Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem ; 93: 117462, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37683572

ABSTRACT

Enteropeptidase is located in the duodenum that involved in intestinal protein digestion. We have reported enteropeptidase inhibitors with low systemic exposure. The aim of this study was to discover novel enteropeptidase inhibitors showing more potent in vivo efficacy while retaining low systemic exposure. Inhibitory mechanism-based drug design led us to cyclize ester 2 to medium-sized lactones, showing potent enteropeptidase inhibitory activity and improving the ester stability, thus increasing fecal protein output in vivo. Optimization on the linker between two benzene rings resulted in discovery of ether lactone 6b, exhibiting further enhanced enteropeptidase inhibitory activity and long duration of inhibitory state. Oral administration of 6b in mice significantly elevated fecal protein output compared with the lead 2. In addition, 6b showed low systemic exposure along with low intestinal absorption. Furthermore, we identified the 10-membered lactonization method for scale-up synthesis of 6b, which does not require high-dilution conditions.


Subject(s)
Drug Design , Enteropeptidase , Animals , Mice , Administration, Oral , Esters , Ethers , Lactones/pharmacology
2.
J Med Chem ; 65(12): 8456-8477, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35686954

ABSTRACT

To discover a novel series of potent inhibitors of enteropeptidase, a membrane-bound serine protease localized to the duodenal brush border, 4-guanidinobenzoate derivatives were evaluated with minimal systemic exposure. The 1c docking model enabled the installation of an additional carboxylic acid moiety to obtain an extra interaction with enteropeptidase, yielding 2a. The oral administration of 2a significantly elevated the fecal protein output, a pharmacodynamic marker, in diet-induced obese (DIO) mice, whereas subcutaneous administration did not change this parameter. Thus, systemic exposure of 2a was not required for its pharmacological effects. Further optimization focusing on the in vitro IC50 value and T1/2, an indicator of dissociation time, followed by enhanced in vivo pharmacological activity based on the ester stability of the compounds, revealed two series of potent enteropeptidase inhibitors, a dihydrobenzofuran analogue ((S)-5b, SCO-792) and phenylisoxazoline (6b), which exhibited potent anti-obesity effects despite their low systemic exposure following their oral administration to DIO rats.


Subject(s)
Enteropeptidase , Obesity , Animals , Benzoates , Enteropeptidase/metabolism , Guanidines/pharmacology , Guanidines/therapeutic use , Mice , Mice, Obese , Obesity/drug therapy , Obesity/metabolism , Rats
3.
Bioorg Med Chem ; 41: 116208, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34010766

ABSTRACT

We undertook an optimization effort involving propan-2-yl 4-({6-[5-(methanesulfonyl)-2,3-dihydro-1H-indol-1-yl]pyrimidin-4-yl}oxy)piperidine-1-carboxylate 1, which we had previously discovered as a novel G protein-coupled receptor 119 (GPR119) agonist. To occupy a presumed hydrophobic space between the pyrimidine and piperidine rings in interaction with GPR119, we replaced the linker oxygen with nitrogen. Subsequently, the introduction of a substituent at the bridging nitrogen atom was explored. We found that the installation of N-trifluoromethyl group 10 not only enhanced GPR119 agonist activity but also considerably improved the human ether-à-go-go-related gene (hERG) inhibition profile. These improvements were not observed for non-fluorinated substituents, such as ethyl analog 8b. The next optimization effort focused on the exploration of a new surrogate structure for the indoline ring and the isosteric replacements of the piperidine N-Boc group to improve solubility, metabolic stability, and oral bioavailability. As a result, N-{1-[3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl]piperidin-4-yl}-6-{[1-(methanesulfonyl)piperidin-4-yl]oxy}-N-(trifluoromethyl)pyrimidin-4-amine (27) was identified as a potent and orally bioavailable GPR119 agonist. This compound augmented insulin secretion and effectively lowered plasma glucose excursion in a diabetic animal model after oral administration. In this study, we discuss the designs, syntheses, and biological activities of a novel series of N-(piperidin-4-yl)-N-(trifluoromethyl)pyrimidin-4-amine derivatives as GPR119 agonists, and to determine the distinctive effect of the N-trifluoromethyl group on hERG inhibition, we also discuss the conformational preference of representative compounds.


Subject(s)
Amines/chemistry , Amines/pharmacology , Gene Expression Regulation/drug effects , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Area Under Curve , Blood Glucose , Diabetes Mellitus, Experimental/drug therapy , Drug Design , Drug Discovery , Insulin/metabolism , Molecular Structure , Rats , Receptors, G-Protein-Coupled/genetics
4.
FEBS Open Bio ; 10(3): 316-326, 2020 03.
Article in English | MEDLINE | ID: mdl-31837122

ABSTRACT

Monoacylglycerol O-acyltransferase 2 (MGAT2) is one of the key enzymes responsible for triglyceride (TG) re-synthesis in the small intestine. We have previously demonstrated that pharmacological inhibition of MGAT2 has beneficial effects on obesity and metabolic disorders in mice. Here, we further investigate the effects of MGAT2 inhibition on (a) fat-induced gut peptide release and fat intake in normal mice and (b) metabolic disorders in high-fat diet (HFD)-fed ob/ob mice, a model of severe obesity and type 2 diabetes mellitus, using an orally bioavailable MGAT2 inhibitor Compound B (CpdB). CpdB inhibited elevation of plasma TG in mice challenged with an oil-supplemented liquid meal. Oil challenge stimulated the secretion of two gut anorectic hormones (peptide tyrosine-tyrosine and glucagon-like peptide-1) into the bloodstream, and these responses were augmented in mice pretreated with CpdB. In a two-choice test using an HFD and a low-fat diet, CpdB selectively inhibited intake of the HFD in normal mice. Administration of CpdB to HFD-fed ob/ob mice for 5 weeks suppressed food intake and body weight gain and inhibited elevation of glycated hemoglobin. These results indicate that pharmacological MGAT2 inhibition modulates fat-induced gut peptide release and fat intake in normal mice and improves obesity and diabetes in HFD-fed ob/ob mice and thus may have potential for development into a treatment of obesity and its related metabolic diseases.


Subject(s)
Acyltransferases/genetics , Acyltransferases/metabolism , Obesity/metabolism , Acyltransferases/antagonists & inhibitors , Animals , Diet, High-Fat/adverse effects , Dietary Fats/metabolism , Dipeptides/pharmacology , Dipeptides/therapeutic use , Eating/drug effects , Eating/physiology , Energy Metabolism , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide 1/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/genetics , Obesity/physiopathology , Triglycerides/metabolism , Weight Gain
5.
PLoS One ; 11(3): e0150976, 2016.
Article in English | MEDLINE | ID: mdl-26938273

ABSTRACT

Monoacylglycerol O-acyltransferase 2 (MGAT2) catalyzes the synthesis of diacylglycerol (DG), a triacylglycerol precursor and potential peripheral target for novel anti-obesity therapeutics. High-throughput screening identified lead compounds with MGAT2 inhibitory activity. Through structural modification, a potent, selective, and orally bioavailable MGAT2 inhibitor, compound A (compA), was discovered. CompA dose-dependently inhibited postprandial increases in plasma triglyceride (TG) levels. Metabolic flux analysis revealed that compA inhibited triglyceride/diacylglycerol resynthesis in the small intestine and increased free fatty acid and acyl-carnitine with shorter acyl chains than originally labelled fatty acid. CompA decreased high-fat diet (HFD) intake in C57BL/6J mice. MGAT2-null mice showed a similar phenotype as compA-treated mice and compA did not suppress a food intake in MGAT2 KO mice, indicating that the anorectic effects were dependent on MGAT2 inhibition. Chronic administration of compA significantly prevented body weight gain and fat accumulation in mice fed HFD. MGAT2 inhibition by CompA under severe diabetes ameliorated hyperglycemia and fatty liver in HFD-streptozotocin (STZ)-treated mice. Homeostatic model assessments (HOMA-IR) revealed that compA treatment significantly improved insulin sensitivity. The proximal half of the small intestine displayed weight gain following compA treatment. A similar phenomenon has been observed in Roux-en-Y gastric bypass-treated animals and some studies have reported that this intestinal remodeling is essential to the anti-diabetic effects of bariatric surgery. These results clearly demonstrated that MGAT2 inhibition improved dyslipidemia, obesity, and diabetes, suggesting that compA is an effective therapeutic for obesity-related metabolic disorders.


Subject(s)
Anti-Obesity Agents/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Enzyme Inhibitors/pharmacology , Hyperlipidemias/drug therapy , Hypoglycemic Agents/pharmacology , Indoles/pharmacology , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Obesity/drug therapy , Sulfonamides/pharmacology , Animals , Anti-Obesity Agents/chemical synthesis , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat , Dietary Fats/metabolism , Diglycerides/antagonists & inhibitors , Diglycerides/biosynthesis , Enzyme Inhibitors/chemical synthesis , Fasting , Gene Expression , High-Throughput Screening Assays , Hyperlipidemias/enzymology , Hyperlipidemias/pathology , Hypoglycemic Agents/chemical synthesis , Indoles/chemical synthesis , Insulin Resistance , Intestine, Small/drug effects , Intestine, Small/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Obesity/enzymology , Obesity/pathology , Streptozocin , Sulfonamides/chemical synthesis , Triglycerides/antagonists & inhibitors , Triglycerides/biosynthesis , Weight Gain/drug effects
6.
Bioorg Med Chem ; 23(15): 4544-4560, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26100443

ABSTRACT

Acyl CoA:monoacylglycerol acyltransferase-2 (MGAT2) has emerged as a potential peripheral target for the treatment of obesity and metabolic disorders. We previously identified a novel series of N-phenylindoline-5-sulfonamide derivatives exemplified by 2 as potent and orally bioavailable MGAT2 inhibitors. Despite its attractive potency, further assessment revealed that this compound exhibited time-dependent inhibition (TDI) of cytochrome P450 3A4 (CYP3A4). To remove the undesirable CYP3A4 TDI activity, structural modification was focused on the 2,4-difluoroaniline moiety on the basis of the assumption that this moiety would be involved in mechanism-based inhibition of CYP3A4 via oxidative metabolism. This led to the finding that the introduction of 4-chloro-2,6-difluoroaniline significantly improved CYP3A4 TDI risk. Further optimization resulted in the discovery of N-(4-chloro-2,6-difluorophenyl)-1-{5-[1-methyl-3-(trifluoromethyl)-1H-pyrazol-5-yl]pyrimidin-2-yl}-7-(2-oxopyrrolidin-1-yl)-2,3-dihydro-1H-indole-5-sulfonamide (27c) with potent MGAT2 inhibitory activity (IC50=7.8 nM) and excellent ADME-Tox profiles including metabolic stability, oral bioavailability, and CYP3A4 TDI. In a mouse oral fat tolerance test, compound 27c effectively and dose-dependently suppressed the elevation of plasma triacylglycerol levels after oral administration at doses of 1 and 3mg/kg. We also discuss mitigation of the phototoxic liability of biaryl derivatives on the basis of the HOMO-LUMO gap hypothesis during the course of optimization efforts.


Subject(s)
Acyltransferases/antagonists & inhibitors , Cytochrome P-450 CYP3A/drug effects , Enzyme Inhibitors/pharmacology , Animals , Enzyme Inhibitors/chemistry , Mice
7.
J Med Chem ; 58(9): 3892-909, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25897973

ABSTRACT

Acyl CoA:monoacylglycerol acyltransferase-2 (MGAT2) has attracted interest as a novel target for the treatment of obesity and metabolic diseases. Starting from N-phenylbenzenesulfonamide derivative 1 with moderate potency for MGAT2 inhibition, we explored an effective location of the hydrophobic group at the 1-position to enhance MGAT2 inhibitory activity. Shifting the hydrophobic group to the adjacent position followed by introduction of a bicyclic central core to restrict the substituent orientation produced N-phenylindoline-5-sulfonamide derivative 10b, which displayed much improved potency, with an IC50 value of 1.0 nM. This compound also exhibited excellent selectivity (greater than 30,000-fold) against related acyltransferases (MGAT3, DGAT1, DGAT2, and ACAT1). Subsequent optimization efforts were directed toward improving pharmacokinetic profiles, which resulted in the identification of 5-[(2,4-difluorophenyl)sulfamoyl]-7-(2-oxopyrrolidin-1-yl)-N-[4-(trifluoromethyl)phenyl]-2,3-dihydro-1H-indole-1-carboxamide (24d) endowed with potent MGAT2 inhibitory activity (IC50 = 3.4 nM) and high oral bioavailability (F = 52%, mouse). In a mouse oral fat tolerance test, oral administration of this compound effectively suppressed the elevation of plasma triacylglycerol levels.


Subject(s)
Acyltransferases/antagonists & inhibitors , Indoles/chemistry , Sulfonamides/chemistry , Administration, Oral , Animals , Benzimidazoles/chemistry , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Benzoxazoles/chemistry , Benzoxazoles/pharmacokinetics , Benzoxazoles/pharmacology , Biological Availability , Cell Line , Humans , Indazoles/chemistry , Indazoles/pharmacokinetics , Indazoles/pharmacology , Indoles/pharmacokinetics , Indoles/pharmacology , Male , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Triglycerides/blood
8.
J Med Chem ; 54(14): 4998-5012, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21657270

ABSTRACT

We previously discovered an orally active human gonadotropin-releasing hormone (GnRH) receptor antagonist, thieno[2,3-d]pyrimidine-2,4-dione derivative 1 (sufugolix). To reduce the cytochrome P450 (CYP) inhibitory activity and improve in vivo GnRH antagonistic activity, further optimization of this scaffold was carried out. We focused our synthetic efforts on chemical modification at the 5 and 3 positions of the thieno[2,3-d]pyrimidine-2,4-dione ring based on computational modeling, which resulted in the discovery of 1-{4-[1-(2,6-difluorobenzyl)-5-[(dimethylamino)methyl]-3-(6-methoxypyridazin-3-yl)-2,4-dioxo-1,2,3,4-tetrahydrothieno[2,3-d]pyrimidin-6-yl]phenyl}-3-methoxyurea (16b) as a highly potent and orally active GnRH antagonist. Compound 16b showed potent in vitro GnRH antagonistic activity in the presence of fetal bovine serum (FBS) without CYP inhibition. Oral administration of 16b maintained the suppressive effect of the plasma luteinizing hormone levels in castrated cynomolgus monkeys at a 3 mg/kg dose for more than 24 h. Compound 16b is currently under clinical development with the code name of TAK-385.


Subject(s)
Phenylurea Compounds/chemical synthesis , Pyrimidinones/chemical synthesis , Receptors, LHRH/antagonists & inhibitors , Administration, Oral , Animals , CHO Cells , Cattle , Cricetinae , Cricetulus , Cytochrome P-450 CYP3A Inhibitors , Humans , Macaca fascicularis , Male , Models, Molecular , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/pharmacology , Pyrimidinones/pharmacokinetics , Pyrimidinones/pharmacology , Radioligand Assay , Rats , Species Specificity , Structure-Activity Relationship
9.
Bioorg Med Chem ; 16(7): 3941-58, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18272372

ABSTRACT

In order to develop an anti-sepsis agent, a series of cyclohexene derivatives were synthesized and evaluated for their biological activities. Through modification of the sulfonamide spacer moiety depicted by formula II, it was found that the benzylsulfone derivative 10a had potent inhibitory activity against the production of NO. Further modifications of the phenyl ring, ester moiety, and benzyl position of benzylsulfone derivatives III were carried out. Among these compounds, (R)-(+)-10a and (6R, 1S)-(+)-22a showed strong inhibitory activity not only against NO production but also against inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) in vitro. Furthermore, (R)-(+)-10a and (6R, 1S)-(+)-22a protected mice from LPS-induced lethality in a dose-dependent manner.


Subject(s)
Cyclohexenes/chemical synthesis , Cyclohexenes/therapeutic use , Interleukin-6/biosynthesis , Nitric Oxide/biosynthesis , Sepsis/drug therapy , Sepsis/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Line , Chemical Phenomena , Chemistry, Physical , Chromatography, High Pressure Liquid , Cross-Linking Reagents/chemistry , Crystallography, X-Ray , Cyclohexenes/chemistry , Drug Design , Lipopolysaccharides/pharmacology , Mice , Models, Molecular , Molecular Structure , Sepsis/chemically induced , Stereoisomerism , Structure-Activity Relationship
10.
Eur J Pharmacol ; 584(1): 40-8, 2008 Apr 14.
Article in English | MEDLINE | ID: mdl-18299127

ABSTRACT

TAK-242, a small-molecule antisepsis agent, has shown to suppress lipopolysaccharide (LPS)-induced inflammation. In this study, we demonstrate that TAK-242 is a selective inhibitor of Toll-like receptor (TLR)-4 signaling. TAK-242 almost completely suppressed production of nitric oxide (NO) or tumor necrosis factor (TNF)-alpha induced by a TLR4-specific ligand, ultra-pure LPS, in mouse RAW264.7, human U-937 and P31/FUJ cells, whereas this agent showed little effect on other TLR ligands, Pam(3)CSK(4) (TLR1/2), peptidoglycan (TLR2/6), double strand RNA (TLR3), R-848 (TLR7) and CpG oligonucleotide (TLR9). Furthermore, TAK-242 potently inhibited nuclear factor (NF)-kappaB activation induced by ultra-pure LPS in HEK293 cells transiently expressing TLR4 and co-receptors, myeloid differentiation protein-2 (MD2) and CD14, whereas this agent showed little effect on other TLRs, TLR1/2, TLR2/6, TLR3, TLR5, TLR7 and TLR9. TAK-242 also inhibited ligand-independent NF-kappaB activation resulting from over-expression of TLR4. Although chimera receptors, which are consist of the extracellular domain of CD4 and the intracellular domain of human or mouse TLR4, showed constitutive NF-kappaB activation, TAK-242 potently inhibited the signaling from CD4-TLR4 chimera receptors. In contrast, the NF-kappaB activation mediated by TLR4 adaptors, myeloid differentiation factor 88 (MyD88), TIR-associated protein (TIRAP), Toll/IL-1R homology (TIR)-domain-containing adaptor protein-inducing interferon-beta (TRIF) or TRIF-related adaptor molecule (TRAM) was not affected by TAK-242. TAK-242 is therefore a selective inhibitor of signaling from the intracellular domain of TLR4 and represents a novel therapeutic approach to the treatment of TLR4-mediated diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Macrophages/drug effects , Signal Transduction/drug effects , Sulfonamides/pharmacology , Toll-Like Receptor 4/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Animals , CD4 Antigens/drug effects , CD4 Antigens/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Humans , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Toll-Like Receptor 4/chemistry , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Transfection , Tumor Necrosis Factor-alpha/metabolism , U937 Cells
11.
Eur J Pharmacol ; 571(2-3): 231-9, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17632100

ABSTRACT

Ethyl (6R)-6-[N-(2-chloro-4-fluorophenyl)sulfamoyl]cyclohex-1-ene-1-carboxylate (TAK-242), a novel small molecule that selectively inhibits Toll-like receptor 4-mediated signaling, inhibits various kinds of inflammatory mediators such as nitric oxide (NO), tumor necrosis factor (TNF)-alpha, interleukin (IL)-1, IL-6, IL-10, macrophage inhibitory protein (MIP)-2 and prostaglandin E2 from lipopolysaccharide (LPS)-stimulated macrophages. The effects of TAK-242 were evaluated in a mouse model of endotoxin shock. Intravenous administration of TAK-242 to mice 1 h before LPS challenge dose-dependently inhibited LPS-induced increases in serum levels of TNF-alpha, IL-1beta, IL-6, IL-10, MIP-2, and NO metabolites. TAK-242 protected mice from LPS-induced lethality in a similar dose-dependent manner, and rescued 100% of mice at a dose of 1 mg/kg. Interestingly, TAK-242 worked quickly, and showed beneficial effects even when administered after LPS challenge. Even though increases in serum levels of IL-6 and hypothermia were already evident 2 h after LPS challenge, TAK-242 administration inhibited further increase in IL-6 levels and decrease in body temperature. LPS-induced increases in serum levels of organ dysfunction markers, such as alanine aminotransferase, total bilirubin, and blood urea nitrogen, were also significantly suppressed by post-treatment as well as pre-treatment. Furthermore, administration of 3 mg/kg TAK-242 significantly increased survival of mice, even when given 4 h after LPS challenge. These results suggest that TAK-242 protects mice against LPS-induced lethality by inhibiting production of multiple cytokines and NO. TAK-242 has a quick onset of action and provides significant benefits by post-treatment, suggesting that it may be a promising drug candidate for the treatment of sepsis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Shock, Septic/drug therapy , Signal Transduction/drug effects , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/therapeutic use , Body Temperature/drug effects , Chemokine CXCL2 , Chemokines/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Interleukins/blood , Lipopolysaccharides , Mice , Mice, Inbred BALB C , Multiple Organ Failure/etiology , Multiple Organ Failure/prevention & control , Nitric Oxide/metabolism , Peptidoglycan , Shock, Septic/chemically induced , Shock, Septic/complications , Shock, Septic/metabolism , Shock, Septic/physiopathology , Staphylococcus aureus , Sulfonamides/therapeutic use , Time Factors , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/blood
12.
Chem Pharm Bull (Tokyo) ; 54(1): 58-62, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16394550

ABSTRACT

Two new synthetic methods were established for the efficient synthesis of optically active cyclohexene antisepsis agent, ethyl (6R)-6-[N-(2-chloro-4-fluorophenyl)sulfamoyl]cyclohex-1-ene-1-carboxylate [(R)-1: TAK-242)]. The first method involved recrystallization from methanol of the diastereomeric mixture (6RS,1'R)-7, obtained by esterification of carboxylic acid 3 with (S)-1-(4-nitrophenyl)ethanol [(S)-5)] to give the desired isomer (6R,1'R)-7 with 99% de in 32% yield. Subsequent catalytic hydrogenolysis and esterification gave (R)-1 with >99% ee. The second method employed enantioselective hydrolysis of acetoxymethyl ester 9a (prepared by alkylation of 3 with bromomethyl acetate) with Lipase PS-D to give the eutomeric enantiomer (R)-9a with excellent enantioselectivity (>99% ee) and high yield (48%). The desired (R)-1 was then obtained by transesterification with ethanol in the presence of concentrated sulfuric acid without loss of ee. Of these, the procedure employing enzymatic kinetic resolution using Lipase PS-D is the more efficient and practical preparation of (R)-1.


Subject(s)
Sepsis/drug therapy , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology , Catalysis , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Indicators and Reagents , Lipase/chemistry , Magnetic Resonance Spectroscopy , Spectrophotometry, Infrared
13.
Mol Pharmacol ; 69(4): 1288-95, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16373689

ABSTRACT

Proinflammatory mediators such as cytokines and NO play pivotal roles in various inflammatory diseases. To combat inflammatory diseases successfully, regulation of proinflammatory mediator production would be a critical process. In the present study, we investigated the in vitro effects of ethyl (6R)-6-[N-(2-chloro-4-fluorophenyl)sulfamoyl]cyclohex-1-ene-1-carboxylate (TAK-242), a novel small molecule cytokine production inhibitor, and its mechanism of action. In RAW264.7 cells and mouse peritoneal macrophages, TAK-242 suppressed lipopolysaccharide (LPS)-induced production of NO, tumor necrosis factor-alpha (TNF-alpha), and interleukin (IL)-6, with 50% inhibitory concentration (IC50) of 1.1 to 11 nM. TAK-242 also suppressed the production of these cytokines from LPS-stimulated human peripheral blood mononuclear cells (PBMCs) at IC50 values from 11 to 33 nM. In addition, the inhibitory effects on the LPS-induced IL-6 and IL-12 production were similar in human PBMCs, monocytes, and macrophages. TAK-242 inhibited mRNA expression of IL-6 and TNF-alpha induced by LPS and interferon-gamma in RAW264.7 cells. The phosphorylation of mitogen-activated protein kinases induced by LPS was also inhibited in a concentration-dependent manner. However, TAK-242 did not antagonize the binding of LPS to the cells. It is noteworthy that TAK-242 suppressed the cytokine production induced by Toll-like receptor (TLR) 4 ligands, but not by ligands for TLR2, -3, and -9. In addition, IL-1beta-induced IL-8 production from human PBMCs was not markedly affected by TAK-242. These data suggest that TAK-242 suppresses the production of multiple cytokines by selectively inhibiting TLR4 intracellular signaling. Finally, TAK-242 is a novel small molecule TLR4 signaling inhibitor and could be a promising therapeutic agent for inflammatory diseases, whose pathogenesis involves TLR4.


Subject(s)
Cytokines/biosynthesis , Signal Transduction/drug effects , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Blotting, Western , Cell Line , Enzyme Activation , MAP Kinase Signaling System , Mice , Nitric Oxide/biosynthesis , Polymerase Chain Reaction , Toll-Like Receptor 4/physiology
14.
J Med Chem ; 48(23): 7457-67, 2005 Nov 17.
Article in English | MEDLINE | ID: mdl-16279805

ABSTRACT

To develop a new therapeutic agent for sepsis, screening of the Takeda chemical library was carried out using mouse macrophages stimulated with lipopolysaccharide (LPS) to identify a new class of small-molecule inhibitors of inflammatory mediator production. The lead compound 5a was discovered, from which a series of novel cyclohexene derivatives I bearing a sulfamoyl and ester group were designed, synthesized and tested for their inhibitory activity against nitric oxide (NO) production. Derivatives I were synthesized by the coupling of sulfonyl chlorides and anilines with concomitant double bond migration in the presence of triethylamine, and phenyl ring substitution and modification of the ester and cyclohexene moieties were carried out. Among the compounds synthesized, ethyl (6R)-6-[N-(2-chloro-4-fluorophenyl)sulfamoyl]cyclohex-1-ene-1-carboxylate [(R)-(+)-5n, TAK-242] was found to exhibit the most potent suppressive activity for the production of not only NO but also inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) induced by LPS-stimulated mouse macrophages with IC50 values of 1.8, 1.9 and 1.3 nM, respectively. It shows marked beneficial effects in vivo also. Intravenous administration of (R)-(+)-5n at doses of 0.1 mg/kg or more suppressed the production of NO and various cytokines [TNF-alpha, IL-6 and IL-1beta] in the mouse endotoxin shock model. Furthermore, it protected mice from death dose-dependently and all mice survived at a dose of 3 mg/kg. The minimum effective dose to protect mice from lethality in this model was 0.3 mg/kg, which was consistent with those for inhibitory effects on the production of NO and cytokines. Compound (R)-(+)-5n is currently undergoing clinical trials for the treatment of sepsis.


Subject(s)
Cytokines/antagonists & inhibitors , Nitric Oxide/antagonists & inhibitors , Sepsis/prevention & control , Sulfonamides/chemical synthesis , Animals , Cell Line , Crystallography, X-Ray , Cytokines/biosynthesis , Female , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Molecular Structure , Nitric Oxide/biosynthesis , Sepsis/metabolism , Sepsis/mortality , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology
15.
Antimicrob Agents Chemother ; 46(5): 1388-93, 2002 May.
Article in English | MEDLINE | ID: mdl-11959573

ABSTRACT

TAK-456 is a novel oral triazole compound with potent and broad-spectrum in vitro antifungal activity and strong in vivo efficacy against Candida albicans and Aspergillus fumigatus. TAK-456 inhibited sterol synthesis of C. albicans and A. fumigatus by 50% at 3 to 11 ng/ml. TAK-456 showed strong in vitro activity against clinical isolates of Candida spp., Aspergillus spp., and Cryptococcus neoformans, except for Candida glabrata. The MICs at which 90% of the isolates tested were inhibited byTAK-456, fluconazole, itraconazole, voriconazole, and amphotericin B were 0.25, 4, 0.5, 0.13, and 0.5 microg/ml, respectively, for clinical isolates of C. albicans and 1, >64, 0.5, 0.5, and 0.5 microg/ml, respectively, for clinical isolates of A. fumigatus. Therapeutic activities of TAK-456 and reference triazoles against systemic lethal infections caused by C. albicans and A. fumigatus in mice were investigated by orally administering drugs once daily for 5 days, and efficacies of the compounds were evaluated by the prolongation of survival. In normal mice, TAK-456 and fluconazole were effective against infection caused by fluconazole-susceptible C. albicans at a dose of 1 mg/kg. In transiently neutropenic mice, therapeutic activity of TAK-456 at 1 mg/kg of body weight against infection with the same strain was stronger than those at 1 mg/kg of fluconazole. TAK-456 was effective against infections with two strains of fluconazole-resistant C. albicans at a dose of 10 mg/kg. TAK-456 also expressed activities similar to or higher than those of itraconazole against the infections caused by two strains of A. fumigatus in neutropenic mice at a dose of 10 mg/kg. These results suggest that TAK-456 is a promising candidate for development for the treatment of candidiasis and aspergillosis in humans.


Subject(s)
Antifungal Agents/pharmacology , Aspergillosis/drug therapy , Candidiasis/drug therapy , Fungi/drug effects , Imidazoles/pharmacology , Tetrazoles/pharmacology , Animals , Antifungal Agents/pharmacokinetics , Antifungal Agents/therapeutic use , Aspergillosis/microbiology , Aspergillus fumigatus/drug effects , Candida albicans/drug effects , Candidiasis/microbiology , Cryptococcus neoformans/drug effects , Female , Imidazoles/pharmacokinetics , Imidazoles/therapeutic use , Mice , Microbial Sensitivity Tests , Tetrazoles/pharmacokinetics , Tetrazoles/therapeutic use
16.
Antimicrob Agents Chemother ; 46(2): 283-7, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11796331

ABSTRACT

TAK-457 is an injectable prodrug of TAK-456, which is a novel oral triazole compound with potent antifungal activity. The in vivo efficacy of TAK-457 was evaluated in two models of invasive pulmonary aspergillosis with CDF(1) mice and CBA/J mice with transient neutropenia induced by cyclophosphamide. Against the infection in CDF(1) mice, treatment with 10 mg of TAK-457 and 1 mg of amphotericin B/kg reduced the fungal burden in lungs and rescued all mice. In the infection model with CBA/J mice, TAK-457 at a dose of 10 mg/kg significantly prolonged the survival time of mice, showing significant reduction of lung chitin levels and the plasma beta-D-glucan levels. On the other hand, amphotericin B at 1 mg/kg which was a maximum tolerable dose showed slight but not significant prolongation of survival time of mice, although it also reduced the lung chitin levels and the plasma beta-D-glucan levels to a lower extent but still significantly. These results suggest that TAK-457 is a promising candidate for development for the treatment of invasive aspergillosis in humans.


Subject(s)
Antifungal Agents/therapeutic use , Aspergillosis, Allergic Bronchopulmonary/drug therapy , Imidazoles/therapeutic use , Triazoles/therapeutic use , Animals , Disease Models, Animal , Female , Injections, Intravenous , Mice , Mice, Inbred CBA , Neutropenia/etiology , Tetrazoles/therapeutic use , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...