Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Pharm Biol ; 59(1): 1133-1138, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34410882

RESUMO

CONTEXT: Ambrisentan is an oral endothelin-receptor antagonist (ERA). However, there is no report on the interaction between ambrisentan and shikonin. OBJECTIVE: To investigate the effect of shikonin on ambrisentan metabolism in vivo and in vitro. MATERIALS AND METHODS: This study developed an ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method for simultaneous determination of ambrisentan and (S)-4-hydroxymethyl ambrisentan in rat plasma. Twelve male Sprague-Dawley (SD) rats were divided into two groups (n = 6): the control group and shikonin (20 mg/kg) group. The pharmacokinetics of ambrisentan (2.5 mg/kg) were investigated after 30 min. Additionally, human and rat liver microsomes were used to investigate the herb-drug interaction. RESULTS: The UPLC-MS/MS method was shown to be accurate, precise and reliable, and was successfully applied to the herb-drug interaction study of ambrisentan with shikonin. When co-administrated with 20 mg/kg shikonin, the Cmax and AUC(0-∞) of ambrisentan were significantly increased by 44.96 and 16.65%, respectively (p < 0.05). In addition, there were modest decreases in (S)-4-hydroxymethyl ambrisentan Cmax and AUC(0-∞) in the presence of shikonin (p < 0.05), which indicated that these results were in accordance with the inhibition of shikonin on ambrisentan metabolism. Moreover, enzyme kinetic study indicated that shikonin had an inhibitory effect on human and rat microsomes where the IC50 values of shikonin were 5.865 and 6.358 µM, respectively. CONCLUSIONS: Our study indicated that shikonin could inhibit ambrisentan metabolism. Further studies need to be carried out to verify whether similar interaction truly apply in humans and whether this interaction has clinical significance.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Naftoquinonas/farmacologia , Fenilpropionatos/farmacocinética , Piridazinas/farmacocinética , Espectrometria de Massas em Tandem/métodos , Animais , Área Sob a Curva , Interações Ervas-Drogas , Humanos , Masculino , Microssomos Hepáticos , Fenilpropionatos/sangue , Piridazinas/sangue , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes
2.
CPT Pharmacometrics Syst Pharmacol ; 10(5): 428-440, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33818908

RESUMO

Tepotinib is a highly selective and potent MET inhibitor in development for the treatment of patients with solid tumors. Given the favorable tolerability and safety profiles up to the maximum tested dose in the first-in-human (FIH) trial, an efficacy-driven translational modeling approach was proposed to establish the recommended phase II dose (RP2D). To study the in vivo pharmacokinetics (PKs)/target inhibition/tumor growth inhibition relationship, a subcutaneous KP-4 pancreatic cell-line xenograft model in mice with sensitivity to MET pathway inhibition was selected as a surrogate tumor model. Further clinical PK and target inhibition data (derived from predose and postdose paired tumor biopsies) from a FIH study were integrated with the longitudinal PKs and target inhibition profiles from the mouse xenograft study to establish a translational PK/pharmacodynamic (PD) model. Preclinical data showed that tumor regression with tepotinib treatment in KP-4 xenograft tumors corresponded to 95% target inhibition. We therefore concluded that a PD criterion of sustained, near-to-complete (>95%) phospho-MET inhibition in tumors should be targeted for tepotinib to be effective. Simulations of dose-dependent target inhibition profiles in human tumors that exceeded the PD threshold in more than 90% of patients established an RP2D of tepotinib 500 mg once daily. This translational mathematical modeling approach supports an efficacy-driven rationale for tepotinib phase II dose selection of 500 mg once daily. Tepotinib at this dose has obtained regulatory approval for the treatment of patients with non-small cell lung cancer harboring MET exon 14 skipping.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Modelos Teóricos , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Piridazinas/farmacologia , Pirimidinas/farmacologia , Administração Oral , Animais , Ensaios Clínicos Fase II como Assunto , Relação Dose-Resposta a Droga , Humanos , Camundongos , Piperidinas/administração & dosagem , Piperidinas/farmacocinética , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacocinética , Piridazinas/administração & dosagem , Piridazinas/farmacocinética , Pirimidinas/administração & dosagem , Pirimidinas/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Pharmacotherapy ; 39(4): 514-520, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30815916

RESUMO

STUDY OBJECTIVE: The ANRS163-ETRAL study showed that etravirine 200 mg/raltegravir 400 mg twice-daily dual therapy was highly effective in the treatment of human immunodeficiency virus (HIV)-infected patients older than 45 years, with virologic and therapeutic success rates at week 48 of 99.4% and 94.5%, respectively. The objective of this study was to determine whether a clinically significant pharmacokinetic interaction between etravirine and raltegravir exists by assessing steady-state total and unbound etravirine, raltegravir, and inactive raltegravir-glucuronide concentrations 12 hours after last intake (C12h ) in blood plasma (BP) and seminal plasma (SP). DESIGN: Pharmacokinetic analysis of data from the ANRS163-ETRAL study. PATIENTS: One hundred forty-six HIV-1-infected patients (of the 165 patients included in the ANRS-163 ETRAL study) who were receiving etravirine 200 mg and raltegravir 400 mg twice daily. MEASUREMENTS AND MAIN RESULTS: Blood was collected from all 146 patients at weeks 2-4, 12, 24, and 48, and semen was collected from 21 patients at week 48. The extent of BP and SP protein binding was determined by using ultrafiltration assay. Total and unbound etravirine, raltegravir, and raltegravir-glucuronide C12h were determined by ultra high performance liquid chromatography coupled with tandem mass spectrometry and interpreted by using the in vitro calculated protein-bound 95% inhibitory concentration (PBIC95 ) for wild-type (WT) HIV: etravirine (116 ng/ml) and raltegravir (15 ng/ml). Median (interquartile range [IQR]) total BP etravirine C12h (536 ng/ml [376-719]) and raltegravir (278 ng/ml [97-690]) were adequate in 99% and 96% of patients, respectively. Median (IQR) SP:BP C12h ratio and BP unbound fraction were etravirine 0.3 (0.2-0.5) and < 1%, respectively, raltegravir 1.8 (1.3-3.3) and 12%, respectively, and raltegravir-glucuronide 12.0 (6.5-17.7) and > 99%, respectively. The BP raltegravir metabolic ratio (raltegravir glucuronide:raltegravir ratio) was 1.7, suggesting only weak induction of raltegravir glucuronidation by etravirine. Only three patients had etravirine and raltegravir C12h < PBIC95 simultaneously. CONCLUSION: No clinically significant pharmacokinetic interaction between etravirine and raltegravir was detected. Total etravirine and raltegravir BP concentrations were adequate in most patients, favoring virologic efficacy and confirming good treatment adherence (> 95%), despite twice-daily administration. The long half-life of etravirine and higher unbound fraction SP of raltegravir (57%) ensured adequate concentrations of dual therapy in genital compartments. Our results indicate that etravirine and raltegravir have good, complementary pharmacokinetic profiles, suggesting that they could be used in a dual-treatment strategy.


Assuntos
Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/farmacocinética , Infecções por HIV/tratamento farmacológico , Piridazinas/administração & dosagem , Piridazinas/farmacocinética , Raltegravir Potássico/administração & dosagem , Raltegravir Potássico/farmacocinética , Sêmen/efeitos dos fármacos , Fármacos Anti-HIV/sangue , Fármacos Anti-HIV/uso terapêutico , Interações Medicamentosas , Quimioterapia Combinada , Feminino , Infecções por HIV/sangue , HIV-1/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Nitrilas , Ligação Proteica , Piridazinas/sangue , Piridazinas/uso terapêutico , Pirimidinas , Raltegravir Potássico/sangue , Raltegravir Potássico/uso terapêutico , Sêmen/metabolismo
4.
Bioorg Med Chem ; 25(14): 3768-3779, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28571972

RESUMO

A lead compound A was identified previously as an stearoyl coenzyme A desaturase (SCD) inhibitor during research on potential treatments for obesity. This compound showed high SCD1 binding affinity, but a poor pharmacokinetic (PK) profile and limited chemical accessibility, making it suboptimal for use in anticancer research. To identify potent SCD1 inhibitors with more promising PK profiles, we newly designed a series of 'non-spiro' 4, 4-disubstituted piperidine derivatives based on molecular modeling studies. As a result, we discovered compound 1a, which retained moderate SCD1 binding affinity. Optimization around 1a was accelerated by analyzing Hansch-Fujita and Hammett constants to obtain 4-phenyl-4-(trifluoromethyl)piperidine derivative 1n. Fine-tuning of the azole moiety of 1n led to compound 1o (T-3764518), which retained nanomolar affinity and exhibited an excellent PK profile. Reflecting the good potency and PK profile, orally administrated compound 1o showed significant pharmacodynamic (PD) marker reduction (at 0.3mg/kg, bid) in HCT116 mouse xenograft model and tumor growth suppression (at 1mg/kg, bid) in 786-O mouse xenograft model. In conclusion, we identified a new series of SCD1 inhibitors, represented by compound 1o, which represents a promising new chemical tool suitable for the study of SCD1 biology as well as the potential development of novel anticancer therapies.


Assuntos
Antineoplásicos/química , Inibidores Enzimáticos/síntese química , Oxidiazóis/síntese química , Piridazinas/síntese química , Estearoil-CoA Dessaturase/antagonistas & inibidores , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Células HCT116 , Humanos , Concentração Inibidora 50 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microssomos Hepáticos/metabolismo , Oxidiazóis/farmacocinética , Oxidiazóis/uso terapêutico , Oxidiazóis/toxicidade , Piperidinas/química , Piperidinas/metabolismo , Piperidinas/farmacologia , Ligação Proteica , Piridazinas/farmacocinética , Piridazinas/uso terapêutico , Piridazinas/toxicidade , Compostos de Espiro/química , Estearoil-CoA Dessaturase/metabolismo , Relação Estrutura-Atividade , Transplante Heterólogo
5.
Bioorg Med Chem Lett ; 27(14): 3101-3106, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28539220

RESUMO

A series of potent dual JAK1/3 inhibitors have been developed from a moderately selective JAK3 inhibitor. Substitution at the C6 position of the pyrrolopyridazine core with aryl groups provided exceptional biochemical potency against JAK1 and JAK3 while maintaining good selectivity against JAK2 and Tyk2. Translation to in vivo efficacy was observed in a murine model of chronic inflammation. X-ray co-crystal structure determination confirmed the presumed inhibitor binding orientation in JAK3. Efforts to reduce hERG channel inhibition will be described.


Assuntos
Janus Quinase 1/antagonistas & inibidores , Janus Quinase 3/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Piridazinas/química , Pirróis/química , Animais , Sítios de Ligação , Domínio Catalítico , Linhagem Celular , Cristalografia por Raios X , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Meia-Vida , Humanos , Inflamação/prevenção & controle , Concentração Inibidora 50 , Janus Quinase 1/metabolismo , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/metabolismo , Janus Quinase 3/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Conformação Molecular , Simulação de Dinâmica Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacocinética , Piridazinas/síntese química , Piridazinas/farmacocinética , Pirróis/síntese química , Pirróis/farmacocinética , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , TYK2 Quinase/antagonistas & inibidores , TYK2 Quinase/metabolismo
6.
Basic Clin Pharmacol Toxicol ; 116(5): 423-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25286744

RESUMO

To evaluate the impact of CYP2C19 polymorphisms on ambrisentan exposure and to assess its modification by St. John's wort (SJW), 20 healthy volunteers (10 CYP2C19 extensive, four poor and six ultrarapid metabolizers) received therapeutic doses of ambrisentan (5 mg qd po) for 20 days and concomitantly SJW (300 mg tid po) for the last 10 days. To quantify changes of CYP3A4 activity, midazolam (3 mg po) as a probe drug was used. Ambrisentan pharmacokinetics was assessed on days 1, 10 and 20, and midazolam pharmacokinetics before and on days 1, 10, 17 and 20. At steady state, ambrisentan exposure was similar in extensive and ultrarapid metabolizers but 43% larger in poor metabolizers (p < 0.01). In all volunteers, SJW reduced ambrisentan exposure and the relative change (17-26%) was similar in all genotype groups. The extent of this interaction did not correlate with the changes in CYP3A activity (midazolam clearance) (rs = 0.23, p = 0.34). Ambrisentan had no effect on midazolam pharmacokinetics. In conclusion, SJW significantly reduced exposure with ambrisentan irrespective of the CYP2C19 genotype. The extent of this interaction was small and thus likely without clinical relevance.


Assuntos
Citocromo P-450 CYP2C19/genética , Indutores do Citocromo P-450 CYP3A/administração & dosagem , Citocromo P-450 CYP3A/biossíntese , Interações Ervas-Drogas , Hypericum , Fenilpropionatos/farmacocinética , Extratos Vegetais/administração & dosagem , Piridazinas/farmacocinética , Administração Oral , Adulto , Estudos Cross-Over , Citocromo P-450 CYP2C19/metabolismo , Esquema de Medicação , Indução Enzimática , Feminino , Genótipo , Alemanha , Voluntários Saudáveis , Humanos , Masculino , Midazolam/administração & dosagem , Midazolam/farmacocinética , Farmacogenética , Fenótipo , Fenilpropionatos/administração & dosagem , Fenilpropionatos/sangue , Piridazinas/administração & dosagem , Piridazinas/sangue
7.
Bioorg Med Chem Lett ; 24(5): 1303-6, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24513042

RESUMO

A series of fused cyclopropyl-4,5-dihydropyridazin-3-one (3,4-diaza-bicyclo[4.1.0]hept-4-en-2-one) phenoxypiperidine analogs was designed and synthesized, leading to the identification of (1R,6S)-5-[4-(1-cyclobutyl-piperidin-4-yloxy)-phenyl]-3,4-diaza-bicyclo[4.1.0]hept-4-en-2-one (R,S-4a) as a second-generation pyridazin-3-one H3R antagonist. Compound R,S-4a was a potent H3R functional antagonist in vivo in the rat dipsogenia model, demonstrated potent wake activity in the rat EEG/EMG model, and enhanced short-term memory in the rat social recognition memory model at doses as low as 0.03-0.3 mg/kg po.


Assuntos
Nootrópicos/química , Piperidinas/química , Piridazinas/química , Receptores Histamínicos H3/química , Animais , Transtornos Cognitivos/tratamento farmacológico , Modelos Animais de Doenças , Cães , Avaliação Pré-Clínica de Medicamentos , Agonismo Inverso de Drogas , Meia-Vida , Haplorrinos , Memória de Curto Prazo/efeitos dos fármacos , Nootrópicos/farmacocinética , Nootrópicos/farmacologia , Nootrópicos/uso terapêutico , Piperidinas/farmacocinética , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Piridazinas/farmacocinética , Piridazinas/farmacologia , Piridazinas/uso terapêutico , Ratos , Receptores Histamínicos H3/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade
9.
Bioorg Med Chem Lett ; 22(22): 6888-95, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23058884

RESUMO

A novel indolizine class of compounds was identified as TRPV1 antagonist from an HTS campaign. However, this indolizine class proved to be unstable and reacted readily with glutathione when exposed to light and oxygen. Reactivity was reduced by the introduction of a nitrogen atom alpha to the indolizine nitrogen. The pyrrolopyridazine core obtained proved to be inert to the action of light and oxygen. The synthesis route followed the one used for the indolizine compounds, and the potency and ADMET profile proved to be similar.


Assuntos
Piridazinas/química , Pirróis/química , Canais de Cátion TRPV/antagonistas & inibidores , Animais , Células CACO-2 , Permeabilidade da Membrana Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Humanos , Indolizidinas/química , Microssomos Hepáticos/metabolismo , Piridazinas/síntese química , Piridazinas/farmacocinética , Pirróis/síntese química , Pirróis/farmacocinética , Ratos , Relação Estrutura-Atividade , Canais de Cátion TRPV/metabolismo
10.
Bioorg Med Chem Lett ; 22(15): 4967-74, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22765895

RESUMO

mTOR is a critical regulator of cellular signaling downstream of multiple growth factors. The mTOR/PI3K/AKT pathway is frequently mutated in human cancers and is thus an important oncology target. Herein we report the evolution of our program to discover ATP-competitive mTOR inhibitors that demonstrate improved pharmacokinetic properties and selectivity compared to our previous leads. Through targeted SAR and structure-guided design, new imidazopyridine and imidazopyridazine scaffolds were identified that demonstrated superior inhibition of mTOR in cellular assays, selectivity over the closely related PIKK family and improved in vivo clearance over our previously reported benzimidazole series.


Assuntos
Inibidores de Proteínas Quinases/química , Piridazinas/química , Piridinas/química , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Benzimidazóis/química , Sítios de Ligação , Ligação Competitiva , Cristalografia por Raios X , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Meia-Vida , Humanos , Imidazóis/química , Masculino , Camundongos , Microssomos Hepáticos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacocinética , Estrutura Terciária de Proteína , Piridazinas/síntese química , Piridazinas/farmacocinética , Piridinas/síntese química , Piridinas/farmacocinética , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR/metabolismo
11.
Xenobiotica ; 42(5): 456-65, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22122353

RESUMO

The R- and S-enantiomer of N-(4-(3-(1-ethyl-3,3-difluoropiperidin-4-ylamino)-1H-pyrazolo[3,4-b]pyridin-4-yloxy)-3-fluorophenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide are novel MET kinase inhibitors that have been investigated as potential anticancer agents. The effect of the chirality of these compounds on preclinical in vivo pharmacokinetics and toxicity was studied. The plasma clearance for the S-enantiomer was low in mice and monkeys (23.7 and 7.8 mL min(-1) kg(-1), respectively) and high in rats (79.2 mL min(-1) kg(-1)). The R/S enantiomer clearance ratio was 1.5 except in rats (0.49). After oral single-dose administration at 5 mg kg(-1) the R/S enantiomer ratio of AUC(inf) was 0.95, 1.9 and 0.41 in mice, rats and monkeys, respectively. In an oral single-dose dose-ranging study at 200 and 500 mg kg(-1) and multi-dose toxicity study in mice plasma AUC exposure was approximately 2- to 3-fold higher for the R-enantiomer compared to the S-enantiomer. Greater toxicity of the S-enantiomer was observed which appeared to be due to high plasma C(min) values and tissue concentrations approximately 24 h after the final dose. Both enantiomers showed low to moderate permeability in MDCKI cells with no significant efflux, no preferential distribution into red blood cells and similar plasma protein binding in vitro. Overall, the differences between the enantiomers with respect to low dose pharmacokinetics and in vitro properties were relatively modest. However, toxicity results warrant further development of the R-enantiomer over the S-enantiomer.


Assuntos
Inibidores de Proteínas Quinases/farmacocinética , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Pirazóis/farmacocinética , Piridazinas/farmacocinética , Administração Oral , Animais , Proteínas Sanguíneas/metabolismo , Peso Corporal , Linhagem Celular , Permeabilidade da Membrana Celular , Cães , Avaliação Pré-Clínica de Medicamentos , Feminino , Macaca fascicularis , Masculino , Camundongos , Ligação Proteica , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/sangue , Inibidores de Proteínas Quinases/química , Proteínas Proto-Oncogênicas c-met/metabolismo , Pirazóis/administração & dosagem , Pirazóis/sangue , Pirazóis/química , Piridazinas/administração & dosagem , Piridazinas/sangue , Piridazinas/química , Ratos , Ratos Sprague-Dawley , Estereoisomerismo , Fatores de Tempo
12.
Bioorg Med Chem Lett ; 21(15): 4550-5, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21705219

RESUMO

We have discovered imidazo[1,2-b]pyridazine derivatives that show suppressive activity of inflammation in arthritis models. We optimized the substructures of imidazo[1,2-b]pyridazine derivatives to combine potent IKKß inhibitory activity, TNFα inhibitory activity in vivo and excellent pharmacokinetics. The compound we have acquired, which had both potent activities and good pharmacokinetic profiles based on improved physicochemical properties, demonstrated efficacy on collagen-induced arthritis models in mice and rats.


Assuntos
Artrite Experimental/tratamento farmacológico , Quinase I-kappa B/antagonistas & inibidores , Imidazóis/química , Inibidores de Proteínas Quinases/química , Piridazinas/química , Administração Oral , Animais , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Quinase I-kappa B/metabolismo , Camundongos , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/uso terapêutico , Piridazinas/farmacocinética , Piridazinas/uso terapêutico , Ratos , Fator de Necrose Tumoral alfa/metabolismo
13.
Xenobiotica ; 41(4): 327-39, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21182395

RESUMO

GNE-A (AR00451896; N-(4-(3-((3S,4R)-1-ethyl-3-fluoropiperidine-4-ylamino)-1H-pyrazolo[3,4-b]pyridin-4-yloxy)-3-fluorophenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide) is a potent, selective MET kinase inhibitor being developed as a potential drug for the treatment of human cancers. Plasma clearance was low in mice and dogs (15.8 and 2.44 mL/min/kg, respectively) and moderate in rats and monkeys (36.6 and 13.9 mL/min/kg, respectively). The volume of distribution ranged from 2.1 to 9.0 L/kg. The mean terminal elimination half-life ranged from 1.67 h in rats to 16.3 h in dogs. Oral bioavailability in rats, mice, monkeys, and dogs were 11.2%, 88.0%, 72.4%, and 55.8%, respectively. Allometric scaling predicted a clearance of 1.3-7.4 mL/min/kg and a volume of distribution of 4.8-11 L/kg in human. Plasma protein binding was high (96.7-99.0% bound). Blood-to-plasma concentration ratios (0.78-1.46) indicated that GNE-A did not preferentially distribute into red blood cells. Transporter studies in MDCKI-MDR1 and MDCKII-Bcrp1 cells suggested that GNE-A is likely a substrate for MDR1 and BCRP. Pharmacokinetic-pharmacodynamic modelling of tumour growth inhibition in MET-amplified EBC-1 human non-small cell lung carcinoma tumour xenograft mice projected oral doses of 5.6 and 13 mg/kg/day for 50% and 90% tumour growth inhibition, respectively. Overall, GNE-A exhibited favourable preclinical properties and projected human dose estimates.


Assuntos
Antineoplásicos/farmacocinética , Modelos Biológicos , Inibidores de Proteínas Quinases/farmacocinética , Pirazóis/farmacocinética , Piridazinas/farmacocinética , Absorção , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cães , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Haplorrinos , Humanos , Masculino , Camundongos , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Pirazóis/metabolismo , Pirazóis/farmacologia , Piridazinas/metabolismo , Piridazinas/farmacologia , Ratos , Ratos Sprague-Dawley
14.
Chem Res Toxicol ; 23(11): 1743-52, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20825217

RESUMO

Compound 1, (7-methoxy-N-((6-(3-methylisothiazol-5-yl)-[1,2,4]triazolo[4,3-b]pyridazin-3-yl)methyl)-1,5-naphthyridin-4-amine) is a potent, selective inhibitor of c-Met (mesenchymal-epithelial transition factor), a receptor tyrosine kinase that is often deregulated in cancer. Compound 1 displayed desirable pharmacokinetic properties in multiple preclinical species. Glutathione trapping studies in liver microsomes resulted in the NADPH-dependent formation of a glutathione conjugate. Compound 1 also exhibited very high in vitro NADPH-dependent covalent binding to microsomal proteins. Species differences in covalent binding were observed, with the highest binding in rats, mice, and monkeys (1100-1300 pmol/mg/h), followed by dogs (400 pmol/mg/h) and humans (144 pmol/mg/h). This covalent binding to protein was abolished by coincubation with glutathione. Together, these in vitro data suggest that covalent binding and glutathione conjugation proceed via bioactivation to a chemically reactive intermediate. The cytochrome (CYP) P450 enzymes responsible for this bioactivation were identified as cytochrome P450 3A4, 1A2, and 2D6 in human and cytochrome P450 2A2, 3A1, and 3A2 in rats. The glutathione metabolite was detected in the bile of rats and mice, thus demonstrating bioactivation occurring in vivo. Efforts to elucidate the structure of the glutathione adduct led to the isolation and characterization of the metabolite by NMR and mass spectrometry. The analytical data confirmed conclusively that the glutathione conjugation was on the 4-C position of the isothiazole ring. Such P450-mediated bioactivation of an isothiazole or thiazole group has not been previously reported. We propose a mechanism of bioactivation via sulfur oxidation followed by glutathione attack at the 4-position with subsequent loss of water resulting in the formation of the glutathione conjugate. Efforts to reduce bioactivation without compromising potency and pharmacokinetics were undertaken in order to minimize the potential risk of toxicity. Because of the exemplary pharmacokinetic/pharmacodynamic (PK/PD) properties of the isothiazole group, initial attempts were focused on introducing alternative metabolic soft spots into the molecule. These efforts resulted in the discovery of 7-(2-methoxyethoxy)-N-((6-(3-methyl-5-isothiazolyl)[1,2,4]triazolo[4,3-b]pyridazin-3-yl)methyl)-1,5-naphthyridin-4-amine (compound 2), with the major metabolic transformation occurring on the naphthyridine ring alkoxy substituent. However, a glutathione conjugate of compound 2 was produced in vitro and in vivo in a manner similar to that observed for compound 1. Furthermore, the covalent binding was high across species (360, 300, 529, 208, and 98 pmol/mg/h in rats, mice, dogs, monkeys, and humans, respectively), but coincubation with glutathione reduced the extent of covalent binding. The second viable alternative in reducing bioactivation involved replacing the isothiazole ring with bioisosteric heterocycles. Replacement of the isothiazole ring with an isoxazole or a pyrazole reduced the bioactivation while retaining the desirable PK/PD characteristics of compounds 1 and 2.


Assuntos
Naftiridinas/metabolismo , Piridazinas/metabolismo , Tiazóis/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Sistema Enzimático do Citocromo P-450/metabolismo , Cães , Avaliação Pré-Clínica de Medicamentos , Glutationa/química , Humanos , Espectroscopia de Ressonância Magnética , Camundongos , Microssomos Hepáticos/metabolismo , Conformação Molecular , Naftiridinas/química , Naftiridinas/farmacocinética , Naftiridinas/toxicidade , Ligação Proteica , Piridazinas/química , Piridazinas/farmacocinética , Piridazinas/toxicidade , Ratos , Fatores de Risco , Espectrometria de Massas por Ionização por Electrospray , Tiazóis/química , Tiazóis/toxicidade
15.
Adv Pharmacol ; 57: 137-85, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20230761

RESUMO

TPA023 is a GABA(A) alpha2/alpha3 subtype-selective modulator which in preclinical species has anxiolytic-like activity but does not produce sedative-like properties and is without abuse potential. It has good oral bioavailability in rat and dog but not in rhesus monkey (respective oral bioavailability values of 36, 54, and 1%), and in all the three species the half-life after i.v. administration was relatively short (0.6-1.5 h). The plasma concentrations of TPA023 required to produce 50% receptor occupancy were 21-25, 19, and 9 ng/mL in rats, baboons, and humans, respectively. In man, TPA023 has a half-life of around 3-6h when administered as an immediate release formulation, but exposure was more prolonged when it was formulated into a controlled release, gel extrusion module (GEM) tablet. In vivo metabolism was via t-butyl hydroxylation and N-deethylation. A drug-drug interaction study with itraconazole confirmed in vitro metabolic results implicating CYP3A enzymes as the major contributors to in vivo oxidative metabolism. The maximum tolerated doses in healthy, normal volunteers were 2 and 8mg for the immediate-release and GEM formulations, respectively. A post hoc analysis of three separate Phase IIa studies, all of which were halted prematurely, showed that TPA023 reduced scores on the Hamilton Anxiety Scale to a significantly greater extent than placebo. In addition, TPA023 has recently been reported to produce a trend toward improved cognitive performance in a small group of schizophrenia patients. Collectively, these data demonstrate that the alpha2/alpha3-selective partial agonist efficacy of TPA023 translates into a novel pharmacological profile.


Assuntos
Desenho de Fármacos , Piridazinas/farmacologia , Receptores de GABA-A/metabolismo , Triazóis/farmacologia , Sequência de Aminoácidos , Animais , Ansiedade/tratamento farmacológico , Avaliação Pré-Clínica de Medicamentos , Humanos , Dados de Sequência Molecular , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Piridazinas/química , Piridazinas/farmacocinética , Piridazinas/uso terapêutico , Receptores de GABA-A/química , Triazóis/química , Triazóis/farmacocinética , Triazóis/uso terapêutico
16.
Expert Opin Drug Metab Toxicol ; 2(3): 447-58, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16863445

RESUMO

Drugs in development for the management of HIV type 1 (HIV-1) infection include agents in existing classes and agents of novel classes. Of existing classes, new protease inhibitors, nucleoside reverse transcriptase inhibitors and non-nucleoside reverse transcriptase inhibitors are in development. Novel therapeutic approaches include the development of chemokine receptor (CCR)5 antagonists, integrase inhibitors and maturation inhibitors. CCR5 antagonists are thought to inhibit HIV-1 entry into host cells by occupying a specific site on the CCR5 receptor, preventing attachment of the HIV-1 envelope protein gp120. Integrase inhibitors are small synthetically prepared molecules that block RNA/DNA interactions and modify protein or enzyme synthesis. Data on the pharmacokinetics and pharmacodynamics of these new antiretroviral agents continue to generate interest. This review reports the known data on the pharmacokinetics of experimental antiretrovirals, and describe the main drug-drug interactions studied so far.


Assuntos
Antirretrovirais/farmacocinética , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Animais , Antirretrovirais/farmacologia , Antirretrovirais/uso terapêutico , Antagonistas dos Receptores CCR5 , Ensaios Clínicos como Assunto , Cicloexanos/farmacocinética , Cicloexanos/farmacologia , Cicloexanos/uso terapêutico , Darunavir , Avaliação Pré-Clínica de Medicamentos , Interações Medicamentosas , Infecções por HIV/enzimologia , Inibidores de Integrase de HIV/farmacocinética , Inibidores de Integrase de HIV/uso terapêutico , Inibidores da Protease de HIV/farmacocinética , Inibidores da Protease de HIV/uso terapêutico , HIV-1/enzimologia , Humanos , Maraviroc , Nitrilas , Compostos Orgânicos/farmacocinética , Compostos Orgânicos/uso terapêutico , Inibidores de Proteases/farmacocinética , Inibidores de Proteases/uso terapêutico , Piridazinas/farmacocinética , Piridazinas/uso terapêutico , Pirimidinas , Pirrolidinonas , Raltegravir Potássico , Receptores CCR5/metabolismo , Inibidores da Transcriptase Reversa/farmacocinética , Inibidores da Transcriptase Reversa/uso terapêutico , Sulfonamidas/farmacocinética , Sulfonamidas/uso terapêutico , Triazóis/farmacocinética , Triazóis/farmacologia , Triazóis/uso terapêutico
17.
J Neurosci ; 26(2): 662-70, 2006 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-16407564

RESUMO

A corollary of the neuroinflammation hypothesis is that selective suppression of neurotoxic products produced by excessive glial activation will result in neuroprotection. We report here that daily oral administration to mice of the brain-penetrant compound 4,6-diphenyl-3-(4-(pyrimidin-2-yl)piperazin-1-yl)pyridazine (MW01-5-188WH), a selective inhibitor of proinflammatory cytokine production by activated glia, suppressed the human amyloid-beta (Abeta) 1-42-induced upregulation of interleukin-1beta, tumor necrosis factor-alpha, and S100B in the hippocampus. Suppression of neuroinflammation was accompanied by restoration of hippocampal synaptic dysfunction markers synaptophysin and postsynaptic density-95 back toward control levels. Consistent with the neuropathophysiological improvements, MW01-5-188WH therapy attenuated deficits in Y maze behavior, a hippocampal-linked task. Oral MW01-5-188WH therapy begun 3 weeks after initiation of intracerebroventricular infusion of human Abeta decreased the numbers of activated astrocytes and microglia and the cytokine levels in the hippocampus without modifying amyloid plaque burden or altering peripheral tissue cytokine upregulation in response to an in vivo inflammatory challenge. The results provide a novel integrative chemical biology proof in support of the neuroinflammation hypothesis of disease progression, demonstrate that neurodegeneration can be attenuated independently of plaque modulation by targeting innate brain proinflammatory cytokine responses, and indicate the feasibility of developing efficacious, safe, and selective therapies for neurodegenerative disorders by targeting key glial activation pathways.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Anti-Inflamatórios não Esteroides/uso terapêutico , Astrócitos/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Citocinas/biossíntese , Hipocampo/efeitos dos fármacos , Microglia/efeitos dos fármacos , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Fragmentos de Peptídeos/toxicidade , Piperazinas/uso terapêutico , Piridazinas/uso terapêutico , Administração Oral , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/antagonistas & inibidores , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacocinética , Anti-Inflamatórios não Esteroides/toxicidade , Astrócitos/metabolismo , Disponibilidade Biológica , Encéfalo/metabolismo , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Citocinas/genética , Depressão Química , Avaliação Pré-Clínica de Medicamentos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiologia , Humanos , Infusões Parenterais , Interleucina-1/biossíntese , Interleucina-1/genética , Lipopolissacarídeos/farmacologia , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microssomos Hepáticos/metabolismo , Fatores de Crescimento Neural/biossíntese , Fatores de Crescimento Neural/genética , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacocinética , Fármacos Neuroprotetores/toxicidade , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/antagonistas & inibidores , Piperazinas/administração & dosagem , Piperazinas/farmacocinética , Piperazinas/toxicidade , Placa Amiloide/patologia , Piridazinas/administração & dosagem , Piridazinas/farmacocinética , Piridazinas/toxicidade , Ratos , Subunidade beta da Proteína Ligante de Cálcio S100 , Proteínas S100/biossíntese , Proteínas S100/genética , Método Simples-Cego , Fator de Necrose Tumoral alfa/biossíntese
18.
Antimicrob Agents Chemother ; 48(12): 4680-6, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15561844

RESUMO

Nonnucleoside reverse transcriptase inhibitors (NNRTIs) are potent inhibitors of human immunodeficiency virus type 1 (HIV-1); however, currently marketed NNRTIs rapidly select resistant virus, and cross-resistance within the class is extensive. A parallel screening strategy was applied to test candidates from a series of diarylpyrimidines against wild-type and resistant HIV strains carrying clinically relevant mutations. Serum protein binding and metabolic stability were addressed early in the selection process. The emerging clinical candidate, TMC125, was highly active against wild-type HIV-1 (50% effective concentration [EC50] = 1.4 to 4.8 nM) and showed some activity against HIV-2 (EC50 = 3.5 microM). TMC125 also inhibited a series of HIV-1 group M subtypes and circulating recombinant forms and a group O virus. Incubation of TMC125 with human liver microsomal fractions suggested good metabolic stability (15% decrease in drug concentration and 7% decrease in antiviral activity after 120 min). Although TMC125 is highly protein bound, its antiviral effect was not reduced by the presence of 45 mg of human serum albumin/ml, 1 mg of alpha1-acid glycoprotein/ml, or 50% human serum. In an initial screen for activity against a panel of 25 viruses carrying single and double reverse transcriptase amino acid substitutions associated with NNRTI resistance, the EC50 of TMC125 was <5 nM for 19 viruses, including the double mutants K101E+K103N and K103N+Y181C. TMC125 also retained activity (EC50 < 100 nM) against 97% of 1,081 recent clinically derived recombinant viruses resistant to at least one of the currently marketed NNRTIs. TMC125 is a potent next generation NNRTI, with the potential for use in individuals infected with NNRTI-resistant virus.


Assuntos
HIV-1/efeitos dos fármacos , Piridazinas/farmacologia , Inibidores da Transcriptase Reversa/farmacologia , Linhagem Celular , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Farmacorresistência Viral , Genótipo , HIV-1/genética , Humanos , Testes de Sensibilidade Microbiana , Microssomos Hepáticos/metabolismo , Mutagênese Sítio-Dirigida , Nitrilas , Piridazinas/farmacocinética , Pirimidinas , Inibidores da Transcriptase Reversa/farmacocinética
19.
Int J Pharm ; 286(1-2): 69-80, 2004 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-15501003

RESUMO

Solid dispersions using Pluronic F-68 as a carrier were studied for improving the dissolution and bioavailability of ABT-963, a poorly water-soluble compound. The solid dispersions were prepared either by evaporation of the ethanol solutions containing ABT-963 and Pluronic, or by cooling the hot melt of the drug in the carrier. The dispersions were characterized using differential scanning calorimetry, powder X-ray diffractometry, scanning electron microscopy, elemental mapping, and by constructing the melting point phase diagram. In vitro dissolution and in vivo oral bioavailability in fasted dogs were compared for the solid dispersion and a conventional IR capsule formulation. Results showed that, at a composition of approximately 7.5%, ABT-963 formed a eutectic mixture with Pluronic F-68. Both the drug and the polymer were crystalline in the solid dispersion with a wide range of composition of each component. The solid dispersion substantially increased the in vitro dissolution rate of ABT-963. Dosing of the dispersion to fasted dogs resulted in a significant increase of oral bioavailability compared with the conventional IR capsule formulation. These results show that solid dispersion is a promising approach for developing ABT-963 drug products.


Assuntos
Química Farmacêutica/métodos , Inibidores de Ciclo-Oxigenase/farmacocinética , Poloxâmero/farmacocinética , Piridazinas/farmacocinética , Sulfonas/farmacocinética , Administração Oral , Algoritmos , Animais , Disponibilidade Biológica , Varredura Diferencial de Calorimetria/métodos , Cápsulas , Técnicas de Química Combinatória/métodos , Inibidores de Ciclo-Oxigenase/administração & dosagem , Inibidores de Ciclo-Oxigenase/química , Cães , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Avaliação Pré-Clínica de Medicamentos/métodos , Alimentos , Teste de Materiais/métodos , Poloxâmero/administração & dosagem , Poloxâmero/química , Piridazinas/administração & dosagem , Piridazinas/química , Solubilidade/efeitos dos fármacos , Sulfonas/administração & dosagem , Sulfonas/química , Difração de Raios X/métodos
20.
Eur J Drug Metab Pharmacokinet ; 26(4): 263-71, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11808869

RESUMO

The excretion and metabolism of (+/-) [6-(3,4-dihydro-3-oxo-1,4[2H]-benzoxazine-yl)-2,3,4,5-tetrahydro-5-methylpyridazin-3-one] (bemoradan; RWJ-22867) have been investigated in male Long-Evans rats and female beagle dogs. Radiolabeled [14C] bemoradan was administered to rats as a singkle 1 mg/kg suspension dose while the dogs received 0.1 mg/kg suspension dose. Plasma (0-24 h; rat and dog), urine (0-72 h; rat and dog) and fecal (0-72 h; rat and dog) samples were collected and analyzed. The terminal half-life of the total radioactivity for rats from plasma was estimate to be 4.3 +/- 0.1 h while for dogs it was 7.5 +/- 1.3 h. Recoveries of total radioactivity in urine and feces for rats were 49.1 +/- 2.4% and 51.1 +/- 4.9% of th dose, respectively. Recoveries of total radioactivity in urine and feces for dogs were 56.2 +/- 12.0% and 42.7 V 9.9% of the dose, respectively. Bemoradan and a total of nine metabolites were isolated and tentatively identified in rat and dog plasma, urine, and fecal extracts. Unchanged bemoradan accounted for approimately < 2% of the dose in rat urine and 20% in rat feces. Unchanged bemoradan accounted for approximately 5% of the dose in urine and 16% in feces in dog. Six proposed pathways were used to describe the metabolites found in rats and dogs: pyridazinyl oxidations, methyl hydroxylation, hydration, N-oxidation, dehydration and phase II conjugations.


Assuntos
Cardiotônicos/farmacocinética , Oxazinas/farmacocinética , Piridazinas/farmacocinética , Animais , Benzoxazinas , Cromatografia Líquida de Alta Pressão , Cromatografia em Camada Fina , Cães , Avaliação Pré-Clínica de Medicamentos , Feminino , Espectroscopia de Ressonância Magnética , Masculino , Espectrometria de Massas , Modelos Químicos , Ratos , Ratos Long-Evans
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA