Abstract
Although the exact prevalence of antiplatelet resistance in ischemic stroke is not known, estimates about the two most widely used antiplatelet agents – aspirin and clopidogrel – suggest that the resistance rate is high, irrespective of the definition used and parameters measured. Inadequate antiplatelet responsiveness correlates with an increased risk of recurrent ischemic vascular events in patients with stroke and acute coronary syndrome. It is not currently known whether tailoring antiplatelet therapy based on platelet function test results translates into a more effective strategy to prevent secondary vascular events after stroke. Large-scale clinical trials using a universally accepted definition and standardized measurement techniques for antiplatelet resistance are needed to demonstrate whether a ‘platelet-function test-guided antiplatelet treatment’ strategy translates into improved stroke care. This article gives an overview of the clinical importance of laboratory antiplatelet resistance, describes the challenges for platelet-function test-guided antiplatelet treatment and discusses practical issues about the management of patients with aspirin and/or clopidogrel resistance.
Keywords: antiplatelet, aspirin, clopidogrel, ischemic stroke, non-response, platelet function assay, point of care, resistance, stroke
The concept of antiplatelet resistance in stroke
Each year, almost 185,000 recurrent strokes occur in the USA and approximately a third to half of them develop while on antiplatelet therapy [1]. The occurrence of thrombotic events despite the use of antiplatelet drugs has led to the concept of ‘antiplatelet resistance’ [2]. Other commonly used terms for antiplatelet resistance include ‘antiplatelet treatment failure’, ‘antiplatelet nonresponsiveness’ and ‘inadequate efficacy’.
The term ‘antiplatelet resistance’, in its most strict sense, refers to insufficient inhibition of platelet aggregation by antiplatelet agents in vitro. Laboratory resistance to aspirin is defined as a failure to achieve reduction in platelet thromboxane A2 (TxA2) formation following inhibition of platelet cyclooxygenase-1 (COX-1) enzyme. Laboratory resistance to thienopyridines refers to the inability to attain reduction in ADP-mediated platelet aggregation after blockade of P2Y12 receptor signaling. Clinical antiplatelet resistance or treatment failure, on the other hand, designates the occurrence of ischemic stroke or other vascular events during antiplatelet treatment. It is important to notice that treatment failure does not necessarily indicate laboratory antiplatelet resistance. Antiplatelet agents commonly used in the prevention of vascular events do not inhibit all pathways of platelet activation and aggregation [3]. Platelets may continue to aggregate and cause a stroke despite full inhibition of one particular pathway as a result of the recruitment of compensatory pathways not blocked by the antiplatelet agent. Therefore, it is critical to test all aspects of platelet function before attributing a clinical recurrence to laboratory resistance. Another point that deserves consideration is that stroke is an etiologically heterogeneous disorder. The extent to which platelets contribute to stroke pathophysiology varies depending on the underlying cause. Mechanisms such as proximal embolism from a cardiac or venous source, inflammatory and noninflammatory vasculopathies, nonthrombotic occluding atheroma, iatrogenic causes or hemodynamic compromise, and arterial vasoconstriction or vasospasm require limited contribution of platelets to vascular occlusion, and therefore can result in clinical–laboratory dissociation (clinical recurrence in the absence of laboratory resistance).
Although insufficient platelet response to anti-platelet treatment has been recognized for a long time, antiplatelet resistance in ischemic stroke has only been the focus of increased interest in recent years. There are several reasons for this: first, acceptable alternatives to aspirin that inhibit alternative pathways of platelet activation, such as clopidogrel, ticlopidine, dipyridamole–aspirin combination and cilostazol, have become available. Second, conventional labor-intensive platelet function methods (light transmission or optical light transmission aggregometry) have been replaced by simple point-of care (POC) tests that rapidly measure platelet function in the whole blood at the bedside [4]. Third, evidence indicating that platelet activity measures provide a predictive value for clinical outcome after stroke has started to emerge [5]. Fourth, combination antiplatelet therapies in individuals with clinical recurrence while on single antiplatelet therapy have been shown to confer an increased risk of clinically significant intra- or extracranial hemorrhage [6,7]. Finally, indications for antiplatelet therapy have evolved to include conditions that are associated with substantial risk of vascular injury and thrombus formation, such as percutaneous neurovascular interventions.
Evaluation of platelet functions to guide antiplatelet therapy may translate into a reduced rate of ischemic and hemorrhagic complications, and thus improved patient outcome. To develop a platelet function test-guided antiplatelet treatment strategy, it is necessary to have a readily available, easily applicable, affordable, reliable and valid method of monitoring the biological effect of antiplatelet drugs. It is also necessary to gain a better understanding of potential sources of variance in platelet aggregation and stroke pathophysiology. The purpose of this article is to introduce the factors that play a role in the variability of platelet response in patients with ischemic stroke and to provide practical considerations for clinical and laboratory aspects of resistance to aspirin and clopidogrel.
Prevalence of laboratory antiplatelet resistance
The rate of antiplatelet resistance in patients with stroke or transient ischemic attack (TIA) is highly variable, ranging from 3 to 85% for aspirin and 28 to 44% for clopidogrel (Table 1) [8]. This high variance can be partly attributed to the lack of correlation among different measurement techniques. The prevalence of antiplatelet resistance is highly assay- and agonist-dependent [9–11]. In a study of 201 patients with stable coronary artery disease using aspirin, laboratory-based aspirin resistance was 60% with Platelet Function Analyzer-100® (PFA-100®; Dade-Behring), 10–52% with light transmittance aggregometry (LTA) using different concentrations of arachidonic acid as agonist, 23% with urinary 11-dehydrothromboxane-B2 (TxB2) assay, 18% with whole-blood aggregometry (Chronolog®), 7% with VerifyNowAspirin® assay and 4% LTA with standard arachidonic acid as agonist [10]. The rate of clopidogrel resistance is also assay-dependent. In a study of 70 patients receiving 150 mg clopidogrel after percutaneous coronary intervention, the clopidogrel resistance was 13% with LTA and ADP as agonist, 39% with vasodilator-stimulated phosphoprotein assay and 33% with the VerifyNow P2Y12®[12]. In general, tests that are directly related to the inhibition of the COX-1 or P2Y12 receptor, such as LTA with arachidonic acid as agonist, VerifyNow Aspirin® and VerifyNow P2Y12, show lower resistance rates compared with nonspecific assays (PFA-100) [13]. In addition to the type of test, multiple technical and clinical factors can also confound the results of platelet function tests. These include posture [14], quality of the blood draw [15], platelet response to endothelial injury that occurs during blood collection[16], time of day [9,17], sample transport conditions [15], platelet count [15], hemolysis [15], exercise [18], smoking [19,20], age [21], gender [22], presence of infection [23], obesity [24,25], glucose control in diabetics [25,26], hemoglobin [27], serum cholesterol [26] and triglyceride [28], as well as use of concomitant agents, such as anticoagulants [29].
Table 1.
Prevalence of antiplatelet resistance in patients with stroke.
| Study (year) | Agent (mg) | n | Prevalence of antiplatelet resistance (%) | Ref. | |||
|---|---|---|---|---|---|---|---|
| LTA | PFA100® | VerifyNow® | Other | ||||
| Grotemeyer (1991) | Aspirin (500) | 82 | 26 | – | – | – | [105] |
| Grotemeyer et al. (1993) | Aspirin (500) | 180 | 33 | – | – | – | [85] |
| Helgason et al. (1993) | Aspirin (<325)† | 113 | 20.5 | – | – | – | [106] |
| Helgason et al. (1994) | Aspirin (<325)† | 228 | 25.5 | – | – | – | [37] |
| Grundman et al. (2003) | Aspirin (100) | 53 | – | 34 | – | – | [34] |
| Grau et al. (2003) | Aspirin (100–300) | 31 | – | 16.1 | – | – | [107] |
| Grau et al. (2003) | Aspirin plus clopidogrel | 31 | – | 3.4 | – | – | [107] |
| Alberts et al. (2004) | Aspirin (75–1300) | 129 | – | 37 | – | – | [108] |
| Macchi et al. (2004) | Aspirin (160) | 37 | – | 24.3 | – | – | [109] |
| Harrison et al. (2005) | Aspirin (75–150) | 100 | 12 | 22 | 17 | 14 (LTA with ADP) | [110] |
| McCabe et al. (2005) | Aspirin (75–300) | 103 | – | 50.5 | – | – | [111] |
| Berrouschot et al. (2006) | Aspirin (300) | 291 | 7.2 | – | – | – | [112] |
| Hohlfeld et al. (2007) | Aspirin (50–1500) | 90 | 15 | – | – | – | [113] |
| Bennet et al. (2008) | Aspirin (100) | 50 | – | – | 30 | – | [114] |
| Englyst et al. (2008) | Aspirin (75) | 40 | – | – | – | 67 (TEG) | [115] |
| Seok et al. (2008) | Aspirin (100) | 88 | – | – | 12 | 25 (Urinary TxB2) | [116] |
| Cha et al. (2008) | Aspirin (100) | 107 | 6.5 | – | – | 25.2 (LTA with ADP) | [117] |
| Gengo et al. (2008) | Aspirin (81–325)† | 653 | 14 | – | – | 17 (LTA with ADP) | [86] |
| Boncoraglio et al. (2009) | Aspirin (75–325) | 129 | – | 20.1 | – | – | [118] |
| Bernstein et al. (2009)‡ | Aspirin (NS) | 60 | 8.3 | – | – | – | [119] |
| Von Lewinski et al. (2009) | Aspirin (100) | 69 | 33 | 62 | – | 84 (LTA with ADP) | [120] |
| Von Lewinski et al. (2009) | Aspirin (300) | 26 | 27 | 58 | – | 85 (LTA with ADP) | [120] |
| Von Lewinski et al. (2009) | Clopidogrel (75) | 36 | 44 | – | – | 64 (LTA with collagen) | [120] |
| Von Lewinski et al. (2009) | Aspirin plus clopidogrel | 11 | 0 | 73 | – | 9 (LTA with ADP) | [120] |
| Lee et al. (2010) | Aspirin (100) | 244 | – | – | 11.5 | – | [121] |
| Jeon et al. (2010) | Aspirin (100) | 117 | – | – | 13.7 | – | [5] |
| Fong et al. (2010) | Aspirin (81–325) | 436 | 28 | – | – | – | [122] |
| Fong et al. (2010) | Clopidogrel (75) | 299 | 28 | – | – | – | [122] |
| Fong et al. (2010) | Aspirin plus clopidogrel | 270 | 9.3 | – | – | – | [122] |
Included primarily aspirin users.
Included patients with stroke, transient ischemic attack or vascular cognitive impairment.
LTA: Light transmittance aggregometry; NS: Not specified; PFA: Platelet function analyzer; TEG: Thromboelastography; TxB2: 11-dehydrothromboxane-B2.
Another source of variability in published rates of antiplatelet resistance is the lack of a standard definition for thresholds used to determine sufficient response [30]. Antiplatelet resistance is not an ‘all or none’ phenomenon; platelet response to antiplatelet treatment is a continuous parameter [31,32]. The definition of ‘resistance’ versus’ ‘nonresistance’ or ‘responder’ versus ‘nonresponder’ is largely dependent upon arbitrary cutoffs used in platelet function tests and is, therefore, highly variable; at least seven different thresholds have been used to define aspirin response in studies using the PFA-100 system [30,33,34]. Further complicating the problem is that antiplatelet resistance is not a stable phenomenon over time [35,36]. A patient who is ‘resistant’ at a specific time point can be found ‘responsive’ at another time point, despite the same treatment [37]. Platelet aggregability can recover despite sustained inhibition of one pathway due to strengthening of alternative pathways. Chronic aspirin use causes increased platelet response to TxA2-independent stimuli, such as ADP, thrombin, epinephrine, collagen and stress increases over time [31]. Similarly, clopidogrel treatment results in the upregulation of P2Y12-independent pathways, such as thrombin, TxA2, collagen and P2Y1 receptor-mediated platelet aggregation [31]. Variation in the timing of platelet function measurement relative to the index event (stroke, acute coronary syndrome, coronary or supra-aortic interventions) is also a potential contributor to the observed variability in the prevalence of antiplatelet resistance among the studies.
Proposed mechanisms of antiplatelet resistance
Several pharmacokinetic and pharmacodynamic factors, including reduced bioavailability, genetic polymorphisms, activation of alternate platelet-stimulation pathways, accelerated platelet turnover and factors associated with antiplatelet-resistant state, contribute to the variability in platelet inhibition by antiplatelet agents (Box 1). Accurate identification of the underlying mechanism of resistance, particularly a distinction between whether diminished platelet response to antiplatelet treatment is primarily due to the lack of antiplatelet drug’s effectiveness in inhibiting its target receptor or recruitment of alternative pathways for platelet activation, is key to the management. The following section describes potential causes of resistance to aspirin or clopidogrel based on their relative contribution to the inhibition of COX-1 and P2Y12 systems.
Box 1. Possible causes of antiplatelet resistance.
Antiplatelet resistance due to inadequate inhibition of COX-1 or P2Y12
-
Reduced bioavailability:
Poor compliance
Inappropriate dosing or underdosing
Reduced absorption
Increased metabolism
Drug–drug interactions
Aspirin: NSAIDs (ibuprofen, indometacin and naproxen), proton pump inhibitors
Clopidogrel: CYP3A4 substrates (atorvastatin, simvastatin and lovastatin) and inhibitors (amlodipine)
CYP2C19 substrates and inhibitors (omeprazole and esomeprazole)
-
Genetic polymorphisms:
-
Aspirin:
Receptors: GPIa/IIa, GPIb α, GPIIIa (PlA1/A2), GPIIbIIIa, GPIb/V/IX, thromboxane and von Willebrand factor receptor
Enzymes: COX-1, COX-2, thromboxane A2 synthase and UDP-glucuronosyltransferases
Factor XIII Val34Leu polymorphism
-
Clopidogrel:
Receptors: P2Y12
Enzymes: CYP3A4, CYP1A2, CYP2C19, ABCB1 (P-glycoprotein), etc.
-
Antiplatelet resistance despite adequate inhibition of COX-1 or P2Y12
-
Activation of alternate platelet stimulation pathways:
Increase epinephrine-mediated platelet activation
Stress-induced COX-2 expression in platelets (aspirin)
Increased platelet sensitivity to ADP and collagen
Increased release of ADP
Red-cell-induced platelet activation
Provide PGH2 to platelets (COX-1 bypass) or direct synthesis of TbXA2, by endothelium and monocytes (aspirin)
Increased P2Y1-dependent platelet aggregation (clopidogrel)
-
Accelerated platelet turnover:
Stress, bleeding and surgery
Acute ischemic syndromes
Acute or chronic infection or inflammation
-
Other:
Severity, duration and control of atherosclerosis
Diabetes mellitus and other vascular risk factors
Enhanced basal platelet reactivity
Antiplatelet resistance due to inadequate inhibition of COX-1 or P2Y12
Reduced availability of antiplatelet drugs
The most common cause of inadequate antiplatelet response in laboratory assays is noncompliance or nonadherence to anti-platelet treatment [38]. Approximately 50% of patients using aspirin or clopidogrel either stop taking medication or fail to adhere to their prescribed dose at 1 year [39–41]. Detection of plasma aspirin, clopidogrel or their metabolite levels (salicylate for aspirin, active thiol or inactive carboxyl metabolite for clopidogrel) can be considered to confirm drug intake when noncompliance is a concern [39]. Alternatively, a shift towards greater platelet inhibition after a period of supervised drug intake strongly indicates that noncompliance is the mechanism responsible for poor platelet response to treatment [42].
Reduced bioavailability due to poor absorption also blunts platelet response to aspirin. Aspirin is served in different pharmaceutical formulations, including plain, enteric-coated, buffered, soluble, suppository, mouth-dispersible and micro-encapsulated forms. Enteric-coated or slow-release aspirin formulations are absorbed in the small intestine. Optimal absorption of aspirin occurs in a pH range of 2–4. The neutral pH environment of the small intestine results in delayed and reduced absorption, and thus inadequate platelet inhibition [43]. Plain preparations are stable in the acidic environment of the stomach and are rapidly absorbed [44]. Soluble buffered, dispersible and chewable preparations are comparable to plain aspirin in terms of absorption rate, onset of effect and magnitude of platelet inhibition [45].
Potential drug interactions may also be important in explaining the reduced response of platelets to antiplatelet therapy. Concomitant use of NSAIDs, particularly ibuprofen, offsets the clinical benefit of aspirin by blocking its docking site on COX-1 [46]. Proton pump inhibitors (PPIs) can reduce absorption of the active form of aspirin through suppression of gastric acid secretion and activation of gastrointestinal mucosal esterases that hydrolyze aspirin to an inactive form [30]. PPIs are substrates and inhibitors of the hepatic enzyme (CYP2C19) that converts clopidogrel to its active metabolite. Therefore, concomitant use of PPIs with clopidogrel can negate clopidogrel’s antiplatelet effect [47–50]. However, published data on the clinical relevance of this interaction are inconsistent; although smaller studies suggest that the clopidogrel–PPI interaction is associated with an increased risk of vascular events after acute coronary syndromes [51,52], more recent data from larger studies fail to provide conclusive evidence for a clinically significant interaction [53,54]. It has been demonstrated that lipophilic statins, such as atorvastatin [55,56], simvastatin [56] and lovastatin [31], impede the antiplatelet effects of clopidogrel. Lipophilic statins inhibit the metabolism of clopidogrel to its active form through competition for the CYP3A4 enzyme. Although registry studies suggest a worse outcome in patients using clopidogrel together with lipophilic statins [31], randomized trials refute these findings by revealing a fairly comparable risk reduction in patients with and without statin therapy [57–59]. Dihydropyridine calcium-channel blockers are also substrates and inhibitors of the CYP3A4 enzyme. In patients treated with percutaneous coronary intervention (PCI), concomitant dihydropyridine calcium-channel blocker therapy correlates with higher platelet reactivity [60–62]. Clinical outcome in patients using clopidogrel and dihydropyridine calcium-channel blockers appears to be worse compared with those who do not use dihydropyridine calcium-channel blockers [62]. Larger randomized trials that take into account other differences among treatment groups are needed to confirm or refute the clinical relevance of this interaction.
Genetic polymorphisms
It is estimated that up to 30% of variability in platelet activity can be explained by genetic factors [63]. Although several polymorphisms have been found to cause resistance to antiplatelet medications, the lack of validation in large studies limits their use in clinically identifying resistant individuals [63,64]. Common polymorphisms for aspirin resistance include polymorphisms in the platelet COX-1 (C50T/A842G polymorphism) and COX-2 genes, the platelet glycoprotein receptor genes (P1A1/A2 polymorphism) [64] and the UDP-glucuronosyltransferase gene (UGT1A6*2). Clopidogrel resistance has been linked to polymorphisms in genes involved in hepatic metabolism (CYP 1A2, CYP3A4 and CYP2C19*2), intestinal absorption (ABCB1 gene, P-glycoprotein gene, C3435T genotype) [65] and platelet surface receptors (P2Y1 and P2Y12) [66]. Individuals with these polymorphisms exhibit higher residual platelet aggregation and increased risk of instent thrombosis and death from myocardial infarction (MI) and stroke after acute coronary syndromes [67,68]. The US FDA has issued a warning regarding clopidogrel, alerting that it can be less effective in individuals carrying the CYP2C19*2 loss-of-function allele (poor metabolizers) [69]. More studies are needed to determine the clinical relevance of genetic polymorphisms in patients with ischemic stroke.
Antiplatelet resistance despite adequate inhibition of COX-1 or P2Y12
Mechanisms that activate platelets through COX-1- or P2Y12-independent pathways can cause increased baseline platelet reactivity and antiplatelet resistance despite full inhibition of the COX-1 or P2Y12 systems. Aspirin reduces TxA2 synthesis through the inhibition of the COX-1 enzyme. However, COX-2 enzyme is inducible under certain conditions, such as inflammation and atherosclerosis, and can continue to provide TxA2 to platelets. Infection and advanced atherosclerosis associated with the formation of macrophage-rich plaques blunt platelet response to aspirin and clopidogrel, despite adequate COX-1 and P2Y12 inhibition [2,70,71].
Another mechanism of poor platelet response to antiplatelet treatment is increased availability of circulating platelets that are not exposed to antiplatelet medication in the bloodstream. The plasma half-life of aspirin is only 20 min and the plasma half-life of the active metabolite of clopidogrel is approximately 8 h. Increased production by the bone marrow, enhanced turnover induced by stress and bleeding, as well as platelet transfusions, thereby, result in circulating platelets that are capable of aggregating in spite of treatment [31].
Diabetes mellitus is also associated with enhanced baseline platelet reactivity [25,29,72]. Several mechanisms have been proposed for the development of the antiplatelet-resistant state in diabetes mellitus. These include a higher rate of circulating immature platelets [73], diabetes-associated hyperfibrinogenemia [74], hyperlipidemia [26], systemic inflammatory stress [23,74,75], reduced conversion of clopidogrel to its active metabolite [76], generalized platelet receptor overexpression [77], reduced formation of platelet-derived nitrous oxide [77], increased platelet sensitivity to ADP [77] and diminished acetylation of the target site of COX-1 [78]. Diabetes-associated antiplatelet resistance is associated with an increased risk of cardiovascular events [72]. Although published data suggest that glycemic control results in improvement of platelet inhibition, translation of this finding to improvement in clinical outcome awaits further studies [25,26]. Other factors that are associated with increased platelet reactivity in spite of COX-1 and P2Y12 inhibition include cigarette smoking, hyperlipidemia, obesity, hypertension, female gender and catecholamine surge in response to mental stress, exercise, sepsis and congestive heart failure [31].
Laboratory tests to assess platelet function
Although several platelet function tests are available to determine ‘laboratory antiplatelet resistance’, none are currently recommended for routine clinical use because of the lack of randomized trial data demonstrating that tailoring antiplatelet therapy based on platelet function tests improves clinical outcome. Nevertheless, the field is rapidly advancing and evidence from trials using newer, less labor-intensive and easy-to-apply tests is becoming available. Table 2 provides an overview of currently available tests to evaluate platelet function. There are two major categories of tests: specific and nonspecific tests. Each test serves a different purpose. Nonspecific tests reflect the global biologic response of platelets to various activators and inhibitors so that they provide an assessment of in vivo platelet activity (e.g., bleeding time and PFA-100®). Tests specific to COX-1 or P2Y12 (e.g., LTA-arachidonic acid, serum TxB2 level and VerifyNow®), on the other hand, measure individual pathway- and drug-specific platelet inhibition.
Table 2.
Methods used for the evaluation of antiplatelet resistance.
| Test | Drugs tested | Limitations | Advantages |
|---|---|---|---|
| Bleeding time | Aspirin, thienopyridines | Poor reproducibility Operator dependent Significant patient discomfort |
Standardized clinical test |
| Light transmission optical aggregometry | Aspirin, thienopyridines, GPIIb/IIIa inhibitors | Labor intensive Time consuming Operator dependent Large sample volume Expensive Difficult to standardize Low sensitivity |
Gold standard |
| Platelet Works®, Helena Laboratories, (TX, USA) | Aspirin, thienopyridines, GPIIb/IIIa inhibitors | Expensive Limited specificity and sensitivity |
Point of care |
| Multiplate® Platelet function Analyser† (Munich, Germany) | Aspirin, thienopyridines, GPIIb/IIIa inhibitors (indirect) | Overlap between non-, low- and high- responders | Point of care |
| VerifyNow® Accumetrics (CA, USA) | Aspirin, thienopyridines, GPIIb/IIIa | Lack of clear cutoff between low and high responders | Point of care |
| Platelet Function analyzer (PFA-100®), Dade Behring | Aspirin | Dependence on hematocrit and von Willebrand factor Insensitive to clopidogrel |
Point of care |
| Impact Cone and Plate(let) Analyzer, DiaMed (Cressier, Switzerland) | Aspirin | Not widely available | Point of care |
| Thromboelastography Thromboelastogram, Haemoscope Corporation, (IL, USA; TEG® or ROTEM®) | Aspirin, thienopyridines, GPIIb/IIIa inhibitors | Not exclusively platelet dependent, mostly evaluates clot properties | Point of care |
| Flow cytometry | Aspirin, thienopyridines, GPIIb/IIIa inhibitors | Expensive, operator dependent | Requires small volume of whole blood |
| Vasodilator-stimulated phosphoprotein phosphorylation (Flow cytometric) | Thienopyridines | Operator dependent Labor intensive |
Requires small volume of whole blood |
| Serum thromboxane B2 level | Aspirin | Not platelet specific, prone to artifact | Widely available |
| Urinary 11-dehydrothromboxane B2 Immunoassay (AspirinWorks®) | Aspirin | Not platelet specific Renal function dependent |
Widely available |
The bleeding time is a simple bedside test used to assess platelet function, but it is not specific and poorly reproducible [4,31]. LTA measures changes in light transmittance that occur after platelet aggregation in response to the addition of agonists. LTA has been the gold standard test of platelet function and is still used to validate newer tests. There are several aspects of this technique that limit its use as a reliable and valid means to assess platelet function. These include operator dependency, poor reproducibility and lack of standardization of results [79]. Measurement of TxA2 pathway metabolites in the serum (TxB2) and urine (11-dehydroTxB2) can be used to assess the antiplatelet effect of aspirin [4]. However, TxA2 metabolites are also dependent on TxA2 production from COX-1-independent sources (e.g., monocytes/macrophages), and are therefore not necessarily platelet-specific. Nonplatelet sources are estimated to be responsible for up to 30% of urinary 11-dehydroTxB2 levels [80]. Flow cytometry is a conjugated monoclonal antibody-based technique that measures in vitro platelet reactivity to agonists. It is a highly specific assay for the assessment of activity of individual surface antigens (receptors) of platelets. However, it is poorly sensitive to changes in overall platelet aggregation [81].
Newer POC assays allow rapid assessment of platelet functions in whole blood at bedside. They are ideal for use in emergency departments or catheter laboratories because they do not require sample transport, pipetting, special handling or a specialized laboratory or technician. PFA-100® simulates the events occurring during a typical vessel wall damage under high shear conditions in the presence of erythrocytes. Shear stress is created by drawing anticoagulant blood through a capillary tube towards a small aperture coated with collagen/epinephrine or collagen/ADP cartridges. As blood flows through the system, a platelet plug forms and gradually occludes the aperture. The time needed to occlude the aperture is called closure time or in vitro bleeding time. The drawbacks of the PFA-100 system include limited sensitivity, dependence of platelet-independent factors, poor reliability and high variability in measurements [82]. Multiple Platelet Function Analyzer (Multiplate®, Munich, Germany) is an automated version of impedance aggregometry. Different reagents, including arachidonic acid (ASPItest®), ADP (ADPtest®), ADP plus prostaglandin E1 (ADP-HS® test), collagen (COLtest®), ristocetin and thrombin receptor activating peptide (TRAP-6) (TRAPtest®), are used to monitor the effect of aspirin, thienopyridines and GPIIb/IIIa inhibitors. The VerifyNow® instrument (Accumetrics, San Diego, CA, USA), which is also known as the Ultegra Rapid Platelet Function Analyzer (RPFA), is a fully automated test using turbidometric aggregometry methodology. Three VerifyNow assays are available: the VerifyNow IIb/IIIa Assay® is sensitive to GPIIb/IIIa antagonists, the VerifyNow Aspirin Assay® is sensitive to aspirin and the VerifyNow P2Y12 Assay® is sensitive to thienopyridines. The Impact® Cone and Plate(let) Analyzer (DiaMed) contains a microscope and performs staining and image analysis of the platelets that have adhered and aggregated on a specially designed plastic plate [83]. POC assays are still new systems. More data regarding correlation between results obtained with these systems and the gold standard LTA are needed [84].
Clinical significance of antiplatelet resistance
Although there is no unified definition for antiplatelet resistance, the published data to date suggest that platelet aggregability is associated with clinical outcome after stroke. Grotemeyer et al. studied platelet reactivity in 180 patients with anterior circulation infarcts 12 h after an oral dose of 500 mg aspirin [85]. A third of patients were diagnosed to have aspirin resistance. All patients were discharged with 500 mg aspirin three-times daily. After a 2-year follow-up, the incidence of stroke, MI or vascular death was 4.4% in aspirin responders and 40% in aspirin nonresponders. Grundmann et al. studied 53 patients using 100 mg/day aspirin for more than 5 years for secondary prevention of cardiovascular events [34]. The rate of aspirin resistance was 34% in patients with, and 0% in patients without, ischemic stroke or TIA during the previous 3 days. In a case–control study of 653 consecutive patients treated with aspirin for secondary stroke prophylaxis, aspirin resistance was diagnosed in 36% of patients with and 17% of patients without prior history of stroke or TIA while on aspirin treatment [86].
There are relatively more data on the link between antiplatelet resistance and clinical outcome in cardiac patients. A meta-analysis of 13 prospective cohort and three case–control studies, which comprised of 2781 patients with cardiovascular disease, demonstrated that the pooled odds ratio (OR) of cardiovascular outcome for laboratory antiplatelet resistance was 3.8 (95% CI: 2.3–6.1) [87]. In another meta-analysis of 20 studies (17 cohort, two case–control and one descriptive study) including a total of 2930 patients with cardiovascular disease, the cardiovascular event rate was 39% in aspirin-resistant patients and 16% in aspirin-sensitive patients (OR: 3.9; 95% CI: 3.1–4.8) [88]. A meta-analysis that included 3688 patients undergoing coronary angioplasty and stenting from 25 studies demonstrated that clopidogrel resistance detected by POC tests was associated with increased risk of cardiovascular events (OR: 8.0; 95% CI: 3.4–19.0) [89]. In a multicenter, prospective, controlled study of 162 patients undergoing coronary angioplasty and stenting, patients randomized to receive additional bolus doses of clopidogrel according to the results of platelet function tests before the procedure had a lower risk of cardiac events (10 vs 0%) during the 1-month follow-up compared with controls who received a standard bolus of clopidogrel not guided by platelet function test results [90]. Although these studies indicate that laboratory antiplatelet resistance may be a clinically important phenomenon, marked heterogeneity among the studies in aspirin dose (75–1500 mg/day), duration of follow-up, type of assay used to determine laboratory resistance, cutoff value to define resistance, distribution of withdrawals, definition of end points and the method of confirmation of compliance limits the validity of their conclusions. The lack of data from well-controlled large-scale randomized trials deters guidelines from recommending the use of platelet function tests for outcome prediction in routine clinical practice. The American College of Cardiology/American Heart Association Guidelines currently only recommend platelet function testing in high-risk patients for coronary stent thrombosis (class IIB, level of evidence C).
Management of laboratory antiplatelet resistance
It is not known whether treatment modification based on laboratory testing is safe, efficient or cost-effective in stroke. More data from large-scale studies are needed to support whether switching from one antiplatelet agent to another, increasing the dose of an antiplatelet drug or administration of a combination of antiplatelet drugs overcomes antiplatelet resistance. However, the lack of high-quality data should not cause a neglect on the side of treating physicians in addressing modifiable causes of antiplatelet resistance. Some simple measures to enhance platelet response to antiplatelet medication include improving patient compliance, selecting the right formulation, avoiding drug interactions and controlling vascular risk factors.
Subjects with antiplatelet resistance may be candidates for a more potent platelet inhibition. One option for this may be to increase the dose of the antiplatelet agent. Platelet response to aspirin seems to be dose-dependent; higher doses (e.g., 300 mg instead of 100 mg) are associated with a lower rate of aspirin resistance [91]. However, there is currently no evidence that platelet function test-guided dose escalation is associated with better outcome. On the contrary, different doses of aspirin (75–1500 mg) are known to provide similar benefit in reducing morbidity and mortality from stroke and other cardiovascular diseases [92]. Moreover, there is a clear dose–hemorrhage relationship for aspirin therapy. Trials are needed to assess the risk and benefit of aggressive aspirin dosing strategies based on platelet function test results. The same conclusion also applies to clopidogrel; although higher loading (1200 mg instead of the usual 600 mg) and maintenance dosing strategies (150 mg/day instead of the routine 75 mg/day) provide greater platelet inhibition in individuals with the CYP2C19 loss-of-function polymorphism [93], it remains to be determined whether increased dosing strategy is safe and effective in clopidogrel-resistant patients with stroke.
More potent platelet inhibition may also be achieved by transition to a different formulation or to a different class of antiplatelet agent. Buffered and enteric-coated aspirin preparations are often prescribed to minimize gastrointestinal side effects, yet, as mentioned before, these preparations suffer from lower bioavailability compared with regular or plain formulations [43]. It is not currently known whether switching from aspirin to clopidogrel on the basis of clinical aspirin failure is an effective strategy because most of the aspirin nonresponders are also less responsive to clopidogrel [94]. Patients with laboratory resistance to clopidogrel may be responsive to other thienopyridines, such as ticlopidine [95,96], prasugrel, cangrelor, ticagrelor and elinogrel [66,97].
Drug–drug interactions should also be carefully taken into consideration in order to ensure optimal platelet response. Elimination of ibuprofen or other NSAIDs, such as naproxen, tiaprofenic acid and indomethacin, may be emphasized in patients using aspirin. If discontinuation is not an option, recommendation for an NSAID that does not interfere with aspirin, such as sulindac [98] and diclofenac [99], can be considered. Alternatively, aspirin can be administered when the plasma concentration of ibuprofen is low. Avoidance of ibuprofen from 8 h before until 30 min after administration of an aspirin dose has been shown to enhance platelet response to aspirin. All PPIs (esomeprazole, lansoprazole, omeprazole, pantoprazole and rabeprazole) reduce the antiplatelet effect of thienopyridines to varying degrees owing to CYP2C19 interaction. The strongest interaction occurs with omeprazole. Pantoprazole exhibits relatively limited interaction with the CYP2C19 system, yet the clinical significance of this interaction is uncertain [48,100]; a recent retrospective cohort study of patients receiving clopidogrel after MI has demonstrated that all PPIs, including pantoprazole, are associated with an increased risk of rehospitalization for MI and coronary stent placement [101]. Until further studies become available, the risks and benefits of concomitant treatment with PPIs and thienopyridines should be carefully considered for each individual patient [102]. Lipophilic statins (atorvastatin, lovastatin and simvastatin) reduce the anti-platelet effect of clopidogrel by inhibiting its activation via interaction with CYP3A4, yet clinical trials do not indicate an increased risk for adverse vascular events by concomitant use of statin and clopidogrel [103]. Switching from lipophilic statins to hydrophilic statins (rosuvastatin and pravastatin) is not justified at this time. Likewise, there is currently no convincing evidence from clinical trials that discontinuation of dihydropyridine calcium channel blockers reduces the rate of vascular events in stroke patients with clopidogrel resistance.
Expert commentary
Increasing evidence suggests that antiplatelet resistance is common and closely correlates with the risk of future vascular events in patients with stroke and coronary artery disease. While individualization of antiplatelet treatment by measuring their effect at the target site is certainly desirable to achieve optimal platelet inhibition, there are several unresolved issues that impede the transition of platelet function measures to routine clinical practice. Laboratory tests measuring platelet responsiveness are currently not optimal. Generally, their results poorly correlate with each other and are subject to high intra-assay variability. In addition, thresholds used to define antiplatelet resistance are inconsistent across the studies and highly assay-dependent. The dynamic nature of platelet aggregability and multiplicity of its activation mechanisms further complicates the problem in terms of when and in whom to measure platelet function. As a result, it is not currently known whether platelet function test-guided anti-platelet therapy translates into a more effective strategy to prevent secondary vascular events after stroke. Well-powered randomized controlled trials are needed to take antiplatelet therapy to the individual level. Such trials will require the availability of platelet function tests that are applicable to a clinical setting, not labor-intensive, have low intra-assay variability, define resistance based on a well-established cutoff, and provide results that correlate with the clinical outcome. Since platelet aggregation is a multidimensional process, there is probably no single test or definition for antiplatelet resistance. Therefore, such trials should consider measuring both global in vivo platelet activity using nonspecific tests and drug-specific platelet inhibition using specific tests. The knowledge of global and specific responses to antiplatelet therapy may help in the development of strategies to achieve maximum platelet inhibition, while keeping the bleeding risk to a minimum.
The vast majority of data published on antiplatelet resistance come from studies of patients with coronary artery disease. While stroke and coronary artery disease share common risk factors and, to some extent, have similar pathophysiology, caution should be exercised before indiscriminate application of data from cardiac literature to stroke. Stroke is an etiologically diverse disease. The relative contribution of platelet activation to the pathophysiology of stroke and coronary artery disease may be different. For instance, atherosclerotic plaque rupture with subsequent platelet aggregation and thrombus formation is the causative mechanism in less than half of all ischemic strokes, whereas up to 90% of acute coronary syndromes are caused by plaque rupture. The safety of treatment modification based on platelet function testing may also be different between stroke and coronary artery disease. Stroke is generally a higher risk condition for developing intracranial hemorrhage by antiplatelet treatment compared with coronary artery disease [104]. It is necessary to test the efficacy and safety of ‘platelet function test-guided antiplatelet therapy’ separately in patients with ischemic stroke before any specific recommendations for this population can be made.
Until evidence from large-scale good quality trials on the utility of platelet function test-guided antiplatelet treatment becomes available, platelet inhibition by antiplatelet treatment can be optimized by careful consideration of assuring adherence to antiplatelet medication, selecting the right dose and formulation, avoiding drug interactions and controlling baseline vascular risk factors. An individualized approach based on platelet function testing may be considered in patients with high and imminent risk of intravascular thrombus formation to prevent a catastrophic stroke. For instance, instent thrombosis in a vertebral or carotid artery could be lethal if the contralateral artery is occluded. Platelet function test results may also influence selection of stent type (drug-eluting or not) and revascularization strategy, such as endarterectomy versus stenting, in patients scheduled for neurovascular intervention.
Five-year view
In recent years, there has been an increasing interest in antiplatelet resistance in ischemic stroke. The number of publications on the prevalence of antiplatelet resistance in ischemic stroke has substantially increased during the last 5 years. It is anticipated that small observational studies exploring the prevalence of anti-platelet resistance will be replaced by larger cohort studies that focus a priori on the clinical relevance of antiplatelet resistance in ischemic stroke. It is also anticipated that large clinical trials will be launched to examine the impact of treatment modification based on platelet function test results on the risk of subsequent vascular events after stroke. Several such trials are currently underway in cardiac patients. The Aspirin Nonresponsiveness and Clopidogrel Endpoint Trial (ASCET) investigates whether clopidogrel treatment in patients with coronary artery disease who are nonresponsive to aspirin according to the PFA-100 method will reduce the risk of composite of unstable angina, MI, stroke or death [201]. The safety and efficacy of platelet function test-guided antiplatelet treatment are also being evaluated in several trials in patients undergoing percutaneous cardiac interventions (Gauging Responsiveness With A VerifyNow Assay – Impact On Thrombosis And Safety [GRAVITAS] [202]; Double Randomization of a Monitoring Adjusted Antiplatelet Treatment Versus a Common Antiplatelet Treatment for DES Implantation and Interruption Versus Continuation of Double Antiplatelet Therapy [ARCTIC] [203]; and Testing Platelet Reactivity In Patients Undergoing Elective Stent Placement on Clopidogrel to Guide Alternative Therapy With Prasugrel [TRIGGER-PCI] [204]). The results of these cardiac trials are expected to provide potentially important implications for the management of antiplatelet therapy in ischemic stroke.
There are several emerging antiplatelet drugs that have more predictable pharmacokinetic and pharmacodynamic properties and, therefore, exhibit a more potent and predictable antiplatelet effect compared with currently available agents. Prasugrel, ticagrelor and cangrelor are new-generation P2Y12 receptor antagonists. Their efficacy is relatively free of hepatic activation, resistant to esterases and less dependent on CYP2C19 oxidation. Accordingly, they exert a more potent and predictable antiplatelet effect. To date, none have been tested exclusively in patients with stroke in a well-powered trial. Therefore, their efficacy and safety in stroke remains to be studied in the future. Future research in stroke will also focus on identifying genes involved in platelet response to antiplatelet treatment. Currently ongoing genome-wide association studies of stroke may reveal additional prevalent and clinically significant genetic abnormalities in the near future.
Footnotes
Financial & competing interests disclosure
Hakan Ay has been supported by NIH grant R01-NS059710. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
No writing assistance was utilized in the production of this manuscript.
References
Papers of special note have been highlighted as:
• of interest
•• of considerable interest
- 1.Lloyd-Jones D, Adams RJ, Brown TM, et al. Heart disease and stroke statistics – 2010 update: a report from the American Heart Association. Circulation. 2010;121(7):e46–e215. doi: 10.1161/CIRCULATIONAHA.109.192667. [DOI] [PubMed] [Google Scholar]
- 2.Hankey GJ, Eikelboom JW. Aspirin resistance. Lancet. 2006;367(9510):606–617. doi: 10.1016/S0140-6736(06)68040-9. [DOI] [PubMed] [Google Scholar]
- 3.Breet NJ, van Werkum JW, Bouman HJ, et al. Comparison of platelet function tests in predicting clinical outcome in patients undergoing coronary stent implantation. JAMA. 2010;303(8):754–762. doi: 10.1001/jama.2010.181. [DOI] [PubMed] [Google Scholar]
- 4.Michelson AD. Methods for the measurement of platelet function. Am J Cardiol. 2009;103(3 Suppl):A20–A26. doi: 10.1016/j.amjcard.2008.11.019. [DOI] [PubMed] [Google Scholar]
- 5.Jeon SB, Song HS, Kim BJ, et al. Biochemical aspirin resistance and recurrent lesions in patients with acute ischemic stroke. Eur Neurol. 2010;64(1):51–57. doi: 10.1159/000315147. [DOI] [PubMed] [Google Scholar]
- 6.Cuisset T, Frere C, Quilici J, Uhry S, Alessi MC, Bonnet JL. Post-PCI fatal bleeding in aspirin and clopidogrel hyper responder: shifting from antiplatelet resistance to bleeding risk assessment? Int J Cardiol. 2010;138(2):212–213. doi: 10.1016/j.ijcard.2008.06.044. [DOI] [PubMed] [Google Scholar]
- 7.May AE, Geisler T, Gawaz M. Individualized antithrombotic therapy in high risk patients after coronary stenting. A double-edged sword between thrombosis and bleeding. Thromb Haemost. 2008;99(3):487–493. doi: 10.1160/TH07-11-0680. [DOI] [PubMed] [Google Scholar]
- 8.Collet JP, Montalescot G. Platelet function testing and implications for clinical practice. J Cardiovasc Pharmacol Ther. 2009;14(3):157–169. doi: 10.1177/1074248409339309. [DOI] [PubMed] [Google Scholar]
- 9.Blais N, Pharand C, Lordkipanidze M, Sia YK, Merhi Y, Diodati JG. Response to aspirin in healthy individuals. Cross-comparison of light transmission aggregometry, VerifyNow system, platelet count drop, thromboelastography (TEG) and urinary 11-dehydrothromboxane B(2) Thromb Haemost. 2009;102(2):404–411. doi: 10.1160/TH09-02-0126. [DOI] [PubMed] [Google Scholar]
- 10.Lordkipanidze M, Pharand C, Schampaert E, Turgeon J, Palisaitis DA, Diodati JG. A comparison of six major platelet function tests to determine the prevalence of aspirin resistance in patients with stable coronary artery disease. Eur Heart J. 2007;28(14):1702–1708. doi: 10.1093/eurheartj/ehm226. [DOI] [PubMed] [Google Scholar]
- 11.Santilli F, Rocca B, De Cristofaro R, et al. Platelet cyclooxygenase inhibition by low-dose aspirin is not reflected consistently by platelet function assays: implications for aspirin ‘resistance’. J Am Coll Cardiol. 2009;53(8):667–677. doi: 10.1016/j.jacc.2008.10.047. [DOI] [PubMed] [Google Scholar]
- 12.Cuisset T, Frere C, Poyet R, et al. Clopidogrel response: head-to-head comparison of different platelet assays to identify clopidogrel non responder patients after coronary stenting. Arch Cardiovasc Dis. 2010;103(1):39–45. doi: 10.1016/j.acvd.2009.11.004. [DOI] [PubMed] [Google Scholar]
- 13.Gurbel PA, Bliden KP, DiChiara J, et al. Evaluation of dose-related effects of aspirin on platelet function: results from the Aspirin-Induced Platelet Effect (ASPECT) study. Circulation. 2007;115(25):3156–3164. doi: 10.1161/CIRCULATIONAHA.106.675587. [DOI] [PubMed] [Google Scholar]
- 14.Broadley AJ, Gapper P, Schmitt M, Frenneaux MP. Supine rest reduces platelet activation and aggregation. Platelets. 2003;14(1):3–7. doi: 10.1080/0953710021000034447. [DOI] [PubMed] [Google Scholar]
- 15.Smock KJ, Rodgers GM. Laboratory evaluation of aspirin responsiveness. Am J Hematol. 2010;85(5):358–360. doi: 10.1002/ajh.21674. [DOI] [PubMed] [Google Scholar]
- 16.Grotemeyer KH. The platelet-reactivity-test – a useful ‘by-product’ of the blood-sampling procedure? Thromb Res. 1991;61(4):423–431. doi: 10.1016/0049-3848(91)90656-h. [DOI] [PubMed] [Google Scholar]
- 17.Dalby MC, Davidson SJ, Burman JF, Davies SW. Diurnal variation in platelet aggregation with the PFA-100 platelet function analyser. Platelets. 2000;11(6):320–324. doi: 10.1080/09537100050144731. [DOI] [PubMed] [Google Scholar]
- 18.Pamukcu B, Oflaz H, Acar RD, et al. The role of exercise on platelet aggregation in patients with stable coronary artery disease: exercise induces aspirin resistant platelet activation. J Thromb Thrombolysis. 2005;20(1):17–22. doi: 10.1007/s11239-005-2318-1. [DOI] [PubMed] [Google Scholar]
- 19.Gremmel T, Steiner S, Seidinger D, Koppensteiner R, Panzer S, Kopp CW. Smoking promotes clopidogrel-mediated platelet inhibition in patients receiving dual antiplatelet therapy. Thromb Res. 2009;124(5):588–591. doi: 10.1016/j.thromres.2009.06.012. [DOI] [PubMed] [Google Scholar]
- 20.Addad F, Chakroune T, Asma A, et al. Clopidogrel but not Aspirin prevents acute smoking-induced platelet aggregation in patients with stable coronary artery disease. Thromb Res. 2009;123(4):640–643. doi: 10.1016/j.thromres.2008.05.001. [DOI] [PubMed] [Google Scholar]
- 21.Gremmel T, Steiner S, Seidinger D, Koppensteiner R, Panzer S, Kopp CW. Adenosine diphosphate-inducible platelet reactivity shows a pronounced age dependency in the initial phase of antiplatelet therapy with clopidogrel. J Thromb Haemost. 2010;8(1):37–42. doi: 10.1111/j.1538-7836.2009.03644.x. [DOI] [PubMed] [Google Scholar]
- 22.Zuern CS, Lindemann S, Gawaz M. Platelet function and response to aspirin: gender-specific features and implications for female thrombotic risk and management. Semin Thromb Hemost. 2009;35(3):295–306. doi: 10.1055/s-0029-1222608. [DOI] [PubMed] [Google Scholar]
- 23.Modica A, Karlsson F, Mooe T. Platelet aggregation and aspirin non-responsiveness increase when an acute coronary syndrome is complicated by an infection. J Thromb Haemost. 2007;5(3):507–511. doi: 10.1111/j.1538-7836.2007.02378.x. [DOI] [PubMed] [Google Scholar]
- 24.Tamminen M, Lassila R, Westerbacka J, Vehkavaara S, Yki-Jarvinen H. Obesity is associated with impaired platelet-inhibitory effect of acetylsalicylic acid in nondiabetic subjects. Int J Obes Relat Metab Disord. 2003;27(8):907–911. doi: 10.1038/sj.ijo.0802312. [DOI] [PubMed] [Google Scholar]
- 25.Ertugrul DT, Tutal E, Yildiz M, et al. Aspirin resistance is associated with glycemic control, the dose of aspirin, and obesity in Type 2 diabetes mellitus. J Clin Endocrinol Metab. 2010;95(6):2897–2901. doi: 10.1210/jc.2009-2392. [DOI] [PubMed] [Google Scholar]
- 26.Hovens MM, Snoep JD, Groeneveld Y, Tamsma JT, Eikenboom JC, Huisman MV. High levels of low-density lipoprotein cholesterol and triglycerides and suboptimal glycemic control predict diminished ex vivo aspirin responsiveness in patients with Type 2 diabetes. J Thromb Haemost. 2007;5(7):1562–1564. doi: 10.1111/j.1538-7836.2007.02596.x. [DOI] [PubMed] [Google Scholar]
- 27.Lee PY, Chen WH, Ng W, et al. Low-dose aspirin increases aspirin resistance in patients with coronary artery disease. Am J Med. 2005;118(7):723–727. doi: 10.1016/j.amjmed.2005.03.041. [DOI] [PubMed] [Google Scholar]
- 28.Karepov V, Tolpina G, Kuliczkowski W, Serebruany V. Plasma triglycerides as predictors of platelet responsiveness to aspirin in patients after first ischemic stroke. Cerebrovasc Dis. 2008;26(3):272–276. doi: 10.1159/000147455. [DOI] [PubMed] [Google Scholar]
- 29.Bouman HJ, van Werkum JW, Hackeng CM, Verheugt FW, Ten Berg JM. The importance of anticoagulant agents in measuring platelet aggregation in patients treated with clopidogrel and aspirin. J Thromb Haemost. 2008;6(6):1040–1042. doi: 10.1111/j.1538-7836.2008.02971.x. [DOI] [PubMed] [Google Scholar]
- 30.Mansour K, Taher AT, Musallam KM, Alam S. Aspirin resistance. Adv Hematol. 2009;2009:937352. doi: 10.1155/2009/937352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Hedegaard SS, Hvas AM, Grove EL, et al. Optical platelet aggregation versus thromboxane metabolites in healthy individuals and patients with stable coronary artery disease after low-dose aspirin administration. Thromb Res. 2009;124(1):96–100. doi: 10.1016/j.thromres.2008.12.034. [DOI] [PubMed] [Google Scholar]
- 32.Alberts MJ. Platelet function testing for aspirin resistance is reasonable to do: yes! Stroke. 2010;41(10):2400–2401. doi: 10.1161/STROKEAHA.110.595637. [DOI] [PubMed] [Google Scholar]
- 33.Ziegler S, Maca T, Alt E, Speiser W, Schneider B, Minar E. Monitoring of antiplatelet therapy with the PFA-100 in peripheral angioplasty patients. Platelets. 2002;13(8):493–497. doi: 10.1080/0953710021000057866. [DOI] [PubMed] [Google Scholar]
- 34.Grundmann K, Jaschonek K, Kleine B, Dichgans J, Topka H. Aspirin non-responder status in patients with recurrent cerebral ischemic attacks. J Neurol. 2003;50(1):63–66. doi: 10.1007/s00415-003-0954-y. [DOI] [PubMed] [Google Scholar]
- 35.Linnemann B, Schwonberg J, Toennes SW, Mani H, Lindhoff-Last E. Variability of residual platelet function despite clopidogrel treatment in patients with peripheral arterial occlusive disease. Atherosclerosis. 2010;209(2):504–509. doi: 10.1016/j.atherosclerosis.2009.05.039. [DOI] [PubMed] [Google Scholar]
- 36.Lev EI. Aspirin resistance transient laboratory finding or important clinical entity? J Coll Cardiol. 2009;53(8):678–680. doi: 10.1016/j.jacc.2008.11.018. [DOI] [PubMed] [Google Scholar]
- 37.Helgason CM, Bolin KM, Hoff JA, et al. Development of aspirin resistance in persons with previous ischemic stroke. Stroke. 1994;25(12):2331–2336. doi: 10.1161/01.str.25.12.2331. [DOI] [PubMed] [Google Scholar]
- 38.Schwartz KA, Schwartz DE, Barber K, Reeves M, De Franco AC. Non-compliance is the predominant cause of aspirin resistance in chronic coronary arterial disease patients. J Transl Med. 2008;6:46. doi: 10.1186/1479-5876-6-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Serebruany V, Cherala G, Williams C, et al. Association of platelet responsiveness with clopidogrel metabolism: role of compliance in the assessment of ‘resistance’. Am Heart J. 2009;158(6):925–932. doi: 10.1016/j.ahj.2009.10.012. [DOI] [PubMed] [Google Scholar]
- 40.Hamann GF, Weimar C, Glahn J, Busse O, Diener HC. Adherence to secondary stroke prevention strategies – results from the German Stroke Data Bank. Cerebrovasc Dis. 2003;15(4):282–288. doi: 10.1159/000069490. [DOI] [PubMed] [Google Scholar]
- 41.Herlitz J, Toth PP, Naesdal J. Low-dose aspirin therapy for cardiovascular prevention: quantification and consequences of poor compliance or discontinuation. Am J Cardiovasc Drugs. 2010;10(2):125–141. doi: 10.2165/11318440-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 42.Cuisset T, Frere C, Quilici J, et al. Aspirin noncompliance is the major cause of ‘aspirin resistance’ in patients undergoing coronary stenting. Am Heart J. 2009;157(5):889–893. doi: 10.1016/j.ahj.2009.02.013. [DOI] [PubMed] [Google Scholar]
- 43.Cox D, Maree AO, Dooley M, Conroy R, Byrne MF, Fitzgerald DJ. Effect of enteric coating on antiplatelet activity of low-dose aspirin in healthy volunteers. Stroke. 2006;37(8):2153–2158. doi: 10.1161/01.STR.0000231683.43347.ec. [DOI] [PubMed] [Google Scholar]
- 44.Maree AO, Fitzgerald DJ. Variable platelet response to aspirin and clopidogrel in atherothrombotic disease. Circulation. 2007;115(16):2196–2207. doi: 10.1161/CIRCULATIONAHA.106.675991. [DOI] [PubMed] [Google Scholar]
- 45.Schwertner HA, McGlasson D, Christopher M, Bush AC. Effects of different aspirin formulations on platelet aggregation times and on plasma salicylate concentrations. Thromb Res. 2006;118(4):529–534. doi: 10.1016/j.thromres.2005.10.001. [DOI] [PubMed] [Google Scholar]
- 46.Catella-Lawson F, Reilly MP, Kapoor SC, et al. Cyclooxygenase inhibitors and the antiplatelet effects of aspirin. N Engl J Med. 2001;345(25):1809–1817. doi: 10.1056/NEJMoa003199. [DOI] [PubMed] [Google Scholar]
- 47.Gilard M, Arnaud B, Cornily JC, et al. Influence of omeprazole on the antiplatelet action of clopidogrel associated with aspirin: the randomized, double-blind OCLA (Omeprazole Clopidogrel Aspirin) study. J Am Coll Cardiol. 2008;51(3):256–260. doi: 10.1016/j.jacc.2007.06.064. [DOI] [PubMed] [Google Scholar]
- 48.Cuisset T, Frere C, Quilici J, et al. Comparison of omeprazole and pantoprazole influence on a high 150-mg clopidogrel maintenance dose the PACA (Proton Pump Inhibitors and Clopidogrel Association) prospective randomized study. J Am Coll Cardiol. 2009;54(13):1149–1153. doi: 10.1016/j.jacc.2009.05.050. [DOI] [PubMed] [Google Scholar]
- 49.Small DS, Farid NA, Payne CD, et al. Effects of the proton pump inhibitor lansoprazole on the pharmacokinetics and pharmacodynamics of prasugrel and clopidogrel. J Clin Pharmacol. 2008;48(4):475–484. doi: 10.1177/0091270008315310. [DOI] [PubMed] [Google Scholar]
- 50.Sibbing D, Morath T, Stegherr J, et al. Impact of proton pump inhibitors on the antiplatelet effects of clopidogrel. Thromb Haemost. 2009;101(4):714–719. [PubMed] [Google Scholar]
- 51.Juurlink DN, Gomes T, Ko DT, et al. A population-based study of the drug interaction between proton pump inhibitors and clopidogrel. CMAJ. 2009;180(7):713–718. doi: 10.1503/cmaj.082001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ho PM, Maddox TM, Wang L, et al. Risk of adverse outcomes associated with concomitant use of clopidogrel and proton pump inhibitors following acute coronary syndrome. JAMA. 2009;301(9):937–944. doi: 10.1001/jama.2009.261. [DOI] [PubMed] [Google Scholar]
- 53.O’Donoghue ML, Braunwald E, Antman EM, et al. Pharmacodynamic effect and clinical efficacy of clopidogrel and prasugrel with or without a proton-pump inhibitor: an analysis of two randomised trials. Lancet. 2009;374(9694):989–997. doi: 10.1016/S0140-6736(09)61525-7. [DOI] [PubMed] [Google Scholar]
- 54.Rassen JA, Choudhry NK, Avorn J, Schneeweiss S. Cardiovascular outcomes and mortality in patients using clopidogrel with proton pump inhibitors after percutaneous coronary intervention or acute coronary syndrome. Circulation. 2009;120(23):2322–2329. doi: 10.1161/CIRCULATIONAHA.109.873497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lau WC, Waskell LA, Watkins PB, et al. Atorvastatin reduces the ability of clopidogrel to inhibit platelet aggregation: a new drug–drug interaction. Circulation. 2003;107(1):32–37. doi: 10.1161/01.cir.0000047060.60595.cc. [DOI] [PubMed] [Google Scholar]
- 56.Neubauer H, Gunesdogan B, Hanefeld C, Spiecker M, Mugge A. Lipophilic statins interfere with the inhibitory effects of clopidogrel on platelet function – a flow cytometry study. Eur Heart J. 2003;24(19):1744–1749. doi: 10.1016/s0195-668x(03)00442-1. [DOI] [PubMed] [Google Scholar]
- 57.Saw J, Steinhubl SR, Berger PB, et al. Lack of adverse clopidogrel–atorvastatin clinical interaction from secondary analysis of a randomized, placebo-controlled clopidogrel trial. Circulation. 2003;108(8):921–924. doi: 10.1161/01.CIR.0000088780.57432.43. [DOI] [PubMed] [Google Scholar]
- 58.Mitsios JV, Papathanasiou AI, Rodis FI, Elisaf M, Goudevenos JA, Tselepis AD. Atorvastatin does not affect the antiplatelet potency of clopidogrel when it is administered concomitantly for 5 weeks in patients with acute coronary syndromes. Circulation. 2004;109(11):1335–1338. doi: 10.1161/01.CIR.0000124581.18191.15. [DOI] [PubMed] [Google Scholar]
- 59.Saw J, Brennan DM, Steinhubl SR, et al. Lack of evidence of a clopidogrel–statin interaction in the CHARISMA trial. J Am Coll Cardiol. 2007;50(4):291–295. doi: 10.1016/j.jacc.2007.01.097. [DOI] [PubMed] [Google Scholar]
- 60.Gremmel T, Steiner S, Seidinger D, Koppensteiner R, Panzer S, Kopp CW. Calcium-channel blockers decrease clopidogrel-mediated platelet inhibition. Heart. 2010;96(3):186–189. doi: 10.1136/hrt.2009.171488. [DOI] [PubMed] [Google Scholar]
- 61.Harmsze AM, Robijns K, van Werkum JW, et al. The use of amlodipine, but not of P-glycoprotein inhibiting calcium channel blockers is associated with clopidogrel poor-response. Thromb Haemost. 2010;103(5):920–925. doi: 10.1160/TH09-08-0516. [DOI] [PubMed] [Google Scholar]
- 62.Siller-Matula JM, Lang I, Christ G, Jilma B. Calcium-channel blockers reduce the antiplatelet effect of clopidogrel. J Am Coll Cardiol. 2008;52(19):1557–1563. doi: 10.1016/j.jacc.2008.07.055. [DOI] [PubMed] [Google Scholar]
- 63.Feher G, Feher A, Pusch G, Lupkovics G, Szapary L, Papp E. The genetics of antiplatelet drug resistance. Clin Genet. 2009;75(1):1–18. doi: 10.1111/j.1399-0004.2008.01105.x. [DOI] [PubMed] [Google Scholar]
- 64.Goodman T, Ferro A, Sharma P. Pharmacogenetics of aspirin resistance: a comprehensive systematic review. Br J Clin Pharmacol. 2008;66(2):222–232. doi: 10.1111/j.1365-2125.2008.03183.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Simon T, Verstuyft C, Mary-Krause M, et al. Genetic determinants of response to clopidogrel and cardiovascular events. N Engl J Med. 2009;360(4):363–375. doi: 10.1056/NEJMoa0808227. [DOI] [PubMed] [Google Scholar]
- 66.Sugunaraj JP, Palaniswamy C, Selvaraj DR, Chaitanya Arudra SK, Sukhija R. Clopidogrel resistance. Am J Ther. 2010;17(2):210–215. doi: 10.1097/MJT.0b013e3181bdc3e4. [DOI] [PubMed] [Google Scholar]
- 67.Collet JP, Hulot JS, Pena A, et al. Cytochrome P450 2C19 polymorphism in young patients treated with clopidogrel after myocardial infarction: a cohort study. Lancet. 2009;373(9660):309–317. doi: 10.1016/S0140-6736(08)61845-0. [DOI] [PubMed] [Google Scholar]
- 68.Patti G, Colonna G, Pasceri V, Pepe LL, Montinaro A, Di Sciascio G. Randomized trial of high loading dose of clopidogrel for reduction of periprocedural myocardial infarction in patients undergoing coronary intervention: results from the ARMYDA-2 (Antiplatelet therapy for Reduction of Myocardial Damage during Angioplasty) study. Circulation. 2005;111(16):2099–2106. doi: 10.1161/01.CIR.0000161383.06692.D4. [DOI] [PubMed] [Google Scholar]
- 69.Seip RL, Duconge J, Ruano G. Implementing genotype-guided antithrombotic therapy. Future Cardiol. 2010;6(3):409–424. doi: 10.2217/fca.10.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Michelson AD, Cattaneo M, Eikelboom JW, et al. Aspirin resistance: position paper of the Working Group on Aspirin Resistance. J Thromb Haemost. 2005;3(6):1309–1311. doi: 10.1111/j.1538-7836.2005.01351.x. [DOI] [PubMed] [Google Scholar]
- 71.Gasparyan AY, Watson T, Lip GY. The role of aspirin in cardiovascular prevention: implications of aspirin resistance. J Am Coll Cardiol. 2008;51(19):1829–1843. doi: 10.1016/j.jacc.2007.11.080. [DOI] [PubMed] [Google Scholar]
- 72.Angiolillo DJ, Suryadevara S. Aspirin and clopidogrel: efficacy and resistance in diabetes mellitus. Best Pract Res Clin Endocrinol Metab. 2009;23(3):375–388. doi: 10.1016/j.beem.2008.12.001. [DOI] [PubMed] [Google Scholar]
- 73.Grove EL, Hvas AM, Kristensen SD. Immature platelets in patients with acute coronary syndromes. Thromb Haemost. 2009;101(1):151–156. [PubMed] [Google Scholar]
- 74.Gaborit B, Frere C, Cuisset T, Alessi MC, Dutour A. Enhanced post-clopidogrel platelet reactivity in diabetic patients is independently related to plasma fibrinogen level but not to glycemic control. J Thromb Haemost. 2009;7(11):1939–1941. doi: 10.1111/j.1538-7836.2009.03599.x. [DOI] [PubMed] [Google Scholar]
- 75.Pamukcu B, Oflaz H, Onur I, et al. Relationship between the serum sCD40L level and aspirin-resistant platelet aggregation in patients with stable coronary artery disease. Circ J. 2008;72(1):61–66. doi: 10.1253/circj.72.61. [DOI] [PubMed] [Google Scholar]
- 76.Erlinge D, Varenhorst C, Braun OO, et al. Patients with poor responsiveness to thienopyridine treatment or with diabetes have lower levels of circulating active metabolite, but their platelets respond normally to active metabolite added ex vivo. J Am Coll Cardiol. 2008;52(24):1968–1977. doi: 10.1016/j.jacc.2008.07.068. [DOI] [PubMed] [Google Scholar]
- 77.Simon DI, Schmaier AH. Sweet and sticky: diabetic platelets, enhanced reactivity, and cardiovascular risk. J Am Coll Cardiol. 2007;50(16):1548–1550. doi: 10.1016/j.jacc.2007.07.023. [DOI] [PubMed] [Google Scholar]
- 78.Watala C, Pluta J, Golanski J, et al. Increased protein glycation in diabetes mellitus is associated with decreased aspirin-mediated protein acetylation and reduced sensitivity of blood platelets to aspirin. J Mol Med. 2005;83(2):148–158. doi: 10.1007/s00109-004-0600-x. [DOI] [PubMed] [Google Scholar]
- 79.Michelson AD, Frelinger AL, 3rd, Furman MI. Current options in platelet function testing. Am J Cardiol. 2006;98(10A):4N–10N. doi: 10.1016/j.amjcard.2006.09.008. [DOI] [PubMed] [Google Scholar]
- 80.Ohmori T, Yatomi Y, Nonaka T, et al. Aspirin resistance detected with aggregometry cannot be explained by cyclooxygenase activity: involvement of other signaling pathway(s) in cardiovascular events of aspirin-treated patients. J Thromb Haemost. 2006;4(6):1271–1278. doi: 10.1111/j.1538-7836.2006.01958.x. [DOI] [PubMed] [Google Scholar]
- 81.Harrison PKD. Laboratory monitoring of antiplatelet therapy. In: Gresele P, Fuster V, Lopez J, Page C, Vermylen J, editors. Platelets in Hematologic and Cardiovascular Disorders – A Clinical Handbook. Cambridge University Press; Cambridge, UK: 2008. pp. 386–407. [Google Scholar]
- 82.Favaloro EJ. Clinical utility of the PFA-100. Semin Thromb Hemost. 2008;34(8):709–733. doi: 10.1055/s-0029-1145254. [DOI] [PubMed] [Google Scholar]
- 83.Harrison PKD. Clinical tests of platelet function. In: Michelson A, editor. Platelets. Elsevier Inc./Academic Press; CA, USA: 2007. pp. 445–474. [Google Scholar]
- 84.Velik-Salchner C, Maier S, Innerhofer P, et al. Point-of-care whole blood impedance aggregometry versus classical light transmission aggregometry for detecting aspirin and clopidogrel: the results of a pilot study. Anesth Analg. 2008;107(6):1798–1806. doi: 10.1213/ane.0b013e31818524c1. [DOI] [PubMed] [Google Scholar]
- 85.Grotemeyer KH, Scharafinski HW, Husstedt IW. Two-year follow-up of aspirin responder and aspirin non responder. A pilot-study including 180 post-stroke patients. Thromb Res. 1993;71(5):397–403. doi: 10.1016/0049-3848(93)90164-j. [DOI] [PubMed] [Google Scholar]
- 86.Gengo FM, Rainka M, Robson M, et al. Prevalence of platelet nonresponsiveness to aspirin in patients treated for secondary stroke prophylaxis and in patients with recurrent ischemic events. J Clin Pharmacol. 2008;48(3):335–343. doi: 10.1177/0091270007313324. [DOI] [PubMed] [Google Scholar]
- 87.Snoep JD, Hovens MM, Eikenboom JC, van der Bom JG, Huisman MV. Association of laboratory-defined aspirin resistance with a higher risk of recurrent cardiovascular events: a systematic review and meta-analysis. Arch Intern Med. 2007;167(15):1593–1599. doi: 10.1001/archinte.167.15.1593. [DOI] [PubMed] [Google Scholar]
- •88.Krasopoulos G, Brister SJ, Beattie WS, Buchanan MR. Aspirin ‘resistance’ and risk of cardiovascular morbidity: systematic review and meta-analysis. BMJ. 2008;336(7637):195–198. doi: 10.1136/bmj.39430.529549.BE. Well-conducted meta-analysis that reports a higher long-term risk of cardiovascular morbidity in patients with aspirin resistance compared with patients responsive to aspirin. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Snoep JD, Hovens MM, Eikenboom JC, van der Bom JG, Jukema JW, Huisman MV. Clopidogrel nonresponsiveness in patients undergoing percutaneous coronary intervention with stenting: a systematic review and meta-analysis. Am Heart J. 2007;154(2):221–231. doi: 10.1016/j.ahj.2007.04.014. [DOI] [PubMed] [Google Scholar]
- 90.Bonello L, Camoin-Jau L, Arques S, et al. Adjusted clopidogrel loading doses according to vasodilator-stimulated phosphoprotein phosphorylation index decrease rate of major adverse cardiovascular events in patients with clopidogrel resistance: a multicenter randomized prospective study. J Am Coll Cardiol. 2008;51(14):1404–1411. doi: 10.1016/j.jacc.2007.12.044. [DOI] [PubMed] [Google Scholar]
- 91.Hovens MM, Snoep JD, Eikenboom JC, van der Bom JG, Mertens BJ, Huisman MV. Prevalence of persistent platelet reactivity despite use of aspirin: a systematic review. Am Heart J. 2007;153(2):175–181. doi: 10.1016/j.ahj.2006.10.040. [DOI] [PubMed] [Google Scholar]
- 92.Antithrombotic Trialists’ Collaboration. Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, and stroke in high risk patients. BMJ. 2002;324(7329):71–86. doi: 10.1136/bmj.324.7329.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Gladding P, Webster M, Zeng I, et al. The pharmacogenetics and pharmacodynamics of clopidogrel response: an analysis from the PRINC (Plavix Response in Coronary Intervention) trial. JACC Cardiovasc Interv. 2008;1(6):620–627. doi: 10.1016/j.jcin.2008.09.008. [DOI] [PubMed] [Google Scholar]
- 94.Lev EI, Patel RT, Maresh KJ, et al. Aspirin and clopidogrel drug response in patients undergoing percutaneous coronary intervention: the role of dual drug resistance. J Am Coll Cardiol. 2006;47(1):27–33. doi: 10.1016/j.jacc.2005.08.058. [DOI] [PubMed] [Google Scholar]
- 95.Milicic D, Skoric B, Lovric D. Drug-specific thienopyridine resistance in patient with recurrent coronary stent thrombosis. J Invasive Cardiol. 2009;21(8):E157–E160. [PubMed] [Google Scholar]
- 96.Campo G, Valgimigli M, Gemmati D, et al. Poor responsiveness to clopidogrel: drug-specific or class-effect mechanism? Evidence from a clopidogrel-to-ticlopidine crossover study. J Am Coll Cardiol. 2007;50(12):1132–1137. doi: 10.1016/j.jacc.2007.04.092. [DOI] [PubMed] [Google Scholar]
- 97.Gladding P, Webster M, Ormiston J, Olsen S, White H. Antiplatelet drug nonresponsiveness. Am Heart J. 2008;155(4):591–599. doi: 10.1016/j.ahj.2007.12.034. [DOI] [PubMed] [Google Scholar]
- 98.Gladding PA, Webster MW, Farrell HB, Zeng IS, Park R, Ruijne N. The antiplatelet effect of six non-steroidal anti-inflammatory drugs and their pharmacodynamic interaction with aspirin in healthy volunteers. Am J Cardiol. 2008;101(7):1060–1063. doi: 10.1016/j.amjcard.2007.11.054. [DOI] [PubMed] [Google Scholar]
- 99.Schroeder WS, Ghobrial L, Gandhi PJ. Possible mechanisms of drug-induced aspirin and clopidogrel resistance. J Thromb Thrombolysis. 2006;22(2):139–150. doi: 10.1007/s11239-006-8670-y. [DOI] [PubMed] [Google Scholar]
- 100.Neubauer H, Engelhardt A, Kruger JC, et al. Pantoprazole does not influence the antiplatelet effect of clopidogrel – a whole blood aggregometry study following coronary stenting. J Cardiovasc Pharmacol. 2010;56(1):91–97. doi: 10.1097/FJC.0b013e3181e19739. [DOI] [PubMed] [Google Scholar]
- 101.Stockl KM, Le L, Zakharyan A, et al. Risk of rehospitalization for patients using clopidogrel with a proton pump inhibitor. Arch Intern Med. 2010;170(8):704–710. doi: 10.1001/archinternmed.2010.34. [DOI] [PubMed] [Google Scholar]
- 102.Abraham NS, Hlatky MA, Antman EM, et al. ACCF/ACG/AHA 2010 Expert Consensus Document on the Concomitant Use of Proton Pump Inhibitors and Thienopyridines: A Focused Update of the ACCF/ACG/AHA 2008 Expert Consensus Document on Reducing the Gastrointestinal Risks of Antiplatelet Therapy and NSAID Use. Circulation. 2010;122(24):2619–2633. doi: 10.1161/CIR.0b013e318202f701. [DOI] [PubMed] [Google Scholar]
- 103.Bhindi R, Ormerod O, Newton J, Banning AP, Testa L. Interaction between statins and clopidogrel: is there anything clinically relevant? Q JM. 2008;101(12):915–925. doi: 10.1093/qjmed/hcn089. [DOI] [PubMed] [Google Scholar]
- 104.Berger PB, Bhatt DL, Fuster V, et al. Bleeding complications with dual antiplatelet therapy among patients with stable vascular disease or risk factors for vascular disease: results from the Clopidogrel for High Atherothrombotic Risk and Ischemic Stabilization, Management, and Avoidance (CHARISMA) trial. Circulation. 2010;121(23):2575–2583. doi: 10.1161/CIRCULATIONAHA.109.895342. [DOI] [PubMed] [Google Scholar]
- 105.Grotemeyer KH. Effects of acetylsalicylic acid in stroke patients. Evidence of nonresponders in a subpopulation of treated patients. Thromb Res. 1991;63(6):587–593. doi: 10.1016/0049-3848(91)90085-b. [DOI] [PubMed] [Google Scholar]
- 106••.Helgason CM, Tortorice KL, Winkler SR, et al. Aspirin response and failure in cerebral infarction. Stroke. 1993;24(3):345–350. doi: 10.1161/01.str.24.3.345. Novel article introducing the concept of aspirin resistance in ischemic stroke. [DOI] [PubMed] [Google Scholar]
- 107.Grau AJ, Reiners S, Lichy C, Buggle F, Ruf A. Platelet function under aspirin, clopidogrel, and both after ischemic stroke: a case-crossover study. Stroke. 2003;34(4):849–854. doi: 10.1161/01.STR.0000064326.65899.AC. [DOI] [PubMed] [Google Scholar]
- 108.Alberts MJ, Bergman DL, Molner E, Jovanovic BD, Ushiwata I, Teruya J. Antiplatelet effect of aspirin in patients with cerebrovascular disease. Stroke. 2004;35(1):175–178. doi: 10.1161/01.STR.0000106763.46123.F6. [DOI] [PubMed] [Google Scholar]
- 109.Macchi L, Petit E, Brizard A, Gil R, Neau JP. Aspirin resistance in vitro and hypertension in stroke patients. J Thromb Haemost. 2004;2(11):2051–2053. doi: 10.1111/j.1538-7836.2004.00953.x. [DOI] [PubMed] [Google Scholar]
- 110•.Harrison P, Segal H, Blasbery K, Furtado C, Silver L, Rothwell PM. Screening for aspirin responsiveness after transient ischemic attack and stroke: comparison of 2 point-of-care platelet function tests with optical aggregometry. Stroke. 2005;36(5):1001–1005. doi: 10.1161/01.STR.0000162719.11058.bd. Demonstrates that the prevalence of aspirin resistance is highly test-dependent in patients with stroke. [DOI] [PubMed] [Google Scholar]
- 111.McCabe DJ, Harrison P, Mackie IJ, et al. Assessment of the antiplatelet effects of low to medium dose aspirin in the early and late phases after ischaemic stroke and TIA. Platelets. 2005;16(5):269–280. doi: 10.1080/09537100400020567. [DOI] [PubMed] [Google Scholar]
- 112.Berrouschot J, Schwetlick B, von Twickel G, et al. Aspirin resistance in secondary stroke prevention. Acta Neurol Scand. 2006;113(1):31–35. doi: 10.1111/j.1600-0404.2005.00419.x. [DOI] [PubMed] [Google Scholar]
- 113.Hohlfeld T, Weber AA, Junghans U, et al. Variable platelet response to aspirin in patients with ischemic stroke. Cerebrovasc Dis. 2007;24(1):43–50. doi: 10.1159/000103115. [DOI] [PubMed] [Google Scholar]
- 114.Bennett D, Yan B, Macgregor L, Eccleston D, Davis SM. A pilot study of resistance to aspirin in stroke patients. J Clin Neurosci. 2008;15(11):1204–1209. doi: 10.1016/j.jocn.2008.01.006. [DOI] [PubMed] [Google Scholar]
- 115.Englyst NA, Horsfield G, Kwan J, Byrne CD. Aspirin resistance is more common in lacunar strokes than embolic strokes and is related to stroke severity. J Cereb Blood Flow Metab. 2008;28(6):1196–1203. doi: 10.1038/jcbfm.2008.9. [DOI] [PubMed] [Google Scholar]
- 116.Seok JI, Joo IS, Yoon JH, et al. Can aspirin resistance be clinically predicted in stroke patients? Clin Neurol Neurosurg. 2008;110(2):110–116. doi: 10.1016/j.clineuro.2007.09.005. [DOI] [PubMed] [Google Scholar]
- 117.Cha JK, Jeon HW, Kang MJ. ADP-induced platelet aggregation in acute ischemic stroke patients on aspirin therapy. Eur J Neurol. 2008;15(12):1304–1308. doi: 10.1111/j.1468-1331.2008.02306.x. [DOI] [PubMed] [Google Scholar]
- 118.Boncoraglio GB, Bodini A, Brambilla C, Corsini E, Carriero MR, Parati EA. Aspirin resistance determined with PFA-100 does not predict new thrombotic events in patients with stable ischemic cerebrovascular disease. Clin Neurol Neurosurg. 2009;111(3):270–273. doi: 10.1016/j.clineuro.2008.11.001. [DOI] [PubMed] [Google Scholar]
- 119.Bernstein PL, Jacobson BF, Connor MD, Becker PJ. Aspirin resistance in South African Caucasian patients with thrombotic cerebrovascular events. J Neurol Sci. 2009;277(1–2):80–82. doi: 10.1016/j.jns.2008.10.013. [DOI] [PubMed] [Google Scholar]
- 120.von Lewinski F, Riggert J, Paulus W. Towards a rationale of platelet aggregation monitoring in stroke prophylaxis? J Stroke Cerebrovasc Dis. 2009;18(2):111–115. doi: 10.1016/j.jstrokecerebrovasdis.2008.09.010. [DOI] [PubMed] [Google Scholar]
- 121.Lee JH, Cha JK, Lee SJ, Ha SW, Kwon SU. Addition of cilostazol reduces biological aspirin resistance in aspirin users with ischaemic stroke: a double-blind randomized clinical trial. Eur J Neurol. 2010;17(3):434–442. doi: 10.1111/j.1468-1331.2009.02837.x. [DOI] [PubMed] [Google Scholar]
- 122.Fong J, Cheng-Ching E, Hussain MS, Katzan I, Gupta R. Predictors of biochemical aspirin and clopidogrel resistance in patients with ischemic stroke. J Stroke Cerebrovasc Dis. 2010 doi: 10.1016/j.jstrokecerebrovasdis.2009.12.004. (Epub ahead of print) [DOI] [PubMed] [Google Scholar]
- 123.Tousoulis D, Siasos G, Stefanadis C. Aspirin resistance: what the cardiologist needs to know? Int J Cardiol. 2009;132(2):153–156. doi: 10.1016/j.ijcard.2008.12.074. [DOI] [PubMed] [Google Scholar]
- 124.Seegmiller A, Sarode R. Laboratory evaluation of platelet function. Hematol Oncol Clin North Am. 2007;21(4):731–742. vii. doi: 10.1016/j.hoc.2007.06.008. [DOI] [PubMed] [Google Scholar]
Websites
- 201.ASCET trial. www.clinicaltrial.gov/ct2/show/NCT00222261?term=NCT00222261&rank=1.
- 202.GRAVITAS trial. www.clinicaltrial.gov/ct2/show/NCT00645918?term=NCT00645918&rank=1.
- 203.ARCTIC trial. www.clinicaltrial.gov/ct2/show/NCT00827411?term=NCT00827411&rank=1.
- 204.TRIGGER-PCI trial. www.clinicaltrial.gov/ct2/show/NCT00910299?term=NCT00910299&rank=1.
