Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
6.
Bioorg Med Chem Lett ; 25(21): 4812-4819, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26195137

ABSTRACT

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.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Hydantoins/chemistry , Hydantoins/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Hydantoins/chemical synthesis , Methylation , Models, Molecular , Molecular Structure , Structure-Activity Relationship
7.
J Phys Condens Matter ; 23(8): 086005, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21411908

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-18413497

ABSTRACT

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.


Subject(s)
Absorbable Implants , Carotid Stenosis/prevention & control , Carotid Stenosis/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Stents , Adenoviridae/genetics , Animals , Aorta/cytology , Cells, Cultured , Cross-Linking Reagents , Green Fluorescent Proteins/genetics , Male , Metals , Muscle, Smooth, Vascular/cytology , Neutralization Tests , Nitric Oxide Synthase Type II/genetics , Rats , Rats, Sprague-Dawley , Secondary Prevention
9.
Xenobiotica ; 33(6): 643-54, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12851040

ABSTRACT

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.


Subject(s)
Benzofurans/pharmacology , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/pharmacokinetics , Indinavir/analogs & derivatives , Indinavir/pharmacology , Piperazines/pharmacology , Animals , Antibodies, Blocking/pharmacology , Area Under Curve , Benzofurans/antagonists & inhibitors , Benzofurans/pharmacokinetics , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System , Dogs , Drug Synergism , Enzyme Inhibitors/pharmacology , HIV Protease Inhibitors/antagonists & inhibitors , Indinavir/antagonists & inhibitors , Indinavir/pharmacokinetics , Injections, Intravenous , Macaca mulatta , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Piperazines/antagonists & inhibitors , Piperazines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Species Specificity , Spectrophotometry, Ultraviolet
10.
Drug Metab Dispos ; 29(12): 1578-87, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11717177

ABSTRACT

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.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Piperazines/pharmacology , Algorithms , Animals , Area Under Curve , Bile/metabolism , Bile Ducts/metabolism , Biotransformation , Blood Proteins/metabolism , Chromatography, High Pressure Liquid , Dogs , Farnesyltranstransferase , Half-Life , Humans , In Vitro Techniques , Intestinal Absorption , Male , Mass Spectrometry , Microsomes, Liver , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL