Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Thromb Haemost ; 104(2): 311-7, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20539903

RESUMO

Niacin is a natural pyridine derivative, proven to favorably modulate the blood lipid profile by increasing levels of high-density lipoprotein (HDL) cholesterol, and by reducing total cholesterol, low-density lipoprotein (LDL) cholesterol, triglycerides, and Lp (a) lipoprotein concentrations. Considering that platelet activity is important in predicting vascular outcomes, and that HDL heavily constitutes platelet cellular membranes, we sought to evaluate the effect of niacin on human platelet activity indices. The blood obtained from 30 aspirin-naïve volunteers was preincubated with escalating concentrations of niacin in vitro. Platelet tests included whole blood and plasma aggregometry, rapid cartridge-based analyser, expression of major surface receptors by flow cytometry, and plasma prostaglandins by ELISA. Preincubation of blood with niacin at 0.3, 1.0 and 3.0 mM resulted in significant inhibition of maximal adenosine diphosphate (ADP)- (p=0.03), and collagen-induced platelet aggregation (p=0.01), and reduced activity by VerifyNow (p=0.007) bedside analyser. Surface platelet PAR-1 (MoAb WEDE-15; p=0.04), and vitronectin (CD51/CD61; p=0.02) receptors were up-regulated. Niacin was associated with a two- to three-fold increase of thromboxane B2, prostaglandins D2, and E2. Formation of platelet-monocyte microparticles (CD14+CD151), and expression of PECAM-1 (CD31), thrombospondin (CD36), GP IIb/IIIa (CD41a) antigen, and activity with MoAb PAC-1, GPIb (CD42b), P-selectin (CD62p), LAMP-3 (CD63), LAMP-1 (CD107a), CD40-ligand (CD154), GP37 (CD165), were not affected by niacin, suggesting no effect on prostacyclin release. In conclusion, niacin in vitro affects platelet activity by mildly inhibiting aggregation, and stimulating significant prostaglandin release, with mostly intact major platelet receptor expression. The effect of niacin is unique, differs from other known antiplatelet agents, and suggests potential opportunities for therapeutic combination, particularly in patients with low levels of HDL-C. These preliminary data, while intriguing, require confirmation in subjects receiving orally dosed extended-release niacin in order to determine whether these findings are clinically relevant.


Assuntos
Plaquetas/efeitos dos fármacos , Niacina/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Difosfato de Adenosina , Adulto , Baltimore , Biomarcadores/sangue , Plaquetas/metabolismo , Colágeno , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Hipolipemiantes/farmacologia , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Razão de Chances , Agregação Plaquetária/efeitos dos fármacos , Testes de Função Plaquetária , Glicoproteínas da Membrana de Plaquetas/metabolismo , Prostaglandinas/sangue
2.
Expert Rev Cardiovasc Ther ; 8(4): 503-7, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20397824

RESUMO

Anticoagulation is an important therapeutic strategy to treat and prevent recurrent thrombotic events in patients after acute myocardial infarction and ischemic stroke. However, available anticoagulants have numerous shortcomings including hemorrhage risks, limited clinical application including heparin-induced thrombocytopenia and modest therapeutic benefit. Each component of the coagulation cascade is a potential target for new drug developments. Representing an attractive new therapeutic class, aptamers are nucleic acid ligand with a high affinity and specificity for proteins involved in the clot growth. Potential antidote control of aptamer activity offers an important advance in anticoagulant safety. Aptamers are a promising class of compounds, both for target validation and therapy. As designer drugs, they exhibit high specificity, high affinity and modifiable bioavailability. The ability to generate inhibitors with such properties against a variety of target proteins will be valuable as the human genome and proteome are deciphered. This article summarizes current knowledge of aptamers developed against coagulation proteins, focusing on the biology, pharmacology, preclinical and early clinical experience with this novel class of anticoagulants.


Assuntos
Anticoagulantes/uso terapêutico , Aptâmeros de Nucleotídeos/uso terapêutico , Doenças Vasculares/tratamento farmacológico , Anticoagulantes/efeitos adversos , Anticoagulantes/farmacologia , Antídotos/uso terapêutico , Aptâmeros de Nucleotídeos/efeitos adversos , Aptâmeros de Nucleotídeos/farmacologia , Disponibilidade Biológica , Coagulação Sanguínea/efeitos dos fármacos , Ensaios Clínicos como Assunto , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Humanos , Doenças Vasculares/fisiopatologia
3.
Am J Ther ; 17(2): 125-32, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20305399

RESUMO

Astaxanthine is a polar carotenoid metabolite derived from a proprietary prodrug, Xancor, which aligns parallel with the membrane phospholipids exhibiting potent antioxidant, anti-inflammatory, and cell protective properties, although the precise mechanism of action is unknown. This prodrug is currently under development for hepatic, neurologic, and vascular disease indications. Considering established links between heart disease and stroke with platelets, coagulation cascade, and fibrinolysis, the aim of the study was to assess the effect of asthaxantine on human biomarkers of hemostasis. The rationale was to test a hypothesis that the drug may diminish activation of hemostasis, making it a potentially attractive addition to treat patients with vascular disease. In vitro effects of whole blood preincubation with escalating concentrations of asthaxantine (0.3 microM, 1 microM, 3 microM, 10 microM, 30 microM, and 100 microM) were assessed from 12 aspirin-naïve and eight aspirin-treated volunteers with multiple risk factors for vascular disease. A total of 25 biomarkers were measured, of which 12 were related to platelet function, 10 to coagulation, and three to fibrinolysis. Platelet aggregation induced by ADP, collagen, and arachidonic acid and expression of CD31, CD41, GP IIb/IIIa, CD51/61, P-selectin, CD63, CD107a, CD151+CD14, and CD154 were not affected. Coagulation indices such as aPTT, prothrombin time, thrombin time, fibrinogen, antithrombin III (antigen and activity), Protein C, Protein S (free and activity), and von Willebrand factor remained unchanged after incubation with astaxanthine. Fibrinolytic activity biomarkers such as plasminogen, D-dimer, and FDP were also not affected after in vitro pretreatment of blood samples with astaxanthine. In the projected subclinical (less than 1 microM), therapeutic (3 microM to 30 microM), and supratherapeutic concentration (100 microM), astaxanthine in vitro does not affect platelet, coagulation, or fibrinolytic indices in either aspirin-naïve or aspirin-treated subjects. These results are important for the assessment of the safety profile, but remain to be confirmed preclinically, in vivo, and ultimately in the clinic.


Assuntos
Antioxidantes/farmacologia , Aspirina/farmacologia , Xantofilas/farmacologia , Adulto , Antioxidantes/administração & dosagem , Antioxidantes/efeitos adversos , Biomarcadores/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Interações Medicamentosas , Feminino , Fibrinólise/efeitos dos fármacos , Hemostasia/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Pró-Fármacos , Fatores de Risco , Método Simples-Cego , Xantofilas/administração & dosagem , Xantofilas/efeitos adversos , Xantofilas/metabolismo
4.
J Thromb Thrombolysis ; 27(1): 48-56, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18188512

RESUMO

BACKGROUND AND PURPOSE: Major depression is an independent risk factor for increased morbidity and mortality in patients with coronary artery disease (CAD). Increased platelet activity and vascular endothelial dysfunction are possible pathways through which depression may increase cardiovascular risk. Citalopram exhibits strong selective inhibition of human platelet activation, but little is known about its effects on vascular endothelium. We assessed whether treatment of depressed CAD patients with citalopram alters platelet/endothelial biomarkers. The study was performed within the framework of the CREATE trial. METHODS: We assessed the effect of citalopram on P-selectin, beta-thromboglobulin (betaTG), soluble intercellular cell adhesion molecule-1 (sICAM-1), and total nitric oxide (tNO). Plasma samples were obtained at baseline and week 12 from subjects randomized to citalopram 20-40 mg daily (n = 36), or placebo (n = 21). Anticoagulants, aspirin, and clopidogrel were permitted. RESULTS: Treatment with citalopram was associated with greater increase in tNO over 12 weeks compared to placebo (P = 0.005). There were no differences for the other biomarkers such as P-selectin (P = 0.70), betaTG (P = 0.46) and ICAM (P = 0.59). CONCLUSION: Treatment with citalopram for 12 weeks in depressed CAD patients is associated with enhanced production of nitric oxide despite the co-administration of commonly prescribed anti-platelet regimens including aspirin and clopidogrel. Clinical implications of these findings are unclear, but improved endothelial function is implied by the increased NO production, suggesting that citalopram may be of particular benefit for patients with comorbid depression and vascular disease including CAD, stroke, peripheral artery disease, and diabetes.


Assuntos
Antidepressivos/farmacologia , Plaquetas/efeitos dos fármacos , Citalopram/farmacologia , Doença das Coronárias/sangue , Transtorno Depressivo Maior/sangue , Endotélio Vascular/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Adulto , Idoso , Antidepressivos/uso terapêutico , Aspirina/administração & dosagem , Aspirina/farmacologia , Aspirina/uso terapêutico , Biomarcadores , Plaquetas/metabolismo , Citalopram/uso terapêutico , Clopidogrel , Comorbidade , Doença das Coronárias/complicações , Transtorno Depressivo Maior/complicações , Transtorno Depressivo Maior/tratamento farmacológico , Transtorno Depressivo Maior/terapia , Interações Medicamentosas , Endotélio Vascular/metabolismo , Feminino , Humanos , Molécula 1 de Adesão Intercelular/sangue , Masculino , Pessoa de Meia-Idade , Óxido Nítrico/biossíntese , Selectina-P/análise , Selectina-P/sangue , Inibidores da Agregação Plaquetária/administração & dosagem , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Psicoterapia , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Ticlopidina/administração & dosagem , Ticlopidina/análogos & derivados , Ticlopidina/farmacologia , Ticlopidina/uso terapêutico , Adulto Jovem , beta-Tromboglobulina/análise
5.
J Thromb Thrombolysis ; 27(4): 438-46, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18521540

RESUMO

BACKGROUND: Oxidative stress play an important role triggering platelet/endothelial activation. AGI-1067 is a novel, phenolic antioxidant, and vascular protectant which dose-dependently inhibits PEA biomarkers in vitro. Whether treatment with AGI-1067 alters platelets in vivo is not known. We serially assessed release of established PEA biomarkers in subjects treated with AGI-1067 versus placebo in the frame of Assessment of Lipoprotein Profiles Randomized Trial (ALPS). METHODS: Healthy subjects (18-65 years) with multiple risk factors for coronary artery disease were randomized 1:1 to receive 300 mg AGI-1067 (n = 112) or matching placebo (n = 117) daily for 12 weeks. Anticoagulants, aspirin, NSAIDS, and COX inhibitors were not permitted in this study. Plasma samples were collected at baseline, and at week 12 after randomization. Platelet factor 4 (PF4), beta-thromboglobulin (betaTG), P-selectin, thromboxane (TxB2), and prostacyclin (6-keto-PGF1a) were measured by ELISA. RESULTS: Treatment with AGI-1067 was associated with a highly significant reduction of TxB2 release (P < 0.0001) when compared to the placebo. There were no differences in PF4, betaTG, P-selectin, and 6-keto-PGF1a between and within groups. AGI-1067 also inhibits TxB2 release from calcium ionophore (A23187)-stimulated human platelets with the IC50 equals 1 microM; but does not interfere with 6-keto-PGF1alpha release in either A23187-, or TXA2-stimulated human aortic endothelial cells. CONCLUSION: AGI-1067 selectively reduces TxB(2 )production from stimulated platelets, and diminishes plasma TxB2 levels in ALPS participants. These data support earlier in vitro, and pilot ex vivo experiments suggesting antiplatelet properties of AGI-1067. Lack of 6-keto-PGF1a down regulation may represent an attractive advantage of AGI-1067 over currently available antiplatelet regimens.


Assuntos
Doença da Artéria Coronariana/prevenção & controle , Lipoproteínas/sangue , Probucol/análogos & derivados , Tromboxanos/antagonistas & inibidores , Tromboxanos/sangue , Adolescente , Adulto , Idoso , Biomarcadores/sangue , Células Cultivadas , Doença da Artéria Coronariana/sangue , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Probucol/uso terapêutico , Fatores de Risco , Doenças Vasculares/sangue , Doenças Vasculares/prevenção & controle , Adulto Jovem
6.
Fundam Clin Pharmacol ; 22(3): 315-21, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18485150

RESUMO

A number of antiplatelet drugs, principally aspirin alone or in combination, have been evaluated in randomized trials of survivors of prior occlusive vascular disease events or of individuals at high risk because of multiple cardiovascular risk factors. In this meta-analysis we compare single and dual antiplatelet regimens to quantitate the risks of bleeding. Data from randomized trials published in English in 1988-2006 were retrieved from MEDLINE, OVID, and CARDIOSOURCE. Inclusion criteria were clinical follow-up for at least 1 month and the presence of data on bleeding complications. Information was compiled on sample size, antiplatelet agents tested, patient characteristics as well as major, minor, fatal and intracranial bleeding. Using these criteria, we identified 18 randomized trials, which included 129,314 patients. For each endpoint, relative risk (RR) and 95% confidence intervals (CI) were calculated. Dual antiplatelet therapy is associated with a significantly increased risk of major (RR 1.47, CI = 1.36-1.60) and minor bleeding events (RR 1.56, CI = 1.47-1.66) compared to single agent therapy. Although based on small numbers, there were no significant differences in fatal (RR 1.10, CI = 0.87-1.40) or intracranial (RR 1.07, CI = 0.85-1.35) bleedings although the CIs are wide to make definite assessments. Patients treated with dual antiplatelet therapy have an approximately 40-50% increase in risks of major and minor bleeding compared to those receiving single agent therapy during the duration of the scrutinized trials. The magnitude of this excess risk is not so remote from the approximately 60% increase observed in trials comparing single antiplatelet agents to placebo. This excess risk should be considered when choosing the optimal antiplatelet strategy for long-term treatment of patients with prior occlusive vascular events or those at high risk of developing occlusive vascular disease.


Assuntos
Hemorragia/induzido quimicamente , Inibidores da Agregação Plaquetária/efeitos adversos , Arteriopatias Oclusivas/prevenção & controle , Quimioterapia Combinada , Humanos , Inibidores da Agregação Plaquetária/uso terapêutico , Medição de Risco
7.
Clin Ther ; 30(2): 249-59, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18343263

RESUMO

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).


Assuntos
Aspirina/uso terapêutico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Dipiridamol/uso terapêutico , Ataque Isquêmico Transitório/tratamento farmacológico , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/uso terapêutico , Ticlopidina/análogos & derivados , Adulto , Idoso , Aspirina/efeitos adversos , Combinação Aspirina e Dipiridamol , Biomarcadores/sangue , Clopidogrel , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Dipiridamol/efeitos adversos , Combinação de Medicamentos , Quimioterapia Combinada , Feminino , Humanos , Ataque Isquêmico Transitório/sangue , Ataque Isquêmico Transitório/complicações , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Inibidores da Agregação Plaquetária/efeitos adversos , Testes de Função Plaquetária , Método Simples-Cego , Ticlopidina/efeitos adversos , Ticlopidina/uso terapêutico , Fatores de Tempo , Resultado do Tratamento
8.
Thromb Haemost ; 99(1): 116-20, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18217142

RESUMO

Randomized trials showed greater stroke prevention with extended release dipyridamole in combination with low dose aspirin than with either aspirin or dipyridamole alone. However, most studies with this formulation (Aggrenox) were carried out in Europe and North America. Considering potential inter-racial differences in drug response, we conducted a small randomized study in healthy Japanese volunteers to compare antiplatelet regimens with regard to the changes in the platelet biomarkers. Thirty healthy volunteers (18-40 years old, 15 male and 15 female) of Japanese descent were randomized to Aggrenox (n = 17) or aspirin 81 mg (n = 13 volunteers) for 30 days. Platelet function was assessed at baseline, and on days 15, and 30 by conventional aggregometry, whole blood flow cytometry, and cartridge-based analyzer. Both Aggrenox and aspirin provided sustained platelet inhibition at Day 15 and Day 30. Therapy with Aggrenox, however, was associated with more prominent and significant inhibition of collagen-induced aggregation (p = 0.08, Day 15), as well as prolongation of the closure time (p = 0.001, Day 30); diminished expression of platelet endothelial cell adhesion molecule-1 (PECAM-1) (p = 0.02, Day 30), glycoprotein IIb (GPIIb) antigen (p = 0.001 and 0.024 for Day 15 and Day 30), and GPIIb/IIIa activity by PAC-1 antibody (p = 0.014 and 0.03), CD62 (P-selectin) (p = 0.03 for Day 15 and Day 30), as well as inhibition of protease activated receptors (PAR-1) associated with intact WEDE-15 (p = 0.002 and 0.003) and SPAN-12 (p = 0.002 and 0.04) thrombin receptors when compared with aspirin. The magnitude and durability of platelet response after Aggrenox in healthy Japanese is similar to those effects observed in Caucasians and African-Americans. A larger study to assess drug efficacy and safety in the Japanese post-stroke patients is warranted.


Assuntos
Aspirina/farmacologia , Plaquetas/efeitos dos fármacos , Dipiridamol/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Adulto , Antígenos CD/sangue , Povo Asiático , Aspirina/administração & dosagem , Combinação Aspirina e Dipiridamol , Biomarcadores/sangue , Plaquetas/imunologia , Plaquetas/metabolismo , Preparações de Ação Retardada , Dipiridamol/administração & dosagem , Combinação de Medicamentos , Feminino , Citometria de Fluxo , Humanos , Masculino , Inibidores da Agregação Plaquetária/administração & dosagem , Testes de Função Plaquetária/métodos , Glicoproteínas da Membrana de Plaquetas/metabolismo , Valores de Referência , Projetos de Pesquisa , Fatores de Tempo
9.
Am Heart J ; 155(1): 93.e1-7, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18082496

RESUMO

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.


Assuntos
Aspirina/administração & dosagem , Diabetes Mellitus Tipo 2/tratamento farmacológico , Angiopatias Diabéticas/tratamento farmacológico , Isquemia Miocárdica/prevenção & controle , Ativação Plaquetária/efeitos dos fármacos , Ticlopidina/análogos & derivados , Adulto , Análise de Variância , Biomarcadores/sangue , Análise Química do Sangue , Clopidogrel , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/diagnóstico , Angiopatias Diabéticas/diagnóstico , Angiopatias Diabéticas/mortalidade , Quimioterapia Combinada , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Agregação Plaquetária/efeitos dos fármacos , Testes de Função Plaquetária , Probabilidade , Prognóstico , Receptor PAR-2/sangue , Valores de Referência , Medição de Risco , Taxa de Sobrevida , Ticlopidina/administração & dosagem , Resultado do Tratamento
10.
J Thromb Thrombolysis ; 25(2): 207-13, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17530178

RESUMO

BACKGROUND: The metabolic syndrome is a matter of ongoing debate with regard to its existence, classification, clinical meaningfulness, and associated risks for vessel occlusion. Considering that persistent platelet activation is a cornerstone for the development of acute vascular events, and that patients with type 2 diabetes consistently exhibit high platelet activity, these characteristics may be critical for distinguishing and triageing specific features of metabolic syndrome among established risk factors for vascular disease. METHODS: We assessed the platelet activity by conventional aggregation, expression of major surface receptors by flow cytometry, and quantitatively by rapid bedside analyzers in 20 aspirin-naïve patients with documented metabolic syndrome, and compared these with 20 untreated subjects with multiple cardiovascular risk factors. RESULTS: Closure time by the PFA-100 analyzer was significantly (P = 0.002) shorter in patients with metabolic syndrome indicating platelet inhibition under high shear conditions. Ultegra analyzer readings revealed increased fibrinogen binding (P = 0.0003) what in combination with the increased expression of PAC-1 (P = 0.32) strongly suggest activation of platelet glycoprotein IIb/IIIa receptor. Surface expression of CD107a (P = 0.014), and SPAN-12 (P = 0.003) were also higher in patients with metabolic syndrome. In contrast, platelet aggregation induced by collagen or ADP, CD31, CD41, CD42b, CD51/61, CD62p, CD63, CD154, CD165, so as formation of platelet-monocyte aggregates, PAR-1 thrombin receptor, and thrombospondin did not differ between groups. CONCLUSION: Patients with metabolic syndrome exhibited a higher degree of platelet activation than subjects with conventional risk factors for vascular disease. Conceptually, applying adequate antiplatelet strategies may reduce the risk of acute thrombotic events in these patients. Further prospective studies exploring this notion are encouraged.


Assuntos
Síndrome Metabólica/fisiopatologia , Ativação Plaquetária/fisiologia , Adulto , Idoso , Biomarcadores/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Testes de Função Plaquetária , Estudos Prospectivos , Fatores de Risco , Doenças Vasculares/fisiopatologia
12.
Expert Rev Cardiovasc Ther ; 5(4): 635-41, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17605642

RESUMO

Oxidation-sensitive signals play an important role in platelet activation. AGI-1067 is a novel, phenolic, intra- and extracellular antioxidant that inhibits the expression of a number of proinflammatory genes involved in atherosclerosis. AGI-1067 is the metabolically stable monosuccinic acid ester of probucol, and a potent phenolic antioxidant representing a novel class of orally bioavailable compounds termed vascular protectants. AGI-1067 exhibits antioxidant activity equipotent to probucol. In addition, animal studies have demonstrated dual pharmacological activities of AGI-1067: the ability to block the expression of oxidation-sensitive inflammatory genes including genes that code for vascular cell adhesion molecule-1 and monocyte chemotactic protein-1. Importantly, AGI-1067 also exhibits mild antiplatelet properties inhibiting surface expression of various key platelet receptors, the formation of platelet monocyte microparticles and PAR-1 thrombin receptors. AGI-1067 is currently being tested in the late trials, and if proven to improve clinical outcomes (ARISE trial), the drug will ultimately be used in patients with different manifestations of atherosclerosis and atherothrombosis.


Assuntos
Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Aterosclerose/tratamento farmacológico , Probucol/análogos & derivados , Animais , Aterosclerose/complicações , Aterosclerose/fisiopatologia , Aterosclerose/prevenção & controle , Plaquetas/efeitos dos fármacos , Doença da Artéria Coronariana/tratamento farmacológico , Doença da Artéria Coronariana/prevenção & controle , Reestenose Coronária/prevenção & controle , Citocinas/metabolismo , Progressão da Doença , Humanos , Hipolipemiantes/farmacologia , Hipolipemiantes/uso terapêutico , Estresse Oxidativo/fisiologia , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Probucol/farmacologia , Probucol/uso terapêutico , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
13.
Clin Chem Lab Med ; 45(4): 513-20, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17439330

RESUMO

BACKGROUND: Selective serotonin reuptake inhibitors (SSRIs) in general, and citalopram/escitalopram in particular, are widely used to treat clinical depression. However, SSRI bioavailability and non-compliance represent major issues, especially in the clinical trials setting. In this context, frequent drug-level measurements for compliance monitoring would be a desirable tool to improve clinical outcomes with SSRIs. However, the liquid chromatography techniques available are expensive, requiring excessive sample preparation, and suffer from high complexity. We sought to develop a rapid method for the measurement of citalopram/escitalopram levels in human plasma by fluorimetry. METHODS: A total of 34 frozen human plasma samples were thawed at room temperature and repeatedly centrifuged in cellulose to remove aggregates, proteins and solids. Fluorescence spectra were measured in the range 270-450 nm with excitation at 240 nm on a FluoroMax 3 spectrofluorimeter. Control samples contained known concentrations of SSRIs. RESULTS: SSRI absorbance spectra were recorded in the range 230-320 nm. The shape of the spectra and the absorbance of citalopram and escitalopram were very similar, with UV maximum absorbance at 239 nm. The maximum extinction coefficient was epsilon239=15,930 M-1 cm-1 for citalopram and epsilon239=13,630 M-1 cm-1 for escitalopram. The fluorescence spectra of SSRIs are unique and are characterized by the presence of two well-defined conjugated spectra with maxima at 300 and 382 nm. CONCLUSIONS: Fluorimetry is very suitable for assessment of plasma SSRI levels. This inexpensive and efficient technique can objectively and reliably quantify drug levels in biological fluids, thereby directly determining the level of patient adherence to the prescribed drug regimen. This method will be useful in a broad spectrum of applications, from compliance/bioavailability assessments in animal and human experiments to utilization in large-scale clinical trials.


Assuntos
Citalopram/sangue , Ensaios Clínicos como Assunto , Fidelidade a Diretrizes , Inibidores Seletivos de Recaptação de Serotonina/sangue , Citalopram/farmacocinética , Humanos , Concentração de Íons de Hidrogênio , Inibidores Seletivos de Recaptação de Serotonina/farmacocinética , Espectrometria de Fluorescência , Espectrofotometria Ultravioleta
14.
Neuropsychopharmacology ; 32(11): 2369-74, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17356575

RESUMO

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.


Assuntos
Citalopram/farmacologia , Doenças Metabólicas/sangue , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Biomarcadores/metabolismo , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo/métodos , Humanos , Técnicas In Vitro , Masculino , Pessoa de Meia-Idade , Testes de Função Plaquetária/métodos , Estudos Prospectivos
15.
Am Heart J ; 153(3): 371-7, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17307414

RESUMO

BACKGROUND: Numerous reports have dichotomized responses after clopidogrel therapy using varying definitions and platelet tests in patients immediately after acute vascular events; however, no large study has assessed platelet characteristics in outpatients receiving long-term treatment for more than 30 days with the maintenance dose (75 mg/d) of clopidogrel. The aim of this study was to describe the responses of ex vivo measures of platelet aggregation and activation to long-term clopidogrel therapy in a large population of outpatients after coronary stenting or ischemic stroke. METHODS: We conducted a secondary post hoc analysis of a data set represented by presumably compliant patients after coronary stenting (n = 237) or a documented ischemic stroke (n = 122) treated with clopidogrel-and-aspirin combination antiplatelet therapy. RESULTS: The mean duration of treatment was 5.8 months (range 1-21 months). Every patient exhibited a significant inhibition of adenosine diphosphate-induced platelet aggregation (mean 52.9%, range 36%-70%) as compared with the preclopidogrel measures. Inhibition of aggregation strongly correlated with a diminished expression of PECAM-1 (platelet/endothelial cell adhesion molecule 1, r = 0.75), glycoprotein IIb/IIIa (r = 0.62), and PAR-1 (protease-activated receptor 1, r = 0.71). None of the patients developed hyporesponsiveness (reduction from the baseline <15%) or profound inhibition (residual platelet activity <10%). CONCLUSIONS: In contrast to the wide variability of responses that exists in the acute setting, long-term therapy with clopidogrel leads to consistent and much less variable platelet inhibition. Lack of nonresponse and profound inhibition with clopidogrel allow for the maintenance of a delicate balance between proven efficacy and acceptable bleeding risks for long-term secondary prevention in outpatients after acute vascular events.


Assuntos
Inibidores da Agregação Plaquetária/administração & dosagem , Ticlopidina/análogos & derivados , Doenças Vasculares/tratamento farmacológico , Idoso , Anti-Inflamatórios não Esteroides/uso terapêutico , Aspirina/uso terapêutico , Clopidogrel , Quimioterapia Combinada , Feminino , Citometria de Fluxo , Humanos , Integrina beta3/sangue , Masculino , Pessoa de Meia-Idade , Cooperação do Paciente , Molécula-1 de Adesão Celular Endotelial a Plaquetas/sangue , Testes de Função Plaquetária , Glicoproteína IIb da Membrana de Plaquetas/sangue , Fatores de Risco , Ticlopidina/administração & dosagem , Doenças Vasculares/epidemiologia , Doenças Vasculares/fisiopatologia
16.
Cardiology ; 107(4): 307-12, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17264511

RESUMO

BACKGROUND: Numerous randomized studies have shown that the combination of clopidogrel with aspirin yields better clinical outcomes than monotherapy in patients with acute vascular events. However, the impact of the aspirin dose on the antiplatelet potency of clopidogrel is unclear. We sought to compare the antiplatelet profile of aspirin 81 mg (n = 252) versus aspirin 325 mg (n = 459) before and during conventional clopidogrel loading (300 mg), and/or clopidogrel maintenance (75 mg/daily) therapy. METHODS: Secondary post hoc analysis of an existing dataset consisting of 711 patients after coronary stenting (n = 601) and ischemic stroke (n = 110) treated previously with aspirin for at least 1 month, and then with aspirin + clopidogrel for at least 7 days was performed. Platelet assessments include conventional and whole blood aggregometry, rapid cartridge-based analyzers, and expression of platelet/endothelial cell adhesion molecule-1, P-selectin, and GPIIb/IIIa activity by flow cytometry measured before and after addition of clopidogrel. RESULTS: There was a small but consistent yet non-significant trend towards more potent platelet inhibition with aspirin 325 mg compared to aspirin 81 mg for every platelet activation parameter before addition of clopidogrel. However, after loading and/or 1 week of chronic treatment with clopidogrel + aspirin, measured platelet parameters became very similar between the groups, and identical for collagen-induced aggregation and PFA-100 analyzer readings. CONCLUSIONS: Before addition of clopidogrel, aspirin 325 mg has a tendency to provide stronger platelet inhibition than aspirin 81 mg. However, when clopidogrel and aspirin are used in combination, the higher aspirin dose does not translate into superior antiplatelet action. Given that the existing body of evidence supports the comparable efficacy and, particularly, superior safety of lower versus higher doses of aspirin, aspirin 81 mg should be the dose used in combination with clopidogrel.


Assuntos
Aspirina/uso terapêutico , Inibidores da Agregação Plaquetária/uso terapêutico , Ticlopidina/análogos & derivados , Adulto , Idoso , Clopidogrel , Doença da Artéria Coronariana/terapia , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Stents , Acidente Vascular Cerebral/tratamento farmacológico , Ticlopidina/uso terapêutico
17.
Blood Coagul Fibrinolysis ; 18(2): 91-6, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17287623

RESUMO

The ability of clopidogrel to inhibit platelet function in patients with congestive heart failure (CHF) was proved by the PLUTO-CHF trial. We retrospectively analyzed platelet characteristics with respect to CHF etiology, class, and ejection fraction in patients enrolled in the PLUTO-CHF study. Twenty-five patients were divided by CHF etiology, severity, and ejection fraction. All patients received aspirin 325 mg for at least 1 month prior to screening. Platelet function studies were performed at baseline and after 30 days of therapy. There were no differences in platelet parameters dependent on clinical characteristics of CHF, except for a significant (P = 0.023) decrease in platelet/endothelial cell adhesion molecule 1 (PECAM-1) expression in the New York Heart Association class III-IV due to the higher baseline values. Therapy with clopidogrel resulted in a significant inhibition of platelet activity assessed by ADP-induced and epinephrine-induced aggregation, closure time, expression of PECAM-1, glycoprotein Ib, glycoprotein IIb/IIIa antigen, glycoprotein IIb/IIIa activity with PAC-1, CD151, and reduced formation of platelet-leukocyte conjugates when compared with baseline. Clopidogrel provides antiplatelet protection in the broad spectrum of patients with CHF independently of its etiology, severity, or myocardial contractility. This uniform platelet inhibition with clopidogrel may be an important consideration in designing future large-scale clinical trials.


Assuntos
Insuficiência Cardíaca/etiologia , Ativação Plaquetária/efeitos dos fármacos , Volume Sistólico , Ticlopidina/análogos & derivados , Idoso , Idoso de 80 Anos ou mais , Aspirina/uso terapêutico , Biomarcadores/sangue , Clopidogrel , Avaliação de Medicamentos , Feminino , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Masculino , Pessoa de Meia-Idade , Contração Miocárdica , Inibidores da Agregação Plaquetária/farmacologia , Testes de Função Plaquetária , Estudos Retrospectivos , Ticlopidina/farmacologia , Ticlopidina/uso terapêutico
18.
Thromb Res ; 119(2): 175-81, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-16546241

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Angiopatias Diabéticas/tratamento farmacológico , Agregação Plaquetária/efeitos dos fármacos , Receptores de Superfície Celular/efeitos dos fármacos , Tirosina/análogos & derivados , Idoso , Plaquetas/química , Plaquetas/citologia , Plaquetas/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Receptores de Superfície Celular/análise , Tirofibana , Tirosina/administração & dosagem , Tirosina/farmacologia
19.
Thromb Res ; 119(3): 277-84, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-16563469

RESUMO

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.


Assuntos
Monitorização Fisiológica , Inibidores da Agregação Plaquetária/administração & dosagem , Agregação Plaquetária/efeitos dos fármacos , Trombose/prevenção & controle , Ticlopidina/análogos & derivados , Difosfato de Adenosina/farmacologia , Adulto , Idoso , Alprostadil/farmacologia , Plaquetas/metabolismo , Clopidogrel , Doença das Coronárias/complicações , Doença das Coronárias/metabolismo , Doença das Coronárias/terapia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Monitorização Fisiológica/instrumentação , Monitorização Fisiológica/métodos , Testes de Função Plaquetária/instrumentação , Testes de Função Plaquetária/métodos , Estudos Prospectivos , Agonistas do Receptor Purinérgico P2 , Antagonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2Y1 , Stents , Trombose/etiologia , Trombose/metabolismo , Ticlopidina/administração & dosagem
20.
J Cardiovasc Pharmacol Ther ; 11(3): 191-6, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17056832

RESUMO

Oxidation-sensitive signals are important in platelet activation. The novel, phenolic, intracellular and extra-cellular antioxidant AGI-1067 inhibits the expression of a number of proinflammatory genes involved in atherosclerosis. The effect of AGI-1067 on human platelets was evaluated. Blood obtained from 20 aspirin-naïve volunteers with multiple risk factors for vascular disease was preincubated with escalating concentrations of AGI-1067 for the assessment of its ex vivo effects on platelet aggregation and expression of major surface receptors flow cytometry, evaluated by flow cytometry. AGI-1067 resulted in significant inhibition of a variety of activation-dependent platelet biomarkers in healthy volunteers, including adenosine diphosphate-induced platelet aggregation and decreased surface platelet expression of glycoprotein IIb/IIIa antigen, activity with PAC-1 antibody, and glycoprotein Ib (CD42b). The effect of AGI-1067 differs from other known antiplatelet agents, suggesting opportunities for therapeutic combination. These data need to be confirmed in subjects receiving orally dosed AGI-1067 to be clinically relevant.


Assuntos
Antioxidantes/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Probucol/análogos & derivados , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/efeitos dos fármacos , Doenças Vasculares/metabolismo , Doenças Vasculares/fisiopatologia , Adulto , Antígenos CD/biossíntese , Antígenos CD/efeitos dos fármacos , Baltimore , Biomarcadores/sangue , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/efeitos dos fármacos , Feminino , Citometria de Fluxo , Humanos , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Glicoproteínas da Membrana de Plaquetas/biossíntese , Glicoproteínas da Membrana de Plaquetas/efeitos dos fármacos , Probucol/farmacologia , Receptores de Superfície Celular/classificação , Fatores de Risco , Doenças Vasculares/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...