WO2020094111A1 - Rho相关蛋白激酶抑制剂、包含其的药物组合物及其用途 - Google Patents

Rho相关蛋白激酶抑制剂、包含其的药物组合物及其用途 Download PDF

Info

Publication number
WO2020094111A1
WO2020094111A1 PCT/CN2019/116453 CN2019116453W WO2020094111A1 WO 2020094111 A1 WO2020094111 A1 WO 2020094111A1 CN 2019116453 W CN2019116453 W CN 2019116453W WO 2020094111 A1 WO2020094111 A1 WO 2020094111A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
ring
compound
group
pharmaceutically acceptable
Prior art date
Application number
PCT/CN2019/116453
Other languages
English (en)
French (fr)
Inventor
赵焰平
王红军
李功
姜媛媛
李想
刘彬
刘凯
仲伟婷
李发洁
赵静
庞建梅
周丽莹
Original Assignee
北京泰德制药股份有限公司
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 北京泰德制药股份有限公司 filed Critical 北京泰德制药股份有限公司
Priority to CN201980073246.2A priority Critical patent/CN112969694B/zh
Priority to US17/292,036 priority patent/US20220002266A1/en
Publication of WO2020094111A1 publication Critical patent/WO2020094111A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • the present invention relates to Rho-related protein kinase inhibitors, pharmaceutical compositions containing the same, and their use for preventing or treating Rho-related protein kinase (ROCK) -mediated diseases.
  • ROCK Rho-related protein kinase
  • Rho-related protein kinase is a serine / threonine protein kinase of the AGC kinase family, which includes two subtypes, ROCK1 and ROCK2.
  • ROCK1 and ROCK2 are differentially expressed and regulated in specific tissues.
  • ROCK1 is generally expressed at relatively high levels, while ROCK2 is preferentially expressed in the heart, brain, and skeletal muscle.
  • ROCK is the first downstream effector of Rho protein to be discovered. It achieves its biological function by phosphorylating downstream effector proteins (MLC, Lin-11, Isl-1, LIMK, ERM, MARCKS, CRMP-2, etc.) .
  • MLC phosphorylating downstream effector proteins
  • Fasudil Fasudil
  • the present invention provides compounds used as ROCK (preferably ROCK2) inhibitors, which have excellent inhibitory activity against ROCK (preferably ROCK2), good selectivity (higher selectivity to ROCK2 relative to ROCK1), and better Physicochemical properties (e.g. solubility, physical and / or chemical stability), improved pharmacokinetic properties (e.g. improved bioavailability, suitable half-life and duration of action), improved safety (lower toxicity And / or less side effects, wider treatment window) and other more excellent properties.
  • ROCK preferably ROCK2
  • good selectivity higher selectivity to ROCK2 relative to ROCK1
  • better Physicochemical properties e.g. solubility, physical and / or chemical stability
  • improved pharmacokinetic properties e.g. improved bioavailability, suitable half-life and duration of action
  • improved safety lower toxicity And / or less side effects, wider treatment window
  • One aspect of the present invention provides compounds or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites, or prodrugs, wherein The compound has the structure of formula (I):
  • Ring C is It is connected to Y by one of the two positions marked by * or **, and to the ring B at the other position;
  • X is selected from CR 8 and N;
  • R 5 and R 6 is independently selected from H, C 1-6 alkyl, C 3-10 cycloalkyl, 3-10 membered heterocyclyl, C 6-10 aryl, 5-14 membered Heteroaryl and C 6-12 aralkyl; or when R 5 and R 6 are attached to the same nitrogen atom, R 5 and R 6 together with the atoms to which they are attached optionally form a 3-12 membered heterocyclic ring or hetero Aromatic ring
  • n is independently an integer of 0, 1, 2 or 3 at each occurrence;
  • n is an integer of 0, 1, 2, 3, or 4;
  • i is an integer of 0, 1, or 2.
  • Another aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a prophylactically or therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt, ester, stereoisomer, polymorph, solvate, N- Oxides, isotope-labeled compounds, metabolites or prodrugs, and one or more pharmaceutically acceptable carriers.
  • the pharmaceutical composition is preferably a solid preparation, semi-solid preparation, liquid preparation, or gaseous preparation.
  • Another aspect of the invention provides compounds of the invention or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or pro Use of the medicament or the pharmaceutical composition of the present invention in the preparation of a medicament for use as a Rho-related protein kinase (ROCK) inhibitor, preferably a selective ROCK2 inhibitor.
  • ROCK Rho-related protein kinase
  • Another aspect of the invention provides compounds of the invention or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or pro
  • the medicine or the pharmaceutical composition of the present invention is used as a Rho-related protein kinase (ROCK) inhibitor, preferably a selective ROCK2 inhibitor.
  • ROCK Rho-related protein kinase
  • Another aspect of the present invention provides a method for preventing or treating Rho-related protein kinase (ROCK) -mediated diseases, which method comprises administering an effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof to an individual in need thereof , Esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs or the pharmaceutical composition of the present invention.
  • ROCK Rho-related protein kinase
  • alkylene means a saturated divalent hydrocarbon group, preferably a saturated divalent hydrocarbon group having 1, 2, 3, 4, 5, or 6 carbon atoms, such as methylene, ethylene, Propylene or butylene.
  • alkyl is defined as a linear or branched saturated aliphatic hydrocarbon.
  • the alkyl group has 1 to 12, for example, 1 to 6 carbon atoms.
  • C 1-6 alkyl refers to a linear or branched group of 1 to 6 carbon atoms (eg, methyl, ethyl, n-propyl, isopropyl, n-butyl Group, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl or n-hexyl), which is optionally substituted by 1 or more (such as 1 to 3) suitable substituents
  • halogen substitution in this case the group is called "haloalkyl” (eg CH 2 F, CHF 2 , CF 3 , CCl 3 , C 2 F 5 , C 2 Cl 5 , CH 2
  • C 1-4 alkyl refers to a linear or branched aliphatic hydrocarbon chain of 1 to 4 carbon atoms (ie, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, Sec-butyl or tert-butyl).
  • alkenyl means a linear or branched monovalent hydrocarbon group that contains a double bond and has 2-6 carbon atoms (“C 2-6 alkenyl”).
  • the alkenyl group is, for example, vinyl, 1-propenyl, 2-propenyl, 2-butenyl, 3-butenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 2 -Hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 2-methyl-2-propenyl and 4-methyl-3-pentenyl.
  • the compound of the present invention contains an alkenylene group, the compound may exist in pure E (enthafen) form, pure Z (zusammen) form, or any mixture thereof.
  • alkynyl refers to a monovalent hydrocarbon group containing one or more triple bonds, which preferably has 2, 3, 4, 5, or 6 carbon atoms, such as ethynyl or propynyl.
  • cycloalkyl refers to a saturated monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (eg, monocyclic, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl) , Cyclooctyl, cyclononyl, or bicyclic, including spiro ring, fused or bridged systems (such as bicyclic [1.1.1] pentyl, bicyclic [2.2.1] heptyl, bicyclic [3.2.1] octyl Or bicyclo [5.2.0] nonyl, decahydronaphthyl, etc.)), which is optionally substituted with 1 or more (such as 1 to 3) suitable substituents.
  • monocyclic such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl
  • the cycloalkyl group has 3 to 15 carbon atoms.
  • C 3-6 cycloalkyl refers to a saturated monocyclic or polycyclic (such as bicyclic) hydrocarbon ring of 3 to 6 ring-forming carbon atoms (eg, cyclopropyl, cyclobutyl, cyclopentyl, or cyclic Hexyl), which is optionally substituted with 1 or more (such as 1 to 3) suitable substituents, for example methyl-substituted cyclopropyl.
  • cyclohydrocarbylene refers to having, for example, 3-10 (suitably 3-8, more suitably 3-6) ring carbons Atomic saturated (ie, “cycloalkylene” and “cycloalkyl”) or unsaturated (ie, having one or more double and / or triple bonds in the ring) monocyclic or polycyclic hydrocarbon ring, which Including but not limited to (cyclo) propyl (cyclo), (cyclo) cyclobutyl (cyclo), (cyclo) cyclopentyl (cyclo), (cyclo) cyclohexyl (cyclo), (cyclo) cycloheptyl ( Ring), (A) cyclooctyl (A), (A) cyclononyl (A), (A) cyclohexenyl (A), etc.
  • Atomic saturated ie, "cycloalkylene” and "cycloalkyl”
  • unsaturated ie, having one or more double and / or triple bonds in
  • heterocyclyl refers to having, for example, 3-10 (suitably 3-8, more suitably 3-6) Ring atoms, at least one of which is a heteroatom selected from N, O, and S and the remaining ring atoms are saturated (ie, heterocycloalkyl) or partially unsaturated (ie, have one or more within the ring) Double bond and / or triple bond) cyclic group.
  • 3-10 membered (sub) heterocyclic (radical) has 2-9 (eg 2, 3, 4, 5, 6, 7, 8 or 9) ring carbon atoms and is independently selected from N , O and S, one or more (for example 1, 2, 3 or 4) saturated or partially unsaturated (sub) heterocyclic (radical) heteroatoms.
  • heterocyclylene and heterocycle include, but are not limited to: (ethylene) ethylene oxide, (aziridinyl), (azetidinyl), (oxygen) Heterobutyl (oxetanyl), (hydrogen) tetrahydrofuranyl, (hydrogen) dioxolyl (dioxolinyl), (hydrogen) pyrrolidinyl, (hydrogen) pyrrolidone, (hydrogen) imidazolidinyl ) Pyrazolidinyl, (pyridinyl), (pyridinyl), tetrahydropyranyl, (piperidinyl), morpholinyl, (diithianyl), (di) Thiomorpholinyl, (piperidinyl) or (trithianyl).
  • the groups also encompass bicyclic systems including spirocyclic, fused or bridged systems (such as 8-azaspiro [4.5] decane, 3,9-diazaspiro [5.5] undecane, 2-nitrogen Heterobicyclo [2.2.2] octane, etc.).
  • Heterocyclylene and heterocycle (radical) can be optionally substituted with one or more (eg 1, 2, 3 or 4) suitable substituents.
  • (arylene) and “aromatic ring” refer to an all-carbon monocyclic or fused ring polycyclic aromatic group having a conjugated pi electron system.
  • C 6-10 (arylene)” and “C 6-10 aromatic ring” mean an aromatic group containing 6 to 10 carbon atoms, such as (phenylene) (Benzene ring) or (methylene) naphthyl (naphthalene ring).
  • (Arylene) and aromatic rings are optionally substituted with 1 or more (such as 1 to 3) suitable substituents (e.g. halogen, -OH, -CN, -NO 2 , C 1-6 alkyl, etc.) .
  • heteroaryl and “heteroaryl ring” refer to monocyclic, bicyclic, or tricyclic aromatic ring systems having 5, 6, 8, 9, 10, 11, 12, 13 or 14 ring atoms, especially 1 or 2 or 3 or 4 or 5 or 6 or 9 or 10 carbon atoms, and it contains at least one heteroatom which may be the same or different (the heteroatom is for example oxygen, nitrogen Or sulfur), and, in addition, may be benzo-fused in each case.
  • “(heteroaryl)" or “heteroaryl ring” is selected from (()) thienyl, (()) furanyl, (()) pyrrolyl, (()) oxazolyl, (()) thiazolyl, (A) imidazolyl, (A) pyrazolyl, (A) isoxazolyl, (A) isothiazolyl, (A) oxadiazolyl, (A) triazolyl, (A) thiadiazolyl Etc., and their benzo derivatives; or (pyridinyl), (pyridazinyl), (pyridinyl), (pyridinyl), (tri) triazinyl, etc., and their benzo derivative.
  • aralkyl preferably refers to an aryl or heteroaryl substituted alkyl, wherein the aryl, heteroaryl, and alkyl are as defined herein.
  • the aryl group may have 6-14 carbon atoms
  • the heteroaryl group may have 5-14 ring atoms
  • the alkyl group may have 1-6 carbon atoms.
  • Exemplary aralkyl groups include but are not limited to benzyl, phenylethyl, phenylpropyl, phenylbutyl.
  • halo or halogen group is defined to include F, Cl, Br or I.
  • substituted means that one or more (eg, one, two, three, or four) hydrogens on the designated atom are replaced by the choice of the indicated group, provided that the designated atom is not exceeded in the current Normal valence in the case and the substitution forms a stable compound. Combinations of substituents and / or variables are only allowed when such combinations form stable compounds.
  • the substituent may be (1) unsubstituted or (2) substituted. If the carbon of the substituent is described as optionally substituted by one or more of the list of substituents, one or more hydrogens on the carbon (to the extent of any hydrogen present) can be independently and / or together Selected optional substituents are substituted. If the nitrogen of the substituent is described as optionally substituted by one or more of the list of substituents, then one or more hydrogens on the nitrogen (to the extent of any hydrogen present) can each be independently selected Substitution.
  • each substituent is selected independently of the other. Therefore, each substituent may be the same as or different from another (other) substituent.
  • one or more means 1 or more than 1 under reasonable conditions, such as 2, 3, 4, 5, or 10.
  • the point of attachment of a substituent can be from any suitable position of the substituent.
  • the present invention also includes all pharmaceutically acceptable isotope-labeled compounds, which are the same as the compounds of the present invention, except that one or more atoms have the same atomic number but the atomic mass or mass number is different from the atomic mass predominant in nature Or mass atom replacement.
  • isotopes suitable for inclusion in the compounds of the invention include, but are not limited to, isotopes of hydrogen (eg, deuterium ( 2 H), tritium ( 3 H)); isotopes of carbon (eg, 11 C, 13 C, and 14 C) ; Isotopes of chlorine (eg 36 Cl); Isotopes of fluorine (eg 18 F); Isotopes of iodine (eg 123 I and 125 I); Isotopes of nitrogen (eg 13 N and 15 N); Isotopes of oxygen (eg 15 O) , 17 O and 18 O); phosphorus isotopes (such as 32 P); and sulfur isotopes (such as 35 S).
  • isotopes of hydrogen eg, deuterium ( 2 H), tritium ( 3 H)
  • isotopes of carbon eg, 11 C, 13 C, and 14 C
  • Isotopes of chlorine eg 36 Cl
  • Certain isotopically-labeled compounds of the invention can be used in drug and / or substrate tissue distribution studies (such as analysis).
  • the radioisotopes tritium (ie 3 H) and carbon-14 (ie 14 C) are particularly useful for this purpose because they are easy to incorporate and easy to detect.
  • Substitution with positron emitting isotopes eg, 11 C, 18 F, 15 O, and 13 N
  • PET positron emission tomography
  • Isotope-labeled compounds of the present invention can be prepared by methods similar to those described in the accompanying routes and / or examples and preparations by using appropriate isotope-labeled reagents instead of the previously employed non-labeled reagents.
  • the pharmaceutically acceptable solvates of the present invention include those in which the crystallization solvent can be replaced by isotopes, for example, D 2 O, acetone-d 6 or DMSO-d 6 .
  • stereoisomer means an isomer formed due to at least one asymmetric center. In compounds with one or more (eg one, two, three or four) asymmetric centers, it can produce racemic mixtures, single enantiomers, diastereomer mixtures and individual Diastereomers. Specific individual molecules may also exist as geometric isomers (cis / trans). Similarly, the compounds of the present invention may exist as a mixture of two or more structurally different forms in rapid equilibrium (commonly referred to as tautomers). Representative examples of tautomers include keto-enol tautomers, phenol-ketone tautomers, nitroso-oxime tautomers, imine-enamine tautomers Wait. It is to be understood that the scope of this application covers all such in any ratio (eg 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99 %) Isomers or mixtures thereof.
  • Solid lines can be used in this article Solid wedge Or virtual wedge
  • the chemical bonds of the compounds of the invention are depicted.
  • the use of a solid line to depict the bond to an asymmetric carbon atom is intended to indicate that all possible stereoisomers at that carbon atom are included (eg, specific enantiomers, racemic mixtures, etc.).
  • the use of solid or imaginary wedges to depict bonds to asymmetric carbon atoms is intended to indicate the presence of the stereoisomers shown.
  • real and virtual wedges are used to define relative stereochemistry, rather than absolute stereochemistry.
  • the compounds of the present invention are intended to be stereoisomers (which include cis and trans isomers, optical isomers (such as R and S enantiomers), diastereomers, Geometric isomers, rotamers, conformers, atropisomers, and mixtures thereof exist.
  • the compounds of the present invention can exhibit more than one type of isomerism and consist of mixtures thereof (e.g. racemic mixtures and diastereomeric pairs).
  • the present invention encompasses all possible crystalline forms or polymorphs of the compounds of the present invention, which may be a single polymorph or a mixture of more than one polymorph in any ratio.
  • compositions of the invention may exist in free form for therapy, or where appropriate, in the form of pharmaceutically acceptable derivatives.
  • pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable salts, esters, solvates, N-oxides, metabolites or prodrugs, which are given to patients in need thereof After the medicine, the compound of the present invention or its metabolite or residue can be provided directly or indirectly. Therefore, when referring to the "compound of the present invention" herein, it is also intended to cover the above-mentioned various derivative forms of the compound.
  • the pharmaceutically acceptable salts of the compounds of the present invention include acid addition salts and base addition salts.
  • Suitable acid addition salts are formed from acids that form pharmaceutically acceptable salts. Examples include acetate, adipate, aspartate, benzoate, benzenesulfonate, bicarbonate / carbonate, bisulfate / sulfate, borate, camphorsulfonate , Citrate, cyclamate, ethanedisulfonate, ethanesulfonate, formate, fumarate, glucoheptonate, gluconate, glucuronate, hexafluorophosphate Salt, hydrobenzoate, hydrochloride / chloride, hydrobromide / bromide, hydroiodide / iodide, isethionate, lactate, malate, maleic acid Salt, malonate, methanesulfonate, methylsulfate, naphthylate, 2-naphthalenesulfonate, nicotinate, nitrate, orotate, ox
  • Suitable base addition salts are formed from bases that form pharmaceutically acceptable salts. Examples include aluminum salt, arginine salt, benzathine penicillin salt, calcium salt, choline salt, diethylamine salt, diethanolamine salt, glycine salt, lysine salt, magnesium salt, meglumine salt, ethanolamine salt, Potassium, sodium, tromethamine and zinc salts.
  • ester means an ester derived from each compound of the general formula in this application, which includes physiologically hydrolyzable esters (which can be hydrolyzed under physiological conditions to release the free acid or alcohol form of the present invention) Compound).
  • the compound of the present invention may itself be an ester.
  • the compound of the present invention may exist in the form of a solvate (preferably 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.
  • Synthetic methods for preparing N-oxides of heterocyclic 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 (MCPBA), hydrogen peroxide, alkyl groups Hydrogen peroxides such as tert-butyl hydroperoxide, sodium perborate and dioxiranes such as dimethyl diepoxide oxidize heterocycles and tertiary amines.
  • MCPBA m-chloroperoxybenzoic acid
  • Hydrogen peroxides such as tert-butyl hydroperoxide
  • sodium perborate and dioxiranes such as dimethyl diepoxide oxidize heterocycles and tertiary amines.
  • metabolites of the compounds of the present invention that is, substances formed in the body when the compounds of the present invention are administered. Such products can be produced, for example, by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymatic hydrolysis, etc. of the administered compound. Accordingly, the present invention includes metabolites of the compounds of the present invention, including compounds prepared by contacting the compounds of the present invention with mammals for a time sufficient to produce their metabolites.
  • the present invention further includes within its scope prodrugs of the compounds of the present invention, which are certain derivatives of the compounds of the present invention which may themselves have less or no pharmacological activity when administered into or on the body It can be converted into a compound of the present invention having a desired activity by, for example, hydrolytic cleavage. 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.
  • prodrugs of the compounds of the present invention which are certain derivatives of the compounds of the present invention which may themselves have less or no pharmacological activity when administered into or on the body It can be converted into a compound of the present invention having a desired activity by, for example, hydrolytic cleavage. 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.
  • prodrugs of the present invention can be used, for example, by those skilled in the art known as "pro-moiety” (such as “Design of Prodrugs", described in H. Bundgaard (Elsevier, 1985)) " They are prepared in place of the appropriate functional groups present in the compounds of the invention.
  • the present invention also encompasses compounds of the present invention containing protecting groups.
  • protecting groups 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 by conventional protecting groups, such as those described in T. W. Greene & P. G. M. Wuts, Protective Groups Organic Synthesis, John Wiley & Sons, 1991, which references are incorporated herein by reference. Using methods known in the art, the protecting group can be removed at an appropriate subsequent stage.
  • the present invention provides compounds or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs , wherein the compound has the structure of formula (I):
  • Ring C is It is connected to Y by one of the two positions marked by * or **, and to the ring B at the other position;
  • X is selected from CR 8 and N;
  • R 5 and R 6 is independently selected from H, C 1-6 alkyl, C 3-10 cycloalkyl, 3-10 membered heterocyclyl, C 6-10 aryl, 5-14 membered Heteroaryl and C 6-12 aralkyl; or when R 5 and R 6 are attached to the same nitrogen atom, R 5 and R 6 together with the atoms to which they are attached optionally form a 3-12 membered heterocyclic ring or hetero Aromatic ring
  • n is independently an integer of 0, 1, 2 or 3 at each occurrence;
  • n is an integer of 0, 1, 2, 3, or 4;
  • i is an integer of 0, 1, or 2.
  • the present invention provides compounds or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs ,
  • the present invention provides compounds or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs ,among them
  • the above group is connected to ring C through one of the two positions marked by # or ##, and the other position is connected to R 1 ,
  • j 0, 1, 2, 3 or 4;
  • Select from The above group is connected to ring C through one of the two positions marked by # or ##, and the other position is connected to R 1 .
  • the present invention provides compounds or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs ,
  • ring C is selected from The above group is connected to Y through one of two positions marked with * or **, and the other position is connected to ring B.
  • the present invention provides compounds or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs ,
  • the present invention provides compounds or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs ,
  • the present invention provides compounds or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs ,among them Select from
  • the present invention provides compounds or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs , Where R 1 is selected from
  • the present invention provides compounds or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs , Where the compound has the structure of any of the following formulas:
  • the present invention encompasses compounds obtained in any combination of various embodiments.
  • the present invention provides compounds or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs , Where the compound has the following structure:
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a prophylactically or therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt, ester, stereoisomer, polymorph, solvate, N-oxides, isotopically labeled compounds, metabolites or prodrugs, and one or more pharmaceutically acceptable carriers
  • the pharmaceutical composition is preferably a solid preparation, semi-solid preparation, liquid preparation, or gaseous preparation.
  • the pharmaceutical composition may further include one or more other therapeutic agents.
  • the present invention provides compounds of the present invention or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites Or a prodrug or the pharmaceutical composition of the present invention for use in the preparation of a medicament for use as a Rho-related protein kinase (ROCK) inhibitor, preferably a selective ROCK2 inhibitor.
  • a Rho-related protein kinase (ROCK) inhibitor preferably a selective ROCK2 inhibitor.
  • the present invention provides compounds of the present invention or pharmaceutically acceptable salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites Or a prodrug or the pharmaceutical composition of the present invention, which is used as a Rho-related protein kinase (ROCK) inhibitor, preferably a selective ROCK2 inhibitor.
  • ROCK Rho-related protein kinase
  • the present invention provides a method of preventing or treating Rho-related protein kinase (ROCK) -mediated diseases, the method comprising administering to an individual in need thereof an effective amount of a compound of the present invention or a pharmaceutically acceptable thereof Salts, esters, stereoisomers, polymorphs, solvates, N-oxides, isotopically labeled compounds, metabolites or prodrugs or pharmaceutical compositions of the present invention.
  • ROCK Rho-related protein kinase
  • the Rho-related protein kinase (ROCK) -mediated diseases include autoimmune diseases (which include rheumatoid arthritis, systemic lupus erythematosus (SLE; lupus), psoriasis, Crohn Disease, atopic dermatitis, eczema, or graft-versus-host disease (GVHD); cardiovascular disorders (including hypertension, atherosclerosis, restenosis, cardiac hypertrophy, cerebral ischemia, cerebral vasospasm, or erectile dysfunction ); Inflammation (which includes asthma, cardiovascular inflammation, ulcerative colitis, or kidney inflammation); central nervous system disorders (which include neuronal degeneration or spinal cord injury; the central nervous system disorders are preferably Huntington's disease, Par Kimson's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS) or multiple sclerosis); arterial thrombotic disorders (which include platelet aggregation or leukocyte aggregation); fibrotic disorders
  • the Rho-related protein kinase (ROCK) -mediated diseases include lupus nephritis, atherosclerosis, rheumatoid arthritis (RA), hemangioma, angiofibroma, pulmonary fibrosis, psoriasis Disease, corneal transplant rejection, insulin-dependent diabetes, multiple sclerosis, myasthenia gravis, Crohn's disease, autoimmune nephritis, primary biliary cirrhosis, acute pancreatitis, foreign body rejection, allergic inflammation, Contact dermatitis, delayed-type hypersensitivity, inflammatory bowel disease, septic shock, osteoporosis, osteoarthritis, neuronal inflammation, Osler-Weber syndrome, restenosis, fungal infection, parasitic infection, and viral infection.
  • RA rheumatoid arthritis
  • hemangioma angiofibroma
  • pulmonary fibrosis psoriasis Disease
  • corneal transplant rejection insulin
  • “Pharmaceutically acceptable carrier” in the present invention refers to a diluent, adjuvant, excipient or vehicle administered together with a therapeutic agent, and it is suitable for contact with humans and / or within the scope of reasonable medical judgment The tissues of other animals 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 compositions of the present invention include, but are not limited to, sterile liquids, such as water and oils, including those of petroleum, animal, plant, or synthetic origin, such as peanut oil, soybean oil, minerals Oil, sesame oil, etc.
  • sterile liquids such as water and oils, including those of petroleum, animal, plant, or synthetic origin, such as peanut oil, soybean oil, minerals 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 a suitable route, for example, by injection (eg, intravenous, intraarterial, subcutaneous, intraperitoneal, intramuscular injection, including drip infusion) or percutaneous administration; or by oral, buccal, or Nasal, transmucosal, topical, in the form of ophthalmic preparations 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.
  • an effective amount refers to an amount of a compound that, after being administered, will alleviate one or more symptoms of the condition being treated to some extent.
  • the dosage regimen can be adjusted to provide the best 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 foregoing 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, more preferably 1-150 mg, particularly preferably 1-50 mg, such as 1.5 mg, 2mg, 4mg, 10mg, 25mg, etc.
  • treating means reversing, alleviating, or inhibiting the development of a disorder or condition to which such terms apply, or the progression of one or more symptoms of such disorder or condition, or Preventing such a condition or condition or one or more symptoms of such condition or condition.
  • “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” in the present invention 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.).
  • the pharmaceutical composition of the present invention may also include one or more additional therapeutic or prophylactic agents.
  • the structure of the compound was confirmed by nuclear magnetic resonance spectroscopy ( 1 H NMR) or mass spectrometry (MS).
  • the chemical shift ( ⁇ ) is given in parts per million (ppm).
  • the measurement of 1 HNMR was performed on Bruker BioSpin GmbH 400 nuclear magnetic instrument.
  • the test solvent was deuterated methanol (CD 3 OD), deuterated chloroform (CDCl 3 ) or hexadeuterated dimethyl sulfoxide (DMSO-d 6 ) Labeled as tetramethylsilane (TMS).
  • Thin layer chromatography was carried out using Huanghai brand HSGF254 (5 ⁇ 20cm) silica gel plates, and thin layer preparative chromatography was carried out using GF254 (0.4-0.5nm) silica gel plates produced in Yantai.
  • the reaction was detected by thin layer chromatography (TLC) or LC-MS.
  • TLC thin layer chromatography
  • the developing system used included dichloromethane and methanol systems, n-hexane and ethyl acetate systems, and petroleum ether and ethyl acetate systems, depending on the compounds to be separated. Different polarities adjust the developing agent system (by adjusting the volume ratio of the solvent or adding triethylamine, etc.).
  • the eluent system includes dichloromethane and methanol system and n-hexane and ethyl acetate system.
  • the eluent system is adjusted according to the polarity of the compound to be separated (by adjusting the volume ratio of the solvent or adding triethylamine, etc. get on).
  • reaction temperature is room temperature (20 ° C to 30 ° C).
  • the reagents used in the examples were purchased from companies such as Acros Organics, Aldrich Chemical Company or Shanghai Bide Pharmaceutical Technology Co., Ltd.
  • Example 4 5- (5-fluoro-4-((1-oxo-1,2-dihydroisoquinolin-6-yl) amino) pyrimidin-2-yl) isoindolin-2-carboxylic acid 3,3-difluorocyclobutyl ester (C10) and 6-((5-fluoro-2- (2- (4,4,4-trifluoro-3-methylbutyryl) isoindoline-5- Of phenyl) pyrimidin-4-yl) amino) isoquinolin-1 (2H) -one (C37)
  • Example 6 2- (5-fluoro-4-((1-oxo-1,2-dihydroisoquinolin-6-yl) amino) pyrimidin-2-yl) -6,7-dihydrothiophene P- [3,2-c] pyridine-5 (4H) -carboxylic acid 3,3-difluorocyclobutyl ester (C27) and 6-((5-fluoro-2- (5- (4,4,4-tri Fluoro-3-methylbutyryl) -4,5,6,7-tetrahydrothieno [3,2-c] pyridin-2-yl) pyrimidin-4-yl) amino) isoquinoline-1 (2H )-Preparation of ketone (C47)
  • reaction solution was diluted with water (20 mL), extracted with ethyl acetate (15 mL ⁇ 2), the organic layers were combined, washed with water (20 mL), saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and reduced After concentration by pressure, the residue was purified by Flash column chromatography (methanol / dichloromethane, 0 to 3%) to obtain compound C38-3 (60 mg, yellow solid, 12.3% yield).
  • Kinase IC 50 determination was performed using a commercial CISBIO kinase detection kit HTRF KinEASE-STK S2 kit (62ST2PEC).
  • the kinase ROCK2 (01-119) used in the reaction was purchased from Carna Biosciences.
  • a 10000nM compound solution was prepared using 1 ⁇ kinase buffer with a DMSO concentration of 2.5%, and the compound was serially diluted using the above-mentioned kinase buffer containing DMSO to obtain 9 test compound solutions with different concentrations.
  • positive wells containing all reagents except compounds
  • negative wells containing all reagents except test compounds and kinases
  • 4 ⁇ L of the test compound solution was added to all reaction wells, and 4 ⁇ L of 2.5% DMSO solution was added to the control wells.
  • ROCK2 IC 50 (nM) ROCK1 IC 50 (nM) C1 129 > 10000 C3 287 > 10000 C5 13 > 10000 C6 76 > 10000 C8 16 > 10000 C9 4 > 10000 C10 11 > 10000 C11 34 > 10000 C12 17 > 10000 C13 38 > 10000 C14 28 > 10000 C15 79 > 10000 C16 19 > 10000 C17 421 > 10000 C18 31 > 10000
  • test compounds IC 50 value was significantly lower than that of ROCK2 value ROCK1 an IC 50, which indicates that the compounds of the present invention has good selectivity for ROCK2.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

式(I)的Rho相关蛋白激酶抑制剂、包含其的药物组合物、及其用于预防或治疗Rho相关蛋白激酶(ROCK)介导的疾病的用途。

Description

Rho相关蛋白激酶抑制剂、包含其的药物组合物及其用途 发明领域
本发明涉及Rho相关蛋白激酶抑制剂、包含其的药物组合物及其用于预防或治疗Rho相关蛋白激酶(ROCK)介导的疾病的用途。
发明背景
Rho相关蛋白激酶(ROCK)为AGC激酶家族的丝氨酸/苏氨酸蛋白激酶,其包括两种亚型,ROCK1和ROCK2。ROCK1和ROCK2在特定组织中被差异性地表达和调节。例如ROCK1以相对高的水平普遍表达,而ROCK2在心脏、脑和骨骼肌中优先表达。ROCK是第一个被发现的Rho蛋白下游效应因子,其通过磷酸化下游的效应蛋白(MLC、Lin-11、Isl-1、LIMK、ERM、MARCKS、CRMP-2等)来实现其生物学功能。研究表明多种疾病(如肺纤维化、心脑血管疾病、神经系统疾病和癌症等)与ROCK介导的通路有关,因此在药物研发中,ROCK被认为是一类重要的靶点。
然而,目前仅有法舒地尔(Fasudil)作为ROCK抑制剂在日本被批准用于治疗脑血管痉挛和缺血。虽然目前还有多种小分子ROCK抑制剂报道,但大多为局部眼科用药,目前没有适用于全身系统给药的小分子ROCK抑制剂。
发明概述
本发明提供用作ROCK(优选ROCK2)抑制剂的化合物,其具有对ROCK(优选ROCK2)的优异的抑制活性、良好的选择性(相对于ROCK1,对ROCK2具有更高的选择性)、更好的物理化学性质(例如溶解度、物理和/或化学稳定性)、改善的药物代谢动力学性质(例如改善的生物利用度、合适的半衰期和作用持续时间)、改善的安全性(较低的毒性和/或较少的副作用,较宽的治疗窗)等更优异的性质。
本发明的一个方面提供化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中所述化合物具有式(I)的结构:
Figure PCTCN2019116453-appb-000001
其中:
V 1选自-CR 2=N-、-N=CR 2-、-NR 3C(=O)-和-C(=O)NR 3-;
V 2选自-CR 2=N-、-N=CR 2-、-(CR 2R 2’) n-和-(CR 2=CR 2’) n-;
Y选自直接键、C(=O)、O、S(=O) i和NR;
R选自H、C 1-6烷基、C 2-6烯基、C 2-6炔基、饱和或部分不饱和的C 3-10环烃基、饱和或部分不饱和的3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基,所述环烃基和杂环基中至多2个环成员为C(=O);
环A和环B各自独立地选自饱和或部分不饱和的C 3-10烃环、饱和或部分不饱和的3-10元杂环、C 6-10芳环和5-14元杂芳环,所述烃环和杂环中至多2个环成员为C(=O);
环C为
Figure PCTCN2019116453-appb-000002
其通过*或**标记的两个位置之一与Y连接,并且另一位置与环B连接;
X选自CR 8和N;
R 1选自-C(=O)R 1a、-C(=O)OR 1a和-C(=O)NR 1aR 1b
R 1a和R 1b各自独立地选自H、卤素、氨基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 3-10 环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基、C 6-12芳烷基、-C(=O)R 5、-OC(=O)R 5、-C(=O)OR 5、-OR 5、-SR 5、-S(=O)R 5、-S(=O) 2R 5、-S(=O) 2NR 5R 6、-NR 5R 6、-C(=O)NR 5R 6、-NR 5-C(=O)R 6、-NR 5-C(=O)OR 6、-NR 5-S(=O) 2-R 6、-NR 5-C(=O)-NR 5R6、-C 1-6亚烷基-NR 5R 6、-C 1-6亚烷基-OR 5和-O-C 1-6亚烷基-NR 5R 6;或者R 1a和R 1b连同其所连接的原子共同构成3-12元杂环或杂芳环;
R 2、R 2’、R 3、R 4、R 7、R 8、R 9和R 10在每次出现时各自独立地选自H、卤素、氨基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 3-10环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基、C 6-12芳烷基、-C(=O)R 5、-OC(=O)R 5、-C(=O)OR 5、-OR 5、-SR 5、-S(=O)R 5、-S(=O) 2R 5、-S(=O) 2NR 5R 6、-NR 5R 6、-C(=O)NR 5R 6、-NR 5-C(=O)R 6、-NR 5-C(=O)OR 6、-NR 5-S(=O) 2-R 6、-NR 5-C(=O)-NR 5R 6、-C 1-6亚烷基-NR 5R 6、-C 1-6亚烷基-O(P=O)(OH) 2和-O-C 1-6亚烷基-NR 5R 6;或者R 7和R 8连同其所连接的基团共同构成C 6-10芳环或5-14元杂芳环;
上述烷基、亚烷基、烯基、亚烯基、炔基、环烃基、烃环、杂环基、杂环、芳基、芳环、杂芳基、杂芳环和芳烷基在每次出现时各自任选地被一个或多个独立地选自下列的取代基取代:卤素、羟基、氧代、氨基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 3-6环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基、C 6-12芳烷基、=N-OR 5、-C(=NH)NH 2、-C(=O)R 5、-OC(=O)R 5、-C(=O)OR 5、-OR 5、-SR 5、-S(=O)R 5、-S(=O) 2R 5、-S(=O) 2NR 5R 6、-NR 5R 6、-C(=O)NR 5R 6、-NR 5-C(=O)R 6、-NR 5-C(=O)OR 6、-NR 5-S(=O) 2-R 6、-NR 5-C(=O)-NR 5R 6、-C 1-6亚烷基-NR 5R 6和-O-C 1-6亚烷基-NR 5R 6,所述烷基、亚烷基、烯基、炔基、环烃基、杂环基、芳基、杂芳基和芳烷基进一步任选地被一个或多个独立地选自下列的取代基取代:卤素、羟基、氧代、氨基、氰基、硝基、C 1-6烷基、C 3-6环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基;
R 5和R 6在每次出现时各自独立地选自H、C 1-6烷基、C 3-10环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基;或者当R 5和R 6与同一氮原子连接时,R 5和R 6连同其所连接的原子任选地共同构成3-12元杂环或杂芳环;
m在每次出现时各自独立地为0、1、2或3的整数;
n为0、1、2、3或4的整数;并且
i为0、1或2的整数。
本发明的另一方面提供药物组合物,其包含预防或治疗有效量的本发明的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药以及一种或多种药学上可接受的载体,所述药物组合物优选为固体制剂、半固体制剂、液体制剂或气态制剂。
本发明的另一方面提供本发明的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药或者本发明的药物组合物在制备用作Rho相关蛋白激酶(ROCK)抑制剂,优选选择性ROCK2抑制剂的药物中的用途。
本发明的另一方面提供本发明的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药或者本发明的药物组合物,其用作Rho相关蛋白激酶(ROCK)抑制剂,优选选择性ROCK2抑制剂。
本发明的另一方面提供预防或治疗Rho相关蛋白激酶(ROCK)介导的疾病的方法,所述方法包括向需要其的个体给药有效量的本发明的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药或者本发明的药物组合物。
发明详细描述
定义
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。提及本文中使用的技术意图指在本领域中通常所理解的技术,包括那些对本领域技术人员显而易见的技术的变化或等效技术的替换。虽然相信以下术语对于本领域技术人员很好理解,但仍然阐述以下定义以更好地解释本发明。
术语“包括”、“包含”、“具有”、“含有”或“涉及”及其在本文中的其它变体形式为包含性的(inclusive)或开放式的,且不排除其它未列举的元素或方法步骤。
如本文中所使用,术语“亚烷基”表示饱和二价烃基,优选表示具有1、2、3、4、5或6个碳原子的饱和二价烃基,例如亚甲基、亚乙基、亚丙基或亚丁基。
如本文中所使用,术语“烷基”定义为线性或支化饱和脂肪族烃。在一些实施方案中,烷基具有1至12个,例如1至6个碳原子。例如,如本文中所使用,术语“C 1-6烷基”指1至6个碳原子的线性 或支化的基团(例如甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、异戊基、新戊基或正己基),其任选地被1或多个(诸如1至3个)适合的取代基如卤素取代(此时该基团被称作“卤代烷基”)(例如CH 2F、CHF 2、CF 3、CCl 3、C 2F 5、C 2Cl 5、CH 2CF 3、CH 2Cl或-CH 2CH 2CF 3等)。术语“C 1-4烷基”指1至4个碳原子的线性或支化的脂肪族烃链(即甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基)。
如本文中所使用,术语“烯基”意指线性的或支化的单价烃基,其包含一个双键,且具有2-6个碳原子(“C 2-6烯基”)。所述烯基为例如乙烯基、1-丙烯基、2-丙烯基、2-丁烯基、3-丁烯基、2-戊烯基、3-戊烯基、4-戊烯基、2-己烯基、3-己烯基、4-己烯基、5-己烯基、2-甲基-2-丙烯基和4-甲基-3-戊烯基。当本发明的化合物含有亚烯基时,所述化合物可以纯E(异侧(entgegen))形式、纯Z(同侧(zusammen))形式或其任意混合物形式存在。
如本文中所使用,术语“炔基”表示包含一个或多个三键的单价烃基,其优选具有2、3、4、5或6个碳原子,例如乙炔基或丙炔基。
如本文中所使用,术语“环烷基”指饱和的单环或多环(诸如双环)烃环(例如单环,诸如环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环壬基,或双环,包括螺环、稠合或桥连系统(诸如双环[1.1.1]戊基、双环[2.2.1]庚基、双环[3.2.1]辛基或双环[5.2.0]壬基、十氢化萘基等)),其任选地被1或多个(诸如1至3个)适合的取代基取代。所述环烷基具有3至15个碳原子。例如,术语“C 3-6环烷基”指3至6个成环碳原子的饱和的单环或多环(诸如双环)烃环(例如环丙基、环丁基、环戊基或环己基),其任选地被1或多个(诸如1至3个)适合的取代基取代,例如甲基取代的环丙基。
如本文中所使用,术语“亚环烃基”、“环烃基”和“烃环”是指具有例如3-10个(适合地具有3-8个,更适合地具有3-6个)环碳原子的饱和(即,“亚环烷基”和“环烷基”)或不饱和的(即在环内具有一个或多个双键和/或三键)单环或多环烃环,其包括但不限于(亚)环丙基(环)、(亚)环丁基(环)、(亚)环戊基(环)、(亚)环己基(环)、(亚)环庚基(环)、(亚)环辛基(环)、(亚)环壬基(环)、(亚)环己烯基(环)等。
如本文中所使用,术语“杂环基”、“亚杂环基”和“杂环”是指具有例如3-10个(适合地具有3-8个,更适合地具有3-6个)环原子、其中至少一个环原子是选自N、O和S的杂原子且其余环原子是C的饱和(即,杂环烷基)或部分不饱和的(即在环内具有一个或多个双键和/或三键)环状基团。例如,“3-10元(亚)杂环(基)”是具有2-9个(如2、3、4、5、6、7、8或9个)环碳原子和独立地选自N、O和S的一个或多个(例如1个、2个、3个或4个)杂原子的饱和或部分不饱和(亚)杂环(基)。亚杂环基和杂环(基)的实例包括但不限于:(亚)环氧乙烷基、(亚)氮丙啶基、(亚)氮杂环丁基(azetidinyl)、(亚)氧杂环丁基(oxetanyl)、(亚)四氢呋喃基、(亚)二氧杂环戊烯基(dioxolinyl)、(亚)吡咯烷基、(亚)吡咯烷酮基、(亚)咪唑烷基、(亚)吡唑烷基、(亚)吡咯啉基、(亚)四氢吡喃基、(亚)哌啶基、(亚)吗啉基、(亚)二噻烷基(dithianyl)、(亚)硫吗啉基、(亚)哌嗪基或(亚)三噻烷基(trithianyl)。所述基团也涵盖双环系统,包括螺环、稠合或桥连系统(诸如8-氮杂螺[4.5]癸烷、3,9-二氮杂螺[5.5]十一烷、2-氮杂双环[2.2.2]辛烷等)。亚杂环基和杂环(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。
如本文中所使用,术语“(亚)芳基”和“芳环”指具有共轭π电子系统的全碳单环或稠合环多环芳族基团。例如,如本文中所使用,术语“C 6-10(亚)芳基”和“C 6-10芳环”意指含有6至10个碳原子的芳族基团,诸如(亚)苯基(苯环)或(亚)萘基(萘环)。(亚)芳基和芳环任选地被1或多个(诸如1至3个)适合的取代基(例如卤素、-OH、-CN、-NO 2、C 1-6烷基等)取代。
如本文中所使用,术语“(亚)杂芳基”和“杂芳环”指单环、双环或三环芳族环系,其具有5、6、8、9、10、11、12、13或14个环原子,特别是1或2或3或4或5或6或9或10个碳原子,且其包含至少一个可以相同或不同的杂原子(所述杂原子是例如氧、氮或硫),并且,另外在每一种情况下可为苯并稠合的。特别地,“(亚)杂芳基”或“杂芳环”选自(亚)噻吩基、(亚)呋喃基、(亚)吡咯基、(亚)噁唑基、(亚)噻唑基、(亚)咪唑基、(亚)吡唑基、(亚)异噁唑基、(亚)异噻唑基、(亚)噁二唑基、(亚)三唑基、(亚)噻二唑基等,以及它们的苯并衍生物;或(亚)吡啶基、(亚)哒嗪基、(亚)嘧啶基、(亚)吡嗪基、(亚)三嗪基等,以及它们的苯并衍生物。
如本文中所使用,术语“芳烷基”优选表示芳基或杂芳基取代的烷基,其中所述芳基、杂芳基和烷基如本文中所定义。通常,所述芳基可具有6-14个碳原子,所述杂芳基可具有5-14个环原子,并且所述烷基可具有1-6个碳原子。示例性芳烷基包括但不限于苄基、苯基乙基、苯基丙基、苯基丁基。
如本文中所使用,术语“卤代”或“卤素”基团定义为包括F、Cl、Br或I。
如本文中所使用,术语“含氮杂环”指饱和或不饱和的单环或双环基团,其在环中具有2、3、4、5、6、7、8、9、10、11、12或13个碳原子和至少一个氮原子,其还可任选地包含一个或多个(例如一个、两个、三个或四个)选自N、O、C=O、S、S=O和S(=O) 2的环成员,其通过所述含氮杂环中 的氮原子以及任一其余环原子与分子的其余部分连接,所述含氮杂环任选地为苯并稠合的,并且优选通过所述含氮杂环中的氮原子以及所稠合的苯环中的任一碳原子与分子的其余部分连接。
术语“取代”指所指定的原子上的一个或多个(例如一个、两个、三个或四个)氢被从所指出的基团的选择代替,条件是未超过所指定的原子在当前情况下的正常原子价并且所述取代形成稳定的化合物。取代基和/或变量的组合仅仅当这种组合形成稳定的化合物时才是允许的。
如果取代基被描述为“任选地被取代”,则取代基可(1)未被取代或(2)被取代。如果取代基的碳被描述为任选地被取代基列表中的一个或多个取代,则碳上的一个或多个氢(至存在的任何氢的程度)可单独和/或一起被独立地选择的任选的取代基替代。如果取代基的氮被描述为任选地被取代基列表中的一个或多个取代,则氮上的一个或多个氢(至存在的任何氢的程度)可各自被独立地选择的任选的取代基替代。
如果取代基被描述为“独立地选自”一组,则各取代基独立于另一者被选择。因此,各取代基可与另一(其他)取代基相同或不同。
如本文中所使用,术语“一个或多个”意指在合理条件下的1个或超过1个,例如2个、3个、4个、5个或10个。
除非指明,否则如本文中所使用,取代基的连接点可来自取代基的任意适宜位置。
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。
本发明还包括所有药学上可接受的同位素标记的化合物,其与本发明的化合物相同,除了一个或多个原子被具有相同原子序数但原子质量或质量数不同于在自然界中占优势的原子质量或质量数的原子替代。适合包含入本发明的化合物中的同位素的实例包括(但不限于)氢的同位素(例如氘( 2H)、氚( 3H));碳的同位素(例如 11C、 13C及 14C);氯的同位素(例如 36Cl);氟的同位素(例如 18F);碘的同位素(例如 123I及 125I);氮的同位素(例如 13N及 15N);氧的同位素(例如 15O、 17O及 18O);磷的同位素(例如 32P);及硫的同位素(例如 35S)。某些同位素标记的本发明的化合物(例如掺入放射性同位素的那些)可用于药物和/或底物组织分布研究(例如分析)中。放射性同位素氚(即 3H)及碳-14(即 14C)因易于掺入且容易检测而特别可用于该目的。用正电子发射同位素(例如 11C、 18F、 15O及 13N)进行取代可在正电子发射断层显像术(PET)研究中用于检验底物受体占据情况。被同位素标记的本发明的化合物可通过与描述于随附路线和/或实施例及制备中的那些类似的方法通过使用适当的被同位素标记的试剂代替之前采用的非标记的试剂来制备。本发明的药学上可接受的溶剂合物包括其中结晶溶剂可被同位素取代的那些,例如,D 2O、丙酮-d 6或DMSO-d 6
术语“立体异构体”表示由于至少一个不对称中心形成的异构体。在具有一个或多个(例如一个、两个、三个或四个)不对称中心的化合物中,其可产生外消旋混合物、单一对映异构体、非对映异构体混合物和单独的非对映异构体。特定个别分子也可以几何异构体(顺式/反式)存在。类似地,本发明的化合物可以两种或更多种处于快速平衡的结构不同的形式的混合物(通常称作互变异构体)存在。互变异构体的代表性实例包括酮-烯醇互变异构体、苯酚-酮互变异构体、亚硝基-肟互变异构体、亚胺-烯胺互变异构体等。要理解,本申请的范围涵盖所有这样的以任意比例(例如60%、65%、70%、75%、80%、85%、90%、95%、96%、97%、98%、99%)的异构体或其混合物。
本文中可使用实线
Figure PCTCN2019116453-appb-000003
实楔形
Figure PCTCN2019116453-appb-000004
或虚楔形
Figure PCTCN2019116453-appb-000005
描绘本发明的化合物的化学键。使用实线以描绘键连至不对称碳原子的键欲表明,包括该碳原子处的所有可能的立体异构体(例如,特定的对映异构体、外消旋混合物等)。使用实或虚楔形以描绘键连至不对称碳原子的键欲表明,存在所示的立体异构体。当存在于外消旋混合物中时,使用实及虚楔形以定义相对立体化学,而非绝对立体化学。除非另外指明,否则本发明的化合物意欲可以立体异构体(其包括顺式及反式异构体、光学异构体(例如R及S对映异构体)、非对映异构体、几何异构体、旋转异构体、构象异构体、阻转异构体及其混合物)的形式存在。本发明的化合物可表现一种以上类型的异构现象,且由其混合物(例如外消旋混合物及非对映异构体对)组成。
本发明涵盖本发明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。
还应当理解,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,以其药学上可接受的衍生物形式存在。在本发明中,药学上可接受的衍生物包括但不限于,药学上可接受的盐、酯、溶剂合物、N-氧化物、代谢物或前药,在将它们向需要其的患者给药后,能够直接或间接提供本发明的化合物或其代谢物或残余物。因此,当在本文中提及“本发明的化合物”时,也意在涵盖化合物的上述各种衍生物形式。
本发明的化合物的药学上可接受的盐包括其酸加成盐及碱加成盐。
适合的酸加成盐由形成药学可接受盐的酸来形成。实例包括乙酸盐、己二酸盐、天冬氨酸盐、苯甲酸盐、苯磺酸盐、碳酸氢盐/碳酸盐、硫酸氢盐/硫酸盐、硼酸盐、樟脑磺酸盐、柠檬酸盐、环己氨磺酸盐、乙二磺酸盐、乙磺酸盐、甲酸盐、延胡索酸盐、葡庚糖酸盐、葡糖酸盐、葡糖醛酸盐、六氟磷酸盐、海苯酸盐、盐酸盐/氯化物、氢溴酸盐/溴化物、氢碘酸盐/碘化物、羟乙基磺酸盐、乳酸盐、苹果酸盐、顺丁烯二酸盐、丙二酸盐、甲磺酸盐、甲基硫酸盐、萘甲酸盐(naphthylate)、2-萘磺酸盐、烟酸盐、硝酸盐、乳清酸盐、草酸盐、棕榈酸盐、双羟萘酸盐、磷酸盐/磷酸氢盐/磷酸二氢盐、焦谷氨酸盐、糖二酸盐、硬脂酸盐、丁二酸盐、单宁酸盐、酒石酸盐、甲苯磺酸盐、三氟乙酸盐及昔萘酸盐(xinofoate)。
适合的碱加成盐由形成药学可接受盐的碱来形成。实例包括铝盐、精氨酸盐、苄星青霉素盐、钙盐、胆碱盐、二乙胺盐、二乙醇胺盐、甘氨酸盐、赖氨酸盐、镁盐、葡甲胺盐、乙醇胺盐、钾盐、钠盐、氨丁三醇盐及锌盐。
适合的盐的综述参见Stahl及Wermuth的“Handbook of Pharmaceutical Salts:Properties,Selection,and Use”(Wiley-VCH,2002)。用于制备本发明的化合物的药学上可接受的盐的方法为本领域技术人员已知的。
如本文中所使用,术语“酯”意指衍生自本申请中各个通式化合物的酯,其包括生理上可水解的酯(可在生理条件下水解以释放游离酸或醇形式的本发明的化合物)。本发明的化合物本身也可以是酯。
本发明的化合物可以溶剂合物(优选水合物)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。
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(MCPBA)、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;A.R.Katritzky和A.J.Boulton,Eds.,Academic Press;以及G.W.H.Cheeseman和E.S.G.Werstiuk,Advances in Heterocyclic Chemistry,vol.22,pp 390-392,A.R.Katritzky和A.J.Boulton,Eds.,Academic Press。
在本发明的范围内还包括本发明的化合物的代谢物,即在给药本发明的化合物时体内形成的物质。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,包括通过使本发明的化合物与哺乳动物接触足以产生其代谢产物的时间的方法制得的化合物。
本发明在其范围内进一步包括本发明的化合物的前药,其为自身可具有较小药理学活性或无药理学活性的本发明的化合物的某些衍生物当被给药至身体中或其上时可通过例如水解裂解转化成具有期望活性的本发明的化合物。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。关于前药的使用的其他信息可参见“Pro-drugs as Novel Delivery Systems”,第14卷,ACS Symposium Series(T.Higuchi及V.Stella)。本发明的前药可例如通过用本领域技术人员已知作为“前-部分(pro-moiety)(例如“Design of Prodrugs”,H.Bundgaard(Elsevier,1985)中所述)”的某些部分替代本发明的化合物中存在的适当官能团来制备。
本发明还涵盖含有保护基的本发明的化合物。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移除保护基。
术语“约”是指在所述数值的±10%范围内,优选±5%范围内,更优选±2%范围内。
化合物
在一些实施方案中,本发明提供化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中所述化合物具有式(I)的结构:
Figure PCTCN2019116453-appb-000006
其中:
V 1选自-CR 2=N-、-N=CR 2-、-NR 3C(=O)-和-C(=O)NR 3-;
V 2选自-CR 2=N-、-N=CR 2-、-(CR 2R 2’) n-和-(CR 2=CR 2’) n-;
Y选自直接键、C(=O)、O、S(=O) i和NR;
R选自H、C 1-6烷基、C 2-6烯基、C 2-6炔基、饱和或部分不饱和的C 3-10环烃基、饱和或部分不饱和的3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基,所述环烃基和杂环基中至多2个环成员为C(=O);
环A和环B各自独立地选自饱和或部分不饱和的C 3-10烃环、饱和或部分不饱和的3-10元杂环、C 6-10芳环和5-14元杂芳环,所述烃环和杂环中至多2个环成员为C(=O);
环C为
Figure PCTCN2019116453-appb-000007
其通过*或**标记的两个位置之一与Y连接,并且另一位置与环B连接;
X选自CR 8和N;
R 1选自-C(=O)R 1a、-C(=O)OR 1a和-C(=O)NR 1aR 1b
R 1a和R 1b各自独立地选自H、卤素、氨基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 3-10环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基、C 6-12芳烷基、-C(=O)R 5、-OC(=O)R 5、-C(=O)OR 5、-OR 5、-SR 5、-S(=O)R 5、-S(=O) 2R 5、-S(=O) 2NR 5R 6、-NR 5R 6、-C(=O)NR 5R 6、-NR 5-C(=O)R 6、-NR 5-C(=O)OR 6、-NR 5-S(=O) 2-R 6、-NR 5-C(=O)-NR 5R 6、-C 1-6亚烷基-NR 5R 6、-C 1-6亚烷基-OR 5和-O-C 1-6亚烷基-NR 5R 6;或者R 1a和R 1b连同其所连接的原子共同构成3-12元杂环或杂芳环;
R 2、R 2’、R 3、R 4、R 7、R 8、R 9和R 10在每次出现时各自独立地选自H、卤素、氨基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 3-10环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基、C 6-12芳烷基、-C(=O)R 5、-OC(=O)R 5、-C(=O)OR 5、-OR 5、-SR 5、-S(=O)R 5、-S(=O) 2R 5、-S(=O) 2NR 5R 6、-NR 5R 6、-C(=O)NR 5R 6、-NR 5-C(=O)R 6、-NR 5-C(=O)OR 6、-NR 5-S(=O) 2-R 6、-NR 5-C(=O)-NR 5R 6、-C 1-6亚烷基-NR 5R 6、-C 1-6亚烷基-O(P=O)(OH) 2和-O-C 1-6亚烷基-NR 5R 6;或者R 7和R 8连同其所连接的基团共同构成C 6-10芳环或5-14元杂芳环;
上述烷基、亚烷基、烯基、亚烯基、炔基、环烃基、烃环、杂环基、杂环、芳基、芳环、杂芳基、杂芳环和芳烷基在每次出现时各自任选地被一个或多个独立地选自下列的取代基取代:卤素、羟基、氧代、氨基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 3-6环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基、C 6-12芳烷基、=N-OR 5、-C(=NH)NH 2、-C(=O)R 5、-OC(=O)R 5、-C(=O)OR 5、-OR 5、-SR 5、-S(=O)R 5、-S(=O) 2R 5、-S(=O) 2NR 5R 6、-NR 5R 6、-C(=O)NR 5R 6、-NR 5-C(=O)R 6、-NR 5-C(=O)OR 6、-NR 5-S(=O) 2-R 6、-NR 5-C(=O)-NR 5R 6、-C 1-6亚烷基-NR 5R 6和-O-C 1-6亚烷基-NR 5R 6,所述烷基、亚烷基、烯基、炔基、环烃基、杂环基、芳基、杂芳基和芳烷基进一步任选地被一个或多个独立地选自下列的取代基取代:卤素、羟基、氧代、氨基、氰基、硝基、C 1-6烷基、C 3-6环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基;
R 5和R 6在每次出现时各自独立地选自H、C 1-6烷基、C 3-10环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基;或者当R 5和R 6与同一氮原子连接时,R 5和R 6连同其所连接的原子任选地共同构成3-12元杂环或杂芳环;
m在每次出现时各自独立地为0、1、2或3的整数;
n为0、1、2、3或4的整数;并且
i为0、1或2的整数。
在一些实施方案中,本发明提供化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中Y选自直接键、C(=O)、O、S、 S(=O)、S(=O) 2、NH和NCH 3
在一些实施方案中,本发明提供化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中
Figure PCTCN2019116453-appb-000008
优选为
Figure PCTCN2019116453-appb-000009
Figure PCTCN2019116453-appb-000010
以上基团通过#或##标记的两个位置之一与环C连接,并且另一位置与R 1连接,
其中
Figure PCTCN2019116453-appb-000011
表示单键或双键且相邻的键不同时为双键;
Z 1、Z 2、Z 3、Z 4、Z 5、Z 6、Z 7、Z 8和Z 9在每次出现时各自独立地选自C、CR 9、C(R 9) 2、CR 10、C(R 10) 2、C(=O)、N、NR 9、NR 10、O和S;优选地,Z 1、Z 2、Z 3、Z 4、Z 5、Z 6、Z 7、Z 8和Z 9在每次出现时各自独立地选自C、CH、CF、CCl、CCH 3、CH 2、C(CH 3) 2、C-OCH 3、C(=O)、N、NH、NCH 3、NCH 2CH 3、NCH(CH 3) 2、NCH=CH 2、NCH 2F、NCHF 2、NCH 2CHF 2、NC(=O)CH 3、NCH 2OH、NCH 2OMe、NCH 2CH 2OMe、NCH 2-O(P=O)(OH) 2
Figure PCTCN2019116453-appb-000012
NCH 2CH 2-N(CH 3) 2、O和S;
j为0、1、2、3或4;
条件是Z 1-Z 9中的至多两个基团同时为C(=O);
R 9和R 10在每次出现时各自独立地选自卤素、甲基、乙基、丙基、乙烯基、环丙基、环丁基、环戊基、氧杂环丁烷基、单氟甲基、二氟甲基、三氟甲基、-CH 2CHF 2、乙酰基、-OCH 3、-CH 2OH、-CH 2OCH 3、-CH 2CH 2OCH 3、-CH 2-O(P=O)(OH) 2
Figure PCTCN2019116453-appb-000013
和-CH 2CH 2-N(CH 3) 2
最优选地,
Figure PCTCN2019116453-appb-000014
选自
Figure PCTCN2019116453-appb-000015
Figure PCTCN2019116453-appb-000016
以上基团通过#或##标记的两个位置之一与环C连接,并且另一位置与R 1连接。
在一些实施方案中,本发明提供化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中环C选自
Figure PCTCN2019116453-appb-000017
Figure PCTCN2019116453-appb-000018
以上基团通过*或**标记的两个位置之一与Y连接,并且另一位置与环B连接。
在一些实施方案中,本发明提供化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中V 1选自-CH=N-、-N=CH-、-C(NH 2)=N-、-N=C(NH 2)-、-NHC(=O)-和-C(=O)NH-。
在一些实施方案中,本发明提供化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中V 2选自-CH=N-、-N=CH-、-CH 2-、-(CH 2) 2-、-(CH=CCl)-、-(CCl=CH)-和-(CH=CH)-。
在一些实施方案中,本发明提供化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中
Figure PCTCN2019116453-appb-000019
选自
Figure PCTCN2019116453-appb-000020
在优选的实施方案中,
Figure PCTCN2019116453-appb-000021
Figure PCTCN2019116453-appb-000022
Figure PCTCN2019116453-appb-000023
在一些实施方案中,本发明提供化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中R 1选自
Figure PCTCN2019116453-appb-000024
Figure PCTCN2019116453-appb-000025
Figure PCTCN2019116453-appb-000026
在一些实施方案中,本发明提供化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中所述化合物具有任意下式的结构:
Figure PCTCN2019116453-appb-000027
本发明涵盖对各个实施方案进行任意组合所得的化合物。
在一些实施方案中,本发明提供化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中所述化合物具有以下结构:
Figure PCTCN2019116453-appb-000028
Figure PCTCN2019116453-appb-000029
Figure PCTCN2019116453-appb-000030
Figure PCTCN2019116453-appb-000031
Figure PCTCN2019116453-appb-000032
Figure PCTCN2019116453-appb-000033
Figure PCTCN2019116453-appb-000034
Figure PCTCN2019116453-appb-000035
Figure PCTCN2019116453-appb-000036
Figure PCTCN2019116453-appb-000037
Figure PCTCN2019116453-appb-000038
药物组合物和治疗方法
在一些实施方案中,本发明提供药物组合物,其包含预防或治疗有效量的本发明的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药以及一种或多种药学上可接受的载体,所述药物组合物优选为固体制剂、半固体制剂、液体制剂或气态制剂。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它治疗剂。
在一些实施方案中,本发明提供本发明的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药或者本发明的药物组合物在制备用作Rho相关蛋白激酶(ROCK)抑制剂,优选选择性ROCK2抑制剂的药物中的用途。
在一些实施方案中,本发明提供本发明的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药或者本发明的药物组合物,其用作Rho相关蛋白激酶(ROCK)抑制剂,优选选择性ROCK2抑制剂。
在一些实施方案中,本发明提供预防或治疗Rho相关蛋白激酶(ROCK)介导的疾病的方法,所述方法包括向需要其的个体给药有效量的本发明的化合物或其药学上可接受的盐、酯、立体异构体、 多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药或者本发明的药物组合物。
在一些实施方案中,所述Rho相关蛋白激酶(ROCK)介导的疾病包括自身免疫性疾病(其包括类风湿性关节炎、全身性红斑狼疮(SLE;狼疮)、银屑病、克罗恩病、特应性皮炎、湿疹或移植物抗宿主病(GVHD));心血管病症(其包括高血压、动脉粥样硬化、再狭窄、心脏肥大、脑缺血、脑血管痉挛或勃起功能障碍);炎症(其包括哮喘、心血管炎症、溃疡性结肠炎或肾脏炎症);中枢神经系统病症(其包括神经元变性或脊髓损伤;所述中枢神经系统病症优选为亨延顿氏病、帕金森氏病、阿尔茨海默病、肌萎缩性脊髓侧索硬化症(ALS)或多发性硬化症);动脉血栓性病症(其包括血小板聚集或白细胞聚集);纤维化病症(其包括肝纤维化、肺纤维化或肾纤维化);瘤性疾病(其包括淋巴瘤、癌(例如鳞状细胞癌、小细胞肺癌、垂体癌、食道癌、非小细胞肺癌、肺腺癌、肺鳞癌、腹膜癌、肝细胞癌、胃肠癌、胰腺癌、胶质母细胞瘤、宫颈癌、卵巢癌、膀胱癌、肝癌、乳腺癌、结肠癌、结直肠癌、子宫内膜癌或子宫癌、唾液腺癌、肾癌、前列腺癌、外阴癌、甲状腺癌、脑癌、子宫内膜癌、睾丸癌、胆管癌、胆囊癌、胃癌、黑色素瘤或头颈部癌症)、白血病、星形细胞瘤、软组织肉瘤、肉瘤或母细胞瘤);代谢综合征;胰岛素抗性;高胰岛素血症;2型糖尿病;葡萄糖耐受不良;骨质疏松症;眼部病症(其包括高眼压、年龄相关性黄斑变性(AMD)、脉络膜新生血管形成(CNV)、糖尿病性黄斑水肿(DME)、虹膜新生血管形成、葡萄膜炎、青光眼(其包括原发性开角型青光眼、急性闭角型青光眼、色素性青光眼、先天性青光眼、正常张力青光眼、继发青光眼或新生血管性青光眼)或早产儿视网膜病变(ROP))。
在一些实施方案中,所述Rho相关蛋白激酶(ROCK)介导的疾病包括狼疮肾炎、动脉粥样硬化、类风湿性关节炎(RA)、血管瘤、血管纤维瘤、肺纤维化、银屑病、角膜移植排斥、胰岛素依赖型糖尿病、多发性硬化症、重症肌无力、克罗恩病、自身免疫性肾炎、原发性胆汁性肝硬化、急性胰腺炎、异体排异、过敏性炎症、接触性皮炎、迟发型超敏反应、炎症性肠病、感染性休克、骨质疏松、骨关节炎、神经元炎症、Osler-Weber综合征、再狭窄、真菌感染、寄生虫感染和病毒感染。
本发明中“药学上可接受的载体”是指与治疗剂一同给药的稀释剂、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/或其它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/风险比相应的其它问题或并发症。
在本发明的药物组合物中可使用的药学上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。适合的药学上可接受的载体的实例如在Remington’s Pharmaceutical Sciences(1990)中所述。
本发明的药物组合物可以系统地作用和/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(如静脉内、动脉内、皮下、腹膜内、肌内注射,包括滴注)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。
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。
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。
如本文中所使用的术语“有效量”指被给药后会在一定程度上缓解所治疗病症的一或多种症状的化合物的量。
可调整给药方案以提供最佳所需响应。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。
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,而在其它情况下,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。
本发明的化合物在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,更优选1-150mg,特别优选1-50mg,例如1.5mg、2mg、4mg、10mg、25mg等。
除非另外说明,否则如本文中所使用,术语“治疗(treating)”意指逆转、减轻、抑制这样的术语所应用的病症或病况或者这样的病症或病况的一或多种症状的进展,或预防这样的病症或病况或者这样的病症或病况的一或多种症状。
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)。
在一些实施方案中,本发明的药物组合物还可以包含一种或多种另外的治疗剂或预防剂。
实施例
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。
化合物的结构通过核磁共振波谱( 1H NMR)或质谱(MS)进行确证。
化学位移(δ)以百万分之一(ppm)为单位给出。 1HNMR的测定在Bruker BioSpin GmbH 400核磁仪上进行,测试溶剂为氘代甲醇(CD 3OD)、氘代氯仿(CDCl 3)或六氘代二甲基亚砜(DMSO-d 6),内标为四甲基硅烷(TMS)。
LC-MS的测定在岛津LC-MS-2020液质联用仪(生产商:岛津,型号:岛津LC-MS-2020)上进行。
制备高效液相色谱法使用waters 2767(waters sunfire,C18,19×250mm 10um色谱柱)进行。
薄层色谱法(TLC)使用黄海牌HSGF 254(5×20cm)硅胶板进行,薄层制备色谱法采用规格为烟台产GF 254(0.4~0.5nm)硅胶板进行。
采用薄层色谱法(TLC)或LC-MS检测反应,使用的展开剂体系包括二氯甲烷和甲醇体系、正己烷和乙酸乙酯体系以及石油醚和乙酸乙酯体系,根据要分离的化合物的极性不同对展开剂体系进行调节(通过调节溶剂的体积比或者加入三乙胺等进行)。
微波反应使用BiotageInitiator+(400W,RT~300℃)微波反应器。
柱色谱法一般使用于成化工200~300目硅胶为固定相。洗脱剂的体系包括二氯甲烷和甲醇体系和正己烷和乙酸乙酯体系,根据要分离的化合物的极性不同对洗脱剂体系进行调节(通过调节溶剂的体积比或者加入三乙胺等进行)。
如在实施例中无特殊说明,反应的温度为室温(20℃~30℃)。
实施例中所使用的试剂购自Acros Organics、Aldrich Chemical Company或上海毕得医药科技有限公司等公司。
本发明中的缩写具有以下含义:
缩写 含义
ACN 乙腈
AcNH 2 乙酰胺
AcOH 乙酸/醋酸
AcOK/KOAc 乙酸钾
aq. 水溶液
Boc 2O 二碳酸二叔丁酯
Cs 2CO 3 碳酸铯
DCM 二氯甲烷
DIEA/DIPEA N,N-二异丙基乙胺
DMAP 二甲基氨基吡啶
DMF N,N-二甲基甲酰胺
DMSO 二甲基亚砜
EA 乙酸乙酯
Et 3N 三乙胺
EtOH 乙醇
FA 甲酸
HATU O-(7-氮杂苯并三唑-1-基)-N,N,N’,N’-四甲基脲六氟磷酸酯
HCl 盐酸
H 2O
K 2CO 3 碳酸钾
KOH 氢氧化钾
m-CPBA 间氯过氧苯甲酸
MeCN 乙腈
MeOH 甲醇
Na 2CO 3 碳酸钠
NaH 氢化钠
NaOH 氢氧化钠
n-BuLi 正丁基锂
NH 4Cl 氯化铵
NMP N-甲基吡咯烷酮
NPC 二(对硝基苯)碳酸酯
Pd/C 钯/碳
Pd 2(dba) 3 三(二亚苄基丙酮)二钯
Pd(dppf)Cl 2 [1,1’-双(二苯基膦)二茂铁]二氯化钯
Pd(OAc) 2 醋酸钯
Pd(PPh 3) 4 四(三苯基膦)钯
Pd(PPh 3) 2Cl 2 双(三苯基膦)二氯化钯
PE 石油醚
POCl 3 三氯氧磷
SOCl 2 氯化亚砜
t-BuOH 叔丁醇
t-BuXPhos 2-二叔丁基膦-2’,4’,6’-三异丙基联苯
TEA 三乙胺
TFA 三氟乙酸
TFAA 三氟乙酸酐
THF 四氢呋喃
Xantphos 4,5-双二苯基膦-9,9-二甲基氧杂蒽
实施例1:6-((2-(5-(3,3-二氟氮杂环丁烷-1-羰基)异吲哚啉-2-基)嘧啶-4-基)氨基)异喹啉-1(2H)-酮(C1)的制备
Figure PCTCN2019116453-appb-000039
第一步:
将C1-1(446mg,2.09mmol)和2-氯嘧啶-4-胺(270mg,2.09mmol)溶于NMP(12mL),加入碳酸铯(1.02g,3.13mmol),反应于微波辅助下,200℃反应0.5小时。LC-MS显示反应完全。室温搅拌下加入二氯甲烷(120mL),继续搅拌0.5小时,过滤,收集固体,干燥后得C1-2(300mg,棕色固体,收率56%)。MS m/z(ESI):257.0[M+H] +.
第二步:
将C1-2(230mg,0.76mmol)、3,3-二氟氮杂环丁烷盐酸盐(197mg,1.52mmol)和HATU(289mg,0.76mmol)溶于DMF(5mL)中,加入DIPEA(294mg,2.28mmol),反应于室温下搅拌1小时,LC-MS显示反应完全。减压蒸除溶剂,残余物以combi-flash硅胶柱色谱法(甲醇/二氯甲烷,0~5%)分离得到C1-3(110mg,棕色固体,收率43.7%)。
1H NMR(400MHz,DMSO-d 6)δ8.74(d,J=4.3Hz,1H),8.51(d,J=8.4Hz,1H),7.80(d,J=4.0Hz,1H),7.73(s,1H),7.64(d,J=8.0Hz,1H),7.11(s,2H),4.80(s,4H),3.65-3.57(m,2H),3.15-3.09(m,2H).MS m/z(ESI):332.0[M+H] +.
第三步:
将C1-3(100mg,0.3mmol)、6-溴异喹啉-1(2H)-酮(67.2mg,0.3mmol)和碳酸铯(293mg,0.9mmol)混合于1,4-二氧六环(5mL)中,以N 2置换3次后,加入Pd 2(dba) 3(27.5mg,0.03mmol)和Xantphos(52mg,0.09mmol),再次以N 2置换3次后置于90℃油浴中反应16小时。LC-MS显示有少量产物。补加Pd 2(dba) 3(27.5mg,0.03mmol)和Xantphos(52mg,0.09mmol),使反应继续于90℃油浴中反应24小时,LC-MS显示产物的转化率有增加。将反应液冷却至室温,过滤,滤液减压浓缩至干,将粗品经制备液相色谱法(乙腈/水(0.1%TFA),20~60%,30分钟)分离得到C1(13mg,浅黄色固体,收率7.4%)。
1H NMR(400MHz,DMSO-d 6)δ11.18(s,1H),10.82(s,1H),8.24(d,J=8.0Hz,1H),8.17(s,1H),8.06(d,J=8.0Hz,1H),7.88-7.78(m,2H),7.73-7.67(m,1H),7.64-7.55(m,1H),7.23-7.17(m,1H),6.64(t,J=6.8Hz,1H),6.47(d,J=6.8Hz,1H),5.08(s,J=8.0Hz,2H),4.95(s,J=8.0Hz,2H),4.79(s,2H),4.51(s,2H).MS m/z(ESI):474.6[M+H] +.
实施例2:6-((2-(2-(3,3-二氟氮杂环丁烷-1-羰基)-1H-吲哚-6-基)-5-氟嘧啶-4-基)氨基)异喹啉-1(2H)-酮(C9)的制备
Figure PCTCN2019116453-appb-000040
第一步:
在1L烧瓶中加入C9-1(31.3g,130mmol)、3,3-二氟氮杂环丁烷盐酸盐(20g,140mmol)、HATU(60g,160mmol)和DMF(330mL),搅拌下加入DIEA(50g,390mmol),并使反应于室温下搅拌过夜。LC-MS显示反应完全。将反应液浓缩,加入水(200mL)、甲醇(20mL)和乙腈(20mL),将此混合物于室温搅拌1小时,过滤,收集固体,干燥后得C9-2(41g,棕色固体,收率100%)。
1H NMR(400MHz,DMSO-d 6)δ11.88(s,1H),7.61(s,1H),7.60(d,J=8.6Hz,1H),7.20(d,J=8.6Hz,1H),6.94(s,1H),4.98(s,2H),4.56(s,2H).MS m/z(ESI):314.9,316.9[M+H] +.
第二步:
将化合物C9-2(1.151g,3.66mmol)、联硼酸频那醇酯(1.024g,4.03mmol)和醋酸钾(1.077g,10.98mmol)溶于1,4-二氧六环(20mL)中,以N 2置换3次后加入Pd(dppf)Cl 2(536mg,0.732mmol),再次以N 2置换3次后加热回流反应16小时。LC-MS显示反应完全。冷却至室温,滤除不溶物,将有机相减压浓缩得到粗品,将粗品经硅胶柱色谱法(二氯甲烷∶甲醇=30∶1)分离得到化合物C9-3(700mg,棕色固体,收率53%)。
1H NMR(400MHz,DMSO-d 6)δ11.90(s,1H),7.83(s,1H),7.62(d,J=8.0Hz,1H),7.35(d,J=8.1Hz,1H),6.92(s,1H),4.99(s,2H),4.58(s,2H),1.31(s,12H).MS m/z(ESI):363.0[M+H] +.
第三步:
将化合物C9-3(300mg,0.83mmol)和2氯-5-氟嘧啶-4-胺(122mg,0.83mmol)溶于1,4-二氧六环(20mL)中,加水(2mL)、碳酸钾(343mg,2.49mmol)和Pd(dppf)Cl 2(61mg,0.08mmol)后,氮气置换三次,反应液在110℃下搅拌16小时。LC-MS检测显示反应完成,反应液冷却至室温,真空旋蒸除去反应溶剂。残留物通过硅胶柱色谱法分离纯化(二氯甲烷∶甲醇=10∶1),得到化合物C9-4粗品(400mg,棕色固体)。
1H NMR(400MHz,DMSO-d6)δ12.00(s,1H),8.38(s,1H),8.25(s,1H),8.03(d,J=8.4Hz,1H),7.66(d,J=8.3Hz,1H),7.31(s,2H),6.94(s,1H).MS m/z(ESI):347.8[M+H] +.
第四步:
将化合物C9-4(100mg,0.29mmol)和6-溴异喹啉-1(2H)-酮(65mg,0.29mmol)溶于1,4-二氧六环(10mL)中,加入碳酸铯(283mg,0.87mmol)、Pd 2(dba) 3(27mg,0.03mmol)和Xantphos(34mg,0.06mmol),氮气置换三次,反应液在微波辅助110℃条件下,搅拌1小时。LC-MS检测显示反应完成,反应液冷却至室温,过滤,真空旋蒸除去反应溶剂,所得粗品用制备HPLC纯化(ACN/H 2O(0.1%TFA),20~60%)得到化合物C9(20mg,黄色固体,收率14%)。
1HNMR(400MHz,DMSO-d 6)δ12.14(s,1H),11.12(s,1H),9.9(s,1H),8.58(s,1H),8.49(s,1H),8.25(d,J=8.2Hz,2H),8.09(s,2H),7.75(d,J=8.6Hz,1H),7.19(s,1H),6.98(s,1H),6.59(d,J=7.5Hz,1H),5.04(s,2H),4.56(s,2H).MS m/z(ESI):490.7[M+H] +.
通过与实施例2中所述类似方法,制备下表中的化合物。
Figure PCTCN2019116453-appb-000041
Figure PCTCN2019116453-appb-000042
*中间体C20-1和C20-2的合成:
Figure PCTCN2019116453-appb-000043
第一步:
将化合物C9-2(9g,28.6mmol)溶于DMF(100mL)中,N 2保护下以冰水浴冷却至0℃,分批加入NaH(60%,1.7g,42.9mmol),搅拌0.5小时后加入溴甲基甲醚(5.4g,42.9mmol)。将反应自然升至室温搅拌16小时,LC-MS显示反应完全。将反应液冷却至0℃,加入水(10mL),浓缩除去溶剂后将所得残留物加二氯甲烷(200mL)溶解,水洗(100mL×3)后,有机相用无水硫酸钠干燥,旋干后得化合物C20-1(10.2g,灰色固体,收率98%)。
1H NMR(400MHz,CDCl 3)δ7.76(s,1H),7.53(d,J=8.4Hz,1H),7.33(d,J=8.5Hz,1H),6.79(s,1H),5.89(s,2H),4.64(t,J=10.5Hz,4H),3.33(s,3H).MS m/z(ESI):380.6,382.7[M+Na] +.
第二步:
将化合物C20-1(10.2g,28.4mmol)、联硼酸频那醇酯(7.94g,31.3mmol)和醋酸钾(8.35g,85.2mmol)溶于1,4-二氧六环(120mL)中,以N 2置换3次后,加入Pd(dppf)Cl 2(2.08g,2.84mmol),再次以N 2置换3次后加热至100℃回流反应16小时,LC-MS显示反应完全。冷却至室温,将反应液减压浓缩得到粗品,将粗品经硅胶柱色谱法(石油醚∶乙酸乙酯=4∶1)分离得到化合物C20-2(11g,白色固体,收率95.4%)。
1H NMR(400MHz,DMSO-d 6)δ7.94(d,J=4.8Hz,1H),7.67(d,J=7.9Hz,1H),7.47(d,J=7.9Hz,1H),7.13(s,1H),5.89(s,2H),4.72(d,J=122.5Hz,4H),3.15(s,3H),1.33(s,12H).MS m/z(ESI):429.1[M+Na] +.
实施例3:6-((2-(2-(3,3-二氟环丁烷-1-羰基)异吲哚啉-5-基)嘧啶-4-基)氨基)异喹啉-1(2H)-酮(C54)的制备
Figure PCTCN2019116453-appb-000044
第一步:
将化合物C54-1(20g,67mmol)溶于二氯甲烷(20mL)中,加入三氟乙酸(50mL),反应液在室温下搅拌16小时,真空旋蒸除去反应溶剂和三氟乙酸,残留物加入20mL水和40mL浓盐酸析出固体,过滤干燥得到化合物C54-2(15.3g,灰色固体)。
1H NMR(400MHz,CDCl 3)δ7.39(s,1H),7.34(d,J=8.2Hz,1H),7.12(d,J=8.0Hz,1H),4.22(d,J=14.1Hz,4H).MS m/z(ESI):197.9,199.8[M-Cl] +.
第二步:
将3,3-二氟环丁酸(3.1g,23mmol)溶于N,N-二甲基甲酰胺(100mL),依次加入HATU(9.5g,25mmol)、二异丙基乙胺(8.7g,67mmol)和C54-2(4.5g,19mmol),反应物在室温下继续搅拌16小时,真空旋蒸除去反应溶剂,残留物溶于20mL二氯甲烷,水洗(20mL×3)后浓缩有机相至有固体析出,待固体完全析出后过滤,干燥得C54-3粗品(4.2g,灰色固体,收率70%)。
MS m/z(ESI):315.7,317.8[M+H] +.
第三步:
将化合物C54-3(4.1g,13mmol)和联硼酸频那醇酯(4.1g,16mmol)溶于N,N-二甲基甲酰胺(100mL)中,加入乙酸钾(4.1g,16mmol)和Pd(dppf)Cl 2(0.95g,1.3mmol),氮气置换三次,反应液在110℃下搅拌16小时。LC-MS检测显示反应完成,反应液冷却至室温,真空旋蒸除去反应溶剂。残留物通过硅胶柱色谱法分离纯化(石油醚∶乙酸乙酯=5∶1),得到化合物C54-4(3.8g,白色固体,收率81%)。
1H NMR(400MHz,CDCl 3)δ7.73-7.79(m,2H),728-7.35(m,1H),4.78-4.85(m,4H),3.09-3.17(m,1H),2.94-3.05(m,2H),2.80-2.85(m,2H),1.38(s,12H).MS m/z(ESI):363.9[M+H] +.
第四步:
将化合物C54-4(545mg,1.5mmol)、2-氯嘧啶-4-胺(176.3mg,1.5mmol)、碳酸钾(621mg,4.5mmol)和Pd(dppf)Cl 2(110mg,0.15mmol)混和于二氧六环(25mL)与水(2.5mL)中,氮气置换3次,在110℃油浴中反应过夜。LC-MS显示反应完全。将反应液冷却至室温,过滤,滤液减压浓缩,将粗品经combi-flash硅胶柱色谱法(甲醇/二氯甲烷=0~5%)分离得化合物C54-5(300mg,浅黄色固体,收率60.6%)。
1H NMR(300MHz,DMSO-d 6)δ8.37-8.16(m,3H),7.45-7.35(m,1H),6.92(s,2H),6.36(d,J=5.6Hz,1H),4.86(s,2H),4.73(s,2H),3.28-3.24(m,1H),2.90-2.79(m,4H).
第五步:
将化合物C54-5(100mg,0.3mmol)、6-溴异喹啉-1(2H)-酮(68mg,0.3mmol)、碳酸铯(196mg,0.6mmol)、Pd 2(dba) 3(27.5mg,0.03mmol)和Xantphos(17.5mg,0.03mmol)混和于二氧六环(2mL)中,氮气置换3次,在110℃油浴中反应过夜。LC-MS显示反应完全。冷至室温,过滤以除掉难溶性盐,将滤液减压浓缩,所得粗品用制备HPLC纯化(ACN/H 2O(0.1%TFA),20~60%),得到化合物C54(8mg,黄色固体,收率4.5%)。
1H NMR(400MHz,DMSO-d 6)δ11.09(d,J=4.0Hz,1H),10.24(s,1H),8.49(dd,J=8.0,2.0Hz,1H),8.34(s,1H),8.30(d,J=12.0Hz,1H),8.21(t,J=2.0Hz,1H),8.19-8.12(m,1H),7.84-7.73(m,1H),7.54(dd,J=16.0,8.0Hz,1H),7.20-7.15(m,1H),6.88(d,J=8.0Hz,1H),6.52(d,J=8.0Hz,1H),4.92(d,J=12.0Hz,2H),4.77(d,J=17.0Hz,2H),3.34-3.27(m,1H),2.90-2.81(m,4H).MS m/z(ESI):473.6[M+H] +.
通过与实施例3中所述类似方法,制备下表中的化合物。
Figure PCTCN2019116453-appb-000045
Figure PCTCN2019116453-appb-000046
实施例4:5-(5-氟-4-((1-氧代-1,2-二氢异喹啉-6-基)氨基)嘧啶-2-基)异吲哚啉-2-甲酸3,3-二氟环丁酯(C10)和6-((5-氟-2-(2-(4,4,4-三氟-3-甲基丁酰基)异吲哚啉-5-基)嘧啶-4-基)氨基)异喹啉-1(2H)-酮(C37)的制备
Figure PCTCN2019116453-appb-000047
第一步:
将化合物C10-1(46g,0.15mol)和联硼酸频那醇酯(46g,0.18mol)溶于N,N-二甲基甲酰胺(800mL)中,加入乙酸钾(46g,0.47mol)和Pd(dppf)Cl 2(10,14mmol),氮气置换三次,反应液在110℃下搅拌16小时。LC-MS检测显示反应完成,反应液冷却至室温,真空旋蒸除去反应溶剂。残留物通过硅胶柱色谱法分离纯化(石油醚∶乙酸乙酯=20∶1~5∶1),得到化合物C10-2(37g,白色固体,66%收率)。
1H NMR(400MHz,CDCl 3)δ7.67-7.72(m,2H),7.22-7.29(m,1H),4.62-4.69(m,4H),1.52(s,9H),1.35(s,12H).MS m/z(ESI):367.9[M+Na] +.
第二步:
将化合物C10-2(1.0g,2.90mmol)和2氯-5-氟嘧啶-4-胺(427mg,2.89mmol)溶于1,4-二氧六环(30mL)中,加水(3mL)、碳酸钾(1.2g,8.7mmol)和Pd(dppf)Cl 2(636mg,0.87mmol),氮气置换三次,反应液在110℃下搅拌16小时,LC-MS检测显示反应完成,反应液冷却至室温,减压浓缩除去溶剂。残留物通过硅胶柱色谱法分离纯化(石油醚∶乙酸乙酯=20∶1~5∶1),得到化合物C10-3(700mg,白色固体,收率66%)。
1H NMR(400MHz,DMSO-d 6)δ8.25(d,J=3.3Hz,1H),8.19-8.12(m,2H),7.40(s,1H),7.34(s,2H),4.63(d,J=8.7Hz,5H),1.07(s,10H).MS m/z(ESI):330.9[M+H] +.
第三步:
将化合物C10-3(560mg,1.7mmol)和6-溴异喹啉-1(2H)-酮(380mg,0.18mol)溶于1,4-二氧六环(20mL)中,加入碳酸铯(1.66g,5.09mmol)、Pd 2(dba) 3(62mg,0.68mmol)和Xantphos(1.18g,2.03mmol),氮气置换三次,反应液在110℃下搅拌16小时,LC-MS检测显示反应完成,反应液冷却至室温,过滤,真空旋蒸除去反应溶剂,得到化合物C10-4(340mg,黄色固体,收率66%)。
MS m/z(ESI):473.8[M+H] +.
第四步:
将化合物C10-4(340mg,0.72mmol)溶于二氯甲烷(10mL)中,加入三氟乙酸(5mL),反应液在室温下搅拌3小时。LC-MS检测显示反应完成,反应液冷却至室温,真空旋蒸除去反应溶剂,得到化合物C10-5粗品(500mg,黄色固体)。
MS m/z(ESI):374.1[M+H] +.
第五步-A:
将3,3-二氟环丁醇(32mg,0.29mmol)和二(对硝基苯)碳酸酯(81mg,0.27mmol)溶于N,N-二甲基甲酰胺(10mL)中,加入三乙胺(162mg,1.61mmol),反应液在室温下搅拌2h,加入化合物C10-5(100mg,0.27mol),反应液继续在室温下搅拌16小时。LC-MS检测显示反应完成,反应液冷却至室温,真空旋蒸除去反应溶剂。残留物通过制备色谱法分离纯化,得到化合物C10(12.32mg,黄色固体,收率9%)。
1H NMR(400MHz,DMSO-d 6)δ11.14(s,1H),10.04(s,1H),8.58(s,1H),8.33-8.16(m,4H),7.95(dd,J=20.3,8.6Hz,1H),7.50(s,1H),7.21-7.16(m,1H),6.52(d,J=6.8Hz,1H),4.92(s,1H),4.75(dd,J=22.6,13.7Hz,4H),3.13-3.05(m,2H),2.82-2.72(m,2H).MS m/z(ESI):507.8[M+H] +.
第五步-B:
将化合物C10-5(100mg,0.268mmol)和HATU(112mg,0.295mmol)溶于N,N-二甲基甲酰胺(5mL)中,加入4,4,4-三氟3-甲基丁酸(42mg,0.268mmol)和二异丙基乙胺(173mg,1.339mmol),室温搅拌16小时。反应液加水(10mL),用二氯甲烷(20mL×3)萃取,有机相减压浓缩后所得残留物用制备HPLC纯化,得到化合物C37(90.15mg,白色固体,收率65%)。
1H NMR(400MHz,DMSO-d 6)δ11.14(s,1H),10.05(s,1H),8.60(s,1H),8.31-8.20(m,4H),7.96(dd,J=25.8,9.0Hz,1H),7.55-7.46(m,1H),7.23-7.13(m,1H),6.52(t,J=6.1Hz,1H),4.97(t,J=9.3Hz,2H),4.76(d,J=16.8Hz,2H),2.93(s,1H),2.83-2.62(m,2H),1.16(d,J=6.7Hz,3H).MS m/z(ESI):511.6[M+H] +.
通过与实施例4中所述类似方法,制备下表中的化合物。
Figure PCTCN2019116453-appb-000048
Figure PCTCN2019116453-appb-000049
Figure PCTCN2019116453-appb-000050
Figure PCTCN2019116453-appb-000051
Figure PCTCN2019116453-appb-000052
Figure PCTCN2019116453-appb-000053
Figure PCTCN2019116453-appb-000054
Figure PCTCN2019116453-appb-000055
实施例5:6-((2-(2-(4,4-二氟环己烷-1-羰基)异吲哚啉-5-基)-5-氟嘧啶-4-基)氨基)异喹啉-1(2H)-酮(C13)的制备
Figure PCTCN2019116453-appb-000056
第一步:
将化合物C13-1(20g,67mmol)溶于二氯甲烷(20mL)中,加入三氟乙酸(50mL),反应液在室温下搅拌16小时,真空旋蒸除去反应溶剂和三氟乙酸,残留物加入水(20mL)和浓盐酸(40mL)析出固体,过滤干燥得到化合物C13-2(15.3g,灰色固体,收率97%)。
1H NMR(400MHz,CDCl 3)δ7.39(s,1H),7.34(d,J=8.2Hz,1H),7.12(d,J=8.0Hz,1H),4.22(d,J=14.1Hz,4H).MS m/z(ESI):197.9,199.8[M-Cl] +.
第二步:
将化合物C13-2(932mg,4mmol)和4,4-二氟环己烷-1-羧酸(656mg,4mmol)溶于N,N-二甲基甲酰胺(30mL)中,加入HATU(1.69g,4.4mmol)和二异丙基乙胺(1.55g,12mmol),在室温下反应过夜。LC-MS显示反应完全。将反应液减压浓缩,残留物溶于二氯甲烷(20mL),用水(20mL×3)洗后旋干有机相,残留物通过硅胶柱色谱法分离(石油醚∶乙酸乙酯=2∶1)纯化得到化合物C13-3(1.1g,白色固体,收率80%)。
1H NMR(400MHz,CDCl 3)δ7.43(dd,J=15.6,8.7Hz,2H),7.16(dd,J=21.7,8.1Hz,1H),4.80(dd,J=33.8,14.4Hz,4H),2.54-2.49(m,1H),2.34-2.17(m,1H),2.03-1.71(m,3H).MS m/z(ESI):343.8,345.8[M+H] +.
第三步:
将化合物C13-3(1.16g,3.37mmol)和联硼酸频那醇酯(941mg,3.7mmol)溶于1,4-二氧六环(35mL)中,加入乙酸钾(980mg,10mmol),用氮气置换3次,加入Pd(dppf)Cl 2(732mg,1.0mmol),用氮气置换3次,将反应物置于90℃油浴中反应过夜。LC-MS检测显示反应完全。将反应液冷却至室温,减压浓缩,将残留物通过硅胶柱色谱法分离(石油醚∶乙酸乙酯=4∶1)纯化得到化合物C13-4(1.3g,白色固体,收率98.6%)。
1H NMR(400MHz,DMSO-d 6)δ7.67-7.59(m,2H),7.36(dd,J=12.2,7.6Hz,1H),4.94(d,J=12.4Hz,2H),4.65(d,J=5.4Hz,2H),2.71(t,J=11.2Hz,1H),2.08(s,2H),1.90(dd,J=21.2,9.7Hz,4H),1.64(d,J=11.5Hz,2H),1.30(s,12H).MS m/z(ESI):391.8[M+H] +.
第四步:
将化合物C13-5(500mg,3.12mmol)和2,4-二氯-5-氟嘧啶(520mg,3.12mmol)加入叔丁醇(12mL)中,加入三氟乙酸(3.56g,31.2mmol),将反应物置于100℃油浴中反应24小时。将反应液冷却至室温,减压浓缩,将残留物通过combi-flash色谱法(石油醚∶乙酸乙酯=4∶1)纯化得到化合物C13-6(420mg,黄色固体,收率46%)。MS m/z(ESI):290.8[M+H] +.
第五步:
将化合物C13-4(129mg,0.33mmol)和C13-6(96mg,0.33mmol)溶于1,4-二氧六环/水(10∶1)的混合液(16.5mL)中,加入碳酸钾(138mg,0.99mmol),氮气置换3次,加入Pd(dppf)Cl 2(74mg,0.099mmol),氮气置换3次,将反应物置于110℃油浴中反应过夜。LC-MS检测显示反应完全。将反应液冷却至室温,减压浓缩,将残留物通过制备薄层色谱法和高效液相色谱法纯化得到化合物C13(11.3mg,淡黄色固体,收率7%)。
1H NMR(400MHz,DMSO-d 6)δ11.14(s,1H),10.05(s,1H),8.58(s,1H),8.33-8.14(m,4H),7.97(dd,J=40.4,8.9Hz,1H),7.50(t,J=8.7Hz,1H),7.19(dd,J=12.5,6.2Hz,1H),6.51(d,J=6.0Hz,1H),5.02(d,J=9.5Hz,2H),4.72(d,J=15.9Hz,2H),2.74(s,1H),2.09(s,2H),2.01-1.85(m,4H),1.70-1.60(m,2H).MS m/z(ESI):519.5[M+H] +.
通过与实施例5中所述类似方法,制备下表中的化合物。
Figure PCTCN2019116453-appb-000057
实施例6:2-(5-氟-4-((1-氧代-1,2-二氢异喹啉-6-基)氨基)嘧啶-2-基)-6,7-二氢噻吩并[3,2-c]吡啶-5(4H)-甲酸3,3-二氟环丁酯(C27)和6-((5-氟-2-(5-(4,4,4-三氟-3-甲基丁酰基)-4,5,6,7-四氢噻吩并[3,2-c]吡啶-2-基)嘧啶-4-基)氨基)异喹啉-1(2H)-酮(C47)的制备
Figure PCTCN2019116453-appb-000058
第一步:
将化合物C27-1(400mg,1.257mmol)溶于无水四氢呋喃(10mL)中,氮气置换三次,将反应液降温至-78℃,加入正丁基锂(1.3M在己烷中,1.2mL),继续在-78℃搅拌2小时。加入异丙醇频哪醇硼酸酯(257mg,1.383mmol),缓慢升温至室温,搅拌16小时。LC-MS检测显示反应完成,加水(3mL)淬灭,真空旋蒸除去反应溶剂,得到化合物C27-2(450mg,黄色固体,收率96%)。MS m/z(ESI):387.8[M+Na] +.
第二步:
向化合物C27-2(450mg,1.232mmol)、2-氯-5-氟嘧啶-4-胺(182mg,1.232mmol)和磷酸钾(784mg,3.700mmol)的混合物加入正丁醇(10mL)和Pd(dppf)Cl 2(450mg,0.616mmol),氮气置换三次后,将反应置于微波反应器中,在110℃条件下搅拌30分钟。LC-MS检测显示反应完成,反应液冷却至室温,真空旋蒸除去反应溶剂。残留物通过flash柱色谱法(EA/PE,0~30%)进行纯化,得到化合物C27-3(140mg,白色固体,收率32%)。
1H NMR(400MHz,DMSO-d 6)δ7.20(s,1H),6.70(s,1H),3.69(s,2H),2.94(s,2H),2.05(s,2H),0.71(s,9H).MS m/z(ESI):350.7[M+H] +.
第三步:
向化合物C27-3(140mg,0.400mmol)、6-溴异喹啉-1(2H)-酮(107mg,0.479mmol)、碳酸铯(390mg,1.199mmol)和Xantphos(1.18g,2.03mmol)的混合物加入1,4-二氧六环(15mL)和Pd 2(dba) 3(62mg,0.68mmol),氮气置换三次,反应液在110℃下搅拌16小时。LC-MS检测显示反应完成,反应液冷却至室温,过滤除去不溶固体,所得滤液真空旋蒸除去反应溶剂。残留物通过flash柱色谱法(MeOH/DCM,0~5%)进行纯化,得到化合物C27-4(64mg,黄色固体,收率32%)。
1H NMR(400MHz,DMSO-d 6)δ11.13(s,1H),10.01(s,1H),8.47(d,J=3.4Hz,1H),8.30(s,1H),8.18(d,J=8.9Hz,1H),7.93(d,J=8.4Hz,1H),7.61(s,1H),7.18(t,J=6.2Hz,1H),6.52(d,J=7.0Hz,1H),4.48(s,2H),3.66(t,J=5.4Hz,2H),2.85(s,2H),1.45(s,9H).MS m/z(ESI):493.6[M+H] +.
第四步:
将化合物C27-4(64mg,0.142mmol)溶于二氯甲烷(10mL)中,加入氯化氢/1,4-二氧六环溶液(4M,5mL),反应液在室温下搅拌16小时。LC-MS检测显示反应完成,减压浓缩,除去溶剂,得到粗品化合物C27-5(80mg,黄色固体)。MS m/z(ESI):393.7[M-Cl] +.
第五步-A:
将3,3-二氟环丁醇(18mg,0.168mmol)和二(对硝基苯)碳酸酯(47mg,0.153mmol)溶于N,N-二甲基甲酰胺(10mL)中,加入三乙胺(93mg,0.915mmol),反应液在45℃搅拌2小时,加入化合物C27-5(60mg,0.153mmol)。反应液在45℃搅拌16h,LC-MS检测显示反应完成,反应液冷却至室温,真空旋蒸除去溶剂。残留物通过制备色谱法(ACN/H 2O(0.1%FA),45~60%)分离纯化,得到化合物C27(16.9mg,白色固体,收率21%)。
1H NMR(400MHz,DMSO-d 6)δ11.13(d,J=5.8Hz,1H),10.01(s,1H),8.47(d,J=3.4Hz,1H),8.28(s,1H),8.18(d,J=8.8Hz,1H),7.93(d,J=7.6Hz,1H),7.62(s,1H),7.18(t,J=6.4Hz,1H),6.52(d,J=7.2Hz,1H),4.88(s,1H),4.55(d,J=23.1Hz,2H),3.72(s,2H),3.10-3.00(m,2H),2.89(s,2H),2.77(td,J=14.4,4.8Hz,2H).MS m/z(ESI):527.8[M+H] +.
第五步-B:
将C27-5(60mg,0.153mmol)和HATU(55mg,0.168mmol)溶于N,N-二甲基甲酰胺(5mL)中,加入4,4,4-三氟-3-甲基丁酸(24mg,0.153mmol)和二异丙基乙胺(99mg,0.763mmol),反应液在室温搅拌16小时。LC-MS检测显示反应完成,反应液冷却至室温,真空旋蒸除去反应溶剂。残留物通过制备色谱(ACN/H 2O(0.1%TFA),40~70%)进行分离纯化,得到化合物C47(18.83mg,白色固体,收率23%)。
1H NMR(400MHz,DMSO-d 6)δ11.13(s,1H),10.01(s,1H),8.47(d,J=3.6Hz,1H),8.30(d,J=14.9Hz,1H),8.18(d,J=8.9Hz,1H),7.93(t,J=9.7Hz,1H),7.63(d,J=11.2Hz,1H),7.21-7.17(m,1H),6.53(d,J=6.4Hz,1H),4.67-4.61(m,2H),3.81(d,J=23.9Hz,3H),2.96(s,2H),2.85(s,2H),1.11(dd,J=6.8,3.6Hz,3H).MS m/z(ESI):531.6[M+H] +.
通过与实施例6中所述类似方法,制备下表中的化合物。
Figure PCTCN2019116453-appb-000059
Figure PCTCN2019116453-appb-000060
Figure PCTCN2019116453-appb-000061
Figure PCTCN2019116453-appb-000062
实施例7:(6-(4-((1-氨基异喹啉-6-基)氨基)嘧啶-2-基)-1-甲基-1H-吲哚-2-基)(3,3-二氟氮杂环丁烷-1-基)甲酮(C38)的制备
Figure PCTCN2019116453-appb-000063
第一步:
将化合物C38-1(0.5g,2.23mmol)溶于三氯氧磷(5mL),将反应液加热至110℃搅拌2小时,LC-MS检测显示反应完成,真空旋蒸除去溶剂,向残留物中加入水(10mL),以饱和碳酸氢钠水溶液中和,用乙酸乙酯(20mL×2)萃取,无水硫酸钠干燥,过滤,减压浓缩得到C38-2(530mg,黄色固体,收率97%)。
1H NMR(400MHz,DMSO-d 6)δ8.30(d,J=5.6Hz,1H),8.21(d,J=9.0Hz,1H),8.03(s,1H),7.77(d,J=9.0Hz,1H),7.52(d,J=5.6Hz,1H)。
第二步:
将化合物C38-2(530mg,2.18mmol)和乙酰胺(1.93g,32.7mmol)溶于NMP(2mL)中,加入碳酸钾(602mg,4.36mmol),加热至110℃搅拌2天。将反应液冷却后加水(20mL)稀释,用乙酸乙酯(15mL×2)萃取,有机层合并,以水(20mL)洗,饱和食盐水(20mL)洗,无水硫酸钠干燥,过滤,减压浓缩,残留物用Flash柱色谱法(甲醇/二氯甲烷,0~3%)纯化,得到化合物C38-3(60mg,黄色固体,收率12.3%)。
1H NMR(400MHz,DMSO-d 6)δ8.13(d,J=8.8Hz,1H),7.96(s,1H),7.82(d,J=5.7Hz,1H),7.58(d,J=8.9Hz,1H),6.91(s,2H),6.87(d,J=5.7Hz,1H)。
第三步:
将化合物C38-3(60mg,0.27mmol)和二碳酸二叔丁酯(71mg,0.33mmol)混合于四氢呋喃(1mL)中,室温搅拌下加入DMAP(33mg,0.27mmol),使反应于室温搅拌2天,LC-MS显示反应完全。减压蒸除溶剂得到C38-4粗品(150mg,黄褐色固体,收率100%),不经纯化直接用于下步反应。
第四步:
将化合物C38-4粗品(130mg,0.27mmol)、化合物C38-5(92.6mg,0.27mmol)和碳酸铯(264mg,0.81mmol)混合于1,4-二氧六环(5mL)中,以N 2置换3次后加入Pd 2(dba) 3(27.5mg,0.03mmol)和Xantphos(52mg,0.09mmol),再次以N 2置换3次后,置于100℃油浴中反应16小时。LC-MS显示有产物生成。将反应液冷却至室温,过滤,滤液减压浓缩至干,将残余物经制备硅胶薄层色谱法分离(甲醇∶二氯甲烷=1∶12)得C38-6(25mg,黄色固体,收率13.5%)。
MS m/z(ESI):686.1[M+H] +.
第五步:
将C38-6(25mg,0.036mmol)溶于二氯甲烷(2mL)中,室温搅拌下加入TFA(0.3mL),使反应于室温搅拌3小时,LC-MS显示反应完全。减压蒸除溶剂,将粗产品经制备液相色谱法(乙腈/水(0.1% TFA),20~60%,30分钟)分离得到C38(13mg,黄色固体,收率60.2%)。
1H NMR(400MHz,DMSO-d 6)δ12.72(s,1H),10.50(s,1H),8.82(s,2H),8.61(d,J=3.6Hz,1H),8.59(s,1H),8.55(d,J=9.2Hz,1H),8.22(d,J=8.3Hz,1H),8.01(d,J=9.6Hz,1H),7.79(d,J=8.4Hz,1H),7.66(d,J=7.0Hz,1H),7.21(d,J=7.0Hz,1H),7.10(s,1H),6.93(d,J=8.0Hz,1H),4.87(s,2H),4.58(s,2H),4.07(s,3H).MS m/z(ESI):486.1[M+H] +.
通过与以上各实施例类似的方法制备以下化合物:
Figure PCTCN2019116453-appb-000064
Figure PCTCN2019116453-appb-000065
生物学测试
采用商品化的CISBIO激酶检测试剂盒HTRF KinEASE-STK S2试剂盒(62ST2PEC)进行激酶IC 50测定。反应所用的激酶ROCK2(01-119)购自Carna Biosciences。
实验开始前,根据需要,按照激酶检测试剂盒说明书用相应试剂配制如下工作液:1×激酶缓冲液,5×STK-S2底物工作液(1.5μM)和5×ATP工作液(1.5μM)、5×ROCK2激酶工作液、4×抗生蛋白链菌素-XL665工作液、4×STK-Ab-穴状化合物2检测液,然后进行如下操作。
采用DMSO浓度为2.5%的1×激酶缓冲液配制得到10000nM的化合物溶液,采用上述含DMSO的激酶缓冲液再对化合物进行逐级稀释,得到9个不同浓度的待测化合物溶液。除测试化合物孔外,设置阳性孔(含有除化合物之外的所有试剂)和阴性孔(含有除待测化合物和激酶外的所有试剂)。除对照孔(阳性孔和阴性孔)外,向所有反应孔中加入4μL的待测化合物溶液,向对照孔中加入4μL 2.5%的DMSO溶液。然后,再向所有反应孔中加入2μM底物(即2μL 5×STK-S2底物工作液)。向除阴性孔外的所有反应孔中加入2μL的5×ROCK2激酶工作液(含1.4ng ROCK2激酶),阴性孔用2μL的1×激酶缓冲液补足体积。向所有反应孔中加入2μL 5×ATP工作液,室温孵育2小时。待激酶反应结束后,向所有反应孔中加入5μL4×抗生蛋白链菌素-XL665工作液,混匀后立即加入5μL 4×STK-Ab-穴状化合物2检测液,室温孵育1小时后,用ENVISION(Perkinelmer)仪器检测荧光信号(激发波长为320nm,发射波长为665nm和615nm)。通过各孔荧光强度值计算出每个孔中的抑制率:ER(Emission Ratio,发射光比率)=(665nm处荧光强度/615nm处荧光强度);抑制率=(ER阳性-ER待测化合物)/(ER阳性-ER阴性)*100%,用软件PRISM 5.0绘制曲线图,同时用PRISM 5.0拟合得到各待测化合物的半数抑制浓度(IC 50)。依照与上述类似的生物学测试方法,还测试了化合物对ROCK1的IC 50值。测试结果如下表中所示。
化合物编号 ROCK2 IC 50(nM) ROCK1 IC 50(nM)
C1 129 >10000
C3 287 >10000
C5 13 >10000
C6 76 >10000
C8 16 >10000
C9 4 >10000
C10 11 >10000
C11 34 >10000
C12 17 >10000
C13 38 >10000
C14 28 >10000
C15 79 >10000
C16 19 >10000
C17 421 >10000
C18 31 >10000
C19 25 >10000
C20 238 >10000
C21 108 >10000
C22 130 >10000
C23 173 >10000
C24 222 >10000
C25 45 >10000
C26 356 >10000
C27 8 >10000
C28 6 >10000
C30 216 >10000
C35 93 >10000
C37 117 >10000
C38 264 >10000
C39 102 >10000
C40 270 >10000
C41 151 >10000
C42 31 >10000
C43 71 >10000
C44 57 >10000
C45 491 >10000
C46 21 >10000
C47 15 >10000
C48 13 >10000
C49 7 >10000
C50 280 >10000
C51 53 >10000
C52 28 >10000
C53 135 >10000
C54 17 >10000
C55 51 >10000
C56 24 >10000
C57 16 >10000
C59 33 >10000
C60 13 >10000
C61 170 >10000
C62 139 >10000
C63 24 >10000
C64 109 >10000
C65 80 >10000
C66 82 >10000
C67 101 >10000
C68 9 >10000
C69 45 >10000
C70 15 >10000
C71 25 >10000
C72 24 >10000
C73 78 >10000
C74 54 >10000
C75 37 >10000
C76 49 >10000
C78 40 >10000
C79 46 >10000
C81 129 >10000
C82 129 >10000
由以上数据可见,所测试化合物对ROCK2的IC 50值显著低于对ROCK1的IC 50值,这表明本发明化合物对于ROCK2具有良好的选择性。
除本文中描述的那些外,根据前述描述,本发明的各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。

Claims (12)

  1. 化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中所述化合物具有式(I)的结构:
    Figure PCTCN2019116453-appb-100001
    其中:
    V 1选自-CR 2=N-、-N=CR 2-、-NR 3C(=O)-和-C(=O)NR 3-;
    V 2选自-CR 2=N-、-N=CR 2-、-(CR 2R 2’) n-和-(CR 2=CR 2’) n-;
    Y选自直接键、C(=O)、O、S(=O) i和NR;
    R选自H、C 1-6烷基、C 2-6烯基、C 2-6炔基、饱和或部分不饱和的C 3-10环烃基、饱和或部分不饱和的3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基,所述环烃基和杂环基中至多2个环成员为C(=O);
    环A和环B各自独立地选自饱和或部分不饱和的C 3-10烃环、饱和或部分不饱和的3-10元杂环、C 6-10芳环和5-14元杂芳环,所述烃环和杂环中至多2个环成员为C(=O);
    环C为
    Figure PCTCN2019116453-appb-100002
    其通过*或**标记的两个位置之一与Y连接,并且另一位置与环B连接;
    X选自CR 8和N;
    R 1选自-C(=O)R 1a、-C(=O)OR 1a和-C(=O)NR 1aR 1b
    R 1a和R 1b各自独立地选自H、卤素、氨基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 3-10环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基、C 6-12芳烷基、-C(=O)R 5、-OC(=O)R 5、-C(=O)OR 5、-OR 5、-SR 5、-S(=O)R 5、-S(=O) 2R 5、-S(=O) 2NR 5R 6、-NR 5R 6、-C(=O)NR 5R 6、-NR 5-C(=O)R 6、-NR 5-C(=O)OR 6、-NR 5-S(=O) 2-R 6、-NR 5-C(=O)-NR 5R 6、-C 1-6亚烷基-NR 5R 6、-C 1-6亚烷基-OR 5和-O-C 1-6亚烷基-NR 5R 6;或者R 1a和R 1b连同其所连接的原子共同构成3-12元杂环或杂芳环;
    R 2、R 2’、R 3、R 4、R 7、R 8、R 9和R 10在每次出现时各自独立地选自H、卤素、氨基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 3-10环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基、C 6-12芳烷基、-C(=O)R 5、-OC(=O)R 5、-C(=O)OR 5、-OR 5、-SR 5、-S(=O)R 5、-S(=O) 2R 5、-S(=O) 2NR 5R 6、-NR 5R 6、-C(=O)NR 5R 6、-NR 5-C(=O)R 6、-NR 5-C(=O)OR 6、-NR 5-S(=O) 2-R 6、-NR 5-C(=O)-NR 5R 6、-C 1-6亚烷基-NR 5R 6、-C 1-6亚烷基-O(P=O)(OH) 2和-O-C 1-6亚烷基-NR 5R 6;或者R 7和R 8连同其所连接的基团共同构成C 6-10芳环或5-14元杂芳环;
    上述烷基、亚烷基、烯基、亚烯基、炔基、环烃基、烃环、杂环基、杂环、芳基、芳环、杂芳基、杂芳环和芳烷基在每次出现时各自任选地被一个或多个独立地选自下列的取代基取代:卤素、羟基、氧代、氨基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 3-6环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基、C 6-12芳烷基、=N-OR 5、-C(=NH)NH 2、-C(=O)R 5、-OC(=O)R 5、-C(=O)OR 5、-OR 5、-SR 5、-S(=O)R 5、-S(=O) 2R 5、-S(=O) 2NR 5R 6、-NR 5R 6、-C(=O)NR 5R 6、-NR 5-C(=O)R 6、-NR 5-C(=O)OR 6、-NR 5-S(=O) 2-R 6、-NR 5-C(=O)-NR 5R 6、-C 1-6亚烷基-NR 5R 6和-O-C 1-6亚烷基-NR 5R 6,所述烷基、亚烷基、烯基、炔基、环烃基、杂环基、芳基、杂芳基和芳烷基进一步任选地被一个或多个独立地选自下列的取代基取代:卤素、羟基、氧代、氨基、氰基、硝基、C 1-6烷基、C 3-6环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基;
    R 5和R 6在每次出现时各自独立地选自H、C 1-6烷基、C 3-10环烃基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基;或者当R 5和R 6与同一氮原子连接时,R 5和R 6连同其所连接的原子任选地共同构成3-12元杂环或杂芳环;
    m在每次出现时各自独立地为0、1、2或3的整数;
    n为0、1、2、3或4的整数;并且
    i为0、1或2的整数。
  2. 权利要求1的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中Y选自直接键、C(=O)、O、S、S(=O)、S(=O) 2、NH和NCH 3
  3. 权利要求1或2的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中
    Figure PCTCN2019116453-appb-100003
    Figure PCTCN2019116453-appb-100004
    优选为
    Figure PCTCN2019116453-appb-100005
    Figure PCTCN2019116453-appb-100006
    以上基团通过#或##标记的两个位置之一与环C连接,并且另一位置与R 1连接,
    其中
    Figure PCTCN2019116453-appb-100007
    表示单键或双键且相邻的键不同时为双键;
    Z 1、Z 2、Z 3、Z 4、Z 5、Z 6、Z 7、Z 8和Z 9在每次出现时各自独立地选自C、CR 9、C(R 9) 2、CR 10、C(R 10) 2、C(=O)、N、NR 9、NR 10、O和S;优选地,Z 1、Z 2、Z 3、Z 4、Z 5、Z 6、Z 7、Z 8和Z 9在每次出现时各自独立地选自C、CH、CF、CCl、CCH 3、CH 2、C(CH 3) 2、C-OCH 3、C(=O)、N、NH、NCH 3、NCH 2CH 3、NCH(CH 3) 2、NCH=CH 2、NCH 2F、NCHF 2、NCH 2CHF 2、NC(=O)CH 3、NCH 2OH、NCH 2OMe、NCH 2CH 2OMe、NCH 2-O(P=O)(OH) 2
    Figure PCTCN2019116453-appb-100008
    NCH 2CH 2-N(CH 3) 2、O和S;
    j为0、1、2、3或4;
    条件是Z 1-Z 9中的至多两个基团同时为C(=O);
    R 9和R 10在每次出现时各自独立地选自卤素、甲基、乙基、丙基、乙烯基、环丙基、环丁基、环戊基、氧杂环丁烷基、单氟甲基、二氟甲基、三氟甲基、-CH 2CHF 2、乙酰基、-OCH 3、-CH 2OH、-CH 2OCH 3、-CH 2CH 2OCH 3、-CH 2-O(P=O)(OH) 2
    Figure PCTCN2019116453-appb-100009
    和-CH 2CH 2-N(CH 3) 2
    最优选地,
    Figure PCTCN2019116453-appb-100010
    选自
    Figure PCTCN2019116453-appb-100011
    Figure PCTCN2019116453-appb-100012
    以上基团通过#或##标记的两个位置之一与环C连接,并且另一位置与R 1连接。
  4. 权利要求1-3中任一项的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂 合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中环C选自
    Figure PCTCN2019116453-appb-100013
    Figure PCTCN2019116453-appb-100014
    以上基团通过*或**标记的两个位置之一与Y连接,并且另一位置与环B连接。
  5. 权利要求1-4中任一项的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中
    Figure PCTCN2019116453-appb-100015
    选自
    Figure PCTCN2019116453-appb-100016
    Figure PCTCN2019116453-appb-100017
    Figure PCTCN2019116453-appb-100018
    优选为
    Figure PCTCN2019116453-appb-100019
    Figure PCTCN2019116453-appb-100020
  6. 权利要求1-5中任一项的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中R 1选自
    Figure PCTCN2019116453-appb-100021
    Figure PCTCN2019116453-appb-100022
    Figure PCTCN2019116453-appb-100023
  7. 权利要求1-6中任一项的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中所述化合物具有任意下式的结构:
    Figure PCTCN2019116453-appb-100024
  8. 权利要求1-7中任一项的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,其中所述化合物具有以下结构:
    Figure PCTCN2019116453-appb-100025
    Figure PCTCN2019116453-appb-100026
    Figure PCTCN2019116453-appb-100027
    Figure PCTCN2019116453-appb-100028
    Figure PCTCN2019116453-appb-100029
    Figure PCTCN2019116453-appb-100030
    Figure PCTCN2019116453-appb-100031
    Figure PCTCN2019116453-appb-100032
    Figure PCTCN2019116453-appb-100033
    Figure PCTCN2019116453-appb-100034
    Figure PCTCN2019116453-appb-100035
  9. 药物组合物,其包含预防或治疗有效量的权利要求1-8中任一项的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药,以及药学上可接受的载体,所述药物组合物优选为固体制剂、半固体制剂、液体制剂或气态制剂。
  10. 权利要求1-8中任一项的化合物或其药学上可接受的盐、酯、立体异构体、多晶型物、溶剂合物、N-氧化物、同位素标记的化合物、代谢物或前药或者权利要求9的药物组合物在制备用作Rho相关蛋白激酶(ROCK)抑制剂,优选选择性ROCK2抑制剂的药物中的用途。
  11. 权利要求10的用途,其中所述药物用于预防或治疗Rho相关蛋白激酶(ROCK)介导的疾病,其包括自身免疫性疾病(其包括类风湿性关节炎、全身性红斑狼疮(SLE;狼疮)、银屑病、克罗恩病、特应性皮炎、湿疹或移植物抗宿主病(GVHD));心血管病症(其包括高血压、动脉粥样硬化、再狭窄、心脏肥大、脑缺血、脑血管痉挛或勃起功能障碍);炎症(其包括哮喘、心血管炎症、溃疡性结肠炎或肾脏炎症);中枢神经系统病症(其包括神经元变性或脊髓损伤;所述中枢神经系统病症优选为亨延顿 氏病、帕金森氏病、阿尔茨海默病、肌萎缩性脊髓侧索硬化症(ALS)或多发性硬化症);动脉血栓性病症(其包括血小板聚集或白细胞聚集);纤维化病症(其包括肝纤维化、肺纤维化或肾纤维化);瘤性疾病(其包括淋巴瘤、癌(例如鳞状细胞癌、小细胞肺癌、垂体癌、食道癌、非小细胞肺癌、肺腺癌、肺鳞癌、腹膜癌、肝细胞癌、胃肠癌、胰腺癌、胶质母细胞瘤、宫颈癌、卵巢癌、膀胱癌、肝癌、乳腺癌、结肠癌、结直肠癌、子宫内膜癌或子宫癌、唾液腺癌、肾癌、前列腺癌、外阴癌、甲状腺癌、脑癌、子宫内膜癌、睾丸癌、胆管癌、胆囊癌、胃癌、黑色素瘤或头颈部癌症)、白血病、星形细胞瘤、软组织肉瘤、肉瘤或母细胞瘤);代谢综合征;胰岛素抗性;高胰岛素血症;2型糖尿病;葡萄糖耐受不良;骨质疏松症;眼部病症(其包括高眼压、年龄相关性黄斑变性(AMD)、脉络膜新生血管形成(CNV)、糖尿病性黄斑水肿(DME)、虹膜新生血管形成、葡萄膜炎、青光眼(其包括原发性开角型青光眼、急性闭角型青光眼、色素性青光眼、先天性青光眼、正常张力青光眼、继发青光眼或新生血管性青光眼)或早产儿视网膜病变(ROP))。
  12. 权利要求10的用途,其中所述药物用于预防或治疗Rho相关蛋白激酶(ROCK)介导的疾病,其包括狼疮肾炎、动脉粥样硬化、类风湿性关节炎(RA)、血管瘤、血管纤维瘤、肺纤维化、银屑病、角膜移植排斥、胰岛素依赖型糖尿病、多发性硬化症、重症肌无力、克罗恩病、自身免疫性肾炎、原发性胆汁性肝硬化、急性胰腺炎、异体排异、过敏性炎症、接触性皮炎、迟发型超敏反应、炎症性肠病、感染性休克、骨质疏松、骨关节炎、神经元炎症、Osler-Weber综合征、再狭窄、真菌感染、寄生虫感染和病毒感染。
PCT/CN2019/116453 2018-11-09 2019-11-08 Rho相关蛋白激酶抑制剂、包含其的药物组合物及其用途 WO2020094111A1 (zh)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201980073246.2A CN112969694B (zh) 2018-11-09 2019-11-08 Rho相关蛋白激酶抑制剂、包含其的药物组合物及其用途
US17/292,036 US20220002266A1 (en) 2018-11-09 2019-11-08 Rho-associated protein kinase inhibitor, pharmaceutical composition comprising same, and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2018114801 2018-11-09
CNPCT/CN2018/114801 2018-11-09

Publications (1)

Publication Number Publication Date
WO2020094111A1 true WO2020094111A1 (zh) 2020-05-14

Family

ID=70610815

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/116453 WO2020094111A1 (zh) 2018-11-09 2019-11-08 Rho相关蛋白激酶抑制剂、包含其的药物组合物及其用途

Country Status (3)

Country Link
US (1) US20220002266A1 (zh)
CN (1) CN112969694B (zh)
WO (1) WO2020094111A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022012409A1 (zh) * 2020-07-14 2022-01-20 武汉朗来科技发展有限公司 一种rock抑制剂及其制备方法和用途
WO2023134688A1 (zh) * 2022-01-13 2023-07-20 武汉朗来科技发展有限公司 一种rock抑制剂的盐及盐的晶型、组合物和药物用途

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101208094A (zh) * 2005-03-25 2008-06-25 表面线段公司 药代动力学改善的化合物
CN101522633A (zh) * 2006-08-18 2009-09-02 欧加农股份有限公司 作为rock-i抑制剂的6-取代的异喹啉衍生物
CN101573338A (zh) * 2006-12-27 2009-11-04 塞诺菲-安万特股份有限公司 环烷基胺取代的异喹诺酮和异喹啉酮衍生物
WO2010011620A1 (en) * 2008-07-21 2010-01-28 Wyeth 4-phenoxy-6-aryl-1h-pyrazolo[3,4-d]pyrimidine and n-aryl-6-aryl-1h-pyrazolo[3,4-d]pyrimidin-4-amine compounds, their use as mtor kinase and pi3 kinase inhibitors, and their syntheses
CN103534249A (zh) * 2011-04-29 2014-01-22 阿玛克姆股份有限公司 新的rock抑制剂
CN104903312A (zh) * 2013-10-07 2015-09-09 卡德门企业有限公司 Rho激酶抑制剂

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11311541B2 (en) * 2014-04-09 2022-04-26 Kadmon Corporation, Llc Treatment of GVHD

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101208094A (zh) * 2005-03-25 2008-06-25 表面线段公司 药代动力学改善的化合物
CN101522633A (zh) * 2006-08-18 2009-09-02 欧加农股份有限公司 作为rock-i抑制剂的6-取代的异喹啉衍生物
CN101573338A (zh) * 2006-12-27 2009-11-04 塞诺菲-安万特股份有限公司 环烷基胺取代的异喹诺酮和异喹啉酮衍生物
WO2010011620A1 (en) * 2008-07-21 2010-01-28 Wyeth 4-phenoxy-6-aryl-1h-pyrazolo[3,4-d]pyrimidine and n-aryl-6-aryl-1h-pyrazolo[3,4-d]pyrimidin-4-amine compounds, their use as mtor kinase and pi3 kinase inhibitors, and their syntheses
CN103534249A (zh) * 2011-04-29 2014-01-22 阿玛克姆股份有限公司 新的rock抑制剂
CN104903312A (zh) * 2013-10-07 2015-09-09 卡德门企业有限公司 Rho激酶抑制剂

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022012409A1 (zh) * 2020-07-14 2022-01-20 武汉朗来科技发展有限公司 一种rock抑制剂及其制备方法和用途
EP4155308A4 (en) * 2020-07-14 2024-04-17 Wuhan LL Science and Technology Development Co., Ltd. ROCK INHIBITOR, METHOD FOR PREPARING IT AND ITS USE
WO2023134688A1 (zh) * 2022-01-13 2023-07-20 武汉朗来科技发展有限公司 一种rock抑制剂的盐及盐的晶型、组合物和药物用途

Also Published As

Publication number Publication date
CN112969694A (zh) 2021-06-15
CN112969694B (zh) 2023-06-13
US20220002266A1 (en) 2022-01-06

Similar Documents

Publication Publication Date Title
JP7311228B2 (ja) Rho-関連プロテインキナーゼ阻害剤、それを含む医薬組成物並びにその調製方法及び使用
CN110582491B (zh) Rho相关蛋白激酶抑制剂、包含其的药物组合物及其制备方法和用途
KR102563325B1 (ko) Rho-관련 단백질 키나아제 억제제, Rho-관련 단백질 키나아제 억제제를 함유하는 약학 조성물, 제조 방법 및 약학 조성물의 용도
CN111936475A (zh) 免疫调节剂及其组合物和制备方法
JP2021526551A (ja) キナーゼ阻害剤としてのヘテロ環式化合物、ヘテロ環式化合物を含む組成物、及びそれらを使用する方法
CN114728962A (zh) 血浆激肽释放酶抑制剂及其用途
TWI793704B (zh) Sos1抑制劑、包含其的藥物組合物及其用途
WO2022121813A1 (zh) Sos1抑制剂、包含其的药物组合物及其用途
WO2022199662A1 (zh) 一种多环化合物及其应用
WO2023202623A1 (zh) Polq抑制剂化合物及其应用
WO2023045960A1 (zh) 一种吡啶类衍生物及其用途
WO2020094111A1 (zh) Rho相关蛋白激酶抑制剂、包含其的药物组合物及其用途
WO2022237676A1 (zh) Shp2磷酸酶抑制剂的制备及其应用
WO2023165581A1 (zh) 一种吡啶类衍生物及其用途
WO2022089219A1 (zh) 芳基酰胺化合物、包含其的药物组合物及其制备方法和用途
KR20170032329A (ko) 피라졸 유도체의 제조 방법
WO2023165574A1 (zh) 用作tyk2抑制剂的化合物、其制备方法及其在医药上的应用
WO2023202642A1 (zh) Rho相关蛋白激酶抑制剂及其制备和应用
EP4358954A1 (en) Cdk2 inhibitors and methods of using the same

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: 19881478

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: 19881478

Country of ref document: EP

Kind code of ref document: A1