WO2024046342A1 - 苯并双环类化合物及其制备方法和应用 - Google Patents

苯并双环类化合物及其制备方法和应用 Download PDF

Info

Publication number
WO2024046342A1
WO2024046342A1 PCT/CN2023/115679 CN2023115679W WO2024046342A1 WO 2024046342 A1 WO2024046342 A1 WO 2024046342A1 CN 2023115679 W CN2023115679 W CN 2023115679W WO 2024046342 A1 WO2024046342 A1 WO 2024046342A1
Authority
WO
WIPO (PCT)
Prior art keywords
methoxy
mmol
chloro
formula
amino
Prior art date
Application number
PCT/CN2023/115679
Other languages
English (en)
French (fr)
Inventor
余天柱
张仕国
关昕宇
杨文谦
王慧
李捍雄
Original Assignee
广州市联瑞制药有限公司
广州一品红制药有限公司
广州润霖医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 广州市联瑞制药有限公司, 广州一品红制药有限公司, 广州润霖医药科技有限公司 filed Critical 广州市联瑞制药有限公司
Publication of WO2024046342A1 publication Critical patent/WO2024046342A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/381Heterocyclic compounds having sulfur as a ring hetero atom having five-membered rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms

Definitions

  • the invention relates to the technical field of chemical medicine, and in particular to a benzobicyclic compound and its preparation method and application.
  • Diabetes is a metabolic disease with multiple causes, characterized by chronic hyperglycemia, accompanied by disorders of sugar, lipid, and protein metabolism caused by defects in insulin secretion or action. Diabetes is a very old disease. It is caused by the absolute or relative lack of insulin in the human body. The concentration of glucose in the blood increases, and then a large amount of sugar is excreted in the urine, and symptoms such as polydipsia, polyuria, polyphagia, and weight loss occur. .
  • diabetes can usually be divided into type I diabetes and type II diabetes.
  • Type I diabetes is caused by the autoimmune system attacking the beta cells of the pancreas, causing it to lose its ability to secrete insulin.
  • Type II diabetes begins with abnormal insulin resistance or cells not responding to insulin. Obesity is one of the main causes of insulin resistance, so obesity can be said to be the main risk factor for type II diabetes.
  • Type II diabetes patients account for about 90% of diabetes patients, so the disease has become a major public health problem in developed countries where obesity is a serious problem, as well as in China, where the number of obesity continues to rise.
  • Glucagon-like peptide-1 (GLP-1) is an incretin hormone secreted by L-cells in the lower gastrointestinal tract. GLP-1 plays a corresponding role by binding to its widespread specific receptors. Currently, the organs in which GLP-1 receptors are clearly present include pancreatic islet cells, gastrointestinal tract, lungs, brain, kidneys, hypothalamus and cardiovascular system, liver There may be GLP-1 receptors in fat tissue and skeletal muscle. GLP-1 not only acts on ⁇ cells to promote insulin secretion, but also acts on ⁇ cells to inhibit glucagon secretion. There is generally no significant difference in serum GLP-1 levels between patients with normal glucose tolerance, impaired glucose tolerance, and type II diabetes.
  • Peptide GLP-1 receptor agonists can reduce fasting and postprandial glucose and improve blood sugar in patients with type II diabetes.
  • Peptide GLP-1 receptor agonists can reduce fasting and postprandial glucose and improve blood sugar in patients with type II diabetes.
  • the peptide GLP-1 has poor oral bioavailability and is inconvenient to take, there is an urgent clinical need for small molecule GLP-1 receptor agonists with good oral bioavailability.
  • the present invention provides a compound, or a pharmaceutical composition thereof, which can serve as an agonist of GLP-1 receptor.
  • the present invention further relates to the use of said compound or a pharmaceutical composition thereof for the preparation of a medicament for the treatment of diseases and/or conditions by agonizing GLP-1 receptors by said compound.
  • the present invention further describes the synthesis method of the compound.
  • the compounds of the present invention exhibit excellent biological activity and pharmacokinetic properties.
  • the present invention relates to a compound, which is a compound represented by formula (I), or a stereoisomer of a compound represented by formula (I), Geometric isomers, tautomers, nitrogen oxides, hydrates, solvates, metabolites, pharmaceutically acceptable salts or prodrugs,
  • X 1 , X 2 and X 3 are each independently N or CR 13 ;
  • Y 1 is N or CR 14 ;
  • Y 2 is N or CR 4 ;
  • Y 3 is N or CR 5 ;
  • Y 4 is N or CR 15 ;
  • L is O or CR 16 R 17 ;
  • Z is O or S
  • R 1 is H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, 3-6 membered heterocyclyl or -C 1-6 alkylene -R 18 , the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, 3-6 membered heterocyclyl and -C 1-6 alkylene
  • the group -R 18 may be independently optionally substituted by 1, 2 or 3 R 1a ;
  • R 18 is C 3-6 cycloalkyl, 3-8 membered heterocyclyl and 5-10 membered heteroaryl;
  • R 1a is D, F, Cl, Br, I, CN, hydroxyl, oxo, amino, nitro, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkylthio, C 1 -6 alkylamino, C 3-6 cycloalkyl or 3-6 membered heterocyclyl, the C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkylthio, C 1-6 Alkylamino, C 3-6 cycloalkyl and 3-6 membered heterocyclyl can be independently optionally substituted by 1, 2 or 3 R 1b ;
  • R 1b is D, F, Cl, Br, I, CN, hydroxyl, oxo, amino, nitro, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkylthio, C 1 -6 alkylamino, C 3-6 cycloalkyl or 3-6 membered heterocyclyl;
  • Each R 13 is independently H, D, F, Cl, Br, I, CN, hydroxyl, amino, nitro, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, C 1-6 alkoxy, C 1-6 alkylamino, C 1-6 haloalkoxy or -C 1-6 alkylene-C 1-6 alkoxy;
  • R 2 , R 3 , R 6 , R 7 , R 16 and R 17 are each independently H, D, F, Cl, Br, I, CN, hydroxyl, oxo, amino, nitro or C 1-6 alkane base, the C 1-6 alkyl group can be independently optionally substituted by 1, 2 or 3 F, Cl, Br, I, CN, hydroxyl, amino or nitro;
  • Ring B is a 5-6 membered heterocyclyl, C 5-6 cycloalkyl, phenyl or 5-6 membered heteroaryl;
  • Each R b is independently H, D, F, Cl, Br, I, CN, hydroxyl, oxo, amino, nitro, C 1-6 alkyl, C 1-6 alkoxy or C 1-6 alkyl Amino group, the C 1-6 alkyl group, C 1-6 alkoxy group and C 1-6 alkylamino group can be independently optionally replaced by 1, 2 or 3 F, Cl, Br, I, CN, hydroxyl, amino or nitro substitution;
  • R 4 , R 5 , R 8 , R 9 , R 10 , R 11 , R 12 , R 14 and R 15 are each independently H, D, F, Cl, Br, I, CN, hydroxyl, nitro, C 1-6 alkyl, C 1-6 alkoxy or C 1-6 alkylamino, the C 1-6 alkyl, C 1-6 alkoxy and C 1-6 alkylamino can be independently optionally 1, 2 or 3 F, Cl, Br, I, CN, hydroxyl, amino or nitro substitution;
  • n 0, 1, 2, 3, 4, 5, 6, 7 or 8.
  • a compound of the invention has a structure represented by Formula (II), Formula (III), Formula (IV), Formula (V), Formula (VI) or Formula (VII):
  • the X1 , X2 , X3 , Y1, Y2 , Y3 , Y4 , Z, R1 , R2 , R3 , R6 , R7 , R8 , R9 , R10 , R 11 , R 12 , R b and n have the meanings described in the present invention.
  • R1 is H, C 1-3 alkyl, C 2-3 alkenyl, C 2-3 alkynyl, C 3-6 cycloalkyl, 3-6 membered heterocyclyl, or -C 1-3 alkylene-R 18 , the C 1-3 alkyl, C 2-3 alkenyl, C 2-3 alkynyl, C 3-6 cycloalkyl, 3-6 membered heterocyclyl and - C 1-3 alkylene-R 18 may be independently optionally substituted by 1, 2 or 3 R 1a ;
  • R 18 is C 3-6 cycloalkyl, 3-6 membered heterocyclyl or 5-10 membered heteroaryl;
  • R 1a is D, F, Cl, Br, I, CN, hydroxyl, oxo, amino, nitro, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 alkylthio, C 1 -3 alkylamino, C 3-6 cycloalkyl or 5-6 membered heterocyclyl, the C 1-3 alkyl, C 1-3 alkoxy, C 1-3 alkylthio, C 1-3 Alkylamino, C 3-6 cycloalkyl and 5-6 membered heterocyclyl can be independently optionally substituted by 1, 2 or 3 R 1b ;
  • R 1b is D, F, Cl, Br, I, CN, hydroxyl, oxo, amino, nitro, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 alkylthio, C 1 -3 alkylamino, C 5-6 cycloalkyl or 5-6 membered heterocyclyl.
  • R1 is H, methyl, ethyl, n-propyl, isopropyl, vinyl, allyl, ethynyl, propargyl, 1-propynyl, cyclopropyl, cyclo Butyl, cyclopentyl, cyclohexyl, oxetanyl, azetidinyl, pyrrolidinyl, tetrahydrofuranyl, piperidinyl, piperazinyl, morpholinyl or -CH 2 R 18 , the methyl group base, ethyl, n-propyl, isopropyl, vinyl, allyl, ethynyl, propargyl, 1-propynyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxa Cyclobutyl, azetidinyl, pyrrolidinyl,
  • R 18 is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxetanyl, azetidinyl, pyrrolidinyl, tetrahydrofuranyl, piperidyl, piperazinyl, morpholinyl,
  • R 1a is D, F, Cl, Br, I, CN, hydroxyl, oxo, amino, nitro, methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, methylthio , N-methylamino, N-ethylamino, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxetanyl, azetidinyl, pyrrolidinyl, tetrahydrofuranyl, piperidyl, piperidyl Azinyl or morpholinyl, the methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, methylthio, N-methylamino, N-ethylamino, cyclopropyl, cyclopropyl, Butyl, cyclopentyl, cyclohexyl,
  • R 1b is D, F, Cl, Br, I, CN, hydroxyl, oxo, amino, nitro, methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, methylthio , N-methylamino, N-ethylamino, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxetanyl, azetidinyl, pyrrolidinyl, tetrahydrofuranyl, piperidyl, piperidyl Azinyl or morpholinyl.
  • R1 is H
  • each R 13 is independently H, D, F, Cl, Br, I, CN, hydroxy, amino, nitro, C 1-6 alkyl, C 1-6 haloalkyl, C 1- 6hydroxyalkyl , C 1-6 aminoalkyl, C 1-6 alkoxy, C 1-6 alkylamino, C 1-6 haloalkoxy or -C 1-6 alkylene-C 1-6 alkyl Oxygen;
  • R 2 , R 3 , R 6 , R 7 , R 16 and R 17 are each independently H, D, F, Cl, Br , I, CN, hydroxyl, oxo, amino, nitro or C 1-3 alkane base, the C 1-3 alkyl group can be independently optionally substituted by 1, 2 or 3 F, Cl, Br, I, CN, hydroxyl, amino or nitro;
  • Each R b is independently H, D, F, Cl, Br, I, CN, hydroxyl, oxo, amino, nitro, C 1-3 alkyl, C 1-3 alkoxy or C 1-3 alkyl Amino group, the C 1-3 alkyl group, C 1-3 alkoxy group and C 1-3 alkylamino group can be independently optionally replaced by 1, 2 or 3 F, Cl, Br, I, CN, hydroxyl, amino or nitro substitution;
  • R 4 , R 5 , R 8 , R 9 , R 10 , R 11 , R 12 , R 14 and R 15 are each independently H, D, F, Cl, Br, I, CN, hydroxyl, nitro, C 1-3 alkyl, C 1-3 alkoxy or C 1-3 alkylamino, the C 1-3 alkyl, C 1-3 alkoxy and C 1-3 alkylamino can be independently optionally 1, 2 or 3 F, Cl, Br, I, CN, hydroxyl, amino or nitro substitutions.
  • each R 13 is independently H, D, F, Cl, Br, I, CN, hydroxy, amino, nitro, methyl, ethyl, n-propyl, isopropyl, -CHF 2 , -CF 3 , -CHFCH 2 F, -CF 2 CHF 2 , -CH 2 CF 3 , -CH 2 CF 2 CHF 2 , hydroxymethyl, hydroxyethyl, aminomethyl, aminoethyl, methoxy, Ethoxy, n-propoxy, isopropoxy, N-methylamino, N-ethylamino, -OCHF 2 , -OCF 3 , -OCHFCH 2 F, -OCF 2 CHF 2 , -OCH 2 CF 3 , - OCH 2 CF 2 CHF 2 , -CH 2 OCH 3 , -CH 2 OCH 2 CH 3 , -CH 2 CH 2 OCH 3 or -CH 2 CH 2 OCH 2
  • R 2 , R 3 , R 6 , R 7 , R 16 and R 17 are each independently H, D, F, Cl, Br, I, CN, hydroxyl, oxo, amino, nitro, methyl, ethyl , n-propyl or isopropyl, the methyl, ethyl, n-propyl and isopropyl can be independently optionally replaced by 1, 2 or 3 F, Cl, Br, I, CN, hydroxyl, amino or Nitro substitution;
  • Each R b is independently H, D, F, Cl, Br, I, CN, hydroxyl, oxo, amino, nitro, methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy base, n-propoxy, isopropoxy, N-methylamino or N-ethylamino, the methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, n-propoxy , Isopropoxy, N-methylamino and N-ethylamino can be independently optionally substituted by 1, 2 or 3 F, Cl, Br, I, CN, hydroxyl, amino or nitro;
  • R 4 , R 5 , R 8 , R 9 , R 10 , R 11 , R 12 , R 14 and R 15 are each independently H, D, F, Cl, Br, I, CN, hydroxyl, nitro, methane base, ethyl, n-propyl, isopropyl, methoxy, ethoxy, n-propoxy, isopropoxy, N-methylamino or N-ethylamino, the methyl, ethyl, n-propyl Propyl, isopropyl, methoxy, ethoxy, n-propoxy, isopropoxy, N-methylamino and N-ethylamino can be independently optionally replaced by 1, 2 or 3 F, Cl, Br, I, CN, hydroxyl, amino or nitro substitution.
  • the compound of the present invention is a compound with one of the following structures or a stereoisomer, geometric isomer, tautomer, nitrogen oxide of a compound with one of the following structures , hydrate, solvate, metabolite, pharmaceutically acceptable salt or its prodrug:
  • the present invention relates to a pharmaceutical composition, which comprises formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI) or formula ( The compound described in VII), or its stereoisomers, geometric isomers, tautomers, nitrogen oxides, hydrates, solvates, metabolites, pharmaceutically acceptable salts or their prodrugs, and pharmaceutically acceptable carriers, excipients, diluents, auxiliaries, vehicles or combinations thereof.
  • the present invention relates to the use of the aforementioned compounds or pharmaceutical compositions thereof in the preparation of medicaments for preventing, treating or alleviating GLP-1 receptor agonist-mediated diseases in patients.
  • the disease mediated by the GLP-1 receptor agonist of the invention is diabetes, non-alcoholic fatty liver disease, or obesity.
  • the diabetes of the present invention is type I diabetes, type II diabetes, gestational diabetes, idiopathic type I diabetes, early-onset type II diabetes, adult-onset diabetes of the young, and adolescent-onset atypical diabetes. , malnutrition-related diabetes or latent autoimmune diabetes in adults.
  • the present invention relates to the preparation, isolation and preparation of compounds comprised by formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI) or formula (VII). Purification methods.
  • substituents When more than one position in a given structural formula can be substituted by one or more substituents selected from a specific group, the substituents may be identically or differently substituted at each position.
  • alkyl as used herein includes 1 to 20 carbon atoms, or 1 to 10 carbon atoms, or 1 to 6 carbon atoms, or 1 to 4 carbon atoms, or 1 to 3 carbon atoms, or A saturated straight-chain or branched-chain monovalent hydrocarbon group with 1-2 carbon atoms, in which the alkyl group can be independently optionally substituted by one or more substituents described in the present invention.
  • alkyl groups include, but are not limited to, methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), n-propyl (n-Pr, -CH 2 CH 2 CH 3 ), isopropyl (i-Pr, -CH(CH 3 ) 2 ), n-butyl (n-Bu, -CH 2 CH 2 CH 2 CH 3 ), isobutyl (i-Bu, -CH 2 CH(CH 3 ) 2 ), sec-butyl (s-Bu, -CH(CH 3 )CH 2 CH 3 ), tert-butyl (t-Bu, -C(CH 3 ) 3 ), n-pentyl (- CH 2 CH 2 CH 2 CH 3 ), 2-pentyl (-CH(CH 3 )CH 2 CH 2 CH 3 ), 3-pentyl (-CH(CH 2 CH 3 ) 2 ), 2-methyl Base-2-butyl (-C(CH 3 ), methyl
  • alkyl and its prefix “alkyl” as used herein include both straight and branched saturated carbon chains.
  • alkylene or “alkylene” as used herein refers to a saturated divalent hydrocarbon radical obtained by eliminating two hydrogen atoms from a linear or branched saturated hydrocarbon, examples of which include, but are not limited to, methylene base, ethylene and isopropylene, etc.
  • alkylene refers to a saturated divalent hydrocarbon radical obtained by removing two hydrogen atoms from a saturated straight or branched chain hydrocarbon radical. Unless otherwise specified, alkylene groups contain 1 to 12 carbon atoms. In some embodiments, the alkylene group contains 1-6 carbon atoms; in other embodiments, the alkylene group contains 1-4 carbon atoms; in still other embodiments, the alkylene group The alkylene group contains 1-3 carbon atoms; in still some embodiments, the alkylene group contains 1-2 carbon atoms. Such examples include methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), isopropylene (-CH(CH 3 )CH 2 -), and the like.
  • alkynyl means a linear or branched monovalent hydrocarbon group of 2 to 12 carbon atoms, or 2 to 8 carbon atoms, or 2 to 6 carbon atoms, or 2 to 4 carbon atoms, in which at least one The position is in an unsaturated state, that is, one CC is an sp triple bond, in which the alkynyl group can be independently and optionally substituted by one or more substituents described in the present invention.
  • Specific examples of the alkynyl group include, but Not limited to, ethynyl (-C ⁇ CH), propargyl (-CH 2 C ⁇ CH), etc.
  • halogen refers to F, Cl, Br or I.
  • unsaturated as used herein means a moiety containing one or more degrees of unsaturation.
  • alkoxy refers to an alkyl group, as defined herein, attached to other parts of the compound molecule through an oxygen atom.
  • the alkoxy group is a C 1-4 alkoxy group; such examples include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, and the like.
  • the alkoxy group may be independently unsubstituted or substituted by one or more substituents described herein.
  • alkylthio refers to an alkyl group, as defined herein, attached to other parts of the compound molecule through a sulfur atom.
  • the alkylthio group is a C 1-4 alkylthio group; such examples include, but are not limited to, methylthio, ethylthio, propylthio, butylthio, and the like.
  • the alkylthio group may be independently unsubstituted or substituted by one or more substituents described herein.
  • alkylamino refers to an alkyl group, as defined herein, attached to other parts of the compound molecule through an N atom.
  • the alkylamino group is C 1-4 alkylamino group; such examples include, but are not limited to, methylamino group, ethylamino group, propylamino group, butylamino group, and the like.
  • the alkylamino groups may be independently unsubstituted or substituted by one or more substituents described in the present invention.
  • cycloalkyl or “cycloalkane” refers to a monovalent or multivalent monocyclic, bicyclic or tricyclic carbocyclic ring system containing 3 to 12 carbon atoms, which is a saturated ring or contains one or more unsaturated bonds. rings, but never contain aromatic rings.
  • the cycloalkyl group contains 3-10 carbon atoms; in another embodiment, the cycloalkyl group contains 3-8 carbon atoms; in yet another embodiment, the cycloalkyl group contains 3-6 carbon atoms carbon atom.
  • Such examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, and the like.
  • the cycloalkyl groups may independently be unsubstituted or substituted with one or more substituents described herein.
  • heterocyclyl and “heterocycle” are used interchangeably herein and both refer to a saturated or partially unsaturated monocyclic, bicyclic or tricyclic ring containing 3 to 12 ring atoms, and never include aromatic rings, where At least one ring atom is a heteroatom.
  • heterocyclyl or “heterocycle” contains 3-10 ring atoms; in one embodiment, “heterocyclyl” or “heterocycle” contains 3-8 ring atoms; in another In one embodiment, “heterocyclyl” or “heterocycle” contains 5-8 ring atoms; in yet another embodiment, “heterocyclyl” or “heterocycle” contains 3-6 ring atoms; further In one embodiment, “heterocyclyl” or “heterocycle” contains 5-6 ring atoms; in yet another embodiment, “heterocyclyl” or “heterocycle” contains 4-6 ring atoms; unless otherwise Note that the heterocyclyl group can be a carbon group or a nitrogen group, and the heteroatom has the meaning as described in the present invention.
  • heterocyclyl groups include, but are not limited to: oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl , pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothiophenyl, dihydrothiophenyl, 1,3-dioxopentyl, disulfide ring Pentyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidyl, morpholinyl, thiomorpholinyl, piperazinyl , dioxanyl, dithianyl, thioxanyl, homopiperazinyl,
  • Examples of oxidized sulfur atoms in heterocyclyl groups include, but are not limited to, sulfolane groups and 1,1-dioxothiomorpholinyl groups.
  • the heterocyclyl group may be optionally substituted with one or more substituents described herein.
  • aryl refers to monocyclic, bicyclic and tricyclic carbocyclic ring systems containing 6 to 14 ring atoms, or 6 to 12 ring atoms, or 6 to 10 ring atoms, in which at least one ring is aromatic Each ring contains a ring of 3-7 atoms and has one or more attachment points to the rest of the molecule.
  • aryl may be used interchangeably with the term “aromatic ring”. Examples of aryl groups may include phenyl, naphthyl, and anthracenyl. The aryl groups may independently be optionally substituted with one or more substituents described herein.
  • heteroaryl means monocyclic, bicyclic and tricyclic ring systems containing 5 to 12 ring atoms, or 5 to 10 ring atoms, or 5 to 6 ring atoms, at least one of which is an aromatic ring, and At least one ring system contains one or more heteroatoms, each of which contains a ring of 5-7 atoms and has one or more points of attachment to the rest of the molecule.
  • heteroaryl may be used interchangeably with the term “heteroaryl ring” or “heteroaromatic compound.”
  • the heteroaryl group is optionally substituted with one or more substituents described herein.
  • the heteroaryl group consisting of 5 to 10 atoms contains 1, 2, 3 or 4 heteroatoms independently selected from O, S and N, wherein the nitrogen atom can be further oxidized.
  • heteroaryl groups include, but are not limited to: furyl, imidazolyl (such as N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl), isoxazolyl, oxazolyl (such as 2-oxazolyl, 4-oxazolyl, 5-oxazolyl), pyrrolyl (such as N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl), pyridyl, pyrimidinyl (such as 2- Pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl), pyridazinyl, thiazolyl (such as 2-thiazolyl, 4-thiazolyl, 5-thiazolyl), tetrazolyl (such as 5-tetrazolyl), Triazolyl, thienyl (such as 2-thienyl, 3-thienyl), pyrazolyl, isothiazolyl, 1,2,3-oxadiazol
  • haloalkyl or haloalkoxy means an alkyl or alkoxy group substituted by one or more halogen atoms. Examples of this include, but are not limited to, trifluoromethyl, trifluoromethoxy wait.
  • hydroxyalkyl means an alkyl group substituted with one or more hydroxyl groups, examples of which include, but are not limited to, hydroxymethyl, hydroxyethyl, and the like.
  • aminoalkyl means an alkyl group substituted with one or more amino groups, examples of which include, but are not limited to, aminomethyl, aminoethyl, and the like.
  • a ring system formed by a substituent bonded to a ring means that the substituent may be substituted at any substitutable position on the ring.
  • formula (a) represents that the substituent R may be mono- or poly-substituted at any possible substituted position on the pyridine ring.
  • a linking bond is attached to a ring to form a ring system (as shown in formula b), which means that the linking bond can be attached to the rest of the molecule at any attachable position on the ring system.
  • Formula b represents that any possible attachment position on the octahydrocyclopenta[c]pyrrole ring can be attached to the rest of the molecule.
  • the structural formulas described in the present invention include all isomeric forms (such as enantiomers, diastereomers, geometric isomers or conformational isomers): for example, R, S containing an asymmetric center Configuration, double bond (Z), (E) isomers, and (Z), (E) conformational isomers. Therefore, individual stereochemical isomers of the compounds of the invention or mixtures of enantiomers, diastereomers, geometric isomers or conformational isomers thereof are within the scope of the invention.
  • the structural formulas and compounds described in the present invention include all isomeric forms (such as enantiomers, diastereomers, geometric isomers or conformational isomers), nitrogen oxides, Hydrates, solvates, metabolites, pharmaceutically acceptable salts and prodrugs.
  • isomeric forms such as enantiomers, diastereomers, geometric isomers or conformational isomers
  • nitrogen oxides, Hydrates, solvates, metabolites pharmaceutically acceptable salts and prodrugs.
  • individual stereochemical isomers, enantiomers, diastereomers, geometric isomers, conformational isomers, nitrogen oxides, hydrates, solvates, metabolites, Pharmaceutically acceptable salts and prodrugs of the compounds are also within the scope of the invention.
  • the structural formulas of the compounds described herein include enriched isotopes of one or more different atoms.
  • Methodabolite refers to the product obtained through metabolism in the body of the specific compound described in the present invention or its pharmaceutically acceptable salt, analog or derivative, which exhibits the same behavior as formula (I) in vivo or in vitro , similar activity to compounds of formula (II), formula (III), formula (IV), formula (V), formula (VI) or formula (VII).
  • the metabolites of a compound can be identified by techniques well known in the art, and its activity can be characterized by assays as described herein. Such products can be obtained by administering compounds through oxidation, reduction, hydrolysis, amidation, deamidation, esterification, delipidation, or enzymatic cleavage, etc. Accordingly, the invention includes metabolites of compounds, including metabolites produced by contacting a compound of the invention with a mammal for a period of time sufficient to do so.
  • stereochemistry used in this invention are generally referred to the following documents: S.P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S ., "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., New York, 1994.
  • the compounds of the present invention may contain asymmetric centers or chiral centers and therefore exist as different stereoisomers. All stereoisomeric forms of the compounds of the present invention, including but not limited to, diastereomers, enantiomers, atropisomers, and mixtures thereof, such as racemic mixtures, constitute the present invention. part.
  • racemic mixture A 50:50 mixture of enantiomers is called a racemic mixture or racemate, which can result in no stereoselectivity or stereospecificity during a chemical reaction.
  • racemic mixture and “racemate” refer to an equimolar mixture of two enantiomers that lacks optical activity.
  • tautomer or “tautomeric form” refers to isomers of structures of different energies that can be converted into each other through a low energy barrier.
  • proton tautomers i.e., proton-shifting tautomers
  • tautomers by proton migration such as keto-enol and imine-enamine isomerizations.
  • Valence (valency) tautomers include recombination of bonding electrons.
  • “Pharmaceutically acceptable salts” used in the present invention refer to organic salts and inorganic salts of the compounds of the present invention.
  • Pharmaceutically acceptable salts are well known in the art, as described in SM Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66:1-19, 1977.
  • Pharmaceutically acceptable salts formed from nontoxic acids include, but are not limited to: inorganic acid salts formed by reaction with amino groups, such as hydrochlorides, hydrobromides, phosphates, sulfates, and perchlorates.
  • Organic acid salts such as acetate, oxalate, maleate, tartrate, citrate, succinate, malonate; or by other methods recorded in books and literature, such as ion exchange method Get these salts.
  • Other pharmaceutically acceptable salts include adipate, malate, 2-hydroxypropionate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, and bisulfate.
  • Salts obtained with appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(C 1-4 alkyl) 4 salts.
  • the present invention also contemplates the formation of quaternary ammonium salts of any compound containing an N group.
  • Water-soluble or oil-soluble or dispersed products can be obtained by quaternization.
  • Alkali or alkaline earth metals that can form salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Pharmaceutically acceptable salts further include appropriate, non-toxic ammonium, quaternary ammonium salts and amine cations that counter counter ion formation, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C1 -8 Sulfonates and aromatic sulfonates.
  • Hydrophilrate in the present invention refers to an association in which solvent molecules are water.
  • Solvents that form solvates include, but are not limited to: water, isopropyl alcohol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, and aminoethanol.
  • esters refers to a compound of formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI) or formula (VII) containing a hydroxyl group that can be hydrolyzed in vivo. of ester.
  • esters are, for example, pharmaceutically acceptable esters which hydrolyze in humans or animals to yield the parent alcohol.
  • Groups of in vivo hydrolyzable esters of compounds of formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI) or formula (VII) containing hydroxyl groups include, but do not Limited to: phosphate, acetoxymethoxy, 2,2-dimethylpropionyloxymethoxy, alkanoyl, benzoyl, phenylacetyl, alkoxycarbonyl, dialkylcarbamoyl and N-(dialkylaminoethyl)-N-alkylcarbamoyl, etc.
  • the "nitrogen oxide” of the present invention means that when the compound contains several amine functional groups, one or more nitrogen atoms can be oxidized to form N-oxide.
  • N-oxides are N-oxides of tertiary amines or N-oxides of nitrogen atoms in nitrogen-containing heterocyclic rings.
  • the corresponding amine can be treated with an oxidizing agent such as hydrogen peroxide or a peracid such as peroxycarboxylic acid to form an N-oxide (see Advanced Organic Chemistry, Wiley Interscience, 4th edition, Jerry March, pages).
  • the N-oxides can be prepared by the method of L.W. Deady (Syn. Comm. 1977, 7, 509-514), for example by reacting an amine compound with m-chloroperoxybenzoic acid (MCPBA) in an inert solvent such as methylene chloride. )reaction.
  • MCPBA m-chloroperoxybenzoic acid
  • prodrug used in the present invention represents a compound that is converted into formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI) or formula ( Compounds shown in VII). Such conversion is affected by hydrolysis of the prodrug in the blood or enzymatic conversion to the parent structure in the blood or tissue.
  • the prodrug compounds of the present invention can be esters.
  • esters that can be used as prodrugs include phenyl esters, aliphatic (C 1-24 ) esters, acyloxymethyl esters, and carbonate esters. , carbamates and amino acid esters.
  • a compound of the present invention contains a hydroxyl group, it can be acylated to obtain a prodrug form of the compound.
  • Other prodrug forms include phosphate esters, which are obtained by phosphorylation of the hydroxyl group of the parent.
  • prodrugs for a complete discussion of prodrugs, please refer to the following literature: T.Higuchi and V.Stella, Pro-drugs as Novel Delivery Systems, Vol.14 of the ACSSymposium Series, Edward B.Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, J.Rautio et al, Prodrugs: Design and Clinical Applications, Nature Review Drug Discovery, 2008, 7, 255-270, and SJHecker et al, Prodrugs of Phosphates and Phosphonates, Journal of Medicinal Chemistry, 2008 ,51, 2328-2345.
  • GLP-1 receptor agonist refers to a substance that agonizes GLP-1 receptor activity.
  • the compounds of the present invention can be prepared by the methods described in the present invention, unless further stated, wherein the substituent The definition is as described in the present invention.
  • the following reaction schemes and examples serve to further illustrate the present invention.
  • the reagents were purchased from commodity suppliers such as Anhui Zesheng Technology Co., Ltd., Shanghai Shaoyuan Reagents Co., Ltd., Shanghai Merrill Chemical Technology Co., Ltd., and Shanghai McLean Biochemical Technology Co., Ltd., and were used without further purification.
  • general reagents were purchased from Shantou Xilong Chemical Factory, Guangdong Guanghua Chemical Reagent Factory, Guangzhou Chemical Reagent Factory, Tianjin Damao Chemical Reagent Factory, Yantai Jiangyou Silica Gel Development Co., Ltd., and Qingdao Ocean Chemical Factory.
  • Anhydrous tetrahydrofuran, N,N-dimethylformamide, 1,4-dioxane, and acetonitrile are obtained by drying with molecular sieves.
  • Dichloromethane, ethyl acetate, petroleum ether, 1,2-dichloroethane and methanol were of analytical grade.
  • reaction bottles are plugged with appropriate rubber stoppers, and the substrate is injected through a syringe. Glassware is dried.
  • the silica gel column was purchased from Tianjin Bona Agel Technology Co., Ltd. using Flash silica gel column.
  • Silica gel (300-400 mesh) was purchased from Qingdao Marine Chemical Factory.
  • 1H NMR spectra were recorded using a Bruker 500MHz nuclear magnetic resonance spectrometer. 1H NMR spectrum uses CDCl 3 , DMSO-d 6 , CD 3 OD or acetone-d 6 as the solvent (in ppm), and TMS (0ppm) or chloroform (7.26ppm) as the reference standard.
  • the measurement conditions for low-resolution mass spectrometry (MS) data are: Agilent G6125C quadrupole HPLC-MS (column model: XBridge BEH C18, 4.6 x 50mm, 2.5 micron, 6min, flow rate 1mL/min.
  • ESI electrospray ionization
  • Ring B' is a 5-6 membered heterocyclic group containing nitrogen atoms.
  • X 1 , X 2 , X 3 , Y 1 , Y 2 , Y 3 , Y 4 , R 4 , R 5 , R 8 , R9 , R10 , R11 , R12 , R18 , Rb , n have the definitions as described in the present invention.
  • Bn is the commonly used protecting group benzyl
  • Tf is the commonly used protecting group trifluoromethanesulfonyl
  • Boc is the commonly used protecting group tert-butoxycarbonyl.
  • the compound represented by formula ( 7 ) can be prepared by reaction scheme 1: the compound represented by formula ( 1 ) and the compound represented by formula ( 2 ) are reacted to obtain the compound represented by formula ( 3 ).
  • the compound represented by formula ( 3 ) reacts with polyphosphoric acid to obtain the compound represented by formula ( 4 ).
  • the compound represented by formula ( 4 ) is brominated to obtain the compound represented by formula ( 5 ).
  • the compound represented by formula ( 5 ) and the compound represented by formula ( 6 ) react to obtain the compound represented by formula ( 7 ).
  • the compound represented by formula ( 13 ) can be prepared by reaction scheme 2: the compound represented by formula ( 8 ) and the compound represented by formula ( 9 ) are reacted to obtain the compound represented by formula ( 10 ).
  • the compound represented by formula ( 10 ) is subjected to reduction reaction to obtain the compound represented by formula ( 11 ).
  • the compound represented by formula ( 11 ) and the compound represented by formula ( 12 ) are reacted to obtain the compound represented by formula ( 13 ).
  • the compound represented by formula ( 21 ) can be prepared by reaction scheme 3: the compound represented by formula ( 14 ) and the compound represented by formula ( 15 ) are reacted to obtain the compound represented by formula ( 16 ).
  • the compound represented by formula ( 16 ) is hydrogenated and reduced to obtain the compound represented by formula ( 17 ).
  • the compound represented by formula ( 17 ) and the compound represented by formula ( 7 ) are reacted to obtain the compound represented by formula ( 18 ).
  • the compound represented by formula ( 18 ) can be removed from the N-Boc protecting group to obtain the compound represented by formula ( 19 ).
  • the compound represented by formula ( 19 ) and the compound represented by formula ( 13 ) are reacted to obtain the compound represented by formula ( 20 ).
  • the compound represented by formula ( 20 ) reacts under alkaline conditions to obtain the compound represented by formula ( 21 ).
  • the compound represented by formula ( 28 ) can be prepared by reaction scheme 4: the compound represented by formula ( 22 ) and the compound represented by formula ( 7 ) are reacted to obtain the compound represented by formula ( 23 ). The compound represented by formula ( 23 ) and the compound represented by formula ( 24 ) are reacted to obtain the compound represented by formula ( 25 ). The compound represented by formula ( 25 ) is removed from the N-Boc protecting group to obtain the compound represented by formula ( 26 ). The compound represented by formula ( 26 ) and the compound represented by formula ( 13 ) are reacted to obtain the compound represented by formula ( 27 ). The compound represented by formula ( 27 ) reacts under alkaline conditions to obtain the compound represented by formula ( 28 ).
  • the compound represented by formula ( 34 ) can be prepared by reaction scheme 5: the compound represented by formula ( 23 ) and the compound represented by formula ( 29 ) are reacted to obtain the compound represented by formula ( 30 ).
  • the compound represented by formula ( 30 ) reacts under basic conditions to obtain the compound represented by formula ( 31 ).
  • the compound represented by formula ( 31 ) and the compound represented by formula ( 11 ) are reacted to obtain the compound represented by formula ( 32 ).
  • the compound represented by formula ( 32 ) forms an intramolecular ring to obtain the compound represented by formula ( 33 ).
  • the compound represented by formula ( 33 ) reacts under basic conditions to obtain the compound represented by formula ( 34 ).
  • the compound represented by formula ( 16 ) can also be prepared by reaction scheme 6: the compound represented by formula ( 35 ) and the compound represented by formula ( 36 ) are reacted to obtain the compound represented by formula ( 37 ). The compound represented by formula ( 37 ) and the compound represented by formula ( 38 ) are reacted to obtain the compound represented by formula ( 39 ). The compound represented by formula ( 39 ) is subjected to a hydrogenation reduction reaction to obtain the compound represented by formula ( 16 ).
  • the compound represented by formula ( 10 ) can also be prepared through the reaction scheme 7: the compound represented by formula ( 40 ) is oxidized to obtain the compound represented by formula ( 41 ). The compound represented by formula ( 41 ) is esterified with methanol to obtain the compound represented by formula ( 42 ). The compound represented by formula ( 42 ) and the compound represented by formula ( 9 ) are reacted to obtain the compound represented by formula ( 10 ).
  • the compound represented by formula ( 13 ) can also be prepared by reaction scheme 8: the compound represented by formula ( 11 ) and the compound represented by formula ( 43 ) are reacted to obtain the compound represented by formula ( 44 ).
  • the compound represented by formula ( 44 ) can undergo an intramolecular cyclization reaction to obtain the compound represented by formula ( 13 ).
  • the compound represented by formula ( 29 ) can be prepared by reaction scheme 9: the compound represented by formula ( 45 ) and the compound represented by formula ( 46 ) are reacted to obtain the compound represented by formula ( 47 ).
  • the compound represented by formula ( 47 ) is subjected to boronation reaction to obtain the compound represented by formula ( 29 ).
  • the compound represented by formula ( 54 ) can be prepared by this reaction scheme 10: the compound represented by formula ( 48 ) and the compound represented by formula ( 15 ) are reacted to obtain the compound represented by formula ( 49 ). The compound represented by formula ( 49 ) and the compound represented by formula ( 50 ) are reacted to obtain the compound represented by formula ( 51 ). The compound represented by formula ( 51 ) is de-N-Boced to obtain the compound represented by formula ( 52 ). The compound represented by formula ( 52 ) and the compound represented by formula ( 13 ) are reacted to obtain the compound represented by formula ( 53 ). The compound represented by formula ( 53 ) reacts under basic conditions to obtain the compound represented by formula ( 54 ).
  • the compound represented by formula ( 62 ) can be prepared by the reaction scheme 11: the compound represented by formula ( 37 ) and the compound represented by formula ( 55 ) are reacted to obtain the compound represented by formula ( 56 ).
  • the compound represented by formula ( 56 ) is reduced to obtain the compound represented by formula ( 57 ).
  • the compound represented by formula ( 57 ) and the compound represented by formula ( 50 ) are reacted to obtain the compound represented by formula ( 58 ).
  • the compound represented by formula ( 58 ) reacts under basic conditions to obtain the compound represented by formula ( 59 ).
  • the compound represented by formula ( 59 ) and the compound represented by formula ( 11 ) are reacted to obtain the compound represented by formula ( 60 ).
  • the compound represented by formula ( 60 ) forms a ring within the molecule to obtain the compound represented by formula ( 61 ).
  • the compound represented by formula ( 61 ) reacts under alkaline conditions to obtain the compound represented by formula ( 62 ).
  • the compound represented by formula ( 69 ) can be prepared by reaction scheme 12: the compound represented by formula ( 35 ) is reacted with bromomethyl methyl ether to obtain the compound represented by formula ( 63 ). The compound represented by formula ( 63 ) and the compound represented by formula ( 64 ) are reacted to obtain the compound represented by formula ( 65 ). The compound represented by formula ( 65 ) is reduced to obtain the compound represented by formula ( 66 ). The compound represented by formula ( 66 ) is oxidized to obtain the compound represented by formula ( 67 ). The compound represented by formula ( 60 ) and the compound represented by formula ( 11 ) are reacted to obtain the compound represented by formula ( 68 ). The compound represented by formula ( 68 ) is deprotected by MOM to obtain the compound represented by formula ( 69 ).
  • the compound represented by formula ( 73 ) can be prepared by reaction scheme 13: the compound represented by formula ( 70 ) and the compound represented by formula ( 71 ) are reacted to obtain the compound represented by formula ( 72 ). The compound represented by formula ( 72 ) is deprotected to obtain the compound represented by formula ( 73 ).
  • the compound represented by formula ( 77 ) can be prepared by this reaction scheme 14: the compound represented by formula ( 63 ) and the compound represented by formula ( 74 ) are reacted to obtain the compound represented by formula ( 75 ). The compound represented by formula ( 75 ) and the compound represented by formula ( 13 ) are reacted to obtain the compound represented by formula ( 76 ). The compound represented by formula ( 76 ) is deprotected to obtain the compound represented by formula ( 77 ).
  • the compound represented by formula ( 82 ) can be prepared by reaction scheme 15: the compound represented by formula ( 5 ) and acetate are reacted to obtain the compound represented by formula ( 78 ).
  • the compound represented by formula ( 78 ) reacts under basic conditions to obtain the compound represented by formula ( 79 ).
  • the compound represented by formula ( 79 ) reacts with the compound represented by formula ( 80 ) to obtain the compound represented by formula ( 81 ).
  • the compound represented by formula ( 76 ) is fluorinated to obtain the compound represented by formula ( 77 ).
  • the compound represented by formula ( 90 ) can be prepared by reaction scheme 16: the compound represented by formula ( 83 ) is reacted with trimethoxymethane to obtain the compound represented by formula ( 84 ). The compound represented by formula ( 84 ) and the compound represented by formula ( 13 ) are reacted to obtain a compound represented by formula ( 85 ). The compound represented by formula ( 85 ) reacts under the action of trifluoroacetic acid to obtain the compound represented by formula ( 86 ). Compounds represented by formula ( 86 ) and N-phenyl The compound represented by formula ( 87 ) is obtained by the reaction of bis(trifluoromethanesulfonyl)imide.
  • the compound represented by formula ( 87 ) and pinacol diboronate are reacted to obtain the compound represented by formula ( 88 ).
  • the compound represented by formula ( 88 ) and the compound represented by formula ( 82 ) are reacted to obtain the compound represented by formula ( 89 ).
  • the compound represented by formula ( 89 ) reacts under alkaline conditions to obtain the compound represented by formula ( 90 ).
  • Step 5 Synthesis of (S)-4-nitro-3-((oxetan-2-ylmethyl)amino)benzoic acid methyl ester
  • Step 6 Synthesis of (S)-4-amino-3-((oxetan-2-ylmethyl)amino)benzoic acid methyl ester
  • Step 7 Synthesis of (S)-2-(chloromethyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 8 Synthesis of tert-butyl 4-(2-hydroxyphenyl)-3,6-dihydropyridine-1(2H)-carboxylate
  • Step 9 Synthesis of tert-butyl 4-(2-hydroxyphenyl)piperidine-1-carboxylate
  • Step 10 Synthesis of tert-butyl 4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)piperidine-1-carboxylate
  • Step 12 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)piperidin-1-yl)methyl) Synthesis of -1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 13 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)piperidin-1-yl)methyl) Synthesis of -1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 3 Synthesis of 4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)-1,2,3,6-tetrahydropyridine
  • Step 4 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)-3,6-dihydropyridine-1 (2H)-yl)methyl)-1-(oxetane Synthesis of alk-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 5 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)-3,6-dihydropyridine-1 Synthesis of (2H)-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of ethyl 2-(4-(((trifluoromethyl)sulfonyl)oxy)cyclohex-3-en-1-yl)acetate
  • Step 2 2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)cyclohex-3-en-1-yl) Synthesis of ethyl acetate
  • Step 4 2-(2'-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-2,3,4,5-tetrahydro-[1,1'-biphenyl Synthesis of ]-4-yl)ethyl acetate
  • Step 5 2-(2'-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-2,3,4,5-tetrahydro-[1,1'-biphenyl Synthesis of ]-4-yl)acetic acid
  • Step 6 4-(2-(2'-((4-chloro-2-fluorobenzofuran-7-yl)methyl)-2,3,4,5-tetrahydro-[1,1'- Synthesis of biphenyl]-4-yl)acetamido)-3-((((S)-oxetan-2-yl)methyl)amino)benzoic acid methyl ester
  • Step 7 2-((2'-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-2,3,4,5-tetrahydro-[1,1'-hydroxy Synthesis of benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 8 2-((2'-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-2,3,4,5-tetrahydro-[1,1'-hydroxy Synthesis of benzo[d]imidazole-6-carboxylic acid
  • Step 2 Synthesis of tert-butyl 4-(2-(benzyloxy)-3-fluorophenyl)-3,6-dihydropyridine-1(2H)-carboxylate
  • Step 3 Synthesis of tert-butyl 4-(3-fluoro-2-hydroxyphenyl)piperidine-1-carboxylate
  • Step 4 Synthesis of tert-butyl 4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidine-1-carboxylate
  • Step 6 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidin-1-yl Synthesis of )methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 7 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidin-1-yl )Methyl)-1-(oxetan-2-yl Synthesis of methyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of 6-chloro-5-nitro-2-carboxylic acid pyridine
  • Step 2 Synthesis of 6-chloro-5-nitro-2-carboxylic acid methyl pyridine
  • Step 5 Synthesis of (S)-5-(2-chloroacetamido)-6-((oxetan-2-ylmethyl)amino)pyridinecarboxylic acid methyl ester
  • Step 7 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidin-1-yl Synthesis of )methyl)-3-(oxetan-2-ylmethyl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid methyl ester
  • Step 8 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidin-1-yl Synthesis of )methyl)-3-(oxetan-2-ylmethyl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid
  • Step 1 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)piperidin-1-yl)methyl) Synthesis of -3-(oxetan-2-ylmethyl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid methyl ester
  • Step 2 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)piperidin-1-yl)methyl) Synthesis of -3-(oxetan-2-ylmethyl)-3H-imidazo[4,5-b]pyridine-5-carboxylic acid
  • Step 2 Synthesis of ethyl 2-(2'-(benzyloxy)-2,3,4,5-tetrahydro-[1,1'-biphenyl]-4-yl)acetate
  • Step 3 Synthesis of ethyl 2-(2'-hydroxy-2,3,4,5-tetrahydro-[1,1'-biphenyl]-4-yl)acetate
  • Step 4 Synthesis of ethyl 2-(4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)cyclohexyl)acetate
  • Step 5 Synthesis of 2-(4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)cyclohexyl)acetic acid
  • Step 6 (S)-4-(2-(4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)cyclohexyl)acetamido)-3 Synthesis of -((oxetan-2-ylmethyl)amino)benzoic acid methyl ester
  • Step 7 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)cyclohexyl)methyl)-1-( Synthesis of oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 8 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)cyclohexyl)methyl)-1-( Synthesis of oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of tert-butyl 4-(2-((4-chlorobenzofuran-7-yl)methoxy)-phenyl)piperazine-1-carboxylate
  • Step 3 (S)-2-((4-(2-((4-chlorobenzofuran-7-yl)methoxy)phenyl)piperazin-1-yl)methyl)-1-( Synthesis of oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 4 (S)-2-((4-(2-((4-chlorobenzofuran-7-yl)methoxy)phenyl)piperazin-1-yl)methyl)-1-( Synthesis of oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of tert-butyl 4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-phenyl)piperazine-1-carboxylate
  • Step 3 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)piperazin-1-yl)methyl) Synthesis of -1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 4 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)piperazin-1-yl)methyl) Synthesis of -1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 2 Synthesis of tert-butyl 4-(2-(benzyloxy)-3-fluorophenyl)piperazine-1-carboxylate
  • Step 3 Synthesis of tert-butyl 4-(3-fluoro-2-hydroxyphenyl)piperazine-1-carboxylate
  • Step 4 Synthesis of tert-butyl 4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperazine-1-carboxylate
  • Step 6 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperazin-1-yl Synthesis of )methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 7 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperazin-1-yl Synthesis of )methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 2-((S)-1-(4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperazine-1 Synthesis of -ethyl)-1-(((S)-(oxetan-2-yl)methyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 2 2-((S)-1-(4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperazine-1 -ethyl)-1-(((S)-oxetane Synthesis of -2-yl)methyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 2-((S)-1-(4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidine-1 Synthesis of -ethyl)-1-(((S)-oxetan-2-yl)methyl)-1H-benzo[d]imidazole-carboxylic acid methyl ester
  • Step 2 2-((S)-1-(4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidine-1 Synthesis of -ethyl)-1-(((S)-oxetan-2-yl)methyl)-1H-benzo[d]imidazole-carboxylic acid
  • Step 1 Synthesis of 3-hydroxy-3',6'-dihydro-(2,4'-bipyridine)-1'(2'H)-carboxylic acid tert-butyl ester
  • Step 2 Synthesis of tert-butyl 4-(3-hydroxypyridin-2-yl)piperidine-1-carboxylate
  • Step 3 Synthesis of tert-butyl 4-(3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)pyridin-2-yl)piperidine-1-carboxylate
  • Step 4 Synthesis of 3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-2-(piperidin-4-yl)pyridine hydrochloride
  • Step 5 (S)-2-((4-(3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)pyridin-2-yl)piperidin-1-yl) Synthesis of Methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate
  • Step 6 (S)-2-((4-(3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)pyridin-2-yl)piperidin-1-yl) Synthesis of methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 2 Synthesis of 2-(benzyloxy)-3',6'-dihydro-[3,4'-bipyridine]-1'(2'H)-carboxylic acid tert-butyl ester
  • Step 3 Synthesis of tert-butyl 4-(2-hydroxypyridin-3-yl)piperidine-1-carboxylate
  • Step 4 Synthesis of tert-butyl 4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)pyridin-3-yl)piperidine-1-carboxylate
  • Step 6 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)pyridin-3-yl)piperidin-1-yl) Synthesis of Methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate
  • Step 7 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)pyridin-3-yl)piperidin-1-yl) Synthesis of methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 2 Synthesis of methyl 2-(3-fluoro-2'-(methoxymethoxy)-[1,1'-biphenyl]-4-yl)acetate
  • Step 3 Synthesis of 2-(3-fluoro-2'-(methoxymethoxy)-[1,1'-biphenyl]-4-yl)ethan-1-ol
  • Step 4 Synthesis of 2-(3-fluoro-2'-(methoxymethoxy)-[1,1'-biphenyl]-4-yl)acetaldehyde
  • Step 5 (S)-2-((3-fluoro-2'-(methoxymethoxy)-[1,1'-biphenyl]-4-yl)methyl)-1-(oxygen Heterocyclobutan-2-ylmethyl)-1H-benzo[d] Synthesis of imidazole-6-carboxylic acid methyl ester
  • Step 6 (S)-2-((3-fluoro-2'-hydroxy-[1,1'-biphenyl]-4-yl)methyl)-1-(oxetane-2- Synthesis of methyl methyl)-1H-benzo[d]imidazole-6-carboxylate
  • Step 7 (S)-2-(4-(6-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)pyridin-2-yl)benzyl)-1-(oxy Synthesis of heterocyclobutan-2-ylmethyl)-1H-thieno[2,3-d]imidazole-5-carboxylic acid methyl ester
  • Step 8 (S)-2-((2'-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluoro-[1,1'-biphenyl] Synthesis of -4-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 2 Synthesis of methyl 2-(3,3'-difluoro-2'-(methoxymethoxy)-[1,1'-biphenyl]-4-yl)acetate
  • Step 3 Synthesis of 2-(3,3'-difluoro-2'-(methoxymethoxy)-[1,1'-biphenyl]-4-yl)ethan-1-ol
  • Step 4 Synthesis of 2-(3,3'-difluoro-2'-(methoxymethoxy)-[1,1'-biphenyl]-4-yl)acetaldehyde
  • Step 5 (S)-2-((3,3'-difluoro-2'-(methoxymethoxy)-[1,1'-biphenyl]-4-yl)methyl)- Synthesis of 1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 6 (S)-2-((3,3'-difluoro-2'-hydroxy-[1,1'-biphenyl]-4-yl)methyl)-1-(oxetane Synthesis of alk-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 7 (S)-2-((2'-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3,3'-difluoro-[1,1'- Synthesis of biphenyl]-4-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 8 (S)-2-((2'-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3,3'-difluoro-[1,1'- Synthesis of biphenyl]-4-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 2-((R)-1-(4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidine-1 Synthesis of -ethyl)-1-(((S)-oxetan-2-yl)methyl)-1H-benzo[d]imidazole-carboxylic acid methyl ester
  • Step 2 2-((R)-1-(4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidine-1 Synthesis of -ethyl)-1-(((S)-oxetan-2-yl)methyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of tert-butyl 4-(2-hydroxy-3-methoxyphenyl)-3,6-dihydropyridine-1(2H)-carboxylate
  • Step 2 Synthesis of tert-butyl 4-(2-hydroxy-3-methoxyphenyl)piperidine-1-carboxylate
  • Step 3 Synthesis of tert-butyl 4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-methoxyphenyl)piperidine-1-carboxylate
  • Step 5 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-methoxyphenyl)piperidine-1 Synthesis of -(yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 6 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-methoxyphenyl)piperidine-1 Synthesis of -(yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of tert-butyl 4-(2-hydroxy-3-methylphenyl)-3,6-dihydropyridine-1(2H)-carboxylate
  • Step 2 Synthesis of tert-butyl 4-(2-hydroxy-3-methylphenyl)piperidine-1-carboxylate
  • Step 3 Synthesis of tert-butyl 4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-methylphenyl)piperidine-1-carboxylate
  • Step 5 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-methylphenyl)piperidine-1- Synthesis of methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate
  • Step 6 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-methylphenyl)piperidine-1- Synthesis of methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 2 Synthesis of 4-(benzyloxy)-3',6'-dihydro-[3,4'-bipyridine]-1'(2'H)-carboxylic acid tert-butyl ester
  • Step 3 Synthesis of tert-butyl 4-(4-hydroxypyridin-3-yl)piperidine-1-carboxylate
  • Step 4 Synthesis of tert-butyl 4-(4-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)pyridin-3-yl)piperidine-1-carboxylate
  • Step 6 (S)-2-((4-(4-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)pyridin-3-yl)piperidin-1-yl) Synthesis of Methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate
  • Step 7 (S)-2-((4-(4-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)pyridin-3-yl)piperidin-1-yl) Synthesis of methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of tert-butyl 4-(2-((2,4-dichlorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidine-1-carboxylate
  • Step 3 (S)-2-((4-(2-((2,4-dichlorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidin-1-yl) Synthesis of methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate
  • Step 4 (S)-2-((4-(2-((2,4-dichlorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidin-1-yl) Synthesis of methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidin-1-yl)methyl) Synthesis of -1-((1-(cyanomethyl)cyclopropyl)methyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 2 2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidin-1-yl)methyl) Synthesis of -1-((1-(cyanomethyl)cyclopropyl)methyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of methyl 3-((2-methoxyethyl)amino)-4-nitrobenzoate
  • Step 2 Synthesis of methyl 4-amino-3-((2-methoxyethyl)amino)benzoate
  • Step 3 Synthesis of 2-(chloromethyl)-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 4 2-((4-(2-(((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidin-1-yl)methyl Synthesis of )-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 5 2-((4-(2-(((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidin-1-yl)methyl Synthesis of )-1-(2-methoxyethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of tert-butyl 4-(3,5-difluoro-2-hydroxyphenyl)-3,6-dihydropyridine-1(2H)-carboxylate
  • Step 2 Synthesis of tert-butyl 4-(3,5-difluoro-2-hydroxyphenyl)piperidine-1-carboxylate
  • Step 3 tert-butyl 4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3,5-difluorophenyl)piperidine-1-carboxylate synthesis
  • Step 4 Synthesis of 4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3,5-difluorophenyl)piperidine
  • Step 5 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3,5-difluorophenyl)piperidine- Synthesis of 1-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 6 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3,5-difluorophenyl)piperidine- Synthesis of 1-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of tert-butyl 4-(4-fluoro-2-hydroxyphenyl)-3,6-dihydropyridine-1(2H)-carboxylate
  • Step 2 Synthesis of tert-butyl 4-(4-fluoro-2-hydroxyphenyl)piperidine-1-carboxylate
  • Step 3 Synthesis of tert-butyl 4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-4-fluorophenyl)piperidine-1-carboxylate
  • Step 5 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-4-fluorophenyl)piperidin-1-yl Synthesis of )methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 6 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-4-fluorophenyl)piperidin-1-yl Synthesis of )methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of tert-butyl 4-(5-fluoro-6-hydroxyphenyl)-3,6-dihydropyridine-1(2H)-carboxylate
  • Step 2 Synthesis of tert-butyl 4-(5-fluoro-2-hydroxyphenyl)piperidine-1-carboxylate
  • Step 3 Synthesis of tert-butyl 4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-5-fluorophenyl)piperidine-1-carboxylate
  • Step 5 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-5-fluorophenyl)piperidin-1-yl Synthesis of )methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 6 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-5-fluorophenyl)piperidin-1-yl Synthesis of )methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 6 Synthesis of tert-butyl 4-(2-((2-chloro-4-cyanobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidine-1-carboxylate
  • Step 8 (S)-2-((4-(2-((2-chloro-4-cyanobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidine-1- Synthesis of methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate
  • Step 9 (S)-2-((4-(2-((2-chloro-4-cyanobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidine-1- Synthesis of methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 2 Synthesis of 3-(benzyloxy)-4-fluoro-3',6'-dihydro-(2,4'-bipyridyl)-1'(2'H)-carboxylic acid tert-butyl ester
  • Step 3 Synthesis of tert-butyl 4-(4-fluoro-3-hydroxypyridin-2-yl)piperidine-1-carboxylate
  • Step 4 Synthesis of tert-butyl 4-(3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-4-fluoropyridin-2-yl)piperidine-1-carboxylate
  • Step 5 Synthesis of 3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-4-fluoro-2-(piperidin-4-yl)pyridine hydrochloride
  • Step 6 (S)-2-((4-(3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-4-fluoropyridin-2-yl)piperidine- Synthesis of 1-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 7 (S)-2-((4-(3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-4-fluoropyridin-2-yl)piperidine- Synthesis of 1-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 2 Synthesis of 3-(benzyloxy)-5-fluoro-3',6'-dihydro-[2,4'-bipyridyl]-1'(2'H)-carboxylic acid tert-butyl ester
  • Step 3 Synthesis of tert-butyl 4-(5-fluoro-3-hydroxypyridin-2-yl)piperidine-1-carboxylate
  • Step 4 4-(3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-5-fluoropyridin-2-yl)piperidine-1-carboxylic acid tert-butyl ester synthesis
  • Step 5 (S)-2-((4-(3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-5-fluoropyridin-2-yl)piperidine- Synthesis of 1-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 6 (S)-2-((4-(3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-5-fluoropyridin-2-yl)piperidine- Synthesis of 1-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of tert-butyl 4-(2-fluoro-6-hydroxyphenyl)-3,6-dihydropyridine-1(2H)-carboxylate
  • Step 2 Synthesis of tert-butyl 4-(2-fluoro-6-hydroxyphenyl)piperidine-1-carboxylate
  • Step 3 Synthesis of tert-butyl 4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-6-fluorophenyl)piperidine-1-carboxylate
  • Step 5 (S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-6-fluorophenyl)piperidin-1-yl )Methyl)-1-(oxetan-2-yl Synthesis of methyl)-1H-benzo[d]imidazole-6-carboxylate
  • Step 6 ((S)-2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-6-fluorophenyl)piperidine-1- Synthesis of methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of 3'-hydroxy-3,6-dihydro-[4,4'-bipyridine]-1(2H)-carboxylic acid tert-butyl ester
  • Step 2 Synthesis of tert-butyl 4-(3-hydroxypyridin-4-yl)piperidine-1-carboxylate
  • Step 3 Synthesis of tert-butyl 4-(3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)pyridin-4-yl)piperidine-1-carboxylate
  • Step 4 Synthesis of 3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-4-(piperidin-4-yl)pyridine
  • Step 5 (S)-2-((4-(3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)pyridin-4-yl)piperidin-1-yl) Synthesis of Methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate
  • Step 6 (S)-2-((4-(3-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)pyridin-4-yl)piperidin-1-yl) Synthesis of methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 2 Synthesis of tert-butyl 4-(3-chloro-2-(methoxymethoxy)phenyl)-3,6-dihydropyridine-1(2H)-carboxylate
  • Step 3 Synthesis of tert-butyl 4-(3-chloro-2-(methoxymethoxy)phenyl)piperidine-1-carboxylate
  • Step 4 Synthesis of tert-butyl 4-(3-chloro-2-hydroxyphenyl)piperidine-1-carboxylate
  • Step 5 Synthesis of tert-butyl 4-(3-chloro-2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)piperidine-1-carboxylate
  • Step 7 (S)-2-((4-(3-chloro-2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)piperidin-1-yl Synthesis of )methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 8 (S)-2-((4-(3-chloro-2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)phenyl)piperidin-1-yl Synthesis of )methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 2 Synthesis of tert-butyl 4-(5-chloro-3-fluoro-2-(methoxymethoxy)phenyl)-3,6-dihydropyridine-1(2H)-carboxylate
  • Step 3 Synthesis of tert-butyl 4-(5-chloro-3-fluoro-2-(methoxymethyloxy)phenyl)piperidine-1-carboxylate
  • Step 4 Synthesis of tert-butyl 4-(5-chloro-3-fluoro-2-hydroxyphenyl)piperidine-1-carboxylate
  • Step 6 Synthesis of 4-(5-chloro-2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidine
  • Step 7 (S)-2-((4-(5-chloro-2-(4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidine- Synthesis of 1-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 8 (S)-2-((4-(5-chloro-2-(4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidine- Synthesis of 1-yl)methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 Synthesis of methyl 3-(((1-ethyl-1H-imidazol-5-yl)methyl)amino)-4-nitrobenzoate
  • Step 2 Synthesis of methyl 4-amino-3-(((1-ethyl-1H-imidazol-5-yl)methyl)amino)benzoate
  • Step 3 Synthesis of methyl 2-(chloromethyl)-1-((1-ethyl-1H-imidazol-5-yl)methyl)-1H-benzo[d]imidazole-6-carboxylate
  • Step 4 2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidin-1-yl)methyl) Synthesis of -1-((1-ethyl-1H-imidazol-5-yl)methyl)-1H-benzo[d]imidazole-6-carboxylic acid methyl ester
  • Step 5 2-((4-(2-((4-chloro-2-fluorobenzofuran-7-yl)methoxy)-3-fluorophenyl)piperidin-1-yl)methyl) Synthesis of -1-((1-ethyl-1H-imidazol-5-yl)methyl)-1H-benzo[d]imidazole-6-carboxylic acid
  • Step 1 2-((4-methoxybenzyl)oxy)-3',6'-dihydro-[3,4'-bipyridyl]-1'(2H)-carboxylic acid tert-butyl ester synthesis
  • Step 2 Synthesis of tert-butyl 4-(2-hydroxypyridin-3-yl)piperidine-1-carboxylate
  • Step 3 Synthesis of tert-butyl 4-(2-((2,4-dichlorobenzofuran-7-yl)methoxy)pyridin-3-yl)piperidine-1-carboxylate
  • Step 5 (S)-2-((4-(2-((2,4-dichlorobenzofuran-7-yl)methoxy)pyridin-3-yl)piperidin-1-yl)methyl Synthesis of methyl)-1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylate
  • Step 6 (S)-2-((4-(2-((2,4-dichlorobenzofuran-7-yl)methoxy)pyridin-3-yl)piperidin-1-yl)methyl Synthesis of 1-(oxetan-2-ylmethyl)-1H-benzo[d]imidazole-6-carboxylic acid

Landscapes

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

Abstract

公开了苯并双环类化合物及其在药物中的应用,具体地,涉及一类新型的苯并双环类化合物及包含该类化合物的药物组合物。还涉及制备所述化合物的方法,以及所述化合物或药物组合物在制备治疗GLP-1受体激动剂介导的疾病和/或病症的药物中的用途,特别是在制备治疗糖尿病、非酒精性脂肪肝病和肥胖的药物中的用途。

Description

苯并双环类化合物及其制备方法和应用
本申请主张如下优先权:
CN202211048114.4,申请日2022年8月30日。
技术领域
本发明涉及化学医药技术领域,具体涉及一种苯并双环类化合物及其制备方法和应用。
背景技术
糖尿病是一种多病因的代谢疾病,特点是慢性高血糖,伴随因胰岛素分泌或作用缺陷引起的糖、脂和蛋白质代谢紊乱。糖尿病是一种非常古老的疾病,是由于人体内胰岛素绝对或相对缺乏而引起的血中葡萄糖浓度升高,进而糖大量从尿中排出,并出现多饮、多尿、多食、消瘦等症状。
根据糖尿病发病机制分类,通常可将糖尿病划分为I型糖尿病和II型糖尿病。I型糖尿病是由于自身免疫***攻击胰岛β细胞从而丧失分泌胰岛素的功能。II型糖尿病则是始于胰岛素抵抗作用异常或细胞对胰岛素没有反应;肥胖是胰岛素抵抗的主要原因之一,因此肥胖可以说是II型糖尿病的主要危险因素。II型糖尿病患者占糖尿病患者人数的90%左右,因此该病在肥胖问题严重的发达国家、以及肥胖人数不断攀升的中国成为主要的公共卫生健康问题。
胰高血糖素样肽-1(GLP-1)是下消化道L-细胞分泌的一种肠降血糖素激素。GLP-1通过与其广泛存在的特异性受体结合而发挥相应的作用,目前明确存在GLP-1受体的器官有胰岛细胞、胃肠、肺、脑、肾脏、下丘脑和心血管***,肝脏、脂肪组积及骨骼肌中可能存在GLP-1受体。GLP-1不仅作用于β细胞促进胰岛素分泌,同时还作用于α细胞抑制胰高血糖素分泌。正常糖耐量、糖耐量受损和II型糖尿病患者中,血清GLP-1水平一般没有明显的差异。但是进食后β细胞对GLP-1的应答存在缺陷,在一定条件下,持续输注GLP-1后这种应答反应明显增强。由于人体自身GLP-1的作用持续时间十分短暂(静脉注射t1/2<1.5分钟),因此人体自身GLP-1并不适合用于糖尿病的临床治疗。
肽类GLP-1受体激动剂(如利拉鲁肽,艾塞那肽等)具有降低空腹和餐后葡萄糖以及改善II型糖尿病患者血糖的作用。然而,因为肽类GLP-1的口服生物利用度差,服用不便,所以临床亟需具有良好口服生物利用度的小分子GLP-1受体的激动剂。
目前,科研工作者已经开展了一些研究以期找到能够有效激动GLP-1受体的治疗药剂。PCT申请WO2018109607、WO2019239319、WO2019239371、WO2020103815、WO2020207474、WO2020263695、WO2021154796、WO2021112538、WO2021096304、WO2021096284、WO2021081207、WO2021018023和WO2021254470、WO2021249492、WO2022007979、WO2022031994、WO2022040600、WO2022068772和WO2022116693披露了诸多小分子化合物,其作为GLP-1受体激动剂用于预防或治疗糖尿病。然而,临床仍然亟需更多更好的GLP-1受体激动剂。
发明内容
本发明提供一种化合物,或其药物组合物,其可作为GLP-1受体的激动剂。本发明进一步涉及所述化合物或其药物组合物用于制备药物的用途,该药物通过所述化合物激动GLP-1受体来治疗疾病和/或病症。本发明又进一步描述了所述化合物的合成方法。本发明的化合物显示出优良的生物活性及药代动力学性质。
具体地说:
一方面,本发明涉及一种化合物,其为如式(I)所示的化合物,或式(I)所示的化合物的立体异构体、 几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
其中:
X1、X2和X3各自独立地为N或CR13
Y1为N或CR14
Y2为N或CR4
Y3为N或CR5
Y4为N或CR15
L为O或CR16R17
Z为O或S;
R1为H、C1-6烷基、C2-6烯基、C2-6炔基、C3-6环烷基、3-6元杂环基或-C1-6亚烷基-R18,所述C1- 6烷基、C2-6烯基、C2-6炔基、C3-6环烷基、3-6元杂环基和-C1-6亚烷基-R18可独立任选地被1、2或3个R1a取代;
R18为C3-6环烷基、3-8元杂环基和5-10元杂芳基;
R1a为D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷氨基、C3-6环烷基或3-6元杂环基,所述C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷氨基、C3-6环烷基和3-6元杂环基可独立任选地被1、2或3个R1b取代;
R1b为D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷氨基、C3-6环烷基或3-6元杂环基;
各R13独立地为H、D、F、Cl、Br、I、CN、羟基、氨基、硝基、C1-6烷基、C1-6卤代烷基、C1-6羟基烷基、C1-6氨基烷基、C1-6烷氧基、C1-6烷氨基、C1-6卤代烷氧基或-C1-6亚烷基-C1-6烷氧基;
R2、R3、R6、R7、R16和R17各自独立地为H、D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基或C1-6烷基,所述C1-6烷基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
环B为5-6元杂环基、C5-6环烷基、苯基或5-6元杂芳基;
各Rb独立地为H、D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、C1-6烷基、C1-6烷氧基或C1-6烷氨基,所述C1-6烷基、C1-6烷氧基和C1-6烷氨基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
R4、R5、R8、R9、R10、R11、R12、R14和R15各自独立地为H、D、F、Cl、Br、I、CN、羟基、硝基、C1-6烷基、C1-6烷氧基或C1-6烷氨基,所述C1-6烷基、C1-6烷氧基和C1-6烷氨基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
n为0、1、2、3、4、5、6、7或8。
在一些实施方案中,本发明化合物具有式(II)、式(III)、式(IV)、式(V)、式(VI)或式(VII)所示结构所示结构:
其中,所述X1、X2、X3、Y1、Y2、Y3、Y4、Z、R1、R2、R3、R6、R7、R8、R9、R10、R11、R12、Rb和n具有本发明所述的含义。
在一些实施方案中,R1为H、C1-3烷基、C2-3烯基、C2-3炔基、C3-6环烷基、3-6元杂环基或-C1-3亚烷基-R18,所述C1-3烷基、C2-3烯基、C2-3炔基、C3-6环烷基、3-6元杂环基和-C1-3亚烷基-R18可独立任选地被1、2或3个R1a取代;
R18为C3-6环烷基、3-6元杂环基或5-10元杂芳基;
R1a为D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、C1-3烷基、C1-3烷氧基、C1-3烷硫基、C1-3烷氨基、C3-6环烷基或5-6元杂环基,所述C1-3烷基、C1-3烷氧基、C1-3烷硫基、C1-3烷氨基、C3-6环烷基和5-6元杂环基可独立任选地被1、2或3个R1b取代;
R1b为D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、C1-3烷基、C1-3烷氧基、C1-3烷硫基、C1-3烷氨基、C5-6环烷基或5-6元杂环基。
在一些实施方案中,R1为H、甲基、乙基、正丙基、异丙基、乙烯基、烯丙基、乙炔基、炔丙基、1-丙炔基、环丙基、环丁基、环戊基、环己基、氧杂环丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基、吗啉基或-CH2R18,所述甲基、乙基、正丙基、异丙基、乙烯基、烯丙基、乙炔基、炔丙基、1-丙炔基、环丙基、环丁基、环戊基、环己基、氧杂环丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基、吗啉基和-CH2R18可独立任选地被1、2或3个R1a取代;
R18为环丙基、环丁基、环戊基、环己基、氧杂环丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基、吗啉基、
R1a为D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、甲硫基、N-甲氨基、N-乙氨基、环丙基、环丁基、环戊基、环己基、氧杂环丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基或吗啉基,所述甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、甲硫基、N-甲氨基、N-乙氨基、环丙基、环丁基、环戊基、环己基、氧杂环丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基和吗啉基可独立任选地被1、2或3个R1b取代;
R1b为D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、甲硫基、N-甲氨基、N-乙氨基、环丙基、环丁基、环戊基、环己基、氧杂环丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基或吗啉基。
在一些实施方案中,R1为H、
在一些实施方案中,各R13独立地为H、D、F、Cl、Br、I、CN、羟基、氨基、硝基、C1-6烷基、C1-6卤代烷基、C1-6羟基烷基、C1-6氨基烷基、C1-6烷氧基、C1-6烷氨基、C1-6卤代烷氧基或-C1-6亚烷基-C1-6烷氧基;
R2、R3、R6、R7、R16和R17各自独立地为H、D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基或C1- 3烷基,所述C1-3烷基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
各Rb独立地为H、D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、C1-3烷基、C1-3烷氧基或C1-3烷氨基,所述C1-3烷基、C1-3烷氧基和C1-3烷氨基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
R4、R5、R8、R9、R10、R11、R12、R14和R15各自独立地为H、D、F、Cl、Br、I、CN、羟基、硝基、C1-3烷基、C1-3烷氧基或C1-3烷氨基,所述C1-3烷基、C1-3烷氧基和C1-3烷氨基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代。
在一些实施方案中,各R13独立地为H、D、F、Cl、Br、I、CN、羟基、氨基、硝基、甲基、乙基、正丙基、异丙基、-CHF2、-CF3、-CHFCH2F、-CF2CHF2、-CH2CF3、-CH2CF2CHF2、羟基甲基、羟基乙基、氨甲基、氨乙基、甲氧基、乙氧基、正丙氧基、异丙氧基、N-甲氨基、N-乙氨基、-OCHF2、-OCF3、-OCHFCH2F、-OCF2CHF2、-OCH2CF3、-OCH2CF2CHF2、-CH2OCH3、-CH2OCH2CH3、-CH2CH2OCH3或-CH2CH2OCH2CH3
R2、R3、R6、R7、R16和R17各自独立地为H、D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、甲基、乙基、正丙基或异丙基,所述甲基、乙基、正丙基和异丙基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
各Rb独立地为H、D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、N-甲氨基或N-乙氨基,所述甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、N-甲氨基和N-乙氨基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
R4、R5、R8、R9、R10、R11、R12、R14和R15各自独立地为H、D、F、Cl、Br、I、CN、羟基、硝基、甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、N-甲氨基或N-乙氨基,所述甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、N-甲氨基和N-乙氨基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代。
在一些实施方案中,本发明所述的化合物,其为具有下列之一结构的化合物或具有下列之一结构的化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或它的前药:


一方面,本发明涉及药物组合物,该药物组合物,包含本发明式(I)、式(II)、式(III)、式(IV)、式(V)、式(VI)或式(VII)所述的化合物,或其立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或它们的前药,及其药学上可接受的载体、赋形剂、稀释剂、辅剂、媒介物或它们的组合。
一方面,本发明涉及前面所述的化合物或其药物组合物在制备药物中的用途,所述药物用于预防、治疗或减轻患者GLP-1受体激动剂介导的疾病。
其中一些实施方案是,本发明所述的GLP-1受体激动剂介导的疾病为糖尿病、非酒精性脂肪肝病或肥胖。
其中一些实施方案是,本发明所述的糖尿病为I型糖尿病、II型糖尿病、妊娠糖尿病、特发性I型糖尿病、早发型II型糖尿病、青年人的成年型糖尿病、青少年发作的非典型糖尿病、营养不良相关性糖尿病或成人隐匿性自身免疫性糖尿病。
另一方面,本发明涉及式(I)、式(II)、式(III)、式(IV)、式(V)、式(VI)或式(VII)所包含的化合物的制备、分离和纯化的方法。
前面所述内容只概述了本发明的某些方面,但并不限于这些方面。这些方面及其他的方面的内容将在下面作更加具体完整的描述。
定义和一般术语
本发明将会把确定的具体化的内容所对应的文献详细列出,实施例都伴随有结构式和化学式的图解。本发明有预期地涵盖所有的选择余地、变体和同等物,这些可能像权利要求所定义的那样包含在现有发明领域。所属领域的技术人员将识别许多类似或等同于在此所描述的方法和物质,这些可以应用于本发明的实践中去。本发明绝非限于方法和物质的描述。有很多文献和相似的物质与本发明申请相区别或抵触,其中包括但绝不限于术语的定义,术语的用法,描述的技术,或像本发明申请所控制的范围。
本发明将应用以下定义除非其他方面表明。根据本发明的目的,化学元素根据元素周期表,CAS版本和化学药品手册,75,thEd,1994来定义。另外,有机化学一般原理见"Organic Chemistry,"Thomas Sorrell,University Science Books,Sausalito:1999,and"March's Advanced Organic Chemistry,"by Michael B.Smith and Jerry March,John Wiley&Sons,New York:2007,因此所有的内容都融合了参考文献。
术语“包含”为开放式表达,即包括本发明所指明的内容,但并不排除其他方面的内容。
像这里所描述的化合物可以任选地被一个或多个取代基所取代,如本发明中的通式化合物,或者像实施例里面特殊的例子,子类,和本发明所包含的一类化合物。应了解“任选取代的”这个术语与“取代或非取代的”这个术语可以交换使用。一般而言,术语“任选地”不论是否位于术语“取代的”之前,表示所给结构中的一个或多个氢原子被具体取代基所取代。除非其他方面表明,一个任选的取代基团可以有一个取代基在基团各个可取代的位置进行取代。当所给出的结构式中不只一个位置能被选自具体基团的一个或多个取代基所取代,那么取代基可以相同或不同地在各个位置取代。其中所述的取代基可以是,但并不限于氢、F、Cl、Br、I、硝基、氰基、氧代(=O)、羟基、烷基、羟基烷基、烷氨基、氨基烷基、卤代烷氧基、环烷基、氨基、芳基、杂环基、杂芳基、烯基、炔基、环烷基氧基、烷氧基、烷氧基烷基、卤代烷基、-COOH、-亚烷基-C(=O)O-烷基、-亚烷基-S(=O)2-烷基、-亚烷基-S(=O)2-氨基、-S(=O)2-烷基、-S(=O)2-氨基、-S(=O)2OH、-O-亚烷基-C(=O)O-烷基、-O-亚烷基-S(=O)2-烷基、-O-亚烷基-S(=O)2-氨基、-O-亚烷基-S(=O)2OH、-C(=O)NH2、-C(=O)NH-烷基、-C(=O)N(烷基)-烷基、-C(=O)NHS(=O)2-烷基、-C(=O)NHS(=O)2-氨基、-C(=O)NHS(=O)2OH、-N(卤代烷基)-烷基、-N(烷基)-S(=O)2-烷基、-NHS(=O)2-烷基、-NHS(=O)2-卤代烷基、-N(烷基)S(=O)2-卤代烷基、-N(烷基)S(=O)2-烷氨基、-NHC(=O)-烷基、-NHC(=O)-卤代烷基、-N(烷基)C(=O)-卤代烷基、-N(烷基)C(=O)-烷氨基、-N(烷基)C(=O)O-烷基、-NHC(=O)O-烷基、-NHC(=O)O-卤代烷基、-N(烷基)C(=O)O-卤代烷基、-N(烷基)C(=O)O-氨基烷基、-NHC(=O)-NH2、-NHC(=O)NH-(烷基)、-NHC(=O)NH(卤代烷基)、-NHC(=O)N(烷基)-烷基、-OC(=O)-烷基、-OC(=O)-氨基、-OC(=O)-烷氨基、-OC(=O)-氨基烷基、-OC(=O)-烷氧基、-C(=O)N(烷基)S(=O)2-烷基、-C(=O)N(烷基)S(=O)2-氨基、-C(=O)NH-S(=O)2OH、-C(=NH)NH2、-C(=NH)NH-烷基、-C(=NH)N(烷基)-烷基、-C(=N-烷基)-NH2、-C(=O)NH-亚烷基-S(=O)2OH、-C(=O)NHC(=O)OH、-C(=O)NHC(=O)O-烷基、-C(=O)N(烷基)C(=O)O-烷基、-C(=O)NH-亚烷基-C(=O)OH和-C(=O)NH-亚烷基-C(=O)O-烷基,等等。
本发明使用的术语“烷基”包括1-20个碳原子,或1-10个碳原子,或1-6个碳原子,或1-4个碳原子,或1-3个碳原子,或1-2个碳原子饱和直链或支链的单价烃基,其中烷基可以独立任选地被一个或多个本发明所描述的取代基所取代。烷基基团更进一步的实例包括,但并不限于甲基(Me,-CH3)、乙基(Et,-CH2CH3)、正丙基(n-Pr,-CH2CH2CH3)、异丙基(i-Pr,-CH(CH3)2)、正丁基(n-Bu,-CH2CH2CH2CH3)、异丁基(i-Bu,-CH2CH(CH3)2)、仲丁基(s-Bu,-CH(CH3)CH2CH3)、叔丁基(t-Bu,-C(CH3)3)、正戊基(-CH2CH2CH2CH2CH3)、2-戊基(-CH(CH3)CH2CH2CH3)、3-戊基(-CH(CH2CH3)2)、2-甲基-2-丁基(-C(CH3)2CH2CH3)、3-甲基-2-丁基(-CH(CH3)CH(CH3)2)、3-甲基-1-丁基(-CH2CH2CH(CH3)2)、2-甲基-1-丁基(-CH2CH(CH3)CH2CH3)、正己基(-CH2CH2CH2CH2CH2CH3)、2-己基(-CH(CH3)CH2CH2CH2CH3)、3-己基(-CH(CH2CH3)(CH2CH2CH3))、2-甲基-2-戊基(-C(CH3)2CH2CH2CH3)、3-甲基-2-戊基(-CH(CH3)CH(CH3)CH2CH3)、4-甲基-2-戊基(-CH(CH3)CH2CH(CH3)2)、3-甲基-3-戊基(-C(CH3)(CH2CH3)2)、2-甲基-3-戊基(-CH(CH2CH3)CH(CH3)2)、2,3-二甲基-2-丁基(-C(CH3)2CH(CH3)2)、3,3-二甲基-2-丁基(-CH(CH3)C(CH3)3)、正庚基和正辛基等等。术语“烷基”和其前缀“烷”在此处使用,都包含直链和支链的饱和碳链。术语“烷撑”或“亚烷基”在此处使用,表示从直链或支链饱和碳氢化物消去两个氢原子得到的饱和二价烃基,这样的实例包括,但并不限于亚甲基、亚乙基和亚异丙基等等。
术语“亚烷基”表示从饱和的直链或支链烃基中去掉两个氢原子所得到的饱和的二价烃基基团。除非另外详细说明,亚烷基基团含有1-12个碳原子。在一些实施方案中,亚烷基基团含有1-6个碳原子;在另一些实施方案中,亚烷基基团含有1-4个碳原子;在又一些实施方案中,亚烷基基团含有1-3个碳原子;还在一些实施方案中,亚烷基基团含有1-2个碳原子。这样的实例包括亚甲基(-CH2-),亚乙基(-CH2CH2-),亚异丙基(-CH(CH3)CH2-)等等。
术语“烯基”表示2-12个碳原子,或2-8个碳原子,或2-6个碳原子,或2-4个碳原子的直链或支链的一价 烃基,其中至少一个位置为不饱和状态,即一个C-C为sp2双键,其中烯基基团可以独立且任选地被一个或多个本发明所描述的取代基所取代,包括基团有“反”,“顺”或“E”,“Z”的定位,其中烯基具体的实例包括,但并不限于,乙烯基(-CH=CH2)、烯丙基(-CH2CH=CH2)等等。
术语“炔基”表示2-12个碳原子,或2-8个碳原子,或2-6个碳原子,或2-4个碳原子的直链或支链的一价烃基,其中至少一个位置为不饱和状态,即一个C-C为sp三键,其中炔基基团可以独立且任选地被一个或多个本发明所描述的取代基所取代,其中炔基具体的实例包括,但并不限于,乙炔基(-C≡CH)、炔丙基(-CH2C≡CH)等等。
术语“杂原子”表示一个或多个O、S、N、P和Si,包括C,N,S和P任何氧化态的形式;伯、仲、叔胺和季铵盐的形式;或者杂环中氮原子上的氢被取代的形式,例如,N(像3,4-二氢-2H-吡咯基中的N),NH(像吡咯烷基中的NH)或NR(像N-取代的吡咯烷基中的NR);或杂环中的-CH2-被氧化,形成-C(=O)-的形式。
术语“卤素”是指F、Cl、Br或I。
术语“氘”是指重氢,D。
在本发明中所使用的术语“不饱和的”表示部分含有一个或多个不饱和度。
本发明中所使用的术语“烷氧基”或“烷基氧基”,涉及到烷基,像本发明所定义的,通过氧原子连接到化合物分子的其它部分上。一些实施例中,烷氧基为C1-4烷氧基;这样的实例包括,但并不限于甲氧基、乙氧基、丙氧基和丁氧基等。并且所述烷氧基可以独立地未被取代或被一个或多个本发明所描述的取代基所取代。
本发明中所使用的术语“烷硫基”或“烷基硫基”,涉及到烷基,像本发明所定义的,通过硫原子连接到化合物分子的其它部分上。一些实施例中,烷硫基为C1-4烷硫基;这样的实例包括,但并不限于甲硫基、乙硫基、丙硫基和丁硫基等。并且所述烷硫基可以独立地未被取代或被一个或多个本发明所描述的取代基所取代。
本发明中所使用的术语“烷氨基”或“烷基氨基”,涉及到烷基,像本发明所定义的,通过N原子连接到化合物分子的其它部分上。一些实施例中,烷氨基为C1-4烷氨基;这样的实例包括,但并不限于甲氨基、乙氨基、丙氨基和丁氨基等。并且所述烷氨基可以独立地未被取代或被一个或多个本发明所描述的取代基所取代。
术语“环烷基”或“环烷烃”表示含有3-12个碳原子的,单价或多价的单环,双环或三环碳环体系,为饱和环或含一个或多个不饱和键的环,但绝不包含芳香环。在一实施方案中,环烷基包含3-10个碳原子;在另一实施方案中,环烷基包含3-8个碳原子;在又一实施方案中,环烷基包含3-6个碳原子。这样的实例包括,但并不限于环丙基、环丁基、环戊基、环己基和环己烯基等。所述环烷基基团可以独立地未被取代或被一个或多个本发明所描述的取代基所取代。
术语“杂环基”和“杂环”在此处可交换使用,都是指包含3-12个环原子的饱和或部分不饱和的单环、双环或三环,绝不包含芳香环,其中至少一个环原子为杂原子。在一实施方案中,“杂环基”或“杂环”包含3-10个环原子;在一实施方案中,“杂环基”或“杂环”包含3-8个环原子;在另一实施方案中,“杂环基”或“杂环”包含5-8个环原子;在又一实施方案中,“杂环基”或“杂环”包含3-6个环原子;还在一实施方案中,“杂环基”或“杂环”包含5-6个环原子;再在一实施方案中,“杂环基”或“杂环”包含4-6个环原子;除非另外说明,杂环基可以是碳基或氮基,杂原子具有如本发明所述的含义。杂环基的实例包括,但不限于:环氧乙烷基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、2-吡咯啉基、3-吡咯啉基、吡唑啉基、吡唑烷基、咪唑啉基、咪唑烷基、四氢呋喃基、二氢呋喃基、四氢噻吩基、二氢噻吩基、1,3-二氧环戊基、二硫环戊基、四氢吡喃基、二氢吡喃基、2H-吡喃基、4H-吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、二噁烷基、二噻烷基、噻噁烷基、高哌嗪基、高哌啶基、氧杂环庚烷基、硫杂环庚烷基、氧氮杂基、二氮 杂基、硫氮杂基、2-氧杂-5-氮杂双环[2.2.1]庚-5-基和1,2,3,6-四氢吡啶基。杂环基中-CH2-基团被-C(=O)-取代的实例包括,但不限于:2-氧代吡咯烷基、氧代-1,3-噻唑烷基、2-哌啶酮基、3,5-二氧代哌啶基、嘧啶二酮基和5,6-二氢吡啶-2(1H)-酮基。杂环基中硫原子被氧化的实例包括,但不限于环丁砜基和1,1-二氧代硫代吗啉基。所述的杂环基基团可以任选地被一个或多个本发明所描述的取代基所取代。
术语“芳基”表示含有6-14个环原子,或6-12个环原子,或6-10个环原子的单环、双环和三环的碳环体系,其中,至少一个环是芳香族的,其中每一个环包含3-7个原子组成的环,且有一个或多个附着点与分子的其余部分相连。术语“芳基”可以和术语“芳环”交换使用。芳基基团的实例可以包括苯基、萘基和蒽基。所述芳基基团可以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”表示含有5-12个环原子,或5-10个环原子,或5-6个环原子的单环、双环和三环体系,其中至少一个环体系是芳香环,且至少一个环体系包含一个或多个杂原子,其中每一个环包含5-7个原子组成的环,且有一个或多个附着点与分子其余部分相连。术语“杂芳基”可以与术语“杂芳环”或“杂芳族化合物”交换使用。所述杂芳基基团任选地被一个或多个本发明所描述的取代基所取代。在一实施方案中,5-10个原子组成的杂芳基包含1、2、3或4个独立选自O,S和N的杂原子,其中氮原子可以被进一步氧化。
杂芳基基团的实例包括,但并不限于:呋喃基、咪唑基(如N-咪唑基、2-咪唑基、4-咪唑基、5-咪唑基)、异噁唑基、恶唑基(如2-噁唑基、4-噁唑基、5-噁唑基)、吡咯基(如N-吡咯基、2-吡咯基、3-吡咯基)、吡啶基、嘧啶基(如2-嘧啶基、4-嘧啶基、5-嘧啶基)、哒嗪基、噻唑基(如2-噻唑基、4-噻唑基、5-噻唑基)、四唑基(如5-四唑基)、***基、噻吩基(如2-噻吩基、3-噻吩基)、吡唑基、异噻唑基、1,2,3-噁二唑基、1,2,5-噁二唑基、1,2,4-噁二唑基、1,2,3-***基、1,2,3-硫代二唑基、1,3,4-硫代二唑基、1,2,5-硫代二唑基、吡嗪基、1,3,5-三嗪基;也包括以下的双环,但绝不限于这些双环:苯并咪唑基、苯并呋喃基、苯并噻吩基、吲哚基(如2-吲哚基)、嘌呤基、喹啉基(如2-喹啉基,3-喹啉基,4-喹啉基)、1,2,3,4-四氢异喹啉基、1,3-苯并二噁茂基、吲哚啉基、异喹啉基(如1-异喹啉基、3-异喹啉基或4-异喹啉基)、咪唑并[1,2-a]吡啶基、吡唑并[1,5-a]吡啶基、吡唑并[1,5-a]嘧啶基、咪唑并[1,2-b]哒嗪基、[1,2,4]***并[4,3-b]哒嗪基、[1,2,4]***并[1,5-a]嘧啶基和[1,2,4]***并[1,5-a]吡啶基,等等。
术语“卤代烷基”或“卤代烷氧基”表示烷基或烷氧基基团被一个或多个卤素原子所取代,这样的实例包含,但并不限于,三氟甲基、三氟甲氧基等。
术语“羟基烷基”表示烷基基团被一个或多个羟基所取代,这样的实例包含,但并不限于,羟基甲基、羟基乙基等。
术语“氨基烷基”表示烷基基团被一个或多个氨基所取代,这样的实例包含,但并不限于,氨甲基、氨乙基等。
像本发明所描述的,取代基画一个键连接到环上形成的环体系代表取代基在该环上任何可取代的位置都可以取代。例如,式(a)代表取代基R可以在吡啶环上任何可能被取代的位置上单取代或多取代。
像本发明所描述的,一个连接键连接到环上形成的环体系(如式b所示)代表连接键可以在环体系上任何可连接的位置与分子其余部分相连。式b代表八氢环戊烯并[c]吡咯环上任何可能连接的位置均可与分子其余部分相连。
另外,需要说明的是,除非以其他方式明确指出,在本文中通篇采用的描述方式“各…和…独立地为”、 “…和…各自独立地为”和“…和…分别独立地为”可以互换,应做广义理解,其既可以是指在不同基团中,相同符号之间所表达的具体选项之间互相不影响,也可以表示在相同的基团中,相同符号之间所表达的具体选项之间互相不影响。
除非其他方面表明,本发明所描述的结构式包括所有的同分异构形式(如对映异构,非对映异构,几何异构或构象异构):例如含有不对称中心的R、S构型,双键的(Z)、(E)异构体,和(Z)、(E)的构象异构体。因此,本发明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、几何异构体或构象异构体的混合物都属于本发明的范围。
除非其他方面表明,本发明所描述的结构式和所述的化合物包括所有的同分异构形式(如对映异构,非对映异构,几何异构或构象异构)、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐和前药。因此,本发明的化合物的单个立体化学异构体、对映异构体、非对映异构体、几何异构体、构象异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐和前药的化合物也属于本发明的范围。另外,除非其他方面表明,本发明所描述的化合物的结构式包括一个或多个不同的原子的富集同位素。
“代谢产物”是指本发明所述的具体的化合物或其药学上可接受的盐、类似物或衍生物在体内通过代谢作用所得到的产物,其在体内或体外表现出与式(I)、式(II)、式(III)、式(IV)、式(V)、式(VI)或式(VII)化合物类似的活性。一个化合物的代谢产物可以通过所属领域公知的技术来进行鉴定,其活性可以通过如本发明所描述的那样采用试验的方法进行表征。这样的产物可以是通过给药化合物经过氧化、还原、水解、酰氨化、脱酰氨作用、酯化、脱脂作用、或酶裂解等等方法得到。相应地,本发明包括化合物的代谢产物,包括将本发明的化合物与哺乳动物充分接触一段时间所产生的代谢产物。
本发明中立体化学的定义和惯例的使用通常参考以下文献:S.P.Parker,Ed.,McGraw-Hill Dictionary of Chemical Terms(1984)McGraw-Hill Book Company,New York;and Eliel,E.and Wilen,S.,"Stereochemistry of Organic Compounds",John Wiley&Sons,Inc.,New York,1994.本发明的化合物可以包含不对称中心或手性中心,因此存在不同的立体异构体。本发明的化合物所有的立体异构形式,包括但绝不限于,非对映体,对映异构体,阻转异构体,和它们的混合物,如外消旋混合物,组成了本发明的一部分。很多有机化合物都以光学活性形式存在,即它们有能力旋转平面偏振光的平面。在描述光学活性化合物时,前缀D、L或R、S用来表示分子手性中心的绝对构型。前缀d、l或(+)、(-)用来命名化合物平面偏振光旋转的符号,(-)或l是指化合物是左旋的,前缀(+)或d是指化合物是右旋的。这些立体异构体的化学结构是相同的,但是它们的立体结构不一样。特定的立体异构体可以是对映体,异构体的混合物通常称为对映异构体混合物。50:50的对映体混合物被称为外消旋混合物或外消旋体,这可能导致化学反应过程中没有立体选择性或立体定向性。术语“外消旋混合物”和“外消旋体”是指等摩尔的两个对映异构体的混合物,缺乏光学活性。
术语“互变异构体”或“互变异构的形式”是指不同能量的结构的同分异构体可以通过低能垒互相转化。例如质子互变异构体(即质子移变的互变异构体)包括通过质子迁移的互变,如酮式-烯醇式和亚胺-烯胺的同分异构化作用。原子价(化合价)互变异构体包括重组成键电子的互变。
本发明所使用的“药学上可接受的盐”是指本发明化合物的有机盐和无机盐。药学上可接受的盐在所属领域是为我们所熟知的,如文献:S.M.Berge et al.,describe pharmaceutically acceptable salts in detail in J.Pharmaceutical Sciences,66:1-19,1977.所记载的。药学上可接受的无毒的酸形成的盐包括,但并不限于:与氨基基团反应形成的无机酸盐,如盐酸盐、氢溴酸盐、磷酸盐、硫酸盐、高氯酸盐;有机酸盐,如乙酸盐、草酸盐、马来酸盐、酒石酸盐、柠檬酸盐、琥珀酸盐、丙二酸盐;或通过书籍文献上所记载的其他方法如离子交换法来得到这些盐。其他药学上可接受的盐包括,己二酸盐、苹果酸盐、2-羟基丙酸盐、藻酸盐、抗坏血酸盐、天冬氨酸盐、苯磺酸盐、苯甲酸盐、重硫酸盐、硼酸盐、丁酸盐、樟脑酸盐、樟脑磺酸盐、环戊基丙酸盐、二葡萄糖酸盐、十二烷基硫酸盐、乙磺酸盐、甲酸盐、反丁烯二酸盐、葡庚糖酸盐、甘油 磷酸盐、葡萄糖酸盐、半硫酸盐、庚酸盐、己酸盐、氢碘酸盐、2-羟基-乙磺酸盐、乳糖醛酸盐、乳酸盐、月桂酸盐、月桂基硫酸盐、苹果酸盐、甲磺酸盐、2-萘磺酸盐、烟酸盐、硝酸盐、油酸盐、棕榈酸盐、扑酸盐、果胶酸盐、过硫酸盐、3-苯基丙酸盐、苦味酸盐、特戊酸盐、丙酸盐、硬脂酸盐、硫氰酸盐、对甲苯磺酸盐、十一酸盐、戊酸盐等。通过适当的碱得到的盐包括,碱金属,碱土金属,铵和N+(C1-4烷基)4的盐。本发明也拟构思了任何所包含N的基团的化合物所形成的季铵盐。水溶性或油溶性或分散产物可以通过季铵化作用得到。可以形成盐的碱金属或碱土金属包括钠、锂、钾、钙、镁等。药学上可接受的盐进一步包括适当的、无毒的铵,季铵盐和抗平衡离子形成的胺阳离子,如卤化物、氢氧化物、羧化物、硫酸化物、磷酸化物、硝酸化物、C1-8磺酸化物和芳香磺酸化物。
本发明的“水合物”是指溶剂分子是水所形成的缔合物。
本发明的“溶剂化物”是指一个或多个溶剂分子与本发明的化合物所形成的缔合物。形成溶剂化物的溶剂包括,但并不限于:水、异丙醇、乙醇、甲醇、二甲亚砜、乙酸乙酯、乙酸、氨基乙醇。
本发明的“酯”是指含有羟基的式(I)、式(II)、式(III)、式(IV)、式(V)、式(VI)或式(VII)化合物形成体内可水解的酯。这样的酯是例如在人或动物体内水解产生母体醇的药学上可接受的酯。含有羟基的式(I)、式(II)、式(III)、式(IV)、式(V)、式(VI)或式(VII)化合物体内可水解的酯的基团包括,但不限于:磷酸基、乙酰氧基甲氧基、2,2-二甲基丙酰氧基甲氧基、烷酰基、苯甲酰基、苯乙酰基、烷氧基羰基、二烷基氨基甲酰基和N-(二烷基氨基乙基)-N-烷基氨基甲酰基等。
本发明的“氮氧化物”是指当化合物含几个胺官能团时,可将1个或大于1个的氮原子氧化形成N-氧化物。N-氧化物的特殊实例是叔胺的N-氧化物或含氮杂环氮原子的N-氧化物。可用氧化剂例如过氧化氢或过酸(例如过氧羧酸)处理相应的胺形成N-氧化物(参见Advanced Organic Chemistry,Wiley Interscience,第4版,Jerry March,pages)。尤其是,N-氧化物可用L.W.Deady的方法制备(Syn.Comm.1977,7,509-514),例如在惰性溶剂(例如二氯甲烷中),使胺化合物与间-氯过氧苯甲酸(MCPBA)反应。
本发明所使用的术语“前药”,代表一个化合物在体内转化为式(I)、式(II)、式(III)、式(IV)、式(V)、式(VI)或式(VII)所示的化合物。这样的转化受前体药物在血液中水解或在血液或组织中经酶转化为母体结构的影响。本发明前体药物类化合物可以是酯,在现有的发明中酯可以作为前体药物的有苯酯类、脂肪族(C1-24)酯类、酰氧基甲基酯类、碳酸酯、氨基甲酸酯类和氨基酸酯类。例如本发明里的一个化合物包含羟基,即可以将其酰化得到前体药物形式的化合物。其他的前体药物形式包括磷酸酯,如这些磷酸酯类化合物是经母体上的羟基磷酸化得到的。关于前体药物完整的讨论可以参考以下文献:T.Higuchi and V.Stella,Pro-drugs as Novel Delivery Systems,Vol.14 of the A.C.S.Symposium Series,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987,J.Rautio et al,Prodrugs:Design and Clinical Applications,Nature Review Drug Discovery,2008,7,255-270,and S.J.Hecker et al,Prodrugs of Phosphates and Phosphonates,Journal of Medicinal Chemistry,2008,51,2328-2345。
除非本文另有说明或者上下文清楚地有相反含义,否则本文所用的术语“一个”、“一种”、“该”以及本发明的上下文中(尤其是在权利要求书的上下文中)所使用的类似术语可以被解释为既包括单数,又包括复数。
本文所用的术语“GLP-1受体激动剂”是指能激动GLP-1受体活性的物质。
一般合成过程
为描述本发明,以下列出了实施例。但需要理解,本发明不限于这些实施例,只是提供实践本发明的方法。
一般地,本发明的化合物可以通过本发明所描述的方法制备得到,除非有进一步的说明,其中取代基 的定义如本发明所述。下面的反应方案和实施例用于进一步举例说明本发明的内容。
所属领域的技术人员将认识到:本发明所描述的化学反应可以用来合适地制备本发明的其他化合物,且用于制备本发明的化合物的其它方法都被认为是在本发明的范围之内。例如,根据本发明那些非例证的化合物的合成可以成功地被所属领域的技术人员通过修饰方法完成,如适当的保护干扰基团,通过利用其他已知的试剂除了本发明所描述的,或将反应条件做一些常规的修改。另外,本发明所公开的反应或已知的反应条件也公认地适用于本发明其他化合物的制备。
下面所描述的实施例,除非其他方面表明所有的温度定为摄氏度。试剂购买于商品供应商如安徽泽升科技有限公司,上海韶远试剂有限公司,上海迈瑞尔化学技术有限公司,上海麦克林生化科技有限公司,使用时都没有经过进一步纯化。除非其他方面表明,一般的试剂从汕头西陇化工厂,广东光华化学试剂厂,广州化学试剂厂,天津大茂化学试剂厂,烟台江友硅胶开发有限公司和青岛海洋化工厂购买得到。
无水四氢呋喃,N,N-二甲基甲酰胺,1,4-二氧六环,乙腈是经过分子筛干燥得到。二氯甲烷,乙酸乙酯,石油醚,1,2-二氯乙烷和甲醇使用分析纯。
以下反应一般是在氮气或氩气正压下或在无水溶剂上套一干燥管(除非其他方面表明),反应瓶都塞上合适的橡皮塞,底物通过注射器打入。玻璃器皿均是经过干燥的。
硅胶柱是使用Flash硅胶柱购于天津博纳艾杰尔科技有限公司。硅胶(300-400目)购于青岛海洋化工厂。
1H NMR谱使用Bruker 500MHz核磁共振谱仪记录。1H NMR谱以CDCl3、DMSO-d6、CD3OD或丙酮-d6为溶剂(以ppm为单位),用TMS(0ppm)或氯仿(7.26ppm)作为参照标准。当出现多重峰的时候,将使用下面的缩写:s(singlet,单峰)、d(doublet,双峰)、t(triplet,三重峰)、q(quartet,四重峰)、m(multiplet,多重峰)、br(broadened,宽峰)、brs(broadened singlet,宽的单峰)、dd(doublet of doublets,双二重峰)、dt(doublet of triplets,双三重峰)。偶合常数J,用赫兹(Hz)表示。
低分辨率质谱(MS)数据的测定条件是:Agilent G6125C四级杆HPLC-MS(色谱柱型号:XBridge BEH C18,4.6 x 50mm,2.5微米,6min,流速为1mL/min。流动相:0%-95%(CH3CN)在(含0.1%甲酸的H2O:CH3CN=90:10)中的比例,采用电喷雾电离(ESI),在210nm/254nm下,用DAD检测。
化合物纯化使用天津博纳艾杰尔科技有限公司Cheetah Pro中压快速纯化制备色谱,在210nm/254nm下,用UV检测测。
下面简写词的使用贯穿本发明:
PE 石油醚                                EtOAc/EA 乙酸乙酯
mg 毫克                                  mmol 毫摩尔
mL 毫升                                  g 克
M 摩尔/升                                rpm 转/分
μM 微摩尔/升                            h 小时
DCM 二氯甲烷                             MeOH 甲醇
min 分钟                                 CDCl3 氘代氯仿
DMSO-d6 氘代二甲基亚砜                   nM 纳摩尔/升
HEPES 4-羟乙基哌嗪乙磺酸                 BSA 牛血清白蛋白
IBMX 磷酸二酯酶抑制剂                    HBSS Hank's平衡盐溶液
nL 纳升                                  DMSO 二甲基亚砜
EDTA-K2 乙二胺四乙酸二钾                 Kolliphor-EL 乙氧基化蓖麻油
HP-β-CD 羟丙基倍他环糊精
下列反应方案描述了制备本发明化合物的步骤。环B'为含氮原子的5-6元杂环基,除非另有说明,各X1、X2、X3、Y1、Y2、Y3、Y4、R4、R5、R8、R9、R10、R11、R12、R18、Rb、n具有如本发明所述的定义。Bn为常用保护基团苄基,Tf为常用保护基团三氟甲磺酰基,Boc为常用保护基团叔丁氧羰基。
反应方案1
式(7)所示的化合物可以通过该反应方案1制备得到:式(1)所示的化合物和式(2)所示的化合物反应得到式(3)所示的化合物。式(3)所示的化合物在多聚磷酸的作用下反应得到式(4)所示的化合物。式(4)所示的化合物经溴代后得到式(5)所示的化合物。式(5)所示的化合物和式(6)所示的化合物反应得到式(7)所示的化合物。
反应方案2
式(13)所示的化合物可以通过该反应方案2制备得到:式(8)所示的化合物和式(9)所示的化合物反应得到式(10)所示的化合物。式(10)所示的化合物经还原反应得到式(11)所示的化合物。式(11)所示的化合物和式(12)所示的化合物反应得到式(13)所示的化合物。
反应方案3
式(21)所示的化合物可以通过该反应方案3制备得到:式(14)所示的化合物和式(15)所示的化合物反应得到式(16)所示的化合物。式(16)所示的化合物加氢还原反应得到式(17)所示的化合物。式(17)所示的化合物和式(7)所示的化合物反应得到式(18)所示的化合物。式(18)所示的化合物脱去N-Boc保护基团得到式(19)所示的化合物。式(19)所示的化合物和式(13)所示的化合物反应得到式(20)所示的化合物。式(20)所示的化合物在碱性条件下反应得到式(21)所示的化合物。
反应方案4
式(28)所示的化合物可以通过该反应方案4制备得到:式(22)所示的化合物和式(7)所示的化合物反应得到式(23)所示的化合物。式(23)所示的化合物和式(24)所示的化合物反应得到式(25)所示的化合物。式(25)所示的化合物脱去N-Boc保护基团得到式(26)所示的化合物。式(26)所示的化合物和式(13)所示的化合物反应得到式(27)所示的化合物。式(27)所示的化合物在碱性条件下反应得到式(28)所示的化合物。
反应方案5
式(34)所示的化合物可以通过该反应方案5制备得到:式(23)所示的化合物和式(29)所示的化合物反应得到式(30)所示的化合物。式(30)所示的化合物碱性条件下反应得到式(31)所示的化合物。式(31)所示的化合物和式(11)所示的化合物反应得到式(32)所示的化合物。式(32)所示的化合物分子内成环得到式(33)所示的化合物。式(33)所示的化合物在碱性条件下反应得到式(34)所示的化合物。
反应方案6
式(16)所示的化合物还可以通过该反应方案6制备得到:式(35)所示的化合物和式(36)所示的化合物反应得到式(37)所示的化合物。式(37)所示的化合物和式(38)所示的化合物反应得到式(39)所示的化合物。式(39)所示的化合物经加氢还原反应得到式(16)所示的化合物。
反应方案7
式(10)所示的化合物还可以通过该反应方案7制备得到:式(40)所示的化合物经氧化反应得到式(41)所示的化合物。式(41)所示的化合物和甲醇酯化反应得到式(42)所示的化合物。式(42)所示的化合物和式(9)所示的化合物反应得到式(10)所示的化合物。
反应方案8
式(13)所示的化合物还可以通过该反应方案8制备得到:式(11)所示的化合物和式(43)所示的化合物反应得到式(44)所示的化合物。式(44)所示的化合物经分子内成环反应得到式(13)所示的化合物。
反应方案9
式(29)所示的化合物可以通过该反应方案9制备得到:式(45)所示的化合物和式(46)所示的化合物反应得到式(47)所示的化合物。式(47)所示的化合物经硼化反应得到式(29)所示的化合物。
反应方案10
式(54)所示的化合物可以通过该反应方案10制备得到:式(48)所示的化合物和式(15)所示的化合物反应得到式(49)所示的化合物。式(49)所示的化合物和式(50)所示的化合物反应得到式(51)所示的化合物。式(51)所示的化合物脱N-Boc得到式(52)所示的化合物。式(52)所示的化合物和式(13)所示的化合物反应得到式(53)所示的化合物。式(53)所示的化合物在碱性条件下反应得到式(54)所示的化合物。
反应方案11
式(62)所示的化合物可以通过该反应方案11制备得到:式(37)所示的化合物和式(55)所示的化合物反应得到式(56)所示的化合物。式(56)所示的化合物经还原得到式(57)所示的化合物。式(57)所示的化合物和式(50)所示的化合物反应得到式(58)所示的化合物。式(58)所示的化合物碱性条件下反应得到式(59)所示的化合物。式(59)所示的化合物和式(11)所示的化合物反应得到式(60)所示的化合物。式(60)所示的化合物分子内成环得到式(61)所示的化合物。式(61)所示的化合物在碱性条件反应得到式(62)所示的化合物。
反应方案12
式(69)所示的化合物可以通过该反应方案12制备得到:式(35)所示的化合物和溴甲基甲基醚反应得到式(63)所示的化合物。式(63)所示的化合物和式(64)所示的化合物反应得到式(65)所示的化合物。式(65)所示的化合物经还原得到式(66)所示的化合物。式(66)所示的化合物经氧化得到式(67)所示的化合物。式(60)所示的化合物和式(11)所示的化合物反应得到式(68)所示的化合物。式(68)所示的化合物脱MOM保护得到式(69)所示的化合物。
反应方案13
式(73)所示的化合物可以通过该反应方案13制备得到:式(70)所示的化合物和和式(71)所示的化合物反应得到式(72)所示的化合物。式(72)所示的化合物脱保护得到式(73)所示的化合物。
反应方案14
式(77)所示的化合物可以通过该反应方案14制备得到:式(63)所示的化合物和式(74)所示的化合物反应得到式(75)所示的化合物。式(75)所示的化合物和式(13)所示的化合物反应得到式(76)所示的化合物。式(76)所示的化合物脱保护得到式(77)所示的化合物。
反应方案15
式(82)所示的化合物可以通过该反应方案15制备得到:式(5)所示的化合物和醋酸盐反应得到式(78)所示的化合物。式(78)所示的化合物碱性条件下反应得到式(79)所示的化合物。式(79)所示的化合物和和式(80)所示的化合物反应得到式(81)所示的化合物。式(76)所示的化合物经氟代得到式(77)所示的化合物。
反应方案16
式(90)所示的化合物可以通过该反应方案16制备得到:式(83)所示的化合物和三甲氧基甲烷反应得到式(84)所示的化合物。式(84)所示的化合物和式(13)所示的化合物反应得到式(85)所示的化合物。式(85)所示的化合物在三氟乙酸作用下反应得到式(86)所示的化合物。式(86)所示的化合物和N-苯基 双(三氟甲烷磺酰)亚胺反应得到式(87)所示的化合物。式(87)所示的化合物和联硼酸频那醇酯反应得到式(88)所示的化合物。式(88)所示的化合物和式(82)所示的化合物反应得到式(89)所示的化合物。式(89)所示的化合物在碱性条件反应得到式(90)所示的化合物。
具体实施方式
下面参考具体实施例,对本发明进行描述,需要说明的是,这些实施例仅仅是描述性的,而不以任何方式限制本发明。
实施例1(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物1)的合成
步骤1:4-氯-2-(2,2-二乙氧基乙氧基)-1-甲基苯的合成
将化合物5-氯-2-甲基苯酚(15g,105.2mmol),2-溴-1,1-二乙氧基乙烷(25g,126.9mmol)和碳酸钾(29.1g,210.4mmol)加入到N,N-二甲基甲酰胺(50mL)中,氮气保护下120℃反应11h。冷却至室温,加入水(100mL),用乙酸乙酯萃取(35mL×3),合并有机相,用饱和食盐水(30mL)洗一次,有机相用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1%)得到浅褐色液体产物21.1g,产率77.5%。
1HNMR(500MHz,CDCl3)δ7.03(d,J=7.9Hz,1H),6.84(dd,J=7.9,2.0Hz,1H),6.81(d,J=2.0Hz,1H),4.84(t,J=5.2Hz,1H),3.98(d,J=5.2Hz,2H),3.81–3.74(m,2H),3.68–3.62(m,2H),2.18(s,3H),1.25(t,J=7.0Hz,6H).
步骤2:4-氯-7-甲基苯并呋喃的合成
将化合物4-氯-2-(2,2-二乙氧基乙氧基)-1-甲基苯(21g,81.2mmol)和多聚磷酸(16.6g,202.9mmol)加入到1,2-二氯乙烷(50mL)中,氮气保护下85℃反应过夜。冷却至室温,减压除去溶剂,加入水(80mL),用乙酸乙酯萃取(50mL×2),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE=100%)得到无色液体产物9.2g,产率68.0%。
1HNMR(500MHz,CDCl3)δ7.63(d,J=2.3Hz,1H),7.12(d,J=7.9Hz,1H),7.00(d,J=7.9Hz,1H),6.84(d,J=2.3Hz,1H),2.48(s,3H).
步骤3:7-(溴甲基)-4-氯苯并呋喃的合成
将化合物4-氯-7-甲基苯并呋喃(4.1g,24.61mmol),偶氮二异丁腈(810mg,4.92mmol)和N-溴代丁二酰亚胺(5.26g,29.53mmol)加入到1,2-二氯乙烷(30mL)中,氮气保护下75℃反应过夜。冷却至室温,加入水(50mL),用二氯甲烷萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE=100%)得到无色液体产物4.3g,产率71.2%。
1HNMR(500MHz,CDCl3)δ7.71(s,1H),7.24–7.20(m,2H),6.89(s,1H),4.75(s,2H).
步骤4:7-(溴甲基)-4-氯-2-氟苯并呋喃的合成
将化合物7-(溴甲基)-4-氯苯并呋喃(3.4g,13.85mmol)和N-氟代双苯磺酰胺(5.24g,16.62mmol)加入到无水四氢呋喃(100mL)中,氮气保护下-50℃冷却,加入二异丙基氨基锂(2.0mol/L的四氢呋喃溶液) (9mL,18mmol),此温度下反应3h。加入水(300mL),用乙酸乙酯萃取(50mL×2),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE=100%)得到白色固体产物523mg,产率14.3%。
1HNMR(500MHz,CDCl3)δ7.24–7.20(m,2H),6.01(d,J=6.6Hz,1H),4.65(s,2H).
步骤5:(S)-4-硝基-3-((氧杂环丁烷-2-基甲基)氨基)苯甲酸甲酯的合成
将化合物3-氟-4-硝基苯甲酸甲酯(500mg,2.51mmol),无水碳酸钾(521mg,3.77mmol)加入到无水四氢呋喃(6mL)中,再加入(S)-氧杂环丁烷-2-甲胺(241mg,2.76mmol),室温反应15h。加入水(25mL)和乙酸乙酯(10mL)搅拌后分离有机相,水相用乙酸乙酯萃取(5mL×2),合并有机相,用无水硫酸钠干燥20min,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:4)得到红褐色固体522mg,产率78.1%。
LC-MS(ESI):[M+H]+=267.1
1HNMR(500MHz,DMSO-d6)δ8.32(t,J=5.8Hz,1H),8.17(d,J=8.9Hz,1H),7.67(d,J=1.7Hz,1H),7.16(dd,J=8.9,1.8Hz,1H),5.03–4.96(m,1H),4.58–4.54(m,1H),4.46–4.42(m,1H),3.88(s,3H),3.71–3.61(m,2H),2.71–2.64(m,1H),2.56–2.50(m,1H).
步骤6:(S)-4-氨基-3-((氧杂环丁烷-2-基甲基)氨基)苯甲酸甲酯的合成
将(S)-4-硝基-3-((氧杂环丁烷-2-基甲基)氨基)苯甲酸甲酯(3.6g,13.52mmol)溶解在甲醇(30mL)和四氢呋喃(15mL)中,加入10%钯碳(含水量60%,1.43g),用氢气置换3次,常温常压下氢气反应过夜。反应液用硅藻土过滤,滤液减压浓缩,得到淡黄色液体产物3.18g,产率99.5%。
LC-MS(ESI):[M+H]+=237.1;
1HNMR(500MHz,DMSO-d6)δ7.17(dd,J=8.1,1.9Hz,1H),7.04(d,J=1.9Hz,1H),6.56(d,J=8.1Hz,1H),5.44(s,2H),4.95–4.90(m,1H),4.67(t,J=5.7Hz,1H),4.56–4.52(m,1H),4.50–4.46(m,1H),3.73(s,3H),3.37–3.25(m,2H),2.70–2.63(m,1H),2.48–2.43(m,1H).
步骤7:(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-4-氨基-3-((氧杂环丁烷-2-基甲基)氨基)苯甲酸甲酯(1.0g,4.23mmol),氯乙基原乙酸三甲酯(790mg,5.08mmol)和对甲苯磺酸一水合物(40mg,0.21mmol)溶于无水四氢呋喃(8mL)中,45℃搅拌反应5h,冷却,减压浓缩除去溶剂,加入二氯甲烷(6mL)溶解,硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:2)得到白色固体产物1.1g,产率88.2%。
LC-MS(ESI):[M+H]+=295.2;
1HNMR(500MHz,DMSO-d6)δ8.35(s,1H),7.87(dd,J=8.5,1.6Hz,1H),7.74(d,J=8.5Hz,1H),5.20–5.08(m,2H),5.09–5.04(m,1H),4.78–4.74(m,1H),4.66–4.63(m,1H),4.50–4.45(m,1H),4.35–4.30(m,1H),3.89(s,3H),2.73–2.66(m,1H),2.41–2.34(m,1H).
步骤8:4-(2-羟苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将化合物2-羟基苯硼酸(1.25g,9.05mmol),4-(((三氟甲基)磺酰基)氧基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(3.0g,9.05mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(370mg,0.45mmol)和碳酸钾(8.85g,27.16mmol)加入到1,4-二氧六环(40mL)中,氮气保护下90℃反应过夜,冷却至室温,加入水(80mL),用乙酸乙酯萃取(20mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=16%)得到白色固体产物1.5g,产率60.2%。
LC-MS(ESI):[M-55]+=220.2;
1HNMR(500MHz,CDCl3)δ7.15(td,J=7.7,1.7Hz,1H),7.08(dd,J=7.8,1.7Hz,1H),6.91–6.87(m,2H),5.85(s,1H),5.46(s,1H),4.10–4.05(m,2H),3.65(t,J=5.7Hz,2H),2.47–2.41(m,2H),1.50(s,9H).
步骤9:4-(2-羟苯基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(2-羟苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(1.5g,5.45mmol),溶解在甲醇(30mL)中, 加入10%钯碳(500mg,含水量40-60%),常温常压下通入氢气反应过夜,过滤出去催化剂,滤液减压浓缩,得到白色固体产物1.4g,产率92.7%。
LC-MS(ESI):[M+H]+=222.1;
1HNMR(500MHz,CDCl3)δ7.13(dd,J=7.6,1.7Hz,1H),7.07(td,J=7.7,1.7Hz,1H),6.94–6.87(m,1H),6.77–6.71(m,1H),4.96(s,1H),4.31–4.18(m,2H),3.06–2.99(m,1H),2.87–2.78(m,2H),1.85–1.79(m,2H),1.67–1.60(m,2H),1.49(s,9H).
步骤10:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-羧酸叔丁酯的合成
将化合物7-(溴甲基)-4-氯苯并呋喃(290mg,1.10mmol)和4-(2-羟苯基)哌啶-1-羧酸叔丁酯(305mg,1.10mmol)加入到四氢呋喃(8mL)中,在冰浴中冷却,加入叔丁醇钾(185mg,1.65mmol),转移到室温反应1.5h。加入水(30mL),用乙酸乙酯萃取(15mL×2),有机相用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=10%)得到褐色油状产物368mg,产率72.7%。
LC-MS(ESI):[M-55]+=404.2;
1HNMR(500MHz,CDCl3)δ7.29–7.23(m,2H),7.21–7.15(m,2H),6.99–6.94(m,2H),6.03(d,J=6.5Hz,1H),5.29(s,2H),4.31–4.12(m,2H),3.18–3.10(m,1H),2.83–2.70(m,2H),1.83–1.76(m,2H),1.64–1.56(m,2H),1.47(s,9H).
步骤11:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-羧酸叔丁酯(660mg,1.43mmol),溶解在二氯甲烷(8mL)中,加入三氟乙酸(3.3mL)室温反应30min,减压除去溶剂,加入饱和碳酸氢钠调节pH=8,用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,得到白色固体产物509mg,产率98.6%。
LC-MS(ESI):[M+H]+=360.2;
1HNMR(500MHz,CDCl3)δ7.29–7.20(m,4H),7.04–6.96(m,2H),6.04(d,J=6.7Hz,1H),5.29(s,2H),3.49(d,J=12.6Hz,2H),3.31–3.22(m,2H),3.04–2.92(m,2H),2.07–1.97(m,4H).
步骤12:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(100mg,0.34mmol),4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶(134mg,0.37mmol),N,N-二异丙基乙胺(219mg,1.69mmol)加入到乙腈(6mL)中,60℃反应过夜。冷却至室温,减压除去溶剂,加入水(20mL),用乙酸乙酯萃取(15mL×2),用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=50%)得到白色固体产物165mg,产率78.7%。
LC-MS(ESI):[M+H]+=618.5;
1HNMR(500MHz,CDCl3)δ8.17(d,J=1.6Hz,1H),7.96(dd,J=8.5,1.6Hz,1H),7.74(d,J=8.5Hz,1H),7.28(s,2H),7.22–7.14(m,2H),6.97(t,J=7.1Hz,2H),6.03(d,J=6.6Hz,1H),5.28(s,2H),5.24–5.18(m,1H),4.78–4.59(m,3H),4.43–4.37(m,1H),3.95(s,5H),3.07–2.91(m,3H),2.77–2.70(m,1H),2.51–2.41(m,1H),2.32–2.20(m,2H),1.86–1.79(m,2H),1.76–1.65(m,2H).
步骤13:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(165mg,0.27mmol)和氢氧化锂一水合物(56mg,1.33mmol)加入到1,4-二氧六环(4mL)和水(2mL)中,40℃反应1h。在冰浴中冷却,加水(15mL),用醋酸调节pH=6,用 乙酸乙酯萃取(15mL×2),合并有机相,用无水硫酸钠干燥,减压浓缩,硅胶柱层析纯化(洗脱剂MeOH:DCM(v/v)=10%)得到白色固体产物69mg,产率40.1%。
LC-MS(ESI):[M+H]+=604.5;
1HNMR(500MHz,DMSO-d6)δ12.74(br.s,1H),8.26(s,1H),7.80(d,J=8.4Hz,1H),7.64(d,J=8.4Hz,1H),7.47–7.40(m,2H),7.22–7.10(m,3H),6.93(t,J=7.4Hz,1H),6.52(d,J=4.2Hz,1H),5.35(s,2H),5.12–5.05(m,1H),4.81–4.74(m,1H),4.67–4.60(m,1H),4.51–4.46(m,1H),4.39–4.33(m,1H),3.91(d,J=13.6Hz,1H),3.76(d,J=13.6Hz,1H),3.01–2.81(m,3H),2.74–2.65(m,1H),2.45–2.36(m,1H),2.20–2.05(m,2H),1.70–1.54(m,4H)。
实施例2(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物2)的合成
步骤1:7-((2-溴苯氧基)甲基)-4-氯-2-氟苯并呋喃的合成
将化合物2-溴苯酚(207mg,1.20mmol)和7-(溴甲基)-4-氯-2-氟苯并呋喃(300mg,1.14mmol)加入到N,N-二甲基甲酰胺(8mL)中,然后加入碳酸钾(472mg,3.42mmol)。60℃反应1h。加入水(50mL)淬灭反应,用乙酸乙酯萃取(15mL×3),合并有机相。饱和食盐水洗涤(5mL×2),有机相用无水硫酸钠干燥,减压浓缩,硅胶柱层析分离纯化(洗脱剂EA:PE(v/v)=5%),得黄色液体产物341mg,产率78.0%。
1H NMR(500MHz,CDCl3)δ7.57(dd,J=7.9,1.7Hz,1H),7.43(d,J=8.2Hz,1H),7.28(d,J=8.5Hz,1H),7.25–7.23(m,1H),7.02–6.97(m,1H),6.87(td,J=7.7,1.4Hz,1H),6.02(d,J=6.6Hz,1H),5.36(s,2H).
步骤2:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将化合物7-((2-溴苯氧基)甲基)-4-氯-2-氟苯并呋喃(280mg,0.79mmol)和N-Boc-1,2,5,6-四氢吡啶-4-硼酸频哪醇酯(244mg,0.79mmol)加入到1,4-二氧六环(6mL)和水(1.5mL)的混合溶液中,然后加入[1,1'-双(二苯基膦)二茂铁]二氯化钯(33mg,0.04mmol)和碳酸钾(220mg,1.59mmol)。氮气保护下加热至90℃反应2h。冷却至室温,加水稀释(30mL),乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥。过滤,浓缩滤液,硅胶柱层析分离纯化(洗脱剂EA:PE(v/v)=5%),得到无色液体355mg,产率98.5%。
LC-MS(ESI):[M-55]+=402.2;
1H NMR(500MHz,CDCl3)δ7.25(s,2H),7.23(s,1H),7.16(dd,J=7.4,1.8Hz,1H),6.96(t,J=7.8Hz,2H),6.02(d,J=6.6Hz,1H),5.73(s,1H),5.27(s,2H),4.01(s,2H),3.51(d,J=5.9Hz,2H),2.48(s,2H),1.48(s,9H).
步骤3:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)-1,2,3,6-四氢吡啶的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(341mg,0.74mmol)加入到二氯甲烷(5mL)中,然后在冰浴下加入三氟乙酸(2.55g,22.34mmol)。恢复至室温反应30min。加饱和碳酸氢钠溶液调节PH=8,用乙酸乙酯萃取(20mL×3)。合并有机相,用无水硫酸钠干燥,过滤,减压除去溶剂,得到棕褐色固体302mg,粗产物直接用于下一步。
LC-MS(ESI):[M+H]+=358.1。
步骤4:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁 烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)-1,2,3,6-四氢吡啶(150mg,0.34mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(100mg,0.34mmol),N,N-二异丙基乙胺(220mg,1.70mmol)加入到乙腈(5mL)中,60℃反应过夜。冷却至室温,加水(20mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=50%),得到黄色固体产物171mg,产率81.8%。
LC-MS(ESI):[M+H]+=616.4;
1H NMR(500MHz,CDCl3)δ8.16(s,1H),8.00–7.94(m,1H),7.75(d,J=8.5Hz,1H),7.28–7.15(m,4H),6.96(d,J=7.9Hz,2H),6.01(d,J=6.6Hz,1H),5.75(s,1H),5.27(s,2H),5.21–5.15(m,1H),4.74–4.53(m,3H),4.40–4.32(m,1H),3.95(s,3H),3.19(s,2H),2.73(t,J=5.7Hz,2H),2.70–2.62(m,1H),2.53(br.s,2H),2.43–2.37(m,1H),2.29–2.18(m,1H),2.06–1.99(m,1H).
步骤5:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(170mg,0.28mmol)和氢氧化锂一水合物(58mg,1.38mmol)加入到1,4-二氧六环(4mL)和水(2mL)中,40℃反应1.5h。冷却至室温,加水稀释(15mL),用醋酸(4M)调节体系pH至6,乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩滤液。硅胶柱层析纯化(洗脱剂MeOH:DCM(v/v)=2%)得到淡黄色固体产物89mg,产率53.6%。
LC-MS(ESI):[M+H]+=602.4;
1H NMR(500MHz,DMSO-d6)δ12.78(br.s,1H),8.25(s,1H),7.81(d,J=8.5Hz,1H),7.65(d,J=8.5Hz,1H),7.41(s,2H),7.24(t,J=8.0Hz,1H),7.17–7.10(m,2H),6.94(t,J=7.5Hz,1H),6.49(d,J=6.3Hz,1H),5.69(s,1H),5.32(s,2H),5.12–4.95(m,1H),4.79–4.69m,1H),4.66–4.55(m,1H),4.46–4.38(m,1H),4.35–4.28(m,1H),4.00(d,J=13.9Hz,1H),3.86(d,J=13.6Hz,1H),3.14–3.02(m,2H),2.74–2.55(m,3H)2.44–2.37(m,2H),2.35–2.27(m,1H).
实施例3 2-((2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-2,3,4,5-四氢-[1,1'-联苯]-4-基)甲基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸(化合物3)的合成
步骤1:2-(4-(((三氟甲基)磺酰基)氧基)环己-3-烯-1-基)乙酸乙酯的合成
将化合物4-氧代环己烷乙酸乙酯(3.0g,16.28mmol)和N-苯基双(三氟甲烷磺酰)亚胺(6.98g,19.54mmol)加入到无水四氢呋喃(30mL)中,氮气保护下-40℃冷却,加双(三甲基硅烷基)氨基锂(1mol/L的THF溶液)(21.2mL,21.2mmol),此温度下反应2h。加入水(50mL)淬灭反应,用乙酸乙酯萃取(30mL×2),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=2%)得到无色液体产物4.1g,产率79.6%。
1HNMR(500MHz,CDCl3)δ5.72(s,1H),4.15(q,J=7.1Hz,2H),2.49–2.39(m,1H),2.38–2.31(m,2H),2.30(d,J=7.1Hz,2H),2.18–2.09(m,1H),1.97–1.89(m,2H),1.57–1.48(m,1H),1.26(t,J=7.1Hz,3H).
步骤2:2-(4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)环己-3-烯-1-基)乙酸乙酯的合成
将化合物2-(4-(((三氟甲基)磺酰基)氧基)环己-3-烯-1-基)乙酸乙酯(3.9g,12.33mmol),联硼酸频那醇酯(3.29g,12.95mmol),四三苯基膦钯(710mg,0.62mmol)和醋酸钾(2.42g,24.66mmol),加入到1,4-二氧六环(30mL)中,氮气保护下90℃反应1h,冷却至室温,加入水(100mL)淬灭反应,用乙酸乙酯萃取(50mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=5%)得到无色液体产物2.0g,产率55.1%。
1HNMR(500MHz,CDCl3)δ6.50(s,1H),4.15–4.10(m,2H),2.30–2.19(m,4H),2.15–2.03(m,2H),1.84–1.73(m,2H),1.40–1.28(m,1H),1.25(s,12H),1.28–1.23(m,3H).
步骤3:7-((2-溴苯氧基)甲基)-4-氯-2-氟苯并呋喃的合成
将化合物7-(溴甲基)-4-氯-2-氟苯并呋喃(320mg,1.20mmol)和2-溴苯酚(208mg,1.20mmol)加入到N,N-二甲基甲酰胺(8mL)中,然后加入碳酸钾(450mg,3.26mmol)。60℃反应1h。加入水(40mL)淬灭反应,用乙酸乙酯萃取(20mL×3),合并有机相。饱和食盐水洗涤(15mL×3),有机相用无水硫酸钠干燥,减压浓缩,硅胶柱层析分离纯化(洗脱剂EA:PE(v/v)=5%),得白色固体产物402mg,产率93.1%。
1H NMR(500MHz,CDCl3)δ7.57(dd,J=7.9,1.7Hz,1H),7.43(d,J=8.2Hz,1H),7.28(d,J=8.5Hz,1H),7.24(d,J=1.7Hz,1H),7.06–6.96(m,1H),6.87(td,J=7.7,1.4Hz,1H),6.02(d,J=6.6Hz,1H),5.36(s,2H).
步骤4:2-(2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-2,3,4,5-四氢-[1,1'-联苯]-4-基)乙酸乙酯的合成
将化合物7-((2-溴苯氧基)甲基)-4-氯-2-氟苯并呋喃(402mg,1.13mmol)和2-(4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)环己-3-烯-1-基)乙酸乙酯(345mg,1.17mmol)加入到1,4-二氧六环(12mL)和水(3mL)的混合溶液中,然后加入[1,1'-双(二苯基膦)二茂铁]二氯化钯(47mg,0.06mmol)和碳酸钾(315mg,2.28mmol)。氮气保护下加热至90℃反应2h。冷却至室温,加水稀释(50ml),乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥。过滤,浓缩滤液,硅胶柱层析分离纯化(洗脱剂EA:PE(v/v)=5%),得到黄色液体366mg,产率73.1%。
LC-MS(ESI):[M-55]+=443.1。
步骤5:2-(2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-2,3,4,5-四氢-[1,1'-联苯]-4-基)乙酸的合成
将化合物2-(2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-2,3,4,5-四氢-[1,1'-联苯]-4-基)乙酸乙酯(366mg,0.83mmol)溶解在1,4-二氧六环(8mL)和水(4mL)中,加入氢氧化锂一水合物(105mg,2.50mmol),40℃反应2h,冷却至室温,加水(8mL)稀释,用稀盐酸调节pH=5,用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,得到米白色固体295mg,产率86.1%。
LC-MS(ESI):[M+H]+=415.1;
1H NMR(500MHz,DMSO-d6)δ12.01(s,1H),7.45–7.37(m,2H),7.22(td,J=7.8,1.8Hz,1H),7.12(d,J=8.2Hz,1H),7.08(dd,J=7.4,1.8Hz,1H),6.92(t,J=7.4Hz,1H),6.51(d,J=6.4Hz,1H),5.59(dd,J=4.8,2.7Hz,1H),5.31(s,2H),2.31(t,J=5.4Hz,2H),2.19(d,J=7.2Hz,2H),1.98–1.90(m,1H),1.83–1.69(m,2H),1.32–1.22(m,2H).
步骤6:4-(2-(2'-((4-氯-2-氟苯并呋喃-7-基)甲基)-2,3,4,5-四氢-[1,1'-联苯]-4-基)乙酰氨基)-3-((((S)-氧杂环丁烷-2-基)甲基)氨基)苯甲酸甲酯的合成
将化合物2-(2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-2,3,4,5-四氢-[1,1'-联苯]-4-基)乙酸(145mg,0.35mmol)和(S)-4-氨基-3-((氧杂环丁烷-2-基甲基)氨基)苯甲酸甲酯(75mg,0.32mmol)加入到二氯甲烷(6mL)和N,N-二甲基甲酰胺(3mL)中,加入N,N,N',N'-四甲基-O-(7-氮杂苯并***-1-基)六氟磷酸脲(182mg,0.48mmol)和N,N-二异丙基乙胺(103mg,0.80mmol),45℃反应过夜。减压除去溶剂,加入水(60mL),用乙酸乙酯萃取(20mL×2),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂 EA:PE(v/v)=20%)得到橙粉色固体137mg,产率68.1%。
LC-MS(ESI):[M+H]+=633.5;
1H NMR(500MHz,DMSO-d6)δ9.40(s,1H),7.43(d,J=8.1Hz,1H),7.41(s,2H),7.30–7.19(m,3H),7.15–7.07(m,2H),6.93(t,J=7.4Hz,1H),6.50(d,J=6.4Hz,1H),5.63(s,1H),5.32(s,2H),5.26(s,1H),4.97–4.88(m,1H),4.54–4.48(m,1H),4.45–4.40(m,1H),3.82(s,3H),3.41–3.34(m,2H),2.68–2.58(m,1H),2.48–2.44(m,1H),2.40–2.30(m,4H),2.29–2.23(m,1H),2.11–2.02(m,1H),1.93–1.74(m,2H).
步骤7:2-((2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-2,3,4,5-四氢-[1,1'-联苯]-4-基)甲基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-(2-(2'-((4-氯-2-氟苯并呋喃-7-基)甲基)-2,3,4,5-四氢-[1,1'-联苯]-4-基)乙酰氨基)-3-((((S)-氧杂环丁烷-2-基)甲基)氨基)苯甲酸甲酯(130mg,0.21mmol)加入到冰醋酸(5mL)中,80℃反应3h。减压除去溶剂,加入水(10mL),用碳酸氢钠溶液(2M)调节体系pH至8,二氯甲烷(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,硅胶柱层析纯化(洗脱剂EA:PE(v/v)=30%)得到无色液体115mg,产率91.1%。
LC-MS(ESI):[M+H]+=615.4。
步骤8:2-((2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-2,3,4,5-四氢-[1,1'-联苯]-4-基)甲基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物2-((2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-2,3,4,5-四氢-[1,1'-联苯]-4-基)甲基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(115mg,0.19mmol)和氢氧化锂一水合物(40mg,0.95mmol)加入到1,4-二氧六环(4mL)和水(2mL)中,40℃反应1.5h。冷却至室温,加水稀释(15mL),用醋酸(4M)调节体系pH至5,乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩滤液。硅胶柱层析纯化(洗脱剂MeOH:DCM(v/v)=5%)得到白色固体产物85mg,产率73.9%。
LC-MS(ESI):[M+H]+=601.3;
1H NMR(500MHz,DMSO-d6)δ12.65(br.s,1H),8.20(s,1H),7.79(d,J=8.4Hz,1H),7.62(d,J=8.4Hz,1H),7.41(s,2H),7.26–7.17(m,1H),7.11(dd,J=12.4,7.7Hz,2H),6.96–6.88(m,1H),6.49(d,J=6.3Hz,1H),5.61(s,1H),5.32(s,2H),5.05–4.95(m,1H),4.69–4.55(m,1H),4.52–4.37(m,2H),4.33–4.24(m,1H),3.01–2.86(m,2H),2.72–2.62(m,1H),2.40–2.22(m,5H),2.03–1.78(m,2H),1.48–1.36(m,1H).
实施例4(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物4)的合成
步骤1:2-(苄氧基)-1-溴-3-氟苯的合成
将化合物7-(溴甲基)-4-氯-2-氟苯并呋喃(500mg,2.62mmol)和溴化苄(450mg,2.62mmol)加入到N,N-二甲基甲酰胺(5mL)中,然后加入碳酸钾(900mg,6.54mmol)。60℃反应4h。加入水(50mL)淬灭反应,用乙酸乙酯萃取(15mL×3),合并有机相。加水洗涤(15mL×2)饱和食盐水洗涤(10mL×1),有机相用无水硫酸钠干燥,减压浓缩,硅胶柱层析分离纯化(洗脱剂EA:PE(v/v)=5%),得淡黄色液体产物730mg,产率99.2%。
LC-MS(ESI):[M+H]+=282.3;
1H NMR(500MHz,CDCl3)δ7.55–7.50(m,2H),7.41–7.29(m,4H),7.09–7.02(m,1H),6.95–6.87(m,1H),5.13(s,2H).
步骤2:4-(2-(苄氧基)-3-氟苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将化合物2-(苄氧基)-1-溴-3-氟苯(730mg,2.60mmol)和N-Boc-1,2,5,6-四氢吡啶-4-硼酸频哪醇酯(840mg,2.73mmol)加入到1,4-二氧六环(20mL)和水(5mL)的混合溶液中,然后加入[1,1'-双(二苯基膦)二茂铁]二氯化钯(110mg,0.13mmol)和碳酸钾(720mg,5.19mmol)。氮气保护下加热至90℃反应2h。冷却至室温,加水稀释(20ml),乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥。过滤,浓缩滤液,硅胶柱层析分离纯化(洗脱剂EA:PE(v/v)=10%),得到浅黄色液体985mg,产率98.8%。
LC-MS(ESI):[M-55]+=328.2;
1H NMR(500MHz,CDCl3)δ7.37–7.28(m,5H),7.06–6.94(m,2H),6.92–6.87(m,1H),5.70(s,1H),4.99(s,2H),3.98(s,2H),3.51(t,J=5.5Hz,2H),2.43–2.39(m,2H),1.49(s,9H).
步骤3:4-(3-氟-2-羟苯基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(2-(苄氧基)-3-氟苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(985mg,2.59mmol)溶解在甲醇(20mL)中,加入钯碳(10%Pd,含55%水)(250mg,),用氢气置换三次,在氢气环境下室温反应过夜。硅藻土过滤,滤液减压浓缩,得淡黄色液体717mg,产率93.6%。
LC-MS(ESI):[M-55]+=240.1。
步骤4:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(3-氟-2-羟苯基)哌啶-1-羧酸叔丁酯(350mg,1.19mmol)和7-(溴甲基)-4-氯-2-氟苯并呋喃(450mg,2.62mmol)加入到N,N-二甲基甲酰胺(8mL)中,然后加入碳酸钾(400mg,2.89mmol)。60℃反应2h。加入水(50mL)淬灭反应,用乙酸乙酯萃取(15mL×3),合并有机相。饱和食盐水洗涤(5mL×2),有机相用无水硫酸钠干燥,减压浓缩,硅胶柱层析分离纯化(洗脱剂EA:PE(v/v)=5%),得黄色液体产物527mg,产率96.8%。
LC-MS(ESI):[M-55]+=422.2;
1H NMR(500MHz,CDCl3)δ7.25(s,2H),7.04–6.95(m,2H),6.92–6.86(m,1H),6.03(d,J=6.6Hz,1H),5.29(s,2H),4.23–4.08(m,2H),3.04–2.95(m,1H),2.71–2.51(m,2H),1.52–1.47(m,4H),1.46(s,9H).
步骤5:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-羧酸叔丁酯(528mg,1.10mmol)加入到二氯甲烷(6mL)中,然后在冰浴下加入三氟乙酸(2.77g,24.30mmol)。升温至室温反应30min。加饱和碳酸氢钠溶液调节PH=8,用乙酸乙酯萃取(10mL×3)。合并有机相,用无水硫酸钠干燥,过滤,减压除去溶剂,得到棕黄色液体412mg,产率98.7%。
LC-MS(ESI):[M+H]+=378.2。
步骤6:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶(140mg,0.37mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(100mg,0.34mmol),N,N-二异丙基乙胺(220mg,1.70mmol)加入到乙腈(5mL)中,60℃反应过夜。冷却至室温,加水(20mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=50%),得到浅黄色油状产物181mg,产率83.9%。
LC-MS(ESI):[M+H]+=636.3。
步骤7:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基 甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(181mg,0.28mmol)和氢氧化锂一水合物(60mg,1.43mmol)加入到1,4-二氧六环(6mL)和水(3mL)中,40℃反应1.5h。冷却至室温,加水稀释(15mL),用醋酸(4M)调节体系pH至5,乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩滤液。硅胶柱层析纯化(洗脱剂MeOH:DCM(v/v)=5%)得到黄色固体产物85mg,产率73.9%。
LC-MS(ESI):[M+H]+=622.4;
1H NMR(500MHz,DMSO-d6)δ8.27(s,1H),7.81(s,1H),7.65(s,1H),7.38(d,J=35.3Hz,2H),7.18–7.05(m,2H),7.00(s,1H),6.55(s,1H),5.27(s,2H),5.09–5.06(m,1H),4.80–4.73(m,1H),4.66–4.60(m,1H),4.54–4.45(m,1H),4.41–4.30(m,1H),3.98–3.83(m,1H),3.80–3.69(m,1H),3.02–2.87(m,1H),2.84–2.61(m,3H),2.47–2.31(m,1H),2.11–1.80(m,3H),1.62–1.31(m,3H).
实施例5(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-3-(氧杂环丁烷-2-基甲基)-3H-咪唑并[4,5-b]吡啶-5-羧酸(化合物5)的合成
步骤1:6-氯-5-硝基-2-甲酸吡啶的合成
将化合物2-氯-6-甲基-3-硝基吡啶(8.0g,46.36mmol)加入到浓硫酸(40mL)中,冰浴中缓慢加入重铬酸钾(20.46g,69.54mmol),室温搅拌反应过夜。冰浴中冷却,缓慢加入冰水(500mL),用乙酸乙酯萃取(150mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,得到淡黄色固体8.26g,产率88.0%。
LC-MS(ESI):[M+H]+=203.0。
步骤2:6-氯-5-硝基-2-甲酸甲酯吡啶的合成
将化合物6-氯-5-硝基-2-甲酸吡啶(200mg,0.99mmol)溶解在甲醇(15mL)中,加入浓硫酸(0.5mL,0.99mmol),加热至68℃反应过夜。浓缩溶剂,加水稀释(30mL),乙酸乙酯萃取三次(15ml×3),合并有机相,无水硫酸钠干燥,过滤,浓缩滤液。用硅胶柱层析纯化(洗脱剂PE:EtOAc(v/v)=2:1)得到白色固体185mg,产率86.5%。
LC-MS(ESI):[M+H]+=217.1;
1H NMR(500MHz,CDCl3)δ8.31(dd,J=8.3,0.8Hz,1H),8.22(dd,J=8.2,0.8Hz,1H),4.04(d,J=0.9Hz,3H).
步骤3:(S)-5-硝基-6-((氧杂环丁烷-2-基甲基)氨基)吡啶甲酸甲酯的合成
将化合物6-氯-5-硝基-2-甲酸甲酯吡啶(1.3g,6.0mmol)和(S)-氧杂环丁烷-2-甲胺(0.58g,6.6mmol)溶于四氢呋喃(20mL),然后加入碳酸钾(1.24g,9.00mmol),室温搅拌过夜。硅藻土过滤,浓缩滤液。用硅胶柱层析纯化(洗脱剂PE:EtOAc(v/v)=2:1)得到黄色固体1.09g,产率68.0%。
LC-MS(ESI):[M+H]+=268.2;
1H NMR(500MHz,DMSO-d6)δ8.58(dd,J=8.4,1.0Hz,1H),8.51(t,J=5.8Hz,1H),7.31(dd,J=8.4,1.0Hz,1H),5.01–4.93(m,1H),4.56–4.52(m,1H),4.48–4.45(m,1H),3.96–3.87(m,4H),3.82(dt,J=13.8,5.2Hz,1H),2.68–2.58(m,1H),2.53–2.49(m,1H).
步骤4:(S)-5-氨基-6-((氧杂环丁烷-2-基甲基)氨基)吡啶甲酸甲酯的合成
将化合物(S)-5-硝基-6-((氧杂环丁烷-2-基甲基)氨基)吡啶甲酸甲酯(1.09g,4.08mmol)溶于四氢呋喃(10mL)和甲醇(10mL)的混合溶液中,然后加入钯碳(10%Pd,含55%水)(185mg,1.74mmol),氢气保护。室温搅拌反应8小时。硅藻土过滤,浓缩滤液。硅胶柱层析分离纯化(洗脱剂DCM:MeOH(v/v)=20:1)得到油黄色粘稠物885mg,产率91.5%。
LC-MS(ESI):[M+H]+=238.1;
1H NMR(500MHz,DMSO-d6)δ7.23(d,J=7.8Hz,1H),6.68(d,J=7.8Hz,1H),5.95(t,J=5.6Hz,1H),5.61(s,2H),4.92–4.87(m,1H),4.54–4.50(m,1H),4.45(dt,J=9.0,5.9Hz,1H),3.72(s,4H),3.62(dt,J=13.8,5.2Hz,1H),2.65–2.57(m,1H),2.44–2.39(m,1H).
步骤5:(S)-5-(2-氯乙酰氨基)-6-((氧杂环丁烷-2-基甲基)氨基)吡啶甲酸甲酯的合成
将化合物(S)-5-氨基-6-((氧杂环丁烷-2-基甲基)氨基)吡啶甲酸甲酯(500mg,2.11mmol)溶解在四氢呋喃(10mL)溶液中。加入N,N-二异丙基乙胺(817mg,6.32mmol),冰浴中冷却,加入氯乙酰氯(285mg,2.52mmol),室温反应3h,加入水(50mL),用乙酸乙酯萃取(20mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc=100%)得到浅褐色液体569mg,产率86%。
LC-MS(ESI):[M+H]+=314.2;
1H NMR(500MHz,CDCl3)δ8.30(br.s,1H),7.88(d,J=7.9Hz,1H),7.53(d,J=7.9Hz,1H),5.11–5.01(m,2H),4.73–4.69(m,1H),4.0–4.53(m,1H),4.22(s,2H),3.93(s,3H),3.80–3.73(m,1H),2.72–2.64(m,1H),2.59–2.49(m,1H).
步骤6:(S)-2-(氯甲基)-3-(氧杂环丁烷-2-基甲基)-3H-咪唑并[4,5-b]吡啶-5-羧酸甲酯的合成
将化合物(S)-5-(2-氯乙酰氨基)-6-((氧杂环丁烷-2-基甲基)氨基)吡啶甲酸甲酯(300mg,0.24mmol)溶于醋酸(5mL)中,加热至60℃,反应过夜。减压除去溶剂,加入饱和碳酸氢钠调节体系pH至9,用乙酸乙酯萃取(20mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,硅胶柱层析纯化(洗脱剂EtOAc=100%),得到淡黄色固体101mg,产率35.7%。
LC-MS(ESI):[M+H]+=296.2;
1H NMR(500MHz,CDCl3)δ8.17–7.11(m,2H),5.26–5.19(m,1H),5.15–5.03(m,2H),4.85–4.71(m,2H),4.65–4.55(m,1H),4.36–4.28(m,1H),4.01(s,3H),2.84–2.73(m,1H),2.49–2.40(m,1H).
步骤7:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-3-(氧杂环丁烷-2-基甲基)-3H-咪唑并[4,5-b]吡啶-5-羧酸甲酯的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶(147mg,0.39mmol),(S)-2-(氯甲基)-3-(氧杂环丁烷-2-基甲基)-3H-咪唑并[4,5-b]吡啶-5-羧酸甲酯(100mg,0.34mmol),N,N-二异丙基乙胺(220mg,1.70mmol)加入到乙腈(10mL)中,60℃反应过夜。冷却至室温,加水(20mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=50%),得到黄色油状产物190mg,产率88.2%。
LC-MS(ESI):[M+H]+=637.4。
步骤8:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-3-(氧杂环丁烷-2-基甲基)-3H-咪唑并[4,5-b]吡啶-5-羧酸的合成
将化合物(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-3-(氧杂环丁烷-2-基甲基)-3H-咪唑并[4,5-b]吡啶-5-羧酸甲酯(190mg,0.30mmol)和氢氧化锂一水合物(63mg,1.50mmol)加入到1,4-二氧六环(6mL)和水(3mL)中,40℃反应1.5h。冷却至室温,加水稀释(15mL),用醋酸(4M)调节体系pH至5,乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩滤液。硅胶柱层析纯化(洗脱剂MeOH:DCM(v/v)=5%)得到淡黄色固体产物45mg,产率22.4%。
LC-MS(ESI):[M+H]+=623.4;
1H NMR(500MHz,DMSO-d6)δ8.15(d,J=8.2Hz,1H),8.00(d,J=8.3Hz,1H),7.41(d,J=8.1Hz,1H),7.34(d,J=8.1Hz,1H),7.17–7.05(m,2H),7.01(d,J=7.8Hz,1H),6.53(d,J=6.3Hz,1H),5.27(s,2H),5.18–5.23(m,1H),4.85–4.81(m,1H),4.73–4.67(m,1H),4.53–4.45(m,1H),4.40–4.32(m,1H),4.00–3.84(m,2H),2.96–2.81(m,2H),2.75–2.65(m,2H),2.50–2.43(m,1H),2.07–1.93(m,2H),1.58–1.46(m,2H),1.37–1.29(m,2H).
实施例6(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-基)甲基)-3-(氧杂环丁烷-2-基甲基)-3H-咪唑并[4,5-b]吡啶-5-羧酸(化合物6)的合成
步骤1:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-基)甲基)-3-(氧杂环丁烷-2-基甲基)-3H-咪唑并[4,5-b]吡啶-5-羧酸甲酯的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶(150mg,0.42mmol),(S)-2-(氯甲基)-3-(氧杂环丁烷-2-基甲基)-3H-咪唑并[4,5-b]吡啶-5-羧酸甲酯(117mg,0.40mmol),N,N-二异丙基乙胺(256mg,1.98mmol)加入到乙腈(10mL)中,60℃反应过夜。冷却至室温,加水(20mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=50%),得到黄色液体产物240mg,产率98.0%。
LC-MS(ESI):[M+H]+=619.5。
步骤2:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-基)甲基)-3-(氧杂环丁烷-2-基甲基)-3H-咪唑并[4,5-b]吡啶-5-羧酸的合成
将化合物(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-基)甲基)-3-(氧杂环丁烷-2-基甲基)-3H-咪唑并[4,5-b]吡啶-5-羧酸甲酯(240mg,0.39mmol)和氢氧化锂一水合物(82mg,1.95mmol)加入到1,4-二氧六环(8mL)和水(4mL)中,40℃反应1.5h。冷却至室温,加水稀释(15mL),用醋酸(4M)调节体系pH至5,乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩滤液。硅胶柱层析纯化(洗脱剂MeOH:DCM(v/v)=5%)得到白色固体产物46mg,产率16.7%。
LC-MS(ESI):[M+H]+=605.4;
1H NMR(500MHz,DMSO-d6)δ8.10(d,J=8.2Hz,1H),7.99(d,J=8.2Hz,1H),7.45–7.41(m,2H),7.21–7.14(m,2H),7.12(d,J=8.2Hz,1H),6.93(t,J=7.4Hz,1H),6.53(d,J=6.4Hz,1H),5.35(s,2H),5.19–5.15(m,1H),4.84–4.71(m,2H),4.52–4.45(m,1H),4.35–4.30(m,1H),3.94(d,J=13.7Hz,1H),3.88(d,J=13.6Hz,1H),2.98–2.87(m,3H),2.73–2.59(m,1H),2.50–2.43(m,1H),2.21–2.11(m,2H),1.72–1.55(m,4H).
实施例7(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)环己基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物7)的合成
步骤1:1-(苄氧基)-2-溴苯的合成
将化合物2-溴苯酚(3.0g,17.34mmol)和溴化苄(3.0mg,17.54mmol)加入到N,N-二甲基甲酰胺(20mL)中,然后加入碳酸钾(6.0g,43.42mmol)。60℃反应3h。加入水(100mL)淬灭反应,用乙酸乙酯萃取(30mL×3),合并有机相。加水洗涤(30mL×2)饱和食盐水洗涤(15mL×1),有机相用无水硫酸钠干燥,减压浓缩,硅胶柱层析分离纯化(洗脱剂EA:PE(v/v)=5%),得无色液体产物4.21g,产率92.3%。
1H NMR(500MHz,CDCl3)δ7.55(d,J=7.6Hz,1H),7.47(d,J=7.6Hz,2H),7.38(t,J=7.6Hz,2H),7.31(t,J=7.4Hz,1H),7.25–7.18(m,1H),6.93(d,J=8.2Hz,1H),6.83(t,J=7.6Hz,1H),5.15(s,2H).
步骤2:2-(2'-(苄氧基)-2,3,4,5-四氢-[1,1'-联苯基]-4-基)乙酸乙酯的合成
将化合物1-(苄氧基)-2-溴苯(1.8g,6.84mmol)和2-(4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)环己-3-烯-1-基)乙酸乙酯(2.0g,6.80mmol)加入到1,4-二氧六环(48mL)和水(12mL)的混合溶液中,然后加入[1,1'-双(二苯基膦)二茂铁]二氯化钯(0.3g,0.37mmol)和碳酸钾(1.9g,13.75mmol)。氮气保护下加热至90℃反应2h。冷却至室温,加水稀释(50ml),乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥。过滤,浓缩滤液,硅胶柱层析分离纯化(洗脱剂EA:PE(v/v)=5%),得到浅黄色液体1.46g,产率61.1%。
LC-MS(ESI):[M+H]+=351.3;
1H NMR(500MHz,CDCl3)δ7.40(d,J=7.0Hz,2H),7.36(t,J=7.7Hz,2H),7.29(t,J=7.2Hz,1H),7.17(td,J=7.7,1.8Hz,1H),7.14(dd,J=7.4,1.8Hz,1H),6.94–6.88(m,2H),5.74–5.70(m,1H),5.06(s,2H),4.15(q,J=7.2Hz,2H),2.51–2.45(m,2H),2.36–2.30(m,3H),2.22–2.16(m,1H),1.96–1.81(m,2H),1.49–1.40(m,1H),1.27(t,J=7.0Hz,3H).
步骤3:2-(2'-羟基-2,3,4,5-四氢-[1,1'-联苯基]-4-基)乙酸乙酯的合成
将化合物2-(2'-(苄氧基)-2,3,4,5-四氢-[1,1'-联苯基]-4-基)乙酸乙酯(1.45g,4.14mmol)溶解在乙醇(20mL)中,加入钯碳(10%Pd,含55%水)(0.4g,1.69mmol),用氢气置换三次,在氢气环境下室温反应过夜。硅藻土过滤,滤液减压浓缩,得灰棕色液体978mg,产率90.1%。
LC-MS(ESI):[M-55]+=263.2;
1H NMR(500MHz,CDCl3)δ7.21–7.14(m,1H),7.08–7.02(m,1H),6.89(t,J=7.4Hz,1H),6.74(d,J=7.9Hz,1H),5.09(br,1H),4.15(q,J=7.1Hz,2H),2.91–2.78(m,1H),2.48(d,J=7.6Hz,1H),2.24(d,J=6.6Hz,1H),1.93–1.85(m,2H),1.75–1.63(m,5H),1.56–1.46(m,1H),1.30–1.24(m,3H),1.23–1.14(m,1H).
步骤4:2-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)环己基)乙酸乙酯的合成
将化合物2-(2'-羟基-2,3,4,5-四氢-[1,1'-联苯基]-4-基)乙酸乙酯(200mg,0.76mmol)和7-(溴甲基)-4-氯-2-氟苯并呋喃(200mg,0.76mmol)加入到N,N-二甲基甲酰胺(6mL)中,然后加入碳酸钾(262mg,1.90mmol)。60℃反应1.5h。加入水(50mL)淬灭反应,用乙酸乙酯萃取(15mL×3),合并有机相。饱和食盐水洗涤(5mL×2),有机相用无水硫酸钠干燥,减压浓缩,硅胶柱层析分离纯化(洗脱剂EA:PE(v/v)=5%),得无色液体产物251mg,产率74.3%。
LC-MS(ESI):[M-55]+=389.3;
1H NMR(500MHz,CDCl3)δ7.34–7.24(m,2H),7.26–7.18(m,1H),7.20–7.12(m,1H),6.96(t,J=7.5Hz,2H),6.02(d,J=6.6Hz,1H),5.28(s,2H),4.18–4.10(m,2H),3.04–2.93(m,1H),2.41(d,J=7.7Hz,1H),2.22(d,J=6.6Hz,1H),1.92–1.83(m,2H),1.64–1.60(m,1H),1.54–1.43(m,1H),1.34–1.29(m,1H),1.29–1.23(m,5H),1.20–1.09(m,1H),0.91–0.83(m,1H).
步骤5:2-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)环己基)乙酸的合成
将化合物2-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)环己基)乙酸乙酯(232mg,0.52mmol)加入到1,4-二氧六环(5mL)和水(2.5mL)中,氢氧化锂一水合物(110mg,2.62mmol),40℃反应2h,冷却至室温,加水(5mL)稀释,用稀盐酸调节pH=5,用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,得到淡粉色液体215mg,产率98.9%。
步骤6:(S)-4-(2-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)环己基)乙酰氨基)-3-((氧杂环丁烷-2-基甲基)氨基)苯甲酸甲酯的合成
将化合物2-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)环己基)乙酸(215mg,0.52mmol)和(S)-4-氨基-3-((氧杂环丁烷-2-基甲基)氨基)苯甲酸甲酯(76mg,0.32mmol)加入到二氯甲烷(6mL)和N,N-二甲基甲酰胺(3mL)中,加入2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(184mg,0.48mmol)和N,N-二异丙基乙胺(104mg,0.80mmol),45℃反应过夜。减压除去溶剂,加入水(60mL),用乙酸乙酯萃取(20mL×2),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=20%)得到棕褐色液体200mg,产率98.4%。
LC-MS(ESI):[M+H]+=635.4;
1H NMR(500MHz,CDCl3)δ8.01(s,1H),7.81–7.76(m,1H),7.70(d,J=19.5Hz,1H),7.56(d,J=8.3Hz,1H),7.52(s,1H),7.33–7.27(m,1H),7.23–7.14(m,1H),7.00–6.94(m,2H),6.04–6.00(m,1H),5.28(s,2H),5.08–5.00(m,1H),4.76–4.69(m,1H),4.62–4.55(m,1H),3.89(s,3H),3.44–3.37(m,1H),3.35–3.29(m,1H),3.06–2.98(m,1H),2.77–2.66(m,1H),2.57–2.47(m,2H),2.46–2.40(m,1H),2.34–2.30(m,1H),2.08–1.88(m,2H),1.57–1.46(m,1H),1.01–0.91(m,1H),0.91–0.82(m,4H).
步骤7:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)环己基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-4-(2-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)环己基)乙酰氨基)-3-((氧杂环丁烷-2-基甲基)氨基)苯甲酸甲酯(230mg,0.36mmol)加入到冰醋酸(9mL)中,80℃反应3h。减压除去溶剂,加入水(20mL),用碳酸氢钠溶液(2M)调节体系pH至8,二氯甲烷(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,硅胶柱层析纯化(洗脱剂EA:PE(v/v)=30%)得到无色液体215mg,产率96.2%。
LC-MS(ESI):[M+H]+=617.4;
1H NMR(500MHz,DMSO-d6)δ8.22(t,J=2.2Hz,1H),7.82–7.78(m,1H),7.64(d,J=8.4Hz,1H),7.43(d,J=2.2Hz,2H),7.30(dd,J=7.6,1.7Hz,1H),7.21–7.09(m,2H),6.99–6.90(m,1H),6.51(dd,J=11.8,6.4Hz,1H),5.35(d,J=6.5Hz,2H),5.06–4.99(m,1H),4.68–4.60(m,1H),4.55–4.43(m,2H),4.32–4.26(m,1H),3.87(s,3H),3.07(dd,J=9.8,7.5Hz,1H),2.97–2.82(m,1H),2.73–2.66(m,1H),2.41–2.33(m,1H),2.03–2.00(m,1H),1.93–1.82(m,1H),1.72(d,J=13.1Hz,4H),1.64–1.38(m,3H),1.28(d,J=20.0Hz,1H).
步骤8:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)环己基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)环己基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(210mg,0.34mmol)和氢氧化锂一水合物(72mg,1.72mmol)加入到1,4-二氧六环(4mL)和水(2mL)中,40℃反应1.5h。冷却至室温,加水稀释(15mL),用醋酸(4M)调节 体系pH至5,乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩滤液。硅胶柱层析纯化(洗脱剂MeOH:DCM(v/v)=5%)得到淡黄色固体产物85mg,产率40.3%。
LC-MS(ESI):[M+H]+=603.5;
1H NMR(500MHz,DMSO-d6)δ12.47(br,1H),8.20(s,1H),7.79(d,J=8.4Hz,1H),7.62(d,J=8.4Hz,1H),7.43(s,2H),7.30(d,J=7.6Hz,1H),7.22–7.09(m,2H),6.99–6.91(m,1H),6.54–6.48(m,1H),5.35(d,J=6.6Hz,2H),5.06–5.00(m,1H),4.67–4.59(m,1H),4.53–4.44(m,2H),4.34–4.28(m,1H),3.13–3.02(m,1H),2.99–2.81(m,2H),2.75–2.66(m,1H),2.42–2.33(m,1H),2.08–1.96(m,1H),1.86(d,J=12.8Hz,1H),1.78–1.68(m,4H),1.66–1.39(m,3H).
实施例8(S)-2-((4-(2-((4-氯苯并呋喃-7-基)甲氧基)苯基)哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物8)的合成
步骤1:4-(2-((4-氯苯并呋喃-7-基)甲氧基)-苯基)哌嗪-1-羧酸叔丁酯的合成
将4-(2-羟基苯基)哌嗪-1-羧酸叔丁酯(500mg,1.80mmol),7-(溴甲基)-4-氯苯并呋喃(440mg,1.79mmol),碳酸钾(496mg,3.59mmol)加入到N,N-二甲基甲酰胺(10mL)中,60℃下反应2小时。TLC检测反应原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=5%)得到无色油状产物789mg,产率99.2%。
LC-MS(ESI):[M+H]+=443.3;
1H NMR(500MHz,CDCl3)δ7.67(d,J=2.2Hz,1H),7.37(d,J=8.0Hz,1H),7.28–7.21(m,1H),7.04–6.90(m,5H),5.39(s,2H),3.56–3.47(m,4H),3.10–3.02(m,4H),1.48(s,9H).
步骤2:1-(2-((4-氯苯并呋喃-7-基)甲氧基)苯基)哌嗪的合成
将叔丁基4-(2-((4-氯苯并呋喃-7-基)甲氧基)-苯基)哌嗪-1-羧酸盐(789mg,1.78mmol),加入到三氟乙酸(5mL)和二氯甲烷(5mL)中,室温搅拌30分钟。TLC检测原料完全消失,停止反应。减压浓缩蒸干溶剂,用饱和的碳酸氢钠水溶液调节体系的PH至7左右,用乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩得到了棕色的固体产物599mg,产率:98.1%。
LC-MS(ESI):[M+H]+=343.2。
步骤3:(S)-2-((4-(2-((4-氯苯并呋喃-7-基)甲氧基)苯基)哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将1-(2-((4-氯苯并呋喃-7-基)甲氧基)苯基)哌嗪(150mg,0.44mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(128mg,0.43mmol),碳酸钾(120mg,0.87mmol),碘化钾(36mg,0.22mmol)加入到乙腈(10mL)中,60℃反应过夜。TLC检测原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=55%)得到无色油状产物162mg,产率61.6%。
LC-MS(ESI):[M+H]+=601.5;
1H NMR(500MHz,CDCl3)δ8.16(s,1H),7.97(dd,J=8.5,1.5Hz,1H),7.75(d,J=8.5Hz,1H),7.68–7.64(m,1H),7.37(d,J=8.0Hz,1H),7.27–7.26(m,1H),7.06–6.84(m,5H),5.39(s,2H),5.26–5.18(m,1H),4.77–4.57(m,3H),4.42–4.35(m,1H),4.02–3.96(m,2H),3.95(s,3H),3.23–3.02(m,4H),2.78–2.59(m,5H),2.49–2.40(m,1H).
步骤4:(S)-2-((4-(2-((4-氯苯并呋喃-7-基)甲氧基)苯基)哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将(S)-2-((4-(2-((4-氯苯并呋喃-7-基)甲氧基)苯基)哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(142mg,0.24mmol),氢氧化锂一水合物(49mg,1.17mmol)加入到水(2mL)和1.4-二氧六环(5mL)的混合溶液中,40℃反应2小时。TLC检测原料完全消失,停止加热。用20%的醋酸水溶液调节体系PH至7左右,用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=20%)得到白色固体产物70mg,产率49.5%。
LC-MS(ESI):[M+H]+=587.4;
1H NMR(500MHz,DMSO-d6)δ12.68(s,1H),8.28(s,1H),8.16(s,1H),7.82(d,J=8.4Hz,1H),7.66(d,J=8.4Hz,1H),7.48(d,J=8.0Hz,1H),7.39(d,J=8.0Hz,1H),7.13–7.05(m,2H),6.98–6.85(m,3H),5.37(s,2H),5.14–5.05(m,1H),4.82–4.73(m,1H),4.68–4.60(m,1H),4.52–4.45(m,1H),4.40–4.33(m,1H),3.95(d,J=13.5Hz,1H),3.79(d,J=13.6Hz,1H),3.07–2.91(m,4H),2.74–2.65(m,1H),2.62–2.47(m,4H),2.46–2.37(m,1H).
实施例9合成(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物9)的合成
步骤1:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-苯基)哌嗪-1-羧酸叔丁酯的合成
将4-(2-羟基苯基)哌嗪-1-羧酸叔丁酯(300mg,1.08mmol),7-(溴甲基)-4-氯-2-氟苯并呋喃(283mg,1.07mmol),碳酸钾(297mg,2.15mmol)加入到10mL的N,N-二甲基甲酰胺中,60℃下反应2小时。TLC检测反应原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=5%)得到无色油状产物469mg,产率94.4%。
1H NMR(500MHz,CDCl3)δ7.34(d,J=8.2Hz,1H),7.30–7.25(m,1H),7.03–6.91(m,4H),6.02(d,J=6.6Hz,1H),5.32(s,2H),3.58–3.47(m,4H),3.10–2.98(m,4H),1.48(s,9H).
步骤2:1-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌嗪的合成
将4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-苯基)哌嗪-1-羧酸叔丁酯(450mg,0.98mmol),加入到4mL的三氟乙酸和6mL的二氯甲烷中,室温搅拌30分钟。TLC检测原料完全消失,停止反应。减压浓缩溶剂,用饱和的碳酸氢钠水溶液调节体系的PH至7左右,用乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩得到了棕色的固体产物315mg,产率89.4%。
LC-MS(ESI):[M+H]+=361.2;
1H NMR(500MHz,CDCl3)δ7.28(s,2H),7.08–6.92(m,4H),6.03(d,J=6.5Hz,1H),5.29(s,2H),3.32–3.21(m,8H).
步骤3:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将1-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌嗪(150mg,0.42mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(147mg,0.50mmol),碳酸钾(114mg,0.82mmol),碘化钾(34mg,0.20mmol)加入到8mL的乙腈溶液中,60℃反应过夜。TLC检测原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=55%)得到无色油状产物249mg,产率96.7%。
LC-MS(ESI):[M+H]+=619.4;
1H NMR(500MHz,CDCl3)δ8.16(d,J=1.6Hz,1H),7.99(d,J=8.5Hz,1H),7.78(d,J=8.5Hz,1H),7.32(d,J=8.2Hz,1H),7.27(d,J=5.5Hz,1H),7.02–6.91(m,4H),6.02(d,J=6.6Hz,1H),5.31(s,2H),5.24–5.18(m,1H),4.84–4.74(m,1H),4.68–4.59(m,2H),4.46–4.38(m,1H),4.20–4.14(m,2H),3.95(s,3H),3.31–3.13(m,4H),2.96–2.80(m,4H),2.79–2.64(m,1H),2.52–2.42(m,1H).
步骤4:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(115mg,0.19mmol),氢氧化锂一水合物(101mg,2.41mmol)加入到4mL水和8mL 1,4-二氧六环的混合溶液中,40℃反应2小时。TLC检测原料完全消失,停止加热。用20%的醋酸水溶液调节体系PH至7左右,用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=20%)得到白色固体产物63mg,产率49.5%。
LC-MS(ESI):[M+H]+=605.4;
1H NMR(500MHz,DMSO-d6)δ12.47(s,1H),8.27(s,1H),7.81(d,J=8.4Hz,1H),7.65(d,J=8.5Hz,1H),7.49–7.43(m,2H),7.09(d,J=7.5Hz,1H),6.98–6.92(m,3H),6.54(d,J=6.4Hz,1H),5.33(s,2H),5.14–5.05(m,1H),4.84–4.74(m,1H),4.70–4.59(m,1H),4.54–4.45(m,1H),4.40–4.33(m,1H),3.95(d,J=13.5Hz,1H),3.79(d,J=13.6Hz,1H),3.02–2.95(m,4H),2.79–2.65(m,1H),2.61–2.52(m,4H),2.47–2.35(m,1H).
实施例10(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物10)的合成
步骤1:2-(苄氧基)-1-溴-3-氟苯的合成
将化合物2-溴-6氟苯酚(2.5g,13.09mmol)、苄溴(2.24g,13.09mmol)和碳酸钾(3.62g,26.18mmol)加入到20mL的N,N-二甲基甲酰胺中,60℃下反应4小时。TLC检测反应原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减 压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=10%)得到无色油状产物3.61g,产率98.1%。
LC-MS(ESI):[M+H]+=282.3;
1H NMR(500MHz,CDCl3)δ7.59–7.45(m,2H),7.43–7.28(m,4H),7.09–7.01(m,1H),6.94–6.86(m,1H),5.13(s,2H).
步骤2:4-(2-(苄氧基)-3-氟苯基)哌嗪-1-羧酸叔丁酯的合成
将2-(苄氧基)-1-溴-3-氟苯(200mg,0.71mmol),哌嗪-1-羧酸叔丁酯(265mg,1.42mmol),1,1'-联萘-2,2'-双二苯膦(88.6mg,0.14mmol),三(二亚苄基丙酮)二钯(66.2mg,0.07mmol),叔丁醇钾(159.6mg,1.42mmol),加入到甲苯(5mL)中,氮气置换三次,置于120℃下反应20分钟。TLC检测反应完全,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=10%)得到无色油状产物186mg,产率67.6%。
LC-MS(ESI):[M+H]+=387.3;
1H NMR(500MHz,CDCl3)δ7.47–7.42(m,2H),7.40–7.29(m,3H),6.98–6.92(m,1H),6.81–6.74(m,1H),6.66(d,J=8.4Hz,1H),5.09(s,2H),3.54–3.56(m,4H),3.07–2.98(m,4H),1.47(s,9H).
步骤3:4-(3-氟-2-羟基苯基)哌嗪-1-羧酸叔丁酯的合成
将4-(2-(苄氧基)-3-氟苯基)哌嗪-1-羧酸叔丁酯(1.5g,3.88mmol),钯碳(410mg),加入到25mL的甲醇溶液中,氢气置换三次,室温搅拌过夜。TLC检测原料反应完全,停止反应。硅藻土抽滤,滤液减压浓缩得到了棕色固体产物1.1g,产率95.6%。
LC-MS(ESI):[M+H]+=297.2;
1H NMR(500MHz,CDCl3)δ6.94–6.85(m,2H),6.82–6.76(m,1H),3.64–3.56(m,4H),2.92–2.79(m,4H),1.49(s,9H).
步骤4:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌嗪-1-羧酸叔丁酯的合成
将4-(3-氟-2-羟基苯基)哌嗪-1-羧酸叔丁酯(500mg,1.69mmol),7-(溴甲基)-4-氯-2-氟苯并呋喃(444mg,1.69mmol),碳酸钾(467mg,3.38mmol)加入到N,N-二甲基甲酰胺(10mL)中,60℃下反应2小时。TLC检测反应原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=5%)得到无色油状产物663mg,产率82%。
1H NMR(500MHz,CDCl3)δ7.30(d,J=8.1Hz,1H),7.28–7.19(m,1H),7.00–6.93(m,1H),6.81–6.74(m,1H),6.68–6.63(m,1H),6.00(d,J=6.6Hz,1H),5.32(s,2H),3.51–3.39(m,4H),3.06–2.92(m,4H),1.47(s,9H).
步骤5:1-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌嗪的合成
将4-(2-((4-氯-2氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌嗪-1-羧酸叔丁酯(593mg,1.24mmol),加入到5mL的三氟乙酸和5mL的二氯甲烷中,室温搅拌30分钟。TLC检测原料完全消失,停止反应。减压浓缩蒸干溶剂,用饱和的碳酸氢钠水溶液调节体系的PH至7左右,用乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩得到了棕色的固体产物450mg,产率:95.9%。
LC-MS(ESI):[M+H]+=379.2;
1H NMR(500MHz,CDCl3)δ7.25(s,2H),7.04–6.97(m,1H),6.89–6.80(m,1H),6.67(d,J=8.3Hz,1H),6.02(d,J=6.6Hz,1H),5.28(s,2H),3.31–3.22(m,4H),3.21–3.10(m,4H).
步骤6:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将1-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌嗪(150mg,0.40mmol),(S)-2-(氯甲基)-1-(氧杂环 丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(116mg,0.39mmol),碳酸钾(110mg,0.8mmol),碘化钾(65mg,0.39mmol)加入到8mL的乙腈溶液中,60℃反应过夜。TLC检测原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=25%)得到无色油状产物177mg,产率70.2%.
LC-MS(ESI):[M+H]+=637.3。
步骤7:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(177mg,0.28mmol),氢氧化锂一水合物(58mg,1.38mmol)加入到水(2mL)和1,4-二氧六环(6mL)的混合溶液中,40℃反应2小时。TLC检测原料完全消失,停止加热。用20%的醋酸水溶液调节体系PH至7左右,用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=20%)得到白色固体产物89mg,产率49.5%。
LC-MS(ESI):[M+H]+=623.4;
1H NMR(500MHz,CDCl3)δ8.26(d,J=1.6Hz,1H),7.81(dd,J=8.5,1.6Hz,1H),7.65(d,J=8.5Hz,1H),7.40–7.31(m,2H),7.01(td,J=8.2,6.1Hz,1H),6.87–6.76(m,1H),6.73(d,J=8.3Hz,1H),6.50(d,J=6.4Hz,1H),5.29(s,2H),5.11–5.04(m,1H),4.77(dd,J=15.3,7.2Hz,1H),4.66–4.59(m,1H),4.52–4.44(m,1H),4.38–4.33(m,1H),3.94(d,J=13.5Hz,1H),3.77(d,J=13.5Hz,1H),3.02–2.89(m,4H),2.73–2.65(m,1H),2.57–2.44(m,4H),2.43–2.35(m,1H).
实施例11 2-((S)-1-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌嗪-1-基)乙基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸(化合物11)的合成
步骤1:2-((S)-1-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌嗪-1-基)乙基)-1-(((S)-(氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将1-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌嗪(122mg,0.40mmol),2-((R)-1-氯乙基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(150mg,0.40mmol),碳酸钾(109mg,0.79mmol),碘化钾(65mg,0.39mmol)加入到10mL的乙腈溶液中,60℃反应过夜。TLC检测原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=25%)得到无色油状产物183mg,产率71%。
LC-MS(ESI):[M+H]+=651.4;
1H NMR(500MHz,CDCl3)δ8.13(s,1H),7.98(dd,J=8.6,1.6Hz,1H),7.81(d,J=8.5Hz,1H),7.26(d,J=10.4Hz,1H),7.19(d,J=8.2Hz,1H),6.97–6.89(m,1H),6.80–6.67(m,1H),6.61(d,J=8.0Hz,1H),5.99(d,J=6.7Hz,1H),5.28(s,2H),5.27–5.23(m,1H),5.00–4.94(m,1H),4.60–4.54(m,1H),4.54–4.46(m,1H),4.45–4.38(m,1H),4.25–4.18(m,1H),3.94(s,3H),3.08–2.97(m,4H),2.68–2.63(m,1H),2.62–2.54(m,4H),2.43–2.29(m,1H),1.59(d,J=6.8Hz,3H).
步骤2:2-((S)-1-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌嗪-1-基)乙基)-1-(((S)-氧杂环丁烷 -2-基)甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将2-((S)-1-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌嗪-1-基)乙基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(180mg,0.28mmol),氢氧化锂一水合物(58mg,1.38mmol)加入到2mL水和6mL 1,4-二氧六环的混合溶液中,40℃反应2小时。TLC检测原料完全消失,停止加热。用20%的醋酸水溶液调节体系PH至6左右,用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=20%)得到白色固体产物56mg,产率28.1%。
LC-MS(ESI):[M+H]+=637.3;
1H NMR(500MHz,DMSO-d6)δ12.70(s,1H),8.27(s,1H),7.80(dd,J=8.5,1.6Hz,1H),7.68(d,J=8.5Hz,1H),7.32–7.26(m,2H),7.05–6.95(m,1H),6.87–6.80(m,1H),6.69(d,J=8.4Hz,1H),6.46(d,J=6.4Hz,1H),5.25(s,2H),5.16–5.09(m,1H),4.79–4.72(m,1H),4.68–4.61(m,1H),4.45–4.36(m,2H),4.20–4.13(m,1H),2.94–2.81(m,4H),2.65–2.58(m,1H),2.47–2.40(m,4H),2.34–2.23(m,1H),1.43(d,J=6.8Hz,3H).
实施例12 2-((S)-1-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)乙基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-羧酸(化合物12)的合成
步骤1:2-((S)-1-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)乙基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-羧酸甲酯的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶(300mg,0.97mmol),2-((R)-1-氯乙基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(300mg,0.79mmol),碳酸钾(220mg,1.59mmol)和碘化钾(132mg,0.80mmol)加入到乙腈(20mL)中,60℃反应2h。冷却至室温,加水(30mL),用乙酸乙酯萃取(20mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=50%),得到黄色粘稠液体产物150mg,产率29.1%。
LC-MS(ESI):[M+H]+=650.4。
步骤2:2-((S)-1-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)乙基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-羧酸的合成
将化合物2-((S)-1-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)乙基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-羧酸甲酯(150mg,0.23mmol)和氢氧化锂一水合物(49mg,1.17mmol)加入到1,4-二氧六环(4mL)和水(2mL)中,40℃反应1.5h。冷却至室温,加水稀释(15mL),用醋酸(4M)调节体系pH至5,乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩滤液。硅胶柱层析纯化(洗脱剂MeOH:DCM(v/v)=5%)得到黄色固体产物56mg,产率37.2%。
LC-MS(ESI):[M+H]+=636.3;
1H NMR(500MHz,DMSO-d6)δ12.71(br,1H),8.28(s,1H),7.82(d,J=8.4Hz,1H),7.69(d,J=8.6Hz,1H),7.36(d,J=8.2Hz,1H),7.27(d,J=8.2Hz,1H),7.15–7.02(m,2H),6.95(d,J=7.8Hz,1H),6.45(d,J=6.4Hz,1H),5.24(s,2H),5.18–5.09(m,1H),4.83–4.74(m,1H),4.70–4.59(m,1H),4.50–4.37(m,2H),4.22–4.12(m,1H),2.94–2.84(m,1H),2.68–2.58(m,1H),2.58–2.54(m,1H),2.36–2.21(m,2H),2.01–1.86(m, 1H),1.56–1.40(m,1H),1.44(s,3H),1.36–1.11(m,4H).
实施例13(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-2-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物13)的合成
步骤1:3-羟基-3',6'-二氢-(2,4'-联吡啶)-1'(2'H)-羧酸叔丁酯的合成
将化合物4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(2.13g,6.90mmol)和2-溴-3-羟基吡啶(1.2g,6.90mmol)加入1,4-二氧六环(20mL)和水(5mL)中搅拌溶解,加入碳酸钠(1.83g,17.24mmol)和四(三苯基膦)钯(0.33g,0.29mmol),氮气保护下升温至120℃反应过夜。降至室温后减压浓缩,加入水(10mL),用乙酸乙酯(10mL×3)萃取,有机相用无水硫酸钠干燥,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:5)得到类白色固体产物600mg,产率37.7%。
1HNMR(400MHz,DMSO-d6)δ10.04(s,1H),8.03–8.01(m,1H),7.22–7.20(m,1H),7.10–7.08(m,1H),6.68(s,1H),4.02(s,2H),3.50(t,J=5.6Hz,2H),2.57(d,J=1.8Hz,2H),1.43(s,9H).
步骤2:4-(3-羟基吡啶-2-基)哌啶-1-羧酸叔丁酯的合成
将化合物3-羟基-3',6'-二氢-(2,4'-联吡啶)-1'(2'H)-羧酸叔丁酯(600mg,2.17mmol)加入甲醇(20mL)中,加入10%钯碳(100mg,0.56mmol),反应液在氢气氛围下室温搅拌过夜。用硅藻土过滤,滤液用甲醇(50mL)洗涤,滤液减压浓缩,得到淡黄色油状产物600mg,产率99.3%。
1HNMR(400MHz,DMSO-d6)δ9.82(s,1H),8.01–7.88(m,1H),7.14–6.97(m,2H),4.05(d,J=11.2Hz,2H),3.18(d,J=7.2Hz,1H),2.83(s,2H),1.72–1.51(m,4H),1.42(s,9H).
步骤3:4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-2-基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(3-羟基吡啶-2-基)哌啶-1-羧酸叔丁酯(600mg,2.16mmol)和7-(溴甲基)-4-氯-2-氟-1-苯并呋喃(568mg,2.16mmol)加入N,N-二甲基甲酰胺(10mL)中,加入碳酸钾(894mg,6.47mmol),反应液在氮气保护下升温至60℃反应3小时。将反应液降温至室温,加入水(50mL),用乙酸乙酯(30mL×3)萃取,有机相用无水硫酸钠干燥,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:3)得到淡黄色固体产物600mg,产率60.4%。
1HNMR(400MHz,CDCl3)δ8.12(m,1H),7.23–7.12(m,3H),7.04(m,1H),5.98(d,J=6.4Hz,1H),5.23(s,2H),4.16(s,2H),3.24(d,J=4.0Hz,1H),2.73(s,2H),1.76(d,J=39.2Hz,4H),1.39(s,9H).
步骤4:3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-2-(哌啶-4-基)吡啶盐酸盐的合成
将化合物4-(3-((4-氯-2-氟代苯并呋喃-7-基)甲氧基)吡啶-2-基)哌啶-1-羧酸叔丁酯加入二氯甲烷(10mL)中,加入盐酸二氧六环溶液(3mL),反应液保持室温反应2小时。减压浓缩得到类白色固体产物450mg,收率87.0%,直接用于下一步反应。
步骤5:(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-2-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-2-(哌啶-4-基)吡啶盐酸盐(300mg,0.83mmol)和(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(245mg,0.83mmol)加入N,N-二甲基甲酰胺(2mL)中,加入碳酸钾(575mg,4.16mmol)和碘化钾(5.0mg,0.03mmol),氮气保护下将反应液升 温至85℃,搅拌2小时。将反应液降至室温,缓慢滴入水(10mL)淬灭,用乙酸乙酯(10mL×3)萃取,有机相用无水硫酸钠干燥,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=1:20),得到淡黄色固体产物128mg,产率24.9%。
LC-MS(ESI):[M+H]+=619.3。
步骤6:(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-2-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-2-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(128mg,0.21mmol)加入二氧六环(2mL)和水(0.5mL)中,加入氢氧化锂(44.0mg,1.05mmol),在氮气保护下升温至40℃反应2小时。反应液降温至室温,用醋酸调节PH值至2~3。减压浓缩,用Prep-HPLC(洗脱剂CH3CN:H2O(v/v)=60%)纯化,得到白色固体产物17.0mg,产率13.6%。
LC-MS(ESI):[M+H]+=605.4;
1HNMR(400MHz,DMSO-d6)δ8.25(s,1H),8.11(d,J=4.4Hz,1H),7.81(d,J=8.0Hz,1H),7.63(d,J=8.4Hz,1H),7.53(d,J=8.4Hz,1H),7.49–7.39(m,2H),7.20(m,1H),6.54(d,J=6.4Hz,1H),5.41(s,2H),5.12–5.06(m,1H),4.85–4.79(m,1H),4.68–4.57(m,1H),4.52–4.46(m,1H),4.40–4.35(m,1H),3.90(d,J=13.6Hz,1H),3.78(d,J=13.6Hz,1H),2.96(d,J=10.8Hz,1H),2.97–2.82(m,1H),3.17–3.11(m,1H),2.78–2.68(m,1H),2.46–2.37(m,1H),2.22–2.02(m,2H),1.783–1.74(m,2H),1.68–1.58(m,2H).
实施例14(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物14)的合成
步骤1:2-(苄氧基)-3-溴吡啶的合成
将化合物3-溴-2-氟吡啶(1.00g,5.68mmol)和苯甲醇(0.68g,6.25mmol)加入到乙腈(5mL)中,然后加入碳酸铯(1.85g,5.68mmol)。室温反应过夜。硅藻土过滤,滤液减压浓缩,硅胶柱层析分离纯化(洗脱剂EA:PE(v/v)=10%),得无色液体产物1.13g,产率75.3%。
1H NMR(500MHz,CDCl3)δ8.09(dd,J=4.9,1.7Hz,1H),7.81(dd,J=7.6,1.7Hz,1H),7.49(d,J=7.5Hz,2H),7.37(t,J=7.6Hz,2H),7.30(d,J=7.4Hz,1H),6.77(dd,J=7.5,4.9Hz,1H),5.46(s,2H).
步骤2:2-(苄氧基)-3',6'-二氢-[3,4'-联吡啶]-1'(2'H)-羧酸叔丁酯的合成
将化合物2-(苄氧基)-3-溴吡啶(0.50g,1.89mmol)和N-Boc-1,2,5,6-四氢吡啶-4-硼酸频哪醇酯(0.59g,1.90mmol)加入到1,4-二氧六环(12mL)和水(3mL)的混合溶液中,然后加入[1,1'-双(二苯基膦)二茂铁]二氯化钯(78mg,0.10mmol)和碳酸钾(524mg,3.79mmol)。氮气保护下加热至90℃反应2h。冷却至室温,加水稀释(20ml),乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥。过滤,浓缩滤液,硅胶柱层析分离纯化(洗脱剂EA:PE(v/v)=30%),得到浅黄色液体0.59g,产率85.2%。
LC-MS(ESI):[M+H]+=367.3;
1H NMR(500MHz,CDCl3)δ8.07(dd,J=5.0,1.9Hz,1H),7.45(dd,J=7.3,1.9Hz,1H),7.42(d,J=6.7Hz,2H),7.36(t,J=7.6Hz,2H),7.32–7.28(m,1H),6.88(dd,J=7.3,5.0Hz,1H),5.91–5.88(m,1H),5.43(s, 2H),4.06–4.02(m,2H),3.59–3.54(m,2H),2.53–2.47(m,2H),1.48(s,9H).
步骤3:4-(2-羟基吡啶-3-基)哌啶-1-羧酸叔丁酯的合成
将化合物2-(苄氧基)-3',6'-二氢-[3,4'-联吡啶]-1'(2'H)-羧酸叔丁酯(0.59g,1.61mmol)溶解在乙醇(13mL)中,加入钯碳(10%Pd,含55%水)(0.19g,0.80mmol),用氢气置换三次,在氢气环境下室温反应过夜。硅藻土过滤,滤液减压浓缩,得白色固体0.36g,产率80.8%。
LC-MS(ESI):[M-55]+=223.0;
1H NMR(500MHz,DMSO-d6)δ11.46(s,1H),7.24–7.19(m,2H),6.12(t,J=6.7Hz,1H),4.08–4.01(m,2H),2.83–2.72(m,3H),1.71(d,J=12.9Hz,2H),1.40(s,9H),1.34(dd,J=12.5,4.0Hz,2H).
步骤4:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(2-羟基吡啶-3-基)哌啶-1-羧酸叔丁酯(360mg,1.29mmol)和(4-氯-2-氟苯并呋喃-7-基)甲醇(220mg,1.10mmol)加入到四氢呋喃(10mL)中,然后加入三苯基膦(400mg,1.53mmol)和偶氮二甲酸二异丙酯(332mg,1.64mmol)。室温反应3h。加入水(30mL)淬灭反应,用乙酸乙酯萃取(15mL×3),合并有机相。无水硫酸钠干燥,减压浓缩,硅胶柱层析分离纯化(洗脱剂EA:PE(v/v)=25%),得无色粘稠液体69mg,产率13.6%。
LC-MS(ESI):[M+H]+=461.3;
1H NMR(500MHz,CDCl3)δ8.03(dd,J=5.0,1.8Hz,1H),7.42(dd,J=7.3,1.9Hz,1H),7.29–7.22(m,2H),6.89(dd,J=7.4,5.0Hz,1H),6.01(d,J=6.6Hz,1H),5.62(s,2H),4.29–4.15(m,2H),3.02–2.95(m,1H),2.82–2.73(m,2H),1.83(d,J=12.8Hz,2H),1.55–1.50(m,2H),1.47(s,9H).
步骤5:2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-(哌啶-4-基)吡啶的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-羧酸叔丁酯(100mg,0.22mmol)加入到二氯甲烷(1.5mL)中,然后在冰浴下加入三氟乙酸(544mg,4.77mmol)。恢复至室温反应30min。加饱和碳酸氢钠溶液调节PH=8,用乙酸乙酯萃取(10mL×3)。合并有机相,用无水硫酸钠干燥,过滤,减压除去溶剂,得到黄色粘稠固体82mg,产率104.8%。
LC-MS(ESI):[M+H]+=361.2。
步骤6:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-(哌啶-4-基)吡啶(82mg,0.23mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(67mg,0.23mmol),N,N-二异丙基乙胺(150mg,1.16mmol)加入到乙腈(5mL)中,60℃反应过夜。冷却至室温,加水(20mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=50%),得到黄色固体产物92mg,产率65.4%。
LC-MS(ESI):[M+H]+=619.4。
步骤7:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(92mg,0.15mmol)和氢氧化锂一水合物(32mg,0.76mmol)加入到1,4-二氧六环(4mL)和水(2mL)中,40℃反应1.5h。冷却至室温,加水稀释(15mL),用醋酸(4M)调节体系pH至5,乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩滤液。硅胶柱层析纯化(洗脱剂MeOH:DCM(v/v)=3%)得到黄色固体产物20mg,产率22.2%。
LC-MS(ESI):[M+H]+=605.4;
1H NMR(500MHz,DMSO-d6)δ12.65(br,1H),8.26(s,1H),8.01(d,J=4.9Hz,1H),7.80(d,J=8.4Hz,1H),7.62(dd,J=18.4,7.9Hz,2H),7.44–7.38(m,2H),6.98(t,J=6.2Hz,1H),6.51(d,J=6.3Hz,1H),5.60(s,2H),5.11–5.04(m,1H),4.77(dd,J=15.3,7.2Hz,1H),4.63(d,J=15.1Hz,1H),4.51–4.45(m,1H),4.38–4.33(m,1H),3.92(d,J=13.5Hz,1H),3.78(d,J=13.6Hz,1H),2.98(d,J=11.1Hz,1H),2.86(d,J=11.4Hz,1H),2.83–2.75(m,1H),2.74–2.65(m,1H),2.45–2.36(m,1H),2.25–2.11(m,2H),1.75(t,J=14.0Hz,2H),1.69–1.56(m,2H).
实施例15(S)-2-((2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物15)的合成
步骤1:1-溴-2-(甲氧基甲氧基)苯的合成
将化合物2-溴苯酚(5g,28.9mmol)加入N,N-二甲基甲酰胺(50mL)中,加入NaH(1.4g,57.8mmol),冰浴搅拌30分钟。加入溴(甲氧基)甲烷(4.3g,34.7mmol),室温搅拌两小时。加入水(50mL),用二氯甲烷(50mL×3)萃取,合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥后,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:20)得到无色油状产物6.15g,产率98.0%。
步骤2:2-(3-氟-2'-(甲氧基甲氧基)-[1,1'-联苯基]-4-基)乙酸甲酯的合成
将化合物1-溴-2-(甲氧基甲氧基)苯(2.0g,9.2mmol)、2-(2-氟-4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)苯基)乙酸甲酯(3.0g,10.12mmol)加入到1,4-二氧六环(20mL)和水(5mL)中。加入碳酸钾(3.8g,27.6mmol),用氮气置换三次,加入四三苯基膦钯(0.3g,0.26mmol),再次氮气置换三次,85℃搅拌过夜。减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:12)得无色油状产物2.4g,产率85.7%。
1HNMR(400MHz,CDCl3)δ7.27–7.19(m,5H),7.15(dd,J=8.7,1.0Hz,1H),7.04–6.99(m,1H),5.07(s,2H),3.66(d,J=9.4Hz,5H),3.34(s,3H).
步骤3:2-(3-氟-2'-(甲氧基甲氧基)-[1,1'-联苯]-4-基)乙-1-醇的合成
将化合物2-(3-氟-2'-(甲氧基甲氧基)-[1,1'-联苯基]-4-基)乙酸甲酯(2.4g,7.9mmol)加入四氢呋喃(20mL)中,0℃搅拌五分钟。加入四氢铝锂(0.87g,23.0mmol),0℃搅拌两小时。加入水(10mL)淬灭,用乙酸乙酯(30mL×3)萃取,合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥后,减压浓缩,用硅胶柱层析进行纯化(洗脱剂EtOAc:PE(v/v)=1:5)得无色油状产物1.2g,产率54.5%。
1HNMR(400MHz,CDCl3)δ7.27–7.15(m,6H),7.01(d,J=1.0Hz,1H),5.07(s,2H),3.84(t,J=6.6Hz,2H),3.35(s,3H),2.89(t,J=6.6Hz,2H).
步骤4:2-(3-氟-2'-(甲氧基甲氧基)-[1,1'-联苯基]-4-基)乙醛的合成
将化合物2-(3-氟-2'-(甲氧基甲氧基)-[1,1'-联苯]-4-基)乙-1-醇(1.2g,4.3mmol)加入到二氯甲烷(20mL)中,加入戴斯马丁试剂(2.8g,6.5mmol)冰浴搅拌1小时,析出大量固体。加入饱和碳酸氢钠(5mL),减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:8)得到无色油状产物550mg,产率46.6%。
1H NMR(400MHz,DMSO-d6)δ9.74(d,J=1.2Hz,1H),7.39–7.30(m,5H),7.22(d,J=7.8Hz,1H),7.12–7.07(m,1H),5.19(s,2H),3.89(s,2H),3.30(d,J=5.5Hz,3H).
步骤5:(S)-2-((3-氟-2'-(甲氧基甲氧基)-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d] 咪唑-6-羧酸甲酯的合成
将化合物2-(3-氟-2'-(甲氧基甲氧基)-[1,1'-联苯基]-4-基)乙醛(550mg,2.0mmol)、(S)-4-氨基-3-((氧杂环丁烷-2-基甲基)氨基)苯甲酸甲酯(520mg,2.2mmol)加入醋酸(20mL)中,通入氧气,60℃搅拌5小时。减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=20:1)得到浅褐色固体产物165mg,产率16.8%。
LC-MS(ESI):[M+H]+=491.3。
步骤6:(S)-2-((3-氟-2'-羟基-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-((3-氟-2'-(甲氧基甲氧基)-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(165mg,0.34mmol)加入二氯甲烷(10mL)中,冷却至-20℃,加入三氟乙酸(5mL),-20℃搅拌2小时。减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=20:1)得到浅褐色固体产物35mg,产率23.3%。
LC-MS(ESI):[M+H]+=447.4。
步骤7:(S)-2-(4-(6-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-2-基)苄基)-1-(氧杂环丁烷-2-基甲基)-1H-噻吩并[2,3-d]咪唑-5-羧酸甲酯的合成
将化合物(S)-2-((3-氟-2'-羟基-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(35mg,0.08mmol)、7-(溴甲基)-4-氯-2-氟-1-苯并呋喃(32.0mg,0.12mmol)加入N,N-二甲基甲酰胺(2mL)中,加入碳酸钾(33.0mg,0.24mmol),60℃搅拌一小时。加入水(5mL),用乙酸乙酯(5mL×3)萃取,合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=10:1)得到白色固体产物28mg,产率56.8%。
LC-MS(ESI):[M+H]+=629.2。
步骤8:(S)-2-((2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-(4-(6-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-2-基)苄基)-1-(氧杂环丁烷-2-基甲基)-1H-噻吩并[2,3-d]咪唑-5-羧酸甲酯(28mg,44.6umol)溶解在二氧六环(2mL)、水(0.5mL)中。加入氢氧化锂(6.5mg,0.27mmol),室温搅拌8小时。用醋酸调节PH至6~7。减压浓缩,用Prep-HPLC(洗脱剂CH3CN:H2O(v/v)=50%)纯化得白色固体产物6.1mg,产率22.3%。
LC-MS(ESI):[M+H]+=615.3;
1H NMR(400MHz,DMSO-d6)δ8.22(s,1H),7.78(d,J=8.6Hz,1H),7.57(d,J=8.4Hz,1H),7.42–7.33(m,5H),7.33–7.24(m,3H),7.09(t,J=7.5Hz,1H),6.50(d,J=6.4Hz,1H),5.38(s,2H),5.00(d,J=5.2Hz,1H),4.68(dd,J=15.5,7.2Hz,1H),4.55(d,J=13.2Hz,1H),4.49–4.40(m,2H),4.39–4.27(m,2H),2.68–2.60(m,1H),2.33(s,1H).
实施例16(S)-2-((2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3,3'-二氟-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物16)的合成
步骤1:1-溴-3-氟-2-(甲氧基甲氧基)苯的合成
将化合物2-溴-6-氟苯酚(5.0g,26.18mmol)加入N,N-二甲基甲酰胺(50mL)中,加入氢化钠(1.57g,39.27mmol),冰浴搅拌十分钟。加入溴(甲氧基)甲烷(3.93g,31.41mmol),室温搅拌一小时。加入水(250mL),用乙酸乙酯(50mL×3)萃取,合并有机相,用饱和食盐水洗一次,无水硫酸钠干燥后,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:20),得到无色油状产物6.0g,产率97.5%。
1HNMR(400MHz,CDCl3)δ7.28–7.27(m,1H),7.02–6.96(m,1H),6.88–6.84(m,1H),5.13(s,2H),3.57(s,3H).
步骤2:2-(3,3'-二氟-2'-(甲氧基甲氧基)-[1,1'-联苯基]-4-基)乙酸甲酯的合成
将化合物1-溴-3-氟-2-(甲氧基甲氧基)苯(1.9g,8.08mmol)、2-(2-氟-4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)苯基)乙酸甲酯(2.85g,9.7mmol)加入到1,4-二氧六环(30mL)和水(7.5mL)中。加入碳酸钾(3.35g,24.24mmol),用氮气置换三次,加入四(三苯基膦)钯(0.47g,0.4mmol),再次氮气置换三次,85℃搅拌过夜。减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:15),得到无色油状产物2.2g,产率84.5%。
1HNMR(400MHz,CDCl3)δ7.23(d,J=6.0Hz,3H),7.07–7.01(m,3H),4.83(s,2H),3.67–3.64(m,5H),3.10(s,3H)
步骤3:2-(3,3'-二氟-2'-(甲氧基甲氧基)-[1,1'-联苯]-4-基)乙-1-醇的合成
将化合物2-(3,3'-二氟-2'-(甲氧基甲氧基)-[1,1'-联苯基]-4-基)乙酸甲酯(2.2g,6.83mmol)加入四氢呋喃(50mL)中,0℃搅拌五分钟。加入四氢铝锂(0.78g,20.5mmol),0℃搅拌两小时。加入水(20mL)淬灭,用乙酸乙酯(30mL×3)萃取,合并有机相,有机相用饱和食盐水洗一次,无水硫酸钠干燥后,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:5),得到无色油状产物1.2g,产率59.7%。
1H NMR(400MHz,DMSO-d6)δ7.40(t,J=8.0Hz,1H),7.33–7.26(m,3H),7.25–7.20(m,2H),4.86(s,2H),4.74(t,J=5.2Hz,1H),3.65–3.61(m,2H),3.09(s,3H),2.80(t,J=7.0Hz,2H).
步骤4:2-(3,3'-二氟-2'-(甲氧基甲氧基)-[1,1'-联苯基]-4-基)乙醛的合成
将化合物2-(3,3'-二氟-2'-(甲氧基甲氧基)-[1,1'-联苯]-4-基)乙-1-醇(1.2g,4.08mmol)加入到二氯甲烷(20mL)中,加入戴斯马丁试剂(2.59g,6.12mmol)冰浴搅拌1小时,析出大量固体。加入水(5mL),过滤后减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:5),得无色油状产物700mg,产率58.7%。
1HNMR(400MHz,DMSO-d6)δ9.73(d,J=1.2Hz,1H),7.40–7.30(m,4H),7.24(dd,J=6.2,3.2Hz,2H),4.88(s,2H),3.92(s,2H),3.08(s,3H).
步骤5:(S)-2-((3,3'-二氟-2’-(甲氧基甲氧基)-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物2-(3,3'-二氟-2'-(甲氧基甲氧基)-[1,1'-联苯基]-4-基)乙醛(700mg,2.39mmol)、(S)-4-氨基-3-((氧杂环丁烷-2-基甲基)氨基)苯甲酸甲酯(566mg,2.4mmol)加入醋酸(10mL)中,60℃搅拌2小时。减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=1:1),得到浅褐色固体产物155mg,产率12.7%。
LC-MS(ESI):[M+H]+=509.4。
步骤6:(S)-2-((3,3'-二氟-2'-羟基-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-((3,3'-二氟-2'-(甲氧基甲氧基)-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(155mg,0.3mmol)加入二氯甲烷(12mL)中,冷却至-20℃,加入三氟乙酸(4mL),-20℃搅拌2小时。减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=20:1),得到浅褐色固体产物100mg,产率70.6%。
LC-MS(ESI):[M+H]+=465.4。
步骤7:(S)-2-((2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3,3'-二氟-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-((3,3'-二氟-2'-羟基-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(100mg,0.22mmol)、7-(溴甲基)-4-氯-2-氟-1-苯并呋喃(80mg,0.3mmol)加入N,N-二甲基甲酰胺(5mL)中,加入碳酸钾(90mg,0.65mmol),60℃搅拌一小时。加入水(10mL),用乙酸乙酯(10mL×3)萃取,合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=10:1),得到类白色固体产物100mg,产率71.8%。
LC-MS(ESI):[M+H]+=647.4。
步骤8:(S)-2-((2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3,3'-二氟-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((2'-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3,3'-二氟-[1,1'-联苯基]-4-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(100mg,0.15mmol)加入到二氧六环(4mL)、水(1mL)中。加入氢氧化锂(22mg,0.92mmol),室温搅拌4小时。用盐酸(0.5N)调节PH至6~7,用乙酸乙酯(5mL×3)萃取,合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥。减压浓缩,用Prep-HPLC(洗脱剂CH3CN:H2O(v/v)=50%)纯化,得到白色固体产物54.0mg,产率55.2%。
LC-MS(ESI):[M+H]+=633.2;
1HNMR(400MHz,DMSO-d6)δ12.74(s,1H),8.25(s,1H),7.79(dd,J=8.4,1.3Hz,1H),7.59(d,J=8.4Hz,1H),7.41–7.29(m,1H),7.28–7.12(m,4H),7.06(dd,J=12.5,9.5Hz,2H),6.96(d,J=8.2Hz,1H),6.38(d,J=6.4Hz,1H),5.12(s,2H),5.04(d,J=6.8Hz,1H),4.71(dd,J=15.6,7.0Hz,1H),4.58(d,J=13.0Hz,1H),4.53–4.47(m,1H),4.43(s,1H),4.40–4.32(m,2H),2.74–2.64(m,1H),2.36(dd,J=18.6,7.9Hz,1H).
实施例17 2-((R)-1-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)乙基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸(化合物17)的合成
步骤1:2-((R)-1-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)乙基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-羧酸甲酯的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶(350mg,0.93mmol),2-((S)-1-氯乙基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(286mg,0.93mmol),碳酸钾(256mg,1.85mmol)和碘化钾(153mg,0.92mmol)加入到乙腈(25mL)中,60℃反应2h。冷却至室温,加水(30mL),用乙酸乙酯萃取(20mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=50%),得到黄色粘稠液体产物123mg,产率20.4%。
LC-MS(ESI):[M+H]+=650.4。
步骤2:2-((R)-1-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)乙基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物2-((R)-1-(4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)乙基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-羧酸甲酯(123mg,0.19mmol)和氢氧化锂一水合物(40mg,0.95mmol)加 入到1,4-二氧六环(4mL)和水(2mL)中,40℃反应1.5h。冷却至室温,加水稀释(15mL),用醋酸(4M)调节体系pH至5,乙酸乙酯萃取三次(15mL×3),合并有机相,无水硫酸钠干燥,过滤,浓缩滤液。硅胶柱层析纯化(洗脱剂MeOH:DCM(v/v)=5%)得到淡黄色固体产物60mg,产率49.2%。
LC-MS(ESI):[M+H]+=636.4;
1H NMR(500MHz,DMSO-d6)δ12.70(br,1H),8.28(s,1H),7.81(d,J=8.4Hz,1H),7.69(d,J=8.5Hz,1H),7.36(d,J=8.1Hz,1H),7.27(d,J=8.2Hz,1H),7.15–7.03(m,2H),6.95(d,J=7.8Hz,1H),6.45(d,J=6.3Hz,1H),5.28–5.21(m,2H),5.16–5.10(m,1H),4.77(d,J=16.6Hz,1H),4.64(dd,J=15.6,5.4Hz,1H),4.48–4.39(m,2H),4.20–4.14(m,1H),2.88(d,J=10.9Hz,1H),2.63(t,J=8.9Hz,1H),2.58–2.52(m,1H),2.37–2.23(m,2H),1.98–1.88(m,1H),1.55–1.47(m,1H),1.43(d,J=6.6Hz,3H),1.36–1.25(m,3H),1.20–1.11(m,1H).
实施例18(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲氧基苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物18)的合成
步骤1:4-(2-羟基-3-甲氧基苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将2-溴-6-甲氧基苯酚(800mg,3.94mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂硼环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(1.22g,3.94mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(320mg,0.39mmol),碳酸钾(1.09g,7.88mmol),加入到3mL的水和12mL的1,4-二氧六环的混合溶液中,氮气置换三次,置于90℃下反应2小时。TLC检测反应完全,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=15%)得到无色油状产物707mg,产率58.8%。
LC-MS(ESI):[M-55]+=250.1。
步骤2:4-(2-羟基-3-甲氧基苯基)哌啶-1-羧酸叔丁酯的合成
将4-(2-羟基-3-甲氧基苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(756mg,2.48mmol),钯碳(400mg),加入到10mL的甲醇溶液中,氢气置换三次,室温搅拌过夜。TLC检测原料反应完全,停止反应。硅藻土抽滤,滤液减压浓缩得到了棕色固体产物631mg,产率82.9%。
LC-MS(ESI):[M-55]+=252.1;
1H NMR(500MHz,CDCl3)δ6.92–6.69(m,3H),5.75(s,1H),4.33–4.14(s,2H),3.89(s,3H),3.12–3.03(m,1H),2.91–2.74(m,2H),1.82(d,J=12.9Hz,2H),1.66–1.59(m,1H),1.48(s,9H).
步骤3:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲氧基苯基)哌啶-1-羧酸叔丁酯的合成
将4-(2-羟基-3-甲氧基苯基)哌啶-1-羧酸叔丁酯(320mg,1.04mmol),7-(溴甲基)-4-氯-2-氟苯并呋喃(250mg,0.95mmol),碳酸钾(131mg,0.95mmol)加入到12mL的N,N-二甲基甲酰胺中,60℃下反应2小时。TLC检测反应原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=5%)得到无色油状产物350mg,产率75.3%
LC-MS(ESI):[M-55]+=434.2。
步骤4:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲氧基苯基)哌啶的合成
将4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲氧基苯基)哌啶-1-羧酸叔丁酯(350mg,0.71mmol),加入到三氟乙酸(4mL)和的二氯甲烷(4mL)溶液中,室温搅拌30分钟。TLC检测原料完全消失,停止反应。减压浓缩蒸干溶剂,用饱和的碳酸氢钠水溶液调节体系的PH至7左右,用乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩得到了褐色油状产物直接用于下一步反应。
步骤5:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲氧基苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲氧基苯基)哌啶(264mg,0.68mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(199mg,0.68mmol),DIPEA(437mg,3.39mmol)加入到5mL的乙腈溶液中,60℃反应过夜。TLC检测原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=40%)得到无色油状产物156mg,产率35.5%。
LC-MS(ESI):[M+H]+=648.4。
步骤6:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲氧基苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲氧基苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(90mg,0.14mmol),氢氧化锂一水合物(28.47mg,0.69mmol)加入到2mL水和6mL 1,4-二氧六环的混合溶液中,40℃反应2小时。TLC检测原料完全消失,停止加热。用20%的醋酸水溶液调节体系PH至6左右,用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=20%)得到白色固体产物28mg,产率31.2%。
LC-MS(ESI):[M+H]+=634.3;
1H NMR(500MHz,DMSO-d6)δ12.70(s,1H),8.25(s,1H),7.80(d,J=8.4Hz,1H),7.64(d,J=8.4Hz,1H),7.38(d,J=7.8Hz,1H),7.29(d,J=8.2Hz,1H),7.02(t,J=7.9Hz,1H),6.92(d,J=8.2Hz,1H),6.73(d,J=7.9Hz,1H),6.52(d,J=6.4Hz,1H),5.19(s,2H),5.11–5.02(m,1H),4.86–4.71(m,1H),4.69–4.56(m,1H),4.55–4.43(m,1H),4.43–4.30(m,1H),3.93–3.80(m,4H),3.71(d,J=13.5Hz,1H),2.89(d,J=11.2Hz,1H),2.81–2.62(m,3H),2.45–2.35(m,1H),2.04–1.83(m,2H),1.55–1.38(m,2H),1.35–1.28(m,2H).
实施例19(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲基苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物19)的合成
步骤1:4-(2-羟基-3-甲基苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将2-溴-6-甲基苯酚(500mg,2.67mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂硼环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(909mg,2.94mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(218mg,0.27mmol),碳酸钾(738mg,5.35mmol),加入到水(3mL)和1,4-二氧六环(12mL)的混合溶液中,氮气置换三次,置于90℃下反应2小时。TLC检测反应完全,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂 PE:EA(v/v)=15%)得到无色油状产物815mg,产率95.9%。
LC-MS(ESI):[M-55]+=234.2;
1H NMR(500MHz,CDCl3)δ7.03(d,J=7.4Hz,1H),6.92(d,J=7.8Hz,1H),6.80(t,J=7.5Hz,1H),5.84(s,1H),4.10–4.00(m,2H),3.67–3.59(m,2H),2.49–2.35(m,2H),2.26(s,3H),1.50(s,9H).
步骤2:4-(2-羟基-3-甲基苯基)哌啶-1-羧酸叔丁酯的合成
将4-(2-羟基-3-甲基苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(0.8g,2.67mmol),钯碳(400mg),加入到12mL的甲醇溶液中,氢气置换三次,室温搅拌过夜。TLC检测原料反应完全,停止反应。硅藻土抽滤,滤液减压浓缩得到了棕色固体产物0.816g,产率101.3%。
LC-MS(ESI):[M-55]+=236.2。
1H NMR(500MHz,CDCl3)δ7.06–6.95(m,2H),6.83(t,J=7.6Hz,1H),4.84(s,1H),4.49–4.17(m,2H),3.13–2.91(m,1H),2.94–2.67(m,2H),2.26(s,3H),1.82(d,J=13.0Hz,2H),1.66–1.55(m,2H),1.48(s,9H).
步骤3:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲基苯基)哌啶-1-羧酸叔丁酯的合成
将4-(2-羟基-3-甲基苯基)哌啶-1-羧酸叔丁酯(232mg,0.8mmol),7-(溴甲基)-4-氯-2氟苯并呋喃(21mg,0.8mmol),碳酸钾(220mg,1.59mmol)加入到12mL的N,N-二甲基甲酰胺中,60℃下反应2小时。TLC检测反应原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=5%)得到无色油状产物144mg,产率38.1%。
LC-MS(ESI):[M-55]+=418.3。
1H NMR(500MHz,CDCl3)δ7.28–7.33(m,2H),7.10–7.06(m,1H),7.05–7.02(m,2H),6.05(d,J=6.6Hz,1H),5.00(s,2H),4.35–4.15(m,2H),3.16–3.07(m,1H),2.83–2.54(m,2H),2.38(s,3H),1.71–1.53(m,4H),1.48(s,9H).
步骤4:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲基苯基)哌啶的合成
将4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲基苯基)哌啶-1-羧酸叔丁酯(290mg,0.61mmol),加入到1.5mL的三氟乙酸和3mL的二氯甲烷中,室温搅拌30分钟。TLC检测原料完全消失,停止反应。减压浓缩蒸干溶剂,用饱和的碳酸氢钠水溶液调节体系的PH至7左右,用乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩得到了褐色油状产物直接用于下一步反应。
1H NMR(500MHz,CDCl3)δ7.33–7.27(m,2H),7.18–7.05(m,3H),6.07(d,J=6.6Hz,1H),4.98(s,2H),3.56–3.40(m,2H),3.28–3.17(m,1H),3.02–2.80(m,2H),2.39(s,3H),2.08–1.93(m,2H),1.89–1.79(m,2H).
步骤5:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲基苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲基苯基)哌啶(150mg,0.4mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(118mg,0.4mmol),N,N-二异丙基乙胺(259mg,2.01mmol)加入到的乙腈(3mL)中,60℃反应过夜。TLC检测原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=35%)得到无色油状产物158mg,产率62.3%。
LC-MS(ESI):[M+H]+=632.4。
步骤6:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲基苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-甲基苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲 基)-1H-苯并[d]咪唑-6-羧酸甲酯(95mg,0.15mmol),氢氧化锂一水合物(31.5mg,0.75mmol)加入到水(2mL)和1,4-二氧六环(6mL)的混合溶液中,40℃反应2小时。TLC检测原料完全消失,停止加热。用20%的醋酸水溶液调节体系PH至6左右,用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=25%)得到白色固体产物34mg,产率36%。
LC-MS(ESI):[M+H]+=618.3。
1H NMR(500MHz,DMSO-d6)δ12.64(br.s,1H),8.27(s,1H),7.80(d,J=8.4Hz,1H),7.64(d,J=8.4Hz,1H),7.47–7.42(m,2H),7.18–6.97(m,3H),6.56(d,J=6.4Hz,1H),5.16–5.05(m,1H),5.00(s,2H),4.85–4.73(m,1H),4.71–4.59(m,1H),4.54–4.46(m,1H),4.41–4.33(m,1H),3.91(d,J=13.5Hz,1H),3.75(d,J=13.5Hz,1H),3.01–2.66(m,4H),2.46–2.38(m,1H),2.33(s,3H),2.12–1.94(m,2H),1.64–1.39(m,4H).
实施例20(S)-2-((4-(4-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物20)的合成
步骤1:4-(苄氧基)-3-溴吡啶的合成
将化合物3-溴-4-羟基吡啶(600mg,3.45mmol),苄溴(589mg,3.45mmol)和碳酸钾(714mg,5.17mmol)加入到N,N-二甲基甲酰胺(20mL)中,60℃下反应2小时。TLC检测反应原料完全消失,停止加热,冷却至室温,加入饱和食盐水(100mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=5%)得到无色油状产物632mg,产率69.4%。
LC-MS(ESI):[M+H]+=264.0;
1H NMR(500MHz,CDCl3)δ7.78(d,J=1.9,1H),7.50–7.39(m,3H),7.35(dd,J=7.5,2.4Hz,1H),7.24–7.18(m,2H),6.49(d,J=7.5Hz,1H),4.97(s,2H).
步骤2:4-(苄氧基)-3',6'-二氢-[3,4'-联吡啶]-1'(2'H)-羧酸叔丁酯的合成
将4-(苄氧基)-3-溴吡啶(500mg,1.89mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂硼环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(644mg,2.08mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(155mg,0.19mmol),碳酸钾(523mg,3.78mmol),加入到水(1mL)和1,4-二氧六环(4mL)的混合溶液中,氮气置换三次,置于90℃下反应2小时。TLC检测反应完全,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=15%)得到棕色油状产物0.605g,产率87.2%。
LC-MS(ESI):[M+H]+=367.3。
步骤3:4-(4-羟基吡啶-3-基)哌啶-1-羧酸叔丁酯的合成
将4-(苄氧基)-3',6'-二氢-[3,4'-联吡啶]-1'(2'H)-羧酸叔丁酯(707mg,1.93mmol),钯碳(350mg),加入到甲醇(12mL)溶液中,氢气置换三次,室温搅拌过夜。TLC检测原料反应完全,停止反应。硅藻土抽滤,滤液减压浓缩得到了棕色固体产物0.499g,产率92.9%
LC-MS(ESI):[M-55]+=223.1。
步骤4:4-(4-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-羧酸叔丁酯的合成
将4-(4-羟基吡啶-3-基)哌啶-1-羧酸叔丁酯(290mg,1.04mmol),7-(溴甲基)-4-氯-2-氟苯并呋喃(250mg,0.95mmol),碳酸钾(262mg,1.9mmol)加入到10mL的N,N-二甲基甲酰胺中,60℃下反应2小时。TLC检测反应原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=5%)得到无色油状产物260mg,产率82.3%。
LC-MS(ESI):[M+H]+=461.3。
1H NMR(500MHz,CDCl3)δ7.38–7.33(m,1H),7.32–7.28(m,1H),7.23(s,1H),7.00(d,J=8.1Hz,1H),6.39(d,J=7.4Hz,1H),6.06(d,J=6.6Hz,1H),5.10(s,2H),4.27–4.10(m,2H),3.15–3.00(m,1H),2.93–2.72(m,2H),1.90–1.85(m,4H),1.46(s,9H).
步骤5:4-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-(哌啶-4-基)吡啶的合成
将4-(4-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-羧酸叔丁酯(232mg,0.5mmol),加入到三氟乙酸(2mL)和二氯甲烷(4mL)中,室温搅拌30分钟。TLC检测原料完全消失,停止反应。减压浓缩蒸干溶剂,用饱和的碳酸氢钠水溶液调节体系的PH至7左右,用乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩得到了棕色的固体产物直接用于下一步反应。
LC-MS(ESI):[M+H]+=361.2。
步骤6:(S)-2-((4-(4-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将4-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-(哌啶-4-基)吡啶(150mg,0.42mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(122mg,0.42mmol),N,N-二异丙基乙胺(268mg,2.08mmol)加入到乙腈(5mL)中,60℃反应过夜。TLC检测原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(5mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=20%)得到无色油状产物116mg,产率45.1%
LC-MS(ESI):[M+H]+=619.4。
步骤7:(S)-2-((4-(4-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将(S)-2-((4-(4-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(116mg,0.19mmol),氢氧化锂一水合物(39.3mg,0.94mmol)加入到水(2mL)和1,4-二氧六环(6mL)的混合溶液中,40℃反应2小时。TLC检测原料完全消失,停止加热。用稀释过的醋酸水溶液调节体系PH至6左右,用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=2 5%)得到白色固体产物53mg,产率44.4%.
LC-MS(ESI):[M+H]+=605.3;
1H NMR(500MHz,CDCl3)δ12.64(s,1H),8.25(s,1H),7.80(d,J=8.4Hz,1H),7.67(s,2H),7.63(d,J=8.5Hz,1H),7.42(d,J=8.2Hz,1H),7.19(d,J=8.2Hz,1H),6.53(d,J=6.4Hz,1H),6.07(d,J=7.5Hz,1H),5.34(s,2H),5.11–5.02(m,1H),4.78(dd,J=15.2,7.1Hz,1H),4.64(d,J=15.0Hz,1H),4.52–4.41(m,1H),4.40–4.33(m,1H),3.92(d,J=13.5Hz,1H),3.77(d,J=13.5Hz,1H),2.99–2.91(m,1H),2.85–2.78(m,1H),2.76–2.57(m,2H),2.44–2.36(m,1H),2.21–2.07(m,2H),1.75–1.67(m,2H),1.50–1.36(m,2H).
实施例21(S)-2-((4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物21)的合成
步骤1:4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(3-氟-2-羟基苯基)哌啶-1-羧酸叔丁酯(160mg,0.54mmol)、7-(溴甲基)-2,4-二氯苯并呋喃(227mg,0.81mmol)加入N,N-二甲基甲酰胺(5mL)中。加入碳酸钾(224mg,1.62mmol),60℃搅拌1小时。减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=1:10),得到浅褐色固体产物157mg,产率73.7%。
1H NMR(400MHz,CDCl3)δ7.21–7.15(m,2H),6.96–6.90(m,2H),6.84–6.80(m,1H),6.67(s,1H),5.25(s,2H),4.11–4.05(m,2H),2.97–2.87(m,1H),2.61–2.51(m,2H),1.55–1.36(m,2H),1.41(s,9H),1.21–1.16(m,2H).
步骤2:4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶的合成
将化合物4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-羧酸叔丁酯(157mg,0.40mmol)加入乙酸乙酯(5mL)中,加入盐酸1.4-二氧六环(5mL),室温搅拌2小时。减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=10:1),得到浅褐色固体产物125mg,产率79.2%。
LC-MS(ESI):[M+H]+=394.3。
步骤3:(S)-2-((4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(68mg,0.23mmol)、碳酸钾(79mg,0.57mmol)、碘化钾(2mg,0.02mmol)加到乙腈(2mL)中。将化合物4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶(125mg,0.19mmol)加入乙腈(2mL)中,滴加二异丙基乙基胺(0.5mL),并加入到反应体系,60℃搅拌2小时。加入水(5mL),用乙酸乙酯(10mL×3)萃取,合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=3:2),得到类白色固体产物80mg,产率64.5%。
LC-MS(ESI):[M+H]+=652.5。
步骤4:(S)-2-((4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(80mg,0.12mmol)加入到二氧六环(2mL)、水(0.5mL)中。加入氢氧化锂(18mg,0.72mmol),40℃搅拌4小时。用盐酸(0.5N)调节PH为6~7,用乙酸乙酯(5mL×3)萃取,合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥。减压浓缩,用Prep-HPLC(洗脱剂CH3CN:H2O(v/v)=50%),纯化得到白色固体产物34.7mg,产率45.3%。
LC-MS(ESI):[M+H]+=638.3;
1H NMR(400MHz,DMSO-d6)δ12.69(s,1H),8.25(s,1H),7.80(d,J=9.8Hz,1H),7.63(d,J=8.4Hz,1H),7.40(d,J=8.0Hz,1H),7.33(d,J=8.0Hz,1H),7.22(s,1H),7.18–7.04(m,2H),6.98(d,J=7.6Hz,1H),5.29(s,2H),5.06(d,J=7.0Hz,1H),4.76(dd,J=15.2,7.0Hz,1H),4.62(d,J=12.8Hz,1H),4.48(dd,J=14.0,7.4Hz,1H),4.36(dt,J=11.8,5.8Hz,1H),3.87(d,J=13.4Hz,1H),3.72(d,J=13.6Hz,1H),2.89(d,J=11.4Hz,1H),2.78–2.59(m,3H),2.42(d,J=8.4Hz,1H),1.99–1.82(m,2H),1.52–1.37(m,2H),1.26(s,2H).
实施例22 2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-((1-(氰甲基)环丙基)甲基)-1H-苯并[d]咪唑-6-羧酸(化合物22)的合成
步骤1:2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-((1-(氰甲基)环丙基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶(96mg,0.25mmol),2-(氯甲基)-1-((1-(氰甲基)环丙基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(96mg,0.3mmol),碳酸钾(105mg,0.76mmol),碘化钾(5mg,0.03mmol)加入到乙腈(4.5mL)中,60℃搅拌1h。减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=10:1),得到淡黄色絮状产物108mg,产率62.0%。
LC-MS(ESI):[M+1]+=659.5。
步骤2:2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-((1-(氰甲基)环丙基)甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-((1-(氰甲基)环丙基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(80mg,0.12mmol)加入到1,4-二氧六环(1.5mL)中,将氢氧化锂(16mg,0.67mmol)加入到水(0.5mL)中完全溶解,再滴加入反应体系,40℃搅拌过夜。加入醋酸(0.5mL)调节PH至弱酸性。减压浓缩,用Prep-HPLC(洗脱剂乙腈:水(v/v)=40%)纯化,得到白色固体产物16.5mg,产率21.1%。
LC-MS(ESI):[M+1]+=645.3;
1H NMR(400MHz,DMSO-d6)δ12.84(s,1H),8.25(s,1H),7.83–7.80(m,1H),7.67–7.65(m,1H),7.42(d,J=8Hz,1H),7.34(d,J=8Hz,1H),7.15–6.97(m,3H),6.56(d,J=6.4Hz,1H),5.27(s,2H),4.59(s,2H),3.80(s,2H),2.92–2.89(m,2H),2.7–2.68(m,3H),1.96–1.9(m,2H),1.51–1.48(m,2H),1.32–1.29(m,2H),0.75–0.67(m,4H).
实施例23 2-((4-(2-(((4-氯-2-氟代苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(2-甲氧乙基)-1H-苯并[d]咪唑-6-羧酸(化合物23)的合成
步骤1:3-((2-甲氧基乙基)氨基)-4-硝基苯甲酸甲酯的合成
将3-氟-4-硝基苯甲酸甲酯(1g,5.02mmol)和2-甲氧基乙-1-胺(0.5mL,6.03mmol)溶于DMF(15mL),再加入碳酸钾(1.39g,10.04mmol)室温搅拌2.5h,加水,用乙酸乙酯萃取三次,干燥,浓缩,经过硅胶柱纯化(PE/EA=5/1)得黄色油状920mg,产率72.1%。
LC-MS(ESI):[M+H]+=255.2。
步骤2:4-氨基-3-((2-甲氧基乙基)氨基)苯甲酸甲酯的合成
将3-((2-甲氧基乙基)氨基)-4-硝基苯甲酸甲酯(920mg,3.62mmol)溶于二氧六环(10mL)和水(2.00mL)中,再加入锌粉(1.2g,18.10mmol)和氯化铵(968mg,18.10mmol),在室温反应2h。反应液用硅藻土过滤,滤液浓缩,得粗品为紫色油状730mg,产率90.0%。
LC-MS(ESI):[M+H]+=225.2。
步骤3:2-(氯甲基)-1-(2-甲氧基乙基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-氨基-3-((2-甲氧乙基)氨基)苯甲酸甲酯(1g,4.46mmol)和2-氯-1,1,1-三甲氧基乙烷(1.03g,6.69mmol)加入四氢呋喃(10mL)中,加入对甲苯磺酸一水合物(0.08g,0.45mmol),氮气保护下升温至50℃反应2小时。降至室温,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=20:1),得到类白色固体产物1.15g,产率91.2%。
1HNMR(400MHz,CDCl3)δ8.05(d,J=0.8Hz,1H),7.93(dd,J=8.4,1.5Hz,1H),7.71(d,J=8.4Hz,1H),4.89(s,2H),4.45(t,J=5.2Hz,2H),3.89(s,3H),3.66(t,J=5.2Hz,2H),3.20(s,3H).
步骤4:2-((4-(2-(((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(2-甲氧基乙基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶和2-(氯甲基)-1-(2-甲氧基乙基)-1H-苯并[d]咪唑-6-羧酸甲酯(72mg,0.25mmol)加入乙腈(5mL)中,加入碳酸钾(32mg,0.23mmol)和碘化钾(38mg,0.23mmol),氮气保护下升温至60℃反应1小时。减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=20:1),得到淡黄色固体产物130mg,产率90.5%。
LC-MS(ESI):[M+H]+=624.5。
步骤5:2-((4-(2-(((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(2-甲氧乙基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物2-((4-(2-(((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(2-甲氧基乙基)-1H-苯并[d]咪唑-6-羧酸甲酯(130mg,0.21mmol)加入1,4-二氧六环(1mL)和水(0.5mL)中。加入氢氧化锂(44mg,1.05mmol)。在氮气保护下升温至40℃反应2小时。加入醋酸(1mL)调节PH至弱酸性。减压浓缩,用Prep-HPLC纯化(洗脱剂CH3CN:H2O(v/v)=60%),得到白色固体产物100mg,产率77.9%。
LC-MS(ESI):[M+H]+=610.3;
1HNMR(400MHz,DMSO-d6)δ8.17(s,1H),7.80(d,J=8.4Hz,1H),7.63(d,J=8.4Hz,1H),7.42(d,J=8.0Hz,1H),7.34(d,J=8.4Hz,1H),7.14–7.08(m,2H),6.98(d,J=7.6Hz,1H),6.56(d,J=6.4Hz,1H),5.27(s,2H),4.58(t,J=5.2Hz,2H),3.78(s,2H),3.72(t,J=5.2Hz,2H),3.22(s,3H),2.84(d,J=11.2Hz,2H),2.74–2.68(m,1H),1.93(t,J=11.2Hz,2H),1.51–1.46(m,2H),1.31(d,J=11.6Hz,2H).
实施例24(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3,5-二氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物24)的合成
步骤1:4-(3,5-二氟-2-羟基苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将2-溴-4,6-二氟苯酚(0.6g,3.14mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂硼环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(1.07g,3.45mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(0.26g,0.31mmol),碳酸钾(0.87g,6.28mmol),加入到水(3mL)和1,4-二氧六环(12mL)的混合溶液中,氮气置换三次,置于90℃下反应2小时。TLC检测反应完全,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=15%)得到棕色油状产物0.896g,产率97.2%。
1H NMR(500MHz,CDCl3)δ6.83–6.77(m,2H),6.04–5.75(m,2H),4.09–4.02(m,2H),3.65–3.58(m,2H),2.51–2.39(m,2H),1.50(s,9H).
步骤2:4-(3,5-二氟-2-羟基苯基)哌啶-1-羧酸叔丁酯的合成
将4-(3,5-二氟-2-羟基苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(1g,3.21mmol),钯碳(550mg),加入到乙醇(10mL)中,氢气置换三次,室温搅拌过夜。TLC检测原料反应完全,停止反应。硅藻土抽滤,滤液减压浓缩得到了棕色固体产物966mg,产率96.0%。
LC-MS(ESI):[M-55]+=258.0;
1H NMR(500MHz,CDCl3)δ6.74–6.60(m,2H),5.61(s,1H),4.41–4.09(m,2H),3.14–3.04(m,1H),2.88-2.74(m,2H),1.81(d,J=12.9Hz,2H),1.60–1.52(m,2H),1.48(s,9H).
步骤3:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3,5-二氟苯基)哌啶-1-羧酸叔丁酯的合成
将4-(3,5-二氟-2-羟基苯基)哌啶-1-羧酸叔丁酯(356mg,1.14mmol),7-(溴甲基)-4-氯-2-氟苯并呋喃(250mg,0.95mmol),碳酸钾(262mg,1.9mmol)加入到10mL的N,N-二甲基甲酰胺中,60℃下反应2小时。TLC检测反应原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=5%)得到无色油状产物309mg,产率54.3%。
1H NMR(500MHz,CDCl3)δ7.19–7.10(m,2H),6.70–6.62(m,1H),6.60–6.52(m,1H),5.96(d,J=6.5Hz,1H),5.16(s,2H),4.18–4.05(m,2H),2.96–2.86(m,1H),2.60–2.42(m,2H),1.61–1.46(m,2H),1.43(s,9H),1.35–1.30(m,2H).
步骤4:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3,5-二氟苯基)哌啶的合成
将4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3,5-二氟苯基)哌啶-1-羧酸叔丁酯(253mg,0.51mmol),加入到三氟乙酸(4mL)和二氯甲烷(4mL)中,室温搅拌30分钟。TLC检测原料完全消失,停止反应。减压浓缩蒸干溶剂,用饱和的碳酸氢钠水溶液调节体系的PH至6左右,用乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩得到了棕色的固体产物187mg,产率92.6%。
LC-MS(ESI):[M+H]+=396.2;
1H NMR(500MHz,CDCl3)δ7.27(s,1H),7.20(d,J=7.9Hz,1H),6.87–6.74(m,1H),6.69(d,J=9.2Hz,1H),6.06(d,J=6.3Hz,1H),5.25(s,2H),3.42(d,J=12.7Hz,2H),3.14–3.02(m,1H),2.78(t,J=11.8Hz,2H),1.94–1.77(m,2H),1.71–1.56(m,2H).
步骤5:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3,5-二氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3,5-二氟苯基)哌啶(150mg,0.38mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(112mg,0.38mmol),N,N-二异丙基乙胺(245mg,1.9mmol)加入到乙腈(10mL)中,60℃反应过夜。TLC检测原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=35%)得到无色油状产物153mg,产率61.6%。
LC-MS(ESI):[M+H]+=654.5;
1H NMR(500MHz,CDCl3)δ8.15(s,1H),8.12(s,1H),8.00(d,J=8.6Hz,1H),7.96(d,J=8.5Hz,1H),7.79(d,J=8.6Hz,1H),7.75(d,J=8.5Hz,1H),7.28–7.20(m,1H),6.03(d,J=6.5Hz,1H),5.21(s,2H),4.77–4.57(m,4H),4.40–4.36(m,1H),3.95(s,2H),3.94(s,3H),2.96–2.82(m,3H),2.80–2.67(m,2H),2.52–2.37(m,2H),2.17–2.07(m,2H),1.57–1.52(m,2H).
步骤6:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3,5-二氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3,5-二氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(153mg,0.23mmol),氢氧化锂一水合物(49mg,1.17mmol)加入到水(1mL)和1,4-二氧六环(4mL)的混合溶液中,40℃反应2小时。TLC检测原料完全消失,停止加热。用20%的醋酸水溶液调节体系PH至7左右,用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=25%)得到白色固体产物101mg,产率64.1%。
LC-MS(ESI):[M+H]+=640.4;
1H NMR(500MHz,DMSO-d6)δ12.59(s,1H),8.27(s,1H),7.81(d,J=8.4Hz,1H),7.65(d,J=8.4Hz,1H),7.42(d,J=8.0Hz,1H),7.34(d,J=8.1Hz,1H),7.22–7.13(m,1H),6.94–6.85(m,1H),6.54(d,J=6.3Hz,1H),5.23(s,2H),5.11–5.02(m,1H),4.83–4.72(m,1H),4.68–4.58(m,1H),4.55–4.46(m,1H),4.39–4.31(m,1H),3.88(d,J=13.5Hz,1H),3.74(d,J=13.5Hz,1H),2.91(d,J=11.1Hz,1H),2.80(d,J=11.3Hz,1H),2.76–2.63(m,2H),2.46–2.34(m,1H),2.05–1.90(m,2H),1.57–1.42(m,2H),1.36–1.26(m,2H).
实施例25(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物25)的合成
步骤1:4-(4-氟-2-羟基苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将2-溴-5-氟苯酚(0.6g,3.14mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂硼环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(1.07g,3.45mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(0.26g,0.31mmol),碳酸钾(0.87g,6.28mmol),加入到水(3mL)和1,4-二氧六环(12mL)的混合溶液中,氮气置换三次,置于90℃下反应2小时。TLC检测反应完全,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=15%)得到棕色油状产物0.697g,产率75.6%。
LC-MS(ESI):[M-55]+=238.2。
步骤2:4-(4-氟-2-羟基苯基)哌啶-1-羧酸叔丁酯的合成
将4-(4-氟-2-羟基苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(789mg,2.69mmol),钯碳(400mg),加入到甲醇(15mL)中,氢气置换三次,室温搅拌过夜。TLC检测原料反应完全,停止反应。硅藻土抽滤,滤液减压浓缩得到了淡黄色固体产物756mg,产率95.2%。
LC-MS(ESI):[M-55]+=240.2;
1H NMR(500MHz,CDCl3)δ7.05–6.99(m,1H),6.60–6.52(m,2H),6.31(s,1H),4.36–4.17(m,2H),3.02–2.92(m,1H),2.89–2.74(m,2H),1.83–1.75(m,2H),1.62–1.54(m,2H),1.49(s,9H).
步骤3:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟苯基)哌啶-1-羧酸叔丁酯的合成
将4-(4-氟-2-羟基苯基)哌啶-1-羧酸叔丁酯(308mg,1.04mmol),7-(溴甲基)-4-氯-2-氟苯并呋喃(250mg,0.95mmol),碳酸钾(262mg,1.9mmol)加入到10mL的N,N-二甲基甲酰胺中,60℃下反应2小时。TLC检测反应原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=5%)得到无色油状产物367mg,产率80.9%。
1H NMR(500MHz,CDCl3)δ7.30–7.23(m,2H),7.09(t,J=8.5Hz,1H),6.75–6.59(m,2H),6.04(d,J=6.6Hz,1H),5.25(s,2H),4.30–4.12(m,2H),3.09–3.01(m,1H),2.81–2.65(m,2H),1.80–1.73(m,2H),1.61–1.49(m,2H),1.47(m,9H).
步骤4:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟苯基)哌啶的合成
将4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟苯基)哌啶-1-羧酸叔丁酯(267mg,0.56mmol),加入到三氟乙酸(5mL)和二氯甲烷(5mL)中,室温搅拌30分钟。TLC检测原料完全消失,停止反应。减压浓缩蒸干溶剂,用饱和的碳酸氢钠水溶液调节体系的PH至7左右,用乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩得到了棕色的固体产物直接用于下一步反应。
LC-MS(ESI):[M+H]+=378.3。
步骤5:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟苯基)哌啶(128mg,0.34mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(100mg,0.34mmol),N,N-二异丙基乙胺(220mg,1.75mmol)加入到乙腈(5mL)中,60℃反应过夜。TLC检测原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=35%)得到无色油状产物151mg,产率69.8%。
LC-MS(ESI):[M+H]+=636.4;
1H NMR(500MHz,CDCl3)δ8.17(s,1H),7.96(dd,J=8.5,1.6Hz,1H),7.75(d,J=8.5Hz,1H),7.30–7.23(m,2H),7.15–7.08(m,1H),6.73–6.61(m,2H),6.04(d,J=6.6Hz,1H),5.25(s,2H),5.23–5.17(m,1H),4.79–4.58(m,3H),4.42–4.35(m,1H),3.96(s,2H),3.94(s,3H),2.80–2.62(m,3H),2.52–2.39(m,1H),2.34–2.18(m,1H),1.86–1.75(m,2H),1.84–1.75(m,2H),1.72–1.62(m,2H)
步骤6:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将甲基(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸盐(92mg,0.14mmol),氢氧化锂一水合物(30.3mg,0.74mmol)加入到2mL水和6mL 1,4-二氧六环的混合溶液中,40℃反应2小时。TLC检测原料完全消失,停止加热。用20%的醋酸水溶液调节体系PH至7左右,用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=25%)得到淡黄色固体产物48mg,产率50.6%。
LC-MS(ESI):[M+H]+=622.3;
1H NMR(500MHz,CDCl3)δ8.25(s,1H),7.80(d,J=8.5Hz,1H),7.63(d,J=8.4Hz,1H),7.50–7.42(m,2H),7.19(t,J=7.7Hz,1H),7.07(d,J=11.3Hz,1H),6.76–6.71(m,1H),6.54(d,J=6.4Hz,1H),5.38(s,2H),5.10–5.03(m,1H),4.80–4.73(m,1H),4.65–4.59(m,1H),4.51–4.45(m,1H),4.38–4.32(m,1H),3.90(d,J =13.5Hz,1H),3.76(d,J=13.6Hz,1H),2.99–2.91(m,1H),2.88–2.80(m,2H),2.74–2.64(m,1H),2.44–2.36(m,1H),2.19–1.96(m,3H),1.68–1.50(m,3H).
实施例26(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物26)的合成
步骤1:4-(5-氟-6-羟基苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将2-溴-4-氟苯酚(600mg,3.14mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂硼环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(1.07g,3.45mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(0.26g,0.31mmol),碳酸钾(0.86g,6.28mmol),加入到水(3mL)和1,4-二氧六环(12mL)的混合溶液中,氮气置换三次,置于90℃下反应2小时。TLC检测反应完全,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=15%)得到无色油状产物896mg,产率97.2%。
LC-MS(ESI):[M-55]+=238.1;
1H NMR(500MHz,CDCl3)δ6.92–6.73(m,3H),6.05–5.80(m,2H),4.07(s,2H),3.63(t,J=5.7Hz,2H),2.46(s,2H),1.50(s,9H).
步骤2:4-(5-氟-2-羟基苯基)哌啶-1-羧酸叔丁酯的合成
将4-(5-氟-6-羟基苯基)-3,6-二氢吡啶-1(2H)-羧酸盐(1.4g,4.77mmol),钯碳(700mg),加入到甲醇(15mL)中,氢气置换三次,室温搅拌过夜。TLC检测原料反应完全,停止反应。硅藻土抽滤,滤液减压浓缩得到棕色固体产物896mg,产率63.6%。
LC-MS(ESI):[M-55]+=240.2;
1H NMR(500MHz,CDCl3)δ6.81(dd,J=9.8,2.7Hz,1H),6.77–6.69(m,2H),6.05(s,1H),4.36–4.08(m,2H),3.07–2.97(m,1H),2.92–2.72(m,2H),1.88–1.76(m,2H),1.59–1.51(m,2H),1.49(s,9H).
步骤3:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟苯基)哌啶-1-羧酸叔丁酯的合成
将4-(2-氟-6-羟基苯基)哌啶-1-羧酸叔丁酯(308mg,1.04mmol),7-(溴甲基)-4-氯-2-氟苯并呋喃(250mg,0.95mmol),碳酸钾(262mg,2mmol)加入到N,N-二甲基甲酰胺(8mL)中,60℃下反应2小时。TLC检测反应原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=5%)得到无色油状产物262mg,产率57.8%。
1H NMR(500MHz,CDCl3)δ7.30–7.22(m,2H),6.92–6.81(m,3H),6.04(d,J=6.6Hz,1H),5.25(s,2H),4.22(s,2H),3.15–3.05(m,1H),2.75(s,2H),1.77(d,J=12.9Hz,2H),1.58–1.49(m,2H),1.47(s,9H).
步骤4:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟苯基)哌啶的合成
将4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟苯基)哌啶-1-羧酸叔丁酯(326mg,0.68mmol),加入到三氟乙酸(5mL)和二氯甲烷(5mL)中,室温搅拌30分钟。TLC检测原料完全消失,停止反应。减压浓缩蒸干溶剂,用饱和的碳酸氢钠水溶液调节体系的PH至7左右,用乙酸乙酯萃取(15mL×3),合并有机相, 无水硫酸钠干燥,减压浓缩得到了褐色油状产物直接用于下一步。
LC-MS(ESI):[M+H]+=378.3;
1H NMR(500MHz,CDCl3)δ7.29–7.19(m,2H),6.97–6.87(m,3H),6.05(d,J=6.8Hz,1H),5.25(s,2H),3.54–3.42(m,2H),3.27–3.18(m,1H),3.01–2.91(m,2H),2.03–1.94(m,2H),1.46–1.37(m,2H).
步骤5:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟苯基)哌啶(192mg,0.51mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(150mg,0.51mmol),N,N-二异丙基乙胺(328mg,2.54mmol)加入到乙腈(10mL)中,60℃反应过夜。TLC检测原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=35%)得到淡黄色油状产物242mg,产率74.8%。
LC-MS(ESI):[M+H]+=636.5;
1H NMR(500MHz,CDCl3)δ8.17(s,1H),7.96(d,J=8.6Hz,1H),7.74(d,J=8.5Hz,1H),7.29–7.21(m,2H),6.93–6.81(m,3H),6.03(d,J=6.6Hz,1H),5.23(s,2H),4.80–4.57(m,4H),4.45–4.36(m,1H),3.96–3.94(m,2H),3.94(s,3H),3.06–2.89(m,3H),2.80–2.69(m,1H),2.50–2.38(m,1H),2.31–2.17(m,2H),1.84–1.75(m,2H),1.69–1.59(m,2H).
步骤6:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(242mg,0.38mmol),氢氧化锂一水合物(79.8mg,1.9mmol)加入到水(3mL)和1,4-二氧六环(9mL)的混合溶液中,40℃反应2小时。TLC检测原料完全消失,停止加热。用20%的醋酸水溶液调节体系PH至7左右,用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=25%)得到白色固体产物106mg,产率40%。
LC-MS(ESI):[M+H]+=622.3;
1H NMR(500MHz,DMSO-d6)δ12.71(s,1H),8.27(s,1H),7.80(d,J=8.4Hz,1H),7.64(d,J=8.4Hz,1H),7.47–7.38(m,2H),7.17–7.14(m,1H),7.05–6.95(m,2H),6.53(d,J=6.4Hz,1H),5.33(s,2H),5.11–4.96(m,1H),4.83–4.73(m,1H),4.68–4.59(m,1H),4.54–4.44(m,1H),4.39–4.31(m,1H),3.91(d,J=13.6Hz,1H),3.77(d,J=13.5Hz,1H),3.00–2.79(m,3H),2.77–2.65(m,1H),2.44–2.33(m,1H),2.18–2.04(m,2H),1.73–1.46(m,4H).
实施例27(S)-2-((4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物27)的合成
步骤1:4-溴-2-(2,2-二乙氧基乙氧基)-1-甲苯的合成
将化合物5-溴-2-甲基苯酚(25g,134.4mmol),2-溴-1,1-二乙氧基乙烷(31.8g,162.3mmol)和碳酸钾(37.1g,268.8mmol)加入到N,N-二甲基甲酰胺(250mL)中,氮气保护下120℃搅拌过夜。加入水(300 mL),用乙酸乙酯(100mL×3)萃取。合并有机相,用饱和食盐水(30mL)洗涤,有机相用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1%),得到浅褐色液体产物40g,产率98.5%。
步骤2:4-溴-7-甲基苯并呋喃的合成
将化合物4-溴-2-(2,2-二乙氧基乙氧基)-1-甲苯(40g,132.2mmol)和多聚磷酸(68g,829.4mmol)加入到1,2-二氯乙烷(400mL)中,氮气保护下85℃反应过夜。减压除去溶剂,加入水(100mL),用二氯甲烷萃取(100mL×2),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE=100%),得到无色液体产物17g,产率60.8%。
1HNMR(400MHz,CDCl3)δ7.68(d,J=4Hz,1H),7.30–7.26(m,1H),6.97(d,J=8.0Hz,1H),6.80(d,J=4Hz,1H),2.48(s,3H).
步骤3:7-甲基苯并呋喃-4-氰基的合成
将化合物4-溴-7-甲基苯并呋喃(16g,75.8mmol)加入到N,N-二甲基甲酰胺(160mL)中,加入氰化亚铜(33.95g,378.5mmol),氮气保护下120℃反应过夜。加入水(500mL),用乙酸乙酯(150mL×3)萃取,合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂:PE=100%),得到无色液体产物8.3g,产率69.7%。
1HNMR(400MHz,CDCl3)δ7.78(d,J=4Hz,1H),7.49(d,J=8Hz,1H),7.16(d,J=8Hz,1H),6.98(s,1H),2.59(s,3H).
步骤4:2-氯-7-甲基苯并呋喃-4-氰基的合成
将化合物7-甲基苯并呋喃-4-氰基(1g,6.4mmol)加入到四氢呋喃(15mL)。在氮气保护下,置于-78℃冷却。在该温度下,加入二异丙基氨基锂(2.6mL),搅拌一小时。加入六氯乙烷(1.6g,6.8mmol),撤去低温泵,搅拌过夜。0℃下加入水(45mL),加入乙酸乙酯(50mL×3)萃取,合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=5%),得到无色液体产物800mg,产率65.6%。
1HNMR(400MHz,CDCl3)δ7.48(d,J=8Hz,1H),7.15(d,J=8Hz,1H),6.81(s,1H),2.56(s,3H).
步骤5:7-(溴甲基)-2-氯苯并呋喃-4-腈的合成
将化合物2-氯-7-甲基苯并呋喃-4-氰基(800mg,4.2mmol),偶氮二异丁腈(137mg,0.8mmol)和N-溴代丁二酰亚胺(1.1g,6.2mmol)加入到1,2-二氯乙烷(36mL)中,氮气保护下85℃反应过夜。加入水(50mL),用二氯甲烷萃取(30mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1%),得到无色液体产物700mg,产率61.9%。
1HNMR(400MHz,CDCl3)δ7.56(d,J=8.0Hz,1H),7.39(d,J=8.0Hz,1H),6.86(s,1H),4.71(s,2H).
步骤6:4-(2-((2-氯-4-氰基苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(3-氟-2-羟基苯基)哌啶-1-羧酸叔丁酯(100mg,0.34mmol)、7-(溴甲基)-2-氯苯并呋喃-4-腈(138mg,0.51mmol)加入N,N-二甲基甲酰胺(5mL)中。加入碳酸钾(141mg,1.02mmol),60℃搅拌1小时。减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=1:10),得到浅褐色固体产物130mg,产率78.8%。
1HNMR(400MHz,CDCl3)δ7.55(d,J=7.8Hz,1H),7.45(d,J=7.8Hz,1H),7.01–6.90(m,2H),6.87(d,J=6.8Hz,1H),6.81(s,1H),5.33(s,2H),4.16–4.07(m,2H),2.97–2.92(m,1H),2.64–2.57(m,2H),1.50–1.38(m,11H),1.21–1.16(m,2H).
步骤7:2-氯-7-((2-氟-6-(哌啶-4-基)苯氧基)甲基)苯并呋喃-4-腈的合成
将化合物4-(2-((2-氯-4-氰基苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-羧酸叔丁酯(130mg,0.27mmol)加入乙酸乙酯(5mL)中,加入盐酸1.4-二氧六环(5mL),室温搅拌2小时。减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=10:1),得到浅褐色固体产物100mg,产率96.5%。
LC-MS(ESI):[M+H]+=385.4。
步骤8:(S)-2-((4-(2-((2-氯-4-氰基苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(84mg,0.31mmol)、碳酸钾(108mg,0.78mmol)、碘化钾(5mg,0.03mmol)加到乙腈(1mL)中,搅拌五分钟,加入化合物2-氯-7-((2-氟-6-(哌啶-4-基)苯氧基)甲基)苯并呋喃-4-腈(100mg,0.26mmol),60℃搅拌2小时。加入水(5mL),用乙酸乙酯(10mL×3)萃取,合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=3:2),得到类白色固体产物100mg,产率59.9%。
LC-MS(ESI):[M+H]+=643.5;
1H NMR(400MHz,CDCl3)δ8.08(s,1H),7.91(d,J=8.6Hz,1H),7.68(d,J=8.5Hz,1H),7.55(d,J=7.8Hz,1H),7.44(d,J=7.8Hz,1H),7.01–6.87(m,3H),6.81(s,1H),5.32(s,2H),5.17–5.09(m,1H),4.74–4.51(m,3H),4.44–4.38(m,1H),3.91–3.85(m,2H),3.88(s,3H),2.92–2.85(m,2H),2.69–2.61(m,1H),2.42–2.38(m,1H),2.06–2.01(m,2H),1.65–1.51(m,5H).
步骤9:(S)-2-((4-(2-((2-氯-4-氰基苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(2-((2-氯-4-氰基苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(100mg,0.16mmol)加入到二氧六环(2mL)和水(0.5mL)中。加入氢氧化锂(23mg,0.96mmol),40℃搅拌4小时。用盐酸(0.5N)调节PH至6~7,用乙酸乙酯(5mL×3)萃取,合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥。减压浓缩,用Prep-HPLC(洗脱剂CH3CN:H2O(v/v)=50%)纯化,得到白色固体产物22.4mg,产率22.3%。
LC-MS(ESI):[M+H]+=629.3;
1HNMR(400MHz,DMSO-d6)δ12.72(s,1H),8.26(s,1H),7.87(d,J=7.8Hz,1H),7.80(d,J=8.5Hz,1H),7.64(d,J=8.4Hz,1H),7.55(d,J=7.8Hz,1H),7.44(s,1H),7.12(dt,J=13.5,8.1Hz,2H),7.01(d,J=7.4Hz,1H),5.39(s,2H),5.18–5.01(m,1H),4.77(dd,J=15.2,7.2Hz,1H),4.63(d,J=13.0Hz,1H),4.55–4.45(m,1H),4.36(dd,J=6.0,3.0Hz,1H),3.89(d,J=13.5Hz,1H),3.74(d,J=13.5Hz,1H),2.92(d,J=10.8Hz,1H),2.79(d,J=10.8Hz,1H),2.76-2.68(m,2H),2.47–2.37(m,1H),1.99–1.91(m,2H),1.58–1.41(m,2H),1.36–1.33(m,2H).
实施例28(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟吡啶-2-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物28)的合成
步骤1:3-(苄氧基)-2-氯-4-氟吡啶的合成
将化合物2-氯-4-氟吡啶-3-醇(500mg,3.39mmol)加入到丙酮(10mL)中,加入碳酸钾(702mg,5.08mmol)搅拌五分钟,加入苄溴(695mg,4.06mmol)置于50℃搅拌过夜。减压浓缩,加入水(10mL),乙酸乙酯(10mL),萃取,有机层干燥浓缩得到粗品,粗品通过柱层析(洗脱剂PE:EA(v/v)=10:1)纯化得到无色油状产物550mg,产率68.3%。
1H NMR(400MHz,CDCl3)δ8.00(dd,J=7.2,5.6Hz,1H),7.41(dd,J=7.6,1.6Hz,2H),7.36–7.24(m,3H),6.95(dd,J=9.6,5.2Hz,1H),5.12(s,2H).
步骤2:3-(苄氧基)-4-氟-3',6'-二氢-(2,4'-联吡啶)-1'(2'H)-羧酸叔丁酯的合成
将化合物3-(苄氧基)-2-氯-4-氟吡啶(500mg,2.10mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂硼环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(650mg,2.10mmol),碳酸钾(581mg,4.20mmol)加入到1,4-二氧六环(10mL)和水(1mL)中,用氮气置换三遍气体,加入1,1'-双二苯基膦二茂铁二氯化钯(15mg,0.02mmol),再次用氮气置换。置于85℃搅拌过夜。减压浓缩,加入水(10mL),乙酸乙酯(10mL)萃取,合并有机相,减压浓缩,通过柱层析(洗脱剂PE:EA(v/v)=5:1)纯化得到橙色油状产物860mg,产率99%。
1H NMR(400MHz,CDCl3)δ8.18(dd,J=6.8,5.6Hz,1H),7.34–7.23(m,5H),6.91(dd,J=10.4,5.2Hz,1H),6.28–6.20(m,1H),4.95(s,2H),3.98(s,2H),3.49(s,2H),2.46(s,2H),1.42(s,9H).
步骤3:4-(4-氟-3-羟基吡啶-2-基)哌啶-1-甲酸叔丁酯的合成
将化合物3-(苄氧基)-4-氟-3',6'-二氢-(2,4'-联吡啶)-1'(2'H)-羧酸叔丁酯(840mg,2.18mmol)加入到甲醇(10mL)中,加入钯碳(84mg,0.79mmol),用氢气置换三遍,置于室温搅拌三十分钟。用硅藻土过滤,加入甲醇(10mL)洗涤,减压浓缩,通过柱层析(洗脱剂PE:EA(v/v)=1:1)纯化得到无色油状产物640mg,产率98.8%。
LC-MS(ESI):[M-55]+=241.3;
1H NMR(400MHz,CDCl3)δ8.03–7.99(m,1H),6.90–6.86(m,1H),4.21–4.16(m,2H),3.38–3.21(m,1H),2.82–2.78(m,2H),1.82–1.71(m,4H),1.40(s,9H).
步骤4:4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟吡啶-2-基)哌啶-1-甲酸叔丁酯的合成
将化合物4-(4-氟-3-羟基吡啶-2-基)哌啶-1-甲酸叔丁酯(300mg,1.01mmol)加入到N,N-二甲基甲酰胺(5mL)中,加入碳酸钾(419mg,3.03mmol),室温搅拌5min,加入7-(溴甲基)-4-氯-2-氟-1-苯并呋喃(293mg,1.11mmol),60℃搅拌两个小时,减压浓缩,通过柱层析(洗脱剂PE:EA(v/v)=8:1)纯化得到淡黄色固体产物390mg,产率80.4%。
1H NMR(400MHz,CDCl3)δ8.22–8.18(m,1H),7.20–7.14(m,2H),6.93–6.91(m,1H),5.98(d,J=6.4Hz,1H),5.27(s,2H),4.21–4.05(m,2H),3.21–2.95(m,1H),2.58(s,2H),1.81–1.65(m,4H),1.38(s,9H).
步骤5:3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟-2-(哌啶-4-基)吡啶盐酸盐的合成
将化合物4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟吡啶-2-基)哌啶-1-甲酸叔丁酯(390mg,0.81mmol)加入到盐酸二氧六环溶液(5mL)中,置于室温搅拌1.5个小时。减压浓缩得到类白色固体产物300mg,产率97.3%。
LC-MS(ESI):[M+H]+=379.3。
步骤6:(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟吡啶-2-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟-2-(哌啶-4-基)吡啶盐酸盐(130mg,0.34mmol)加入到N,N-二甲基甲酰胺(3mL)中,加入碳酸钾(142mg,1.03mmol)室温搅拌五分钟,加入(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(99mg,0.38mmol),置于60℃搅拌1.5小时。减压浓缩,通过柱层析(洗脱剂PE:EA(v/v)=1:1)纯化得到淡黄色固体产物132mg,产率60.4%。
1H NMR(400MHz,CDCl3)δ8.18(dd,J=7.1,5.6Hz,1H),8.07(s,1H),7.89(d,J=8.3Hz,1H),7.67(d,J=8.5Hz,1H),7.20–7.14(m,2H),6.88(dd,J=10.9,5.3Hz,1H),5.97(d,J=6.6Hz,1H),5.24(s,2H),5.14–5.08(m,1H),4.86–4.44(m,3H),4.35–4.30(m,1H),3.88(s,3H),2.99–2.96(m,1H),2.93–2.86(m,2H),2.74–2.57(m,1H),2.41–4.36(m,1H),2.10–1.98(m,2H),1.82–1.75(m,2H),1.58–1.38(m,4H).
步骤7:(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟吡啶-2-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-氟吡啶-2-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(110mg,0.17mmol)加入到1,4-二氧六环(1.2mL)和水(0.2mL)中,置于40℃搅拌四个小时。加入醋酸(0.1mL)调节PH至5~6,用Prep-HPLC纯化(洗脱剂乙腈:水(v/v)=60%),得到白色固体产物73mg,产率67.9%。
LC-MS(ESI):[M+H]+=623.3;
1H NMR(400MHz,DMSO-d6)δ12.75(s,1H),8.29–8.27(m,2H),7.80(dd,J=8.4,1.6Hz,1H),7.64(d,J=8.4Hz,1H),7.42(d,J=8.0Hz,1H),7.36(d,J=8.4Hz,1H),7.27(dd,J=11.2,5.2Hz,1H),6.57(d,J=6.4Hz,1H),5.34(s,2H),5.14–4.99(m,1H),4.80(dd,J=15.2,7.2Hz,1H),4.65(dd,J=15.2,2.4Hz,1H),4.48(dd,J=13.6,7.6Hz,1H),4.43–4.36(m,1H),3.89(d,J=13.6Hz,1H),3.72(d,J=13.6Hz,1H),2.88–2.84(m,2H),2.79–2.63(m,2H),2.46–2.39(m,1H),2.03–1.86(m,2H),1.77–1.55(m,2H),1.27–1.24(m,2H).
实施例29(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟吡啶-2-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物29)的合成
步骤1:3-(苄氧基)-2-氯-5-氟吡啶的合成
将化合物2-氯-5-氟吡啶-3-醇(2g,13.56mmol),苄溴(2.78g,16.27mmol),碳酸钾(3.75g,27.11mmol)加入到N,N-二甲基甲酰胺(20mL)中,反应液于室温下搅拌过夜,将溶液过滤,减压下浓缩滤液,硅胶柱层析(洗脱剂:PE/EA(v/v)=10:1)得到白色固体产物2.6g,产率80.7%。
LC-MS(ESI):[M+H]+=238.2;
1HNMR(400MHz,CDCl3)δ7.82(d,J=4.0Hz,1H),7.39–7.26(m,5H),6.96–6.92(m,1H),5.09(s,2H).
步骤2:3-(苄氧基)-5-氟-3',6'-二氢-[2,4'-联吡啶]-1'(2'H)-羧酸叔丁酯的合成
将化合物加入3-(苄氧基)-2-氯-5-氟吡啶(2g,8.42mmol),4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(3.1g,10.03mmol),(1,1'-双(二苯基膦)二茂铁)二氯钯(II)(0.18g,0.25mmol)加入到1,4-二氧六环(30mL)和水(8mL)中,溶液用氮气置换三次,反应液于90℃下搅拌过夜,冷却至室温,减压下浓缩反应液,硅胶柱层析(洗脱剂:PE/EA(v/v)=5:1),得到无色油状液体2.9g,产率89.6%。
LC-MS(ESI):[M+H]+=385.5;
1H NMR(400MHz,CDCl3)δ8.00(d,J=2.4Hz,1H),7.38–7.23(m,5H),6.94(dd,J=10.2,2.4Hz,1H),6.36(d,J=29.8Hz,1H),5.03(s,2H),4.06–3.99(m,2H),3.53(t,J=5.6Hz,2H),2.61–2.54(m,2H),1.41(s,9H).
步骤3:4-(5-氟-3-羟基吡啶-2-基)哌啶-1-羧酸叔丁酯的合成
将化合物3-(苄氧基)-5-氟-3',6'-二氢-[2,4'-联吡啶]-1'(2'H)-羧酸叔丁酯(1g,2.60mmol),钯碳(0.2g,1.88mmol)加入到甲醇(50mL)中,溶液用氢气置换三次,于氢气氛围下反应2小时,用硅藻土过滤, 浓缩滤液得到淡黄色固体产物0.7g,产率90.8%。
LC-MS(ESI):[M+H-56]+=241.3;
1HNMR(400MHz,DMSO-d6)δ7.96(d,J=2.4Hz,1H),7.00(dd,J=10.4,2.6Hz,1H),4.03(d,J=12.2Hz,2H),3.20–3.11(m,1H),2.92–2.72(m,2H),1.71–1.54(m,4H),1.41(s,9H).
步骤4:4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟吡啶-2-基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(5-氟-3-羟基吡啶-2-基)哌啶-1-羧酸叔丁酯(300mg,1.01mmol),7-(溴甲基)-4-氯-2-氟-1-苯并呋喃(400mg,1.52mmol),碳酸钾(279mg,2.02mmol)加入到N,N-二甲基甲酰胺(8mL),溶液于60℃下反应2小时,冷却至室温,减压下浓缩反应液,加入水(20mL),乙酸乙酯萃取(20mL×3),无水硫酸钠干燥,硅胶柱层析(洗脱剂:PE/EA(v/v)=5:1),得到无色油状液体330mg,产率68.1%。
LC-MS(ESI):[M-55]+=423.4;
1HNMR(400MHz,DMSO-d6)δ8.12(d,J=2.4Hz,1H),7.66(dd,J=11.0Hz,2.4Hz,1H),7.46(s,2H),6.56(d,J=6.4Hz,1H),5.46(s,2H),4.10–3.94(m,2H),3.25-3.14(m,1H),2.76(s,2H),1.70–1.51(m,4H),1.39(s,9H).
步骤5:(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟吡啶-2-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟吡啶-2-基)哌啶-1-羧酸叔丁酯(200mg,0.42mmol)加入到盐酸/1,4-二氧六环溶液(4M,2mL)中,室温下反应1小时,减压下浓缩反应液,往溶液中加入乙腈(6mL),N,N-二异丙基乙胺(1mL),碳酸钾(174mg,1.26mmol),碘化钾(7mg,0.04mmol),溶液于60℃下反应2小时,冷却至室温,减压下浓缩反应液,硅胶柱纯化(洗脱剂:DCM/MeOH(v/v)=20:1),得到淡黄色固体产物140mg,产率42.1%。
LC-MS(ESI):[M+H]+=637.5。
步骤6:(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟吡啶-2-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-5-氟吡啶-2-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(70mg,0.11mmol),氢氧化锂(46mg,1.10mmol)加入到1,4-二氧六环(3mL)和水(1mL)中,溶液于40℃下搅拌2小时,冷却至室温,缓慢滴加醋酸至溶液为弱酸性,减压下浓缩反应液,制备纯化(洗脱剂:ACN/H2O(v/v)=1:1),得到白色固体产物16mg,产率23.4%。
LC-MS(ESI):[M+H]+=623.4;
1HNMR(400MHz,DMSO-d6)δ8.23(s,1H),8.12(d,J=2.4Hz,1H),7.80(d,J=8.4Hz,1H),7.69–7.57(m,2H),7.49–7.41(m,2H),6.56(d,J=6.4Hz,1H),5.45(s,2H),5.14–5.05(m,1H),4.84–4.73(m 1H),4.73–4.60(m,1H),4.52–4.45(m,1H),4.41–4.33(m,1H),3.91(d,J=13.4Hz,1H),3.75(d,J=13.4Hz,1H),3.12–2.88(m,2H),2.85–2.80(m,1H),2.79–2.63(m,1H),2.45–2.40(m,1H),2.21–2.03(m,2H),1.83–1.56(m,4H).
实施例30(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-6-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物30)的合成
步骤1:4-(2-氟-6-羟基苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将2-溴-3-氟苯酚(600mg,3.14mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂硼环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(1.08g,3.46mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(0.25g,0.31mmol),碳酸钾(0.86g,3.28mmol),加入到水(4mL)和1,4-二氧六环(12mL)的混合溶液中,氮气置换3次,置于90℃下反应2小时。TLC检测反应完全,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=15%)得到无色油状产物591mg,产率64.1%。
步骤2:4-(2-氟-6-羟基苯基)哌啶-1-羧酸叔丁酯的合成
将4-(2-氟-6-羟基苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(591mg,2.01mmol),钯碳(300mg),加入到甲醇(15mL)中,氢气置换三次,室温搅拌过夜。TLC检测原料反应完全,停止反应。硅藻土抽滤,滤液减压浓缩得到了棕色固体产物563mg,产率94.6%。
LC-MS(ESI):[M+H-56]+=240.2;
1H NMR(500MHz,CDCl3)δ6.98(td,J=8.2,6.3Hz,1H),6.64–6.50(m,3H),4.30–4.09(m,2H),3.23–3.15(m,1H),2.85–4.70(m,2H),2.25–4.12(m,2H),1.63–1.53(m,2H),1.49(s,9H).
步骤3:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-6-氟苯基)哌啶-1-羧酸叔丁酯的合成
将4-(2-氟-6-羟基苯基)哌啶-1-羧酸叔丁酯(308mg,1.04mmol),7-(溴甲基)-4-氯-2-氟苯并呋喃(250mg,0.95mmol),碳酸钾(262mg,2mmol)加入到10mL的N,N-二甲基甲酰胺中,60℃下反应2小时。TLC检测反应原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=5%)得到无色油状产物262mg,产率57.8%。
LC-MS(ESI):[M-55]+=422.2;
1H NMR(500MHz,CDCl3)δ7.35–7.20(m,2H),7.14–7.07(m,1H),6.76(d,J=8.3Hz,1H),6.71–6.58(m,1H),6.03(d,J=6.6Hz,1H),5.28(s,2H),4.32–4.14(m,2H),3.32–3.23(m,1H),2.82–2.61(m,2H),2.17–2.06(m,2H),1.62–1.53(m,2H),1.46(s,9H).
步骤4:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-6-氟苯基)哌啶的合成
将4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-6-氟苯基)哌啶-1-羧酸叔丁酯(362mg,0.76mmol),加入到盐酸/1,4-二氧六环溶液(4M,4mL)中,室温搅拌30分钟。TLC检测原料完全消失,停止反应。减压浓缩蒸干溶剂,用饱和的碳酸氢钠水溶液调节体系的PH至7左右,用乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩得到了棕色的固体产物256mg,产率89.5%。
LC-MS(ESI):[M+H]+=378.3;
1H NMR(500MHz,CDCl3)δ7.39–7.19(m,2H),7.16–7.08(m,1H),6.74(d,J=8.4Hz,1H),6.69(dd,J=10.5,8.4Hz,1H),6.03(d,J=6.6Hz,1H),5.32(s,2H),3.49(d,J=12.5Hz,2H),3.42–3.32(m,1H),3.00–2.89(m,2H),2.57–2.43(m,2H),1.83(d,J=14.0Hz,2H).
步骤5:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-6-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基 甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-6-氟苯基)哌啶(150mg,0.4mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(117mg,0.4mmol),N,N-二异丙基乙胺(256mg,1.9mmol)加入到乙腈(10mL)中,60℃反应过夜。TLC检测原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=35%)得到无色油状产物124mg,产率49%。
LC-MS(ESI):[M+H]+=636.4;
1H NMR(500MHz,CDCl3)δ8.19(s,1H),7.96(dd,J=8.5,1.5Hz,1H),7.73(d,J=8.5Hz,1H),7.33–7.21(m,2H),7.14–7.05(m,1H),6.75(d,J=8.3Hz,1H),6.71–6.58(m,1H),6.03(d,J=6.6Hz,1H),5.27(s,2H),5.23–5.18(m,1H),4.74–4.58(m,3H),4.42–4.36(m,1H),3.95(s,3H),3.21–3.13(m,1H),2.98–2.81(m,2H),2.80–2.61(m,1H),2.51–2.38(m,1H),2.26–2.12(m,4H),1.74–1.60(m,4H).
步骤6:((S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-6-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-6-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(126mg,0.2mmol),氢氧化锂一水合物(41.5mg,0.99mmol)加入到水(2mL)和1,4-二氧六环(6mL)的混合溶液中,40℃反应2小时。TLC检测原料完全消失,停止加热。用20%的醋酸水溶液调节体系PH至7左右,用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=25%)得到白色固体产物89mg,产率64.8%。
LC-MS(ESI):[M+H]+=622.4;
1H NMR(500MHz,DMSO-d6)δ8.26(s,1H),7.83–7.77(m,1H),7.63(d,J=8.4Hz,1H),7.45–7.40(m,2H),7.22–7.17(m,1H),7.01(d,J=8.3Hz,1H),6.80–6.72(m,1H),6.52(d,J=6.4Hz,1H),5.38(s,2H),5.10–5.05(m,1H),4.73–4.69(m,1H),4.59–4.56(m,1H),4.50–4.43(m,1H),4.37–4.32(m,1H),3.89(d,J=13.5Hz,1H),3.73(d,J=13.6Hz,1H),3.11–3.03(m,1H),2.97–2.90(m,1H),2.84–2.77(m,1H),2.70–2.62(m,1H),2.42–2.33(m,1H),2.16–1.96(m,4H),1.51–1.39(m,2H).
实施例31(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-4-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物31)的合成
步骤1:3'-羟基-3,6-二氢-[4,4'-联吡啶]-1(2H)-羧酸叔丁酯的合成
将4-溴吡啶-3-醇(1g,5.75mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂硼环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(1.78g,5.75mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(0.47g,0.57mmol),碳酸钾(1.59g,2mmol),加入到水(3mL)和1,4-二氧六环(12mL)的混合溶液中,氮气置换三次,置于90℃下反应2小时。TLC检测反应完全,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取 (15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=15%)得到棕色油状产物579mg,产率62.1%。
LC-MS(ESI):[M+H]+=277.2;
1H NMR(500MHz,CDCl3)δ8.30(s,1H),8.05(d,J=5.0Hz,1H),7.13(d,J=4.9Hz,1H),6.23(s,1H),4.15–4.01(m,2H),3.69–3.57(m,2H),2.61–2.50(m,2H),1.50(s,9H).
步骤2:4-(3-羟基吡啶-4-基)哌啶-1-羧酸叔丁酯的合成
将3'-羟基-3,6-二氢-[4,4'-联吡啶]-1(2H)-羧酸叔丁酯(927mg,3.35mmol),钯碳(400mg),加入到甲醇(15mL)中,氢气置换三次,室温搅拌过夜。TLC检测原料反应完全,停止反应。硅藻土抽滤,滤液减压浓缩得到了棕色固体产物901mg,产率96.5%。
LC-MS(ESI):[M+H]+=279.3;
1H NMR(500MHz,CDCl3)δ8.41–8.17(s,1H),8.04(d,J=5.0Hz,1H),7.13(d,J=4.8Hz,1H),4.35–4.15(m,2H),3.22–3.03(m,1H),2.95–2.67(m,2H),1.87(d,J=12.9Hz,2H),1.65–1.54(m,2H),1.48(s,9H).
步骤3:4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-4-基)哌啶-1-羧酸叔丁酯的合成
将4-(3-羟基吡啶-4-基)哌啶-1-羧酸叔丁酯(147.5mg,0.53mmol),7-(溴甲基)-4-氯-2-氟苯并呋喃(140mg,0.53mmol),碳酸钾(146.8mg,2mmol)加入到N,N-二甲基甲酰胺(10mL)中,60℃下反应2小时。TLC检测反应原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=5%)得到无色油状产物114mg,产率46.5%。
LC-MS(ESI):[M+H]+=461.3;
1H NMR(500MHz,CDCl3)δ8.33(s,1H),8.23(d,J=4.9Hz,1H),8.01(s,1H),7.30–7.24(m,2H),7.10(d,J=4.8Hz,1H),6.04(d,J=6.6Hz,1H),5.38(s,2H),4.22(s,2H),3.14–3.06(m,1H),2.81–2.69(m,2H),1.78(d,J=13.0Hz,2H),1.60–1.53(m,1H),1.47(s,9H).
步骤4:3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-(哌啶-4-基)吡啶的合成
将4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-4-基)哌啶-1-羧酸叔丁酯(100mg,0.22mmol),加入到三氟乙酸(4mL)和二氯甲烷(4mL)中,室温搅拌30分钟。TLC检测原料完全消失,停止反应。减压浓缩蒸干溶剂,用饱和的碳酸氢钠水溶液调节体系的PH至7左右,用乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩得到了棕色的固体产物直接用于下一步反应。
LC-MS(ESI):[M+H]+=361.1。
步骤5:(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-4-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-4-(哌啶-4-基)吡啶(100mg,0.28mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(81mg,0.28mmol),N,N-二异丙基乙胺(179mg,1.39mmol)加入到10mL的乙腈溶液中,60℃反应过夜。TLC检测原料完全消失,停止加热,冷却至室温,加入饱和食盐水(50mL),用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=35%)得到无色油状产物87mg,产率50.7%。
LC-MS(ESI):[M+H]+=619.4;
1H NMR(500MHz,CDCl3)δ8.32(s,1H),8.23(d,J=4.9Hz,1H),8.16–8.14(m,1H),7.97(d,J=8.5Hz,1H),7.75(d,J=8.5Hz,1H),7.30–7.23(m,2H),7.12(d,J=4.9Hz,1H),6.04(d,J=6.7Hz,1H),5.37(s,2H),5.24–5.17(m,1H),4.76–4.50(m,3H),4.42–4.35(m,1H),3.98–3.95(m,2H),3.95(s,3H),3.03–2.91(m,3H),2.78–2.69(m,1H),2.49–2.40(m,1H),2.32–2.16(m,2H),1.85–1.75(m,2H),1.70–1.64(m,2H).
步骤6:(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-4-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将(S)-2-((4-(3-((4-氯-2-氟苯并呋喃-7-基)甲氧基)吡啶-4-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(80mg,0.13mmol),氢氧化锂一水合物(27.1mg,0.65mmol)加入到水(2mL)和1,4-二氧六环(6mL)的混合溶液中,40℃反应2小时。TLC检测原料完全消失,停止加热。用20%的醋酸水溶液调节体系PH至7左右,用乙酸乙酯萃取(10mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=25%)得到白色固体产物41mg,产率51%。
LC-MS(ESI):[M+H]+=605.4;
1H NMR(500MHz,CDCl3)δ8.36(s,1H),8.25(d,J=4.9Hz,1H),8.19(s,1H),8.05(d,J=8.5Hz,1H),7.80(d,J=8.5Hz,1H),7.30–7.24(m,2H),7.14(d,J=5.0Hz,1H),6.04(d,J=6.6Hz,1H),5.38(s,2H),5.24–5.16(m,1H),4.81–4.51(m,3H),4.42–4.34(m,1H),4.02(s,2H),3.09–2.96(m,3H),2.77–2.67(m,1H),2.49–2.38(m,1H),2.38–2.25(m,2H),1.87–1.77(m,2H),1.76–1.64(m,2H).
实施例32(S)-2-((4-(3-氯-2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物32)的合成
步骤1:1-溴-3-氯-2-(甲氧基甲氧基)苯的合成
将化合物2-溴-6-氯苯酚(5.0g,24.10mmol)加入DMF(50mL)中,加入氢化钠(1.27g,53.02mmol),冰浴搅拌十分钟,室温搅拌半小时。加入溴(甲氧基)甲烷(4.48g,36.15mmol),室温搅拌一小时。加入水(100mL),用乙酸乙酯(100mL×3)萃取,合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥后,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:40)得到无色油状产物6.0g,产率99.0%。
1H NMR(400MHz,DMSO-d6)δ7.48(dd,J=8.2Hz,1.4Hz,1H),7.35(dd,J=8.2,1.4Hz,1H),6.94(t,J=8.2Hz,1H),5.19(s,2H),3.71(s,3H).
步骤2:4-(3-氯-2-(甲氧基甲氧基)苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将化合物1-溴-3-氯-2-(甲氧基甲氧基)苯(2.0g,7.95mmol)、4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(2.95g,9.54mmol)加入到1,4-二氧六环(20mL)和水(5mL)中。加入碳酸钾(3.29g,23.85mmol),用氮气置换三次,加入四(三苯基膦)钯(184mg,0.16mmol),再次氮气置换三次,85℃搅拌过夜。减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:10)得无色油状产物2.7g,产率95.7%。
1H NMR(400MHz,DMSO-d6)δ7.23(dd,J=7.6Hz,2.0Hz,1H),6.95(s,2H),5.74(s,1H),4.95(s,2H),3.98(d,J=2.8Hz,2H),3.54(s,2H),3.50(s,3H),2.41(d,J=1.8Hz,2H),1.42(s,9H).
步骤3:4-(3-氯-2-(甲氧基甲氧基)苯基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(3-氯-2-(甲氧基甲氧基)苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(530mg,1.50mmol)加入甲醇(6mL)中。加入钯碳(53mg,0.48mmol),用氢气置换三次,室温下搅拌半小时。把钯碳滤去,加水(10mL)淬灭,用乙酸乙酯(20mL×3)萃取,合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥后,减压浓缩,用硅胶柱层析进行纯化(洗脱剂PE:DCM(v/v)=1:5)得无色油状产物320mg,产率59.9%。
LC-MS(ESI):[M+H]+=356.3。
步骤4:4-(3-氯-2-羟基苯基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(3-氯-2-(甲氧基甲氧基)苯基)哌啶-1-羧酸叔丁酯(320mg,0.90mmol)加入到乙酸(5mL)中,80℃搅拌过夜。减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=1:20)得无色油状产物75mg,产率26.7%。
LC-MS(ESI):[M-55]+=256.3。
步骤5:4-(3-氯-2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(3-氯-2-羟基苯基)哌啶-1-羧酸叔丁酯(75mg,0.24mmol)、7-(溴甲基)-4-氯-2-氟苯并呋喃(108mg,0.41mmol)加入二甲基甲酰胺(4mL)中。再加入碳酸钾(100mg,0.72mmol),60℃搅拌1小时。减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=1:10)得到浅褐色固体产物70mg,产率59.0%。
1H NMR(400MHz,DMSO-d6)δ7.34(d,J=8.2Hz,1H),7.31–7.27(m,2H),7.09–7.06(m,2H),6.05(d,J=6.6Hz,1H),5.19(s,2H),4.18(d,J=13.2Hz,2H),3.11–3.01(m,1H),2.63(dd,J=18.0,7.4Hz,2H),1.48(s,2H),1.47(s,9H),1.30–1.25(m,2H).
步骤6:4-(3-氯-2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶的合成
将化合物4-(3-氯-2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-羧酸叔丁酯(70mg,0.14mmol)加入乙酸乙酯(3mL)中,再加入盐酸1.4-二氧六环溶液(4M,3mL),常温下搅拌2小时。减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=10:1)得到浅褐色固体产物60mg,产率99.0%。
LC-MS(ESI):[M+H]+=394.3。
步骤7:(S)-2-((4-(3-氯-2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(53mg,0.18mmol)、碳酸钾(62mg,0.45mmol)、碘化钾(2mg,0.02mmol)加到乙腈(2ml)中。将化合物4-(3-氯-2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶(60mg,0.15mmol)加入乙腈(2ml)中,再滴加二异丙基乙基胺(0.5ml),把该溶液滴加到前一个溶液中,60℃搅拌2小时。加入水(5mL),用乙酸乙酯(10mL×3)萃取,合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=3:2)得到类白色固体产物60mg,产率61.3%。
LC-MS(ESI):[M+H]+=652.4。
步骤8:(S)-2-((4-(3-氯-2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(3-氯-2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(60mg,0.09mmol)溶解在二氧六环(2mL)、水(0.5mL)中。加入氢氧化锂(13mg,0.54mmol),40℃搅拌4小时。用盐酸(0.5N)调节PH为6~7,用乙酸乙酯(5mL×3)萃取,合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥。减压浓缩,用prep-HPLC(洗脱剂CH3CN:H2O(v/v)=50%)纯化得白色固体产物7.8mg,产率13.6%。
LC-MS(ESI):[M+H]+=638.2;
1H NMR(400MHz,DMSO-d6)δ12.73(s,1H),8.28(s,1H),7.81(d,J=8.4Hz,1H),7.65(d,J=8.3Hz,1H),7.48–7.34(m,3H),7.25–7.12(m,2H),6.57(dd,J=6.3Hz,3.2Hz,1H),5.19(s,2H),5.07(d,J=6.8Hz,1H),4.81–4.76(m,1H),4.64(d,J=13.2Hz,1H),4.50(dd,J=13.6Hz,7.5Hz,1H),4.36–4.34(m,1H),3.90–3.75(m,2H),2.96–2.70(m,4H),2.43–2.39(m,1H),1.99–1.98(m,2H),1.51–1.44(m,4H).
实施例33(S)-2-((4-(5-氯-2-(4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁 烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物33)的合成
步骤1:1-溴-5-氯-3-氟-2-(甲氧基甲氧基)苯的合成
将化合物2-溴-4-氯-6-氟苯酚(3.00g,13.31mmol)加入四氢呋喃(75mL)中,降温至0℃,加入氢化钠(0.8g,33.27mmol),0℃反应1小时。加入溴(甲氧基)甲烷(2.00g,15.97mmol),保持0℃反应1小时。加入水(50mL)淬灭,用乙酸乙酯(50mL×2)萃取,有机层用无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=10:1)纯化,得到无色油状产物3.31g,产率92.3%。
1HNMR(400MHz,CDCl3)δ7.29(t,J=2.0Hz,1H),7.03(dd,J=10.4,2.4Hz,1H),5.10(s,2H),3.56(s,3H).
步骤2:4-(5-氯-3-氟-2-(甲氧基甲氧基)苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将化合物1-溴-5-氯-3-氟-2-(甲氧基甲氧基)苯(1.65g,6.12mmol)和4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(1.89g,6.12mmol)加入到1,4-二氧六环(33mL)和水(8.25mL)中。加入碳酸钾(1.69g,12.25mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(0.22g,0.31mmol),氮气保护下升温至85℃反应过夜。冷却至室温,减压浓缩,用硅胶柱层析纯化(洗脱剂PE:EA(v/v)=10:1)纯化,得到淡黄色油状产物1.71g,产率75.1%。
1HNMR(400MHz,CDCl3)δ6.97(dd,J=10.4,2.4Hz,1H),6.92–6.84(m,1H),5.78(s,1H),4.95(s,2H),3.98(d,J=2.8Hz,2H),3.52(t,J=5.6Hz,2H),3.43(s,3H),2.40(d,J=1.6Hz,2H),1.42(s,9H).
步骤3:4-(5-氯-3-氟-2-(甲氧基甲基氧基)苯基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(5-氯-3-氟-2-(甲氧基甲氧基)苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(640mg,1.72mmol)加入乙酸乙酯(10mL)中,加入二氧化铂(64mg,0.13mmol),氢气置换后升温至45℃反应30分钟。用硅藻土过滤,减压浓缩,得到无色油状产物640mg,产率99.5%。
1HNMR(400MHz,CDCl3)δ6.91(dd,J=10.4,2.4Hz,1H),6.89–6.82(m,1H),5.03(s,2H),4.19(d,J=12.8Hz,2H),3.51(s,3H),3.11–3.06(m,1H),2.73(t,J=12.0Hz,2H),1.76–1.69(m,2H),1.52–1.43(m,2H),1.42(s,9H).
步骤4:4-(5-氯-3-氟-2-羟基苯基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(5-氯-3-氟-2-(甲氧基甲基氧基)苯基)哌啶-1-羧酸叔丁酯(644mg,1.72mmol)加入醋酸(10mL)中,氮气保护下升温至85℃反应过夜。降温至室温,减压浓缩,用硅胶柱层析纯化(洗脱剂:PE:EA(v/v)=5:1)纯化,得到淡黄色油状产物179mg,产率31.5%。
1HNMR(400MHz,CDCl3)δ6.91(dd,J=9.6,2.4Hz,1H),6.85(d,J=11.6Hz,1H),5.23(d,J=5.2Hz,1H),4.17(d,J=13.2Hz,2H),2.99(tt,J=12.0,3.2Hz,1H),2.75(t,J=12.0Hz,2H),1.74(d,J=12.8Hz,2H),1.57–1.50(m,2H),1.41(s,9H).
步骤5:4-(5-氯-2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(5-氯-3-氟-2-羟基苯基)哌啶-1-羧酸叔丁酯(160mg,0.49mmol)和7-(溴甲基)-4-氯-2-氟-1-苯并呋喃(153mg,0.58mmol)加入N,N-二甲基甲酰胺(5mL)中,加入碳酸钾(201mg,1.45mmol),氮 气保护下升温至60℃反应2小时。降温至室温,减压浓缩,用硅胶柱层析纯化(洗脱剂:PE:EA(v/v)=3:1),得到淡黄色油状产物206mg,产率82.9%。
1HNMR(400MHz,CDCl3)δ7.21–7.15(m,2H),6.95(dd,J=10.8,2.4Hz,1H),6.81(d,J=1.6Hz,1H),5.97(d,J=6.4Hz,1H),5.19(s,2H),4.08(dd,J=10.4,6.4Hz,2H),2.95–2.82(m,1H),2.52(s,2H),1.41(s,2H),1.39(s,9H),0.83–0.74(m,2H).
步骤6:4-(5-氯-2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶的合成
将化合物4-(5-氯-2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-羧酸叔丁酯(100mg,0.20mmol)加入乙酸乙酯(6mL)中,加入盐酸二氧六环溶液(4M,6mL),氮气保护下室温反应2小时。减压浓缩,得到类白色固体产物80mg,产率99.4%。
LC-MS(ESI):[M+H]+=412.3。
步骤7:(S)-2-((4-(5-氯-2-(4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-(5-氯-2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶(80mg,0.19mmol)和2-(氯甲基)-3-(((2S)-恶烷-2-基)甲基)苯并[d]咪唑-5-羧酸甲酯(60mg,0.20mmol)加入乙腈(5mL)中,加入碳酸钾(134mg,0.97mmol)和碘化钾(0.0mL,0.03mmol),氮气保护下升温至60℃反应2小时。降温至室温,减压浓缩,用硅胶柱层析纯化(洗脱剂:DCM:MeOH(v/v)=20:1),得到淡黄色固体产物60mg,产率46.1%。
LC-MS(ESI):[M+H]+=670.4。
步骤8:(S)-2-((4-(5-氯-2-(4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(5-氯-2-(4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(60mg,0.09mmol)加入1,4-二氧六环(2mL)和水(0.5mL)中。加入氢氧化锂(4.0mg,0.10mmol),氮气保护下升温至40℃反应2小时。降温至室温,用醋酸调节PH至6~7。减压浓缩,用Prep-HPLC纯化(洗脱剂CH3CN:H2O(v/v)=50%),得到白色固体产物20mg,产率34.0%。
LC-MS(ESI):[M+H]+=656.2;
1H NMR(400MHz,DMSO-d6)δ8.26(s,1H),7.81(d,J=8.4Hz,1H),7.64(d,J=8.4Hz,1H),7.38(ddd,J=14.4,13.4,8.0Hz,3H),7.06(s,1H),6.56(d,J=6.4Hz,1H),5.28(s,2H),5.09–5.04(m,1H),4.78(dd,J=15.2,7.2Hz,1H),4.63(d,J=13.2Hz,1H),4.49(dd,J=13.6,7.5Hz,1H),4.36–4.33(m,1H),3.88(d,J=13.6Hz,1H),3.73(d,J=13.6Hz,1H),2.91–2.65(m,4H),2.43–2.37(m,1H),2.02–1.81(m,2H),1.59–1.38(m,2H),1.33–1.23(m,2H).
实施例34 2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-((1-乙基-1H-咪唑-5-基)甲基)-1H-苯并[d]咪唑-6-羧酸(化合物34)的合成
步骤1:3-(((1-乙基-1H-咪唑-5-基)甲基)氨基)-4-硝基苯甲酸甲酯的合成
将化合物3-氟-4-硝基苯甲酸甲酯(150mg,0.75mmol),(3-乙基咪唑-4-基)甲胺(94mg,0.75mmol),碳酸钾(312mg,2.26mmol),N,N-二异丙基乙胺(195mg,1.51mmol)加入到N,N-二甲基甲酰胺(10mL) 中,50℃下搅拌3小时。冷却至室温,减压浓缩,用硅胶柱层析纯化(洗脱剂:DCM/MeOH(v/v)=15:1),得到黄色固体产物220mg,产率96.0%。
LC-MS(ESI):[M+H]+=305.1。
步骤2:4-氨基-3-(((1-乙基-1H-咪唑-5-基)甲基)氨基)苯甲酸甲酯的合成
将化合物3-(((1-乙基-1H-咪唑-5-基)甲基)氨基)-4-硝基苯甲酸甲酯(220mg,0.72mmol),钯碳(100mg,0.94mmol)加入到甲醇(20mL)中,用氢气置换三次,反应1小时。用硅藻土过滤,减压浓缩,得到褐色固体产物192mg,产率96.8%。
LC-MS(ESI):[M+H]+=275.1。
步骤3:2-(氯甲基)-1-((1-乙基-1H-咪唑-5-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-氨基-3-(((1-乙基-1H-咪唑-5-基)甲基)氨基)苯甲酸甲酯(220mg,0.80mmol),2-氯-1,1,1-三甲氧基乙烷(248mg,1.60mmol),一水对甲苯磺酸(76mg,0.40mmol),加入到乙腈(10mL)中,60℃下反应2小时。降至室温,减压浓缩,用硅胶柱层析纯化(洗脱剂:DCM/MeOH(v/v)=1:1),得到淡黄色固体产物200mg,产率74.9%。
LC-MS(ESI):[M+H]+=333.2;
1HNMR(400MHz,DMSO-d6)δ7.98–7.76(m,2H),7.50–7.46(m,2H),7.13–7.08(m,1H),5.10(s,2H),4.05–3.97(m,2H),3.91–3.87(m,2H),3.86(s,3H),1.17(t,J=8.0Hz,3H).
步骤4:2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-((1-乙基-1H-咪唑-5-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶(110mg,0.29mmol),2-(氯甲基)-1-((1-乙基-1H-咪唑-5-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(110mg,0.29mmol),碘化钾(5mg,0.03mmol),碳酸钾(121mg,0.88mmol)加入到乙腈(10mL)中,60℃下反应2小时。冷却至室温,减压浓缩,用硅胶柱层析纯化(洗脱剂:DCM/MeOH(v/v)=15:1),得到淡黄色固体产物45mg,产率22.9%。
LC-MS(ESI):[M+H]+=674.1。
步骤5:2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-((1-乙基-1H-咪唑-5-基)甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-((1-乙基-1H-咪唑-5-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(45mg,0.07mmol),氢氧化锂(10mg,0.42mmol)加入到1,4-二氧六环(2mL)和水(0.5mL)中,40℃搅拌2小时。冷却至室温,缓慢滴加醋酸至溶液PH为弱酸性。减压浓缩,用Prep-HPLC纯化(洗脱剂:ACN/H2O(v/v)=1:1),得到白色固体产物13.7mg,产率31.1%。
LC-MS(ESI):[M+H]+=660.2;
1HNMR(400MHz,DMSO-d6)δ12.83(s,1H),8.11–8.08(m,1H),7.83–7.79(m,1H),7.71–7.66(m,2H),7.43–7.39(m,1H),7.34–7.30(m,1H),7.16–7.05(m,2H),6.88–6.84(m,1H),6.57(t,J=8.0Hz,1H),6.36(s,1H),5.70(s,2H),5.25(s,2H),4.05–3.95(m,2H),3.75(s,2H),2.80–2.73(m,2H),2.68–2.58(m,1H),1.90–1.80(m,2H),1.25–1.11(m,7H).
实施例35(S)-2-((4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物35)的合成
步骤1:2-((4-甲氧基苄基)氧基)-3',6'-二氢-[3,4'-联吡啶]-1'(2H)-羧酸叔丁酯的合成
将化合物3-溴-2-((4-甲氧基苄基)氧基)吡啶(1.0g,3.40mmol)、4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(1.26g,4.08mmol)加入到1,4-二氧六环(10mL)和水(2.5mL)中。加入碳酸钾(1.41g,10.2mmol),用氮气置换三次,加入1,1'-双二苯基膦二茂铁二氯化钯(124mg,0.17mmol),再次氮气置换三次,85℃搅拌过夜。减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:10)得到无色油状产物1.14g,产率84.6%。
1HNMR(400MHz,CDCl3)δ8.08–8.07(m,1H),7.43(s,1H),7.36(d,J=8Hz,2H),6.91–6.86(m,3H),5.87(s,1H),5.35(s,2H),4.03(s,2H),3.82(s,3H),3.55(s,2H),2.52(s,2H),1.48(s,9H).
步骤2:4-(2-羟基吡啶-3-基)哌啶-1-羧酸叔丁酯的合成
将化合物2-((4-甲氧基苄基)氧基)-3',6'-二氢-[3,4'-联吡啶]-1'(2'H)-羧酸叔丁酯(1.1g,2.77mmol)加入甲醇(11mL)中。加入钯碳(110mg,0.89mmol),用氢气置换三次,室温下搅拌半小时。用硅藻土过滤,加入水(10mL),用乙酸乙酯(20mL×3)萃取。合并有机相,有机相使用饱和食盐水洗一次,用无水硫酸钠干燥。减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc=100%),得到无色油状产物685mg,产率88.8%。
LC-MS(ESI):[M+H]+=279.3。
步骤3:4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-羧酸叔丁酯的合成
将化合物4-(2-羟基吡啶-3-基)哌啶-1-羧酸叔丁酯(685mg,2.46mmol)、7-(溴甲基)-2,4-二氯苯并呋喃(972mg,3.69mmol)加入N,N-二甲基甲酰胺(10mL)中。加入碳酸钾(1.0g,7.38mmol),60℃搅拌1小时。减压浓缩,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=1:5),得到浅褐色固体产物140mg,产率11.9%。
LC-MS(ESI):[M+H]+=477.3;
1HNMR(400MHz,CDCl3)δ7.98(d,J=4Hz,1H),7.37(d,J=8Hz,1H),7.24–7.15(m,2H),6.85(d,J=4Hz,1H),6.65(s,1H),5.59(s,2H),3.32–3.29(m,1H),2.71(t,J=12Hz,2H),1.78(d,J=8Hz,2H),1.55–1.46(m,2H),1.40(s,9H),1.24–1.21(m,2H).
步骤4:2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-(哌啶-4-基)吡啶的合成
将化合物4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-羧酸叔丁酯(140mg,0.29mmol)加入乙酸乙酯(6mL)中,加入盐酸1.4-二氧六环(6mL),室温搅拌2小时。减压浓缩,用硅胶柱层析纯化(洗脱剂DCM:MeOH(v/v)=10:1),得到浅褐色固体产物110mg,直接用于下一步反应。
LC-MS(ESI):[M+H]+=377.3。
步骤5:(S)-2-((4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(103mg,0.35mmol)、碳酸钾(121mg,0.87mmol)、碘化钾(5mg,0.03mmol)加入乙腈(4mL)中。将化合物2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-(哌啶-4-基)吡啶(110mg,0.29mmol)、二异丙基乙胺(0.5mL)加入乙腈(4mL)中,充分溶解后,滴加入反应体系。60℃搅拌2小时。加入水(5mL),用乙酸乙酯(10mL×3)萃取, 合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥,用硅胶柱层析纯化(洗脱剂EA:PE(v/v)=3:2),得到类白色固体产物60mg,产率32.6%。
LC-MS(ESI):[M+H]+=635.5。
步骤6:(S)-2-((4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)吡啶-3-基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(60mg,0.09mmol)加入到二氧六环(2mL)、水(0.5mL)中。加入氢氧化锂(13mg,0.54mmol),40℃搅拌4小时。用盐酸(0.5N)调节PH至6~7,用乙酸乙酯(5mL×3)萃取。合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥。减压浓缩,用Prep-HPLC(洗脱剂CH3CN:H2O(v/v)=50%)纯化,得到白色固体产物21.5mg,产率38.4%。
LC-MS(ESI):[M+H]+=621.2;
1HNMR(400MHz,DMSO-d6)δ12.76(s,1H),8.28(d,J=6.4Hz,1H),8.02–7.80(m,2H),7.65–7.59(m,2H),7.42–7.17(m,3H),6.97–6.95(m,1H),5.62(s,2H),5.08–5.07(m,1H),4.79–4.33(m,4H),3.93–3.73(m,2H),2.99–2.68(m,4H),2.42–2.38(m,1H),2.18–2.14(m,2H),1.74–1.60(m,4H).
实施例36(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-氧代哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物36)的合成
步骤1:(S)-2-((4-(2-(苄氧基)-3-氟苯基)-2-氧代哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-1-(氧杂环丁烷-2-基甲基)-2-((2-氧代哌嗪-1-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(200mg,0.6mmol),2-(苄氧基)-1-溴-3-氟苯(236mg,0.8mmol),1,1-联萘-2,2-双二苯膦(64mg,0.1mmol),碳酸铯(365mg,1.1mmol)加入到甲苯(50mL)中,加入三二亚苄基丙酮二钯(50mg,0.1mmol),氮气置换后120℃反应过夜。减压浓缩,加入水(30mL),用乙酸乙酯(30mL×3)萃取,合并有机相,减压浓缩,用薄层色谱纯化(洗脱剂MeOH:DCM(v/v)=5%),得到淡黄色固体产物172mg,产率55.2%。
LC-MS:[M+H]+=559.5;
1H NMR(400MHz,CDCl3)δ8.18(s,1H),8.02(d,J=8.3Hz,1H),7.79(d,J=8.3Hz,1H),7.36–6.31(m,2H),7.26–21(m,3H),6.95–6.89(m,1H),6.85–6.80(m,1H),6.62(d,J=8.2Hz,1H),5.18–5.10(m,2H),5.03(s,2H),4.90–4.83(m,2H),4.61–4.46(m,3H),3.97(s,3H),3.81(s,2H),3.61–3.55(m,2H),3.42–3.387(m,2H),2.78–2.75(m,1H),2.49–2.42(m,1H).
步骤2:(S)-2-((4-(3-氟-2-羟基苯基)-2-氧代哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-((4-(2-(苄氧基)-3-氟苯基)-2-氧代哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(100mg,0.2mmol)加入到甲醇(1mL),加入钯碳(10mg)。氢气置换3次,常温反应2小时。硅藻土过滤,滤液减压浓缩,得到白色固体产物85mg,产率100%。
LC-MS:[M+H]+=469.4。
步骤3:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-氧代哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-((4-(3-氟-2-羟基苯基)-2-氧代哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(85mg,0.2mmol),7-(溴甲基)-4-氯-2-氟苯并呋喃(57mg,0.2mmol)和碳酸钾(75mg,0.6mmol)加入到N,N-二甲基甲酰胺(4.5mL)中,氮气保护下60℃反应。加入水(30mL),用二氯甲烷萃取(30mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,用薄层色谱纯化(洗脱剂MeOH:DCM(v/v)=5%),得到黄色液体产物43mg,产率36.4%。
LC-MS:[M+H]+=651.5。
步骤4:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-氧代哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-氧代哌嗪-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(43mg,0.06mmol)、氢氧化锂(14mg,0.6mmol)加入到1,4-二氧六环(0.8mL)和水(0.2mL)中,40℃反应三小时。加入醋酸(0.2mL)调节pH至6~7。减压浓缩,用Prep-HPLC(洗脱剂乙腈:水(v/v)=70%)纯化,得到白色固体产物10mg,产率23.8%。
LC-MS:[M+H]+=637.2;
1H NMR(400MHz,DMSO-d6)δ12.76(s,1H),8.26(d,J=0.8Hz,1H),7.81(dd,J=8.4,1.6Hz,1H),7.61(d,J=8.4Hz,1H),7.33(d,J=8.0Hz,1H),7.21(d,J=8.0Hz,1H),7.06(td,J=8.4,6.4Hz,1H),6.95–6.87(m,1H),6.80(d,J=8.0Hz,1H),6.46(d,J=6.4Hz,1H),5.26(d,J=12.0Hz,2H),5.09–4.97(m,2H),4.86–4.71(m,2H),4.60(dd,J=15.2,2.4Hz,1H),4.47(dd,J=13.6,7.6Hz,1H),4.34(dt,J=8.8,6.0Hz,1H),3.72(s,2H),3.44(t,J=5.2Hz,2H),3.33(s,2H),2.75–2.66(m,1H),2.38–2.27(m,1H).
实施例37(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-(三氟甲基)苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物37)的合成
步骤1:2-(苄氧基)-1-溴-3-(三氟甲基)苯的合成
将化合物2-溴-6-(三氟甲基)苯酚(550mg,2.28mmol)和苄溴(468mg,2.74mmol)加入N,N-二甲基甲酰胺(10mL)搅拌溶解,然后加入碳酸钾(946mg,6.85mmol),反应液氮气保护下升温至60℃反应2小时,反应液加入水(30mL)淬灭反应,用乙酸乙酯(20mL×3)萃取,有机层干燥浓缩得到粗品,粗品通过柱层析(洗脱剂PE:EA(v/v)=1:1)纯化得到类白色固体产物460mg,产率60.9%。
1H NMR(400MHz,CDCl3)δ7.73(dd,J=8.0Hz,1.2Hz,1H),7.58–7.46(m,3H),7.40–7.26(m,3H),7.06(t,J=8.0Hz,1H),5.03(s,2H).
步骤2:4-(2-(苄氧基)-3-(三氟甲基)苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将化合物2-(苄氧基)-1-溴-3-(三氟甲基)苯(460mg,1.39mmol)和4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(516mg,1.67mmol)加入二氧六环(10mL)中搅拌溶解,然后加入碳酸钾(576mg,4.17mmol),四三苯基膦钯(80mg,0.07mmol)和水(2mL),反应液氮气保护下升温至85℃反应过夜。将反应液降温至室温,减压浓缩得到粗品。粗品通过柱层析(洗脱剂PE:EA (v/v)=1:1)纯化得到类白色固体产物383mg,产率63.6%。
1H NMR(400MHz,CDCl3)δ7.48(dd,J=7.6Hz,1.3Hz,1H),7.39–7.23(m,6H),7.13(s,1H),5.87(s,1H),4.79(br.s,2H),3.98(s,2H),3.49(br.s,2H),2.44(br.s,2H),1.42(s,9H).
步骤3:4-(2-羟基-3-(三氟甲基)苯基)哌啶-1-甲酸叔丁酯的合成
将化合物4-(2-(苄氧基)-3-(三氟甲基)苯基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(383mg,0.88mmol)加入甲醇(10mL)搅拌溶解,然后加入钯碳(40mg,0.38mmol),反应液氢气氛围下室温反应3小时。反应液过滤浓缩得到无色油状产物307mg,收率100%。
1H NMR(400MHz,CDCl3)δ7.37–7.22(m,2H),6.92(t,J=7.6Hz,1H),4.19(d,J=13.2Hz,2H),3.12–2.97(m,1H),2.77(td,J=13.2Hz,2.4Hz,2H),1.76(d,J=13.2Hz,2H),1.60–1.48(m,2H),1.41(s,9H).
步骤4:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-(三氟甲基)苯基)哌啶-1-甲酸叔丁酯的合成
将化合物4-(2-羟基-3-(三氟甲基)苯基)哌啶-1-甲酸叔丁酯(287mg,0.83mmol)和7-(溴甲基)-4-氯-2-氟-1-苯并呋喃(219mg,0.83mmol)加入N,N-二甲基甲酰胺(5mL)搅拌溶解,然后加入碳酸钾(345mg,2.50mmol),反应液氮气保护下升温至60℃下搅拌3小时。反应液减压浓缩,通过柱层析(洗脱剂PE:EA(v/v)=1:1)纯化得到类白色固体产物307mg,产率70.0%。
1H NMR(400MHz,CDCl3)δ7.49–7.42(m,1H),7.39(d,J=7.6Hz,1H),7.33(d,J=8.4Hz,1H),7.26(d,J=8.4Hz,1H),7.18(d,J=9.2Hz,1H),5.99(d,J=6.8Hz,1H),5.02(s,2H),4.18(d,J=13.2Hz,2H),3.30–3.16(m,1H),2.69(td,J=13.2,2.0Hz,2H),1.70(d,J=12.4Hz,2H),1.58–1.47(m,2H),1.42(s,9H).
步骤5:4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-(三氟甲基)苯基)哌啶的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-(三氟甲基)苯基)哌啶-1-甲酸叔丁酯(220mg,0.42mmol)加入二氧六环(4mL)搅拌溶解,然后加入盐酸二氧六环溶液(4M,4mL),反应液氮气保护下室温搅拌2小时。反应液减压浓缩得到类白色固体产物150mg,收率:84.1%,直接用于下一步。
LC-MS(ESI):[M+H]+=428.3。
步骤6:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-(三氟甲基)苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-(三氟甲基)苯基)哌啶(150mg,0.35mmol)和2-(氯甲基)-3-(((2S)-氧杂环丁烷-2-基)甲基)苯并[d]咪唑-5-羧酸甲酯(104mg,0.35mmol)加入N,N-二甲基甲酰胺(5mL)搅拌溶解,然后加入碳酸钾(243mg,1.76mmol),反应液氮气保护下升温至60℃反应2小时。反应液减压浓缩,通过柱层析(洗脱剂DCM:MeOH(v/v)=20:1)纯化得到类白色固体产物150mg,产率62.4%。
1H NMR(400MHz,CDCl3)δ8.07(s,1H),7.88(d,J=8.0Hz,1H),7.66(dd,J=8.4,2.8Hz,1H),7.42(d,J=7.6Hz,2H),7.31(d,J=8.4Hz,1H),7.28–7.18(m,1H),7.15(d,J=7.6Hz,1H),5.98(d,J=6.4Hz,1H),5.15–5.05(m,1H),4.98(s,2H),4.74–4.26(m,5H),3.95–3.75(m,2H),3.86(s,3H),3.17–3.08(m,1H),2.94–2.88(m 1H),2.69–2.51(m,1H),2.47–2.04(m,3H),1.75–1.70(m,4H).
步骤7:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-(三氟甲基)苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-(三氟甲基)苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(150mg,0.22mmol)加入二氧六环(2mL)搅拌溶解,然后将氢氧化锂(46mg,1.10mmol)加入水(1mL)中溶解后加入反应液中,反应液氮气保护下升温至40℃反应2小时。加入醋酸(0.1mL)调节PH至5~6,用Prep-HPLC纯化(洗脱剂乙腈:水(v/v)=60%),得到白色固体产物97mg,产率66.0%。
1H NMR(400MHz,DMSO-d6)δ12.81(s,1H),8.28(s,1H),7.81(dd,J=8.4,1.6Hz,1H),7.70(d,J=7.6Hz,1H),7.64(d,J=8.4Hz,1H),7.59(d,J=7.2Hz,1H),7.52(d,J=8.0Hz,1H),7.44(d,J=8.4Hz,1H),7.38(t,J=8.0Hz,1H),6.62(d,J=6.4Hz,1H),5.13–5.11(m,1H),5.12(s,2H),4.82(dd,J=15.2Hz,7.2Hz,1H),4.77–4.62(m,1H),4.56–4.51(m,1H),4.41–4.27(m,1H),3.95(d,J=13.6Hz,1H),3.79(d,J=13.6Hz,1H),3.15–3.00(m,2H),2.94–2.88(m,1H),2.81–2.70(m,1H),2.47–2.37(m,1H),2.25–2.06(m,2H),1.73–1.63(m,4H).
实施例38(S)-2-((4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物38)的合成
步骤1:4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-甲酸叔丁酯的合成
将化合物4-(3-氟-2-羟基苯基)哌啶-1-甲酸叔丁酯(500mg,1.69mmol),7-(溴甲基)-2-氯-4-氟苯并呋喃(450mg,1.69mmol)和碳酸铯(1.10g,3.39mmol)溶于DMF(8mL)中。室温反应2h,加入水(30mL)淬灭反应,乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩,硅胶柱层析分离纯化(洗脱剂EtOAc:PE(v/v)=30%),得到无色液体710mg,产率81.8%。
1H NMR(500MHz,DMSO-d6)δ7.40–7.37(m,1H),7.28(s,1H),7.22–7.13(m,2H),7.13–7.03(m,1H),6.98(d,J=7.8Hz,1H),5.29(s,2H),3.98(d,J=12.9Hz,2H),2.88–2.82(m,1H),2.51–2.50(m,2H),1.39(s,9H),1.38–1.30(m,4H).
步骤2:4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶的合成
将化合物4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-甲酸叔丁酯(710mg,1.49mmol),溶于二氯甲烷(5mL)中后,加入三氟乙酸(1mL)室温反应30min。减压除去溶剂,加饱和碳酸氢钠调节PH=9,二氯甲烷萃取(8mL×3),合并有机相,无水硫酸钠干燥,减压浓缩得到白色固体546mg,产率97.3%。
1H NMR(500MHz,DMSO-d6)δ7.40–7.37(m,1H),7.27(s,1H),7.25–7.06(m,3H),6.97(d,J=7.6Hz,1H),5.28(s,2H),3.05(d,J=12.8Hz,2H),2.88–2.84(m,1H),2.51–2.45(m,2H),1.51–1.46(m,2H),1.36–1.30(m,2H).
步骤3:(S)-2-((4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶(340mg,0.9mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(318mg,1.08mmol)溶于乙腈(5mL)中后,加入N,N'-二异丙基乙胺(582mg,4.5mmol)60℃反应过夜。减压除去溶剂,加水10mL,乙酸乙酯萃取(10mL×3),无水硫酸钠干燥,减压浓缩。硅胶柱层析洗脱剂EtOAc:PE(v/v)=50%),得到白色固体449mg,产率78.4%。
1H NMR(500MHz,DMSO-d6)δ8.28(d,J=1.6Hz,1H),7.81(dd,J=8.4,1.7Hz,1H),7.67(d,J=8.5Hz,1H),7.33(dd,J=8.3,5.2Hz,1H),7.28(s,1H),7.19–7.08(m,2H),7.09–7.05(m,1H),6.97(d,J=7.7Hz,1H), 5.28(s,2H),5.07(dd,J=7.2,2.7Hz,1H),4.78(dd,J=15.3,7.2Hz,1H),4.65–4.62(m,1H),4.52–4.45(m,1H),4.37–4.33(m,1H),3.92–3.88(m,1H),3.87(s,3H),3.73(d,J=13.5Hz,1H),2.90(d,J=11.0Hz,1H),2.77(d,J=11.2Hz,1H),2.75–2.61(m,2H),2.46–2.35(m,1H),2.04–1.86(m,2H),1.49–1.39(m,2H),1.36–1.22(m,2H).
步骤4:(S)-2-((4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(0.44g,0.69mmol)和氢氧化锂一水合物(0.09g,2.08mmol)加入到1,4-二氧六环(10mL)和水(5mL)中,40℃反应1h。在冰浴中冷却,用三氟乙酸调节pH=6,用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,硅胶柱层析纯化(洗脱剂MeOH:DCM(v/v)=1:30)得到淡黄色固体产物380mg,产率83.2%。
LC-MS(ESI):[M+H]+=622.2;
1H NMR(500MHz,DMSO-d6)δ12.71(s,1H),8.27–8.24(m,1H),7.80(dd,J=8.4,1.6Hz,1H),7.64(d,J=8.4Hz,1H),7.33(dd,J=8.3,5.2Hz,1H),7.28(s,1H),7.18–7.08(m,2H),7.07–7.05(m,1H),6.98(d,J=7.8Hz,1H),5.28(s,2H),5.06(dd,J=7.1,2.8Hz,1H),4.76(dd,J=15.3,7.2Hz,1H),4.62(dd,J=15.2,2.8Hz,1H),4.51–4.46(m,1H),4.39–4.30(m,1H),3.88(d,J=13.5Hz,1H),3.72(d,J=13.6Hz,1H),2.90(d,J=11.1Hz,1H),2.77(d,J=11.0Hz,1H),2.75–2.63(m,2H),2.46–2.39(m,1H),2.00–1.86(m,2H),1.56–1.39(m,2H),1.27(t,J=12.6Hz,2H).
实施例39 2-(((2S)-4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶-1-基)甲基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸(化合物39)的合成
步骤1:(S)-2-甲基-4-(((三氟甲基)磺酰基)氧基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将化合物(S)-2-甲基-4-氧代哌啶-1-羧酸叔丁酯(3.00g,13.78mmol)溶解无水四氢呋喃(30mL)中,氮气氛围移置-78℃搅拌反应5分钟后,滴加双(三甲基硅烷基)氨基锂(17.9mL,17.92mmol),然后缓慢滴加N-苯基双(三氟甲烷磺酰)亚胺(5.53g,15.16mmol)的四氢呋喃溶液,滴加完成后继续反应30分钟。此温度下加入50mL饱和氯化铵水淬灭,分离有机相,水相用乙酸乙酯萃取(10mL×3),合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,经硅胶柱层析纯化(EtOAc:PE(v/v)=10%)得到无色透明液体4.68g,产率98.3%。
步骤2:(S)-2-甲基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将(S)-2-甲基-4-(((三氟甲基)磺酰基)氧基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(4.68g,13.55mmol),联硼酸频呐醇酯(3.69g,14.23mmol),四三苯基膦钯(0.79g,0.68mmol)和醋酸钾(2.69g,27.10mmol)加入至1,4-二氧六环(80mL)中,氮气氛围下加热至90℃反应20分钟。自然冷却至室温,加水(250mL),乙酸乙酯萃取(15mL×3),无水硫酸钠干燥,减压浓缩,经硅胶柱层析(EtOAc:PE(v/v)=10%)得无色液体产物4.08g,产率93.1%。
步骤3:(S)-4-(2-(苄氧基)-3-氟苯基)-2-甲基-3,6-二氢吡啶-1(2H)-羧酸叔丁酯的合成
将化合物(S)-2-甲基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼环戊烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(1.26g,3.90mmol),2-(苄氧基)-1-溴-3-氟苯(1.10g,3.90mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(0.16g,0.19mmol)和碳酸钾(1.08g,7.74mmol)加入1,4-二氧六环(40mL)和水(8mL)的混合溶液中,氮气氛围下加热至90℃反应2h。自然降温至室温,加水(100mL)和乙酸乙酯萃取(15mL×3),无水硫酸钠干燥,减压浓缩,硅胶柱层析(EtOAc:PE(v/v)=10%)得无色透明油状物1.23g,产率79.4%。
1H NMR(500MHz,CDCl3)δ7.47–7.32(m,5H),7.11–6.99(m,2H),7.00–6.90(m,1H),5.82–5.68(m,1H),5.08–4.89(m,2H),4.73–4.43(m,1H),4.37–4.24(m,0.5H),4.19–4.02(m,0.5H),3.72–3.57(m,0.5H),2.97–2.83(m,0.6H),2.81–2.72(m,0.4H),2.56–2.43(m,0.5H),2.31–2.13(m,1H),1.52(s,9H),1.22(d,J=6.7Hz,2H),1.10(d,J=6.8Hz,1H).
步骤4:(2S)-4-(3-氟-2-羟基苯基)-2-甲基哌啶-1-羧酸叔丁酯的合成
将化合物(S)-4-(2-(苄氧基)-3-氟苯基)-2-甲基-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(1.23g,3.09mmol)和钯碳(0.24g,mmol)加入甲醇(10mL)中,氢气氛围下室温下反应过夜。反应液用硅藻土过滤,滤液减压浓缩,得到无色透明油状物产物0.95g,产率99.0%。
LC-MS(ESI):[M-55]+=254.1;
1H NMR(500MHz,CDCl3)δ6.99–6.90(m,2H),6.87–6.77(m,1H),5.57–5.38(m,1H),4.05–3.90(m,1H),3.87–3.79(m,1H),3.34–3.25(m,1H),3.19–3.08(m,1H),2.26–2.12(m,1H),1.93–1.82(m,1H),1.65–1.56(m,1H),1.51(s,9H),1.26(d,J=6.3Hz,3H).
步骤5:(2S)-4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶-1-羧酸叔丁酯的合成
将化合物(2S)-4-(3-氟-2-羟基苯基)-2-甲基哌啶-1-羧酸叔丁酯(300mg,0.97mmol),7-(溴甲基)-4-氯-2-氟苯并呋喃(255mg,0.97mmol)和碳酸铯(638mg,1.94mmol)溶于DMF(5mL)中。室温反应2h,加水(30mL),乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩,经硅胶柱层析分离纯化(洗脱剂EtOAc:PE(v/v)=30%),得到无色液体420mg,产率88.0%。
1H NMR(500MHz,CDCl3)δ7.28–7.25(m,2H),7.06–6.91(m,3H),6.05–6.03(m,1H),5.37–5.28(m,2H),3.75–3.562(m,2H),3.24–3.17(m,1H),3.08–3.03(m,1H),2.06–1.92(m,1H),1.56–1.46(m,3H),1.52–1.45(m,9H),1.17(d,J=6.4Hz,3H).
步骤6:(2S)-4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶的合成
将化合物(2S)-4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶-1-羧酸叔丁酯(420mg,0.91mmol)加入二氯甲烷(5mL)中溶解,加入三氟乙酸(1mL,13.46mmol),室温反应30min。减压除去溶剂,加水(10mL),冰浴下滴加饱和碳酸氢钠水溶液调节PH=9,二氯甲烷萃取(8mL×3),无水硫酸钠干燥,减压浓缩,得淡黄色油状物328mg,产率98.1%。
LC-MS(ESI):[M+H]+=392.1。
步骤7:2-(((2S)-4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶-1-基)甲基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(2S)-4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶(328mg,0.84mmol)和(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(296mg,1.0mmol)溶于乙腈(10mL)中后,室温搅拌下加入N,N'-二异丙基乙胺(541mg,4.14mmol),加热至60℃反应过夜。减压除去溶剂,加入水(50mL)和乙酸乙酯萃取(15mL×3)萃取,无水硫酸钠干燥,减压浓缩,经硅胶柱层析(洗脱剂EtOAc:PE(v/v)=50%)得无色透明油状产物540mg,产率99.2%。
LC-MS(ESI):[M+H]+=650.2;
1H NMR(500MHz,DMSO-d6)δ8.28(d,J=1.7Hz,1H),7.82(dd,J=8.5,1.6Hz,1H),7.68(d,J=8.5Hz,1H),7.39(d,J=8.1Hz,1H),7.30(d,J=8.1Hz,1H),7.14–7.02(m,2H),6.97(d,J=7.8Hz,1H),6.52(d,J=6.4Hz,1H),5.34–5.20(m,2H),5.18–5.08(m,1H),4.85–4.64(m,2H),4.49–4.40(m,2H),4.26–4.19(m,1H),3.88(s,3H),3.45(d,J=13.9Hz,1H),2.76–2.60(m,3H),2.40–2.29(m,1H),2.15–2.06(m,1H),1.43–1.12(m,5H),1.04(d,J=6.1Hz,3H).
步骤8:2-(((2S)-4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶-1-基)甲基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物2-(((2S)-4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶-1-基)甲基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(540mg,0.83mmol)和氢氧化锂一水合物(174mg,4.11mmol)溶于1,4-二氧六环(5mL)和水(1mL)的混合溶剂中,40℃反应1.5h,冰浴环境下滴加稀冰醋酸调节pH=6左右,用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,经硅胶柱层析纯化(洗脱剂MeOH:DCM(v/v)=10%)得到白色固体产物113mg,产率19.9%。
LC-MS(ESI):[M+H]+=636.2;
1H NMR(500MHz,DMSO-d6)δ12.71(br.s,1H),8.25(s,1H),7.80(dd,J=8.4,1.6Hz,1H),7.65(d,J=8.4Hz,1H),7.40(d,J=8.1Hz,1H),7.31(d,J=8.1Hz,1H),7.16–7.09(m,1H),7.09–7.04(m,1H),6.97(d,J=7.8Hz,1H),6.52(d,J=6.5Hz,1H),5.32–5.22(m,2H),5.17–5.11(m,1H),4.82–4.75(m,1H),4.74–4.66(m,1H),4.51–4.40(m,2H),4.26–4.20(m,1H),3.48–3.40(m,1H),2.79–2.61(m,3H),2.38–2.30(m,1H),2.14–2.05(m,1H),2.05–1.92(m,2H),1.43–1.35(m,1H),1.22–1.18(m,1H),1.13–1.07(m,1H),1.07–1.03(m,3H).
实施例40 2-(((2S)-4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶-1-基)甲基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸(化合物40)的合成
步骤1:(2S)-4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶-1-羧酸叔丁酯的合成
将化合物(2S)-4-(3-氟-2-羟基苯基)-2-甲基哌啶-1-羧酸叔丁酯(300mg,0.97mmol),7-(溴甲基)-2-氯-4-氟苯并呋喃(255mg,0.97mmol)和碳酸铯(638mg,1.94mmol)溶于DMF(5mL)中。室温反应2h,加水(30mL),乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩,经硅胶柱层析分离纯化(洗脱剂EtOAc:PE(v/v)=30%),得到无色液体450mg,产率94.3%。
1H NMR(500MHz,CDCl3)δ7.35–7.29(m,1H),7.06–6.91(m,4H),6.75–6.71(m,1H),5.36–5.31(m,2H),3.75–3.57(m,2H),3.29–3.17(m,1H),3.14–3.02(m,1H),2.01–1.90(m,1H),1.56–1.46(m,3H),1.52–1.45(m,9H),1.18(d,J=6.4Hz,3H).
步骤2:(2S)-4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶的合成
将化合物(2S)-4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶-1-羧酸叔丁酯(450mg,0.91mmol)加入二氯甲烷(5mL)中溶解,加入三氟乙酸(1mL,13.46mmol),室温反应30min。减压除去溶剂,加入水(10mL),冰浴下滴加饱和碳酸氢钠水溶液调节PH=9,二氯甲烷萃取(8mL×3),无水硫酸钠干燥,减压浓缩,得淡黄色油状物355mg,产率99.0%。
LC-MS(ESI):[M+H]+=392.2;
1H NMR(500MHz,CDCl3)δ7.18–7.14(m,1H),7.01–6.92(m,2H),6.91–6.82(m,2H),6.66(d,J=2.2Hz,1H),5.24(s,2H),3.32–3.24(m,1H),3.11–3.00(m,1H),2.95–2.84(m,1H),2.78–2.68(m,1H),1.81–1.67(m,1H),1.59–1.43(m,3H),1.20–1.15(m,3H).
步骤3:2-(((2S)-4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶-1-基)甲基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(2S)-4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶(355mg,0.91mmol)和(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(320mg,1.09mmol)溶于乙腈(10mL)中后,室温搅拌下加入N,N'-二异丙基乙胺(585mg,4.53mmol),加热至60℃反应过夜。减压除去溶剂,加入水(50mL)和乙酸乙酯萃取(15mL×3),无水硫酸钠干燥,减压浓缩,经硅胶柱层析(洗脱剂EtOAc:PE(v/v)=50%)得无色透明油状产物528mg,产率89.6%。
LC-MS(ESI):[M+H]+=650.3;
1H NMR(500MHz,DMSO-d6)δ8.28(s,1H),7.82(d,J=8.5,1.6Hz,1H),7.68(d,J=8.5Hz,1H),7.31(dd,J=8.3,5.2Hz,1H),7.25(s,1H),7.17–7.10(m,2H),7.05(d,J=7.9,5.3Hz,1H),6.95(d,J=7.7Hz,1H),5.28(q,J=11.7Hz,2H),5.19–5.10(m,1H),4.84–4.77(m,1H),4.71(dd,J=15.4,6.1Hz,1H),4.51–4.41(m,2H),4.26–4.19(m,1H),3.88(s,3H),3.45(d,J=13.9Hz,1H),2.76–2.60(m,3H),2.40–2.29(m,1H),2.15–2.06(m,1H),1.98(s,1H),1.43–1.29(m,1H),1.27–1.20(m,2H),1.16–1.10(m,1H),1.04(d,J=6.1Hz,3H).
步骤4:2-(((2S)-4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶-1-基)甲基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物2-(((2S)-4-(2-((2-氯-4-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-2-甲基哌啶-1-基)甲基)-1-(((S)-氧杂环丁烷-2-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(528mg,0.81mmol)和氢氧化锂一水合物(170mg,4.05mmol)溶于1,4-二氧六环(5mL)和水(1mL)的混合溶剂中,40℃反应1.5h,冰浴环境下滴加稀冰醋酸调节pH=6左右,用乙酸乙酯萃取(15mL×3),合并有机相,用无水硫酸钠干燥,减压浓缩,经硅胶柱层析纯化(洗脱剂MeOH:DCM(v/v)=10%)得到白色固体产物314mg,产率58.1%。
LC-MS(ESI):[M+H]+=636.2;
1H NMR(500MHz,DMSO-d6)δ12.71(br.s,1H),8.25(s,1H),7.80(d,J=8.6Hz,1H),7.40(d,J=8.1Hz,1H),7.31(d,J=8.1Hz,1H),7.19–7.02(m,2H),6.97(d,J=7.8Hz,1H),6.57–6.47(m,1H),5.37–5.21(m,2H),5.18–5.09(m,1H),4.83–4.74(m,1H),4.72–4.65(m,1H),4.50–4.39(m,2H),4.27–4.18(m,1H),3.49–3.41(m,1H),2.78–2.58(m,3H),2.40–2.30(m,1H),2.12–1.93(m,3H),1.31–1.20(m,4H),1.08–1.02(m,3H).
实施例41(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物41)的合成
步骤1:4-(3-氟-2-(甲氧基甲氧基)苯基)-5,6-二氢吡啶-2(1H)-酮的合成
将化合物1-溴-3-氟-2-(甲氧基甲氧基)苯(1.5g,6.38mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)- 5,6-二氢吡啶-2(1H)-酮(1.42g,6.38mmol),四三苯基磷钯(0.22g,0.19mmol),醋酸钠(1.35g,12.76mmol)加入到1,4-二氧六环(40mL)和水(10mL)中,80℃下搅拌4小时。过滤反应液,减压浓缩,用硅胶柱层析纯化(PE:EA(v/v)=10:1),得到无色油状产物1.28g,产率80.2%。
LC-MS(ESI):[M+H]+=252.4;
1HNMR(400MHz,CDCl3)δ7.50–7.37(m,1H),7.08–6.93(m,2H),6.04(s,1H),5.05(s,2H),3.48(t,J=8.0Hz,2H),3.44(s,3H),2.72(t,J=8.0Hz,2H).
步骤2:(S)-2-((4-(3-氟-2-(甲氧基甲氧基)苯基)-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-(3-氟-2-(甲氧基甲氧基)苯基)-5,6-二氢吡啶-2(1H)-酮(350mg,1.39mmol)加入到四氢呋喃(10mL)中,冷却至0℃。加入氢化钠(133mg,5.54mmol),0℃搅拌半小时。加入(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(410mg,1.39mmol),升温至室温反应2小时。加入水(5mL)淬灭反应,用乙酸乙酯(5mL×2)萃取。有机层用无水硫酸钠干燥,减压浓缩,用硅胶柱层析(洗脱剂:PE:EA(v/v)=5:1)纯化,得到类白色固体产物190mg,产率26.8%。
LC-MS(ESI):[M+H]+=510.2。
步骤3:(S)-2-((4-(3-氟-2-羟基苯基)-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并(d)咪唑-6-羧酸甲酯的合成
将化合物(S)-2-((4-(3-氟-2-(甲氧基甲氧基)苯基)-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并(d)咪唑-6-羧酸甲酯(190mg,0.22mmol)加入到二氯甲烷(30mL)中,冷却至-10℃后滴加三氟乙酸(5mL),溶液于-10℃搅拌过夜。减压浓缩,用硅胶柱层析纯化(洗脱剂:PE:EA(v/v)=3:1),得到淡黄色固体产物65mg,产率62.4%。
LC-MS(ESI):[M+H]+=466.2;
1HNMR(400MHz,DMSO-d6)δ10.02(s,1H),8.28(s,1H),7.86–7.80(m,1H),7.74–7.70(m,1H),7.24–7.15(m,1H),7.13–7.07(m,1H),6.89–6.80(m,1H),6.16(s,1H),5.13–5.00(m,2H),4.93–4.70(m,2H),4.68–4.58(m,1H),4.52–4.42(m,1H),4.40–4.31(m,1H),3.88(s,3H),3.66(t,J=8.0Hz,2H),2.78(t,J=8.0Hz,2H),2.84–2.65(m,1H),2.42–2.29(m,1H).
步骤4:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-((4-(3-氟-2-羟基苯基)-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(65mg,0.14mmol),7-(溴甲基)-4-氯-2-氟代苯并呋喃(73mg,0.28mmol),碳酸钾(58mg,0.42mmol)加入到N,N-二甲基甲酰胺(3mL)中,氮气保护下升温至60℃反应两小时。冷却至室温,用乙酸乙酯(10mL×2)萃取,有机层用无水硫酸钠干燥,减压浓缩,用硅胶柱层析(洗脱剂:DCM:MeOH(v/v)=20:1)纯化,得到淡黄色固体产物70mg,产率77.4%。
LC-MS(ESI):[M+H]+=648.4。
步骤5:(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-3-氟苯基)-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(70mg,0.11mmol)加入到1,4-二氧六环(4mL)和水(1mL)中,加入氢氧化锂(23mg,0.55mmol),氮气保护下升温至40℃,搅拌2小时。反应液冷却至室温,滴加醋酸至溶液PH=6,减压浓缩,用Prep-HPLC纯化(洗脱剂ACN:H2O(v/v)=50%)得到白色固体产物10.3mg,产率15.0%。
LC-MS(ESI):[M+H]+=634.2;
1HNMR(400MHz,DMSO-d6)δ8.26(s,1H),7.86–7.80(m,1H),7.69–7.61(m,1H),7.39–7.08(m,3H),7.19–7.08(m,2H),6.35(d,J=4.0Hz,1H),5.82(s,1H),5.27(s,2H),5.06–4.95(m,2H),4.84–4.71(m,2H),4.65–4.56(m,1H),4.50–4.42(m,1H),4.38–4.30(m,1H),2.75–2.64(m,2H),2.60–2.53(m,2H),2.39–2.26(m,2H).
实施例42(S)-2-((4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-氟苯基)-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸(化合物42)的合成
步骤1:(S)-2-((4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-氟苯基)-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-((4-(3-氟-2-羟基苯基)-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(67mg,0.14mmol)加入到N,N-二甲基甲酰胺(2mL),加入碳酸钾(60mg,0.43mmol)搅拌5min,加入7-(溴甲基)-2,4-二氯苯并呋喃(60mg,0.21mmol)置于60℃搅拌1.5个小时。减压浓缩,加入水(5mL)和乙酸乙酯(5mL)萃取。合并有机相,有机相使用饱和食盐水洗一次,无水硫酸钠干燥后,减压浓缩,用硅胶柱层析纯化(洗脱剂EtOAc:PE(v/v)=1:1)得到淡黄色絮状产物90mg,产率94.6%。
LC-MS(ESI):[M+H]+=664.4;
1HNMR(400MHz,CDCl3)δ8.12(s,1H),7.95(d,J=8.4Hz,1H),7.71(d,J=8.4Hz,1H),7.09–7.03(m,3H),6.99–6.94(m,1H),6.84(d,J=7.6Hz,1H),6.46(s,1H),5.81(s,1H),5.23(s,2H),5.1–5.04(m,2H),4.85–4.74(m,2H),4.54–4.39(m,3H),3.89(s,3H),3.51–3.48(m,2H),2.72–2.59(m,1H),2.57–2.40(m,2H),2.38–2.35(m,1H).
步骤2:(S)-2-((4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-氟苯基)-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((4-(2-((2,4-二氯苯并呋喃-7-基)甲氧基)-3-氟苯基)-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(90mg,0.14mmol)加入到1,4-二氧六环(1.2mL)和水(0.1mL),加入氢氧化锂(16mg,0.67mmol)置于40℃搅拌过夜。加入醋酸(0.1mL)调节PH至6~7。减压浓缩,用Prep-HPLC纯化(洗脱剂乙腈:水(v/v)=0~60%),得到白色固体产物35.2mg,产率40%。
LC-MS(ESI):[M+H]+=650.2;
1H NMR(400MHz,DMSO-d6)δ12.73(s,1H),8.27(s,1H),7.76(d,J=8.4Hz,1H),7.66(d,J=8.4Hz,1H),7.38–7.29(m,3H),7.17–7.0(m,3H),5.77(s,1H),5.30(s,2H),5.02–4.98(m,2H),4.82–4.76(m,2H),4.64–4.63(m,1H),4.47–4.45(m,1H),4.35–4.33(m,1H),3.46(t,J=7.0Hz,2H),2.74–2.55(m,3H),2.33–2.29(m,1H).
实施例43 2-((4-(2-(((4-氯-2-氟-1-苯并呋喃-7-基)甲基)氧基)吡啶-3-基)-6-氧代-1,2,3,6-四氢吡啶-1-基)甲基)-3-(((2S)-氧杂环-2-基)甲基)苯并[d]咪唑-5-羧酸(化合物43)的合成
步骤1:(4-氯苯并呋喃-7-基)乙酸甲酯的合成
将化合物7-(溴甲基)-4-氯苯并呋喃(0.50g,2.04mmol),醋酸钾(2.00g,20.37mmol)加入到N,N-二甲基甲酰胺(5mL)中,室温搅拌过夜。过滤溶液,减压浓缩,用乙酸乙酯萃取(20mL×3),无水硫酸钠干燥。减压浓缩,得到淡黄色固体产物粗品600mg,直接用于下一步反应。
LC-MS(ESI):[M+H]+=225.1。
步骤2:(4-氯苯并呋喃-7-基)甲醇的合成
将化合物(4-氯苯并呋喃-7-基)乙酸甲酯(600mg,2.67mmol),氢氧化锂(641mg,26.71mmol)加入到1,4-二氧六环(6mL)和水(2mL)中,40℃下反应2小时,冷却至室温,用乙酸乙酯萃取(20mL×3),无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂:PE/EA(v/v)=1:1),得到白色固体产物330mg,产率67.7%。
LC-MS(ESI):[M+1]+=183.1;
1HNMR(400MHz,DMSO-d6)δ8.15–8.10(m,1H),7.38–7.30(m,2H),7.04–6.98(m,1H),4.78(s,2H),3.38(s,1H).
步骤3:3-溴-2-((4-氯苯并呋喃-7-基)甲氧基)吡啶的合成
将化合物(4-氯苯并呋喃-7-基)甲醇(342mg,1.87mmol)加入到四氢呋喃(25mL)中,加入氢化钠(225mg,5.63mmol),搅拌10min后,加入3-溴-2-氟吡啶(330mg,1.88mmol),室温搅拌1小时,缓慢滴加水(5mL)淬灭,加入水(5mL),用乙酸乙酯(20mL×3)萃取,无水硫酸钠干燥,用硅胶柱层析纯化(洗脱剂:PE/EA(v/v)=10:1),得到白色固体产物500mg,产率78.8%。
LC-MS(ESI):[M+H]+=338.2;
1HNMR(400MHz,DMSO-d6)δ8.22–8.17(m,2H),8.10–8.06(m,1H),7.47–7.37(m,2H),7.08–7.06(m,1H),7.03–6.99(m,1H),5.71(m,2H).
步骤4:3-溴-2-((4-氯-2-氟-1-苯并呋喃-7-基)甲氧基)吡啶的合成
将化合物3-溴-2-((4-氯苯并呋喃-7-基)甲氧基)吡啶(400mg,1.18mmol),N-氟代双苯磺酰胺(447mg,1.42mmol)加入到四氢呋喃(10mL)中,-50℃下滴加二异丙基氨基锂(253mg,2.36mmol),搅拌过夜取。在-50℃下滴加水(5mL)淬灭反应,用乙酸乙酯萃取(20mL×3),无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂:PE/EA(v/v)=20:1),得到淡黄色固体产物100mg,产率23.7%。
LC-MS(ESI):[M+H]+=356.1;
1HNMR(400MHz,DMSO-d6)δ8.23–8.16(m,1H),8.12–8.05(m,1H),7.48–7.40(m,2H),7.04–6.98(m,1H),6.54(d,J=4.0Hz,1H),5.65(s,2H).
步骤5:4-甲氧基-5,6-二氢吡啶-2(1H)-酮的合成
将化合物哌啶-2,4-二酮(1g,8.84mmol),三甲氧基甲烷(1.88g,17.68mmol),一水对甲苯磺酸(0.42g,2.21mmol)加入到甲醇(10mL)中,70℃下搅拌5小时。冷却至室温,减压浓缩,用硅胶柱层析纯化(洗脱剂:DCM/MeOH(v/v)=20:1),得到白色固体产物0.6g,产率53.4%。
LC-MS(ESI):[M+H]+=128.2
1HNMR(400MHz,DMSO-d6)δ5.93(s,1H),5.00(s,1H),3.63(s,3H),3.36(t,J=4.0Hz,2H),2.39(t,J=8.0Hz,2H).
步骤6:(S)-2-((4-甲氧基-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物4-甲氧基-5,6-二氢吡啶-2(1H)-酮(86mg,0.68mmol),(S)-2-(氯甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(200mg,0.68mmol),加入到四氢呋喃(6mL)中,加入氢化钠(135mg,3.38mmol),室温下反应2小时。加入水(5mL),用乙酸乙酯萃取(20mL×3),无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂:DCM/MeOH(v/v)=20:1),得到淡黄色固体产物250mg,产率95.9%。
LC-MS(ESI):[M+H]+=386.2;
1HNMR(400MHz,DMSO-d6)δ8.28–8.26(m,1H),7.84–7.80(m,1H),7.70–7.66(m,1H),5.08(s,1H),5.04–4.95(m,2H),4.81–4.70(m,2H),4.63–4.55(m,1H),4.50–4.42(m,1H),4.37–4.30(m,1H),3.87(s,3H),3.66(s,3H),3.55(t,J=8.0Hz,2H),2.74–2.64(m,1H),2.53–2.43(m,2H),2.38–2.28(m,1H).
步骤7:(S)-2-((2,4-二氧代哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-((4-甲氧基-6-氧代-3,6-二氢吡啶-1(2H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(250mg,0.65mmol),加入到四氢呋喃(2mL)中,加入三氟乙酸(2mL,26.93mmol),室温搅拌过夜,冰浴下滴加饱和碳酸氢钠溶液至溶液为弱碱性,用二氯甲烷萃取(20mL×3),无水硫酸钠干燥,减压浓缩,用硅胶柱层析纯化(洗脱剂:DCM/MeOH(v/v)=20:1),得到白色固体产物100mg,产率41.5%。
LC-MS(ESI):[M+H]+=372.4;
1HNMR(400MHz,DMSO-d6)δ8.30–8.26(m,1H),7.84–7.80(m,1H),7.73–7.66(m,1H),5.11–4.89(m,3H),4.81–4.70(m,1H),4.63–4.55(m,1H),4.50–4.42(m,1H),4.37–4.30(m,1H),3.88(s,3H),3.73(t,J=4.0Hz,1H),3.55–3.47(m,1H),3.46–3.36(m,2H),2.76–2.64(m,1H),2.59(t,J=4.0Hz,1H),2.45–2.27(m,2H).
步骤8:(S)-1-(氧杂环丁烷-2-基甲基)-2-((6-氧代-4-(((三氟甲基)磺酰基)氧基)-3,6-二氢吡啶-1(2H)-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-2-((2,4-二氧代哌啶-1-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(100mg,0.27mmol),N-苯基双(三氟甲烷磺酰)亚胺(115mg,0.32mmol),三乙胺(0.1mL,0.54mmol)加入到四氢呋喃(3mL)中。室温下反应5小时,减压浓缩,用硅胶柱层析纯化(洗脱剂:DCM/MeOH(v/v)=20:1),得到浅白色固体产物100mg,产率73.8%。
LC-MS(ESI):[M+H]+=504.3。
步骤9:(S)-1-(氧杂环丁烷-2-基甲基)-2-((6-氧代-4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-3,6-二氢吡啶-1(2H)-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
将化合物(S)-1-(氧杂环丁烷-2-基甲基)-2-((6-氧代-4-(((三氟甲基)磺酰基)氧基)-3,6-二氢吡啶-1(2H)-基)甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(100mg,0.20mmol),联硼酸频那醇酯(56mg,0.22mmol),(1,1'-双(二苯基膦基)二茂铁)二氯化钯(7mg,0.01mmol),醋酸钾(39mg,0.40mmol)加入到1,4-二氧六环(5mL)中,用氮气置换三次,于70℃下反应1.5小时,直接投入下一步反应。
步骤10:(S)-2-((2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-6'-氧代-3',6'-二氢-[3,4'-联吡啶]-1'(2'H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯的合成
往步骤9的反应液中加入3-溴-2-(((4-氯-2-氟-1-苯并呋喃-7-基)甲基)氧)吡啶(71mg,0.20mmol),四三苯基膦钯(11mg,0.01mmol),碳酸钠(42mg,0.40mmol)和水(1mL),用氮气置换三次,于85℃下 反应3小时。过滤溶液,减压浓缩,用硅胶柱层析纯化(洗脱剂:DCM/MeOH(v/v)=20:1),得到淡黄色固体产物50mg,产率39.5%。
LC-MS(ESI):[M+H]+=631.4。
步骤11:(S)-2-((2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-6'-氧代-3',6'-二氢-[3,4'-联吡啶]-1'(2'H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸的合成
将化合物(S)-2-((2-((4-氯-2-氟苯并呋喃-7-基)甲氧基)-6'-氧代-3',6'-二氢-[3,4'-联吡啶]-1'(2'H)-基)甲基)-1-(氧杂环丁烷-2-基甲基)-1H-苯并[d]咪唑-6-羧酸甲酯(50mg,0.08mmol),氢氧化锂(20mg,0.48mmol)加入到1,4-二氧六环(2mL)和水(0.5mL)中,40℃下反应2小时。冷却至室温,缓慢滴加醋酸至溶液pH=6,减压浓缩,用Prep-HPLC纯化(洗脱剂:ACN/H2O(v/v)=50%),得到白色固体产物17.7mg,产率36.2%。
LC-MS(ESI):[M+H]+=617.2;
1HNMR(400MHz,DMSO-d6)δ12.80(s,1H),8.28–8.19(m,2H),7.88–7.78(m,2H),7.64(d,J=8.0Hz,1H),7.45–7.37(m,2H),7.16–7.10(m,1H),6.51(d,J=8.0Hz,1H),6.23(s,1H),5.65(s,2H),5.04–4.96(m,2H),4.89–4.82(m,1H),4.77–4.69(m,1H),4.61–4.54(m,1H),4.45–4.38(m,1H),4.34–4.26(m,1H),3.59(t,J=8.0Hz,2H),2.74(t,J=8.0Hz,2H),2.70–2.59(m,1H),2.35–2.24(m,1H).
实验例1体外细胞活性测试
1.材料
(1)细胞株
该细胞由上海药明康德新药开发有限公司构建,见下表1。
表1
(2)试剂见下表2。
表2
(3)仪器见下表3。
表3

(4)化合物信息
用DMSO将化合物配制为100μM或10μM的工作浓度化合物溶液。做10个点4倍稀释(稀释后最高浓度为1000nM或100nM),自动移液工作站完成稀释。
2.方法
(1)实验材料
实验缓冲液见下表4。
表4
检测试剂制备的终浓度见下表5。
表5
(2)实验方法
(a)配制化合物板:
待测化合物做10个点4倍稀释,起始浓度为100μM,自动移液工作站完成稀释
(b)转移化合物:
(i)使用液体处理仪转移100nL 100x化合物至384孔板。
(ii)将384孔板在1000rpm离心5秒。
(c)细胞悬液的制备
(i)将一支高表达hGLP-1R的HEK293细胞冻存管迅速置于37℃温水中解冻。
(ii)将细胞悬液转移至15mL离心管中,用10mL HBSS轻柔冲洗。
(iii)将离心管在1000rpm室温离心1分钟。
(iv)弃去上清。
(v)再用10ml HBSS轻柔冲洗,离心沉降细胞,最后用实验缓冲液重悬细胞。
(vi)利用细胞计数仪测量细胞密度与活度。
(vii)用实验缓冲液将GLP-1R细胞浓度稀释至1.0*105/mL。
(viii)在384孔板中转入10μL稀释好的细胞悬液。
(ix)室温孵育30分钟。
(d)检测:
(i)在384孔板空孔中加入10μL 800nM梯度稀释好的cAMP标准品。
(ii)加入10μL cAMP检测试剂。
(iii)室温孵育60分钟后,在酶标仪读数。
结果参见表1,表1为本发明部分实施例提供的化合物的激动作用(EC50)。
表1.本发明部分实施例提供的化合物的激动作用实验结果
结果与讨论:本发明的化合物表现出较优的对GLP-1受体的激动能力。
实验例2:本发明化合物小鼠药代动力学研究
1、实验材料
C57BL/6小鼠:雄性,6-8周龄,体重20-30克,购自维通利华(北京)实验动物科技有限公司。
试剂:色谱级乙腈购自于Thermo Fisher Scientific公司,色谱级甲酸购自于迪科马公司,实验用水为超纯水,其余试剂均为市售分析纯。
仪器:AB LCMS-5500串联质谱仪
2、实验方法
称取化合物溶于10%DMSO/5%Kolliphor-EL/85%HP-β-CD(20%)或其它适当体系中,制剂均呈澄清溶液,小鼠静脉或灌胃给药,于给药后5min、15min、30min、1h、2h、4h、6h、8h、24h采集血样。将每个PK采样点采全血30μL至EDTA-K2抗凝采血管中30min内于4℃离心取血浆。离心前全血样品置于湿冰上。所有采集的血浆样品保存在干冰上或冷冻保存中直至分析检测。
称取约1mg的化合物溶解于DMSO中,涡旋并超声,获得1mg/mL标准储备溶液。将标准储备溶液用50%乙腈水溶液稀释,得到浓度为5、10、20、50、100、500、1000、5000和10000ng/mL的标准工作溶液。用相同的稀释方法制备浓度为10、20、500和8000ng/mL的质控工作液。3μL浓度为(5、10、20、50、100、500、1000、5000和10000ng/mL)的标准工作液加入30μL空白C57BL/6小鼠血浆中,获得总体积为33μL,浓度为0.5-1000ng/mL(0.5、1、2、5、10、50、100、500、1000ng/mL)的标准曲线样品。浓度为(1ng/ml(低-1)和2ng/ml(低-2)、50ng/ml(中)、800ng/ml(高))的质控样品另外配置。
33μL标准样品、33μL质控样品或33μL未知样品(30μL血浆和3μL空白溶液)加入200μL含内标 (***)的乙腈沉淀蛋白。然后样品涡旋30秒,在4000g,4℃下离心15分钟后,取上清用水稀释3倍,10μL上清稀释液注入LC-MS/MS***进行定量分析,检测条件如下:
色谱柱:Raptor Biphenyl 2.7μm 2.1×50mm
流动相:溶液A:100%水(0.1%甲酸);溶液B:95%乙腈(0.1%甲酸,5%水),按下表进行梯度洗脱。
3、数据处理
使用Phoenix TM8.3软件的非房室模型进行药代动力学数据分析。
结果参见表2,表2为小鼠的药代动力学参数。
表2.本发明部分实施例提供的化合物的药代动力学参数实验结果
结果与讨论:本实验同时测定了CN114591308A中公开的化合物3的药代动力学性质,实验结果表明,本发明化合物在小鼠口服吸收较好,具有较高的暴露量且明显高于CN114591308A中公开的化合物3。
实验例3:大鼠药代动力学研究
3、实验材料
SD大鼠:雄性,6-8周龄,体重200-300克,购自斯贝福(北京)实验动物科技有限公司。
试剂:色谱级乙腈购自于Thermo Fisher Scientific公司,色谱级甲酸购自于迪科马公司,实验用水为超纯水,其余试剂均为市售分析纯。
仪器:AB LCMS-5500串联质谱仪
2、实验方法
称取化合物溶于10%DMSO/5%Kolliphor-EL/85%HP-β-CD(20%)或其它适当体系中,制剂均呈澄清溶液,大鼠静脉或灌胃给药,于给药后5min、15min、30min、1h、2h、4h、6h、8h、24h采集血样。将每个PK采样点采全血200μL至EDTA-K2抗凝采血管中30min内于4℃离心取血浆。离心前全血样品置于湿冰上。所有采集的血浆样品保存在干冰上或冷冻保存中直至分析检测。
称取约1mg的本申请化合物溶解于DMSO中,涡旋并超声,获得1mg/mL标准储备溶液。将标准储备溶液用50%乙腈水溶液稀释,得到浓度为5、10、20、50、100、500、1000、5000和10000ng/mL的标准工作溶液。用相同的稀释方法制备浓度为10、20、500和8000ng/mL的质控工作液。这些质控样品在分析当天以与标准曲线样品相同的方法制备。5μL浓度为(5、10、20、50、100、500、1000、5000和10000ng/mL)的标准工作液加入50μL空白SD大鼠血浆中,获得总体积为55μL,浓度为0.5-1000ng/mL(0.5、1、2、5、10、50、100、500、1000ng/mL)的标准曲线样品。浓度为(1ng/ml(低-1)和2ng/ml(低-2)、 50ng/ml(中)、800ng/ml(高))的质控样品另外配置。
55μL标准样品,55μL QC样品或55μL未知样本(50μL血浆样品加入5μL 50%乙腈水溶液)加入200μL含内标(***)的乙腈沉淀蛋白。然后样品涡旋30秒,在4000g,4℃下离心15分钟后,取上清用水稀释3倍,5μL上清稀释液注入LC-MS/MS***进行定量分析,检测条件如下:
色谱柱:HALO 90A Phenly-Hexyl,2μm 2.1×30mm
流动相:溶液A:100%水(0.1%甲酸);溶液B:95%乙腈(0.1%甲酸,5%水),按下表进行梯度洗脱。
3、数据处理
使用Phoenix TM8.3软件的非房室模型进行药代动力学数据分析。
结果参见表3,表3为大鼠的药代动力学参数。
表3.本发明部分实施例提供的化合物的药代动力学参数实验结果
结果与讨论:本实验同时测定了CN114591308A中公开的化合物3的药代动力学性质,实验结果表明,本发明化合物在大鼠口服吸收较好,具有较高的暴露量且明显高于CN114591308A中公开的化合物3。
在本说明书的描述中,参考术语“一个实施例”、“一些实施例”、“示例”、“具体示例”、或“一些示例”等的描述意指结合该实施例或示例描述的具体特征、结构、材料或者特点包含于本发明的至少一个实施例或示例中。在本说明书中,对上述术语的示意性表述不必须针对的是相同的实施例或示例。而且,描述的具体特征、结构、材料或者特点可以在任一个或多个实施例或示例中以合适的方式结合。此外,在不相互矛盾的情况下,本领域的技术人员可以将本说明书中描述的不同实施例或示例以及不同实施例或示例的特征进行结合和组合。

Claims (12)

  1. 一种化合物,其为如式(I)所示的化合物,或式(I)所示的化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或前药,
    其中:
    X1、X2和X3各自独立地为N或CR13
    Y1为N或CR14
    Y2为N或CR4
    Y3为N或CR5
    Y4为N或CR15
    L为O或CR16R17
    Z为O或S;
    R1为H、C1-6烷基、C2-6烯基、C2-6炔基、C3-6环烷基、3-6元杂环基或-C1-6亚烷基-R18,所述C1-6烷基、C2-6烯基、C2-6炔基、C3-6环烷基、3-6元杂环基和-C1-6亚烷基-R18可独立任选地被1、2或3个R1a取代;
    R18为C3-6环烷基、3-8元杂环基和5-10元杂芳基;
    R1a为D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷氨基、C3-6环烷基或3-6元杂环基,所述C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷氨基、C3-6环烷基和3-6元杂环基可独立任选地被1、2或3个R1b取代;
    R1b为D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、C1-6烷基、C1-6烷氧基、C1-6烷硫基、C1-6烷氨基、C3-6环烷基或3-6元杂环基;
    各R13独立地为H、D、F、Cl、Br、I、CN、羟基、氨基、硝基、C1-6烷基、C1-6卤代烷基、C1-6羟基烷基、C1-6氨基烷基、C1-6烷氧基、C1-6烷氨基、C1-6卤代烷氧基或-C1-6亚烷基-C1-6烷氧基;
    R2、R3、R6、R7、R16和R17各自独立地为H、D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基或C1-6烷基,所述C1-6烷基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
    环B为5-6元杂环基、C5-6环烷基、苯基或5-6元杂芳基;
    各Rb独立地为H、D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、C1-6烷基、C1-6烷氧基或C1-6烷氨基,所述C1-6烷基、C1-6烷氧基和C1-6烷氨基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
    R4、R5、R8、R9、R10、R11、R12、R14和R15各自独立地为H、D、F、Cl、Br、I、CN、羟基、硝基、C1-6烷基、C1-6烷氧基或C1-6烷氨基,所述C1-6烷基、C1-6烷氧基和C1-6烷氨基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
    n为0、1、2、3、4、5、6、7或8。
  2. 根据权利要求1所示的化合物,其具有式(II)、式(III)、式(IV)、式(V)、式(VI)或式(VII)所示结构:

  3. 根据权利要求1或2所述的化合物,其中R1为H、C1-3烷基、C2-3烯基、C2-3炔基、C3-6环烷基、3-6元杂环基或-C1-3亚烷基-R18,所述C1-3烷基、C2-3烯基、C2-3炔基、C3-6环烷基、3-6元杂环基和-C1-3亚烷基-R18可独立任选地被1、2或3个R1a取代;
    R18为C3-6环烷基、3-6元杂环基或5-10元杂芳基;
    R1a为D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、C1-3烷基、C1-3烷氧基、C1-3烷硫基、C1-3烷氨基、C3-6环烷基或5-6元杂环基,所述C1-3烷基、C1-3烷氧基、C1-3烷硫基、C1-3烷氨基、C3-6环烷基和5-6元杂环基可独立任选地被1、2或3个R1b取代;
    R1b为D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、C1-3烷基、C1-3烷氧基、C1-3烷硫基、C1-3烷氨基、C5-6环烷基或5-6元杂环基。
  4. 根据权利要求1-3任意一项所述的化合物,其中R1为H、甲基、乙基、正丙基、异丙基、乙烯基、烯丙基、乙炔基、炔丙基、1-丙炔基、环丙基、环丁基、环戊基、环己基、氧杂环丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基、吗啉基或-CH2R18,所述甲基、乙基、正丙基、异丙基、乙烯基、烯丙基、乙炔基、炔丙基、1-丙炔基、环丙基、环丁基、环戊基、环己基、氧杂环丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基、吗啉基和-CH2R18可独立任选地被1、2或3个R1a取代;
    R18为环丙基、环丁基、环戊基、环己基、氧杂环丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基、吗啉基、
    R1a为D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、甲硫基、N-甲氨基、N-乙氨基、环丙基、环丁基、环戊基、环己基、氧杂环丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基或吗啉基,所述甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、甲硫基、N-甲氨基、N-乙氨基、环丙基、环丁基、环戊基、环己基、氧杂环丁基、氮杂环丁基、吡咯烷基、四氢呋喃基、哌啶基、哌嗪基和吗啉基可独立任选地被1、2或3个R1b取代;
    R1b为D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、甲硫基、N-甲氨基、N-乙氨基、环丙基、环丁基、环戊基、环己基、氧杂环丁基、氮杂环丁基、 吡咯烷基、四氢呋喃基、哌啶基、哌嗪基或吗啉基。
  5. 根据权利要求1-4任意一项所述的化合物,其中R1为H、
  6. 根据权利要求1-5任意一项所述的化合物,其中各R13独立地为H、D、F、Cl、Br、I、CN、羟基、氨基、硝基、C1-6烷基、C1-6卤代烷基、C1-6羟基烷基、C1-6氨基烷基、C1-6烷氧基、C1-6烷氨基、C1-6卤代烷氧基或-C1-6亚烷基-C1-6烷氧基;
    R2、R3、R6、R7、R16和R17各自独立地为H、D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基或C1-3烷基,所述C1-3烷基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
    各Rb独立地为H、D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、C1-3烷基、C1-3烷氧基或C1-3烷氨基,所述C1-3烷基、C1-3烷氧基和C1-3烷氨基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
    R4、R5、R8、R9、R10、R11、R12、R14和R15各自独立地为H、D、F、Cl、Br、I、CN、羟基、硝基、C1-3烷基、C1-3烷氧基或C1-3烷氨基,所述C1-3烷基、C1-3烷氧基和C1-3烷氨基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代。
  7. 根据权利要求1-6任意一项所述的化合物,其中各R13独立地为H、D、F、Cl、Br、I、CN、羟基、氨基、硝基、甲基、乙基、正丙基、异丙基、-CHF2、-CF3、-CHFCH2F、-CF2CHF2、-CH2CF3、-CH2CF2CHF2、羟基甲基、羟基乙基、氨甲基、氨乙基、甲氧基、乙氧基、正丙氧基、异丙氧基、N-甲氨基、N-乙氨基、-OCHF2、-OCF3、-OCHFCH2F、-OCF2CHF2、-OCH2CF3、-OCH2CF2CHF2、-CH2OCH3、-CH2OCH2CH3、-CH2CH2OCH3或-CH2CH2OCH2CH3
    R2、R3、R6、R7、R16和R17各自独立地为H、D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、甲基、乙基、正丙基或异丙基,所述甲基、乙基、正丙基和异丙基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
    各Rb独立地为H、D、F、Cl、Br、I、CN、羟基、氧代、氨基、硝基、甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、N-甲氨基或N-乙氨基,所述甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、N-甲氨基和N-乙氨基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代;
    R4、R5、R8、R9、R10、R11、R12、R14和R15各自独立地为H、D、F、Cl、Br、I、CN、羟基、硝基、甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、N-甲氨基或N-乙氨基,所述甲基、乙基、正丙基、异丙基、甲氧基、乙氧基、正丙氧基、异丙氧基、N-甲氨基和N-乙氨基可独立任选地被1、2或3个F、Cl、Br、I、CN、羟基、氨基或硝基取代。
  8. 根据权利要求1-7任意一项所述的化合物,其为具有下列之一结构的化合物或具有下列之一结构的化合物的立体异构体、几何异构体、互变异构体、氮氧化物、水合物、溶剂化物、代谢产物、药学上可接受的盐或它的前药:


  9. 一种药物组合物,包含权利要求1-8任意一项所述的化合物;所述药物组合物任选地进一步包含药学上可接受的赋形剂、载体、佐剂或它们的任意组合。
  10. 权利要求1-8任意一项所述的化合物或者权利要求9所述的药物组合物在制备药物中的用途,所述药物用于预防、治疗或减轻患者GLP-1受体激动剂介导的疾病。
  11. 根据权利要求10所述的用途,其中,所述GLP-1受体激动剂介导的疾病为糖尿病、非酒精性脂肪肝病或肥胖。
  12. 根据权利要求11所述的用途,其中,所述糖尿病为I型糖尿病、II型糖尿病、妊娠糖尿病、特发性I型糖尿病、早发型II型糖尿病、青年人的成年型糖尿病、青少年发作的非典型糖尿病、营养不良相关性糖尿病或成人隐匿性自身免疫性糖尿病。
PCT/CN2023/115679 2022-08-30 2023-08-30 苯并双环类化合物及其制备方法和应用 WO2024046342A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211048114 2022-08-30
CN202211048114.4 2022-08-30

Publications (1)

Publication Number Publication Date
WO2024046342A1 true WO2024046342A1 (zh) 2024-03-07

Family

ID=90100378

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/115679 WO2024046342A1 (zh) 2022-08-30 2023-08-30 苯并双环类化合物及其制备方法和应用

Country Status (1)

Country Link
WO (1) WO2024046342A1 (zh)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113480534A (zh) * 2021-07-23 2021-10-08 广州必贝特医药技术有限公司 苯并咪唑或氮杂苯并咪唑-6-羧酸类化合物及其应用
CN114591308A (zh) * 2020-12-03 2022-06-07 苏州闻泰医药科技有限公司 一类glp-1r受体激动剂化合物及其用途
CN114630823A (zh) * 2019-10-25 2022-06-14 吉利德科学公司 Glp-1r调节化合物
CN114716423A (zh) * 2022-05-20 2022-07-08 中国科学院上海药物研究所 5,6-二氢-1,2,4-三嗪类化合物及其作为glp-1受体激动剂的药学用途
CN116120296A (zh) * 2021-11-12 2023-05-16 四川科伦博泰生物医药股份有限公司 一种多环化合物、包含其的药物组合物、其制备方法及其用途
CN116574092A (zh) * 2022-05-20 2023-08-11 成都地奥九泓制药厂 苯并咪唑或氮杂苯并咪唑类化合物、其制备方法及其应用

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114630823A (zh) * 2019-10-25 2022-06-14 吉利德科学公司 Glp-1r调节化合物
CN114591308A (zh) * 2020-12-03 2022-06-07 苏州闻泰医药科技有限公司 一类glp-1r受体激动剂化合物及其用途
CN113480534A (zh) * 2021-07-23 2021-10-08 广州必贝特医药技术有限公司 苯并咪唑或氮杂苯并咪唑-6-羧酸类化合物及其应用
CN116120296A (zh) * 2021-11-12 2023-05-16 四川科伦博泰生物医药股份有限公司 一种多环化合物、包含其的药物组合物、其制备方法及其用途
CN114716423A (zh) * 2022-05-20 2022-07-08 中国科学院上海药物研究所 5,6-二氢-1,2,4-三嗪类化合物及其作为glp-1受体激动剂的药学用途
CN116574092A (zh) * 2022-05-20 2023-08-11 成都地奥九泓制药厂 苯并咪唑或氮杂苯并咪唑类化合物、其制备方法及其应用

Similar Documents

Publication Publication Date Title
JP7248665B2 (ja) 化学化合物
TWI431001B (zh) 作為圓滑蛋白(smo)抑制劑的有機化合物
KR20210042265A (ko) 헌팅턴병 치료를 위한 헤테로사이클릭 및 헤테로아릴 화합물
CN108430998A (zh) 氮杂双环衍生物及其制备方法和用途
CN105384687B (zh) 喹啉酮类化合物及其在药物中应用
TWI576346B (zh) 雙環吡嗪酮衍生物
JP6688372B2 (ja) キヌレニン−3−モノオキシゲナーゼ阻害薬、医薬組成物、及びこれらの使用方法
CN114728962A (zh) 血浆激肽释放酶抑制剂及其用途
WO2011097079A1 (en) Apoptosis signal-regulating kinase 1 inhibitors
CN101951775A (zh) 四氢噻吩并吡啶
WO2022228399A1 (zh) 三并环类usp1抑制剂及其用途
WO2021032004A1 (zh) 氮杂芳基化合物及其应用
WO2022237676A1 (zh) Shp2磷酸酶抑制剂的制备及其应用
KR20220038696A (ko) 폴리방향족 우레아 유도체 및 근육 질환 치료에서의 이들의 용도
WO2023217230A1 (zh) 驱动蛋白kif18a抑制剂及其应用
WO2020078402A1 (zh) 作为TGF-βR1抑制剂的化合物及其应用
WO2024046342A1 (zh) 苯并双环类化合物及其制备方法和应用
WO2023274396A1 (zh) 苯并氮杂环类化合物及其在药物中的应用
EP4029501A1 (en) Combination of polyaromatic urea derivatives and glucocorticoid or hdac inhibitor for the treatment of diseases or conditions associated with muscle cells and/or satellite cells
WO2024041609A1 (zh) 苯并双环类化合物及其制备方法和应用
WO2023169436A1 (zh) 苯并双环类化合物及其制备方法和应用
WO2024061366A1 (zh) 具有磷酰化芳基结构的小分子化合物及其应用
WO2023109883A1 (zh) 一类芳杂环取代的化合物及其制备方法和用途
WO2024032661A1 (zh) Kif18a抑制剂及其用途
WO2023134713A1 (zh) 氮杂并环衍生物、其药物组合物及其用途

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: 23859371

Country of ref document: EP

Kind code of ref document: A1