Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Pharm ; 523(1): 343-356, 2017 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-28330645

RESUMEN

The aim of this study was to investigate a fixed dose combination (FDC) of telmisartan (TEL) and pravastatin sodium (PRA) in enteric-coated bilayer tablets, which was designed for once-daily bedtime dose in order to match circadian rhythmic variations of hypertension and cholesterol synthesis and optimize the patient friendly dosing treatment. Due to the poor aqueous solubility of TEL, ternary solid dispersions (SD) consisting of TEL, polyethylene glycol 6000 (PEG 6000) and magnesium oxide (MgO) were designed to enhance its dissolution rate in intestinal fluid. MgO was added as an effective alkalizer to maintain the high microenvironmental pH of the saturated solution in the immediate vicinity of TEL particles because TEL is known to be ionizable but poorly soluble in intestinal fluid. In contrast, PRA is known to be very unstable in low pH conditions. In the SD system, TEL was present in an amorphous structure and formed an intermolecular hydrogen bonding with MgO, giving complete drug release without precipitation in intestinal fluid. In addition, the amount of hydrophilic carrier (PEG 6000) was also a factor. In the design of tablet formulation, the diluents and superdisintegrants could play a key role in release profiles. Then, to fulfill the unmet needs of the two model drugs and match circadian rhythmic variations of hypertension and cholesterol synthesis, enteric-coated bilayer tablet consisting of TEL SD and PRA was finally prepared using Acryl-EZE® as an enteric coating material. Prior to enteric coating, a seal coating layer (Opadry®, 2% weight gains) was firstly introduced to separate the core bilayer tablet from the acidic enteric coating polymers to avoid premature degradation. Dissolution profiles of finished tablets revealed that enteric-coated bilayer tablets with 6% weight gains remained intact in acidic media (pH 1.0) for 2h and then released drugs completely within 45min after switching to the intestinal media (pH 6.8). It was observed that enteric-coated bilayer tablets were stable during 3 month under the accelerated condition of 40°C/75% RH. The delayed drug release and bedtime dosage regimen using enteric-coated bilayer tablet containing TEL and PRA, matching the circadian rhythms of hypertension and hyperlipidemia can provide therapeutic benefits for elderly patients in terms of maximizing the therapeutic effects.


Asunto(s)
Anticolesterolemiantes/química , Antihipertensivos/química , Bencimidazoles/química , Benzoatos/química , Pravastatina/química , Anticolesterolemiantes/administración & dosificación , Antihipertensivos/administración & dosificación , Bencimidazoles/administración & dosificación , Benzoatos/administración & dosificación , Cronoterapia de Medicamentos , Combinación de Medicamentos , Liberación de Fármacos , Estabilidad de Medicamentos , Difracción de Polvo , Pravastatina/administración & dosificación , Comprimidos Recubiertos , Telmisartán , Difracción de Rayos X
2.
PLoS One ; 9(10): e106570, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25329899

RESUMEN

Statins, 3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitors, are associated with the prevention of atrial fibrillation (AF) by pleiotropic effects. Recent clinical trial studies have demonstrated conflicting results on anti-arrhythmia between lipophilic and hydrophilic statins. However, the underlying mechanisms responsible for anti-arrhythmogenic effects of statins are largely unexplored. In this study, we evaluated the different roles of lipophilic and hydrophilic statins (simvastatin and pravastatin, respectively) in acetylcholine (100 µM)-activated K+ current (IKACh, recorded by nystatin-perforated whole cell patch clamp technique) which are important for AF initiation and maintenance in mouse atrial cardiomyocytes. Our results showed that simvastatin (1-10 µM) inhibited both peak and quasi-steady-state IKACh in a dose-dependent manner. In contrast, pravastatin (10 µM) had no effect on IKACh. Supplementation of substrates for the synthesis of cholesterol (mevalonate, geranylgeranyl pyrophosphate or farnesyl pyrophosphate) did not reverse the effect of simvastatin on IKACh, suggesting a cholesterol-independent effect on IKACh. Furthermore, supplementation of phosphatidylinositol 4,5-bisphosphate, extracellular perfusion of phospholipase C inhibitor or a protein kinase C (PKC) inhibitor had no effect on the inhibitory activity of simvastatin on IKACh. Simvastatin also inhibits adenosine activated IKACh, however, simvastatin does not inhibit IKACh after activated by intracellular loading of GTP gamma S. Importantly, shortening of the action potential duration by acetylcholine was restored by simvastatin but not by pravastatin. Together, these findings demonstrate that lipophilic statins but not hydrophilic statins attenuate IKACh in atrial cardiomyocytes via a mechanism that is independent of cholesterol synthesis or PKC pathway, but may be via the blockade of acetylcholine binding site. Our results may provide important background information for the use of statins in patients with AF.


Asunto(s)
Acetilcolina/farmacología , Fibrilación Atrial/prevención & control , Fenómenos Electrofisiológicos/efectos de los fármacos , Atrios Cardíacos/citología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Miocitos Cardíacos/efectos de los fármacos , Potasio/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Atrios Cardíacos/fisiopatología , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Miocitos Cardíacos/citología , Pravastatina/química , Pravastatina/farmacología , Simvastatina/química , Simvastatina/farmacología
3.
J Pharmacol Exp Ther ; 347(1): 193-202, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23926287

RESUMEN

We developed a pravastatin derivative, sodium (3R,5R)-3,5-dihydroxy-7-((1S,2S,6S,8S)-6-hydroxy-2-methyl-8-((1-[(11)C]-(E)-2-methyl-but-2-enoyl)oxy)-1,2,6,7,8,8a-hexahydronaphthalen-1-yl)heptanoate ([(11)C]DPV), as a positron emission tomography (PET) probe for noninvasive measurement of hepatobiliary transport, and conducted pharmacokinetic analysis in rats as a feasibility study for future clinical study. Transport activities of DPV in freshly isolated rat hepatocytes and rodent multidrug resistance-associated protein 2 (rMrp2; human, MRP2)-expressing membrane vesicles were similar to those of pravastatin. Rifampicin diminished the uptake of DPV and pravastatin by the hepatocytes, with similar inhibition potency. [(11)C]DPV underwent biotransformation to produce at least two metabolites in rat, but metabolism of [(11)C]DPV occurred negligibly in human hepatocytes during a 90-minute incubation. After intravenous injection, [(11)C]DPV was mainly distributed to the liver and kidneys, where the tissue uptake clearances (CLuptake,liver and CLuptake,kidney) were blood-flow-limited (73.6 ± 4.8 and 24.6 ± 0.6 ml/min per kilogram, respectively). Systemic elimination of [(11)C]DPV was delayed in rifampicin-treated rat and an Mrp2-deficient mutant rat, Eisai hyperbilirubinemic mutant rat (EHBR). Rifampicin treatment decreased both CLuptake,liver and CLuptake,kidney of [(11)C]DPV by 30% (P < 0.05), whereas these parameters were unchanged in EHBR. Meanwhile, the canalicular efflux clearance (CLint,bile) of [(11)C]DPV, which was 12.2 ± 1.5 ml/min per kilogram in the control rat, decreased by 60% and 89% in rifampicin-treated rat and EHBR (P < 0.05), respectively. These results indicate that [(11)C]DPV is taken up into the liver by organic anion-transporting polypeptides (rodent, Oatps; human, OATP) and excreted into bile by Mrp2 in rat, and that rifampicin may inhibit Mrp2 as well as Oatps, and consequently increase systemic exposure of [(11)C]DPV. PET using [(11)C]DPV is feasible for studies prior to the future clinical investigation of OATP and MRP2 functionality, especially for personalized medicine.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Sistema Biliar/metabolismo , Hepatocitos/metabolismo , Transportadores de Anión Orgánico/metabolismo , Tomografía de Emisión de Positrones , Pravastatina/metabolismo , Transportadoras de Casetes de Unión a ATP/fisiología , Animales , Sistema Biliar/diagnóstico por imagen , Radioisótopos de Carbono/metabolismo , Técnicas de Cocultivo , Evaluación Preclínica de Medicamentos/métodos , Estudios de Factibilidad , Hepatocitos/diagnóstico por imagen , Humanos , Masculino , Transportadores de Anión Orgánico/fisiología , Tomografía de Emisión de Positrones/métodos , Pravastatina/química , Ratas , Ratas Sprague-Dawley
4.
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
5.
Ann Ital Med Int ; 10 Suppl: 35S-42S, 1995 Oct.
Artículo en Italiano | MEDLINE | ID: mdl-8562263

RESUMEN

With the increasing knowledge on the pathogenesis of atherosclerosis, it appears that the prevention of cardiovascular disease in the future will involve, besides risk factor correction, direct pharmacological control of processes occurring in the arterial wall. Among them, a pivotal role is played by smooth muscle cell migration and proliferation that, together with lipid deposition, are prominent features of atherogenesis and restenosis after angioplasty. Mevalonate and other intermediates (isoprenoids) of cholesterol synthesis are essential for cell growth, hence drugs affecting this metabolic pathway are potential antiatherosclerotic agents. Recently we provided evidence that fluvastatin, simvastatin, lovastatin, but not pravastatin, dose-dependently decrease smooth muscle cell migration and proliferation, independently of their hypocholesterolemic properties. The in vitro inhibition of cell migration and proliferation induced by simvastatin and fluvastatin (70-90% decrease) was completely prevented by the addition of mevalonate and partially (80%) by farnesol and geranylgeraniol, confirming the specific role of isoprenoid metabolites--probably through prenylated proteins--in regulating these cellular events. The present results provide evidence that HMG-CoA reductase inhibitors interfere directly with processes involved in atherogenesis--beyond their effects on plasma lipids--partially through local inhibition of isoprenoid biosynthesis.


Asunto(s)
Arteriosclerosis/tratamiento farmacológico , Ácidos Grasos Monoinsaturados/farmacología , Indoles/farmacología , Lovastatina/análogos & derivados , Lovastatina/farmacología , Pravastatina/farmacología , Animales , Arteriosclerosis/patología , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Ácidos Grasos Monoinsaturados/química , Ácidos Grasos Monoinsaturados/uso terapéutico , Fluvastatina , Indoles/química , Indoles/uso terapéutico , Lovastatina/química , Lovastatina/uso terapéutico , Pravastatina/química , Pravastatina/uso terapéutico , Ratas , Ratas Sprague-Dawley , Simvastatina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA