WO2020114307A1 - Isoxazole derivative, preparation method therefor and use thereof - Google Patents

Isoxazole derivative, preparation method therefor and use thereof Download PDF

Info

Publication number
WO2020114307A1
WO2020114307A1 PCT/CN2019/121534 CN2019121534W WO2020114307A1 WO 2020114307 A1 WO2020114307 A1 WO 2020114307A1 CN 2019121534 W CN2019121534 W CN 2019121534W WO 2020114307 A1 WO2020114307 A1 WO 2020114307A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
pharmaceutical composition
metabolites
stereoisomers
Prior art date
Application number
PCT/CN2019/121534
Other languages
French (fr)
Chinese (zh)
Inventor
刘金明
蔡家强
吴勇勇
崔洪
王超磊
田强
宋宏梅
薛彤彤
王利春
王晶翼
Original Assignee
四川科伦博泰生物医药股份有限公司
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 四川科伦博泰生物医药股份有限公司 filed Critical 四川科伦博泰生物医药股份有限公司
Priority to CN201980066095.8A priority Critical patent/CN112805279B/en
Publication of WO2020114307A1 publication Critical patent/WO2020114307A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/422Oxazoles not condensed and containing further heterocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • the present invention relates generally to compounds for the treatment of diseases or conditions mediated by farnesoid X receptor (FXR), and more specifically to FXR agonist compounds, as well as their stereoisomers, tautomers, polymorphs Crystal forms, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N-oxides, and chemically protected forms and prodrugs thereof.
  • FXR farnesoid X receptor
  • the invention also relates to preparation methods, intermediates, pharmaceutical compositions and kits containing the compounds, and their therapeutic uses.
  • Farnesoid X receptor (FXR, NR1H4) is expressed in the liver, the entire gastrointestinal tract including the esophagus, stomach, duodenum, small intestine, and colon (Kuipers, F. et al., The Farnesoid X Receptor (FXR) as Modulator of Bile Acid Metabolism, Rev. Endocrine Metab. Disorders, 2004, 5: 319-326).
  • FXR is a member of a transcription factor known to be a ligand-activated nuclear receptor.
  • Bile acids such as chenodeoxycholic acid (CDCA) or its taurine or glycine amide conjugates are endogenous ligands of FXR.
  • the bile acid binds to FXR and activates FXR. It controls the expression of various genes through heterodimer complex with retinol X receptor (RXR), including the involvement of bile acid, cholesterol, triglyceride in liver and circulatory system.
  • RXR retinol X receptor
  • Gene expression of acid glyceride and lipoprotein homeostasis (Kalaany, NY; Mangelsdorf, DJ; LXRS and FXR: the yin and yang of cholesterol and fat metabolism, Annu. Rev. Physiol., 2006, 68, 159-191; Calkin , AC; Tontonoz, P.; Transcriptional integration of metabolism by the nuclear sterol-activated receptors LXR and FXR, Nat. Rev. Mol.
  • FXR also appears to be involved in paracrine and endocrine signaling by up-regulating fibroblast growth factor 15 (rodent) or fibroblast growth factor 19 (monkey, human) (T. Inagaki et al., Fibroblast growth factor 15 functions) aserohepatic signaling Regulated acid homeostasis. Cell Metab., 2005, 2(4), 217-225).
  • Bile acids are amphiphilic molecules that form micelles and emulsify the lipids in the diet. If the bile acid concentration is too high, cytotoxicity will also occur, so physiologically there is a mechanism to strictly control the bile acid concentration. FXR plays a key role in controlling the bile acid to maintain a stable state in the body (Makishima, M.; Nuclear Receptors as Targets for Drug Development: Regulation of Cholesterol and Bile Acid Metabolism by Nuclear Receptors, J. Pharmacol. Sci., 2005, 97 : 177-183).
  • FXR has been shown to regulate complex biological processes beyond metabolism, such as liver regeneration or the integrity of the intestinal barrier. FXR also controls the immune system of the intestine and liver and has a certain anti-inflammatory effect (Modica, S.; Gadaleta, RM; Moschetta, A.; Deciphering the nuclear acceptor FXR paradigm, Nucl. Recept. Signal., 2010 , 8, e005).
  • Obeticholic acid (6-Et CDCA) is an FXR receptor agonist that is more active than the endogenous ligand CDCA and has been shown in a phase IIa clinical study of non-alcoholic fatty liver disease (NAFLD) Significant improvement in insulin sensitivity and other beneficial effects on metabolism disease, Gastroenterology, 2013, 145, 574-582).
  • a phase IIb study of obeticholic acid showed that 72-week treatment is also beneficial for the improvement of histopathology of non-alcoholic hepatitis (NASH).
  • NASH non-alcoholic hepatitis
  • WO2012087519 discloses an agonist or partial agonist of FXR for the treatment of conditions mediated by FXR.
  • the FXR agonist compounds disclosed in the prior art still have defects in pharmacodynamics or pharmacokinetic properties.
  • the present invention relates generally to compounds of general formula (I) or their stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs,
  • X is CH or N
  • R is selected from hydrogen, halogen, C 1-6 alkyl and C 1-6 haloalkyl.
  • Another aspect of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising the compound of the general formula (I) or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceuticals Acceptable salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, and one or more pharmaceutically acceptable carriers.
  • the pharmaceutical composition may further comprise one or more other therapeutic agents suitable for preventing or treating diseases or conditions mediated by FXR.
  • the present invention also includes a method of preventing or treating a disease or condition mediated by FXR, the method comprising administering to a subject in need thereof a therapeutically effective amount of the compound of general formula (I) or its stereoisomer, Tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, or the pharmaceutical composition .
  • the present invention also includes a kit for preventing or treating a disease or condition mediated by FXR, which includes:
  • a) a first container which contains as a first therapeutic agent at least one compound of the general formula (I) or its stereoisomers, tautomers, polymorphs, solvates (eg hydrated) Substances), pharmaceutically acceptable salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, or the pharmaceutical composition as the first pharmaceutical composition;
  • the present invention also includes the compound of the general formula (I) or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites , N-oxide, its chemically protected form or prodrug, or the pharmaceutical composition, which is used to prevent or treat a disease or condition mediated by FXR.
  • the present invention also includes the compound of the general formula (I) or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites , N-oxide, its chemically protected form or prodrug, or the use of the pharmaceutical composition in the preparation of a medicament for the prevention or treatment of a disease or disorder mediated by FXR.
  • the invention also includes methods and corresponding intermediates for preparing the compounds of the invention.
  • the compound of the general formula (I) of the present invention has excellent in vivo or in vitro pharmacodynamics or pharmacokinetic properties, shows good FXR activation activity and activation, and excellent liver microsomal stability, and thus has good Drug activity and metabolic advantages.
  • Figure 1 shows the effect of administering compound C3 in a mouse fatty liver model, where a represents p ⁇ 0.05 compared to the control group; b represents p ⁇ 0.05 compared to the model group.
  • alkyl refers to a saturated linear or branched hydrocarbon group having 1 to 6 carbon atoms (C 1-6 ), wherein the alkyl group may be optionally substituted by one or more (eg, 1 , 2, 3, or 4) suitable substituents.
  • the alkyl group has 1 to 6 carbon atoms (C 1-6 ).
  • the alkyl group has 1 to 4 carbon atoms (C 1-4 ), especially 1 to 3 carbon atoms (C 1-3 ) or 1 to 2 carbon atoms (C 1-2 ) .
  • alkyl groups include, but are not limited to: methyl (Me), ethyl (Et), 1-propyl (n-Pr), 2-propyl (i-Pr or isopropyl), 1-butyl ( n-Bu or n-butyl), 2-methyl-1-propyl (i-Bu or isobutyl), 2-butyl (s-Bu or sec-butyl), 2-methyl-2-propyl (T-Bu or tert-butyl), 1-pentyl (n-pentyl), 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-2-butyl , 3-methyl-1-butyl, 2-methyl-1-butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl (-C(CH 3 ) 2 CH 2 CH 2 CH 3 ), 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2-methyl
  • halo or halogen as used herein includes F, Cl, Br or I.
  • Halo includes but is not limited to mono-, di- or tri-substitution, and the halogen atoms used for substitution may be the same or different.
  • haloalkyl as used herein includes fluoroalkyl, chloroalkyl, bromoalkyl, or iodoalkyl.
  • chiral refers to molecules that have non-superimposability of mirror image pairs
  • achiral refers to molecules that can overlap on their mirror image pairs
  • stereoisomer refers to compounds that have the same chemical composition but differ in the spatial arrangement of atoms or groups.
  • Diastereomer refers to a stereoisomer that has two or more chiral centers and whose molecules are not mirror images of each other. Diastereomers have different physical properties, such as melting point, boiling point, spectral properties, and reactivity. The mixture of diastereomers can be separated by high-resolution analytical methods such as electrophoresis and chromatography.
  • Enantiomer refers to two stereoisomers of a compound that are non-overlapping mirror images of each other.
  • the stereochemical definitions and rules used in this article generally follow SPParker, Ed., McGraw-Hill Dictionary of Chemicals (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., "Stereochemistry” of Organic Compounds”, John Wiley & Sons, Inc., New York, 1994.
  • the compounds of the present invention may contain asymmetric or chiral centers and therefore exist in different stereoisomeric forms. All stereoisomeric forms of the compounds of the invention, including but not limited to their diastereomers, enantiomers and atropisomers, and mixtures thereof, such as racemic mixtures, are intended to constitute the invention a part of.
  • optically active compounds that is, they have the ability to rotate the plane of plane polarized light.
  • the prefixes D and L, or R and S are used to indicate the absolute configuration of the chiral center of the molecule.
  • the prefixes d and l or (+) and (-) are used to indicate the sign that the compound rotates plane polarized light, where (-) or l means that the compound is levorotatory.
  • Compounds with the prefix (+) or d are right-handed. For a specific chemical structure, these stereoisomers are the same, but they are mirror images of each other.
  • a specific stereoisomer can also be called an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture.
  • a 50:50 mixture of enantiomers is called a racemic mixture or a racemate, which can occur in a chemical reaction or process without stereoselectivity or stereospecificity.
  • the terms "racemic mixture” and “racemate” refer to an equimolar mixture of two enantiomers, which is not optically active.
  • the stereoisomers of the invention may exist in a predominant form, for example, greater than 50%ee (enantiomeric excess), greater than 80%ee, greater than 90%ee, greater than 95%ee, or greater than 99% ee.
  • the method for preparing the compound of the present invention produces a mixture of stereoisomers
  • these isomers can be separated by conventional techniques such as preparative chromatography.
  • the compound can be prepared in racemic form, or a single enantiomer can be prepared by selective synthesis of enantiomers or by resolution.
  • diastereoisomers are formed by standard techniques, for example by forming salts with optically active acids such as (-)-di-p-toluoyl-d-tartaric acid and/or (+)-di-p-toluoyl-l-tartaric acid Pairs, followed by step crystallization and free base regeneration, the compounds can be resolved into their enantiomeric components.
  • the compounds can also be resolved by forming diastereomeric esters or amides, followed by chromatographic purification and removal of chiral auxiliaries. Alternatively, the compound can be resolved using a chiral HPLC column.
  • tautomer or “tautomeric form” refers to structural isomers of different energies that can be interconverted by a low energy barrier.
  • proton tautomers also known as proton transfer tautomers
  • Valence tautomers include interconversion through the recombination of some bonding electrons.
  • the present invention encompasses all possible crystalline forms or polymorphs of the compound of general formula (I), which may be a single polymorph or a mixture of more than one polymorph in any ratio.
  • pharmaceutically acceptable derivatives include, but are not limited to: pharmaceutically acceptable salts, esters, solvates, metabolites, N-oxides, and chemically protected forms and prodrugs. After administration to an individual in need, the compound of the present invention or its metabolite or residue can be provided directly or indirectly.
  • pharmaceutically acceptable salts refers to pharmaceutically acceptable organic or inorganic salts of the compounds of the present invention.
  • Exemplary salts include but are not limited to isonicotinate, salicylate, acid citrate, oleate, tannate, pantothenate, gluconate, glucuronate, glucuronic acid Salts and parabens (ie 1,1'-methylene-bis(2-hydroxy-3-naphthoate)), etc.
  • the pharmaceutically acceptable salt may include the inclusion of another molecule of counter ion.
  • the counter ion can be any organic or inorganic ion that stabilizes the charge on the parent compound.
  • pharmaceutically acceptable salts may have more than one charged atom in their structure. Where multiple charged atoms are part of a pharmaceutically acceptable salt, there may be multiple counter ions.
  • a pharmaceutically acceptable salt can have one or more charged atoms and/or one or more counter ions.
  • the desired pharmaceutically acceptable salt can be prepared by any suitable method available in the art, for example, with an inorganic acid or with an organic acid such as salicylic acid, pyranosidic acid such as glucose
  • an inorganic acid or with an organic acid such as salicylic acid, pyranosidic acid such as glucose
  • pyranosidic acid such as glucose
  • the free base is treated with uronic acid or galacturonic acid, ⁇ -hydroxy acid amino acid, aromatic acid, etc.
  • the desired pharmaceutically acceptable salt can be prepared by any suitable method, for example, using an inorganic or organic base such as an amine (primary, secondary, or tertiary amine), alkali metal hydroxide, or Alkaline earth metal hydroxides, etc. treat free acids.
  • an inorganic or organic base such as an amine (primary, secondary, or tertiary amine), alkali metal hydroxide, or Alkaline earth metal hydroxides, etc. treat free acids.
  • suitable salts include, but are not limited to, organic salts derived from amino acids and cyclic amines, and inorganic salts.
  • pharmaceutically acceptable means that the substance or composition must be chemically and/or toxicologically compatible with the other components making up the formulation and/or the mammal treated with it.
  • ester means an ester derived from a compound of general formula (I), including physiologically hydrolyzable esters, which can be hydrolyzed under physiological conditions to release the free acid or alcohol form of the general formula of the present invention ( I) Compound.
  • the compound of the general formula (I) of the present invention may itself be an ester.
  • the compound of the present invention may exist in the form of a solvate (such as a hydrate), wherein the compound of the present invention contains a polar solvent as a structural element of the crystal lattice of the compound, particularly, for example, water, methanol, or ethanol.
  • a polar solvent as a structural element of the crystal lattice of the compound, particularly, for example, water, methanol, or ethanol.
  • the amount of polar solvents, especially water can be present in stoichiometric or non-stoichiometric ratios.
  • a “metabolite” is a product produced by metabolism of a specific compound or its salt in the body. Metabolites of compounds can be identified using conventional techniques known in the art and tests such as those described herein can be used to determine their activity. Such products can be produced, for example, by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymatic hydrolysis, etc. of the administered compound. Therefore, the present invention includes metabolites of the compounds of the present invention, including compounds produced by a method including contacting a compound of the general formula (I) of the present invention with a mammal for a period of time sufficient to produce its metabolite.
  • Synthetic methods for preparing N-oxides of heterocycles and tertiary amines are well known to those skilled in the art and include the use of peroxyacids such as peroxyacetic acid and m-chloroperoxybenzoic acid (m-CPBA), hydrogen peroxide, Alkyl hydrogen peroxides such as t-butyl hydroperoxide, sodium perborate and dioxiranes such as dimethyl dioxirane oxidize heterocycles and tertiary amines.
  • peroxyacids such as peroxyacetic acid and m-chloroperoxybenzoic acid (m-CPBA)
  • hydrogen peroxide Alkyl hydrogen peroxides such as t-butyl hydroperoxide
  • sodium perborate and dioxiranes such as dimethyl dioxirane oxidize heterocycles and tertiary amines.
  • any process for preparing the compounds of the present invention it may be necessary and/or desirable to protect sensitive or reactive groups on any relevant molecules, thereby forming a chemically protected form of the compounds of the present invention.
  • This can be achieved through conventional protecting groups, for example, as described in Protective Groups in Organic Chemistry, ed. JFW McOmie, Plenum Press, 1973; and TW Greene & PGMWuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991 Those protecting groups, these references are incorporated herein by reference.
  • the protecting group can be removed at an appropriate subsequent stage.
  • the invention further includes within its scope prodrugs of the compounds of the invention.
  • a prodrug will be a functional group derivative of the compound, which is easily converted into the desired therapeutically active compound in vivo. Therefore, in these cases, the term "administration" used in the treatment method of the present invention should include the treatment of various diseases or conditions with one or more prodrug forms of the claimed compounds, but After the individual is administered, the prodrug form is converted into the above compound in vivo.
  • “Design of Prodrug” ed. H. Bundgaard, Elsevier, 1985, a conventional method for selecting and preparing suitable prodrug derivatives is described.
  • any general formula or structure shown herein, including the compound of general formula (I), is also intended to represent the unlabeled form and the isotopically labeled form of the compound.
  • Isotope-labeled compounds have the structure shown in the molecular formula given herein, except that one or more atoms are replaced with atoms having a selected atomic mass or mass number.
  • isotopes that may be included in the compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine, and chlorine, such as, but not limited to, 2 H (deuterium, D), 3 H (tritium), 11 C, 13 C, 14 C, 15 N, 18 F, 31 P, 32 P, 35 S, 36 Cl and 125 I.
  • isotope-labeled compounds of the present invention for example, those containing radioisotopes such as 3 H, 13 C, and 14 C.
  • isotopically labeled compounds can be used in metabolic studies, reaction kinetic studies, detection or imaging techniques, such as positron emission tomography (PET) or single photon emission tomography (SPECT), including drug or substrate tissue distribution Determination, or used for radiotherapy of patients.
  • PET positron emission tomography
  • SPECT single photon emission tomography
  • the deuterium-labeled or substituted therapeutic compounds of the invention may have improved DMPK (pharmacokinetic and pharmacokinetic) properties related to distribution, metabolism and excretion (ADME).
  • substitution with heavier isotopes such as deuterium may provide certain therapeutic advantages due to greater metabolic stability, for example, increased half-life in the body, or reduced dose requirements.
  • 18 F labeled compounds can be used for PET or SPECT studies.
  • the isotopically-labeled compounds and prodrugs of the present invention can generally be prepared by replacing the non-isotopically-labeled reagents with readily available isotopically-labeled reagents by the methods disclosed in the implementation route or examples and the following preparation methods.
  • substitution with heavier isotopes, especially deuterium (ie, 2 H or D) may provide certain therapeutic advantages due to greater metabolic stability, such as increased half-life in vivo or reduced dose requirements or improved therapeutic index.
  • deuterium is regarded as a substituent in the compound of formula (I).
  • concentration of such heavier isotopes, especially deuterium can be defined by the isotope enrichment factor.
  • any atom not specifically designated as a specific isotope is intended to mean any stable isotope of that atom.
  • any atom clearly deuterium (D) is intended to represent deuterium.
  • composition includes products containing a therapeutically effective amount of a compound of formula (I) of the present invention, as well as any product produced directly or indirectly from a combination of compounds of formula (I) of the present invention.
  • the present invention provides compounds of general formula (I) or their stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters , Metabolites, N-oxides, their chemically protected forms or prodrugs,
  • X is CH or N
  • R is selected from hydrogen, halogen, C 1-6 alkyl and C 1-6 haloalkyl.
  • the present invention provides compounds or their stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N- Oxides, their chemically protected forms or prodrugs, where R is selected from hydrogen, fluorine, methyl and trifluoromethyl.
  • the present invention provides compounds or their stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N- Oxides, their chemically protected forms or prodrugs, where X is CH; and R is selected from hydrogen, fluorine, methyl and trifluoromethyl.
  • the present invention provides compounds or their stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N- Oxides, their chemically protected forms or prodrugs, where X is N; and R is hydrogen.
  • the present invention provides compounds or their stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N- Oxides, their chemically protected forms or prodrugs, wherein the compound is selected from:
  • the compound of the general formula (I) of the present invention may contain an asymmetric center or a chiral center, and thus may exist in different stereoisomeric forms. All stereoisomeric forms of the compounds of the present invention, including but not limited to their diastereomers, enantiomers, and atropisomers, and mixtures thereof, such as racemic mixtures, are intended to constitute the present invention portion.
  • the present invention covers all diastereomers, including cis-trans (geometric) isomers and conformational isomers.
  • cis-trans geometric isomers
  • conformational isomers for example, if the compound of general formula (I) contains a double bond or a fused ring, the cis and trans forms and mixtures thereof are covered within the scope of the present invention.
  • the stereochemistry is indicated by a solid wedge or dashed line representing a specific configuration, then the stereoisomer is so designated and defined.
  • the compounds of the present invention can exist in unsolvated forms as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and the present invention is intended to encompass both solvated and unsolvated forms.
  • the compounds of the invention can also exist in different tautomeric forms, and all such forms are encompassed within the scope of the invention.
  • Another aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising at least one compound of the general formula (I) of the present invention as described above or a stereoisomer, tautomer, polymorph, or solvate thereof (Eg hydrate), pharmaceutically acceptable salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, and one or more pharmaceutically acceptable carriers.
  • the pharmaceutical composition may further include one or more other therapeutic agents, such as other therapeutic agents suitable for preventing or treating diseases or conditions mediated by FXR.
  • “Pharmaceutically acceptable carrier” in the present invention refers to diluents, adjuvants, excipients or vehicles administered together with the active ingredient, and it is suitable for contact with humans and/or others within the scope of reasonable medical judgment Animal tissue without excessive toxicity, irritation, allergic reactions or other problems or complications corresponding to a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable carriers that can be used in the pharmaceutical composition of the present invention include, but are not limited to, sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil , Sesame oil, etc.
  • sterile liquids such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil , Sesame oil, etc.
  • water is an exemplary carrier.
  • Physiological saline and aqueous solutions of glucose and glycerin can also be used as liquid carriers, especially for injections.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, maltose, chalk, silica gel, sodium stearate, glyceryl monostearate, talc, sodium chloride, skimmed milk powder, glycerin, propylene glycol, water, Ethanol, etc.
  • the composition may also contain small amounts of wetting agents, emulsifying agents or pH buffering agents as needed.
  • Oral preparations may contain standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, cellulose, magnesium carbonate, and the like. Examples of suitable pharmaceutically acceptable carriers are described in Remington's Pharmaceuticals (1990).
  • the pharmaceutical composition of the present invention can act systemically and/or locally.
  • they can be administered by suitable routes, for example by injection, intravenous, intraarterial, subcutaneous, intraperitoneal, intramuscular or transdermal administration; or by oral, buccal, nasal, transmucosal, topical, It is administered in the form of an ophthalmic preparation or by inhalation.
  • the pharmaceutical composition of the present invention can be administered in a suitable dosage form.
  • the dosage form includes but is not limited to tablets, capsules, lozenges, hard candy, powder, spray, cream, ointment, suppository, gel, paste, lotion, ointment, aqueous suspension , Injectable solutions, elixirs, syrups.
  • Another aspect of the invention provides therapeutic uses of the compounds and pharmaceutical compositions.
  • the present invention relates to a method of preventing or treating a disease or disorder mediated by FXR, the method comprising administering to a subject in need thereof a therapeutically effective amount of at least one general formula of the present invention ( I)
  • the present invention relates to at least one compound of the general formula (I) of the present invention or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), Pharmaceutically acceptable salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, or the pharmaceutical composition of the invention in the preparation of a medicament for the prevention or treatment of diseases or conditions mediated by FXR the use of.
  • FXR diseases or conditions mediated by FXR include but are not limited to:
  • Inflammatory bowel disease dyslipidemia, atherosclerosis, diabetes and related diseases; lipid and lipoprotein disorders; type II diabetes and clinical complications of type I and type II diabetes, including diabetic nephropathy, diabetic neuropathy , Diabetic retinopathy, and other clinically apparent effects of long-term diabetes; chronic fatty and fibrotic degeneration due to forced lipid accumulation, especially triglycerides, and subsequent activation of pro-fibrotic pathways
  • diseases such as non-alcoholic fatty liver disease (NAFLD) or non-alcoholic steatohepatitis (NASH); obesity or metabolic syndrome (combined disorders of dyslipidemia, diabetes and abnormally high body mass index);
  • treatment means reversing, alleviating, or inhibiting the indicated disease or disorder or the progression of one or more symptoms of such disease or disorder.
  • “Individual” as used herein includes human or non-human animals.
  • Exemplary human individuals include human individuals (referred to as patients) or normal individuals with a disease (such as the diseases described herein).
  • “non-human animals” include all vertebrates, such as non-mammals (eg birds, amphibians, reptiles) and mammals, such as non-human primates, domestic animals and/or domesticated animals (eg sheep, dogs) , Cats, cows, pigs, etc.).
  • therapeutically effective amount refers to the amount of the compound that will achieve the above-mentioned therapeutic efficacy after being administered.
  • the dosage regimen can be adjusted to provide the most desired response. For example, a single bolus injection can be administered, several divided doses can be administered over time, or the dosage can be reduced or increased proportionally as indicated by the urgent need for the treatment. It should be noted that the dose value may vary with the type and severity of the condition to be reduced, and may include single or multiple doses. It is to be further understood that for any particular individual, the specific dosing regimen should be adjusted over time based on the individual's needs and the professional judgment of the person administering the composition or supervising the administration of the composition.
  • the amount of the compound of the present invention administered will depend on the individual, the severity of the condition or condition being treated, the rate of administration, the treatment of the compound, and the judgment of the prescribing physician.
  • the effective dose is about 0.0001 to about 50 mg per kg of body weight per day, for example about 0.01 to about 10 mg/kg/day (single or divided doses). For a 70 kg person, this would add up to about 0.007 mg/day to about 3500 mg/day, such as about 0.7 mg/day to about 700 mg/day.
  • a dose level not higher than the lower limit of the aforementioned range may be sufficient, while in other cases, a larger dose may still be used without causing any harmful side effects, provided that the larger The dose is divided into several smaller doses to be administered throughout the day.
  • the content or amount of the compound of the present invention in the pharmaceutical composition may be about 0.01 mg to about 1000 mg, suitably 0.1-500 mg, preferably 0.5-300 mg and the like.
  • the compound of general formula (I) may be used alone or in combination with one or more other therapeutic agents suitable for preventing or treating diseases or conditions mediated by FXR.
  • the compound of general formula (I) in the pharmaceutical composition or as a dosing regimen for combination therapy, is combined with other therapeutic agents having anti-hyperproliferative efficacy, for example.
  • the other therapeutic agent may be, for example, a chemotherapeutic agent.
  • the pharmaceutical composition or other therapeutic agents of the administration regimen preferably have complementary activities to the compound of general formula (I) so that they do not adversely affect each other. Such compounds are suitably present in combination in amounts effective for the intended purpose.
  • Combination therapy can be administered simultaneously or sequentially. When administered sequentially, the combination can be administered in two or more doses.
  • Combined administration includes simultaneous administration using separate pharmaceutical compositions or a single pharmaceutical composition containing a compound of general formula (I) and other therapeutic agents, and sequential administration in any order, with two (or all) preferably present Active agents simultaneously exert their biological activity for a period of time.
  • Suitable doses of any of the above-mentioned concurrently administered agents are those currently in use, and can be reduced due to the combined (synergistic) effect of the newly identified drug with other therapeutic agents or treatments.
  • Combination therapy can provide "synergy” and prove to be “synergistic", that is, the effects achieved when the active ingredients are used together are greater than the sum of the effects produced when the compounds are used separately.
  • the active ingredient (1) is co-formulated in a combined unit dose formulation and is simultaneously administered or delivered; (2) is delivered as a separate formulation alternately or in parallel; or (3) by some other regimen, Can achieve synergy.
  • a synergistic effect can be achieved when the compounds are administered or delivered sequentially, for example by separate injections in separate syringes, by separate pills or capsules, or by separate infusions.
  • an effective dose of each active ingredient is administered sequentially, that is, continuously, while in combination therapy, an effective dose of two or more active ingredients is administered together.
  • the compound of general formula (I) or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, Esters, metabolites, N-oxides, their chemically protected forms or prodrugs can be combined with other therapeutic agents such as those described herein, and can also be combined with surgical treatment and radiotherapy. Therefore, the combination therapy of the present invention includes administration of at least one compound of general formula (I) or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable Salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, and the use of at least one other treatment. In order to achieve the desired combined therapeutic effect, the amount of the compound of general formula (I) and other therapeutic agents and the relative timing of administration are selected.
  • the present invention includes metabolites of the compound of the general formula (I), including compounds prepared by contacting the compound of the present invention with a mammal for a time sufficient to produce its metabolite.
  • Metabolites are usually prepared by preparing a compound labeled with a radioisotope of the present invention (for example, 14 C or 3 H) and administering it to animals such as rats, mice, guinea pigs, and monkeys in a detectable dose (for example, greater than about 0.5 mg/kg) Or human parenteral administration, metabolism for a sufficient period of time (usually about 30 seconds to 30 hours), and then its transformation product is isolated from urine, blood or other biological samples for identification. These products are easy to isolate because they are labeled (others are separated by using antibodies that can bind to the remaining epitopes in the metabolites).
  • the metabolite structure is determined by conventional methods, for example by MS, LC/MS or NMR analysis. The analysis of metabolites is performed in the same way as conventional drug metabolism studies well known to those skilled in the art. Metabolites, as long as they are not found in the body, can be used in diagnostic assays to administer the compounds of the present invention therapeutically.
  • kits containing materials for treating the above diseases or conditions.
  • the kit includes a container containing the compound of general formula (I), its stereoisomers, tautomers, polymorphs, solvates (such as hydrates) as the first therapeutic agent , A pharmaceutically acceptable salt, ester, metabolite, N-oxide, or a chemically protected form or prodrug thereof, or a pharmaceutical composition of the present invention containing the first pharmaceutical composition.
  • the kit may also include a label or package insert on or accompanying the container.
  • the term "package insert” refers to the instructions usually included in commercial packages of therapeutic products, which contain information on indications, usage, dosage, administration, contraindications and/or warnings related to the use of the therapeutic product.
  • Suitable containers include, for example, bottles, vials, syringes, blister packs, and the like.
  • the container may be made of various materials such as glass and plastic.
  • the container may contain a compound of general formula (I) or a preparation thereof that is effective for treating a condition, and may have a sterile inlet (for example, the container may be an intravenous solution sachet or have a pierceable by a hypodermic injection needle) Corks).
  • the label or package insert indicates that the composition is used to treat a selected condition, such as cancer.
  • the label or package insert may indicate that the patient to be treated is a patient with a disease or condition such as cirrhosis, hyperproliferative disorder, atherosclerosis, type I diabetes, or the label or package insert may also indicate the composition Can be used to treat other conditions.
  • the kit further includes a second container containing, as a second therapeutic agent, at least one other therapeutic agent suitable for preventing or treating a disease or condition mediated by FXR, or as a second A pharmaceutical composition comprising the other therapeutic agent. Therefore, in some embodiments, the kit may include instructions for administering the first therapeutic agent or first pharmaceutical composition and the second therapeutic agent or second pharmaceutical composition (if present).
  • the kit may also include simultaneous, sequential or Instructions for administering the first pharmaceutical composition and the second pharmaceutical composition separately.
  • the kit may further include a third container containing a pharmaceutically acceptable buffer such as bacteriostatic water for injection (BWFI), phosphate buffered saline, Ringer's solution, and glucose solution.
  • BWFI bacteriostatic water for injection
  • the kit may also include other materials desirable for commercial and user purposes, including other buffers, diluents, fillers, injection needles, and syringes.
  • the kit is suitable for the delivery of solid oral forms of compounds of general formula (I) such as tablets or capsules.
  • a kit preferably includes multiple unit doses.
  • Such kits may include cards with dosages positioned for their intended use.
  • An example of such a kit is a "blister pack".
  • Blister packaging is well known in the packaging industry and is widely used for packaging pharmaceutical unit dosage forms.
  • memory aids can be provided in the form of numbers, letters, or other indicia or calendar inserts that specify the date of administration in the treatment schedule, for example.
  • the present invention provides a method of preparing a compound of general formula (I) of the present invention, the method comprising the following steps:
  • Hal 1 , Hal 2 and Hal 3 are each independently the same or different halogen, such as F, Cl, Br or I, preferably Cl or Br;
  • PG 1 is an amino protecting group, preferably tert-butoxycarbonyl (Boc);
  • PG 2 is a carboxyl protecting group, preferably C 1-6 alkyl, and more preferably methyl;
  • Y is a boric acid or borate group, preferably -B(OH) 2 or
  • reaction conditions of each step are as follows:
  • Step A reacting compound IN-1 with compound IN-2 to obtain compound IN-3;
  • the reaction is preferably carried out in a suitable organic solvent.
  • the organic solvent can be selected from linear or cyclic ethers (such as tetrahydrofuran or diethyl ether), N-methylpyrrolidone, N,N-dimethylformamide, N,N-dimethylacetamide, 1, 4-Dioxane, dimethyl sulfoxide and any combination thereof, preferably tetrahydrofuran or N,N-dimethylformamide.
  • the reaction is preferably carried out in the presence of a suitable base (eg alkali metal alkoxide or carbonate) and/or catalyst.
  • the catalyst may be a catalyst system including a crown ether, and the crown ether may be selected from 15-crown-5 and 18-crown-6; the alkali metal carbonate is, for example, potassium carbonate or cesium carbonate; The alkali metal alkoxide may be selected from sodium tert-butoxide, potassium tert-butoxide, sodium methoxide, potassium methoxide, sodium ethoxide, potassium ethoxide.
  • the alkali metal alkoxide and catalyst are a combination of sodium t-butoxide and/or potassium t-butoxide and 15-crown-5 and/or 18-crown-6, preferably sodium t-butoxide and 15 -Combination of crown ether-5 or potassium tert-butoxide and 18-crown-6.
  • the reaction is preferably carried out at a suitable temperature.
  • the temperature is preferably room temperature (20-30°C) or 50-100°C (eg 50-80°C).
  • the reaction is preferably carried out for a suitable time, for example 1-24 hours, for example 5-15 hours.
  • Step B Remove the PG 1 group in compound IN-3 to obtain compound IN-4;
  • the reaction is preferably carried out in a suitable organic solvent.
  • the organic solvent may be selected from halogenated hydrocarbons (for example, dichloromethane, chloroform, ethyl chloride, dichloroethane, trichloroethane), N,N-dimethylformamide, N,N-di Methylacetamide and any combination thereof are preferably dichloromethane.
  • the reaction can be carried out under acidic conditions, such as 1,4-dioxane in hydrogen chloride; or suitable organic acids (such as carboxylic acids or halogenated acids, including but not limited to formic acid, fluoroacetic acid, difluoro Acetic acid, trifluoroacetic acid, chloroacetic acid, dichloroacetic acid, trichloroacetic acid and combinations thereof, preferably trifluoroacetic acid).
  • the reaction is preferably carried out at a suitable temperature.
  • the temperature is preferably room temperature (20-30°C).
  • the reaction is preferably carried out for a suitable time, such as 1-5 hours or 6-15 hours, such as 2 hours, 4 hours or overnight.
  • Step C reacting compound IN-4 with compound IN-a to obtain compound IN-5;
  • compound IN-4 is substituted with compound IN-a to give compound IN-5.
  • the substitution reaction is preferably carried out in a suitable organic solvent.
  • the organic solvent may be selected from N,N-dimethylformamide, N,N-dimethylacetamide, tetrahydrofuran, N-methylpyrrolidone, dimethylsulfoxide and any combination thereof, preferably N,N- Dimethylformamide or N,N-dimethylacetamide.
  • the substitution reaction is preferably carried out in the presence of a suitable base.
  • the base is an organic base (eg organic amines such as triethylamine, N,N-diisopropylethylamine, N-methylmorpholine or pyridine, preferably triethylamine or N,N-diiso Propylethylamine) or inorganic bases (eg alkali metal salts, preferably potassium carbonate).
  • organic bases eg organic amines such as triethylamine, N,N-diisopropylethylamine, N-methylmorpholine or pyridine, preferably triethylamine or N,N-diiso Propylethylamine
  • inorganic bases eg alkali metal salts, preferably potassium carbonate.
  • the substitution reaction is preferably carried out at a suitable temperature.
  • the temperature may be 20-150°C, for example 30-140°C, preferably 25°C, 50°C, 100°C or 130°C, preferably 80°C.
  • the substitution reaction is
  • the compound IN-4 is coupled with the compound IN-a to give compound IN-5.
  • the coupling reaction is preferably carried out in the presence of a metal catalyst and a base.
  • the metal catalyst is a palladium metal catalyst, such as tris(dibenzylideneacetone) dipalladium, [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride, triphenyl Phosphorus palladium and palladium acetate, preferably tris(dibenzylideneacetone) dipalladium.
  • the base is an inorganic base such as potassium carbonate, cesium carbonate, sodium carbonate, sodium bicarbonate, potassium bicarbonate, preferably cesium carbonate.
  • the coupling reaction is carried out in the presence of an organophosphorus compound derived from biphenyl, which is selected from BINAP, RuPhos and XPhos, preferably BINAP.
  • the coupling reaction is carried out in a suitable organic solvent, which may be selected from benzene, toluene and xylene, for example toluene.
  • the coupling reaction is carried out under a suitable protective atmosphere (for example, a nitrogen atmosphere).
  • the coupling reaction is carried out at a suitable temperature, which may be 70-100°C, preferably 80°C.
  • the coupling reaction is carried out for a suitable time, such as 1-3 hours, such as 2 hours.
  • Step D reacting compound IN-5 with compound IN-b to obtain compound IN-6;
  • the compound IN-5 and the compound IN-b undergo a metal-catalyzed coupling reaction to obtain the compound IN-6.
  • the metal-catalyzed coupling reaction is carried out by conventional methods.
  • compound IN-5 and compound IN-b are dissolved in a solvent (such as water, an organic solvent, or a mixed solvent of organic solvent and water), a palladium catalyst and a base are added, optionally under nitrogen protection, at 50°C to
  • the reaction is carried out at a temperature of 120°C (preferably 80°C or 90°C) for 8 to 24 hours (preferably 8 hours or 12 hours).
  • the organic solvent is N,N-dimethylformamide, tetrahydrofuran, 1,4-dioxane, toluene or DME.
  • the palladium catalyst is tris(dibenzylideneacetone) dipalladium, [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride, triphenylphosphine palladium, palladium acetate, preferably [ 1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride or tris(dibenzylideneacetone)dipalladium etc.
  • the base is preferably an inorganic base, such as potassium carbonate, cesium carbonate, sodium carbonate, sodium bicarbonate, potassium bicarbonate, or the like.
  • Step E Remove the PG 2 group in compound IN-6 to obtain the compound of general formula (I);
  • the reaction is preferably in a suitable organic solvent (the organic solvent may be selected from linear or cyclic ethers (such as tetrahydrofuran or diethyl ether, etc.), N-methylpyrrolidone, N,N-dimethylformamide, N, N-dimethylacetamide, 1,4-dioxane, dimethylsulfoxide and any combination thereof, preferably tetrahydrofuran).
  • the reaction is preferably carried out in the presence of alcohol or water and a base.
  • the alcohol may be, for example, methanol or ethanol.
  • the base may be selected from alkali metal hydroxides, and the alkali metal hydroxide may be selected from lithium hydroxide, sodium hydroxide, and potassium hydroxide.
  • the reaction is preferably carried out at a suitable temperature.
  • the temperature may be room temperature to 80°C, for example 25°C or 40-60°C.
  • the reaction is preferably carried out for a suitable time, such as 2-5 hours or 6-15 hours, such as 2, 3 or 4 hours or overnight.
  • the present invention provides compounds of general formula (IN-6) or pharmaceutically acceptable salts thereof,
  • the compound is preferably selected from:
  • suitable means that the choice of a specific compound or condition will depend on the specific synthetic operation to be performed and the characteristics of the molecule or molecules to be converted, but the choice is within the ability of those skilled in the art . All process/method steps described herein are performed under conditions sufficient to provide the product shown. Those skilled in the art will understand that all reaction conditions (including, for example, reaction solvent, reaction time, reaction temperature, and whether the reaction should be carried out under anhydrous or inert atmosphere, etc.) can be modified to optimize the desired product yield, and these Variations are within the capabilities of those skilled in the art.
  • the examples provide exemplary methods for preparing compounds of general formula (I). Those skilled in the art will understand that other synthetic routes can be used to synthesize compounds of general formula (I). Although specific raw materials and reagents are described and discussed in the examples, other raw materials and reagents can be substituted to provide various derivatives and/or reaction conditions. In addition, with reference to the present disclosure, many of the example compounds prepared by the method can be further modified using conventional chemistry well known to those skilled in the art.
  • the structure of the compound is determined by nuclear magnetic resonance ( 1 H-NMR) or mass spectrometry (MS).
  • the 1 H-NMR shift ( ⁇ ) is given in units of parts per million (ppm). Chemical shifts are given in units of 10 -6 (ppm).
  • the MS was measured using an Agilent (ESI) mass spectrometer.
  • Thin layer chromatography silica gel plate uses Merck aluminum plate (20 ⁇ 20cm), and thin layer chromatography separation and purification uses GF 254 (0.4 ⁇ 0.5mm).
  • the reaction is monitored by thin layer chromatography (TLC) or LC-MS.
  • TLC thin layer chromatography
  • the developing system used is: dichloromethane and methanol system, n-hexane and ethyl acetate system or petroleum ether and ethyl acetate system.
  • the volume ratio of the solvent needs to be adjusted, or triethylamine or the like needs to be further added to separate and purify the product.
  • microwave reaction Initiator+ 400W, RT ⁇ 300 °C microwave reactor.
  • Column chromatography generally uses 200-300 mesh silica gel as a carrier.
  • the eluent system includes: dichloromethane and methanol system, n-hexane and ethyl acetate system, the volume ratio of the solvent is adjusted according to the polarity of the compound, and a small amount of triethylamine can also be added for adjustment.
  • reaction temperature of the examples is room temperature (20°C to 30°C).
  • the reagents used in the present invention were purchased from Acros Organics, Aldrich Chemical Company, Shanghai Tebo Chemical Technology Co., Ltd. and so on.
  • T1-a 2-(trifluoromethoxy)benzaldehyde (150g, 788.98mmol) in a mixed solvent of ethanol (1000mL) and water (1000mL), add hydroxylamine hydrochloride (65.79g, 946.77) with mechanical stirring mmol), a white solid precipitated, and continued to add 1M aqueous sodium hydroxide solution (789 mL, 788.98 mmol).
  • reaction was reacted at 25°C for 2 hours, 1M hydrochloric acid (2000 mL) was added to the mixture to adjust the pH of the system to about 5, directly filtered with suction, and dried overnight at 50°C to obtain compound (T1-b) (150g, collected Rate: 92%).
  • Step 3 Preparation of 5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazole-4-carboxylic acid methyl ester (T1-d)
  • Step 5 Preparation of 4-(chloromethyl)-5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazole (T1-f)
  • Benzotriazole (59.65g, 500.74mmol) was dissolved in dichloromethane, the reaction was stirred at -5 °C, dichlorosulfoxide (59.65g, 501.39mmol) was added dropwise, after stirring at room temperature for half an hour, the compound was added A dichloromethane solution (500 mL) of (T1-e) (100 g, 334.17 mmol) was reacted at room temperature for 6 hours. The reaction solution was filtered with suction, and the filtrate was spin-dried to obtain compound (T1-f) (106 g, yield: 94%).
  • reaction solution was spin-dried, ethyl acetate (1500 mL) and water (1500 mL) were added to the residue.
  • Step 8 4-((((1R,3r,5S)-8-(4-bromothiazol-2-yl)-8-azabicyclo[3.2.1]oct-3-yl)oxy)methan Yl)-5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazole (T1)
  • Example 1 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)benzoic acid (C1)
  • the first step 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)benzoic acid methyl ester (C1-a)
  • compound (T1) (1 g, 1.75 mmol) was dissolved in 1,4-dioxane (20 mL), followed by addition of 4-(4,4,5,5-tetramethyl-1,3, 2-Dioxaborolan-2-yl)benzoic acid methyl ester (532.50mg, 1.93mmol), [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride ( 143.16mg, 195.66 ⁇ mol) and potassium carbonate (483.85mg, 3.50mmol), nitrogen substitution 2-3 times. Reaction at 80°C for 8 hours.
  • the second step 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)benzoic acid (C1)
  • Example 2 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-3-methylbenzoic acid (C2)
  • the first step 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-3-methylbenzoic acid methyl ester (C2-a)
  • compound (T1) 500 mg, 876.55 mmol was dissolved in 1,4-dioxane (20 mL), followed by addition of 3-methyl-4-(4,4,5,5-tetramethyl -1,3,2-dioxaborolan-2-yl)benzoic acid methyl ester (266.25 mg, 964.20 ⁇ mol), [1,1′-bis(diphenylphosphino)ferrocene] Palladium dichloride (71.58 mg, 97.83 ⁇ mol) and potassium carbonate (241.93 mg, 1.75 mmol) were replaced with nitrogen gas 2-3 times. Reaction at 80°C for 8 hours.
  • the second step 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-3-methylbenzoic acid (C2)
  • Example 3 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl]thiazol-4-yl)-3-fluorobenzoic acid (C3)
  • the first step 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-3-fluorobenzoic acid methyl ester (C3-a)
  • compound (T1) (650 mg, 1.14 mmol) was dissolved in 1,4-dioxane (20 mL), followed by addition of 3-fluoro-4-(4,4,5,5-tetramethyl- 1,3,2-Dioxaborolan-2-yl)benzoic acid methyl ester (248.13mg, 1.25mmol), [1,1′-bis(diphenylphosphino)ferrocene]di Palladium chloride (93.06 mg, 127.18 ⁇ mol) and potassium carbonate (314.50 mg, 2.28 mmol) were replaced with nitrogen gas 2-3 times. Reaction at 80°C for 8 hours.
  • the second step 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl]thiazol-4-yl)-3-fluorobenzoic acid (C3)
  • Example 4 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-2-fluorobenzoic acid (C4)
  • the first step 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-2-fluorobenzoic acid methyl ester (C4-a)
  • compound (T1) 400 mg, 701.24 ⁇ mol was dissolved in 1,4-dioxane (20 mL), followed by addition of 2-fluoro-4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)benzoic acid methyl ester (152.70 mg, 771.36 ⁇ mol), [1,1′-bis(diphenylphosphino)ferrocene]di Palladium chloride (57.27 mg, 78.26 ⁇ mol) and potassium carbonate (193.54 mg, 1.40 mmol) were replaced with nitrogen gas 2-3 times. Reaction at 80°C for 8 hours.
  • the second step 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-2-fluorobenzoic acid (C4)
  • Example 5 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-2-(trifluoromethyl)benzoic acid (C5)
  • the first step 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-2-(trifluoromethyl)benzoic acid methyl ester (C5-a)
  • compound (T1) (0.4 g, 701.24 ⁇ mol) was dissolved in 1,4-dioxane (20 mL), followed by addition of 4-(4,4,5,5-tetramethyl-1,3 , 2-dioxaborolane-2-yl)-2-(trifluoromethyl)benzoic acid methyl ester (277.78 mg, 841.48 ⁇ mol), [1,1′-bis(diphenylphosphino ) Ferrocene] palladium dichloride (57.27 mg, 70.12 ⁇ mol) and potassium carbonate (193.83 mg, 1.40 mmol), nitrogen substitution 2-3 times. Reaction at 80°C for 8 hours.
  • the second step 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-2-(trifluoromethyl)benzoic acid (C5)
  • the first step 5-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Preparation of methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)picolinic acid methyl ester (C6-a)
  • compound (T1) 50 mg, 87.65 ⁇ mol was dissolved in 1,4-dioxane (20 mL), followed by addition of 5-(4,4,5,5-tetramethyl-1,3,2 -Dioxaborolan-2-yl)picolinic acid methyl ester (27.67mg, 105.19 ⁇ mol), [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride (7.16 mg, 8.77 ⁇ mol) and potassium carbonate (12.11 mg, 87.65 ⁇ mol), nitrogen substitution 2-3 times. The reaction was carried out at 80°C for 8 hours.
  • reaction solution was filtered with celite, and the filtrate was washed with ethyl acetate (100 mL ⁇ 3).
  • the second step 5-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)picolinic acid (C6)
  • Emax and Emin are the upper and lower asymptotic estimates of the fitted curve, respectively, x is the log concentration of the compound, and Hillslope is the slope of the curve.
  • Emax represents the maximum activation effect value of the tested compound of the present invention
  • Emax' represents the maximum activation effect value of CDCA, both of which are calculated by the formula shown above.
  • Table 1 shows that the compounds tested have a lower EC 50 (0.002-0.133 ⁇ M) relative to an EC 50 of chenodeoxycholic acid (CDCA) value of 4.43 ⁇ M, indicating that the compounds of the present invention have a greater effect on FXR Good activation activity.
  • Human embryonic kidney cells HEK293 were cultured in DMEM medium containing 10% FBS. Co-transfect the plasmid to make it highly express FXR and human BSEP luciferase reporter gene. The transfected cells were digested, resuspended, counted, and then seeded in multi-well plates. Add 10 ⁇ L of different concentrations of the test compound to the multi-well plate to make the final concentration of 64 ⁇ M, 16 ⁇ M, 4 ⁇ M, 1 ⁇ M, 0.25 ⁇ M, 0.0625 ⁇ M, 0.0156 ⁇ M, 0.0039 ⁇ M, 0.000975 ⁇ M, 0.000244 ⁇ M, 0 ⁇ M, the final concentration of DMSO is 0.5%.
  • test compound 50 ⁇ L was mixed with various liver microsomes (100 ⁇ L). After pre-incubation at 37°C for 5 minutes, NADPH (50 ⁇ L) was added and incubated for 0, 30, and 60 minutes. The test compound, NADPH and liver microsome enzymes The incubation concentrations are 1 ⁇ M, 1mM and 0.5mg/mL, respectively. After adding ice acetonitrile (200 ⁇ L) to terminate the reaction, an appropriate volume of internal standard (rivaroxaban) was added, and the supernatant was collected by vortexing and centrifugation for detection.
  • rivaroxaban internal standard
  • LC-MS/MS mass spectrometer
  • liquid phase is Shimadzu LC-30AD system.
  • the chromatographic column is Hypersil C18, 1.9 ⁇ m particle size, 50 ⁇ 2.1mm; mobile phase A is water + 0.1% formic acid, phase B is acetonitrile; flow rate is 0.55mL/min, column temperature is 40 °C.
  • the ion source is the positive ion mode of the ESI source, and the scanning mode is multiple reaction monitoring (MRM).
  • the rate constant is obtained by plotting "Ln (% of drug residue)" versus "incubation time”, thereby calculating the half-life and liver clearance of the drug. Evaluate the metabolic stability of the drug in liver microsomes.
  • the compounds of the present invention were administered to male SD rats by intravenous (IV) and intragastric (PO), respectively, to investigate the pharmacokinetic characteristics.
  • IV and PO are 1 mg/kg and 5 mg/kg, respectively, the IV administration vehicle system is 5% DMSO: 5% Solutol: 90% saline, and the PO administration vehicle system is 0.5% MC (methyl cellulose ).
  • IV and PO administration blood was collected at different time points. The blood was anticoagulated with K2-EDTA. After centrifugation, plasma samples were obtained and stored at -80°C. Plasma samples were treated with precipitated proteins and analyzed by LC-MS/MS.
  • WinNonlin 6.3 software was used to calculate the pharmacokinetic parameters using a non-compartmental model. The results are shown in Tables 6 and 7.
  • Table 6 shows that the compounds C3 and C5 of the present invention administered IV at a dose of 1 mg/kg have excellent drug exposure in rats.
  • Table 7 shows that the compounds C3 and C5 of the present invention administered by PO at a dose of 5 mg/kg have excellent drug exposure and bioavailability in rats.
  • the compounds C3 and C5 of the present invention have excellent plasma drug exposure and oral bioavailability in rats by IV and PO administration.
  • Other compounds of the present invention also have better AUC last value, C max value and bioavailability, and have better pharmacokinetic properties in rats.
  • mice Eight to ten-week-old male C57 mice (purchased from Viton Lihua) were used. Animals were randomly divided into 5 groups according to body weight before modeling, which were normal group (group 1), model group (group 2), and C3 compound. Administration group (30mg/kg). The normal group was given normal feed, and the model group and C3 compound-administered group were fed with MCD feed (methionine choline-deficient feed, Research Diets). Simultaneously with the modeling, the C3 compound group was administered by intragastric administration once a day for 28 consecutive days. The normal group and the model group were given 0.5% MC (methyl cellulose). See Table 8 for details.
  • Drug Efficacy Index Animals were dissected after administration on day 28, and liver HE staining was performed for pathological scoring (the scoring criteria refer to "Chinese Non-alcoholic Fatty Liver Disease Diagnosis and Treatment Guide”).
  • Pharmacokinetic indicators 0.5h, 1h, 4h after administration on day 28, blood, liver and small intestine tissue samples were taken from the animals, and the drug concentration was detected using LC-MS/MS.
  • Compound C3 can inhibit liver fat lesions induced by MCD feed, and has a significant improvement relative to the model group.
  • the distribution of compound C3 in the liver is higher than that in plasma, indicating that it is effectively enriched at the target site.

Abstract

The present invention relates to an isoxazole derivative, a preparation method therefor and a use thereof. Specifically, provided are a farnesoid X receptor (FXR) agonist compound and stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N-oxides, chemically protected forms and prodrugs thereof. Further provided are a method for preparing the described compound, intermediates, pharmaceutical compositions and kits containing the compound, and a use thereof in treating diseases or conditions mediated by FXR.

Description

异噁唑衍生物及其制备方法和用途Isoxazole derivative and its preparation method and use 发明领域Field of invention
本发明一般涉及用于治疗由类法尼醇X受体(FXR)介导的疾病或病症的化合物,更具体地涉及FXR激动剂化合物,以及其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物以及其化学保护的形式和前药。本发明还涉及所述化合物的制备方法、中间体、包含所述化合物的药物组合物和药盒以及它们的治疗用途。The present invention relates generally to compounds for the treatment of diseases or conditions mediated by farnesoid X receptor (FXR), and more specifically to FXR agonist compounds, as well as their stereoisomers, tautomers, polymorphs Crystal forms, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N-oxides, and chemically protected forms and prodrugs thereof. The invention also relates to preparation methods, intermediates, pharmaceutical compositions and kits containing the compounds, and their therapeutic uses.
发明背景Background of the invention
类法尼醇X受体(FXR,NR1H4)在肝、包括食道、胃、十二指肠、小肠、结肠在内的整个胃肠道、肾和肾上腺中表达(Kuipers,F.等,The Farnesoid X Receptor(FXR)as Modulator of Bile Acid Metabolism,Rev.Endocrine Metab.Disorders,2004,5:319-326)。FXR是已知作为核受体的配体活化的转录因子的一员。胆汁酸如鹅脱氧胆酸(CDCA)或者其牛磺酸或甘氨酸酰胺偶联物是FXR的内源性配体。胆汁酸与FXR结合后激活FXR,通过与视黄醇类X受体(RXR)的异二聚体复合物来控制多种基因的表达,包括在肝脏和循环***中参与胆汁酸、胆固醇、三酸甘油酯、脂蛋白动态平衡的基因表达(Kalaany,N.Y.;Mangelsdorf,D.J.;LXRS and FXR:the yin and yang of cholesterol and fat metabolism,Annu.Rev.Physiol.,2006,68,159-191;Calkin,A.C.;Tontonoz,P.;Transcriptional integration of metabolism by the nuclear sterol-activated receptors LXR and FXR,Nat.Rev.Mol.Cell Biol.,2012,13,213-224)。FXR似乎还通过上调成纤维细胞生长因子15(啮齿动物)或成纤维细胞生长因子19(猴、人)参与旁分泌和内分泌信号传导(T.Inagaki等,Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis.Cell Metab.,2005,2(4),217-225)。Farnesoid X receptor (FXR, NR1H4) is expressed in the liver, the entire gastrointestinal tract including the esophagus, stomach, duodenum, small intestine, and colon (Kuipers, F. et al., The Farnesoid X Receptor (FXR) as Modulator of Bile Acid Metabolism, Rev. Endocrine Metab. Disorders, 2004, 5: 319-326). FXR is a member of a transcription factor known to be a ligand-activated nuclear receptor. Bile acids such as chenodeoxycholic acid (CDCA) or its taurine or glycine amide conjugates are endogenous ligands of FXR. The bile acid binds to FXR and activates FXR. It controls the expression of various genes through heterodimer complex with retinol X receptor (RXR), including the involvement of bile acid, cholesterol, triglyceride in liver and circulatory system. Gene expression of acid glyceride and lipoprotein homeostasis (Kalaany, NY; Mangelsdorf, DJ; LXRS and FXR: the yin and yang of cholesterol and fat metabolism, Annu. Rev. Physiol., 2006, 68, 159-191; Calkin , AC; Tontonoz, P.; Transcriptional integration of metabolism by the nuclear sterol-activated receptors LXR and FXR, Nat. Rev. Mol. Cell Biol., 2012, 13, 213-224). FXR also appears to be involved in paracrine and endocrine signaling by up-regulating fibroblast growth factor 15 (rodent) or fibroblast growth factor 19 (monkey, human) (T. Inagaki et al., Fibroblast growth factor 15 functions) aserohepatic signaling Regulated acid homeostasis. Cell Metab., 2005, 2(4), 217-225).
胆汁酸是两亲性分子,它们形成胶束并将膳食中的脂质乳化。如果胆汁酸浓度过高也会产生细胞毒性,因此生理上有严格控制胆汁酸浓度的机制。FXR在控制胆汁酸在体内保持稳定状态中起着关键作用(Makishima,M.;Nuclear Receptors as Targets for Drug Development:Regulation of Cholesterol and Bile Acid Metabolism by Nuclear Receptors,J.Pharmacol.Sci.,2005,97:177-183)。Bile acids are amphiphilic molecules that form micelles and emulsify the lipids in the diet. If the bile acid concentration is too high, cytotoxicity will also occur, so physiologically there is a mechanism to strictly control the bile acid concentration. FXR plays a key role in controlling the bile acid to maintain a stable state in the body (Makishima, M.; Nuclear Receptors as Targets for Drug Development: Regulation of Cholesterol and Bile Acid Metabolism by Nuclear Receptors, J. Pharmacol. Sci., 2005, 97 : 177-183).
此外,FXR还被证明调节超出代谢的复杂生物流程,如肝再生或肠屏障的完整性。FXR还对肠和肝脏的免疫***有控制,有一定的抗炎作用(Modica,S.;Gadaleta,R.M.;Moschetta,A.;Deciphering the nuclear bile acid receptor FXR paradigm,Nucl.Recept.Signal.,2010,8,e005)。In addition, FXR has been shown to regulate complex biological processes beyond metabolism, such as liver regeneration or the integrity of the intestinal barrier. FXR also controls the immune system of the intestine and liver and has a certain anti-inflammatory effect (Modica, S.; Gadaleta, RM; Moschetta, A.; Deciphering the nuclear acceptor FXR paradigm, Nucl. Recept. Signal., 2010 , 8, e005).
奥贝胆酸(Obeticholic Acid,6-Et CDCA)是一种比内源性配体CDCA活性更高的FXR受体激动剂,在非酒精性脂肪性肝病(NAFLD)的IIa期临床研究中显示出对胰岛素敏感性有显著改善以及其他代谢方面的有益作用(Mudaliar,S.;Henry,R.R.;Sanyal,A.J.等,Efficacy and safety of the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty liver disease,Gastroenterology,2013,145,574-582)。奥贝胆酸的IIb期研究显示72周的治疗对非酒精性肝炎(NASH)的组织病理学的改进也有益。在原发性胆汁性肝硬化(PBC)的III期研究中,患者的肝功能损害得到改善(Nevens,F.,Andreone,P.,Mazzella,G.等,The first primary biliary cirrhosis(PBC)phase 3 trial in two decades-an international study of the FXR agonist obeticholic acid in PBC patients,J.Hepatol.,2014,60,S525-S526)。Obeticholic acid (6-Et CDCA) is an FXR receptor agonist that is more active than the endogenous ligand CDCA and has been shown in a phase IIa clinical study of non-alcoholic fatty liver disease (NAFLD) Significant improvement in insulin sensitivity and other beneficial effects on metabolism disease, Gastroenterology, 2013, 145, 574-582). A phase IIb study of obeticholic acid showed that 72-week treatment is also beneficial for the improvement of histopathology of non-alcoholic hepatitis (NASH). In a phase III study of primary biliary cirrhosis (PBC), the patient's liver function impairment was improved (Nevens, F., Andreone, P., Mazzella, G., etc., The first primary biliary cirrhosis (PBC) phase 3 trial in two colleges-an international study of the FXR agonist obticholic acid in PBC patients, J. Hepatol., 2014, 60, S525-S526).
WO2012087519中公开了一种FXR的激动剂或部分激动剂用于治疗由FXR介导的病症的方法。然而,现有技术中公开的FXR激动剂化合物在药效动力学或药代动力学性质方面仍存在缺陷。WO2012087519 discloses an agonist or partial agonist of FXR for the treatment of conditions mediated by FXR. However, the FXR agonist compounds disclosed in the prior art still have defects in pharmacodynamics or pharmacokinetic properties.
发明概述Summary of the invention
本发明概括地涉及通式(I)的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,The present invention relates generally to compounds of general formula (I) or their stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs,
Figure PCTCN2019121534-appb-000001
Figure PCTCN2019121534-appb-000001
Figure PCTCN2019121534-appb-000002
Figure PCTCN2019121534-appb-000002
其中:among them:
X为CH或N;并且X is CH or N; and
R选自氢、卤素、C 1-6烷基和C 1-6卤代烷基。 R is selected from hydrogen, halogen, C 1-6 alkyl and C 1-6 haloalkyl.
本发明的另一方面是药物组合物,其包含所述通式(I)的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,以及一种或多种药学可接受的载体。所述药物组合物还可包含适于预防或治疗由FXR介导的疾病或病症的一种或多种其他治疗剂。Another aspect of the invention is a pharmaceutical composition comprising the compound of the general formula (I) or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceuticals Acceptable salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, and one or more pharmaceutically acceptable carriers. The pharmaceutical composition may further comprise one or more other therapeutic agents suitable for preventing or treating diseases or conditions mediated by FXR.
本发明还包括预防或治疗由FXR介导的疾病或病症的方法,所述方法包括向有此需要的个体给药治疗有效量的所述通式(I)的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药或者所述药物组合物。The present invention also includes a method of preventing or treating a disease or condition mediated by FXR, the method comprising administering to a subject in need thereof a therapeutically effective amount of the compound of general formula (I) or its stereoisomer, Tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, or the pharmaceutical composition .
本发明还包括用于预防或治疗由FXR介导的疾病或病症的药盒,其包括:The present invention also includes a kit for preventing or treating a disease or condition mediated by FXR, which includes:
a)第一容器,其包含作为第一治疗剂的至少一种所述通式(I)的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药或者作为第一药物组合物的所述药物组合物;a) a first container, which contains as a first therapeutic agent at least one compound of the general formula (I) or its stereoisomers, tautomers, polymorphs, solvates (eg hydrated) Substances), pharmaceutically acceptable salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, or the pharmaceutical composition as the first pharmaceutical composition;
b)任选存在的第二容器,其包含作为第二治疗剂的至少一种其他治疗剂,或者作为第二药物组合物的包含所述其他治疗剂的药物组合物;和b) an optional second container containing at least one other therapeutic agent as a second therapeutic agent, or a pharmaceutical composition containing the other therapeutic agent as a second pharmaceutical composition; and
c)任选存在的包装说明书。c) optional package inserts.
本发明还包括所述通式(I)的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药或者所述药物组合物,其用于预防或治疗由FXR介导的疾病或病症。The present invention also includes the compound of the general formula (I) or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites , N-oxide, its chemically protected form or prodrug, or the pharmaceutical composition, which is used to prevent or treat a disease or condition mediated by FXR.
本发明还包括所述通式(I)的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药或者所述药物组合物在制备用于预防或治疗由FXR介导的疾病或病症的药物中的用途。The present invention also includes the compound of the general formula (I) or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites , N-oxide, its chemically protected form or prodrug, or the use of the pharmaceutical composition in the preparation of a medicament for the prevention or treatment of a disease or disorder mediated by FXR.
本发明还包括制备本发明的化合物的方法和相应中间体。The invention also includes methods and corresponding intermediates for preparing the compounds of the invention.
本发明的通式(I)的化合物具有优良的体内或体外药效动力学或药代动力学性质,显示出良好的FXR激活活性和激活作用以及优良的肝微粒体稳定性,因而具有良好的药物活性和代谢优势。The compound of the general formula (I) of the present invention has excellent in vivo or in vitro pharmacodynamics or pharmacokinetic properties, shows good FXR activation activity and activation, and excellent liver microsomal stability, and thus has good Drug activity and metabolic advantages.
附图简要说明Brief description of the drawings
图1显示在小鼠脂肪肝模型中给药化合物C3的效果,其中a表示与对照组相比p≤0.05;b表示与模型组相比p≤0.05。Figure 1 shows the effect of administering compound C3 in a mouse fatty liver model, where a represents p≤0.05 compared to the control group; b represents p≤0.05 compared to the model group.
发明详细描述Detailed description of the invention
现详细描述本发明的某些实施方案,其实例在随附结构和分子式中说明。虽然结合列举的实施方案来描述本发明,但是应理解本发明并不限于那些实施方案。相反,本发明意在涵盖可包括在本发明的由权利要求书限定的范围内的所有替代方案、修改和等同。本领域技术人员会认识到,与本文中所述的那些相似或等同的许多方法和材料可用于实施本发明。本发明绝不限于所述的方法和材料。在所引的文献、专利及相似的材料中的一者或多者与本申请(包括但不限于定义的术语、术语用法、所述的技术等)不同或矛盾的情况下,以本申请为准。Some embodiments of the present invention will now be described in detail, examples of which are illustrated in the accompanying structure and molecular formula. Although the invention has been described in conjunction with the enumerated embodiments, it should be understood that the invention is not limited to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents that may be included within the scope of the invention as defined by the claims. Those skilled in the art will recognize that many methods and materials similar or equivalent to those described herein can be used in the practice of the present invention. The invention is in no way limited to the methods and materials described. In the case where one or more of the cited documents, patents, and similar materials are different or contradictory to this application (including but not limited to defined terms, term usage, described techniques, etc.), this application is regarded as quasi.
定义definition
除非在下文中另有定义,本文中所用的所有技术术语和科学术语的含义意图与本领域技术人员通常所理解的相同。Unless otherwise defined below, all technical and scientific terms used herein are intended to have the same meaning as commonly understood by those skilled in the art.
术语“包括”、“包含”、“具有”、“含有”或“涉及”及其在本文中的其他变体形式是包含性的或开放式的,且不排除其他未列举的元素或方法步骤。The terms "including", "comprising", "having", "containing" or "involving" and other variations thereof herein are inclusive or open-ended and do not exclude other unlisted elements or method steps .
术语“烷基”用于本文中是指具有1至6个碳原子(C 1-6)的饱和直链或支链烃基,其中所述烷基可任选地被一个或多个(例如1个、2个、3个或4个)适合的取代基取代。在一些实施方案中,烷基具有1至6个碳原子(C 1-6)。在另一些实施方案中,烷基具有1至4个碳原子(C 1-4),特别是1至3个碳原子(C 1-3)或者1至2个碳原子(C 1-2)。烷基的实例包括但不限于:甲基(Me)、乙基(Et)、1-丙基(n-Pr)、2-丙基(i-Pr或异丙基)、1-丁基(n-Bu或正丁基)、2-甲基-1-丙基(i-Bu或异丁基)、2-丁基(s-Bu或仲丁基)、2-甲基-2-丙基(t-Bu或叔丁基)、1-戊基(正戊基)、2-戊基、3-戊基、2-甲基-2-丁基、3-甲基-2-丁基、3-甲基-1-丁基、2-甲基-1-丁基、1-己基、2-己基、3-己基、2-甲基-2-戊基(-C(CH 3) 2CH 2CH 2CH 3)、3-甲基-2-戊基、4-甲基-2-戊基、3-甲基-3-戊基、2-甲基-3-戊基、2,3-二甲基-2-丁基、3,3-二甲基-2- 丁基、1-庚基、1-辛基等。 The term "alkyl" as used herein refers to a saturated linear or branched hydrocarbon group having 1 to 6 carbon atoms (C 1-6 ), wherein the alkyl group may be optionally substituted by one or more (eg, 1 , 2, 3, or 4) suitable substituents. In some embodiments, the alkyl group has 1 to 6 carbon atoms (C 1-6 ). In other embodiments, the alkyl group has 1 to 4 carbon atoms (C 1-4 ), especially 1 to 3 carbon atoms (C 1-3 ) or 1 to 2 carbon atoms (C 1-2 ) . Examples of alkyl groups include, but are not limited to: methyl (Me), ethyl (Et), 1-propyl (n-Pr), 2-propyl (i-Pr or isopropyl), 1-butyl ( n-Bu or n-butyl), 2-methyl-1-propyl (i-Bu or isobutyl), 2-butyl (s-Bu or sec-butyl), 2-methyl-2-propyl (T-Bu or tert-butyl), 1-pentyl (n-pentyl), 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-2-butyl , 3-methyl-1-butyl, 2-methyl-1-butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl (-C(CH 3 ) 2 CH 2 CH 2 CH 3 ), 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2-methyl-3-pentyl, 2, 3-dimethyl-2-butyl, 3,3-dimethyl-2-butyl, 1-heptyl, 1-octyl, etc.
本文中使用的术语“卤代”或“卤素”包括F、Cl、Br或I。“卤代”包括但不限于单取代、二取代或三取代,而且,用于取代的卤原子可以相同或不同。本文中使用的术语“卤代烷基”包括氟代烷基、氯代烷基、溴代烷基或碘代烷基。The term "halo" or "halogen" as used herein includes F, Cl, Br or I. "Halo" includes but is not limited to mono-, di- or tri-substitution, and the halogen atoms used for substitution may be the same or different. The term "haloalkyl" as used herein includes fluoroalkyl, chloroalkyl, bromoalkyl, or iodoalkyl.
当取代基的键显示为穿过环中连接两个原子的键时,则这样的取代基可键连至该可取代的环中的任一成环原子。When a bond of a substituent is shown to pass through a bond connecting two atoms in the ring, then such substituent may be bonded to any ring-forming atom in the substitutable ring.
术语“手性”是指具有镜像对的不可重叠性的分子,而术语“非手性”是指可在它们的镜像对上重叠的分子。The term "chiral" refers to molecules that have non-superimposability of mirror image pairs, and the term "achiral" refers to molecules that can overlap on their mirror image pairs.
术语“立体异构体”是指具有相同的化学组成但原子或基团的空间排列不同的化合物。The term "stereoisomer" refers to compounds that have the same chemical composition but differ in the spatial arrangement of atoms or groups.
“非对映异构体”是指具有两个或多个手性中心并且其分子彼此不互为镜像的立体异构体。非对映异构体具有不同的物理性质,例如熔点、沸点、光谱性质和反应性。非对映异构体的混合物可通过高分辨率的分析方法例如电泳法和色谱法进行分离。"Diastereomer" refers to a stereoisomer that has two or more chiral centers and whose molecules are not mirror images of each other. Diastereomers have different physical properties, such as melting point, boiling point, spectral properties, and reactivity. The mixture of diastereomers can be separated by high-resolution analytical methods such as electrophoresis and chromatography.
“对映异构体”是指化合物的彼此呈不可重叠的镜像的两种立体异构体。"Enantiomer" refers to two stereoisomers of a compound that are non-overlapping mirror images of each other.
本文中所用的立体化学定义和规则一般遵循S.P.Parker,Ed.,McGraw-Hill Dictionary of Chemical Terms(1984)McGraw-Hill Book Company,New York;和Eliel,E.和Wilen,S.,“Stereochemistry of Organic Compounds”,John Wiley & Sons,Inc.,New York,1994。本发明的化合物可包含非对称的或手性的中心,因此以不同的立体异构体形式存在。本发明的化合物的所有立体异构体形式,包括但不限于其非对映异构体、对映异构体和阻转异构体以及它们的混合物例如外消旋混合物,意在构成本发明的一部分。许多有机化合物以旋光形式存在,即,它们具有使平面偏振光的平面旋转的能力。在表述旋光化合物时,前缀D和L,或者R和S,用来表示分子的手性中心的绝对构型。前缀d和l或者(+)和(-)用来指示化合物使平面偏振光旋转的符号,其中(-)或l是指化合物是左旋的。具有前缀(+)或d的化合物是右旋的。对于特定的化学结构,这些立体异构体是相同的,只是它们彼此互为镜像。特定的立体异构体还可称为对映异构体,并且这样的异构体的混合物常称为对映异构体混合物。对映异构体的50∶50混合物称为外消旋混合物或外消旋物,其可在化学反应或过程没有立体选择性或立体专一性的情况下出现。术语“外消旋混合物”和“外消旋物”是指两种对映异构体的等摩尔混合物,不具有旋光性。在一方面,本发明的立体异构体可以占主导的形式存在,例如,大于50%ee(对映体过量),大于80%ee,大于90%ee,大于95%ee,或者大于99%ee。The stereochemical definitions and rules used in this article generally follow SPParker, Ed., McGraw-Hill Dictionary of Chemicals (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., "Stereochemistry" of Organic Compounds”, John Wiley & Sons, Inc., New York, 1994. The compounds of the present invention may contain asymmetric or chiral centers and therefore exist in different stereoisomeric forms. All stereoisomeric forms of the compounds of the invention, including but not limited to their diastereomers, enantiomers and atropisomers, and mixtures thereof, such as racemic mixtures, are intended to constitute the invention a part of. Many organic compounds exist in optically active forms, that is, they have the ability to rotate the plane of plane polarized light. When expressing optically active compounds, the prefixes D and L, or R and S, are used to indicate the absolute configuration of the chiral center of the molecule. The prefixes d and l or (+) and (-) are used to indicate the sign that the compound rotates plane polarized light, where (-) or l means that the compound is levorotatory. Compounds with the prefix (+) or d are right-handed. For a specific chemical structure, these stereoisomers are the same, but they are mirror images of each other. A specific stereoisomer can also be called an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is called a racemic mixture or a racemate, which can occur in a chemical reaction or process without stereoselectivity or stereospecificity. The terms "racemic mixture" and "racemate" refer to an equimolar mixture of two enantiomers, which is not optically active. In one aspect, the stereoisomers of the invention may exist in a predominant form, for example, greater than 50%ee (enantiomeric excess), greater than 80%ee, greater than 90%ee, greater than 95%ee, or greater than 99% ee.
在用于制备本发明的化合物的方法产生立体异构体的混合物的情况中,通过常规技术例如制备色谱可以分离这些异构体。所述化合物可以制备成外消旋的形式,或者,通过对映体选择性合成或者通过拆分可以制备单一的对映异构体。例如,通过标准技术,例如通过与旋光性的酸如(-)-二对甲苯酰基-d-酒石酸和/或(+)-二对甲苯酰基-l-酒石酸形成盐来形成非对映异构体对,然后进行分步结晶和游离碱再生,可以将所述化合物拆分成它们的对映异构体成分。通过形成非对映的酯或酰胺,然后进行色谱纯化和除去手性助剂,也可以拆分所述化合物。或者,使用手性HPLC柱可以拆分所述化合物。In the case where the method for preparing the compound of the present invention produces a mixture of stereoisomers, these isomers can be separated by conventional techniques such as preparative chromatography. The compound can be prepared in racemic form, or a single enantiomer can be prepared by selective synthesis of enantiomers or by resolution. For example, diastereoisomers are formed by standard techniques, for example by forming salts with optically active acids such as (-)-di-p-toluoyl-d-tartaric acid and/or (+)-di-p-toluoyl-l-tartaric acid Pairs, followed by step crystallization and free base regeneration, the compounds can be resolved into their enantiomeric components. The compounds can also be resolved by forming diastereomeric esters or amides, followed by chromatographic purification and removal of chiral auxiliaries. Alternatively, the compound can be resolved using a chiral HPLC column.
术语“互变异构体”或“互变异构体形式”是指可通过低能垒互相转化的能量不同的结构异构体。例如,质子互变异构体(也称为质子转移互变异构体)包括通过质子迁移互相转化,例如酮-烯醇和亚胺-烯胺异构化。价键互变异构体包括通过一些成键电子的重组互相转化。The term "tautomer" or "tautomeric form" refers to structural isomers of different energies that can be interconverted by a low energy barrier. For example, proton tautomers (also known as proton transfer tautomers) include interconversion through proton migration, such as keto-enol and imine-enamine isomerization. Valence tautomers include interconversion through the recombination of some bonding electrons.
本发明涵盖通式(I)的化合物的所有可能的结晶形式或多晶型物,其可为单一多晶型物或多于一种多晶型物的任意比例的混合物。The present invention encompasses all possible crystalline forms or polymorphs of the compound of general formula (I), which may be a single polymorph or a mixture of more than one polymorph in any ratio.
应当理解,本发明的某些化合物可以游离形式存在用于治疗,或适当时,以其药学可接受的衍生物形式存在。根据在本发明中,药学可接受的衍生物包括但不限于:药学可接受的盐、酯、溶剂合物、代谢物、N-氧化物以及化学保护的形式和前药,在将它们向有此需要的个体给药后,能够直接或间接提供本发明的化合物或者其代谢物或残余物。It should be understood that certain compounds of the invention may exist in free form for therapy, or where appropriate, in the form of pharmaceutically acceptable derivatives. According to the present invention, pharmaceutically acceptable derivatives include, but are not limited to: pharmaceutically acceptable salts, esters, solvates, metabolites, N-oxides, and chemically protected forms and prodrugs. After administration to an individual in need, the compound of the present invention or its metabolite or residue can be provided directly or indirectly.
因此,当在本文中提及“通式(I)的化合物”、“本发明的化合物”或“本发明的通式(I)的化合物”时,也意在涵盖所述通式(I)的化合物的溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物以及其化学保护的形式和前药。Therefore, when referring to "a compound of general formula (I)", "a compound of the present invention" or "a compound of general formula (I) of the present invention" herein, it is also intended to cover the general formula (I) Solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N-oxides, and chemically protected forms and prodrugs of the compounds of
术语“药学可接受的盐”用于本文中是指本发明的化合物的药学可接受的有机或无机盐。示例性的盐包括但不限于异烟酸盐、水杨酸盐、酸式柠檬酸盐、油酸盐、鞣酸盐、泛酸盐、葡糖酸盐、葡糖醛酸盐、糖质酸盐和扑酸盐(即1,1’-亚甲基-双(2-羟基-3-萘甲酸盐))等。药学可接受的盐可包括反荷离子的另一种分子的包合。所述反荷离子可以是使母体化合物上的电荷稳定的任何有机或无机离子。此外,药学可接受的盐在其结构中可具有多于一个带电荷的原子。多个带电荷的原子为药学可接受的盐的部分的情况可具有多个反荷离子。因此,药学可接受的盐可具有一个或多个带电荷的原 子和/或一个或多个反荷离子。The term "pharmaceutically acceptable salts" as used herein refers to pharmaceutically acceptable organic or inorganic salts of the compounds of the present invention. Exemplary salts include but are not limited to isonicotinate, salicylate, acid citrate, oleate, tannate, pantothenate, gluconate, glucuronate, glucuronic acid Salts and parabens (ie 1,1'-methylene-bis(2-hydroxy-3-naphthoate)), etc. The pharmaceutically acceptable salt may include the inclusion of another molecule of counter ion. The counter ion can be any organic or inorganic ion that stabilizes the charge on the parent compound. In addition, pharmaceutically acceptable salts may have more than one charged atom in their structure. Where multiple charged atoms are part of a pharmaceutically acceptable salt, there may be multiple counter ions. Thus, a pharmaceutically acceptable salt can have one or more charged atoms and/or one or more counter ions.
若本发明的化合物是碱,期望的药学可接受的盐可通过本领域中可利用的任何适合的方法制备,例如,用无机酸或者用有机酸例如水杨酸、吡喃糖苷基酸例如葡糖醛酸或半乳糖醛酸、α-羟基酸氨基酸、芳香酸等处理游离的碱。If the compound of the present invention is a base, the desired pharmaceutically acceptable salt can be prepared by any suitable method available in the art, for example, with an inorganic acid or with an organic acid such as salicylic acid, pyranosidic acid such as glucose The free base is treated with uronic acid or galacturonic acid, α-hydroxy acid amino acid, aromatic acid, etc.
若本发明的化合物是酸,期望的药学可接受的盐可通过任何适合的方法制备,例如,用无机碱或有机碱例如胺(伯胺、仲胺或叔胺)、碱金属氢氧化物或碱土金属氢氧化物等处理游离的酸。适合的盐的示例性实例包括但不限于,得自氨基酸以及环胺的有机盐,以及无机盐。If the compound of the present invention is an acid, the desired pharmaceutically acceptable salt can be prepared by any suitable method, for example, using an inorganic or organic base such as an amine (primary, secondary, or tertiary amine), alkali metal hydroxide, or Alkaline earth metal hydroxides, etc. treat free acids. Illustrative examples of suitable salts include, but are not limited to, organic salts derived from amino acids and cyclic amines, and inorganic salts.
术语“药学可接受的”是指物质或组合物必须与构成制剂的其他组分和/或用其治疗的哺乳动物在化学和/或毒理学上相容。The term "pharmaceutically acceptable" means that the substance or composition must be chemically and/or toxicologically compatible with the other components making up the formulation and/or the mammal treated with it.
本文所使用的术语“酯”意指衍生自通式(I)化合物的酯,包括生理上可水解的酯,其可在生理条件下水解以释放游离酸或醇形式的本发明的通式(I)化合物。本发明的通式(I)化合物本身也可以是酯。As used herein, the term "ester" means an ester derived from a compound of general formula (I), including physiologically hydrolyzable esters, which can be hydrolyzed under physiological conditions to release the free acid or alcohol form of the general formula of the present invention ( I) Compound. The compound of the general formula (I) of the present invention may itself be an ester.
本发明的化合物可以溶剂合物(如水合物)的形式存在,其中本发明的化合物包含作为所述化合物晶格的结构要素的极性溶剂,特别是例如水、甲醇或乙醇。极性溶剂特别是水的量可以化学计量比或非化学计量比存在。The compound of the present invention may exist in the form of a solvate (such as a hydrate), wherein the compound of the present invention contains a polar solvent as a structural element of the crystal lattice of the compound, particularly, for example, water, methanol, or ethanol. The amount of polar solvents, especially water, can be present in stoichiometric or non-stoichiometric ratios.
“代谢物”是特定的化合物或其盐通过体内代谢产生的产物。化合物的代谢物可利用本领域已知的常规技术进行鉴定并且可利用诸如本文中所述的那些试验测定它们的活性。这样的产物可由例如被给药的化合物的氧化、还原、水解、酰胺化、脱酰胺化、酯化、酶解等产生。因此,本发明包括本发明的化合物的代谢物,包括通过包括使本发明的通式(I)的化合物与哺乳动物接触足以产生其代谢物的时间段的方法产生的化合物。A "metabolite" is a product produced by metabolism of a specific compound or its salt in the body. Metabolites of compounds can be identified using conventional techniques known in the art and tests such as those described herein can be used to determine their activity. Such products can be produced, for example, by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymatic hydrolysis, etc. of the administered compound. Therefore, the present invention includes metabolites of the compounds of the present invention, including compounds produced by a method including contacting a compound of the general formula (I) of the present invention with a mammal for a period of time sufficient to produce its metabolite.
本领域技术人员会理解,由于氮需要可用的孤对电子来氧化成氧化物,因此并非所有的含氮杂环都能够形成N-氧化物;本领域技术人员会识别能够形成N-氧化物的含氮杂环。本领域技术人员还会认识到叔胺能够形成N-氧化物。用于制备杂环和叔胺的N-氧化物的合成方法是本领域技术人员熟知的,包括用过氧酸如过氧乙酸和间氯过氧苯甲酸(m-CPBA)、过氧化氢、烷基过氧化氢如叔丁基过氧化氢、过硼酸钠和双环氧乙烷(dioxirane)如二甲基双环氧乙烷来氧化杂环和叔胺。这些用于制备N-氧化物的方法已在文献中得到广泛描述和综述,参见例如:T.L.Gilchrist,Comprehensive Organic Synthesis,vol.7,pp 748-750;S.V.Ley,Ed.,Pergamon Press;M.Tisler和B.Stanovnik,Comprehensive Heterocyclic Chemistry,vol.3,pp 18-20。Those skilled in the art will understand that because nitrogen requires available lone pairs to oxidize to oxides, not all nitrogen-containing heterocycles can form N-oxides; those skilled in the art will recognize those capable of forming N-oxides Nitrogen-containing heterocycle. Those skilled in the art will also recognize that tertiary amines can form N-oxides. Synthetic methods for preparing N-oxides of heterocycles and tertiary amines are well known to those skilled in the art and include the use of peroxyacids such as peroxyacetic acid and m-chloroperoxybenzoic acid (m-CPBA), hydrogen peroxide, Alkyl hydrogen peroxides such as t-butyl hydroperoxide, sodium perborate and dioxiranes such as dimethyl dioxirane oxidize heterocycles and tertiary amines. These methods for preparing N-oxides have been widely described and reviewed in the literature, see for example: TLGilchrist, Comprehensive, Organic, Synthesis, vol. 7, pp 748-750; SVLey, Ed., Pergamon Press; M. Tisler and B. Stanovnik, Comprehensive Heterocyclic Chemistry, vol. 3, pp 18-20.
在制备本发明的化合物的任何过程中,保护在任何有关分子上的敏感基团或反应基团可能是必需的和/或期望的,由此形成本发明的化合物的化学保护的形式。这可以通过常规的保护基实现,例如,在Protective Groups in Organic Chemistry,ed.J.F.W.McOmie,Plenum Press,1973;和T.W.Greene & P.G.M.Wuts,Protective Groups in Organic Synthesis,John Wiley & Sons,1991中所述的那些保护基,这些参考文献通过援引加入本文。使用本领域已知的方法,在适当的后续阶段可以除去保护基。In any process for preparing the compounds of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any relevant molecules, thereby forming a chemically protected form of the compounds of the present invention. This can be achieved through conventional protecting groups, for example, as described in Protective Groups in Organic Chemistry, ed. JFW McOmie, Plenum Press, 1973; and TW Greene & PGMWuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991 Those protecting groups, these references are incorporated herein by reference. Using methods known in the art, the protecting group can be removed at an appropriate subsequent stage.
本发明在其范围内进一步包括本发明的化合物的前药。通常这样的前药会是所述化合物的官能团衍生物,其易于在体内转化成期望的治疗活性化合物。因此,在这些情况中,用于本发明的治疗方法的术语“给药”应包括用所要求保护的化合物中的一种或多种的前药形式来治疗各种疾病或病症,但是在向个体给药后所述前药形式在体内转化成上述化合物。例如,在“Design of Prodrug”,ed.H.Bundgaard,Elsevier,1985中,描述了选择和制备适合的前药衍生物的常规方法。The invention further includes within its scope prodrugs of the compounds of the invention. Usually such a prodrug will be a functional group derivative of the compound, which is easily converted into the desired therapeutically active compound in vivo. Therefore, in these cases, the term "administration" used in the treatment method of the present invention should include the treatment of various diseases or conditions with one or more prodrug forms of the claimed compounds, but After the individual is administered, the prodrug form is converted into the above compound in vivo. For example, in "Design of Prodrug", ed. H. Bundgaard, Elsevier, 1985, a conventional method for selecting and preparing suitable prodrug derivatives is described.
本文中所示的任何通式或结构,包括通式(I)的化合物在内,还意在表示所述化合物的未标记形式和同位素标记的形式。同位素标记的化合物具有本文给出的分子式所示的结构,除了一个或多个原子被具有选定原子质量或质量数的原子替代。本发明的化合物中可包含的同位素的实例包括氢、碳、氮、氧、磷、氟和氯的同位素,例如,但不限于 2H(氘,D)、 3H(氚)、 11C、 13C、 14C、 15N、 18F、 31P、 32P、 35S、 36Cl和 125I。各种同位素标记的本发明的化合物,例如,其中包含诸如 3H、 13C和 14C的放射性同位素的那些。这样的同位素标记的化合物可用于代谢研究、反应动力学研究、检测或显像技术,例如正电子发射断层摄影术(PET)或单光子发射断层摄影术(SPECT),包括药物或底物组织分布测定,或者用于患者的放射性治疗。本发明的氘标记的或取代的治疗性化合物可具有改进的有关分布、代谢和***(ADME)的DMPK(药物代谢和药物动力学)性质。用较重的同位素例如氘取代可能由于较大的代谢稳定性而提供某些治疗优点,例如,体内半衰期增长,或者剂量要求减小。 18F标记的化合物可用于PET或SPECT研究。本发明的同位素标记的化合物及其前药一般可通过实施路线或实施例中公开的方法和下述制备方法,以易得的同位素标记的试剂替代非同位素标记的试剂进行制备。此外,用较重的同位素特别是氘(即 2H或D)取代可由于较大的代谢稳定性而提供某些治疗优点,例如,体内半衰期增长或者剂量要求减小或者治疗指数改进。应理解,在此情况中氘被视为式(I)的化合物中的取代基。可通过同位素富集系数定义这样的较重的同位素特别是氘的浓度。在本 发明的化合物中,未明确指明为特定同位素的任何原子意在表示该原子的任何稳定的同位素。除非另外说明,当明确以“H”或“氢”标明某位置时,应理解为该位置具有其天然丰度同位素组成的氢。因此,在本发明的化合物中,明确标明氘(D)的任何原子意在表示氘。 Any general formula or structure shown herein, including the compound of general formula (I), is also intended to represent the unlabeled form and the isotopically labeled form of the compound. Isotope-labeled compounds have the structure shown in the molecular formula given herein, except that one or more atoms are replaced with atoms having a selected atomic mass or mass number. Examples of isotopes that may be included in the compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine, and chlorine, such as, but not limited to, 2 H (deuterium, D), 3 H (tritium), 11 C, 13 C, 14 C, 15 N, 18 F, 31 P, 32 P, 35 S, 36 Cl and 125 I. Various isotope-labeled compounds of the present invention, for example, those containing radioisotopes such as 3 H, 13 C, and 14 C. Such isotopically labeled compounds can be used in metabolic studies, reaction kinetic studies, detection or imaging techniques, such as positron emission tomography (PET) or single photon emission tomography (SPECT), including drug or substrate tissue distribution Determination, or used for radiotherapy of patients. The deuterium-labeled or substituted therapeutic compounds of the invention may have improved DMPK (pharmacokinetic and pharmacokinetic) properties related to distribution, metabolism and excretion (ADME). Substitution with heavier isotopes such as deuterium may provide certain therapeutic advantages due to greater metabolic stability, for example, increased half-life in the body, or reduced dose requirements. 18 F labeled compounds can be used for PET or SPECT studies. The isotopically-labeled compounds and prodrugs of the present invention can generally be prepared by replacing the non-isotopically-labeled reagents with readily available isotopically-labeled reagents by the methods disclosed in the implementation route or examples and the following preparation methods. In addition, substitution with heavier isotopes, especially deuterium (ie, 2 H or D), may provide certain therapeutic advantages due to greater metabolic stability, such as increased half-life in vivo or reduced dose requirements or improved therapeutic index. It should be understood that in this case deuterium is regarded as a substituent in the compound of formula (I). The concentration of such heavier isotopes, especially deuterium, can be defined by the isotope enrichment factor. In the compounds of the present invention, any atom not specifically designated as a specific isotope is intended to mean any stable isotope of that atom. Unless otherwise stated, when a position is explicitly marked with "H" or "hydrogen", it should be understood that the position has hydrogen with its natural abundance isotopic composition. Therefore, in the compound of the present invention, any atom clearly deuterium (D) is intended to represent deuterium.
本文使用的术语“药物组合物”包括包含治疗有效量的本发明的通式(I)的化合物的产品,以及直接地或间接地由本发明的通式(I)化合物的组合产生的任何产品。The term "pharmaceutical composition" as used herein includes products containing a therapeutically effective amount of a compound of formula (I) of the present invention, as well as any product produced directly or indirectly from a combination of compounds of formula (I) of the present invention.
化合物Chemical compound
在一些实施方案中,本发明提供通式(I)的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,In some embodiments, the present invention provides compounds of general formula (I) or their stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters , Metabolites, N-oxides, their chemically protected forms or prodrugs,
Figure PCTCN2019121534-appb-000003
Figure PCTCN2019121534-appb-000003
其中:among them:
X为CH或N;并且X is CH or N; and
R选自氢、卤素、C 1-6烷基和C 1-6卤代烷基。 R is selected from hydrogen, halogen, C 1-6 alkyl and C 1-6 haloalkyl.
在一些实施方案中,本发明提供化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,其中R选自氢、氟、甲基和三氟甲基。In some embodiments, the present invention provides compounds or their stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N- Oxides, their chemically protected forms or prodrugs, where R is selected from hydrogen, fluorine, methyl and trifluoromethyl.
在一些实施方案中,本发明提供化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,其中X为CH;并且R选自氢、氟、甲基和三氟甲基。In some embodiments, the present invention provides compounds or their stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N- Oxides, their chemically protected forms or prodrugs, where X is CH; and R is selected from hydrogen, fluorine, methyl and trifluoromethyl.
在一些实施方案中,本发明提供化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,其中X为N;并且R为氢。In some embodiments, the present invention provides compounds or their stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N- Oxides, their chemically protected forms or prodrugs, where X is N; and R is hydrogen.
在一些实施方案中,本发明提供化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,其中所述化合物选自:In some embodiments, the present invention provides compounds or their stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N- Oxides, their chemically protected forms or prodrugs, wherein the compound is selected from:
Figure PCTCN2019121534-appb-000004
Figure PCTCN2019121534-appb-000004
本发明的通式(I)的化合物可包含不对称中心或手性中心,因而可以不同的立体异构体形式存在。本发明化合物的所有立体异构体形式,包括但不限于其非对映异构体、对映异构体和阻转异构体以及它们的混合物例如外消旋混合物,意在构成本发明的一部分。The compound of the general formula (I) of the present invention may contain an asymmetric center or a chiral center, and thus may exist in different stereoisomeric forms. All stereoisomeric forms of the compounds of the present invention, including but not limited to their diastereomers, enantiomers, and atropisomers, and mixtures thereof, such as racemic mixtures, are intended to constitute the present invention portion.
此外,本发明涵盖所有的非对映异构体,包括顺-反(几何)异构体和构象异构体。例如,若通式(I)的化合物包含双键或稠合环,则顺式和反式形式以及其混合物被涵盖在本发明的范围内。在本文 所示的结构中,若未指明任何具体手性原子的立体化学,则所有的立体异构体被视为并被包含为本发明的化合物。若以表示具体构型的楔形实线或虚线指明立体化学,则如此指明和定义该立体异构体。In addition, the present invention covers all diastereomers, including cis-trans (geometric) isomers and conformational isomers. For example, if the compound of general formula (I) contains a double bond or a fused ring, the cis and trans forms and mixtures thereof are covered within the scope of the present invention. In the structures shown herein, if no stereochemistry of any specific chiral atom is indicated, then all stereoisomers are considered and included as compounds of the invention. If the stereochemistry is indicated by a solid wedge or dashed line representing a specific configuration, then the stereoisomer is so designated and defined.
本发明的化合物可以非溶剂化的形式、以及用药学可接受的溶剂例如水、乙醇等溶剂化的形式存在,并且本发明意在涵盖溶剂化的和非溶剂化的形式。The compounds of the present invention can exist in unsolvated forms as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and the present invention is intended to encompass both solvated and unsolvated forms.
本发明的化合物还可以不同的互变异构体形式存在,并且所有这样的形式都被涵盖在本发明的范围内。The compounds of the invention can also exist in different tautomeric forms, and all such forms are encompassed within the scope of the invention.
还应理解,实施方案中的任意两个或更多个实施方案的组合也包括在本发明的范围内。It should also be understood that combinations of any two or more of the embodiments are also included within the scope of the present invention.
药物组合物Pharmaceutical composition
本发明另一方面提供药物组合物,其包含至少一种如上所述的本发明的通式(I)的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,以及一种或多种药学可接受的载体。在一些实施方案中,所述药物组合物还可包含一种或多种其他治疗剂,例如适于预防或治疗由FXR介导的疾病或病症的其他治疗剂。Another aspect of the present invention provides a pharmaceutical composition comprising at least one compound of the general formula (I) of the present invention as described above or a stereoisomer, tautomer, polymorph, or solvate thereof (Eg hydrate), pharmaceutically acceptable salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, and one or more pharmaceutically acceptable carriers. In some embodiments, the pharmaceutical composition may further include one or more other therapeutic agents, such as other therapeutic agents suitable for preventing or treating diseases or conditions mediated by FXR.
本发明中“药学可接受的载体”是指与活性成分一同给药的稀释剂、辅剂、赋形剂或媒介物,并且其在合理的医学判断的范围内适于接触人类和/或其他动物的组织而没有过度的毒性、刺激、过敏反应或与合理的益处/风险比相应的其他问题或并发症。"Pharmaceutically acceptable carrier" in the present invention refers to diluents, adjuvants, excipients or vehicles administered together with the active ingredient, and it is suitable for contact with humans and/or others within the scope of reasonable medical judgment Animal tissue without excessive toxicity, irritation, allergic reactions or other problems or complications corresponding to a reasonable benefit/risk ratio.
在本发明的药物组合物中可使用的药学可接受的载体包括但不限于无菌液体,例如水和油,包括那些石油、动物、植物或合成来源的油,例如花生油、大豆油、矿物油、芝麻油等。当所述药物组合物通过静脉内给药时,水是示例性载体。还可以使用生理盐水和葡萄糖及甘油水溶液作为液体载体,特别是用于注射液。适合的药物赋形剂包括淀粉、葡萄糖、乳糖、蔗糖、明胶、麦芽糖、白垩、硅胶、硬脂酸钠、单硬脂酸甘油酯、滑石、氯化钠、脱脂奶粉、甘油、丙二醇、水、乙醇等。所述组合物还可以视需要包含少量的湿润剂、乳化剂或pH缓冲剂。口服制剂可以包含标准载体,如药物级的甘露醇、乳糖、淀粉、硬脂酸镁、糖精钠、纤维素、碳酸镁等。适合的药学可接受的载体的实例如在Remington’s Pharmaceutical Sciences(1990)中所述。Pharmaceutically acceptable carriers that can be used in the pharmaceutical composition of the present invention include, but are not limited to, sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil , Sesame oil, etc. When the pharmaceutical composition is administered intravenously, water is an exemplary carrier. Physiological saline and aqueous solutions of glucose and glycerin can also be used as liquid carriers, especially for injections. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, maltose, chalk, silica gel, sodium stearate, glyceryl monostearate, talc, sodium chloride, skimmed milk powder, glycerin, propylene glycol, water, Ethanol, etc. The composition may also contain small amounts of wetting agents, emulsifying agents or pH buffering agents as needed. Oral preparations may contain standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, cellulose, magnesium carbonate, and the like. Examples of suitable pharmaceutically acceptable carriers are described in Remington's Pharmaceuticals (1990).
本发明的药物组合物可以***地作用和/或局部地作用。为此目的,它们可以适合的途径给药,例如通过注射、静脉内、动脉内、皮下、腹膜内、肌内或经皮给药;或通过口服、含服、经鼻、透粘膜、局部、以眼用制剂的形式或通过吸入给药。The pharmaceutical composition of the present invention can act systemically and/or locally. For this purpose, they can be administered by suitable routes, for example by injection, intravenous, intraarterial, subcutaneous, intraperitoneal, intramuscular or transdermal administration; or by oral, buccal, nasal, transmucosal, topical, It is administered in the form of an ophthalmic preparation or by inhalation.
对于这些给药途径,可以适合的剂型给药本发明的药物组合物。For these administration routes, the pharmaceutical composition of the present invention can be administered in a suitable dosage form.
所述剂型包括但不限于片剂、胶囊剂、锭剂、硬糖剂、散剂、喷雾剂、乳膏剂、软膏剂、栓剂、凝胶剂、糊剂、洗剂、软膏剂、水性混悬剂、可注射溶液剂、酏剂、糖浆剂。The dosage form includes but is not limited to tablets, capsules, lozenges, hard candy, powder, spray, cream, ointment, suppository, gel, paste, lotion, ointment, aqueous suspension , Injectable solutions, elixirs, syrups.
治疗用途Therapeutic use
本发明的另一方面提供所述化合物和药物组合物的治疗用途。Another aspect of the invention provides therapeutic uses of the compounds and pharmaceutical compositions.
因此,在一些实施方案中,本发明涉及预防或治疗由FXR介导的疾病或病症的方法,所述方法包括向有此需要的个体给药治疗有效量的至少一种本发明的通式(I)的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,或者给药本发明的药物组合物。Therefore, in some embodiments, the present invention relates to a method of preventing or treating a disease or disorder mediated by FXR, the method comprising administering to a subject in need thereof a therapeutically effective amount of at least one general formula of the present invention ( I) The compound or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N-oxides, and their chemical protection Form or prodrug, or administration of the pharmaceutical composition of the present invention.
在另一些实施方案中,本发明涉及至少一种本发明的通式(I)的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药、或者本发明的药物组合物在制备用于预防或治疗由FXR介导的疾病或病症的药物中的用途。In other embodiments, the present invention relates to at least one compound of the general formula (I) of the present invention or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), Pharmaceutically acceptable salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, or the pharmaceutical composition of the invention in the preparation of a medicament for the prevention or treatment of diseases or conditions mediated by FXR the use of.
所述由FXR介导的疾病或病症包括但不限于:The diseases or conditions mediated by FXR include but are not limited to:
慢性肝内或某些形式的肝外胆汁淤积性病症;肝纤维化;肝的梗阻性或慢性炎性病症;肝硬化;脂肪肝及并发症;与酒精引发的肝硬化或与病毒传染性形式的肝炎相关的胆汁淤积性和纤维变性效果;在部分肝切除术后的肝衰竭或肝缺血;化疗相关的脂肪性肝炎(CASH);急性肝衰竭;Chronic intrahepatic or some forms of extrahepatic cholestasis; liver fibrosis; obstructive or chronic inflammatory conditions of the liver; cirrhosis of the liver; fatty liver and complications; liver cirrhosis caused by alcohol or infectious forms of the virus Hepatitis-related cholestatic and fibrotic effects; liver failure or liver ischemia after partial hepatectomy; chemotherapy-related steatohepatitis (CASH); acute liver failure;
炎性肠道疾病、血脂异常、动脉粥样硬化、糖尿病和相关疾病;脂质和脂蛋白病症;II型糖尿病以及I型和II型糖尿病的临床并发症,包括糖尿病性肾病、糖尿病性神经病变、糖尿病性视网膜病、及临床上明显的长期糖尿病的其他观察到的效果;由于强迫的脂质、特别是甘油三酯蓄积以及随后的促纤维化途径激活而导致的慢性脂肪性和纤维性变性引起的病症和疾病,例如非酒精性脂肪肝病(NAFLD)或非酒精性脂肪性肝炎(NASH);肥胖或代谢综合征(血脂障碍、糖尿病和体重指数异 常高的合并病症);Inflammatory bowel disease, dyslipidemia, atherosclerosis, diabetes and related diseases; lipid and lipoprotein disorders; type II diabetes and clinical complications of type I and type II diabetes, including diabetic nephropathy, diabetic neuropathy , Diabetic retinopathy, and other clinically apparent effects of long-term diabetes; chronic fatty and fibrotic degeneration due to forced lipid accumulation, especially triglycerides, and subsequent activation of pro-fibrotic pathways Caused conditions and diseases, such as non-alcoholic fatty liver disease (NAFLD) or non-alcoholic steatohepatitis (NASH); obesity or metabolic syndrome (combined disorders of dyslipidemia, diabetes and abnormally high body mass index);
急性心肌梗塞、急性中风或作为慢性梗阻性动脉粥样硬化终点发生的血栓形成;非恶性过度增殖性病症和恶性过度增殖性病症,特别是肝细胞癌、结肠腺瘤和息肉病、结肠腺癌、乳腺癌、胰腺癌、巴特氏食管癌和胃肠道和肝脏的其他形式的肿瘤性疾病。Acute myocardial infarction, acute stroke, or thrombosis that occurs as the end point of chronic obstructive atherosclerosis; non-malignant hyperproliferative disorders and malignant hyperproliferative disorders, especially hepatocellular carcinoma, colon adenomas, and polyposis, colon adenocarcinoma , Breast cancer, pancreatic cancer, Barth's esophageal cancer and other forms of neoplastic diseases of the gastrointestinal tract and liver.
除非另外说明,本文中所使用的术语“治疗”意指逆转、减轻、抑制所指示的疾病或病症或者这样的疾病或病症的一或多种症状的进展。Unless otherwise stated, the term "treatment" as used herein means reversing, alleviating, or inhibiting the indicated disease or disorder or the progression of one or more symptoms of such disease or disorder.
如本文所使用的“个体”包括人或非人动物。示例性人个体包括患有疾病(例如本文所述的疾病)的人个体(称为患者)或正常个体。本发明中“非人动物”包括所有脊椎动物,例如非哺乳动物(例如鸟类、两栖动物、爬行动物)和哺乳动物,例如非人灵长类、家畜和/或驯化动物(例如绵羊、犬、猫、奶牛、猪等)。"Individual" as used herein includes human or non-human animals. Exemplary human individuals include human individuals (referred to as patients) or normal individuals with a disease (such as the diseases described herein). In the present invention, "non-human animals" include all vertebrates, such as non-mammals (eg birds, amphibians, reptiles) and mammals, such as non-human primates, domestic animals and/or domesticated animals (eg sheep, dogs) , Cats, cows, pigs, etc.).
如本文中所使用的术语“治疗有效量”指被给药后会实现上述治疗效力的化合物的量。The term "therapeutically effective amount" as used herein refers to the amount of the compound that will achieve the above-mentioned therapeutic efficacy after being administered.
可调整给药方案以提供最期望的应答。例如,可给药单次推注,可随时间给药数个分剂量,或可如治疗情况的急需所表明而按比例减少或增加剂量。要注意,剂量值可随要减轻的病况的类型及严重性而变化,且可包括单次或多次剂量。要进一步理解,对于任何特定个体,具体的给药方案应根据个体需要及给药组合物或监督组合物的给药的人员的专业判断来随时间调整。The dosage regimen can be adjusted to provide the most desired response. For example, a single bolus injection can be administered, several divided doses can be administered over time, or the dosage can be reduced or increased proportionally as indicated by the urgent need for the treatment. It should be noted that the dose value may vary with the type and severity of the condition to be reduced, and may include single or multiple doses. It is to be further understood that for any particular individual, the specific dosing regimen should be adjusted over time based on the individual's needs and the professional judgment of the person administering the composition or supervising the administration of the composition.
所给药的本发明的化合物的量会取决于所治疗的个体、病症或病况的严重性、给药的速率、化合物的处置及处方医师的判断。一般而言,有效剂量在每日每kg体重约0.0001至约50mg,例如约0.01至约10mg/kg/日(单次或分次给药)。对70kg的人而言,这会合计为约0.007mg/日至约3500mg/日,例如约0.7mg/日至约700mg/日。在一些情况下,不高于前述范围的下限的剂量水平可以是足够的,而在其他情况下,仍可在不引起任何有害副作用的情况下采用较大剂量,条件是首先将所述较大剂量分成数个较小剂量以在一整天中给药。The amount of the compound of the present invention administered will depend on the individual, the severity of the condition or condition being treated, the rate of administration, the treatment of the compound, and the judgment of the prescribing physician. In general, the effective dose is about 0.0001 to about 50 mg per kg of body weight per day, for example about 0.01 to about 10 mg/kg/day (single or divided doses). For a 70 kg person, this would add up to about 0.007 mg/day to about 3500 mg/day, such as about 0.7 mg/day to about 700 mg/day. In some cases, a dose level not higher than the lower limit of the aforementioned range may be sufficient, while in other cases, a larger dose may still be used without causing any harmful side effects, provided that the larger The dose is divided into several smaller doses to be administered throughout the day.
本发明的化合物在所述药物组合物中的含量或用量可以是约0.01mg至约1000mg,适合地是0.1-500mg,优选0.5-300mg等。The content or amount of the compound of the present invention in the pharmaceutical composition may be about 0.01 mg to about 1000 mg, suitably 0.1-500 mg, preferably 0.5-300 mg and the like.
组合治疗Combination therapy
通式(I)的化合物可单独使用,或者与适于预防或治疗由FXR介导的疾病或病症的一种或多种其他治疗剂组合使用。在一些实施方案中,在所述药物组合物或者作为组合治疗的给药方案中,将通式(I)的化合物与例如具有抗过度增殖效力的其他治疗剂组合。所述其他治疗剂可以是例如化疗剂。所述药物组合物或给药方案的其他治疗剂优选地具有与通式(I)的化合物互补的活性,从而它们不会不利地相互影响。这样的化合物适合地以对预期目的有效的量组合存在。The compound of general formula (I) may be used alone or in combination with one or more other therapeutic agents suitable for preventing or treating diseases or conditions mediated by FXR. In some embodiments, in the pharmaceutical composition or as a dosing regimen for combination therapy, the compound of general formula (I) is combined with other therapeutic agents having anti-hyperproliferative efficacy, for example. The other therapeutic agent may be, for example, a chemotherapeutic agent. The pharmaceutical composition or other therapeutic agents of the administration regimen preferably have complementary activities to the compound of general formula (I) so that they do not adversely affect each other. Such compounds are suitably present in combination in amounts effective for the intended purpose.
组合治疗可以同时或依次给药的方案施用。当依次施用时,该组合可以在两次或多次给药中施用。组合给药包括使用分开的药物组合物或者包含通式(I)的化合物和其他治疗剂的单一药物组合物同时给药,以及以任意顺序相继地给药,其中优选存在两种(或所有)活性剂同时发挥它们的生物活性的时间段。Combination therapy can be administered simultaneously or sequentially. When administered sequentially, the combination can be administered in two or more doses. Combined administration includes simultaneous administration using separate pharmaceutical compositions or a single pharmaceutical composition containing a compound of general formula (I) and other therapeutic agents, and sequential administration in any order, with two (or all) preferably present Active agents simultaneously exert their biological activity for a period of time.
上述同时给药的药剂中的任一种的适合的剂量是当前使用的那些,并且由于新鉴定的药物与其他治疗剂或治疗的组合(协同)作用,可以降低。Suitable doses of any of the above-mentioned concurrently administered agents are those currently in use, and can be reduced due to the combined (synergistic) effect of the newly identified drug with other therapeutic agents or treatments.
组合治疗可提供“协同作用”并证明是“协同的”,即,活性成分在一起使用时所达到的效果大于分开使用所述化合物时所产生的效果之和。当所述活性成分:(1)在组合的单位剂量制剂中共同配制并同时给药或者递送时;(2)作为分开的制剂交替或平行地递送时;或者(3)通过一些其他方案时,可达到协同效果。当在交替疗法中递送时,当所述化合物例如通过在分开的注射器中分别注射、通过分开的丸剂或胶囊剂、或通过分开的输注依次给药或递送时,可达到协同效果。通常在交替疗法中,相继地,即连续地,给药有效剂量的各活性成分,而在组合治疗中,一起给药有效剂量的两种或多种活性成分。Combination therapy can provide "synergy" and prove to be "synergistic", that is, the effects achieved when the active ingredients are used together are greater than the sum of the effects produced when the compounds are used separately. When the active ingredient: (1) is co-formulated in a combined unit dose formulation and is simultaneously administered or delivered; (2) is delivered as a separate formulation alternately or in parallel; or (3) by some other regimen, Can achieve synergy. When delivered in alternation therapy, a synergistic effect can be achieved when the compounds are administered or delivered sequentially, for example by separate injections in separate syringes, by separate pills or capsules, or by separate infusions. Usually in alternating therapy, an effective dose of each active ingredient is administered sequentially, that is, continuously, while in combination therapy, an effective dose of two or more active ingredients is administered together.
在治疗的一个具体的实施方案中,通式(I)的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药可以与例如本文所述的那些其他治疗剂组合,还可与外科治疗和放疗组合。因此,本发明的组合治疗包括给药至少一种通式(I)的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,以及使用至少一种其他治疗方法。为了达到期望的组合治疗效果,选择通式(I)的化合物和其他治疗剂的量以及给药的相对时机。In a specific embodiment of treatment, the compound of general formula (I) or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, Esters, metabolites, N-oxides, their chemically protected forms or prodrugs can be combined with other therapeutic agents such as those described herein, and can also be combined with surgical treatment and radiotherapy. Therefore, the combination therapy of the present invention includes administration of at least one compound of general formula (I) or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable Salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, and the use of at least one other treatment. In order to achieve the desired combined therapeutic effect, the amount of the compound of general formula (I) and other therapeutic agents and the relative timing of administration are selected.
通式(I)的化合物的代谢物Metabolites of compounds of general formula (I)
本文所述的通式(I)的化合物的体内代谢物也在本发明的范围内。这样的产物可由例如被给药的 化合物的氧化、还原、水解、酰胺化、脱酰胺化、酯化、酶解等产生。因此,本发明包括通式(I)的化合物的代谢物,包括通过使本发明的化合物与哺乳动物接触足以产生其代谢物的时间的方法制得的化合物。In vivo metabolites of the compounds of general formula (I) described herein are also within the scope of the present invention. Such products can be produced, for example, by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymatic hydrolysis, etc. of the administered compound. Therefore, the present invention includes metabolites of the compound of the general formula (I), including compounds prepared by contacting the compound of the present invention with a mammal for a time sufficient to produce its metabolite.
代谢物通常通过制备本发明的放射性同位素(例如 14C或 3H)标记的化合物,将其以可检测的剂量(例如大于约0.5mg/kg)向动物例如大鼠、小鼠、天竺鼠、猴或人肠胃外给药,代谢足够的时间(通常约30秒至30小时),然后从尿、血液或其他生物样品分离其转化产物进行鉴定。这些产物易于分离,因为它们是标记的(其他的通过使用能够结合代谢物中存余的表位的抗体进行分离)。代谢物结构以常规方法测定,例如通过MS、LC/MS或NMR分析。通过以与本领域技术人员公知的常规药物代谢研究相同的方式进行代谢物的分析。代谢物,只要未在体内发现它们,可用于诊断测定中,以治疗性给药本发明的化合物。 Metabolites are usually prepared by preparing a compound labeled with a radioisotope of the present invention (for example, 14 C or 3 H) and administering it to animals such as rats, mice, guinea pigs, and monkeys in a detectable dose (for example, greater than about 0.5 mg/kg) Or human parenteral administration, metabolism for a sufficient period of time (usually about 30 seconds to 30 hours), and then its transformation product is isolated from urine, blood or other biological samples for identification. These products are easy to isolate because they are labeled (others are separated by using antibodies that can bind to the remaining epitopes in the metabolites). The metabolite structure is determined by conventional methods, for example by MS, LC/MS or NMR analysis. The analysis of metabolites is performed in the same way as conventional drug metabolism studies well known to those skilled in the art. Metabolites, as long as they are not found in the body, can be used in diagnostic assays to administer the compounds of the present invention therapeutically.
药盒Pillbox
在本发明的另一些实施方案中,提供包含用于治疗上述疾病或病症的材料的“药盒”。所述药盒包括容器,所述容器包含作为第一治疗剂的通式(I)的化合物、其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、或者其化学保护的形式或前药,或者包含作为第一药物组合物的本发明的药物组合物。在一些实施方案中,所述药盒还可包括在所述容器上或伴随所述容器的标签或包装说明书。术语“包装说明书”是指治疗产品的商业包装中通常包含的说明书,其包含使用该治疗产品相关的适应征、用法、剂量、给药、禁忌和/或警示的信息。适合的容器包括,例如,瓶、小瓶、注射器、泡罩包装等。所述容器可以由各种材料例如玻璃和塑料制成。所述容器可容纳对治疗病症有效的通式(I)的化合物或其制剂,并且可具有无菌入口(例如,所述容器可以是静脉内溶液剂袋或者具有可被皮下注射针刺穿的瓶塞的小瓶)。标签或包装说明书指明所述组合物用于治疗所选的病症例如癌症。此外,标签或包装说明书可指明要治疗的患者是患有诸如肝硬化、过度增殖性病症、动脉粥样硬化、I型糖尿病之类疾病或病症的患者标签或包装说明书还可指明所述组合物可用来治疗其他病症。在另一些实施方案中,所述药盒还包括第二容器,其包含作为第二治疗剂的适于预防或治疗由FXR介导的疾病或病症的至少一种其他治疗剂,或者作为第二药物组合物的包含所述其他治疗剂的药物组合物。因此,在一些实施方案中,所述药盒可包括给药所述第一治疗剂或第一药物组合物和所述第二治疗剂或第二药物组合物(若存在)的说明书。例如,若所述药盒包括含有通式(I)的化合物的第一组合物和包含其他治疗剂的第二药物组合物,则该药盒还可包括向有此需要的个体同时、相继或分开地给药所述第一药物组合物和第二药物组合物的说明书。替代地或者额外的,所述药盒还可包括第三容器,其包含药学可接受的缓冲剂例如抑菌的注射用水(BWFI)、磷酸盐缓冲盐水、林格氏液和葡萄糖溶液。所述药盒还可包括就商业和用户而言令人期望的其他材料,包括其他缓冲剂、稀释剂、填料、注射针和注射器。In other embodiments of the present invention, a "kit" containing materials for treating the above diseases or conditions is provided. The kit includes a container containing the compound of general formula (I), its stereoisomers, tautomers, polymorphs, solvates (such as hydrates) as the first therapeutic agent , A pharmaceutically acceptable salt, ester, metabolite, N-oxide, or a chemically protected form or prodrug thereof, or a pharmaceutical composition of the present invention containing the first pharmaceutical composition. In some embodiments, the kit may also include a label or package insert on or accompanying the container. The term "package insert" refers to the instructions usually included in commercial packages of therapeutic products, which contain information on indications, usage, dosage, administration, contraindications and/or warnings related to the use of the therapeutic product. Suitable containers include, for example, bottles, vials, syringes, blister packs, and the like. The container may be made of various materials such as glass and plastic. The container may contain a compound of general formula (I) or a preparation thereof that is effective for treating a condition, and may have a sterile inlet (for example, the container may be an intravenous solution sachet or have a pierceable by a hypodermic injection needle) Corks). The label or package insert indicates that the composition is used to treat a selected condition, such as cancer. In addition, the label or package insert may indicate that the patient to be treated is a patient with a disease or condition such as cirrhosis, hyperproliferative disorder, atherosclerosis, type I diabetes, or the label or package insert may also indicate the composition Can be used to treat other conditions. In other embodiments, the kit further includes a second container containing, as a second therapeutic agent, at least one other therapeutic agent suitable for preventing or treating a disease or condition mediated by FXR, or as a second A pharmaceutical composition comprising the other therapeutic agent. Therefore, in some embodiments, the kit may include instructions for administering the first therapeutic agent or first pharmaceutical composition and the second therapeutic agent or second pharmaceutical composition (if present). For example, if the kit includes a first composition containing a compound of general formula (I) and a second pharmaceutical composition containing other therapeutic agents, the kit may also include simultaneous, sequential or Instructions for administering the first pharmaceutical composition and the second pharmaceutical composition separately. Alternatively or additionally, the kit may further include a third container containing a pharmaceutically acceptable buffer such as bacteriostatic water for injection (BWFI), phosphate buffered saline, Ringer's solution, and glucose solution. The kit may also include other materials desirable for commercial and user purposes, including other buffers, diluents, fillers, injection needles, and syringes.
在另一些实施方案中,所述药盒适合于递送通式(I)的化合物的固体口服形式例如片剂或胶囊剂。这样的药盒优选地包括多个单位剂量。这样的药盒可包括具有以它们的预期用途定位的剂量的卡片。这样的药盒的一个实例是“泡罩包装”。泡罩包装在包装工业中是公知的并且广泛用于包装药学单位剂量形式。若期望,可以例如指定在治疗时间表中可给药之日的数字、字母或其他标记或者日历插页的形式提供记忆辅助工具。In other embodiments, the kit is suitable for the delivery of solid oral forms of compounds of general formula (I) such as tablets or capsules. Such a kit preferably includes multiple unit doses. Such kits may include cards with dosages positioned for their intended use. An example of such a kit is a "blister pack". Blister packaging is well known in the packaging industry and is widely used for packaging pharmaceutical unit dosage forms. If desired, memory aids can be provided in the form of numbers, letters, or other indicia or calendar inserts that specify the date of administration in the treatment schedule, for example.
化合物的制备方法Compound preparation method
在一些实施方案中,本发明提供制备本发明的通式(I)的化合物的方法,所述方法包括以下步骤:In some embodiments, the present invention provides a method of preparing a compound of general formula (I) of the present invention, the method comprising the following steps:
Figure PCTCN2019121534-appb-000005
Figure PCTCN2019121534-appb-000005
其中:among them:
Hal 1、Hal 2和Hal 3各自独立地为相同或不同的卤素,例如F、Cl、Br或I,优选为Cl或Br; Hal 1 , Hal 2 and Hal 3 are each independently the same or different halogen, such as F, Cl, Br or I, preferably Cl or Br;
PG 1为氨基保护基,优选为叔丁氧羰基(Boc); PG 1 is an amino protecting group, preferably tert-butoxycarbonyl (Boc);
PG 2为羧基保护基,优选为C 1-6烷基,更优选为甲基; PG 2 is a carboxyl protecting group, preferably C 1-6 alkyl, and more preferably methyl;
Y为硼酸或硼酸酯基团,优选为-B(OH) 2或者
Figure PCTCN2019121534-appb-000006
Y is a boric acid or borate group, preferably -B(OH) 2 or
Figure PCTCN2019121534-appb-000006
其余基团如上文所定义;The remaining groups are as defined above;
各步骤的反应条件如下:The reaction conditions of each step are as follows:
步骤A:使化合物IN-1与化合物IN-2反应以得到化合物IN-3;Step A: reacting compound IN-1 with compound IN-2 to obtain compound IN-3;
所述反应优选在适合的有机溶剂中进行。所述有机溶剂可选自直链或环状醚类(例如四氢呋喃或***等)、N-甲基吡咯烷酮、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、1,4-二氧六环、二甲基亚砜及其任意组合,优选四氢呋喃或N,N-二甲基甲酰胺。所述反应优选在适合的碱(例如碱金属的醇盐或碳酸盐)和/或催化剂的存在下进行。所述催化剂可以是包括冠醚的催化剂***,所述冠醚可选自15-冠醚-5和18-冠醚-6;所述碱金属的碳酸盐为例如碳酸钾或碳酸铯;所述碱金属的醇盐可选自叔丁醇钠、叔丁醇钾、甲醇钠、甲醇钾、乙醇钠、乙醇钾。优选地,所述碱金属的醇盐和催化剂是叔丁醇钠和/或叔丁醇钾与15-冠醚-5和/或18-冠醚-6的组合,优选叔丁醇钠与15-冠醚-5的组合或叔丁醇钾与18-冠醚-6的组合。所述反应优选在适合的温度下进行。所述温度优选为室温(20-30℃)或50-100℃(例如50-80℃)。所述反应优选进行合适的时间,例如1-24小时,例如5-15小时。The reaction is preferably carried out in a suitable organic solvent. The organic solvent can be selected from linear or cyclic ethers (such as tetrahydrofuran or diethyl ether), N-methylpyrrolidone, N,N-dimethylformamide, N,N-dimethylacetamide, 1, 4-Dioxane, dimethyl sulfoxide and any combination thereof, preferably tetrahydrofuran or N,N-dimethylformamide. The reaction is preferably carried out in the presence of a suitable base (eg alkali metal alkoxide or carbonate) and/or catalyst. The catalyst may be a catalyst system including a crown ether, and the crown ether may be selected from 15-crown-5 and 18-crown-6; the alkali metal carbonate is, for example, potassium carbonate or cesium carbonate; The alkali metal alkoxide may be selected from sodium tert-butoxide, potassium tert-butoxide, sodium methoxide, potassium methoxide, sodium ethoxide, potassium ethoxide. Preferably, the alkali metal alkoxide and catalyst are a combination of sodium t-butoxide and/or potassium t-butoxide and 15-crown-5 and/or 18-crown-6, preferably sodium t-butoxide and 15 -Combination of crown ether-5 or potassium tert-butoxide and 18-crown-6. The reaction is preferably carried out at a suitable temperature. The temperature is preferably room temperature (20-30°C) or 50-100°C (eg 50-80°C). The reaction is preferably carried out for a suitable time, for example 1-24 hours, for example 5-15 hours.
步骤B:移除化合物IN-3中的PG 1基团,以得到化合物IN-4; Step B: Remove the PG 1 group in compound IN-3 to obtain compound IN-4;
所述反应优选在适合的有机溶剂中进行。所述有机溶剂可选自卤代烃类(例如,二氯甲烷、氯仿、氯乙烷、二氯乙烷、三氯乙烷)、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺及其任意组合,优选二氯甲烷。所述反应可在酸性条件下进行,例如在1,4-二氧六环的氯化氢溶液中;或者合适的有机酸(例如羧酸或卤代酸,包括但不限于甲酸、氟乙酸、二氟乙酸、三氟乙酸、氯乙酸、二氯乙酸、三氯乙酸及其组合,优选三氟乙酸)的存在下进行。所述反应优选在适合的温度下进行。所述温度优选为室温(20-30℃)。所述反应优选进行合适的时间,例如1-5小时或6-15小时,例如2小时、4小时或过夜反应。The reaction is preferably carried out in a suitable organic solvent. The organic solvent may be selected from halogenated hydrocarbons (for example, dichloromethane, chloroform, ethyl chloride, dichloroethane, trichloroethane), N,N-dimethylformamide, N,N-di Methylacetamide and any combination thereof are preferably dichloromethane. The reaction can be carried out under acidic conditions, such as 1,4-dioxane in hydrogen chloride; or suitable organic acids (such as carboxylic acids or halogenated acids, including but not limited to formic acid, fluoroacetic acid, difluoro Acetic acid, trifluoroacetic acid, chloroacetic acid, dichloroacetic acid, trichloroacetic acid and combinations thereof, preferably trifluoroacetic acid). The reaction is preferably carried out at a suitable temperature. The temperature is preferably room temperature (20-30°C). The reaction is preferably carried out for a suitable time, such as 1-5 hours or 6-15 hours, such as 2 hours, 4 hours or overnight.
步骤C:使化合物IN-4与化合物IN-a反应以得到化合物IN-5;Step C: reacting compound IN-4 with compound IN-a to obtain compound IN-5;
在一些实施方案中,使化合物IN-4与化合物IN-a发生取代反应以得到化合物IN-5。所述取代反应优选在适合的有机溶剂中进行。所述有机溶剂可选自N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、四氢呋喃、N-甲基吡咯烷酮、二甲基亚砜及其任意组合,优选N,N-二甲基甲酰胺或N,N-二甲基乙酰胺。所述取代反应优选在适合的碱的存在下进行。优选地,所述碱是有机碱(例如有机胺类如三乙胺、N,N-二异丙基乙胺、N-甲基吗啉或吡啶,优选三乙胺或N,N-二异丙基乙胺)或无机碱(例如碱金属盐,优选碳酸钾)。所述取代反应优选在适合的温度下进行。所述温度可以是20-150℃,例如30-140℃,优选25℃、50℃、100℃或130℃,优选80℃。所述取代反应优选进行合适的时间,例如2-24小时、2-18小时或2-12小时,例如5、8或10小时。In some embodiments, compound IN-4 is substituted with compound IN-a to give compound IN-5. The substitution reaction is preferably carried out in a suitable organic solvent. The organic solvent may be selected from N,N-dimethylformamide, N,N-dimethylacetamide, tetrahydrofuran, N-methylpyrrolidone, dimethylsulfoxide and any combination thereof, preferably N,N- Dimethylformamide or N,N-dimethylacetamide. The substitution reaction is preferably carried out in the presence of a suitable base. Preferably, the base is an organic base (eg organic amines such as triethylamine, N,N-diisopropylethylamine, N-methylmorpholine or pyridine, preferably triethylamine or N,N-diiso Propylethylamine) or inorganic bases (eg alkali metal salts, preferably potassium carbonate). The substitution reaction is preferably carried out at a suitable temperature. The temperature may be 20-150°C, for example 30-140°C, preferably 25°C, 50°C, 100°C or 130°C, preferably 80°C. The substitution reaction is preferably carried out for a suitable time, such as 2-24 hours, 2-18 hours or 2-12 hours, such as 5, 8 or 10 hours.
在另一些实施方案中,使化合物IN-4与化合物IN-a发生偶联反应以得到化合物IN-5。所述偶联反应优选在金属催化剂和碱的存在下进行。优选地,所述金属催化剂是钯金属催化剂,例如三(二亚苄基丙酮)二钯、[1,1′-双(二苯基膦基)二茂铁]二氯化钯、三苯基膦钯、醋酸钯,优选三(二亚苄基丙酮)二钯。所述碱是无机碱,例如碳酸钾、碳酸铯、碳酸钠、碳酸氢钠、碳酸氢钾,优选碳酸铯。优选地,所述偶联反应在衍生自联苯的有机磷化合物的存在下进行,所述有机磷化合物选自BINAP、RuPhos和XPhos,优选BINAP。优选地,所述偶联反应在适合的有机溶剂中进行,所述有机溶剂可选自苯、甲苯和二甲苯,例如是甲苯。优选地,所述偶联反应在适合的保护气氛(例如氮气环境)下进行。优选地,所述偶联反应在适合的温度下进行,所述温度可以是70-100℃,优选80℃。优选地,所述偶联反应进行合适的时间,例如1-3小时,例如2小时。In other embodiments, the compound IN-4 is coupled with the compound IN-a to give compound IN-5. The coupling reaction is preferably carried out in the presence of a metal catalyst and a base. Preferably, the metal catalyst is a palladium metal catalyst, such as tris(dibenzylideneacetone) dipalladium, [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride, triphenyl Phosphorus palladium and palladium acetate, preferably tris(dibenzylideneacetone) dipalladium. The base is an inorganic base such as potassium carbonate, cesium carbonate, sodium carbonate, sodium bicarbonate, potassium bicarbonate, preferably cesium carbonate. Preferably, the coupling reaction is carried out in the presence of an organophosphorus compound derived from biphenyl, which is selected from BINAP, RuPhos and XPhos, preferably BINAP. Preferably, the coupling reaction is carried out in a suitable organic solvent, which may be selected from benzene, toluene and xylene, for example toluene. Preferably, the coupling reaction is carried out under a suitable protective atmosphere (for example, a nitrogen atmosphere). Preferably, the coupling reaction is carried out at a suitable temperature, which may be 70-100°C, preferably 80°C. Preferably, the coupling reaction is carried out for a suitable time, such as 1-3 hours, such as 2 hours.
步骤D:使化合物IN-5与化合物IN-b反应以得到化合物IN-6;Step D: reacting compound IN-5 with compound IN-b to obtain compound IN-6;
优选地,使化合物IN-5与化合物IN-b发生金属催化偶联反应得到化合物IN-6。所述金属催化偶联反应采用常规的方法进行。例如:化合物IN-5与化合物IN-b在溶剂(例如水、有机溶剂、或者有机溶剂与水的混合溶剂)中溶解,加入钯催化剂和碱,任选地在氮气保护下,在50℃到120℃的温度(优选80℃或90℃)下反应8到24小时(优选8小时或12小时)。所述有机溶剂为N,N-二甲基甲酰胺、四氢呋喃、1,4-二氧六环、甲苯或DME等。所述钯催化剂为三(二亚苄基丙酮)二钯、[1,1′-双(二苯基膦基)二茂铁]二氯化钯、三苯基膦钯、醋酸钯,优选[1,1′-双(二苯基膦基)二茂铁]二氯化钯或三(二亚苄基丙酮)二钯等。所述碱优选是无机碱,例如碳酸钾、碳酸铯、碳酸钠、碳酸氢钠或碳酸氢钾等。Preferably, the compound IN-5 and the compound IN-b undergo a metal-catalyzed coupling reaction to obtain the compound IN-6. The metal-catalyzed coupling reaction is carried out by conventional methods. For example: compound IN-5 and compound IN-b are dissolved in a solvent (such as water, an organic solvent, or a mixed solvent of organic solvent and water), a palladium catalyst and a base are added, optionally under nitrogen protection, at 50°C to The reaction is carried out at a temperature of 120°C (preferably 80°C or 90°C) for 8 to 24 hours (preferably 8 hours or 12 hours). The organic solvent is N,N-dimethylformamide, tetrahydrofuran, 1,4-dioxane, toluene or DME. The palladium catalyst is tris(dibenzylideneacetone) dipalladium, [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride, triphenylphosphine palladium, palladium acetate, preferably [ 1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride or tris(dibenzylideneacetone)dipalladium etc. The base is preferably an inorganic base, such as potassium carbonate, cesium carbonate, sodium carbonate, sodium bicarbonate, potassium bicarbonate, or the like.
步骤E:移除化合物IN-6中的PG 2基团,以得到通式(I)的化合物; Step E: Remove the PG 2 group in compound IN-6 to obtain the compound of general formula (I);
所述反应优选在适合的有机溶剂(所述有机溶剂可选自直链或环状醚类(例如四氢呋喃或***等)、N-甲基吡咯烷酮、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、1,4-二氧六环、二甲基亚砜及其任意组合,优选四氢呋喃)中进行。所述反应优选在醇或水以及碱的存在下进行。所述醇可以是例如甲醇或乙醇。所述碱可以选自碱金属氢氧化物,所述碱金属氢氧化物可以选自氢氧化锂、氢氧化钠和氢氧化钾。所述反应优选在适合的温度下进行。所述温度可以是室温至80℃,例如25℃或40-60℃。所述反应优选进行合适的时间,例如2-5小时或6-15小时,例如2、3或4小时或过夜。The reaction is preferably in a suitable organic solvent (the organic solvent may be selected from linear or cyclic ethers (such as tetrahydrofuran or diethyl ether, etc.), N-methylpyrrolidone, N,N-dimethylformamide, N, N-dimethylacetamide, 1,4-dioxane, dimethylsulfoxide and any combination thereof, preferably tetrahydrofuran). The reaction is preferably carried out in the presence of alcohol or water and a base. The alcohol may be, for example, methanol or ethanol. The base may be selected from alkali metal hydroxides, and the alkali metal hydroxide may be selected from lithium hydroxide, sodium hydroxide, and potassium hydroxide. The reaction is preferably carried out at a suitable temperature. The temperature may be room temperature to 80°C, for example 25°C or 40-60°C. The reaction is preferably carried out for a suitable time, such as 2-5 hours or 6-15 hours, such as 2, 3 or 4 hours or overnight.
在另一些实施方案中,本发明提供通式(IN-6)的化合物或其药学上可接受的盐,In other embodiments, the present invention provides compounds of general formula (IN-6) or pharmaceutically acceptable salts thereof,
Figure PCTCN2019121534-appb-000007
Figure PCTCN2019121534-appb-000007
其中各基团如上述所定义;Each group is as defined above;
所述化合物优选自:The compound is preferably selected from:
Figure PCTCN2019121534-appb-000008
Figure PCTCN2019121534-appb-000008
本文所用的术语“适合的”意指对具体化合物或条件的选择会取决于所要进行的特定合成操作以及所要转化的一个或多个分子的特性,但该选择在本领域技术人员的能力范围内。本文所述的所有工艺/方法的步骤均在足以提供所示产物的条件下进行。本领域技术人员会理解,可以改变所有反应条件(包括例如反应溶剂、反应时间、反应温度以及反应是否应在无水或惰性气氛下进行,等等)以优化期望的产物的收率,且这些变化在本领域技术人员的能力范围内。The term "suitable" as used herein means that the choice of a specific compound or condition will depend on the specific synthetic operation to be performed and the characteristics of the molecule or molecules to be converted, but the choice is within the ability of those skilled in the art . All process/method steps described herein are performed under conditions sufficient to provide the product shown. Those skilled in the art will understand that all reaction conditions (including, for example, reaction solvent, reaction time, reaction temperature, and whether the reaction should be carried out under anhydrous or inert atmosphere, etc.) can be modified to optimize the desired product yield, and these Variations are within the capabilities of those skilled in the art.
实施例提供制备通式(I)的化合物的示例性方法。本领域技术人员会理解,其他合成路线可用来合成通式(I)的化合物。虽然在实施例中描述和讨论了具体的原料和试剂,但是可替换成其他原料和试剂以提供各种衍生物和/或反应条件。此外,还可参考本公开,利用本领域技术人员公知的常规化学对通过所述方法制得的许多实施例化合物进一步进行修饰。The examples provide exemplary methods for preparing compounds of general formula (I). Those skilled in the art will understand that other synthetic routes can be used to synthesize compounds of general formula (I). Although specific raw materials and reagents are described and discussed in the examples, other raw materials and reagents can be substituted to provide various derivatives and/or reaction conditions. In addition, with reference to the present disclosure, many of the example compounds prepared by the method can be further modified using conventional chemistry well known to those skilled in the art.
在制备通式(I)的化合物时,可能需要保护中间体的远端官能团(例如羧基或氨基)。对这种保护的需要可随着远端官能团的性质以及制备方法的条件而改变。本领域技术人员容易地确定这样保护的必要性。关于保护基的概述及它们的用途,参见T.W.Greene,Protective Groups in Organic Synthesis,John Wiley & Sons,New York,1991。In the preparation of compounds of general formula (I), it may be necessary to protect the distal functional groups of the intermediates (eg carboxyl or amino). The need for such protection can vary with the nature of the remote functional group and the conditions of the preparation method. Those skilled in the art can easily determine the need for such protection. For an overview of protecting groups and their use, see T.W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.
实施例Examples
以下结合实施例进一步描述本发明,但提供这些实施例并非意在限制本发明的范围。The present invention is further described below in conjunction with examples, but these examples are not intended to limit the scope of the present invention.
化合物的结构是通过核磁共振( 1H-NMR)或质谱(MS)来确定的。 1H-NMR位移(δ)以百万分之一(ppm)的单位给出。化学位移是以10 -6(ppm)作为单位给出。 The structure of the compound is determined by nuclear magnetic resonance ( 1 H-NMR) or mass spectrometry (MS). The 1 H-NMR shift (δ) is given in units of parts per million (ppm). Chemical shifts are given in units of 10 -6 (ppm).
MS的测定是使用Agilent(ESI)质谱仪。The MS was measured using an Agilent (ESI) mass spectrometer.
制备高效液相使用岛津制备高效液相色谱仪。Preparation of high-performance liquid chromatography Shimadzu preparation of high-performance liquid chromatography was used.
薄层色谱硅胶板(TLC)使用Merck产的铝板(20×20cm),薄层色谱法分离纯化采用的是GF 254(0.4~0.5mm)。Thin layer chromatography silica gel plate (TLC) uses Merck aluminum plate (20×20cm), and thin layer chromatography separation and purification uses GF 254 (0.4~0.5mm).
反应的监测采用薄层色谱法(TLC)或LC-MS,使用的展开剂体系有:二氯甲烷和甲醇体系、正己烷和乙酸乙酯体系或者石油醚和乙酸乙酯体系。根据化合物的极性不同,需要调节溶剂的体积比,或者需要进一步加入三乙胺等,以实现产物的分离纯化。The reaction is monitored by thin layer chromatography (TLC) or LC-MS. The developing system used is: dichloromethane and methanol system, n-hexane and ethyl acetate system or petroleum ether and ethyl acetate system. Depending on the polarity of the compound, the volume ratio of the solvent needs to be adjusted, or triethylamine or the like needs to be further added to separate and purify the product.
微波反应使用
Figure PCTCN2019121534-appb-000009
Initiator+(400W,RT~300℃)微波反应器。
Use of microwave reaction
Figure PCTCN2019121534-appb-000009
Initiator+ (400W, RT ~ 300 ℃) microwave reactor.
柱色谱法一般使用200~300目硅胶为载体。洗脱剂的体系包括:二氯甲烷和甲醇体系,正己烷和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的三乙胺进行调节。Column chromatography generally uses 200-300 mesh silica gel as a carrier. The eluent system includes: dichloromethane and methanol system, n-hexane and ethyl acetate system, the volume ratio of the solvent is adjusted according to the polarity of the compound, and a small amount of triethylamine can also be added for adjustment.
除非特别指出,实施例的反应温度为室温(20℃~30℃)。Unless otherwise specified, the reaction temperature of the examples is room temperature (20°C to 30°C).
本发明所使用的试剂购自Acros Organics、Aldrich Chemical Company、上海特伯化学科技有限公司等。The reagents used in the present invention were purchased from Acros Organics, Aldrich Chemical Company, Shanghai Tebo Chemical Technology Co., Ltd. and so on.
在常规的合成法以及实施例和中间体制备例中,各缩写具有以下含义。In the conventional synthesis method and the examples and intermediate preparation examples, each abbreviation has the following meanings.
缩写abbreviation 含义meaning 缩写abbreviation 含义meaning
TLCTLC 薄层色谱法Thin layer chromatography LC-MSLC-MS 液相色谱-质谱联用仪Liquid chromatography-mass spectrometry
DMFDMF N,N-二甲基甲酰胺N,N-dimethylformamide DMEDME 乙二醇二甲醚Ethylene glycol dimethyl ether
中间体制备例:Examples of intermediate preparation:
中间体制备例1:4-((((1R,3r,5S)-8-(4-溴噻唑-2-基)-8-氮杂双环[3.2.1]辛-3-基)氧基)甲基)-5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑(T1)的制备Intermediate preparation example 1: 4-((((1R,3r,5S)-8-(4-bromothiazol-2-yl)-8-azabicyclo[3.2.1]oct-3-yl)oxy )Methyl)-5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazole (T1)
Figure PCTCN2019121534-appb-000010
Figure PCTCN2019121534-appb-000010
第一步:(E)-2-(三氟甲氧基)苯甲醛肟(T1-b)的制备Step 1: Preparation of (E)-2-(trifluoromethoxy)benzaldehyde oxime (T1-b)
将2-(三氟甲氧基)苯甲醛(T1-a)(150g,788.98mmol)溶于乙醇(1000mL)和水(1000mL)的混合溶剂中,机械搅拌下加入盐酸羟胺(65.79g,946.77mmol),有白色固体析出,继续加入1M的氢氧化钠水溶液(789mL,788.98mmol)。将反应物在25℃下反应2小时,向混合物加入1M的盐酸(2000mL)调节体系pH为5左右,直接进行抽滤,50℃下烘干过夜,得到化合物(T1-b)(150g,收率:92%)。Dissolve 2-(trifluoromethoxy)benzaldehyde (T1-a) (150g, 788.98mmol) in a mixed solvent of ethanol (1000mL) and water (1000mL), add hydroxylamine hydrochloride (65.79g, 946.77) with mechanical stirring mmol), a white solid precipitated, and continued to add 1M aqueous sodium hydroxide solution (789 mL, 788.98 mmol). The reaction was reacted at 25°C for 2 hours, 1M hydrochloric acid (2000 mL) was added to the mixture to adjust the pH of the system to about 5, directly filtered with suction, and dried overnight at 50°C to obtain compound (T1-b) (150g, collected Rate: 92%).
第二步:(Z)-N-羟基-2-(三氟甲氧基)亚胺苄基氯(T1-c)的制备Step 2: Preparation of (Z)-N-hydroxy-2-(trifluoromethoxy)imine benzyl chloride (T1-c)
将化合物(T1-b)(150g,731.23mmol)溶于DMF(1000mL),在0℃机械搅拌下加入N-氯代丁二酰亚胺(117.17g,877.48mmol),0℃反应1小时。向反应物中加入水(2000mL),随后用乙酸乙酯(1000mL×3)萃取。将有机层用无水硫酸钠(500g)干燥,过滤浓缩后得到化合物(T1-c)(160g,收率:91%)。Compound (T1-b) (150 g, 731.23 mmol) was dissolved in DMF (1000 mL), N-chlorosuccinimide (117.17 g, 877.48 mmol) was added under mechanical stirring at 0°C, and reacted at 0°C for 1 hour. Water (2000 mL) was added to the reaction, followed by extraction with ethyl acetate (1000 mL×3). The organic layer was dried over anhydrous sodium sulfate (500g), filtered and concentrated to obtain compound (T1-c) (160g, yield: 91%).
第三步:5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-甲酸甲酯(T1-d)的制备Step 3: Preparation of 5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazole-4-carboxylic acid methyl ester (T1-d)
将3-环丙基-3-氧代丙酸甲酯(189.87g,1.34mol)加入化合物(T1-c)(160g,667.84mmol)中,反应物在-5℃搅拌,滴加三乙胺(500mL),-5℃反应过夜。将反应液倒入水(20L)中,机械搅拌30分钟,有固体出现,抽滤得到黄色固体,50℃下烘干过夜,得到化合物(T1-d)(120g,收率:55%)。Methyl 3-cyclopropyl-3-oxopropionate (189.87g, 1.34mol) was added to compound (T1-c) (160g, 667.84mmol), the reaction was stirred at -5°C, and triethylamine was added dropwise (500mL), -5 °C overnight. The reaction solution was poured into water (20 L) and mechanically stirred for 30 minutes. A solid appeared. Suction filtration gave a yellow solid, which was dried overnight at 50°C to obtain compound (T1-d) (120 g, yield: 55%).
第四步:(5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲醇(T1-e)的制备Step 4: Preparation of (5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl)methanol (T1-e)
将化合物(T1-d)(120g,366.69mmol)溶于甲苯中,反应物在-10℃搅拌,滴加2M的二异丁基氢化铝(550mL,1.10mol),室温反应过夜。将反应液倒入加有冰块的甲醇(1000mL)中,机械搅拌下,加入水(3000mL),进行抽滤,得到黄色固体,滤液用乙酸乙酯(2000mL×3)萃取,无水硫酸钠(500g)干燥,过滤,旋干有机相得到化合物(T1-e)(100g,收率:91%)。Compound (T1-d) (120 g, 366.69 mmol) was dissolved in toluene, the reaction was stirred at -10°C, 2M diisobutylaluminum hydride (550 mL, 1.10 mol) was added dropwise, and the reaction was carried out overnight at room temperature. Pour the reaction solution into methanol (1000 mL) with ice cubes, add water (3000 mL) under mechanical stirring, and perform suction filtration to obtain a yellow solid. The filtrate is extracted with ethyl acetate (2000 mL×3), anhydrous sodium sulfate (500g) Dry, filter and spin-dry the organic phase to obtain compound (T1-e) (100g, yield: 91%).
第五步:4-(氯甲基)-5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑(T1-f)的制备Step 5: Preparation of 4-(chloromethyl)-5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazole (T1-f)
将苯并三氮唑(59.65g,500.74mmol)溶于二氯甲烷中,反应物在-5℃搅拌,滴加二氯亚砜(59.65g,501.39mmol),室温搅拌半小时后,加入化合物(T1-e)(100g,334.17mmol)的二氯甲烷溶液(500mL),室温反应6小时。将反应液进行抽滤,滤液旋干得到化合物(T1-f)(106g,收率:94%)。Benzotriazole (59.65g, 500.74mmol) was dissolved in dichloromethane, the reaction was stirred at -5 °C, dichlorosulfoxide (59.65g, 501.39mmol) was added dropwise, after stirring at room temperature for half an hour, the compound was added A dichloromethane solution (500 mL) of (T1-e) (100 g, 334.17 mmol) was reacted at room temperature for 6 hours. The reaction solution was filtered with suction, and the filtrate was spin-dried to obtain compound (T1-f) (106 g, yield: 94%).
第六步:(1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛烷-8-甲酸叔丁酯(T1-g)的制备The sixth step: (1R, 3r, 5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl)methoxy)- Preparation of 8-azabicyclo[3.2.1]octane-8-carboxylic acid tert-butyl ester (T1-g)
将化合物(T1-f)(74.77g,328.94mmol)溶于四氢呋喃(500mL)中,随后加入18-冠醚-6(118.56g,448.55mmol)。反应物在0℃搅拌,加入叔丁醇钾(50.33g,448.55mmol),随后移至室温搅拌,加入(1R,3r,5S)-3-羟基-8-氮杂双环[3.2.1]辛烷-8-甲酸叔丁酯(95g,299.03mmol),室温反应过夜。将反应液旋干,残余物加入乙酸乙酯(1500mL)和水(1500mL),有机相用饱和食盐水(1500mL)洗两次,有机相浓缩后,经硅胶柱色谱法纯化(洗脱剂:石油醚/乙酸乙酯=10/1-5/1)得到化合物(T1-g)(85g,收率:56%)。Compound (T1-f) (74.77 g, 328.94 mmol) was dissolved in tetrahydrofuran (500 mL), and then 18-crown-6 (118.56 g, 448.55 mmol) was added. The reaction was stirred at 0°C, potassium tert-butoxide (50.33g, 448.55mmol) was added, and then moved to room temperature and stirred, (1R, 3r, 5S)-3-hydroxy-8-azabicyclo[3.2.1]octane was added Tert-Butyl alkane-8-carboxylate (95 g, 299.03 mmol), react overnight at room temperature. The reaction solution was spin-dried, ethyl acetate (1500 mL) and water (1500 mL) were added to the residue. The organic phase was washed twice with saturated brine (1500 mL). After the organic phase was concentrated, it was purified by silica gel column chromatography (eluent: Petroleum ether/ethyl acetate = 10/1-5/1) to obtain compound (T1-g) (85g, yield: 56%).
第七步:4-(((1R,3r,5S)-8-氮杂双环[3.2.1]辛-3-基氧基)甲基)-5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑(T1-h)的制备Seventh step: 4-(((1R,3r,5S)-8-azabicyclo[3.2.1]oct-3-yloxy)methyl)-5-cyclopropyl-3-(2-( Preparation of trifluoromethoxy)phenyl)isoxazole (T1-h)
将化合物(T1-g)(85g,167.15mmol)溶于二氯甲烷(500mL)中,随后加入盐酸二氧六环溶液(4M,500mL)。室温反应过夜。将反应液旋干得到化合物(T1-h)(60g,收率:81%)。Compound (T1-g) (85 g, 167.15 mmol) was dissolved in dichloromethane (500 mL), followed by the addition of dioxane hydrochloride solution (4M, 500 mL). Reaction at room temperature overnight. The reaction solution was spin-dried to obtain compound (T1-h) (60 g, yield: 81%).
第八步:4-((((1R,3r,5S)-8-(4-溴噻唑-2-基)-8-氮杂双环[3.2.1]辛-3-基)氧基)甲基)-5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑(T1)的制备Step 8: 4-((((1R,3r,5S)-8-(4-bromothiazol-2-yl)-8-azabicyclo[3.2.1]oct-3-yl)oxy)methan Yl)-5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazole (T1)
将化合物(T1-h)(7g,15.73mmol)溶于DMF(50mL)中,随后加入N,N-二异丙基乙胺(10.17g,78.67mmol)和2,4-二溴噻唑(4.59g,18.88mmol),100℃反应12小时。反应液加水(300mL),用乙酸乙酯(1000mL),无水硫酸钠(20g)干燥,过滤,有机相浓缩后,经硅胶柱色谱法纯化(洗脱剂:石油醚/乙酸乙酯=10/1-6/1)得到化合物(T1)(2.8g,收率:31%)。Compound (T1-h) (7 g, 15.73 mmol) was dissolved in DMF (50 mL), followed by addition of N,N-diisopropylethylamine (10.17 g, 78.67 mmol) and 2,4-dibromothiazole (4.59 g, 18.88 mmol), and reacted at 100°C for 12 hours. The reaction solution was added with water (300 mL), dried with ethyl acetate (1000 mL), anhydrous sodium sulfate (20 g), filtered, and the organic phase was concentrated, then purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate=10 /1-6/1) Compound (T1) (2.8 g, yield: 31%) was obtained.
实施例1:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)苯甲酸(C1)的制备Example 1: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)benzoic acid (C1)
Figure PCTCN2019121534-appb-000011
Figure PCTCN2019121534-appb-000011
第一步:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)苯甲酸甲酯(C1-a)的制备The first step: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)benzoic acid methyl ester (C1-a)
室温下,将化合物(T1)(1g,1.75mmol)溶于1,4-二氧六环(20mL)中,随后加入4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)苯甲酸甲酯(532.50mg,1.93mmol)、[1,1′-双(二苯基膦基)二茂铁]二氯化钯(143.16mg,195.66μmol)和碳酸钾(483.85mg,3.50mmol),氮气置换2-3遍。80℃反应8小时。反应液用硅藻土抽滤,滤液用乙酸乙酯(100mL×3)洗涤,所得有机相用无水硫酸钠(20g)干燥后过滤浓缩,所得剩余物经硅胶柱色谱法纯化(洗脱剂:石油醚/乙酸乙酯=10/1-6/1)得到本步的标题化合物(1.12g)。At room temperature, compound (T1) (1 g, 1.75 mmol) was dissolved in 1,4-dioxane (20 mL), followed by addition of 4-(4,4,5,5-tetramethyl-1,3, 2-Dioxaborolan-2-yl)benzoic acid methyl ester (532.50mg, 1.93mmol), [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride ( 143.16mg, 195.66μmol) and potassium carbonate (483.85mg, 3.50mmol), nitrogen substitution 2-3 times. Reaction at 80°C for 8 hours. The reaction solution was suction-filtered with celite, the filtrate was washed with ethyl acetate (100 mL×3), the resulting organic phase was dried over anhydrous sodium sulfate (20 g), filtered and concentrated, and the obtained residue was purified by silica gel column chromatography (eluent : Petroleum ether/ethyl acetate=10/1-6/1) to obtain the title compound of this step (1.12g).
MS m/z(ESI):626.1[M+H] +MS m/z (ESI): 626.1 [M+H] + .
第二步:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)苯甲酸(C1)的制备The second step: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)benzoic acid (C1)
将化合物(C1-a)(390mg,625.31μmol)溶于四氢呋喃(5mL)中,加入氢氧化钠(25.01mg,625.31μmol)的水溶液(2mL),25℃反应4小时。反应液用1M的稀盐酸调至pH为4,用乙酸乙酯(100mL×3)萃取,有机相用无水硫酸钠干燥,过滤,浓缩,所得剩余物经硅胶柱色谱法纯化(洗脱剂:石油醚/乙酸乙酯=10/1-6/1)得到标题化合物(150mg)。Compound (C1-a) (390 mg, 625.31 μmol) was dissolved in tetrahydrofuran (5 mL), an aqueous solution (2 mL) of sodium hydroxide (25.01 mg, 625.31 μmol) was added, and the mixture was reacted at 25° C. for 4 hours. The reaction solution was adjusted to pH 4 with 1M dilute hydrochloric acid, extracted with ethyl acetate (100 mL×3), the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated, and the resulting residue was purified by silica gel column chromatography (eluent : Petroleum ether/ethyl acetate = 10/1-6/1) to obtain the title compound (150 mg).
MS m/z(ESI):612.1[M+H] +MS m/z (ESI): 612.1 [M+H] + .
1H-NMR(400MHz,DMSO-d 6)δ:7.96(s,1H),7.68(dd,J=18.6,7.7Hz,1H),7.58(dd,J=13.9,6.8Hz,1H),7.45(s,1H),4.36(s,1H),4.15(s,1H),2.36(s,1H),2.04(d,J=15.2Hz,1H),1.82(s,1H),1.70(d,J=14.9Hz,1H),1.39-1.26(m,1H),1.13(d,J=22.2Hz,1H)。 1 H-NMR (400 MHz, DMSO-d 6 ) δ: 7.96 (s, 1H), 7.68 (dd, J=18.6, 7.7 Hz, 1H), 7.58 (dd, J=13.9, 6.8 Hz, 1H), 7.45 (s, 1H), 4.36 (s, 1H), 4.15 (s, 1H), 2.36 (s, 1H), 2.04 (d, J = 15.2 Hz, 1H), 1.82 (s, 1H), 1.70 (d, J=14.9 Hz, 1H), 1.39-1.26 (m, 1H), 1.13 (d, J=22.2 Hz, 1H).
实施例2:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)-3-甲基苯甲酸(C2)的制备Example 2: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-3-methylbenzoic acid (C2)
Figure PCTCN2019121534-appb-000012
Figure PCTCN2019121534-appb-000012
第一步:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)-3-甲基苯甲酸甲酯(C2-a)的制备The first step: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-3-methylbenzoic acid methyl ester (C2-a)
室温下,将化合物(T1)(500mg,876.55mmol)溶于1,4-二氧六环(20mL)中,随后加入3-甲基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)苯甲酸甲酯(266.25mg,964.20μmol)、[1,1′-双(二苯基膦基)二茂铁]二氯化钯(71.58mg,97.83μmol)和碳酸钾(241.93mg,1.75mmol),氮气置换2-3遍。80℃反应8小时。反应液用硅藻土抽滤,滤液用乙酸乙酯(100mL×3)洗涤,所得有机相用无水硫酸钠(20g)干燥后浓缩,所得剩余物经硅胶柱色谱法纯化(洗脱剂:石油醚/乙酸乙酯=10/1-6/1)得到本步的标题化合物(400mg)。At room temperature, compound (T1) (500 mg, 876.55 mmol) was dissolved in 1,4-dioxane (20 mL), followed by addition of 3-methyl-4-(4,4,5,5-tetramethyl -1,3,2-dioxaborolan-2-yl)benzoic acid methyl ester (266.25 mg, 964.20 μmol), [1,1′-bis(diphenylphosphino)ferrocene] Palladium dichloride (71.58 mg, 97.83 μmol) and potassium carbonate (241.93 mg, 1.75 mmol) were replaced with nitrogen gas 2-3 times. Reaction at 80°C for 8 hours. The reaction solution was suction filtered with celite, the filtrate was washed with ethyl acetate (100 mL×3), the resulting organic phase was dried over anhydrous sodium sulfate (20 g) and concentrated, and the obtained residue was purified by silica gel column chromatography (eluent: Petroleum ether/ethyl acetate = 10/1-6/1) to obtain the title compound (400 mg) of this step.
MS m/z(ESI):640.2[M+H] +MS m/z (ESI): 640.2 [M+H] + .
第二步:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)-3-甲基苯甲酸(C2)的制备The second step: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-3-methylbenzoic acid (C2)
将化合物(C2-a)(300mg,468.98μmol)溶于四氢呋喃(5mL)中,加入氢氧化钠(18.76mg,468.98μmol)的水溶液(2mL),25℃下反应4小时。反应液用1M的稀盐酸调至pH为4,用乙酸乙酯(100mL×3)萃取,有机相用无水硫酸钠干燥,过滤,浓缩,所得剩余物经硅胶柱色谱法纯化(洗脱剂:石油醚∶乙酸乙酯=10∶1-6∶1)得到标题化合物(280mg)。Compound (C2-a) (300 mg, 468.98 μmol) was dissolved in tetrahydrofuran (5 mL), an aqueous solution (2 mL) of sodium hydroxide (18.76 mg, 468.98 μmol) was added, and the mixture was reacted at 25° C. for 4 hours. The reaction solution was adjusted to pH 4 with 1M dilute hydrochloric acid, extracted with ethyl acetate (100 mL×3), the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated, and the resulting residue was purified by silica gel column chromatography (eluent : Petroleum ether: ethyl acetate = 10: 1-6: 1) to obtain the title compound (280 mg).
MS m/z(ESI):612.1[M+H] +MS m/z (ESI): 612.1 [M+H] + .
1H-NMR(400MHz,DMSO-d 6)δ:7.84(s,1H),7.80(d,J=8.1Hz,1H),7.74-7.68(m,1H),7.66(d,J=6.2Hz,1H),7.58(dd,J=13.7,6.4Hz,1H),7.06(s,1H),4.35(s,1H),4.11(s,1H),2.51(d,J=15.3Hz,4H),2.41-2.32(m,1H),2.04(d,J=14.7Hz,1H),1.94(s,1H),1.82(s,2H),1.69(d,J=14.6Hz,1H),1.19-1.14(m,2H),1.10(m,2H). 1 H-NMR (400 MHz, DMSO-d 6 ) δ: 7.84 (s, 1H), 7.80 (d, J=8.1 Hz, 1H), 7.74-7.68 (m, 1H), 7.66 (d, J=6.2 Hz , 1H), 7.58 (dd, J = 13.7, 6.4Hz, 1H), 7.06 (s, 1H), 4.35 (s, 1H), 4.11 (s, 1H), 2.51 (d, J = 15.3Hz, 4H) , 2.41-2.32 (m, 1H), 2.04 (d, J = 14.7 Hz, 1H), 1.94 (s, 1H), 1.82 (s, 2H), 1.69 (d, J = 14.6 Hz, 1H), 1.19- 1.14(m, 2H), 1.10(m, 2H).
实施例3:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基]噻唑-4-基)-3-氟苯甲酸(C3)的制备Example 3: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl]thiazol-4-yl)-3-fluorobenzoic acid (C3)
Figure PCTCN2019121534-appb-000013
Figure PCTCN2019121534-appb-000013
第一步:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)-3-氟苯甲酸甲酯(C3-a)的制备The first step: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-3-fluorobenzoic acid methyl ester (C3-a)
室温下,将化合物(T1)(650mg,1.14mmol)溶于1,4-二氧六环(20mL)中,随后加入3-氟-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)苯甲酸甲酯(248.13mg,1.25mmol)、[1,1′-双(二苯基膦基)二茂铁]二氯化钯(93.06mg,127.18μmol)和碳酸钾(314.50mg,2.28mmol),氮气置换2-3遍。80℃反应8小时。反应液用硅藻土抽滤,滤液用乙酸乙酯(100mL×3)洗涤,所得有机相用无水硫酸钠(20g)干燥后过滤浓缩,所得剩余物经硅胶柱色谱法纯化(洗脱剂:石油醚/乙酸乙酯=10/1)得到本步的标题化合物(350mg)。At room temperature, compound (T1) (650 mg, 1.14 mmol) was dissolved in 1,4-dioxane (20 mL), followed by addition of 3-fluoro-4-(4,4,5,5-tetramethyl- 1,3,2-Dioxaborolan-2-yl)benzoic acid methyl ester (248.13mg, 1.25mmol), [1,1′-bis(diphenylphosphino)ferrocene]di Palladium chloride (93.06 mg, 127.18 μmol) and potassium carbonate (314.50 mg, 2.28 mmol) were replaced with nitrogen gas 2-3 times. Reaction at 80°C for 8 hours. The reaction solution was suction-filtered with celite, the filtrate was washed with ethyl acetate (100 mL×3), the resulting organic phase was dried over anhydrous sodium sulfate (20 g), filtered and concentrated, and the obtained residue was purified by silica gel column chromatography (eluent : Petroleum ether/ethyl acetate = 10/1) to obtain the title compound (350 mg) of this step.
MS m/z(ESI):644.1[M+H] +MS m/z (ESI): 644.1 [M+H] + .
第二步:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基]噻唑-4-基)-3-氟苯甲酸(C3)的制备The second step: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl]thiazol-4-yl)-3-fluorobenzoic acid (C3)
将化合物(C3-a)(390mg,625.31μmol)溶于四氢呋喃(5mL)中,加入氢氧化钠(25.01mg,625.31μmol)的水溶液(2mL),25℃下反应4小时。反应液用1M的稀盐酸调至pH为4,用乙酸乙酯(100mL×3)萃取,有机相用无水硫酸钠干燥浓缩,所得剩余物经硅胶柱色谱法纯化(洗脱剂:石油醚/乙酸乙酯=10/1-6/1)得到标题化合物(1.01g)。Compound (C3-a) (390 mg, 625.31 μmol) was dissolved in tetrahydrofuran (5 mL), an aqueous solution (2 mL) of sodium hydroxide (25.01 mg, 625.31 μmol) was added, and the mixture was reacted at 25° C. for 4 hours. The reaction solution was adjusted to pH 4 with 1M dilute hydrochloric acid, extracted with ethyl acetate (100 mL×3), the organic phase was dried and concentrated over anhydrous sodium sulfate, and the resulting residue was purified by silica gel column chromatography (eluent: petroleum ether) /Ethyl acetate=10/1-6/1) to obtain the title compound (1.01 g).
MS m/z(ESI):620.1[M+H] +MS m/z (ESI): 620.1 [M+H] + .
1H-NMR(400MHz,DMSO-d 6)δ:7.71(t,J=7.8Hz,1H),7.66(d,J=7.4Hz,1H),7.63-7.54(m,2H),7.50(d,J=11.7Hz,1H),7.33(s,1H),4.35(s,1H),4.12(s,1H),2.39-2.33(m,1H),2.03(d,J=14.5Hz,1H),1.80(s,2H),1.69(d,J=14.7Hz,1H),1.33-1.24(m,1H),1.16(m,1H),1.10(m,1H)。 1 H-NMR (400 MHz, DMSO-d 6 ) δ: 7.71 (t, J=7.8 Hz, 1H), 7.66 (d, J=7.4 Hz, 1H), 7.63-7.54 (m, 2H), 7.50 (d , J = 11.7Hz, 1H), 7.33 (s, 1H), 4.35 (s, 1H), 4.12 (s, 1H), 2.39-2.33 (m, 1H), 2.03 (d, J = 14.5Hz, 1H) , 1.80 (s, 2H), 1.69 (d, J = 14.7 Hz, 1H), 1.33-1.24 (m, 1H), 1.16 (m, 1H), 1.10 (m, 1H).
实施例4:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环 [3.2.1]辛-8-基)噻唑-4-基)-2-氟苯甲酸(C4)的制备Example 4: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-2-fluorobenzoic acid (C4)
Figure PCTCN2019121534-appb-000014
Figure PCTCN2019121534-appb-000014
第一步:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)-2-氟苯甲酸甲酯(C4-a)的制备The first step: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-2-fluorobenzoic acid methyl ester (C4-a)
室温下,将化合物(T1)(400mg,701.24μmol)溶于1,4-二氧六环(20mL)中,随后加入2-氟-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)苯甲酸甲酯(152.70mg,771.36μmol)、[1,1′-双(二苯基膦基)二茂铁]二氯化钯(57.27mg,78.26μmol)和碳酸钾(193.54mg,1.40mmol),氮气置换2-3遍。80℃反应8小时。反应液用硅藻土抽滤,滤液用乙酸乙酯(100mL×3)洗涤,所得有机相用无水硫酸钠(20g)干燥后过滤浓缩,所得剩余物经硅胶柱色谱法纯化(洗脱剂:石油醚/乙酸乙酯=10/1)得到本步的标题化合物(200mg)。At room temperature, compound (T1) (400 mg, 701.24 μmol) was dissolved in 1,4-dioxane (20 mL), followed by addition of 2-fluoro-4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)benzoic acid methyl ester (152.70 mg, 771.36 μmol), [1,1′-bis(diphenylphosphino)ferrocene]di Palladium chloride (57.27 mg, 78.26 μmol) and potassium carbonate (193.54 mg, 1.40 mmol) were replaced with nitrogen gas 2-3 times. Reaction at 80°C for 8 hours. The reaction solution was suction-filtered with celite, the filtrate was washed with ethyl acetate (100 mL×3), the resulting organic phase was dried over anhydrous sodium sulfate (20 g), filtered and concentrated, and the obtained residue was purified by silica gel column chromatography (eluent : Petroleum ether/ethyl acetate=10/1) to obtain the title compound (200 mg) of this step.
MS m/z(ESI):630.1[M+H] +MS m/z (ESI): 630.1 [M+H] + .
第二步:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)-2-氟苯甲酸(C4)的制备The second step: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-2-fluorobenzoic acid (C4)
将化合物(C4-a)(200.00mg,310.73μmol)溶于四氢呋喃(5mL)中,加入氢氧化钠(12.43mg,310.73μmol)的水溶液(2mL),25℃下反应4小时。反应液用1M的稀盐酸调至pH为4,用乙酸乙酯(100mL×3)萃取,有机相用无水硫酸钠干燥浓缩,所得剩余物经硅胶柱色谱法纯化(洗脱剂:石油醚/乙酸乙酯=10/1-6/1)得到标题化合物(170mg)。Compound (C4-a) (200.00 mg, 310.73 μmol) was dissolved in tetrahydrofuran (5 mL), an aqueous solution (2 mL) of sodium hydroxide (12.43 mg, 310.73 μmol) was added, and the reaction was carried out at 25° C. for 4 hours. The reaction solution was adjusted to pH 4 with 1M dilute hydrochloric acid, extracted with ethyl acetate (100 mL×3), the organic phase was dried and concentrated over anhydrous sodium sulfate, and the resulting residue was purified by silica gel column chromatography (eluent: petroleum ether) /Ethyl acetate=10/1-6/1) to obtain the title compound (170 mg).
MS m/z(ESI):620.1[M+H] +MS m/z (ESI): 620.1 [M+H] + .
1H-NMR(400MHz,DMSO-d 6)δ:7.85(d,J=8.1Hz,1H),7.72(dd,J=17.5,10.3Hz,1H),7.66(d,J=7.3Hz,1H),7.58(dd,J=13.5,6.3Hz,1H),7.37(d,J=1.9Hz,1H),4.36(s,1H),4.15(s,1H),2.37(d,J=4.9Hz,1H),2.03(d,J=14.7Hz,1H),1.82(s,2H),1.70(d,J=14.6Hz,1H),1.33-1.24(m,1H),1.15(dd,J=11.0,6.1Hz,1H),1.10(d,J=3.0Hz,1H)。 1 H-NMR (400 MHz, DMSO-d 6 ) δ: 7.85 (d, J=8.1 Hz, 1H), 7.72 (dd, J=17.5, 10.3 Hz, 1H), 7.66 (d, J=7.3 Hz, 1H ), 7.58 (dd, J = 13.5, 6.3 Hz, 1H), 7.37 (d, J = 1.9 Hz, 1H), 4.36 (s, 1H), 4.15 (s, 1H), 2.37 (d, J = 4.9 Hz) , 1H), 2.03 (d, J = 14.7 Hz, 1H), 1.82 (s, 2H), 1.70 (d, J = 14.6 Hz, 1H), 1.33-1.24 (m, 1H), 1.15 (dd, J = 11.0, 6.1 Hz, 1H), 1.10 (d, J=3.0 Hz, 1H).
实施例5:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)-2-(三氟甲基)苯甲酸(C5)的制备Example 5: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-2-(trifluoromethyl)benzoic acid (C5)
Figure PCTCN2019121534-appb-000015
Figure PCTCN2019121534-appb-000015
第一步:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)-2-(三氟甲基)苯甲酸甲酯(C5-a)的制备The first step: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-2-(trifluoromethyl)benzoic acid methyl ester (C5-a)
室温下,将化合物(T1)(0.4g,701.24μmol)溶于1,4-二氧六环(20mL)中,随后加入4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-2-(三氟甲基)苯甲酸甲酯(277.78mg,841.48μmol)、[1,1′-双(二苯基膦基)二茂铁]二氯化钯(57.27mg,70.12μmol)和碳酸钾(193.83mg,1.40mmol),氮气置换2-3遍。80℃反应8小时。反应液用硅藻土抽滤,滤液用乙酸乙酯(100mL×3)洗涤,所得有机相用无水硫酸钠(20g)干燥后过滤浓缩,所得剩余物经硅胶柱色谱法纯化(洗脱剂:石油醚/乙酸乙酯=15/1-8/1)得到本步的标题化合物(0.36g)。At room temperature, compound (T1) (0.4 g, 701.24 μmol) was dissolved in 1,4-dioxane (20 mL), followed by addition of 4-(4,4,5,5-tetramethyl-1,3 , 2-dioxaborolane-2-yl)-2-(trifluoromethyl)benzoic acid methyl ester (277.78 mg, 841.48 μmol), [1,1′-bis(diphenylphosphino ) Ferrocene] palladium dichloride (57.27 mg, 70.12 μmol) and potassium carbonate (193.83 mg, 1.40 mmol), nitrogen substitution 2-3 times. Reaction at 80°C for 8 hours. The reaction solution was suction-filtered with celite, the filtrate was washed with ethyl acetate (100 mL×3), the resulting organic phase was dried over anhydrous sodium sulfate (20 g), filtered and concentrated, and the obtained residue was purified by silica gel column chromatography (eluent : Petroleum ether/ethyl acetate=15/1-8/1) to obtain the title compound of this step (0.36g).
MS m/z(ESI):694.1[M+H] +MS m/z (ESI): 694.1 [M+H] + .
第二步:4-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)-2-(三氟甲基)苯甲酸(C5)的制备The second step: 4-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)-2-(trifluoromethyl)benzoic acid (C5)
将化合物(C5-a)(360.00mg,518.99μmol)溶于四氢呋喃(5mL)中,加入氢氧化钠(20.71mg,518.99μmol)的水溶液(2mL),25℃下反应4小时。反应液用1M的稀盐酸调至pH为4,用乙酸乙酯(100mL×3)萃取,有机相用无水硫酸钠干燥浓缩,所得剩余物经硅胶柱色谱法纯化(洗脱剂:石油醚/乙酸乙酯=10/1-6/1)得到标题化合物(45mg)。Compound (C5-a) (360.00 mg, 518.99 μmol) was dissolved in tetrahydrofuran (5 mL), an aqueous solution (2 mL) of sodium hydroxide (20.71 mg, 518.99 μmol) was added, and the reaction was carried out at 25° C. for 4 hours. The reaction solution was adjusted to pH 4 with 1M dilute hydrochloric acid, extracted with ethyl acetate (100 mL×3), the organic phase was dried and concentrated over anhydrous sodium sulfate, and the resulting residue was purified by silica gel column chromatography (eluent: petroleum ether) /Ethyl acetate=10/1-6/1) to obtain the title compound (45 mg).
MS m/z(ESI):680.1[M+H] +MS m/z (ESI): 680.1 [M+H] + .
1H-NMR(400MHz,DMSO-d 6)δ:8.21(s,1H),8.16(d,J=7.3Hz,1H),7.83(d,J=7.2Hz,1H),7.64(dd,J=18.9,11.7Hz,1H),7.57(d,J=12.8Hz,1H),4.33(s,1H),4.12(s,1H),2.33(s,1H),2.01(d,J=12.4Hz,1H),1.79(s,1H),1.68(m,1H),1.19-1.01(m,1H)。 1 H-NMR (400 MHz, DMSO-d 6 ) δ: 8.21 (s, 1H), 8.16 (d, J=7.3 Hz, 1H), 7.83 (d, J=7.2 Hz, 1H), 7.64 (dd, J = 18.9, 11.7 Hz, 1H), 7.57 (d, J = 12.8 Hz, 1H), 4.33 (s, 1H), 4.12 (s, 1H), 2.33 (s, 1H), 2.01 (d, J = 12.4 Hz , 1H), 1.79 (s, 1H), 1.68 (m, 1H), 1.19-1.01 (m, 1H).
实施例6:5-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)吡啶甲酸(C6)的制备Example 6: 5-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)picolinic acid (C6)
Figure PCTCN2019121534-appb-000016
Figure PCTCN2019121534-appb-000016
第一步:5-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)吡啶甲酸甲酯(C6-a)的制备The first step: 5-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Preparation of methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)picolinic acid methyl ester (C6-a)
室温下,将化合物(T1)(50mg,87.65μmol溶于1,4-二氧六环(20mL)中,随后加入5-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)吡啶甲酸甲酯(27.67mg,105.19μmol)、[1,1′-双(二苯基膦基)二茂铁]二氯化钯(7.16mg,8.77μmol)和碳酸钾(12.11mg,87.65μmol),氮气置换2-3遍。80℃反应8小时。反应液用硅藻土抽滤,滤液用乙酸乙酯(100mL×3)洗涤,所得有机相用无水硫酸钠(20g)干燥后过滤浓缩,所得剩余物经硅胶柱色谱法纯化(洗脱剂:石油醚/乙酸乙酯=10/1-6/1)得到本步的标题化合物(20mg)。At room temperature, compound (T1) (50 mg, 87.65 μmol was dissolved in 1,4-dioxane (20 mL), followed by addition of 5-(4,4,5,5-tetramethyl-1,3,2 -Dioxaborolan-2-yl)picolinic acid methyl ester (27.67mg, 105.19μmol), [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride (7.16 mg, 8.77 μmol) and potassium carbonate (12.11 mg, 87.65 μmol), nitrogen substitution 2-3 times. The reaction was carried out at 80°C for 8 hours. The reaction solution was filtered with celite, and the filtrate was washed with ethyl acetate (100 mL×3). The obtained organic phase was dried over anhydrous sodium sulfate (20g), filtered and concentrated, and the obtained residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate=10/1-6/1) to obtain the title of this step Compound (20mg).
MS m/z(ESI):626.1[M+H] +MS m/z (ESI): 626.1 [M+H] + .
第二步:5-(2-((1R,3r,5S)-3-((5-环丙基-3-(2-(三氟甲氧基)苯基)异噁唑-4-基)甲氧基)-8-氮杂双环[3.2.1]辛-8-基)噻唑-4-基)吡啶甲酸(C6)的制备The second step: 5-(2-((1R,3r,5S)-3-((5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl)isoxazol-4-yl ) Methoxy)-8-azabicyclo[3.2.1]oct-8-yl)thiazol-4-yl)picolinic acid (C6)
将化合物(C6-a)(200mg,316.61μmol)溶于四氢呋喃(5mL)中,加入氢氧化钠(12.66mg,316.61μmol)的水溶液(2mL),25℃下反应4小时。反应液用1M的稀盐酸调至pH为4,用乙酸乙酯(100mL×3)萃取,有机相用无水硫酸钠干燥后过滤浓缩,所得剩余物经硅胶柱色谱法纯化(洗脱剂:石油醚/乙酸乙酯=10/1-6/1)得到标题化合物(25mg)。Compound (C6-a) (200 mg, 316.61 μmol) was dissolved in tetrahydrofuran (5 mL), an aqueous solution (2 mL) of sodium hydroxide (12.66 mg, 316.61 μmol) was added, and the mixture was reacted at 25° C. for 4 hours. The reaction solution was adjusted to pH 4 with 1M dilute hydrochloric acid, and extracted with ethyl acetate (100 mL×3). The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated. The obtained residue was purified by silica gel column chromatography (eluent: Petroleum ether/ethyl acetate = 10/1-6/1) to give the title compound (25mg).
MS m/z(ESI):618.1[M+H] +MS m/z (ESI): 618.1 [M+H] + .
1H-NMR(400MHz,DMSO-d 6)δ:8.88(s,1H),8.20(dd,J=8.1,2.1Hz,1H),7.93(d,J=8.2Hz,1H),7.71-7.61(m,1H),7.55(t,J=7.5Hz,1H),7.44(s,1H),4.32(s,1H),4.11(s,1H),3.52(s,1H),2.33(dd,J=10.8,5.9Hz,1H),2.01(d,J=13.7Hz,1H),1.79(s,2H),1.67(d,J=14.5Hz,1H),1.13(m,1H),1.09-1.03(m,1H)。 1 H-NMR (400 MHz, DMSO-d 6 ) δ: 8.88 (s, 1H), 8.20 (dd, J=8.1, 2.1 Hz, 1H), 7.93 (d, J=8.2 Hz, 1H), 7.71-7.61 (m, 1H), 7.55 (t, J = 7.5 Hz, 1H), 7.44 (s, 1H), 4.32 (s, 1H), 4.11 (s, 1H), 3.52 (s, 1H), 2.33 (dd, J = 10.8, 5.9 Hz, 1H), 2.01 (d, J = 13.7 Hz, 1H), 1.79 (s, 2H), 1.67 (d, J = 14.5 Hz, 1H), 1.13 (m, 1H), 1.09- 1.03 (m, 1H).
生物学测定Biological assay
实验例1:胆汁酸受体FXR辅激活因子结合实验Experimental Example 1: Bile acid receptor FXR coactivator binding experiment
1.试验方法1. Test method
采用Invitrogen·LanthaScreen TM TR-FRET Farnesoid X Receptor Coactivator Assay试剂盒测定化合物对FXR的激活作用。 Invitrogen·LanthaScreen TR-FRET Farnesoid X Receptor Coactivator Assay kit was used to determine the FXR activation effect of the compound.
将受体与不同浓度的待测化合物在室温下孵育后,加入荧光标记的辅激活因子短肽及铽标记的抗体,在室温下反应后检测FRET信号。以无受体蛋白组为空白,使用四参数拟合方程计算待测化合物对FXR的激活活性EC 50及最大激活效应值E maxAfter incubating the receptor with test compounds at different concentrations at room temperature, fluorescently labeled coactivator short peptides and terbium-labeled antibodies are added, and the FRET signal is detected after reaction at room temperature. With the receptor-free proteome as a blank, use the four-parameter fitting equation to calculate the activation activity EC 50 and maximum activation effect value E max of the test compound for FXR:
y=Emin+(Emax-Emin)/(1+(x/EC 50)^(-Hillslope)) y=Emin+(Emax-Emin)/(1+(x/EC 50 )^(-Hillslope))
其中y为FRET结合信号,Emax和Emin分别为拟合曲线的上渐近线估值和下渐近线估值,x为化合物的对数浓度,Hillslope为曲线斜率。Where y is the FRET binding signal, Emax and Emin are the upper and lower asymptotic estimates of the fitted curve, respectively, x is the log concentration of the compound, and Hillslope is the slope of the curve.
另外,以鹅去氧胆酸(即CDCA)为阳性对照,通过以下公式计算本发明所测试化合物的相对激活效应:In addition, using chenodeoxycholic acid (ie, CDCA) as a positive control, the relative activation effect of the tested compounds of the present invention was calculated by the following formula:
相对激活效应(%)=(Emax/Emax’)×100%Relative activation effect (%)=(Emax/Emax’)×100%
其中,Emax代表本发明所测试化合物的最大激活效应值,Emax’代表CDCA的最大激活效应值,二者均通过上文所示的公式计算得到。Among them, Emax represents the maximum activation effect value of the tested compound of the present invention, and Emax' represents the maximum activation effect value of CDCA, both of which are calculated by the formula shown above.
2.试验结果2. Test results
表1.本发明所测试的化合物对FXR的EC 50 Table 1. EC 50 of compounds tested by the present invention for FXR
化合物编号Compound number EC 50(μM) EC 50 (μM)
CDCACDCA 4.434.43
C3C3 0.1330.133
C4C4 0.0220.022
C5C5 0.0020.002
C6C6 0.0310.031
表1的数据显示,相对于鹅去氧胆酸(CDCA)的值为4.43μM的EC 50,所测试的化合物具有更低的EC 50(0.002-0.133μM),表明本发明化合物对FXR具有更好的激活活性。 The data in Table 1 shows that the compounds tested have a lower EC 50 (0.002-0.133 μM) relative to an EC 50 of chenodeoxycholic acid (CDCA) value of 4.43 μM, indicating that the compounds of the present invention have a greater effect on FXR Good activation activity.
表2.本发明所测试的化合物对FXR的相对激活效应Table 2. Relative activation effects of compounds tested by the present invention on FXR
化合物编号Compound number 相对激活效应Relative activation effect
CDCACDCA 100%100%
C3C3 154%154%
C4C4 137%137%
C5C5 196%196%
C6C6 177%177%
表2的数据显示,本发明所测试的化合物的最大激活效应值较鹅去氧胆酸(CDCA)的最大激活效应值更高,说明本发明化合物对FXR具有很好的最大激活效应。The data in Table 2 shows that the maximum activation effect value of the tested compounds of the present invention is higher than that of chenodeoxycholic acid (CDCA), indicating that the compounds of the present invention have a very good maximum activation effect on FXR.
综合表1和表2的EC 50值和相对激活效应数据显示,本发明的化合物对FXR具有较好的激活作用。 Combining the EC 50 values and relative activation effect data in Table 1 and Table 2 shows that the compound of the present invention has a better activation effect on FXR.
实验例2.荧光素酶报告基因检测实验Experimental example 2. Luciferase reporter gene detection experiment
1.试验方法1. Test method
人胚胎肾细胞HEK293培养于含有10%FBS的DMEM培养基中。共转染质粒,使其高表达FXR和人源BSEP荧光素酶报告基因。将转染细胞消化、重悬,计数,然后接种于多孔板中。加入10μL不同浓度待测化合物于多孔板中,使其终浓度分别为64μM、16μM、4μM、1μM、0.25μM、0.0625μM、0.0156μM、0.0039μM、0.000975μM、0.000244μM、0μM,DMSO终浓度为0.5%。待测化合物与细胞孵育18h后,加入Brigh-GloTM检测试剂,用多功能全自动酶标仪检测化学发光单位值(RLU),以空白孔(不含待测化合物)信号值为100%,计算各待测化合物浓度下的相对信号百分比(%)。用SigmaPlot 10软件采用四参数模型拟合所测试的化合物EC 50和最大激动效应Emax(相对信号百分比)。 Human embryonic kidney cells HEK293 were cultured in DMEM medium containing 10% FBS. Co-transfect the plasmid to make it highly express FXR and human BSEP luciferase reporter gene. The transfected cells were digested, resuspended, counted, and then seeded in multi-well plates. Add 10 μL of different concentrations of the test compound to the multi-well plate to make the final concentration of 64 μM, 16 μM, 4 μM, 1 μM, 0.25 μM, 0.0625 μM, 0.0156 μM, 0.0039 μM, 0.000975 μM, 0.000244 μM, 0 μM, the final concentration of DMSO is 0.5%. After incubating the test compound with the cells for 18h, add Brigh-GloTM detection reagent, use a multi-functional automatic microplate reader to detect the chemiluminescence unit value (RLU), and the signal value of the blank well (excluding the test compound) is 100%. The relative signal percentage (%) at each test compound concentration. Model using a four parameter fit using SigmaPlot 10 software tested compound EC 50 and maximum agonistic effect Emax (relative percentage of signal).
2.试验结果2. Test results
测定结果列于下表3。The measurement results are shown in Table 3 below.
表3table 3
化合物编号Compound number EC 50(μM) EC 50 (μM) EmaxEmax
C1C1 0.058±0.0230.058±0.023 252%252%
C2C2 0.033±0.0180.033±0.018 320%320%
C3C3 0.037±0.0190.037±0.019 343%343%
C4C4 0.021±0.0060.021±0.006 322%322%
C5C5 0.014±0.0050.014±0.005 301%301%
表3的数据显示,所测试的的化合物在体外细胞测定中的EC 50值在0.014μM~0.058μM之间,Emax值大于250%。表明本发明的化合物在体外细胞测定中具有良好的FXR激活活性。 Data in Table 3 show, EC 50 values of the compounds tested in an in vitro cell assay between 0.014μM ~ 0.058μM, Emax values greater than 250%. It shows that the compound of the present invention has good FXR activation activity in in vitro cell assay.
实验例3:肝微粒体稳定性实验Experimental Example 3: Liver microsome stability experiment
1.实验方法:1. Experimental method:
待测化合物(50μL)与各种属肝微粒体(100μL)混合,37℃预孵5分钟后,加入NADPH(50μL),孵育0、30、60分钟,待测化合物、NADPH和肝微粒体酶的孵育浓度分别为1μM、1mM和0.5mg/mL。加入冰乙腈(200μL)终止反应后加入适当体积内标(利伐沙班),涡旋、离心取上清,检测。The test compound (50 μL) was mixed with various liver microsomes (100 μL). After pre-incubation at 37°C for 5 minutes, NADPH (50 μL) was added and incubated for 0, 30, and 60 minutes. The test compound, NADPH and liver microsome enzymes The incubation concentrations are 1μM, 1mM and 0.5mg/mL, respectively. After adding ice acetonitrile (200 μL) to terminate the reaction, an appropriate volume of internal standard (rivaroxaban) was added, and the supernatant was collected by vortexing and centrifugation for detection.
检测方法:Detection method:
LC-MS/MS,质谱为API 5500,液相为Shimadzu LC-30AD***。色谱柱为Hypersil GOLD C18,1.9μm粒径,50×2.1mm;流动相A相为水+0.1%甲酸,B相为乙腈;流速为0.55mL/min,柱温为40℃。采用离子源为ESI源正离子模式,扫描方式为多重反应监测(MRM)。LC-MS/MS, mass spectrometer is API5500, liquid phase is Shimadzu LC-30AD system. The chromatographic column is Hypersil C18, 1.9μm particle size, 50×2.1mm; mobile phase A is water + 0.1% formic acid, phase B is acetonitrile; flow rate is 0.55mL/min, column temperature is 40 ℃. The ion source is the positive ion mode of the ESI source, and the scanning mode is multiple reaction monitoring (MRM).
通过测定不同孵育时间的样品浓度,以“Ln(药物残留量%)”对“孵育时间”作图获得速率常数,从而计算出药物的半衰期与肝清除率,以药物半衰期与肝清除率值来评价药物在肝微粒体中的代谢稳定性。By measuring the sample concentration at different incubation times, the rate constant is obtained by plotting "Ln (% of drug residue)" versus "incubation time", thereby calculating the half-life and liver clearance of the drug. Evaluate the metabolic stability of the drug in liver microsomes.
2.实验结果:2. Experimental results:
表4Table 4
Figure PCTCN2019121534-appb-000017
Figure PCTCN2019121534-appb-000017
Figure PCTCN2019121534-appb-000018
Figure PCTCN2019121534-appb-000018
表5table 5
Figure PCTCN2019121534-appb-000019
Figure PCTCN2019121534-appb-000019
结论:从表4和表5的实验数据可以看出,本发明的化合物在肝微粒体中清除较慢,稳定性较好,具有良好的药物代谢动力学性质。Conclusion: From the experimental data in Table 4 and Table 5, it can be seen that the compound of the present invention is cleared slowly in liver microsomes, has good stability, and has good pharmacokinetic properties.
实验例4.大鼠药代动力学(PK)研究Experimental Example 4. Rat Pharmacokinetic (PK) Study
分别通过静脉(IV)和灌胃(PO)给予雄性SD大鼠本发明的化合物,考察药代动力学特点。IV和PO的给药剂量分别是1mg/kg和5mg/kg,IV给药溶媒***为5%DMSO:5%Solutol:90%生理盐水,PO给药溶媒***为0.5%MC(甲基纤维素)。IV和PO给药后在不同时间点收集血液,血液采用K2-EDTA抗凝,离心后得到血浆样品,保存于-80℃。血浆样品经沉淀蛋白处理后进行LC-MS/MS分析。The compounds of the present invention were administered to male SD rats by intravenous (IV) and intragastric (PO), respectively, to investigate the pharmacokinetic characteristics. The doses of IV and PO are 1 mg/kg and 5 mg/kg, respectively, the IV administration vehicle system is 5% DMSO: 5% Solutol: 90% saline, and the PO administration vehicle system is 0.5% MC (methyl cellulose ). After IV and PO administration, blood was collected at different time points. The blood was anticoagulated with K2-EDTA. After centrifugation, plasma samples were obtained and stored at -80°C. Plasma samples were treated with precipitated proteins and analyzed by LC-MS/MS.
应用WinNonlin 6.3软件,采用非房室模型计算药代动力学参数,结果见表6和7。The WinNonlin 6.3 software was used to calculate the pharmacokinetic parameters using a non-compartmental model. The results are shown in Tables 6 and 7.
表6.IV给药的化合物在大鼠体内的药代动力学参数Table 6. Pharmacokinetic parameters of IV administered compounds in rats
Figure PCTCN2019121534-appb-000020
Figure PCTCN2019121534-appb-000020
表6的数据显示,通过以1mg/kg的剂量IV给药的本发明化合物C3、C5在大鼠体内具有优良的药物暴露量。The data in Table 6 shows that the compounds C3 and C5 of the present invention administered IV at a dose of 1 mg/kg have excellent drug exposure in rats.
表7.PO给药的化合物在大鼠体内的药代动力学参数Table 7. Pharmacokinetic parameters of PO-administered compounds in rats
Figure PCTCN2019121534-appb-000021
Figure PCTCN2019121534-appb-000021
表7的数据显示,通过以5mg/kg的剂量PO给药的本发明化合物C3、C5在大鼠体内具有优良的药物暴露量和生物利用度。The data in Table 7 shows that the compounds C3 and C5 of the present invention administered by PO at a dose of 5 mg/kg have excellent drug exposure and bioavailability in rats.
综合表6和表7,本发明的化合物C3、C5通过IV和PO给药在大鼠体内具有优良的血浆药物暴露量和口服生物利用度。Combining Table 6 and Table 7, the compounds C3 and C5 of the present invention have excellent plasma drug exposure and oral bioavailability in rats by IV and PO administration.
本发明的其它化合物也具有较好的AUC last值、C max值以及生物利用度,在大鼠体内具有较好的药代动力学性质。 Other compounds of the present invention also have better AUC last value, C max value and bioavailability, and have better pharmacokinetic properties in rats.
实验例5.小鼠脂肪肝模型药效试验Experimental Example 5. Drug effect test of mouse fatty liver model
试验方法experiment method
采用8-10周龄的雄性C57小鼠(购自维通利华),动物造模前按体重随机分为5组,分别是正常组(组1)、模型组(组2)、C3化合物给药组(30mg/kg)。正常组给予正常饲料,模型组和C3化合物给药组喂食MCD饲料(蛋氨酸胆碱缺乏饲料,Research Diets)。造模的同时,C3化合物组每天单次灌胃给药,连续给药28天。正常组和模型组给予0.5%MC(甲基纤维素)。详见表8。Eight to ten-week-old male C57 mice (purchased from Viton Lihua) were used. Animals were randomly divided into 5 groups according to body weight before modeling, which were normal group (group 1), model group (group 2), and C3 compound. Administration group (30mg/kg). The normal group was given normal feed, and the model group and C3 compound-administered group were fed with MCD feed (methionine choline-deficient feed, Research Diets). Simultaneously with the modeling, the C3 compound group was administered by intragastric administration once a day for 28 consecutive days. The normal group and the model group were given 0.5% MC (methyl cellulose). See Table 8 for details.
表8 分组及给药表Table 8 Grouping and administration table
Figure PCTCN2019121534-appb-000022
Figure PCTCN2019121534-appb-000022
检测指标Detection Indicator
药效指标:第28天给药后解剖动物,取肝脏HE染色,进行病理评分(评分标准参考《中国非酒精性脂肪性肝病诊疗指南》)。Drug Efficacy Index: Animals were dissected after administration on day 28, and liver HE staining was performed for pathological scoring (the scoring criteria refer to "Chinese Non-alcoholic Fatty Liver Disease Diagnosis and Treatment Guide").
药代动力学指标:第28天给药后0.5h、1h、4h,向动物取血、肝脏和小肠组织样品,使用LC-MS/MS检测药物浓度。Pharmacokinetic indicators: 0.5h, 1h, 4h after administration on day 28, blood, liver and small intestine tissue samples were taken from the animals, and the drug concentration was detected using LC-MS/MS.
试验结果test results
药效结果:由图1可见,模型组脂肪变评分显著高于正常组,说明成功获得小鼠脂肪肝模型。此外,C3化合物30mg/kg剂量组脂肪变评分与模型组相比有显著差异。Pharmacodynamic results: It can be seen from Figure 1 that the model group's steatosis score is significantly higher than that of the normal group, indicating that the mouse fatty liver model was successfully obtained. In addition, the C3 compound 30mg/kg dose group steatosis score was significantly different from the model group.
药代动力学结果:实验中获得的药代动力学数据如表9中所示。Pharmacokinetic results: The pharmacokinetic data obtained in the experiment are shown in Table 9.
表9 由C3体内药效试验获得的PK数据(30mg/kg)Table 9 PK data (30mg/kg) obtained from C3 in vivo efficacy test
样本sample 血浆plasma liver 小肠Small intestine
时间点/浓度Time point/concentration ng/mlng/ml ng/gng/g ng/gng/g
0.25h0.25h 373±242373±242 3263±14293263±1429 1115±4881115±488
1h1h 1353±2041353±204 12700±426012700±4260 4940±574940±57
4h4h 2212±4622212±462 13040±421813040±4218 6105±24716105±2471
由上表数据可见,化合物C3在肝肠均有分布,且肝肠暴露量均高于血浆,肝脏暴露量更高。It can be seen from the data in the above table that compound C3 is distributed in the liver and intestine, and the exposure of liver and intestine are higher than that of plasma, and the exposure of liver is higher.
试验结论Test Conclusions
化合物C3能够抑制MCD饲料诱导的肝脏脂肪病变,且相对于模型组有显著改善。化合物C3在肝脏分布高于血浆,表明其有效地在靶点部位富集。Compound C3 can inhibit liver fat lesions induced by MCD feed, and has a significant improvement relative to the model group. The distribution of compound C3 in the liver is higher than that in plasma, indicating that it is effectively enriched at the target site.
除本文中描述的那些实施方案外,根据前述描述,本发明的多种修改对本领域技术人员而言会是显而易见的。这样的修改也意图落入所附权利要求书的范围内。本申请中所引用的各参考文献(包括所有专利、专利申请、期刊文章、书籍及任何其它公开)均以其整体援引加入本文。In addition to those embodiments described herein, various modifications of the present invention will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference cited in this application (including all patents, patent applications, journal articles, books, and any other publications) is incorporated by reference in its entirety.

Claims (9)

  1. 通式(I)的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,Compounds of general formula (I) or their stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N-oxides, Its chemically protected form or prodrug,
    Figure PCTCN2019121534-appb-100001
    Figure PCTCN2019121534-appb-100001
    其中:among them:
    X为CH或N;并且X is CH or N; and
    R选自氢、卤素、C 1-6烷基和C 1-6卤代烷基。 R is selected from hydrogen, halogen, C 1-6 alkyl and C 1-6 haloalkyl.
  2. 权利要求1的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,其中R选自氢、氟、甲基和三氟甲基。The compound of claim 1 or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N-oxides, their chemistry A protected form or prodrug, wherein R is selected from hydrogen, fluorine, methyl and trifluoromethyl.
  3. 权利要求1或2中的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,其中所述化合物选自:The compound of claim 1 or 2 or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N-oxides , Its chemically protected form or prodrug, wherein the compound is selected from:
    Figure PCTCN2019121534-appb-100002
    Figure PCTCN2019121534-appb-100002
  4. 药物组合物,其包含至少一种权利要求1-3中任一项的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,以及一种或多种药学可接受的载体。A pharmaceutical composition comprising at least one compound according to any one of claims 1-3 or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable Salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, and one or more pharmaceutically acceptable carriers.
  5. 权利要求4的药物组合物,其为选自以下的形式:片剂、胶囊剂、锭剂、硬糖剂、散剂、喷雾剂、乳膏剂、软膏剂、栓剂、凝胶剂、糊剂、洗剂、软膏剂、水性混悬剂、可注射溶液剂、酏剂和糖浆剂。The pharmaceutical composition according to claim 4, which is in a form selected from the group consisting of tablets, capsules, lozenges, hard candy, powders, sprays, creams, ointments, suppositories, gels, pastes, lotions Agents, ointments, aqueous suspensions, injectable solutions, elixirs and syrups.
  6. 药盒,其包括:Pill box, which includes:
    a)第一容器,其包含作为第一治疗剂的至少一种权利要求1-3中任一项的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药,或作为第一药物组合物的权利要求4或5的药物组合物;a) A first container containing at least one compound according to any one of claims 1-3 or its stereoisomers, tautomers, polymorphs, solvates as a first therapeutic agent ( Such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N-oxides, their chemically protected forms or prodrugs, or the pharmaceutical composition of claim 4 or 5 as the first pharmaceutical composition;
    b)任选存在的第二容器,其包含作为第二治疗剂的至少一种其他治疗剂,或者作为第二药物组合物的包含所述其他治疗剂的药物组合物;和b) an optional second container containing at least one other therapeutic agent as a second therapeutic agent, or a pharmaceutical composition containing the other therapeutic agent as a second pharmaceutical composition; and
    c)任选存在的包装说明书。c) optional package inserts.
  7. 权利要求1-3中任一项的化合物或者其立体异构体、互变异构体、多晶型物、溶剂合物(如水合物)、药学可接受的盐、酯、代谢物、N-氧化物、其化学保护的形式或前药、或者权利要求4或5的药物组合物在制备用于预防或治疗由类法尼醇X受体介导的疾病或病症的药物中的用途。The compound of any one of claims 1-3, or its stereoisomers, tautomers, polymorphs, solvates (such as hydrates), pharmaceutically acceptable salts, esters, metabolites, N -Use of an oxide, its chemically protected form or prodrug, or the pharmaceutical composition of claim 4 or 5 for the preparation of a medicament for the prevention or treatment of a disease or condition mediated by farnesoid X receptor.
  8. 制备权利要求1-3中任一项的通式(I)的化合物的方法,所述方法包括以下步骤:A method for preparing a compound of general formula (I) according to any one of claims 1-3, the method comprising the following steps:
    Figure PCTCN2019121534-appb-100003
    Figure PCTCN2019121534-appb-100003
    其中:among them:
    Hal 1、Hal 2和Hal 3各自独立地为相同或不同的卤素,例如F、Cl、Br或I,优选为Cl或Br; Hal 1 , Hal 2 and Hal 3 are each independently the same or different halogen, such as F, Cl, Br or I, preferably Cl or Br;
    PG 1为氨基保护基,优选为叔丁氧羰基(Boc); PG 1 is an amino protecting group, preferably tert-butoxycarbonyl (Boc);
    PG 2为羧基保护基,优选为C 1-6烷基,更优选为甲基; PG 2 is a carboxyl protecting group, preferably C 1-6 alkyl, and more preferably methyl;
    Y为硼酸或硼酸酯基团,优选为-B(OH) 2或者
    Figure PCTCN2019121534-appb-100004
    Y is a boric acid or borate group, preferably -B(OH) 2 or
    Figure PCTCN2019121534-appb-100004
    其余基团如权利要求1或2中所定义;The remaining groups are as defined in claim 1 or 2;
    各步骤的反应条件如下:The reaction conditions of each step are as follows:
    步骤A:使化合物IN-1与化合物IN-2反应以得到化合物IN-3;Step A: reacting compound IN-1 with compound IN-2 to obtain compound IN-3;
    步骤B:移除化合物IN-3中的PG 1基团,以得到化合物IN-4; Step B: Remove the PG 1 group in compound IN-3 to obtain compound IN-4;
    步骤C:使化合物IN-4与化合物IN-a反应以得到化合物IN-5;Step C: reacting compound IN-4 with compound IN-a to obtain compound IN-5;
    步骤D:使化合物IN-5与化合物IN-b反应以得到化合物IN-6;以及Step D: reacting compound IN-5 with compound IN-b to obtain compound IN-6; and
    步骤E:移除化合物IN-6中的PG 2基团,以得到通式(I)的化合物。 Step E: Remove the PG 2 group in compound IN-6 to obtain the compound of general formula (I).
  9. 通式(IN-6)的化合物或其药学上可接受的盐,A compound of general formula (IN-6) or a pharmaceutically acceptable salt thereof,
    Figure PCTCN2019121534-appb-100005
    Figure PCTCN2019121534-appb-100005
    其中各基团如权利要求1、2或8中所定义;Where each group is as defined in claims 1, 2 or 8;
    所述化合物优选自:The compound is preferably selected from:
    Figure PCTCN2019121534-appb-100006
    Figure PCTCN2019121534-appb-100006
    Figure PCTCN2019121534-appb-100007
    Figure PCTCN2019121534-appb-100007
PCT/CN2019/121534 2018-12-07 2019-11-28 Isoxazole derivative, preparation method therefor and use thereof WO2020114307A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201980066095.8A CN112805279B (en) 2018-12-07 2019-11-28 Isoxazole derivative, preparation method and application thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201811492827 2018-12-07
CN201811492827.3 2018-12-07

Publications (1)

Publication Number Publication Date
WO2020114307A1 true WO2020114307A1 (en) 2020-06-11

Family

ID=70975172

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/121534 WO2020114307A1 (en) 2018-12-07 2019-11-28 Isoxazole derivative, preparation method therefor and use thereof

Country Status (2)

Country Link
CN (1) CN112805279B (en)
WO (1) WO2020114307A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021144330A1 (en) 2020-01-15 2021-07-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of fxr agonists for treating an infection by hepatitis d virus
WO2022068815A1 (en) * 2020-09-30 2022-04-07 中国科学院上海药物研究所 Fxr small-molecule agonist, and preparation method therefor and use thereof
WO2022152770A1 (en) 2021-01-14 2022-07-21 Enyo Pharma Synergistic effect of a fxr agonist and ifn for the treatment of hbv infection
WO2022229302A1 (en) 2021-04-28 2022-11-03 Enyo Pharma Strong potentiation of tlr3 agonists effects using fxr agonists as a combined treatment
EP4245365A1 (en) * 2022-03-16 2023-09-20 Cascade Pharmaceuticals, Inc. A fxr small molecule agonist, the preparation and use thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012087521A1 (en) * 2010-12-20 2012-06-28 Irm Llc Compositions and methods for modulating farnesoid x receptors
WO2018067704A1 (en) * 2016-10-04 2018-04-12 Enanta Pharmaceuticals, Inc. Isoxazole analogs as fxr agonists and methods of use thereof
WO2019120088A1 (en) * 2017-12-22 2019-06-27 四川科伦博泰生物医药股份有限公司 Isoxazole derivative, preparation method therefor, and use thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012087521A1 (en) * 2010-12-20 2012-06-28 Irm Llc Compositions and methods for modulating farnesoid x receptors
WO2018067704A1 (en) * 2016-10-04 2018-04-12 Enanta Pharmaceuticals, Inc. Isoxazole analogs as fxr agonists and methods of use thereof
WO2019120088A1 (en) * 2017-12-22 2019-06-27 四川科伦博泰生物医药股份有限公司 Isoxazole derivative, preparation method therefor, and use thereof

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021144330A1 (en) 2020-01-15 2021-07-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of fxr agonists for treating an infection by hepatitis d virus
WO2022068815A1 (en) * 2020-09-30 2022-04-07 中国科学院上海药物研究所 Fxr small-molecule agonist, and preparation method therefor and use thereof
WO2022152770A1 (en) 2021-01-14 2022-07-21 Enyo Pharma Synergistic effect of a fxr agonist and ifn for the treatment of hbv infection
WO2022229302A1 (en) 2021-04-28 2022-11-03 Enyo Pharma Strong potentiation of tlr3 agonists effects using fxr agonists as a combined treatment
EP4245365A1 (en) * 2022-03-16 2023-09-20 Cascade Pharmaceuticals, Inc. A fxr small molecule agonist, the preparation and use thereof

Also Published As

Publication number Publication date
CN112805279A (en) 2021-05-14
CN112805279B (en) 2023-09-12

Similar Documents

Publication Publication Date Title
WO2020114307A1 (en) Isoxazole derivative, preparation method therefor and use thereof
CN111263759B (en) Isoxazole derivative and preparation method and application thereof
TWI731392B (en) Fxr (nr1h4) modulating compounds
EP3233831B1 (en) Novel fxr (nr1h4) modulating compounds
WO2018133730A1 (en) Heterocyclic compound, preparation method and use therefor
TWI643855B (en) Fxr (nr1h4) modulating compounds
WO2018059314A1 (en) Azabicycle derivatives and preparation method and use thereof
TWI733307B (en) Fxr (nr1h4) modulating compounds
KR101712466B1 (en) Novel fxr(nr 1h4) binding and activity modulating compounds
CN112955448B (en) Aromatic ring or aromatic heterocyclic compound and preparation method and medical application thereof
CN111868056B (en) 1,2, 4-oxadiazole compound and preparation method and medical application thereof
CN112812114A (en) Isoxazole derivative, pharmaceutical composition containing isoxazole derivative, and preparation method and application of isoxazole derivative
WO2020249064A1 (en) Compounds for modulating fxr

Legal Events

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

Ref document number: 19892293

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19892293

Country of ref document: EP

Kind code of ref document: A1