Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Expert Rev Cardiovasc Ther ; 13(12): 1333-44, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26513059

ABSTRACT

The optimal utilization of antiplatelet therapy in patients with renal impairment (RI) following acute coronary syndromes (ACS) represents an urgent, unmet and yet unsolved need with regards to the choice of agents, duration of treatment and potential dose/regimen adjustment. The lack of any large randomized trials designed and powered specifically in such high-risk patients, absence of the uniformed efficacy and safety data reporting policy to the FDA and endless overoptimistic publications based on post hoc analyses of primary trials sometimes exaggerating benefits and hiding risks, clouds reality. In addition, triaging RI patients is problematic due to ongoing kidney deterioration and the fact that such patients are prone to both vascular occlusions and bleeding. The authors summarize available FDA-confirmed evidence from the latest trials with approved antiplatelet agents, namely clopidogrel (CAPRIE, CURE, CREDO, CLARITY, CHARISMA); prasugrel (TRITON, TRILOGY); ticagrelor (PLATO, and PEGASUS); and vorapaxar (TRACER and TRA2P) in RI patient cohorts on top of aspirin as part of dual antiplatelet therapy (DAPT). We deliberately avoided any results unless they were verified by the FDA, with the exception of the recent PEGASUS, since Agency reviews are not yet available. Despite differences among the trials and DAPT choices, RI patients universally experience much higher (HR = 1.3-3.1) rates of primary endpoint events, and bleeding risks (HR = 1.7-3.6). However, only ticagrelor increases creatinine and uric acid levels above that of clopidogrel; has the worst incidence of serious adverse events, more adverse events, and inferior outcomes in patients with severe (eGFR <30 ml/min), especially in the lowest (eGFR <15 ml/min) RI subsets. Clopidogrel, prasugrel and vorapaxar appear safer. Moreover, less aggressive half dose (5 mg/daily) prasugrel and strict DAPT, are well justified in RI, but not predominantly triple strategies with vorapaxar as tested in TRA2P and especially in TRACER. In conclusion, data from clinical trials, their sub-studies and affiliated FDA reviews indicate that RI cause more vascular occlusions and bleeding in ACS patients treated with DAPT. Among the novel antiplatelet agents, prasugrel and vorapaxar, but probably not ticagrelor, offer advantage in RI patients.


Subject(s)
Acute Coronary Syndrome , Adenosine/analogs & derivatives , Lactones/pharmacology , Prasugrel Hydrochloride/pharmacology , Pyridines/pharmacology , Renal Insufficiency , Ticlopidine/analogs & derivatives , Acute Coronary Syndrome/complications , Acute Coronary Syndrome/drug therapy , Adenosine/pharmacology , Clinical Trials as Topic , Clopidogrel , Comparative Effectiveness Research , Glomerular Filtration Rate , Hemorrhage/chemically induced , Hemorrhage/prevention & control , Humans , Patient Selection , Platelet Aggregation Inhibitors/pharmacology , Renal Insufficiency/complications , Renal Insufficiency/diagnosis , Ticagrelor , Ticlopidine/pharmacology
2.
Am J Ther ; 21(6): 482-90, 2014.
Article in English | MEDLINE | ID: mdl-23698186

ABSTRACT

Valsartan is known to inhibit platelet activity in both in vitro and ex vivo clinical setting, whereas aliskiren in vitro modulates antithrombin-III in plasma. The authors tested how aliskiren and valsartan combination versus aliskiren monotherapy will affect hemostatic biomarkers in mild-to-moderate hypertensive diabetics in the frame of the Aliskiren and Valsartan Impact in Diabetics (AVID) trial. A total of 52 patients with type 2 diabetes and mild-to-moderate hypertension were equally randomized to aliskiren (150-300 mg/d) and valsartan (160 mg/d) versus aliskiren (150-300 mg/d) alone for 4 weeks. A total of 25 biomarkers were serially measured, of which 16 are related to platelet function, 6 to coagulation, and 3 to fibrinolysis. Aliskiren monotherapy has no significant impact on any of the assessed biomarkers. In contrast, valsartan on top of aliskiren provided significant inhibition of ADP-induced platelet aggregation (P=0.032), decreased shear-induced activation measured with PFA-100 analyzer (P=0.041), and diminished expression of GP IIb/IIIa activity (P=0.027) measured by PAC-1 antibody, GP Ib (CD42b, P=0.033), vitronectin receptor (CD51/61, P=0.046), P-selectin (CD62p, P=0.026), lysosome-associated membrane protein (CD107a, P=0.042), and CD40-ligand (CD154, P=0.048). In AVID trial, valsartan in combination with aliskiren mildly but significantly inhibited platelets, confirming previous observations. In contrast, aliskiren monotherapy does not enhance antithrombin activity, suggesting that previous data probably represent a laboratory artifact. Importantly, these randomized data were generated on top of low-dose daily aspirin, supporting extra benefit for combination use of angiotensin receptor blockers and renin inhibitors in high-risk diabetic population.


Subject(s)
Amides/therapeutic use , Antihypertensive Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Fumarates/therapeutic use , Hypertension/drug therapy , Tetrazoles/therapeutic use , Valine/analogs & derivatives , Amides/administration & dosage , Antihypertensive Agents/administration & dosage , Aspirin/administration & dosage , Biomarkers/metabolism , Blood Platelets/drug effects , Diabetes Mellitus, Type 2/complications , Double-Blind Method , Drug Therapy, Combination , Female , Follow-Up Studies , Fumarates/administration & dosage , Humans , Male , Middle Aged , Pilot Projects , Renin/antagonists & inhibitors , Tetrazoles/administration & dosage , Valine/administration & dosage , Valine/therapeutic use , Valsartan
3.
Thromb Haemost ; 111(1): 3-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24285121

ABSTRACT

The recently published, largest trial with cangrelor, the Cangrelor versus Standard Therapy to Achieve Optimal Management of Platelet Inhibition (CHAMPION)-PHOENIX, suggested that the experimental agent significantly reduced the rate of stent thrombosis (ST) and myocardial infarction (MI) during PCI at 48 hours (h) and 30 days. However, the declared impressive cangrelor vascular non-fatal benefit was contradicted by identical deaths at 48 h, and a trend toward excess mortality at 30 days. We analysed the mismatch between outcomes in the CHAMPION-PHOENIX trial. The trial reported identical mortality (18 death in each arm; odds ratio [OR] 1.00 (0.52-1.92); p>0.999) at 48 h, but more deaths, 60 vs 55, after cangrelor at 30 days. There was a significant reduction of ST from 0.8% (n=46) of the patients in the cangrelor group versus 1.4% (n=74) in the clopidogrel group (odds ratio, 0.62; 95% CI, 0.43 to 0.90; p= 0.01) at 48 h, and a persistent but less impressive ST prevention benefit OR of 0.68 (0.50=0.92, p = 0.01) at 30 days. There were also 48 less MI's following cangrelor usage enforced by a significant difference (odds ratio 0.80 (0.67-0.97) p = 0.02), which was also less prevalent at 30 days (OR 0.82 (0.68-0.98), p = 0.03). The reported ST/MI advantage should result in at least a trend towards numerically less deaths after cangrelor at 30 days follow-up, which was opposite of the results reported in CHAMPION-PHOENIX trial. Efficacy of cangrelor is challenged by the disproportional "reduction" of ST and MI conflicting with identical mortality at 48 h and worsened at day 30 fatalities. The dissociation between vascular mortality and non-fatal vascular ischaemic occlusions, unless compensated by some other unreported cause(s) of death, should be explored and explained. Unadjudicated 30-day outcomes, and all ST types should be fully disclosed. The ongoing FDA cangrelor review should focus on appropriate event count and/or possible mismatch between site-reported and extra adjudicated events in the CHAMPION-PHOENIX trial.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Blood Platelets/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Randomized Controlled Trials as Topic/methods , Adenosine Monophosphate/adverse effects , Adenosine Monophosphate/therapeutic use , Clopidogrel , Data Interpretation, Statistical , Humans , Myocardial Infarction/mortality , Myocardial Infarction/prevention & control , Odds Ratio , Percutaneous Coronary Intervention , Proportional Hazards Models , Research Design , Stents/adverse effects , Thrombosis/mortality , Thrombosis/prevention & control , Ticlopidine/adverse effects , Ticlopidine/analogs & derivatives , Ticlopidine/therapeutic use , Time Factors , Treatment Outcome
4.
Cardiology ; 118(3): 187-94, 2011.
Article in English | MEDLINE | ID: mdl-21701167

ABSTRACT

BACKGROUND: Prescription omega-3-acid ethyl esters (PO-3A) have been tested for outcome benefits in patients with coronary artery disease (CAD), arrhythmias and heart failure. Some evidence suggests that PO-3A may exert their benefit via inhibiting platelets. We tested the hypothesis that PO-3A may inhibit platelet activity in patients with documented stable CAD, beyond the antiplatelet properties of aspirin and statins. METHODS: Thirty patients with documented CAD and triglycerides over 250 mg/dl treated with aspirin (70-160 mg/daily) and statins (simvastatin equivalence dose: 5-40 mg/daily) were randomized 1:1:1 to Omacor™ 1 g/day (DHA/EPA ratio 1.25:1.0), Omacor 2 g/day, or a placebo for 2 weeks. Platelet tests including aggregometry and flow cytometry and cartridge analyzer readings were performed at baseline and at 1 and 2 weeks following PO-3A therapy. RESULTS: ADP-induced platelet aggregation (p = 0.037), GP IIb/IIIa antigen (p = 0.031) and activity (p = 0.024), and P-selectin (p = 0.041) were significantly reduced after PO-3A, while platelet/endothelial cell adhesion molecule (p = 0.09), vitronectin receptor (p = 0.16), formation of platelet-monocyte microparticles (p = 0.19) and the VerifyNow IIb/IIIa test (p = 0.27) only exhibited nonsignificant trends suggestive of reduced platelet activity. Finally, collagen- and arachidonic acid-induced aggregation, closure time with the PFA-100 device and expression of thrombospondin (CD36), GP Ib (CD42b), LAMP-3 (CD63), LAMP-1 (CD107a), CD40-ligand (CD154), GP37 (CD165), and PAR-1 receptor intact (SPAN 12) and cleaved (WEDE-15) epitopes were not affected by 2 weeks of PO-3A. CONCLUSION: Independently of the dose and already at 1 week, short-term therapy with PO-3A provided a modest reduction of platelet activity biomarkers, despite concomitant aspirin and statin therapy, when compared to a placebo. The effect of PO-3A is unique, differs from other known antiplatelet agents and suggests potential pleiotropism. These preliminary randomized data call for confirmation in prospective studies.


Subject(s)
Coronary Artery Disease/blood , Docosahexaenoic Acids/therapeutic use , Eicosapentaenoic Acid/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Hypertriglyceridemia/blood , Platelet Membrane Glycoproteins/metabolism , Aged , Biomarkers/blood , Coronary Artery Disease/complications , Coronary Artery Disease/drug therapy , Double-Blind Method , Drug Combinations , Female , Humans , Hypertriglyceridemia/complications , Hypertriglyceridemia/drug therapy , Male , Middle Aged , Platelet Activation/physiology , Platelet Function Tests
5.
J Cardiovasc Pharmacol Ther ; 15(4): 344-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20938039

ABSTRACT

BACKGROUND: we performed the first test in humans of whether aspirin at clinically relevant doses increases nitric oxide (NO) formation. METHODS: seventy primary prevention patients with metabolic syndrome were randomly assigned to 81 mg, 162.5 mg, 325 mg, 650 mg, or 1300 mg aspirin daily for 12 weeks to test changes in heme oxygenase (HO-1), a downstream target of NO formation and asymmetrical dimethylarginine (ADMA), a competitive inhibitor of NO synthase. FINDINGS: for HO-1, the mean was 29.37 nanograms per milliliter at baseline and 57.45 at 12 weeks giving a mean ratio (MR) of 1.96 (P < .001) and 95% confidence interval (CI) from 1.91 to 2.00. There was no effect modification by dose or gender (P = .341). For ADMA, the mean was 1.70 micromoles per liter at baseline and 0.81 at 12 weeks, giving an MR of 0.48 (P < .001) and CI from 0.46 to 0.49. There was no effect modification by dose but a possible difference by gender (P = .055). INTERPRETATION: in high-risk primary prevention patients, aspirin significantly increases markers of NO formation. All doses produce similar increases in HO-1 and decreases in ADMA. The antiplatelet properties of aspirin to irreversibly inhibit platelet dependent cyclooxygenase are sufficient to explain benefits in patients with occlusive vascular diseases. Nonetheless, these data contribute to the formulation of the hypothesis that aspirin has additional beneficial effects mediated through NO formation. Further research, including direct randomized comparisons on atherosclerosis using noninvasive techniques as well as on occlusive vascular disease events, is necessary to test whether this hypothesis has clinical or public health relevance.


Subject(s)
Aspirin/therapeutic use , Cardiovascular Diseases/prevention & control , Myocardial Infarction/prevention & control , Nitric Oxide/metabolism , Platelet Aggregation Inhibitors/therapeutic use , Primary Prevention , Adult , Aged , Aged, 80 and over , Aspirin/administration & dosage , Aspirin/adverse effects , Biomarkers , Cardiovascular Diseases/drug therapy , Female , Humans , Male , Middle Aged , Myocardial Infarction/drug therapy , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/adverse effects , Risk Factors , Treatment Outcome , Vascular Diseases/drug therapy , Vascular Diseases/prevention & control
6.
Clin Ther ; 30(2): 249-59, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18343263

ABSTRACT

BACKGROUND: Clopidogrel, aspirin (ASA), and the fixed-dose combination of extended-release dipyridamole and ASA (ER-DP+ASA) are widely used in post-stroke regimens. OBJECTIVE: This study compared serial changes in multiple biomarkers of platelet activation with ER-DP+ASA and clopidogrel with or without ASA in patients with type 2 diabetes mellitus and a history of transient ischemic attack (TIA). METHODS: This was a randomized, single-blind pilot study conducted at an outpatient center in the United States. Eligible patients were aged 40 years and had a diagnosis of type 2 diabetes and a history of TIA. Patients were allocated to receive ER-DP+ASA 200/25 mg BID, clopidogrel 75 mg/d, or clopidogrel 75 mg/d plus ASA 81 mg/d. Multiple platelet bio-markers were assessed at baseline, day 15, and day 30 using aggregometry, cartridge-based platelet function analyzers, and flow cytometry. The primary end point was the change in platelet receptor expression after 30 days of therapy. Compliance and tolerability were monitored by measuring plasma dipyridamole levels and recording all episodes of headache and vomiting. RESULTS: The study enrolled 60 consecutive patients (20 per treatment arm), all of whom completed the study. There were no significant differences between treatment arms, although the ER-DP+ASA group had a numerically greater mean age, higher proportion of men, and a greater prevalence of vascular disease and smoking compared with the other groups. There were no deaths or serious adverse events during the study, including symptoms attributable to cerebral ischemia, worsening of diabetes, or cerebral or systemic bleeding. Three patients in the ER-DP+ASA group and 1 in the clopidogrel plus ASA group reported headache during the first several days of therapy; 1 patient in the clopidogrel monotherapy group experienced transitory nausea and vomiting. ER-DP+ASA was associated with a significantly delayed (day 30) reduction in expression of glyco-protein (GP) Ilb/IIIa activity (P = 0.02), platelet-endothelial cell adhesion molecule 1 (PECAM-1) (P = 0.03), GP Ib (P = 0.001), vitronectin (P = 0.001), P-selectin (P = 0.001), lysosome-associated membrane protein 1 (P = 0.001), and cluster of differentiation 40 ligand (P = 0.01), as well as significant inhibition of the intact (P = 0.01) and cleaved (P = 0.01) epitopes of protease-activated receptor 1. Clopidogrel monotherapy, on the other hand, was associated with significant inhibition of adenosine diphosphate-induced platelet aggregation (P = 0.001), closure-time prolongation (P = 0.01), and reduction in measurements on the rapid platelet function assay-ASA at day 15 (P = 0.001). Expression of PECAM-1 (P = 0.03) and GP IIb/IIIa activity (P = 0.01) was reduced at day 15 in clopidogrel-treated patients. The addition of ASA to clopidogrel was associated with significant inhibition of collagen-induced platelet aggregation (P = 0.001) and diminished formation of platelet-monocyte microparticles at days 15 (P = 0.02) and 30 (P = 0.03). CONCLUSIONS: In these patients with type 2 diabetes and a history of TIA, patterns of platelet inhibition differed significantly according to whether treatment was with ER-DP+ASA or clopidogrel with or without ASA. The antiplatelet activity of clopidogrel was more potent and occurred earlier (15 days), whereas ER-DP+ASA was associated with moderate downregulation of multiple activation-dependent platelet receptors that occurred later (30 days).


Subject(s)
Aspirin/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Dipyridamole/therapeutic use , Ischemic Attack, Transient/drug therapy , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Ticlopidine/analogs & derivatives , Adult , Aged , Aspirin/adverse effects , Aspirin, Dipyridamole Drug Combination , Biomarkers/blood , Clopidogrel , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Dipyridamole/adverse effects , Drug Combinations , Drug Therapy, Combination , Female , Humans , Ischemic Attack, Transient/blood , Ischemic Attack, Transient/complications , Male , Middle Aged , Pilot Projects , Platelet Aggregation Inhibitors/adverse effects , Platelet Function Tests , Single-Blind Method , Ticlopidine/adverse effects , Ticlopidine/therapeutic use , Time Factors , Treatment Outcome
7.
Am Heart J ; 155(1): 93.e1-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18082496

ABSTRACT

BACKGROUND: Clopidogrel is widely used in diabetic patients after vascular events; however, the ability of this thienopyridine to yield additional antiplatelet protection on top of aspirin has never been explored in a controlled study with comprehensive assessment of platelet activity. The objective of this study was to compare the antiplatelet profiles of clopidogrel + aspirin in combination (C + ASA) versus aspirin alone (ASA) in patients with type 2 diabetes mellitus. METHODS: Seventy patients with documented diabetes already treated with antecedent aspirin were randomly assigned to receive C + ASA or ASA in the PLUTO-Diabetes trial. Platelet studies included adenosine diphosphate-, collagen-, and arachidonic acid-induced aggregometry; PFA-100 (Dade-Behring, Miami, FL) and Ultegra (Accumetrics, San Diego, CA) analyzers; and expression of 6 major receptors by flow cytometry at baseline and at day 30 after randomization. RESULTS: There were no differences in the baseline clinical and platelet characteristics between the C + ASA and ASA groups, or subsequent significant changes in platelet biomarkers in the ASA group, except for diminished collagen-induced aggregation (P = .02). In contrast, when compared with the ASA group, therapy with C + ASA resulted in significant inhibition of platelet activity assessed by adenosine diphosphate aggregation (P = .0001); closure time prolongation (P = .0003) and reduction of platelet activation units with Ultegra (P = .0001); and expression of platelet/endothelial cell adhesion molecule 1 (P = .002), glycoprotein IIb/IIIa antigen (P = .0002), and activity (P = .0001). CONCLUSION: Treatment with C + ASA for 1 month provides significantly greater inhibition of platelet activity than ASA alone in diabetic patients in this small randomized trial. However, despite dual antiplatelet regimen, diabetic patients exhibit high residual activity of some platelet biomarkers, including unaffected protease-activated receptor 1 receptor expression.


Subject(s)
Aspirin/administration & dosage , Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/drug therapy , Myocardial Ischemia/prevention & control , Platelet Activation/drug effects , Ticlopidine/analogs & derivatives , Adult , Analysis of Variance , Biomarkers/blood , Blood Chemical Analysis , Clopidogrel , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diabetic Angiopathies/diagnosis , Diabetic Angiopathies/mortality , Drug Therapy, Combination , Female , Flow Cytometry , Humans , Male , Middle Aged , Pilot Projects , Platelet Aggregation/drug effects , Platelet Function Tests , Probability , Prognosis , Receptor, PAR-2/blood , Reference Values , Risk Assessment , Survival Rate , Ticlopidine/administration & dosage , Treatment Outcome
8.
Neuropsychopharmacology ; 32(11): 2369-74, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17356575

ABSTRACT

There is an increasing body of evidence suggesting that selective serotonin reuptake inhibitors exhibit clinical benefit beyond treating depression, by simultaneously inhibiting platelet activity. We recently demonstrated that escitalopram (ESC), but not its major metabolites, inhibits multiple platelet biomarkers in healthy volunteers. Considering that the metabolic syndrome represents one of the major risk factors for vascular disease, we here determined how ESC affects platelet activity in such patients. We assessed the in vitro effects of preincubation with escalating (50-200 nM/l) concentrations of ESC on platelet aggregation, expression of major surface receptors by flow cytometry, and quantitatively by platelet function analyzers. Blood samples were obtained from 20 aspirin-naïve patients with documented metabolic syndrome. Pretreatment of blood samples with medium (150 nM/l), or high (200 nM/l) doses of ESC resulted in a significant inhibition of platelet aggregation induced by ADP (p=0.007) and by collagen (p=0.004). Surface platelet expression of GPIb (CD42, p=0.03), LAMP-3 (CD63, p=0.04), and GP37 (CD165, p=0.03) was decreased in the ESC-pretreated samples. Closure time by the PFA-100 analyzer was prolonged after the 200 nM/l dose (p=0.02), indicating platelet inhibition under high shear conditions. On the other hand, the lowest tested concentration of ESC (50 nM/l) did not affect platelet activity in these patients. The in vitro antiplatelet characteristics of ESC in patients with the metabolic syndrome are similar to those in healthy volunteers. However, higher ESC doses are required to induce equally potent platelet inhibition. These data justify prospective ex vivo studies with the highest therapeutic dose to determine the potential clinical advantage of ESC in high-risk patients with vascular disease.


Subject(s)
Citalopram/pharmacology , Metabolic Diseases/blood , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Adult , Aged , Aged, 80 and over , Analysis of Variance , Biomarkers/metabolism , Dose-Response Relationship, Drug , Female , Flow Cytometry/methods , Humans , In Vitro Techniques , Male , Middle Aged , Platelet Function Tests/methods , Prospective Studies
9.
Thromb Res ; 119(2): 175-81, 2007.
Article in English | MEDLINE | ID: mdl-16546241

ABSTRACT

INTRODUCTION: Ongoing search for the optimal dosing regimens, and valid concerns that some GPIIb/IIIa inhibitors may cause rebound platelet activation are limiting the use of these agents in patients with acute vascular events. MATERIALS AND METHODS: We assessed the in vitro effects of preincubation with escalating (12.5-200 ng/mL) concentrations of tirofiban on platelet biomarkers in 20 diabetic patients. Platelet activity was assessed by ADP-, and collagen-induced conventional plasma aggregometry, and by whole blood flow cytometry measuring expression of PECAM-1, GPIb, GP IIb/IIIa antigen and activity, vitronectin, P-selectin, LAMP-1, GP 37, LAMP-3, activated and intact PAR-1 thrombin receptors, GPIV, and platelet-monocyte formation. All patients were treated with aspirin (at least 81 mg daily for 1 month); other antiplatelet agents were not allowed. RESULTS: Significant decrease of ADP-induced platelet aggregation was observed starting at the low 12.5 ng/mL concentration (p=0.0001), with total inhibition occurring at 50 ng/mL of tirofiban dose. Inhibition of collagen-induced platelet aggregability requires 25 ng/ml of tirofiban (p=0.002), and was complete at 100 ng/mL. Dose-dependent blockade of GP IIb/IIIa activity was observed with tirofiban concentrations over 50 ng/mL (p=0.003). Other receptors were unaffected even with the high doses of tirofiban (100-200 ng/mL). CONCLUSION: Tirofiban completely inhibits ADP- and, with the higher dose, collagen-induced platelet aggregation. Higher loading dose of tirofiban used in the ongoing TENACITY trial (100 ng/mL) may be superior with regard to clinical outcomes to the regimens used in PRISM-PLUS (25 ng/mL), or TARGET (50 ng/mL). Selective inhibition of GPIIb/IIIa activity, and lack of alternative platelet activation beyond the GP IIb/IIIa blockade may represent the therapeutic advantage of tirofiban over other agents.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/drug therapy , Platelet Aggregation/drug effects , Receptors, Cell Surface/drug effects , Tyrosine/analogs & derivatives , Aged , Blood Platelets/chemistry , Blood Platelets/cytology , Blood Platelets/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Female , Flow Cytometry , Humans , Male , Middle Aged , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Receptors, Cell Surface/analysis , Tirofiban , Tyrosine/administration & dosage , Tyrosine/pharmacology
10.
Thromb Res ; 119(3): 277-84, 2007.
Article in English | MEDLINE | ID: mdl-16563469

ABSTRACT

INTRODUCTION: Clopidogrel inhibits platelet P2Y12 ADP receptors, while ADP, as an inductor of aggregation, stimulates both P2Y12 and P2Y1 platelet receptors. Despite a clinical loading dose routine with clopidogrel, some patients still experience coronary stent thrombosis suggesting persistent platelet activation. The VerifyNow-P2Y12 is a rapid assay that test platelet activity over 3 min and uses of the combination of ADP and prostaglandin E1 (PGE1) to directly measure the effects of clopidogrel on the P2Y12 receptor. ADP is used to maximally activate the platelets by binding to the P2Y1 and P2Y12 platelet receptors, while PGE1 is used to suppress the ADP-induced P2Y1-mediated increase in intracellular calcium levels. OBJECTIVE: The VERIfy Thrombosis risk ASsessment (VERITAS) was a prospective study designed to measure platelet response to clopidogrel therapy in subjects with multiple risk factors or history of vascular disease using this novel point-of-care assay. METHODS: 166 participants were enrolled in 4 participating sites. Data from 147 participants were analyzed after exclusion of 19 patients due to protocol violations. Platelets were assessed twice at baseline (before clopidogrel) and at 24 h post-loading 450 mg (110 participants) or 7 days after chronic clopidogrel treatment (75 mg/day) (37 patients). All participants received aspirin 81-325 mg for at least 2 days before the study enrollment. Results from the VerifyNow-P2Y12 assay are reported in P2Y12 reaction units (PRU). RESULTS: Clopidogrel therapy resulted in a mean 64.0+/-25.3% PRU reduction. No participant reached PRU inhibition below 10% of baseline. Distribution of PRU values for the VerifyNow-P2Y12 assay shows a separation from baseline to post-clopidogrel assay values with some overlap due to high inter-individual variations in response. CONCLUSIONS: VerifyNow-P2Y12 is a reliable, fast and sensitive device suitable for monitoring of platelet inhibition during clopidogrel therapy.


Subject(s)
Monitoring, Physiologic , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation/drug effects , Thrombosis/prevention & control , Ticlopidine/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adult , Aged , Alprostadil/pharmacology , Blood Platelets/metabolism , Clopidogrel , Coronary Disease/complications , Coronary Disease/metabolism , Coronary Disease/therapy , Female , Humans , Male , Middle Aged , Monitoring, Physiologic/instrumentation , Monitoring, Physiologic/methods , Platelet Function Tests/instrumentation , Platelet Function Tests/methods , Prospective Studies , Purinergic P2 Receptor Agonists , Purinergic P2 Receptor Antagonists , Receptors, Purinergic P2Y1 , Stents , Thrombosis/etiology , Thrombosis/metabolism , Ticlopidine/administration & dosage
11.
J Clin Psychopharmacol ; 26(2): 172-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16633147

ABSTRACT

BACKGROUND: Clinical depression has been identified as an independent risk factor for increased mortality during follow-up in patients suffered from acute coronary events, whereas increased platelet activity has been proposed as one of the mechanisms for this association. Some evidence suggests that selective serotonin reuptake inhibitors and/or their metabolites exhibit potent antiplatelet properties. METHODS: We assessed the in vitro effects of preincubation with escalating (50-200 nmol/L) concentrations of escitalopram (ESC) S-desmethyl-citalopram (S-DCT), and S-di-desmethyl-citalopram, (S-DCT) on platelet aggregation through the expression of major surface receptors using flow cytometry and quantitatively using platelet function analyzers in 20 healthy volunteers. RESULTS: Pretreatment of blood samples with ESC with ESC resulted in a significant inhibition of platelet aggregation induced by ADP (P = 0.0001) and by collagen with the highest dose (P = 0.001). Surface platelet expressions of glycoprotein Ib (CD42) (P = 0.04), lysosome associated membrane protein-3 (CD63) (P = 0.02), and GP37 (CD165) (P = 0.03) was decreased in the ESC-pretreated samples. Closure time by the Platelet Function Analyzer-100 analyzer was prolonged for the 200 nmol/L dose (P = 0.02), indicating platelet inhibition under high shear conditions. Two major metabolites of ESC, namely S-DCT and S-DDCT, did not affect platelet activity. CONCLUSION: Escitalopram, but not its metabolites, exhibited selective inhibition of human platelet properties. The direct antiplatelet effect of ESC requires further prospective or ex vivo testing to determine the possible clinical advantage of this finding.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Blood Platelets/drug effects , Citalopram/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation , Adult , Antigens, Neoplasm/metabolism , Blood Platelets/immunology , Citalopram/metabolism , Female , Flow Cytometry , Humans , In Vitro Techniques , Lysosomal Membrane Proteins/metabolism , Male , Neoplasm Proteins/metabolism , Platelet Function Tests , Platelet Glycoprotein GPIb-IX Complex/metabolism
12.
Am J Cardiol ; 97(9): 1332-6, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16635606

ABSTRACT

We investigated whether, in primary prevention patients with metabolic syndrome, statins affect the platelet protease-activated receptor-1 (PAR-1) thrombin receptor by performing serial measurements of its activity and the antigen expression level by flow cytometry before and during treatment. Recent data from randomized trials of statins are compatible with the possibility of clinically relevant pleiotropic effects. The use of statins is associated with a reduced thrombosis burden and diminished platelet activity, as shown in animal models and in vitro studies. Seventy patients with the metabolic syndrome who were not taking antiplatelet agents were assigned consecutively at starting doses at the discretion of the responsible clinician to 1 of 6 statins (atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin, or simvastatin) or to a no-statin group for 6 weeks. Platelet expression of intact (SPAN12 antibody) and cleaved (WEDE15) PAR-1 thrombin receptors were assessed by flow cytometry at baseline and at weeks 4 and 6 of treatment. At baseline, no difference was found in receptor expression. However, after 4 weeks of treatment, all statins had significantly inhibited (46% to 55%) the activated epitope of PAR-1 expression. After 6 weeks, inhibition remained, despite a slight rebound (22% to 37%). Also, a delayed pattern of inhibition of the intact PAR-1 receptor epitope was found. In conclusion, all statins inhibited the activity and antigen level of the platelet PAR-1 thrombin receptor, which has a major role in regulating platelet activity and thrombin formation. These observational data offer a plausible mechanism for the recently demonstrated pleiotropic effects of statins that may contribute to early clinical benefit.


Subject(s)
Blood Platelets/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Metabolic Syndrome/drug therapy , Receptor, PAR-1/antagonists & inhibitors , Blood Platelets/metabolism , Female , Flow Cytometry , Humans , Male , Middle Aged , Prospective Studies , Receptor, PAR-1/metabolism
13.
Am Heart J ; 151(1): 92-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16368298

ABSTRACT

BACKGROUND: Previous in vitro studies have suggested that valsartan produces significant inhibition of human platelets, probably targeting angiotensin I platelet receptors. To test whether valsartan inhibits platelet activity in mild to moderate hypertensives we conducted the randomized Valsartan Inhibits Platelets (VIP) trial. METHODS AND RESULTS: Seventy-five patients with mild to moderate hypertension were randomized to valsartan 80 (n = 25), valsartan 160 (n = 29), or valsartan 320 mg/d (n = 21) for 9 weeks. Platelet function was assessed at baseline, week 5, and week 9 by aggregometry, flow cytometry, and cartridge-based analyzer. Independently of dose and duration, valsartan provided early sustained significant inhibition of adenosine diphosphate-induced platelet aggregation, decreased shear-induced activation measured with PFA-100 analyzer, and diminished expression of GP IIb/IIIa activity measured by PAC-1 antibody, GPIb (CD42b), vitronectin receptor (CD51/61), P-selectin (CD62p), lysosome-associated membrane protein (CD107a), and CD40-ligand (CD154). The antiplatelet properties of valsartan were more profound in patients with diabetes (n = 28) when compared with the nondiabetic group (n = 47). In subgroup analyses of patients with diabetes there appeared to be stronger inhibition of the platelet receptors, a significant decrease of adenosine diphosphate- and collagen-induced platelet aggregation, and more profound inhibition of GP IIb/IIIa activity. CONCLUSIONS: In the randomized VIP trial, valsartan produced sustained inhibition of platelet aggregation and major platelet receptors. The antiplatelet properties of valsartan were not dose or time dependent. In subgroup analyses patients with diabetes with mild to moderate hypertension tended to have greater platelet inhibition, a finding which, if confirmed in future studies suggests possible additional advantages for using valsartan in this high-risk population.


Subject(s)
Antihypertensive Agents/administration & dosage , Blood Platelets/drug effects , Hypertension/blood , Hypertension/drug therapy , Tetrazoles/administration & dosage , Valine/analogs & derivatives , Aged , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Severity of Illness Index , Valine/administration & dosage , Valsartan
14.
Cerebrovasc Dis ; 21(1-2): 98-105, 2006.
Article in English | MEDLINE | ID: mdl-16340184

ABSTRACT

BACKGROUND: Although controversial, the phenomenon of aspirin resistance (AR) has been correlated in some small studies with poor clinical outcomes in patients with coronary artery disease. Even less is known regarding the role of AR in the post stroke population. The reason for and the underlying mechanism of AR is unknown. We hypothesized that excessive formation of thrombin on the platelet surface may contribute to this phenomenon and assessed how dipyridamole affects multiple platelet and thrombin generation biomarkers in AR patients after ischemic stroke. METHODS: Whole blood samples from 20 post stroke AR patients were pretreated with dipyridamole, simulating the therapeutic range, and then incubated for 45 min at 37 degrees C. Platelet characteristics were assessed by aggregometry, cartridge-based analyzer, and receptor expression by flow cytometry. Markers of thrombin generation were measured in the autologous plasma by ELISA. RESULTS: Pretreatment of blood with dipyridamole resulted in 22-26% diminished expression of intact PAR-1 receptor (p=0.021 and p=0.024) and 28-31% decrease of annexin V binding (p=0.031 and p=0.02) after incubation with 2 microg/ml and 4 microg/ml of dipyridamole, respectively. Platelet aggregation and thrombin generation markers were not affected in vitro by dipyridamole. CONCLUSIONS: Dipyridamole may be capable of overcoming increased prothrombinase complex formation and be in part able to compensate for AR in patients with moderate carotid stenosis. This phenomenon may explain the clinical advantages of Aggrenox, known to reduce ischemic events in post stroke patients as proven in clinical trials, though an additional antithrombotic benefit beyond the platelet inhibition by aspirin alone.


Subject(s)
Aspirin , Dipyridamole/pharmacology , Drug Resistance , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Stroke/physiopathology , Aged , Cell Culture Techniques , Dipyridamole/administration & dosage , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Platelet Aggregation Inhibitors/administration & dosage , Receptor, PAR-1/drug effects , Receptors, Peptide/drug effects
15.
Stroke ; 36(10): 2289-92, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16151033

ABSTRACT

BACKGROUND AND PURPOSE: Clopidogrel is widely used in patients after recent ischemic stroke; however, its ability to yield additional antiplatelet protection on top of aspirin has never been explored in a controlled study. To determine whether clopidogrel with aspirin (C+ASA) will produce more potent platelet inhibition than aspirin alone (ASA) in patients after ischemic stroke, we conducted the Plavix Use for Treatment of Stroke trial. METHODS: Seventy patients after ischemic stroke were randomly assigned to C+ASA or ASA groups. Platelet studies included aggregometry; cartridge-based analyzers; expression of PECAM-1, P-selectin, GP IIb/IIIa (antigen and activity), vitronectin receptor, and formation of platelet-leukocyte microparticles by flow cytometry. Platelet tests were performed at baseline and after 30 days after randomization. RESULTS: There were no deaths, hospitalizations, or serious adverse events. There were no differences in the baseline platelet characteristics between C+ASA and ASA groups, or significant changes in platelet parameters in the ASA group, except diminished collagen-induced aggregation (P=0.001). In contrast, therapy with C+ASA resulted in a significant inhibition of platelet activity assessed by ADP- (P=0.00001) and collagen-induced (P=0.02) aggregation; closure time prolongation (P=0.03), and reduction of platelet activation units with Ultegra (P=0.00001); expression of PECAM-1 (P=0.01), and GP IIb/IIIa activity with PAC-1 (P=0.02) when compared with ASA group. Therapy with C+ASA also resulted in the reduced formation of platelet-leukocyte microparticles (P=0.02). CONCLUSIONS: Treatment with C+ASA for 1 month provides significantly greater inhibition of platelet activity than ASA alone in patients after recent ischemic stroke in the frame of the small randomized trial.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Aspirin/administration & dosage , Brain Ischemia/drug therapy , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Stroke/drug therapy , Ticlopidine/analogs & derivatives , Aged , Algorithms , Biomarkers/metabolism , Blood Platelets/drug effects , Brain Ischemia/pathology , Clopidogrel , Drug Therapy, Combination , Female , Flow Cytometry , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Function Tests , Stroke/pathology , Ticlopidine/administration & dosage , Time Factors , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...