Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Eur J Med Chem ; 276: 116686, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39053192

ABSTRACT

With an objective to improve the profiles of the 1st generation non-basic MCHR1 antagonists, a lean design approach of replacing the bicyclic thienopyrimidine core with a monocyclic pyrrol-2-one chemotype was examined in the context of reducing aromatic ring count, while also contemplating enhanced flexibility as a means of decreasing flat character. The new compounds exhibited potent antagonism up to the sub-nanomolar range, thereby implying that the monocyclic ring could effectively serve as an effective bioisostere of the bicyclic system. The prototype compound 2m offered benefits like improved potency, reduced half-life, and enhanced solubility, while also demonstrating >5% reduction in weight gain in rats, thereby providing proof-of-concept for this new class of compounds as anti-obesity agents.


Subject(s)
Pyrroles , Animals , Rats , Structure-Activity Relationship , Humans , Molecular Structure , Pyrroles/pharmacology , Pyrroles/chemistry , Pyrroles/chemical synthesis , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/chemistry , Dose-Response Relationship, Drug , Drug Discovery , Male , Rats, Sprague-Dawley , Receptors, Somatostatin
2.
Toxicol Appl Pharmacol ; 438: 115885, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35090952

ABSTRACT

In a Phase 2 clinical trial, BMS-986020, a lysophosphatidic acid receptor-1 (LPA1) antagonist, produced hepatobiliary toxicity (increased ALT, AST, and ALP; cholecystitis) and increases in plasma bile acids (BA). Nonclinical investigations conducted to identify a potential mechanism(s) for this toxicity examined BMS-986020 and two LPA1 antagonists structurally distinct from BMS-986020 (BMS-986234 and BMS-986278). BMS-986020 inhibited hepatic BA efflux transporters BSEP (IC50 1.8 µM), MRP3 (IC50 22 µM), and MRP4 (IC50 6.2 µM) and inhibited BA canalicular efflux in human hepatocytes (68% at 10 µM). BMS-986020 inhibited mitochondrial function (basal and maximal respiration, ATP production, and spare capacity) in human hepatocytes and cholangiocytes at ≥10 µM and inhibited phospholipid efflux in human hepatocytes (MDR3 IC50 7.5 µM). A quantitative systems toxicology analysis (DILIsym®), considering pharmacokinetics, BA homeostasis, mitochondrial function, oxidative phosphorylation, and reactive intermediates performed for BMS-986020 recapitulated clinical findings ascribing the effects to BA transporter and mitochondrial electron transport chain inhibition. BMS-986234 and BMS-986278 minimally inhibited hepatic BA transporters (IC50 ≥20 µM) and did not inhibit MDR3 activity (IC50 >100 µM), nor did BMS-986234 inhibit BA efflux (≤50 µM) or mitochondrial function (≤30 µM) (BMS-986278 not evaluated). Multiple mechanisms may be involved in the clinical toxicity observed with BMS-986020. The data indicate that this toxicity was unrelated to LPA1 antagonism since the mechanisms that likely influenced the adverse clinical outcome of BMS-986020 were not observed with equipotent LPA1 antagonists BMS-986234 and BMS-986278. This conclusion is consistent with the lack of hepatobiliary toxicity in nonclinical and clinical safety studies with BMS-986278.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Digestive System Diseases/chemically induced , Idiopathic Pulmonary Fibrosis/drug therapy , Liver/drug effects , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Bile Acids and Salts/metabolism , Biological Transport/physiology , Cell Line , Cell Line, Tumor , Electron Transport/physiology , HEK293 Cells , Hep G2 Cells , Hepatocytes/drug effects , Humans , Mitochondria/drug effects
3.
J Med Chem ; 64(21): 15549-15581, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34709814

ABSTRACT

The oxycyclohexyl acid BMS-986278 (33) is a potent lysophosphatidic acid receptor 1 (LPA1) antagonist, with a human LPA1 Kb of 6.9 nM. The structure-activity relationship (SAR) studies starting from the LPA1 antagonist clinical compound BMS-986020 (1), which culminated in the discovery of 33, are discussed. The detailed in vitro and in vivo preclinical pharmacology profiles of 33, as well as its pharmacokinetics/metabolism profile, are described. On the basis of its in vivo efficacy in rodent chronic lung fibrosis models and excellent overall ADME (absorption, distribution, metabolism, excretion) properties in multiple preclinical species, 33 was advanced into clinical trials, including an ongoing Phase 2 clinical trial in patients with lung fibrosis (NCT04308681).


Subject(s)
Drug Discovery , Pulmonary Fibrosis/drug therapy , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Male , Mice , Molecular Structure , Pulmonary Fibrosis/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Lysophosphatidic Acid/metabolism , Structure-Activity Relationship
4.
Nat Commun ; 12(1): 3770, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34145282

ABSTRACT

Circulating cell-free DNA from blood plasma of cancer patients can be used to non-invasively interrogate somatic tumor alterations. Here we develop MSK-ACCESS (Memorial Sloan Kettering - Analysis of Circulating cfDNA to Examine Somatic Status), an NGS assay for detection of very low frequency somatic alterations in 129 genes. Analytical validation demonstrated 92% sensitivity in de-novo mutation calling down to 0.5% allele frequency and 99% for a priori mutation profiling. To evaluate the performance of MSK-ACCESS, we report results from 681 prospective blood samples that underwent clinical analysis to guide patient management. Somatic alterations are detected in 73% of the samples, 56% of which have clinically actionable alterations. The utilization of matched normal sequencing allows retention of somatic alterations while removing over 10,000 germline and clonal hematopoiesis variants. Our experience illustrates the importance of analyzing matched normal samples when interpreting cfDNA results and highlights the importance of cfDNA as a genomic profiling source for cancer patients.


Subject(s)
Biomarkers, Tumor/genetics , Circulating Tumor DNA/genetics , Genetic Markers/genetics , Neoplasms/genetics , DNA Mutational Analysis/methods , Gene Frequency/genetics , High-Throughput Nucleotide Sequencing , Humans , Mutation/genetics , Neoplasms/blood , Neoplasms/pathology
5.
J Am Soc Nephrol ; 28(11): 3300-3311, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28739650

ABSTRACT

Lysophosphatidic acid (LPA) functions through activation of LPA receptors (LPARs). LPA-LPAR signaling has been implicated in development of fibrosis. However, the role of LPA-LPAR signaling in development of diabetic nephropathy (DN) has not been studied. We examined whether BMS002, a novel dual LPAR1 and LPAR3 antagonist, affects development of DN in endothelial nitric oxide synthase-knockout db/db mice. Treatment of these mice with BMS002 from 8 to 20 weeks of age led to a significant reduction in albuminuria, similar to that observed with renin-angiotensin system inhibition (losartan plus enalapril). LPAR inhibition also prevented the decline in GFR observed in vehicle-treated mice, such that GFR at week 20 differed significantly between vehicle and LPAR inhibitor groups (P<0.05). LPAR inhibition also reduced histologic glomerular injury; decreased the expression of profibrotic and fibrotic components, including fibronectin, α-smooth muscle actin, connective tissue growth factor, collagen I, and TGF-ß; and reduced renal macrophage infiltration and oxidative stress. Notably, LPAR inhibition slowed podocyte loss (podocytes per glomerulus ±SEM at 8 weeks: 667±40, n=4; at 20 weeks: 364±18 with vehicle, n=7, and 536±12 with LPAR inhibition, n=7; P<0.001 versus vehicle). Finally, LPAR inhibition minimized the production of 4-hydroxynonenal (4-HNE), a marker of oxidative stress, in podocytes and increased the phosphorylation of AKT2, an indicator of AKT2 activity, in kidneys. Thus, the LPAR antagonist BMS002 protects against GFR decline and attenuates development of DN through multiple mechanisms. LPAR antagonism might provide complementary beneficial effects to renin-angiotensin system inhibition to slow progression of DN.


Subject(s)
Diabetic Nephropathies/prevention & control , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Animals , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/etiology , Disease Models, Animal , Mice , Receptors, Lysophosphatidic Acid/physiology
6.
J Med Chem ; 60(14): 6166-6190, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28635286

ABSTRACT

Agonism of the 5-HT2C receptor represents one of the most well-studied and clinically proven mechanisms for pharmacological weight reduction. Selectivity over the closely related 5-HT2A and 5-HT2B receptors is critical as their activation has been shown to lead to undesirable side effects and major safety concerns. In this communication, we report the development of a new screening paradigm that utilizes an active site mutant D134A (D3.32) 5-HT2C receptor to identify atypical agonist structures. We additionally report the discovery and optimization of a novel class of nonbasic heterocyclic amide agonists of 5-HT2C. SAR investigations around the screening hits provided a diverse set of potent agonists at 5-HT2C with high selectivity over the related 5-HT2A and 5-HT2B receptor subtypes. Further optimization through replacement of the amide with a variety of five- and six-membered heterocycles led to the identification of 6-(1-ethyl-3-(quinolin-8-yl)-1H-pyrazol-5-yl)pyridazin-3-amine (69). Oral administration of 69 to rats reduced food intake in an ad libitum feeding model, which could be completely reversed by a selective 5-HT2C antagonist.


Subject(s)
Arginine/analogs & derivatives , Flavones/chemistry , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin 5-HT2 Receptor Agonists/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Animals , Arginine/chemical synthesis , Arginine/chemistry , Arginine/pharmacology , Brain/metabolism , Caco-2 Cells , Cell Membrane Permeability , Feeding Behavior/drug effects , Flavones/chemical synthesis , Flavones/pharmacology , HEK293 Cells , Humans , Male , Membranes, Artificial , Mice, Knockout , Microsomes, Liver/metabolism , Mutation , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2B/metabolism , Receptor, Serotonin, 5-HT2C/genetics , Serotonin 5-HT2 Receptor Agonists/chemical synthesis , Serotonin 5-HT2 Receptor Agonists/pharmacokinetics , Serotonin 5-HT2 Receptor Agonists/pharmacology , Structure-Activity Relationship
7.
J Med Chem ; 60(4): 1417-1431, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28112924

ABSTRACT

A novel series of pyrrolidine-containing GPR40 agonists is described as a potential treatment for type 2 diabetes. The initial pyrrolidine hit was modified by moving the position of the carboxylic acid, a key pharmacophore for GPR40. Addition of a 4-cis-CF3 to the pyrrolidine improves the human GPR40 binding Ki and agonist efficacy. After further optimization, the discovery of a minor enantiomeric impurity with agonist activity led to the finding that enantiomers (R,R)-68 and (S,S)-68 have differential effects on the radioligand used for the binding assay, with (R,R)-68 potentiating the radioligand and (S,S)-68 displacing the radioligand. Compound (R,R)-68 activates both Gq-coupled intracellular Ca2+ flux and Gs-coupled cAMP accumulation. This signaling bias results in a dual mechanism of action for compound (R,R)-68, demonstrating glucose-dependent insulin and GLP-1 secretion in vitro. In vivo, compound (R,R)-68 significantly lowers plasma glucose levels in mice during an oral glucose challenge, encouraging further development of the series.


Subject(s)
Hypoglycemic Agents/pharmacology , Pyrrolidines/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Blood Glucose/analysis , Blood Glucose/metabolism , Cell Line , Cells, Cultured , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/therapeutic use , Insulin/metabolism , Male , Mice, Inbred C57BL , Models, Molecular , Pyrrolidines/chemistry , Pyrrolidines/pharmacokinetics , Pyrrolidines/therapeutic use , Rats , Receptors, G-Protein-Coupled/metabolism
8.
J Med Chem ; 59(19): 8848-8858, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27564419

ABSTRACT

The potent MCHR1 in vitro and in vivo antagonist activity of a series of cyclic tertiary alcohols derived from compound 2b is described. Subsequent pharmacokinetic and pharmacodynamic studies identified BMS-814580 (compound 10) as a highly efficacious antiobesity agent with a relatively clean in vitro and in vivo safety profile.


Subject(s)
Anti-Obesity Agents/chemistry , Anti-Obesity Agents/therapeutic use , Obesity/drug therapy , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Receptors, Somatostatin/antagonists & inhibitors , Animals , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/pharmacology , Dogs , Halogenation , Humans , Macaca fascicularis , Male , Mice , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Receptors, Somatostatin/metabolism , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 25(20): 4412-8, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26386604

ABSTRACT

Non-basic azolotriazinones were explored using an empirical free brain exposures-driven approach to identify potent MCHR1 antagonists for evaluation in in vivo efficacy studies. An optimized lead from this series, 1j (rMCHR1 Ki=1.8 nM), demonstrated a 6.9% reduction in weight gain relative to vehicle in a rat model at 30 mg/kg after 4 days of once-daily oral treatment as a glycine prodrug. Despite a promising efficacy profile, an assessment of the biliary toxicity risk of this compound rendered this compound non-progressible.


Subject(s)
Brain/drug effects , Obesity/drug therapy , Receptors, Somatostatin/antagonists & inhibitors , Triazines/pharmacology , Animals , Brain/metabolism , Dose-Response Relationship, Drug , Humans , Molecular Structure , Obesity/metabolism , Rats , Structure-Activity Relationship , Triazines/administration & dosage , Triazines/chemistry
10.
Bioorg Med Chem Lett ; 25(14): 2793-9, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26022839

ABSTRACT

Our investigation of the structure-activity and structure-liability relationships for dihydropyrrolopyrazol-6-one MCHR1 antagonists revealed that off-rate characteristics, inferred from potencies in a FLIPR assay following a 2 h incubation, can impact in vivo efficacy. The in vitro and exposure profiles of dihydropyrrolopyrazol-6-ones 1b and 1e were comparable to that of the thienopyrimidinone counterparts 41 and 43 except for a much faster MCHR1 apparent off-rate. The greatly diminished dihydropyrrolopyrazol-6-one anti-obesity response may be the consequence of this rapid off-rate.


Subject(s)
Anti-Obesity Agents/chemistry , Pyrazoles/chemistry , Receptors, Somatostatin/antagonists & inhibitors , Animals , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Half-Life , Humans , Obesity/drug therapy , Protein Binding , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Somatostatin/metabolism , Structure-Activity Relationship , Weight Loss/drug effects
11.
J Med Chem ; 57(18): 7509-22, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25165888

ABSTRACT

Identification of MCHR1 antagonists with a preclinical safety profile to support clinical evaluation as antiobesity agents has been a challenge. Our finding that a basic moiety is not required for MCHR1 antagonists to achieve high affinity allowed us to explore structures less prone to off-target activities such as hERG inhibition. We report the SAR evolution of hydroxylated thienopyrimidinone ethers culminating in the identification of 27 (BMS-819881), which entered obesity clinical trials as the phosphate ester prodrug 35 (BMS-830216).


Subject(s)
Anti-Obesity Agents/pharmacology , Drug Discovery , Obesity/drug therapy , Receptors, Somatostatin/antagonists & inhibitors , Animals , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/therapeutic use , Dogs , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Humans , Male , Rats
12.
J Med Chem ; 56(23): 9586-600, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24182233

ABSTRACT

Several strategies have been employed to reduce the long in vivo half-life of our lead CB1 antagonist, triazolopyridazinone 3, to differentiate the pharmacokinetic profile versus the lead clinical compounds. An in vitro and in vivo clearance data set revealed a lack of correlation; however, when compounds with <5% free fraction were excluded, a more predictable correlation was observed. Compounds with log P between 3 and 4 were likely to have significant free fraction, so we designed compounds in this range to give more predictable clearance values. This strategy produced compounds with desirable in vivo half-lives, ultimately leading to the discovery of compound 46. The progression of compound 46 was halted due to the contemporaneous marketing and clinical withdrawal of other centrally acting CB1 antagonists; however, the design strategy successfully delivered a potent CB1 antagonist with the desired pharmacokinetic properties and a clean off-target profile.


Subject(s)
Pyridazines/pharmacokinetics , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Triazoles/pharmacokinetics , Animals , Cytochrome P-450 Enzyme System/metabolism , Drug Discovery , Half-Life , Protein Binding , Pyridazines/chemistry , Rats , Structure-Activity Relationship , Triazoles/chemistry
13.
Cancer Biol Ther ; 9(6): 469-76, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20087061

ABSTRACT

We previously identified metal-responsive transcription factor-1 (MTF-1) as a positive contributor to mouse fibrosarcoma growth through effects on cell survival, proliferation, tumor angiogenesis and extracellular matrix remodeling. In the present study, we investigated MTF-1 protein expression in human tissues by specific immunostaining of both normal and tumor tissue samples. Immunohistochemical (IHC) staining of a human tissue microarray (TMA), using a unique anti-human MTF-1 antibody, indicated constitutive MTF-1 expression in most normal tissues, with liver and testis displaying comparatively high levels of expression. Nevertheless, MTF-1 protein levels were found to be significantly elevated in diverse human tumor types, including breast, lung and cervical carcinomas. IHC analysis of a separate panel of full-size tissue sections of human breast cancers, including tumor and normal adjacent, surrounding tissue, confirmed and extended the results of the TMA analysis. Taken with our previous findings, this new study suggests a role for MTF-1 in human tumor development, growth or spread. Moreover, the study suggests that MTF-1 could be a novel therapeutic target that offers the opportunity to manipulate metal or redox homeostasis in tumor cells.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasms/metabolism , Transcription Factors/metabolism , Breast/chemistry , Breast/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , DNA-Binding Proteins/genetics , Female , Humans , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Transcription Factors/genetics , Transcription Factor MTF-1
14.
Cancer Lett ; 289(1): 111-8, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19751965

ABSTRACT

Tissue inhibitor of metalloproteinase-2 (TIMP-2), the endogenous inhibitor of matrix metalloproteinase-2 (MMP-2), regulates tumor invasion by modulating the activity of MMP-2. In addition, TIMP-2 is involved in the direct regulation of cell growth and angiogenesis, independent of MMP inhibition. Therefore, the development of therapeutic agents that increase TIMP-2 levels may offer a novel and broad approach to anti-neoplastic therapy. We report that a novel small molecule synthetic flavanoid SR13179, which inhibits the invasion of a highly metastatic human breast cancer cell line MCF-10CA1a through Matrigel, significantly increases protein and mRNA levels of TIMP-2 in a time- and dose-dependent manner. SR13179 inhibits the gelatinolytic activity of MMP-2 by >50% but has no effect on MMP-2 protein expression. SR13179 also possesses potent anti-tumor activity in several tumor cell lines regardless of their hormone receptor, p53 or multi-drug resistance status. Given the multifunctional inhibitory activity of TIMP-2 on tumor growth and invasion, the observed increase in TIMP-2 expression by SR13179 may play a central role in the anti-tumor and anti-invasive activity of this novel small molecule. Modulation of TIMP-2 protein expression may be a new molecular target for anti-metastatic adjuvant therapy for breast and other cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Flavonoids/pharmacology , Matrix Metalloproteinase Inhibitors , Tissue Inhibitor of Metalloproteinase-2/metabolism , Antineoplastic Agents/chemical synthesis , Blotting, Western , Breast Neoplasms/enzymology , Cell Line, Tumor , Female , Flavonoids/chemical synthesis , Humans , Matrix Metalloproteinase 2/metabolism , Neoplasm Invasiveness , Up-Regulation/drug effects
15.
Cancer Biol Ther ; 8(13): 1252-61, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19633434

ABSTRACT

The peroxisome proliferator-activated receptor delta (PPARdelta) is a ligand-activated, nuclear receptor transcription factor that has a documented role in glucose and lipid homeostasis. Recent studies have implicated this nuclear receptor in numerous aspects of oncogenesis. We report herein the characterization of a novel small-molecule (SR13904) that inhibits PPARdelta agonist-induced transactivation and functions as a PPARdelta antagonist. SR13904 also antagonizes PPARgamma transactivation, albeit with much weaker potency. SR13904 displays inhibitory effects on cellular proliferation and survival in several human carcinoma lines, including lung, breast and liver. These inhibitory effects of SR13904 on tumor cells were linked to a G(1)/S cell cycle block and increased apoptosis. Molecular studies show that SR13904 treatment of a lung cancer cell line, A549, results in markedly reduced levels of a number of cell cycle proteins including cyclin A and D, and cyclin dependent kinase (CDK) 2 and 4. The inhibitory effects on CDK2 appear to be transcriptional. Several of these cell cycle-related genes are known to be upregulated by PPARdelta. The antitumor activities of SR13904 suggest that antagonism of PPARdelta-mediated transactivation may inhibit tumorigenesis and that pharmacological inhibition of PPARdelta may be a potential strategy for treatment or prevention of cancer.


Subject(s)
Cell Cycle/drug effects , Cell Proliferation/drug effects , Growth Inhibitors/pharmacology , PPAR delta/antagonists & inhibitors , Thiazoles/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Cyclin A/genetics , Cyclin A/metabolism , Cyclin D/genetics , Cyclin D/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Dose-Response Relationship, Drug , Growth Inhibitors/chemistry , Humans , Inhibitory Concentration 50 , PPAR delta/genetics , PPAR delta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thiazoles/chemistry
16.
J Pharmacol Exp Ther ; 329(3): 1178-86, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19252061

ABSTRACT

The orexigenic peptide ghrelin has been shown to have prokinetic activity in the gastrointestinal (GI) system of several species, including humans. In this series of experiments, we have evaluated the prokinetic activity of novel, small-molecule ghrelin receptor (GhrR) agonists after parenteral and peroral dosing in mice and rats. Gastric emptying, small intestinal transport, and fecal output were determined after intraperitoneal and intracerebroventricular dosing of GhrR agonists, using ghrelin as a positive control. These same parameters were evaluated after oral gavage dosing of the synthetic agonists. Regardless of dose route, GhrR agonist treatment increased gastric emptying, small intestinal transit, and fecal output. However, fecal output was only increased by GhrR agonist treatment if mice were able to feed during the stimulatory period. Thus, GhrR agonists can stimulate upper GI motility, and the orexigenic action of the compounds can indirectly contribute to prokinetic activity along the entire GI tract. The orexigenic and prokinetic effects of either ghrelin or small-molecule GhrR agonists were selective for the GhrR because they were absent when evaluated in GhrR knockout mice. We next evaluated the efficacy of the synthetic GhrR agonists dosed in a model of opiate-induced bowel dysfunction induced by a single injection of morphine. Oral dosing of a GhrR agonist normalized GI motility in opiate-induced dysmotility. These data demonstrate the potential utility of GhrR agonists for treating gastrointestinal hypomotility disorders.


Subject(s)
Gastrointestinal Motility/drug effects , Ghrelin/administration & dosage , Ghrelin/pharmacology , Peptide Hormones/administration & dosage , Peptide Hormones/pharmacology , Receptors, Ghrelin/agonists , Administration, Oral , Animals , Body Weight/drug effects , Bowen's Disease/chemically induced , Bowen's Disease/drug therapy , Bowen's Disease/physiopathology , Central Nervous System/drug effects , Defecation/drug effects , Eating/drug effects , Gastric Emptying/drug effects , Gastrointestinal Transit/drug effects , Intestine, Small/drug effects , Intestine, Small/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/pharmacology , Peptide Hormones/blood , Rats , Rats, Sprague-Dawley , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism
17.
Bioorg Med Chem Lett ; 18(18): 5083-6, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18722770

ABSTRACT

The discovery and optimization of a novel series of prolinol-derived GHSR agonists is described. This series emerged from a 11,520-member solid-phase library targeting the GPCR protein superfamily, and the rapid optimization of low micromolar hits into single-digit nanomolar leads can be attributed to the solid-phase synthesis of matrix libraries, which revealed multiple non-additive structure-activity relationships. In addition, the separation of potent diastereomers highlighted the influence of the alpha-methyl stereochemistry of the phenoxyacetamide sidechain on GHSR activity.


Subject(s)
Pyrrolidines/chemical synthesis , Pyrrolidines/pharmacology , Receptors, G-Protein-Coupled/drug effects , Receptors, Ghrelin/agonists , Combinatorial Chemistry Techniques , Molecular Structure , Pyrrolidines/chemistry , Stereoisomerism , Structure-Activity Relationship
18.
Bioorg Med Chem Lett ; 18(14): 4072-4, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18554903

ABSTRACT

The first enantioselective synthesis of (D)-2-tert-butoxycarbonylamino-5,5-difluoro-5-phenyl-pentanoic acid 3 was achieved. The incorporation of the titled compound into growth hormone secretagogue (GHS) compounds resulted in new analogs 10 and 16, both of which had significantly increased in vitro potency. The compound 10 also showed improved in vivo efficacy as well as pharmacokinetic properties in rat models.


Subject(s)
Growth Hormone/metabolism , Pentanoic Acids/chemical synthesis , Administration, Oral , Animals , Area Under Curve , Biological Availability , Carbamates/pharmacology , Chemistry, Pharmaceutical/methods , Drug Design , Indoles/pharmacology , Models, Chemical , Pentanoic Acids/chemistry , Pentanoic Acids/pharmacology , Peptide Hormones/chemistry , Rats , Spiro Compounds/pharmacology , Stereoisomerism , Tetrazoles/pharmacology
19.
Bioorg Med Chem Lett ; 18(8): 2536-9, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18378446

ABSTRACT

A novel series of N1 substituted tetrazole amides were prepared and showed to be potent growth hormone (GH) secretagogues. Among them, hydroxyl containing analog 31 displayed excellent in vivo activity by increasing plasma GH 10-fold in an anesthetized IV rat model.


Subject(s)
Amides/chemical synthesis , Amides/pharmacology , Growth Hormone/metabolism , Tetrazoles/chemistry , Amides/chemistry , Animals , Cell Line , Glioma/metabolism , Growth Hormone/blood , Molecular Structure , Rats , Structure-Activity Relationship
20.
Mol Cancer Res ; 6(3): 483-90, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18337454

ABSTRACT

Mammalian metallothionein (MT) genes are transcriptionally activated by the essential metal zinc as well as by environmental stresses, including toxic metal overload and redox fluctuations. In addition to playing a key role in zinc homeostasis, MT proteins can protect against metal- and oxidant-induced cellular damage, and may participate in other fundamental physiologic and pathologic processes such as cell survival, proliferation, and neoplasia. Previously, our group reported a requirement for metal-responsive transcription factor-1 (MTF-1) in hypoxia-induced transcription of mouse MT-I and human MT-IIA genes. Here, we provide evidence that the protumorigenic hypoxia-inducible transcription factor-1alpha (HIF-1alpha) is essential for induction of MT-1 by hypoxia, but not zinc. Chromatin immunoprecipitation assays revealed that MTF-1 and HIF-1alpha are both recruited to the mouse MT-I promoter in response to hypoxia, but not zinc. In the absence of HIF-1alpha, MTF-1 is recruited to the MT-I promoter but fails to activate MT-I gene expression in response to hypoxia. Thus, HIF-1alpha seems to function as a coactivator of MT-I gene transcription by interacting with MTF-1 during hypoxia. Coimmunoprecipitation studies suggest interaction between MTF-1 and HIF-1alpha, either directly or as mediated by other factors. It is proposed that association of these important transcription factors in a multiprotein complex represents a common strategy to control unique sets of hypoxia-inducible genes in both normal and diseased tissue.


Subject(s)
Cell Hypoxia/physiology , DNA-Binding Proteins/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Metallothionein/genetics , Transcription Factors/physiology , 3T3 Cells , Animals , Cell Hypoxia/drug effects , Cell Line, Transformed , Gene Deletion , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Metallothionein/biosynthesis , Mice , RNA Interference , Zinc/pharmacology , Transcription Factor MTF-1
SELECTION OF CITATIONS
SEARCH DETAIL