Quick viewing(Text Mode)

Platelets and New Antiplatelet Drugs

Platelets and New Antiplatelet Drugs

REVIEW

Platelets and new antiplatelet

Rosemarie A Reiter & play an important, life-saving role in and clotting at sites of Bernd Jilma† vascular injury. However, unwanted activation and arterial formation are †Author for correspondence implicated in the onset of , and other cardiovascular diseases. Medical University of Vienna, Department of Clinical Different mechanisms, such as vascular damage, the development of mural platelet thrombi , Währinger as a response to injury and the biochemical effects of intraplatelet substances that are Gürtel 18–20, A-1090 released in response to damage, may be involved. Thus, antiplatelet therapy has become a Vienna, Austria Tel.: +43 140 400 2981 mainstay of treatment for these conditions and the benefit of antiplatelet drugs is Fax: +43 140 400 2998 documented across a wide spectrum of clinical conditions. has been regarded as the [email protected] prototype antiplatelet and is still the most widely used agent. Aspirin’s antiplatelet effect is directly due to irreversible inactivation of arachidonic and suppression of

A2 synthesis. However, platelet activation occurs via several pathways that do not rely on amplification by released . Therefore, a number of other compounds have been developed to complement the beneficial effect of aspirin. Four main classes of antiplatelet agents are currently available for clinical use: aspirin, phosphodiesterase

inhibitors, and the platelet glycoprotein αIIbβ3 receptor antagonists. This review discusses state-of-the-art antiplatelet therapies and recent advances, using aspirin as the reference standard.

Platelets, & receptors. Moreover, platelet activation allows atherothrombotic vascular disease interplatelet contact and the formation of plate- Arterial thrombosis rarely occurs in the presence let aggregates. This process is mediated by the of a healthy or undamaged vascular wall of the integrin-αIIbβIIIa (GPIIb/IIIa) complex in the active involvement of endothelial cells, which pro- platelet membrane, which acts as a promiscuous vide a thromboresistant surface for flowing receptor for several ligands such as fibrinogen, blood [1]. Platelet accumulation in the arterial wall vWF and fibronectin [7]. On the surface of the is currently well accepted as a rather early and key activated platelet, the GPIIb/IIIa receptor under- event, both in initiation and perpetuation of goes a conformational change [8], allowing plate- atherosclerotic lesions. lets to bind fibrinogen and thus crosslink, which One of the early events following vascular is important for the final common pathway of injury is the adhesion of circulating platelets to stable platelet aggregation. exposed subendothelium [2]. Both superficial intimal injury, caused by endothelial denuda- Platelet adhesion tion, and deep injury, caused by plaque rupture, Inhibition of GPIb–von Willebrand expose subendothelial collagen and von Wille- factor interaction brand factor (vWF) to platelets of the circulating The initial step of thrombus formation consists blood [3]. Platelet adhesion is mediated by the of platelet adhesion to an injured vessel wall. interaction of platelet glycoprotein (GP)Ib/IX Especially under the high shear-stress conditions receptor with the subendothelial vWF under observed in stenosed coronary , the Keywords: antiplatelet, aspirin, , high shear conditions [4] and by the platelet interaction between platelet GPIb and vWF GPIa/IIa receptor binding to collagen under plays a crucial role in platelet-mediated throm- glycoprotein iib/iiia, both stasis and flow [5]. This further triggers a bus formation [9]. Blocking GPIb [10–13] or vWF prevention, thrombosis network of signaling events inducing the release [14–16] with antibodies results in the inability of and local accumulation of soluble platelet ago- the platelets to attach to the exposed suben- nists (thromboxane [TX]A2, serotonin and ADP dothelium and, thus, the inhibition of the part of and ) [6] and further induces activation GPIb–vWF interaction has focused considera- and aggregation of platelets by activating specific ble attention as an attractive target for the

10.2217/14750708.2.3.465 © 2005 Future Medicine Ltd ISSN 1475-0708 http://www.futuremedicine.com Therapy (2005) 2(3), 465–502 465 REVIEW – Reiter & Jilma

prevention of thrombus formation in stenosed activation and aggregation [24,25]. The binding site arteries [13,16]. Moreover, pharmacologic block- for collagen on vWF is localized to the A3 domain ade of the GPIb–vWF interaction may show a and blocking this domain from binding to colla- lower bleeding risk than GPIIb/IIIa gen by a specific antibody reduced thrombus blockade [13,17]. As shown in a baboon model, growth in vivo and prolonged skin-bleeding time the combination of a low dose of a GPIb inhibi- [26]. Thus, collagen antagonists could be potent tor with a low-dose of a GPIIb/IIIa inhibitor has antiplatelet agents, targeting platelet adhesion by a potent action with minimal inhibiting the collagen–platelet interaction. How- effects on the bleeding time [13]. Given the ever, as reviewed recently, these selective antago- growing awareness of the importance of inflam- nists might have potential limitations as they mation in influencing the outcomes of cardio- could have limited antithrombotic protection in vascular disease [18], GPIb inhibitors that reduce patients with arterial thrombosis [27]. Indeed, it has platelet–leukocyte interactions might have addi- been shown that most patients with acute myocar- tional therapeutic benefits. To summarize, anti- dial infarction (MI) have plaque rupture and non- GPIb and anti-vWF may be useful compounds occlusive thrombus formation up to a week before in the therapy of thrombosis and cardiovascular the clinical event [28] and, thus, collagen antago- disease. However, the clinical use of GPIb inhib- nists might be restricted to specific subsets of itors could be limited by potential pathogenic patients with vascular disease. effects on megakaryocytes [19,20] and antibody- induced thrombocytopenia [11,21] and, moreo- Platelet aggregation ver, only rare data on the use of such inhibitors Aspirin & related inhibitors in clinical trials are currently available [22]. or thromboxane antagonists Aspirin Inhibition of collagen–platelet interactions Aspirin has been regarded as the prototype Not only the interaction between platelet GPIb , with multiple dose-depend- and vWF but also the interaction between colla- ent therapeutic effects, and is still the most gen and vWF plays a crucial role under conditions widely used and studied agent. Aspirin irrevers- of high shear rates, which is typically found in the ibly inhibits arachidonate cyclooxygenase arteriolar circulation and at sites of arterial stenosis (COX) activity in platelets, thereby reducing [23]. Collagen, the most thrombogenic component the extent of TXA2 formation that occurs after of the extracellular matrix, directly binds to recep- activation of phospholipase A2 and release of tors that mediate platelet adhesion, and induces (AA) (Figure 1) [29]. However, although aspirin effectively reduces plate- Table 1. Antiplatelet agents. let secretion and aggregation, it is a relatively weak platelet inhibitor [30]. Other platelet-dependent Pathways Agents prothrombotic mechanisms are less affected or not Platelet adhesion modified at all at doses that block platelet-depend- von Willebrand factor GPlb-receptor antibody, von Willebrand ent TX formation. For example, aspirin does not factor antibody inhibit shear stress-induced platelet activation and Platelet aggregation adhesion [31], does not inhibit α-granule secretion Cyclooxygenase pathway in response to ADP and other agonists, and does • Cyclooxygenase Aspirin, NO-aspirin, gingerols not inhibit fibrinogen binding [32,33]. Moreover, • Thromboxane A2 synthase Dazoxiban aspirin usage is associated with potentially life- threatening side effects such as gastric hemorrhage • Thromboxane A2 receptor Vapiprost [34]. Thus, researchers have been encouraged to • Thromboxane synthase and Ridogrel, , investigate and develop new antiplatelet drugs that receptor are equivalent in strength to aspirin, but with • , , ebaprostanol fewer, or no, adverse events (Table 1). Phosphodiesterase Dipyridamole and ADP receptor antagonists and P2Y1 Acute therapy & secondary prevention of receptor antagonists cardiovascular disease & stroke GPllb/llla Murine– human antibodies, synthetic Since the 1950s when it was recognized that aspi- peptide forms and synthetic nonpeptide rin could reduce the incidence of MI [35], multiple, forms randomized, controlled clinical trials have shown a GP: Glycoprotein; NO: Nitric oxide. clinically significant decrease in cardiovascular

466 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

Figure 1. Platelet receptors and their antagonists.

Fibrinogen ADP GPIIb/IIIa antagonists

Thienopyridines

Thromboxane A2

αIIbβ3 P2Y12 Gi

Gq Inhibition

G12/13 Activation Asprin Phospholipase C Adenylate of GP ↑ Ca2+ cyclase IIb/IIIa receptor

Arachidonic Thromboxane A2 ↓ cAMP acid

Cyclooxygenase Platelet aggregation Promotion Final common pathway 5´AMP cAMP of platelet of platelet aggregation 5´GMP cGMP aggregation and plug stabilization

Phosphodiesterase inhibitors

Thromboxane A2 is produced from arachidonic acid via cyclooxygenase and is released from the platelet to work with the thromboxane receptor. The thromboxane receptor (a G-protein coupled receptor) causes platelet aggregation via both Gq and G12/13. Aspirin irreversibly inhibits cyclooxygenase, for the lifetime of the platelets, preventing thromboxane production. Phosphodiesterase inhibitors prevent the breakdown of cAMP and cGMP. Therefore cAMP and cGMP levels are maintained inhibiting platelet aggregation. The ADP

P2Y12 receptor (a G-protein coupled receptor) is linked to adenylate cyclase via Gi. Upon stimulation the adenylate cyclase is inhibited, leading to a fall in cAMP and promotion of platelet aggregation. Thienopyridines irreversibly bind to the ADP P2Y12 receptor preventing the inhibition of adenylate cyclase, therefore maintaining cAMP levels and inhibiting platelet aggregation. The GPIIb/IIIa antagonists

prevent fibrinogen binding to the IIbß3 receptors and therefore inhibit the ‘final common pathway of platelet aggregation’. cAMP: Cyclic adenosine monophosphate; cGMP: Cyclic guanosine monophosphate. morbidity and mortality in patients at risk of randomized studies of various antiplatelet recurrent atherothrombotic events [36–39]. regimes, which demonstrated that aspirin caused a The most accurate data regarding the efficacy 25% reduction in cardiovascular outcomes (non- of aspirin comes from the Antiplatelet Trialists’ fatal MI, or vascular death) in patients Collaboration (ATC), a meta-analysis of 287 with pre-existing cardiovascular disease [39]. The

www.futuremedicine.com 467 REVIEW – Reiter & Jilma

Second International Study of Infarct Survival less clear. A meta-analysis of five randomized con- (ISIS-2) has established the benefit in acute trolled trials of aspirin for primary prevention MI [36,37]. Patients (n = 17187) with acute MI demonstrated that aspirin was associated with a were randomized to one of four arms of therapy statistically significant, 32%, reduction in the risk consisting of , aspirin, or of a first MI and a significant, 15%, reduction in streptokinase plus aspirin. At the end of 5 weeks, the risk of all important vascular events, but had patients receiving aspirin therapy alone had a sig- no significant effects on nonfatal stroke or vascu- nificant (23%) reduction in vascular mortality lar death [46,47]. Interestingly, the absolute benefit and a nearly 50% reduction in the risk of nonfatal of aspirin clearly increases with the risk of cardio- reinfarction and nonfatal stroke. Administration vascular events in the treatment group [48]. Addi- of streptokinase alone was associated with a 25% tionally, aspirin increases the risk for hemorrhagic reduction in vascular deaths, and the combination strokes and major gastrointestinal bleeding [46]. of streptokinase and aspirin was significantly bet- Data regarding the use of aspirin for the primary ter than either agent alone (42% reduction in vas- prevention of strokes in high-risk patients are not cular mortality with combination therapy). encouraging [49,50]. Overall, low-dose aspirin Interestingly, the mortality benefit of combined appears to decrease the risk of MI in men with aspirin and streptokinase therapy was maintained little effect on the risk of stroke. after 10 years’ follow-up [37]. Since three of the previous five primary pre- In contrast to the secondary prevention of MI, vention trials [49–53] exclusively evaluated men, the therapeutic value of aspirin in preventing and fewer than 180 of the 2042 vascular events ischemic stroke is less clear and appears to be occurred in women, the currently published related to the cause and severity of cerebral results of the Women’s Health Study are of ischemia [40–42]. Two large, randomized trials of interest [54]. Indeed, the current recommenda- aspirin use in the setting of acute, ischemic tions for the use of aspirin in primary preven- stroke have demonstrated that the use of aspirin tion in women are based on limited direct data reduces both recurrent stroke and the combined from women [46,55,56] incidence of death or nonfatal stroke In the Women’s Health Study [54] there was a [43,44](Table 2). The relative risk reduction (RRR) nonsignificant RRR of 9% in the primary out- in fatal or nonfatal vascular events was only 10% come of first major cardiovascular events (i.e., in this setting. Accordingly, a meta-analysis MI, nonfatal stroke or death from a cardio- showed a modest 13% RRR of vascular events in vascular event). Regarding the individual end patients with cerebral ischemia of arterial points, women in the aspirin group had a 17% origin [45]. Overall, the benefit of aspirin in acute RRR (p = 0.04) in stroke, compared with the pla- stroke treatment and secondary prevention of cebo group, owing to a 24% RRR of ischemic stroke are definite but modest. stroke (p = 0.009) and a nonsignificant increase in the risk of hemorrhagic stroke. Moreover, aspi- Primary prevention of cardiovascular disease rin therapy was associated with a 22% RRR of & stroke transient ischemic attack (TIA) (p = 0.01) and a Although the beneficial effect of aspirin in the significant 19% RRR of nonfatal stroke secondary prevention of ischemic events is well (p = 0.02). However, compared with placebo, established [39], the role of primary prevention is aspirin had no significant effect on the risk of

Table 2. Trials of aspirin therapy in acute ischemic stroke [48].

End point Chinese acute stroke trial [43] International stroke trial [44] Aspirin (%) No aspirin (%) 2P Aspirin (%) No aspirin (%) 2P Death 3.3 3.9 0.04 9.0 9.4 NS Death and 5.3 5.9 0.03 11.3 12.4 <0.05 nonfatal CVA Recurrent 1.6 2.1 0.01 2.8 3.9 <0.001 CVA Hemorrhagic 1.1 0.9 NS 0.9 0.8 NS CVA Values are given as percentages with the exception of 2P values. CVA: Cerebrovascular accident. NS: Not significant.

468 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

fatal stroke, fatal or nonfatal MI or death from in patients with chronic stable angina (Table 3). cardiovascular causes. In a subgroup analysis, the Low-dose aspirin (75–150 mg) is an effective most consistent benefit of aspirin was observed antiplatelet regimen for long-term prevention of among the subgroup of women 65 years of age or serious vascular events, whereas in clinical situa- older at study entry. This subgroup showed a tions, where an immediate antithrombotic effect 26% RRR (p = 0.008) in major cardiovascular is required (such as acute MI, stroke or unstable events, a 30% RRR (p = 0.05) in ischemic stroke, angina pectoris), a loading dose of at least and a 34% RRR (p = 0.04) in MI. As expected, 150 to 325 mg is recommended [39,48,60,63–65]. For the rates of any gastrointestinal (GI) bleeding primary prevention, no clear indications currently were higher in the aspirin group than in the pla- exist, although low-dose aspirin is recommended, cebo group (4.6 vs. 3.8%, p < 0.001) but without especially in those patients believed to be at high significantly more episodes of fatal GI bleeding. risk for the development of cardiovascular disease In summary, questions remain concerning the and stroke [48,55,61,65]. optimal therapeutic dose of aspirin, and concern- In conclusion, individual trial data shows sub- ing its use in the primary prevention of vascular stantial heterogeneity in the treatment of cardio- disease. Moreover, the reasons for any sex-based vascular/cerebrovascular diseases but the reason differences in the efficacy of aspirin for primary for these differences in aspirin therapy is still prevention are unclear and require further explo- unknown. However, increased platelet-depend- rations. In patients with a relatively low risk of ent TX generation may only occur in a minority developing cardiovascular (or cerebrovascular dis- (30%) of patients with acute ischemic ease), the risk of prophylactic aspirin therapy may stroke [66,67] and a variable importance of TXA2 be outweighed by the risk of hemorrhagic compli- as a mechanism to amplify the hemostatic cation. Conversely, in high-risk patients, the bene- response to plaque destabilization in different fits of therapy may outweigh the risks of clinical settings has been suggested [62]. Thus, one hemorrhagic complications. Indeed, recent stud- may speculate that the TX-mediated amplifica- ies have suggested that the indications for aspirin tion of platelet response to acute vascular injury use should be expanded to primary prevention in plays a more important role in cardiovascular populations at high risk, including diabetes, than in cerebrovascular diseases, which might carotid stenosis, peripheral vascular disease, end- therefore explain the superior benefit of aspirin in stage renal disease [47,57,58] or polycythemia the treatment of cardiovascular diseases. vera [59]. Notwithstanding these results, it remains essential to balance the cardio/cerebrovascular risk Optimal dose of aspirin profile against the risk of potential bleeding com- The overall results of clinical trials in which differ- plications for each patient (irrespective of gender) ent doses of aspirin have been tested indicate that when prescribing aspirin [46,54,60–62]. lower doses of aspirin decrease the prevalence of GI side effects [62]. A direct comparison on the preva- Recommendations for aspirin use lence of GI toxicity among patients using 300 and Overall, aspirin remains the background template 1200 mg aspirin once daily in the UK-TIA therapy, both for acute ischemic syndromes and trial [68] showed that both subjective GI com- secondary prevention after MI, stroke or TIA and plaints and GI bleeding were more frequent at 1200 mg/day than at 300 mg/day. In the 30-mg Table 3. Summary of recommended uses for aspirin. group of the Dutch TIA [69] trial, major bleeding complications (requiring hospital attendance) were Clinical indication Recommended dose slightly less common than in the 300-mg group Acute MI, stroke, or unstable Loading dose of at least 150 mg (RRR 23%) and significantly fewer minor bleeds angina pectoris (up to 325 mg) occurred (RRR 42%). Gastric discomfort or Secondary prevention after MI, Daily therapy with 75–150 mg unspecified side effects were reported significantly nonhemorrhagic stroke, or TIA less often with 30 mg than with 300 mg of aspirin. and in patients with chronic Another study comparing clopidogrel with placebo stable angina on top of a ‘background’ of aspirin therapy (aspirin Primary prevention No clear indication at this time. Consider doses ranging from 75 to 352 mg/day were used in therapy with 75–160 mg/day in patients the Clopidogrel in to prevent believed to be at high risk for the Recurrent ischemic Events [CURE] trial), demon- development of cardiovascular disease. strated that bleeding risk increased with increasing MI: Myocardial infarction; TIA: Transient ischemic attack. aspirin dose, with or without clopidogrel [70]. www.futuremedicine.com 469 REVIEW – Reiter & Jilma

In contrast, the accumulated evidence from inhibit endothelial COX, such as aspirin, may clinical trials makes it clear that aspirin doses reduce the synthesis of vasodilatory PGs. from 75 to 1300 mg do not (generally) alter the Accordingly, the inhibition of COX may clinical benefit [39] (Table 4). For example, The reduce the efficacy of ACE inhibition and Acetylsalicylic Acid and Carotid Endarterectomy therefore the safety of combination therapy trial [71] reported that the risk of the composite with aspirin and ACE inhibitors has been outcome of MI, stroke, or death within questioned because both drugs affect a related three months of carotid endarterectomy was sig- PG-mediated pathway. nificantly lower among patients taking 81 or However, the results of several trials are con- 325 mg aspirin daily than in those taking 625 to troversial concerning the negative interaction of 1300 mg. CURE investigators showed that the the combined therapy of ACE inhibitors with bleeding risk increased with increasing doses of aspirin. Post hoc analysis of two large, multi- aspirin, with or without clopidogrel, but without center trials have suggested that aspirin causes any increase in efficacy [70]. Moreover, aspirin blunting, or even complete abrogation, of the doses of less than 75 mg/day have been less benefit of ACE inhibitors on mortality in widely assessed than those of 75 to 1500 mg/day patients with failure [77,78]. In another ret- and seemed to have a somewhat smaller effect at rospective analysis, ACE inhibitors were associ- doses lower than 75 mg/day [39] (Table 4). ated with an increased mortality in patients who Summarizing, any effective antiplatelet dose took aspirin following percutaneous coronary of aspirin is associated with an increased risk of intervention (PCI) [79]. More recently, no evi- bleeding and thus, the individual benefit:risk dence of a negative therapeutic interaction ratio determines the dose of the compound and between aspirin and ACE inhibitors was found use of the lowest effective dose of aspirin (Table 3) in stable patients with chronic heart failure is probably the most rational strategy to related to left-ventricular systolic maximize efficacy and minimize toxicity. dysfunction [80]. Similar interactions have not been found in patients with MI [81,82] and the Negative effects of aspirin on negative hemodynamic effects may be seen with angiotensin-converting enzyme inhibitors high doses of aspirin only. Indeed, several stud- Aspirin and angiotensin-converting enzyme ies have found that a negative interaction with (ACE) inhibitors are widely used in combina- ACE inhibitors is present with high doses of tion to treat a wide spectrum of cardiac disor- aspirin (i.e., 325 mg) [79,83–85], but not with low ders. There is experimental evidence showing doses (i.e., 100 mg) [80,86,87]. Although the con- that ACE inhibitors limit the development of troversy has not yet been resolved, the recom- infarct size, reduce the incidence of ischemic mendation of low-dose aspirin in patients with and reperfusion arrhythmias, and enhance the chronic heart failure seems to be justified [80,88]. recovery of contractile function of stunned myocardium [72]. The cardioprotective effects Nitric oxide-releasing aspirin of ACE inhibitors are mediated by an attenu- As aspirin can cause severe damage to the stom- ated degradation of bradykinin [73,74]. Bradyki- ach, a nitric oxide (NO)-releasing derivate nin, a potent vasodilator on its own, activates (NCX-4016) has been developed that might have vascular endothelial B2-kinin receptors, which clinical promise in the protection from athero- promote the formation of vasodilatory prostag- sclerosis without the unwanted effects on the landins (PGs) through the action of phosphol- stomach. NO protects the gastric mucosa, ipase-A2 and COX [75,76]. Thus, drugs that induces vasodilatation, and inhibits platelet

Table 4. Aspirin doses reducing vascular events in high-risk patients*. Dose of aspirin (mg/day) Number of trials Number of patients Odds reduction‡ (%) <75 3 3.655 13 ± 8 75–150 12 6.776 32 ± 6 160–325 19 26.513 26 ± 3 500–1500 34 22.451 19 ± 3 *Data from Antithrombotic Trialists Collaboration [39]. ‡Data are presented as mean ± standard deviation.

470 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

aggregation, inflammation, cellular proliferation drugs should be used for cardioprotection in the and apoptosis through both a cyclic guanosine absence of evidence from randomized controlled monophosphate (cGMP)-dependent and -inde- trials to lend support to such a practice. The only pendent mechanism [89]. In experimental animal nonaspirin NSAIDs that have been tested for their studies, NO aspirin had antiatherosclerotic and antithrombotic efficacy in randomized trials are antioxidant effects in the arterial wall of hyperc- sulfinpyrazone, , and triflu- holesterol mice and inhibited restenosis after PCI sal; however, none of these drugs have been in rats [90,91]. In another rat model, NO-releasing approved as an antiplatelet drug since the overall aspirin reduced brain damage after focal cerebral results of these trials were not satisfactory [39,62]. ischemia, indicating that NO release associated Another interesting point of nonaspirin with aspirin confers neuroprotective effects NSAIDs is the fact that they may interact with the against ischemic injury [92]. cardioprotective effects of aspirin. Based on epide- Recently, a randomized, parallel-group, clini- miologic data, some people have recommended cal trial was designed to assess whether NO- avoiding in patients taking aspirin and releasing aspirin could have broader anti-inflam- suggested that ibuprofen reverses the cardioprotec- matory and antithrombotic effects, as well as tion offered by aspirin [99]. Indeed, results from an better gastric tolerability than aspirin [93]. A total in vivo study demonstrated an interaction between of 48 healthy subjects were randomized to aspirin and ibuprofen on platelet function, but no receive NO-releasing aspirin 800 mg twice daily, such interaction was reported with , NO-releasing aspirin mg twice daily plus aspirin or [100]. The hypothesis 325 mg, aspirin 325 mg, or placebo for 21 days. that ibuprofen may interact with the cardio- In this study, NO-releasing aspirin was equally protective effects of aspirin, at least in patients effective as aspirin in inhibiting COX activity with established cardiovascular disease, is a theory but caused less gastric damage. These beneficial that has been supported by the findings of a effects of NO-releasing aspirin, combined with recently published trial [101]. This study involved the inhibition of TXA2, are very interesting. As 7107 patients who received aspirin alone, aspirin atherogenesis and plaque rupture are points on and ibuprofen, aspirin plus diclofenac or aspirin the continuum of vascular inflammation, one plus other NSAIDs. Patients in the aspirin plus may speculate that the NO aspirin, NCX-4016, ibuprofen group had a significantly higher risk of could be superior to aspirin in the primary and all-cause mortality (p = 0.0011) and cardiovascu- secondary prevention of vascular events and in lar mortality (p = 0.0305) than those in the aspi- gastric tolerability. Furthermore, NO-releasing rin-alone group. No such increased risk was noted aspirin may offer an attractive alternative in in users of aspirin plus diclofenac or aspirin plus patients in whom COX inhibition with aspirin other NSAIDs. may be poorly tolerated. Thus, larger multi- However, as reviewed very recently [102] no center clinical trials are warranted in order to randomized, controlled trials addressing this par- establish the clinical efficacy and safety of the ticular issue exist at the moment and the data NO-releasing aspirin, NCX-4016. obtained from observational and epidemiologic studies are conflicting and limited. Nonaspirin, nonsteroidal anti-inflammatory drugs in cardiovascular disease Cyclooxygenase-2 inhibitors in Although a variety of traditional nonsteroidal anti- cardiovascular disease inflammatory drugs (NSAIDs) can inhibit TXA2- Selective COX-2 inhibitors (coxibs) differ from dependent platelet function through competitive, traditional NSAIDs in two major ways. Coxibs are reversible inhibition of COX-1 the effects of these less likely to result in NSAID-induced gastropathy, NSAIDs on cardiovascular disease are controver- and they do not inhibit platelet function [103,104]. sial. An excellent review has recently been pub- The major benefits of coxibs are the reduction in lished that addresses this problem [62]. For gastric ulcer formation and bleeding from those instance, was shown to have a protective ulcers, as reported by the VIoxx Gastrointestinal effect against acute MI [94] and to reduce the risk of Outcomes Research (VIGOR) trial [105]. Another thromboembolic cardiovascular events among benefit of the platelet-sparing coxibs is their use as patients with rheumatoid arthritis [95]. However, and anti-inflammatory agents in other population-based studies [96–98] have failed to situations in which bleeding may limit the use of detect protective effects of naproxen or other non- traditional NSAIDs, such as trauma and surgical aspirin NSAIDs, considering that none of these procedures [106,107]. Otherwise, coxibs are www.futuremedicine.com 471 REVIEW – Reiter & Jilma

supposed to have several effects that could increase Drug Administration (FDA), demonstrated an the risk of cardiovascular disease, including a increase in cardiovascular events among patients decrease in prostacyclin levels, increasing blood receiving [401]. pressure, decreasing angiogenesis and destabilizing Summarizing, since different coxibs were found plaque [108]. to be associated with cardiovascular complica- Recently, this class of drugs has come under tions, it appears that this is a class effect. The risk scrutiny due to clinical reports of an associated of serious cardiovascular events will need to be increased risk of serious cardiovascular weighed against any potential benefits of coxibs in events [109,110]. As reported in the VIGOR trial, preventing colorectal neoplasia and in relieving there were more cardiovascular events among pain. The lesson to be learnt from this observation patients given a high dose of rofecoxib than among has been discussed very recently [119]. those patients given naproxen, a nonselective NSAID with platelet-inhibiting properties of Aspirin-related drugs unclear clinical relevance [105]. In contrast, pooled Given the presumed understanding of aspirin’s data from other randomized trials have not shown mode of antiplatelet effect, it should be possible to a significant difference in cardiovascular risk design a more specific drug to interfere with the between rofecoxib and placebo or other nonselec- COX/TX pathway. Aspirin, by nonselectively tive NSAIDs [111–113]. Indeed, most of the earlier blocking COX both in platelets and in endothelial trials of coxibs did not appear to show an increase cells, not only inhibits the TXA2 pathway of in cardiovascular events [114–116]; however, these platelet activation but, at the same time, also the trials were generally short-term studies designed to generation of vasodilating and platelet-inhibitory assess the use of this class of drug for pain relief and , such as PGI2 (or prostacyclin), by the to evaluate associated adverse GI events. endothelial cells. This is of importance because Overall, studies have provided conflicting data there is an increased intravascular prostacyclin on the association of coxibs with cardiovascular generation in patients at advanced stages of risk but only limited long-term data have been atherosclerosis, which is paralleled by the degree available for analysis so far. However, this obser- of platelet activation [120]. Even 40-mg doses of vation has been changed since the results of aspirin are sufficient to inhibit the local produc- three long-term trials have recently been tion of antithrombotic PG generation in the published [108,117,118]. The Adenomatous Polyp blood vessels of atherosclerotic patients [121]. This PRevention On Vioxx (APPROVe) study, which appears to be a limitation to the antithrombotic was designed to determine the effect of 3 years’ efficacy of aspirin and was one of the reasons to treatment with rofecoxib on the risk of recurrent look for alternatives, specifically, more selective neoplastic polyps among patients with a history inhibitors of TX formation and action. of colorectal adenomas, showed an increased car- diovascular risk associated with the long-term Agents interfering directly with thromboxane use of rofecoxib [108]. The Coronary formation &/or action Bypass Grafting (CABG) surgery study showed TX synthase inhibitors and TX receptor antago- that cardiovascular events (including MI, cardiac nists directly interfere with TX formation (syn- arrest, stoke and pulmonary embolism) were thase inhibitors) and action (receptor blockers). more frequent among the patients given TX synthase inhibitors do not reduce vascular and than among those PGI2 formation but enhance it by shifting accu- given placebo (2 vs. 5%; risk ratio: 3.7; p = 0.03) mulating PG endoperoxidase into this pathway [118] . The Adenoma Prevention with Celecoxib [122]. In fact, TX synthase inhibitors and TX (APC) study reviewed all potentially serious car- receptor blockers have been demonstrated to diovascular events among 2035 patients with a exhibit potent antithrombotic effects in several history of colorectal neoplasia who were enrolled animal studies [123–129]. In general, inhibition of in a trial comparing two doses of celecoxib TX synthase appears to be more efficient than (200 and 400 mg twice daily) with placebo for the blockade of TX receptors [126,130]. To sum- the prevention of colorectal adenomas [117]. marize, all of these data suggest that the selective There was a dose-related increase in cardiovascu- inhibition of TX synthase, in particular if com- lar events for celecoxib when compared with pla- bined with the blockade of TX receptors, would cebo. Of interest, the results of a randomized, be a useful approach to antiplatelet therapy. controlled of celecoxib with Alzhe- Picotamide, a dual TXA2 synthase inhibi- imer’s disease, reported to the US Food and tor/receptor antagonist, slowed the evolution of

472 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

early carotid lesions in a controlled study in dia- smooth muscle proliferation [143]. Thus, one may betic patients [131]. However, more well-proven speculate that this class of drugs could also have randomized controlled trials are required to fur- therapeutic potential in the treatment of cardio- ther investigate the possible benefit of picota- vascular or cerebrovascular diseases. mide in preventing the formation of arterial The PGI2 analog iloprost was, however, una- thrombus in such patients. ble to prevent reocclusions of stenosed dog coro- The Coronary Artery Restenosis Prevention nary arteries after electrical injury or On Repeated Thromboxane–antagonism (CAR- [144]. This could be due to receptor PORT) study compared the TX receptor antago- desensitization owing to high levels of PGI2. nist vapiprost with aspirin in patients Importantly, platelet receptors may become undergoing percutaneous transluminal coronary downregulated if there is a significant increase in angioplasty (PTCA). After a 6-month follow-up PGI2 production, for example, in unstable period, no effect on the incidence of restenosis angina and acute MI [145,146]. Moreover, only a was observed [132]. small number of larger clinical trials with PGI2- The combined TX synthase inhibitor and TX related compounds [147,148] exist and the results, receptor antagonist, ridogrel, was compared with in general, were not encouraging. In a more aspirin in patients with acute MI receiving recent study the orally active PGI2 analog, berap- streptokinase in the Ridogrel versus Aspirin rost, was compared with placebo in patients with Patient Trial (RAPT) [133]. Ridogrel was not intermittent claudication [149]. Although the superior to aspirin in enhancing the fibrinolytic incidence of critical cardiovascular events was efficacy of streptokinase, although there was a not significantly reduced in those patients lower rate of new ischemic events noted in a assigned to beraprost, there was a significant post hoc analysis. reduction in the combination of cardiovascular Moreover, terbogrel, another combined TX death and MI. Critical cardiovascular events synthase inhibitor/receptor antagonist failed to were defined as: live up to expectations in a trial of patients with • Death of cardiovascular origin, nonfatal MI or primary pulmonary hypertension [134]. Although unstable angina terbogrel was able to reduce TX metabolism by • Stroke or TIA 98% with a modest, but statistically insignificant • Critical leg ischemia, subacute critical (39%) rise in PGI2 metabolites, it was associated with severe leg pain, thus limiting its clinical ischemia, peripheral angioplasty, peripheral utility. In contrast, another study of TX synthase bypass surgery or amputation at any level inhibition in patients with primary pulmonary Nonetheless, judgements on whether PGI2 disease did not describe leg pain as a side analogs could have potential beneficial cardiopro- effect [135] and, in a recently published trial, ter- tective effects should not be made from this one bogrel was shown to be well tolerated without trial. Moreover, major problems include the obvious adverse effects in healthy receptor-mediated nature of response and the low volunteers [136]. selectivity for the platelet, which may result in In summary, despite showing considerable nonspecific effects, for example hypotension [122]. promise in preclinical studies, TX synthase To summarize, the future of these drugs as a inhibitors and TX receptor antagonists have possible potential new approach to the treatment been disappointing in clinical trials and have of vascular diseases is, indeed, questionable. not demonstrated a benefit over aspirin. There- fore, these newer drugs could provide an alter- Gingerols & related analogs native approach to antiplatelet therapy, but Gingerols, the active components of ginger (the more clinical trials are required. rhizome of Zingiber officinale, Roscoe), were shown to selectively inhibit secondary platelet Prostacyclin-related agents activation and ATP release from platelets in PGI2, known to have potent antiplatelet and human platelet-rich plasma, which is due to vasodilatatory activities [137], occurs naturally in inhibition of AA metabolism and COX the vascular endothelium and has been approved activity [150]. Gingerols and other synthetic ana- by the FDA for the management of pulmonary logs were also shown to have a strong COX-1 artery hypertension [138–140]. PGI2 relaxes vascu- inhibitory activity in rat basophilic leukemia lar smooth muscle [141,142], inhibits platelet cells [151], a cell line with COX-1 expression [152]. aggregation [141,142] and also suppresses vascular More recently, the inhibition of AA-induced www.futuremedicine.com 473 REVIEW – Reiter & Jilma

platelet activation in human blood was studied. For Effectively avoiding Second Strokes trial Gingerol and gingerol analogs dose-dependently (PRoFESS) [161]. Irrespective of the outcome of inhibited COX-1 activity and the COX-1 inhib- this study, there is one major limitation in the itory effect of these substances was more potent design of the PRoFESS study, as the two treat- than aspirin [153]. In a rat model, ginger extract ment arms will not be compared against aspirin. and other ginger preparations showed antiulcer This aspect is important since the evidence of the activity [154,155]. Thus, these substances could be superiority of aspirin/ERDP over aspirin alone useful well-tolerated platelet activation inhibi- hinges on a single trial. Moreover, in high-risk tors; however, data on these substances are rare patients with recent TIA or ischemic stroke, and additional experiments are needed to gain a clopidogrel plus aspirin was not superior to clopi- better insight into the effect of platelet inhibition dogrel alone, as demonstrated in the Manage- by gingerols. ment of ATherothrombosis with Clopidogrel in High-risk patients with recent TIA or ischemic Phosphodiesterase inhibitors stroke (MATCH) study, but was not compared Dipyridamole with aspirin alone. Thus, the results of the PRo- Both the inhibition of cyclic FESS trial will not enable a judgement to be phosphodiesterase (PDE) (the enzyme that made on the benefits of a combination of differ- degrades cyclic adenosine monophosphate ent antiplatelet drugs as a therapeutic approach [cAMP] to 5´-AMP, resulting in the intraplatelet for patients with cerebrovascular diseases. accumulation of cAMP, a platelet inhibitor) and In conclusion, aspirin/ERDP has been FDA the blockade of the uptake of adenosine (which approved and is usually classified as a potential acts at A2 receptors for adenosine to stimulate first-line therapy in the secondary prevention of platelet adenyl cyclase and, thus, increases the ischemic stroke and TIA, especially in patients level of cAMP), have been suggested [156]. More- with lower cardiovascular comorbidity [162,163] but over, dipyridamole blocks the enzyme cGMP the current feeling is that there is not yet sufficient PDE, thereby inhibiting the breakdown of evidence to justify adoption of aspirin and dipyri- cGMP (Figure 1)[157]. Raised levels of cAMP and damole as a first-line treatment for the secondary cGMP within platelets potentiate inherent prevention of stroke. Thus, aspirin should be the mechanisms, resulting in vasodilatation and first-line antiplatelet therapy in the secondary pre- inhibition of aggregation [158]. vention of stroke and TIA [39,164,165] (Table 3) until prospective, randomized clinical trials have shown Dipyridamole in a sustained benefit of aspirin/ERDP or clopidog- Dipyridamole appears to have similar efficacy to rel plus aspirin over the gold-standard treatment low-dose aspirin in preventing stroke [159]. In with aspirin. patients with cerebrovascular disease, the Euro- pean Stroke Prevention Study (ESPS)-2 demon- Dipyridamole & coronary artery steal strated that the combination of low-dose aspirin Perfusion imaging during coronary vasodilata- (50 mg daily) and extended-release dipyridamole tion with either adenosine or dipyridamole is (ERDP) (400 mg daily) was superior in prevent- widely used for the diagnosis of coronary artery ing nonfatal stroke than either drug alone [159]. disease (CAD) [166–168]. Dipyridamole, admin- However, comparing dipyridamole plus aspirin istered intravenously, represents a well-estab- with aspirin alone was associated with only a lished that induces dilatation of nonsignificant reduction in serious vascular coronary arteries by inhibiting the degradation events [39,160]. Indeed, the apparent reduction in of adenosine. nonfatal stroke was derived mainly from the Vasodilatation in nonischemic regions can ESPS-2 study but this result was not supported divert blood from already underperfused by the findings for nonfatal stroke in other stud- regions to parallel-perfused regions where the ies or by the overall findings for nonfatal MI or vasodilator reserve has not been exhausted vascular death [39]. Finally, headaches limited the (coronary steal) [169]. Coronary steal has been use of aspirin/ERDP as they occurred in 37% of well described in canine studies of CAD after treated patients in the ESPS-2 study and resulted intravenous dipyridamole [170]. Moreover, in a high rate of noncompliance. myocardial ischemia due to coronary steal is Currently, aspirin/ERDP is being compared generally believed to be manifested clinically with clopidogrel in the largest ever secondary by ST segment depression following coronary stroke-prevention trial: the Prevention Regimen vasodilatation [171,172].

474 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

Indeed, there are some reports concerning proliferation was greater in the cilostazol than in patients who developed angina pectoris manifested the group [192]. Moreover, a meta-anal- by ischemic electrocardiographic changes and per- ysis of five clinical studies comparing cilostazol fusion defects, which occurred after dipyridamole (plus aspirin) with ticlopidine (plus aspirin) administration [173–175]. Moreover, a significant showed no difference regarding the effectiveness difference on coronary angiography between and safety for a 1-month period when used as an patients with dipyridamole-induced angina pec- adjunctive therapy after coronary stenting [193]. toris and those without angina pectoris was found Despite the obvious beneficial effect of cilosta- in the presence of collaterals (p < 0.05) [173]. zol in these clinical settings, there may be con- Therefore, it has been suggested that angina pec- cerns regarding the study designs of these trials. toris during dipyridamole stress test is due to Indeed, these trials were single-center studies ischemia, which is not related to the severity of with a relatively small number of patients [186–192] CAD but is probably owing to coronary steal to and some of them were not randomized [190] or the collateralized territory in patients without double blinded [188,192]. Thus, large-scale, multi- transmural MI. Thus, dipyridamole-induced center, randomized trials are needed to confirm angina pectoris could be predictive for collaterals the efficacy of cilostazol in other patients groups and may indicate viability in patients with MI [173]. of different ethnicity. Overall, since dipyridamole has a vasodilatatory In animal studies, cilostazol was shown to effect, it should be used with caution in patients decrease ischemic brain infarction [194–196]. In a with severe CAD (e.g., unstable angina and MI) recently published study of guinea pig and and chest pain may be aggravated in patients human cerebral arteries, it has been suggested underlying CAD who are receiving dipyridamole. that cilostazol is still effective under conditions with possible dysfunctional NO-cGMP path- Cilostazol way, such as in ischemic stroke or cerebral vasos- Cilostazol, a potent inhibitor of platelet aggrega- pasm [197]. Thus, it is of interest whether this tion [176] with vasodilating properties, inhibits drug could also have therapeutic potential in cAMP-selective PDE-III [177], thereby increasing patients with cerebrovascular disorders. the intracellular level of cAMP. Cilostazol revers- In a previous report [185], it could be demon- ibly inhibits platelet aggregation induced by a strated that cilostazol prevented the progression variety of stimuli, including thrombin, ADP, col- of carotid intima thickness in patients with lagen, AA, epinephrine and shear stress [178]. Type II diabetes mellitus. The group without Cilostazol has also been shown to be a potent cilostazol had a significant increase in infarct-like antiplatelet agent with antiproliferative proper- lesions which was positively correlated with the ties [179], that increases peripheral blood flow [180] intima media thickness. The intima media thick- and ameliorates insulin resistance [181,182]. Thus, ness of the carotid artery is used as a surrogate of it has been suggested that cilostazol could pre- definite atherosclerosis with a high risk of vascu- vent both thrombosis and restenosis after coro- lar events [198–200]. The results of a secondary nary stenting when coadministered with prevention study using cilostazol 200 mg/day aspirin [183,184] but it could also be effective in showed a reduction of stroke by 43.3% com- the primary prevention of ischemic stroke in pared with the placebo group [201]. However, subjects with Type II diabetes mellitus [185]. cilostazol was compared with placebo without Several reports have demonstrated that cilosta- administering a standard antiplatelet therapy. zol can prevent subacute thrombosis after stent Indeed, as this was a secondary prevention study implantation and may reduce restenosis after coro- of patients (n = 1052) who suffered from cere- nary interventions [186–189]. As reported [190], after bral infarction 1 to 6 months prior to enrolment, coronary angioplasty, the restenosis rate was signif- this appears inadequate. As the place of aspirin in icantly lower in groups with cilostazol than in the secondary prevention of stroke/TIA has been groups with aspirin or ticlopidine. Moreover, established to the satisfaction of most cilostazol has been shown to have an outstanding authors [39,164,165,202], the design of this study has effect on the prevention of acute or subacute to be criticised. Moreover, the antithrombotic thrombotic complication after coronary stenting, effects of cilostazol after stent implantation may equal to ticlopidine [191]. A very recent study dem- be somewhat overstated, particularly with drug- onstrated that ticlopidine showed significantly less eluting stents. Indeed, patients receiving cilosta- subacute thrombosis after stenting compared with zol had more acute/subacute stent thrombosis cilostazol but the inhibition of neointimal compared with those receiving clopidogrel. www.futuremedicine.com 475 REVIEW – Reiter & Jilma

Taken together, cilostazol has been routinely Agents interfering with ADP-mediated used as an antithrombotic agent for the treatment platelet reactions of peripheral arterial occlusive disease in Japan Thienopyridines (ticlopidine & clopidogrel) and some Asian countries for more than 15 years ADP is an important platelet agonist, which [203,204] and a new indication for stroke preven- activates platelets by binding to purinergic tion has been recently approved in Japan [204]. In receptors on the platelet surface. There are contrast, cilostazol has been available for the three recognized subtypes of P2 receptors on treatment of intermittent claudication in the platelet membranes, namely P2X1, Y1 and USA since 1999 and in the UK since 2000 [204] Y12 [208–210]. The latter is the target of the but is not generally considered an antithrombotic antiplatelet thienopyridines, ticlopidine and agent in Western countries, perhaps due to the clopidogrel [211,212]. Both drugs selectively bulk of its antithrombotic preclinical and clinical inhibit ADP-induced platelet aggregation with development being conducted in Japan. no direct effects on AA metabolism (Figure 1) [213]. While not studied as extensively Phosphodiesterase inhibitors in perspective as aspirin, several clinical trials have confirmed NSP-513 is a novel selective PDE-III inhibitor the ability of thienopyridines to reduce cardio- on PDE isozyme activities and in vitro platelet vascular events in patients with several different aggregation and in vivo thrombus formation types of cardiovascular disease (Table 5) [214,215]. were investigated as reported recently [205]. Moreover, ticlopidine has been established as an NSP-513 selectively inhibited human platelet alternative to aspirin in the prevention of recur- PDE-III isozyme in vitro. In a mouse pulmonary rent cerebral ischemia and stroke [216] but its thromboembolism model, orally administered use has been limited due to potentially detri- NSP-513 showed in vivo antithrombotic effects mental side effects, including fatal severe neu- that were 320- to 470-times more potent than tropenia and thrombotic thrombocytopenic those of cilostazol. In a rat carotid arterial throm- purpura [217] as well as aplastic anemia [218]. As a bosis model, intraduodenally administered result, it has been replaced by clopidogrel NSP-513, cilostazol and aspirin reduced throm- [62,214,219]. Indeed, clopidogrel has become a bus formation by 75, 66 and 48%, respectively. mainstay in antiplatelet therapy [99,220,221] and In contrast, intravenously administered dipyrid- several studies have been preformed using this amole did not significantly prevent thrombus drug (Table 5). formation and therefore NSP-513 is suggested to have the potential to prevent, not only in vitro Clinical studies of patients with vascular diseases, platelet aggregation, but also in vivo thrombus particularly patients with cardiovascular diseases formation. Of interest, this study shows that the The Clopidogrel versus Aspirin in Patients at antiplatelet and antithrombotic activities of Risk of Ischemic Events (CAPRIE) trial, involv- NSP-513 are greater than those of cilostazol, ing 19185 subjects, was the first randomized, dipyridamole or aspirin, thus, may have thera- double-blinded, international trial to evaluate the peutic potential in the treatment of arterial efficacy of aspirin 325 mg once daily versus clopi- thrombotic disorders. dogrel 75 mg once daily in patients with recent KW-7, a new inhibitor of cyclic nucleotide ischemic stroke or MI, or established peripheral PDE, was shown to inhibit cAMP- and cGMP- disease [214]. An intent-to-treat analysis of all ran- PDE activities as well as AA-stimulated TXA2 domized patients showed a modest 8.7% RRR production [206]. This was associated with an (p = 0.043) in the primary outcome of stroke, MI increase in PGD2 levels, indicating that KW-7 is or vascular death. Interestingly, in a subgroup also an inhibitor of TX synthase. In a very recent analysis, patients with peripheral arterial disease study, it was demonstrated that cilostazol and derived the greatest benefit from the drug dipyridamole synergistically inhibited platelet (RRR: 23.8%; p = 0.0028) whereas a nonsignifi- aggregation in vitro and ex vivo, compared with cant 7.3% RRR in patients with stroke and a treatment with either drug alone [207]. Although nonsignificant 3.7% risk increase in the primary the dual inhibition of KW-7 on PDE and TX outcome of patients with MI was obtained. Over- synthase and the combination of cilostazol and all, the safety and tolerability of clopidogrel and dipyridamole might provide an attractive target in aspirin were similar and, therefore, clopidogrel developing new antiplatelet drugs, only few data was established as an alternative antiplatelet to exist and no data on the use of such inhibitors in aspirin for secondary prevention across a wide clinical trials are currently available. spectrum of patients with vascular disease.

476 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

Table 5. Clinical studies of dual antiplatelet therapy with clopidogrel Trial Patients Subjects Treatment Primary end point Result Ref. (n)

CLASSICS Patients 1020 Clopidogrel loading dose Major peripheral or 50% RRR in the occurrence [219] after 300 mg + 325 mg aspirin, bleeding complications, of primary endpoint in favor coronary followed by clopidogrel neutropenia, of clopidogrel stenting 75 mg/day + aspirin thrombocytopenia or (p = 0.005) 325 mg/day vs. early discontinuation of clopidogrel 75 mg/day study drug as the result + aspirin 325 mg/day of noncardiac adverse vs. ticlopidine 250 mg event during the study twice daily + aspirin treatment period 325 mg/day CREDO Patients 2116 Clopidogrel loading dose 1-year incidence of the Following PCI, long-term [224] undergoing 300 mg (Group A) or composite of death, MI clopidogrel therapy PCI placebo (Group B) before or stoke in the intent- significantly reduced the PCI plus aspirin to-treat population and risk of ischemic events 325 mg/day 28-day incidence of the (27% RRR; p = 0.02) (Group A + Group B), composite of death, MI, A loading dose of followed by or urgent target-vessel clopidogrel given before Group A: clopidogrel revascularization in the PCI did not reduce 75 mg/day + aspirin per-protocol population ischemic events at 28 days 325 mg/day for 12 (18.5% RRR; p = 0.23). months However, a subgroup of vs. Group B: clopidogrel patients receiving 75mg/day for 28 days; clopidogrel >6 h before from day 29 through 12 PCI experienced a 38.6% months placebo + aspirin RRR (p = 0.051) compared 325 mg/day for 12 with no reduction in months patients receiving clopidogrel less than 6 h before PCI. CURE Unstable 12562 Clopidogrel loading dose Composite of 20% RRR in ischemic events [222] angina or 300 mg, followed by cardiovascular death, in favour of clopidogrel non-ST clopidogrel 75mg/day MI or stroke and the (p < 0.001) segment + aspirin 325 mg/day, composite of There were significantly elevation vs. placebo loading dose, cardiovascular death, more patients with major MI followed by placebo MI, stroke or refractory bleeding in the clopidogrel + aspirin 325 mg/day ischemia group than in the placebo group (3.7 vs 2.7%, 1.38 RRR; p < 0.001) but there were not significantly more patients with episodes of life-threatening bleeding (2.1 vs 1.8%; p = 0.13) or hemorrhagic strokes.

CREDO: Clopidogrel for Reduction of Events During Observation trial; CURE: Clopidogrel in Unstable angina to prevent Recurrent Events trial; MATCH: Management of Atherothrombosis with clopidogrel in high-risk patients with recent Transient isCHemic attacks or ischemic stroke trial; MI: Myocardial infarction; PCI: Percutaneous coronary intervention; PCI-CURE: Percutaneous coronary intervention – Clopidogrel in Unstable angina to prevent Recurrent Events trial; RRR: Relative risk reduction.

www.futuremedicine.com 477 REVIEW – Reiter & Jilma

Table 5. Clinical studies of dual antiplatelet therapy with clopidogrel (Cont.). Trial Patients Subjects Treatment Primary end point Result Ref. (n)

PCI-CURE Subset of 2658 Clopidogrel loading dose Composite of 30% RRR in ischemic events [70] CURE 300 mg, followed by cardiovascular death, from PCI to 30 days in favor undergoing clopidogrel 75 mg/day + MI or urgent target- of clopidogrel (p = 0.03). PCI aspirin 325 mg/day vessel revascularization Overall (including ischemic vs placebo loading dose, within 30 days of PCI events before and after PCI) followed by placebo + there was a 31% RRR in aspirin 325 mg/day cardiovascular death or MI After PCI, patients (p = 0.002) received either clopidogrel or ticlopidine + aspirin for 2 to 4 weeks, after which administration of the randomly assigned study medication was started until the end of the scheduled follow-up MATCH Recent 7599 Clopidogrel 75 mg/day First reoccurrence of There was a nonsignificant [225] ischemic + aspirin 75 mg/day ischemic stroke, MI, 6.4% RRR in the primary stroke or vs clopidogrel 75 mg/day vascular death or endpoint when adding transient + placebo rehospitalisation for an aspirin to clopidogrel ischemic acute ischemic event (p = 0.244). attack with Adding aspirin to at least one clopidogrel was associated additional with increased life-threating vascular bleedings (absolute risk risk factor increase 1.26%; p < 0.0001) and increased major bleeding (absolute risk increase 1.36%; p < 0.0001). No difference between groups was recorded in mortality. CREDO: Clopidogrel for Reduction of Events During Observation trial; CURE: Clopidogrel in Unstable angina to prevent Recurrent Events trial; MATCH: Management of Atherothrombosis with clopidogrel in high-risk patients with recent Transient isCHemic attacks or ischemic stroke trial; MI: Myocardial infarction; PCI: Percutaneous coronary intervention; PCI-CURE: Percutaneous coronary intervention – Clopidogrel in Unstable angina to prevent Recurrent Events trial; RRR: Relative risk reduction.

Moreover, CLASSICS demonstrated the supe- There was a RRR of 20% in (9.3% patients in rior efficacy and safety of clopidogrel plus aspirin the clopidogrel group vs. 11.4% in the placebo compared with ticlopidine plus aspirin in group; p < 0.001) in the composite primary out- patients undergoing coronary stenting [219]. This come of death from cardiovascular causes, non- was the first randomized trial of clopidogrel in fatal MI or stroke. The rates of bleeding were coronary stenting and the first study to evaluate higher in the clopidogrel group than in the pla- clopidogrel–aspirin combination therapy and a cebo group (3.7 vs. 2.7%; p = 0.001), but there loading dose of clopidogrel. were not significantly more episodes of life- The CURE trial investigated the effect of threatening bleeding. The PCI-CURE, a sub- clopidogrel combined with aspirin in the treat- study of CURE, showed that the benefit of ment of patients with acute coronary syndromes clopidogrel over placebo was also seen in patients (ACSs) that included unstable angina and MI receiving PCI [70]. Overall, there was a 31% without ST-segment elevation [222]. A total of reduction in cardiovascular death or MI 12562 patients presenting within 24 h after the (p = 0.002). Moreover, a meta-analysis of ten onset of symptoms received clopidogrel (300 mg studies, comparing clopidogrel plus aspirin ver- immediately, followed by 75 mg once daily) or sus ticlopidine plus aspirin after - placebo in addition to aspirin for 3 to 12 months. ing demonstrated that clopidogrel, in addition to

478 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

better tolerability and fewer side effects, is at The results of this study are of interest, as the RRR least as effective as ticlopidine in reducing in a subgroup analysis for stroke alone was not sig- 30-day major adverse cardiac events [223]. Thus, nificant in the CAPRIE study [214]. However, the clopidogrel plus aspirin should replace ticlo- CAPRIE study was not designed to specifically pidine plus aspirin as the standard antiplatelet address patients who had cerebrovascular disease. regimen after stent deployment. Indeed, the The MATCH trial showed a nonsignificant combination of aspirin and clopidogrel has RRR of 6.4% (16% patients in the clopidogrel become standard treatment up to 12 months and aspirin arm vs. 17% patients in the clopi- after coronary stent implantation [99]. dogrel-only arm; p = 0.244) in the composite The CREDO trial was designed to evaluate primary outcome of ischemic stroke, MI, vascu- the benefit of clopidogrel pretreatment and lar death, or rehospitalization for an acute long-term therapy to a more stable population ischemic event. The 6.4% RRR in favor of aspi- undergoing coronary stenting [224]. Patients rin plus clopidogrel in the intent-to-treat-analy- receiving 1 year instead of 1 month of clopidog- sis among all randomized patients is in the range rel showed a significant 27% RRR in the com- that was reported about the stroke subgroup posite of death, MI or stroke. Clopidogrel population (7.3%) in the CAPRIE study [214]. pretreatment did not significantly reduce the The rates of life-threatening bleeding were combined risk of death, MI or urgent target ves- higher in the clopidogrel plus aspirin arm versus sel revascularization at 28 days. However, in a the clopidogrel-only arm (3 vs. 1%; p < 0.0001) subgroup analysis, patients who received a load- but in both treatment arms, no hemorrhagic ing dose of 300 mg at least 6 h before PCI expe- transformation of ischemic stroke was reported rienced a 38.6% RRR (p = 0.051) for this end as life-threatening bleeding and no significant point compared with no reduction with treat- difference was recorded in the incidence of ment less than 6 h before percutaneous inter- fatal bleeding. However, several vention. Risk of major bleeding at 1 year trials [70,222,224] have demonstrated a clear benefit increased, but not significantly. of the combination of clopidogrel and aspirin To summarize, the findings of these rand- over aspirin alone for the prevention of vascular omized controlled trials have shown the sustained end points in patients with coronary heart dis- benefit of clopidogrel in addition to standard ease. Moreover, the increase in bleeding risk with treatment including aspirin, especially in patients the combination was smaller than in MATCH. with coronary manifestation of atherothrombo- The differences between the MATCH trial sis. The overall safety profile of clopidogrel is at and the cardiology trials could be due to differ- least as good as that of medium-dose aspirin and ent designs. Whereas in the cardiology trials is superior to that of ticlopidine. clopidogrel was added to treatment, in the MATCH trial aspirin was added to clopidog- Clinical studies addressed to patients with rel. Consequently, the MATCH trial provided cerebrovascular diseases or heart failure a measure of the benefit-to-risk ratio of aspirin Data from the MATCH trial have recently been in addition to clopidogrel, not for clopidogrel published [225]. The trial, involving added to aspirin as in the cardiology trials. One 7599 patients, was a first randomized, double- major limitation of this study is that clopidog- blinded trial that was designed to assess whether rel was used as the standard therapy for the addition of aspirin 75 mg once daily to clopi- patients with cerebrovascular diseases although dogrel 75 mg once daily could have a greater aspirin presently is considered the treatment of benefit than clopidogrel alone in reducing the choice for secondary prevention of disorders risk of recurrent ischemic vascular events in associated with arterial thrombosis [39,164,165]. high-risk patients after TIA or ischemic stroke. Indeed, a meta-analysis of more than 287 clin- Patients were included if they had an ischemic ical trials showed that, overall, aspirin reduces stroke or TIA in the previous 3 months and had the risk of stroke, MI and vascular death by one or more of five additional risk factors: approximately 23% in patients with various • Previous ischemic stroke cardiovascular and cerebrovascular diseases [39]. •Previous MI Thus, recommendations for clopidogrel are usually made to patients who are intolerant of • Angina pectoris aspirin, who have had a recurrent ischemic • Diabetes mellitus event while on aspirin, or who are at vascular • Symptomic peripheral arterial disease high risk [39,65,99,162,163,202,220]. www.futuremedicine.com 479 REVIEW – Reiter & Jilma

In summary, the outcome of the MATCH prior ischemic events [230–232]. Clopidogrel, in trial indicates that the combination therapy of combination with aspirin, has been demon- aspirin plus clopidogrel is not superior to clopi- strated to be more efficacious than aspirin alone dogrel alone, and that the addition of aspirin to in patients presenting with ACS, with a favorable clopidogrel results in significantly higher bleed- safety profile [222,224,233]. ing rates. As a consequence, the combination of As most of the trials were addressed to patients aspirin and clopidogrel for cerebrovascular pre- with cardiovascular diseases, numerous trials vention should only be given within controlled have now been initiated to study the benefits of studies. Indeed, the design of the PRoFESS trial combining aspirin and clopidogrel for other indi- has been changed [402] following the announce- cations such as stroke or atrial ment of the results of the MATCH trial and this fibrillation (Table 6). One of these interesting trial is no longer utilizing a combination of studies is the Clopidogrel for High Atherothrom- clopidogrel plus aspirin as the comparator – it is botic Risk and Ischemic Stabilization, Manage- now clopidogrel alone. Unfortunately, whether ment, and Avoidance (CHARISMA) study clopidogrel is superior to aspirin in the treatment (Table 6) which was designed to evaluate the effi- of cerebrovascular diseases cannot be concluded cacy and safety of clopidogrel plus aspirin versus from the MATCH trial and therefore no recom- placebo plus aspirin in patients with established mendation can yet be given for the primary use coronary, cerebral or peripheral arterial disease or of clopidogrel in these patients. Additional infor- in patients with multiple risk factors for athero- mation could be obtained by a further study thrombosis who have not yet suffered from an comparing clopidogrel versus aspirin addressed ischemic event [234]. The results of this study will especially to patients with cerebrovascular dis- be of interest since it is unknown whether dual eases. Indeed, this is of interest as several contro- antiplatelet therapy is superior to aspirin mono- versial recommendations and guidelines therapy for high-risk primary and secondary pre- concerning the use of antiplatelet drugs in vention. This large-scale trial of patients at a high patients with TIA or stroke have been risk for atherothrombotic events will allow deter- published [162–165,202,226–228] but there are no mination of the value of the strategy of adding data available regarding how these recommenda- clopidogrel to the current standard of care, tions translate into clinical practice and which including low-dose aspirin, for a wide spectrum affect the choice of antiplatelet drugs in patients of patients with atherothrombosis. with a recent ischemic cerebrovascular event. The and Antiplatelet Therapy in Clopidogrel: a better antiplatelet drug for the Chronic Heart failure (WATCH) trial was prevention of gastrointestinal bleeding? designed to determine the optimal antiplate- An overview of randomized trials of aspirin let/thrombotic agent for heart failure. Patients therapy found that GI toxicity (both major and were randomized to open-label warfarin (target minor) was dose related, with daily doses international normalized ratio [INR]: 2.5–3.0) between 30 and 1300 mg [235]. Even when or double-blind antiplatelet therapy with aspirin administered at very low doses (30–50 mg/day), 162 or clopidogrel 75 mg. Unfortunately, the aspirin can cause serious GI bleeding [69,159]. Pro- trial had to be terminated after a study period of ton-pump inhibitors reduce the risk of aspirin- 18 months due to poor enrolment, with a result- induced ulcera-bleeding [236,237] and thus, con- ing reduction of its power to achieve its original current therapy with these drugs has become a objectives [229]. standard treatment for patients at risk for ulcera bleeding who are taking aspirin [238,239]. Thienopyridines in perspective An alternative strategy is to replace aspirin Thienopyridines have become a mainstay in with another antiplatelet drug that does not antiplatelet therapy. Ticlopidine is currently used induce GI ulcera. Clopidogrel has been shown as a reserve drug due to its unfavourable side- to be potentially superior to aspirin in patients effect profile (neutropenia and thrombocyto- with atherothrombotic disease [214]. Moreover, penic purpura) and has been replaced by clopi- administration of clopidogrel resulted in a dogrel. Clopidogrel has been shown to be slightly lower rate of GI bleeding when com- superior to aspirin in patients with atherothrom- pared with aspirin (0.5 vs. 0.7%) [214] and did botic disease [214]. The benefit of clopidogrel not induce gastric damage in healthy volun- appears particularly pronounced in patients teers [240]. Thus, clopidogrel has recently been with diabetes, prior revascularization, and recommended for patients unable to take

480 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

Table 6. Ongoing trials with clopidogrel. Trial Patients Number of Study design subjects ACTIVE A 7500 Patients with a contraindication to warfarin or who refuse therapy will be randomized to receive either clopidogrel 75 mg/day + aspirin 75–100 mg/day or aspirin 75–100 mg/day ACTIVE W Atrial fibrillation 6500 Patients eligible for will be randomized to receive either clopidogrel 75 mg/day + aspirin 75–100 mg/day or adjusted dose K antagonist (INR 2.0–3.0) ARCH Aortic arch atheroma 1500 Patients will be randomized to receive either warfarin (INR 2.0–3.0) or aspirin 75–325 mg/day + clopidogrel 75 mg/day CAMPER Peripheral arterial 2000 Unknown intervention CASPAR Peripheral arterial bypass 1460 Unknown surgery CCS- Acute ST-segment 45000 Clopidogrel 75 mg/day + aspirin versus aspirin alone 2/COMMIT elevation MI CHARISMA Secondary and high-risk 15603 Two treatment groups will be randomized to receive either clopidogrel primary prevention 75 mg/day or placebo, in combination with aspirin 75–162 mg/day CLARITY Acute ST-segment 3000 Clopidogrel 75mg/day + aspirin vs aspirin alone elevation MI FASTER Acute TIA and minor 7500 All patients will be on aspirin. Patients will be randomized to ischemic stroke within 12 clopidogrel (300 mg loading dose + 75 mg/day) or placebo, and to hours of onset simvastatin (40 mg/day) or placebo PRoFESS Secondary prevention of 15500 Patients will be randomized to either of two antiplatelet combination stroke regimens: aspirin/ERDP (25 mg aspirin/200 mg extended-release dipyridamole) or clopidogrel 75 mg/d, and randomized to treatment with telmisartan 80 mg or placebo SPS3 Secondary prevention of 2500 Patients will be randomized to receive either aspirin 325 mg/day or a stroke, major vascular combination of aspirin 325 mg/day + clopidogrel 75 mg/day. events and cognitive Hypertensive S3 patients will be randomized to receive decline among patients antihypertensive therapy with a target systolic blood pressure of either with small subcortical <150 mmHg or stroke <130 mmHg. ACTIVE: Atrial fibrillation Clopidogrel Trial with Irbesartan for prevention of Vascular Events A trial; ARCH: Aortic arch Related Cerebral Hazard; CAMPER: Clopidogrel and Aspirin in the Management of Peripheral Endovascular Revascularization; CASPAR: Clopidogrel and Aspirin in bypass Surgery for Peripheral ARterial disease; CCS2/COMMIT: Second Chinese Cardiac Study/ClOpidogrel and Metoprolol Myocardial Infarction Trial; CHARISMA: Clopidogrel for High Atherothrombotic Risk and Ischemic Stabilization, Management and Avoidance; CLARITY: CLopidogrel as Adjunctive ReperfusIon TherapY; FASTER: Fast Assessment of Stroke and TIA to prevent Early Recurrence study INR: International normalized ratio; MI: Myocardial infarction: PRoFESS: Prevention Regimen For Effectively avoiding Second Strokes trial; SPS3: Secondary Prevention of Stroke 3 trial; TIA: Transient ischemic attack.

aspirin owing to previous GI intolerance which clopidogrel (75 mg/day) was compared [65,220]. However, although one study found a with aspirin (80 mg/day) plus esomeprazole lower incidence of GI bleeding among patients (20 mg twice daily) in high-risk patients who receiving clopidogrel than in those receiving had a -induced upper GI bleed- aspirin, a relatively high dose of aspirin ing, have been reported [241]. Over a 1-year fol- (325 mg/day) was used for the secondary pre- low-up period, the incidence of recurrent ulcera vention of cardiovascular/cerebrovascular bleeding was significantly higher in patients tak- events in this study [214]. ing clopidogrel when compared with those tak- Interestingly, until very recently, there has ing aspirin plus esomeprazole (8.6 vs. 0.7%). been no prospective trial available to assess Among patients with a history of aspirin- whether clopidogrel is an alternative to aspirin induced ulcera bleeding, aspirin plus esomepra- plus a proton-pump inhibitor for patients at risk zole was definitely superior to clopidogrel for the for ulcera. Currently, the results of a study in prevention of recurrent GI bleeding.

www.futuremedicine.com 481 REVIEW – Reiter & Jilma

At present the current recommendation for the European Society of Cardiology (ESC) in patients who already have had a GI complication August 2004, evaluated 904 patients. Subjects while taking aspirin is to replace aspirin with received aspirin 325 mg daily and were assigned clopidogrel [39,65,99,220]. However, although there to either clopidogrel (300-mg loading dose and are several potential limitations of the recently 75 mg maintenance doses for 1 month) or one published study [241,242], the observation of this of three loading- and maintenance- study clearly do not support the current recom- dose regimens [403]. There was no significant mendation that clopidogrel should be used for difference in bleeding between combined patients who already have had GI complications prasugrel groups and clopidogrel (1.7% prasug- while taking aspirin. rel vs. 1.2% clopidogrel; p = 0.77). Death, MI, stroke, clinical target vessel thrombosis and Development of new drugs interfering with severe recurrent ischemia nonsignificantly ADP-mediated platelet reaction favored prasugrel (7.2% prasugrel vs. 9.4% The development of new ADP-receptor antago- clopidogrel; p = 0.31) without a dose–response nists possessing high potency and platelet selec- effect [252]. Despite limitations, predominantly tivity provides a new pharmacologic approach due to the exploratory nature of the trial, the for modulating platelet reactivity with the results are interesting and promising and clini- potential to prevent arterial thrombosis. cally significant differences in efficacy have to A more recent and related P2Y12 receptor be established in the future by the Triton antagonists, AR-C69931MX [243], is found to be a TIMI-38 trial. Patients will be followed on highly potent and selective antagonist at the maintenance therapy for 12 months. In this P2Y12 receptor that, unlike clopidogrel, is active trial, 13.000 patients with ACS will be enroled in vitro [243–245]. AR-C69931MX reversibly inhib- and randomized to either prasugrel or standard its aggregation, granule secretion, P-selectin doses of clopidogrel. The primary end point expression and procoagulant activity induced by will be the composite of cardiovascular death, agonists other than ADP, including , MI, and stroke. The secondary end point will thrombin receptor-activating peptide and colla- include bleeding, recurrent ischemia and urgent gen [244,246]. In a canine model, AR-C69931MX target-vessel revascularization [403]. administered as an intravenous infusion Since it is known that P2Y1-null mice display (4.0 µg/kg/min) was shown to prevent occlusive strong resistance to the thromboembolism arterial thrombus formation in vivo in response to induced by intravenous injection of ADP, a mix- a deep arterial lesion in the canine carotid artery ture of collagen and adrenalin [253,254] and [247]. The first Phase II study of intravenous AR- thromboplastin [255] the P2Y1 receptor represents C69931MX in patients with ACS showed that it a potential pharmacological target for anti- was well tolerated as adjunctive therapy, including thrombotic drugs. Indeed, the administration of and aspirin, with effective inhibition of the reversible P2Y1 receptor antagonist ADP-induced platelet aggregation, rapid onset of MRS2179 to mice was shown to strongly inhibit action and a plasma half-life of only several min- ADP-induced aggregation [253,255,256]. As both utes [245]. Moreover, it was demonstrated that, in P2Y1 and Y12 antagonists alone can potently both healthy volunteers and patients with ACS, inhibit ADP-induced platelet activation [257,258] AR-C69931MX inhibited ADP-induced platelet it was of interest to investigate whether there is a aggregation, P-selectin expression and platelet- synergistic effect of these antagonists. In a leukocyte conjugate formation in vitro and recently published study a synergistic effect in vivo, whereas aspirin had no effect on any of regarding inhibition of ADP-induced platelet these responses [248]. activation was obtained with the combination of Another novel P2Y12 receptor the P2Y12 antagonist AR-C69931MX and the antagonist is CS-747 [249]. CS-747 inactivates P2Y1 antagonist MRS2179 [259]. Moreover, a the P2Y12 receptor through its active metabo- strong synergistic effect in inhibition of lite, by the same mechanism such as clopidogrel thrombin-induced platelet activation with com- [249–251]. Currently, CS-747 (prasugrel) is being bination of AR-C69931MX and the thrombin investigated for the treatment of patients with inhibitor melagatran could also be shown. ACS who undergo PCI. The Phase IIb Whether the synergistic effect in vitro also results Utilization of to Block platelets in an improved antithrombotic effect in vivo Optimally – Thrombolysis In Myocardial Inf- with or without an increased risk of bleeding arction (JUMBO-TIMI) 26 trial, presented at should be established in further studies.

482 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

Another promising target of ADP-receptor therapy with GPIIb/IIIa antagonists has recently antagonists could be the P2X1 receptor. An excel- been published [266]. Recommendations for the lent review has recently been published focusing use of GPIIb/IIIa antagonists in patients with on the role of P2X1 receptors in platelet activa- cardiovascular diseases are available from tion [210]. Indeed, results from functional evidenced-based guidelines [99,221,220]. genomic studies showed that P2X1 receptors sig- nificantly increased the risk of thrombosis PIIb/IIIa antagonists in cardiovascular disease whereas knockout of P2X1 receptors led to a Intravenous GPIIb/IIIa antagonists for PCI in high-risk reduced risk of thrombosis [260]. Moreover, it has patients and in patients with ACS without been suggested from both knock-in and -out persistent ST-segment elevation studies, that this receptor appears to contribute to As previously reported, there is outstanding evi- platelet activation under condition of high shear dence supporting the utility of intravenous rates such as found in the arterial circulation [210]. GPIIb/IIIa inhibition, mainly for , as an To summarize, the ATP analogs of the AR-C adjunct to aspirin and heparin in high-risk series, which are potent competitive ADP-recep- patients undergoing PCI and in patients with tor antagonists, have proved to be efficient plate- ACS without persistent ST-segment elevation let drugs in animal models and some of them are before revascularization [266]. Indeed, the use of in Phase II clinical trials for the treatment of GPIIb/IIIa antagonists has been validated in dedi- ACS. Moreover, in addition to the clopidogrel or cated trials for patients undergoing PCI with or the AR-C compound-sensitive P2Y12 receptor, without stenting [267–271]. However, a recent clini- the P2Y1 and X1 receptor are promising potential cal trial was designed to evaluate the possible addi- targets for new antithrombotic drugs. tional benefit of the GPIIb/IIIa antagonist abciximab in 2159 patients at low-to-intermedi- GPIIb/IIIa receptor antagonists ate risk undergoing PCI pretreated with 600 mg It is well established that the expression of of clopidogrel [272]. The trial showed no addi- GPIIb/IIIa on the platelet surface is the final tional benefit of abciximab in the composite pri- common pathway of platelet aggregation [261]. mary outcome of death, MI or target-vessel These receptors recognise an arginine-glycine- revascularization within the first 30 days in those aspartic-acid sequence contained in adhesive patients. Moreover, the role in the purely medical molecules such as fibrinogen and von Wille- management of patients with ACS without brand factor (vWF). When platelets get acti- persistent ST-segment elevation is less certain [266]. vated, GPIIb/IIIa is converted into a functional receptor, binding these proteins and allowing Intravenous GPIIb/IIIa antagonists for platelets to aggregate and form a hemostatic plug primary percutaneous transluminal coronary [8]. Thus, this receptor has become the target of angioplasty (with or without stenting) in acute novel antiplatelet drugs [262]. Murine mono- myocardial infarction clonal antibodies were the first antagonists of the On the basis of trials investigating the possible GPIIb/IIIa receptor to be developed [263]. Plate- benefit of intravenous GPIIb/IIIa antagonists for let aggregation is highly inhibited by the block- primary PTCA in acute MI it is suggested that ade of 80% of the surface GPIIb/IIIa receptors there is no definite indication for these drugs as an [264]. Three classes of GPIIb/IIIa antagonists adjunct to primary PTCA, as this treatment have been developed [265]: modality was demonstrated to have variable • Murine–human chimeric antibodies, such as effects [266]. Indeed, there had been debate in this abciximab field since the results of the four largest trials [273–276] did not support the benefit of abcix- • Synergistic peptide forms, such as imab at the primary end point timing of • Synthetic nonpeptide forms, such as 6 months. Moreover, the study designs and meth- and lamifiban odology were quite variable in the clinical setting The development of this new class of drugs that of these trials. Finally, the data on tirofiban and blocks fibrinogen binding to the GPIIb/IIIa eptifibatide in primary PCI are far more limited receptors (Figure 1) has raised the possibility that than for abciximab [99,277]. Thus, given the size these potent agents may reduce thrombotic com- and limitations of the available data set, the rou- plications in ACS or after PCI. A large number tine administration of GPIIb/IIIa antagonists of trials have been carried out so far and an excel- with PCI (with or without stenting) in patients lent review on the current status of antiplatelet with MI is still a matter of debate [266,278]. www.futuremedicine.com 483 REVIEW – Reiter & Jilma

However, a meta-analysis of pooled data from coronary patency with favorable trends for clini- the previous four trials [277] and of the more cal outcomes. The early administration of recent Abciximab and Carbostent Evaluation GPIIb/IIIa antagonists was associated with a (ACE) trial (Table 7) [279] showed an overall 46% 28% relative reduction of mortality from 4.7 to reduction in death, reinfarction and target vessel 3.4%, which was not significant but consistent revascularization; a 34% reduction in death or with similar trends for reinfarction and the reinfarction; and a 26% reduction in death at 30 composite ischemic end point. days [277]. In all of the trials except the Controlled To summarize, although there has been con- Abciximab and Device Investigation to Lower troversy in the routine administration of Late Angioplasty Complications trial (CADIL- GPIIb/IIIa antagonists with PCI [277,278], it is LAC), there was support for abciximab treatment now recommended that treatment with abcixi- benefit for reduction of death or reinfarction at mab is started as early as possible in patients 6 months’ follow-up, and in both Abciximab undergoing primary PCI (with or without stent- before Direct angioplasty and stenting in Myocar- ing) [277,281] and treatment with tirofiban or epti- dial Infarction Regarding Acute and Long-term fibatide may be considered before primary PCI follow-up (ADMIRAL) and ACE the differences (with or without stenting) [99]. Moreover, the were statistically significant (Table 7). meta-analysis by Topol and colleagues [277] clearly Moreover, concerning the effects of early ver- indicates that catheter-based reperfusion with sus delayed administration of GPIIb/IIIa antago- adjunctive abciximab should be considered the nists in patients with ST-segment elevation MI preferred reperfusion therapy for acute MI and undergoing PCI a meta-analysis of 6 randomized do not justify a different level of recommendation trials [283] showed that the early administration for the use of GPIIb/IIIa inhibitors in acute MI of abciximab or tirofiban appeared to improve according to whether or not a stent is implanted.

Table 7. Clinical studies with intravenous GPIIb/IIIa antagonists for PTCA in acute MI Trial Patients Subjects Treatment Primary end point Result Ref. (n)

RAPPORT Acute MI within 483 Abciximab 0.25 mg/kg Composite of death No difference in primary [273] 12 h and ST- bolus followed by a from any cause, 6-month endpoint, segment elevation 12-h infusion of nonfatal reinfarction, although abciximab in two contiguous 0.125 µg/kg/min + and any TVR within significantly reduced leads or a new stenting vs. placebo + 6 months, or death, reinfarction or complete left stenting composite of death, urgent TVR at all time bundle-branch reinfarction, and points measured. Major block pattern, urgent TVR at 7 and bleeding was significantly referred for 30 days. higher with abciximab coronary than with placebo (16.6 angioplasty vs. 9.5%; p = 0.02) ISAR-2 Patients 401 Abciximab 0.25 mg/kg Composite of death, The primary 30-day end [274] undergoing bolus followed by 12-h reinfarction, and TVR point was reached in stenting within 48 infusion of 10 µg/min + at 30 days 5.0% of the abciximab h after onsets of reduced-dose heparin + group and in 10.5% of symptoms of acute stenting vs. standard the control group MI and ST- dose heparin+ stenting (p = 0.038). During 1-year segment elevation follow-up, there was no in two or more additional benefit from a contiguous leads reduction in TVR nor did abciximab reduce angiographic restenosis.

ACE: Abciximab and Carbostent Evaluation trial; ACS: Acute coronary syndromes; ADMIRAL: Abciximab before Direct angioplasty and stenting in Myocardial Infarction Regarding Acute and Long-term follow-up trial; CADILLAC: Controlled Abciximab and Device Investigation to Lower Late Angioplasty Complications trial; ECG: Electrocardiography; ISAR: Intracoronary Stenting and Angiographic Results trial; MI: Myocardial infarction; PTCA: Percutaneous transluminal coronary angioplasty; RAPPORT: ReoPro And Primary PTCA Organization and Randomized Trial; TVR: Target vessel revascularization.

484 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

Table 7. Clinical studies with intravenous GPIIb/IIIa antagonists for PTCA in acute MI (Cont.). Trial Patients Subjects Treatment Primary end point Result Ref. (n) ADMIRAL Patients 300 Abciximab 0.25 mg/kg Composite of death, Significant difference in [275] undergoing bolus, followed by a reinfarction, or primary 30-day endpoint stenting within 12-h infusion of urgent TVR at (6% for abciximab and 12 h after onset of 0.125 µg/kg/min + 30 days 14,6% for placebo; stenting vs. placebo + p = 0.01) which remained stenting significant through 6-month follow up (7.4 vs. 15.9%; p=0.02) One major bleeding event occurred in the abciximab group compared to none in the placebo group The primary 6-month endpoint occurred in 20.0% after PTCA, 16.5% after PTCA + abciximab, 11.5 % after stenting, and 10.2% after stenting + abciximab (p < 0.001) CADILLAC symptoms of acute 2082 Abciximab + PTCA vs. Composite of death, No differences among the [276] MI and ST-segment PTCA alone vs. reinfarction, groups in the rates of elevation in abciximab + multiLink disabling stroke, and death, stroke or two contiguous stent vs. multiLink stent ischemia-driven reinfarction, although leads alone revascularization of abciximab did reduce Symptoms of MI the target vessel at rates of repeated >30min within 6months revascularization during 12 h and lytic the first week postinitial eligible ECG procedure. The primary 30-day endpoint occurred in 10.5% after stenting, and 4.5% after abciximab + stenting (p = 0.023) ACE Symptoms of MI 400 Abciximab 0.25 mg/kg Composite of death At 6 months, the [279] >30 min bolus, followed by a from any cause, cumulative difference in associated with ST- 12-h infusion of reinfarction, TVR, mortality between the segment elevation 0.125 µg/kg/min + and stroke at 30 days groups increased (4.5 vs. in two or more stenting vs. stenting 8%), and the incidence of contiguous leads alone the composite of within 6 h or 6-month death and between 6 and reinfarction was lower in 24 h if there was the abciximab group than evidence of in the stent only group continuing (5.5% and 13.5%,p = ischemia. Patients 0.006). Six-month repeat with cardiogenic TVR and restenosis rates shock due to were similar between the predominant two groups. ventricular failure were also included ACE: Abciximab and Carbostent Evaluation trial; ACS: Acute coronary syndromes; ADMIRAL: Abciximab before Direct angioplasty and stenting in Myocardial Infarction Regarding Acute and Long-term follow-up trial; CADILLAC: Controlled Abciximab and Device Investigation to Lower Late Angioplasty Complications trial; ECG: Electrocardiography; ISAR: Intracoronary Stenting and Angiographic Results trial; MI: Myocardial infarction; PTCA: Percutaneous transluminal coronary angioplasty; RAPPORT: ReoPro And Primary PTCA Organization and Randomized Trial; TVR: Target vessel revascularization.

www.futuremedicine.com 485 REVIEW – Reiter & Jilma

IIntravenous GPIIb/IIIa antagonists as adjuncts to SM-20302 [288,289], ME3277 [290,291], murine lytic therapy in acute myocardial infarction 7E3 F(ab´)(2) [292,293] and SDZ–GPI 562 [294] The use of GPIIb/IIIa antagonists as adjuncts to have been reported to preserve microvascular lytic therapy in acute MI is not recommended patency in different animal models of acute [266] as the results of the large GUSTO V study ischemic stroke and they may have neuroprotec- were disappointing with no difference in mortal- tive properties. More recently, FK419, a novel ity rates [282]. Indeed, the clinical efficacy of nonpeptide GPIIb/IIIa antagonist dose-depend- GPIIb/IIIa antagonists has been equivocal in ently shortened the time to first reperfusion and terms of mortality in patients with ST-elevated the total middle cerebral artery occlusion time MI, although a subgroup analysis of the ADMI- and reduced ischemic brain damage in a guinea- RAL trial has suggested that the early adminis- pig model [295]. Thus, GPIIb/IIIa antagonists tration of abciximab may provide further benefit may be suitable as a single therapeutic or as an [280]. A meta-analysis of 16 randomized, control- adjunct therapeutic to thrombolysis with led trials demonstrated that the parenteral for the treatment of stroke, although GPIIb/IIIa antagonists significantly reduced bleeding risk will be a major concern. mortality at 48 to 96 h but mortality benefits of Results of pilot trials in the setting of acute 30 days and 6 months were not statistically sig- ischemic stroke with the three GPIIb/IIIa nificant [284]. However, the meta-analysis showed antagonists abciximab, tirofiban, and eptifi- that GPIIb/IIIa antagonists provided a consist- batide are promising [288,296–299]. To evaluate ent and sustained therapeutic benefit on death, the safety of abciximab in acute ischemic stroke MI, and revascularization in patients with a randomized, double-blind, placebo-control- ischemic heart disease. led, dose-escalation trial was conducted in patients presenting within 24 h following Limitations of intravenous GPIIb/IIIa antagonists ischemic stroke onset [300] who were rand- Although GPIIb/IIIa antagonists have been omized to receive either an escalating dose of shown to be beneficial, the drugs have a narrow abciximab or placebo. There were no identified therapeutic window with regard to potential cases of fatal or nonfatal major intracranial complications such as increased bleeding risk or hemorrhage within either 5 days or 3 months thrombocytopenia particularly for abciximab [265]. of randomization. Asymptomatic parenchymal However, across all clinical trials, the rate of life- hemorrhages were detected on poststudy agent threatening bleeding and intracranial haemor- computed tomography in 7% of patients rhage was less than 0.2% and the most common treated with abciximab and 5% of the placebo- bleeding site was the vascular access site [286]. treated patients. Overall, abciximab is consid- Critical thrombocytopenia, eventually leading to ered safe when administered up to 24 h after uncontrolled bleeding, has been estimated in stroke onset. Analysis of the pooled abciximab 0.4 to 1.6% patients treated with abciximab and data provided some preliminary evidence that severe thrombocytopenia does even occur in this agent might improve outcome after stroke, healthy volunteers [287]. In trials with other since the proportion of patients with minimal GPIIb/IIIa antagonists, the incidence of throm- residual disability at 90 days was higher in the bocytopenia is generally lower than 1% [287]. abciximab group. Overall, intravenous GPIIb/IIIa antagonists have The currently ongoing Safety of Tirofiban in become a mainstay in the treatment of patients acute Ischemic Stroke (SaTIS) trial, in which undergoing PCI and in these patients with ACS patients receive either tirofiban or placebo, will undergoing revascularization [266]. help to further evaluate the safety and efficacy of GPIIb/IIIa antagonists in the treatment of GPIIb/IIIa antagonists in cerebrovascular disease acute stroke [301]. Moreover, GPIIb/IIIa antago- Compared with coronary arterial diseases, only nists in combination with reduced doses of few trials have evaluated the efficacy and tolera- thrombolytic agents may have the potential for bility of platelet GPIIb/IIIa antagonists in improving safety and efficacy compared with patients with cerebrovascular diseases. On the standard recombinant tissue plasminogen acti- basis of experience in ACS, parenteral vator [298]. However, GPIIb/IIIa antagonists GPIIb/IIIa antagonists may have potential appli- cannot be recommended for general use in cations in the treatment of acute ischemic stroke patients with cerebrovascular disease before and as adjunctive therapy to carotid angioplasty. prospective randomized placebo-controlled Indeed, the administration of agents such as clinical trials are completed.

486 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

Table 8. Clinical studies with oral GPIIb/IIIa antagonists. Trial Patients Subjects Treatment Primary end Result Ref. (n) point

OPUS-TIMI 16 ACS within 10302 Orbofiban 50 mg Death, MI, Trial terminated prematurely [302] 72 h; history of twice daily vs. recurrent due to an unexpected increase cardiovascular orbofiban 50 mg ischemia, urgent in 30-day mortality in the disease, positive twice daily for revascularisatio orbofiban group cardiac markers 30 days, followed by or stroke Major or severe bleeding was or ECG changes, 30 mg twice daily higher with orbofiban; it peripheral or vs. placebo occurred in 2.0%, 3.7% and cerebrovascular 4.5% of patients disease, or DM SYMPHONY ACS after 9233 Aspirin 80 mg twice Death, MI, and No difference between aspirin [303] stabilisation daily vs. low-dose severe recurrent group (9.8%), low- (10.1%), or twice daily ischemia at high-dose sibrafiban (10.1%). vs. high-dose 90 days Sibrafiban was associated with sibrafiban twice daily increased major bleeding SYMPHONY-2 ACS after 6671 Aspirin 80mg twice Death, MI and No difference between aspirin [304] stabilisation daily vs. low-dose severe recurrent (9.3%), low-dose sibrafiban sibrafiban plus ischemia plus aspirin (9.2%) or high-dose aspirin 80 mg twice sibrafiban (10.5%) daily vs. high-dose Aspirin with low-dose sibrafiban twice daily sibrafiban caused more bleeding than aspirin alone. There was a trend toward increased mortality in this group and a significant increase in the high-dose arm EXCITE Patients 7232 Xemilofiban 20 mg Death, MI and No difference between placebo [305] undergoing PCI or placebo before recurrent (13.5%), 10 mg (13.9%) or PCI, followed by revascularization 20 mg xemilofiban (12.7%) at Xemilofiban 10 mg at 30 and 182 days. Significant increase in three-times daily vs,. 182 days major bleeding in the xemilofiban 20 mg xemilofiban group three-times daily vs. placebo BRAVO Recent ACS, 9200 Lotrafiban 30 mg + Death, MI, Stopped at interim analysis [306] stroke, TIA, or aspirin twice daily vs. stroke, recurrent because lotrafibran had a peripheral lotrafiban 50 mg+ ischemia higher mortality than placebo vascular disease aspirin twice daily requiring (2.7 vs. 2.0%). Lotrafiban was vs. placebo hospitalization associated with a higher and urgent mortality, more major bleeding, revascularization and a greater risk of serious thrombocytopenia ACS: Acute coronary syndromes; BRAVO: Blockade of the glycoprotein IIb/IIIa Receptor to Avoid Vascular Occlusion trial; DM: Diabetes mellitus; ECG: Electrocardiography; EXCITE: Evaluation of oral Xemilofiban in ControllIng Thrombotic Events; MI: Myocardial infarction; OPUS-TIMI: Orbofiban in Patients with Unstable coronary Syndromes – Thrombolysis In Myocardial Infarction; PCI: Percutaneous coronary intervention; SYMPHONY: Sibrafiban versus aspirin to Yield Maximum Protection from ischemic Heart Events post–acute CorONary sYndromes trial; TIA: Transient ischemic attack.

Oral antagonists of GPIIb/IIIa receptors of patients with ischemic heart disease treated To further test the clinical efficacy of GPIIb/IIIa with oral GPIIb/IIIa antagonists (Table 8) [302–306]. antagonists, oral agents of this interesting class In contrast to the favorable results of the intrave- of drugs have been developed but only led to nous GPIIb/IIIa antagonists, the long-term use of disappointing clinical results. oral GPIIb/IIIa antagonists showed no benefit in To date, five large Phase III trials including reducing major adverse cardiac events, and signifi- 45,523 patients investigated the clinical outcomes cantly increased mortality in these patients. www.futuremedicine.com 487 REVIEW – Reiter & Jilma

Indeed, a meta-analysis of trials with oral An excellent review on antiplatelet therapy GPIIb/IIIa antagonists demonstrated a 31% provides some possible explanations for this ‘dark increase in mortality rate [307]. Despite the lack of side’ of GPIIb/IIIa antagonists [312]. In general, clinical benefit, excess bleeding complications the disappointing results of the oral agents were associated with the oral GPIIb/IIIa antago- sibrafiban, orbofiban and xemilofiban may be nists in these trials and, as a consequence, new oral due to suboptimal levels of platelet inhibition, drugs of this class were synthesized and tested in leading to paradoxical agonist-induced platelet dose–response and safety studies. activation and/or vascular inflammation [312–315]. The safety and tolerability of roxifiban, a sec- Indeed, it could be possible that the short half-life ond-generation oral nonpeptide platelet of oral GPIIb/IIIa antagonists function as antago- GPIIb/IIIa receptor antagonist, was tested in nists at peak concentrations (causing bleeding) 98 patients with a history of chronic stable but at lower concentrations they might act as par- angina pectoris [308]. Roxifiban-induced inhibi- tial agonists via a mechanism known as ‘platelet tion of platelet aggregation was dose dependent escape’ [316,317]. Thus, these agents may cause and sustained throughout the study period: bleeding at peak levels and promote thrombosis higher drug dosages correlated with higher levels at trough levels in the same patient. Indeed, pre- of platelet inhibition and a higher incidence of vious studies have demonstrated that chronic minor bleeding events. No serious adverse inhibition of platelets with oral GPIIb/IIIa antag- events were observed at any dosage. However, onists might be associated with platelet receptor the Roxifiban Oral Compound Kinetics Evalua- activation, continued procoagulant activity or tion Trials (ROCKET)-I trial demonstrated enhanced platelet–neutrophil interaction that, despite achieving sustained inhibition of [318–321]. Apart from that, even intravenous platelet aggregation, therapy with roxifiban was GPIIb/IIIa antagonists such as tirofiban have lim- associated with over expression or phasic ited efficacy under conditions of platelet activa- changes of major platelet receptors in patients tion, high vWF release, high shear stress and with CAD [309]. Another second-generation oral physiological calcium concentrations [322]. platelet GPIIb/IIIa receptor antagonist is the In a more recent study, it has been demonstrated prodrug UR-3216. UR-3216 maintains a high that the oral GPIIb/IIIa antagonist roxifiban signif- level of inhibition of platelet aggregation and, icantly decreased ADP- and collagen-induced due to a small peak-to-trough ratio, severe platelet aggregation, although platelet receptors bleeding is avoided [310]. Moreover, UR-3216 such as GPIIb/IIIa, P-selectin, and plate- induces no prothrombotic activity in human let/endothelial cell-adhesion molecule (PECAM)- platelets, distinctly different from orbofiban and 1 were paradoxically activated, monotherapy with other small molecule antagonists [311]. Although aspirin resulted in a mild, but consistent, inhibi- UR-3216 may be a promising orally-active tion of these receptors. Moreover, oral GPIIb/IIIa GPIIb/IIIa antagonist, hardly any data on this antagonists may have proinflammatory potency at drug are available. subtherapeutic levels, which could be an important To summarize, oral GPIIb/IIIa antagonists are factor in the toxicity of these agents [312]. not effective in reducing ischemic events when Overall, various factors have been proposed to used on a long-term basis after ACS and, cur- explain their failure, such as: rently, it seems unlikely that second-generation • Low affinity for the receptor oral platelet GPIIb/IIIa receptor antagonists will • Large peak-to-trough ratio be brought into Phase III testing. • Low bioavailability Dark side of GPIIb/IIIa antagonists & • Partial agonist activity future directions • Proaggregatory effect Paradoxical platelet activation after treatment • The fact that oral GPIIb/IIIa antagonists don’t with oral GPIIb/IIIa antagonists may be seem to entice the clinical market responsible for and explain the increased rate of mortality and high incidence of acute Monitoring of platelet function thrombotic events in patients with CAD. Although the role of antiplatelet drugs in the Moreover, the inability to maintain stable sus- treatment of vascular disease is well established, tained platelet inhibition for chronic regimes there remains concern about aspirin contributes substantially to the withdrawal of resistance [323], the less well known clopidogrel this class of drugs. resistance [324], and nonresponsiveness to other

488 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

antiplatelet drugs such as GPIIb/IIIa antagonists stenting were resistant to clopidogrel (mean [366,325]. Thus, the monitoring of antiplatelet ther- ADP-induced platelet aggregation on day 6 of apy is becoming increasingly important and many treatment: 103 ± 8% of baseline) and, therefore, instruments have been, or will be, utilized as point- might be at an increased risk for recurrent cardio- of-care instruments for monitoring antiplatelet vascular events [332]. Otherwise, from a secondary therapy or for the assessment of bleeding risk [326]. post-hoc analysis it has been demonstrated that A single dose of 160 mg completely abolishes pretreatment platelet activity and clinical charac- platelet TXA2 production [324] and the same teristics were not associated with responsiveness effect can be progressively achieved with the to clopidogrel [333]. In this study, platelet func- chronic administration of daily doses of 30 to tion before and after clopidogrel therapy was 50 mg [29]; however, variable platelet responses to analyzed in all 544 individuals by conventional aspirin have been described. Indeed, based on aggregometry. Hypo- (4.8%) and hyper- measurements of platelet aggregation in response responders (4.3%) to clopidogrel, as determined to arachidonate and ADP, 5 and 24% of by change in ADP-induced platelet aggregation, patients, respectively with stable cardiovascular did not significantly differ in clinical characteris- disease who were receiving 325 mg aspirin once tics from those whose responses were within the daily were defined as being ‘resistant’ and ‘sem- standard range. Moreover, platelet activity before iresponders’ [327]. The aspirin-resistant group the administration of clopidogrel, which was had an increased risk of death, MI, or cerebro- defined by baseline platelet aggregation response vascular accident during almost 2 years’ follow- to ADP, did not appear to be associated with the up. Another study using different techniques to response to clopidogrel. measure platelet aggregation, showed that 57% Measurement of platelet function by the rapid of a group of 88 patients with documented heart platelet function assay (Ultegra) has been found failure who had been treated with aspirin. to predict therapeutic response to GPIIb/IIIa 325 mg/day for over a month, showed ‘aspirin antagonists in patients with PCI [334]. Moreover, nonresponsiveness’ [328]. there have been several examples of the applica- The ADP receptor blocker clopidogrel reduces tion of the FDA-approved platelet function ana- the incidence of recurrent ischemic events in lyzer 100 (PFA-100) in therapeutic monitoring patients with ACS [220] and after coronary stent- [335]. In patients suffering from peripheral arterial ing [222]. Platelet inhibition with clopidogrel is occlusive disease poor responders to clopidogrel evident within two hours of an initial dose and (detected by the PFA-100) had an increased like- the maximal effect with an initial dose has been lihood of experiencing restenosis after percutane- noted to occur at 400 mg (40% platelet inhibi- ous angioplasty [336]. Patients with ST-elevated tion). In healthy volunteers ADP-induced plate- MI had significantly enhanced platelet function let aggregation was inhibited within 2 days when measured under high shear rates with the (platelet inhibition by ∼30%), and reached a PFA-100 [337] and standard doses of GPIIb/IIIa maximal inhibition (60% platelet inhibition) antagonists, particularly tirofiban, had a limited within 4 to 7 days of continued dosing impact on high shear-induced platelet formation (50–100 mg/day) of clopidogrel [156]. However, at physiologic Ca2+ concentration [322]. Moreover, clopidogrel ‘nonresponsiveness’ has been reported since GPIIb/IIIa antagonists have been shown to to be present in as few as 5% to as many as 56% dose dependently inhibit platelet of patients who are undergoing coronary stent- aggregation [322,338] one may speculate that the ing. Previous studies [329–331] labeled patients as inefficacy of these antiplatelet drugs could be due nonresponders based on the arbitrary definitions to insufficient therapeutic dosing. Indeed, as sug- of the change in ADP-induced platelet aggrega- gested by the Enhanced Suppression of the Plate- tion before and after the start of clopidogrel ther- let IIb/IIIa Receptor with Integrilin Therapy apy. Indeed, a significant number of (ESPRIT) trial [271], both medically treated cardiovascular events continue to occur [23,222]. In patients and patients undergoing PCI may bene- a more recent study, the antiplatelet effect of fit from higher dosages of GPIIb/IIIa antagonists. clopidogrel was studied prospectively in 60 con- However, the use and benefit of the monitoring secutive patients who underwent PCI with stent- of antiplatelet drugs in clinical settings are cur- ing for acute MI to determine whether variability rently controversially discussed [324] and further in response to clopidogrel affects clinical out- well-designed studies are needed to demonstrate comes. This study demonstrated that up to 25% its utility in different populations of patients with of patients undergoing primary PCI with vascular disease. www.futuremedicine.com 489 REVIEW – Reiter & Jilma

Potential • Administer a DDAVP infusion When bleeding occurs under treatment with • Transfuse platelet concentrates in case of antiplatelet agents, evaluation of platelet func- major or life-threatening bleeding or urgent tion may also help to guide further decision need for normalization of platelet function in making. In the acute event, platelet concentrates case of surgery. (PC) or desmopressin (DDAVP) may be helpful. Concerning the use of PC the pharmaco- Expert opinion & outlook kinetics of GPIIb/IIIa inhibitors is of impor- An ideal antiplatelet agent should specifically block tant practical implication [265]. The plasma thrombogenic platelet-dependent mechanisms in levels of unbound abciximab drop very rapidly vascular diseases without interfering with normal after administration. The rapid disappearance platelet functions that are required in hemostasis of free abciximab in plasma is an important and wound healing. Additionally, these agents consideration for reversing the therapeutic should be free of any major adverse events. effect. In the acute situation, the transfusion of Although, several antiplatelet strategies have platelets results in an immediate and partial already been developed or are under preclinical or normalization of platelet function, since the clinical investigation (Table 1) none of the available amount of unbound plasma abciximab availa- antiplatelet drugs meet all of these criteria. ble to inhibit transfused platelets is very small. Aspirin has been, and will be, the standard ref- In contrast to abciximab, the plasma concen- erence compound for long-term oral treatment of tration of eptifibatide molecules at peak dosing platelet hyper-reactivity, most notably in the sec- is very high relative to the number of ondary prevention of cardiovascular diseases [39]. GPIIb/IIIa molecules. As a result, newly trans- However, aspirin is neither selective for platelets fused platelets would probably be rapidly nor a potent antiplatelet compound. As platelet inhibited. Tirofiban is intermediate between activation occurs via several pathways that do not abciximab and eptifibatide in its peak molecu- rely on amplification by released TXA2, adding a lar concentration relative to the number of second antiplatelet drug to aspirin might have GPIIb/IIIa molecules. Thus, platelet transfu- additional benefits in some clinical circumstances, sions are possibly far more effective after abcix- but more research into this strategy is required. imab administration than following tirofiban The combination of modified-released dipyri- or eptifibatide infusion. damole and low-dose aspirin therapy has been DDAVP has been shown to accelerate approved by the FDA but recommendations for normalization of in vitro platelet dysfunction this class of antiplatelet drugs are induced by GPIIb/IIIa antagonists [238]. Com- controversial [162–165,202]. The reduction in non- bined use of platelet concentrates and DDAVP fatal stroke was derived merely from one large has been shown to additively enhance recovery trial [159], but this result was not supported by of normal platelet function after infusion of the findings for nonfatal stroke in other studies GPIIb/IIIa antagonists [338]. Thus, based on our or by the overall findings for nonfatal MI or vas- previous reports [338,339] and based on the poten- cular death [39]. Although dipyridamoles are con- tial necessity to administer large numbers of PC sidered to have some benefit in cerebrovascular [340] in case of bleeding induced by GPIIb/IIIa disease they have not been proven as an effective inhibitors cautious recommendations may be agent in cardiovascular disease. given: DDAVP should be used whenever bleed- Thienopyridines such as clopidogrel have been ing is suspected to stem from aspirin or shown to be beneficial in the treatment of vascular GPIIb/IIIa inhibitors. As long as there are no disease. Moreover, thienopyridines are the thera- clinical trials in bleeding patients, the authors peutic alternative in aspirin tolerance or resist- cautiously recommend the following course of ance. In combination with low-dose aspirin, action: clopidogrel has been shown to be more efficacious • Stop the GPIIb/IIIa infusion than aspirin alone, especially in patients with ACS [222,224,234] and several studies are currently • Obtain a platelet count and monitor activated planned or ongoing (Table 6). Newer ADP recep- partial thromboplastin (aPTT) or anti-FXa tor antagonists will probably become part of activity levels when anticoagulants are used clinical practice in the next few years. concomitantly Currently, the intravenous GPIIb/IIIa antago- • If possible, measure the degree of platelet nists are the most potent inhibitors of platelet inhibition with a rapid bedside test aggregation. Their use is restricted to patients

490 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

undergoing PCI and to patients with ACS before To summarize, there is a better understanding of revascularization but the role of GPIIb/IIIa the molecular events regulating thrombogenesis antagonists in the nonintervential management and ongoing investigations are exploring the value of ACS is more controversial [266]. Oral of novel antiplatelet agents in various preclinical or GPIIb/IIIa antagonists have failed to be benefi- clinical trials. Future developments might probably cial in the treatment of vascular disease and it include combined-mode agents targeting one or seems unlikely that there will be any further more steps in the thrombotic process to optimize development of these drugs. the efficacy and safety of antiplatelet therapy.

Highlights

• Aspirin: multiple, randomized, controlled clinical trials have shown a clinically significant decrease in cardiovascular morbidity and mortality in patients at risk of recurrent atherothrombotic events. Although the beneficial effect of aspirin in secondary prevention of ischemic events is well established, the role of primary prevention is less clear, particularly in women. • Dipyridamole: the combination of modified-released dipyridamole and low-dose aspirin therapy has been approved by the US Food and Drug Administration; however, recommendations for this class of antiplatelet drugs are controversial. • Thienopyridines: clopidogrel has been shown to be more efficacious than aspirin alone especially in patients with and several studies are currently planned or ongoing. Clopidogrel should not be used when aspirin causes gastrointestinal (GI) intolerance, because aspirin plus esomeprazole was definitely superior to clopidogrel for the prevention of recurrent GI bleeding among patients with a history of aspirin-induced ulcer bleeding. • GP IIb/IIIa antagonists: a large number of trials have been carried out with GPIIb/IIIa antagonists and several studies have shown the benefit of GP IIb/IIIa inhibitors in patients undergoing percutaneous coronary intervention (PCI) and in patients with acute coronary syndrome (ACS) before revascularisation. In contrast, the oral GPIIb/IIIa antagonists have failed to be beneficial in the treatment of vascular disease and it seems unlikely that there will be any further development of these drugs. • Monitoring of platelet function: with the increasing number of anti-platelet drugs, the monitoring of antiplatelet therapy is becoming increasingly important and many devices have been or will be studied as point-of-care instruments for monitoring antiplatelet therapy or assessing bleeding risk • Potential antidotes: when bleeding occurs under treatment with antiplatelet agents evaluation of platelet function may help to guide further decision making. Combined use of platelet concentrates and desmopressin has been shown to additively enhance recovery of normal platelet function after infusion of GPIIb/IIIa antagonists, thus, in the acute event or desmopressin and/or platelet concentrates may be helpful.

Bibliography 6. Santos MT, Valles J, Marcus AJ et al. antibody Fab fragments in nonhuman Papers of special note have been highlighted as of Enhancement of platelet reactivity and primates. Arterioscler. Thromb. Vasc. Biol. interest (•) or of considerable interest (••) to modulation of production by intact 20(5), 1347–1353 (2000). readers. erythrocytes. A new approach to platelet 12. Yeh CH, Chang MC, Peng HC, Huang TF. 1. Pearson JD. Vessel wall interactions regulating activation and recruitment. J. Clin. Invest. Pharmacological characterization and thrombosis. Br. Med. Bull. 50(4), 776–788 87(2), 571–580 (1991). antithrombotic effect of agkistin, a platelet (1994). 7. Plow EF, D’Souza SE, Ginsberg MH. glycoprotein Ib antagonist. Br. J. Pharmacol. 2. Ashby B, Daniel JL, Smith JB. Mechanisms of binding to GPIIb-IIIa: a status 132(4), 843–850 (2001). platelet activation and inhibition. Hematol. report. Semin. Thromb. Hemost. 18(3), 13. Wu D, Meiring M, Kotze HF, Deckmyn Oncol. Clin. North Am. 4(1), 1–26 (1990). 324–332 (1992). H, Cauwenberghs N. Inhibition of 3. Davies MJ, Thomas A. Thrombosis and acute 8. Clemetson KJ. Platelet activation: signal platelet glycoprotein Ib, coronary-artery lesions in sudden cardiac transduction via membrane receptors. glycoprotein IIb/IIIa, or both by ischemic death. N. Engl. J. Med. 310(18), Thromb. Haemost. 74(1), 111–126 (1995). monoclonal antibodies prevents arterial 1137–1140 (1984). 9. Ruggeri ZM, Saldivar E. Platelets, thrombosis in baboons. Arterioscler. 4. Fressinaud E, Sakariassen KS, Rothschild C, hemostasis and thrombosis. In: Von Thromb. Vasc. Biol. 22(2), 323–328 Baumgartner HR, Meyer D. Shear rate- Willebrand Factor and the Mechanisms of (2002). dependent impairment of thrombus growth Platelet Functions. Ruggeri ZM (Ed.). 14. Gurevitz O, Goldfarb A, Hod H et al. on collagen in nonanticoagulated blood from Springer, Berlin, Germany 1–32 (1998). Recombinant von Willebrand factor patients with and 10. Chang MC, Lin HK, Peng HC, Huang TF. fragment AR545C inhibits platelet hemophilia A. Blood 80(4), 988–994 (1992). Antithrombotic effect of crotalin, a platelet aggregation and enhances thrombolysis 5. Saelman EU, Nieuwenhuis HK, Hese KM membrane glycoprotein Ib antagonist from with rtPA in a rabbit thrombosis model. et al. Platelet adhesion to collagen types I venom of Crotalus atrox. Blood 91(5), Arterioscler. Thromb. Vasc. Biol. 18(2), through VIII under conditions of stasis and 1582–1589 (1998). 200–207 (1998). flow is mediated by GPIa/IIa (α2 β 1- 11. Cauwenberghs N, Meiring M, Vauterin S 15. Kageyama S, Yamamoto H, Nagano M, integrin). Blood 83(5), 1244–1250 (1994). et al. Antithrombotic effect of platelet Arisaka H, Kayahara T, Yoshimoto R. glycoprotein Ib-blocking monoclonal Antithrombotic effects and bleeding risk www.futuremedicine.com 491 REVIEW – Reiter & Jilma

of AJvW-2, a monoclonal antibody rupture and/or thrombi until the onset of 39. Antithrombotic Trialists’ Collaboration. against human von Willebrand factor. Br. acute myocardial infarction in humans: Collaborative meta-analysis of randomized J. Pharmacol. 122(1), 165–171 (1997). coronary angiographic findings within 1 week trials of antiplatelet therapy for prevention 16. Yamamoto H, Vreys I, Stassen JM, before the onset of infarction. Circulation of death, myocardial infarction, and stroke Yoshimoto R, Vermylen J, Hoylaerts MF. 102(17), 2063–2069 (2000). in high risk patients. Br. Med. J. 324(7329), Antagonism of vWF inhibits both injury 29. Patrono C. Aspirin as an antiplatelet drug. 71–86 (2002). induced arterial and venous thrombosis in N. Engl. J. Med. 330(18), 1287–1294 •• Largest ever meta-analysis – provides an the hamster. Thromb. Haemost. 79(1), (1994). update of trials of antiplatelet therapy, 202–210 (1998). 30. Folts JD, Schafer AI, Loscalzo J, Willerson particularly aspirin. 17. Kageyama S, Yamamoto H, Nakazawa H, JT, Muller JE. A perspective on the potential 40. Carne X, Arnau JM, Laporte JR. Aspirin, Yoshimoto R. Antihuman vWF monoclonal problems with aspirin as an antithrombotic ticlopidine, and stroke. Lancet 2(8660), antibody, AJvW-2 Fab, inhibits repetitive agent: a comparison of studies in an animal 442–443 (1989). coronary artery thrombosis without bleeding model with clinical trials. J. Am. Coll. 41. Dyken ML, Barnett HJ, Easton JD, Fields time prolongation in dogs. Thromb. Res. Cardiol. 33(2), 295–303 (1999). WS, Fuster V, Hachinski V, Norris JW, 101(5), 395–404 (2001). 31. Maalej N, Folts JD. Increased shear stress Sherman DG. Low-dose aspirin and stroke. 18. Libby P. Inflammation in atherosclerosis. overcomes the antithrombotic platelet ‘It ain’t necessarily so’. Stroke 23(10), Nature 420(6917), 868–874 (2002). inhibitory effect of aspirin in stenosed dog 1395–1399 (1992). 19. Takahashi R, Sekine N, Nakatake T. Influence coronary arteries. Circulation 93(6), 42. Matchar DB, McCrory DC, Barnett HJ, of monoclonal antiplatelet glycoprotein 1201–1205 (1996). Feussner JR. Medical treatment for stroke antibodies on in vitro human megakaryocyte 32. Rinder CS, Student LA, Bonan JL, Rinder prevention. Ann. Intern. Med. 12(1), 41–53 colony formation and proplatelet formation. HM, Smith BR. Aspirin does not inhibit (1994). Blood 93(6), 1951–1958 (1999). adenosine diphosphate-induced platelet 43. Chinese Acute Stroke Trial Collaborative 20. Alimardani G, Guichard J, Fichelson S, α-granule release. Blood 82(2), 505–512 Group. CAST: randomized placebo- Cramer EM. Pathogenic effects of (1993). controlled trial of early aspirin use in antiglycoprotein Ib antibodies on 33. Chronos NA, Wilson DJ, Janes SL, Hutton 20,000 patients with acute ischemic stroke. megakaryocytes and platelets. Thromb. RA, Buller NP, Goodall AH. Aspirin does Lancet 349(9066), 1641–649 (1997). Haemost. 88(6), 1039–1046 (2002). not affect the flow cytometric detection of 44. International Stroke Trial Collaborative 21. Cadroy Y, Hanson SR, Kelly AB et al. Relative fibrinogen binding to, or release of alpha- Group. The International Stroke Trial antithrombotic effects of monoclonal granules or lysosomes from, human (IST): a randomized trial of aspirin, antibodies targeting different platelet platelets. Clin. Sci. (Lond.) 87(5), 575–580 subcutaneous heparin, both, or neither glycoprotein-adhesive molecule interactions in (1994). among 19435 patients with acute ischemic nonhuman primates. Blood 83(11), 34. Lloyd J. Aspirin: how low is low dose? Aust. stroke. Lancet 349(9065), 1569–1581 3218–3224 (1994). Prescriber 19, 79–81 (1996). (1997). 22. Machin SJ, Clarke C, Ikemura O et al. A 35. Craven LL. Experiences with aspirin 45. Algra A, van Gijn J. Cumulative meta- humanized monoclonal antibody against vWF (Acetylsalicylic acid) in the nonspecific analysis of aspirin efficacy after cerebral A1 domain inhibits vWF:RcO of activity and prophylaxis of coronary thrombosis. Miss. ischemia of arterial origin. J. Neurol. platelet adhesion in human volunteers. Valley Med. J. 75(1), 38–44 (1953). Neurosurg. Psychiat. 66(2), 255 (1999). J. Thromb. Haemost. 1(Suppl. 1), OC328 36. ISIS-2 (Second International Study of 46. Hayden M, Pignone M, Phillips C, Mulrow (2003). Infarct Survival) Collaborative Group. C. Aspirin for the primary prevention of 23. Savage B, Almus-Jacobs F, Ruggeri ZM. Randomized trial of intravenous cardiovascular events: a summary of the Specific synergy of multiple substrate-receptor streptokinase, oral aspirin, both, or neither evidence for the US Preventive Services interactions in platelet thrombus formation among 17,187 cases of suspected acute Task Force. Ann. Intern. Med. 136(2), under flow. Cell 94(5), 657–666 (1998). myocardial infarction: ISIS-2. Lancet 161–172 (2002). 24. Kehrel B. Platelet-collagen interactions. Semin. 2(8607), 349–360 (1988). 47. Eidelman RS, Hebert PR, Weisman SM, Thromb. Hemost. 21(2), 123–129 (1995). 37. Baigent C, Collins R, Appleby P, Parish S, Hennekens CH. An update on aspirin in 25. Alberio L, Dale GL. Flow cytometric analysis Sleight P, Peto R. ISIS-2: 10 year survival the primary prevention of cardiovascular of platelet activation by different collagen types among patients with suspected acute disease. Ann. Intern. Med. 163(17), present in the vessel wall. Br. J. Haematol. myocardial infarction in randomized 2006–2010 (2003). 102(5), 1212–1218 (1998). comparison of intravenous streptokinase, 48. Awtry EH, Loscalzo J. Aspirin. Circulation 26. Wu D, Vanhoorelbeke K, Cauwenberghs N oral aspirin, both, or neither. The ISIS-2 101(10), 1206–1218 (2000). et al. Inhibition of the von Willebrand (Second International Study of Infarct 49. Peto R, Gray R, Collins R et al. (VWF)-collagen interaction by an antihuman Survival) Collaborative Group. Br. Med. J. Randomized trial of prophylactic daily VWF monoclonal antibody results in abolition 316(7141), 1337–1343 (1998). aspirin in British male doctors. Br. Med. J. of in vivo arterial platelet thrombus formation 38. Antiplatelet Trialists’ Collaboration. (Clin. Res. Ed.). 296(6618), 313–316 in baboons. Blood 99(10), 3623–3628 (2002). Collaborative overview of randomized trials (1988). 27. Jackson SP, Schoenwaelder SM. Antiplatelet of antiplatelet therapy-III: Reduction in 50. Steering Committee of the Physicians’ therapy: in search of the ‘magic bullet’. Nature venous thrombosis and pulmonary Health Study Research Group. Final report Rev. Drug Discov. 2(10), 775–789 (2003). embolism by antiplatelet prophylaxis among on the aspirin component of the ongoing 28. Ojio S, Takatsu H, Tanaka T et al. surgical and medical patients. Br. Med. J. Physicians’ Health Study. N. Engl. J. Med. Considerable time from the onset of plaque 308(6923), 235–246 (1994). 321(3), 129–135 (1989).

492 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

51. Medical Research Council’s General 61. US Preventive Services Task Force. Aspirin 71. Taylor DW, Barnett HJ, Haynes RB et al. Practice Research Framework. Thrombosis for the primary prevention of Low-dose and high-dose acetylsalicylic acid prevention trial: randomized trial of low- cardiovascular events: recommendation for patients undergoing carotid intensity oral anticoagulation with warfarin and rationale. Ann. Intern. Med. 136(2), endarterectomy: a randomized controlled and low-dose aspirin in the primary 157–160 (2002). trial. ASA and Carotid Endarterectomy prevention of ischemic heart disease in men 62. Patrono C, Coller B, FitzGerald GA, Hirsh J, (ACE) Trial Collaborators. Lancet at increased risk. Lancet 351(9098), Roth G. Platelet-active drugs: the relationships 353(9171), 2179–2184 (1999). 233–241 (1998). among dose, effectiveness, and side effects: the 72. Bertrand ME. Provision of cardiovascular 52. Hansson L, Zanchetti A, Carruthers SG Seventh ACCP Conference on protection by ACE inhibitors: a review of et al. Effects of intensive blood-pressure Antithrombotic and Thrombolytic Therapy. recent trials. Curr. Med. Res. Opin. 20(10), lowering and low-dose aspirin in patients Chest 126(Suppl. 3), S234–S264 (2004). 1559–1569 (2004). with hypertension: principal results of the 63. Buerke M, Pittroff W, Meyer J, Darius H. 73. Stauss HM, Zhu YC, Redlich T et al. Hypertension Optimal Treatment (HOT) Aspirin therapy: optimized platelet Angiotensin-converting enzyme inhibition in randomized trial. HOT Study Group. inhibition with different loading and infarct-induced heart failure in rats: Lancet 351(9118), 1755–1762 (1998). maintenance doses. Am. Heart J. 130(3 bradykinin versus angiotensin II. 53. de Gaetano G; Collaborative Group of the Pt 1), 465–472 (1995). J. Cardiovasc. Risk. 1(3), 255–262 (1994). Primary Prevention Project. Low-dose 64. Adams HP Jr, Adams RJ, Brott T et al. and 74. Zhu P, Zaugg CE, Hornstein PS, Allegrini aspirin and vitamin E in people at the Stroke Council of the American Stroke PR, Buser PT. Bradykinin-dependent cardiovascular risk: a randomized trial in Association. Guidelines for the early cardioprotective effects of losartan against general practice. Collaborative Group of management of patients with ischemic ischemia and reperfusion in rat . the Primary Prevention Project. Lancet stroke: a scientific statement from the Stroke J. Cardiovasc. Pharmacol. 33, 785–790 357(9250), 89–95 (2001). Council of the American Stroke Association. (1999). 54. Ridker PM, Cook NR, Lee IM et al. A Stroke 34(4), 1056–1083 (2003). 75. Blais C Jr, Drapeau G, Raymond P et al. randomized trial of low-dose aspirin in the 65. Harrington RA, Becker RC, Ezekowitz M Contribution of angiotensin-converting primary prevention of cardiovascular et al. Antithrombotic therapy for coronary enzyme to the cardiac metabolism of disease in women. N. Engl. J. Med. artery disease: the Seventh ACCP bradykinin: an interspecies study. Am. J. 352(13), 1293–1304 (2005). Conference on Antithrombotic and Physiol. 273(5 Pt 2), H2263–H2271 (1997). 55. Pearson TA, Blair SN, Daniels SR et al. Thrombolytic Therapy. Chest 76. Gainer JV, Morrow JD, Loveland A, King AHA Guidelines for Primary Prevention of 126(Suppl. 3), S513–S548 (2004). DJ, Brown NJ. Effect of bradykinin-receptor Cardiovascular Disease and Stroke: 2002 66. Lee TK, Chen YC, Lien IN, Liu MC, blockade on the response to angiotensin- Update: Consensus Panel Guide to Huang ZS. Inhibitory effect of converting- in normotensive Comprehensive Risk Reduction for Adult acetylsalicylic acid on platelet function in and hypertensive subjects. N. Engl. J. Med. Patients Without Coronary or Other patients with completed stroke or reversible 339(18), 1285–1292 (1998). Atherosclerotic Vascular Diseases. ischemic neurologic deficit. Stroke 19(5), 77. Nguyen KN, Aursnes I, Kjekshus J. American Heart Association Science 566–570 (1988). Interaction between enalapril and aspirin on Advisory and Co-ordinating Committee. 67. Koudstaal PJ, Ciabattoni G, van Gijn J et al. mortality after acute myocardial infarction: Circulation 106(3), 388–391 (2002). Increased thromboxane biosynthesis in subgroup analysis of the Co-operative New 56. Mosca L, Appel LJ, Benjamin EJ et al. and patients with acute cerebral ischemia. Stroke Scandinavian Enalapril Survival Study II the American Heart Association. Evidence- 24(2), 219–223 (1993). (CONSENSUS II) Am. J. Cardiol. 79(2), based guidelines for cardiovascular disease 68. Farrell B, Godwin J, Richards S, Warlow C. 115–119 (1997). prevention in women. Circulation 109(5), The United Kingdom transient ischemic 78. Al-Khadra AS, Salem DN, Rand WM, 672–693 (2004). attack (UK-TIA) aspirin trial: final results. Udelson JE, Smith JJ, Konstam MA. 57. Lauer MS. Clinical practice. Aspirin for J. Neurol. Neurosurg. Psychiat. 54(12), Antiplatelet agents and survival: a cohort primary prevention of coronary events. 1044–1054 (1991). analysis from the Studies of Left Ventricular N. Engl. J. Med. 346(19), 1468–1474 69. Dutch TIA Trial Study Group. A Dysfunction (SOLVD) trial. J. Am. Coll. (2002). comparison of two doses of aspirin (30 vs. Cardiol. 31(2), 419–425 (1998). 58. Marso SP. Optimizing the diabetic 283 mg a day) in patients after a transient 79. Peterson JG, Topol EJ, Sapp SK, Young JB, formulary: beyond aspirin and insulin. ischemic attack or minor ischemic stroke. Lincoff AM, Lauer MS. Evaluation of the J. Am. Coll. Cardiol. 40(4), 652–661 N. Engl. J. Med. 325(18), 1261–1266 effects of aspirin combined with angiotensin- (2002). (1991). converting enzyme inhibitors in patients with 59. Landolfi R, Marchioli R, Kutti J et al.; 70. Mehta SR, Yusuf S, Peters RJ et al.; . Am. J. Med. 109(5), European collaboration on low-dose Clopidogrel in Unstable angina to prevent 371–377 (2000). aspirin in polycythemia vera investigators. Recurrent Events trial (CURE) 80. Aumegeat V, Lamblin N, de Groote P et al. Efficacy and safety of low-dose aspirin in Investigators. Effects of pretreatment with Aspirin does not adversely affect survival in polycythemia vera. N. Engl. J. Med. 350(2), clopidogrel and aspirin followed by long- patients with stable congestive heart failure 114–124 (2004). term therapy in patients undergoing treated with Angiotensin-converting enzyme 60. Hennekens CH. Update on aspirin in the percutaneous coronary intervention: the inhibitors. Chest 124(4), 1250–1258 (2003). treatment and prevention of cardiovascular PCI-CURE study. Lancet 358(9281), 81. Oosterga M, Anthonio RL, de Kam PJ, disease. Am. J. Manag. Care 8(Suppl. 22), 527–533 (2001). Kingma JH, Crijns HJ, van Gilst WH. S691–S700 (2002). Effects of aspirin on angiotensin-converting www.futuremedicine.com 493 REVIEW – Reiter & Jilma

enzyme inhibition and left ventricular 92. Fredduzzi S, Mariucci G, Tantucci M, Del 103. Hawkey CJ. COX-2 inhibitors. Lancet dilation one year after acute myocardial Soldato P, Ambrosini MV. Nitroaspirin 353(9149), 307–314 (1999). infarction. Am. J. Cardiol. 81(10), (NCX4016) reduces brain damage induced 104. Homoncik M, Malec M, Marsik C et al. 1178–1181 (1998). by focal cerebral ischemia in the rat. Rofecoxib exerts no effect on platelet plug 82. Leor J, Reicher-Reiss H, Goldbourt U et al. Neurosci. Lett. 302(2–3), 121–124 (2001). formation in healthy volunteers. Clin. Aspirin and mortality in patients treated with 93. Di Napoli M, Papa F. NCX-4016 NicOx. Exp. Rheumatol. 21(2), 229–231 (2003). angiotensin-converting enzyme inhibitors: a Curr. Opin. Investig. Drugs 4(9), 1126–1139 105. Bombardier C, Laine L, Reicin A, Shapiro cohort study of 11,575 patients with (2003). D, Burgos-Vargas R, Davis B et al. coronary artery disease. J. Am. Coll. Cardiol. 94. Rahme E, Pilote L, LeLorier J. Association Comparison of upper gastrointestinal 33(7), 1920–1925 (1999). between naproxen use and protection toxicity of rofecoxib and naproxen in 83. Hall D, Zeitler H, Rudolph W. against acute myocardial infarction. Ann. patients with rheumatoid arthritis. Counteraction of the vasodilator effects of Intern. Med. 162(10), 1111–1115 (2002). VIGOR Study Group. N. Engl. J. Med. enalapril by aspirin in severe heart failure. 95. Watson DJ, Rhodes T, Cai B, Guess HA. 343(21), 1520–1528 (2000). J. Am. Coll. Cardiol. 20(7), 1549–1555 Lower risk of thromboembolic 106. Ott E, Nussmeier NA, Duke PC et al. and (1992). cardiovascular events with naproxen the Multicenter Study of Perioperative 84. Spaulding C, Charbonnier B, Cohen-Solal among patients with rheumatoid arthritis. Ischaemia (McSPI) Research Group; A et al. Acute hemodynamic interaction of Ann. Intern. Med. 162(10), 1105–1110 Ischaemia Research and Education aspirin and ticlopidine with enalapril: results (2002). Foundation (IREF) Investigators. Efficacy of a double-blind, randomized comparative 96. Garcia Rodriguez LA, Varas C, Patrono C. and safety of the cyclooxygenase-2 trial. Circulation 98(8), 757–765 (1998). Differential effects of aspirin and non- inhibitors parecoxib and valdecoxib in 85. Lapane KL, Hume AL, Barbour MM, aspirin nonsteroidal anti-inflammatory patients undergoing coronary artery Lipsitz LA. Does aspirin attenuate the effect drugs in the primary prevention of bypass surgery. J. Thorac. Cardiovasc. Surg. of angiotensin-converting enzyme inhibitors myocardial infarction in postmenopausal 125(6), 1481–1492 (2003). on health outcomes of very old patients with women. Epidemiology 11(4), 382–387 107. Joshi GP, Viscusi ER, Gan TJ et al. Effective heart failure? J. Am. Geriatr. Soc. 50(7), (2000). treatment of laparoscopic cholecystectomy 1198–1204 (2002). 97. Ray WA, Stein CM, Hall K, Daugherty pain with intravenous followed by oral 86. Guazzi MD, Campodonico J, Celeste F JR, Griffin MR. Nonsteroidal anti- COX-2 specific inhibitor. Anesth. Analg. et al. Antihypertensive efficacy of inflammatory drugs and risk of serious 98(2), 336–334 (2004). angiotensin converting enzyme inhibition coronary heart disease: an observational 108. Bresalier RS, Sandler RS, Quan H et al. and aspirin counteraction. Clin. Pharmacol. cohort study. Lancet 359(9301), 118–123 Cardiovascular events associated with Ther. 63(1), 79–86 (1998). (2002). rofecoxib in a colorectal adenoma 87. Boger RH, Bode-Boger SM, Kramme P, 98. Solomon DH, Glynn RJ, Avorn J. Non- chemoprevention trial. N. Engl. J. Med. Tsikas D, Gutzki FM, Frolich JC. Effect of steroidal anti-inflammatory drugs and risk 352(11), 1092–1102 (2005). captopril on prostacyclin and nitric oxide of serious coronary heart disease. Lancet 109. Fitzgerald GA. Coxibs and cardiovascular formation in healthy human subjects: 360(9326), 90 (2002). disease. N. Engl. J. Med. 351(17), interaction with low dose acetylsalicylic 99. Antman EM, Anbe DT, Armstrong PW 1709–1711 (2004). acid. Br. J. Clin. Pharmacol. 42(6), 721–727 et al. ACC/AHA guidelines for the 110. Topol EJ, Falk GW. A coxib a day won’t (1996). management of patients with ST-elevation keep the doctor away. Lancet 364(9435), 88. Brunner-La Rocca HP. Interaction of myocardial infarction; a report of the 639–640 (2004). angiotensin-converting enzyme inhibition American College of 111. Konstam MA, Weir MR, Reicin A, Shapiro and aspirin in congestive heart failure: long Cardiology/American Heart Association D, Sperling RS, Barr E, Gertz BJ. controversy finally resolved? Chest 124(4), Task Force on Practice Guidelines Cardiovascular thrombotic events in 1192–1194 (2003). (Committee to Revise the 1999 controlled, clinical trials of rofecoxib. 89. Wallace JL, Ignarro LJ, Fiorucci S. Potential Guidelines for the Management of Circulation 104(19), 2280–2288 (2001). cardioprotective actions of no-releasing patients with acute myocardial infarction). 112. Weir MR, Sperling RS, Reicin A, Gertz BJ. aspirin. Nature Rev. Drug Discov. 1(5), J. Am. Coll. Cardiol. 44(3), E1–E211 Selective COX-2 inhibition and 375–382 (2002). (2004). cardiovascular effects: a review of the 90. Napoli C, Ackah E, De Nigris F et al. 100. Catella-Lawson F, Reilly MP, Kapoor SC rofecoxib development program. Am. Chronic treatment with nitric oxide- et al. Cyclooxygenase inhibitors and the Heart J. 146(4), 591–604 (2003). releasing aspirin reduces plasma low-density antiplatelet effects of aspirin. N. Engl. J. 113. Reicin AS, Shapiro D, Sperling RS, Barr E, lipoprotein oxidation and oxidative stress, Med. 345(25), 1809–1817 (2001). Yu Q. Comparison of cardiovascular arterial oxidation-specific epitopes, and 101. MacDonald TM, Wei L. Effect of thrombotic events in patients with atherogenesis in hypercholesterolemic mice. ibuprofen on cardioprotective effect of osteoarthritis treated with rofecoxib versus Proc. Natl Acad. Sci. USA 99(19), aspirin. Lancet 361(9357), 573–574 nonselective nonsteroidal anti-inflammatory 12467–2470 (2002). (2003). drugs (ibuprofen, diclofenac, and 91. Napoli C, Aldini G, Wallace JL et al. 102. Cheema AA. Should people on aspirin ). Am. J. Cardiol. 89(2), Efficacy and age-related effects of nitric avoid Ibuprofen? A review of the 204–209 (2002). oxide-releasing aspirin on experimental literature. Cardiol. Rev. 12(3), 174–176 114. Silverstein FE, Faich G, Goldstein JL et al. restenosis. Proc. Natl Acad. Sci. USA 99(3), (2004). Gastrointestinal toxicity with celecoxib vs. 1689–1694 (2002). nonsteroidal anti-inflammatory drugs for

494 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

osteoarthritis and rheumatoid arthritis: the thrombosis in a baboon model by Bay myocardial infarction. Circulation 89(2), CLASS study: a randomized controlled trial. U3405, a thromboxane A2-receptor 588–595 (1994). Celecoxib Long-term Arthritis Safety Study. antagonist. Thromb. Haemost. 70(4), 134. Langleben D, Christman BW, Barst RJ et al. J. Am. Med. Assoc. 284(10), 1247–1255 672–675 (1993). Effects of the thromboxane synthetase (2000). 126. Rote WE, Mu DX, Lucchesi BR. inhibitor and receptor antagonist terbogrel 115. Farkouh ME, Kirshner H, Harrington RA Thromboxane antagonism in experimental in patients with primary pulmonary et al. and the TARGET Study Group. canine carotid artery thrombosis. Stroke hypertension. Am. Heart J. 143(5), E4 Comparison of with naproxen 24(6), 820–827 (1993). (2002). and ibuprofen in the Therapeutic Arthritis 127. White BP, Sullivan AT, Lumley P. 135. Rich S, Hart K, Kieras K, Brundage BH. Research and Gastrointestinal Event Trial Prevention of intra-coronary thrombosis in Thromboxane synthetase inhibition in (TARGET), cardiovascular outcomes: the anesthetized dog: the importance of primary pulmonary hypertension. Chest randomized controlled trial. Lancet thromboxane A2 and thrombin. Thromb. 91(3), 356–360 (1987). 364(9435), 675–684 (2004). Haemost. 71(3), 366–374 (1994). 136. Guth BD, Narjes H, Schubert HD, Tanswell 116. Solomon DH, Schneeweiss S, Glynn RJ 128. Shirakura S, Higo K, Takeda M, Karasawa P, Riedel A, Nehmiz G. Pharmacokinetics et al. Relationship between selective A. Antithrombotic effects of KW-3635, a and pharmacodynamics of terbogrel, a cyclooxygenase-2 inhibitors and acute thromboxane A2-receptor antagonist, in combined thromboxane A2 receptor and myocardial infarction in older adults. guinea-pigs. Jpn J. Pharmacol. 65(2), 93–98 synthase inhibitor, in healthy subjects. Br. J. Circulation 109(17), 2068–2073 (2004). (1994). Clin. Pharmacol. 58(1), 40–51 (2004). 117. Solomon SD, McMurray JJ, Pfeffer MA 129. Imamura T, Kiguchi S, Kobayashi K, 137. Nickolson RC, Town MH, Vorbruggen H. et al. Cardiovascular risk associated with Ichikawa K, Yamazaki Y, Kojima M. Effect Prostacyclin-analogs. Med. Res. Rev. 5(1), celecoxib in a clinical trial for colorectal of , a selective thromboxane A2- 1–53 (1985). adenoma prevention. N. Engl. J. Med. synthetase inhibitor, on cerebral infarction 138. Galie N, Manes A, Branzi A. The new clinical 352(11), 1071–1080 (2005). in rats. Comparative study with trials on pharmacological treatment in 118. Nussmeier NA, Whelton AA, Brown MT norphenazone, a free-radical scavenger. pulmonary arterial hypertension. Eur. Respir. et al. Complications of the COX-2 Arzneimittelforschung 53(10), 688–694 J. 20(4), 1037–1049 (2002). inhibitors parecoxib and valdecoxib after (2003). 139. McLaughlin VV, Gaine SP, Barst RJ et al. and cardiac surgery. N. Engl. J. Med. 352(11), 130. Golino P, Rosolowsky M, Yao SK et al. the Study Group. Efficacy and 1081–1091 (2005). Endogenous endoperoxides safety of treprostinil: an epoprostenol analog 119. Drazen JM. COX-2 Inhibitors – a lesson in and prostacyclin modulate the thrombolytic for primary pulmonary hypertension. unexpected problems. N. Engl. J. Med. activity of tissue . J. Cardiovasc. Pharmacol. 41(2), 293–299 352(11), 1131–1132 (2005). Effects of simultaneous inhibition of (2003). 120. FitzGerald GA, Smith B, Pedersen AK, thromboxane A2 synthase and blockade of 140. Badesch DB, McLaughlin VV, Delcroix M Brash AR. Increased prostacyclin thromboxane A2/ et al. therapy for pulmonary biosynthesis in patients with severe receptors in a canine model of coronary arterial hypertension. J. Am. Coll. Cardiol. atherosclerosis and platelet activation. thrombosis. J. Clin. Invest. 86(4), 43(Suppl. 12), S56–S61 (2004). N. Engl. J. Med. 310(17), 1065–1068 1095–1102 (1990). 141. Vane JR, Botting RM. Pharmacodynamic (1984). 131. Cocozza M, Picano T, Oliviero U, Russo N, profile of prostacyclin. Am. J. Cardiol. 75(3), 121. Carlsson I, Benthin G, Petersson AS, Coto V, Milani M. Effects of picotamide, an A3–A10 (1995). Wennmalm A. Differential inhibition of antithromboxane agent, on carotid 142. Friedman R, Mears JG, Barst RJ. Continuous thromboxane A2 and prostacyclin synthesis atherosclerotic evolution. A two-year, infusion of prostacyclin normalizes plasma by low dose acetylsalicylic acid in double-blind, placebo-controlled study in markers of endothelial cell injury and platelet atherosclerotic patients. Thromb. Res. 57, diabetic patients. Stroke 26(4), 97–601 aggregation in primary pulmonary 437–444 (1990). (1995). hypertension. Circulation 96(9), 2782–2784 122. Schrör K. Antiplatelet drugs. Drugs 50(1), 132. Serruys PW, Rutsch W, Heyndrickx GR (1997). 7–28 (1995). et al. Prevention of restenosis after 143. Willis AL, Smith DL, Vigo C, Kluge AF. 123. Mickelson JK, Simpson PJ, Gallas MT, percutaneous transluminal coronary Effects of prostacyclin and orally-active stable Lucchesi BR. Thromboxane synthetase angioplasty with thromboxane A2-receptor mimetic agent RS-93427–007 on basic inhibition with CGS 13080 improves blockade. A randomized, double-blind, mechanisms of atherogenesis. Lancet coronary blood flow after streptokinase- placebo-controlled trial. Coronary Artery 2(8508), 682–683 (1986). induced thrombolysis. Am. Heart J. 113(6), Restenosis Prevention On Repeated 144. Nichols WW, Nicolini FA, Khan S, Saldeen 1345–1352 (1987). Thromboxane–antagonism study TG, Mehta JL. Failure of prostacyclin analog 124. Takiguchi Y, Wada K, Nakashima M. (CARPORT). Circulation 84(4), iloprost to sustain coronary blood flow after Comparison of the inhibitory effects of the 1568–1580 (1991). recombinant tissue-type plasminogen- TXA2 receptor antagonist, vapiprost, and 133. Ridogrel Versus Aspirin Patency Trial induced thrombolysis in dogs. Am. Heart J. other antiplatelet drugs on arterial (RAPT). Randomized trial of ridogrel, a 126(2), 285–292 (1993).

thrombosis in rats: possible role of TXA2. combined thromboxane A2 synthase 145. Mueller HS, Rao PS, Greenberg MA et al. Thromb. Haemost. 688(4), 460–463 (1992). inhibitor and thromboxane Systemic and transcardiac platelet activity in 125. Kotze HF, Lamprecht S, Badenhorst PN, A2/prostaglandin endoperoxide receptor acute myocardial infarction in man: resistance van Wyk V, Roodt JP, Alexander K. In vivo antagonist, versus aspirin as adjunct to to prostacyclin. Circulation 72(6), inhibition of acute platelet-dependent thrombolysis in patients with acute 1336–1345 (1985). www.futuremedicine.com 495 REVIEW – Reiter & Jilma

146. Jaschonek K, Karsch KR, Weisenberger H, therapy. Neurol. Res. 24(4), 381–388 169. Bache RJ, Duncker DJ. Coronary steal. Tidow S, Faul C, Renn W. Platelet (2002). ACC Curr. J. Rev. 9–12 (1994). prostacyclin binding in coronary artery 158. Vasodilating agents. Dipyridamole. In: 170. Becker LC. Conditions for vasodilator- disease. J. Am. Coll. Cardiol. 8(2), 259–266 AHFS Drug Information. McEvoy GK (Ed.). induced coronary steal in experimental (1986). American Society of Health-System myocardial ischemia. Circulation 57(6), 147. Gershlick AH, Spriggins D, Davies SW et al. Pharmicists MD, USA. 1654–1656 (1999). 1103–1110 (1978). Failure of epoprostenol (prostacyclin, PGI2) 159. Diener HC, Cunha L, Forbes C, Sivenius J, 171. Gould KL. Coronary collateral function to inhibit platelet aggregation and to prevent Smets P, Lowenthal A. European Stroke assessed by PET. In: Coronary Artery restenosis after coronary angioplasty: results Prevention Study. 2. Dipyridamole and Stenosis: A Textbook of Coronary Pathology, of a randomized placebo-controlled trial. Br. acetylsalicylic acid in the secondary Quantitative Coronary Angiography, Heart J. 71(1), 7–15 (1994). prevention of stroke. J. Neurol. Sci. Perfusion Imaging and Reversal of Coronary 148. Bath PM, Bath FJ. Prostacyclin and 143(1–2), 1–13 (1996). Artery Diseases. Gould KL (Ed.). Elsevier analogues for acute ischemic stroke. 160. De Schryver EL, Algra A, van Gijn J. Science, NY, USA 169–178 (1991). Cochrane Database Syst. Rev. 2, CD000177 Cochrane review: dipyridamole for 172. Nishimura S, Kimball KT, Mahmarian JJ, (2000). preventing major vascular events in Verani MS. Angiographic and 149. Mohler ER 3rd, Hiatt WR, Olin JW, Wade patients with vascular disease. Stroke hemodynamic determinants of myocardial M, Jeffs R, Hirsch AT. Treatment of 34(8), 2072–2080 (2003). ischemia during adenosine thallium-201 intermittent claudication with beraprost 161. PRoFESS study presented. Change in scintigraphy in coronary artery disease. sodium, an orally-active prostaglandin I2 secondary prevention. MMW Fortschr. Circulation 87(4), 1211–1219 (1993). analogue: a double-blinded, randomized, Med. 145(Suppl. 2), 96–97 (2003). 173. Rosseel M, Dendale P, De Sadeleer C, controlled trial. J. Am. Coll. Cardiol. 41(10), 162. Moonis M, Fisher M. Antiplatelet Schoors D, Block P, Franken PR. 1679–1686 (2003). treatment for secondary prevention of Dipyridamole-induced angina pectoris 150. Guh JH, Ko FN, Jong TT, Teng CM. acute ischemic stroke and transient during sestamibi stress test in patients Antiplatelet effect of gingerol isolated from ischemic attacks: mechanisms, choices and with significant coronary artery disease: Zingiber officinale. J. Pharm. Pharmacol. possible emerging patterns of use. Expert clinical, angiographic, and nuclear 47(4), 329–332 (1995). Rev. Cardiovasc. Ther. 1(4), 611–615 determinants. Angiology 48(4), 301–307 151. Koo KL, Ammit AJ, Tran VH, Duke CC, (2003). (1997). Roufogalis BD. Gingerols and related 163. Schellinger PD, Juttler E, Meyding- 174. Hansen CL, Williams E. Severe analogs inhibit arachidonic acid-induced Lamade UK, Schwark C. The value of transmural myocardial ischemia after human platelet serotonin release and platelet inhibitors in the secondary dipyridamole administration implicating aggregation. Thromb. Res. 103(5), 387–397 prophylaxis of stroke – a review. Fortschr. coronary steal. Clin. Cardiol. 21(4), (2001). Neurol. Psychiatr. 72(5), 270–281 (2004). 293–296 (1998). 152. Hamasaki Y, Miyazaki S, Tai HH. 164. Warlow C. Aspirin should be first-line 175. Keltz TN, Innerfield M, Gitler B, Cooper Regulatory mechanism of arachidonic acid antiplatelet therapy in the secondary JA. Dipyridamole-induced myocardial metabolism in rat basophilic leukemia prevention of stroke. Stroke 33(8), ischemia. J. Am. Med. Assoc. 257(11), (RBL)-2H3 cells. Nippon Ketsueki Gakkai 2137–2138 (2002). 1515–1516 (1987). Zasshi 50(4), 813–818 (1987). 165. Diener HC. Aspirin therapy should be 176. Kimura Y, Tani T, Kanbe T, Watanabe K. 153. Nurtjahja-Tjendraputra E, Ammit AJ, first-line treatment in secondary Effect of cilostazol on platelet aggregation Roufogalis BD, Tran VH, Duke CC. prevention of stroke-against. Stroke 33(8), and experimental thrombosis. Effective antiplatelet and COX-1 enzyme 2138–2139 (2002). Arzneimittelforschung 35(7A), 1144–1149 inhibitors from pungent constituents of 166. Leppo J, Boucher CA, Okada RD, Newell (1985). ginger. Thromb. Res. 111(4–5), 259–265 JB, Strauss HW, Pohost GM. Serial 177. Umekawa H, Tanaka T, Kimura Y, Hidaka (2003). thallium-201 myocardial imaging after H. Purification of cyclic adenosine 154. Yamahara J, Mochizuki M, Rong HQ, dipyridamole infusion: diagnostic utility monophosphate phosphodiesterase from Matsuda H, Fujimura H. The antiulcer in detecting coronary stenoses and human platelets using new-inhibitor effect in rats of ginger constituents. relationship to regional wall motion. Sepharose chromatography. Biochem. J. Ethnopharmacol. 23(2–3), 299–304 Circulation 66(3), 649–657 (1982). Pharmacol. 33(21), 3339–3344 (1984). (1988). 167. Boucher CA, Brewster DC, Darling RC, 178. Uehara S, Hirayama A. Effects of 155. al-Yahya MA, Rafatullah S, Mossa JS, Ageel Okada RD, Strauss HW, Pohost GM. cilostazol on platelet function. AM, Parmar NS, Tariq M. Gastroprotective Determination of cardiac risk by Arzneimittelforschung 39(12), 1531–1534 activity of ginger zingiber officinale rosc. in dipyridamole-thallium imaging before (1989). albino rats. Am. J. Chin. Med. 17(1–2), peripheral vascular surgery. N. Engl. J. 179. Ishizaka N, Taguchi J, Kimura Y et al. 51–56 (1989). Med. 312(7), 389–394 (1985). Effects of a single local administration of 156. Patrono C, Coller B, Dalen JE et al. Platelet- 168. Homma S, Callahan RJ, Ameer B et al. cilostazol on neointimal formation in active drugs : the relationships among dose, Usefulness of oral dipyridamole balloon-injured rat carotid artery. effectiveness, and side effects. Chest suspension for stress thallium imaging Atherosclerosis 142(1), 41–46 (1999). 119(Suppl. 1), S39–S63 (2001). without exercise in the detection of 180. Hotta N, Koh N, Sakakibara F et al. Nerve 157. Neill KK, Luer MS. Ischemic stroke coronary artery disease. Am. J. Cardiol. function and blood flow in Otsuka Long- prevention: an update on antiplatelet 57(8), 503–508 (1986). Evans Tokushima fatty rats with sucrose

496 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

feeding: effect of an anticoagulant. Eur. J. 192. Sekiguchi M, Hoshizaki H, Adachi H, thrombolytic therapy for ischemic stroke: Pharmacol. 313(3), 201–209 (1996). Ohshima S, Taniguchi K, Kurabayashi M. the Seventh ACCP Conference on 181. Wesslau C, Eriksson JW, Smith U. Cellular Effects of antiplatelet agents on subacute Antithrombotic and Thrombolytic Therapy. cyclic AMP levels modulate insulin thrombosis and restenosis after successful Chest 126(Suppl. 3), S483–S512 (2004). sensitivity and responsiveness-evidence coronary stenting: a randomized 203. Liu Y, Shakur Y, Yoshitake M, Kambayashi against a significant role of Gi in insulin comparison of ticlopidine and cilostazol. Ji J. Cilostazol (pletal): a dual inhibitor of signal transduction. Biochem. Biophys. Res. Circ. J. 68(7), 610–614 (2004). cyclic nucleotide phosphodiesterase Type 3 Commun. 196(1), 287–293 (1993). 193. Hashiguchi M, Ohno K, Nakazawa R, and adenosine uptake. Cardiovasc. Drug Rev. 182. Nakaya Y, Minami A, Sakamoto S et al. Kishino S, Mochizuki M, Shiga T. 19(4), 369–386 (2001). Cilostazol, a phosphodiesterase inhibitor, Comparison of cilostazol and ticlopidine for 204. Kambayashi J, Liu Y, Sun B, Shakur Y, improves insulin sensitivity in the Otsuka one-month effectiveness and safety after Yoshitake M, Czerwiec F. Cilostazol as a Long-Evans Tokushima Fatty Rat, a elective coronary stenting. Cardiovasc. Drugs unique antithrombotic agent. Curr. Pharm. model of spontaneous NIDDM. Diabetes Ther. 18(3), 211–217 (2004). Des. 9(28), 2289–2302 (2003). Obes. Metab. 1(1), 37–41 (1999). 194. Choi JM, Shin HK, Kim KY, Lee JH, Hong 205. Hirose H, Kimura T, Okada M et al. 183. Park SW, Lee CW, Kim HS et al. KW. Neuroprotective effect of cilostazol Antiplatelet and antithrombotic effects of a Comparison of cilostazol versus against focal cerebral ischemia via anti- novel selective phosphodiesterase 3 ticlopidine therapy after stent apoptotic action in rats. J. Pharmacol. Exp. inhibitor, NSP-513, in mice and rats. Jpn J. implantation. Am. J. Cardiol. 84(5), Ther. 300(3), 787–793 (2002). Pharmacol. 82(3), 188–198(2000). 511–514 (1999). 195. Lee JH, Lee YK, Ishikawa M et al. Cilostazol 206. Wu CC, Wang WY, Kuo RY, Chang FR, 184. Kozuma K, Hara K, Yamasaki M et al. reduces brain lesion induced by focal Wu YC. Antiplatelet effects of KW-7, a new Effects of cilostazol on late lumen loss and cerebral ischemia in rats-an MRI study. inhibitor of cyclic nucleotide repeat revascularization after Palmaz–Schatz Brain Res. 994(1), 91–98 (2003). phosphodiesterases. Eur. J. Pharmacol. coronary stent implantation. Am. Heart J. 196. Lee JH, Kim KY, Lee YK et al. Cilostazol 483(2–3), 187–194 (2004). 141(1), 124–130 (2001). prevents focal cerebral ischemic injury by 207. Liu Y, Cone J, Le SN et al. Cilostazol and 185. Shinoda-Tagawa T, Yamasaki Y, Yoshida S enhancing casein kinase 2 phosphorylation dipyridamole synergistically inhibit human et al. A phosphodiesterase inhibitor, and suppression of phosphatase and tensin platelet aggregation. J. Cardiovasc. cilostazol, prevents the onset of silent brain homolog deleted from chromosome 10 Pharmacol. 44(2), 266–273 (2004). infarction in Japanese subjects with Type II phosphorylation in rats. J. Pharmacol. Exp. 208. Kunapuli SP. Multiple P2 receptor subtypes diabetes. Diabetologia 45(2), 188–194 Ther. 308(3), 896–903 (2004). on platelets: a new interpretation of their (2002). 197. Birk S, Edvinsson L, Olesen J, Kruuse C. function. Trends Pharmacol. Sci. 19(10), 186. Yoshitomi Y, Kojima S, Sugi T, Yano M, Analysis of the effects of phosphodiesterase 391–394 (1998). Matsumoto Y, Kuramochi M. Antiplatelet Type 3 and 4 inhibitors in cerebral arteries. 209. Savage B, Cattaneo M, Ruggeri ZM. treatment with cilostazol after stent Eur. J. Pharmacol. 489(1–2), 93–100 Mechanisms of platelet aggregation. Curr. implantation. Heart 80(4), 393–396 (1998). (2004). Opin. Hematol. 8(5), 270–276 (2001). 187. Take S, Matsutani M, Ueda H et al. Effect 198. Chambless LE, Heiss G, Folsom AR et al. 210. Mahaut-Smith MP, Tolhurst G, Evans RJ. of cilostazol in preventing restenosis after Association of coronary heart disease Emerging roles for P2X1 receptors in percutaneous transluminal coronary incidence with carotid arterial wall thickness platelet activation. Platelets 15(3), 131–144 angioplasty. Am. J. Cardiol. 79(8), and major risk factors: the Atherosclerosis (2004). 1097–1099 (1997). Risk in Communities (ARIC) Study, • Overview of the role of P2X1 receptors in 188. Sekiya M, Funada J, Watanabe K, Miyagawa 1987–1993. Am. J. Epidemiol. 146(6), platelet activation. M, Akutsu H. Effects of probucol and 483–494 (1997). 211. Hechler B, Eckly A, Ohlmann P, Cazenave cilostazol alone and in combination on 199. Hodis HN, Mack WJ, LaBree L et al. The JP, Gachet C. The P2Y1 receptor, necessary frequency of poststenting restenosis. Am. J. role of carotid arterial intima-media but not sufficient to support full ADP- Cardiol. 82(2), 144–147 (1998). thickness in predicting clinical coronary induced platelet aggregation, is not the 189. Tsuchikane E, Fukuhara A, Kobayashi T events. Ann. Intern. Med. 128(4), 262–269 target of the drug clopidogrel. Br. J. et al. Impact of cilostazol on restenosis after (1998). Haematol. 103(3), 858–866 (1998). percutaneous coronary balloon angioplasty. 200. O’Leary DH, Polak JF, Kronmal RA, 212. Geiger J, Brich J, Honig-Liedl P et al. Circulation 100(1), 21–26 (1999). Manolio TA, Burke GL, Wolfson SK Jr. Specific impairment of human platelet 190. Tanabe Y, Ito E, Nakagawa I, Suzuki K. Carotid-artery intima and media thickness P2Y(AC) ADP receptor-mediated signaling Effect of cilostazol on restenosis after as a risk factor for myocardial infarction and by the antiplatelet drug clopidogrel. coronary angioplasty and stenting in stroke in older adults. Cardiovascular Health Arterioscler. Thromb. Vasc. Biol. 19(8), comparison to conventional coronary artery Study Collaborative Research Group. 2007–2011 (1999). stenting with ticlopidine. Int. J. Cardiol. N. Engl. J. Med. 340(1), 14–22 (1999). 213. Quinn MJ, Fitzgerald DJ. Ticlopidine and 78(3), 285–291 (2001). 201. Gotoh FG, Tohgi H, Hirai S et al. Cilostazol clopidogrel. Circulation 100(15), 191. Kamishirado H, Inoue T, Mizoguchi K et al. stroke prevention study: a placebo- 1667–1672 (1999). Randomized comparison of cilostazol versus controlled double-blind trial for secondary 214. CAPRIE Steering Committee. A ticlopidine hydrochloride for antiplatelet prevention of cerebral infarction. J. Stroke randomized, blinded, trial of clopidogrel therapy after coronary stent implantation Cerebrovasc. Dis. 9(4), 147–157 (2000). versus aspirin in patients at risk of ischemic for prevention of late restenosis. Am. 202. Albers GW, Amarenco P, Easton JD, Sacco events (CAPRIE). Lancet 348(9038), Heart J. 144(2), 303–308 (2002). RL, Teal P. Antithrombotic and 1329–1339 (1996). www.futuremedicine.com 497 REVIEW – Reiter & Jilma

•• Landmark study which demonstrated the clopidogrel in addition to aspirin in 232. Ringleb PA, Bhatt DL, Hirsch AT, Topol EJ, superiority of clopidogrel over aspirin. patients with acute coronary syndromes Hacke W and the Clopidogrel Versus 215. Solet DJ, Zacharski LR, Plehn JF. The role without ST-segment elevation. N. Engl. J. Aspirin in Patients at Risk of Ischemic of adenosine 5´-diphosphate receptor Med. 345(7), 494–502 (2001). Events Investigators Benefit of clopidogrel blockade in patients with cardiovascular 223. Bhatt DL, Bertrand ME, Berger PB et al. over aspirin is amplified in patients with a disease. Am. J. Med. 111(1), 45–53 (2001). Meta-analysis of randomized and registry history of ischemic events. Stroke 35(2), 216. Hass WK, Easton JD, Adams HP Jr et al. A comparisons of ticlopidine with clopidogrel 528–532 (2004). randomized trial comparing ticlopidine after stenting. J. Am. Coll. Cardiol. 39(1), 233. Mehta P. Aspirin in the prophylaxis of hydrochloride with aspirin for the 9–14 (2002). coronary artery disease. Curr. Opin. Cardiol. prevention of stroke in high-risk patients. 224. Steinhubl SR, Berger PB, Mann JT 3rd 17(5), 552–558 (2002). Ticlopidine Aspirin Stroke Study Group. et al. and the CREDO Investigators. 234. Bhatt DL, Topol EJ and the Clopidogrel for N. Engl. J. Med. 321(8), 501–507 (1989). Clopidogrel for the reduction of events High Atherothrombotic Risk and Ischemic 217. Noble S, Goa KL. Ticlopidine. A review of during observation. Early and sustained Stabilization, Management, and Avoidance its pharmacology, clinical efficacy and dual oral antiplatelet therapy following Executive Committee. Clopidogrel added to tolerability in the prevention of cerebral percutaneous coronary intervention: a aspirin versus aspirin alone in secondary ischemia and stroke. Drugs Aging 8(3), randomized controlled trial. J. Am. Med. prevention and high-risk primary 214–232 (1996). Assoc. 288(19), 2411–2420 (2002). prevention: rationale and design of the 218. Symeonidis A, Kouraklis-Symeonidis A, 225. Diener HC, Bogousslavsky J, Brass LM Clopidogrel for High Atherothrombotic Seimeni U et al. Ticlopidine-induced et al. and the MATCH investigators. Risk and Ischemic Stabilization, aplastic anemia: two new case reports, Aspirin and clopidogrel compared with Management, and Avoidance review, and meta-analysis of 55 additional clopidogrel alone after recent ischemic (CHARISMA) trial. Am. Heart J. 148(2), cases. Am. J. Hematol. 71(1), 24–32 stroke or transient ischemic attack in high- 263–268 (2004). (2002). risk patients (MATCH): randomized, 235. Roderick PJ, Wilkes HC, Meade TW. The 219. Bertrand ME, Rupprecht HJ, Urban P, double-blind, placebo-controlled trial. gastrointestinal toxicity of aspirin: an Gershlick AH, Investigators FT. Double- Lancet 364(9431), 331–337 (2004). overview of randomized controlled trials. blind study of the safety of clopidogrel with •• First study demonstrating that aspirin plus Br. J. Clin. Pharmacol. 35(3), 219–226 and without a loading dose in combination clopidogrel is not superior to clopidogrel (1993). with aspirin compared with ticlopidine in alone in reducing major vascular events in 236. Lai KC, Lam SK, Chu KM et al. combination with aspirin after coronary high-risk patients with recent transient Lansoprazole for the prevention of stenting: the CLopidogrel ASpirin Stent ischemic attack or ischemic stroke. recurrences of ulcer complications from International Co-operative Study 226. Hankey GJ, Warlow CP. Treatment and long-term low-dose aspirin use. N. Engl. J. (CLASSICS). Circulation 102(6), 624–629 secondary prevention of stroke: evidence, Med. 346(26), 2033–2038 (2002). (2000). costs, and effects on individuals and 237. Chan FK, Chung SC, Suen BY et al. 220. Braunwald E, Antman EM, Beasley JW populations. Lancet 354(9188), 1457–1463 Preventing recurrent upper et al. and the American College of (1999). gastrointestinal bleeding in patients with Cardiology/American Heart Association 227. Sacco RL, Elkind MS. Update on Helicobacter pylori who are Task Force on Practice Guidelines antiplatelet therapy for stroke prevention. taking low-dose aspirin or naproxen. (Committee on the Management of Ann. Intern. Med. 160(11), 1579–1582 N. Engl. J. Med. 344(13), 967–973 Patients With Unstable Angina). (2000). (2001). ACC/AHA guideline update for the 228. Gorelick PB, Born GV, D’Agostino RB, 238. Laine L. Approaches to nonsteroidal anti- management of patients with unstable Hanley DF Jr, Moye L, Pepine CJ. inflammatory drug use in the high-risk angina and non-ST-segment elevation Therapeutic benefit. Aspirin revisited in patient. Gastroenterology 120(3), 594–606 myocardial infarction-2002: summary light of the introduction of clopidogrel. (2001). article: a report of the American College of Stroke 30(8), 1716–1721 (1999). 239. Peura DA, Malfertheiner P. Chairmen’s Cardiology/American Heart Association 229. Massie BM, Krol WF, Ammon SE et al. The summary: dichotomies and directions in Task Force on Practice Guidelines Warfarin and Antiplatelet Therapy in Heart acid-related disorders. Aliment. (Committee on the Management of Failure trial (WATCH): rationale, design, Pharmacol. Ther. 19(Suppl. 1), 77–80 Patients With Unstable Angina). and baseline patient characteristics. J. Card. (2004). Circulation 106(14), 1893–1900 (2002). Fail. 10(2), 101–112 (2004). 240. Fork FT, Lafolie P, Toth E, Lindgarde F. 221. Pollack CV Jr, Roe MT, Peterson ED. 2002 230. Bhatt DL, Chew DP, Hirsch AT, Ringleb Gastroduodenal tolerance of 75 mg update to the ACC/AHA guidelines for the PA, Hacke W, Topol EJ. Superiority of clopidogrel versus 325 mg aspirin in management of patients with unstable clopidogrel versus aspirin in patients with healthy volunteers. A gastroscopic study. angina and non-ST-segment elevation prior cardiac surgery. Circulation 103(3), Scand. J. Gastroenterol. 35(5), 464–469 myocardial infarction: implications for 363–368 (2001). (2000). emergency department practice. Ann. 231. Bhatt DL, Marso SP, Hirsch AT, Ringleb 241. Chan FK, Ching JY, Hung LC et al. Emerg. Med. 41(3), 355–369 (2003). PA, Hacke W, Topol EJ. Amplified benefit Clopidogrel versus aspirin and 222. Yusuf S, Zhao F, Mehta SR, Chrolavicius S, of clopidogrel versus aspirin in patients with esomeprazole to prevent recurrent ulcer Tognoni G, Fox KK and the Clopidogrel in diabetes mellitus. Am. J. Cardiol. 90(6), bleeding. N. Engl. J. Med. 352(3), Unstable Angina to Prevent Recurrent 625–628 (2002). 238–244 (2005). Events Trial Investigators. Effects of • Shows that aspirin plus a proton-pump

498 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

inhibitor is superior to clopidogrel for the cysteine residues, Cys17 and Cys270. GPIIb/IIIa antagonists. Thromb. Haemost. prevention of recurrent bleeding. Blood 101(10), 3908–3914 (2003). 74(1), 302–308 (1995). 242. Cryer B. Reducing the risks of 252. Schellekens S, Verheugt FW. Hotline 263. Coller BS. A new murine monoclonal gastrointestinal bleeding with antiplatelet sessions of the 26th European Congress of antibody reports an activation-dependent therapies. N. Engl. J. Med. 352(3), Cardiology. Eur. Heart J. 25(23), change in the conformation and/or 287–289 (2005). 2164–2166 (2004). microenvironment of the platelet 243. Ingall AH, Dixon J, Bailey A et al. 253. Leon C, Hechler B, Freund M et al. glycoprotein IIb/IIIa complex. J. Clin. Antagonists of the platelet P2T receptor: a Defective platelet aggregation and Invest. 76(1), 101–108 (1985). novel approach to antithrombotic therapy. increased resistance to thrombosis in 264. Coller BS. GPIIb/IIIa antagonists:

J. Med. Chem. 42(2), 213–220 (1999). purinergic P2Y1 receptor null mice. pathophysiologic and therapeutic insights 244. Storey RF, Sanderson HM, White AE, J. Clin. Invest. 104(12), 1731–1737 from studies of c7E3 Fab. Thromb. May JA, Cameron KE, Heptinstall S. The (1999). Haemost. 78(1), 730–735 (1997).

central role other P2T receptor in 254. Fabre JE, Nguyen M, Latour A et al. 265. Coller BS. AntiGPIIb/IIIa drugs: current amplification of human platelet Decreased platelet aggregation, increased strategies and future directions. Thromb. activation, aggregation, secretion and bleeding time and resistance to Haemost. 86(1), 427–443 (2001). procoagulant activity. Br. J. Haematol. thromboembolism in P2Y1 -deficient •• Review of the experiences with the three 110(4), 925–934 (2000). mice. Nature Med. 5(10), 1199–1202 platelet GPIIb/IIIa receptor antagonists 245. Storey RF, Oldroyd KG, Wilcox RG. (1999). abciximab, tirofiban and epifibatide. Open multicentre study of the P2T 255. Leon C, Freund M, Ravanat C, Baurand 266. Behan MW, Storey RF. Antiplatelet receptor antagonist AR-C69931MX A, Cazenave JP, Gachet C. Key role of the therapy in cardiovascular disease. Postgrad assessing safety, tolerability and activity in P2Y(1) receptor in tissue factor-induced Med. J. 80(941), 155–164 (2004). patients with acute coronary syndromes. thrombin-dependent acute • Up-to-date review of the current status of Thromb. Haemost. 85(3), 401–407 thormboemdolism: studies in P2Y(1)- antiplatelet therapy in cardiovascular (2001). knockout mice and mice treated with a disease.

246. Storey F. The P2Y12 receptor as a P2Y(1) antagonists. Circulation 103(5), 267. The EPIC Investigation. Use of a therapeutic target in cardiovascular 718–723 (2001). monoclonal antibody directed against the disease. Platelets 12(4), 197–209 (2001). 256. Baurand A, Raboisson P, Freund M et al. platelet glycoprotein IIb/IIIa receptor in •• Overview of the role of the P2Y12 receptor Inhibition of platelet function by high-risk coronary angioplasty. N. Engl. J.

as therapeutic target in cardiovascular administration of MRS2179, a P2Y1 Med. 330(14), 956–961 (1994). disease. receptor antagonist. Eur. J. Pharmacol. 268. EPILOG Investigators. Platelet 247. Huang J, Driscoll EM, Gonzales ML, Park 412(3), 213–221 (2001). glycoprotein IIb/IIIa receptor blockade AM, Lucchesi BR. Prevention of arterial 257. Jin J, Daniel JL, Kunapuli SP. Molecular and low-dose heparin during thrombosis by intravenously administered basis for ADP-induced platelet activation. percutaneous coronary revascularization. platelet P2T receptor antagonist AR- II. The P2Y1 receptor mediates ADP- N. Engl. J. Med. 336(24), 1689–1696 C69931MX in a canine model. induced intracellular calcium mobilization (1997). J. Pharmacol. Exp. Ther. 295(2), 492–499 and shape change in platelets. J. Biol. 269. Randomized placebo-controlled trial of (2000). Chem. 273(4), 2030–2034 (1998). abciximab before and during coronary 248. Storey RF, Judge HM, Wilcox RG, 258. Eckly A, Gendrault JL, Hechler B, intervention in refractory unstable angina: Heptinstall S. Inhibition of ADP-induced Cazenave JP, Gachet C. Differential the CAPTURE Study. Lancet 349(9063), P-selectin expression and platelet- involvement of the P2Y1 and P2YT 1429–1435 (1997). leukocyte conjugate formation by receptors in the morphological changes of 270. Topol EJ, Mark DB, Lincoff AM et al.

clopidogrel and the P2Y12 receptor platelet aggregation. Thromb. Haemost. Outcomes at 1 year and economic antagonist AR-C69931MX but not 85(4), 694–701 (2001). implications of platelet glycoprotein IIb/IIIa aspirin. Thromb. Haemost. 88(3), 259. Nylander S, Mattsson C, Ramstrom S, blockade in patients undergoing coronary 488–494 (2002). Lindahl TL. Synergistic action between stenting: results from a multicentre 249. Sugidachi A, Asai F, Ogawa T, Inoue T, inhibition of P2Y12/P2Y1 and randomized trial. EPISTENT Investigators. Koike H. The in vivo pharmacological P2Y12/thrombin in ADP- and thrombin- Evaluation of Platelet IIb/IIIa Inhibitor for profile of CS-747, a novel antiplatelet induced human platelet activation. Br. J. STENTing. Lancet 354(9195), 2019–2024 agent with platelet ADP receptor Pharmacol. 142(8), 1325–1331 (2004). (1999). antagonist properties. Br. J. Pharmacol. 260. Oury C, Kuijpers MJ, Toth-Zsamboki E 271. ESPRIT Investigators. Enhanced 129(7), 1439–1446 (2000). et al. Overexpression of the platelet P2X1 Suppression of the Platelet IIb/IIIa Receptor 250. Sugidachi A, Asai F, Yoneda K et al. ion channel in transgenic mice generates a with Integrilin Therapy. Novel dosing Antiplatelet action of R-99224, an active novel prothrombotic phenotype. Blood regimen of eptifibatide in planned coronary metabolite of a novel thienopyridine-type 101(10), 3969–3976 (2003). stent implantation (ESPRIT): a G(i)-linked P2T antagonist, CS-747. Br. 261. Nurden AT, Nurden P. A review of the role randomized, placebo-controlled trial. Lancet J. Pharmacol. 132(1), 47–54 (2001). of platelet membrane glycoproteins in the 356(9247), 2037–2044 (2000). 251. Ding Z, Kim S, Dorsam RT, Jin J, platelet-vessel wall interaction. Baillieres 272. Kastrati A, Mehilli J, Schuhlen H et al. and Kunapuli SP. Inactivation of the human Clin. Haematol. 6(3), 653–690 (1993). the Intracoronary Stenting and P2Y12 receptor by thiol reagents requires 262. Coller BS, Anderson K, Weisman HF. Antithrombotic Regimen-Rapid Early interaction with both extracellular New antiplatelet agents: platelet Action for Coronary Treatment Study www.futuremedicine.com 499 REVIEW – Reiter & Jilma

Investigators. A clinical trial of abciximab in infarction. J. Am. Coll. Cardiol. 42(11), model of transient cerebral ischemia. elective percutaneous coronary intervention 1879–1885 (2003). Thromb. Res. 101(3), 119–126 (2001). after pretreatment with clopidogrel. N. Engl. 280. Montalescot G, Borentain M, Payot L, 290. Kawano K, Ikeda Y, Kondo K, Umemura J. Med. 350(3), 232–238 (2004). Collet JP, Thomas D. Early vs. late K. Superiority of platelet integrin GPIIb- 273. Brener SJ, Barr LA, Burchenal JE et al. administration of glycoprotein IIb/IIIa IIIa receptor antagonist over aspirin in Randomized, placebo-controlled trial of inhibitors in primary percutaneous preventing cyclic flow reductions in the platelet glycoprotein IIb/IIIa blockade with coronary intervention of acute ST- guinea-pig middle cerebral artery. Eur. J. primary angioplasty for acute myocardial segment elevation myocardial infarction: a Pharmacol. 374(3), 377–385 (1999). infarction. ReoPro And Primary PTCA meta-analysis. J. Am. Med. Assoc. 292(3), 291. Kawano KI, Fujishima K, Ikeda Y, Kondo Organization and Randomized Trial 362–366 (2004). K, Umemura K. ME3277, a GPIIb/IIIa (RAPPORT) Investigators. Circulation 281. Montalescot G, Andersen HR, Antoniucci antagonist reduces cerebral infarction 98(8), 734–741(1998). D et al. Recommendations on without enhancing intracranial 274. Neumann FJ, Kastrati A, Schmitt C et al. percutaneous coronary intervention for hemorrhage in photothrombotic Effect of glycoprotein IIb/IIIa receptor the reperfusion of acute ST elevation occlusion of rabbit middle cerebral artery. blockade with abciximab on clinical and myocardial infarction. Heart 90(6), e37 J. Cereb. Blood Flow Metab. 20(6), angiographic restenosis rate after the (2004). 988–997 (2000). placement of coronary stents following acute 282. Topol EJ and the GUSTO V 292. Yang Y, Li Q, Nakada MT, Yang T, Shuaib myocardial infarction. J. Am. Coll. Cardiol. Investigators. Reperfusion therapy for A. Angiographic evaluation of middle 35(4), 915–921 (2000). acute myocardial infarction with cerebral artery reperfusion caused by 275. Montalescot G, Barragan P, Wittenberg O fibrinolytic therapy or combination platelet glycoprotein IIb/IIIa receptor et al. and the ADMIRAL Investigators. reduced fibrinolytic therapy and platelet complex antagonist murine 7E3 F(ab’)2 in Abciximab before Direct angioplasty and glycoprotein IIb/IIIa inhibition: the a model of focal cerebral ischemia in rats. stenting in Myocardial Infarction Regarding GUSTO V randomized trial. Lancet J. Neurosurg. 94(4), 582–588 (2001). Acute and Long-term follow-up. Platelet 357(9272), 1905–1914 (2001). 293. Shuaib A, Yang Y, Nakada MT, Li Q, Yang glycoprotein IIb/IIIa inhibition with 283. Lincoff AM. Targeting the use of platelet T. Glycoprotein IIb/IIIa antagonist, coronary stenting for acute myocardial IIb/IIIa inhibitors: treatment of acute murine 7E3 F(ab’) 2, and tissue infarction. N. Engl. J. Med. 344(25), myocardial infarction. Rev. Cardiovasc. plasminogen activator in focal ischemia: 1895–1903 (2001). Med. 3(Suppl. 1), S13–S19 (2002). evaluation of efficacy and risk of 276. Stone GW, Grines CL, Cox DA et al. and 284. Kong DF, Califf RM, Miller DP et al. hemorrhage with combination therapy. the Controlled Abciximab and Device Clinical outcomes of therapeutic agents J. Cereb. Blood Flow Metab. 22(2), Investigation to Lower Late Angioplasty that block the platelet glycoprotein 215–222 (2002). Complications (CADILLAC) IIb/IIIa integrin in ischemic heart disease. 294. Choudhri TF, Hoh BL, Zerwes HG et al. investigators. Comparison of angioplasty Circulation 98(25), 2829–2835 (1998). Reduced microvascular thrombosis and with stenting, with or without abciximab, 285. Kam PC, Egan MK. Platelet glycoprotein improved outcome in acute murine stroke in acute myocardial infarction. N. Engl. J. IIb/IIIa antagonists: pharmacology and by inhibiting GP IIb/IIIa receptor- Med. 346(13), 957–966 (2002). clinical developments. Anesthesiology mediated platelet aggregation. J. Clin. 277. Topol EJ, Neumann FJ, Montalescot G. A 96(5), 1237–1249 (2002). Invest. 102(7), 1301–1310 (1998). preferred reperfusion strategy for acute 286. Abernethy DR, Pezzullo J, Mascelli MA, 295. Moriguchi A, Mihara K, Aoki T et al. myocardial infarction. J. Am. Coll. Frederick B, Kleiman NS, Freedman J. Restoration of middle cerebral artery Cardiol. 42(11), 1886–1889 (2003). Pharmacodynamics of abciximab during thrombosis by novel glycoprotein IIb/IIIa •• Demonstrates that abciximab treatment angioplasty: comparison to healthy antagonist FK419 in guinea-pig. Eur. J. reduces death or reinfarction in patients subjects. Clin. Pharmacol. Ther. 71(3), Pharmacol. 498(1–3), 179–188 (2004). with myocardial infarction undergoing 186–195 (2002). 296. Junghans U, Seitz RJ, Aulich A, Freund HJ, percutaneous intervention. 287. Tcheng JE. Clinical challenges of platelet Siebler M. Bleeding risk of tirofiban, a 278. Van de Werf F, Ardissino D, Betriu A et al. glycoprotein IIb/IIIa receptor inhibitor nonpeptide GPIIb/IIIa platelet receptor and the Task Force on the Management of therapy: bleeding, reversal, antagonist in progressive stroke: an open Acute Myocardial Infarction of the thrombocytopenia and retreatment. Am. pilot study. Cerebrovasc. Dis. 12(4), European Society of Cardiology. Heart J. 139(2 Pt 2), S38–S45 (2000). 308–312 (2001). Management of acute myocardial 288. Lapchak PA, Araujo DM, Song D, Zivin 297. Junghans U, Siebler M. Cerebral infarction in patients presenting with ST- JA. The nonpeptide glycoprotein IIb/IIIa microembolism is blocked by tirofiban, a segment elevation. The Task Force on the platelet receptor antagonist SM-20302 selective nonpeptide platelet glycoprotein Management of Acute Myocardial reduces tissue plasminogen activator- IIb/IIIa receptor antagonist. Circulation Infarction of the European Society of induced intracerebral hemorrhage after 107(21), 2717–2721 (2003). Cardiology. Eur. Heart J. 24(1), 28–66 thromboembolic stroke. Stroke 33(1), 298. Straub S, Junghans U, Jovanovic V, Wittsack (2003). 147–152 (2002). HJ, Seitz RJ, Siebler M. Systemic 279. Antoniucci D, Rodriguez A, Hempel A 289. Horisawa S, Kaneko M, Sakurama T. thrombolysis with recombinant tissue et al. A randomized trial comparing Protective effects of SM-20302, an orally- plasminogen activator and tirofiban in acute primary infarct artery stenting with or active GPIIb/IIIa antagonist, in an middle cerebral artery occlusion. Stroke without abciximab in acute myocardial ADP/epinephrine-induced guinea-pig 35(3), 705–709 (2004).

500 Therapy (2005) 2(3) Platelets and new antiplatelet drugs – REVIEW

299. Haerten K, Krabbe C, Raiber M. Efficacy platelet aggregation and major receptor substudy. Orbofiban in patients with and safety of treatment of acute ischemic expression in patients with coronary artery unstable coronary syndromes. thrombolysis stroke with glycoprotein IIb/IIIa receptor disease for the Roxifiban Oral Compound in myocardial infarction. Am. J. Cardiol. blocker in routine clinical practice Dtsch Kinetics Evaluation Trial-I (ROCKET-I 85(4), 491–493, A10 (2000). Med. Wochenschr. 129(12), 607–610 (2004). Platelet Substudy). Am. Heart J. 146(1), 321. Xiao Z, Theroux P, Frojmovic M. 300. Abciximab in Ischemic Stroke Investigators. 91–98 (2003). Modulation of platelet-neutrophil Abciximab in acute ischemic stroke: a 310. Baba K, Aga Y, Nakanishi T, Motoyama T, interaction with pharmacological inhibition randomized, double-blind, placebo- Ueno H. UR-3216: a manageable oral of fibrinogen binding to platelet GP IIb/IIIa controlled, dose-escalation study. Stroke GPIIb/IIIa antagonist. Cardiovasc. Drug Rev. receptor. Thromb. Haemost. 81(2), 281–285 31(3), 601–609 (2000). 19(1), 25–40 (2001). (1999). 301. Major ongoing stroke trials. Stroke 35(10), 311. Aga Y, Baba K, Tam S, Nakanishi T, Yoneda 322. Derhaschnig U, Pachinger C, Jilma B. E359–E368 (2004). K, Kita J, Ueno H. UR-3216: a new Variable inhibition of high-shear-induced 302. Cannon CP, McCabe CH, Wilcox RG et al. generation oral platelet GPIIb/IIIa platelet plug formation by eptifibatide and Oral glycoprotein IIb/IIIa inhibition with antagonist. Curr. Pharm. Des. 10(14), tirofiban under conditions of platelet orbofiban in patients with unstable coronary 1597–1601 (2004). activation and high von Willebrand release: syndromes (OPUS-TIMI 16) trial. 312. Bhatt DL, Topol EJ. Scientific and a randomized, placebo-controlled, clinical Circulation 102(2), 149–156 (2000). therapeutic advances in antiplatelet therapy. trial. Am. Heart J. 147(4), E17 (2004). 303. The SYMPHONY Investigators. Nature Rev. Drug Discov. 2(1), 15–28 323. Chen WH, Lee PY, Ng W, Tse HF, Lau CP. Comparison of sibrafiban with aspirin for (2003). Aspirin resistance is associated with a high prevention of cardiovascular events after •• Overview of scientific and therapeutic incidence of myonecrosis after non-urgent acute coronary syndromes: a randomized advances in antiplatelet therapy. percutaneous intervention despite trial. Sibrafiban versus aspirin to yield 313. Chew DP, Bhatt DL, Topol E. Oral clopidogrel pretreatment. J. Am. Coll. maximum protection from ischemic heart glycoprotein IIb/IIIa inhibitors. Why don’t Cardiol. 43(6), 1122–1126 (2004). events postacute coronary syndromes. they work? Am. J. Cardiovasc. Drugs 1(6), 324. Cattaneo M. Aspirin and clopidogrel: Lancet 355(9201), 337–345 (2000). 421–428. (2001) efficacy, safety, and the issue of drug 304. Second SYMPHONY Investigators. 314. Newby LK, Califf RM, White HD. The resistance. Arterioscler. Thromb. Vasc. Biol. Randomized trial of aspirin, sibrafiban, or failure of orally administered glycoprotein 24(11), 1980–1987 (2004). both for secondary prevention after acute IIb/IIIa inhibitors to prevent recurrent • Overview of aspirin resistance and coronary syndromes. Circulation 103(13), cardiac events. Am. J. Med. 112(8), 647–658 clopidogrel resistance. 1727–1733 (2001). (2002). 325. Schriger DL, Herbert ME. Platelet 305. O’Neill WW, Serruys P, Knudtson M et al. 315. Quinn MJ, Cox D, Foley BJ, Fitzgerald DJ. glycoprotein inhibitors in patients with Long-term treatment with a platelet Glycoprotein IIb/IIIa receptor number and medically managed acute coronary glycoprotein-receptor antagonist after occupancy during chronic administration of syndrome: does the enthusiasm exceed the percutaneous coronary revascularization. an oral antagonist. J. Pharmacol. Exp. Ther. science? Ann. Emerg. Med. 38(3), 249–255 EXCITE Trial Investigators. Evaluation of 295(2), 670–676 (2000). (2001). oral xemilofiban in controlling thrombotic 316. Cox D, Smith R, Quinn M, Theroux P, 326. Harrison P. Progress in the assessment of events. N. Engl. J. Med. 342(18), Crean P, Fitzgerald DJ. Evidence of platelet platelet function. Br. J. Haematol. 111(3), 1316–1324 (2000). activation during treatment with a 733–744 (2000). 306. Topol EJ, Easton JD, Amarenco P et al. GPIIb/IIIa antagonist in patients presenting • Overview of the importance of monitoring Design of the blockade of the glycoprotein with acute coronary syndromes. J. Am. Coll. antiplatelet therapy or assessing bleeding IIb/IIIa receptor to avoid vascular occlusion Cardiol. 36(5), 1514–1519 (2000). risk. (BRAVO) trial. Am. Heart J. 139(6), 317. Quinn MJ, Plow EF, Topol EJ. Platelet 327. Gum PA, Kottke-Marchant K, Poggio ED 927–933 (2000). glycoprotein IIb/IIIa inhibitors: recognition et al. Profile and prevalence of aspirin 307. Chew DP, Bhatt DL, Sapp S, Topol EJ. of a two-edged sword? Circulation 106(3), resistance in patients with cardiovascular Increased mortality with oral platelet 379–385 (2002). disease. Am. J. Cardiol. 88(3), 230–235 glycoprotein IIb/IIIa antagonists: a meta- 318. Serebruany VL, Gurbel PA. The relations of (2001). analysis of Phase III multicenter randomized major platelet receptor expression during 328. Sane DC, McKee SA, Malinin AI, trials. Circulation 103(2), 201–206 (2001). myocardial infarction: monitoring efficacy Serebruany VL. Frequency of aspirin •• Initial report that oral glycoprotein IIb/IIIa of GPIIb/IIIa inhibitors by measuring P- resistance in patients with congestive heart inhibitors increase the rate of mortality. selectin? Thromb. Haemost. 81(2), 314–316 failure treated with antecedent aspirin. 308. Murphy J, Wright RS, Gussak I et al. The (1999). Am. J. Cardiol. 90(8), 893–895 (2002). Use of Roxifiban (DMP754), a Novel Oral 319. Peter K, Schwarz M, Ylanne J et al. 329. Jaremo P, Lindahl TL, Fransson SG, Platelet Glycoprotein IIb/IIIa Receptor Induction of fibrinogen binding and platelet Richter A. Individual variations of platelet Inhibitor, in Patients with Stable Coronary aggregation as a potential intrinsic property inhibition after loading doses of Artery Disease. Am. J. Cardiovasc. Drugs of various glycoprotein IIb/IIIa (αIIbβa3) clopidogrel. J. Intern. Med. 252(3), 3(2), 101–112 (2003). inhibitors. Blood 92(9), 3240–3249 (1998). 233–238 (2002). 309. Serebruany VL, Malinin AI, O’connor CM, 320. Holmes MB, Sobel BE, Cannon CP, 330. Muller I, Besta F, Schulz C, Massberg S, Gurbel PA and the Roxifiban Oral Schneider DJ. Increased platelet reactivity in Schonig A, Gawaz M. Prevalence of Compound Kinetics Evaluation Trial-I patients given orbofiban after an acute clopidogrel non-responders among Platelet Substudy. Effects of roxifiban on coronary syndrome: an OPUS-TIMI 16 patients with stable angina pectoris www.futuremedicine.com 501 REVIEW – Reiter & Jilma

scheduled for elective coronary stent 336. Ziegler S, Maca T, Alt E, Speiser W, Websites placement. Thromb. Haemost. 89(5), Schneider B, Minar E. Monitoring of 401. Celecoxib. PhRMA IQ5–97–02–001 783–787 (2003). antiplatelet therapy with the PFA-100 in www.clinicalstudyresults.org/documents/com 331. Gurbel PA, Bliden KP, Hiatt BL, O’Connor peripheral angioplasty patients. Platelets pany-study_76_0.pdf (Accessed April 2005) CM. Clopidogrel for coronary stenting: 13(8), 493–497 (2002). 402. Change of the PRoFESS trial design, News response variability, drug resistance, and the 337. Frossard M, Fuchs I, Leitner JM et al. Releases-Boerhinger Ingelheim effect of pretreatment platelet reactivity. Platelet function predicts myocardial http://www.boehringer- Circulation 107(23), 2908–2913 (2003). damage in patients with acute myocardial ingelheim.ca/news_releases/2004/2004–06–0 332. Matetzky S, Shenkman B, Guetta V et al. infarction. Circulation 4.asp (Accessed April 2005) Clopidogrel resistance is associated with 110(11)1392–1397 (2004). 403. Wiviott SD. JUMBO-TIMI 26: Joint increased risk of recurrent atherothrombotic 338. Reiter RA, Mayr F, Blazicek H et al. utilization of medication to block platelets events in patients with acute myocardial Desmopressin antagonizes the in vitro optimally.The ESC Congress 2004 Munich infarction. Circulation 109(25), 3171–3175 platelet dysfunction induced by www.escardio.org/knowledge/education/slides (2004). GPIIb/IIIa inhibitors and aspirin. Blood /ESC_Congress_2004_Munich/SDWiviottFP 333. Serebruany VL, Steinhubl SR, Berger PB, 102(13), 4594–4599 (2003). 714 (Accessed April 2005) Malinin AI, Bhatt DL, Topol EJ. Variability in 339. Reiter R, Jilma-Stohlawetz P, Horvath M, platelet responsiveness to clopidogrel among Jilma B. Additive effects between platelet Affiliations 544 individuals. J. Am. Coll. Cardiol. 45(2), concentrates and desmopressin in Rosemarie A Reiter 246–251 (2005). antagonizing the platelet GPIIb/IIIa Medical University of Vienna, 334. Steinhubl SR, Talley JD, Braden GA et al. inhibitor eptifibatide. Transfusion 45(3), Department of Clinical Pharmacology, Point-of-care measured platelet inhibition 420–426 (2005). Währinger Gürtel 18–20, correlates with a reduced risk of an adverse 340. Jubelirer SJ, Koenig BA, Bates MC. Acute A-1090 Vienna, Austria cardiac event after percutaneous coronary profound thrombocytopenia following Bernd Jilma intervention: results of the GOLD (AU- C7E3 Fab (Abciximab) therapy: case Medical University of Vienna, Assessing Ultegra) multicenter study. reports, review of the literature and Department of Clinical Pharmacology, Circulation 103(21), 2572–2578 (2001). implications for therapy. Am. J. Hematol. Währinger Gürtel 18–20, 335. Jilma B. Platelet function analyzer (PFA-100): 61(3), 205–208 (1999). A-1090 Vienna, Austria A tool to quantify congenital or acquired Tel.: +43 140 400 2981 platelet dysfunction. J. Lab. Clin. Med. 138(3), Fax: +43 140 400 2998 152–163 (2001). [email protected]

502 Therapy (2005) 2(3)