WO2017152677A1 - Cholanic acid compound for preventing or treating fxr-mediated diseases - Google Patents

Cholanic acid compound for preventing or treating fxr-mediated diseases Download PDF

Info

Publication number
WO2017152677A1
WO2017152677A1 PCT/CN2016/109851 CN2016109851W WO2017152677A1 WO 2017152677 A1 WO2017152677 A1 WO 2017152677A1 CN 2016109851 W CN2016109851 W CN 2016109851W WO 2017152677 A1 WO2017152677 A1 WO 2017152677A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
disease
acid
hydroxy
evaporated
Prior art date
Application number
PCT/CN2016/109851
Other languages
French (fr)
Chinese (zh)
Inventor
王义汉
任兴业
Original Assignee
深圳市塔吉瑞生物医药有限公司
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 深圳市塔吉瑞生物医药有限公司 filed Critical 深圳市塔吉瑞生物医药有限公司
Publication of WO2017152677A1 publication Critical patent/WO2017152677A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/575Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of three or more carbon atoms, e.g. cholane, cholestane, ergosterol, sitosterol
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J9/00Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane
    • C07J9/005Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane containing a carboxylic function directly attached or attached by a chain containing only carbon atoms to the cyclopenta[a]hydrophenanthrene skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/007Steroids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J9/00Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Definitions

  • the invention belongs to the field of medicine.
  • the present invention relates to a cyanic acid compound and a pharmaceutical composition thereof for use in the prevention or treatment of FXR-mediated diseases and uses thereof.
  • Farnesol X receptor a member of the orphan nuclear receptor family, was first discovered by Forman et al in 1995, and its transcriptional activity can be named by the increase in superphysiological concentration of farnesol.
  • Northern and in situ analysis revealed extensive expression of FXR in the lung, intestine, kidney, and adrenal glands.
  • FXR forms a heterodimer with the 9-cis retinoic acid receptor (RXR) to bind to DNA.
  • the FXR/RXR heterodimer preferentially binds to a component consisting of a binuclear receptor half site that shares AG(G/T) TCA, which forms an inverted repeat and a single nucleoside separation (IR-1 motif).
  • Cholic acids as FXR ligands include chenodeoxycholic acid (CDCA), deoxycholic acid (DCA), lithocholic acid (LCA), and taurine and glycine conjugates of these cholic acids.
  • Cholic acid is a cholesterol metabolite that forms in the liver and is secreted into the duodenum, which plays an important role in the dissolution and absorption of food fats and vitamins.
  • Cholic acid downregulates the transcription of cytochrome P4507a (CYP7a), which encodes an enzyme that catalyzes the rate-limiting step of bile acid biosynthesis.
  • CYP7a cytochrome P4507a
  • FXR regulates the various enzymes of bile acid metabolism and bile salt carriers under the control of corresponding ligands, synergistic activating factors and hormones.
  • chenodeoxycholic acid (CDCA) is a commonly used natural agonist.
  • 6-ECDCA (5 ⁇ -3 ⁇ ,7 ⁇ -dihydroxy-6 ⁇ -ethyl-cholanoic acid), a derivative of CDCA, is a potentially potent FXR agonist, two orders of magnitude higher than CDCA.
  • PBC Primary biliary cirrhosis
  • ursodeoxycholic acid was the only FDA approved drug for the treatment of PBC, which effectively improved the level of abnormal liver biochemical markers and reduced the incidence of liver fibrosis and cirrhosis.
  • INT-747 is a novel drug that is used in patients who do not respond adequately or tolerate the old standard treatment drug ursodeoxycholic acid.
  • the drug has been granted orphan drug status by the US FDA, and if approved for marketing, it will receive a seven-year market franchise.
  • the FDA accelerated the approval of INT-747 (Obecholic Acid), which can be used alone to treat primary biliary cholangitis (PBC) that is intolerant to the existing standard therapy drug ursodeoxycholic acid.
  • PBC primary biliary cholangitis
  • Patients, or in combination with ursodeoxycholic acid were treated with PBC patients who did not respond adequately to ursodeoxycholic acid therapy.
  • the G protein coupled receptor 5 (TGR5) receptor is a G protein coupled receptor that has been identified as a Cell surface receptors that respond to bile acids (BA).
  • BA bile acids
  • the primary structure of the TGR5 and its responsiveness to bile acids have been found between human, bovine, rabbit, rat, and mouse G protein-coupled receptor 5 (TGR5). It is highly conserved and therefore suggests that TGR5 has important physiological functions.
  • TGR5 is involved in the intracellular accumulation of cyclic adenosine monophosphate (cAMP), which is widely expressed in a variety of different cell types.
  • cAMP cyclic adenosine monophosphate
  • the activation of the membrane receptor described in macrophages can reduce the production of pro-inflammatory cytokines
  • the stimulation of TGR5 by bile acids in fat cells and muscle cells can be increased. Energy consumption.
  • the latter effect involves cAMP-dependent induction of the type 2 potassium iodide adenine deiodinase (D2), which induces increased thyroid hormone activity by local conversion of T4 to T3 ( See Maruyama, T. et al., 2006, J. Endocrinol, Journal of Endocrinology, 191, 197-205).
  • D2 potassium iodide adenine deiodinase
  • TGR5 is an attractive targeting for the treatment of metabolic diseases, which may be obesity, diabetes, and metabolic syndrome.
  • Deuterated modification is a potentially attractive strategy for improving the metabolic properties of drugs.
  • Helium is a safe, stable, non-radiative isotope of hydrogen. Compared with the C-H bond, the C-D bond formed by ruthenium and carbon is stronger because of the lower vibration frequency.
  • the "heavy hydrogen" version of the drug may be more stable to degradation and longer in the body. Incorporating hydrazine to replace hydrogen can improve the pharmacodynamics and pharmacokinetic profile of the drug, altering the metabolic fate while maintaining the pharmacological activity and selectivity of the physiologically active compound.
  • Deuterated drugs have a positive impact on safety, efficacy and tolerance, and have excellent research prospects.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 And R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 and R 29 are each independently selected from "hydrogen (H), hydrazine (D) ""group;
  • X 1 , X 2 , X 3 , X 4 are independently selected from the group consisting of "hydrogen (H), hydrazine (D), methyl, CH 2 D, CHD 2 , CD 3 , CH 2 CH 3 , CHDCH 3 , CHDCH 2 a group consisting of D, CHDCHD 2 , CHDCD 3 , CD 2 CH 3 , CD 2 CH 2 D, CD 2 CHD 2 , CD 2 CD 3 ”;
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently hydrazine or hydrogen.
  • R 6 , R 7 , R 8 , R 9 , R 10 and R 11 are each independently hydrazine or hydrogen.
  • R 12 , R 13 , R 14 , R 15 and R 16 are each independently hydrazine or hydrogen.
  • R 17 , R 18 and R 29 are each independently hydrazine or hydrogen.
  • R 19 and R 20 are each independently hydrazine or hydrogen.
  • R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 and R 28 are each independently hydrazine or hydrogen.
  • X 1 , X 2 , X 3 , X 4 may be independently selected from alkyl groups which are deuterated one or more times.
  • R 1 , R 2 , R 3 , R 4 , R 5 are deuterium.
  • R 6 , R 7 , R 8 , R 9 , R 10 , R 11 are deuterium.
  • R 17 and R 18 are deuterium.
  • R 19 and R 20 are deuterium.
  • R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 are ⁇ .
  • the compound is selected from the group consisting of a compound or a pharmaceutically acceptable salt thereof, but is not limited to the following compounds:
  • the cerium isotope content of the cerium in the deuterated position is at least greater than the natural strontium isotope content (0.015%), preferably greater than 30%, more preferably greater than 50%, and even more preferably greater than 75%. More preferably greater than 95%, more preferably greater than 99%.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 and R 14 , R 15 , R 16 , R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 and R 29 are each in the metamorphic position
  • the isotope content is at least 5%, preferably more than 10%, more preferably more than 15%, more preferably more than 20%, more preferably more than 25%, more preferably more than 30%, more preferably more than 35%, more Preferably more than 40%, more preferably more than 45%, more preferably more than 50%, more preferably more than 55%, more preferably more than 60%, more preferably more than 65%, more preferably more than 70%, more preferably
  • the ground is greater than 75%, more preferably greater than 80%, more
  • the compound does not include a non-deuterated compound.
  • a method of preparing a pharmaceutical composition comprising the steps of: pharmaceutically acceptable carrier and a compound of the first aspect of the invention, or a crystalline form thereof, pharmaceutically acceptable
  • the accepted salt, hydrate or solvate is mixed to form a pharmaceutical composition.
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of the first aspect of the invention, or a crystalline form thereof, a pharmaceutically acceptable salt, hydrated Or a solvate.
  • Pharmaceutically acceptable carriers that can be used in the pharmaceutical compositions of the present invention include, but are not limited to, any glidants, sweeteners, diluents, preservatives, dyes/colorants, flavor enhancers, surfactants, wetting agents A dispersing agent, a disintegrating agent, a suspending agent, a stabilizer, an isotonic agent, a solvent or an emulsifier.
  • the pharmaceutical composition of the present invention can be formulated into solid, semi-solid, liquid or gaseous preparations, such as tablets, pills, capsules, powders, granules, ointments, emulsions, suspensions, solutions, suppositories, injections, inhalants, coagulation Glues, microspheres and aerosols.
  • Typical routes of administration of the pharmaceutical compositions of the invention include, but are not limited to, oral, rectal, transmucosal, enteral, or topical, transdermal, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal , intramuscular, subcutaneous, intravenous administration. Oral administration or injection administration is preferred.
  • the pharmaceutical composition of the present invention can be produced by a method known in the art, such as a conventional mixing method, a dissolution method, a granulation method, a sugar-coating method, a pulverization method, an emulsification method, a freeze-drying method, and the like.
  • halogen means F, Cl, Br, and I unless otherwise specified. More preferably, the halogen atom is selected from the group consisting of F, Cl and Br.
  • deuterated means that one or more hydrogens in the compound or group are replaced by deuterium; deuteration may be monosubstituted, disubstituted, polysubstituted or fully substituted.
  • deuteration may be monosubstituted, disubstituted, polysubstituted or fully substituted.
  • deuterated is used interchangeably with “one or more deuterated”.
  • non-deuterated compound means a compound containing a proportion of germanium atoms not higher than the natural helium isotope content (0.015%).
  • the invention also includes isotopically labeled compounds, equivalent to the original compounds disclosed herein.
  • isotopes which may be listed as compounds of the present invention include hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine isotopes such as 2 H, 3 H, 13 C, 14 C, 15 N, 17 O, 18 O, respectively. , 31 P, 32 P, 35 S, 18 F and 36 Cl. a compound, or an enantiomer, a diastereomer, an isomer, or a pharmaceutically acceptable salt or solvate of the present invention, wherein an isotope or other isotopic atom containing the above compound is within the scope of the present invention .
  • isotopically-labeled compounds of the present invention such as the radioisotopes of 3 H and 14 C, are also useful therein, and are useful in tissue distribution experiments of drugs and substrates. ⁇ , ie 3 H and carbon-14, ie 14 C, are easier to prepare and detect and are preferred in isotopes.
  • Isotopically labeled compounds can be prepared in a conventional manner by substituting a readily available isotopically labeled reagent with a non-isotopic reagent using the protocol of the examples.
  • Pharmaceutically acceptable salts include inorganic and organic salts.
  • a preferred class of salts are the salts of the compounds of the invention with acids.
  • Suitable acids for forming salts include, but are not limited to, mineral acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid; formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, Organic acids such as fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, benzoic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, benzenesulfonic acid, naphthalenesulfonic acid; Amino acids such as amino acid, phenylalanine, aspartic acid, and glutamic acid.
  • salts of the compounds of the invention with bases such as alkali metal salts (for example sodium or potassium salts), alkaline earth metal salts (for example magnesium or calcium salts), ammonium salts (for example lower alkanolammonium).
  • bases such as alkali metal salts (for example sodium or potassium salts), alkaline earth metal salts (for example magnesium or calcium salts), ammonium salts (for example lower alkanolammonium).
  • Salts and other pharmaceutically acceptable amine salts such as methylamine, ethylamine, propylamine, dimethylamine, trimethylamine, diethylamine, triethylamine, tert-butyl
  • a base amine salt an ethylenediamine salt, a hydroxyethylamine salt, a dihydroxyethylamine salt, a trihydroxyethylamine salt, and an amine salt formed of morpholine, piperazine, and lysine, respectively.
  • the compounds of the invention may have a chiral center, for example, a chiral carbon atom.
  • the compounds of the invention thus include racemic mixtures of all stereoisomers, including enantiomers, diastereomers and atropisomers. Additionally, the compounds of the invention include enriched or resolved optical isomers on any or all of the asymmetric chiral atoms. In other words, the chiral centers apparent from the description are provided as chiral isomers or racemic mixtures. Racemic mixtures and diastereomeric mixtures, as well as enantiomerically or diastereomeric partners which are substantially free of them, isolated or synthesized individual optical isomers, are all within the scope of the invention.
  • the racemic mixture is separated into their individual, substantially optical, by known techniques, for example, by separation of diastereomeric salts formed with optically active auxiliaries such as acids or bases, followed by conversion to optically active materials. Pure isomers. In most cases, the desired optical isomer is synthesized by stereospecific reaction starting from the appropriate stereoisomer of the desired starting material.
  • solvate refers to a complex of a compound of the invention that is coordinated to a solvent molecule to form a specific ratio.
  • Hydrophilate means a complex formed by the coordination of a compound of the invention with water.
  • the FXR or TGR5 mediated disease or condition is selected from the group consisting of chronic liver disease, gastrointestinal disease, kidney disease, cardiovascular disease, cholestatic disorder, metabolic disease; preferably, the kidney disease is diabetic nephropathy;
  • the disease is selected from the group consisting of arteriosclerosis, dyslipidemia, hypercholesterolemia, hypertriglyceridemia, in which arteriosclerosis is atherosclerosis.
  • the FXR or TGR5 mediated disease or condition is cardiovascular disease, atherosclerosis, arteriosclerosis, hypercholesterolemia, hyperlipidemia, chronic hepatitis disease, gastrointestinal disease, kidney disease, cardiovascular disease, metabolism Disease, cancer (eg, colorectal cancer), or signs of nerves such as stroke.
  • the chronic liver disease is primary sclerosis, cerebral palsy xanthomatosis, primary sclerosing gallbladder Inflammation, drug-induced cholestasis, intrahepatic cholestasis of pregnancy, parenteral absorption-related cholestasis, bacterial overgrowth or sepsis cholestasis, autoimmune hepatitis, chronic viral hepatitis, alcoholic liver disease, nonalcoholic fatty liver disease , nonalcoholic steatohepatitis, graft-versus-host disease associated with liver transplantation, regeneration of living donor liver transplantation, congenital liver fibrosis, common bile duct stones, granulomatous liver disease, intrahepatic- or extra-malignant tumor, Sjogren syndrome, Sarcoidosis, Wilson's disease, Gaucher's disease, hemochromatosis, or blood pressure nephropathy, chronic glomerulonephritis, chronic allograft glomerulopathy, chronic interstitial
  • the cardiovascular disease is arteriosclerosis, arteriosclerosis, dyslipidemia, hypercholesterolemia, or hypertriglyceridemia.
  • the metabolic disease is insulin resistance, type I and type II diabetes, or obesity.
  • the present invention has the beneficial effects that the compound of the present invention can be used to activate the farnesoid X receptor, indirectly inhibit the gene expression of cytochrome 7A1 (CYP7A1), and promote the synthesis of cholic acid. Furthermore, the compounds of the invention are capable of better modulating G protein coupled receptor 5 (TGR5). By deuteration this technique changes the metabolism of the compound in the organism, giving the compound a better pharmacokinetic parameter characteristic. In this case, the dosage can be changed and a long-acting preparation can be formed to improve the applicability.
  • TGR5 G protein coupled receptor 5
  • Replacing a hydrogen atom in a compound with hydrazine can increase the drug concentration of the compound in an animal to improve the efficacy of the drug due to its strontium isotope effect. Substitution of a hydrogen atom in a compound with hydrazine may increase the safety of the compound due to inhibition of certain metabolites.
  • each reaction is usually carried out in an inert solvent at room temperature to reflux temperature (e.g., 0 ° C to 100 ° C, preferably 0 ° C to 80 ° C).
  • the reaction time is usually from 0.1 to 60 hours, preferably from 0.5 to 24 hours.
  • Step 1 Synthesis of methyl ⁇ -hydroxy-7-keto-5 ⁇ -cholanoate (Compound 2).
  • Step 2 Synthesis of methyl ⁇ -7 ⁇ -di-trimethylsiloxy-5 ⁇ -cholanoate (Compound 3).
  • Trimethylchlorosilane (7.15 mL, 57.0 mmol) was added dropwise to a solution of lithium diisopropylamide (LDA 2M, 34.2 mL, 68.4 mmol) in anhydrous tetrahydrofuran (THF, 70 mL). minute. Then a mixture of methyl 3 ⁇ -hydroxy-7-keto-5 ⁇ -cholanoate (2.30 g, 5.7 mmol) in anhydrous tetrahydrofuran (30 mL) was added dropwise to the reaction mixture over 15 min. The reaction was further stirred for 1.5 hours.
  • LDA 2M lithium diisopropylamide
  • THF anhydrous tetrahydrofuran
  • Triethylamine (15 mL, 103 mmol) was added, and the reaction mixture was stirred at -78 ° C for further 1.5 hours. The reaction was warmed to -20 ° C, saturated aqueous sodium bicarbonate (20 mL) was then evaporated and evaporated. The organic layer was separated and the aqueous extracted with ethyl acetate (100 mL The combined organic layers were washed with a saturated aqueous solution of sodium bicarbonate (100 mL), water (100mL) Dry over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure.
  • Step 3 Synthesis of 3 ⁇ -hydroxy-6-ethylidene-7-keto-5 ⁇ -cholanoic acid methyl ester (Compound 4).
  • Step 4 Synthesis of 3 ⁇ -hydroxy-6 ⁇ -ethyl-7-keto-5 ⁇ -cholanoic acid methyl ester (Compound 5).
  • Step 5 Synthesis of 3 ⁇ -hydroxy-6 ⁇ -ethyl-7-keto-5 ⁇ -cholanoic acid (Compound 6).
  • Step 6 Synthesis of 3 ⁇ ,7 ⁇ -di-hydroxy-7-d-6 ⁇ -ethyl-5 ⁇ -cholanoic acid (Compound 7).
  • Step 1 Synthesis of 3 ⁇ -hydroxy-6 ⁇ -(ethyl-1-d)-6-d-7-keto-5 ⁇ -cholanoic acid methyl ester (Compound 8).
  • Step 2 Synthesis of 3 ⁇ -hydroxy-6 ⁇ -(ethyl-1-d)-7-keto-5 ⁇ -cholanoic acid (Compound 9).
  • Step 3 Synthesis of 3 ⁇ ,7 ⁇ -di-hydroxy-6 ⁇ -(ethyl-1-d)-5 ⁇ -cholanoic acid (Compound 10).
  • Step 1 Synthesis of 3 ⁇ -hydroxy-6 ⁇ -ethyl-6-d-7-keto-5 ⁇ -cholanoic acid (Compound 11).
  • Step 3 Synthesis of 3 ⁇ ,7 ⁇ -di-hydroxy-6 ⁇ -(ethyl-1-d)-5 ⁇ -cholanoic acid (Compound 12).
  • Step 1 Synthesis of 3 ⁇ -hydroxy-6 ⁇ -(ethyl-1-d)-6-d-7-keto-5 ⁇ -cholanoic acid (Compound 13).
  • Step 2 Synthesis of 3 ⁇ ,7 ⁇ -di-hydroxy-6 ⁇ -(ethyl-1-d)-6-d-5 ⁇ -cholanoic acid (Compound 14).
  • Step 1 Synthesis of 3 ⁇ -hydroxy-6-(ethylidene-1,2,2,2-d 4 )-7-keto-5 ⁇ -cholanoic acid methyl ester (Compound 16).
  • Step 2 Synthesis of 3 ⁇ -hydroxy-6 ⁇ -(ethyl-1,2,2,2-d 4 )-7-keto-5 ⁇ -cholanoic acid methyl ester (Compound 17).
  • Step 3 Synthesis of 3 ⁇ -hydroxy-6 ⁇ -(ethyl-1,2,2,2-d 4 )-7-keto-5 ⁇ -cholanoic acid (Compound 18).
  • Step 4 Synthesis of 3 ⁇ ,7 ⁇ -dihydroxy-6 ⁇ -(ethyl-1,2,2,2-d 4 )-ethyl-5 ⁇ -cholanoic acid (Compound 19).
  • Step 1 Synthesis of 3 ⁇ -hydroxy-6 ⁇ -(ethyl-1,2,2,2-d 4 )-6-d-7-keto-5 ⁇ -cholanoic acid (Compound 21).
  • Step 2 Synthesis of 3 ⁇ ,7 ⁇ -di-hydroxy-6 ⁇ -(ethyl-1,2,2,2-d 4 )-6-d-5 ⁇ -cholanoic acid (Compound 22).
  • Step 1 Synthesis of 3 ⁇ -hydroxy-6 ⁇ -(ethyl-1,1,2,2,2-d 5 )-6-d-7-keto-5 ⁇ -cholanoic acid methyl ester (Compound 23).
  • Step 2 Synthesis of 3 ⁇ -hydroxy-6 ⁇ -(ethyl-1,1,2,2,2-d 5 )-6-d-7-keto-5 ⁇ -cholanoic acid (Compound 24).
  • Step 3 Synthesis of 3 ⁇ ,7 ⁇ -di-hydroxy-7-d-6 ⁇ -(ethyl-1,1,2,2,2-d 5 )-6-d-5 ⁇ -cholanoic acid (Compound 25) .
  • Step 1 Synthesis of 3 ⁇ -hydroxy-6 ⁇ -(ethyl-1,1,2,2,2-d 5 )-7-keto-5 ⁇ -cholanoic acid (Compound 26).
  • Step 2 Synthesis of 3 ⁇ ,7 ⁇ -di-hydroxy-6 ⁇ -(ethyl-1,1,2,2,2-d 5 )-5 ⁇ -cholanoic acid (Compound 27).
  • FXR farnesoid X receptor
  • TGR5 G protein coupled receptor 5
  • FXR farnesoid X receptor
  • FRET fluorescence resonance energy transfer
  • TGR5 G protein coupled receptor 5
  • Luciferase activity is measured in Chinese hamster ovary (CHO) cells, wherein the CHO cells stably express human G protein-coupled receptor 5 (hTGR5), or the cells are utilized an hTGR5 expression vector And a luciferase receptor gene driven by the cAMP responsive element (CRE) was transiently co-transfected.
  • CHO Chinese hamster ovary
  • CRE cAMP responsive element
  • Plasmid The National Institutes of Health (NIH) mammalian gene-preserving clone MGC: 40597 (also known as pCMVSPORT6/hTGR5 or pTGR5) and pcDNA 3.1 (+) were obtained from Invitrogen (Carlsbad, CA).
  • pCRE-Luc cyclized adenosine monophosphate responsive element driven luciferase receptor plasmid
  • pCMV ⁇ were obtained from Clontech (Palo Alto, CA).
  • Transient transfection Chinese hamster ovary (CHO) cells were placed in 96-well culture plates at a density of 3.5 x 10 4 cells/well for 24 hours, and after that, 150 ng of humans were utilized in each well.
  • G protein coupled receptor 5 expression plasmid pCMVSPORT6/hTGR5
  • CRE circularized adenosine monophosphate
  • pCRE-Luc luciferase reporter plasmid
  • the cells were washed once with phosphate buffered saline (PBS) and the medium was exchanged for DMEM containing 0.1% (w/v) bovine serum albumin (BSA). After another 18 hours of incubation, the cells were treated with different concentrations of each compound for 5 hours, wherein the compound was present in fresh 0.1% (w/v) bovine serum albumin (BSA).
  • DMEM phosphate buffered saline
  • EDTA ethylenediaminetetraacetic acid
  • disulfide 50 ⁇ l of lysis buffer (25 mmol of Tris hydrochloric acid (pH 7.6), 2 mmol of ethylenediaminetetraacetic acid (EDTA), 1 mmol of disulfide was used through a freeze-thaw cycle.
  • Threitol (DTT), 10% (v/v) glycerol and 1% (v/v) Triton X-100) lyse the cells and render the method as described below
  • Luciferase assay and beta-galactosidase assay For luciferase assay, 20 ⁇ l of cell lysate with 100 ⁇ l of luciferase buffer 235 ⁇ M of luciferin, 265 ⁇ M Adenosine triphosphate (ATP) and 135 micromolar coenzyme A (CoA) were mixed and luminescence was measured using CentroXS3LB960 (Berthold Technologies, Bad Wildbad, Germany).
  • ATP Adenosine triphosphate
  • CoA micromolar coenzyme A
  • CHO-TGR5 cells Chinese hamster ovary (CHO-TGR5 cells) capable of stably expressing human G protein-coupled receptor 5: using Lipofectamin 2000, using 3.8 ⁇ g of hTGR5 expression plasmid (pCMVSPORT6/hTGR5), 3.8 ⁇ g of cAMP responsiveness
  • pCMVSPORT6/hTGR5 expression plasmid 3.8 ⁇ g of cAMP responsiveness
  • pCRE-Luc CRE-driven luciferase reporter plasmid
  • pcDNA3.1(+) a neomycin-resistant gene expression plasmid
  • the transfectants were screened with 400 ⁇ g/ml of G418 sulfate and individual clones were grown independently in 96-well culture plates.
  • the Chinese hamster ovary (CHO) cell line expressing G protein coupled receptor 5 (TGR5) was screened by life cycle assessment (LCA) treatment, after which luciferase assay was performed.
  • EC 50 50% effective concentration
  • the EC 50 value is determined by probabilistic unit analysis.
  • the efficiency was determined by the following method: For the study of TGR5 agonist, by calculating the percentage of the value of 10 micromoles of lithocholic acid (LCA). After performing the conversion algorithm, the completion of the comparison of the calculated average EC 50 and EC for various compounds 50 has performed.
  • Microsomal experiments human liver microsomes: 0.5 mg/mL, Xenotech; rat liver microsomes: 0.5 mg/mL, Xenotech; coenzyme (NADPH/NADH): 1 mM, Sigma Life Science; magnesium chloride: 5 mM, 100 mM phosphate buffer Agent (pH 7.4).
  • phosphate buffer 100 mM, pH 7.4.
  • the pH was adjusted to 7.4, diluted 5 times with ultrapure water before use, and magnesium chloride was added to obtain a phosphate buffer (100 mM) containing 100 mM potassium phosphate, 3.3 mM magnesium chloride, and a pH of 7.4.
  • NADPH regeneration system containing 6.5 mM NADP, 16.5 mM G-6-P, 3 U/mL G-6-P D, 3.3 mM magnesium chloride was prepared and placed on wet ice before use.
  • Formulation stop solution acetonitrile solution containing 50 ng/mL propranolol hydrochloride and 200 ng/mL tolbutamide (internal standard). Take 25057.5 ⁇ L of phosphate buffer (pH 7.4) into a 50 mL centrifuge tube, add 812.5 ⁇ L of human liver microsomes, and mix to obtain a liver microsome dilution with a protein concentration of 0.625 mg/mL. 25057.5 ⁇ L of phosphate buffer (pH 7.4) was taken into a 50 mL centrifuge tube, and 812.5 ⁇ L of SD rat liver microsomes were added and mixed to obtain a liver microsome dilution having a protein concentration of 0.625 mg/mL.
  • the corresponding compound had a reaction concentration of 1 ⁇ M and a protein concentration of 0.5 mg/mL.
  • 100 ⁇ L of the reaction solution was taken at 10, 30, and 90 min, respectively, and added to the stopper, and the reaction was terminated by vortexing for 3 min.
  • the plate was centrifuged at 5000 x g for 10 min at 4 °C.
  • 100 ⁇ L of the supernatant was taken into a 96-well plate to which 100 ⁇ L of distilled water was previously added, mixed, and sample analysis was performed by LC-MS/MS.
  • Rats were fed a standard diet and given water. Fasting began 16 hours before the test.
  • the drug was dissolved with PEG400 and dimethyl sulfoxide. Blood was collected from the eyelids at a time point of 0.083 hours, 0.25 hours, 0.5 hours, 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, and 24 hours after administration.
  • Rats were briefly anesthetized after inhalation of ether, and 300 ⁇ L of blood samples were collected from the eyelids in test tubes. There was 30 ⁇ L of 1% heparin salt solution in the test tube. The tubes were dried overnight at 60 ° C before use. After the blood sample collection was completed at a later time point, the rats were anesthetized with ether and sacrificed.
  • Plasma samples were centrifuged at 5000 rpm for 5 minutes at 4 ° C to separate plasma from red blood cells. Pipette 100 ⁇ L of plasma into a clean plastic centrifuge tube to indicate the name and time point of the compound. Plasma was stored at -80 °C prior to analysis. The concentration of the compound of the invention in plasma was determined by LC-MS/MS. Pharmacokinetic parameters were calculated based on the plasma concentration of each animal at different time points.

Abstract

The present invention relates to a cholanic acid compound for preventing or treating FXR-mediated diseases. Specifically, disclosed are a cholanic acid compound represented by formula (I) and a pharmaceutical composition containing the compound, or a crystalline form, a pharmaceutically-acceptable salt, a prodrug, a tautomer, a stereoisomer, an enantiomer, and a hydrate or a solvate thereof. The compound in the present invention can be used as a farnesol X receptor agonist and can therefore be applied to the preparation of drugs for treating FXR-related diseases (for example, fatty liver).

Description

用于预防或治疗FXR-介导疾病的胆烷酸化合物Cholenic acid compounds for the prevention or treatment of FXR-mediated diseases 技术领域Technical field
本发明属于医药领域。具体地,本发明涉及一种用于预防或治疗FXR-介导疾病的胆烷酸化合物及其药物组合物及用途。The invention belongs to the field of medicine. In particular, the present invention relates to a cyanic acid compound and a pharmaceutical composition thereof for use in the prevention or treatment of FXR-mediated diseases and uses thereof.
背景技术Background technique
法尼醇X受体(FXR)是孤儿核受体家族成员,最早由Forman等于1995年发现,因其转录活性可被超生理浓度的法尼醇增强而命名。Northern和原位分析显示FXR在肺,肠,肾,和肾上腺中大量表达。FXR与9-顺式维生素A酸受体(RXR)形成异源二聚体与DNA结合。FXR/RXR异源二聚体优先与由共有AG(G/T)TCA的双核受体半位点组成的成分结合,其形成反向重复并祓单一核苷分离(IR-1模体)。然而,这些化合物无法激活小鼠和人类FXR,使得内源性FXR配基的自然性还不确定。一些自然发生的胆酸在生理浓度下结合并激活FXR。作为FXR配基的胆酸包括鹅脱氧胆酸(CDCA),脱氧胆酸(DCA),石胆酸(LCA),和这些胆酸的牛磺酸及氨基乙酸共轭物。Farnesol X receptor (FXR), a member of the orphan nuclear receptor family, was first discovered by Forman et al in 1995, and its transcriptional activity can be named by the increase in superphysiological concentration of farnesol. Northern and in situ analysis revealed extensive expression of FXR in the lung, intestine, kidney, and adrenal glands. FXR forms a heterodimer with the 9-cis retinoic acid receptor (RXR) to bind to DNA. The FXR/RXR heterodimer preferentially binds to a component consisting of a binuclear receptor half site that shares AG(G/T) TCA, which forms an inverted repeat and a single nucleoside separation (IR-1 motif). However, these compounds fail to activate mouse and human FXR, making the natural nature of endogenous FXR ligands uncertain. Some naturally occurring bile acids bind to and activate FXR at physiological concentrations. Cholic acids as FXR ligands include chenodeoxycholic acid (CDCA), deoxycholic acid (DCA), lithocholic acid (LCA), and taurine and glycine conjugates of these cholic acids.
胆酸是在肝脏中形成并分泌入十二指肠的胆固醇代谢产物,其在食物脂肪和维生素的溶解和吸收中具有重要作用。胆酸下调细胞色素P4507a(CYP7a)的转录,其编码催化胆酸生物合成限速步骤的酶。数据显示FXR与胆酸对CYP7a表达的抑制有关,虽然精确的机制仍未明确。FXR在相应配体、协同活化因子及激素的调控下,对胆汁酸代谢的多种酶和胆盐载体进行精密的调控。近年来发现生理浓度的多种初级和次级胆汁酸可激活FXR,例如,鹅去氧胆酸(chenodexycholic acid,CDCA)是常用的一种天然激动剂。6-ECDCA(5β-3α,7α-二羟基-6α-乙基-胆烷酸)作为CDCA的衍生物,是一种潜在的高效FXR激动剂,比CDCA高两个数量级。Cholic acid is a cholesterol metabolite that forms in the liver and is secreted into the duodenum, which plays an important role in the dissolution and absorption of food fats and vitamins. Cholic acid downregulates the transcription of cytochrome P4507a (CYP7a), which encodes an enzyme that catalyzes the rate-limiting step of bile acid biosynthesis. The data show that FXR is associated with the inhibition of CYP7a expression by cholic acid, although the precise mechanism remains unclear. FXR regulates the various enzymes of bile acid metabolism and bile salt carriers under the control of corresponding ligands, synergistic activating factors and hormones. In recent years, various primary and secondary bile acids at physiological concentrations have been found to activate FXR. For example, chenodeoxycholic acid (CDCA) is a commonly used natural agonist. 6-ECDCA (5β-3α,7α-dihydroxy-6α-ethyl-cholanoic acid), a derivative of CDCA, is a potentially potent FXR agonist, two orders of magnitude higher than CDCA.
原发性胆汁性肝硬化(PBC)是一种常见的自身免疫性肝胆疾病,主要发生于40岁以上的中年女性,发病率为0.1%。PBC与胆汁淤积有关,主要由胆汁摄取、合成或分泌障碍引起,也可因胆道梗阻形成,并逐步发展为肝内小胆管炎症损伤,最终导致硬化。在INT747上市之前,熊去氧胆酸是FDA唯一批准用于治疗PBC的药物,它能够有效改善肝脏异常生化指标的水平,并降低肝纤维化和肝硬化的发病率。INT-747是一种新型药物,研究用于那些对旧标准治疗药物熊去氧胆酸没有充分应答或不能耐受的患者。该药物已被美国FDA授予孤儿药资格,若获得上市批准,它将获得7年的市场专营权。2016年5月27日,FDA加速批准INT-747(奥贝胆酸)上市,可以单独使用治疗对现有标准疗法药物熊去氧胆酸不耐受的原发性胆汁性胆管炎(PBC)患者,或者与熊去氧胆酸联用治疗对熊去氧胆酸治疗没有充分应答的PBC患者。Primary biliary cirrhosis (PBC) is a common autoimmune hepatobiliary disease that occurs mainly in middle-aged women over the age of 40 with an incidence of 0.1%. PBC is associated with cholestasis, mainly caused by bile uptake, synthesis or secretion disorders, but also due to biliary obstruction, and gradually develop into intrahepatic small bile duct inflammation damage, which eventually leads to hardening. Prior to the INT747, ursodeoxycholic acid was the only FDA approved drug for the treatment of PBC, which effectively improved the level of abnormal liver biochemical markers and reduced the incidence of liver fibrosis and cirrhosis. INT-747 is a novel drug that is used in patients who do not respond adequately or tolerate the old standard treatment drug ursodeoxycholic acid. The drug has been granted orphan drug status by the US FDA, and if approved for marketing, it will receive a seven-year market franchise. On May 27, 2016, the FDA accelerated the approval of INT-747 (Obecholic Acid), which can be used alone to treat primary biliary cholangitis (PBC) that is intolerant to the existing standard therapy drug ursodeoxycholic acid. Patients, or in combination with ursodeoxycholic acid, were treated with PBC patients who did not respond adequately to ursodeoxycholic acid therapy.
G蛋白偶联受体5(TGR5)受体是一种G蛋白偶联受体,其已经被识别为是一种 细胞表面受体,对胆汁酸(BA)进行应答。在人类、牛、兔、大鼠、以及小鼠的G蛋白偶联受体5(TGR5)之间,已经发现了所述的TGR5所具有的一级结构以及它对胆汁酸所产生的应答性是高度保守的,并且因此提示了TGR5具有重要的生理学功能。The G protein coupled receptor 5 (TGR5) receptor is a G protein coupled receptor that has been identified as a Cell surface receptors that respond to bile acids (BA). The primary structure of the TGR5 and its responsiveness to bile acids have been found between human, bovine, rabbit, rat, and mouse G protein-coupled receptor 5 (TGR5). It is highly conserved and therefore suggests that TGR5 has important physiological functions.
TGR5与环化腺核苷一磷酸(cAMP)的细胞内积累有关,其中所述的cAMP在各种不同的细胞类型中进行广泛的表达。尽管所述的这种膜受体在巨噬细胞中所具有的活化作用能够降低促炎性细胞因子的生成,但是胆汁酸在脂肪细胞以及肌细胞中造成的所述的TGR5的刺激作用能够增加能量的消耗。后一种作用涉及到所述的2型碘化钾腺氨酸脱碘酶(D2)的cAMP依赖性诱导作用,这种诱导作用通过局部的将T4转化成T3的方式引起增加的甲状腺激素的活性(参见Maruyama,T.等人于2006年在J.Endocrinol《内分泌学杂志》191,197-205中发表的文章)。据此,TGR5是一种用于进行代谢疾病的治疗的有吸引力的靶向作用,其中所述的代谢疾病可以是,肥胖症,糖尿病以及代谢综合症。TGR5 is involved in the intracellular accumulation of cyclic adenosine monophosphate (cAMP), which is widely expressed in a variety of different cell types. Although the activation of the membrane receptor described in macrophages can reduce the production of pro-inflammatory cytokines, the stimulation of TGR5 by bile acids in fat cells and muscle cells can be increased. Energy consumption. The latter effect involves cAMP-dependent induction of the type 2 potassium iodide adenine deiodinase (D2), which induces increased thyroid hormone activity by local conversion of T4 to T3 ( See Maruyama, T. et al., 2006, J. Endocrinol, Journal of Endocrinology, 191, 197-205). Accordingly, TGR5 is an attractive targeting for the treatment of metabolic diseases, which may be obesity, diabetes, and metabolic syndrome.
氘代修饰是改进药物代谢性质的一种有潜在吸引力的策略。氘是氢的安全、稳定、非辐射性的同位素。与C-H键相比,氘与碳形成的C-D键因为振动频率较低,所以较强。此外,药物的“重氢”型式可能对降解更稳定且在生物体内维持更长时间。并入氘来代替氢可改善药物的药效学和药代动力学概况,改变代谢归宿,同时保持生理学活性化合物的药理活性和选择性。氘化药物可对安全性、功效及耐受性都有正面影响,具有优良的研究前景。Deuterated modification is a potentially attractive strategy for improving the metabolic properties of drugs. Helium is a safe, stable, non-radiative isotope of hydrogen. Compared with the C-H bond, the C-D bond formed by ruthenium and carbon is stronger because of the lower vibration frequency. In addition, the "heavy hydrogen" version of the drug may be more stable to degradation and longer in the body. Incorporating hydrazine to replace hydrogen can improve the pharmacodynamics and pharmacokinetic profile of the drug, altering the metabolic fate while maintaining the pharmacological activity and selectivity of the physiologically active compound. Deuterated drugs have a positive impact on safety, efficacy and tolerance, and have excellent research prospects.
发明内容Summary of the invention
本发明的目的是提供一种用于预防或治疗由FXR或者TGR5介导疾病的取代的胆烷酸化合物及其药物组合物及用途。It is an object of the present invention to provide a substituted clavonic acid compound for use in the prevention or treatment of a disease mediated by FXR or TGR5, and a pharmaceutical composition thereof and use thereof.
对此,本发明采用的技术方案如下:In this regard, the technical solution adopted by the present invention is as follows:
本发明的第一方面中,提供了一种式(I)所示的胆烷酸化合物,或其晶型、药学上可接受的盐、水合物或溶剂化合物。In a first aspect of the invention, there is provided a cholanoic acid compound of the formula (I), or a crystalline form thereof, a pharmaceutically acceptable salt, a hydrate or a solvent compound.
Figure PCTCN2016109851-appb-000001
Figure PCTCN2016109851-appb-000001
式中: In the formula:
R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15、R16、R17、R18、R19、R20、R21、R22、R23、R24、R25、R26、R27、R28和R29相互独立地选自由“氢(H)、氘(D)”组成的组;R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 And R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 and R 29 are each independently selected from "hydrogen (H), hydrazine (D) ""group;
X1、X2、X3、X4相互独立地选自由“氢(H)、氘(D)、甲基、CH2D、CHD2、CD3、CH2CH3、CHDCH3、CHDCH2D、CHDCHD2、CHDCD3、CD2CH3、CD2CH2D、CD2CHD2、CD2CD3”组成的组;X 1 , X 2 , X 3 , X 4 are independently selected from the group consisting of "hydrogen (H), hydrazine (D), methyl, CH 2 D, CHD 2 , CD 3 , CH 2 CH 3 , CHDCH 3 , CHDCH 2 a group consisting of D, CHDCHD 2 , CHDCD 3 , CD 2 CH 3 , CD 2 CH 2 D, CD 2 CHD 2 , CD 2 CD 3 ”;
及其生理学上可接受的盐、前药、互变异构体和立体异构体、水合物或溶剂合物,包括这些化合物以所有比例形成的混合物。And physiologically acceptable salts, prodrugs, tautomers and stereoisomers, hydrates or solvates thereof, including mixtures of these compounds in all ratios.
作为本发明的进一步改进,R1、R2、R3、R4、R5各自独立地为氘或氢。As a further improvement of the present invention, R 1 , R 2 , R 3 , R 4 and R 5 are each independently hydrazine or hydrogen.
作为本发明的进一步改进,R6、R7、R8、R9、R10、R11各自独立地为氘或氢。As a further improvement of the present invention, R 6 , R 7 , R 8 , R 9 , R 10 and R 11 are each independently hydrazine or hydrogen.
作为本发明的进一步改进,R12、R13、R14、R15、R16各自独立地为氘或氢。As a further improvement of the present invention, R 12 , R 13 , R 14 , R 15 and R 16 are each independently hydrazine or hydrogen.
作为本发明的进一步改进,R17、R18和R29各自独立地为氘或氢。As a further improvement of the present invention, R 17 , R 18 and R 29 are each independently hydrazine or hydrogen.
作为本发明的进一步改进,R19、R20各自独立地为氘或氢。As a further improvement of the present invention, R 19 and R 20 are each independently hydrazine or hydrogen.
作为本发明的进一步改进,R21、R22、R23、R24、R25、R26、R27、R28各自独立地为氘或氢。As a further improvement of the present invention, R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 and R 28 are each independently hydrazine or hydrogen.
作为本发明的进一步改进,X1、X2、X3、X4可独立选自一次或多次氘代的烷基。As a further improvement of the present invention, X 1 , X 2 , X 3 , X 4 may be independently selected from alkyl groups which are deuterated one or more times.
在另一选例中,R1、R2、R3、R4、R5是氘。In another alternative, R 1 , R 2 , R 3 , R 4 , R 5 are deuterium.
在另一选例中,R6、R7、R8、R9、R10、R11是氘。In another alternative, R 6 , R 7 , R 8 , R 9 , R 10 , R 11 are deuterium.
在另一选例中,R17、R18是氘。In another alternative, R 17 and R 18 are deuterium.
在另一选例中,R19、R20是氘。In another alternative, R 19 and R 20 are deuterium.
在另一选例中,R21、R22、R23、R24、R25、R26、R27、R28是氘。In another alternative, R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 are 氘.
在另一选例中,所述化合物选自如下组化合物或其药学上可接受的盐,但不局限于下列化合物:In another embodiment, the compound is selected from the group consisting of a compound or a pharmaceutically acceptable salt thereof, but is not limited to the following compounds:
Figure PCTCN2016109851-appb-000002
Figure PCTCN2016109851-appb-000002
Figure PCTCN2016109851-appb-000003
Figure PCTCN2016109851-appb-000003
Figure PCTCN2016109851-appb-000004
Figure PCTCN2016109851-appb-000004
Figure PCTCN2016109851-appb-000005
Figure PCTCN2016109851-appb-000005
Figure PCTCN2016109851-appb-000006
Figure PCTCN2016109851-appb-000006
反应在另一优选例中,氘在氘代位置的氘同位素含量至少是大于天然氘同位素含量(0.015%),较佳地大于30%,更佳地大于50%,更佳地大于75%,更佳地大于95%,更佳地大于99%。In another preferred embodiment, the cerium isotope content of the cerium in the deuterated position is at least greater than the natural strontium isotope content (0.015%), preferably greater than 30%, more preferably greater than 50%, and even more preferably greater than 75%. More preferably greater than 95%, more preferably greater than 99%.
具体地说,在本发明中R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15、R16、R17、R18、R19、R20、R21、R22、R23、R24、R25、R26、R27、R28和R29各氘代位置中氘同位素含量至少是5%,较佳地大于10%,更佳地大于15%,更佳地大于20%,更佳地大于25%,更佳地大于30%,更佳地大于35%,更佳地大于40%,更佳地大于45%,更佳地大于50%,更佳地大于55%,更佳地大于60%,更佳地大于65%,更佳地大于70%,更佳地大于75%,更佳地大于80%,更佳地大于85%,更佳地大于90%,更佳地大于95%,更佳地大于99%。Specifically, in the present invention, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 and R 14 , R 15 , R 16 , R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 and R 29 are each in the metamorphic position The isotope content is at least 5%, preferably more than 10%, more preferably more than 15%, more preferably more than 20%, more preferably more than 25%, more preferably more than 30%, more preferably more than 35%, more Preferably more than 40%, more preferably more than 45%, more preferably more than 50%, more preferably more than 55%, more preferably more than 60%, more preferably more than 65%, more preferably more than 70%, more preferably The ground is greater than 75%, more preferably greater than 80%, more preferably greater than 85%, more preferably greater than 90%, more preferably greater than 95%, and even more preferably greater than 99%.
在另一优选例中,式(I)中化合物的R R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15、R16、R17、R18、R19、R20、R21、R22、R23、R24、R25、R26、R27、R28和R29,至少其中一个R含氘,更佳地两个R含氘,更佳地三个R含氘,更佳地四个R含氘,更佳地五个R含氘,更佳地六个R含氘,更佳地七个R含氘,更佳地八个R含氘,更佳地九个R含氘,更佳地十个R含氘,更佳地十一个R含氘,更佳地十二个R含氘,更佳地十三个R含氘,更佳地十四个R含氘,更佳地十五个R含氘,更佳地十六个R含氘,更佳地十七个R含氘,更佳地十八个R含氘,更佳地十九个R含氘,更佳地二十个R含氘,更佳地二十一个R含氘,更佳地二十二个R含氘,更佳地二十三个R含氘,更佳地二十四个R含氘,更佳地二十五个R含氘,更佳地二十六个R含氘,更佳地二十七个R含氘,更佳地二十八个R含氘,更佳地二十九个R含氘。In another preferred embodiment, R R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 of the compound of formula (I), R 13 , R 14 , R 15 , R 16 , R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 and R 29 , at least one of R contains ruthenium, more preferably two R contains ruthenium, more preferably three R contains ruthenium, more preferably four R contains ruthenium, more preferably five R contains ruthenium, more preferably six R氘, preferably seven R 氘, more preferably eight R 氘, more preferably nine R 氘, more preferably ten R 氘, more preferably eleven R 氘, more Preferably, twelve R 氘, preferably thirteen R 氘, more preferably fourteen R 氘, more preferably fifteen R 氘, more preferably sixteen R 氘, more The seventeen Rs contain 氘, preferably 18 R, more preferably 19 R, more preferably 20 氘, more preferably 21 R More preferably twenty-two R containing bismuth, more preferably twenty-three R containing bismuth, more preferably twenty-four R containing bismuth, more preferably twenty-five R containing bismuth, more preferably twenty-six R contains 氘, better two Seventeen R-containing strontiums, more preferably twenty-eight R 氘, more preferably twenty-nine R 氘.
在另一优选例中,所述化合物不包括非氘代化合物。 In another preferred embodiment, the compound does not include a non-deuterated compound.
在本发明的第二方面中,提供了一种制备药物组合物的方法,包括步骤:将药学上可接受的载体与本发明第一方面中所述的化合物,或其晶型、药学上可接受的盐、水合物或溶剂合物进行混合,从而形成药物组合物。In a second aspect of the invention, there is provided a method of preparing a pharmaceutical composition comprising the steps of: pharmaceutically acceptable carrier and a compound of the first aspect of the invention, or a crystalline form thereof, pharmaceutically acceptable The accepted salt, hydrate or solvate is mixed to form a pharmaceutical composition.
在本发明的第三方面中,提供了一种药物组合物,它含有药学上可接受的载体和本发明第一方面中所述的化合物,或其晶型、药学上可接受的盐、水合物或溶剂合物。In a third aspect of the invention, there is provided a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of the first aspect of the invention, or a crystalline form thereof, a pharmaceutically acceptable salt, hydrated Or a solvate.
可用于本发明药物组合物中的药学上可接受的载体包括但不限于任何助流剂、增甜剂、稀释剂、防腐剂、染料/着色剂、矫味增强剂、表面活性剂、润湿剂、分散剂、崩解剂、助悬剂、稳定剂、等渗剂、溶剂或乳化剂。Pharmaceutically acceptable carriers that can be used in the pharmaceutical compositions of the present invention include, but are not limited to, any glidants, sweeteners, diluents, preservatives, dyes/colorants, flavor enhancers, surfactants, wetting agents A dispersing agent, a disintegrating agent, a suspending agent, a stabilizer, an isotonic agent, a solvent or an emulsifier.
本发明药物组合物可配制成固态、半固态、液态或气态制剂,如片剂、丸剂、胶囊剂、粉剂、颗粒剂、膏剂、乳剂、悬浮剂、溶液剂、栓剂、注射剂、吸入剂、凝胶剂、微球及气溶胶等。The pharmaceutical composition of the present invention can be formulated into solid, semi-solid, liquid or gaseous preparations, such as tablets, pills, capsules, powders, granules, ointments, emulsions, suspensions, solutions, suppositories, injections, inhalants, coagulation Glues, microspheres and aerosols.
给予本发明药物组合物的典型途径包括但不限于口服、直肠、透黏膜、经肠给药,或者局部、经皮、吸入、肠胃外、舌下、阴道内、鼻内、眼内、腹膜内、肌内、皮下、静脉内给药。优选口服给药或注射给药。Typical routes of administration of the pharmaceutical compositions of the invention include, but are not limited to, oral, rectal, transmucosal, enteral, or topical, transdermal, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal , intramuscular, subcutaneous, intravenous administration. Oral administration or injection administration is preferred.
本发明的药物组合物可以采用本领域周知的方法制造,如常规的混合法、溶解法、制粒法、制糖衣药丸法、磨细法、乳化法、冷冻干燥法等。The pharmaceutical composition of the present invention can be produced by a method known in the art, such as a conventional mixing method, a dissolution method, a granulation method, a sugar-coating method, a pulverization method, an emulsification method, a freeze-drying method, and the like.
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。It is to be understood that within the scope of the present invention, the various technical features of the present invention and the various technical features specifically described hereinafter (as in the embodiments) may be combined with each other to constitute a new or preferred technical solution. Due to space limitations, we will not repeat them here.
本文中,如无特别说明,“卤素”指F、Cl、Br、和I。更佳地,卤原子选自F、Cl和Br。Herein, "halogen" means F, Cl, Br, and I unless otherwise specified. More preferably, the halogen atom is selected from the group consisting of F, Cl and Br.
本文中,如无特别说明,“氘代”指化合物或基团中的一个或多个氢被氘所取代;氘代可以是一取代、二取代、多取代或全取代。术语“一个或多个氘代的”与“一次或多次氘代”可互换使用。As used herein, unless otherwise specified, "deuterated" means that one or more hydrogens in the compound or group are replaced by deuterium; deuteration may be monosubstituted, disubstituted, polysubstituted or fully substituted. The terms "one or more deuterated" are used interchangeably with "one or more deuterated".
本文中,如无特别说明,“非氘代的化合物”是指含氘原子比例不高于天然氘同位素含量(0.015%)的化合物。As used herein, unless otherwise specified, "non-deuterated compound" means a compound containing a proportion of germanium atoms not higher than the natural helium isotope content (0.015%).
本发明还包括同位素标记的化合物,等同于原始化合物在此公开。可以列为本发明的化合物同位素的例子包括氢,碳,氮,氧,磷,硫,氟和氯同位素,分别如2H,3H,13C,14C,15N,17O,18O,31P,32P,35S,18F以及36Cl。本发明中的化合物,或对映体,非对映体,异构体,或药学上可接受的盐或溶剂化物,其中含有上述化合物的同位素或其他其他同位素原子都在本发明的范围之内。本发明中某些同位素标记化合物,例如3H和14C的放射性同位素也在其中,在药物和底物的组织分布实验中是有 用的。氚,即3H和碳-14,即14C,它们的制备和检测比较容易,是同位素中的首选。同位素标记的化合物可以用一般的方法,通过用易得的同位素标记试剂替换为非同位素的试剂,用示例中的方案可以制备。The invention also includes isotopically labeled compounds, equivalent to the original compounds disclosed herein. Examples of isotopes which may be listed as compounds of the present invention include hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine isotopes such as 2 H, 3 H, 13 C, 14 C, 15 N, 17 O, 18 O, respectively. , 31 P, 32 P, 35 S, 18 F and 36 Cl. a compound, or an enantiomer, a diastereomer, an isomer, or a pharmaceutically acceptable salt or solvate of the present invention, wherein an isotope or other isotopic atom containing the above compound is within the scope of the present invention . Certain isotopically-labeled compounds of the present invention, such as the radioisotopes of 3 H and 14 C, are also useful therein, and are useful in tissue distribution experiments of drugs and substrates.氚, ie 3 H and carbon-14, ie 14 C, are easier to prepare and detect and are preferred in isotopes. Isotopically labeled compounds can be prepared in a conventional manner by substituting a readily available isotopically labeled reagent with a non-isotopic reagent using the protocol of the examples.
药学上可接受的盐包括无机盐和有机盐。一类优选的盐是本发明化合物与酸形成的盐。适合形成盐的酸包括但并不限于:盐酸、氢溴酸、氢氟酸、硫酸、硝酸、磷酸等无机酸;甲酸、乙酸、三氟乙酸、丙酸、草酸、丙二酸、琥珀酸、富马酸、马来酸、乳酸、苹果酸、酒石酸、柠檬酸、苦味酸、苯甲酸、甲磺酸、乙磺酸、对甲苯磺酸、苯磺酸、萘磺酸等有机酸;以及脯氨酸、苯丙氨酸、天冬氨酸、谷氨酸等氨基酸。另一类优选的盐是本发明化合物与碱形成的盐,例如碱金属盐(例如钠盐或钾盐)、碱土金属盐(例如镁盐或钙盐)、铵盐(如低级的烷醇铵盐以及其它药学上可接受的胺盐),例如甲胺盐、乙胺盐、丙胺盐、二甲基胺盐、三甲基胺盐、二乙基胺盐、三乙基胺盐、叔丁基胺盐、乙二胺盐、羟乙胺盐、二羟乙胺盐、三羟乙胺盐,以及分别由吗啉、哌嗪、赖氨酸形成的胺盐。Pharmaceutically acceptable salts include inorganic and organic salts. A preferred class of salts are the salts of the compounds of the invention with acids. Suitable acids for forming salts include, but are not limited to, mineral acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid; formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, Organic acids such as fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, benzoic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, benzenesulfonic acid, naphthalenesulfonic acid; Amino acids such as amino acid, phenylalanine, aspartic acid, and glutamic acid. Another preferred class of salts are the salts of the compounds of the invention with bases, such as alkali metal salts (for example sodium or potassium salts), alkaline earth metal salts (for example magnesium or calcium salts), ammonium salts (for example lower alkanolammonium). Salts and other pharmaceutically acceptable amine salts), such as methylamine, ethylamine, propylamine, dimethylamine, trimethylamine, diethylamine, triethylamine, tert-butyl A base amine salt, an ethylenediamine salt, a hydroxyethylamine salt, a dihydroxyethylamine salt, a trihydroxyethylamine salt, and an amine salt formed of morpholine, piperazine, and lysine, respectively.
本发明化合物可具有手性中心,例如,手性碳原子。本发明化合物因此包括所有立体异构体的外消旋混合物,包括对映体、非对映体和阻转异构体。另外,本发明化合物包括在任意或全部不对称手性原子上的富集的或拆分的旋光异构体。换句话说,从描述中显而易见的手性中心被作为手性异构体或外消旋混合物提供。外消旋混合物和非对映的混合物,以及实质上不含它们的对映或非对映的配偶体、分离或合成的单独旋光异构体,全部落在本发明的保护范围内。通过已知技术,例如,分离用旋光活性的助剂例如,酸或碱形成的非对映的盐,接着转回为旋光活性物质,外消旋混合物被分离成他们的单个的、实质上光学纯的同分异构体。在大多数情况下,想得到的旋光异构体是通过立体特异反应,从希望的原料的适宜立体异构体开始进行合成的。The compounds of the invention may have a chiral center, for example, a chiral carbon atom. The compounds of the invention thus include racemic mixtures of all stereoisomers, including enantiomers, diastereomers and atropisomers. Additionally, the compounds of the invention include enriched or resolved optical isomers on any or all of the asymmetric chiral atoms. In other words, the chiral centers apparent from the description are provided as chiral isomers or racemic mixtures. Racemic mixtures and diastereomeric mixtures, as well as enantiomerically or diastereomeric partners which are substantially free of them, isolated or synthesized individual optical isomers, are all within the scope of the invention. The racemic mixture is separated into their individual, substantially optical, by known techniques, for example, by separation of diastereomeric salts formed with optically active auxiliaries such as acids or bases, followed by conversion to optically active materials. Pure isomers. In most cases, the desired optical isomer is synthesized by stereospecific reaction starting from the appropriate stereoisomer of the desired starting material.
术语“溶剂合物”指本发明化合物与溶剂分子配位形成特定比例的配合物。“水合物”是指本发明化合物与水进行配位形成的配合物。The term "solvate" refers to a complex of a compound of the invention that is coordinated to a solvent molecule to form a specific ratio. "Hydrate" means a complex formed by the coordination of a compound of the invention with water.
优选地,所述FXR或者TGR5介导的疾病或状况选自慢性肝病,胃肠疾病,肾病,心血管疾病,淤胆失调,代谢疾病;优选地,所述肾病为糖尿病肾病;所述心血管疾病选自动脉硬化,血脂障碍,高胆固醇血症,高甘油三酯血症,其中动脉硬化为动脉粥样硬化。Preferably, the FXR or TGR5 mediated disease or condition is selected from the group consisting of chronic liver disease, gastrointestinal disease, kidney disease, cardiovascular disease, cholestatic disorder, metabolic disease; preferably, the kidney disease is diabetic nephropathy; The disease is selected from the group consisting of arteriosclerosis, dyslipidemia, hypercholesterolemia, hypertriglyceridemia, in which arteriosclerosis is atherosclerosis.
在特定实施方案中,FXR或者TGR5介导的疾病或状况是心血管疾病,动脉粥样硬化,动脉硬化,高胆甾醇血,高血脂慢性肝炎疾病,胃肠疾病,肾病,心血管疾病,代谢疾病,癌症(例如,结直肠癌),或神经迹象如中风。In a specific embodiment, the FXR or TGR5 mediated disease or condition is cardiovascular disease, atherosclerosis, arteriosclerosis, hypercholesterolemia, hyperlipidemia, chronic hepatitis disease, gastrointestinal disease, kidney disease, cardiovascular disease, metabolism Disease, cancer (eg, colorectal cancer), or signs of nerves such as stroke.
在特定实施方案中,慢性肝病是原发性硬化,脑腱性黄瘤症,原发性硬化性胆囊 炎,药物导致的胆汁郁积,妊娠肝内胆汁淤积症,肠外吸收相关胆汁郁积,细菌过度生长或脓血症胆汁郁积,自身免疫肝炎,慢性病毒性肝炎,酒精性肝病,非酒精性脂肪肝疾病,非酒精性脂肪性肝炎,肝移植相关移植物抗宿主病,活供体肝移植再生,先天性肝纤维化,胆总管结石,肉芽性肝病,肝内-或外恶性肿瘤,Sjogren综合征,结节病,Wilson’s疾病,Gaucher’s疾病,血色病,或血压肾病,慢性肾小球炎,慢性移植性肾小球病,慢性间质性肾炎,或多囊肾病。In a specific embodiment, the chronic liver disease is primary sclerosis, cerebral palsy xanthomatosis, primary sclerosing gallbladder Inflammation, drug-induced cholestasis, intrahepatic cholestasis of pregnancy, parenteral absorption-related cholestasis, bacterial overgrowth or sepsis cholestasis, autoimmune hepatitis, chronic viral hepatitis, alcoholic liver disease, nonalcoholic fatty liver disease , nonalcoholic steatohepatitis, graft-versus-host disease associated with liver transplantation, regeneration of living donor liver transplantation, congenital liver fibrosis, common bile duct stones, granulomatous liver disease, intrahepatic- or extra-malignant tumor, Sjogren syndrome, Sarcoidosis, Wilson's disease, Gaucher's disease, hemochromatosis, or blood pressure nephropathy, chronic glomerulonephritis, chronic allograft glomerulopathy, chronic interstitial nephritis, or polycystic kidney disease.
在特定实施方案中,心血管疾病是动脉硬化症,动脉硬化,血脂障碍,高胆固醇血症,或高甘油三酯血症。In a particular embodiment, the cardiovascular disease is arteriosclerosis, arteriosclerosis, dyslipidemia, hypercholesterolemia, or hypertriglyceridemia.
在特定实施方案中,代谢疾病是胰岛素抗性,I型和II型糖尿病,或肥胖。In a particular embodiment, the metabolic disease is insulin resistance, type I and type II diabetes, or obesity.
与现有技术相比,本发明的有益效果为:本发明化合物可用于活化法尼醇X受体,间接抑制细胞色素7A1(CYP7A1)的基因表达,促进胆酸的合成。此外本发明化合物同时能够更好地调节G蛋白偶联受体5(TGR5)。通过氘化这一技术改变化合物在生物体中的代谢,使化合物具有更好的药代动力学参数特性。在这种情况下,可以改变剂量并形成长效制剂,改善适用性。用氘取代化合物中的氢原子,由于其氘同位素效应,能够提高化合物在动物体内的药物浓度,以提高药物疗效。用氘取代化合物中的氢原子,由于某些代谢产物被抑制,可能提高化合物的安全性。Compared with the prior art, the present invention has the beneficial effects that the compound of the present invention can be used to activate the farnesoid X receptor, indirectly inhibit the gene expression of cytochrome 7A1 (CYP7A1), and promote the synthesis of cholic acid. Furthermore, the compounds of the invention are capable of better modulating G protein coupled receptor 5 (TGR5). By deuteration this technique changes the metabolism of the compound in the organism, giving the compound a better pharmacokinetic parameter characteristic. In this case, the dosage can be changed and a long-acting preparation can be formed to improve the applicability. Replacing a hydrogen atom in a compound with hydrazine can increase the drug concentration of the compound in an animal to improve the efficacy of the drug due to its strontium isotope effect. Substitution of a hydrogen atom in a compound with hydrazine may increase the safety of the compound due to inhibition of certain metabolites.
具体实施方式detailed description
下面更具体地描述本发明式I结构化合物的制备方法,但这些具体方法不对本发明构成任何限制。本发明化合物还可以任选将在本说明书中描述的或本领域已知的各种合成方法组合起来而方便地制得,这样的组合可由本发明所属领域的技术人员容易地进行。The preparation of the structural compound of the formula I of the present invention is more specifically described below, but these specific methods do not constitute any limitation to the present invention. The compounds of the present invention may also be conveniently prepared by combining various synthetic methods described in the specification or known in the art, and such combinations are readily made by those skilled in the art to which the present invention pertains.
通常,在制备流程中,各反应通常在惰性溶剂中,在室温至回流温度(如0℃~100℃,优选0℃~80℃)下进行。反应时间通常为0.1小时-60小时,较佳地为0.5-24小时。Usually, in the preparation scheme, each reaction is usually carried out in an inert solvent at room temperature to reflux temperature (e.g., 0 ° C to 100 ° C, preferably 0 ° C to 80 ° C). The reaction time is usually from 0.1 to 60 hours, preferably from 0.5 to 24 hours.
实施例1制备3α,7α-二-羟基-7-d-6β-乙基-5β-胆烷酸(化合物7),具体合成步骤如Example 1 Preparation of 3α,7α-di-hydroxy-7-d-6β-ethyl-5β-cholanoic acid (Compound 7), the specific synthesis steps are as follows 下:under:
Figure PCTCN2016109851-appb-000007
Figure PCTCN2016109851-appb-000007
步骤1:α-羟基-7-酮基-5β-胆烷酸甲酯(化合物2)的合成。Step 1: Synthesis of methyl α-hydroxy-7-keto-5β-cholanoate (Compound 2).
将两滴浓硫酸滴加到3α-羟基-7-酮基-5β-胆烷酸(5.00g,12.80mmol)的甲醇(12mL)溶液中、回流反应(65℃)3小时。减压浓缩除去甲醇,浓缩液柱层析得到4.60g无色固体(化合物2),收率:88.3%。LC-MS(APCI):m/z=405.3[M+1]+Two drops of concentrated sulfuric acid were added dropwise to a solution of 3?-hydroxy-7-keto-5?-cholanoic acid (5.00 g, 12.80 mmol) in methanol (12 mL). The methanol was concentrated under reduced pressure, and the residue was applied tojjjjjjjjjjj LC-MS (APCI): m / z = 405.3 [M + 1] +.
步骤2:α-7α-二-三甲基甲硅烷氧基-5β-胆烷酸甲酯(化合物3)的合成。Step 2: Synthesis of methyl α-7α-di-trimethylsiloxy-5β-cholanoate (Compound 3).
-78℃下,将三甲基氯硅烷(7.15mL,57.0mmol)滴加到二异丙氨基锂(LDA 2M,34.2mL,68.4mmol)的无水四氢呋喃(THF,70mL)溶液中,搅拌30分钟。然后将3α-羟基-7-酮基-5β-胆烷酸甲酯(2.30g,5.7mmol)的无水四氢呋喃(30mL)混合物在15分钟内滴加到反应液中,反应液在-78℃下继续搅拌反应1.5小时。加入三乙胺(15mL,103mmol),反应液在-78℃下继续搅拌反应1.5小时。将反应升温至-20℃,加入饱和的碳酸氢钠溶液(20mL),缓慢升温至室温,搅拌2小时。有机层分离,水相用乙酸乙酯(100mL x 3)萃取。合并有机层用饱和碳酸氢钠溶液(100mL x 3),水(100mL),饱和食盐水(100mL)洗涤。无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析得到3.8g米白色固体,产率:100%。Trimethylchlorosilane (7.15 mL, 57.0 mmol) was added dropwise to a solution of lithium diisopropylamide (LDA 2M, 34.2 mL, 68.4 mmol) in anhydrous tetrahydrofuran (THF, 70 mL). minute. Then a mixture of methyl 3α-hydroxy-7-keto-5β-cholanoate (2.30 g, 5.7 mmol) in anhydrous tetrahydrofuran (30 mL) was added dropwise to the reaction mixture over 15 min. The reaction was further stirred for 1.5 hours. Triethylamine (15 mL, 103 mmol) was added, and the reaction mixture was stirred at -78 ° C for further 1.5 hours. The reaction was warmed to -20 ° C, saturated aqueous sodium bicarbonate (20 mL) was then evaporated and evaporated. The organic layer was separated and the aqueous extracted with ethyl acetate (100 mL The combined organic layers were washed with a saturated aqueous solution of sodium bicarbonate (100 mL), water (100mL) Dry over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure.
步骤3:3α-羟基-6-亚乙基-7-酮基-5β-胆烷酸甲酯(化合物4)的合成。Step 3: Synthesis of 3α-hydroxy-6-ethylidene-7-keto-5β-cholanoic acid methyl ester (Compound 4).
-60℃下,将三氟化硼乙醚(BF3·OEt2,14mL,110mmol)缓慢滴加到3α-7α-二-三甲基甲硅烷氧基-5β-胆烷酸甲酯(6.15g,11mmol)和乙醛(1.5mL,37.11mmol)的无水二氯甲烷(70mL)混合物中。反应液在-60℃下,搅拌反应2.5小时,升温至室温。用饱和碳酸氢钠溶液(50mL)淬灭反应。有机层分离,水相用二氯甲烷(50mL x 3)萃取。合并有机层用饱和食盐水(50mL)洗涤。无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到1.8g油状物,产率:33.3%。LC-MS(APCI):m/z=431.3[M+1]+Boron trifluoride etherate (BF 3 ·OEt 2 , 14 mL, 110 mmol) was slowly added dropwise to methyl αα-7α-di-trimethylsiloxy-5β-cholanoate (6.15 g) at -60 °C. , 11 mmol) and a mixture of acetaldehyde (1.5 mL, 37.11 mmol) in anhydrous dichloromethane (70 mL). The reaction solution was stirred at -60 ° C for 2.5 hours and warmed to room temperature. The reaction was quenched with saturated aqueous sodium bicarbonate (50 mL). The organic layer was separated and the aqueous extracted with dichloromethane (50 mL×3). The combined organic layers were washed with brine (50 mL). Dry over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure. LC-MS (APCI): m / z = 431.3 [M + 1] +.
步骤4:3α-羟基-6β-乙基-7-酮基-5β-胆烷酸甲酯(化合物5)的合成。Step 4: Synthesis of 3α-hydroxy-6β-ethyl-7-keto-5β-cholanoic acid methyl ester (Compound 5).
将Pd/C(10%,90mg)加入到3α-羟基-6-亚乙基-7-酮基-5β-胆烷酸甲酯(900mg,2.09mmol)的无水四氢呋喃和无水甲醇(30mL,1:1v/v)的混合溶剂中。氢气置换空气,氢气下搅拌反应过夜。硅藻土过滤,减压浓缩,浓缩液柱层析纯化得到630mg淡黄色油状物,产率:70.0%。LC-MS(APCI):m/z=433.3[M+1]+Add Pd/C (10%, 90 mg) to 3α-hydroxy-6-ethylidene-7-keto-5β-cholanoic acid methyl ester (900 mg, 2.09 mmol) in anhydrous tetrahydrofuran and anhydrous methanol (30 mL) , 1:1v/v) in a mixed solvent. The air was replaced with hydrogen, and the reaction was stirred overnight under hydrogen. The mixture was filtered through Celite, and evaporated. LC-MS (APCI): m / z = 433.3 [M + 1] +.
步骤5:3α-羟基-6β-乙基-7-酮基-5β-胆烷酸(化合物6)的合成。 Step 5: Synthesis of 3α-hydroxy-6β-ethyl-7-keto-5β-cholanoic acid (Compound 6).
将氢氧化钠(205mg,3.50mmol)加入到3α-羟基-6β-乙基-7-酮基-5β-胆烷酸甲酯(630mg,1.46mmol)的甲醇和水(20mL,1:1v/v)的混合溶剂中,反应搅拌过夜,减压除去甲醇,用水(20mL)稀释,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到430mg白色固体,产率:70.0%。LC-MS(APCI):m/z=417.3[M-1]-Add sodium hydroxide (205 mg, 3.50 mmol) to 3α-hydroxy-6β-ethyl-7-keto-5β-cholanoic acid methyl ester (630 mg, 1.46 mmol) in methanol and water (20 mL, 1:1 v/ In a mixed solvent of v), the reaction was stirred overnight, EtOAc was evaporated, evaporated, evaporated, evaporated, evaporated, evaporated, evaporated dry. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 417.3 [M-1] -.
步骤6:3α,7α-二-羟基-7-d-6β-乙基-5β-胆烷酸(化合物7)的合成。Step 6: Synthesis of 3α,7α-di-hydroxy-7-d-6β-ethyl-5β-cholanoic acid (Compound 7).
将硼氘化钠(195mg,5.00mmol)在冰浴下,分批加入到3α-羟基-6β-乙基-7-酮基-5β-胆烷酸(210mg,0.50mmol)的氘代甲醇-d(5mL)中,反应液在室温下搅拌反应过夜。将反应液倒入20mL的冰水中,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到130mg白色固体,产率:62.0%。LC-MS(APCI):m/z=420.3[M-1]-Sodium borohydride (195 mg, 5.00 mmol) was added portionwise to a deuterated methanol of 3α-hydroxy-6β-ethyl-7-keto-5β-cholanoic acid (210 mg, 0.50 mmol) in an ice bath. In d (5 mL), the reaction solution was stirred at room temperature overnight. The reaction mixture was poured into 20 mL of ice water, and then evaporated and evaporated. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 420.3 [M-1] -.
实施例2制备3α,7α-二-羟基-6β-(乙基-1-d)-5β-胆烷酸(化合物10),具体合成步Example 2 Preparation of 3α,7α-di-hydroxy-6β-(ethyl-1-d)-5β-cholanoic acid (Compound 10), specific synthesis steps 骤如下:The steps are as follows:
Figure PCTCN2016109851-appb-000009
Figure PCTCN2016109851-appb-000009
步骤1:3α-羟基-6β-(乙基-1-d)-6-d-7-酮基-5β-胆烷酸甲酯(化合物8)的合成。Step 1: Synthesis of 3α-hydroxy-6β-(ethyl-1-d)-6-d-7-keto-5β-cholanoic acid methyl ester (Compound 8).
将Pd/C(10%,55%in D2O,60mg)加入到3α-羟基-6-亚乙基-7-酮基-5β-胆烷酸甲酯(600mg,1.39mmol)的无水四氢呋喃和氘代甲醇(20mL,1:1v/v)的混合溶剂中。氘气置换空气,氘气下搅拌反应过夜。硅藻土过滤,减压浓缩,浓缩液柱层析纯化得到420mg淡黄色油状物,产率:70.0%。LC-MS(APCI):m/z=435.3[M+1]+Add Pd/C (10%, 55% in D 2 O, 60 mg) to anhydrous 3α-hydroxy-6-ethylidene-7-keto-5β-cholanoate (600 mg, 1.39 mmol) A mixed solvent of tetrahydrofuran and deuterated methanol (20 mL, 1:1 v/v). The helium was replaced with air and the reaction was stirred overnight under helium. The mixture was filtered through Celite, EtOAc evaporated. LC-MS (APCI): m / z = 435.3 [M + 1] +.
步骤2:3α-羟基-6β-(乙基-1-d)-7-酮基-5β-胆烷酸(化合物9)的合成。Step 2: Synthesis of 3α-hydroxy-6β-(ethyl-1-d)-7-keto-5β-cholanoic acid (Compound 9).
将氢氧化钠(120mg,2.90mmol)加入到3α-羟基-6β-(乙基-1-d)-6-d-7-酮基-5β-胆烷酸甲酯(420mg,0.97mmol)的甲醇和水(20mL,1:1v/v)的混合溶剂中,反应搅拌过夜,减压除去甲醇,用水(20mL)稀释,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到270mg白色固体,产率:66.4%。LC-MS(APCI):m/z=418.1[M-1]-Add sodium hydroxide (120 mg, 2.90 mmol) to 3α-hydroxy-6β-(ethyl-1-d)-6-d-7-keto-5β-cholanoic acid methyl ester (420 mg, 0.97 mmol) In a mixed solvent of methanol and water (20 mL, 1:1 v/v), the mixture was stirred and evaporated, evaporated, evaporated, evaporated, evaporated, evaporated, evaporated Wash with saturated brine and dry over anhydrous sodium sulfate. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 418.1 [M-1] -.
步骤3:3α,7α-二-羟基-6β-(乙基-1-d)-5β-胆烷酸(化合物10)的合成。Step 3: Synthesis of 3α,7α-di-hydroxy-6β-(ethyl-1-d)-5β-cholanoic acid (Compound 10).
将硼氢化钠(170mg,5.50mmol)在冰浴下,分批加入到3α-羟基-6β-(乙基-1-d)-7-酮基-5β-胆烷酸(230mg,0.55mmol)的无水甲醇(5mL)中,反应液在室温下搅拌反应过夜。将反应液倒入20mL的冰水中,用2N HCl酸化后,乙酸乙酯萃取(50mLx 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到70mg白色固体,产率:30.3%。LC-MS(APCI):m/z=420.3[M-1]-Sodium borohydride (170 mg, 5.50 mmol) was added portionwise to 3α-hydroxy-6β-(ethyl-1-d)-7-keto-5β-cholanoic acid (230 mg, 0.55 mmol). The reaction solution was stirred at room temperature overnight under anhydrous methanol (5 mL). The reaction solution was poured into 20 mL of ice water, and then evaporated, evaporated, evaporated The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 420.3 [M-1] -.
实施例3制备3α,7α-二-羟基-6β-乙基-6-d-5β-胆烷酸(化合物12),具体合成步骤Example 3 Preparation of 3α,7α-di-hydroxy-6β-ethyl-6-d-5β-cholanoic acid (Compound 12), specific synthesis steps 如下:as follows:
Figure PCTCN2016109851-appb-000010
Figure PCTCN2016109851-appb-000010
步骤1:3α-羟基--6β-乙基-6-d-7-酮基-5β-胆烷酸(化合物11)的合成。Step 1: Synthesis of 3α-hydroxy-6β-ethyl-6-d-7-keto-5β-cholanoic acid (Compound 11).
将氘氧化钠(120mg,2.90mmol)加入到3α-羟基-6β-乙基-7-酮基-5β-胆烷酸甲酯(420mg,0.97mmol)的氘代甲醇和重水(20mL,1:1v/v)的混合溶剂中,反应搅拌过夜,减压除去甲醇,用水(20mL)稀释,用2N HCl酸化后,乙酸乙酯萃取(50mL x3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到280mg白色固体,产率:68.8%。LC-MS(APCI):m/z=418.1[M-1]-Sodium bismuth oxide (120 mg, 2.90 mmol) was added to methyl 3α-hydroxy-6β-ethyl-7-keto-5β-cholanoate (420 mg, 0.97 mmol) in deuterated methanol and heavy water (20 mL, 1: In a mixed solvent of 1v/v), the reaction was stirred overnight, EtOAc was evaporated, evaporated, evaporated, evaporated, evaporated, evaporated. Sodium is dry. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 418.1 [M-1] -.
步骤3:3α,7α-二-羟基-6β-(乙基-1-d)-5β-胆烷酸(化合物12)的合成。Step 3: Synthesis of 3α,7α-di-hydroxy-6β-(ethyl-1-d)-5β-cholanoic acid (Compound 12).
将硼氢化钠(260mg,6.60mmol)在冰浴下,分批加入到3α-羟基-6β-(乙基-1-d)-7-酮基-5β-胆烷酸(280mg,0.66mmol)的无水甲醇(5mL)中,反应液在室温下搅拌反应过夜。将反应液倒入20mL的冰水中,用2N HCl酸化后,乙酸乙酯萃取(50mL x3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到70mg白色固体,产率:25.2%。LC-MS(APCI):m/z=420.3[M-1]-Sodium borohydride (260 mg, 6.60 mmol) was added portionwise to 3α-hydroxy-6β-(ethyl-1-d)-7-keto-5β-cholanoic acid (280 mg, 0.66 mmol). The reaction solution was stirred at room temperature overnight under anhydrous methanol (5 mL). The reaction mixture was poured into 20 mL of ice water, and then evaporated, evaporated, evaporated The organic layer was concentrated under reduced pressure. LC-MS (APCI): m / z = 420.3 [M-1] -.
实施例4制备3α,7α-二-羟基-6β-(乙基-1-d)-6-d-5β-胆烷酸(化合物14),具体合Example 4 Preparation of 3α,7α-di-hydroxy-6β-(ethyl-1-d)-6-d-5β-cholanoic acid (Compound 14), specifically 成步骤如下:The steps are as follows:
Figure PCTCN2016109851-appb-000011
Figure PCTCN2016109851-appb-000011
步骤1:3α-羟基-6β-(乙基-1-d)-6-d-7-酮基-5β-胆烷酸(化合物13)的合成。Step 1: Synthesis of 3α-hydroxy-6β-(ethyl-1-d)-6-d-7-keto-5β-cholanoic acid (Compound 13).
将氘氧化钠(150mg,3.52mmol)加入到3α-羟基-6β-(乙基-1-d)-6-d-7-酮基-5β-胆烷酸甲酯(510mg,1.17mmol)的氘代甲醇和重水(10mL,1:1v/v)的混合溶剂中,反应搅拌过夜,减压除去甲醇,用水(20mL)稀释,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到490mg白色固体,产率:95.0%。LC-MS(APCI):m/z=419.1[M-1]-Sodium bismuth oxide (150 mg, 3.52 mmol) was added to methyl 3?-hydroxy-6?-(ethyl-1-d)-6-d-7-keto-5?-cholanoate (510 mg, 1.17 mmol) The reaction mixture was stirred with EtOAc (EtOAc) (EtOAc (EtOAc) The organic layer was washed with brine and dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 419.1 [M-1] -.
步骤2:3α,7α-二-羟基-6β-(乙基-1-d)-6-d-5β-胆烷酸(化合物14)的合成。Step 2: Synthesis of 3α,7α-di-hydroxy-6β-(ethyl-1-d)-6-d-5β-cholanoic acid (Compound 14).
将硼氢化钠(225mg,6.00mmol)在冰浴下,分批加入到3α-羟基-6β-(乙基-1-d)-6-d-7-酮基-5β-胆烷酸(250mg,0.60mmol)的无水甲醇(5mL)中,反应液在室温下搅拌反应过夜。将反应液倒入20mL的冰水中,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到53mg白色固体,产率:21.0%。LC-MS(APCI):m/z=421.4[M-1]-Sodium borohydride (225 mg, 6.00 mmol) was added in portions to 3α-hydroxy-6β-(ethyl-1-d)-6-d-7-keto-5β-cholanoic acid (250 mg). The reaction solution was stirred at room temperature overnight under anhydrous methanol (5 mL). The reaction mixture was poured into 20 mL of ice water, and then evaporated and evaporated. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 421.4 [M-1] -.
实施例5制备3α,7α-二-羟基-7-d-6β-(乙基-1-d)-6-d-5β-胆烷酸(化合物15),具Example 5 Preparation of 3α,7α-di-hydroxy-7-d-6β-(ethyl-1-d)-6-d-5β-cholanoic acid (Compound 15), 体合成步骤如下:The body synthesis steps are as follows:
Figure PCTCN2016109851-appb-000012
Figure PCTCN2016109851-appb-000012
将硼氘化钠(225mg,6.00mmol)在冰浴下,分批加入到3α-羟基-6β-(乙基-1-d)-6-d-7-酮基-5β-胆烷酸(250mg,0.60mmol)的氘代甲醇(5mL)中,反应液在室温下搅拌反应过夜。将反应液倒入20mL的冰水中,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到31mg白色固体,产率:12.3%。LC-MS(APCI):m/z=422.4[M-1]-Sodium borohydride (225 mg, 6.00 mmol) was added portionwise to 3α-hydroxy-6β-(ethyl-1-d)-6-d-7-keto-5β-cholanoic acid in an ice bath ( 250 mg, 0.60 mmol) of deuterated methanol (5 mL), the reaction mixture was stirred at room temperature overnight. The reaction mixture was poured into 20 mL of ice water, and then evaporated and evaporated. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 422.4 [M-1] -.
实施例6制备3α,7α-二-羟基-6β-(乙基-1,2,2,2-d4)-5β-胆烷酸(化合物19),具体Example 6 Preparation of 3α,7α-di-hydroxy-6β-(ethyl-1,2,2,2-d 4 )-5β-cholanoic acid (Compound 19), specific 实施步骤如下:The implementation steps are as follows:
Figure PCTCN2016109851-appb-000013
Figure PCTCN2016109851-appb-000013
步骤1:3α-羟基-6-(亚乙基-1,2,2,2-d4)-7-酮基-5β-胆烷酸甲酯(化合物16)的合成。Step 1: Synthesis of 3α-hydroxy-6-(ethylidene-1,2,2,2-d 4 )-7-keto-5β-cholanoic acid methyl ester (Compound 16).
-60℃下,将三氟化硼乙醚(BF3·OEt2,7.20mL,11.4mmol)缓慢滴加到3α-7α-二-三甲基甲硅烷氧基-5β-胆烷酸甲酯(3.10g,5.70mmol)和氘代乙醛-d4(1.00mL)的无水二氯甲烷(58mL)混合物中。反应液在-60℃下,搅拌反应2.5小时,升温至室温。用饱和碳酸氢钠溶液(50mL)淬灭反应。有机层分离,水相用二氯甲烷(50mL x3)萃取。合并有机层用饱和食盐水(50mL)洗涤。无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到1.3g油状物,产率:52.5%。LC-MS(APCI):m/z=435.5[M+1]+Boron trifluoride etherate (BF 3 ·OEt 2 , 7.20 mL, 11.4 mmol) was slowly added dropwise to methyl 3α-7α-di-trimethylsiloxy-5β-cholanoate at -60 °C. 3.10g, 5.70mmol) and deuterated acetaldehyde -d 4 (1.00mL) in dry dichloromethane (58 mL) mixture. The reaction solution was stirred at -60 ° C for 2.5 hours and warmed to room temperature. The reaction was quenched with saturated aqueous sodium bicarbonate (50 mL). The organic layer was separated and the aqueous extracted with dichloromethane (50 mL×3). The combined organic layers were washed with brine (50 mL). Dry over anhydrous sodium sulfate. The organic layer was concentrated under reduced pressure. LC-MS (APCI): m / z = 435.5 [M + 1] +.
步骤2:3α-羟基-6β-(乙基-1,2,2,2-d4)-7-酮基-5β-胆烷酸甲酯(化合物17)的合成。Step 2: Synthesis of 3α-hydroxy-6β-(ethyl-1,2,2,2-d 4 )-7-keto-5β-cholanoic acid methyl ester (Compound 17).
将Pd/C(10%,118mg)加入到3α-羟基-6-(亚乙基-1,2,2,2-d4)-7-酮基-5β-胆烷酸甲 酯(1.18g,2.72mmol)的无水四氢呋喃和无水甲醇(30mL,1:1v/v)的混合溶剂中。氢气置换空气,氢气下搅拌反应过夜。硅藻土过滤,减压浓缩,浓缩液柱层析纯化得到900mg淡黄色油状物,产率:75.8%。LC-MS(APCI):m/z=437.4[M+1]+Add Pd/C (10%, 118 mg) to 3α-hydroxy-6-(ethylidene-1,2,2,2-d 4 )-7-keto-5β-cholanoic acid methyl ester (1.18 g) , 2.72 mmol) in a mixed solvent of anhydrous tetrahydrofuran and anhydrous methanol (30 mL, 1:1 v/v). The air was replaced with hydrogen, and the reaction was stirred overnight under hydrogen. The mixture was filtered through Celite, and evaporated. LC-MS (APCI): m / z = 437.4 [M + 1] +.
步骤3:3α-羟基-6β-(乙基-1,2,2,2-d4)-7-酮基-5β-胆烷酸(化合物18)的合成。Step 3: Synthesis of 3α-hydroxy-6β-(ethyl-1,2,2,2-d 4 )-7-keto-5β-cholanoic acid (Compound 18).
将氢氧化钠(192mg,4.80mmol)加入到3α-羟基-6β-(乙基-1,2,2,2-d4)-7-酮基-5β-胆烷酸甲酯(600mg,1.37mmol)的甲醇和水(20mL,1:1v/v)的混合溶剂中,反应搅拌过夜,减压除去甲醇,用水(20mL)稀释,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到360mg白色固体,产率:62.2%。LC-MS(APCI):m/z=421.3[M-1]-Add sodium hydroxide (192 mg, 4.80 mmol) to 3α-hydroxy-6β-(ethyl-1,2,2,2-d 4 )-7-keto-5β-cholanoic acid methyl ester (600 mg, 1.37) Methyl acetate and water (20 mL, 1:1 v/v) were stirred in EtOAc EtOAc (EtOAc) The organic layer was washed with brine and dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 421.3 [M-1] -.
步骤4:3α,7α-二羟基-6β-(乙基-1,2,2,2-d4)-乙基-5β-胆烷酸(化合物19)的合成。Step 4: Synthesis of 3α,7α-dihydroxy-6β-(ethyl-1,2,2,2-d 4 )-ethyl-5β-cholanoic acid (Compound 19).
将硼氢化钠(160mg,4.10mmol)在冰浴下,分批加入到3α-羟基-6β-(乙基-1,2,2,2-d4)-7-酮基-5β-胆烷酸(175mg,0.41mmol)的无水甲醇(5mL)中,反应液在室温下搅拌反应过夜。将反应液倒入20mL的冰水中,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到110mg白色固体,产率:60.5%。LC-MS(APCI):m/z=423.3[M-1]-Sodium borohydride (160 mg, 4.10 mmol) was added portionwise to 3α-hydroxy-6β-(ethyl-1,2,2,2-d 4 )-7-keto-5β-cholane in an ice bath The reaction mixture was stirred at room temperature overnight with EtOAc (EtOAc,EtOAc. The reaction mixture was poured into 20 mL of ice water, and then evaporated and evaporated. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 423.3 [M-1] -.
实施例7制备3α,7α-二-羟基-7-d--6β-(乙基-1,2,2,2-d4)-5β-胆烷酸(化合物20),Example 7 Preparation of 3α,7α-di-hydroxy-7-d--6β-(ethyl-1,2,2,2-d 4 )-5β-cholanoic acid (Compound 20), 具体合成步骤如下:The specific synthesis steps are as follows:
Figure PCTCN2016109851-appb-000014
Figure PCTCN2016109851-appb-000014
将硼氘化钠(174mg,4.10mmol)在冰浴下,分批加入到3α-羟基-6β-(乙基-1,2,2,2-d4)-7-酮基-5β-胆烷酸(175mg,0.41mmol)的氘代甲醇(5mL)中,反应液在室温下搅拌反应过夜。将反应液倒入20mL的冰水中,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到85mg白色固体,产率:48.5%。LC-MS(APCI):m/z=424.3[M-1]-Sodium borohydride (174 mg, 4.10 mmol) was added in portions to 3α-hydroxy-6β-(ethyl-1,2,2,2-d 4 )-7-keto-5β-biliary in an ice bath The alkanoic acid (175 mg, 0.41 mmol) in EtOAc (5 mL) was evaporated. The reaction mixture was poured into 20 mL of ice water, and then evaporated and evaporated. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 424.3 [M-1] -.
实施例8制备3α,7α-二-羟基-6β-(乙基-1,2,2,2-d4)-6-d-5β-胆烷酸(化合物22),Example 8 Preparation of 3α,7α-di-hydroxy-6β-(ethyl-1,2,2,2-d 4 )-6-d-5β-cholanoic acid (Compound 22), 具体合成步骤如下:The specific synthesis steps are as follows:
Figure PCTCN2016109851-appb-000015
Figure PCTCN2016109851-appb-000015
步骤1:3α-羟基-6β-(乙基-1,2,2,2-d4)-6-d-7-酮基-5β-胆烷酸(化合物21)的合成。Step 1: Synthesis of 3α-hydroxy-6β-(ethyl-1,2,2,2-d 4 )-6-d-7-keto-5β-cholanoic acid (Compound 21).
将氢氧化钠(97mg,2.36mmol)加入到3α-羟基-6β-(乙基-1,2,2,2-d4)-7-酮基-5β-胆烷酸甲酯(300mg,0.68mmol)的氘代甲醇和重水(10mL,1:1v/v)的混合溶剂中,反应搅拌过夜,减压除去甲醇,用水(20mL)稀释,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到150mg白色固体,产率:51.8%。LC-MS(APCI):m/z=422.3[M-1]-Add sodium hydroxide (97 mg, 2.36 mmol) to 3α-hydroxy-6β-(ethyl-1,2,2,2-d 4 )-7-keto-5β-cholanoic acid methyl ester (300 mg, 0.68) Methyl acetate in a mixed solvent of deuterated methanol and heavy water (10 mL, 1:1 v/v). The mixture was stirred overnight. The solvent was evaporated in vacuo, diluted with water (20 mL), EtOAc. 3) The organic layer was washed with brine and dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 422.3 [M-1] -.
步骤2:3α,7α-二-羟基-6β-(乙基-1,2,2,2-d4)-6-d-5β-胆烷酸(化合物22)的合成。Step 2: Synthesis of 3α,7α-di-hydroxy-6β-(ethyl-1,2,2,2-d 4 )-6-d-5β-cholanoic acid (Compound 22).
将硼氢化钠(134mg,3.50mmol)在冰浴下,分批加入到3α-羟基-6β-(乙基-1,2,2,2-d4)-6-d-7-酮基-5β-胆烷酸(150mg,0.35mmol)的无水甲醇(5mL)中,反应液在室温下搅拌反应过夜。将反应液倒入20mL的冰水中,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到25mg灰色固体,产率:16.8%。LC-MS(APCI):m/z=424.3[M-1]-Sodium borohydride (134 mg, 3.50 mmol) was added portionwise to the 3α-hydroxy-6β-(ethyl-1,2,2,2-d 4 )-6-d-7-keto group in an ice bath. 5β-cholanoic acid (150 mg, 0.35 mmol) in anhydrous methanol (5 mL). The reaction mixture was poured into 20 mL of ice water, and then evaporated and evaporated. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 424.3 [M-1] -.
实施例9制备3α,7α-二-羟基-7-d-6β-(乙基-1,1,2,2,2-d5)-6-d-5β-胆烷酸(化合物25),Example 9 Preparation of 3α,7α-di-hydroxy-7-d-6β-(ethyl-1,1,2,2,2-d 5 )-6-d-5β-cholanoic acid (Compound 25), 具体合成步骤如下:The specific synthesis steps are as follows:
Figure PCTCN2016109851-appb-000016
Figure PCTCN2016109851-appb-000016
步骤1:3α-羟基-6β-(乙基-1,1,2,2,2-d5)-6-d-7-酮基-5β-胆烷酸甲酯(化合物23)的合成。Step 1: Synthesis of 3α-hydroxy-6β-(ethyl-1,1,2,2,2-d 5 )-6-d-7-keto-5β-cholanoic acid methyl ester (Compound 23).
将Pd/C(10%,50%in D2O,110mg)加入到3α-羟基-6-(亚乙基-1,2,2,2-d4)-7-酮基-5β-胆烷酸甲酯(1.10g,2.53mmol)的无水四氢呋喃和氘代甲醇(30mL,1:1v/v)的混合溶剂中。氘气置换空气,氘气下搅拌反应过夜。硅藻土过滤,减压浓缩,浓缩液柱层析纯化得到840mg无色油状物,产率:76.0%。LC-MS(APCI):m/z=439.4[M+1]+Add Pd/C (10%, 50% in D 2 O, 110 mg) to 3α-hydroxy-6-(ethylidene-1,2,2,2-d 4 )-7-keto-5β-biliary A mixture of methyl alkanoate (1.10 g, 2.53 mmol) in anhydrous tetrahydrofuran and deuterated methanol (30 mL, 1:1 v/v). The helium was replaced with air and the reaction was stirred overnight under helium. The mixture was filtered through Celite, EtOAc evaporated. LC-MS (APCI): m / z = 439.4 [M + 1] +.
步骤2:3α-羟基-6β-(乙基-1,1,2,2,2-d5)-6-d-7-酮基-5β-胆烷酸(化合物24)的合成。Step 2: Synthesis of 3α-hydroxy-6β-(ethyl-1,1,2,2,2-d 5 )-6-d-7-keto-5β-cholanoic acid (Compound 24).
将氘氧化钠(96mg,2.40mmol)加入到3α-羟基-6β-(乙基-1,1,2,2,2-d5)-6-d-7-酮基-5β-胆烷酸甲酯(300mg,0.68mmol)的氘代甲醇和重水(10mL,1:1v/v)的混合溶剂中,反应搅拌过夜,减压除去甲醇,用水(20mL)稀释,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到260mg白色固体,产率:90.1%。LC-MS(APCI):m/z=423.3[M-1]-Add sodium niobate (96 mg, 2.40 mmol) to 3α-hydroxy-6β-(ethyl-1,1,2,2,2-d 5 )-6-d-7-keto-5β-cholanoic acid Methyl ester (300 mg, 0.68 mmol) in a mixed solvent of deuterated methanol and heavy water (10 mL, 1:1 v/v), and the mixture was stirred overnight. The methanol was evaporated under reduced pressure, diluted with water (20mL), acidified with 2N HCl, acetic acid The organic layer was washed with brine and dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 423.3 [M-1] -.
步骤3:3α,7α-二-羟基-7-d-6β-(乙基-1,1,2,2,2-d5)-6-d-5β-胆烷酸(化合物25)的合成。 Step 3: Synthesis of 3α,7α-di-hydroxy-7-d-6β-(ethyl-1,1,2,2,2-d 5 )-6-d-5β-cholanoic acid (Compound 25) .
将硼氘化钠(134mg,3.50mmol)在冰浴下,分批加入到3α-羟基-6β-(乙基-1,1,2,2,2-d5)-6-d-7-酮基-5β-胆烷酸(150mg,0.35mmol)的氘代甲醇(5mL)中,反应液在室温下搅拌反应过夜。将反应液倒入20mL的冰水中,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到70mg白色固体,产率:46.8%。LC-MS(APCI):m/z=426.3[M-1]-Sodium borohydride (134 mg, 3.50 mmol) was added in portions to 3α-hydroxy-6β-(ethyl-1,1,2,2,2-d 5 )-6-d-7- in an ice bath. The keto-5β-cholanoic acid (150 mg, 0.35 mmol) in deuterated methanol (5 mL) was stirred and stirred at room temperature overnight. The reaction mixture was poured into 20 mL of ice water, and then evaporated and evaporated. The organic layer was concentrated under reduced pressure. LC-MS (APCI): m / z = 426.3 [M-1] -.
实施例10制备3α,7α-二-羟基-6β-(乙基-1,1,2,2,2-d5)-5β-胆烷酸(化合物27),具Example 10 Preparation of 3α,7α-di-hydroxy-6β-(ethyl-1,1,2,2,2-d 5 )-5β-cholanoic acid (Compound 27), 体合成步骤如下:The body synthesis steps are as follows:
Figure PCTCN2016109851-appb-000017
Figure PCTCN2016109851-appb-000017
步骤1:3α-羟基-6β-(乙基-1,1,2,2,2-d5)-7-酮基-5β-胆烷酸(化合物26)的合成。Step 1: Synthesis of 3α-hydroxy-6β-(ethyl-1,1,2,2,2-d 5 )-7-keto-5β-cholanoic acid (Compound 26).
将氢氧化钠(85mg,2.04mmol)加入到3α-羟基-6β-(乙基-1,1,2,2,2-d5)-6-d-7-酮基-5β-胆烷酸甲酯(255mg,0.58mmol)的氘代甲醇和重水(10mL,1:1v/v)的混合溶剂中,反应搅拌过夜,减压除去甲醇,用水(20mL)稀释,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到220mg白色固体,产率:86.2%。LC-MS(APCI):m/z=422.3[M-1]-Add sodium hydroxide (85 mg, 2.04 mmol) to 3α-hydroxy-6β-(ethyl-1,1,2,2,2-d 5 )-6-d-7-keto-5β-cholanoic acid Methyl ester (255 mg, 0.58 mmol) in a mixed solvent of deuterated methanol and heavy water (10 mL, 1:1 v/v), and the mixture was stirred overnight. The methanol was evaporated under reduced pressure, diluted with water (20mL), acidified with 2N HCl, acetic acid The organic layer was washed with brine and dried over anhydrous sodium sulfate. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 422.3 [M-1] -.
步骤2:3α,7α-二-羟基-6β-(乙基-1,1,2,2,2-d5)-5β-胆烷酸(化合物27)的合成。Step 2: Synthesis of 3α,7α-di-hydroxy-6β-(ethyl-1,1,2,2,2-d 5 )-5β-cholanoic acid (Compound 27).
将硼氢化钠(200mg,5.20mmol)在冰浴下,分批加入到3α-羟基-6β-(乙基-1,1,2,2,2-d5)-7-酮基-5β-胆烷酸(220mg,0.52mmol)的无水甲醇(5mL)中,反应液在室温下搅拌反应过夜。将反应液倒入20mL的冰水中,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到120mg白色固体,产率:53.5%。LC-MS(APCI):m/z=424.3[M-1]-Sodium borohydride (200 mg, 5.20 mmol) was added in portions to 3α-hydroxy-6β-(ethyl-1,1,2,2,2-d 5 )-7-keto-5β- in an ice bath. The reaction solution was stirred at room temperature overnight under EtOAc (EtOAc, EtOAc) The reaction mixture was poured into 20 mL of ice water, and then evaporated and evaporated. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 424.3 [M-1] -.
实施例11制备3α,7α-二-羟基-7-d-6β-(乙基-1,1,2,2,2-d5)-5β-胆烷酸(化合物28)具体合成步骤如下: Example 11 Preparation of 3α,7α-di-hydroxy-7-d-6β-(ethyl-1,1,2,2,2-d 5 )-5β-cholanoic acid (Compound 28) , the specific synthetic steps are as follows :
Figure PCTCN2016109851-appb-000018
Figure PCTCN2016109851-appb-000018
将硼氘化钠(80mg,1.90mmol)在冰浴下,分批加入到3α-羟基-6β-(乙基-1,1,2,2,2-d5)-7-酮基-5β-胆烷酸(80mg,0.19mmol)的氘代甲醇(5mL)中,反应液在 室温下搅拌反应过夜。将反应液倒入20mL的冰水中,用2N HCl酸化后,乙酸乙酯萃取(50mL x 3),有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩有机层,浓缩液柱层析纯化得到40mg淡黄色固体,产率:49.5%。LC-MS(APCI):m/z=425.4[M-1]-Sodium borohydride (80 mg, 1.90 mmol) was added in portions to 3α-hydroxy-6β-(ethyl-1,1,2,2,2-d 5 )-7-keto-5β in an ice bath - Cyanic acid (80 mg, 0.19 mmol) in deuterated methanol (5 mL), and the reaction mixture was stirred at room temperature overnight. The reaction mixture was poured into 20 mL of ice water, and then evaporated and evaporated. The organic layer was concentrated under reduced vacuo. LC-MS (APCI): m / z = 425.4 [M-1] -.
生物活性测试。Biological activity test.
法尼酯X受体(FXR)以及G蛋白偶联受体5(TGR5)的体外活性测试。In vitro activity assay of farnesoid X receptor (FXR) and G protein coupled receptor 5 (TGR5).
法尼酯X受体(FXR)的体外活性测试。In vitro activity assay of farnesoid X receptor (FXR).
通过荧光共振能量转移(FRET)检测对在FXR上的活性进行评价,其中所述的FRET用以检测所述的SRC-1肽在人类FXR中的募集,其中使用到一种不含有细胞的酶联免疫吸附检测(ELISA)。Activity on FXR was assessed by fluorescence resonance energy transfer (FRET) assay to detect recruitment of the SRC-1 peptide in human FXR using an enzyme that does not contain cells Combined immunosorbent assay (ELISA).
G蛋白偶联受体5(TGR5)的体外活性测试。In vitro activity assay of G protein coupled receptor 5 (TGR5).
在中国仓鼠卵巢(CHO)细胞中对荧光素酶的活性进行测定,其中所述的CHO细胞稳定的表达人类G蛋白偶联受体5(hTGR5),或者所述的细胞被利用一个hTGR5表达载体以及一个由cAMP应答性元件(CRE)驱动的荧光素酶受体基因进行了瞬间的共转染。具体方法如下。Luciferase activity is measured in Chinese hamster ovary (CHO) cells, wherein the CHO cells stably express human G protein-coupled receptor 5 (hTGR5), or the cells are utilized an hTGR5 expression vector And a luciferase receptor gene driven by the cAMP responsive element (CRE) was transiently co-transfected. The specific method is as follows.
质粒:从Invitrogen(Carlsbad,CA)处获得了所述的美国国立卫生研究院(NIH)哺乳动物基因保藏克隆MGC:40597(也被称之为pCMVSPORT6/hTGR5或者pTGR5)以及pcDNA 3.1(+)。pCRE-Luc(环化腺核苷一磷酸应答性元件驱动的荧光素酶受体质粒)以及pCMVβ是从Clontech(Palo Alto,CA)处获得的。Plasmid: The National Institutes of Health (NIH) mammalian gene-preserving clone MGC: 40597 (also known as pCMVSPORT6/hTGR5 or pTGR5) and pcDNA 3.1 (+) were obtained from Invitrogen (Carlsbad, CA). pCRE-Luc (cyclized adenosine monophosphate responsive element driven luciferase receptor plasmid) and pCMVβ were obtained from Clontech (Palo Alto, CA).
瞬间转染:将中国仓鼠卵巢(CHO)细胞以3.5x 104个细胞/孔的密度放置于96孔培养板内,培养24小时,并且在此之后在每一个孔内利用150纳克的人类(h)G蛋白偶联受体5表达质粒(pCMVSPORT6/hTGR5)以及100纳克的环化腺核苷一磷酸(cAMP)应答性元件(CRE)驱动的荧光素酶报告质粒(pCRE-Luc)进行转染,其中在所述的转染中依照制造商的说明书使用Lipofectamine 2000试剂(Invitrogen)。经过6小时的培养之后,利用磷酸盐缓冲的生理盐水(PBS)对细胞进行一次洗涤并且将培养基调换为含有0.1%(重量/体积)的牛血清白蛋白(BSA)的DMEM。又经过了18小时的培养之后,利用每一种化合物的不同浓度对细胞进行5小时的处理,其中所述的化合物存在于含有0.1%(重量/体积)的牛血清白蛋白(BSA)的新鲜的DMEM之中。经过处理之后,通过一个冷冻-解冻循环利用50微升的裂解缓冲液(25毫摩的Tris盐酸(pH7.6),2毫摩的乙二胺四乙酸(EDTA),1毫摩的二硫苏糖醇(DTT),10%(体积/体积)的丙三醇以及1%(体积/体积)的Triton X-100)对所述的细胞进行裂解并且按照下文中所描述的方法使所述的细胞进行荧光素酶的检测。 Transient transfection: Chinese hamster ovary (CHO) cells were placed in 96-well culture plates at a density of 3.5 x 10 4 cells/well for 24 hours, and after that, 150 ng of humans were utilized in each well. (h) G protein coupled receptor 5 expression plasmid (pCMVSPORT6/hTGR5) and 100 ng of circularized adenosine monophosphate (cAMP) responsive element (CRE) driven luciferase reporter plasmid (pCRE-Luc) Transfection was performed in which Lipofectamine 2000 reagent (Invitrogen) was used in the transfection as described in accordance with the manufacturer's instructions. After 6 hours of incubation, the cells were washed once with phosphate buffered saline (PBS) and the medium was exchanged for DMEM containing 0.1% (w/v) bovine serum albumin (BSA). After another 18 hours of incubation, the cells were treated with different concentrations of each compound for 5 hours, wherein the compound was present in fresh 0.1% (w/v) bovine serum albumin (BSA). Among the DMEM. After treatment, 50 μl of lysis buffer (25 mmol of Tris hydrochloric acid (pH 7.6), 2 mmol of ethylenediaminetetraacetic acid (EDTA), 1 mmol of disulfide was used through a freeze-thaw cycle. Threitol (DTT), 10% (v/v) glycerol and 1% (v/v) Triton X-100) lyse the cells and render the method as described below The cells were tested for luciferase.
荧光素酶检测以及β-半乳糖苷酶检测:为了进行荧光素酶检测,将20微升的细胞裂解液与100微升的荧光素酶反应缓冲液235微摩的虫荧光素,265微摩的三磷酸腺苷(ATP)以及135微摩的辅酶A(CoA)进行混合并且利用CentroXS3LB960(Berthold Technologies,Bad Wildbad,德国)对发光性进行测定。为了进行β-半乳糖苷酶检测,将10微升的细胞裂解液与100微升的缓冲液Z[60毫摩的磷酸氢二钠,10毫摩的氯化钾,1毫摩的硫酸镁,50毫摩的β-巯基乙醇以及0.75毫克/毫升的邻硝基苯基-β-D-吡喃半乳糖苷(ONPG)]进行混合并且在37℃下进行0.5至3小时的培养。通过添加50微升的终止缓冲液(1M的碳酸钠)对反应进行终止并且在420纳米下对所述的光学密度进行测定。Luciferase assay and beta-galactosidase assay: For luciferase assay, 20 μl of cell lysate with 100 μl of luciferase buffer 235 μM of luciferin, 265 μM Adenosine triphosphate (ATP) and 135 micromolar coenzyme A (CoA) were mixed and luminescence was measured using CentroXS3LB960 (Berthold Technologies, Bad Wildbad, Germany). For beta-galactosidase assay, 10 μl of cell lysate with 100 μl of buffer Z [60 mmol of disodium hydrogen phosphate, 10 mmol of potassium chloride, 1 mmol of magnesium sulfate) 50 μM of β-mercaptoethanol and 0.75 mg/ml of o-nitrophenyl-β-D-galactopyranoside (ONPG) were mixed and cultured at 37 ° C for 0.5 to 3 hours. The reaction was terminated by the addition of 50 microliters of stop buffer (1 M sodium carbonate) and the optical density was determined at 420 nm.
建立能够稳定的表达人类G蛋白偶联受体5的中国仓鼠卵巢(CHO)细胞(CHO-TGR5cells):使用Lipofectamin 2000,利用3.8微克的hTGR5表达质粒(pCMVSPORT6/hTGR5),3.8微克的cAMP应答性元件(CRE)驱动的荧光素酶报告质粒(pCRE-Luc)以及0.4微克的新霉素抵抗的基因表达质粒[pcDNA3.1(+)]对中国仓鼠卵巢(CHO)细胞进行转染。利用400微克/毫升的G418硫酸盐对所述的转染体进行筛选并且使单一的克隆物独立的生长在96孔培养板内。通过生命周期评价(LCA)的处理方式对表达G蛋白偶联受体5(TGR5)的中国仓鼠卵巢(CHO)细胞系进行筛选,在此之后进行荧光素酶的检测。Establishment of Chinese hamster ovary (CHO) cells (CHO-TGR5 cells) capable of stably expressing human G protein-coupled receptor 5: using Lipofectamin 2000, using 3.8 μg of hTGR5 expression plasmid (pCMVSPORT6/hTGR5), 3.8 μg of cAMP responsiveness A Chinese hamster ovary (CHO) cell was transfected with a CRE-driven luciferase reporter plasmid (pCRE-Luc) and 0.4 μg of a neomycin-resistant gene expression plasmid [pcDNA3.1(+)]. The transfectants were screened with 400 μg/ml of G418 sulfate and individual clones were grown independently in 96-well culture plates. The Chinese hamster ovary (CHO) cell line expressing G protein coupled receptor 5 (TGR5) was screened by life cycle assessment (LCA) treatment, after which luciferase assay was performed.
50%有效浓度(EC50)以及效率的测定:在每一种条件下以一式三份或者一式四份的方式进行检测。通过概率单位分析来确定EC50值。通过下述方式对效率进行测定:对于TGR5激动剂的研究而言,通过计算10微摩的石胆酸(LCA)的数值的百分含量的方式。在进行了运算法则的转换之后,完成了对平均的EC50的计算以及对各种不同的化合物所具有的EC50所进行的比较。50% effective concentration (EC 50) and the measurement efficiency: detecting in triplicate or quadruplicate manner under each condition. The EC 50 value is determined by probabilistic unit analysis. The efficiency was determined by the following method: For the study of TGR5 agonist, by calculating the percentage of the value of 10 micromoles of lithocholic acid (LCA). After performing the conversion algorithm, the completion of the comparison of the calculated average EC 50 and EC for various compounds 50 has performed.
实验结果如下表1所示,说明本发明化合物同INT-747相比,能够更好地活化法尼酯X受体(FXR)和G蛋白偶联受体5(TGR5)。The experimental results are shown in Table 1 below, indicating that the compound of the present invention can better activate farnesyl ester X receptor (FXR) and G protein coupled receptor 5 (TGR5) as compared with INT-747.
表1实施例化合物对FXR受体和TGR5受体的作用Table 1 Effect of Example Compounds on FXR Receptor and TGR5 Receptor
Figure PCTCN2016109851-appb-000019
Figure PCTCN2016109851-appb-000019
Figure PCTCN2016109851-appb-000020
Figure PCTCN2016109851-appb-000020
代谢稳定性评价。Metabolic stability evaluation.
微粒体实验:人肝微粒体:0.5mg/mL,Xenotech;大鼠肝微粒体:0.5mg/mL,Xenotech;辅酶(NADPH/NADH):1mM,Sigma Life Science;氯化镁:5mM,100mM磷酸盐缓冲剂(pH为7.4)。Microsomal experiments: human liver microsomes: 0.5 mg/mL, Xenotech; rat liver microsomes: 0.5 mg/mL, Xenotech; coenzyme (NADPH/NADH): 1 mM, Sigma Life Science; magnesium chloride: 5 mM, 100 mM phosphate buffer Agent (pH 7.4).
储备液的配制:精密称取一定量的实施例化合物,并用DMSO分别溶解至5mM。Preparation of the stock solution: A certain amount of the compound of the example was accurately weighed and dissolved to 5 mM with DMSO.
磷酸盐缓冲液(100mM,pH7.4)的配制:取预先配好的0.5M磷酸二氢钾150mL和700mL的0.5M磷酸氢二钾溶液混合,再用0.5M磷酸氢二钾溶液调节混合液pH值至7.4,使用前用超纯水稀释5倍,加入氯化镁,得到磷酸盐缓冲液(100mM),其中含100mM磷酸钾,3.3mM氯化镁,pH为7.4。Preparation of phosphate buffer (100 mM, pH 7.4): Mix 150 mL of pre-formed 0.5 M potassium dihydrogen phosphate and 700 mL of 0.5 M potassium dihydrogen phosphate solution, and adjust the mixture with 0.5 M potassium dihydrogen phosphate solution. The pH was adjusted to 7.4, diluted 5 times with ultrapure water before use, and magnesium chloride was added to obtain a phosphate buffer (100 mM) containing 100 mM potassium phosphate, 3.3 mM magnesium chloride, and a pH of 7.4.
配制NADPH再生系统溶液(含有6.5mM NADP,16.5mM G-6-P,3U/mL G-6-P D,3.3mM氯化镁),使用前置于湿冰上。A solution of NADPH regeneration system (containing 6.5 mM NADP, 16.5 mM G-6-P, 3 U/mL G-6-P D, 3.3 mM magnesium chloride) was prepared and placed on wet ice before use.
配制终止液:含有50ng/mL盐酸普萘洛尔和200ng/mL甲苯磺丁脲(内标)的乙腈溶液。取25057.5μL磷酸盐缓冲液(pH7.4)至50mL离心管中,分别加入812.5μL人肝微粒体,混匀,得到蛋白浓度为0.625mg/mL的肝微粒体稀释液。取25057.5μL磷酸盐缓冲液(pH7.4)至50mL离心管中,分别加入812.5μL SD大鼠肝微粒体,混匀,得到蛋白浓度为0.625mg/mL的肝微粒体稀释液。Formulation stop solution: acetonitrile solution containing 50 ng/mL propranolol hydrochloride and 200 ng/mL tolbutamide (internal standard). Take 25057.5 μL of phosphate buffer (pH 7.4) into a 50 mL centrifuge tube, add 812.5 μL of human liver microsomes, and mix to obtain a liver microsome dilution with a protein concentration of 0.625 mg/mL. 25057.5 μL of phosphate buffer (pH 7.4) was taken into a 50 mL centrifuge tube, and 812.5 μL of SD rat liver microsomes were added and mixed to obtain a liver microsome dilution having a protein concentration of 0.625 mg/mL.
样品的孵育:用含70%乙腈的水溶液将相应化合物的储备液分别稀释至0.25mM,作为工作液,备用。分别取398μL的人肝微粒体或者大鼠肝微粒体稀释液加入96孔孵育板中(N=2),分别加入2μL 0.25mM的的工作液中,混匀。Incubation of the sample: The stock solution of the corresponding compound was diluted to 0.25 mM with an aqueous solution containing 70% acetonitrile as a working solution, and was used. 398 μL of human liver microsomes or rat liver microsome dilutions were added to 96-well incubation plates (N=2), and 2 μL of 0.25 mM working solution was added and mixed.
代谢稳定性的测定:在96孔深孔板的每孔中加入300μL预冷的终止液,并置于冰上,作为终止板。将96孔孵育板和NADPH再生系统置于37℃水浴箱中,100转/分钟震荡,预孵5min。从孵育板每孔取出80μL孵育液加入终止板,混匀,补充20μL NADPH再生系统溶液,作为0min样品。再向孵育板每孔加入80μL的NADPH再生系统溶液,启动反应,开始计时。相应化合物的反应浓度为1μM,蛋白浓度为0.5mg/mL。分别于反应10、30、90min时,各取100μL反应液,加入终止板中,涡旋3min终止反应。将终止板于5000×g,4℃条件下离心10min。取100μL上清液至预先加入100μL蒸馏水的96孔板中,混匀,采用LC-MS/MS进行样品分析。Determination of metabolic stability: 300 μL of pre-cooled stop solution was added to each well of a 96-well deep well plate and placed on ice as a stop plate. The 96-well incubation plate and the NADPH regeneration system were placed in a 37 ° C water bath, shaken at 100 rpm, and pre-incubated for 5 min. 80 μL of the incubation solution was taken from each well of the incubation plate, added to the stopper plate, and mixed, and 20 μL of the NADPH regeneration system solution was added as a sample of 0 min. Then, 80 μL of the NADPH regeneration system solution was added to each well of the incubation plate to start the reaction and start timing. The corresponding compound had a reaction concentration of 1 μM and a protein concentration of 0.5 mg/mL. 100 μL of the reaction solution was taken at 10, 30, and 90 min, respectively, and added to the stopper, and the reaction was terminated by vortexing for 3 min. The plate was centrifuged at 5000 x g for 10 min at 4 °C. 100 μL of the supernatant was taken into a 96-well plate to which 100 μL of distilled water was previously added, mixed, and sample analysis was performed by LC-MS/MS.
数据分析:通过LC-MS/MS系统检测相应化合物及内标的峰面积,计算化合物与内标峰面积比值。通过化合物剩余量的百分率的自然对数与时间作图测得斜率,并根 据以下公式计算t1/2和CLint,其中V/M即等于1/蛋白浓度。Data analysis: The peak area of the corresponding compound and the internal standard was detected by LC-MS/MS system, and the ratio of the peak area of the compound to the internal standard was calculated. The slope is measured by the natural logarithm of the percentage of the remaining amount of the compound versus time, and t 1/2 and CL int are calculated according to the following formula, where V/M is equal to 1/protein concentration.
Figure PCTCN2016109851-appb-000021
Figure PCTCN2016109851-appb-000021
实验结果如下表2所示,本发明化合物在人肝微粒体与大鼠肝微粒体实验中都表现出优异的代谢稳定性。The experimental results are shown in Table 2 below. The compounds of the present invention exhibited excellent metabolic stability in both human liver microsomes and rat liver microsome experiments.
表2实施例化合物肝微粒代谢作用Table 2 Example compound liver particle metabolism
Figure PCTCN2016109851-appb-000022
Figure PCTCN2016109851-appb-000022
大鼠中的药代动力学评价。Pharmacokinetic evaluation in rats.
6只雄性Sprague-Dawley大鼠,7-8周龄,体重约210g,分成2组,每组3只,经静脉或口服单个剂量的化合物(经静脉3mg/kg,口服10mg/kg),比较其药代动力学差异。Six male Sprague-Dawley rats, 7-8 weeks old, weighing 210 g, divided into 2 groups, 3 in each group, intravenously or orally administered a single dose of compound (3 mg/kg intravenously, 10 mg/kg orally). Its pharmacokinetic differences.
大鼠采用标准饲料饲养,给予水。试验前16小时开始禁食。药物用PEG400和二甲亚砜溶解。眼眶采血,采血的时间点为给药后0.083小时,0.25小时、0.5小时、1小时、2小时、4小时、6小时、8小时、12小时和24小时。 Rats were fed a standard diet and given water. Fasting began 16 hours before the test. The drug was dissolved with PEG400 and dimethyl sulfoxide. Blood was collected from the eyelids at a time point of 0.083 hours, 0.25 hours, 0.5 hours, 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, and 24 hours after administration.
大鼠吸入乙醚后短暂麻醉,眼眶采集300μL血样于试管。试管内有30μL1%肝素盐溶液。使用前,试管于60℃烘干过夜。在随后一个时间点血样采集完成之后,大鼠乙醚麻醉后处死。Rats were briefly anesthetized after inhalation of ether, and 300 μL of blood samples were collected from the eyelids in test tubes. There was 30 μL of 1% heparin salt solution in the test tube. The tubes were dried overnight at 60 ° C before use. After the blood sample collection was completed at a later time point, the rats were anesthetized with ether and sacrificed.
血样采集后,立即温和地颠倒试管至少5次,保证混合充分后放置于冰上。血样在4℃5000rpm离心5分钟,将血浆与红细胞分离。用移液器吸出100μL血浆到干净的塑料离心管中,表明化合物的名称和时间点。血浆在进行分析前保存在-80℃。用LC-MS/MS测定血浆中本发明化合物的浓度。药代动力学参数基于每只动物在不同时间点的血药浓度进计算。Immediately after the blood sample is collected, gently invert the tube at least 5 times to ensure adequate mixing and place on ice. Blood samples were centrifuged at 5000 rpm for 5 minutes at 4 ° C to separate plasma from red blood cells. Pipette 100 μL of plasma into a clean plastic centrifuge tube to indicate the name and time point of the compound. Plasma was stored at -80 °C prior to analysis. The concentration of the compound of the invention in plasma was determined by LC-MS/MS. Pharmacokinetic parameters were calculated based on the plasma concentration of each animal at different time points.
实验结果如下表3所示,相对于对照化合物INT-747,本发明化合物19的口服利用率大幅度提高,说明其在动物体内具有更好的药物动力学。The experimental results are shown in Table 3 below. Compared with the control compound INT-747, the oral utilization rate of the compound 19 of the present invention was greatly improved, indicating that it has better pharmacokinetics in animals.
表3大鼠药代动力学实验Table 3 Rat pharmacokinetic experiments
Figure PCTCN2016109851-appb-000023
Figure PCTCN2016109851-appb-000023
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,实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则份数和百分比为重量份和重量百分比。It is to be understood that the examples are merely illustrative of the invention and are not intended to limit the scope of the invention, and the experimental methods in which the specific conditions are not indicated in the examples are generally in accordance with conventional conditions or in accordance with the conditions suggested by the manufacturer. Parts and percentages are parts by weight and percentage by weight unless otherwise stated.
以上内容是结合具体的优选实施方式对本发明所作的进一步详细说明,不能认定本发明的具体实施只局限于这些说明。对于本发明所属技术领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干简单推演或替换,都应当视为属于本发明的保护范围。 The above is a further detailed description of the present invention in connection with the specific preferred embodiments, and the specific embodiments of the present invention are not limited to the description. It will be apparent to those skilled in the art that the present invention may be made without departing from the spirit and scope of the invention.

Claims (10)

  1. 一种法尼醇X受体激动剂,其特征在于:如通式(I)所示取代的胆烷酸化合物、或其晶型、药学上可接受的盐:A farnesoid X-receptor agonist characterized by a substituted choline acid compound represented by the formula (I), or a crystalline form thereof, or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2016109851-appb-100001
    Figure PCTCN2016109851-appb-100001
    式中:R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15、R16、R17、R18、R19、R20、R21、R22、R23、R24、R25、R26、R27、R28和R29相互独立地选自由“氢(H)、氘(D)”组成的组;Wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 And R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 and R 29 are each independently selected from "hydrogen (H), a group consisting of 氘(D)";
    X1、X2、X3、X4相互独立地选自由“氢(H)、氘(D)、甲基、CH2D、CHD2、CD3、CH2CH3、CHDCH3、CHDCH2D、CHDCHD2、CHDCD3、CD2CH3、CD2CH2D、CD2CHD2、CD2CD3”组成的组;X 1 , X 2 , X 3 , X 4 are independently selected from the group consisting of "hydrogen (H), hydrazine (D), methyl, CH 2 D, CHD 2 , CD 3 , CH 2 CH 3 , CHDCH 3 , CHDCH 2 a group consisting of D, CHDCHD 2 , CHDCD 3 , CD 2 CH 3 , CD 2 CH 2 D, CD 2 CHD 2 , CD 2 CD 3 ”;
    及其生理学上可接受的盐、前药、互变异构体和立体异构体、对映异构体、水合物或溶剂合物,包括这些化合物以所有比例形成的混合物;And physiologically acceptable salts, prodrugs, tautomers and stereoisomers, enantiomers, hydrates or solvates thereof, including mixtures of these compounds in all ratios;
    附加条件是,所述化合物不包括非氘代化合物。Additionally, the compound does not include a non-deuterated compound.
  2. 如权利要求1所述化合物,其特征在于,X1、X2、X3、X4可独立选自一次或多次氘代的烷基。A compound according to claim 1 wherein X 1 , X 2 , X 3 , X 4 are independently selected from alkyl groups which are deuterated one or more times.
  3. 如权利要求1所述化合物,其特征在于,R17、R18、R19、R20各自独立地选自氢或氘。The compound of claim 1 wherein R 17 , R 18 , R 19 , R 20 are each independently selected from hydrogen or hydrazine.
  4. 如权利要求1所述化合物,其特征在于,选自以下组内的氘代胆烷酸化合物或其药学上可接受的盐,但不局限于如下化合物:The compound according to claim 1, which is selected from the group consisting of a deuterated cholic acid compound or a pharmaceutically acceptable salt thereof, but is not limited to the following compounds:
    Figure PCTCN2016109851-appb-100002
    Figure PCTCN2016109851-appb-100002
    Figure PCTCN2016109851-appb-100003
    Figure PCTCN2016109851-appb-100003
    Figure PCTCN2016109851-appb-100004
    Figure PCTCN2016109851-appb-100004
    Figure PCTCN2016109851-appb-100005
    Figure PCTCN2016109851-appb-100005
    Figure PCTCN2016109851-appb-100006
    Figure PCTCN2016109851-appb-100006
  5. 一种包含根据权利要求1所述的胆烷酸化合物或其药学上可接受盐以及药学上可接受载体的药物组合物,其特征在于,含有权利要求1所述的氘代化合物或其药学上可接受的盐作为有效成分,并含有常规药用载体。A pharmaceutical composition comprising the bile acid compound according to claim 1 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, comprising the deuterated compound according to claim 1 or a pharmaceutical thereof An acceptable salt is used as an active ingredient and contains a conventional pharmaceutical carrier.
  6. 如权利要求5所述的药物组合物,其特征在于,所述的药物组合物可以用于治疗、预防或消除各种FXR或者TGR5介导的相关病症;包含这些化合物的药物组合物用于在不同治疗领域诸如癌症中治疗、预防疾病或障碍或减慢所述疾病或障碍进程。The pharmaceutical composition according to claim 5, wherein said pharmaceutical composition is useful for treating, preventing or eliminating various FXR or TGR5 mediated related disorders; a pharmaceutical composition comprising these compounds is used in Treatment, prevention of disease or disorder, or slowing down the progression of the disease or disorder in different therapeutic areas, such as cancer.
  7. 权利要求1-4所述化合物用于制备治疗或者预防疾病的药物中的用途,所述疾病由FXR或者TGR5介导,可选自自慢性肝病,胃肠疾病,肾病,心血管疾病,淤胆失调,代谢疾病。Use of a compound according to claims 1-4 for the manufacture of a medicament for the treatment or prevention of a disease mediated by FXR or TGR5, which may be selected from the group consisting of chronic liver disease, gastrointestinal disease, kidney disease, cardiovascular disease, cholestatic Disorder, metabolic disease.
  8. 如权利要求7所述的用途,其中所述的慢性肝病为原发性胆汁性肝硬化和非酒精性脂肪性肝炎。The use according to claim 7, wherein said chronic liver disease is primary biliary cirrhosis and nonalcoholic steatohepatitis.
  9. 如权利要求7所述的用途,其中所述的代谢疾病为肥胖症和糖尿病。The use according to claim 7, wherein the metabolic diseases are obesity and diabetes.
  10. 一种在受试者中治疗和/或预防与各种FXR或者TGR5介导的疾病的方法,所述方法包括向所述受试者给药如权利要求1~4任意一项所述的式(I)化合物或其多晶型、药学上可接受的盐、前药、立体异构体、同位素变体、水合物或溶剂化合物,或者权利要求5中的药物组合物。 A method of treating and/or preventing a disease mediated by various FXR or TGR5 in a subject, the method comprising administering to the subject a formula according to any one of claims 1 to 4. (I) a compound or a polymorphic form thereof, a pharmaceutically acceptable salt, a prodrug, a stereoisomer, an isotope variant, a hydrate or a solvent compound, or the pharmaceutical composition of claim 5.
PCT/CN2016/109851 2016-03-11 2016-12-14 Cholanic acid compound for preventing or treating fxr-mediated diseases WO2017152677A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201610141005 2016-03-11
CN201610141005.5 2016-03-11
CN201610409536.8 2016-06-12
CN201610409536.8A CN106008639B (en) 2016-03-11 2016-06-12 For preventing or treating the disease mediated cholane acid compound of FXR-

Publications (1)

Publication Number Publication Date
WO2017152677A1 true WO2017152677A1 (en) 2017-09-14

Family

ID=57090959

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/109851 WO2017152677A1 (en) 2016-03-11 2016-12-14 Cholanic acid compound for preventing or treating fxr-mediated diseases

Country Status (2)

Country Link
CN (2) CN109467585A (en)
WO (1) WO2017152677A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113774142A (en) * 2021-10-11 2021-12-10 北京化工大学 Application of TGR5 in preparation of medicines for treating intestinal cancer and diagnostic kit

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109467585A (en) * 2016-03-11 2019-03-15 深圳市塔吉瑞生物医药有限公司 For preventing or treating the disease mediated cholane acid compound of FXR-
TW201802103A (en) * 2016-07-13 2018-01-16 江蘇恆瑞醫藥股份有限公司 Preparation method of obeticholic acid and intermediates thereof
MX2020000873A (en) * 2017-07-24 2020-07-22 Intercept Pharmaceuticals Inc Isotopically labeled bile acid derivatives.
US11267810B2 (en) 2017-08-30 2022-03-08 Shenzhen Targetrx, Inc. Aminopyrimidine compound and composition comprising same and use thereof
JP7150356B2 (en) * 2017-12-22 2022-10-11 深▲チェン▼市塔吉瑞生物医薬有限公司 Substituted pyrazolo[1,5-a]pyrimidine compounds and pharmaceutical compositions and uses thereof
JP7156722B2 (en) 2018-01-23 2022-10-19 深▲チェン▼市塔吉瑞生物医薬有限公司 substituted pyridazinone compounds
WO2019144885A1 (en) 2018-01-23 2019-08-01 深圳市塔吉瑞生物医药有限公司 Substituted pyrazolo[1,5-a]pyrimidine macrocyclic compound
TW202021595A (en) 2018-08-10 2020-06-16 德商菲尼克斯製藥股份有限公司 Isolithocholic acid or isoallolithocholic acid and deuterated derivatives thereof for preventing and treating clostridium difficile-associated diseases
JP7441475B2 (en) * 2020-07-21 2024-03-01 国立大学法人 東京大学 Deuterated labeled compound and its production method
CN113061070A (en) * 2021-03-29 2021-07-02 阿尔塔(天津)标准物质研究院有限公司 Deuterium-labeled metinuron stable isotope labeled compound

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002072598A1 (en) * 2001-03-12 2002-09-19 Roberto Pellicciari Steroids as agonists for fxr
WO2015061421A1 (en) * 2013-10-22 2015-04-30 Metselex, Inc. Deuterated bile acids
CN106008639A (en) * 2016-03-11 2016-10-12 深圳市塔吉瑞生物医药有限公司 Ursodeoxycholic acid compound for preventing or treating FXR (farnesol X receptor)-mediated disease

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104370990A (en) * 2008-11-19 2015-02-25 英特塞普特医药品公司 TGR5 modulators and methods of use thereof
AU2013277429B2 (en) * 2012-06-19 2016-01-14 Intercept Pharmaceuticals, Inc. Preparation, uses and solid forms of obeticholic acid
CN105985396A (en) * 2015-02-16 2016-10-05 苏州泽璟生物制药有限公司 Deuterated chenodeoxycholic acid derivative and pharmaceutical composition containing same

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002072598A1 (en) * 2001-03-12 2002-09-19 Roberto Pellicciari Steroids as agonists for fxr
WO2015061421A1 (en) * 2013-10-22 2015-04-30 Metselex, Inc. Deuterated bile acids
CN106008639A (en) * 2016-03-11 2016-10-12 深圳市塔吉瑞生物医药有限公司 Ursodeoxycholic acid compound for preventing or treating FXR (farnesol X receptor)-mediated disease

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
JIANG, WENFENG ET AL.: "Application of Deuteration in Drug Research", QILU PHARMACEUTICAL AFFAIRS, vol. 29, no. 11, 31 December 2010 (2010-12-31), pages 682, XP008173943 *
MOLECULAR NUCLEAR MEDICINE, 31 May 2001 (2001-05-31), pages 389 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113774142A (en) * 2021-10-11 2021-12-10 北京化工大学 Application of TGR5 in preparation of medicines for treating intestinal cancer and diagnostic kit

Also Published As

Publication number Publication date
CN106008639B (en) 2019-01-08
CN109467585A (en) 2019-03-15
CN106008639A (en) 2016-10-12

Similar Documents

Publication Publication Date Title
WO2017152677A1 (en) Cholanic acid compound for preventing or treating fxr-mediated diseases
US20230234968A1 (en) Methyl-substituted benzobisoxazole compound and use thereof
CN102666504B (en) Compound as lpa receptor antagonist
WO2019144835A1 (en) Substituted pyridazinone compound
WO2019007418A1 (en) Fxr receptor agonist
JP6158817B2 (en) Pyridazine derivatives useful for treatment
CN102245584A (en) Antagonists of lysophosphatidic acid receptors
BRPI0610183A2 (en) COMPOUND OR PHARMACEUTICALLY ACCEPTABLE SALT THEREOF, PHARMACEUTICAL COMPOSITION, AND USE OF COMPOUND OR PHARMACEUTICALLY ACCEPTABLE SALT THEREOF
MX2010012436A (en) Tetrahydrocyclopenta[b]indole androgen receptor modulators.
CN112839940A (en) Preparation and application of aminourea sensitive amine oxidase inhibitor
EP3831812B1 (en) Novel 3, 5-disubstituted pyridine and 3, 5-disubstituted pyridazine derivatives and pharmaceutical use of same
WO2020156459A1 (en) Pyrrolopyrimidine derivative and use thereof
JP6842474B2 (en) Steroid compounds, compositions containing the compounds and their use
CN112334467B (en) FXR receptor agonists
WO2020020377A1 (en) Fused ring derivative used as fgfr4 inhibitor
WO2019104748A1 (en) Use of compound in preparing drug
JP7265817B2 (en) Liver-targeting compounds with thyroid hormone receptor agonist properties and pharmaceutical compositions thereof
CN114057701A (en) Deuterated pyridazinone compound and application thereof
KR20230006563A (en) 4-(2,6-difluorophenoxy)-6-(trifluoromethyl)pyrimidin-2-amine derivatives as enhancers of the hMrgX1 receptor for the treatment of pain
WO2022184177A1 (en) Aromatic compound, preparation method therefor, and application thereof
WO2022184172A1 (en) New-type benzazepine fused ring derivative
WO2018214872A1 (en) Substituted pyrimidinetrione compound, composition comprising compound, and use thereof
CN115894456A (en) Deuterated pyrazole aminopyrimidine compound, pharmaceutical composition and application
JP2024501429A (en) Preparation method of pralsetinib
JP2023518399A (en) Crystalline Forms of Farnesoid X Receptor Agonists

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 16893320

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 16893320

Country of ref document: EP

Kind code of ref document: A1