Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Pharmacol ; 41(4): 397-403, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11304896

RESUMO

The pharmacokinetics of 1, 10, 23.25, and 139.5 mg doses of atrasentan was assessed in a placebo-controlled, double-blind, single oral dose study in 24 healthy male subjects. Atrasentan was well tolerated. Atrasentan pharmacokinetics was linear in the 1 to 23.25 mg dose range, with some dose dependency in the highest dose group. Harmonic mean terminal half-life was similar across all dose groups (20-25 h). Apparent oral clearance was low (12 L/h) for the highest dose group compared with the other three dose groups (21-27 L/h). The apparent volume of distribution was large (approximately 6 L/kg), consistent with extensive tissue distribution.


Assuntos
Antagonistas dos Receptores de Endotelina , Administração Oral , Adolescente , Adulto , Análise de Variância , Pressão Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Método Duplo-Cego , Endotelina-1/efeitos dos fármacos , Meia-Vida , Humanos , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Placebos
2.
Drug Metab Dispos ; 27(8): 902-8, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10421617

RESUMO

ABT-378 is a potent in vitro inhibitor of the HIV protease and is currently being developed for coadministration with another HIV protease inhibitor, ritonavir, as an oral therapeutic treatment for HIV infection. In the present study, the effect of ritonavir, a potent inhibitor of cytochrome P-450 (CYP) 3A, on the in vitro metabolism of ABT-378 was examined. Furthermore, the effect of ABT-378-ritonavir combinations on several CYP-dependent monooxygenase activities in human liver microsomes was also examined. ABT-378 was found to undergo NADPH- and CYP3A4/5-dependent metabolism to three major metabolites, M-1 (4-oxo) and M-3/M-4 (4-hydroxy epimers), as well as several minor oxidative metabolites in human liver microsomes. The mean apparent K(m) and V(max) values for the metabolism of ABT-378 by human liver microsomes were 6.8 +/- 3.6 microM and 9.4 +/- 5.5 nmol of ABT-378 metabolized/mg protein/min, respectively. Ritonavir inhibited human liver microsomal metabolism of ABT-378 potently (K(i) = 0.013 microM). The combination of ABT-378 and ritonavir was much weaker in inhibiting CYP-mediated biotransformations than ritonavir alone, and the inhibitory effect appears to be primarily due to the ritonavir component of the combination. The ABT-378-ritonavir combinations (at 3:1 and 29:1 ratios) inhibited CYP3A (IC(50) = 1.1 and 4.6 microM), albeit less potently than ritonavir (IC(50) = 0.14 microM). Metabolic reactions mediated by CYP1A2, CYP2A6, and CYP2E1 were not affected by the ABT-378-ritonavir combinations. The inhibitory effects of ABT-378-ritonavir combinations on CYP2B6 (IC(50) = >30 microM), CYP2C9 (IC(50) = 13.7 and 23.0 microM), CYP2C19 (IC(50) = 28.7 and 38.0 microM), and CYP2D6 (IC(50) = 13.5 and 29.0 microM) were marginal and are not likely to produce clinically significant drug-drug interactions.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/metabolismo , Inibidores da Protease de HIV/metabolismo , Inibidores da Protease de HIV/farmacologia , Microssomos Hepáticos/enzimologia , Oxirredutases N-Desmetilantes/antagonistas & inibidores , Oxirredutases N-Desmetilantes/metabolismo , Pirimidinonas/metabolismo , Pirimidinonas/farmacologia , Ritonavir/metabolismo , Ritonavir/farmacologia , Anticorpos Bloqueadores/farmacologia , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP3A , Interações Medicamentosas , Humanos , Técnicas In Vitro , Isoenzimas/metabolismo , Cinética , Lopinavir , Microssomos Hepáticos/efeitos dos fármacos
3.
Antimicrob Agents Chemother ; 42(12): 3107-12, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9835499

RESUMO

The potential interaction between fluoxetine, a known inhibitor of cytochrome P-450 isoform 2D6 (CYP2D6), and ritonavir, a human immunodeficiency virus type 1 protease inhibitor, was evaluated in this open-label study. Sixteen male and female subjects ranging in age from 18 to 40 years completed the study. Subjects received single doses of 600 mg of ritonavir on days 1 and 10. On study days 3 to 10, all subjects received 30 mg of fluoxetine every 12 h for a total of 16 consecutive doses. Serial blood samples for determination of ritonavir concentrations in plasma were collected after the administration of ritonavir on days 1 and 10. A limited number of blood samples for determination of fluoxetine and norfluoxetine concentrations were collected after administration of the morning dose on day 10. A statistically significant increase (19%) in the ritonavir area under the concentration-time curve (AUC) was observed with concomitant fluoxetine administration, with individual changes ranging from -12 to +56%. The change in the ritonavir AUC with concomitant fluoxetine administration was positively correlated with the norfluoxetine 24-h AUC (AUC24) (r2 = 0.42), the norfluoxetine/fluoxetine AUC24 ratio (r2 = 0.53), and the fluoxetine elimination rate constant (r2 = 0.65), with larger increases in the ritonavir AUC tending to occur with higher norfluoxetine concentrations and higher fluoxetine elimination rate constants. The effect of fluoxetine appeared to be larger in subjects with the CYP2D6 wt/wt genotype. There was little or no effect on the time to maximum drug concentration (Cmax) in serum, Cmax, and the elimination rate constant of ritonavir with concomitant fluoxetine administration. Considering the magnitude of the change observed, no ritonavir dose adjustment is recommended during concomitant fluoxetine administration.


Assuntos
Fármacos Anti-HIV/farmacocinética , Antidepressivos de Segunda Geração/farmacologia , Inibidores Enzimáticos/farmacologia , Fluoxetina/farmacologia , Inibidores da Protease de HIV/farmacocinética , Ritonavir/farmacocinética , Adolescente , Adulto , Área Sob a Curva , Citocromo P-450 CYP2D6/genética , Inibidores do Citocromo P-450 CYP2D6 , Interações Medicamentosas , Feminino , Genótipo , HIV-1 , Meia-Vida , Humanos , Masculino , Pessoa de Meia-Idade
4.
Antimicrob Agents Chemother ; 42(12): 3218-24, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9835517

RESUMO

The valine at position 82 (Val 82) in the active site of the human immunodeficiency virus (HIV) protease mutates in response to therapy with the protease inhibitor ritonavir. By using the X-ray crystal structure of the complex of HIV protease and ritonavir, the potent protease inhibitor ABT-378, which has a diminished interaction with Val 82, was designed. ABT-378 potently inhibited wild-type and mutant HIV protease (Ki = 1.3 to 3.6 pM), blocked the replication of laboratory and clinical strains of HIV type 1 (50% effective concentration [EC50], 0.006 to 0.017 microM), and maintained high potency against mutant HIV selected by ritonavir in vivo (EC50, 50-fold after 8 h. In healthy human volunteers, coadministration of a single 400-mg dose of ABT-378 with 50 mg of ritonavir enhanced the area under the concentration curve of ABT-378 in plasma by 77-fold over that observed after dosing with ABT-378 alone, and mean concentrations of ABT-378 exceeded the EC50 for >24 h. These results demonstrate the potential utility of ABT-378 as a therapeutic intervention against AIDS.


Assuntos
Fármacos Anti-HIV/farmacologia , Inibidores da Protease de HIV/farmacologia , Pirimidinonas/farmacologia , Animais , Fármacos Anti-HIV/metabolismo , Fármacos Anti-HIV/farmacocinética , Área Sob a Curva , Cristalografia por Raios X , Cães , Interações Medicamentosas , Feminino , Protease de HIV/química , Inibidores da Protease de HIV/metabolismo , Inibidores da Protease de HIV/farmacocinética , HIV-1/efeitos dos fármacos , Humanos , Técnicas In Vitro , Lopinavir , Macaca fascicularis , Masculino , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Pirimidinonas/metabolismo , Pirimidinonas/farmacocinética , Ratos , Ratos Sprague-Dawley , Ritonavir/química , Ritonavir/farmacologia
5.
Clin Pharmacol Ther ; 64(4): 355-62, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9797791

RESUMO

BACKGROUND: Because ritonavir, a human immunodeficiency virus (HIV) protease inhibitor, and clarithromycin, a macrolide antibiotic used in the treatment of disseminated infection caused by Mycobacterium avium complex, are likely to be administered concurrently for treatment of patients with HIV and acquired immunodeficiency syndrome (AIDS), the drug interaction potential of these 2 agents was evaluated. Both clarithromycin and ritonavir are metabolized to a significant extent through cytochrome P450-mediated biotransformation and are potential inhibitors of these enzymes. OBJECTIVE: To evaluate the pharmacokinetic effects of concomitant administration of multiple doses of ritonavir and clarithromycin. METHODS: This was an open-label, randomized, 3-period crossover study. Ritonavir alone (200 mg every 8 hours), clarithromycin alone (500 mg every 12 hours), and ritonavir and clarithromycin in combination were administered to 22 healthy volunteers. Blood samples were collected on day 4 for determination of ritonavir, clarithromycin, and its metabolite 14-(R)-hydroxyclarithromycin. RESULTS: Ritonavir practically completely inhibited the formation of 14-(R)-hydroxyclarithromycin. The mean area under the plasma concentration-time curve (AUC) for clarithromycin increased by 77% with concomitant ritonavir, and the harmonic mean terminal half-life increased from 5 hours to 14 hours. Statistically significant increases in peak plasma concentration (31%) and minimum plasma concentration (182%) were also observed. The effect of concomitant clarithromycin administration on ritonavir pharmacokinetics was statistically significant but small, with a 12.5% increase in mean AUC and a 15.3% increase in peak plasma concentration. The terminal half-life increased from 3.47 to 3.87 hours with concomitant clarithromycin. CONCLUSIONS: No adjustment of the ritonavir dose is necessary when administered with clarithromycin. In addition, no changes in clarithromycin dose are warranted in patients with normal renal function.


Assuntos
Antibacterianos/farmacocinética , Claritromicina/farmacocinética , Inibidores da Protease de HIV/farmacocinética , Ritonavir/farmacocinética , Infecções Oportunistas Relacionadas com a AIDS/tratamento farmacológico , Adulto , Antibacterianos/administração & dosagem , Antibacterianos/sangue , Claritromicina/administração & dosagem , Claritromicina/sangue , Estudos Cross-Over , Esquema de Medicação , Interações Medicamentosas , Feminino , Inibidores da Protease de HIV/administração & dosagem , Inibidores da Protease de HIV/sangue , Meia-Vida , Humanos , Hidroxilação , Masculino , Pessoa de Meia-Idade , Infecção por Mycobacterium avium-intracellulare/tratamento farmacológico , Valores de Referência , Ritonavir/administração & dosagem , Ritonavir/sangue
6.
Clin Pharmacokinet ; 35(4): 275-91, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9812178

RESUMO

Ritonavir is 1 of the 4 potent synthetic HIV protease inhibitors, approved by the US Food and Drug Administration (FDA) between 1995 and 1997, that have revolutionised HIV therapy. The extent of oral absorption is high and is not affected by food. Within the clinical concentration range, ritonavir is approximately 98 to 99% bound to plasma proteins, including albumin and alpha 1-acid glycoprotein. Cerebrospinal fluid (CSF) drug concentrations are low in relation to total plasma concentration. However, parallel decreases in the viral burden have been observed in the plasma, CSF and other tissues. Ritonavir is primarily metabolised by cytochrome P450 (CYP) 3A isozymes and, to a lesser extent, by CYP2D6. Four major oxidative metabolites have been identified in humans, but are unlikely to contribute to the antiviral effect. About 34% and 3.5% of a 600 mg dose is excreted as unchanged drug in the faeces and urine, respectively. The clinically relevant t1/2 beta is about 3 to 5 hours. Because of autoinduction, plasma concentrations generally reach steady state 2 weeks after the start of administration. The pharmacokinetics of ritonavir are relatively linear after multiple doses, with apparent oral clearance averaging 7 to 9 L/h. In vitro, ritonavir is a potent inhibitor of CYP3A. In vivo, ritonavir significantly increases the AUC of drugs primarily eliminated by CYP3A metabolism (e.g. clarithromycin, ketoconazole, rifabutin, and other HIV protease inhibitors, including indinavir, saquinavir and nelfinavir) with effects ranging from an increase of 77% to 20-fold in humans. It also inhibits CYP2D6-mediated metabolism, but to a significantly lesser extent (145% increase in desipramine AUC). Since ritonavir is also an inducer of several metabolising enzymes [CYP1A4, glucuronosyl transferase (GT), and possibly CYP2C9 and CYP2C19], the magnitude of drug interactions is difficult to predict, particularly for drugs that are metabolised by multiple enzymes or have low intrinsic clearance by CYP3A. For example, the AUC of CYP3A substrate methadone was slightly decreased and alprazolam was unaffected. Ritonavir is minimally affected by other CYP3A inhibitors, including ketoconazole. Rifampicin (rifampin), a potent CYP3A inducer, decreased the AUC of ritonavir by only 35%. The degree and duration of suppression of HIV replication is significantly correlated with the plasma concentrations. Thus, the large increase in the plasma concentrations of other protease inhibitors when coadministered with ritonavir forms the basis of rational dual protease inhibitor regimens, providing patients with 2 potent drugs at significantly reduced doses and less frequent dosage intervals. Combination treatment of ritonavir with saquinavir and indinavir results in potent and sustained clinical activity. Other important factors with combination regimens include reduced interpatient variability for high clearance agents, and elimination of the food effect on the bioavailibility of indinavir.


Assuntos
Fármacos Anti-HIV/farmacocinética , Inibidores da Protease de HIV/farmacocinética , Ritonavir/farmacocinética , Animais , Fármacos Anti-HIV/farmacologia , Interações Medicamentosas , Inibidores da Protease de HIV/farmacologia , Humanos , Ritonavir/farmacologia
7.
Antimicrob Agents Chemother ; 42(11): 2784-91, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9797204

RESUMO

The pharmacokinetic interaction between indinavir and ritonavir was evaluated in five groups of healthy adult volunteers to explore the potential for twice-daily (b.i.d.) dosing of this combination. All subjects received 800 mg of indinavir every 8 h (q8h) on day 2. In addition, subjects in group I received one dose of 800 mg of indinavir on day 1 and 800 mg of indinavir q8h on day 17. Subjects in Groups II and IV each received one dose of 600 mg of indinavir on days 1 and 17, and subjects in groups III and V each received one dose of 400 mg of indinavir on days 1 and 17. During days 3 to 17, ritonavir placebo or ritonavir at 200, 300, 300, or 400 mg q12h was given to groups I, II, III, IV, and V, respectively. Ritonavir at steady state probably inhibited the cytochrome P-450 3A metabolism of indinavir and substantially increased plasma indinavir concentrations, with the area under the plasma concentration-time curve (AUC) increasing up to 475% and the peak concentration in serum (Cmax) increasing up to 110%. The Cmax/trough concentration ratio decreased from 50 in standard q8h regimens to less than 14 when indinavir was administered with ritonavir. For a constant indinavir dose, an increase in the ritonavir dose yielded similar indinavir AUCs, Cmaxs, and concentrations at 12 h (C12s). For a constant ritonavir dose, an increase in the indinavir dose resulted in approximately proportional increases in the indinavir AUC, less than proportional increases in Cmax, and slightly more than proportional increases in C12. Ritonavir reduced between-subject variability in the indinavir AUC and trough concentrations and did not affect indinavir renal clearance. With the altered pharmacokinetic profile, indinavir likely could be given as a b.i.d. combination regimen with ritonavir. This could potentially improve patient compliance and thereby reduce treatment failures.


Assuntos
Fármacos Anti-HIV/farmacologia , Inibidores da Protease de HIV/farmacologia , Indinavir/farmacocinética , Ritonavir/farmacologia , Adolescente , Adulto , Fármacos Anti-HIV/farmacocinética , Relação Dose-Resposta a Droga , Esquema de Medicação , Interações Medicamentosas , Quimioterapia Combinada , Feminino , Inibidores da Protease de HIV/farmacocinética , Humanos , Indinavir/efeitos adversos , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Ritonavir/efeitos adversos , Ritonavir/farmacocinética
9.
Br J Clin Pharmacol ; 46(2): 111-6, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9723818

RESUMO

AIMS: To assess the effects of the protease inhibitor ritonavir on the pharmacokinetics of ethinyl oestradiol in healthy female volunteers. METHODS: This was an open-label, single centre study in 23 subjects who received two single doses of oral contraceptive containing 50 microg ethinyl oestradiol on Day 1 (alone) and on Day 29 during concomitant ritonavir. Each subject received 16 days of every 12 h doses of ritonavir from Day 15 through Day 30. Blood samples were collected for serum ethinyl oestradiol concentrations for 48 h after each dose and for plasma ritonavir on Day 29 at 0 and 4 h postdose. RESULTS: Statistically significant decreases in ethinyl oestradiol mean Cmax (-32%) and mean AUC (-41%), and a statistically significant increase in the mean terminal elimination rate constant (+31%) were observed during concomitant ritonavir. The harmonic mean terminal half-life decreased from 17 h to 13 h during concomitant ritonavir. No statistically significant change was noted in tmax. The ratios of means (95% confidence intervals) for Cmax and AUC were 0.682 (0.612-0.758) and 0.595 (0.506-0.694), respectively. The changes in ethinyl oestradiol pharmacokinetics were consistent with an increase in clearance from enzymatic induction of glucuronidation and/or cytochrome P450 hydroxylation. Mean steady-state ritonavir concentrations of 6.5 and 13.4 microg ml(-1) were observed at 0 and 4 h postdose, respectively. CONCLUSIONS: Considering the extent of the decrease in ethinyl oestradiol concentrations, the use of alternate contraceptive measures should be considered when ritonavir is being administered.


PIP: The protease inhibitor ritonavir has demonstrated broad-spectrum ability against HIV 1 and 2. The present study investigated the drug interaction potential of steady-state ritonavir on single dose ethinyl estradiol pharmacokinetics. 23 healthy women (mean age, 34 years) received an oral contraceptive (OC) containing 50 mcg of ethinyl estradiol and 1 mg of ethynodiol on days 1 and 29, while 500 mg of ritonavir was administered every 12 hours on days 15-30. After administration of a single dose of OC, serum ethinyl estradiol concentrations peaked at 4 hours and declined thereafter, with a typical half-life of 17 hours. Administration of the second OC on day 29, after 16 days of continuous ritonavir, resulted in a 32% lower ethinyl estradiol mean maximum concentration (p 0.001) and a 41% lower mean area under curve (p 0.001) compared with OC administration alone. In addition, the mean terminal elimination rate constant increased by 31% (p 0.001) with concomitant ritonavir. The changes in ethinyl estradiol pharmacokinetics were consistent with an increase in clearance from enzymatic induction of glucuronidation and/or cytochrome P450 hydroxylation. Mean steady-state ritonavir concentrations of 6.5 and 13.4 mcg/ml were observed at 0 and 4 hours post-dose, respectively. The interaction observed in this study is likely to be clinically significant, with an increased risk of OC failure. Thus, use of alternate contraceptive measures should be recommended when ritonavir is being administered.


Assuntos
Fármacos Anti-HIV/farmacologia , Anticoncepcionais Orais/farmacocinética , Congêneres do Estradiol/farmacocinética , Etinilestradiol/farmacocinética , Inibidores da Protease de HIV/farmacologia , Ritonavir/farmacologia , Adulto , Área Sob a Curva , Interações Medicamentosas , Feminino , Meia-Vida , Humanos , Pessoa de Meia-Idade
10.
Epilepsia ; 39(8): 868-73, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9701378

RESUMO

PURPOSE: In two open-label long-term safety studies, we determined tiagabine (TGB) pharmacokinetics in patients with epilepsy. METHODS: In all, 2,147 plasma samples from 511 patients who participated in the studies were available. The total daily dose ranged from 2 mg administered once daily to 80 mg administered in four doses. A one-compartment model with first-order absorption and elimination was used to fit the TGB plasma concentration-time data, with a population pharmacokinetic approach. RESULTS: The patients' average (+/-SD) weight and age were 73.8+/-20.7 kg and 32.1+/-12.3 years. The most significantly factor affecting TGB pharmacokinetics was concomitant administration of other antiepileptic drugs (AEDs). The central clearance value in patients receiving AEDs known to induce hepatic drug metabolism was 21.4 L/h, a value 67% higher than the central clearance estimate obtained for the patients receiving AEDs not known to affect hepatic drug metabolism (12.8 L/h). There was no evidence of any dose or time effect, indicating that TGB pharmacokinetics are linear. TGB pharmacokinetics were not different in white, black, or Hispanic patients, although our ability to explore racial effects was limited since 90% of the patients were white. No other demographic variables (including age and smoking) or any clinical chemistry measurements (including bilirubin, SGOT, and SGPT) were important in explaining the variability in the clearance estimates. CONCLUSIONS: TGB pharmacokinetics are linear, influenced by enzyme-inducing AEDs, and largely unaffected by other demographic variables.


Assuntos
Anticonvulsivantes/farmacocinética , Epilepsia/sangue , Ácidos Nipecóticos/farmacocinética , Adolescente , Adulto , Fatores Etários , Idoso , Anticonvulsivantes/sangue , Anticonvulsivantes/uso terapêutico , Disponibilidade Biológica , Peso Corporal , Criança , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Indução Enzimática/efeitos dos fármacos , Epilepsia/tratamento farmacológico , Epilepsia/metabolismo , Feminino , Humanos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Pessoa de Meia-Idade , Modelos Estatísticos , Ácidos Nipecóticos/sangue , Ácidos Nipecóticos/uso terapêutico , Análise de Regressão , Estudos Retrospectivos , Fatores Sexuais , Fumar/metabolismo , Tiagabina
11.
Clin Pharmacol Ther ; 63(4): 453-64, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9585800

RESUMO

OBJECTIVE: To assess the pharmacokinetic interaction between ritonavir and saquinavir. METHODS: Ritonavir and saquinavir were administered in single doses to six groups of healthy volunteers in a two-way (saquinavir alone and ritonavir plus saquinavir for groups I through V) and a three-way (ritonavir alone, saquinavir alone, and ritonavir plus saquinavir for group VI) crossover manner with the following doses: group I, 200 mg saquinavir and 300 mg ritonavir; group II, 200 mg saquinavir and 600 mg ritonavir; group III, 400 mg saquinavir and 300 mg ritonavir; group IV, 400 mg saquinavir and 600 mg ritonavir; group V; 600 mg saquinavir and 200 mg ritonavir; group VI, 600 mg saquinavir and 600 mg ritonavir. RESULTS: Coadministration of ritonavir markedly increased the area under the plasma concentration-time curve (AUC) and peak concentration of saquinavir (> 50-fold and 22-fold, respectively). For a constant ritonavir dose, the pharmacokinetics of saquinavir were relatively proportional to dose. For a constant saquinavir dose, the increase in saquinavir concentration tended to be less than proportional to ritonavir dose. Ritonavir reduced intersubject variability in the saquinavir AUC from 60% to 28%. The in vivo inhibition constant was 0.025 +/- 0.020 micrograms/ml with noncompartmental estimation and 0.0164 +/- 0.0004 micrograms/ml with nonlinear mixed-effects model compartmental analysis. Saquinavir showed no clinically significant effect on the pharmacokinetics of ritonavir (+6.4% in AUC). The regimens were well tolerated. CONCLUSIONS: The large effect of ritonavir on the pharmacokinetics of saquinavir is consistent with a large reduction of saquinavir first-pass metabolism and postabsorptive clearance. Given the limited bioavailability of saquinavir given in the hard gelatin capsule formulation, this drug interaction is expected to have implications in the use of protease inhibitors in the management of human immunodeficiency virus infection.


Assuntos
Fármacos Anti-HIV/farmacocinética , Inibidores da Protease de HIV/farmacocinética , Ritonavir/farmacocinética , Saquinavir/farmacocinética , Adulto , Análise de Variância , Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/sangue , Área Sob a Curva , Estudos Cross-Over , Esquema de Medicação , Interações Medicamentosas , Feminino , Inibidores da Protease de HIV/administração & dosagem , Inibidores da Protease de HIV/sangue , Humanos , Masculino , Valores de Referência , Ritonavir/administração & dosagem , Ritonavir/sangue , Saquinavir/administração & dosagem , Saquinavir/sangue
12.
Int J Clin Pharmacol Ther ; 36(3): 146-51, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9562230

RESUMO

The effect of sertindole (a new selective antipsychotic compound) on the pharmacokinetic disposition of terfenadine was investigated. Thirteen subjects who completed the study received a single 120 mg dose of terfenadine alone or with concomitant 20 mg sertindole daily. The mean values for terfenadine Cmax (alone: 2.42 +/- 1.48 ng/ml, in combination: 2.99 +/- 1.85 ng/ml) and AUC (29.6 +/- 18.9 vs 37.9 +/- 23.4 ng x hr/ml) did not change statistically significant in the presence of sertindole (p > 0.05). Similarly, the mean Cmax (531 +/- 195 vs 506 +/- 190 ng/ml) and AUC (3,728 +/- 1,163 vs 4,003 +/- 1,739 ng x hr/ml) values of carboxyterfenadine did not change statistically significant in the presence of sertindole (p > 0.05). The other pharmacokinetic parameters of terfenadine and carboxyterfenadine such as Tmax, t1/2, as well as the carboxyterfenadine to terfenadine Cmax and AUC ratios did not change in the presence of sertindole. Although terfenadine is a substrate for CYP3A (cytochrome P-450 3A), while sertindole is a substrate for both CYP2D6 and CYP3A4, the results in this study suggest that sertindole, at a clinical dose, is not an inhibitor of the metabolism of terfenadine.


Assuntos
Antipsicóticos/farmacologia , Hidrocarboneto de Aril Hidroxilases , Inibidores das Enzimas do Citocromo P-450 , Antagonistas dos Receptores Histamínicos H1/farmacocinética , Imidazóis/farmacologia , Indóis/farmacologia , Oxirredutases N-Desmetilantes/antagonistas & inibidores , Terfenadina/farmacocinética , Absorção , Administração Oral , Adulto , Antipsicóticos/farmacocinética , Área Sob a Curva , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP2D6/química , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/química , Interações Medicamentosas , Feminino , Antagonistas dos Receptores Histamínicos H1/farmacologia , Humanos , Imidazóis/farmacocinética , Indóis/farmacocinética , Masculino , Espectrometria de Massas , Oxigenases de Função Mista/química , Especificidade por Substrato , Terfenadina/farmacologia
13.
Psychopharmacology (Berl) ; 135(3): 236-41, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9498726

RESUMO

The effect of sertindole (a new selective antipsychotic compound) on the pharmacokinetic disposition of alprazolam was investigated. Fourteen subjects who completed the study received a single 1 mg dose of alprazolam without or with concomitant sertindole 12 mg daily. Coadministration of sertindole and alprazolam led to a half-hour decrease (P < 0.05) in mean Tmax value (alone: 1.2 h, in combination: 0.7 h) and a 1.6-h increase in the mean t1/2 value (12.5 +/- 3.2 versus 14.3 +/- 3.4 h, P < 0.05) of alprazolam. The mean Cmax (18.5 +/- 4.9 versus 18.5 +/- 4.8 ng/ml) and AUC (266 +/- 68 versus 275 +/- 57 ng x h/ml) values of alprazolam did not change statistically significantly in the presence of sertindole (P > 0.05). These pharmacokinetic changes are minor and not considered to be of clinical significance. Although both sertindole and alprazolam are substrate for CYP3A4 (cytochrome P-450 3A4), the results in this study suggest that sertindole is not an inhibitor of the metabolism of alprazolam.


Assuntos
Alprazolam/farmacocinética , Ansiolíticos/farmacocinética , Antipsicóticos/farmacologia , Imidazóis/farmacologia , Indóis/farmacologia , Adulto , Antipsicóticos/administração & dosagem , Área Sob a Curva , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP2D6/metabolismo , Interações Medicamentosas , Feminino , Genótipo , Meia-Vida , Humanos , Imidazóis/administração & dosagem , Indóis/administração & dosagem , Masculino
14.
Am J Ther ; 5(2): 73-9, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10099041

RESUMO

This single-center, open-label study examined the safety and potential effect of tiagabine on valproate pharmacokinetics under steady-state conditions. Twelve adult patients with seizures controlled by an individualized fixed dosage of valproate participated in the study. On day 1, the pharmacokinetics of valproic acid were determined. On days 2 through 14, tiagabine was titrated from 8 to 24 mg/d (or the maximum tolerated dose up to 24 mg/d), and the patients continued to take their usual fixed dosage of valproate. Valproic acid pharmacokinetics were assessed again on day 14. The mean maximum plasma concentration (Cmax ) and area under the plasma concentration-time curve from time zero to the end of the dosing interval (AUC0-tau ) for valproic acid were reduced approximately 10% and 12%, respectively (p < or = 0.05), when valproate and tiagabine were administered concomitantly, compared with the mean values when valproate was administered alone. The concomitant administration of these drugs was generally well tolerated. Ten patients reported treatment-emergent adverse events during the study, the most common of which was dizziness(n = 8). Only one patient experienced events that were considered to be severe. There were no clinically important effects on laboratory values, vital signs, or physical exam findings. The small decreases in mean valproic acid Cmax and AUC0-tau observed during the concomitant administration of tiagabine and valproate are probably of limited clinical importance, given the broad therapeutic range of valproate (50-100 microg/mL).


Assuntos
Anticonvulsivantes/administração & dosagem , Anticonvulsivantes/farmacocinética , Agonistas GABAérgicos/administração & dosagem , Agonistas GABAérgicos/farmacocinética , Ácidos Nipecóticos/administração & dosagem , Ácidos Nipecóticos/farmacocinética , Convulsões/tratamento farmacológico , Ácido Valproico/farmacocinética , Adolescente , Adulto , Anticonvulsivantes/sangue , Tontura/induzido quimicamente , Interações Medicamentosas , Monitoramento de Medicamentos , Quimioterapia Combinada , Feminino , Humanos , Masculino , Rinite/induzido quimicamente , Fases do Sono/efeitos dos fármacos , Tiagabina , Fatores de Tempo , Tremor/induzido quimicamente , Ácido Valproico/sangue
15.
J Pharm Sci ; 87(12): 1629-31, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10189278

RESUMO

The pharmacokinetics of sertindole were studied in young, healthy volunteers after single and multiple oral dose administered under an escalating manner. In a low-dose study (study 1), subjects received 4-8 mg with a maintenance dose period of 7 days. In a high-dose study (study 2), subjects received 4 mg daily for 2 days, and the dose was increased by 4 mg increments every third day until reaching 20 mg daily. The mean terminal t 1,2 was 73 h after the final 8 mg dose in study 1 and 60 h after the 20 mg dose in study 2. The terminal elimination phase appeared to be monophasic in all the study subjects, suggesting that Michaelis-Menten saturable metabolism was not involved in the elimination of sertindole. Compartmental analyses suggested that the disproportional increase of the Cmax and AUC values from 4 mg to 20 mg during multiple dosing may be explained by saturable presystemic elimination of sertindole, leading to a higher fraction of sertindole available for absorption at higher doses.


Assuntos
Antipsicóticos/farmacocinética , Imidazóis/farmacocinética , Indóis/farmacocinética , Administração Oral , Adolescente , Adulto , Antipsicóticos/administração & dosagem , Esquema de Medicação , Meia-Vida , Humanos , Imidazóis/administração & dosagem , Indóis/administração & dosagem , Masculino
16.
Clin Pharmacol Ther ; 62(4): 426-35, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9357394

RESUMO

BACKGROUND: Seratrodast, a potent thromboxane receptor antagonist, is approved in Japan for the treatment of asthma and currently is being developed in the United States. METHODS: This was a phase II, randomized, double-blind, parallel-group, placebo-controlled 15-center study of seratrodast in patients with mild to moderate asthma. A total of 183 patients were randomly assigned to receive daily doses of either placebo, or 80 mg seratrodast, or 120 mg seratrodast for 8 weeks. Pharmacokinetic and pharmacodynamic modeling was carried out by means of the population approach. A two-compartment model with zero-order input and first-order elimination best fitted the plasma concentration-time data. RESULTS AND CONCLUSIONS: The pharmacokinetics of seratrodast were linear after single and multiple dosing for 8 weeks. The population estimates for oral clearance and apparent volume of distribution were 8.5 ml/hr/kg and 43.3 ml/kg, respectively. All pharmacokinetic parameters (the oral central compartment clearance, the volumes of distribution of the central and peripheral compartments, and the intercompartmental clearance) were estimated with a precision of 10% or less and were found to be associated with body weight. The residual variability was 30%. The values of oral clearance estimated in this study in male patients were similar to those previously estimated in healthy male subjects. Seratrodast at a dose of 120 mg daily produced an increase in forced expiratory volume in 1 second (FEV1) from baseline that was linearly correlated with its plasma concentrations. The average slope of the concentration-effect relationship was 0.222% and 0.470% per microgram/ml after single and multiple dosing, respectively. Interpatient variability in response was mainly affected by the initial severity of the disease. A lower percentage of predicted FEV1 (i.e., more severe obstruction) was associated with higher slopes, and greater increases in FEV1.


Assuntos
Antiasmáticos/farmacologia , Asma/sangue , Asma/tratamento farmacológico , Benzoquinonas/farmacologia , Ácidos Heptanoicos/farmacologia , Adulto , Idoso , Antiasmáticos/farmacocinética , Asma/fisiopatologia , Benzoquinonas/farmacocinética , Método Duplo-Cego , Feminino , Volume Expiratório Forçado/efeitos dos fármacos , Ácidos Heptanoicos/farmacocinética , Humanos , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Fatores de Tempo , Resultado do Tratamento
17.
Biopharm Drug Dispos ; 18(6): 533-41, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9267685

RESUMO

The pharmacokinetic disposition and relative bioavailability of sertindole administered as a tablet dosage form under fasting conditions, in the presence of food, in the presence of antacid, and as solution was studied in a four-way crossover in young healthy male volunteers. Overall, tablet dosing after a meal or Maalox had no effect on t1/2 or AUC values when compared to fasting conditions, but increased the tmax and decreased the Cmax values slightly. The mean relative bioavailabilities of sertindole administered as tablets after fasting, with food, and with Maalox are 99, 104, and 98%, respectively, compared to sertindole solution. Therefore, sertindole can be administered with and without food and without regard to coadministration of antacids.


Assuntos
Hidróxido de Alumínio/farmacologia , Antiácidos/farmacologia , Antipsicóticos/farmacocinética , Interações Alimento-Droga , Imidazóis/farmacocinética , Indóis/farmacocinética , Hidróxido de Magnésio/farmacologia , Administração Oral , Adulto , Análise de Variância , Antipsicóticos/administração & dosagem , Antipsicóticos/sangue , Área Sob a Curva , Disponibilidade Biológica , Estudos Cross-Over , Combinação de Medicamentos , Jejum/sangue , Humanos , Imidazóis/administração & dosagem , Imidazóis/sangue , Indóis/administração & dosagem , Indóis/sangue , Masculino , Soluções , Comprimidos
18.
Clin Pharmacol Ther ; 62(2): 157-64, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9284851

RESUMO

OBJECTIVE: The pharmacokinetic disposition of oral sertindole, a new selective antipsychotic compound, in young and elderly male and female subjects was investigated. STUDY DESIGN: A total of 46 subjects (12 young males, 11 elderly males, 11 young females, and 12 elderly females) received 4 mg/day sertindole (once a day; days 1 through 3) for 3 consecutive days, 8 mg/day sertindole for 3 consecutive days (days 4 through 6), and 12 mg/day sertindole for 10 consecutive days (days 7 through 16). RESULTS: Age and gender did not appear to have any effect on the plasma binding of sertindole, despite a lower albumin concentration in elderly subjects. After multiple dosing of 12 mg sertindole, the mean peak plasma concentration (Cmax) values for young and elderly female subjects were 20% and 31% higher than those observed for male subjects of comparable age (p < 0.05). The mean values for area under the plasma concentration-time curve [AUC(0-24)] of female subjects were 29% higher than those observed in male subjects of similar age (p < 0.05). There were no statistically significant age-related differences in Cmax and AUC(0-24) (or apparent total plasma clearance), and there were no gender- or age-related differences for the elimination rate constant or values for apparent volume of distribution during the terminal elimination phase after the last 12 mg dose on day 16 (p > 0.05). CONCLUSIONS: There are no differences between young and elderly subjects in the absorption and elimination of sertindole. The higher Cmax and AUC values in females may be a result of a higher extent of absorption or a dependence of sertindole clearance on lean body mass.


Assuntos
Envelhecimento/metabolismo , Antipsicóticos/farmacocinética , Imidazóis/farmacocinética , Indóis/farmacocinética , Absorção , Administração Oral , Adolescente , Adulto , Idoso , Área Sob a Curva , Feminino , Humanos , Masculino , Caracteres Sexuais
19.
Antimicrob Agents Chemother ; 41(5): 898-905, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9145841

RESUMO

The multiple-dose pharmacokinetics of ritonavir were investigated in four groups of human immunodeficiency virus-positive male subjects (with 16 subjects per group) under nonfasting conditions; a 3:1 ritonavir:placebo ratio was used. Ritonavir was given at 200 (group I), 300 (group II), 400 (group III), or 500 (group IV) mg every 12 h for 2 weeks. The multiple-dose pharmacokinetics of ritonavir were moderately dose dependent, with the clearance for group IV (6.8 +/- 2.7 liters/h) being an average of 32% lower than that for group I (10.0 +/- 3.2 liters/h). First-pass metabolism should be minimal for ritonavir. The functional half-life, estimated from peak and trough concentrations, were similar among the dosage groups, averaging 3.1 and 5.7 h after the morning and evening doses, respectively. The area under the concentration-time curve at 24 h (AUC24) and apparent terminal-phase elimination rate constant remained relatively time invariant, but predose concentrations decreased 30 to 70% over time. Concentration-dependent autoinduction is the most likely mechanism for the time-dependent pharmacokinetics. The Km and initial maximum rate of metabolism (Vmax) values estimated from population pharmacokinetic modeling (nonlinear mixed-effects models) were 3.43 microg/ml and 46.9 mg/h, respectively. The group IV Vmax increased to 68 mg/h after 2 weeks. The maximum concentration of ritonavir in serum (Cmax) and AUC after the evening doses were an average of 30 to 40% lower than the values after the morning doses, while the concentration at 12 h was an average of 32% lower than the predose concentration, probably due to protracted absorption. Less than 2% of the dose was eliminated unchanged in the urine. Triglyceride levels increased from the levels at the baseline, and the levels were correlated with baseline triglyceride levels and AUC, Cmax, or predose concentrations.


Assuntos
Infecções por HIV/metabolismo , Inibidores da Protease de HIV/farmacocinética , Ritonavir/farmacocinética , Adulto , Área Sob a Curva , Proteínas Sanguíneas/metabolismo , Relação Dose-Resposta a Droga , Método Duplo-Cego , Inibidores da Protease de HIV/administração & dosagem , Humanos , Masculino , Pessoa de Meia-Idade , Ritonavir/administração & dosagem
20.
Neurology ; 48(4): 1032-7, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9109895

RESUMO

We report an open-label study of 25 children with complex partial seizures that assessed the pharmacokinetics and safety of a single dose of approximately 0.1 mg/kg tiagabine. The children received their usual individualized regimen of one concomitant antiepilepsy drug (AED) throughout the study. Seventeen children were receiving an inducing AED (carbamazepine or phenytoin); eight were receiving valproate. Tiagabine was well tolerated. Dose-normalized Cmax was higher in children taking valproate (18.2 +/- 5.0 ng/mL/mg) than in the induced children (14.8 +/- 6.9 ng/mL/mg), but the difference was not statistically significant. Dose-normalized area under the plasma concentration-time curve from time zero to infinite time was significantly higher (p = 0.002) in children taking valproate (176.5 +/- 54.7 ng.hr/mL/mg) than in induced children (92.4 +/- 56.7 ng.hr/mL/mg). Similarly, oral clearance in the children taking valproate (96 +/- 39 mL/min) was half that of the induced children (207 +/- 91 mL/min). Half-life in children taking valproate (5.7 hr) was almost twice that for the induced children (3.2 hr), and the elimination rate constant was significantly lower (p < 0.02) for the children taking valproate than for the induced children. Volume of distribution was similar in the children taking valproate (52 +/- 9 L) and the induced children (59 +/- 29 L). This is consistent with observations in adults taking tiagabine with inducing AEDs or valproate. Exploratory regressions on these data in children and previous data in adults showed fairly strong relationships between body size and tiagabine clearance and volume of distribution, with body size explaining about 40 to 50% of the variability. When adjusted per kg body weight, clearance and volume were greater in children than adults. When adjusted per m2 body surface area, clearance and volume were more similar in adults and children.


Assuntos
Anticonvulsivantes/farmacocinética , Epilepsia Parcial Complexa/metabolismo , Ácidos Nipecóticos/farmacocinética , Anticonvulsivantes/administração & dosagem , Anticonvulsivantes/uso terapêutico , Constituição Corporal , Criança , Pré-Escolar , Quimioterapia Combinada , Epilepsia Parcial Complexa/tratamento farmacológico , Feminino , Humanos , Masculino , Ácidos Nipecóticos/administração & dosagem , Ácidos Nipecóticos/uso terapêutico , Fases do Sono , Tiagabina , Ácido Valproico/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...