Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
N Engl J Med ; 345(25): 1809-17, 2001 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-11752357

RESUMO

BACKGROUND: Patients with arthritis and vascular disease may receive both low-dose aspirin and other nonsteroidal antiinflammatory drugs. We therefore investigated potential interactions between aspirin and commonly prescribed arthritis therapies METHODS: We administered the following combinations of drugs for six days: aspirin (81 mg every morning) two hours before ibuprofen (400 mg every morning) and the same medications in the reverse order; aspirin two hours before acetaminophen (1000 mg every morning) and the same medications in the reverse order; aspirin two hours before the cyclooxygenase-2 inhibitor rofecoxib (25 mg every morning) and the same medications in the reverse order; enteric-coated aspirin two hours before ibuprofen (400 mg three times a day); and enteric-coated aspirin two hours before delayed-release diclofenac (75 mg twice daily) RESULTS: Serum thromboxane B(2) levels (an index of cyclooxygenase-1 activity in platelets) and platelet aggregation were maximally inhibited 24 hours after the administration of aspirin on day 6 in the subjects who took aspirin before a single daily dose of any other drug, as well as in those who took rofecoxib or acetaminophen before taking aspirin. In contrast, inhibition of serum thromboxane B(2) formation and platelet aggregation by aspirin was blocked when a single daily dose of ibuprofen was given before aspirin, as well as when multiple daily doses of ibuprofen were given. The concomitant administration of rofecoxib, acetaminophen, or diclofenac did not affect the pharmacodynamics of aspirin CONCLUSIONS: The concomitant administration of ibuprofen but not rofecoxib, acetaminophen, or diclofenac antagonizes the irreversible platelet inhibition induced by aspirin. Treatment with ibuprofen in patients with increased cardiovascular risk may limit the cardioprotective effects of aspirin.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/farmacologia , Inibidores de Ciclo-Oxigenase/farmacologia , Isoenzimas/antagonistas & inibidores , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Acetaminofen/farmacologia , Adulto , Analgésicos não Narcóticos/farmacologia , Aspirina/antagonistas & inibidores , Estudos Cross-Over , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Diclofenaco/farmacologia , Dinoprostona/sangue , Interações Medicamentosas , Quimioterapia Combinada , Humanos , Ibuprofeno/farmacologia , Lactonas/farmacologia , Proteínas de Membrana , Pessoa de Meia-Idade , Prostaglandina-Endoperóxido Sintases , Sulfonas , Tromboxano B2/sangue
2.
Neurology ; 57(5 Suppl 2): S5-7, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11552047

RESUMO

The antithrombotic effect of aspirin has long been recognized, and administration of low doses (80-160 mg/day) for the prevention of ischemic events in patients with coronary artery disease (CAD) is now generally considered to be routine practice. The action of aspirin derives mostly from the selective inhibition of cyclo-oxygenases (Cox). These enzymes (Cox-1 and Cox-2) catalyze the synthesis of eicosanoids, which play an important part in platelet-vessel wall interactions. Cox-1 catalyzes the synthesis of thromboxane A2 (Tx-A2), which causes platelet activation, vasoconstriction, and smooth muscle proliferation. Tx-A2 levels are elevated in conditions associated with platelet activation, including unstable angina and cerebral ischemia. Conversely, Cox-2 controls the synthesis of prostacyclin (PGI2), a local platelet regulator with an effect opposite to that of Tx-A2. PGI2 is produced as a compensatory response to increases in Tx-A2 during ischemic events. Aspirin is a more potent inhibitor of Cox-1 than of Cox-2, unlike other non-steroidal anti-inflammatory drugs (NSAIDs), which have limited selectivity. Aspirin at low doses selectively inhibits the formation of Tx-A2 without inhibiting the basal biosynthesis of cardioprotective PGI2. Furthermore, aspirin causes complete enzyme inhibition, without the recovery of enzyme activity at trough drug levels associated with conventional NSAIDs. The effect of aspirin in the prevention of ischemic events has been well documented in many recent clinical trials involving more than 50,000 patients with CAD. It is clear from these studies that aspirin, alone or in combination with other antithrombotics, significantly reduces the incidence of cardiovascular death, stroke, and myocardial infarction.


Assuntos
Aspirina/uso terapêutico , Trombose Intracraniana/tratamento farmacológico , Inibidores da Agregação Plaquetária/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Plaquetas/citologia , Plaquetas/efeitos dos fármacos , Comunicação Celular/efeitos dos fármacos , Artérias Cerebrais/citologia , Artérias Cerebrais/efeitos dos fármacos , Humanos , Trombose Intracraniana/fisiopatologia , Acidente Vascular Cerebral/fisiopatologia
3.
Am J Cardiol ; 88(3): 236-42, 2001 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-11472700

RESUMO

Dose-finding studies and trials of interaction of oral glycoprotein IIb/IIIa antagonists with other antiplatelet agents have been limited. We hypothesized that these detailed assessments could be first performed in patients with stable coronary artery disease (CAD) and then extrapolated to the target population. To this end, we performed 2 sequential studies. The first study examined the dose-related effects on indexes of platelet and vascular function induced by the oral inhibitor RPR 109891, when given alone and in combination with aspirin, in patients (n = 100) with stable CAD. The second study (the Antagonism of the FIbrinogen Receptor after Myocardial Events trial) assessed the pharmacodynamics and safety of derived regimens in patients (n = 320) with unstable coronary syndromes. In patients with stable CAD, platelet aggregation was dose dependently inhibited by RPR 109891, and the dose-response relation was shifted to the right by the concomitant administration of aspirin (p = 0.0001). The degree of platelet inhibition induced by 3 doses of RPR 109891 (plus aspirin) was lower in patients with unstable than stable CAD. No drug-related major bleeding occurred in either study. RPR 109891 treatment was associated with acute and delayed thrombocytopenia. In conclusion, chronic treatment with an oral glycoprotein IIb/IIIa antagonist (1) induces antiplatelet effects that are potentiated by concomitant administration of aspirin, (2) may require dose adjustment in syndromes of platelet activation, (3) is associated with a low rate of clinically significant bleeding when doses inducing incomplete inhibition of platelet aggregation are used, and (4) requires frequent monitoring of platelet count unless reliable predictors of delayed thrombocytopenia become available.


Assuntos
Aspirina/administração & dosagem , Doença das Coronárias/tratamento farmacológico , Peptídeos/administração & dosagem , Peptídeos/farmacocinética , Inibidores da Agregação Plaquetária/administração & dosagem , Agregação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Administração Oral , Adulto , Idoso , Aspirina/efeitos adversos , Doença das Coronárias/sangue , Relação Dose-Resposta a Droga , Método Duplo-Cego , Quimioterapia Combinada , Humanos , Pessoa de Meia-Idade , Peptídeos/efeitos adversos , Inibidores da Agregação Plaquetária/efeitos adversos , Trombocitopenia/induzido quimicamente , Tromboxano B2/sangue
4.
Blood ; 97(10): 3093-9, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11342435

RESUMO

The polymorphism responsible for the Pl(A2) alloantigen on the beta(3)-component of beta(3)-containing integrins is reported to be a risk factor for coronary thrombosis. This study examined the effect of Pl(A2) on the function of beta(3)-integrins using platelets from subjects homozygous and heterozygous for Pl(A1) and Pl(A2). There was overlap in the distribution of the dissociation constant (K(d)) and maximum fibrinogen binding (B(max)) values for fibrinogen binding to alpha(IIb)beta(3) on platelets from Pl(A1) and Pl(A2) homozygotes and Pl(A1)/Pl(A2) heterozygotes. However, whereas there was no statistical difference in these values for the Pl(A1) homozygotes and Pl(A2) heterozygotes, the K(d) for the Pl(A2) homozygotes was significantly lower than that for the Pl(A1)/Pl(A2) heterozygotes, but was not statistically different from that for the Pl(A1) homozygotes. No differences were detected in ADP sensitivity between platelets from Pl(A1) homozygotes and Pl(A1)/Pl(A2) heterozygotes, in the IC(50) for RGDS inhibition of fibrinogen binding to alpha(IIb)beta(3), in the alpha(v)beta(3)-mediated adhesion of platelets to osteopontin and vitronectin, and in the phorbol ester-stimulated adhesion to fibrinogen of B lymphocytes expressing alpha(IIb)beta(3) containing either the Pl(A1) or the Pl(A2) polymorphism. Finally, no differential effects of Pl(A2) on turbidometric platelet aggregation, platelet secretion, or platelet thrombus formation were found as measured in the PFA-100. Because no differences were detected in the ability of beta(3)-integrins to interact with ligands based on the presence or absence of the Pl(A2) polymorphism, the results suggest that factors unrelated to beta(3)-integrin function may account for the reported association of the Pl(A2) allele with coronary thrombosis.


Assuntos
Antígenos CD/genética , Antígenos CD/fisiologia , Antígenos de Plaquetas Humanas/genética , Antígenos de Plaquetas Humanas/fisiologia , Plaquetas/fisiologia , Glicoproteínas da Membrana de Plaquetas/genética , Glicoproteínas da Membrana de Plaquetas/fisiologia , Polimorfismo Genético , Difosfato de Adenosina/farmacologia , Adulto , Idoso , Linfócitos B/fisiologia , Plaquetas/efeitos dos fármacos , Plaquetas/imunologia , Trombose Coronária/genética , Feminino , Fibrinogênio/metabolismo , Heterozigoto , Homozigoto , Humanos , Integrina beta3 , Masculino , Pessoa de Meia-Idade , Osteopontina , Adesividade Plaquetária , Agregação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Sialoglicoproteínas/metabolismo , Transfecção , Vitronectina/metabolismo
5.
Am J Med ; 110 Suppl 3A: 28S-32S, 2001 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-11173047

RESUMO

Cyclooxygenase (COX)-1 and COX-2 catalyze the formation of prothrombotic and antithrombotic eicosanoids, respectively. Aspirin, conventional nonsteroidal anti-inflammatory drugs (NSAIDs), and COX-2-specific inhibitors exhibit different patterns of inhibition of COX-1-mediated thromboxane biosynthesis and COX-2-mediated prostacyclin biosynthesis. The relationship between the pharmacologic inhibition of these vasoactive eicosanoids and the thromboprophylaxis or thrombogenicity exhibited by different therapeutic agents is currently unclear. Future studies are needed to assess the antithrombotic properties of commonly used NSAIDs, the hypothetical thrombogenicity of COX-2-specific inhibitors in high-risk patients, the need for concomitant aspirin with selective versus nonselective COX inhibitors, and the antiplatelet and gastric toxicity of the aspirin/COX-2-specific inhibitor combination in comparison with the aspirin/conventional NSAID combination.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Inibidores de Ciclo-Oxigenase/uso terapêutico , Epoprostenol/antagonistas & inibidores , Epoprostenol/biossíntese , Isoenzimas/antagonistas & inibidores , Isoenzimas/fisiologia , Prostaglandina-Endoperóxido Sintases/fisiologia , Trombose/prevenção & controle , Tromboxano A2/antagonistas & inibidores , Tromboxano A2/biossíntese , Anti-Inflamatórios não Esteroides/farmacologia , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/prevenção & controle , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Interações Medicamentosas , Humanos , Proteínas de Membrana , Fatores de Risco , Trombose/etiologia
6.
Arthritis Rheum ; 43(8): 1891-6, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10943882

RESUMO

Specific inhibitors of cyclooxygenase 2 (COX-2) have been approved for the treatment of osteoarthritis and rheumatoid arthritis. Unlike nonsteroidal anti-inflammatory drugs, specific COX-2 inhibitors do not inhibit platelet activation. However, these agents significantly reduce systemic production of prostacyclin. As a result, theoretical concerns have been raised that specific COX-2 inhibitors could shift the hemostatic balance toward a prothrombotic state. Patients with connective tissue diseases (CTD), who may be predisposed to vasculopathy and thrombosis, often have arthritis or pain syndromes requiring treatment with antiinflammatory agents. Herein we describe 4 patients with CTD who developed ischemic complications after receiving celecoxib. All patients had a history of Raynaud's phenomenon, as well as elevated anticardiolipin antibodies, lupus anticoagulant, or a history compatible with antiphospholipid syndrome. It was possible to measure a urinary metabolite of thromboxane A2 in 2 of the patients as an indicator of in vivo platelet activation, and this was markedly elevated in both. In addition, the patients had evidence of ongoing inflammation as indicated by elevated erythrocyte sedimentation rate, hypocomplementemia, and/or elevated levels of anti-DNA antibodies. The findings in these 4 patients suggest that COX-2 inhibitor-treated patients with diseases that predispose to thrombosis should be monitored carefully for this complication.


Assuntos
Doenças do Tecido Conjuntivo/complicações , Doenças do Tecido Conjuntivo/tratamento farmacológico , Inibidores de Ciclo-Oxigenase/uso terapêutico , Sulfonamidas/uso terapêutico , Trombose/complicações , Trombose/etiologia , Adulto , Anticorpos Anticardiolipina/sangue , Celecoxib , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/administração & dosagem , Inibidores de Ciclo-Oxigenase/efeitos adversos , Feminino , Humanos , Isoenzimas/antagonistas & inibidores , Proteínas de Membrana , Pessoa de Meia-Idade , Prostaglandina-Endoperóxido Sintases , Pirazóis , Sulfonamidas/administração & dosagem , Sulfonamidas/efeitos adversos , Trombose/induzido quimicamente
8.
J Pharmacol Exp Ther ; 289(2): 735-41, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10215647

RESUMO

Conventional nonsteroidal anti-inflammatory drugs inhibit both cyclooxygenase (Cox) isoforms (Cox-1 and Cox-2) and may be associated with nephrotoxicity. The present study was undertaken to assess the renal effects of the specific Cox-2 inhibitor, MK-966. Healthy older adults (n = 36) were admitted to a clinical research unit, placed on a fixed sodium intake, and randomized under double-blind conditions to receive the specific Cox-2 inhibitor, MK-966 (50 mg every day), a nonspecific Cox-1/Cox-2 inhibitor, indomethacin (50 mg t.i.d.), or placebo for 2 weeks. All treatments were well tolerated. Both active regimens were associated with a transient but significant decline in urinary sodium excretion during the first 72 h of treatment. Blood pressure and body weight did not change significantly in any group. The glomerular filtration rate (GFR) was decreased by indomethacin but was not changed significantly by MK-966 treatment. Thromboxane biosynthesis by platelets was inhibited by indomethacin only. The urinary excretion of the prostacyclin metabolite 2,3-dinor-6-keto prostaglandin F1alpha was decreased by both MK-966 and indomethacin and was unchanged by placebo. Cox-2 may play a role in the systemic biosynthesis of prostacyclin in healthy humans. Selective inhibition of Cox-2 by MK-966 caused a clinically insignificant and transient retention of sodium, but no depression of GFR. Inhibition of both Cox isoforms by indomethacin caused transient sodium retention and a decline in GFR. Our data suggest that acute sodium retention by nonsteroidal anti-inflammatory drugs in healthy elderly subjects is mediated by the inhibition of Cox-2, whereas depression of GFR is due to inhibition of Cox-1.


Assuntos
Inibidores de Ciclo-Oxigenase/toxicidade , Eicosanoides/metabolismo , Hemodinâmica/efeitos dos fármacos , Isoenzimas/metabolismo , Lactonas , Prostaglandina-Endoperóxido Sintases/metabolismo , Sódio/metabolismo , Equilíbrio Hidroeletrolítico/efeitos dos fármacos , 6-Cetoprostaglandina F1 alfa/análogos & derivados , 6-Cetoprostaglandina F1 alfa/sangue , Idoso , Idoso de 80 Anos ou mais , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Método Duplo-Cego , Feminino , Taxa de Filtração Glomerular/efeitos dos fármacos , Humanos , Indometacina/toxicidade , Masculino , Proteínas de Membrana , Pessoa de Meia-Idade , Sódio/urina , Sulfonas , Tromboxano B2/análogos & derivados , Tromboxano B2/sangue , Tromboxano B2/urina
9.
Proc Natl Acad Sci U S A ; 96(1): 272-7, 1999 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-9874808

RESUMO

Prostaglandins (PG) are synthesized by two isoforms of the enzyme PG G/H synthase [cyclooxygenase (COX)]. To examine selectivity of tolerated doses of an inhibitor of the inducible COX-2 in humans, we examined the effects of celecoxib on indices of COX-1-dependent platelet thromboxane (Tx) A2 and on systemic biosynthesis of prostacyclin in vivo. Volunteers received doses of 100, 400, or 800 mg of celecoxib or 800 mg of a nonselective inhibitor, ibuprofen. Ibuprofen, but not celecoxib, significantly inhibited TxA2-dependent aggregation, induced ex vivo by arachidonic acid (83 +/- 11% vs. 11. 9 +/- 2.2%; P < 0.005) and by collagen. Neither agent altered aggregation induced by thromboxane mimetic, U46619. Ibuprofen reduced serum TxB2 (-95 +/- 2% vs. -6.9 +/- 4.2%; P < 0.001) and urinary excretion of the major Tx metabolite, 11-dehydro TxB2 (-70 +/- 9.9% vs. -20.3 +/- 5.3%; P < 0.05) when compared with placebo. Despite a failure to suppress TxA2-dependant platelet aggregation, celecoxib had a modest but significant inhibitory effect on serum TxB2 4 hr after dosing. By contrast, both ibuprofen and celecoxib suppressed a biochemical index of COX-2 activity (endotoxin induced PGE2 in whole blood ex vivo) to a comparable degree (-93.3 +/- 2% vs. -83 +/- 6.1%). There was no significant difference between the doses of celecoxib on COX-2 inhibition. Celecoxib and ibuprofen suppressed urinary excretion of the prostacyclin metabolite 2,3 dinor 6-keto PGF1alpha. These data suggest that (i) platelet COX-1-dependent aggregation is not inhibited by up to 800 mg of celecoxib; (ii) comparable COX-2 inhibition is attained by celecoxib (100-800 mg) and ibuprofen (800 mg) after acute dosing; and (iii) COX-2 is a major source of systemic prostacyclin biosynthesis in healthy humans.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Epoprostenol/biossíntese , Isoenzimas/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Sulfonamidas/farmacologia , 6-Cetoprostaglandina F1 alfa/análogos & derivados , 6-Cetoprostaglandina F1 alfa/urina , Adulto , Celecoxib , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Dinoprostona/metabolismo , Feminino , Humanos , Ibuprofeno/farmacocinética , Ibuprofeno/farmacologia , Isoenzimas/sangue , Isoenzimas/efeitos dos fármacos , Masculino , Proteínas de Membrana , Pessoa de Meia-Idade , Monócitos/enzimologia , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Prostaglandina-Endoperóxido Sintases/sangue , Prostaglandina-Endoperóxido Sintases/efeitos dos fármacos , Pirazóis , Sulfonamidas/farmacocinética , Tromboxano B2/análogos & derivados , Tromboxano B2/sangue , Tromboxano B2/urina
10.
J Cardiovasc Pharmacol ; 33(1): 12-8, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9890391

RESUMO

Platelet inhibition significantly reduces the risk of cardiovascular mortality and morbidity. However, current antiplatelet therapies have limitations, and more efficacious agents are needed. E5510 is a novel compound that has multiple platelet inhibitory effects in in vitro studies. We compared the in vivo, pharmacodynamic effects of maximal antiplatelet doses of E5510 (20 mg) with 300 mg aspirin in a placebo-controlled, triple crossover trial in nine healthy volunteers. Collagen-induced platelet aggregation and serum thromboxane B2 (TxB2) were similarly inhibited by both compounds in the first 12 h but showed recovery at 24 h in the E5510 group only (p < 0.05). Thrombin and U46619-induced platelet aggregation, as well as basal and prostaglandin E2 (PGE2)-stimulated platelet cyclic adenosine monophosphate (cAMP) levels were unchanged after ingestion of either agent. E5510 and aspirin reduced systemic thromboxane formation without affecting prostacyclin biosynthesis. Neither E5510 nor aspirin inhibited the excretion of 8-epi PGF2alpha and 5,6-DHET, two indices of cyclooxygenase-independent arachidonate metabolism. In conclusion, (a) E55 10 in vivo most likely induces a reversible inhibition of cyclooxygenase, without affecting thromboxane synthetase, phosphodiesterase, thrombin, or thromboxane receptor-mediated signaling; (b) single doses of aspirin or E5510 affect thromboxane/prostacyclin profiles favorably, supporting their use in acute coronary syndromes. This study outlines a comprehensive and minimally invasive approach for the assessment of the in vivo mechanism of action of novel antiplatelet agents.


Assuntos
Aspirina/farmacologia , Plaquetas/efeitos dos fármacos , Ácidos Graxos Monoinsaturados/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Adulto , Aspirina/farmacocinética , Tempo de Sangramento , Plaquetas/metabolismo , Estudos Cross-Over , AMP Cíclico/metabolismo , Método Duplo-Cego , Eicosanoides/urina , Ácidos Graxos Monoinsaturados/farmacocinética , Humanos , Masculino , Inibidores da Agregação Plaquetária/farmacocinética , Tromboxanos/sangue
11.
Circulation ; 98(16): 1610-5, 1998 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-9778325

RESUMO

BACKGROUND: Parenteral heparin is the anticoagulant of choice in hospitalized patients. Continued anticoagulation is achieved by subcutaneous administration of low-molecular-weight heparin or with an orally active anticoagulant such as warfarin. An oral heparin formulation would avoid the inconvenience of subcutaneous injection and the unfavorable drug interactions and adverse events associated with warfarin. A candidate delivery agent, sodium N-[8(-2-hydroxybenzoyl)amino]caprylate (SNAC), was evaluated with escalating oral heparin doses in a randomized, double-blind, controlled clinical study for safety, tolerability, and effects on indexes of anticoagulation. METHODS AND RESULTS: Increases in activated partial thromboplastin time (aPTT), anti-factors IIa and Xa, and tissue factor pathway inhibitor (TFPI) concentrations were detected when normal volunteers were dosed with 10.5 g SNAC/20000 IU heparin by gavage in some subjects. For the entire group, 30000 IU SNAC and heparin elevated TFPI from 74.9+/-7.6 to 254.2+/-12.3 mg/mL (P<0.001) 1 hour after dosing (P<0.001). Similar changes occurred in anti-factor IIa and anti-factor Xa. aPTT rose from 28+/-0.5 to 42.2+/-6.3 seconds 2 hours after dosing (P<0.01). No significant changes in vital signs, physical examination, ECGs, or clinical laboratory values were observed. Neither 30000 IU heparin alone nor 10.5 g SNAC alone altered the hemostatic parameters. Emesis was associated with 10.5 g SNAC. A taste-masked preparation of SNAC 2.25 g was administered orally with heparin 30000 to 150000 IU. Both aPTT and anti-factor Xa increased with escalating doses of heparin. This preparation was well tolerated. Conclusions-Heparin, administered orally in combination with the delivery agent SNAC, produces significant elevations in 4 indexes of anticoagulant effect in healthy human volunteers. These results establish the feasibility of oral delivery of anticoagulant doses of heparin in humans and may have broader implications for the absorption of macromolecules.


Assuntos
Anticoagulantes/uso terapêutico , Caprilatos/farmacologia , Sistemas de Liberação de Medicamentos , Heparina/uso terapêutico , Administração Oral , Anticoagulantes/efeitos adversos , Anticoagulantes/farmacocinética , Glicemia/metabolismo , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Heparina/efeitos adversos , Heparina/farmacocinética , Humanos , Insulina/sangue , Absorção Intestinal/efeitos dos fármacos , Masculino , Tempo de Tromboplastina Parcial , Valores de Referência , Resultado do Tratamento
12.
Coron Artery Dis ; 8(2): 105-11, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9211051

RESUMO

Heparin, the most widely used antithrombin, suffers several limitations, including high inter-individual variability of anticoagulant response, a nonlinear dose-response curve, inability to inactivate clot-bound thrombin, a requirement for endogenous cofactors and inactivation by platelet factor 4 and heparinase. These shortcomings may explain its suboptimal efficacy and safety in the prevention of arterial vessel occlusion. Heparin's drawbacks may be overcome by direct thrombin inhibitors. The development of these specific antithrombins has been a major therapeutic goal of the past decade. The high expectations generated by the use of these compounds in experimental models of arterial thrombosis appeared to be confirmed by the initial phase I and II clinical studies. However, large phase III trials have been highly discouraging: three trials with hirudin have been interrupted as a result of a high incidence of serious adverse events. Two of these trials were subsequently restarted at lower doses and did not support an incremental efficacy of hirudin over heparin. Two trials in the setting of angioplasty (one with hirudin and one with hirulog) have also failed to demonstrate the superiority of these compounds over heparin. Is this the result of a very narrow therapeutic range of these agents or the consequence of poor design of the phase II studies leading to the selection of inappropriate doses for the comparative efficacy trials? This review focuses on the clinical development of two specific antithrombins: hirudin and hirulog. The experimental pharmacology and human studies of Argatroban are discussed in a review by Fitzgerald and Murphy.


Assuntos
Antitrombinas/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Terapia com Hirudina , Hirudinas/análogos & derivados , Fragmentos de Peptídeos/uso terapêutico , Trombina/antagonistas & inibidores , Animais , Antitrombinas/farmacologia , Doenças Cardiovasculares/sangue , Avaliação de Medicamentos , Hirudinas/farmacologia , Humanos , Fragmentos de Peptídeos/farmacologia , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Trombina/metabolismo , Resultado do Tratamento
14.
Diabetes Res Clin Pract ; 30 Suppl: 13-8, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8964187

RESUMO

The role of oxidative stress and platelet activation in the development and evolution of diabetic vascular complications is unclear. They can both be the consequence of established vascular disease or a contributing factor to the evolution of atherosclerosis. Free radical generation may both lead to or result from platelet activation, suggesting that oxidative stress and platelet activation may be closely interrelated. Controversial results may partly reflect methodological constraints. Novel techniques for the measurement of in vivo indices of oxidant injury have been developed and will be used in conjunction with pharmacological probes to establish whether oxidative stress is enhanced in diabetes and whether this preceeds the onset of micro and macrovascular disease.


Assuntos
Diabetes Mellitus/fisiopatologia , Estresse Oxidativo , Ativação Plaquetária , Animais , Arteriosclerose/etiologia , Angiopatias Diabéticas/etiologia , Produtos Finais de Glicação Avançada/metabolismo , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...