WO2022007882A1 - 一种atx抑制剂及其制备方法和应用 - Google Patents

一种atx抑制剂及其制备方法和应用 Download PDF

Info

Publication number
WO2022007882A1
WO2022007882A1 PCT/CN2021/105180 CN2021105180W WO2022007882A1 WO 2022007882 A1 WO2022007882 A1 WO 2022007882A1 CN 2021105180 W CN2021105180 W CN 2021105180W WO 2022007882 A1 WO2022007882 A1 WO 2022007882A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
amino
ethyl
methyl
pyridin
Prior art date
Application number
PCT/CN2021/105180
Other languages
English (en)
French (fr)
Inventor
彭程
钱梦飞
吕遐师
武进
尹伟
刘奉友
金羿
邹罡
袁海卿
邬征
Original Assignee
苏州爱科百发生物医药技术有限公司
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
Priority claimed from CN202010658085.8A external-priority patent/CN113912600A/zh
Application filed by 苏州爱科百发生物医药技术有限公司 filed Critical 苏州爱科百发生物医药技术有限公司
Priority to EP21838673.8A priority Critical patent/EP4180430A4/en
Priority to CN202180046308.8A priority patent/CN115916777A/zh
Priority to US18/015,261 priority patent/US20230295159A1/en
Priority to JP2023501105A priority patent/JP2023533294A/ja
Publication of WO2022007882A1 publication Critical patent/WO2022007882A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Definitions

  • the present invention requires an invention patent application with an application number of 202010658085.8 submitted in China on July 9, 2020, entitled “An ATX inhibitor and its preparation method and application”, and an invention patent application submitted in China on March 23, 2021, The priority of the invention patent application entitled “An ATX inhibitor and its preparation method and application” with the application number of 202110308656.X is incorporated herein by reference in its entirety.
  • the invention belongs to the field of medicinal chemistry, in particular to an ATX inhibitor and a preparation method thereof.
  • Autotaxin (ATX, also known as ENPP2 [exonucleotide pyrophosphatase/phosphodiesterase 2] or lysophospholipase D) is a member of the exonucleotide pyrophosphatase/phosphodiesterase (ENPP) family.
  • ATX is capable of converting lysophosphatidylcholine (LPC) to biologically active lysophosphatidic acid (LPA).
  • LPA consists of a glycerol chain, a phosphate group, and a fatty acyl chain of varying length and saturation. Once produced, LPA can exert its biological activity mediated by six cell surface-specific receptor proteins (LPA1-6), namely G protein-coupled receptors.
  • LPC lysophosphatidylcholine
  • LPA biologically active lysophosphatidic acid
  • LPA consists of a glycerol chain, a phosphate group, and a fatty acyl chain
  • ATX is the major source of LPA in the blood.
  • the ATX-LPA signaling pathway is involved in cell survival, migration, and proliferation, and thus has important links to many serious diseases, such as fibrotic diseases (mainly idiopathic pulmonary fibrosis, IPF), cancer, proliferative diseases, inflammatory diseases, autologous Immune diseases, cardiovascular diseases, neurodegenerative diseases, etc.
  • fibrotic diseases mainly idiopathic pulmonary fibrosis, IPF
  • cancer proliferative diseases
  • inflammatory diseases mainly autologous Immune diseases
  • cardiovascular diseases mainly glioblastoma.
  • GLPG-1690 has inhibitory effect on ATX and has entered the phase III clinical trial for the treatment of idiopathic pulmonary fibrosis (IPF).
  • IPF idiopathic pulmonary fibrosis
  • GLPG-1690 has inhibitory activity on ATX, its inhibitory activity is not very high. Therefore, more inhibitors with higher inhibitory activity on ATX still need to be developed.
  • the present invention provides a new class of heteroaromatic ring compounds, which have high inhibitory activity against ATX, and at the same time have excellent pharmacodynamics, in vitro/in vivo pharmacokinetic properties and safety.
  • the application prospect is high.
  • the present invention provides the following technical solutions:
  • n is any integer from 0 to 5;
  • each R 1 is independently selected from hydrogen, deuterium, cyano, halogen, amino, hydroxy, -COOH, -CHO, -NO 2 , C 1-6 alkylamino, C 1-6 alkoxy and C 1-6 alkyl, C 3-7 cycloalkyl, unsubstituted or substituted with substituents selected from cyano, amino, hydroxy, halogen, C 1-6 alkyl and C 1-6 alkoxy and 3-7 membered heterocycloalkyl;
  • R 3 is selected from hydrogen, deuterium, C 1-6 alkyl, C 3-7 cycloalkyl and selected from cyano, amino, hydroxyl, halogen, C 1-6 alkyl and C 1-6 alkoxy C 1-6 alkyl, C 3-7 cycloalkyl and 3-7 membered heterocycloalkyl substituted by substituents;
  • Ring B is any of the following structures:
  • R 4 is selected from hydrogen, deuterium, halogen, cyano, hydroxyl, amino, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkylamino, C 3-7 cycloalkyl and 3- 7-membered heterocycloalkyl;
  • X 1 , X 2 , X 3 and X 4 are each independently N or CR 5 ;
  • R 5 is selected from hydrogen, deuterium, halo, C 1-6 alkyl and are selected from cyano, amino, hydroxy, halo, C 1-6 alkyl and C 1-6 alkoxy substituents of C 1-6 alkyl, C 3-7 cycloalkyl and 3-7 membered heterocycloalkyl;
  • Ring A is selected from substituted or unsubstituted C 6-10 aryl and 5-10 membered heteroaryl, the substitution is 1 to 3 selected from hydroxy, amino, halogen, cyano, C 1-6 alkyl , C 1-6 haloalkyl, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 1-6 alkylamino, C 1-6 alkoxy substituent;
  • R 6 is -L 1 -L 2 -W 1 ;
  • n, R 1 , R 2 , R 3 , R 4 , R 6 , X 1 , X 2 , X 3 and Ring A are as defined in formula I′.
  • n, R 1 , R 2 , R 3 , R 4 , R 6 and ring A are as defined in formula I'.
  • n, R 1 , R 2 , R 3 , R 4 , R 6 , X 1 , X 2 , X 3 and Ring A are as defined in formula I′.
  • n, R 1 , R 2 , R 3 , R 4 , R 6 and ring A are as defined in formula I'.
  • n, R 1 , R 2 , R 3 , R 4 , R 6 , X 1 , X 2 , X 3 and Ring A are as defined in formula I′.
  • n, R 1 , R 2 , R 3 , R 4 , R 6 and ring A are as defined in formula I'.
  • the present invention also provides the following compounds:
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising the above-mentioned compound having the structure of formula I', formula I, formula I-1, formula II, formula II-1, formula III or formula III-1 or pharmaceutically acceptable compounds thereof Accepted salts, esters, isomers, solvates, prodrugs or isotopic labels.
  • the present invention also provides a pharmaceutical preparation, which comprises the above-mentioned compound having the structure of formula I', formula I, formula I-1, formula II, formula II-1, formula III or formula III-1 or a pharmaceutically acceptable compound thereof
  • a pharmaceutical preparation which comprises the above-mentioned compound having the structure of formula I', formula I, formula I-1, formula II, formula II-1, formula III or formula III-1 or a pharmaceutically acceptable compound thereof
  • the salts, esters, isomers, solvates, prodrugs or isotopic labels or the above-mentioned pharmaceutical compositions, the preparations are tablets, capsules, injections, granules, powders, suppositories, pills, creams, pastes , any of gels, powders, oral solutions, inhalants, suspensions, dry suspensions, patches, and lotions.
  • the present invention also provides the above-mentioned compounds having the structures of formula I', formula I, formula I-1, formula II, formula II-1, formula III or formula III-1 or their pharmaceutically acceptable salts, esters, isomers body, solvate, prodrug or isotope label, or the above-mentioned pharmaceutical composition or pharmaceutical preparation, which are used for the prevention and/or treatment of related diseases with pathological features of increased ATX expression; preferably, the said with increased ATX expression Diseases associated with pathological features include: cancer, fibrotic disease, metabolic disease, myelodysplastic syndrome, respiratory disease, cardiovascular disease, autoimmune disease, inflammation, dermatological disease, neurological disease or pain; more Preferably, the related disease with pathological features of increased ATX expression is pulmonary fibrosis, renal fibrosis or liver fibrosis.
  • the present invention also provides the above-mentioned compounds having the structures of formula I', formula I, formula I-1, formula II, formula II-1, formula III or formula III-1 or their pharmaceutically acceptable salts, esters, isoforms Constituents, solvates, prodrugs or isotopic labels, or the use of the above-mentioned pharmaceutical compositions or pharmaceutical preparations in the preparation of medicines for the prevention and/or treatment of related diseases with pathological features of increased ATX expression; preferably,
  • the associated diseases with pathological features of increased ATX expression include: cancer, fibrotic diseases, metabolic diseases, myelodysplastic syndromes, respiratory diseases, cardiovascular diseases, autoimmune diseases, inflammation, dermatological diseases, neurological diseases. Systemic disease or pain; more preferably, the associated disease with pathological features of increased ATX expression is pulmonary fibrosis, renal fibrosis or liver fibrosis.
  • the present invention also provides a method for preventing and/or treating a related disease with pathological characteristics of increased ATX expression, comprising the steps of: adding a therapeutically effective amount of the above-mentioned formula I', formula I, formula I-1, The compound of formula II, formula II-1, formula III or formula III-1 structure or a pharmaceutically acceptable salt, ester, isomer, solvate, prodrug or isotopic label, or the above-mentioned pharmaceutical composition or drug The formulation is administered to a patient in need thereof.
  • the compound of the present invention Compared with the existing ATX inhibitor GLPG-1690, the compound of the present invention has higher inhibitory activity, and meanwhile has excellent pharmacodynamics, in vitro/in vivo pharmacokinetic properties and safety, and has high clinical application prospect.
  • C 1-6 alkyl alone or in combination denotes a saturated straight or branched alkyl group containing 1-6, especially 1-4 carbon atoms, including methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-2- Butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, n-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2 -pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2-methyl-3-pentyl, 2,3-dimethyl-2-butyl, 3,3 ,-dimethyl-2-butyl, etc.
  • "C 1-6 alkyl” is any one of methyl, ethyl, propyl
  • C 1-3 alkylene represent a saturated alkylene straight chain or branched, containing from 1 to 3 carbon atoms, and includes methylene (-CH 2 -), ethylene ( -CH 2 CH 2 -), n-propylidene (-CH 2 CH 2 CH 2 -), isopropylidene (-(CH 3 ) 2 C-) and the like.
  • C 3-7 cycloalkyl alone or in combination denotes a saturated cycloalkyl group having 3-7, especially 3-6 carbon atoms, including cyclopropyl, cyclobutyl, cyclopentyl, cyclo Hexyl, cycloheptyl, etc.
  • C 3 - 7 cycloalkyl are cyclopropyl, cyclopentyl, cyclohexyl and the like.
  • C 1-6 alkoxy alone or in combination refers to the group C 1 - 6 alkyl group -O-, wherein "C 1-6 alkyl” as defined above.
  • halogen alone or in combination denotes fluorine, chlorine, bromine or iodine, in particular fluorine, chlorine or bromine.
  • amino alone or in combination, signifies a primary amino group (-NH 2), secondary amino group (-NH-) or tertiary amino group
  • C 1-6 alkylamino also called “C 1-6 alkylamino” alone or in combination means “amino” as defined above, wherein the hydrogen atom on the amino group is replaced by at least one C 1-6 alkyl group substituted, wherein “C 1-6 alkyl” is as defined above.
  • C 1-6 alkylamino includes methylamino, ethylamino, propylamino, isopropylamino, n-butylamino, isobutylamino, 2-butylamino, tert-butylamino , n-pentylamino, 2-pentylamino, 3-pentylamino, 2-methyl-2-butylamino, 3-methyl-2-butylamino, 3-methyl-1-butylamino , 2-methyl-1-butylamino, n-hexylamino, 2-hexylamino, 3-hexylamino, 2-methyl-2-pentylamino, 3-methyl-2-pentylamino, 4- Methyl-2-pentylamino, 3-methyl-3-pentylamino, 2-methyl-3-pentylamino, 2,3-dimethyl-2-butylamino, 3,3-di Methyl-2-buty
  • C 1-6 alkylamino is methylamino, ethylamino, isopropylamino, tert-butylamino and the like.
  • the C 1-6 alkylamino group may be further optionally substituted by a substituent group common in the art.
  • heterocycloalkyl also known as “heterocyclyl” refers to a saturated or partially unsaturated (containing 1 or 2 double bond) non-aromatic cyclic composed of carbon atoms and heteroatoms such as nitrogen, oxygen or sulfur
  • the number of carbon atoms in the heterocycloalkyl group is 2-11, and the number of heteroatoms is preferably 1, 2, 3 or 4,
  • a nitrogen, carbon or sulfur atom in a heterocycloalkyl group can be optionally oxidized.
  • the hydrogen atoms on “heterocycloalkyl” are independently optionally substituted with one or more substituents described herein.
  • heterocycloalkyl can be linked to the parent molecule through any ring atom in the ring.
  • the terms “3-6 membered heterocycloalkyl” and “3-7 membered heterocycloalkyl” refer to saturated or partially non-isolated groups containing 3-6 and 3-7 carbon atoms and a heteroatom or heteroatom group, respectively.
  • aryl refers to any stable 6-10 membered monocyclic or bicyclic aromatic group, including phenyl, naphthyl, tetrahydronaphthyl, 2,3-indenyl, or biphenyl, and the like.
  • the hydrogen atoms on the "aryl group” are independently optionally substituted with one or more substituents described herein.
  • heteroaryl refers to an aromatic ring group formed by replacing a carbon atom on the ring with at least one heteroatom selected from sulfur, oxygen or nitrogen, and the aromatic ring group may be a 5-7 membered monocyclic or 7-12 bicyclic group.
  • the number of heteroatoms in the heteroaryl group is preferably 1, 2, 3 or 4, such as thienyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyridin-2(1H)-one, Pyridin-4(1H)-one, pyrrolyl, pyrazolyl, thiazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, imidazolyl, tetrazolyl, isothiazole base, oxazolyl, isoxazolyl, thiadiazolyl, oxadiazolyl, naphthyl, benzothienyl, indolyl, benzimidazolyl, benzothiazolyl, benzofuranyl, quinoline base, isoquinolinyl, quinazolinyl, etc.
  • the hydrogen atoms on a "heteroaryl group" are independently optionally optionally
  • C6-10 aryl refers to an aryl group having 6-10 carbon atoms, wherein “aryl” is as defined above.
  • heteroaryl refers to a heteroaryl group having 5-10 atoms, wherein “heteroaryl” is as defined above.
  • cyano alone or in combination refers to the group -CN.
  • hydroxy alone or in combination refers to the group -OH.
  • stereoisomers includes all isomeric forms, including enantiomers, diastereomers and geometric isomers (cis-trans isomers). Therefore, individual stereochemical isomers of the compounds designed in the present invention or mixtures of enantiomers, diastereomers, or geometric isomers (or cis-trans isomers) belong to the present invention range.
  • pharmaceutically acceptable salts means that the compounds of the present invention exist in the form of their pharmaceutically acceptable salts, including acid addition salts and base addition salts.
  • pharmaceutically acceptable non-toxic acid addition salts refer to salts formed by the compounds of the present invention with organic or inorganic acids, including but not limited to hydrochloric acid, sulfuric acid, hydrobromic acid, hydroiodide Acid, phosphoric acid, nitric acid, perchloric acid, acetic acid, oxalic acid, maleic acid, fumaric acid, tartaric acid, benzenesulfonic acid, methanesulfonic acid, salicylic acid, succinic acid, citric acid, lactic acid, propionic acid, benzoic acid, p-toluenesulfonic acid, malic acid, etc.
  • Non-toxic base addition salts refer to salts formed by the compounds of the present invention with organic or inorganic bases, including but not limited to alkali metal salts such as lithium, sodium or potassium salts; alkaline earth metal salts such as calcium Or magnesium salts; organic base salts, such as ammonium salts or N + (C 1-6 alkyl) 4 salts formed with organic bases containing N groups.
  • alkali metal salts such as lithium, sodium or potassium salts
  • alkaline earth metal salts such as calcium Or magnesium salts
  • organic base salts such as ammonium salts or N + (C 1-6 alkyl) 4 salts formed with organic bases containing N groups.
  • solvate refers to an association of one or more solvent molecules with a compound of the present invention.
  • Solvates forming solvents include, but are not limited to, water, methanol, ethanol, isopropanol, ethyl acetate, tetrahydrofuran, N,N-dimethylformamide, dimethylsulfoxide, and the like.
  • hydrate refers to the association of water with the compounds of the present invention.
  • prodrug refers to a chemical derivative of a compound of the present invention, which is converted into a compound represented by general formula I by undergoing a chemical reaction in vivo.
  • isotopic derivatives refers to isotopic derivatives obtained by substituting 1-6 deuterium atoms for hydrogen atoms in general formula I, and isotopic derivatives obtained by replacing carbon atoms in general formula I with 1-3 carbon 14 atoms thing.
  • n is any integer from 0 to 5;
  • each R 1 is independently selected from hydrogen, deuterium, cyano, halogen, amino, hydroxy, -COOH, -CHO, -NO 2 , C 1-6 alkylamino, C 1-6 alkoxy and C 1-6 alkyl, C 3-7 cycloalkyl, unsubstituted or substituted with substituents selected from cyano, amino, hydroxy, halogen, C 1-6 alkyl and C 1-6 alkoxy and 3-7 membered heterocycloalkyl;
  • R 3 is selected from hydrogen, deuterium, C 1-6 alkyl, C 3-7 cycloalkyl and selected from cyano, amino, hydroxyl, halogen, C 1-6 alkyl and C 1-6 alkoxy C 1-6 alkyl, C 3-7 cycloalkyl and 3-7 membered heterocycloalkyl substituted by substituents;
  • Ring B is any of the following structures:
  • R 4 is selected from hydrogen, deuterium, halogen, cyano, hydroxyl, amino, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkylamino, C 3-7 cycloalkyl and 3- 7-membered heterocycloalkyl;
  • X 1 , X 2 , X 3 and X 4 are each independently N or CR 5 ;
  • R 5 is selected from hydrogen, deuterium, halo, C 1-6 alkyl and are selected from cyano, amino, hydroxy, halo, C 1-6 alkyl and C 1-6 alkoxy substituents of C 1-6 alkyl, C 3-7 cycloalkyl and 3-7 membered heterocycloalkyl;
  • Ring A is selected from substituted or unsubstituted C 6-10 aryl and 5-10 membered heteroaryl, the substitution is 1 to 3 selected from hydroxy, amino, halogen, cyano, C 1-6 alkyl , C 1-6 haloalkyl, C 3-7 cycloalkyl, 3-7 membered heterocycloalkyl, C 1-6 alkylamino, C 1-6 alkoxy substituent;
  • R 6 is -L 1 -L 2 -W 1 ;
  • n is any integer from 0 to 2, preferably n is 1; if present, each R 1 is independently selected from hydrogen, cyano and halogen, preferably halogen, more Fluorine is preferred.
  • R 2 is cyano
  • R 3 is C 1-6 alkyl, preferably methyl or ethyl.
  • R 4 is selected from hydrogen, deuterium, methyl, ethyl, isopropyl and cyclopropyl, preferably ethyl.
  • X 1 , X 2 , X 3 and X 4 are each independently N or CR 5 , and R 5 is selected from hydrogen, deuterium, halogen, methyl and halomethyl, preferably hydrogen.
  • Ring A is selected from phenylene, pyridylene, pyrazinylene, pyridazinylene, pyrimidinylene, pyrazolylide, imidazolyl, oxazolylide , isoxazolylide, thiazolylidene, thiadiazolylidene and oxadiazolylide.
  • Ring A is selected from any of the following structures:
  • Ring A is substituted with 1 to 3, preferably 1, substituents each independently selected from the group consisting of hydroxy, amino, halogen, cyano and C1-6alkane groups, preferably halogen and C 1-6 alkyl, more preferably fluorine and methyl.
  • W is an unsubstituted or substituted 1 or 2 substituents selected from hydroxy, amino, hydroxymethyl, acetoxymethyl, and aminomethyl substituents substituted following groups: Methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, aziridinyl, azetidinyl, tetrahydropyrrolyl, oxopyrrolidinyl, piperidine base, piperazinyl, morpholinyl, thiomorpholinyl, oxetanyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, 1,1-dioxothiomorpholinyl, 2- Azaspiro[3.3]heptyl, 1,1-dioxyisothiazolidinyl or
  • n 1;
  • R 1 is fluorine
  • R 2 is cyano
  • R 3 is methyl or ethyl
  • Ring B is any of the following structures:
  • R 4 is ethyl
  • X 1 , X 2 , X 3 and X 4 are each independently N or CH;
  • Ring A is selected from any of the following structures:
  • Ring A is unsubstituted or substituted with 1 fluorine or methyl
  • R 6 is -L 1 -L 2 -W 1 ;
  • W 1 is any of the following groups:
  • the above-mentioned compound of formula I' is a compound having the structure of formula I,
  • n, R 1 , R 2 , R 3 , R 4 , R 6 , X 1 , X 2 , X 3 and Ring A are as defined in formula I′.
  • the above-mentioned compound of formula I' is a compound having the structure of formula I-1,
  • n, R 1 , R 2 , R 3 , R 4 , R 6 and ring A are as defined in formula I'.
  • the above-mentioned compound of formula I' is a compound having the structure of formula II,
  • n, R 1 , R 2 , R 3 , R 4 , R 6 , X 1 , X 2 , X 3 and Ring A are as defined in formula I′.
  • the above-mentioned compound of formula I' is a compound having the structure of formula II-1,
  • n, R 1 , R 2 , R 3 , R 4 , R 6 and ring A are as defined in formula I'.
  • the above-mentioned compound of formula I' is a compound having the structure of formula III,
  • n, R 1 , R 2 , R 3 , R 4 , R 6 , X 1 , X 2 , X 3 and Ring A are as defined in formula I′.
  • the above-mentioned compound of formula I' is a compound having the structure of formula III-1,
  • n, R 1 , R 2 , R 3 , R 4 , R 6 and ring A are as defined in formula I'.
  • the present invention also provides a typical synthesis method of the above-mentioned compound of formula I, to further describe the technical scheme of the present invention, and the synthesis method comprises the following steps:
  • Step 1 Compound 1 reacts with the corresponding aldehyde and 1,1,3,3-tetramethylbutylisonitrile under the catalysis of magnesium chloride to generate intermediate 2;
  • Step 2 The intermediate 2 is heated and deprotected in formic acid to obtain the intermediate 3;
  • Step 3 Optionally, intermediate 3 and corresponding halogenated hydrocarbon are subjected to nucleophilic substitution reaction to obtain intermediate 4;
  • Step 4 Intermediate 4 is hydrolyzed to obtain Intermediate 5;
  • Step 5 Intermediate 5 and intermediate 6 undergo nucleophilic substitution reaction under alkaline conditions to obtain intermediate 7;
  • intermediate 6 is prepared from compound 11 through a two-step reaction: first, intermediate 12 is obtained from compound 11 and thiourea through ring closure; intermediate 12 is obtained under the action of tert-butyl nitrite and copper chloride to obtain intermediate 6 ;
  • Step 6 Intermediate 7 and Intermediate 10 are reacted by Suzuki coupling to obtain Intermediate 8;
  • the intermediate 8 can also be produced by the intermediate 7 to generate the boronic acid reagent 13 first, and then the intermediate 13 and the intermediate 14 (X can be Cl, Br, I) are prepared by the Suzuki reaction;
  • Step 7 Intermediate 8 is obtained by hydrolysis to obtain Intermediate 9;
  • Step 8 The intermediate 9 is subjected to condensation reaction with amine to obtain the compound of formula I, or the product with tert-butoxycarbonyl protection of formula I, which is further deprotected under the condition of trifluoroacetic acid to obtain the compound of formula I.
  • the synthetic route is as follows:
  • R 1 -R 6 , X 1 -X 3 , L 1 , n, and A have the same definitions as above, and R 9 is methyl or ethyl.
  • the present invention also provides a typical synthetic method of the above-mentioned compound of formula II (wherein X 1 is N) to further describe the technical scheme of the present invention, and the synthetic method comprises the following steps:
  • Step 1 compound 1 reacts with hydrogen peroxide in trifluoroacetic acid solution to generate intermediate 2;
  • Step 2 Intermediate 2 is reacted with trimethylsilyl cyanide under alkaline conditions to obtain Intermediate 3;
  • Step 3 Optionally, intermediate 3 is closed with the corresponding substituted hydrazine under basic conditions to obtain intermediate 4;
  • Step 4 Intermediate 4 and intermediate 12 undergo nucleophilic substitution reaction under alkaline conditions to obtain intermediate 5;
  • intermediate 12 is prepared from compound 10 through a two-step reaction: first, intermediate 11 is obtained from compound 10 and thiourea through ring closure; intermediate 11 is obtained under the action of tert-butyl nitrite and copper chloride to obtain intermediate 12 ;
  • Step 5 Intermediate 5 is subjected to nucleophilic substitution reaction with the corresponding halogenated hydrocarbon to obtain intermediate 6;
  • Step 6 intermediate 6 and intermediate 9 are reacted by Suzuki coupling to obtain the compound of formula II, or the product with tert-butoxycarbonyl protection of formula II, which is further deprotected under the condition of trifluoroacetic acid to obtain the compound of formula II;
  • intermediate 9 is prepared from compound 7 through a two-step reaction: firstly, intermediate 8 is obtained by nucleophilic substitution reaction between compound 7 and the corresponding amine; intermediate 8 is generated by boronic acid reagent under the catalyst to obtain intermediate 9.
  • the synthetic route is as follows:
  • R 1 -R 6 , X 2 -X 3 , n, A are the same as above.
  • the present invention also provides a typical synthesis method of the above-mentioned compound of formula III (wherein X 1 is N) to further describe the technical scheme of the present invention, and the synthesis method comprises the following steps:
  • Step 1 Compound 1 and compound 2 are ring-closed under basic conditions to generate intermediate 3;
  • Step 2 Intermediate 3 undergoes a nitration reaction to obtain Intermediate 4;
  • Step 3 Intermediate 4 undergoes a chlorination reaction under phosphorus oxychloride conditions to obtain intermediate 5;
  • Step 4 Intermediate 5 undergoes a reduction reaction to obtain intermediate 6;
  • Step 5 Intermediate 6 and intermediate 14 undergo nucleophilic substitution reaction under alkaline conditions to obtain intermediate 7;
  • intermediate 14 is prepared from compound 12 through a two-step reaction: intermediate 13 is obtained from compound 12 and thiourea through ring closure; intermediate 13 is obtained under the action of tert-butyl nitrite and copper chloride to obtain intermediate 14 ;
  • Step 6 Intermediate 7 is subjected to nucleophilic substitution reaction with the corresponding halogenated hydrocarbon to obtain intermediate 8;
  • Step 7 intermediate 8 and intermediate 11 are reacted by Suzuki coupling to obtain the compound of formula III, or the product with tert-butoxycarbonyl protection of formula III, which is further deprotected under the condition of trifluoroacetic acid to obtain the compound of formula I;
  • intermediate 11 is prepared from compound 9 through a two-step reaction: first, intermediate 10 is obtained by nucleophilic substitution reaction between compound 9 and the corresponding amine; intermediate 10 is generated by boronic acid reagent under catalyst to obtain intermediate 11.
  • the synthetic route is as follows:
  • R 1 -R 6 , X 2 -X 3 , n, A are the same as above.
  • pharmaceutical composition refers to a composition that can be used as a medicament, comprising a pharmaceutically active ingredient (API) and optionally one or more pharmaceutically acceptable carriers.
  • pharmaceutically acceptable carrier refers to pharmaceutical excipients that are compatible with active pharmaceutical ingredients and are harmless to subjects, including (but not limited to) diluents (or fillers), binders, disintegrants Agents, lubricants, wetting agents, thickeners, glidants, flavoring agents, olfactory agents, preservatives, antioxidants, pH adjusters, solvents, cosolvents, surfactants, etc.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the above-mentioned compound having the structure of formula I', formula I, formula I-1, formula II, formula II-1, formula III or formula III-1 or pharmaceutically acceptable compounds thereof of salts, esters, isomers, solvates, prodrugs or isotopic labels.
  • the above-mentioned pharmaceutical composition further comprises a pharmaceutically acceptable carrier.
  • the present invention also provides a pharmaceutical preparation, which comprises the above-mentioned compound having the structure of formula I', formula I, formula I-1, formula II, formula II-1, formula III or formula III-1 or its pharmaceutically acceptable
  • a pharmaceutical preparation which comprises the above-mentioned compound having the structure of formula I', formula I, formula I-1, formula II, formula II-1, formula III or formula III-1 or its pharmaceutically acceptable
  • the salts, esters, isomers, solvates, prodrugs or isotopic labels or the above-mentioned pharmaceutical compositions, the preparations are tablets, capsules, injections, granules, powders, suppositories, pills, creams, pastes Any of the formulations, gels, powders, oral solutions, inhalants, suspensions, dry suspensions, patches, and lotions.
  • the above-mentioned compound having the structure of formula I', formula I, formula I-1, formula II, formula II-1, formula III or formula III-1 or its pharmaceutically acceptable salts, esters, isomers, solvents Compounds, prodrugs or isotopic labels, or the above-mentioned pharmaceutical compositions or pharmaceutical preparations can be used for the prevention and treatment of related diseases with pathological features of increased ATX expression; preferably, the pathological features with increased ATX expression Related diseases include: cancer, fibrotic disease, metabolic disease, myelodysplastic syndrome, respiratory disease, cardiovascular disease, autoimmune disease, inflammation, dermatological disease, neurological disease or pain; more preferably, the The related diseases described as pathological features of increased ATX expression are pulmonary fibrosis, renal fibrosis or liver fibrosis.
  • the present invention also provides the above-mentioned compounds having the structures of formula I', formula I, formula I-1, formula II, formula II-1, formula III or formula III-1 or their pharmaceutically acceptable salts, esters, Isomers, solvates, prodrugs or isotopic labels, and the use of the above-mentioned pharmaceutical compositions or pharmaceutical preparations in the preparation of medicaments for the prevention and/or treatment of related diseases with pathological features of increased ATX expression, preferably
  • the related diseases with pathological features of increased ATX expression include: cancer, fibrotic diseases, metabolic diseases, myelodysplastic syndromes, respiratory diseases, cardiovascular diseases, autoimmune diseases, inflammation, dermatological diseases , neurological disease or pain, more preferably, the related disease with pathological features of increased ATX expression is pulmonary fibrosis, renal fibrosis or liver fibrosis.
  • the present invention provides a method for preventing and/or treating a related disease with pathological features of increased ATX expression, comprising the steps of: adding an effective amount of the above-mentioned formula I', formula I, formula I-1, formula II , the compound of formula II-1, formula III or formula III-1 structure or a pharmaceutically acceptable salt, ester, isomer, solvate, prodrug or isotopic label, or the above-mentioned pharmaceutical composition or pharmaceutical preparation administration for patients in need.
  • terapéuticaally effective amount refers to a dose of a pharmaceutically active ingredient capable of eliciting a biological or medical response in a cell, tissue, organ or organism (eg, a patient).
  • administration refers to the application of a pharmaceutically active ingredient (such as a compound of the present invention) or a pharmaceutical composition comprising a pharmaceutically active ingredient (eg, a pharmaceutical composition of the present invention) to a patient or a site of its cells, tissues, organs, biological fluids, etc. , the process of contacting a patient or its cells, tissues, organs, biological fluids, etc. with active pharmaceutical ingredients or pharmaceutical compositions.
  • a pharmaceutically active ingredient such as a compound of the present invention
  • a pharmaceutical composition comprising a pharmaceutically active ingredient (eg, a pharmaceutical composition of the present invention) to a patient or a site of its cells, tissues, organs, biological fluids, etc.
  • Common modes of administration include, but are not limited to, oral, subcutaneous, intramuscular, subperitoneal, ocular, nasal, sublingual, rectal, vaginal, and the like.
  • in need of it refers to a physician or other caregiver's judgment that a patient needs or will benefit from a prophylactic and/or therapeutic process based on the physician's or other caregiver's various areas of expertise. a factor.
  • patient refers to a human or non-human animal (eg, a mammal).
  • the reactions in the examples are generally carried out under nitrogen protection.
  • MS Mass spectrometry
  • MS uses an ESI source and only indicates the molecular weight M of the parent molecule, usually reporting [M+H] + .
  • Injection volume was determined by sample concentration; flow rate: 0.8 mL/min; HPLC peaks were read by recording UV-Vis wavelengths at 220 nm and 254 nm.
  • the mobile phases were 0.01% formic acid in ultrapure water (mobile phase A) and 0.01% formic acid in acetonitrile (mobile phase B).
  • the gradient elution conditions are shown in Tables 1 and 2 below:
  • NMR spectra were obtained by using a Varian 400MHz nuclear magnetic resonance spectrometer, often with CDCl 3 and DMSO-d 6 as solvents, and chemical shifts were reported in ppm.
  • the various peaks are described as follows: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), dd (doublet). Coupling constants are expressed in Hz.
  • Step 1a Preparation of 2-amino-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 1a To a solution of 3-(4-fluorophenyl)-3-oxopropionitrile (6.0 g, 36.78 mmol) in ethanol (72 mL) was added pyridine (2.97 mL, 36.78 mmol) at room temperature. The mixture was stirred at 70°C for 15 minutes and then cooled to room temperature. Thiourea (5.61 g, 73.56 mmol) and iodine (9.33 g, 6.78 mmol) in EtOH (36 mL) were then slowly added dropwise. After stirring for one hour at room temperature, was added a 1M Na 2 S 2 O 3 (36mL ) quench the reaction. After filtration, the filter cake was washed with water and dried to give 2-amino-4-(4-fluorophenyl)thiazole-5-carbonitrile (4.2 g) as a white solid.
  • Step 1b Preparation of 2-chloro-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 1b At room temperature, to a solution of copper chloride (3.09 g, 23.0 mmol) in acetonitrile (42 mL) was added tert-butyl nitrite (3.57 g, 34.6 mmol). The mixture was stirred at room temperature for 30 minutes, then 2-amino-4-(4-fluorophenyl)thiazole-5-carbonitrile (4.2 g, 19.15 mmol) was added and stirred at room temperature for 1 hour. The reaction was quenched by adding 1M HCl (63 mL) and extracted with ethyl acetate (100 mL ⁇ 2).
  • Step 1c Preparation of 6-bromo-2-ethyl-N-(2,4,4-trimethylpent-2-yl)imidazo[1,2-a]pyridin-3-amine
  • step 1c 5-bromopyridin-2-amine (4.0 g, 23.1 mmol), propionaldehyde (4.4 mL, 25.4 mmol), 1,1,3,3-tetramethylbutyl isocyanide (4.0 mL, 57.8 mmol) and magnesium chloride (220 mg, 2.31 mmol) were dissolved in n-butanol (40 mL), heated to 130°C and stirred for 3 hours. After cooling, the reaction solution was concentrated and diluted with water (30 mL).
  • Step 1d Preparation of N-(6-bromo-2-ethylimidazo[1,2-a]pyridin-3-yl)carboxamide
  • step 1d 6-bromo-2-ethyl-N-(2,4,4-trimethylpentan-2-yl)imidazo[1,2-a]pyridin-3-amine ( 8.1 g, 23.0 mmol) was dissolved in formic acid (30 mL) and refluxed for 2 hours. After the reaction solution was cooled, it was rotated to dryness, and methyl tert-butyl ether (30 mL) was added.
  • step 1e at zero degrees, under nitrogen atmosphere, to N-(6-bromo-2-ethylimidazo[1,2-a]pyridin-3-yl)carboxamide (6g, 22.4mmol) in DMF solution (50 mL) 60% sodium hydride (1.3 g, 33.6 mmol) was added portionwise. After the reaction solution was stirred at room temperature for 1 hour, iodomethane (4.8 g, 33.6 mmol) was added.
  • Step 1f Preparation of 6-bromo-2-ethyl-N-methylimidazo[1,2-a]pyridin-3-amine
  • step 1f To a solution of N-(6-bromo-2-ethylimidazo[1,2-a]pyridin-3-yl)-N-methylformamide (4.5 g, 16.0 mmol) in methanol (25 mL) was added 10M sodium hydroxide solution (24 mL, 240 mmol).
  • Step 1g Preparation of 2-((6-Bromo-2-ethylimidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5 - Formonitrile
  • step 1g under zero degree, under nitrogen atmosphere, add DMF solution of 6-bromo-2-ethyl-N-methylimidazo[1,2-a]pyridin-3-amine (2.5g, 9.84mmol) (30 mL) solution was added 60% sodium hydride (0.79 g, 19.67 mmol). The mixture was stirred at room temperature for 30 minutes, then a solution of 2-chloro-4-(4-fluorophenyl)thiazole-5-carbonitrile (4.8 g, 20.11 mmol) in DMF (10 mL) was added slowly.
  • Step 1h Preparation of 2-(4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]Pyridin-6-yl)phenyl)acetate methyl ester
  • step 1h under nitrogen atmosphere, add 2-((6-bromo-2-ethylimidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4- Fluorophenyl)thiazole-5-carbonitrile (100 mg, 0.219 mmol) and 2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaboran-2-yl) )phenyl)methyl acetate (79 mg, 0.285 mmol) in DMF (5 mL) was added [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (18 mg, 0.022 mmol) and phosphoric acid Potassium (93 mg, 0.438 mmol).
  • Step 1i Preparation of 2-(4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]pyridin-6-yl)phenyl)acetic acid
  • step 1i To 2-(4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo A solution of methyl [1,2-a]pyridin-6-yl)phenyl)acetate (68 mg, 0.129 mmol) in tetrahydrofuran (3 mL) and water (1 mL) was added sodium hydroxide (16 mg, 0.387 mmol). The mixture was stirred at room temperature for 4 hours, adjusted to pH 3-4 with 1M hydrochloric acid solution, and extracted with ethyl acetate (10 mL ⁇ 3).
  • Step 1j Preparation of 2-((2-ethyl-6-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)phenyl)imidazo[1 ,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 1j To 2-(4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]Pyridin-6-yl)phenyl)acetic acid (55 mg, 0.11 mmol) and azetidin-3-ol (24 mg, 0.32 mmol) in DMF (2 mL) were added DIEA (42 mg, 0.324 mmol) and HATU (62 mg, 0.162 mmol). The mixture was stirred at room temperature for 2 hours, diluted with water (10 mL), and extracted with ethyl acetate (10 mL x 3).
  • Step 2a Preparation of ethyl 2-(5-(4,4,5,5-tetramethyl-1,3,2-dioxaboran-2-yl)pyridin-2-yl)acetate
  • step 2a under nitrogen protection, to 2-(5-bromopyridin-2-yl) ethyl acetate (200mg, 0.82mmol), biboronic acid pinacol ester (229mg, 0.90mmol), potassium acetate (120mg , 1.23 mmol) in 1,4-dioxane (2 mL) was added [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (58 mg, 0.08 mmol). The mixture was heated to 85°C and stirred for 2 hours, diluted with water (15 mL), and extracted with ethyl acetate (20 mL ⁇ 2).
  • Step 2b Preparation of 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]pyridin-6-yl)pyridin-2-yl)ethyl acetate
  • step 2b 2-((6-bromo-2-ethylimidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl) Thiazole-5-carbonitrile (100 mg, 0.22 mmol) and 2-(5-(4,4,5,5-tetramethyl-1,3,2-dioxaboran-2-yl)pyridine-2 -yl)ethyl acetate (55 mg, 0.26 mmol) was prepared according to the method of Example 1, step 1h to obtain a brown solid 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazole- 2-yl)(methyl)amino)-2-ethylimidazo[1,2-a]pyridin-6-yl)pyridin-2-yl)acetate (60 mg).
  • Step 2c Preparation of 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]pyridin-6-yl)pyridin-2-yl)acetic acid
  • step 2c 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazolium [1,2-a]pyridin-6-yl)pyridin-2-yl)ethyl acetate (50 mg, 0.09 mmol) and sodium hydroxide (11 mg, 0.27 mmol) were prepared according to the method of Example 1, step 1i to give a brown solid 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a] Pyridin-6-yl)pyridin-2-yl)acetic acid (45 mg).
  • Step 2d Preparation of 2-((2-ethyl-6-(6-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)pyridin-3-yl)imidazole [1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 2d 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]Pyridin-6-yl)pyridin-2-yl)acetic acid (45 mg, 0.09 mmol) and azetidin-3-ol (20 mg, 0.27 mmol) as in Example 1, Step 1j Prepared as a white solid (2-((2-ethyl-6-(6-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)pyridin-3-yl) Imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (28.9 mg).
  • Step 3a Preparation of 1-(tert-butyl)3-ethyl 2-(5-bromo-3-fluoropyridin-2-yl)malonate
  • step 3a 60% sodium hydride (893 mg, 22.3 mmol) was added portionwise to a solution of tert-butyl ethyl malonate (3.5 g, 18.6 mmol) in dry DMF (30 mL) under a nitrogen atmosphere at zero degrees. After the mixture was stirred at zero degrees for half an hour, 5-bromo-2,3-difluoropyridine (3 g, 15.5 mmol) was added, and the reaction was stirred at 80° C. for 3 hours.
  • Step 3b Preparation of ethyl 2-(5-bromo-3-fluoropyridin-2-yl)acetate
  • step 3b at zero degrees, to 1-(tert-butyl) 3-ethyl 2-(5-bromo-3-fluoropyridin-2-yl) malonate (3 g, 8.30 mmol) in dichloromethane
  • methane solution 5 mL
  • trifluoroacetic acid 5 mL
  • the mixture was further stirred at room temperature for 2 hours, concentrated, diluted with water (15 mL), and extracted with ethyl acetate (30 mL x 3).
  • the combined organic phases were washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain the crude product.
  • the crude product was separated and purified by column chromatography to obtain ethyl 2-(5-bromo-3-fluoropyridin-2-yl)acetate (1.2 g) as a yellow oil.
  • Step 3c Preparation of Ethyl 2-(3-Fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaboran-2-yl)pyridin-2-yl)acetate ester
  • step 3c under nitrogen protection, to ethyl 2-(5-bromo-3-fluoropyridin-2-yl)acetate (100 mg, 0.38 mmol), biboronic acid pinacol ester (115 mg, 0.46 mmol), A solution of potassium acetate (56 mg, 0.57 mmol) in 1,4-dioxane (2 mL) was added [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (29 mg, 0.04 mmol) . The mixture was heated to 85°C and stirred for 3 hours, diluted with water (15 mL), and extracted with ethyl acetate (20 mL ⁇ 2).
  • Step 3d Preparation of 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]pyridin-6-yl)-3-fluoropyridin-2-yl)ethyl acetate
  • step 3d 2-((6-bromo-2-ethylimidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl) Thiazole-5-carbonitrile (100 mg, 0.22 mmol) and 2-(3-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaboran-2-yl) ) ethyl pyridin-2-yl)acetate (100 mg, 0.33 mmol) was prepared according to the method of Example 1, step 1h to obtain 2-(5-(3-((5-cyano-4-(4-fluorobenzene) as a brown solid.
  • Step 3e Preparation of 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]pyridin-6-yl)-3-fluoropyridin-2-yl)acetic acid
  • step 3e 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo Ethyl [1,2-a]pyridin-6-yl)-3-fluoropyridin-2-yl)acetate (50 mg, 0.09 mmol) and sodium hydroxide (11 mg, 0.27 mmol) were as in Example 1, Step 1i 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]Pyridin-6-yl)-3-fluoropyridin-2-yl)acetic acid (45 mg).
  • Step 3f Preparation of 2-((2-ethyl-6-(5-fluoro-6-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)pyridine-3 -yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 3f 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]pyridin-6-yl)-3-fluoropyridin-2-yl)acetic acid (45 mg, 0.08 mmol) and azetidin-3-ol (10 mg, 0.13 mmol) as in Example 1
  • the method of step 1j prepared 2-((2-ethyl-6-(5-fluoro-6-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl) as a white solid )pyridin-3-yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (18 mg).
  • Step 4a Preparation of 1-(tert-butyl)3-ethyl 2-(5-bromopyrazin-2-yl)malonate
  • step 4a 2,5-dibromopyrazine (1 g, 4.20 mmol) and tert-butyl ethyl malonate (870 mg, 4.62 mmol) were prepared according to the method of Example 3, step 3a to obtain a yellow solid 1- (tert-Butyl) 3-ethyl 2-(5-bromopyrazin-2-yl)malonate (1.5 g).
  • Step 4b Preparation of ethyl 2-(5-bromopyrazin-2-yl)acetate
  • step 4b 1-(tert-butyl) 3-ethyl 2-(5-bromopyrazin-2-yl) malonate (1.5g, 4.35mmol) is according to the method of embodiment 3 step 3b Ethyl 2-(5-bromopyrazin-2-yl)acetate (400 mg) was prepared as a yellow oil.
  • Step 4c Preparation of (3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a]pyridine- 6-yl)boronic acid
  • step 4c 2-((6-bromo-2-ethylimidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl) Thiazole-5-carbonitrile (100 mg, 0.22 mmol) and pinacol diboronate (67 mg, 0.26 mmol) were prepared as in Example 3, step 3c to give a brown oil (3-((5-cyano-4- (4-Fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a]pyridin-6-yl)boronic acid (100 mg).
  • Step 4d Preparation of 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]Pyridin-6-yl)pyrazin-2-yl)ethyl acetate
  • step 4d (3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a ]pyridin-6-yl)boronic acid (100mg, 0.22mmol) and ethyl 2-(5-bromopyrazin-2-yl)acetate (87mg, 0.36mmol) were prepared according to the method of Example 1, step 1h to give a brown solid 2 -(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a]pyridine ethyl oxazin-6-yl)pyrimidin-2-yl)acetate (50 mg).
  • Step 4e Preparation of 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]pyridin-6-yl)pyrazin-2-yl)acetic acid
  • step 4e 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazolium [1,2-a]pyrazin-6-yl)pyrimidin-2-yl)ethyl acetate (50 mg, 0.09 mmol) and sodium hydroxide (11 mg, 0.27 mmol) were prepared according to the method of Example 1, step 1i to give a brown color Solid 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a ]pyridin-6-yl)pyrazin-2-yl)acetic acid (40 mg).
  • Step 4f Preparation of 2-((2-ethyl-6-(5-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)pyrazin-2-yl) Imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 4f 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]Pyridin-6-yl)pyrazin-2-yl)acetic acid (40 mg, 0.08 mmol) and azetidin-3-ol (18 mg, 0.23 mmol) according to Example 1, Step 1j method to prepare off-white solid (2-((2-ethyl-6-(5-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)pyrazine-2- yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (7 mg).
  • Step 5a Preparation of 1-(tert-butyl)3-ethyl 2-(6-chloropyridazin-3-yl)malonate
  • step 5a 3,6-dichloropyridazine (1 g, 6.70 mmol) and tert-butyl ethyl malonate (1.3 g, 6.70 mmol) were prepared according to the method of step 3a of Example 3 to prepare yellow liquid 1 -(tert-butyl) 3-ethyl 2-(6-chloropyridazin-3-yl)malonate (1.0 g).
  • Step 5b Preparation of ethyl 2-(6-chloropyridazin-3-yl)acetate
  • step 5b 1-(tert-butyl) 3-ethyl 2-(6-chloropyridazin-3-yl) malonate (1.0 g, 3.33 mmol) was prepared according to the method of step 3b of Example 3 Ethyl 2-(6-chloropyridazin-3-yl)acetate (400 mg) was prepared as a yellow oil.
  • Step 5c Preparation of 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]Pyridin-6-yl)pyridazin-3-yl)ethyl acetate
  • step 5c 2-((6-bromo-2-ethylimidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl) Thiazole-5-carbonitrile (100 mg, 0.22 mmol) and ethyl 2-(6-chloropyridazin-3-yl)acetate (60 mg, 0.30 mmol) were prepared as a brown solid 2-( 6-(3-((5-Cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a]pyridine-6 -yl)pyridazin-3-yl)ethyl acetate (90 mg).
  • Step 5d Preparation of 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]pyridin-6-yl)pyridazin-3-yl)acetic acid
  • step 5d To 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo A solution of ethyl [1,2-a]pyridin-6-yl)pyridazin-3-yl)acetate (90 mg, 0.166 mmol) in tetrahydrofuran (1 mL) and water (0.2 mL) was added lithium hydroxide monohydrate (10 mg, 0.250 mmol).
  • Step 5e Preparation of 2-((2-ethyl-6-(6-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)pyridazin-3-yl) Imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 5e 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazolium
  • a solution of [1,2-a]pyridin-6-yl)pyridazin-3-yl)acetic acid (90 mg, 0.175 mmol) and HATU (80 mg, 0.211 mmol) in DMF was stirred at 0° C. for 0.5 h before adding to it Azetidine-3-ol (18 mg, 0.23 mmol). The mixture was stirred at room temperature for 1 hour, diluted with water (10 mL), and extracted with ethyl acetate (10 mL x 3).
  • Step 6a Preparation of 1-(tert-butyl)3-ethyl 2-(5-bromo-6-methylpyridin-2-yl)malonate
  • step 6a 3-bromo-6-fluoro-2-methylpyridine (0.5 g, 2.63 mmol) and tert-butyl ethyl malonate (1.5 mL, 7.89 mmol) were prepared according to Example 3, step 3a.
  • Step 6b Preparation of ethyl 2-(5-bromo-6-methylpyridin-2-yl)acetate
  • step 6b 1-(tert-butyl) 3-ethyl 2-(5-bromo-6-methylpyridin-2-yl) malonate (0.3g, 1.58mmol) was prepared according to Example 3
  • the method of step 3b prepared ethyl 2-(5-bromo-6-methylpyridin-2-yl)acetate (100 mg) as a yellow oil.
  • Step 6c Preparation of (3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a]pyridine- 6-yl)boronic acid
  • step 6c 2-((6-bromo-2-ethylimidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl) Thiazole-5-carbonitrile (200 mg, 0.44 mmol) and pinacol diboronate (134 mg, 0.52 mmol) were prepared as in Example 3, step 3c to give a brown oil (3-((5-cyano-4- (4-Fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a]pyridin-6-yl)boronic acid (200 mg).
  • Step 6d Preparation of 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]Pyridin-6-yl)-6-methylpyridin-2-yl)ethyl acetate
  • step 6d (3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a ]pyridin-6-yl)boronic acid (90mg, 0.21mmol) and ethyl 2-(5-bromo-6-methylpyridin-2-yl)acetate (66mg, 0.26mmol) were prepared according to the procedure of Example 1, step 1h 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2 was obtained as a brown solid -a] Ethyl pyridin-6-yl)-6-methylpyridin-2-yl)acetate (27 mg).
  • Step 6e Preparation of 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]pyridin-6-yl)-6-methylpyridin-2-yl)acetic acid
  • step 6e 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazolium [1,2-a]pyridin-6-yl)-6-methylpyridin-2-yl)ethyl acetate (27 mg, 0.05 mmol) and sodium hydroxide (8.0 mg, 0.20 mmol) according to Example 1, Step 1i 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[ 1,2-a]pyridin-6-yl)-6-methylpyridin-2-yl)acetic acid (25.6 mg).
  • Step 6f Preparation of 2-((2-ethyl-6-(6-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)-2-methylpyridine- 3-yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 6f 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]Pyridin-6-yl)-6-methylpyridin-2-yl)acetic acid (25.6 mg, 0.05 mmol) and azetidin-3-ol (7 mg, 0.1 mmol) were as performed
  • step 1j prepared off-white solid 2-((2-ethyl-6-(6-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)- 2-Methylpyridin-3-yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (6.2 mg ).
  • Example 7 2-((2-ethyl-6-(5-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)pyridin-2-yl)imidazole [1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (Compound 7)
  • step 7a 2-bromo-5-methylpyridine (0.5g, 2.91mmol) was dissolved in 5mL carbon tetrachloride, then azobisisobutyronitrile (48mg, 0.29mmol) and NBS (0.67 g, 3.78 mmol), heated to 70 °C for 3 hours under nitrogen protection. The reaction solution was concentrated and purified by column chromatography to obtain 2-bromo-5-(bromomethyl)pyridine (0.4 g) as a colorless oil.
  • step 7b 2-bromo-5-(bromomethyl)pyridine (300 mg, 1.2 mmol) was dissolved in 5 mL of acetonitrile, tetrabutylammonium cyanide (320 mg, 1.2 mmol) was added, and the reaction was carried out at room temperature for 2 hours.
  • the reaction solution was concentrated, 20 mL of water was added, extracted with ethyl acetate (50 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated.
  • the concentrated reaction liquid was purified by column chromatography to give 2-(6-bromopyridin-3-yl)acetonitrile (200 mg) as a colorless oil.
  • Step 7c Preparation of methyl 2-(6-bromopyridin-3-yl)acetate
  • step 7c Dissolve 2-(6-bromopyridin-3-yl)acetonitrile (200 mg, 1.02 mmol) in 3 mL of methanol, add thionyl chloride (0.4 mL, 5.10 mmol) under nitrogen protection, and react at room temperature 5 hours. 20 mL of water was added, extracted with ethyl acetate (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The concentrated reaction liquid was purified by column chromatography to give methyl 2-(6-bromopyridin-3-yl)acetate (200 mg) as a colorless oil.
  • Step 7d Preparation of 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]Pyridin-6-yl)pyridin-3-yl)acetate methyl ester
  • step 7d (3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2- a] Pyridin-6-yl)boronic acid (150mg, 0.36mmol) and methyl 2-(6-bromopyridin-3-yl)acetate (98mg, 0.43mmol) were prepared according to the method of Example 1, step 1h to give a brown solid 2 -(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a]pyridine -6-yl)pyridin-3-yl)acetate methyl ester (70m).
  • Step 7e Preparation of 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]pyridin-6-yl)pyridin-3-yl)acetic acid
  • step 7e 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]Pyridin-6-yl)pyridin-3-yl acetate methyl ester (70 mg, 0.13 mmol) and sodium hydroxide (16 mg, 0.40 mmol) were prepared according to the method of Example 1, step 1i to obtain a brown solid 2 -(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a]pyridine -6-yl)pyridin-3-yl)acetic acid (68 mg).
  • Step 7f Preparation of 2-((2-ethyl-6-(5-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)pyridin-2-yl)imidazole [1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 7f 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]pyridin-6-yl)pyridin-3-yl)acetic acid (66 mg, 0.13 mmol) and azetidin-3-ol (14 mg, 0.2 mmol) as in Example 1, Step 1j 2-((2-ethyl-6-(5-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)pyridin-2-yl)imidazole was prepared as off-white solid [1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (6.3 mg).
  • Example 8 2-((2-Ethyl-6-(5-(3-hydroxyazetidine-1-carbonyl)pyrazin-2-yl)imidazo[1,2-a]pyridine- 3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (Compound 8)
  • Step 8a Preparation of methyl 5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2 -a]Pyridin-6-yl)pyrazine-2-carboxylate methyl ester
  • step 8a 3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a ] pyridin-6-yl)boronic acid (210mg, 0.50mmol) and methyl 5-bromopyrazine-2-carboxylate (216mg, 1.0mmol) were prepared according to the method of Example 1, step 1h to obtain a brown solid methyl 5-( 3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a]pyridin-6-yl) Methyl pyrazine-2-carboxylate (143 mg).
  • Step 8b Preparation of 5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a ]pyridin-6-yl)pyrazine-2-carboxylic acid
  • step 8b at room temperature, methyl 5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethyl Methyl imidazo[1,2-a]pyridin-6-yl)pyrazine-2-carboxylate (143 mg, 0.28 mmol) and sodium hydroxide (24 mg, 0.60 mmol) were prepared according to the method of Example 1, step 1i 5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a]pyridine as brown solid -6-yl)pyrazine-2-carboxylic acid (139 mg).
  • Step 8c Preparation of 2-((2-ethyl-6-(5-(3-hydroxyazetidine-1-carbonyl)pyrazin-2-yl)imidazo[1,2-a]pyridine- 3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 8c at room temperature, 5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]Pyridin-6-yl)pyrazine-2-carboxylic acid (46 mg, 0.092 mmol) and azetidin-3-ol (14 mg, 0.2 mmol) according to the procedure of Example 1, Step 1j 2-((2-ethyl-6-(5-(3-hydroxyazetidine-1-carbonyl)pyrazin-2-yl)imidazo[1,2-a]pyridine- 3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (14.2 mg).
  • Step 9a Preparation of ethyl 2-(5-bromopyrazin-2-yl)-2-methylpropanoate
  • step 9a under a nitrogen atmosphere at zero degrees, 60% sodium hydride (69 mg, 1.73 mmol) was added in portions to ethyl 2-(5-bromopyrazin-2-yl)acetate (200 mg, 0.82 mmol). DMF solution (5 mL) was dried. After the mixture was continued to stir at zero for half an hour, iodomethane (244 mg, 1.73 mmol) was added and the reaction was stirred at room temperature for 2 hours.
  • the reaction was quenched by adding water (15 mL), extracted with ethyl acetate (20 mL ⁇ 3), then the combined organic phases were washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain the crude product.
  • the crude product was purified by flash column chromatography to give ethyl 2-(5-bromopyrazin-2-yl)-2-methylpropanoate (120 mg) as a yellow oily liquid.
  • Step 9b Preparation of 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]Pyridin-6-yl)pyrazin-2-yl)-2-methylpropionic acid ethyl ester
  • step 9b (3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2- a] Pyridin-6-yl)boronic acid (100 mg, 0.24 mmol) and 2-(5-bromopyrazin-2-yl)-2-methylpropionic acid ethyl ester (97 mg, 0.36 mmol) according to Example 1 step 1h 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[ 1,2-a]Pyridin-6-yl)pyrazin-2-yl)-2-methylpropionic acid ethyl ester (100 mg).
  • Step 9c Preparation of 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]Pyridin-6-yl)pyrazin-2-yl)-2-methylpropionic acid
  • step 9c at room temperature, 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethyl ethylimidazo[1,2-a]pyridin-6-yl)pyrazin-2-yl)-2-methylpropanoate (100 mg, 0.18 mmol) and lithium hydroxide monohydrate (15 mg, 0.35 mmol)
  • the brown solid 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2 was prepared according to the method of Example 5, step 5d.
  • -Ethylimidazo[1,2-a]pyridin-6-yl)pyrazin-2-yl)-2-methylpropionic acid 110 mg).
  • Step 9d Preparation of 2-((2-ethyl-6-(5-(1-(3-hydroxyazetidin-1-yl)-2-methyl-1-oxopropan-2-yl )pyrazin-2-yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 9d at room temperature, 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethyl imidazo[1,2-a]pyridin-6-yl)pyrazin-2-yl)-2-methylpropionic acid (100 mg, 0.18 mmol) and azetidin-3-ol (27 mg, 0.37 mmol) was prepared according to the method of step 5e in Example 5 to obtain a white solid 2-((2-ethyl-6-(5-(1-(3-hydroxyazetidine-1-yl)-2-methyl) -1-Oxopropan-2-yl)pyrazin-2-yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole -5-carbonitrile (45 mg).
  • Step 10a Preparation of 5-(bromomethyl)-2-chloro-3-fluoropyridine
  • step 10a Dissolve 2-chloro-3-fluoro-5-methylpyridine (1.0 g, 6.90 mmol) in 10 mL of carbon tetrachloride, then add azobisisobutyronitrile (113 mg, 0.69 mmol) And NBS (1.35g, 7.59mmol), under nitrogen protection, heated to 70 °C and reacted for 3 hours.
  • the reaction solution was concentrated and purified by column chromatography to obtain 5-(bromomethyl)-2-chloro-3-fluoropyridine (0.45 g) as a colorless oil.
  • step 10b 5-(bromomethyl)-2-chloro-3-fluoropyridine (450mg, 2.0mmol) was dissolved in 5mL acetonitrile, tetrabutylammonium cyanide (644mg, 2.4mmol) was added, room temperature React for 2 hours.
  • the reaction solution was concentrated, 20 mL of water was added, extracted with ethyl acetate (50 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated.
  • the concentrated reaction liquid was purified by column chromatography to give 2-(6-chloro-5-fluoropyridin-3-yl)acetonitrile (275 mg) as a colorless oil.
  • Step 10c Preparation of methyl 2-(6-chloro-5-fluoropyridin-3-yl)acetate
  • step 10c 2-(6-chloro-5-fluoropyridin-3-yl)acetonitrile (275 mg, 1.62 mmol) was dissolved in 3 mL of methanol, and thionyl chloride (0.6 mL, 8.10 mmol) was added under nitrogen protection. ) for 5 hours at room temperature. 20 mL of water was added, extracted with ethyl acetate (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The concentrated reaction liquid was purified by column chromatography to give methyl 2-(6-chloro-5-fluoropyridin-3-yl)acetate (253 mg) as a colorless oil.
  • Step 10d Preparation of 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, Methyl 2-a]pyridin-6-yl)-5-fluoropyridin-3-ylacetate
  • step 10d (3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a ]pyridin-6-yl)boronic acid (351mg, 0.83mmol) and methyl 2-(6-chloro-5-fluoropyridin-3-yl)acetate (253mg, 1.25mmol) were prepared according to the method of Example 1, step 1h Brown solid 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2- a] Methyl pyridin-6-yl)-5-fluoropyridin-3-ylacetate (110 mg).
  • Step 10e Preparation of 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]pyridin-6-yl)-5-fluoropyridin-3-yl)acetic acid
  • step 10e 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazolium Methyl [1,2-a]pyridin-6-yl)-5-fluoropyridin-3-ylacetate (110 mg, 0.20 mmol) and sodium hydroxide (16 mg, 0.40 mmol) were prepared according to the procedure of Example 1, Step 1i 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2 was obtained as a brown solid -a]Pyridin-6-yl)-5-fluoropyridin-3-yl)acetic acid (106 mg).
  • Step 10f Preparation of 2-((2-ethyl-6-(3-fluoro-5-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)pyridine-2 -yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 10f 2-(6-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]pyridin-6-yl)-5-fluoropyridin-3-yl)acetic acid (106 mg, 0.2 mmol) and azetidin-3-ol (27 mg, 0.37 mmol) as in Example 1
  • the method of step 1j produced 2-((2-ethyl-6-(3-fluoro-5-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl) as an off-white solid )pyridin-2-yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (29.4 mg).
  • Step 11a Preparation of (S)-2-((2-ethyl-6-(5-(2-(3-hydroxypyrrolidin-1-yl)-2-oxoethyl)pyrazin-2-yl )imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 11a 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]Pyridin-6-yl)pyrazin-2-yl)acetic acid (40 mg, 0.08 mmol) and (S)-3-pyrrolidinol (8.14 mg, 0.093 mmol) according to Example 1, Step 1j The method of preparing off-white solid (S)-2-((2-ethyl-6-(5-(2-(3-hydroxypyrrolidin-1-yl)-2-oxoethyl)pyrazine-2 -yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (20.5 mg).
  • Example 12 2-((2-Ethyl-6-(5-(2-(3-(hydroxymethyl)azetidin-1-yl)-2-oxoethyl)pyrazine- 2-yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (Compound 12)
  • Step 12a Preparation of 2-((2-ethyl-6-(5-(2-(3-(hydroxymethyl)azetidin-1-yl)-2-oxoethyl)pyrazine- 2-yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 12a 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]Pyridin-6-yl)pyrazin-2-yl)acetic acid (40 mg, 0.08 mmol) and 3-(hydroxymethyl)azetidine hydrochloride (11.55 mg, 0.093 mmol) The off-white solid 2-((2-ethyl-6-(5-(2-(3-(hydroxymethyl)azetidin-1-yl)-2- oxoethyl)pyrazin-2-yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile ( 12.5 mg).
  • Step 13a Preparation of (R)-2-((2-ethyl-6-(5-(2-(3-hydroxypyrrolidin-1-yl)-2-oxoethyl)pyrazin-2-yl )imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 13a 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]Pyridin-6-yl)pyrazin-2-yl)acetic acid (40 mg, 0.08 mmol) and (R)-pyrrolidin-3-ol (14 mg, 0.16 mmol) following the procedure of Example 1 1j method to prepare off-white solid (R)-2-((2-ethyl-6-(5-(2-(3-hydroxypyrrolidin-1-yl)-2-oxoethyl)pyrazine- 2-yl)imidazo[1,2-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (16 mg).
  • Step 14a Preparation of 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]Pyridin-6-yl)pyrazin-2-yl)-N-((5-oxopyrrolidin-2-yl)methyl)acetamide
  • step 14a 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]Pyridin-6-yl)pyrazin-2-yl)acetic acid (40 mg, 0.08 mmol) and 5-(aminomethyl)pyrrolidin-2-one (18 mg, 0.16 mmol) as per example 1
  • the method of step 1j gave 2-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethyl as an off-white solid Imidazo[1,2-a]pyridin-6-yl)pyrazin-2-yl)-N-((5-oxopyrrolidin-2-yl)methyl)acetamide (20 mg).
  • Step 15a Preparation of 6-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]Pyridin-6-yl)pyrazine-2-carboxamide)-2-azaspiro[3.3]heptane-2-carboxylic acid tert-butyl ester
  • step 15a 5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]Pyridin-6-yl)pyrazine-2-carboxylic acid (40 mg, 0.08 mmol) and tert-butyl 6-amino-2-azaspiro[3.3]heptane-2-carboxylate (34 mg, 0.16 mmol) was prepared according to the method of step 1j of Example 1 to obtain off-white solid 6-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino) - 2-Ethylimidazo[1,2-a]pyridin-6-yl)pyrazine-2-carboxamide)-2-azaspiro[3.3]heptane-2-carboxylic acid tert-butyl ester (32 mg).
  • Step 15b Preparation of 5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a ]pyridin-6-yl)-N-(2-azaspiro[3.3]heptan-6-yl)pyrazine-2-carboxamide
  • step 15b To 6-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo [1,2-a]Pyridin-6-yl)pyrazine-2-carboxamide)-2-azaspiro[3.3]heptane-2-carboxylic acid tert-butyl ester (32 mg, 0.046 mmol) in dichloromethane ( 1 ml) solution was added trifluoroacetic acid (2 ml), stirred at room temperature for 1 hour, quenched with saturated aqueous sodium bicarbonate solution, and washed with dichloromethane (10 mL ⁇ 3).
  • Step 16a Preparation of (5-fluoro-6-(methoxycarbonyl)pyridin-3-yl)boronic acid
  • step 16a 5-bromo-3-fluoropicolinate methyl ester (100mg, 0.429mmol) and biboronic acid pinacol ester (130mg, 0.52mmol) were prepared according to the method of step 2a of Example 2 to obtain a black oily substance (5-Fluoro-6-(methoxycarbonyl)pyridin-3-yl)boronic acid (140 mg).
  • Step 16b Preparation of 5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a ]pyridin-6-yl)-3-fluoropicolinate methyl ester
  • step 16b (5-fluoro-6-(methoxycarbonyl)pyridin-3-yl)boronic acid (85 mg, 0.43 mmol) and 2-((6-bromo-2-ethylimidazo[1, 2-a]Pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (391 mg, 0.86 mmol) was prepared according to the method of Example 1, step 1h to obtain a yellow solid 5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a]pyridine-6 -yl)-3-fluoropicolinate methyl ester (150 mg).
  • Step 16c 5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a] Pyridin-6-yl)-3-fluoropicolinic acid
  • step 16c 5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1, 2-a]Pyridin-6-yl)-3-fluoropicolinate methyl ester (150mg, 0.28mmol) and sodium hydroxide (24mg, 0.60mmol) were prepared according to the method of Example 1 step 1i to obtain a white solid 5-(3 -((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2-a]pyridin-6-yl)- 3-Fluoropicolinic acid (191 mg).
  • Step 16d Preparation of 2-((2-ethyl-6-(5-fluoro-6-(3-hydroxyazetidine-1-carbonyl)pyridin-3-yl)imidazo[1,2-a ]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 16d 5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethylimidazo[1,2 -a]Pyridin-6-yl)-3-fluoropicolinic acid (146 mg, 0.28 mmol) and azetidine-3-ol (27 mg, 0.37 mmol) were prepared according to the method of Example 1, step 1j to obtain a white solid 2 -((2-ethyl-6-(5-fluoro-6-(3-hydroxyazetidine-1-carbonyl)pyridin-3-yl)imidazo[1,2-a]pyridine-3- (methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (65.8 mg).
  • Example 17 2-((5-(2-(1,1-Dioxyisothiazolidine-2-yl)pyrimidin-5-yl)-2-ethyl-2H-pyrazolo[4,3- b] Pyridin-3-yl)(ethyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (Compound 17)
  • Step 17a Preparation of 2-bromo-5-fluoro-1-hydroxypyridine-1-onium
  • step 17a at room temperature, hydrogen peroxide (80 mL) was added to a solution of 2-bromo-5-fluoropyridine (50.0 g, 36.78 mmol) in trifluoroacetic acid (400 mL). The mixture was stirred at 85°C for 10 hours and then cooled to room temperature, quenched by the addition of sodium thiosulfate (120 g), and filtered. The mother liquor was adjusted to pH 7-8 with aqueous sodium hydroxide solution and extracted with dichloromethane (800ml x 3). The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give 2-bromo-5-fluoro-1-hydroxypyridine-1-onium (50 g) as a gray solid.
  • step 1b at room temperature, to 2-bromo-5-fluoro-1-hydroxypyridine-1-onium (50 g, 0.25 mol) in acetonitrile solution (400 mL) was added triethylamine (126 g, 1.25 mol) and triethylamine Methylsilyl cyanide (125 g, 1.25 mol). The mixture was heated to 90°C for 12 hours, then concentrated, and the crude product was purified by flash column chromatography to give 6-bromo-3-fluoro-2-cyanopyridine (22 g) as an off-white solid.
  • Step 17c Preparation of 5-bromo-2-ethyl-2H-pyrazolo[4,3-b]pyridin-3-amine
  • step 17c To a solution of 6-bromo-3-fluoro-2-cyanopyridine (22g, 0.11mol) and hydrazine oxalate (25g, 0.16mol) in dimethyl sulfoxide (110ml) was added Potassium carbonate (53g, 0.38mol), the reaction was heated to 110°C for 2h. The reaction solution was cooled to room temperature, diluted with ethyl acetate (800ml), the organic phase was washed with water (200ml ⁇ 3) and saturated brine (200ml), dried, filtered and concentrated. The crude product was purified by flash column chromatography to give 5-bromo-2-ethyl-2H-pyrazolo[4,3-b]pyridin-3-amine (3.2 g) as a yellow solid.
  • Step 17d Preparation of 2-((5-Bromo-2-ethyl-2H-pyrazolo[4,3-b]pyridin-3-yl)amino)-4-(4-fluorophenyl)thiazole-5 - Formonitrile
  • step 17d at zero degrees, under nitrogen atmosphere, a solution of 5-bromo-2-ethyl-2H-pyrazolo[4,3-b]pyridin-3-amine (3.2 g, 13 mmol) in DMF (30 mL ) was added portionwise 60% sodium hydride (1.1 g, 26.4 mmol). The reaction solution was stirred at 0°C for 15 minutes, and then 2-chloro-4-(4-fluorophenyl)thiazole-5-carbonitrile (3.4 g, 14.5 mmol) was added in portions. The mixture was allowed to react at room temperature for 1 hour before being quenched with water (60 mL) and filtered.
  • Step 17e Preparation of 2-((5-Bromo-2-ethyl-2H-pyrazolo[4,3-b]pyridin-3-yl)(ethyl)amino)-4-(4-fluorophenyl ) thiazole-5-carbonitrile
  • step 17e at zero degrees, under nitrogen atmosphere, 2-((5-bromo-2-ethyl-2H-pyrazolo[4,3-b]pyridin-3-yl)amino)-4-( 4-Fluorophenyl)thiazole-5-carbonitrile (6 g, 13.6 mmol) in DMF (50 mL) was added portionwise 60% sodium hydride (1.4 g, 33.9 mmol). The reaction solution was stirred at room temperature for 0.5 hours, and then iodoethane (4.2 g, 27.1 mmol) was added.
  • step 17f under nitrogen atmosphere, to a solution of 3-chloropropane-1-sulfonamide (1.85 g, 7.87 mmol) and 2,5-dibromopyrimidine (954 mg, 6.05 mmol) in tert-butanol (10 mL) was added Tris(dibenzylideneacetone)dipalladium (277 mg, 0.30 mmol), 2-(di-tert-butylphosphine)biphenyl (180 mg, 0.61 mmol), 4,5-bisdiphenylphosphine-9,9-diphenyl Methylxanthene (350 mg, 0.61 mmol) and cesium carbonate (3.94 g, 12.11 mmol).
  • Step 17g Preparation of (2-(1,1-Dioxyisothiazolidine-2-yl)pyrimidin-5-yl)boronic acid
  • step 17g under nitrogen protection, to 2-(5-bromopyrimidin-2-yl)isothiazolidine 1,1-dioxide (5g, 18.0mmol), biboronic acid pinacol ester (5.0g, 19.8 mmol), potassium acetate (5.3 g, 54.1 mmol) in 1,4-dioxane solution (50 mL) was added [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride ( 661 mg, 0.9 mmol). The mixture was heated to 85°C and stirred for 2 hours, diluted with water (80 mL), and extracted with ethyl acetate (100 mL ⁇ 2).
  • Step 17h Preparation of 2-((5-(2-(1,1-Dioxyisothiazolidin-2-yl)pyrimidin-5-yl)-2-ethyl-2H-pyrazolin[4,3- b]Pyridin-3-yl)(ethyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 17h under nitrogen atmosphere, add 2-((5-bromo-2-ethyl-2H-pyrazolo[4,3-b]pyridin-3-yl)(ethyl)amino)-4 -(4-Fluorophenyl)thiazole-5-carbonitrile (5 g, 10.6 mmol) and (2-(1,1-dioxyisothiazolidine-2-yl)pyrimidin-5-yl)boronic acid (3.1 g, [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (389 mg, 0.53 mmol) was added to a solution of 12.8 mmol) in 1,4-dioxane (50 mL) and water (7 ml).
  • Step 18a Preparation of 2-ethylpyrazolo[1,5-a]pyrimidin-5(4H)-one
  • step 18a 3-ethyl-1H-pyrazol-5-amine (5 g, 45 mmol) and 1,3-dimethylpyrimidine-2,4(1H,3H)-dione ( To a solution of 7.6 g, 54 mmol) in ethanol (50 mL) was added sodium ethoxide (9.2 g, 135 mmol). The mixture was stirred at 90°C for 5 hours, cooled to room temperature, concentrated and adjusted to pH 2-3 by adding dilute hydrochloric acid (2M). Filtration and drying of the filter cake gave 2-ethylpyrazolo[1,5-a]pyrimidin-5(4H)-one (5g) as a white solid.
  • step 18b at zero temperature, to 2-ethylpyrazolo[1,5-a]pyrimidin-5(4H)-one (5g, 0.25mol) in concentrated sulfuric acid solution (20mL) was slowly added dropwise concentrated Nitric acid (2.2ml). The mixture was continued to react at 0 °C for 2 hours, then the reaction solution was slowly added to ice water to quench, filtered and dried to obtain 2-ethyl-3-nitropyrazolo[1,5-a]pyrimidine-5 as a pale yellow solid (4H)-ketone (5 g).
  • step 18c To a solution of 2-ethyl-3-nitropyrazolo[1,5-a]pyrimidin-5(4H)-one (5g, 24mol) in phosphorus oxychloride (30ml) was added N,N-dimethylaniline (4.4g, 36mol), the reaction was heated to 90°C for 2h. The reaction solution was cooled to room temperature, concentrated, quenched with water, and extracted with ethyl acetate (100ml). The organic phase was washed with 1M sodium bicarbonate solution (40ml), washed with saturated brine (40ml), dried and filtered. Concentration gave 5-chloro-2-ethyl-3-nitropyrazo[1,5-a]pyrimidine (4.5 g) as a yellow solid.
  • step 18d to the ethanol solution (40 mL) of 5-chloro-2-ethyl-3-nitropyrazo[1,5-a]pyrimidine (4.5 g, 13 mmol) was added iron powder (1.1 g, 26.4 mmol) and saturated ammonium chloride solution (20 ml). The reaction solution was heated to 45°C, reacted for 2 hours, filtered and concentrated. The residue was extracted with ethyl acetate (50 mL ⁇ 3), the combined organic phases were washed with saturated brine, dried and concentrated to obtain the crude product. The crude product was purified by flash column chromatography to give 5-chloro-2-ethylpyrazolo[1,5-a]pyrimidin-3-amine (3 g) as a yellow solid.
  • Step 18e Preparation of 2-((5-Chloro-2-ethylpyrazolo[1,5-a]pyrimidin-3-yl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 18e at zero degrees, under nitrogen atmosphere, to the THF solution (30 mL) of 5-chloro-2-ethylpyrazolo[1,5-a]pyrimidin-3-amine (3.0 g, 15 mmol) was added in batches 60% Sodium hydride (1.2 g, 30.1 mmol). The reaction solution was stirred at 0°C for 15 minutes, and then 2-chloro-4-(4-fluorophenyl)thiazole-5-carbonitrile (4.3 g, 18.4 mmol) was added in portions.
  • Step 18f Preparation of 2-((5-Chloro-2-ethylpyrazolo[1,5-a]pyrimidin-3-yl)(ethyl)amino)-4-(4-fluorophenyl)thiazole- 5-carbonitrile
  • step 18f at zero degrees, under nitrogen atmosphere, to 2-((5-chloro-2-ethylpyrazolo[1,5-a]pyrimidin-3-yl)amino)-4-(4-fluoro Phenyl)thiazole-5-carbonitrile (600 mg, 1.5 mmol) in DMF (6 mL) was added cesium carbonate (733 mg, 2.2 mmol). The reaction solution was stirred at room temperature for ten minutes, and then iodoethane (305 mg, 1.9 mmol) was added.
  • Step 18g Preparation of 2-((5-(2-(1,1-Dioxyisothiazolidine-2-yl)pyrimidin-5-yl)-2-ethylpyrazolo[1,5-a]pyrimidine -3-yl)(ethyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 18g under nitrogen atmosphere, 2-((5-chloro-2-ethylpyrazolo[1,5-a]pyrimidin-3-yl)(ethyl)amino)-4-(4 -Fluorophenyl)thiazole-5-carbonitrile (122 mg, 0.258 mmol) and (2-(1,1-dioxyisothiazolidine-2-yl)pyrimidin-5-yl)boronic acid (94 mg, 0.388 mmol) according to The method of Example 17, step 17h prepared yellow solid 2-((5-(2-(1,1-dioxyisothiazolidine-2-yl)pyrimidin-5-yl)-2-ethylpyrazolo[ 1,5-a]pyrimidin-3-yl)(ethyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (108 mg).
  • Step 19a Preparation of 2-(5-Bromopyrazin-2-yl)isothiazolidine 1,1-dioxide
  • step 19a under nitrogen atmosphere, 2,5-dibromopyrazine (900mg, 3.82mmol) and 3-chloropropane-1-sulfonamide (400mg, 2.54mmol) were prepared according to the method of step 17f of Example 17 2-(5-Bromopyrazin-2-yl)isothiazolidine 1,1-dioxide (364 mg) was obtained as a brown solid.
  • Step 19b Preparation of 2-((6-(5-(1,1-Dioxyisothiazolidine-2-yl)pyrazin-2-yl)-2-ethylimidazo[1,2-a]pyridine -3-yl)(ethyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 19b under nitrogen atmosphere, 2-(5-bromopyrazin-2-yl)isothiazolidine 1,1-dioxide (170 mg, 0.614 mmol) and (3-((5-cyano -4-(4-Fluorophenyl)thiazol-2-yl)(ethyl)amino)-2-ethylimidazo[1,2-a]pyridin-6-yl)boronic acid (400 mg, 0.921 mmol) according to The method of Example 17, step 17h prepared a white solid 2-((6-(5-(1,1-dioxyisothiazolidine-2-yl)pyrazin-2-yl)-2-ethylimidazo[ 1,2-a]pyridin-3-yl)(ethyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (74.8 mg).
  • Step 20a Preparation of N-(5-bromopyrimidin-2-yl)-N-methylmethanesulfonamide
  • step 20a in nitrogen atmosphere, N-methylmethanesulfonamide (77mg, 0.707mmol) and 2,5-dibromopyrimidine (200mg, 0.848mmol) were prepared according to the method of step 17f of Example 17 to prepare N- (5-Bromopyrimidin-2-yl)-N-methylmethanesulfonamide (114 mg).
  • Step 20b Preparation of N-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(ethyl)amino)-2-ethylimidazo[1, 2-a]Pyridin-6-yl)pyrimidin-2-yl)-N-methylmethanesulfonamide
  • step 20b under nitrogen atmosphere, N-(5-bromopyrimidin-2-yl)-N-methylmethanesulfonamide (70 mg, 0.263 mmol) and (3-((5-cyano-4- (4-Fluorophenyl)thiazol-2-yl)(ethyl)amino)-2-ethylimidazo[1,2-a]pyridin-6-yl)boronic acid (172 mg, 0.395 mmol) as in Example 17
  • the method of step 17h prepared N-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(ethyl)amino)-2-ethylimidazole as a brown solid [1,2-a]pyridin-6-yl)pyrimidin-2-yl)-N-methylmethanesulfonamide (45 mg).
  • Step 21a Preparation of 2-(5-Bromopyrimidin-2-yl)-1,2-thiazinane 1,1-dioxide
  • step 21a at room temperature, under nitrogen atmosphere, 1,2-thiazinane 1,1-dioxide (300mg, 2.22mmol) and 2,5-dibromopyrimidine (792mg, 3.33mmol) were carried out according to Example 17
  • the procedure of step 17f prepared 2-(5-bromopyrimidin-2-yl)-1,2-thiazinane 1,1-dioxide (500 mg) as a brown-like solid.
  • Step 21b Preparation of (2-(1,1-Dioxy-1,2-thiazin-2-yl)pyrimidin-5-yl)boronic acid
  • step 21b under nitrogen atmosphere, to 2-(5-bromopyrimidin-2-yl)-1,2-thiazinane 1,1-dioxide (200 mg, 0.68 mmol) and pinacol biboronate Ester (191 mg, 0.75 mmol) was prepared according to the procedure of Example 17, step 17g to give (2-(1,1-dioxy-1,2-thiazin-2-yl)pyrimidin-5-yl)boronic acid (176mg) .
  • Step 21c Preparation of 2-((5-(2-(1,1-Dioxy-1,2-thiazin-2-yl)pyrimidin-5-yl)-2-ethyl-2H-pyrazolo [4,3-b]pyridin-3-yl)(ethyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 21c under nitrogen atmosphere, (2-(1,1-dioxy-1,2-thiazin-2-yl)pyrimidin-5-yl)boronic acid (175 mg, 0.68 mmol) and 2- ((5-Bromo-2-ethyl-2H-pyrazolo[4,3-b]pyridin-3-yl)(ethyl)amino)-4-(4-fluorophenyl)thiazole-5-methyl Nitrile (320 mg, 0.68 mmol) was prepared according to the method of Example 17, step 17h to give 2-((5-(2-(1,1-dioxy-1,2-thiazin-2-yl)pyrimidine) as an off-white solid -5-yl)-2-ethyl-2H-pyrazolo[4,3-b]pyridin-3-yl)(ethyl)amino)-4-(4-fluorophenyl)thiazole-5-methyl Nitrile (60 mg).
  • step 22a at room temperature, ((2-bromoethoxy)methyl)benzene (5 g, 23.36 mmol) was added to an aqueous solution (170 mL) of sodium sulfite (3.62 g, 28.67 mmol). The mixture was stirred at 100°C for 6 hours and concentrated to give a white solid. Methanol (170 mL) was then added to the white solid, the mixture was heated to 50°C and stirred for 20 minutes, filtered, and the filtrate was concentrated to give 2-(benzyloxy)ethane-1-sulfonic acid (6.8 g) as a white solid.
  • step 22b at room temperature, under nitrogen atmosphere, DMF (163 mg, 2.3 mmol) and thionyl chloride (5.1 mL, 90.81 mmol) were added to 2-(benzyloxy)ethane-1-sulfonic acid (5.0 g, 23.14 mmol) in tetrahydrofuran (20 mL). The mixture was stirred at 65°C for 5 hours. Filtration and concentration of the filtrate gave 2-(benzyloxy)ethane-1-sulfonyl chloride (5.6 g) as a yellow oil.
  • DMF 163 mg, 2.3 mmol
  • thionyl chloride 5.1 mL, 90.81 mmol
  • Step 22c Preparation of 2-(benzyloxy)-N-(5-bromopyrimidin-2-yl)ethane-1-sulfonamide
  • step 22c 5-bromopyridin-2-amine (6.0 g, 34.69 mmol) and triethylamine (6.4 mL, 46.15 mmol) were added to 2-(benzyloxy)ethane-1-sulfonic acid at room temperature
  • the acid chloride (5.4 g, 23.08 mmol) in tetrahydrofuran solution (50 mL) was heated to 80°C and stirred for 10 hours. After cooling, it was diluted with water (50 mL).
  • the aqueous phase was extracted with ethyl acetate (80 mL ⁇ 3), the combined organic phases were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • the crude product was purified by flash column chromatography to give 2-(benzyloxy)-N-(5-bromopyrimidin-2-yl)ethane-1-sulfonamide (560 mg) as a white solid.
  • Step 22d Preparation of 2-(benzyloxy)-N-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(ethyl)amino)-2 -Ethylimidazo[1,2-a]pyridin-6-yl)pyrimidin-2-yl)ethane-1-sulfonamide
  • step 22d under nitrogen atmosphere, (3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(ethyl)amino)-2-ethylimidazo[ 1,2-a]pyridin-6-yl)boronic acid (985 mg, 2.26 mmol) and 2-(benzyloxy)-N-(5-bromopyrimidin-2-yl)ethane-1-sulfonamide (371 mg, 1.51 mmol) was prepared according to the method of Example 17, step 17h to obtain a brown solid 2-(benzyloxy)-N-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazole-2- yl)(ethyl)amino)-2-ethylimidazo[1,2-a]pyridin-6-yl)pyrimidin-2-yl)ethane-1-sulfonamide (530 mg).
  • Step 22e Preparation of N-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(ethyl)amino)-2-ethylimidazo[1, 2-a]pyridin-6-yl)pyrimidin-2-yl)-2-hydroxyethane-1-sulfonamide
  • step 22e To 2-(benzyloxy)-N-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(ethyl)amino )-2-ethylimidazo[1,2-a]pyridin-6-yl)pyrimidin-2-yl)ethane-1-sulfonamide (530 mg, 0.777 mmol) was added with trifluoroacetic acid (5 mL). The mixture was heated to 90°C in the microwave and stirred for 1 hour. Then, the pH was adjusted to 8-9 with saturated sodium bicarbonate solution, and extracted with ethyl acetate (10 mL ⁇ 3).
  • Step 23a Preparation of 1-(5-Bromopyrimidin-2-yl)azetidin-3-ol
  • step 23a at room temperature, azetidin-3-ol (149 mg, 2.03 mmol) and potassium carbonate ( 511 mg, 3.7 mmol). The mixture was stirred at 80 °C for 1 h. 50 mL of water was added, filtered, and the filter cake was spin-dried to obtain 1-(5-bromopyrimidin-2-yl)azetidin-3-ol (360 mg) as an off-white solid.
  • Step 23b Preparation of (2-(3-hydroxyazetidin-1-yl)pyrimidin-5-yl)boronic acid
  • step 23b under nitrogen atmosphere, 1-(5-bromopyrimidin-2-yl)azetidin-3-ol (100 mg, 0.43 mmol) and biboronic acid pinacol ester (121 mg, 0.48 mmol) were mixed together ) was prepared according to the method of Example 17, step 17g to obtain (2-(3-hydroxyazetidin-1-yl)pyrimidin-5-yl)boronic acid (85mg).
  • Step 23c Preparation of 2-(ethyl(2-ethyl-5-(2-(3-hydroxyazetidin-1-yl)pyrimidin-5-yl)-2H-pyrazolo[4,3 -b]pyridin-3-yl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 23c under nitrogen atmosphere, (2-(3-hydroxyazetidin-1-yl)pyrimidin-5-yl)boronic acid (85mg, 0.43mmol) and 2-((5-bromo- 2-ethyl-2H-pyrazolo[4,3-b]pyridin-3-yl)(ethyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (205 mg, 0.43 mmol ) was prepared according to the method of step 17h of Example 17 to obtain 2-(ethyl(2-ethyl-5-(2-(3-hydroxyazetidin-1-yl)pyrimidin-5-yl)- 2H-pyrazolo[4,3-b]pyridin-3-yl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (38 mg).
  • Step 24a Preparation of (1-(5-Bromopyrimidin-2-yl)azetidin-3-yl)methanol
  • step 24a at room temperature, azetidin-3-ylmethanol hydrochloride (368 mg, 2.97 mmol) was added to a DMF solution (5 mL) of 5-bromo-2-chloropyrimidine (500 mg, 2.58 mmol). ) and triethylamine (1.1 mL, 7.75 mmol). The mixture was stirred at 60°C for 4 hours. After cooling, the reaction solution was concentrated and diluted with water (60 mL). The aqueous phase was extracted with ethyl acetate (20 mL ⁇ 3), the combined organic phases were washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by flash chromatography to give (1-(5-bromopyrimidin-2-yl)azetidin-3-yl)methanol (618 mg) as a yellow solid.
  • Step 24b Preparation of (2-(3-(hydroxymethyl)azetidin-1-yl)pyrimidin-5-yl)boronic acid
  • step 24b (1-(5-bromopyrimidin-2-yl)azetidin-3-yl)methanol (500 mg, 1.70 mmol) and pinacol biboronate (781 mg, 2.21 mmol) (2-(3-(hydroxymethyl)azetidin-1-yl)pyrimidin-5-yl)boronic acid (529 mg) was prepared according to the procedure of Example 17, step 17g.
  • Step 24c Preparation of 2-(ethyl(2-ethyl-5-(2-(3-(hydroxymethyl)azetidin-1-yl)pyrimidin-5-yl)-2H-pyrazolo [4,3-b]pyridin-3-yl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 24c 2-((5-bromo-2-ethyl-2H-pyrazolo[4,3-b]pyridin-3-yl)(ethyl)amino)-4-(4- Fluorophenyl)thiazole-5-carbonitrile (100 mg, 0.21 mmol) and (2-(3-(hydroxymethyl)azetidin-1-yl)pyrimidin-5-yl)boronic acid (133 mg, 0.32 mmol) ) was prepared according to the method of step 17h of Example 17 to obtain 2-(ethyl(2-ethyl-5-(2-(3-(hydroxymethyl)azetidin-1-yl)pyrimidine-5) as a white solid -yl)-2H-pyrazolo[4,3-b]pyridin-3-yl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (40 mg).
  • Step 25a Preparation of 1-(5-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(ethyl)amino)-2-ethyl-2H-pyrazole [4,3-b]pyridin-5-yl)pyrimidin-2-yl)azetidin-3-yl acetate
  • step 25a 2-(ethyl(2-ethyl-5-(2-(3-hydroxyazetidin-1-yl)pyrimidin-5-yl)-2H-pyrazolo[ 4,3-b]pyridin-3-yl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (200 mg, 0.37 mmol) and triethylamine (112 mg, 1.11 mmol) in dichloromethane (3 mL), under nitrogen protection, was added acetyl chloride (58 mg, 0.74 mmol) at 0°C.
  • Step 26a Preparation of 1-(5-Bromopyrimidin-2-yl)piperidin-4-ol
  • step 26a 2,5-dibromopyrimidine (440mg, 1.85mmol), 4-hydroxypiperidine (206mg, 2.03mmol) and potassium carbonate (511mg, 3.7mmol) were prepared according to the method of step 23a of Example 23 1-(5-Bromopyrimidin-2-yl)piperidin-4-ol (350 mg) was obtained as an off-white solid.
  • step 26b under nitrogen atmosphere, 1-(5-bromopyrimidin-2-yl)piperidin-4-ol (110 mg, 0.43 mmol) and biboronic acid pinacol ester (130 mg, 0.51 mmol) were carried out according to Example 23
  • the method of step 23b prepared (2-(4-hydroxypiperidin-1-yl)pyrimidin-5-yl)boronic acid (95 mg).
  • Step 26c Preparation of 2-(ethyl(2-ethyl-5-(2-(4-hydroxypiperidin-1-yl)pyrimidin-5-yl)-2H-pyrazolo[4,3-b] Pyridin-3-yl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 26c under nitrogen atmosphere, combine (2-(4-hydroxypiperidin-1-yl)pyrimidin-5-yl)boronic acid (95 mg, 0.43 mmol) and 2-((5-bromo-2-ethyl) yl-2H-pyrazolo[4,3-b]pyridin-3-yl)(ethyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (205 mg, 0.43 mmol) as carried out
  • Example 23 The method of step 23c prepared off-white solid 2-(ethyl(2-ethyl-5-(2-(4-hydroxypiperidin-1-yl)pyrimidin-5-yl)-2H-pyrazolo [4,3-b]pyridin-3-yl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (50 mg).
  • Step 27a Preparation of (S)-1-(5-bromopyrimidin-2-yl)pyrrolidin-3-ol
  • step 27a at room temperature, add (S)-pyrrolidin-3-ol (0.4 g, 4.7 mmol) to a DMF solution (8 mL) of 2,5-dibromopyrimidine (1 g, 4.7 mmol) as carried out
  • a DMF solution 8 mL
  • 2,5-dibromopyrimidine 1 g, 4.7 mmol
  • Example 23 The method of step 23a prepared (S)-1-(5-bromopyrimidin-2-yl)pyrrolidin-3-ol (800 mg) as a white solid.
  • Step 27b Preparation of (S)-(2-(3-hydroxypyrrolidin-1-yl)pyrimidin-5-yl)boronic acid
  • step 27b under nitrogen atmosphere, (S)-1-(5-bromopyrimidin-2-yl)pyrrolidin-3-ol (200 mg, 0.82 mmol) and pinacol biboronate (230 mg, 0.9 mmol) according to the method of Example 23, step 23b to prepare (S)-(2-(3-hydroxypyrrolidin-1-yl)pyrimidin-5-yl)boronic acid (400 mg) as a black solid.
  • Step 27c Preparation of (S)-2-(Ethyl(2-ethyl-5-(2-(3-hydroxypyrrolidin-1-yl)pyrimidin-5-yl)-2H-pyrazolo[4, 3-b]pyridin-3-yl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 27c under nitrogen atmosphere, (S)-(2-(3-hydroxypyrrolidin-1-yl)pyrimidin-5-yl)boronic acid (200 mg, 0.96 mmol) and 2-((5-bromo -2-Ethyl-2H-pyrazolo[4,3-b]pyridin-3-yl)(ethyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (200 mg, 0.42 mmol) was prepared according to the method of Example 23, Step 23c to obtain a white solid (S)-2-(ethyl(2-ethyl-5-(2-(3-hydroxypyrrolidin-1-yl)pyrimidin-5-yl) )-2H-pyrazolo[4,3-b]pyridin-3-yl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (100 mg).
  • Step 28a Preparation of (R)-1-(5-bromopyrimidin-2-yl)pyrrolidin-3-ol
  • step 28a at room temperature, 5-bromo-2-chloropyrimidine (500mg, 2.10mmol) and (R)-pyrrolidin-3-ol (210mg, 2.42mmol) were prepared according to the method of step 23a of Example 23 (R)-1-(5-bromopyrimidin-2-yl)pyrrolidin-3-ol (472 mg) was obtained as a white solid.
  • Step 28b Preparation of (R)-(2-(3-hydroxypyrrolidin-1-yl)pyrimidin-5-yl)boronic acid
  • step 28b (R)-1-(5-bromopyrimidin-2-yl)pyrrolidin-3-ol (200mg, 0.82mmol) and pinacol biboronate (312mg, 1.23mmol) were carried out according to Example 23
  • the method of step 23b prepared (R)-(2-(3-hydroxypyrrolidin-1-yl)pyrimidin-5-yl)boronic acid (171 mg).
  • Step 28c Preparation of (R)-2-(Ethyl(2-ethyl-5-(2-(3-hydroxypyrrolidin-1-yl)pyrimidin-5-yl)-2H-pyrazolo[4, 3-b]pyridin-3-yl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
  • step 28c 2-((5-bromo-2-ethyl-2H-pyrazolo[4,3-b]pyridin-3-yl)(ethyl)amino)-4-(4- Fluorophenyl)thiazole-5-carbonitrile (100 mg, 0.21 mmol) and (R)-(2-(3-hydroxypyrrolidin-1-yl)pyrimidin-5-yl)boronic acid (67 mg, 0.32 mmol) as per implementation
  • Example 23 The method of step 23c prepared a white solid (R)-2-(ethyl(2-ethyl-5-(2-(3-hydroxypyrrolidin-1-yl)pyrimidin-5-yl)-2H- Pyrazolo[4,3-b]pyridin-3-yl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (56 mg).
  • step 29a at room temperature, 5-bromo-2-chloropyrimidine (500mg, 2.10mmol) and morpholine (200mg, 2.3mmol) were prepared according to the method of step 23a of Example 23 to prepare a white solid 4-(5- bromopyrimidin-2-yl)morpholine (400 mg).
  • step 29b (4-(5-bromopyrimidin-2-yl)morpholine (200 mg, 0.82 mmol) and pinacol biboronate (312 mg, 1.23 mmol) were prepared according to the method of step 23b of Example 23 (2-morpholinopyrimidin-5-yl)boronic acid (180 mg) was obtained.
  • Step 29c Preparation of 2-(Ethyl(2-ethyl-5-(2-morpholinopyrimidin-5-yl)-2H-pyrazolo[4,3-b]pyridin-3-yl)amino) -4-(4-Fluorophenyl)thiazole-5-carbonitrile
  • step 29c 2-((5-bromo-2-ethyl-2H-pyrazolo[4,3-b]pyridin-3-yl)(ethyl)amino)-4-(4- Fluorophenyl)thiazole-5-carbonitrile (100 mg, 0.21 mmol) and (2-morpholinopyrimidin-5-yl)boronic acid (84 mg, 0.40 mmol) were prepared according to the method of Example 23, Step 23c to give a white solid 2- (Ethyl(2-ethyl-5-(2-morpholinopyrimidin-5-yl)-2H-pyrazolo[4,3-b]pyridin-3-yl)amino)-4-(4- Fluorophenyl)thiazole-5-carbonitrile (60 mg).
  • test compounds were dissolved in DMSO to make a 20 mM stock solution, and 4-fold serial dilutions were carried out with DMSO, with a starting concentration of 20 ⁇ M, for a total of 8 concentration gradients.
  • Example IC 50 ( ⁇ M) Example IC 50 ( ⁇ M) 1 0.030 2 0.029 3 0.0052 4 0.021 5 0.048 6 0.029 7 0.025 8 0.0044 9 0.028 10 0.018 11 0.025 12 0.019 13 0.013 14 0.042 15 0.040 16 0.039 17 0.0074 18 0.015 19 0.017 20 0.045 twenty one 0.011 twenty three 0.011 twenty four 0.011 25 0.012 26 0.0055 27 0.0046 28 0.0038 29 0.023 GLPG1690 0.112
  • Metabolism experiments of SD rats are carried out to the compounds of various embodiments of the present invention, respectively taking a single intravenous injection (IV, dose 2mg/kg) and a single oral administration (PO, dose 10mg/kg), respectively. Blood was collected before administration and at 5 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours, 12 hours and 24 hours after administration to measure the concentration of the test compound in plasma.
  • the pharmacokinetic data of the control compound and Examples 17, 26, 28 and 29 of the present invention in rat plasma are shown in Tables 4-8.
  • Example 3 In vitro micronucleus assay of Chinese hamster ovary cells
  • Example 26 The compound of Example 26 and GLPG1690 induced micronucleus formation in Chinese hamster ovary cells (CHO-WBL) with or without exogenous metabolic activation system ( ⁇ -naphthoflavone and phenobarbital-induced rat liver S9) ability to evaluate its clastogenic/aneuploidy-inducing potential.
  • CHO-WBL Chinese hamster ovary cells
  • ⁇ -naphthoflavone and phenobarbital-induced rat liver S9 ability to evaluate its clastogenic/aneuploidy-inducing potential.
  • CHO-WBL cells were exposed to at least 3 concentrations of the test substance, and a vehicle and a positive control were set up at the same time. Each dose group was set up with 2 wells of cells. In the non-activated test system, the dosing time was 3 and 24 hours, and in the S9 activated test system, the test substance exposure time was 3 hours.
  • the upper limit of the test substance tested depends on its solubility in the medium, but generally does not exceed the maximum concentration of 1 mM or 0.5 mg/mL, whichever is lower.
  • the cytotoxicity of the test substance dose group should not exceed 50% too much compared to the corresponding vehicle control group. If the highest concentration is not limited by cytotoxicity, in order to reach the upper limit of solubility in the medium, multiple doses containing visible precipitates can be tested, and the highest dose group selected for micronucleus analysis showed a small amount of identifiable precipitates in the medium.
  • test results are evaluated according to the following criteria:
  • test substance meets the following criteria at the same time, the test substance can be considered as positive:
  • a significant increase in the frequency of micronucleated cells was observed in one or more concentration groups.
  • the frequency of micronucleated cells observed in one or more concentration groups was outside the range of laboratory negative historical data.
  • micronucleus frequency had a dose-response relationship.
  • test substance can be considered as negative.
  • the frequency of binucleate cells containing micronuclei was significantly increased at AKEX0070 concentrations of 7 and 8 ⁇ g/mL of GLPG1690 in the non-metabolically activated dosing 24-hour series, and the increase in the rate of micronuclei was seen to be dose-dependent relation.
  • the positive controls cyclophosphamide monohydrate and mitomycin C
  • the potential of GLPG1690 to induce micronucleated cells in CHO-WBL cells was positive.
  • GLPG1690 is genotoxic, and the compound of Example 26 has a better safety profile than GLPG1690.
  • CYP450 enzyme single-concentration point inhibition test use 320 ⁇ L of human liver microsomes (for 3A4 subtype, the final concentration is 0.05 mg/mL) for direct inhibition incubation, the system contains NADPH (final concentration 1.3 mM), 10 ⁇ M compound, Positive inhibitor (ketoconazole 0.1 ⁇ M), negative control (BPS in 0.1% DMSO) and mixed probe substrate (midazolam 5 ⁇ M) were incubated for 5 min to terminate the reaction. The relative enzyme activity was calculated by measuring the relative production of metabolites.
  • the single-point inhibition data of CYP450 enzyme at 10 ⁇ M of the control compound GLPG1690 and the compounds of Examples 17, 23, 26 and 27 of the present invention are shown in the following table.
  • the results showed that the inhibitory activity of Examples 17, 23, 26, and 27 on the 3A4 subtype was lower than that of GLPG1690, and the risk of drug-drug interaction in clinical practice was lower.
  • Rapidly activated human delayed rectifier outward potassium current (I Kr ) is mainly mediated by the hERG ion channel and is involved in human cardiomyocyte repolarization. Drugs to block this current will lead to the clinical appearance of QT interval prolongation syndrome, which is easy to induce acute arrhythmia and even sudden death.
  • the method of manual patch clamp was used to test the effect of several example compounds on hERG potassium current on stable cell lines transfected with hERG potassium channel, so as to determine whether the test substance has an inhibitory effect on hERG ion channel.
  • hERG currents were recorded by whole-cell patch clamp technique.
  • the cell suspension was added to a small petri dish and placed on an inverted microscope stage. After the cells have adhered, perfuse with extracellular fluid.
  • the recommended flow rate is 1–2 mL/min.
  • the glass micro-electrode is drawn by a micro-electrode drawing apparatus in two steps, and the resistance value of the glass micro-electrode is 2-5M ⁇ after filling with the liquid in the electrode. After establishing the whole-cell recording mode, hold the clamp potential at -80mV.
  • a depolarization voltage was given to +60mV for 850ms, followed by repolarization to -50mV for 1275ms to elicit hERG tail currents.
  • IR 100% ⁇ (peak tail current before administration-peak tail current after administration)/peak tail current before administration.
  • IC 50 values were obtained by fitting the Hill equation (if applicable), and no IC 50 values were calculated if the maximum inhibition rate at all concentrations of the test substance was less than 50%.
  • CYP450 enzyme 3A4 isoform testosterone substrate time-dependent inhibition assay (IC 50 shift): pre-incubated with 100 ⁇ L of human liver microsomes (final concentration 0.2 mg/mL) with or without NADPH for 30 min and then supplemented with 20 ⁇ L NADPH was incubated for inhibition, and the final concentration of NADPH in the system was 1.3 mM.
  • the concentration of compound series starts from 100 ⁇ M, and there are 8 concentration points (including 0 concentration point) in 3-fold dilution.
  • the positive inhibitor was mifepristone, with serial concentrations starting from 10 ⁇ M and 3-fold dilution for a total of 8 concentration points (including 0 concentration point).
  • the probe substrate was testosterone at a concentration of 50 ⁇ M.
  • the reaction was terminated after the system was incubated for 10 min.
  • the IC 50 of the compound on the enzyme inhibition rate was calculated by measuring the relative production of metabolites, and the IC 50 shift value was calculated from the IC 50 results of the incubation groups with and without NADPH.
  • the time-dependent inhibition data of the compounds of Examples 4, 17, 23, 26, 27, 28 and 29 of the present invention to CYP450 enzyme 3A4 are shown in the following table.
  • the results showed that the time-dependent inhibition of CYP3A4 by the compounds of Examples 4, 17, 23, 26, 27, 28 and 29 was weaker than that of GLPG1690, and the risk of drug-drug interaction in clinical practice was lower.
  • the aforementioned compounds represented by formula I', formula I, formula I-1, formula II, formula II-1, formula III or formula III-1, formula I', formula I Different realization modes of compounds represented by formula I-1, formula II, formula II-1, formula III or formula III-1 and formula I', formula I, formula I-1, formula II, formula II-1, formula III
  • all the compounds involved in the specific examples of the compound represented by formula III-1 can be made into corresponding isomers, solvates, hydrates, prodrugs, stable isotope derivatives and pharmaceutically acceptable salts .
  • the compound is formulated into a pharmaceutically acceptable derivative, the derivative being any of prodrugs, salts, esters, amides, salts of esters, salts of amides, and metabolites.
  • pharmaceutically acceptable salts include those obtained by using inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid or phosphoric acid, or using organic acids such as acetic acid, propionic acid, succinic acid, benzoic acid, p-aminobenzenesulfonic acid, 2-Acetoxy-benzoic acid, cinnamic acid, mandelic acid, salicylic acid, glycolic acid, lactic acid, oxalic acid, malic acid, maleic acid, malonic acid, fumaric acid, tartaric acid, citric acid, p-toluenesulfonic acid acid, methanesulfonic acid, ethanesulfonic acid or benzenesulfonic acid) conventional non-toxic salts obtained by salifying any of the compounds involved in the present invention.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid or phosphoric acid
  • organic acids such as acetic acid, propionic acid, succ
  • stable isotope derivatives can introduce isotopes into any compound involved in the present invention, and the introduced isotopes can be 2 H, 3 H, 13 C, 14 C, 15 N, 17 O, 18 O, 31 P , 32 P, 35 S, 18 F, 36 Cl, and specific isotopic derivatives can be prepared by conventional techniques.
  • an actual product it can also be made into tablets, capsules, injections, granules, powders, suppositories, pills, creams, pastes, gel powders, oral solutions, inhalants, suspensions, Any of dry suspension, patch and lotion.
  • a mixture can also be formed with any of the following substances: a pharmaceutically acceptable carrier or adjuvant or excipient.
  • All the compounds involved in the present invention and mixtures, compositions and the like containing the compounds of the present invention can be administered to a living body via any route of administration.
  • the route of administration can be oral administration, intravenous injection, intramuscular injection, subcutaneous injection, rectal administration, vaginal administration, sublingual administration, nasal inhalation, oral inhalation, eye drop, and local or systemic transdermal administration.
  • All the compounds involved in the present invention and mixtures, compositions, etc. containing the compounds of the present invention can be formulated into a single dose, which contains the active compounds of the present invention, carriers, excipients, etc., and the dosage form can be tablets, capsules, etc. , injections, granules, powders, suppositories, pills, creams, pastes, gels, powders, oral solutions, inhalants, suspensions, dry suspensions, patches, lotions, etc.
  • These dosage forms may contain ingredients commonly used in pharmaceutical preparations, such as diluents, absorbents, wetting agents, binders, disintegrants, colorants, pH adjusters, antioxidants, bacteriostatic agents, isotonicity adjusters, Anti-sticking agent, etc.
  • ingredients commonly used in pharmaceutical preparations such as diluents, absorbents, wetting agents, binders, disintegrants, colorants, pH adjusters, antioxidants, bacteriostatic agents, isotonicity adjusters, Anti-sticking agent, etc.
  • Suitable formulations for the various dosage forms described above are available from published sources, for example, Remington: The Science and Practice of Pharmacy, 21st Edition, Lippincott Williams & Wilkins, 2006 and Rowe, Raymond C. Handbook of Pharmaceutical Excipients, Chicago, Pharmaceutical Press, 2005 Published in 2000. Therefore, it can be easily prepared by those skilled in the art.
  • Different dosages can be selected according to the nature, intensity, age, sex, body weight, route of administration and other factors of the diseases suffered by different individuals.
  • the dosage of the compounds of the present invention can be 0.01 to 500 mg/kg per day.
  • the preferred daily dose is 1-100 mg/kg, which can be administered in single or multiple doses.
  • fibrotic disease fibrotic disease, metabolic disease, myelodysplastic syndrome, respiratory disease, cardiovascular disease, autoimmune disease, inflammation, dermatological disease, neurological disease or pain.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Hospice & Palliative Care (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pulmonology (AREA)
  • Cardiology (AREA)
  • Psychiatry (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Immunology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

一种ATX抑制剂及其制备方法和应用,属于药物化学技术领域。具体而言,所述ATX抑制剂为具备式I'结构的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物。相对于现有的ATX抑制剂GLPG-1690,所述的ATX抑制剂具有更高的抑制活性,同时具有优异的药效、体外/体内药代动力学性质和安全性,临床应用前景高。

Description

一种ATX抑制剂及其制备方法和应用
相关申请的引用
本发明要求2020年7月9日在中国提交的,名称为“一种ATX抑制剂及其制备方法和应用”、申请号为202010658085.8的发明专利申请和2021年3月23日在中国提交的,名称为“一种ATX抑制剂及其制备方法和应用”、申请号为202110308656.X的发明专利申请的优先权,通过引用的方式将该专利申请的全部内容并入本文。
技术领域
本发明属于药物化学领域,具体涉及一种ATX抑制剂及其制备方法。
背景技术
Autotaxin(ATX,又称ENPP2[外核苷酸焦磷酸酶/磷酸二酯酶2]或溶血磷脂酶D)是外核苷酸焦磷酸酶/磷酸二酯酶(ENPP)家族中的一员。ATX能够将溶血磷脂酰胆碱(LPC)转化为生物活性的溶血磷脂酸(LPA)。LPA由一个甘油链、一个磷酸基和一个长度及饱和度不同的脂肪酰基链组成。LPA一旦生成,能够通过六个细胞表面特异的受体蛋白(LPA1-6),即G蛋白偶联受体介导发挥其生物学活性。许多证据表明ATX是血液中LPA的主要来源。ATX-LPA信号通路参与细胞存活、迁移和增殖,从而与许多严重疾病有着重要联系,如纤维化疾病(主要是特发性肺纤维化,IPF)、癌症、增殖性疾病、炎症性疾病、自身免疫疾病、心血管疾病、神经变性疾病等。事实上,ATX的过度表达经常出现在癌症组织中,如乳腺癌、结肠直肠癌和胶质母细胞瘤等。
目前,GLPG-1690对ATX有抑制作用,已进入临床III期试验阶段,用于治疗特发性肺纤维化(IPF)。一些证据表明,通过调节肺上皮细胞、成纤维细胞和平滑肌细胞的生物学,ATX/LPA信号在各种肺部疾病中发挥作用。
然而,GLPG-1690对ATX虽然有抑制活性,但是其抑制活性并不是很高,因此,仍需开发更多的对ATX具有更高抑制活性的抑制剂。
发明内容
发明要解决的问题
为了解决上述技术问题,本发明提供了一类新的杂芳环类化合物,其对ATX具有很高的抑制活性,同时具有优异的药效、体外/体内药代动力学性质和安全性,临床应用前景高。
用于解决问题的方案
为了解决上述技术问题,本发明提供了以下技术方案:
一种具备式I’结构的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物:
Figure PCTCN2021105180-appb-000001
其中,
n为0至5中的任一整数;
若存在,每一个R 1各自独立地选自氢、氘、氰基、卤素、氨基、羟基、-COOH、-CHO、-NO 2、C 1-6烷基氨基、C 1-6烷氧基和未取代或被选自氰基、氨基、羟基、卤素、C 1-6烷基和C 1-6烷氧基的取代基所取代的C 1-6烷基、C 3-7环烷基和3-7元杂环烷基;
R 2选自氢、氘、卤素、氰基、氨基、羟基、-COOH、-CHO、-NO 2、C 1-6烷基、C 1-6烷氧基、C 1-6烷基氨基、-C(=O)NH 2、-NH(C=O)CH 3和3-7元杂环烷基;
R 3选自氢、氘、C 1-6烷基、C 3-7环烷基和被选自氰基、氨基、羟基、卤素、C 1-6烷基和C 1-6烷氧基的取代基所取代的C 1-6烷基、C 3-7环烷基和3-7元杂环烷基;
环B为下列结构的任一种:
Figure PCTCN2021105180-appb-000002
R 4选自氢、氘、卤素、氰基、羟基、氨基、C 1-6烷基、C 1-6烷氧基、C 1-6烷基氨基、C 3-7环烷基和3-7元杂环烷基;
X 1、X 2、X 3和X 4各自独立地为N或CR 5
R 5选自氢、氘、卤素、C 1-6烷基和被选自氰基、氨基、羟基、卤素、C 1-6烷基和C 1-6烷氧基的取代基所取代的C 1-6烷基、C 3-7环烷基和3-7元杂环烷基;
环A选自取代或未取代的C 6-10芳基和5-10元杂芳基,所述取代是被1至3个选自羟基、氨基、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3-7元杂环烷基、C 1-6烷基氨基、C 1-6烷氧基的取代基所取代;
R 6为-L 1-L 2-W 1
L 1选自化学键、C 1-3亚烷基、-O-、-C(=O)-、-C(=O)C(R 8) 2-、-C(=O)O-、-C(=O)NH-、-OC(=O)-、-NH-、-SO 2-、-NHSO 2-、-SO 2NH-;其中,R 8为氢或C 1-6烷基;
L 2选自化学键、-O-、-C(=O)-、-C(=O)CH 2-、-C(=O)O-、-OC(=O)-、-C(=O)NH-、-NR 7-、-NHCH 2-、-SO 2-、-NR 7SO 2-、-SO 2NR 7-、-C(=O)NR 7-和-NR 7C(=O)-;其中,R 7为氢或C 1-6烷基;
W 1为取代或未取代的下列基团:氢、氘、氨基、氰基、C 1-6烷基、卤素、羟基、C 1-6烷氧基、C 1- 6烷基氨基、羧基、C 6-10芳基、5-10元杂芳基、C 3-7环烷基或3-7元杂环烷基;所述取代是被1或2个选自氧代、羟基、氨基、羟甲基、氨基甲基、氰基、C 1-6烷基、C 1-6烷基-C(=O)O-和卤素的取代基所取代。
进一步地,上述式I’化合物为具备式I结构的化合物,
Figure PCTCN2021105180-appb-000003
其中,n、R 1、R 2、R 3、R 4、R 6、X 1、X 2、X 3和环A如式I’中所定义。
更进一步地,上述式I’化合物为具备式I-1结构的化合物,
Figure PCTCN2021105180-appb-000004
其中,n、R 1、R 2、R 3、R 4、R 6和环A如式I’中所定义。
进一步地,上述式I’化合物为具备式II结构的化合物,
Figure PCTCN2021105180-appb-000005
其中,n、R 1、R 2、R 3、R 4、R 6、X 1、X 2、X 3和环A如式I’中所定义。
更进一步地,上述式I’化合物为具备式II-1结构的化合物,
Figure PCTCN2021105180-appb-000006
其中,n、R 1、R 2、R 3、R 4、R 6和环A如式I’中所定义。
进一步地,上述式I’化合物为具备式III结构的化合物,
Figure PCTCN2021105180-appb-000007
其中,n、R 1、R 2、R 3、R 4、R 6、X 1、X 2、X 3和环A如式I’中所定义。
更进一步地,上述式I’化合物为具备式III-1结构的化合物,
Figure PCTCN2021105180-appb-000008
其中,n、R 1、R 2、R 3、R 4、R 6和环A如式I’中所定义。
本发明还提供了下列化合物:
(1)2-((2-乙基-6-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)苯基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(2)2-((2-乙基-6-(6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(3)2-((2-乙基-6-(5-氟-6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(4)2-((2-乙基-6-(5-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(5)2-((2-乙基-6-(6-2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哒嗪-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(6)2-((2-乙基-6-(6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)-2-甲基吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(7)2-((2-乙基-6-(5-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(8)2-((2-乙基-6-(5-(3-羟基氮杂环丁烷-1-羰基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(9)2-((2-乙基-6-(5-(1-(3-羟基氮杂环丁烷-1-基)-2-甲基-1-氧代丙-2-基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(10)2-((2-乙基-6-(3-氟-5-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(11)(S)-2-((2-乙基-6-(5-(2-(3-羟基吡咯烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(12)2-((2-乙基-6-(5-(2-(3-(羟甲基)氮杂环丁烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(13)(R)-2-((2-乙基-6-(5-(2-(3-羟基吡咯烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(14)2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)-N-((5-氧代吡咯烷-2-基)甲基)乙酰胺;
(15)5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-N-(2-氮杂螺[3.3]庚烷-6-基)吡嗪-2-甲酰胺;
(16)2-((2-乙基-6-(5-氟-6-(3-羟基氮杂环丁烷-1-羰基)吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(17)2-((5-(2-(1,1-二氧化异噻唑烷-2-基)嘧啶-5-基)-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(18)2-((5-(2-(1,1-二氧化异噻唑烷-2-基)嘧啶-5-基)-2-乙基吡唑并[1,5-a]嘧啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(19)2-((6-(5-(1,1-二氧化异噻唑烷-2-基)吡嗪-2-基)-2-乙基咪唑并[1,2-a]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(20)N-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)-N-甲基甲磺酰胺;
(21)2-((5-(2-(1,1-二氧化-1,2-噻嗪烷-2-基)嘧啶-5-基)-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(22)N-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)-2-羟基乙-1-磺酰胺;
(23)2-(乙基(2-乙基-5-(2-(3-羟基氮杂环丁烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(24)2-(乙基(2-乙基-5-(2-(3-(羟甲基)氮杂环丁烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(25)1-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基-2H-吡唑并[4,3-b]吡啶-5-基)嘧啶-2-基)氮杂环丁烷-3-基乙酸酯;
(26)2-(乙基(2-乙基-5-(2-(4-羟基哌啶-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(27)(S)-2-(乙基(2-乙基-5-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;
(28)(R)-2-(乙基(2-乙基-5-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈;和
(29)2-(乙基(2-乙基-5-(2-吗啉基嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈。
本发明还提供了一种药物组合物,其包含上述具备式I’、式I、式I-1、式II、式II-1、式III或式III-1结构的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物。
本发明还提供了一种药物制剂,其包括上述具有式I’、式I、式I-1、式II、式II-1、式III或式III-1结构的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物或者上述药物组合物,所述制剂为片剂、胶囊剂、注射剂、颗粒剂、粉剂、栓剂、丸剂、乳膏剂、糊剂、凝胶剂、散剂、口服溶液、吸入剂、混悬剂、干悬剂、贴剂、洗剂中的任一种。
本发明还提供了上述具有式I’、式I、式I-1、式II、式II-1、式III或式III-1结构的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,或者上述药物组合物或者药物制剂,其用于预防和/或治疗具有ATX表达增加的病理学特征的相关疾病;优选的,所述具有ATX表达增加的病理学特征的相关疾病包括:癌症、纤维化疾病、代谢疾病、骨髓增生异常综合征、呼吸系统疾病、心血管疾病、自身免疫性疾病、炎症、皮肤学疾病、神经系统疾病或疼痛;更优选的,所述具有ATX表达增加的病理学特征的相关疾病为肺纤维化、肾纤维化或肝纤维化。
本发明还提供给了上述具有式I’、式I、式I-1、式II、式II-1、式III或式III-1结构的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,或者上述药物组合物或者药物制剂在制备用于预防和/或治疗具有ATX表达增加的病理学特征的相关疾病的药物中的应用;优选的,所述具有ATX表达增加的病理学特征的相关疾病包括:癌症、纤维化疾病、代谢疾病、骨髓增生异常综合征、呼吸系统疾病、心血管疾病、自身免疫性疾病、炎症、皮肤学疾病、神经系统疾病或疼痛;更优选的,所述具有ATX表达增加的病理学特征的相关疾病为肺纤维化、肾纤维化或肝纤维化。
本发明还提供了一种预防和/或治疗具有ATX表达增加的病理学特征的相关疾病的方法,其包括下列步骤:将治疗有效量的上述具有式I’、式I、式I-1、式II、式II-1、式III或式III-1结构的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,或者上述药物组合物或者药物制剂施用于对其有需求的患者。
发明的效果
本发明的化合物相对于现有的ATX抑制剂GLPG-1690,具有更高的抑制活性,同时具有优异的药效、体外/体内药代动力学性质和安全性,临床应用前景高。
具体实施方式
在进一步描述本发明之前,应当理解,本发明不限于本文中所述的特定实施方案;还应该理解,本文中所使用的术语仅用于描述而非限制特定实施方案。
[术语定义]
除非另有说明,下列术语的含义如下。
术语“C 1-6烷基”单独或者以组合方式表示包含1-6个、特别是1-4个碳原子的饱和直链或支链的烷基,包括甲基、乙基、丙基、异丙基、丁基、仲丁基、异丁基、叔丁基、正戊基、2-戊基、3-戊基、2-甲基-2-丁基、3-甲基-2-丁基、3-甲基-1-丁基、2-甲基-1-丁基、正己基、2-己基、3-己基、2-甲基-2-戊基、3-甲基-2-戊基、4-甲基-2-戊基、3-甲基-3-戊基、2-甲基-3-戊基、2,3-二甲基-2-丁基、3,3,-二甲基-2-丁基等。优选地,“C 1-6烷基”是甲基、乙基、异丙基、叔丁基中的任一种。
术语“C 1-3亚烷基”单独或者以组合方式表示包含1-3个碳原子的饱和直链或支链的亚烷基,包括亚甲基(-CH 2-)、亚乙基(-CH 2CH 2-)、亚正丙基(-CH 2CH 2CH 2-)和亚异丙基(-(CH 3) 2C-)等。
术语“C 3-7环烷基”单独或者以组合方式表示具有3-7个、特别是3-6个碳原子的饱和环烷基,包括环丙基、环丁基、环戊基、环己基、环庚基等。特别的“C 3- 7环烷基”是环丙基、环戊基、环己基等。
术语“C 1-6烷氧基”单独或者以组合方式表示基团C 1- 6烷基-O-,其中“C 1-6烷基”如以上所定义。
术语“卤素”单独或者以组合方式表示氟、氯、溴或碘,特别的是氟、氯或溴。
术语“氨基”单独或者以组合方式表示伯氨基(-NH 2)、仲氨基(-NH-)或叔氨基
Figure PCTCN2021105180-appb-000009
术语“C 1-6烷基氨基”,也叫“C 1-6烷氨基”单独或者以组合方式表示如上所定义的“氨基”,其中氨基上的氢原子被至少一个C 1-6烷基所取代,其中“C 1-6烷基”如以上所定义。相应地,“C 1-6烷基氨基”包括甲基氨基,乙基氨基,丙基氨基,异丙基氨基,正丁基氨基,异丁基氨基,2-丁基氨基,叔丁基氨基,正戊基氨基、2-戊基氨基、3-戊基氨基、2-甲基-2-丁基氨基、3-甲基-2-丁基氨基、3-甲基-1-丁基氨基、2-甲基-1-丁基氨基、正己基氨基、2-己基氨基、3-己基氨基、2-甲基-2-戊基氨基、3-甲基-2-戊基氨基、 4-甲基-2-戊基氨基、3-甲基-3-戊基氨基、2-甲基-3-戊基氨基、2,3-二甲基-2-丁基氨基、3,3-二甲基-2-丁基氨基等。特别的,“C 1-6烷基氨基”是甲基氨基、乙基氨基、异丙基氨基、叔丁基氨基等。在本发明中,所述C 1-6烷基氨基可进一步任选被本领域常见的取代基团所取代。
术语“羰基”,又称“-C(=O)-”,是指二价的基团,其仅由一个碳原子和一个氧原子构成,碳原子和氧原子之间通过双键连接,并且自身结构中的碳原子还分别通过单键连接至其他两个片段。
术语“杂环烷基”,又称“杂环基”,指由碳原子与氮、氧或硫等杂原子组成的饱和或部分不饱和(包含1或2个双键)的非芳香环状基团,此环状基团可以是单环或双环基团,在本发明中,杂环烷基中碳原子个数为2-11个,杂原子个数优选1、2、3或4,杂环烷基中的氮、碳或硫原子可任选地被氧化。“杂环烷基”上的氢原子独立任选地被一个或多个本发明所描述的取代基所取代。“杂环烷基”可以通过环上任意的环原子链接到母体分子上。术语“3-6元杂环烷基”和“3-7元杂环烷基”分别是指包含3-6个和3-7个碳原子和杂原子或杂原子基团的饱和或部分不饱和单环或多环杂环烷基,所述杂原子或杂原子基团选自N、O、S(O) m(其中m是0至2中的任一整数);例如吖丙啶基、吖丁啶基、氧杂环丁基、四氢吡咯基、氧代吡咯烷基、四氢呋喃基、四氢噻吩基、哌啶基、吗啉基、哌嗪基、硫代吗啉基、四氢吡喃基、1,1-二氧化硫代吗啉基和
Figure PCTCN2021105180-appb-000010
等。
术语“芳基”表示任何稳定的6-10元单环或双环芳香族基团,包括苯基、萘基、四氢萘基、2,3-二氢化茚基或联苯基等。“芳基”上的氢原子独立任选地被一个或多个本发明所描述的取代基所取代。
术语“杂芳基”表示环上的碳原子被至少一个选自硫、氧或氮的杂原子置换形成的芳香环基团,此芳香环基团可以是5-7元单环或7-12双环基团。在本发明中,杂芳基中杂原子个数优选1、2、3或4,例如噻吩基、吡啶基、嘧啶基、吡嗪基、哒嗪基、吡啶-2(1H)-酮基、吡啶-4(1H)-酮基、吡咯基、吡唑基、噻唑基、1,2,3-三唑基、1,2,4-三唑基、咪唑基、四氮唑基、异噻唑基、噁唑基、异噁唑基、噻二唑基、噁二唑基、萘基、苯并噻吩基、吲哚基、苯并咪唑基、苯并噻唑基、苯并呋喃基、喹啉基、异喹啉基、喹唑啉基等。“杂芳基”上的氢原子独立任选地被一个或多个本发明所描述的取代基所取代。
术语“C 6-10芳基”表示具有6-10个碳原子的芳基,其中“芳基”如以上所定义。
术语“5-10元杂芳基”表示具有5-10个原子的芳杂基,其中“杂芳基”如以上所定义。
术语“氰基”单独或者以组合方式表示基团-CN。
术语“羧基”单独或者以组合方式表示基团-COOH。
术语“羟基”单独或者以组合方式表示基团-OH。
术语“立体异构体”包含所有的同分异构形式,包括对映异构体、非对映异构体和几何异构体(顺反异构体)。因此,本发明中所设计的化合物的单个立体化学异构体或其对映异构体、非对映异构体、或几何异构体(或顺反异构体)的混合物都属于本发明的范围。
术语“药学上可接受的盐”表示本发明的化合物以它们的药用盐的形式存在,包括酸加成盐和碱加成盐。在本发明中,药学上可接受的无毒的酸加成盐表示本发明中的化合物与有机或无机酸形成的盐,有机或无机酸包括但不限于盐酸、硫酸、氢溴酸、氢碘酸、磷酸、硝酸、高氯酸、乙酸、草酸、马来酸、富马酸、酒石酸、苯磺酸、甲磺酸、水杨酸、琥珀酸、柠檬酸、乳酸、丙酸、苯甲酸、对甲苯磺酸、苹果酸等。药学上可接受的无毒的碱加成盐表示本发明中的化合物与有机或无机碱所形成的盐,包括但不限于碱金属盐,例如锂、钠或钾盐;碱土金属盐,例如钙或镁盐;有机碱盐,例如通过与含N基团的有机碱形成的铵盐或N +(C 1-6烷基) 4盐。
术语“溶剂化物”表示一个或多个溶剂分子与本发明中的化合物所形成的缔合物。形成溶剂化物的溶剂包括但不限于水、甲醇、乙醇、异丙醇、乙酸乙酯、四氢呋喃、N,N-二甲基甲酰胺、二甲亚砜等。
术语“水合物”是指水与本发明中的化合物形成的缔合物。
术语“前药”表示作为本发明的化合物的化学衍生物,该衍生物在体内通过发生化学反应转换成通式I所表示的化合物。
术语“同位素衍生物”表示通式I中的氢原子被1-6个氘原子所取代得到的同位素衍生物、通式I中的碳原子被1-3个碳14原子所取代得到的同位素衍生物。
以上对本发明的涉及的术语进行了定义,本领域技术人员还可以结合现有技术对以上术语进行理解,以下基于本发明的内容以及对术语的定义进一步进行描述。
[通式化合物]
一种具备式I’结构的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物:
Figure PCTCN2021105180-appb-000011
n为0至5中的任一整数;
若存在,每一个R 1各自独立地选自氢、氘、氰基、卤素、氨基、羟基、-COOH、-CHO、-NO 2、C 1-6烷基氨基、C 1-6烷氧基和未取代或被选自氰基、氨基、羟基、卤素、C 1-6烷基和C 1-6烷氧基的取代基所取代的C 1-6烷基、C 3-7环烷基和3-7元杂环烷基;
R 2选自氢、氘、卤素、氰基、氨基、羟基、-COOH、-CHO、-NO 2、C 1-6烷基、C 1-6烷氧基、C 1-6烷基氨基、-C(=O)NH 2、-NH(C=O)CH 3和3-7元杂环烷基;
R 3选自氢、氘、C 1-6烷基、C 3-7环烷基和被选自氰基、氨基、羟基、卤素、C 1-6烷基和C 1-6烷氧基的取代基所取代的C 1-6烷基、C 3-7环烷基和3-7元杂环烷基;
环B为下列结构的任一种:
Figure PCTCN2021105180-appb-000012
R 4选自氢、氘、卤素、氰基、羟基、氨基、C 1-6烷基、C 1-6烷氧基、C 1-6烷基氨基、C 3-7环烷基和3-7元杂环烷基;
X 1、X 2、X 3和X 4各自独立地为N或CR 5
R 5选自氢、氘、卤素、C 1-6烷基和被选自氰基、氨基、羟基、卤素、C 1-6烷基和C 1-6烷氧基的取代基所取代的C 1-6烷基、C 3-7环烷基和3-7元杂环烷基;
环A选自取代或未取代的C 6-10芳基和5-10元杂芳基,所述取代是被1至3个选自羟基、氨基、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3-7元杂环烷基、C 1-6烷基氨基、C 1-6烷氧基的取代基所取代;
R 6为-L 1-L 2-W 1
L 1选自化学键、C 1-3亚烷基、-O-、-C(=O)-、-C(=O)C(R 8) 2-、-C(=O)O-、-C(=O)NH-、-OC(=O)-、-NH-、-SO 2-、-NHSO 2-、-SO 2NH-;其中,R 8为氢或C 1-6烷基;
L 2选自化学键、-O-、-C(=O)-、-C(=O)CH 2-、-C(=O)O-、-OC(=O)-、-C(=O)NH-、-NR 7-、-NHCH 2-、-SO 2-、-NR 7SO 2-、-SO 2NR 7-、-C(=O)NR 7-和-NR 7C(=O)-;其中,R 7为氢或C 1-6烷基;
W 1为取代或未取代的下列基团:氢、氘、氨基、氰基、C 1-6烷基、卤素、羟基、C 1-6烷氧基、C 1- 6烷基氨基、羧基、C 6-10芳基、5-10元杂芳基、C 3-7环烷基或3-7元杂环烷基;所述取代是被1或2个选自氧代、羟基、氨基、羟甲基、氨基甲基、氰基、C 1-6烷基、C 1-6烷基-C(=O)O-和卤素的取代基所取代。
在本发明的一些优选的实施方案中,n为0至2中的任一整数,优选n为1;若存在,每一个R 1各自独立地选自氢、氰基和卤素,优选卤素,更优选氟。
在本发明的一些优选的实施方案中,其特征在于,R 2为氰基。
在本发明的一些优选的实施方案中,R 3为C 1-6烷基,优选甲基或乙基。
在本发明的一些优选的实施方案中,R 4选自氢、氘、甲基、乙基、异丙基和环丙基,优选乙基。
在本发明的一些优选的实施方案中,X 1、X 2、X 3和X 4各自独立地为N或CR 5,R 5选自氢、氘、卤素、甲基和卤代甲基,优选氢。
在本发明的一些优选的实施方案中,环A选自亚苯基、亚吡啶基、亚吡嗪基、亚哒嗪基、亚嘧啶基、亚吡唑基、亚咪唑基、亚噁唑基、亚异噁唑基、亚噻唑基、亚噻二唑基和亚噁二唑基。
在本发明的一些更优选的实施方案中,环A选自下列结构的任一种:
Figure PCTCN2021105180-appb-000013
优选下列结构中的任一种:
Figure PCTCN2021105180-appb-000014
在本发明的一些优选的实施方案中,环A被1至3个,优选1个取代基所取代,所述取代基各自独立地选自羟基、氨基、卤素、氰基和C 1-6烷基,优选卤素和C 1-6烷基,更优选氟和甲基。
在本发明的一些优选的实施方案中,L 1选自化学键、-CH 2-、-CH(CH 3)-、-C(CH 3) 2-、-C(=O)-、-SO 2-、-C(=O)CH 2-和-C(=O)NH-,优选化学键、-CH 2-、-C(CH 3) 2-和-C(=O)CH 2-。
在本发明的一些优选的实施方案中,L 2选自化学键、-NH-、-C(=O)-、-SO 2-、-NHCH 2-、-C(=O)CH 2-、-C(=O)NH-、-NHSO 2-和-N(CH 3)SO 2-,优选化学键、-C(=O)-、-NHCH 2-、-C(=O)NH-、-NHSO 2-和-N(CH 3)SO 2-。
在本发明的一些优选的实施方案中,W 1为取代或未取代的下列基团:C 1-6烷基、C 6-10芳基、5-10元杂芳基、C 3-7环烷基或3-7元杂环烷基,所述取代是被1或2个选自氧代、羟基、氨基、羟甲基、 氨基甲基、卤素、氰基、C 1-6烷基-C(=O)O-和C 1-6烷基的取代基所取代。
在本发明的一些更优选的实施方案中,W 1为未取代或被1或2个选自羟基、氨基、羟甲基、氨基甲基和C 1-6烷基-C(=O)O-的取代基所取代的C 1-6烷基、C 3-7环烷基或3-7元杂环烷基。
在本发明的一些更优选的实施方案中,W 1为未取代或被1或2个选自羟基、氨基、羟甲基、氨基甲基和乙酰氧基的取代基所取代的下列基团:甲基、乙基、正丙基、异丙基、环丙基、环丁基、环戊基、环己基、吖丙啶基、吖丁啶基、四氢吡咯基、氧代吡咯烷基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、氧杂环丁基、四氢呋喃基、四氢噻吩基、四氢吡喃基、1,1-二氧硫代吗啉基、2-氮杂螺[3.3]庚基、1,1-二氧化异噻唑烷基或1,1-二氧化-1,2-噻嗪烷基,优选为甲基、氧代吡咯烷基、2-氮杂螺[3.3]庚基、1,1-二氧化异噻唑烷基、1,1-二氧化-1,2-噻嗪烷基或被选自羟基、羟甲基和乙酰氧基的取代基所取代的乙基、吖丁啶基、环丁基、四氢吡咯基或哌啶基。
在本发明的一些优选的实施方案中,在式I’化合物中,
n为1;
R 1为氟;
R 2为氰基;
R 3为甲基或乙基;
环B为下列结构的任一种:
Figure PCTCN2021105180-appb-000015
R 4为乙基;
X 1、X 2、X 3和X 4各自独立地为N或CH;
环A选自下列结构的任一种:
Figure PCTCN2021105180-appb-000016
环A未取代或被1个氟或甲基所取代;
R 6为-L 1-L 2-W 1
L 1选自化学键、-CH 2-、-C(CH 3) 2-和-C(=O)CH 2-;
L 2选自化学键、-C(=O)-、-NHCH 2-、-C(=O)NH-、-NHSO 2-和-N(CH 3)SO 2-;
W 1为下列基团中的任一种:
CH 3-、
Figure PCTCN2021105180-appb-000017
Figure PCTCN2021105180-appb-000018
在本发明的一些优选的实施方案中,上述式I’化合物为具备式I结构的化合物,
Figure PCTCN2021105180-appb-000019
其中,n、R 1、R 2、R 3、R 4、R 6、X 1、X 2、X 3和环A如式I’中所定义。
在本发明的一些更优选的实施方案中,上述式I’化合物为具备式I-1结构的化合物,
Figure PCTCN2021105180-appb-000020
其中,n、R 1、R 2、R 3、R 4、R 6和环A如式I’中所定义。
在本发明的一些优选的实施方案中,上述式I’化合物为具备式II结构的化合物,
Figure PCTCN2021105180-appb-000021
其中,n、R 1、R 2、R 3、R 4、R 6、X 1、X 2、X 3和环A如式I’中所定义。
在本发明的一些更优选的实施方案中,上述式I’化合物为具备式II-1结构的化合物,
Figure PCTCN2021105180-appb-000022
其中,n、R 1、R 2、R 3、R 4、R 6和环A如式I’中所定义。
在本发明的一些优选的实施方案中,上述式I’化合物为具备式III结构的化合物,
Figure PCTCN2021105180-appb-000023
其中,n、R 1、R 2、R 3、R 4、R 6、X 1、X 2、X 3和环A如式I’中所定义。
在本发明的一些更优选的实施方案中,上述式I’化合物为具备式III-1结构的化合物,
Figure PCTCN2021105180-appb-000024
其中,n、R 1、R 2、R 3、R 4、R 6和环A如式I’中所定义。
另外,本发明还提供了下列化合物:
Figure PCTCN2021105180-appb-000025
Figure PCTCN2021105180-appb-000026
Figure PCTCN2021105180-appb-000027
Figure PCTCN2021105180-appb-000028
[制备方法]
本发明还提供了上述式I化合物的一种典型的合成方法,以进一步描述本发明的技术方案,所述合成方法包括以下步骤:
步骤1:化合物1与相应醛和1,1,3,3-四甲基丁基异腈在氯化镁催化下反应生成中间体2;
步骤2:中间体2在甲酸中加热去保护得中间体3;
步骤3:任选的,中间体3与相应卤代烃经亲核取代反应得中间体4;
步骤4:中间体4通过水解得中间体5;
步骤5:中间体5与中间体6在碱性条件下发生亲核取代反应得中间体7;
其中,中间体6由化合物11通过两步反应制得:先由化合物11与硫脲通过关环得中间体12;中间体12在亚硝酸叔丁酯和氯化铜的作用下得中间体6;
步骤6:中间体7与中间体10通过Suzuki偶联反应得中间体8;
作为另一种可以替换的技术方案,中间体8也可通过中间体7先生成硼酸试剂13,再由中间体13与中间体14(X可为Cl、Br、I)经过Suzuki反应制得;
步骤7:中间体8通过水解得中间体9;
步骤8:中间体9与胺进行缩合反应得式I化合物,或式I的带叔丁氧羰基保护的产物,其进一步在三氟乙酸条件下脱保护可得式I化合物。
所述合成路线如下所示:
Figure PCTCN2021105180-appb-000029
其中,R 1-R 6,X 1-X 3,L 1,n,A的定义与上述相同,R 9为甲基或乙基。
本发明还提供了上述式II化合物(其中X 1为N)的一种典型的合成方法,以进一步描述本发明的技术方案,所述合成方法包括以下步骤:
步骤1:化合物1与双氧水在三氟乙酸溶液中反应生成中间体2;
步骤2:中间体2与三甲基氰硅烷在碱性条件下反应得中间体3;
步骤3:任选的,中间体3与相应的取代的肼在碱性条件下关环得中间体4;
步骤4:中间体4与中间体12在碱性条件下发生亲核取代反应得中间体5;
其中,中间体12由化合物10通过两步反应制得:先由化合物10与硫脲通过关环得中间体11;中间体11在亚硝酸叔丁酯和氯化铜的作用下得中间体12;
步骤5:中间体5与相应的卤代烃发生亲核取代反应得中间体6;
步骤6:中间体6与中间体9通过Suzuki偶联反应得式II化合物,或式II的带叔丁氧羰基保护的产物,其进一步在三氟乙酸条件下脱保护可得式II化合物;
其中,中间体9由化合物7通过两步反应制得:先由化合物7与相应的胺通过亲核取代反应得中间体8;中间体8在催化剂下生成硼酸试剂得中间体9。
所述合成路线如下所示:
Figure PCTCN2021105180-appb-000030
其中,R 1-R 6,X 2-X 3,n,A的定义与上述相同。
本发明还提供了上述式III化合物(其中X 1为N)的一种典型的合成方法,以进一步描述本发明的技术方案,所述合成方法包括以下步骤:
步骤1:化合物1与化合物2在碱性条件下关环生成中间体3;
步骤2:中间体3发生硝化反应得中间体4;
步骤3:中间体4在三氯氧磷条件下发生氯代反应得中间体5;
步骤4:中间体5发生还原反应得中间体6;
步骤5:中间体6与中间体14在碱性条件下发生亲核取代反应得中间体7;
其中,中间体14由化合物12通过两步反应制得:先由化合物12与硫脲通过关环得中间体13;中间体13在亚硝酸叔丁酯和氯化铜的作用下得中间体14;
步骤6:中间体7与相应的卤代烃发生亲核取代反应得中间体8;
步骤7:中间体8与中间体11通过Suzuki偶联反应得式III化合物,或式III的带叔丁氧羰基保护的产物,其进一步在三氟乙酸条件下脱保护可得式I化合物;
其中,中间体11由化合物9通过两步反应制得:先由化合物9与相应的胺通过亲核取代反应得中间体10;中间体10在催化剂下生成硼酸试剂得中间体11。
所述合成路线如下所示:
Figure PCTCN2021105180-appb-000031
其中,R 1-R 6,X 2-X 3,n,A的定义与上述相同。
[药物组合物]
术语“药物组合物”是指可以用作药物的组合物,其包含药物活性成分(API)以及可选的一种或多种药学上可接受载体。术语“药学上可接受的载体”是指与药物活性成分相容并且对受试者无害的药用辅料,包括(但不限于)稀释剂(或称填充剂)、粘合剂、崩解剂、润滑剂、润湿剂、增稠剂、助流剂、矫味剂、矫嗅剂、防腐剂、抗氧化剂、pH调节剂、溶剂、助溶剂、表面活性剂等。
本发明提供了一种药物组合物,其包含上述具备式I’、式I、式I-1、式II、式II-1、式III或式III-1结构的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物。
在本发明的一些实施方案中,上述药物组合物还包含药学上可接受的载体。
[药物制剂]
本发明还提供了一种药物制剂,其包含上述具有式I’、式I、式I-1、式II、式II-1、式III或式III-1结构的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物或者上述的药物组合物,所述制剂为片剂、胶囊剂、注射剂、颗粒剂、粉剂、栓剂、丸剂、乳膏剂、糊剂、凝胶剂、散剂、口服溶液、吸入剂、混悬剂、干悬剂、贴剂、洗剂中的任一种。
[医药用途]
无论是上述具有式I’、式I、式I-1、式II、式II-1、式III或式III-1结构的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,还是上述药物组合物或者药物制剂,都能够用作预防和治疗具有ATX表达增加的病理学特征的相关疾病;优选地,所述具有ATX表达增加的病理学特征的相关疾病包括:癌症、纤维化疾病、代谢疾病、骨髓增生异常综合征、呼吸系统疾病、心血管疾病、自身免疫性疾病、炎症、皮肤学疾病、神经系统疾病或疼痛;更优选地,所述具有ATX表达增加的病理学特征的相关疾病为肺纤维化、肾纤维化或肝纤维化。
另外,本发明还提供了上述具有式I’、式I、式I-1、式II、式II-1、式III或式III-1结构的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,以及上述的药物组合物或者药物制剂在制备用于预防和/或治疗具有ATX表达增加的病理学特征的相关疾病的药物中的应用,优选的,所述具有ATX表达增加的病理学特征的相关疾病包括:癌症、纤维化疾病、代谢疾病、骨髓增生异常综合征、呼吸系统疾病、心血管疾病、自身免疫性疾病、炎症、皮肤学疾病、神经系统疾病或疼痛,更优选的,所述具有ATX表达增加的病理学特征的相关疾病为肺纤维化、肾纤维化或肝纤维化。
[治疗方法]
本发明提供了一种预防和/或治疗具有ATX表达增加的病理学特征的相关疾病的方法,其包括下列步骤:将有效量的上述具有式I’、式I、式I-1、式II、式II-1、式III或式III-1结构的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,或者上述药物组合物或者药物制剂施用于对其有需求的患者。
术语“治疗有效量”是指能够诱发细胞、组织、器官或生物体(例如患者)产生生物或医学反应的药物活性成分的剂量。
术语“施用”是指将药物活性成分(比如本发明的化合物)或包含药物活性成分的药物组合物(例如本发明的药物组合物)应用于患者或其细胞、组织、器官、生物流体等部位,以便使药物活性成分或药物组合物与患者或其细胞、组织、器官、生物流体等部位接触的过程。常见的施用方式包括(但不限于)口服施用、皮下施用、肌内施用、腹膜下施用、眼部施用、鼻部施用、舌下施用、直肠施用、阴道施用等。
术语“对其有需求”是指医生或其他护理人员对患者需要或者将要从预防和/或治疗过程中获益的判断,该判断的得出基于医生或其他护理人员在其专长领域中的各种因素。
术语“患者”(或称受试者)是指人类或非人类的动物(例如哺乳动物)。
下面的实施例可以对本发明做进一步的描述,然而,这些实施例不应作为对本发明的范围的限制。
本发明中使用的缩写如下:
AcOH:乙酸;HCOOH:甲酸;I 2:碘;t-BuONO:亚硝酸叔丁酯;CuCl 2:氯化铜;n-BuOH:正丁醇;MgCl 2:氯化镁;NaH:氢化钠;CH 3I:碘甲烷;Pd(dppf)Cl 2:[1,1'-双(二苯基膦)二茂铁]二氯化钯;Pd(PPh 3) 4:四(三苯基膦)钯;KOAc:醋酸钾;B 2(Pin) 2:联硼酸频那醇酯;CDCl 3:氘代氯仿;dioxane:1,4-二氧六环;CO 2:二氧化碳;conc.H 2SO 4:浓硫酸;DCM:二氯甲烷;DIEA:N,N-二异丙基乙胺;DMF:N,N-二甲基甲酰胺;DMSO:二甲亚砜;DMSO-d 6:氘代二甲亚砜;EtOH:乙醇; g:克;HATU:2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯;Hz:赫兹;h:小时;IC 50:半数最大抑制浓度;LiOH:氢氧化锂;MeOH:甲醇;mg:毫克;mL:毫升;mmol:毫摩尔;MHz:兆赫兹;NaOH:氢氧化钠;NMR:核磁共振;M:摩尔/升;PBS:磷酸缓冲盐溶液;TLC:薄层色谱;μM:微摩尔/升;μg:微克;μL:微升;δ:化学位移。
以下描述本发明实施例中通用试验条件:
首先,实施例中的反应一般在氮气保护下进行。
进一步地,中间体和最终产物通过色谱柱、制备色谱板和ICSO快速制备色谱系统分离纯化。
进一步地,LC-MS液质联用色谱仪使用Waters公司ACQUITY Arc配备QDa Detector。质谱(MS)采用ESI源,仅指示母体分子的分子量M,通常汇报[M+H] +。注射体积是通过样品浓度来确定;流速为:0.8mL/min;HPLC的峰值是通过在220nm和254nm处的UV-Vis波长来记录读取的。流动相为0.01%甲酸的超纯水溶液(流动相A)和0.01%甲酸的乙腈溶液(流动相B)。梯度洗脱条件如下表1和表2所示:
表1:梯度洗脱条件1
时间(min) A(H 2O,0.01%HCOOH) B(CH 3CN,0.01%HCOOH)
0.0-0.3 95-85 5-15
0.3-3.2 85-20 15-80
3.2-3.8 20-5 80-95
3.8-3.81 5-95 95-5
3.81-4.0 95 5
表2:梯度洗脱条件2
时间(min) A(H 2O,0.01%HCOOH) B(CH 3CN,0.01%HCOOH)
0.00-5.90 95-5 5-95
5.90-5.91 5-95 95-5
5.91-6.00 95 5
进一步地,NMR谱图采用Varian 400MHz核磁共振谱仪获得数据,常以CDCl 3,DMSO-d 6作为溶剂,以ppm报告化学位移。各种峰的描述如下:s(单峰),d(双峰),t(三重峰),q(四重峰),m(多重峰),dd(双二重峰)。偶合常数使用Hz表示。
实施例1:2-((2-乙基-6-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)苯基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物1)
Figure PCTCN2021105180-appb-000032
步骤1a:制备2-氨基-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000033
步骤1a的具体方式:室温下,在3-(4-氟苯基)-3-氧代丙腈(6.0g,36.78mmol)的乙醇(72mL) 溶液中加入吡啶(2.97mL,36.78mmol)。将混合物在70℃下搅拌15分钟后冷却至室温。然后缓慢滴加硫脲(5.61g,73.56mmol)和碘(9.33g,6.78mmol)的EtOH(36mL)溶液。室温搅拌一小时后,加入1M的Na 2S 2O 3(36mL)淬灭反应。过滤,用水洗涤滤饼,干燥得白色固体2-氨基-4-(4-氟苯基)噻唑-5-甲腈(4.2g)。
步骤1b:制备2-氯-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000034
步骤1b的具体方式:室温下,向氯化铜(3.09g,23.0mmol)的乙腈溶液(42mL)加入亚硝酸叔丁酯(3.57g,34.6mmol)。将混合物在室温下搅30分钟,然后加入2-氨基-4-(4-氟苯基)噻唑-5-甲腈(4.2g,19.15mmol),室温搅拌1小时。加入1M HCl(63mL)淬灭反应,用乙酸乙酯萃取(100mL×2)。合并有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗产品。粗产品经过快速柱层析色谱纯化得白色固体2-氯-4-(4-氟苯基)噻唑-5-甲腈(4.8g)。
步骤1c:制备6-溴-2-乙基-N-(2,4,4-三甲基戊-2-基)咪唑并[1,2-a]吡啶-3-胺
Figure PCTCN2021105180-appb-000035
步骤1c的具体方式:将5-溴吡啶-2-胺(4.0g,23.1mmol)、丙醛(4.4mL,25.4mmol)、1,1,3,3-四甲基丁基异氰(4.0mL,57.8mmol)和氯化镁(220mg,2.31mmol)溶于正丁醇(40mL)中,加热至130℃搅拌反应3小时。冷却后,将反应液浓缩,加水稀释(30mL)。水相用乙酸乙酯萃取(80mL×3),将合并的有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤浓缩得棕色固体6-溴-2-乙基-N-(2,4,4-三甲基戊烷-2-基)咪唑并[1,2-a]吡啶-3-胺(8.8g)。
步骤1d:制备N-(6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)甲酰胺
Figure PCTCN2021105180-appb-000036
步骤1d的具体方式:将6-溴-2-乙基-N-(2,4,4-三甲基戊烷-2-基)咪唑并[1,2-a]吡啶-3-胺(8.1g,23.0mmol)溶于甲酸(30mL),回流2小时。待反应液冷却,旋干,加入甲基叔丁基醚(30mL)。搅拌15分钟后过滤,滤饼用甲基叔丁基醚(10mL)洗涤,真空干燥得灰白色固体N-(6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)甲酰胺(5.45g)。
步骤1e:制备N-(6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)-N-甲基甲酰胺
Figure PCTCN2021105180-appb-000037
步骤1e的具体方式:零度下,氮气氛围中向N-(6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)甲酰胺(6g,22.4mmol)的DMF溶液(50mL)分批加入60%氢化钠(1.3g,33.6mmol)。反应液在室温下搅拌1小时后加入碘甲烷(4.8g,33.6mmol)。混合物在室温下继续反应3小时后加水(50mL)淬灭,用乙酸乙酯萃取(100mL×3),合并的有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤浓缩减压浓缩得到粗产品。粗产品通过快速柱层析色谱纯化得灰色固体N-(6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)- N-甲基甲酰胺(5g)。
步骤1f:制备6-溴-2-乙基-N-甲基咪唑并[1,2-a]吡啶-3-胺
Figure PCTCN2021105180-appb-000038
步骤1f的具体方式:向N-(6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)-N-甲基甲酰胺(4.5g,16.0mmol)的甲醇溶液(25mL)加入10M的氢氧化钠溶液(24mL,240mmol)。反应液在室温下搅拌3小时后浓缩,加入乙酸乙酯(100mL)稀释,有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得灰白色固体6-溴-2-乙基-N-甲基咪唑并[1,2-a]吡啶-3-胺(4.1g)。
步骤1g:制备2-((6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000039
步骤1g的具体方式:零度下,氮气氛围中向6-溴-2-乙基-N-甲基咪唑并[1,2-a]吡啶-3-胺(2.5g,9.84mmol)的DMF溶液(30mL)溶液加入60%氢化钠(0.79g,19.67mmol)。将混合物在室温下搅拌30分钟,然后缓慢加入2-氯-4-(4-氟苯基)噻唑-5-甲腈(4.8g,20.11mmol)的DMF溶液(10mL)。混合物在80℃下搅拌4小时后用饱和氯化铵水溶液淬灭,并用乙酸乙酯(80mL×3)萃取。将合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗产品。粗产品经过快速柱层析色谱纯化得黄色固体2-((6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-腈(2.6g)。
步骤1h:制备2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)苯基)乙酸甲酯
Figure PCTCN2021105180-appb-000040
步骤1h的具体方式:氮气氛围中,向2-((6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(100mg,0.219mmol)和2-(4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯基)乙酸甲酯(79mg,0.285mmol)的DMF溶液(5mL)加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(18mg,0.022mmol)和磷酸钾(93mg,0.438mmol)。混合物在85℃搅拌4小时后加水(10mL)稀释,并用乙酸乙酯萃取(15mL×3)。将合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗产品。粗产品经过快速柱层析色谱纯化得棕色固体2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)苯基)乙酸甲酯(68mg)。
步骤1i:制备2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)苯基)乙酸
Figure PCTCN2021105180-appb-000041
步骤1i的具体方式:向2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)苯基)乙酸甲酯(68mg,0.129mmol)的四氢呋喃(3mL)和水(1mL)溶液加入氢氧化钠(16mg,0.387mmol)。混合物在室温搅拌4小时后用1M盐酸溶液调节pH至3~4,用乙酸乙酯(10mL×3)萃取。将合并的有机相用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得棕色固体2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)苯基)乙酸(55mg)。
步骤1j:制备2-((2-乙基-6-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)苯基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000042
步骤1j的具体方式:向2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)苯基)乙酸(55mg,0.11mmol)和氮杂环丁烷-3-醇(24mg,0.32mmol)的DMF溶液(2mL)加入DIEA(42mg,0.324mmol)和HATU(62mg,0.162mmol)。混合物在室温下搅拌2小时后加水(10mL)稀释,并用乙酸乙酯萃取(10mL×3)。将合并的有机相用饱和食盐水洗涤(10mL),无水硫酸钠干燥,过滤,减压浓缩得到粗产品。粗产品经过快速柱层析色谱纯化得白色固体2-((2-乙基-6-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)苯基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(25.4mg)。
1H NMR(400MHz,DMSO-d 6):δ8.72(br.s,1H),8.09-8.06(m,2H),7.89(d,J=8.8Hz,1H),7.80(d,J=9.2Hz,1H),7.72(d,J=8.0Hz,2H),7.42(d,J=8.8Hz,2H),7.34(d,J=8.0Hz,2H),4.46-4.41(m,1H),4.36(d,J=7.8Hz,1H),4.04-3.99(m,1H),3.91-3.88(m,1H),3.65(s,3H),3.58-3.54(m,1H),3.47(s,2H),2.75-2.70(q,J=7.6Hz,2H),1.38(t,J=7.6Hz,3H)。
MS实测值(ESI +)[(M+H) +]:567。
实施例2:2-((2-乙基-6-(6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物2)
Figure PCTCN2021105180-appb-000043
步骤2a:制备2-(5-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)吡啶-2-基)乙酸乙酯
Figure PCTCN2021105180-appb-000044
步骤2a的具体方式:氮气保护下,向2-(5-溴吡啶-2-基)乙酸乙酯(200mg,0.82mmol),联硼酸频那醇酯(229mg,0.90mmol),醋酸钾(120mg,1.23mmol)的1,4-二氧六环溶液(2mL)加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(58mg,0.08mmol)。将混合物加热到85℃搅拌2小时后,加水(15mL)稀释,用乙酸乙酯(20mL×2)萃取。将合并的有机相用饱和食盐水(15mL)洗涤,无水硫酸钠干燥,过滤,浓缩得到棕色油状物2-(5-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)吡啶-2-基)乙酸乙酯(171mg)。
步骤2b:制备2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡啶-2-基)乙酸乙酯
Figure PCTCN2021105180-appb-000045
步骤2b的具体方式:将2-((6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(100mg,0.22mmol)和2-(5-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)吡啶-2-基)乙酸乙酯(55mg,0.26mmol)按照实施例1步骤1h的方法制备得棕色固体2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡啶-2-基)乙酸乙酯(60mg)。
步骤2c:制备2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡啶-2-基)乙酸
Figure PCTCN2021105180-appb-000046
步骤2c的具体方式:将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡啶-2-基)乙酸乙酯(50mg,0.09mmol)和氢氧化钠(11mg,0.27mmol)按照实施例1步骤1i的方法制备得到棕色固体2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡啶-2-基)乙酸(45mg)。
步骤2d:制备2-((2-乙基-6-(6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000047
步骤2d的具体方式:将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a] 吡啶-6-基)吡啶-2-基)乙酸(45mg,0.09mmol)和氮杂环丁烷-3-醇(20mg,0.27mmol)按照实施例1步骤1j的方法制备得到白色固体(2-((2-乙基-6-(6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(28.9mg)。
1H NMR(400MHz,CDCl 3):δ8.68(d,J=2.0Hz,1H),8.15(dd,J=8.8,5.4Hz,2H),7.92(s,1H),7.81(dd,J=8.1,2.2Hz,1H),7.74(d,J=9.4Hz,1H),7.51(dd,J=9.4,1.4Hz,1H),7.46(d,J=8.2Hz,1H),7.20–7.15(m,2H),4.71-4.65(m,1H),4.54-4.48(m,1H),4.29-4.24(m,1H),4.16-4.13(m,1H),3.91–3.88(m,1H),3.70(s,2H),3.68(s,3H),3.17(br.s,1H),2.80(q,J=7.4Hz,2H),1.40(t,J=7.6Hz,3H)。
MS实测值(ESI +)[(M+H) +]:568。
实施例3:2-((2-乙基-6-(5-氟-6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物3)
Figure PCTCN2021105180-appb-000048
步骤3a:制备1-(叔丁基)3-乙基2-(5-溴-3-氟吡啶-2-基)丙二酸酯
Figure PCTCN2021105180-appb-000049
步骤3a的具体方式:在零度下氮气氛围中,将60%氢化钠(893mg,22.3mmol)分批加入丙二酸叔丁基乙酯(3.5g,18.6mmol)的干燥DMF溶液(30mL)。混合物继续在零度下搅拌半小时后,加入5-溴-2,3-二氟吡啶(3g,15.5mmol),然后将反应液在80℃下搅拌3小时。反应加水(35mL)淬灭,用乙酸乙酯(30mL×3)萃取,然后将合并的有机相用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得棕色油状物1-(叔丁基)3-乙基2-(5-溴-3-氟吡啶-2-基)丙二酸酯(3g)。
步骤3b:制备2-(5-溴-3-氟吡啶-2-基)乙酸乙酯
Figure PCTCN2021105180-appb-000050
步骤3b的具体方式:零度下,向1-(叔丁基)3-乙基2-(5-溴-3-氟吡啶-2-基)丙二酸酯(3g,8.30mmol)的二氯甲烷溶液(5mL)加入三氟乙酸(5mL)。混合物室温下继续搅拌2小时,浓缩,加水稀释(15mL),用乙酸乙酯萃取(30mL×3)。将合并的有机相用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗产品。粗产品经过柱层析分离纯化得黄色油状物2-(5-溴-3-氟吡啶-2-基)乙酸乙酯(1.2g)。
步骤3c:制备2-(3-氟-5-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)吡啶-2-基)乙酸乙酯
Figure PCTCN2021105180-appb-000051
步骤3c的具体方式:氮气保护下,向2-(5-溴-3-氟吡啶-2-基)乙酸乙酯(100mg,0.38mmol),联 硼酸频那醇酯(115mg,0.46mmol),醋酸钾(56mg,0.57mmol)的1,4-二氧六环溶液(2mL)加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(29mg,0.04mmol)。将混合物加热到85℃搅拌3小时后,加水(15mL)稀释,用乙酸乙酯(20mL×2)萃取。将合并的有机相用饱和食盐水(15mL)洗涤,无水硫酸钠干燥,过滤,浓缩得到棕色固体2-(3-氟-5-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)吡啶-2-基)乙酸乙酯(100mg)。
步骤3d:制备2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-3-氟吡啶-2-基)乙酸乙酯
Figure PCTCN2021105180-appb-000052
步骤3d的具体方式:将2-((6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(100mg,0.22mmol)和2-(3-氟-5-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)吡啶-2-基)乙酸乙酯(100mg,0.33mmol)按照实施例1步骤1h的方法制备得棕色固体2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-3-氟吡啶-2-基)乙酸乙酯(50mg)。
步骤3e:制备2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-3-氟吡啶-2-基)乙酸
Figure PCTCN2021105180-appb-000053
步骤3e的具体方式:将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-3-氟吡啶-2-基)乙酸乙酯(50mg,0.09mmol)和氢氧化钠(11mg,0.27mmol)按照实施例1步骤1i的方法制备得棕色固体2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-3-氟吡啶-2-基)乙酸(45mg)。
步骤3f:制备2-((2-乙基-6-(5-氟-6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000054
步骤3f的具体方式:将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-3-氟吡啶-2-基)乙酸(45mg,0.08mmol)和氮杂环丁烷-3-醇(10mg,0.13mmol)按照实施例1步骤1j的方法制备得白色固体2-((2-乙基-6-(5-氟-6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-腈(18mg)。
1H NMR(400MHz,CDCl 3):δ8.54(s,1H),8.17-8.13(m,2H),7.92(s,1H),7.75(d,J=8.0Hz,1H),7.52(dd,J=8.0Hz,2.0Hz,2H),7.17(t,J=8.0Hz,2H),4.70(d,J=4.0Hz,1H),4.49(t,J=8.0Hz,1H),4.32-4.28(m,1H),4.14(dd,J=8.0,4.0Hz,1H),3.91(dd,J=8.0,4.0Hz,1H),3.76(s,2H),3.72(d, J=8.0Hz,1H),3.68(s,3H),2.80(q,J=8.0Hz,2H),1.39(t,J=8.0Hz,3H)。
MS实测值(ESI +)[(M+H) +]:586。
实施例4:2-((2-乙基-6-(5-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物4)
Figure PCTCN2021105180-appb-000055
步骤4a:制备1-(叔丁基)3-乙基2-(5-溴吡嗪-2-基)丙二酸酯
Figure PCTCN2021105180-appb-000056
步骤4a的具体方式:将2,5-二溴吡嗪(1g,4.20mmol)和丙二酸叔丁基乙酯(870mg,4.62mmol)按照实施例3步骤3a的方法制备得黄色固体1-(叔丁基)3-乙基2-(5-溴吡嗪-2-基)丙二酸酯(1.5g)。
步骤4b:制备2-(5-溴吡嗪-2-基)乙酸乙酯
Figure PCTCN2021105180-appb-000057
步骤4b的具体方式:将1-(叔丁基)3-乙基2-(5-溴吡嗪-2-基)丙二酸酯(1.5g,4.35mmol)按照实施例3步骤3b的方法制备得黄色油状物2-(5-溴吡嗪-2-基)乙酸乙酯(400mg)。
步骤4c:制备(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑[1,2-a]吡啶-6-基)硼酸
Figure PCTCN2021105180-appb-000058
步骤4c的具体方式:将2-((6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(100mg,0.22mmol)和联硼酸频那醇酯(67mg,0.26mmol)按照实施例3步骤3c的方法制备得棕色油状物(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑[1,2-a]吡啶-6-基)硼酸(100mg)。
步骤4d:制备2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)乙酸乙酯
Figure PCTCN2021105180-appb-000059
步骤4d的具体方式:将(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑[1,2-a]吡啶-6-基)硼酸(100mg,0.22mmol)和2-(5-溴吡嗪-2-基)乙酸乙酯(87mg,0.36mmol)按照实施例1步骤1h的方法制备得棕色固体2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡嗪-6-基)嘧啶-2-基)乙酸乙酯(50mg)。
步骤4e:制备2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)乙酸
Figure PCTCN2021105180-appb-000060
步骤4e的具体方式:将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡嗪-6-基)嘧啶-2-基)乙酸乙酯(50mg,0.09mmol)和氢氧化钠(11mg,0.27mmol)按照实施例1步骤1i的方法制备得棕色固体2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)乙酸(40mg)。
步骤4f:制备2-((2-乙基-6-(5-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000061
步骤4f的具体方式:将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)乙酸(40mg,0.08mmol)和氮杂环丁烷-3-醇(18mg,0.23mmol)按照实施例1步骤1j的方法制备得灰白色固体(2-((2-乙基-6-(5-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(7mg)。
1H NMR(400MHz,CDCl 3):δ8.91(s,1H),8.66(s,1H),8.56(s,1H),8.15(dd,J=8.8,5.2Hz,2H),7.94(d,J=9.6Hz,1H),7.86(d,J=8.8Hz,1H),7.18(t,J=8.6Hz,2H),4.75-4.71(m,1H),4.58-4.51(m,1H),4.34-4.27(m,1H),4.23-4.17(m,1H),3.95-3.89(m,1H),3.73(s,2H),3.70(s,3H),2.82(q,J=7.7Hz,2H),1.40(t,J=7.4Hz,3H)。
MS实测值(ESI +)[(M+H) +]:569。
实施例5:2-((2-乙基-6-(6-2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哒嗪-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物5)
Figure PCTCN2021105180-appb-000062
步骤5a:制备1-(叔丁基)3-乙基2-(6-氯哒嗪-3-基)丙二酸酯
Figure PCTCN2021105180-appb-000063
步骤5a的具体方式:将3,6-二氯哒嗪(1g,6.70mmol)和丙二酸叔丁基乙酯(1.3g,6.70mmol)按照实施例3步骤3a的方法制备得黄色液体1-(叔丁基)3-乙基2-(6-氯哒嗪-3-基)丙二酸酯(1.0g)。
步骤5b:制备2-(6-氯哒嗪-3-基)乙酸乙酯
Figure PCTCN2021105180-appb-000064
步骤5b的具体方式:将1-(叔丁基)3-乙基2-(6-氯哒嗪-3-基)丙二酸酯(1.0g,3.33mmol)按照实施例3步骤3b的方法制备得黄色油状物2-(6-氯哒嗪-3-基)乙酸乙酯(400mg)。
步骤5c:制备2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)哒嗪-3-基)乙酸乙酯
Figure PCTCN2021105180-appb-000065
步骤5c的具体方式:将2-((6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(100mg,0.22mmol)和2-(6-氯哒嗪-3-基)乙酸乙酯(60mg,0.30mmol)按照实施例1步骤1h的方法制备得棕色固体2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)哒嗪-3-基)乙酸乙酯(90mg)。
步骤5d:制备2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)哒嗪-3-基)乙酸
Figure PCTCN2021105180-appb-000066
步骤5d的具体方式:向2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)哒嗪-3-基)乙酸乙酯(90mg,0.166mmol)的四氢呋喃(1mL)和水(0.2mL)溶液加入一水合氢氧化锂(10mg,0.250mmol)。混合物在室温搅拌2小时后用油泵减压浓缩得棕色固体2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)哒嗪-3-基)乙酸(100mg)。
步骤5e:制备2-((2-乙基-6-(6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哒嗪-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000067
步骤5e的具体方式:将2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)哒嗪-3-基)乙酸(90mg,0.175mmol)和HATU(80mg,0.211mmol)的DMF溶液在0℃下搅拌0.5小时,随后向其中加入氮杂环丁烷-3-醇(18mg,0.23mmol)。混合物在室温下搅拌1小时后加水(10mL)稀释,并用乙酸乙酯(10mL×3)萃取。将合并的有机相用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗产品。粗产品经过快速柱层析色谱纯化得白色固体2-((2-乙基-6-(6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哒嗪-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(15.0mg)。
1H NMR(400MHz,CDCl 3):δ8.78(s,1H),8.16-8.12(m,2H),7.84-7.75(m,4H),7.18(t,J=8.0Hz,2H),4.69(br.s,1H),4.59(t,J=8.0Hz,1H),4.28-4.18(m,2H),3.92-3.91(m,1H),3.88(s,2H),3.69(s,3H),2.80(q,J=8.0Hz,2H),1.40(t,J=8.0Hz,3H)。
MS实测值(ESI +)[(M+H) +]:569。
实施例6:2-((2-乙基-6-(6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)-2-甲基吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物6)
Figure PCTCN2021105180-appb-000068
步骤6a:制备1-(叔丁基)3-乙基2-(5-溴-6-甲基吡啶-2-基)丙二酸酯
Figure PCTCN2021105180-appb-000069
步骤6a的具体方式:将3-溴-6-氟-2-甲基吡啶(0.5g,2.63mmol)和丙二酸叔丁基乙酯(1.5mL,7.89mmol)按照实施例3步骤3a的方法制备得黄色固体1-(叔丁基)3-乙基2-(5-溴-6-甲基吡啶-2-基)丙二酸酯(0.3g)。
步骤6b:制备2-(5-溴-6-甲基吡啶-2-基)乙酸乙酯
Figure PCTCN2021105180-appb-000070
步骤6b的具体方式:将1-(叔丁基)3-乙基2-(5-溴-6-甲基吡啶-2-基)丙二酸酯(0.3g,1.58mmol)按照实施例3步骤3b的方法制备得黄色油状物2-(5-溴-6-甲基吡啶-2-基)乙酸乙酯(100mg)。
步骤6c:制备(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑[1,2-a]吡啶-6-基)硼酸
Figure PCTCN2021105180-appb-000071
步骤6c的具体方式:将2-((6-溴-2-乙基咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(200mg,0.44mmol)和联硼酸频那醇酯(134mg,0.52mmol)按照实施例3步骤3c的方法制 备得棕色油状物(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑[1,2-a]吡啶-6-基)硼酸(200mg)。
步骤6d:制备2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-6-甲基吡啶-2-基)乙酸乙酯
Figure PCTCN2021105180-appb-000072
步骤6d的具体方式:将(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑[1,2-a]吡啶-6-基)硼酸(90mg,0.21mmol)和2-(5-溴-6-甲基吡啶-2-基)乙酸乙酯(66mg,0.26mmol)按照实施例1步骤1h的方法制备得棕色固体2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-6-甲基吡啶-2-基)乙酸乙酯(27mg)。
步骤6e:制备2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-6-甲基吡啶-2-基)乙酸
Figure PCTCN2021105180-appb-000073
步骤6e的具体方式:将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-6-甲基吡啶-2-基)乙酸乙酯(27mg,0.05mmol)和氢氧化钠(8.0mg,0.20mmol)按照实施例1步骤1i的方法制备得棕色固体2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-6-甲基吡啶-2-基)乙酸(25.6mg)。
步骤6f:制备2-((2-乙基-6-(6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)-2-甲基吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000074
步骤6f的具体方式:将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-6-甲基吡啶-2-基)乙酸(25.6mg,0.05mmol)和氮杂环丁烷-3-醇(7mg,0.1mmol)按照实施例1步骤1j的方法制备得灰白色固体2-((2-乙基-6-(6-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)-2-甲基吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(6.2mg)。
1H NMR(400MHz,CDCl 3):δ8.12(dd,J=8.8,5.4Hz,2H),7.73-7.64(m,2H),7.52-7.47(m,1H),7.32-7.26(m,2H),7.17-7.13(m,2H),4.70-4.64(m,1H),4.56-4.49(m,1H),4.28-4.24(m,1H),4.17-4.13(m,1H),3.90-3.86(m,1H),3.66(s,2H),3.64(s,3H),2.79(q,J=7.6Hz,2H),2.46(s,3H),1.39(t,J=7.60Hz,3H)。
MS实测值(ESI +)[(M+H) +]:582。
实施例7:2-((2-乙基-6-(5-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-2-基)咪唑并[1,2-a]吡啶- 3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物7)
Figure PCTCN2021105180-appb-000075
步骤7a:制备2-溴-5-(溴甲基)吡啶
Figure PCTCN2021105180-appb-000076
步骤7a的具体方式:将2-溴-5-甲基吡啶(0.5g,2.91mmol)溶于5mL四氯化碳中,然后加入偶氮二异丁腈(48mg,0.29mmol)和NBS(0.67g,3.78mmol),氮气保护下,加热到70℃反应3小时。反应液浓缩,柱层析纯化方法制备得无色油状物2-溴-5-(溴甲基)吡啶(0.4g)。
步骤7b:制备2-(6-溴吡啶-3-基)乙腈
Figure PCTCN2021105180-appb-000077
步骤7b的具体方式:将2-溴-5-(溴甲基)吡啶(300mg,1.2mmol)溶于5mL乙腈中,加入四丁基氰化铵(320mg,1.2mmol),室温反应2小时。将反应液浓缩,加入20mL水,乙酸乙酯萃取(50mL×3),无水硫酸钠干燥,过滤,浓缩。将浓缩的反应液柱层析纯化得无色油状物2-(6-溴吡啶-3-基)乙腈(200mg)。
步骤7c:制备2-(6-溴吡啶-3-基)乙酸甲酯
Figure PCTCN2021105180-appb-000078
步骤7c的具体方式:将2-(6-溴吡啶-3-基)乙腈(200mg,1.02mmol)溶于3mL甲醇中,氮气保护下加入二氯亚砜(0.4mL,5.10mmol),室温反应5小时。加入20mL水,乙酸乙酯萃取(50mL),无水硫酸钠干燥,过滤,浓缩。将浓缩的反应液柱层析纯化得无色油状物2-(6-溴吡啶-3-基)乙酸甲酯(200mg)。
步骤7d:制备2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡啶-3-基)乙酸甲酯
Figure PCTCN2021105180-appb-000079
步骤7d的具体方式:将(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)硼酸(150mg,0.36mmol)和2-(6-溴吡啶-3-基)乙酸甲酯(98mg,0.43mmol)按照实施例1步骤1h的方法制备得棕色固体2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡啶-3-基)乙酸甲酯(70m)。
步骤7e:制备2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡啶-3-基)乙酸
Figure PCTCN2021105180-appb-000080
步骤7e的具体方式:将2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡啶-3-基乙酸甲酯(70mg,0.13mmol)与氢氧化钠(16mg,0.40mmol)按照实施例1步骤1i的方法制备得棕色固体2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡啶-3-基)乙酸(68mg)。
步骤7f:制备2-((2-乙基-6-(5-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000081
步骤7f的具体方式:将2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡啶-3-基)乙酸(66mg,0.13mmol)和氮杂环丁烷-3-醇(14mg,0.2mmol)按照实施例1步骤1j的方法制备得灰白色固体2-((2-乙基-6-(5-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(6.3mg)。
1H NMR(400MHz,CDCl 3):δ8.53(d,J=8.5Hz,2H),8.17(dd,J=8.5,5.4Hz,2H),7.87(d,J=8.8Hz,1H),7.78(d,J=7.7Hz,1H),7.69(dd,J=19.1,8.8Hz,2H),7.19(t,J=8.6Hz,2H),4.75-4.61(m,1H),4.44-4.39(m,1H),4.31-4.26(m,1H),4.08-4.06(m,1H),3.92-3.88(m,1H),3.70(s,3H),3.50(s,2H),2.79(q,J=7.6Hz,2H),1.40(t,J=7.2Hz,3H)。
MS实测值(ESI +)[(M+H) +]:568。
实施例8:2-((2-乙基-6-(5-(3-羟基氮杂环丁烷-1-羰基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物8)
Figure PCTCN2021105180-appb-000082
步骤8a:制备甲基5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-羧酸甲酯
Figure PCTCN2021105180-appb-000083
步骤8a的具体方式:将3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)硼酸(210mg,0.50mmol)和5-溴吡嗪-2-羧酸甲酯(216mg,1.0mmol)按照实施例1步骤1h的方法制备得棕色固体甲基5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-羧酸甲酯(143mg)。
步骤8b:制备5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-羧酸
Figure PCTCN2021105180-appb-000084
步骤8b的具体方式:室温下,将甲基5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-羧酸甲酯(143mg,0.28mmol)与氢氧化钠(24mg,0.60mmol)按照实施例1步骤1i的方法制备得棕色固体5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-羧酸(139mg)。
步骤8c:制备2-((2-乙基-6-(5-(3-羟基氮杂环丁烷-1-羰基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000085
步骤8c的具体方式:室温下,将5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-羧酸(46mg,0.092mmol)与氮杂环丁烷-3-醇(14mg,0.2mmol)按照实施例1步骤1j的方法制备得白色固体2-((2-乙基-6-(5-(3-羟基氮杂环丁烷-1-羰基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(14.2mg)。
1H NMR(400MHz,CDCl 3):δ9.34(s,1H),8.97(s,1H),8.66(s,1H),8.17(dd,J=8.8,5.6Hz,2H),7.95(d,J=9.6Hz,1H),7.79(d,J=9.6Hz,1H),7.20(t,J=8.8Hz,2H),4.99-4.94(m,1H),4.80-4.79(m,1H),4.58-4.51(m,2H),4.13(dd,J=12.0,3.2Hz,1H),3.73(s,3H),2.82(q,J=7.6Hz,2H),1.42(t,J=7.6Hz,3H)。
MS实测值(ESI +)[(M+H) +]:555。
实施例9:2-((2-乙基-6-(5-(1-(3-羟基氮杂环丁烷-1-基)-2-甲基-1-氧代丙-2-基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物9)
Figure PCTCN2021105180-appb-000086
步骤9a:制备2-(5-溴吡嗪-2-基)-2-甲基丙酸乙酯
Figure PCTCN2021105180-appb-000087
步骤9a的具体方式:在零度下氮气氛围中,将60%氢化钠(69mg,1.73mmol)分批加入2-(5-溴吡嗪-2-基)乙酸乙酯(200mg,0.82mmol)的干燥DMF溶液(5mL)。混合物继续在零度下搅拌半小时后,加入碘甲烷(244mg,1.73mmol),然后将反应液在室温下搅拌2小时。反应加水(15mL)淬灭,用乙酸乙酯(20mL×3)萃取,然后将合并的有机相用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得粗产品。粗产品通过快速柱层析色谱纯化得黄色油状液体2-(5-溴吡嗪-2-基)-2-甲基丙酸乙酯(120mg)。
步骤9b:制备2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)-2-甲基丙酸乙酯
Figure PCTCN2021105180-appb-000088
步骤9b的具体方式:将(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)硼酸(100mg,0.24mmol)和2-(5-溴吡嗪-2-基)-2-甲基丙酸乙酯(97mg,0.36mmol)按照实施例1步骤1h的方法制备得棕色固体2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)-2-甲基丙酸乙酯(100mg)。
步骤9c:制备2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)-2-甲基丙酸
Figure PCTCN2021105180-appb-000089
步骤9c的具体方式:室温下,将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)-2-甲基丙酸乙酯(100mg,0.18mmol)与一水合氢氧化锂(15mg,0.35mmol)按照实施例5步骤5d的方法制备得棕色固体2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)-2-甲基丙酸(110mg)。
步骤9d:制备2-((2-乙基-6-(5-(1-(3-羟基氮杂环丁烷-1-基)-2-甲基-1-氧代丙烷-2-基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000090
步骤9d的具体方式:室温下,将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)-2-甲基丙酸(100mg,0.18mmol)与氮杂环丁烷-3-醇(27mg,0.37mmol)按照实施例5步骤5e的方法制备得白色固体2-((2-乙基-6-(5-(1-(3-羟基氮杂环丁烷-1-基)-2-甲基-1-氧 代丙烷-2-基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(45mg)。
1H NMR(400MHz,CDCl 3):δ8.95(s,1H),8.63(s,1H),8.58(s,1H),8.16(dd,J=8.8,5.6Hz,2H),7.90(d,J=9.6Hz,1H),7.76(d,J=9.2Hz,1H),7.20(t,J=8.8Hz,2H),4.50-4.48(m,1H),4.24-4.19(m,1H),3.91-3.80(m,2H),3.70(s,3H),3.40-3.30(m,1H),2.81(q,J=7.6Hz,2H),1.63(s,6H),1.42(t,J=7.6Hz,3H)。
MS实测值(ESI +)[(M+H) +]:597。
实施例10:2-((2-乙基-6-(3-氟-5-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物10)
Figure PCTCN2021105180-appb-000091
步骤10a:制备5-(溴甲基)-2-氯-3-氟吡啶
Figure PCTCN2021105180-appb-000092
步骤10a的具体方式:将2-氯-3-氟-5-甲基吡啶(1.0g,6.90mmol)溶于10mL四氯化碳中,然后加入偶氮二异丁腈(113mg,0.69mmol)和NBS(1.35g,7.59mmol),氮气保护下,加热到70℃反应3小时。反应液浓缩,柱层析纯化方法制备得无色油状物5-(溴甲基)-2-氯-3-氟吡啶(0.45g)。
步骤10b:制备2-(6-氯-5-氟吡啶-3-基)乙腈
Figure PCTCN2021105180-appb-000093
步骤10b的具体方式:将5-(溴甲基)-2-氯-3-氟吡啶(450mg,2.0mmol)溶于5mL乙腈中,加入四丁基氰化铵(644mg,2.4mmol),室温反应2小时。将反应液浓缩,加入20mL水,乙酸乙酯(50mL×3)萃取,无水硫酸钠干燥,过滤,浓缩。将浓缩的反应液柱层析纯化得无色油状物2-(6-氯-5-氟吡啶-3-基)乙腈(275mg)。
步骤10c:制备2-(6-氯-5-氟吡啶-3-基)乙酸甲酯
Figure PCTCN2021105180-appb-000094
步骤10c的具体方式:将2-(6-氯-5-氟吡啶-3-基)乙腈(275mg,1.62mmol)溶于3mL甲醇中,氮气保护下加入二氯亚砜(0.6mL,8.10mmol),室温反应5小时。加入20mL水,乙酸乙酯萃取(50mL),无水硫酸钠干燥,过滤,浓缩。将浓缩的反应液柱层析纯化得无色油状物2-(6-氯-5-氟吡啶-3-基)乙酸甲酯(253mg)。
步骤10d:制备2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-5-氟吡啶-3-基乙酸甲酯
Figure PCTCN2021105180-appb-000095
步骤10d的具体方式:(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)硼酸(351mg,0.83mmol)和2-(6-氯-5-氟吡啶-3-基)乙酸甲酯(253mg,1.25mmol)按照实施例1步骤1h的方法制备得棕色固体2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-5-氟吡啶-3-基乙酸甲酯(110mg)。
步骤10e:制备2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-5-氟吡啶-3-基)乙酸
Figure PCTCN2021105180-appb-000096
步骤10e的具体方式:将2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-5-氟吡啶-3-基乙酸甲酯(110mg,0.20mmol)与氢氧化钠(16mg,0.40mmol)按照实施例1步骤1i的方法制备得棕色固体2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-5-氟吡啶-3-基)乙酸(106mg)。
步骤10f:制备2-((2-乙基-6-(3-氟-5-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-2-基)咪唑[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000097
步骤10f的具体方式:将2-(6-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-5-氟吡啶-3-基)乙酸(106mg,0.2mmol)与氮杂环丁烷-3-醇(27mg,0.37mmol)按照实施例1步骤1j的方法制备得灰白色固体2-((2-乙基-6-(3-氟-5-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)吡啶-2-基)咪唑[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(29.4mg)。
1H NMR(400MHz,CDCl 3):δ8.48(s,1H),8.37(s,1H),8.19-8.16(dd,J=8.8,5.2Hz,2H),8.05(d,J=9.6Hz,1H),7.72(d,J=9.2Hz,1H),7.57(d,J=12.0Hz,1H),7.19(t,J=8.8Hz,2H),4.74(br.s,1H),4.45(t,J=8.4Hz,1H),4.32-4.28(m,1H),4.14-4.09(m,1H),3.95-3.90(m,1H),3.70(s,3H),2.81(q,J=7.6Hz,2H),2.52(s,2H),1.41(t,J=7.6Hz,3H).
MS实测值(ESI +)[(M+H) +]:586。
实施例11:(S)-2-((2-乙基-6-(5-(2-(3-羟基吡咯烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物11)
Figure PCTCN2021105180-appb-000098
步骤11a:制备(S)-2-((2-乙基-6-(5-(2-(3-羟基吡咯烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000099
步骤11a的具体方式:将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)乙酸(40mg,0.08mmol)和(S)-3-吡咯烷醇(8.14mg,0.093mmol)按照实施例1步骤1j的方法制备得灰白色固体(S)-2-((2-乙基-6-(5-(2-(3-羟基吡咯烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(20.5mg)。
1H NMR(400MHz,CDCl 3):δ8.90(s,1H),8.66(s,1H),8.55(s,1H),8.17(dd,J=8.4,5.6Hz,2H),7.90(d,J=9.2Hz,1H),7.77(d,J=9.2Hz,1H),7.19(t,J=8.4Hz,2H),4.63-4.48(m,1H),3.94(s,2H),3.86-3.73(m,2H),3.71(s,3H),3.69-3.63(m,2H),2.81(q,J=7.6Hz,2H),2.02-1.99(m,2H),1.40(t,J=7.6Hz,3H)。
MS实测值(ESI +)[(M+H) +]:583。
实施例12:2-((2-乙基-6-(5-(2-(3-(羟甲基)氮杂环丁烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物12)
Figure PCTCN2021105180-appb-000100
步骤12a:制备2-((2-乙基-6-(5-(2-(3-(羟甲基)氮杂环丁烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000101
步骤12a的具体方式:将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a] 吡啶-6-基)吡嗪-2-基)乙酸(40mg,0.08mmol)和3-(羟甲基)氮杂环丁烷盐酸盐(11.55mg,0.093mmol)按照实施例1步骤1j的方法制备得灰白色固体2-((2-乙基-6-(5-(2-(3-(羟甲基)氮杂环丁烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(12.5mg)。
1H NMR(400MHz,CDCl 3):δ8.91(s,1H),8.66(s,1H),8.55(s,1H),8.17(dd,J=8.4,5.6Hz,2H),7.90(d,J=9.2Hz,1H),7.76(d,J=9.2Hz,1H),7.19(t,J=8.4Hz,2H),4.41-4.30(m,1H),4.17-4.07(m,2H),3.88-3.79(m,3H),3.73(s,2H),3.70(s,3H),2.90-2.86(m,1H),2.82(q,J=7.6Hz,2H),1.41(t,J=7.60Hz,3H)。
MS实测值(ESI +)[(M+H) +]:583。
实施例13:(R)-2-((2-乙基-6-(5-(2-(3-羟基吡咯烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物13)
Figure PCTCN2021105180-appb-000102
步骤13a:制备(R)-2-((2-乙基-6-(5-(2-(3-羟基吡咯烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000103
步骤13a的具体方式:将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)乙酸(40mg,0.08mmol)和(R)-吡咯烷丁-3-醇(14mg,0.16mmol)按照实施例1步骤1j的方法制备得灰白色固体(R)-2-((2-乙基-6-(5-(2-(3-羟基吡咯烷-1-基)-2-氧代乙基)吡嗪-2-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(16mg)。
1H NMR(400MHz,CDCl 3):δ8.90(s,1H),8.67(s,1H),8.55(s,1H),8.17(dd,J=8.4,5.6Hz,2H),7.90(d,J=9.2Hz,1H),7.78-7.76(m,1H),7.19(t,J=8.4Hz,2H),4.61-4.52(m,1H),3.94(s,2H),3.86-3.73(m,2H),3.73(s,3H),3.69-3.63(m,2H),2.81(q,J=7.6Hz,2H),2.13-1.97(m,2H),1.41(t,J=7.6Hz,3H)。
MS实测值(ESI +)[(M+H) +]:583。
实施例14:2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)-N-((5-氧代吡咯烷-2-基)甲基)乙酰胺(化合物14)
Figure PCTCN2021105180-appb-000104
步骤14a:制备2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)-N-((5-氧代吡咯烷-2-基)甲基)乙酰胺
Figure PCTCN2021105180-appb-000105
步骤14a的具体方式:将2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)乙酸(40mg,0.08mmol)和5-(氨基甲基)吡咯烷-2-酮(18mg,0.16mmol)按照实施例1步骤1j的方法制备得灰白色固体2-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-基)-N-((5-氧代吡咯烷-2-基)甲基)乙酰胺(20mg)。
1H NMR(400MHz,CDCl 3):δ8.83-8.79(m,1H),8.62-8.51(m,2H),8.16(dd,J=8.5,5.4Hz,2H),7.89(d,J=9.3Hz,1H),7.75(d,J=7.5Hz,1H),7.70-7.55(m,1H),7.18(t,J=8.6Hz,2H),6.93-6.79(m,1H),3.91-3.90(m,1H),3.77(s,2H),3.71(s,3H),3.59-3.49(m,1H),3.26-3.16(m,1H),2.81(q,J=7.6Hz,2H),2.32-2.17(m,2H),1.84-1.73(m,2H),1.41(t,J=7.6Hz,3H)。
MS实测值(ESI +)[(M+H) +]:610。
实施例15:5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-N-(2-氮杂螺[3.3]庚烷-6-基)吡嗪-2-甲酰胺(化合物15)
Figure PCTCN2021105180-appb-000106
步骤15a:制备6-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-甲酰胺)-2-氮杂螺[3.3]庚烷-2-甲酸叔丁酯
Figure PCTCN2021105180-appb-000107
步骤15a的具体方式:将5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-羧酸(40mg,0.08mmol)和6-氨基-2-氮杂螺[3.3]庚烷-2-羧酸叔丁酯(34mg,0.16mmol)按照实施例1步骤1j的方法制备得灰白色固体6-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-甲酰胺)-2-氮杂螺[3.3]庚烷-2-甲酸叔丁酯(32mg)。
步骤15b:制备5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-N-(2-氮杂螺[3.3]庚烷-6-基)吡嗪-2-甲酰胺
Figure PCTCN2021105180-appb-000108
步骤15b的具体方式:向6-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)吡嗪-2-甲酰胺)-2-氮杂螺[3.3]庚烷-2-甲酸叔丁酯(32mg,0.046mmol)的二氯甲烷(1ml)溶液中加入三氟乙酸(2ml),室温搅拌1小时后用饱和碳酸氢钠水溶液淬灭,并用二氯甲烷(10mL×3)。萃取将合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗产品。粗产品经过快速柱层析色谱纯化得白色固体5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-N-(2-氮杂螺[3.3]庚-6-基)吡嗪-2-甲酰胺(10.4mg)。
1H NMR(400MHz,DMSO d 6):δ9.38(s,1H),8.91(s,1H),8.68(s,1H),8.19-8.16(m,2H),7.94(d,J=9.2Hz,1H),7.81(t,J=10.0Hz,1H),7.20(t,J=8.8Hz,2H),4.50-4.44(m,1H),3.82(s,2H),3.73(s,3H),3.70(s,2H),2.81(q,J=7.6Hz,2H),2.79-2.74(m,2H),2.27-2.19(m,2H),1.42(t,J=7.6Hz,3H)。
MS实测值(ESI +)[(M+H) +]:594。
实施例16:2-((2-乙基-6-(5-氟-6-(3-羟基氮杂环丁烷-1-羰基)吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物16)
Figure PCTCN2021105180-appb-000109
步骤16a:制备(5-氟-6-(甲氧羰基)吡啶-3-基)硼酸
Figure PCTCN2021105180-appb-000110
步骤16a的具体方式:将5-溴-3-氟吡啶甲酸甲酯(100mg,0.429mmol)和联硼酸频那醇酯(130mg,0.52mmol)按照实施例2步骤2a的方法制备得黑色油状物(5-氟-6-(甲氧羰基)吡啶-3-基)硼酸(140mg)。
步骤16b:制备5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-3-氟吡啶甲酸甲酯
Figure PCTCN2021105180-appb-000111
步骤16b的具体方式:将(5-氟-6-(甲氧羰基)吡啶-3-基)硼酸(85mg,0.43mmol)和2-((6-溴-2-乙 基咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(391mg,0.86mmol)按照实施例1步骤1h的方法制备得黄色固体5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-3-氟吡啶甲酸甲酯(150mg)。
步骤16c:5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-3-氟吡啶甲酸
Figure PCTCN2021105180-appb-000112
步骤16c的具体方式:将5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-3-氟吡啶甲酸甲酯(150mg,0.28mmol)与氢氧化钠(24mg,0.60mmol)按照实施例1步骤1i的方法制备得白色固体5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-3-氟吡啶甲酸(191mg)。
步骤16d:制备2-((2-乙基-6-(5-氟-6-(3-羟基氮杂环丁烷-1-羰基)吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000113
步骤16d的具体方式:5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)-3-氟吡啶甲酸(146mg,0.28mmol)与氮杂环丁烷-3-醇(27mg,0.37mmol)按照实施例1步骤1j的方法制备得白色固体2-((2-乙基-6-(5-氟-6-(3-羟基氮杂环丁烷-1-羰基)吡啶-3-基)咪唑并[1,2-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(65.8mg)。
1H NMR(400MHz,CDCl 3):δ8.59(s,1H),8.15-8.11(m,2H),8.00(s,1H),7.80(d,J=9.2Hz,1H),7.66(d,J=10.4Hz,1H),7.52(d,J=8.8Hz,1H),7.16(t,J=8.8Hz,2H),4.74(s,1H),4.60-4.56(m,1H),4.51-4.46(m,,1H),4.27-4.23(m,1H),4.11-4.07(m,1H),3.68(s,3H),2.79(q,J=7.6Hz,2H),1.39(t,J=7.6Hz,3H)。
MS实测值(ESI +)[(M+H) +]:572。
实施例17:2-((5-(2-(1,1-二氧化异噻唑烷-2-基)嘧啶-5-基)-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物17)
Figure PCTCN2021105180-appb-000114
步骤17a:制备2-溴-5-氟-1-羟基吡啶-1-鎓
Figure PCTCN2021105180-appb-000115
步骤17a的具体方式:室温下,在2-溴-5-氟吡啶(50.0g,36.78mmol)的三氟乙酸(400mL)溶液中加入双氧水(80mL)。将混合物在85℃下搅拌10小时后冷却至室温,加入硫代硫酸钠(120g)淬灭,并过滤。母液用氢氧化钠水溶液调节pH至7-8,用二氯甲烷(800ml x 3)萃取。有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤并浓缩得灰色固体2-溴-5-氟-1-羟基吡啶-1-鎓(50g)。
步骤17b:制备6-溴-3-氟-2-氰基吡啶
Figure PCTCN2021105180-appb-000116
步骤1b的具体方式:室温下,向2-溴-5-氟-1-羟基吡啶-1-鎓(50g,0.25mol)的乙腈溶液(400mL)加入三乙胺(126g,1.25mol)和三甲基氰硅烷(125g,1.25mol)。将混合物加热至90℃反应12小时,然后浓缩,粗产品经过快速柱层析色谱纯化得类白色固体6-溴-3-氟-2-氰基吡啶(22g)。
步骤17c:制备5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-胺
Figure PCTCN2021105180-appb-000117
步骤17c的具体方式:向6-溴-3-氟-2-氰基吡啶(22g,0.11mol)和乙肼草酸盐(25g,0.16mol)的二甲基亚砜(110ml)溶液中加入碳酸钾(53g,0.38mol),反应加热至110℃反应2h。将反应液冷却至室温,用乙酸乙酯(800ml)稀释,有机相分别用水(200ml x 3),饱和食盐水(200ml)洗,干燥,过滤浓缩。粗产品经过快速柱层析色谱纯化得黄色固体5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-胺(3.2g)。
步骤17d:制备2-((5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000118
步骤17d的具体方式:零度下,氮气氛围中向5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-胺(3.2g,13mmol)的DMF溶液(30mL)分批加入60%氢化钠(1.1g,26.4mmol)。反应液在0℃下搅拌15分钟后分批加入2-氯-4-(4-氟苯基)噻唑-5-甲腈(3.4g,14.5mmol)。混合物在室温下继续反应1小时后加水(60mL)淬灭,并过滤。滤饼用乙酸乙酯(20mL×3)洗涤,然后干燥得黄色固体2-((5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(6g)。
步骤17e:制备2-((5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000119
步骤17e的具体方式:零度下,氮气氛围中向2-((5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(6g,13.6mmol)的DMF溶液(50mL)分批加入60%氢化钠(1.4g,33.9 mmol)。反应液在室温下搅拌0.5小时后加入碘乙烷(4.2g,27.1mmol)。混合物在室温下继续反应3小时后加水(200mL)淬灭,用乙酸乙酯萃取(100mL×3),合并的有机相用饱和食盐水洗涤(50mL),干燥,过滤后减压浓缩得到粗产品。粗产品通过快速柱层析色谱纯化得黄色固体2-((5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(5.5g)。
步骤17f:制备2-(5-溴嘧啶-2-基)异噻唑烷1,1-二氧化物
Figure PCTCN2021105180-appb-000120
步骤17f的具体方式:氮气氛围中,向3-氯丙烷-1-磺酰胺(1.85g,7.87mmol)和2,5-二溴嘧啶(954mg,6.05mmol)的叔丁醇溶液(10mL)加入三(二亚苄基丙酮)二钯(277mg,0.30mmol),2-(二叔丁基膦)联苯(180mg,0.61mmol),4,5-双二苯基膦-9,9-二甲基氧杂蒽(350mg,0.61mmol)和碳酸铯(3.94g,12.11mmol)。混合物在100℃搅拌4小时后加水(40mL)和乙酸乙酯(3mL)稀释并在室温下搅拌10分钟后过滤,用水洗涤滤饼,然后用乙酸乙酯/石油醚(1:5,10mLx2)洗涤滤饼,干燥得2-(5-溴嘧啶-2-基)异噻唑烷1,1-二氧化物(1.5g)。
步骤17g:制备(2-(1,1-二氧化异噻唑烷-2-基)嘧啶-5-基)硼酸
Figure PCTCN2021105180-appb-000121
步骤17g的具体方式:氮气保护下,向2-(5-溴嘧啶-2-基)异噻唑烷1,1-二氧化物(5g,18.0mmol),联硼酸频那醇酯(5.0g,19.8mmol),醋酸钾(5.3g,54.1mmol)的1,4-二氧六环溶液(50mL)加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(661mg,0.9mmol)。将混合物加热到85℃搅拌2小时后,加水(80mL)稀释,用乙酸乙酯萃取(100mL×2)。将合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,浓缩得到棕色油状物(2-(1,1-二氧化异噻唑烷-2-基)嘧啶-5-基)硼酸(5g)。
步骤17h:制备2-((5-(2-(1,1-二氧化异噻唑烷-2-基)嘧啶-5-基)-2-乙基-2H-吡唑啉[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000122
步骤17h的具体方式:氮气氛围中,向2-((5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(5g,10.6mmol)和(2-(1,1-二氧化异噻唑烷-2-基)嘧啶-5-基)硼酸(3.1g,12.8mmol)的1,4-二氧六环溶液(50mL)和水(7ml)中加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(389mg,0.53mmol)和磷酸钾(2.3g,17.0mmol)。混合物在75℃搅拌2小时后加水(50mL)稀释,并用乙酸乙酯萃取(50mL×3)。将合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到粗产品。粗产品经过快速柱层析色谱纯化得棕色固体2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)苯基)乙酸甲酯(4.5g)。
1H NMR(400MHz,DMSO-d 6):δ9.26(s,2H),8.20(d,J=9.2Hz,1H),8.13(dd,J=8.8,5.2Hz,2H),7.70(d,J=9.2Hz,1H),7.17(t,J=8.4Hz,2H),4.50-4.65(m,1H),4.44(q,J=7.2Hz,2H),4.12(t,J=6.4Hz,2H),3.98-3.84(m,1H),3.50(q,J=7.2Hz,2H),2.57-2.51(m,2H),1.68(t,J=7.2Hz,3H), 1.48(t,J=7.2Hz,3H)。
MS实测值(ESI +)[(M+H) +]:590。
实施例18:2-((5-(2-(1,1-二氧化异噻唑烷-2-基)嘧啶-5-基)-2-乙基吡唑并[1,5-a]嘧啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物18)
Figure PCTCN2021105180-appb-000123
步骤18a:制备2-乙基吡唑并[1,5-a]嘧啶-5(4H)-酮
Figure PCTCN2021105180-appb-000124
步骤18a的具体方式:室温下,向3-乙基-1H-吡唑-5-胺(5g,45mmol)和1,3-二甲基嘧啶-2,4(1H,3H)-二酮(7.6g,54mmol)的乙醇(50mL)溶液中加入乙醇钠(9.2g,135mmol)。将混合物在90℃下搅拌5小时后冷却至室温,浓缩后加入稀盐酸(2M)调节pH至2-3。过滤,滤饼干燥得白色固体2-乙基吡唑并[1,5-a]嘧啶-5(4H)-酮(5g)。
步骤18b:制备2-乙基-3-硝基吡唑并[1,5-a]嘧啶-5(4H)-酮
Figure PCTCN2021105180-appb-000125
步骤18b的具体方式:零度下,向2-乙基吡唑并[1,5-a]嘧啶-5(4H)-酮(5g,0.25mol)的浓硫酸溶液(20mL)中缓慢滴加浓硝酸(2.2ml)。将混合物在0℃下继续反应2小时,然后反应液缓慢加入冰水中淬灭,过滤,干燥得淡黄色固体2-乙基-3-硝基吡唑并[1,5-a]嘧啶-5(4H)-酮(5g)。
步骤18c:制备5-氯-2-乙基-3-硝基吡唑[1,5-a]嘧啶
Figure PCTCN2021105180-appb-000126
步骤18c的具体方式:向2-乙基-3-硝基吡唑并[1,5-a]嘧啶-5(4H)-酮(5g,24mol)的三氯氧磷(30ml)溶液中加入N,N-二甲基苯胺(4.4g,36mol),反应加热至90℃反应2h。将反应液冷却至室温,并将其浓缩,加水淬灭,并用乙酸乙酯(100ml)萃取,有机相分别用1M碳酸氢钠溶液(40ml)洗涤,饱和食盐水(40ml)洗涤,干燥,过滤浓缩得黄色固体5-氯-2-乙基-3-硝基吡唑[1,5-a]嘧啶(4.5g)。
步骤18d:制备5-氯-2-乙基吡唑[1,5-a]嘧啶-3-胺
Figure PCTCN2021105180-appb-000127
步骤18d的具体方式:向5-氯-2-乙基-3-硝基吡唑[1,5-a]嘧啶(4.5g,13mmol)的乙醇溶液(40mL)中加入铁粉(1.1g,26.4mmol)和饱和氯化铵溶液(20ml)。将反应液加热至45℃,反应2小时后过滤浓缩。残留物用乙酸乙酯(50mL×3)萃取,合并的有机相用饱和食盐水洗涤,干燥,浓缩得到粗产品。粗产品通过快速柱层析色谱纯化得黄色固体5-氯-2-乙基吡唑[1,5-a]嘧啶-3-胺(3g)。
步骤18e:制备2-((5-氯-2-乙基吡唑并[1,5-a]嘧啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000128
步骤18e的具体方式:零度下,氮气氛围中向5-氯-2-乙基吡唑[1,5-a]嘧啶-3-胺(3.0g,15mmol)的THF溶液(30mL)分批加入60%氢化钠(1.2g,30.1mmol)。反应液在0℃下搅拌15分钟后分批加入2-氯-4-(4-氟苯基)噻唑-5-甲腈(4.3g,18.4mmol)。混合物在90℃下继续反应6小时后冷却至室温,加水(60mL)淬灭,用乙酸乙酯萃取(30mL×3),合并的有机相用盐水洗涤(50mL),干燥,过滤浓缩减压浓缩得到粗产品。粗产品通过快速柱层析色谱纯化得黄色固体2-((5-氯-2-乙基吡唑并[1,5-a]嘧啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(600mg)。
步骤18f:制备2-((5-氯-2-乙基吡唑并[1,5-a]嘧啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000129
步骤18f的具体方式:零度下,氮气氛围中向2-((5-氯-2-乙基吡唑并[1,5-a]嘧啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(600mg,1.5mmol)的DMF溶液(6mL)加入碳酸铯(733mg,2.2mmol)。反应液在室温下搅拌十分钟后加入碘乙烷(305mg,1.9mmol)。混合物在室温下继续反应2小时后加水(20mL)淬灭,用乙酸乙酯萃取(20mL×3),合并的有机相用盐水洗涤(20mL),干燥,过滤后减压浓缩得到粗产品。粗产品通过快速柱层析色谱纯化得黄色固体2-((5-氯-2-乙基吡唑并[1,5-a]嘧啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(400mg)。
步骤18g:制备2-((5-(2-(1,1-二氧化异噻唑烷-2-基)嘧啶-5-基)-2-乙基吡唑并[1,5-a]嘧啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000130
步骤18g的具体方式:氮气氛围中,将2-((5-氯-2-乙基吡唑并[1,5-a]嘧啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(122mg,0.258mmol)和(2-(1,1-二氧化异噻唑烷-2-基)嘧啶-5-基)硼酸(94mg,0.388mmol)按照实施例17步骤17h的方法制备得黄色固体2-((5-(2-(1,1-二氧化异噻唑烷-2-基)嘧啶-5-基)-2-乙基吡唑并[1,5-a]嘧啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(108mg)。
1H NMR(400MHz,CDCl 3):δ9.27(s,2H),8.72(d,J=7.6Hz,1H),8.16(dd,J=8.8,5.6Hz,2H),7.26(br.s.,1H),7.18(t,J=8.8Hz,2H),4.36-4.32(m,1H),4.13(t,J=6.4Hz,2H),3.95-3.83(m,1H),3.51(t,J=7.2Hz,2H),2.90-2.80(m,2H),2.59-2.54(m,2H),1.47-1.34(m,6H)。
MS实测值(ESI +)[(M+H) +]:590。
实施例19:2-((6-(5-(1,1-二氧化异噻唑烷-2-基)吡嗪-2-基)-2-乙基咪唑并[1,2-a]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物19)
Figure PCTCN2021105180-appb-000131
步骤19a:制备2-(5-溴吡嗪-2-基)异噻唑烷1,1-二氧化物
Figure PCTCN2021105180-appb-000132
步骤19a的具体方式:氮气氛围中,将2,5-二溴吡嗪(900mg,3.82mmol)和3-氯丙烷-1-磺酰胺(400mg,2.54mmol)按照实施例17步骤17f的方法制备得棕色固体2-(5-溴吡嗪-2-基)异噻唑烷1,1-二氧化物(364mg)。
步骤19b:制备2-((6-(5-(1,1-二氧化异噻唑烷-2-基)吡嗪-2-基)-2-乙基咪唑并[1,2-a]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000133
步骤19b的具体方式:氮气氛围中,将2-(5-溴吡嗪-2-基)异噻唑烷1,1-二氧化物(170mg,0.614mmol)和(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)硼酸(400mg,0.921mmol)按照实施例17步骤17h的方法制备得白色固体2-((6-(5-(1,1-二氧化异噻唑烷-2-基)吡嗪-2-基)-2-乙基咪唑并[1,2-a]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(74.8mg)。
1H NMR(400MHz,DMSO-d 6):δ8.76(s,1H),8.67(s,1H),8.52(s,1H),8.18-8.15(m,2H),7.84(d,J=9.2Hz,1H),7.74(d,J=9.2Hz,1H),7.20(t,J=8.4Hz,2H),4.35-4.26(m,1H),4.08-4.03(m,3H),3.48(t,J=7.2Hz,2H),2.81-2.76(m,2H),2.66-2.59(m,2H),1.44(t,J=7.2Hz,3H),1.38(t,J=7.6Hz,3H)。
MS实测值(ESI +)[(M+H) +]:589。
实施例20:N-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)-N-甲基甲磺酰胺(化合物20)
Figure PCTCN2021105180-appb-000134
步骤20a:制备N-(5-溴嘧啶-2-基)-N-甲基甲磺酰胺
Figure PCTCN2021105180-appb-000135
步骤20a的具体方式:氮气氛围中,将N-甲基甲磺酰胺(77mg,0.707mmol)和2,5-二溴嘧啶(200mg,0.848mmol)按照实施例17步骤17f的方法制备得N-(5-溴嘧啶-2-基)-N-甲基甲磺酰胺(114mg)。
步骤20b:制备N-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)-N-甲基甲磺酰胺
Figure PCTCN2021105180-appb-000136
步骤20b的具体方式:氮气氛围中,将N-(5-溴嘧啶-2-基)-N-甲基甲磺酰胺(70mg,0.263mmol)和(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)硼酸(172mg,0.395mmol)按照实施例17步骤17h的方法制备得棕色固体N-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)-N-甲基甲磺酰胺(45mg)。
1H NMR(400MHz,CDCl 3):δ8.77(s,2H),8.16(dd,J=8.8,5.6Hz,2H),7.92(br.s,1H),7.80(d,J=9.2Hz,1H),7.48(dd,J=9.2,1.2Hz,1H),7.19(t,J=8.8Hz,2H),4.26-4.21(m,1H),4.10-4.05(m,1H),3.60(s,3H),3.53(s,3H),2.81(q,J=7.6Hz,2H),1.44(t,J=7.6Hz,3H),1.38(t,J=7.2Hz,3H)。
MS实测值(ESI +)[(M+H) +]:577。
实施例21:2-((5-(2-(1,1-二氧化-1,2-噻嗪烷-2-基)嘧啶-5-基)-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物21)
Figure PCTCN2021105180-appb-000137
步骤21a:制备2-(5-溴嘧啶-2-基)-1,2-噻嗪烷1,1-二氧化物
Figure PCTCN2021105180-appb-000138
步骤21a的具体方式:室温下,氮气氛围中,将1,2-噻嗪烷1,1-二氧化物(300mg,2.22mmol)和2,5-二溴嘧啶(792mg,3.33mmol)按照实施例17步骤17f的方法制备得类棕色固体2-(5-溴嘧啶-2-基)-1,2-噻嗪烷1,1-二氧化物(500mg)。
步骤21b:制备(2-(1,1-二氧化-1,2-噻嗪烷-2-基)嘧啶-5-基)硼酸
Figure PCTCN2021105180-appb-000139
步骤21b的具体方式:氮气氛围中,向2-(5-溴嘧啶-2-基)-1,2-噻嗪烷1,1-二氧化物(200mg,0.68mmol)和联硼酸频那醇酯(191mg,0.75mmol)按照实施例17步骤17g的方法制备得到(2-(1,1-二氧化-1,2-噻嗪烷-2-基)嘧啶-5-基)硼酸(176mg)。
步骤21c:制备2-((5-(2-(1,1-二氧化-1,2-噻嗪烷-2-基)嘧啶-5-基)-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000140
步骤21c的具体方式:氮气氛围中,将(2-(1,1-二氧化-1,2-噻嗪烷-2-基)嘧啶-5-基)硼酸(175mg,0.68mmol)和2-((5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(320mg,0.68mmol)按照实施例17步骤17h的方法制备得类白色固体2-((5-(2-(1,1-二氧化-1,2-噻嗪烷-2-基)嘧啶-5-基)-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(60mg)。
1H NMR(400MHz,CDCl3):δ9.30(s,2H),8.22(d,J=9.2Hz,1H),8.12(dd,J=8.4,5.2Hz,2H),7.72(d,J=9.2Hz,1H),7.19-7.15(m,2H),4.63-4.49(m,1H),4.44(q,J=7.2Hz,2H),4.38-4.32(m,2H),3.93-3.89(m,1H),3.39-3.30(m,2H),2.38-2.30(m,2H),1.89-1.80(m,2H),1.68(t,J=7.2Hz,3H),1.47(t,J=7.2Hz,3H)。
MS实测值(ESI +)[(M+H) +]:604。
实施例22:N-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)-2-羟基乙-1-磺酰胺(化合物22)
Figure PCTCN2021105180-appb-000141
步骤22a:制备2-(苄氧基)乙-1-磺酸
Figure PCTCN2021105180-appb-000142
步骤22a的具体方式:室温下,在亚硫酸钠(3.62g,28.67mmol)的水溶液(170mL)中加入((2-溴乙氧基)甲基)苯(5g,23.36mmol)。将混合物在100℃下搅拌6小时,浓缩得到白色固体。然后向白色固体中加入甲醇(170mL),将混合物加热至50℃搅拌20分钟,过滤,滤液浓缩得白色固体2-(苄氧基)乙-1-磺酸(6.8g)。
步骤22b:制备2-(苄氧基)乙-1-磺酰氯
Figure PCTCN2021105180-appb-000143
步骤22b的具体方式:室温下,氮气氛围中,将DMF(163mg,2.3mmol)和氯化亚砜(5.1mL,90.81mmol)加入到2-(苄氧基)乙-1-磺酸(5.0g,23.14mmol)的四氢呋喃溶液(20mL)中。将混合物 在65℃下搅5小时。过滤,滤液浓缩得黄色油状2-(苄氧基)乙-1-磺酰氯(5.6g)。
步骤22c:制备2-(苄氧基)-N-(5-溴嘧啶-2-基)乙-1-磺酰胺
Figure PCTCN2021105180-appb-000144
步骤22c的具体方式:室温下,将5-溴吡啶-2-胺(6.0g,34.69mmol)和三乙胺(6.4mL,46.15mmol)加入到2-(苄氧基)乙-1-磺酰氯(5.4g,23.08mmol)的四氢呋喃溶液(50mL)中,加热至80℃搅拌反应10小时。冷却后,加水稀释(50mL)。水相用乙酸乙酯萃取(80mL×3),将合并的有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤浓缩得粗产品。粗产品通过快速柱层析色谱纯化得白色固体2-(苄氧基)-N-(5-溴嘧啶-2-基)乙-1-磺酰胺(560mg)。
步骤22d:制备2-(苄氧基)-N-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)乙-1-磺酰胺
Figure PCTCN2021105180-appb-000145
步骤22d的具体方式:氮气氛围中,将(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)硼酸(985mg,2.26mmol)和2-(苄氧基)-N-(5-溴嘧啶-2-基)乙-1-磺酰胺(371mg,1.51mmol)按照实施例17步骤17h的方法制备得棕色固体2-(苄氧基)-N-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)乙-1-磺酰胺(530mg)。
步骤22e:制备N-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)-2-羟基乙-1-磺酰胺
Figure PCTCN2021105180-appb-000146
步骤22e的具体方式:向2-(苄氧基)-N-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)乙-1-磺酰胺(530mg,0.777mmol)加入三氟乙酸(5mL)。混合物在微波中加热到90℃后搅拌1小时。然后用饱和碳酸氢钠溶液调节pH至8~9,用乙酸乙酯萃取(10mL×3)。将合并的有机相用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得棕色固体N-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)-2-羟基乙-1-磺酰胺(25.6mg)。
1H NMR(400MHz,DMSO-d 6):δ8.82(s,2H),8.17-8.13(m,2H),7.95(br.s,1H),7.81(d,J=9.2Hz,1H),7.47(d,J=9.2Hz,1H),7.19(t,J=8.4Hz,2H),4.27-4.18(m,1H),4.12-4.06(m,3H),3.86-3.85(m,2H),2.81(q,J=7.2Hz,2H),1.44(t,J=7.2Hz,3H),1.39(t,J=7.2Hz,3H)。
MS实测值(ESI +)[(M+H) +]:593。
实施例23:2-(乙基(2-乙基-5-(2-(3-羟基氮杂环丁-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物23)
Figure PCTCN2021105180-appb-000147
步骤23a:制备1-(5-溴嘧啶-2-基)氮杂环丁烷-3-醇
Figure PCTCN2021105180-appb-000148
步骤23a的具体方式:室温下,在2,5-二溴嘧啶(440mg,1.85mmol)的DMF(5mL)溶液中加入氮杂环丁烷-3-醇(149mg,2.03mmol)和碳酸钾(511mg,3.7mmol)。将混合物在80℃下搅拌1h。加入50mL水,过滤,滤饼旋干得类白色固体1-(5-溴嘧啶-2-基)氮杂环丁烷-3-醇(360mg)。
步骤23b:制备(2-(3-羟基氮杂环丁烷-1-基)嘧啶-5-基)硼酸
Figure PCTCN2021105180-appb-000149
步骤23b的具体方式:氮气氛围中,将1-(5-溴嘧啶-2-基)氮杂环丁烷-3-醇(100mg,0.43mmol)和联硼酸频那醇酯(121mg,0.48mmol)按照实施例17步骤17g的方法制备得到(2-(3-羟基氮杂环丁烷-1-基)嘧啶-5-基)硼酸(85mg)。
步骤23c:制备2-(乙基(2-乙基-5-(2-(3-羟基氮杂环丁烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000150
步骤23c的具体方式:氮气氛围中,将(2-(3-羟基氮杂环丁烷-1-基)嘧啶-5-基)硼酸(85mg,0.43mmol)和2-((5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(205mg,0.43mmol)按照实施例17步骤17h的方法制备得白色固体2-(乙基(2-乙基-5-(2-(3-羟基氮杂环丁烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(38mg)。
1H NMR(400MHz,CDCl3):δ9.02(s,2H),8.14-8.11(m,3H),7.64(d,J=9.2Hz,1H),7.20-7.15(m,2H),4.86-4.82(m,1H),4.51-4.47(m,2H),4.42(q,J=7.2Hz,2H),4.11-4.08(m,2H),3.91-3.87(m,1H),2.28-2.24(m,1H),1.67(t,J=7.2Hz,3H),1.48(t,J=7.2Hz,3H)。
MS实测值(ESI +)[(M+H) +]:542。
实施例24:2-(乙基(2-乙基-5-(2-(3-(羟甲基)氮杂环丁烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物24)
Figure PCTCN2021105180-appb-000151
步骤24a:制备(1-(5-溴嘧啶-2-基)氮杂环丁烷-3-基)甲醇
Figure PCTCN2021105180-appb-000152
步骤24a的具体方式:室温下,向5-溴-2-氯嘧啶(500mg,2.58mmol)的DMF溶液(5mL)中加入氮杂环丁烷-3-基甲醇盐酸盐(368mg,2.97mmol)和三乙胺(1.1mL,7.75mmol)。将混合物在60℃下搅拌4小时。冷却后,将反应液浓缩,加水稀释(60mL)。水相用乙酸乙酯萃取(20mL×3),将合并的有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤浓缩。粗品用快速色谱法分离纯化得黄色固体(1-(5-溴嘧啶-2-基)氮杂环丁烷-3-基)甲醇(618mg)。
步骤24b:制备(2-(3-(羟甲基)氮杂环丁烷-1-基)嘧啶-5-基)硼酸
Figure PCTCN2021105180-appb-000153
步骤24b的具体方式:将(1-(5-溴嘧啶-2-基)氮杂环丁烷-3-基)甲醇(500mg,1.70mmol)和联硼酸频那醇酯(781mg,2.21mmol)按照实施例17步骤17g的方法制备得(2-(3-(羟甲基)氮杂环丁烷-1-基)嘧啶-5-基)硼酸(529mg)。
步骤24c:制备2-(乙基(2-乙基-5-(2-(3-(羟甲基)氮杂环丁烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000154
步骤24c的具体方式:将2-((5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(100mg,0.21mmol)和(2-(3-(羟甲基)氮杂环丁烷-1-基)嘧啶-5-基)硼酸(133mg,0.32mmol)按照实施例17步骤17h的方法制备得白色固体2-(乙基(2-乙基-5-(2-(3-(羟甲基)氮杂环丁烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(40mg)。
1H NMR(400MHz,CDCl 3):δ9.00(s,2H),8.13-8.10(m,3H),7.62(d,J=8.8Hz,1H),7.16(m,2H),4.65-3.76(m,10H),2.95(br.s.,1H),1.41-1.69(m,6H)。
MS实测值(ESI +)[(M+H) +]:556。
实施例25:1-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基-2H-吡唑并[4,3-b]吡啶-5-基)嘧啶-2-基)氮杂环丁烷-3-基乙酸酯(化合物25)
Figure PCTCN2021105180-appb-000155
步骤25a:制备1-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基-2H-吡唑并[4,3-b]吡啶-5-基)嘧啶-2-基)氮杂环丁烷-3-基乙酸酯
Figure PCTCN2021105180-appb-000156
步骤25a的具体方式:将2-(乙基(2-乙基-5-(2-(3-羟基氮杂环丁烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(200mg,0.37mmol)和三乙胺(112mg,1.11mmol)的二氯甲烷(3mL)溶液中,氮气保护下,在0℃条件下加入乙酰氯(58mg,0.74mmol)。混合物在室温下搅拌1小时后加水(20mL)稀释,并用二氯甲烷(30mL×2)萃取。将合并的有机相用无水硫酸钠干燥,过滤,减压浓缩得到粗产品,柱层析纯化得类白色固体1-(5-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(乙基)氨基)-2-乙基-2H-吡唑并[4,3-b]吡啶-5-基)嘧啶-2-基)氮杂环丁烷-3-基乙酸酯(100mg)。
1H NMR(400MHz,CDCl 3):δ9.02(s,2H),8.18-8.09(m,3H),7.64(d,J=9.3Hz,1H),7.20-7.14(m,2H),5.38-5,34(m,1H),4.57-4.52(m,3H),4.41(q,J=7.1Hz,2H),4.19-4.15(m,2H),3.90-3.86(m,1H),2.14(s,3H),1.66(t,J=7.4Hz,3H),1.47(t,J=7.1Hz,3H)。
MS实测值(ESI +)[(M+H) +]:584。
实施例26:2-(乙基(2-乙基-5-(2-(4-羟基哌啶-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物26)
Figure PCTCN2021105180-appb-000157
步骤26a:制备1-(5-溴嘧啶-2-基)哌啶-4-醇
Figure PCTCN2021105180-appb-000158
步骤26a的具体方式:将2,5-二溴嘧啶(440mg,1.85mmol),4-羟基哌啶(206mg,2.03mmol)和碳酸钾(511mg,3.7mmol)按照实施例23步骤23a的方法制备得类白色固体1-(5-溴嘧啶-2-基)哌啶-4-醇(350mg)。
步骤26b:制备(2-(4-羟基哌啶-1-基)嘧啶-5-基)硼酸
Figure PCTCN2021105180-appb-000159
步骤26b的具体方式:氮气氛围中,将1-(5-溴嘧啶-2-基)哌啶-4-醇(110mg,0.43mmol)和联硼酸频那醇酯(130mg,0.51mmol)按照实施例23步骤23b的方法制备得到(2-(4-羟基哌啶-1-基)嘧啶-5-基)硼酸(95mg)。
步骤26c:制备2-(乙基(2-乙基-5-(2-(4-羟基哌啶-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000160
步骤26c的具体方式:氮气氛围中,将(2-(4-羟基哌啶-1-基)嘧啶-5-基)硼酸(95mg,0.43mmol)和2-((5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(205mg,0.43mmol)按照实施例23步骤23c的方法制备得类白色固体2-(乙基(2-乙基-5-(2-(4-羟基哌啶-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(50mg)。
1H NMR(400MHz,CDCl 3):δ9.00(s,2H),8.17-8.07(m,3H),7.64(d,J=9.3Hz,1H),7.19-7.15(m,2H),4.55-4.35(m,5H),4.03-3.85(m,2H),3.47-3.41(m,2H),2.00-1.95(m,2H),1.66(t,J=7.2Hz,3H),1.59-1.52(m,2H),1.47(t,J=7.2Hz,3H)。
MS实测值(ESI +)[(M+H) +]:570。
实施例27:(S)-2-(乙基(2-乙基-5-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物27)
Figure PCTCN2021105180-appb-000161
步骤27a:制备(S)-1-(5-溴嘧啶-2-基)吡咯烷-3-醇
Figure PCTCN2021105180-appb-000162
步骤27a的具体方式:室温下,将2,5-二溴嘧啶(1g,4.2mmol)的DMF溶液(8mL)中加入(S)-吡咯烷-3-醇(0.4g,4.7mmol)按照实施例23步骤23a的方法制备得到白色固体(S)-1-(5-溴嘧啶-2-基)吡咯烷-3-醇(800mg)。
步骤27b:制备(S)-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)硼酸
Figure PCTCN2021105180-appb-000163
步骤27b的具体方式:氮气氛围中,将(S)-1-(5-溴嘧啶-2-基)吡咯烷-3-醇(200mg,0.82mmol)和联硼酸频那醇酯(230mg,0.9mmol)按照实施例23步骤23b的方法制备得黑色固体(S)-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)硼酸(400mg)。
步骤27c:制备(S)-2-(乙基(2-乙基-5-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000164
步骤27c的具体方式:氮气氛围中,将(S)-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)硼酸(200mg,0.96mmol)和2-((5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(200mg,0.42mmol)按照实施例23步骤23c的方法制备得白色固体(S)-2-(乙基(2-乙基-5-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(100mg)。
1H NMR(400MHz,DMSO-d 6):δ9.04(br.s,2H),8.16-8.11(m,3H),7.65(d,J=8.8Hz,1H),7.19(t,J=8.8Hz,2H),4.66(s,1H),4.61-4.45(m,1H),4.44-4.39(m,2H),4.01-3.92(m,1H),3.85-3.81(m,2H),3.78-3.73(m,2H),2.24-2.10(m,2H),1.78(d,J=2.8Hz,1H),1.68(t,J=7.2Hz,3H),1.49(t,J=7.2Hz,3H)。
MS实测值(ESI +)[(M+H) +]:556。
实施例28:(R)-2-(乙基(2-乙基-5-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物28)
Figure PCTCN2021105180-appb-000165
步骤28a:制备(R)-1-(5-溴嘧啶-2-基)吡咯烷-3-醇
Figure PCTCN2021105180-appb-000166
步骤28a的具体方式:室温下,将5-溴-2-氯嘧啶(500mg,2.10mmol)和(R)-吡咯烷-3-醇(210mg,2.42mmol)按照实施例23步骤23a的方法制备得白色固体(R)-1-(5-溴嘧啶-2-基)吡咯烷-3-醇(472mg)。
步骤28b:制备(R)-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)硼酸
Figure PCTCN2021105180-appb-000167
步骤28b的具体方式:将(R)-1-(5-溴嘧啶-2-基)吡咯烷-3-醇(200mg,0.82mmol)和联硼酸频那醇酯(312mg,1.23mmol)按照实施例23步骤23b的方法制备得(R)-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)硼酸(171mg)。
步骤28c:制备(R)-2-(乙基(2-乙基-5-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000168
步骤28c的具体方式:将2-((5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(100mg,0.21mmol)和(R)-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)硼酸(67mg,0.32mmol)按照实施例23步骤23c的方法制备得白色固体(R)-2-(乙基(2-乙基-5-(2-(3-羟基吡咯烷-1-基)嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(56mg)。
1H NMR(400MHz,CDCl 3):δ9.03(s,2H),8.25-7.98(m,3H),7.64(d,J=9.0Hz,1H),7.17(t,J=8.4Hz,2H),4.65(br.s.,1H),4.53(br.s.,1H),4.40(d,J=7.0Hz,2H),3.90-3.64(m,5H),2.19-2.05(m,2H),1.67(t,J=7.1Hz,3H),1.47(t,J=7.1Hz,3H)。
MS实测值(ESI +)[(M+H) +]:556。
实施例29:2-(乙基(2-乙基-5-(2-吗啉基嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物29)
Figure PCTCN2021105180-appb-000169
步骤29a:制备4-(5-溴嘧啶-2-基)吗啉
Figure PCTCN2021105180-appb-000170
步骤29a的具体方式:室温下,将5-溴-2-氯嘧啶(500mg,2.10mmol)和吗啉(200mg,2.3mmol)按照实施例23步骤23a的方法制备得白色固体4-(5-溴嘧啶-2-基)吗啉(400mg)。
步骤29b:制备(2-吗啉基嘧啶-5-基)硼酸
Figure PCTCN2021105180-appb-000171
步骤29b的具体方式:将(4-(5-溴嘧啶-2-基)吗啉(200mg,0.82mmol)和联硼酸频那醇酯(312mg, 1.23mmol)按照实施例23步骤23b的方法制备得(2-吗啉基嘧啶-5-基)硼酸(180mg)。
步骤29c:制备2-(乙基(2-乙基-5-(2-吗啉基嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈
Figure PCTCN2021105180-appb-000172
步骤29c的具体方式:将2-((5-溴-2-乙基-2H-吡唑并[4,3-b]吡啶-3-基)(乙基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(100mg,0.21mmol)和(2-吗啉基嘧啶-5-基)硼酸(84mg,0.40mmol)按照实施例23步骤23c的方法制备得白色固体2-(乙基(2-乙基-5-(2-吗啉基嘧啶-5-基)-2H-吡唑并[4,3-b]吡啶-3-基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(60mg)。
1H NMR(400MHz,CDCl 3):δ9.03(s,2H),8.15–8.11(m,3H),7.64(d,J=9.2Hz,1H),7.17(t,J=8.4Hz,2H),4.40(q,J=7.2Hz,2H),3.91-3.88(m.,5H),3.79-3.77(m,5H),1.66(t,J=7.2Hz,3H),1.47(t,J=7.2Hz,3H)。
MS实测值(ESI +)[(M+H) +]:556。
以下具体描述了生物学实施数据,以进一步阐述本发明技术方案。
实验例1:ATX酶活性抑制试验
实验步骤:
受试化合物用DMSO溶解成20mM储备液,用DMSO进行4倍梯度稀释,起始浓度为20μM,共8个浓度梯度。使用Echo550仪器将20nL稀释后的化合物转移到384-孔板,并向孔中加入5μL使用4×Tris-HCl反应缓冲液配置的ATX酶溶液,然后将384-孔板1000rpm离心1分钟并在室温预孵育15分钟。随后向每孔加入5μL的上述4×反应缓冲液配制的16:0-LPC和10μL使用同样反应缓冲液配置的含有2×Amplex UltraRed试剂、胆碱氧化酶和辣根过氧化物酶(HRP)的检测液,然后将384-孔板1000rpm离心1分钟。使用Synergy 2仪器读取激发光530nm,发射光590nm的荧光信号。根据荧光比值计算化合物对酶反应的抑制率,用GraphPad Prism 5软件分析计算出化合物的IC 50
检测结果:
按上述方法测定本发明的实施例1至实施例29所涉及的化合物以及GLPG-1690对于抑制ATX的活性,结果总结在表3中。
表3:实施例化合物的活性数据
实施例 IC 50(μM) 实施例 IC 50(μM)
1 0.030 2 0.029
3 0.0052 4 0.021
5 0.048 6 0.029
7 0.025 8 0.0044
9 0.028 10 0.018
11 0.025 12 0.019
13 0.013 14 0.042
15 0.040 16 0.039
17 0.0074 18 0.015
19 0.017 20 0.045
21 0.011 23 0.011
24 0.011 25 0.012
26 0.0055 27 0.0046
28 0.0038 29 0.023
GLPG1690 0.112    
结论:
由表3可知,本发明中的实施例化合物对ATX酶抑制活性均优于GLPG1690。
实验例2:大鼠药代动力学试验
对本发明中多个实施例的化合物进行SD大鼠的代谢动力学实验,分别采取单次静脉注射(IV,剂量2mg/kg)和单次口服给药(PO,剂量10mg/kg),分别于给药前及给药后5分钟、15分钟、30分钟、1小时、2小时、4小时、8小时、12小时和24小时采血测定血浆中待测化合物的浓度。对照化合物及本发明中实施例17、26、28和29在大鼠血浆中的药代动力学数据如表4-8所示。
结果显示,实施例17、26、28和29在大鼠中单次静脉注射和单次口服给药后的体内暴露量均高于GLPG1690,且清除率更低。
表4:对照化合物GLPG1690的大鼠体内PK参数
Figure PCTCN2021105180-appb-000173
表5:实施例17化合物的大鼠体内PK参数
Figure PCTCN2021105180-appb-000174
表6:实施例26化合物的大鼠体内PK参数
Figure PCTCN2021105180-appb-000175
表7:实施例28化合物的大鼠体内PK参数
Figure PCTCN2021105180-appb-000176
表8:实施例29化合物的大鼠体内PK参数
Figure PCTCN2021105180-appb-000177
实施例3:中国仓鼠卵巢细胞体外微核试验
考察实施例26化合物和GLPG1690在加或不加外源性代谢活化系统(β-萘黄酮和苯巴比妥诱导的大鼠肝脏S9)条件下诱导中国仓鼠卵巢细胞(CHO-WBL)形成微核的能力,评价其致染色体断裂/诱导非整倍体的潜力。
微核主试验将CHO-WBL细胞暴露于受试物的至少3个浓度下,同时设立溶媒和阳性对照,每个剂量组均设2孔细胞。在非活化测试系统中,给药时间为3和24小时,在S9活化测试系统中,受试物暴露时间为3小时。
受试物测试的上限取决于它在培养基中的溶解度,但总的来说不会超过最大浓度1mM或0.5mg/mL,取较低者。选择微核频率分析的最高剂量时,该受试物剂量组与相应溶媒对照组相比细胞毒性应不超过50%太多。若最高浓度不受限于细胞毒性,为了达到培养基中溶解度上限,可以测试多个含有可见沉淀的剂量,且选作微核分析的最高剂量组培养基中可见少量可识别的沉淀。
在试验有效前提下,根据以下标准评估试验结果:
1.阳性
若受试物同时满足以下标准,可认为该受试物为阳性:
在一个或多个浓度组中观察到微核细胞频率显著性增加。
在一个或多个浓度组中观察到微核细胞频率超出试验室阴性历史数据范围。
微核频率的增加具有剂量效应关系。
2.阴性
若以上三点均不符合,可认为该受试物为阴性。
Figure PCTCN2021105180-appb-000178
Figure PCTCN2021105180-appb-000179
与平行溶媒对照组相比,实施例26化合物各给药系列中所分析的浓度含有微核的双核细胞频率未见显著增加。也未见微核率有剂量效应关系。而阳性对照(环磷酰胺一水合物和丝裂霉素C)含有微核的双核细胞频率均显著增加,证明了实验系统的有效性。实施例26化合物在CHO-WBL细胞中诱导微核细胞的潜能为阴性。
与平行溶媒对照组相比,GLPG1690在非代谢活化给药24小时系列中所分析的AKEX0070浓度为7和8μg/mL时含有微核的双核细胞频率显著增加,且可见微核率增加有剂量效应关系。而阳性对照(环磷酰胺一水合物和丝裂霉素C)含有微核的双核细胞频率均显著增加,证明了实验系统的有效性。GLPG1690在CHO-WBL细胞中诱导微核细胞的潜能为阳性。
基于以上结果,GLPG1690具有遗传毒性,实施例26化合物具有比GLPG1690更优的安全性。
实验例4:CYP抑制试验
CYP450酶单浓度点抑制试验:用体系为320μL的人肝微粒体(对于3A4亚型,终浓度为0.05mg/mL)进行直接抑制温孵,体系含NADPH(终浓度1.3mM)、10μM化合物、阳性抑制剂(酮康唑0.1μM)、阴性对照(0.1%DMSO的BPS)和混合探针底物(咪达唑仑5μM),温孵5min后终止反应。通过测定代谢物的相对生成量计算酶相对活性。
按上述方法,对照化合物GLPG1690及本发明中实施例17、23、26、27化合物在10μM下CYP450酶单点抑制数据如下表所示。结果显示,实施例17、23、26、27对3A4亚型的抑制活性低于GLPG1690,临床中药物-药物相互作用的风险更低。
化合物 3A4亚型
GLPG1690 73.5
实施例17 18.6
实施例23 39.0
实施例26 31.1
实施例27 47.7
实施例5:hERG抑制试验
快速激活人延迟整流外向钾电流(I Kr)主要由hERG离子通道介导,参与人类心肌细胞复极化。药物阻断这一电流将导致临床上出现QT间期延长综合征,易诱发急性心律失常甚至猝死。本研究应用手动膜片钳的方法在转染hERG钾通道的稳定细胞株上测试多个实施例化合物对hERG钾电流的作用,从而确定受试物是否对hERG离子通道具有抑制作用。
实验方法:采用全细胞膜片钳技术记录hERG电流。取细胞悬液加于小培养皿中,置于倒置显微镜载物台上。待细胞贴壁后,用细胞外液灌流,推荐流速为1–2mL/min。玻璃微电极由微电极拉制仪两步拉制,充灌电极内液后其入水电阻值为2-5MΩ。建立全细胞记录模式后,保持钳制电位为-80mV。给予去极化电压至+60mV持续850ms,然后复极化至-50mV维持1275ms引出hERG尾电流。这样一组脉冲程序每15秒钟重复一次,贯穿整个实验。电流稳定后采用从低浓度到高浓度胞外连续灌流给药的方式。从低浓度开始,持续灌流至药效稳定,然后进行下一浓度的灌流。本实验将分别测试各浓度供试品和阳性对照对hERG尾电流的阻断效应(N≥2)。随后通过PatchMaster软件进行刺激发放及信号采集;膜片钳放大器放大信号。使用FitMaster,EXCEL,Graphpad Prism和SPSS 21.0等进行进一步数据分析和曲线拟合。在数据处理中,判断对hERG的阻断效应时,将尾电流的峰值和其基线进行校正。用尾流的抑制率(inhibition rate,IR)表示不同浓度下各化合物的作用。
IR=100%×(给药前尾电流峰值-给药后尾电流峰值)/给药前尾电流峰值。
IC 50数值由Hill方程进行拟合所得(如果适用),若受试物所有浓度的最大抑制率小于50%,则不计算IC 50数值。
Figure PCTCN2021105180-appb-000180
其中:
y:I/I control;max:为100%;min:为0%;[drug]:供试品浓度;n H:Hill斜率;IC 50:测试物的最大半数抑制浓度
按上述方法,对照化合物及本发明中实施例17、26、27和29对hERG电流抑制作用的IC 50数据如下表所示。结果显示,对照化合物GLPG1690和实施例17、26、27和29对hERG电流抑制作用均大于30μM,均无抑制作用。
化合物 hERG IC 50[μM]
GLPG1690 >30
实施例17 >30
实施例26 >30
实施例27 >30
实施例29 >30
实施例6:3A4时间依赖性抑制试验
CYP450酶3A4亚型睾酮底物时间依赖性抑制实验(IC 50shift):用体系为100μL的人肝微粒体(终浓度为0.2mg/mL)加或不加NADPH预孵育30min后再补加20μL NADPH进行抑制温孵,体系NADPH终浓度1.3mM。化合物系列浓度从100μM起始,3倍稀释共8个浓度点(包含0浓度点)。阳性抑制剂为米非司酮,系列浓度从10μM起始,3倍稀释共8个浓度点(包含0浓度点)。探针底物为睾酮,浓度50μM。体系温孵10min后终止反应。通过测定代谢物的相对生成量计算化合物对酶抑制率的IC 50,通过加与不加NADPH孵育组的IC 50结果计算IC 50shift值。
按上述方法,本发明中实施例4、17、23、26、27、28和29化合物对CYP450酶3A4时间依懒性抑制数据如下表所示。结果显示,实施例4、17、23、26、27、28和29化合物对CYP3A4的时间依赖性抑制作用均弱于GLPG1690,临床中药物-药物相互作用的风险更低。
化合物 IC 50 shift
实施例4 1.04
实施例17 IC 50>100μM/IC 50>100μM
实施例23 IC 50>100μM/IC 50>100μM
实施例26 1.09
实施例27 0.63
实施例28 1.28
实施例29 IC 50>100μM/IC 50>100μM
GLPG1690 1.64
进一步地,本领域技术人员理解,前述涉及的式I’、式I、式I-1、式II、式II-1、式III或式III-1所示化合物、式I’、式I、式I-1、式II、式II-1、式III或式III-1所示化合物不同的实现方式以及式I’、式I、式I-1、式II、式II-1、式III或式III-1所示化合物的具体实施例所涉及的全部化合物,均可以被制成对应的异构体、溶剂化物、水合物、前药、稳定的同位素衍生物及药学上可接受的盐。优选地,所述化合物被制成药学上可接受的衍生物,所述衍生物为前药、盐、酯、酰胺、酯类的盐、酰胺类的盐、代谢产物中的任一种。
进一步地,药学上可接受的盐包括通过用无机酸(例如盐酸、氢溴酸、硫酸或者磷酸),或者用有机酸(例如乙酸、丙酸、琥珀酸、苯甲酸、对氨基苯磺酸、2-乙酰氧基-苯甲酸、肉桂酸、苦杏仁酸、水杨酸、乙醇酸、乳酸、草酸、苹果酸、马来酸、丙二酸、富马酸、酒石酸、柠檬酸、对甲苯磺酸、甲磺酸、乙磺酸或苯磺酸)使本发明涉及的任一化合物成盐获得的常规非毒性盐。对于合适的药用盐的综述可以参考Berge S.M等,J.Pharm.Sci.1977,66,1-19;Gould P.L.Int.J.Pharm 1986,33,201-277以及Bighley等,药学技术百科全书,Marcel Dekker Inc,纽约1996,第13卷,第453-497页。
进一步地,稳定的同位素衍生物,可以将同位素引入本发明涉及的任一化合物中,引入的同位素可以是 2H, 3H, 13C, 14C, 15N, 17O, 18O, 31P, 32P, 35S, 18F, 36Cl,具体的同位素衍生物可以通过常规技术制备。
进一步地,其作为实际产品还可以被制成片剂、胶囊剂、注射剂、颗粒剂、粉剂、栓剂、丸剂、 乳膏剂、糊剂、凝胶剂散剂、口服溶液、吸入剂、混悬剂、干悬剂、贴剂、洗剂中的任一种。
进一步地,在上述基础上,还可以与以下任一物质形成混合物:药学上可接受的载体或者辅剂或者赋形剂。
本发明所涉及的全部化合物及包含本发明化合物的混合物、组合物等,可以经任一给药途径给予到生物体内。给药途径可以是口服给药,静脉注射,肌肉注射,皮下注射,直肠给药,阴道给药,舌下含化,鼻腔吸入,口腔吸入,滴眼,也可局部或全身经皮给药。
本发明所涉及的全部化合物及包含本发明化合物的混合物、组合物等,可以配制成单一剂量,其中含有本发明的活性化合物以及载体、赋形剂等,给药剂型可以是片剂,胶囊剂,注射剂,颗粒剂,粉剂,栓剂,丸剂,乳膏剂,糊剂,凝胶剂,散剂,口服溶液,吸入剂,混悬剂,干悬剂,贴剂,洗剂等。这些剂型中可以含有药物制剂常用的成分,例如稀释剂,吸收剂,润湿剂,粘合剂,崩解剂,着色剂,pH调节剂,抗氧剂,抑菌剂,等渗调节剂,抗粘剂等。
上述各类剂型的合适配方可从公开途径获得,例如Remington:The Science and Practice of Pharmacy,第21版,Lippincott Williams&Wilkins于2006年出版和Rowe,Raymond C.Handbook of Pharmaceutical Excipients,Chicago,Pharmaceutical Press于2005年出版.因此本领域的技术人员可以容易的制备。
根据不同个体所患疾病的性质,强度,患者的年龄、性别、体重,给药途径等因素,可以选择不同的给药剂量,本发明的化合物的给药剂量可以为每日0.01至500mg/kg,优选每日剂量为1-100mg/kg,可单次或多次给药。
本领域技术人员理解,作为本发明的所涉及的全部化合物及包含本发明化合物的混合物、组合物等,其典型的应用为医学应用,特别是用于预防或治疗哺乳动物具有ATX表达增加的病理学特征的疾病,具体的适应症如下:
癌症、纤维化疾病、代谢疾病、骨髓增生异常综合征、呼吸系统疾病、心血管疾病、自身免疫性疾病、炎症、皮肤学疾病、神经系统疾病或疼痛。
以上对本发明的具体实施例进行了描述。需要理解的是,本发明并不局限于上述特定实施方式,本领域技术人员可以在权利要求的范围内做出各种变形或修改,这并不影响本发明的实质内容。

Claims (25)

  1. 式I’化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物:
    Figure PCTCN2021105180-appb-100001
    其中,
    n为0至5中的任一整数;
    若存在,每一个R 1各自独立地选自氢、氘、氰基、卤素、氨基、羟基、-COOH、-CHO、-NO 2、C 1-6烷基氨基、C 1-6烷氧基和未取代或被选自氰基、氨基、羟基、卤素、C 1-6烷基和C 1-6烷氧基的取代基所取代的C 1-6烷基、C 3-7环烷基和3-7元杂环烷基;
    R 2选自氢、氘、卤素、氰基、氨基、羟基、-COOH、-CHO、-NO 2、C 1-6烷基、C 1-6烷氧基、C 1-6烷基氨基、-C(=O)NH 2、-NH(C=O)CH 3和3-7元杂环烷基;
    R 3选自氢、氘、C 1-6烷基、C 3-7环烷基和被选自氰基、氨基、羟基、卤素、C 1-6烷基和C 1-6烷氧基的取代基所取代的C 1-6烷基、C 3-7环烷基和3-7元杂环烷基;
    环B为下列结构的任一种:
    Figure PCTCN2021105180-appb-100002
    R 4选自氢、氘、卤素、氰基、羟基、氨基、C 1-6烷基、C 1-6烷氧基、C 1-6烷基氨基、C 3-7环烷基和3-7元杂环烷基;
    X 1、X 2、X 3和X 4各自独立地为N或CR 5
    R 5选自氢、氘、卤素、C 1-6烷基和被选自氰基、氨基、羟基、卤素、C 1-6烷基和C 1-6烷氧基的取代基所取代的C 1-6烷基、C 3-7环烷基和3-7元杂环烷基;
    环A选自取代或未取代的C 6-10芳基和5-10元杂芳基,所述取代是被1至3个选自羟基、氨基、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3-7元杂环烷基、C 1-6烷基氨基、C 1-6烷氧基的取代基所取代;
    R 6为-L 1-L 2-W 1
    L 1选自化学键、C 1-3亚烷基、-O-、-C(=O)-、-C(=O)C(R 8) 2-、-C(=O)O-、-C(=O)NH-、-OC(=O)-、-NH-、-SO 2-、-NHSO 2-和-SO 2NH-;其中,R 8为氢或C 1-6烷基;
    L 2选自化学键、-O-、-C(=O)-、-C(=O)CH 2-、-C(=O)O-、-OC(=O)-、-C(=O)NH-、-NR 7-、-NHCH 2-、-SO 2-、-NR 7SO 2-、-SO 2NR 7-、-C(=O)NR 7-和-NR 7C(=O)-;其中,R 7为氢或C 1-6烷基;
    W 1为取代或未取代的下列基团:氢、氘、氨基、氰基、C 1-6烷基、卤素、羟基、C 1-6烷氧基、C 1- 6烷基氨基、羧基、C 6-10芳基、5-10元杂芳基、C 3-7环烷基或3-7元杂环烷基;所述取代是被1或2个选自氧代、羟基、氨基、羟甲基、氨基甲基、氰基、C 1-6烷基、C 1-6烷基-C(=O)O-和卤素的取代基所取代。
  2. 如权利要求1所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标 记物,其特征在于,
    n为0至2中的任一整数,优选n为1;
    若存在,每一个R 1各自独立地选自氢、氰基和卤素,优选卤素,更优选氟。
  3. 如权利要求1或2所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,
    R 2为氰基。
  4. 如权利要求1至3中任一项所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,
    R 3为C 1-6烷基,优选甲基或乙基。
  5. 如权利要求1至4中任一项所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,
    环B为下列结构的任一种:
    Figure PCTCN2021105180-appb-100003
    X 1、X 2、X 3、X 4各自独立地为N或CR 5
    R 5选自氢、氘、卤素、甲基和卤代甲基,优选氢;
    R 4选自氢、氘、甲基、乙基、异丙基和环丙基,优选乙基。
  6. 如权利要求1至5中任一项所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,
    环A选自亚苯基、亚吡啶基、亚吡嗪基、亚哒嗪基、亚嘧啶基、亚吡唑基、亚咪唑基、亚噁唑基、亚异噁唑基、亚噻唑基、亚噻二唑基和亚噁二唑基;
    优选地,环A选自下列结构的任一种:
    Figure PCTCN2021105180-appb-100004
    Figure PCTCN2021105180-appb-100005
    更优选地,环A选自下列结构中的任一种:
    Figure PCTCN2021105180-appb-100006
  7. 如权利要求6所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,
    环A被1至3个,优选1个取代基所取代,所述取代基各自独立地选自羟基、氨基、卤素、氰基和C 1-6烷基,优选卤素和C 1-6烷基,更优选氟和甲基。
  8. 如权利要求1至7中任一项所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,
    L 1选自化学键、-CH 2-、-CH(CH 3)-、-C(CH 3) 2-、-C(=O)-、-SO 2-、-C(=O)CH 2-和-C(=O)NH-,优选化学键、-CH 2-、-C(CH 3) 2-和-C(=O)CH 2-。
  9. 如权利要求1至8中任一项所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,
    L 2选自化学键、-NH-、-C(=O)-、-SO 2-、-NHCH 2-、-C(=O)CH 2-、-C(=O)NH-、-NHSO 2-和-N(CH 3)SO 2-,优选化学键、-C(=O)-、-NHCH 2-、-C(=O)NH-、-NHSO 2-和-N(CH 3)SO 2-。
  10. 如权利要求1至9中任一项所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,
    W 1为取代或未取代的下列基团:W 1为取代或未取代的下列基团:C 1-6烷基、C 6-10芳基、5-10元杂芳基、C 3-7环烷基或3-7元杂环烷基,所述取代是被1或2个选自氧代、羟基、氨基、羟甲基、氨基甲基、卤素、氰基、C 1-6烷基-C(=O)O-和C 1-6烷基的取代基所取代。
  11. 如权利要求10所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,
    W 1为未取代或被1或2个选自羟基、氨基、羟甲基、氨基甲基和C 1-6烷基-C(=O)O-的取代基所取代的C 1-6烷基、C 3-7环烷基或3-7元杂环烷基。
  12. 如权利要求11所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,
    W 1为未取代或被1或2个选自羟基、氨基、羟甲基、氨基甲基和乙酰氧基的取代基所取代的下列基团:甲基、乙基、正丙基、异丙基、环丙基、环丁基、环戊基、环己基、吖丙啶基、吖丁啶基、四氢吡咯基、氧代吡咯烷基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、氧杂环丁基、四氢呋喃基、四氢噻吩基、四氢吡喃基、1,1-二氧硫代吗啉基、2-氮杂螺[3.3]庚基、1,1-二氧化异噻唑烷基或1,1-二氧化-1,2-噻嗪烷基,优选为甲基、氧代吡咯烷基、2-氮杂螺[3.3]庚基、1,1-二氧化异噻唑烷基、1,1-二氧 化-1,2-噻嗪烷基或被选自羟基、羟甲基和乙酰氧基的取代基所取代的乙基、吖丁啶基、环丁基、四氢吡咯基或哌啶基。
  13. 如权利要求1至12中任一项所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,
    n为1;
    R 1为氟;
    R 2为氰基;
    R 3为甲基或乙基;
    环B为下列结构的任一种:
    Figure PCTCN2021105180-appb-100007
    R 4为乙基;
    X 1、X 2、X 3和X 4各自独立地为N或CH;
    环A选自下列结构的任一种:
    Figure PCTCN2021105180-appb-100008
    环A未取代或被1个氟或甲基所取代;
    R 6为-L 1-L 2-W 1
    L 1选自化学键、-CH 2-、-C(CH 3) 2-和-C(=O)CH 2-;
    L 2选自化学键、-C(=O)-、-NHCH 2-、-C(=O)NH-、-NHSO 2-和-N(CH 3)SO 2-;
    W 1为下列基团中的任一种:
    Figure PCTCN2021105180-appb-100009
  14. 如权利要求1所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,所述化合物为具备式I结构的化合物,
    Figure PCTCN2021105180-appb-100010
    其中,n、R 1、R 2、R 3、R 4、R 6、X 1、X 2、X 3和环A如权利要求1所定义。
  15. 如权利要求1所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,所述化合物为具备式I-1结构的化合物,
    Figure PCTCN2021105180-appb-100011
    其中,n、R 1、R 2、R 3、R 4、R 6和环A如权利要求1所定义。
  16. 如权利要求1所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,所述化合物为具备式II结构的化合物,
    Figure PCTCN2021105180-appb-100012
    其中,n、R 1、R 2、R 3、R 4、R 6、X 1、X 2、X 3和环A如权利要求1所定义。
  17. 如权利要求1所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,所述化合物为具备式II-1结构的化合物,
    Figure PCTCN2021105180-appb-100013
    其中,n、R 1、R 2、R 3、R 4、R 6和环A如权利要求1所定义。
  18. 如权利要求1所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,所述化合物为具备式III结构的化合物,
    Figure PCTCN2021105180-appb-100014
    其中,n、R 1、R 2、R 3、R 4、R 6、X 1、X 2、X 3和环A如权利要求1所定义。
  19. 如权利要求1所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物,其特征在于,所述化合物为具备式III-1结构的化合物,
    Figure PCTCN2021105180-appb-100015
    其中,n、R 1、R 2、R 3、R 4、R 6和环A如权利要求1所定义。
  20. 下列化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物:
    Figure PCTCN2021105180-appb-100016
    Figure PCTCN2021105180-appb-100017
    Figure PCTCN2021105180-appb-100018
    Figure PCTCN2021105180-appb-100019
  21. 一种药物组合物,其包含如权利要求1至20中任一项所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物。
  22. 一种药物制剂,其包含如权利要求1至20中任一项所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物或者如权利要求15所述的药物组合物;
    优选地,所述制剂为片剂、胶囊剂、注射剂、颗粒剂、粉剂、栓剂、丸剂、乳膏剂、糊剂、凝胶剂、散剂、口服溶液、吸入剂、混悬剂、干悬剂、贴剂、洗剂中的任一种。
  23. 如权利要求1至20中任一项所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物或者如权利要求21所述的药物组合物或者如权利要求22所述的药物制剂在制备用于预防和/或治疗具有ATX表达增加的病理学特征的相关疾病的药物中的应用;
    优选地,所述具有ATX表达增加的病理学特征的相关疾病包括:癌症、纤维化疾病、代谢疾病、骨髓增生异常综合征、呼吸系统疾病、心血管疾病、自身免疫性疾病、炎症、皮肤学疾病、神经系统疾病或疼痛;
    更优选地;所述具有ATX表达增加的病理学特征的相关疾病为肺纤维化、肾纤维化或肝纤维化。
  24. 如权利要求1至20中任一项所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物或者如权利要求21所述的药物组合物或者如权利要求22所述的药物制剂,其用 于预防和/或治疗具有ATX表达增加的病理学特征的相关疾病;
    优选地,所述具有ATX表达增加的病理学特征的相关疾病包括:癌症、纤维化疾病、代谢疾病、骨髓增生异常综合征、呼吸系统疾病、心血管疾病、自身免疫性疾病、炎症、皮肤学疾病、神经系统疾病或疼痛;
    更优选地,所述具有ATX表达增加的病理学特征的相关疾病为肺纤维化、肾纤维化或肝纤维化。
  25. 一种预防和/或治疗具有ATX表达增加的病理学特征的相关疾病的方法,其包括下列步骤:将有效量的如权利要求1至20中任一项所述的化合物或其药学上可接受的盐、酯、异构体、溶剂化物、前药或同位素标记物或者如权利要求21所述的药物组合物或者如权利要求22所述的药物制剂施用于对其有需求的患者。
PCT/CN2021/105180 2020-07-09 2021-07-08 一种atx抑制剂及其制备方法和应用 WO2022007882A1 (zh)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP21838673.8A EP4180430A4 (en) 2020-07-09 2021-07-08 ATX INHIBITOR AND PRODUCTION METHOD THEREOF AND USE THEREOF
CN202180046308.8A CN115916777A (zh) 2020-07-09 2021-07-08 一种atx抑制剂及其制备方法和应用
US18/015,261 US20230295159A1 (en) 2020-07-09 2021-07-08 Atx inhibitor, and preparation method therefor and use thereof
JP2023501105A JP2023533294A (ja) 2020-07-09 2021-07-08 Atx阻害剤及びその製造方法並びに使用

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202010658085.8A CN113912600A (zh) 2020-07-09 2020-07-09 一种atx抑制剂及其制备方法和应用
CN202010658085.8 2020-07-09
CN202110308656.X 2021-03-23
CN202110308656 2021-03-23

Publications (1)

Publication Number Publication Date
WO2022007882A1 true WO2022007882A1 (zh) 2022-01-13

Family

ID=79552799

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/105180 WO2022007882A1 (zh) 2020-07-09 2021-07-08 一种atx抑制剂及其制备方法和应用

Country Status (6)

Country Link
US (1) US20230295159A1 (zh)
EP (1) EP4180430A4 (zh)
JP (1) JP2023533294A (zh)
CN (1) CN115916777A (zh)
TW (1) TW202202499A (zh)
WO (1) WO2022007882A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023185979A1 (zh) * 2022-03-31 2023-10-05 苏州爱科百发生物医药技术有限公司 一种atx抑制剂或其盐的结晶形式及其制备方法与应用
US11970493B2 (en) 2020-10-06 2024-04-30 Ildong Pharmaceutical Co., Ltd. Autotaxin inhibitor compounds

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014202763A1 (en) * 2013-06-20 2014-12-24 Ab Science Benzimidazole derivatives as selective proteine kinase inhibitors
CN105143221A (zh) * 2013-03-14 2015-12-09 加拉帕戈斯股份有限公司 用于治疗炎性障碍的化合物及其药物组合物
CN105339370A (zh) * 2013-06-19 2016-02-17 加拉帕戈斯股份有限公司 用于治疗炎症性疾病的新化合物和及其药物组合物
CN109384803A (zh) * 2017-08-09 2019-02-26 广州市恒诺康医药科技有限公司 Atx抑制剂及其制备方法和应用
WO2019158107A1 (zh) * 2018-02-14 2019-08-22 苏州信诺维医药科技有限公司 一种杂环化合物、其应用及含其的药物组合物
WO2019228403A1 (en) * 2018-05-29 2019-12-05 Fronthera U.S. Pharmaceuticals Llc Autotaxin inhibitors and uses thereof
WO2020177729A1 (zh) * 2019-03-06 2020-09-10 江苏恒瑞医药股份有限公司 稠合芳香环类衍生物、其制备方法及其在医药上的应用

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105143221A (zh) * 2013-03-14 2015-12-09 加拉帕戈斯股份有限公司 用于治疗炎性障碍的化合物及其药物组合物
CN105339370A (zh) * 2013-06-19 2016-02-17 加拉帕戈斯股份有限公司 用于治疗炎症性疾病的新化合物和及其药物组合物
WO2014202763A1 (en) * 2013-06-20 2014-12-24 Ab Science Benzimidazole derivatives as selective proteine kinase inhibitors
CN109384803A (zh) * 2017-08-09 2019-02-26 广州市恒诺康医药科技有限公司 Atx抑制剂及其制备方法和应用
WO2019158107A1 (zh) * 2018-02-14 2019-08-22 苏州信诺维医药科技有限公司 一种杂环化合物、其应用及含其的药物组合物
WO2019228403A1 (en) * 2018-05-29 2019-12-05 Fronthera U.S. Pharmaceuticals Llc Autotaxin inhibitors and uses thereof
WO2020177729A1 (zh) * 2019-03-06 2020-09-10 江苏恒瑞医药股份有限公司 稠合芳香环类衍生物、其制备方法及其在医药上的应用

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
"Remington: The Science and Practice of Pharmacy", 2006, LIPPINCOTT WILLIAMS & WILKINS
BERGE S. M. ET AL., J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
BIGHLEY ET AL.: "Encyclopedia of Pharmaceutical Technology", vol. 13, 1996, MARCEL DEKKER INC., pages: 453 - 497
GOULD P. L., INT. J. PHARM., vol. 33, 1986, pages 201 - 277
JONCOUR, A. ET AL.: "Discovery, Structure-Activity Relationship, and Binding Mode of an Imidazo[l, 2-a]pyridine Series of Autotaxin Inhibitors", JOURNAL OF MEDICINAL CHEMISTRY, vol. 60, 21 July 2017 (2017-07-21), pages 7371 - 7392, XP055517662, DOI: 10.1021/acs.jmedchem.7b00647 *
ROWE, RAYMOND C.: "Handbook of Pharmaceutical Excipients", 2005, PHARMACEUTICAL PRESS
See also references of EP4180430A4

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11970493B2 (en) 2020-10-06 2024-04-30 Ildong Pharmaceutical Co., Ltd. Autotaxin inhibitor compounds
WO2023185979A1 (zh) * 2022-03-31 2023-10-05 苏州爱科百发生物医药技术有限公司 一种atx抑制剂或其盐的结晶形式及其制备方法与应用

Also Published As

Publication number Publication date
EP4180430A1 (en) 2023-05-17
JP2023533294A (ja) 2023-08-02
EP4180430A4 (en) 2023-12-20
TW202202499A (zh) 2022-01-16
US20230295159A1 (en) 2023-09-21
CN115916777A (zh) 2023-04-04

Similar Documents

Publication Publication Date Title
WO2022111624A1 (zh) 一种苯并咪唑类衍生物及其制备方法和医药用途
WO2018157856A1 (zh) 酰胺类衍生物抑制剂及其制备方法和应用
KR20190039591A (ko) Glp-1 수용체 아고니스트 작용을 갖는 피라졸로피리딘 유도체
CN105566321B (zh) 杂芳化合物及其在药物中的应用
WO2019062733A1 (zh) Pde9 抑制剂及其用途
CN114423753A (zh) 作为cd38抑制剂的杂双环酰胺
WO2022007882A1 (zh) 一种atx抑制剂及其制备方法和应用
CN110546150B (zh) 含吡唑基的三并环类衍生物、其制备方法和应用
WO2020143763A1 (zh) 卤代烯丙基胺类化合物及其应用
WO2019085894A1 (zh) 一类含氮稠环化合物及其制备方法和用途
AU2012261077B2 (en) 2-amino-3-(imidazol-2-yl)-pyridin-4-one derivatives and their use as VEGF receptor kinase inhibitors
WO2015074516A1 (zh) 咪唑酮类衍生物、其药物组合物和用途
CN113912600A (zh) 一种atx抑制剂及其制备方法和应用
CN114437116A (zh) 杂环化合物及其制备方法、药物组合物和应用
WO2022017408A1 (zh) 芳胺类衍生物及其制备方法和医药用途
WO2021147940A1 (zh) 一种pd-1/pd-l1抑制剂及其制备方法和用途
CN114144410A (zh) 多芳基脲衍生物及其在治疗肌肉疾病中的用途
WO2023280254A1 (zh) 一种tead抑制剂
CN113166141A (zh) 一类六元并六元杂环化合物及其作为蛋白受体激酶抑制剂的用途
WO2022078356A1 (zh) 杂芳环类AhR抑制剂
WO2021249417A1 (zh) 杂环化合物及其衍生物
JP7216105B2 (ja) Erkキナーゼ阻害活性を有する化合物及びその使用
CN113493453A (zh) 稠合芳香环类衍生物、其制备方法及其在医药上的应用
CN106749203B (zh) 一种嘧啶类杂环化合物、嘧啶类杂环化合物盐以及制备方法和应用
WO2023125845A1 (zh) 芳杂双环化合物及其抗病毒用途

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: 21838673

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2023501105

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021838673

Country of ref document: EP

Effective date: 20230209