Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
Mol Pharmacol ; 92(6): 694-706, 2017 12.
Article En | MEDLINE | ID: mdl-28974538

Bromodomain (BD) and extra-terminal domain containing proteins (BET) are chromatin adapters that bind acetylated histone marks via two tandem BDs, BD1 and BD2, to regulate gene transcription. BET proteins are involved in transcriptional reprogramming in response to inflammatory stimuli. BET BD inhibitors (BETis) that are nonselective for BD1 or BD2 have recognized anti-inflammatory properties in vitro and counter pathology in models of inflammation or autoimmune disease. Although both BD1 and BD2 bind acetylated histone residues, they may independently regulate the expression of BET-sensitive genes. Here we characterized the ability of RVX-297, a novel orally active BETi with selectivity for BD2, to modulate inflammatory processes in vitro, in vivo, and ex vivo. RVX-297 suppressed inflammatory gene expression in multiple immune cell types in culture. Mechanistically, RVX-297 displaced BET proteins from the promoters of sensitive genes and disrupted recruitment of active RNA polymerase II, a property shared with pan-BETis that nonselectively bind BET BDs. In the lipopolysaccharide model of inflammation, RVX-297 reduced proinflammatory mediators assessed in splenic gene expression and serum proteins. RVX-297 also countered pathology in three rodent models of polyarthritis: rat and mouse collagen-induced arthritis, and mouse collagen antibody-induced arthritis. Further, RVX-297 prevented murine experimental autoimmune encephalomyelitis (a model of human multiple sclerosis) disease development when administered prophylactically and reduced hallmarks of pathology when administered therapeutically. We show for the first time that a BD2-selective BETi maintains anti-inflammatory properties and is effective in preclinical models of acute inflammation and autoimmunity.


Anti-Inflammatory Agents/pharmacology , Arthritis/drug therapy , Autoimmune Diseases/drug therapy , Proteins/antagonists & inhibitors , Quinazolinones/therapeutic use , Acute Disease , Animals , Anti-Inflammatory Agents/therapeutic use , Antibodies/immunology , Arthritis/chemically induced , Arthritis/immunology , Arthritis/pathology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cells, Cultured , Collagen/immunology , Cytokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Male , Mice, Inbred C57BL , Rats, Inbred Lew , Spleen/drug effects , Spleen/metabolism , Spleen/pathology , U937 Cells
2.
Bioorg Med Chem Lett ; 25(14): 2818-23, 2015 Jul 15.
Article En | MEDLINE | ID: mdl-26022843

Bromodomains are key transcriptional regulators that are thought to be druggable epigenetic targets for cancer, inflammation, diabetes and cardiovascular therapeutics. Of particular importance is the first of two bromodomains in bromodomain containing 4 protein (BRD4(1)). Protein-ligand docking in BRD4(1) was used to purchase a small, focused screening set of compounds possessing a large variety of core structures. Within this set, a small number of weak hits each contained a dihydroquinoxalinone ring system. We purchased other analogs with this ring system and further validated the new hit series and obtained improvement in binding inhibition. Limited exploration by new analog synthesis showed that the binding inhibition in a FRET assay could be improved to the low µM level making this new core a potential hit-to-lead series. Additionally, the predicted geometries of the initial hit and an improved analog were confirmed by X-ray co-crystallography with BRD4(1).


Drug Design , Ligands , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Binding Sites , Cell Cycle Proteins , Crystallography, X-Ray , Drug Evaluation, Preclinical , Humans , Molecular Docking Simulation , Nuclear Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Quinoxalines/chemistry , Quinoxalines/metabolism , Structure-Activity Relationship , Transcription Factors/metabolism
3.
Atherosclerosis ; 236(1): 91-100, 2014 Sep.
Article En | MEDLINE | ID: mdl-25016363

Despite the benefit of statins in reducing cardiovascular risk, a sizable proportion of patients still remain at risk. Since HDL reduces CVD risk through a process that involves formation of pre-beta particles that facilitates the removal of cholesterol from the lipid-laden macrophages in the arteries, inducing pre-beta particles, may reduce the risk of CVD. A novel BET bromodomain antagonist, RVX-208, was reported to raise apoA-I and increase preß-HDL particles in non-human primates and humans. In the present study, we investigated the effect of RVX-208 on aortic lesion formation in hyperlipidemic apoE(-/-) mice. Oral treatments of apoE(-/-) mice with 150 mg/kg b.i.d RVX-208 for 12 weeks significantly reduced aortic lesion formation, accompanied by 2-fold increases in the levels of circulating HDL-C, and ∼50% decreases in LDL-C, although no significant changes in plasma apoA-I were observed. Circulating adhesion molecules as well as cytokines also showed significant reduction. Haptoglobin, a proinflammatory protein, known to bind with HDL/apoA-I, decreased >2.5-fold in the RVX-208 treated group. With a therapeutic dosing regimen in which mice were fed Western diet for 10 weeks to develop lesions followed by switching to a low fat diet and concurrent treatment with RVX-208 for 14 weeks, RVX-208 similarly reduced lesion formation by 39% in the whole aorta without significant changes in the plasma lipid parameters. RVX-208 significantly reduced the proinflammatory cytokines IP-10, MIP1(®) and MDC. These results show that the antiatherogenic activity of BET inhibitor, RVX-208, occurs via a combination of lipid changes and anti-inflammatory activities.


Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Hyperlipidemias/drug therapy , Quinazolines/therapeutic use , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/etiology , Aortic Diseases/pathology , Apolipoprotein A-I/blood , Apolipoproteins E/deficiency , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/pathology , Cell Line , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Cytokines/blood , Diet, Fat-Restricted , Diet, Western/adverse effects , Drug Evaluation, Preclinical , Endothelial Cells , Gene Expression Profiling , Humans , Hyperlipidemias/blood , Hyperlipidemias/complications , Hyperlipidemias/diet therapy , Hyperlipidemias/genetics , Inflammation/blood , Inflammation/prevention & control , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Quinazolines/pharmacology , Quinazolinones , RNA, Messenger/analysis , U937 Cells
4.
Eur J Med Chem ; 64: 121-8, 2013 Jun.
Article En | MEDLINE | ID: mdl-23644195

The structures of the two predominant metabolites (M4 and M5) of RVX-208, observed both in in vitro human and animal liver microsomal incubations, as well as in plasma from animal in vivo studies, were determined. A panel of biocatalytic systems was tested to identify biocatalysts suitable for milligram scale production of metabolite M4 from RVX-208. Rabbit liver S9 fraction was selected as the most suitable system, primarily based on pragmatic metrics such as catalyst cost and estimated yield of M4 (∼55%). Glucuronidation of RVX-208 catalyzed by rabbit liver S9 fraction was optimized to produce M4 in amounts sufficient for structural characterization. Structural studies using LC/MS/MS analysis and (1)H NMR spectroscopy showed the formation of a glycosidic bond between the primary hydroxyl group of RVX-208 and glucuronic acid. NMR results suggested that the glycosidic bond has the ß-anomeric configuration. A synthetic sample of M4 confirmed the proposed structure. Metabolite M5, hypothesized to be the carboxylate of RVX-208, was prepared using human liver microsomes, purified by HPLC, and characterized by LC/MS/MS and (1)H NMR. The structure was confirmed by comparison to a synthetic sample. Both samples confirmed M5 as a product of oxidation of primary hydroxyl group of RVX-208 to carboxylic acid.


Quinazolines/isolation & purification , Quinazolines/metabolism , Animals , Humans , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Quinazolines/chemistry , Quinazolinones , Rabbits
5.
PLoS One ; 8(12): e83190, 2013.
Article En | MEDLINE | ID: mdl-24391744

Increased synthesis of Apolipoprotein A-I (ApoA-I) and HDL is believed to provide a new approach to treating atherosclerosis through the stimulation of reverse cholesterol transport. RVX-208 increases the production of ApoA-I in hepatocytes in vitro, and in vivo in monkeys and humans, which results in increased HDL-C, but the molecular target was not previously reported. Using binding assays and X-ray crystallography, we now show that RVX-208 selectively binds to bromodomains of the BET (Bromodomain and Extra Terminal) family, competing for a site bound by the endogenous ligand, acetylated lysine, and that this accounts for its pharmacological activity. siRNA experiments further suggest that induction of ApoA-I mRNA is mediated by BET family member BRD4. These data indicate that RVX-208 increases ApoA-I production through an epigenetic mechanism and suggests that BET inhibition may be a promising new approach to the treatment of atherosclerosis.


Apolipoprotein A-I/biosynthesis , Nuclear Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Transcription Factors/antagonists & inhibitors , Animals , Apolipoprotein A-I/genetics , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Binding Sites , Cell Cycle Proteins , Cell Line , Crystallography, X-Ray , Epigenesis, Genetic/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Models, Molecular , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Quinazolines/chemistry , Quinazolinones , RNA, Small Interfering/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
6.
J Am Coll Cardiol ; 55(23): 2580-9, 2010 Jun 08.
Article En | MEDLINE | ID: mdl-20513599

OBJECTIVES: The aim of this study was to determine whether a novel small molecule RVX-208 affects apolipoprotein (apo)A-I and high-density lipoprotein cholesterol (HDL-C) levels in vitro and in vivo. BACKGROUND: Increased apoA-I and HDL-C levels are potential therapeutic targets for reducing atherosclerotic disease. METHODS: HepG2 cells were treated with 0 to 60 mumol/l RVX-208 followed by assays for apoA-I and HDL-C production. For in vivo studies, African green monkeys (AGMs) received 15 to 60 mg/kg/day RVX-208, and the serum was analyzed for lipoprotein levels, HDL-subparticle distribution, cholesterol efflux, and activity of lipid-modifying enzymes. A phase I clinical trial was conducted in healthy volunteers (given 1 to 20 mg/kg/day of RVX-208) to assess safety, tolerability, and pharmacokinetics. RESULTS: The RVX-208 induced apoA-I messenger ribonucleic acid and protein synthesis in HepG2 cells, leading to increased levels of pre-beta-migrating and alpha-lipoprotein particles containing apoA-I (LpA-I) in spent media. Similarly, in AGMs, RVX-208 treatment for 63 days increased serum apoA-I and HDL-C levels (60% and 97%, respectively). In addition, the levels of pre-beta(1)-LpA-I and alpha1-LpA-I HDL-subparticles were increased as well as adenosine triphosphate binding cassette AI, adenosine triphosphate binding cassette G1, and scavenger receptor class B type I-dependent cholesterol efflux. These changes were not mediated by cholesteryl-ester-transfer protein. Treatment of humans for 1 week with oral RVX-208 increased apoA-I, pre-beta-HDL, and HDL functionality. CONCLUSIONS: RVX-208 increases apoA-I and HDL-C in vitro and in vivo. In AGMs, RVX-208 raises serum pre-beta(1)-LpA-I and alpha-LpA-I levels and enhances cholesterol efflux. Data in humans point to beneficial features of RVX-208 that might be useful for treating atherosclerosis.


Apolipoprotein A-I/blood , Apolipoprotein A-I/drug effects , Cholesterol, HDL/blood , Cholesterol, HDL/drug effects , Quinazolines/pharmacology , Animals , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/metabolism , Cells, Cultured , Chlorocebus aethiops , Cholesterol, HDL/metabolism , Cricetinae , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Follow-Up Studies , Hep G2 Cells/drug effects , Hep G2 Cells/metabolism , Humans , In Vitro Techniques , Macaca fascicularis , Male , Molecular Weight , Probability , Quinazolines/chemistry , Quinazolinones , Random Allocation , Risk Assessment
...