Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Regul Toxicol Pharmacol ; 63(1): 29-39, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22343256

RESUMEN

Anidulafungin and voriconazole are potent antifungal agents that may provide a powerful therapeutic option over current therapies when coadministered. A non-clinical combination toxicity study was required as part of the voriconazole Paediatric Investigation Plan. Rats received anidulafungin or voriconazole alone or in combination once daily from postnatal day (PND) 21-56 with a recovery period to PND 84. Doses used were based upon the approximate adult rat no observed adverse-effect level (NOAEL). Transient and reversible reductions in bodyweight, haematology, serum chemistry, liver weight and minimal liver changes were associated with anidulafungin. Voriconazole caused an increase in gamma-glutamyltransferase in female rats only. No increased toxicity was observed with the combination. Toxicokinetics were determined using a validated dual-analyte bioanalytical method. Systemic exposure at juvenile rat NOAELs was comparable to that found with adult rats in previous studies. There were no drug-drug interactions affecting exposure of either drug. Juvenile rats were not more sensitive to each drug dosed alone compared with adult rat data on the single drugs. No novel, additive or synergistic toxicities were noted with the combination in juvenile rats. This study will support future studies of the combination of voriconazole and anidulafungin in children with invasive fungal infection.


Asunto(s)
Antifúngicos/farmacocinética , Antifúngicos/toxicidad , Equinocandinas/farmacocinética , Equinocandinas/toxicidad , Pirimidinas/farmacocinética , Pirimidinas/toxicidad , Triazoles/farmacocinética , Triazoles/toxicidad , Administración Oral , Anidulafungina , Animales , Antifúngicos/administración & dosificación , Área Bajo la Curva , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos/métodos , Equinocandinas/administración & dosificación , Femenino , Inyecciones Subcutáneas , Masculino , Pirimidinas/administración & dosificación , Ratas , Ratas Endogámicas F344 , Proyectos de Investigación , Factores Sexuales , Pruebas de Toxicidad/métodos , Triazoles/administración & dosificación , Voriconazol
2.
J Med Chem ; 52(23): 7446-57, 2009 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-19775168

RESUMEN

Respiratory tract bacterial strains are becoming increasingly resistant to currently marketed macrolide antibiotics. The current alternative telithromycin (1) from the newer ketolide class of macrolides addresses resistance but is hampered by serious safety concerns, hepatotoxicity in particular. We have discovered a novel series of azetidinyl ketolides that focus on mitigation of hepatotoxicity by minimizing hepatic turnover and time-dependent inactivation of CYP3A isoforms in the liver without compromising the potency and efficacy of 1.


Asunto(s)
Azetidinas/química , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Cetólidos/química , Cetólidos/farmacología , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Animales , Bacterias/efectos de los fármacos , Infecciones Comunitarias Adquiridas/tratamiento farmacológico , Susceptibilidad a Enfermedades , Descubrimiento de Drogas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Cetólidos/efectos adversos , Cetólidos/síntesis química , Cetólidos/uso terapéutico , Ratones , Pruebas de Sensibilidad Microbiana
3.
Drug Metab Dispos ; 37(7): 1339-54, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19389860

RESUMEN

Cytochrome P450 (P450) induction is one of the factors that can affect the pharmacokinetics of a drug molecule upon multiple dosing, and it can result in pharmacokinetic drug-drug interactions with coadministered drugs causing potential therapeutic failures. In recent years, various in vitro assays have been developed and used routinely to assess the potential for drug-drug interactions due to P450 induction. There is a desire from the pharmaceutical industry and regulatory agencies to harmonize assay methodologies, data interpretation, and the design of clinical drug-drug interaction studies. In this article, a team of 10 scientists from nine Pharmaceutical Research and Manufacturers of America (PhRMA) member companies conducted an anonymous survey among PhRMA companies to query current practices with regards to the conduct of in vitro induction assays, data interpretation, and clinical induction study practices. The results of the survey are presented in this article, along with reviews of current methodologies of in vitro assays and in vivo studies, including modeling efforts in this area. A consensus recommendation regarding common practices for the conduct of P450 induction studies is included.


Asunto(s)
Sistema Enzimático del Citocromo P-450/biosíntesis , Hepatocitos/metabolismo , Receptores de Esteroides/metabolismo , Américas , Biología Computacional , Sistema Enzimático del Citocromo P-450/metabolismo , Recolección de Datos , Evaluación Preclínica de Medicamentos , Industria Farmacéutica , Interacciones Farmacológicas , Inducción Enzimática/fisiología , Predicción , Humanos , Receptor X de Pregnano , Receptores de Esteroides/genética , Proyectos de Investigación , Activación Transcripcional
4.
Bioorg Med Chem Lett ; 17(19): 5447-54, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17707640

RESUMEN

A series of pyrimidine benzamide-based thrombopoietin receptor agonists is described. The lead molecule contains a 2-amino-5-unsubstituted thiazole, a group that has been associated with idiosyncratic toxicity. The potential for metabolic oxidation at C-5 of the thiazole, the likely source of toxic metabolites, was removed by substitution at C-5 or by replacing the thiazole with a thiadiazole. Potency in the series was improved by modifying the substituents on the pyrimidine and/or on the thiazole or thiadiazole pendant aryl ring. In vivo examination revealed that compounds from the series are not highly bioavailable. This is attributed to low solubility and poor permeability.


Asunto(s)
Benzamidas/síntesis química , Benzamidas/farmacología , Pirimidinas/síntesis química , Pirimidinas/farmacología , Receptores de Trombopoyetina/agonistas , Antígenos CD34/metabolismo , Benzamidas/farmacocinética , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Fenómenos Químicos , Química Física , Simulación por Computador , Reacciones Cruzadas , Evaluación Preclínica de Medicamentos , Humanos , Peso Molecular , Pirimidinas/farmacocinética , Solubilidad , Relación Estructura-Actividad
6.
Chem Res Toxicol ; 19(12): 1643-9, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17173378

RESUMEN

The roles of flavin-containing monooxygenases (FMOs) in the oxidation of seleno-l-methionine (SeMet) to l-methionine selenoxide (MetSeO) were investigated using cDNA-expressed human FMOs, purified rat liver FMOs, and rat liver microsomes. MetSeO and the N-2,4-dinitrophenyl-derivatives of SeMet and MetSeO were synthesized and characterized by 1H-NMR and ESI/MS. These reference compounds were then used to develop a sensitive HPLC assay to monitor SeMet oxidation to MetSeO. The formation of MetSeO in rat liver microsomes was time-, protein concentration-, SeMet concentration-, and NADPH-dependent. The microsomal activity exhibited a SeMet Km value (mean +/- S.D.; n = 4) of 0.91 +/- 0.29 mM and a Vmax value of 44 +/- 8.0 nmol MetSeO/mg protein/min. The inclusion of 1-benzylimidazole, superoxide dismutase, or deferoxamine caused no inhibition of the rat liver microsomal activity. Because these results suggested the involvement of FMOs in the oxidation of SeMet in rat liver microsomes, the formation of MetSeO was also examined using cDNA-expressed human and purified rat FMOs. The results showed that both rat and human FMO1 and FMO3 but not FMO5 can catalyze the reaction. The SeMet kinetic constants were obtained with purified rat liver FMO3 (Km = 0.11 mM, Vmax = 280 nmol/mg protein/min) and rat liver FMO1 (Km = 7.8 mM, Vmax = 1200 nmol/mg protein/min). Because SeMet has anti-cancer, chemopreventive, and toxic properties, the kinetic results suggest that FMO3 is likely to play a role in the biological activities of SeMet at low exposure conditions.


Asunto(s)
Metionina/análogos & derivados , Microsomas Hepáticos/metabolismo , Compuestos de Organoselenio/metabolismo , Oxigenasas/metabolismo , Selenometionina/metabolismo , Animales , Cromatografía Líquida de Alta Presión , ADN Complementario/genética , Dinitrobencenos/química , Humanos , Técnicas In Vitro , Espectroscopía de Resonancia Magnética , Fase I de la Desintoxicación Metabólica , Metionina/química , Metionina/metabolismo , Microsomas Hepáticos/enzimología , Compuestos de Organoselenio/química , Oxigenasas/genética , Ratas , Selenometionina/química , Espectrometría de Masa por Ionización de Electrospray
7.
Mol Pharmacol ; 63(3): 722-31, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12606783

RESUMEN

Dehydroepiandrosterone (DHEA) is a C-19 adrenal steroid precursor to the gonadal steroids. In humans, circulating levels of DHEA, as its sulfated conjugate, are high at puberty and throughout early adulthood but decline with age. Dietary supplementation to maintain high levels of DHEA purportedly has beneficial effects on cognitive memory, the immune system, and fat and carbohydrate metabolism. In rodents, DHEA is a peroxisome proliferator that induces genes for the classical peroxisomal and microsomal enzymes associated with this response. These effects are mediated through activation of peroxisome proliferator-activated receptor alpha (PPAR alpha). However, DHEA can affect the expression of genes independently of PPAR alpha, including the gene for the major inducible drug and xenobiotic metabolizing enzyme, cytochrome P450 3A23. To elucidate the biochemistry associated with DHEA treatment, we employed a cDNA gene expression array using liver RNA from rats treated with DHEA or the classic peroxisome proliferator nafenopin. Principal components analysis identified 30 to 35 genes whose expression was affected by DHEA and/or nafenopin. Some were genes previously identified as PPAR-responsive genes. Changes in expression of several affected genes were verified by quantitative reverse transcriptase-polymerase chain reaction. These included aquaporin 3, which was induced by DHEA and to a lesser extent nafenopin, nuclear tyrosine phosphatase, which was induced by both agents, and 11 beta-hydroxysteroid dehydrogenase 1, which was decreased by treatment with DHEA in a dose-dependent fashion. Regulation of 11 beta-hydroxysteroid dehydrogenase 1 expression is important since the enzyme is believed to amplify local glucocorticoid signaling, and its repression may cause some of the metabolic effects associated with DHEA.


Asunto(s)
Deshidroepiandrosterona/farmacología , Expresión Génica/efectos de los fármacos , Hidroxiesteroide Deshidrogenasas/biosíntesis , Hígado/efectos de los fármacos , 11-beta-Hidroxiesteroide Deshidrogenasas , Animales , Acuaporina 3 , Acuaporinas/biosíntesis , Acuaporinas/genética , Perfilación de la Expresión Génica , Hidroxiesteroide Deshidrogenasas/genética , Hipolipemiantes/farmacología , Hígado/enzimología , Masculino , Nafenopina/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA