Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Pharm Biol ; 60(1): 359-373, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35171063

RESUMEN

CONTEXT: Patients with non-alcoholic steatohepatitis (NASH) may have a simultaneous intake of pravastatin and evodiamine-containing herbs. OBJECTIVE: The effect of evodiamine on the pharmacokinetics of pravastatin and its potential mechanisms were investigated in NASH rats. MATERIALS AND METHODS: The NASH model was conducted with feeding a methionine choline-deficient (MCD) diet for 8 weeks. Sprague-Dawley rats were randomised equally (n = 6) into NASH group, evodiamine group (10 mg/kg), pravastatin group (10 mg/kg), and evodiamine (10 mg/kg) + pravastatin (10 mg/kg) group. Normal control rats were fed a standard diet. Effects of evodiamine on the pharmacokinetics, distribution, and uptake of pravastatin were investigated. RESULTS: Evodiamine decreased Cmax (159.43 ± 26.63 vs. 125.61 ± 22.17 µg/L), AUC0-t (18.17 ± 2.52 vs. 14.91 ± 2.03 mg/min/L) and AUC0-∞ (22.99 ± 2.62 vs. 19.50 ± 2.31 mg/min/L) of orally administered pravastatin in NASH rats, but had no significant effect in normal rats. Evodiamine enhanced the uptake (from 154.85 ± 23.17 to 198.48 ± 26.31 pmol/mg protein) and distribution (from 736.61 ± 108.07 to 911.89 ± 124.64 ng/g tissue) of pravastatin in NASH rat liver. The expression of Oatp1a1, Oatp1a4, and Oatp1b2 was up-regulated 1.48-, 1.38-, and 1.51-fold by evodiamine. Evodiamine decreased the levels of IL-1ß, IL-6, and TNF-α by 27.82%, 24.76%, and 29.72% in NASH rats, respectively. DISCUSSION AND CONCLUSIONS: Evodiamine decreased the systemic exposure of pravastatin by up-regulating the expression of OATPs. These results provide a reference for further validation of this interaction in humans.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Transportadores de Anión Orgánico/genética , Pravastatina/farmacocinética , Quinazolinas/farmacología , Animales , Área Bajo la Curva , Interacciones de Hierba-Droga , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Masculino , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos
2.
Expert Opin Drug Metab Toxicol ; 9(4): 459-72, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23331046

RESUMEN

INTRODUCTION: Membrane transporters have been recognized to play a key role in determining the absorption, distribution and elimination processes of drugs. The organic anion-transporting polypeptide (OATP)1B1 and OATP1B3 isoforms are selectively expressed in the human liver and are known to cause significant drug-drug interactions (DDIs), as observed with an increasing number of drugs. It is evident that DDIs involving hepatic transporters are capable of altering systemic, as well as tissue-specific, exposure of drug substrates resulting in marked differences in drug safety and/or efficacy. It is therefore essential to quantitatively predict such interactions early in the drug development to mitigate clinical risks. AREAS COVERED: The role of hepatic uptake transporters in drug disposition and clinical DDIs has been reviewed with an emphasis on the current state of the models applicable for quantitative predictions. The readers will also gain insight into the in vitro experimental tools available to characterize transport kinetics, while appreciating the knowledge gaps in the in vitro-in vivo extrapolation (IVIVE), which warrant further investigation. EXPERT OPINION: Static and dynamic models can be convincingly applied to quantitatively predict drug interactions, early in drug discovery, to mitigate clinical risks as well as to avoid unnecessary clinical studies. Compared to basic models, which focus on individual processes, mechanistic models provide the ability to assess DDI potential for compounds with systemic disposition determined by both transporters and metabolic enzymes. However, complexities in the experimental tools and an apparent disconnect in the IVIVE of transport kinetics have limited the physiologically based pharmacokinetic modeling strategies. Emerging data on the expression of transporter proteins and tissue drug concentrations are expected to help bridge these gaps. In addition, detailed characterization of substrate kinetics can facilitate building comprehensive mechanistic models.


Asunto(s)
Hígado/citología , Hígado/efectos de los fármacos , Proteínas de Transporte de Membrana/efectos de los fármacos , Línea Celular , Fenómenos Químicos , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Fluorobencenos/farmacocinética , Fluorobencenos/uso terapéutico , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Cinética , Hígado/metabolismo , Modelos Teóricos , Pravastatina/farmacocinética , Pravastatina/uso terapéutico , Pirimidinas/farmacocinética , Pirimidinas/uso terapéutico , Rosuvastatina Cálcica , Sulfonamidas/farmacocinética , Sulfonamidas/uso terapéutico
3.
AAPS PharmSciTech ; 12(2): 673-82, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21594729

RESUMEN

The purpose of research was to develop a mucoadhesive multiparticulate sustained drug delivery system of pravastatin sodium, a highly water-soluble and poorly bioavailable drug, unstable at gastric pH. Mucoadhesive microparticles were formulated using eudragit S100 and ethyl cellulose as mucoadhesive polymers. End-step modification of w/o/o double emulsion solvent diffusion method was attempted to improve the purity of the product, that can affect the dose calculations of sustained release formulations and hence bioavailability. Microparticles formed were discrete, free flowing, and exhibited good mucoadhesive properties. DSC and DRS showed stable character of drug in microparticles and absence of drug polymer interaction. The drug to polymer ratio and surfactant concentration had significant effect on mean particle size, drug release, and entrapment efficiency. Microparticles made with drug: eudragit S100 ratio of 1:3 (F6) exhibited maximum entrapment efficiency of 72.7% and ex vivo mucoadhesion time of 4.15 h. In vitro permeation studies on goat intestinal mucosa demonstrated a flux rate (1,243 µg/cm(2)/h) that was 169 times higher than the flux of pure drug. The gastric instability problem was overcome by formulating the optimized microparticles as enteric-coated capsules that provided a sustained delivery of the highly water-soluble drug for 12 h beyond the gastric region. The release mechanism was identified as fickian diffusion (n = 0.4137) for the optimized formulation F6. Conclusively, a drug delivery system was successfully developed that showed delayed and sustained release up to 12 h and could be potentially useful to overcome poor bioavailability problems associated with pravastatin sodium.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Diseño de Fármacos , Mucosa Intestinal/metabolismo , Microesferas , Pravastatina/química , Animales , Sistemas de Liberación de Medicamentos/normas , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/normas , Cabras , Mucosa Intestinal/efectos de los fármacos , Tamaño de la Partícula , Ácidos Polimetacrílicos/administración & dosificación , Ácidos Polimetacrílicos/química , Ácidos Polimetacrílicos/farmacocinética , Pravastatina/administración & dosificación , Pravastatina/farmacocinética
4.
Eur J Pharm Sci ; 41(5): 729-35, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-20933082

RESUMEN

Garlic extracts have been shown to decrease drug exposure for saquinavir, a P-glycoprotein and cytochrome P450 3A4 substrate. In order to explore the underlying mechanisms and to study the effects of garlic on pre-systemic drug elimination, healthy volunteers were administered garlic extract for 21 days. Prior to and at the end of this period, expression of duodenal P-glycoprotein and cytochrome P450 3A4 protein were assayed and normalized to villin, while hepatic cytochrome P450 3A4 function and simvastatin, pravastatin and saquinavir pharmacokinetics were also evaluated. Ingestion of garlic extract increased expression of duodenal P-glycoprotein to 131% (95% CI, 105-163%), without increasing the expression of cytochrome P450 3A4 which amounted to 87% (95% CI, 67-112%), relative to baseline in both cases. For the erythromycin breath test performed, the average result was 96% (95% CI, 83-112%). Ingestion of garlic extract had no effect on drug and metabolite AUCs following a single dose of simvastatin or pravastatin, although the average area under the plasma concentration curve (AUC) of saquinavir decreased to 85% (95% CI, 66-109%), and changes in intestinal P-glycoprotein expression negatively correlated with this change. In conclusion, garlic extract induces intestinal expression of P-glycoprotein independent of cytochrome P450 3A4 in human intestine and liver.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Citocromo P-450 CYP3A/biosíntesis , Duodeno/enzimología , Hígado/enzimología , Extractos Vegetales/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Adulto , Área Bajo la Curva , Citocromo P-450 CYP3A/metabolismo , Duodeno/efectos de los fármacos , Ajo , Regulación de la Expresión Génica , Humanos , Hígado/efectos de los fármacos , Masculino , Pravastatina/farmacocinética , Saquinavir/farmacocinética , Simvastatina/farmacocinética
5.
J Pharm Sci ; 96(2): 459-72, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17075867

RESUMEN

A preclinical canine model capable of predicting a compound's potential for a human food effect was developed. The beagle dog was chosen as the in vivo model. A validation set of compounds with known propensities for human food effect was studied. Several diets were considered including high-fat dog food and various quantities of the human FDA meal. The effect of pentagastrin pretreatment was also investigated. The high-fat dog food did not predict human food effect and was discontinued from further evaluation. The amount of FDA meal in the dog was important in the overall prediction of the magnitude of human food effect. Fed/fasted Cmax and AUC ratios using a 50-g aliquot of the FDA meal in the dog were in the closest qualitative agreement to human data. Pentagastrin pretreatment did not affect the AUC in the fed state, but increased the fasted AUC for weakly basic compounds. Pentagastrin pretreatment and a 50-g aliquot of the FDA meal in the dog predicted the human food effect for a validation set of compounds. This model, which is intended for compound screening, will be helpful for determining food effect as a liability when compounds progress from discovery to clinical development.


Asunto(s)
Grasas de la Dieta , Evaluación Preclínica de Medicamentos/métodos , Interacciones Alimento-Droga , Modelos Animales , Pentagastrina/farmacología , Animales , Sulfato de Atazanavir , Disponibilidad Biológica , Perros , Inhibidores de la Proteasa del VIH/farmacocinética , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Oligopéptidos/farmacocinética , Pravastatina/farmacocinética , Piridinas/farmacocinética
6.
Clin Pharmacol Ther ; 70(6): 518-24, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11753267

RESUMEN

OBJECTIVE: St John's Wort, a widely used herbal product, is an inducer of CYP3A4 and it decreases blood concentrations of CYP3A4 substrates. The effects of St John's Wort on the pharmacokinetics of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors simvastatin (an inactive lactone pro-drug) and pravastatin were determined in this study. METHODS: Sixteen healthy male subjects (n = 8 in group 1 and n = 8 in group 2) took a St John's Wort caplet (300 mg) or matching placebo three times a day for 14 days in a double-blind, crossover study. On day 14, a single oral dose of 10 mg simvastatin and 20 mg pravastatin was given to subjects in group 1 and group 2, respectively. Blood samples were obtained during a 24-hour period after the administration of each drug. RESULTS: Repeated St John's Wort treatment tended to lower plasma simvastatin concentration and significantly (P <.05) lowered concentrations of simvastatin hydroxy acid, its active metabolite. The peak concentration in plasma (ratio, 0.72 of placebo) of simvastatin hydroxy acid tended to be decreased and its area under the plasma concentration-time curve between time zero and 24 hours after administration (ratio, 0.48 of placebo) was significantly decreased (P <.05) by St John's Wort. On the other hand, St John's Wort did not influence plasma pravastatin concentration. No significant differences were observed in the elimination half-life of simvastatin or pravastatin between the placebo and St John's Wort trials. CONCLUSIONS: This study showed that St John's Wort decreases plasma concentrations of simvastatin but not of pravastatin. Because simvastatin is extensively metabolized by CYP3A4 in the intestinal wall and liver, which are induced by St John's Wort, it is likely that this interaction is partly caused by the enhancement of the CYP3A4-mediated first-pass metabolism of simvastatin in the small intestine and liver.


Asunto(s)
Anticolesterolemiantes/farmacocinética , Hypericum/efectos adversos , Fitoterapia/efectos adversos , Pravastatina/farmacocinética , Simvastatina/farmacocinética , Adulto , Área Bajo la Curva , Biotransformación , Cromatografía Liquida , Estudios Cruzados , Método Doble Ciego , Interacciones Farmacológicas , Femenino , Humanos , Masculino , Espectrometría de Masas
7.
J Pharmacokinet Biopharm ; 26(3): 329-47, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10098103

RESUMEN

The tissue-selective inhibition of cholesterol synthesis by pravastatin was evaluated pharmacokinetically and pharmacodynamically. Plasma, tissue, urine, and bile concentrations were measured after i.v. bolus injection of pravastatin to rats at various doses. The total body clearance and steady state volume of distribution decreased with increasing dose. A saturable biliary excretion was also observed. The time course of plasma and liver concentrations was described by a three-compartment model, consisting of a central compartment, a deep compartment with an nonsaturable uptake process, and a shallow compartment with saturable uptake and nonsaturable elimination processes. It suggests that a mechanism for the decrease in the total body clearance and distribution volume might be explained by a saturation of pravastatin uptake into the liver. Plasma concentration data after oral administration was also fitted to the same model by connecting an absorption compartment to the shallow compartment. The inhibitory activity of pravastatin against cholesterol synthesis in liver could be related to the concentration in the shallow compartment via a sigmoidal Emax model and the obtained pharmacodynamic parameters were comparable to those in vitro. Results suggest that the carrier-mediated hepatic uptake of pravastatin is actually responsible for the hepatoselective inhibition of cholesterol synthesis under physiological conditions.


Asunto(s)
Anticolesterolemiantes/farmacología , Colesterol/biosíntesis , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Pravastatina/farmacología , Administración Oral , Animales , Anticolesterolemiantes/farmacocinética , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Inyecciones Intravenosas , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Dinámicas no Lineales , Especificidad de Órganos , Pravastatina/farmacocinética , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA