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

RESUMEN

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.


Asunto(s)
Hepatopatías/metabolismo , Piperazinas/farmacocinética , Inhibidores de Agregación Plaquetaria/farmacocinética , Tiofenos/farmacocinética , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Piperazinas/efectos adversos , Piperazinas/farmacología , Clorhidrato de Prasugrel , Tiofenos/efectos adversos , Tiofenos/farmacología
2.
J Clin Pharm Ther ; 34(5): 585-94, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19744014

RESUMEN

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.


Asunto(s)
Enfermedades Renales/metabolismo , Fallo Renal Crónico/metabolismo , Piperazinas/farmacocinética , Inhibidores de Agregación Plaquetaria/farmacocinética , Tiofenos/farmacocinética , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Piperazinas/efectos adversos , Piperazinas/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Clorhidrato de Prasugrel , Unión Proteica , Tiofenos/efectos adversos , Tiofenos/farmacología
3.
Clin Pharmacol Ther ; 81(5): 735-41, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17361128

RESUMEN

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.


Asunto(s)
Inhibidores del Citocromo P-450 CYP3A , Inhibidores Enzimáticos/farmacología , Cetoconazol/farmacología , Piperazinas/farmacocinética , Inhibidores de Agregación Plaquetaria/farmacocinética , Tiofenos/farmacocinética , Ticlopidina/análogos & derivados , Adulto , Área Bajo la Curva , Clopidogrel , Estudios Cruzados , Método Doble Ciego , Interacciones Farmacológicas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Piperazinas/administración & dosificación , Piperazinas/farmacología , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/farmacología , Clorhidrato de Prasugrel , Tiofenos/administración & dosificación , Tiofenos/farmacología , Ticlopidina/administración & dosificación , Ticlopidina/farmacocinética , Ticlopidina/farmacología
4.
Clin Pharmacol Ther ; 69(3): 96-103, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11240972

RESUMEN

BACKGROUND AND OBJECTIVE: Nefazodone inhibits CYP3A; therefore coadministration with CYP3A substrates such as terfenadine or loratadine may result in increased exposure to these drugs. A potential pharmacodynamic consequence is electrocardiographic QTc prolongation, which has been associated with torsade de pointes cardiac arrhythmia. Therefore a clinical pharmacokinetic-pharmacodynamic evaluation of this potential interaction was conducted. METHODS: A randomized, double-blind, double-dummy, parallel group, multiple-dose design was used. Healthy men and women who were given doses of 60 mg of terfenadine every 12 hours, 20 mg of loratadine once daily, and 300 mg of nefazodone every 12 hours were studied. Descriptive pharmacokinetics (time to maximum concentration, maximum concentration, and area under the plasma concentration-time curve) were used for the examination of interactions among the respective parent drugs and metabolites. QTc prolongation (mean value over the dosing interval) was the pharmacodynamic parameter measured. Kinetic and dynamic analysis was used for the examination of pooled concentration and QTc data with the use of a linear model. RESULTS: Concomitant nefazodone treatment markedly increased the dose interval area under the plasma concentration-time curve of both terfenadine (mean value, 17.3 +/- 8.5 ng. mL/h versus 97.4 +/- 48.9 ng. mL/h; P <.001) and carboxyterfenadine (mean value, 1.69 +/- 0.48 microg. h/mL versus 2.88 +/- 0.53 microg. h/mL; P <.001) and moderately increased the dose interval area under the plasma concentration-time curve of both loratadine (mean value, 31.5 +/- 27.9 ng. h/mL versus 43.7 +/- 25.9 ng. h/mL; P <.014) and descarboethoxyloratadine (mean value, 73.4 +/- 54.9 ng. h/mL versus 81.9 +/- 26.2 ng. h/mL; P <.002). The mean QTc was unchanged with terfenadine alone; however, it was markedly prolonged with concomitant nefazodone and terfenadine (mean [90% confidence interval] prolongation 42.4 ms [34.2, 50.6 ms]; P <.05). Similarly, the mean QTc was unchanged with loratadine alone; however, it was prolonged with concomitant nefazodone and loratadine (21.6 ms [13.7, 29.4 ms]; P <.05). Nefazodone alone did not change mean QTc. QTc was positively correlated with terfenadine plasma concentration (r (2) = 0.21; P =.0001). Similarly, QTc was positively correlated with loratadine plasma concentration (r (2) = 0.056; P =.0008) but with a flatter slope. There was no relationship between QTc and nefazodone plasma concentration during treatment with nefazodone alone (r (2) = 0.002, not significant). CONCLUSIONS: In healthy men and women, concomitant nefazodone treatment at a therapeutic dose increases exposure to both terfenadine and carboxyterfenadine. This increased exposure is associated with marked QTc prolongation, which is correlated with terfenadine plasma concentration. A similar interaction occurs with loratadine, although it is of lesser magnitude. Concomitant administration of nefazodone with terfenadine may have predisposed individuals to the arrhythmia associated with QTc prolongation, torsade de pointes, when terfenadine was available for clinical use. However, a new finding is that in the context of higher than clinically recommended daily doses (20 mg) of loratadine concomitant administration with a metabolic inhibitor such as nefazodone can also result in QTc prolongation.


Asunto(s)
Antidepresivos de Segunda Generación/farmacología , Electrocardiografía/efectos de los fármacos , Antagonistas de los Receptores Histamínicos H1/farmacocinética , Loratadina/farmacocinética , Terfenadina/farmacocinética , Triazoles/farmacología , Adulto , Antidepresivos de Segunda Generación/farmacocinética , Área Bajo la Curva , Método Doble Ciego , Interacciones Farmacológicas , Femenino , Antagonistas de los Receptores Histamínicos H1/farmacología , Humanos , Modelos Lineales , Loratadina/farmacología , Masculino , Persona de Mediana Edad , Piperazinas , Terfenadina/farmacología , Triazoles/farmacocinética
5.
Am J Med ; 84(6): 1053-60, 1988 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-3376975

RESUMEN

Existing methods for predicting creatinine clearance provide accurate estimates for normal-weight patients but not for patients who are obese. Studies into this problem began with an animal model of obesity, the obese overfed rat. Mean creatinine clearance was found to vary in direct proportion to fat-free body mass, determined in both obese and normal animals. The relevance of this observation to renal function in humans was evaluated by analyzing published studies reporting creatinine clearance and creatinine excretion rates in obese and normal persons. Measured creatinine clearance correlated well with estimated fat-free body mass (r = 0.772, p less than 0.02), and urinary excretion of creatinine normalized to fat-free mass correlated impressively with age (r = 0.960). Formulas derived from these observations allow for the prediction of creatinine clearance at steady state: (formula; see text) In initial tests of these formulas, their predictions appeared to be as accurate as existing methods for the normal-weight population and far superior to these methods when applied to the obese population. Therefore, when creatinine clearance is not measured in obese patients, the estimation of this parameter with the proposed formulas should improve the ability to select the appropriate dose for drugs that are cleared principally by renal filtration.


Asunto(s)
Composición Corporal , Creatinina/metabolismo , Riñón/metabolismo , Obesidad/metabolismo , Tejido Adiposo/patología , Animales , Masculino , Tasa de Depuración Metabólica , Obesidad/patología , Ratas , Ratas Endogámicas
6.
J Clin Pharmacol ; 39(7): 685-94, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10392323

RESUMEN

Avitriptan, a selective 5-HT1-like receptor agonist, is an effective compound for the treatment of migraine headaches with a prolonged duration of response. A double-blind, placebo-controlled, parallel-group, ascending-dose study in 24 healthy subjects was designed to assess the safety, tolerance, pharmacokinetics, and pharmacodynamics of avitriptan. This antimigraine drug was administered as two consecutive constant-rate IV infusions at three dose levels (12.7, 25.3, and 38.0 mg), which were targeted to produce plasma concentrations in and above the therapeutic range. The best fitting of the plasma concentration-time data was obtained by using a triexponential function yielding a terminal t1/2 of 8 hours. The areas under the plasma concentration versus time curves were proportional to dose, indicating linear pharmacokinetics. Moreover, the clearance and steady-state volume of distribution values were independent of the dose. The change in pulse rate and supine systolic and diastolic blood pressure was determined as pharmacodynamic effects of avitriptan. A "threshold log-linear" model, which accounts for the linear increase in pharmacodynamic effect with the log of plasma concentrations when the latter was higher than a certain threshold value, adequately described the pharmacodynamic data. The threshold plasma drug concentrations for the pulse rate and the diastolic and systolic blood pressure were 14, 74, and 161 ng/ml, respectively. Overall, avitriptan has consistent, linear pharmacokinetics and increases systolic and diastolic blood pressure in a predictable manner at a higher plasma concentration. However, this drug does not produce a significant change in pulse rate at the dose levels (12.7-38 mg) evaluated in this study.


Asunto(s)
Indoles/farmacocinética , Agonistas de Receptores de Serotonina/farmacocinética , Sulfonamidas/farmacocinética , Adulto , Ansiedad/inducido químicamente , Área Bajo la Curva , Presión Sanguínea/efectos de los fármacos , Diástole , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Electrocardiografía , Femenino , Humanos , Indoles/efectos adversos , Infusiones Intravenosas , Masculino , Parestesia/inducido químicamente , Enfermedades Faríngeas/inducido químicamente , Presión , Sensación/efectos de los fármacos , Agonistas de Receptores de Serotonina/efectos adversos , Sulfonamidas/efectos adversos , Sístole , Triptaminas
7.
J Clin Pharmacol ; 37(9): 799-809, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9549633

RESUMEN

The objective of this study was to assess the pharmacokinetics and pharmacodynamics of the dextro (d-) isomer of sotalol, a class III antiarrhythmic agent, in healthy young men and women after a single intravenous bolus dose. The design was open-label, randomized, parallel group. Each group (4 men and 4 women) received either 0.5, 1.5, or 3.0 mg/kg d-sotalol as an intravenous infusion for 2 minutes. Serial measurements of the d-sotalol plasma concentration and the Q-Tc interval data were recorded before, during, and for 72 hours after drug administration. The pharmacokinetics of d-sotalol were found to be well described by a three-compartment model with linear elimination clearance from the central compartment. There were no significant differences in the elimination clearance or volume of the central compartment between dose levels or between men and women. However, women were found to have a lower steady-state volume of distribution than men (1.20 L/Kg versus 1.43 L/Kg). The Q-Tc versus d-sotalol plasma concentration data were fitted to a model that assumed a distinct "effect compartment" and sigmoidal Emax response. The baseline Q-Tc, determined from the fittings, was found to be significantly higher in women (0.40 versus 0.38 seconds). The effect compartment clearance was found to be highly variable, with a median of 12.3 (range, 0.2-671,300) L/h. There were statistically significant differences in the effect compartment clearance by dose among men and by gender at a dose of 1.5 mg/kg. There were no significant differences detected between dose groups or genders for the d-sotalol effect site concentration at one half the maximum Q-Tc prolongation from baseline (EC50), EMAX, (the maximum Q-Tc prolongation from baseline) or the Hill coefficient. In conclusion, the pharmacokinetics of d-sotalol after intravenous administration are independent of dose and gender, because the difference between men and women in volume of distribution at steady-state is not clinically significant. The pharmacodynamics of Q-Tc prolongation produced by d-sotalol appear to be independent of dose and gender; however, there is considerable variability in the time course of effects on Q-Tc between individuals.


Asunto(s)
Antiarrítmicos/farmacología , Electrocardiografía/efectos de los fármacos , Sotalol/farmacología , Adolescente , Adulto , Femenino , Humanos , Infusiones Intravenosas , Masculino , Sotalol/administración & dosificación , Sotalol/farmacocinética
8.
J Clin Pharmacol ; 35(7): 730-8, 1995 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7560254

RESUMEN

Nefazodone, an antidepressant with serotonin and norepinephrine receptor modulating activity, is highly protein bound and eliminated by oxidative metabolism. This study evaluated the potential for clinically significant drug interactions with warfarin and nefazodone coadministration. Eighteen subjects received warfarin daily for 14 days, achieving steady-state warfarin concentrations and a stable prothrombin ratio. Nefazodone 200 mg every 12 hours (n = 12) or placebo every 12 hours (n = 6) was then added to the daily warfarin dose for the next 7 days in a double-blind, randomized design. No serious or unexpected adverse events or events suggestive of abnormal bleeding occurred during coadministration. The addition of nefazodone had no effect on the unbound fraction of total warfarin in plasma or on the steady-state pharmacokinetics of R-warfarin based on within-subject or comparison to placebo-treated subjects. The steady-state AUCTAU over the dosing interval and Cmax of S-warfarin decreased by 12%; however, this change is clinically insignificant because the prothrombin ratio and bleeding time remained unchanged. The steady-state minimum concentrations for nefazodone and metabolites, achieved on coadministration day 3, were typical of healthy men treated with this nefazodone dosage. In conclusion, warfarin and nefazodone coadministration was safe and well-tolerated with no clinically significant interactions.


Asunto(s)
Anticoagulantes/farmacología , Antidepresivos/farmacología , Triazoles/farmacología , Warfarina/farmacología , Adulto , Anticoagulantes/administración & dosificación , Anticoagulantes/farmacocinética , Antidepresivos/administración & dosificación , Antidepresivos/farmacocinética , Método Doble Ciego , Esquema de Medicación , Interacciones Farmacológicas , Humanos , Masculino , Piperazinas , Comprimidos , Triazoles/administración & dosificación , Triazoles/farmacocinética , Warfarina/administración & dosificación , Warfarina/farmacocinética
9.
J Clin Pharmacol ; 35(11): 1109-18, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8626885

RESUMEN

Potential interactions between nefazodone (200 mg every 12 hours) and propranolol (40 mg every 12 hours) were assessed in 18 healthy male volunteers in an open-label, randomized, three-way crossover study. The nature, frequency, and severity of adverse events during coadministration of nefazodone and propranolol were similar to those observed with either treatment alone. There were no clinically significant effects on vital signs, electrocardiographic results, or laboratory parameters. With coadministration, the maximum peak concentration (Cmax) and area under the concentration-time curve over the dosing interval (AUC tau) of propranolol decreased 29% and 14%, respectively; Cmax and AUC tau of 4-hydroxy-propranolol decreased 15% and 21%, respectively. Despite decreased plasma concentrations of the beta-antagonists, the reduction in exercise-induced tachycardia and post-exercise double product was slightly greater with coadministration than with propranolol alone. Administration of nefazodone alone did not significantly affect either pharmacologic parameter. The pharmacokinetics of nefazodone and its metabolites were largely unaffected during coadministration. Coadministration of propranolol and nefazodone results in modest pharmacokinetic inequivalencies, but no clinically significant alterations of the pharmacodynamics of propranolol.


Asunto(s)
Agonistas Adrenérgicos beta/farmacocinética , Antidepresivos de Segunda Generación/farmacocinética , Propranolol/farmacocinética , Triazoles/farmacocinética , Adolescente , Agonistas Adrenérgicos beta/administración & dosificación , Agonistas Adrenérgicos beta/sangre , Adulto , Antidepresivos de Segunda Generación/administración & dosificación , Antidepresivos de Segunda Generación/sangre , Antidepresivos de Segunda Generación/farmacología , Estudios Cruzados , Interacciones Farmacológicas , Quimioterapia Combinada , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino , Piperazinas , Propranolol/administración & dosificación , Propranolol/sangre , Triazoles/administración & dosificación , Triazoles/sangre , Triazoles/farmacología
10.
J Clin Pharmacol ; 41(12): 1351-8, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11762563

RESUMEN

A 21-day, open-label, multisite, dose escalation study comprising three demographic groups (children, adolescents, and adults) was performed to determine the pharmacokinetics and tolerability of orally administered buspirone. Thirteen children and 12 adolescents with anxiety disorder and 14 normal healthy adults were escalated from 5 to 30 mg buspirone bid over the 3-week study. Pharmacokinetic analysis revealed that buspirone was rapidly absorbed in all study groups, reaching peak levels at about 1 hour after administration. Peak plasma buspirone concentrations (Cmax) were highest in children and lowest in adults at all three dose levels (7.5, 15, 30 mg bid). However, 1-pyrimidinylpiperazine (1-PP), the primary metabolite of buspirone, exhibited a different plasma concentration-time profile; Cmax was significantly higher in children than in either adolescents or adults at all concentrations. In addition, TAUC0-T for 1-PP was significantly higher in the children cohort relative to adolescents and adults. Buspirone was generally safe and well tolerated at doses up to 30 mg bid in adolescents and adults and most of the children. The most frequently reported adverse events in children and adolescents were lightheadedness (68%), headache (48%), and dyspepsia (20%); 2 children withdrewfrom the study at the higher doses (15 mg and 30 mg bid) due to adverse effects. In adults, the most common adverse effect was somnolence (21.4%); lightheadedness, nausea, vomiting, and diarrhea were also reported, although these were mild in intensity.


Asunto(s)
Ansiolíticos/efectos adversos , Ansiolíticos/farmacocinética , Trastornos de Ansiedad/metabolismo , Buspirona/efectos adversos , Buspirona/farmacocinética , Adolescente , Adulto , Ansiolíticos/administración & dosificación , Trastornos de Ansiedad/tratamiento farmacológico , Área Bajo la Curva , Buspirona/administración & dosificación , Niño , Electrocardiografía/efectos de los fármacos , Femenino , Semivida , Humanos , Masculino
11.
Clin Pharmacol Ther ; 91(2): 250-6, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22205195

RESUMEN

Modeling and simulation were used extensively in the development of an indication for the use of olmesartan medoxomil in pediatric patients with hypertension. Simulations based on models developed in adult patients indicated that two dose groups were sufficient to estimate a dose-response relationship, thereby reducing by one-third the number of subjects required for the phase III pediatric study. Model-based predictions for blood pressure reduction agreed with the observed results of the subsequent phase III study, showing statistically significant dose-response relationships with respect to both systolic and diastolic blood pressure. Previously established pharmacokinetic and exposure-response relationships in adults, adjusted for the influence of body weight on clearance (wt(0.80)), were confirmed in the pediatric population. Together, these findings support an olmesartan dosing recommendation in pediatric subjects aged 6 to 16 years of 10 mg for subjects weighing <35 kg and 20 mg for those weighing ≥35 kg.


Asunto(s)
Ensayos Clínicos Fase I como Asunto/estadística & datos numéricos , Ensayos Clínicos Fase III como Asunto/estadística & datos numéricos , Simulación por Computador/estadística & datos numéricos , Imidazoles/uso terapéutico , Modelos Estadísticos , Tetrazoles/uso terapéutico , Adolescente , Antihipertensivos/administración & dosificación , Antihipertensivos/uso terapéutico , Presión Sanguínea/efectos de los fármacos , Niño , Preescolar , Relación Dosis-Respuesta a Droga , Humanos , Hipertensión/tratamiento farmacológico , Imidazoles/administración & dosificación , Imidazoles/farmacocinética , Lactante , Olmesartán Medoxomilo , Tetrazoles/administración & dosificación , Tetrazoles/farmacocinética
14.
Curr Med Res Opin ; 25(8): 1821-9, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19530977

RESUMEN

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.


Asunto(s)
Antibióticos Antituberculosos/farmacología , Piperazinas/farmacología , Piperazinas/farmacocinética , Rifampin/farmacología , Tiofenos/farmacología , Tiofenos/farmacocinética , Síndrome Coronario Agudo , Adolescente , Adulto , Antibióticos Antituberculosos/administración & dosificación , Interacciones Farmacológicas , Humanos , Masculino , Persona de Mediana Edad , Piperazinas/administración & dosificación , Clorhidrato de Prasugrel , Rifampin/administración & dosificación , Tiofenos/administración & dosificación , Adulto Joven
15.
J Pharmacol Exp Ther ; 248(1): 17-22, 1989 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-2913269

RESUMEN

Obese humans suffer from excessive organ dysfunction, altered drug pharmacokinetics and may be at increased risk of various drug toxicities. A recent report shows that gentamicin nephrotoxicity in critically ill patients is more frequent and more severe than usual in individuals who are substantially overweight. The present study utilizes an overfed rat model to examine the influence of obesity on the nephrotoxic potential of gentamicin. After 52 weeks on an energy-dense diet, obese animals outweighed pellet controls by more than 80% (913 +/- 86 vs. 507 +/- 52 g; X +/- S.D., n = 7). When animals were treated twice daily for 6 days with 30 mg/kg of gentamicin i.p. based on total body mass, obese rats sustained more cortical necrosis than control (median score 3+ vs. 0), higher serum creatinine (4.36 +/- 2.72 vs. 0.71 +/- 0.17) and greater creatinine adjusted N-acetyl hexosaminidase excretion. The impact of obesity on intrinsic susceptibility to gentamicin nephrotoxicity was assessed by dosing animals for 5 days to ideal body mass plus 40% of excess body mass, the current clinical practice for achieving normal gentamicin concentrations in obese patients. Obese rats again sustained more frequent and severe cortical necrosis (2+ vs. 0) and excreted more N-acetyl hexosaminidase than control animals. Urine pH averaged 1.7 U below normal in obese animals, but restoration to normal values by 2 weeks on the pellet diet did not diminish the toxicity increase. Results from the overfed rat closely resemble the recent clinical observation that obese patients sustain more frequent and severe kidney damage from aminoglycoside antibiotics.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Gentamicinas/toxicidad , Riñón/efectos de los fármacos , Obesidad/complicaciones , Acetilglucosaminidasa/orina , Animales , Gentamicinas/metabolismo , Concentración de Iones de Hidrógeno , Riñón/metabolismo , Riñón/patología , Masculino , Obesidad/metabolismo , Obesidad/patología , Ratas , Ratas Endogámicas , Factores de Riesgo
16.
Biochem Biophys Res Commun ; 157(1): 315-20, 1988 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-3196341

RESUMEN

The obese overfed rat effectively models many of the pharmacological changes in human obesity. Recent data show that the obese rat is unusually susceptible to liver damage by several metabolically activated drugs that may be more toxic in obese humans. Results of the present study suggest a specific molecular locus for this interaction. In obese rats, P450 content of liver and the microsomal concentration of P450 were elevated 88% and 31%, respectively, over nonobese controls. Increases in microsomal ethanol oxidation were of identical magnitude. The ethanol-inducible form of P450 that is responsible for microsomal ethanol oxidation, P450IIE1, bioactivates several drugs that are shown to cause increased injury in obese rats. Collectively, these findings indicate that specific forms of P450 may become up-regulated in obesity, increasing the risk of a biochemically defined spectrum of drug-induced organ injuries.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Etanol/metabolismo , Microsomas Hepáticos/enzimología , Oxigenasas de Función Mixta/metabolismo , Obesidad/enzimología , Alcohol Deshidrogenasa/metabolismo , Aldehído Deshidrogenasa/metabolismo , Animales , Peso Corporal , Oxidación-Reducción , Ratas , Factores de Riesgo
17.
Int J Obes ; 13(1): 69-79, 1989.
Artículo en Inglés | MEDLINE | ID: mdl-2703295

RESUMEN

The present study was undertaken to examine the appropriateness of the obese overfed rat and the obese Zucker rat as animal models for evaluating drug disposition changes in human obesity. It was found that 11 of 12 characteristics that control or influence drug clearance and volume of drug distribution in obese humans were qualitatively reproduced in the obese overfed rat. In contrast, existing literature shows that the obese Zucker rat resembles the obese human in only five of 12 characteristics, with meaningful discrepancies in fat-free mass, creatinine clearance, and thyroid function. Perhaps of greatest significance were changes in hepatic cytochrome P-450, which increased in proportion to total body mass in the obese overfed rat but remain unchanged in the Zucker rat. Although P-450 status in human obesity is unknown, the overfed rat model provides an opportunity for examining increased oxidative drug elimination that appears as an established feature of human obesity. In conclusion, the obese overfed rat appears to be superior to the obese Zucker rat as an animal model for evaluating the pharmacological consequences of human obesity, particularly those in which reproducing drug pharmacokinetics is an important consideration.


Asunto(s)
Modelos Animales de Enfermedad , Obesidad/metabolismo , Farmacocinética , Ratas Endogámicas/metabolismo , Ratas Mutantes/metabolismo , Ratas Zucker/metabolismo , Animales , Ingestión de Alimentos , Tasa de Depuración Metabólica , Ratas , Distribución Tisular
18.
Toxicol Appl Pharmacol ; 98(1): 12-24, 1989 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-2467406

RESUMEN

Effects of the diuretic drug furosemide were examined in obese animals to evaluate the hypothesis that organ damage by reactive drug metabolites may be potentiated by this disease. Obese overfed Sprague-Dawley rats that were treated ip with 450 mg/kg furosemide on the basis of total body mass suffered a 58% mortality rate over 24 h. This contrasted with 0% mortality in animals of normal body mass. On the basis of median histopathology scores, organ necrosis was judged to be greater in the liver (2+) and kidneys (1+) of obese rats than in the liver (1+) and kidneys (less than 1+) of normal controls (p less than 0.05). Obese animals demonstrated a fourfold rise in fat mass over controls. The low solubility of furosemide in lipid makes it probable that aggravated drug toxicity in obese rats dosed to total body mass resulted in part from elevated furosemide concentrations in lean body mass. In a subsequent study designed to minimize this possibility, furosemide was administered on the basis of fat-free body mass to equalize initial drug exposure in obese and control rats. Even with this downward dosage adjustment, obese animals suffered increased hepatic necrosis (median score of 2+ versus 0 in treated controls), greater impairment of renal function (plasma creatinine concentration of 2.41 mg/dl versus 0.96 mg/dl in treated controls), and more extensive enzymuria (enzyme excretion 175-300% more elevated than in treated controls). In conclusion, obese rats appear to be at increased risk of furosemide-induced liver and kidney injury due to at least two factors: (1) increased exposure of target organs in lean body mass to furosemide when the dosing of this poorly lipophilic drug was based on total body mass, and (2) increased susceptibility of target organs in lean body mass to furosemide injury when dosing was adjusted downward to reflect fat-free body mass and to equalize initial drug exposure.


Asunto(s)
Furosemida/toxicidad , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Obesidad/fisiopatología , Animales , Dieta , Furosemida/farmacocinética , Riñón/patología , Hígado/patología , Masculino , Obesidad/sangre , Ratas , Ratas Endogámicas , Factores de Riesgo , Coloración y Etiquetado
19.
Biochem Biophys Res Commun ; 149(1): 203-7, 1987 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-2446617

RESUMEN

The status of Na+ regulation was examined during early stages of alkylation insult to rat liver. Na+/K+-ATPase activity in plasma membranes declined by 52% within 3 hr of treatment with 850 mg/kg acetaminophen. This loss preceded the release of alanine aminotransferase (2880 +/- 1550 U/ml) and necrosis (2+) seen at 24 hr. Activities of 5'-nucleotidase and Mg2+-ATPase and recovery of plasma membranes were comparatively unchanged at 3 hr. Because damage to Na+/K+-ATPase appeared early in the pathogenesis of acetaminophen hepatotoxicity, loss of hepatocellular Na+ regulation could represent one of the critical molecular consequences of lethal alkylation by acetaminophen.


Asunto(s)
Acetaminofén/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas , Canales Iónicos/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , 5'-Nucleotidasa , Acetaminofén/farmacología , Alquilación , Animales , Membrana Celular/enzimología , Hígado/enzimología , Hepatopatías/enzimología , Masculino , Nucleotidasas/metabolismo , Ratas , Ratas Endogámicas , Sodio/metabolismo
20.
Drug Metab Dispos ; 20(3): 402-6, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1355715

RESUMEN

Obese human patients and obese overfed rats treated chronically with gentamicin suffer greater renal injury than nonobese patients and control animals. To understand the mechanism of this heightened susceptibility to the nephrotoxic effects of gentamicin, this study examines the plasma-time course and renal uptake of gentamicin in control and obese overfed rats following a single bolus dose. Gentamicin was administered ip to control rats at 30 mg/kg total body mass and to obese rats at 30 mg/kg ideal body mass plus 40% of excess body mass. Following gentamicin dosing, only 1 of 11 concentration-time points taken over 6/hr postdosing was different between control and obese groups. In addition, the area under the plasma concentration curve extrapolated to infinite time was not different between obese and control rats (mean +/- SD of 4.47 +/- 0.85 vs. 4.13 +/- 0.35 mg.min.ml-1, p greater than 0.5). The gentamicin plasma concentrations after 6 hr were less than 1 microgram/ml and not different between the groups; however, the concentration of gentamicin in the kidneys was 33% greater in obese than control rats at this time (324 +/- 66.9 vs. 244 +/- 34.7 micrograms/g, p less than 0.05). The fraction of dose and the total amount of drug that accumulated in the kidneys were also greater in the obese rats (42 and 72% increases). Considered with the results of previous studies, it appears that obese overfed rats sustain more severe nephrotoxicity following comparable plasma gentamicin exposure because of increased renal uptake and/or retention of drug.


Asunto(s)
Gentamicinas/farmacocinética , Riñón/efectos de los fármacos , Obesidad/metabolismo , Absorción , Animales , Peso Corporal/efectos de los fármacos , Creatinina/sangre , Gentamicinas/sangre , Gentamicinas/toxicidad , Riñón/metabolismo , Masculino , Ratas , Ratas Endogámicas , Análisis de Regresión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA