Skip to main content
British Journal of Clinical Pharmacology logoLink to British Journal of Clinical Pharmacology
. 2011 Oct;72(4):634–646. doi: 10.1111/j.1365-2125.2011.04034.x

Anti-platelet therapy: phosphodiesterase inhibitors

Paolo Gresele 1, Stefania Momi 1, Emanuela Falcinelli 1
PMCID: PMC3195739  PMID: 21649691

Abstract

Inhibition of platelet aggregation can be achieved either by the blockade of membrane receptors or by interaction with intracellular signalling pathways. Cyclic adenosine 3′,5′-monophosphate (cAMP) and cyclic guanosine 3′,5′-monophosphate (cGMP) are two critical intracellular second messengers provided with strong inhibitory activity on fundamental platelet functions. Phosphodiesterases (PDEs), by catalysing the hydrolysis of cAMP and cGMP, limit the intracellular levels of cyclic nucleotides, thus regulating platelet function. The inhibition of PDEs may therefore exert a strong platelet inhibitory effect. Platelets possess three PDE isoforms (PDE2, PDE3 and PDE5), with different selectivity for cAMP and cGMP. Several nonselective or isoenzyme-selective PDE inhibitors have been developed, and some of them have entered clinical use as antiplatelet agents. This review focuses on the effect of PDE2, PDE3 and PDE5 inhibitors on platelet function and on the evidence for an antithrombotic action of some of them, and in particular of dipyridamole and cilostazol.

Keywords: cilostazol, dipyridamole, phosphodiesterase inhibitor, platelet

Introduction

Inhibition of platelet aggregation has shown great benefit for the treatment and prevention of ischaemic cardiovascular disease. Platelet inhibition can be achieved either by blockade of membrane receptors or by interaction with intracellular signalling pathways. While receptor antagonism may provide high specificity, the inhibition of platelet signal transduction may display broader effects, suppressing platelet activation regardless of the initial stimulus. Inhibition of signalling can be obtained either by interfering with platelet-activating second messengers or by amplifying the action of physiological platelet inhibitors, such as endothelium-derived prostacyclin (PGI2) and nitric oxide (NO), which act by activating adenylyl and guanylyl cyclases respectively, thus increasing intraplatelet cyclic adenosine 3′,5′-monophosphate (cAMP) and cyclic guanosine 3′,5′-monophosphate (cGMP). cAMP and cGMP are two critical inhibitory intracellular second messengers regulating fundamental platelet processes. In fact, elevation of platelet cyclic nucleotides interferes basically with all known platelet activatory signalling pathways and thus blocks cytoskeletal rearrangement, fibrinogen receptor activation, degranulation and expression of pro-inflammatory mediators. Indeed, cAMP and cGMP activate protein kinases that phosphorylate specific substrates (i.e. Rap1, MLCK, vasodilator-stimulated phosphoprotein, etc.) thus interfering with receptor/G-protein activation, phospholipase C, protein kinase C and mitogen-activated protein kinase activation, and blocking cytosolic Ca2+ elevation and the reorganization of the cytoskeleton [1].

Phosphodiesterases

Phosphodiesterases (PDEs), by catalysing the hydrolysis of cAMP and cGMP to inactive 5′-AMP and 5′-GMP, limit intracellular levels of cyclic nucleotides and thus regulate the amplitude, duration and compartmentation of cyclic nucleotide signalling [2].

To date, more than 60 different isoforms of PDE have been described in mammalian tissues, grouped into 11 broad families (PDE1–PDE11) based on differences in their structure, kinetics, regulatory properties and sensitivity to chemical inhibitors. Alternative splicing and transcription start sites also contribute to the multiplicity of the different isoforms, many of which possess species-specific tissue and/or cellular distribution. Current data suggest that individual isozymes modulate distinct regulatory pathways in cells.

Given that PDEs are associated with many physiological functions, targeting pathological conditions by modulating individual PDEs has been the object of intense study (Table 1).

Table 1.

Phosphodiesterase (PDE) families, tissue expression, their inhibitors and their role in disease

Family Substrate Tissue expression Inhibitors Disease targets
PDE1 cGMP > cAMP Heart, vascular smooth muscle and brain Vinpocetine, IC86340 Cerebrovascular disorders and age-related memory impairment, cardiac hypertrophy
PDE2 cGMP = cAMP Platelets, heart and endothelial cells EHNA, EHNA analogues: BAY 60-7550, PDP Memory impairment, endothelial permeability in inflammatory conditions
PDE3 cAMP > cGMP Platelets, vascular smooth muscle, corpus cavernosum and heart Cilostazol, milrinone, vesnarinone, lixazinone, anagrelide Peripheral vascular disease, congestive heart failure, airways disease, fertility, ischaemic cardiovascular disease
PDE4 cAMP Lung, heart, vascular smooth muscle, brain, inflammatory and immune cells Rolipram, etazolate, zardaverine Chronic obstructive pulmonary disease, asthma, allergic disease
PDE5 cGMP Platelets, vascular smooth muscle and corpus cavernosum Sildenafil, vardenafil, tadalafil, zaprinast, dypiridamole Erectile dysfunction, ischaemic cardiovascular disease
PDE6 cGMP > cAMP Retinal rods and cones Sildenafil, zaprinast, dypiridamole None
PDE7 cAMP > cGMP T cell, B cell, skeletal muscle and heart BRL 50481, IC242, dypiridamole Inflammation, osteoporosis
PDE8 cAMP Testis, eye, liver, kidney, skeletal muscle, embryo, ovary and brain Zaprinast None
PDE9 cGMP Brain, small intestinal smooth muscle, liver, kidney, lung, testis, skeletal muscle and heart BAY 73-6691 Alzheimer's disease
PDE10 cAMP > cGMP Testis and brain None None
PDE11 cAMP = cGMP Skeletal muscle, prostate, kidney, liver, pituitary, salivary glands and testis None None

In bold are the drugs discussed in detail in the manuscript. PDE, phosphodiesterase; cAMP, cyclic adenosine 3′,5′-monophosphate; cGMP, cyclic guanosine 3′,5′-monophosphate.

Functionally, phosphodiesterases can be classified in terms of their affinity and rates of degradation for cAMP and cGMP. The isoforms PDE1, 2 and 3 hydrolyse both cAMP and cGMP, whereas PDE4 and PDE8 specifically hydrolyse cAMP, and PDE5 specifically hydrolyses cGMP. Concerning affinity for cyclic nucleotides, PDE3 isoforms are high-affinity enzymes, with KD values <150 nm, while other families, such as PDE1 and PDE5, have affinities in the micromolar range. For many PDEs, affinity for one substrate is so much higher than for the other as to render them functionally monospecific. The isoform PDE3 has a relatively high affinity for both cAMP and cGMP, but a much lower efficacy of hydrolysis for cGMP, behaving essentially as a pure cAMP PDE; it has also been referred to as cGMP-inhibited cAMP PDE because cGMP may competitively inhibit AMP hydrolysis [3]. Thus, in platelets, agents that activate guanylyl cyclase potentiate the effects of activators of adenylyl cyclase [4].

Platelets express three PDE isoenzymes: PDE2, PDE3 and PDE5 (Table 2). Hidaka and Asano [5] were the first to resolve the PDE activity of platelets into three distinct peaks, as follows: the first prefers cGMP as a substrate, with a Km of about 1 µm, and is selectively inhibited by PDE5 inhibitors; the second hydrolyses cAMP and cGMP equally well [6] and is selectively inhibited by PDE2 inhibitors; and the third has a high affinity for both cAMP and cGMP, but hydrolyses cAMP much more rapidly than cGMP, and is selectively inhibited by PDE3 inhibitors. Thus, in platelets cAMP is hydrolysed by PDE3 and PDE2, and cGMP is hydrolysed by PDE5 and PDE2 (Figure 1); these isozymes account for at least 90% of platelet PDE activity.

Table 2.

Phosphodiesterases (PDEs) in platelets and their inhibitors

Family Substrate Inhibitors Phase of development Indication
PDE2 cGMP = cAMP EHNA Preclinical Endotoxaemia
BAY 60-7550 Preclinical Cognitive impairment
PDP Preclinical Cognitive impairment
PDE3 cAMP > cGMP Cilostazol Registered Intermittent claudication; peripheral arterial disease
Milrinone Registered Congestive heart failure
Vesnarinone Phase II Sarcoma Kaposi; HIV infection
Lixazinone Preclinical Polycystic kidney disease
Anagrelide Registered Essential thrombocythaemia
PDE5 cGMP Dypiridamole Registered In association with aspirin for second prevention of ischaemic cerebrovascular disease; as adjunct to coumarin anticoagulants in the prevention of thromboembolic complications in patients with prosthetic heart valves
Sildenafil Registered Erectile dysfunction
Vardenafil Registered Erectile dysfunction
Tadalafil Registered Pulmonary arterial hypertension; erectile dysfunction
Zaprinast Preclinical Allergic disease

In bold are the drugs discussed in detail in the manuscript. PDE2, phosphodiesterase inhibitor-2; PDE3, phosphodiesterase inhibitor-3; PDE5, phosphodiesterase inhibitor-5; cAMP, cyclic adenosine 3′,5′-monophosphate; cGMP, cyclic guanosine 3′,5′-monophosphate; EHNA, erythro-9-(2-hydroxy-3-nonyl)adenine; PDP, -(6-phenyl-2-oxohex-3-yl)-2-(3,4-dimethoxybenzyl)-purin-6-one.

Figure 1.

Figure 1

Schematic representation of a platelet and the mechanisms regulating intraplatelet levels of cyclic nucleotides (cAMP and cGMP); the three isoforms of phosphodiesterases so far described in platelets and their pharmacological inhibitors are described. In bold are the drugs discussed in detail. AC, adenylate cyclase; sCG, solubile guanylate cyclase; ATP, adenosine triphoasphate; GTP, guanosine triphosphate; cAMP, cyclic adenosine 3′,5′-monophosphate; cGMP, cyclic guanosine 3′,5′-monophosphate; AMP, adenosine 3′,5′-monophosphate; GMP, guanosine 3′,5′-monophosphate; PDE2, phosphodiesterase inhibitor-2; PDE3, phosphodiesterase inhibitor-3; PDE5, phosphodiesterase inhibitor-5

Phosphodiesterase inhibitors

Soon after the discovery of PDEs, it was found that caffeine is an effective inhibitor of PDE activity, and a number of nonselective PDE inhibitors, including the caffeine analogue theophylline, entered clinical use. Since then, several isoenzyme-selective PDE inhibitors have been developed as therapeutics or are currently under investigation for various disorders.

This review focuses on the effects of PDE2, PDE3 and PDE5 inhibitors on platelet function and on their potential as antithrombotic agents.

Nonselective PDE inhibitors: methylxanthines and pentoxifylline

Naturally occurring methylxanthines were the earliest PDE inhibitors to be discovered, and among them, caffeine (1,3,7-trimethylxanthine) was the first [7]. Some years later, theophilline (1,3-dimethylxanthine) and theobromine, more effective PDE inhibitors, were characterized [8]. Ardlie and co-workers documented inhibition of platelet aggregation by caffeine and theophylline in 1967 [9]. In a milestone study in 1971, Mills and Smith showed that adenosine increases cAMP in platelets and that methylxanthines prevent the conversion of cAMP to AMP, acting as PDE inhibitors, thus greatly increasing the inhibitory effects of adenosine on platelet aggregation [10]. Caffeine and theophylline act also as nonselective adenosine receptor antagonists [11]. In healthy volunteers, 250 mg caffeine orally three times a day for 7 days reduced platelet aggregation, increased intracellular cAMP and upregulated the platelet adenosine A2A receptors [12].

Pentoxifylline (3,7-dimethyl-1-(5-oxohexyl)-xanthine), a vasoactive drug used in patients with intermittent claudication [13] and reported to reduce whole blood viscosity and to improve erythrocyte deformability, was shown to inhibit platelet aggregation in vitro in platelet-rich plasma, but at concentrations higher than those attainable in vivo[14]. However, given that the platelet inhibitory effect was potentiated by PGI2, it was suggested that the in vivo antiplatelet activity could be stronger [15]; moreover, pentoxifylline was found to inhibit platelet aggregation in whole blood more effectively than in platelet-rich plasma, due to the contribution of an adenosine uptake-inhibitory effect on erythrocytes [16]. So far, there is no evidence that pentoxyfilline reduces ischaemic cardiovascular events [13].

PDE2 inhibitors

The isoform PDE2 hydrolyses both cAMP and cGMP, and high concentrations of cGMP stimulate PDE2 [2].

Inhibitors of PDE2 have been used mainly as research tools, but ongoing studies investigate their effectiveness for memory impairment and prevention of endothelial permeability in inflammation [2].

Erythro-9-(2-hydroxy-3-nonyl) adenine (EHNA), an inhibitor of adenosine deaminase (ADA), was shown to act as a selective PDE2 inhibitor with an IC50 of 3 µm and at least 50-fold selectivity over other PDEs [17]. EHNA (20 µm) has no direct effect on platelet aggregation, but it potentiates the inhibition of thrombin-induced platelet aggregation by nitroprusside, a guanylyl cyclase stimulator [17]. The use of EHNA as a tool to assess the role of PDE2 in platelets is limited by the low PDE2-inhibitory potency and by the concomitant adenosine deaminase inhibitory action.

Recently, a new series of thienylacylhydrazone derivatives synthesized from natural safrole (4-allyl-1,2-methyldioxybenzene), a Brazilian natural product obtained from Ocotea pretiosa, have been studied as PDE2 inhibitors for their antiplatelet activity [18]. The most potent of these inhibited platelet aggregation induced by arachidonic acid (IC50 0.2–3.1 µm) and collagen (IC50 0.9–3.4 µm), effects enhanced by sodium nitroprusside without interference with ADP-induced aggregation, ATP release and thromboxane (Tx)B2 production [19]. Moreover, one of these induced a concentration-dependent relaxation of intact rat aortic rings (IC50 74 µm) [20].

Novel selective PDE2 inhibitors with nanomolar potency, such as 9-(6-phenyl-2-oxohex-3-yl)-2-(3,4-dimethoxybenzyl)-purin-6one (PDP), have recently been developed but not tested on platelets [21].

PDE3 inhibitors

The isoform PDE3 comprises two subfamilies, PDE3A and PDE3B, showing distinct and overlapping tissue and subcellular distributions. In situ hybridization studies have shown that PDE3A is highly expressed in the cardiovascular system, including the myocardium, vascular smooth muscle cells and megakaryocytes, while PDE3B mRNA is detected in adipocytes [22]. Recently, it has been shown that PDE3A is the main subtype of PDE3 expressed in platelets [23].

Anagrelide

Anagrelide (Agrylin/Xagrid; BL-4162A; 6,7-dichloro-1,5-dihydroimidazo[2, 1–6] quinazolin-2[3H]one monohydrochloride hydrate) is a potent inhibitor of PDE3 and a potent and broad-spectrum inhibitor of platelet aggregation (IC50 = 36 nm) [24].

During studies in humans, however, anagrelide was found to produce thrombocytopoenia [25]; although the mechanisms through which anagrelide inhibits the megakaryocytes maturation are not completely understoood, the drug has entered clinical use for patients with essential thrombocythemia [26].

Cilostazol

Cilostazol, a 2-oxo-quinoline derivative (6-(4-(1-cyclohexyl-1H-tetrazol-5-yl)butoxy)-3,4-dihydro-2(1H)-quinolinone), was registered in Japan and other Asiatic countries in 1988 and approved in the USA in 1999 for the treatment of intermittent claudication [27].

Cilostazol is a specific and strong inhibitor of PDE3 in platelets (IC50 = 0.2 µm) and smooth muscle cells, where it diminishes intracellular calcium, causing smooth muscle cell relaxation and inhibition of platelet activation [28]. Cilostazol also inhibits adenosine uptake, thus enhancing adenosine levels that in turn enhance intracellular cAMP, resulting in additional increases in cAMP [28].

Platelet inhibition

Cilostazol inhibits both primary and secondary platelet aggregation induced by collagen, ADP, arachidonic acid and adrenaline with IC50 values ranging from 3.6 to 15.0 µm, depending on the agonist [29, 30]. It also suppresses the expression of P-selectin (IC50 25 µm) [31], TxB2 production, platelet factor 4 and platelet-derived growth factor release, and enhances the antiplatelet effects of PGI2[32]. Cilostazol inhibits shear stress-induced platelet activation in vitro, with an IC50 of 15.0 µm[33], and ex vivo is not dissimilar from ticlopidine plus aspirin [34]. Among patients with acute myocardial infarction undergoing coronary stenting, the association of cilostazol with clopidogrel plus aspirin resulted in a greater antiplatelet effect in comparison to clopidogrel plus aspirin [35]. Moreover, the addition of cilostazol to clopidogrel gives greater inhibition of platelet aggregation than clopidogrel, either at the standard or at double dose, in clopidogrel low responders [36].

One potential benefit of the use of cilostazol over conventional antiplatelet therapy is the relatively short recovery time of platelet function [37].

Cilostazol inhibits the expression of monocyte chemoattractant protein-1, an initial trigger in the development of atherosclerosis, in human umbilical vein endothelial cells by increasing intracellular cAMP [38].

Cilostazol is rapidly absorbed, and reaches peak plasma concentrations (775 ng ml−1 = 2.09 µm) at about 2.4 h after oral administration. In plasma it is largely bound to proteins (95–98%), primarily albumin [39, 40]. Specifically, metabolism of cilostazol occurs primarily via CYP3A5 and, to a lesser extent, CYP2C19, while <1% of the administered dose is excreted unchanged in urine [41]. The CYP3A5 and CYP2C19 polymorphisms explain the substantial interindividual variability in the metabolism of cilostazol, with a coefficient of variation of about 40–60% [39, 42].

After oral administration, cilostazol and its metabolites, 3,4-dehydrocilostazol (OPC-13015) and 4-trans-hydroxy-cilostazol (OPC-13213), show a half-life of approximately 10 h, with a twofold accumulation during repeated administration [28].

Effect on bleeding time

There is no evidence that cilostazol prolongs the bleeding time when compared with aspirin, clopidogrel or ticlopidine [43, 44], even when used in combination with these antiplatelet drugs [44].

In a study in healthy men comparing ticlopidine, aspirin and cilostazol, bleeding time was significantly prolonged by aspirin and ticlopidine but not by cilostazol [45].

In patients with peripheral arterial disease, aspirin or clopidogrel alone significantly prolonged bleeding time, and even more when used in combination. In contrast, no prolongation of the bleeding time was observed with cilostazol alone, nor was there a further prolongation when added to aspirin or clopidogrel [46].

Animal models

Cilostazol exerts antithrombotic activity in different animal models. Pulmonary thromboembolism induced by ADP and collagen in mice was reduced by cilostazol at 3 and 10 mg kg−1per os, more effectively than by aspirin and pentoxifylline [29]. Cerebral infarction induced by the injection of arachidonic acid into the internal carotid artery of rabbits was reduced by 1 mg kg−1 cilostazol [47].

In a porcine model of totally occlusive thrombosis of the carotid artery induced by electrical injury, cilostazol significantly prolonged the time to occlusion and decreased thrombus weight more than ticlopidine [48]. In FeCl3 venous thrombosis model in rats, oral administration of 50 mg kg−1 cilostazol significantly decreased venous thrombus weight [49].

Clinical studies

Several studies have shown the superiority of cilostazol vs placebo and also vs pentoxifylline in increasing the walking distance [50] and quality of life of peripheral arterial disease patients [51].

Cilostazol was tested for secondary prevention of stroke in a randomized, placebo-controlled, double-blind trial in 1052 patients in Japan [52]. Cilostazol significantly reduced the recurrence of ischaemic stroke (−41.7%; confidence interval (CI) −9.2–62.5%) compared with placebo, and the incidence of the combined end-point of myocardial infarction, transient ischaemic attack and intracranial haemorrhage. The benefits were achieved without a significant increase of bleeding (2.8 vs 2.1%) and with no fatal intracranial haemorrhage [52]. These data seem to confirm a low bleeding risk with cilostazol. In the second Cilostazol Stroke Prevention Study (CSPS 2), in 2757 patients with a noncardioembolic cerebral infarction within the previous 26 weeks, cilostazol 100 mg twice daily resulted to be non-inferior, and possibly superior, to aspirin 81 mg once daily, in preventing stroke recurrence (yearly rate of 2·76% (n = 82) in the cilostazol group and 3·71% (n = 119) in the aspirin group, hazard ratio 0·743, 95% CI 0·564–0·981; P = 0·0357) and was associated with fewer haemorrhagic events [53]. A recent meta-analysis of 12 double-bind, placebo-controlled trials in patients with atherothrombotic disease concluded that cilostazol is associated with a significant risk reduction of cerebrovascular events (relative risk [RR] 0.86; 95% CI 0.74–0.99; P = 0.038), with no associated increase of bleeding [54]. There was no significant difference in the incidence of cardiac events (RR 0.99; 95% CI 0.83–1.17; P = 0.908) [54].

In a study in 1212 patients with acute coronary syndromes undergoing percutaneous coronary intervention, cilostazol added to aspirin plus clopidogrel reduced the composite end-point of cardiac death, myocardial infarction and stroke, in comparison to aspirin plus clopidogrel [55].

Recently, however, a multicentre randomized trial in 960 patients undergoing coronary drug-eluting stent implantation showed that the addition of cilostazol to aspirin and clopidogrel for 6 months did not reduce the composite adverse outcome of cardiac death, nonfatal myocardial infarction, ischaemic stroke or target lesion revascularization [56].

Cilostazol has also been suggested to prevent post-stent restenosis [57]. Cilostazol was compared with ticlopidine, both in addition to aspirin, in 397 patients undergoing elective coronary stenting for the rate of restenosis; restenosis tended to be lower with cilostazol, but not significantly [58].

A recent study investigated whether periprocedural cilostazol affects the incidence of in-stent restenosis (ISR) or target vessel revascularization (TVR) after carotid artery stenting. The study group comprised 553 patients undergoing carotid artery stenting followed for 30 months, 207 of whom (37.4%) were treated with cilostazol. The incidence of ISR or TVR was significantly lower in the cilostazol-treated group than in the untreated group (1.4 vs 6.4%; log-rank P = 0.006) [59]. A metanalysis of five randomized controlled trials comparing triple therapy cilostazol plus aspirin plus thienopyridines with aspirin plus clopidogrel, involving 1597 patients undergoing coronary stenting, showed that triple therapy significantly reduced the 6 months restenosis rate (12.7 vs 21.9%; odds ratio 0.5; 95% CI 0.38–0.66; P < 0.001) [60].

Finally, in a small study, cilostazol administration improved long-term patency after carotid artery stenting in 97 patients with high-grade carotid stenosis monitored for a 12 month period (0 vs 15.7%, P = 0.03) [61].

The most common adverse effects of cilostazol are headache, tachycardia, palpitations, soft stools and diarrhoea [62].

Milrinone

Milrinone, a specific PDE3A inhibitor, inhibits arachidonic acid-induced platelet shape change and platelet aggregation induced by ADP, U46619, collagen and calcium ionophore, both in whole blood and in platelet-rich plasma [63], with IC50 values as low as 1 µmol l−1, a concentration lower than the concentration achievable after therapeutic administration (1.5 µmol l−1), indicating that an antiaggregating effect of milrinone can occur in vivo[64]. Milrinone induces an elevation of intraplatelet cAMP in a dose-dependent manner, resulting in the inhibition of platelet aggregation [65].

Milrinone is currently in clinical use for congestive heart failure [66].

PDE3–PDE5 inhibitors

Dipyridamole

Dipyridamole (2,6-bis (diethanolamino)-4,8-dipiperidino-pyrimido 5,4-d pyrimidine) was synthesized more than 50 years ago and initially used as a coronary vasodilator. However, it was soon shown that dipyridamole inhibited platelet aggregation [67, 68] and thrombus formation in rabbits [69], opening the way to the use of dipyridamole as an antithrombotic agent [70].

Mechanisms of action

Dipyridamole affects platelet function by acting on the following different targets: it inhibits the reuptake of adenosine by red blood cells, in this way enhancing plasma levels of this vasodilator and platelet inhibitory nucleoside; it acts as an inhibitor of PDE5 and PDE3, thus increasing intraplatelet cAMP and/or cGMP; and it acts as a antioxidant by scavenging free radicals that inactivate cyclo-oxygenase, thus enhancing PGI2 biosynthesis.

Adenosine is released by tissues in the extracellular space as a breakdown product of ATP during ischaemia, or by erythrocytes stressed by elevated shear forces, and is then taken up avidly by erythrocytes to keep plasma levels low. Inhibition of adenosine reuptake by dipyridamole is concentration dependent and reaches 90% at 1 µm of dipyridamole in whole blood [71], a concentration in the range of those attained after oral administration to humans (0.5–6 µm). Dipyridamole inhibits platelet aggregation in whole blood, as assessed by impedance aggregometry, but not in platelet-rich plasma both in vitro and ex vivo, by blocking the reuptake of adenosine [72, 73].

A stronger antiplatelet effect of dipyridamole in whole blood than in plasma was confirmed using different laboratory methods [7477].

The slight PDE3-inhibitory action of dipyridamole increases the effects of adenosine and PGI2, both stimulators of adenylylcyclase, leading to inhibition of platelet activation [78, 79].

Dipyridamole potentiates the inhibitory effects of NO on human and rabbit platelets also [80] by inhibiting PDE5 and thus increasing production of vasodilator-stimulated phosphoprotein, an established marker of the NO/cGMP effects [81], both in vitro and in vivo[82]. However, the inhibition of PDE5 is detectable in vitro only at concentrations (100–200 µm) much higher than those attainable after oral administration.

The antioxidant properties of dipyridamole may contribute to its antithrombotic effect. In fact, not only does dipyridamole enhance extracellular adenosine, which reduces superoxide anion generation by human neutrophils and directly scavenges oxygen- and hydroxyl-radicals, but it also has direct antioxidant effects. In fact, dipyridamole has better antioxidant properties than ascorbic acid, α-tocopherol and probucol [83, 84].

The antioxidant activity of dipyridamole leads to the inhibition of leukotriene B4 production in vitro by stimulated white blood cells [85]. Moreover, based on its antioxidant properties, dipyridamole was shown to prolong prostacyclin production by ‘exhausted’ vessel walls, preventing the autoinactivation of cyclo-oxygenase caused by enhanced peroxide formation [86], an activity similar to the so-called physiological prostacyclin-regulating plasma factor [87].

The redox state of dypiridamole regulates its antioxidant properties, which appear to be mediated in vascular cells by suppression of nuclear factor κB signalling [88].

Additional pharmacological effects of dipyridamole that may be of relevance for prevention of atherothrombosis are the inhibition of vascular smooth muscle cell proliferation, the prevention of endothelium–leukocyte interactions [89], the increase of antithrombotic properties of endothelial cells [90], the interference with interaction of leukocytes with endothelium, the enhancement of interleukin-1-stimulated NO production by smooth muscle cells [91], and the inhibition of inflammatory gene expression in platelet–monocyte aggregates [92]. Very recently, an additional interesting observation has shown that inhibition of multidrug resistance protein-4-mediated transport by dipyridamole increases aspirin entrapment in platelets and its in vitro effect on cyclo-oxygenase-1 activity [93], thus explaining in part the pharmacodynamic [94] and therapeutic [95, 96] synergism with aspirin.

Antithrombotic properties

The antithrombotic effects of dipyridamole were evaluated in several animal models. Thrombus formation on air-injured carotid arteries of rabbits was reduced by dipyridamole, while salicylate increased it [97]. In rabbits, the accumulation of radioactive fibrinogen at balloon angioplasty-treated carotid arteries was significantly reduced by dipyridamole (0.45 mg kg h−1 for 4 h) in comparison to heparin [98].

More recently, in a model of chronic hindlimb ischaemia induced by the ligation of the common femoral artery branches in mice, dipyridamole (200 mg kg−1 twice daily, for 7 days) was shown to enhance ischaemia-induced arteriogenesis, thereby quickly restoring blood flow through a protein kinase A-dependent NO pathway [99].

Clinical studies

There is little clinical evidence that dipyridamole alone exerts an antithrombotic effect. Several studies instead have been carried out in combination with aspirin. Dipyridamole in combination with low-dose aspirin is associated with greater stroke risk reduction in patients with ischaemic cerebrovascular disease. This was confirmed in two large clinical studies: the ESPS2 [95], in which 6602 patients with prior stroke or transient ischaemic attack were randomized to aspirin (50 mg daily), slow release dipyridamole (200 mg twice a day), the two agents in a combined formulation, or placebo; and the ESPRIT trial [96], in which 2739 patients with previous transient ischaemic attack were randomized to aspirin (30–325 mg day) with or without dipyridamole (200 mg twice daily). A recent meta-analysis supports the higher efficacy of combination therapy over aspirin alone [100]. Based on these findings, the 2008 American College of Chest Physicians (ACCP) guidelines recommend dual therapy with extended-release dipyridamole plus aspirin over aspirin monotherapy for stroke prevention after a first transient ischaemic attack or stroke [101]. More recently, a large clinical trial, the PRoFESS study, carried out to investigate whether aspirin 25 mg and extended-release dypiridamole 200 mg twice a day was not inferior to clopidogrel 75 mg once a day in 20 332 patients with previous stroke, did not meet predefined criteria for non-inferiority, but showed similar rates of recurrent stroke with the two treatments.

There were more haemorrhagic strokes with aspirin plus extended-release dipyridamole than with clopidogrel [102]. The degree of functional impairment (disability and cognitive decline) at 3 months after stroke was similar across treatment arms [103].

Another condition for which there is evidence of efficacy of dipyridamole is secondary prophylaxis of thromboembolic events in patients with mechanical heart valves in combination with warfarin [104]. Concerning the effects in other clinical indications, a recent overview of 27 randomized long-term secondary prevention trials involving 23 019 patients presenting with an arterial vascular disease (cardiac disease, cerebral ischaemia, thrombosis of haemodialysis fistula, patients prone to develop atherosclerosis) comparing dipyridamole, alone or in combination with an antiplatelet drug other than dipyridamole (chiefly aspirin), with placebo found that there was no evidence that dipyridamole reduced the risk of vascular death, while it significantly reduced the risk of further vascular events (RR 0.88; 95% CI 0.81–0.95); however, this benefit was statistically significant only in patients presenting after cerebral ischaemia [105].

PDE5 inhibitors

To date, only one family of PDE5, PDE5A, has been described. Three splice variants of PDE5A, PDE5A1/2/3, have been found in humans, differing in their N-terminal region, but with similar Km for cGMP [5].

Three PDE5 inhibitors, sildenafil (Viagra), vardenafil (Levitra) and tadalafil (Cialis), are currently in clinical use for erectile dysfunction.

Actually, the first developed PDE5 inhibitor to be used in humans was zaprinast (M&B22948), originally designed as a mast cell stabilizing drug for the treatment of allergic diseases [106]. The observation that cGMP elevation induced by zaprinast was associated with smooth muscle cell relaxation opened the possibility to use PDE5 inhibitors in cardiovascular diseases [107]. Zaprinast inhibits human platelet PDE5 with an IC50 of 0.3 µm and PDE2A with an IC50 of 42 µm. In association with sodium nitroprusside (1 µm), zaprinast led to a complete concentration-dependent inhibition of serotonin release in washed platelets (IC50∼1.6 µm) [108]. This compound was an unsuccessful clinical drug candidate, turning out to be weak and poorly selective for PDE5. Consequently, a variety of compounds with key variations in the heterocyclic ring system of zaprinast were prepared, leading to the identification in 1989 of sildenafil as a PDE5 inhibitor, which was 100 times as potent as zaprinast and highly specific in its action.

Sildenafil (1-[4-ethoxy-3-(6,7-dihydro-1-methyl-7-oxo-3-propyl-1H-pyrazolo [4,3-d]pyrimidin-5-yl) phenylsulphonyl]-4-methylpiperazine) is a potent PDE5 inhibitor, with high selectivity for human PDE5 over PDE2, PDE3 and PDE4 (>1000-fold), and moderate selectivity (>80-fold) over PDE1 [109]. Sildenafil instead is only ∼10-fold more potent for PDE5 than for PDE6 (an enzyme found in the photoreceptors of the human retina), and this explains the frequent adverse effect of flashes of blue light with its use. Platelet PDE5 is inhibited by sildenafil with a IC50 of 6.3 nm[110]. While sildenafil alone had no effect on platelet function, it potentiated the inhibition by sodium nitroprusside of rabbit and human platelets [110].

Sildenafil is rapidly absorbed after oral administration, with ∼40% bioavailability. Plasma concentrations peak within 30–120 min (median, 60 min), and the drug is primarily metabolized by hepatic cytochrome P450 3A4 (major route) and 2C9 (minor route), which convert it to an active N-desmethyl metabolite that possesses 50% of the parent drug's potency for PDE5 [111].

The administration of sildenafil (100 and 50 mg in healthy volunteers) inhibited collagen-induced aggregation (100 mg), with an additive effect when combined with nitrates (isosorbide di-nitrate, 10 mg) [112]. Moreover, ADP-stimulated glycoprotein IIb/IIIa receptor activation was inhibited by sildenafil (100 mg) [113]. A study in healthy men showed that bleeding time was significantly prolonged (+72%) 1 h after sildenafil intake (100 mg), with recovery after 4 h, whereas a lower dose (50 mg) did not alter the bleeding time [112]. Sildenafil was also reported to increase the incidence of epistaxis in patients on concomitant vitamin K antagonists and in combination with heparin had an additive effect on bleeding time in rabbits [114]. Given the relatively low selectivity of sildenafil for PDE5, other more selective PDE5 inhibitors have been developed: vardenafil (KD of 0.6–0.7 nm; selectivity over PDE6, approximately 16-fold) [115] and tadalafil (KD range of 0.9–6.7 nm; 200–700 times more selective for PDE5 than PDE6) [116]. In patients with erectile dysfunction associated with cardiovascular risk factors (dyslipidaemia, hypertension and smoking), tadalafil enhanced cGMP and significantly reduced P-selectin expression in platelets [117].

Conclusions

Antiplatelet therapy is the mainstay of treatment of patients with acute cardiovascular ischaemic events and of secondary prophylaxis of patients with a previous ischaemic event. Despite great progress, even optimal antithrombotic therapy still does not offer satisfactory protection against cardiovascular events and is associated with serious risk of haemorrhagic adverse effects. Agents targeting thromboxane production or activity, ADP and fibrinogen binding to platelets have proved of benefit but have also shown limitations.

An alternative, appealing strategy is to develop agents interfering with intracellular signalling pathways [118]. Phosphodiesterase inhibitors, by increasing crucial intraplatelet second messengers, such as cGMP and cAMP, have theoretically great potential for platelet inhibition. In fact, differently from aspirin or clopidogrel, not only they may inhibit platelet activation induced by whatever stimulus, but they may exert beneficial cardiovascular effects though their capacity to regulate the interaction of platelets with vascular cells in the setting of ischaemic cardiovascular disease. However, the widespread distribution of PDE in the body renders it difficult for an effective antiplatelet action be achieved without significant unwanted effects. Moreover, the reversibility of the effect of most clinically available PDE inhibitors on their target may represent a serious limitation for their antithrombotic effectiveness in long-term secondary prophylaxis. It is now established that continuous platelet inhibition is required in order to obtain an effective antithrombotic protection, and the reversibility of the blockade of the target, with temporary restoration of platelet function, in patients at high cardiovascular risk may be associated with enhanced ischaemic events, as shown by the negative experience with orally active glycoprotein IIb/IIIa antagonists [119]. Is not a chance that the antiplatelet agents currently used in long-term secondary prevention of ischaemic events, i.e. aspirin and thyenopiridines, both act by irreversibly blocking their target. A deeper understanding of the physiology of PDEs in platelets and other tissues, the development of techniques allowing the targeting of PDE inhibition to platelets and the development of irreversible or long-acting isoenzyme-selective PDE inhibitors may potentially lead to advances in antiplatelet therapy.

Acknowledgments

This work was supported in part by research grants from Fondazione Cassa di Risparmio di Perugia (protocol n. 2002.020.0097, 2010.020.0161 and 2009.010.0478) to Paolo Gresele.

The expert editorial assistance of Dr S. Orsini is gratefully acknowledged.

Competing Interests

There are no competing interests to declare.

REFERENCES

  • 1.Daniel JL, Ashby B, Pulcinelli F. Platelet signaling: cAMP and cGMP. In: Gresele P, Page C, Fuster V, Vermylen J, editors. Platelets in Thrombotic and Non-Thrombotic Disorders. Cambridge: Cambridge University Press; 2002. pp. 290–304. [Google Scholar]
  • 2.Bender AT, Beavo JA. Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol Rev. 2006;58:488–520. doi: 10.1124/pr.58.3.5. [DOI] [PubMed] [Google Scholar]
  • 3.Grant PG, Colman RW. Purification and characterization of a human platelet cyclic nucleotide phosphodiesterase. Biochemistry. 1984;23:1801–7. doi: 10.1021/bi00303a034. [DOI] [PubMed] [Google Scholar]
  • 4.Maurice DH, Haslam RJ. Molecular basis of the synergistic inhibition of platelet function by nitrovasodilators and activators of adenylate cyclase: inhibition of cyclic AMP breakdown by cyclic GMP. Mol Pharmacol. 1990;37:671–81. [PubMed] [Google Scholar]
  • 5.Hidaka H, Asano T. Human blood platelet 3′:5′-cyclic nucleotide phosphodiesterase. Isolation of low-Km and high-Km phosphodiesterase. Biochim Biophys Acta. 1976;429:485–97. doi: 10.1016/0005-2744(76)90296-5. [DOI] [PubMed] [Google Scholar]
  • 6.Haslam RJ, Dickinson NT, Jang EK. Cyclic nucleotides and phosphodiesterases in platelets. Thromb Haemost. 1999;82:412–23. [PubMed] [Google Scholar]
  • 7.Sutherland EW, Rall TW. Fractionation and characterization of a cyclic adenine ribonucleotide formed by tissue particles. J Biol Chem. 1958;232:1077–91. [PubMed] [Google Scholar]
  • 8.Butcher RW, Sutherland EW. Adenosine 3′-5′-phosphate in biological materials. Purification and properties of cyclic 3′-5′-nucleotide phosphodiesterase and use of this enzyme to characterize adenosine 3′-5′-phosphate in human urine. J Biol Chem. 1962;237:1244–50. [PubMed] [Google Scholar]
  • 9.Ardlie NG, Glew G, Schultz BG, Schwartz CJ. Inhibition and reversal of platelet aggregation by methylxanthines. Thromb Diath Haemorrh. 1967;18:670–3. [PubMed] [Google Scholar]
  • 10.Mills DCB, Smith JB. The influence on platelet aggregation of drugs that affect the accumulation of adenosine 3′:5′-cyclic monophosphate in platelets. Biochem J. 1971;121:185–96. doi: 10.1042/bj1210185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Evoniuk G, Jacobson KA, Shamim MT, Daly JW, Wurtman RJ. A1- and A2-selective adenosine antagonists: in vivo characterization of cardiovascular effects. J Pharmacol Exp Ther. 1987;242:882–7. [PMC free article] [PubMed] [Google Scholar]
  • 12.Varani K, Portaluppi F, Merighi S, Ongini E, Belardinelli L, Borea PA. Caffeine alters A2A adenosine receptors and their function in human platelets. Circulation. 1999;99:2499–502. doi: 10.1161/01.cir.99.19.2499. [DOI] [PubMed] [Google Scholar]
  • 13.Jackson MR, Clagett GP. Antithrombotic therapy in peripheral arterial occlusive disease. Chest. 2001;119:283S–99S. doi: 10.1378/chest.119.1_suppl.283s. [DOI] [PubMed] [Google Scholar]
  • 14.Nenci GG, Gresele P, Agnelli G, Ballatori E. Effect of pentoxifylline on platelet aggregation. Pharmatherapeutica. 1981;2:532–8. [PubMed] [Google Scholar]
  • 15.Weithmann KU. The influence of pentoxifylline on interaction between blood vessel wall and platelets. IRCS Med Sci. 1980;8:293–4. [Google Scholar]
  • 16.Manrique RV, Manrique V. Platelet resistance to prostacyclin. Enhancement of the antiaggergatory effect of prostacyclin by pentoxifylline. Angiology. 1987;38:101–8. doi: 10.1177/000331978703800202. [DOI] [PubMed] [Google Scholar]
  • 17.Dickinson NT, Jang EK, Haslam RJ. Activation of cGMP-stimulated phosphodiesterase by nitroprusside limits cAMP accumulation in human platelets: effects on platelet aggregation. Biochem J. 1997;323:371–7. doi: 10.1042/bj3230371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lima LM, Ormelli CB, Brito FF, Miranda AL, Fraga CA, Barreiro EJ. Synthesis and antiplatelet evaluation of novel aryl-sulfonamide derivatives, from natural safrole. Pharm Acta Helv. 1999;73:281–92. doi: 10.1016/s0031-6865(99)00004-7. [DOI] [PubMed] [Google Scholar]
  • 19.Brito FC, Kummerle AE, Lugnier C, Fraga CA, Barreiro EJ, Miranda AL. Novel thienylacylhydrazone derivatives inhibit platelet aggregation through cyclic nucleotides modulation and thromboxane A2 synthesis inhibition. Eur J Pharmacol. 2010;638:1–3. doi: 10.1016/j.ejphar.2010.04.003. [DOI] [PubMed] [Google Scholar]
  • 20.Silva CL, Noël F, Barreiro EJ. Cyclic GMP-dependent vasodilatory properties of LASSBio 294 in rat aorta. Br J Pharmacol. 2002;135:293–8. doi: 10.1038/sj.bjp.0704473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Diebold I, Djordjevic T, Petry A, Hatzelmann A, Tenor H, Hess J, Görlach A. Phosphodiesterase 2 mediates redox sensitive endothelial cell proliferation and angiogenesis by thrombin via Rac1 and NADPH oxidase 2. Circ Res. 2009;104:1169–77. doi: 10.1161/CIRCRESAHA.109.196592. [DOI] [PubMed] [Google Scholar]
  • 22.Park H, Young Lee S, Lee DS, Yim M. Phosphodiesterase 4 inhibitor regulates the TRANCE/OPG ratio via COX-2 expression in a manner similar to PTH in osteoblasts. Biochem Biophys Res Commun. 2007;354:178–83. doi: 10.1016/j.bbrc.2006.12.174. [DOI] [PubMed] [Google Scholar]
  • 23.Sun B, Li H, Shakur Y, Hensley J, Hockman S, Kambayashi J, Manganiello VC, Liu Y. Role of phosphodiesterase type 3A and 3B in regulating platelet and cardiac function using subtype-selective knockout mice. Cell Signal. 2007;19:1765–71. doi: 10.1016/j.cellsig.2007.03.012. [DOI] [PubMed] [Google Scholar]
  • 24.Seiler S, Arnold AJ, Grove RI, Fifer CA, Keely SL, Jr, Stanton HC. Effects of anagrelide on platelet cAMP levels, cAMP-dependent protein kinase and thrombin-induced Ca++ fluxes. J Pharmacol Exp Ther. 1987;243:767–74. [PubMed] [Google Scholar]
  • 25.Thiele J, Kvasnicka HM, Schmitt-Graeff A. Effects of anagrelide on megakaryopoiesis and platelet production. Semin Thromb Hemost. 2006;32:352–61. doi: 10.1055/s-2006-942756. [DOI] [PubMed] [Google Scholar]
  • 26.Silverstein MN, Petitt RM, Solberg LA, Fleming JS, Knight RC, Schacter LP. Anagrelide: a new drug for treating thrombocytosis. N Engl J Med. 1988;318:1292–4. doi: 10.1056/NEJM198805193182002. [DOI] [PubMed] [Google Scholar]
  • 27.Faxon DP, Creager MA, Smith SC, Pasternak RC, Olin JW, Bettmann MA, Criqui MH, Milani RV, Loscalzo J, Kaufman JA, Jones DW, Pearce WH. American Heart Association. 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. doi: 10.1161/01.CIR.0000128517.52533.DB. [DOI] [PubMed] [Google Scholar]
  • 28.Shrör K. The pharmacology of cilostazol. Diabetes Obes Metab. 2002;4:14–9. doi: 10.1046/j.1463-1326.2002.0040s2s14.x. [DOI] [PubMed] [Google Scholar]
  • 29.Kimura Y, Tani T, Kanbe T, Watanabe K. Effect of cilostazol on platelet aggregation and experimental thrombosis. Arzneimittelforschung. 1985;35:1144–9. [PubMed] [Google Scholar]
  • 30.Yasunaga K, Mase K. Antiaggregatory effect of oral cilostazol and recovery of platelet aggregability in patients with cerebrovascular disease. Arzneimittelforschung. 1985;35:1189–92. [PubMed] [Google Scholar]
  • 31.Kariyazono H, Nakamura K, Shinkawa T, Yamaguchi T, Sakata R, Yamada K. Inhibition of platelet aggregation and the release of P-selectin from platelets by cilostazol. Thromb Res. 2001;101:445–53. doi: 10.1016/s0049-3848(00)00415-1. [DOI] [PubMed] [Google Scholar]
  • 32.Igawa T, Tani T, Chijiwa T, Shiragiku T, Shimidzu S, Kawamura K, Kato S, Unemi F, Kimura Y. Potentiation of anti-platelet aggregating activity of cilostazol with vascular endothelial cells. Thromb Res. 1990;57:617–23. doi: 10.1016/0049-3848(90)90079-r. [DOI] [PubMed] [Google Scholar]
  • 33.Minami N, Suzuki Y, Yamamoto M, Kihira H, Imai E, Wada H, Kimura Y, Ikeda Y, Shiku H, Nishikawa M. Inhibition of shear stress-induced platelet aggregation by cilostazol, a specific inhibitor of cGMP-inhibited phosphodiesterase, in vitro and ex vivo. Life Sci. 1997;61:PL383–9. doi: 10.1016/s0024-3205(97)00986-7. [DOI] [PubMed] [Google Scholar]
  • 34.Tanigawa T, Nishikawa M, Kitai T, Ueda Y, Okinaka T, Makino K, Ito M, Isaka N, Ikeda Y, Shiku H, Nakano T. 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. doi: 10.1016/s0002-9149(00)00695-0. [DOI] [PubMed] [Google Scholar]
  • 35.Jeong YH, Hwang JY, Kim IS, Park Y, Hwang SJ, Lee SW, Kwak CH, Park SW. Adding cilostazol to dual antiplatelet therapy achieves greater platelet inhibition than high maintenance dose clopidogrel in patients with acute myocardial infarction: results of the adjunctive cilostazol versus high maintenance dose clopidogrel in patients with AMI (ACCEL-AMI) study. Circ Cardiovasc Interv. 2010;3:17–26. doi: 10.1161/CIRCINTERVENTIONS.109.880179. [DOI] [PubMed] [Google Scholar]
  • 36.Lee K, Kim JY, Yoo BS, Yoon J, Hong MK, Ahn MS, Choe H, Lee SH. Cilostazol augments the inhibition of platelet aggregation in clopidogrel low-responders. J Thromb Haemost. 2010;8:2577–9. doi: 10.1111/j.1538-7836.2010.04019.x. [DOI] [PubMed] [Google Scholar]
  • 37.Iwamoto T, Kin K, Miyazaki K, Shin K, Takasaki M. Recovery of platelet function after withdrawal of cilostazol administered orally for a long period. J Atheroscler Thromb. 2003;10:348–54. doi: 10.5551/jat.10.348. [DOI] [PubMed] [Google Scholar]
  • 38.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. doi: 10.1055/s-2007-979086. [DOI] [PubMed] [Google Scholar]
  • 39.Bramer SL, Forbes WP, Mallikaarjun S. Cilostazol pharmacokinetics after single and multiple oral doses in healthy males and patients with intermittent claudication resulting from peripheral arterial disease. Clin Pharmacokinet. 1999;37(Suppl. 2):1–11. doi: 10.2165/00003088-199937002-00001. [DOI] [PubMed] [Google Scholar]
  • 40.Suri A, Forbes WP, Bramer SL. Pharmacokinetics of multiple-dose oral cilostazol in middle-age and elderly men and women. J Clin Pharmacol. 1998;38:144–50. doi: 10.1002/j.1552-4604.1998.tb04403.x. [DOI] [PubMed] [Google Scholar]
  • 41.Akiyama H, Kudo S, Shimizu T. The metabolism of a new antithrombotic and vasodilating agent, cilostazol, in rat, dog and man. Arzneimittelforschung. 1985;35:1133–40. [PubMed] [Google Scholar]
  • 42.Yoo HD, Cho HY, Lee YB. Population pharmacokinetic analysis of cilostazol in healthy subjects with genetic polymorphisms of CYP3A5, CYP2C19 and ABCB1. Br J Clin Pharmacol. 2010;69:27–37. doi: 10.1111/j.1365-2125.2009.03558.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.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. doi: 10.1159/000022512. [DOI] [PubMed] [Google Scholar]
  • 44.Wilhite DB, Comerota AJ, Schmieder FA, Throm RC, Gaughan JP, Rao AK. Managing PAD with multiple platelet inhibitors: the effect of combination therapy on bleeding time. J Vasc Surg. 2003;38:710–3. doi: 10.1016/s0741-5214(03)01029-2. [DOI] [PubMed] [Google Scholar]
  • 45.Ikeda Y, Kikuchi M, Murakami H, Satoh K, Murata M, Watanabe K, Ando Y. Comparison of the inhibitory effects of cilostazol, acetylsalicylic acid and ticlopidine on platelet functions ex vivo: randomized, double-blind cross-over study. Arzneimittelforschung. 1987;37:563–6. [PubMed] [Google Scholar]
  • 46.Comerota AJ. Effect on platelet function of cilostazol, clopidogrel, and aspirin, each alone or in combination. Atheroscler. 2005;6(Suppl.):13–9. doi: 10.1016/j.atherosclerosissup.2005.09.005. [DOI] [PubMed] [Google Scholar]
  • 47.Watanabe K, Nakase H, Kimura Y. Effect of cilostazol on experimental cerebral infarction in rabbits. Arzneimittelforschung. 1986;36:1022–4. [PubMed] [Google Scholar]
  • 48.Kohda N, Tani T, Nakayama S, Adachi T, Marukawa K, Ito R, Ishida K, Matsumoto Y, Kimura Y. Effect of cilostazol, a phosphodiesterase III inhibitor, on experimental thrombosis in the porcine carotid artery. Thromb Res. 1999;96:261–8. doi: 10.1016/s0049-3848(99)00109-7. [DOI] [PubMed] [Google Scholar]
  • 49.Kim CW, Yun JW, Bae IH, Park YH, Jeong YS, Park JW, Chung JH, Park YH, Lim KM. Evaluation of anti-platelet and anti-thrombotic effects of cilostazol with PFA-100(®) and Multiplate(®) whole blood aggregometer in vitro, ex vivo and FeCl(3)-induced thrombosis models in vivo. Thromb Res. 2011;127:656–70. doi: 10.1016/j.thromres.2011.02.004. [DOI] [PubMed] [Google Scholar]
  • 50.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. doi: 10.1016/s0002-9149(02)02869-2. [DOI] [PubMed] [Google Scholar]
  • 51.Regensteiner JG, Ware JE, Jr, McCarthy WJ, Zhang P, Forbes WP, Heckman J, Hiatt WR. 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. doi: 10.1046/j.1532-5415.2002.50604.x. [DOI] [PubMed] [Google Scholar]
  • 52.Gotoh F, Tohgi H, Hirai S, Terashi A, Fukuuchi Y, Otomo E, Shinohara Y, Itoh E, Matsuda T, Sawada T, Yamaguchi T, Nishimaru K, Ohashi Y. Cilostazol Stroke Prevention Study: a placebo-controlled double-blind trial for secondary prevention of cerebral infarction. J Stroke Cerebrovasc Dis. 2000;9:147–57. doi: 10.1053/jscd.2000.7216. [DOI] [PubMed] [Google Scholar]
  • 53.Shinohara Y, Katayama Y, Uchiyama S, Yamaguchi T, Handa S, Matsuoka K, Ohashi Y, Tanahashi N, Yamamoto H, Genka C, Kitagawa Y, Kusuoka H, Nishimaru K, Tsushima M, Koretsune Y, Sawada T, Hamada C. CSPS 2 group. Cilostazol for prevention of secondary stroke (CSPS 2): an aspirin-controlled, double-blind, randomised non-inferiority trial. Lancet Neurol. 2010;90:959–68. doi: 10.1016/S1474-4422(10)70198-8. [DOI] [PubMed] [Google Scholar]
  • 54.Uchiyama S, Demaerschalk BM, Goto S, Shinohara Y, Gotoh F, Stone WM, Money SR, Kwon SU. Stroke prevention by cilostazol in patients with atherothrombosis: meta-analysis of placebo-controlled randomized trials. J Stroke Cerebrovasc Dis. 2009;18:482–90. doi: 10.1016/j.jstrokecerebrovasdis.2009.07.010. [DOI] [PubMed] [Google Scholar]
  • 55.Han Y, Li Y, Wang S, Jing Q, Wang Z, Wang D, Shu Q, Tang X. Cilostazol in addition to aspirin and clopidogrel improves long-term outcomes after percutaneous coronary intervention in patients with acute coronary syndromes: a randomized, controlled study. Am Heart J. 2009;157:733–9. doi: 10.1016/j.ahj.2009.01.006. [DOI] [PubMed] [Google Scholar]
  • 56.Suh JW, Lee SP, Park KW, Lee HY, Kang HJ, Koo BK, Cho YS, Youn TJ, Chae IH, Choi DJ, Rha SW, Bae JH, Kwon TG, Bae JW, Cho MC, Kim HS. Multicenter randomized trial evaluating the efficacy of cilostazol on ischemic vascular complications after drug-eluting stent implantation for coronary heart disease: results of the CILON-T (Influence of CILostazol-based triple antiplatelet therapy ON ischemic complication after drug-eluting stenT implantation) trial. J Am Coll Cardiol. 2011;57:280–9. doi: 10.1016/j.jacc.2010.08.631. [DOI] [PubMed] [Google Scholar]
  • 57.Weintraub WS, Foster J, Culler SD, Becker ER, Parker K, Zhang Z, Kolm P, Douglas JS, Jr, Cilostazol for RESTenosis trial Cilostazol for RESTenosis trial: methods for the economic and quality of life supplement to the cilostazol for RESTenosis (CREST) trial. J Invasive Cardiol. 2004;16:257–9. [PubMed] [Google Scholar]
  • 58.Ge J, Han H, Jiang H, Sun B, Chen J, Zhang S, Du Z. RACTS (Randomized Prospective Antiplatelet Trial of Cilostazol Versus Ticlopidine in Patients Undergoing Coronary Stenting) Trial Investigators 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. doi: 10.1097/01.fjc.0000167012.82930.8f. [DOI] [PubMed] [Google Scholar]
  • 59.Yamagami H, Sakai N, Matsumaru Y, Sakai C, Kai Y, Sugiu K, Fujinaka T, Matsumoto Y, Miyachi S, Yoshimura S, Hyogo T, Kuwayama N, Hyodo A. Periprocedural cilostazol treatment and restenosis after carotid artery stenting: the retrospective study of in-stent restenosis after carotid artery stenting (ReSISteR-CAS) J Stroke Cerebrovasc Dis. 2010 doi: 10.1016/j.jstrokecerebrovasdis.2010.06.007. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 60.Jennings DL, Kalus JS. Addition of cilostazol to aspirin and a thienopyridine for prevention of restenosis after coronary artery stenting: a meta-analysis. Clin Pharmacol. 2010;50:415–21. doi: 10.1177/0091270009338940. [DOI] [PubMed] [Google Scholar]
  • 61.Takigawa T, Matsumaru Y, Hayakawa M, Nemoto S, Matsumura A. Cilostazol reduces restenosis after carotid artery stenting. J Vasc Surg. 2010;51:51–6. doi: 10.1016/j.jvs.2009.08.040. [DOI] [PubMed] [Google Scholar]
  • 62.Sorkin EM, Markham A. Cilostazol. Drugs Aging. 1999;14:63–71. doi: 10.2165/00002512-199914010-00005. [DOI] [PubMed] [Google Scholar]
  • 63.Barradas MA, Jagroop A, O'Donoghue S, Jeremy JY, Mikhailidis DP. Effect of milrinone in human platelet shape change, aggregation and thromboxane A2 synthesis: an in vitro study. Thromb Res. 1993;71:227–36. doi: 10.1016/0049-3848(93)90097-8. [DOI] [PubMed] [Google Scholar]
  • 64.Anfossi G, Massucco P, Piretto V, Mularoni E, Cavalot F, Mattiello L, Trovati M. Interplay between milrinone and adenosine in the inhibition of human platelet response. Gen Pharmacol. 1996;27:1149–54. doi: 10.1016/s0306-3623(96)00050-x. [DOI] [PubMed] [Google Scholar]
  • 65.Manns JM, Brenna KJ, Colman RW, Sheth SB. Differential regulation of human platelet responses by cGMP inhibited and stimulated cAMP phosphodiesterases. Thromb Haemost. 2002;87:873–9. [PubMed] [Google Scholar]
  • 66.Colucci WS. Cardiovascular effects of milrinone. Am Heart J. 1991;121:1945–7. doi: 10.1016/0002-8703(91)90829-7. [DOI] [PubMed] [Google Scholar]
  • 67.Born GVR, Cross MJ. Inhibition of the aggregation of blood platelets by substances related to adenosine diphosphate. J Physiol. 1963;166:29P–30P. [Google Scholar]
  • 68.Elkeles RS, Hampton JR, Honour AJ, Mitchell JR Prichard JS. Effect of a pyrido-pyrimidine compound on platelet behaviour in vitro and in vivo. Lancet. 1968;2:751–4. doi: 10.1016/s0140-6736(68)90952-5. [DOI] [PubMed] [Google Scholar]
  • 69.Ahn HS, Crim W, Romano M, Sybertz E, Pitts B. Effects of selective inhibitors on cyclic nucleotide phosphodieterase of rabbit aorta. Biochem Pharmacol. 1989;38:3331–9. doi: 10.1016/0006-2952(89)90631-x. [DOI] [PubMed] [Google Scholar]
  • 70.Schwartz L, Bourassa G, Lesperance J, Eastwood C, Kazim F. Aspirin and dipyridamole in the prevention of restenosis after percutaneous transluminal coronary angioplasty. N Engl J Med. 1988;318:1714–9. doi: 10.1056/NEJM198806303182603. [DOI] [PubMed] [Google Scholar]
  • 71.Klabunde RE. Dipyridamole inhibition of adenosine metabolism in human blood. Eur J Pharmacol. 1993;93:21–6. doi: 10.1016/0014-2999(83)90026-2. [DOI] [PubMed] [Google Scholar]
  • 72.Gresele P, Zoja C, Deckmyn H, Arnout J, Vermylen J, Verstraete M. Dipyridamole inhibits platelet aggregation in whole blood. Thromb Haemost. 1983;30:852–6. [PubMed] [Google Scholar]
  • 73.Gresele P, Arnout J, Deckmyn H, Vermylen J. Mechanism of the antiplatelet action of dipyridamole in whole blood: modulation of adenosine concentration and activity. Thromb Haemost. 1986;55:12–8. [PubMed] [Google Scholar]
  • 74.De La Cruz JP, Garcia PJ, Sanchez F, Cuesta F. Dipyridamole inhibits platelet aggregation induced by oxygen-derived free radicals. Thromb Res. 1992;66:277–85. doi: 10.1016/0049-3848(92)90278-i. [DOI] [PubMed] [Google Scholar]
  • 75.Perez Requeio JL, Santos MT, Valles J, Aznar J. Antiplatelet activity of dipirydamole in non anticoagulated whole blood. Thromb Res. 1988;52:279–86. doi: 10.1016/0049-3848(88)90069-2. [DOI] [PubMed] [Google Scholar]
  • 76.Muller TH, Su CA, Weisemberg H, Brickl R, Nehmiz G, Eisert WG. Dipyridamole alone or combined with low-dose acetylsalicylic acid inhibits platelet aggregation in human whole blood ex vivo. Br J Clin Pharmacol. 1990;30:179–86. doi: 10.1111/j.1365-2125.1990.tb03763.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Saniabadi AR, Fisher TC, McLaren M, Belch JF, Forbes CD. Effects of dipyridamole alone or in combination with aspirin on whole blood platelet aggregation, PGI2 generation, and red cell deformability ex vivo in man. Cardiovasc Res. 1991;25:177–83. doi: 10.1093/cvr/25.3.177. [DOI] [PubMed] [Google Scholar]
  • 78.Soderback U, Sollevi A, Wallen NH, Larsson PT, Hjemdhal P. Anti-aggregatory effects of physiological concentrations of adenosine in human whole blood as assessed by filtragometry. Clin Sci. 1991;81:691–4. doi: 10.1042/cs0810691. [DOI] [PubMed] [Google Scholar]
  • 79.Harker LA, Kadatz RA. Mechanism of action of dipyridamole. Thromb Res. 1983;4(Suppl.):39–46. doi: 10.1016/0049-3848(83)90356-0. [DOI] [PubMed] [Google Scholar]
  • 80.Sakuma I, Akaishi Y, Fukao M, Makita Y, Makita MA, Kobayashi T, Matsuno K, Miyazaki T, Yasuda H. Dipyridamole potentiates the anti-aggregating effect of endothelium-derived relaxing factor. Thromb Res. 1990;60:87–90. doi: 10.1016/0049-3848(90)90444-h. [DOI] [PubMed] [Google Scholar]
  • 81.Aktas B, Utz A, Hoenig-Liedl P, Walter U, Geiger J. Dipyridamole enhances NO/cGMP-mediated vasodilator-stimulated phosphoprotein phosphorylation and signaling in human platelets: in vitro and in vivo/ex vivo studies. Stroke. 2003;34:764–9. doi: 10.1161/01.STR.0000056527.34434.59. [DOI] [PubMed] [Google Scholar]
  • 82.Utz A, Aktas B, Honig-Liedl P, Walter U, Geriger J. Dipyridamole effects at physiolgical plasma concentrations on platelets in vitro and in vivo. Naunyn Schmiedebergs Arch Pharmacol. 2001;363:3. [Google Scholar]
  • 83.Iuliano L, Colavita AR, Camastra P, Bello V, Quintarelli C, Alessandroni M, Piovella F, Violi F. Protection of low density lipoprotein oxidation at chemical and cellular level by the antioxidant drug dipyridamole. Br J Pharmacol. 1996;119:1438–43. doi: 10.1111/j.1476-5381.1996.tb16056.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Pascual C, Romay C. Effect of antioxidant and chemiluminescence produced by reactive oxygen species. J Biolumin Chemilumin. 1992;7:123–32. doi: 10.1002/bio.1170070205. [DOI] [PubMed] [Google Scholar]
  • 85.Gresele P, Arnout J, Vermylen J. Dipyridamole inhibits leukotriene B4 synthesis. Thromb Haemost. 1987;57:235. [PubMed] [Google Scholar]
  • 86.Deckmyn H, Gresele P, Arnout J, Todisco A, Vermylen J. Prolonging prostacyclin production by nafazatrom or dipyridamole. Lancet. 1984;2:410–1. doi: 10.1016/s0140-6736(84)90582-8. [DOI] [PubMed] [Google Scholar]
  • 87.Deckmyn H, Zoja D, Arnout J, Todisco A, Vanden Bulcke F, D'Hondt L, Hendrickx N, Gresele P, Vermylen J. Partial isolation and function of the prostacyclin regulating plasma factor. Clin Sci. 1985;69:383–93. doi: 10.1042/cs0690383. [DOI] [PubMed] [Google Scholar]
  • 88.Chakrabarti S, Freedman JE. Dipyridamole, cerebrovascular disease, and the vasculature. Vasc Pharmacol. 2008;48:143–9. doi: 10.1016/j.vph.2007.12.004. [DOI] [PubMed] [Google Scholar]
  • 89.Kim HH, Liao JK. Translational therapeutics of dipyridamole. Arterioscler Thromb Vasc Biol. 2008;28:s39–42. doi: 10.1161/ATVBAHA.107.160226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Eisert WG. Near-field amplification of antithrombotic effects of dipyridamole through vessel wall cells. Neurology. 2001;57:S20–3. doi: 10.1212/wnl.57.suppl_2.s20. [DOI] [PubMed] [Google Scholar]
  • 91.Iimura O, Kusano E, Amemiya M, Muto S, Ikeda U, Shimada K, Asano Y. Dipyridamole enhances interleukin 1 beta stimulated nitric oxide production by cultured rat vascular smooth muscle cells. Eur J Pharmacol. 1996;296:319–26. doi: 10.1016/0014-2999(95)00712-1. [DOI] [PubMed] [Google Scholar]
  • 92.Weyrich AS, Denis MM, Kuhlmann-Eyre JR, Spencer ED, Dixon DA, Marathe GK, McIntyre TM, Zimmerman GA, Prescott SM. Dipyridamole selectively inhibits inflammatory gene expression in platelet-monocyte aggregates. Circulation. 2005;111:633–42. doi: 10.1161/01.CIR.0000154607.90506.45. [DOI] [PubMed] [Google Scholar]
  • 93.Mattiello T, Guerriero R, Lotti LV, Trifirò E, Felli MP, Barbarulo A, Pucci B, Gazzaniga P, Gaudio C, Frati L, Pulcinelli FM. Aspirin extrusion from human platelets though multidrug resistance protein-4 mediated transport: evidence of a reduced drug action in patients after coronary artery bypass grafting. J Am Coll Cardiol. 2011 doi: 10.1016/j.jacc.2011.03.049. in press. [DOI] [PubMed] [Google Scholar]
  • 94.Gresele P, Arnout J, Deckmyn H, Vermylen J. Combining antiplatelet agents: potentiation between aspirin and dipyridamole. Lancet. 1985;1:937–8. doi: 10.1016/s0140-6736(85)91718-0. [DOI] [PubMed] [Google Scholar]
  • 95.Diener HC, Cunha L, Forbes C, Sivenius J, Smets P, Lowenthal A. European Stroke Prevention Study 2. Dipyridamole and acetylsalicylic acid in the secondary prevention of stroke. J Neurol Sci. 1996;143:1–13. doi: 10.1016/s0022-510x(96)00308-5. [DOI] [PubMed] [Google Scholar]
  • 96.Halkes PH, van Gijn J, Kappelle LJ, Koudstaal PJ, Algra A, ESPRIT Study Group Aspirin plus dipyridamole versus aspirin alone after cerebral ischaemia of arterial origin (ESPRIT): randomised controlled trial. Lancet. 2006;367:1665–73. doi: 10.1016/S0140-6736(06)68734-5. [DOI] [PubMed] [Google Scholar]
  • 97.Weber E, Haas TA, Muller TH, Eisert WG, Hirsh J, Richardson M, Buchanan MR. Relationship between vessel wall 13-HODE synthesis and vessel wall thrombogenicity following injury: influence of salicylate and dipyridamole treatment. Thromb Res. 1990;57:383–92. doi: 10.1016/0049-3848(90)90254-a. [DOI] [PubMed] [Google Scholar]
  • 98.Van Ryn JM, Lorenz M, Merk H, Buchanan MR, Eisert WG. Accumulation of radiolabelled platelets and fibrin on the carotid artery of rabbits after angioplasty: effects of heparin and dipyridamole. Thromb Haemost. 2003;90:1179–86. doi: 10.1160/TH03-05-0305. [DOI] [PubMed] [Google Scholar]
  • 99.Venkatesh PK, Pattillo CB, Branch B, Hood J, Thoma S, Illum S, Pardue S, Teng X, Patel RP, Kevil CG. Dipyridamole enhances ischaemia-induced arteriogenesis through an endocrine nitrite/nitric oxide-dependent pathway. Cardiovasc Res. 2010;85:661–70. doi: 10.1093/cvr/cvq002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Verro P, Gorelick PB, Nguyen D. Aspirin plus dipyridamole versus aspirin for prevention of vascular events after stroke or TIA: a meta-analysis. Stroke. 2008;39:1358–63. doi: 10.1161/STROKEAHA.107.496281. [DOI] [PubMed] [Google Scholar]
  • 101.Adams RJ, Albers G, Alberts MJ, Benavente O, Furie K, Goldstein LB, Gorelick P, Halperin J, Harbaugh R, Johnston SC, Katzan I, Kelly-Hayes M, Kenton EJ, Marks M, Sacco RL, Schwamm LH. American Heart Association; American Stroke Association. Update to the AHA/ASA recommendations for the prevention of stroke in patients with stroke and transient ischemic attack. Stroke. 2008;39:1647–52. doi: 10.1161/STROKEAHA.107.189063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Sacco RL, Diener HC, Yusuf S, Cotton D, Ounpuu S, Lawton WA, Palesch Y, Martin RH, Albers GW, Bath P, Bornstein N, Chan BP, Chen ST, Cunha L, Dahlöf B, De Keyser J, Donnan GA, Estol C, Gorelick P, Gu V, Hermansson K, Hilbrich L, Kaste M, Lu C, Machnig T, Pais P, Roberts R, Skvortsova V, Teal P, Toni D, Vandermaelen C, Voigt T, Weber M, Yoon BW, PRoFESS Study Group Aspirin and extended-release dipyridamole versus clopidogrel for recurrent stroke. N Engl J Med. 2008;359:1238–51. doi: 10.1056/NEJMoa0805002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Diener HC, Sacco RL, Yusuf S, Cotton D, Ounpuu S, Lawton WA, Palesch Y, Martin RH, Albers GW, Bath P, Bornstein N, Chan BP, Chen ST, Cunha L, Dahlöf B, De Keyser J, Donnan GA, Estol C, Gorelick P, Gu V, Hermansson K, Hilbrich L, Kaste M, Lu C, Machnig T, Pais P, Roberts R, Skvortsova V, Teal P, Toni D, VanderMaelen C, Voigt T, Weber M, Yoon BW, Prevention Regimen for Effectively Avoiding Second Strokes (PRoFESS) study group Effects of aspirin plus extended-release dipyridamole versus clopidogrel and telmisartan on disability and cognitive function after recurrent stroke in patients with ischaemic stroke in the Prevention Regimen for Effectively Avoiding Second Strokes (PRoFESS) trial: a double-blind, active and placebo-controlled study. Lancet Neurol. 2008;7:875–84. doi: 10.1016/S1474-4422(08)70198-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Stein PD, Alpert JS, Bussey HI, Dalen JE, Turpie AG. Antithrombotic therapy in patients with mechanical and biological prosthetic heart valves. Chest. 2001;119:220S–7S. doi: 10.1378/chest.119.1_suppl.220s. [DOI] [PubMed] [Google Scholar]
  • 105.De Schryver EL, Algra A, van Gijn J. Dipyridamole for preventing stroke and other vascular events in patients with vascular disease. Cochrane Database Syst Rev. 2006;(2) doi: 10.1002/14651858.CD001820.pub2. CD001820. 1–43. [DOI] [PubMed] [Google Scholar]
  • 106.Murray KJ. Phosphodiesterase Va inhibitors. Drug News Perspect. 1993;6:150–6. [Google Scholar]
  • 107.Rudd RM, Gellert AR, Studdy PR, Geddes DM. Inhibition of exercise induced asthma by an orally absorbed mast cell stabilizer (M&B22948) Br J Dis Chest. 1983;77:78–86. [PubMed] [Google Scholar]
  • 108.Dunkern TR, Hatzelmann A. The effect of Sildenafil on human platelet secretory function is controlled by a complex interplay between phosphodiesterases 2, 3 and 5. Cell Signal. 2005;17:331–9. doi: 10.1016/j.cellsig.2004.07.007. [DOI] [PubMed] [Google Scholar]
  • 109.Schwartz BG, Kloner RA. Drug interactions with phosphodiesterase-5 inhibitors used for the treatment of erectile dysfunction or pulmonary hypertension. Circulation. 2010;122:88–95. doi: 10.1161/CIRCULATIONAHA.110.944603. [DOI] [PubMed] [Google Scholar]
  • 110.Wallis RM, Corbin JD, Francis SH, Ellis P. Tissue distribution of phosphodiesterase families and the effects of sildenafil on tissue cyclic nucleotides, platelet function, and the contractile responses of trabeculae carneae and aortic rings in vitro. Am J Cardiol. 1999;83:3C–12C. doi: 10.1016/s0002-9149(99)00042-9. [DOI] [PubMed] [Google Scholar]
  • 111.Cheitlin MD, Hutter AM, Jr, Brindis RG, Ganz P, Kaul S, Russell RO, Jr, Zusman RM. Use of sildenafil (Viagra) in patients with cardiovascular disease. Technology and practice executive committee. Circulation. 1999;99:168–77. doi: 10.1161/01.cir.99.1.168. [DOI] [PubMed] [Google Scholar]
  • 112.Berkels R, Klotz T, Sticht G, Englemann U, Klaus W. Modulation of human platelet aggregation by the phosphodiesterase type 5 inhibitor sildenafil. J Cardiovasc Pharmacol. 2001;37:413–21. doi: 10.1097/00005344-200104000-00008. [DOI] [PubMed] [Google Scholar]
  • 113.Halcox JP, Nour KR, Zalos G, Mincemoyer RA, Waclawiw M, Rivera CE, Willie G, Ellahham S, Quyyumi AA. The effect of sildenafil on human vascular function, platelet activation, and myocardial ischemia. J Am Coll Cardiol. 2002;40:1232–40. doi: 10.1016/s0735-1097(02)02139-3. [DOI] [PubMed] [Google Scholar]
  • 114.Galie N, Rubin LJ, Simonneau G. Phosphodiesterase inhibitors for pulmonary hypertension. N Engl J Med. 2010;362:559–60. [PubMed] [Google Scholar]
  • 115.Young JM. Expert opinion: vardenafil. Expert Opin Investig Drugs. 2002;1:1487–96. doi: 10.1517/13543784.11.10.1487. [DOI] [PubMed] [Google Scholar]
  • 116.Corbin JD, Francis SH. Pharmacology of phosphodiesterase-5 inhibitors. Int J Clin Pract. 2002;56:453–9. [PubMed] [Google Scholar]
  • 117.De Bon E, Bonanni G, Saggiorato G, Bassi P, Cella G. Effects of tadalafil on platelets and endothelium in patients with erectile dysfunction and cardiovascular risk factors: a pilot study. Angiology. 2010;61:602–6. doi: 10.1177/0003319710362977. [DOI] [PubMed] [Google Scholar]
  • 118.Gresele P, Falcinelli E, Momi S. Potentiation and priming of platelet activation: a potential target for antiplatelet therapy. Trends Pharmacol Sci. 2008;29:352–60. doi: 10.1016/j.tips.2008.05.002. [DOI] [PubMed] [Google Scholar]
  • 119.Curtin R, FitzGerald DJ. A cold start for oral glycoprotein IIb/IIIa antagonists. Eur Heart J. 2000;21:1992–4. doi: 10.1053/euhj.2000.2458. [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Clinical Pharmacology are provided here courtesy of British Pharmacological Society

RESOURCES