Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Clin Pharm Ther ; 34(5): 575-83, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19744013

RESUMO

BACKGROUND AND OBJECTIVE: Prasugrel is a thienopyridine antiplatelet agent under investigation for the prevention of atherothrombotic events in patients with acute coronary syndrome who undergo percutaneous coronary intervention. Patients with chronic liver disease are among those in the target population for prasugrel. As hepatic enzymes play a key role in formation of prasugrel's active metabolite, hepatic impairment could affect the safety and/or efficacy of prasugrel in such patients. METHODS: This was a parallel-design, open-label, multiple dose study of 30 subjects, 10 with moderate hepatic impairment (Child-Pugh Class B) and 20 with normal hepatic function. Prasugrel was administered orally as a 60-mg loading dose (LD) and daily 10-mg maintenance doses (MDs) for 5 days. Pharmacokinetic parameters (AUC(0-t), C(max) and t(max)) and maximal platelet aggregation (MPA) by light transmission aggregometry were assessed after the LD and final MD. RESULTS AND DISCUSSION: Exposure to prasugrel's active metabolite was comparable between healthy subjects and those with moderate hepatic impairment. Point estimates for the ratios of geometric least square means for AUC(0-t) and C(max) after the LD and last MD ranged from 0.91 to 1.14. MPA to 20 microm ADP was similar between subjects with moderate hepatic impairment and healthy subjects for both the LD and MD. Prasugrel was well tolerated by all subjects, and adverse events were mild in severity. CONCLUSION: Moderate hepatic impairment appears to have no effect on exposure to prasugrel's active metabolite. Furthermore, MPA results suggest that moderate hepatic impairment has little or no effect on platelet aggregation relative to healthy controls. Overall, these results suggest that a dose adjustment would not be required in moderately hepatically impaired patients taking prasugrel.


Assuntos
Hepatopatias/metabolismo , Piperazinas/farmacocinética , Inibidores da Agregação Plaquetária/farmacocinética , Tiofenos/farmacocinética , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Piperazinas/efeitos adversos , Piperazinas/farmacologia , Cloridrato de Prasugrel , Tiofenos/efeitos adversos , Tiofenos/farmacologia
2.
J Clin Pharm Ther ; 34(5): 585-94, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19744014

RESUMO

OBJECTIVE: The pharmacokinetic (PK) and pharmacodynamic (PD) responses to prasugrel were compared in three studies of healthy subjects vs. those with moderate or end-stage renal impairment. METHODS: Two of the three protocols were parallel-design, open-label, single dose (60-mg prasugrel) studies in subjects with end-stage renal disease (ESRD; n = 12) or moderate renal impairment (n = 10) and matched healthy subjects with normal renal function (n = 10). The third protocol was an open-label, single-dose escalation (5, 10, 30 and 60 mg prasugrel) study in subjects with ESRD (n = 16) and matched healthy subjects with normal renal function (n = 16). Plasma concentrations of prasugrel's active metabolite were determined and pharmacokinetic parameter estimates were derived. Maximum platelet aggregation (MPA) was measured by light transmission aggregometry using 20 mum adenosine diphosphate as agonist. RESULTS: Across all studies, prasugrel's C(max) and AUC(0-t) were 51% and 42% lower in subjects with ESRD than in healthy subjects. AUC(0-t) did not differ between healthy subjects and subjects with moderate renal impairment. The magnitude of change and time-course profiles of MPA was similar for healthy subjects compared with subjects with moderate renal impairment and those with ESRD. Prasugrel was well-tolerated in all subjects. CONCLUSION: There was no difference in pharmacokinetics or PD responses between subjects with moderate renal impairment and healthy subjects. Despite significantly lower exposure to prasugrel's active metabolite in subjects with ESRD, MPA did not differ between healthy subjects and those with ESRD.


Assuntos
Nefropatias/metabolismo , Falência Renal Crônica/metabolismo , Piperazinas/farmacocinética , Inibidores da Agregação Plaquetária/farmacocinética , Tiofenos/farmacocinética , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Piperazinas/efeitos adversos , Piperazinas/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Cloridrato de Prasugrel , Ligação Proteica , Tiofenos/efeitos adversos , Tiofenos/farmacologia
3.
Clin Pharmacol Ther ; 81(5): 735-41, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17361128

RESUMO

Prasugrel and clopidogrel inhibit platelet aggregation through active metabolite formation. Prasugrel's active metabolite (R-138727) is formed primarily by cytochrome P450 (CYP) 3A and CYP2B6, with roles for CYP2C9 and CYP2C19. Clopidogrel's activation involves two sequential steps by CYP3A, CYP1A2, CYP2C9, CYP2C19, and/or CYP2B6. In a randomized crossover study, healthy subjects received a loading dose (LD) of prasugrel (60 mg) or clopidogrel (300 mg), followed by five daily maintenance doses (MDs) (15 and 75 mg, respectively) with or without the potent CYP3A inhibitor ketoconazole (400 mg/day). Subjects had a 2-week washout between periods. Ketoconazole decreased R-138727 and clopidogrel active metabolite Cmax (maximum plasma concentration) 34-61% after prasugrel and clopidogrel dosing. Ketoconazole did not affect R-138727 exposure or prasugrel's inhibition of platelet aggregation (IPA). Ketoconazole decreased clopidogrel's active metabolite AUC0-24 (area under the concentration-time curve to 24 h postdose) 22% (LD) to 29% (MD) and reduced IPA 28% (LD) to 33% (MD). We conclude that CYP3A4 and CYP3A5 inhibition by ketoconazole affects formation of clopidogrel's but not prasugrel's active metabolite. The decreased formation of clopidogrel's active metabolite is associated with reduced IPA.


Assuntos
Inibidores do Citocromo P-450 CYP3A , Inibidores Enzimáticos/farmacologia , Cetoconazol/farmacologia , Piperazinas/farmacocinética , Inibidores da Agregação Plaquetária/farmacocinética , Tiofenos/farmacocinética , Ticlopidina/análogos & derivados , Adulto , Área Sob a Curva , Clopidogrel , Estudos Cross-Over , Método Duplo-Cego , Interações Medicamentosas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Piperazinas/administração & dosagem , Piperazinas/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/administração & dosagem , Inibidores da Agregação Plaquetária/farmacologia , Cloridrato de Prasugrel , Tiofenos/administração & dosagem , Tiofenos/farmacologia , Ticlopidina/administração & dosagem , Ticlopidina/farmacocinética , Ticlopidina/farmacologia
4.
J Thromb Haemost ; 5(12): 2429-36, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17900275

RESUMO

BACKGROUND: Thienopyridines are metabolized to active metabolites that irreversibly inhibit the platelet P2Y(12) adenosine diphosphate receptor. The pharmacodynamic response to clopidogrel is more variable than the response to prasugrel, but the reasons for variation in response to clopidogrel are not well characterized. OBJECTIVE: To determine the relationship between genetic variation in cytochrome P450 (CYP) isoenzymes and the pharmacokinetic/pharmacodynamic response to prasugrel and clopidogrel. METHODS: Genotyping was performed for CYP1A2, CYP2B6, CYP2C19, CYP2C9, CYP3A4 and CYP3A5 on samples from healthy subjects participating in studies evaluating pharmacokinetic and pharmacodynamic responses to prasugrel (60 mg, n = 71) or clopidogrel (300 mg, n = 74). RESULTS: In subjects receiving clopidogrel, the presence of the CYP2C19*2 loss of function variant was significantly associated with lower exposure to clopidogrel active metabolite, as measured by the area under the concentration curve (AUC(0-24); P = 0.004) and maximal plasma concentration (C(max); P = 0.020), lower inhibition of platelet aggregation at 4 h (P = 0.003) and poor-responder status (P = 0.030). Similarly, CYP2C9 loss of function variants were significantly associated with lower AUC(0-24) (P = 0.043), lower C(max) (P = 0.006), lower IPA (P = 0.046) and poor-responder status (P = 0.024). For prasugrel, there was no relationship observed between CYP2C19 or CYP2C9 loss of function genotypes and exposure to the active metabolite of prasugrel or pharmacodynamic response. CONCLUSIONS: The common loss of function polymorphisms of CYP2C19 and CYP2C9 are associated with decreased exposure to the active metabolite of clopidogrel but not prasugrel. Decreased exposure to its active metabolite is associated with a diminished pharmacodynamic response to clopidogrel.


Assuntos
Hidrocarboneto de Aril Hidroxilases/metabolismo , Plaquetas/efeitos dos fármacos , Oxigenases de Função Mista/metabolismo , Piperazinas/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Polimorfismo Genético , Pró-Fármacos/farmacologia , Tiofenos/farmacologia , Ticlopidina/análogos & derivados , Adulto , Área Sob a Curva , Hidrocarboneto de Aril Hidroxilases/genética , Plaquetas/metabolismo , Ensaios Clínicos como Assunto , Clopidogrel , Estudos Cross-Over , Citocromo P-450 CYP2C19 , Citocromo P-450 CYP2C9 , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Oxigenases de Função Mista/genética , Fenótipo , Piperazinas/sangue , Piperazinas/farmacocinética , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/sangue , Inibidores da Agregação Plaquetária/farmacocinética , Cloridrato de Prasugrel , Pró-Fármacos/farmacocinética , Antagonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y12 , Valores de Referência , Projetos de Pesquisa , Estudos Retrospectivos , Tiofenos/sangue , Tiofenos/farmacocinética , Ticlopidina/sangue , Ticlopidina/farmacocinética , Ticlopidina/farmacologia
5.
Thromb Res ; 59(2): 279-93, 1990 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-2146766

RESUMO

To elucidate interactions responsible for inhibition of aggregation of platelets in platelet-rich plasma (PRP) harvested from whole blood preincubated with t-PA, experiments were performed with PRP and washed platelets under diverse conditions of preincubation. Both ADP and collagen induced aggregation were inhibited in PRP unless aprotinin had been added to the preincubated whole blood concomitantly with t-PA. However, in washed platelets prepared after the same exposure aggregation was intact. When washed platelets were supplemented with fibrinogen degradation products (FDPs) in concentrations simulating those in whole blood preincubated with t-PA, aggregation induced with either ADP or collagen was inhibited. Thus, the inhibition in PRP depended on generation of FDPs by activated plasminogen. The functional integrity of surface glycoprotein (GP) IIb/IIIa receptors in washed platelets was documented by autoradiography after SDS-PAGE of surface labeled GPs and by fibrinogen binding despite preincubation of the whole blood or washed platelets themselves with t-PA and plasminogen as long as exogenous calcium (greater than or equal to 0.1 microM) was present. In contrast, when calcium was absent, the platelet GP IIb/IIIa receptor was rendered susceptible to degradation by plasmin, and aggregation was inhibited by preincubation at 37 degrees C even if aprotinin was present when aggregation was being assayed. These observations reconcile disparate results in the literature from studies in vivo and in vitro by demonstrating that inhibition of aggregation of platelets in PRP and in whole blood reflects indirect effects of plasminogen activation rather than direct effects of t-PA or plasmin on the platelets themselves.


Assuntos
Inibidores da Agregação Plaquetária , Ativador de Plasminogênio Tecidual/farmacologia , Cálcio/farmacologia , Fibrinogênio/metabolismo , Fibrinolisina/farmacologia , Humanos , Radioisótopos do Iodo , Plasminogênio/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/efeitos dos fármacos
6.
Thromb Haemost ; 112(3): 589-97, 2014 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-25008027

RESUMO

CYP2C19 genotype has been shown to impact response to clopidogrel 75-mg but not prasugrel 10-mg. Here, we assessed effects of CYP2C19 metaboliser status on pharmacokinetics (PK) and pharmacodynamic (PD) responses to prasugrel 5-mg and 10-mg and clopidogrel 75-mg using data from two PK/PD studies in stable coronary artery disease (CAD) patients (GENERATIONS and FEATHER). Active metabolite concentrations (area under the curve, AUC[0-tlast]), maximum platelet aggregation (MPA) measured by light transmission aggregometry, vasodilator-stimulated phosphoprotein platelet reactivity index, and VerifyNow P2Y12-platelet reaction units (VN-PRU) were analysed by CYP2C19-predicted phenotype (extensive metaboliser [EM; N=154], *2-*8 non-carriers, vs reduced metaboliser [RM; N=41],*2-*8 carriers/*17 non-carriers). AUC(0-tlast) was unaffected by metaboliser status for prasugrel 5-mg and 10-mg (geometric mean EM/RM ratios 1.00, 95% confidence interval [CI]: 0.86,1.17, p>0.99; and 0.97, 95% CI:0.85,1.12, p=0.71, respectively), but was lower among RMs receiving clopidogrel 75-mg (1.37, 95% CI:1.14,1.65, p<0.001). Platelet reactivity was not significantly affected by CYP2C19 metaboliser status for prasugrel 5-mg, or for prasugrel 10-mg by MPA and VN-PRU, but for clopidogrel 75-mg was significantly higher in reduced metabolisers (all measures p<0.01). Prasugrel 10-mg showed greater antiplatelet effects vs clopidogrel 75-mg (all comparisons p<0.001). Prasugrel 5-mg showed greater antiplatelet effects vs clopidogrel 75-mg in RMs (all p<0.001), and comparable effects in EMs (all p≥0.37). In contrast to clopidogrel, prasugrel active metabolite PK was not influenced by CYP2C19 genotype. Antiplatelet effect for prasugrel 10-mg was greater irrespective of metaboliser status and for prasugrel 5-mg was greater for RMs and comparable for EMs as compared to clopidogrel 75-mg.


Assuntos
Doença da Artéria Coronariana/tratamento farmacológico , Citocromo P-450 CYP2C19/genética , Piperazinas/farmacocinética , Inibidores da Agregação Plaquetária/farmacocinética , Tiofenos/farmacocinética , Ticlopidina/análogos & derivados , Idoso , Protocolos Clínicos , Clopidogrel , Doença da Artéria Coronariana/genética , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Piperazinas/administração & dosagem , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/genética , Inibidores da Agregação Plaquetária/administração & dosagem , Polimorfismo Genético , Cloridrato de Prasugrel , Tiofenos/administração & dosagem , Ticlopidina/administração & dosagem , Ticlopidina/farmacocinética , Resultado do Tratamento
7.
Curr Med Res Opin ; 25(8): 1821-9, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19530977

RESUMO

OBJECTIVE: Prasugrel is a thienopyridine antiplatelet agent for the prevention of atherothrombotic events in patients with acute coronary syndrome undergoing percutaneous coronary intervention. Since cytochrome P450 enzymes CYP3A4 and CYP2B6 play a major role in prasugrel's active metabolite formation, the effect of potent CYP induction by rifampin on the pharmacokinetics of prasugrel and on the pharmacodynamic response to prasugrel was evaluated in healthy male subjects. RESEARCH DESIGN AND METHODS: This was an open-label, two-period, fixed-sequence study conducted at a single clinical research center. In the first treatment period, subjects received prasugrel as an oral 60-mg loading dose (LD) on the first day followed by ten oral, 10-mg daily maintenance doses. After a 2-week washout period, subjects received oral rifampin alone (600 mg once daily) for 8 days, followed by coadministration of oral rifampin with prasugrel, given as a 60-mg LD on the first day followed by five daily 10-mg MDs. Blood collection for pharmacokinetic and pharmacodynamic analyses occurred after the LD and fifth MD of prasugrel in both periods. CLINICAL TRIAL SYNOPSIS: clinicalstudyresults.org ID #8976 RESULTS: Rifampin coadministration (600 mg daily) did not affect exposure to prasugrel's active metabolite (R-138727). However, at 2 and 4 h after the prasugrel loading dose (60 mg), rifampicin coadministration was associated with a 6-9 percentage point decrease (p < 0.01) in the magnitude of platelet inhibition; similarly, a 5-17 percentage point decrease (p < 0.05) was observed with rifampin coadministration during the prasugrel maintenance dose (10 mg) period. Post hoc in vitro experiments demonstrated a dose-dependent R-138727-rifampin interaction at the P2Y(12) level unrelated to enzyme induction. A limitation of this study is that while results of the in vitro post hoc study indicate a pharmacodynamic interaction with rifampin, the mechanism underlying this interaction has not been elucidated. CONCLUSIONS: Dose adjustment should not be necessary when prasugrel is administered with CYP inducers since formation of prasugrel's active metabolite is not affected by potent enzyme induction with rifampin.


Assuntos
Antibióticos Antituberculose/farmacologia , Piperazinas/farmacologia , Piperazinas/farmacocinética , Rifampina/farmacologia , Tiofenos/farmacologia , Tiofenos/farmacocinética , Síndrome Coronariana Aguda , Adolescente , Adulto , Antibióticos Antituberculose/administração & dosagem , Interações Medicamentosas , Humanos , Masculino , Pessoa de Meia-Idade , Piperazinas/administração & dosagem , Cloridrato de Prasugrel , Rifampina/administração & dosagem , Tiofenos/administração & dosagem , Adulto Jovem
8.
J Thromb Haemost ; 6(2): 359-65, 2008 02.
Artigo em Inglês | MEDLINE | ID: mdl-18021304

RESUMO

BACKGROUND: Prasugrel is a novel antiplatelet prodrug of the same thienopyridine class as clopidogrel and ticlopidine. Metabolism of prasugrel generates the active metabolite R-138727, an antagonist of the platelet P2Y(12) adenosine diphosphate (ADP) receptor, leading to inhibition of ADP-mediated platelet activation and aggregation. ADP also enhances the platelet response to collagen, and these two agonists contribute to the generation of platelet procoagulant activity. We therefore examined whether R-138727 inhibits ADP- and collagen-triggered platelet procoagulant activities. METHODS AND RESULTS: As shown by whole blood flow cytometry, R-138727 inhibited surface phosphatidylserine expression on ADP plus collagen-stimulated platelets and tissue factor (TF) expression on ADP-, collagen-, and ADP plus collagen-stimulated monocyte-platelet aggregates. R-138727 reduced monocyte-platelet aggregate formation, thereby further inhibiting TF expression. ADP, collagen, and ADP plus collagen accelerated the kinetics of thrombin generation in recalcified whole blood and R-138727 significantly inhibited this acceleration. Clot strength in a modified thromboelastograph system was also inhibited by R-138727 (IC50 0.7 +/- 0.1 microM). CONCLUSIONS: In addition to its previously known inhibitory effects on platelet activation and aggregation, the active metabolite of prasugrel, R-138727, inhibits platelet procoagulant activity in whole blood (as determined by phosphatidylserine expression on platelets and TF expression on monocyte-platelet aggregates), resulting in the functional consequences of delayed thrombin generation and impaired clot development.


Assuntos
Fatores de Coagulação Sanguínea/antagonistas & inibidores , Plaquetas/efeitos dos fármacos , Retração do Coágulo/efeitos dos fármacos , Piperazinas/farmacologia , Piperazinas/farmacocinética , Ativação Plaquetária/efeitos dos fármacos , Pró-Fármacos/farmacocinética , Antagonistas do Receptor Purinérgico P2 , Tiofenos/farmacocinética , Difosfato de Adenosina/farmacologia , Anexina A5/metabolismo , Biotransformação , Colágeno/farmacologia , Humanos , Monócitos/metabolismo , Fosfatidilserinas/sangue , Cloridrato de Prasugrel , Receptores Purinérgicos P2Y12 , Tromboelastografia , Trombina/biossíntese , Tromboplastina/biossíntese
10.
Blood ; 76(8): 1546-57, 1990 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-2169924

RESUMO

The effects of activation of plasminogen by streptokinase and tissue-type-plasminogen activator on platelet activation and the membrane glycoproteins (GPs) that mediate platelet adhesion and aggregation are not yet fully defined. To clarify effects on platelets during activation of plasminogen in vitro, we used monoclonal antibodies (MoAbs), flow cytometry, and platelets surface-labeled with 125I to characterize changes in receptors for fibrinogen (GPIIb-IIIa), von Willebrand factor (GPIb), and collagen (GPIa-IIa). Activation of plasminogen in plasma with pharmacologic concentrations of plasminogen activators did not degrade GPIIb-IIIa or GPIb, and caused only a modest decrease in GPIa. In washed platelets GPIIb-IIIa was extensively degraded by plasmin at 37 degrees C in the absence of exogenous Ca2+, conditions that destabilize the IIb-IIIa complex. Degradation of GPIb in washed platelets displayed a similar although less-marked dependence on temperature and the absence of Ca2+. The binding of activation-specific MoAbs did not increase during activation of plasminogen in plasma. We conclude that during pharmacologic fibrinolysis, reported inhibition of platelet function in plasma is not due to degradation of platelet-adhesive receptors. In addition, platelet activation observed during thrombolytic therapy does not appear to be a direct consequence of plasminogen activation.


Assuntos
Cálcio/farmacologia , Fibrinolisina/metabolismo , Adesividade Plaquetária/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores de Superfície Celular/metabolismo , Anticorpos Monoclonais , Ativação Enzimática/efeitos dos fármacos , Citometria de Fluxo , Humanos , Radioisótopos do Iodo , Plasminogênio/metabolismo , Receptores de Colágeno , Estreptoquinase/farmacologia , Temperatura , Ativador de Plasminogênio Tecidual/farmacologia
11.
Blood ; 81(7): 1778-86, 1993 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-8461464

RESUMO

Divalent cation-dependent platelet adhesion to fibronectin (FN) is mediated by the integrin receptors alpha 5 beta 1 (GP Ic-IIa) and alpha IIb beta 3 (GP IIb-IIIa), which recognize the RGD (Arg-Gly-Asp) sequence in the cell-binding domain. However, FN can also support divalent cation-independent platelet adhesion. To determine which domain of FN mediates divalent cation-independent adhesion, proteolysis with thermolysin and affinity chromatography were used to isolate the cell-binding, gelatin-binding, and heparin-binding domains of FN. Unactivated and thrombin-activated platelets adhered to intact FN and the 45-Kd gelatin-binding domain in the presence of either Ca2+ or EDTA. Platelet spreading was mediated only by the 105-Kd cell-binding domain and required divalent cations. The heparin-binding domains did not support platelet adhesion. Reduction of intrachain disulfide bonds or removal of carbohydrate side chains on the gelatin-binding domain did not alter the ability to support platelet adhesion. Divalent cation-independent adhesion to the 45-Kd gelatin-binding domain was not inhibited by RGDS (Arg-Gly-Asp-Ser) synthetic peptides or monoclonal antibodies (MoAbs) directed against known platelet receptors. We conclude that platelets can adhere but not spread on the gelatin-binding domain of FN by a novel divalent cation-independent mechanism.


Assuntos
Cálcio/sangue , Proteínas de Transporte/sangue , Fibronectinas/sangue , Gelatina/sangue , Adesividade Plaquetária , Sequência de Aminoácidos , Cátions Bivalentes/sangue , Comunicação Celular , Ácido Edético/farmacologia , Humanos , Dados de Sequência Molecular , Fragmentos de Peptídeos/sangue , Adesividade Plaquetária/efeitos dos fármacos , Ligação Proteica
12.
Circulation ; 84(4): 1552-60, 1991 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1833087

RESUMO

BACKGROUND: Platelet activation occurs in vivo during pharmacologic thrombolysis and may contribute to recurrent thrombosis. Plasmin does not directly activate platelets except at high concentrations; thus, the mechanisms for platelet activation during thrombolysis remain undefined. Increases in thrombin activity also occur in patients treated with fibrinolytic agents and may contribute to activation of platelets. We have shown that one mechanism for increased thrombin activity is activation of the coagulation system by plasmin. METHODS AND RESULTS: In the present study we sought to determine whether activation of platelets in response to pharmacologic activation of plasminogen in plasma is due primarily to plasmin or mediated by increased thrombin activity. Platelet-rich citrated plasma (PRP) was recalcified and incubated with 1,000 IU/ml of streptokinase or 1.0 caseinolytic units/ml of plasmin. Concentrations of fibrinopeptide A, a marker of thrombin activity, increased markedly over 10 minutes in plasma incubated with streptokinase or plasmin, but not in PRP incubated without plasminogen activator. Platelet activation characterized by the secretion of 14C-serotonin occurred within 2-4 minutes after thrombin activity increased. In stirred recalcified PRP, platelet aggregation was accelerated from 3.6 +/- 0.5 to 2.5 +/- 0.3 minutes (p less than 0.01) when incubated with 1,000 IU/ml of streptokinase. Leupeptin and aprotinin, inhibitors of plasmin activity, markedly attenuated platelet activation in response to pharmacologic activation of plasminogen. However, inhibition of thrombin with heparin, hirudin, or D-Phe-D-Pro-L-Arg-chloromethylketone was more effective in inhibiting the acceleration of platelet activation induced by plasminogen activation, despite the elaboration of plasmin activity. CONCLUSIONS: Activation of platelets during coronary thrombolysis may be due in part to increased procoagulant activity induced by plasminogen activation as well as other factors that promote platelet activation in vivo.


Assuntos
Fibrinolisina/fisiologia , Plasminogênio/efeitos dos fármacos , Ativação Plaquetária/fisiologia , Estreptoquinase/farmacologia , Trombina/fisiologia , Fibrinopeptídeo A/metabolismo , Humanos , Técnicas In Vitro , Plasminogênio/metabolismo , Agregação Plaquetária/fisiologia , Terapia Trombolítica , Fatores de Tempo
13.
Catheter Cardiovasc Interv ; 46(3): 357-62, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10348141

RESUMO

Hydrophilic coatings improve the handling characteristics of angioplasty equipment, but until recently this technology has not been available for intra-aortic balloon (IAB) catheters. To determine whether a new hydrophilic coating is associated with a reduction in IAB-related complications, we prospectively evaluated 188 patients undergoing insertion of this new IAB catheter. Complications related to IAB insertion were recorded and compared to data from 682 patients previously treated with an uncoated IAB catheter of the same shaft diameter and balloon size. By multivariate analysis the hydrophilic-coated IAB catheter was associated with a 72% reduction in ischemic vascular complications (relative risk 0.28, 95% CI 0.08-0.96, P= 0.04). There were also trends toward reductions in IAB rupture and the need for vascular surgical repair for bleeding or ischemia. While further investigation is warranted, hydrophilic coatings that reduce the coefficient of friction during IAB catheter insertion may also reduce subsequent ischemic vascular complications.


Assuntos
Materiais Revestidos Biocompatíveis , Balão Intra-Aórtico , Idoso , Desenho de Equipamento , Feminino , Humanos , Balão Intra-Aórtico/efeitos adversos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Estudos Prospectivos , Fatores de Risco
14.
Cathet Cardiovasc Diagn ; 39(4): 433-7, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8958440

RESUMO

Serial sampling from the coronary sinus is an attractive technique for drawing blood samples to be used in the characterization of procoagulant activity during coronary interventions. We have developed a modified Simmons catheter for rapid cannulation of the coronary sinus from the femoral approach. The catheter design incorporates heparin bonding and distal side-holes to minimize blood sampling artifacts. Coronary sinus cannulation was performed via the femoral approach in 186 patients by use of a multipurpose catheter (n = 8), an unmodified Simmons I or II catheter (n = 64), or the modified Simmons catheter (n = 114). The coronary sinus was cannulated successfully with the modified Simmons catheter in 97% of patients; the success rate with the unmodified Simmons II catheter was 87% (P = 0.02). The modified Simmons catheter represents an improved technique for cannulation of the coronary sinus from the femoral vein.


Assuntos
Cateterismo Periférico/instrumentação , Vasos Coronários , Veia Femoral , Heparina/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Sangue , Cateterismo Periférico/métodos , Cateterismo Periférico/estatística & dados numéricos , Distribuição de Qui-Quadrado , Desenho de Equipamento , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Manejo de Espécimes/instrumentação , Manejo de Espécimes/métodos , Manejo de Espécimes/estatística & dados numéricos
15.
Circulation ; 94(9): 2064-71, 1996 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-8901652

RESUMO

BACKGROUND: Acute thrombosis is thought to contribute to abrupt coronary occlusion during percutaneous coronary revascularization despite the administration of heparin and aspirin. This study was designed to detect the presence of heparin-resistant thrombin activity and to define its relationship to the acute ischemic complications of coronary interventions. METHODS AND RESULTS: Plasma levels of fibrinopeptide A (FPA) and prothrombin fragment 1.2 (F1.2), markers of thrombin and factor Xa activity, respectively, were measured in the coronary sinus with heparin-bonded catheters in 58 patients undergoing coronary interventions. Activated coagulation times were maintained > 300 seconds by the Hemochron method. Mean FPA levels decreased significantly, from 7.0 +/- 0.9 nmol/L before the procedure to 5.2 +/- 0.5 nmol/L after the heparin bolus and to 2.9 +/- 0.2 nmol/L after the procedure (P = .0001). In 26 patients (45%), FPA levels remained above the threshold for suppression angioplasty of thrombin activity determined during angiography in 7 patients without coronary artery disease (> 3.0 nmol/L). FPA concentrations after coronary interventions were increased in patients with intracoronary thrombus (P = .01), abrupt coronary occlusion (P = .06), postprocedural non-Q-wave myocardial infarction (P = .04), and clinically unsuccessful procedures (P = .04). F1.2 levels were relatively low before the procedures and did not change significantly. CONCLUSIONS: Heparin administration suppresses thrombin activity in most but not all patients undergoing coronary interventions. Heparin-resistant thrombin activity is associated with angiographic evidence of intracoronary thrombus and ischemic complications of coronary interventions.


Assuntos
Cateterismo Cardíaco/efeitos adversos , Heparina/farmacologia , Isquemia Miocárdica/complicações , Trombina/antagonistas & inibidores , Trombina/metabolismo , Doença Aguda , Idoso , Angiografia , Coleta de Amostras Sanguíneas , Inibidores do Fator Xa , Feminino , Fibrinopeptídeo A/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Isquemia Miocárdica/etiologia , Isquemia Miocárdica/metabolismo , Fragmentos de Peptídeos/metabolismo , Protrombina/metabolismo , Reprodutibilidade dos Testes , Trombose/tratamento farmacológico , Trombose/etiologia , Tempo de Coagulação do Sangue Total
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA