Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-29226625

ABSTRACT

Therapies that suppress RIPK1 kinase activity are emerging as promising therapeutic agents for the treatment of multiple inflammatory disorders. The ability to directly measure drug binding of a RIPK1 inhibitor to its target is critical for providing insight into pharmacokinetics, pharmacodynamics, safety and clinical efficacy, especially for a first-in-class small-molecule inhibitor where the mechanism has yet to be explored. Here, we report a novel method for measuring drug binding to RIPK1 protein in cells and tissues. This TEAR1 (Target Engagement Assessment for RIPK1) assay is a pair of immunoassays developed on the principle of competition, whereby a first molecule (ie, drug) prevents the binding of a second molecule (ie, antibody) to the target protein. Using the TEAR1 assay, we have validated the direct binding of specific RIPK1 inhibitors in cells, blood and tissues following treatment with benzoxazepinone (BOAz) RIPK1 inhibitors. The TEAR1 assay is a valuable tool for facilitating the clinical development of the lead RIPK1 clinical candidate compound, GSK2982772, as a first-in-class RIPK1 inhibitor for the treatment of inflammatory disease.


Subject(s)
Antibodies/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , HT29 Cells , Humans , Immunoassay , Macaca fascicularis , Male , Protein Binding/drug effects , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Small Molecule Libraries/pharmacology
2.
PLoS One ; 11(2): e0147979, 2016.
Article in English | MEDLINE | ID: mdl-26870941

ABSTRACT

BACKGROUND: Psoriasis is a chronic inflammatory skin disorder involving marked immunological changes. IL-17-targeting biologics have been successful in reducing the disease burden of psoriasis patients with moderate-to-severe disease. Unfortunately, the stratum corneum prevents penetration of large molecule weight proteins, including monoclonal antibodies. Thus, for the majority of psoriasis patients ineligible for systemic treatments, a small molecule targeting RORγt, the master regulator of IL-17 family cytokines, may represent an alternative topical medicine with biologic-like efficacy. METHODS AND FINDINGS: The preclinical studies described in this manuscript bridge the gap from bench to bedside to provide the scientific foundation for a compound entering clinical trials for patients with mild to moderate psoriasis. In addition to several ex vivo reporter assays, primary T cell cultures, and the imiquimod mouse model, we demonstrate efficacy in a newly developed human ex vivo skin assay, where Th17-skewed cytokine expression is induced from skin-resident immune cells. Importantly, the skin barrier remains intact allowing for the demonstration of topical drug delivery. With the development of this novel assay, we demonstrate potent compound activity in the target tissue: human skin. Finally, target engagement by this small molecule was confirmed in ex vivo lesional psoriatic skin. CONCLUSIONS: Our work describes a progressive series of assays to demonstrate the potential clinical value of a novel RORγ inverse agonist small molecule with high potency and selectivity, which will enter clinical trials in late 2015 for psoriasis patients.


Subject(s)
Immunologic Factors/pharmacology , Interleukin-17/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Psoriasis/drug therapy , Skin/drug effects , Small Molecule Libraries/pharmacology , Administration, Cutaneous , Aminoquinolines , Animals , Drug Evaluation, Preclinical , Female , Gene Expression , Genes, Reporter , Humans , Imiquimod , Immunologic Factors/chemical synthesis , Interleukin-17/genetics , Interleukin-17/immunology , Jurkat Cells , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Permeability , Primary Cell Culture , Psoriasis/chemically induced , Psoriasis/immunology , Psoriasis/pathology , Skin/immunology , Skin/pathology , Small Molecule Libraries/chemical synthesis , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/pathology , Translational Research, Biomedical
3.
Cell Rep ; 11(9): 1446-57, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26004183

ABSTRACT

AMPK is a master regulator of cellular metabolism that exerts either oncogenic or tumor suppressor activity depending on context. Here, we report that the specific AMPK agonist GSK621 selectively kills acute myeloid leukemia (AML) cells but spares normal hematopoietic progenitors. This differential sensitivity results from a unique synthetic lethal interaction involving concurrent activation of AMPK and mTORC1. Strikingly, the lethality of GSK621 in primary AML cells and AML cell lines is abrogated by chemical or genetic ablation of mTORC1 signaling. The same synthetic lethality between AMPK and mTORC1 activation is established in CD34-positive hematopoietic progenitors by constitutive activation of AKT or enhanced in AML cells by deletion of TSC2. Finally, cytotoxicity in AML cells from GSK621 involves the eIF2α/ATF4 signaling pathway that specifically results from mTORC1 activation. AMPK activation may represent a therapeutic opportunity in mTORC1-overactivated cancers.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Enzyme Activation/drug effects , Imidazoles/pharmacology , Leukemia, Myeloid, Acute/metabolism , Multiprotein Complexes/agonists , Pyrimidinones/pharmacology , Animals , Fluorescent Antibody Technique , Heterografts , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Nude , Microscopy, Electron, Transmission , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA Interference , Signal Transduction/drug effects , TOR Serine-Threonine Kinases
4.
J Med Chem ; 57(19): 8111-31, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25249180

ABSTRACT

Through their function as epigenetic readers of the histone code, the BET family of bromodomain-containing proteins regulate expression of multiple genes of therapeutic relevance, including those involved in tumor cell growth and inflammation. BET bromodomain inhibitors have profound antiproliferative and anti-inflammatory effects which translate into efficacy in oncology and inflammation models, and the first compounds have now progressed into clinical trials. The exciting biology of the BETs has led to great interest in the discovery of novel inhibitor classes. Here we describe the identification of a novel tetrahydroquinoline series through up-regulation of apolipoprotein A1 and the optimization into potent compounds active in murine models of septic shock and neuroblastoma. At the molecular level, these effects are produced by inhibition of BET bromodomains. X-ray crystallography reveals the interactions explaining the structure-activity relationships of binding. The resulting lead molecule, I-BET726, represents a new, potent, and selective class of tetrahydroquinoline-based BET inhibitors.


Subject(s)
Aminoquinolines/chemical synthesis , Anti-Inflammatory Agents/chemical synthesis , Apolipoprotein A-I/metabolism , Benzoates/chemical synthesis , Nuclear Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinolines/chemical synthesis , Transcription Factors/antagonists & inhibitors , Aminoquinolines/pharmacokinetics , Aminoquinolines/pharmacology , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Benzoates/pharmacokinetics , Benzoates/pharmacology , Cell Cycle Proteins , Drug Discovery , Humans , Mice , Quinolines/pharmacokinetics , Quinolines/pharmacology , Structure-Activity Relationship
5.
J Med Chem ; 56(19): 7501-15, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24015967

ABSTRACT

The bromo and extra C-terminal domain (BET) family of bromodomains are involved in binding epigenetic marks on histone proteins, more specifically acetylated lysine residues. This paper describes the discovery and structure-activity relationships (SAR) of potent benzodiazepine inhibitors that disrupt the function of the BET family of bromodomains (BRD2, BRD3, and BRD4). This work has yielded a potent, selective compound I-BET762 that is now under evaluation in a phase I/II clinical trial for nuclear protein in testis (NUT) midline carcinoma and other cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Benzodiazepines/pharmacology , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Apolipoprotein A-I/biosynthesis , Benzodiazepines/chemical synthesis , Benzodiazepines/pharmacokinetics , Cell Cycle Proteins , Dogs , Epigenesis, Genetic , Humans , Macaca fascicularis , Mice , Models, Molecular , Permeability , Protein Structure, Tertiary , Rats , Stereoisomerism , Structure-Activity Relationship
6.
ACS Med Chem Lett ; 4(7): 632-6, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-24900722

ABSTRACT

AMP-activated protein kinase (AMPK) is an evolutionarily conserved fuel-sensing enzyme that is activated in shortage of energy and suppressed in its surfeit. AMPK activation stimulates fatty acid oxidation, enhances insulin sensitivity, alleviates hyperglycemia and hyperlipidemia, and inhibits proinflammatory changes. Thus, AMPK is a well-received therapeutic target for type 2 diabetes and other metabolic disorders. Here, we will report the discovery of pyrrolopyridone derivatives as AMPK direct activators. We will illustrate the synthesis and structure-activity relationships of the series as well as some pharmacokinetic results. Some compounds exhibited encouraging oral exposure and were evaluated in a mouse diabetic model. Compound 17 showed oral activity at 30 mg/kg on blood glucose.

8.
J Med Chem ; 54(11): 3827-38, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21568322

ABSTRACT

Epigenetic mechanisms of gene regulation have a profound role in normal development and disease processes. An integral part of this mechanism occurs through lysine acetylation of histone tails which are recognized by bromodomains. While the biological and structural characterization of many bromodomain containing proteins has advanced considerably, the therapeutic tractability of this protein family is only now becoming understood. This paper describes the discovery and molecular characterization of potent (nM) small molecule inhibitors that disrupt the function of the BET family of bromodomains (Brd2, Brd3, and Brd4). By using a combination of phenotypic screening, chemoproteomics, and biophysical studies, we have discovered that the protein-protein interactions between bromodomains and acetylated histones can be antagonized by selective small molecules that bind at the acetylated lysine recognition pocket. X-ray crystal structures of compounds bound into bromodomains of Brd2 and Brd4 elucidate the molecular interactions of binding and explain the precisely defined stereochemistry required for activity.


Subject(s)
Apolipoprotein A-I/genetics , Benzodiazepines/metabolism , Benzodiazepines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Acetylation , Amino Acid Sequence , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/metabolism , Benzodiazepines/chemical synthesis , Benzodiazepines/chemistry , Binding Sites , Crystallography, X-Ray , Drug Discovery , Epigenomics , Hep G2 Cells , Histones/chemistry , Histones/genetics , Histones/metabolism , Humans , Lysine/chemistry , Lysine/genetics , Lysine/metabolism , Models, Molecular , Molecular Sequence Data , Molecular Structure , Molecular Targeted Therapy , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Stereoisomerism , Transcription Factors , Up-Regulation
9.
J Cardiovasc Pharmacol Ther ; 15(2): 196-202, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20332533

ABSTRACT

Cholesteryl ester transfer protein (CETP) plays a key role in high-density lipoprotein (HDL) cholesterol metabolism, but normal mice are deficient in CETP. In this study, transgenic mice expressing both human apolipoprotein B 100 (ApoB-100) and human CETP (hApoB100/hCETP) were used to characterize the effects of CETP inhibition and peroxisome proliferator-activated receptor alpha (PPARalpha) agonism on lipid profiles. Torcetrapib (3, 10, and 30 mg/kg), a CETP inhibitor, fenofibrate (30 mg/kg), a weak PPARalpha agonist, and GW590735 (3 and 10 mg/kg), a potent and selective PPARalpha agonist were given orally for 14 days to hApoB100/hCETP mice and lipid profiles were assessed. The average percentages of HDL, low-density lipoprotein (LDL), and very-low-density lipoprotein (VLDL) cholesterol fractions in hApoB100/hCETP mice were 34.8%, 61.6%, and 3.6%, respectively, which is similar to those of normolipidemic humans. Both torcetrapib and fenofibrate significantly increased HDL cholesterol and reduced LDL cholesterol, and there was a tendency for torcetrapib to reduce VLDL cholesterol and triglycerides. GW590735 significantly increased HDL cholesterol, decreased LDL and VLDL cholesterol, and significantly reduced triglycerides. Maximal increases in HDL cholesterol were 37%, 53%, and 84% with fenofibrate, torcetrapib, and GW590735, respectively. These results, in mice that exhibit a more human-like lipid profile, demonstrate an improved lipid profile with torcetrapib, fenofibrate, and GW590735, and support the use of selective PPARalpha agonism for the treatment of lipid disorders. In addition, these data demonstrate the use of hApoB100/hCETP transgenic mice to identify, characterize, and screen compounds that increase HDL cholesterol.


Subject(s)
Anticholesteremic Agents/pharmacology , Apolipoprotein B-100/genetics , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cholesterol, HDL/blood , Cholesterol, LDL/blood , PPAR alpha/agonists , Animals , Cholesterol Ester Transfer Proteins/genetics , Cholesterol, VLDL/blood , Dose-Response Relationship, Drug , Fenofibrate/pharmacology , Humans , Mice , Mice, Transgenic , Propionates/pharmacology , Quinolines/administration & dosage , Quinolines/pharmacology , Thiazoles/pharmacology
10.
Eur J Pharmacol ; 618(1-3): 28-36, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19616540

ABSTRACT

Stearoyl-CoA Desaturase 1 (SCD1) is a central enzyme that catalyzes the biosynthesis of monounsaturated fatty acids from saturated fatty acids. SCD1 is an emerging target in obesity and insulin resistance due to the improved metabolic profile obtained when the enzyme is genetically inactivated. Here, we have investigated if the pharmacological inhibition of SCD1 could elicit the same profile. We have identified a small molecule, GSK993 and characterized it as a potent and orally available SCD1 inhibitor. In Zucker(fa/fa) rats, GSK993 exerted a marked reduction in hepatic lipids as well as a significant improvement of glucose tolerance. Furthermore, in a diet-induced insulin resistant rat model, GSK993 induced a very strong reduction in Triton-induced hepatic Very Low Density Lipoprotein-Triglyceride production. In addition, following a hyperinsulinemic-euglycemic clamp in GSK993-treated animals, we observed an improvement in the whole body insulin sensitivity as reflected by an increase in the glucose infusion rate. Taken together, these findings demonstrate that the pharmacological inhibition of SCD1 translates into improved lipid and glucose metabolic profiles and raises the interest of SCD1 inhibitors as potential new drugs for the treatment of insulin resistance.


Subject(s)
Enzyme Inhibitors/pharmacology , Insulin Resistance , Insulin/pharmacology , Isoquinolines/pharmacology , Pyrazoles/pharmacology , Stearoyl-CoA Desaturase/antagonists & inhibitors , Animals , Cattle , Cell Line, Tumor , Diet/adverse effects , Disease Models, Animal , Drug Evaluation, Preclinical , Glucose/metabolism , Humans , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Rats
11.
J Lipid Res ; 47(6): 1281-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16508037

ABSTRACT

The alkaloid drug berberine (BBR) was recently described to decrease plasma cholesterol and triglycerides (TGs) in hypercholesterolemic patients by increasing expression of the hepatic low density lipoprotein receptor (LDLR). Using HepG2 human hepatoma cells, we found that BBR inhibits cholesterol and TG synthesis in a similar manner to the AMP-activated protein kinase (AMPK) activator 5-aminoimidazole-4-carboxamide 1-beta-ribofuranoside (AICAR). Significant increases in AMPK phosphorylation and AMPK activity were observed when the cells were incubated with BBR. Activation of AMPK was also demonstrated by measuring the phosphorylation of acetyl-CoA carboxylase, a substrate of AMPK, correlated with a subsequent increase in fatty acid oxidation. All of these effects were abolished by the mitogen-activated protein kinase kinase inhibitor PD98059. Treatment of hyperlipidemic hamsters with BBR decreased plasma LDL cholesterol and strongly reduced fat storage in the liver. These findings indicate that BBR, in addition to upregulating the LDLR, inhibits lipid synthesis in human hepatocytes through the activation of AMPK. These effects could account for the strong reduction of plasma TGs observed with this drug in clinical trials.


Subject(s)
Adenylate Kinase/metabolism , Berberine/pharmacology , Hypolipidemic Agents/pharmacology , Lipids/biosynthesis , Acetyl-CoA Carboxylase/metabolism , Aminoimidazole Carboxamide/pharmacology , Animals , Cell Line, Tumor , Cricetinae , Enzyme Activation/drug effects , Fatty Acids/metabolism , Humans , Lipid Metabolism/drug effects , Male , Phosphorylation/drug effects , Receptors, LDL/metabolism
12.
Br J Pharmacol ; 145(2): 166-77, 2005 May.
Article in English | MEDLINE | ID: mdl-15723089

ABSTRACT

1 Chronic liver disease is characterized by an exacerbated accumulation of matrix, causing progressive fibrosis, which may lead to cirrhosis. Transforming growth factor beta (TGF-beta), a well-known profibrotic cytokine, transduces its signal through the ALK5 ser/thr kinase receptor, and increases transcription of different genes including PAI-1 and collagens. The identification of GW6604 (2-phenyl-4-(3-pyridin-2-yl-1H-pyrazol-4-yl)pyridine), an ALK5 inhibitor, allowed us to evaluate the therapeutic potential of inhibiting TGF-beta pathway in different models of liver disease. 2 A cellular assay was used to identify GW6604 as a TGF-beta signaling pathway inhibitor. This ALK5 inhibitor was then tested in a model of liver hepatectomy in TGF-beta-overexpressing transgenic mice, in an acute model of liver disease and in a chronic model of dimethylnitrosamine (DMN)-induced liver fibrosis. 3 In vitro, GW6604 inhibited autophosphorylation of ALK5 with an IC(50) of 140 nM and in a cellular assay inhibited TGF-beta-induced transcription of PAI-1 (IC(50): 500 nM). In vivo, GW6604 (40 mg kg(-1) p.o.) increased liver regeneration in TGF-beta-overexpressing mice, which had undergone partial hepatectomy. In an acute model of liver disease, GW6604 reduced by 80% the expression of collagen IA1. In a chronic model of DMN-induced fibrosis where DMN was administered for 6 weeks and GW6604 dosed for the last 3 weeks (80 mg kg(-1) p.o., b.i.d.), mortality was prevented and DMN-induced elevations of mRNA encoding for collagen IA1, IA2, III, TIMP-1 and TGF-beta were reduced by 50-75%. Inhibition of matrix genes overexpression was accompanied by reduced matrix deposition and reduction in liver function deterioration, as assessed by bilirubin and liver enzyme levels. 4 Our results suggest that inhibition of ALK5 could be an attractive new approach to treatment of liver fibrotic diseases by both preventing matrix deposition and promoting hepatocyte regeneration.


Subject(s)
Activin Receptors, Type I/antagonists & inhibitors , Dimethylnitrosamine , Liver Cirrhosis/prevention & control , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Activin Receptors, Type I/metabolism , Acute Disease , Animals , Cell Line, Tumor , Chronic Disease , Hepatectomy , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Liver Regeneration/drug effects , Male , Mice , Mice, Transgenic , Phosphorylation , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/genetics , Protein Serine-Threonine Kinases , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transcription, Genetic , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...