Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Xenobiotica ; 48(5): 467-477, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28485193

RESUMO

1. Breast cancer resistance protein (BCRP) plays an important role in drug absorption, distribution and excretion. It is challenging to evaluate BCRP functions in preclinical models because commonly used BCRP inhibitors are nonspecific or unstable in animal plasma. 2. In this work, in vitro absorption, distribution, metabolism and elimination (ADME) assays and pharmacokinetic (PK) experiments in Bcrp knockout (KO) (Abcg2-/-) and wild-type (WT) FVB mice and Wistar rats were conducted to characterize the preclinical properties of a novel selective BCRP inhibitor (ML753286, a Ko143 analog). 3. ML753286 is a potent inhibitor for BCRP, but not for P-glycoprotein (P-gp), organic anion-transporting polypeptide (OATP) or major cytochrome P450s (CYPs). It has high permeability, but is not an efflux transporter substrate. ML753286 has low to medium clearance in rodent and human liver S9 fractions, and is stable in plasma cross species. Bcrp inhibition affects oral absorption and clearance of sulfasalazine in rodents. A single dose of ML753286 at 50-300 mg/kg orally, and at 20 mg/kg intravenously or 25 mg/kg orally inhibits Bcrp functions in mice and rats, respectively. 4. These findings confirm that ML753286 is a useful selective inhibitor to evaluate BCRP/Bcrp activity in vitro and in rodent model systems.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Absorção Fisiológica , Neoplasias da Mama/tratamento farmacológico , Dicetopiperazinas/farmacocinética , Dicetopiperazinas/uso terapêutico , Proteínas de Neoplasias/antagonistas & inibidores , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Células CACO-2 , Permeabilidade da Membrana Celular/efeitos dos fármacos , Dicetopiperazinas/sangue , Dicetopiperazinas/química , Cães , Feminino , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Macaca fascicularis , Masculino , Camundongos Knockout , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Proteínas de Neoplasias/metabolismo , Ratos , Sulfassalazina/farmacologia , Sulfassalazina/uso terapêutico , Fatores de Tempo
2.
Bioorg Med Chem Lett ; 26(2): 551-555, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26642765

RESUMO

The design, synthesis, in vitro inhibitory potency, and pharmacokinetic (PK) profiles of Ko143 analogs are described. Compared to commonly used Ko143, the new breast cancer resistance protein (BCRP) inhibitor (compound A) showed the same potency and a significantly improved PK profile in rats (lower clearance [1.54L/h/kg] and higher bioavailability [123%]). Ko143 on the other hand suffers from poor bioavailability. Compared to Ko143, compound A would be a useful probe for delineating the role of BCRP during in vivo studies in animals.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Dicetopiperazinas/síntese química , Dicetopiperazinas/farmacocinética , Compostos Heterocíclicos de 4 ou mais Anéis/síntese química , Compostos Heterocíclicos de 4 ou mais Anéis/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Animais , Células CACO-2 , Estrona/análogos & derivados , Estrona/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/sangue , Humanos , Ratos , Estereoisomerismo , Relação Estrutura-Atividade
3.
Biopharm Drug Dispos ; 35(9): 543-52, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25264242

RESUMO

Orteronel is a nonsteroidal, selective inhibitor of 17,20-lyase that was recently in phase 3 clinical development as a treatment for castration-resistant prostate cancer. In humans, the primary clearance route for orteronel is renal excretion. Human liver microsomal studies indicated that orteronel weakly inhibits CYP1A2, 2C8, 2C9 and 2C19, with IC50 values of 17.8, 27.7, 30.8 and 38.8 µm, respectively, whereas orteronel does not inhibit CYP2B6, 2D6 or 3A4/5 (IC50 > 100 µm). Orteronel also does not exhibit time-dependent inhibition of CYP1A2, 2B6, 2C8, 2C9, 2C19, 2D6 or 3A4/5. The results of a static model indicated an [I]/Ki ratio >0.1 for CYP1A2, 2C8, 2C9 and 2C19. Therefore, a physiologically based pharmacokinetic (PBPK) model was developed to assess the potential for drug-drug interactions (DDIs) between orteronel and theophylline, repaglinide, (S)-warfarin and omeprazole, which are sensitive substrates of CYP1A2, 2C8, 2C9 and 2C19, respectively. Simulation of the area under the plasma concentration-time curve (AUC) of these four CYP substrates in the presence and absence of orteronel revealed geometric mean AUC ratios <1.25. Therefore, in accordance with the 2012 US FDA Draft Guidance on DDIs, orteronel can be labeled a 'non-inhibitor' and further clinical DDI evaluation is not required. In PBPK models of moderate and severe renal impairment, the AUC of orteronel was predicted to increase by 52% and 83%, respectively. These results are in agreement with those of a clinical trial in which AUC increases of 38% and 87% were observed in patients with moderate and severe renal impairment, respectively.


Assuntos
Antineoplásicos/farmacocinética , Simulação por Computador , Sistema Enzimático do Citocromo P-450/metabolismo , Imidazóis/farmacocinética , Modelos Biológicos , Naftalenos/farmacocinética , Insuficiência Renal/metabolismo , Absorção Fisiológica , Idoso , Antineoplásicos/sangue , Antineoplásicos/química , Interações Medicamentosas , Humanos , Imidazóis/sangue , Imidazóis/química , Taxa de Depuração Metabólica , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Pessoa de Meia-Idade , Estrutura Molecular , Peso Molecular , Naftalenos/sangue , Naftalenos/química , Preparações Farmacêuticas/administração & dosagem , Preparações Farmacêuticas/metabolismo , Valor Preditivo dos Testes , Insuficiência Renal/enzimologia , Especificidade por Substrato
4.
J Labelled Comp Radiopharm ; 57(9): 574-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24992204

RESUMO

[(13) CD3 ]-TAK-459 (1A), an HSP90 inhibitor, was synthesized from [(13) CD3 ]-sodium methoxide in three steps in an overall yield of 29%. The key intermediate [(13) CD3 ]-2-methoxy-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine was synthesized in two steps from 2,6-dibromopyridine and stable isotope-labeled sodium methoxide. [(14) C]-TAK-459 (1B) was synthesized from [(14) C(U)]-guanidine hydrochloride in five steps in an overall radiochemical yield of 5.4%. The key intermediate, [(14) C]-(R)-2-amino-7-(2-bromo-4-fluorophenyl)-4-methyl-7,8-dihydropyrido[4,3-d]pyrimidin-5(6H)-one, was prepared by microwave-assisted condensation.


Assuntos
Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Piridinas/síntese química , Pirimidinas/síntese química , Compostos Radiofarmacêuticos/síntese química , Radioisótopos de Carbono/química , Piridinas/química , Pirimidinas/química
5.
Drug Metab Dispos ; 41(4): 727-34, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23298863

RESUMO

6-Ethyl-N-[1-(hydroxyacetyl)piperidin-4-yl]-1-methyl-4-oxo-5-(2-oxo-2-phenylethyl)-3-(2,2,2-trifluoroethoxy)-4,5-dihydro-1H-pyrrolo[3,2-c]pyridine-2-carboxamide (TAK-441) is a potent, selective hedgehog signaling pathway inhibitor that binds to Smo and is being developed for the treatment of cancer. The objectives of these studies were to explore the possibility of establishing of a link between the pharmacokinetics of TAK-441 and the responses of Gli1 mRNA in tumor-associated stromal or skin cells and the antitumor effect of hedgehog inhibition. To this end, we built pharmacokinetic and pharmacodynamic models that describe the relationship of the concentrations of TAK-441 plasma to the responses of Gli1 mRNA in the tumor (target) and skin (surrogate) and to tumor growth inhibition in mice bearing xenografts of human pancreatic tumors (PAN-04). The responses of Gli1 mRNA and tumor growth were described by an indirect response model and an exponential tumor growth model, respectively. The IC50 values for Gli1 mRNA inhibition in the tumor and skin by TAK-441 were estimated to be 0.0457 and 0.113 µg/ml, respectively. The IC90 value for tumor growth inhibition was estimated to be 0.68 µg/ml. These results suggest that a >83% inhibition of Gli1 mRNA expression in the skin or a >94% inhibition of Gli1 mRNA expression in the tumor would be required to sufficiently inhibit (>90%) hedgehog-related tumor growth in the xenografted model mice. We conclude that Gli1 mRNA expression in the tumor and skin could be a useful biomarker for predicting the antitumor effect of hedgehog inhibitors.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/farmacocinética , Expressão Gênica/efeitos dos fármacos , Proteínas Hedgehog/antagonistas & inibidores , Proteínas Oncogênicas/genética , Piridinas/farmacologia , Piridinas/farmacocinética , Pirróis/farmacologia , Pirróis/farmacocinética , Transativadores/genética , Animais , Relação Dose-Resposta a Droga , Feminino , Humanos , Camundongos , Modelos Biológicos , Neoplasias/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Pele/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína GLI1 em Dedos de Zinco
6.
J Labelled Comp Radiopharm ; 56(9-10): 475-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24285524

RESUMO

L-MTP-PE (1), an immunomodulator and its metabolite MDP (4) were synthesized from labeled l-alanine and its protected derivative, respectively. The key intermediate product for the labeled L-MTP-PE synthesis, [(13) C3 ,D4 ]-alanyl-cephalin (2A), was synthesized from [(13) C3 ,D4 ]-l-alanine (3A) in three steps. The key intermediate product for labeled MDP synthesis, amine 11, was prepared from [(13) C3 ,(15) N]-Boc-l-alanine (5A) in two steps.


Assuntos
Acetilmuramil-Alanil-Isoglutamina/análogos & derivados , Acetilmuramil-Alanil-Isoglutamina/química , Acetilmuramil-Alanil-Isoglutamina/síntese química , Fosfatidiletanolaminas/química , Fosfatidiletanolaminas/síntese química , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Técnicas de Química Sintética , Marcação por Isótopo , Fosfatidiletanolaminas/metabolismo
7.
J Labelled Comp Radiopharm ; 56(9-10): 464-70, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24285522

RESUMO

MLN9708 (ixazomib citrate) is an investigational, orally bioavailable proteasome inhibitor that is under development by Millennium in clinical studies in both hematologic and nonhematologic malignancies. The stable isotope-labeled MLN9708 was required for bio-analytical studies. [(13) C9 ]-MLN9708 (11) was synthesized in seven steps from the uniformly labeled [(13) C6 ]-1,4-dichlorobenzene (3) and [1-(13) C]-acetyl chloride. Because of the presence of two chlorine atoms and a boron atom, compound 6 was further reacted with [(13) C2 ]-glycine to provide an internal standard that is well separated from the parent compound during mass spectrometric analysis. The radiolabeled version was prepared to support metabolite profiling and whole body autoradiography studies in experimental animals. [(14) C]-MLN9708 (19) was synthesized in six steps from commercially available [(14) C]-barium carbonate. The key intermediate, [carboxyl-(14) C]-2,5-dichlorobenzoic acid (14), was prepared by selective lithiation of 1-bromo-2,5-dichlorobenzene (12) followed by carbonation with [(14) C]-barium carbonate. In preparation for a one-time human absorption, distribution, metabolism and excretion (ADME) study, the stability of [(14) C]-MLN9708 and its precursors were also evaluated.


Assuntos
Compostos de Boro/química , Compostos de Boro/síntese química , Glicina/análogos & derivados , Inibidores de Proteassoma/química , Inibidores de Proteassoma/síntese química , Isótopos de Carbono/química , Radioisótopos de Carbono/química , Técnicas de Química Sintética , Glicina/síntese química , Glicina/química , Marcação por Isótopo
8.
J Pharmacol Exp Ther ; 332(2): 562-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19889796

RESUMO

A novel in vitro model was recently developed in our laboratories for the prediction of magnitude of clinical pharmacokinetic drug-drug interactions (DDIs), based on reversible hepatic cytochrome P450 (P450) inhibition. This approach, using inhibition data from human hepatocytes incubated in human plasma, and quantitative P450 phenotyping data from hepatic microsomal incubations, successfully predicted DDIs for 15 marketed drugs with ketoconazole, a strong competitive inhibitor of CYP3A4/5, generally used to demonstrate a "worst-case scenario" for CYP3A inhibition. In addition, this approach was successfully extended to DDI predictions with the moderate competitive CYP3A inhibitor fluconazole for nine marketed drugs. In the current report, the general applicability of the model has been demonstrated by prospectively predicting the degree of inhibition and then conducting DDI studies in the clinic for an investigational CCR1 antagonist MLN3897, which is cleared predominantly by CYP3A. The clinical studies involved treatment of healthy volunteers (n = 17-20), in a crossover design, with ketoconazole (200 mg b.i.d.) or fluconazole (400 mg once a day), while receiving MLN3897. Administration of MLN3897 and ketoconazole led to an average 8.28-fold increase in area under the curve of plasma concentration-time plot (AUC) of MLN3897 at steady state, compared with the 8.33-fold increase predicted from the in vitro data. Similarly for fluconazole, an average increase of 3.93-fold in AUC was observed for MLN3897 in comparison with a predicted value of 3.26-fold. Thus, our model reliably predicted the exposure changes for MLN3897 in interaction studies with competitive CYP3A inhibitors in humans, further strengthening the utility of our in vitro model.


Assuntos
Inibidores das Enzimas do Citocromo P-450 , Interações Medicamentosas , Inibidores Enzimáticos/administração & dosagem , Hepatócitos/enzimologia , Microssomos Hepáticos/enzimologia , Receptores CCR1/antagonistas & inibidores , Adulto , Estudos Cross-Over , Inibidores do Citocromo P-450 CYP3A , Feminino , Fluconazol/administração & dosagem , Humanos , Cetoconazol/administração & dosagem , Masculino , Modelos Biológicos
9.
Drug Metab Dispos ; 36(7): 1261-6, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18381488

RESUMO

Whereas ketoconazole is often used to study the worst-case scenario for clinical pharmacokinetic drug-drug interactions (DDIs) for drugs that are primarily metabolized by CYP3A4, fluconazole is considered to be a moderate inhibitor of CYP3A4, providing assessment of the moderate-case scenario of CYP3A-based DDIs. Fluconazole is also a moderate inhibitor of CYP2C9 and CYP2C19. For predicting clinical DDIs using conventional approaches, determining the in vivo inhibitor concentration at the enzymatic site [I], a critical parameter, is still not practical. In our previous study, a novel method involving hepatocyte suspension in plasma was used to circumvent the need to determine the elusive [I] value. In this study, the CYP1A2, 2C9, 2C19, 2D6, and 3A4 activities remaining in the presence of fluconazole were determined in human hepatocytes suspended in human plasma, covering a range of fluconazole clinical plasma concentrations (C(avg) and C(max)). Because the protein-binding effect of fluconazole is expected to be close to that in vivo, the inhibition observed in vitro will be similar to that in vivo. This inhibition information was then applied to the cytochrome P450 (P450) phenotypic data to predict DDIs. Using the available P450 phenotypic information on theophylline, tolbutamide, omeprazole, S-warfarin, phenytoin, cyclosporine, and midazolam and that determined in this study for sirolimus and tacrolimus, we found that the predictions for area under the curve increases for most of these drugs in the presence of fluconazole were remarkably similar (within 35%) to the observed clinical values. This study proves the general applicability of our approach using human hepatocyte incubation in human plasma to predict DDIs.


Assuntos
Antifúngicos/farmacocinética , Fluconazol/farmacocinética , Hepatócitos/efeitos dos fármacos , Antifúngicos/sangue , Área Sob a Curva , Interações Medicamentosas , Fluconazol/sangue , Hepatócitos/enzimologia , Humanos
10.
Drug Metab Dispos ; 36(6): 1102-10, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18347085

RESUMO

The role of the intestine in the elimination of (2R)-2-{(3R)-3-amino-3-[4-(2-methylquinolin-4-ylmethoxy)phenyl]-2-oxopyrrolidin-1-yl}-N-hydroxy-4-methylpentanamide (DPC 333), a potent inhibitor of tissue necrosis factor alpha-converting enzyme, was investigated in mice and rats in vivo and in vitro. In Madine-Darby canine kidney cells stably transfected with P-glycoprotein (P-gp) and DPC 333, the transport from B-->A reservoirs exceeded the transport from A-->B by approximately 7-fold. In Caco-2 monolayers and isolated rat ileal mucosa, DPC 333 was transported from basolateral to apical reservoirs in a concentration-dependent, saturable manner, and transport was blocked by 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), confirming the contribution of P-gp/breast cancer resistance protein in B-->A efflux of DPC 333. In quantitative whole body autoradiography studies with [(14)C]DPC 333 in mice and rats, radioactivity was distributed throughout the small intestine in both species. In GF120918-pretreated bile duct-cannulated rats, radioactivity in feces was reduced 60%. Using the in situ perfused rat intestine model, approximately 20% of an i.v. dose of [(14)C]DPC 333 was measured in the intestinal lumen within 3 h postdose, 12% as parent. Kinetic analysis of data suggested that excreted DPC 333 may be further metabolized in the gut. Intestinal clearance was 0.2 to 0.35 l/h/kg. The above data suggest that in the rodent the intestine serves as an organ of DPC 333 excretion, mediated in part by the transporter P-gp.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Mucosa Intestinal/metabolismo , Quinolinas/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Células CACO-2 , Linhagem Celular , Cães , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Quinolinas/sangue , Ratos , Ratos Sprague-Dawley , Transfecção
11.
Clin Pharmacol Drug Dev ; 5(3): 180-7, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27163496

RESUMO

This study evaluated the absorption, distribution, and excretion of orteronel, an investigational, nonsteroidal, reversible, selective 17,20-lyase inhibitor. Six healthy male subjects received a single 400-mg dose of radiolabeled [(14) C]-orteronel (18.5 kBq). The pharmacokinetics of [(14) C]-radioactivity, orteronel, and the primary metabolite M-I were characterized by ultra-performance liquid chromatography-tandem mass spectrometry, and mass balance recovery of [(14) C]-radioactivity was determined by liquid scintillation counting and accelerator mass spectrometry. Median time to maximum observed concentration of [(14) C]-radioactivity was 2.5 hours (plasma/whole blood) and of orteronel was 1 hour (plasma). Mean terminal half-life for [(14) C]-radioactivity in plasma and whole blood was 9.46 and 7.39 hours, respectively. For [(14) C]-radioactivity, the geometric mean whole blood-to-plasma ratios for maximum observed plasma/whole-blood concentration, area under the plasma concentration-time curve from time 0 to last quantifiable concentration (AUC0-last ), and AUC0-inf (AUC from time 0 to infinity) were 1.04, 0.92, and 0.93, respectively. Dose recovery accounted for 95.9% of the administered orteronel dose; the majority of excretion occurred by 96 hours postdose. The principal excretion route was via urine (mean, 77.5%; including 49.7% unchanged drug and 16.3% M-I) compared with 18.4% via feces. Three mild adverse events were reported; none were considered serious or related to orteronel.


Assuntos
Inibidores das Enzimas do Citocromo P-450/farmacocinética , Imidazóis/farmacocinética , Naftalenos/farmacocinética , Adulto , Área Sob a Curva , Cromatografia Líquida , Inibidores das Enzimas do Citocromo P-450/efeitos adversos , Meia-Vida , Humanos , Imidazóis/efeitos adversos , Masculino , Naftalenos/efeitos adversos , Esteroide 17-alfa-Hidroxilase/antagonistas & inibidores , Espectrometria de Massas em Tandem , Adulto Jovem
12.
Drug Metab Lett ; 10(1): 22-37, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26031460

RESUMO

UNLABELLED: MLN3897 is a small molecule antagonist of the C-C chemokine receptor-1. Since preclinical studies showed that the molecule was metabolized into two halves, the metabolism, excretion, and pharmacokinetics of MLN3897 were investigated in humans using MLN3897 14C-radiolabeled either on the chlorophenyl (CP) or the tricyclic (TC) half of MLN3897 after an oral dose. OBJECTIVE: To evaluate the mass balance, metabolism and pharmacokinetics of MLN3897 in two cohorts of six randomized healthy subjects. METHOD: After receiving informed consent, subjects were dosed after an overnight fast of 10-hours followed by at least 4- hours after dosing on day-1. Each cohort received a single 29 mg oral dose of either the CP or the TC as an oral solution in water. Serial blood samples, urine and feces were collected over a 10-day period post-dose. RESULTS: For both radiolabeled moieties, 55-59% of the dose was recovered in feces and 32% recovered in urine. MLN3897 was metabolized extensively in humans, with minor amounts of intact MLN3897 detected in the urine and feces. N-oxidation of the tricyclic moiety (M28) and N-dealkylation of the piperidinyl moiety were the primary metabolic pathways leading to further formation of the carboxylic acid metabolite (M19) and the (4-(4-chlorophenyl)-3,3- dimethylpiperidin-4-ol) metabolite (M40). Oxidative metabolites M11, M19, M28, M44 were present at >10% of the total circulating radioactivity and also at >25% of MLN3897 exposure. Metabolites resulting from the chlorophenyl-labeled moiety (M40) had significantly more systemic exposure compared to the tricyclic-labeled moiety (M19).


Assuntos
Anti-Inflamatórios/farmacocinética , Receptores CCR1/antagonistas & inibidores , Administração Oral , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/sangue , Anti-Inflamatórios/urina , Benzoxepinas/administração & dosagem , Benzoxepinas/sangue , Benzoxepinas/farmacocinética , Benzoxepinas/urina , Biotransformação , Ácidos Carboxílicos/metabolismo , Remoção de Radical Alquila , Fezes/química , Feminino , Humanos , Eliminação Intestinal , Espectroscopia de Ressonância Magnética , Masculino , Oxirredução , Piridinas/administração & dosagem , Piridinas/sangue , Piridinas/farmacocinética , Piridinas/urina , Ratos Sprague-Dawley , Receptores CCR1/metabolismo , Eliminação Renal
13.
J Neurosci ; 24(12): 2942-52, 2004 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-15044533

RESUMO

gamma-Secretase is a multimeric complex consisted of presenilins (PSs) and three other proteins. PSs appear to be key contributors for the enzymatic center, the potential target of a number of recently developed gamma-secretase inhibitors. Using radiolabeled and unlabeled inhibitors as ligands, this study was aimed to determine the in situ distribution of gamma-secretase in the brain. Characterization using PS-1 knock-out mouse embryos revealed 50 and 80% reductions of gamma-secretase inhibitor binding density in the heterozygous (PS-1(+/-)) and homozygous (PS-1-/-) embryos, respectively, relative to the wild type (PS-1(+/+)). The pharmacological profile from competition binding assays suggests that the ligands may target at the N- and C-terminal fragments of PS essential for gamma-secretase activity. In the adult rat brain, the binding sites existed mostly in the forebrain, the cerebellum, and discrete brainstem areas and were particularly abundant in areas rich in neuronal terminals, e.g., olfactory glomeruli, CA3-hilus area, cerebellar molecular layer, and pars reticulata of the substantia nigra. In the developing rat brain, diffuse and elevated expression of binding sites occurred at the early postnatal stage relative to the adult. The possible association of binding sites with neuronal terminals in the adult brain was further investigated after olfactory deafferentation. A significant decrease with subsequent recovery of binding sites was noted in the olfactory glomeruli after chemical damage of the olfactory epithelium. The findings in this study support a physiological role of PS or gamma-secretase complex in neuronal and synaptic development and plasticity.


Assuntos
Encéfalo/metabolismo , Endopeptidases/efeitos dos fármacos , Endopeptidases/metabolismo , Inibidores Enzimáticos/metabolismo , Vias Aferentes/efeitos dos fármacos , Vias Aferentes/fisiologia , Fatores Etários , Secretases da Proteína Precursora do Amiloide , Animais , Ácido Aspártico Endopeptidases , Autorradiografia , Sítios de Ligação/fisiologia , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Denervação , Inibidores Enzimáticos/farmacocinética , Ligantes , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Bulbo Olfatório/efeitos dos fármacos , Bulbo Olfatório/fisiologia , Presenilina-1 , Ratos , Ratos Sprague-Dawley , Sulfato de Zinco/farmacologia
14.
Drug Metab Lett ; 4(4): 201-12, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20670210

RESUMO

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.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas de Neoplasias/metabolismo , Piperazinas/farmacocinética , Inibidores de Proteínas Quinases/farmacocinética , Quinazolinas/farmacocinética , Subfamília B de Transportador de Cassetes de Ligação de ATP/antagonistas & inibidores , Subfamília B de Transportador de Cassetes de Ligação de ATP/deficiência , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Transportadores de Cassetes de Ligação de ATP/genética , Acridinas/farmacologia , Administração Oral , Animais , Transporte Biológico , Encéfalo/metabolismo , Células CACO-2 , Humanos , Injeções Intravenosas , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/antagonistas & inibidores , Piperazinas/administração & dosagem , Piperazinas/sangue , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/sangue , Quinazolinas/administração & dosagem , Quinazolinas/sangue , Ratos , Ratos Sprague-Dawley , Tetra-Hidroisoquinolinas/farmacologia , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
15.
Drug Metab Dispos ; 35(1): 79-85, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17020957

RESUMO

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.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Interações Medicamentosas , Modelos Biológicos , Inibidores das Enzimas do Citocromo P-450 , Inibidores Enzimáticos/metabolismo , Hepatócitos/metabolismo , Humanos , Cetoconazol/metabolismo , Fenótipo , Plasma
16.
Chem Res Toxicol ; 20(11): 1709-17, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17939741

RESUMO

BMS-A78277 (1) is a 5,10-dihydrobenzo[beta][1,8]naphthyridine-N-oxide compound that resides in a class of novel non-nucleoside reverse transcriptase inhibitors (NNRTIs), displaying improved activity against clinically relevant mutants of HIV-1 and possessing pharmacokinetic profiles amenable to once-daily dosing. In the course of investigating the nonclinical metabolism of 1, a circulating metabolite specific to the cynomolgus monkey was identified and subsequently characterized as the carboxyindole metabolite 2. The present investigation describes the biotransformation of this NNRTI in cynomolgus monkey, one which results in a net ring contraction of 1. The use of mass spectrometry and high field NMR analysis aided in the structural characterization of metabolite 2, the source of which originated from the urine and bile of cynomolgus monkeys receiving oral doses of 1. Preparation of a synthetic standard of 2 not only provided ultimate structural confirmation but also afforded ample material for biological testing. The metabolism of 1 was investigated in monkey hepatocytes and hepatic subcellular fractions. While microsomes were incapable of generating metabolite 2, incubation of 1 in monkey S9 fractions as well as hepatocytes resulted in measurable levels of the carboxyindole metabolite. Consequently, incubation of 1 in monkey hepatocytes, which were suspended in media containing (18)O-labeled water, resulted in the incorporation of (18)O into the carboxyindole metabolite, 2. These data implicate a mechanism involving the bioactivation of 1 to an electrophilic intermediate that upon hydrolysis undergoes a concerted ring contraction, resulting in the formation of 2. Previously confined to discussions regarding the metabolism of natural products and select aliphatic heterocycles, the present investigation extends the discussion of metabolism-mediated ring contraction to aromatics such as the present naphthyridine compound, 1.


Assuntos
Fármacos Anti-HIV/metabolismo , Óxidos N-Cíclicos/metabolismo , Sistema Enzimático do Citocromo P-450/fisiologia , Naftiridinas/metabolismo , Inibidores da Transcriptase Reversa/metabolismo , Sulfotransferases/fisiologia , Animais , Fármacos Anti-HIV/química , Biotransformação , Óxidos N-Cíclicos/química , Cães , Transcriptase Reversa do HIV/antagonistas & inibidores , Hepatócitos/metabolismo , Macaca fascicularis , Masculino , Microssomos Hepáticos/metabolismo , Naftiridinas/química , Ratos , Ratos Sprague-Dawley , Inibidores da Transcriptase Reversa/química , Especificidade da Espécie
17.
Chem Res Toxicol ; 15(1): 63-75, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11800598

RESUMO

The in vivo and in vitro disposition of DPC 423, a highly potent, selective, and orally bioavailable inhibitor of blood coagulation factor Xa, has recently been described. Several metabolites, some of which were considered potentially reactive, were identified in rats. A novel GSH adduct, the structure of which was not determined conclusively, was isolated from bile of rats dosed with DPC 423. Herein, we describe the complete structural elucidation of this unique GSH conjugate employing LC/MS and high-field NMR. Similar GSH adducts of DPC 602, [13CD2]DPC 602, and SX 737, all structural analogues of DPC 423, were isolated, characterized spectroscopically, and shown to have identical mass fragmentation pathways. The structures of these conjugates were initially suspected to be either an amide with N-S bond or a nitrogen-oxygen juxtaposed amide with a C-S bond. Studies conducted with [13CD2]DPC 602 indicated an aldoxime structure. The concluding evidence came from HMBC NMR spectrum of the conjugate, which showed strong correlation of the cysteine methylene protons with the imino carbon. Further spectroscopic studies with chemically prepared GSH adduct from benzaldehyde oxime confirmed this pattern of correlation. In vivo and in vitro studies with the synthetic oxime intermediate from DPC 423 showed an adduct identical to the one isolated from the bile of rats dosed with DPC 423. This supported the intermediacy of an aldoxime as a precursor to the GSH adducts. It is postulated that the benzylamine moiety of DPC 423 (and its analogues) is oxidized to a hydroxylamine, which is subsequently converted to a nitroso intermediate. Subsequent rearrangement of the nitroso leads to an aldoxime which in turn is metabolized by P450 to a reactive intermediate. The formation of oxime from DPC 423 (and its analogues) was found to be mediated by rat CYP 3A1/2, which were also responsible for converting the oxime to the GSH trappable reactive intermediate. It is postulated that the aldoxime produces a radical or a nitrile oxide intermediate that reacts with GSH and hence produces this unusual GSH adduct. On the basis of synthetic analogy, it is more likely that the nitrile oxide resulting from two-electron oxidation of the aldoxime is the reactive intermediate. Intramolecular kinetic isotope effects were studied with [13CD2]DPC 602 to assess the importance of the metabolic cleavage of the aminomethyl carbon-hydrogen bond in forming this GSH adduct. The lack of isotope effect in forming the aldoxime from [13CD2]DPC 602 suggests its formation does not occur through the imine intermediate. Instead the data supports the postulated mechanism of hydroxylamine and nitroso intermediates as precursors to the aldoxime. However, the formation of the GSH adduct from [13CD2]DPC 602 did show a significant intramolecular kinetic isotope effect (kH/kD = 2.3) since a carbon-deuterium bond had to be broken on the aldoxime prior to the formation of the adduct. A stable nitrile oxide derived from DPC 602 was postulated as the reactive intermediate responsible for forming this unique GSH adduct.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Inibidores do Fator Xa , Fibrinolíticos/metabolismo , Glutationa/metabolismo , Oximas/metabolismo , Pirazóis/metabolismo , Sulfonas/metabolismo , Animais , Relação Dose-Resposta a Droga , Fibrinolíticos/análise , Masculino , Microssomos Hepáticos/metabolismo , Pirazóis/análise , Ratos , Ratos Sprague-Dawley , Sulfonas/análise
18.
Chem Res Toxicol ; 15(1): 48-62, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11800597

RESUMO

The in vitro and in vivo disposition of DPC 423 was investigated in mice, rats, dogs and humans and the metabolites characterized by LC/MS, LC/NMR and high field-NMR. The rodents produced several metabolites that included an aldehyde (M1), a carboxylic acid (M2), a benzyl alcohol (M3), glutamate conjugates (M4 and M5), an acyl glucuronide (M6) and its isomers; a carbamyl glucuronide (M7); a phenol (M8) and its glucuronide conjugate (M9), two glutathione adducts (M10 and M11), a sulfamate conjugate (M12), isomers of an oxime metabolite (M13), and an amide (M14). Humans and dogs produced less complex metabolite profiles than rats. While unchanged DPC 423 was the major component in plasma and urine samples, differences in the metabolic disposition of this compound among species were noted. M1 is believed to be rapidly oxidized to the carboxylic acid (M2), which forms the potentially reactive acyl glucuronide (M6). The formation of novel glutamate conjugates (M4 and M5) and their role in depleting endogenous glutathione have been described previously. The carbamyl glucuronide M7, found as the major metabolite in rats and in other species, was considered nonreactive and was easily hydrolyzed to the parent compound in the presence of beta-glucuronidase. The identification of GSH adducts M10 and M11 led us to postulate the existence of at least two reactive intermediates responsible for their formation, an epoxide and possibly a nitrile oxide, respectively. Although the formation of GSH adducts such as M10 from epoxides has been described before, there are no reports to date describing the existence of a GSH adduct (M11) of an oxime. The formation of a sulfamate conjugate (M12) formed by direct coupling of sulfate to the nitrogen of benzylamine is described. A mechanism is proposed for the formation of the oxime (M13) that involves sequential oxidation of the benzylamine to the corresponding hydroxylamine and nitroso intermediate. The rearrangement of the nitroso intermediate is believed to produce the oxime (M13). In vitro studies suggested that both the oxime (M13) and the aldehyde (M1) were precursors to the carboxylic acid (M2). This is the first demonstration of carboxylic acid formation via an oxime intermediate produced from an amine. The stability of DPC423 in plasma obtained from several species was studied. Significant species differences in the plasma stability of DPC 423 were observed. The formation of the aldehyde metabolite (M1) was found to be catalyzed by a semicarbazide-sensitive monoamine oxidase (SSAO) found in plasma of rabbits, dogs, and rhesus monkeys. Rat, chimpanzee, and human plasma did not form M1.


Assuntos
Inibidores do Fator Xa , Fibrinolíticos/farmacocinética , Pirazóis/farmacocinética , Sulfonas/farmacocinética , Adulto , Idoso , Animais , Cromatografia Líquida de Alta Pressão , Cães , Fibrinolíticos/análise , Humanos , Técnicas In Vitro , Espectroscopia de Ressonância Magnética , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Microssomos Hepáticos/metabolismo , Pessoa de Meia-Idade , Pirazóis/análise , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Sulfonas/análise
19.
J Pharmacol Exp Ther ; 305(1): 57-69, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12649353

RESUMO

The in vitro pharmacological profile of a novel small molecule corticotropin-releasing factor 1 (CRF(1)) receptor antagonist, (+/-)-N-[2-methyl-4-methoxyphenyl]-1-(1-(methoxymethyl)propyl)-6-methyl-1H-1,2,3-triazolo[4,5-c]pyridin-4-amine (SN003), and the characteristics of its radioligand ([(3)H]SN003) are described. SN003 has high affinity and selectivity for CRF(1) receptors expressed in rat cortex, pituitary, and recombinant HEK293EBNA (HEK293e) cells with respective radiolabeled ovine CRF ([(125)I]oCRF) binding K(i) values of 2.5, 7.9, and 6.8 nM. SN003 was shown to be a CRF(1) receptor antagonist inasmuch as it inhibited CRF-induced cAMP accumulation in human CRF(1)HEK293e cells and CRF-stimulated adrenocorticotropin hormone release from rat pituitary cells without agonist activities. Significant decreases in the B(max) of [(125)I]oCRF binding by SN003 suggest that this antagonist is not simply competitive. To further explore the interaction of SN003 with the CRF(1) receptors, [(3)H]SN003 binding to rat cortex and human CRF(1)HEK293e cell membranes was characterized and shown to be reversible and saturable, with K(D) values of 4.8 and 4.6 nM, and B(max) values of 0.142 and 7.42 pmol/mg protein, respectively. The association and dissociation rate constants of [(3)H]SN003 (k(+1) 0.292 nM(-1) min(-1) and k(-1) 0.992 x 10(-2) min(-1)) were also assessed using human CRF(1)HEK293e cell membranes, giving an equilibrium dissociation constant of 3.4 nM. Moreover, [(3)H]SN003 binding displayed a single affinity state and insensitivity to 5'-guanylylimidodiphosphate, consistent with characteristics of antagonist binding. Incomplete inhibition of [(3)H]SN003 binding by CRF peptides also suggests that SN003 is not simply competitive with CRF at CRF(1) receptors. The distribution of [(3)H]SN003 binding sites was consistent with the expression pattern of CRF(1) receptors in rat brain regions. Small molecule CRF(1) antagonist radioligands like [(3)H]SN003 should enable a better understanding of small molecule interactions with the CRF(1) receptor.


Assuntos
Piridinas/farmacologia , Compostos Radiofarmacêuticos/farmacologia , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Triazóis/farmacologia , Hormônio Adrenocorticotrópico/metabolismo , Animais , Sítios de Ligação , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Humanos , Masculino , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Trítio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA