Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Pharmacol ; 59(1): 139-152, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30192390

RESUMEN

Maraviroc is a C-C chemokine receptor type-5 antagonist approved for the treatment of HIV-1. Previous studies show that cytochrome P450 3A5 (CYP3A5) plays a role in maraviroc metabolism. CYP3A5 is subject to a genetic polymorphism. The presence of 2 functional alleles (CYP3A5*1/*1) confers the extensive metabolism phenotype, which is rare in whites but common in blacks. The effect of CYP3A5 genotype on maraviroc and/or metabolite pharmacokinetics was evaluated in 2 clinical studies: a post hoc analysis from a phase 2b/3 study (NCT00098293) conducted in 494 HIV-1-infected subjects (study 1) in which the impact on maraviroc efficacy in 303 subjects was also assessed, and a study conducted in 47 healthy volunteers (study 2). In study 2 (NCT02625207), extensive metabolizers had 26% to 37% lower mean area under the concentration-time curve compared with poor metabolizers (no CYP3A5*1 alleles). This effect diminished to 17% in the presence of potent CYP3A inhibition. The effect of CYP3A5 genotype was greatest in the formation of the metabolite (1S,2S)-2-hydroxymaraviroc. In study 1, the CYP3A5*1/*1 genotype unexpectedly had higher maraviroc area under the curve predictions (20%) compared with those with no CYP3A5*1 alleles. The reason for this disparity remains unclear. The proportions of subjects with viral loads <50 and <400 copies/mL for maraviroc were comparable among all 3 CYP3A5 genotypes. In both studies maraviroc exposures were in the range of near-maximal viral inhibition in the majority of subjects. These results demonstrate that although CYP3A5 contributes to the metabolism of maraviroc, CYP3A5 genotype does not affect the clinical response to maraviroc in combination treatment of HIV-1 infection at approved doses.


Asunto(s)
Citocromo P-450 CYP3A/genética , Inhibidores de Fusión de VIH/farmacocinética , Inhibidores de Fusión de VIH/uso terapéutico , Infecciones por VIH , VIH-1 , Maraviroc/farmacocinética , Maraviroc/uso terapéutico , Adulto , Método Doble Ciego , Femenino , Genotipo , Inhibidores de Fusión de VIH/sangre , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , Voluntarios Sanos , Humanos , Masculino , Maraviroc/sangre , Persona de Mediana Edad , Polimorfismo Genético , Resultado del Tratamiento , Adulto Joven
2.
Drug Metab Dispos ; 46(5): 493-502, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29475834

RESUMEN

Maraviroc (MVC) is a CCR5 coreceptor antagonist indicated in combination with other antiretroviral agents for the treatment of CCR5-tropic human immunodefinciency virus-1 infection. In this study, the metabolism of MVC was investigated in human liver microsomes to delineate the relative roles of CYP3A4 and CYP3A5. MVC is metabolized to five hydroxylated metabolites, all of which were biosynthesized and identified using mass and NMR spectroscopy. The sites of metabolism were the 2- and 3-positions of the 4,4-difluorocyclohexyl moiety and the methyl of the triazole moiety. Absolute configurations were ultimately ascertained by comparison to authentic standards. The biosynthesized metabolites were used for quantitative in vitro experiments in liver microsomes using cyp3cide, a selective inactivator of CYP3A4. (1S,2S)-2-OH-MVC was the main metabolite representing approximately half of the total metabolism, and CYP3A5 contributed approximately 40% to that pathway in microsomes from CYP3A5*1/*1 donors. The other four metabolites were almost exclusively metabolized by CYP3A4. (1S,2S)-2-hydroxylation also correlated to T-5 N-oxidation, a CYP3A5-specific activity. These data are consistent with clinical pharmacokinetic data wherein CYP3A5 extensive metabolizer subjects showed a modestly lower exposure to MVC.


Asunto(s)
Ciclohexanos/metabolismo , Citocromo P-450 CYP3A/metabolismo , Triazoles/metabolismo , Ciclohexanos/farmacocinética , Humanos , Hidroxilación/fisiología , Cinética , Maraviroc , Microsomas Hepáticos/metabolismo , Oxidación-Reducción , Pirazoles/metabolismo , Pirazoles/farmacocinética , Pirimidinas/metabolismo , Pirimidinas/farmacocinética , Triazoles/farmacocinética
3.
Toxicol Pathol ; 43(3): 354-65, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25122632

RESUMEN

Administration of lersivirine, a nonnucleotide reverse transcriptase inhibitor, daily by oral gavage to Sprague-Dawley rats for up to 2 yr was associated with decreased survival, decreased body weights, and an increase in neoplasms and related proliferative lesions in the liver, thyroid, kidney, and urinary bladder. Thyroid follicular adenoma and carcinoma, the associated thyroid follicular hypertrophy/hyperplasia, hepatocellular adenoma/adenocarcinoma, altered cell foci, and hepatocellular hypertrophy were consistent with lersivirine-related induction of hepatic microsomal enzymes. Renal tubular adenoma and renal tubular hyperplasia were attributed to the lersivirine-related exacerbation of chronic progressive nephropathy (CPN), while urinary bladder hyperplasia and transitional cell carcinoma in the renal pelvis and urinary bladder were attributed to urinary calculi. Renal tubular neoplasms associated with increased incidence and severity of CPN, neoplasms of transitional epithelium attributed to crystalluria, and thyroid follicular and hepatocellular neoplasms related to hepatic enzyme induction have low relevance for human risk assessment.


Asunto(s)
Carcinógenos/toxicidad , Nitrilos/toxicidad , Pirazoles/toxicidad , Inhibidores de la Transcriptasa Inversa/toxicidad , Animales , Peso Corporal/efectos de los fármacos , Pruebas de Carcinogenicidad , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Femenino , Estimación de Kaplan-Meier , Neoplasias Renales/inducido químicamente , Neoplasias Renales/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/patología , Masculino , Nitrilos/farmacocinética , Pirazoles/farmacocinética , Ratas , Ratas Sprague-Dawley , Inhibidores de la Transcriptasa Inversa/farmacocinética , Análisis de Supervivencia , Neoplasias de la Tiroides/inducido químicamente , Neoplasias de la Tiroides/patología , Urinálisis , Neoplasias de la Vejiga Urinaria/inducido químicamente , Neoplasias de la Vejiga Urinaria/patología
4.
Drug Metab Lett ; 4(3): 162-72, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20642449

RESUMEN

The quinuclidine PHA-0568487(1) is an agonist of the alpha 7 nicotinic acetylcholine receptor that was designed to mitigate the bioactivation associated with the core scaffold and subsequently remove associated liabilities with in vivo tolerability. The drug metabolites of 1 in nonclinical species were identified in plasma and urine of rats, dogs and monkeys receiving oral administrations of 1. The in vitro biotransformation of 1 was subsequently investigated in multiple species employing cryopreserved hepatocytes, hepatic subcellular fractions and recombinantly-expressed human P450 enzymes. In addition, in vitro metabolism of synthetically prepared metabolite precursors were instrumental in the elucidation of several secondary metabolites. The results indicated that the principal biotransformation of 1 was oxidation of the benzo[1,4]dioxane moiety (M8, M10) followed by subsequent oxidation to a range of secondary metabolites (M1-7, M9, M11, M13-15, and M17-18). The carboxylic acids M1 and M2 resulting from the oxidative cleavage of the dioxane ring were the principal metabolites observed in the plasma, urine and hepatocyte incubations across all species (M1 & M2). Quinuclidine oxidation was another pathway of importance, yielding an N-oxide (M12) which was also observed in all species.P450 2D6 and FMO1 catalyze the oxidation of the quinuclidine nitrogen. The N oxidation of the quinuclidine moiety is consistent with previously published accounts of this scaffold's metabolism and, interestingly, may implicate the uncommon quinuclidine moiety as an entity directing the metabolism of this scaffold (e.g., 1) via FMO1 and P450 2D6 oxidation.


Asunto(s)
Compuestos Aza/farmacocinética , Dioxinas/farmacocinética , Agonistas Nicotínicos/farmacocinética , Quinuclidinas/farmacocinética , Receptores Nicotínicos/efectos de los fármacos , Administración Oral , Animales , Compuestos Aza/administración & dosificación , Compuestos Aza/sangre , Compuestos Aza/orina , Biotransformación , Cromatografía Liquida , Citocromo P-450 CYP2D6/metabolismo , Dioxinas/administración & dosificación , Dioxinas/sangre , Dioxinas/orina , Perros , Haplorrinos , Hepatocitos/enzimología , Humanos , Espectroscopía de Resonancia Magnética , Agonistas Nicotínicos/administración & dosificación , Agonistas Nicotínicos/sangre , Agonistas Nicotínicos/orina , Oxidación-Reducción , Oxigenasas/metabolismo , Quinuclidinas/administración & dosificación , Quinuclidinas/sangre , Quinuclidinas/orina , Ratas , Proteínas Recombinantes/metabolismo , Espectrometría de Masas en Tándem , Receptor Nicotínico de Acetilcolina alfa 7
5.
ACS Med Chem Lett ; 1(5): 219-23, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-24900198

RESUMEN

As part of a strategy to deliver short-acting calcium-sensing receptor (CaSR) antagonists, the metabolically labile thiomethyl functionality was incorporated into the zwitterionic amino alcohol derivative 3 with the hope of increasing human clearance through oxidative metabolism, while delivering a pharmacologically inactive sulfoxide metabolite. The effort led to the identification of thioanisoles 22 and 23 as potent and orally active CaSR antagonists with a rapid onset of action and short pharmacokinetic half-lives, which led to a rapid and transient stimulation of parathyroid hormone in a dose-dependent fashion following oral administration to rats. On the basis of the balance between target pharmacology, safety, and human disposition profiles, 22 and 23 were advanced as clinical candidates for the treatment of osteoporosis.

6.
Expert Rev Clin Pharmacol ; 1(4): 515-31, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24410554

RESUMEN

It is generally accepted that bioactivation of relatively inert functional groups (toxicophores) to reactive metabolites is an obligatory step in the pathogenesis of certain idiosyncratic adverse drug reactions (IADRs). IADRs cannot be detected in regulatory animal toxicity studies and, given their low frequency of occurrence in humans (1 in 10,000 to 1 in 100,000), they are often not detected until the drug has gained broad exposure in a large patient population. The detection of IADRs during late clinical trials or after a drug has been released can lead to an unanticipated restriction in its use, and even in its withdrawal. To date, there is neither a consistent nor a well-defined link between bioactivation and IADRs; however, the potential does exist for these processes to be causally related. Thus, the formation of reactive metabolites with a drug candidate is generally considered a liability in most pharmaceutical companies. Procedures have been implemented to evaluate bioactivation potential of new drug candidates with the goal of eliminating or minimizing reactive metabolite formation by rational structural modification of the lead chemical class. While such studies have proven extremely useful in the retrospective analysis of bioactivation pathways of toxic drugs and defining toxicophores, their ability to accurately predict the IADR potential of new drug candidates has been challenged, given that several commercially successful drugs form reactive metabolites, yet, they are not associated with a significant incidence of IADRs. In this article, we review the basic methodology that is currently utilized to evaluate the bioactivation potential of new compounds, with particular emphasis on the advantages and limitation of these assays. Plausible reasons for the excellent safety record of certain drugs susceptible to bioactivation are also explored. Overall, these observations provide valuable guidance in the proper use of bioactivation assessments when selecting drug candidates for development.

7.
Toxicol Appl Pharmacol ; 225(2): 221-8, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17889094

RESUMEN

Parabens are esters of 4-hydroxybenzoic acid and used as anti-microbial agents in a wide variety of toiletries, cosmetics and pharmaceuticals. It is of interest to understand the dermal absorption and hydrolysis of parabens, and to evaluate their disposition after dermal exposure and their potential to illicit localised toxicity. The use of minipig as a surrogate model for human dermal metabolism and toxicity studies, justifies the comparison of paraben metabolism in human and minipig skin. Parabens are hydrolysed by carboxylesterases to 4-hydroxybenzoic acid. The effects of the carboxylesterase inhibitors paraoxon and bis-nitrophenylphosphate provided evidence of the involvement of dermal carboxylesterases in paraben hydrolysis. Loperamide, a specific inhibitor of human carboxylesterase-2 inhibited butyl- and benzylparaben hydrolysis in human skin but not methylparaben or ethylparaben. These results show that butyl- and benzylparaben are more selective substrates for human carboxylesterase-2 in skin than the other parabens examined. Parabens applied to the surface of human or minipig skin were absorbed to a similar amount and metabolised to 4-hydroxybenzoic acid during dermal absorption. These results demonstrate that the minipig is a suitable model for man for assessing dermal absorption and hydrolysis of parabens, although the carboxylesterase profile in skin differs between human and minipig.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Modelos Animales , Parabenos/farmacocinética , Conservadores Farmacéuticos/farmacocinética , Absorción Cutánea , Adulto , Animales , Citosol/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Hidrólisis , Masculino , Microsomas/metabolismo , Parabenos/metabolismo , Piel/metabolismo , Especificidad de la Especie , Porcinos , Porcinos Enanos
8.
Toxicol Lett ; 173(2): 118-23, 2007 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-17719730

RESUMEN

Skin esterases serve an important pharmacological function as they can be utilised for activation of topically applied ester prodrugs. Understanding the nature of these enzymes, with respect to their role and local activity, is essential to defining the efficacy of ester prodrugs. Minipigs are used as models to study the kinetics of absorption of topically applied drugs. Their skin has structural properties very similar to human skin. However, regional distribution differences in esterase activity from site-to-site could influence cross-species extrapolation. Investigation of the regional site variation of minipig skin esterase activity will facilitate standardization of topically applied drug studies. Furthermore, the characterization of regional skin variation, will aid in translation of minipig results to better predictions of human esterase activity. Here we report the variation in rates of hydrolysis by minipig skin taken from different regional sites, using the esterase-selective substrates: phenyl valerate (carboxylesterase), phenyl acetate (arylesterase) and p-nitrophenyl acetate (general esterase). Skin from ears and back of male minipig showed higher activity than female. Skin from minipig ears and the back showed the highest level of esterase activity and was similar to human breast skin used in vitro absorption studies. These results suggest that skin from the minipig back is an appropriate model for preclinical human skin studies, particularly breast skin. This study supports the use of the minipig, with topical application to the back, as a model for the investigation of pharmacokinetics and metabolism of ester prodrugs.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Esterasas/metabolismo , Hígado/enzimología , Modelos Animales , Piel/enzimología , Porcinos Enanos/metabolismo , Porcinos/metabolismo , Animales , Carboxilesterasa/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Citosol/enzimología , Femenino , Hidrólisis , Cinética , Masculino , Microsomas/enzimología , Nitrofenoles/metabolismo , Fenilacetatos/metabolismo , Reproducibilidad de los Resultados , Factores Sexuales , Especificidad por Sustrato , Valeratos/metabolismo
9.
Drug Metab Dispos ; 35(11): 2015-22, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17664252

RESUMEN

The capacity of human, minipig, and rat skin and liver subcellular fractions to hydrolyze the anesthetic ester procaine was compared with carboxylesterase substrates 4-methylumbelliferyl-acetate, phenylvalerate, and para-nitrophenylacetate and the arylesterase substrate phenylacetate. Rates of procaine hydrolysis by minipig and human skin microsomal and cytosolic fractions were similar, with rat displaying higher activity. Loperamide inhibited procaine hydrolysis by human skin, suggesting involvement of human carboxylesterase hCE2. The esterase activity and inhibition profiles in the skin were similar for minipig and human, whereas rat had a higher capacity to metabolize esters and a different inhibition profile. Minipig and human liver and skin esterase activity was inhibited principally by paraoxon and bis-nitrophenyl phosphate, classical carboxylesterase inhibitors. Rat skin and liver esterase activity was inhibited additionally by phenylmethylsulfonyl fluoride and the arylesterase inhibitor mercuric chloride, indicating a different esterase profile. These results have highlighted the potential of skin to hydrolyze procaine following topical application, which possibly limits its pharmacological effect. Skin from minipig used as an animal model for assessing transdermal drug preparations had similar capacity to hydrolyze esters to human skin.


Asunto(s)
Esterasas/metabolismo , Hígado/enzimología , Procaína/metabolismo , Piel/enzimología , Animales , Inhibidores Enzimáticos/farmacología , Esterasas/antagonistas & inhibidores , Ésteres , Femenino , Humanos , Hidrólisis/efectos de los fármacos , Loperamida/farmacología , Masculino , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Estructura Molecular , Nitrofenoles/química , Nitrofenoles/metabolismo , Nitrofenoles/farmacología , Paraoxon/farmacología , Ácidos Pentanoicos/química , Ácidos Pentanoicos/metabolismo , Fenilacetatos/química , Fenilacetatos/metabolismo , Fluoruro de Fenilmetilsulfonilo/farmacología , Procaína/química , Procaína/farmacocinética , Ratas , Ratas Wistar , Porcinos , Porcinos Enanos , Umbeliferonas/química , Umbeliferonas/metabolismo
10.
Drug Metab Dispos ; 34(9): 1615-23, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16790552

RESUMEN

The metabolism and excretion of N-(3R)-1-azabicyclo[2.2.2]oct-3-ylfuro[2,3-c]pyridine-5-carboxamide (1), an agonist of the alpha7 nicotinic acetylcholinergic receptor, were determined in both Sprague-Dawley rats and beagle dogs using [3H]1. Initially, 3-tritio-furanopyridine 1 ([3H]1a) was evaluated in pilot mass balance studies by determining total radioactivity recovery and pharmacokinetics in lyophilized excreta and nonlyophilized plasma, respectively. Lower mass balance and much greater circulatory radioactivity exposures were observed in rats than in dogs, with urinary tritiated water (HTO) only detected in rats. The 133-h half-life in rats, possibly due to very slowly eliminated metabolites, was more likely attributable to HTO formed from [3H]1a because of site-specific chemical and/or metabolic 3H instability, which was confirmed by urinary HTO. In contrast, dog data supported 3H stability within [3H]1a. Conflicting cross-species data with [3H]1a suggested species-specific metabolic fates for 1, requiring a 3H form of 1 resistant to 3H loss in rats. Therefore, tritiation of 1 at its furanopyridine C7, a site predicted to be both chemically and metabolically stable, yielded 7-tritio-N-(3R)-1-azabicyclo[2.2.2]oct-3-ylfuro[2,3-c]pyridine-5-carboxamide ditrifluoroacetate ([3H]1b), which allowed in both species the determination of all excretory pathways, total radioactivity pharmacokinetics, and major excretory and circulatory metabolites with complete radioactivity recovery without HTO generation. Definitive metabolite elucidation for 1 using [3H]1b confirmed the suspected species-dependent metabolic susceptibility for 3H loss from [3H]1a in rats, but not dogs, since the majority of rat metabolites resulted from furanopyridine biotransformation. The described studies explore the evaluation of tritium exchange risk from a mechanistic biotransformation perspective and highlight the need for careful deliberation when considering and designing 3H compounds for radiolabeled metabolism studies.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/farmacocinética , Evaluación Preclínica de Medicamentos , Marcaje Isotópico , Quinuclidinas/farmacocinética , Tritio , Animales , Biotransformación , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Perros , Evaluación Preclínica de Medicamentos/métodos , Estabilidad de Medicamentos , Heces/química , Estructura Molecular , Quinuclidinas/química , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie
11.
Biol Pharm Bull ; 26(5): 573-8, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12736492

RESUMEN

Our study objectives were: To quantitatively determine the effect of radiolabel instability on terminal phase radioactive tissue residues in a multiple dose tissue distribution study. To quantitatively compare tissue residue artifacts (non drug-related radioactivity) from two chemically-distinct radiolabel locations. To conduct a definitive multiple dose tissue distribution study using the better of the two radiolabeled compounds. We compared the excretion and tissue distribution in rats of [(14)C]linezolid, radiolabeled in two different locations, after 7 consecutive once daily [(14)C] oral doses. The radiolabels were in the acetamide (two carbon) and oxazolidinone (isolated carbon) functional groups. Terminal phase tissue residue and excretion data were compared to data from rats dosed orally with [(14)C]sodium acetate. Drug-related radioactivity was excreted rapidly over 24 h. After a single dose, the acetamide and oxazolidinone radiolabel sites both gave 3% of dose as exhaled (14)CO(2). After 7 daily [(14)C] oral doses, terminal phase radioactive tissue residues were higher from the acetamide radiolabel, relative to the oxazolidinone radiolabel, and were primarily not drug-related. In the definitive tissue distribution study, low concentrations of drug-related radioactivity in skin and thyroid were observed. We conclude that although small amounts of radiolabel instability do not significantly affect single dose tissue radioactivity C(max) and AUC, artifacts arising from radiolabel instability can prolong the apparent terminal phase half life and complicate study data interpretation. When possible, it is always preferable to use a completely stable radiolabel site.


Asunto(s)
Acetamidas/farmacocinética , Antibacterianos/farmacocinética , Radioisótopos de Carbono/farmacocinética , Oxazolidinonas/farmacocinética , Acetamidas/administración & dosificación , Administración Oral , Animales , Antibacterianos/administración & dosificación , Área Bajo la Curva , Estabilidad de Medicamentos , Semivida , Linezolid , Masculino , Tasa de Depuración Metabólica , Oxazolidinonas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Acetato de Sodio/farmacocinética , Factores de Tiempo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...