Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Pharm Res ; 35(8): 159, 2018 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-29915999

RESUMEN

PURPOSE: The renal clearance of fampridine (Fampyra®, or Ampyra®) significantly exceeds the glomerular filtration rate, suggesting active renal secretion is likely the major elimination pathway. The goal of this study was to identify the renal transporters that are involved in the renal active secretion, and elucidate the active renal secretion mechanism of fampridine. METHODS: The uptake of fampridine to HEK-293 cells overexpressing human OCT2, MATE1 or MATE2K was determined in the absence and presence of Cimetidine, the prototypical inhibitor of the transporters. The inhibition potential of fampridine on the renal transporters was evaluated by determining the uptake of TEA and Metformin, the probe substrates of the transporters of OCT2 and MATEs, respectively, in the absence or presence of fampridine. RESULTS: Significant time- and concentration-dependent uptake of fampridine by human OCT2 was observed. The Km and Vmax were determined as 51.0 ± 17.1 µM and 1107 ± 136 pmole/min/106 cells, respectively. Fampridine also inhibited OCT2 mediated uptake of Metformin with estimated IC50 of 66.8 µM. In contrast, there was not significant uptake of fampridine by human MATE1 or MATE2K, and fampridine did not inhibit MATE1 or MATE2K mediated uptake of TEA. CONCLUSION: The studies indicated fampridine is a substrate and inhibitor of OCT2, but not MATE1 or MATE2K. Results from the study suggested the active renal secretion of fampridine is mediated by human OCT2 but not MATE1 or MATE2K. To our knowledge, fampridine is the first reported substrate specific to OCT2 but not to MATE1 or MATE2K.


Asunto(s)
4-Aminopiridina/farmacocinética , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 2 de Cátion Orgánico/metabolismo , Bloqueadores de los Canales de Potasio/farmacocinética , 4-Aminopiridina/metabolismo , 4-Aminopiridina/farmacología , Transporte Biológico/efectos de los fármacos , Células HEK293 , Humanos , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacocinética , Metformina/metabolismo , Metformina/farmacocinética , Transportador 2 de Cátion Orgánico/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología
2.
Drug Metab Dispos ; 40(4): 779-87, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22266779

RESUMEN

The objectives of the study were to characterize the selectivity of dantrolene to breast cancer resistance protein (Bcrp) and to evaluate whether cerebrospinal fluid (CSF) can be used as a surrogate to assess brain exposures of BCRP and P-glycoprotein (Pgp) substrates. The impact of Bcrp and Pgp on dantrolene exposures in brain and CSF was examined in Bcrp and Mdr1a/1b knockout mice and was further investigated in wild-type mice in the presence of the Bcrp inhibitor (3S,6S,12aS)-1,2,3,4,6,7,12,12a-octahydro-9-methoxy-6-(2-methylpropyl)-1,4-dioxopyrazino[1',2':1,6]pyrido[3,4-b]indole-3-propanoic acid 1,1-dimethylethyl ester (Ko143), the Pgp inhibitor 6-[(2S,4R,6E)-4-methyl-2-(methylamino)-3-oxo-6-octenoic acid]-7-l-valine-cyclosporine A (PSC833), and the dual inhibitor N-(4-[2-(1,2,3,4-tetrahydro-6,7-dimethoxy-2-isoquinolinyl)ethyl]-phenyl)-9,10-dihydro-5-methoxy-9-oxo-4-acridine carboxamide (GF120918). The effect of Bcrp and Pgp on digoxin exposures in brain and CSF was investigated in wild-type mice in the presence of the inhibitors. In vivo studies showed dantrolene exposures in brain and CSF, but not the blood, increased in Bcrp(-/-) and Mdr1a/1b(-/-)/Bcrp(-/-) mice, or in the presence of the Bcrp inhibitors Ko143 or GF120918. Inhibition of Pgp by GF120918 and PSC833 significantly increased digoxin exposures in brain, CSF, and blood to a lesser extent. Results from the present study demonstrated that inhibition of Bcrp and Pgp increased not only the exposures of dantrolene and digoxin in brain, but also the exposures in CSF. In addition, the change of exposures in CSF reflected the changes in brain. The present study strongly suggests that the dantrolene and digoxin exposures in CSF are primarily determined by the rapid transport from brain to CSF, and inhibition of Bcrp and Pgp exhibits little impact on using CSF as surrogates to assess brain exposures of Bcrp and Pgp substrates.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Barrera Hematoencefálica/metabolismo , Preparaciones Farmacéuticas , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/genética , Animales , Transporte Biológico , Barrera Hematoencefálica/efectos de los fármacos , Células CACO-2 , Dantroleno/administración & dosificación , Dantroleno/sangre , Dantroleno/líquido cefalorraquídeo , Digoxina/administración & dosificación , Digoxina/sangre , Digoxina/líquido cefalorraquídeo , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Ratones Noqueados , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/sangre , Preparaciones Farmacéuticas/líquido cefalorraquídeo , Factores de Tiempo , Distribución Tisular
3.
Drug Metab Dispos ; 39(6): 1054-7, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21393461

RESUMEN

A refined cytochrome P450 (P450) enzyme IC50 shift assay for more accurately screening CYP3A time-dependent inhibitors (TDIs) is presented. In contrast to the regular IC50 shift assay, in which only one pair of P450 inhibition curves is generated, this modified method generates two pairs of inhibition curves; one pair of curves is created from human liver microsomal incubations with the test article in the presence or absence of NADPH (curves 1 and 2) (same as the traditional assay), and the other pair is created from new microsomal incubations with extract (compound/metabolites) of previous incubations (curves 3 and 4). To assess the true CYP3A time-dependent inhibition, we propose a new parameter, the vertical IC50 curve shift (VICS), represented by vertical shift difference between the two sets of curves divided by inhibitor concentration at which maximal vertical shift of curves 1 and 2 is observed. A shift in the curves 1 and 2 could mean a time-dependent inhibition or formation of a more active inhibitory metabolite(s). The new method provides more reliable characterization of the shift as a result of a true TDI- or metabolite-mediated reversible inhibition. Nine known TDI drugs were evaluated using this refined shift assay. The derived VICS values correlated well with the reported k(inact)/K(I) values derived via the conventional dilution assay method. Thus, the refined assay can be used to identify a true TDI and quantitatively assess the inactivation potential of TDIs in a high-throughput fashion. This assay can be invaluable to screen for true P450 TDIs in the early drug discovery.


Asunto(s)
Inhibidores del Citocromo P-450 CYP3A , Inhibidores Enzimáticos/efectos adversos , Ensayos Analíticos de Alto Rendimiento/métodos , Citocromo P-450 CYP3A , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Interacciones Farmacológicas , Inhibidores Enzimáticos/metabolismo , Humanos , Concentración 50 Inhibidora , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Factores de Tiempo
4.
Drug Metab Dispos ; 39(4): 693-702, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21177487

RESUMEN

Oxymetazoline (6-tert-butyl-3-(2-imidazolin-2-ylmethyl)-2,4-dimethylphenol) has been widely used as a nonprescription nasal vasoconstrictor for >40 years; however, its metabolic pathway has not been investigated. This study describes the in vitro metabolism of oxymetazoline in human, rat, and rabbit liver postmitochondrial supernatant fraction from homogenized tissue (S9) fractions and their microsomes supplemented with NADPH. The metabolites of oxymetazoline identified by liquid chromatography (LC)/UV/tandem mass spectrometry (MS/MS), included M1 (monohydroxylation of the t-butyl group), M2 (oxidative dehydrogenation of the imidazoline to an imidazole moiety), M3 (monohydroxylation of M2), M4 (dihydroxylation of oxymetazoline), and M5 (dihydroxylation of M2). Screening with nine human expressed cytochromes P450 (P450s) identified CYP2C19 as the single P450 isoform catalyzing the formation of M1, M2, and M3. Glutathione conjugates of oxymetazoline (M6) and M2 (M7) were identified in the liver S9 fractions, indicating the capability of oxymetazoline to undergo bioactivation to reactive intermediate species. M6 and M7 were not detected in those liver S9 incubations without NADPH. Cysteine conjugates (M8 and M9) derived from glutathione conjugates and hydroxylated glutathione conjugates (M10 and M11) were also identified. The reactive intermediate of oxymetazoline was trapped with glutathione and N-acetyl cysteine and identified by LC/MS/MS. M6 was isolated and identified by one-dimensional or two-dimensional NMR as the glutathione conjugate of a p-quinone methide. We have shown the tendency of oxymetazoline to form p-quinone methide species via a bioactivation mechanism involving a CYP2C19-catalyzed two-electron oxidation. Nevertheless, we conclude that the formation of this reactive species might not be a safety concern for oxymetazoline nasal products because of the typical low-dose and brief dosage regimen limited to nasal delivery.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Glutatión/metabolismo , Oximetazolina/metabolismo , Simpatomiméticos/metabolismo , Acetilcisteína/metabolismo , Animales , Hidrocarburo de Aril Hidroxilasas/metabolismo , Citocromo P-450 CYP2C19 , Humanos , Hidroxilación , Técnicas In Vitro , Indolquinonas/metabolismo , Hígado/metabolismo , Masculino , Microsomas Hepáticos/metabolismo , NADP/metabolismo , Oxidación-Reducción , Oximetazolina/química , Conejos , Ratas , Simpatomiméticos/química
5.
Bioorg Med Chem Lett ; 21(18): 5633-7, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21798738

RESUMEN

A novel class of pyrazolopyrimidine-sulfonamides was discovered as selective dual inhibitors of aurora kinase A (AKA) and cyclin-dependent kinase 1 (CDK1). These inhibitors were originally designed based on an early lead (compound I). SAR development has led to the discovery of potent inhibitors with single digit nM IC(50)s towards both AKA and CDK1. An exemplary compound 1a has demonstrated good efficacy in an HCT116 colon cancer xenograft model.


Asunto(s)
Antineoplásicos/farmacología , Proteína Quinasa CDC2/antagonistas & inhibidores , Neoplasias del Colon/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Sulfonamidas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Aurora Quinasa A , Aurora Quinasas , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Técnicas de Química Sintética , Neoplasias del Colon/patología , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Pirimidinas/síntesis química , Pirimidinas/química , Estereoisomerismo , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/química , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cancer Chemother Pharmacol ; 62(1): 117-22, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17899085

RESUMEN

PURPOSE: The objective of this study was to understand the biochemical mechanisms by which a haloenol lactone (HEL) derivative potentiates cisplatin-induced cytotoxicity in vitro. HEL was originally designed and synthesized as a site-directed inactivator of glutathione S-transferase pi isozyme (GST-pi). Over-expression of GST-pi has been found to be associated with chemotherapy resistance. METHODS: A concentration-dependent GST inhibition was assessed after UOK130 cells were exposed to HEL at concentrations of 10 and 20 muM. Potentiated cytotoxicity was evaluated by treatment of UOK130 cells with a selection of alkylating agents in the presence or absence of HEL. Intracellular glutathione (GSH) was determined after exposure to HEL. Protective effect of GSH was examined by co-treatment with GSH ester in UOK130 cells exposed with a combination of cisplatin and HEL. Multiple resistance-associated protein (MRP) 1-3 activity was assayed by determining the rate of (3)H-LTC(4) and (3)H-E(2)17betaG through the MRPs into recombinant membrane vesicles. RESULTS: Exposure of HEL at 10 and 20 muM caused 28 and 41% of inhibition of cellular GST activity. Cytotoxicity of cisplatin, chlorambucil, and melphalan was enhanced 1.8-2.7-fold by HEL at 10 muM. No significant protection effect by GSH ester exposure was observed on cisplatin toxicity co-treated with HEL. HEL was found to inhibit MRP1, MRP2, and MRP3 with IC(50) of 1.30, 28.2, and 3.66 muM, respectively. CONCLUSION: Haloenol lactone showed inhibitory effect on GST-pi and MRP1-3 (selective inhibition of MRP1 and MRP3), and it was also found to deplete intracellular GSH.


Asunto(s)
4-Butirolactona/análogos & derivados , Antineoplásicos/farmacología , 4-Butirolactona/farmacología , Antineoplásicos Alquilantes/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Estradiol/biosíntesis , Glutatión/metabolismo , Gutatión-S-Transferasa pi/antagonistas & inhibidores , Gutatión-S-Transferasa pi/biosíntesis , Humanos , Leucotrieno C4/biosíntesis , Membranas/efectos de los fármacos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/biosíntesis , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética
7.
Fluids Barriers CNS ; 15(1): 10, 2018 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-29558954

RESUMEN

BACKGROUND: Many studies have focused on the challenges of small molecule uptake across the blood-brain barrier, whereas few in-depth studies have assessed the challenges with the uptake of antibodies into the central nervous system (CNS). In drug development, cerebrospinal fluid (CSF) sampling is routinely used as a surrogate for assessing CNS drug exposure and biomarker levels. In this report, we have studied the kinetic correlation between CSF and serum drug concentration-time profiles for five humanized monoclonal antibodies in rats and cynomolgus monkeys and analyzed factors that affect their CSF exposure. RESULTS: Upon intravenous (IV) bolus injection, antibodies entered the CNS slowly and reached maximum CSF concentration ( CSF T max ) in one to several days in both rats and monkeys. Antibody serum and CSF concentration-time curves converged until they became parallel after CSF T max was reached. Antibody half-lives in CSF ( CSF t ½ ) approximated their serum half-lives ( serum t ½ ). Although the intended targets of these antibodies were different, the steady-state CSF to serum concentration ratios were similar at 0.1-0.2% in both species. Independent of antibody target and serum concentration, CSF-to-serum concentration ratios for individual monkeys ranged by up to tenfold from 0.03 to 0.3%. CONCLUSION: Upon systemic administration, average antibodies CSF-to-serum concentration ratios in rats and monkeys were 0.1-0.2%. The CSF t ½ of the antibodies was largely determined by their long systemic t ½ ( systemic t ½ ).


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/líquido cefalorraquídeo , Administración Intravesical , Animales , Anticuerpos Monoclonales/sangre , Encéfalo/metabolismo , Líquido Cefalorraquídeo/metabolismo , Humanos , Cinética , Macaca fascicularis , Masculino , Ratas Sprague-Dawley , Médula Espinal/metabolismo , Factores de Tiempo
8.
Curr Drug Metab ; 8(4): 341-63, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17504223

RESUMEN

Drug transporters, including efflux transporters (the ATP binding cassette (ABC) proteins) and uptake transporters (the solute carrier proteins (SLC)), have an important impact on drug disposition, efficacy, drug-drug interactions and toxicity. Identification of the interactions of chemical scaffolds with transporters at the early stages of drug development can assist in the optimization and selection of new drug candidates. In this review, we discuss current in vitro and in vivo models used to investigate the interactions between drugs and transporters such as P-gp, MRP, BCRP, BSEP, OAT, OATP, OCT, NTCP, PEPT1/2 and NT. In vitro models including cell-based, cell-free, and yeast systems as well as in vivo models such as genetic knockout, gene deficient and chemical knockout animals are discussed and compared. The applications, throughput, advantages and limitations of each model are also addressed in this review.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Proteínas de Transporte de Membrana/metabolismo , Preparaciones Farmacéuticas/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Autorradiografía , Unión Competitiva , Bioensayo , Transporte Biológico , Células Cultivadas , Colorantes Fluorescentes/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Noqueados , Microscopía Confocal , Transportadores de Anión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Tomografía de Emisión de Positrones , Fracciones Subcelulares , Simportadores/metabolismo , Tomografía Computarizada de Emisión de Fotón Único , Transfección
9.
Curr Top Med Chem ; 5(11): 1033-8, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16181128

RESUMEN

The high-throughput screening in drug discovery for absorption, distribution, metabolism and excretion (ADME) properties has become the norm in the industry. Only a few years ago it was ADME properties that were attributed to more failure of drugs than efficacy or safety in the clinic trials. With the realization of new techniques and refinement of existing techniques better projections for the pharmacokinetic properties of compounds in humans are being made, shifting the drug failure attributes more to the safety and efficacy properties of drug candidates. There are a tremendous number of tools available to discovery scientists to screen compounds for optimization of ADME properties and selection of better candidates. However, the use of these tools has generally been to characterize these compounds rather than to select among them. This report discusses applications of the available ADME tools to better understand the clinical implication of these properties, and to optimize these properties. It also provides tracts for timing of studies with respect to the stage of the compound during discovery, by means of a discovery assay by stage (DABS) paradigm. The DABS provide the team with a rationale for the types of studies to be done during hit-to-lead, early and late lead optimization stages of discovery, as well as outlining the deliverables (objectives) at those stages. DABS has proven to be optimal for efficient utilization of resources and helped the discovery team to track the progress of compounds and projects.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Industria Farmacéutica/métodos , Métodos , Preparaciones Farmacéuticas/metabolismo , Farmacocinética
10.
Expert Opin Drug Metab Toxicol ; 1(4): 595-611, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16863427

RESUMEN

Breast cancer resistance protein (BCRP), also known as ABCG2, ABCP and MXR, is a member of the ATP-binding cassette transporter G family. BCRP functions as a biological barrier that extrudes xenobiotics out of cells. The broad substrate specificity and tissue distributions of BCRP in the body make this transporter one of the major efflux transporters in chemotherapy. Recent studies have demonstrated that BCRP exerts a great impact on drug absorption and disposition. This review focuses on the role of BCRP in pharmacokinetics as well as in vitro and in vivo strategies to evaluate hepatic/intestinal BCRP-mediated drug transports and drug-drug interactions. The impacts of polymorphism and gender difference of BCRP are also discussed.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Interacciones Farmacológicas , Proteínas de Neoplasias/fisiología , Farmacocinética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/análisis , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/genética , Absorción , Área Bajo la Curva , Barrera Hematoencefálica , Interacciones Alimento-Droga , Humanos , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Polimorfismo Genético
11.
Curr Drug Metab ; 5(6): 483-505, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15578943

RESUMEN

Cytochrome P450 3A4 (CYP3A4), an enzyme that is highly expressed in the human liver and small intestine, plays a major role in the metabolism of a large variety of xenobiotics, including an estimated 50% of therapeutic drugs, as well as many endogenous compounds. The expression of CYP3A4 can be induced by xenobiotics. Such induction leads to accelerated metabolism of the xenobiotics themselves (autoinduction) or of concomitantly administered CYP3A4 substrates/drugs, thereby significantly altering their pharmacokinetic and pharmacodynamic profiles. During the past decade, much progress has been made in our understanding of the biological mechanisms responsible for regulation of CYP3A4 expression. It is now known that many xenobiotics induce CYP3A4 expression via the pregnane X receptor (PXR) pathway, while others are thought to act through the constitutive androstane receptor (CAR) and the vitamin D receptor (VDR). As a result, most pharmaceutical companies have recognized that it is important to evaluate CYP3A4 induction potential preclinically and are using primary cultures of human hepatocytes and/or PXR reporter gene assays. In general, the results from these two assay methods correlate well. The reporter gene assays in particular can be used to rapidly screen hundreds of drug candidates, whereas methods using primary human hepatocyte cultures may more accurately assess the potential for CYP3A4 induction in vivo. Although it is important to consider CYP3A4 induction in the early stages of the drug development process, it should be recognized that the assessment of induction potential preclinically is a difficult and imprecise endeavor and can be complicated by many factors.


Asunto(s)
Sistema Enzimático del Citocromo P-450/biosíntesis , Diseño de Fármacos , Preparaciones Farmacéuticas/metabolismo , Xenobióticos/metabolismo , Animales , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/genética , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Humanos , Preparaciones Farmacéuticas/administración & dosificación , Xenobióticos/química , Xenobióticos/farmacología
12.
J Pharm Sci ; 102(9): 2953-94, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23798314

RESUMEN

The importance of plasma protein binding (PPB) in modulating the effective drug concentration at pharmacological target sites has been the topic of significant discussion and debate amongst drug development groups over the past few decades. Free drug theory, which states that in absence of energy-dependent processes, after steady state equilibrium has been attained, free drug concentration in plasma is equal to free drug concentration at the pharmacologic target receptor(s) in tissues, has been used to explain pharmacokinetics/pharmacodynamics relationships in a large number of cases. Any sudden increase in free concentration of a drug could potentially cause toxicity and may need dose adjustment. Free drug concentration is also helpful to estimate the effective concentration of drugs that potentially can precipitate metabolism (or transporter)-related drug-drug interactions. Disease models are extensively validated in animals to progress a compound into development. Unbound drug concentration, and therefore PPB information across species is very informative in establishing safety margins and guiding selection of First in Human (FIH) dose and human efficacious dose. The scope of this review is to give an overview of reported role of PPB in several therapeutic areas, highlight cases where PPB changes are clinically relevant, and provide drug metabolism and pharmacokinetics recommendations in discovery and development settings.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Preparaciones Farmacéuticas/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Interacciones Farmacológicas , Humanos , Preparaciones Farmacéuticas/sangre , Unión Proteica
13.
Int J Cell Biol ; 2013: 703545, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840214

RESUMEN

Transport of macromolecules across the blood-brain-barrier (BBB) requires both specific and nonspecific interactions between macromolecules and proteins/receptors expressed on the luminal and/or the abluminal surfaces of the brain capillary endothelial cells. Endocytosis and transcytosis play important roles in the distribution of macromolecules. Due to the tight junction of BBB, brain delivery of traditional therapeutic proteins with large molecular weight is generally not possible. There are multiple pathways through which macromolecules can be taken up into cells through both specific and nonspecific interactions with proteins/receptors on the cell surface. This review is focused on the current knowledge of receptor-mediated endocytosis/transcytosis and brain delivery using the Angiopep-2-conjugated system and the molecular Trojan horses. In addition, the role of neonatal Fc receptor (FcRn) in regulating the efflux of Immunoglobulin G (IgG) from brain to blood, and approaches to improve the pharmacokinetics of therapeutic biologics by generating Fc fusion proteins, and increasing the pH dependent binding affinity between Fc and FcRn, are discussed.

14.
J Clin Pharmacol ; 52(4): 543-51, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21610206

RESUMEN

The objective of the study was to evaluate the effect of hepatic impairment on the pharmacokinetics of tonapofylline. Patients with mild or moderate hepatic impairment were enrolled in parallel with demographically matched healthy subjects. All study participants received a single 75-mg oral tonapofylline capsule. The pharmacokinetic parameters for both tonapofylline and its active metabolite, acyl-glucuronide (tonapofylline-AG), were affected by hepatic impairment significantly (P < .1) except for time to peak plasma concentration (t(max)), terminal half-life (t(½)), and apparent volume of distribution based on the terminal phase (Vdz/F). In the mild group, peak plasma concentration (C(max)), area under the time-concentration curve from time 0 to 48 hours postdose (AUC(48 h)), and from time 0 to infinity (AU(Cinf)) of tonapofylline modestly increased as compared with the control healthy subjects (GMR 1.62, 1.57, and 1.53, respectively). The extent of increase of these parameters for tonapofylline-AG was more profound than tonapofylline with geometric mean ratio (GMR) ranging from 2.02 to 2.08. Moderate hepatic impairment was also associated with modest increases of C(max), AUC(48 h), and AUC(inf) of tonapofylline (GMR 1.41, 1.98, and 2.08, respectively). Similar to the mild group, the increase of these parameters were higher for tonapofylline-AG with GMR ranging from 2.80 to 3.86. Single oral 75-mg tonapofylline was safe and well tolerated in patients with mild or moderate hepatic impairment.


Asunto(s)
Antagonistas del Receptor de Adenosina A1/farmacocinética , Hepatopatías/fisiopatología , Xantinas/farmacocinética , Antagonistas del Receptor de Adenosina A1/efectos adversos , Administración Oral , Anciano , Área Bajo la Curva , Estudios de Casos y Controles , Femenino , Glucurónidos/efectos adversos , Glucurónidos/farmacocinética , Semivida , Humanos , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Xantinas/efectos adversos
15.
J Pharm Sci ; 100(2): 784-93, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20669329

RESUMEN

The incubation of oxymetazoline, a nonprescription nasal decongestant, with human liver microsomes (HLMs) supplemented with uridine-5-diphosphoglucuronic acid (UDPGA) generated glucuronide metabolite as observed by LC/MS/MS. The uridine glucuronosyltransferases (UGTs) responsible for the O-glucuronidation of oxymetazoline remain thus far unidentified. The glucuronide formed in HLMs was identified by LC/MS/MS and characterized by one- and two-dimensional NMR to be the ß-O-glucuronide of oxymetazoline. UGT screening with expressed UGTs identified UGT1A9 as the single UGT isoform catalyzing O-glucuronidation of oxymetazoline. Oxymetazoline O-glucuronidation by using HLMs was best fitted to the allosteric sigmoidal model. The derived S(50) and V(max) values were 2.42 ± 0.40 mM and 8.69 ± 0.58 pmole/(min mg of protein), respectively, and maximum clearance (CL(max)) was 3.61 L/min/mg. Oxymetazoline O-glucuronidation by using expressed UGT1A9 was best fitted to the substrate inhibition model. The derived K(m) and V(max) values were 2.53 ± 1.03 mM and 54.18 ± 16.92 pmole/(min mg of protein), respectively, and intrinsic clearance (CL(int)) was 21.41 L/(min mg). Our studies indicate that oxymetazoline is not glucuronidated at its nanomolar intranasal dose and thus is eliminated unchanged, because UGT1A9 would only contribute to its elimination at the toxic plasma concentrations.


Asunto(s)
Glucurónidos/metabolismo , Glucuronosiltransferasa/metabolismo , Microsomas Hepáticos/metabolismo , Descongestionantes Nasales/metabolismo , Oximetazolina/metabolismo , Uridina Difosfato Ácido Glucurónico/metabolismo , Cromatografía Liquida , Humanos , Cinética , Microsomas Hepáticos/enzimología , Isoformas de Proteínas/metabolismo , Espectrometría de Masas en Tándem , UDP Glucuronosiltransferasa 1A9
16.
Drug Metab Lett ; 4(4): 201-12, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20670210

RESUMEN

Tandutinib is a tyrosine kinase inhibitor under investigation for the treatment of solid and hematological tumors. We evaluated efflux transporter substrate specificity of tandutinib in Caco-2 cells, and the role of efflux transporters in the disposition of tandutinib in rats and efflux transporter knock-out mice. These studies demonstrated that tandutinib is a substrate of P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) in Caco-2 cells. In rats, administration of GF120918, before treatment with tandutinib orally resulted in approximately a seven-fold increase in the mean plasma area under the concentration-versus-time curve (AUC) compared to the vehicle control group. In mice, after intravenous administration of tandutinib, the mean plasma AUC values in the Bcrp1(-/-) mice and Mdr1a/b(-/-) mice was 1.53- and 1.20-fold greater than that of the wild type (WT) mice, respectively. After oral administration, the drug exposure in Mdr1a/b(-/-), Bcrp1(-/-), and Mdr1a/b(-/-)/Bcrp1(-/-) mice was higher than in the WT mice. The brain to plasma exposure ratio (B/P) of tandutinib in Mdr1a/b(-/-) mice increased by 2- to 3-fold over that in the WT mice. There was a 13-fold increase in B/P in Mdr1a/b(-/-)/Bcrp1(-/-) mice. This finding illustrates that P-gp and Bcrp play a role in oral absorption, systemic clearance, and brain penetration of tandutinib in the rodents. P-gp affected oral absorption and brain penetration of tandutinib to a greater extent than Bcrp, but Bcrp contribution to systemic clearance of tandutinib was greater than P-gp. Thus, co-administration of efflux pump inhibitors may be a useful strategy to enhance tandutinib absorption and brain penetration clinically.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas de Neoplasias/metabolismo , Piperazinas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Quinazolinas/farmacocinética , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/deficiencia , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/genética , Acridinas/farmacología , Administración Oral , Animales , Transporte Biológico , Encéfalo/metabolismo , Células CACO-2 , Humanos , Inyecciones Intravenosas , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas de Neoplasias/antagonistas & inhibidores , Piperazinas/administración & dosificación , Piperazinas/sangre , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/sangre , Quinazolinas/administración & dosificación , Quinazolinas/sangre , Ratas , Ratas Sprague-Dawley , Tetrahidroisoquinolinas/farmacología , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
17.
Drug Metab Dispos ; 35(7): 1203-8, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17452418

RESUMEN

Our previous studies described a newly identified potential of grapefruit juice (GFJ) in mediating pharmacokinetic drug interactions due to its capability of esterase inhibition. The current study identifies the active components in GFJ responsible for its esterase-inhibitory effect. The esterase-inhibitory potential of 10 constitutive flavonoids and furanocoumarins toward p-nitrophenylacetate (PNPA) hydrolysis was investigated. The furanocoumarins bergamottin, 6',7'-dihydroxybergamottin, and bergapten, and the glycoside flavonoids naringin and hesperidin, at concentrations found in GFJ or higher, did not inhibit the hydrolysis of PNPA by purified porcine esterase and human liver microsomes. However, the flavonoid aglycones morin, galangin, kaempferol, quercetin, and naringenin showed appreciable inhibition of PNPA hydrolysis in purified porcine esterase, and human and rat liver systems. In Caco-2 cells, demonstrated to contain minimal CYP3A activity, the permeability coefficient of the prodrugs lovastatin and enalapril was increased in the presence of the active flavonoids kaempferol and naringenin, consistent with inhibition of esterase activity. In rats, oral coadministration of kaempferol and naringenin with these prodrugs led to significant increases in plasma exposure to the active acids. In addition, in portal vein-cannulated rats, coadministration of lovastatin with kaempferol (10 mg/kg) led to a 154% and a 113% increase in the portal plasma exposure to the prodrug and active acid, respectively, compared with coadministration with water. The contribution of CYP3A inhibition was demonstrated to be minimal. Overall, a series of flavonoids present in GFJ are identified as esterase inhibitors, of which kaempferol and naringenin are shown to mediate pharmacokinetic drug interaction with the prodrugs lovastatin and enalapril due to their capability of esterase inhibition.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacocinética , Bebidas/análisis , Citrus paradisi , Inhibidores Enzimáticos/farmacología , Esterasas/antagonistas & inhibidores , Flavonoides/farmacología , Interacciones Alimento-Droga , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Administración Oral , Inhibidores de la Enzima Convertidora de Angiotensina/administración & dosificación , Inhibidores de la Enzima Convertidora de Angiotensina/sangre , Animales , Células CACO-2 , Relación Dosis-Respuesta a Droga , Enalapril/administración & dosificación , Enalapril/farmacocinética , Inhibidores Enzimáticos/aislamiento & purificación , Esterasas/metabolismo , Flavanonas/farmacología , Flavonoides/aislamiento & purificación , Frutas , Humanos , Hidrólisis , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/sangre , Técnicas In Vitro , Absorción Intestinal/efectos de los fármacos , Quempferoles/farmacología , Lovastatina/administración & dosificación , Lovastatina/farmacocinética , Masculino , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Nitrofenoles/metabolismo , Ratas , Ratas Sprague-Dawley
18.
Drug Metab Dispos ; 35(1): 79-85, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17020957

RESUMEN

Ketoconazole has generally been used as a standard inhibitor for studying clinical pharmacokinetic drug-drug interactions (DDIs) of drugs that are primarily metabolized by CYP3A4/5. However, ketoconazole at therapeutic, high concentrations also inhibits cytochromes P450 (P450) other than CYP3A4/5, which has made the predictions of DDIs less accurate. Determining the in vivo inhibitor concentration at the enzymatic site is critical for predicting the clinical DDI, but it remains a technical challenge. Various approaches have been used in the literature to estimate the human hepatic free concentrations of this inhibitor, and application of those to predict DDIs has shown some success. In the present study, a novel approach using cryopreserved human hepatocytes suspended in human plasma was applied to mimic the in vivo concentration of ketoconazole at the enzymatic site. The involvement of various P450s in the metabolism of compounds of interest was quantitatively determined (reactive phenotyping). Likewise, the effect of ketoconazole on various P450s was quantitated. Using this information, P450-mediated change in the area under the curve has been predicted without the need of estimating the inhibitor concentrations at the enzyme active site or the K(i). This approach successfully estimated the magnitude of the clinical DDI of an investigational compound, MLX, which is cleared by multiple P450-mediated metabolism. It also successfully predicted the pharmacokinetic DDIs for several marketed drugs (theophylline, tolbutamide, omeprazole, desipramine, midazolam, alprazolam, cyclosporine, and loratadine) with a correlation coefficient (r(2)) of 0.992. Thus, this approach provides a simple method to more precisely predict the DDIs for P450 substrates when coadministered with ketoconazole or any other competitive P450 inhibitors in humans.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Modelos Biológicos , Inhibidores Enzimáticos del Citocromo P-450 , Inhibidores Enzimáticos/metabolismo , Hepatocitos/metabolismo , Humanos , Cetoconazol/metabolismo , Fenotipo , Plasma
19.
Drug Metab Dispos ; 35(4): 576-82, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17220244

RESUMEN

The objective of this study was to investigate whether cyclosporin A (CsA) is a modulator for breast cancer resistance protein (BCRP). The interactions between CsA and BCRP were evaluated by using both membrane- and cell-based assays. CsA inhibited BCRP or BCRP R482T mutant-associated ATPase with an IC(50) of 26.1 and 7.3 microM (31,388 and 8779 ng/ml), respectively, indicating that CsA is a modulator for BCRP and its R482T mutant. The apparent permeability (P(app)) of CsA was not affected by the BCRP-specific inhibitor Ko143 in both apical-to-basolateral (A-to-B) and basolateral-to-apical (B-to-A) directions in hBCRP- or mBcrp-transfected MDCKII cells, whereas CsA at 50 microM significantly increased the A-to-B transport and decreased B-to-A transport of BCRP substrates, [(3)H]estrone-3-sulfate ([(3)H]E3S) and [(3)H]methotrexate ([(3)H]MTX), in hBCRP- and mBcrp1-trasfected MDCKII cells. Similar to cellular transport studies, CsA did not exhibit ATP-dependent uptake in BCRP-expressed membrane vesicles but inhibited the ATP-mediated E3S and MTX uptake in the same vesicles. The inhibitory constant (K(i)) of CsA toward BCRP was 6.7 microM (8507 ng/ml) and 7.8 microM (9380 ng/ml) when using E3S or MTX, respectively, as a BCRP substrate. The inhibitory potency of CsA on BCRP wild type or its R482T mutant was lower than that on P-glycoprotein. The present studies demonstrate that CsA is an inhibitor but not a substrate for BCRP, and has low potential to cause drug-drug interactions with BCRP substrate drugs due to its weak inhibitory effect on BCRP and BCRP R482T mutant at its normal therapeutic blood concentrations (200-400 ng/ml) (Blood 91:362-363, 1998).


Asunto(s)
Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Adenosina Trifosfatasas/antagonistas & inhibidores , Membrana Celular/efectos de los fármacos , Ciclosporina/farmacología , Inhibidores Enzimáticos/farmacología , Inmunosupresores/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Línea Celular , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Ciclosporina/metabolismo , Daunorrubicina/farmacología , Perros , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inhibidores Enzimáticos/metabolismo , Estrona/análogos & derivados , Estrona/metabolismo , Femenino , Humanos , Inmunosupresores/metabolismo , Metotrexato/metabolismo , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transfección
20.
Drug Metab Dispos ; 35(7): 1023-31, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17392396

RESUMEN

This report describes a newly identified potential of grapefruit juice (GFJ) in mediating pharmacokinetic drug interactions due to its capability to inhibit esterase. The study demonstrates that GFJ inhibits purified porcine esterase activity toward p-nitrophenyl acetate and the prodrugs lovastatin and enalapril. In rat and human hepatic or gut S9 fractions and rat gut lumen, GFJ inhibited the hydrolysis of enalapril and lovastatin, which are known to be metabolized principally by esterases, lovastatin being metabolized also by CYP3A. In Caco-2 cells, with minimal CYP3A activity, permeability of these prodrugs was increased in the presence of GFJ. In rats, oral coadministration of GFJ or an esterase inhibitor, bis-(p-nitrophenylphosphate), with the prodrugs led to respective increases in plasma area under the curve by 70% or 57% for enalaprilat and 279% or 141% for lovastatin acid. In addition, portal vein-cannulated rats pretreated with GFJ at -15 and -2 h before lovastatin administration (10 mg/kg p.o.) as a solution, 1) in water and 2) in GFJ, showed, respectively, a 49% increase (CYP3A-inhibited) and a 116% increase (both CYP3A and gut esterase-inhibited) in the portal plasma exposure to the active acid, compared with a non-GFJ pretreatment group. Overall, along with the CYP3A inactivation by GFJ, the decreased esterase activity also played a significant role in increasing the metabolic stability and permeability of esters leading to enhancement of exposure to the active drugs in rats. These new esterase inhibition findings indicate that the potential of drug interaction between ester prodrugs and GFJ should also be considered in the clinic.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacocinética , Bebidas , Citrus paradisi , Inhibidores Enzimáticos/farmacología , Esterasas/antagonistas & inhibidores , Interacciones Alimento-Droga , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Administración Oral , Inhibidores de la Enzima Convertidora de Angiotensina/administración & dosificación , Animales , Células CACO-2 , Permeabilidad de la Membrana Celular/efectos de los fármacos , Enalapril/farmacocinética , Inhibidores Enzimáticos/administración & dosificación , Esterasas/metabolismo , Frutas , Humanos , Hidrólisis , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inyecciones Intravenosas , Absorción Intestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Intestinos/enzimología , Hígado/efectos de los fármacos , Hígado/enzimología , Lovastatina/farmacocinética , Masculino , Nitrofenoles/metabolismo , Nitrofenoles/farmacología , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/enzimología , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA