WO2007143951A1 - COMPOSÉS DOTÉS D'UNE ACTIVITÉ D'AGONISTE DU PPARγ ET LEUR APPLICATION - Google Patents

COMPOSÉS DOTÉS D'UNE ACTIVITÉ D'AGONISTE DU PPARγ ET LEUR APPLICATION Download PDF

Info

Publication number
WO2007143951A1
WO2007143951A1 PCT/CN2007/070089 CN2007070089W WO2007143951A1 WO 2007143951 A1 WO2007143951 A1 WO 2007143951A1 CN 2007070089 W CN2007070089 W CN 2007070089W WO 2007143951 A1 WO2007143951 A1 WO 2007143951A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
butyl
compound
biphenyl
chloro
Prior art date
Application number
PCT/CN2007/070089
Other languages
English (en)
French (fr)
Inventor
Jianhui Guo
Yong Jiang
Original Assignee
Shanghai Allist Pharmaceutical., Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Allist Pharmaceutical., Inc. filed Critical Shanghai Allist Pharmaceutical., Inc.
Publication of WO2007143951A1 publication Critical patent/WO2007143951A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/68Halogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/10Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings

Definitions

  • the present invention relates to compounds having PPAR gamma agonist activity, methods for their preparation, and their clinical use in the treatment of PPAR ⁇ receptor related diseases such as diabetes or related complications. Background technique
  • Diabetes is a disease caused by a variety of genetic disorders, which plague a large part of the world.
  • Type 11 diabetes or non-insulin dependent diabetes mellitus (MDDM) type 11
  • IDDM insulin-dependent diabetes mellitus
  • MDDM non-insulin dependent diabetes mellitus
  • the plasma insulin concentration in diabetic patients is basically the same as that in normal people. However, the patient's body is resistant to insulin, which further affects the metabolism of sugar and fat in insulin-sensitive tissues, muscle, liver and adipose tissue, and the concentration of insulin in plasma in patients with type 2 diabetes is insufficient to overcome this. Resistance. 90% of people with diabetes are type 2 diabetes.
  • PPAR peroxisome proliferator-activated receptor
  • PPAR belongs to a member of the ligand-activated transcription factor-nuclear hormone receptor superfamily and is divided into three subtypes, PPAR a, PPAR Y and PPAR S.
  • PPAR and the retinoic acid X receptor (RXR) form a heterodimer and bind to a hormone response element on the target gene to activate gene expression.
  • RXR heterodimers play an important role in controlling cellular lipid homeostasis and adipocyte differentiation.
  • PPAR ⁇ plays an important regulatory role in the expression and differentiation of adipose tissue-related genes, and is also an important regulator of glucose and lipid metabolism target genes. PPAR a stimulates the proliferation of peroxidase, accelerates the oxidation of fatty acids, and thus reduces the fatty acid content in the blood. PPAR ci agonists such as Fibrates are also used to treat dyslipidemia.
  • Oral hypoglycemic drugs currently on the market mainly include insulin, sulfonylureas, biguanides, glucosidase inhibitors and thiazolidinedione (TZD) drugs.
  • TZD compounds are novel insulin sensitizers that target PPAR ⁇ , which can improve the body's sensitivity to insulin, thereby improving abnormal glucose metabolism, reducing high glucose toxicity, and not exhibiting hypoglycemia.
  • the compounds also have the advantages of preventing loss of islet cells and long duration of treatment.
  • TZD regulates the differentiation of adipocytes and increases sensitivity to insulin by activating PPAR gamma.
  • Such drugs that agonize PPAR gamma receptors such as rosiglitazone (Avandia) and pioglitazone (Austin, Actos;).
  • TZD compounds have been clinically proven It is a very effective anti-type 2 diabetes drug, but because these two drugs can cause side effects such as weight gain, edema, adipose tissue ablation, and changes in bone marrow fatty acids, it is necessary to find a new class of PPAR Y agonists.
  • the present invention discloses a class of compounds or salts thereof which selectively activate PPAR gamma.
  • the invention also discloses a process for the preparation of the compound or a salt thereof, and a clinical application of the compound as a PPAR gamma receptor related disease, such as diabetes or related complications.
  • X is a halogen, preferably fluorine or chlorine, more preferably chlorine;
  • alkyl group is a CC 4 alkyl group having 1 to 3 substituents, and the substituent is 1 to 3 groups selected from the group consisting of halogen, -OH, CC 4 alkyl, Alkoxy, CC 4 alkyl-OH, dC 4 alkoxymethyl, C 2 -C 4 ester, or sulfonate;
  • R2 includes:
  • R', R" are independently selected from substituted or unsubstituted CC 4 straight or branched alkyl groups (eg, methyl, ethyl, butyl, n-propyl, isopropyl, n-butyl, iso) Butyl, tert-butyl), wherein the substituted alkyl group is a CC 4 alkyl group having 1 to 3 substituents, and the substituent is 1-3 selected from the group consisting of Group of groups: halogen, -OH, dC 4 alkyl, dC 4 alkoxy, dC 4 alkyl-OH, dC 4 alkoxymethyl, C 2 -C 4 ester, or sulfonate;
  • OR' and OR" together with adjacent C atoms form a substituted or unsubstituted five- or six-membered ring;
  • the substituent is, for example, one to three groups selected from the group consisting of: halogen, -OH, CC 4 alkane a group, a CC 4 alkoxy group, a dC 4 alkyl-OH group, a dC 4 alkoxymethyl group, a C 2 -C 4 ester group, or a sulfonate;
  • Arl, Ar2 are each an aromatic ring (preferably a benzene ring), having no substituent on the ring or having one or more (e.g., 1-3;) substituents, wherein the substituent is 3 ⁇ 4 Alkane, an alkyl group or an alkoxy group, preferably a halogen substitution, more preferably a fluorine or chlorine substitution;
  • the alkyl group is an alkyl group which is unsubstituted or has 1-3 substituents, and the substituent is 1-3 groups selected from the group consisting of halogen, -OH, dC 4 alkyl, CC 4 alkoxy, dC 4 alkyl-OH, CC 4 alkoxymethyl, C 2 -C 4 ester, or sulfonate;
  • the alkoxy group is an alkoxy group in which CC 6 is unsubstituted or has 1-3 substituents, and the substituent is 1-3 groups selected from the group consisting of halogen, -OH, CC 4 alkyl, CC 4 alkoxy, CC 4 alkyl-OH, dC 4 alkoxymethyl, C 2 -C 4 ester, or sulfonate;
  • N :
  • R4 is hydrogen or dC 4 alkyl, preferably, the alkyl groups are unsubstituted or substituted with 1-3 substituents CC 4 alkyl, the substituent is selected from the group 1-3 Group: halogen, -OH, CC 4 alkyl, alkoxy, CC 4 alkyl-OH, dC 4 alkoxymethyl, C 2 -C 4 ester, or sulfonate, most preferably methyl.
  • R3 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • R3 is -COOR4, wherein R4 is hydrogen or methyl
  • X is chlorine.
  • R1 is n-butane.
  • R2 is -CHO; or
  • OR' and OR" together with adjacent C atoms form a substituted or unsubstituted five or six membered ring; said substituted five or six membered ring having from 1 to 3 substituents selected from the group consisting of:
  • the substituent is 1-3 groups selected from the group consisting of halogen, -OH, dC 4 alkyl, dC 4 alkoxy, dC 4 alkyl-OH, dC 4 alkoxymethyl, C 2 -C 4 ester group, or sulfonate; or
  • R 2 is a benzene ring, the ring is unsubstituted or has one or more substituents, and the substituent is fluorine or chlorine; or
  • Preferred compounds of the invention are selected from the group consisting of:
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, excipient or diluent.
  • a compound of the present invention or a pharmaceutically acceptable salt thereof for the preparation of a medicament for treating a PPAR Y receptor-related disease: a disease associated with abnormal blood glucose elevation, diabetes, a lipid disorder , metabolic syndrome, cardiovascular disease, coronary artery disease, high blood cholesterol or obesity.
  • the present invention also provides a method for treating or preventing a PPAR Y receptor-associated disease, comprising the steps of: administering to a subject in need thereof, for example, 0.05 to 200 mg/kg body weight per day of a compound of the present invention or a pharmaceutically acceptable compound thereof salt.
  • the PPAR Y receptor-related diseases include diseases associated with abnormally elevated blood glucose, diabetes, lipid disorders, metabolic syndrome, cardiovascular disease, coronary artery disease, hypercholesterolemia or obesity.
  • a method of preparing a pharmaceutical composition comprising mixing a compound of the invention, or a pharmaceutically acceptable salt thereof, with a pharmaceutically acceptable carrier to form a pharmaceutical composition.
  • the inventors have extensively and extensively studied and screened a large number of compounds, and found for the first time that the compound of the formula (I) not only has a good activation effect on the ⁇ receptor, but also has substantially no activation effect on the PPARoc receptor, and thus Compared with drugs, the side effects are small and do not cause a significant increase in body weight.
  • the present invention has been completed on the basis of this. Active ingredient
  • the term "compound of the invention” refers to a compound of formula (I).
  • the term also encompasses various crystalline forms, pharmaceutically acceptable salts, hydrates or solvates of the compounds of formula (I).
  • the compound in the present invention, can be converted into a "pharmaceutically acceptable salt" form.
  • the salt refers to a relatively non-toxic inorganic acid addition salt or an organic acid addition salt. These salts can be prepared in situ during the final isolation and purification of the compound, or by reacting the purified compound with the appropriate organic or inorganic acid in its free base form and separating the formed salt.
  • the compounds of the invention can be prepared by the following routes:
  • each substituent is as described above.
  • the imidazole aldehyde and the biphenylbenzyl bromide undergo nucleophilic substitution reaction under suitable conditions to obtain the aldehyde target compound, and the structural formula is as shown in FIG. 1; the compound of the formula 1 is reacted with a monohydric or polyhydric alcohol under suitable conditions to obtain Acetal target compound 2; compound of formula 1 is reacted with an aromatic amine Schiff base under suitable conditions to obtain imine target compound 3; under suitable conditions, compound of formula 1 is reduced to an alcohol, and the resulting alcohol
  • the substitution reaction with the 3 ⁇ 4 generation aromatic compound gives the ether target compound 4.
  • each reaction is usually carried out in an inert solvent at room temperature to reflux temperature (e.g., 0 ° C to 90 V, preferably 0 ° C to 60 ° C).
  • the reaction time is usually from 0.1 to 60 hours, preferably from 0.5 to 48 hours.
  • the compound obtained by the present invention or a salt thereof can be administered to a human, and the compound can be administered alone or in combination with other pharmaceutically acceptable compounds. It is to be noted that the compounds of the invention may be administered in admixture. Oral, rectal, parenteral (intravenous, intramuscular or subcutaneous;), topical (powder, Ointment or drops;).
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the solid dosage form usually contains 0.5 to 50% of the active ingredient, preferably 1 to 20% of the active ingredient, and most preferably 1 to 10% of the active ingredient.
  • the active compound is mixed with at least one conventional inert excipient (or carrier;) such as: (a) a filler or compatibilizer, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose, and acacia; (c) humectants, for example, glycerin; (d) disintegrants, for example, Agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) a slow solvent such as paraffin; (f) an absorption accelerator, for
  • Solid dosage forms such as tablets, troches, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other materials known in the art. They may contain opacifying agents and the release of the active compound or compound in such compositions may be released in a portion of the digestive tract in a delayed manner. If necessary, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or elixirs.
  • the liquid dosage form may contain inert diluents conventionally employed in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or a mixture of these substances.
  • inert diluents conventionally employed in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethyl
  • compositions may contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents, and flavoring agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents, and flavoring agents.
  • the suspension may contain a suspending agent, for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan ester, microcrystalline cellulose, aluminum methoxide and agar or a mixture of these and the like.
  • a suspending agent for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan ester, microcrystalline cellulose, aluminum methoxide and agar or a mixture of these and the like.
  • compositions for parenteral injection may comprise a physiologically acceptable sterile aqueous or nonaqueous solution, dispersion, suspension or emulsion, and a sterile powder for reconstitution into a sterile injectable solution or dispersion.
  • Suitable aqueous and nonaqueous vehicles, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.
  • Dosage forms for the compounds of the invention for topical administration include ointments, powders, propellants and inhalants.
  • the active ingredient is under sterile conditions with a physiologically acceptable carrier and any preservative, buffer, or Mix the propellants that may be needed if necessary.
  • therapeutically effective amount of the invention is meant an amount which is functional or active against humans and/or animals and which is acceptable to humans and/or animals.
  • a therapeutically effective amount of the composition of the invention is between 0.01 and 200 mg/kg body weight per day. Any amount within the above range is an effective amount of the invention.
  • the “therapeutically effective amount” can be used for single or combination therapy of the disease in question. Those skilled in the art will appreciate that the amount administered at the time of actual administration may be higher or lower than the above dosage range.
  • the "therapeutically effective amount” and specific treatment regimen for a subject can be affected by a number of factors, including the pharmacodynamic activity of the compound or prodrug used, the age, weight, and general Conditions, gender, diet, time of administration, susceptibility to disease, progression of the disease, and judgment of the attending physician.
  • the compounds of the present invention can be administered clinically to mammals (including humans) by oral or injection means, particularly preferably orally.
  • the dosage is 0.01 to 200 mg/kg body weight per day, and the preferred dosage is 0.01 to 100 mg/kg body weight per day.
  • the optimal dosage is 0.01 to 50 mg/kg body weight per day. Meanwhile, the optimal dose is quantified according to the individual.
  • the compound of the present invention or a salt thereof can be used for the treatment or prevention of diseases associated with the regulation of RXR and ⁇ ⁇ nuclear receptors, and can also be used for the treatment of diseases associated with abnormally elevated blood glucose.
  • the compound of the present invention or a salt thereof can be used for the treatment of diabetes, or for the treatment of lipid disorders, metabolic syndrome, cardiovascular disease, coronary artery disease, hypercholesterolemia or obesity.
  • the main advantage of the present invention is that the compounds of the present invention have partial agonistic activity on the ⁇ ⁇ receptor compared to a complete ⁇ ⁇ receptor agonist such as rosiglitazone and do not cause an increase in body weight of a diabetic animal or patient.
  • Example 3 Product 170 mg was dissolved in 30 ml of dioxane, 2 ml of a 10% NaOH solution was added, and reacted at 90 ° C for 6 h. Concentration under reduced pressure, the residue was added water to a solution, and ethyl acetate was evaporated. The aqueous phase precipitated a white solid, adjusted to pH neutral, and filtered to dryness.
  • Imidazole aldehyde 1.86 g, 10 mmol was dissolved in 15 ml of DMF, and 410 mg of 60% NaH, 2.50 g, 10 mmol of p-bromobenzyl bromide solid was allowed to react at room temperature overnight. Diluted with 300 ml of ethyl acetate, washed well with ice water to remove DMF, dried organic phase and purified by chromatography.
  • Example 9 190 mg of the compound of Example 9 was dissolved in 30 ml of dioxane, and 2.5 ml of a 10% NaOH solution was added thereto, and reacted at 90 ° C for 6 hours. Concentration under reduced pressure, the residue was added water to a solution, and ethyl acetate was evaporated. The aqueous phase precipitated as a white solid, adjusted to pH neutral, and filtered to dryness.
  • ⁇ ⁇ cDNA was cloned from adipose tissue by RT-PCR, and the amplified PCR product was inserted into the pcDNA3.1 expression vector and sequenced.
  • the reporter gene was constructed using Promega's luciferase assay vector pGL3-Promoter. Transfection experiments were performed with U2OS cells in 96-well plates. The RXR and ⁇ genes were co-transfected at the same time as the transfection of the reporter gene. After transfection for 24 hours, the compound to be tested was added and the final concentration of the solvent DMSO was maintained at 0.1. %.
  • the cells were lysed and tested for luciferase activity.
  • the intensity of activation of the nuclear receptor by the compound can be known by observing the intensity of the luminescence.
  • GFP plasmid was also co-transfected as an internal reference.
  • the luminescence values of all the test wells were corrected by GFP value in the analysis of the experimental results.
  • the test results are expressed as relative activation multiples, and the solvent control has a value of 1, and a larger value indicates a higher activation ability.
  • the activation of the receptor at 6 different concentrations was observed, which comprehensively reflected the pharmacological properties of the compound and calculated the corresponding semi-effective concentration (EC 5() ;).
  • the full-length PPAR cDNA was cloned from adipose tissue by conventional RT-PCR, and the amplified PCR product was inserted into a commercially available pcDNA3.1 expression vector and sequenced.
  • the reporter gene was constructed using Promega's luciferase assay vector pGL3-Promoter. Transfection experiments were performed with U2OS cells in 96-well plates. The RXR and PPAR a genes were co-transfected at the same time as the transfection of the reporter gene.
  • the compound to be tested was added 24 hours after transfection, and the final concentration of the solvent DMSO was maintained at 0.1. %. After 24 hours of compound action, cells were lysed and tested for luciferase activity.
  • the intensity of activation of the nuclear receptor by the compound can be known by observing the intensity of the luminescence.
  • GFP plasmid was also co-transfected as an internal reference. The luminescence values of all the test wells were corrected by GFP values in the analysis of the experimental results.
  • the test results are expressed as relative activation multiples, and the value of the solvent control is 1. The larger the value, the higher the activation ability.
  • the experimental results show that the compounds of the examples of the present invention have different degrees of activation on the ⁇ receptor, but have no activation effect on the PPARo receptor; compared with the complete ⁇ receptor agonist rosiglitazone, the present invention
  • the compound has partial agonistic activity at the PPARj receptor.
  • Healthy spontaneous type 2 diabetes animal model male GK rats, 200g, animals fasted for 12 hours, blood glucose meter to determine fasting blood glucose levels, followed by continuous intragastric administration of compounds 1-12, rosiglitazone (20 mg / kg) Or control solution 0.5% CMC (10 ml/kg), and the fasting blood glucose level was measured again after 10 days.
  • the above materials were uniformly mixed according to a conventional method, and then filled into ordinary gelatin capsules to obtain 1000 capsules.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Diabetes (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Child & Adolescent Psychology (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Description

一类具有 PPAR γ激动剂活性的化合物及其应用 技术领域
本发明涉及具有 PPAR γ激动剂活性的化合物、制备方法及其在治疗 PPAR Υ受体相关疾病, 如糖尿病或相关并发症方面的临床应用。 背景技术
糖尿病是多种基因紊乱而导致的疾病, 困扰着全球相当一部分人群。 分为 两种类型: (1)1型糖尿病或胰岛素依赖型糖尿病( IDDM;), 患者分泌极少或根 本不分泌胰岛素; (2)11型糖尿病或非胰岛素依赖型糖尿病( MDDM ),11型糖尿 病患者血浆胰岛素的浓度与正常人基本相同。 但是, 患者的机体却对胰岛素具 有抵抗作用, 因而会进一步影响糖和脂肪在对胰岛素敏感的组织即肌肉、 肝脏 和脂肪组织中的代谢, 并且 II型糖尿病患者血浆中胰岛素的浓度不足以克服这 种抵抗作用。 糖尿病患者中 90%是 II型糖尿病。
近年来发现, II 型糖尿病与过氧化物酶增殖体活化受体 (; peroxisome proliferator— activated receptor, PPAR)存在密切的关系。 PPAR属于由配体激活 的转录因子- -核激素受体超级家族中的一员,分为 3个亚型,即 PPAR a、PPAR Y和 PPAR S。 PPAR和维甲酸 X受体 (RXR)形成异二聚体, 并与目标基因上的 激素响应元件结合而激活基因表达。 PPAR / RXR异二聚体在控制细胞脂质动 态平衡和脂肪细胞分化方面起着重要作用。 PPAR γ主要在脂肪组织相关基因 的表达和分化方面起着重要的调控作用, 也是葡萄糖和脂类代谢靶基因的重要 调节因子。 PPAR a剌激过氧化物酶的增殖, 加速脂肪酸的氧化, 从而减少血 液中的脂肪酸含量, PPAR ci激动剂如 Fibrates也因此而用于治疗血脂异常。
目前市场上的口服降血糖药物主要包括胰岛素、 磺酰脲类药物、 双胍类药 物、 葡萄糖苷酶抑制剂类药物和噻唑烷二酮 (thiazolidinedione, TZD)类药物。 TZD类化合物是以 PPAR γ为作用靶点的新型的胰岛素增敏剂, 能够提高机体 对胰岛素的敏感性, 从而改善糖代谢异常, 降低高糖毒性, 并且不出现低血糖 现象。 另外, 该类化合物还具有预防胰岛细胞的损失、疗效持续时间长等优点。 TZD通过激动 PPAR γ来调节脂肪细胞的分化、 提高对胰岛素的敏感性。 这类 能激动 PPAR γ受体、已上市的药物有罗格列酮 (rosiglitazone,文迪雅, Avandia) 和吡格列酮 (pioglitazone, 艾汀, Actos;)。 虽然 TZD类化合物在临床上已被证明 是非常有效的抗 II型糖尿病药物, 但由于此两种药物能造成病人体重增加、 水 肿、脂肪组织激增、骨髓脂肪酸改变等副作用, 因此有必要寻找一类新的 PPAR Y激动剂。 发明内容
本发明公开了一类具有选择性激活 PPAR γ的化合物或其盐。
本发明还公开了所述化合物或其盐的制备方法, 以及所述化合物作为治疗 PPAR γ受体相关疾病, 如糖尿病或相关并发症的临床应用。 在本发明的第一方面, 提供了一种通式 (I)所示化合物, 或其晶型、 药学上 可接受的盐、 水合物或溶剂合物:
Figure imgf000003_0001
其中, X为卤素, 优选氟、 氯, 更优选氯;
1为 (:1- 4直链或支链烷基, 例如甲基、 乙基、 丁基、 正丙基、 异丙基、 正丁基、 异丁基、 叔丁基, R1优选正丁烷基; 或者, 所述的烷基是具有 1-3个 取代基的 C C4烷基, 所述取代基为 1-3个选自下组的基团: 卤素、 -OH、 C C4 烷基、 烷氧基、 C C4烷基 -OH、 d-C4烷氧基甲基、 C2-C4酯基、 或磺酸 酯;
R2包括:
a)-CHO
Figure imgf000003_0002
其中, R'、 R"分别独立的选自取代或未取代的 C C4直链或支链烷基 (例 如甲基、 乙基、 丁基、 正丙基、 异丙基、 正丁基、 异丁基、 叔丁基) , 其中, 所述的取代烷基是具有 1-3个取代基的 C C4烷基,所述取代基为 1-3个选自下 组的基团: 卤素、 -OH、 d-C4烷基、 d-C4烷氧基、 d-C4烷基 -OH、 d-C4烷 氧基甲基、 C2-C4酯基、 或磺酸酯;
或 OR'和 OR"与相邻 C原子一起形成取代或非取代五元或六元环; 所述 取代基例如是 1-3个选自下组的基团: 卤素、 -OH、 C C4烷基、 C C4烷氧基、 d-C4烷基 -OH、 d-C4烷氧基甲基、 C2-C4酯基、 或磺酸酯;
c)
d)
式中, n=0或 1, Arl、 Ar2分别为芳香环 (优选苯环) , 环上无取代基 或有一个或多个 (例如 1-3 个;)取代基, 所述取代基为 ¾素、 烷基或烷氧基, 优 选卤素取代, 更优选氟、 氯取代;
在另一优选例中, 所述的烷基是 C 未取代或具有 1-3个取代基的烷 基, 所述取代基为 1-3 个选自下组的基团: 卤素、 -OH、 d-C4烷基、 C C4烷 氧基、 d-C4烷基 -OH、 C C4烷氧基甲基、 C2-C4酯基、 或磺酸酯;
在另一优选例中, 所述的烷氧基是 C C6未取代或具有 1-3个取代基的 烷氧基, 所述取代基为 1-3个选自下组的基团: 卤素、 -OH、 C C4烷基、 C C4 烷氧基、 C C4烷基 -OH、 d-C4烷氧基甲基、 C2-C4酯基、 或磺酸酯;
H
N- -N
1/ —— COOR4
\ 或
\
N= : N
其中, R4为氢或 d-C4烷基, 优选地, 所述的烷基是未取代或具有 1-3个 取代基的 C C4烷基, 所述取代基为 1-3个选自下组的基团: 卤素、 -OH、 C C4 烷基、 烷氧基、 C C4烷基 -OH、 d-C4烷氧基甲基、 C2-C4酯基、 或磺酸 酯, 最优选甲基。
在另一优选例中, R3为
Figure imgf000004_0001
在另一优选例中, R3为- COOR4, 其中 R4为氢或甲基,
在另一优选例中, X为氯。 在另一优选例中, R1为正丁烷基。
在另一优选例中, R2为- CHO; 或
R2为
OR"
式中, R'、 R"分别独立地选自 C C4直链或支链烷基, 所述的烷基是 C C4 未取代或具有 1-3个取代基的烷基, 所述取代基为 1-3个选自下组的基团: 卤 素、 -OH、 d-C4烷基、 d-C4烷氧基、 C C4烷基 -OH、 d-C4烷氧基甲基、 C2-C4 酯基、 或磺酸酯;
或 OR'和 OR"与相邻 C原子一起形成取代或非取代五元或六元环;所述的取 代的五元或六元环具有 1-3个选自下组的取代基: 所述取代基为 1-3个选自下 组的基团: 卤素、 -OH、 d-C4烷基、 d-C4烷氧基、 d-C4烷基 -OH、 d-C4烷 氧基甲基、 C2-C4酯基、 或磺酸酯; 或
R2为 其中, Arl为苯环, 环上无取代基或有一个或多个取代基, 取代基为氟或 氯; 或
R2为
其中, Ar2 为苯环, 环上无取代基或有一个或多个取代基, 取代基为氟或 氯, n=0或 1。
本发明的优选化合物选自下组:
2-丁基 -4-氯 -1-[[2'-(1Η-四唑 -5-基) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑 -5-(5, 5- 二乙醚;);
2-丁基 -4-氯 -1-[[2'-(1Η-四唑 -5-基) [Ι, Γ-联苯] -4-基]甲基] -1Η-咪唑 -5-(1, 3- 二氧戊环 -4(5)-甲基 -2-基);
2-丁基 -4-氯 -1-[[2'- (甲酸甲酯 )[1, 1,-联苯] -4-基]甲基] -1Η-咪唑 -5-(1, 3-二氧 戊环);
2-丁基 -4-氯 -1-[[2'-甲酸 -[1, 1'-联苯] -4-基]甲基] -1Η-咪唑 -5-(1,3-二氧戊环); 2-丁基 -4-氯 -1-[[2'- (甲酸甲酯 )[1, 1,-联苯] -4-基]甲基] -1Η-咪唑 -5-甲醛; 2-丁基 -4-氯 -1-[4'- (甲酸甲酯) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑 -5-甲醛;
2-丁基 -4-氯 -1-[[2'- (甲酸甲酯) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑 -5-甲基 -苯甲 醚;
2-丁基 -4-氯 -1-[[2'-(1Η-四唑 -5-基) [Ι, Γ-联苯] -4-基]甲基] -1Η-咪唑 -5-甲基- 间氟苯甲醚;
2-丁基 -4-氯 -1-[[2'- (甲酸甲酯) [Ι, Γ-联苯] -4-基]甲基] -1Η-咪唑 -5- (亚甲基-苯 甲胺);
2-丁基 -4-氯 -1-[[2'- (甲酸甲酯) [Ι, Γ-联苯] -4-基]甲基] -1Η-咪唑 -5- (亚甲基对 氯苯胺);
2-丁基 -4-氯 -1-[[2'-甲酸 -[1, 1'-联苯] -4-基]甲基] -1Η-咪唑 -5-甲基-间氟苯甲 醚;
2-丁基 -4-氯 -1-[[2'-甲酸 -[1, 1 '-联苯] -4-基]甲基] -1Η-咪唑 -5- (亚甲基 -苯甲 胺;); 或上述各化合物的盐。
在本发明的第二方面, 提供了一种药物组合物, 它包含治疗有效量的本发 明所述化合物或其药学上可接受的盐以及药学上可接受的载体、 赋形剂或稀释 剂。
在本发明的第三方面, 提供了本发明所述化合物或其药学上可接受的盐在 制备治疗 PPAR Y受体相关疾病的药物中的应用: 血糖异常升高相关疾病、 糖 尿病、 脂质紊乱、 代谢综合症、 心血管疾病、 冠状动脉疾病、 高血胆固醇或肥 胖症。
本发明还提供了一种治疗或预防 PPAR Y受体相关疾病的方法,包括步骤: 给需要的对象施用例如 0.05-200mg/kg体重 /天的本发明所述的化合物或其药学 上可接受的盐。
在另一优选例中, 所述 PPAR Y受体相关疾病包括、 血糖异常升高相关疾 病、 糖尿病、 脂质紊乱、 代谢综合症、 心血管疾病、 冠状动脉疾病、 高血胆固 醇或肥胖症。
在本发明的第四方面, 提供了一种制备药物组合物的方法, 包括将本发明 所述化合物或其药学上可接受的盐与药学上可接受的载体混合, 从而形成药物 组合物。 具体实施方式
本发明人经过广泛而深入的研究, 筛选了大量的化合物, 首次发现式(I) 化合物不仅对 ΡΡΑΙίγ受体有较好的激活作用, 而且对 PPARoc受体基本上无激活 作用, 因此与现有药物相比, 其副作用小, 不会导致患者体重明显增加。 在此 基础上完成了本发明。 活性成分
如本文所用,术语"本发明化合物 "指式 (I)所示的化合物。该术语还包括及式(I) 化合物的各种晶型形式、 药学上可接受的盐、 水合物或溶剂合物。
本发明中, 可以将化合物转化成 "药学上可接受的盐" 的形式。 所述盐是 指相对无毒的无机酸加成盐或有机酸加成盐。 这些盐可在化合物最后的分离和 提纯过程中现场制备, 或者是使纯化的化合物以其游离碱形式与适宜的有机或 无机酸进行反应, 再将形成的盐分离而制成。代表性盐包括氢溴酸盐、盐酸盐、 硫酸盐、 亚硫酸盐、 乙酸盐、 草酸盐、 戊酸盐、 油酸盐、 棕榈酸盐、 硬脂酸盐、 月桂酸盐、 硼酸盐、 苯甲酸盐、 乳酸盐、 磷酸盐、 柠檬酸盐、 马来酸盐、 富马 酸盐、 琥珀酸盐、 酒石酸盐、 苯甲酸盐、 甲磺酸盐、 葡萄糖酸盐、 乳糖酸盐和 月桂基磺酸盐等。 它们可包含基于碱金属和碱土金属的阳离子, 如钠、 锂、 钾、 钙、 镁等, 以及无毒胺、 季胺和胺阳离子等。 制备方法
本发明所述化合物可以通过以下工艺路线制备:
Figure imgf000008_0001
Figure imgf000008_0002
在上述路线中, 各取代基如上文所述。 咪唑醛与联苯苄溴在合适的条件 下发生亲核取代反应, 得到醛类目标化合物, 结构通式如 1所示; 通式 1化合 物在合适的条件下与一元或多元醇起反应, 得到缩醛类目标化合物 2 ; 通式 1 化合物在合适的条件下与芳香胺发生希夫碱反应, 得到亚胺类目标化合物 3 ; 在合适的条件下, 将通式 1化合物还原为醇, 所得醇与 ¾代芳香族化合物发生 取代反应, 得到醚类目标化合物 4。
在该流程中, 各反应通常在惰性溶剂中, 在室温至回流温度(如 0°C〜90 V, 优选 0°C〜60°C )下进行。 反应时间通常为 0. 1 小时一 60小时, 较佳地为 0. 5-48小时。 药物组合物和施用方法
利用本发明所得的化合物或其盐可给药于人, 所述化合物可以单独给药, 或者与其他药学上可接受的化合物联合给药。 需要指出, 本发明的化合物可以 混合给药。 可以口服、 直肠、 肠胃外 (静脉内、 肌肉内或皮下;)、 局部给药 (粉剂、 软膏剂或滴剂;)。
用于口服给药的固体剂型包括胶囊剂、 片剂、 丸剂、 散剂和颗粒剂。 固 体剂型中通常含有 0.5〜50%的活性成分, 较佳的含有 1〜20 %的活性成分, 最 佳的含有 1〜10 %的活性成分。 在这些固体剂型中, 活性化合物与至少一种常 规惰性赋形剂 (或载体;)混合, 如: (a) 填料或增容剂, 例如, 淀粉、 乳糖、 蔗糖、 葡萄糖、 甘露醇和硅酸; (b) 粘合剂, 例如, 羟甲基纤维素、 藻酸盐、 明胶、 聚乙烯基吡咯烷酮、 蔗糖和阿拉伯胶; (c) 保湿剂, 例如, 甘油; (d) 崩解剂, 例如, 琼脂、 碳酸钙、 马铃薯淀粉或木薯淀粉、 藻酸、 某些复合硅酸盐、 和碳 酸钠; (e) 缓溶剂, 例如石蜡; (f) 吸收加速剂, 例如, 季胺化合物; (g) 润湿 剂, 例如鲸蜡醇和单硬脂酸甘油酯; (h) 吸附剂, 例如, 高岭土; 和 (; i) 润滑剂, 例如, 滑石、 硬脂酸钙、 硬脂酸镁、 固体聚乙二醇、 十二烷基硫酸钠, 或其混 合物。
固体剂型如片剂、 糖丸、 胶囊剂、 丸剂和颗粒剂可采用包衣和壳材制备, 如肠衣和其它本领域公知的材料。 它们可包含不透明剂, 并且, 这种组合物中 活性化合物或化合物的释放可以延迟的方式在消化道内的某一部分中释放。 必 要时, 活性化合物也可与上述赋形剂中的一种或多种形成微胶囊形式。
用于口服给药的液体剂型包括药学上可接受的乳液、 溶液、 悬浮液、 糖 浆或酊剂。 除了活性化合物外, 液体剂型可包含本领域中常规采用的惰性稀释 剂, 如水或其它溶剂, 增溶剂和乳化剂, 例知, 乙醇、 异丙醇、 碳酸乙酯、 乙 酸乙酯、 丙二醇、 1 , 3 -丁二醇、 二甲基甲酰胺以及油, 特别是棉籽油、花生油、 玉米胚油、 橄榄油、 蓖麻油和芝麻油或这些物质的混合物等。
除了这些惰性稀释剂外, 组合物也可包含助剂, 如润湿剂、 乳化剂和悬 浮剂、 甜味剂、 矫味剂和香料。
除了活性化合物外, 悬浮液可包含悬浮剂, 例如, 乙氧基化异十八烷醇、 聚氧乙烯山梨醇和脱水山梨醇酯、 微晶纤维素、 甲醇铝和琼脂或这些物质的混 合物等。
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、 分散液、 悬浮液或乳液, 和用于重新溶解成无菌的可注射溶液或分散液的无菌 粉末。 适宜的含水和非水载体、 稀释剂、 溶剂或赋形剂包括水、 乙醇、 多元醇 及其适宜的混合物。
用于局部给药的本发明化合物的剂型包括软膏剂、 散剂、 喷射剂和吸入 剂。 活性成分在无菌条件下与生理上可接受的载体及任何防腐剂、 缓冲剂, 或 必要时可能需要的推进剂一起混合。
本发明的 "治疗有效量" 是指可对人和 /或动物产生功能或活性的且可被 人和 /或动物所接受的量。 治疗有效量的本发明的组合物介于 0.01-200mg/kg体 重 /天之间。 任何介于上述范围之内的用量皆为本发明的有效量。 所述的 "治疗 有效量" 可用于相关疾病的单一用药或联合用药治疗。 本领域的专业人员能够 理解, 在实际给药时的用量可高于或低于上述剂量范围。针对某一对象 (如哺乳 动物-人;)的 "治疗有效量" 和具体治疗方案可受诸多因素的影响, 包括所用化 合物或其前药的药效活性、 给药对象的年龄、 体重、 一般情况、 性别、 饮食、 给药时间、 疾病易感性、 疾病进程以及收治医师的判断等。
本发明所述化合物在临床上可以通过口服或注射方式对哺乳动物 (;包括人;) 进行用药, 其中尤以口服方式最佳。 用药剂量为每日 0.01〜 200mg/kg体重, 较佳用药剂量为每日 0.01〜100mg/kg 体重, 最佳用药剂量为每日 0.01〜 50mg/kg体重, 同时, 最佳剂量视个体而定量。
本发明化合物或其盐可用于治疗或预防与 RXR和 ΡΡΑΙ γ核受体调节相关 的疾病, 还可用于治疗血糖异常升高相关的疾病。 具体地说, 本发明化合物或 其盐可用于治疗糖尿病, 或用于治疗脂质紊乱、 代谢综合症、 心血管疾病、 冠 状动脉疾病、 高血胆固醇或肥胖症。 本发明的主要优点在于: 与完全的 ΡΡΑΙ γ受体激动剂如罗格列酮相比, 本 发明化合物对 ΡΡΑΙ γ受体具有部分激动活性,并且不会导致糖尿病动物或患者 体重的增加。 以下结合具体实施例, 进一步阐明本发明。 应理解, 这些实施例仅用于说 明本发明而不用于限制本发明的范围。 下列实施例中未注明具体条件的实验方 法, 通常按照常规条件, 例如是 《贝尔斯坦有机化学手册》 (化学工业出版社, 1996 年;)中的条件, 或按照制造厂商所建议的条件。 比例和百分比基于重量, 除非特别说明。
除非另有定义或说明, 本文中所使用的所有专业与科学用语与本领域技术 熟练人员所熟悉的意义相同。 此外任何与所记载内容相似或均等的方法及材料 皆可应用于本发明方法中。 本发明的最佳实施方案 实施例 1 : 2-丁基 -4-氯 -1-[[2'-(1Η-四唑 -5-基) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑
Figure imgf000011_0001
在装有分水器、冷凝管的 100ml烧瓶中加入 5ml的乙醇、 105mg(0.25mmol) 的 2-丁基 -4-氯 -1-[[2'-(1Η-四唑 -5-基) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑 -5-甲醛, 15ml正己烷, 加入 4eq的 CuCl22H2O, 回流反应 14h。 停止反应后蒸干乙醇, 加入 5ml 的乙酸乙酯后用 (5ml X 3)饱和的氢氧化钠水溶液和水 (5ml X 2)洗涤后 拌硅胶过柱, 得 53mg产物 (41.67%)。
H-NMR(CDC13) δ 8.08(s, lH), δ 7.60-7.55(t,2H), δ 7.41-7.40(t,2H), δ 7.26-7.15(d,2H), δ 7.07-7.00(d,2H), δ 5.30(s,2H), δ 3.3-3.27(t,2H), δ 2.63(d,2H), δ 2.38(t,2H), δ 1.34-1.28(t,2H), δ 1.26-1.25(t,3H), δ 1.08-1.04(t,3H), δ 0.90-0.82(t,3H) 实施例 2: 2-丁基 -4-氯 -1-[[2'-(1Η-四唑 -5-基) [Ι, Γ-联苯] -4-基]甲基] -1H-咪 唑 -5-(1, 3-二氧戊环 -4(5)-甲基 -2-基)的制备
Figure imgf000011_0002
在 100ml烧瓶中加入 2ml的 1, 2-丙二醇、 105mg(0.25mmol)的 2-丁基 -4- 氯 -1-[[2'-(1Η-四唑 -5-基) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑 -5-甲醛、 15ml正己烷, 搅拌使之溶解, 加入 4eq的 CuCl2 2H2O, 回流反应 12h。 停止反应后蒸干 1, 2-丙二醇, 加入 5ml 的乙酸乙酯后用 (5ml X 3)饱和的氢氧化钠水溶液和水 (5ml X 2)洗涤后拌硅胶过柱, 得 37mg产物 (30.8%)。
H-NMR(CDC13) δ 7.60-7.57(t,2H), δ 7.51-7.47(t,2H) , δ 7.08-7.04(s,4H) , δ 5.44(s,2H), δ 4.10-4.02(t,2H), δ 3.40-3.39(d, lH), δ 2.49-2.45(t,2H), δ 1.47-1.43(t,2H), δ 1.28-1.21(t,3H), δ 1.19-1.1 l(t,2H), δ 0.84-0.81(t,3H). 实施例 3 : 2-丁基 -4-氯 -1-[[2'- (甲酸甲酯) [1, 1'-联苯] -4-基]甲基] -1H-咪唑
Figure imgf000012_0001
在 100ml烧瓶中加入 2ml的乙二醇、 105mg(0.25mmol)的 2-丁基 -4-氯 -1-[[2'- (甲酸甲酯) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑 -5-甲醛、 15ml正己烷, 搅拌使之 溶解, 加入 4eq的 CuCl2 2H2O, 回流反应 12h。 停止反应后蒸干乙二醇, 加入 5ml 的乙酸乙酯后用 (5ml X 3)饱和的氢氧化钠水溶液和水 (5ml X 2)洗涤后拌硅 胶过柱, 得 37mg产物 (30.8%)。
1H NMR(400Mz, CDC13): δ9.77(1Η, s), 7.83(1H, dd, J=1.10Hz, 7.70Hz), 7.56-7.49(lH, m), 7.45-7.39(lH, m), 7.36-7.26(3H, m), 7.09(2H, d, J=8.43Hz), 5.17(1H, s),5.02(2H, s), 4.29-4.23(2H, t,), 4.19-4.13(2H, t,) 3.63(3H, s), 2.66(2H, t, J=7.70Hz), 1.73-1.64(2H, m), 1.41-1.32(2H, m), 0.89(3H, t, J=7.33Hz) 实施例 4: 2-丁基 -4-氯 -1-[[2'-甲酸 -[1, 1'-联苯] -4-基]甲基] -1H-咪唑 -5-(l,
Figure imgf000012_0002
实施例 3产物 170mg溶于 30ml二氧六环, 加入 10% NaOH溶液 2ml, 90 °C反应 6h。 减压浓缩, 残余物加水形成溶液, 乙酸乙酯提取一次。 水相析出白 色固体, 调 PH成中性, 过滤干燥得产物。
1H NMR(400Mz, CDC13): δ14.12(1Η, s), 8.29(1H, s), 7.44-7.40(2H, dd, J=1.10Hz, 7.70Hz), 7.22-7.20(2H, m), 7.12-7.10(2H, m), 7.09(1H, d, J=8.43Hz), 5.17(1H, s), 4.29-4.23(2H, t,), 4.19-4.13(2H, t,) 3.63(3H, s), 2.66(2H, t, J=7.70Hz), 1.73-1.64(2H, m), 1.41-1.32(2H, m), 0.89(3H, t, J=7.33Hz) 实施例 5 : 2-丁基 -4-氯 -1-[[2'- (甲酸甲酯) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑 -5- 甲醛
Figure imgf000013_0001
咪唑醛 6.30g溶于 80ml DMF, 加入 NaH 3.5g, 有大量气体产生, 室温搅 拌反应 lh, 接着加入联苯苄溴 9.36g, 40度反应 12h。 原料转化完全后, 加压 蒸去大部分 DMF 后, 乙酸乙酯稀释, 水洗, 有机相干燥后层析纯化得粘稠状 产物。
1H NMR(400Mz, CDC13): δ9.77(1Η, s), 7.83(1H, dd, J=1.10Hz, 7.70Hz), 7.56-7.49(lH, m), 7.45-7.39(lH, m), 7.36-7.26(3H, m), 7.09(2H, d, J=8.43Hz), 5.59(2H, s), 3.63(3H, s), 2.66(2H, t, J=7.70Hz), 1.73-1.64(2H, m), 1.41-1.32(2H, m), 0.89(3H, t, J=7.33Hz) 实施例 6: 2-丁基 -4-氯 -1-[4'- (甲酸甲酯) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑 -5- 甲醛
Figure imgf000013_0002
COOMe
咪唑醛 1.86g, lOmmol 溶于 15ml DMF 中, 加入 410mg 60% NaH, 2.50g , 10mmol对溴苄溴固体, 室温反应过夜。 300ml乙酸乙酯稀释, 冰水充分 洗涤以除去 DMF, 有机相干燥, 层析纯化得油状物。
取 533mg上述油状物, 对甲酸甲酯苯基硼酸 360mg,和 K2CO3 固体粉末 414mg,—起溶于 THF/EtOH(l: l)混合溶液 40ml中, Ar保护下, 迅速加入催化 /:/:/ O 6800/-0/-00si>l£ ϊ£6εΗ/-00ίAV
0§((D)8¾9 ¾0∞£ΐ∞ ¾£0£¾面 Ηΐs : ¾-.··
Figure imgf000014_0001
奪 t画%ς6£Ξ 。
ss蝴 ώ【m8 "-- 叵蝴^ ώί--- 三苯甲基氯沙坦 665mg,溶于 10ml DMF中, 加入 44mg 60% NaH, 室温 反应半小时, 加入间氟苄溴 122μ1,反应 12h。 常规后处理后层析纯化得产物。
1H NMR(400Mz, CDC13): δ7.94(1Η, m), 7.52-7.46(2H, m), 7.36-7.30(4H, m) 7.28-7.21(8H, m), 7.09(2H, d, J=8.05Hz), 7.00-6.89(8H, m), 6.70(2H, d, J=8.24Hz) 5.04(2H, s), 4.35(2H, s), 4.21(2H, s), 2.48(2H, t, J=7.87Hz), 1.70-1.61(2H, m), 1.36-1.20(2H, m), 0.85(3H, t, J=7.32Hz).
上述中间产物 105mg溶于 10ml 无水甲醇中, 加入 45mg的 KOH, 回流 反应 2h, 原料转化完全, 浓缩后层析纯化得固体产物。
1H NMR(400Mz, CDC13): δ7.87(1Η, d, J=7.32Hz), 7.63-7.50(2H, m), 7.37(1H, d, J=7.47Hz), 7.26-7.19(1H, m), 7.02(2H, d, J=7.62Hz), 6.98-6.85(3 H, m) 6.71(2H, d, J=7.78Hz), 5.08(2H, s), 4.39(2H, s), 4.21(2H,s), 2.29(2H, t, J=7.48Hz), 1.59-1.40(2H, m), 1.30-1.18(2H, m), 0.82(3H, t, J=7.25Hz). 实施例 9 : 2-丁基 -4-氯 -1-[[2'- (甲酸甲酯) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑 -5- (亚甲基-苯甲胺)的制备
Figure imgf000015_0001
在 100ml 烧瓶中加入 206mg 0.5mmoi;)的 2-丁基 -4-氯 -1-[[2'- (;甲酸甲 酯) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑 -5-甲醛, lOOmg苄胺, 加入 5ml甲醇搅拌使 之溶解, 加入 50 μ ΐ 的冰醋酸, 室温搅拌 16h。 析出白色固体, 过滤后用水洗 涤(10ml X 3)烘干得 153mg产物 (51.5%)。
H-NMR(CDC13): δ 8.38-8.37(s, lH), δ 7.84-7.55(d, lH), δ 7.84-7.81(d, lH), δ 7.55-7.51(t, lH), δ 7.44-7.39(t, lH), δ 7.27-7.16(m,5H), δ 7.03-7.01(d,2H), δ 7.15-7.14(d,2H), δ 5.81(s,2H), δ 4.69(s,2H), δ 3.58(s,3H), δ 2.64-2.60(t,2H), δ 1.71-1.65(t,2H)
δ 1.39-1.30(m,2H), δ 0.90-0.86(t,3H). 实施例 10: 2-丁基 -4-氯 -1-[[2'- (甲酸甲酯) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑
-5- (亚甲基对氯苯胺)的制备
Figure imgf000016_0001
在 100ml 烧瓶中加入 206mg 0.5mmoi;)的 2-丁基 -4-氯 -1-[[2'- (;甲酸甲 酯;) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑 -5-甲醛, lOOmg对氯苯胺, 加入 5ml甲醇搅 拌使之溶解, 加入 30 μ 1的冰醋酸, 室温搅拌 16h。 蒸干溶剂后加 10ml乙酸乙 酯溶解, 用饱和的 NaHCO3水溶液洗涤 (10ml X 3)后过柱得 30mg产物。
H-NMR(CDC13) : δ 8.38(s, lH), δ 7.84-7.81(d, lH) , δ 7.55-7.49(t, lH), δ 7.43-7.38(t, lH), δ 7.34-7.25(m,5H), δ 7.13-7.10(d,2H), δ 7.03-6.98(d,2H), δ 5.85(s,2H), δ 3.58(s,3H), δ 2.71-2.66(t,2H), δ 1.74-1.69(t,2H) , δ 1.39-1.35(m,2H), δ 0.93-0.89(t,3H). 实施例 11 : 2-丁基 -4-氯 -1-[[2'-甲酸 -[1, 1'-联苯] -4-基]甲基] -1H-咪唑 -5-甲基
-间氟苯甲醚
Figure imgf000016_0002
实施例 5化合物 1.50g溶于 THF/MeOH(2: 1)混合溶液 40ml中,加入 NaBH4 固体 276mg 体系放热, 有气体产生。 lOmin后, 原料转化完全。 浓缩去大部分 溶剂, 残余物用乙酸乙酯溶解, 水洗。 有机相干燥纯化得醛基还原产物固体。
取上述还原产物 0.6g, 溶于 8ml无水 DMF中,加入 69mg NaH搅拌反应半 小时后, 滴加 139μ1 间氟苄溴室温反应 5小时, TLC显示已经大部分转化。 乙 酸乙酯稀释体系后, 水洗, 有机相干燥层析纯化得油状产物。 1H NMR(400Mz, CDC13): δ7.87(1Η, d, J=7.32Hz), 7.63-7.50(2H, m),
7.37(1H, d, J=7.47Hz), 7.26-7.19(1H, m), 7.02(2H, d, J=7.62Hz), 6.98-6.85(3 H, m): 6.71(2H, d, J=7.78Hz), 5.08(2H, s), 4.39(2H, s), 4.21(2H,s), 3.63(3H, s), 2.29(2H, t J=7.48Hz), 1.59-1.40(2H, m), 1.30-1.18(2H, m), 0.82(3H, t, J=7.25Hz). 取上述产物 lg, 溶于 40ml THF中, 加入 10%NaOH溶液 4ml, 回流反应 6 小时, TLC显示原料转化完全后, 浓缩体系, 残余物用水溶解, IN HC1 溶液 调体系 PH值 2-3, 析出大量固体。 抽滤干燥得产物 (标题化合物)。
1H NMR(400Mz, CDC13): 512. 1( 1H,S),7.87( 1H, d, J=7.32Hz), 7.63 -7.50(2H, m), 7.37( 1H, d, J=7.47Hz), 7.26-7.19( 1H, m), 7.02(2H, d, J=7.62Hz), 6.98-6.85(3 H m), 6.71(2H, d, J=7.78Hz), 5.08(2H, s), 4.39(2H, s), 4.21 (2H,s), 3.63(3H, s), 2.29(2H, t, J=7.48Hz), 1.59- 1.40(2H, m), 1.30- 1. 18(2H, m), 0.82(3H, t, J=7.25Hz).
实施例 12 : 2-丁基 -4-氯 -1-[[2'-甲酸 -[Ι, Γ-联苯] -4-基]甲基] -1H-咪唑 -5- (亚 甲基 -苯甲胺;)的制备
Figure imgf000017_0001
将实施例 9化合物 190mg溶于 30ml二氧六环,加入 10% NaOH溶液 2.5ml, 90°C反应 6h。 减压浓缩, 残余物加水形成溶液, 乙酸乙酯提取一次。 水相析出 白色固体, 调 PH成中性, 过滤干燥得产物。
1H NMR(400Mz, CDC13): δ14.12(1Η, s), 9.19(1H, s), δ 8.29(s, lH), δ 7.43-7.39(m,3H) , δ 7.27-7.21(m,3H) , δ 5.06(s,2H), δ 4.74(s,2H), δ 2.64-2.60(t,2H), δ 1.71- 1.65(t,2H), δ 1.39-1.30(m,2H), δ 0.90-0.86(t,3H). 实施例 13 活性测定
对于实施例 1-12制备的化合物 (;即化合物 1- 12), 通过细胞学实验, 测定 了其对 ΡΡΑΙ γ、 PPARoc受体的激动活性, 得到了浓度 /效应关系, 并计算出相 应的 EC5Q值。
13.1 试验方法
采用报告基因的方法, 利用核受体活化后能激活它下游基因转录的原理 设计了一种活细胞内筛选核受体激活剂的筛选模型, 用来验证化合物激活 PPAR受体的活性, 实验方法如下:
对 PPARY受体的激活 用 RT-PCR 方法从脂肪组织中克隆到全长的 ΡΡΑΙ γ cDNA, 将扩增到的 PCR 产物插入 pcDNA3.1 表达载体后测序鉴定。 报告基因用 Promega 公司的 荧光素酶检测载体 pGL3-Promoter 构建。 转染实验用 U2OS细胞在 96 孔板中 进行, 在转染报告基因的同时共转染 RXR和 ΡΡΑΙ γ 基因, 转染 24 小时后加 入待检测的化合物,并使溶剂 DMSO 的终浓度保持在 0.1 %。化合物作用 24 小 时后裂解细胞并进行荧光素酶活性的检测。 通过观察发光的强度可以得知化合 物对核受体的激活强度。 为了校正转染效率、 细胞接种数量及化合物毒性等因 素造成的试验误差, 还同时共转染了 GFP质粒作为内参, 在实验结果分析时所 有试验孔的发光值都用 GFP值进行了校正。试验结果用相对激活倍数表示, 溶 剂对照的值为 1, 值越大表明激活能力越高。 在模型筛选中, 观察了样品在 6 种不同浓度条件下对受体的激活情况, 较全面地反映了化合物的药理特性, 并 计算出相应的半有效浓度 (EC5();)。 对 PPAR a受体的激活
萤火虫荧光素酶报告基因系统对化合物激活 RXR/PPAR ci异二聚体活性 的分析。 用常规 RT-PCR 方法从脂肪组织中克隆到全长的 PPAR cDNA, 将 扩增到的 PCR 产物插入市售的 pcDNA3.1 表达载体后测序鉴定。 报告基因用 Promega 公司的荧光素酶检测载体 pGL3-Promoter 构建。 转染实验用 U2OS细 胞在 96 孔板中进行, 在转染报告基因的同时共转染 RXR和 PPAR a基因, 转 染 24 小时后加入待检测的化合物, 并使溶剂 DMSO 的终浓度保持在 0.1 %。 化合物作用 24 小时后裂解细胞并进行荧光素酶活性的检测。 通过观察发光的 强度可以得知化合物对核受体的激活强度。 为了校正转染效率、 细胞接种数量 及化合物毒性等因素造成的试验误差, 还同时共转染了 GFP质粒作为内参, 在 实验结果分析时所有试验孔的发光值都用 GFP值进行了校正。试验结果用相对 激活倍数表示, 溶剂对照的值为 1, 值越大表明激活能力越高。
13.2 结果
实验结果表明, 本发明实施例化合物对 ΡΡΑΙ γ受体有不同程度的激活作 用,而对 PPARo 受体无激活作用;与完全的 ΡΡΑΙ γ受体激动剂罗格列酮相比, 本发明实施例化合物对 PPARj受体具有部分激动活性。
结果详见下表 1和表 2: 表 1: PPARy:
Figure imgf000019_0001
表 2: PPAR :
Figure imgf000019_0002
Ia=无活性 (inactivity) 实施例 14 降糖药效实验
健康自发性 2型糖尿病动物模型,雄性 GK大鼠, 200g,动物禁食 12小时, 血糖仪测定空腹血糖值, 之后连续灌胃分别给予化合物 1-12、 罗格列酮 (20 mg/kg)或对照液 0.5%CMC (10ml/kg), 10天后再次测定空腹血糖值。
结果表明: 本发明实施例化合物具有良好的降血糖活性。 具体实验数据如 下表 3 :
表 3
Figure imgf000020_0001
实施例 15 长期给药体重增加实验
健康自发性 2型糖尿病动物模型, 雄性 GK大鼠, 200g, 连续给予含有实 施例化合物 3、 4、 7或罗格列酮的饲料 (20 mg/kg/天 以给予正常饲料组为对 照, 40天后观察体重增加情况。 实验结果: 空白对照组动物体重增长 23.2%; 给予实施例化合物饲料组体重增长分别为 26.7%、 26.2%、 25.5%; 给予罗格列 酮饲料组的体重增长为 38.7%。
结果表明: 与完全的 PPARY受体激动剂罗格列酮相比, 本发明化合物基 本上不会导致糖尿病动物体重的增加。 实施例 16 药物组合物
化合物 1 23g
淀粉 140g
微晶纤维素 67g
按常规方法, 将上述物质混合均匀后, 装入普通明胶胶囊, 得到 1000 颗 胶囊。
按类似方法, 分别制得含化合物 2-12的胶囊。 在本发明提及的所有文献都在本申请中引用作为参考, 就如同每一篇文献 被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后, 本领域技术人员可以对本发明作各种改动或修改, 这些等价形式同样落于本申 请所附权利要求书所限定的范围。

Claims

权 利 要 求
1、 结构通式 (I)所示化合物或其药学上可接受的盐:
Figure imgf000022_0001
其中, X为卤素;
R1为 C C4直链或支链烷基;
R2包括:
a)-CHO;
b)
OR'
OR"
其中, R'、 R"分别独立地选自取代或未取代的 C C4直链或支链烷基; 或 者 OR'和 OR"与相邻 C原子一起形成取代或非取代五元或六元环;
c)
Figure imgf000022_0002
d)
式中, Arl、 Ar2分别为芳香环, 环上无取代基或有一个或多个取代基, 所述取代基为卤素、 烷基或烷氧基, n=0或 1 ;
R3为以下基团的取代: 或 COOR
Figure imgf000022_0003
其中, R4为氢或 d-C4烷基。
2、 如权利要求 1所述的化合物或其药学上可接受的盐, 其特征在于, H
N-N
R3为 N=N; 或 R3为- COOR4, 其中 R4为氢或甲基。
3、 如权利要求 2所述化合物或其药学上可接受的盐, 其特征在于, X为 氯; 和 /或 R1为正丁烷基。
4、 如权利要求 3所述的化合物或其药学上可接受的盐, 其特征在于, R2为 -CHO; 或
R2为
Figure imgf000023_0001
式中, R'、 R"分别独立地选自 C C4直链或支链烷基, 所述的烷基是 C C4 未取代或具有 1-3个取代基的烷基, 所述取代基为 1-3个选自下组的基团: 卤 素、 -OH、 d-C4烷基、 d-C4烷氧基、 C C4烷基 -OH、 d-C4烷氧基甲基、 C2-C4 酯基、 或磺酸酯;
或 OR'和 OR"与相邻 C原子一起形成取代或非取代五元或六元环;所述的取 代的五元或六元环具有 1-3个选自下组的取代基: 所述取代基为 1-3个选自下 组的基团: 卤素、 -OH、 d-C4烷基、 d-C4烷氧基、 d-C4烷基 -OH、 d-C4烷 氧基甲基、 C2-C4酯基、 或磺酸酯; 或
R2为 其中, Arl为苯环, 环上无取代基或有一个或多个取代基, 取代基为氟或 氯; 或
R2为
其中, Ar2为苯环,环上无取代基或有一个或多个取代基,取代基为氟或氯, n=0或 1。
5、 如权利要求 1 所述的化合物或其药学上可接受的盐, 其特征在于, 所 述的化合物选自下组:
2-丁基 -4-氯 -1-[[2'-(1Η-四唑 -5-基) [Ι,Γ-联苯] -4-基]甲基] -1H-咪唑 -5-(5, 5- 二乙醚;);
2-丁基 -4-氯 -1-[[2'-(1Η-四唑 -5-基) [Ι,Γ-联苯] -4-基]甲基] -1Η-咪唑 -5-(1, 3- 二氧戊环 -4(5)-甲基 -2-基); 2-丁基 -4-氯 -1-[[2'- (甲酸甲酯) [1, 1 '-联苯] -4-基]甲基] -1H-咪唑 -5-(1, 3- 二氧戊环 ));
2-丁基 -4-氯 -1-[[2'-甲酸 -[1, 1'-联苯] -4-基]甲基] -1H-咪唑 -5-(1,3-二氧戊环); 2-丁基 -4-氯 -1-[[2'- (甲酸甲酯 )[1, 1,-联苯] -4-基]甲基] -1H-咪唑 -5-甲醛; 2-丁基 -4-氯 -1-[4'- (甲酸甲酯) [Ι, Γ-联苯] -4-基]甲基] -1H-咪唑 -5-甲醛;
2-丁基 -4-氯 -1-[[2'- (甲酸甲酯) [Ι, Γ-联苯] -4-基]甲基] -1Η-咪唑 -5-甲基 -苯甲 醚;
2-丁基 -4-氯 -1-[[2'-(1Η-四唑 -5-基) [Ι, Γ-联苯] -4-基]甲基] -1Η-咪唑 -5-甲基- 间氟苯甲醚;
2-丁基 -4-氯 -1-[[2'- (甲酸甲酯) [Ι, Γ-联苯] -4-基]甲基] -1Η-咪唑 -5- (亚甲基-苯 甲胺);
2-丁基 -4-氯 -1-[[2'- (甲酸甲酯) [Ι, Γ-联苯] -4-基]甲基] -1Η-咪唑 -5- (亚甲基对 氯苯胺);
2-丁基 -4-氯 -1-[[2'-甲酸 -[1, 1'-联苯] -4-基]甲基] -1Η-咪唑 -5-甲基-间氟苯甲 醚; 或
2-丁基 -4-氯 -1-[[2'-甲酸 -[1, 1 '-联苯] -4-基]甲基] -1Η-咪唑 -5- (亚甲基 -苯甲 胺)。
6、 一种药物组合物, 其特征在于, 包含治疗有效量的权利要求 1 所述化 合物或其药学上可接受的盐以及药学上可接受的载体、 赋形剂或稀释剂。
7、权利要求 1所述的化合物或其药学上可接受的盐在制备治疗 PPAR y受 体相关疾病的药物中的应用。
8、 一种治疗或预防 PPAR γ受体相关疾病的方法, 包括步骤: 给需要的对 象施用 0.05-200mg/kg体重 /天的权利要求 1所述的化合物或其药学上可接受的
9、 如权利要求 8所述的方法, 其特征在于, 所述的 PPAR γ受体相关疾病 选自下组: 血糖异常升高相关疾病、 糖尿病、 脂质紊乱、 代谢综合症、 心血管 疾病、 冠状动脉疾病、 高血胆固醇或肥胖症。
10、 一种制备药物组合物的方法, 其特征在于, 将权利要求 1所述的化合 物或其药学上可接受的盐与药学上可接受的载体混合, 从而形成药物组合物。
PCT/CN2007/070089 2006-06-07 2007-06-07 COMPOSÉS DOTÉS D'UNE ACTIVITÉ D'AGONISTE DU PPARγ ET LEUR APPLICATION WO2007143951A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN200610027344.7 2006-06-07
CNA2006100273447A CN101085772A (zh) 2006-06-07 2006-06-07 一类具有PPARγ激动剂活性的化合物及其应用

Publications (1)

Publication Number Publication Date
WO2007143951A1 true WO2007143951A1 (fr) 2007-12-21

Family

ID=38831424

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2007/070089 WO2007143951A1 (fr) 2006-06-07 2007-06-07 COMPOSÉS DOTÉS D'UNE ACTIVITÉ D'AGONISTE DU PPARγ ET LEUR APPLICATION

Country Status (2)

Country Link
CN (1) CN101085772A (zh)
WO (1) WO2007143951A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11358954B2 (en) 2016-04-13 2022-06-14 Mitobridge, Inc. PPAR agonists, compounds, pharmaceutical compositions, and methods of use thereof
US11578052B2 (en) 2015-10-07 2023-02-14 Mitobridge, Inc. PPAR agonists, compounds, pharmaceutical compositions, and methods of use thereof

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101260103A (zh) * 2007-03-09 2008-09-10 上海艾力斯医药科技有限公司 一类具有部分PPARγ激动剂活性的化合物及其应用
CN101475564B (zh) * 2009-01-22 2012-05-23 江苏德峰药业有限公司 一种氯沙坦钾的制备方法

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0253310A2 (en) * 1986-07-11 1988-01-20 E.I. Du Pont De Nemours And Company Angiotensin II receptor blocking imidazoles
US5538987A (en) * 1992-07-28 1996-07-23 Istituto Luso Farmaco D'italia S.P.A. Imidazole ethers having a II antagonist activity
US5962500A (en) * 1994-03-31 1999-10-05 Eide; Ivar K. Insulin sensitivity with angiotensin II receptor blocking imidazoles

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0253310A2 (en) * 1986-07-11 1988-01-20 E.I. Du Pont De Nemours And Company Angiotensin II receptor blocking imidazoles
US5538987A (en) * 1992-07-28 1996-07-23 Istituto Luso Farmaco D'italia S.P.A. Imidazole ethers having a II antagonist activity
US5962500A (en) * 1994-03-31 1999-10-05 Eide; Ivar K. Insulin sensitivity with angiotensin II receptor blocking imidazoles

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11578052B2 (en) 2015-10-07 2023-02-14 Mitobridge, Inc. PPAR agonists, compounds, pharmaceutical compositions, and methods of use thereof
US11358954B2 (en) 2016-04-13 2022-06-14 Mitobridge, Inc. PPAR agonists, compounds, pharmaceutical compositions, and methods of use thereof

Also Published As

Publication number Publication date
CN101085772A (zh) 2007-12-12

Similar Documents

Publication Publication Date Title
EP2595952B1 (en) Agonists of gpr40
WO2008006319A1 (fr) DÉRIVÉS DE BIBENZIMIDAZOLE À ACTIVITÉ AGONISTE PPARγ ET LEURS APPLICATIONS
JP2892838B2 (ja) アンジオテンシン▲ii▼受容体拮抗薬としてのイミダゾールカルボン酸のプロドラッグ
EA015516B1 (ru) Ингибиторы 11-бета-гидроксистероид дегидрогеназы 1
JP3199380B2 (ja) 化合物
WO2019226991A1 (en) Androgen receptor modulators and methods for their use
WO1993014070A2 (fr) Nouveaux derives de l'imidazole, leur preparation et leurs applications therapeutiques
EA021025B1 (ru) Соединения, эффективные в качестве ингибиторов ксантиноксидазы, способ их получения и содержащая их фармацевтическая композиция
TW200936576A (en) Carboxyl-or hydroxylsubstituted benzimidazole derivatives
JP2007506773A (ja) 線溶系の障害および血栓症の治療用のpai−1阻害剤としての、4−(フェニル−エチリデンアミノキシ−プロポキシ)−フェニル−酢酸誘導体および関連化合物
JPH06199811A (ja) ジアザ化合物
JP2009514816A (ja) 新規ベータ−アゴニスト、それらの調製方法及び薬物としてのそれらの使用
WO2008110062A1 (en) Compounds with partial agonist activity of pparϝ and application thereof
JP2007535512A (ja) 新規なβ−アゴニスト、その製造方法及びその薬物としての使用
CA2794176C (en) Novel benzamide derivatives
JPH09503503A (ja) 化合物、組成物ならびにアレルギーおよび炎症の治療
WO2007143951A1 (fr) COMPOSÉS DOTÉS D'UNE ACTIVITÉ D'AGONISTE DU PPARγ ET LEUR APPLICATION
JP2007538102A (ja) 糖尿病の処置に有用な5−アニリノ−4−ヘテロアリールピラゾール誘導体
US20090176841A1 (en) Novel 6-5 system bicyclic heterocyclic derivative and its pharmaceutical utility
WO2008044729A1 (en) Carboxylic acid derivative
US20220177418A1 (en) Inhibitors of the n-terminal domain of the androgen receptor
JP4146641B2 (ja) 3−アミノ−1−フェニル−1h−[1,2,4]トリアゾールの新規な分岐状置換アミノ誘導体、それらの製造法およびそれらを含む医薬組成物
US20230124268A1 (en) Nicotinamide phosphoribosyltransferase inhibitors and methods for use of the same
US20110112158A1 (en) Benzisoxazole analogs as glycogen synthase activators
WO2010095462A1 (ja) 新規な3-(5-アルコキシピリミジン-2-イル)ピリミジン-4(3h)-オン構造を有する化合物及びこれを含有する医薬

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07721710

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 07721710

Country of ref document: EP

Kind code of ref document: A1