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










Publication year range
1.
Biochim Biophys Acta Gen Subj ; 1865(2): 129800, 2021 02.
Article in English | MEDLINE | ID: mdl-33246032

ABSTRACT

BACKGROUND: Due to the importance of both prostaglandins (PGs) and leukotrienes (LTs) as pro-inflammatory mediators, and the potential for eicosanoid shunting in the presence of pathway target inhibitors, we have investigated an approach to inhibiting the formation of both PGs and LTs as part of a multi-targeted drug discovery effort. METHODS: We generated ligand-protein X-ray crystal structures of known inhibitors of microsomal prostaglandin E2 synthase-1 (mPGES-1) and the 5-Lipoxygenase Activating Protein (FLAP), with their respective proteins, to understand the overlapping pharmacophores. We subsequently used molecular modeling and structure-based drug design (SBDD) to identify hybrid structures intended to inhibit both targets. RESULTS: This work enabled the preparation of compounds 4 and 5, which showed potent in vitro inhibition of both targets. SIGNIFICANCE: Our findings enhance the structural understanding of mPGES-1 and FLAP's unique ligand binding pockets and should accelerate the discovery of additional dual inhibitors for these two important integral membrane protein drug targets.


Subject(s)
5-Lipoxygenase-Activating Protein Inhibitors/pharmacology , Drug Discovery , Eicosanoids/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Prostaglandin-E Synthases/antagonists & inhibitors , 5-Lipoxygenase-Activating Protein Inhibitors/chemistry , 5-Lipoxygenase-Activating Proteins/metabolism , Eicosanoids/metabolism , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Prostaglandin-E Synthases/metabolism , Structure-Activity Relationship
2.
Pharmacol Res Perspect ; 5(3): e00316, 2017 06.
Article in English | MEDLINE | ID: mdl-28603634

ABSTRACT

Prostaglandin (PG) E2 is the key driver of inflammation associated with arthritic conditions. Inhibitors of PGE 2 production (NSAIDs and Coxibs) are used to treat these conditions, but carry significant side effect risks due to the inhibition of all prostanoids that play important physiological function. The activities of PGE 2 are transduced through various receptor sub-types. Prostaglandin E2 type 4 receptor (EP4) is associated with the development of inflammation and autoimmunity. We therefore are interested in identifying novel EP4 antagonists to treat the signs and symptoms of arthritis without the potential side effects of PGE 2 modulators such as NSAIDs and Coxibs. Novel EP4 antagonists representing distinct chemical scaffolds were identified using a variety of in vitro functional assays and were shown to be selective and potent. The compounds were shown to be efficacious in animal models of analgesia, inflammation, and arthritis.

3.
Bioorg Med Chem Lett ; 27(6): 1478-1483, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28190634

ABSTRACT

We describe a novel class of acidic mPGES-1 inhibitors with nanomolar enzymatic and human whole blood (HWB) potency. Rational design in conjunction with structure-based design led initially to the identification of anthranilic acid 5, an mPGES-1 inhibitor with micromolar HWB potency. Structural modifications of 5 improved HWB potency by over 1000×, reduced CYP2C9 single point inhibition, and improved rat clearance, which led to the selection of [(cyclopentyl)ethyl]benzoic acid compound 16 for clinical studies. Compound 16 showed an IC80 of 24nM for inhibition of PGE2 formation in vitro in LPS-stimulated HWB. A single oral dose resulted in plasma concentrations of 16 that exceeded its HWB IC80 in both rat (5mg/kg) and dog (3mg/kg) for over twelve hours.


Subject(s)
Benzoates/chemistry , Benzoates/pharmacology , Drug Discovery , Microsomes/drug effects , Prostaglandin-E Synthases/antagonists & inhibitors , Animals , Crystallography, X-Ray , Dogs , Microsomes/enzymology , Prostaglandin-E Synthases/chemistry , Rats
4.
Bioorg Med Chem Lett ; 26(19): 4824-4828, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27554445

ABSTRACT

Here we report on novel, potent 3,3-dimethyl substituted N-aryl piperidine inhibitors of microsomal prostaglandin E synthases-1(mPGES-1). Example 14 potently inhibited PGE2 synthesis in an ex vivo human whole blood (HWB) assay with an IC50 of 7nM. In addition, 14 had no activity in human COX-1 or COX-2 assays at 30µM, and failed to inhibit human mPGES-2 at 62.5µM in a microsomal prep assay. These data are consistent with selective mPGES-1-mediated reduction of PGE2. In dog, 14 had oral bioavailability (74%), clearance (3.62mL/(min*kg)) and volume of distribution (Vd,ss=1.6L/kg) values within our target ranges. For these reasons, 14 was selected for further study.


Subject(s)
Piperidines/chemistry , Piperidines/pharmacology , Prostaglandin-E Synthases/antagonists & inhibitors , A549 Cells , Animals , Crystallography, X-Ray , Dogs , Humans , Piperidines/pharmacokinetics , Rats , Species Specificity , Structure-Activity Relationship
5.
Bioorg Med Chem Lett ; 26(9): 2303-7, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27020304

ABSTRACT

Continued SAR optimization of a series of 3-methylpyridine-2-carbonyl amino-2,4-dimethyl-benzoic acid led to the selection of compound 4f for clinical studies. Compound 4f showed an IC50 of 123nM for inhibition of PGE2-induced TNFα reduction in an ex vivo LPS-stimulated human whole blood assay (showing >10-fold increase over clinical compound CJ-023,423). Pharmacokinetic profile, selectivity and in vivo efficacy comparing 4f to NSAID diclofenac in the monoiodoacetic acid (MIA) pain model and adjuvant induced arthritis (AIA) inflammatory model are included.


Subject(s)
Benzoates/pharmacology , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Animals , Benzoates/chemistry , Rats , X-Ray Diffraction
6.
J Pharmacol Exp Ther ; 356(3): 635-44, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26740668

ABSTRACT

Prostaglandin (PG) E2 plays a critical role in eliciting inflammation. Nonsteroidal anti-inflammatory drugs and selective inhibitors of cyclooxygenase, which block PGE2 production, have been used as key agents in treating inflammation and pain associated with arthritis and other conditions. However, these agents have significant side effects such as gastrointestinal bleeding and myocardial infarction, since they also block the production of prostanoids that are critical for other normal physiologic functions. Microsomal prostaglandin E2 synthase-1 is a membrane-bound terminal enzyme in the prostanoid pathway, which acts downstream of cyclooxygenase 2 and is responsible for PGE2 production during inflammation. Thus, inhibition of this enzyme would be expected to block PGE2 production without inhibiting other prostanoids and would provide analgesic efficacy without the side effects. In this report, we describe novel microsomal prostaglandin E2 synthase-1 inhibitors that are potent in blocking PGE2 production and are efficacious in a guinea pig monoiodoacetate model of arthralgia. These molecules may be useful in treating the signs and symptoms associated with arthritis.


Subject(s)
Analgesics/chemistry , Analgesics/pharmacology , Imidazoles/chemistry , Imidazoles/pharmacology , Intramolecular Oxidoreductases/antagonists & inhibitors , Microsomes/drug effects , Phenanthrenes/chemistry , Phenanthrenes/pharmacology , Analgesia/methods , Animals , Celecoxib/chemistry , Celecoxib/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Guinea Pigs , Humans , Intramolecular Oxidoreductases/metabolism , Male , Microsomes/enzymology , Pain Measurement/drug effects , Pain Measurement/methods , Prostaglandin-E Synthases , Rats
7.
Bioorg Med Chem Lett ; 26(3): 931-935, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26764191

ABSTRACT

Two new series of EP4 antagonists containing a 3-methylaryl-2-carbonyl core have been identified. One series has a 3-substituted-phenyl core, while the other one incorporates a 3-substituted pyridine. Both series led to compounds with potent activity in functional and human whole blood (hWB) assays. In the pyridine series, compound 7a was found to be a highly potent and selective EP4 antagonist, with suitable rat and dog pharmacokinetic profiles.


Subject(s)
Benzoic Acid/chemistry , Picolines/chemistry , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Animals , Benzoic Acid/pharmacokinetics , Benzoic Acid/therapeutic use , Disease Models, Animal , Dogs , Drug Evaluation, Preclinical , Half-Life , Humans , Inhibitory Concentration 50 , Pain/drug therapy , Protein Binding , Rats , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Structure-Activity Relationship
8.
J Med Chem ; 59(1): 194-205, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26653180

ABSTRACT

As part of a program aimed at the discovery of antinociceptive therapy for inflammatory conditions, a screening hit was found to inhibit microsomal prostaglandin E synthase-1 (mPGES-1) with an IC50 of 17.4 µM. Structural information was used to improve enzyme potency by over 1000-fold. Addition of an appropriate substituent alleviated time-dependent cytochrome P450 3A4 (CYP3A4) inhibition. Further structure-activity relationship (SAR) studies led to 8, which had desirable potency (IC50 = 12 nM in an ex vivo human whole blood (HWB) assay) and absorption, distribution, metabolism, and excretion (ADME) properties. Studies on the formulation of 8 identified 8·H3PO4 as suitable for clinical development. Omission of a lipophilic portion of the compound led to 26, a readily orally bioavailable inhibitor with potency in HWB comparable to celecoxib. Furthermore, 26 was selective for mPGES-1 inhibition versus other mechanisms in the prostanoid pathway. These factors led to the selection of 26 as a second clinical candidate.


Subject(s)
Analgesics/chemical synthesis , Analgesics/pharmacology , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/pharmacology , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Intramolecular Oxidoreductases/antagonists & inhibitors , Microsomes/enzymology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biological Availability , Celecoxib/pharmacology , Cyclooxygenase Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors/chemical synthesis , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Dogs , Drug Discovery , Humans , Microsomes/drug effects , Models, Molecular , Prostaglandin-E Synthases , Rats , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 26(1): 105-9, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26608552

ABSTRACT

A novel series of EP4 antagonists, based on a quinoline scaffold, has been discovered. Medicinal chemistry efforts to optimize the potency of the initial hit are described. A highly potent compound in a clinically relevant human whole blood assay was identified. Selectivity and pharmacokinetic profiles of this compound are discussed.


Subject(s)
Benzoates/pharmacology , Drug Discovery , Naphthalenes/pharmacology , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Benzoates/chemical synthesis , Benzoates/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Naphthalenes/chemical synthesis , Naphthalenes/chemistry , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 25(16): 3176-8, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26091726

ABSTRACT

EP4 is a prostaglandin E2 receptor that is a target for potential anti-nociceptive therapy. Described herein is a class of amphoteric EP4 antagonists which reverses PGE2-induced suppression of TNFα production in human whole blood. From this class, a potent and highly bioavailable compound (6) has been selected for potential clinical studies. EP4 binding and functional data, selectivity, and pharmacokinetic properties of this compound are included.


Subject(s)
Analgesics/chemistry , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Analgesics/metabolism , Analgesics/pharmacokinetics , Animals , Blood Cells/cytology , Blood Cells/drug effects , Blood Cells/metabolism , Dogs , Half-Life , Humans , Lipopolysaccharides/toxicity , Protein Binding , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism
11.
J Med Chem ; 58(11): 4727-37, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25961169

ABSTRACT

Microsomal prostaglandin E synthase 1 (mPGES-1) is an α-helical homotrimeric integral membrane inducible enzyme that catalyzes the formation of prostaglandin E2 (PGE2) from prostaglandin H2 (PGH2). Inhibition of mPGES-1 has been proposed as a therapeutic strategy for the treatment of pain, inflammation, and some cancers. Interest in mPGES-1 inhibition can, in part, be attributed to the potential circumvention of cardiovascular risks associated with anti-inflammatory cyclooxygenase 2 inhibitors (coxibs) by targeting the prostaglandin pathway downstream of PGH2 synthesis and avoiding suppression of antithrombotic prostacyclin production. We determined the crystal structure of mPGES-1 bound to four potent inhibitors in order to understand their structure-activity relationships and provide a framework for the rational design of improved molecules. In addition, we developed a light-scattering-based thermal stability assay to identify molecules for crystallographic studies.


Subject(s)
Analgesics/chemistry , Anti-Inflammatory Agents/chemistry , Drug Design , Enzyme Inhibitors/chemistry , Imidazoles/chemistry , Intramolecular Oxidoreductases/chemistry , Amino Acid Sequence , Analgesics/metabolism , Analgesics/therapeutic use , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/therapeutic use , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Humans , Intramolecular Oxidoreductases/metabolism , Microsomes/enzymology , Models, Molecular , Molecular Sequence Data , Molecular Structure , Prostaglandin-E Synthases , Protein Conformation , Sequence Homology, Amino Acid
12.
J Orthop Res ; 32(3): 471-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24243768

ABSTRACT

We compared the effect of a sclerostin antibody to that of a clinically relevant dose of parathyroid hormone (PTH) in a rat model for metaphyseal bone healing. Screws of steel or poly methyl methacrylate (PMMA) were inserted bilaterally into the proximal tibia of young male rats. During 4 weeks the animals then received injections of either phosphate buffered saline (control), sclerostin antibody (25 mg/kg, twice weekly) or PTH (5 µg/kg, daily). The healing response around the screws was then assessed by mechanical testing and X-ray microtomography (µCT). To distinguish between effects on healing and general effects on the skeleton, other untraumatized bone sites and serum biomarkers were also assessed. After 4 weeks of treatment, PTH yielded a 48% increase in screw pull-out force compared to control (p = 0.03), while the antibody had no significant effect. In contrast, the antibody increased femoral cortical and vertebral strength where PTH had no significant effect. µCT showed only slight changes that were statistically significant for the antibody mainly at cortical sites. The results suggest that a relatively low dose of PTH stimulates metaphyseal repair (screw fixation) specifically, whereas the sclerostin antibody has wide-spread effects, mainly on cortical bone, with less influence on metaphyseal healing.


Subject(s)
Antibodies/therapeutic use , Bone Density Conservation Agents/therapeutic use , Bone Morphogenetic Proteins/immunology , Fracture Healing/drug effects , Fractures, Bone/drug therapy , Genetic Markers/immunology , Teriparatide/therapeutic use , Animals , Bone Density Conservation Agents/pharmacology , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Screws , Drug Evaluation, Preclinical , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Teriparatide/pharmacology
13.
Endocrinology ; 152(5): 1767-78, 2011 May.
Article in English | MEDLINE | ID: mdl-21343258

ABSTRACT

We compared teriparatide (TPTD) and strontium ranelate (SR) efficacy on bone formation activity in a mature rat model of estrogen-deficiency bone loss. Rats were ovariectomized (OVX) at age 6 months and permitted to lose bone for 2 months to establish osteopenia before initiation of treatment with TPTD (5 or 15 µg/kg · d sc) or SR (150 or 450 mg/kg · d oral gavage). After 3 wk, RT-PCR analyses of bone formation genes in the distal femur metaphysis showed significant elevation of collagen 1α2, osteocalcin, bone sialoprotein, alkaline phosphatase, and Runx2 gene expression at both TPTD doses, relative to OVX controls. SR had no significant effect on expression of these genes. TPTD treatment for 12 wk dose dependently increased lumbar vertebral (LV) and femoral midshaft bone mineral content (BMC) and bone mineral density over pretreatment and age-matched OVX controls. SR 150 increased BMC, and SR 450 increased BMC and bone mineral density of femoral midshaft and LV over OVX controls. There were significant dose-dependent TPTD increases of LV and femoral neck strength, and TPTD 15 also increased midshaft strength compared with pretreatment and age-matched OVX controls. SR did not enhance bone strength relative to pretreatment or age-matched OVX controls. Histomorphometry of the proximal tibial metaphysis showed dose-dependent effects of TPTD on trabecular area, number, width, and osteoblast surface, bone mineralizing surface, and bone formation rate relative to pretreatment and age-matched OVX controls, whereas SR had no effect on these parameters. These findings confirmed the bone anabolic efficacy of teriparatide, but not SR in mature, osteopenic, OVX rats.


Subject(s)
Bone Diseases, Metabolic/prevention & control , Bone and Bones/drug effects , Ovariectomy , Teriparatide/pharmacology , Alkaline Phosphatase/genetics , Anabolic Agents/pharmacology , Animals , Bone Density/drug effects , Bone Density Conservation Agents/pharmacology , Bone Diseases, Metabolic/blood , Bone Diseases, Metabolic/pathology , Bone and Bones/metabolism , Bone and Bones/pathology , Collagen Type I/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Dose-Response Relationship, Drug , Female , Femur/drug effects , Femur/metabolism , Gene Expression/drug effects , Humans , Integrin-Binding Sialoprotein/genetics , Lumbar Vertebrae/drug effects , Lumbar Vertebrae/metabolism , Organometallic Compounds/pharmacology , Osteocalcin/blood , Osteocalcin/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Thiophenes/pharmacology , Time Factors
14.
J Proteome Res ; 6(11): 4218-29, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17924680

ABSTRACT

Type-I procollagen aminoterminal propeptide (PINP) is a useful biomarker for bone formation activity that is used to monitor treatment of bone disorders including osteoporosis. Studies with human patients under long-term therapy for osteoporosis by daily injection of parathyroid hormone (PTH) demonstrated that the circulating level of PINP at 3 months of treatment, measured by radioimmunoassay, was a good predictor for bone mineral density (BMD) at 18 months. It is important to have PINP assays for other species to elucidate processes of bone formation and for the development of new therapeutic options that can enhance bone formation activity. Currently, only a human PINP radioimmunoassay is commercially available for clinical use, which may not be cross reactive with PINP from other species. For example, rat PINP has little amino acid sequence homology to human PINP. Therefore, we developed a new, highly sensitive, high-throughput mass spectrometry-based assay for PINP from rat plasma or serum that does not rely on antibody reagents. Circulating levels of PINP showed age-dependent changes in rats. Prednisolone treatment, which is known to retard bone formation activity, led to a significant decrease in PINP, whereas PTH treatment dose-dependently increased PINP. The throughput of the assay parallels that of most antibody-based assays so that it can handle a large number of samples that are generated from preclinical animal studies. PINP in rats may serve as a biomarker for bone formation activity, and this assay could be instrumental in studying bone physiology in rat experimental models.


Subject(s)
Biomarkers , Bone Development , Bone and Bones/metabolism , Mass Spectrometry/methods , Peptide Fragments/chemistry , Procollagen/chemistry , Animals , Cattle , Dogs , Dose-Response Relationship, Drug , Goats , Guinea Pigs , Horses , Parathyroid Hormone/metabolism , Prednisolone/pharmacology , Rabbits , Radioimmunoassay/methods , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity , Sheep , Swine
15.
Osteoarthritis Cartilage ; 14(8): 749-58, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16549373

ABSTRACT

OBJECTIVE: To investigate mitogen activated protein (MAP) kinase pathways for their ability to differentially regulate the expression of matrix metalloprotease (MMP)-1 and -13 in human chondrosarcoma cells using pathway-selective inhibitors. DESIGN: Human chondrosarcoma cell lines (SW1353 and JJ012) and human articular chondrocytes (HACs) were treated with cytokines (IL-1beta and TNFalpha) and the expression of MMP-1 and -13 was analyzed. The effects of MAP kinase inhibitors on cytokine-induced expression of MMP-1 and -13 were evaluated using ELISA and Western blot analyses. The possible involvement of the Runx2 pathway in mediating p38 effects on MMP-13 expression was analyzed using promoter-reporter assays, ELISA and immunoprecipitation analyses. RESULTS: IL-1beta and TNFalpha strongly induced the expression of MMP-1 and -13 in SW1353 cells and HACs, whereas only TNFalpha was found to induce the expression of these two MMPs in JJ012 cells. Cytokine treatment did not result in a significant increase in the activity of MMPs because of the excess production of endogenous tissue inhibitors of metalloproteases (TIMPs). Treatment with p38 kinase inhibitors (SB203580 and SB242235) strongly inhibited cytokine-induced MMP-13 expression in a dose-dependent fashion while having a somewhat weaker inhibitory effect on MMP-1 expression. In contrast, inhibitors of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) pathways did not inhibit the expression of either MMP. Overexpression of Runx2 robustly stimulated the transcriptional activation of MMP-13 but had no effect on MMP-1 expression. Furthermore, IL-1beta induced the phosphorylation of Runx2, and this effect was blocked by a p38 kinase inhibitor. Our data suggest that Runx2 is likely to be a key downstream mediator of p38 effects in the differential regulation of IL-1beta induced MMP-13 expression. CONCLUSIONS: These studies demonstrate the differential inhibition of cytokine-induced MMP-1 and -13 expression by p38 kinase inhibitors in human chondrosarcoma cells. Our studies also suggest the involvement of Runx2, at least in part, in mediating the effects of p38 on MMP-13 expression.


Subject(s)
Chondrosarcoma/enzymology , Chondrosarcoma/immunology , Core Binding Factor Alpha 1 Subunit/pharmacology , Interleukin-1beta/pharmacology , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 1/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Blotting, Western/methods , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay/methods , Humans , Immunoprecipitation , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 13/genetics , Phosphorylation , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...