Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Pharmacol Res Perspect ; 10(6): e01030, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36424908

RESUMO

UDP-glucuronosyltransferases (UGTs) catalyze the conjugation of various substrates with sugars. Since the UGT2 family forms a large cluster spanning 1.5 Mb, transgenic mouse lines carrying the entire human UGT2 family have not been constructed because of limitations in conventional cloning techniques. Therefore, we made a humanized mouse model for UGT2 by chromosome engineering technologies. The results showed that six UGT2 isoforms examined were expressed in the liver of adult humanized UGT2 (hUGT2) mice. Thus, the functions of human UGT2B7 in the liver of hUGT2 mice were evaluated. Glucuronide of azidothymidine (AZT, zidovudine), a typical UGT2B7 substrate, was formed in the liver microsomes of hUGT2 mice but not in the liver microsomes of wild-type and Ugt2-knockout mice. When AZT was intravenously administered, AZT glucuronide was detected in the bile and urine of hUGT2 mice, but it was not detected in the bile and urine of wild-type and Ugt2-knockout mice. These results indicated that the hUGT2 mice express functional human UGT2B7 in the liver. This finding was also confirmed by using gemfibrozil as an alternative UGT2B7 substrate. Gemfibrozil glucuronide was formed in the liver microsomes of hUGT2 mice and was mainly excreted in the bile of hUGT2 mice after intravenous dosing of gemfibrozil. This hUGT2 mouse model will enable improved predictions of pharmacokinetics, urinary and biliary excretion and drug-drug interactions mediated by human UGT2, at least UGT2B7, in drug development research and basic research.


Assuntos
Glucuronídeos , Zidovudina , Humanos , Camundongos , Animais , Glucuronídeos/metabolismo , Genfibrozila , Camundongos Knockout , Camundongos Transgênicos , Cromossomos/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(8): 3072-3081, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30718425

RESUMO

Although "genomically" humanized animals are invaluable tools for generating human disease models as well as for biomedical research, their development has been mainly restricted to mice via established transgenic-based and embryonic stem cell-based technologies. Since rats are widely used for studying human disease and for drug efficacy and toxicity testing, humanized rat models would be preferred over mice for several applications. However, the development of sophisticated humanized rat models has been hampered by the difficulty of complex genetic manipulations in rats. Additionally, several genes and gene clusters, which are megabase range in size, were difficult to introduce into rats with conventional technologies. As a proof of concept, we herein report the generation of genomically humanized rats expressing key human drug-metabolizing enzymes in the absence of their orthologous rat counterparts via the combination of chromosome transfer using mouse artificial chromosome (MAC) and genome editing technologies. About 1.5 Mb and 700 kb of the entire UDP glucuronosyltransferase family 2 and cytochrome P450 family 3 subfamily A genomic regions, respectively, were successfully introduced via the MACs into rats. The transchromosomic rats were combined with rats carrying deletions of the endogenous orthologous genes, achieved by genome editing. In the "transchromosomic humanized" rat strains, the gene expression, pharmacokinetics, and metabolism observed in humans were well reproduced. Thus, the combination of chromosome transfer and genome editing technologies can be used to generate fully humanized rats for improved prediction of the pharmacokinetics and drug-drug interactions in humans, and for basic research, drug discovery, and development.


Assuntos
Citocromo P-450 CYP3A/genética , Edição de Genes , Glucuronosiltransferase/genética , Inativação Metabólica/genética , Animais , Técnicas de Transferência de Genes , Genoma , Humanos , Taxa de Depuração Metabólica/genética , Camundongos , Camundongos Transgênicos , Ratos
3.
J Epidemiol ; 29(10): 384-390, 2019 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-30224580

RESUMO

BACKGROUND: It is unclear whether either neighborhood collective efficacy or school collective efficacy is associated with adolescent alcohol use. This study aimed to examine the relative contributions of collective efficacy, both in school and in the neighborhood contexts, to alcohol use among Japanese adolescents. METHODS: A cross-sectional study was conducted in public high schools across Okinawa and Ibaraki Prefectures in Japan in 2016. The study participants consisted of 3,291 students in grades 10 through 12 cross-nested in 51 schools and 107 neighborhoods. Alcohol use was measured as current alcohol drinking, which was defined as self-reported drinking on at least 1 day in the past 30 days. Collective efficacy was measured using scales of social cohesion and informal social control in school and the neighborhood. Contextual-level collective efficacy was measured using aggregated school-level and neighborhood-level individual responses, respectively. We used non-hierarchical multilevel models to fit the cross-nested data. RESULTS: Significant variation in alcohol use was shown between schools but not between neighborhoods. After adjusting for covariates, school collective efficacy at individual- and contextual-levels was protectively associated with alcohol drinking (odds ratio [OR] for the increase of one standard deviation from the mean 0.72; 95% confidence interval [CI], 0.63-0.82 and OR 0.61; 95% CI, 0.49-0.75, respectively), whereas neighborhood collective efficacy at individual- and contextual-levels was not associated with alcohol consumption. CONCLUSION: The school-level associations of collective efficacy with adolescent alcohol use may have the greater impact than the neighborhood-level associations. Adolescent drinking prevention efforts should include enhancing school collective efficacy.


Assuntos
Comportamento do Adolescente/psicologia , Consumo de Bebidas Alcoólicas/psicologia , Influência dos Pares , Características de Residência/estatística & dados numéricos , Instituições Acadêmicas , Autoeficácia , Estudantes/psicologia , Consumo de Álcool por Menores/psicologia , Adolescente , Comportamento do Adolescente/etnologia , Consumo de Bebidas Alcoólicas/etnologia , Estudos Transversais , Feminino , Humanos , Masculino , Análise Multinível , Fatores Socioeconômicos , Estudantes/estatística & dados numéricos , Inquéritos e Questionários , Consumo de Álcool por Menores/etnologia , Consumo de Álcool por Menores/estatística & dados numéricos
4.
Xenobiotica ; 46(11): 967-76, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26899628

RESUMO

1. In prior studies, it has been shown that tivantinib is extensively metabolized in humans to many oxidative metabolites and glucuronides. In order to identify the responsible enzymes, we investigated the in vitro metabolism of tivantinib and its four major circulating metabolites. 2. The primary isoforms involved in the elimination of tivantinib were CYP2C19 and CYP3A4/5. CYP2C19 showed catalytic activity for the formation of M5 (hydroxylated metabolite), but not for M4 (a stereoisomer of M5), whereas CYP3A4/5 catalyzed the formation of both metabolites. For the elimination of M4, M5 and M8 (keto-metabolite), CYP3A4/5 was the major cytochrome P450 isoform and UGT1A9 was mainly involved in the glucuronidation of M4 and M5. 3. ADH4 was identified as one of the major alcohol dehydrogenase isoforms contributing to the formation of M6 (sequential keto-metabolite of M4 and M5) and M8. The substrate preference of ADH for M4, and not M5, was observed in the formation of M6. 4. In conclusion, CYP2C19, CYP3A4/5, UGT1A9 and ADH4 were the primary drug metabolizing enzymes involved in the in vitro metabolism of tivantinib and its metabolites. The stereoselective hydroxylation by CYP2C19 and substrate stereoselectivity of ADH4-catalyzed oxidation in the in vitro metabolism of tivantinib was discovered.


Assuntos
Álcool Desidrogenase/metabolismo , Antineoplásicos/metabolismo , Citocromo P-450 CYP2C19/metabolismo , Pirrolidinonas/metabolismo , Quinolinas/metabolismo , Humanos , Hidroxilação , Oxirredução
5.
Drug Metab Dispos ; 44(1): 115-23, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26558823

RESUMO

Prasugrel is a thienopyridine antiplatelet prodrug that undergoes rapid hydrolysis in vivo to a thiolactone metabolite by human carboxylesterase-2 (hCE2) during gastrointestinal absorption. The thiolactone metabolite is further converted to a pharmacologically active metabolite by cytochrome P450 isoforms. The aim of the current study was to elucidate hydrolases other than hCE2 involved in the bioactivation step of prasugrel in human intestine. Using size-exclusion column chromatography of a human small intestinal S9 fraction, another peak besides the hCE2 peak was observed to have prasugrel hydrolyzing activity, and this protein was found to have a molecular weight of about 20 kDa. This prasugrel hydrolyzing protein was successfully purified from a monkey small intestinal cytosolic fraction by successive four-step column chromatography and identified as Raf-1 kinase inhibitor protein (RKIP) by liquid chromatography-tandem mass spectrometry. Second, we evaluated the enzymatic kinetic parameters for prasugrel hydrolysis using recombinant human RKIP and hCE2 and estimated the contributions of these two hydrolyzing enzymes to the prasugrel hydrolysis reaction in human intestine, which were approximately 40% for hRKIP and 60% for hCE2. Moreover, prasugrel hydrolysis was inhibited by anti-hRKIP antibody and carboxylesterase-specific chemical inhibitor (bis p-nitrophenyl phosphate) by 30% and 60%, respectively. In conclusion, another protein capable of hydrolyzing prasugrel to its thiolactone metabolite was identified as RKIP, and this protein may play a significant role with hCE2 in prasugrel bioactivation in human intestine. RKIP is known to have diverse functions in many intracellular signaling cascades, but this is the first report describing RKIP as a hydrolase involved in drug metabolism.


Assuntos
Hidrolases/metabolismo , Intestino Delgado/enzimologia , Proteína de Ligação a Fosfatidiletanolamina/metabolismo , Inibidores da Agregação Plaquetária/metabolismo , Cloridrato de Prasugrel/metabolismo , Ativação Metabólica , Animais , Carboxilesterase/antagonistas & inibidores , Carboxilesterase/metabolismo , Catálise , Cromatografia em Gel , Cromatografia Líquida , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Hidrolases/antagonistas & inibidores , Hidrólise , Intestino Delgado/efeitos dos fármacos , Cinética , Macaca fascicularis , Masculino , Modelos Biológicos , Proteína de Ligação a Fosfatidiletanolamina/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Espectrometria de Massas em Tandem
6.
Biopharm Drug Dispos ; 36(6): 352-363, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25765700

RESUMO

Loxoprofen (LX) is a prodrug-type non-steroidal anti-inflammatory drug which is used not only as an oral drug but also as a transdermal formulation. As a pharmacologically active metabolite, the trans-alcohol form of LX (trans-OH form) is generated after oral administration to humans. The objectives of this study were to evaluate the generation of the trans-OH form in human in vitro skin and to identify the predominant enzyme for its generation. In the permeation and metabolism study using human in vitro skin, both the permeation of LX and the formation of the trans-OH form increased in a time- and dose-dependent manner after the application of LX gel to the skin. In addition, the characteristics of permeation and metabolism of both LX and the trans-OH form were examined by a mathematical pharmacokinetic model. The Km value was calculated to be 10.3 mm in the human in vitro skin. The predominant enzyme which generates the trans-OH form in human whole skin was identified to be carbonyl reductase 1 (CBR1) by immunodepletion using the anti-human CBR1 antibody. The results of the enzyme kinetic study using the recombinant human CBR1 protein demonstrated that the Km and Vmax values were 7.30 mm and 402 nmol/min/mg protein, respectively. In addition, it was found that no unknown metabolites were generated in the human in vitro skin. This is the first report in which LX is bioactivated to the trans-OH form in human skin by CBR1. Copyright © 2015 John Wiley & Sons, Ltd.

8.
Xenobiotica ; 44(11): 1026-38, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24916900

RESUMO

1. Loxoprofen (LX), is a prodrug of the pharmacologically active form, trans-alcohol metabolite (trans-OH form), which shows very potent analgesic effect. In this study, the pharmacokinetics and metabolism of [(14)C]LX-derived radioactivity after dermal application of [(14)C]LX gel (LX-G) to rats were evaluated. 2. The area under concentration-time curve (AUC0-∞) of radioactivity in the plasma after the dermal application was 13.6% of that of the oral administration (p < 0.05). 3. After the dermal application, the radioactivity remained in the skin and skeletal muscle at the treated site for 168 h, whereas the AUC0-168 h of the radioactivity concentration in every tissue examined except the treated site was statistically lower than that after the oral administration (p < 0.05). 4. The trans-OH form was observed at high levels in the treated skin site at 0.5 h. Metabolite profiles in plasma, non-treated skin site and urine after the dermal application were comparable with those after the oral administration. 5. Renal excretion was the main route of elimination after the dermal application. 6. In conclusion, compared to the oral administration, the dermal application of [(14)C]LX-G showed lower systemic and tissue exposure with higher exposure in the therapeutic target site. The radioactivity revealed similar metabolite profiles in both administration routes.


Assuntos
Fenilpropionatos/farmacocinética , Administração Cutânea , Animais , Radioisótopos de Carbono/análise , Masculino , Metaboloma , Ratos , Ratos Wistar , Absorção Cutânea , Distribuição Tecidual
9.
Drug Metab Dispos ; 42(7): 1138-45, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24733788

RESUMO

Prasugrel, a thienopyridine antiplatelet drug, is converted in animals and humans to the pharmacologically active metabolite R-138727 [(2Z)-{1-[(1RS)-2-cyclopropyl-1-(2-fluorophenyl)-2-oxoethyl]-4-sulfanylpiperidin-3-ylidene}ethanoic acid], which has two chiral centers, occurring as a mixture of four isomers. The RS and RR isomers are more active than the SS and SR isomers (RS > RR > > SR = SS). The pharmacologically active metabolite is further metabolized to an S-methylated metabolite that is the major identified inactive metabolite in humans. In rat, dog, and human liver microsomes supplemented with S-adenosyl methione, the SS and SR isomers of the active metabolite were extensively S-methylated while the RS and RR isomers were not. Addition of 2,3-dichloromethyl benzylamine (50 µM) completely inhibited the S-methylation reaction, indicating that the microsomal and cytosolic thiol methyltransferase but not the cytosolic thiopurine S-methyltransferase is involved in the methylation. The hepatic intrinsic clearance values for methylation of the RS, RR, SS, and SR isomers (ml/min/kg) were 0, 0, 40.4, and 37.6, respectively, in rat liver microsomes, 0, 0, 11.6, and 2.5, respectively, in dog liver microsomes, and 0, 0, 17.3, and 17.7, respectively, in human liver microsomes, indicating that the RS and RR isomers are not methylated in vitro and that the methylation of SS and SR isomers is high with rat > human > dog. This finding in vitro agreed well with the in vivo observation in rats and dogs, where the S-methylated SS and SR isomers were the major metabolites in the plasma whereas negligible amounts of S-methylated RS and RR isomers were detected after intravenous administration of the pharmacologically active metabolites.


Assuntos
Metiltransferases/metabolismo , Microssomos Hepáticos/enzimologia , Piperazinas/metabolismo , Antagonistas do Receptor Purinérgico P2Y/metabolismo , Tiofenos/metabolismo , Animais , Cromatografia Líquida , Cães , Humanos , Masculino , Metilação , Cloridrato de Prasugrel , Ratos , Ratos Sprague-Dawley , Espectrofotometria Ultravioleta
10.
Drug Metab Dispos ; 42(4): 685-94, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24487120

RESUMO

6ß-Hydroxycortisol (6ß-OHF) is a substrate of the organic anion transporter 3 (OAT3) and the multidrug and toxin extrusion proteins MATE1 and MATE-2K in the corresponding cDNA-transfected cells. This study aimed to examine the contribution of OAT3 and MATEs to the urinary excretion of 6ß-OHF in humans using the appropriate in vivo inhibitors, probenecid and pyrimethamine, for OAT3 and MATEs, respectively. Oat3(-/-) mice showed significantly reduced renal clearance of 6ß-OHF (CL(renal, 6ß-OHF)) compared with wild-type mice (18.1 ± 1.5 versus 7.60 ± 1.8 ml/min/kg). 6ß-OHF uptake by human kidney slices was inhibited significantly by probenecid to 20-45% of the control values and partly by 1-methyl-4-phenylpyridinium. 6ß-OHF plasma concentration and the amount of 6ß-OHF excreted into the urine (X(6ß-OHF)) were measured in healthy subjects enrolled in drug-drug interaction studies of benzylpenicillin alone or with probenecid (study 1), adefovir alone or with probenecid (study 2), and metformin alone or with pyrimethamine (study 3). Probenecid treatment caused a 57 and 76% increase in the area under the plasma concentration-time curve for 6ß-OHF (AUC(6ß-OHF)) in studies 1 and 2, respectively, but did not affect X(6ß-OHF). Consequently, CL(renal, 6ß-OHF) (milliliters per minute) decreased significantly from 231 ± 11 to 135 ± 9 and from 225 ± 26 to 141 ± 12 after probenecid administration in studies 1 and 2, respectively. By contrast, neither AUC(6ß-OHF) nor CL(renal, 6ß-OHF) was significantly altered by pyrimethamine administration. Taken together, these data suggest that OAT3 plays a significant role in the urinary excretion of 6ß-OHF, and that 6ß-OHF can be used to investigate the perpetrators of the pharmacokinetic drug interactions involving OAT3 in humans.


Assuntos
Interações Medicamentosas , Hidrocortisona/análogos & derivados , Rim/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Preparações Farmacêuticas/metabolismo , Adulto , Animais , Biomarcadores/sangue , Biomarcadores/urina , Técnicas de Cultura de Células , Feminino , Células HEK293 , Voluntários Saudáveis , Humanos , Hidrocortisona/sangue , Hidrocortisona/urina , Rim/efeitos dos fármacos , Masculino , Taxa de Depuração Metabólica , Camundongos , Camundongos Knockout , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Especificidade por Substrato , Adulto Jovem
11.
Drug Metab Dispos ; 41(5): 1156-62, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23471504

RESUMO

Olmesartan medoxomil (OM) is a prodrug-type angiotensin II type 1 receptor antagonist. OM is rapidly converted into its active metabolite olmesartan by multiple hydrolases in humans, and we recently identified carboxymethylenebutenolidase homolog (CMBL) as one of the OM bioactivating hydrolases. In the present study, we further investigated the interindividual variability of mRNA and protein expression of CMBL and OM-hydrolase activity using 40 individual human liver and 30 intestinal specimens. In the intestinal samples, OM-hydrolase activity strongly correlated with the CMBL protein expression, clearly indicating that CMBL is a major contributor to the prodrug bioactivation in human intestine. The protein and activity were highly distributed in the proximal region (duodenum and jejunum) and decreased to the distal region of the intestine. Although there was high interindividual variability (16-fold) in both the protein and activity in the intestinal segments from the duodenum to colon, the interindividual variability in the duodenum and jejunum was relatively small (3.0- and 2.4-fold, respectively). In the liver samples, the interindividual variability in the protein and activity was 4.1- and 6.8-fold, respectively. No sex differences in the protein and activity were shown in the human liver or intestine. A genetically engineered Y155C mutant of CMBL, which was caused by a single nucleotide polymorphism rs35489000, showed significantly lower OM-hydrolase activity than the wild-type protein although no minor allele was genotyped in the 40 individual liver specimens.


Assuntos
Hidrolases de Éster Carboxílico/metabolismo , Intestinos/enzimologia , Fígado/enzimologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Hidrolases de Éster Carboxílico/genética , Primers do DNA , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/genética , Adulto Jovem
12.
Pharm Res ; 30(2): 447-57, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23073666

RESUMO

PURPOSE: To examine the effect of the fluoroquinolone DX-619 on CYP3A4 and urinary excretion of 6ß-hydroxycortisol, an endogenous probe of hepatic CYP3A4 activity, in healthy subjects. METHODS: The effect of DX-619 on CYP3A4 was examined in human liver microsomes. The apparent formation and renal clearance of 6ß-hydroxycortisol (CL(6ß-OHF) and CL(renal,6ß-OHF), respectively) were determined in placebo- and DX-619-treated subjects. 6ß-hydroxycortisol uptake was determined in HEK293 cells expressing OAT1, OAT3, OCT2, MATE1, and MATE2-K. RESULTS: DX-619 was a mechanism-based inhibitor of CYP3A4, with K(I) and k(inact) of 67.9 ± 7.3 µmol/l and 0.0730 ± 0.0033 min(-1), respectively. Pharmacokinetic simulation suggested in vivo relevance of CYP3A4 inhibition by DX-619. CL(6ß-OHF) and CL(renal,6ß-OHF) were decreased 72% and 70%, respectively, on day 15 in DX-619-treated group compared with placebo (P < 0.05). 6ß-hydroxycortisol was a substrate of OAT3 (K(m) = 183 ± 25 µmol/l), OCT2, MATE1, and MATE2-K. Maximum unbound concentration of DX-619 (9.1 ± 0.4 µmol/l) was above K(i) of DX-619 for MATE1 (4.32 ± 0.79 µmol/l). CONCLUSIONS: DX-619 caused a moderate inhibition of hepatic CYP3A4-mediated formation and significant inhibition of MATE-mediated efflux of 6ß-hydroxycortisol into urine. Caution is needed in applying CL(6ß-OHF) as an index of hepatic CYP3A4 activity without evaluating CL(renal,6ß-OHF).


Assuntos
Antibacterianos/metabolismo , Citocromo P-450 CYP3A/metabolismo , Hidrocortisona/análogos & derivados , Microssomos Hepáticos/metabolismo , Pirrolidinas/metabolismo , Quinolonas/metabolismo , Feminino , Células HEK293 , Humanos , Hidrocortisona/metabolismo , Hidrocortisona/urina , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Transportador 2 de Cátion Orgânico
13.
Pharm Res ; 29(11): 3143-55, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22729371

RESUMO

PURPOSE: Recombinant osteoprotegerin (OPG) has been proven to be useful for treating various bone disorders such as osteoporosis. To improve its in vivo pharmacological effect, OPG was conjugated to novel comb-shaped co-polymers of polyethylene glycol (PEG) allylmethylether and maleamic acid (poly(PEG), 5 kDa). Biodistribution and bioactivity were evaluated. METHODS: OPG was conjugated via lysine to poly(PEG) and to linear PEG (0.5 kDa and 5 kDa). Poly(PEG)-OPG was compared with linear PEG0.5k-OPG and PEG5k-OPG in terms of in vitro and in vivo efficacy and bone distribution. RESULTS: The in vitro receptor binding study showed that poly(PEG)-OPG could be the most bioactive among the three PEG-OPG derivatives. Pharmacokinetic studies in ovariectomized (OVX) rats showed that serum half-life and AUC of poly(PEG)-OPG were comparable with those of linear PEG-OPG derivatives. For in vivo pharmacological effect, poly(PEG)-OPG showed the strongest inhibitory effect on bone resorption activity in OVX rats. Poly(PEG)-OPG demonstrated enhanced bone marrow distribution with higher selectivity than linear PEG5k-OPG. CONCLUSION: Poly(PEG) modification could provide longer residence time in serum and higher bone-marrow specific delivery of OPG, leading to a higher in vivo pharmacological effect.


Assuntos
Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoprotegerina/administração & dosagem , Osteoprotegerina/química , Polietilenoglicóis/administração & dosagem , Administração Intravenosa , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/metabolismo , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Maleatos/administração & dosagem , Maleatos/química , Osteoclastos/metabolismo , Osteoprotegerina/farmacocinética , Ovariectomia/métodos , Polietilenoglicóis/química , Polímeros/administração & dosagem , Polímeros/química , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética , Relação Estrutura-Atividade , Distribuição Tecidual
14.
Drug Metab Dispos ; 40(9): 1854-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22733806

RESUMO

Clopidogrel is a thienopyridine antiplatelet agent that is converted to the active metabolite, R-361015, in vivo. Clopidogrel is first oxidized to a thiolactone intermediate R-115991. R-115991 is thought to be metabolized to a GSH conjugate of R-361015 (R-361015-SG) and then is reduced to R-361015 in the presence of GSH. In this study, we investigated the enzyme-mediated formation of R-361015 from R-361015-SG in human liver microsomes and cytosols. After incubation of R-115991 in human liver microsomes, the formation of R-361015-SG, and subsequently of R-361015, was observed. The apparent formation rate of R-361015-SG was markedly decreased when human liver cytosols were added. Fitting the data to the kinetic model showed that the rate constant of R-361015-SG reduction to R-361015 in human liver microsomes was approximately 20-fold higher in the presence of human liver cytosols (6.56 min⁻¹) than in the absence of cytosols (0.326 min⁻¹). In addition, the formation rate of R-361015 from R-361015-SG was higher in human liver cytosols (2843 ± 1176 pmol · min⁻¹ · mg⁻¹) compared with in human liver microsomes (508 ± 396 pmol · min⁻¹ · mg⁻¹). The formation of R-361015 from R-361015-SG in human liver microsomes or cytosols was inhibited by anti-human glutaredoxin antibody in a concentration-dependent manner. Recombinant human glutaredoxin mediated the formation of R-361015 from R-361015-SG with the K(m) and V(max) values of 30.0 ± 1.3 µM and 381.6 ± 209.8 pmol · min⁻¹ · µg⁻¹, respectively. The intrinsic clearance value (V(max)/K(m)) was 12.9 ± 7.5 µl · min⁻¹ · µg⁻¹. In conclusion, we found that human glutaredoxin is a main contributor to the formation of the pharmacologically active metabolite of clopidogrel from its GSH conjugate in human liver.


Assuntos
Glutarredoxinas/metabolismo , Glutationa/metabolismo , Fígado/enzimologia , Inibidores da Agregação Plaquetária/metabolismo , Pró-Fármacos/metabolismo , Ticlopidina/análogos & derivados , Biotransformação , Clopidogrel , Inibidores Enzimáticos/farmacologia , Glutarredoxinas/antagonistas & inibidores , Glutationa/análogos & derivados , Humanos , Cinética , Fígado/efeitos dos fármacos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Modelos Biológicos , Oxirredução , Proteínas Recombinantes/metabolismo , Ticlopidina/metabolismo
15.
Bioanalysis ; 4(3): 263-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22303830

RESUMO

BACKGROUND: Drug-induced toxicity such as idiosyncratic drug toxicity is believed to be reduced when either reactive metabolite formation or exposure to a drug is minimized. The objective of the present study was therefore to clarify the relationship between the daily doses, the formation rates of reactive metabolite adduct with (35)S-glutathione (RM-GS) and the safety profiles of compounds. RESULTS: The RM-GS formation rates for 113 test compounds were determined by incubation with human liver microsomes, and the test compounds were classified into three categories of safe, warning and withdrawn/black box warning. A total of 23 out of 28 withdrawn/black box warning drugs showed both a RM-GS formation rate of over 1 pmol/30 min/mg protein and a dose of over 100 mg. CONCLUSION: These results suggest that when compounds are plotted in this region, the compounds would have a relatively high idiosyncratic drug toxicity potential.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Glutationa/química , Preparações Farmacêuticas/metabolismo , Diclofenaco/química , Diclofenaco/metabolismo , Diclofenaco/toxicidade , Esquema de Medicação , Glutationa/metabolismo , Humanos , Microssomos Hepáticos/metabolismo , Radioisótopos de Enxofre/química , Ticlopidina/química , Ticlopidina/metabolismo , Ticlopidina/toxicidade
16.
Drug Metab Pharmacokinet ; 27(3): 354-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22146109

RESUMO

We have previously reported that human total body clearance (CL) and steady-state volume of distribution (Vss) of monoclonal antibodies (mAbs) could be predicted reasonably well from monkey data alone using simple allometry with scaling exponents of 0.79 and 1.12 (for soluble targets), and 0.96 and 1.00 (for membrane-bound targets). In the present study, to predict the plasma concentration-time profiles of mAbs in humans, we employed simple dose-normalization and species-invariant time methods (elementary Dedrick plot and complex Dedrick plot), based on the monkey data and the scaling exponents we previously determined. The results demonstrated that the species-invariant time methods were able to provide higher accuracy of prediction than simple dose-normalization, regardless of the type of target antigens (soluble or membrane-bound). The accuracy between elementary Dedrick plot and complex Dedrick plot was nearly equivalent. The predicted human CL and Vss using species-invariant time methods were within mostly 2-fold differences from the observed values. The prediction not only of pharmacokinetic (PK) parameters but also of the plasma concentration-time profile in humans can serve as guidelines for better planning of clinical studies on mAbs.


Assuntos
Anticorpos Monoclonais/farmacocinética , Modelos Biológicos , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/sangue , Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais Murinos/administração & dosagem , Anticorpos Monoclonais Murinos/sangue , Anticorpos Monoclonais Murinos/farmacocinética , Antígenos/química , Relação Dose-Resposta a Droga , Meia-Vida , Humanos , Macaca fascicularis , Macaca mulatta , Taxa de Depuração Metabólica , Pan troglodytes , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/sangue , Proteínas Recombinantes de Fusão/farmacocinética , Solubilidade , Especificidade da Espécie
17.
Drug Metab Dispos ; 40(2): 374-80, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22086979

RESUMO

Olmesartan medoxomil (OM) is a prodrug-type angiotensin II type 1 receptor antagonist. The OM-hydrolyzing enzyme responsible for prodrug bioactivation was purified from human plasma through successive column chromatography and was molecularly identified through N-terminal amino acid sequencing, which resulted in a sequence of 20 amino acids identical to that of human paraoxonase 1 (PON1). Two recombinant allozymes of human PON1 (PON1(192QQ) and PON1(192RR)) were constructed and were clearly demonstrated to hydrolyze OM; hydrolysis by the latter allozyme was slightly faster than that by the former. In addition, we evaluated the contribution of PON1 to OM bioactivation in human plasma. Enzyme kinetic studies demonstrated that OM was hydrolyzed more effectively by the recombinant PON1 proteins than by purified albumin. The OM-hydrolyzing activities of the recombinant PON1 proteins and diluted plasma were greatly reduced in the absence of calcium ions. Immunoprecipitation with anti-PON1 IgG completely abolished the OM-hydrolyzing activity in human plasma, whereas the activity was partially inhibited with anti-albumin IgG. The distribution pattern of the OM-hydrolyzing activity in human serum lipoprotein fractions and lipoprotein-deficient serum was examined and showed that most of the OM-hydrolyzing activity was located in the high-density lipoprotein fraction, with which PON1 is closely associated. In conclusion, we identified PON1 as the OM-bioactivating hydrolase in human plasma on a molecular basis and demonstrated that PON1, but not albumin, plays a major role in OM bioactivation in human plasma.


Assuntos
Bloqueadores do Receptor Tipo 2 de Angiotensina II/metabolismo , Anti-Hipertensivos/metabolismo , Arildialquilfosfatase/metabolismo , Imidazóis/metabolismo , Pró-Fármacos/metabolismo , Tetrazóis/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Arildialquilfosfatase/química , Arildialquilfosfatase/genética , Arildialquilfosfatase/isolamento & purificação , Cálcio/metabolismo , Humanos , Hidrólise , Isoenzimas/sangue , Isoenzimas/química , Isoenzimas/isolamento & purificação , Isoenzimas/metabolismo , Cinética , Lipoproteínas HDL/metabolismo , Proteínas Mutantes/sangue , Proteínas Mutantes/química , Proteínas Mutantes/isolamento & purificação , Proteínas Mutantes/metabolismo , Olmesartana Medoxomila , Plasma/enzimologia , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Análise de Sequência de Proteína , Especificidade da Espécie , Especificidade por Substrato
18.
Drug Metab Dispos ; 39(10): 1946-53, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21712433

RESUMO

KAI-9803 is composed of a selective δ-protein kinase C (δPKC) inhibitor peptide derived from the δV1-1 portion of δPKC (termed "cargo peptide"), conjugated reversibly to the cell-penetrating peptide 11-amino acid, arginine-rich sequence of the HIV type 1 transactivator protein (TAT47₋57; termed "carrier peptide") via a disulfide bond. KAI-9803 administration at the end of ischemia has been found to reduce cardiac damage caused by ischemia-reperfusion in a rat model of acute myocardial infarction. In the study presented here, we examined the TAT47₋57-mediated distribution of KAI-9803 in rats after a single intravenous bolus administration (1 mg/kg). ¹4C-KAI-9803 was rapidly delivered to many tissues, including the heart (1.21 µg eq/g tissue), while being quickly cleared from the systemic circulation. The microautoradiography analysis showed that ¹4C-KAI-9803 was effectively delivered into various cells, including cardiac myocytes and cardiac endothelial cells within 1 min after dosing. The tissue distribution of ¹²5I-labeled KAI-9803 was compared to that of ¹²5I-labeled cargo peptide; this comparison demonstrated that the distribution of KAI-9803 to tissues such as the liver, kidney, and heart was facilitated by the reversible conjugation to TAT47₋57. In an in vitro cardiomyocyte study, the extent of ¹²5I-KAI-9803 internalization was greater at 37°C than that at 4°C, whereas the internalization of the ¹²5I-cargo peptide at 37°C was not observed, indicating that the uptake of ¹²5I-KAI-9803 into the cardiomyocytes was mediated by the TAT47₋57 carrier. Our studies demonstrated that after a single intravenous administration, KAI-9803 can be delivered into the target cells in the liver, kidney, and heart by a TAT47₋57-mediated mechanism.


Assuntos
Peptídeos/farmacocinética , Proteína Quinase C-delta/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacocinética , Animais , Autorradiografia/métodos , Peptídeos Penetradores de Células/metabolismo , Células Cultivadas , Produtos do Gene tat/metabolismo , Injeções Intravenosas , Masculino , Miócitos Cardíacos/metabolismo , Peptídeos/administração & dosagem , Proteína Quinase C-delta/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
19.
Drug Metab Pharmacokinet ; 26(4): 423-30, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21606605

RESUMO

Interspecies allometric scaling is a useful tool for calculating human pharmacokinetic (PK) parameters from data in animals. In this study, in order to determine the scaling exponent in a simple allometric equation that can predict human clearance (CL) and distribution volume at steady state (Vss) of monoclonal antibodies (mAbs) from monkey data alone, PK data of 24 mAbs were collected and analyzed according to the types of targeted antigens (soluble or membrane-bound antigens). Based on the observed PK data in humans (at clinical doses) and monkeys (at >1 mg/kg), where the PK is expected to be linear, the mean scaling exponents in the allometric equation for CL and Vss, respectively, against body weight were calculated to be 0.79 and 1.12 [95% confidence intervals (CIs): 0.69-0.89 and 0.96-1.28] for soluble antigens, and 0.96 and 1.00 (95% CIs: 0.83-1.09 and 0.87-1.13) for membrane-bound antigens. Using these exponents and monkey PK data (at >1 mg/kg) alone, both human CL and Vss of mAbs can be predicted with reasonable accuracy, i.e., within 2-fold of the observed values. Compared with traditional allometric scaling using PK data from three or more preclinical species, this approach is simple, quick, resource-saving, and useful in drug discovery and development.


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais/farmacocinética , Adalimumab , Animais , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais Humanizados/sangue , Tamanho Corporal , Peso Corporal , Cetuximab , Bases de Dados Factuais , Relação Dose-Resposta a Droga , Descoberta de Drogas , Haplorrinos , Humanos , Modelos Animais , Especificidade da Espécie
20.
Drug Metab Dispos ; 39(5): 820-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21282406

RESUMO

The aim of the current study was to evaluate the accuracy of allometric scaling methods for drugs metabolized by UDP-glucuronosyltransferases (UGTs), such as ketoprofen, imipramine, lorazepam, levofloxacin, zidovudine, diclofenac, furosemide, raloxifene, gemfibrozil, mycophenolic acid, indomethacin, and telmisartan. Human plasma clearance (CL) predictions were conducted from preclinical in vivo data by using multiple-species allometry with the rule of exponents and single-species allometric scaling (SSS) of mice, rats, monkeys, or dogs. Distribution volume at a steady state (V(ss)) was predicted by multiple-species allometry or SSS of V(ss). Oral plasma clearance (CL(po)) was calculated under the assumption that F(a) × F(g) was equivalent across species. Each of the results was compared with the observed parameter calculated from the clinical data after intravenous or oral administration. Multiple-species allometry and SSS of mice, rats, and dogs resulted in a similar accuracy of CL and CL(po) predictions. Monkeys tended to provide the most accurate predictions of human CL and CL(po). The ability to predict the half-life, which was determined from CL and V(ss) predictions, was more accurate in SSS of rats and monkeys. The in vivo fraction metabolized by glucuronidation (f(m,UGT)) in bile duct-cannulated monkeys was relatively similar to that of humans compared with other animal species, which likely contributed to the highest accuracy of SSS prediction of monkeys. On the basis of the current results, monkeys would be more reliable than other animal species in predicting human pharmacokinetics and f(m,UGT) for drugs metabolized by UGTs.


Assuntos
Anti-Infecciosos/farmacocinética , Anti-Inflamatórios não Esteroides/farmacocinética , Anti-Hipertensivos/farmacocinética , Avaliação Pré-Clínica de Medicamentos/métodos , Glucuronosiltransferase/metabolismo , Administração Oral , Animais , Anti-Infecciosos/química , Anti-Infecciosos/metabolismo , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/metabolismo , Anti-Hipertensivos/química , Anti-Hipertensivos/metabolismo , Área Sob a Curva , Sistema Enzimático do Citocromo P-450/química , Sistema Enzimático do Citocromo P-450/metabolismo , Cães , Glucuronídeos/metabolismo , Glucuronosiltransferase/química , Meia-Vida , Humanos , Macaca fascicularis , Masculino , Camundongos , Preparações Farmacêuticas/administração & dosagem , Preparações Farmacêuticas/metabolismo , Valor Preditivo dos Testes , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...