Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 336(2): 468-78, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21036913

ABSTRACT

Nitric oxide (NO) derived from neuronal nitric-oxide synthase (nNOS) and inducible nitric-oxide synthase (iNOS) plays a key role in various pain and inflammatory states. KLYP961 (4-((2-cyclobutyl-1H-imidazo[4,5-b]pyrazin-1-yl)methyl)-7,8-difluoroquinolin-2(1H)-one) inhibits the dimerization, and hence the enzymatic activity of human, primate, and murine iNOS and nNOS (IC(50) values 50-400 nM), with marked selectivity against endothelial nitric-oxide synthase (IC(50) >15,000 nM). It has ideal drug like-properties, including excellent rodent and primate pharmacokinetics coupled with a minimal off-target activity profile. In mice, KLYP961 attenuated endotoxin-evoked increases in plasma nitrates, a surrogate marker of iNOS activity in vivo, in a sustained manner (ED(50) 1 mg/kg p.o.). KLYP961 attenuated pain behaviors in a mouse formalin model (ED(50) 13 mg/kg p.o.), cold allodynia in the chronic constriction injury model (ED(50) 25 mg/kg p.o.), or tactile allodynia in the spinal nerve ligation model (ED(50) 30 mg/kg p.o.) with similar efficacy, but superior potency relative to gabapentin, pregabalin, or duloxetine. Unlike morphine, the antiallodynic activity of KLYP961 did not diminish upon repeated dosing. KLYP961 also attenuated carrageenin-induced edema and inflammatory hyperalgesia and writhing response elicited by phenylbenzoquinone with efficacy and potency similar to those of celecoxib. In contrast to gabapentin, KLYP961 did not impair motor coordination at doses as high as 1000 mg/kg p.o. KLYP961 also attenuated capsaicin-induced thermal allodynia in rhesus primates in a dose-related manner with a minimal effective dose (≤ 10 mg/kg p.o.) and a greater potency than gabapentin. In summary, KLYP961 represents an ideal tool with which to probe the physiological role of NO derived from iNOS and nNOS in human pain and inflammatory states.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Enzyme Inhibitors/pharmacology , Fluoroquinolones/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type I/antagonists & inhibitors , Pyrazines/pharmacology , Analgesics/pharmacology , Animals , Cells, Cultured , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/toxicity , Fluoroquinolones/pharmacokinetics , Fluoroquinolones/toxicity , Gastrointestinal Transit/drug effects , Humans , Macaca mulatta , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Motor Activity/drug effects , Protein Multimerization , Pyrazines/pharmacokinetics , Pyrazines/toxicity
2.
J Med Chem ; 53(21): 7739-55, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20931971

ABSTRACT

Three isoforms of nitric oxide synthase (NOS), dimeric enzymes that catalyze the formation of nitric oxide (NO) from arginine, have been identified. Inappropriate or excessive NO produced by iNOS and/or nNOS is associated with inflammatory and neuropathic pain. Previously, we described the identification of a series of amide-quinolinone iNOS dimerization inhibitors that although potent, suffered from high clearance and limited exposure in vivo. By conformationally restricting the amide of this progenitor series, we describe the identification of a novel series of benzimidazole-quinolinone dual iNOS/nNOS inhibitors with low clearance and sustained exposure in vivo. Compounds were triaged utilizing an LPS challenge assay coupled with mouse and rhesus pharmacokinetics and led to the identification of 4,7-imidazopyrazine 42 as the lead compound. 42 (KD7332) (J. Med. Chem. 2009, 52, 3047 - 3062) was confirmed as an iNOS dimerization inhibitor and was efficacious in the mouse formalin model of nociception and Chung model of neuropathic pain, without showing tolerance after repeat dosing. Further 42 did not affect motor coordination up to doses of 1000 mg/kg, demonstrating a wide therapeutic margin.


Subject(s)
Analgesics/chemical synthesis , Fluoroquinolones/chemical synthesis , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type I/antagonists & inhibitors , Pain/drug therapy , Pyrazines/chemical synthesis , Administration, Oral , Analgesics/chemistry , Analgesics/pharmacology , Animals , Cell Line , Drug Tolerance , Fluoroquinolones/chemistry , Fluoroquinolones/pharmacology , Humans , In Vitro Techniques , Mice , Microsomes, Liver/metabolism , Pain/etiology , Pain Measurement , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/etiology , Protein Multimerization , Pyrazines/chemistry , Pyrazines/pharmacology , Rotarod Performance Test , Structure-Activity Relationship
3.
Mol Pharmacol ; 76(1): 153-62, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19364813

ABSTRACT

Nitric-oxide synthases (NOS) generate nitric oxide (NO) through the oxidation of l-arginine. Inappropriate or excessive production of NO by NOS is associated with the pathophysiology of various disease states. Efforts to treat these disorders by developing arginine mimetic, substrate-competitive NOS inhibitors as drugs have met with little success. Small-molecule-mediated inhibition of NOS dimerization represents an intriguing alternative to substrate-competitive inhibition. An ultra-high-throughput cell-based screen of 880,000 small molecules identified a novel quinolinone with inducible NOS (iNOS) inhibitory activity. Exploratory chemistry based on this initial screening hit resulted in the synthesis of KLYP956, which inhibits iNOS at low nanomolar concentrations. The iNOS inhibitory potency of KLYP956 is insensitive to changes in concentrations of the substrate arginine, or the cofactor tetrahydrobiopterin. Mechanistic analysis suggests that KLYP956 binds the oxygenase domain in the vicinity of the active site heme and inhibits iNOS and neuronal NOS (nNOS) by preventing the formation of enzymatically active dimers. Oral administration of KLYP956 [N-(3-chlorophenyl)-N-((8-fluoro-2-oxo-1,2-dihydroquinolin-4-yl)methyl)-4-methylthiazole-5-carboxamide] inhibits iNOS activity in a murine model of endotoxemia and blocks pain behaviors in a formalin model of nociception. KLYP956 thus represents the first nonimidazole-based inhibitor of iNOS and nNOS dimerization and provides a novel pharmaceutical alternative to previously described substrate competitive inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Fluoroquinolones/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Thiazoles/pharmacology , Administration, Oral , Animals , Cells, Cultured , Dimerization , Humans , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide Synthase Type II/chemistry , Pain/drug therapy , Species Specificity
4.
J Med Chem ; 52(9): 3047-62, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19374401

ABSTRACT

There are three isoforms of dimeric nitric oxide synthases (NOS) that convert arginine to citrulline and nitric oxide. Inducible NOS is implicated in numerous inflammatory diseases and, more recently, in neuropathic pain states. The majority of existing NOS inhibitors are either based on the structure of arginine or are substrate competitive. We describe the identification from an ultra high-throughput screen of a novel series of quinolinone small molecule, nonarginine iNOS dimerization inhibitors. SAR studies on the screening hit, coupled with an in vivo lipopolysaccharide (LPS) challenge assay measuring plasma nitrates and drug levels, rapidly led to the identification of compounds 12 and 42--potent inhibitors of the human and mouse iNOS enzyme that were highly selective over endothelial NOS (eNOS). Following oral dosing, compounds 12 and 42 gave a statistical reduction in pain behaviors in the mouse formalin model, while 12 also statistically reduced neuropathic pain behaviors in the chronic constriction injury (Bennett) model.


Subject(s)
Drug Discovery , Fluoroquinolones/administration & dosage , Fluoroquinolones/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Pain/drug therapy , Protein Multimerization/drug effects , Pyrazines/administration & dosage , Pyrazines/pharmacology , Quinolones/administration & dosage , Quinolones/pharmacology , Administration, Oral , Animals , Cell Line , Constriction, Pathologic/chemically induced , Constriction, Pathologic/drug therapy , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Fluoroquinolones/chemistry , Fluoroquinolones/therapeutic use , Formaldehyde/toxicity , Humans , Inhibitory Concentration 50 , Lipopolysaccharides/toxicity , Mice , Nitric Oxide Synthase Type II/chemistry , Nitric Oxide Synthase Type II/metabolism , Protein Structure, Quaternary , Pyrazines/chemistry , Pyrazines/therapeutic use , Quinolones/chemistry , Quinolones/therapeutic use , Structure-Activity Relationship , Substrate Specificity
5.
Curr Chem Genomics ; 2: 1-9, 2008 Sep 27.
Article in English | MEDLINE | ID: mdl-20161838

ABSTRACT

The transcription of inducible nitric oxide synthase (iNOS) is activated by a network of proinflammatory signaling pathways. Here we describe the identification of a small molecule that downregulates the expression of iNOS mRNA and protein in cytokine-activated cells and suppresses nitric oxide production in vivo. Mechanistic analysis suggests that this small molecule, erstressin, also activates the unfolded protein response (UPR), a signaling pathway triggered by endoplasmic reticulum stress. Erstressin induces rapid phosphorylation of eIF2alpha and the alternative splicing of XBP-1, hallmark initiating events of the UPR. Further, erstressin activates the transcription of multiple genes involved in the UPR. These data suggest an inverse relationship between UPR activation and iNOS mRNA and protein expression under proinflammatory conditions.

6.
Biomed Microdevices ; 6(1): 11-21, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15307440

ABSTRACT

Optophoresis is a non-invasive cell analysis technique that is based on the interaction of live whole cells with optical gradient fields, typically generated by a near-infrared laser. The magnitude of the interaction depends upon the intrinsic physical properties of the cells, such as their refractive index, composition, size, and morphology. Time-of-flight (TOF) optophoresis is an implementation of this technique in a microfluidic environment. It measures cell travel times through a fixed distance with and without irradiation from a laser beam. The magnitude of the optical force from the laser, and therefore the change in transit time introduced by the presence of the infrared laser provides a signature for the cell. By accumulating such measurements for a population of cells (typically 200-300 cells per population), different cell types, drug treatments, or biological states can be compared quantitatively without the need for external labels or markers. An integrated TOF system has been constructed and characterized. The system typically uses square capillaries with 50-100 microm internal diameter and uses a syringe-pump-based flow system that generates initial bulk flow velocities between 200 and 600 microm/sec. Using this TOF technique, we have been able to consistently detect significant differences between normal skin and melanoma cell lines, CCD-1037 and A375, respectively. We have also been able to measure consistent differences in a cell differentiation model (HL60 cell line with DMSO treatment). These early results indicate the potential biological sensitivity of the TOF measurement technique for cellular analysis and cancer diagnostic applications.


Subject(s)
Cell Movement , Flow Cytometry/instrumentation , Microfluidic Analytical Techniques/instrumentation , Micromanipulation/instrumentation , Neoplasms/classification , Neoplasms/pathology , Physical Stimulation/instrumentation , Cell Line, Tumor , Equipment Design , Equipment Failure Analysis , Flow Cytometry/methods , Humans , Lasers , Microfluidic Analytical Techniques/methods , Micromanipulation/methods , Physical Stimulation/methods , Reproducibility of Results , Sensitivity and Specificity , Systems Integration
SELECTION OF CITATIONS
SEARCH DETAIL
...