Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Br J Clin Pharmacol ; 84(1): 179-188, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28865237

RESUMO

AIMS: LY3031207, a novel microsomal prostaglandin E synthase 1 inhibitor, was evaluated in a multiple ascending dose study after nonclinical toxicology studies and a single ascending dose study demonstrated an acceptable toxicity, safety and tolerability profile. METHODS: Healthy subjects were randomized to receive LY3031207 (25, 75 and 275 mg), placebo or celecoxib (400 mg) once daily for 28 days. The safety, tolerability and pharmacokinetic and pharmacodynamic profiles of LY3031207 were evaluated. RESULTS: The study was terminated when two subjects experienced drug-induced liver injury (DILI) after they had received 225 mg LY3031207 for 19 days. Liver biopsy from these subjects revealed acute liver injury with eosinophilic infiltration. Four additional DILI cases were identified after LY3031207 dosing had been stopped. All six DILI cases shared unique presentations of hepatocellular injury with hypersensitivity features and demonstrated a steep dose-dependent trend. Prompt discontinuation of the study drug and supportive medical care resulted in full recovery. Metabolites from metabolic activation of the imidazole ring were observed in plasma and urine samples from all subjects randomized to LY3031207 dosing. CONCLUSIONS: This study emphasized the importance of careful safety monitoring and serious adverse events management in phase I trials. Metabolic activation of the imidazole ring may be involved in the development of hepatotoxicity of LY3031207.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/etiologia , Hipersensibilidade a Drogas/etiologia , Inibidores Enzimáticos/efeitos adversos , Imidazóis/efeitos adversos , Prostaglandina-E Sintases/antagonistas & inibidores , Administração Oral , Adulto , Área Sob a Curva , Celecoxib/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/patologia , Inibidores de Ciclo-Oxigenase 2/efeitos adversos , Relação Dose-Resposta a Droga , Método Duplo-Cego , Esquema de Medicação , Hipersensibilidade a Drogas/patologia , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Feminino , Meia-Vida , Voluntários Saudáveis , Humanos , Imidazóis/administração & dosagem , Imidazóis/farmacocinética , Imidazóis/farmacologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/patologia , Masculino , Pessoa de Meia-Idade , Dor/tratamento farmacológico , Suspensão de Tratamento
2.
Pharm Res ; 33(12): 2920-2929, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27528391

RESUMO

PURPOSE: Determine the pharmacokinetics of insulin peglispro (BIL) in 5/6-nephrectomized rats and study the absorption in lymph duct cannulated (LDC) sheep. METHODS: BIL is insulin lispro modified with 20-kDa linear PEG at lysine B28 increasing the hydrodynamic size to 4-fold larger than insulin lispro. Pharmacokinetics of BIL and insulin lispro after IV administration were compared in 5/6-nephrectomized and sham rats. BIL was administered IV or SC into the interdigital space of the hind leg, and peripheral lymph and/or serum samples were collected from both LDC and non-LDC sheep to determine pharmacokinetics and absorption route of BIL. RESULTS: The clearance of BIL was similar in 5/6-nephrectomized and sham rats, while the clearance of insulin lispro was 3.3-fold slower in 5/6-nephrectomized rats than in the sham rats. In non-LDC sheep, the terminal half-life after SC was about twice as long vs IV suggesting flip-flop pharmacokinetics. In LDC sheep, bioavailability decreased to <2%; most of the dose was absorbed via the lymphatic system, with 88% ± 19% of the dose collected in the lymph after SC administration. CONCLUSION: This work demonstrates that increasing the hydrodynamic size of insulin lispro through PEGylation can impact both absorption and clearance to prolong drug action.


Assuntos
Hipoglicemiantes/química , Insulina Lispro/química , Linfa/efeitos dos fármacos , Polietilenoglicóis/química , Animais , Disponibilidade Biológica , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Meia-Vida , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/farmacocinética , Injeções Intravenosas , Injeções Subcutâneas , Insulina Lispro/administração & dosagem , Insulina Lispro/farmacocinética , Cinética , Masculino , Peso Molecular , Ratos Sprague-Dawley , Ovinos
3.
Drug Metab Dispos ; 40(12): 2354-64, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22961682

RESUMO

The disposition and metabolism of isopropyl N-[(2S)-7-cyano-4-(2-pyridylmethyl)-2,3-dihydro-1H-cyclopenta[b]indol-2-yl]carbamate (LY2452473; a selective androgen receptor modulator) in humans was characterized after a single 15-mg (100 µCi) oral dose of [¹4C]LY2452473 to six healthy male subjects. LY2452473 was absorbed rapidly (time to reach maximum plasma concentration for both LY2452473 and total radioactivity was 2-3 h) and cleared slowly (plasma terminal t(½) of 27 h for LY2452473 and 51 h for the total radioactivity). LY2452473 and metabolites S5 (acetylamine) and S12 (hydroxylation on the cyclopentene) were major circulating entities in plasma, accounting for approximately 42, 21, and 35% of the total radioactivity exposure, respectively, as calculated from relative area under the concentration versus time curves from zero to 48 h derived from the plasma radiochromatograms. The radioactive dose was almost completely recovered after 312 h with 47.9% of the dose eliminated in urine and 46.6% in feces. Minimal LY2452473 was detected in excreta, indicating that metabolic clearance was the main route of elimination. Multiple metabolic pathways were observed with no single metabolic pathway accounting for more than 30% of the dose in excreta. Metabolite S10 (a diol across the cyclopenta-indole linkage) was the largest excretory metabolite (approximately 14% of the dose). S10 displayed interesting chemical and chromatographic properties, undergoing conversion to the corresponding epoxide under acidic conditions and conversion back to the diol under neutral conditions. An in vitro phenotyping approach indicated that CYP3A4 was the largest contributor to LY2452473 depletion.


Assuntos
Carbamatos/farmacocinética , Receptores Androgênicos/metabolismo , Adulto , Área Sob a Curva , Citocromo P-450 CYP3A/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
4.
Xenobiotica ; 41(12): 1108-21, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21892880

RESUMO

LY377604 has a potential to form 4-hydroxycarbazole, which was reported in the literature as a mutagen. This safety concern led to our investigation of the metabolism and carcinogenicity of LY377604. In in vitro studies with LY377604, 4-hydroxycarbazole was detected in the presence of liver microsomes prepared from different species. When incubated with liver slices, only the conjugate of 4-hydroxycarbazole was detected. Subsequent in vivo radio-labelled studies were conducted to characterise the formation of 4-hydroxycarbazole from LY377604. Free 4-hydroxycarbazole was not detected in vivo, but the O-glucuronide conjugate was identified as a minor metabolite in urine samples, representing 0.2% and 0.9% of the radioactive dose in rats and monkeys. The low level of circulating 4-hydroxycarbazole glucuronide conjugate was also detected in plasma. LY377604 was negative in all genetic toxicology assays and was not associated with tumour induction in a 6-month carcinogenicity study using RasH2+/- mouse model. The exposure to free 4-hydroxycarbazole was not measurable after one dose and was about 0.1%-0.2% of the parent exposure at the end of the 6-month study. These data suggested that 4-hydroxycarbazole was formed as a minor metabolite in vivo, but it was primarily conjugated and excreted in urine as the glucuronide conjugate. The absence of tumours in the carcinogenicity study combined with the exposure data suggested that the low level of free 4-hydroxycarbazole did not represent a carcinogenic risk.


Assuntos
Carbazóis/metabolismo , Carbazóis/toxicidade , Carcinógenos/toxicidade , Receptores Adrenérgicos beta/metabolismo , Administração Oral , Animais , Carbazóis/administração & dosagem , Carbazóis/química , Testes de Carcinogenicidade , Cromatografia Líquida , Feminino , Fluorescência , Espectroscopia de Ressonância Magnética , Masculino , Espectrometria de Massas , Redes e Vias Metabólicas/efeitos dos fármacos , Mutagênicos/toxicidade , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Ratos Sprague-Dawley , Análise de Sobrevida
5.
J Med Chem ; 59(1): 194-205, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26653180

RESUMO

As part of a program aimed at the discovery of antinociceptive therapy for inflammatory conditions, a screening hit was found to inhibit microsomal prostaglandin E synthase-1 (mPGES-1) with an IC50 of 17.4 µM. Structural information was used to improve enzyme potency by over 1000-fold. Addition of an appropriate substituent alleviated time-dependent cytochrome P450 3A4 (CYP3A4) inhibition. Further structure-activity relationship (SAR) studies led to 8, which had desirable potency (IC50 = 12 nM in an ex vivo human whole blood (HWB) assay) and absorption, distribution, metabolism, and excretion (ADME) properties. Studies on the formulation of 8 identified 8·H3PO4 as suitable for clinical development. Omission of a lipophilic portion of the compound led to 26, a readily orally bioavailable inhibitor with potency in HWB comparable to celecoxib. Furthermore, 26 was selective for mPGES-1 inhibition versus other mechanisms in the prostanoid pathway. These factors led to the selection of 26 as a second clinical candidate.


Assuntos
Analgésicos/síntese química , Analgésicos/farmacologia , Inibidores de Ciclo-Oxigenase/síntese química , Inibidores de Ciclo-Oxigenase/farmacologia , Imidazóis/síntese química , Imidazóis/farmacologia , Oxirredutases Intramoleculares/antagonistas & inibidores , Microssomos/enzimologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Disponibilidade Biológica , Celecoxib/farmacologia , Inibidores de Ciclo-Oxigenase/farmacocinética , Citocromo P-450 CYP3A , Inibidores das Enzimas do Citocromo P-450/síntese química , Inibidores das Enzimas do Citocromo P-450/farmacologia , Cães , Descoberta de Drogas , Humanos , Microssomos/efeitos dos fármacos , Modelos Moleculares , Prostaglandina-E Sintases , Ratos , Relação Estrutura-Atividade
6.
Drug Metab Dispos ; 34(11): 1909-17, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16896067

RESUMO

Ruboxistaurin is a potent and specific inhibitor of the beta isoforms of protein kinase C (PKC) that is being developed for the treatment of diabetic microvascular complications. The disposition of [(14)C]ruboxistaurin was determined in six healthy male subjects who received a single oral dose of 64 mg of [(14)C]ruboxistaurin in solution. There were no clinically significant adverse events during the study. Whole blood, urine, and feces were collected at frequent intervals after dosing. Metabolites were profiled by high performance liquid chromatography with radiometric detection. The total mean recovery of the radioactive dose was approximately 87%, with the majority of the radioactivity (82.6 +/- 1.1%) recovered in the feces. Urine was a minor pathway of elimination (4.1 +/- 0.3%). The major route of ruboxistaurin metabolism was to the N-desmethyl ruboxistaurin metabolite (LY338522), which has been shown to be active and equipotent to ruboxistaurin in the inhibition of PKC(beta). In addition, multiple hydroxylated metabolites were identified by liquid chromatography-mass spectrometry in all matrices. Pharmacokinetics were conducted for both ruboxistaurin and LY338522 (N-desmethyl ruboxistaurin, 1). These moieties together accounted for approximately 52% of the radiocarbon measured in the plasma. The excreted radioactivity was profiled using radiochromatography, and approximately 31% was structurally characterized as ruboxistaurin or N-desmethyl ruboxistaurin. These data demonstrate that ruboxistaurin is metabolized primarily to N-desmethyl ruboxistaurin (1) and multiple other oxidation products, and is excreted primarily in the feces.


Assuntos
Inibidores Enzimáticos/farmacocinética , Indóis/farmacocinética , Maleimidas/farmacocinética , Proteína Quinase C/antagonistas & inibidores , Administração Oral , Adulto , Radioisótopos de Carbono , Cromatografia Líquida , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/urina , Fezes/química , Humanos , Indóis/sangue , Indóis/metabolismo , Indóis/urina , Masculino , Maleimidas/sangue , Maleimidas/metabolismo , Maleimidas/urina , Pessoa de Meia-Idade , Estrutura Molecular , Proteína Quinase C beta , Espectrometria de Massas em Tandem , Distribuição Tecidual
7.
Drug Metab Dispos ; 34(2): 213-24, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16258078

RESUMO

Ruboxistaurin (LY333531), a potent and isoform-selective protein kinase C beta inhibitor, is currently undergoing clinical trials as a therapeutic agent for the treatment of diabetic microvascular complications. The present study describes the disposition and metabolism of [14C]ruboxistaurin following administration of an oral dose to dogs, mice, and rats. The study revealed that ruboxistaurin was highly metabolized in all species. Furthermore, the results from the bile duct-cannulated study revealed that ruboxistaurin was well absorbed in rats. The primary route of excretion of ruboxistaurin and its metabolites was through feces in all species. The major metabolite detected consistently in all matrices for all species was the N-desmethyl metabolite 1, with the exception of rat bile, in which hydroxy N-desmethyl metabolite 5 was detected as the major metabolite. Other significant metabolites detected in dog plasma were 2, 3, 5, and 6 and in mouse plasma 2, 5, and 19. The structures of the metabolites were proposed by tandem mass spectrometry with the exception of 1, 2, 3, 5, and 6, which were additionally confirmed either by direct comparison with authentic standards or by nuclear magnetic resonance spectroscopy. To assist identification by nuclear magnetic resonance spectroscopy, metabolites 3 and 5 were produced via biotransformation using recombinant human CYP2D6 and, likewise, metabolite 6 and compound 4 (regioisomer of 3 which did not correlate to metabolites found in vivo) were produced using a microbe, Mortierella zonata. The unambiguous identification of metabolites enabled the proposal of clear metabolic pathways of ruboxistaurin in dogs, mice, and rats.


Assuntos
Inibidores Enzimáticos/farmacocinética , Indóis/farmacocinética , Maleimidas/farmacocinética , Animais , Bile/química , Biotransformação , Radioisótopos de Carbono , Cromatografia Líquida , Citocromo P-450 CYP2D6/metabolismo , Cães , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/urina , Fezes/química , Feminino , Indóis/sangue , Indóis/urina , Espectroscopia de Ressonância Magnética , Masculino , Maleimidas/sangue , Maleimidas/urina , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos , Mortierella/metabolismo , Proteína Quinase C/antagonistas & inibidores , Ratos , Ratos Endogâmicos F344
8.
Antimicrob Agents Chemother ; 47(5): 1700-6, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12709343

RESUMO

The pharmacokinetics and pharmacodynamics of oritavancin (LY333328), a glycopeptide antibiotic with concentration-dependent bactericidal activity against gram-positive pathogens, in a neutropenic-mouse thigh model of Staphylococcus aureus infection were studied. Plasma radioequivalent concentrations of oritavancin were determined by using [(14)C]oritavancin at doses ranging from 0.5 to 20 mg/kg of body weight. Peak plasma radioequivalent concentrations after an intravenous dose were 7.27, 12.56, 69.29, and 228.83 micro g/ml for doses of 0.5, 1, 5, and 20 mg/kg, respectively. The maximum concentration of drug in serum (C(max)) and the area under the concentration-time curve (AUC) increased linearly in proportion to the dose. Neither infection nor neutropenia was seen to affect the pharmacokinetics of oritavancin. Intravenous administration resulted in much higher concentrations in plasma than the concentrations obtained with subcutaneous administration. Single-dose dose-ranging studies suggested a sigmoid maximum effect (E(max)) dose-response relationship, with a maximal effect evident at single doses exceeding 2 mg/kg. The oritavancin dose (stasis dose) that resulted in a 24-h colony count similar to the pretreatment count was 1.53 (standard error [SE], 0.35) mg/kg. The single oritavancin dose that resulted in 50% of maximal bacterial killing (ED(50)) was 0.95 (SE, 0.20) mg/kg. Dose fractionation studies suggested that single doses of 0.5, 1, 2, 4, and 16 mg/kg appeared to have greater bactericidal efficacy than the same total dose subdivided and administered multiple times during the 24-h treatment period. When using an inhibitory E(max) model, C(max) appears to correlate better with bactericidal activity than do the time during which the concentration in plasma exceeds the MIC (T>MIC) and AUC. These data suggest that optimal oritavancin dosing strategies will require regimens that favor high C(max) concentrations rather than long periods during which unbound concentrations in plasma exceed the MIC.


Assuntos
Antibacterianos/farmacocinética , Glicopeptídeos , Neutropenia/complicações , Infecções Estafilocócicas/tratamento farmacológico , Animais , Antibacterianos/administração & dosagem , Antibacterianos/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Lipoglicopeptídeos , Camundongos , Camundongos Endogâmicos ICR , Modelos Biológicos , Infecções Estafilocócicas/metabolismo , Coxa da Perna
9.
Drug Metab Dispos ; 30(9): 957-61, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12167559

RESUMO

Studies were performed to determine the cytochromes P450 (P450) responsible for the biotransformation of (S)-13[(dimethylamino)methyl]-10,11,14,15-tetrahydro-4,9:16,21-dimetheno-1H, 13H-dibenzo[e,k]pyrrolo[3,4-h][1,4,13]oxadiazacyclohexadecene-1,3(2H)-dione (LY333531) to its equipotent metabolite, N-desmethyl LY333531, and to examine the ability of these two compounds to inhibit P450-mediated metabolism. Kinetic studies indicated that a single enzyme in human liver microsomes was able to form N-desmethyl LY333531 with an apparent K(M) value of approximately 1 microM. The formation rate of N-desmethyl LY333531 was correlated with markers of nine P450s in a bank of 20 human liver microsomes. The only significant correlation observed was with the form-selective activity for CYP3A. Of the nine cDNA-expressed P450s examined, only CYP3A4 and CYP2D6 formed N-desmethyl LY333531. However, CYP3A4 formed N-desmethyl LY333531 at a rate 57-fold greater than that observed with CYP2D6. In incubations with human liver microsomes, quinidine, an inhibitor of CYP2D6, demonstrated little inhibition of metabolite formation while ketoconazole, an inhibitor of CYP3A, demonstrated almost complete inhibition. Thus, CYP3A is responsible for the formation of N-desmethyl LY333531. LY333531 and N-desmethyl LY333531 were also examined for their ability to inhibit metabolism mediated by CYP2D6, CYP2C9, CYP3A, and CYP1A2. LY333531 and N-desmethyl LY333531 were found to competitively inhibit CYP2D6 with calculated K(i) values of 0.17 and 1.0 microM, respectively. Less potent inhibition by these compounds of metabolism mediated by the other three P450s examined was observed. In conclusion, CYP3A is primarily responsible for forming N-desmethyl LY333531. Therefore, alterations in the activity of this enzyme have the potential to affect LY333531 clearance. In addition, LY333531 and its metabolite are predicted to be potential inhibitors of CYP2D6-mediated reactions in vivo.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Isoenzimas/antagonistas & inibidores , Maleimidas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Hidrocarboneto de Aril Hidroxilases/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP3A , Inibidores Enzimáticos/metabolismo , Humanos , Técnicas In Vitro , Indóis/metabolismo , Maleimidas/metabolismo , Microssomos Hepáticos/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Proteína Quinase C beta
10.
Drug Metab Dispos ; 30(11): 1266-73, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12386134

RESUMO

Previous results demonstrating homotropic activation of human UDP-glucuronosyltransferase (UGT) 1A1-catalyzed estradiol-3-glucuronidation led us to investigate the effects of 16 compounds on estradiol glucuronidation by human liver microsomes (HLM). In confirmation of previous work using alamethicin-treated HLM pooled from four livers, UGT1A1-catalyzed estradiol-3-glucuronidation demonstrated homotropic activation kinetics (S(50) = 22 microM, Hill coefficient, n = 1.9) whereas estradiol-17-glucuronidation (catalyzed by other UGT enzymes) followed Michaelis-Menten kinetics (K(m) = 7 microM). Modulatory effects of the following compounds were investigated: bilirubin, eight flavonoids, 17alpha-ethynylestradiol (17alpha-EE), estriol, 2-amino-1-methyl-6-phenylimidazo [4,5-b]pyridine (PhIP), anthraflavic acid, retinoic acid, morphine, and ibuprofen. Although the classic UGT1A1 substrate bilirubin was a weak competitive inhibitor of estradiol-3-glucuronidation, the estrogens and anthraflavic acid activated or inhibited estradiol-3-glucuronidation dependent on substrate and effector concentrations. For example, at substrate concentrations of 5 and 10 microM, estradiol-3-glucuronidation activity was stimulated by as much as 80% by low concentrations of 17alpha-EE but was unaltered by flavanone. However, at higher substrate concentrations (25-100 microM) estradiol-3-glucuronidation was inhibited by about 55% by both compounds. Anthraflavic acid and PhIP were also stimulators of estradiol 3-glucuronidation at low substrate concentrations. The most potent inhibitor of estradiol 3-glucuronidation was the flavonoid tangeretin. The UGT2B7 substrates morphine and ibuprofen had no effect on estradiol 3-glucuronidation, whereas retinoic acid was slightly inhibitory. Estradiol-17-glucuronidation was inhibited by 17alpha-EE, estriol, and naringenin but was not activated by any compound. This study demonstrates that the interactions of substrates and inhibitors at the active site of UGT1A1 are complex, yielding both activation and competitive inhibition kinetics.


Assuntos
Estradiol/metabolismo , Glucuronosiltransferase/metabolismo , Microssomos Hepáticos/enzimologia , Algoritmos , Bilirrubina/farmacologia , Estrogênios/farmacologia , Flavonoides/farmacologia , Glucuronídeos/metabolismo , Humanos , Técnicas In Vitro , Cinética , Relação Estrutura-Atividade , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA