Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 130(12): 6510-6522, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32853180

ABSTRACT

The sodium-phosphate cotransporter NPT2a plays a key role in the reabsorption of filtered phosphate in proximal renal tubules, thereby critically contributing to phosphate homeostasis. Inadequate urinary phosphate excretion can lead to severe hyperphosphatemia as in tumoral calcinosis and chronic kidney disease (CKD). Pharmacological inhibition of NPT2a may therefore represent an attractive approach for treating hyperphosphatemic conditions. The NPT2a-selective small-molecule inhibitor PF-06869206 was previously shown to reduce phosphate uptake in human proximal tubular cells in vitro. Here, we investigated the acute and chronic effects of the inhibitor in rodents and report that administration of PF-06869206 was well tolerated and elicited a dose-dependent increase in fractional phosphate excretion. This phosphaturic effect lowered plasma phosphate levels in WT mice and in rats with CKD due to subtotal nephrectomy. PF-06869206 had no effect on Npt2a-null mice, but promoted phosphate excretion and reduced phosphate levels in normophophatemic mice lacking Npt2c and in hyperphosphatemic mice lacking Fgf23 or Galnt3. In CKD rats, once-daily administration of PF-06869206 for 8 weeks induced an unabated acute phosphaturic and hypophosphatemic effect, but had no statistically significant effect on FGF23 or PTH levels. Selective pharmacological inhibition of NPT2a thus holds promise as a therapeutic option for genetic and acquired hyperphosphatemic disorders.


Subject(s)
Hyperphosphatemia/metabolism , Phosphates/metabolism , Renal Insufficiency, Chronic/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa , Animals , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hyperphosphatemia/genetics , Hyperphosphatemia/pathology , Male , Mice , Mice, Knockout , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Sodium-Phosphate Cotransporter Proteins, Type IIa/antagonists & inhibitors , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Polypeptide N-acetylgalactosaminyltransferase
2.
J Biol Chem ; 295(10): 3115-3133, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32005658

ABSTRACT

The fortuitously discovered antiaging membrane protein αKlotho (Klotho) is highly expressed in the kidney, and deletion of the Klotho gene in mice causes a phenotype strikingly similar to that of chronic kidney disease (CKD). Klotho functions as a co-receptor for fibroblast growth factor 23 (FGF23) signaling, whereas its shed extracellular domain, soluble Klotho (sKlotho), carrying glycosidase activity, is a humoral factor that regulates renal health. Low sKlotho in CKD is associated with disease progression, and sKlotho supplementation has emerged as a potential therapeutic strategy for managing CKD. Here, we explored the structure-function relationship and post-translational modifications of sKlotho variants to guide the future design of sKlotho-based therapeutics. Chinese hamster ovary (CHO)- and human embryonic kidney (HEK)-derived WT sKlotho proteins had varied activities in FGF23 co-receptor and ß-glucuronidase assays in vitro and distinct properties in vivo Sialidase treatment of heavily sialylated CHO-sKlotho increased its co-receptor activity 3-fold, yet it remained less active than hyposialylated HEK-sKlotho. MS and glycopeptide-mapping analyses revealed that HEK-sKlotho is uniquely modified with an unusual N-glycan structure consisting of N,N'-di-N-acetyllactose diamine at multiple N-linked sites, one of which at Asn-126 was adjacent to a putative GalNAc transfer motif. Site-directed mutagenesis and structural modeling analyses directly implicated N-glycans in Klotho's protein folding and function. Moreover, the introduction of two catalytic glutamate residues conserved across glycosidases into sKlotho enhanced its glucuronidase activity but decreased its FGF23 co-receptor activity, suggesting that these two functions might be structurally divergent. These findings open up opportunities for rational engineering of pharmacologically enhanced sKlotho therapeutics for managing kidney disease.


Subject(s)
Glucuronidase/metabolism , Renal Insufficiency, Chronic/pathology , Animals , CHO Cells , Catalytic Domain , Chromatography, High Pressure Liquid , Cricetinae , Cricetulus , Fibroblast Growth Factor-23 , Glomerular Filtration Rate/drug effects , Glucuronidase/chemistry , Glucuronidase/genetics , Glycopeptides/analysis , HEK293 Cells , Half-Life , Humans , Klotho Proteins , Mass Spectrometry , Mutagenesis, Site-Directed , Protein Processing, Post-Translational , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Renal Insufficiency, Chronic/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Reperfusion Injury/veterinary , Structure-Activity Relationship
3.
ACS Med Chem Lett ; 9(5): 440-445, 2018 May 10.
Article in English | MEDLINE | ID: mdl-29795756

ABSTRACT

Sodium-phosphate cotransporter 2a, or NaPi2a (SLC34A1), is a solute-carrier (SLC) transporter located in the kidney proximal tubule that reabsorbs glomerular-filtered phosphate. Inhibition of NaPi2a may enhance urinary phosphate excretion and correct maladaptive mineral and hormonal derangements associated with increased cardiovascular risk in chronic kidney disease-mineral and bone disorder (CKD-MBD). To date, only nonselective NaPi inhibitors have been described. Herein, we detail the discovery of the first series of selective NaPi2a inhibitors, resulting from optimization of a high-throughput screening hit. The oral PK profile of inhibitor PF-06869206 (6f) in rodents allows for the exploration of the pharmacology of selective NaPi2a inhibition.

4.
J Med Chem ; 61(8): 3685-3696, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29627981

ABSTRACT

C-X-C chemokine receptor type 7 (CXCR7) is involved in cardiac and immune pathophysiology. We report the discovery of a novel 1,4-diazepine CXCR7 modulator, demonstrating for the first time the role of pharmacological CXCR7 intervention in cardiac repair. Structure-activity-relationship (SAR) studies demonstrated that a net reduction in lipophilicity (log D) and an incorporation of saturated ring systems yielded compounds with good CXCR7 potencies and improvements in oxidative metabolic stability in human-liver microsomes (HLM). Tethering an ethylene amide further improved the selectivity profile (e.g., for compound 18, CXCR7 Ki = 13 nM, adrenergic α 1a Kb > 10 000 nM, and adrenergic ß 2 Kb > 10 000 nM). The subcutaneous administration of 18 in mice led to a statistically significant increase in circulating concentrations of plasma stromal-cell-derived factor 1α (SDF-1α) of approximately 2-fold. Chronic dosing of compound 18 in a mouse model of isoproterenol-induced cardiac injury further resulted in a statistically significant reduction of cardiac fibrosis.


Subject(s)
Acetamides/therapeutic use , Azepines/therapeutic use , Cardiotonic Agents/therapeutic use , Fibrosis/drug therapy , Heart Diseases/drug therapy , Receptors, CXCR/metabolism , Acetamides/chemical synthesis , Acetamides/chemistry , Acetamides/pharmacology , Animals , Azepines/chemical synthesis , Azepines/chemistry , Azepines/pharmacology , Cardiotonic Agents/chemical synthesis , Cardiotonic Agents/chemistry , Cardiotonic Agents/pharmacology , Dogs , Fibrosis/chemically induced , Heart Diseases/chemically induced , Humans , Hydrophobic and Hydrophilic Interactions , Isoproterenol , Madin Darby Canine Kidney Cells , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Molecular Structure , Structure-Activity Relationship
5.
J Am Soc Nephrol ; 27(11): 3459-3468, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27113485

ABSTRACT

Diabetic nephropathy (DN) is the leading cause of ESRD worldwide. Reduced bioavailability or uncoupling of nitric oxide in the kidney, leading to decreased intracellular levels of the nitric oxide pathway effector molecule cyclic guanosine monophosphate (cGMP), has been implicated in the progression of DN. Preclinical studies suggest that elevating the cGMP intracellular pool through inhibition of the cGMP-hydrolyzing enzyme phosphodiesterase type 5 (PDE5) might exert renoprotective effects in DN. To test this hypothesis, the novel, highly specific, and long-acting PDE5 inhibitor, PF-00489791, was assessed in a multinational, multicenter, randomized, double-blind, placebo-controlled, parallel group trial of subjects with type 2 diabetes mellitus and overt nephropathy receiving angiotensin converting enzyme inhibitor or angiotensin receptor blocker background therapy. In total, 256 subjects with an eGFR between 25 and 60 ml/min per 1.73 m2 and macroalbuminuria defined by a urinary albumin-to-creatinine ratio >300 mg/g, were randomly assigned 3:1, respectively, to receive PF-00489791 (20 mg) or placebo orally, once daily for 12 weeks. Using the predefined primary assessment of efficacy (Bayesian analysis with informative prior), we observed a significant reduction in urinary albumin-to-creatinine ratio of 15.7% (ratio 0.843; 95% credible interval 0.73 to 0.98) in response to the 12-week treatment with PF-00489791 compared with placebo. PF-00489791 was safe and generally well tolerated in this patient population. Most common adverse events were mild in severity and included headache and upper gastrointestinal events. In conclusion, the safety and efficacy profile of PDE5 inhibitor PF-00489791 supports further investigation as a novel therapy to improve renal outcomes in DN.


Subject(s)
Albuminuria/drug therapy , Albuminuria/etiology , Diabetic Nephropathies/complications , Heterocyclic Compounds, 2-Ring/therapeutic use , Phosphodiesterase 5 Inhibitors/therapeutic use , Sulfonamides/therapeutic use , Adult , Aged , Aged, 80 and over , Albuminuria/enzymology , Double-Blind Method , Female , Humans , Male , Middle Aged
6.
Nat Biotechnol ; 29(11): 1028-33, 2011 Oct 23.
Article in English | MEDLINE | ID: mdl-22020385

ABSTRACT

Effective therapies are needed to control excessive bleeding in a range of clinical conditions. We improve hemostasis in vivo using a conformationally pliant variant of coagulation factor Xa (FXa(I16L)) rendered partially inactive by a defect in the transition from zymogen to active protease. Using mouse models of hemophilia, we show that FXa(I16L) has a longer half-life than wild-type FXa and does not cause excessive activation of coagulation. Once clotting mechanisms are activated to produce its cofactor FVa, FXa(I16L) is driven to the protease state and restores hemostasis in hemophilic animals upon vascular injury. Moreover, using human or murine analogs, we show that FXa(I16L) is more efficacious than FVIIa, which is used to treat bleeding in hemophilia inhibitor patients. FXa(I16L) may provide an effective strategy to enhance blood clot formation and act as a rapid pan-hemostatic agent for the treatment of bleeding conditions.


Subject(s)
Enzyme Precursors/therapeutic use , Factor Xa/therapeutic use , Hemophilia A/drug therapy , Hemostatics/therapeutic use , Animals , Blood Coagulation/genetics , Disease Models, Animal , Enzyme Precursors/pharmacokinetics , Factor VIIa/genetics , Factor VIIa/metabolism , Factor Xa/pharmacokinetics , Gene Expression , HEK293 Cells , Hemorrhage/drug therapy , Hemostasis/genetics , Hemostatics/pharmacokinetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Thrombelastography , Thrombin/metabolism
7.
J Med Chem ; 53(16): 6003-17, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20718494

ABSTRACT

Previously, we reported the discovery of PSI-697 (1a), a C-2 benzyl substituted quinoline salicylic acid-based P-selectin inhibitor. It is active in a variety of animal models of cardiovascular disease. Compound 1a has also been shown to be well tolerated and safe in healthy volunteers at doses of up to 1200 mg in a phase 1 single ascending dose study. However, its oral bioavailability was low. Our goal was to identify a back up compound with equal potency, increased solubility, and increased exposure. We expanded our structure-activity studies in this series by branching at the alpha position of the C-2 benzyl side chain and through modification of substituents on the carboxylic A-ring of the quinoline. This resulted in discovery of PSI-421 with marked improvement in aqueous solubility and pharmacokinetic properties. This compound has shown oral efficacy in animal models of arterial and venous injury and was selected as a preclinical development compound for potential treatment of such diseases as atherosclerosis and deep vein thrombosis.


Subject(s)
Carotid Artery Injuries/drug therapy , Hydroxyquinolines/chemical synthesis , P-Selectin/antagonists & inhibitors , Salicylates/chemical synthesis , Venous Thrombosis/drug therapy , Administration, Oral , Animals , Caco-2 Cells , Cell Membrane Permeability , Dogs , Drug Stability , Humans , Hydroxyquinolines/pharmacokinetics , Hydroxyquinolines/pharmacology , Leukocyte Rolling/drug effects , Macaca fascicularis , Mice , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Models, Molecular , Papio , Rats , Rats, Sprague-Dawley , Salicylates/chemistry , Salicylates/pharmacology , Solubility , Structure-Activity Relationship
8.
J Lipid Res ; 50(12): 2358-70, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19318684

ABSTRACT

Liver X receptors (LXRs) are ligand-activated transcription factors that coordinate regulation of gene expression involved in several cellular functions but most notably cholesterol homeostasis encompassing cholesterol transport, catabolism, and absorption. WAY-252623 (LXR-623) is a highly selective and orally bioavailable synthetic modulator of LXR, which demonstrated efficacy for reducing lesion progression in the murine LDLR(-/-) atherosclerosis model with no associated increase in hepatic lipogenesis either in this model or Syrian hamsters. In nonhuman primates with normal lipid levels, WAY-252623 significantly reduced total (50-55%) and LDL-cholesterol (LDLc) (70-77%) in a time- and dose-dependent manner as well as increased expression of the target genes ABCA1/G1 in peripheral blood cells. Statistically significant decreases in LDLc were noted as early as day 7, reached a maximum by day 28, and exceeded reductions observed for simvastatin alone (20 mg/kg). Transient increases in circulating triglycerides and liver enzymes reverted to baseline levels over the course of the study. Complementary microarray analysis of duodenum and liver gene expression revealed differential activation of LXR target genes and suggested no direct activation of hepatic lipogenesis. WAY-252623 displays a unique and favorable pharmacological profile suggesting synthetic LXR ligands with these characteristics may be suitable for evaluation in patients with atherosclerotic dyslipidemia.


Subject(s)
Atherosclerosis/drug therapy , Cholesterol, LDL/drug effects , Cholesterol, LDL/metabolism , Indazoles/pharmacology , Lipid Metabolism/drug effects , Macaca fascicularis/metabolism , Orphan Nuclear Receptors/agonists , Animals , Atherosclerosis/metabolism , Caco-2 Cells , Cricetinae , Disease Models, Animal , Humans , Indazoles/blood , Indazoles/chemistry , Ligands , Liver/enzymology , Liver/metabolism , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors/metabolism
9.
J Med Chem ; 51(22): 7161-8, 2008 Nov 27.
Article in English | MEDLINE | ID: mdl-18973288

ABSTRACT

A series of substituted 2-benzyl-3-aryl-7-trifluoromethylindazoles were prepared as LXR modulators. These compounds were partial agonists in transactivation assays when compared to 1 (T0901317) and were slightly weaker with respect to potency and efficacy on LXRalpha than on LXRbeta. Lead compounds in this series 12 (WAY-252623) and 13 (WAY-214950) showed less lipid accumulation in HepG2 cells than potent full agonists 1 and 3 (WAY-254011) but were comparable in efficacy to 1 and 3 with respect to cholesterol efflux in THP-1 foam cells, albeit weaker in potency. Compound 13 reduced aortic lesion area in LDLR knockout mice equivalently to 3 or positive control 2 (GW3965). In a 7-day hamster model, compound 13 showed a lesser propensity for plasma TG elevation than 3, when the compounds were compared at doses in which they elevated ABCA1 and ABCG1 gene expression in duodenum and liver at equal levels. In contrast to results previously published for 2, the lack of TG effect of 13 correlated with its inability to increase liver fatty acid synthase (FAS) gene expression, which was up-regulated 4-fold by 3. These results suggest indazoles such as 13 may have an improved profile for potential use as a therapeutic agent.


Subject(s)
Arteriosclerosis/drug therapy , DNA-Binding Proteins/agonists , Indazoles/pharmacology , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Triglycerides/biosynthesis , Animals , Arteriosclerosis/metabolism , Cell Differentiation/drug effects , Cell Line , Cricetinae , Crystallography, X-Ray , DNA-Binding Proteins/metabolism , Humans , Hydrogen Bonding , Indazoles/chemical synthesis , Indazoles/chemistry , Ligands , Liver/drug effects , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Models, Molecular , Molecular Structure , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Structure-Activity Relationship , Triglycerides/blood
10.
Atherosclerosis ; 201(1): 53-66, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18377911

ABSTRACT

OBJECTIVE: The present study was conducted to characterize the expression of the cysteine protease legumain in murine and human atherosclerotic tissues, and to explore the molecular mechanisms by which legumain may contribute to the pathophysiology of atherosclerosis. METHODS AND RESULTS: Using microarray analysis, legumain mRNA expression was found to increase with development of atherosclerosis in the aorta of aging Apolipoprotein E deficient mice while expression remained at low level and unchanged in arteries of age-matched C57BL/6 control mice. In situ hybridization and immunohistochemical analysis determined that legumain was predominantly expressed by macrophages in the atherosclerotic aorta, in lesions at the aortic sinus and in injured carotid arteries of Apolipoprotein E deficient mice as well as in inflamed areas in advanced human coronary atherosclerotic plaques. In vitro, M-CSF differentiated human primary macrophages were shown to express legumain and the protein could also be detected in the culture media. When tested in migration assays, legumain induced chemotaxis of primary human monocytes and human umbilical vein endothelial cells. CONCLUSIONS: Legumain is expressed in both murine and human atherosclerotic lesions. The macrophage-specific expression of legumain in vivo and ability of legumain to induce chemotaxis of monocytes and endothelial cells in vitro suggest that legumain may play a functional role in atherogenesis.


Subject(s)
Aortic Diseases/enzymology , Aortic Diseases/etiology , Atherosclerosis/enzymology , Atherosclerosis/etiology , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Age Factors , Animals , Aortic Diseases/physiopathology , Apolipoproteins E/physiology , Atherosclerosis/physiopathology , Disease Models, Animal , Endothelial Cells/physiology , Female , Humans , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Monocytes/physiology , RNA, Messenger/metabolism
11.
J Pharmacol Exp Ther ; 324(2): 497-506, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18024786

ABSTRACT

P-selectin plays a significant and well documented role in vascular disease by mediating leukocyte and platelet rolling and adhesion. This study characterizes the in vitro activity, pharmacokinetic properties, and the anti-inflammatory and antithrombotic efficacy of the orally active P-selectin small-molecule antagonist PSI-697 [2-(4-chlorobenzyl)-3-hydroxy-7,8,9,10-tetrahydrobenzo[h] quinoline-4-carboxylic acid; molecular mass, 367.83]. Biacore and cell-based assays were used to demonstrate the ability of PSI-697 to dose dependently inhibit the binding of human P-selectin to human P-selectin glycoprotein ligand-1, inhibiting 50% of binding at 50 to 125 microM. The pharmacokinetics of PSI-697 in rats were characterized by low clearance, short half-life, low volume of distribution, and moderate apparent oral bioavailability. A surgical inflammation model, using exteriorized rat cremaster venules, demonstrated that PSI-697 (50 mg/kg p.o.) significantly reduced the number of rolling leukocytes by 39% (P < 0.05) versus vehicle control. In a rat venous thrombosis model, PSI-697 (100 mg/kg p.o.) reduced thrombus weight by 18% (P < 0.05) relative to vehicle, without prolonging bleeding time. Finally, in a rat carotid injury model, PSI-697 (30 or 15 mg/kg p.o.) administered 1 h before arterial injury and once daily thereafter for 13 days resulted in dose-dependent decreases in intima/media ratios of 40.2% (P = 0.025) and 25.7% (P = 0.002) compared with vehicle controls. These data demonstrate the activity of PSI-697 in vitro and after oral administration in animal models of both arterial and venous injury and support the clinical evaluation of this novel antagonist of P-selectin in atherothrombotic and venous thrombotic indications.


Subject(s)
Disease Models, Animal , Hydroxyquinolines/therapeutic use , P-Selectin , Vasculitis/drug therapy , Venous Thrombosis/drug therapy , Animals , HL-60 Cells , Humans , Hydroxyquinolines/chemistry , Hydroxyquinolines/pharmacology , Male , P-Selectin/metabolism , Protein Binding/drug effects , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Vasculitis/metabolism , Venous Thrombosis/metabolism
12.
Bioorg Med Chem ; 15(10): 3321-33, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17391964

ABSTRACT

A series of phenyl acetic acid based quinolines was prepared as LXR modulators. An SAR study in which the C-3 and C-8 positions of the quinoline core were varied led to the identification of two potent LXR agonists 23 and 27. Both compounds displayed good binding affinity for LXRbeta and LXRalpha, and increased expression of ABCA1 in THP-1 cells. These two compounds also had desirable pharmacokinetic profiles in mice and displayed in vivo efficacy in a 12-week Apo E knockout mouse lesion model.


Subject(s)
Atherosclerosis/prevention & control , DNA-Binding Proteins/agonists , Phenylacetates/chemical synthesis , Phenylacetates/pharmacology , Quinolines/chemical synthesis , Quinolines/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , CHO Cells , Carboxylic Acids/chemical synthesis , Carboxylic Acids/pharmacology , Chromatography, High Pressure Liquid , Cricetinae , Cricetulus , DNA-Binding Proteins/genetics , Humans , Indicators and Reagents , Liver X Receptors , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Proteins/metabolism , Solvents , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship , Transcriptional Activation/genetics
13.
J Med Chem ; 50(1): 40-64, 2007 Jan 11.
Article in English | MEDLINE | ID: mdl-17201409

ABSTRACT

P-selectin-PSGL-1 interaction causes rolling of leukocytes on the endothelial cell surface, which subsequently leads to firm adherence and leukocyte transmigration through the vessel wall into the surrounding tissues. P-selectin is upregulated on the surface of both platelets and endothelium in a variety of atherosclerosis-associated conditions. Consequently, inhibition of this interaction by means of a small molecule P-selectin antagonist is an attractive strategy for the treatment of atherosclerosis. High-throughput screening and subsequent analoging had led to the identification of compound 1 as the lead candidate. Herein, we report the continuation of this work and the discovery of a second-generation series, the tetrahydrobenzoquinoline salicylic acids. These compounds have improved pharmacokinetic properties, and a number of them have shown oral efficacy in mouse and rat models of atherogenesis and vascular injury. The lead 31 (PSI-697), is currently in clinical development for the treatment of atherothrombotic vascular events.


Subject(s)
Atherosclerosis/prevention & control , Fibrinolytic Agents/chemical synthesis , Hydroxyquinolines/chemical synthesis , P-Selectin/metabolism , Quinolines/chemical synthesis , Salicylates/chemical synthesis , Administration, Oral , Animals , Apolipoproteins E/genetics , Carotid Stenosis/prevention & control , Dogs , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacology , Hydroxyquinolines/pharmacokinetics , Hydroxyquinolines/pharmacology , Indoles/chemistry , Indoles/pharmacokinetics , Indoles/pharmacology , Leukocyte Rolling/drug effects , Male , Mice , Mice, Knockout , Quinolines/pharmacokinetics , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Salicylates/pharmacokinetics , Salicylates/pharmacology , Structure-Activity Relationship
14.
J Med Chem ; 49(21): 6151-4, 2006 Oct 19.
Article in English | MEDLINE | ID: mdl-17034119

ABSTRACT

A structure-based approach was used to optimize our new class of quinoline LXR modulators leading to phenyl acetic acid substituted quinolines 15 and 16. Both compounds displayed good binding affinity for LXRbeta and LXRalpha and were potent activators in LBD transactivation assays. The compounds also increased expression of ABCA1 and stimulated cholesterol efflux in THP-1 cells. Quinoline 16 showed good oral bioavailability and in vivo efficacy in a LDLr knockout mouse model for lesions.


Subject(s)
Anticholesteremic Agents/chemical synthesis , Atherosclerosis/drug therapy , DNA-Binding Proteins/agonists , Phenylacetates/chemical synthesis , Quinolines/chemical synthesis , Receptors, Cytoplasmic and Nuclear/agonists , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/biosynthesis , Animals , Anticholesteremic Agents/chemistry , Anticholesteremic Agents/pharmacology , Binding Sites , Biological Availability , Cell Line , Cholesterol/metabolism , DNA-Binding Proteins/genetics , Drug Stability , Female , Humans , In Vitro Techniques , Ligands , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Orphan Nuclear Receptors , Phenylacetates/chemistry , Phenylacetates/pharmacology , Protein Structure, Tertiary , Quinolines/chemistry , Quinolines/pharmacology , Receptors, Cytoplasmic and Nuclear/genetics , Structure-Activity Relationship , Transcriptional Activation
15.
Tissue Eng ; 9(3): 461-72, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12857414

ABSTRACT

Traditional approaches to generating tissue-engineered arteries in vitro rely on expansion of cells in culture to seed appropriate scaffolds. In most envisioned applications, small autologous blood vessels would be harvested and used as a source for these cells. We propose that small autologous arteries, not the cells derived from them, may be an attractive starting point for engineered arteries. This approach capitalizes on the ability of intact arteries to grow and remodel in response to chronic changes in their mechanical environment. Carotid arteries from juvenile (approximately 30-kg) pigs were stretched longitudinally in an ex vivo perfusion system over 9 days. This resulted in a 40% increase in artery length at physiological longitudinal stress and a 20 +/- 3% increase when unstressed. Control arteries were perfused for 9 days ex vivo at their physiological loaded length. Control and elongated arteries displayed native appearance (macroscopic and histological), excellent viability (cellularity and mitochondrial activity), normal vasoactivity, and similar mechanical properties (ultimate stress and ultimate strain) as compared with freshly harvested arteries. Growth, as opposed to just redistribution of existing mass, contributed to elongation as evidenced by an increase in artery weight. Results on elongation of arteries from neonatal and adolescent pigs are also presented and discussed.


Subject(s)
Carotid Arteries , Tissue Engineering/methods , Animals , Arteries , Perfusion/instrumentation , Perfusion/methods , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...