WO2023143194A1 - Ccr4 small molecule antagonist and use thereof - Google Patents

Ccr4 small molecule antagonist and use thereof Download PDF

Info

Publication number
WO2023143194A1
WO2023143194A1 PCT/CN2023/072403 CN2023072403W WO2023143194A1 WO 2023143194 A1 WO2023143194 A1 WO 2023143194A1 CN 2023072403 W CN2023072403 W CN 2023072403W WO 2023143194 A1 WO2023143194 A1 WO 2023143194A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
room temperature
isomer
formula
Prior art date
Application number
PCT/CN2023/072403
Other languages
French (fr)
Chinese (zh)
Inventor
陈鑫德
柳梦林
宋云鹏
郝欣
Original Assignee
瑞石生物医药有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 瑞石生物医药有限公司 filed Critical 瑞石生物医药有限公司
Priority to CN202380016094.9A priority Critical patent/CN118475581A/en
Publication of WO2023143194A1 publication Critical patent/WO2023143194A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the invention belongs to the field of medicine, and in particular relates to a CCR4 small molecule antagonist and its application.
  • CCR4 (C-C chemokine receptor type 4) belongs to G protein-coupled receptors and is a member of the chemokine receptor family. It is widely expressed on the surface of immune cells, especially Th2 cells and Treg cells.
  • the main endogenous ligands of CCR4 include CCL22 (also known as macrophage-derived chemokine, MDC) and CCL17 (also known as thymus activation-regulated chemokine, TARC).
  • CCL22 also known as macrophage-derived chemokine, MDC
  • CCL17 also known as thymus activation-regulated chemokine, TARC.
  • the combination of CCR4 and endogenous ligand can activate the coupled G protein, and then a cascade of cell activation effects occurs. Through a series of information transmission, chemotactic target cells migrate to a specific location and exert biological effects.
  • CCR4 is closely related to the occurrence and development of immune-related diseases such as atopic dermatitis, asthma, allergic rhinitis, atopic dermatitis, systemic lupus erythematosus and rheumatoid arthritis.
  • CCR4 plays a regulatory role in the tumor microenvironment, inducing tumor immune escape and promoting tumor cell metastasis. Therefore, CCR4 has become an important drug target for the treatment of immune-related diseases and tumor immunotherapy, and the development of CCR4 small molecule antagonists will provide new options for the treatment of the above diseases.
  • Mogamulizumab a CCR4 monoclonal antibody
  • Mogamulizumab a CCR4 monoclonal antibody
  • WO2018/022992 involves FLX475, which is currently the fastest-growing small-molecule antagonist of CCR4 in clinical practice.
  • RPT193 is another small-molecule antagonist of CCR4 in clinical research.
  • most small-molecule antagonists of CCR4 are in the stage of biological activity testing. It can be seen that the development of small molecule antagonists of CCR4 has a good application prospect.
  • the purpose of the present invention is to provide a new type of CCR4 small molecule antagonist, this type of compound has good activity in inhibiting CCR4, and exhibits excellent effects and functions.
  • the present invention provides a compound of formula (I) or an isotopically labeled compound thereof, or an optical isomer, geometric isomer, tautomer or isomer mixture thereof, or a pharmaceutically acceptable salts, or their prodrugs, or their metabolites,
  • X 1 and X 2 are each independently selected from C or N;
  • X 3 , X 4 and X 5 are each independently selected from CR b or N;
  • Each R 1 is independently selected from H, halogen, C 1 -C 3 alkylsulfonyl, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, C 1 -C 3 alkoxy, C 3 -C 4 cycloalkyl or cyano;
  • R 2 is selected from H, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl or C 3 -C 4 cycloalkyl;
  • R 3 is selected from H, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl or C 3 -C 4 cycloalkyl;
  • R4 is selected from
  • Y is selected from CH2 or O
  • R a is selected from H, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 3 -C 6 cycloalkyl, 4-7 membered heterocyclyl, 5-6 membered heteroaryl, phenyl or cyano, wherein said C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 6 cycloalkyl, 4-7 membered heterocyclic , 5-6 membered heteroaryl or phenyl are each independently optionally selected from one or more groups selected from halogen, hydroxyl, cyano, C 1 -C 3 alkyl or C 1 -C 3 alkoxy replaced by
  • Each R b is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyl or cyano, wherein the C 1 -C 3 alk radical, C 1 -C 3 alkoxy or C 3 -C 6 cycloalkyl each independently optionally selected from halogen, hydroxy, amino, cyano, C 1 -C 3 alkyl or C 1 -C 3 One or more groups of alkoxy are substituted;
  • Each R c is independently selected from H, halogen, C 1 -C 3 alkyl or C 3 -C 4 cycloalkyl, wherein each of the C 1 -C 3 alkyl or C 3 -C 4 cycloalkyl is independently optionally substituted by one or more groups selected from halogen, hydroxyl, cyano, C 1 -C 3 alkyl or C 1 -C 3 alkoxy;
  • Each R d is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyl or cyano, wherein the C 1 -C 3 alk radical, C 1 -C 3 alkoxy or C 3 -C 6 cycloalkyl each independently optionally selected from halogen, hydroxy, amino, cyano, C 1 -C 3 alkyl or C 1 -C 3 One or more groups of alkoxy are substituted;
  • each Re is independently -LR f ;
  • Each R f is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 1 -C 3 haloalkyl, C 3 -C 6 cycloalkyl, 4-7 membered Heterocyclyl, R g C(O)NH-, R g NHC(O)-, R g OC(O)NH-, R g S(O) 2 NH-, R g NHS(O) 2 -, R g g S(O) 2 -, -(CH 2 ) n -hydroxyl, -(CH 2 ) n -amino, -(CH 2 ) n -cyano, -(CH 2 ) n -carboxy or 2-oxa- Spiro[3,3]heptyl, wherein the C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyl, 4-7 membere
  • Each R g is independently selected from C 1 -C 3 alkyl, C 3 -C 6 cycloalkyl or 3-6 membered heterocyclyl;
  • Each L is independently selected from a chemical bond, a 4-7 membered heterocyclic group, a C 1 -C 3 alkylene group or a C 3 -C 6 cycloalkylene group, wherein the 4-7 membered heterocyclic group, C 1 -C 3 alkylene or C 3 -C 6 cycloalkylene are each independently optionally selected Substituted by one or more groups from halogen, hydroxyl, amino, cyano, C 1 -C 3 alkyl or C 1 -C 3 alkoxy;
  • Z 1 is any integer from 0 to 3;
  • Z 2 is any integer from 0 to 3;
  • Z 3 is any integer from 0 to 4.
  • Z 4 is any integer from 0 to 2;
  • n is independently any integer from 0 to 3.
  • One of X1 and X2 is N, the other is C,
  • At least one of X 3 , X 4 and X 5 is N.
  • the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt , or its prodrug, or its metabolite is a compound of formula (II) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical acceptable salts, or prodrugs thereof, or metabolites thereof,
  • R 1 -R 3 , X 1 -X 5 , Y, R a , R c , R d , R e , Z 1 , Z 2 , Z 3 and Z 4 are as defined in formula (I);
  • R c , R d , Re , Z 2 and Z 3 are as defined in formula (I);
  • R 1 -R 3 , R a , R d , R e , Z 1 and Z 3 are as defined in formula (I).
  • the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt , or its prodrug, or its metabolite is a compound of formula (III) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical acceptable salts, or prodrugs thereof, or metabolites thereof,
  • R 1 -R 3 , R a , R e and Z 1 are as defined in formula (I).
  • the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt , or its prodrug, or its metabolite is a compound of formula (IV) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical acceptable salts, or prodrugs thereof, or metabolites thereof,
  • R 1 -R 3 , R a , R e and Z 1 are as defined in formula (I).
  • each Re is independently -LR f ;
  • Each R f is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 1 -C 3 haloalkyl, C 3 -C 6 cycloalkyl, 4-7 membered hetero Cyclo, R g C(O)NH-, R g NHC(O)-, R g OC(O)NH-, R g S(O) 2 NH-, R g NHS(O) 2 -, R g S(O) 2 -, -(CH 2 ) n -hydroxyl, -(CH 2 ) n -amino, -(CH 2 ) n -cyano, -(CH 2 ) n -carboxy or 2-oxa-spiro [3,3] Heptyl, wherein the C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyl, 4-7 membered heterocyclyl
  • R g is selected from C 1 -C 3 alkyl, C 3 -C 4 cycloalkyl or 3-4 membered heterocyclic group, preferably selected from methyl, ethyl, cyclopropyl, cyclobutanyl or oxa cyclobutanyl;
  • each n is independently any integer from 0 to 3
  • L is selected from the group consisting of chemical bond, methylene optionally substituted by methyl or ethyl, ethylene, propylene, cyclopropylene, cyclobutylene, cyclopentylene, cyclohexylene, azepine Cyclobutane, pyrrolidinylene or piperidinylene;
  • Each Re is independently selected from H, methyl, ethyl, n-propyl, isopropyl, cyclopropyl,
  • Each R 1 is independently selected from H, halogen, C 1 -C 3 alkylsulfonyl, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl or C 1 -C 3 alkoxy, preferably selected from H, fluorine, chlorine, bromine, methyl, ethyl, n-propyl, isopropyl or methanesulfonyl; and/or
  • R is selected from H or C 1 -C 3 alkyl, preferably from H or methyl;
  • R 3 is selected from H or C 1 -C 3 alkyl, preferably from H or methyl.
  • Each R a is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, C 1 -C 3 alkoxy, C 3 -C 4 cycloalkyl or cyano, wherein The C 1 -C 3 alkyl, C 1 -C 3 alkoxy or C 3 -C 4 cycloalkyl are each independently optionally replaced by halogen, hydroxyl, cyano, C 1 -C 3 alkyl or C 1 - C3alkoxy substitution, R is preferably selected from H, fluorine, chlorine, bromine, methyl, ethyl, isopropyl, cyclopropyl, difluoromethyl, trifluoromethyl, trifluoroethyl, Methoxy, ethoxy, hydroxymethyl, 1,1-dimethylhydroxymethyl, cyano, or methoxymethylene; and/or
  • Each R b is independently selected from H, halogen, C 1 -C 3 alkyl, C 3 -C 4 cycloalkyl or cyano, wherein the C 1 -C 3 alkyl or C 3 -C 4 cycloalkyl Each is independently optionally substituted by halogen, hydroxyl, amino, cyano or C 1 -C 3 alkyl, R b is preferably selected from H, fluorine, chlorine, bromine, methyl, ethyl, isopropyl, cyclopropyl group, difluoromethyl, trifluoromethyl, trifluoroethyl or cyano; and/or
  • Each R c is independently selected from H, halogen or C 1 -C 3 alkyl, preferably selected from H, fluorine, chlorine, methyl or ethyl; and/or
  • Each R d is independently selected from H, halogen, C 1 -C 3 alkyl or cyano, preferably selected from H, fluorine, chlorine, bromine, methyl, ethyl, n-propyl, isopropyl or cyano .
  • Z 1 is 2; and/or
  • Z2 is 0 or 1;
  • Z3 is 0 or 1;
  • Z 4 is 1; and/or
  • n is independently 0 or 1.
  • the "compound of formula (I), (II), (III), (IV)” or “compound of the present invention” described hereinafter may also cover formula (I), (II), (III), (IV) Any isotope-labeled compound of the compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its Metabolites.
  • optical isomer means that when a compound has one or more chiral centers, each chiral center can have an R configuration or an S configuration, and the various isomers thus constituted are optical isomers. Construct. Optical isomers include all diastereoisomers, enantiomers, mesoforms, racemates or mixtures thereof. For example, optical isomers can be separated by chiral chromatographic columns or by chiral synthesis.
  • Geometric isomer means that when a double bond exists in the compound, the compound may exist as a cis-isomer, a trans-isomer, an E-isomer and a Z-isomer. Geometric isomers include cis isomers, trans isomers, E isomers, Z isomers, or mixtures thereof.
  • tautomer refers to isomers that result from the rapid movement of an atom in a molecule between two positions. Those skilled in the art can understand that tautomers can transform into each other, and may reach an equilibrium state and coexist in a certain state.
  • references herein to "compounds of formula (I)" or “compounds of the present invention” also encompass isotopically labeled compounds in which any atom in the compound is replaced by its isotopic atom.
  • the present invention includes all pharmaceutically acceptable isotope-labeled compounds of compounds of formula (I) in which one or more atoms are replaced by atoms having the same atomic number but a different atomic mass or mass number as atoms normally found in nature. replace.
  • isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2 H(D) and 3 H(T), isotopes of carbon, such as 11 C, 13 C and 14 C, isotopes of chlorine, such as 36 Cl, isotopes of fluorine such as 18 F, isotopes of iodine such as 123 I and 125 I, isotopes of nitrogen such as 13 N and 15 N, isotopes of oxygen such as 15 O, 17 O and 18 O, and of sulfur Isotopes such as35S .
  • isotopes of hydrogen such as 2 H(D) and 3 H(T)
  • isotopes of carbon such as 11 C, 13 C and 14 C
  • isotopes of chlorine such as 36 Cl
  • isotopes of fluorine such as 18 F
  • isotopes of iodine such as 123 I and 125 I
  • isotopes of nitrogen such as 13
  • Isotope-labeled compounds of formula (I), (II), (III), (IV) can generally be prepared by conventional techniques known to those skilled in the art or by using a suitable isotope-labeled reagent instead of a previously used non-labeled reagent The preparation was carried out as described in the Examples and Preparations appended hereto.
  • Formula (I), (II), (III), (IV) compound can exist in the form of pharmaceutically acceptable salt, for example, the acid addition of formula (I), (II), (III), (IV) compound Salt formation and/or base addition salts.
  • pharmaceutically acceptable salt includes acid addition salts or base addition salts that may occur in compounds of formula (I), (II), (III), (IV).
  • the pharmaceutically acceptable salts of the compound of formula (I), (II), (III), (IV) include its acid plus Salt and base addition salt.
  • Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include, but are not limited to: acetate, adipate, aspartate, benzoate, benzenesulfonate, bicarbonate/carbonate, bisulfate/sulfate, borate , camphorsulfonate, citrate, cyclamate, ethanedisulfonate, formate, fumarate, glucoheptonate, gluconate, glucuronate, six Fluorophosphate, 2-(4-Hydroxybenzyl)benzoate, Hydrochloride/Chloride, Hydrobromide/Bromide, Hydroiodide/Iodide, 2-Isethionate, Lactate , malate, maleate, malonate, methanesulfonate, methylsulfate,
  • Suitable base addition salts are formed from bases which form non-toxic salts. Examples include, but are not limited to: aluminum, arginine, calcium, choline, diethylamine, diethanolamine, glycine, lysine, magnesium, meglumine, ethanolamine, potassium, sodium, tromethamine, and zinc salts. Half-salts of acids and bases, such as the hemisulfate and hemicalcium salts, may also be formed.
  • suitable salts see Handbook of Pharmaceutical Salts: Properties, Selection and Use by Stahl and Wermuth (Wiley-VCH, 2002). Methods for preparing pharmaceutically acceptable salts of the compounds described herein are known to those skilled in the art.
  • Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms.
  • compounds of formula (I), (II), (III), (IV) are included within the scope of the present invention whether they exist in solvated or unsolvated form.
  • Certain compounds of the present invention may exist in different crystalline or amorphous forms, and no matter in which form they exist, the compounds of formula (I), (II), (III) and (IV) are included within the scope of the present invention.
  • pharmaceutically acceptable means that the corresponding compound, carrier or molecule is suitable for administration to a human.
  • the term refers to the use in mammals, preferably humans, certified by any national regulatory agency such as CFDA (China), EMEA (Europe), FDA (USA), etc.
  • Prodrug refers to a derivative that is converted into the compound of the present invention by reacting with enzymes, gastric acid, etc. under physiological conditions in vivo, for example, by oxidation, reduction, hydrolysis, etc. catalyzed by enzymes.
  • Metal refers to all molecules derived from any compound of the invention in a cell or organism, preferably a human.
  • hydroxyl refers to -OH; the term “amino” refers to -NH2 ; the term “nitro” refers to -NO2 ; and the term “cyano” refers to -CN; the term “carboxy” refers to -COOH.
  • substituted means that one or more (preferably 1 to 5, more preferably 1 to 3, even more effectively 1 or 2) hydrogen atoms in a group are independently replaced by the corresponding number of substituents are substituted.
  • the term “optional” or “optionally” means that the event it describes can or cannot occur.
  • a group “optionally substituted” means that the group can be unsubstituted or substituted.
  • alkyl refers to saturated aliphatic hydrocarbons, including straight and branched chains.
  • an alkyl group has 1-8, or 1-6, or 1-3 carbon atoms.
  • C 1-8 alkyl refers to a straight-chain or branched chain radical having 1-8 carbon atoms
  • C 1-6 alkyl refers to a straight-chain or branched chain having 1-6 carbon atoms.
  • a branched chain radical the term “C 1-3 alkyl” refers to a straight chain or branched chain radical having 1 to 3 carbon atoms.
  • C 1-8 alkyl includes within its definition the terms “C 1-6 alkyl", “C 1 -C 3 alkyl” and "C 1 -C 4 alkyl”.
  • alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, 2-pentyl, 3-pentyl, iso Pentyl, neopentyl, (R)-2-methylbutyl, (S)-2-methylbutyl, 3-methylbutyl, 2,3-dimethylpropyl, 2,3- Dimethylbutyl, hexyl, etc.
  • Alkyl groups may optionally be substituted with one or more (eg, 1 to 5, preferably 1 to 3, even more effectively 1 or 2) suitable substituents.
  • haloalkyl refers to an alkyl group having one or more halogen substituents (up to perhaloalkyl, i.e., each hydrogen atom of the alkyl group is replaced by a halogen atom) .
  • C 1- C 6 haloalkyl refers to a C 1- C 6 alkyl group (up to perhaloalkyl, i.e., each hydrogen atom of the alkyl group has one or more halogen substituents replaced by halogen atoms).
  • C 1- C 4 haloalkyl refers to a C 1- C 4 alkyl group having one or more halogen substituents (up to perhaloalkyl, i.e., each hydrogen of the alkyl group atoms are all replaced by halogen atoms);
  • C 1- C 3 haloalkyl refers to a C 1- C 3 alkyl group (up to perhaloalkyl, i.e., alkyl each hydrogen atom of the group is replaced by a halogen atom);
  • C 1- C 2 haloalkyl refers to a C 1- C 2 alkyl group having one or more halogen substituents (i.e., methyl or ethyl) (up to perhaloalkyl, ie, each hydrogen atom of the alkyl group is replaced by a halogen atom).
  • C haloalkyl refers to a methyl group having 1, 2 or 3 halo substituents .
  • haloalkyl groups include: CF3 , C2F5 , CHF2 , CH2F , CH2CF3 , CH2Cl , and the like.
  • alkylene refers to a divalent alkyl group, wherein alkyl is as defined above.
  • the alkylene group is preferably an alkylene group having 1-6 carbon atoms (i.e. C 1- C 6 alkylene group), more preferably an alkylene group having 1-3 carbon atoms (i.e. C 1- C 3 alkylene group alkyl).
  • alkylene groups include, but are not limited to -CH2- , -CH( CH3 ) -, -C( CH3 ) 2- , -CH2CH2- , -CH( CH2CH3 )-, -CH 2 CH(CH 3 )-, -CH 2 C(CH 3 ) 2 -, -CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 -, etc.
  • n-membered cycloalkyl refers to an all-carbocyclic ring system having n carbon atoms.
  • C 3 -C 6 cycloalkyl refers to a fully carbocyclic ring system having 3-6 carbon atoms
  • C 3 -C 4 cycloalkyl refers to a fully carbocyclic ring system having 3-4 carbon atoms .
  • Examples of C 3 -C 6 cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl.
  • cycloalkyl groups may be optionally substituted with one or more suitable substituents.
  • cycloalkylene refers to a divalent cycloalkyl group, wherein cycloalkyl is as defined above.
  • n-membered heterocyclyl refers to a heterocycloalkyl group having m ring-forming carbon atoms and (nm) ring-forming heteroatoms selected from O, S, S(O) 2 and N.
  • 4-7 membered heterocyclic groups include, but are not limited to, oxetane, thietane, azetidine, tetrahydrofuran, tetrahydrothiophene, pyrrolidine, tetrahydropyran, tetrahydrothiopyran, Piperidine, morpholine, piperazine, oxepane, thiepane, azepane, 1,1-dioxothietane.
  • heterocyclyl may be optionally substituted with one or more suitable substituents.
  • heterocyclylene refers to a divalent heterocyclyl group, wherein heterocyclyl is as defined above.
  • n-membered heteroaryl refers to a heteroaryl group having m carbon atoms forming an aromatic ring and (n-m) heteroatoms forming an aromatic ring selected from O, S and N.
  • 5-6 membered heteroaryl groups include, but are not limited to, pyrazine, pyrazole, pyrrole, furan, thiophene, thiazole, pyridine.
  • heteroaryl groups may be optionally substituted with one or more suitable substituents.
  • the number ranges related to the number of substituents, the number of carbon atoms, and the number of ring atoms represent enumeration of all integers within the range one by one, and the range is only used as a simplified representation.
  • “1-4 substituents” means 1, 2, 3 or 4 substituents
  • "3-8 ring atoms” means 3, 4, 5, 6, 7 or 8 ring atoms . Therefore, the number ranges related to the number of substituents, the number of carbon atoms, and the number of ring atoms also cover any sub-range thereof, and each sub-range is also deemed to be disclosed herein.
  • the compounds of the present invention can be prepared in a variety of ways known to those skilled in the art of organic synthesis. Those skilled in the art can refer to the synthetic routes of the specific compounds in the specific examples of the present invention, and make appropriate adjustments to the reaction raw materials and reaction conditions to obtain the synthetic methods of other compounds.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I), (II), (III), (IV) or its isotope-labeled compound, or its optical isomers, geometric isomers isomer, tautomer or isomer mixture, or a pharmaceutically acceptable salt thereof, or a prodrug thereof, or a metabolite thereof, and a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers can be organic or inorganic inert carrier materials, for example, suitable carriers include water, gelatin, gum arabic, lactose, starch, magnesium stearate, talc, vegetable oils, polyalkylene glycols, petrolatum, Mannitol, cellulose, cellulose derivatives, sodium saccharin, glucose, sucrose, magnesium carbonate, saline, glycerin, ethanol, etc.
  • the pharmaceutical composition may also contain other pharmaceutical additives, such as flavoring agents, preservatives, stabilizers, emulsifiers, buffers, diluents, binders, wetting agents, disintegrants, lubricants, glidants and the like.
  • the dosage form of the pharmaceutical composition of the present invention may be a liquid dosage form, a solid dosage form or a semi-solid dosage form.
  • Liquid dosage forms can be solutions (including true solutions and colloid solutions), emulsions (including o/w type, w/o type and double emulsion), suspensions, injections (including aqueous injections, powder injections and infusion solutions), eye drops Agents, nasal drops, lotions and liniments, etc.
  • solid dosage forms can be tablets (including ordinary tablets, enteric-coated tablets, buccal tablets, dispersible tablets, chewable tablets, effervescent tablets, orally disintegrating tablets), capsules (including hard capsules, soft capsules, enteric-coated capsules), granules, powders, pills, suppositories, films, patches, aerosols, sprays, etc.
  • semi-solid dosage forms can be ointments, Gels, pastes, etc.
  • the pharmaceutical composition of the present invention can be made into common preparations, sustained
  • the dosage form of the pharmaceutical composition is selected from tablet, granule, powder, syrup, inhalation and injection.
  • Solid dosage forms for oral administration may include capsules, tablets, pills, powders and granules.
  • the active compound is mixed with at least one inert excipient (or carrier) (for example, sodium citrate or dicalcium phosphate), which may also include: (a) fillers or mixing agents (for example, starch, lactose, sucrose, glucose, mannitol, and silicic acid); (b) binders (e.g., carboxymethylcellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose, and gum arabic); (c) Moisturizers (eg, glycerol); (d) disintegrants (eg, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain synthetic silicates, sodium carbonate); (e) solutions Blocking agents (e.g., paraffin); (f) absorption enhancers (e.g., quaternary ammonium compounds); (g) we
  • Formulations suitable for parenteral administration may include aqueous and nonaqueous isotonic sterile solutions suitable for injection, as well as aqueous and nonaqueous sterile suspensions.
  • the parenteral formulations provided herein are optionally contained in unit-dose or multi-dose sealed containers, such as ampoules, and can be stored in freeze-dried (lyophilized) containers that require only the addition of a sterile liquid carrier (such as water for injection) immediately before use. ) conditions.
  • Suitable diluents for reconstitution of the pharmaceutical composition include bacteriostatic water for injection, 5% dextrose in water, phosphate buffered saline, Ringer's solution, saline, sterile water, Deionized water and combinations thereof.
  • Sprays can contain excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • Inhalants may contain excipients such as lactose, or aqueous solutions containing, for example, polyethylene oxide-9-lauryl ether, glycocholate and deoxycholate, or oily solutions as nasal drops or spray , or in gel form.
  • the content of the compound of the present invention in its pharmaceutical composition can be adjusted according to actual needs (such as dosage form, administration method, administration object, etc.), such as 0.1-95% by weight, such as 1-95% by weight, 5-90% by weight , 10-80% by weight, etc.
  • 0.01-10 g (eg, 0.05 g, 0.1 g, 0.5 g, 1 g or 5 g, etc.) of the compound of the present invention may be included in the pharmaceutical composition of the present invention.
  • the present invention relates to compounds of formula (I), (II), (III), (IV) or isotopically labeled compounds thereof, or optical isomers, geometric isomers, tautomers or isomers thereof Use of a mixture of conformers, or a pharmaceutically acceptable salt thereof, or a prodrug thereof, or a metabolite thereof in the preparation of a medicament for treating or preventing a disease or condition mediated by CCR4 as a CCR4 antagonist.
  • Compounds of formula (I), (II), (III), (IV) or their isotope-labeled compounds, or their optical isomers, geometric isomers, Tautomers or mixtures of isomers, or pharmaceutically acceptable salts thereof, or prodrugs thereof, or metabolites thereof may be used to treat or prevent diseases mediated by CCR4 in a subject in need thereof or illness.
  • subject refers to any human or non-human organism that could potentially benefit from treatment with a compound of formula (I), (II), (III), (IV).
  • exemplary subjects include humans or mammals of any age.
  • the subject is a human.
  • treatment includes treating a disease or condition in a mammal, especially a human and includes: (a) inhibiting an infection, disease or condition, i.e. arresting or delaying the development of an infection, disease or condition; (b) alleviating an infection, disease or condition; A disease or condition, ie causing regression of the disease or condition, and/or (c) cure of the infection, disease or condition.
  • prevention includes prophylactic therapy in mammals, especially humans, aimed at reducing the likelihood of an infection, disease or condition occurring. Patients may be selected for prophylactic therapy based on an increased risk of infection or having a disease or condition compared to the general population as a factor. "Preventing” can include treating a subject who has not yet exhibited an infection or clinical condition, and preventing a second occurrence of the same or similar infection or clinical condition.
  • the compounds of the present invention can inhibit CCR4-mediated cell migration. Therefore, the compounds of the present invention can be used in the prevention or treatment of diseases or conditions mediated by CCR4.
  • the disease or condition mediated by CCR4 may be selected from one or more of atopic dermatitis, asthma, allergic rhinitis, atopic dermatitis, systemic lupus erythematosus and rheumatoid arthritis.
  • atopic dermatitis asthma, allergic rhinitis, atopic dermatitis, systemic lupus erythematosus and rheumatoid arthritis.
  • Various immune-related diseases may be cancer.
  • the cancer is preferably selected from cholangiocarcinoma, liver cancer, breast cancer, prostate cancer, lung cancer, nasopharyngeal cancer, thyroid cancer, gastric cancer, ovarian cancer, colorectal cancer, endometrial cancer, urothelial cell carcinoma, testicular cancer, Cervical cancer, leukemia, skin cancer, squamous cell carcinoma, basal cell carcinoma, bladder cancer, esophageal cancer, head and neck cancer, kidney cancer, pancreatic cancer, bone cancer, lymphoma, melanoma, sarcoma, peripheral neuroepithelial tumor, glia tumors, ependymomas, neuroblastomas, ganglioneuromas, medulloblastomas, pineal cell tumors, meningiomas, neurofibromas, schwannomas, and Wilms tumors.
  • the present invention provides a method of treating or preventing a disease or condition mediated by CCR4, the method comprising administering to a subject in need a therapeutically effective amount of formula (I), (II), (III), (IV) compound or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its precursor drugs, or their metabolites.
  • the present invention relates to compounds of formula (I), (II), (III), (IV) or isotopically labeled compounds thereof, or optical isomers, geometric isomers, tautomers or isomers thereof A mixture of conformers, or a pharmaceutically acceptable salt thereof, or a prodrug thereof, or a metabolite thereof, wherein it is used for preventing or treating a disease or condition mediated by CCR4.
  • the compounds of the present invention can be administered orally, parenterally, intravenously, intramuscularly, subcutaneously, nasally, buccally, ocularly, via the lungs, via the respiratory tract, vaginally, rectally, intraperitoneally, intralesionally, Administer around the lesion and other routes.
  • a “therapeutically effective amount” refers to an amount of a compound of the present invention effective to treat or prevent a disease or condition mediated by CCR4 when administered alone or in combination.
  • the daily dose of the compound of the invention may specifically be 0.001-150 mg/kg body weight (eg 0.1 mg/kg body weight, 1 mg/kg body weight, 10 mg/kg body weight or 100 mg/kg body weight etc.).
  • the specific administration frequency can be determined by those skilled in the art, for example, once a day, once a day, once a day, once a day, once a day, once a day, once a day, once a day, twice a day, 1 day 3 times etc.
  • Figure 1 shows the inhibitory effect of test compounds on FITC-induced ear swelling in an animal model.
  • Figure 2 shows the effect of test compounds on body weight in FITC-induced animal models.
  • Figure 3 shows the inhibitory effect of test compounds on OXA-induced ear swelling in animal models.
  • the compounds of formula (I) of the present invention can be synthesized by various methods familiar to those skilled in the art of organic synthesis. Some exemplary synthetic methods of compounds of formula (I) are given in the following specific examples, and these methods are well known in the field of synthetic chemistry. Consequently, with reference to the exemplary schemes in this patent, those skilled in the art can easily design the synthetic routes of other compounds of formula (I) by appropriately adjusting the reactants, reaction conditions and protecting groups.
  • compound B-1 (25.00g, 158.95mmol) and tert-butyl 3-iodoazetidine-1-carboxylate (25.00g, 88.31mmol) were dissolved in isopropanol (400mL), and added Nickel iodide (2.76g, 8.83mmol), (1R,2R)-2-aminocyclohexanol hydrochloride (1.34g, 8.83mmol) and sodium bis(trimethylsilyl)amide (176.60mL, 176.61mmol , 1mol/L tetrahydrofuran solution), the reaction mixture was stirred at 80°C for 18 hours under nitrogen protection.
  • compound B-2 26.00g, 96.75mmol
  • (S)-4-isopropyloxazolidin-2-one 24.99g, 193.50mmol
  • anhydrous toluene 400mL
  • tris(dibenzylideneacetone)dipalladium 4.43g, 4.84mmol
  • 2-dicyclohexylphosphine-2,4,6-triisopropylbiphenyl 2.31g, 4.84mmol
  • cesium carbonate 31.52 g, 96.75mmol
  • reaction mixture was cooled to room temperature, filtered, the filter cake was washed with methyl tert-butyl ether (500 mL), the organic phase was washed with saturated brine (500 mL ⁇ 4), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a residue
  • compound B-3 (10.00g, 27.67mmol) was dissolved in anhydrous acetic acid (200mL), under the protection of nitrogen, slowly added platinum dioxide (1.00g, 4.40mmol), hydrogen replacement, the reaction mixture in hydrogen (15Psi ) atmosphere at room temperature for 15 hours.
  • the reaction solution was filtered, the filter cake was washed with methanol (100 mL), and the filtrate was concentrated under reduced pressure.
  • the crude product was dissolved in dichloromethane (50 mL), and 1 mol/L aqueous sodium hydroxide solution (80 mL) was added to adjust the pH to 13.
  • the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain the crude intermediate B.
  • the crude intermediate B was dissolved in methyl tert-butyl ether (1000 mL), and S-mandelic acid (13.61 g, 89.45 mmol) was added in portions under reflux, and the reaction mixture was stirred at room temperature for 16 hours.
  • a large amount of white solid precipitated, filtered, and the filter cake was washed with methyl tert-butyl ether (400 mL), and dissolved in dichloromethane (500 mL) and 1 mol/L sodium hydroxide aqueous solution (800 mL).
  • the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain the crude product.
  • the crude product was dissolved in methyl tert-butyl ether (500 mL), and S-mandelic acid (8.23 g, 54.09 mmol) was added in portions under reflux, and the reaction mixture was reacted at room temperature for 16 hours.
  • intermediate B (10.00g, 36.13mmol) and 2-iodoethanol (12.43g, 72.25mmol) were dissolved in acetonitrile (100mL), potassium carbonate (14.98g, 108.38mmol) was slowly added, and the reaction solution was stirred at 80°C 16 hours. After the reaction solution was cooled to room temperature, dichloromethane (200 mL) was added to dilute, filtered, and the filtrate was concentrated under reduced pressure.
  • intermediate B (3.00g, 12.48mmol), compound D-2 (1.76g, 13.73mmol) and sodium triacetoxyborohydride (3.95g, 18.72mmol) were dissolved in 1,2-dichloroethyl Acetic acid (0.70 mL, 12.48 mmol) was slowly added dropwise to the reaction solution at -5°C, and stirred at 20°C for 15 hours after the addition was complete.
  • compound D-3 (2.60g, 7.38mmol) was dissolved in a mixed solvent of dichloromethane (30mL) and methanol (3mL), and trimethylsilyldiazomethane (14.70mL , 29.51 mmol), followed by stirring at 20°C for 2 hours. After completion of the reaction, acetic acid was slowly added dropwise to the reaction solution until the color of the reaction solution changed from yellow to colorless and no longer bubbled, diluted with water (30mL), extracted with dichloromethane (30mL), saturated aqueous sodium chloride ( 25 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound D-4.
  • compound D-4 (1.75g, 4.78mmol) was dissolved in 1,4-dioxane (30mL), and 4mol/L dioxane hydrochloride solution (6.00mL, 24.00mmol) was slowly added to react The solution was stirred at 20°C for 15 hours. The reaction solution was concentrated under reduced pressure to obtain intermediate D. The crude product was directly used in the next reaction without further purification.
  • Example 5 4-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-(3-((R)-1-(2-hydroxyethyl)piper Pyridin-3-yl)azetidin-1-yl)pyrazol[1,5-a][1,3,5]triazine-7-carbonitrile (5)
  • reaction solution was cooled to room temperature, concentrated under reduced pressure, and the resulting residue was subjected to preparative high-performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge 150*25mm*5 ⁇ m; mobile phase: water (10mM ammonium bicarbonate ), acetonitrile; gradient ratio: acetonitrile phase (0-8min, 52-82%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 5.
  • MS-ESI m/z 515.2 [M+H] + .
  • Example 7 2-((R)-3-(1-(8-chloro-4-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)pyrazol[1 ,5-a][1,3,5]triazin-2-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (7)
  • compound 7-2 (170mg, 0.39mmol), intermediate C (131mg, 0.59mmol), N,N-diisopropylethylamine (203mg, 1.57mmol) were dissolved in acetonitrile (6mL) and dimethyl sulfoxide (2 mL), and the reaction solution was stirred at 30°C for 16 hours.
  • reaction solution was concentrated under reduced pressure, and the resulting residue was subjected to preparative high-performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge C18 150*25mm*5 ⁇ m; mobile phase: water (10mM ammonium bicarbonate), acetonitrile ; Gradient ratio: acetonitrile phase (0-10min, 50-80%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 7.
  • MS-ESI m/z 524.0 [M+H] + .
  • Example 8 4-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-(3-((R)-1-(2-hydroxyethyl)piper Pyridin-3-yl)azetidin-1-yl)pyrazol[1,5-a][1,3,5]triazine-8-carbonitrile (8)
  • compound 8-1 (300 mg, 0.62 mmol) was added to 1-methyl-2-pyrrolidone (2 mL), followed by cuprous cyanide (277 mg, 3.09 mmol), replaced by nitrogen, and reacted at 140°C for 3 hours. After the reaction was completed, the reaction solution was cooled to room temperature, poured into water (30 mL) for dilution, extracted with ethyl acetate (20 mL ⁇ 2), combined the organic phases, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure.
  • reaction solution was poured into water (30 mL) for dilution, washed with ethyl acetate (20 mL ⁇ 2), the organic phase was discarded, the aqueous phase was concentrated under reduced pressure, and the resulting residue was subjected to preparative high performance liquid chromatography (formic acid/acetonitrile/water system, Chromatographic column: Waters Xbridge C18 150*25mm*5 ⁇ m; mobile phase: water (0.0225% formic acid), acetonitrile; gradient ratio: acetonitrile phase (0-10min, 37-67%); flow rate: 25mL/min; column temperature: room temperature) to obtain compound 9.
  • MS-ESI m/z 557.2 [M+H] + .
  • compound 10-8 (330mg, 0.29mmol, 53% purity) and lithium hydroxide monohydrate (36mg, 0.87mmol) were dissolved in tetrahydrofuran (3mL) and water (3mL), and the reaction mixture was stirred at room temperature for 3 hours .
  • reaction solution was cooled to room temperature, poured into water (10 mL) for dilution, and washed with dichloromethane (20 mL ⁇ 2).
  • the organic phase was discarded, and the aqueous phase was subjected to preparative high-performance liquid chromatography (formic acid/acetonitrile/water system, chromatographic column: Phenomenex C18 75*30mm*3 ⁇ m; mobile phase: water (10mM formic acid), acetonitrile; gradient ratio: acetonitrile phase ( 0-7min, 8-38%); flow rate: 25mL/min; column temperature: room temperature) to obtain compound 11.
  • MS-ESI m/z 557.2 [M+H] + .
  • compound 12-3 (2.63g, crude product) and N,N-diisopropylethylamine (2.90mL, 17.57mmol) were dissolved in a mixed solvent of methanol (20mL) and ethanol (20mL), and then separated Hydroxylamine hydrochloride (814mg, 11.71mmol) was added in batches, and the reaction solution was reacted at 45°C for 2 hours.
  • MS-ESI m/z 357.0 [M+H] + .
  • compound 15-1 (5.00g, 59.47mmol) was dissolved in N,N-dimethylformamide (50mL), and ethoxycarbonyl isothiocyanate (7.80g, 59.47mmol) was slowly added dropwise, The reaction solution was reacted at 25° C. for 15 hours. After the reaction was completed, the reaction solution was poured into water (500 mL), filtered, the filter cake was washed with water (100 mL), and dried in vacuo to obtain compound 15-2.
  • reaction solution was cooled to room temperature, filtered, and the filter cake was dissolved by adding water (10mL), and the pH was adjusted to 6-7 with 1mol/L hydrochloric acid aqueous solution, and the mixture was passed through a reverse-phase column (acetonitrile/water system, chromatographic column: C18spherical20 -35 ⁇ m 40g, mobile phase: water, acetonitrile; gradient ratio: acetonitrile phase (0-8min, 0-5%); flow rate: 35mL/min; column temperature: room temperature) separation to obtain compound 15-3.
  • 1 H NMR 400 MHz, DMSO-d 6 ) ⁇ 11.53 (brs, 1H), 7.92 (s, 1H).
  • reaction solution was cooled to room temperature, concentrated under reduced pressure, and the resulting residue was separated by preparative thin-layer chromatography on silica gel (pure methanol), and then separated by preparative high-performance liquid chromatography (formic acid/acetonitrile/water system, chromatographic column: Unisil 3-100C18Ultra 150* 50mm*3 ⁇ m; mobile phase: water (0.5% formic acid), acetonitrile; gradient ratio: acetonitrile phase (0-7min, 10-40%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 15.
  • MS-ESI m/z 491.2 [M+H] + .
  • Example 17 2-(R)-3-(1-(7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-(trifluoromethyl)- [1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)azetidin-3-yl)piperidin-1-yl)ethane -1-ol(17)
  • Example 20 was prepared referring to the synthesis method of Example 19. MS-ESI: m/z 504.2 [M+H] + .
  • 1 H NMR 400MHz, CD 3 OD
  • Example 22 2-(R)-3-(1-(7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-(methoxymethyl) -[1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (22)
  • reaction solution was poured into water (30 mL), extracted with dichloromethane (20 mL ⁇ 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the obtained residue was subjected to preparative high performance liquid chromatography ( Acetonitrile/ammonium bicarbonate system, chromatographic column: ACSWH-GX-A Waters Xbridge 150*25mm*5 ⁇ m; mobile phase: water (0.1% ammonium bicarbonate), acetonitrile; gradient ratio: acetonitrile phase (0-9min,60- 90%); flow rate: 25mL/min; column temperature: room temperature) to obtain compound 25.
  • preparative high performance liquid chromatography Acetonitrile/ammonium bicarbonate system, chromatographic column: ACSWH-GX-A Waters Xbridge 150*25mm*5 ⁇ m; mobile phase: water (0.1% ammonium bicarbonate), acetonitrile; gradient ratio: acetonit
  • compound 25-2 (100mg, 0.22mmol) was dissolved in acetonitrile (6mL), then 3-iodopropanol (83mg, 0.44mmol) and potassium carbonate (92mg, 0.67mmol) were added, and the reaction mixture was heated at 80°C Stir for 3 hours.
  • reaction solution was concentrated under reduced pressure, and the resulting residue was subjected to preparative high-performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge C18 150*25mm*5 ⁇ m; mobile phase: water (10mM ammonium bicarbonate), acetonitrile ; Gradient ratio: acetonitrile phase (0-10min, 35-65%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 26.
  • MS-ESI m/z 504.3 [M+H] + .
  • Example 28 N-((R)-1-(2,4-dichlorophenyl)ethyl)-5-(3-(R)-2-(2-(methylsulfonyl)ethyl) Piperidin-3-yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (28)
  • reaction solution was concentrated under reduced pressure, and the obtained residue was subjected to preparative high performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge C18 150*25mm*5 ⁇ m; mobile phase: water (10mM ammonium bicarbonate), acetonitrile; Gradient ratio: acetonitrile phase (0-10min, 46-76%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 29.
  • MS-ESI m/z 542.3[M+H] + .
  • Example 31 and Example 32 (1R,4r)-4-((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl) Amino)-[1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclohexyl alkan-1-ol and (1S,4s)-4-((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2 ,4] Triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclohexane-1-alcohol (31 and 32)
  • compound 31-2 (1.00g, 4.65mmol, 80% purity) was dissolved in tetrahydrofuran (15mL), then 1mol/L dilute hydrochloric acid (8.00mL, 8.00mmol) was added, and the reaction solution was stirred at 25°C for 16 hours .
  • Example 33 (1S,3s)-3-((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[ 1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclobutane-1- Base (33) and Example 34: (1R,3r)-3-((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl )amino)-[1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)-1-methyl ring Butan-1-ol(34)
  • compound 25-2 200 mg, 0.44 mmol was dissolved in methanol (6 mL), followed by addition of compound 33-1 (147 mg, 0.88 mmol, 60% purity), sodium cyanoborohydride (83 mg, 1.32 mmol) and acetic acid (79mg, 1.32mmol), the reaction mixture was stirred at 25°C for 16 hours. The reaction solution was filtered, and the filtrate was concentrated under reduced pressure to obtain compound 33-2.
  • Compound 33-2 (200mg, 90% purity) was subjected to normal phase liquid chromatography (column: DAICEL CHIRALPAK IC (250mm*50mm*10 ⁇ m); mobile phase: hexane, ethanol (0.1% ammonia water); gradient ratio: ethanol Phase 70%; flow rate: 100mL/min; column temperature: room temperature) separation, and then separated by preparative thin-layer chromatography (pure methanol) to obtain compound 33 and compound 34.
  • Compound 33 MS-ESI: m/z 530.2 [M+H] + .
  • Example 35 and Example 36 N-((R)-1-(2,4-dichlorophenyl)ethyl)-5-(3-(R)-1-((1s,3S)-3 -Methoxycyclobutyl)piperidin-3-yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine and N - ((R)-1-(2,4-dichlorophenyl)ethyl)-5-(3-(R)-2-((1r,3R)-3-methoxycyclobutyl)piperidine -3-yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (35 and 36)
  • Compound 35-1 (70mg, 0.13mmol) was resolved by supercritical fluid chromatography (column: DAICEL CHIRALCEL OJ (250mm*30mm*10 ⁇ m); mobile phase: supercritical carbon dioxide, ethanol (0.1% ammonia monohydrate); gradient Ratio: methanol phase 25%; flow rate: 55mL/min; column temperature: room temperature), compound 35 and compound 36 were obtained. (Compound 35 was the first eluting peak, compound 36 was the second eluting peak). Compound 35: MS-ESI: m/z 530.2 [M+H] + .
  • Example 37 and Example 38 N-((R)-1-(2,4-dichlorophenyl)ethyl)-5-(3-(R)-1-((1s,3S)-3 -(methylsulfonyl)cyclobutyl)piperidin-3-yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidine-7- Amine and N-((R)-1-(2,4-dichlorophenyl)ethyl)-5-(3-(R)-1-((1r,3R)-3-(methylsulfonyl )cyclobutyl)piperidin-3-yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (37 and 38)
  • compound 37-1 (5.00 g, 28.37 mmol) was dissolved in methanol (20 mL), sodium borohydride (1.14 g, 30.14 mmol) was added in three batches, and the reaction mixture was stirred at 0°C for 1 hour. After the reaction was completed, it was quenched by adding water (20 mL), and concentrated under reduced pressure. The residue was diluted with ethyl acetate (200 mL), washed with water (30 mL ⁇ 3). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound 37-2.
  • compound 37-2 (4.50g, 25.25mmol), methanesulfonic anhydride (6.60g, 37.87mmol) and triethylamine (10.50mL, 75.75mmol) were dissolved in dichloromethane (50mL), and the reaction The mixture was stirred at room temperature for 4 hours. After the reaction was completed, it was diluted with dichloromethane (100 mL), and washed with water (50 mL ⁇ 3). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound 37-3.
  • compound 37-4 (1.00g, 4.80mmol) was dissolved in dichloromethane (10mL), then m-chloroperoxybenzoic acid (2.44g, 12.00mmol, 85% purity) was added, and the reaction mixture was stirred at room temperature for 2 Hour.
  • Compound 37-8 was separated by supercritical fluid chromatography (column: DAICEL CHIRALPAK AS (250mm ⁇ 30mm*10 ⁇ m); mobile phase: supercritical carbon dioxide, ethanol (0.1% ammonia monohydrate); gradient ratio: ethanol phase 40 %; flow rate: 70mL/min; column temperature: room temperature), compound 37 and compound 38 were obtained. (Compound 37 was the first eluting peak, compound 38 was the second eluting peak). Compound 37: MS-ESI: m/z 578.4 [M+H] + .
  • Example 39 and Example 40 (R)-1-((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino) -[1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)propan-2-ol and (S)- 1-((R)-3- (1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4]triazolo[1,5-a]pyrimidine-5 -yl)azetidin-3-yl)piperidin-1-yl)propan-2-ol (39 and 40)
  • Compound 39-1 (140mg, 0.28mmol) was resolved by supercritical fluid chromatography (column: DAICEL CHIRALCEL OJ (250mm*30mm*10 ⁇ m); mobile phase: supercritical carbon dioxide, ethanol (0.1% ammonia monohydrate); gradient Ratio: ethanol phase 25%; flow rate: 60mL/min; column temperature: room temperature), compound 39 and compound 40 were obtained. (Compound 39 was the first eluting peak, compound 40 was the second eluting peak). Compound 39: MS-ESI: m/z 504.4 [M+H] + .
  • Example 41 1-(((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4 ]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)methyl)cyclopropan-1-ol (41)
  • compound 41-1 (1.80g, 13.83mmol) was dissolved in N,N-dimethylformamide (30mL), the temperature was lowered to 0°C, and sodium hydrogen (830mg, 20.75mmol, 60% purity ), the reaction solution was stirred at 0°C for 20 minutes, p-methoxybenzyl chloride (2.38 g, 15.21 mmol) was added, and stirred at room temperature for 3 hours. After the reaction was completed, saturated ammonium chloride aqueous solution (50 mL) was added to the reaction liquid to quench, and ethyl acetate (50 mL ⁇ 2) was extracted.
  • Example 43 3-(((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4 ]triazol[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)methyl)thietane-1,1-dioxide ( 43)
  • Example 45 (1R,3r)-3-(4-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2, 4] Triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclobutane-1-ol (45) and
  • Example 46 (1S,3s)-3-(4-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2 ,4] Triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclobutane-1-alcohol (46 )
  • compound 45-5 150 mg, 0.34 mmol
  • compound 33-1 48 mg, 0.34 mmol, 70% purity
  • acetic acid 2 mg, 0.03 mmol
  • methanol 3 mL
  • Sodium cyanoborohydride 63 mg, 1.01 mmol
  • the reaction solution was concentrated under reduced pressure, and the residue was separated by silica gel preparative thin-layer chromatography (pure methanol) to obtain compound 45-6.
  • Compound 45-6 was resolved by preparative supercritical fluid chromatography (column: DAICEL CHIRALPAK AD (250mm*30mm*10 ⁇ m); mobile phase: supercritical carbon dioxide, methanol (0.1% ammonia monohydrate); gradient ratio: methanol phase 40 %; flow rate: 70mL/min; column temperature: room temperature), compound 45 and compound 46 were obtained.
  • compound 52-3 (180mg, 0.34mmol, 93% purity) was dissolved in acetonitrile (8mL), then 2-iodoethanol (116mg, 0.67mmol) and potassium carbonate (140mg, 1.01mmol) were added, and the reaction mixture Stir at 80°C for 16 hours.
  • reaction solution was concentrated under reduced pressure, and the resulting residue was subjected to preparative high-performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge C18 150*25mm*5 ⁇ m; mobile phase: water (10mM ammonium bicarbonate), acetonitrile ; Gradient ratio: acetonitrile phase (0-10min, 36-66%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 52.
  • MS-ESI m/z 504.3 [M+H] + .
  • Example 53 and Example 54 2-((R)-3-(1-(7-(((R)-1-(2-chloro-4-methylphenyl)ethyl)amino)-[ 1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol and 2-(R) -3-(1-(7-((R)-1-(4-chloro-2-methylphenyl)ethyl)amino)-[1,2,4]triazolo[1,5-a ]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (53 and 54)
  • compound 53-1 500mg, 3.30mmol was dissolved in toluene (6mL), replaced with nitrogen, and a solution of methylmagnesium bromide in tetrahydrofuran (3.30mL, 9.90mmol, 3mol/L) was added, and the reaction mixture was heated at 110 °C and stirred for 18 hours. After the reaction solution was cooled to 0°C, dilute hydrochloric acid was slowly added dropwise to pH ⁇ 2, the temperature was raised to reflux and stirred for 1 hour, and then cooled to room temperature.
  • compound 53-3 (450 mg, 1.52 mmol, 62% purity) was dissolved in tetrahydrofuran (10 mL), slowly added dropwise into borane dimethyl sulfide solution (0.50 mL, 5.00 mmol, 10 mol/L), and the reaction mixture Stir at 60°C for 16 hours. The reaction solution was cooled to 0°C, and methanol (10 mL) was slowly added dropwise until no obvious bubbles were generated. Stirring was continued at room temperature for 1 hour, and concentrated under reduced pressure to obtain compound 53-4. MS-ESI: m/z 153.0 [M-NH 3 +H] + .
  • Example 58 2-(R)-2-(1-(7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-(trifluoromethyl)- [1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)morpholinyl)ethan-1-ol (58)
  • compound 58-1 (3.00g, 16.20mmol) was dissolved in anhydrous methanol (30mL), under ice cooling, nitromethane (2.97g, 48.59mmol) and triethylamine (3.28g, 32.39mmol) were added ), and the reaction mixture was stirred at room temperature for 16 hours.
  • compound 58-2 (3.60 g, 14.62 mmol) and platinum dioxide (664 mg, 2.92 mmol) were added into methanol (50 mL), and reacted at room temperature for 16 hours under hydrogen atmosphere. The reaction solution was filtered and concentrated under reduced pressure to obtain compound 58-3.
  • compound 58-3 (3.10g, 14.33mmol) and triethylamine (3.99mL, 28.67mmol) were dissolved in dichloromethane (50mL), and chloroacetyl chloride (1.78g, 15.77mmol) was slowly added dropwise, at room temperature React for 16 hours.
  • the reaction solution was diluted with water (50 mL), extracted with dichloromethane (50 mL ⁇ 2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound 58-4.
  • compound 58-6 (2.60g, 6.66mmol) was dissolved in anhydrous tetrahydrofuran (50mL), and borane dimethyl sulfide (3.30mL, 33.29mmol, 10mol/L) was added dropwise under ice cooling, and the reaction mixture was Stir at 50°C for 16 hours.
  • Compound 58-7 (1.67g) was resolved by supercritical fluid chromatography (chromatographic column: Phenomenex-Cellulose-2 (250mm*30mm, 10 ⁇ m); mobile phase: supercritical carbon dioxide, methanol (0.1% ammonia monohydrate); gradient formulation Ratio: methanol phase 30%; flow rate: 60mL/min; column temperature: room temperature), to obtain compound 58-8 and compound 58-9.
  • Compound 58-8 was the first eluting peak
  • compound 58-9 was the second eluting peak
  • the in vitro CCR4 receptor cell functional assay was used to detect the activity of the compound to be tested by using a cell line stably expressing CCR4 and FLIPR assay.
  • the initial concentration of the test compound was 10 ⁇ M, diluted 3 times, and a total of 10 concentrations were tested in duplicate wells.
  • a cell line stably expressing CCR4 (CCR4-HEK293) was cultured in DMEM medium supplemented with fetal bovine serum, G418 and penicillin-streptomycin. Configure 250mM FLIPR experimental buffer, 2 ⁇ Fluo-4Direct TM loading buffer. Collect and resuspend CCR4-HEK293 cells to 1x10 6 /mL, and add 20 ⁇ L of cell suspension to each reaction well of a 384-well cell culture microplate. Cells were cultured overnight. The next day, discard the supernatant from the cell reaction plate, and add 20 ⁇ L of assay buffer and 20 ⁇ L of Fluo-4Direct TM loading buffer.
  • Compound plate pre-gradiently diluted in 384-well PP microplate
  • 10 ⁇ L of the dilution of the compound to be tested was added to the cell reaction plate with 10 ⁇ L of the dilution of the compound to be tested, incubated at 37°C in a 5% carbon dioxide environment for 50 minutes, and then incubated at room temperature for 10 minutes.
  • the reaction plate was transferred to a FLIPR Tetra System (Molecular Devices).
  • the inhibitory activity of the disclosed compounds on CCR4 receptor cell calcium flow was determined by the above tests, and the measured IC 50 values are shown in Table 1.
  • the experimental results show that the test compound of the present invention has significant inhibitory activity on the calcium flow of CCR4 receptor cells.
  • Comparative Example 1 was prepared according to WO2018/02299; Comparative Example 2 was prepared according to Example 38 of WO2019/147862
  • Centrifuge the CCRF-CEM cells remove the supernatant, resuspend in FBS, adjust the cell density to 2 ⁇ 10 6 /mL, and divide the cells into 96-well cell plates, 99 ⁇ L per well.
  • hCCL22 solution with a concentration of 2.5 nM
  • hCCL22 Using DPBS as a solvent, prepare a hCCL22 solution with a concentration of 2.5 nM, take out the lower cell plate of ChemoTX, and dispense hCCL22 into the cell plate, 30 ⁇ L per well. Cover the lower plate with a ChemoTX microporous membrane, remove the cells from the incubator, transfer 50 ⁇ L of cells to the microporous membrane per well, cover the lid, and incubate for another 1.5 hours at 37°C in a 5% carbon dioxide incubator .
  • the inhibitory activity of the compounds of the present invention on CCR4-mediated chemotaxis of CCRF-CEM cells was determined by the above tests, and the measured IC 50 values are shown in Table 2.
  • the experimental results show that the test compounds of the present invention have significant inhibitory activity on CCRF-CEM cell chemotaxis, and the inhibitory activity of most compounds is obviously stronger than that of Comparative Example 1 and Comparative Example 2.
  • Test Example 3 CCR4-mediated Th2 and Treg cell chemotaxis inhibitory activity test
  • the hTh2 (or hTreg) cells were centrifuged to remove the culture medium, resuspended in 100% fetal bovine serum, adjusted the cell density to 2 ⁇ 10 6 /mL, and split the cells into 96-well cell plates, 99 ⁇ L per well.
  • DPBS DPBS
  • a recombinant human CCL22 solution with a concentration of 2.5 nM take out the lower cell plate of ChemoTX, and dispense recombinant human CCL22 into the cell plate, 30 ⁇ L per well.
  • Cover the lower plate with a ChemoTX microporous membrane take out the cells from the incubator, transfer 50 ⁇ L of cells per well to the microporous membrane, cover the lid, and store at 37°C in a fine atmosphere of 5% carbon dioxide. Incubate for another 1.5 hours in the incubator.
  • the inhibitory activity of the compounds of the present invention on CCR4-mediated chemotaxis of Th2 and Treg cells was determined by the above tests, and the measured IC 50 values are shown in Table 3. Experimental results show that the test compound of the present invention has significant inhibitory activity on both Th2 cell chemotaxis and Treg cell chemotaxis.
  • Test Example 4 Evaluation of the occupancy rate of the compound on the cell surface CCR4 receptor
  • the CCRF-CEM cells were centrifuged at 300 rcf to remove the medium, resuspended in 100% fetal calf serum, adjusted the cell density to 2 ⁇ 10 6 /mL, and split the cells into 96-well cell plates, 99 ⁇ L per well.
  • Two wells were set for each concentration of the compound, 1 ⁇ L of the compound to be tested was transferred to each well of the 96-well plate, and placed in a cell culture incubator with 5% carbon dioxide at 37° C. for 0.5 hours.
  • DPBS DPBS
  • hCCL22 solution with a concentration of 20 ⁇ M was prepared, and the 96-well plate was taken out of the cell culture incubator, and 1 ⁇ L of hCCL22 solution was added to one well of each concentration of the compound, and 1 ⁇ L of DPBS solution was added to the other well, and placed at 37 °C again. , and incubate for 0.5 hr in a 5% carbon dioxide incubator. Take out the cell plate, add 1 ⁇ L of PE-conjugated anti-human CD194 (CCR4) antibody to each cell well, and incubate in a refrigerator at 4°C in the dark for 0.5 hours.
  • CCR4 PE-conjugated anti-human CD194
  • the occupancy rate of the compound of the present invention on the CCR4 receptor on the cell surface was determined by the above method, and the experimental results are shown in Table 4.
  • the experimental results show that the test compound of the present invention can show a higher occupancy rate of CCR4 at a lower concentration, and under the premise of producing the same CCR4 occupancy rate, the required concentration of the test compound of the present invention is significantly lower than that of Comparative Example 1 and Comparative example 2.
  • test compound was dissolved in 5% DMSO + 5% Solutol + 90% saline, vortexed and sonicated to prepare a clear solution of corresponding concentration, which was filtered through a microporous membrane for use.
  • Balb/c male mice were selected, and the candidate compound solution was administered intravenously at a dose of 1 mg/kg.
  • the test compound was dissolved in 4% DMSO+96% (0.5% HPMC+0.1% TW-80) or 5% DMSO+40% PEG400+10% Solutol+45% water, vortexed and sonicated to prepare the corresponding concentration of clear
  • the solution was filtered through a microporous membrane for later use.
  • Balb/c male mice were selected, and the candidate compound solution was orally administered at a dose of 10 mg/kg or 50 mg/kg.
  • Whole blood was collected at a certain time point, and plasma was prepared, and the drug concentration was analyzed by LC-MS/MS method, and the pharmacokinetic parameters were calculated.
  • the experimental results are shown in Table 5.
  • the experimental results show that the test compound of the present invention has better pharmacokinetic properties in mice.
  • test compound was dissolved in 5% DMSO + 5% Solutol + 90% saline, vortexed and sonicated to prepare a clear solution of corresponding concentration, which was filtered through a microporous membrane for use.
  • Male SD rats were selected, and the candidate compound solution was administered intravenously at a dose of 1 mg/kg.
  • the test compound was dissolved in 5% DMSO + 40% PEG400 + 10% Solutol + 45% water, vortexed and sonicated to prepare a clear solution of corresponding concentration, which was filtered through a microporous membrane for use.
  • Male SD rats were selected, and the candidate compound solution was orally administered at a dose of 50 mg/kg.
  • Whole blood was collected at a certain time point, and plasma was prepared, and the drug concentration was analyzed by LC-MS/MS method, and the pharmacokinetic parameters were calculated.
  • the experimental results are shown in Table 6.
  • the experimental results show that the test compound of the present invention has better pharmacokinetic properties in rats.
  • test compound was dissolved in 5% DMSO + 5% Solutol + 90% saline, vortexed and sonicated to prepare a clear solution of corresponding concentration, which was filtered through a microporous membrane for use.
  • Male Beagle dogs were selected, and the candidate compound solution was administered intravenously at a dose of 1 mg/kg.
  • the test compound was dissolved in 5% DMSO + 40% PEG400 + 10% Solutol + 45% water, vortexed and sonicated to prepare a clear solution of corresponding concentration, which was filtered through a microporous membrane for use.
  • Male Beagle dogs were selected, and the candidate compound solution was orally administered at a dose of 5 mg/kg. Collect whole blood at a certain time point, prepare plasma, analyze drug concentration by LC-MS/MS method, and calculate pharmacokinetics mechanical parameters.
  • the experimental results are shown in Table 7.
  • the experimental results show that the test compound of the present invention has better in vivo pharmacokinetic properties in Beagle dogs.
  • Test Example 8 Evaluation of the Inhibitory Activity of Compounds on the hERG Potassium Ion Channel
  • CHO cells stably expressing hERG were cultured in a cell culture dish with a diameter of 35 mm, placed in an incubator at 37°C and 5% CO 2 , and subcultured at a ratio of 1:5 every 48 hours.
  • the medium formula: 90% Ham's F12 culture Base, 10% fetal bovine serum, 100g/mL geneticin and 100g/mL hygromycin. Aspirate the cell culture medium, rinse with extracellular fluid once, add 0.25% trypsin-EDTA solution, and digest at room temperature for 3-5 minutes. Aspirate the digestion solution, resuspend the cells with extracellular solution, and transfer the cells to a laboratory dish for electrophysiological recording.
  • hERG potassium channel currents were recorded by whole-cell voltage-clamp technique at room temperature.
  • the resistance of the tip after perfusion with the inner solution of the electrode is about 2-5M ⁇ , and the glass microelectrode is inserted into the amplifier probe to connect to the patch clamp amplifier.
  • the clamping voltage and data recording are controlled and recorded by the pClamp software through the computer, the sampling frequency is 10kHz, and the filtering frequency is 2kHz.
  • the cell was clamped at -100mV, and the step voltage of the evoked hERG potassium current was given a 2s depolarization voltage from -100mV to +20mV, then repolarized to -50mV, and returned to - for 1s. 100mV. Give this voltage stimulation every 5s, and start the administration process after confirming that the hERG potassium current is stable (at least 1 minute), and give each test concentration of the compound at least 1 minute until the steady state of action.
  • Data analysis and processing used pClamp 10, GraphPad and Excel software.
  • the experimental results are shown in Table 8.
  • the experimental results showed that the test compounds had weak inhibitory activity on the hERG potassium ion channel, and the IC 50 values of most test compounds were greater than 10 ⁇ M, showing a low risk of hERG potassium ion channel inhibition.
  • the compound of Comparative Example 1 has strong inhibitory activity on the hERG potassium ion channel, with an IC 50 value of 0.15 ⁇ M.
  • Test Example 9 Pharmacodynamic experiment of FITC-induced delayed-type hypersensitivity in mice in vivo
  • mice 7-week-old BALB/c mice (Victoria Lihua) acclimatized for 1 week after arriving at the animal room. According to body weight, they were divided into model control group, positive control dexamethasone group and test compound group. On days 0 and 1, BALB/c mice were anesthetized with isoflurane, and their abdomens were shaved with 400 ⁇ L of 0.5% FITC (dissolved in 50% acetone + 50% dibutyl phthalate). sensitization. On days 6, 7, 8, and 9, 20 ⁇ L of 0.5% FITC was applied to the inner and outer sides of the left ear of the mice for modeling stimulation.
  • 0.5% FITC dissolved in 50% acetone + 50% dibutyl phthalate
  • Test Example 10 Pharmacodynamic experiment of OXA-induced mouse atopic dermatitis in vivo
  • mice 7-week-old BALB/c mice (Shanghai Jihui) acclimatized for 1 week after arriving in the animal room. According to body weight, they were divided into model control group, positive control dexamethasone group and test compound group. On day 0, BALB/c mice were anesthetized with isoflurane, shaved their abdomen, and sensitized with 100 ⁇ L of 1.5% OXA (dissolved in 80% acetone + 20% olive oil). On day 7, 20 ⁇ L of 1.5% OXA was applied to the inner and outer sides of the left ear of the mice for modeling stimulation.
  • OXA dissolved in 80% acetone + 20% olive oil

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (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)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Hydrogenated Pyridines (AREA)

Abstract

The present invention relates to a CCR4 small molecule antagonist and the use thereof. Specifically, the present invention relates to a compound of formula (I) or isotope labeled compounds thereof, or optical isomers, geometric isomers, tautomers or isomer mixtures thereof, or pharmaceutically acceptable salts thereof, or prodrugs thereof, or metabolites thereof, and the use thereof in the preparation of a drug for treating or preventing CCR4-mediated diseases or disorders.

Description

一种CCR4小分子拮抗剂及其用途A kind of CCR4 small molecule antagonist and its use 技术领域technical field
本发明属于医药领域,具体地,涉及用于一种CCR4小分子拮抗剂及其用途。The invention belongs to the field of medicine, and in particular relates to a CCR4 small molecule antagonist and its application.
背景技术Background technique
CCR4(C-C趋化因子受体4型)属于G蛋白偶联受体,是趋化因子受体家族成员之一,广泛表达于免疫细胞表面,特别是Th2细胞及Treg细胞。CCR4主要的内源性配体包括CCL22(又名巨噬细胞衍生的趋化因子,MDC)以及CCL17(又名胸腺活化调节趋化因子,TARC)。CCR4与内源性配体结合,可激活偶联的G蛋白,进而发生级联细胞激活效应,通过一系列的信息传递实现趋化靶细胞向特定位置迁移,发挥生物学效应。CCR4 (C-C chemokine receptor type 4) belongs to G protein-coupled receptors and is a member of the chemokine receptor family. It is widely expressed on the surface of immune cells, especially Th2 cells and Treg cells. The main endogenous ligands of CCR4 include CCL22 (also known as macrophage-derived chemokine, MDC) and CCL17 (also known as thymus activation-regulated chemokine, TARC). The combination of CCR4 and endogenous ligand can activate the coupled G protein, and then a cascade of cell activation effects occurs. Through a series of information transmission, chemotactic target cells migrate to a specific location and exert biological effects.
研究证实,CCR4与特应性皮炎、哮喘、过敏性鼻炎、异位性皮炎、系统性红斑狼疮和类风湿性关节炎等免疫相关性疾病的发生发展密切相关。同时,CCR4在肿瘤微环境中起到调控作用,诱导肿瘤的免疫逃逸和促进肿瘤细胞的转移等。因此,CCR4已经成为治疗免疫相关性疾病和肿瘤免疫治疗的重要的药物靶标,研发CCR4小分子拮抗剂将为上述疾病的治疗提供新的选择。Studies have confirmed that CCR4 is closely related to the occurrence and development of immune-related diseases such as atopic dermatitis, asthma, allergic rhinitis, atopic dermatitis, systemic lupus erythematosus and rheumatoid arthritis. At the same time, CCR4 plays a regulatory role in the tumor microenvironment, inducing tumor immune escape and promoting tumor cell metastasis. Therefore, CCR4 has become an important drug target for the treatment of immune-related diseases and tumor immunotherapy, and the development of CCR4 small molecule antagonists will provide new options for the treatment of the above diseases.
目前为止,有一款CCR4的单克隆抗体Mogamulizumab被批准上市,用于治疗复发或难治性CCR4阳性的成人T细胞白血病淋巴瘤,但仍未有CCR4小分子拮抗剂被批准上市。WO2018/022992涉及FLX475,是目前临床进展最快的CCR4小分子拮抗剂,RPT193是另一个处于临床研究的CCR4小分子拮抗剂,另外大部分CCR4小分子拮抗剂都处于生物活性测试阶段。由此可见,研发CCR4小分子拮抗剂具有良好的应用前景。So far, Mogamulizumab, a CCR4 monoclonal antibody, has been approved for marketing for the treatment of relapsed or refractory CCR4-positive adult T-cell leukemia-lymphoma, but no CCR4 small molecule antagonist has been approved for marketing. WO2018/022992 involves FLX475, which is currently the fastest-growing small-molecule antagonist of CCR4 in clinical practice. RPT193 is another small-molecule antagonist of CCR4 in clinical research. In addition, most small-molecule antagonists of CCR4 are in the stage of biological activity testing. It can be seen that the development of small molecule antagonists of CCR4 has a good application prospect.
发明内容Contents of the invention
本发明的目的在于提供一种新型的CCR4小分子拮抗剂,此类化合物在抑制CCR4方面具有良好的活性,并表现出优异的效果和作用。The purpose of the present invention is to provide a new type of CCR4 small molecule antagonist, this type of compound has good activity in inhibiting CCR4, and exhibits excellent effects and functions.
在第一方面,本发明提供了式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,
In a first aspect, the present invention provides a compound of formula (I) or an isotopically labeled compound thereof, or an optical isomer, geometric isomer, tautomer or isomer mixture thereof, or a pharmaceutically acceptable salts, or their prodrugs, or their metabolites,
其中,in,
X1和X2各自独立地选自C或N;X 1 and X 2 are each independently selected from C or N;
X3、X4和X5各自独立地选自CRb或N;X 3 , X 4 and X 5 are each independently selected from CR b or N;
R1各自独立地选自H、卤素、C1-C3烷基磺酰基、C1-C3烷基、C1-C3卤代烷基、C1-C3烷氧基、C3-C4环烷基或氰基;Each R 1 is independently selected from H, halogen, C 1 -C 3 alkylsulfonyl, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, C 1 -C 3 alkoxy, C 3 -C 4 cycloalkyl or cyano;
R2选自H、C1-C3烷基、C1-C3卤代烷基或C3-C4环烷基;R 2 is selected from H, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl or C 3 -C 4 cycloalkyl;
R3选自H、C1-C3烷基、C1-C3卤代烷基或C3-C4环烷基;R 3 is selected from H, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl or C 3 -C 4 cycloalkyl;
R4选自 R4 is selected from
Y选自CH2或O;Y is selected from CH2 or O;
Ra选自H、卤素、C1-C6烷基、C1-C6卤代烷基、C1-C6烷氧基、C3-C6环烷基、4-7元杂环基、5-6元杂芳基、苯基或氰基,其中所述C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-7元杂环基、5-6元杂芳基或苯基各自独立地任选地被选自卤素、羟基、氰基、C1-C3烷基或C1-C3烷氧基的一个或多个基团所取代;R a is selected from H, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 3 -C 6 cycloalkyl, 4-7 membered heterocyclyl, 5-6 membered heteroaryl, phenyl or cyano, wherein said C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 6 cycloalkyl, 4-7 membered heterocyclic , 5-6 membered heteroaryl or phenyl are each independently optionally selected from one or more groups selected from halogen, hydroxyl, cyano, C 1 -C 3 alkyl or C 1 -C 3 alkoxy replaced by
每个Rb独立地选自H、卤素、C1-C3烷基、C1-C3烷氧基、C3-C6环烷基或氰基,其中所述C1-C3烷基、C1-C3烷氧基或C3-C6环烷基各自独立地任选地被选自卤素、羟基、氨基、氰基、C1-C3烷基或C1-C3烷氧基的一个或多个基团所取代;Each R b is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyl or cyano, wherein the C 1 -C 3 alk radical, C 1 -C 3 alkoxy or C 3 -C 6 cycloalkyl each independently optionally selected from halogen, hydroxy, amino, cyano, C 1 -C 3 alkyl or C 1 -C 3 One or more groups of alkoxy are substituted;
每个Rc独立地选自H、卤素、C1-C3烷基或C3-C4环烷基,其中所述C1-C3烷基或C3-C4环烷基各自独立地任选地被选自卤素、羟基、氰基、C1-C3烷基或C1-C3烷氧基的一个或多个基团所取代;Each R c is independently selected from H, halogen, C 1 -C 3 alkyl or C 3 -C 4 cycloalkyl, wherein each of the C 1 -C 3 alkyl or C 3 -C 4 cycloalkyl is independently optionally substituted by one or more groups selected from halogen, hydroxyl, cyano, C 1 -C 3 alkyl or C 1 -C 3 alkoxy;
每个Rd独立地选自H、卤素、C1-C3烷基、C1-C3烷氧基、C3-C6环烷基或氰基,其中所述C1-C3烷基、C1-C3烷氧基或C3-C6环烷基各自独立地任选地被选自卤素、羟基、氨基、氰基、C1-C3烷基或C1-C3烷氧基的一个或多个基团所取代;Each R d is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyl or cyano, wherein the C 1 -C 3 alk radical, C 1 -C 3 alkoxy or C 3 -C 6 cycloalkyl each independently optionally selected from halogen, hydroxy, amino, cyano, C 1 -C 3 alkyl or C 1 -C 3 One or more groups of alkoxy are substituted;
每个Re独立地为-L-Rfeach Re is independently -LR f ;
每个Rf独立地选自H、卤素、C1-C3烷基、C1-C3烷氧基、C1-C3卤代烷基、C3-C6环烷基、4-7元杂环基、RgC(O)NH-、RgNHC(O)-、RgOC(O)NH-、RgS(O)2NH-、RgNHS(O)2-、RgS(O)2-、-(CH2)n-羟基、-(CH2)n-氨基、-(CH2)n-氰基、-(CH2)n-羧基或2-氧杂-螺[3,3]庚烷基,其中所述C1-C3烷基、C1-C3烷氧基、C3-C6环烷基、4-7元杂环基、-(CH2)n-羟基、-(CH2)n-氨基、-(CH2)n-氰基或-(CH2)n-羧基各自独立地任选地被选自卤素、羟基、氨基、氰基、C1-C3烷基或C1-C3烷氧基的一个或多个基团所取代;Each R f is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 1 -C 3 haloalkyl, C 3 -C 6 cycloalkyl, 4-7 membered Heterocyclyl, R g C(O)NH-, R g NHC(O)-, R g OC(O)NH-, R g S(O) 2 NH-, R g NHS(O) 2 -, R g g S(O) 2 -, -(CH 2 ) n -hydroxyl, -(CH 2 ) n -amino, -(CH 2 ) n -cyano, -(CH 2 ) n -carboxy or 2-oxa- Spiro[3,3]heptyl, wherein the C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyl, 4-7 membered heterocyclyl, -(CH 2 ) n -hydroxyl, -(CH 2 ) n -amino, -(CH 2 ) n -cyano or -(CH 2 ) n -carboxyl are each independently optionally selected from halogen, hydroxyl, amino, cyano , C 1 -C 3 alkyl or one or more groups of C 1 -C 3 alkoxy;
每个Rg独立地选自C1-C3烷基、C3-C6环烷基或3-6元杂环基;Each R g is independently selected from C 1 -C 3 alkyl, C 3 -C 6 cycloalkyl or 3-6 membered heterocyclyl;
每个L独立地选自化学键、4-7元亚杂环基、C1-C3亚烷基或C3-C6亚环烷基,其中所述4-7元亚杂环基、C1-C3亚烷基或C3-C6亚环烷基各自独立地任选地被选 自卤素、羟基、氨基、氰基、C1-C3烷基或C1-C3烷氧基的一个或多个基团所取代;Each L is independently selected from a chemical bond, a 4-7 membered heterocyclic group, a C 1 -C 3 alkylene group or a C 3 -C 6 cycloalkylene group, wherein the 4-7 membered heterocyclic group, C 1 -C 3 alkylene or C 3 -C 6 cycloalkylene are each independently optionally selected Substituted by one or more groups from halogen, hydroxyl, amino, cyano, C 1 -C 3 alkyl or C 1 -C 3 alkoxy;
Z1为0至3的任一整数;Z 1 is any integer from 0 to 3;
Z2为0至3的任一整数;Z 2 is any integer from 0 to 3;
Z3为0至4的任一整数;Z 3 is any integer from 0 to 4;
Z4为0至2的任一整数;并且Z 4 is any integer from 0 to 2; and
每个n独立地为0至3的任一整数。Each n is independently any integer from 0 to 3.
在本发明一些实施方案中,在式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物中,In some embodiments of the present invention, in the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its metabolite,
X1和X2中的一个为N,另一个为C,One of X1 and X2 is N, the other is C,
X3、X4和X5中的至少一个为N。At least one of X 3 , X 4 and X 5 is N.
在本发明一些实施方案中,在式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物中,In some embodiments of the present invention, in the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its metabolite,
选自 selected from
在本发明一些实施方案中,式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物为式(II)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,
In some embodiments of the present invention, the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt , or its prodrug, or its metabolite is a compound of formula (II) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical acceptable salts, or prodrugs thereof, or metabolites thereof,
其中,R1-R3、X1-X5、Y、Ra、Rc、Rd、Re、Z1、Z2、Z3和Z4如式(I)中所定义; Wherein, R 1 -R 3 , X 1 -X 5 , Y, R a , R c , R d , R e , Z 1 , Z 2 , Z 3 and Z 4 are as defined in formula (I);
特别地,In particular,
选自 和/或 selected from and / or
选自 Rc、Rd、Re、Z2和Z3如式(I)中所定义; selected from R c , R d , Re , Z 2 and Z 3 are as defined in formula (I);
更特别地,其为式(II-1)、式(II-2)、式(II-3)、式(II-4)、式(II-5)、式(II-6)、式(II-7)或式(II-8)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,

More particularly, it is formula (II-1), formula (II-2), formula (II-3), formula (II-4), formula (II-5), formula (II-6), formula ( II-7) or formula (II-8) compound or its isotope labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrugs, or its metabolites,

其中,R1-R3、Ra、Rd、Re、Z1和Z3如式(I)中所定义。Wherein, R 1 -R 3 , R a , R d , R e , Z 1 and Z 3 are as defined in formula (I).
在本发明一些实施方案中,式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物为式(III)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,
In some embodiments of the present invention, the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt , or its prodrug, or its metabolite is a compound of formula (III) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical acceptable salts, or prodrugs thereof, or metabolites thereof,
其中,R1-R3、Ra、Re和Z1如式(I)中所定义。Wherein, R 1 -R 3 , R a , R e and Z 1 are as defined in formula (I).
在本发明一些实施方案中,式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物为式(IV)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,
In some embodiments of the present invention, the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt , or its prodrug, or its metabolite is a compound of formula (IV) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical acceptable salts, or prodrugs thereof, or metabolites thereof,
其中,R1-R3、Ra、Re和Z1如式(I)中所定义。Wherein, R 1 -R 3 , R a , R e and Z 1 are as defined in formula (I).
在本发明一些实施方案中,在式(I)、(II)、(III)、(IV)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物中,In some embodiments of the present invention, in the formula (I), (II), (III), (IV) compound or its isotope labeled compound, or its optical isomer, geometric isomer, tautomer or Isomer mixtures, or pharmaceutically acceptable salts thereof, or prodrugs thereof, or metabolites thereof,
每个Re独立地为-L-Rfeach Re is independently -LR f ;
Rf各自独立地选自H、卤素、C1-C3烷基、C1-C3烷氧基、C1-C3卤代烷基、C3-C6环烷基、4-7元杂环基、RgC(O)NH-、RgNHC(O)-、RgOC(O)NH-、RgS(O)2NH-、RgNHS(O)2-、RgS(O)2-、-(CH2)n-羟基、-(CH2)n-氨基、-(CH2)n-氰基、-(CH2)n-羧基或2-氧杂-螺[3,3]庚烷基,其中所述C1-C3烷基、C1-C3烷氧基、C3-C6环烷基、4-7元杂环基、-(CH2)n-羟基、-(CH2)n-氨基、-(CH2)n-氰基或-(CH2)n-羧基各自独立地任选地被选自羟基或C1-C3烷基的一个或多个基团所取代;优选地Rf各自独立地选自H、氟、氯、溴、甲基、乙基、正丙基、异丙基、二氟甲基、三氟甲基、羟基、羧基、甲氧基、氧杂环丁烷基、氮杂环丁烷基、1,1-二氧代硫杂环丁烷基、吗啉基、吡咯烷基、哌啶基、四氢吡喃基、乙酰胺基、甲磺酰基、CH3S(O)2NH-或2-氧杂-螺[3,3]庚烷基;Each R f is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 1 -C 3 haloalkyl, C 3 -C 6 cycloalkyl, 4-7 membered hetero Cyclo, R g C(O)NH-, R g NHC(O)-, R g OC(O)NH-, R g S(O) 2 NH-, R g NHS(O) 2 -, R g S(O) 2 -, -(CH 2 ) n -hydroxyl, -(CH 2 ) n -amino, -(CH 2 ) n -cyano, -(CH 2 ) n -carboxy or 2-oxa-spiro [3,3] Heptyl, wherein the C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyl, 4-7 membered heterocyclyl, -(CH 2 ) n -hydroxyl, -(CH 2 ) n -amino, -(CH 2 ) n -cyano or -(CH 2 ) n -carboxyl are each independently optionally selected from hydroxyl or C 1 -C 3 alkyl Substituted by one or more groups; preferably each R f is independently selected from H, fluorine, chlorine, bromine, methyl, ethyl, n-propyl, isopropyl, difluoromethyl, trifluoromethyl , hydroxyl, carboxyl, methoxy, oxetanyl, azetidinyl, 1,1-dioxothietanyl, morpholinyl, pyrrolidinyl, piperidinyl, tetra Hydropyranyl, acetamido, methanesulfonyl, CH 3 S(O) 2 NH- or 2-oxa-spiro[3,3]heptyl;
Rg选自C1-C3烷基、C3-C4环烷基或3-4元杂环基,优选地选自甲基、乙基、环丙烷基、环丁烷基或氧杂环丁烷基;R g is selected from C 1 -C 3 alkyl, C 3 -C 4 cycloalkyl or 3-4 membered heterocyclic group, preferably selected from methyl, ethyl, cyclopropyl, cyclobutanyl or oxa cyclobutanyl;
每个n独立地为0至3的任一整数,并且each n is independently any integer from 0 to 3, and
L选自化学键、任选地被甲基或乙基取代的亚甲基、亚乙基、亚丙基、亚环丙基、亚环丁基、亚环戊基、亚环己基、亚氮杂环丁烷基、亚吡咯烷基或亚哌啶烷基;L is selected from the group consisting of chemical bond, methylene optionally substituted by methyl or ethyl, ethylene, propylene, cyclopropylene, cyclobutylene, cyclopentylene, cyclohexylene, azepine Cyclobutane, pyrrolidinylene or piperidinylene;
特别地,In particular,
每个Re独立地选自H、甲基、乙基、正丙基、异丙基、环丙基、 Each Re is independently selected from H, methyl, ethyl, n-propyl, isopropyl, cyclopropyl,
在本发明一些实施方案中,在式(I)、(II)、(III)、(IV)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物中,In some embodiments of the present invention, in the formula (I), (II), (III), (IV) compound or its isotope labeled compound, or its optical isomer, geometric isomer, tautomer or Isomer mixtures, or pharmaceutically acceptable salts thereof, or prodrugs thereof, or metabolites thereof,
R1各自独立地选自H、卤素、C1-C3烷基磺酰基、C1-C3烷基、C1-C3卤代烷基或C1-C3烷氧基,优选地选自H、氟、氯、溴、甲基、乙基、正丙基、异丙基或甲磺酰基;和/或Each R 1 is independently selected from H, halogen, C 1 -C 3 alkylsulfonyl, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl or C 1 -C 3 alkoxy, preferably selected from H, fluorine, chlorine, bromine, methyl, ethyl, n-propyl, isopropyl or methanesulfonyl; and/or
R2选自H或C1-C3烷基,优选地选自H或甲基;和/或R is selected from H or C 1 -C 3 alkyl, preferably from H or methyl; and/or
R3选自H或C1-C3烷基,优选地选自H或甲基。R 3 is selected from H or C 1 -C 3 alkyl, preferably from H or methyl.
在本发明一些实施方案中,在式(I)、(II)、(III)、(IV)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物中,In some embodiments of the present invention, in the formula (I), (II), (III), (IV) compound or its isotope labeled compound, or its optical isomer, geometric isomer, tautomer or Isomer mixtures, or pharmaceutically acceptable salts thereof, or prodrugs thereof, or metabolites thereof,
Ra各自独立地选自H、卤素、C1-C3烷基、C1-C3卤代烷基、C1-C3烷氧基、C3-C4环烷基或氰基,其中所述C1-C3烷基、C1-C3烷氧基或C3-C4环烷基各自独立地任选被卤素、羟基、氰基、C1-C3烷基或C1-C3烷氧基取代,Ra优选地选自H、氟、氯、溴、甲基、乙基、异丙基、环丙基、二氟甲基、三氟甲基、三氟乙基、甲氧基、乙氧基、羟甲基、1,1-二甲基羟甲基、氰基或甲氧基亚甲基;和/或Each R a is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, C 1 -C 3 alkoxy, C 3 -C 4 cycloalkyl or cyano, wherein The C 1 -C 3 alkyl, C 1 -C 3 alkoxy or C 3 -C 4 cycloalkyl are each independently optionally replaced by halogen, hydroxyl, cyano, C 1 -C 3 alkyl or C 1 - C3alkoxy substitution, R is preferably selected from H, fluorine, chlorine, bromine, methyl, ethyl, isopropyl, cyclopropyl, difluoromethyl, trifluoromethyl, trifluoroethyl, Methoxy, ethoxy, hydroxymethyl, 1,1-dimethylhydroxymethyl, cyano, or methoxymethylene; and/or
Rb各自独立地选自H、卤素、C1-C3烷基、C3-C4环烷基或氰基,其中所述C1-C3烷基或C3-C4环烷基各自独立地任选被卤素、羟基、氨基、氰基或C1-C3烷基取代,Rb优选地选自H、氟、氯、溴、甲基、乙基、异丙基、环丙基、二氟甲基、三氟甲基、三氟乙基或氰基;和/或Each R b is independently selected from H, halogen, C 1 -C 3 alkyl, C 3 -C 4 cycloalkyl or cyano, wherein the C 1 -C 3 alkyl or C 3 -C 4 cycloalkyl Each is independently optionally substituted by halogen, hydroxyl, amino, cyano or C 1 -C 3 alkyl, R b is preferably selected from H, fluorine, chlorine, bromine, methyl, ethyl, isopropyl, cyclopropyl group, difluoromethyl, trifluoromethyl, trifluoroethyl or cyano; and/or
Rc各自独立地选自H、卤素或C1-C3烷基,优选地选自H、氟、氯、甲基或乙基;和/或Each R c is independently selected from H, halogen or C 1 -C 3 alkyl, preferably selected from H, fluorine, chlorine, methyl or ethyl; and/or
Rd各自独立地选自H、卤素、C1-C3烷基或氰基,优选地选自H、氟、氯、溴、甲基、乙基、正丙基、异丙基或氰基。Each R d is independently selected from H, halogen, C 1 -C 3 alkyl or cyano, preferably selected from H, fluorine, chlorine, bromine, methyl, ethyl, n-propyl, isopropyl or cyano .
在本发明一些实施方案中,在式(I)、(II)、(III)、(IV)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物中,In some embodiments of the present invention, in the formula (I), (II), (III), (IV) compound or its isotope labeled compound, or its optical isomer, geometric isomer, tautomer or Isomer mixtures, or pharmaceutically acceptable salts thereof, or prodrugs thereof, or metabolites thereof,
Z1为2;和/或Z 1 is 2; and/or
Z2为0或1;和/或 Z2 is 0 or 1; and/or
Z3为0或1;和/或 Z3 is 0 or 1; and/or
Z4为1;和/或 Z 4 is 1; and/or
每个n独立地为0或1。Each n is independently 0 or 1.
本发明典型的式(I)、(II)、(III)、(IV)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物包括但不限于:



Typical compounds of formula (I), (II), (III), (IV) of the present invention or their isotope-labeled compounds, or their optical isomers, geometric isomers, tautomers or isomer mixtures, Or its pharmaceutically acceptable salt, or its prodrug, or its metabolites include but not limited to:



为了简明起见,后文所述“式(I)、(II)、(III)、(IV)化合物”或“本发明的化合物”也可以涵盖式(I)、(II)、(III)、(IV)化合物的任意同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物。For the sake of brevity, the "compound of formula (I), (II), (III), (IV)" or "compound of the present invention" described hereinafter may also cover formula (I), (II), (III), (IV) Any isotope-labeled compound of the compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its Metabolites.
术语“光学异构体”意指,当化合物具有一个或更多个手性中心时,每个手性中心可以存在R构型或S构型,由此构成的各种异构体为光学异构体。光学异构体包括所有的非对映异构体、对映异构体、内消旋体、外消旋体或其混合物形式。例如,通过手性色谱柱或通过手性合成可以分离光学异构体。The term "optical isomer" means that when a compound has one or more chiral centers, each chiral center can have an R configuration or an S configuration, and the various isomers thus constituted are optical isomers. Construct. Optical isomers include all diastereoisomers, enantiomers, mesoforms, racemates or mixtures thereof. For example, optical isomers can be separated by chiral chromatographic columns or by chiral synthesis.
术语“几何异构体”意指,当化合物中存在双键时,该化合物可以存在顺式异构体、反式异构体、E型异构体和Z型异构体。几何异构体包括顺式异构体、反式异构体、E型异构体、Z型异构体或其混合物形式。The term "geometric isomer" means that when a double bond exists in the compound, the compound may exist as a cis-isomer, a trans-isomer, an E-isomer and a Z-isomer. Geometric isomers include cis isomers, trans isomers, E isomers, Z isomers, or mixtures thereof.
术语“互变异构体”指因分子中某一原子在两个位置迅速移动而产生的异构体。本领域技术人员可以理解:互变异构体之间可以互相转变,在某一状态下可能会达到一种平衡状态而共存。The term "tautomer" refers to isomers that result from the rapid movement of an atom in a molecule between two positions. Those skilled in the art can understand that tautomers can transform into each other, and may reach an equilibrium state and coexist in a certain state.
除非另有指明,本文提到“式(I)化合物”或“本发明的化合物”时也涵盖该化合物中任一个原子被其同位素原子代替而得到的同位素标记化合物。本发明包括式(I)化合物的所有药学上可接受的同位素标记化合物,其中,一个或者多个原子被具有与通常在自然界中所发现的原子相同原子序数但是不同原子质量或者质量数的原子所替换。Unless otherwise specified, references herein to "compounds of formula (I)" or "compounds of the present invention" also encompass isotopically labeled compounds in which any atom in the compound is replaced by its isotopic atom. The present invention includes all pharmaceutically acceptable isotope-labeled compounds of compounds of formula (I) in which one or more atoms are replaced by atoms having the same atomic number but a different atomic mass or mass number as atoms normally found in nature. replace.
适用于包含在本发明的化合物中的同位素的实例包括氢的同位素,诸如2H(D)和3H(T),碳的同位素,诸如11C、13C和14C,氯的同位素,诸如36Cl,氟的同位素,诸如18F,碘的同位素,诸如123I和125I,氮的同位素,诸如13N和15N,氧的同位素,诸如15O、17O和18O,以及硫的同位素,诸如35S。Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2 H(D) and 3 H(T), isotopes of carbon, such as 11 C, 13 C and 14 C, isotopes of chlorine, such as 36 Cl, isotopes of fluorine such as 18 F, isotopes of iodine such as 123 I and 125 I, isotopes of nitrogen such as 13 N and 15 N, isotopes of oxygen such as 15 O, 17 O and 18 O, and of sulfur Isotopes such as35S .
式(I)、(II)、(III)、(IV)的同位素标记化合物一般可以通过本领域技术人员已知的常规技术或者通过使用合适的同位素标记试剂代替先前使用的非标记试剂以类似于在本文所附的实例和制备中所描述的方法,来进行制备。Isotope-labeled compounds of formula (I), (II), (III), (IV) can generally be prepared by conventional techniques known to those skilled in the art or by using a suitable isotope-labeled reagent instead of a previously used non-labeled reagent The preparation was carried out as described in the Examples and Preparations appended hereto.
式(I)、(II)、(III)、(IV)化合物可以药学上可接受的盐的形式存在,比如,式(I)、(II)、(III)、(IV)化合物的酸加成盐和/或碱加成盐。除非另有指明,否则本文所用的“药学上可接受的盐”包括可出现于式(I)、(II)、(III)、(IV)化合物内的酸加成盐或碱加成盐。Formula (I), (II), (III), (IV) compound can exist in the form of pharmaceutically acceptable salt, for example, the acid addition of formula (I), (II), (III), (IV) compound Salt formation and/or base addition salts. As used herein, unless otherwise indicated, "pharmaceutically acceptable salt" includes acid addition salts or base addition salts that may occur in compounds of formula (I), (II), (III), (IV).
式(I)、(II)、(III)、(IV)化合物的药学上可接受的盐类包括其酸加 成盐和碱加成盐。适当的酸加成盐是由形成无毒性盐的酸所形成的。其实例包括但不限于:乙酸盐、己二酸盐、天冬氨酸盐、苯甲酸盐、苯磺酸盐、碳酸氢盐/碳酸盐、硫酸氢盐/硫酸盐、硼酸盐、樟脑磺酸盐、柠檬酸盐、环己胺磺酸盐、乙二磺酸盐、甲酸盐、反丁烯二酸盐、葡萄庚糖酸盐、葡萄糖酸盐、葡萄糖醛酸盐、六氟磷酸盐、2-(4-羟苄基)苯甲酸盐、氢氯化物/氯化物、氢溴化物/溴化物、氢碘化物/碘化物、2-羟乙磺酸盐、乳酸盐、苹果酸盐、顺丁烯二酸盐、丙二酸盐、甲磺酸盐、甲基硫酸盐、萘酸盐、2-萘磺酸盐、烟碱酸盐、硝酸盐、乳清酸盐、草酸盐、十六酸盐、磷酸盐/磷酸氢盐/磷酸二氢盐、焦谷氨酸盐、葡萄糖二酸盐、硬脂酸盐、水杨酸盐、单宁酸盐、酒石酸盐、甲苯磺酸盐和三氟乙酸盐。适当的碱加成盐是由形成无毒性盐的碱所形成的。其实例包括但不限于:铝、精氨酸、钙、胆碱、二乙胺、二乙醇胺、甘氨酸、赖氨酸、镁、葡甲胺、乙醇胺、钾、钠、氨丁三醇和锌盐。还可形成酸和碱的半盐,例如半硫酸盐和半钙盐。关于合适的盐的综述,参见Handbook of Pharmaceutical Salts:Properties,Selection and Use by Stahl and Wermuth(Wiley-VCH,2002)。用于制备本文中所述的化合物的药学上可接受的盐的方法是本领域技术人员已知的。The pharmaceutically acceptable salts of the compound of formula (I), (II), (III), (IV) include its acid plus Salt and base addition salt. Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include, but are not limited to: acetate, adipate, aspartate, benzoate, benzenesulfonate, bicarbonate/carbonate, bisulfate/sulfate, borate , camphorsulfonate, citrate, cyclamate, ethanedisulfonate, formate, fumarate, glucoheptonate, gluconate, glucuronate, six Fluorophosphate, 2-(4-Hydroxybenzyl)benzoate, Hydrochloride/Chloride, Hydrobromide/Bromide, Hydroiodide/Iodide, 2-Isethionate, Lactate , malate, maleate, malonate, methanesulfonate, methylsulfate, naphthenate, 2-naphthalenesulfonate, nicotine, nitrate, orotate , Oxalate, Cetate, Phosphate/Hydrogen Phosphate/Dihydrogen Phosphate, Pyroglutamate, Gluconate, Stearate, Salicylate, Tannin, Tartrate , tosylate and trifluoroacetate. Suitable base addition salts are formed from bases which form non-toxic salts. Examples include, but are not limited to: aluminum, arginine, calcium, choline, diethylamine, diethanolamine, glycine, lysine, magnesium, meglumine, ethanolamine, potassium, sodium, tromethamine, and zinc salts. Half-salts of acids and bases, such as the hemisulfate and hemicalcium salts, may also be formed. For a review of suitable salts, see Handbook of Pharmaceutical Salts: Properties, Selection and Use by Stahl and Wermuth (Wiley-VCH, 2002). Methods for preparing pharmaceutically acceptable salts of the compounds described herein are known to those skilled in the art.
本发明的某些化合物可以以非溶剂化形式以及溶剂化形式(包括水合形式)存在。一般而言,式(I)、(II)、(III)、(IV)化合物无论以溶剂化形式存在或以未溶剂化形式存在,其都包括在本发明的范围内。Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, compounds of formula (I), (II), (III), (IV) are included within the scope of the present invention whether they exist in solvated or unsolvated form.
本发明的某些化合物可以不同晶型或不定型形式存在,无论以何种形式存在,式(I)、(II)、(III)、(IV)化合物都包括在本发明的范围内。Certain compounds of the present invention may exist in different crystalline or amorphous forms, and no matter in which form they exist, the compounds of formula (I), (II), (III) and (IV) are included within the scope of the present invention.
为了避免歧义,下面对本文中所使用的术语给出定义。除非另有说明,本文所用术语的含义如下。To avoid ambiguity, definitions of terms used herein are given below. Unless otherwise specified, the meanings of the terms used herein are as follows.
术语“药学上可接受的”是指相应的化合物、载体或分子适于给予人。优选地,该术语是指由管理机构例如CFDA(中国)、EMEA(欧洲)、FDA(美国)等任意国家管理机构认证的用于哺乳动物优选人。The term "pharmaceutically acceptable" means that the corresponding compound, carrier or molecule is suitable for administration to a human. Preferably, the term refers to the use in mammals, preferably humans, certified by any national regulatory agency such as CFDA (China), EMEA (Europe), FDA (USA), etc.
“前体药”是指通过与酶、胃酸等在生理条件下在活体内例如通过各自在酶催化下进行的氧化、还原、水解等反应转化为本发明化合物的衍生物。"Prodrug" refers to a derivative that is converted into the compound of the present invention by reacting with enzymes, gastric acid, etc. under physiological conditions in vivo, for example, by oxidation, reduction, hydrolysis, etc. catalyzed by enzymes.
“代谢物”是指在细胞或有机体优选人中源自本发明任意化合物的所有分子。"Metabolite" refers to all molecules derived from any compound of the invention in a cell or organism, preferably a human.
术语“羟基”是指-OH;术语“氨基”是指-NH2;术语“硝基”是指-NO2;并且术语“氰基”是指-CN;术语“羧基”是指-COOH。The term "hydroxyl" refers to -OH; the term "amino" refers to -NH2 ; the term "nitro" refers to -NO2 ; and the term "cyano" refers to -CN; the term "carboxy" refers to -COOH.
术语“酰基”是指-C(=O)-;术语“酰胺基”是指-C(=O)-NH2;术语“磺酰基”是指-S(=O)2-;并且术语“磺酰胺基”是指-S(=O)2-NH2The term "acyl" refers to -C(=O)-; the term "amido" refers to -C(=O) -NH2 ; the term "sulfonyl" refers to -S(=O) 2- ; and the term ""Sulfonamido" means -S(=O) 2 -NH2 .
在本文中使用时,术语“被取代”是指基团中的一个或多个(优选1至5个,更优选1至3个,甚至更有效1个或2个)氢原子独立地被相应数目的取代基所代替。As used herein, the term "substituted" means that one or more (preferably 1 to 5, more preferably 1 to 3, even more effectively 1 or 2) hydrogen atoms in a group are independently replaced by the corresponding number of substituents are substituted.
在本文中使用时,术语“各自独立地”是指当取代基的个数超过一个时,这 些取代基可以相同也可以不同。When used herein, the term "independently" means that when the number of substituents is more than one, the These substituents may be the same or different.
在本文中使用时,术语“任选”或“任选地”表示其所描述的事件可以发生或不发生。例如,一个基团“任选地被取代”表示:该基团可以是未被取代的,也可以是被取代的。As used herein, the term "optional" or "optionally" means that the event it describes can or cannot occur. For example, a group "optionally substituted" means that the group can be unsubstituted or substituted.
在本文中使用时,术语“烷基”是指饱和的脂族烃,包括直链及支链。在一些实施方案中,烷基基团具有1-8个、或1-6个、或1-3个碳原子。例如,术语“C1-8烷基”是指具有1-8个碳原子的直链或支链原子团,术语“C1-6烷基”是指具有1-6个碳原子的直链或支链原子团,术语“C1-3烷基”是指具有1-3个碳原子的直链或支链原子团。术语“C1-8烷基”在其定义中包括术语“C1-6烷基”、“C1-C3烷基”和“C1-C4烷基”。烷基的实例包括但不限于甲基、乙基、丙基、异丙基、丁基、异丁基、仲丁基、叔丁基、戊基、2-戊基、3-戊基、异戊基、新戊基、(R)-2-甲基丁基、(S)-2-甲基丁基、3-甲基丁基、2,3-二甲基丙基、2,3-二甲基丁基、己基等。烷基基团可任选地被一或多个(例如,1至5个,优选1至3个,甚至更有效1个或2个)适当的取代基所取代。As used herein, the term "alkyl" refers to saturated aliphatic hydrocarbons, including straight and branched chains. In some embodiments, an alkyl group has 1-8, or 1-6, or 1-3 carbon atoms. For example, the term "C 1-8 alkyl" refers to a straight-chain or branched chain radical having 1-8 carbon atoms, and the term "C 1-6 alkyl" refers to a straight-chain or branched chain having 1-6 carbon atoms. A branched chain radical, the term "C 1-3 alkyl" refers to a straight chain or branched chain radical having 1 to 3 carbon atoms. The term "C 1-8 alkyl" includes within its definition the terms "C 1-6 alkyl", "C 1 -C 3 alkyl" and "C 1 -C 4 alkyl". Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, 2-pentyl, 3-pentyl, iso Pentyl, neopentyl, (R)-2-methylbutyl, (S)-2-methylbutyl, 3-methylbutyl, 2,3-dimethylpropyl, 2,3- Dimethylbutyl, hexyl, etc. Alkyl groups may optionally be substituted with one or more (eg, 1 to 5, preferably 1 to 3, even more effectively 1 or 2) suitable substituents.
在本文中使用时,术语“卤代烷基”是指具有一或多个卤素取代基的烷基基团(至多全卤代烷基,即,烷基基团的每个氢原子均被卤素原子所取代)。例如,术语“C1-C6卤代烷基”是指具有一或多个卤素取代基的C1-C6烷基基团(至多全卤代烷基,即,烷基基团的每个氢原子均被卤素原子所取代)。另举一例,术语“C1-C4卤代烷基”是指具有一或多个卤素取代基的C1-C4烷基基团(至多全卤代烷基,即,烷基基团的每个氢原子均被卤素原子所取代);术语“C1-C3卤代烷基”是指具有一或多个卤素取代基的C1-C3烷基基团(至多全卤代烷基,即,烷基基团的每个氢原子均被卤素原子所取代);且术语“C1-C2卤代烷基”是指具有一或多个卤素取代基的C1-C2烷基基团(即,甲基或乙基)(至多全卤代烷基,即,烷基基团的每个氢原子均被卤素原子所取代)。再另举一例,术语“C1卤代烷基”是指具有1、2或3个卤素取代基的甲基基团。卤代烷基基团的例子包括:CF3、C2F5、CHF2、CH2F、CH2CF3、CH2Cl等。As used herein, the term "haloalkyl" refers to an alkyl group having one or more halogen substituents (up to perhaloalkyl, i.e., each hydrogen atom of the alkyl group is replaced by a halogen atom) . For example, the term "C 1- C 6 haloalkyl" refers to a C 1- C 6 alkyl group (up to perhaloalkyl, i.e., each hydrogen atom of the alkyl group has one or more halogen substituents replaced by halogen atoms). As another example, the term "C 1- C 4 haloalkyl" refers to a C 1- C 4 alkyl group having one or more halogen substituents (up to perhaloalkyl, i.e., each hydrogen of the alkyl group atoms are all replaced by halogen atoms); the term "C 1- C 3 haloalkyl" refers to a C 1- C 3 alkyl group (up to perhaloalkyl, i.e., alkyl each hydrogen atom of the group is replaced by a halogen atom); and the term "C 1- C 2 haloalkyl" refers to a C 1- C 2 alkyl group having one or more halogen substituents (i.e., methyl or ethyl) (up to perhaloalkyl, ie, each hydrogen atom of the alkyl group is replaced by a halogen atom). As yet another example, the term "C haloalkyl" refers to a methyl group having 1, 2 or 3 halo substituents . Examples of haloalkyl groups include: CF3 , C2F5 , CHF2 , CH2F , CH2CF3 , CH2Cl , and the like.
在本文中使用时,术语“亚烷基”是指二价烷基,其中烷基如上所定义。所述亚烷基优选具有1-6个碳原子的亚烷基(即C1-C6亚烷基),更优选具有1-3个碳原子的亚烷基(即C1-C3亚烷基)。亚烷基的例子包括但不限于-CH2-、-CH(CH3)-、-C(CH3)2-、-CH2CH2-、-CH(CH2CH3)-、-CH2CH(CH3)-、-CH2C(CH3)2-、-CH2CH2CH2-、-CH2CH2CH2CH2-等。As used herein, the term "alkylene" refers to a divalent alkyl group, wherein alkyl is as defined above. The alkylene group is preferably an alkylene group having 1-6 carbon atoms (i.e. C 1- C 6 alkylene group), more preferably an alkylene group having 1-3 carbon atoms (i.e. C 1- C 3 alkylene group alkyl). Examples of alkylene groups include, but are not limited to -CH2- , -CH( CH3 ) -, -C( CH3 ) 2- , -CH2CH2- , -CH( CH2CH3 )-, -CH 2 CH(CH 3 )-, -CH 2 C(CH 3 ) 2 -, -CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 -, etc.
在本文中使用时,术语“n元环烷基”是指具有n个碳原子的全碳环系统。术语“C3-C6环烷基”是指具有3-6个碳原子的全碳环系统,“C3-C4环烷基”是指具有3-4个碳原子的全碳环系统。C3-C6环烷基的例子包括但不限于环丙基、环丁基、环戊基、环戊烯基、环己基。此外,环烷基可任选地被一或多个适当的取代基所取代。 As used herein, the term "n-membered cycloalkyl" refers to an all-carbocyclic ring system having n carbon atoms. The term "C 3 -C 6 cycloalkyl" refers to a fully carbocyclic ring system having 3-6 carbon atoms, and "C 3 -C 4 cycloalkyl" refers to a fully carbocyclic ring system having 3-4 carbon atoms . Examples of C 3 -C 6 cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl. In addition, cycloalkyl groups may be optionally substituted with one or more suitable substituents.
术语“亚环烷基”指二价环烷基,其中环烷基如上所定义。The term "cycloalkylene" refers to a divalent cycloalkyl group, wherein cycloalkyl is as defined above.
在本文中使用时,术语“n元杂环基”是指具有m个形成环的碳原子和(n-m)个形成环的杂原子的杂环烷基,所述杂原子选自O、S、S(O)2及N。例如,4-7元杂环基包括但不限于氧杂环丁烷、硫杂环丁烷、氮杂环丁烷、四氢呋喃、四氢噻吩、吡咯烷、四氢吡喃、四氢噻喃、哌啶、吗啉、哌嗪、氧杂环庚烷、硫杂环庚烷、氮杂环庚烷、1,1-二氧代硫杂环丁烷。此外,杂环基可任选地被一或多个适当的取代基所取代。As used herein, the term "n-membered heterocyclyl" refers to a heterocycloalkyl group having m ring-forming carbon atoms and (nm) ring-forming heteroatoms selected from O, S, S(O) 2 and N. For example, 4-7 membered heterocyclic groups include, but are not limited to, oxetane, thietane, azetidine, tetrahydrofuran, tetrahydrothiophene, pyrrolidine, tetrahydropyran, tetrahydrothiopyran, Piperidine, morpholine, piperazine, oxepane, thiepane, azepane, 1,1-dioxothietane. In addition, heterocyclyl may be optionally substituted with one or more suitable substituents.
术语“亚杂环基”指二价杂环基,其中杂环基如上所定义。The term "heterocyclylene" refers to a divalent heterocyclyl group, wherein heterocyclyl is as defined above.
在本文中使用时,术语“n元杂芳基”是指具有m个形成芳环的碳原子和(n-m)个形成芳环的杂原子的杂芳基,所述杂原子选自O、S及N。例如,5-6元杂芳基包括但不限于吡嗪、吡唑、吡咯、呋喃、噻吩、噻唑、吡啶。此外,杂芳基可任选地被一或多个适当的取代基所取代。As used herein, the term "n-membered heteroaryl" refers to a heteroaryl group having m carbon atoms forming an aromatic ring and (n-m) heteroatoms forming an aromatic ring selected from O, S and N. For example, 5-6 membered heteroaryl groups include, but are not limited to, pyrazine, pyrazole, pyrrole, furan, thiophene, thiazole, pyridine. In addition, heteroaryl groups may be optionally substituted with one or more suitable substituents.
本文中,与取代基个数、碳原子个数、环原子个数相关的数目范围表示该范围内所有整数的逐个列举,而范围仅是作为一种简化的表示法。例如:“1-4个取代基”表示1、2、3或4个取代基;“3-8个环原子”表示3个、4个、5个、6个、7个或8个环原子。因此,与取代基个数、碳原子个数、环原子个数相关的数目范围也涵盖其任意一个子范围,且每一个子范围也视为被本文公开。Herein, the number ranges related to the number of substituents, the number of carbon atoms, and the number of ring atoms represent enumeration of all integers within the range one by one, and the range is only used as a simplified representation. For example: "1-4 substituents" means 1, 2, 3 or 4 substituents; "3-8 ring atoms" means 3, 4, 5, 6, 7 or 8 ring atoms . Therefore, the number ranges related to the number of substituents, the number of carbon atoms, and the number of ring atoms also cover any sub-range thereof, and each sub-range is also deemed to be disclosed herein.
本发明化合物可按有机合成领域技术人员已知的多种方式制备。本领域技术人员可以参照本发明具体实施例的具体化合物的合成路线,对反应原料和反应条件进行适当调整而得到其它化合物的合成方法。The compounds of the present invention can be prepared in a variety of ways known to those skilled in the art of organic synthesis. Those skilled in the art can refer to the synthetic routes of the specific compounds in the specific examples of the present invention, and make appropriate adjustments to the reaction raw materials and reaction conditions to obtain the synthetic methods of other compounds.
在第二方面,本发明提供了一种药物组合物,其包含式(I)、(II)、(III)、(IV)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,以及药学上可接受的载体。In a second aspect, the present invention provides a pharmaceutical composition comprising a compound of formula (I), (II), (III), (IV) or its isotope-labeled compound, or its optical isomers, geometric isomers isomer, tautomer or isomer mixture, or a pharmaceutically acceptable salt thereof, or a prodrug thereof, or a metabolite thereof, and a pharmaceutically acceptable carrier.
药学上可接受的载体可以是有机或无机惰性载体材料,例如,合适的载体包括水、明胶、阿拉伯树胶、乳糖、淀粉、硬脂酸镁、滑石、植物油、聚亚烷基二醇、凡士林、甘露醇、纤维素、纤维素衍生物、糖精钠、葡萄糖、蔗糖、碳酸镁、盐水、甘油、乙醇等。此外,药物组合物还可含有其他药物添加剂,例如调味剂、防腐剂、稳定剂、乳化剂、缓冲剂、稀释剂、黏合剂、润湿剂、崩解剂、润滑剂、助流剂等。Pharmaceutically acceptable carriers can be organic or inorganic inert carrier materials, for example, suitable carriers include water, gelatin, gum arabic, lactose, starch, magnesium stearate, talc, vegetable oils, polyalkylene glycols, petrolatum, Mannitol, cellulose, cellulose derivatives, sodium saccharin, glucose, sucrose, magnesium carbonate, saline, glycerin, ethanol, etc. In addition, the pharmaceutical composition may also contain other pharmaceutical additives, such as flavoring agents, preservatives, stabilizers, emulsifiers, buffers, diluents, binders, wetting agents, disintegrants, lubricants, glidants and the like.
本发明的药物组合物的剂型可以是液体剂型、固体剂型或半固体剂型。液体剂型可以是溶液剂(包括真溶液和胶体溶液)、乳剂(包括o/w型、w/o型和复乳)、混悬剂、注射剂(包括水针剂、粉针剂和输液)、滴眼剂、滴鼻剂、洗剂和搽剂等;固体剂型可以是片剂(包括普通片、肠溶片、含片、分散片、咀嚼片、泡腾片、口腔崩解片)、胶囊(包括硬胶囊、软胶囊、肠溶胶囊)、颗粒剂、散剂、丸剂、栓剂、膜剂、贴片、气雾剂、喷雾剂等;半固体剂型可以是软膏剂、 凝胶剂、糊剂等。本发明的药物组合物可以制成普通制剂、也制成是缓释制剂、控释制剂、靶向制剂及各种微粒给药系统。The dosage form of the pharmaceutical composition of the present invention may be a liquid dosage form, a solid dosage form or a semi-solid dosage form. Liquid dosage forms can be solutions (including true solutions and colloid solutions), emulsions (including o/w type, w/o type and double emulsion), suspensions, injections (including aqueous injections, powder injections and infusion solutions), eye drops Agents, nasal drops, lotions and liniments, etc.; solid dosage forms can be tablets (including ordinary tablets, enteric-coated tablets, buccal tablets, dispersible tablets, chewable tablets, effervescent tablets, orally disintegrating tablets), capsules (including hard capsules, soft capsules, enteric-coated capsules), granules, powders, pills, suppositories, films, patches, aerosols, sprays, etc.; semi-solid dosage forms can be ointments, Gels, pastes, etc. The pharmaceutical composition of the present invention can be made into common preparations, sustained-release preparations, controlled-release preparations, targeted preparations and various particle delivery systems.
在一些实施方案中,所述药物组合物的剂型选自片剂、颗粒剂、散剂、糖浆剂、吸入剂和注射剂。In some embodiments, the dosage form of the pharmaceutical composition is selected from tablet, granule, powder, syrup, inhalation and injection.
用于口服的固体剂型可以包括胶囊、片剂、丸剂、粉末和颗粒。在这种固体剂型中,将活性化合物与至少一种惰性赋型剂(或载体)(例如,柠檬酸钠或磷酸二钙)混合,其中还可以包括:(a)填料或混合剂(例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸);(b)粘结剂(例如,羧基甲基纤维素、褐藻酸酯、凝胶、聚乙烯基吡咯烷酮、蔗糖和阿拉伯树胶);(c)保湿剂(例如,丙三醇);(d)崩解剂(例如,琼脂-琼脂、碳酸钙、马铃薯或木薯淀粉、褐藻酸、某些合成的硅酸酯、碳酸钠);(e)溶液阻滞剂(例如,石蜡);(f)吸收促进剂(例如,季铵化合物);(g)润湿剂(例如,十六烷醇和单硬脂酸丙三醇酯);(h)吸附剂(例如,高岭土和斑脱土)和(i)润滑剂(例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、月桂基硫酸钠)或其混合物混合。Solid dosage forms for oral administration may include capsules, tablets, pills, powders and granules. In such solid dosage forms, the active compound is mixed with at least one inert excipient (or carrier) (for example, sodium citrate or dicalcium phosphate), which may also include: (a) fillers or mixing agents (for example, starch, lactose, sucrose, glucose, mannitol, and silicic acid); (b) binders (e.g., carboxymethylcellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose, and gum arabic); (c) Moisturizers (eg, glycerol); (d) disintegrants (eg, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain synthetic silicates, sodium carbonate); (e) solutions Blocking agents (e.g., paraffin); (f) absorption enhancers (e.g., quaternary ammonium compounds); (g) wetting agents (e.g., cetyl alcohol and glyceryl monostearate); (h) adsorbents (eg, kaolin and bentonite) and (i) lubricants (eg, talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium lauryl sulfate) or mixtures thereof.
适于肠胃外施用的制剂例如注射剂可以包括适于注射的水性和非水性等渗无菌溶液,以及水性和非水性无菌混悬剂。本文提供的肠胃外制剂任选地包含在单位剂量或多剂量密封容器(例如安瓿)中,并且可以储存在仅需要于即将使用前添加无菌液体载体(例如注射用水)的冷冻干燥(冻干)条件下。用于重构药物组合物(例如在注射前)的合适稀释剂的实例包括抑菌性注射用水、5%葡萄糖水溶液、磷酸盐缓冲盐水、林格氏(Ringer's)溶液、盐水、无菌水、去离子水及其组合。Formulations suitable for parenteral administration, such as injections, may include aqueous and nonaqueous isotonic sterile solutions suitable for injection, as well as aqueous and nonaqueous sterile suspensions. The parenteral formulations provided herein are optionally contained in unit-dose or multi-dose sealed containers, such as ampoules, and can be stored in freeze-dried (lyophilized) containers that require only the addition of a sterile liquid carrier (such as water for injection) immediately before use. ) conditions. Examples of suitable diluents for reconstitution of the pharmaceutical composition (eg, prior to injection) include bacteriostatic water for injection, 5% dextrose in water, phosphate buffered saline, Ringer's solution, saline, sterile water, Deionized water and combinations thereof.
喷雾剂可含有赋形剂,如乳糖、滑石、硅酸、氢氧化铝、硅酸钙和聚酰胺粉剂,或这些物质的混合物。喷雾剂可另外含有常规推进剂,例如氯氟烃和挥发性未经取代的烃,例如丁烷和丙烷。吸入剂可包含赋形剂如乳糖,或是包含如聚环氧乙烷-9-月桂基醚,甘氨胆酸盐和脱氧胆酸盐的含水溶液,或是油性溶液以鼻滴剂或喷雾,或凝胶形式施用。Sprays can contain excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane. Inhalants may contain excipients such as lactose, or aqueous solutions containing, for example, polyethylene oxide-9-lauryl ether, glycocholate and deoxycholate, or oily solutions as nasal drops or spray , or in gel form.
本发明的化合物在其药物组合物中的含量可以根据实际需要(例如剂型、施用方式、施用对象等)进行调整,例如为0.1-95重量%,例如1-95重量%,5-90重量%,10-80重量%等。The content of the compound of the present invention in its pharmaceutical composition can be adjusted according to actual needs (such as dosage form, administration method, administration object, etc.), such as 0.1-95% by weight, such as 1-95% by weight, 5-90% by weight , 10-80% by weight, etc.
具体地,本发明的药物组合物中可以特别地包含0.01-10g(例如0.05g、0.1g、0.5g、1g或5g等)的本发明的化合物。Specifically, 0.01-10 g (eg, 0.05 g, 0.1 g, 0.5 g, 1 g or 5 g, etc.) of the compound of the present invention may be included in the pharmaceutical composition of the present invention.
在第三方面,本发明涉及式(I)、(II)、(III)、(IV)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物在制备用于治疗或预防由CCR4介导的疾病或病症的作为CCR4拮抗剂的药物中的用途。式(I)、(II)、(III)、(IV)化合物或其同位素标记化合物、或其光学异构体、几何异构体、 互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物可以用于在有需要的受试者中治疗或预防由CCR4介导的疾病或病症。In a third aspect, the present invention relates to compounds of formula (I), (II), (III), (IV) or isotopically labeled compounds thereof, or optical isomers, geometric isomers, tautomers or isomers thereof Use of a mixture of conformers, or a pharmaceutically acceptable salt thereof, or a prodrug thereof, or a metabolite thereof in the preparation of a medicament for treating or preventing a disease or condition mediated by CCR4 as a CCR4 antagonist. Compounds of formula (I), (II), (III), (IV) or their isotope-labeled compounds, or their optical isomers, geometric isomers, Tautomers or mixtures of isomers, or pharmaceutically acceptable salts thereof, or prodrugs thereof, or metabolites thereof may be used to treat or prevent diseases mediated by CCR4 in a subject in need thereof or illness.
本发明使用的术语“受试者”是指可潜在地从用式(I)、(II)、(III)、(IV)化合物进行的治疗中受益的任何人类或非人类生物体。示例性受试者包括任何年龄的人类或哺乳动物。优选地,所述受试者是人。The term "subject" as used herein refers to any human or non-human organism that could potentially benefit from treatment with a compound of formula (I), (II), (III), (IV). Exemplary subjects include humans or mammals of any age. Preferably, the subject is a human.
本文使用的术语“治疗”包括在哺乳动物、尤其人类中治疗疾病或病症且包括:(a)抑制感染、疾病或病症,即遏制或延缓感染、疾病或病症的发展;(b)缓解感染、疾病或病症,即引起疾病或病症的消退,和/或(c)感染、疾病或病症的治愈。The term "treatment" as used herein includes treating a disease or condition in a mammal, especially a human and includes: (a) inhibiting an infection, disease or condition, i.e. arresting or delaying the development of an infection, disease or condition; (b) alleviating an infection, disease or condition; A disease or condition, ie causing regression of the disease or condition, and/or (c) cure of the infection, disease or condition.
本文使用的术语“预防”包括在哺乳动物、尤其人类中进行预防性疗法以旨在降低感染、疾病或病症发生的可能性。可以根据与一般群体相比感染或患有疾病或病症的风险增加为因素来选择接受预防性疗法的患者。“预防”可以包括对尚未呈现感染或临床病况的受试者进行处置,和预防相同或类似感染或临床病况的第二次出现。As used herein, the term "prevention" includes prophylactic therapy in mammals, especially humans, aimed at reducing the likelihood of an infection, disease or condition occurring. Patients may be selected for prophylactic therapy based on an increased risk of infection or having a disease or condition compared to the general population as a factor. "Preventing" can include treating a subject who has not yet exhibited an infection or clinical condition, and preventing a second occurrence of the same or similar infection or clinical condition.
发明人发现,本发明的化合物能够实现对CCR4介导的细胞迁移的抑制。因此,本发明的化合物可以用于预防或治疗由CCR4介导的疾病或病症。The inventors found that the compounds of the present invention can inhibit CCR4-mediated cell migration. Therefore, the compounds of the present invention can be used in the prevention or treatment of diseases or conditions mediated by CCR4.
在一些实施方案中,所述由CCR4介导的疾病或病症可以选自特应性皮炎、哮喘、过敏性鼻炎、异位性皮炎、系统性红斑狼疮和类风湿性关节炎中的一种或多种免疫相关性疾病。在另一些实施方案中,所述由CCR4介导的疾病或病症可以为癌症。所述的癌症优选选自胆管癌、肝癌、乳腺癌、前列腺癌、肺癌、鼻咽癌、甲状腺癌、胃癌、卵巢癌、结直肠癌、子宫内膜癌、尿路上皮细胞癌、睾丸癌、宫颈癌、白血病、皮肤癌、鳞状细胞癌、基底细胞癌、膀胱癌、食管癌、头颈癌、肾癌、胰腺癌、骨癌、淋巴瘤、黑色素瘤、肉瘤、外周神经上皮瘤、胶质瘤、室管膜瘤、成神经细胞瘤、神经节细胞瘤、成神经管细胞瘤、松果体细胞肿瘤、脑膜瘤、神经纤维瘤、神经鞘瘤和维尔姆斯瘤。In some embodiments, the disease or condition mediated by CCR4 may be selected from one or more of atopic dermatitis, asthma, allergic rhinitis, atopic dermatitis, systemic lupus erythematosus and rheumatoid arthritis. Various immune-related diseases. In other embodiments, the disease or condition mediated by CCR4 may be cancer. The cancer is preferably selected from cholangiocarcinoma, liver cancer, breast cancer, prostate cancer, lung cancer, nasopharyngeal cancer, thyroid cancer, gastric cancer, ovarian cancer, colorectal cancer, endometrial cancer, urothelial cell carcinoma, testicular cancer, Cervical cancer, leukemia, skin cancer, squamous cell carcinoma, basal cell carcinoma, bladder cancer, esophageal cancer, head and neck cancer, kidney cancer, pancreatic cancer, bone cancer, lymphoma, melanoma, sarcoma, peripheral neuroepithelial tumor, glia tumors, ependymomas, neuroblastomas, ganglioneuromas, medulloblastomas, pineal cell tumors, meningiomas, neurofibromas, schwannomas, and Wilms tumors.
在第四方面,本发明提供了一种治疗或预防由CCR4介导的疾病或病症的方法,所述方法包括向有需要的受试者施用治疗有效量的式(I)、(II)、(III)、(IV)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物。In a fourth aspect, the present invention provides a method of treating or preventing a disease or condition mediated by CCR4, the method comprising administering to a subject in need a therapeutically effective amount of formula (I), (II), (III), (IV) compound or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its precursor drugs, or their metabolites.
在第五方面,本发明涉及式(I)、(II)、(III)、(IV)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,其中用于预防或治疗由CCR4介导的疾病或病症。In a fifth aspect, the present invention relates to compounds of formula (I), (II), (III), (IV) or isotopically labeled compounds thereof, or optical isomers, geometric isomers, tautomers or isomers thereof A mixture of conformers, or a pharmaceutically acceptable salt thereof, or a prodrug thereof, or a metabolite thereof, wherein it is used for preventing or treating a disease or condition mediated by CCR4.
在一些实施方案中,本发明化合物可以通过口服、肠胃外、静脉注射、肌肉注射、皮下注射、鼻腔、口腔粘膜、眼、经肺、经呼吸道、经阴道、经直肠、腹膜内、病灶内、病灶周围等途径施用。 In some embodiments, the compounds of the present invention can be administered orally, parenterally, intravenously, intramuscularly, subcutaneously, nasally, buccally, ocularly, via the lungs, via the respiratory tract, vaginally, rectally, intraperitoneally, intralesionally, Administer around the lesion and other routes.
“治疗有效量”是指本发明化合物当单独或组合给药时有效治疗或预防由CCR4介导的疾病或病症的量。A "therapeutically effective amount" refers to an amount of a compound of the present invention effective to treat or prevent a disease or condition mediated by CCR4 when administered alone or in combination.
具体施用剂量将取决于施用途径、疾病的严重程度、患者的年龄和体重,以及主治医师在确定最适合特定患者的个体方案和剂量水平时通常考虑的其他因素。例如,本发明的化合物的日剂量可以特别地为0.001-150mg/kg体重(例如0.1mg/kg体重、1mg/kg体重、10mg/kg体重或100mg/kg体重等)。The specific dosage administered will depend on the route of administration, the severity of the disease, the age and weight of the patient, and other factors normally considered by the attending physician in determining the individual regimen and dosage level most suitable for a particular patient. For example, the daily dose of the compound of the invention may specifically be 0.001-150 mg/kg body weight (eg 0.1 mg/kg body weight, 1 mg/kg body weight, 10 mg/kg body weight or 100 mg/kg body weight etc.).
具体的施用频率可以由相关领域的技术人员确定,例如为1天1次、2天1次、3天1次、4天1次、5天1次、6天1次、1天2次、1天3次等。The specific administration frequency can be determined by those skilled in the art, for example, once a day, once a day, once a day, once a day, once a day, once a day, once a day, once a day, twice a day, 1 day 3 times etc.
本领域技术人员能够理解,在本发明的一个方面中描述的定义和优选项同样适用于其他方面。本领域技术人员能够明了本发明各个方面的实施方案可以以各种方式组合,而不偏离本发明的主题和思想,这些组合也包括在本发明的范围内。Those skilled in the art can understand that the definitions and preferences described in one aspect of the present invention are also applicable to other aspects. Those skilled in the art can understand that the embodiments of various aspects of the present invention can be combined in various ways without departing from the subject and idea of the present invention, and these combinations are also included in the scope of the present invention.
附图说明Description of drawings
图1显示测试化合物对FITC诱导的动物模型的耳肿胀的抑制效应。Figure 1 shows the inhibitory effect of test compounds on FITC-induced ear swelling in an animal model.
图2显示测试化合物对FITC诱导的动物模型的体重影响。Figure 2 shows the effect of test compounds on body weight in FITC-induced animal models.
图3显示测试化合物对OXA诱导的动物模型的耳肿胀的抑制效应。Figure 3 shows the inhibitory effect of test compounds on OXA-induced ear swelling in animal models.
具体实施方式Detailed ways
本发明式(I)化合物可以用有机合成领域的技术人员所熟悉的多种方法合成。以下具体实施例中给出了一些示例性的式(I)化合物的合成方法,这些方法是合成化学领域所公知的。显然,参照本专利中的示例性方案,本领域技术人员可以适当调整反应物、反应条件和保护基团而容易地设计其他式(I)化合物的合成路线。The compounds of formula (I) of the present invention can be synthesized by various methods familiar to those skilled in the art of organic synthesis. Some exemplary synthetic methods of compounds of formula (I) are given in the following specific examples, and these methods are well known in the field of synthetic chemistry. Apparently, with reference to the exemplary schemes in this patent, those skilled in the art can easily design the synthetic routes of other compounds of formula (I) by appropriately adjusting the reactants, reaction conditions and protecting groups.
下面进一步结合实施例来阐述本发明;但这些实施例并不限制本发明的范围。除非另有声明,各实施例中所用的所有反应物均从商业途径获得;合成实验和产物分析检测中所用仪器设备等均为有机合成中通常使用的常规仪器和设备。The present invention is further described below in conjunction with examples; but these examples do not limit the scope of the present invention. Unless otherwise stated, all reactants used in each example were obtained from commercial sources; the instruments and equipment used in the synthesis experiment and product analysis and detection were conventional instruments and equipment commonly used in organic synthesis.
中间体A:(R)-1-(2,4-二氯苯基)乙烷-1-胺
Intermediate A: (R)-1-(2,4-dichlorophenyl)ethan-1-amine
室温下,将S-扁桃酸(56.00g,368.29mmol)溶于异丙醇(420mL)和乙醇(280mL)的混合溶剂中,升温至60℃,缓慢滴加化合物A-1(70.00g,368.29mmol),反应混合物在60℃搅拌30分钟,然后室温搅拌24小时。大量白色固体析出,过滤,滤饼用丙酮(105mL)洗涤,真空干燥后,加入异丙醇(315mL)和乙醇(210mL) 的混合溶剂中,60℃搅拌30分钟,冷却到室温,搅拌12小时。大量白色固体析出,过滤,滤饼用丙酮(70mL)洗涤,真空干燥,得到化合物A-2。室温下,将化合物A-2(45.00g,131.49mmol)溶于二氯甲烷(400mL)和4mol/L氢氧化钠水溶液(120mL)中,反应混合物在室温下搅拌1小时。静止分液,有机相用水(300mL×3)洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到中间体A。1H NMR(400MHz,DMSO-d6)δ7.71(d,J=8.8Hz,1H),7.48(d,J=2.4Hz,1H),7.41(dd,J=8.4,2.0Hz,1H),4.35-4.24(m,1H),1.92(s,2H),1.20(d,J=6.4Hz,3H)。At room temperature, dissolve S-mandelic acid (56.00g, 368.29mmol) in a mixed solvent of isopropanol (420mL) and ethanol (280mL), raise the temperature to 60°C, and slowly add compound A-1 (70.00g, 368.29 mmol), the reaction mixture was stirred at 60°C for 30 minutes, then at room temperature for 24 hours. A large amount of white solid precipitated, filtered, the filter cake was washed with acetone (105mL), dried in vacuo, and isopropanol (315mL) and ethanol (210mL) were added in a mixed solvent, stirred at 60°C for 30 minutes, cooled to room temperature, and stirred for 12 hours. A large amount of white solid was precipitated, filtered, the filter cake was washed with acetone (70 mL), and dried in vacuo to obtain compound A-2. Compound A-2 (45.00 g, 131.49 mmol) was dissolved in dichloromethane (400 mL) and 4 mol/L sodium hydroxide aqueous solution (120 mL) at room temperature, and the reaction mixture was stirred at room temperature for 1 hour. After static liquid separation, the organic phase was washed with water (300 mL×3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain Intermediate A. 1 H NMR (400MHz, DMSO-d 6 )δ7.71(d, J=8.8Hz, 1H), 7.48(d, J=2.4Hz, 1H), 7.41(dd, J=8.4, 2.0Hz, 1H) , 4.35-4.24 (m, 1H), 1.92 (s, 2H), 1.20 (d, J=6.4Hz, 3H).
中间体B:(R)-3-(哌啶-3-基)氮杂环丁烷-1-羧酸叔丁酯
Intermediate B: tert-butyl (R)-3-(piperidin-3-yl)azetidine-1-carboxylate
1)化合物B-2的合成1) Synthesis of compound B-2
室温下,将化合物B-1(25.00g,158.95mmol)和3-碘氮杂环丁烷-1-羧酸叔丁酯(25.00g,88.31mmol)溶于异丙醇(400mL)中,加入碘化镍(2.76g,8.83mmol),(1R,2R)-2-氨基环己醇盐酸盐(1.34g,8.83mmol)和双(三甲基硅基)氨基钠(176.60mL,176.61mmol,1mol/L四氢呋喃溶液),反应混合物氮气保护下,80℃搅拌18小时。反应液冷却至室温,缓慢倒入冰水(250mL)中淬灭,用甲基叔丁基醚萃取(250mL×4)。合并有机相,饱和食盐水洗涤(150mL×3),无水硫酸钠干燥,过滤,减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=3/1)分离,得到化合物B-2。MS-ESI:m/z 269.1[M+H]+1H NMR(400MHz,CDCl3)δ8.30(d,J=2.4Hz,1H),7.71(dd,J=8.0,2.4Hz,1H),7.35(d,J=8.4Hz,1H),4.38(t,J=8.8Hz,2H),3.96-3.88(m,2H),3.78-3.69(m,1H),1.47(s,9H)。At room temperature, compound B-1 (25.00g, 158.95mmol) and tert-butyl 3-iodoazetidine-1-carboxylate (25.00g, 88.31mmol) were dissolved in isopropanol (400mL), and added Nickel iodide (2.76g, 8.83mmol), (1R,2R)-2-aminocyclohexanol hydrochloride (1.34g, 8.83mmol) and sodium bis(trimethylsilyl)amide (176.60mL, 176.61mmol , 1mol/L tetrahydrofuran solution), the reaction mixture was stirred at 80°C for 18 hours under nitrogen protection. The reaction solution was cooled to room temperature, slowly poured into ice water (250 mL) to quench, and extracted with methyl tert-butyl ether (250 mL×4). The organic phases were combined, washed with saturated brine (150mL×3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to obtain compound B-2. MS-ESI: m/z 269.1 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ8.30 (d, J = 2.4Hz, 1H), 7.71 (dd, J = 8.0, 2.4Hz, 1H), 7.35 (d, J = 8.4Hz, 1H), 4.38 (t, J=8.8Hz, 2H), 3.96-3.88(m, 2H), 3.78-3.69(m, 1H), 1.47(s, 9H).
2)化合物B-3的合成2) Synthesis of Compound B-3
室温下,将化合物B-2(26.00g,96.75mmol)和(S)-4-异丙基噁唑烷-2-酮(24.99g,193.50mmol)溶于无水甲苯(400mL)中,随后加入三(二亚苄基丙酮)二钯(4.43g,4.84mmol),2-二环己基磷-2,4,6-三异丙基联苯(2.31g,4.84mmol)和碳酸铯(31.52g,96.75mmol),氮气置换,反应混合物在100℃搅拌16小时。反应混合物冷却至室温,过滤,滤饼用甲基叔丁基醚(500mL)洗涤,有机相用饱和食盐水(500mL×4)洗涤,无水硫酸钠干燥,过滤,减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=1/1)分离,所得粗品再用石油醚(200mL)打浆,过滤,滤饼再用石油醚(30mL)洗涤,减压浓缩,得到化合物B-3。1H NMR(400MHz,CD3OD)δ8.29(d,J=2.0Hz,1H),8.11(d,J=8.8Hz,1H),7.88(dd,J=8.4,2.4Hz,1H),4.92-4.85(m,1H),4.47-4.41(m,1H),4.40-4.33(m,3H),3.98-3.90(m,2H),3.89-3.81(m,1H),2.51-2.40(m,1H),1.47(s,9H),0.94(d,J=7.2Hz,3H),0.82(d,J=6.8Hz,3H)。At room temperature, compound B-2 (26.00g, 96.75mmol) and (S)-4-isopropyloxazolidin-2-one (24.99g, 193.50mmol) were dissolved in anhydrous toluene (400mL), followed by Add tris(dibenzylideneacetone)dipalladium (4.43g, 4.84mmol), 2-dicyclohexylphosphine-2,4,6-triisopropylbiphenyl (2.31g, 4.84mmol) and cesium carbonate (31.52 g, 96.75mmol), replaced with nitrogen, and the reaction mixture was stirred at 100°C for 16 hours. The reaction mixture was cooled to room temperature, filtered, the filter cake was washed with methyl tert-butyl ether (500 mL), the organic phase was washed with saturated brine (500 mL×4), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a residue After separation by silica gel column chromatography (petroleum ether/ethyl acetate=1/1), the resulting crude product was slurried with petroleum ether (200 mL), filtered, and the filter cake was washed with petroleum ether (30 mL) and concentrated under reduced pressure to obtain compound B -3. 1 H NMR (400MHz, CD 3 OD) δ8.29 (d, J = 2.0Hz, 1H), 8.11 (d, J = 8.8Hz, 1H), 7.88 (dd, J = 8.4, 2.4Hz, 1H), 4.92-4.85(m,1H),4.47-4.41(m,1H),4.40-4.33(m,3H),3.98-3.90(m,2H),3.89-3.81(m,1H),2.51-2.40(m , 1H), 1.47 (s, 9H), 0.94 (d, J=7.2Hz, 3H), 0.82 (d, J=6.8Hz, 3H).
3)中间体B的合成 3) Synthesis of Intermediate B
室温下,将化合物B-3(10.00g,27.67mmol)溶于无水乙酸(200mL),氮气保护下,缓慢加入二氧化铂(1.00g,4.40mmol),氢气置换,反应混合物在氢气(15Psi)氛围下室温反应15小时。反应液过滤,滤饼用甲醇(100mL)洗涤,滤液减压浓缩。所得残余物经硅胶柱层析(乙酸乙酯/甲醇=0/1)分离得到粗品。粗品溶于二氯甲烷(50mL),加入1mol/L的氢氧化钠水溶液(80mL)调节pH~13。有机相用无水硫酸钠干燥,过滤,减压浓缩得到中间体B的粗品。室温下,将中间体B的粗品溶于甲基叔丁基醚(1000mL),回流状态下,分批加入S-扁桃酸(13.61g,89.45mmol),反应混合物室温搅拌16小时。大量白色固体析出,过滤,滤饼用甲基叔丁基醚(400mL)洗涤后,溶于二氯甲烷(500mL)和1mol/L的氢氧化钠水溶液(800mL)中。有机相用无水硫酸钠干燥,过滤,减压浓缩,得到粗品。室温下,再将粗品溶于甲基叔丁基醚(500mL),回流状态下,分批加入S-扁桃酸(8.23g,54.09mmol),反应混合物室温反应16小时。大量白色固体析出,反应混合液过滤,滤饼用甲基叔丁基醚(300mL)洗涤后,溶于二氯甲烷(500mL)和1mol/L的氢氧化钠水溶液(800mL)。有机相用无水硫酸钠干燥,过滤,减压浓缩,得到中间体B。1H NMR(400MHz,CD3OD)δ4.00-3.88(m,2H),3.70-3.60(m,2H),3.00-2.89(m,2H),2.50(td,J=12.0,2.8Hz,1H),2.35-2.23(m,1H),2.15(dd,J=12.0,10.4Hz,1H),1.85-1.75(m,1H),1.72-1.56(m,2H),1.55-1.45(m,1H),1.43(s,9H),1.07-0.93(m,1H)。At room temperature, compound B-3 (10.00g, 27.67mmol) was dissolved in anhydrous acetic acid (200mL), under the protection of nitrogen, slowly added platinum dioxide (1.00g, 4.40mmol), hydrogen replacement, the reaction mixture in hydrogen (15Psi ) atmosphere at room temperature for 15 hours. The reaction solution was filtered, the filter cake was washed with methanol (100 mL), and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (ethyl acetate/methanol=0/1) to obtain a crude product. The crude product was dissolved in dichloromethane (50 mL), and 1 mol/L aqueous sodium hydroxide solution (80 mL) was added to adjust the pH to 13. The organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain the crude intermediate B. At room temperature, the crude intermediate B was dissolved in methyl tert-butyl ether (1000 mL), and S-mandelic acid (13.61 g, 89.45 mmol) was added in portions under reflux, and the reaction mixture was stirred at room temperature for 16 hours. A large amount of white solid precipitated, filtered, and the filter cake was washed with methyl tert-butyl ether (400 mL), and dissolved in dichloromethane (500 mL) and 1 mol/L sodium hydroxide aqueous solution (800 mL). The organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain the crude product. At room temperature, the crude product was dissolved in methyl tert-butyl ether (500 mL), and S-mandelic acid (8.23 g, 54.09 mmol) was added in portions under reflux, and the reaction mixture was reacted at room temperature for 16 hours. A large amount of white solid precipitated out, and the reaction mixture was filtered, and the filter cake was washed with methyl tert-butyl ether (300 mL), and dissolved in dichloromethane (500 mL) and 1 mol/L sodium hydroxide aqueous solution (800 mL). The organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain Intermediate B. 1 H NMR (400MHz, CD 3 OD) δ4.00-3.88(m, 2H), 3.70-3.60(m, 2H), 3.00-2.89(m, 2H), 2.50(td, J=12.0, 2.8Hz, 1H),2.35-2.23(m,1H),2.15(dd,J=12.0,10.4Hz,1H),1.85-1.75(m,1H),1.72-1.56(m,2H),1.55-1.45(m, 1H), 1.43(s, 9H), 1.07-0.93(m, 1H).
中间体C:(R)-2-(3-(氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇盐酸盐
Intermediate C: (R)-2-(3-(azetidin-3-yl)piperidin-1-yl)ethan-1-ol hydrochloride
1)化合物C-1的合成1) Synthesis of Compound C-1
室温下,中间体B(10.00g,36.13mmol)和2-碘乙醇(12.43g,72.25mmol)溶于乙腈(100mL),缓慢加入碳酸钾(14.98g,108.38mmol),反应液在80℃搅拌16小时。反应液冷却至室温后,加入二氯甲烷(200mL)稀释,过滤,滤液减压浓缩,所得残余物经硅胶柱层析(乙酸乙酯/甲醇=0/1)分离得到粗品,粗品再用二氯甲烷(150mL)溶解,过滤,滤液减压浓缩,得到化合物C-1。1H NMR(400MHz,CD3OD)δ4.01-3.85(m,2H),3.70(t,J=6.0Hz,2H),3.67-3.57(m,2H),3.10-2.94(m,2H),2.66(t,J=6.0Hz,2H),2.37-2.16(m,2H),1.95-1.52(m,5H),1.39(s,9H),0.98-0.81(m,1H)。At room temperature, intermediate B (10.00g, 36.13mmol) and 2-iodoethanol (12.43g, 72.25mmol) were dissolved in acetonitrile (100mL), potassium carbonate (14.98g, 108.38mmol) was slowly added, and the reaction solution was stirred at 80°C 16 hours. After the reaction solution was cooled to room temperature, dichloromethane (200 mL) was added to dilute, filtered, and the filtrate was concentrated under reduced pressure. The resulting residue was separated by silica gel column chromatography (ethyl acetate/methanol=0/1) to obtain a crude product, which was then purified with two Chloromethane (150 mL) was dissolved, filtered, and the filtrate was concentrated under reduced pressure to obtain compound C-1. 1 H NMR (400MHz, CD 3 OD) δ4.01-3.85 (m, 2H), 3.70 (t, J = 6.0Hz, 2H), 3.67-3.57 (m, 2H), 3.10-2.94 (m, 2H) , 2.66 (t, J=6.0Hz, 2H), 2.37-2.16 (m, 2H), 1.95-1.52 (m, 5H), 1.39 (s, 9H), 0.98-0.81 (m, 1H).
2)中间体C的合成2) Synthesis of Intermediate C
室温下,将化合物C-1(3.00g,9.49mmol)溶于二氧六环(25mL)中,缓慢加入4mol/L盐酸二氧六环溶液(25mL),反应混合液在室温下搅拌4小时。反应完毕 后,反应液减压浓缩得到中间体C。粗品未经纯化,直接用于下一步反应。MS-ESI:m/z 185.1[M+H]+At room temperature, compound C-1 (3.00g, 9.49mmol) was dissolved in dioxane (25mL), and 4mol/L hydrochloric acid dioxane solution (25mL) was slowly added, and the reaction mixture was stirred at room temperature for 4 hours . The reaction is complete Afterwards, the reaction solution was concentrated under reduced pressure to obtain intermediate C. The crude product was directly used in the next reaction without further purification. MS-ESI: m/z 185.1 [M+H] + .
中间体D:(1R,3r)-3-((R)-3-(氮杂环丁烷-3-基)哌啶-1-基)-1-甲基环丁烷-1-甲酸甲酯盐酸盐
Intermediate D: (1R,3r)-3-((R)-3-(azetidin-3-yl)piperidin-1-yl)-1-methylcyclobutane-1-carboxylic acid methyl Ester hydrochloride
1)化合物D-2的合成1) Synthesis of compound D-2
室温下,将化合物D-1(5.00g,35.17mmol)溶于四氢呋喃(30mL)和水(30mL)的混合物溶剂中,随后加入氢氧化锂(4.43g,105.52mmol),反应液在25℃搅拌15小时。用1mol/L盐酸水溶液调节反应液pH~4,二氯甲烷萃取(50mL×3)。合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物D-2。粗品未经纯化,直接用于下一步反应。1H NMR(400MHz,DMSO-d6)δ12.57(s,1H),3.42-3.37(m,2H),2.99-2.92(m,2H),1.47(s,3H)。At room temperature, compound D-1 (5.00g, 35.17mmol) was dissolved in a mixture solvent of tetrahydrofuran (30mL) and water (30mL), then lithium hydroxide (4.43g, 105.52mmol) was added, and the reaction solution was stirred at 25°C 15 hours. Adjust the pH of the reaction solution to ~4 with 1mol/L hydrochloric acid aqueous solution, and extract with dichloromethane (50mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound D-2. The crude product was directly used in the next reaction without further purification. 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.57 (s, 1H), 3.42-3.37 (m, 2H), 2.99-2.92 (m, 2H), 1.47 (s, 3H).
2)化合物D-3的合成2) Synthesis of Compound D-3
室温下,将中间体B(3.00g,12.48mmol),化合物D-2(1.76g,13.73mmol)和三乙酰氧基硼氢化钠(3.95g,18.72mmol)溶于1,2-二氯乙烷(150mL),反应液在-5℃下缓慢滴加醋酸(0.70mL,12.48mmol),滴加完毕后,在20℃搅拌15小时。补加化合物D-2(800mg,6.24mmol)和三乙酰氧基硼氢化钠(1316mg,6.24mmol),反应液在20℃搅拌3小时,补加化合物D-2(800mg,6.24mmol)和三乙酰氧基硼氢化钠(1316mg,6.24mmol)。反应完成后,反应液减压浓缩,所得残余物经硅胶柱层析(二氯甲烷/甲醇=10/1)分离,得到化合物D-3。1H NMR(400MHz,CD3OD)δ4.04-3.95(m,2H),3.77-3.67(m,2H),3.24-3.16(m,1H),2.77-2.68(m,2H),2.61-2.53(m,1H),2.43-2.36(m,1H),2.29(t,J=12.0Hz,1H),2.13-2.02(m,2H),1.96-1.81(m,4H),1.79-1.68(m,1H),1.43(s,9H),1.42-1.40(m,1H),1.39(s,3H),1.17-1.05(m,1H)。At room temperature, intermediate B (3.00g, 12.48mmol), compound D-2 (1.76g, 13.73mmol) and sodium triacetoxyborohydride (3.95g, 18.72mmol) were dissolved in 1,2-dichloroethyl Acetic acid (0.70 mL, 12.48 mmol) was slowly added dropwise to the reaction solution at -5°C, and stirred at 20°C for 15 hours after the addition was complete. Compound D-2 (800mg, 6.24mmol) and sodium triacetoxyborohydride (1316mg, 6.24mmol) were added, the reaction solution was stirred at 20°C for 3 hours, and compound D-2 (800mg, 6.24mmol) and three Sodium acetoxyborohydride (1316 mg, 6.24 mmol). After the reaction was completed, the reaction solution was concentrated under reduced pressure, and the resulting residue was separated by silica gel column chromatography (dichloromethane/methanol=10/1) to obtain compound D-3. 1 H NMR (400MHz, CD 3 OD) δ4.04-3.95(m,2H),3.77-3.67(m,2H),3.24-3.16(m,1H),2.77-2.68(m,2H),2.61- 2.53(m,1H),2.43-2.36(m,1H),2.29(t,J=12.0Hz,1H),2.13-2.02(m,2H),1.96-1.81(m,4H),1.79-1.68( m, 1H), 1.43(s, 9H), 1.42-1.40(m, 1H), 1.39(s, 3H), 1.17-1.05(m, 1H).
3)化合物D-4的合成3) Synthesis of Compound D-4
室温下,将化合物D-3(2.60g,7.38mmol)溶于二氯甲烷(30mL)和甲醇(3mL)的混合溶剂中,在0℃下缓慢加入三甲基硅基重氮甲烷(14.70mL,29.51mmol),之后在20℃搅拌2小时。反应完毕后,将醋酸缓慢滴加到反应液,直至反应液的颜色由黄色变成无色且不再冒气泡,加水(30mL)稀释,二氯甲烷(30mL)萃取,饱和氯化钠水溶液(25mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物D-4。1H NMR(400MHz,CD3OD)δ4.00-3.89(m,2H),3.70(s,3H),3.68-3.61(m,2H),2.87-2.79(m,1H),2.77-2.67(m,2H),2.60-2.51(m,2H),2.36-2.25(m,1H),1.89-1.80(m,2H),1.79-1.64(m,4H),1.61-1.49(m,1H),1.43(s,9H),1.39(d,J=10.8Hz,1H),1.36(s,3H),0.93-0.78(m,1H)。At room temperature, compound D-3 (2.60g, 7.38mmol) was dissolved in a mixed solvent of dichloromethane (30mL) and methanol (3mL), and trimethylsilyldiazomethane (14.70mL , 29.51 mmol), followed by stirring at 20°C for 2 hours. After completion of the reaction, acetic acid was slowly added dropwise to the reaction solution until the color of the reaction solution changed from yellow to colorless and no longer bubbled, diluted with water (30mL), extracted with dichloromethane (30mL), saturated aqueous sodium chloride ( 25 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound D-4. 1 H NMR (400MHz, CD 3 OD) δ4.00-3.89(m,2H),3.70(s,3H),3.68-3.61(m,2H),2.87-2.79(m,1H),2.77-2.67( m,2H),2.60-2.51(m,2H),2.36-2.25(m,1H),1.89-1.80(m,2H),1.79-1.64(m,4H),1.61-1.49(m,1H), 1.43 (s, 9H), 1.39 (d, J=10.8Hz, 1H), 1.36 (s, 3H), 0.93-0.78 (m, 1H).
4)中间体D的合成 4) Synthesis of Intermediate D
室温下,将化合物D-4(1.75g,4.78mmol)溶于1,4-二氧六环(30mL)中,缓慢加入4mol/L盐酸二氧六环溶液(6.00mL,24.00mmol),反应液在20℃搅拌15小时。反应液减压浓缩,得到中间体D。粗品未经纯化,直接用于下一步反应。At room temperature, compound D-4 (1.75g, 4.78mmol) was dissolved in 1,4-dioxane (30mL), and 4mol/L dioxane hydrochloride solution (6.00mL, 24.00mmol) was slowly added to react The solution was stirred at 20°C for 15 hours. The reaction solution was concentrated under reduced pressure to obtain intermediate D. The crude product was directly used in the next reaction without further purification.
中间体E:(R)-3-(氮杂环丁烷-3-基)哌啶-1-甲酸烯丙酯盐酸盐
Intermediate E: Allyl (R)-3-(azetidin-3-yl)piperidine-1-carboxylate hydrochloride
1)中间体E-1的合成1) Synthesis of intermediate E-1
室温下,将中间体B(500mg,2.08mmol)溶于二氯甲烷(10mL),加入碳酸钠(330mg,3.12mmol)和氯甲酸烯丙酯(376mg,3.12mmol),反应混合物在25℃搅拌16小时。加水(50mL)稀释,乙酸乙酯(50mL×3)萃取,无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=2/1)分离,得到化合物E-1。1H NMR(400MHz,CDCl3)δ6.01-5.89(m,1H),5.30(d,J=17.2Hz,1H),5.22(d,J=10.4Hz,1H),4.59(d,J=5.2Hz,2H),4.02-3.91(m,3H),3.88-3.70(m,1H),3.65(dd,J=8.4,5.6Hz,1H),2.98-2.85(m,1H),2.70-2.41(m,1H),2.35-2.23(m,1H),1.85-1.75(m,1H),1.73-1.63(m,2H),1.62-1.57(m,1H),1.55-1.47(m,1H),1.44(s,9H),1.14-1.02(m,1H)。Intermediate B (500mg, 2.08mmol) was dissolved in dichloromethane (10mL) at room temperature, sodium carbonate (330mg, 3.12mmol) and allyl chloroformate (376mg, 3.12mmol) were added, and the reaction mixture was stirred at 25°C 16 hours. Dilute with water (50 mL), extract with ethyl acetate (50 mL×3), dry over anhydrous sodium sulfate, filter, concentrate the filtrate under reduced pressure, and separate the resulting residue by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) , to obtain compound E-1. 1 H NMR (400MHz, CDCl 3 ) δ6.01-5.89(m, 1H), 5.30(d, J=17.2Hz, 1H), 5.22(d, J=10.4Hz, 1H), 4.59(d, J= 5.2Hz, 2H), 4.02-3.91(m, 3H), 3.88-3.70(m, 1H), 3.65(dd, J=8.4, 5.6Hz, 1H), 2.98-2.85(m, 1H), 2.70-2.41 (m,1H),2.35-2.23(m,1H),1.85-1.75(m,1H),1.73-1.63(m,2H),1.62-1.57(m,1H),1.55-1.47(m,1H) ,1.44(s,9H),1.14-1.02(m,1H).
2)中间体E的合成2) Synthesis of Intermediate E
室温下,将化合物E-1(650mg,2.00mmol)溶于盐酸二氧六环溶液(4mL,4mol/L)中,反应液在25℃搅拌2小时。反应完毕后,减压浓缩,得到中间体E。粗品不经纯化,直接用于下一步反应。1H NMR(400MHz,CDCl3)δ9.80(s,1H),9.56(s,1H),6.01-5.87(m,1H),5.29(dd,J=17.2,1.2Hz,1H),5.23(dd,J=10.0,0.8Hz,1H),4.63-4.52(m,2H),4.17-4.03(m,2H),3.95-3.85(m,2H),3.84-3.72(m,2H),3.08-2.98(m,1H),2.86-2.73(m,1H),2.72-2.61(m,1H),1.96-1.83(m,1H),1.81-1.71(m,1H),1.70-1.60(m,1H),1.54-1.42(m,1H),1.17-1.05(m,1H)。Compound E-1 (650 mg, 2.00 mmol) was dissolved in dioxane hydrochloride solution (4 mL, 4 mol/L) at room temperature, and the reaction solution was stirred at 25° C. for 2 hours. After the reaction was completed, it was concentrated under reduced pressure to obtain intermediate E. The crude product was directly used in the next reaction without purification. 1 H NMR (400MHz, CDCl 3 ) δ9.80(s, 1H), 9.56(s, 1H), 6.01-5.87(m, 1H), 5.29(dd, J=17.2, 1.2Hz, 1H), 5.23( dd,J=10.0,0.8Hz,1H),4.63-4.52(m,2H),4.17-4.03(m,2H),3.95-3.85(m,2H),3.84-3.72(m,2H),3.08- 2.98(m,1H),2.86-2.73(m,1H),2.72-2.61(m,1H),1.96-1.83(m,1H),1.81-1.71(m,1H),1.70-1.60(m,1H ), 1.54-1.42(m,1H), 1.17-1.05(m,1H).
中间体F:(R)-3-(氮杂环丁烷-3-基)哌啶-1-甲酸烯丙酯盐酸盐
Intermediate F: Allyl (R)-3-(azetidin-3-yl)piperidine-1-carboxylate hydrochloride
1)化合物F-1的合成1) Synthesis of Compound F-1
室温下,依次将化合物C-1(3.00g,9.49mmol,90%纯度)和叔丁基二苯基氯硅烷(3.70mL,14.24mmol)加入二氯甲烷(20mL)和N,N-二甲基甲酰胺(20mL)中,搅拌均匀后,加入咪唑(2.59g,37.98mmol),反应液在60℃搅拌16小时。反应液用乙酸乙酯(150mL)稀释,水(100mL×3)洗涤,有机相用无水硫酸钠干燥,过滤, 滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=1/1)分离,得到化合物F-1。MS-ESI:m/z 523.3[M+H]+At room temperature, sequentially add compound C-1 (3.00 g, 9.49 mmol, 90% purity) and tert-butyldiphenylchlorosilane (3.70 mL, 14.24 mmol) into dichloromethane (20 mL) and N,N-dimethyl After stirring evenly, imidazole (2.59 g, 37.98 mmol) was added, and the reaction solution was stirred at 60° C. for 16 hours. The reaction solution was diluted with ethyl acetate (150 mL), washed with water (100 mL×3), the organic phase was dried over anhydrous sodium sulfate, filtered, The filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to obtain compound F-1. MS-ESI: m/z 523.3 [M+H] + .
2)中间体F的合成2) Synthesis of Intermediate F
冰浴下,将化合物F-1(3200mg,5.20mmol,85%纯度),溶于二氯甲烷(30mL)中,缓慢加入三氟乙酸(3.00mL),反应液在室温下反应16小时。反应液用甲醇(30mL)稀释,加入碱性树脂调节溶液的pH~8,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(乙酸乙酯/甲醇=0/1)分离,得到中间体F。MS-ESI:m/z 423.2[M+H]+Under ice-cooling, compound F-1 (3200 mg, 5.20 mmol, 85% purity) was dissolved in dichloromethane (30 mL), trifluoroacetic acid (3.00 mL) was slowly added, and the reaction solution was reacted at room temperature for 16 hours. The reaction solution was diluted with methanol (30 mL), and a basic resin was added to adjust the pH of the solution to ~8, filtered, and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (ethyl acetate/methanol=0/1) to obtain Intermediate F. MS-ESI: m/z 423.2 [M+H] + .
实施例1:2-(R)-3-(1-(4-((R)-1-(2,4-二氯苯基)乙基)氨基)吡唑并[1,5-a][1,3,5]三嗪-2-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(1)
Example 1: 2-(R)-3-(1-(4-((R)-1-(2,4-dichlorophenyl)ethyl)amino)pyrazolo[1,5-a] [1,3,5]triazin-2-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (1)
1)化合物1-2的合成1) Synthesis of compound 1-2
室温下,将化合物1-1(1.00g,5.49mmol)溶于三氯氧磷(10.00mL)中,缓慢滴加N,N-二甲基苯胺(0.33g,2.76mmol),反应液在110℃搅拌5小时。反应完毕后,反应液冷却至室温,减压浓缩,所得残余物用水(20mL)稀释,甲苯(30mL×2)萃取,合并有机相,用饱和氯化钠水溶液(20mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到化合物1-2。1H NMR(400MHz,CDCl3)δ8.16(d,J=2.0Hz,1H),6.52(d,J=2.0Hz,1H),2.62(s,3H)。At room temperature, compound 1-1 (1.00g, 5.49mmol) was dissolved in phosphorus oxychloride (10.00mL), and N,N-dimethylaniline (0.33g, 2.76mmol) was slowly added dropwise. °C and stirred for 5 hours. After completion of the reaction, the reaction solution was cooled to room temperature, concentrated under reduced pressure, the resulting residue was diluted with water (20 mL), extracted with toluene (30 mL×2), the organic phases were combined, washed with saturated aqueous sodium chloride (20 mL), anhydrous sodium sulfate Dry, filter, and concentrate under reduced pressure to obtain compound 1-2. 1 H NMR (400MHz, CDCl 3 ) δ8.16 (d, J=2.0Hz, 1H), 6.52 (d, J=2.0Hz, 1H), 2.62 (s, 3H).
2)化合物1-3的合成2) Synthesis of compound 1-3
室温下,将化合物1-2(600mg,2.99mmol),中间体A(568mg,2.99mmol)和N,N-二异丙基乙胺(773mg,5.98mmol)溶于乙腈(15mL)中,反应液在25℃搅拌2小时。反应完毕后,加水(20mL)稀释,乙酸乙酯(30mL×2)萃取,饱和氯化钠水溶液(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=3/1)分离,得到化合物1-3。MS-ESI:m/z 354.1[M+H]+1H NMR(400MHz,CDCl3)δ7.91(d,J=2.4Hz,1H),7.42(d,J=2.0Hz,1H),7.35(d,J=8.4Hz,1H),7.25(dd,J=8.4,2.0Hz,1H),6.89(d,J=7.6Hz,1H),6.23(d,J=2.0Hz,1H),5.76-5.64(m,1H),2.47(s,3H),1.68(d,J=7.2Hz,3H)。At room temperature, compound 1-2 (600mg, 2.99mmol), intermediate A (568mg, 2.99mmol) and N,N-diisopropylethylamine (773mg, 5.98mmol) were dissolved in acetonitrile (15mL), and the reaction The solution was stirred at 25°C for 2 hours. After the reaction was completed, dilute with water (20mL), extract with ethyl acetate (30mL×2), wash with saturated aqueous sodium chloride solution (30mL), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure. (petroleum ether/ethyl acetate=3/1) to obtain compound 1-3. MS-ESI: m/z 354.1 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ7.91(d, J=2.4Hz, 1H), 7.42(d, J=2.0Hz, 1H), 7.35(d, J=8.4Hz, 1H), 7.25(dd ,J=8.4,2.0Hz,1H),6.89(d,J=7.6Hz,1H),6.23(d,J=2.0Hz,1H),5.76-5.64(m,1H),2.47(s,3H) , 1.68 (d, J=7.2Hz, 3H).
3)化合物1-4的合成3) Synthesis of compound 1-4
室温下,将化合物1-3(100mg,0.28mmol)溶于二氯甲烷(5mL)中,分批加入间氯过氧苯甲酸(172mg,0.85mmol,85%纯度),反应液在25℃搅拌2小时。反应完毕后,反应液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=2/1)分离,得到化合物1-4。MS-ESI:m/z 386.1[M+H]+1H NMR(400MHz,CDCl3)δ8.13(d, J=2.0Hz,1H),7.44(d,J=2.4Hz,1H),7.40(d,J=8.4Hz,1H),7.37(d,J=7.6Hz,1H),7.31-7.27(m,1H),6.70(d,J=2.4Hz,1H),5.86-5.76(m,1H),3.23(s,3H),1.77(d,J=7.2Hz,3H)。At room temperature, compound 1-3 (100mg, 0.28mmol) was dissolved in dichloromethane (5mL), m-chloroperoxybenzoic acid (172mg, 0.85mmol, 85% purity) was added in batches, and the reaction solution was stirred at 25°C 2 hours. After the reaction was completed, the reaction solution was concentrated under reduced pressure, and the resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to obtain compound 1-4. MS-ESI: m/z 386.1 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ8.13(d, J=2.0Hz, 1H), 7.44(d, J=2.4Hz, 1H), 7.40(d, J=8.4Hz, 1H), 7.37(d, J=7.6Hz, 1H), 7.31-7.27(m, 1H), 6.70 (d, J = 2.4Hz, 1H), 5.86-5.76 (m, 1H), 3.23 (s, 3H), 1.77 (d, J = 7.2Hz, 3H).
4)化合物1的合成4) Synthesis of Compound 1
室温下,将化合物1-4(80mg,0.21mmol),N,N-二异丙基乙胺(80mg,0.62mmol)和中间体C(46mg,0.21mmol)溶于乙腈(10mL)中,反应液在50℃搅拌2小时。反应完毕后,反应液减压浓缩,所得残余物经制备级高效液相色谱(碳酸氢铵/乙腈/水体系,色谱柱:Waters Xbridge 150*25mm*5μm;流动相:水(10mM碳酸氢铵),乙腈;梯度配比:乙腈相(0-8min,44-74%);流速:25mL/min;柱温:室温)分离,得到化合物1。MS-ESI:m/z 490.3[M+H]+1H NMR(400MHz,CD3OD)δ7.80(d,J=2.0Hz,1H),7.49(d,J=8.4Hz,1H),7.47(d,J=2.0Hz,1H),7.30(dd,J=8.4,2.0Hz,1H),5.83(d,J=2.4Hz,1H),5.67-5.58(m,1H),4.08(t,J=8.8Hz,1H),4.03-3.92(m,1H),3.82-3.73(m,1H),3.69(t,J=6.0Hz,2H),3.64-3.47(m,1H),2.99-2.81(m,2H),2.55(t,J=6.0Hz,2H),2.44-2.31(m,1H),2.04(t,J=12.0Hz,1H),1.79-1.66(m,4H),1.63-1.56(m,4H),0.97-0.80(m,1H)。At room temperature, compound 1-4 (80mg, 0.21mmol), N,N-diisopropylethylamine (80mg, 0.62mmol) and intermediate C (46mg, 0.21mmol) were dissolved in acetonitrile (10mL), and the reaction The solution was stirred at 50°C for 2 hours. After completion of the reaction, the reaction solution was concentrated under reduced pressure, and the resulting residue was subjected to preparative high-performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge 150*25mm*5 μm; mobile phase: water (10mM ammonium bicarbonate ), acetonitrile; gradient ratio: acetonitrile phase (0-8min, 44-74%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 1. MS-ESI: m/z 490.3 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.80(d, J=2.0Hz, 1H), 7.49(d, J=8.4Hz, 1H), 7.47(d, J=2.0Hz, 1H), 7.30( dd, J=8.4, 2.0Hz, 1H), 5.83(d, J=2.4Hz, 1H), 5.67-5.58(m, 1H), 4.08(t, J=8.8Hz, 1H), 4.03-3.92(m ,1H),3.82-3.73(m,1H),3.69(t,J=6.0Hz,2H),3.64-3.47(m,1H),2.99-2.81(m,2H),2.55(t,J=6.0 Hz,2H),2.44-2.31(m,1H),2.04(t,J=12.0Hz,1H),1.79-1.66(m,4H),1.63-1.56(m,4H),0.97-0.80(m, 1H).
实施例2:2-(R)-3-(1-(4-((R)-1-(2,4-二氯苯基)乙基)氨基)-7-甲基吡唑[1,5-a][1,3,5]三嗪-2-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(2)
Example 2: 2-(R)-3-(1-(4-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-7-methylpyrazol[1, 5-a][1,3,5]triazin-2-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (2)
1)化合物2-2的合成1) Synthesis of compound 2-2
在室温下,将化合物2-1(25.00g,280.61mmol)溶于1,2-二氯乙烷(125mL)中,缓慢滴加草酰氯(33.20mL,392.86mmol),反应混合物在60℃搅拌6小时。反应完毕后,过滤,滤液减压浓缩,得到化合物2-2。粗品未经纯化,直接用于下一步反应。1H NMR(400MHz,CDCl3)δ4.25(q,J=7.2Hz,2H),1.27(t,J=7.2Hz,3H)。At room temperature, compound 2-1 (25.00g, 280.61mmol) was dissolved in 1,2-dichloroethane (125mL), and oxalyl chloride (33.20mL, 392.86mmol) was slowly added dropwise, and the reaction mixture was stirred at 60°C 6 hours. After the reaction was completed, it was filtered, and the filtrate was concentrated under reduced pressure to obtain compound 2-2. The crude product was directly used in the next reaction without further purification. 1 H NMR (400MHz, CDCl 3 ) δ 4.25 (q, J=7.2Hz, 2H), 1.27 (t, J=7.2Hz, 3H).
2)化合物2-3的合成2) Synthesis of compound 2-3
室温下,将3-氨基-5-甲基吡唑(2.50g,25.74mmol)溶于乙腈(25mL)中,加入化合物2-2(8.08g,38.61mmol,55%纯度),反应混合物在室温搅拌4小时。反应完 毕后,加水(50mL)稀释,乙酸乙酯(60mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=1/1)分离,得到化合物2-3。MS-ESI:m/z 213.1[M+H]+At room temperature, 3-amino-5-methylpyrazole (2.50g, 25.74mmol) was dissolved in acetonitrile (25mL), compound 2-2 (8.08g, 38.61mmol, 55% purity) was added, and the reaction mixture was Stir for 4 hours. finished responding After completion, dilute with water (50mL), extract with ethyl acetate (60mL×3), combine the organic phases, dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure. The resulting residue is subjected to silica gel column chromatography (petroleum ether/ethyl acetate Ester = 1/1) separation to obtain compound 2-3. MS-ESI: m/z 213.1 [M+H] + .
3)化合物2-4的合成3) Synthesis of compound 2-4
室温下,将化合物2-3(1.00g,4.43mmol)和20%乙醇钠乙醇溶液(2.26g,6.64mmol)溶于乙醇(30mL)中,反应液在80℃搅拌3小时。反应完毕后,反应液冷却至室温,缓慢滴加到1mol/L盐酸(100mL)水溶液中,过滤,滤饼真空干燥,得到化合物2-4。1H NMR(400MHz,DMSO-d6)δ11.78(s,1H),11.45(s,1H),5.65(s,1H),2.19(s,3H)。Compound 2-3 (1.00 g, 4.43 mmol) and 20% sodium ethoxide ethanol solution (2.26 g, 6.64 mmol) were dissolved in ethanol (30 mL) at room temperature, and the reaction solution was stirred at 80° C. for 3 hours. After the reaction was completed, the reaction liquid was cooled to room temperature, slowly added dropwise to 1 mol/L hydrochloric acid (100 mL) aqueous solution, filtered, and the filter cake was vacuum-dried to obtain compound 2-4. 1 H NMR (400 MHz, DMSO-d 6 ) δ 11.78 (s, 1H), 11.45 (s, 1H), 5.65 (s, 1H), 2.19 (s, 3H).
4)化合物2-5的合成4) Synthesis of compound 2-5
室温下,将化合物2-4(200mg,1.20mmol)和N,N-二乙基苯胺(539mg,3.61mmol)缓慢加到三氯氧磷(2.20mL)中,反应混合物在100℃搅拌3小时。反应完毕后,反应液减压浓缩,得到化合物2-5。粗品不经纯化,直接用于下一步反应。MS-ESI:m/z 203.0[M+H]+At room temperature, compound 2-4 (200mg, 1.20mmol) and N,N-diethylaniline (539mg, 3.61mmol) were slowly added to phosphorus oxychloride (2.20mL), and the reaction mixture was stirred at 100°C for 3 hours . After the reaction, the reaction solution was concentrated under reduced pressure to obtain compound 2-5. The crude product was directly used in the next reaction without purification. MS-ESI: m/z 203.0 [M+H] + .
5)化合物2-6的合成5) Synthesis of compound 2-6
室温下,将化合物2-5(244mg,1.20mmol),中间体A(343mg,1.77mmol)和吡啶(1.00mL,12.04mmol)溶于乙腈(4mL)中,反应混合物在50℃搅拌1小时。反应完毕后,加水(30mL)稀释,乙酸乙酯(50mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=3/1)分离,得到化合物2-6。MS-ESI:m/z 335.9[M+H]+Compound 2-5 (244mg, 1.20mmol), Intermediate A (343mg, 1.77mmol) and pyridine (1.00mL, 12.04mmol) were dissolved in acetonitrile (4mL) at room temperature, and the reaction mixture was stirred at 50°C for 1 hour. After the reaction was completed, dilute with water (30mL), extract with ethyl acetate (50mL×3), combine the organic phases, dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure. The resulting residue was subjected to silica gel column chromatography (petroleum ether/acetic acid Ethyl ester=3/1) separation to obtain compound 2-6. MS-ESI: m/z 335.9 [M+H] + .
6)化合物2的合成6) Synthesis of Compound 2
室温下,将化合物2-6(160mg,0.40mmol,90%纯度),中间体C(89mg,0.40mmol)和氟化铯(123mg,0.81mmol)混于二甲亚砜(4mL)中,反应混合物在60℃搅拌4小时。反应完毕后,加水(20mL)稀释,乙酸乙酯(30mL×3)萃取,合并有机相,用水(15mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶薄层层析(纯甲醇)分离,再经制备级高效液相色谱(甲酸/乙腈/水体系,色谱柱:Unisil 3-100C18Ultra 150*50mm*3μm;流动相:水(0.5%甲酸),乙腈;梯度配比:乙腈相(0-7min,11-41%);流速:25mL/min;柱温:室温)分离,得到化合物2。MS-ESI:m/z 504.3[M+H]+1H NMR(400MHz,DMSO-d6)δ8.99(d,J=7.2Hz,1H),7.65(d,J=8.8Hz,1H),7.59(d,J=2.0Hz,1H),7.43(dd,J=8.4,2.0Hz,1H),5.67(s,1H),5.64-5.53(m,1H),4.02-3.83(m,2H),3.82-3.55(m,4H),3.15-2.86(m,2H),2.64-2.53(m,4H),2.42-2.35(m,1H),2.30(s,3H),1.88-1.58(m,4H),1.52(d,J=7.2Hz,3H),1.06-0.90(m,1H)。At room temperature, compound 2-6 (160 mg, 0.40 mmol, 90% purity), intermediate C (89 mg, 0.40 mmol) and cesium fluoride (123 mg, 0.81 mmol) were mixed in dimethyl sulfoxide (4 mL), and the reaction The mixture was stirred at 60°C for 4 hours. After the reaction is complete, dilute with water (20 mL), extract with ethyl acetate (30 mL×3), combine the organic phases, wash with water (15 mL×3), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure. Thin layer chromatography (pure methanol) separation, and then preparative high performance liquid chromatography (formic acid/acetonitrile/water system, chromatographic column: Unisil 3-100C18Ultra 150*50mm*3μm; mobile phase: water (0.5% formic acid), acetonitrile ; Gradient ratio: acetonitrile phase (0-7min, 11-41%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 2. MS-ESI: m/z 504.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ8.99(d, J=7.2Hz, 1H), 7.65(d, J=8.8Hz, 1H), 7.59(d, J=2.0Hz, 1H), 7.43 (dd,J=8.4,2.0Hz,1H),5.67(s,1H),5.64-5.53(m,1H),4.02-3.83(m,2H),3.82-3.55(m,4H),3.15-2.86 (m,2H),2.64-2.53(m,4H),2.42-2.35(m,1H),2.30(s,3H),1.88-1.58(m,4H),1.52(d,J=7.2Hz,3H ), 1.06-0.90(m,1H).
实施例3:2-((R)-3-(1-(4-(((R)-1-(2,4-二氯苯基)乙基)氨基)-7-(三氟甲基)吡唑[1,5-a][1,3,5]三嗪-2-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(3)
Example 3: 2-((R)-3-(1-(4-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-7-(trifluoromethyl )pyrazol[1,5-a][1,3,5]triazin-2-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (3)
1)化合物3-2的合成1) Synthesis of compound 3-2
室温下,将化合物3-1(1.80g,11.91mmol)和乙氧羰基异硫氰酸酯(1.56g,11.91mmol)溶于乙酸乙酯(20mL)中,反应混合物在80℃搅拌1小时。反应混合物冷却至室温,减压浓缩,所得残余物用石油醚和乙酸乙酯的混合溶剂(10/1,15mL)打浆,过滤,滤饼真空干燥,得到化合物3-2。MS-ESI:m/z 283.1[M+H]+Compound 3-1 (1.80 g, 11.91 mmol) and ethoxycarbonyl isothiocyanate (1.56 g, 11.91 mmol) were dissolved in ethyl acetate (20 mL) at room temperature, and the reaction mixture was stirred at 80° C. for 1 hour. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The resulting residue was slurried with a mixed solvent of petroleum ether and ethyl acetate (10/1, 15 mL), filtered, and the filter cake was dried in vacuum to obtain compound 3-2. MS-ESI: m/z 283.1 [M+H] + .
2)化合物3-3的合成2) Synthesis of compound 3-3
室温下,将化合物3-2(2.00g,6.82mmol)溶于2mol/L氢氧化钠水溶液(20mL)中,室温搅拌2小时。反应完毕后,将反应混合物倒入1mol/L盐酸水溶液(20mL)中,乙酸乙酯(20mL×3)萃取。合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物3-3。MS-ESI:m/z 236.9[M+H]+Compound 3-2 (2.00 g, 6.82 mmol) was dissolved in 2 mol/L sodium hydroxide aqueous solution (20 mL) at room temperature, and stirred at room temperature for 2 hours. After the reaction was completed, the reaction mixture was poured into 1 mol/L hydrochloric acid aqueous solution (20 mL), and extracted with ethyl acetate (20 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound 3-3. MS-ESI: m/z 236.9 [M+H] + .
3)化合物3-4的合成3) Synthesis of compound 3-4
室温下,将化合物3-3(350mg,1.48mmol)和碘甲烷(210mg,1.48mmol)溶于N,N-二甲基甲酰胺(4mL),反应混合物在50℃搅拌5小时。反应混合物冷却到室温,减压浓缩,加水(20mL)稀释,乙酸乙酯(30mL×3)萃取,合并有机相,饱和氯化钠水溶液(15mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物3-4。1H NMR(400MHz,DMSO-d6)δ6.75(s,1H),2.53(s,3H)。Compound 3-3 (350 mg, 1.48 mmol) and iodomethane (210 mg, 1.48 mmol) were dissolved in N,N-dimethylformamide (4 mL) at room temperature, and the reaction mixture was stirred at 50° C. for 5 hours. The reaction mixture was cooled to room temperature, concentrated under reduced pressure, diluted with water (20 mL), extracted with ethyl acetate (30 mL×3), combined the organic phases, washed with saturated aqueous sodium chloride solution (15 mL×3), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain compound 3-4. 1 H NMR (400 MHz, DMSO-d 6 ) δ 6.75 (s, 1H), 2.53 (s, 3H).
4)化合物3-5的合成4) Synthesis of compound 3-5
室温下,将化合物3-4(0.25g,0.10mmol),N,N-二乙基苯胺(0.45g,3.00mmol)和三氯氧磷(1.90mL,19.98mmol)溶于甲苯(2mL)中,反应混合物在100℃搅拌2小时。反应混合物冷却至室温后,减压浓缩,得到化合物3-5。粗品不经过纯化,直接用于下一步反应。MS-ESI:m/z 268.9[M+H]+At room temperature, compound 3-4 (0.25g, 0.10mmol), N,N-diethylaniline (0.45g, 3.00mmol) and phosphorus oxychloride (1.90mL, 19.98mmol) were dissolved in toluene (2mL) , the reaction mixture was stirred at 100°C for 2 hours. After the reaction mixture was cooled to room temperature, it was concentrated under reduced pressure to obtain compound 3-5. The crude product was directly used in the next reaction without further purification. MS-ESI: m/z 268.9 [M+H] + .
5)化合物3-6的合成 5) Synthesis of compound 3-6
室温下,将化合物3-5(268mg,0.10mmol),中间体A(287mg,1.48mmol)和吡啶(0.80mL,9.99mmol)溶于乙腈(10mL)中,反应混合物在50℃搅拌0.5小时。冷却至室温,减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=20/1)分离,得到化合物3-6。MS-ESI:m/z 422.0[M+H]+Compound 3-5 (268mg, 0.10mmol), Intermediate A (287mg, 1.48mmol) and pyridine (0.80mL, 9.99mmol) were dissolved in acetonitrile (10mL) at room temperature, and the reaction mixture was stirred at 50°C for 0.5 hours. Cooled to room temperature, concentrated under reduced pressure, and the obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to obtain compound 3-6. MS-ESI: m/z 422.0 [M+H] + .
6)化合物3-7的合成6) Synthesis of compound 3-7
室温下,将化合物3-6(180mg,0.43mmol)溶于二氯甲烷(10mL)中,分批加入间氯过氧苯甲酸(260mg,1.28mmol,85%纯度),反应混合物在20℃搅拌3小时。反应完毕后,反应液经硅胶柱层析(石油醚/乙酸乙酯=5/1)分离,得到化合物3-7。MS-ESI:m/z 453.9[M+H]+1H NMR(400MHz,CDCl3)δ7.48(d,J=2.0Hz,1H),7.42(d,J=8.4Hz,1H),7.37(d,J=7.6Hz,1H),7.31(dd,J=8.4,2.0Hz,1H),6.92(s,1H),5.90-5.77(m,1H),3.26(s,3H),1.80(d,J=7.2Hz,3H)。At room temperature, compound 3-6 (180mg, 0.43mmol) was dissolved in dichloromethane (10mL), m-chloroperoxybenzoic acid (260mg, 1.28mmol, 85% purity) was added in portions, and the reaction mixture was stirred at 20°C 3 hours. After the reaction was completed, the reaction solution was separated by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to obtain compound 3-7. MS-ESI: m/z 453.9 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ7.48(d, J=2.0Hz, 1H), 7.42(d, J=8.4Hz, 1H), 7.37(d, J=7.6Hz, 1H), 7.31(dd , J=8.4, 2.0Hz, 1H), 6.92(s, 1H), 5.90-5.77(m, 1H), 3.26(s, 3H), 1.80(d, J=7.2Hz, 3H).
7)化合物3的合成7) Synthesis of compound 3
室温下,将化合物3-7(200mg,0.44mmol)溶于乙腈(8mL)中,加入中间体C(194mg,0.88mmol)和N,N-二异丙基乙胺(227mg,1.76mmol),反应混合物在50℃搅拌3小时。反应完毕后,冷却到室温,减压浓缩,剩余物经制备级高效液相色谱(碳酸氢铵/乙腈/水体系,色谱柱:Waters Xbridge C18 150*25mm*5μm;流动相:水(10mM碳酸氢铵),乙腈;梯度配比:乙腈相(0-10min,52-82%);流速:25mL/min;柱温:室温)分离,得到化合物3。MS-ESI:m/z 558.0[M+H]+1H NMR(400MHz,CD3OD)δ7.54-7.45(m,2H),7.31(dd,J=8.8,2.4Hz,1H),6.07(s,1H),5.70-5.57(m,1H),4.20-3.91(m,2H),3.86-3.50(m,4H),3.00-2.83(m,2H),2.54(t,J=5.6Hz,2H),2.46-2.36(m,1H),2.04(t,J=11.6Hz,1H),1.86-1.68(m,4H),1.65-1.53(m,4H),0.97-0.80(m,1H)。Compound 3-7 (200mg, 0.44mmol) was dissolved in acetonitrile (8mL) at room temperature, Intermediate C (194mg, 0.88mmol) and N,N-diisopropylethylamine (227mg, 1.76mmol) were added, The reaction mixture was stirred at 50°C for 3 hours. After the reaction is completed, cool to room temperature, concentrate under reduced pressure, and the residue is subjected to preparative high-performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge C18 150*25mm*5 μm; mobile phase: water (10mM carbonic acid ammonium hydrogen), acetonitrile; gradient ratio: acetonitrile phase (0-10min, 52-82%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 3. MS-ESI: m/z 558.0 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.54-7.45(m, 2H), 7.31(dd, J=8.8, 2.4Hz, 1H), 6.07(s, 1H), 5.70-5.57(m, 1H) ,4.20-3.91(m,2H),3.86-3.50(m,4H),3.00-2.83(m,2H),2.54(t,J=5.6Hz,2H),2.46-2.36(m,1H),2.04 (t, J=11.6Hz, 1H), 1.86-1.68(m, 4H), 1.65-1.53(m, 4H), 0.97-0.80(m, 1H).
实施例4:2-((R)-3-(1-(7-氯-4-(((R)-1-(2,4-二氯苯基)乙基)氨基)吡唑[1,5-a][1,3,5]三嗪-2-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(4)
Example 4: 2-((R)-3-(1-(7-chloro-4-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)pyrazol[1 ,5-a][1,3,5]triazin-2-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (4)
1)化合物4-2的合成1) Synthesis of compound 4-2
室温下,将化合物4-1(6.50g,39.88mmol)和乙氧羰基异硫氰酸酯(6.28g,47.86mmol)溶于乙酸乙酯(50mL),氮气置换,反应混合物在80℃搅拌1小时。冷却至室温,过滤,滤饼真空干燥,得到化合物4-2。MS-ESI:m/z 292.9[M+H]+At room temperature, compound 4-1 (6.50g, 39.88mmol) and ethoxycarbonyl isothiocyanate (6.28g, 47.86mmol) were dissolved in ethyl acetate (50mL), replaced with nitrogen, and the reaction mixture was stirred at 80°C for 1 Hour. Cool to room temperature, filter, and vacuum-dry the filter cake to obtain compound 4-2. MS-ESI: m/z 292.9 [M+H] + .
2)化合物4-3的合成2) Synthesis of compound 4-3
室温下,将化合物4-2(17.00g,50.45mmol,92%纯度)溶于氢氧化钠水溶液(50mL,2mol/L),反应混合物在室温搅拌2小时。反应完毕后,将反应混合物倒入1mol/L盐酸水溶液(100mL)中,过滤,滤饼真空干燥,得到化合物4-3。MS-ESI:m/z 246.9[M+H]+Compound 4-2 (17.00 g, 50.45 mmol, 92% purity) was dissolved in aqueous sodium hydroxide solution (50 mL, 2 mol/L) at room temperature, and the reaction mixture was stirred at room temperature for 2 hours. After the reaction was completed, the reaction mixture was poured into 1 mol/L hydrochloric acid aqueous solution (100 mL), filtered, and the filter cake was vacuum-dried to obtain compound 4-3. MS-ESI: m/z 246.9 [M+H] + .
3)化合物4-4的合成3) Synthesis of compound 4-4
室温下,将化合物4-3(13.40g,53.15mmol)和碘甲烷(1.89g,13.28mmol)溶于N,N-二甲基甲酰胺(150mL),反应混合物在室温搅拌4小时,然后在50℃搅拌3小时。反应完毕后,将反应混合物冷却至室温,减压浓缩。加水(300mL)稀释,乙酸乙酯(150mL×3)萃取,饱和氯化钠水溶液(500mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物4-4。1H NMR(400MHz,DMSO-d6)δ13.08(brs,1H),6.53(s,1H),2.53(s,3H).At room temperature, compound 4-3 (13.40g, 53.15mmol) and iodomethane (1.89g, 13.28mmol) were dissolved in N,N-dimethylformamide (150mL), and the reaction mixture was stirred at room temperature for 4 hours, and then Stir at 50°C for 3 hours. After the reaction was completed, the reaction mixture was cooled to room temperature and concentrated under reduced pressure. Dilute with water (300 mL), extract with ethyl acetate (150 mL×3), wash with saturated aqueous sodium chloride solution (500 mL×3), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure to obtain compound 4-4. 1 H NMR (400MHz,DMSO-d 6 )δ13.08(brs,1H),6.53(s,1H),2.53(s,3H).
4)化合物4-5的合成4) Synthesis of compound 4-5
室温下,将化合物4-4(1.50g,5.75mmol),N,N-二乙基苯胺(2.57g,17.23mmol)和三氯氧磷(10.70mL,114.90mmol)溶于甲苯(20mL)中,反应混合物在100℃搅拌2小时。反应完毕后,将反应混合物冷却至室温,减压浓缩,得到化合物4-5。粗品不经过纯化,直接用于下一步反应。MS-ESI:m/z 278.8[M+H]+At room temperature, compound 4-4 (1.50g, 5.75mmol), N,N-diethylaniline (2.57g, 17.23mmol) and phosphorus oxychloride (10.70mL, 114.90mmol) were dissolved in toluene (20mL) , the reaction mixture was stirred at 100°C for 2 hours. After completion of the reaction, the reaction mixture was cooled to room temperature and concentrated under reduced pressure to obtain compound 4-5. The crude product was directly used in the next reaction without further purification. MS-ESI: m/z 278.8 [M+H] + .
5)化合物4-6的合成 5) Synthesis of compound 4-6
室温下,将化合物4-5(1.60g,5.74mmol),中间体A(1.65g,8.62mmol)和吡啶(4.60mL,57.45mmol)溶于乙腈(30mL)中,反应混合物在50℃搅拌0.5小时。反应完毕后,反应混合物冷却到室温,减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=20/1)分离,得到化合物4-6。MS-ESI:m/z 431.9[M+H]+At room temperature, compound 4-5 (1.60g, 5.74mmol), intermediate A (1.65g, 8.62mmol) and pyridine (4.60mL, 57.45mmol) were dissolved in acetonitrile (30mL), and the reaction mixture was stirred at 50°C for 0.5 Hour. After the reaction was completed, the reaction mixture was cooled to room temperature and concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to obtain compound 4-6. MS-ESI: m/z 431.9 [M+H] + .
6)化合物4-7的合成6) Synthesis of compound 4-7
室温下,将中间体4-6(400mg,0.92mmol)溶于1-甲基-2-吡咯烷酮(15mL)中,加入氯化亚铜(457mg,4.62mmol),氮气置换,微波加热至150℃反应16小时。反应液冷却至室温,加水(40mL)和乙酸乙酯(80mL)稀释,过滤,滤饼用乙酸乙酯(10mL×3)洗涤,滤液用乙酸乙酯(20mL×3)萃取,无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经制备级高效液相色谱(盐酸/乙腈/水体系,色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:水(0.225%甲酸),乙腈;梯度配比:乙腈相(0-10min,70-100%);流速:25mL/min;柱温:室温)分离,得到化合物4-7。MS-ESI:m/z 388.0[M+H]+Dissolve Intermediate 4-6 (400mg, 0.92mmol) in 1-methyl-2-pyrrolidone (15mL) at room temperature, add cuprous chloride (457mg, 4.62mmol), replace with nitrogen, and heat to 150°C with microwave React for 16 hours. The reaction solution was cooled to room temperature, diluted with water (40mL) and ethyl acetate (80mL), filtered, the filter cake was washed with ethyl acetate (10mL×3), the filtrate was extracted with ethyl acetate (20mL×3), anhydrous sodium sulfate Dry, filter, and concentrate the filtrate under reduced pressure. The resulting residue is subjected to preparative high-performance liquid chromatography (hydrochloric acid/acetonitrile/water system, chromatographic column: Phenomenex luna C18 150*25mm*10 μm; mobile phase: water (0.225% formic acid), acetonitrile ; Gradient ratio: acetonitrile phase (0-10min, 70-100%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 4-7. MS-ESI: m/z 388.0 [M+H] + .
7)化合物4-8的合成7) Synthesis of compound 4-8
室温下,将化合物4-7(20mg,0.05mmol)溶于二氯甲烷(2mL),加入间氯过氧苯甲酸(31mg,0.15mmol,85%纯度),反应混合物在25℃搅拌1.5小时。反应液过滤,滤液经硅胶柱层析(石油醚/乙酸乙酯=2/1)分离,得到化合物4-8。MS-ESI:m/z 419.8[M+H]+Compound 4-7 (20 mg, 0.05 mmol) was dissolved in dichloromethane (2 mL) at room temperature, m-chloroperoxybenzoic acid (31 mg, 0.15 mmol, 85% purity) was added, and the reaction mixture was stirred at 25°C for 1.5 hours. The reaction solution was filtered, and the filtrate was separated by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to obtain compound 4-8. MS-ESI: m/z 419.8 [M+H] + .
8)化合物4的合成8) Synthesis of Compound 4
室温下,将化合物4-8(12mg,0.03mmol)和中间体C(8mg,0.03mmol)溶于乙腈(2mL)中,加入二异丙基乙胺(37mg,0.29mmol),反应混合物在50℃反应2小时。反应液冷却至室温,减压浓缩,所得残余物经硅胶制备薄层色谱(乙酸乙酯/甲醇=1/1)分离,得到化合物4。MS-ESI:m/z 524.1[M+H]+1H NMR(400MHz,CD3OD)δ7.50-7.45(m,2H),7.31(dd,J=8.4,2.0Hz,1H),5.78(s,1H),5.64-5.57(m,1H),4.15-4.06(m,1H),4.05-3.94(m,1H),3.85-3.71(m,3H),3.19-3.03(m,2H),2.80-2.72(m,2H),2.48-2.37(m,1H),2.35-2.25(m,1H),2.07-1.96(m,1H),1.87-1.72(m,3H),1.70-1.62(m,1H),1.58(d,J=6.8Hz,3H),1.35-1.27(m,1H),1.03-0.89(m,1H)。At room temperature, compound 4-8 (12mg, 0.03mmol) and intermediate C (8mg, 0.03mmol) were dissolved in acetonitrile (2mL), diisopropylethylamine (37mg, 0.29mmol) was added, and the reaction mixture was heated at 50 °C for 2 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the obtained residue was separated by preparative thin-layer silica gel chromatography (ethyl acetate/methanol=1/1) to obtain compound 4. MS-ESI: m/z 524.1 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.50-7.45 (m, 2H), 7.31 (dd, J = 8.4, 2.0Hz, 1H), 5.78 (s, 1H), 5.64-5.57 (m, 1H) ,4.15-4.06(m,1H),4.05-3.94(m,1H),3.85-3.71(m,3H),3.19-3.03(m,2H),2.80-2.72(m,2H),2.48-2.37( m,1H),2.35-2.25(m,1H),2.07-1.96(m,1H),1.87-1.72(m,3H),1.70-1.62(m,1H),1.58(d,J=6.8Hz, 3H), 1.35-1.27(m, 1H), 1.03-0.89(m, 1H).
实施例5:4-(((R)-1-(2,4-二氯苯基)乙基)氨基)-2-(3-((R)-1-(2-羟乙基)哌啶-3-基)氮杂环丁烷-1-基)吡唑[1,5-a][1,3,5]三嗪-7-甲腈(5)
Example 5: 4-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-(3-((R)-1-(2-hydroxyethyl)piper Pyridin-3-yl)azetidin-1-yl)pyrazol[1,5-a][1,3,5]triazine-7-carbonitrile (5)
1)化合物5-1的合成1) Synthesis of Compound 5-1
室温下,将中间体4-6(1.50g,3.43mmol),1,1-双(二苯基磷)二茂铁氯化钯(0.25g,0.34mmol)和三乙胺(1.40mL,10.28mmol)溶于甲醇(20mL)中,反应混合物在一氧化碳(50psi)氛围下,80℃搅拌16小时。反应混合物冷却至室温,减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=5/1)分离,得到化合物5-1。MS-ESI:m/z 412.0[M+H]+At room temperature, Intermediate 4-6 (1.50g, 3.43mmol), 1,1-bis(diphenylphosphino)ferrocenepalladium chloride (0.25g, 0.34mmol) and triethylamine (1.40mL, 10.28 mmol) was dissolved in methanol (20 mL), and the reaction mixture was stirred at 80° C. for 16 hours under an atmosphere of carbon monoxide (50 psi). The reaction mixture was cooled to room temperature, concentrated under reduced pressure, and the resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to obtain compound 5-1. MS-ESI: m/z 412.0 [M+H] + .
2)化合物5-2的合成2) Synthesis of compound 5-2
室温下,将甲醇(5mL)置于100mL三口瓶中,氮气置换,将反应液的温度降至-65℃,通入氨气。室温下,加入化合物5-1(400mg,0.97mmol),反应混合物在20℃搅拌16小时。反应液减压浓缩,所得残余物用石油醚和甲基叔丁基醚的混合溶剂(1/1,20mL)打浆,过滤,滤饼真空干燥,得到化合物5-2。MS-ESI:m/z397.0[M+H]+At room temperature, methanol (5 mL) was placed in a 100 mL three-neck flask, replaced with nitrogen, the temperature of the reaction solution was lowered to -65° C., and ammonia gas was introduced. At room temperature, compound 5-1 (400mg, 0.97mmol) was added, and the reaction mixture was stirred at 20°C for 16 hours. The reaction solution was concentrated under reduced pressure, and the resulting residue was slurried with a mixed solvent of petroleum ether and methyl tert-butyl ether (1/1, 20 mL), filtered, and the filter cake was vacuum-dried to obtain compound 5-2. MS-ESI: m/z 397.0 [M+H] + .
3)化合物5-3的合成3) Synthesis of compound 5-3
室温下,将化合物5-2(330mg,0.83mmol),伯吉斯试剂(396mg,1.66mmol)溶于无水二氯甲烷(10mL)中,反应混合物在20℃搅拌16小时。将反应混合物倒入水(20mL)中,二氯甲烷(20mL×2)萃取,有机相依次用水(20mL),饱和氯化钠水溶液(20mL)洗涤,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=3/1)分离,得到化合物5-3。1H NMR(400MHz,DMSO-d6)δ9.99(s,1H),7.63(d,J=8.4Hz,1H),7.60(s,1H),7.43(d,J=8.4Hz,1H),6.97(s,1H),5.70-5.58(m,1H),2.41(s,3H),1.56(d,J=6.8Hz,3H)。Compound 5-2 (330 mg, 0.83 mmol), Burgess reagent (396 mg, 1.66 mmol) were dissolved in anhydrous dichloromethane (10 mL) at room temperature, and the reaction mixture was stirred at 20°C for 16 hours. Pour the reaction mixture into water (20mL), extract with dichloromethane (20mL×2), wash the organic phase with water (20mL) and saturated aqueous sodium chloride solution (20mL) successively, combine the organic phases, dry over anhydrous sodium sulfate, and filter , and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to obtain compound 5-3. 1 H NMR (400MHz,DMSO-d 6 )δ9.99(s,1H),7.63(d,J=8.4Hz,1H),7.60(s,1H),7.43(d,J=8.4Hz,1H) , 6.97 (s, 1H), 5.70-5.58 (m, 1H), 2.41 (s, 3H), 1.56 (d, J=6.8Hz, 3H).
4)化合物5-4的合成4) Synthesis of Compound 5-4
室温下,将化合物5-3(180mg,0.48mmol),间氯过氧苯甲酸(289mg,1.42mmol,85%纯度)溶于二氯甲烷(3mL)中,反应混合物在20℃搅拌1小时。反应液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=3/1)分离,得到化合物5-4。MS-ESI:m/z 411.0[M+H]+Compound 5-3 (180 mg, 0.48 mmol), m-chloroperoxybenzoic acid (289 mg, 1.42 mmol, 85% purity) were dissolved in dichloromethane (3 mL) at room temperature, and the reaction mixture was stirred at 20° C. for 1 hour. The reaction solution was concentrated under reduced pressure, and the obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to obtain compound 5-4. MS-ESI: m/z 411.0 [M+H] + .
5)化合物5的合成 5) Synthesis of Compound 5
室温下,将化合物5-4(190mg,0.38mmol,83%纯度),中间体C(171mg,0.77mmol),N,N-二异丙基乙胺(0.20mL,1.15mmol)溶于乙腈(5mL)中,反应混合物在50℃搅拌2小时。反应液冷却至室温,减压浓缩,所得残余物经制备级高效液相色谱(碳酸氢铵/乙腈/水体系,色谱柱:Waters Xbridge 150*25mm*5μm;流动相:水(10mM碳酸氢铵),乙腈;梯度配比:乙腈相(0-8min,52-82%);流速:25mL/min;柱温:室温)分离,得到化合物5。MS-ESI:m/z 515.2[M+H]+1H NMR(400MHz,CD3OD)δ7.52-7.42(m,2H),7.28(dd,J=8.4,2.0Hz,1H),6.18(s,1H),5.70-5.52(m,1H),4.16-3.90(m,2H),3.85-3.45(m,4H),2.99-2.78(m,2H),2.60-2.48(m,2H),2.45-2.30(m,1H),2.01(t,J=10.8Hz,1H),1.87-1.65(m,4H),1.63-1.51(m,4H),0.96-0.76(m,1H)。At room temperature, compound 5-4 (190mg, 0.38mmol, 83% purity), intermediate C (171mg, 0.77mmol), N,N-diisopropylethylamine (0.20mL, 1.15mmol) were dissolved in acetonitrile ( 5 mL), the reaction mixture was stirred at 50 °C for 2 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the resulting residue was subjected to preparative high-performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge 150*25mm*5 μm; mobile phase: water (10mM ammonium bicarbonate ), acetonitrile; gradient ratio: acetonitrile phase (0-8min, 52-82%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 5. MS-ESI: m/z 515.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.52-7.42(m, 2H), 7.28(dd, J=8.4, 2.0Hz, 1H), 6.18(s, 1H), 5.70-5.52(m, 1H) ,4.16-3.90(m,2H),3.85-3.45(m,4H),2.99-2.78(m,2H),2.60-2.48(m,2H),2.45-2.30(m,1H),2.01(t, J=10.8Hz, 1H), 1.87-1.65(m, 4H), 1.63-1.51(m, 4H), 0.96-0.76(m, 1H).
实施例6:2-((R)-3-(1-(4-(((R)-1-(2,4-二氯苯基)乙基)氨基)-7-氟吡唑[1,5-a][1,3,5]三嗪-2-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(6)
Example 6: 2-((R)-3-(1-(4-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-7-fluoropyrazol[1 ,5-a][1,3,5]triazin-2-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (6)
参照实施例3的合成方法制备化合物6。MS-ESI:m/z 508.2[M+H]+1H NMR(400MHz,CD3OD)δ8.55(s,1H),7.52-7.46(m,2H),7.33(dd,J=8.4,2.0Hz,1H),5.67-5.58(m,1H),5.45(d,J=4.8Hz,1H),4.19-4.09(m,1H),4.08-3.93(m,1H),3.91-3.53(m,4H),3.43-3.35(m,1H),3.30-3.22(m,1H),3.06-2.91(m,2H),2.67-2.53(m,1H),2.51-2.40(m,1H),2.39-2.23(m,1H),1.98-1.68(m,4H),1.59(d,J=7.2Hz,3H),1.14-1.10(m,1H)。Compound 6 was prepared according to the synthesis method of Example 3. MS-ESI: m/z 508.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.55(s, 1H), 7.52-7.46(m, 2H), 7.33(dd, J=8.4, 2.0Hz, 1H), 5.67-5.58(m, 1H) ,5.45(d,J=4.8Hz,1H),4.19-4.09(m,1H),4.08-3.93(m,1H),3.91-3.53(m,4H),3.43-3.35(m,1H),3.30 -3.22(m,1H),3.06-2.91(m,2H),2.67-2.53(m,1H),2.51-2.40(m,1H),2.39-2.23(m,1H),1.98-1.68(m, 4H), 1.59 (d, J=7.2Hz, 3H), 1.14-1.10 (m, 1H).
实施例7:2-((R)-3-(1-(8-氯-4-(((R)-1-(2,4-二氯苯基)乙基)氨基)吡唑[1,5-a][1,3,5]三嗪-2-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(7)
Example 7: 2-((R)-3-(1-(8-chloro-4-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)pyrazol[1 ,5-a][1,3,5]triazin-2-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (7)
1)化合物7-1的合成1) Synthesis of compound 7-1
室温下,将化合物1-3(300mg,0.80mmol)溶于氯仿(6mL)中,随后加入N-氯代丁二酰亚胺(129mg,0.97mmol),反应混合物在50℃搅拌6小时。反应液冷却 至室温,减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=2/1)分离,得到化合物7-1。MS-ESI:m/z 389.9[M+H]+Compound 1-3 (300mg, 0.80mmol) was dissolved in chloroform (6mL) at room temperature, then N-chlorosuccinimide (129mg, 0.97mmol) was added, and the reaction mixture was stirred at 50°C for 6 hours. Reaction liquid cooling After reaching room temperature, it was concentrated under reduced pressure, and the resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to obtain compound 7-1. MS-ESI: m/z 389.9 [M+H] + .
2)化合物7-2的合成2) Synthesis of Compound 7-2
室温下,将化合物7-1(250mg,0.49mmol,76%纯度)溶于二氯甲烷(10mL)中,随后加入间氯过氧苯甲酸(298mg,1.47mmol,85%纯度),反应液在20℃搅拌3小时。反应结束后,反应液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=1/1)分离,得到化合物7-2。MS-ESI:m/z 421.9[M+H]+At room temperature, compound 7-1 (250mg, 0.49mmol, 76% purity) was dissolved in dichloromethane (10mL), then m-chloroperoxybenzoic acid (298mg, 1.47mmol, 85% purity) was added, and the reaction solution was Stir at 20°C for 3 hours. After the reaction, the reaction solution was concentrated under reduced pressure, and the obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to obtain compound 7-2. MS-ESI: m/z 421.9 [M+H] + .
3)化合物7的合成3) Synthesis of compound 7
室温下,将化合物7-2(170mg,0.39mmol),中间体C(131mg,0.59mmol),N,N-二异丙基乙胺(203mg,1.57mmol)溶于乙腈(6mL)和二甲基亚砜(2mL)中,反应液在30℃搅拌16小时。反应液减压浓缩,所得残余物经制备级高效液相色谱(碳酸氢铵/乙腈/水体系,色谱柱:Waters Xbridge C18 150*25mm*5μm;流动相:水(10mM碳酸氢铵),乙腈;梯度配比:乙腈相(0-10min,50-80%);流速:25mL/min;柱温:室温)分离,得到化合物7。MS-ESI:m/z 524.0[M+H]+1H NMR(400MHz,CD3OD)δ7.78(s,1H),7.52-7.46(m,2H),7.30(dd,J=8.4,2.0Hz,1H),5.68-5.57(m,1H),4.28-3.74(m,4H),3.70(t,J=6.0Hz,2H),3.01-2.85(m,2H),2.55(t,J=6.0Hz,2H),2.45-2.33(m,1H),2.14-1.99(m,1H),1.86-1.67(m,4H),1.66-1.55(m,4H),0.97-0.81(m,1H)。At room temperature, compound 7-2 (170mg, 0.39mmol), intermediate C (131mg, 0.59mmol), N,N-diisopropylethylamine (203mg, 1.57mmol) were dissolved in acetonitrile (6mL) and dimethyl sulfoxide (2 mL), and the reaction solution was stirred at 30°C for 16 hours. The reaction solution was concentrated under reduced pressure, and the resulting residue was subjected to preparative high-performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge C18 150*25mm*5μm; mobile phase: water (10mM ammonium bicarbonate), acetonitrile ; Gradient ratio: acetonitrile phase (0-10min, 50-80%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 7. MS-ESI: m/z 524.0 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.78(s, 1H), 7.52-7.46(m, 2H), 7.30(dd, J=8.4, 2.0Hz, 1H), 5.68-5.57(m, 1H) ,4.28-3.74(m,4H),3.70(t,J=6.0Hz,2H),3.01-2.85(m,2H),2.55(t,J=6.0Hz,2H),2.45-2.33(m,1H ), 2.14-1.99 (m, 1H), 1.86-1.67 (m, 4H), 1.66-1.55 (m, 4H), 0.97-0.81 (m, 1H).
实施例8:4-(((R)-1-(2,4-二氯苯基)乙基)氨基)-2-(3-((R)-1-(2-羟乙基)哌啶-3-基)氮杂环丁烷-1-基)吡唑[1,5-a][1,3,5]三嗪-8-甲腈(8)
Example 8: 4-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-(3-((R)-1-(2-hydroxyethyl)piper Pyridin-3-yl)azetidin-1-yl)pyrazol[1,5-a][1,3,5]triazine-8-carbonitrile (8)
1)化合物8-1的合成1) Synthesis of Compound 8-1
室温下,将化合物1-3(300mg,0.85mmol)溶于N,N-二甲基甲酰胺(5mL)中,冰浴降到0℃,随后加入N-碘代丁二酰亚胺(191mg,0.85mmol),反应混合物在25℃搅拌2小时。反应完毕后,反应液冷却至室温,倒入水(50mL)中稀释,用乙酸乙酯(10mL×2)萃取。合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=3/1)分离,得到化合物8-1。MS-ESI:m/z 480.0[M+H]+At room temperature, compound 1-3 (300mg, 0.85mmol) was dissolved in N,N-dimethylformamide (5mL), the ice bath was lowered to 0°C, and N-iodosuccinimide (191mg , 0.85mmol), the reaction mixture was stirred at 25°C for 2 hours. After the reaction was completed, the reaction solution was cooled to room temperature, poured into water (50 mL) for dilution, and extracted with ethyl acetate (10 mL×2). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to obtain compound 8-1. MS-ESI: m/z 480.0 [M+H] + .
2)化合物8-2的合成2) Synthesis of Compound 8-2
室温下,将化合物8-1(300mg,0.62mmol)加入1-甲基-2-吡咯烷酮(2mL)中,随后加入氰化亚銅(277mg,3.09mmol),氮气置换,140℃反应3小时。反应完毕后,反应液冷却至室温,倒入水(30mL)中稀释,用乙酸乙酯(20mL×2)萃取,合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=1/2)分离,粗品再用甲基叔丁基醚(30mL)溶解,用饱和食盐水(10mL×4)洗涤,有机相经无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物8-2。MS-ESI:m/z 379.3[M+H]+At room temperature, compound 8-1 (300 mg, 0.62 mmol) was added to 1-methyl-2-pyrrolidone (2 mL), followed by cuprous cyanide (277 mg, 3.09 mmol), replaced by nitrogen, and reacted at 140°C for 3 hours. After the reaction was completed, the reaction solution was cooled to room temperature, poured into water (30 mL) for dilution, extracted with ethyl acetate (20 mL×2), combined the organic phases, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/2), and the crude product was dissolved in methyl tert-butyl ether (30 mL), washed with saturated brine (10 mL×4), and the organic phase was washed with Dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure to obtain compound 8-2. MS-ESI: m/z 379.3 [M+H] + .
3)化合物8-3的合成3) Synthesis of Compound 8-3
室温下,将化合物8-2(45mg,0.08mmol,70%纯度)溶于二氯甲烷(5mL)中,加入间氯过氧苯甲酸(34mg,0.17mmol,85%纯度),反应混合物在25℃搅拌2小时。反应完毕后,反应液经硅胶柱层析(石油醚/乙酸乙酯=1/2)分离,得到化合物8-3。MS-ESI:m/z 411.2[M+H]+At room temperature, compound 8-2 (45mg, 0.08mmol, 70% purity) was dissolved in dichloromethane (5mL), m-chloroperoxybenzoic acid (34mg, 0.17mmol, 85% purity) was added, and the reaction mixture was heated at 25 °C and stirred for 2 hours. After the reaction was completed, the reaction solution was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/2) to obtain compound 8-3. MS-ESI: m/z 411.2 [M+H] + .
4)化合物8的合成4) Synthesis of Compound 8
室温下,将化合物8-3(40mg,0.08mmol,85%纯度)和中间体C(37mg,0.17mmol)溶于1,2-二氯乙烷(5mL)中,加入N,N-二异丙基乙胺(53mg,0.41mmol),反应液在80℃搅拌12小时。反应完毕后,反应液减压浓缩。所得残余物经硅胶制备薄层色谱(纯甲醇)分离,得到化合物8。MS-ESI:m/z 515.2[M+H]+1H NMR(400MHz,CD3OD)δ8.08(s,1H),7.51-7.43(m,2H),7.31(dd,J=8.4,2.0Hz,1H),5.68-5.58(m,1H),4.28-4.15(m,1H),4.04-3.78(m,4H),3.66-3.53(m,1H),3.52-3.40(m,1H),3.24-3.16(m,2H),2.95-2.80(m,1H),2.78-2.55(m,1H),2.54-2.43(m,1H),2.06-1.93(m,2H),1.92-1.71(m,2H),1.59(d,J=6.8Hz,3H),1.33-1.30(m,1H),1.21-1.07(m,1H)。At room temperature, compound 8-3 (40 mg, 0.08 mmol, 85% purity) and intermediate C (37 mg, 0.17 mmol) were dissolved in 1,2-dichloroethane (5 mL), and N,N-diiso Propylethylamine (53mg, 0.41mmol), the reaction solution was stirred at 80°C for 12 hours. After the reaction was completed, the reaction solution was concentrated under reduced pressure. The obtained residue was separated by silica gel preparative thin-layer chromatography (pure methanol) to obtain compound 8. MS-ESI: m/z 515.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.08(s, 1H), 7.51-7.43(m, 2H), 7.31(dd, J=8.4, 2.0Hz, 1H), 5.68-5.58(m, 1H) ,4.28-4.15(m,1H),4.04-3.78(m,4H),3.66-3.53(m,1H),3.52-3.40(m,1H),3.24-3.16(m,2H),2.95-2.80( m,1H),2.78-2.55(m,1H),2.54-2.43(m,1H),2.06-1.93(m,2H),1.92-1.71(m,2H),1.59(d,J=6.8Hz, 3H), 1.33-1.30(m, 1H), 1.21-1.07(m, 1H).
实施例9:(1R,3r)-3-((R)-3-(1-(5-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑[1,5-a]吡啶-7-基)氮杂环丁烷-3-基)哌啶-1-基)-1-甲基环丁烷-1-甲酸(9)
Example 9: (1R,3r)-3-((R)-3-(1-(5-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[ 1,2,4]triazol[1,5-a]pyridin-7-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclobutane-1-carboxylic acid (9)
1)化合物9-2的合成1) Synthesis of compound 9-2
室温下,将化合物9-1(8.00g,35.26mmol),(2,4-二甲氧苯基)甲胺(8.84g,52.89mmol)和氟化铯(10.71g,70.52mmol)加入二甲基亚砜(50mL)中,反应液在 100℃搅拌16小时。反应液加水稀释(50mL),用乙酸乙酯萃取(80mL×2),有机相用饱和食盐水洗涤(50mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩。剩余物经硅胶柱层析(石油醚/乙酸乙酯=20/1)分离,得到化合物9-2。MS-ESI:m/z 357.0[M+H]+At room temperature, compound 9-1 (8.00g, 35.26mmol), (2,4-dimethoxyphenyl) methylamine (8.84g, 52.89mmol) and cesium fluoride (10.71g, 70.52mmol) were added into dimethyl In base sulfoxide (50mL), the reaction solution was Stir at 100°C for 16 hours. The reaction solution was diluted with water (50 mL), extracted with ethyl acetate (80 mL×2), the organic phase was washed with saturated brine (50 mL×2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to obtain compound 9-2. MS-ESI: m/z 357.0 [M+H] + .
2)化合物9-3的合成2) Synthesis of compound 9-3
室温下,将化合物9-2(1.80g,4.58mmol,91%纯度)加入二氯甲烷(10mL)中,缓慢滴加三氟乙酸(10.00mL),反应液在室温搅拌5小时。反应液减压浓缩,用甲基叔丁基醚和石油醚(1/1,100mL)打浆,过滤,滤液减压浓缩,得到化合物9-3。MS-ESI:m/z 207.0[M+H]+At room temperature, compound 9-2 (1.80 g, 4.58 mmol, 91% purity) was added into dichloromethane (10 mL), trifluoroacetic acid (10.00 mL) was slowly added dropwise, and the reaction solution was stirred at room temperature for 5 hours. The reaction solution was concentrated under reduced pressure, slurried with methyl tert-butyl ether and petroleum ether (1/1, 100 mL), filtered, and the filtrate was concentrated under reduced pressure to obtain compound 9-3. MS-ESI: m/z 207.0 [M+H] + .
3)化合物9-4的合成3) Synthesis of Compound 9-4
室温下,依次将化合物9-3(1.50g,3.73mmol,80%纯度)和乙氧羰基异硫氰酸酯(0.98g,7.47mmol)加入到二氧六环(15mL)中,反应液室温搅拌16小时。反应液减压浓缩,粗品用甲基叔丁基醚和石油醚(1/1,50mL)打浆,过滤,固体干燥得到化合物9-4。MS-ESI:m/z 337.9[M+H]+At room temperature, sequentially add compound 9-3 (1.50g, 3.73mmol, 80% purity) and ethoxycarbonyl isothiocyanate (0.98g, 7.47mmol) into dioxane (15mL), and the reaction solution was Stir for 16 hours. The reaction solution was concentrated under reduced pressure, and the crude product was slurried with methyl tert-butyl ether and petroleum ether (1/1, 50 mL), filtered, and dried to obtain compound 9-4. MS-ESI: m/z 337.9 [M+H] + .
4)化合物9-5的合成4) Synthesis of compound 9-5
室温下,依次将化合物9-4(1.10g,3.09mmol,95%纯度),N,N-二异丙基乙胺(1.50mL,9.26mmol)和盐酸羟胺(0.64g,9.26mmol)加入到甲醇(10mL)和乙醇(10mL)中,反应混合物在60℃反应16小时。反应液减压浓缩,所得粗品用水(40mL)打浆,过滤,固体干燥得到化合物9-5。MS-ESI:m/z 246.9[M+H]+At room temperature, compound 9-4 (1.10g, 3.09mmol, 95% purity), N,N-diisopropylethylamine (1.50mL, 9.26mmol) and hydroxylamine hydrochloride (0.64g, 9.26mmol) were added to In methanol (10 mL) and ethanol (10 mL), the reaction mixture was reacted at 60° C. for 16 hours. The reaction solution was concentrated under reduced pressure, and the resulting crude product was slurried with water (40 mL), filtered, and dried to obtain compound 9-5. MS-ESI: m/z 246.9 [M+H] + .
5)化合物9-6的合成5) Synthesis of compound 9-6
室温下,依次将化合物9-5(0.80g,2.55mmol,79%纯度)和亚硝酸叔丁酯(0.79g,7.66mmol)加入到四氢呋喃(10mL)中,室温反应3小时。反应液减压浓缩,剩余物经硅胶柱层析(石油醚/乙酸乙酯=20:1)纯化,得到化合物9-6。MS-ESI:m/z232.0[M+H]+At room temperature, compound 9-5 (0.80 g, 2.55 mmol, 79% purity) and tert-butyl nitrite (0.79 g, 7.66 mmol) were sequentially added into tetrahydrofuran (10 mL), and reacted at room temperature for 3 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20:1) to obtain compound 9-6. MS-ESI: m/z 232.0 [M+H] + .
6)化合物9-7的合成6) Synthesis of compound 9-7
室温下,将化合物9-6(300mg,1.29mmol)和中间体A(442mg,2.32mmol)加入二甲基亚砜(10mL)中,随后加入氟化铯(294mg,1.94mmol),反应液在100℃搅拌16小时。反应液冷却到室温,加水(30mL)稀释,乙酸乙酯(40mL×2)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩。粗品经硅胶柱层析(石油醚/乙酸乙酯=10:1)分离,得到化合物9-7。MS-ESI:m/z384.9[M+H]+At room temperature, compound 9-6 (300mg, 1.29mmol) and intermediate A (442mg, 2.32mmol) were added to dimethyl sulfoxide (10mL), followed by cesium fluoride (294mg, 1.94mmol), and the reaction solution was Stir at 100°C for 16 hours. The reaction solution was cooled to room temperature, diluted with water (30 mL), extracted with ethyl acetate (40 mL×2), combined organic phases, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was separated by silica gel column chromatography (petroleum ether/ethyl acetate=10:1) to obtain compound 9-7. MS-ESI: m/z 384.9 [M+H] + .
7)化合物9-8的合成7) Synthesis of compound 9-8
室温下,将化合物9-7(150mg,0.33mmol,85%纯度),中间体D(110mg,0.36mmol)和叔丁醇钾(111mg,0.99mmol),2,2-二(二苯基膦基)-1,1-联萘(41.13mg,0.066mmol)溶于叔丁醇(5mL)和乙二醇二甲醚(5mL)中,氮气置换,再加入三(二亚苄基丙酮)二钯(30mg,0.03mmol),反应混合物在100℃搅拌16小时。反应液 冷却到室温,减压浓缩,得到化合物9-8。粗品不经纯化,直接用于下一步反应。MS-ESI:m/z 571.2[M+H]+At room temperature, compound 9-7 (150mg, 0.33mmol, 85% purity), intermediate D (110mg, 0.36mmol) and potassium tert-butoxide (111mg, 0.99mmol), 2,2-bis(diphenylphosphine Base)-1,1-binaphthalene (41.13mg, 0.066mmol) was dissolved in tert-butanol (5mL) and ethylene glycol dimethyl ether (5mL), replaced with nitrogen, and then added tris(dibenzylideneacetone) di Palladium (30mg, 0.03mmol), the reaction mixture was stirred at 100°C for 16 hours. The reaction solution Cool to room temperature and concentrate under reduced pressure to obtain compound 9-8. The crude product was directly used in the next reaction without purification. MS-ESI: m/z 571.2 [M+H] + .
8)化合物9的合成8) Synthesis of Compound 9
室温下,将化合物9-8(189mg,0.33mmol)加入到水(5mL)和甲醇(5mL)中,缓慢加入一水合氢氧化锂(69mg,1.65mmol),反应混合物在室温搅拌16小时。反应液倒入水(30mL)中稀释,用乙酸乙酯(20mL×2)洗涤,有机相丢弃,水相减压浓缩,所得残余物经制备级高效液相色谱(甲酸/乙腈/水体系,色谱柱:Waters Xbridge C18 150*25mm*5μm;流动相:水(0.0225%甲酸),乙腈;梯度配比:乙腈相(0-10min,37-67%);流速:25mL/min;柱温:室温)分离,得到化合物9。MS-ESI:m/z 557.2[M+H]+1H NMR(400MHz,CD3OD)δ8.06(s,1H),7.52(d,J=2.0Hz,1H),7.49(d,J=8.4Hz,1H),7.30(dd,J=8.4,2.0Hz,1H),5.76(d,J=1.2Hz,1H),5.13-5.06(m,1H),4.76(d,J=1.2Hz,1H),3.97(t,J=8.0Hz,1H),3.86(t,J=7.6Hz,1H),3.72-3.64(m,1H),3.59-3.53(m,1H),3.48(t,J=8.4Hz,1H),3.38-3.33(m,1H),3.28-3.20(m,1H),2.81-2.69(m,2H),2.59-2.43(m,2H),2.28(t,J=12.0Hz,1H),2.12-1.99(m,2H),1.98-1.82(m,3H),1.78-1.67(m,1H),1.64(d,J=6.8Hz,3H),1.38(s,3H),1.15-1.06(m,1H)。Compound 9-8 (189 mg, 0.33 mmol) was added to water (5 mL) and methanol (5 mL) at room temperature, lithium hydroxide monohydrate (69 mg, 1.65 mmol) was added slowly, and the reaction mixture was stirred at room temperature for 16 hours. The reaction solution was poured into water (30 mL) for dilution, washed with ethyl acetate (20 mL×2), the organic phase was discarded, the aqueous phase was concentrated under reduced pressure, and the resulting residue was subjected to preparative high performance liquid chromatography (formic acid/acetonitrile/water system, Chromatographic column: Waters Xbridge C18 150*25mm*5μm; mobile phase: water (0.0225% formic acid), acetonitrile; gradient ratio: acetonitrile phase (0-10min, 37-67%); flow rate: 25mL/min; column temperature: room temperature) to obtain compound 9. MS-ESI: m/z 557.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.06(s, 1H), 7.52(d, J=2.0Hz, 1H), 7.49(d, J=8.4Hz, 1H), 7.30(dd, J=8.4 ,2.0Hz,1H),5.76(d,J=1.2Hz,1H),5.13-5.06(m,1H),4.76(d,J=1.2Hz,1H),3.97(t,J=8.0Hz,1H ),3.86(t,J=7.6Hz,1H),3.72-3.64(m,1H),3.59-3.53(m,1H),3.48(t,J=8.4Hz,1H),3.38-3.33(m, 1H),3.28-3.20(m,1H),2.81-2.69(m,2H),2.59-2.43(m,2H),2.28(t,J=12.0Hz,1H),2.12-1.99(m,2H) , 1.98-1.82 (m, 3H), 1.78-1.67 (m, 1H), 1.64 (d, J=6.8Hz, 3H), 1.38 (s, 3H), 1.15-1.06 (m, 1H).
实施例10:(1R,3r)-3-((R)-3-(1-(2-氯-5-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]吡啶-7-基)氮杂环丁烷-3-基)哌啶-1-基)-1-甲基环丁烷-1-甲酸(10)
Example 10: (1R,3r)-3-((R)-3-(1-(2-chloro-5-(((R)-1-(2,4-dichlorophenyl)ethyl) Amino)-[1,2,4]triazolo[1,5-a]pyridin-7-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclobutane Alkane-1-carboxylic acid (10)
1)化合物10-2的合成1) Synthesis of compound 10-2
室温下,将化合物10-1(15.00g,66.11mmol),(2,4-二甲氧苯基)甲胺(16.58g,99.17mmol)和氟化铯(20.00g,132.23mmol)加入二甲基亚砜(100mL)中,反应混合物在100℃搅拌16小时。反应液加水(100mL)稀释,用乙酸乙酯(100mL×3)萃 取,有机相用饱和食盐水洗涤(100mL×3),无水硫酸钠干燥,过滤,滤液减压浓缩。剩余物经硅胶柱层析(石油醚/乙酸乙酯=20/1)分离,得到化合物10-2。MS-ESI:m/z 357.1[M+H]+At room temperature, compound 10-1 (15.00g, 66.11mmol), (2,4-dimethoxyphenyl) methylamine (16.58g, 99.17mmol) and cesium fluoride (20.00g, 132.23mmol) were added into dimethyl sulfoxide (100 mL), the reaction mixture was stirred at 100°C for 16 hours. The reaction solution was diluted with water (100mL), extracted with ethyl acetate (100mL×3) The organic phase was washed with saturated brine (100 mL×3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to obtain compound 10-2. MS-ESI: m/z 357.1 [M+H] + .
2)化合物10-3的合成2) Synthesis of compound 10-3
室温下,将化合物10-2(4.50g,12.58mmol)加入二氯甲烷(20mL)中,缓慢滴加三氟乙酸(20.00mL),反应液在室温搅拌5小时。反应液减压浓缩,用甲基叔丁基醚和石油醚(1/1,100mL)打浆,过滤,滤液减压浓缩,得到化合物10-3。MS-ESI:m/z 206.8[M+H]+At room temperature, compound 10-2 (4.50 g, 12.58 mmol) was added into dichloromethane (20 mL), trifluoroacetic acid (20.00 mL) was slowly added dropwise, and the reaction solution was stirred at room temperature for 5 hours. The reaction solution was concentrated under reduced pressure, slurried with methyl tert-butyl ether and petroleum ether (1/1, 100 mL), filtered, and the filtrate was concentrated under reduced pressure to obtain compound 10-3. MS-ESI: m/z 206.8 [M+H] + .
3)化合物10-4的合成3) Synthesis of Compound 10-4
室温下,依次将化合物10-3(3.42g,10.64mmol)和乙氧羰基异硫氰酸酯(2.79g,21.28mmol)加入到二氧六环(40mL)中,室温搅拌16小时。反应液减压浓缩,粗品用甲基叔丁基醚和石油醚(1/1,50mL)打浆,过滤,固体干燥,得到化合物10-4。MS-ESI:m/z 337.9[M+H]+At room temperature, compound 10-3 (3.42 g, 10.64 mmol) and ethoxycarbonyl isothiocyanate (2.79 g, 21.28 mmol) were sequentially added to dioxane (40 mL), and stirred at room temperature for 16 hours. The reaction solution was concentrated under reduced pressure, the crude product was slurried with methyl tert-butyl ether and petroleum ether (1/1, 50 mL), filtered, and the solid was dried to obtain compound 10-4. MS-ESI: m/z 337.9 [M+H] + .
4)化合物10-5的合成4) Synthesis of Compound 10-5
室温下,依次将化合物10-4(3.50g,10.34mmol),N,N-二异丙基乙胺(5.10mL,31.01mmol)和盐酸羟胺(2.15g,31.01mmol)加入到甲醇(30mL)和乙醇(30mL)中,反应混合物在60℃反应16小时。反应液减压浓缩,所得粗品用水(50mL)打浆,过滤,固体干燥,得到化合物10-5。MS-ESI:m/z 246.9[M+H]+At room temperature, compound 10-4 (3.50g, 10.34mmol), N,N-diisopropylethylamine (5.10mL, 31.01mmol) and hydroxylamine hydrochloride (2.15g, 31.01mmol) were added to methanol (30mL) in sequence and ethanol (30 mL), the reaction mixture was reacted at 60° C. for 16 hours. The reaction solution was concentrated under reduced pressure, and the obtained crude product was slurried with water (50 mL), filtered, and the solid was dried to obtain compound 10-5. MS-ESI: m/z 246.9 [M+H] + .
5)化合物10-6的合成5) Synthesis of Compound 10-6
在0℃和氮气保护下,将亚硝酸叔丁酯(362mg,3.52mmol)缓慢滴加至化合物10-5(500mg,1.76mmol,87%纯度)和氯化铜(283mg,2.11mmol)的乙腈(5mL)溶液中,反应混合物在60℃搅拌1小时。反应完毕后,向反应混合物中加水(50mL)稀释,乙酸乙酯(50mL×3)萃取。合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=3/1)分离,得到化合物10-6。MS-ESI:m/z 265.9[M+H]+Under nitrogen protection at 0°C, tert-butyl nitrite (362mg, 3.52mmol) was slowly added dropwise to compound 10-5 (500mg, 1.76mmol, 87% purity) and copper chloride (283mg, 2.11mmol) in acetonitrile (5 mL) solution, the reaction mixture was stirred at 60°C for 1 hour. After the reaction was completed, water (50 mL) was added to the reaction mixture to dilute, and ethyl acetate (50 mL×3) was extracted. The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to obtain compound 10-6. MS-ESI: m/z 265.9 [M+H] + .
6)化合物10-7的合成6) Synthesis of compound 10-7
室温下,将化合物10-6(340mg,1.27mmol)和中间体A(363mg,1.91mmol)加入二甲基亚砜(6mL)中,随后加入氟化铯(387mg,2.55mmol),反应液在100℃搅拌1小时。反应完毕后,待反应液冷却至室温,加水(30mL)稀释,乙酸乙酯(30mL×3)萃取。合并有机相,用饱和食盐水(30mL×3)洗涤,经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=5/1)分离,得到化合物10-7。At room temperature, compound 10-6 (340 mg, 1.27 mmol) and intermediate A (363 mg, 1.91 mmol) were added to dimethyl sulfoxide (6 mL), followed by cesium fluoride (387 mg, 2.55 mmol), and the reaction solution was Stir at 100°C for 1 hour. After the reaction was completed, the reaction solution was cooled to room temperature, diluted with water (30 mL), and extracted with ethyl acetate (30 mL×3). The organic phases were combined, washed with saturated brine (30mL×3), dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) , to obtain compound 10-7.
7)化合物10-8的合成7) Synthesis of compound 10-8
室温及氮气保护下,将化合物10-7(150mg,0.30mmol,85%纯度),中间体D(138mg,0.45mmol)和碳酸铯(296mg,0.91mmol)混于叔戊醇(3mL)中,氮气置换,加入甲烷磺酸(4,5-双二苯基膦-9,9-二甲基氧杂蒽)(2'-甲胺基-1,1'-联苯-2-基)钯 (II)(49mg,0.03mmol),反应混合物在90℃搅拌16小时。反应完毕后,待反应液冷却至室温,加水(30mL)稀释,乙酸乙酯(30mL×3)萃取。合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=0/1)分离,得到化合物10-8。粗品未经纯化,直接用于下一步反应。MS-ESI:m/z 605.2[M+H]+Compound 10-7 (150mg, 0.30mmol, 85% purity), intermediate D (138mg, 0.45mmol) and cesium carbonate (296mg, 0.91mmol) were mixed in tert-amyl alcohol (3mL) at room temperature under the protection of nitrogen. Nitrogen replacement, adding methanesulfonic acid (4,5-bisdiphenylphosphine-9,9-dimethylxanthene) (2'-methylamino-1,1'-biphenyl-2-yl) palladium (II) (49mg, 0.03mmol), the reaction mixture was stirred at 90°C for 16 hours. After the reaction was completed, the reaction solution was cooled to room temperature, diluted with water (30 mL), and extracted with ethyl acetate (30 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=0/1) to obtain compound 10-8. The crude product was directly used in the next reaction without further purification. MS-ESI: m/z 605.2 [M+H] + .
8)化合物10的合成8) Synthesis of Compound 10
室温下,将化合物10-8(330mg,0.29mmol,53%纯度)和一水合氢氧化锂(36mg,0.87mmol)溶于四氢呋喃(3mL)和水(3mL)中,反应混合物在室温搅拌3小时。反应完毕后,反应液减压浓缩,所得残余物经制备级高效液相色谱(甲酸/乙腈/水体系,色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:水(0.5%甲酸),乙腈;梯度配比:乙腈相(0-10min,30-60%);流速:25mL/min;柱温:室温)分离,得到化合物10。MS-ESI:m/z 591.3[M+H]+1H NMR(400MHz,CD3OD)δ7.51(d,J=2.0Hz,1H),7.49(d,J=8.4Hz,1H),7.31(dd,J=8.4,2.0Hz,1H),5.67(d,J=2.0Hz,1H),5.10-5.03(m,1H),4.94(d,J=2.0Hz,1H),3.98(t,J=7.6Hz,1H),3.87(t,J=8.4Hz,1H),3.72-3.64(m,1H),3.60-3.53(m,1H),3.52-3.41(m,1H),3.37-3.32(m,1H),3.25-3.18(m,1H),2.78-2.68(m,2H),2.59-2.44(m,2H),2.32-2.21(m,1H),2.07-1.82(m,5H),1.77-1.65(m,1H),1.62(d,J=6.8Hz,3H),1.38(s,3H),1.17-1.02(m,1H)。At room temperature, compound 10-8 (330mg, 0.29mmol, 53% purity) and lithium hydroxide monohydrate (36mg, 0.87mmol) were dissolved in tetrahydrofuran (3mL) and water (3mL), and the reaction mixture was stirred at room temperature for 3 hours . After completion of the reaction, the reaction solution was concentrated under reduced pressure, and the resulting residue was subjected to preparative high-performance liquid chromatography (formic acid/acetonitrile/water system, chromatographic column: Phenomenex luna C18 150*25mm*10 μm; mobile phase: water (0.5% formic acid), Acetonitrile; gradient ratio: acetonitrile phase (0-10min, 30-60%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 10. MS-ESI: m/z 591.3 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.51(d, J=2.0Hz, 1H), 7.49(d, J=8.4Hz, 1H), 7.31(dd, J=8.4, 2.0Hz, 1H), 5.67(d,J=2.0Hz,1H),5.10-5.03(m,1H),4.94(d,J=2.0Hz,1H),3.98(t,J=7.6Hz,1H),3.87(t,J =8.4Hz,1H),3.72-3.64(m,1H),3.60-3.53(m,1H),3.52-3.41(m,1H),3.37-3.32(m,1H),3.25-3.18(m,1H ),2.78-2.68(m,2H),2.59-2.44(m,2H),2.32-2.21(m,1H),2.07-1.82(m,5H),1.77-1.65(m,1H),1.62(d , J=6.8Hz, 3H), 1.38(s, 3H), 1.17-1.02(m, 1H).
实施例11:(1R,3r)-3-((R)-3-(1-(7-(((R)-1-(2,4-二氯苯基)乙基)氨基)吡唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)-1-甲基环丁烷-1-甲酸(11)
Example 11: (1R,3r)-3-((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)pyrazole And[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclobutane-1-carboxylic acid (11)
1)化合物11-2的合成1) Synthesis of Compound 11-2
室温下,将化合物11-1(600mg,3.19mmol),中间体A(619mg,3.19mmol)和N,N-二异丙基乙胺(825mg,6.38mmol)溶于乙腈(10mL)中,反应混合物在50℃搅拌12小时。反应完毕后,反应混合物减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=3/1)分离,得到化合物11-2。MS-ESI:m/z 341.0[M+H]+1H NMR(400MHz,DMSO-d6)δ8.97(d,J=7.6Hz,1H),8.18(d,J=2.4Hz,1H),8.65(d,J=2.0Hz,1H),8.62(d,J=8.4Hz,1H),7.44(dd,J=8.4,2.0Hz,1H),6.45(d,J=2.4Hz,1H),5.74(s,1H),5.19-5.07(m,1H),1.62(d,J=6.8Hz,3H)。At room temperature, compound 11-1 (600mg, 3.19mmol), intermediate A (619mg, 3.19mmol) and N,N-diisopropylethylamine (825mg, 6.38mmol) were dissolved in acetonitrile (10mL), and the reaction The mixture was stirred at 50°C for 12 hours. After the reaction was completed, the reaction mixture was concentrated under reduced pressure, and the resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to obtain compound 11-2. MS-ESI: m/z 341.0 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ8.97(d, J=7.6Hz, 1H), 8.18(d, J=2.4Hz, 1H), 8.65(d, J=2.0Hz, 1H), 8.62 (d,J=8.4Hz,1H),7.44(dd,J=8.4,2.0Hz,1H),6.45(d,J=2.4Hz,1H),5.74(s,1H),5.19-5.07(m, 1H), 1.62 (d, J=6.8Hz, 3H).
2)化合物11的合成2) Synthesis of Compound 11
室温下,将化合物11-2(100mg,0.29mmol),中间体D(97mg,0.32mmol),叔丁醇钾(98mg,0.87mmol),(±)-2,2-双(二苯膦基)-11-联萘(36mg,0.06mmol)加 入到叔丁醇(3mL)和乙二醇二甲醚(5mL)中,氮气置换,再将三(二亚苄基丙酮)二钯(26mg,0.03mmol)加入到反应液,反应混合物在100℃搅拌12小时。反应液冷却至室温,倒入水(10mL)中稀释,二氯甲烷(20mL×2)洗涤。有机相丢弃,水相经制备级高效液相色谱(甲酸/乙腈/水体系,色谱柱:Phenomenex C18 75*30mm*3μm;流动相:水(10mM甲酸),乙腈;梯度配比:乙腈相(0-7min,8-38%);流速:25mL/min;柱温:室温)分离,得到化合物11。MS-ESI:m/z 557.2[M+H]+1H NMR(400MHz,CD3OD)δ7.83-7.79(m,1H),7.53-7.43(m,2H),7.32(dd,J=8.4,2.0Hz,1H),5.99-5.93(m,1H),5.17-5.08(m,1H),4.71(s,1H),4.07(t,J=8.4Hz,1H),3.96(t,J=7.6Hz,1H),3.76(dd,J=8.4,5.6Hz,1H),3.71-3.63(m,1H),3.60-3.50(m,1H),3.39-3.33(m,1H),3.26-3.20(m,1H),2.81-2.67(m,2H),2.62-2.47(m,2H),2.41-2.28(m,1H),2.10-1.97(m,3H),1.96-1.85(m,2H),1.77-1.67(m,1H),1.66-1.61(m,3H),1.38(s,3H),1.15-1.04(m,1H)。At room temperature, compound 11-2 (100mg, 0.29mmol), intermediate D (97mg, 0.32mmol), potassium tert-butoxide (98mg, 0.87mmol), (±)-2,2-bis(diphenylphosphino )-11-binaphthalene (36mg, 0.06mmol) plus into tert-butanol (3mL) and ethylene glycol dimethyl ether (5mL), nitrogen replacement, then tris(dibenzylideneacetone) dipalladium (26mg, 0.03mmol) was added to the reaction solution, and the reaction mixture was heated at 100 °C and stirred for 12 hours. The reaction solution was cooled to room temperature, poured into water (10 mL) for dilution, and washed with dichloromethane (20 mL×2). The organic phase was discarded, and the aqueous phase was subjected to preparative high-performance liquid chromatography (formic acid/acetonitrile/water system, chromatographic column: Phenomenex C18 75*30mm*3μm; mobile phase: water (10mM formic acid), acetonitrile; gradient ratio: acetonitrile phase ( 0-7min, 8-38%); flow rate: 25mL/min; column temperature: room temperature) to obtain compound 11. MS-ESI: m/z 557.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.83-7.79(m, 1H), 7.53-7.43(m, 2H), 7.32(dd, J=8.4, 2.0Hz, 1H), 5.99-5.93(m, 1H), 5.17-5.08(m, 1H), 4.71(s, 1H), 4.07(t, J=8.4Hz, 1H), 3.96(t, J=7.6Hz, 1H), 3.76(dd, J=8.4 ,5.6Hz,1H),3.71-3.63(m,1H),3.60-3.50(m,1H),3.39-3.33(m,1H),3.26-3.20(m,1H),2.81-2.67(m,2H ),2.62-2.47(m,2H),2.41-2.28(m,1H),2.10-1.97(m,3H),1.96-1.85(m,2H),1.77-1.67(m,1H),1.66-1.61 (m,3H), 1.38(s,3H), 1.15-1.04(m,1H).
实施例12:2-(R)-3-(1-(5-((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-c]嘧啶-7-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(12)
Example 12: 2-(R)-3-(1-(5-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4]triazole And[1,5-c]pyrimidin-7-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (12)
1)化合物12-2的合成1) Synthesis of compound 12-2
室温下,依次将化合物12-1(3.50g,18.23mmol),中间体A(2.99g,18.23mmol)和N,N-二异丙基乙胺(4.50mL,27.34mmol)溶于异丙醇(500mL),100℃搅拌32个小时。反应液减压浓缩,用乙酸乙酯(60mL)和水(40mL)稀释,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=2/1)分离,得到化合物12-2。MS-ESI:m/z 317.1[M+H]+At room temperature, sequentially dissolve compound 12-1 (3.50g, 18.23mmol), intermediate A (2.99g, 18.23mmol) and N,N-diisopropylethylamine (4.50mL, 27.34mmol) in isopropanol (500 mL), stirred at 100°C for 32 hours. The reaction solution was concentrated under reduced pressure, diluted with ethyl acetate (60 mL) and water (40 mL), the organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was subjected to silica gel column chromatography (petroleum ether/ethyl acetate Ester = 2/1) separation to obtain compound 12-2. MS-ESI: m/z 317.1 [M+H] + .
2)化合物12-3的合成2) Synthesis of compound 12-3
室温下,将化合物12-2(2.00g,5.86mmol)溶于二氧六环(40mL)中,随后加入乙氧羰基异硫氰酸酯(1.54g,11.71mmol),反应液在90℃搅拌16小时。反应液减压浓缩,得到化合物12-3。粗品不经纯化,直接用于下一步反应。MS-ESI:m/z448.0[M+H]+At room temperature, compound 12-2 (2.00g, 5.86mmol) was dissolved in dioxane (40mL), then ethoxycarbonyl isothiocyanate (1.54g, 11.71mmol) was added, and the reaction solution was stirred at 90°C 16 hours. The reaction solution was concentrated under reduced pressure to obtain compound 12-3. The crude product was directly used in the next reaction without purification. MS-ESI: m/z 448.0 [M+H] + .
3)化合物12-4的合成 3) Synthesis of compound 12-4
室温下,将化合物12-3(2.63g,粗品)和N,N-二异丙基乙胺(2.90mL,17.57mmol)溶于甲醇(20mL)和乙醇(20mL)的混合溶剂中,随后分批加入盐酸羟胺(814mg,11.71mmol),反应液在45℃反应2个小时。反应结束后,反应液减压浓缩,用水(20mL)稀释,乙酸乙酯(60mL×2)萃取,合并有机相,无水硫酸钠干燥,过滤,减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=2/1)分离,得到化合物12-4。MS-ESI:m/z 357.0[M+H]+At room temperature, compound 12-3 (2.63g, crude product) and N,N-diisopropylethylamine (2.90mL, 17.57mmol) were dissolved in a mixed solvent of methanol (20mL) and ethanol (20mL), and then separated Hydroxylamine hydrochloride (814mg, 11.71mmol) was added in batches, and the reaction solution was reacted at 45°C for 2 hours. After the reaction, the reaction liquid was concentrated under reduced pressure, diluted with water (20 mL), extracted with ethyl acetate (60 mL×2), combined the organic phases, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, and the resulting residue was subjected to silica gel column chromatography (petroleum ether/ethyl acetate=2/1) separation to obtain compound 12-4. MS-ESI: m/z 357.0 [M+H] + .
4)化合物12-5的合成4) Synthesis of compound 12-5
0℃下,将化合物12-4(1.00g,2.71mmol)溶于四氢呋喃(20mL),缓慢滴加亚硝酸叔丁酯(0.84g,8.14mmol),反应液在0℃搅拌2小时。反应液加水(50mL)稀释,乙酸乙酯(50mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=2/1)分离,得到化合物12-5。MS-ESI:m/z 341.9[M+H]+At 0°C, compound 12-4 (1.00g, 2.71mmol) was dissolved in tetrahydrofuran (20mL), tert-butyl nitrite (0.84g, 8.14mmol) was slowly added dropwise, and the reaction solution was stirred at 0°C for 2 hours. The reaction solution was diluted with water (50mL), extracted with ethyl acetate (50mL×3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was subjected to silica gel column chromatography (petroleum ether/ethyl acetate =2/1) separation to obtain compound 12-5. MS-ESI: m/z 341.9 [M+H] + .
5)化合物12的合成5) Synthesis of Compound 12
室温下,将化合物12-5(150mg,0.44mmol),中间体C(145mg,0.59mmol)和氟化铯(133mg,0.88mmol)混于二甲亚砜(3mL),反应混合物在60℃搅拌16小时。反应完毕后,过滤,滤液经制备级高效液相色谱(碳酸氢铵/乙腈/水体系,色谱柱:Unisil 3-100C18Ultra 150*50mm*3μm;流动相:水(0.5%甲酸),乙腈;梯度配比:乙腈相(0-7min,10-40%);流速:25mL/min;柱温:室温)分离,再经硅胶制备薄层色谱(纯甲醇)分离,得到化合物12。MS-ESI:m/z 490.1[M+H]+1H NMR(400MHz,CD3OD)δ8.03(s,1H),7.51-7.46(m,2H),7.29(dd,J=8.4,2.0Hz,1H),5.62-5.55(m,1H),5.54(s,1H),4.00(t,J=8.4Hz,1H),3.93(t,J=8.4Hz,1H),3.74-3.66(m,3H),3.52-3.47(m,1H),2.98-2.88(m,2H),2.58-2.53(m,2H),2.49-2.41(m,1H),2.10-2.01(m,1H),1.78-1.71(m,4H),1.61(d,J=7.2Hz,4H),0.95-0.85(m,1H)。At room temperature, compound 12-5 (150mg, 0.44mmol), intermediate C (145mg, 0.59mmol) and cesium fluoride (133mg, 0.88mmol) were mixed in dimethylsulfoxide (3mL), and the reaction mixture was stirred at 60°C 16 hours. After the reaction is completed, filter, and the filtrate is subjected to preparative high-performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Unisil 3-100C18Ultra 150*50mm*3μm; mobile phase: water (0.5% formic acid), acetonitrile; gradient Proportion: acetonitrile phase (0-7min, 10-40%); flow rate: 25mL/min; column temperature: room temperature) separation, and then separated by silica gel preparative thin-layer chromatography (pure methanol) to obtain compound 12. MS-ESI: m/z 490.1 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.03(s, 1H), 7.51-7.46(m, 2H), 7.29(dd, J=8.4, 2.0Hz, 1H), 5.62-5.55(m, 1H) ,5.54(s,1H),4.00(t,J=8.4Hz,1H),3.93(t,J=8.4Hz,1H),3.74-3.66(m,3H),3.52-3.47(m,1H), 2.98-2.88(m,2H),2.58-2.53(m,2H),2.49-2.41(m,1H),2.10-2.01(m,1H),1.78-1.71(m,4H),1.61(d,J =7.2Hz, 4H), 0.95-0.85(m, 1H).
实施例13:(1R,3r)-3-((R)-3-(1-(2-氯-5-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-c]嘧啶-7-基)氮杂环丁烷-3-基)哌啶-1-基)-1-甲基环丁烷-1-甲酸(13)
Example 13: (1R,3r)-3-((R)-3-(1-(2-chloro-5-(((R)-1-(2,4-dichlorophenyl)ethyl) Amino)-[1,2,4]triazolo[1,5-c]pyrimidin-7-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclobutane Alkane-1-carboxylic acid (13)
1)化合物13-1的合成1) Synthesis of compound 13-1
0℃下,将化合物12-4(400mg,1.12mmol)和氯化铜(180mg,1.34mmol)溶于乙腈(2mL)中,缓慢滴加亚硝酸叔丁酯(231mg,2.24mmol),反应混合物在0℃搅拌2小时。反应完毕后,反应混合物加水(50mL)稀释,用乙酸乙酯(50mL×3)萃取。合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=1/1)分离,得到化合物13-1。MS-ESI:m/z 375.9[M+H]+At 0°C, compound 12-4 (400mg, 1.12mmol) and copper chloride (180mg, 1.34mmol) were dissolved in acetonitrile (2mL), and tert-butyl nitrite (231mg, 2.24mmol) was slowly added dropwise, and the reaction mixture was Stir at 0°C for 2 hours. After the reaction was complete, the reaction mixture was diluted with water (50 mL), and extracted with ethyl acetate (50 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to obtain compound 13-1. MS-ESI: m/z 375.9 [M+H] + .
2)化合物13-2的合成2) Synthesis of compound 13-2
室温下,依次将化合物13-1(200mg,0.53mmol),中间体D(241mg,0.80mmol)和氟化铯(161mg,1.06mmol)混于二甲基亚砜(4mL)中,反应混合物在80℃搅拌16小时。反应完毕后,反应混合物加水(30mL)稀释,乙酸乙酯(50mL×3)萃取。合并有机相,用水(30mL×3)洗涤,经无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶制备薄层色谱(乙酸乙酯/甲醇=5/1)分离,得到化合物13-2。MS-ESI:m/z 606.0[M+H]+At room temperature, sequentially mix compound 13-1 (200mg, 0.53mmol), intermediate D (241mg, 0.80mmol) and cesium fluoride (161mg, 1.06mmol) in dimethyl sulfoxide (4mL), and the reaction mixture was Stir at 80°C for 16 hours. After the reaction was completed, the reaction mixture was diluted with water (30 mL), and extracted with ethyl acetate (50 mL×3). The organic phases were combined, washed with water (30 mL×3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The obtained residue was separated by preparative thin-layer chromatography on silica gel (ethyl acetate/methanol=5/1) to obtain compound 13-2. MS-ESI: m/z 606.0 [M+H] + .
3)化合物13的合成3) Synthesis of compound 13
室温下,将化合物13-2(170mg,0.28mmol)和一水合氢氧化锂(35mg,0.84mmol)溶于甲醇(2mL)和水(2mL)中,反应混合物在室温搅拌2小时。反应完毕后,反应液减压浓缩,所得残余物经制备级高效液相色谱(甲酸/乙腈/水体系,色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:水(0.5%甲酸),乙腈;梯度配比:乙腈相(0-10min,19-49%);流速:25mL/min;柱温:室温)分离,得到化合物13。MS-ESI:m/z 592.1[M+H]+1H NMR(400MHz,CD3OD)δ8.42(s,1H),7.51-7.46(m,2H),7.30(dd,J=8.4,2.0Hz,1H),5.61-5.54(m,1H),5.49(s,1H),4.06(t,J=8.4Hz,1H),3.96(t,J=8.4Hz,1H),3.78-3.69(m,1H),3.65-3.54(m,2H),3.43-3.36(m,1H),3.28-3.21(m,1H),2.82-2.75(m,2H),2.67-2.57(m,1H),2.56-2.48(m,1H),2.42-2.31(m,1H),2.13-1.98(m,3H),1.95-1.83(m,2H),1.80-1.68(m,1H),1.59(d,J=7.2Hz,3H),1.41(s,3H),1.19-1.07(m,1H)。Compound 13-2 (170 mg, 0.28 mmol) and lithium hydroxide monohydrate (35 mg, 0.84 mmol) were dissolved in methanol (2 mL) and water (2 mL) at room temperature, and the reaction mixture was stirred at room temperature for 2 hours. After completion of the reaction, the reaction solution was concentrated under reduced pressure, and the resulting residue was subjected to preparative high-performance liquid chromatography (formic acid/acetonitrile/water system, chromatographic column: Phenomenex luna C18 150*25mm*10 μm; mobile phase: water (0.5% formic acid), Acetonitrile; gradient ratio: acetonitrile phase (0-10min, 19-49%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 13. MS-ESI: m/z 592.1 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.42(s, 1H), 7.51-7.46(m, 2H), 7.30(dd, J=8.4, 2.0Hz, 1H), 5.61-5.54(m, 1H) ,5.49(s,1H),4.06(t,J=8.4Hz,1H),3.96(t,J=8.4Hz,1H),3.78-3.69(m,1H),3.65-3.54(m,2H), 3.43-3.36(m,1H),3.28-3.21(m,1H),2.82-2.75(m,2H),2.67-2.57(m,1H),2.56-2.48(m,1H),2.42-2.31(m ,1H),2.13-1.98(m,3H),1.95-1.83(m,2H),1.80-1.68(m,1H),1.59(d,J=7.2Hz,3H),1.41(s,3H), 1.19-1.07(m,1H).
实施例14:2-(R)-3-(1-(5-((R)-1-(2,4-二氯苯基)乙基)氨基)-8-氟-[1,2,4]三唑并[1,5-c]嘧啶-7-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(14)
Example 14: 2-(R)-3-(1-(5-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-8-fluoro-[1,2, 4] Triazolo[1,5-c]pyrimidin-7-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (14)
1)化合物14-1的合成1) Synthesis of compound 14-1
室温下,将化合物12-4(500mg,1.40mmol)溶于甲醇(1.5mL)和乙腈(1.5mL)中,然后加入1-氟-4-甲基-1,4-二氮杂双环[2.2.2]辛烷四氟硼酸盐(670mg,2.10mmol),反应液在50℃搅拌1小时。反应完毕后,将反应液冷却至室温,减压浓 缩。所得残余物经硅胶制备薄层色谱(石油醚/乙酸乙酯=1/1)分离,得到化合物14-1。MS-ESI:m/z 375.1[M+H]+At room temperature, compound 12-4 (500mg, 1.40mmol) was dissolved in methanol (1.5mL) and acetonitrile (1.5mL), and then 1-fluoro-4-methyl-1,4-diazabicyclo[2.2 .2] Octane tetrafluoroborate (670mg, 2.10mmol), the reaction solution was stirred at 50°C for 1 hour. After the reaction was completed, the reaction solution was cooled to room temperature, and concentrated under reduced pressure. shrink. The resulting residue was separated by preparative thin-layer chromatography on silica gel (petroleum ether/ethyl acetate=1/1) to obtain compound 14-1. MS-ESI: m/z 375.1 [M+H] + .
2)化合物14-2的合成2) Synthesis of compound 14-2
室温下,将化合物14-1(80mg,0.16mmol,77%纯度)溶于四氢呋喃(1mL)中,冰浴下加入亚硝酸叔丁酯(51mg,0.49mmol),反应液在0℃搅拌20分钟。反应完毕后,反应液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=2/1)分离,得到化合物14-2。MS-ESI:m/z 359.9[M+H]+At room temperature, compound 14-1 (80mg, 0.16mmol, 77% purity) was dissolved in tetrahydrofuran (1mL), tert-butyl nitrite (51mg, 0.49mmol) was added under ice cooling, and the reaction solution was stirred at 0°C for 20 minutes . After the reaction was completed, the reaction solution was concentrated under reduced pressure, and the resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to obtain compound 14-2. MS-ESI: m/z 359.9 [M+H] + .
3)化合物14-3的合成3) Synthesis of compound 14-3
室温下,将化合物14-2(59mg,0.16mmol)和中间体E(86mg,0.33mmol)溶于二甲亚砜(2mL)中,加入氟化铯(75mg,0.49mmol),反应混合物在80℃搅拌16小时。将反应液冷却至室温,倒入水(50mL)中稀释,用乙酸乙酯(30mL×3)萃取。合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=1/2)分离,得到化合物14-3。MS-ESI:m/z 548.2[M+H]+At room temperature, compound 14-2 (59mg, 0.16mmol) and intermediate E (86mg, 0.33mmol) were dissolved in dimethylsulfoxide (2mL), cesium fluoride (75mg, 0.49mmol) was added, and the reaction mixture was heated at 80 °C and stirred for 16 hours. The reaction solution was cooled to room temperature, poured into water (50 mL) for dilution, and extracted with ethyl acetate (30 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/2) to obtain compound 14-3. MS-ESI: m/z 548.2 [M+H] + .
4)化合物14-4的合成4) Synthesis of compound 14-4
室温下,将化合物14-3(30mg,0.06mmol)和四三苯基膦钯(6mg,0.01mmol)混于二氯甲烷(2mL)中,缓慢加入苯硅烷(59mg,0.55mmol),反应混合物在25℃搅拌2小时。反应完毕后,所得残余物经硅胶柱层析(三乙胺/甲醇=1/20)分离,得到化合物14-4。MS-ESI:m/z 464.2[M+H]+At room temperature, compound 14-3 (30mg, 0.06mmol) and tetrakistriphenylphosphine palladium (6mg, 0.01mmol) were mixed in dichloromethane (2mL), and phenylsilane (59mg, 0.55mmol) was added slowly, and the reaction mixture Stir at 25°C for 2 hours. After the reaction was completed, the obtained residue was separated by silica gel column chromatography (triethylamine/methanol=1/20) to obtain compound 14-4. MS-ESI: m/z 464.2 [M+H] + .
5)化合物14-5的合成5) Synthesis of compound 14-5
室温下,将化合物14-4(10mg,0.02mmol,89%纯度),醋酸(12mg,0.19mmol)和叔丁基二甲基硅氧烷基乙醛(5mg,0.03mmol)溶于甲醇(1mL)中,搅拌30分钟后,再加入氰基硼氢化钠(2mg,0.04mmol),反应混合物在25℃反应2小时。反应完毕后,反应液减压浓缩,得到化合物14-5,粗品不经纯化,直接用于下一步反应。MS-ESI:m/z 622.3[M+H]+At room temperature, compound 14-4 (10 mg, 0.02 mmol, 89% purity), acetic acid (12 mg, 0.19 mmol) and tert-butyldimethylsiloxane acetaldehyde (5 mg, 0.03 mmol) were dissolved in methanol (1 mL ), after stirring for 30 minutes, sodium cyanoborohydride (2mg, 0.04mmol) was added, and the reaction mixture was reacted at 25°C for 2 hours. After the reaction was completed, the reaction solution was concentrated under reduced pressure to obtain compound 14-5. The crude product was directly used in the next reaction without further purification. MS-ESI: m/z 622.3 [M+H] + .
6)化合物14的合成6) Synthesis of compound 14
室温下,将化合物14-5(13mg,0.02mmol,84%纯度)溶于甲醇(1mL),加入三氟乙酸(1.00mL),反应液30℃反应2小时。反应完毕后,反应液减压浓缩。所得残余物溶于甲醇(1mL)中,缓慢滴加氨水至pH~8后,减压浓缩。所得残余物经硅胶制备薄层色谱(纯甲醇)分离,得到化合物14。MS-ESI:m/z 508.3[M+H]+1H NMR(400MHz,CD3OD)δ8.08(s,1H),7.49-7.43(m,2H),7.28(dd,J=8.4,2.0Hz,1H),5.56-5.47(m,1H),4.15-4.05(m,2H),3.88-3.82(m,1H),3.72(t,J=6.0Hz,2H),3.65-3.58(m,1H),3.07-2.93(m,2H),2.64(t,J=5.2Hz,2H),2.53-2.42(m,1H),2.23-2.12(m,1H),1.84-1.70(m,4H),1.68-1.61(m,1H),1.58(d,J=7.2Hz,3H),0.97-0.89(m,1H)。Compound 14-5 (13 mg, 0.02 mmol, 84% purity) was dissolved in methanol (1 mL) at room temperature, trifluoroacetic acid (1.00 mL) was added, and the reaction solution was reacted at 30° C. for 2 hours. After the reaction was completed, the reaction solution was concentrated under reduced pressure. The resulting residue was dissolved in methanol (1 mL), and aqueous ammonia was slowly added dropwise to pH ~ 8, then concentrated under reduced pressure. The resulting residue was separated by silica gel preparative thin-layer chromatography (pure methanol) to obtain Compound 14. MS-ESI: m/z 508.3 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.08(s, 1H), 7.49-7.43(m, 2H), 7.28(dd, J=8.4, 2.0Hz, 1H), 5.56-5.47(m, 1H) ,4.15-4.05(m,2H),3.88-3.82(m,1H),3.72(t,J=6.0Hz,2H),3.65-3.58(m,1H),3.07-2.93(m,2H),2.64 (t,J=5.2Hz,2H),2.53-2.42(m,1H),2.23-2.12(m,1H),1.84-1.70(m,4H),1.68-1.61(m,1H),1.58(d , J=7.2Hz, 3H), 0.97-0.89(m, 1H).
实施例15:2-(R)-3-(1-(7-((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a][1,3,5]三嗪-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(15)
Example 15: 2-(R)-3-(1-(7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4]triazole And[1,5-a][1,3,5]triazin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (15)
1)化合物15-2的合成1) Synthesis of compound 15-2
室温下,将化合物15-1(5.00g,59.47mmol)溶于N,N-二甲基甲酰胺(50mL)中,缓慢滴加乙氧羰基异硫氰酸酯(7.80g,59.47mmol),反应液在25℃反应15小时。反应完毕后,将反应液倒入水(500mL)中,过滤,滤饼用水(100mL)洗涤,真空干燥,得到化合物15-2。1H NMR(400MHz,DMSO-d6)δ14.00(s,1H),12.15-11.68(m,1H),11.45(s,1H),8.54(s,1H),4.21(q,J=7.2Hz,2H),1.25(t,J=7.2Hz,3H)。At room temperature, compound 15-1 (5.00g, 59.47mmol) was dissolved in N,N-dimethylformamide (50mL), and ethoxycarbonyl isothiocyanate (7.80g, 59.47mmol) was slowly added dropwise, The reaction solution was reacted at 25° C. for 15 hours. After the reaction was completed, the reaction solution was poured into water (500 mL), filtered, the filter cake was washed with water (100 mL), and dried in vacuo to obtain compound 15-2. 1 H NMR (400MHz, DMSO-d 6 )δ14.00(s,1H),12.15-11.68(m,1H),11.45(s,1H),8.54(s,1H),4.21(q,J=7.2 Hz, 2H), 1.25 (t, J = 7.2Hz, 3H).
2)化合物15-3的合成2) Synthesis of compound 15-3
室温下,将化合物15-2(5.00g,23.23mmol)溶于乙醇(80mL)中,加入氢氧化钠水溶液(23.23mL,2mol/L,46.46mmol),反应混合物在78℃搅拌30分钟。反应完毕后,将反应液冷却至室温,过滤,滤饼加水(10mL)溶解,用1mol/L的盐酸水溶液调节pH至6~7,混合物经反相柱(乙腈/水体系,色谱柱:C18spherical20-35μm40g,流动相:水,乙腈;梯度配比:乙腈相(0-8min,0-5%);流速:35mL/min;柱温:室温)分离,得到化合物15-3。1H NMR(400MHz,DMSO-d6)δ11.53(brs,1H),7.92(s,1H)。Compound 15-2 (5.00g, 23.23mmol) was dissolved in ethanol (80mL) at room temperature, aqueous sodium hydroxide solution (23.23mL, 2mol/L, 46.46mmol) was added, and the reaction mixture was stirred at 78°C for 30 minutes. After completion of the reaction, the reaction solution was cooled to room temperature, filtered, and the filter cake was dissolved by adding water (10mL), and the pH was adjusted to 6-7 with 1mol/L hydrochloric acid aqueous solution, and the mixture was passed through a reverse-phase column (acetonitrile/water system, chromatographic column: C18spherical20 -35μm 40g, mobile phase: water, acetonitrile; gradient ratio: acetonitrile phase (0-8min, 0-5%); flow rate: 35mL/min; column temperature: room temperature) separation to obtain compound 15-3. 1 H NMR (400 MHz, DMSO-d 6 ) δ 11.53 (brs, 1H), 7.92 (s, 1H).
3)化合物15-4的合成3) Synthesis of compound 15-4
室温下,将化合物15-3(3.40g,20.10mmol)和碘甲烷(2.85g,20.10mmol)溶于N,N-二甲基甲酰胺(34mL)中,反应混合物室温搅拌6小时。反应完毕后,反应液减压浓缩,得到化合物15-4。粗品不经纯化,直接用于下一步反应。1H NMR(400MHz,DMSO-d6)δ8.32(s,1H),2.57(s,3H)。Compound 15-3 (3.40 g, 20.10 mmol) and iodomethane (2.85 g, 20.10 mmol) were dissolved in N,N-dimethylformamide (34 mL) at room temperature, and the reaction mixture was stirred at room temperature for 6 hours. After completion of the reaction, the reaction solution was concentrated under reduced pressure to obtain compound 15-4. The crude product was directly used in the next reaction without purification. 1 H NMR (400 MHz, DMSO-d 6 ) δ 8.32 (s, 1H), 2.57 (s, 3H).
4)化合物15-5的合成4) Synthesis of compound 15-5
室温下,将化合物15-4(1.00g,5.46mmol)和N,N-二乙基苯胺(2.44g,16.38mmol)溶于三氯氧磷(10.10mL,109.17mmol)中,反应混合物在100℃搅拌2小时。反应完毕后,反应混合物冷却至室温,减压浓缩,得到化合物15-5。粗品不经纯化,直接用于下一步反应。MS-ESI:m/z 201.9[M+H]+At room temperature, compound 15-4 (1.00g, 5.46mmol) and N,N-diethylaniline (2.44g, 16.38mmol) were dissolved in phosphorus oxychloride (10.10mL, 109.17mmol), and the reaction mixture was heated at 100 °C and stirred for 2 hours. After completion of the reaction, the reaction mixture was cooled to room temperature and concentrated under reduced pressure to obtain compound 15-5. The crude product was directly used in the next reaction without purification. MS-ESI: m/z 201.9 [M+H] + .
5)化合物15-6的合成5) Synthesis of compound 15-6
室温下,将化合物15-5(1.10g,5.46mmol),中间体A(1.56g,8.19mmol)和吡啶(1.32mL,16.38mmol)溶于乙腈(11mL)中,反应混合物在50℃搅拌0.5小时。 反应完毕后,反应混合物冷却至室温,减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=2/1)分离,得到化合物15-6。MS-ESI:m/z 355.0[M+H]+At room temperature, compound 15-5 (1.10g, 5.46mmol), intermediate A (1.56g, 8.19mmol) and pyridine (1.32mL, 16.38mmol) were dissolved in acetonitrile (11mL), and the reaction mixture was stirred at 50°C for 0.5 Hour. After the reaction was completed, the reaction mixture was cooled to room temperature and concentrated under reduced pressure. The resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to obtain compound 15-6. MS-ESI: m/z 355.0 [M+H] + .
6)化合物15-7的合成6) Synthesis of Compound 15-7
室温下,将化合物15-6(450mg,1.27mmol)和间氯过氧苯甲酸(819mg,3.80mmol,80%纯度)溶于二氯甲烷(10mL)中,室温搅拌2小时。反应完毕后,反应液经硅胶柱层析(石油醚/乙酸乙酯=1/1)分离,得到化合物15-7。MS-ESI:m/z 387.0[M+H]+Compound 15-6 (450 mg, 1.27 mmol) and m-chloroperoxybenzoic acid (819 mg, 3.80 mmol, 80% purity) were dissolved in dichloromethane (10 mL) at room temperature, and stirred at room temperature for 2 hours. After the reaction was completed, the reaction solution was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to obtain compound 15-7. MS-ESI: m/z 387.0 [M+H] + .
7)化合物15的合成7) Synthesis of Compound 15
室温下,将化合物15-7(100mg,0.14mmol,54%纯度),中间体C(46mg,0.21mmol)和N,N-二异丙基乙胺(54mg,0.42mmol)溶于乙腈(2mL),反应液在60℃搅拌4小时。反应液冷却至室温,减压浓缩,所得残余物经硅胶制备薄层色谱(纯甲醇)分离,再经制备级高效液相色谱(甲酸/乙腈/水体系,色谱柱:Unisil 3-100C18Ultra 150*50mm*3μm;流动相:水(0.5%甲酸),乙腈;梯度配比:乙腈相(0-7min,10-40%);流速:25mL/min;柱温:室温)分离,得到化合物15。MS-ESI:m/z 491.2[M+H]+1H NMR(400MHz,CD3OD)δ8.54(s,1H),8.05(s,1H),7.54-7.47(m,2H),7.34(dd,J=8.4,2.0Hz,1H),5.71-5.61(m,1H),4.31-4.09(m,2H),4.05-3.58(m,5H),3.48-3.41(m,1H),3.14-3.04(m,2H),2.72(t,J=11.6Hz,1H),2.55-2.40(m,2H),2.00-1.73(m,4H),1.62(d,J=6.8Hz,3H),1.18-1.05(m,1H)。Compound 15-7 (100 mg, 0.14 mmol, 54% purity), Intermediate C (46 mg, 0.21 mmol) and N,N-diisopropylethylamine (54 mg, 0.42 mmol) were dissolved in acetonitrile (2 mL ), and the reaction solution was stirred at 60° C. for 4 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the resulting residue was separated by preparative thin-layer chromatography on silica gel (pure methanol), and then separated by preparative high-performance liquid chromatography (formic acid/acetonitrile/water system, chromatographic column: Unisil 3-100C18Ultra 150* 50mm*3μm; mobile phase: water (0.5% formic acid), acetonitrile; gradient ratio: acetonitrile phase (0-7min, 10-40%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 15. MS-ESI: m/z 491.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.54(s, 1H), 8.05(s, 1H), 7.54-7.47(m, 2H), 7.34(dd, J=8.4, 2.0Hz, 1H), 5.71 -5.61(m,1H),4.31-4.09(m,2H),4.05-3.58(m,5H),3.48-3.41(m,1H),3.14-3.04(m,2H),2.72(t,J= 11.6Hz, 1H), 2.55-2.40(m, 2H), 2.00-1.73(m, 4H), 1.62(d, J=6.8Hz, 3H), 1.18-1.05(m, 1H).
实施例16:2-(R)-3-(1-(7-((R)-1-(2,4-二氯苯基)乙基)氨基)-2-甲基-[1,2,4]三唑并[1,5-a][1,3,5]三嗪-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(16)
Example 16: 2-(R)-3-(1-(7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-methyl-[1,2 ,4] Triazolo[1,5-a][1,3,5]triazin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (16)
参照实施例15的合成方法制备化合物16。MS-ESI:m/z 505.2[M+H]+1H NMR(400MHz,DMSO-d6)δ9.27(s,1H),7.65-7.56(m,2H),7.41(dd,J=8.0,2.0Hz,1H),5.61-5.46(m,1H),4.38-4.28(m,1H),4.14-3.57(m,4H),3.54-3.42(m,2H),2.79-2.64(m,2H),2.41-2.33(m,3H),2.31(s,3H),1.98-1.84(m,1H),1.72-1.53(m,4H),1.50(d,J=6.8Hz,3H),1.46-1.33(m,1H),0.85-0.68(m,1H)。Compound 16 was prepared according to the synthesis method of Example 15. MS-ESI: m/z 505.2 [M+H] + . 1 H NMR (400MHz,DMSO-d 6 )δ9.27(s,1H),7.65-7.56(m,2H),7.41(dd,J=8.0,2.0Hz,1H),5.61-5.46(m,1H ),4.38-4.28(m,1H),4.14-3.57(m,4H),3.54-3.42(m,2H),2.79-2.64(m,2H),2.41-2.33(m,3H),2.31(s ,3H),1.98-1.84(m,1H),1.72-1.53(m,4H),1.50(d,J=6.8Hz,3H),1.46-1.33(m,1H),0.85-0.68(m,1H ).
实施例17:2-(R)-3-(1-(7-((R)-1-(2,4-二氯苯基)乙基)氨基)-2-(三氟甲基)-[1,2,4]三唑并[1,5-a][1,3,5]三嗪-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(17)
Example 17: 2-(R)-3-(1-(7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-(trifluoromethyl)- [1,2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)azetidin-3-yl)piperidin-1-yl)ethane -1-ol(17)
参照实施例15的合成方法制备化合物17。MS-ESI:m/z 559.5[M+H]+1H NMR(400MHz,CD3OD)δ7.52-7.46(m,2H),7.34-7.29(m,1H),5.71-5.57(m,1H),4.24-4.07(m,1H),4.02-3.75(m,2H),3.74-3.54(m,3H),2.98-2.82(m,2H),2.59-2.50(m,2H),2.48-2.37(m,1H),2.12-1.99(m,1H),1.82-1.66(m,4H),1.64-1.55(m,4H),0.96-0.81(m,1H)。Compound 17 was prepared according to the synthesis method of Example 15. MS-ESI: m/z 559.5 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.52-7.46(m,2H),7.34-7.29(m,1H),5.71-5.57(m,1H),4.24-4.07(m,1H),4.02- 3.75(m,2H),3.74-3.54(m,3H),2.98-2.82(m,2H),2.59-2.50(m,2H),2.48-2.37(m,1H),2.12-1.99(m,1H ), 1.82-1.66 (m, 4H), 1.64-1.55 (m, 4H), 0.96-0.81 (m, 1H).
实施例18:2-(R)-3-(1-(2-环丙基-7-((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a][1,3,5]三嗪-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(18)
Example 18: 2-(R)-3-(1-(2-cyclopropyl-7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1, 2,4]triazolo[1,5-a][1,3,5]triazin-5-yl)azetidin-3-yl)piperidin-1-yl)ethane-1- Alcohol(18)
参照实施例15的合成方法制备化合物18。MS-ESI:m/z 531.1[M+H]+1H NMR(400MHz,CD3OD)δ7.52-7.43(m,2H),7.35-7.28(m,1H),5.66-5.55(m,1H),4.21-4.09(m,1H),4.03-3.73(m,4H),3.72-3.36(m,3H),3.24-3.08(m,2H),2.93-2.73(m,1H),2.67-2.42(m,2H),2.16-1.71(m,5H),1.59(d,J=7.2Hz,3H),1.23-1.09(m,1H),1.08-0.99(m,4H)。Compound 18 was prepared according to the synthesis method of Example 15. MS-ESI: m/z 531.1 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.52-7.43(m,2H),7.35-7.28(m,1H),5.66-5.55(m,1H),4.21-4.09(m,1H),4.03- 3.73(m,4H),3.72-3.36(m,3H),3.24-3.08(m,2H),2.93-2.73(m,1H),2.67-2.42(m,2H),2.16-1.71(m,5H ), 1.59 (d, J=7.2Hz, 3H), 1.23-1.09 (m, 1H), 1.08-0.99 (m, 4H).
实施例19:2-(R)-3-(1-(7-((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(19)
Example 19: 2-(R)-3-(1-(7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4]triazole And[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (19)
1)化合物19-1的合成 1) Synthesis of Compound 19-1
室温下,将化合物15-1(5000mg,59.47mmol)和丙二酸二乙酯(11.43g,71.36mmol)溶于N,N-二甲基乙酰胺(50mL)中,缓慢加入钠氢(4.76g,118.93mmol,60%纯度),反应混合物在80℃搅拌2小时。反应完毕后,反应液冷却至室温,倒入水中(200mL)震荡溶清,用乙酸乙酯(50mL×3)洗涤,有机相丢弃。水相缓慢滴加2mol/L稀盐酸溶液至pH~2后,用乙酸乙酯(50mL×3)洗涤,有机相丢弃。剩余的水相减压浓缩得到化合物19-1。粗品不经纯化,直接用于下一步反应。MS-ESI:m/z 151.1[M-H]-At room temperature, compound 15-1 (5000mg, 59.47mmol) and diethyl malonate (11.43g, 71.36mmol) were dissolved in N,N-dimethylacetamide (50mL), and sodium hydrogen (4.76 g, 118.93 mmol, 60% purity), the reaction mixture was stirred at 80°C for 2 hours. After the reaction was completed, the reaction solution was cooled to room temperature, poured into water (200 mL) and shaken to dissolve, washed with ethyl acetate (50 mL×3), and the organic phase was discarded. The aqueous phase was slowly added dropwise with 2 mol/L dilute hydrochloric acid solution to pH~2, washed with ethyl acetate (50 mL×3), and the organic phase was discarded. The remaining aqueous phase was concentrated under reduced pressure to obtain compound 19-1. The crude product was directly used in the next reaction without purification. MS-ESI: m/z 151.1 [MH] - .
2)化合物19-2的合成2) Synthesis of compound 19-2
室温下,将化合物19-1(9.05g,59.46mmol)溶于三氯氧磷(50.00mL)中,加入N,N-二甲基苯胺(14.41g,118.93mmol),反应混合物在100℃搅拌3小时。反应完毕后,反应混合物冷却至室温,减压浓缩。所得残余物加入二氯甲烷(100mL)稀释,饱和食盐水(30mL×3)洗涤,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=2/1)分离,得到化合物19-2。MS-ESI:m/z 189.1[M+H]+1H NMR(400MHz,CDCl3)δ8.57(s,1H),7.28(s,1H)。At room temperature, compound 19-1 (9.05g, 59.46mmol) was dissolved in phosphorus oxychloride (50.00mL), N,N-dimethylaniline (14.41g, 118.93mmol) was added, and the reaction mixture was stirred at 100°C 3 hours. After the reaction was completed, the reaction mixture was cooled to room temperature and concentrated under reduced pressure. The resulting residue was diluted with dichloromethane (100 mL), washed with saturated brine (30 mL×3), the organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to obtain compound 19-2. MS-ESI: m/z 189.1 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 8.57 (s, 1H), 7.28 (s, 1H).
3)化合物19-3的合成3) Synthesis of compound 19-3
室温下,将化合物19-2(300mg,1.59mmol)和中间体A(302mg,1.57mmol)溶于二甲基亚砜(10mL)中,加入氟化铯(362mg,2.38mmol),反应混合物在100℃搅拌2小时。反应完毕后,反应液冷却至室温,倒入水(150mL)中稀释,用乙酸乙酯(50mL×3)萃取。合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=2/1)分离,得到化合物19-3。MS-ESI:m/z342.1[M+H]+At room temperature, compound 19-2 (300mg, 1.59mmol) and intermediate A (302mg, 1.57mmol) were dissolved in dimethyl sulfoxide (10mL), cesium fluoride (362mg, 2.38mmol) was added, and the reaction mixture was Stir at 100°C for 2 hours. After the reaction was completed, the reaction solution was cooled to room temperature, poured into water (150 mL) for dilution, and extracted with ethyl acetate (50 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to obtain compound 19-3. MS-ESI: m/z 342.1 [M+H] + .
4)化合物19的合成4) Synthesis of compound 19
室温下,将化合物19-3(200mg,0.42mmol,72%纯度)和中间体C(93mg,0.42mmol)溶于乙腈(10mL)中,加入N,N-二异丙基乙胺(163mg,1.26mmol),反应混合物在80℃搅拌1小时。反应完毕后,反应混合物冷却至室温,减压浓缩。所得残余物经硅胶制备薄层色谱(纯甲醇)分离,得到化合物19。MS-ESI:m/z 490.1[M+H]+1H NMR(400MHz,CD3OD)δ8.08(s,1H),7.54(d,J=2.0Hz,1H),5.53(d,J=8.8Hz,1H),7.36(dd,J=8.4,2.0Hz,1H),5.18-5.11(m,1H),4.83(s,1H),4.20-3.95(m,2H),3.90-3.82(m,3H),3.78-3.67(m,1H),3.46-3.36(m,2H),3.14-3.01(m,2H),2.79-2.64(m,1H),2.63-2.38(m,2H),2.15-2.01(m,1H),1.98-1.81(m,3H),1.67(d,J=6.8Hz,3H),1.17-1.06(m,1H)。At room temperature, compound 19-3 (200mg, 0.42mmol, 72% purity) and intermediate C (93mg, 0.42mmol) were dissolved in acetonitrile (10mL), and N,N-diisopropylethylamine (163mg, 1.26 mmol), and the reaction mixture was stirred at 80°C for 1 hour. After the reaction was completed, the reaction mixture was cooled to room temperature and concentrated under reduced pressure. The obtained residue was separated by silica gel preparative thin-layer chromatography (pure methanol) to obtain Compound 19. MS-ESI: m/z 490.1 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.08(s, 1H), 7.54(d, J=2.0Hz, 1H), 5.53(d, J=8.8Hz, 1H), 7.36(dd, J=8.4 ,2.0Hz,1H),5.18-5.11(m,1H),4.83(s,1H),4.20-3.95(m,2H),3.90-3.82(m,3H),3.78-3.67(m,1H), 3.46-3.36(m,2H),3.14-3.01(m,2H),2.79-2.64(m,1H),2.63-2.38(m,2H),2.15-2.01(m,1H),1.98-1.81(m , 3H), 1.67 (d, J=6.8Hz, 3H), 1.17-1.06 (m, 1H).
实施例20:2-(R)-3-(1-(7-((R)-1-(2,4-二氯苯基)乙基)氨基)-2-甲基-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(20)
Example 20: 2-(R)-3-(1-(7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-methyl-[1,2 ,4] Triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (20)
参照实施例19的合成方法制备实施例20。MS-ESI:m/z 504.2[M+H]+1H NMR(400MHz,CD3OD)δ7.53(d,J=2.4Hz,1H),7.50(d,J=8.4Hz,1H),7.34(dd,J=8.8,2.4Hz,1H),5.14-5.06(m,1H),4.75(s,1H),4.08(t,J=8.4Hz,1H),4.04-3.87(m,1H),3.82-3.56(m,4H),3.16-3.01(m,2H),2.71(t,J=5.6Hz,2H),2.55-2.42(m,1H),2.40(s,3H),2.32-2.21(m,1H),2.01-1.75(m,5H),1.64(d,J=7.2Hz,3H),1.01-0.88(m,1H)。Example 20 was prepared referring to the synthesis method of Example 19. MS-ESI: m/z 504.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.53 (d, J = 2.4Hz, 1H), 7.50 (d, J = 8.4Hz, 1H), 7.34 (dd, J = 8.8, 2.4Hz, 1H), 5.14-5.06(m,1H),4.75(s,1H),4.08(t,J=8.4Hz,1H),4.04-3.87(m,1H),3.82-3.56(m,4H),3.16-3.01( m,2H),2.71(t,J=5.6Hz,2H),2.55-2.42(m,1H),2.40(s,3H),2.32-2.21(m,1H),2.01-1.75(m,5H) , 1.64 (d, J=7.2Hz, 3H), 1.01-0.88 (m, 1H).
实施例21:2-(R)-3-(1-(7-((R)-1-(2,4-二氯苯基)乙基)氨基)-2-(三氟甲基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(21)
Example 21: 2-(R)-3-(1-(7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-(trifluoromethyl)- [1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (21)
参照实施例19的合成方法制备化合物21。MS-ESI:m/z 558.2[M+H]+1H NMR(400MHz,CD3OD)δ7.54(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.8,2.0Hz,1H),5.17-5.09(m,1H),4.89(s,1H),4.16-3.96(m,2H),3.81(dd,J=8.8,6.0Hz,1H),3.75-3.60(m,3H),2.97-2.88(m,2H),2.54(t,J=6.0Hz,2H),2.51-2.42(m,1H),2.08-2.00(m,1H),1.86-1.68(m,5H),1.65(d,J=6.8Hz,3H),1.62-1.52(m,1H),0.97-0.82(m,1H)。Compound 21 was prepared according to the synthesis method of Example 19. MS-ESI: m/z 558.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.54 (d, J = 2.0Hz, 1H), 7.51 (d, J = 8.4Hz, 1H), 7.34 (dd, J = 8.8, 2.0Hz, 1H), 5.17-5.09(m,1H),4.89(s,1H),4.16-3.96(m,2H),3.81(dd,J=8.8,6.0Hz,1H),3.75-3.60(m,3H),2.97- 2.88(m,2H),2.54(t,J=6.0Hz,2H),2.51-2.42(m,1H),2.08-2.00(m,1H),1.86-1.68(m,5H),1.65(d, J=6.8Hz, 3H), 1.62-1.52(m, 1H), 0.97-0.82(m, 1H).
实施例22:2-(R)-3-(1-(7-((R)-1-(2,4-二氯苯基)乙基)氨基)-2-(甲氧基甲基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(22)
Example 22: 2-(R)-3-(1-(7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-(methoxymethyl) -[1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (22)
参照实施例19的合成方法制备化合物22。MS-ESI:m/z 534.2[M+H]+1H NMR(400MHz,CD3OD)δ7.53(d,J=2.0Hz,1H),7.50(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.17-5.07(m,1H),4.79(s,1H),4.55(s,2H),4.08(t,J=8.4Hz,1H),4.04-3.91(m,1H),3.82-3.63(m,4H),3.46(s,3H),3.04-2.91(m,2H),2.59(t,J=6.0Hz,2H),2.52-2.39(m,1H),2.17-2.04(m,1H),1.85-1.70(m,4H),1.67-1.58(m,4H),0.98-0.82(m,1H)。Compound 22 was prepared according to the synthesis method of Example 19. MS-ESI: m/z 534.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.53 (d, J = 2.0Hz, 1H), 7.50 (d, J = 8.4Hz, 1H), 7.34 (dd, J = 8.4, 2.0Hz, 1H), 5.17-5.07(m,1H),4.79(s,1H),4.55(s,2H),4.08(t,J=8.4Hz,1H),4.04-3.91(m,1H),3.82-3.63(m, 4H),3.46(s,3H),3.04-2.91(m,2H),2.59(t,J=6.0Hz,2H),2.52-2.39(m,1H),2.17-2.04(m,1H),1.85 -1.70(m,4H),1.67-1.58(m,4H),0.98-0.82(m,1H).
实施例23:2-(R)-3-(1-(7-((R)-1-(2,4-二氯苯基)乙基)氨基)-2-乙基-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(23)
Example 23: 2-(R)-3-(1-(7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-ethyl-[1,2 ,4] Triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (23)
参照实施例19的合成方法制备化合物23。MS-ESI:m/z 518.2[M+H]+1H NMR(400MHz,CD3OD)δ7.53(d,J=2.4Hz,1H),7.51(t,J=8.4Hz,1H),7.34(dd,J=8.4,2.4Hz,1H),5.17-5.07(m,1H),4.76(s,1H),4.07(t,J=8.8Hz,1H),4.02-3.86(m,1H),3.80-3.73(m,1H),3.73-3.53(m,3H),2.98-2.86(m,2H),2.76(q,J=7.6Hz,2H),2.53(t,J=6.0Hz,2H),2.50-2.41(m,1H),2.08-1.99(m,1H),1.81-1.67(m,4H),1.64(d,J=6.8Hz,3H),1.62-1.53(m,1H),1.35(t,J=7.6Hz,3H),0.95-0.83(m,1H)。Compound 23 was prepared according to the synthesis method of Example 19. MS-ESI: m/z 518.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.53 (d, J = 2.4Hz, 1H), 7.51 (t, J = 8.4Hz, 1H), 7.34 (dd, J = 8.4, 2.4Hz, 1H), 5.17-5.07(m,1H),4.76(s,1H),4.07(t,J=8.8Hz,1H),4.02-3.86(m,1H),3.80-3.73(m,1H),3.73-3.53( m,3H),2.98-2.86(m,2H),2.76(q,J=7.6Hz,2H),2.53(t,J=6.0Hz,2H),2.50-2.41(m,1H),2.08-1.99 (m,1H),1.81-1.67(m,4H),1.64(d,J=6.8Hz,3H),1.62-1.53(m,1H),1.35(t,J=7.6Hz,3H),0.95- 0.83(m,1H).
实施例24:2-(R)-3-(1-(2-环丙基-7-((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(24)
Example 24: 2-(R)-3-(1-(2-cyclopropyl-7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1, 2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (24)
参照实施例19的合成方法制备化合物24。MS-ESI:m/z 530.3[M+H]+1H NMR(400MHz,CD3OD)δ7.52(d,J=2.0Hz,1H),7.50(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.13-5.07(m,1H),4.74(s,1H),4.08(t,J=8.4Hz,1H),4.04-3.94(m,1H),3.86-3.75(m,3H),3.74-3.64(m,1H),3.43-3.34(m,1H),3.04-2.97(m,2H),2.69-2.58(m,1H),2.57-2.45(m,1H),2.42-2.31(m,1H),2.11-2.02(m,1H),2.01-1.95(m,1H),1.93-1.73(m,3H),1.64(d,J=6.8Hz,3H),1.39-1.21(m,1H),1.13-1.04(m,1H),1.03-0.98(m,4H)。Compound 24 was prepared according to the synthesis method of Example 19. MS-ESI: m/z 530.3 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.52(d, J=2.0Hz, 1H), 7.50(d, J=8.4Hz, 1H), 7.34(dd, J=8.4, 2.0Hz, 1H), 5.13-5.07(m,1H),4.74(s,1H),4.08(t,J=8.4Hz,1H),4.04-3.94(m,1H),3.86-3.75(m,3H),3.74-3.64( m,1H),3.43-3.34(m,1H),3.04-2.97(m,2H),2.69-2.58(m,1H),2.57-2.45(m,1H),2.42-2.31(m,1H), 2.11-2.02(m,1H),2.01-1.95(m,1H),1.93-1.73(m,3H),1.64(d,J=6.8Hz,3H),1.39-1.21(m,1H),1.13- 1.04(m,1H),1.03-0.98(m,4H).
实施例25:N-(R)-1-(2,4-二氯苯基)乙基)-5-(3-(R)-2-(四氢-2H-吡喃-4-基)哌啶-3-基)氮杂环丁烷-1-基)-[1,2,4]三唑并[1,5-a]嘧啶-7-胺(25)
Example 25: N-(R)-1-(2,4-dichlorophenyl)ethyl)-5-(3-(R)-2-(tetrahydro-2H-pyran-4-yl) Piperidin-3-yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (25)
1)化合物25-1的合成1) Synthesis of compound 25-1
室温下,将化合物19-3(7.30g,21.31mmol)溶于乙腈(80mL)中,随后加入N,N-二异丙基乙胺(10.50mL,63.92mmol)和中间体E(6.67g,25.57mmol),反应液在60℃反应16小时。反应完毕后,反应液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=1/1)分离,得到化合物25-1。MS-ESI:m/z 530.4[M+H]+At room temperature, compound 19-3 (7.30g, 21.31mmol) was dissolved in acetonitrile (80mL), then N,N-diisopropylethylamine (10.50mL, 63.92mmol) and intermediate E (6.67g, 25.57mmol), and the reaction solution was reacted at 60°C for 16 hours. After the reaction was completed, the reaction solution was concentrated under reduced pressure, and the resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to obtain compound 25-1. MS-ESI: m/z 530.4 [M+H] + .
2)化合物25-2的合成2) Synthesis of compound 25-2
室温下,将化合物25-1(2.70g,5.09mmol)溶于二氯甲烷(50mL)中,随后加入苯硅烷(5.51g,50.90mmol)和四(三苯基膦)钯(590mg,0.51mmol),反应液在室温 搅拌15分钟。反应完毕后,反应液经硅胶柱层析(甲醇/三乙胺=5/1)分离,得到化合物25-2。MS-ESI:m/z 446.1[M+H]+Compound 25-1 (2.70g, 5.09mmol) was dissolved in dichloromethane (50mL) at room temperature, followed by the addition of phenylsilane (5.51g, 50.90mmol) and tetrakis(triphenylphosphine)palladium (590mg, 0.51mmol ), the reaction solution at room temperature Stir for 15 minutes. After the reaction was completed, the reaction solution was separated by silica gel column chromatography (methanol/triethylamine=5/1) to obtain compound 25-2. MS-ESI: m/z 446.1 [M+H] + .
3)化合物25的合成3) Synthesis of compound 25
室温下,将化合物25-2(100mg,0.22mmol),四氢吡喃-4-酮(67mg,0.67mmol)和乙酸(13mg,0.22mmol)溶于甲醇(5mL)中,反应混合物在25℃搅拌1小时,随后将氰基硼氢化钠(42mg,0.67mmol)加入到反应液中,反应混合物在25℃搅拌12小时。反应液倒入水(30mL)中,用二氯甲烷(20mL×3)萃取,合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经制备级高效液相色谱(乙腈/碳酸氢胺体系,色谱柱:ACSWH-GX-A Waters Xbridge 150*25mm*5μm;流动相:水(0.1%碳酸氢胺),乙腈;梯度配比:乙腈相(0-9min,60-90%);流速:25mL/min;柱温:室温)分离,得到化合物25。MS-ESI:m/z 530.2[M+H]+1H NMR(400MHz,CD3OD)δ8.03(s,1H),7.52(d,J=2.0Hz,1H),7.50(d,J=8.4Hz,1H),7.33(dd,J=8.0,2.0Hz,1H),5.18-5.12(m,1H),4.80(s,1H),4.08(t,J=8.4Hz,1H),3.98(dd,J=11.2,3.6Hz,3H),3.77(dd,J=8.8,6.0Hz,1H),3.72-3.51(m,1H),3.38(t,J=12.0Hz,2H),2.97-2.86(m,2H),2.57-2.41(m,2H),2.13(td,J=12.0,2.8Hz,1H),1.87-1.68(m,6H),1.64(d,J=6.4Hz,3H),1.62-1.48(m,3H),0.95-0.82(m,1H)。At room temperature, compound 25-2 (100mg, 0.22mmol), tetrahydropyran-4-one (67mg, 0.67mmol) and acetic acid (13mg, 0.22mmol) were dissolved in methanol (5mL), and the reaction mixture was heated at 25°C After stirring for 1 hour, sodium cyanoborohydride (42 mg, 0.67 mmol) was added to the reaction solution, and the reaction mixture was stirred at 25° C. for 12 hours. The reaction solution was poured into water (30 mL), extracted with dichloromethane (20 mL×3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the obtained residue was subjected to preparative high performance liquid chromatography ( Acetonitrile/ammonium bicarbonate system, chromatographic column: ACSWH-GX-A Waters Xbridge 150*25mm*5μm; mobile phase: water (0.1% ammonium bicarbonate), acetonitrile; gradient ratio: acetonitrile phase (0-9min,60- 90%); flow rate: 25mL/min; column temperature: room temperature) to obtain compound 25. MS-ESI: m/z 530.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.03(s, 1H), 7.52(d, J=2.0Hz, 1H), 7.50(d, J=8.4Hz, 1H), 7.33(dd, J=8.0 ,2.0Hz,1H),5.18-5.12(m,1H),4.80(s,1H),4.08(t,J=8.4Hz,1H),3.98(dd,J=11.2,3.6Hz,3H),3.77 (dd, J=8.8,6.0Hz,1H),3.72-3.51(m,1H),3.38(t,J=12.0Hz,2H),2.97-2.86(m,2H),2.57-2.41(m,2H ), 2.13(td, J=12.0, 2.8Hz, 1H), 1.87-1.68(m, 6H), 1.64(d, J=6.4Hz, 3H), 1.62-1.48(m, 3H), 0.95-0.82( m, 1H).
实施例26:3-(R)-3-(1-(7-((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)丙烷-1-醇(26)
Example 26: 3-(R)-3-(1-(7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4]triazole And[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)propan-1-ol (26)
室温下,将化合物25-2(100mg,0.22mmol)溶于乙腈(6mL)中,随后加入3-碘丙醇(83mg,0.44mmol)和碳酸钾(92mg,0.67mmol),反应混合物在80℃搅拌3小时。反应液减压浓缩,所得残余物经制备级高效液相色谱(碳酸氢铵/乙腈/水体系,色谱柱:Waters Xbridge C18 150*25mm*5μm;流动相:水(10mM碳酸氢铵),乙腈;梯度配比:乙腈相(0-10min,35-65%);流速:25mL/min;柱温:室温)分离,得到化合物26。MS-ESI:m/z 504.3[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.53(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.18-5.10(m,1H),4.81(s,1H),4.09(t,J=8.4Hz,1H),4.05-3.89(m,1H),3.78(dd,J=8.4,5.6Hz,1H),3.74-3.55(m,3H),2.98-2.83(m,2H),2.53-2.41(m,3H),2.03-1.91(m,1H),1.82-1.70(m,5H),1.69-1.61(m,4H),1.61-1.49(m,1H),0.98-0.82(m,1H)。 At room temperature, compound 25-2 (100mg, 0.22mmol) was dissolved in acetonitrile (6mL), then 3-iodopropanol (83mg, 0.44mmol) and potassium carbonate (92mg, 0.67mmol) were added, and the reaction mixture was heated at 80°C Stir for 3 hours. The reaction solution was concentrated under reduced pressure, and the resulting residue was subjected to preparative high-performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge C18 150*25mm*5μm; mobile phase: water (10mM ammonium bicarbonate), acetonitrile ; Gradient ratio: acetonitrile phase (0-10min, 35-65%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 26. MS-ESI: m/z 504.3 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.53(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz,1H),5.18-5.10(m,1H),4.81(s,1H),4.09(t,J=8.4Hz,1H),4.05-3.89(m,1H),3.78(dd,J= 8.4,5.6Hz,1H),3.74-3.55(m,3H),2.98-2.83(m,2H),2.53-2.41(m,3H),2.03-1.91(m,1H),1.82-1.70(m, 5H), 1.69-1.61(m, 4H), 1.61-1.49(m, 1H), 0.98-0.82(m, 1H).
实施例27:N-(2-((R)-3-(1-(7-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙基)甲磺酰胺(27)
Example 27: N-(2-((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2 ,4] Triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethyl)methanesulfonamide (27)
1)化合物27-1的合成1) Synthesis of compound 27-1
室温下,将化合物25-2(200mg,0.45mmol),(2-溴乙基)氨基甲酸叔丁酯(100mg,0.45mmol)和碳酸钾(186mg,1.34mmol)溶于乙腈(10mL)中,反应混合物在80℃搅拌2小时。反应完毕后,加水(30mL)稀释,用乙酸乙酯(30mL×3)萃取。合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=0/1)分离,得到化合物27-1。MS-ESI:m/z 589.2[M+H]+Compound 25-2 (200 mg, 0.45 mmol), tert-butyl (2-bromoethyl)carbamate (100 mg, 0.45 mmol) and potassium carbonate (186 mg, 1.34 mmol) were dissolved in acetonitrile (10 mL) at room temperature, The reaction mixture was stirred at 80°C for 2 hours. After the reaction was completed, it was diluted with water (30 mL), and extracted with ethyl acetate (30 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=0/1) to obtain compound 27-1. MS-ESI: m/z 589.2 [M+H] + .
2)化合物27-2的合成2) Synthesis of compound 27-2
室温下,将化合物27-1(300mg,0.43mmol,85%纯度)溶于二氯甲烷(5mL)中,随后加入三氟乙酸(1.00mL),反应混合物在室温搅拌1小时。反应完毕后,加水(30mL)稀释,用乙酸乙酯(30mL×3)洗涤,水相用饱和碳酸氢钠水溶液调节至pH~8,用乙酸乙酯(30mL×3)萃取。合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物27-2。MS-ESI:m/z 489.3[M+H]+Compound 27-1 (300 mg, 0.43 mmol, 85% purity) was dissolved in dichloromethane (5 mL) at room temperature, then trifluoroacetic acid (1.00 mL) was added, and the reaction mixture was stirred at room temperature for 1 hour. After the reaction was completed, it was diluted with water (30 mL), washed with ethyl acetate (30 mL×3), the aqueous phase was adjusted to pH~8 with saturated aqueous sodium bicarbonate solution, and extracted with ethyl acetate (30 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound 27-2. MS-ESI: m/z 489.3 [M+H] + .
3)化合物27合成3) Synthesis of Compound 27
冰浴下,将化合物27-2(55mg,0.10mmol,89%纯度),甲基磺酸酐(16mg,0.09mmol)和三乙胺(30mg,0.30mmol)溶于二氯甲烷(2mL)中,反应混合物在0℃下搅拌1.5小时。反应完毕后,反应液减压浓缩。剩余物经制备级高效液相色谱(碳酸氢铵/乙腈/水体系,色谱柱:Waters Xbridge 150*25mm*5μm;流动相:水(0.5%碳酸氢铵),乙腈;梯度配比:乙腈相(0-9min,45-75%);流速:25mL/min;柱温:室温)分离,得到化合物27。MS-ESI:m/z 567.3[M+H]+1H NMR(400MHz,DMSO-d6)δ8.34(d,J=6.8Hz,1H),8.11(s,1H),7.65(d,J=2.0Hz,1H),7.63(d,J=8.4Hz,1H),7.44(dd,J=8.4,2.0Hz,1H),6.91-6.76(m,1H),5.06-4.96(m,1H),4.84(s,1H),3.99(t,J=8.8Hz,1H),3.95-3.86(m,1H),3.73-3.69(m,1H),3.64-3.56(m,1H),3.04(t,J=6.4Hz,2H),2.92(s,3H),2.79-2.70(m,2H),2.62-2.57(m,1H),2.38(t,J=6.8Hz,2H),1.96-1.87(m,1H),1.68-1.60(m,3H),1.59-1.55(m,4H),1.49-1.36(m,1H),0.87-0.74(m,1H)。Under ice-cooling, compound 27-2 (55 mg, 0.10 mmol, 89% purity), methanesulfonic anhydride (16 mg, 0.09 mmol) and triethylamine (30 mg, 0.30 mmol) were dissolved in dichloromethane (2 mL), The reaction mixture was stirred at 0°C for 1.5 hours. After the reaction was completed, the reaction solution was concentrated under reduced pressure. The residue was subjected to preparative high-performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge 150*25mm*5 μm; mobile phase: water (0.5% ammonium bicarbonate), acetonitrile; gradient ratio: acetonitrile phase (0-9min, 45-75%); flow rate: 25mL/min; column temperature: room temperature) to obtain compound 27. MS-ESI: m/z 567.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 )δ8.34(d, J=6.8Hz, 1H), 8.11(s, 1H), 7.65(d, J=2.0Hz, 1H), 7.63(d, J= 8.4Hz,1H),7.44(dd,J=8.4,2.0Hz,1H),6.91-6.76(m,1H),5.06-4.96(m,1H),4.84(s,1H),3.99(t,J =8.8Hz,1H),3.95-3.86(m,1H),3.73-3.69(m,1H),3.64-3.56(m,1H),3.04(t,J=6.4Hz,2H),2.92(s, 3H), 2.79-2.70(m, 2H), 2.62-2.57(m, 1H), 2.38(t, J=6.8Hz, 2H), 1.96-1.87(m, 1H), 1.68-1.60(m, 3H) ,1.59-1.55(m,4H),1.49-1.36(m,1H),0.87-0.74(m,1H).
实施例28:N-((R)-1-(2,4-二氯苯基)乙基)-5-(3-(R)-2-(2-(甲基磺酰基)乙基)哌啶-3-基)氮杂环丁烷-1-基)-[1,2,4]三唑并[1,5-a]嘧啶-7-胺(28)
Example 28: N-((R)-1-(2,4-dichlorophenyl)ethyl)-5-(3-(R)-2-(2-(methylsulfonyl)ethyl) Piperidin-3-yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (28)
1)化合物28-2的合成1) Synthesis of compound 28-2
室温下,将化合物28-1(1.00g,7.24mmol)溶于吡啶(10mL)中,将体系降温到0℃,加入对甲苯磺酰氯(1.69g,8.86mmol),反应混合物在25℃搅拌12小时。反应液倒入水(20mL)中淬灭,用乙酸乙酯(30mL)萃取。有机相分别用1mol/L的稀盐酸(30mL),饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(二氯甲烷/甲醇=0/1)分离,得到化合物28-2,粗品不经纯化,直接用于下一步反应。At room temperature, compound 28-1 (1.00g, 7.24mmol) was dissolved in pyridine (10mL), the system was cooled to 0°C, p-toluenesulfonyl chloride (1.69g, 8.86mmol) was added, and the reaction mixture was stirred at 25°C for 12 Hour. The reaction solution was quenched by pouring into water (20 mL), and extracted with ethyl acetate (30 mL). The organic phase was washed with 1 mol/L dilute hydrochloric acid (30 mL) and saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting residue was separated by silica gel column chromatography (dichloromethane/methanol = 0/1) to obtain compound 28-2. The crude product was directly used in the next reaction without further purification.
2)化合物28的合成2) Synthesis of compound 28
室温下,将化合物28-2(488mg,1.75mmol)和化合物25-2(300mg,0.59mmol)溶于乙腈(10mL)中,加入N,N-二异丙基乙胺(227mg,1.75mmol),混合物在80℃搅拌12小时。反应完成后,将反应液冷却到室温,倒入水(20mL)中稀释,用二氯甲烷(10mL×2)萃取。合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶柱层析(乙酸乙酯/甲醇=0/1)分离,得到化合物28。MS-ESI:m/z552.1[M+H]+1H NMR(400MHz,CD3OD)δ8.03(s,1H),7.52(d,J=2.0Hz,1H),7.49(d,J=8.4Hz,1H),7.32(dd,J=8.4,2.0Hz,1H),5.15-5.07(m,1H),4.79(s,1H),4.07(t,J=8.4Hz,1H),4.03-3.91(m,1H),3.78(dd,J=8.4,6.0Hz,1H),3.74-3.56(m,1H),3.29-3.25(m,2H),3.03(s,3H),2.89-2.78(m,4H),2.55-2.41(m,1H),2.10-1.98(m,1H),1.80-1.66(m,4H),1.64(d,J=7.2Hz,3H),1.61-1.47(m,1H),0.97-0.83(m,1H)。At room temperature, compound 28-2 (488mg, 1.75mmol) and compound 25-2 (300mg, 0.59mmol) were dissolved in acetonitrile (10mL), and N,N-diisopropylethylamine (227mg, 1.75mmol) was added , and the mixture was stirred at 80°C for 12 hours. After the reaction was completed, the reaction solution was cooled to room temperature, poured into water (20 mL) for dilution, and extracted with dichloromethane (10 mL×2). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting residue was separated by silica gel column chromatography (ethyl acetate/methanol=0/1) to obtain compound 28. MS-ESI: m/z 552.1 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.03(s, 1H), 7.52(d, J=2.0Hz, 1H), 7.49(d, J=8.4Hz, 1H), 7.32(dd, J=8.4 ,2.0Hz,1H),5.15-5.07(m,1H),4.79(s,1H),4.07(t,J=8.4Hz,1H),4.03-3.91(m,1H),3.78(dd,J= 8.4,6.0Hz,1H),3.74-3.56(m,1H),3.29-3.25(m,2H),3.03(s,3H),2.89-2.78(m,4H),2.55-2.41(m,1H) , 2.10-1.98 (m, 1H), 1.80-1.66 (m, 4H), 1.64 (d, J=7.2Hz, 3H), 1.61-1.47 (m, 1H), 0.97-0.83 (m, 1H).
实施例29:5-(3-((R)-1-(2-氧杂螺[3.3]庚烷-6-基)哌啶-3-基)氮杂环丁烷-1-基)-N-((R-1-(2,4-二氯苯基)乙基)-[1,2,4]三唑并[1,5-a]嘧啶-7-胺(29)
Example 29: 5-(3-((R)-1-(2-oxaspiro[3.3]heptan-6-yl)piperidin-3-yl)azetidin-1-yl)- N-((R-1-(2,4-dichlorophenyl)ethyl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (29)
室温下,将化合物25-2(100mg,0.22mmol)溶于甲醇(5mL)中,随后加入2-氧杂螺[3.3]庚烷-6-酮(74mg,0.51mmol),氰基硼氢化钠(41mg,0.66mmol)和醋酸(40mg,0.66mmol),反应混合物在25℃搅拌16小时。反应液减压浓缩,所得残余物经制备级高效液相色谱(碳酸氢铵/乙腈/水体系,色谱柱:Waters Xbridge C18150*25mm*5μm;流动相:水(10mM碳酸氢铵),乙腈;梯度配比:乙腈相(0-10min,46-76%);流速:25mL/min;柱温:室温)分离,得到化合物29。MS-ESI: m/z 542.3[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.54(d,J=2.4Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.16-5.11(m,1H),4.81(s,1H),4.72(s,2H),4.59(s,2H),4.08(t,J=8.4Hz,1H),4.04-3.86(m,1H),3.82-3.75(m,1H),3.74-3.54(m,1H),2.88-2.72(m,2H),2.61-2.51(m,1H),2.50-2.37(m,3H),2.11-1.98(m,2H),1.83-1.68(m,4H),1.65(d,J=6.8Hz,3H),1.59-1.48(m,1H),1.42(t,J=10.4Hz,1H),0.97-0.81(m,1H)。Compound 25-2 (100mg, 0.22mmol) was dissolved in methanol (5mL) at room temperature, followed by the addition of 2-oxaspiro[3.3]heptane-6-one (74mg, 0.51mmol), sodium cyanoborohydride (41mg, 0.66mmol) and acetic acid (40mg, 0.66mmol), the reaction mixture was stirred at 25°C for 16 hours. The reaction solution was concentrated under reduced pressure, and the obtained residue was subjected to preparative high performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge C18 150*25mm*5μm; mobile phase: water (10mM ammonium bicarbonate), acetonitrile; Gradient ratio: acetonitrile phase (0-10min, 46-76%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 29. MS-ESI: m/z 542.3[M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.54(d, J=2.4Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz,1H),5.16-5.11(m,1H),4.81(s,1H),4.72(s,2H),4.59(s,2H),4.08(t,J=8.4Hz,1H),4.04 -3.86(m,1H),3.82-3.75(m,1H),3.74-3.54(m,1H),2.88-2.72(m,2H),2.61-2.51(m,1H),2.50-2.37(m, 3H), 2.11-1.98(m, 2H), 1.83-1.68(m, 4H), 1.65(d, J=6.8Hz, 3H), 1.59-1.48(m, 1H), 1.42(t, J=10.4Hz ,1H), 0.97-0.81(m,1H).
实施例30:3-((R)-3-(1-(7-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)硫杂环丁烷-1,1-二氧化物(30)
Example 30: 3-((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4] Triazol[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)thietane-1,1-dioxide (30)
1)化合物30-2的合成1) Synthesis of compound 30-2
冰浴下,将化合物30-1(500mg,4.09mmol),甲基磺酸酐(1.07g,6.14mmol)和三乙胺(1.70mL,12.28mmol)溶于二氯甲烷(10mL)中,反应混合物室温搅拌1小时,得到化合物30-2,反应液直接用于下一步反应。Under ice-cooling, compound 30-1 (500mg, 4.09mmol), methanesulfonic anhydride (1.07g, 6.14mmol) and triethylamine (1.70mL, 12.28mmol) were dissolved in dichloromethane (10mL), and the reaction mixture Stir at room temperature for 1 hour to obtain compound 30-2, and the reaction solution was directly used in the next reaction.
2)化合物30的合成2) Synthesis of compound 30
室温下,将化合物25-2(100mg,0.22mmol),化合物30-2(224mg,1.12mmol,粗品,理论量)和碳酸铯(146mg,0.45mmol)溶于N,N-二甲基甲酰胺(2mL)中,反应混合物在室温搅拌8小时。反应完毕后,向反应混合物中加水(20mL)稀释,用乙酸乙酯(30mL×3)萃取,合并有机相,用饱和氯化钠水溶液(30mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经制备级高效液相色谱(甲酸/乙腈/水体系,色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:水(0.5%甲酸),乙腈;梯度配比:乙腈相(0-10min,18-48%);流速:25mL/min;柱温:室温)分离,得到化合物30。MS-ESI:m/z 550.4[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.53(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.17-5.10(m,1H),4.81(s,1H),4.21-4.14(m,2H),4.12-3.94(m,4H),3.83-3.78(m,1H),3.76-3.62(m,1H),3.23-3.13(m,1H),2.85-2.73(m,2H),2.60-2.47(m,1H),2.04-1.93(m,1H),1.81-1.72(m,3H),1.71-1.67(m,1H),1.65(d,J=6.8Hz,3H),1.62-1.53(m,1H),0.99-0.86(m,1H)。At room temperature, compound 25-2 (100mg, 0.22mmol), compound 30-2 (224mg, 1.12mmol, crude product, theoretical amount) and cesium carbonate (146mg, 0.45mmol) were dissolved in N,N-dimethylformamide (2 mL), the reaction mixture was stirred at room temperature for 8 hours. After the reaction was complete, add water (20 mL) to the reaction mixture to dilute, extract with ethyl acetate (30 mL×3), combine the organic phases, wash with saturated aqueous sodium chloride solution (30 mL×3), dry over anhydrous sodium sulfate, filter, The filtrate was concentrated under reduced pressure. The obtained residue was subjected to preparative high-performance liquid chromatography (formic acid/acetonitrile/water system, chromatographic column: Phenomenex luna C18 150*25mm*10 μm; mobile phase: water (0.5% formic acid), acetonitrile; gradient ratio: acetonitrile phase (0 -10min, 18-48%)); flow rate: 25mL/min; column temperature: room temperature) to obtain compound 30. MS-ESI: m/z 550.4 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.53(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz,1H),5.17-5.10(m,1H),4.81(s,1H),4.21-4.14(m,2H),4.12-3.94(m,4H),3.83-3.78(m,1H), 3.76-3.62(m,1H),3.23-3.13(m,1H),2.85-2.73(m,2H),2.60-2.47(m,1H),2.04-1.93(m,1H),1.81-1.72(m , 3H), 1.71-1.67(m, 1H), 1.65(d, J=6.8Hz, 3H), 1.62-1.53(m, 1H), 0.99-0.86(m, 1H).
实施例31和实施例32:(1R,4r)-4-((R)-3-(1-(7-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)-1-甲基环己烷-1-醇 和(1S,4s)-4-((R)-3-(1-(7-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)-1-甲基环己烷-1-醇(31和32)
Example 31 and Example 32: (1R,4r)-4-((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl) Amino)-[1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclohexyl alkan-1-ol and (1S,4s)-4-((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2 ,4] Triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclohexane-1-alcohol (31 and 32)
1)化合物31-2的合成1) Synthesis of compound 31-2
冰浴下,将化合物31-1(1000mg,6.40mmol)溶于四氢呋喃(15mL)中,氮气置换,随后加入3mol/L甲基溴化镁(2.50mL,7.50mmol)的乙醚溶液,反应液在0℃搅拌2小时。反应结束后,将反应液缓慢倒入饱和氯化铵的水溶液(50mL)中淬灭,乙酸乙酯(50mL)萃取,饱和食盐水(40mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物31-2。1H NMR(400MHz,CDCl3)δ3.97-3.86(m,4H),1.94-1.81(m,2H),1.73-1.52(m,6H),1.27-1.23(m,3H)。Under ice-cooling, compound 31-1 (1000mg, 6.40mmol) was dissolved in tetrahydrofuran (15mL), replaced with nitrogen, and then a 3mol/L methylmagnesium bromide (2.50mL, 7.50mmol) ether solution was added, and the reaction solution was Stir at 0°C for 2 hours. After the reaction, the reaction solution was slowly poured into saturated ammonium chloride aqueous solution (50mL) to quench, extracted with ethyl acetate (50mL), washed with saturated brine (40mL×3), dried over anhydrous sodium sulfate, filtered, and the filtrate Concentration under reduced pressure gave compound 31-2. 1 H NMR (400 MHz, CDCl 3 ) δ 3.97-3.86 (m, 4H), 1.94-1.81 (m, 2H), 1.73-1.52 (m, 6H), 1.27-1.23 (m, 3H).
2)化合物31-3的合成2) Synthesis of compound 31-3
室温下,将化合物31-2(1.00g,4.65mmol,80%纯度)溶于四氢呋喃(15mL)中,随后加入1mol/L稀盐酸(8.00mL,8.00mmol),反应液在25℃搅拌16小时。反应液用乙酸乙酯(50mL)萃取,饱和食盐水(40mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=1/1)分离,得到化合物31-3。1H NMR(400MHz,CDCl3)δ2.75-2.66(m,2H),2.26-2.19(m,2H),1.99-1.93(m,2H),1.88-1.80(m,2H),1.35(s,3H)。At room temperature, compound 31-2 (1.00g, 4.65mmol, 80% purity) was dissolved in tetrahydrofuran (15mL), then 1mol/L dilute hydrochloric acid (8.00mL, 8.00mmol) was added, and the reaction solution was stirred at 25°C for 16 hours . The reaction solution was extracted with ethyl acetate (50 mL), washed with saturated brine (40 mL×3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to obtain compound 31-3. 1 H NMR (400MHz, CDCl 3 ) δ2.75-2.66(m,2H),2.26-2.19(m,2H),1.99-1.93(m,2H),1.88-1.80(m,2H),1.35(s ,3H).
3)化合物31和化合物32的合成3) Synthesis of compound 31 and compound 32
室温下,将化合物31-3(209mg,1.30mmol,80%纯度)溶于1,2-二氯乙烷(10mL)中,随后加入化合物25-2(300mg,0.65mmol)和醋酸(117mg,1.96mmol),反应混合物在25℃搅拌2小时,然后加入醋酸硼氢化钠(413mg,1.96mmol),反应混合物在50℃下搅拌14小时。反应结束后,反应液减压浓缩,所得残余物经硅胶柱层析(乙酸乙酯/甲醇=0/1)分离,再经硅胶制备薄层色谱(纯甲醇)分离,得到化合物31-4。再经制备超临界流体色谱(色谱柱:DAICEL CHIRALPAK IC(250mm*50mm*10μm);流动相:己烷,乙醇(0.1%氨水);梯度配比:乙醇相70%;流速:100mL/min;柱温:室温)分离,得到化合物31和化合物32。(化合物31为第一个洗脱的峰,化合物32为第二个洗脱的峰)。化合物31:MS-ESI:m/z558.3[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.54(d,J=2.0Hz,1H),7.51(d,J=8.8Hz,1H),7.34(dd,J=8.8,2.4Hz,1H),5.17-5.09(m,1H),4.81(s,1H),4.09(t,J=8.4Hz,1H),4.04-3.88(m,1H),3.81-3.73(m,1H),3.72-3.54(m,1H),2.98-2.82(m,2H),2.52-2.42(m,1H),2.41-2.31(m,1H),2.30-2.21(m,1H),1.98-1.90(m,1H),1.82-1.69(m,6H),1.67-1.62(m,4H),1.61-1.54(m,1H),1.46-1.26(m,4H),1.18(s,3H),0.92-0.84(m,1H)。化合物32:MS-ESI:m/z 558.3[M+1]+1H NMR(400 MHz,CD3OD)δ8.04(s,1H),7.54(d,J=2.4Hz,1H),7.51(d,J=8.8Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.19-5.11(m,1H),4.81(s,1H),4.09(t,J=8.4Hz,1H),4.05-3.92(m,1H),3.81-3.74(m,1H),3.72-3.54(m,1H),2.97-2.82(m,2H),2.53-2.42(m,1H),2.39-2.30(m,1H),2.22-2.13(m,1H),1.91-1.80(m,3H),1.79-1.68(m,5H),1.65(d,J=10.8Hz,3H),1.53-1.41(m,3H),1.40-1.27(m,2H),1.22(s,3H),0.94-0.84(m,1H)。At room temperature, compound 31-3 (209 mg, 1.30 mmol, 80% purity) was dissolved in 1,2-dichloroethane (10 mL), then compound 25-2 (300 mg, 0.65 mmol) and acetic acid (117 mg, 1.96mmol), the reaction mixture was stirred at 25°C for 2 hours, then sodium acetate borohydride (413mg, 1.96mmol) was added, and the reaction mixture was stirred at 50°C for 14 hours. After the reaction, the reaction solution was concentrated under reduced pressure, and the obtained residue was separated by silica gel column chromatography (ethyl acetate/methanol=0/1), and then separated by preparative silica gel thin layer chromatography (pure methanol) to obtain compound 31-4. Then prepare supercritical fluid chromatography (column: DAICEL CHIRALPAK IC (250mm*50mm*10μm); mobile phase: hexane, ethanol (0.1% ammonia water); gradient ratio: ethanol phase 70%; flow rate: 100mL/min; Column temperature: room temperature) separation to obtain compound 31 and compound 32. (Compound 31 is the first eluting peak, compound 32 is the second eluting peak). Compound 31: MS-ESI: m/z 558.3 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.54(d, J=2.0Hz, 1H), 7.51(d, J=8.8Hz, 1H), 7.34(dd, J=8.8 ,2.4Hz,1H),5.17-5.09(m,1H),4.81(s,1H),4.09(t,J=8.4Hz,1H),4.04-3.88(m,1H),3.81-3.73(m, 1H),3.72-3.54(m,1H),2.98-2.82(m,2H),2.52-2.42(m,1H),2.41-2.31(m,1H),2.30-2.21(m,1H),1.98- 1.90(m,1H),1.82-1.69(m,6H),1.67-1.62(m,4H),1.61-1.54(m,1H),1.46-1.26(m,4H),1.18(s,3H), 0.92-0.84 (m, 1H). Compound 32: MS-ESI: m/z 558.3 [M+1] + . 1 H NMR (400 MHz, CD 3 OD) δ8.04(s, 1H), 7.54(d, J=2.4Hz, 1H), 7.51(d, J=8.8Hz, 1H), 7.34(dd, J=8.4, 2.0Hz, 1H),5.19-5.11(m,1H),4.81(s,1H),4.09(t,J=8.4Hz,1H),4.05-3.92(m,1H),3.81-3.74(m,1H),3.72 -3.54(m,1H),2.97-2.82(m,2H),2.53-2.42(m,1H),2.39-2.30(m,1H),2.22-2.13(m,1H),1.91-1.80(m, 3H),1.79-1.68(m,5H),1.65(d,J=10.8Hz,3H),1.53-1.41(m,3H),1.40-1.27(m,2H),1.22(s,3H),0.94 -0.84(m,1H).
实施例33:(1S,3s)-3-((R)-3-(1-(7-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)-1-甲基环丁烷-1-基(33)和实施例34:(1R,3r)-3-((R)-3-(1-(7-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)-1-甲基环丁烷-1-醇(34)
Example 33: (1S,3s)-3-((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[ 1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclobutane-1- Base (33) and Example 34: (1R,3r)-3-((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl )amino)-[1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)-1-methyl ring Butan-1-ol(34)
室温下,将化合物25-2(200mg,0.44mmol)溶于甲醇(6mL)中,随后加入化合物33-1(147mg,0.88mmol,60%纯度),氰基硼氢化钠(83mg,1.32mmol)和醋酸(79mg,1.32mmol),反应混合物在25℃搅拌16小时。反应液过滤,滤液减压浓缩,得到化合物33-2。化合物33-2(200mg,90%纯度)经正相液相色谱(色谱柱:DAICEL CHIRALPAK IC(250mm*50mm*10μm);流动相:己烷,乙醇(0.1%氨水);梯度配比:乙醇相70%;流速:100mL/min;柱温:室温)分离,再经制备薄层色谱(纯甲醇)分离,得到化合物33和化合物34。化合物33:MS-ESI:m/z530.2[M+H]+1H NMR(400MHz,CD3OD)δ8.06(s,1H),7.58-7.46(m,2H),7.34(dd,J=8.8,2.4Hz,1H),5.17-5.11(m,1H),4.59(s,1H),4.13(t,J=8.4Hz,1H),4.08-3.93(m,1H),3.92-3.84(m,1H),3.83-3.64(m,2H),3.53-3.37(m,2H),2.81-2.64(m,1H),2.62-2.30(m,6H),2.23-2.11(m,1H),2.02-1.82(m,3H),1.66(d,J=6.8Hz,3H),1.40(s,3H),1.24-1.09(m,1H)。化合物34:MS-ESI:m/z 530.3[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.52(d,J=1.6Hz,1H),7.50(d,J=8.4Hz,1H),7.33(dd,J=8.4,1.6Hz,1H),5.19-5.06(m,1H),4.80(s,1H),4.08(t,J=8.4Hz,1H),4.03-3.88(m,1H),3.82-3.74(m,1H),3.73-3.54(m,1H),2.90-2.74(m,2H),2.53-2.40(m,1H),2.38-2.28(m,1H),2.24-2.12(m,2H),2.00-1.87(m,2H),1.83-1.68(m,4H),1.65(d,J=6.8Hz,3H),1.60-1.51(m,1H),1.50-1.40(m,1H),1.30(s,3H),0.92-0.80(m,1H)。At room temperature, compound 25-2 (200 mg, 0.44 mmol) was dissolved in methanol (6 mL), followed by addition of compound 33-1 (147 mg, 0.88 mmol, 60% purity), sodium cyanoborohydride (83 mg, 1.32 mmol) and acetic acid (79mg, 1.32mmol), the reaction mixture was stirred at 25°C for 16 hours. The reaction solution was filtered, and the filtrate was concentrated under reduced pressure to obtain compound 33-2. Compound 33-2 (200mg, 90% purity) was subjected to normal phase liquid chromatography (column: DAICEL CHIRALPAK IC (250mm*50mm*10μm); mobile phase: hexane, ethanol (0.1% ammonia water); gradient ratio: ethanol Phase 70%; flow rate: 100mL/min; column temperature: room temperature) separation, and then separated by preparative thin-layer chromatography (pure methanol) to obtain compound 33 and compound 34. Compound 33: MS-ESI: m/z 530.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.06(s, 1H), 7.58-7.46(m, 2H), 7.34(dd, J=8.8, 2.4Hz, 1H), 5.17-5.11(m, 1H) ,4.59(s,1H),4.13(t,J=8.4Hz,1H),4.08-3.93(m,1H),3.92-3.84(m,1H),3.83-3.64(m,2H),3.53-3.37 (m,2H),2.81-2.64(m,1H),2.62-2.30(m,6H),2.23-2.11(m,1H),2.02-1.82(m,3H),1.66(d,J=6.8Hz ,3H), 1.40(s,3H), 1.24-1.09(m,1H). Compound 34: MS-ESI: m/z 530.3 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.52(d, J=1.6Hz, 1H), 7.50(d, J=8.4Hz, 1H), 7.33(dd, J=8.4 ,1.6Hz,1H),5.19-5.06(m,1H),4.80(s,1H),4.08(t,J=8.4Hz,1H),4.03-3.88(m,1H),3.82-3.74(m, 1H),3.73-3.54(m,1H),2.90-2.74(m,2H),2.53-2.40(m,1H),2.38-2.28(m,1H),2.24-2.12(m,2H),2.00- 1.87(m,2H),1.83-1.68(m,4H),1.65(d,J=6.8Hz,3H),1.60-1.51(m,1H),1.50-1.40(m,1H),1.30(s, 3H), 0.92-0.80 (m, 1H).
实施例35和实施例36:N-((R)-1-(2,4-二氯苯基)乙基)-5-(3-(R)-1-((1s,3S)-3-甲氧基环丁基)哌啶-3-基)氮杂环丁烷-1-基)-[1,2,4]三唑并[1,5-a]嘧啶-7-胺和N- ((R)-1-(2,4-二氯苯基)乙基)-5-(3-(R)-2-((1r,3R)-3-甲氧基环丁基)哌啶-3-基)氮杂环丁烷-1-基)-[1,2,4]三唑并[1,5-a]嘧啶-7-胺(35和36)
Example 35 and Example 36: N-((R)-1-(2,4-dichlorophenyl)ethyl)-5-(3-(R)-1-((1s,3S)-3 -Methoxycyclobutyl)piperidin-3-yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine and N - ((R)-1-(2,4-dichlorophenyl)ethyl)-5-(3-(R)-2-((1r,3R)-3-methoxycyclobutyl)piperidine -3-yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (35 and 36)
室温下,将化合物25-2(150mg,0.34mmol)和3-甲氧基环丁烷-1-酮(34mg,0.34mmol)溶于甲醇(2mL),加入醋酸(2mg,0.03mmol),反应液室温搅拌20分钟,随后加入氰基硼氢化钠(32mg,0.50mmol),室温继续搅拌1小时。反应完毕后,反应液减压浓缩。所得残余物经硅胶制备薄层色谱(乙酸乙酯/甲醇=5/1)分离,得到化合物35-1。化合物35-1(70mg,0.13mmol)经超临界流体色谱拆分(色谱柱:DAICEL CHIRALCEL OJ(250mm*30mm*10μm);流动相:超临界二氧化碳,乙醇(0.1%一水合氨);梯度配比:甲醇相25%;流速:55mL/min;柱温:室温),得到化合物35和化合物36。(化合物35为第一个洗脱的峰,化合物36为第二个洗脱的峰)。化合物35:MS-ESI:m/z 530.2[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.54(d,J=2.4Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.8,2.4Hz,1H),5.18-5.10(m,1H),4.82(s,1H),4.09(t,J=8.8Hz,1H),4.04-3.95(m,1H),3.94-3.89(m,1H),3.78(dd,J=8.0,6.4Hz,1H),3.73-3.58(m,1H),3.24(s,3H),2.98-2.76(m,3H),2.54-2.41(m,1H),2.24-2.01(m,4H),1.82-1.70(m,4H),1.65(d,J=6.8Hz,3H),1.60-1.51(m,1H),1.46-1.39(m,1H),0.96-0.81(m,1H)。化合物36:MS-ESI:m/z 530.2[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.54(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.18-5.11(m,1H),4.81(s,1H),4.09(t,J=8.8Hz,1H),4.05-3.90(m,1H),3.80(dd,J=8.8,5.6Hz,1H),3.74-3.56(m,2H),3.24(s,3H),2.89-2.76(m,2H),2.54-2.41(m,3H),2.39-2.29(m,1H),1.84-1.69(m,6H),1.65(d,J=6.8Hz,3H),1.61-1.51(m,1H),1.46(t,J=10.8Hz,1H),0.96-0.81(m,1H)。At room temperature, compound 25-2 (150mg, 0.34mmol) and 3-methoxycyclobutane-1-one (34mg, 0.34mmol) were dissolved in methanol (2mL), acetic acid (2mg, 0.03mmol) was added, and the reaction The solution was stirred at room temperature for 20 minutes, then sodium cyanoborohydride (32 mg, 0.50 mmol) was added, and stirring was continued at room temperature for 1 hour. After the reaction was completed, the reaction solution was concentrated under reduced pressure. The obtained residue was separated by preparative thin-layer chromatography on silica gel (ethyl acetate/methanol=5/1) to obtain compound 35-1. Compound 35-1 (70mg, 0.13mmol) was resolved by supercritical fluid chromatography (column: DAICEL CHIRALCEL OJ (250mm*30mm*10μm); mobile phase: supercritical carbon dioxide, ethanol (0.1% ammonia monohydrate); gradient Ratio: methanol phase 25%; flow rate: 55mL/min; column temperature: room temperature), compound 35 and compound 36 were obtained. (Compound 35 was the first eluting peak, compound 36 was the second eluting peak). Compound 35: MS-ESI: m/z 530.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.54(d, J=2.4Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.8 ,2.4Hz,1H),5.18-5.10(m,1H),4.82(s,1H),4.09(t,J=8.8Hz,1H),4.04-3.95(m,1H),3.94-3.89(m, 1H), 3.78(dd, J=8.0, 6.4Hz, 1H), 3.73-3.58(m, 1H), 3.24(s, 3H), 2.98-2.76(m, 3H), 2.54-2.41(m, 1H) ,2.24-2.01(m,4H),1.82-1.70(m,4H),1.65(d,J=6.8Hz,3H),1.60-1.51(m,1H),1.46-1.39(m,1H),0.96 -0.81(m,1H). Compound 36: MS-ESI: m/z 530.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.54(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz,1H),5.18-5.11(m,1H),4.81(s,1H),4.09(t,J=8.8Hz,1H),4.05-3.90(m,1H),3.80(dd,J= 8.8,5.6Hz,1H),3.74-3.56(m,2H),3.24(s,3H),2.89-2.76(m,2H),2.54-2.41(m,3H),2.39-2.29(m,1H) ,1.84-1.69(m,6H),1.65(d,J=6.8Hz,3H),1.61-1.51(m,1H),1.46(t,J=10.8Hz,1H),0.96-0.81(m,1H ).
实施例37和实施例38:N-((R)-1-(2,4-二氯苯基)乙基)-5-(3-(R)-1-((1s,3S)-3-(甲基磺酰基)环丁基)哌啶-3-基)氮杂环丁烷-1-基)-[1,2,4]三唑并[1,5-a]嘧啶-7-胺和N-((R)-1-(2,4-二氯苯基)乙基)-5-(3-(R)-1-((1r,3R)-3-(甲基磺酰基)环丁基)哌啶-3-基)氮杂环丁烷-1-基)-[1,2,4]三唑并[1,5-a]嘧啶-7-胺(37和38)
Example 37 and Example 38: N-((R)-1-(2,4-dichlorophenyl)ethyl)-5-(3-(R)-1-((1s,3S)-3 -(methylsulfonyl)cyclobutyl)piperidin-3-yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidine-7- Amine and N-((R)-1-(2,4-dichlorophenyl)ethyl)-5-(3-(R)-1-((1r,3R)-3-(methylsulfonyl )cyclobutyl)piperidin-3-yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (37 and 38)
1)化合物37-2的合成1) Synthesis of Compound 37-2
冰浴下,将化合物37-1(5.00g,28.37mmol)溶于甲醇(20mL),分三批次加入硼氢化钠(1.14g,30.14mmol),反应混合物在0℃搅拌1小时。反应完毕后,加水(20mL)淬灭,减压浓缩。残余物加乙酸乙酯(200mL)稀释,用水(30mL×3)洗涤。有机相经无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物37-2。1H NMR(400MHz,CDCl3)δ7.38-7.32(m,4H),7.31-7.27(m,1H),4.43(s,2H),3.91-3.86(m,1H),3.68-3.60(m,1H),2.78-2.67(m,2H),1.98-1.90(m,2H)。Under ice bath, compound 37-1 (5.00 g, 28.37 mmol) was dissolved in methanol (20 mL), sodium borohydride (1.14 g, 30.14 mmol) was added in three batches, and the reaction mixture was stirred at 0°C for 1 hour. After the reaction was completed, it was quenched by adding water (20 mL), and concentrated under reduced pressure. The residue was diluted with ethyl acetate (200 mL), washed with water (30 mL×3). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound 37-2. 1 H NMR (400MHz, CDCl 3 )δ7.38-7.32(m,4H),7.31-7.27(m,1H),4.43(s,2H),3.91-3.86(m,1H),3.68-3.60(m ,1H), 2.78-2.67(m,2H), 1.98-1.90(m,2H).
2)化合物37-3的合成2) Synthesis of compound 37-3
冰浴下,将化合物37-2(4.50g,25.25mmol),甲基磺酸酐(6.60g,37.87mmol)和三乙胺(10.50mL,75.75mmol)溶于二氯甲烷(50mL)中,反应混合物在室温搅拌4小时。反应完毕后,加二氯甲烷(100mL)稀释,用水(50mL×3)洗涤。有机相经无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物37-3。1H NMR(400MHz,CDCl3)δ7.37-7.31(m,5H),4.69-4.61(m,1H),4.44(s,2H),3.78-3.71(m,1H),2.98(s,3H),2.89-2.79(m,2H),2.40-2.28(m,2H)。Under ice bath, compound 37-2 (4.50g, 25.25mmol), methanesulfonic anhydride (6.60g, 37.87mmol) and triethylamine (10.50mL, 75.75mmol) were dissolved in dichloromethane (50mL), and the reaction The mixture was stirred at room temperature for 4 hours. After the reaction was completed, it was diluted with dichloromethane (100 mL), and washed with water (50 mL×3). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound 37-3. 1 H NMR (400MHz, CDCl 3 )δ7.37-7.31(m,5H),4.69-4.61(m,1H),4.44(s,2H),3.78-3.71(m,1H),2.98(s,3H ), 2.89-2.79(m,2H), 2.40-2.28(m,2H).
3)化合物37-4的合成3) Synthesis of compound 37-4
室温下,将化合物37-3(6.20g,24.19mmol)和甲硫醇钠(3.39g,48.38mmol)溶于N,N-二甲基甲酰胺(65mL)中,反应混合物在室温搅拌16小时。反应完毕后,加水(100mL)稀释,用乙酸乙酯(100mL×3)萃取。合并有机相,饱和食盐水(100mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=5/1)分离,得到化合物37-4。1H NMR(400MHz,CDCl3)δ7.36-7.33(m,4H),7.32-7.27(m,1H),4.42(s,2H),4.38-4.30(m,1H),3.43-3.35(m,1H),2.47-2.39(m,2H),2.29-2.22(m,2H),2.06(s,3H)。At room temperature, compound 37-3 (6.20g, 24.19mmol) and sodium thiomethoxide (3.39g, 48.38mmol) were dissolved in N,N-dimethylformamide (65mL), and the reaction mixture was stirred at room temperature for 16 hours . After the reaction was completed, it was diluted with water (100 mL), and extracted with ethyl acetate (100 mL×3). The organic phases were combined, washed with saturated brine (100 mL×3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to obtain compound 37-4. 1 H NMR (400MHz, CDCl 3 ) δ7.36-7.33(m, 4H), 7.32-7.27(m, 1H), 4.42(s, 2H), 4.38-4.30(m, 1H), 3.43-3.35(m ,1H), 2.47-2.39(m,2H), 2.29-2.22(m,2H), 2.06(s,3H).
4)化合物37-5的合成4) Synthesis of compound 37-5
室温下,将化合物37-4(1.00g,4.80mmol)溶于二氯甲烷(10mL),再加入间氯过氧苯甲酸(2.44g,12.00mmol,85%纯度),反应混合物在室温搅拌2小时。反应液过滤,滤液经硅胶柱层析(石油醚/乙酸乙酯=3/1)分离,得到化合物37-5。1H NMR(400MHz,CDCl3)δ7.38-7.31(m,5H),4.45(s,2H),4.42-4.36(m,1H),3.74-3.62(m,1H),2.84(s,3H),2.82-2.74(m,2H),2.51-2.42(m,2H)。 At room temperature, compound 37-4 (1.00g, 4.80mmol) was dissolved in dichloromethane (10mL), then m-chloroperoxybenzoic acid (2.44g, 12.00mmol, 85% purity) was added, and the reaction mixture was stirred at room temperature for 2 Hour. The reaction solution was filtered, and the filtrate was separated by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to obtain compound 37-5. 1 H NMR (400MHz, CDCl 3 )δ7.38-7.31(m,5H),4.45(s,2H),4.42-4.36(m,1H),3.74-3.62(m,1H),2.84(s,3H ), 2.82-2.74(m,2H), 2.51-2.42(m,2H).
5)化合物37-6的合成5) Synthesis of compound 37-6
室温下,将化合物37-5(1.00g,4.16mmol)溶于甲醇(20mL)中,氮气保护下加入湿钯碳(100mg,0.94mmol,10%纯度)和湿氢氧化钯(100mg,0.71mmol),氮气置换,氢气置换,反应液在氢气氛围(50psi)下,在30℃搅拌16小时。反应完毕后,反应液经硅藻土过滤,用甲醇(20mL×3)洗涤,滤液减压浓缩,得到化合物37-6。1H NMR(400MHz,CDCl3)δ4.73-4.61(m,1H),3.75-3.63(m,1H),2.90-2.80(m,5H),2.47-2.36(m,2H)。At room temperature, compound 37-5 (1.00g, 4.16mmol) was dissolved in methanol (20mL), and wet palladium on carbon (100mg, 0.94mmol, 10% purity) and wet palladium hydroxide (100mg, 0.71mmol) were added under nitrogen protection ), nitrogen replacement, hydrogen replacement, and the reaction solution was stirred at 30° C. for 16 hours under a hydrogen atmosphere (50 psi). After the reaction was completed, the reaction solution was filtered through diatomaceous earth, washed with methanol (20 mL×3), and the filtrate was concentrated under reduced pressure to obtain compound 37-6. 1 H NMR (400 MHz, CDCl 3 ) δ 4.73-4.61 (m, 1H), 3.75-3.63 (m, 1H), 2.90-2.80 (m, 5H), 2.47-2.36 (m, 2H).
6)化合物37-7的合成6) Synthesis of compound 37-7
室温下,将化合物37-6(510mg,3.40mmol)溶于二氯甲烷(10mL)中,0℃下加入戴斯-马丁氧化剂(4.32g,10.19mmol),反应混合物在室温搅拌16小时。反应完毕后,反应混合物经硅藻土过滤,滤饼用二氯甲烷(20mL×2)洗涤,滤液减压浓缩。所得残余物用乙酸乙酯(10mL)打浆,过滤,滤液减压浓缩,得到化合物37-7。1H NMR(400MHz,CDCl3)δ3.94-3.86(m,1H),3.74-3.63(m,2H),3.49-3.37(m,2H),2.99(s,3H)。Compound 37-6 (510 mg, 3.40 mmol) was dissolved in dichloromethane (10 mL) at room temperature, Dess-Martin oxidant (4.32 g, 10.19 mmol) was added at 0°C, and the reaction mixture was stirred at room temperature for 16 hours. After the reaction was completed, the reaction mixture was filtered through diatomaceous earth, the filter cake was washed with dichloromethane (20 mL×2), and the filtrate was concentrated under reduced pressure. The obtained residue was slurried with ethyl acetate (10 mL), filtered, and the filtrate was concentrated under reduced pressure to obtain compound 37-7. 1 H NMR (400 MHz, CDCl 3 ) δ 3.94-3.86 (m, 1H), 3.74-3.63 (m, 2H), 3.49-3.37 (m, 2H), 2.99 (s, 3H).
7)化合物37和化合物38的合成7) Synthesis of compound 37 and compound 38
室温下,将化合物25-2(200mg,0.45mmol),化合物37-7(266mg,0.90mmol,50%纯度)和醋酸(3μL,0.04mmol)溶于甲醇(5mL)中,反应混合物在室温搅拌30分钟,加入氰基硼氢化钠(84mg,1.34mmol),反应混合物室温搅拌15.5小时。反应完毕后,反应液减压浓缩,所得残余物经硅胶制备薄层色谱(纯甲醇)分离,得到化合物37-8。化合物37-8再经超临界流体色谱拆分(色谱柱:DAICEL CHIRALPAK AS(250mm×30mm*10μm);流动相:超临界二氧化碳,乙醇(0.1%一水合氨);梯度配比:乙醇相40%;流速:70mL/min;柱温:室温),得到化合物37和化合物38。(化合物37为第一个洗脱的峰,化合物38为第二个洗脱的峰)。化合物37:MS-ESI:m/z 578.4[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.53(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.17-5.09(m,1H),4.81(s,1H),4.08(t,J=8.4Hz,1H),4.04-3.88(m,1H),3.82-3.75(m,1H),3.74-3.53(m,2H),2.86(s,3H),2.85-2.73(m,3H),2.54-2.37(m,3H),2.36-2.24(m,2H),1.88-1.80(m,1H),1.79-1.68(m,3H),1.65(d,J=6.8Hz,3H),1.61-1.48(m,2H),0.96-0.82(m,1H)。化合物38:MS-ESI:m/z 578.4[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.53(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.17-5.10(m,1H),4.81(s,1H),4.09(t,J=8.4Hz,1H),4.05-3.91(m,1H),3.82-3.76(m,1H),3.75-3.55(m,2H),2.94-2.78(m,6H),2.54-2.40(m,3H),2.39-2.26(m,2H),1.97-1.87(m,1H),1.84-1.69(m,3H),1.65(d,J=6.8Hz,3H),1.63-1.50(m,2H),0.99-0.83(m,1H)。At room temperature, compound 25-2 (200 mg, 0.45 mmol), compound 37-7 (266 mg, 0.90 mmol, 50% purity) and acetic acid (3 μL, 0.04 mmol) were dissolved in methanol (5 mL), and the reaction mixture was stirred at room temperature After 30 minutes, sodium cyanoborohydride (84 mg, 1.34 mmol) was added, and the reaction mixture was stirred at room temperature for 15.5 hours. After the reaction was completed, the reaction solution was concentrated under reduced pressure, and the obtained residue was separated by silica gel preparative thin-layer chromatography (pure methanol) to obtain compound 37-8. Compound 37-8 was separated by supercritical fluid chromatography (column: DAICEL CHIRALPAK AS (250mm×30mm*10μm); mobile phase: supercritical carbon dioxide, ethanol (0.1% ammonia monohydrate); gradient ratio: ethanol phase 40 %; flow rate: 70mL/min; column temperature: room temperature), compound 37 and compound 38 were obtained. (Compound 37 was the first eluting peak, compound 38 was the second eluting peak). Compound 37: MS-ESI: m/z 578.4 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.53(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz,1H),5.17-5.09(m,1H),4.81(s,1H),4.08(t,J=8.4Hz,1H),4.04-3.88(m,1H),3.82-3.75(m, 1H),3.74-3.53(m,2H),2.86(s,3H),2.85-2.73(m,3H),2.54-2.37(m,3H),2.36-2.24(m,2H),1.88-1.80( m, 1H), 1.79-1.68 (m, 3H), 1.65 (d, J=6.8Hz, 3H), 1.61-1.48 (m, 2H), 0.96-0.82 (m, 1H). Compound 38: MS-ESI: m/z 578.4 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.53(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz,1H),5.17-5.10(m,1H),4.81(s,1H),4.09(t,J=8.4Hz,1H),4.05-3.91(m,1H),3.82-3.76(m, 1H),3.75-3.55(m,2H),2.94-2.78(m,6H),2.54-2.40(m,3H),2.39-2.26(m,2H),1.97-1.87(m,1H),1.84- 1.69 (m, 3H), 1.65 (d, J=6.8Hz, 3H), 1.63-1.50 (m, 2H), 0.99-0.83 (m, 1H).
实施例39和实施例40:(R)-1-((R)-3-(1-(7-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)丙-2-醇和(S)-1-((R)-3- (1-(7-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)丙-2-醇(39和40)
Example 39 and Example 40: (R)-1-((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino) -[1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)propan-2-ol and (S)- 1-((R)-3- (1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4]triazolo[1,5-a]pyrimidine-5 -yl)azetidin-3-yl)piperidin-1-yl)propan-2-ol (39 and 40)
室温下,将化合物25-2(200mg,0.44mmol)和1-溴丙-2-醇(92mg,0.66mmol)溶于乙腈(10mL)中,加入二异丙基乙胺(172mg,1.33mmol),反应液在80℃搅拌16小时。反应液冷却至室温,减压浓缩。所得残余物经硅胶制备薄层色谱(乙酸乙酯/甲醇=3/1)分离,得到化合物39-1。化合物39-1(140mg,0.28mmol)经超临界流体色谱拆分(色谱柱:DAICEL CHIRALCEL OJ(250mm*30mm*10μm);流动相:超临界二氧化碳,乙醇(0.1%一水合氨);梯度配比:乙醇相25%;流速:60mL/min;柱温:室温),得到化合物39和化合物40。(化合物39为第一个洗脱的峰,化合物40为第二个洗脱的峰)。化合物39:MS-ESI:m/z 504.4[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.54(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.17-5.10(m,1H),4.81(s,1H),4.10(t,J=8.0Hz,1H),4.07-3.92(m,2H),3.79(dd,J=8.4,6.4Hz,1H),3.76-3.61(m,1H),3.12-2.96(m,2H),2.57-2.40(m,3H),2.37-2.23(m,1H),1.97-1.74(m,4H),1.69-1.60(m,4H),1.16(d,J=6.4Hz,3H),1.04-0.89(m,1H)。化合物40:MS-ESI:m/z 504.3[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.54(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.4Hz,1H),5.19-5.11(m,1H),4.81(s,1H),4.14-4.05(m,1H),4.01-3.87(m,2H),3.82-3.75(m,1H),3.74-3.57(m,1H),3.00-2.81(m,2H),2.56-2.45(m,1H),2.41-2.26(m,2H),2.06-1.94(m,1H),1.85-1.69(m,4H),1.65(d,J=6.8Hz,3H),1.63-1.50(m,1H),1.14(d,J=6.0Hz,3H),0.99-0.82(m,1H)。At room temperature, compound 25-2 (200mg, 0.44mmol) and 1-bromopropan-2-ol (92mg, 0.66mmol) were dissolved in acetonitrile (10mL), and diisopropylethylamine (172mg, 1.33mmol) was added , and the reaction solution was stirred at 80° C. for 16 hours. The reaction solution was cooled to room temperature and concentrated under reduced pressure. The obtained residue was separated by preparative thin-layer chromatography on silica gel (ethyl acetate/methanol=3/1) to obtain compound 39-1. Compound 39-1 (140mg, 0.28mmol) was resolved by supercritical fluid chromatography (column: DAICEL CHIRALCEL OJ (250mm*30mm*10μm); mobile phase: supercritical carbon dioxide, ethanol (0.1% ammonia monohydrate); gradient Ratio: ethanol phase 25%; flow rate: 60mL/min; column temperature: room temperature), compound 39 and compound 40 were obtained. (Compound 39 was the first eluting peak, compound 40 was the second eluting peak). Compound 39: MS-ESI: m/z 504.4 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.54(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz,1H),5.17-5.10(m,1H),4.81(s,1H),4.10(t,J=8.0Hz,1H),4.07-3.92(m,2H),3.79(dd,J= 8.4,6.4Hz,1H),3.76-3.61(m,1H),3.12-2.96(m,2H),2.57-2.40(m,3H),2.37-2.23(m,1H),1.97-1.74(m, 4H), 1.69-1.60 (m, 4H), 1.16 (d, J=6.4Hz, 3H), 1.04-0.89 (m, 1H). Compound 40: MS-ESI: m/z 504.3 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.54(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.4Hz,1H),5.19-5.11(m,1H),4.81(s,1H),4.14-4.05(m,1H),4.01-3.87(m,2H),3.82-3.75(m,1H), 3.74-3.57(m,1H),3.00-2.81(m,2H),2.56-2.45(m,1H),2.41-2.26(m,2H),2.06-1.94(m,1H),1.85-1.69(m , 4H), 1.65 (d, J=6.8Hz, 3H), 1.63-1.50 (m, 1H), 1.14 (d, J=6.0Hz, 3H), 0.99-0.82 (m, 1H).
实施例41:1-(((R)-3-(1-(7-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)甲基)环丙烷-1-醇(41)
Example 41: 1-(((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4 ]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)methyl)cyclopropan-1-ol (41)
1)化合物41-2的合成1) Synthesis of Compound 41-2
室温下,将化合物41-1(1.80g,13.83mmol)溶于N,N-二甲基甲酰胺(30mL),温度降至0℃,分批加入钠氢(830mg,20.75mmol,60%纯度),反应液在0℃搅拌20分钟,加入对甲氧基氯苄(2.38g,15.21mmol),室温搅拌3小时。反应完毕后,向反应液中加入饱和氯化铵水溶液(50mL)淬灭,乙酸乙酯(50mL×2)萃取。合并有机相,经饱和食盐水(30mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。粗品经硅胶柱层析(乙酸乙酯/石油醚=5/1)分离,得到化合物41-2。1H NMR(400MHz,CDCl3)δ7.30(d,J=8.4Hz,2H),6.88(d,J=8.8Hz,2H),4.59(s,2H),4.24(q,J=6.8Hz,2H),3.80(s,3H),1.35-1.33(m,2H),1.32-1.26(m,3H),1.25-1.20(m,2H)。At room temperature, compound 41-1 (1.80g, 13.83mmol) was dissolved in N,N-dimethylformamide (30mL), the temperature was lowered to 0°C, and sodium hydrogen (830mg, 20.75mmol, 60% purity ), the reaction solution was stirred at 0°C for 20 minutes, p-methoxybenzyl chloride (2.38 g, 15.21 mmol) was added, and stirred at room temperature for 3 hours. After the reaction was completed, saturated ammonium chloride aqueous solution (50 mL) was added to the reaction liquid to quench, and ethyl acetate (50 mL×2) was extracted. The organic phases were combined, washed with saturated brine (30 mL×3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was separated by silica gel column chromatography (ethyl acetate/petroleum ether=5/1) to obtain compound 41-2. 1 H NMR (400MHz, CDCl 3 ) δ7.30(d, J=8.4Hz, 2H), 6.88(d, J=8.8Hz, 2H), 4.59(s, 2H), 4.24(q, J=6.8Hz ,2H), 3.80(s,3H), 1.35-1.33(m,2H), 1.32-1.26(m,3H), 1.25-1.20(m,2H).
2)化合物41-3的合成2) Synthesis of Compound 41-3
室温下,将化合物41-2(1.00g,4.00mmol)溶于四氢呋喃(30mL),冷却至0℃,缓慢滴加2.5mol/L氢化铝锂四氢呋喃溶液(2.40mL,6.00mmol),0℃搅拌0.5小时。反应完毕后,在0℃下缓慢滴加饱和酒石酸钾钠水溶液(50mL)淬灭,乙酸乙酯(50mL×3)萃取。合并有机相,用饱和食盐水(50mL)洗涤,过滤,滤液减压浓缩,得到化合物41-3。1H NMR(400MHz,CDCl3)δ7.25(d,J=8.4Hz,2H),6.88(d,J=8.8Hz,2H),4.54(s,2H),3.81(s,3H),3.71(s,2H),0.99-0.94(m,2H),0.66-0.61(m,2H)。Dissolve compound 41-2 (1.00g, 4.00mmol) in tetrahydrofuran (30mL) at room temperature, cool to 0°C, slowly add 2.5mol/L lithium aluminum hydride tetrahydrofuran solution (2.40mL, 6.00mmol) dropwise, and stir at 0°C 0.5 hours. After the reaction was completed, it was quenched by slowly adding saturated potassium sodium tartrate aqueous solution (50 mL) dropwise at 0° C., and extracted with ethyl acetate (50 mL×3). The organic phases were combined, washed with saturated brine (50 mL), filtered, and the filtrate was concentrated under reduced pressure to obtain compound 41-3. 1 H NMR (400MHz, CDCl 3 ) δ7.25(d, J=8.4Hz, 2H), 6.88(d, J=8.8Hz, 2H), 4.54(s, 2H), 3.81(s, 3H), 3.71 (s, 2H), 0.99-0.94 (m, 2H), 0.66-0.61 (m, 2H).
3)化合物41-4的合成3) Synthesis of compound 41-4
室温下,将化合物41-3(150mg,0.72mmol)和吡啶(342mg,4.32mmol)溶于二氯甲烷(6mL)中,冰水浴降温至0℃,加入甲基磺酸酐(126mg,0.72mmol),反应液在室温搅拌1小时。反应完毕后,反应液加二氯甲烷(50mL)稀释,用水(10mL×3)洗涤,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物41-4。粗品未经纯化,直接用于下一步反应。1H NMR(400MHz,CDCl3)δ7.24(d,J=8.4Hz,2H),6.87(d,J=8.8Hz,2H),4.56(s,2H),4.40(s,2H),3.81(s,3H),3.07(s,3H),1.11-1.06(m,2H),0.83-0.78(m,2H)。At room temperature, compound 41-3 (150mg, 0.72mmol) and pyridine (342mg, 4.32mmol) were dissolved in dichloromethane (6mL), cooled to 0°C in an ice-water bath, and methanesulfonic anhydride (126mg, 0.72mmol) was added , and the reaction solution was stirred at room temperature for 1 hour. After the reaction was completed, the reaction solution was diluted with dichloromethane (50 mL), washed with water (10 mL×3), the organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound 41-4. The crude product was directly used in the next reaction without further purification. 1 H NMR (400MHz, CDCl 3 ) δ7.24(d, J=8.4Hz, 2H), 6.87(d, J=8.8Hz, 2H), 4.56(s, 2H), 4.40(s, 2H), 3.81 (s,3H), 3.07(s,3H), 1.11-1.06(m,2H), 0.83-0.78(m,2H).
4)化合物41-5的合成4) Synthesis of Compound 41-5
室温下,将化合物25-2(130mg,0.29mmol)和化合物41-4(167mg,0.58mmol)溶于乙腈(5mL)中,随后加入N,N-二异丙基乙胺(188mg,1.46mmol),反应液在80℃搅拌14小时。反应完毕后,反应液减压浓缩,剩余物经硅胶制备薄层色谱(乙酸乙酯/甲醇=1/1)分离,得到化合物41-5。MS-ESI:m/z 636.4[M+H]+At room temperature, compound 25-2 (130mg, 0.29mmol) and compound 41-4 (167mg, 0.58mmol) were dissolved in acetonitrile (5mL), then N,N-diisopropylethylamine (188mg, 1.46mmol ), and the reaction solution was stirred at 80° C. for 14 hours. After the reaction was completed, the reaction liquid was concentrated under reduced pressure, and the residue was separated by silica gel preparative thin-layer chromatography (ethyl acetate/methanol=1/1) to obtain compound 41-5. MS-ESI: m/z 636.4 [M+H] + .
5)化合物41的合成5) Synthesis of Compound 41
室温下,将化合物41-5(50mg,0.05mmol,66%纯度)溶于二氯甲烷(1mL),随后加入三氟乙酸(1.00mL),反应液在室温搅拌3小时。反应完毕后,反应液减压浓缩,加少量甲醇稀释,用氨水调节pH~9,剩余物经硅胶制备薄层色谱(纯甲醇)分离,得到化合物41。MS-ESI:m/z 516.2[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.54(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz 1H),5.17-5.10(m,1H),4.81(s,1H),4.10(t,J=8.4Hz,1H),4.05-3.93(m,1H),3.81(dd,J=8.4,6.0Hz 1H),3.74-3.61(m,1H),3.15-2.98(m,2H),2.59-2.43(m,3H),2.14-2.04(m,1H),1.85-1.68(m,4H),1.65(d,J=6.8Hz,3H),1.63-1.56(m,1H),0.96-0.87(m,1H),0.75-0.68(m,2H),0.54-0.46(m,2H)。Compound 41-5 (50 mg, 0.05 mmol, 66% purity) was dissolved in dichloromethane (1 mL) at room temperature, then trifluoroacetic acid (1.00 mL) was added, and the reaction solution was stirred at room temperature for 3 hours. After the reaction was completed, the reaction solution was concentrated under reduced pressure, diluted with a small amount of methanol, adjusted to pH ~ 9 with ammonia water, and the residue was separated by preparative silica gel thin-layer chromatography (pure methanol) to obtain compound 41. MS-ESI: m/z 516.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.54(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz 1H), 5.17-5.10(m, 1H), 4.81(s, 1H), 4.10(t, J=8.4Hz, 1H), 4.05-3.93(m, 1H), 3.81(dd, J=8.4, 6.0Hz 1H),3.74-3.61(m,1H),3.15-2.98(m,2H),2.59-2.43(m,3H),2.14-2.04(m,1H),1.85-1.68(m,4H),1.65( d, J=6.8Hz, 3H), 1.63-1.56(m, 1H), 0.96-0.87(m, 1H), 0.75-0.68(m, 2H), 0.54-0.46(m, 2H).
实施例42:N-((R)-1-(2,4-二氯苯基)乙基)-5-(3-(R)-2-(氧杂环丁烷-3-基甲基)哌啶-3-基)氮杂环丁烷-1-基)-[1,2,4]三唑并[1,5-a]嘧啶-7-胺(42)
Example 42: N-((R)-1-(2,4-dichlorophenyl)ethyl)-5-(3-(R)-2-(oxetan-3-ylmethyl) )piperidin-3-yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (42)
室温下,将化合物25-2(150mg,0.34mmol)溶于乙腈(5mL)中,随后加入3-溴甲基氧杂环丁烷(101mg,0.67mmol)和碳酸钾(139mg,1.01mmol),反应液在80℃搅拌14小时。反应完毕后,反应液减压浓缩。剩余物经硅胶制备薄层色谱(纯甲醇)分离,得到化合物42。MS-ESI:m/z 516.4[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.54(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz 1H),5.17-5.10(m,1H),4.82-4.77(m,3H),4.41(t,J=6.0Hz,2H),4.08(t,J=8.4Hz,1H),4.04-3.90(m,1H),3.77(dd,J=8.4,6.0Hz 1H),3.72-3.56(m,1H),3.29-3.26(m,1H),2.80-2.67(m,4H),2.53-2.41(m,1H),2.02-1.91(m,1H),1.80-1.66(m,4H),1.65(d,J=6.8Hz,3H),1.60-1.50(m,1H),0.94-0.83(m,1H)。Compound 25-2 (150 mg, 0.34 mmol) was dissolved in acetonitrile (5 mL) at room temperature, and then 3-bromomethyloxetane (101 mg, 0.67 mmol) and potassium carbonate (139 mg, 1.01 mmol) were added, The reaction solution was stirred at 80°C for 14 hours. After the reaction was completed, the reaction solution was concentrated under reduced pressure. The residue was separated by preparative thin-layer chromatography on silica gel (pure methanol) to obtain compound 42. MS-ESI: m/z 516.4 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.54(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz 1H),5.17-5.10(m,1H),4.82-4.77(m,3H),4.41(t,J=6.0Hz,2H),4.08(t,J=8.4Hz,1H),4.04- 3.90(m,1H),3.77(dd,J=8.4,6.0Hz 1H),3.72-3.56(m,1H),3.29-3.26(m,1H),2.80-2.67(m,4H),2.53-2.41 (m,1H),2.02-1.91(m,1H),1.80-1.66(m,4H),1.65(d,J=6.8Hz,3H),1.60-1.50(m,1H),0.94-0.83(m ,1H).
实施例43:3-(((R)-3-(1-(7-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)甲基)硫杂环丁烷-1,1-二氧化物(43)
Example 43: 3-(((R)-3-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4 ]triazol[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)methyl)thietane-1,1-dioxide ( 43)
1)化合物43-2的合成1) Synthesis of compound 43-2
室温下,将化合物43-1(25.00g,211.63mmol)和四氢吡咯(30.10g,423.26mmol)溶于甲基叔丁基醚(300mL)中,随后加入硫酸镁(6.00g,49.85mmol),反应液在室温搅拌16小时。反应完毕后,低温减压浓缩掉大部分溶剂。剩余物经减 压蒸馏(60℃-64℃,0.03帕),得到化合物43-2。1H NMR(400MHz,CDCl3)δ4.68(s,1H),3.88(s,1H),3.60(s,1H),3.35(s,6H),3.19-3.09(m,4H),1.87-1.78(m,4H)。At room temperature, compound 43-1 (25.00 g, 211.63 mmol) and tetrahydropyrrole (30.10 g, 423.26 mmol) were dissolved in methyl tert-butyl ether (300 mL), then magnesium sulfate (6.00 g, 49.85 mmol) was added , and the reaction solution was stirred at room temperature for 16 hours. After the reaction was completed, most of the solvent was concentrated under reduced pressure at low temperature. The remainder is reduced Pressure distillation (60°C-64°C, 0.03 Pa) gave compound 43-2. 1 H NMR (400MHz, CDCl 3 )δ4.68(s,1H),3.88(s,1H),3.60(s,1H),3.35(s,6H),3.19-3.09(m,4H),1.87- 1.78 (m, 4H).
2)化合物43-3的合成2) Synthesis of compound 43-3
室温下,将化合物43-2(5.00g,29.20mmol)和三乙胺(3.84g,37.96mmol)溶于甲基叔丁基醚(25mL)中,降温至0℃,随后滴加甲基磺酰氯(4.10g,35.80mmol)的甲基叔丁基醚(25mL)溶液,控制内温不超过5℃,滴加完毕后,反应液室温搅拌1小时。反应完毕后,将反应液倒入饱和碳酸氢钠水溶液(50ml)中淬灭,用二氯甲烷(50ml×3)萃取。合并有机相,经无水硫酸钠干燥,过滤,滤液在40℃减压浓缩,得到化合物43-3。粗品不经纯化,直接用于下一步反应。1H NMR(400MHz,CDCl3)δ4.45(s,1H),4.28-4.25(m,1H),4.24-4.21(m,1H),4.07-4.05(m,1H),4.04-4.01(m,1H),3.54(s,6H),2.84-2.71(m,4H),1.84-1.75(m,4H)。At room temperature, compound 43-2 (5.00g, 29.20mmol) and triethylamine (3.84g, 37.96mmol) were dissolved in methyl tert-butyl ether (25mL), cooled to 0°C, and then methylsulfonate was added dropwise Acyl chloride (4.10g, 35.80mmol) in methyl tert-butyl ether (25mL) solution, the internal temperature was controlled not to exceed 5°C, after the dropwise addition, the reaction solution was stirred at room temperature for 1 hour. After the reaction was completed, the reaction solution was poured into saturated aqueous sodium bicarbonate solution (50ml) to quench, and extracted with dichloromethane (50ml×3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure at 40° C. to obtain compound 43-3. The crude product was directly used in the next reaction without purification. 1 H NMR (400MHz, CDCl 3 )δ4.45(s,1H),4.28-4.25(m,1H),4.24-4.21(m,1H),4.07-4.05(m,1H),4.04-4.01(m ,1H), 3.54(s,6H), 2.84-2.71(m,4H), 1.84-1.75(m,4H).
3)化合物43-4的合成3) Synthesis of Compound 43-4
室温下,将化合物43-3(3.00g,12.03mmol)溶于二氯甲烷(20mL)中,降温至0℃,加入三氟甲磺酸甲酯(2.17g,13.24mmol),控制内温不超过5℃,滴加完毕后,反应液室温反应14小时。然后0℃缓慢滴加三乙胺(1.34g,13.24mmol),滴加完毕后,反应液回流搅拌1小时。反应完毕后,反应液冷却到室温,依次用水(20mL),1mol/L的盐酸水溶液(10mL)和饱和碳酸钠水溶液(10mL)洗涤。有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物43-4。粗品不经纯化,直接用于下一步反应。1H NMR(400MHz,CDCl3)δ6.76(d,J=1.2Hz,1H),5.16(d,J=0.8Hz,1H),4.46(s,2H),3.38(s,6H)。At room temperature, compound 43-3 (3.00g, 12.03mmol) was dissolved in dichloromethane (20mL), cooled to 0°C, and methyl trifluoromethanesulfonate (2.17g, 13.24mmol) was added to control the internal temperature. When the temperature exceeds 5°C, after the dropwise addition is completed, the reaction solution is reacted at room temperature for 14 hours. Then triethylamine (1.34 g, 13.24 mmol) was slowly added dropwise at 0° C. After the dropwise addition, the reaction solution was refluxed and stirred for 1 hour. After the reaction was completed, the reaction solution was cooled to room temperature, and washed successively with water (20 mL), 1 mol/L aqueous hydrochloric acid (10 mL) and saturated aqueous sodium carbonate (10 mL). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound 43-4. The crude product was directly used in the next reaction without purification. 1 H NMR (400MHz, CDCl 3 ) δ6.76 (d, J = 1.2Hz, 1H), 5.16 (d, J = 0.8Hz, 1H), 4.46 (s, 2H), 3.38 (s, 6H).
4)化合物43-5的合成4) Synthesis of compound 43-5
室温下,将化合物43-4(1.00g,5.61mmol)溶于乙醇(20mL)中,随后加入湿钯碳(0.20g,10%纯度)和氢氧化钯碳(0.20g),氢气置换(50psi),反应液在50℃搅拌14小时。反应完毕后,反应液冷却到室温,过滤,滤液减压浓缩,得到化合物43-5。粗品不经纯化,直接用于下一步反应。1H NMR(400MHz,CDCl3)δ4.52(d,J=6.4Hz,1H),4.15-4.01(m,4H),3.41(s,6H),2.84-1.76(m,1H)。At room temperature, compound 43-4 (1.00g, 5.61mmol) was dissolved in ethanol (20mL), then wet palladium on carbon (0.20g, 10% purity) and palladium hydroxide on carbon (0.20g) were added, hydrogen replacement (50psi ), and the reaction solution was stirred at 50° C. for 14 hours. After the reaction was completed, the reaction liquid was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure to obtain compound 43-5. The crude product was directly used in the next reaction without purification. 1 H NMR (400MHz, CDCl 3 ) δ 4.52 (d, J=6.4Hz, 1H), 4.15-4.01 (m, 4H), 3.41 (s, 6H), 2.84-1.76 (m, 1H).
5)化合物43-6的合成5) Synthesis of Compound 43-6
室温下,将化合物43-5(150mg,0.83mmol)溶于1mol/L盐酸水溶液(3mL)中,反应液在80℃搅拌1小时。反应完毕后,待反应液冷却至室温,倒入水(10mL)中稀释,用乙酸乙酯(10mL×2)萃取。合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物43-6。1H NMR(400MHz,CDCl3)δ9.82(s,1H),4.47-4.36(m,2H),4.35-4.24(m,2H),3.43-3.29(m,1H)。Compound 43-5 (150 mg, 0.83 mmol) was dissolved in 1 mol/L hydrochloric acid aqueous solution (3 mL) at room temperature, and the reaction solution was stirred at 80° C. for 1 hour. After the reaction was completed, the reaction liquid was cooled to room temperature, poured into water (10 mL) for dilution, and extracted with ethyl acetate (10 mL×2). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound 43-6. 1 H NMR (400 MHz, CDCl 3 ) δ9.82 (s, 1H), 4.47-4.36 (m, 2H), 4.35-4.24 (m, 2H), 3.43-3.29 (m, 1H).
6)化合物43的合成6) Synthesis of compound 43
室温下,将化合物43-6(54.9mg,0.41mmol),化合物25-2(70mg,0.14mmol)和醋酸(8.19mg,0.14mmol)溶于甲醇(10mL)中,反应混合物在25℃搅拌0.5小时,随后加入氰基硼氢化钠(25.7mg,0.41mmol),反应混合物在25℃搅拌12小 时。反应完毕后,反应液倒入水(30mL)中稀释,用二氯甲烷(10mL×2)萃取。合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(二氯甲烷/甲醇=0/1)分离,得到化合物43。MS-ESI:m/z 564.2[M+H]+1H NMR(400MHz,CD3OD)δ8.03(s,1H),7.53(d,J=2.0Hz,1H),7.50(d,J=8.4Hz,1H),7.33(dd,J=8.4,2.0Hz,1H),5.18-5.07(m,1H),4.79(s,1H),4.24-4.12(m,2H),4.07(t,J=8.8Hz,1H),4.04-3.90(m,1H),3.86-3.73(m,3H),3.70-3.56(m,1H),2.84-2.72(m,3H),2.65-2.56(m,2H),2.55-2.43(m,1H),2.13-1.98(m,1H),1.80-1.67(m,4H),1.64(d,J=6.8Hz,3H),1.61-1.46(m,1H),0.99-0.84(m,1H)。At room temperature, compound 43-6 (54.9mg, 0.41mmol), compound 25-2 (70mg, 0.14mmol) and acetic acid (8.19mg, 0.14mmol) were dissolved in methanol (10mL), and the reaction mixture was stirred at 25°C for 0.5 hours, then sodium cyanoborohydride (25.7mg, 0.41mmol) was added, and the reaction mixture was stirred at 25°C for 12 hours hour. After the reaction was completed, the reaction solution was poured into water (30 mL) for dilution, and extracted with dichloromethane (10 mL×2). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (dichloromethane/methanol=0/1) to obtain compound 43. MS-ESI: m/z 564.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.03(s, 1H), 7.53(d, J=2.0Hz, 1H), 7.50(d, J=8.4Hz, 1H), 7.33(dd, J=8.4 ,2.0Hz,1H),5.18-5.07(m,1H),4.79(s,1H),4.24-4.12(m,2H),4.07(t,J=8.8Hz,1H),4.04-3.90(m, 1H),3.86-3.73(m,3H),3.70-3.56(m,1H),2.84-2.72(m,3H),2.65-2.56(m,2H),2.55-2.43(m,1H),2.13- 1.98 (m, 1H), 1.80-1.67 (m, 4H), 1.64 (d, J=6.8Hz, 3H), 1.61-1.46 (m, 1H), 0.99-0.84 (m, 1H).
实施例44:N-((R)-1-(2,4-二氯苯基)乙基)-5-(3-(R)-2-(2-(吡咯烷-1-基)乙基)哌啶-3-基)氮杂环丁烷-1-基)-[1,2,4]三唑并[1,5-a]嘧啶-7-胺(44)
Example 44: N-((R)-1-(2,4-dichlorophenyl)ethyl)-5-(3-(R)-2-(2-(pyrrolidin-1-yl)ethyl) Base) piperidin-3-yl) azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (44)
室温下,将化合物25-2(100mg,0.22mmol),1-(2-氯乙基)吡咯烷(75mg,0.44mmol)和N,N-二异丙基乙胺(229mg,1.77mmol)溶于乙腈(5mL)中,反应混合物在60℃搅拌12小时。反应完毕后,反应液冷却到室温,减压浓缩。所得残余物经硅胶制备薄层色谱(纯甲醇)分离,得到化合物44。MS-ESI:m/z 543.4[M+H]+1H NMR(400MHz,CD3OD)δ8.03(s,1H),7.53(d,J=2.0Hz,1H),7.50(d,J=8.4Hz,1H),7.33(dd,J=8.4,2.0Hz,1H),5.17-5.09(m,1H),4.80(s,1H),4.08(t,J=8.4Hz,1H),4.04-3.91(m,1H),3.77(dd,J=8.8,6.0Hz,1H),3.74-3.57(m,1H),2.95-2.82(m,2H),2.74-2.66(m,2H),2.65-2.58(m,4H),2.57-2.51(m,2H),2.50-2.41(m,1H),2.05-1.96(m,1H),1.84-1.78(m,4H),1.77-1.67(m,4H),1.64(d,J=6.8Hz,3H),1.60-1.49(m,1H),0.99-0.82(m,1H)。At room temperature, compound 25-2 (100mg, 0.22mmol), 1-(2-chloroethyl)pyrrolidine (75mg, 0.44mmol) and N,N-diisopropylethylamine (229mg, 1.77mmol) were dissolved In acetonitrile (5 mL), the reaction mixture was stirred at 60 °C for 12 hours. After the reaction was completed, the reaction solution was cooled to room temperature and concentrated under reduced pressure. The obtained residue was separated by silica gel preparative thin layer chromatography (pure methanol) to obtain compound 44. MS-ESI: m/z 543.4 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.03(s, 1H), 7.53(d, J=2.0Hz, 1H), 7.50(d, J=8.4Hz, 1H), 7.33(dd, J=8.4 ,2.0Hz,1H),5.17-5.09(m,1H),4.80(s,1H),4.08(t,J=8.4Hz,1H),4.04-3.91(m,1H),3.77(dd,J= 8.8,6.0Hz,1H),3.74-3.57(m,1H),2.95-2.82(m,2H),2.74-2.66(m,2H),2.65-2.58(m,4H),2.57-2.51(m, 2H), 2.50-2.41(m, 1H), 2.05-1.96(m, 1H), 1.84-1.78(m, 4H), 1.77-1.67(m, 4H), 1.64(d, J=6.8Hz, 3H) ,1.60-1.49(m,1H),0.99-0.82(m,1H).
实施例45:(1R,3r)-3-(4-(1-(7-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)-1-甲基环丁烷-1-醇(45)和实施例46:(1S,3s)-3-(4-(1-(7-(((R)-1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)-1-甲基环丁烷-1-醇(46)
Example 45: (1R,3r)-3-(4-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2, 4] Triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclobutane-1-ol (45) and Example 46: (1S,3s)-3-(4-(1-(7-(((R)-1-(2,4-dichlorophenyl)ethyl)amino)-[1,2 ,4] Triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)-1-methylcyclobutane-1-alcohol (46 )
1)化合物45-2的合成1) Synthesis of compound 45-2
室温下,将化合物45-1(200mg,0.83mmol),氯甲酸烯丙酯(120mg,1.00mmol)和碳酸钠(176mg,1.66mmol)混于二氯甲烷(5mL)中,反应混合物室温搅拌16小时。反应完毕后,反应液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=2/1)分离,得到化合物45-2。1H NMR(400MHz,CDCl3)δ6.02-5.88(m,1H),5.34-5.26(m,1H),5.24-5.18(m,1H),4.59(d,J=5.6Hz,2H),4.30-4.13(m,2H),3.96(t,J=8.4Hz,2H),3.64(dd,J=8.8,6.0Hz,2H),2.89-2.67(m,2H),2.31-2.17(m,1H),1.71-1.60(m,3H),1.44(s,9H),1.12-0.96(m,2H)。At room temperature, compound 45-1 (200mg, 0.83mmol), allyl chloroformate (120mg, 1.00mmol) and sodium carbonate (176mg, 1.66mmol) were mixed in dichloromethane (5mL), and the reaction mixture was stirred at room temperature for 16 Hour. After the reaction was completed, the reaction solution was concentrated under reduced pressure, and the resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to obtain compound 45-2. 1 H NMR (400MHz, CDCl 3 )δ6.02-5.88(m,1H),5.34-5.26(m,1H),5.24-5.18(m,1H),4.59(d,J=5.6Hz,2H), 4.30-4.13(m,2H),3.96(t,J=8.4Hz,2H),3.64(dd,J=8.8,6.0Hz,2H),2.89-2.67(m,2H),2.31-2.17(m, 1H), 1.71-1.60 (m, 3H), 1.44 (s, 9H), 1.12-0.96 (m, 2H).
2)化合物45-3的合成2) Synthesis of compound 45-3
室温下,将化合物45-2(269mg,0.83mmol)溶于二氧六环(3mL)中,加入4mol/L盐酸二氧六环溶液(3mL),反应混合物在室温搅拌1小时。反应完毕后,反应液减压浓缩,得到化合物45-3。粗品不经纯化,直接用于下一步反应。MS-ESI:m/z 225.2[M+H]+Compound 45-2 (269 mg, 0.83 mmol) was dissolved in dioxane (3 mL) at room temperature, 4 mol/L dioxane hydrochloride solution (3 mL) was added, and the reaction mixture was stirred at room temperature for 1 hour. After the reaction was completed, the reaction solution was concentrated under reduced pressure to obtain compound 45-3. The crude product was directly used in the next reaction without purification. MS-ESI: m/z 225.2 [M+H] + .
3)化合物45-4的合成3) Synthesis of compound 45-4
室温下,将化合物19-3(245mg,0.69mmol),化合物45-3(215mg,0.82mmol)和N,N-二异丙基乙胺(0.30mL,2.06mmol)溶于乙腈(4mL),反应混合物在60℃搅拌16小时。反应完毕后,向反应混合物中加水(30mL)稀释,乙酸乙酯(30mL×3)萃取。合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=1/1)分离,得到化合物45-4。MS-ESI:m/z 530.2[M+H]+At room temperature, compound 19-3 (245 mg, 0.69 mmol), compound 45-3 (215 mg, 0.82 mmol) and N,N-diisopropylethylamine (0.30 mL, 2.06 mmol) were dissolved in acetonitrile (4 mL), The reaction mixture was stirred at 60°C for 16 hours. After the reaction was completed, water (30 mL) was added to the reaction mixture to dilute, and ethyl acetate (30 mL×3) was extracted. The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to obtain compound 45-4. MS-ESI: m/z 530.2 [M+H] + .
4)化合物45-5的合成4) Synthesis of compound 45-5
室温下,将化合物45-4(375mg,0.71mmol),四(三苯基膦)钯(41mg,0.04mmol)和苯硅烷(765mg,7.07mmol)混于二氯甲烷(5mL)中,反应混合物在室温搅拌3小时。反应完毕后,反应液经硅胶柱层析(三乙胺/甲醇=10/1)分离,得到化合物45-5。MS-ESI:m/z 446.2[M+H]+At room temperature, compound 45-4 (375 mg, 0.71 mmol), tetrakis(triphenylphosphine) palladium (41 mg, 0.04 mmol) and phenylsilane (765 mg, 7.07 mmol) were mixed in dichloromethane (5 mL), and the reaction mixture Stir at room temperature for 3 hours. After the reaction was completed, the reaction solution was separated by silica gel column chromatography (triethylamine/methanol=10/1) to obtain compound 45-5. MS-ESI: m/z 446.2 [M+H] + .
5)化合物45和化合物46的合成 5) Synthesis of compound 45 and compound 46
室温下,将化合物45-5(150mg,0.34mmol),化合物33-1(48mg,0.34mmol,70%纯度)和醋酸(2mg,0.03mmol)溶于甲醇(3mL)中,室温搅拌0.5小时,随后加入氰基硼氢化钠(63mg,1.01mmol),反应混合物在室温继续搅拌15.5小时。反应完毕后,反应液减压浓缩,剩余物经硅胶制备薄层色谱(纯甲醇)分离,得到化合物45-6。化合物45-6经制备超临界流体色谱拆分(色谱柱:DAICEL CHIRALPAK AD(250mm*30mm*10μm);流动相:超临界二氧化碳,甲醇(0.1%一水合氨);梯度配比:甲醇相40%;流速:70mL/min;柱温:室温),得到化合物45和化合物46。化合物45:MS-ESI:m/z 530.3[M+H]+1H NMR(400MHz,CD3OD)δ8.03(s,1H),7.53(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.17-5.10(m,1H),4.81(s,1H),4.08(t,J=8.4Hz,1H),4.04-3.93(m,1H),3.79-3.73(m,1H),3.72-3.59(m,1H),2.96-2.86(m,3H),2.54-2.42(m,1H),2.18-2.11(m,2H),1.93-1.86(m,2H),1.84-1.77(m,2H),1.75-1.69(m,2H),1.65(d,J=6.8Hz,3H),1.56-1.51(m,1H),1.31(s,3H),1.21-1.15(m,2H)。化合物46:MS-ESI:m/z 530.4[M+H]+1H NMR(400MHz,CD3OD)δ8.03(s,1H),7.53(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.1Hz,1H),5.17-5.10(m,1H),4.81(s,1H),4.08(t,J=8.4Hz,1H),4.04-3.89(m,1H),3.80-3.73(m,1H),3.72-3.56(m,1H),2.95-2.88(m,2H),2.53-2.42(m,1H),2.40-2.29(m,1H),2.22-2.15(m,2H),1.98-1.90(m,2H),1.88-1.79(m,2H),1.77-1.68(m,2H),1.65(d,J=6.8Hz,3H),1.55-1.47(m,1H),1.29(s,3H),1.25-1.15(m,2H)。At room temperature, compound 45-5 (150 mg, 0.34 mmol), compound 33-1 (48 mg, 0.34 mmol, 70% purity) and acetic acid (2 mg, 0.03 mmol) were dissolved in methanol (3 mL), stirred at room temperature for 0.5 hours, Sodium cyanoborohydride (63 mg, 1.01 mmol) was then added and the reaction mixture was stirred at room temperature for a further 15.5 hours. After the reaction was completed, the reaction solution was concentrated under reduced pressure, and the residue was separated by silica gel preparative thin-layer chromatography (pure methanol) to obtain compound 45-6. Compound 45-6 was resolved by preparative supercritical fluid chromatography (column: DAICEL CHIRALPAK AD (250mm*30mm*10μm); mobile phase: supercritical carbon dioxide, methanol (0.1% ammonia monohydrate); gradient ratio: methanol phase 40 %; flow rate: 70mL/min; column temperature: room temperature), compound 45 and compound 46 were obtained. Compound 45: MS-ESI: m/z 530.3 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.03(s, 1H), 7.53(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz,1H),5.17-5.10(m,1H),4.81(s,1H),4.08(t,J=8.4Hz,1H),4.04-3.93(m,1H),3.79-3.73(m, 1H),3.72-3.59(m,1H),2.96-2.86(m,3H),2.54-2.42(m,1H),2.18-2.11(m,2H),1.93-1.86(m,2H),1.84- 1.77(m,2H),1.75-1.69(m,2H),1.65(d,J=6.8Hz,3H),1.56-1.51(m,1H),1.31(s,3H),1.21-1.15(m, 2H). Compound 46: MS-ESI: m/z 530.4 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.03(s, 1H), 7.53(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.1Hz,1H),5.17-5.10(m,1H),4.81(s,1H),4.08(t,J=8.4Hz,1H),4.04-3.89(m,1H),3.80-3.73(m, 1H),3.72-3.56(m,1H),2.95-2.88(m,2H),2.53-2.42(m,1H),2.40-2.29(m,1H),2.22-2.15(m,2H),1.98- 1.90(m,2H),1.88-1.79(m,2H),1.77-1.68(m,2H),1.65(d,J=6.8Hz,3H),1.55-1.47(m,1H),1.29(s, 3H), 1.25-1.15 (m, 2H).
实施例47:(R)-2-(4-(1-(7-((1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(47)
Example 47: (R)-2-(4-(1-(7-((1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4]triazolo[ 1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (47)
1)化合物47-2的合成1) Synthesis of compound 47-2
室温下,将化合物19-3(90mg,0.26mmol),N,N-二异丙基乙胺(0.10mL,0.79mmol)和化合物47-1(65mg,0.27mmol)溶于乙腈(2mL)中,反应混合物在80℃搅拌2小时。反应完毕后,向反应液中加水(30mL)稀释,用乙酸乙酯(30mL×3)萃取,合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=3/1)分离,得到化合物47-2。MS-ESI:m/z 546.2[M+H]+At room temperature, compound 19-3 (90 mg, 0.26 mmol), N,N-diisopropylethylamine (0.10 mL, 0.79 mmol) and compound 47-1 (65 mg, 0.27 mmol) were dissolved in acetonitrile (2 mL) , the reaction mixture was stirred at 80 °C for 2 hours. After the reaction was completed, add water (30mL) to the reaction liquid for dilution, extract with ethyl acetate (30mL×3), combine the organic phases, dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure. (petroleum ether/ethyl acetate=3/1) to obtain compound 47-2. MS-ESI: m/z 546.2 [M+H] + .
2)化合物47-3的合成2) Synthesis of compound 47-3
室温下,将化合物47-2(135mg,0.25mmol)溶于二氯甲烷(4mL)中,加入三氟乙酸(0.80mL),反应混合物在室温搅拌1小时。反应完毕后,反应液减压浓缩, 得到化合物47-3。粗品不经纯化,直接用于下一步反应。MS-ESI:m/z 446.1[M+H]+Compound 47-2 (135 mg, 0.25 mmol) was dissolved in dichloromethane (4 mL) at room temperature, trifluoroacetic acid (0.80 mL) was added, and the reaction mixture was stirred at room temperature for 1 hour. After completion of the reaction, the reaction solution was concentrated under reduced pressure, Compound 47-3 was obtained. The crude product was directly used in the next reaction without purification. MS-ESI: m/z 446.1 [M+H] + .
3)化合物47的合成3) Synthesis of Compound 47
室温下,将化合物47-3(138mg,0.25mmol),碘乙醇(127mg,0.74mmol)和碳酸钾(102mg,0.74mmol)混于乙腈(10mL)中,反应混合物在60℃搅拌2小时。反应完毕后,反应液过滤,滤液经硅胶柱层析(纯甲醇)分离,得到化合物47。MS-ESI:m/z 490.2[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.54(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.17-5.09(m,1H),4.81(s,1H),4.09(t,J=8.8Hz,1H),4.05-3.89(m,1H),3.80-3.74(m,1H),3.73-3.60(m,3H),3.15-3.05(m,2H),2.64(t,J=5.6Hz,2H),2.54-2.43(m,1H),2.29-2.16(m,2H),1.78-1.70(m,2H),1.65(d,J=6.8Hz,3H),1.61-1.49(m,1H),1.32-1.21(m,2H)。Compound 47-3 (138mg, 0.25mmol), iodoethanol (127mg, 0.74mmol) and potassium carbonate (102mg, 0.74mmol) were mixed in acetonitrile (10mL) at room temperature, and the reaction mixture was stirred at 60°C for 2 hours. After the reaction was completed, the reaction solution was filtered, and the filtrate was separated by silica gel column chromatography (pure methanol) to obtain compound 47. MS-ESI: m/z 490.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.54(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz,1H),5.17-5.09(m,1H),4.81(s,1H),4.09(t,J=8.8Hz,1H),4.05-3.89(m,1H),3.80-3.74(m, 1H), 3.73-3.60(m, 3H), 3.15-3.05(m, 2H), 2.64(t, J=5.6Hz, 2H), 2.54-2.43(m, 1H), 2.29-2.16(m, 2H) , 1.78-1.70 (m, 2H), 1.65 (d, J=6.8Hz, 3H), 1.61-1.49 (m, 1H), 1.32-1.21 (m, 2H).
实施例48:(R)-5-(3-(1-(2-氧杂螺[3.3]庚烷-6-基)哌啶-4-基)氮杂环丁烷-1-基)-N-(1-(2,4-二氯苯基)乙基)-[1,2,4]三唑并[1,5-a]嘧啶-7-胺(48)
Example 48: (R)-5-(3-(1-(2-oxaspiro[3.3]heptan-6-yl)piperidin-4-yl)azetidin-1-yl)- N-(1-(2,4-dichlorophenyl)ethyl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (48)
1)化合物48的合成1) Synthesis of Compound 48
室温下,将化合物45-5(40mg,0.09mmol),2-氧杂螺[3.3]庚烷-6-酮(12mg,0.11mmol)和醋酸(1mg,0.01mmol)溶于甲醇(1mL)中,搅拌10分钟,加入氰基硼氢化钠(17mg,0.27mmol),反应混合物在室温搅拌16小时。反应完毕后,向反应混合物中加水(20mL)稀释,用乙酸乙酯(30mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶制备薄层色谱(纯甲醇)分离,得到化合物48。MS-ESI:m/z 542.3[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.52(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.17-5.08(m,1H),4.80(s,1H),4.72(s,2H),4.60(s,2H),4.09(t,J=8.4Hz,1H),4.05-3.90(m,1H),3.79-3.73(m,1H),3.72-3.60(m,1H),3.09-3.00(m,2H),2.88-2.77(m,1H),2.55-2.43(m,3H),2.19-2.12(m,2H),2.11-2.01(m,2H),1.84-1.74(m,2H),1.65(d,J=6.8Hz,3H),1.62-1.52(m,1H),1.27-1.16(m,2H)。Compound 45-5 (40mg, 0.09mmol), 2-oxaspiro[3.3]heptan-6-one (12mg, 0.11mmol) and acetic acid (1mg, 0.01mmol) were dissolved in methanol (1mL) at room temperature , stirred for 10 minutes, sodium cyanoborohydride (17 mg, 0.27 mmol) was added, and the reaction mixture was stirred at room temperature for 16 hours. After the reaction is complete, add water (20 mL) to the reaction mixture to dilute, extract with ethyl acetate (30 mL×3), combine the organic phases, dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure, and prepare a thin layer of the residue on silica gel. Chromatography (pure methanol) yields compound 48. MS-ESI: m/z 542.3 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.52(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz,1H),5.17-5.08(m,1H),4.80(s,1H),4.72(s,2H),4.60(s,2H),4.09(t,J=8.4Hz,1H),4.05 -3.90(m,1H),3.79-3.73(m,1H),3.72-3.60(m,1H),3.09-3.00(m,2H),2.88-2.77(m,1H),2.55-2.43(m, 3H), 2.19-2.12(m, 2H), 2.11-2.01(m, 2H), 1.84-1.74(m, 2H), 1.65(d, J=6.8Hz, 3H), 1.62-1.52(m, 1H) ,1.27-1.16(m,2H).
实施例49:(R)-N-(1-(2,4-二氯苯基)乙基)-5-(3-(1-(3-(甲基磺酰基)丙基)哌啶-4-基)氮杂环丁烷-1-基)-[1,2,4]三唑并[1,5-a]嘧啶-7-胺(49)
Example 49: (R)-N-(1-(2,4-dichlorophenyl)ethyl)-5-(3-(1-(3-(methylsulfonyl)propyl)piperidine- 4-yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (49)
室温下,将化合物45-5(100mg,0.22mmol)和化合物49-1(59mg,0.20mmol)溶于乙腈(10mL)中,随后加入N,N-二异丙基乙胺(87mg,0.67mmol),反应液在80℃搅拌16小时。反应液冷却至室温,减压浓缩。所得残余物经制备级高效液相色谱(甲酸/乙腈/水体系,色谱柱:Phenomenex Luna C18 150*25mm*10μm;流动相:水(0.1%甲酸),乙腈;梯度配比:乙腈相(0-10min,20-40%);流速:25mL/min;柱温:室温)分离,得到化合物49。MS-ESI:m/z 566.2[M+H]+1H NMR(400MHz,CD3OD)δ8.05(s,1H),7.53(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.16-5.09(m,1H),4.81(s,1H),4.10(t,J=8.4Hz,1H),4.05-3.93(m,1H),3.78(dd,J=8.4,6.0Hz,1H),3.73-3.62(m,1H),3.27-3.17(m,4H),2.99(s,3H),2.87-2.79(m,2H),2.56-2.39(m,3H),2.16-2.07(m,2H),1.89-1.79(m,2H),1.65(d,J=6.8Hz,4H),1.38-1.23(m,2H)。At room temperature, compound 45-5 (100mg, 0.22mmol) and compound 49-1 (59mg, 0.20mmol) were dissolved in acetonitrile (10mL), then N,N-diisopropylethylamine (87mg, 0.67mmol ), and the reaction solution was stirred at 80° C. for 16 hours. The reaction solution was cooled to room temperature and concentrated under reduced pressure. The resulting residue was subjected to preparative high-performance liquid chromatography (formic acid/acetonitrile/water system, chromatographic column: Phenomenex Luna C18 150*25mm*10 μm; mobile phase: water (0.1% formic acid), acetonitrile; gradient ratio: acetonitrile phase (0 -10min, 20-40%); flow rate: 25mL/min; column temperature: room temperature) to obtain compound 49. MS-ESI: m/z 566.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.05(s, 1H), 7.53(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz,1H),5.16-5.09(m,1H),4.81(s,1H),4.10(t,J=8.4Hz,1H),4.05-3.93(m,1H),3.78(dd,J= 8.4,6.0Hz,1H),3.73-3.62(m,1H),3.27-3.17(m,4H),2.99(s,3H),2.87-2.79(m,2H),2.56-2.39(m,3H) , 2.16-2.07 (m, 2H), 1.89-1.79 (m, 2H), 1.65 (d, J=6.8Hz, 4H), 1.38-1.23 (m, 2H).
实施例50:(R)-N-(1-(2,4-二氯苯基)乙基)-5-(3-(1-(氧杂环丁烷-3-基)哌啶-4-基)氮杂环丁烷-1-基)-[1,2,4]三唑并[1,5-a]嘧啶-7-胺(50)
Example 50: (R)-N-(1-(2,4-dichlorophenyl)ethyl)-5-(3-(1-(oxetan-3-yl)piperidine-4 -yl)azetidin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7-amine (50)
室温下,将化合物45-5(80mg,0.18mmol)和氧杂环丁烷酮(26mg,0.36mmol)溶于甲醇(3mL)中,随后加入醋酸(11mg,0.18mmol)和氰基硼氢化钠(23mg,0.36mmol),反应液在室温搅拌14小时。反应完毕后,反应液用乙酸乙酯(30mL)稀释,饱和食盐水(10mL×3)洗涤,有机相经无水硫酸钠干燥,过滤,滤液浓缩。剩余物经硅胶制备薄层色谱(纯甲醇)分离,得到化合物50。MS-ESI:m/z 502.2[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.53(d,J=2.0Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.17-5.09(m,1H),4.81(s,1H),4.68(t,J=6.8Hz,2H),4.62-4.58(m,2H),4.09(t,J=8.4Hz,1H),4.04-3.95(m,1H),3.80-3.73(m,1H),3.72-3.61(m,1H),3.53-3.44(m,1H),2.87-2.76(m,2H),2.56-2.43(m,1H),1.93-1.82(m,2H),1.79-1.69(m,2H),1.65(d,J=6.8Hz,3H),1.59-1.45(m,1H),1.28-1.17(m,2H)。 Compound 45-5 (80 mg, 0.18 mmol) and oxetanone (26 mg, 0.36 mmol) were dissolved in methanol (3 mL) at room temperature, followed by the addition of acetic acid (11 mg, 0.18 mmol) and sodium cyanoborohydride (23mg, 0.36mmol), the reaction solution was stirred at room temperature for 14 hours. After the reaction was completed, the reaction solution was diluted with ethyl acetate (30 mL), washed with saturated brine (10 mL×3), the organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated. The residue was separated by silica gel preparative thin-layer chromatography (pure methanol) to obtain compound 50. MS-ESI: m/z 502.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.53(d, J=2.0Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz,1H),5.17-5.09(m,1H),4.81(s,1H),4.68(t,J=6.8Hz,2H),4.62-4.58(m,2H),4.09(t,J= 8.4Hz,1H),4.04-3.95(m,1H),3.80-3.73(m,1H),3.72-3.61(m,1H),3.53-3.44(m,1H),2.87-2.76(m,2H) ,2.56-2.43(m,1H),1.93-1.82(m,2H),1.79-1.69(m,2H),1.65(d,J=6.8Hz,3H),1.59-1.45(m,1H),1.28 -1.17(m,2H).
实施例51:(R)-2-(3-(1-(7-((1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)哌啶-4-基)氮杂环丁烷-1-基)乙烷-1-醇(51)
Example 51: (R)-2-(3-(1-(7-((1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4]triazolo[ 1,5-a]pyrimidin-5-yl)piperidin-4-yl)azetidin-1-yl)ethan-1-ol (51)
1)化合物51-2的合成1) Synthesis of compound 51-2
室温下,将化合物19-3(200mg,0.58mmol),化合物51-1(168mg,0.70mmol)和N,N-二异丙基乙胺(0.30mL,1.75mmol)溶于乙腈(3mL)中,反应混合物在80℃搅拌2小时。反应混合物加水(50mL)稀释,用乙酸乙酯(50mL×3)萃取,合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=3/1)分离,得到化合物51-2。1H NMR(400MHz,DMSO-d6)δ8.29(d,J=7.6Hz,1H),8.13(s,1H),7.67-7.62(m,2H),7.44(dd,J=8.4,2.0Hz,1H),5.26(s,1H),5.14-5.03(m,1H),4.32-4.13(m,2H),3.91-3.74(m,2H),3.63-3.50(m,2H),2.92-2.77(m,2H),2.24-2.11(m,1H),1.74-1.60(m,3H),1.58(d,J=6.8Hz,3H),1.36(s,9H),0.99-0.72(m,2H)。At room temperature, compound 19-3 (200mg, 0.58mmol), compound 51-1 (168mg, 0.70mmol) and N,N-diisopropylethylamine (0.30mL, 1.75mmol) were dissolved in acetonitrile (3mL) , the reaction mixture was stirred at 80 °C for 2 hours. The reaction mixture was diluted with water (50 mL), extracted with ethyl acetate (50 mL×3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was subjected to silica gel column chromatography (petroleum ether/acetic acid Ethyl ester=3/1) separation to obtain compound 51-2. 1 H NMR (400MHz, DMSO-d 6 ) δ8.29(d, J=7.6Hz, 1H), 8.13(s, 1H), 7.67-7.62(m, 2H), 7.44(dd, J=8.4, 2.0 Hz,1H),5.26(s,1H),5.14-5.03(m,1H),4.32-4.13(m,2H),3.91-3.74(m,2H),3.63-3.50(m,2H),2.92- 2.77(m,2H),2.24-2.11(m,1H),1.74-1.60(m,3H),1.58(d,J=6.8Hz,3H),1.36(s,9H),0.99-0.72(m, 2H).
2)化合物51-3的合成2) Synthesis of compound 51-3
室温下,将化合物51-2(290mg,0.53mmol)溶于二氯甲烷(4mL)中,加入三氟乙酸(1.60mL),反应混合物在室温搅拌1小时。反应完毕后,减压浓缩,得到化合物51-3。粗品不经纯化,直接用于下一步反应。MS-ESI:m/z 446.1[M+H]+Compound 51-2 (290 mg, 0.53 mmol) was dissolved in dichloromethane (4 mL) at room temperature, trifluoroacetic acid (1.60 mL) was added, and the reaction mixture was stirred at room temperature for 1 hour. After the reaction was completed, it was concentrated under reduced pressure to obtain compound 51-3. The crude product was directly used in the next reaction without purification. MS-ESI: m/z 446.1 [M+H] + .
3)化合物51-5的合成3) Synthesis of compound 51-5
室温下,将化合物51-3(280mg,0.50mmol),化合物51-4(105mg,0.60mmol)和醋酸(3mg,0.05mmol)溶于甲醇(5mL)中,反应混合物搅拌10分钟后,再加入氰基硼氢化钠(94mg,1.50mmol),反应混合物在室温搅拌16小时。反应完毕后,减压浓缩,所得残余物加水(20mL)稀释,乙酸乙酯(30mL×3)萃取,合并有机相,经无水硫酸钠干燥,减压浓缩,得到化合物51-5。粗品不经纯化,直接用于下一步反应。MS-ESI:m/z 604.3[M+H]+At room temperature, compound 51-3 (280 mg, 0.50 mmol), compound 51-4 (105 mg, 0.60 mmol) and acetic acid (3 mg, 0.05 mmol) were dissolved in methanol (5 mL), and the reaction mixture was stirred for 10 minutes before adding Sodium cyanoborohydride (94mg, 1.50mmol), the reaction mixture was stirred at room temperature for 16 hours. After the reaction was completed, it was concentrated under reduced pressure, and the obtained residue was diluted with water (20 mL), extracted with ethyl acetate (30 mL×3), the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain compound 51-5. The crude product was directly used in the next reaction without purification. MS-ESI: m/z 604.3 [M+H] + .
4)化合物51的合成4) Synthesis of compound 51
室温下,将化合物51-5(290mg,0.40mmol,84%纯度)溶于二氯甲烷(2.5mL)中,再加入三氟乙酸(1.00mL),反应混合物在室温搅拌1小时。反应完毕后,用氨水将反应混合物pH调为中性,减压浓缩,所得残余物经制备级高效液相色谱 (碳酸氢氨/乙腈/水体系,色谱柱:Waters Xbridge 150*25mm*5μm;流动相:水(0.5%碳酸氢氨),乙腈;梯度配比:乙腈相(0-8min,25-55%);流速:25mL/min;柱温:室温)分离,再经硅胶制备薄层色谱(乙酸乙酯/甲醇=0/1)分离,得到化合物51。MS-ESI:m/z 490.2[M+H]+1H NMR(400MHz,CD3OD)δ8.05(s,1H),7.56-7.49(m,2H),7.34(dd,J=8.4,2.0Hz,1H),5.20(s,1H),5.19-5.13(m,1H),4.41-4.20(m,2H),3.58-3.47(m,4H),2.97(t,J=7.6Hz,2H),2.94-2.80(m,2H),2.61(t,J=6.0Hz,2H),2.28-2.13(m,1H),1.79-1.67(m,2H),1.65(d,J=6.8Hz,3H),1.63-1.57(m,1H),1.06-0.93(m,1H),0.91-0.78(m,1H)。Compound 51-5 (290 mg, 0.40 mmol, 84% purity) was dissolved in dichloromethane (2.5 mL) at room temperature, and trifluoroacetic acid (1.00 mL) was added, and the reaction mixture was stirred at room temperature for 1 hour. After the reaction was completed, the pH of the reaction mixture was adjusted to neutral with ammonia water, concentrated under reduced pressure, and the resulting residue was subjected to preparative high performance liquid chromatography. (ammonia bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge 150*25mm*5μm; mobile phase: water (0.5% ammonium bicarbonate), acetonitrile; gradient ratio: acetonitrile phase (0-8min, 25-55% ); flow rate: 25mL/min; column temperature: room temperature), and then separated by silica gel preparative thin-layer chromatography (ethyl acetate/methanol=0/1) to obtain compound 51. MS-ESI: m/z 490.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.05(s, 1H), 7.56-7.49(m, 2H), 7.34(dd, J=8.4, 2.0Hz, 1H), 5.20(s, 1H), 5.19 -5.13(m,1H),4.41-4.20(m,2H),3.58-3.47(m,4H),2.97(t,J=7.6Hz,2H),2.94-2.80(m,2H),2.61(t ,J=6.0Hz,2H),2.28-2.13(m,1H),1.79-1.67(m,2H),1.65(d,J=6.8Hz,3H),1.63-1.57(m,1H),1.06- 0.93(m,1H),0.91-0.78(m,1H).
实施例52:(R)-2-(9-(7-((1-(2,4-二氯苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)-3,9-二氮杂螺[5.5]十一烷-3-基)乙烷-1-醇(52)
Example 52: (R)-2-(9-(7-((1-(2,4-dichlorophenyl)ethyl)amino)-[1,2,4]triazolo[1,5 -a]pyrimidin-5-yl)-3,9-diazaspiro[5.5]undec-3-yl)ethan-1-ol (52)
1)化合物52-2的合成1) Synthesis of compound 52-2
室温下,将化合物19-3(300mg,0.88mmol)和化合物52-1(223mg,0.88mmol)溶于乙腈(8mL)中,随后加入N,N-二异丙基乙胺(339mg,2.63mmol),反应混合物在80℃搅拌2小时。反应结束后,反应液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=1/5)分离,得到化合物52-2。MS-ESI:m/z 560.0[M+H]+At room temperature, compound 19-3 (300mg, 0.88mmol) and compound 52-1 (223mg, 0.88mmol) were dissolved in acetonitrile (8mL), then N,N-diisopropylethylamine (339mg, 2.63mmol ), and the reaction mixture was stirred at 80°C for 2 hours. After the reaction, the reaction solution was concentrated under reduced pressure, and the resulting residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/5) to obtain compound 52-2. MS-ESI: m/z 560.0 [M+H] + .
2)化合物52-3的合成2) Synthesis of compound 52-3
室温下,将化合物52-2(300mg,0.44mmol,83%纯度)溶于二氧六环(5mL)中,搅拌均匀后,缓慢加入4mol/L盐酸二氧六环溶液(5.00mL),反应液在20℃搅拌3小时。反应液减压浓缩,得到化合物52-3。粗品不经纯化,直接用于下一步反应。MS-ESI:m/z 460.1[M+H]+At room temperature, compound 52-2 (300mg, 0.44mmol, 83% purity) was dissolved in dioxane (5mL), and after stirring evenly, 4mol/L dioxane hydrochloride solution (5.00mL) was slowly added, and the reaction The solution was stirred at 20°C for 3 hours. The reaction solution was concentrated under reduced pressure to obtain compound 52-3. The crude product was directly used in the next reaction without purification. MS-ESI: m/z 460.1 [M+H] + .
3)化合物52的合成3) Synthesis of compound 52
室温下,将化合物52-3(180mg,0.34mmol,93%纯度)溶于乙腈(8mL)中,随后加入2-碘乙醇(116mg,0.67mmol)和碳酸钾(140mg,1.01mmol),反应混合物在80℃搅拌16小时。反应液减压浓缩,所得残余物经制备级高效液相色谱(碳酸氢铵/乙腈/水体系,色谱柱:Waters Xbridge C18 150*25mm*5μm;流动相:水(10mM碳酸氢铵),乙腈;梯度配比:乙腈相(0-10min,36-66%);流速:25mL/min;柱温:室温)分离,得到化合物52。MS-ESI:m/z 504.3[M+H]+1H NMR(400MHz,CD3OD)δ8.05(s,1H),7.54(d,J=2.4Hz,1H),7.51(d,J=8.4Hz,1H),7.34(dd,J=8.4,2.0Hz,1H),5.21(s,1H),5.19-5.13(m,1H),3.69(t,J=6.4Hz,2H),3.65- 3.47(m,4H),2.61-2.46(m,6H),1.66(d,J=6.8Hz,3H),1.56(t,J=5.6Hz,4H),1.52-1.37(m,4H)。At room temperature, compound 52-3 (180mg, 0.34mmol, 93% purity) was dissolved in acetonitrile (8mL), then 2-iodoethanol (116mg, 0.67mmol) and potassium carbonate (140mg, 1.01mmol) were added, and the reaction mixture Stir at 80°C for 16 hours. The reaction solution was concentrated under reduced pressure, and the resulting residue was subjected to preparative high-performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge C18 150*25mm*5μm; mobile phase: water (10mM ammonium bicarbonate), acetonitrile ; Gradient ratio: acetonitrile phase (0-10min, 36-66%); flow rate: 25mL/min; column temperature: room temperature) separation to obtain compound 52. MS-ESI: m/z 504.3 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.05(s, 1H), 7.54(d, J=2.4Hz, 1H), 7.51(d, J=8.4Hz, 1H), 7.34(dd, J=8.4 ,2.0Hz,1H),5.21(s,1H),5.19-5.13(m,1H),3.69(t,J=6.4Hz,2H),3.65- 3.47(m, 4H), 2.61-2.46(m, 6H), 1.66(d, J=6.8Hz, 3H), 1.56(t, J=5.6Hz, 4H), 1.52-1.37(m, 4H).
实施例53和实施例54:2-((R)-3-(1-(7-(((R)-1-(2-氯-4-甲基苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇和2-(R)-3-(1-(7-((R)-1-(4-氯-2-甲基苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(53和54)
Example 53 and Example 54: 2-((R)-3-(1-(7-(((R)-1-(2-chloro-4-methylphenyl)ethyl)amino)-[ 1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol and 2-(R) -3-(1-(7-((R)-1-(4-chloro-2-methylphenyl)ethyl)amino)-[1,2,4]triazolo[1,5-a ]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (53 and 54)
1)化合物53-2的合成1) Synthesis of Compound 53-2
室温下,将化合物53-1(500mg,3.30mmol)溶于甲苯(6mL)中,氮气置换,加入甲基溴化镁的四氢呋喃溶液(3.30mL,9.90mmol,3mol/L),反应混合物在110℃搅拌18小时。反应液冷却至0℃后,缓慢滴加稀盐酸至pH~2,升温至回流搅拌1小时,冷却至室温。反应液缓慢加入氢氧化钠至pH~7,乙酸乙酯萃取(30mL×3),合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=20/1)分离,得到化合物53-2。MS-ESI:m/z 169.1[M+H]+1H NMR(400MHz,CDCl3)δ7.51(d,J=7.6Hz,1H),7.25(s,1H),7.15-7.11(m,1H),2.64(s,3H),2.37(s,3H)。At room temperature, compound 53-1 (500mg, 3.30mmol) was dissolved in toluene (6mL), replaced with nitrogen, and a solution of methylmagnesium bromide in tetrahydrofuran (3.30mL, 9.90mmol, 3mol/L) was added, and the reaction mixture was heated at 110 °C and stirred for 18 hours. After the reaction solution was cooled to 0°C, dilute hydrochloric acid was slowly added dropwise to pH~2, the temperature was raised to reflux and stirred for 1 hour, and then cooled to room temperature. The reaction solution was slowly added sodium hydroxide to pH ~ 7, extracted with ethyl acetate (30 mL × 3), combined organic phases, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to obtain compound 53-2. MS-ESI: m/z 169.1 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ7.51(d, J=7.6Hz, 1H), 7.25(s, 1H), 7.15-7.11(m, 1H), 2.64(s, 3H), 2.37(s, 3H).
2)化合物53-3的合成2) Synthesis of compound 53-3
室温下,将化合物53-2(550mg,2.28mmol,70%纯度)和碳酸氢钠(575mg,6.85mmol)溶于乙醇(2mL)中,随后加入盐酸羟胺(317mg,4.57mmol),反应混合物在25℃下搅拌12小时。反应液倒入水中(30mL)稀释,用乙酸乙酯(50mL×3)萃取。合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品化合物53-3。MS-ESI:m/z 184.1[M+H]+At room temperature, compound 53-2 (550mg, 2.28mmol, 70% purity) and sodium bicarbonate (575mg, 6.85mmol) were dissolved in ethanol (2mL), then hydroxylamine hydrochloride (317mg, 4.57mmol) was added, and the reaction mixture was Stir at 25°C for 12 hours. The reaction solution was poured into water (30 mL) for dilution, and extracted with ethyl acetate (50 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain crude compound 53-3. MS-ESI: m/z 184.1 [M+H] + .
3)化合物53-4的合成3) Synthesis of Compound 53-4
室温下,将化合物53-3(450mg,1.52mmol,62%纯度)溶于四氢呋喃(10mL)中,缓慢滴入硼烷二甲硫醚溶液(0.50mL,5.00mmol,10mol/L),反应混合物在60℃搅拌16小时。反应液冷却至0℃,缓慢滴入甲醇(10mL),至无明显气泡产生后, 室温下继续搅拌1小时,减压浓缩,得到化合物53-4。MS-ESI:m/z 153.0[M-NH3+H]+At room temperature, compound 53-3 (450 mg, 1.52 mmol, 62% purity) was dissolved in tetrahydrofuran (10 mL), slowly added dropwise into borane dimethyl sulfide solution (0.50 mL, 5.00 mmol, 10 mol/L), and the reaction mixture Stir at 60°C for 16 hours. The reaction solution was cooled to 0°C, and methanol (10 mL) was slowly added dropwise until no obvious bubbles were generated. Stirring was continued at room temperature for 1 hour, and concentrated under reduced pressure to obtain compound 53-4. MS-ESI: m/z 153.0 [M-NH 3 +H] + .
4)化合物53-5的合成4) Synthesis of compound 53-5
室温下,将化合物53-4(200mg,0.84mmol,68%纯度),化合物19-2(158mg,0.84mmol)和N,N-二异丙基乙胺(0.40mL,2.51mmol)溶于乙腈(5mL)中,反应混合物在60℃搅拌4小时。反应完毕后,加水(20mL)稀释,用乙酸乙酯(30mL×3)萃取,合并有机相,经无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经硅胶柱层析(石油醚/乙酸乙酯=2/1)分离,得到化合物53-5。粗品未经纯化,直接用于下一步反应。MS-ESI:m/z 322.0[M+H]+At room temperature, compound 53-4 (200mg, 0.84mmol, 68% purity), compound 19-2 (158mg, 0.84mmol) and N,N-diisopropylethylamine (0.40mL, 2.51mmol) were dissolved in acetonitrile (5 mL), the reaction mixture was stirred at 60°C for 4 hours. After the reaction was completed, it was diluted with water (20 mL), extracted with ethyl acetate (30 mL×3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to obtain compound 53-5. The crude product was directly used in the next reaction without further purification. MS-ESI: m/z 322.0 [M+H] + .
5)化合物53和化合物54的合成5) Synthesis of compound 53 and compound 54
室温下,将化合物53-5(110mg,0.27mmol,78%纯度),中间体C(89mg,0.40mmol)和氟化铯(81mg,0.53mmol)溶于二甲亚砜(3mL)中,反应混合物在80℃搅拌16小时。反应完毕后,过滤,滤液经反相色谱柱(乙腈/水体系,色谱柱:C18spherical 20-35μm40g,流动相:水(0.2%甲酸),乙腈;梯度配比:乙腈相(0-8min,50-65%);流速:40mL/min;柱温:室温)分离,得到化合物53-6。再经超临界流体色谱拆分(色谱柱:DAICEL CHIRALCEL OX(250mm*30mm*10μm);流动相:超临界二氧化碳,甲醇(0.1%一水合氨);梯度配比:甲醇相60%;流速:80mL/min;柱温:室温),得到化合物53和化合物54。(化合物53为第一个洗脱的峰,化合物54为第二个洗脱的峰)。化合物53:MS-ESI:m/z 470.2[M+H]+1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.37(d,J=7.6Hz,1H),7.28(s,1H),7.13(d,J=7.6Hz,1H),5.15-5.08(m,1H),4.84(s,1H),4.08(t,J=8.4Hz,1H),4.03-3.93(m,1H),3.80-3.75(m,1H),3.74-3.68(m,3H),3.05-2.95(m,2H),2.63(t,J=5.6Hz,2H),2.51-2.43(m,1H),2.32(s,3H),2.20-2.12(m,1H),1.86-1.73(m,4H),1.63(d,J=6.8Hz,3H),1.62-1.56(m,1H),0.96-0.88(m,1H)。化合物54:MS-ESI:m/z 470.2[M+H]+1H NMR(400MHz,CD3OD)δ8.05(s,1H),7.39(d,J=8.0Hz,1H),7.29(s,1H),7.14(d,J=8.0Hz,1H),5.17-5.10(m,1H),4.86(s,1H),4.10(t,J=8.4Hz,1H),4.06-3.95(m,1H),3.83-3.78(m,1H),3.76-3.65(m,3H),3.06-2.94(m,2H),2.64(t,J=6.0Hz,2H),2.54-2.45(m,1H),2.33(s,3H),2.20-2.11(m,1H),1.87-1.75(m,4H),1.65(d,J=6.8Hz,3H),1.64-1.57(m,1H),0.97-0.90(m,1H)。At room temperature, compound 53-5 (110 mg, 0.27 mmol, 78% purity), intermediate C (89 mg, 0.40 mmol) and cesium fluoride (81 mg, 0.53 mmol) were dissolved in dimethyl sulfoxide (3 mL), and the reaction The mixture was stirred at 80°C for 16 hours. After completion of the reaction, filter, and the filtrate passes through a reversed-phase chromatographic column (acetonitrile/water system, chromatographic column: C18spherical 20-35 μm 40g, mobile phase: water (0.2% formic acid), acetonitrile; gradient ratio: acetonitrile phase (0-8min, 50-65%); flow rate: 40mL/min; column temperature: room temperature) separation to obtain compound 53-6. Then separated by supercritical fluid chromatography (chromatographic column: DAICEL CHIRALCEL OX (250mm*30mm*10μm); mobile phase: supercritical carbon dioxide, methanol (0.1% ammonia monohydrate); gradient ratio: methanol phase 60%; flow rate: 80mL/min; column temperature: room temperature), to obtain compound 53 and compound 54. (Compound 53 was the first eluting peak, compound 54 was the second eluting peak). Compound 53: MS-ESI: m/z 470.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.04(s, 1H), 7.37(d, J=7.6Hz, 1H), 7.28(s, 1H), 7.13(d, J=7.6Hz, 1H), 5.15-5.08(m,1H),4.84(s,1H),4.08(t,J=8.4Hz,1H),4.03-3.93(m,1H),3.80-3.75(m,1H),3.74-3.68( m,3H),3.05-2.95(m,2H),2.63(t,J=5.6Hz,2H),2.51-2.43(m,1H),2.32(s,3H),2.20-2.12(m,1H) , 1.86-1.73 (m, 4H), 1.63 (d, J=6.8Hz, 3H), 1.62-1.56 (m, 1H), 0.96-0.88 (m, 1H). Compound 54: MS-ESI: m/z 470.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.05(s, 1H), 7.39(d, J=8.0Hz, 1H), 7.29(s, 1H), 7.14(d, J=8.0Hz, 1H), 5.17-5.10(m,1H),4.86(s,1H),4.10(t,J=8.4Hz,1H),4.06-3.95(m,1H),3.83-3.78(m,1H),3.76-3.65( m,3H),3.06-2.94(m,2H),2.64(t,J=6.0Hz,2H),2.54-2.45(m,1H),2.33(s,3H),2.20-2.11(m,1H) , 1.87-1.75 (m, 4H), 1.65 (d, J=6.8Hz, 3H), 1.64-1.57 (m, 1H), 0.97-0.90 (m, 1H).
实施例55:2-(R)-3-(1-(7-((R)-1-(2,4-二氟苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(55)
Example 55: 2-(R)-3-(1-(7-((R)-1-(2,4-difluorophenyl)ethyl)amino)-[1,2,4]triazole And[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (55)
参照实施例19的合成方法制备化合物55。MS-ESI:m/z 470.2[M+H]+1H NMR(400MHz,CD3OD)δ8.03(s,1H),7.36(d,J=8.4Hz,1H),7.25(d,J=2.0Hz,1H),7.18(dd,J=8.4,2.0Hz,1H),4.97-4.93(m,1H),4.73(s,1H),4.12-4.04(m,1H),4.03-3.87(m,1H),3.82-3.74(m,1H),3.73-3.60(m,3H),3.01-2.86(m,2H),2.56(t,J=6.0Hz,2H),2.52-2.39(m,4H),2.13-2.01(m,1H),1.83-1.69(m,4H),1.67-1.53(m,4H),0.96-0.85(m,1H)。Compound 55 was prepared according to the synthesis method of Example 19. MS-ESI: m/z 470.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.03(s, 1H), 7.36(d, J=8.4Hz, 1H), 7.25(d, J=2.0Hz, 1H), 7.18(dd, J=8.4 ,2.0Hz,1H),4.97-4.93(m,1H),4.73(s,1H),4.12-4.04(m,1H),4.03-3.87(m,1H),3.82-3.74(m,1H), 3.73-3.60(m,3H),3.01-2.86(m,2H),2.56(t,J=6.0Hz,2H),2.52-2.39(m,4H),2.13-2.01(m,1H),1.83- 1.69 (m, 4H), 1.67-1.53 (m, 4H), 0.96-0.85 (m, 1H).
实施例56:2-(R)-3-(1-(7-((R)-1-(4-氯-2-氟苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(56)
Example 56: 2-(R)-3-(1-(7-((R)-1-(4-chloro-2-fluorophenyl)ethyl)amino)-[1,2,4]tri Azolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (56)
参照实施例19的合成方法制备化合物56。MS-ESI:m/z 458.2[M+H]+1H NMR(400MHz,CD3OD)δ8.03(s,1H),7.56-7.45(m,1H),7.08-6.93(m,2H),5.09-5.03(m,1H),4.97(s,1H),4.11(t,J=8.4Hz,1H),4.07-3.91(m,1H),3.84-3.78(m,1H),3.77-3.65(m,3H),3.04-2.91(m,2H),2.59(t,J=6.0Hz,2H),2.53-2.45(m,1H),2.10(t,J=11.2Hz,1H),1.85-1.71(m,4H),1.70-1.57(m,4H),0.96-0.86(m,1H)。Compound 56 was prepared according to the synthesis method of Example 19. MS-ESI: m/z 458.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.03(s, 1H), 7.56-7.45(m, 1H), 7.08-6.93(m, 2H), 5.09-5.03(m, 1H), 4.97(s, 1H), 4.11(t, J=8.4Hz, 1H), 4.07-3.91(m, 1H), 3.84-3.78(m, 1H), 3.77-3.65(m, 3H), 3.04-2.91(m, 2H) ,2.59(t,J=6.0Hz,2H),2.53-2.45(m,1H),2.10(t,J=11.2Hz,1H),1.85-1.71(m,4H),1.70-1.57(m,4H ),0.96-0.86(m,1H).
实施例57:2-(R)-3-(1-(7-((R)-1-(2-氯-4-氟苯基)乙基)氨基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)哌啶-1-基)乙烷-1-醇(57)
Example 57: 2-(R)-3-(1-(7-((R)-1-(2-chloro-4-fluorophenyl)ethyl)amino)-[1,2,4]tri Azolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)piperidin-1-yl)ethan-1-ol (57)
参照实施例19的合成方法制备化合物57。MS-ESI:m/z 474.2[M+H]+1H NMR(400MHz,CD3OD)δ8.03(s,1H),7.46(t,J=8.4Hz,1H),7.26(dd,J=10.4,1.6Hz,1H),7.21(d,J=8.4Hz,1H),5.10-5.03(m,1H),4.95(s,1H),4.11(t,J=8.4Hz,1H),4.05-3.93(m,1H),3.80(dd,J=8.4,6.0Hz,1H),3.74-3.63(m,3H),2.92(t,J=9.2Hz,1H),2.54(t,J=6.0Hz,2H),2.50-2.41(m,1H),2.09-1.98(m,1H),1.84-1.70(m,4H),1.68(d,J=6.8Hz,3H),1.63-1.53(m,1H),0.94-0.85(m,1H)。Compound 57 was prepared according to the synthesis method of Example 19. MS-ESI: m/z 474.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ8.03(s, 1H), 7.46(t, J=8.4Hz, 1H), 7.26(dd, J=10.4, 1.6Hz, 1H), 7.21(d, J =8.4Hz,1H),5.10-5.03(m,1H),4.95(s,1H),4.11(t,J=8.4Hz,1H),4.05-3.93(m,1H),3.80(dd,J= 8.4,6.0Hz,1H),3.74-3.63(m,3H),2.92(t,J=9.2Hz,1H),2.54(t,J=6.0Hz,2H),2.50-2.41(m,1H), 2.09-1.98 (m, 1H), 1.84-1.70 (m, 4H), 1.68 (d, J=6.8Hz, 3H), 1.63-1.53 (m, 1H), 0.94-0.85 (m, 1H).
实施例58:2-(R)-2-(1-(7-((R)-1-(2,4-二氯苯基)乙基)氨基)-2-(三氟甲基)-[1,2,4]三唑并[1,5-a]嘧啶-5-基)氮杂环丁烷-3-基)吗啉基)乙烷-1-醇(58)
Example 58: 2-(R)-2-(1-(7-((R)-1-(2,4-dichlorophenyl)ethyl)amino)-2-(trifluoromethyl)- [1,2,4]triazolo[1,5-a]pyrimidin-5-yl)azetidin-3-yl)morpholinyl)ethan-1-ol (58)
1)化合物58-2的合成1) Synthesis of compound 58-2
室温下,将化合物58-1(3.00g,16.20mmol)溶于无水甲醇(30mL)中,冰浴下,加入硝基甲烷(2.97g,48.59mmol)和三乙胺(3.28g,32.39mmol),反应混合物在室温搅拌16小时。反应液减压浓缩,所得残余物经硅胶柱层析(石油醚/乙酸乙酯=1/1)分离,得到化合物58-2。1H NMR(400MHz,CDCl3)δ4.52-4.45(m,1H),4.39-4.32(m,2H),4.00(td,J=8.8,1.6Hz,2H),3.93(dd,J=8.8,5.2Hz,1H),3.74(dd,J=8.8,5.6Hz,1H),3.32(brs,1H),2.68-2.57(m,1H),1.44(s,9H)。At room temperature, compound 58-1 (3.00g, 16.20mmol) was dissolved in anhydrous methanol (30mL), under ice cooling, nitromethane (2.97g, 48.59mmol) and triethylamine (3.28g, 32.39mmol) were added ), and the reaction mixture was stirred at room temperature for 16 hours. The reaction solution was concentrated under reduced pressure, and the obtained residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to obtain compound 58-2. 1 H NMR (400MHz, CDCl 3 ) δ4.52-4.45 (m, 1H), 4.39-4.32 (m, 2H), 4.00 (td, J=8.8, 1.6Hz, 2H), 3.93 (dd, J=8.8 , 5.2Hz, 1H), 3.74 (dd, J = 8.8, 5.6Hz, 1H), 3.32 (brs, 1H), 2.68-2.57 (m, 1H), 1.44 (s, 9H).
2)化合物58-3的合成2) Synthesis of compound 58-3
室温下,将化合物58-2(3.60g,14.62mmol)和二氧化铂(664mg,2.92mmol)加入甲醇(50mL)中,氢气氛围下,室温反应16小时。反应液过滤,减压浓缩,得到化合物58-3。1H NMR(400MHz,CDCl3)δ4.01-3.88(m,3H),3.71(dd,J=8.4,5.6Hz,1H),3.65(td,J=8.0,3.2Hz,1H),2.82(dd,J=8.8,3.6Hz,1H),2.57-2.38(m,1H),2.45(dd,J=12.8,8.4Hz,1H),1.44(s,9H)。At room temperature, compound 58-2 (3.60 g, 14.62 mmol) and platinum dioxide (664 mg, 2.92 mmol) were added into methanol (50 mL), and reacted at room temperature for 16 hours under hydrogen atmosphere. The reaction solution was filtered and concentrated under reduced pressure to obtain compound 58-3. 1 H NMR (400MHz, CDCl 3 ) δ4.01-3.88 (m, 3H), 3.71 (dd, J=8.4, 5.6Hz, 1H), 3.65 (td, J=8.0, 3.2Hz, 1H), 2.82( dd, J=8.8, 3.6Hz, 1H), 2.57-2.38(m, 1H), 2.45(dd, J=12.8, 8.4Hz, 1H), 1.44(s, 9H).
3)化合物58-4的合成3) Synthesis of compound 58-4
室温下,将化合物58-3(3.10g,14.33mmol)和三乙胺(3.99mL,28.67mmol)溶入二氯甲烷(50mL),缓慢滴加氯乙酰氯(1.78g,15.77mmol),室温反应16小时。反应液加水(50mL)稀释,二氯甲烷萃取(50mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物58-4。1H NMR(400MHz,CDCl3)δ7.13-7.03(m,1H),4.00(s,2H),3.96(td,J=12.8,2.8Hz,2H),3.94-3.86(m,2H),3.73(dd,J=8.8,5.6Hz,1H),3.50-3.41(m,1H),3.26-3.17(m,1H),3.64-2.52(m,1H),1.44(s,9H)。At room temperature, compound 58-3 (3.10g, 14.33mmol) and triethylamine (3.99mL, 28.67mmol) were dissolved in dichloromethane (50mL), and chloroacetyl chloride (1.78g, 15.77mmol) was slowly added dropwise, at room temperature React for 16 hours. The reaction solution was diluted with water (50 mL), extracted with dichloromethane (50 mL×2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain compound 58-4. 1 H NMR (400MHz, CDCl 3 ) δ7.13-7.03 (m, 1H), 4.00 (s, 2H), 3.96 (td, J=12.8, 2.8Hz, 2H), 3.94-3.86 (m, 2H), 3.73 (dd, J = 8.8, 5.6 Hz, 1H), 3.50-3.41 (m, 1H), 3.26-3.17 (m, 1H), 3.64-2.52 (m, 1H), 1.44 (s, 9H).
4)中间体58-5的合成 4) Synthesis of intermediate 58-5
室温下,将化合物58-4(3.10g,10.59mmol)和叔丁醇钾(1.19g,10.59mmol)溶入四氢呋喃(20mL)中,反应液在60℃反应2小时。反应完毕后,反应液冷却至室温,减压浓缩,残余物经硅胶柱层析(乙酸乙酯/甲醇=10/1)纯化,得到中间体58-5。1H NMR(400MHz,CDCl3)δ6.57(s,1H),4.37-4.16(m,2H),3.98(td,J=8.4,3.6Hz,2H),3.90(dd,J=8.8,5.6Hz,1H),3.87-3.81(m,1H),3.74(dd,J=16.0,10.0Hz,1H),3.32-3.17(m,2H),3.69-2.60(m,1H),1.44(s,9H)。Compound 58-4 (3.10 g, 10.59 mmol) and potassium tert-butoxide (1.19 g, 10.59 mmol) were dissolved in tetrahydrofuran (20 mL) at room temperature, and the reaction solution was reacted at 60° C. for 2 hours. After completion of the reaction, the reaction liquid was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (ethyl acetate/methanol=10/1) to obtain intermediate 58-5. 1 H NMR (400MHz, CDCl 3 ) δ6.57(s, 1H), 4.37-4.16(m, 2H), 3.98(td, J=8.4, 3.6Hz, 2H), 3.90(dd, J=8.8, 5.6 Hz,1H),3.87-3.81(m,1H),3.74(dd,J=16.0,10.0Hz,1H),3.32-3.17(m,2H),3.69-2.60(m,1H),1.44(s, 9H).
5)化合物58-6的合成5) Synthesis of Compound 58-6
室温下,依次加入钠氢(749mg,18.73mmol,60%纯度)和N,N-二甲基甲酰胺(30mL),氮气保护下加入中间体58-5(3.20g,12.49mmol),冰浴下反应0.5小时,加入苄基-2-溴乙基醚(4.03g,18.73mmol),反应混合物在室温搅拌1小时。反应完毕后,向反应液中缓慢加入饱和氯化铵水溶液(100mL)淬灭,用乙酸乙酯(80mL×2)萃取。合并有机相,饱和食盐水洗涤(80mL×3),无水硫酸钠干燥,过滤,滤液减压浓缩。剩余物经硅胶柱层析(石油醚/乙酸乙酯=0/1)分离,得到化合物58-6。1H NMR(400MHz,CDCl3)δ7.39-7.34(m,2H),7.33-7.25(m,3H),4.51(s,2H),4.35-4.14(m,2H),3.96(td,J=8.4,2.0Hz,2H),3.88(dd,J=8.8,5.6Hz,1H),3.85-3.78(m,1H),3.73-3.67(m,3H),3.63-3.56(m,2H),3.36-3.26(m,2H),2.64-2.54(m,1H),1.45(s,9H)。At room temperature, sodium hydrogen (749mg, 18.73mmol, 60% purity) and N,N-dimethylformamide (30mL) were added sequentially, and intermediate 58-5 (3.20g, 12.49mmol) was added under nitrogen protection, and ice bath After reacting for 0.5 hours, benzyl-2-bromoethyl ether (4.03 g, 18.73 mmol) was added, and the reaction mixture was stirred at room temperature for 1 hour. After the reaction was completed, saturated ammonium chloride aqueous solution (100 mL) was slowly added to the reaction liquid to quench, and extracted with ethyl acetate (80 mL×2). The organic phases were combined, washed with saturated brine (80 mL×3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate=0/1) to obtain compound 58-6. 1 H NMR (400MHz, CDCl 3 )δ7.39-7.34(m,2H),7.33-7.25(m,3H),4.51(s,2H),4.35-4.14(m,2H),3.96(td,J =8.4,2.0Hz,2H),3.88(dd,J=8.8,5.6Hz,1H),3.85-3.78(m,1H),3.73-3.67(m,3H),3.63-3.56(m,2H), 3.36-3.26 (m, 2H), 2.64-2.54 (m, 1H), 1.45 (s, 9H).
6)化合物58-8和化合物58-9的合成6) Synthesis of compound 58-8 and compound 58-9
室温下,将化合物58-6(2.60g,6.66mmol)溶于无水四氢呋喃(50mL)中,冰浴下滴加硼烷二甲硫醚(3.30mL,33.29mmol,10mol/L),反应混合物在50℃搅拌16小时。反应液冷却至室温,向反应液中缓慢滴加甲醇(10mL)淬灭硼烷,直至无气泡冒出,反应液减压浓缩。剩余物经硅胶柱层析(石油醚/乙酸乙酯=1/1)分离,减压蒸馏,得到粗品,加入甘油(2mL)和甲醇(20mL),室温搅拌过夜,减压浓缩,所得剩余物经硅胶柱层析(石油醚/乙酸乙酯=1/1)分离,得到化合物58-7。MS-ESI:m/z 377.1[M+H]+。化合物58-7(1.67g)经超临界流体色谱拆分(色谱柱:Phenomenex-Cellulose-2(250mm*30mm,10μm);流动相:超临界二氧化碳,甲醇(0.1%一水合氨);梯度配比:甲醇相30%;流速:60mL/min;柱温:室温),得到化合物58-8和化合物58-9。(化合物58-8为第一个洗脱的峰,化合物58-9为第二个洗脱的峰)。At room temperature, compound 58-6 (2.60g, 6.66mmol) was dissolved in anhydrous tetrahydrofuran (50mL), and borane dimethyl sulfide (3.30mL, 33.29mmol, 10mol/L) was added dropwise under ice cooling, and the reaction mixture was Stir at 50°C for 16 hours. The reaction solution was cooled to room temperature, methanol (10 mL) was slowly added dropwise to the reaction solution to quench borane until no bubbles emerged, and the reaction solution was concentrated under reduced pressure. The residue was separated by silica gel column chromatography (petroleum ether/ethyl acetate = 1/1), and distilled under reduced pressure to obtain the crude product. Glycerol (2 mL) and methanol (20 mL) were added, stirred overnight at room temperature, and concentrated under reduced pressure to obtain the residue After separation by silica gel column chromatography (petroleum ether/ethyl acetate=1/1), compound 58-7 was obtained. MS-ESI: m/z 377.1 [M+H] + . Compound 58-7 (1.67g) was resolved by supercritical fluid chromatography (chromatographic column: Phenomenex-Cellulose-2 (250mm*30mm, 10μm); mobile phase: supercritical carbon dioxide, methanol (0.1% ammonia monohydrate); gradient formulation Ratio: methanol phase 30%; flow rate: 60mL/min; column temperature: room temperature), to obtain compound 58-8 and compound 58-9. (Compound 58-8 was the first eluting peak, compound 58-9 was the second eluting peak).
7)化合物58-10的合成7) Synthesis of compound 58-10
室温下,将化合物58-9(500mg,1.33mmol)溶于二氯甲烷(10mL)中,氮气置换,反应液冷却至-78℃,缓慢滴入三溴化硼(3.33g,13.28mmol),反应液在-78℃搅拌30分钟,随后加入甲醇(2mL),反应混合物-78℃继续搅拌1小时。反应完毕后,反应液减压浓缩,得到化合物58-10。粗品不经纯化,直接用于下一步反应。MS-ESI:m/z 187.1[M+H]+At room temperature, compound 58-9 (500mg, 1.33mmol) was dissolved in dichloromethane (10mL), replaced with nitrogen, the reaction solution was cooled to -78°C, and boron tribromide (3.33g, 13.28mmol) was slowly added dropwise, The reaction was stirred at -78°C for 30 minutes, then methanol (2 mL) was added, and the reaction mixture was stirred at -78°C for an additional 1 hour. After completion of the reaction, the reaction solution was concentrated under reduced pressure to obtain compound 58-10. The crude product was directly used in the next reaction without purification. MS-ESI: m/z 187.1 [M+H] + .
8)化合物58的合成 8) Synthesis of Compound 58
室温下,将化合物21-3(100mg,0.17mmol,71%纯度)和化合物58-10(46mg,0.17mmol)溶于二氯乙烷(10mL)中,随后加入二异丙基乙胺(112mg,0.87mmol),反应液在80℃搅拌16小时。反应液冷却至室温,减压浓缩。所得残余物经制备级高效液相色谱(碳酸氢铵/乙腈/水体系,色谱柱:Waters Xbridge 150*25mm*5μm;流动相:水(10mM碳酸氢铵),乙腈;梯度配比:乙腈相(0-8min,45-75%);流速:25mL/min;柱温:室温)分离,得到化合物58。MS-ESI:m/z 560.2[M+H]+1H NMR(400MHz,CD3OD)δ7.54(d,J=2.0Hz,1H),7.51(d,J=8.8Hz,1H),7.35(dd,J=8.4,1.6Hz,1H),5.18-5.09(m,1H),4.87-4.86(m,1H),4.09-3.97(m,2H),3.94-3.78(m,3H),3.73-3.61(m,4H),2.86-2.71(m,3H),2.53(t,J=5.6Hz,2H),2.20(td,J=11.2,2.8Hz,1H),1.84(t,J=11.2Hz,1H),1.64(d,J=6.8Hz,3H)。At room temperature, compound 21-3 (100mg, 0.17mmol, 71% purity) and compound 58-10 (46mg, 0.17mmol) were dissolved in dichloroethane (10mL), then diisopropylethylamine (112mg ,0.87mmol), the reaction solution was stirred at 80°C for 16 hours. The reaction solution was cooled to room temperature and concentrated under reduced pressure. The resulting residue was subjected to preparative high-performance liquid chromatography (ammonium bicarbonate/acetonitrile/water system, chromatographic column: Waters Xbridge 150*25mm*5 μm; mobile phase: water (10mM ammonium bicarbonate), acetonitrile; gradient ratio: acetonitrile phase (0-8min, 45-75%); flow rate: 25mL/min; column temperature: room temperature) to obtain compound 58. MS-ESI: m/z 560.2 [M+H] + . 1 H NMR (400MHz, CD 3 OD) δ7.54 (d, J = 2.0Hz, 1H), 7.51 (d, J = 8.8Hz, 1H), 7.35 (dd, J = 8.4, 1.6Hz, 1H), 5.18-5.09(m,1H),4.87-4.86(m,1H),4.09-3.97(m,2H),3.94-3.78(m,3H),3.73-3.61(m,4H),2.86-2.71(m ,3H),2.53(t,J=5.6Hz,2H),2.20(td,J=11.2,2.8Hz,1H),1.84(t,J=11.2Hz,1H),1.64(d,J=6.8Hz ,3H).
生物学评价biological evaluation
以下结合测试例进一步描述解释本发明的内容,但这些测试例并非意味着限制本发明的范围。The content of the present invention is further described and explained in conjunction with test examples below, but these test examples are not meant to limit the scope of the present invention.
测试例1:体外CCR4钙流活性检测实验Test Example 1: In Vitro CCR4 Calcium Flux Activity Detection Experiment
1.1、实验材料
1.1. Experimental materials
1.2、实验步骤1.2. Experimental steps
体外CCR4受体细胞功能性实验通过使用稳定表达CCR4的细胞株及FLIPR实验检测待测化合物活性。实验中,受试化合物测试起始浓度为10μM,3倍稀释,共10个浓度,复孔测试。The in vitro CCR4 receptor cell functional assay was used to detect the activity of the compound to be tested by using a cell line stably expressing CCR4 and FLIPR assay. In the experiment, the initial concentration of the test compound was 10 μM, diluted 3 times, and a total of 10 concentrations were tested in duplicate wells.
稳定表达CCR4的细胞株(CCR4-HEK293)使用DMEM培养基并添加胎牛血清、G418和青霉素-链霉素进行培养。配置250mM FLIPR实验缓冲液,2×Fluo-4DirectTM加样缓冲液。收集并重悬CCR4-HEK293细胞至1x106/mL,在384孔细胞培养微孔板的每个反应孔中加入20μL细胞悬液。细胞培养过夜。第二天,细胞反应板弃上清,加入20μL实验缓冲液和20μL Fluo-4DirectTM加样缓冲液。从 化合物板(预梯度稀释在384孔PP微孔板)取10μL待测化合物稀释液加到细胞反应板,在37℃ 5%二氧化碳环境中孵育50分钟,再在室温孵育10分钟。将反应板转移到FLIPR Tetra System(Molecular Devices)。取10μL 6×EC80重组人CCL22稀释液加到细胞反应板,震荡2次。读取荧光值,使用Graphpad软件分析数据,计算化合物对CCR4受体细胞钙流抑制活性的IC50A cell line stably expressing CCR4 (CCR4-HEK293) was cultured in DMEM medium supplemented with fetal bovine serum, G418 and penicillin-streptomycin. Configure 250mM FLIPR experimental buffer, 2×Fluo-4Direct TM loading buffer. Collect and resuspend CCR4-HEK293 cells to 1x10 6 /mL, and add 20 μL of cell suspension to each reaction well of a 384-well cell culture microplate. Cells were cultured overnight. The next day, discard the supernatant from the cell reaction plate, and add 20 μL of assay buffer and 20 μL of Fluo-4Direct TM loading buffer. from Compound plate (pre-gradiently diluted in 384-well PP microplate) was added to the cell reaction plate with 10 μL of the dilution of the compound to be tested, incubated at 37°C in a 5% carbon dioxide environment for 50 minutes, and then incubated at room temperature for 10 minutes. The reaction plate was transferred to a FLIPR Tetra System (Molecular Devices). Add 10 μL of 6×EC 80 recombinant human CCL22 dilution to the cell reaction plate and shake twice. Read the fluorescence value, use Graphpad software to analyze the data, and calculate the IC 50 of the compound on the calcium flow inhibitory activity of CCR4 receptor cells.
1.3、实验结果1.3. Experimental results
公开的化合物对CCR4受体细胞钙流抑制活性通过以上的试验进行测定,测得的IC50值见表1。实验结果表明,本发明测试化合物对CCR4受体细胞钙流具有显著的抑制活性。The inhibitory activity of the disclosed compounds on CCR4 receptor cell calcium flow was determined by the above tests, and the measured IC 50 values are shown in Table 1. The experimental results show that the test compound of the present invention has significant inhibitory activity on the calcium flow of CCR4 receptor cells.
表1:测试化合物对钙流的抑制活性
Table 1: Inhibitory activity of test compounds on calcium flux
注:比较例1根据WO2018/02299制备;比较例2根据WO2019/147862实施例38制备
Note: Comparative Example 1 was prepared according to WO2018/02299; Comparative Example 2 was prepared according to Example 38 of WO2019/147862
测试例2:CCR4介导的CCRF-CEM细胞趋化抑制活性测试Test Example 2: CCR4-mediated CCRF-CEM Cell Chemotaxis Inhibition Activity Test
2.1、实验材料

2.1. Experimental materials

2.2、实验步骤2.2. Experimental steps
将CCRF-CEM细胞离心,去除上清液,重悬于FBS中,调整细胞密度为2×106/mL,将细胞分液到96孔细胞板中,每孔99μL。在试管中以DMSO为溶剂配制浓度为10mM的化合物储备溶液,然后以DMSO为溶剂,根据需要梯度稀释样品。向96孔板中的细胞每孔转移1μL待测化合物,于37℃,5%二氧化碳的细胞培养箱中孵育0.5小时。以DPBS为溶剂,配制浓度为2.5nM的hCCL22溶液,取出ChemoTX的下层细胞板,将hCCL22分液至细胞板中,每孔30μL。用ChemoTX的微孔膜盖住下层板,从培养箱中取出细胞,每孔转移50μL细胞至微孔膜上,将盖子盖好,于37℃,5%二氧化碳的细胞培养箱中再孵育1.5小时。取出细胞板,取下微孔膜,向下层板中的细胞加入30μL ATP结合剂CellTiter-Glo试剂,用排枪混匀后每孔转移30μL至白色不透明的96孔板中,避光孵育10分钟后,用Envision多模式读板仪读取发光(Luminescence)信号,用Graphpad软件拟合得出化合物对细胞迁移的抑制率,并计算得到IC50Centrifuge the CCRF-CEM cells, remove the supernatant, resuspend in FBS, adjust the cell density to 2×10 6 /mL, and divide the cells into 96-well cell plates, 99 μL per well. Prepare compound stock solutions with a concentration of 10 mM in test tubes using DMSO as a solvent, and then use DMSO as a solvent to serially dilute the samples as required. Transfer 1 μL of the compound to be tested to each well of the cells in the 96-well plate, and incubate for 0.5 hour at 37° C. in a 5% carbon dioxide incubator. Using DPBS as a solvent, prepare a hCCL22 solution with a concentration of 2.5 nM, take out the lower cell plate of ChemoTX, and dispense hCCL22 into the cell plate, 30 μL per well. Cover the lower plate with a ChemoTX microporous membrane, remove the cells from the incubator, transfer 50 μL of cells to the microporous membrane per well, cover the lid, and incubate for another 1.5 hours at 37°C in a 5% carbon dioxide incubator . Take out the cell plate, remove the microporous membrane, add 30 μL ATP binding agent CellTiter-Glo reagent to the cells in the lower plate, mix well with a row gun, transfer 30 μL per well to a white opaque 96-well plate, and incubate in the dark for 10 minutes , using the Envision multi-mode plate reader to read the luminescence (Luminescence) signal, using Graphpad software to fit the inhibitory rate of the compound on cell migration, and calculate the IC 50 .
2.3、实验结果2.3. Experimental results
本发明的化合物对CCR4介导的CCRF-CEM细胞趋化的抑制活性通过以上的试验进行测定,测得的IC50值见表2。实验结果表明,本发明测试化合物对CCRF-CEM细胞趋化具有显著的抑制活性,并且多数化合物抑制活性明显强于比较例1和比较例2。The inhibitory activity of the compounds of the present invention on CCR4-mediated chemotaxis of CCRF-CEM cells was determined by the above tests, and the measured IC 50 values are shown in Table 2. The experimental results show that the test compounds of the present invention have significant inhibitory activity on CCRF-CEM cell chemotaxis, and the inhibitory activity of most compounds is obviously stronger than that of Comparative Example 1 and Comparative Example 2.
表2:测试化合物对CCRF-CEM细胞趋化的抑制活性

Table 2: Inhibitory activity of test compounds on chemotaxis of CCRF-CEM cells

测试例3:CCR4介导的Th2和Treg细胞趋化抑制活性测试Test Example 3: CCR4-mediated Th2 and Treg cell chemotaxis inhibitory activity test
3.1、实验材料
3.1. Experimental materials
3.2、实验步骤3.2. Experimental steps
将hTh2(或hTreg)细胞离心去除培养基,重悬于100%胎牛血清中,调整细胞密度为2×106/mL,将细胞分液到96孔细胞板中,每孔99μL。在试管中以DMSO为溶剂配制浓度为10mM的化合物储备溶液,然后以DMSO为溶剂,根据需要梯度稀释样品。向96孔板中的细胞每孔转移1μL待测化合物,于37℃,5%二氧化碳的细胞培养箱中孵育0.5小时。以DPBS为溶剂,配制浓度为2.5nM的重组人CCL22溶液,取出ChemoTX的下层细胞板,将重组人CCL22分液至细胞板中,每孔30μL。用ChemoTX的微孔膜盖住下层板,从培养箱中取出细胞,每孔转移50μL细胞至微孔膜上,将盖子盖好,于37℃,5%二氧化碳的细 胞培养箱中再孵育1.5小时。取出细胞板,小心取下微孔膜,向下层板中的细胞加入30μL ATP结合剂CellTiter-Glo试剂,用排枪混匀后每孔转移30μL至白色不透明的96孔板中,避光孵育10分钟后,用Envision多模式读板仪读取发光信号,用Graphpad软件拟合得出化合物抑制率。The hTh2 (or hTreg) cells were centrifuged to remove the culture medium, resuspended in 100% fetal bovine serum, adjusted the cell density to 2×10 6 /mL, and split the cells into 96-well cell plates, 99 μL per well. Prepare compound stock solutions with a concentration of 10 mM in test tubes using DMSO as a solvent, and then use DMSO as a solvent to serially dilute the samples as needed. Transfer 1 μL of the compound to be tested to each well of the cells in the 96-well plate, and incubate for 0.5 hour at 37° C. in a 5% carbon dioxide incubator. Using DPBS as a solvent, prepare a recombinant human CCL22 solution with a concentration of 2.5 nM, take out the lower cell plate of ChemoTX, and dispense recombinant human CCL22 into the cell plate, 30 μL per well. Cover the lower plate with a ChemoTX microporous membrane, take out the cells from the incubator, transfer 50 μL of cells per well to the microporous membrane, cover the lid, and store at 37°C in a fine atmosphere of 5% carbon dioxide. Incubate for another 1.5 hours in the incubator. Take out the cell plate, carefully remove the microporous membrane, add 30 μL ATP binding agent CellTiter-Glo reagent to the cells in the lower plate, mix well with a row gun, transfer 30 μL per well to a white opaque 96-well plate, and incubate in the dark for 10 minutes Finally, the luminescent signal was read with the Envision multi-mode plate reader, and the inhibition rate of the compound was obtained by fitting with Graphpad software.
3.3、实验结果3.3. Experimental results
本发明的化合物对CCR4介导的Th2和Treg细胞趋化的抑制活性通过以上的试验进行测定,测得的IC50值见表3。实验结果表明,本发明测试化合物对Th2细胞趋化以及Treg细胞趋化都具有显著的抑制活性。The inhibitory activity of the compounds of the present invention on CCR4-mediated chemotaxis of Th2 and Treg cells was determined by the above tests, and the measured IC 50 values are shown in Table 3. Experimental results show that the test compound of the present invention has significant inhibitory activity on both Th2 cell chemotaxis and Treg cell chemotaxis.
表3:测试化合物对Th2和Treg细胞趋化的抑制活性
Table 3: Inhibitory activity of test compounds on Th2 and Treg cell chemotaxis
测试例4:化合物对细胞表面CCR4受体占据率评价Test Example 4: Evaluation of the occupancy rate of the compound on the cell surface CCR4 receptor
4.1、实验材料
4.1. Experimental materials
4.2、实验步骤4.2. Experimental steps
将CCRF-CEM细胞300rcf离心去除培养基,重悬于100%的胎牛血清中,调整细胞密度为2×106/mL,将细胞分液到96孔细胞板中,每孔99μL。在试管中以DMSO为溶剂配制浓度为10mM的化合物储备溶液,然后以DMSO为溶剂,根据需要梯度稀释样品。化合物每个浓度设置2个孔,向96孔板中每孔转移1μL待测化合物,置于37℃,5%二氧化碳的细胞培养箱中孵育0.5小时。以 DPBS为溶剂,配制浓度为20μM的hCCL22溶液,从细胞培养箱中取出96孔板,化合物每个浓度中的一个孔加入1μL的hCCL22溶液,另一个孔加入1μL的DPBS溶液,再次置于37℃,5%二氧化碳的细胞培养箱中孵育0.5小时。取出细胞板,向每个细胞孔中加入1μL的PE偶联抗人CD194(CCR4)抗体,置于4℃冰箱中避光孵育0.5小时。取出细胞板,4℃300rcf离心去除培养基,向每孔加入200μL提前4℃预冷的DPBS溶液,重悬细胞后再次4℃300rcf离心去除上清液,再次加入200μL的DPBS洗涤一次。离心去除上清后,用预冷的100μL的DPBS重悬细胞。使用Cytek Aurora流式细胞仪检测每孔PE荧光信号的强度,用Graphpad软件拟合得到化合物剂量对CCR4占据率的曲线。The CCRF-CEM cells were centrifuged at 300 rcf to remove the medium, resuspended in 100% fetal calf serum, adjusted the cell density to 2×10 6 /mL, and split the cells into 96-well cell plates, 99 μL per well. Prepare compound stock solutions with a concentration of 10 mM in test tubes using DMSO as a solvent, and then use DMSO as a solvent to serially dilute the samples as required. Two wells were set for each concentration of the compound, 1 μL of the compound to be tested was transferred to each well of the 96-well plate, and placed in a cell culture incubator with 5% carbon dioxide at 37° C. for 0.5 hours. by DPBS was used as the solvent, and hCCL22 solution with a concentration of 20 μM was prepared, and the 96-well plate was taken out of the cell culture incubator, and 1 μL of hCCL22 solution was added to one well of each concentration of the compound, and 1 μL of DPBS solution was added to the other well, and placed at 37 °C again. , and incubate for 0.5 hr in a 5% carbon dioxide incubator. Take out the cell plate, add 1 μL of PE-conjugated anti-human CD194 (CCR4) antibody to each cell well, and incubate in a refrigerator at 4°C in the dark for 0.5 hours. Remove the cell plate, centrifuge at 4°C and 300 rcf to remove the medium, add 200 μL of DPBS solution that was pre-cooled at 4°C to each well, resuspend the cells and centrifuge at 4°C and 300 rcf to remove the supernatant, add 200 μL of DPBS to wash again. After removing the supernatant by centrifugation, resuspend the cells with 100 μL of pre-cooled DPBS. The intensity of PE fluorescence signal in each well was detected by Cytek Aurora flow cytometer, and the curve of compound dose versus CCR4 occupancy was obtained by fitting with Graphpad software.
4.3、实验结果4.3. Experimental results
本发明的化合物对细胞表面CCR4受体占据率通过以上方法进行测定,实验结果见表4。实验结果表明,本发明测试化合物在较低浓度下即可表现出对CCR4较高的占据率,并且在产生相同CCR4占据率的前提下,本发明测试化合物所需浓度明显低于比较例1和比较例2。The occupancy rate of the compound of the present invention on the CCR4 receptor on the cell surface was determined by the above method, and the experimental results are shown in Table 4. The experimental results show that the test compound of the present invention can show a higher occupancy rate of CCR4 at a lower concentration, and under the premise of producing the same CCR4 occupancy rate, the required concentration of the test compound of the present invention is significantly lower than that of Comparative Example 1 and Comparative example 2.
表4:测试化合物对细胞表面CCR4受体占据率评价
Table 4: Evaluation of test compounds on cell surface CCR4 receptor occupancy
测试例5:小鼠药代动力学性质评价Test Example 5: Evaluation of Pharmacokinetic Properties in Mice
5.1、实验方法5.1. Experimental method
测试化合物溶于5%DMSO+5%Solutol+90%食盐水中,涡旋并超声,制备得到相应浓度的澄清溶液,微孔滤膜过滤后备用。选取Balb/c雄性小鼠,静脉注射给予候选化合物溶液,剂量为1mg/kg。受试化合物溶于4%DMSO+96%(0.5%HPMC+0.1%TW-80)或5%DMSO+40%PEG400+10%Solutol+45%水中,涡旋并超声,制备得到相应浓度的澄清溶液,微孔滤膜过滤后备用。选取Balb/c雄性小鼠,口服给予候选化合物溶液,剂量为10mg/kg或50mg/kg。收集一定时间点的全血,制备得到血浆,以LC-MS/MS方法分析药物浓度,并计算药代动力学参数。The test compound was dissolved in 5% DMSO + 5% Solutol + 90% saline, vortexed and sonicated to prepare a clear solution of corresponding concentration, which was filtered through a microporous membrane for use. Balb/c male mice were selected, and the candidate compound solution was administered intravenously at a dose of 1 mg/kg. The test compound was dissolved in 4% DMSO+96% (0.5% HPMC+0.1% TW-80) or 5% DMSO+40% PEG400+10% Solutol+45% water, vortexed and sonicated to prepare the corresponding concentration of clear The solution was filtered through a microporous membrane for later use. Balb/c male mice were selected, and the candidate compound solution was orally administered at a dose of 10 mg/kg or 50 mg/kg. Whole blood was collected at a certain time point, and plasma was prepared, and the drug concentration was analyzed by LC-MS/MS method, and the pharmacokinetic parameters were calculated.
5.2、实验结果5.2. Experimental results
实验结果见表5。实验结果表明,本发明受试化合物具有较好的小鼠体内药代动力学性质。The experimental results are shown in Table 5. The experimental results show that the test compound of the present invention has better pharmacokinetic properties in mice.
表5:测试化合物小鼠体内药代动力学性质

Table 5: Pharmacokinetic properties of test compounds in mice

测试例6:大鼠药代动力学性质评价Test Example 6: Evaluation of Pharmacokinetic Properties in Rats
6.1、实验方法6.1. Experimental method
测试化合物溶于5%DMSO+5%Solutol+90%食盐水中,涡旋并超声,制备得到相应浓度的澄清溶液,微孔滤膜过滤后备用。选取雄性SD大鼠,静脉注射给予候选化合物溶液,剂量为1mg/kg。受试化合物溶于5%DMSO+40%PEG400+10%Solutol+45%水中,涡旋并超声,制备得到相应浓度的澄清溶液,微孔滤膜过滤后备用。选取雄性SD大鼠,口服给予候选化合物溶液,剂量为50mg/kg。收集一定时间点的全血,制备得到血浆,以LC-MS/MS方法分析药物浓度,并计算药代动力学参数。The test compound was dissolved in 5% DMSO + 5% Solutol + 90% saline, vortexed and sonicated to prepare a clear solution of corresponding concentration, which was filtered through a microporous membrane for use. Male SD rats were selected, and the candidate compound solution was administered intravenously at a dose of 1 mg/kg. The test compound was dissolved in 5% DMSO + 40% PEG400 + 10% Solutol + 45% water, vortexed and sonicated to prepare a clear solution of corresponding concentration, which was filtered through a microporous membrane for use. Male SD rats were selected, and the candidate compound solution was orally administered at a dose of 50 mg/kg. Whole blood was collected at a certain time point, and plasma was prepared, and the drug concentration was analyzed by LC-MS/MS method, and the pharmacokinetic parameters were calculated.
6.2、实验结果6.2. Experimental results
实验结果见表6。实验结果表明,本发明受试化合物具有较好的大鼠体内药代动力学性质。The experimental results are shown in Table 6. The experimental results show that the test compound of the present invention has better pharmacokinetic properties in rats.
表6:测试化合物大鼠体内药代动力学性质
Table 6: Pharmacokinetic properties of test compounds in rats
测试例7:比格犬药代动力学性质评价Test Example 7: Evaluation of Pharmacokinetic Properties of Beagle Dogs
7.1、实验方法7.1. Experimental method
测试化合物溶于5%DMSO+5%Solutol+90%食盐水中,涡旋并超声,制备得到相应浓度的澄清溶液,微孔滤膜过滤后备用。选取雄性比格犬,静脉注射给予候选化合物溶液,剂量为1mg/kg。受试化合物溶于5%DMSO+40%PEG400+10%Solutol+45%水中,涡旋并超声,制备得到相应浓度的澄清溶液,微孔滤膜过滤后备用。选取雄性比格犬,口服给予候选化合物溶液,剂量为5mg/kg。收集一定时间点的全血,制备得到血浆,以LC-MS/MS方法分析药物浓度,并计算药代动 力学参数。The test compound was dissolved in 5% DMSO + 5% Solutol + 90% saline, vortexed and sonicated to prepare a clear solution of corresponding concentration, which was filtered through a microporous membrane for use. Male Beagle dogs were selected, and the candidate compound solution was administered intravenously at a dose of 1 mg/kg. The test compound was dissolved in 5% DMSO + 40% PEG400 + 10% Solutol + 45% water, vortexed and sonicated to prepare a clear solution of corresponding concentration, which was filtered through a microporous membrane for use. Male Beagle dogs were selected, and the candidate compound solution was orally administered at a dose of 5 mg/kg. Collect whole blood at a certain time point, prepare plasma, analyze drug concentration by LC-MS/MS method, and calculate pharmacokinetics mechanical parameters.
7.2、实验结果7.2. Experimental results
实验结果见表7。实验结果表明,本发明受试化合物具有较好的比格犬体内药代动力学性质。The experimental results are shown in Table 7. The experimental results show that the test compound of the present invention has better in vivo pharmacokinetic properties in Beagle dogs.
表7:测试化合物比格犬体内药代动力学性质
Table 7: In vivo pharmacokinetic properties of test compounds in Beagle dogs
测试例8:化合物对hERG钾离子通道的抑制活性评价Test Example 8: Evaluation of the Inhibitory Activity of Compounds on the hERG Potassium Ion Channel
8.1、实验材料
8.1. Experimental materials
8.2、实验步骤8.2. Experimental steps
8.2.1、细胞培养和处理8.2.1. Cell culture and treatment
稳定表达hERG的CHO细胞培养于直径35mm的细胞培养皿中,置于37℃,5%CO2的培养箱培养,每48小时按1:5比例进行传代,培养基配方:90%Ham’s F12培养基,10%胎牛血清,100g/mL遗传霉素和100g/mL潮霉素。吸走细胞培养液,用细胞外液淋洗一遍后加入0.25%胰蛋白酶-EDTA溶液,在室温下消化3-5分钟。吸走消化液,用细胞外液重悬后将细胞转移到用于电生理记录的实验皿中备用。CHO cells stably expressing hERG were cultured in a cell culture dish with a diameter of 35 mm, placed in an incubator at 37°C and 5% CO 2 , and subcultured at a ratio of 1:5 every 48 hours. The medium formula: 90% Ham's F12 culture Base, 10% fetal bovine serum, 100g/mL geneticin and 100g/mL hygromycin. Aspirate the cell culture medium, rinse with extracellular fluid once, add 0.25% trypsin-EDTA solution, and digest at room temperature for 3-5 minutes. Aspirate the digestion solution, resuspend the cells with extracellular solution, and transfer the cells to a laboratory dish for electrophysiological recording.
8.2.2、电生理记录过程8.2.2. Electrophysiological recording process
稳定表达hERG钾通道的CHO细胞,在室温下用全细胞电压钳技术记录hERG钾通道电流。灌注电极内液后的尖端电阻为2-5MΩ左右,将玻璃微电极插入放大器探头即可连接至膜片钳放大器。钳制电压和数据记录由pClamp软件通过电脑控制和记录,采样频率为10kHz,滤波频率为2kHz。在得到全细胞记录后,细胞钳制在-100mV,诱发hERG钾电流的步阶电压从-100mV给予一个2s的去极化电压到+20mV,再复极化到-50mV,持续1s后回到-100mV。每5s给予此电压刺激,确定hERG钾电流稳定后(至少1分钟)开始给药过程,化合物每个测试浓度至少给予1分钟至作用稳态。数据分析处理采用pClamp 10,GraphPad和Excel软件。 In CHO cells stably expressing hERG potassium channels, hERG potassium channel currents were recorded by whole-cell voltage-clamp technique at room temperature. The resistance of the tip after perfusion with the inner solution of the electrode is about 2-5MΩ, and the glass microelectrode is inserted into the amplifier probe to connect to the patch clamp amplifier. The clamping voltage and data recording are controlled and recorded by the pClamp software through the computer, the sampling frequency is 10kHz, and the filtering frequency is 2kHz. After obtaining the whole-cell recording, the cell was clamped at -100mV, and the step voltage of the evoked hERG potassium current was given a 2s depolarization voltage from -100mV to +20mV, then repolarized to -50mV, and returned to - for 1s. 100mV. Give this voltage stimulation every 5s, and start the administration process after confirming that the hERG potassium current is stable (at least 1 minute), and give each test concentration of the compound at least 1 minute until the steady state of action. Data analysis and processing used pClamp 10, GraphPad and Excel software.
8.3、实验结果8.3. Experimental results
实验结果见表8。实验结果表明,测试化合物对hERG钾离子通道抑制活性较弱,多数测试化合物IC50值大于10μM,表现出较低的hERG钾离子通路抑制风险。比较例1化合物,对hERG钾离子通道抑制活性较强,IC50值为0.15μΜ。The experimental results are shown in Table 8. The experimental results showed that the test compounds had weak inhibitory activity on the hERG potassium ion channel, and the IC 50 values of most test compounds were greater than 10 μM, showing a low risk of hERG potassium ion channel inhibition. The compound of Comparative Example 1 has strong inhibitory activity on the hERG potassium ion channel, with an IC 50 value of 0.15 μM.
表8:测试化合物对hERG钾离子通道的抑制活性
Table 8: Inhibitory activity of test compounds on hERG potassium ion channel
测试例9:体内FITC诱导的小鼠迟发型超敏反应的药效学实验Test Example 9: Pharmacodynamic experiment of FITC-induced delayed-type hypersensitivity in mice in vivo
9.1、实验材料
9.1. Experimental materials
9.2、实验步骤9.2. Experimental steps
7周龄BALB/c小鼠(维通利华)到达动物房后适应1周。根据体重分为模型对照、阳性对照地塞米松组和待测化合物组。在第0天和第1天,BALB/c小鼠用异氟烷进行麻醉后,进行腹部剃毛,用400μL 0.5%FITC(溶解在50%丙酮+50%邻苯二甲酸二丁酯)进行致敏。在第6、7、8、9天,在小鼠左耳的内、外侧分别涂抹20μL 0.5%FITC进行造模刺激。化合物19和比较例1化合物在溶媒(5%DMSO+40%PEG400+10%Solutol+45%水)中进行溶解,比较例2化合物在溶媒(10%PEG400+90%水)中进行溶解,地塞米松在0.5%MC中进行溶解。第5天及第6、7、8、9天造模刺激前30分钟进行口服给药,一天一次。在第6、7、8、9天FITC刺激前和最后一次刺激24小时后,对左耳用螺旋测微仪进行耳厚测量。所有动物在试验期间每天进行一次称重。所有实验步骤均已通过IACUC评审。7-week-old BALB/c mice (Victoria Lihua) acclimatized for 1 week after arriving at the animal room. According to body weight, they were divided into model control group, positive control dexamethasone group and test compound group. On days 0 and 1, BALB/c mice were anesthetized with isoflurane, and their abdomens were shaved with 400 μL of 0.5% FITC (dissolved in 50% acetone + 50% dibutyl phthalate). sensitization. On days 6, 7, 8, and 9, 20 μL of 0.5% FITC was applied to the inner and outer sides of the left ear of the mice for modeling stimulation. Compound 19 and the compound of Comparative Example 1 were dissolved in the vehicle (5% DMSO+40% PEG400+10% Solutol+45% water), and the compound of Comparative Example 2 was dissolved in the vehicle (10% PEG400+90% water). Semethasone was dissolved in 0.5% MC. On day 5 and day 6, 7, 8, and 9, oral administration was performed 30 minutes before modeling stimulation, once a day. Before FITC stimulation on days 6, 7, 8, and 9 and 24 hours after the last stimulation, ear thickness measurements were performed on the left ear with a spiral micrometer. All animals were weighed once a day during the experiment. All experimental procedures have been reviewed by IACUC.
9.3、实验结果9.3. Experimental results
实验结果见图1和图2。实验结果表明,口服给予化合物19,能够显著抑制该动物模型的耳肿胀。并且,在同等剂量及相同给药途径下,化合物19表现出明显优于比较例1和比较例2的药效作用。 The experimental results are shown in Figure 1 and Figure 2. The experimental results showed that oral administration of Compound 19 could significantly inhibit ear swelling in this animal model. Moreover, at the same dose and the same route of administration, Compound 19 exhibited significantly better pharmacodynamic effects than those of Comparative Example 1 and Comparative Example 2.
测试例10:体内OXA诱导的小鼠特异性皮炎的药效学实验Test Example 10: Pharmacodynamic experiment of OXA-induced mouse atopic dermatitis in vivo
10.1、实验材料
10.1. Experimental materials
10.2、实验步骤10.2. Experimental steps
7周龄BALB/c小鼠(上海吉辉)到达动物房后适应1周。根据体重分为模型对照、阳性对照地塞米松组和待测化合物组。在第0天,BALB/c小鼠用异氟烷进行麻醉后,进行腹部剃毛,用100μL 1.5%OXA(溶解在80%丙酮+20%橄榄油)进行致敏。在第7天,在小鼠左耳的内、外侧分别涂抹20μL 1.5%OXA进行造模刺激。化合物19和比较例1化合物在溶媒(5%DMSO+40%PEG400+10%Solutol+45%水)中进行溶解,比较例2化合物在溶媒(10%PEG400+90%水)中进行溶解,地塞米松在0.5%MC中进行溶解。第6、7天连续给药进行口服给药,一天一次。在第6天和第8天(刺激24小时后)对左耳用螺旋测微仪进行耳厚测量。所有动物在试验期间每天进行一次称重。所有实验步骤均已通过IACUC评审。7-week-old BALB/c mice (Shanghai Jihui) acclimatized for 1 week after arriving in the animal room. According to body weight, they were divided into model control group, positive control dexamethasone group and test compound group. On day 0, BALB/c mice were anesthetized with isoflurane, shaved their abdomen, and sensitized with 100 μL of 1.5% OXA (dissolved in 80% acetone + 20% olive oil). On day 7, 20 μL of 1.5% OXA was applied to the inner and outer sides of the left ear of the mice for modeling stimulation. Compound 19 and the compound of Comparative Example 1 were dissolved in the vehicle (5% DMSO+40% PEG400+10% Solutol+45% water), and the compound of Comparative Example 2 was dissolved in the vehicle (10% PEG400+90% water). Semethasone was dissolved in 0.5% MC. On the 6th and 7th day, continuous administration was performed orally, once a day. Ear thickness measurements were taken with a screw micrometer on the left ear on days 6 and 8 (24 hours after stimulation). All animals were weighed once a day during the experiment. All experimental procedures have been reviewed by IACUC.
10.3、实验结果10.3. Experimental results
实验结果见图3。实验结果表明,口服给予化合物19,能够显著抑制该动物模型的耳肿胀。并且,在同等剂量及相同给药途径下,化合物19表现出明显优于比较例1和比较例2的药效作用。 The experimental results are shown in Figure 3. The experimental results showed that oral administration of Compound 19 could significantly inhibit ear swelling in this animal model. Moreover, at the same dose and the same route of administration, Compound 19 exhibited significantly better pharmacodynamic effects than those of Comparative Example 1 and Comparative Example 2.

Claims (14)

  1. 式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,
    The compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its metabolites,
    其中,in,
    X1和X2各自独立地选自C或N;X 1 and X 2 are each independently selected from C or N;
    X3、X4和X5各自独立地选自CRb或N;X 3 , X 4 and X 5 are each independently selected from CR b or N;
    R1各自独立地选自H、卤素、C1-C3烷基磺酰基、C1-C3烷基、C1-C3卤代烷基、C1-C3烷氧基、C3-C4环烷基或氰基;Each R 1 is independently selected from H, halogen, C 1 -C 3 alkylsulfonyl, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, C 1 -C 3 alkoxy, C 3 -C 4 cycloalkyl or cyano;
    R2选自H、C1-C3烷基、C1-C3卤代烷基或C3-C4环烷基;R 2 is selected from H, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl or C 3 -C 4 cycloalkyl;
    R3选自H、C1-C3烷基、C1-C3卤代烷基或C3-C4环烷基;R 3 is selected from H, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl or C 3 -C 4 cycloalkyl;
    R4选自 R4 is selected from
    Y选自CH2或O;Y is selected from CH2 or O;
    Ra选自H、卤素、C1-C6烷基、C1-C6卤代烷基、C1-C6烷氧基、C3-C6环烷基、4-7元杂环基、5-6元杂芳基、苯基或氰基,其中所述C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-7元杂环基、5-6元杂芳基或苯基各自独立地任选地被选自卤素、羟基、氰基、C1-C3烷基或C1-C3烷氧基的一个或多个基团所取代;R a is selected from H, halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 3 -C 6 cycloalkyl, 4-7 membered heterocyclyl, 5-6 membered heteroaryl, phenyl or cyano, wherein said C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 6 cycloalkyl, 4-7 membered heterocyclic , 5-6 membered heteroaryl or phenyl are each independently optionally selected from one or more groups selected from halogen, hydroxyl, cyano, C 1 -C 3 alkyl or C 1 -C 3 alkoxy replaced by
    每个Rb独立地选自H、卤素、C1-C3烷基、C1-C3烷氧基、C3-C6环烷基或氰基,其中所述C1-C3烷基、C1-C3烷氧基或C3-C6环烷基各自独立地任选地被选自卤素、羟基、氨基、氰基、C1-C3烷基或C1-C3烷氧基的一个或多个基团所取代;Each R b is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyl or cyano, wherein the C 1 -C 3 alk radical, C 1 -C 3 alkoxy or C 3 -C 6 cycloalkyl each independently optionally selected from halogen, hydroxy, amino, cyano, C 1 -C 3 alkyl or C 1 -C 3 One or more groups of alkoxy are substituted;
    每个Rc独立地选自H、卤素、C1-C3烷基或C3-C4环烷基,其中所述C1-C3烷基或C3-C4环烷基各自独立地任选地被选自卤素、羟基、氰基、C1-C3烷基或C1-C3烷氧基的一个或多个基团所取代;Each R c is independently selected from H, halogen, C 1 -C 3 alkyl or C 3 -C 4 cycloalkyl, wherein each of the C 1 -C 3 alkyl or C 3 -C 4 cycloalkyl is independently optionally substituted by one or more groups selected from halogen, hydroxyl, cyano, C 1 -C 3 alkyl or C 1 -C 3 alkoxy;
    每个Rd独立地选自H、卤素、C1-C3烷基、C1-C3烷氧基、C3-C6环烷基或氰基,其中所述C1-C3烷基、C1-C3烷氧基或C3-C6环烷基各自独立地任选地被选自卤素、羟基、氨基、氰基、C1-C3烷基或C1-C3烷氧基的一个或多个基团所取代; Each R d is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyl or cyano, wherein the C 1 -C 3 alk radical, C 1 -C 3 alkoxy or C 3 -C 6 cycloalkyl each independently optionally selected from halogen, hydroxy, amino, cyano, C 1 -C 3 alkyl or C 1 -C 3 One or more groups of alkoxy are substituted;
    每个Re独立地为-L-Rfeach Re is independently -LR f ;
    每个Rf独立地选自H、卤素、C1-C3烷基、C1-C3烷氧基、C1-C3卤代烷基、C3-C6环烷基、4-7元杂环基、RgC(O)NH-、RgNHC(O)-、RgOC(O)NH-、RgS(O)2NH-、RgNHS(O)2-、RgS(O)2-、-(CH2)n-羟基、-(CH2)n-氨基、-(CH2)n-氰基、-(CH2)n-羧基或2-氧杂-螺[3,3]庚烷基,其中所述C1-C3烷基、C1-C3烷氧基、C3-C6环烷基、4-7元杂环基、-(CH2)n-羟基、-(CH2)n-氨基、-(CH2)n-氰基或-(CH2)n-羧基各自独立地任选地被选自卤素、羟基、氨基、氰基、C1-C3烷基或C1-C3烷氧基的一个或多个基团所取代;Each R f is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 1 -C 3 haloalkyl, C 3 -C 6 cycloalkyl, 4-7 membered Heterocyclyl, R g C(O)NH-, R g NHC(O)-, R g OC(O)NH-, R g S(O) 2 NH-, R g NHS(O) 2 -, R g g S(O) 2 -, -(CH 2 ) n -hydroxyl, -(CH 2 ) n -amino, -(CH 2 ) n -cyano, -(CH 2 ) n -carboxy or 2-oxa- Spiro[3,3]heptyl, wherein the C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyl, 4-7 membered heterocyclyl, -(CH 2 ) n -hydroxyl, -(CH 2 ) n -amino, -(CH 2 ) n -cyano or -(CH 2 ) n -carboxyl are each independently optionally selected from halogen, hydroxyl, amino, cyano , C 1 -C 3 alkyl or one or more groups of C 1 -C 3 alkoxy;
    每个Rg独立地选自C1-C3烷基、C3-C6环烷基或3-6元杂环基;Each R g is independently selected from C 1 -C 3 alkyl, C 3 -C 6 cycloalkyl or 3-6 membered heterocyclyl;
    每个L独立地选自化学键、4-7元亚杂环基、C1-C3亚烷基或C3-C6亚环烷基,其中所述4-7元亚杂环基、C1-C3亚烷基或C3-C6亚环烷基各自独立地任选地被选自卤素、羟基、氨基、氰基、C1-C3烷基或C1-C3烷氧基的一个或多个基团所取代;Each L is independently selected from a chemical bond, a 4-7 membered heterocyclic group, a C 1 -C 3 alkylene group or a C 3 -C 6 cycloalkylene group, wherein the 4-7 membered heterocyclic group, C 1 -C 3 alkylene or C 3 -C 6 cycloalkylene are each independently optionally selected from halogen, hydroxy, amino, cyano, C 1 -C 3 alkyl or C 1 -C 3 alkoxy Substituted by one or more groups of the group;
    Z1为0至3的任一整数;Z 1 is any integer from 0 to 3;
    Z2为0至3的任一整数;Z 2 is any integer from 0 to 3;
    Z3为0至4的任一整数;Z 3 is any integer from 0 to 4;
    Z4为0至2的任一整数;并且Z 4 is any integer from 0 to 2; and
    每个n独立地为0至3的任一整数。Each n is independently any integer from 0 to 3.
  2. 根据权利要求1所述的式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,其中,According to claim 1, the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its metabolite, wherein,
    X1和X2中的一个为N,另一个为C,One of X1 and X2 is N, the other is C,
    X3、X4和X5中的至少一个为N。At least one of X 3 , X 4 and X 5 is N.
  3. 根据权利要求1或2所述的式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,其中,
    选自
    According to claim 1 or 2, the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrug, or its metabolite, wherein,
    selected from
  4. 根据权利要求1-3中任一项所述的式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受 的盐、或其前体药、或其代谢物,其为式(II)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,
    The compound of formula (I) or its isotope-labeled compound according to any one of claims 1-3, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical Acceptable A salt, or a prodrug thereof, or a metabolite thereof, which is a compound of formula (II) or an isotope-labeled compound thereof, or an optical isomer, a geometric isomer, a tautomer or a mixture of isomers thereof, or a pharmaceutically acceptable salt thereof, or a prodrug thereof, or a metabolite thereof,
    其中,R1-R3、X1-X5、Y、Ra、Rc、Rd、Re、Z1、Z2、Z3和Z4如权利要求1中所定义;Wherein, R 1 -R 3 , X 1 -X 5 , Y, R a , R c , R d , R e , Z 1 , Z 2 , Z 3 and Z 4 are as defined in claim 1;
    特别地,其中,
    选自 和/或
    选自 Rc、Rd、Re、Z2和Z3如权利要求1中所定义;
    In particular, among them,
    selected from and / or
    selected from Rc , Rd , Re , Z2 and Z3 are as defined in claim 1;
    更特别地,其为式(II-1)、式(II-2)、式(II-3)、式(II-4)、式(II-5)、式(II-6)、式(II-7)或式(II-8)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,
    More particularly, it is formula (II-1), formula (II-2), formula (II-3), formula (II-4), formula (II-5), formula (II-6), formula ( II-7) or formula (II-8) compound or its isotope labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutically acceptable salt, or its prodrugs, or its metabolites,
    其中,R1-R3、Ra、Rd、Re、Z1和Z3如权利要求1中所定义。Wherein, R 1 -R 3 , R a , R d , R e , Z 1 and Z 3 are as defined in claim 1.
  5. 根据权利要求1-3中任一项所述的式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,其为式(III)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,
    The compound of formula (I) or its isotope-labeled compound according to any one of claims 1-3, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical An acceptable salt, or a prodrug thereof, or a metabolite thereof, which is a compound of formula (III) or an isotope-labeled compound thereof, or an optical isomer, geometric isomer, tautomer or isomer thereof body mixture, or a pharmaceutically acceptable salt thereof, or a prodrug thereof, or a metabolite thereof,
    其中,R1-R3、Ra、Re和Z1如权利要求1中所定义。Wherein, R 1 -R 3 , R a , R e and Z 1 are as defined in claim 1.
  6. 根据权利要求1-3中任一项所述的式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,其为式(IV)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,
    The compound of formula (I) or its isotope-labeled compound according to any one of claims 1-3, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical An acceptable salt, or a prodrug thereof, or a metabolite thereof, which is a compound of formula (IV) or an isotope-labeled compound thereof, or an optical isomer, geometric isomer, tautomer or isomer thereof body mixture, or a pharmaceutically acceptable salt thereof, or a prodrug thereof, or a metabolite thereof,
    其中,R1-R3、Ra、Re和Z1如权利要求1中所定义。Wherein, R 1 -R 3 , R a , R e and Z 1 are as defined in claim 1.
  7. 根据权利要求1-6中任一项所述的式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,其中,According to any one of claims 1-6, the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical Acceptable salts, or prodrugs thereof, or metabolites thereof, wherein,
    每个Re独立地为-L-Rfeach Re is independently -LR f ;
    Rf各自独立地选自H、卤素、C1-C3烷基、C1-C3烷氧基、C1-C3卤代烷基、C3-C6环烷基、4-7元杂环基、RgC(O)NH-、RgNHC(O)-、RgOC(O)NH-、RgS(O)2NH-、RgNHS(O)2-、RgS(O)2-、-(CH2)n-羟基、-(CH2)n-氨基、-(CH2)n-氰基、-(CH2)n-羧基或2-氧杂-螺[3,3]庚烷基,其中所述C1-C3烷基、C1-C3烷氧基、C3-C6环烷基、4-7元杂环基、-(CH2)n-羟基、-(CH2)n-氨基、-(CH2)n-氰基或-(CH2)n-羧基各自独立地任选地被选自羟基或C1-C3烷基的一个或多个基团所取代;优选地Rf各自独立地选自H、氟、氯、溴、甲基、乙基、正丙基、异丙基、二氟甲基、三氟甲基、羟基、羧基、甲氧基、氧杂环丁烷基、氮杂环丁烷基、1,1-二氧代硫杂环丁烷基、吗啉基、吡咯烷基、哌啶基、四氢吡喃基、乙酰胺基、甲磺酰基、CH3S(O)2NH-或2-氧杂-螺[3,3]庚烷基;Each R f is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 1 -C 3 haloalkyl, C 3 -C 6 cycloalkyl, 4-7 membered hetero Cyclo, R g C(O)NH-, R g NHC(O)-, R g OC(O)NH-, R g S(O) 2 NH-, R g NHS(O) 2 -, R g S(O) 2 -, -(CH 2 ) n -hydroxyl, -(CH 2 ) n -amino, -(CH 2 ) n -cyano, -(CH 2 ) n -carboxy or 2-oxa-spiro [3,3] Heptyl, wherein the C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyl, 4-7 membered heterocyclyl, -(CH 2 ) n -hydroxyl, -(CH 2 ) n -amino, -(CH 2 ) n -cyano or -(CH 2 ) n -carboxyl are each independently optionally selected from hydroxyl or C 1 -C 3 alkyl Substituted by one or more groups; preferably each R f is independently selected from H, fluorine, chlorine, bromine, methyl, ethyl, n-propyl, isopropyl, difluoromethyl, trifluoromethyl , hydroxyl, carboxyl, methoxy, oxetanyl, azetidinyl, 1,1-dioxothietanyl, morpholinyl, pyrrolidinyl, piperidinyl, tetra Hydropyranyl, acetamido, methanesulfonyl, CH 3 S(O) 2 NH- or 2-oxa-spiro[3,3]heptyl;
    Rg选自C1-C3烷基、C3-C4环烷基或3-4元杂环基,优选地选自甲基、乙基、 环丙烷基、环丁烷基或氧杂环丁烷基;R g is selected from C 1 -C 3 alkyl, C 3 -C 4 cycloalkyl or 3-4 membered heterocyclic group, preferably selected from methyl, ethyl, Cyclopropyl, cyclobutanyl or oxetanyl;
    每个n独立地为0至3的任一整数,并且each n is independently any integer from 0 to 3, and
    L选自化学键、任选地被甲基或乙基取代的亚甲基、亚乙基、亚丙基、亚环丙基、亚环丁基、亚环戊基、亚环己基、亚氮杂环丁烷基、亚吡咯烷基或亚哌啶烷基;L is selected from the group consisting of chemical bond, methylene optionally substituted by methyl or ethyl, ethylene, propylene, cyclopropylene, cyclobutylene, cyclopentylene, cyclohexylene, azepine Cyclobutane, pyrrolidinylene or piperidinylene;
    特别地,In particular,
    每个Re独立地选自H、甲基、乙基、正丙基、异丙基、环丙基、 Each Re is independently selected from H, methyl, ethyl, n-propyl, isopropyl, cyclopropyl,
  8. 根据权利要求1-7中任一项所述的式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,其中,The compound of formula (I) or its isotope-labeled compound according to any one of claims 1-7, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical Acceptable salts, or prodrugs thereof, or metabolites thereof, wherein,
    R1各自独立地选自H、卤素、C1-C3烷基磺酰基、C1-C3烷基、C1-C3卤代烷基或C1-C3烷氧基,优选地选自H、氟、氯、溴、甲基、乙基、正丙基、异丙基或甲磺酰基;和/或Each R 1 is independently selected from H, halogen, C 1 -C 3 alkylsulfonyl, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl or C 1 -C 3 alkoxy, preferably selected from H, fluorine, chlorine, bromine, methyl, ethyl, n-propyl, isopropyl or methanesulfonyl; and/or
    R2选自H或C1-C3烷基,优选地选自H或甲基;和/或R is selected from H or C 1 -C 3 alkyl, preferably from H or methyl; and/or
    R3选自H或C1-C3烷基,优选地选自H或甲基。R 3 is selected from H or C 1 -C 3 alkyl, preferably from H or methyl.
  9. 根据权利要求1-8中任一项所述的式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,其中,According to any one of claims 1-8, the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical Acceptable salts, or prodrugs thereof, or metabolites thereof, wherein,
    Ra各自独立地选自H、卤素、C1-C3烷基、C1-C3卤代烷基、C1-C3烷氧基、C3-C4环烷基或氰基,其中所述C1-C3烷基、C1-C3烷氧基或C3-C4环烷基各自独立地任选被卤素、羟基、氰基、C1-C3烷基或C1-C3烷氧基取代,Ra优选地选自H、氟、氯、溴、甲基、乙基、异丙基、环丙基、二氟甲基、三氟甲基、三氟乙基、 甲氧基、乙氧基、羟甲基、1,1-二甲基羟甲基、氰基或甲氧基亚甲基;和/或Each R a is independently selected from H, halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, C 1 -C 3 alkoxy, C 3 -C 4 cycloalkyl or cyano, wherein The C 1 -C 3 alkyl, C 1 -C 3 alkoxy or C 3 -C 4 cycloalkyl are each independently optionally replaced by halogen, hydroxyl, cyano, C 1 -C 3 alkyl or C 1 - C3alkoxy substitution, R is preferably selected from H, fluorine, chlorine, bromine, methyl, ethyl, isopropyl, cyclopropyl, difluoromethyl, trifluoromethyl, trifluoroethyl, Methoxy, ethoxy, hydroxymethyl, 1,1-dimethylhydroxymethyl, cyano, or methoxymethylene; and/or
    Rb各自独立地选自H、卤素、C1-C3烷基、C3-C4环烷基或氰基,其中所述C1-C3烷基或C3-C4环烷基各自独立地任选被卤素、羟基、氨基、氰基或C1-C3烷基取代,Rb优选地选自H、氟、氯、溴、甲基、乙基、异丙基、环丙基、二氟甲基、三氟甲基、三氟乙基或氰基;和/或Each R b is independently selected from H, halogen, C 1 -C 3 alkyl, C 3 -C 4 cycloalkyl or cyano, wherein the C 1 -C 3 alkyl or C 3 -C 4 cycloalkyl Each is independently optionally substituted by halogen, hydroxyl, amino, cyano or C 1 -C 3 alkyl, R b is preferably selected from H, fluorine, chlorine, bromine, methyl, ethyl, isopropyl, cyclopropyl group, difluoromethyl, trifluoromethyl, trifluoroethyl or cyano; and/or
    Rc各自独立地选自H、卤素或C1-C3烷基,优选地选自H、氟、氯、甲基或乙基;和/或Each R c is independently selected from H, halogen or C 1 -C 3 alkyl, preferably selected from H, fluorine, chlorine, methyl or ethyl; and/or
    Rd各自独立地选自H、卤素、C1-C3烷基或氰基,优选地选自H、氟、氯、溴、甲基、乙基、正丙基、异丙基或氰基。Each R d is independently selected from H, halogen, C 1 -C 3 alkyl or cyano, preferably selected from H, fluorine, chlorine, bromine, methyl, ethyl, n-propyl, isopropyl or cyano .
  10. 根据权利要求1-9中任一项所述的式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,其中,According to any one of claims 1-9, the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical Acceptable salts, or prodrugs thereof, or metabolites thereof, wherein,
    Z1为2;和/或Z 1 is 2; and/or
    Z2为0或1;和/或 Z2 is 0 or 1; and/or
    Z3为0或1;和/或 Z3 is 0 or 1; and/or
    Z4为1;和/或Z 4 is 1; and/or
    每个n独立地为0或1。Each n is independently 0 or 1.
  11. 根据权利要求1-10中任一项所述的式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,其选自:



    According to any one of claims 1-10, the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical An acceptable salt, or a prodrug thereof, or a metabolite thereof, selected from:



  12. 一种药物组合物,其包含根据权利要求1-11中任一项所述的式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物,以及药学上可接受的载体。A pharmaceutical composition comprising the compound of formula (I) according to any one of claims 1-11 or its isotope-labeled compound, or its optical isomers, geometric isomers, tautomers or A mixture of isomers, or a pharmaceutically acceptable salt thereof, or a prodrug thereof, or a metabolite thereof, and a pharmaceutically acceptable carrier.
  13. 根据权利要求1-11中任一项所述的式(I)化合物或其同位素标记化合物、或其光学异构体、几何异构体、互变异构体或异构体混合物、或其药学上可接受的盐、或其前体药、或其代谢物在制备作为CCR4拮抗剂的药物中的用途,特别地,所述药物用于治疗或预防由CCR4介导的疾病或病症。According to any one of claims 1-11, the compound of formula (I) or its isotope-labeled compound, or its optical isomer, geometric isomer, tautomer or isomer mixture, or its pharmaceutical Use of an above-acceptable salt, or a prodrug thereof, or a metabolite thereof in the preparation of a medicament as a CCR4 antagonist, in particular, the medicament is used for treating or preventing a disease or condition mediated by CCR4.
  14. 根据权利要求13所述的用途,其中所述由CCR4介导的疾病或病症选自特应性皮炎、哮喘、过敏性鼻炎、异位性皮炎、系统性红斑狼疮、类风湿性关节 炎中的一种或多种免疫相关性疾病;The use according to claim 13, wherein the disease or disease mediated by CCR4 is selected from atopic dermatitis, asthma, allergic rhinitis, atopic dermatitis, systemic lupus erythematosus, rheumatoid arthritis One or more immune-related diseases in inflammation;
    或者,or,
    所述由CCR4介导的疾病或病症为癌症。 The disease or condition mediated by CCR4 is cancer.
PCT/CN2023/072403 2022-01-25 2023-01-16 Ccr4 small molecule antagonist and use thereof WO2023143194A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202380016094.9A CN118475581A (en) 2022-01-25 2023-01-16 CCR4 small molecule antagonist and application thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202210087598 2022-01-25
CN202210087598.7 2022-01-25
CN202210928357 2022-08-03
CN202210928357.0 2022-08-03

Publications (1)

Publication Number Publication Date
WO2023143194A1 true WO2023143194A1 (en) 2023-08-03

Family

ID=87470522

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/072403 WO2023143194A1 (en) 2022-01-25 2023-01-16 Ccr4 small molecule antagonist and use thereof

Country Status (3)

Country Link
CN (1) CN118475581A (en)
TW (1) TW202340197A (en)
WO (1) WO2023143194A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1592624A (en) * 2000-06-30 2005-03-09 惠氏公司 Substituted-triazolopyrimidines as anticancer agents
CN101268038A (en) * 2005-09-22 2008-09-17 塞诺菲-安万特股份有限公司 Amino-alkyl-amide derivatives as CCR3 receptor ligands
WO2008146914A1 (en) * 2007-06-01 2008-12-04 Mitsubishi Tanabe Pharma Corporation Heterocyclic compound
US20090131666A1 (en) * 2006-03-24 2009-05-21 Astellas Pharma Inc. Acylaminopiperidine compound
CN103974950A (en) * 2011-12-01 2014-08-06 凯莫森特里克斯股份有限公司 Substituted benzimidazoles and benzopyrazoles as CCR(4) antagonists
CN111971279A (en) * 2018-01-26 2020-11-20 拉普特医疗公司 Chemokine receptor modulators and uses thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1592624A (en) * 2000-06-30 2005-03-09 惠氏公司 Substituted-triazolopyrimidines as anticancer agents
CN101268038A (en) * 2005-09-22 2008-09-17 塞诺菲-安万特股份有限公司 Amino-alkyl-amide derivatives as CCR3 receptor ligands
US20090131666A1 (en) * 2006-03-24 2009-05-21 Astellas Pharma Inc. Acylaminopiperidine compound
WO2008146914A1 (en) * 2007-06-01 2008-12-04 Mitsubishi Tanabe Pharma Corporation Heterocyclic compound
CN103974950A (en) * 2011-12-01 2014-08-06 凯莫森特里克斯股份有限公司 Substituted benzimidazoles and benzopyrazoles as CCR(4) antagonists
CN111971279A (en) * 2018-01-26 2020-11-20 拉普特医疗公司 Chemokine receptor modulators and uses thereof

Also Published As

Publication number Publication date
CN118475581A (en) 2024-08-09
TW202340197A (en) 2023-10-16

Similar Documents

Publication Publication Date Title
EP3932919B1 (en) Jak inhibitor compound and use thereof
JP6948659B1 (en) Pyridadinyl thiaazole carboxamide compound
JP6494624B2 (en) Substituted 4,5,6,7-tetrahydropyrazolo [1,5-A] pyrazine derivatives as casein kinase 1D / E inhibitors
EP3674307B1 (en) Macrocycle containing aminopyrazole and pyrimidine and pharmaceutical composition and use thereof
CN114349750A (en) KRAS mutein inhibitors
EP2315521B1 (en) Diamino-pyridine, pyrimidine, and pyridazine modulators of the histamine h 4 receptor
CN112552295A (en) KRAS mutein inhibitors
EP3766882A1 (en) Phthalazine isoxazole alkoxy derivatives, preparation method thereof, pharmaceutical composition and use thereof
WO2023041055A1 (en) Kif18a inhibitor
EP3971176A1 (en) Aromatic amine ar and bet targeting protein degradation chimera compound and use
JP2023536891A (en) JAK inhibitor compounds for treating severe pneumonia
CN113121509A (en) JAK inhibitor compounds and uses thereof
WO2016091207A1 (en) Nitromidazole compound, preparation method therefor and use thereof in drug manufacturing
WO2023143194A1 (en) Ccr4 small molecule antagonist and use thereof
CN111315750B (en) Pyridopyrimidines as mTORC1/2 dikinase inhibitors
WO2022135390A1 (en) Ketohexokinase inhibitor and use thereof
WO2024094190A1 (en) Irak4 degradation agent and use thereof
WO2024120424A1 (en) Compound targeting pan-kras protein degradation agent and use thereof
RU2779497C2 (en) Macrocycle containing aminopyrazole and pyrimidine, and its pharmaceutical composition and use
WO2024193464A1 (en) Nitrogen-containing tricyclic derivative and use thereof in medicine
WO2023143384A1 (en) Compound for inhibiting or degrading hpk1 kinase and medical use thereof
WO2024175024A1 (en) New fused heterocyclic compound as cdks inhibitor and use thereof
WO2024125532A1 (en) Novel heterocyclic compound acting as cdks inhibitor, and use thereof
WO2023151635A1 (en) Compound based on quinazoline-substituted glutarimide skeleton and use thereof
TW202311260A (en) Sulfoximine-containing ATR inhibitor compound

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: 23746076

Country of ref document: EP

Kind code of ref document: A1