Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
6.
Bioorg Med Chem Lett ; 25(21): 4812-4819, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26195137

RESUMO

The IC50 of a beta-secretase (BACE-1) lead compound was improved ∼200-fold from 11 µM to 55 nM through the addition of a single methyl group. Computational chemistry, small molecule NMR, and protein crystallography capabilities were used to compare the solution conformation of the ligand under varying pH conditions to its conformation when bound in the active site. Chemical modification then explored available binding pockets adjacent to the ligand. A strategically placed methyl group not only maintained the required pKa of the piperidine nitrogen and filled a small hydrophobic pocket, but more importantly, stabilized the conformation best suited for optimized binding to the receptor.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Hidantoínas/química , Hidantoínas/farmacologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Humanos , Hidantoínas/síntese química , Metilação , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade
7.
J Phys Condens Matter ; 23(8): 086005, 2011 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-21411908

RESUMO

The destruction of the ferromagnetism of alloyed SrRuO(3) can be caused by electron localization at the substitution sites. Among all the non-magnetic cations that enter the B site, Zr(4+) is the least disruptive to conductivity and ferromagnetism. This is because Zr(4+) does not cause any charge disorder, and its empty d electron states which are poorly matched in energy with the Ru t(2g)(4) states cause the least resonance scattering of Ru's d electrons. Conducting Sr(Ru, Zr)O(3) may be used as an electrode for perovskite-based thin film devices, while its insulating counterpart provides unprecedented magnetoresistance, seldom seen in other non-manganite and non-cobaltite perovskites.

8.
Circulation ; 117(16): 2096-103, 2008 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-18413497

RESUMO

BACKGROUND: Local drug delivery from polymer-coated stents has demonstrated efficacy for preventing in-stent restenosis; however, both the inflammatory effects of polymer coatings and concerns about late outcomes of drug-eluting stent use indicate the need to investigate innovative approaches, such as combining localized gene therapy with stent angioplasty. Thus, we investigated the hypothesis that adenoviral vectors (Ad) could be delivered from the bare-metal surfaces of stents with a synthetic complex for reversible vector binding. METHODS AND RESULTS: We synthesized the 3 components of a gene vector binding complex: (1) A polyallylamine bisphosphonate with latent thiol groups (PABT), (2) a polyethyleneimine (PEI) with pyridyldithio groups for amplification of attachment sites [PEI(PDT)], and (3) a bifunctional (amine- and thiol-reactive) cross-linker with a labile ester bond (HL). HL-modified Ad attached to PABT/PEI(PDT)-treated steel surfaces demonstrated both sustained release in vitro over 30 days and localized green fluorescent protein expression in rat arterial smooth muscle cell cultures, which were not sensitive to either inhibition by neutralizing anti-Ad antibodies or inactivation after storage at 37 degrees C. In rat carotid studies, deployment of steel stents configured with PABT/PEI(PDT)/HL-tethered adenoviral vectors demonstrated both site-specific arterial Ad(GFP) expression and adenovirus-luciferase transgene activity per optical imaging. Rat carotid stent delivery of adenovirus encoding inducible nitric oxide synthase resulted in significant inhibition of restenosis. CONCLUSIONS: Reversible immobilization of adenovirus vectors on the bare-metal surfaces of endovascular stents via a synthetic complex represents an efficient, tunable method for sustained release of gene vectors to the vasculature.


Assuntos
Implantes Absorvíveis , Estenose das Carótidas/prevenção & controle , Estenose das Carótidas/terapia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Stents , Adenoviridae/genética , Animais , Aorta/citologia , Células Cultivadas , Reagentes de Ligações Cruzadas , Proteínas de Fluorescência Verde/genética , Masculino , Metais , Músculo Liso Vascular/citologia , Testes de Neutralização , Óxido Nítrico Sintase Tipo II/genética , Ratos , Ratos Sprague-Dawley , Prevenção Secundária
9.
Xenobiotica ; 33(6): 643-54, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12851040

RESUMO

1. The use of a beneficial interaction between indinavir and compound A, a potent investigational HIV protease inhibitor to enhance systemic exposure of compound A, was investigated. 2. When administrated alone, compound A underwent extensive hepatic first-pass metabolism in rats and monkeys, resulting in low oral bioavailability. 3. In vitro studies with liver microsomes revealed that compound A metabolism was mediated exclusively by CYP3A enzymes in rats, dogs and monkeys. Indinavir, which also was metabolized predominantly by CYP3A enzymes, extensively inhibited compound A metabolism in microsomes, whereas compound A showed weak inhibitory potency on indinavir metabolism. 4. Consistent with in vitro observations, co-administration of the two compounds resulted in a 17-fold increase in oral AUC of compound A in rats owing to the inhibition of metabolism of compound A by indinavir, whereas compound A did not affect indinavir metabolism as indicated by the unchanged indinavir AUC. Similarly, the systemic exposure of compound A in dogs and monkeys was increased substantially following oral co-administration with indinavir by 7- and > 50-fold, respectively. 5. Enhancement in compound A systemic exposure by indinavir in humans, as predicted based on the in vivo animal and in vitro human liver microsomal data, was confirmed in subsequent clinical studies.


Assuntos
Benzofuranos/farmacologia , Inibidores da Protease de HIV/farmacologia , Inibidores da Protease de HIV/farmacocinética , Indinavir/análogos & derivados , Indinavir/farmacologia , Piperazinas/farmacologia , Animais , Anticorpos Bloqueadores/farmacologia , Área Sob a Curva , Benzofuranos/antagonistas & inibidores , Benzofuranos/farmacocinética , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP3A , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450 , Cães , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Inibidores da Protease de HIV/antagonistas & inibidores , Indinavir/antagonistas & inibidores , Indinavir/farmacocinética , Injeções Intravenosas , Macaca mulatta , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Piperazinas/antagonistas & inibidores , Piperazinas/farmacocinética , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Espectrofotometria Ultravioleta
10.
Drug Metab Dispos ; 29(12): 1578-87, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11717177

RESUMO

Compound I (1-(3-chlorophenyl)-4-[(1-(4-cyanobenzyl)-1H-imidazol-5-yl)methyl]piperazin-2-one) is a potent and selective inhibitor of farnesyl-protein transferase (FPTase). The pharmacokinetics and metabolism of compound I displayed species differences in rats and dogs. After oral administration, the drug was well absorbed in dogs but less so in rats. Following i.v. administration, compound I was cleared rapidly in rats in a polyphasic manner with a terminal t(1/2) of 41 min. The plasma clearance (CL(p)) and volume of distribution (V(dss)) were 41.2 ml/min/kg and 1.2 l/kg, respectively. About 1% of the dose was excreted in rat bile and urine as unchanged drug over a period of 24 h, suggesting that biotransformation is the major route of elimination of compound I. Using liquid chromatography (LC)-tandem mass spectometry, nineteen metabolites of compound I were identified in urine and bile from dogs and rats. Structures of two major metabolites were confirmed by LC-NMR. N-Dealkylation and phase II metabolism were the major metabolic pathways. Animal and human liver microsomal intrinsic clearance values were scaled to predict hepatic clearance and half-life in humans, and the predicted values were in good agreement to the in vivo data.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Imidazóis/farmacologia , Piperazinas/farmacologia , Algoritmos , Animais , Área Sob a Curva , Bile/metabolismo , Ductos Biliares/metabolismo , Biotransformação , Proteínas Sanguíneas/metabolismo , Cromatografia Líquida de Alta Pressão , Cães , Farnesiltranstransferase , Meia-Vida , Humanos , Técnicas In Vitro , Absorção Intestinal , Masculino , Espectrometria de Massas , Microssomos Hepáticos , Ratos , Ratos Sprague-Dawley
12.
Bioorg Med Chem Lett ; 11(4): 537-40, 2001 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-11229765

RESUMO

The evaluation of SAR associated with the insertion of carbonyl groups at various positions of N-arylpiperazinone farnesyltransferase inhibitors is described herein. 1-Aryl-2,3-diketopiperazine derivatives exhibited the best balance of potency and pharmacokinetic profile relative to the parent 1-aryl-2-piperazinones.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Piperazinas/farmacologia , Animais , Cães , Inibidores Enzimáticos/farmacocinética , Farnesiltranstransferase , Relação Estrutura-Atividade
13.
Org Lett ; 2(22): 3473-6, 2000 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-11082012

RESUMO

[reaction: see text] Synthesis of the 8-amino-5,6,7,8-tetrahydroimidazo[1,5-a]pyridine ring system was accomplished by intramolecular cyclization of an iminium ion, derived from condensation of an amine and a substituted gamma-(1-imidazolyl)butyraldehyde. The reaction was used to produce conformationally restricted farnesyltransferase inhibitor analogues which exhibit improved in vivo metabolic stability.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Inibidores Enzimáticos/síntese química , Imidazóis/síntese química , Piridinas/síntese química , Administração Oral , Animais , Cães , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Farnesiltranstransferase , Imidazóis/química , Imidazóis/farmacocinética , Imidazóis/farmacologia , Indicadores e Reagentes , Modelos Moleculares , Conformação Molecular , Piridinas/química , Piridinas/farmacocinética , Piridinas/farmacologia , Relação Estrutura-Atividade
14.
J Med Chem ; 43(18): 3386-99, 2000 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-10978186

RESUMO

Recent results from human clinical trials have established the critical role of HIV protease inhibitors in the treatment of acquired immune-deficiency syndrome (AIDS). However, the emergence of viral resistance, demanding treatment protocols, and adverse side effects have exposed the urgent need for a second generation of HIV protease inhibitors. The continued exploration of our hydroxylaminepentanamide (HAPA) transition-state isostere series of HIV protease inhibitors, which initially resulted in the identification of Crixivan (indinavir sulfate, MK-639, L-735,524), has now yielded MK-944a (L-756,423). This compound is potent, is selective, and competitively inhibits HIV-1 PR with a K(i) value of 0.049 nM. It stops the spread of the HIV(IIIb)-infected MT4 lymphoid cells at 25.0-50.0 nM, even in the presence of alpha(1) acid glycoprotein, human serum albumin, normal human serum, or fetal bovine serum. MK-944a has a longer half-life in several animal models (rats, dogs, and monkeys) than indinavir sulfate and is currently in advanced human clinical trials.


Assuntos
Antivirais/síntese química , Inibidores da Protease de HIV/síntese química , HIV-1/efeitos dos fármacos , Indanos/síntese química , Piperazinas/síntese química , Animais , Antivirais/química , Antivirais/farmacocinética , Antivirais/farmacologia , Bovinos , Técnicas de Cultura de Células , Cães , Avaliação Pré-Clínica de Medicamentos , Resistência Microbiana a Medicamentos , Inibidores da Protease de HIV/química , Inibidores da Protease de HIV/farmacocinética , Inibidores da Protease de HIV/farmacologia , Haplorrinos , Humanos , Indanos/química , Indanos/farmacocinética , Indanos/farmacologia , Masculino , Piperazinas/química , Piperazinas/farmacocinética , Piperazinas/farmacologia , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Cálculos Urinários/induzido quimicamente , Cálculos Urinários/urina
16.
Drug Metab Dispos ; 28(4): 460-6, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10725315

RESUMO

L-754,394, a furanopyridine derivative, is an experimental HIV protease inhibitor. Previous studies from this laboratory have demonstrated that L-754,394 is cleared very rapidly in animals, and that this drug is a potent mechanism-based inactivator (suicide inhibitor) for CYP3A4 in human liver microsomes. Because L-754,394 is a high-clearance drug and an enzyme inactivator, it is expected that this drug will be subject to significant first-pass metabolism, and that the degree of enzyme inactivation will be dependent not only on the dose, but also on the route of administration. The purpose of this study is to examine the effects of dose and route of administration on the kinetics of L-754,394 using rats and dogs as animal models. In both rats and dogs, L-754,394 exhibited marked dose-dependent pharmacokinetics after i.v. and oral administration. Irrespective of i.v. or oral administration, the area under the plasma concentration-time curve from zero to infinity increased with dose in a greater than proportional manner. However, the magnitude of area under the plasma concentration-time curve from zero to infinity increase was much greater after oral dosing than after i.v. administration, indicating route-dependent pharmacokinetics. Data from in vitro and in vivo studies suggested that the dose- and route-dependent pharmacokinetics were due mainly to the inactivation (destruction) of the enzymes responsible for its own metabolism.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Inibidores da Protease de HIV/farmacocinética , Indanos/farmacocinética , Piperazinas/farmacocinética , Administração Oral , Animais , Anticorpos Bloqueadores/farmacologia , Área Sob a Curva , Cromatografia Líquida de Alta Pressão , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/metabolismo , Cães , Relação Dose-Resposta a Droga , Inibidores da Protease de HIV/administração & dosagem , Inibidores da Protease de HIV/sangue , Técnicas In Vitro , Indanos/administração & dosagem , Indanos/sangue , Injeções Intravenosas , Fígado/enzimologia , Fígado/metabolismo , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Dinâmica não Linear , Perfusão , Piperazinas/administração & dosagem , Piperazinas/sangue , Ratos , Ratos Sprague-Dawley , Esteroide Hidroxilases/antagonistas & inibidores , Esteroide Hidroxilases/metabolismo , Testosterona/sangue
17.
Drug Metab Dispos ; 27(10): 1187-93, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10497146

RESUMO

Indinavir, a potent and specific inhibitor of HIV protease, is a known substrate of cytochrome P-450 (CYP) 3A and p-glycoprotein. The purpose of this study is to investigate and compare the inducing effect of dexamethasone (DEX) on CYP3A and p-glycoprotein in the hepatic and intestinal first-pass metabolism of indinavir in rats. Pretreatment of rats with DEX had little effect on the pharmacokinetics (Cl and T(1/2)) after i.v. administration of indinavir, whereas DEX markedly altered the peak concentration (C(max)) and bioavailability of indinavir after oral dosing. The C(max) decreased from 2.8 microM in control rats to 0.28 microM in DEX-treated rats, and bioavailability decreased from 28 to 12.4%. The decreased bioavailability after DEX pretreatment was due mainly to an increase in first-pass metabolism. Intestinal first-pass metabolism (E(G)) increased from 6% in control rats to 34% in DEX-treated rats, and hepatic first-pass metabolism (E(H)) increased from 65 to 82%. Analysis of in vitro kinetic data revealed that the increased intestinal and hepatic metabolism by DEX was attributed to an increase in the V(max), as a result of CYP3A induction, without a significant change in the K(m) values. DEX pretreatment also induced p-glycoprotein in the intestine and liver of rats. p-Glycoprotein appeared to increase the intestinal metabolism of indinavir whereas it had little effect on the hepatic metabolism of indinavir. Although it has been suggested that the role of intestinal metabolism for some drugs is quantitatively greater than that of hepatic metabolism in the overall first-pass metabolism, the contribution of intestinal metabolism to the overall first-pass metabolism of indinavir in rats is not quantitatively as important as the hepatic metabolism, regardless of DEX induction.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Fármacos Anti-HIV/farmacocinética , Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/biossíntese , Dexametasona/farmacologia , Indinavir/farmacocinética , Mucosa Intestinal/metabolismo , Oxirredutases N-Desmetilantes/biossíntese , Animais , Western Blotting , Citocromo P-450 CYP3A , Fígado/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley
18.
J Med Chem ; 41(23): 4466-74, 1998 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-9804686

RESUMO

We have addressed the key deficiency of noncovalent pyridinone acetamide thrombin inhibitor L-374,087 (1), namely, its modest half-lives in animals, by making a chemically stable 3-alkylaminopyrazinone bioisostere for its 3-sulfonylaminopyridinone core. Compound 3 (L-375,378), the closest aminopyrazinone analogue of 1, has comparable selectivity and slightly decreased efficacy but significantly improved pharmacokinetics in rats, dogs, and monkeys to 1. We have developed an efficient and versatile synthesis of 3, and this compound has been chosen for further preclinical and clinical development.


Assuntos
Aminopiridinas/síntese química , Peptídeos/química , Pirazinas/síntese química , Piridonas/síntese química , Trombina/antagonistas & inibidores , Aminopiridinas/química , Aminopiridinas/farmacocinética , Aminopiridinas/farmacologia , Animais , Disponibilidade Biológica , Cristalografia por Raios X , Cães , Macaca mulatta , Modelos Moleculares , Mimetismo Molecular , Pirazinas/química , Pirazinas/farmacocinética , Pirazinas/farmacologia , Piridonas/química , Piridonas/farmacocinética , Piridonas/farmacologia , Ratos , Relação Estrutura-Atividade
19.
Biochem Pharmacol ; 56(2): 223-30, 1998 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-9698076

RESUMO

P450 complex formation and the unusual pharmacokinetics of methylenedioxyphenyl HIV protease inhibitors were examined by in vitro studies using human and rat liver microsomes and by in vivo oral dosing studies. In vitro spectral studies indicated that the formation of a P450 complex having absorbance maxima at 425 and 456 nm was time and concentration dependent; 27-60% of the total P450 was complexed in dexamethasone-induced rat liver microsomes after a 30-min incubation with 100 microM HIV protease inhibitors. Methoxy substitution on the phenyl ring of the methylenedioxyphenyl moiety increased formation of the P450 complex, whereas chlorine substitution markedly decreased the P450 complexation. Kinetic studies on the P450 complex formation indicated that both methoxy and chlorine substitution affected the maximum complex formation rate (Vmax), while it had little effect on Km values (approximately 10 microM). This complexation in human liver microsomes was inhibited markedly by an anti-CYP3A1 antibody. Furthermore, the P450 complex formation resulted in a time-dependent loss of CYP3A-catalyzed marker activities (testosterone 2beta/6beta-hydroxylase) in both rat and human liver microsomes. Collectively, these results point to the involvement of CYP3A isoforms in P450 complexation by methylenedioxyphenyl HIV protease inhibitors. Additionally, after oral administration to rats, one of these HIV protease inhibitors (Compound I), which complexed P450 to the greatest extent, showed no elimination over a period of 500 min after administration of the highest dose. It is suggested that formation of a quasi-irreversible metabolite-CYP3A complex with methylenedioxyphenyl HIV protease inhibitors was responsible for the CYP3A-selective time-dependent loss of catalytic function and the unusual dose-dependent pharmacokinetics after oral administration.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/metabolismo , Inibidores da Protease de HIV/farmacologia , Isoenzimas/metabolismo , Microssomos Hepáticos/efeitos dos fármacos , Animais , Biotransformação , Catálise , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP3A , Inibidores da Protease de HIV/metabolismo , Inibidores da Protease de HIV/farmacocinética , Meia-Vida , Humanos , Hidroxilação , Masculino , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Ratos , Ratos Sprague-Dawley
20.
J Med Chem ; 41(17): 3210-9, 1998 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-9703466

RESUMO

As part of an ongoing effort to prepare therapeutically useful orally active thrombin inhibitors, we have synthesized a series of compounds that utilize nonbasic groups in the P1 position. The work is based on our previously reported lead structure, compound 1, which was discovered via a resin-based approach to varying P1. By minimizing the size and lipophilicity of the P3 group and by incorporating hydrogen-bonding groups on the N-terminus or on the 2-position of the P1 aromatic ring, we have prepared a number of derivatives in this series that exhibit subnanomolar enzyme potency combined with good in vivo antithrombotic and bioavailability profiles. The oxyacetic amide compound 14b exhibited the best overall profile of in vitro and in vivo activity, and crystallographic studies indicate a unique mode of binding in the thrombin active site.


Assuntos
Cicloexilaminas/síntese química , Dipeptídeos/síntese química , Fibrinolíticos/síntese química , Trombina/antagonistas & inibidores , Administração Oral , Animais , Sítios de Ligação , Disponibilidade Biológica , Simulação por Computador , Cristalografia por Raios X , Cicloexilaminas/química , Cicloexilaminas/farmacocinética , Dipeptídeos/química , Dipeptídeos/farmacocinética , Cães , Desenho de Fármacos , Fibrinolíticos/química , Fibrinolíticos/farmacocinética , Fibrinolíticos/farmacologia , Ligação de Hidrogênio , Macaca fascicularis , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Ratos , Resinas Vegetais , Relação Estrutura-Atividade , Trombina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA