Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Drug Metab Dispos ; 47(9): 1004-1012, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31201213

RESUMEN

2-(N-acetyl-D-tyrosyl-trans-4-hydroxy-L-prolyl-L-asparaginyl-L-threonyl-L-phenylalanyl) hydrazinocarbonyl-L-leucyl-Nω-methyl-L-arginyl-L-tryptophanamide monoacetate (TAK-448, RVT-602), a kisspeptin analog, has been developed as a therapeutic agent for prostate cancer. The purpose of the present study is to clarify the mechanism of the less than dose-proportional nonlinear pharmacokinetics of TAK-448 after subcutaneous administration to rats. The plasma pharmacokinetics of TAK-448 and radiolabeled TAK-448 ([14C]TAK-448) were examined after subcutaneous and intravenous administrations to rats. [14C]TAK-448 was also subcutaneously injected together with protease inhibitors. The effects of the protease inhibitors on the in vitro metabolism of [14C]TAK-448 were investigated using rat skin homogenates. In a dose-ascending study, less than dose-proportional nonlinear pharmacokinetics were observed after subcutaneous administration with limited absorption of TAK-448 at the highest dose level contrary to the linear pharmacokinetics following intravenous dosing, indicating enhancement of subcutaneous metabolism with dose escalation. The systemic absorption of unchanged TAK-448 recovered when protease inhibitors were subcutaneously coadministered, suggested the involvement of subcutaneous proteases in the first-pass metabolism. An in vitro metabolism study suggests that serine protease could be responsible for the subcutaneous metabolism of TAK-448. Dose-dependent enhancement of first-pass metabolism appears to contribute to the less than dose-proportional nonlinear pharmacokinetics of TAK-448 after subcutaneous administrations to rats.


Asunto(s)
Antineoplásicos/farmacocinética , Kisspeptinas/farmacocinética , Tejido Subcutáneo/metabolismo , Administración Intravenosa , Animales , Antineoplásicos/administración & dosificación , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Humanos , Inyecciones Subcutáneas , Kisspeptinas/administración & dosificación , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/farmacocinética , Ratas
2.
Drug Metab Dispos ; 47(5): 525-534, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30765394

RESUMEN

Fasiglifam, a potent and highly selective agonist of G protein-coupled receptor 40, was developed for the treatment of type 2 diabetes mellitus. However, phase III clinical programs were terminated owing to liver safety concerns. Fasiglifam-related liver toxicity was also observed in repeat-dose dog toxicology studies, characterized by granulomatous inflammation with crystal formation in the liver and/or bile ducts. These histopathological changes were not observed in rat toxicology studies. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry analysis of dog liver sections obtained from a repeat-dose toxicology study indicated that the crystalline material in the affected dog liver contained fasiglifam and fasiglifam glucuronide (fasiglifam-G). Nonclinical mechanistic studies indicated that after 14 days of repeated oral dosing with [14C]fasiglifam at 200 mg/kg per day to dogs, the concentrations of fasiglifam and fasiglifam-G in the bile exceeded the solubility limit of these compounds in the bile (approximately 3000 µg/ml). After single oral 2- and 200-mg/kg doses administered to rats and dogs, fasiglifam and fasiglifam-G concentrations in dog bile were 5- to 10-fold higher than those in rat bile for the same dose of fasiglifam, while the bile flow rate adjusted by body weight was 4- to 8-fold lower in dogs than in rats. High fasiglifam and fasiglifam-G concentrations in dog bile together with lower bile flow rate could cause crystal formation in dog bile, resulting in secondary granulomatous inflammation in the dog liver.


Asunto(s)
Benzofuranos/efectos adversos , Benzofuranos/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Hígado/efectos de los fármacos , Sulfonas/efectos adversos , Sulfonas/metabolismo , Animales , Bilis/metabolismo , Perros , Hígado/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley
3.
Xenobiotica ; 49(7): 833-839, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30044673

RESUMEN

Disposition of 2-(N-acetyl-d-tyrosyl-trans-4-hydroxy-l-prolyl-l-asparaginyl-l-threonyl-l-phenylalanyl) hydrazinocarbonyl-L-leucyl-Nω-methyl-l-arginyl-l-tryptophanamide monoacetate (TAK-448, RVT-602), a synthetic kisspeptin analog, was investigated after parenteral dosing of radiolabeled TAK-448 ([d-Tyr-14C]TAK-448) to rats and dogs, and it was confirmed if the radiolabeling position at d-Tyr was eligible for assessment of in vivo disposition. Dosed radioactivity was rapidly and well absorbed after subcutaneous administration and an appreciable amount of unchanged TAK-448 (TAK-448F) and a hydrolyzed metabolite, M-I, were detected in the plasma of rats and dogs. After intravenous administration of [d-Tyr-14C]TAK-448 to rats, the radioactivity widely distributed to tissues with relatively higher concentrations in kidney and urinary bladder. The radioactivity was decreased rapidly from the tissues. After subcutaneous administration of [d-Tyr-14C]TAK-448 to rats and dogs, the dosed radioactivity was almost completely recovered by 48 and 72 h in rats and dogs, respectively, and most of the radioactivity was excreted in urine after extensive metabolism in the two species. These results suggest that TAK-448 has an acceptable pharmacokinetic profile for clinical evaluation and development, and demonstrate that the synthesized [D-Tyr-14C]TAK-448 used in this study represents a favorable labeling position to evaluate disposition properties of this compound.


Asunto(s)
Riñón/metabolismo , Kisspeptinas , Vejiga Urinaria/metabolismo , Animales , Radioisótopos de Carbono/farmacocinética , Radioisótopos de Carbono/farmacología , Perros , Marcaje Isotópico , Kisspeptinas/farmacocinética , Kisspeptinas/farmacología , Ratas , Ratas Sprague-Dawley
4.
Basic Clin Pharmacol Toxicol ; 122(6): 577-587, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29345044

RESUMEN

TAK-063 is currently being developed to treat schizophrenia. In this study, we investigated the absorption, distribution, metabolism and excretion (ADME) properties of TAK-063 using several paradigms. Following oral administration of TAK-063 at 0.3 mg/kg, bioavailability of TAK-063 was 27.4% in rats and 49.5% in dogs with elimination half-lives of 3.1 hr in rats and 3.7 hr in dogs. TAK-063 is a highly permeable compound without P-glycoprotein (P-gp) or breast cancer resistance protein substrate liability and can be readily absorbed into systemic circulation via the intestine. TAK-063 can also cross the blood-brain barrier. TAK-063 was metabolized mainly by CYP2C8 and CYP3A4/5, while incubation with human liver microsomes produced the major human metabolite, M-I as well as several unknown minor metabolites. Metabolism of TAK-063 to M-I occurs through hydroxylation of the mono-substituted pyrazole moiety. In vitro, TAK-063 was observed to inhibit CYP2C8, CYP2C19 and P-gp with IC50 values of 8.4, 12 and 7.13 µM, respectively. TAK-063 was primarily excreted in the faeces in rats and dogs with M-I as a predominant component. The pre-clinical data from these ADME studies demonstrate a favourable pharmacokinetic profile for TAK-063 with good brain distribution supporting the feasibility of targeting central nervous system regions involved in schizophrenia pathophysiology. TAK-063 has recently been investigated in a phase 2 clinical trial (NCT02477020).


Asunto(s)
Antipsicóticos/farmacocinética , Inhibidores de Fosfodiesterasa/farmacocinética , Hidrolasas Diéster Fosfóricas/efectos de los fármacos , Pirazoles/farmacocinética , Piridazinas/farmacocinética , Animales , Antipsicóticos/metabolismo , Biotransformación , Células CACO-2 , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Perros , Semivida , Humanos , Absorción Intestinal , Masculino , Inhibidores de Fosfodiesterasa/metabolismo , Unión Proteica , Pirazoles/metabolismo , Piridazinas/metabolismo , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Distribución Tisular
5.
Drug Metab Dispos ; 46(3): 204-213, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29269409

RESUMEN

N-{4-Chloro-2-[(1-oxidopyridin-4-yl)carbonyl]phenyl}-4-(propan-2-yloxy)benzenesulfonamide (MLN3126) is an orally available chemokine C-C motif receptor 9 selective antagonist. In nonclinical pharmacokinetic studies of MLN3126, nonextractable radioactivity was observed in plasma after oral administration of 14C-labeled MLN3126 ([14C]MLN3126) to Sprague-Dawley (SD) rats. In this study, the nonextractable radioactive component was digested with trypsin or a nonspecific protease, pronase, after chemical reduction to obtain drug-peptide adducts or drug-amino acid adducts. The chemical structure of these adducts was characterized by liquid chromatography/mass spectrometry. The results demonstrated that the major part of the nonextractable radioactivity was accounted for by covalent binding via the Schiff base formed specifically between the ε-amino group of lysine residue 199 in rat serum albumin and the carbonyl group of MLN3126. The half-life (t1/2) of the total radioactivity in plasma during and after 21 daily multiple oral administrations of [14C]MLN3126 to SD rats was approximately 5-fold shorter than the reported t1/2 of albumin in rats. The data indicated that the covalent binding was reversible under physiologic conditions. The formation of the covalent binding was also confirmed in in vitro incubations with serum albumins from rats, humans, and dogs in the same manner, indicating that there are no qualitative interspecies differences in the formation of the Schiff base.


Asunto(s)
Receptores CCR/antagonistas & inhibidores , Albúmina Sérica/metabolismo , Sulfonamidas/metabolismo , Administración Oral , Animales , Perros , Humanos , Masculino , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , Bencenosulfonamidas
6.
Xenobiotica ; 47(3): 255-266, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27225050

RESUMEN

1. Following oral administration of [14C]TAK-438, the radioactivity was rapidly absorbed in rats and dogs. The apparent absorption of the radioactivity was high in both species. 2. After oral administration of [14C]TAK-438 to rats, the radioactivity in most tissues reached the maximum at 1-hour post-dose. By 168-hour post-dose, the concentrations of the radioactivity were at very low levels in nearly all the tissues. In addition, TAK-438F was the major component in the stomach, whereas TAK-438F was the minor component in the plasma and other tissues. High accumulation of TAK-438F in the stomach was observed after oral and intravenous administration. 3. TAK-438F was a minor component in the plasma and excreta in both species. Its oxidative metabolite (M-I) and the glucuronide of a secondary metabolite formed by non-oxidative metabolism of M-I (M-II-G) were the major components in the rat and dog plasma, respectively. The glucuronide of M-I (M-I-G) and M-II-G were the major components in the rat bile and dog urine, respectively, and most components in feces were other unidentified metabolites. 4. The administered radioactive dose was almost completely recovered. The major route of excretion of the drug-derived radioactivity was via the feces in rats and urine in dogs.


Asunto(s)
Inhibidores de la Bomba de Protones/metabolismo , Pirroles/metabolismo , Sulfonamidas/metabolismo , Animales , Bilis/metabolismo , Perros , Heces , Inhibidores de la Bomba de Protones/farmacocinética , Pirroles/farmacocinética , Ratas , Sulfonamidas/farmacocinética , Distribución Tisular
7.
Xenobiotica ; 47(12): 1027-1034, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27414183

RESUMEN

1. TAK-438, vonoprazan fumarate, is a novel orally active potassium-competitive acid blocker, developed as an antisecretory drug. In this study, we investigated the in vitro metabolism of 14C-labeled TAK-438. In human hepatocytes, M-I, M-II, M-III and M-IV-Sul were mainly formed, and these were also detected in clinical studies. N-demethylated TAK-438 was also formed as an in vitro specific metabolite. Furthermore, CYP3A4 mainly contributed to the metabolism of TAK-438 to M-I, M-III, and N-demethylated TAK-438, and CYP2B6, CYP2C19 and CYP2D6 partly catalyzed the metabolism of TAK-438. The sulfate conjugation by SULT2A1 also contributed to the metabolism of TAK-438 to form TAK-438 N-sulfate, and CYP2C9 mediated the formation of M-IV-Sul from TAK-438 N-sulfate. The metabolite M-IV, which could be another possible intermediate in the formation of M-IV-Sul, was not observed as a primary metabolite of TAK-438 in any of the in vitro studies. 2. In conclusion, TAK-438 was primarily metabolized by multiple metabolizing enzymes including CYP3A4, CYP2B6, CYP2C19, CYP2D6, and a non-CYP enzyme SULT2A1, and the influence of the CYP2C19 genotype status on gastric acid suppression post TAK-438 dosing could be small. The multiple metabolic pathways could also minimize the effects of co-administrated CYP inhibitors or inducers on the pharmacokinetics of TAK-438.


Asunto(s)
Fármacos Gastrointestinales/farmacocinética , Pirroles/farmacocinética , Sulfonamidas/farmacocinética , Citocromo P-450 CYP2C19/metabolismo
8.
Xenobiotica ; 43(2): 182-92, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22867273

RESUMEN

The absorption process in animals of TAK-491, designed as ester-based prodrug with medoxomil moiety, was evaluated. In the plasma of rats and dogs, TAK-536, the pharmacologically active metabolite, was present as the main component with hardly detectable concentrations of TAK-491 after oral administration of TAK-491. In the rat portal plasma, TAK-536 was also present as the main component with hardly detectable concentrations of TAK-491 after jejunal loop injection of TAK-491, suggesting TAK-491 was absorbed from small intestine and hydrolyzed almost completely during absorption. Caco-2 study indicated the permeability of TAK-491 was improved by prodrug modification and the compound could be mainly transferred as TAK-491. This is well consistent with the facts that the AUC and T(max) of TAK-536 after oral administration of TAK-491 were higher and shorter than those after oral administration of TAK-536 in dogs Hydrolysis of TAK-491 is observed not only by the intestinal and hepatic S9 fraction, but also by plasma and human serum albumin. However, medoxomil alcohol wasn't detected during the hydrolysis of TAK-491. These metabolic features of TAK-491 were similar to olmesartan medoxomil, suggesting the hydrolytic pathway and enzymes for TAK-491 when catalyzing to TAK-536 would be the same as olmesartan medoxomil.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacocinética , Bencimidazoles/farmacocinética , Oxadiazoles/farmacocinética , Administración Oral , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/metabolismo , Células CACO-2 , Radioisótopos de Carbono/sangre , Permeabilidad de la Membrana Celular , Perros , Humanos , Hidrólisis , Absorción Intestinal , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Masculino , Oxadiazoles/administración & dosificación , Oxadiazoles/metabolismo , Ratas , Ratas Wistar , Albúmina Sérica
9.
J Mass Spectrom ; 47(8): 1015-22, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22899510

RESUMEN

We investigated the application of a high-resolution Orbitrap mass spectrometer equipped with an electrospray ionization (ESI) source and a matrix-assisted laser desorption/ionization-time-of-flight (MALDI-TOF) mass spectrometer to the metabolite profiling of a model small interfering RNA (siRNA) duplex TSR#34 and compared their functions and capabilities. TSR#34 duplex was incubated in human serum in vitro, and the duplex and its metabolites were then purified by ion exchange chromatography in order to remove the biological matrices. The fraction containing the siRNA duplex and its metabolites was collected and desalted and then subjected to high-performance liquid chromatography (HPLC) equipped with a reversed phase column. The siRNA and its metabolites were separated into single strands by elevated chromatographic temperature and analyzed using the ESI-Orbitrap or the MALDI-TOF mass spectrometer. Using this method, the 5' and/or 3' truncated metabolites of each strand were detected in the human serum samples. The ESI-Orbitrap mass spectrometer enabled differentiation between two possible RNA-based sequences, a monoisotopic molecular mass difference which was less than 2 Da, with an intrinsic mass resolving power. In-source decay (ISD) analysis using a MALDI-TOF mass spectrometer allowed the sequencing of the RNA metabolite with characteristic fragment ions, using 2,4-dihydroxyacetophenone (2,4-DHAP) as a matrix. The ESI-Orbitrap mass spectrometer provided the highest mass accuracy and the benefit of on-line coupling with HPLC for metabolite profiling. Meanwhile, the MALDI-TOF mass spectrometer, in combination with 2,4-DHAP, has the potential for the sequencing of RNA by ISD analysis. The combined use of these methods will be beneficial to characterize the metabolites of therapeutic siRNA compounds.


Asunto(s)
ARN Interferente Pequeño/química , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Acetofenonas/química , Secuencia de Bases , Cromatografía Líquida de Alta Presión , Cromatografía de Fase Inversa , Humanos , Modelos Químicos , Datos de Secuencia Molecular , ARN Interferente Pequeño/sangre , Análisis de Secuencia de ARN
10.
J Med Chem ; 55(8): 3960-74, 2012 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-22490067

RESUMEN

G protein-coupled receptor 40 (GPR40)/free fatty acid receptor 1 (FFA1) is a free fatty acid (FFA) receptor that mediates FFA-amplified glucose-stimulated insulin secretion in pancreatic ß-cells. We previously identified (2,3-dihydro-1-benzofuran-3-yl)acetic acid derivative 2 as a candidate, but it had relatively high lipophilicity. Adding a polar functional group on 2 yielded several compounds with lower lipophilicity and little effect on caspase-3/7 activity at 30 µM (a marker of toxicity in human HepG2 hepatocytes). Three optimized compounds showed promising pharmacokinetic profiles with good in vivo effects. Of these, compound 16 had the lowest lipophilicity. Metabolic analysis of 16 showed a long-acting PK profile due to high resistance to ß-oxidation. Oral administration of 16 significantly reduced plasma glucose excursion and increased insulin secretion during an OGTT in type 2 diabetic rats. Compound 16 (TAK-875) is being evaluated in human clinical trials for the treatment of type 2 diabetes.


Asunto(s)
Benzofuranos/farmacología , Glucosa/farmacología , Insulina/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Sulfonas/farmacología , Animales , Benzofuranos/síntesis química , Benzofuranos/farmacocinética , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Femenino , Prueba de Tolerancia a la Glucosa , Células Hep G2 , Humanos , Secreción de Insulina , Masculino , Ratas , Receptores Acoplados a Proteínas G/efectos de los fármacos , Estereoisomerismo , Sulfonas/síntesis química , Sulfonas/farmacocinética
11.
Biopharm Drug Dispos ; 32(7): 408-25, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21887787

RESUMEN

Ethyl (6R)-6-[N-(2-chloro-4-fluorophenyl)sulfamoyl]cyclohex-1-ene-1-carboxylate (TAK-242) was metabolized to cyclohexene and phenyl ring moieties in non-clinical pharmacokinetic studies and it was suggested that the cyclohexene ring moiety of TAK-242 is tightly bound to endogenous macromolecules. After incubation of TAK-242 and glutathione (GSH) in phosphate buffer (pH 7.4) at 37 °C, TAK-242 reacted with GSH to produce a glutathione conjugate of the cyclohexene ring moiety of TAK-242, which had been observed as a metabolite (M-SG) in non-clinical pharmacokinetic studies. Formation of M-SG was time dependent with a first order reaction and M-I, a metabolite from the phenyl ring moiety of TAK-242, was also produced in parallel. The formation of M-SG was accelerated with increasing pH, therefore it was indicated that TAK-242 reacted with GSH by a nucleophilic substitution reaction. Because glutathione transferase (GST) enhanced M-SG formation in vitro, it is expected that the conjugation of TAK-242 with GSH is also facilitated by GST in vivo in addition to a spontaneous chemical reaction. When radio-labeled TAK-242 ([cyclohexene ring-U-¹4C]TAK-242) was incubated with rat serum albumin (RSA) or human serum albumin (HSA) in vitro, the radioactive material was covalently bound to RSA and HSA, and M-I was generated simultaneously in the reaction mixture. The chemical structure of the TAK-242 adduct covalently bound to HSA was characterized by the accurate mass spectra that cyclohexene ring moiety of TAK-242 was covalently bound to the lysine residue in HSA. The adduct was also detected in the plasma of rats and humans after single i.v. dosing of TAK-242 (in vivo).


Asunto(s)
Sulfonamidas/química , Sulfonamidas/metabolismo , Animales , Glutatión , Humanos , Concentración de Iones de Hidrógeno , Masculino , Estructura Molecular , Ratas , Albúmina Sérica/química , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacocinética , Receptor Toll-Like 4/antagonistas & inhibidores
12.
Nat Struct Biol ; 9(4): 252-6, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11875518

RESUMEN

The Maf family proteins, which constitute a subgroup of basic region-leucine zipper (bZIP) proteins, function as transcriptional regulators of cellular differentiation. Together with the basic region, the Maf extended homology region (EHR), conserved only within the Maf family, defines the DNA binding specific to Mafs. Here we present the first NMR-derived structure of the DNA-binding domain (residues 1-76) of MafG, which contains the EHR and the basic region. The structure consists of three alpha-helices and resembles the fold of the DNA-binding domain of Skn-1, a developmental transcription factor of Caenorhabditis elegans. The structural similarity between MafG and Skn-1 enables us to propose a possible mechanism by which Maf family proteins recognize their consensus DNA sequences.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , ADN/genética , Proteínas de Unión al ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Factor de Transcripción MafG , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Represoras/genética , Alineación de Secuencia , Soluciones , Relación Estructura-Actividad , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA