[go: up one dir, main page]

WO2025092973A1 - Parp1-targeting compound and use thereof - Google Patents

Parp1-targeting compound and use thereof Download PDF

Info

Publication number
WO2025092973A1
WO2025092973A1 PCT/CN2024/129351 CN2024129351W WO2025092973A1 WO 2025092973 A1 WO2025092973 A1 WO 2025092973A1 CN 2024129351 W CN2024129351 W CN 2024129351W WO 2025092973 A1 WO2025092973 A1 WO 2025092973A1
Authority
WO
WIPO (PCT)
Prior art keywords
mmol
compound
methyl
int
compound according
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
PCT/CN2024/129351
Other languages
French (fr)
Chinese (zh)
Inventor
贾涛
王太津
李钢
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chengdu Zenitar Biomedical Technology Co Ltd
Original Assignee
Chengdu Zenitar Biomedical Technology Co Ltd
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 Chengdu Zenitar Biomedical Technology Co Ltd filed Critical Chengdu Zenitar Biomedical Technology Co Ltd
Publication of WO2025092973A1 publication Critical patent/WO2025092973A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • 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
    • 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
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • C07D491/14Ortho-condensed systems
    • C07D491/147Ortho-condensed systems the condensed system containing one ring with oxygen as ring hetero atom and two rings with nitrogen as ring hetero atom

Definitions

  • the present invention belongs to the field of chemical medicine and relates to a class of PARP1 targeting compounds and uses thereof.
  • PARP inhibitors have shown excellent clinical efficacy in patients with homologous recombination-deficient cancers. However, hematological toxicity (anemia, neutropenia, and thrombocytopenia) and other toxicities limit the application of these drugs, whether used as a single agent or in combination therapy.
  • Related studies have shown that (Harris P A, Boloor A, Cheung M, et al. Discovery of 5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2-methyl-benzenesulfonamide (Pazopanib), a novel and potent vascular endothelial growth factor receptor inhibitor. [J].
  • the object of the present invention is to provide a class of PARP1 targeted compounds and uses thereof, which have unexpectedly higher selectivity for PARP1 than other PARP family members (such as PARP2, PARP3, PARP5a and PARP6), and can achieve highly selective and highly effective prevention or treatment of diseases related to PARP function; in particular, the PARP1 targeted compounds of the present invention are surprisingly able to penetrate the blood-brain barrier (BBB); therefore, the PARP1 targeted compounds of the present invention can be used to treat diseases and conditions occurring in central nervous system tissues (such as the brain and spinal cord).
  • BBBB blood-brain barrier
  • the present invention provides a compound of formula I or a pharmaceutically acceptable form thereof, the structure of which is as follows:
  • X is selected from N or C, and when X is selected from N, represents a single bond, when X is selected from C, represents a double bond;
  • X1 is selected from N or C( R8a ), X2 is selected from N or C( R8b ), X3 is selected from N or C( R8c ), and at most one of X1 , X2 and X3 is selected from N;
  • R1 is selected from C1-8 alkylthio, C1-8 halogenated alkylthio, C1-8 deuterated alkylthio, C1-8 halogenated alkoxy, C1-8 halogenated alkyl or C2-8 alkenyl;
  • R2 is selected from hydrogen, fluorine, chlorine, C1-4 alkyl, C1-4 fluoroalkyl or C1-4 deuterated alkyl;
  • R 3 and R 4 are independently selected from hydrogen, deuterium, fluorine, C 1-4 alkyl, C 1-4 fluoroalkyl or C 1-4 deuterated alkyl;
  • R 3 , R 4 together with the atoms to which they are attached, form a 3-6 membered alkyl ring;
  • R 5 is selected from hydrogen, fluorine, chlorine, cyano, C 1-4 alkyl, C 1-4 fluoroalkyl or C 1-4 deuterated alkyl;
  • R 4 , R 5 and the atoms to which they are connected together form a 5-7 membered alkyl heterocycle; when R 4 , R 5 and the atoms to which they are connected are connected to form a ring, the 5-7 membered alkyl heterocycle contains 1 O heteroatom in addition to X on the main ring;
  • R 3 , R 4 and the atoms to which they are attached, and R 4 , R 5 and the atoms to which they are attached, will not form a ring at the same time;
  • R 6 is selected from -C(O)-NH-R 6a ;
  • R 6a is selected from C 1-4 alkyl, C 1-4 fluoroalkyl, C 1-4 deuterated alkyl or 3-6 membered cycloalkyl;
  • R7 is selected from hydrogen, fluorine, chlorine, cyano, C1-4 alkyl, C1-4 fluoroalkyl or C1-4 deuterated alkyl;
  • R 8a , R 8b , and R 8c are independently selected from hydrogen, fluorine, chlorine, C 1-4 alkyl, C 1-4 deuterated alkyl, and C 1-4 fluoroalkyl;
  • the pharmaceutically acceptable form is selected from pharmaceutically acceptable salts, esters, stereoisomers, tautomers, polymorphs, solvates, N-oxides, isotopically labeled substances, metabolites or prodrugs.
  • X1 is selected from N
  • X2 is selected from C( R8b )
  • X3 is selected from C( R8c ); or X1 is selected from C( R8a ), X2 is selected from N, and X3 is selected from C( R8c ); or X1 is selected from C( R8a ), X2 is selected from C( R8b ), and X3 is selected from C( R8c ).
  • R1 is selected from C1-4 alkylthio, C1-4 haloalkylthio, C1-4 deuterated alkylthio, C1-4 haloalkoxy, C1-4 haloalkyl, C2-4 alkenyl; in R1 , the halogen is selected from F, Cl, and Br.
  • R 1 is selected from methylthio, halogenated methylthio, deuterated methylthio, halogenated methoxy, halogenated methyl, halogenated ethyl or vinyl; in R 1 , the halogenated halogen is selected from F and Cl.
  • R2 is selected from hydrogen, fluorine, chlorine, methyl, fluoromethyl or deuterated methyl.
  • R 3 and R 4 when R 3 and R 4 do not form a ring with the atoms to which they are attached, R 3 and R 4 are independently selected from hydrogen, deuterium, fluorine, methyl, fluoromethyl or deuterated methyl.
  • R 5 is selected from hydrogen, fluorine, chlorine, cyano, methyl, fluoromethyl or deuterated methyl.
  • R 7 is selected from hydrogen, fluorine, chlorine, cyano, methyl, fluoromethyl or deuterated methyl.
  • R 8a , R 8a , R 8c are independently selected from hydrogen, fluorine, chlorine, methyl, fluoromethyl or deuterated methyl.
  • R 6a is selected from methyl, fluoromethyl, deuterated methyl or cyclopropyl.
  • R 6a is selected from methyl, -CF 3 , -CD 3 or cyclopropyl.
  • the structural unit (connected to the main ring from left to right, that is, the N end is connected to the main ring methylene, and the X end is connected to the main ring pyridyl) is selected from:
  • the structural unit in formula I Replace with (Both are connected to the main ring in the same way, that is, the N end is connected to the main ring methylene, and the O end is connected to the main ring pyridyl) (In this case, R 5 does not form a ring with the connected atoms).
  • the present invention also provides some specific compounds, which are selected from:
  • the structural unit in formula I Replace with (Both are connected to the main ring in the same way, that is, the N end is connected to the methylene group of the main ring, and the CH end is connected to the pyridyl group of the main ring) (In this case, R 5 does not form a ring with the connected atoms).
  • the present invention also provides some specific compounds, which are selected from:
  • the structural unit in formula I Replace with (Connected to the main ring from left to right, that is, the N-terminal is connected to the methylene group of the main ring, and the alkynyl end is connected to the pyridyl group of the main ring).
  • the structural unit in formula I Replace with R9 is selected from H.
  • the present invention also provides some specific compounds, which are selected from:
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the aforementioned compound or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, nitrogen oxide, isotope-labeled substance, metabolite or prodrug as an active ingredient, supplemented with a pharmaceutically acceptable carrier.
  • a further object of the present invention is to provide a method for preparing the pharmaceutical composition of the present invention, which comprises combining any compound of Formula I or a pharmaceutically acceptable form thereof, or a mixture thereof, with one or more pharmaceutically acceptable carriers.
  • the pharmaceutically acceptable carrier that can be used in the pharmaceutical composition of the present invention is a pharmaceutically acceptable carrier.
  • suitable pharmaceutically acceptable carriers are described in Remington’s Pharmaceutical Sciences (2005).
  • the pharmaceutical composition can be administered in any form as long as it prevents, alleviates, prevents or cures the symptoms of a human or animal patient.
  • it can be prepared into various suitable dosage forms according to the administration route.
  • the administration of the compounds or pharmaceutical compositions of the present invention may be combined with another therapeutic method.
  • the other therapeutic method may be selected from, but not limited to: radiotherapy, chemotherapy, immunotherapy, or a combination thereof.
  • the present invention also relates to a pharmaceutical preparation comprising any compound of formula I or a pharmaceutically acceptable form thereof, or a mixture thereof as an active ingredient, or a pharmaceutical composition of the present invention.
  • the preparation is in the form of a solid preparation, a semisolid preparation, a liquid preparation or a gaseous preparation.
  • the present invention provides the use of the aforementioned compounds, compounds of formula I, and related specific compounds or pharmaceutically acceptable forms thereof, or the pharmaceutical compositions of the present invention in the preparation of drugs for preventing or treating PARP1 enzyme-related diseases.
  • the present invention provides a method for preventing or treating PARP1 enzyme-related diseases, comprising administering a compound of formula I or a pharmaceutically acceptable form thereof, or a pharmaceutical composition of the present invention to an individual in need thereof.
  • the present invention provides a compound of formula I or a pharmaceutically acceptable form thereof, or a pharmaceutical composition of the present invention for use in preventing or treating PARP1 enzyme-related diseases.
  • the present invention provides a method for preventing or treating PARP1 enzyme-related diseases in combination with a compound of formula I or a pharmaceutically acceptable form thereof or a pharmaceutical composition of the present invention, wherein the additional treatment method includes but is not limited to: radiotherapy, chemotherapy, immunotherapy, or a combination thereof.
  • the PARP1 enzyme-related disease is a disease that is sensitive or responsive to PARP1 enzyme inhibition.
  • the PARP1 enzyme-related disease is a tumor disorder.
  • the tumor-like disorder lacks the HR-dependent DNA DSB repair pathway.
  • the tumor-like disorder comprises one or more cancer cells having reduced or absent ability to repair DNA DSB via HR relative to normal cells.
  • the cancer cells have a BRCA1 or BRCA2 deficient phenotype.
  • the PARP1 enzyme-related disease is a tumor-like disorder, including but not limited to solid and hematological malignancies.
  • the tumor-like disorder includes but is not limited to breast cancer, colorectal cancer, colon cancer, lung cancer (including small cell lung cancer, non-small cell lung cancer and bronchioloalveolar carcinoma) and prostate cancer, as well as bile duct cancer, bone cancer, bladder cancer, head and neck cancer, kidney cancer, liver cancer, gastrointestinal tissue cancer, esophageal cancer, ovarian cancer, pancreatic cancer, skin cancer, testicular cancer, thyroid cancer, uterine cancer, cervical cancer and vulvar cancer, as well as leukemia (including chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL) and chronic myeloid leukemia (CML)), multiple myeloma, lymphoma, meningioma, pituitary tumor, craniopharyngioma, schwannoma, gli
  • CLL chronic lympho
  • the PARP1 enzyme-related disease is breast cancer, ovarian cancer, pancreatic cancer, prostate cancer, blood cancer, gastrointestinal cancer, lung cancer or glioma.
  • the compounds of the present invention can be used in combination with chemoradiotherapy or immunotherapy to prevent or treat cancer.
  • the present invention provides the use of the aforementioned compounds, compounds of formula I, and related specific compounds or pharmaceutically acceptable forms thereof, or the pharmaceutical compositions of the present invention in the preparation of PARP1 inhibitors.
  • the present invention provides a novel class of highly active and highly selective PARP1 inhibitors, which can achieve at least one of the following technical effects: (1) high inhibitory activity against PARP1 enzyme; (2) selective inhibition of PARP1 enzyme, with high selectivity for other PARP family enzymes such as PARP2, PARP5a, and PARP5b; (3) strong inhibitory activity against homologous recombination-deficient tumor cells, and weak inhibitory effect on non-homologous recombination-deficient cells; (4) excellent pharmacokinetic properties (e.g., good bioavailability, suitable half-life and duration of action); (5) excellent safety (lower toxicity and/or fewer side effects, wider therapeutic window), etc.
  • the PARP1 targeted compounds described in the present invention are surprisingly able to penetrate the blood-brain barrier (BBB) and can be used to treat diseases and conditions occurring in central nervous system tissues (e.g., brain and spinal cord).
  • BBB blood-brain barrier
  • C 1-8 should be understood to include any sub-ranges and each point value therein, such as C 2-5 , C 3-4 , C 1-2 , C 1-3 , C 1-4 , C 1-5 , etc., as well as C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , etc.
  • the expression “C 2-8 " should also be understood in a similar manner, for example, it can include any sub-ranges and point values contained therein, such as C 6-8 , C 6-7 , C 7-8 , etc., as well as C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , etc.
  • 3-6 yuan should be understood to include any sub-ranges and each point value therein, such as 3-4 yuan, 3-5 yuan, 3-6 yuan, 4-5 yuan, 4-6 yuan, etc., as well as 3, 4, 5, 6 yuan, etc.
  • halogen means fluorine, chlorine, bromine or iodine.
  • alkyl includes a linear or branched monovalent saturated hydrocarbon group.
  • alkyl includes methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 3-(2-methyl)butyl, 2-pentyl, 2-methylbutyl, neopentyl, n-hexyl, 2-hexyl, 2-methylpentyl, etc.
  • C1-4 in “ C1-4 alkyl” refers to a group containing 1, 2, 3 or 4 carbon atoms in a linear or branched form.
  • cycloalkyl refers to a saturated or partially saturated, monocyclic or polycyclic (such as bicyclic) non-aromatic hydrocarbon group.
  • Common cycloalkyl groups include (but are not limited to) monocyclic cycloalkyl groups, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclobutene, cyclopentene, cyclohexene, etc.; or bicyclic cycloalkyl groups, including fused rings, bridged rings or spiro rings, such as bicyclo[1.1.1]pentyl, bicyclo[2.2.1]heptyl, bicyclo[3.2.1]octyl, bicyclo[5.2.0]nonyl, decalinyl, etc.
  • monocyclic cycloalkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cycl
  • C 3-12 cycloalkyl refers to a cycloalkyl group having 3-12 ring carbon atoms (such as 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12).
  • the cycloalkyl or cycloalkylene group in the present invention is optionally substituted by one or more substituents described in the present invention.
  • haloalkyl refers to an alkyl group as described above, in which one or more hydrogen atoms are present. Atoms are replaced by halogen.
  • C 1-6 haloalkyl refers to a C 1-6 alkyl group optionally substituted by one or more (e.g., 1-3) halogens. It will be appreciated by those skilled in the art that when there are more than one halogen substituent, the halogens may be the same or different and may be located on the same or different C atoms.
  • haloalkyl groups include, for example, -CH 2 F, -CHF 2 , -CF 3 , -CCl 3 , -C 2 F 5 , -C 2 Cl 5 , -CH 2 CF 3 , -CH 2 Cl or -CH 2 CH 2 CF 3.
  • the haloalkyl group in the present invention is optionally substituted by one or more substituents described herein.
  • fluoroalkyl refers to the alkyl group described above, wherein one or more hydrogen atoms are replaced by fluorine atoms.
  • C 1-4 fluoroalkyl refers to a C 1-4 alkyl group optionally substituted by one or more (e.g., 1-3) fluorine atoms. It will be understood by those skilled in the art that when there are more than one fluorine atom substituents, the fluorine atoms may be the same or different, and may be located on the same or different C atoms.
  • haloalkyl groups include, for example, -CH 2 F, -CHF 2 , -CF 3 , -C 2 F 5 , -CH 2 CF 3 , -CH 2 CH 2 CF 3 , etc.
  • the fluoroalkyl group in the present invention is optionally substituted by one or more substituents described in the present invention.
  • C2-4 alkenyl refers to an alkenyl group having 2 to 4 carbon atoms.
  • Common alkenyl groups include (but are not limited to) vinyl, propenyl, n-butenyl, 3-methylbut-2-enyl, n-pentenyl, n-octenyl, n-decenyl, etc.
  • the alkenyl group in the present invention is optionally substituted by one or more substituents described in the present invention.
  • substituted means that one or more hydrogen atoms in the group are replaced by the same or different substituents, respectively.
  • the present invention also includes all pharmaceutically acceptable isotopically labeled compounds, which are identical to the compounds of the present invention except that one or more atoms are replaced by atoms having the same atomic number but an atomic mass or mass number different from the atomic mass or mass number predominant in nature.
  • isotopes suitable for inclusion in the compounds of the present invention include, but are not limited to, isotopes of hydrogen (e.g., deuterium ( 2H ), tritium ( 3H )); isotopes of carbon (e.g., 13C and 14C ); isotopes of chlorine (e.g., 37Cl); isotopes of iodine (e.g., 125I ); isotopes of nitrogen (e.g., 13N and 15N ); isotopes of oxygen (e.g., 17O and 18O ); isotopes of phosphorus (e.g., 32P ); and isotopes of sulfur (e.g., 34S ).
  • isotopes of hydrogen e.g., deuterium ( 2H ), tritium ( 3H )
  • isotopes of carbon e.g., 13C and 14C
  • isotopes of chlorine e.g.
  • pharmaceutical composition refers to a preparation of the compound of the present invention and a medium generally accepted in the art for delivering biologically active compounds to mammals (e.g., humans).
  • the medium includes a pharmaceutically acceptable carrier.
  • the purpose of the pharmaceutical composition is to promote administration of the organism, facilitate the absorption of the active ingredient, and thus exert biological activity.
  • pharmaceutically acceptable carrier includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersant, suspending agent, stabilizer, isotonic agent, solvent or emulsifier approved or accepted by relevant governmental regulatory authorities for use in humans or livestock.
  • drug combination refers to medical treatments obtained by mixing or combining more than one active ingredient, including fixed and non-fixed combinations of active ingredients.
  • fixed combination refers to the simultaneous administration of at least one compound described herein and at least one synergistic agent to a patient in the form of a single entity or a single dosage form.
  • non-fixed combination refers to the simultaneous administration, combined administration or sequential administration of at least one compound described herein and at least one synergistic agent to a patient in the form of separate entities, at the same time, in combination or at variable intervals. These also apply to cocktail therapies, for example, the administration of three or more active ingredients.
  • tumor includes but is not limited to leukemia, gastrointestinal stromal tumor, histiocytic lymphoma, non-small cell lung cancer, small cell lung cancer, pancreatic cancer, squamous cell carcinoma of the lung, adenocarcinoma of the lung, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, cervical cancer, ovarian cancer, intestinal cancer, rhinitis cancer, brain cancer, bone cancer, esophageal cancer, melanoma, kidney cancer, oral cancer, brain cancer and central nervous system cancer.
  • treating means reversing, alleviating, inhibiting the progression of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.
  • FIG1 is a curve showing the change in tumor volume in a NOD-SCID mouse subcutaneous tumor model of human breast cancer 436 cells after administration of compounds 47 and 64.
  • Figure 2 is a physical picture of the change in tumor volume in the human breast cancer 436 cell NOD-SCID mouse subcutaneous tumor model after administration of compounds 47 and 64.
  • FIG3 is a graph showing the changes in tumor volume in the breast cancer MDA-MB-436 nude mouse model after administration of compound 11.
  • FIG4 is a graph showing the effect of compound 11 administration on the intracranial fluorescence intensity of the human breast cancer MDA-MB-436 mouse model.
  • FIG6 is a graph showing the effect of compound 11 administration on the body weight of human breast cancer MDA-MB-436 mice.
  • the reagents and raw materials used in the examples of the present invention are commercially available.
  • the structures of the compounds of the present invention are determined by nuclear magnetic resonance (NMR) or mass spectrometry (MS). NMR measurements are performed using a Bruker AVANCE-400 nuclear magnetic spectrometer, with deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD) as the solvent, tetramethylsilane (TMS) as the internal standard, and chemical shifts are given in units of 10 -6 (ppm).
  • DMSO-d 6 deuterated dimethyl sulfoxide
  • CDCl 3 deuterated chloroform
  • CD 3 OD deuterated methanol
  • TMS tetramethylsilane
  • MS was measured using an Agilent SQD (ESI) mass spectrometer (manufacturer: Agilent, signal: 6110).
  • HPLC determination used Agilent 1200DAD high pressure liquid chromatograph (Sunfirc C18, 150X 4.6mm, 5wn, chromatographic column) and Waters 2695-2996 high pressure liquid chromatograph (Gimini C18, 150X 4.5mm, 5ym chromatographic column).
  • the thin layer chromatography silica gel plate used was Qingdao Ocean GF254 silica gel plate.
  • the silica gel plate used in thin layer chromatography (TLC) had a specification of 0.15mm-0.2mm, and the thin layer chromatography separation and purification product used a 0.4mm-0.5mm silica gel plate.
  • the reactions were carried out under an argon atmosphere or a nitrogen atmosphere.
  • Argon atmosphere or nitrogen atmosphere means that the reaction bottle is connected to an argon or nitrogen balloon with a volume of about 1 L.
  • Hydrogen atmosphere means that the reaction bottle is connected to a hydrogen balloon with a volume of about 1 L.
  • the hydrogenation reaction is usually evacuated, filled with hydrogen, and the operation is repeated 3 times.
  • Step 1 Add int-1a (500g, 1.99mol) and 2.5L of methanol under nitrogen protection, and cool to 0-5°C. Add a solution of sodium methoxide (118g) in methanol (1.0L) dropwise at 0-5°C. After complete addition, warm to room temperature and stir for 1 hour. Add water (2.0L) to the reaction system, stir for 30min, and then concentrate under reduced pressure at 40°C until no liquid is discharged. Then add ethyl acetate (4.0L), stir and separate, add ethyl acetate to the aqueous layer for extraction, and separate the layers. Combine the organic layers, add saturated brine to wash, separate the layers, and concentrate the organic layer under reduced pressure at 40°C to obtain int-1b (480g) as a white solid.
  • int-1b 480g
  • Step 2 Weigh compound int-1b (475 g, 1.93 mol) and add it to DMF (2.85 L), then add DMF-DMA (2.85 L) dropwise. After adding, heat to 100 °C and stir for 2 h. After the reaction is complete, cool to 70-80 °C, concentrate under reduced pressure until no liquid is produced, then add to water and stir to precipitate. Cool to 20-30 °C, stir for 1 h, then filter, and dry the filter cake in a vacuum oven at 70 °C to constant weight to obtain a red solid int-1c (612 g, yield 95.3%).
  • Step 3 Add compound int-1c (500g, 1.91mol) to THF (2.56L) and stir to dissolve. Add an aqueous solution (2.56L) of sodium periodate (805g, 3.72mol) to the reaction system. Stir at room temperature for 2-4h. After the reaction is completed, add ethyl acetate (4.0L) and water (4.0L) to the reaction system, stir and separate the layers, and extract the aqueous layer with ethyl acetate (2.0L) twice. After the organic layers are combined, add saturated sodium thiosulfate solution and saturated brine to wash in turn. The organic layer is concentrated under reduced pressure at 40-45°C until there is no fraction to obtain 500g of oily int-1d, which is directly used in the next step.
  • Step 4 Add compounds int-1d (512 g, 1.69 mol) and int-1e (1457.0 g, 7.61 mol) to anhydrous ethanol (7.5 L), stir to dissolve, and add SnCl 2 (1815.0 g, 9.57 mol) to the reaction system in batches at room temperature. After the addition, heat to reflux and stir for 1-2 h. Cool the reaction system to 45-50 ° C, concentrate under reduced pressure until there is no fraction, add ethyl acetate to the system and stir to dissolve, and then adjust the pH to 7-8 with saturated sodium bicarbonate. During the process, gas is released violently and a large amount of solid is precipitated. The reaction solution is centrifuged, and the filtrate is collected and allowed to stand for stratification. The organic layer is concentrated under reduced pressure at 40-45 ° C until there is no fraction, and 200-300 mesh silica gel is added to purify by column to obtain flocculent solid int-1 (230 g, yield 36.5%).
  • Step 1 Add compound int-2a (1g, 4.6mmol), int-2b (1.7g, 5.5mmol), Pd(dppf)Cl 2 (0.3g, 0.46mmol) and potassium carbonate (1.6g, 11.5mmol) to a mixed solvent of 7ml dioxane, 3ml anhydrous ethanol and 4ml water, then replace nitrogen three times, and react at 90°C for 2h under nitrogen protection. After TLC detection, the reaction is cooled to room temperature, 30ml dichloromethane and 20ml water are added, and the layers are separated in a separatory funnel.
  • the aqueous phase is extracted twice with dichloromethane, and the organic phases are combined and then washed with water, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and dried.
  • the crude product is purified by column chromatography to obtain compound int-2c (1g, white solid).
  • Step 2 Add int-2c (1 g, 3 mmol), methylamine aqueous solution (5 g, 161.3 mmol) and anhydrous methanol (20 ml) to a 100 ml reaction bottle and stir overnight at room temperature. After the reaction is complete as monitored by TLC, the reaction solution is concentrated under reduced pressure to dryness to obtain compound int-2d (0.8 g, white solid).
  • Step 3 Add compound int-2d (0.5 g, 1.5 mmol) into 10 ml of anhydrous methanol, and then add 10 ml of 4 mol/L hydrochloric acid dioxane solution. Stir at room temperature for 0.5-1 h. After the reaction is complete as monitored by TLC, concentrate the reaction solution under reduced pressure to obtain compound int-2 (0.5 g, white solid).
  • Step 1 Weigh int-2c (1.4 g, 4.5 mmol), add 20 mL MeOH to dissolve, add 5 mL water, add lithium hydroxide (570 mg, 13.5 mmol), react at room temperature for 12 h, monitor the reaction by TLC, add 2 M HCl to adjust the pH to 6 after the reaction is complete, add (3 x 25 mL) EA to extract, combine the organic phases, dry over anhydrous sodium sulfate, and vacuum dry. The product was spin dried in vacuo to obtain int-5a (460 mg, light yellow solid).
  • Step 2 Weigh int-5a (180 mg, 0.6 mmol), EDCI (144 mg, 0.75 mmol), HOBT (100 mg, 0.75 mmol) 2 mL DMF, N-methylmorpholine (290 mg, 3.2 mmol), cyclopropylamine (34 mg, 0.6 mmol), react at warm temperature for 12 h, monitor by TLC, dilute with water after the raw materials are completely consumed, extract with EA, wash the organic phase with water 5 times, dry over anhydrous sodium sulfate, and spin dry to obtain the crude product int-5b (180 mg, yellow oily liquid).
  • Step 1 Add compound int-2a (5.66 g, 26.21 mmol, 1.05 eq), compound int-9a (4.65 g, 25.0 mmol, 1.00 eq), Cs 2 CO 3 (16.27 g, 50.0 mmol, 2.00 eq) and RuPhos-Pd-G3 (1.04 g, 1.25 mmol, 0.05 eq) to 1,4-dioxane (50 mL) and stir overnight at 120 ° C under nitrogen atmosphere. After the reaction is completed by LCMS, the mixture is allowed to cool to room temperature. The reaction solution is diluted with water (100 mL) and then extracted with ethyl acetate (2 x 100 mL).
  • Step 1 Weigh int-9b (1.4 g, 4.5 mmol), add 20 mL MeOH to dissolve, add 5 mL water, add lithium hydroxide (570 mg, 13.5 mmol), react at room temperature for 12 h, monitor the reaction by TLC, add 2 M HCl to adjust the pH to 6 after the reaction is complete, add (3x 25 mL) EA to extract, combine the organic phases, dry over anhydrous sodium sulfate, and vacuum dry to obtain the product int-12a (845 mg).
  • the preparation method of intermediates int-13 to int-15 refers to intermediate int-12.
  • triphenylphosphine (20.54 g, 0.0783 mol)
  • DEAD 11.36 g, 0.06525 mol
  • LC-MS: ESI [M+H] + 333.2.
  • Step 3 Dissolve the compound int-17d (0.61 g, 1.84 mmol) in DCM (20 mL), slowly add DIBAL-H (1.0 M, 5.0 mL, 5.0 mmol) at 0°C, and stir at 0°C for 1 h. Slowly pour the reaction solution into 50 mL of saturated ammonium chloride solution to quench, filter with diatomaceous earth, extract with 4*30 mL of EA, and dry with sodium sulfate to obtain a yellow oily compound int-17e (0.45 g, crude).
  • LC-MS: ESI [M+H] + 291.1.
  • Step 1 Place compound int-1 (5.0 g, 16.07 mmol) in a sealed tube and add DMSO (50 mL) to dissolve. After solution, CuI (3.0 g, 15.7 mmol) and Zn(OAc) 2 (6.0 g, 32.78 mmol) were added, and N 2 was replaced three times, and then stirred at 150°C for 15 h.
  • Step 2 Dissolve compound int-18a (0.3 g, 1.08 mmol) in DCM (20 mL), slowly add DIBAL-H (1.0 M, 5.0 mL, 5.0 mmol) at 0°C, and stir at 0°C for 1 h. Slowly pour the reaction solution into 50 mL saturated ammonium chloride solution to quench, filter with diatomaceous earth, extract with 4*30 mL EA, and dry with sodium sulfate to obtain yellow oily compound int-18b (0.3 g, crude).
  • LC-MS: ESI [M+H] + 237.2.
  • Step 3 Dissolve compound int-18b (0.3 g, 1.26 mmol) in DCM (10 mL), add TEA (0.5 mL, 4.32 mmol) and MsCl (0.3 mL, 2.62 mmol) dropwise at 0°C, and stir at 0°C for 1 h. Slowly pour 20 ml of saturated sodium bicarbonate solution into the reaction solution to quench, extract with 3*30 ml of DCM, dry with sodium sulfate, filter and concentrate under reduced pressure to obtain compound int-18 (0.3 g, crude).
  • LC-MS: ESI [M+H] + 315.2.
  • Step 1 Dissolve compound int-19a (4.2 g, 19.4 mmol) in dichloromethane (100 mL), add triethylamine (117 mg, 38.9 mmol), tert-butyldimethylsilyl chloride (2.17 g, 14.39 mmol), 4-dimethylaminopyridine (3.94 g, 0.94 mmol), stir and react for 14 hours, concentrate the reaction solution under reduced pressure, and purify the residue by silica gel column chromatography to obtain compound int-19b (5.2 g).
  • LC-MS: ESI [M+H] + 331.2.
  • Step 3 Dissolve compound int-19c (1 g, 2.06 mmol) in tetrahydrofuran (6 mL), add tetrabutylammonium fluoride tetrahydrofuran solution (1 M/L, 6 mL), stir and react for 2 hours, and concentrate the reaction solution under reduced pressure to obtain the crude title compound int-19d (722 mg, yield: 99%), which is used directly in the next step without purification.
  • LC-MS: ESI [M+H] + 350.2.
  • Step 4 Dissolve the crude product compound int-19d (722 mg, 2.06 mmol) in a solution of methylamine (8 mL, 25-30 wt% aqueous solution) in methanol (7 mL), stir and react for 2 hours, and concentrate the reaction solution under reduced pressure to obtain the crude title compound int-19e (700 mg, yield: 97%), which is used directly in the next step without purification.
  • LC-MS: ESI [M+H] + 349.2.
  • the first step Weigh the compound int-20a (0.25 g, 0.83 mmol), add 20 ml of tetrahydrofuran, replace nitrogen, cool to -78 ° C, add lithium diisopropylamide (0.46 mL, 0.91 mmol) dropwise, stir for 30 minutes, add N-phenylbis(trifluoromethanesulfonyl)imide (0.36 g, 1.0 mmol) dissolved in 10 mL of tetrahydrofuran dropwise, warm to room temperature and react for 2 hours. After TLC detection, the reaction is complete, saturated ammonium chloride aqueous solution is added to quench, and extracted three times with ethyl acetate. After the organic phases are combined, they are dried over anhydrous sodium sulfate, filtered, and spin-dried. Purification by column chromatography gives compound int-20b (0.25 g, colorless liquid).
  • Step 2 Add compound int-20b (0.25 g, 0.6 mmol), compound int-20c (0.14 g, 0.66 mmol), Pd(dppf)Cl 2 (0.04 g, 0.06 mmol) and potassium carbonate (0.16 g, 1.2 mmol) to a mixed solvent of 20 ml dioxane and 2 ml water, then replace nitrogen, and react at 80°C for 3 hours under nitrogen protection. After TLC detection, the reaction is cooled to room temperature, concentrated by rotary evaporation, and extracted three times by adding 30 ml ethyl acetate and 20 ml water.
  • Step 1 Sodium chlorodifluoroacetate int-22a (2.88 g, 18.9 mmol, 2 eq) and K 2 CO 3 (1.95 g, 14.1 mmol, 1.5 eq) were added to a 100 mL two-necked flask and dried under high vacuum for 1 hour. DMF (99%, 30 mL) was added at room temperature, and int-17b (2.5 g, 9.43 mmol) was added in batches. The reaction mixture was stirred at 95° C. for 15 minutes (exothermic reaction) and cooled to room temperature. Water (130 mL) and ethyl acetate (130 mL) were added.
  • Step 2 Dissolve compound int-22b (0.58 g, 1.84 mmol) in DCM (20 mL), slowly add DIBAL-H (1.0 M, 5.0 mL, 5.0 mmol) at 0°C, and stir at 0°C for 1 h. Slowly pour the reaction solution into 50 mL saturated ammonium chloride solution to quench, filter with diatomaceous earth, extract with 4*30 mL EA, and dry with sodium sulfate to obtain yellow oily compound int-22c (0.42 g, crude).
  • LC-MS: ESI [M+H] + 273.1.
  • Step 3 Dissolve compound int-12c (0.42 g, 1.55 mmol) in DCM (30 mL), add TEA (0.7 mL, 7.36 mmol) and MsCl (0.5 mL, 3.6 mmol) dropwise at 0°C, and stir at 0°C for 1 h. Slowly pour 30 ml of saturated sodium bicarbonate solution into the reaction solution to quench, extract with 3*30 ml of DCM, dry with sodium sulfate, filter and concentrate under reduced pressure to obtain compound int-22 (0.5 g, crude).
  • LC-MS: ESI [M+H] + 351.2.
  • Step 1 Dissolve compound int-24a (0.85 g, 4.564 mmol) and compound int-9a (1.0 g, 4.149 mmol) in dioxane (40 mL), add RuPphos-Pd-G 3 (0.35 g, 0.415 mmol), Cs 2 CO 3 (5.41 g, 16.595 mmol), replace with N 2 three times, and stir at 100°C for 15 h.
  • Step 1 Dissolve compound int-25a (1 g, 3.102 mmol) in DCM (30 mL), add compound int-25b (0.36 g, 3.723 mmol), HATU (1.42 g, 3.723 mmol), DIPEA (1.20 g, 9.307 mmol), and stir at 25°C for 1 h.
  • Step 2 Dissolve compound int-25c (1.2 g, 2.989 mmol) in MeOH (20 mL) and add Pd/C 10% (1.2 g, 11.276 mmol), H 2 replaced 3 times, stirred at 25°C for 1 h. Silica gel was filtered off under reduced pressure, and dried under reduced pressure to obtain compound int-25d (1.1 g, 2.726 mmol, 91.21%) as a light yellow solid.
  • LC-MS: ESI [M+H] + 404.2.
  • Step 1 Dissolve compound int-26a (1 g, 2.9 mmol) in THF (10 mL), H 2 O (10 mL), add LiOH (0.62 g, 14.7 mmol), and stir at 0°C for 2 h. Pour the reaction solution into saturated aqueous ammonium chloride solution (30 mL). Extract with DCM (3*30 mL), dry with anhydrous sodium sulfate, and concentrate under reduced pressure to obtain compound int-26b (1 g, 3.1 mmol, crude) as a white solid.
  • LC-MS: ESI [M+H] + 326.2.
  • Step 2 Compound int-26b (1 g, 3.1 mmol) was dissolved in DCM (30 mL), and compound int-25b (0.36 g, 3.723 mmol), HATU (1.42 g, 3.7 mmol), and DIPEA (1.20 g, 9.3 mmol) were added, and stirred at 25°C for 1 h.
  • Step 3 Dissolve compound int-26c (1.1 g, 2.7 mmol) in HCl/MeOH (10 mL, 4.0 M) and stir at 25°C for 0.5 h. Concentrate under reduced pressure to obtain compound int-26 (0.8 g, 2.6 mmol, 96.65%) as a white solid.
  • LC-MS: ESI [M+H] + 305.2.
  • Step 1 Dissolve compound int-25a (1 g, 3.102 mmol) in MeOH (15 mL), add Pd/C 10% (1 g, 9.397 mmol), replace with H 2 three times, and stir at 25°C for 1 h. Filter through silica gel pad, and dry the filtrate under reduced pressure to obtain compound int-27a (1 g, 3.083 mmol, 99.38%) as a light yellow solid.
  • LC-MS: ESI [M+H] + 325.2.
  • Step 3 Dissolve compound int-27b (1.0 g, 2.752 mmol) in dioxane hydrochloride solution (10 mL, 4.0 M) and stir at 25°C for 1 h. Concentrate under reduced pressure to obtain compound int-27 (0.7 g, 2.658 mmol, 96.61%) as a white solid.
  • LC-MS: ESI [M+H] + 264.3.
  • N,6-dimethyl-5-(piperidin-4-yl)picolinamide refers to the intermediate int-23.
  • LC-MS: ESI [M+H] + 234.2.
  • Step 1 Dissolve compound int-31a (2 g, 9.899 mmol) and compound int-9a (2.21 g, 11.878 mmol) in 1,4-dioxane (50 mL), add Ruphos-Pd-G3 (0.83 g, 0.990 mmol), Cs 2 CO 3 (9.68 g, 29.696 mmol), replace with N 2 three times, and stir at 100°C for 12 h.
  • Step 2 Dissolve compound int-31b (1.7 g, 5.530 mmol) in methanolic hydrochloric acid solution (20 mL) and stir at 25°C for 1 h. Concentrate under reduced pressure to obtain compound int-31 (1.1 g, 5.307 mmol, 95.96%) as a white solid.
  • LC-MS: ESI [M+H] + 208.2.
  • Step 1 Add (R)-3-(Hydroxymethyl)piperazine-1-carboxylic acid tert-butyl ester (5 g, 23.118 mmol) to DMF (50 mL), add p-methoxybenzyl chloride (3.62 g, 23.118 mmol) and potassium carbonate (9.58 g, 69.355 mmol), raise the temperature to 60 ° C and react for 24 hours. After the reaction is completed, water is added to quench the reaction, and ethyl acetate is added for extraction. The organic phase is washed with water 3 times, and the organic phase is concentrated under reduced pressure.
  • Step 2 Add compound int-32b (6 g, 17.834 mmol) and 6-bromo-2-(bromomethyl)-3-fluoropyridine (4.80 g, 17.834 mmol) to DMF (100 mL), cool to 0°C, add sodium hydrogen sulfide (1.07 g, 26.751 mmol), react at room temperature for 12 h, and when the reaction is complete, add water to quench the reaction, add ethyl acetate for extraction, and wash the organic phase with water for 3 hours.
  • Step 3 Compound int-32d (4.2 g, 8.009 mmol), PdCl 2 (PPh 3 ) 2 (1.12 g, 1.602 mmol) and DIEA (3.11 g, 24.026 mmol) were added to DMF (30 mL) and ethanol (30 mL), and the temperature was raised to 90°C for reaction for 12 h under the protection of carbon monoxide. The reaction solution was added to water, and ethyl acetate was added for extraction. The organic phase was washed with water for 3 times, and the organic phase was concentrated under reduced pressure.
  • LC-MS: ESI [M+H] + 398.2.
  • Step 5 Add compound int-32f (85 mg, 0.214 mmol) and DIEA (82.93 mg, 0.642 mmol) to NMP (3 mL), heat to 180°C and stir for 6 hours. After the reaction is completed, add the reaction solution to water, extract with ethyl acetate, wash the organic phase with water three times, and concentrate the organic phase under reduced pressure to obtain compound int-32g (80 mg, yield: 99.11%).
  • LC-MS: ESI [M+H] + 378.2.
  • Step 6 Add compound int-32g (80 mg, 0.212 mmol) and methylamine alcohol solution (2 mL) into methanol (2 mL), stir at room temperature for 1 h, and after the reaction, concentrate the reaction solution under reduced pressure to obtain crude compound int-32h (80 mg, yield: 100%).
  • LC-MS: ESI [M+H] + 363.4.
  • Step 5 Add compound int-33e (2.0 g, 6.223 mmol) to dichloromethane (20 mL), add trifluoroacetic acid (5 mL), react at room temperature for 1 h, and after the reaction is completed, concentrate under reduced pressure and collect the product to obtain compound int-33 trifluoroacetate.
  • LC-MS: ESI [M+H] + 222.1.
  • Step 3 Add compound int-34c (86 mg, 0.258 mmol) into the bottle, dissolve in DCM (10 mL), add trifluoroacetic acid (2 mL, 26.118 mmol), stir at room temperature for 5 h, and spin dry to obtain the crude trifluoroacetate of compound int-34 (130 mg), which was used directly in the next step without further purification.
  • LC-MS: [M+H]+ 234.1.
  • LC-MS: [M+H] + 331.2.
  • Step 2 Add compound int-37b (529 mg, 1.601 mmol) to the reaction flask, dissolve in methanol (15 mL), add methylamine (15 mL), stir at room temperature for 5 h, and spin dry the reaction solution to obtain crude compound int-37c (529 mg) as a light yellow oil.
  • LC-MS: [M+H] + 330.2, without further purification, directly used in the next step.
  • Step 3 Compound int-37c (529 mg, 1.606 mmol) was dissolved in DCM (20 mL), TFA (5 mL, 65.296 mmol) was added, and the mixture was stirred at room temperature for 2 h. The reaction solution was spin-dried to obtain a crude product of compound int-37 (1.25 g) as a yellow oil (containing TFA), which was used directly in the next step without further purification.
  • LC-MS: [M+H] + 230.2.
  • Step 4 Add compound int-38d (118 mg, 0.354 mmol) to the bottle, dissolve in DCM (10 mL), add TFA (3 mL, 39.177 mmol), stir at room temperature for 2 h, LC-MS shows: the reaction is complete, the reaction solution is concentrated under reduced pressure to obtain compound int-38 (181 mg, containing TFA) as a colorless oil, which is used directly in the next step without further purification.
  • LC-MS: [M+H] + 234.2.
  • Example 1 1'-((7-(1,1-difluoroethyl)-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)-N-methyl-1',2',3',6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide
  • Step 1 Dissolve compound int-1 (1.80 g, 1.61 mmol) in dioxane (30 mL), add XPhos-Pd-G 2 (0.40 g, 0.51 mmol), tributyl (1-ethoxyethylene) tin (2.0 g, 11.6 mmol), heat at 80 ° C and stir for 2 h under N 2 atmosphere. Pour the reaction solution into 30 mL saturated sodium bicarbonate solution, extract 5*30 mL with ethyl acetate, dry with sodium sulfate, filter and concentrate under reduced pressure, add THF (10 mL), HCl (3.0 M, 20 mL), and stir for 12 h.
  • LC-MS: ESI[M+H] + 297.2.
  • LC-MS: ESI [M+H] + 241.2.
  • Step 7 Compound 1f (0.05 g, 0.09 mmol) was dissolved in MeOH (5 mL), KOH (5 mL, 3.0 M) was added and stirred at 25°C for 1 h. 30 ml of saturated ammonium chloride solution was slowly poured into the reaction solution to quench, 4*30 ml of EA was extracted, dried over sodium sulfate, filtered and concentrated under reduced pressure, and the white solid compound 1 (6.0 mg, 0.013 mmol, yield was 13.5%) was prepared and purified by reverse phase.
  • LC-MS: ESI [M+H] + 443.2.
  • Example 2 1'-((7-(1,1-difluoroethyl)-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)-2-fluoro-N-methyl-1',2',3',6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide
  • Step 1 Dissolve compound 1b (0.68 g, 2.30 mmol) in DCM (30 mL), slowly add DIBAL-H (1 M, 11.5 mL, 11.5 mmol) at 0°C under N2 atmosphere, and stir at 25°C for 1 h. Slowly pour 100 mL of saturated sodium potassium tartrate solution into the reaction solution to quench, extract with DCM 5*50 mL, dry with sodium sulfate, filter and concentrate under reduced pressure to obtain compound 2a (0.59 g, crude).
  • LC-MS: ESI [M+H] + 257.2.
  • Step 4 Compound 2c (0.11 g, 0.23 mmol) was dissolved in dioxane (10 mL), HBr (46% in H 2 O, 5.0 mL) was slowly added dropwise, and stirred at 70°C for 1 h. The reaction solution was cooled and slowly poured into 100 mL of saturated sodium bicarbonate solution for quenching, extracted with DCM/MeOH (20/1) for 5*30 mL, dried over sodium sulfate, and purified by reverse preparation to obtain white solid compound 2 (20 mg, 0.043 mmol, yield 19%).
  • LC-MS: ESI [M+H] + 458.2.
  • Example 2 The preparation methods of compounds 3 to 9 of Examples 3 to 9 can be referred to Example 2.
  • LC-MS: ESI[M+H] + 436.2.
  • Step 2 Compound 10a (0.09 g, 0.21 mmol) was dissolved in dioxane (10 mL), HBr (46% in H 2 O, 5.0 mL) was slowly added dropwise, and stirred at 70°C for 1 h. The reaction solution was cooled and slowly poured into 100 mL of saturated sodium bicarbonate solution for quenching, extracted with 4*50 mL of DCM, dried with sodium sulfate, and purified by reverse preparation to obtain white solid compound 10 (30 mg, 0.07 mmol, yield 34%).
  • LC-MS: ESI [M+H] + 422.2.
  • Example 10 The preparation methods of compounds 11 to 30 of Examples 11 to 30 can be referred to Example 10.
  • Example 31 2-Fluoro-N-methyl-1'-((6-oxo-7-((trifluoromethyl)thio)-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)-1'-, 2', 3', 6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide
  • LC-MS: ESI[M+H] + 436.2.
  • Step 2 Compound 31a (0.09 g, 0.18 mmol) was dissolved in dioxane (10 mL), HBr (46% in H 2 O, 5.0 mL) was slowly added dropwise, and stirred at 70°C for 1 h. The reaction solution was cooled and slowly poured into 100 mL of saturated sodium bicarbonate solution for quenching, extracted with 4*50 mL of DCM, dried with sodium sulfate, and purified by reverse preparation to obtain white solid compound 31 (20 mg, 0.04 mmol, yield 22%).
  • LC-MS: ESI [M+H] + 494.2.
  • Example 31 The preparation methods of compounds 32-40 of Examples 32-40 can be referred to Example 31.
  • Example 41 1'-((7-(difluoromethyl)thio)-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)-N-methyl-1',2',3',6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide
  • LC-MS: ESI[M+H] + 472.2.
  • Step 2 Compound 41a (0.084 g, 0.18 mmol) was dissolved in dioxane (10 mL), HBr (46% in H 2 O, 5.0 mL) was slowly added dropwise, and stirred at 70°C for 1 h. The reaction solution was cooled and slowly poured into 100 mL of saturated sodium bicarbonate solution for quenching, extracted with 4*50 mL of DCM, dried with sodium sulfate, and purified by reverse preparation to obtain white solid compound 41 (18 mg, 0.04 mmol, yield 22%).
  • LC-MS: ESI [M+H] + 458.2.
  • Example 41 The preparation methods of compounds 42 to 49 of Examples 42 to 49 can be referred to Example 41.
  • Example 50 2-Fluoro-N-methyl-1'-((6-oxo-7-vinyl-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)-1', 2', 3',6'-Tetrahydro-[3,4'-bipyridine]-6-carboxamide
  • Step 1 Compound int-1 (0.91 g, 2.92 mmol) was added to anhydrous THF (300 mL), the reaction solution was cooled to -20 ° C, DIBAL-H (4.9 mL, 7.3 mmol, 1.5 M toluene solution) was added under a N 2 atmosphere at -20 ° C, and the reaction mixture was further stirred between -15-0 ° C for 3 hours. TLC showed that the reaction was complete. The reaction was slowly quenched with 3N NaOH aqueous solution between -15-0 ° C, and the internal temperature was kept at no more than 0 ° C.
  • Step 2 SOCl 2 (357 mg, 3.0 mmol) was slowly added to a DMF (100 mL) solution of compound 50a (0.54 g, 2.0 mmol) at 0-5°C under nitrogen atmosphere, and the mixture was stirred at 25°C for 3 h until the starting material was completely consumed.
  • the reaction mixture was stirred at room temperature for 1 hour, and the gray-white precipitate formed was collected by filtration, washed with water (10 mL*3), and dried in vacuo to obtain compound 50b (0.32 g).
  • LC-MS: ESI [M+H] + 287.0.
  • Step 3 Compound 50b (0.42 g, 1.45 mmol) was dissolved in 10 mL of ethanol, and compound int-3 (0.35 g, 1.45 mmol) and KI (0.10 g, 0.6 mmol) were added. DIPEA (3.0 mL, 23 mmol) was added dropwise, and the mixture was stirred at 80°C for 2 h.
  • LC-MS: ESI[M+H] + 482.2/484.1.
  • Example 50 The preparation methods of compounds 51-58 of Examples 51-58 can be referred to Example 50.
  • LC-MS: ESI[M+H] + 439.
  • Step 1 Dissolve compound int-1 (5.0 g, 16.08 mmol) in DCM (100 mL), add DIBAL-H (1.0 M, 50 mL, 50.0 mmol) dropwise at 0°C, and stir at 0°C for 1 h. Slowly add 100 mL of saturated sodium potassium tartrate solution, extract with DCM 4*30 mL, dry with sodium sulfate, and concentrate under reduced pressure to obtain compound 60a (4.34 g, crude).
  • LC-MS: ESI [M+H] + 270.1.
  • Step 5 Compound 60d (1.0 g, 3.76 mmol) was dissolved in MeCN (10 mL), Cs 2 CO 3 (21.2 g, 3.76 mmol) was added, and iodomethane was added dropwise to the reaction solution, and stirred at 25° C. for 3 hours.
  • LC-MS: ESI[M+H] + 299.2.
  • Step 6 Compound 60e (0.65 g, 2.17 mmol) was dissolved in HCl (4.0 M in MeOH, 10 mL), stirred at 25 °C for 2 h, concentrated under reduced pressure, quenched with 100 mL of saturated sodium bicarbonate solution, extracted with DCM for 4*30 mL, The residue was dried over sodium sulfate and concentrated under reduced pressure to give compound 60f (0.6 g, crude).
  • LC-MS: ESI [M+H] + 255.2.
  • Step 7 Compound 60f (0.6 g, 2.35 mmol) was dissolved in DCM (10 mL), DMF (0.01 mL) was added, SOCl 2 (0.4 mL, 4.70 mmol) was added dropwise at 25°C, and the mixture was stirred for 1 h at 25°C. The mixture was filtered and concentrated under reduced pressure to obtain compound 60g (0.6 g, crude).
  • LC-MS: ESI [M+H] + 273.2.
  • Step 8 Dissolve compound 60g (0.60g, 2.20mmol) in dioxane (10mL), slowly add HBr (48% in AcOH, 3.0mL) dropwise, and stir at 40°C for 1h. After cooling, slowly pour the reaction solution into 100mL saturated sodium bicarbonate solution to quench, and filter to obtain white solid compound 60h (0.70g, crude).
  • LC-MS: ESI [M+H] + 259.1.
  • Example 60 The preparation methods of compounds 61-63 of Examples 61-63 can be referred to Example 60.
  • Step 1 Dissolve compound int-18b (0.59 g, 2.50 mmol) in DCM (10 mL), add DMF (0.01 mL) dropwise, add SOCl 2 (0.60 mL, 5.00 mmol) dropwise at 0°C, and stir at 25°C for 1 h. Concentrate under reduced pressure to remove the solvent to obtain compound 64a (0.64 g, crude).
  • LC-MS: ESI [M+H] + 255.2.
  • Step 3 Compound 64b (0.1 g, 0.42 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (0.3 mL, 2.1 mmol), KI (0.02 g), and compound int-23 (0.1 g, 0.42 mmol) were added and stirred at 80°C for 2 h. After concentration under reduced pressure, the mixture was prepared by reverse phase to obtain compound 64 (0.03 g, 0.068 mmol, yield 16%).
  • LC-MS: ESI [M+H] + 442.2.
  • Example 65 5-(4-(((7-(1,1-difluoroethyl)-6-oxo-56-dihydro-1,5-naphthyridin-3-yl)methyl)piperazin-1-yl)-6-fluoro-N-methylpicolinamide
  • Step 1 Dissolve compound 2a (0.12 g, 0.47 mmol) in DCM (10 mL), add DMF (0.01 mL), add SOCl 2 (0.1 mL, 0.87 mmol) dropwise at 25°C, and stir at 25°C for 1 h. Concentrate under reduced pressure to remove the solvent to obtain compound 65a (0.12 g, crude).
  • LC-MS: ESI [M+H] + 273.2.
  • Step 3 Compound 65b (0.06 g, 0.23 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (0.15 mL, 1.0 mmol), KI (0.01 g), and compound int-10 (0.05 g, 0.21 mmol) were added and stirred at 80°C for 2 h. The solvent was removed by concentration under reduced pressure, and the residue was purified by Pre-HPLC to obtain white solid compound 65 (0.02 g, 0.043 mmol, yield 19%).
  • LC-MS: ESI [M+H] + 461.2.
  • Step 1 Dissolve compound int-22c (1.0 g, 3.47 mmol) in DCM (10 mL), add DMF (0.01 mL), add SOCl 2 (1.0 mL, 6.94 mmol) dropwise at 25°C, and stir at 25°C for 1 h. Concentrate under reduced pressure to remove the solvent to obtain compound 67a (0.6 g, crude).
  • LC-MS: ESI [M+H] + 291.2.
  • Step 1 Compound 69a (1.0 g, 5.0 mmol) was dissolved in DCM (20 mL), MnO 2 (7.0 g, 80.0 mmol) was added, and the mixture was stirred at 25° C. for 15 h. The mixture was filtered through celite, and the filtrate was collected and concentrated under reduced pressure to obtain compound 69b (1.0 g, crude).
  • LC-MS: ESI [M+H] + 200.1/202.1.
  • Step 2 Compound 69b (1.0 g, 5.0 mmol) was dissolved in DCM (30 mL), methylthioacetic acid (0.4 mL, 5.0 mmol) was added, the temperature was lowered to 0°C, pyridine (4.0 mL, 50.0 mmol) and POCl 3 (2.3 mL, 25 mmol) were added dropwise in sequence, and the temperature was raised to 25°C and stirred for 1 h.
  • Step 5 Compound 69e (0.17 g, 0.77 mmol) was dissolved in toluene (10 mL), DMF (0.01 mL) was added, SOCl 2 (0.06 mL, 0.85 mmol) was added dropwise at 25°C, and the mixture was stirred for 1 h at 25°C. The mixture was concentrated under reduced pressure to obtain compound 69f (0.2 g, crude).
  • LC-MS: ESI [M+H] + 240.2.
  • Step 7 Compound 69f (0.1 g, 0.42 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (0.3 mL, 1.68 mmol), KI (0.02 g), and compound int-3 (0.1 g, 0.42 mmol) were added and stirred at 80°C for 2 h. The solvent was removed by concentration under reduced pressure, and the residue was purified by Pre-HPLC to obtain white solid compound 69 (0.03 g, 0.07 mmol, yield 16%).
  • LC-MS: ESI [M+H] + 439.2.
  • Example 69 The preparation methods of compounds 70-74 of Examples 70-74 can be referred to Example 69.
  • Example 75 N-methyl-5-(4-((7-(methyl-d 3 )thio)-6-oxo-5,6-dihydro-1,5-naphthopyridin-3-yl)methyl)piperazin-1-yl)picolinamide
  • Step 2 Compound 76b (10 g, 45.446 mmol) was dissolved in DCM (200 mL), MnO 2 (19.76 g, 227.231 mmol) was added, and the mixture was stirred at 25° C. for 3 h. The mixture was filtered through Celite pad, and the filtrate was collected and concentrated under reduced pressure to obtain compound 76c (7.0 g, crude).
  • LC-MS: ESI [M+H] + 217.2/219.2.
  • Step 3 Compound 76c (5 g, 22.933 mmol) was dissolved in THF (150 mL), ethyl 2-methylthioacetate (3.08 g, 22.933 mmol), N 2 was replaced, LiHMDS (68.798 mL, 68.798 mmol) was added dropwise at -78°C, and stirred at -78°C for 1 h.
  • LC-MS: ESI[M+H] + 288.2/290.2.
  • Step 5 Compound 76e (1.35 g, 5.64 mmol) was dissolved in toluene (30 mL), DMF (0.02 mL) was added, SOCl 2 (0.81 g, 6.771 mmol) was added dropwise at 25°C, and stirred at 25°C for 1 h. The mixture was concentrated under reduced pressure to obtain compound 76f (1.3 g, crude).
  • LC-MS: ESI [M+H] + 258.2.
  • Example 76 The preparation methods of compounds 77-90, 96, 98-99 and 100-107 of Examples 77-90, 96, 98-99 and 100-107 can be referred to Example 76.
  • Step 2 Dissolve compound 91b (0.9 g, 5.15 mmol) in THF (30 mL), compound ethyl 2-methylthioacetate (0.83 g, 6.18 mmol), replace with N 2 , add LiHMDS (25.7 mL, 25.7 mmol) dropwise at -78 °C, and stir at -78 °C for 1 h. Then raise the temperature to 25 °C and stir for 12 h. Slowly pour the reaction solution into saturated ammonium chloride solution (100 mL) under ice-water bath to quench the reaction, and collect the filter cake by filtration to obtain the target compound 91c (2.8 g, crude).
  • LC-MS: ESI [M+H] + 245.2.
  • PARP1 protein BPS, Cat. No. 80501
  • PARP2 protein BPS, Cat. No. 80502
  • PARP5A protein BPS, Cat. No. 80504
  • Biotin-NAD+ R&D, Cat. No. 6573
  • Strep-HRP Thermo Pierce, Cat. No. 21127
  • NAD+ TCI, Cat. No. D0919-5G
  • quantitative enhanced chemiluminescence HRP substrate kit Thermo Pierce, Cat. No. 15159
  • histone Active Motif, Cat. No. 81167
  • activated DNA Genescript, Cat. No. L05182-01&02&03
  • anti-rabbit IgG HRP-linked Antibody
  • the compounds of the present invention have a significant inhibitory effect on PARP1.
  • BRCA mutant MDA-MB-436 cells were cultured in DMEM medium containing 10% fetal bovine serum, 100U/mL penicillin, and 100 ⁇ g/mL streptomycin, and cultured in a 5% saturated CO2 incubator at 37°C. When the cells grew to 80% confluence, the cells were collected, centrifuged at 300g for 10min, and plated on a 96-well plate at 1200 cells/well. After 24h, different final concentrations of PARPi (0, 0.01, 0.1, 1, 10, 100, 1000nM) were added and cultured for 72h. The cells were treated with a replacement medium (PARPi was added again at the same final concentration) and cultured for 96h.
  • PARPi 0.05, 0.01, 0.1, 1, 10, 100, 1000nM
  • inhibition rate % 1-(mean OD value of the drug group-mean OD value of the Blank group)/(mean OD value of the Control group-mean OD value of the Blank group)*100%.
  • DLD-1 cells were cultured in RPMI-1640 medium containing 10% fetal bovine serum, 100U/mL penicillin, and 100 ⁇ g/mL streptomycin, and cultured in a 5% saturated CO2 incubator at 37°C. When the cells grew to 80% confluence, the cells were collected, centrifuged at 300g for 10min, and plated at 1000 cells/well in a 96-well plate. After 24h, different final concentrations of PARPi (0, 1, 10 ⁇ M) were added and cultured for another 72h. The cells were treated with a change of medium (PARPi was re-added at the same final concentration) and cultured for another 96h. The 96-well plate was taken out, and the OD value at a wavelength of 450nM was detected by CCK8 method, and the cell inhibition rate was calculated:
  • Inhibition rate % 1-(average OD value of the drug administration group-average OD value of the Blank group)/(average OD value of the Control group-average OD value of the Blank group)*100%.
  • the compounds of the present invention have significant inhibitory effect on BRCA mutant MDA-MB-436 cells, but have no significant inhibitory effect on BRCA wild-type DLD-1 cells, indicating that the compounds of the present invention specifically inhibit homologous recombination-deficient tumor cells, and a large number of preferred compounds of the present invention, such as 2, 11, 17, 21, 28, 59, etc., have significantly better activity than the reference compounds AZD5305 and AZD9574.
  • CHO cells stably expressing hERG were cultured in a cell culture flask and placed in an incubator at 37°C and 5% CO 2. When the cell density grew to 60-80%, the cell culture medium was removed, and the cells were washed once with PBS and digested with Detachin. After complete digestion, the cells were neutralized with culture medium, centrifuged, the supernatant was removed, and the cells were resuspended with culture medium. The cell density was adjusted to 2-5 ⁇ 10 6 /mL for later use.
  • Compound preparation Dilute the compound stock solution with 100% DMSO, that is, take 10 ⁇ L of the compound stock solution and add it to 20 ⁇ L DMSO, and dilute it 3 times continuously to 6 concentrations. Take 4 ⁇ L of the 6 concentrations of the compound respectively and add it to 396 ⁇ L of extracellular fluid, that is, dilute it 100 times to get 6 intermediate concentrations. Then take 80 ⁇ L of the 6 intermediate concentration compounds respectively and add them to 320 ⁇ L of extracellular fluid, that is, dilute it 5 times to the final concentration to be tested. The highest test concentration is 40 ⁇ M, and there are 6 concentrations of 40, 13.33, 4.44, 1.48, 0.49 and 0.16 ⁇ M respectively. The DMSO content in the final test concentration does not exceed 0.2%, and this concentration of DMSO has no effect on the hERG potassium channel.
  • the compound preparation is completed by the Bravo instrument throughout the dilution process.
  • Electrophysiological recording process The single-cell high-impedance sealing and whole-cell pattern formation process are all automatically completed by the Qpatch instrument. After obtaining the whole-cell recording mode, the cell is clamped at -80 mV. Before giving a 5-second +40 mV depolarizing stimulus, a 50-millisecond -50 mV pre-voltage is given, and then repolarizes to -50 mV for 5 seconds, and then returns to -80 mV. This voltage stimulus is applied every 15 seconds. After recording for 2 minutes, the extracellular solution is given for 5 minutes, and then the drug administration process begins. The compound concentration starts from the lowest test concentration, and each test concentration is given for 2.5 minutes. After all concentrations are given continuously, the positive control compound 3 ⁇ M Cisapride is given. At least 3 cells (n ⁇ 3) are tested for each concentration.
  • X is the Log value of the test sample detection concentration
  • Y is the inhibition percentage at the corresponding concentration
  • Bottom and Top are the minimum and maximum inhibition percentages, respectively.
  • Experimental plan The brain-blood distribution of the compound was investigated by monitoring the content of the compound in the mouse brain and plasma.
  • mice Male mice, were orally administered at 10 mg ⁇ kg -1.
  • Whole blood was centrifuged at 3500 rpm for 15 min, and the supernatant plasma was collected.
  • the weight of the centrifuge tube was weighed as M1
  • the weight of the centrifuge tube containing the whole brain was weighed as M2
  • the weight of the centrifuge tube after adding water homogenate was weighed as M3
  • the weight of the centrifuge tube after taking out 30 ⁇ L of homogenate was weighed as M4.
  • CO2 cell culture box Yamato brand, model IP610; biological safety cabinet: Suzhou Antai brand, model BSC-1304IIA2; normal temperature centrifuge: Thermo Scientific brand, model SORVALLST 16;
  • Water purifier American Millipore brand, model F7PNO9748; vertical high pressure sterilizer: Yamato brand, model DKN812C; 1mL disposable sterile syringe: Shanghai Kangdeli Enterprise Development Group Co., Ltd.; 1mL disposable sterile insulin syringe: BD brand; weight scale: Shanghai Hengping Instrument Factory, model JY2002; analytical balance: SARTORIUS brand, model BCE95I-1CEU;
  • Graphpad Prism version 6.0, Graphpad Software Ltd., Graphpad Prism software is used to organize and quantify data and to create bar graphs.
  • Tumor cell culture and preparation For routine tumor cell line passage and culture, the culture medium contains 10% fetal bovine serum, 100U/mL penicillin, and 100 ⁇ g/mL streptomycin. When the cells grow to 80% confluence, collect the cells and centrifuge at 300g for 10min. Wash with pre-cooled PBS three times, collect the cells by centrifugation at 300g for 10min, resuspend the cells in PBS, count the cells with a hemocytometer, adjust the cell concentration, and pre-cool the cell suspension on ice after cell collection. Take 100 ⁇ L of the cell suspension and inject it subcutaneously in the dorsal armpit of the mouse. Start group treatment when the tumor volume reaches 200-300mm3 .
  • Observation animals All surviving experimental animals planned for observation; Observation time: Twice a day; Observation content: including but not limited to general manifestations, behavioral status, eyes, mouth, nose and mouth, ears, hair, feces, urine, genitals and other toxic symptoms. If any abnormalities are found, detailed descriptions are required.
  • Tumor volume (TV), relative tumor volume (RTV) and relative tumor proliferation rate of subcutaneous transplanted tumor model Use a vernier caliper to measure the long and short diameters of the tumor every 2 days, and dynamically observe the anti-tumor effect of the test drug.
  • the relative tumor volume (RTV) was calculated based on the measurement results.
  • Tumor weight determination and calculation of tumor inhibition rate (%) At the end of treatment, the animals were killed, the tumor was dissected and removed, the tumor weight was weighed, and photographed. The tumor inhibition rate (%) was calculated according to the following formula:
  • Tumor inhibition rate (average tumor weight of negative control group (g) - average tumor weight of drug administration group (g)) / average tumor weight of negative control group (g) ⁇ 100%;
  • mice 30 female NOD/SCID mice were randomly divided into blank control group, compound 47 (0.3 mg/kg) dose group, compound 64 (0.3 mg/kg) dose group, reference compound AZD5305 0.3 mg/kg and reference compound Olaparib 100 mg/kg dose group, a total of 5 groups, 6 mice in each group, all were orally administered once a day.
  • the body weight was weighed every 2 days and the length and width of the tumor were measured with a vernier caliper.
  • the tumor-bearing mice were anesthetized and killed, the tumor tissue was peeled off, weighed and photographed, and the tumor inhibition rate was calculated.
  • each drug-treated group was continuously administered for 30 days. Compared with the blank control group, each drug-treated group (AZD5305 0.3 mg/kg, compound 47 0.3 mg/kg, compound 64 0.3 mg/kg, Olaparib 100 mg/kg) had a significant inhibitory effect on the growth of human breast cancer cell MM436 cell line NOD/SCID mouse subcutaneous tumor model.
  • the anti-tumor effects of the compound 47 0.3 mg/kg dose group and the compound 64 0.3 mg/kg dose group were significantly better than the reference compound Olaparib 100 mg/kg dose group, and were equivalent to the AZD5305 0.3 mg/kg dose group; in addition, after 30 days of administration, the tumors of one mouse were completely regressed in the compound 64 0.3 mg/kg and AZD5305 0.3 mg/kg dose groups, respectively, showing excellent anti-tumor efficacy.
  • CD34+ hematopoietic stem cells (hmPB34-P-SC) were cultured overnight in RPMI 1640 complete medium supplemented with 25ng/mL IL-3, 25ng/mL IL-6, 25ng/ml SCF and 10% fetal bovine serum at 37°C and 5% CO2. The next day, the cells were resuspended and seeded in 96-well plates at a concentration of 1000 cells per well, and different concentrations of drugs were added at the same time, with 3 replicates. After 5 days of culture, the number of live cells in each well was determined using the CellTiter-Glo 2.0 kit and detected by a microplate reader.
  • the compounds of the present invention have weak inhibitory effects on CD34+ hematopoietic stem progenitor cells hmPB34-P-SC, especially compounds 76, 83, 89 and 95, whose IC 50 values for hmPB34-P-SC inhibitory activity are greater than 1000nM, which are significantly better than the positive drug Olaparib.
  • Some compounds of the present invention have good brain penetration potential, especially compounds 81, 83, 88, 89, 90, 95, 96 and 101, which have higher distribution concentrations in rat cerebrospinal fluid.
  • mice 18 female NOD/SCID mice were randomly divided into a blank control group, a compound AZD9574 (3 mg/kg) group, and a compound 11 (1 mg/kg) group, a total of 3 groups, 6 mice in each group, all of which were orally administered once a day.
  • the body weight was weighed every 2 days and the length and width of the tumor were measured with a vernier caliper.
  • the tumor-bearing mice were anesthetized and killed, the tumor tissue was peeled off, weighed and photographed, and the tumor inhibition rate was calculated.
  • Compound AZD9574 was synthesized by referring to the synthesis method of Example 20 of patent WO2021260092.
  • mice Ten days after inoculation, group treatment began based on the results of in vivo imaging. Eighteen female NOD/SCID mice were used in the experiment and randomly divided into a blank control group, a compound 11 3mg/kg dose group, and a reference compound AZD9574 3mg/kg group, for a total of 3 groups, with 6 mice in each group. Each group was orally administered once a day. The body weight was measured every 2 days and the fluorescence intensity of the mouse brain was measured every 7 days. The drug was stopped after 42 days of administration, and the changes in the fluorescence intensity of the mouse brain and the survival rate of the mice were counted.
  • the median survival time of the blank control group, AZD9574 3 mg/kg group, and compound 11 3 mg/kg group was NA, 37.5 days, and 61.5 days, respectively.
  • the survival rate of compound 11 was significantly better than that of the reference compound AZD9574, and the weight loss of compound 11 was weaker than that of the reference compound AZD9574, indicating that compound 11 has a better therapeutic effect on intracranial tumors.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A PARP1-targeting compound represented by formula (I) and a use thereof, relating to the technical field of chemical medicines. The PARP1-targeting compound can be used as a PARP1 inhibitor, has the advantages of high activity and high selectivity, and also has excellent pharmacokinetic properties, excellent safety, and brain penetration potential.

Description

PARP1靶向化合物及其用途PARP1 targeting compounds and uses thereof 技术领域Technical Field

本发明属于化学医药领域,涉及一类PARP1靶向化合物及其用途。The present invention belongs to the field of chemical medicine and relates to a class of PARP1 targeting compounds and uses thereof.

背景技术Background Art

细胞在生长过程中,它的DNA会不断受到内在和周遭各种不利因素的损害。在DNA损伤中,最严重的损伤类型是单链断裂和双链断裂,其中单链断裂更为常见,这些断裂如果得不到及时准确的修复,会导致基因组不稳定,进而引起癌变,甚至直接引起细胞死亡。对于DNA的单链断裂而言,它的修复主要依赖于PARP酶。对于双链断裂而言,双链DNA的修复方式,一种是非同源末端链接修复,另外一种是同源重组修复。同源重组修复是一种高保真,无错误的修复方式,也是双链DNA修复的主要途径。同源重组修复参与蛋白众多,其中最为人熟知的是BRCA蛋白。2005年的两项研究(Farmer H,Mccabe N,et al.Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy[J].Nature,2005,434(7035):917-921.Bryant,H.,Schultz,N.,Thomas,H.et al.Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose)polymerase.Nature 434,913–917(2005))表明,缺乏BRCA1或BRCA2的肿瘤细胞会被PARP抑制剂选择性地抑制。根据这一研究结果,学者提出了合成致死的概念:BRCA和PARP两种基因中的任何一种缺失本身都不致命,但两者同时失活会导致细胞死亡。基于合成致死理论,PARP抑制剂(PARPi)被开发用于选择性靶向BRCA1/2突变的癌细胞。During the growth of cells, their DNA will be constantly damaged by various internal and surrounding adverse factors. Among DNA damage, the most serious types of damage are single-strand breaks and double-strand breaks, among which single-strand breaks are more common. If these breaks are not repaired promptly and accurately, they will lead to genomic instability, causing cancer and even directly causing cell death. For single-strand breaks of DNA, its repair mainly depends on the PARP enzyme. For double-strand breaks, there are two ways to repair double-stranded DNA: non-homologous end joining repair and homologous recombination repair. Homologous recombination repair is a high-fidelity, error-free repair method, and it is also the main way to repair double-stranded DNA. There are many proteins involved in homologous recombination repair, among which the most well-known is the BRCA protein. Two studies in 2005 (Farmer H, McCabe N, et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy [J]. Nature, 2005, 434 (7035): 917-921. Bryant, H., Schultz, N., Thomas, H. et al. Specific killing of BRCA2-deficient tumors with inhibitors of poly (ADP-ribose) polymerase. Nature 434, 913–917 (2005)) showed that tumor cells lacking BRCA1 or BRCA2 would be selectively inhibited by PARP inhibitors. Based on this research result, scholars proposed the concept of synthetic lethality: the loss of either BRCA or PARP gene itself is not lethal, but the simultaneous inactivation of both will lead to cell death. Based on the theory of synthetic lethality, PARP inhibitors (PARPi) have been developed to selectively target cancer cells with BRCA1/2 mutations.

PARP抑制剂在同源重组缺陷癌症患者中已经表现出了优异的临床疗效,然而无论是单药使用还是联合疗法,血液学毒性(贫血、中性粒细胞减少和血小板减少)和其他毒性限制了这类药物的应用。相关研究表明(Harris P A,Boloor A,Cheung M,et al.Discovery of 5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2-methyl-benzenesulfonamide(Pazopanib),a novel and potent vascular endothelial growth factor receptor inhibitor.[J].Journal of Medicinal Chemistry,2008,51(15):4632.)这部分不良反应可能来源于已上市PARP抑制剂对于PARP2的抑制,而PARP2并非疗效所必须。高选择性PARP1抑制剂可以降低血液毒性,提高治疗安全窗,增加与其他化疗或者靶向药联用的潜力。PARP inhibitors have shown excellent clinical efficacy in patients with homologous recombination-deficient cancers. However, hematological toxicity (anemia, neutropenia, and thrombocytopenia) and other toxicities limit the application of these drugs, whether used as a single agent or in combination therapy. Related studies have shown that (Harris P A, Boloor A, Cheung M, et al. Discovery of 5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2-methyl-benzenesulfonamide (Pazopanib), a novel and potent vascular endothelial growth factor receptor inhibitor. [J]. Journal of Medicinal Chemistry, 2008, 51(15): 4632.) These adverse reactions may be due to the inhibition of PARP2 by the PARP inhibitors on the market, which is not necessary for efficacy. Highly selective PARP1 inhibitors can reduce hematologic toxicity, improve the therapeutic safety window, and increase the potential for combination with other chemotherapy or targeted drugs.

因此,对于有效且安全的PARP抑制剂存在未满足的临床需求,特别是对PARP1 具有选择性的PARP抑制剂。Therefore, there is an unmet clinical need for effective and safe PARP inhibitors, especially for PARP1 Selective PARP inhibitor.

发明内容Summary of the invention

本发明的目的在于提供一类PARP1靶向化合物及其用途,其对PARP1的选择性比其他PARP家族成员(如PARP2、PARP3、PARP5a和PARP6)出人意料地高,能够实现高选择性、高效预防或治疗与PARP功能相关的疾病;特别是,本发明所述的PARP1靶向化合物令人惊讶地能够穿透血脑屏障(BBB);因此,本发明所述的PARP1靶向化合物可用于治疗发生在中枢神经系统组织(例如脑和脊髓)中的疾病和病症。The object of the present invention is to provide a class of PARP1 targeted compounds and uses thereof, which have unexpectedly higher selectivity for PARP1 than other PARP family members (such as PARP2, PARP3, PARP5a and PARP6), and can achieve highly selective and highly effective prevention or treatment of diseases related to PARP function; in particular, the PARP1 targeted compounds of the present invention are surprisingly able to penetrate the blood-brain barrier (BBB); therefore, the PARP1 targeted compounds of the present invention can be used to treat diseases and conditions occurring in central nervous system tissues (such as the brain and spinal cord).

第一方面,本发明提供一种式I所示的化合物或其药学上可接受的形式,其结构如下:
In a first aspect, the present invention provides a compound of formula I or a pharmaceutically acceptable form thereof, the structure of which is as follows:

其中,in,

X选自N或者C,且当X选自N时,表示单键,当X选自C时,表示双键;X is selected from N or C, and when X is selected from N, represents a single bond, when X is selected from C, represents a double bond;

X1选自N或者C(R8a),X2选自N或者C(R8b),X3选自N或者C(R8c),且X1、X2和X3中至多有一个选自N; X1 is selected from N or C( R8a ), X2 is selected from N or C( R8b ), X3 is selected from N or C( R8c ), and at most one of X1 , X2 and X3 is selected from N;

R1选自C1~8烷硫基、C1~8卤代烷硫基、C1~8氘代烷硫基、C1~8卤代烷氧基、C1~8卤代烷基或者C2~8烯基; R1 is selected from C1-8 alkylthio, C1-8 halogenated alkylthio, C1-8 deuterated alkylthio, C1-8 halogenated alkoxy, C1-8 halogenated alkyl or C2-8 alkenyl;

R2选自氢、氟、氯、C1-4烷基、C1-4氟代烷基或者C1-4氘代烷基; R2 is selected from hydrogen, fluorine, chlorine, C1-4 alkyl, C1-4 fluoroalkyl or C1-4 deuterated alkyl;

R3、R4独立地选自氢、氘、氟、C1-4烷基、C1-4氟代烷基或者C1-4氘代烷基;R 3 and R 4 are independently selected from hydrogen, deuterium, fluorine, C 1-4 alkyl, C 1-4 fluoroalkyl or C 1-4 deuterated alkyl;

或者,R3、R4与它们所连接的原子一起形成3-6元烷基环;Alternatively, R 3 , R 4 , together with the atoms to which they are attached, form a 3-6 membered alkyl ring;

R5选自氢、氟、氯、氰基、C1-4烷基、C1-4氟代烷基或者C1-4氘代烷基;R 5 is selected from hydrogen, fluorine, chlorine, cyano, C 1-4 alkyl, C 1-4 fluoroalkyl or C 1-4 deuterated alkyl;

或者,R4、R5与它们所连接的原子一起形成5-7元烷基杂环;R4、R5与它们所连接的原子相连成环时,所述5-7元烷基杂环中,除主环上的X外,还含有1个O杂原子;Alternatively, R 4 , R 5 and the atoms to which they are connected together form a 5-7 membered alkyl heterocycle; when R 4 , R 5 and the atoms to which they are connected are connected to form a ring, the 5-7 membered alkyl heterocycle contains 1 O heteroatom in addition to X on the main ring;

R3、R4与它们所连接的原子,和R4、R5与它们所连接的原子,两者不会同时成环;R 3 , R 4 and the atoms to which they are attached, and R 4 , R 5 and the atoms to which they are attached, will not form a ring at the same time;

R6选自-C(O)-NH-R6a;R6a选自C1-4烷基、C1-4氟代烷基、C1-4氘代烷基或者3-6元环烷基;R 6 is selected from -C(O)-NH-R 6a ; R 6a is selected from C 1-4 alkyl, C 1-4 fluoroalkyl, C 1-4 deuterated alkyl or 3-6 membered cycloalkyl;

R7选自氢、氟、氯、氰基、C1-4烷基、C1-4氟代烷基或者C1-4氘代烷基; R7 is selected from hydrogen, fluorine, chlorine, cyano, C1-4 alkyl, C1-4 fluoroalkyl or C1-4 deuterated alkyl;

R8a、R8b、R8c独立地选自氢、氟、氯、C1-4烷基、C1-4氘代烷基、C1-4氟代烷基;R 8a , R 8b , and R 8c are independently selected from hydrogen, fluorine, chlorine, C 1-4 alkyl, C 1-4 deuterated alkyl, and C 1-4 fluoroalkyl;

所述药学上可接受的形式选自药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、氮氧化物、同位素标记物、代谢物或前药。The pharmaceutically acceptable form is selected from pharmaceutically acceptable salts, esters, stereoisomers, tautomers, polymorphs, solvates, N-oxides, isotopically labeled substances, metabolites or prodrugs.

在本发明的一些实施例中,X1选自N,X2选自C(R8b),X3选自C(R8c);或者X1选自C(R8a),X2选自N,X3选自C(R8c);或者X1选自C(R8a),X2选自者C(R8b),X3选自C(R8c)。In some embodiments of the present invention, X1 is selected from N, X2 is selected from C( R8b ), and X3 is selected from C( R8c ); or X1 is selected from C( R8a ), X2 is selected from N, and X3 is selected from C( R8c ); or X1 is selected from C( R8a ), X2 is selected from C( R8b ), and X3 is selected from C( R8c ).

在本发明的一些实施例中,R1选自C1~4烷硫基、C1~4卤代烷硫基、C1~4氘代烷硫基、C1~4卤代烷氧基、C1~4卤代烷基、C2~4烯基;R1中,所述卤代的卤选自F、Cl、Br。In some embodiments of the present invention, R1 is selected from C1-4 alkylthio, C1-4 haloalkylthio, C1-4 deuterated alkylthio, C1-4 haloalkoxy, C1-4 haloalkyl, C2-4 alkenyl; in R1 , the halogen is selected from F, Cl, and Br.

在本发明的一些优选实施例中,R1选自甲硫基、卤代甲硫基、氘代甲硫基、卤代甲氧基、卤代甲基、卤代乙基或者乙烯基;R1中,所述卤代的卤选自F、Cl。In some preferred embodiments of the present invention, R 1 is selected from methylthio, halogenated methylthio, deuterated methylthio, halogenated methoxy, halogenated methyl, halogenated ethyl or vinyl; in R 1 , the halogenated halogen is selected from F and Cl.

在本发明的一些更实施例中,R1选自-S-CH3、-S-CF2H、-S-CF3、-S-CF2Cl、-S-CD3、-O-CF2H、-O-CF3、-O-CF2Cl、-CF2-CH3或者-CH=CH2In some further embodiments of the present invention, R1 is selected from -S- CH3 , -S- CF2H , -S- CF3 , -S- CF2Cl , -S-CD3, -O- CF2H , -O- CF3 , -O- CF2Cl , -CF2 - CH3 or -CH = CH2 .

在本发明的一些实施例中,R2选自氢、氟、氯、甲基、氟代甲基或者氘代甲基。In some embodiments of the present invention, R2 is selected from hydrogen, fluorine, chlorine, methyl, fluoromethyl or deuterated methyl.

在本发明的一些实施例中,当R3、R4不与它们所连接的原子成环时,R3、R4独立地选自氢、氘、氟、甲基、氟代甲基或者氘代甲基。In some embodiments of the present invention, when R 3 and R 4 do not form a ring with the atoms to which they are attached, R 3 and R 4 are independently selected from hydrogen, deuterium, fluorine, methyl, fluoromethyl or deuterated methyl.

在本发明的一些实施例中,当R4、R5不与它们所连接的原子成环时,R5选自氢、氟、氯、氰基、甲基、氟代甲基或者氘代甲基。In some embodiments of the present invention, when R 4 and R 5 do not form a ring with the atoms to which they are attached, R 5 is selected from hydrogen, fluorine, chlorine, cyano, methyl, fluoromethyl or deuterated methyl.

在本发明的一些实施例中,R7选自氢、氟、氯、氰基、甲基、氟代甲基或者氘代甲基。In some embodiments of the present invention, R 7 is selected from hydrogen, fluorine, chlorine, cyano, methyl, fluoromethyl or deuterated methyl.

在本发明的一些实施例中,R8a、R8a、R8c独立地选自氢、氟、氯、甲基、氟代甲基或者氘代甲基。In some embodiments of the present invention, R 8a , R 8a , R 8c are independently selected from hydrogen, fluorine, chlorine, methyl, fluoromethyl or deuterated methyl.

在本发明的一些实施例中,R6a选自甲基、氟代甲基、氘代甲基或者环丙基。In some embodiments of the present invention, R 6a is selected from methyl, fluoromethyl, deuterated methyl or cyclopropyl.

在本发明的一些优选实施例中,R6a选自甲基、-CF3、-CD3或者环丙基。In some preferred embodiments of the present invention, R 6a is selected from methyl, -CF 3 , -CD 3 or cyclopropyl.

在本发明的一些实施例中,结构单元选自:

In some embodiments of the present invention, the structural unit Selected from:

在本发明的一些实施例中,当R4、R5不与它们所连接的原子成环时,结构单元(从左到右与主环连接,即N端与主环亚甲基相连,X端与主环吡啶基相连)选自: In some embodiments of the present invention, when R 4 and R 5 do not form a ring with the atoms to which they are attached, the structural unit (connected to the main ring from left to right, that is, the N end is connected to the main ring methylene, and the X end is connected to the main ring pyridyl) is selected from:

在本发明的一些实施例中,当R4、R5不与它们所连接的原子成环时,结构单元选自: In some embodiments of the present invention, when R 4 and R 5 do not form a ring with the atoms to which they are attached, the structural unit Selected from:

在本发明的一些实施例中,当R4、R5与它们所连接的原子成环时,结构单元选自: In some embodiments of the present invention, when R 4 , R 5 and the atoms to which they are attached form a ring, the structural unit Selected from:

在本发明的一些优选实施例中,当R4、R5与它们所连接的原子成环时,结构单元 选自:
In some preferred embodiments of the present invention, when R 4 , R 5 and the atoms to which they are attached form a ring, the structural unit Selected from:

在本发明的一些实施例中,将式Ⅰ中结构单元替换为(两者与主环的连接方式一致,即N端与主环亚甲基相连,O端与主环吡啶基相连)(此时R5不与相连原子成环)。In some embodiments of the present invention, the structural unit in formula I Replace with (Both are connected to the main ring in the same way, that is, the N end is connected to the main ring methylene, and the O end is connected to the main ring pyridyl) (In this case, R 5 does not form a ring with the connected atoms).

本发明还提供了一些具体化合物,所述化合物选自:




The present invention also provides some specific compounds, which are selected from:




在本发明的一些实施例中,R1选自-S-CH3、-S-CF2H、-S-CF3、-S-CF2Cl、-S-CD3、-O-CF2H、-O-CF3、-O-CF2Cl、-CF2-CH3、-CH=CH2、-S-CFH2或者-O-CFH2In some embodiments of the invention, R1 is selected from -S- CH3 , -S- CF2H , -S- CF3 , -S- CF2Cl , -S- CD3 , -O- CF2H , -O- CF3 , -O- CF2Cl , -CF2 - CH3 , -CH= CH2 , -S- CFH2 or -O- CFH2 .

在本发明的一些实施例中,结构单元选自:

In some embodiments of the present invention, the structural unit Selected from:

在本发明的一些实施例中,将式Ⅰ中结构单元替换为(两者与主环的连接方式一致,即N端与主环亚甲基相连,CH端与主环吡啶基相连)(此时R5不与相连原子成环)。In some embodiments of the present invention, the structural unit in formula I Replace with (Both are connected to the main ring in the same way, that is, the N end is connected to the methylene group of the main ring, and the CH end is connected to the pyridyl group of the main ring) (In this case, R 5 does not form a ring with the connected atoms).

本发明还提供了一些具体化合物,所述化合物选自:

The present invention also provides some specific compounds, which are selected from:

在本发明的一些实施例中,当R4、R5不与它们所连接的原子成环时,结构单元选自: In some embodiments of the present invention, when R 4 and R 5 do not form a ring with the atoms to which they are attached, the structural unit Selected from:

在本发明的一些实施例中,当R4、R5与它们所连接的原子成环时,结构单元 选自: In some embodiments of the present invention, when R 4 , R 5 and the atoms to which they are attached form a ring, the structural unit Selected from:

在本发明的一些优选实施例中,当R4、R5与它们所连接的原子成环时,结构单元选自:
In some preferred embodiments of the present invention, when R 4 , R 5 and the atoms to which they are attached form a ring, the structural unit Selected from:

在本发明的一些实施例中,将式Ⅰ中结构单元替换为 (从左到右与主环连接,即N端与主环亚甲基相连,炔基端与主环吡啶基相连)。In some embodiments of the present invention, the structural unit in formula I Replace with (Connected to the main ring from left to right, that is, the N-terminal is connected to the methylene group of the main ring, and the alkynyl end is connected to the pyridyl group of the main ring).

在本发明的一些实施例中,将式Ⅰ中结构单元替换为 In some embodiments of the present invention, the structural unit in formula I Replace with

在本发明的一些实施例中,将式Ⅰ中结构单元替换为 R9选自H。In some embodiments of the present invention, the structural unit in formula I Replace with R9 is selected from H.

本发明还提供了一些具体化合物,所述化合物选自:


The present invention also provides some specific compounds, which are selected from:


第二方面,本发明提供了一种药物组合物,其以前述化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、氮氧化物、同位素标记物、代谢物或前药为活性成分,辅以药学上可接受的载体。In a second aspect, the present invention provides a pharmaceutical composition comprising the aforementioned compound or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, nitrogen oxide, isotope-labeled substance, metabolite or prodrug as an active ingredient, supplemented with a pharmaceutically acceptable carrier.

本发明的进一步的目的在于提供一种制备本发明的药物组合物的方法,所述方法包括将含式I的任意化合物或其药学上可接受的形式、或者它们的混合物、与一种或多种药学上可接受的载体组合。A further object of the present invention is to provide a method for preparing the pharmaceutical composition of the present invention, which comprises combining any compound of Formula I or a pharmaceutically acceptable form thereof, or a mixture thereof, with one or more pharmaceutically acceptable carriers.

在本发明的药物组合物中可使用的药学上可接受的载体为药学上可接受的载体,适合的药学上可接受的载体的实例如在Remington’s Pharmaceutical Sciences(2005)中所述。The pharmaceutically acceptable carrier that can be used in the pharmaceutical composition of the present invention is a pharmaceutically acceptable carrier. Examples of suitable pharmaceutically acceptable carriers are described in Remington’s Pharmaceutical Sciences (2005).

药物组合物可以以任意形式施用,只要其实现预防、减轻、防止或者治愈人类或动物患者的症状即可。例如,可根据给药途径制成各种适宜的剂型。The pharmaceutical composition can be administered in any form as long as it prevents, alleviates, prevents or cures the symptoms of a human or animal patient. For example, it can be prepared into various suitable dosage forms according to the administration route.

在另一些实施方案中,本发明的化合物或药物组合物的施用可以与另外的治疗方法组合。所述另外的治疗方法可以选自,但不限于:放射疗法、化疗疗法、免疫疗法,或其组合。In other embodiments, the administration of the compounds or pharmaceutical compositions of the present invention may be combined with another therapeutic method. The other therapeutic method may be selected from, but not limited to: radiotherapy, chemotherapy, immunotherapy, or a combination thereof.

本发明还涉及一种药物制剂,其包含式I的任意化合物或其药学上可接受的形式、或它们的混合物作为活性成分,或者本发明的药物组合物。在一些实施方案中,所述制剂的形式为固体制剂、半固体制剂、液体制剂或气态制剂。The present invention also relates to a pharmaceutical preparation comprising any compound of formula I or a pharmaceutically acceptable form thereof, or a mixture thereof as an active ingredient, or a pharmaceutical composition of the present invention. In some embodiments, the preparation is in the form of a solid preparation, a semisolid preparation, a liquid preparation or a gaseous preparation.

第三方面,本发明提供前述化合物,式I化合物,及相关具体化合物或其药学上可接受的形式、或者本发明的药物组合物在制备用于预防或治疗PARP1酶相关疾病的药物中的用途。In a third aspect, the present invention provides the use of the aforementioned compounds, compounds of formula I, and related specific compounds or pharmaceutically acceptable forms thereof, or the pharmaceutical compositions of the present invention in the preparation of drugs for preventing or treating PARP1 enzyme-related diseases.

本发明提供一种用于预防或治疗PARP1酶相关疾病的方法,所述方法包括向有此需要的个体施用式I的化合物或其药学上可接受的形式、或者本发明的药物组合物。The present invention provides a method for preventing or treating PARP1 enzyme-related diseases, comprising administering a compound of formula I or a pharmaceutically acceptable form thereof, or a pharmaceutical composition of the present invention to an individual in need thereof.

本发明提供式I化合物或其药学上可接受的形式、或者本发明的药物组合物,用于预防或治疗PARP1酶相关疾病。The present invention provides a compound of formula I or a pharmaceutically acceptable form thereof, or a pharmaceutical composition of the present invention for use in preventing or treating PARP1 enzyme-related diseases.

本发明提供式I的化合物或其药学上可接受的形式或者本发明的药物组合物与另外的治疗方法组合用于预防或治疗PARP1酶相关疾病的方法,所述另外的治疗方法包括但不限于:放射疗法、化疗疗法,免疫疗法、或其组合。 The present invention provides a method for preventing or treating PARP1 enzyme-related diseases in combination with a compound of formula I or a pharmaceutically acceptable form thereof or a pharmaceutical composition of the present invention, wherein the additional treatment method includes but is not limited to: radiotherapy, chemotherapy, immunotherapy, or a combination thereof.

在一些实施方案中,所述PARP1酶相关疾病为对PARP1酶抑制敏感或有响应的疾病。In some embodiments, the PARP1 enzyme-related disease is a disease that is sensitive or responsive to PARP1 enzyme inhibition.

在一些实施方案,所述PARP1酶相关疾病为肿瘤类病症。In some embodiments, the PARP1 enzyme-related disease is a tumor disorder.

在一些优选的实施方案,所述肿瘤类病症缺乏HR依赖性DNA DSB修复途径。In some preferred embodiments, the tumor-like disorder lacks the HR-dependent DNA DSB repair pathway.

在一些优选的实施方案,所述肿瘤类病症包含一种或多种癌细胞,所述癌细胞相对于正常细胞具有降低的或缺失的通过HR修复DNA DSB的能力。In some preferred embodiments, the tumor-like disorder comprises one or more cancer cells having reduced or absent ability to repair DNA DSB via HR relative to normal cells.

在一些优选的实施方案,所述癌细胞具有BRCA1或BRCA2缺陷表型。In some preferred embodiments, the cancer cells have a BRCA1 or BRCA2 deficient phenotype.

在一些实施方案中,所述PARP1酶相关疾病为肿瘤类病症,包括但不限于实体和血液恶性肿瘤。在进一步的实施方案中,所述肿瘤类病症包括但不限于乳腺癌、结肠直肠癌、结肠癌、肺癌(包括小细胞肺癌、非小细胞肺癌和细支气管肺泡癌)和前列腺癌,以及胆管癌、骨癌、膀胱癌、头颈癌、肾癌、肝癌、胃肠组织癌、食道癌、卵巢癌、胰腺癌、皮肤癌、睾丸癌、甲状腺癌、子宫癌、宫颈癌和外阴癌,以及白血病(包括慢性淋巴细胞性白血病(CLL)、急性淋巴细胞性白血病(ALL)和慢性骨髓性白血病(CML))、多发性骨髓瘤、淋巴瘤、脑膜瘤、垂体瘤、颅咽管瘤、神经鞘瘤、胶质瘤、室管膜瘤、原始神经外胚层肿瘤、中枢神经系统淋巴瘤、生殖细胞肿瘤、转移瘤、脑癌或中枢神经系统癌。In some embodiments, the PARP1 enzyme-related disease is a tumor-like disorder, including but not limited to solid and hematological malignancies. In further embodiments, the tumor-like disorder includes but is not limited to breast cancer, colorectal cancer, colon cancer, lung cancer (including small cell lung cancer, non-small cell lung cancer and bronchioloalveolar carcinoma) and prostate cancer, as well as bile duct cancer, bone cancer, bladder cancer, head and neck cancer, kidney cancer, liver cancer, gastrointestinal tissue cancer, esophageal cancer, ovarian cancer, pancreatic cancer, skin cancer, testicular cancer, thyroid cancer, uterine cancer, cervical cancer and vulvar cancer, as well as leukemia (including chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL) and chronic myeloid leukemia (CML)), multiple myeloma, lymphoma, meningioma, pituitary tumor, craniopharyngioma, schwannoma, glioma, ependymoma, primitive neuroectodermal tumor, central nervous system lymphoma, germ cell tumor, metastasis, brain cancer or central nervous system cancer.

在一些优选的实施方案,所述PARP1酶相关疾病为乳腺癌、卵巢癌、胰腺癌、前列腺癌、血液癌、胃肠道癌、肺癌或者胶质瘤。In some preferred embodiments, the PARP1 enzyme-related disease is breast cancer, ovarian cancer, pancreatic cancer, prostate cancer, blood cancer, gastrointestinal cancer, lung cancer or glioma.

在进一步优选的实施方案中,本发明的化合物可以与放化疗或免疫疗法联用以预防或治疗癌症。In a further preferred embodiment, the compounds of the present invention can be used in combination with chemoradiotherapy or immunotherapy to prevent or treat cancer.

第四方面,本发明提供前述化合物,式I化合物,及相关具体化合物或其药学上可接受的形式、或者本发明的药物组合物在制备PARP1抑制剂中的用途。In a fourth aspect, the present invention provides the use of the aforementioned compounds, compounds of formula I, and related specific compounds or pharmaceutically acceptable forms thereof, or the pharmaceutical compositions of the present invention in the preparation of PARP1 inhibitors.

本发明的有益效果:Beneficial effects of the present invention:

本发明提供一类新型的高活性高选择性PARP1抑制剂,能够实现下述至少一种技术效果:(1)对PARP1酶的高抑制活性;(2)选择性抑制PARP1酶,对PARP2、PARP5a、PARP5b等PARP家族其他酶具有高选择性;(3)对同源重组缺陷型肿瘤细胞具有强抑制活性,对非同源重组缺陷型细胞抑制作用弱;(4)优异的药物代谢动力学性质(例如良好的生物利用度、合适的半衰期和作用持续时间);(5)优异的安全性(较低的毒性和/或较少的副作用,较宽的治疗窗)等;本发明所述的PARP1靶向化合物令人惊讶地能够穿透血脑屏障(BBB),可用于治疗发生在中枢神经系统组织(例如脑和脊髓)中的疾病和病症。 The present invention provides a novel class of highly active and highly selective PARP1 inhibitors, which can achieve at least one of the following technical effects: (1) high inhibitory activity against PARP1 enzyme; (2) selective inhibition of PARP1 enzyme, with high selectivity for other PARP family enzymes such as PARP2, PARP5a, and PARP5b; (3) strong inhibitory activity against homologous recombination-deficient tumor cells, and weak inhibitory effect on non-homologous recombination-deficient cells; (4) excellent pharmacokinetic properties (e.g., good bioavailability, suitable half-life and duration of action); (5) excellent safety (lower toxicity and/or fewer side effects, wider therapeutic window), etc. The PARP1 targeted compounds described in the present invention are surprisingly able to penetrate the blood-brain barrier (BBB) and can be used to treat diseases and conditions occurring in central nervous system tissues (e.g., brain and spinal cord).

术语定义:Definition of terms:

除非在下文中另有定义,本文中所用的所有技术术语和科学术语的含义意图与本领域技术人员通常所理解的相同。术语“包括”、“包含”、“具有”、“含有”或“涉及”及其在本文中的其它变体形式为包含性的或开放式的,且不排除其它未列举的元素或方法步骤。本领域技术人员应当理解,上述术语如“包括”涵盖“由…组成”的含义。Unless otherwise defined below, the meanings of all technical and scientific terms used herein are intended to be the same as those commonly understood by those skilled in the art. The terms "include," "comprises," "has," "contains," or "involves," and other variations thereof herein, are inclusive or open-ended and do not exclude other unlisted elements or method steps. It should be understood by those skilled in the art that the above terms, such as "includes," encompass the meaning of "consisting of."

在本发明中,“一”、“一个”、“该”、“至少一个”和“一个或多个”可互换使用。因此,例如,包含“一种”药学上可接受的赋型剂的组合物,可以被解释为表示该组合物包括“一种或多种”药学上可接受的赋型剂。In the present invention, "a", "an", "the", "at least one" and "one or more" are used interchangeably. Thus, for example, a composition comprising "a" pharmaceutically acceptable excipient can be interpreted as indicating that the composition includes "one or more" pharmaceutically acceptable excipients.

当公开了数值范围的下限和上限时,落入该范围中的任何数值和任何包括的范围都被具体公开。特别地,本文公开的值的每个取值范围(以形式“约a至b”,或同等的,“大约a至b”,或同等的,“约a-b”),应理解为表示涵盖于较宽范围中的每个数值和范围。When the lower and upper limits of a numerical range are disclosed, any value and any included range falling within the range are specifically disclosed. In particular, each range of values disclosed herein (in the form of "about a to b", or equivalently, "approximately a to b", or equivalently, "about a-b"), should be understood to represent each value and range encompassed in the broader range.

例如,表述“C1-8”应理解为涵盖其中的任意亚范围以及每个点值,例如C2-5、C3-4、C1-2、C1-3、C1-4、C1-5等,以及C1、C2、C3、C4、C5、C6等。例如,表述“C2-8”也应当以类似的方式理解,例如可以涵盖包含于其中的任意亚范围和点值,例如C6-8、C6-7、C7-8等以及C3、C4、C5、C6、C7、C8等。又例如,表述“3-6元”应理解为涵盖其中的任意亚范围以及的每个点值,例如3-4元、3-5元、3-6元、4-5元、4-6元等以及3、4、5、6元等。For example, the expression "C 1-8 " should be understood to include any sub-ranges and each point value therein, such as C 2-5 , C 3-4 , C 1-2 , C 1-3 , C 1-4 , C 1-5 , etc., as well as C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , etc. For example, the expression "C 2-8 " should also be understood in a similar manner, for example, it can include any sub-ranges and point values contained therein, such as C 6-8 , C 6-7 , C 7-8 , etc., as well as C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , etc. For another example, the expression "3-6 yuan" should be understood to include any sub-ranges and each point value therein, such as 3-4 yuan, 3-5 yuan, 3-6 yuan, 4-5 yuan, 4-6 yuan, etc., as well as 3, 4, 5, 6 yuan, etc.

在本发明中,除非另有说明,卤素是指氟、氯、溴或碘。In the present invention, unless otherwise specified, halogen means fluorine, chlorine, bromine or iodine.

在本发明中,除非另有说明,“烷基”包括直链或支链的一价饱和烃基。例如烷基包括甲基、乙基、丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、3-(2-甲基)丁基、2-戊基、2-甲基丁基、新戊基、正己基、2-己基、2-甲基戊基等。类似的,“C1-4烷基”中的C1-4是指包含有1、2、3或4个碳原子的直链或支链形式排列的基团。In the present invention, unless otherwise specified, "alkyl" includes a linear or branched monovalent saturated hydrocarbon group. For example, alkyl includes methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 3-(2-methyl)butyl, 2-pentyl, 2-methylbutyl, neopentyl, n-hexyl, 2-hexyl, 2-methylpentyl, etc. Similarly, C1-4 in " C1-4 alkyl" refers to a group containing 1, 2, 3 or 4 carbon atoms in a linear or branched form.

在本发明中,除非另有说明,“环烷基”、“碳环”或“亚环烷基”是指饱和或部分饱和的,单环或多环(诸如双环)的非芳香族烃基。常见的环烷基包括(但不限于)单环环烷基,诸如环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环壬基、环丁烯、环戊烯、环己烯等;或双环环烷基,包括稠环、桥环或螺环,诸如双环[1.1.1]戊基、双环[2.2.1]庚基、双环[3.2.1]辛基、双环[5.2.0]壬基、十氢化萘基等。例如,“C 3-12环烷基”指具有3-12个环碳原子(如3、4、5、6、7、8、9、10、11或12个)的环烷基。本发明中的环烷基或亚环烷基任选地被一个或多个本发明所描述的取代基取代。In the present invention, unless otherwise specified, "cycloalkyl", "carbocycle" or "cycloalkylene" refers to a saturated or partially saturated, monocyclic or polycyclic (such as bicyclic) non-aromatic hydrocarbon group. Common cycloalkyl groups include (but are not limited to) monocyclic cycloalkyl groups, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclobutene, cyclopentene, cyclohexene, etc.; or bicyclic cycloalkyl groups, including fused rings, bridged rings or spiro rings, such as bicyclo[1.1.1]pentyl, bicyclo[2.2.1]heptyl, bicyclo[3.2.1]octyl, bicyclo[5.2.0]nonyl, decalinyl, etc. For example, "C 3-12 cycloalkyl" refers to a cycloalkyl group having 3-12 ring carbon atoms (such as 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12). The cycloalkyl or cycloalkylene group in the present invention is optionally substituted by one or more substituents described in the present invention.

在本发明中,除非另有说明,“卤代烷基”指上文所述的烷基,其中一个或多个氢 原子被卤素代替。例如,术语“C1-6卤代烷基”指任选地被一个或多个(如1-3个)卤素取代的C1-6烷基。本领域技术人员应当理解,当卤素取代基多于一个时,卤素可以相同也可以不同,并且可以位于相同或不同的C原子上。卤代烷基的实例有例如-CH2F、-CHF2、-CF3、-CCl3、-C2F5、-C2Cl5、-CH2CF3、-CH2Cl或-CH2CH2CF3等。本发明中的卤代烷基任选地被一个或多个本发明所描述的取代基取代。In the present invention, unless otherwise specified, "haloalkyl" refers to an alkyl group as described above, in which one or more hydrogen atoms are present. Atoms are replaced by halogen. For example, the term "C 1-6 haloalkyl" refers to a C 1-6 alkyl group optionally substituted by one or more (e.g., 1-3) halogens. It will be appreciated by those skilled in the art that when there are more than one halogen substituent, the halogens may be the same or different and may be located on the same or different C atoms. Examples of haloalkyl groups include, for example, -CH 2 F, -CHF 2 , -CF 3 , -CCl 3 , -C 2 F 5 , -C 2 Cl 5 , -CH 2 CF 3 , -CH 2 Cl or -CH 2 CH 2 CF 3. The haloalkyl group in the present invention is optionally substituted by one or more substituents described herein.

在本发明中,除非另有说明,“氟代烷基”指上文所述的烷基,其中一个或多个氢原子被氟原子代替。例如,术语“C1-4氟代烷基”指任选地被一个或多个(如1-3个)氟原子取代的C1-4烷基。本领域技术人员应当理解,当氟原子取代基多于一个时,氟原子可以相同也可以不同,并且可以位于相同或不同的C原子上。卤代烷基的实例有例如-CH2F、-CHF2、-CF3、-C2F5、-CH2CF3、-CH2CH2CF3等。本发明中的氟代烷基任选地被一个或多个本发明所描述的取代基取代。In the present invention, unless otherwise specified, "fluoroalkyl" refers to the alkyl group described above, wherein one or more hydrogen atoms are replaced by fluorine atoms. For example, the term "C 1-4 fluoroalkyl" refers to a C 1-4 alkyl group optionally substituted by one or more (e.g., 1-3) fluorine atoms. It will be understood by those skilled in the art that when there are more than one fluorine atom substituents, the fluorine atoms may be the same or different, and may be located on the same or different C atoms. Examples of haloalkyl groups include, for example, -CH 2 F, -CHF 2 , -CF 3 , -C 2 F 5 , -CH 2 CF 3 , -CH 2 CH 2 CF 3 , etc. The fluoroalkyl group in the present invention is optionally substituted by one or more substituents described in the present invention.

在本发明中,除非另有说明,“烯基”是指具有至少一个C=C双键的、直链或支链的脂肪族烃基。例如,“C2-4烯基”是指具有2至4个碳原子的烯基。常见的烯基包括(但不限于)乙烯基、丙烯基、正丁烯基、3-甲基丁-2-烯基、正戊烯基、正辛烯基、正癸烯基等。本发明中的烯基任选地被一个或多个本发明所描述的取代基取代。In the present invention, unless otherwise specified, "alkenyl" refers to a straight or branched aliphatic hydrocarbon group having at least one C=C double bond. For example, " C2-4 alkenyl" refers to an alkenyl group having 2 to 4 carbon atoms. Common alkenyl groups include (but are not limited to) vinyl, propenyl, n-butenyl, 3-methylbut-2-enyl, n-pentenyl, n-octenyl, n-decenyl, etc. The alkenyl group in the present invention is optionally substituted by one or more substituents described in the present invention.

在本发明中,除非另有说明,“取代的”是指基团中的一个或多个氢原子分别被相同的或者不同的取代基所取代。In the present invention, unless otherwise specified, "substituted" means that one or more hydrogen atoms in the group are replaced by the same or different substituents, respectively.

本发明还包括所有药学上可接受的同位素标记的化合物,其与本发明的化合物相同,除了一个或多个原子被具有相同原子序数但原子质量或质量数不同于在自然界中占优势的原子质量或质量数的原子替代。适合包含入本发明的化合物中的同位素的实例包括(但不限于)氢的同位素(例如氘(2H)、氚(3H));碳的同位素(例如13C及14C);氯的同位素(例如37Cl);碘的同位素(例如125I);氮的同位素(例如13N及15N);氧的同位素(例如17O及18O);磷的同位素(例如32P);及硫的同位素(例如34S)。The present invention also includes all pharmaceutically acceptable isotopically labeled compounds, which are identical to the compounds of the present invention except that one or more atoms are replaced by atoms having the same atomic number but an atomic mass or mass number different from the atomic mass or mass number predominant in nature. Examples of isotopes suitable for inclusion in the compounds of the present invention include, but are not limited to, isotopes of hydrogen (e.g., deuterium ( 2H ), tritium ( 3H )); isotopes of carbon (e.g., 13C and 14C ); isotopes of chlorine (e.g., 37Cl); isotopes of iodine (e.g., 125I ); isotopes of nitrogen (e.g., 13N and 15N ); isotopes of oxygen (e.g., 17O and 18O ); isotopes of phosphorus (e.g., 32P ); and isotopes of sulfur (e.g., 34S ).

在本申请中,“药物组合物”是指本发明化合物与本领域通常接受的用于将生物活性化合物输送至哺乳动物(例如人)的介质的制剂。该介质包括药学上可接受的载体。药物组合物的目的是促进生物体的给药,利于活性成分的吸收,进而发挥生物活性。In this application, "pharmaceutical composition" refers to a preparation of the compound of the present invention and a medium generally accepted in the art for delivering biologically active compounds to mammals (e.g., humans). The medium includes a pharmaceutically acceptable carrier. The purpose of the pharmaceutical composition is to promote administration of the organism, facilitate the absorption of the active ingredient, and thus exert biological activity.

在本申请中,“药学上可接受的载体”包括但不限于任何被相关的政府管理部门许可或为接受供人类或家畜使用的佐剂、载体、赋型剂、助流剂、增甜剂、稀释剂、防腐剂、染料/着色剂、矫味剂、表面活性剂、润湿剂、分散剂、助悬剂、稳定剂、等渗剂、溶剂或乳化剂。In this application, "pharmaceutically acceptable carrier" includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersant, suspending agent, stabilizer, isotonic agent, solvent or emulsifier approved or accepted by relevant governmental regulatory authorities for use in humans or livestock.

本文所使用术语“药物组合”、“药物联用”、“联合用药”、“施用其他治疗”、 “施用其他治疗剂”等是指通过混合或组合不止一种活性成分而获得的药物治疗,其包括活性成分的固定和不固定组合。术语“固定组合”是指以单个实体或单个剂型的形式向患者同时施用至少一种本文所述的化合物和至少一种协同药剂。术语“不固定组合”是指以单独实体的形式向患者同时施用、合用或以可变的间隔时间顺次施用至少一种本文所述的化合物和至少一种协同制剂。这些也应用到鸡尾酒疗法中,例如施用三种或更多种活性成分。As used herein, the terms "drug combination", "drug combination", "combination therapy", "administering other treatments", "Administering other therapeutic agents" and the like refers to medical treatments obtained by mixing or combining more than one active ingredient, including fixed and non-fixed combinations of active ingredients. The term "fixed combination" refers to the simultaneous administration of at least one compound described herein and at least one synergistic agent to a patient in the form of a single entity or a single dosage form. The term "non-fixed combination" refers to the simultaneous administration, combined administration or sequential administration of at least one compound described herein and at least one synergistic agent to a patient in the form of separate entities, at the same time, in combination or at variable intervals. These also apply to cocktail therapies, for example, the administration of three or more active ingredients.

在本发明中,除非另有说明,“肿瘤”包括但不限于白血病、胃肠间质瘤、组织细胞性淋巴瘤、非小细胞肺癌、小细胞肺癌、胰腺癌、肺鳞癌、肺腺癌、乳腺癌、前列腺癌、肝癌、皮肤癌、上皮细胞癌、宫颈癌、卵巢癌、肠癌、鼻炎癌、脑癌、骨癌、食道癌、黑色素瘤、肾癌、口腔癌、脑癌和中枢神经系统癌等疾病。In the present invention, unless otherwise specified, "tumor" includes but is not limited to leukemia, gastrointestinal stromal tumor, histiocytic lymphoma, non-small cell lung cancer, small cell lung cancer, pancreatic cancer, squamous cell carcinoma of the lung, adenocarcinoma of the lung, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, cervical cancer, ovarian cancer, intestinal cancer, rhinitis cancer, brain cancer, bone cancer, esophageal cancer, melanoma, kidney cancer, oral cancer, brain cancer and central nervous system cancer.

在本发明中,除非另有说明,“治疗”意指逆转、减轻、抑制这样的术语所应用的病症或病况或者这样的病症或病况的一种或多种症状的进展,或预防这样的病症或病况或者这样的病症或病况的一种或多种症状。In the present invention, unless otherwise indicated, "treating" means reversing, alleviating, inhibiting the progression of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.

在不违背本领域常识的基础上,上述各优选条件,可任意组合,即得本发明各较佳实例。Without violating the common sense in the art, the above-mentioned preferred conditions can be arbitrarily combined to obtain the preferred embodiments of the present invention.

附图说明BRIEF DESCRIPTION OF THE DRAWINGS

图1为化合物47和64给药后人乳腺癌436细胞NOD-SCID小鼠皮下瘤模型肿瘤体积变化曲线。FIG1 is a curve showing the change in tumor volume in a NOD-SCID mouse subcutaneous tumor model of human breast cancer 436 cells after administration of compounds 47 and 64.

图2为化合物47和64给药后人乳腺癌436细胞NOD-SCID小鼠皮下瘤模型肿瘤体积变化实物图。Figure 2 is a physical picture of the change in tumor volume in the human breast cancer 436 cell NOD-SCID mouse subcutaneous tumor model after administration of compounds 47 and 64.

图3为化合物11给药后乳腺癌MDA-MB-436裸小鼠模型肿瘤体积的变化图。FIG3 is a graph showing the changes in tumor volume in the breast cancer MDA-MB-436 nude mouse model after administration of compound 11.

图4为化合物11给药后对人乳腺癌MDA-MB-436小鼠模型颅内荧光强度的影响图。FIG4 is a graph showing the effect of compound 11 administration on the intracranial fluorescence intensity of the human breast cancer MDA-MB-436 mouse model.

图5为化合物11给药后对人乳腺癌MDA-MB-436小鼠生存期的影响图。FIG5 is a graph showing the effect of compound 11 administration on the survival of human breast cancer MDA-MB-436 mice.

图6为化合物11给药后对人乳腺癌MDA-MB-436小鼠体重的影响图。FIG6 is a graph showing the effect of compound 11 administration on the body weight of human breast cancer MDA-MB-436 mice.

具体实施方式DETAILED DESCRIPTION

下面将结合实施例对本发明的方案进行解释。本领域技术人员将会理解,下面的实施例仅用于说明本发明,而不应视为限定本发明的范围。实施例中未注明具体技术或条件的,按照本领域内的文献所描述的技术或条件或者按照产品说明书进行。The scheme of the present invention will be explained below in conjunction with embodiments. It will be appreciated by those skilled in the art that the following embodiments are only used to illustrate the present invention and should not be considered as limiting the scope of the present invention. Where specific techniques or conditions are not indicated in the embodiments, the techniques or conditions described in the literature in this area or the product specifications are followed.

本发明实施例中所用试剂和原料均市售可得。The reagents and raw materials used in the examples of the present invention are commercially available.

表1本发明中字母缩写及其含义

Table 1 Abbreviations and their meanings in the present invention

本发明所述化合物的结构是通过核磁共振(NMR)或质谱(MS)来确定的。NMR的测定是用Bruker AVANCE-400核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6)、氘代氯仿(CDCl3)、氘代甲醇(CD3OD)内标为四甲基硅烷(TMS)化学位移是以10-6(ppm)作为单位给出。The structures of the compounds of the present invention are determined by nuclear magnetic resonance (NMR) or mass spectrometry (MS). NMR measurements are performed using a Bruker AVANCE-400 nuclear magnetic spectrometer, with deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD) as the solvent, tetramethylsilane (TMS) as the internal standard, and chemical shifts are given in units of 10 -6 (ppm).

MS的测定用Agilent SQD(ESI)质谱仪(生产商:Agilent,信号:6110)。 MS was measured using an Agilent SQD (ESI) mass spectrometer (manufacturer: Agilent, signal: 6110).

HPLC的测定使用安捷伦1200DAD高压液相色谱仪(Sunfirc C18,150X 4.6mm,5wn,色谱柱)和Waters 2695-2996高压液相色谱仪(Gimini C18,150X 4.5mm,5ym色谱柱)。HPLC determination used Agilent 1200DAD high pressure liquid chromatograph (Sunfirc C18, 150X 4.6mm, 5wn, chromatographic column) and Waters 2695-2996 high pressure liquid chromatograph (Gimini C18, 150X 4.5mm, 5ym chromatographic column).

薄层层析硅胶板使用青岛海洋GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm-0.2mm,薄层层析分离纯化产品采用的规格是0.4mm-0.5mm硅胶板。The thin layer chromatography silica gel plate used was Qingdao Ocean GF254 silica gel plate. The silica gel plate used in thin layer chromatography (TLC) had a specification of 0.15mm-0.2mm, and the thin layer chromatography separation and purification product used a 0.4mm-0.5mm silica gel plate.

柱层析一般使用青岛海洋100-200、200-300目硅胶为载体。Column chromatography generally uses Qingdao Ocean 100-200, 200-300 mesh silica gel as the carrier.

以下实施例中无特殊说明,反应均在氩气氛围或氮气氛围下进行。氩气氛围或氮气氛围是指反应瓶连接一个约1L容积的氩气或氮气气球。氢气氛围是指反应瓶连接一个约1L容积的氢气气球。氢化反应通常抽真空,充入氢气,反复操作3次。Unless otherwise specified in the following examples, the reactions were carried out under an argon atmosphere or a nitrogen atmosphere. Argon atmosphere or nitrogen atmosphere means that the reaction bottle is connected to an argon or nitrogen balloon with a volume of about 1 L. Hydrogen atmosphere means that the reaction bottle is connected to a hydrogen balloon with a volume of about 1 L. The hydrogenation reaction is usually evacuated, filled with hydrogen, and the operation is repeated 3 times.

中间体int-1:7-溴-6-甲氧基-1,5-萘吡啶-3-羧酸乙酯
Intermediate int-1: 7-bromo-6-methoxy-1,5-naphthyridine-3-carboxylic acid ethyl ester

第一步:氮气保护下加入int-1a(500g,1.99mol)和甲醇2.5L,降温至0-5℃。在0-5℃滴加甲醇钠(118g)的甲醇(1.0L)溶液。滴加完全后升至室温搅拌1小时。向反应体系中加入水(2.0L),搅拌30min后于40℃下减压浓缩,浓缩至不出液,后加入乙酸乙酯(4.0L),搅拌分层,水层加入乙酸乙酯萃取,分液。合并有机层加入饱和食盐水洗涤,分液,有机层于40℃下减压浓缩至得到白色固体int-1b(480g)。Step 1: Add int-1a (500g, 1.99mol) and 2.5L of methanol under nitrogen protection, and cool to 0-5°C. Add a solution of sodium methoxide (118g) in methanol (1.0L) dropwise at 0-5°C. After complete addition, warm to room temperature and stir for 1 hour. Add water (2.0L) to the reaction system, stir for 30min, and then concentrate under reduced pressure at 40°C until no liquid is discharged. Then add ethyl acetate (4.0L), stir and separate, add ethyl acetate to the aqueous layer for extraction, and separate the layers. Combine the organic layers, add saturated brine to wash, separate the layers, and concentrate the organic layer under reduced pressure at 40°C to obtain int-1b (480g) as a white solid.

第二步:称取化合物int-1b(475g,1.93mol)加入DMF(2.85L)中,滴加DMF-DMA(2.85L)。加完后升温至100℃搅拌2h。反应完全后降温至70-80℃,减压浓缩,浓缩至不出液,后加入水中,搅拌析出。降温至20-30℃,搅拌1h后过滤,滤饼于70℃真空干燥箱中干燥至恒重,得红色固体int-1c(612g,收率95.3%)。Step 2: Weigh compound int-1b (475 g, 1.93 mol) and add it to DMF (2.85 L), then add DMF-DMA (2.85 L) dropwise. After adding, heat to 100 °C and stir for 2 h. After the reaction is complete, cool to 70-80 °C, concentrate under reduced pressure until no liquid is produced, then add to water and stir to precipitate. Cool to 20-30 °C, stir for 1 h, then filter, and dry the filter cake in a vacuum oven at 70 °C to constant weight to obtain a red solid int-1c (612 g, yield 95.3%).

第三步:将化合物int-1c(500g,1.91mol)加入THF(2.56L)中,搅拌溶清。向反应体系中滴加高碘酸钠(805g,3.72mol)的水溶液(2.56L)。室温搅拌2-4h,反应结束后向反应体系中加入乙酸乙酯(4.0L)和水(4.0L),搅拌分层,水层乙酸乙酯(2.0L)萃取两次。有机层合并后依次加入饱和硫代硫酸钠溶液、饱和食盐水洗涤,有机层于40-45℃下减压浓缩至无馏分,得到500g油状物int-1d,直接用于下一步。Step 3: Add compound int-1c (500g, 1.91mol) to THF (2.56L) and stir to dissolve. Add an aqueous solution (2.56L) of sodium periodate (805g, 3.72mol) to the reaction system. Stir at room temperature for 2-4h. After the reaction is completed, add ethyl acetate (4.0L) and water (4.0L) to the reaction system, stir and separate the layers, and extract the aqueous layer with ethyl acetate (2.0L) twice. After the organic layers are combined, add saturated sodium thiosulfate solution and saturated brine to wash in turn. The organic layer is concentrated under reduced pressure at 40-45°C until there is no fraction to obtain 500g of oily int-1d, which is directly used in the next step.

第四步:将化合物int-1d(512g,1.69mol)和int-1e(1457.0g,7.61mol)加入无水乙醇(7.5L)中,搅拌溶清,室温下分批次向反应体系中加入SnCl2(1815.0g,9.57mol)。加毕后升温至回流,搅拌1-2h。将反应体系降温至45-50℃,减压浓缩至无馏分,向体系中加入乙酸乙酯搅拌溶解,后用饱和碳酸氢钠调节pH=7-8,过程剧烈放气,析出大量固体,反应液离心,收集滤液静置分层,有机层于40-45℃下减压浓缩至无馏分,加入200-300目硅胶过柱纯化得到絮状固体int-1(230g,收率36.5%)。 Step 4: Add compounds int-1d (512 g, 1.69 mol) and int-1e (1457.0 g, 7.61 mol) to anhydrous ethanol (7.5 L), stir to dissolve, and add SnCl 2 (1815.0 g, 9.57 mol) to the reaction system in batches at room temperature. After the addition, heat to reflux and stir for 1-2 h. Cool the reaction system to 45-50 ° C, concentrate under reduced pressure until there is no fraction, add ethyl acetate to the system and stir to dissolve, and then adjust the pH to 7-8 with saturated sodium bicarbonate. During the process, gas is released violently and a large amount of solid is precipitated. The reaction solution is centrifuged, and the filtrate is collected and allowed to stand for stratification. The organic layer is concentrated under reduced pressure at 40-45 ° C until there is no fraction, and 200-300 mesh silica gel is added to purify by column to obtain flocculent solid int-1 (230 g, yield 36.5%).

中间体int-2:N-甲基-1',2',3',6'-四氢-[3,4'-联吡啶]-6-甲酰胺
Intermediate int-2: N-methyl-1',2',3',6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide

第一步:在7ml二氧六环、3ml无水乙醇和4ml水的混合溶剂中加入化合物int-2a(1g,4.6mmol)、int-2b(1.7g,5.5mmol)、Pd(dppf)Cl2(0.3g,0.46mmol)和碳酸钾(1.6g,11.5mmol),然后置换氮气三次,在氮气保护下90℃下反应2h。TLC检测反应完全后,将反应降至室温,加入30ml二氯甲烷和20ml水,在分液漏斗中分层。水相用二氯甲烷萃取两次,合并有机相后再分别水洗、饱和食盐水洗、无水硫酸钠干燥、过滤、旋干。所得粗品经过柱层析纯化得到化合物int-2c(1g,白色固体)。Step 1: Add compound int-2a (1g, 4.6mmol), int-2b (1.7g, 5.5mmol), Pd(dppf)Cl 2 (0.3g, 0.46mmol) and potassium carbonate (1.6g, 11.5mmol) to a mixed solvent of 7ml dioxane, 3ml anhydrous ethanol and 4ml water, then replace nitrogen three times, and react at 90°C for 2h under nitrogen protection. After TLC detection, the reaction is cooled to room temperature, 30ml dichloromethane and 20ml water are added, and the layers are separated in a separatory funnel. The aqueous phase is extracted twice with dichloromethane, and the organic phases are combined and then washed with water, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and dried. The crude product is purified by column chromatography to obtain compound int-2c (1g, white solid).

第二步:在100ml反应瓶中加入int-2c(1g,3mmol)、甲胺水溶液(5g,161.3mmol)和无水甲醇(20ml),室温下搅拌过夜。TLC监测反应完全后,将反应液减压浓缩干得到化合物int-2d(0.8g,白色固体)。Step 2: Add int-2c (1 g, 3 mmol), methylamine aqueous solution (5 g, 161.3 mmol) and anhydrous methanol (20 ml) to a 100 ml reaction bottle and stir overnight at room temperature. After the reaction is complete as monitored by TLC, the reaction solution is concentrated under reduced pressure to dryness to obtain compound int-2d (0.8 g, white solid).

第三步:将化合物int-2d(0.5g,1.5mmol)加入到10ml无水甲醇中,随后再加入10ml 4mol/L盐酸二氧六环溶液,室温搅拌0.5-1h,TLC监测反应完全后,将反应液减压浓缩干得到化合物int-2(0.5g,白色固体)。Step 3: Add compound int-2d (0.5 g, 1.5 mmol) into 10 ml of anhydrous methanol, and then add 10 ml of 4 mol/L hydrochloric acid dioxane solution. Stir at room temperature for 0.5-1 h. After the reaction is complete as monitored by TLC, concentrate the reaction solution under reduced pressure to obtain compound int-2 (0.5 g, white solid).

中间体int-3和中间体int-4的制备参考中间体int-2。The preparation of intermediates int-3 and int-4 refers to intermediate int-2.

表2中间体int-3~int-4
Table 2 Intermediates int-3 to int-4

中间体int-5:N-环丙基-1',2',3',6'-四氢-[3,4'-联吡啶]-6-甲酰胺
Intermediate int-5: N-cyclopropyl-1',2',3',6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide

第一步:称取int-2c(1.4g,4.5mmol),加入20mL MeOH溶解,加入5mL水,加入氢氧化锂(570mg,13.5mmol),室温反应12h,TLC监测反应,反应完全后加入2M HCl调pH至6,加入(3x 25mL)EA萃取,合并有机相,无水硫酸钠干燥后,真 空旋干,得到产物int-5a(460mg,淡黄色固体)。Step 1: Weigh int-2c (1.4 g, 4.5 mmol), add 20 mL MeOH to dissolve, add 5 mL water, add lithium hydroxide (570 mg, 13.5 mmol), react at room temperature for 12 h, monitor the reaction by TLC, add 2 M HCl to adjust the pH to 6 after the reaction is complete, add (3 x 25 mL) EA to extract, combine the organic phases, dry over anhydrous sodium sulfate, and vacuum dry. The product was spin dried in vacuo to obtain int-5a (460 mg, light yellow solid).

第二步:称取int-5a(180mg,0.6mmol),EDCI(144mg,0.75mmol),HOBT(100mg,0.75mmol)2mL DMF,N-甲基吗啉(290mg,3.2mmol),环丙胺(34mg,0.6mmol),温反应12h,TLC监测,原料消耗完全后,加水稀释,EA萃取,有机相用水洗5次,无水硫酸钠干燥,旋干,得到粗产物int-5b(180mg,黄色油状液体)。Step 2: Weigh int-5a (180 mg, 0.6 mmol), EDCI (144 mg, 0.75 mmol), HOBT (100 mg, 0.75 mmol) 2 mL DMF, N-methylmorpholine (290 mg, 3.2 mmol), cyclopropylamine (34 mg, 0.6 mmol), react at warm temperature for 12 h, monitor by TLC, dilute with water after the raw materials are completely consumed, extract with EA, wash the organic phase with water 5 times, dry over anhydrous sodium sulfate, and spin dry to obtain the crude product int-5b (180 mg, yellow oily liquid).

第三步:将int-5b的粗产物(171mg,0.5mmol)加入甲醇(5mL)溶解,随后加入4M HCl 1,4-二氧六环(0.9mL)溶液,室温反应12h,TLC监测反应,反应完全后加入碳酸钾,搅拌30分钟后,过滤除去碳酸钾,得到化合物int-5的粗产物(180mg,黄褐色固体)。MS/ESI[M+H]+=244.1。Step 3: The crude product of int-5b (171 mg, 0.5 mmol) was dissolved in methanol (5 mL), followed by addition of 4M HCl 1,4-dioxane (0.9 mL) solution, and reacted at room temperature for 12 h. The reaction was monitored by TLC. After the reaction was complete, potassium carbonate was added, stirred for 30 minutes, and potassium carbonate was removed by filtration to obtain the crude product of compound int-5 (180 mg, yellow-brown solid). MS/ESI[M+H] + =244.1.

中间体int-6~int-8的制备参考中间体int-5。The preparation of intermediates int-6 to int-8 refers to intermediate int-5.

表3中间体int-6~int-8
Table 3 Intermediates int-6 to int-8

中间体int-9:N-甲基-5-(哌嗪-1-基)吡啶酰胺
Intermediate int-9: N-methyl-5-(piperazine-1-yl)pyridineamide

第一步:将化合物int-2a(5.66g,26.21mmol,1.05eq),化合物int-9a(4.65g,25.0mmol,1.00eq),Cs2CO3(16.27g,50.0mmol,2.00eq)和RuPhos-Pd-G3(1.04g,1.25mmol,0.05eq)加入1,4-二氧六环(50mL)中,在氮气气氛下120℃搅拌过夜。用LCMS检测反应完成后让混合物冷却到室温。将反应液用水(100mL)稀释,然后用乙酸乙酯(2x 100mL)提取。合并后的有机层用盐水(2x 50mL)洗涤,用无水Na2SO4 干燥,过滤,滤液在减压下浓缩。残留物经硅胶柱层析纯化得到化合物int-9b(白色固体,4.5g)。MS/ESI[M+H]+=322.1。Step 1: Add compound int-2a (5.66 g, 26.21 mmol, 1.05 eq), compound int-9a (4.65 g, 25.0 mmol, 1.00 eq), Cs 2 CO 3 (16.27 g, 50.0 mmol, 2.00 eq) and RuPhos-Pd-G3 (1.04 g, 1.25 mmol, 0.05 eq) to 1,4-dioxane (50 mL) and stir overnight at 120 ° C under nitrogen atmosphere. After the reaction is completed by LCMS, the mixture is allowed to cool to room temperature. The reaction solution is diluted with water (100 mL) and then extracted with ethyl acetate (2 x 100 mL). The combined organic layer is washed with brine (2 x 50 mL) and washed with anhydrous Na 2 SO 4 The mixture was dried, filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to obtain compound int-9b (white solid, 4.5 g). MS/ESI [M+H] + = 322.1.

第二步:将化合物int-9b(1.92g,6.0mmol,1.0eq)加入甲胺(8mL,25-30wt%水溶液)在甲醇(7mL)的溶液中,在氮气气氛下室温搅拌3h。反应通过LCMS进行监测。在室温下用饱和氯化铵水溶液(30mL)猝灭反应。所得混合物用二氯甲烷(3x 50mL)萃取。合并后的有机层用盐水(2x 50mL)洗涤,用无水Na2SO4干燥。过滤后,在减压下浓缩滤液,得到浅黄色油状的化合物int-9c(1.65g)。MS/ESI[M+H]+=321.3。Step 2: Compound int-9b (1.92 g, 6.0 mmol, 1.0 eq) was added to a solution of methylamine (8 mL, 25-30 wt% aqueous solution) in methanol (7 mL) and stirred at room temperature for 3 h under a nitrogen atmosphere. The reaction was monitored by LCMS. The reaction was quenched with a saturated aqueous solution of ammonium chloride (30 mL) at room temperature. The resulting mixture was extracted with dichloromethane (3 x 50 mL). The combined organic layer was washed with brine (2 x 50 mL) and dried over anhydrous Na 2 SO 4. After filtration, the filtrate was concentrated under reduced pressure to obtain compound int-9c (1.65 g) as a light yellow oil. MS/ESI[M+H] + =321.3.

第三步:将化合物int-9c(482mg,1.50mmol,1.00eq)和HCl的1,4-二恶烷溶液(3.7mL,15.0mmol,10.0eq,4.0M)在室温下搅拌2小时。LCMS监控反应完成后将所得混合物减压浓缩,得到黄色固体的化合物int-9(392mg,粗品)。将粗产物直接用于下一步骤中,而无需进一步纯化。MS/ESI[M+H]+=221.3。Step 3: Compound int-9c (482 mg, 1.50 mmol, 1.00 eq) and 1,4-dioxane solution of HCl (3.7 mL, 15.0 mmol, 10.0 eq, 4.0 M) were stirred at room temperature for 2 hours. After the reaction was completed, the resulting mixture was concentrated under reduced pressure by LCMS monitoring to obtain compound int-9 (392 mg, crude product) as a yellow solid. The crude product was used directly in the next step without further purification. MS/ESI[M+H] + =221.3.

中间体int-10和int-11的制备参考中间体int-9。The preparation of intermediates int-10 and int-11 refers to intermediate int-9.

表4中间体int-10和int-11
Table 4 Intermediates int-10 and int-11

中间体INT12:N-环丙基-5-(哌嗪-1-基)吡啶酰胺
Intermediate INT12: N-cyclopropyl-5-(piperazine-1-yl)pyridineamide

第一步:称取int-9b(1.4g,4.5mmol),加入20mL MeOH溶解,加入5mL水,加入氢氧化锂(570mg,13.5mmol),室温反应12h,TLC监测反应,反应完全后加入2M HCl调pH至6,加入(3x 25mL)EA萃取,合并有机相,无水硫酸钠干燥后,真空旋干,得到产物int-12a(845mg)。Step 1: Weigh int-9b (1.4 g, 4.5 mmol), add 20 mL MeOH to dissolve, add 5 mL water, add lithium hydroxide (570 mg, 13.5 mmol), react at room temperature for 12 h, monitor the reaction by TLC, add 2 M HCl to adjust the pH to 6 after the reaction is complete, add (3x 25 mL) EA to extract, combine the organic phases, dry over anhydrous sodium sulfate, and vacuum dry to obtain the product int-12a (845 mg).

第二步:称取int-12a(180mg,0.6mmol),EDCI(144mg,0.75mmol),HOBT(100mg,0.75mmol)2mL DMF,N-甲基吗啉(290mg,3.2mmol),环丙胺(34mg,0.6mmol),室温反应12h,TLC监测,原料消耗完全后,加水稀释,EA萃取,有机相用水洗5次,无水硫酸钠干燥,减压浓缩除去溶剂,得到粗产物int-12b(165mg)。 MS/ESI[M+H]+=347.2。Step 2: Weigh int-12a (180 mg, 0.6 mmol), EDCI (144 mg, 0.75 mmol), HOBT (100 mg, 0.75 mmol) 2 mL DMF, N-methylmorpholine (290 mg, 3.2 mmol), cyclopropylamine (34 mg, 0.6 mmol), react at room temperature for 12 h, monitor by TLC, after the raw materials are completely consumed, dilute with water, extract with EA, wash the organic phase with water 5 times, dry over anhydrous sodium sulfate, and concentrate under reduced pressure to remove the solvent to obtain the crude product int-12b (165 mg). MS/ESI [M+H] + = 347.2.

第三步:将int-12b的粗产物(150mg,0.4mmol)加入5mL MeOH溶解,随后加入0.9mL 4M HCl in dioxane溶液,室温反应12h,TLC监测反应,反应完全后加入碳酸钾,搅拌30分钟后,过滤除去碳酸钾,得到化合物int-12的粗产物(120mg,黄褐色固体)。MS/ESI[M+H]+=247.1。Step 3: The crude product of int-12b (150 mg, 0.4 mmol) was dissolved in 5 mL MeOH, followed by the addition of 0.9 mL 4M HCl in dioxane solution, and the reaction was carried out at room temperature for 12 h. The reaction was monitored by TLC. After the reaction was complete, potassium carbonate was added, and after stirring for 30 minutes, potassium carbonate was removed by filtration to obtain the crude product of compound int-12 (120 mg, yellow-brown solid). MS/ESI[M+H] + =247.1.

中间体int-13~int-15的制备方法参考中间体int-12。The preparation method of intermediates int-13 to int-15 refers to intermediate int-12.

表5中间体int-13~int-15
Table 5 Intermediates int-13 to int-15

中间体int-16:N-甲基-5-(((2R,3S)-2-甲基氮杂环丁烷-3-基)氧基)吡啶酰胺
Intermediate int-16: N-methyl-5-(((2R,3S)-2-methylazetidin-3-yl)oxy)picolinamide

第一步:将化合物int-16a(2.0g,0.01305mol)、化合物int-16b(3.393g,0.0195mol)加入四氢呋喃(200mL)中,将反应液降温0℃,加入三苯基磷(20.54g,0.0783mol),加完氮气置换3次,搅拌1h,加入DEAD(11.36g,0.06525mol),加完,室温搅拌12h,反应结束后,将反应液减压浓缩,浓缩物柱层析(PE:EA=10:1~1:1),收集产物,得粗品化合物int-16c,LCMS=341.1。Step 1: Add compound int-16a (2.0 g, 0.01305 mol) and compound int-16b (3.393 g, 0.0195 mol) to tetrahydrofuran (200 mL), cool the reaction solution to 0°C, add triphenylphosphine (20.54 g, 0.0783 mol), replace with nitrogen three times, stir for 1 hour, add DEAD (11.36 g, 0.06525 mol), stir at room temperature for 12 hours, after the reaction is completed, concentrate the reaction solution under reduced pressure, and the concentrate is subjected to column chromatography (PE:EA=10:1~1:1), and the product is collected to obtain a crude compound int-16c, LCMS=341.1.

第二步:将化合物int-16c(4.5g,0.0146mol)加入甲醇(20mL)中,加入甲胺醇溶液(20mL),加完,室温反应1h,反应结束后,将反应液减压浓缩,得粗品化合物int-16d,LCMS=340.2。Step 2: Add compound int-16c (4.5 g, 0.0146 mol) to methanol (20 mL), add methylamine alcohol solution (20 mL), and react at room temperature for 1 h. After the reaction is completed, concentrate the reaction solution under reduced pressure to obtain a crude compound int-16d, LCMS = 340.2.

第三步:将化合物int-16d(4.5g,0.0146mol)加入二氯甲烷(20mL)中,加入盐酸二氧六环溶液(20mL),加完,室温反应1h,反应结束后,将反应液减压浓缩,向 浓缩物中加入乙酸乙酯打浆,过滤,收集滤固,得化合物int-16盐酸盐,LCMS=240.2。Step 3: Add compound int-16d (4.5 g, 0.0146 mol) to dichloromethane (20 mL), add dioxane hydrochloride solution (20 mL), and react at room temperature for 1 h. After the reaction is completed, the reaction solution is concentrated under reduced pressure and Ethyl acetate was added to the concentrate for slurrying, and the solid was collected by filtration to obtain compound int-16 hydrochloride, LCMS=240.2.

中间体int-17:(6-甲氧基-7-((三氟甲基)硫代)-1,5-萘啶-3-基)甲基甲磺酸盐
Intermediate int-17: (6-methoxy-7-((trifluoromethyl)thio)-1,5-naphthyridin-3-yl)methyl methanesulfonate

第一步:将化合物int-1(3.0g,9.64mmol)溶于无水甲苯(30mL)中,加入化合物int-17a(4.0g,18.34mmol),Pd2(dba)3(1.0g,1.05mmol),Xantphos(1.1g,1.9mmol),DIPEA(4.0mL,24.24mmol),N2置换三次后,在110℃下搅拌12h。将反应液冷却后缓慢倒入100mL饱和氯化铵溶液中淬,EA萃取4*50mL,硫酸钠干燥,柱层析分离(PE/EA=5/1)。将得到的化合物溶于无水EtOH(30mL)中,加入NaOEt(1.3g,19.11mmol),于25℃下搅拌1h。将反应液缓慢倒入HCl(1.0M,50mL)中淬灭,EA萃取5*30mL,硫酸钠干燥,浓缩后用EA打浆,得到化合物int-17b(2.5g,crude)LC-MS:ESI[M+H]+=265.2。Step 1: Dissolve compound int-1 (3.0 g, 9.64 mmol) in anhydrous toluene (30 mL), add compound int-17a (4.0 g, 18.34 mmol), Pd 2 (dba) 3 (1.0 g, 1.05 mmol), Xantphos (1.1 g, 1.9 mmol), DIPEA (4.0 mL, 24.24 mmol), replace with N 2 three times, and stir at 110 ° C for 12 h. After cooling the reaction solution, slowly pour it into 100 mL saturated ammonium chloride solution for quenching, extract with EA 4*50 mL, dry with sodium sulfate, and separate by column chromatography (PE/EA=5/1). The obtained compound is dissolved in anhydrous EtOH (30 mL), NaOEt (1.3 g, 19.11 mmol) is added, and stirred at 25 ° C for 1 h. The reaction solution was slowly poured into HCl (1.0 M, 50 mL) for quenching, extracted with EA 5*30 mL, dried over sodium sulfate, concentrated and slurried with EA to obtain compound int-17b (2.5 g, crude) LC-MS: ESI [M+H] + = 265.2.

第二步:将化合物int-17b(2.5g,9.43mmol)溶于DCM(30mL)中,25℃下加入化合物int-17c(2.7g,8.2mmol),在20℃下搅拌1h。浓缩过柱(PE/EA=1/5)得到黄色油状化合物int-17d(0.61g,1.84mmol,20%)。LC-MS:ESI[M+H]+=333.2。Step 2: Dissolve compound int-17b (2.5 g, 9.43 mmol) in DCM (30 mL), add compound int-17c (2.7 g, 8.2 mmol) at 25°C, and stir at 20°C for 1 h. Concentrate and pass through a column (PE/EA=1/5) to obtain yellow oily compound int-17d (0.61 g, 1.84 mmol, 20%). LC-MS: ESI [M+H] + = 333.2.

第三步:将化合物int-17d(0.61g,1.84mmol)溶于DCM(20mL)中,0℃下缓慢滴加DIBAL-H(1.0M,5.0mL,5.0mmol),在0℃下搅拌1h。将反应液缓慢倒入50mL饱和氯化铵溶液中淬灭,硅藻土抽滤后EA萃取4*30mL,硫酸钠干燥,得到黄色油状化合物int-17e(0.45g,crude)。LC-MS:ESI[M+H]+=291.1。Step 3: Dissolve the compound int-17d (0.61 g, 1.84 mmol) in DCM (20 mL), slowly add DIBAL-H (1.0 M, 5.0 mL, 5.0 mmol) at 0°C, and stir at 0°C for 1 h. Slowly pour the reaction solution into 50 mL of saturated ammonium chloride solution to quench, filter with diatomaceous earth, extract with 4*30 mL of EA, and dry with sodium sulfate to obtain a yellow oily compound int-17e (0.45 g, crude). LC-MS: ESI [M+H] + = 291.1.

第四步:将化合物int-17e(0.45g,1.55mmol)溶于DCM(30mL)中,0℃下滴加TEA(0.7mL,7.36mmol),MsCl(0.5mL,3.6mmol),0℃搅拌1h。在反应液缓慢倒入30ml饱和碳酸氢钠溶液淬灭,DCM萃取3*30ml,硫酸钠干燥,抽滤减压浓缩得到化合物int-17(0.6g,crude)。LC-MS:ESI[M+H]+=369.2。Step 4: Compound int-17e (0.45 g, 1.55 mmol) was dissolved in DCM (30 mL), TEA (0.7 mL, 7.36 mmol) and MsCl (0.5 mL, 3.6 mmol) were added dropwise at 0°C, and stirred at 0°C for 1 h. 30 ml of saturated sodium bicarbonate solution was slowly poured into the reaction solution for quenching, and DCM was extracted for 3*30 ml, dried over sodium sulfate, filtered and concentrated under reduced pressure to obtain compound int-17 (0.6 g, crude). LC-MS: ESI [M+H] + = 369.2.

中间体int-18:(6-甲氧基-7-(甲硫基)-1,5-萘啶-3-基)甲基甲磺酸盐
Intermediate int-18: (6-methoxy-7-(methylthio)-1,5-naphthyridin-3-yl)methyl methanesulfonate

第一步:将化合物int-1(5.0g,16.07mmol)置于封管中,加入DMSO(50mL)溶 解,加入CuI(3.0g,15.7mmol),Zn(OAc)2(6.0g,32.78mmol),N2置换三次后,在150℃下搅拌15h。将反应液冷却后缓慢倒入100mL饱和氯化铵溶液中淬灭,经硅藻土抽滤,EA萃取5*50mL,硫酸钠干燥,柱层析分离(PE/EA=5/1)得到黄色固体化合物int-18a(0.3g,1.08mmol,收率为8%)。LC-MS:ESI[M+H]+=279.2。Step 1: Place compound int-1 (5.0 g, 16.07 mmol) in a sealed tube and add DMSO (50 mL) to dissolve. After solution, CuI (3.0 g, 15.7 mmol) and Zn(OAc) 2 (6.0 g, 32.78 mmol) were added, and N 2 was replaced three times, and then stirred at 150°C for 15 h. After cooling, the reaction solution was slowly poured into 100 mL of saturated ammonium chloride solution for quenching, filtered through diatomaceous earth, extracted with EA 5*50 mL, dried with sodium sulfate, and separated by column chromatography (PE/EA=5/1) to obtain a yellow solid compound int-18a (0.3 g, 1.08 mmol, yield 8%). LC-MS: ESI[M+H] + =279.2.

第二步:将化合物int-18a(0.3g,1.08mmol)溶于DCM(20mL)中,0℃下缓慢滴加DIBAL-H(1.0M,5.0mL,5.0mmol),在0℃下搅拌1h。将反应液缓慢倒入50mL饱和氯化铵溶液中淬灭,硅藻土抽滤后EA萃取4*30mL,硫酸钠干燥,得到黄色油状化合物int-18b(0.3g,crude)。LC-MS:ESI[M+H]+=237.2。Step 2: Dissolve compound int-18a (0.3 g, 1.08 mmol) in DCM (20 mL), slowly add DIBAL-H (1.0 M, 5.0 mL, 5.0 mmol) at 0°C, and stir at 0°C for 1 h. Slowly pour the reaction solution into 50 mL saturated ammonium chloride solution to quench, filter with diatomaceous earth, extract with 4*30 mL EA, and dry with sodium sulfate to obtain yellow oily compound int-18b (0.3 g, crude). LC-MS: ESI [M+H] + = 237.2.

第三步:将化合物int-18b(0.3g,1.26mmol)溶于DCM(10mL)中,0℃下滴加TEA(0.5mL,4.32mmol),MsCl(0.3mL,2.62mmol),0℃搅拌1h。在反应液缓慢倒入20ml饱和碳酸氢钠溶液淬灭,DCM萃取3*30ml,硫酸钠干燥,抽滤减压浓缩得到化合物int-18(0.3g,crude)。LC-MS:ESI[M+H]+=315.2。Step 3: Dissolve compound int-18b (0.3 g, 1.26 mmol) in DCM (10 mL), add TEA (0.5 mL, 4.32 mmol) and MsCl (0.3 mL, 2.62 mmol) dropwise at 0°C, and stir at 0°C for 1 h. Slowly pour 20 ml of saturated sodium bicarbonate solution into the reaction solution to quench, extract with 3*30 ml of DCM, dry with sodium sulfate, filter and concentrate under reduced pressure to obtain compound int-18 (0.3 g, crude). LC-MS: ESI [M+H] + = 315.2.

中间体int-19:(R)-N-甲基-1,2,3,4,4a,5-六氢吡嗪并[1,2-d]吡啶并[2,3-b][1,4]恶嗪-8-羧酰胺
Intermediate int-19: (R)-N-methyl-1,2,3,4,4a,5-hexahydropyrazino[1,2-d]pyrido[2,3-b][1,4]oxazine-8-carboxamide

第一步:将化合物int-19a(4.2g,19.4mmol)溶于二氯甲烷(100mL),加入三乙胺(117mg,38.9mmol),叔丁基二甲基氯硅烷(2.17g,14.39mmol),4-二甲氨基吡啶(3.94g,0.94mmol),搅拌反应14小时,反应液减压浓缩,残余物用硅胶柱色谱法纯化得到化合物int-19b(5.2g)。LC-MS:ESI[M+H]+=331.2。Step 1: Dissolve compound int-19a (4.2 g, 19.4 mmol) in dichloromethane (100 mL), add triethylamine (117 mg, 38.9 mmol), tert-butyldimethylsilyl chloride (2.17 g, 14.39 mmol), 4-dimethylaminopyridine (3.94 g, 0.94 mmol), stir and react for 14 hours, concentrate the reaction solution under reduced pressure, and purify the residue by silica gel column chromatography to obtain compound int-19b (5.2 g). LC-MS: ESI [M+H] + = 331.2.

第二步:将化合物int-19b(1.3g,5.55mmol),化合物int-3a(2.02g,6.1mmol),溶于1,4-二氧六环(15mL),加入甲磺酸(2-二环己基膦基-2',6'-二异丙氧基-1,1'-联苯基)(2-氨基-1,1'-联苯-2-基)钯(II)(464mg,554.7μmol),碳酸铯(3.6g,11.1mmol),氮气氛围下,110℃反应14小时,反应液减压浓缩,残余物用硅胶柱色谱法纯化得到化合物int-19c(1g,产率:37.2%)。LC-MS:ESI[M+H]+=484.2。 Step 2: Compound int-19b (1.3 g, 5.55 mmol) and compound int-3a (2.02 g, 6.1 mmol) were dissolved in 1,4-dioxane (15 mL), and methanesulfonic acid (2-dicyclohexylphosphino-2',6'-diisopropoxy-1,1'-biphenyl) (2-amino-1,1'-biphenyl-2-yl) palladium (II) (464 mg, 554.7 μmol) and cesium carbonate (3.6 g, 11.1 mmol) were added. The mixture was reacted at 110° C. for 14 hours under nitrogen atmosphere. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography to obtain compound int-19c (1 g, yield: 37.2%). LC-MS: ESI [M+H] + = 484.2.

第三步:将化合物int-19c(1g,2.06mmol)溶于四氢呋喃(6mL),加入四丁基氟化铵的四氢呋喃溶液(1M/L,6mL),搅拌反应2小时,反应液减压浓缩后得到粗品标题化合物int-19d(722mg,产率:99%),产品不经纯化直接用于下步反应。LC-MS:ESI[M+H]+=350.2。Step 3: Dissolve compound int-19c (1 g, 2.06 mmol) in tetrahydrofuran (6 mL), add tetrabutylammonium fluoride tetrahydrofuran solution (1 M/L, 6 mL), stir and react for 2 hours, and concentrate the reaction solution under reduced pressure to obtain the crude title compound int-19d (722 mg, yield: 99%), which is used directly in the next step without purification. LC-MS: ESI [M+H] + = 350.2.

第四步:将粗产品化合物int-19d(722mg,2.06mmol)溶于甲胺(8mL,25-30wt%水溶液)在甲醇(7mL)的溶液中,搅拌反应2小时,反应液减压浓缩后得到粗品标题化合物int-19e(700mg,产率:97%),产品不经纯化直接用于下步反应。LC-MS:ESI[M+H]+=349.2。Step 4: Dissolve the crude product compound int-19d (722 mg, 2.06 mmol) in a solution of methylamine (8 mL, 25-30 wt% aqueous solution) in methanol (7 mL), stir and react for 2 hours, and concentrate the reaction solution under reduced pressure to obtain the crude title compound int-19e (700 mg, yield: 97%), which is used directly in the next step without purification. LC-MS: ESI [M+H] + = 349.2.

第五步:将化合物int-19e的粗产物(140mg,0.4mmol)加入5mL MeOH溶解,随后加入0.9mL 4M HCl in dioxane溶液,室温反应12h,TLC监测反应,反应完全后加入碳酸钾,搅拌30分钟后,过滤除去碳酸钾,得到化合物int-19的粗产物(120mg)。MS/ESI[M+H]+=249.1。Step 5: The crude product of compound int-19e (140 mg, 0.4 mmol) was dissolved in 5 mL MeOH, followed by the addition of 0.9 mL 4M HCl in dioxane solution, and the reaction was carried out at room temperature for 12 h. The reaction was monitored by TLC. After the reaction was complete, potassium carbonate was added, stirred for 30 minutes, and potassium carbonate was removed by filtration to obtain the crude product of compound int-19 (120 mg). MS/ESI [M+H] + = 249.1.

中间体int-20:(R)-N-甲基-3,4,4a,5-四氢-2H-吡喃并[2,3-b:5,4-c']二吡啶-8-羧酰胺
Intermediate int-20: (R)-N-methyl-3,4,4a,5-tetrahydro-2H-pyrano[2,3-b:5,4-c']bipyridine-8-carboxamide

第一步:称取化合物int-20a(0.25g,0.83mmol),加入四氢呋喃20ml,置换氮气,降温至-78℃,滴加二异丙基氨基锂(0.46mL,0.91mmol),搅拌30分钟后滴加溶于10mL四氢呋喃的N-苯基双(三氟甲烷磺酰)亚胺(0.36g,1.0mmol),升至室温反应2小时,TLC检测反应完全后,加入饱和氯化铵水溶液淬灭,用乙酸乙酯萃取三次,合并有机相后用无水硫酸钠干燥、过滤、旋干经过柱层析纯化得到化合物int-20b(0.25g,无色液体)。The first step: Weigh the compound int-20a (0.25 g, 0.83 mmol), add 20 ml of tetrahydrofuran, replace nitrogen, cool to -78 ° C, add lithium diisopropylamide (0.46 mL, 0.91 mmol) dropwise, stir for 30 minutes, add N-phenylbis(trifluoromethanesulfonyl)imide (0.36 g, 1.0 mmol) dissolved in 10 mL of tetrahydrofuran dropwise, warm to room temperature and react for 2 hours. After TLC detection, the reaction is complete, saturated ammonium chloride aqueous solution is added to quench, and extracted three times with ethyl acetate. After the organic phases are combined, they are dried over anhydrous sodium sulfate, filtered, and spin-dried. Purification by column chromatography gives compound int-20b (0.25 g, colorless liquid).

第二步:在20ml二氧六环和2ml水的混合溶剂中加入化合物int-20b(0.25g,0.6mmol)、化合物int-20c(0.14g,0.66mmol)、Pd(dppf)Cl2(0.04g,0.06mmol)和碳酸钾(0.16g,1.2mmol),然后置换氮气,在氮气保护下80℃下反应3小时。TLC检测反应完全后,将反应降至室温,旋蒸浓缩,加入30ml乙酸乙酯和20ml水萃取三次,合并有机相后用饱和食盐水洗涤、无水硫酸钠干燥、过滤、旋干。所得粗品经过柱层析纯化,用石油醚打浆固化后过滤,烘干得到化合物int-20d(180mg,淡黄色液体);LC-MS:ESI[M+H]+=434.6。Step 2: Add compound int-20b (0.25 g, 0.6 mmol), compound int-20c (0.14 g, 0.66 mmol), Pd(dppf)Cl 2 (0.04 g, 0.06 mmol) and potassium carbonate (0.16 g, 1.2 mmol) to a mixed solvent of 20 ml dioxane and 2 ml water, then replace nitrogen, and react at 80°C for 3 hours under nitrogen protection. After TLC detection, the reaction is cooled to room temperature, concentrated by rotary evaporation, and extracted three times by adding 30 ml ethyl acetate and 20 ml water. After combining the organic phases, wash with saturated brine, dry with anhydrous sodium sulfate, filter, and spin dry. The obtained crude product is purified by column chromatography, solidified by pulping with petroleum ether, filtered, and dried to obtain compound int-20d (180 mg, light yellow liquid); LC-MS: ESI[M+H] + =434.6.

第三步:在25ml反应瓶中加入int-20d(180mg,0.4mmol)、甲胺水溶液(0.12mL,1.6mmol)和无水甲醇(15ml),室温下搅拌反应3小时。LC-MS监测反应完全后,将 反应液减压浓缩,用石油醚打浆,过滤烘干得到化合物int-20e(160mg,淡黄色固体);LC-MS:ESI[M+H]+=439.6。Step 3: Add int-20d (180 mg, 0.4 mmol), methylamine aqueous solution (0.12 mL, 1.6 mmol) and anhydrous methanol (15 mL) to a 25 mL reaction bottle and stir at room temperature for 3 hours. After the reaction is complete, the LC-MS monitoring The reaction solution was concentrated under reduced pressure, slurried with petroleum ether, filtered and dried to obtain compound int-20e (160 mg, light yellow solid); LC-MS: ESI [M+H] + = 439.6.

第四步:将化合物int-20e(160mg,0.38mmol)加入到10ml无水甲醇中,随后再加入0.4ml 4mol/L盐酸二氧六环溶液,室温搅拌2小时,TLC监测反应完全后,加入二氧六环析出固体,过滤,烘干后得到化合物int-20(80mg,黄色固体);LC-MS:ESI[M+H]+=246.3。Step 4: Compound int-20e (160 mg, 0.38 mmol) was added to 10 ml of anhydrous methanol, followed by 0.4 ml of 4 mol/L hydrochloric acid dioxane solution, and stirred at room temperature for 2 hours. After the reaction was completed as monitored by TLC, dioxane was added to precipitate a solid, which was filtered and dried to obtain compound int-20 (80 mg, yellow solid); LC-MS: ESI [M+H] + = 246.3.

中间体int-21:(6-甲氧基-7-((甲基-d3)硫代)-1,5-萘啶-3-基)甲基甲磺酸盐
Intermediate int-21: (6-methoxy-7-((methyl-d 3 )thio)-1,5-naphthyridin-3-yl)methyl methanesulfonate

(6-甲氧基-7-((甲基-d3)硫代)-1,5-萘啶-3-基)甲基甲磺酸盐的制备参考中间体int-18。LC-MS:ESI[M+H]+=318.3。Preparation of (6-methoxy-7-((methyl-d 3 )thio)-1,5-naphthyridin-3-yl)methyl methanesulfonate Reference intermediate int-18. LC-MS: ESI [M+H] + =318.3.

中间体int-22:(7-((二氟甲基)硫代)-6-甲氧基-1,5-萘啶-3-基)甲基甲磺酸盐
Intermediate int-22: (7-((difluoromethyl)thio)-6-methoxy-1,5-naphthyridin-3-yl)methyl methanesulfonate

第一步:在100mL双颈烧瓶中加入氯二氟乙酸钠int-22a(2.88g,18.9mmol,2eq)和K2CO3(1.95g,14.1mmol,1.5eq),并在高真空下干燥1小时。在室温下加入DMF(99%,30mL),分批加入int-17b(2.5g,9.43mmol)。将反应混合物在95℃下搅拌15分钟(放热反应)并冷却至室温。加入水(130毫升)和乙酸乙酯(130毫升)。分离各层并用水(4x 130mL)洗涤有机层。用MgSO4干燥并在真空下浓缩,柱层析纯化(PE/EA=1/5)得到黄色油状化合物int-22b(1.2g)。LC-MS:ESI[M+H]+=315.1。Step 1: Sodium chlorodifluoroacetate int-22a (2.88 g, 18.9 mmol, 2 eq) and K 2 CO 3 (1.95 g, 14.1 mmol, 1.5 eq) were added to a 100 mL two-necked flask and dried under high vacuum for 1 hour. DMF (99%, 30 mL) was added at room temperature, and int-17b (2.5 g, 9.43 mmol) was added in batches. The reaction mixture was stirred at 95° C. for 15 minutes (exothermic reaction) and cooled to room temperature. Water (130 mL) and ethyl acetate (130 mL) were added. The layers were separated and the organic layer was washed with water (4 x 130 mL). Drying with MgSO 4 and concentrating under vacuum, column chromatography purification (PE/EA=1/5) gave yellow oily compound int-22b (1.2 g). LC-MS: ESI[M+H] + =315.1.

第二步:将化合物int-22b(0.58g,1.84mmol)溶于DCM(20mL)中,0℃下缓慢滴加DIBAL-H(1.0M,5.0mL,5.0mmol),在0℃下搅拌1h。将反应液缓慢倒入50mL饱和氯化铵溶液中淬灭,硅藻土抽滤后EA萃取4*30mL,硫酸钠干燥,得到黄色油状化合物int-22c(0.42g,crude)。LC-MS:ESI[M+H]+=273.1。Step 2: Dissolve compound int-22b (0.58 g, 1.84 mmol) in DCM (20 mL), slowly add DIBAL-H (1.0 M, 5.0 mL, 5.0 mmol) at 0°C, and stir at 0°C for 1 h. Slowly pour the reaction solution into 50 mL saturated ammonium chloride solution to quench, filter with diatomaceous earth, extract with 4*30 mL EA, and dry with sodium sulfate to obtain yellow oily compound int-22c (0.42 g, crude). LC-MS: ESI [M+H] + = 273.1.

第三步:将化合物int-12c(0.42g,1.55mmol)溶于DCM(30mL)中,0℃下滴加TEA(0.7mL,7.36mmol),MsCl(0.5mL,3.6mmol),0℃搅拌1h。在反应液缓慢倒入30ml饱和碳酸氢钠溶液淬灭,DCM萃取3*30ml,硫酸钠干燥,抽滤减压浓缩得到化合物int-22(0.5g,crude)。LC-MS:ESI[M+H]+=351.2。Step 3: Dissolve compound int-12c (0.42 g, 1.55 mmol) in DCM (30 mL), add TEA (0.7 mL, 7.36 mmol) and MsCl (0.5 mL, 3.6 mmol) dropwise at 0°C, and stir at 0°C for 1 h. Slowly pour 30 ml of saturated sodium bicarbonate solution into the reaction solution to quench, extract with 3*30 ml of DCM, dry with sodium sulfate, filter and concentrate under reduced pressure to obtain compound int-22 (0.5 g, crude). LC-MS: ESI [M+H] + = 351.2.

中间体int-23:6-氟-N-甲基-5-(哌啶-4-基)吡啶甲酰胺
Intermediate int-23: 6-Fluoro-N-methyl-5-(piperidin-4-yl)picolinamide

将化合物int-3(0.2g,0.84mmol)溶于DCM(20mL)中,加入Pd/C(10%,0.1g),氢气置换三次后,氢气氛围下在25℃搅12h。反应液垫硅藻土过滤,滤液减压浓缩除去溶剂得化合物int-23(0.2g,crude)。LC-MS:ESI[M+H]+=238.2。Compound int-3 (0.2 g, 0.84 mmol) was dissolved in DCM (20 mL), Pd/C (10%, 0.1 g) was added, and the mixture was replaced with hydrogen three times, and stirred at 25°C for 12 h under a hydrogen atmosphere. The reaction solution was filtered through diatomaceous earth, and the filtrate was concentrated under reduced pressure to remove the solvent to obtain compound int-23 (0.2 g, crude). LC-MS: ESI [M+H] + = 238.2.

中间体int-24:6-氰基-N-甲基-5-(哌嗪-1-基)吡啶甲酰胺
Intermediate int-24: 6-cyano-N-methyl-5-(piperazin-1-yl)picolinamide

第一步:将化合物int-24a(0.85g,4.564mmol),化合物int-9a(1.0g,4.149mmol)溶于dioxane(40mL),加入RuPphos-Pd-G3(0.35g,0.415mmol),Cs2CO3(5.41g,16.595mmol),N2置换3次,于100℃下搅拌15h。将反应液倒入饱和碳酸氢钠(30mL)中萃取,EA萃取(3*30mL),硫酸钠干燥,减压浓缩,柱层析(PE/EA=3/1)纯化,得到化合物int-24b(0.7g,2.021mmol),白色固体。LC-MS:ESI[M+H]+=347.2。Step 1: Dissolve compound int-24a (0.85 g, 4.564 mmol) and compound int-9a (1.0 g, 4.149 mmol) in dioxane (40 mL), add RuPphos-Pd-G 3 (0.35 g, 0.415 mmol), Cs 2 CO 3 (5.41 g, 16.595 mmol), replace with N 2 three times, and stir at 100°C for 15 h. Pour the reaction solution into saturated sodium bicarbonate (30 mL), extract with EA (3*30 mL), dry with sodium sulfate, concentrate under reduced pressure, and purify by column chromatography (PE/EA=3/1) to obtain compound int-24b (0.7 g, 2.021 mmol) as a white solid. LC-MS: ESI[M+H]+=347.2.

第二步:将化合物int-24b(0.7g,2.021mmol)溶于MeOH(10mL)中,加入甲胺甲醇溶液(10mL)。于25℃下搅拌1h。将反应液减压浓缩,得到化合物int-24c(0.7g,2.03mmol,crude),LC-MS:ESI[M+H]+=346.2。Step 2: Dissolve compound int-24b (0.7 g, 2.021 mmol) in MeOH (10 mL), add methylamine methanol solution (10 mL), stir at 25°C for 1 h. Concentrate the reaction solution under reduced pressure to obtain compound int-24c (0.7 g, 2.03 mmol, crude), LC-MS: ESI [M+H] + = 346.2.

第三步:将化合物int-24c(0.7g,2.027mmol)溶于HCl/dioxane(10mL,4.0M)中,25℃下搅拌1h。将反应液减压浓缩,得到化合物int-24(0.6g,2.130mmol,crude),LC-MS:ESI[M+H]+=246.2。Step 3: Dissolve compound int-24c (0.7 g, 2.027 mmol) in HCl/dioxane (10 mL, 4.0 M) and stir at 25°C for 1 h. Concentrate the reaction solution under reduced pressure to obtain compound int-24 (0.6 g, 2.130 mmol, crude), LC-MS: ESI [M+H] + = 246.2.

中间体int-25:6-氟-N-(1-甲基-1H-吡唑-4-基)-5-(哌啶-4-基)吡啶甲酰胺
Intermediate int-25: 6-fluoro-N-(1-methyl-1H-pyrazol-4-yl)-5-(piperidin-4-yl)picolinamide

第一步:将化合物int-25a(1g,3.102mmol)溶于DCM(30mL),加入化合物int-25b(0.36g,3.723mmol),HATU(1.42g,3.723mmol),DIPEA(1.20g,9.307mmol),于25℃下搅拌1h。将反应液倒入饱和碳酸氢钠(30mL)中萃取,DCM萃取(3*30mL),无水硫酸钠干燥,减压浓缩,柱层析(PE/EA=1/1)纯化得到化合物int-25c(1.2g,2.989mmol,96.36%),淡黄色油状液体,LC-MS:ESI[M+H]+=402.2。Step 1: Dissolve compound int-25a (1 g, 3.102 mmol) in DCM (30 mL), add compound int-25b (0.36 g, 3.723 mmol), HATU (1.42 g, 3.723 mmol), DIPEA (1.20 g, 9.307 mmol), and stir at 25°C for 1 h. Pour the reaction solution into saturated sodium bicarbonate (30 mL), extract with DCM (3*30 mL), dry with anhydrous sodium sulfate, concentrate under reduced pressure, and purify by column chromatography (PE/EA=1/1) to obtain compound int-25c (1.2 g, 2.989 mmol, 96.36%), light yellow oily liquid, LC-MS: ESI[M+H] + =402.2.

第二步:将化合物int-25c(1.2g,2.989mmol)溶于MeOH(20mL)中,加入Pd/C 10%(1.2g,11.276mmol),H2置换3次,25℃下搅拌1h。硅胶减压抽滤,减压干燥,得到化合物int-25d(1.1g,2.726mmol,91.21%),淡黄色固体。LC-MS:ESI[M+H]+=404.2。Step 2: Dissolve compound int-25c (1.2 g, 2.989 mmol) in MeOH (20 mL) and add Pd/C 10% (1.2 g, 11.276 mmol), H 2 replaced 3 times, stirred at 25°C for 1 h. Silica gel was filtered off under reduced pressure, and dried under reduced pressure to obtain compound int-25d (1.1 g, 2.726 mmol, 91.21%) as a light yellow solid. LC-MS: ESI [M+H] + = 404.2.

第三步:将化合物int-25d(1.1g,2.726mmol)溶于HCl/MeOH(10mL,4.0M)中,于25℃下搅拌0.5h减压浓缩,得到化合物int-25(1g,2.943mmol,98.95%),白色固体。LC-MS:ESI[M+H]+=304.2。Step 3: Compound int-25d (1.1 g, 2.726 mmol) was dissolved in HCl/MeOH (10 mL, 4.0 M), stirred at 25°C for 0.5 h and concentrated under reduced pressure to obtain compound int-25 (1 g, 2.943 mmol, 98.95%) as a white solid. LC-MS: ESI [M+H] + = 304.2.

中间体int-26:6-氟-N-(1-甲基-1H-吡唑-4-基)-5-(哌嗪-1-基)吡啶甲酰胺
Intermediate int-26: 6-fluoro-N-(1-methyl-1H-pyrazol-4-yl)-5-(piperazin-1-yl)picolinamide

第一步:将化合物int-26a(1g,2.9mmol)溶于THF(10mL),H2O(10mL),加入LiOH(0.62g,14.7mmol),于0℃下搅拌2h。将反应液倒入饱和氯化铵水溶液(30mL)中。DCM萃取(3*30mL),无水硫酸钠干燥,减压浓缩,得到化合物int-26b(1g,3.1mmol,crude),白色固体。LC-MS:ESI[M+H]+=326.2。Step 1: Dissolve compound int-26a (1 g, 2.9 mmol) in THF (10 mL), H 2 O (10 mL), add LiOH (0.62 g, 14.7 mmol), and stir at 0°C for 2 h. Pour the reaction solution into saturated aqueous ammonium chloride solution (30 mL). Extract with DCM (3*30 mL), dry with anhydrous sodium sulfate, and concentrate under reduced pressure to obtain compound int-26b (1 g, 3.1 mmol, crude) as a white solid. LC-MS: ESI [M+H] + = 326.2.

第二步:将化合物int-26b(1g,3.1mmol)溶于DCM(30mL),加入化合物int-25b(0.36g,3.723mmol),HATU(1.42g,3.7mmol),DIPEA(1.20g,9.3mmol),于25℃下搅拌1h。将反应液倒入饱和碳酸氢钠(30mL)中萃取,DCM萃取(3*30mL),无水硫酸钠干燥,减压浓缩,柱层析(PE/EA=1/1)纯化得到化合物化合物int-26c(1.1g,2.7mmol,88.48%),淡黄色油状液体,LC-MS:ESI[M+H]+=405.2。Step 2: Compound int-26b (1 g, 3.1 mmol) was dissolved in DCM (30 mL), and compound int-25b (0.36 g, 3.723 mmol), HATU (1.42 g, 3.7 mmol), and DIPEA (1.20 g, 9.3 mmol) were added, and stirred at 25°C for 1 h. The reaction solution was poured into saturated sodium bicarbonate (30 mL) for extraction, extracted with DCM (3*30 mL), dried over anhydrous sodium sulfate, concentrated under reduced pressure, and purified by column chromatography (PE/EA=1/1) to obtain compound int-26c (1.1 g, 2.7 mmol, 88.48%), a light yellow oily liquid, LC-MS: ESI[M+H] + =405.2.

第三步:将化合物int-26c(1.1g,2.7mmol)溶于HCl/MeOH(10mL,4.0M)中,于25℃下搅拌0.5h。减压浓缩,得到化合物int-26(0.8g,2.6mmol,96.65%),白色固体。LC-MS:ESI[M+H]+=305.2。Step 3: Dissolve compound int-26c (1.1 g, 2.7 mmol) in HCl/MeOH (10 mL, 4.0 M) and stir at 25°C for 0.5 h. Concentrate under reduced pressure to obtain compound int-26 (0.8 g, 2.6 mmol, 96.65%) as a white solid. LC-MS: ESI [M+H] + = 305.2.

中间体int-27:N-环丙基-6-氟-5-(哌啶-4-基)吡啶甲酰胺
Intermediate int-27: N-cyclopropyl-6-fluoro-5-(piperidin-4-yl)picolinamide

第一步:将化合物int-25a(1g,3.102mmol)溶于MeOH(15mL),加入Pd/C 10%(1g,9.397mmol),H2置换3次,25℃下搅拌1h。垫硅胶抽滤,滤液减压干燥,得到化合物int-27a(1g,3.083mmol,99.38%),淡黄色固体。LC-MS:ESI[M+H]+=325.2。Step 1: Dissolve compound int-25a (1 g, 3.102 mmol) in MeOH (15 mL), add Pd/C 10% (1 g, 9.397 mmol), replace with H 2 three times, and stir at 25°C for 1 h. Filter through silica gel pad, and dry the filtrate under reduced pressure to obtain compound int-27a (1 g, 3.083 mmol, 99.38%) as a light yellow solid. LC-MS: ESI [M+H] + = 325.2.

第二步:将化合物int-27a(1g,3.083mmol)溶于DCM(30mL)中,加入环丙胺盐酸盐(0.58g,6.166mmol),DIPEA(1.20g,9.249mmol),HATU(1.76g,4.625mmol),于25℃下搅拌1h。将反应液倒入饱和碳酸氢钠(30mL)中萃取,DCM萃取(3*30mL),无水硫酸钠干燥,减压浓缩,柱层析(PE/EA=1/1)纯化得到化合物int-27b(1g,2.752 mmol,89.25%),淡黄色固体,LC-MS:ESI[M+H]+=364.2。Step 2: Dissolve compound int-27a (1 g, 3.083 mmol) in DCM (30 mL), add cyclopropylamine hydrochloride (0.58 g, 6.166 mmol), DIPEA (1.20 g, 9.249 mmol), HATU (1.76 g, 4.625 mmol), and stir at 25 ° C for 1 h. Pour the reaction solution into saturated sodium bicarbonate (30 mL), extract with DCM (3*30 mL), dry over anhydrous sodium sulfate, concentrate under reduced pressure, and purify by column chromatography (PE/EA=1/1) to obtain compound int-27b (1 g, 2.752 mmol, 89.25%), light yellow solid, LC-MS: ESI [M+H] + = 364.2.

第三步:将化合物int-27b(1.0g,2.752mmol)溶于盐酸二氧六环溶液(10mL,4.0M)中,25℃下搅拌1h。减压浓缩,得到化合物int-27(0.7g,2.658mmol,96.61%),白色固体。LC-MS:ESI[M+H]+=264.3。Step 3: Dissolve compound int-27b (1.0 g, 2.752 mmol) in dioxane hydrochloride solution (10 mL, 4.0 M) and stir at 25°C for 1 h. Concentrate under reduced pressure to obtain compound int-27 (0.7 g, 2.658 mmol, 96.61%) as a white solid. LC-MS: ESI [M+H] + = 264.3.

中间体int-28:N,6-二甲基-5-(哌啶-4-基)吡啶甲酰胺
Intermediate int-28: N,6-dimethyl-5-(piperidin-4-yl)picolinamide

N,6-二甲基-5-(哌啶-4-基)吡啶甲酰胺的制备方法参考中间体int-23。LC-MS:ESI[M+H]+=234.2。The preparation method of N,6-dimethyl-5-(piperidin-4-yl)picolinamide refers to the intermediate int-23. LC-MS: ESI [M+H] + = 234.2.

中间体int-29:2-氯-N-甲基-1',2',3',6'-四氢-[3,4'-联吡啶]-6-甲酰胺
Intermediate int-29: 2-Chloro-N-methyl-1',2',3',6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide

2-氯-N-甲基-1',2',3',6'-四氢-[3,4'-联吡啶]-6-甲酰胺的制备方法参考中间体int-2。LC-MS:ESI[M+H]+=252.7。The preparation method of 2-chloro-N-methyl-1',2',3',6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide refers to the intermediate int-2. LC-MS: ESI [M+H] + = 252.7.

中间体int-30:6-氯-N-甲基-5-(哌啶-4-基)吡啶甲酰胺
Intermediate int-30: 6-chloro-N-methyl-5-(piperidin-4-yl)picolinamide

6-氯-N-甲基-5-(哌啶-4-基)吡啶甲酰胺的制备方法参考中间体int-28。LC-MS:ESI[M+H]+=254.2。The preparation method of 6-chloro-N-methyl-5-(piperidin-4-yl)picolinamide refers to intermediate int-28. LC-MS: ESI [M+H] + = 254.2.

中间体int-31:6-氯-N-甲基-5-(哌嗪-1-基)吡啶甲酰胺
Intermediate int-31: 6-chloro-N-methyl-5-(piperazin-1-yl)picolinamide

6-氯-N-甲基-5-(哌嗪-1-基)吡啶甲酰胺的制备方法参考中间体int-9。LC-MS:ESI[M+H]+=255.2。The preparation method of 6-chloro-N-methyl-5-(piperazin-1-yl)picolinamide refers to the intermediate int-9. LC-MS: ESI [M+H] + = 255.2.

中间体int-32:1-(5-甲氧基-3-甲基吡啶-2-基)哌嗪
Intermediate int-32: 1-(5-methoxy-3-methylpyridin-2-yl)piperazine

第一步:将化合物int-31a(2g,9.899mmol),化合物int-9a(2.21g,11.878mmol)溶于1,4-dioxane(50mL),加入Ruphos-Pd-G3(0.83g,0.990mmol),Cs2CO3(9.68g,29.696mmol),N2置换3次,于100℃下搅拌12h。将反应液倒入饱和碳酸氢钠(30mL)中萃取,EA萃取(3*30mL),无水硫酸钠干燥,减压浓缩,柱层析(PE/EA=3/1)纯化得到化合物int-31b(1.7g,5.530mmol,55.87%),淡黄色液体。LC-MS:ESI[M+H]+=308.2。Step 1: Dissolve compound int-31a (2 g, 9.899 mmol) and compound int-9a (2.21 g, 11.878 mmol) in 1,4-dioxane (50 mL), add Ruphos-Pd-G3 (0.83 g, 0.990 mmol), Cs 2 CO 3 (9.68 g, 29.696 mmol), replace with N 2 three times, and stir at 100°C for 12 h. Pour the reaction solution into saturated sodium bicarbonate (30 mL), extract with EA (3*30 mL), dry with anhydrous sodium sulfate, concentrate under reduced pressure, and purify by column chromatography (PE/EA=3/1) to obtain compound int-31b (1.7 g, 5.530 mmol, 55.87%), a light yellow liquid. LC-MS: ESI[M+H] + =308.2.

第二步:将化合物int-31b(1.7g,5.530mmol)溶于盐酸甲醇溶液(20mL)中,25℃下搅拌1h。减压浓缩,得到化合物int-31(1.1g,5.307mmol,95.96%),白色固体。LC-MS:ESI[M+H]+=208.2。Step 2: Dissolve compound int-31b (1.7 g, 5.530 mmol) in methanolic hydrochloric acid solution (20 mL) and stir at 25°C for 1 h. Concentrate under reduced pressure to obtain compound int-31 (1.1 g, 5.307 mmol, 95.96%) as a white solid. LC-MS: ESI [M+H] + = 208.2.

中间体int-32:(R)-N-甲基-1,2,3,4,4a,5-六氢-7H-吡嗪并[2,1-c]吡啶并[3,2-e][1,4]恶唑烷-9-甲酰胺盐酸盐的制备
Intermediate int-32: Preparation of (R)-N-methyl-1,2,3,4,4a,5-hexahydro-7H-pyrazino[2,1-c]pyrido[3,2-e][1,4]oxazolidine-9-carboxamide hydrochloride

第一步:将(R)-3-(羟甲基)哌嗪-1-甲酸叔丁酯(5g,23.118mmol)加入DMF(50mL)中,加入对甲氧基苄氯(3.62g,23.118mmol)和碳酸钾(9.58g,69.355mmol)升温60℃反应24h,反应结束,加入水淬灭反应,加入乙酸乙酯萃取,有机相水洗3次,有机相减压浓缩,所得粗品经过柱层析纯化(石油醚:乙酸乙酯=10:1~5:1),得化合物int-32b(6g,收率:77.14%)。LC-MS:ESI[M+H]+=337.4.1H NMR(400MHz,Chloroform-d)δ7.14(d,J=8.3Hz,2H),6.79(d,J=8.3Hz,2H),3.88(d,J=13.2Hz,1H),3.81–3.76(m,1H),3.72(s,3H),3.60(m,1H),3.51(m,2H),3.28(d,J=13.1Hz,2H),3.07(s,1H),2.69(d,J=12.3Hz,1H),2.56–2.42(m,1H),2.18(m,1H),1.38(s,9H)。Step 1: Add (R)-3-(Hydroxymethyl)piperazine-1-carboxylic acid tert-butyl ester (5 g, 23.118 mmol) to DMF (50 mL), add p-methoxybenzyl chloride (3.62 g, 23.118 mmol) and potassium carbonate (9.58 g, 69.355 mmol), raise the temperature to 60 ° C and react for 24 hours. After the reaction is completed, water is added to quench the reaction, and ethyl acetate is added for extraction. The organic phase is washed with water 3 times, and the organic phase is concentrated under reduced pressure. The crude product is purified by column chromatography (petroleum ether: ethyl acetate = 10: 1 to 5: 1) to obtain compound int-32b (6 g, yield: 77.14%). LC-MS: ESI[M+H] + =337.4. 1 H NMR(400MHz,Chloroform-d)δ7.14(d,J=8.3Hz,2H),6.79(d,J=8.3Hz,2H),3.88(d,J=13.2Hz,1H),3.81–3.76(m,1H),3.72(s,3H), 3.60(m,1H),3.51(m,2H),3.28(d,J=13.1Hz,2H),3.07(s,1H),2.69(d,J=12.3Hz,1H),2.56–2.42(m,1H),2.18(m,1H),1.38(s,9H).

第二步:将化合物int-32b(6g,17.834mmol)和6-溴-2-(溴甲基)-3-氟吡啶(4.80g,17.834mmol)加入DMF(100mL)中,降温0℃,加入钠氢(1.07g,26.751mmol)加完,室温反应12h,反应结束,加入水淬灭反应,加入乙酸乙酯萃取,有机相水洗3 次,有机相减压浓缩,所得粗品经过柱层析纯化(石油醚:乙酸乙酯=5:1~1:1),得化合物int-32d(4.2g)。LC-MS:ESI[M+H]+=525.4。1H NMR(400MHz,Chloroform-d)δ7.37-7.34(m,1H),7.23-7.19(m,1H),7.16–7.10(m,2H),6.79–6.73(m,2H),4.66–4.51(m,2H),3.84(s,1H),3.72(s,3H),3.59-3.55(m,2H),3.43(d,J=13.1Hz,1H),3.29(d,J=13.3Hz,1H),3.11(s,2H),2.61-2.53(m,2H),2.09(s,1H),1.37(s,9H)。Step 2: Add compound int-32b (6 g, 17.834 mmol) and 6-bromo-2-(bromomethyl)-3-fluoropyridine (4.80 g, 17.834 mmol) to DMF (100 mL), cool to 0°C, add sodium hydrogen sulfide (1.07 g, 26.751 mmol), react at room temperature for 12 h, and when the reaction is complete, add water to quench the reaction, add ethyl acetate for extraction, and wash the organic phase with water for 3 hours. The organic phase was concentrated under reduced pressure, and the obtained crude product was purified by column chromatography (petroleum ether:ethyl acetate=5:1-1:1) to obtain compound int-32d (4.2 g). LC-MS: ESI[M+H] + =525.4. 1H NMR(400MHz,Chloroform-d)δ7.37-7.34(m,1H),7.23-7.19(m,1H),7.16–7.10(m,2H),6.79–6.73(m,2H),4.66–4.51(m,2H),3.84(s,1H), 3.72(s,3H),3.59-3.55(m,2H),3.43(d,J=13.1Hz,1H),3.29(d,J=13.3Hz,1H),3.11(s,2H),2.61-2.53(m,2H),2.09(s,1H),1.37(s,9H).

第三步:将化合物int-32d(4.2g,8.009mmol),PdCl2(PPh3)2(1.12g,1.602mmol)和DIEA(3.11g,24.026mmol)加入DMF(30mL)和乙醇(30mL)中,一氧化碳保护下,升温90℃反应12h,将反应液加入水中,加入乙酸乙酯萃取,有机相水洗3次,有机相减压浓缩,所得粗品经过柱层析纯化(石油醚:乙酸乙酯=5:1~2:1),得化合物int-32e(2g,收率:48.25%)。LC-MS:ESI[M+H]+=518.2。Step 3: Compound int-32d (4.2 g, 8.009 mmol), PdCl 2 (PPh 3 ) 2 (1.12 g, 1.602 mmol) and DIEA (3.11 g, 24.026 mmol) were added to DMF (30 mL) and ethanol (30 mL), and the temperature was raised to 90°C for reaction for 12 h under the protection of carbon monoxide. The reaction solution was added to water, and ethyl acetate was added for extraction. The organic phase was washed with water for 3 times, and the organic phase was concentrated under reduced pressure. The obtained crude product was purified by column chromatography (petroleum ether: ethyl acetate = 5:1 to 2:1) to obtain compound int-32e (2 g, yield: 48.25%). LC-MS: ESI [M+H] + = 518.2.

第四步:将化合物int-32e(500mg,0.966mmol)加入乙腈(10mL)和水(10mL)中,降温0℃,加入硝酸铈铵(2.647g,4.83mmol),室温搅拌12小时,反应结束后,将反应液加入饱和碳酸氢钠水溶液中调pH=8~9,加入乙酸乙酯萃取,有机相水洗3次,有机相减压浓缩,所得粗品经过柱层析纯化(二氯甲烷:甲醇=8:1),得粗品化合物int-32f(100mg,收率:26.05%)。LC-MS:ESI[M+H]+=398.2。Step 4: Add compound int-32e (500 mg, 0.966 mmol) to acetonitrile (10 mL) and water (10 mL), cool to 0°C, add cerium ammonium nitrate (2.647 g, 4.83 mmol), stir at room temperature for 12 hours, after the reaction is completed, add the reaction solution to saturated sodium bicarbonate aqueous solution to adjust pH to 8-9, add ethyl acetate for extraction, wash the organic phase with water 3 times, concentrate the organic phase under reduced pressure, and purify the obtained crude product by column chromatography (dichloromethane: methanol = 8: 1) to obtain crude compound int-32f (100 mg, yield: 26.05%). LC-MS: ESI [M+H] + = 398.2.

第五步:将化合物int-32f(85mg,0.214mmol)和DIEA(82.93mg,0.642mmol)加入NMP(3mL)中,升温180℃搅拌6小时,反应结束后,将反应液加入水中,乙酸乙酯萃取,有机相水洗3次,有机相减压浓缩,得化合物int-32g(80mg,收率:99.11%)。LC-MS:ESI[M+H]+=378.2。Step 5: Add compound int-32f (85 mg, 0.214 mmol) and DIEA (82.93 mg, 0.642 mmol) to NMP (3 mL), heat to 180°C and stir for 6 hours. After the reaction is completed, add the reaction solution to water, extract with ethyl acetate, wash the organic phase with water three times, and concentrate the organic phase under reduced pressure to obtain compound int-32g (80 mg, yield: 99.11%). LC-MS: ESI [M+H] + = 378.2.

第六步:将化合物int-32g(80mg,0.212mmol)和甲胺醇溶液(2mL)加入甲醇(2mL)中,室温搅拌1h,反应结束后,将反应液减压浓缩,得粗品化合物int-32h(80mg,收率:100%)。LC-MS:ESI[M+H]+=363.4。Step 6: Add compound int-32g (80 mg, 0.212 mmol) and methylamine alcohol solution (2 mL) into methanol (2 mL), stir at room temperature for 1 h, and after the reaction, concentrate the reaction solution under reduced pressure to obtain crude compound int-32h (80 mg, yield: 100%). LC-MS: ESI [M+H] + = 363.4.

第七步:将化合物int-32h(80mg,0.221mmol)加入二氧六环(2mL)中,加入盐酸二氧六环(1mL),室温搅拌1小时,反应结束后,将反应液减压浓缩,得化合物int-32(盐酸盐80mg),LC-MS:ESI[M+H]+=263.2。Step 7: Add compound int-32h (80 mg, 0.221 mmol) to dioxane (2 mL), add dioxane hydrochloride (1 mL), stir at room temperature for 1 hour, after the reaction is completed, concentrate the reaction solution under reduced pressure to obtain compound int-32 (hydrochloride 80 mg), LC-MS: ESI [M+H] + = 263.2.

中间体int-33:5-(氮杂环丁烷-3-基)-N-甲基-5,6-二氢吡咯并[3,4-c]吡唑-2(4H)-甲酰胺
Intermediate int-33: 5-(azetidin-3-yl)-N-methyl-5,6-dihydropyrrolo[3,4-c]pyrazole-2(4H)-carboxamide

第一步:将化合物int-33a(4.0g,19.116mmol)加入二氯甲烷(80mL)中,加入碳酸钠(5.07g,47.790mmol)和氯乙酸4-硝基苯酯(4.24g,21.027mmol),加完,室温反应12h,反应结束后,加入水淬灭反应,加入二氯甲烷萃取,有机相水洗,有机相无水硫酸钠干燥,过滤,收集滤液,滤液浓干,得化合物int-33b(收率:83.79%),LC-MS:ESI[M+H]+=375.2。Step 1: Add compound int-33a (4.0 g, 19.116 mmol) to dichloromethane (80 mL), add sodium carbonate (5.07 g, 47.790 mmol) and 4-nitrophenyl chloroacetate (4.24 g, 21.027 mmol), and react at room temperature for 12 h. After the reaction is completed, add water to quench the reaction, add dichloromethane to extract, wash the organic phase with water, dry the organic phase over anhydrous sodium sulfate, filter, collect the filtrate, and concentrate the filtrate to obtain compound int-33b (yield: 83.79%), LC-MS: ESI [M+H] + = 375.2.

第二步:将化合物int-33b(6.0g,16.028mmol)加入二氯甲烷(100mL)中,加入三乙胺(4.87g,48.083mmol)和甲胺盐酸盐(1.30g,19.233mmol),加完,室温反应12h,反应结束后,加入水淬灭反应,加入二氯甲烷萃取,有机相水洗,有机相无水硫酸钠干燥,过滤,收集滤液,滤液浓干,浓缩物柱层析(PE:EA=5:1~1:1),收集产物,得化合物int-33c(收率:46.86%),LC-MS:ESI[M+H]+=267.2。Step 2: Add compound int-33b (6.0 g, 16.028 mmol) to dichloromethane (100 mL), add triethylamine (4.87 g, 48.083 mmol) and methylamine hydrochloride (1.30 g, 19.233 mmol), react at room temperature for 12 h, add water to quench the reaction, add dichloromethane to extract, wash the organic phase with water, dry the organic phase with anhydrous sodium sulfate, filter, collect the filtrate, concentrate the filtrate, and perform column chromatography (PE:EA=5:1~1:1) to collect the product to obtain compound int-33c (yield: 46.86%), LC-MS: ESI[M+H] + =267.2.

第三步:将化合物int-33c(2.0g,7.51mmol)加入二氯甲烷(20mL)中,加入三氟乙酸(5mL),室温反应1h,反应结束后,减压浓缩,收集产物,得化合物int-33d三氟乙酸盐,LC-MS:ESI[M+H]+=167.20。Step 3: Add compound int-33c (2.0 g, 7.51 mmol) to dichloromethane (20 mL), add trifluoroacetic acid (5 mL), react at room temperature for 1 h, and after the reaction is completed, concentrate under reduced pressure and collect the product to obtain compound int-33d trifluoroacetate, LC-MS: ESI [M+H] + = 167.20.

第四步:将化合物int-33d三氟乙酸盐(2.3g,8.208mmol)加入二氯甲烷(20mL)中,加入三乙胺调pH=6~7,加入3-氧亚基氮杂环丁烷-1-甲酸叔丁酯(2.81g,16.416mmol),加完,室温反应1h,加入三乙酰氧基硼氢化钠(3.48g,16.416mmol),反应结束后,加入水淬灭反应,加入二氯甲烷萃取,有机相水洗,有机相无水硫酸钠干燥,过滤,收集滤液,滤液浓干,浓缩物柱层析(PE;EA=2:1~1:1),收集产物,得化合物int-33e(收率:75.82%),LC-MS:ESI[M+H]+=322.2。Step 4: Add compound int-33d trifluoroacetate (2.3 g, 8.208 mmol) to dichloromethane (20 mL), add triethylamine to adjust the pH to 6-7, add tert-butyl 3-oxyylideneazetidine-1-carboxylate (2.81 g, 16.416 mmol), add, react at room temperature for 1 h, add sodium triacetoxyborohydride (3.48 g, 16.416 mmol), after the reaction is completed, add water to quench the reaction, add dichloromethane to extract, wash the organic phase with water, dry the organic phase with anhydrous sodium sulfate, filter, collect the filtrate, concentrate the filtrate, and column chromatograph the concentrate (PE; EA = 2:1-1:1) to collect the product to obtain compound int-33e (yield: 75.82%), LC-MS: ESI [M+H] + = 322.2.

第五步:将化合物int-33e(2.0g,6.223mmol)加入二氯甲烷(20mL)中,加入三氟乙酸(5mL),室温反应1h,反应结束后,减压浓缩,收集产物,得化合物int-33三氟乙酸盐,LC-MS:ESI[M+H]+=222.1。Step 5: Add compound int-33e (2.0 g, 6.223 mmol) to dichloromethane (20 mL), add trifluoroacetic acid (5 mL), react at room temperature for 1 h, and after the reaction is completed, concentrate under reduced pressure and collect the product to obtain compound int-33 trifluoroacetate. LC-MS: ESI [M+H] + = 222.1.

中间体int-34:5-(氮杂环丁烷-3-基乙炔基)-6-氟-N-甲基吡啶甲酰胺
Intermediate int-34: 5-(azetidin-3-ylethynyl)-6-fluoro-N-methylpicolinamide

第一步:在瓶中加入化合物int-3a(537.47mg,2.306mmol),化合物int-34a(418mg,2.306mmol),CuI(87.85mg,0.461mmol),双三苯基磷二氯化钯(161.88mg,0.231mmol),TEA(0.962mL,6.919mmol),DMF(10mL)溶解,N2置换,升温至80℃搅拌过夜,反应液倒入冰水淬灭,EA萃取,有机相干燥,旋干,残余物EA/PE=0-25%柱层析得产物int-34b(357mg,色固体),LC-MS:[M+H]+=335.2。Step 1: Add compound int-3a (537.47 mg, 2.306 mmol), compound int-34a (418 mg, 2.306 mmol), CuI (87.85 mg, 0.461 mmol), bistriphenylphosphine palladium dichloride (161.88 mg, 0.231 mmol), TEA (0.962 mL, 6.919 mmol), DMF (10 mL) to a bottle for dissolution, N2 replacement, warm to 80 ° C and stir overnight, pour the reaction solution into ice water for quenching, extract with EA, dry the organic phase, spin dry, and the residue is EA/PE = 0-25% column chromatography to obtain the product int-34b (357 mg, color solid), LC-MS: [M+H] + = 335.2.

第二步:在瓶中加入化合物int-34b(314mg,0.939mmol),DCM(10mL)溶清,降温至0-10℃,滴加甲胺甲醇溶液(0.8mL,0.120mmol),保温搅拌1.5h,缓慢恢复至室温,继续搅拌3h,控温在10-20℃减压浓缩,残余物EA/PE=0-26%柱层析得产物int-34c(110mg,油状液体),LC-MS:[M+H]+=334.2。Step 2: Add compound int-34b (314 mg, 0.939 mmol) into a bottle, dissolve in DCM (10 mL), cool to 0-10°C, add methylamine methanol solution (0.8 mL, 0.120 mmol) dropwise, keep stirring for 1.5 h, slowly return to room temperature, continue stirring for 3 h, control the temperature at 10-20°C, concentrate under reduced pressure, and the residue is purified by column chromatography with EA/PE = 0-26% to obtain the product int-34c (110 mg, oily liquid), LC-MS: [M+H]+ = 334.2.

第三步:在瓶中加入化合物int-34c(86mg,0.258mmol),DCM(10mL)溶清,加入三氟乙酸(2mL,26.118mmol),室温搅拌5h,旋干得化合物int-34(130mg)三氟醋酸盐粗品,未进一步纯化,直接用于下一步,LC-MS:[M+H]+=234.1。Step 3: Add compound int-34c (86 mg, 0.258 mmol) into the bottle, dissolve in DCM (10 mL), add trifluoroacetic acid (2 mL, 26.118 mmol), stir at room temperature for 5 h, and spin dry to obtain the crude trifluoroacetate of compound int-34 (130 mg), which was used directly in the next step without further purification. LC-MS: [M+H]+=234.1.

中间体int-35:(R)-N-环丙基-1,2,3,4,4a,5-六氢-7H-吡嗪并[2,1-c]吡啶并[3,2-e][1,4]恶唑烷-9-甲酰胺
Intermediate int-35: (R)-N-cyclopropyl-1,2,3,4,4a,5-hexahydro-7H-pyrazino[2,1-c]pyrido[3,2-e][1,4]oxazolidine-9-carboxamide

第一步:将化合物int-32g(210mg,0.578mmol)加入甲醇(2.00mL),四氢呋喃(2mL),水(2mL)中,加入氢氧化锂一水合物(121.35mg,2.889mmol),加完,室温反应2h。反应结束后,反应液用1N盐酸调pH=3~4,乙酸乙酯萃取,浓缩干,得化合物int-35a(160mg,收率:79.25%),LC-MS:[M+H]+=350.1。Step 1: Add compound int-32g (210 mg, 0.578 mmol) to methanol (2.00 mL), tetrahydrofuran (2 mL), and water (2 mL), add lithium hydroxide monohydrate (121.35 mg, 2.889 mmol), add, and react at room temperature for 2 hours. After the reaction is completed, the reaction solution is adjusted to pH 3-4 with 1N hydrochloric acid, extracted with ethyl acetate, and concentrated to dryness to obtain compound int-35a (160 mg, yield: 79.25%), LC-MS: [M+H] + = 350.1.

第二步:将化合物int-35a(130mg,0.372mmol),环丙胺(34.81mg,0.372mmol),DIEA(144.27mg,1.116mmol)加入DMF(3mL),加入HATU(212.22mg,0.558mmol),室温反应1h。将反应液加入水中,加EA萃取,浓缩除去溶剂得化合物int-35b(120mg,收率:83.02%),LC-MS:[M+H]+=389.2。Step 2: Add compound int-35a (130 mg, 0.372 mmol), cyclopropylamine (34.81 mg, 0.372 mmol), DIEA (144.27 mg, 1.116 mmol) to DMF (3 mL), add HATU (212.22 mg, 0.558 mmol), and react at room temperature for 1 h. Add the reaction solution into water, extract with EA, and concentrate to remove the solvent to obtain compound int-35b (120 mg, yield: 83.02%), LC-MS: [M+H] + = 389.2.

第三步:将化合物int-35b(120mg,0.309mmol)加入二氯甲烷(3mL),加入盐酸二氧六环溶液(3mL),室温反应1h。将反应液减压浓缩干,得粗品化合物int-35盐酸盐(120 mg),LC-MS:[M+H]+=289.2。Step 3: Add compound int-35b (120 mg, 0.309 mmol) to dichloromethane (3 mL), add hydrochloric acid dioxane solution (3 mL), and react at room temperature for 1 h. Concentrate the reaction solution under reduced pressure to dryness to obtain crude compound int-35 hydrochloride (120 mg, 0.309 mmol). mg), LC-MS: [M+H] + =289.2.

中间体int-36:N-甲基-5-((2S,3R)-2-甲基氮杂环丁烷-3-基)-5,6-二氢吡咯并[3,4-c]吡唑-2(4H)-甲酰胺
Intermediate int-36: N-methyl-5-((2S,3R)-2-methylazetidin-3-yl)-5,6-dihydropyrrolo[3,4-c]pyrazole-2(4H)-carboxamide

第一步:在反应瓶中加入化合物int-33d(1.1g,2.790mmol),DCM(25mL)溶解,加入三乙胺(1.801mL,12.958mmol),搅拌10min,加入化合物int-36a(0.4g,2.160mmol),搅拌10min,加入三乙酰氧基硼氢化钠(1.6g,7.559mmol),室温搅拌5h,将反应液倒入冰水中,分出有机相,水相DCM萃取,合并有机相,干燥,旋干,减压浓缩,残余物用EA/PE=0-95%柱层析得油状物化合物int-36b(315mg,白色固体)。Step 1: Add compound int-33d (1.1 g, 2.790 mmol) to the reaction bottle, dissolve in DCM (25 mL), add triethylamine (1.801 mL, 12.958 mmol), stir for 10 min, add compound int-36a (0.4 g, 2.160 mmol), stir for 10 min, add sodium triacetoxyborohydride (1.6 g, 7.559 mmol), stir at room temperature for 5 h, pour the reaction solution into ice water, separate the organic phase, extract the aqueous phase with DCM, combine the organic phases, dry, spin dry, and concentrate under reduced pressure. The residue is purified by EA/PE = 0-95% column chromatography to obtain an oily compound int-36b (315 mg, white solid).

第二步:向反应瓶中加入化合物int-36b(315mg,0.94mmol),DCM(10mL)溶解,降温T=0-10℃,加入TFA(2mL,26.118mmol),室温搅拌3h,LC-MS显示:反应完成,旋干得化合物int-36粗品(766mg,含TFA),LC-MS:[M+H]+=236.2,未进一步纯化,直接用于下一步。Step 2: Add compound int-36b (315 mg, 0.94 mmol) to the reaction bottle, dissolve in DCM (10 mL), cool to T = 0-10°C, add TFA (2 mL, 26.118 mmol), stir at room temperature for 3 h, LC-MS shows: the reaction is complete, and the crude compound int-36 (766 mg, containing TFA) is obtained by spin drying, LC-MS: [M+H] + = 236.2, without further purification, directly used in the next step.

中间体int-37:N-甲基-5-(吡咯烷-3-基乙炔基)吡啶甲酰胺
Intermediate int-37: N-methyl-5-(pyrrolidin-3-ylethynyl)picolinamide

第一步:反应瓶中加入化合物int-37a(565mg,2.894mmol),化合物int-2a(625.10mg,2.894mmol),双三苯基磷二氯化钯(203.10mg,0.289mmol),TEA(1.207mL,8.681mmol),CuI(110.22mg,0.579mmol),DMF(10mL)溶解,N2置换,升温T=80℃搅拌20h,反应液过滤,滤液旋干,残余物用EA/PE=0-40%柱层析得化合物int-37b(529mg)黄色油状物。LC-MS:[M+H]+=331.2。Step 1: Add compound int-37a (565 mg, 2.894 mmol), compound int-2a (625.10 mg, 2.894 mmol), bistriphenylphosphine palladium dichloride (203.10 mg, 0.289 mmol), TEA (1.207 mL, 8.681 mmol), CuI (110.22 mg, 0.579 mmol), dissolve in DMF (10 mL), replace with N2, heat to T = 80 ° C and stir for 20 h, filter the reaction solution, spin dry the filtrate, and use EA/PE = 0-40% column chromatography to obtain compound int-37b (529 mg) as a yellow oil. LC-MS: [M+H] + = 331.2.

第二步:反应瓶中加入化合物int-37b(529mg,1.601mmol),甲醇(15mL)溶解,加入甲胺(15mL),室温搅拌5h,反应液旋干得化合物int-37c粗品(529mg)淡黄色油状物。LC-MS:[M+H]+=330.2,未进一步纯化,直接用于下一步。Step 2: Add compound int-37b (529 mg, 1.601 mmol) to the reaction flask, dissolve in methanol (15 mL), add methylamine (15 mL), stir at room temperature for 5 h, and spin dry the reaction solution to obtain crude compound int-37c (529 mg) as a light yellow oil. LC-MS: [M+H] + = 330.2, without further purification, directly used in the next step.

第三步:化合物int-37c(529mg,1.606mmol),DCM(20mL)溶解,加入TFA(5mL,65.296mmol),室温搅拌2h,反应液旋干得化合物int-37粗品(1.25g)黄色油状物(含TFA),未进一步纯化,直接用于下一步。LC-MS:[M+H]+=230.2。 Step 3: Compound int-37c (529 mg, 1.606 mmol) was dissolved in DCM (20 mL), TFA (5 mL, 65.296 mmol) was added, and the mixture was stirred at room temperature for 2 h. The reaction solution was spin-dried to obtain a crude product of compound int-37 (1.25 g) as a yellow oil (containing TFA), which was used directly in the next step without further purification. LC-MS: [M+H] + = 230.2.

中间体int-38:5-((3-氟氮杂环丁烷-3-基)乙炔基)-N-甲基吡啶甲酰胺
Intermediate int-38: 5-((3-fluoroazetidin-3-yl)ethynyl)-N-methylpicolinamide

第一步:反应瓶中加入化合物int-38a(400mg,1.852mmol),化合物int-2a(438.23mg,2.222mmol),双三苯基磷二氯化钯(64.98mg,0.093mmol),碘化亚铜(29.38mg,0.093mmol),TEA(1.029mL,7.406mmol),DMF(5mL),N2置换,升温T=50℃搅拌过夜,反应液中加入水,EA萃取,有机相干燥,旋干,残余物用EA/PE=0-90%柱层析得化合物int-38b(529mg)棕色油状物,LC-MS:[M+H]+=333.1。Step 1: Add compound int-38a (400 mg, 1.852 mmol), compound int-2a (438.23 mg, 2.222 mmol), bistriphenylphosphine palladium dichloride (64.98 mg, 0.093 mmol), cuprous iodide (29.38 mg, 0.093 mmol), TEA (1.029 mL, 7.406 mmol), DMF (5 mL) into the reaction flask, replace with N2, raise the temperature to T = 50 ° C and stir overnight, add water to the reaction solution, extract with EA, dry the organic phase, spin dry, and chromatograph the residue with EA/PE = 0-90% column to obtain compound int-38b (529 mg) as a brown oil, LC-MS: [M+H] + = 333.1.

第二步:瓶中加入化合物int-38b(339mg,1.020mmol),甲醇(10mL)溶解,加入甲胺(33%,甲醇溶液),室温搅拌过夜。反应液旋干,残余物MeOH/DCM=0-9%柱层析得化合物int-38c(321mg)淡黄色油状物,LC-MS:[M+H]+=332.2。Step 2: Add compound int-38b (339 mg, 1.020 mmol) to the bottle, dissolve in methanol (10 mL), add methylamine (33%, methanol solution), and stir at room temperature overnight. The reaction solution was spin-dried, and the residue was subjected to column chromatography with MeOH/DCM = 0-9% to obtain compound int-38c (321 mg) as a light yellow oil, LC-MS: [M+H] + = 332.2.

第三步:瓶中加入化合物int-38c(321mg,0.969mmol),DCM(20mL)溶解,降温T=-78℃,滴加DAST(0.256mL,1.937mmol),保温搅拌1h。将反应液中加入水,分液,水相DCM萃取,合并有机相,干燥,旋干。残余物EA/PE=0-40%柱层析得化合物int-38d(229mg)无色油状物,LC-MS:[M+H]+=334.2。Step 3: Add compound int-38c (321 mg, 0.969 mmol) to the bottle, dissolve in DCM (20 mL), cool to T = -78 ° C, add DAST (0.256 mL, 1.937 mmol) dropwise, and stir for 1 h. Add water to the reaction solution, separate the liquids, extract the aqueous phase with DCM, combine the organic phases, dry, and spin dry. The residue was subjected to EA/PE = 0-40% column chromatography to obtain compound int-38d (229 mg) as a colorless oil, LC-MS: [M+H] + = 334.2.

第四步:瓶中加入化合物int-38d(118mg,0.354mmol),DCM(10mL)溶解,加入TFA(3mL,39.177mmol),室温搅拌2h,LC-MS显示:反应完成,将反应液减压浓缩得化合物int-38(181mg,含TFA)无色油状物,未进一步纯化,直接用于下一步。LC-MS:[M+H]+=234.2。Step 4: Add compound int-38d (118 mg, 0.354 mmol) to the bottle, dissolve in DCM (10 mL), add TFA (3 mL, 39.177 mmol), stir at room temperature for 2 h, LC-MS shows: the reaction is complete, the reaction solution is concentrated under reduced pressure to obtain compound int-38 (181 mg, containing TFA) as a colorless oil, which is used directly in the next step without further purification. LC-MS: [M+H] + = 234.2.

中间体int-39:4-(1,5-二甲基-1H-咪唑-4-基)-1,2,3,6-四氢吡啶
Intermediate int-39: 4-(1,5-dimethyl-1H-imidazol-4-yl)-1,2,3,6-tetrahydropyridine

第一步:将化合物int-2b(350mg,1.132mmol),化合物int-39a(200mg,1.143mmol)溶于H2O(5mL),dioxane(20mL),加入Pd(dppf)Cl2(82.82mg,0.113mmol),K2CO3(312.86mg,2.26mmol),N2置换3次,于100℃下搅拌12h。减压浓缩,柱层析(EA/PE=1/1,DCM/MeOH=20/1),得到化合物int-39b(300mg,1.08mmol,95.55%)。 LC-MS:ESI[M+H]+=278.2。Step 1: Dissolve compound int-2b (350 mg, 1.132 mmol) and compound int-39a (200 mg, 1.143 mmol) in H 2 O (5 mL) and dioxane (20 mL), add Pd(dppf)Cl 2 (82.82 mg, 0.113 mmol) and K 2 CO 3 (312.86 mg, 2.26 mmol), replace with N 2 three times, stir at 100°C for 12 h. Concentrate under reduced pressure, and perform column chromatography (EA/PE=1/1, DCM/MeOH=20/1) to obtain compound int-39b (300 mg, 1.08 mmol, 95.55%). LC-MS: ESI [M+H] + = 278.2.

第二步:将化合物int-39b(300mg,1.08mmol)溶于盐酸-甲醇标准溶液(4.0M,10mL),25℃下搅拌1h。减压浓缩,得到化合物化合物int-39(180mg,1.016mmol,93.89%)。LC-MS:ESI[M+H]+=178.2。Step 2: Dissolve compound int-39b (300 mg, 1.08 mmol) in a standard hydrochloric acid-methanol solution (4.0 M, 10 mL), and stir at 25°C for 1 h. Concentrate under reduced pressure to obtain compound int-39 (180 mg, 1.016 mmol, 93.89%). LC-MS: ESI [M+H] + = 178.2.

实施例1:1'-((7-(1,1-二氟乙基)-6-氧代-5,6-二氢-1,5-萘啶-3-基)甲基)-N-甲基-1',2',3',6'-四氢-[3,4'-联吡啶]-6-甲酰胺
Example 1: 1'-((7-(1,1-difluoroethyl)-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)-N-methyl-1',2',3',6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide

第一步:将化合物int-1(1.80g,1.61mmol)溶于dioxane(30mL)中,加入XPhos-Pd-G2(0.40g,0.51mmol),三丁基(1-乙氧基乙烯)锡(2.0g,11.6mmol),N2氛围下,80℃加热并搅拌2h。将反应液倒入30mL饱和碳酸氢钠溶液中,乙酸乙酯萃取5*30mL,硫酸钠干燥,抽滤减压浓缩后,加入THF(10mL),HCl(3.0M,20mL),搅拌12h。将反应液倒入30mL饱和碳酸氢钠溶液中,乙酸乙酯萃取3*40mL,硫酸钠干燥过滤。柱层析(EA/PE=1/5),得到黄色油状化合物1a(2.30g)。LC-MS:ESI[M+H]+=275.2。Step 1: Dissolve compound int-1 (1.80 g, 1.61 mmol) in dioxane (30 mL), add XPhos-Pd-G 2 (0.40 g, 0.51 mmol), tributyl (1-ethoxyethylene) tin (2.0 g, 11.6 mmol), heat at 80 ° C and stir for 2 h under N 2 atmosphere. Pour the reaction solution into 30 mL saturated sodium bicarbonate solution, extract 5*30 mL with ethyl acetate, dry with sodium sulfate, filter and concentrate under reduced pressure, add THF (10 mL), HCl (3.0 M, 20 mL), and stir for 12 h. Pour the reaction solution into 30 mL saturated sodium bicarbonate solution, extract 3*40 mL with ethyl acetate, dry with sodium sulfate and filter. Column chromatography (EA/PE=1/5) gave yellow oily compound 1a (2.30 g). LC-MS: ESI[M+H] + =275.2.

第二步:将化合物1a(0.37g,1.34mmol)溶于DCM(10mL)中,0℃下加入DAST(0.4mL,4.02mmol),缓慢升温至25℃,搅拌12h。将反应液缓慢倒入30mL饱和碳酸氢钠中淬灭,DCM萃取3*30mL,硫酸钠干燥,抽滤,减压浓缩后,Pre-TLC(EA/PE=1/3)纯化得到白色固体化合物1b(0.09g,0.30mmol,收率为22.7%)。LC-MS:ESI[M+H]+=297.2。Step 2: Compound 1a (0.37 g, 1.34 mmol) was dissolved in DCM (10 mL), DAST (0.4 mL, 4.02 mmol) was added at 0°C, the temperature was slowly raised to 25°C, and stirred for 12 h. The reaction solution was slowly poured into 30 mL of saturated sodium bicarbonate to quench, extracted with DCM for 3*30 mL, dried with sodium sulfate, filtered, concentrated under reduced pressure, and purified by Pre-TLC (EA/PE=1/3) to obtain white solid compound 1b (0.09 g, 0.30 mmol, yield 22.7%). LC-MS: ESI[M+H] + =297.2.

第三步:将化合物1b(0.09g,0.30mmol)溶于dioxane(10mL)中,缓慢滴加HBr(46%in H2O,10mL),在70℃下搅拌1h。将反应液冷却后缓慢倒入100mL饱和碳酸氢钠溶液中淬灭,EA萃取4*40mL,硫酸钠干燥,Pre-TLC(DCM/EA=1/1)得到白色固体化合物1c(0.09g)。LC-MS:ESI[M+H]+=283.1。Step 3: Dissolve compound 1b (0.09 g, 0.30 mmol) in dioxane (10 mL), slowly add HBr (46% in H 2 O, 10 mL), and stir at 70°C for 1 h. After cooling, slowly pour the reaction solution into 100 mL saturated sodium bicarbonate solution to quench, extract with EA 4*40 mL, dry with sodium sulfate, and obtain white solid compound 1c (0.09 g) by Pre-TLC (DCM/EA=1/1). LC-MS: ESI[M+H] + =283.1.

第四步:将化合物1c(0.09g,0.31mmol)溶于DCM(10mL)中,0℃,N2氛围下,缓慢滴加DIBAL-H(1M,3.0mL,3.0mmol),在0℃下搅拌1h。在反应液缓慢倒入30mL 饱和酒石酸钾钠溶液淬灭,DCM萃取4*40mL,硫酸钠干燥,Pre-TLC(DCM/MeOH=15/1)得到白色固体化合物1d(0.026g,0.11mmol,收率为34.8%)。LC-MS:ESI[M+H]+=241.2。Step 4: Dissolve compound 1c (0.09 g, 0.31 mmol) in DCM (10 mL), slowly add DIBAL-H (1 M, 3.0 mL, 3.0 mmol) dropwise at 0°C under N2 atmosphere, and stir at 0°C for 1 h. Slowly pour 30 mL of The mixture was quenched with saturated sodium potassium tartrate solution, extracted with 4*40 mL of DCM, dried with sodium sulfate, and Pre-TLC (DCM/MeOH=15/1) gave white solid compound 1d (0.026 g, 0.11 mmol, yield 34.8%). LC-MS: ESI [M+H] + = 241.2.

第五步:将化合物1d(0.026g,0.11mmol)溶于DCM(20mL)中,0℃下滴加DIPEA(0.1mL,0.56mmol),MsCl(0.02mL,0.2mmol)升至25℃搅拌1h。在反应液缓慢倒入20ml饱和碳酸氢钠溶液淬灭,DCM萃取3*30ml,硫酸钠干燥,抽滤减压浓缩得到化合物1e(0.05g)。LC-MS:ESI[M+H]+=397.2。Step 5: Compound 1d (0.026 g, 0.11 mmol) was dissolved in DCM (20 mL), and DIPEA (0.1 mL, 0.56 mmol) and MsCl (0.02 mL, 0.2 mmol) were added dropwise at 0°C, and the mixture was heated to 25°C and stirred for 1 h. 20 ml of saturated sodium bicarbonate solution was slowly poured into the reaction solution for quenching, and 3*30 ml of DCM was extracted, dried over sodium sulfate, and filtered and concentrated under reduced pressure to obtain compound 1e (0.05 g). LC-MS: ESI [M+H] + = 397.2.

第六步:将化合物1e(0.05g,0.12mmol)溶于ACN(10mL)中,加入DIPEA(0.5mL,2.80mmol),KI(0.02g),化合物int-6(0.02g,0.09mmol)在80℃搅拌2h。在反应液缓慢倒入20ml饱和氯化铵溶液淬灭,EA萃取4*30ml,硫酸钠干燥,抽滤减压浓缩得到化合物1f(0.05g)。LC-MS:ESI[M+H]+=522.2。Step 6: Compound 1e (0.05 g, 0.12 mmol) was dissolved in ACN (10 mL), and DIPEA (0.5 mL, 2.80 mmol), KI (0.02 g), and compound int-6 (0.02 g, 0.09 mmol) were added and stirred at 80°C for 2 h. 20 ml of saturated ammonium chloride solution was slowly poured into the reaction solution for quenching, EA was extracted with 4*30 ml, dried over sodium sulfate, filtered and concentrated under reduced pressure to obtain compound 1f (0.05 g). LC-MS: ESI [M+H] + = 522.2.

第七步:将化合物1f(0.05g,0.09mmol)溶于MeOH(5mL)中,加入KOH(5mL,3.0M在25℃搅拌1h。在反应液缓慢倒入30ml饱和氯化铵溶液淬灭,EA萃取4*30ml,硫酸钠干燥,抽滤减压浓缩,反相制备纯化白色固体化合物1(6.0mg,0.013mmol,收率为13.5%)。LC-MS:ESI[M+H]+=443.2。1H NMR(400MHz,MeOD)δ8.68(s,1H),8.59(s,1H),8.22(s,1H),8.03(d,J=8.4Hz,1H),7.95(dd,J=8.4,2.0Hz,1H),7.80(s,1H),6.36(s,1H),3.85(s,2H),3.01(s,1H),2.84(t,J=5.6Hz,2H),2.65(s,2H),2.19(t,J=7.6Hz,1H),2.07(t,J=14.4Hz,3H)。Step 7: Compound 1f (0.05 g, 0.09 mmol) was dissolved in MeOH (5 mL), KOH (5 mL, 3.0 M) was added and stirred at 25°C for 1 h. 30 ml of saturated ammonium chloride solution was slowly poured into the reaction solution to quench, 4*30 ml of EA was extracted, dried over sodium sulfate, filtered and concentrated under reduced pressure, and the white solid compound 1 (6.0 mg, 0.013 mmol, yield was 13.5%) was prepared and purified by reverse phase. LC-MS: ESI [M+H] + = 443.2. 1 H NMR (400MHz, MeOD) δ8.68(s,1H),8.59(s,1H),8.22(s,1H),8.03(d,J=8.4Hz,1H),7.95(dd,J=8.4,2.0Hz,1H),7.80(s,1H) ,6.36(s,1H),3.85(s,2H),3.01(s,1H),2.84(t,J=5.6Hz,2H),2.65(s,2H),2.19(t,J=7.6Hz,1H),2.07(t,J=14.4Hz,3H).

实施例2:1'-((7-(1,1-二氟乙基)-6-氧代-5,6-二氢-1,5-萘啶-3-基)甲基)-2-氟-N-甲基-1',2',3',6'-四氢-[3,4'-联吡啶]-6-甲酰胺
Example 2: 1'-((7-(1,1-difluoroethyl)-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)-2-fluoro-N-methyl-1',2',3',6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide

第一步:将化合物1b(0.68g,2.30mmol)溶于DCM(30mL)中,0℃,N2氛围下,缓慢滴加DIBAL-H(1M,11.5mL,11.5mmol),在25℃下搅拌1h。在反应液缓慢倒入100mL饱和酒石酸钾钠溶液淬灭,DCM萃取5*50mL,硫酸钠干燥,抽滤减压浓缩得到化合物2a(0.59g,crude)。LC-MS:ESI[M+H]+=257.2。Step 1: Dissolve compound 1b (0.68 g, 2.30 mmol) in DCM (30 mL), slowly add DIBAL-H (1 M, 11.5 mL, 11.5 mmol) at 0°C under N2 atmosphere, and stir at 25°C for 1 h. Slowly pour 100 mL of saturated sodium potassium tartrate solution into the reaction solution to quench, extract with DCM 5*50 mL, dry with sodium sulfate, filter and concentrate under reduced pressure to obtain compound 2a (0.59 g, crude). LC-MS: ESI [M+H] + = 257.2.

第二步:将化合物2a(0.59g,2.32mmol)溶于DCM(20mL)中,0℃下滴加TEA(2.0mL,13.0mmol),MsCl(0.8mL,7.10mmol)升至25℃搅拌1h。在反应液缓慢倒入20ml饱和碳酸氢钠溶液淬灭,DCM萃取4*30ml,硫酸钠干燥,抽滤减压浓缩得到化合 物2b(0.60g,crude)。LC-MS:ESI[M+H]+=333.2。Step 2: Compound 2a (0.59 g, 2.32 mmol) was dissolved in DCM (20 mL), TEA (2.0 mL, 13.0 mmol) and MsCl (0.8 mL, 7.10 mmol) were added dropwise at 0 °C, and the mixture was heated to 25 °C and stirred for 1 h. 20 ml of saturated sodium bicarbonate solution was slowly poured into the reaction solution to quench, and 4*30 ml of DCM was extracted, dried over sodium sulfate, and filtered and concentrated under reduced pressure to obtain compound Material 2b (0.60g, crude). LC-MS: ESI[M+H] + =333.2.

第三步:将化合物2b(0.20g,0.60mmol)溶于乙腈(10mL)中,加入DIPEA(0.2mL,1.55mmol),化合物int-3(0.2g,0.85mmol)在80℃搅拌2h。在反应液缓慢倒入20ml饱和氯化铵溶液淬灭,EA萃取5*30ml,硫酸钠干燥,抽滤减压浓缩,Pre-TLC(DCM/MeOH=20/1)得到白色固体化合物2c(0.11g,0.23mmol,收率为38%)。LC-MS:ESI[M+H]+=472.1。Step 3: Compound 2b (0.20 g, 0.60 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (0.2 mL, 1.55 mmol) was added. Compound int-3 (0.2 g, 0.85 mmol) was stirred at 80°C for 2 h. 20 ml of saturated ammonium chloride solution was slowly poured into the reaction solution for quenching, 5*30 ml of EA was extracted, dried over sodium sulfate, filtered and concentrated under reduced pressure, and Pre-TLC (DCM/MeOH=20/1) obtained white solid compound 2c (0.11 g, 0.23 mmol, yield 38%). LC-MS: ESI[M+H] + =472.1.

第四步:将化合物2c(0.11g,0.23mmol)溶于dioxane(10mL)中,缓慢滴加HBr(46%in H2O,5.0mL),在70℃下搅拌1h。将反应液冷却后缓慢倒入100mL饱和碳酸氢钠溶液中淬灭,DCM/MeOH(20/1)萃取5*30mL,硫酸钠干燥,反向制备纯化得到白色固体化合物2(20mg,0.043mmol,收率为19%)。LC-MS:ESI[M+H]+=458.2。1H NMR(400MHz,CDCl3)δ8.52(d,J=1.6Hz,1H),8.24(s,1H),7.99(dd,J=7.6,1.6Hz,1H),7.86–7.71(m,2H),7.65(s,1H),6.16(s,1H),3.75(s,2H),3.23(d,J=3.2Hz,2H),2.97(d,J=5.2Hz,3H),2.73(m,4H),2.06(t,J=18.8Hz,3H)。Step 4: Compound 2c (0.11 g, 0.23 mmol) was dissolved in dioxane (10 mL), HBr (46% in H 2 O, 5.0 mL) was slowly added dropwise, and stirred at 70°C for 1 h. The reaction solution was cooled and slowly poured into 100 mL of saturated sodium bicarbonate solution for quenching, extracted with DCM/MeOH (20/1) for 5*30 mL, dried over sodium sulfate, and purified by reverse preparation to obtain white solid compound 2 (20 mg, 0.043 mmol, yield 19%). LC-MS: ESI [M+H] + = 458.2. 1 H NMR (400MHz, CDCl 3 )δ8.52(d,J=1.6Hz,1H),8.24(s,1H),7.99(dd,J=7.6,1.6Hz,1H),7.86–7.71(m,2H),7.65(s,1H),6.16 (s,1H),3.75(s,2H),3.23(d,J=3.2Hz,2H),2.97(d,J=5.2Hz,3H),2.73(m,4H),2.06(t,J=18.8Hz,3H).

实施例3-实施例9的化合物3~9的制备方法参考实施例2。The preparation methods of compounds 3 to 9 of Examples 3 to 9 can be referred to Example 2.

表6化合物3~9

Table 6 Compounds 3 to 9

实施例10:N-甲基-1'-((7-(甲硫基)-6-氧代-5,6-二氢-1,5-萘啶-3-基)甲基)-1'-,2',3',6'-四氢-[3,4'-联吡啶]-6-甲酰胺
Example 10: N-methyl-1'-((7-(methylthio)-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)-1'-, 2', 3', 6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide

第一步:将化合物int-18(0.1g,0.3mmol)溶于乙腈(5mL)中,加入DIPEA(0.3mL,1.80mmol),KI(0.02g),化合物int-2(0.1g,0.46mmol)在80℃搅拌2h。在反应液缓慢倒入20ml饱和氯化铵溶液淬灭,EA萃取4*30ml,硫酸钠干燥,Prep-TLC(DCM/MeOH=20/1)纯化得到黄色油状化合物10a(0.09g,0.21mmol,收率为68%)。LC-MS:ESI[M+H]+=436.2。Step 1: Dissolve compound int-18 (0.1 g, 0.3 mmol) in acetonitrile (5 mL), add DIPEA (0.3 mL, 1.80 mmol), KI (0.02 g), and compound int-2 (0.1 g, 0.46 mmol) and stir at 80 ° C for 2 h. Slowly pour 20 ml of saturated ammonium chloride solution into the reaction solution for quenching, extract with EA 4*30 ml, dry with sodium sulfate, and purify by Prep-TLC (DCM/MeOH=20/1) to obtain yellow oil compound 10a (0.09 g, 0.21 mmol, yield 68%). LC-MS: ESI[M+H] + =436.2.

第二步:将化合物10a(0.09g,0.21mmol)溶于二恶烷(10mL)中,缓慢滴加HBr(46%in H2O,5.0mL),在70℃下搅拌1h。将反应液冷却后缓慢倒入100mL饱和碳酸氢钠溶液中淬灭,DCM萃取4*50mL,硫酸钠干燥,反向制备纯化得到白色固体化合物10(30mg,0.07mmol,收率为34%)。LC-MS:ESI[M+H]+=422.2。1H NMR(400MHz,MeOD)δ8.70(s,1H),8.51(d,J=1.8Hz,1H),8.07–7.93(m,2H),7.78(s,1H),7.62(s,1H),6.38(s,1H),3.82(s,2H),2.96(s,3H),2.84(t,J=5.7Hz,2H),2.65(s,2H),2.53(s,3H),2.20(t,J=7.6Hz,2H)。Step 2: Compound 10a (0.09 g, 0.21 mmol) was dissolved in dioxane (10 mL), HBr (46% in H 2 O, 5.0 mL) was slowly added dropwise, and stirred at 70°C for 1 h. The reaction solution was cooled and slowly poured into 100 mL of saturated sodium bicarbonate solution for quenching, extracted with 4*50 mL of DCM, dried with sodium sulfate, and purified by reverse preparation to obtain white solid compound 10 (30 mg, 0.07 mmol, yield 34%). LC-MS: ESI [M+H] + = 422.2. 1H NMR(400MHz,MeOD)δ8.70(s,1H),8.51(d,J=1.8Hz,1H),8.07–7.93(m,2H),7.78(s,1H),7.62(s,1H),6.38 (s,1H),3.82(s,2H),2.96(s,3H),2.84(t,J=5.7Hz,2H),2.65(s,2H),2.53(s,3H),2.20(t,J=7.6Hz,2H).

实施例11-实施例30的化合物11~30的制备方法参考实施例10。The preparation methods of compounds 11 to 30 of Examples 11 to 30 can be referred to Example 10.

表7化合物11~30





Table 7 Compounds 11 to 30





实施例31:2-氟-N-甲基-1'-((6-氧代-7-((三氟甲基)硫代)-5,6-二氢-1,5-萘吡啶-3-基)甲基)-1'-,2',3',6'-四氢-[3,4'-联吡啶]-6-甲酰胺
Example 31: 2-Fluoro-N-methyl-1'-((6-oxo-7-((trifluoromethyl)thio)-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)-1'-, 2', 3', 6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide

第一步:将化合物int-17(0.2g,0.54mmol)溶于乙腈(5mL)中,加入DIPEA(0.45mL,2.70mmol),KI(0.02g),化合物int-3(0.2g,0.85mmol)在80℃搅拌2h。在反应液缓慢倒入20ml饱和氯化铵溶液淬灭,EA萃取4*30ml,硫酸钠干燥,Pre-TLC(DCM/MeOH=20/1)纯化得到黄色油状化合物31a(0.09g,0.18mmol,收率为32%)。LC-MS:ESI[M+H]+=436.2。Step 1: Dissolve compound int-17 (0.2 g, 0.54 mmol) in acetonitrile (5 mL), add DIPEA (0.45 mL, 2.70 mmol), KI (0.02 g), and compound int-3 (0.2 g, 0.85 mmol) and stir at 80°C for 2 h. Slowly pour 20 ml of saturated ammonium chloride solution into the reaction solution for quenching, extract with EA 4*30 ml, dry with sodium sulfate, and purify by Pre-TLC (DCM/MeOH=20/1) to obtain yellow oily compound 31a (0.09 g, 0.18 mmol, yield 32%). LC-MS: ESI[M+H] + =436.2.

第二步:将化合物31a(0.09g,0.18mmol)溶于二恶烷(10mL)中,缓慢滴加HBr(46%in H2O,5.0mL),在70℃下搅拌1h。将反应液冷却后缓慢倒入100mL饱和碳酸氢钠溶液中淬灭,DCM萃取4*50mL,硫酸钠干燥,反向制备纯化得到白色固体化合物31(20mg,0.04mmol,收率为22%)。LC-MS:ESI[M+H]+=494.2。1H NMR(400MHz,DMSO)δ8.64(d,J=4.9Hz,1H),8.54(d,J=1.5Hz,1H),8.33(s,1H),8.09(dd,J=9.8,7.8Hz,1H),7.96–7.83(m,1H),7.75(s,1H),6.26(s,1H),3.78(s,2H),3.19(d,J=2.5Hz,2H),2.79(d,J=4.8Hz,3H),2.70(t,J=5.5Hz,2H),2.52(s,2H)。Step 2: Compound 31a (0.09 g, 0.18 mmol) was dissolved in dioxane (10 mL), HBr (46% in H 2 O, 5.0 mL) was slowly added dropwise, and stirred at 70°C for 1 h. The reaction solution was cooled and slowly poured into 100 mL of saturated sodium bicarbonate solution for quenching, extracted with 4*50 mL of DCM, dried with sodium sulfate, and purified by reverse preparation to obtain white solid compound 31 (20 mg, 0.04 mmol, yield 22%). LC-MS: ESI [M+H] + = 494.2. 1H NMR (400MHz, DMSO) δ8.64(d,J=4.9Hz,1H),8.54(d,J=1.5Hz,1H),8.33(s,1H),8.09(dd,J=9.8,7.8Hz,1H),7.96–7.83(m,1 H), 7.75 (s, 1H), 6.26 (s, 1H), 3.78 (s, 2H), 3.19 (d, J = 2.5Hz, 2H), 2.79 (d, J = 4.8Hz, 3H), 2.70 (t, J = 5.5Hz, 2H), 2.52 (s, 2H).

实施例32-实施例40的化合物32~40的制备方法参考实施例31。The preparation methods of compounds 32-40 of Examples 32-40 can be referred to Example 31.

表8化合物32~40


Table 8 Compounds 32-40


实施例41:1'-((7-(二氟甲基)硫基)-6-氧代-5,6-二氢-1,5-萘啶-3-基)甲基)-N-甲基-1',2',3',6'-四氢-[3,4'-联吡啶]-6-甲酰胺
Example 41: 1'-((7-(difluoromethyl)thio)-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)-N-methyl-1',2',3',6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide

第一步:将化合物int-22(0.2g,0.5mmol)溶于乙腈(5mL)中,加入DIPEA(0.45mL,2.70mmol),KI(0.02g),化合物int-2(0.2g,0.85mmol)在80℃搅拌2h。在反应液缓慢倒入20ml饱和氯化铵溶液淬灭,EA萃取4*30ml,硫酸钠干燥,Pre-TLC(DCM/MeOH=20/1)纯化得到黄色油状化合物41a(0.1g,0.22mmol,收率为32%)。LC-MS:ESI[M+H]+=472.2。Step 1: Dissolve compound int-22 (0.2 g, 0.5 mmol) in acetonitrile (5 mL), add DIPEA (0.45 mL, 2.70 mmol), KI (0.02 g), and compound int-2 (0.2 g, 0.85 mmol) and stir at 80°C for 2 h. Slowly pour 20 ml of saturated ammonium chloride solution into the reaction solution for quenching, extract with EA 4*30 ml, dry with sodium sulfate, and purify by Pre-TLC (DCM/MeOH=20/1) to obtain yellow oily compound 41a (0.1 g, 0.22 mmol, yield 32%). LC-MS: ESI[M+H] + =472.2.

第二步:将化合物41a(0.084g,0.18mmol)溶于二恶烷(10mL)中,缓慢滴加HBr(46%in H2O,5.0mL),在70℃下搅拌1h。将反应液冷却后缓慢倒入100mL饱和碳酸氢钠溶液中淬灭,DCM萃取4*50mL,硫酸钠干燥,反向制备纯化得到白色固体化合物41(18mg,0.04mmol,收率为22%)。LC-MS:ESI[M+H]+=458.2。1H NMR(400MHz,DMSO)δ8.69(s,2H),8.48(s,1H),8.08(s,1H),7.99(dd,J=6.0,4.0Hz,2H),7.85(t,J=46.9Hz,1H),7.69(s,1H),6.43(s,1H),3.75(s,2H),3.17(s,3H),2.81(d,J=4.8Hz,3H),2.72(t,J=5.5Hz,2H),2.54(d,J=13.7Hz,2H)。Step 2: Compound 41a (0.084 g, 0.18 mmol) was dissolved in dioxane (10 mL), HBr (46% in H 2 O, 5.0 mL) was slowly added dropwise, and stirred at 70°C for 1 h. The reaction solution was cooled and slowly poured into 100 mL of saturated sodium bicarbonate solution for quenching, extracted with 4*50 mL of DCM, dried with sodium sulfate, and purified by reverse preparation to obtain white solid compound 41 (18 mg, 0.04 mmol, yield 22%). LC-MS: ESI [M+H] + = 458.2. 1H NMR (400MHz, DMSO) δ8.69(s,2H),8.48(s,1H),8.08(s,1H),7.99(dd,J=6.0,4.0Hz,2H),7.85(t,J=46.9Hz,1H),7.69 (s,1H),6.43(s,1H),3.75(s,2H),3.17(s,3H),2.81(d,J=4.8Hz,3H),2.72(t,J=5.5Hz,2H),2.54(d,J=13.7Hz,2H).

实施例42-实施例49的化合物42~49的制备方法参考实施例41。The preparation methods of compounds 42 to 49 of Examples 42 to 49 can be referred to Example 41.

表9化合物42~49


Table 9 Compounds 42 to 49


实施例50:2-氟-N-甲基-1'-((6-氧代-7-乙烯基-5,6-二氢-1,5-萘啶-3-基)甲基)-1',2', 3',6'-四氢-[3,4'-联吡啶]-6-甲酰胺
Example 50: 2-Fluoro-N-methyl-1'-((6-oxo-7-vinyl-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)-1', 2', 3',6'-Tetrahydro-[3,4'-bipyridine]-6-carboxamide

第一步:将化合物int-1(0.91g,2.92mmol)加入无水THF(300mL)中,将反应液冷却至-20℃,在-20℃的N2气氛下加入DIBAL-H(4.9mL,7.3m mol,1.5M甲苯溶液),将反应混合物在-15-0℃之间进一步搅拌3小时。TLC显示反应完全。用3N NaOH水溶液在-15-0℃之间缓慢猝灭反应,并保持内部温度不超过0℃。在25℃下减压除去挥发性物质,用乙酸乙酯(30mL*3)萃取所得物,用水(30mL)、盐水(30ml)洗涤合并的有机相,用无水Na2SO4干燥并蒸发至干。粗产物通过硅胶柱色谱法纯化(纯DCM,然后DCM/丙酮=30:1至10:1)。得到黄色固体的产物50a(0.54g)。LC-MS:ESI[M+H]+=269.2。Step 1: Compound int-1 (0.91 g, 2.92 mmol) was added to anhydrous THF (300 mL), the reaction solution was cooled to -20 ° C, DIBAL-H (4.9 mL, 7.3 mmol, 1.5 M toluene solution) was added under a N 2 atmosphere at -20 ° C, and the reaction mixture was further stirred between -15-0 ° C for 3 hours. TLC showed that the reaction was complete. The reaction was slowly quenched with 3N NaOH aqueous solution between -15-0 ° C, and the internal temperature was kept at no more than 0 ° C. The volatile substances were removed under reduced pressure at 25 ° C, and the resultant was extracted with ethyl acetate (30 mL*3), and the combined organic phase was washed with water (30 mL) and brine (30 ml), dried with anhydrous Na 2 SO 4 and evaporated to dryness. The crude product was purified by silica gel column chromatography (pure DCM, then DCM/acetone=30:1 to 10:1). The product 50a (0.54 g) was obtained as a yellow solid. LC-MS: ESI [M+H] + = 269.2.

第二步:在0-5℃氮气氛围下,向化合物50a(0.54g,2.0mmol)的DMF(100mL)溶液中缓慢加入SOCl2(357mg,3.0mmol),将混合物在25℃下搅拌3h,直到原料完全消耗掉。用冰水浴将反应混合物冷却至0-5℃,并用1N NaOH淬灭至pH=9,然后在搅拌下加入水(10mL)。将反应混合物在室温下搅拌1小时,通过过滤收集形成的灰白色沉淀物,用水(10mL*3)洗涤,真空干燥得化合物50b(0.32g)。LC-MS:ESI[M+H]+=287.0。Step 2: SOCl 2 (357 mg, 3.0 mmol) was slowly added to a DMF (100 mL) solution of compound 50a (0.54 g, 2.0 mmol) at 0-5°C under nitrogen atmosphere, and the mixture was stirred at 25°C for 3 h until the starting material was completely consumed. The reaction mixture was cooled to 0-5°C with an ice-water bath, quenched with 1N NaOH to pH = 9, and then water (10 mL) was added under stirring. The reaction mixture was stirred at room temperature for 1 hour, and the gray-white precipitate formed was collected by filtration, washed with water (10 mL*3), and dried in vacuo to obtain compound 50b (0.32 g). LC-MS: ESI [M+H] + = 287.0.

第三步:将化合物50b(0.42g,1.45mmol)溶于已经(10mL)中,加入化合物int-3(0.35g,1.45mmol),KI(0.10g,0.6mmol),滴加DIPEA(3.0mL,23mmol),在80℃下搅拌2h。将反应液冷却后缓慢倒入30mL饱和氯化铵溶液中淬灭,EA萃取4*30mL,硫酸钠干燥,柱层析分离(DCM/MeOH=20/1,添加5%三乙胺)得到白色固体化合物50c(0.47g,0.98mmol,收率为67%)。LC-MS:ESI[M+H]+=482.2/484.1。Step 3: Compound 50b (0.42 g, 1.45 mmol) was dissolved in 10 mL of ethanol, and compound int-3 (0.35 g, 1.45 mmol) and KI (0.10 g, 0.6 mmol) were added. DIPEA (3.0 mL, 23 mmol) was added dropwise, and the mixture was stirred at 80°C for 2 h. The reaction solution was cooled and slowly poured into 30 mL of saturated ammonium chloride solution for quenching, extracted with EA 4*30 mL, dried with sodium sulfate, and separated by column chromatography (DCM/MeOH=20/1, 5% triethylamine was added) to obtain white solid compound 50c (0.47 g, 0.98 mmol, yield was 67%). LC-MS: ESI[M+H] + =482.2/484.1.

第四步:将化合物50c(0.3g,0.62mmol)溶于dioxane(10mL)中,缓慢滴加HBr(46%in H2O,5.0mL),在70℃下搅拌1h。将反应液冷却后缓慢倒入100mL饱和碳酸氢钠溶液中淬灭,DCM萃取4*30mL,硫酸钠干燥,EA/PE(10/1)打浆得到白色固体化合物50d(0.35g,crude)。LC-MS:ESI[M+H]+=469.2/470.2。Step 4: Compound 50c (0.3 g, 0.62 mmol) was dissolved in dioxane (10 mL), HBr (46% in H 2 O, 5.0 mL) was slowly added dropwise, and stirred at 70°C for 1 h. The reaction solution was cooled and slowly poured into 100 mL of saturated sodium bicarbonate solution for quenching, extracted with 4*30 mL of DCM, dried with sodium sulfate, and slurried with EA/PE (10/1) to obtain white solid compound 50d (0.35 g, crude). LC-MS: ESI [M+H] + = 469.2/470.2.

第五步:将化合物50d(0.2g,0.423mmol)溶于dioxane(10mL)中,加入三正丁基乙烯基锡(0.27g,0.86mmol),XPhos-Pd-G2(0.05g,0.06mmol),N2置换三次后,于90℃ 下搅拌15h。冷却后浓缩,Pre-TLC(DCM/MeOH=20/1纯化后再经反相制备纯化得到化合物50(20mg,0.05mmol,收率为12%)。LC-MS:ESI[M+H]+=416.2。1H NMR(400MHz,CDCl3)δ10.75(s,1H),8.57(d,J=1.7Hz,1H),8.11(s,1H),8.05(d,J=5.0Hz,1H),7.96(d,J=7.9Hz,1H),7.67(s,1H),7.53(d,J=7.9Hz,1H),7.00(dd,J=17.7,11.4Hz,1H),6.24(dd,J=17.7,1.2Hz,1H),5.65(s,1H),5.54(dd,J=11.3,1.1Hz,1H),3.80(s,2H),3.22(d,J=2.9Hz,2H),3.03(d,J=5.1Hz,5H),2.77(s,2H),2.54(s,3H)。Step 5: Compound 50d (0.2 g, 0.423 mmol) was dissolved in dioxane (10 mL), tri-n-butyl vinyl tin (0.27 g, 0.86 mmol), XPhos-Pd-G 2 (0.05 g, 0.06 mmol) were added, and after N 2 substitution three times, the mixture was heated at 90°C. The mixture was stirred for 15 h. After cooling, it was concentrated and purified by Pre-TLC (DCM/MeOH=20/1) and then by reverse phase preparative purification to obtain compound 50 (20 mg, 0.05 mmol, yield 12%). LC-MS: ESI [M+H] + = 416.2. 1 H NMR (400 MHz, CDCl 3 )δ10.75(s,1H),8.57(d,J=1.7Hz,1H),8.11(s,1H),8.05(d,J=5.0Hz,1H),7.96 (d,J=7.9Hz,1H),7.67(s,1H),7.53(d,J=7.9Hz,1H),7.00(dd,J=17.7,11.4Hz,1 H),6.24(dd,J=17.7,1.2Hz,1H),5.65(s,1H),5.54(dd,J=11.3,1.1Hz,1H),3.8 0(s,2H),3.22(d,J=2.9Hz,2H),3.03(d,J=5.1Hz,5H),2.77(s,2H),2.54(s,3H).

实施例51-实施例58的化合物51~58的制备方法参考实施例50。The preparation methods of compounds 51-58 of Examples 51-58 can be referred to Example 50.

表10化合物51~58

Table 10 Compounds 51 to 58

实施例59:N-甲基-5-(4-(((7-(甲硫基)-6-氧代-5,6-二氢-1,5-萘啶-3-基)甲基)哌嗪-1-基)吡啶甲酰胺
Example 59: N-methyl-5-(4-(((7-(methylthio)-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)piperazin-1-yl)picolinamide

第一步:将化合物int-18(0.1g,0.3mmol)溶于ACN(10mL)中,加入DIPEA(0.3mL,1.8mmol),KI(0.02g),化合物int-9(0.1g,0.45mmol)在80℃搅拌2h。在反应液缓慢倒入20ml饱和氯化铵溶液淬灭,EA萃取4*30ml,硫酸钠干燥,Prep-TLC(DCM/MeOH=10/1)纯化得到白色固体化合物59a(0.12g,0.27mmol,收率为91%)。LC-MS:ESI[M+H]+=439。Step 1: Dissolve compound int-18 (0.1 g, 0.3 mmol) in ACN (10 mL), add DIPEA (0.3 mL, 1.8 mmol), KI (0.02 g), and compound int-9 (0.1 g, 0.45 mmol) and stir at 80°C for 2 h. Slowly pour 20 ml of saturated ammonium chloride solution into the reaction solution to quench, extract with 4*30 ml of EA, dry with sodium sulfate, and purify with Prep-TLC (DCM/MeOH=10/1) to obtain white solid compound 59a (0.12 g, 0.27 mmol, yield 91%). LC-MS: ESI[M+H] + =439.

将化合物59a(0.12g,0.27mmol)溶于dioxane(10mL)中,缓慢滴加HBr(46%in H2O,5.0mL),在70℃下搅拌1h。将反应液冷却后缓慢倒入100mL饱和碳酸氢钠溶液中淬灭,DCM萃取4x 30mL,硫酸钠干燥,反向制备纯化得到白色固体化合物59(35mg,0.082mmol,收率为31%)。LC-MS:ESI[M+H]+=425.2。1H NMR(400MHz,CDCl3)δ8.52(s,1H),8.16(d,J=2.4Hz,1H),8.05(d,J=8.7Hz,1H),7.77(s,1H),7.64(d,J=12.2Hz,2H),7.22(d,J=8.8Hz,1H),3.70(s,2H),3.36(s,4H),3.01(d,J=5.0Hz,3H),2.67(s,4H),2.50(s,3H)。Compound 59a (0.12 g, 0.27 mmol) was dissolved in dioxane (10 mL), HBr (46% in H 2 O, 5.0 mL) was slowly added dropwise, and stirred at 70° C. for 1 h. The reaction solution was cooled and slowly poured into 100 mL of saturated sodium bicarbonate solution for quenching, extracted with DCM 4 x 30 mL, dried with sodium sulfate, and purified by reverse preparation to obtain white solid compound 59 (35 mg, 0.082 mmol, yield 31%). LC-MS: ESI [M+H] + = 425.2. 1 H NMR (400MHz, CDCl 3 )δ8.52(s,1H),8.16(d,J=2.4Hz,1H),8.05(d,J=8.7Hz,1H),7.77(s,1H),7.64(d,J=12.2Hz,2H), 7.22(d,J=8.8Hz,1H),3.70(s,2H),3.36(s,4H),3.01(d,J=5.0Hz,3H),2.67(s,4H),2.50(s,3H).

实施例60:2-氟-1-((7-((氟甲基)硫基)-6-氧代-5,6-二氢-1,5-萘啶-3-基)甲基)-N-甲基-1',2',3',6'-四氢-[3,4'-联吡啶]-6-甲酰胺
Example 60: 2-Fluoro-1-((7-((fluoromethyl)thio)-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)-N-methyl-1',2',3',6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide

第一步:将化合物int-1(5.0g,16.08mmol)溶于DCM(100mL)中,0℃下滴加DIBAL-H(1.0M,50mL,50.0mmol),0℃搅拌1h。缓慢滴加100mL饱和酒石酸钾钠溶液,DCM萃取4*30mL,硫酸钠干燥,减压浓缩得到化合物60a(4.34g,crude)。LC-MS:ESI[M+H]+=270.1。Step 1: Dissolve compound int-1 (5.0 g, 16.08 mmol) in DCM (100 mL), add DIBAL-H (1.0 M, 50 mL, 50.0 mmol) dropwise at 0°C, and stir at 0°C for 1 h. Slowly add 100 mL of saturated sodium potassium tartrate solution, extract with DCM 4*30 mL, dry with sodium sulfate, and concentrate under reduced pressure to obtain compound 60a (4.34 g, crude). LC-MS: ESI [M+H] + = 270.1.

第二步:将化合物60a(4.34g,16.08mmol)溶于DCM(100mL)中,滴加DIPEA(10mL,80.4mmol),在0℃缓慢滴加MOMCl(2.5mL,32.16mmol),于0℃下搅拌2h。将反应液冷却后缓慢倒入100mL饱和碳酸氢钠溶液中淬灭,DCM萃取4*30mL,硫酸钠干燥,减压浓缩,柱层析(EA/PE=1/3),得到化合物60b(2.44g,7.79mmol)。LC-MS:ESI[M+H]+=313.1/315.1。Step 2: Compound 60a (4.34 g, 16.08 mmol) was dissolved in DCM (100 mL), and DIPEA (10 mL, 80.4 mmol) was added dropwise. MOMCl (2.5 mL, 32.16 mmol) was slowly added dropwise at 0°C, and stirred at 0°C for 2 h. The reaction solution was cooled and slowly poured into 100 mL of saturated sodium bicarbonate solution for quenching, extracted with DCM 4*30 mL, dried with sodium sulfate, concentrated under reduced pressure, and column chromatography (EA/PE=1/3) was performed to obtain compound 60b (2.44 g, 7.79 mmol). LC-MS: ESI[M+H] + =313.1/315.1.

第三步:将化合物60b(2.44g,7.79mmol)溶于甲苯(20mL)中,加入DIPEA(3mL,18.18mmol),Pd2(dba)3(0.74g,0.8mmol),Xantphos(0.9g,1.56mmol),化合物int-17(3.4g,15.63mmol),N2氛围下,在110℃搅拌12h。减压浓缩,柱层析(EA/PE=1/3)得到化合物60c(3.2g,7.34mmol,收率为91%)。LC-MS:ESI[M+H]+=437.1。Step 3: Compound 60b (2.44 g, 7.79 mmol) was dissolved in toluene (20 mL), and DIPEA (3 mL, 18.18 mmol), Pd2(dba)3 (0.74 g, 0.8 mmol), Xantphos (0.9 g, 1.56 mmol), and compound int-17 (3.4 g, 15.63 mmol) were added. The mixture was stirred at 110°C for 12 h under N2 atmosphere. The mixture was concentrated under reduced pressure and column chromatography (EA/PE=1/3) was performed to obtain compound 60c (3.2 g, 7.34 mmol, yield 91%). LC-MS: ESI[M+H] + =437.1.

第四步:将化合物60c(3.2g,7.32mmol)溶于EtOH(50mL)中,加入EtONa(1.5g,22.05mmol),在25℃搅拌2h。将反应液缓慢倒入HCl(0.5M,100mL)水溶液中淬灭,EA萃取5*50mL,硫酸钠干燥,减压浓缩得到化合物60d(3.0g,crude)。LC-MS:ESI[M+H]+=267.2。Step 4: Compound 60c (3.2 g, 7.32 mmol) was dissolved in EtOH (50 mL), EtONa (1.5 g, 22.05 mmol) was added, and the mixture was stirred at 25°C for 2 h. The reaction solution was slowly poured into an aqueous solution of HCl (0.5 M, 100 mL) for quenching, extracted with EA 5*50 mL, dried over sodium sulfate, and concentrated under reduced pressure to obtain compound 60d (3.0 g, crude). LC-MS: ESI [M+H] + = 267.2.

第五步:将化合物60d(1.0g,3.76mmol)溶于MeCN(10mL)中,加入Cs2CO3(21.2g,3.76mmol),滴加氟碘甲烷至该反应液中,25℃下搅拌3小时。将反应液倒入饱和氯化铵(50mL)中淬灭,EA萃取5*30mL,硫酸钠干燥,减压浓缩后柱层析(EA/PE=1/2),得到化合物60e(0.65g,2.17mmol,59%)。LC-MS:ESI[M+H]+=299.2。Step 5: Compound 60d (1.0 g, 3.76 mmol) was dissolved in MeCN (10 mL), Cs 2 CO 3 (21.2 g, 3.76 mmol) was added, and iodomethane was added dropwise to the reaction solution, and stirred at 25° C. for 3 hours. The reaction solution was poured into saturated ammonium chloride (50 mL) for quenching, extracted with EA 5*30 mL, dried over sodium sulfate, concentrated under reduced pressure, and column chromatographed (EA/PE=1/2) to obtain compound 60e (0.65 g, 2.17 mmol, 59%). LC-MS: ESI[M+H] + =299.2.

第六步:将化合物60e(0.65g,2.17mmol)溶于HCl(4.0M in MeOH,10mL)中,25℃下搅拌2h,减压浓缩后加入100mL饱和碳酸氢钠溶液淬灭,DCM萃取4*30mL, 硫酸钠干燥,减压浓缩得到化合物60f(0.6g,crude)。LC-MS:ESI[M+H]+=255.2。Step 6: Compound 60e (0.65 g, 2.17 mmol) was dissolved in HCl (4.0 M in MeOH, 10 mL), stirred at 25 °C for 2 h, concentrated under reduced pressure, quenched with 100 mL of saturated sodium bicarbonate solution, extracted with DCM for 4*30 mL, The residue was dried over sodium sulfate and concentrated under reduced pressure to give compound 60f (0.6 g, crude). LC-MS: ESI [M+H] + = 255.2.

第七步:将化合物60f(0.6g,2.35mmol)溶于DCM(10mL)中,加入DMF(0.01mL),25℃下滴加SOCl2(0.4mL,4.70mmol),25℃搅拌1h。抽滤减压浓缩得到化合物60g(0.6g,crude)。LC-MS:ESI[M+H]+=273.2。Step 7: Compound 60f (0.6 g, 2.35 mmol) was dissolved in DCM (10 mL), DMF (0.01 mL) was added, SOCl 2 (0.4 mL, 4.70 mmol) was added dropwise at 25°C, and the mixture was stirred for 1 h at 25°C. The mixture was filtered and concentrated under reduced pressure to obtain compound 60g (0.6 g, crude). LC-MS: ESI [M+H] + = 273.2.

第八步:将化合物60g(0.60g,2.20mmol)溶于dioxane(10mL)中,缓慢滴加HBr(48%in AcOH,3.0mL),在40℃下搅拌1h。将反应液冷却后缓慢倒入100mL饱和碳酸氢钠溶液中淬灭,抽滤得到白色固体化合物60h(0.70g,crude)。LC-MS:ESI[M+H]+=259.1。Step 8: Dissolve compound 60g (0.60g, 2.20mmol) in dioxane (10mL), slowly add HBr (48% in AcOH, 3.0mL) dropwise, and stir at 40°C for 1h. After cooling, slowly pour the reaction solution into 100mL saturated sodium bicarbonate solution to quench, and filter to obtain white solid compound 60h (0.70g, crude). LC-MS: ESI [M+H] + = 259.1.

第九步:将化合物60h(0.1g,0.39mmol)溶于ACN(10mL)中,加入DIPEA(0.2mL,1.21mmol),KI(0.02g),化合物7(0.1g,0.42mmol)在80℃搅拌2h。减压旋干反相制备得到白色固体化合物60(0.08g,0.17mmol,收率为45%)。LC-MS:ESI[M+H]+=458.1。1H NMR(400MHz,DMSO)δ12.66(s,1H),8.76-8.54(m,2H),8.14(dd,J=9.8,7.8Hz,1H),8.04-7.92(m,2H),7.87(d,J=1.3Hz,1H),6.32-6.05(m,3H),4.59(s,2H),3.90(s,3H),3.36(s,1H),2.80(d,J=4.8Hz,5H)。Step 9: Compound 60h (0.1 g, 0.39 mmol) was dissolved in ACN (10 mL), and DIPEA (0.2 mL, 1.21 mmol), KI (0.02 g), and compound 7 (0.1 g, 0.42 mmol) were added and stirred at 80°C for 2 h. The mixture was dried under reduced pressure and the mixture was reversed to obtain a white solid compound 60 (0.08 g, 0.17 mmol, yield 45%). LC-MS: ESI [M+H] + = 458.1. 1H NMR (400MHz, DMSO) δ12.66 (s, 1H), 8.76-8.54 (m, 2H), 8.14 (dd, J = 9.8, 7.8Hz, 1H), 8.04-7.92 (m, 2H), 7 .87(d,J=1.3Hz,1H),6.32-6.05(m,3H),4.59(s,2H),3.90(s,3H),3.36(s,1H),2.80(d,J=4.8Hz,5H).

实施例61-实施例63的化合物61~63的制备方法参考实施例60。The preparation methods of compounds 61-63 of Examples 61-63 can be referred to Example 60.

表11化合物61~63

Table 11 Compounds 61-63

实施例64:6-氟-N-甲基-5-(1-((7-(甲硫基)-6-氧代-56-二氢-1,5-萘啶-3-基)甲基)哌啶-4-基)吡啶甲酰胺
Example 64: 6-Fluoro-N-methyl-5-(1-((7-(methylthio)-6-oxo-5-(6-dihydro-1,5-naphthyridin-3-yl)methyl)piperidin-4-yl)picolinamide

第一步:将化合物int-18b(0.59g,2.50mmol)溶于DCM(10mL)中,滴加DMF(0.01mL),0℃下滴加SOCl2(0.60mL,5.00mmol),25℃搅拌1h。减压浓缩除去溶剂得到化合物64a(0.64g,crude)。LC-MS:ESI[M+H]+=255.2。Step 1: Dissolve compound int-18b (0.59 g, 2.50 mmol) in DCM (10 mL), add DMF (0.01 mL) dropwise, add SOCl 2 (0.60 mL, 5.00 mmol) dropwise at 0°C, and stir at 25°C for 1 h. Concentrate under reduced pressure to remove the solvent to obtain compound 64a (0.64 g, crude). LC-MS: ESI [M+H] + = 255.2.

第二步:将化合物64a(0.64g,2.50mmol)溶于dioxane(10mL)中,缓慢滴加HBr(38%in AcOH,2.0mL),在40℃下搅拌1h。将反应液冷却后缓慢倒入100mL饱和碳酸 氢钠溶液中淬灭,过滤得到滤饼,DCM/MeOH(20/1)洗涤滤饼,滤液减压浓缩得到化合物64b(0.6g,crude)。LC-MS:ESI[M+H]+=241.1。Step 2: Compound 64a (0.64 g, 2.50 mmol) was dissolved in dioxane (10 mL), HBr (38% in AcOH, 2.0 mL) was slowly added dropwise, and stirred at 40°C for 1 h. After the reaction solution was cooled, 100 mL of saturated carbonic acid was slowly poured into the mixture. The mixture was quenched in sodium hydrogen solution, filtered to obtain a filter cake, washed with DCM/MeOH (20/1), and the filtrate was concentrated under reduced pressure to obtain compound 64b (0.6 g, crude). LC-MS: ESI [M+H] + = 241.1.

第三步:将化合物64b(0.1g,0.42mmol)溶于乙腈(10mL)中,加入DIPEA(0.3mL,2.1mmol),KI(0.02g),化合物int-23(0.1g,0.42mmol)在80℃搅拌2h。减压浓缩后反相制备,得到化合物64(0.03g,0.068mmol,收率为16%)。LC-MS:ESI[M+H]+=442.2。1H NMR(400MHz,DMSO)δ12.11(s,1H),8.61(q,J=4.4Hz,1H),8.40(d,J=1.6Hz,1H),8.06(dd,J=9.5,7.9Hz,1H),7.96-7.82(m,1H),7.62(d,J=1.1Hz,1H),7.54(s,1H),3.62(s,2H),2.94(d,J=11.4Hz,2H),2.79(t,J=8.8Hz,4H),2.45(s,3H)2.13(dd,J=11.4,9.0Hz,2H),1.81-1.62(m,4H)。Step 3: Compound 64b (0.1 g, 0.42 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (0.3 mL, 2.1 mmol), KI (0.02 g), and compound int-23 (0.1 g, 0.42 mmol) were added and stirred at 80°C for 2 h. After concentration under reduced pressure, the mixture was prepared by reverse phase to obtain compound 64 (0.03 g, 0.068 mmol, yield 16%). LC-MS: ESI [M+H] + = 442.2. 1H NMR (400MHz, DMSO) δ12.11(s,1H),8.61(q,J=4.4Hz,1H),8.40(d,J=1.6Hz,1H),8.06(dd,J=9.5,7.9Hz,1H),7.96-7.82(m,1H),7.62(d,J=1.1 Hz,1H),7.54(s,1H),3.62(s,2H),2.94(d,J=11.4Hz,2H),2.79(t,J=8.8Hz,4H),2.45(s,3H)2.13(dd,J=11.4,9.0Hz,2H),1.81-1.62(m,4H).

实施例65:5-(4-(((7-(1,1-二氟乙基)-6-氧代-56-二氢-1,5-萘啶-3-基)甲基)哌嗪-1-基)-6-氟-N-甲基吡啶酰胺
Example 65: 5-(4-(((7-(1,1-difluoroethyl)-6-oxo-56-dihydro-1,5-naphthyridin-3-yl)methyl)piperazin-1-yl)-6-fluoro-N-methylpicolinamide

第一步:将化合物2a(0.12g,0.47mmol)溶于DCM(10mL)中,加入DMF(0.01mL),25℃下滴加SOCl2(0.1mL,0.87mmol),25℃搅拌1h。减压浓缩除去溶剂得到化合物65a(0.12g,crude)。LC-MS:ESI[M+H]+=273.2。Step 1: Dissolve compound 2a (0.12 g, 0.47 mmol) in DCM (10 mL), add DMF (0.01 mL), add SOCl 2 (0.1 mL, 0.87 mmol) dropwise at 25°C, and stir at 25°C for 1 h. Concentrate under reduced pressure to remove the solvent to obtain compound 65a (0.12 g, crude). LC-MS: ESI [M+H] + = 273.2.

第二步:将化合物65a(0.12g,0.43mmol)溶于dioxane(10mL)中,缓慢滴加HBr(38%in AcOH,2.0mL),在40℃下搅拌1h。将反应液冷却后缓慢倒入100mL饱和碳酸氢钠溶液中淬灭,减压过滤得到白色固体化合物65b(0.12g,crude)。LC-MS:ESI[M+H]+=259.2。Step 2: Compound 65a (0.12 g, 0.43 mmol) was dissolved in dioxane (10 mL), HBr (38% in AcOH, 2.0 mL) was slowly added dropwise, and stirred at 40°C for 1 h. The reaction solution was cooled and slowly poured into 100 mL of saturated sodium bicarbonate solution for quenching, and filtered under reduced pressure to obtain white solid compound 65b (0.12 g, crude). LC-MS: ESI [M+H] + = 259.2.

第三步:将化合物65b(0.06g,0.23mmol)溶于乙腈(10mL)中,加入DIPEA(0.15mL,1.0mmol),KI(0.01g),化合物int-10(0.05g,0.21mmol)在80℃搅拌2h。减压浓缩除去溶剂,残留物Pre-HPLC纯化得到白色固体化合物65(0.02g,0.043mmol,收率为19%)。LC-MS:ESI[M+H]+=461.2。1H NMR(400MHz,DMSO)δ12.24(s,1H),8.53(d,J=1.7Hz,1H),8.41(q,J=4.6Hz,1H),8.09(s,1H),7.85(dd,J=8.0,1.2Hz,1H),7.71(d,J=1.1Hz,1H),7.58(dd,J=10.6,8.2Hz,1H),3.72(s,2H),3.19(d,J=4.7Hz,4H),2.77(d,J=4.8Hz,3H),2.60(d,J=4.3Hz,4H),2.06(t,J=19.4Hz,3H)。 Step 3: Compound 65b (0.06 g, 0.23 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (0.15 mL, 1.0 mmol), KI (0.01 g), and compound int-10 (0.05 g, 0.21 mmol) were added and stirred at 80°C for 2 h. The solvent was removed by concentration under reduced pressure, and the residue was purified by Pre-HPLC to obtain white solid compound 65 (0.02 g, 0.043 mmol, yield 19%). LC-MS: ESI [M+H] + = 461.2. 1H NMR (400MHz, DMSO) δ12.24(s,1H),8.53(d,J=1.7Hz,1H),8.41(q,J=4.6Hz,1H),8.09(s,1H),7.85(dd,J=8.0,1.2Hz,1H),7.71(d,J=1.1Hz, 1H), 7.58 (dd, J=10.6, 8.2Hz, 1H), 3.72 (s, 2H), 3.19 (d, J=4.7Hz, 4H), 2.77 (d, J=4.8Hz, 3H), 2.60 (d, J=4.3Hz, 4H), 2.06 (t, J=19.4Hz, 3H).

实施例66:5-(1-(((7-(1,1-二氟乙基)-6-氧代-56-二氢-1,5-萘啶-3-基)甲基)哌啶-4-基)-6-氟-N-甲基吡啶酰胺
Example 66: 5-(1-(((7-(1,1-difluoroethyl)-6-oxo-56-dihydro-1,5-naphthyridin-3-yl)methyl)piperidin-4-yl)-6-fluoro-N-methylpicolinamide

将化合物65b(0.06g,0.23mmol)溶于乙腈(10mL)中,加入DIPEA(0.15mL,1.0mmol),KI(0.01g),化合物int-23(0.06g,0.21mmol)在80℃搅拌2h。减压浓缩除去溶剂,残留物Pre-HPLC纯化得到化合物66(0.02g,0.043mmol,收率为19%)。LC-MS:ESI[M+H]+=460.2。1H NMR(400MHz,DMSO)δ8.61(d,J=4.8Hz,1H),8.51(d,J=1.7Hz,1H),8.12-7.99(m,2H),7.90(dd,J=7.6,1.3Hz,1H),7.69(d,J=1.0Hz,1H),3.67(s,2H),2.94(d,J=11.4Hz,2H),2.78(d,J=4.8Hz,4H),2.16(dd,J=11.3,8.7Hz,2H),2.05(dd,J=23.9,14.9Hz,3H),1.75(dt,J=12.0,6.9Hz,4H)。Compound 65b (0.06 g, 0.23 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (0.15 mL, 1.0 mmol), KI (0.01 g), and compound int-23 (0.06 g, 0.21 mmol) were added and stirred at 80°C for 2 h. The solvent was removed by concentration under reduced pressure, and the residue was purified by Pre-HPLC to obtain compound 66 (0.02 g, 0.043 mmol, yield 19%). LC-MS: ESI [M+H] + = 460.2. 1H NMR (400MHz, DMSO) δ8.61(d,J=4.8Hz,1H),8.51(d,J=1.7Hz,1H),8.12-7.99(m,2H),7.90(dd,J=7.6,1.3Hz,1H),7.69(d,J=1.0Hz,1H),3.67(s ,2H),2.94(d,J=11.4Hz,2H),2.78(d,J=4.8Hz,4H),2.16(dd,J=11.3,8.7Hz,2H),2.05(dd,J=23.9,14.9Hz,3H),1.75(dt,J=12.0,6.9Hz,4H).

实施例67:5-(4-((7-(二氟甲基)硫基)-6-氧代-5,6-二氢-1,5-萘啶-3-基)甲基)哌嗪-1-基)-N-甲基吡啶酰胺
Example 67: 5-(4-((7-(difluoromethyl)thio)-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)piperazin-1-yl)-N-methylpicolinamide

第一步:将化合物int-22c(1.0g,3.47mmol)溶于DCM(10mL)中,加入DMF(0.01mL),25℃下滴加SOCl2(1.0mL,6.94mmol),25℃搅拌1h。减压浓缩除去溶剂得到化合物67a(0.6g,crude)。LC-MS:ESI[M+H]+=291.2。Step 1: Dissolve compound int-22c (1.0 g, 3.47 mmol) in DCM (10 mL), add DMF (0.01 mL), add SOCl 2 (1.0 mL, 6.94 mmol) dropwise at 25°C, and stir at 25°C for 1 h. Concentrate under reduced pressure to remove the solvent to obtain compound 67a (0.6 g, crude). LC-MS: ESI [M+H] + = 291.2.

第二步:将化合物67a(1.0,3.44mmol)溶于dioxane(10mL)中,缓慢滴加HBr(38%in AcOH,3.0mL),在40℃下搅拌1h。将反应液冷却后缓慢倒入100mL饱和碳酸氢钠溶液中淬灭,抽滤得到白色固体化合物67b(0.90g,crude)。LC-MS:ESI[M+H]+=277.2。Step 2: Compound 67a (1.0, 3.44 mmol) was dissolved in dioxane (10 mL), HBr (38% in AcOH, 3.0 mL) was slowly added dropwise, and stirred at 40°C for 1 h. The reaction solution was cooled and slowly poured into 100 mL of saturated sodium bicarbonate solution for quenching, and filtered to obtain white solid compound 67b (0.90 g, crude). LC-MS: ESI [M+H] + = 277.2.

第三步:将化合物67b(0.1g,0.36mmol)溶于乙腈(10mL)中,加入DIPEA(0.2mL,1.21mmol),KI(0.02g),化合物int-2(0.1g,0.45mmol)在80℃搅拌2h。减压浓缩除去溶剂,残留物Pre-HPLC纯化得到化合物67(0.05g,0.11mmol,收率为30%)。LC-MS:ESI[M+H]+=461.2。1H NMR(400MHz,DMSO)δ8.48(d,J=1.3Hz,1H),8.39(d,J=4.9Hz,1H),8.27(d,J=2.8Hz,1H),8.08(s,1H),8.03-7.62(m,3H),7.39(dd,J=8.8,2.8Hz,1H),3.68(s,2H),3.38(s,4H),2.78(d,J=4.8Hz,3H),2.60-2.51(m,4H)。 Step 3: Compound 67b (0.1 g, 0.36 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (0.2 mL, 1.21 mmol), KI (0.02 g), and compound int-2 (0.1 g, 0.45 mmol) were added and stirred at 80°C for 2 h. The solvent was removed by concentration under reduced pressure, and the residue was purified by Pre-HPLC to obtain compound 67 (0.05 g, 0.11 mmol, yield 30%). LC-MS: ESI [M+H] + = 461.2. 1H NMR (400MHz, DMSO) δ8.48(d,J=1.3Hz,1H),8.39(d,J=4.9Hz,1H),8.27(d,J=2.8Hz,1H),8.08(s,1H),8.03-7 .62(m,3H),7.39(dd,J=8.8,2.8Hz,1H),3.68(s,2H),3.38(s,4H),2.78(d,J=4.8Hz,3H),2.60-2.51(m,4H).

实施例68:5-(1-((7-(二氟甲基)硫基)-6-氧代-5,6-二氢-1,5-萘啶-3-基)甲基)哌啶-4-基)-6-氟-N-甲基吡啶酰胺
Example 68: 5-(1-((7-(difluoromethyl)thio)-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl)piperidin-4-yl)-6-fluoro-N-methylpicolinamide

将化合物67b(0.1g,0.36mmol)溶于乙腈(10mL)中,加入DIPEA(0.2mL,1.21mmol),KI(0.02g),化合物int-23(0.1g,0.41mmol)在80℃搅拌2h。减压浓缩除去溶剂,残留物Pre-HPLC纯化得到化合物68(0.04g,0.08mmol,收率为23%)。LC-MS:ESI[M+H]+=478.2。1H NMR(400MHz,DMSO)δ12.79(s,1H),8.74-8.46(m,2H),8.14(s,1H),7.95(ddd,J=66.2,41.2,26.0Hz,4H),4.54(s,2H),3.56(d,J=11.6Hz,2H),3.28-3.04(m,3H),2.80(d,J=4.7Hz,3H),2.11-1.84(m,4H)。Compound 67b (0.1 g, 0.36 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (0.2 mL, 1.21 mmol), KI (0.02 g), and compound int-23 (0.1 g, 0.41 mmol) were added and stirred at 80°C for 2 h. The solvent was removed by concentration under reduced pressure, and the residue was purified by Pre-HPLC to obtain compound 68 (0.04 g, 0.08 mmol, yield 23%). LC-MS: ESI [M+H] + = 478.2. 1H NMR (400MHz, DMSO) δ12.79 (s, 1H), 8.74-8.46 (m, 2H), 8.14 (s, 1H), 7.95 (ddd, J=66.2, 41.2, 26.0Hz, 4 H), 4.54 (s, 2H), 3.56 (d, J = 11.6Hz, 2H), 3.28-3.04 (m, 3H), 2.80 (d, J = 4.7Hz, 3H), 2.11-1.84 (m, 4H).

实施例69:2-氟-N-甲基-1'-(3-(甲硫基)-2-氧代-1,2-二氢喹啉-7-基)甲基)-1',2',3',6'-四氢-[3,4'-联吡啶]-6-甲酰胺
Example 69: 2-Fluoro-N-methyl-1'-(3-(methylthio)-2-oxo-1,2-dihydroquinolin-7-yl)methyl)-1',2',3',6'-tetrahydro-[3,4'-bipyridine]-6-carboxamide

第一步:将化合物69a(1.0g,5.0mmol)溶于DCM(20mL)中,加入MnO2(7.0g,80.0mmol),25℃下搅拌15h。垫硅藻土过滤,收集滤液,减压浓缩后得到化合物69b(1.0g,crude)。LC-MS:ESI[M+H]+=200.1/202.1。Step 1: Compound 69a (1.0 g, 5.0 mmol) was dissolved in DCM (20 mL), MnO 2 (7.0 g, 80.0 mmol) was added, and the mixture was stirred at 25° C. for 15 h. The mixture was filtered through celite, and the filtrate was collected and concentrated under reduced pressure to obtain compound 69b (1.0 g, crude). LC-MS: ESI [M+H] + = 200.1/202.1.

第二步:将化合物69b(1.0g,5.0mmol)溶于DCM(30mL)中,加入甲基硫代乙酸(0.4mL,5.0mmol),降温至0℃依次滴加吡啶(4.0mL,50.0mmol),POCl3(2.3mL,25mmol),加完后升至25℃下搅拌1h。冰水浴下将反应液缓慢倒入饱和碳酸氢钠溶液(100mL)中淬灭反应,DCM萃取5*30mL,硫酸钠干燥,减压浓缩得到化合物69c(1.4g,crude)。LC-MS:ESI[M+H]+=290.2/292.2。Step 2: Compound 69b (1.0 g, 5.0 mmol) was dissolved in DCM (30 mL), methylthioacetic acid (0.4 mL, 5.0 mmol) was added, the temperature was lowered to 0°C, pyridine (4.0 mL, 50.0 mmol) and POCl 3 (2.3 mL, 25 mmol) were added dropwise in sequence, and the temperature was raised to 25°C and stirred for 1 h. The reaction solution was slowly poured into a saturated sodium bicarbonate solution (100 mL) under an ice-water bath to quench the reaction, and DCM was used for extraction (5*30 mL), dried over sodium sulfate, and concentrated under reduced pressure to obtain compound 69c (1.4 g, crude). LC-MS: ESI [M+H] + = 290.2/292.2.

第三步:将化合物69c(1.4g,5.0mmol)溶于MeOH(20mL)中,加入MeONa(0.8g,15.0mmol),25℃下搅拌21h。将反应液浓缩后加入饱和氯化铵溶液(20mL)淬灭,DCM萃取4*30mL,硫酸钠干燥,减压浓缩后柱层析(EA/PE=1/1)纯化得到化合物69d(0.7g,2.57mmol,51%)。LC-MS:ESI[M+H]+=270.2/272.1。Step 3: Compound 69c (1.4 g, 5.0 mmol) was dissolved in MeOH (20 mL), MeONa (0.8 g, 15.0 mmol) was added, and the mixture was stirred at 25°C for 21 h. The reaction solution was concentrated and quenched by adding saturated ammonium chloride solution (20 mL), extracted with DCM 4*30 mL, dried with sodium sulfate, concentrated under reduced pressure, and purified by column chromatography (EA/PE=1/1) to obtain compound 69d (0.7 g, 2.57 mmol, 51%). LC-MS: ESI[M+H] + =270.2/272.1.

第四步:将化合物69d(0.7g,2.57mmol)溶于dioxane(30mL)中,加入三丁基锡 甲醇(1.0mL,3.11mmol),XPHOS-Pd-G2(0.2g,0.26mmol),N2氛围下于100℃搅拌12h。反应完全后垫硅藻土过滤,滤液旋干后Prep-TLC纯化得到化合物69e(0.17g,0.77mmol,30%)。LC-MS:ESI[M+H]+=222.2。Step 4: Dissolve compound 69d (0.7 g, 2.57 mmol) in dioxane (30 mL) and add tributyltin Methanol (1.0 mL, 3.11 mmol), XPHOS-Pd-G 2 (0.2 g, 0.26 mmol), stirred at 100° C. for 12 h under N 2 atmosphere. After the reaction was complete, the mixture was filtered through celite pad, and the filtrate was dried by spin drying and purified by Prep-TLC to obtain compound 69e (0.17 g, 0.77 mmol, 30%). LC-MS: ESI [M+H] + = 222.2.

第五步:将化合物69e(0.17g,0.77mmol)溶于甲苯(10mL)中,加入DMF(0.01mL),25℃下滴加SOCl2(0.06mL,0.85mmol),25℃搅拌1h。减压浓缩得到化合物69f(0.2g,crude)。LC-MS:ESI[M+H]+=240.2。Step 5: Compound 69e (0.17 g, 0.77 mmol) was dissolved in toluene (10 mL), DMF (0.01 mL) was added, SOCl 2 (0.06 mL, 0.85 mmol) was added dropwise at 25°C, and the mixture was stirred for 1 h at 25°C. The mixture was concentrated under reduced pressure to obtain compound 69f (0.2 g, crude). LC-MS: ESI [M+H] + = 240.2.

第七步:将化合物69f(0.1g,0.42mmol)溶于乙腈(10mL)中,加入DIPEA(0.3mL,1.68mmol),KI(0.02g),化合物int-3(0.1g,0.42mmol)在80℃搅拌2h。减压浓缩除去溶剂,残留物Pre-HPLC纯化得到白色固体化合物69(0.03g,0.07mmol,收率为16%)。LC-MS:ESI[M+H]+=439.2。1H NMR(400MHz,DMSO)δ11.96(s,1H),8.67-8.55(m,1H),8.08(dd,J=9.9,7.8Hz,1H),7.92(dd,J=7.7,1.7Hz,1H),7.65-7.49(m,2H),7.30(d,J=16.6Hz,1H),7.17(dd,J=8.1,1.2Hz,1H),6.26(s,1H),3.66(s,2H),3.14(d,J=2.8Hz,2H),2.79(d,J=4.8Hz,3H),2.67(t,J=5.5Hz,2H),2.51(d,J=1.7Hz,2H),2.41(s,3H)。Step 7: Compound 69f (0.1 g, 0.42 mmol) was dissolved in acetonitrile (10 mL), and DIPEA (0.3 mL, 1.68 mmol), KI (0.02 g), and compound int-3 (0.1 g, 0.42 mmol) were added and stirred at 80°C for 2 h. The solvent was removed by concentration under reduced pressure, and the residue was purified by Pre-HPLC to obtain white solid compound 69 (0.03 g, 0.07 mmol, yield 16%). LC-MS: ESI [M+H] + = 439.2. 1H NMR (400MHz, DMSO) δ11.96 (s, 1H), 8.67-8.55 (m, 1H), 8.08 (dd, J = 9.9, 7.8Hz, 1H),7.92(dd,J=7.7,1.7Hz,1H),7.65-7.49(m,2H),7.30(d,J=16.6Hz,1H),7. 17(dd,J=8.1,1.2Hz,1H),6.26(s,1H),3.66(s,2H),3.14(d,J=2.8Hz,2H),2. 79 (d, J = 4.8 Hz, 3H), 2.67 (t, J = 5.5 Hz, 2H), 2.51 (d, J = 1.7 Hz, 2H), 2.41 (s, 3H).

实施例70-实施例74的化合物70~74的制备方法参考实施例69。The preparation methods of compounds 70-74 of Examples 70-74 can be referred to Example 69.

表12化合物70~74


Table 12 Compounds 70-74


实施例75:N-甲基-5-(4-((7-(甲基-d3)硫代)-6-氧代-5,6-二氢-1,5-萘并吡啶-3-基)甲基)哌嗪-1-基)吡啶甲酰胺
Example 75: N-methyl-5-(4-((7-(methyl-d 3 )thio)-6-oxo-5,6-dihydro-1,5-naphthopyridin-3-yl)methyl)piperazin-1-yl)picolinamide

N-甲基-5-(4-((7-(甲基-d3)硫代)-6-氧代-5,6-二氢-1,5-萘并吡啶-3-基)甲基)哌嗪-1-基)吡啶甲酰胺的制备参考实施例59,得到化合物75(0.04g,0.09mmol,收率为23%)。LC-MS:ESI[M+H]+=428.5。1H NMR(400MHz,CDCl3)δ11.49(s,1H),8.53(s,1H),8.16(d,J=2.4Hz,1H),8.05(d,J=8.7Hz,1H),7.78(d,J=4.5Hz,1H),7.72(s,1H),7.64(s,1H),7.22(dd,J=8.8,2.7Hz,1H),3.71(s,2H),3.36(d,J=4.6Hz,4H),3.01(d,J=5.0Hz,3H),2.68(d,J=4.5Hz,4H)。Preparation of N-methyl-5-(4-((7-(methyl-d 3 )thio)-6-oxo-5,6-dihydro-1,5-naphthopyridin-3-yl)methyl)piperazin-1-yl)picolinamide Referring to Example 59, Compound 75 (0.04 g, 0.09 mmol, yield 23%) was obtained. LC-MS: ESI [M+H] + = 428.5. 1 H NMR (400MHz, CDCl 3 )δ11.49(s,1H),8.53(s,1H),8.16(d,J=2.4Hz,1H),8.05(d,J=8.7Hz,1H),7.78(d,J=4.5Hz,1H),7.72(s,1H),7.64( s, 1H), 7.22 (dd, J = 8.8, 2.7Hz, 1H), 3.71 (s, 2H), 3.36 (d, J = 4.6Hz, 4H), 3.01 (d, J = 5.0Hz, 3H), 2.68 (d, J = 4.5Hz, 4H).

实施例76:2-氯-1'-((8-氟-3-(甲硫基)-2-氧代-1,2-二氢喹啉-7-基)甲基)-N-甲基-1',2',3',6'-四氢-[3,4-联吡啶]-6-甲酰胺
Example 76: 2-Chloro-1'-((8-fluoro-3-(methylthio)-2-oxo-1,2-dihydroquinolin-7-yl)methyl)-N-methyl-1',2',3',6'-tetrahydro-[3,4-bipyridine]-6-carboxamide

第一步:将化合物76a(10g,42.731mmol)溶于THF(200mL),加入BF3 .Et2O(18.19g,128.194mmol),0℃下加入NaBH4(4.85g,128.194mmol),于0℃下搅拌2h。将反应液缓慢倒入100mL饱和氯化铵溶液中淬灭反应。减压浓缩,EA萃取5*50mL。无水硫酸钠干燥,减压浓缩,得到化合物76b粗产品(10.0g),LC-MS: ESI[M+H]+=220.2/222.2。Step 1: Dissolve compound 76a (10 g, 42.731 mmol) in THF (200 mL), add BF 3 . Et 2 O (18.19 g, 128.194 mmol), add NaBH 4 (4.85 g, 128.194 mmol) at 0°C, and stir at 0°C for 2 h. Slowly pour the reaction solution into 100 mL of saturated ammonium chloride solution to quench the reaction. Concentrate under reduced pressure, extract with EA 5*50 mL. Dry over anhydrous sodium sulfate, and concentrate under reduced pressure to obtain a crude product of compound 76b (10.0 g), LC-MS: ESI [M+H] + = 220.2/222.2.

第二步:将化合物76b(10g,45.446mmol)溶于DCM(200mL)中,加入MnO2(19.76g,227.231mmol),25℃下搅3h。垫硅藻土过滤,收集滤液,减压浓缩后得到化合物76c(7.0g,crude)。LC-MS:ESI[M+H]+=217.2/219.2。Step 2: Compound 76b (10 g, 45.446 mmol) was dissolved in DCM (200 mL), MnO 2 (19.76 g, 227.231 mmol) was added, and the mixture was stirred at 25° C. for 3 h. The mixture was filtered through Celite pad, and the filtrate was collected and concentrated under reduced pressure to obtain compound 76c (7.0 g, crude). LC-MS: ESI [M+H] + = 217.2/219.2.

第三步:将化合物76c(5g,22.933mmol)溶于THF(150mL)中,2-甲硫基乙酸乙酯(3.08g,22.933mmol),N2置换,-78℃下滴加LiHMDS(68.798mL,68.798mmol),于-78℃下搅拌1h。冰水浴下将反应液缓慢倒入饱和氯化铵溶液(100mL)中淬灭反应,EA萃取5*30mL,硫酸钠干燥,减压浓缩,柱层析分离(EA/PE=1/3)得到化合物76d(7.0g,crude)。LC-MS:ESI[M+H]+=288.2/290.2。Step 3: Compound 76c (5 g, 22.933 mmol) was dissolved in THF (150 mL), ethyl 2-methylthioacetate (3.08 g, 22.933 mmol), N 2 was replaced, LiHMDS (68.798 mL, 68.798 mmol) was added dropwise at -78°C, and stirred at -78°C for 1 h. The reaction solution was slowly poured into a saturated ammonium chloride solution (100 mL) under an ice-water bath to quench the reaction, extracted with EA 5*30 mL, dried with sodium sulfate, concentrated under reduced pressure, and separated by column chromatography (EA/PE=1/3) to obtain compound 76d (7.0 g, crude). LC-MS: ESI[M+H] + =288.2/290.2.

第四步:将化合物76d(2g,6.941mmol)溶于dioxane(30mL)中,加入三丁基锡甲醇(2.23g,6.941mmol),XPHOS-Pd-G2(0.55g,0.694mmol),N2氛围下,于100℃下搅拌12h。Prep-TLC纯化得到化合物76e(1.35g,5.64mmol,81%)。LC-MS:ESI[M+H]+=240.2。Step 4: Compound 76d (2 g, 6.941 mmol) was dissolved in dioxane (30 mL), tributyltin methanol (2.23 g, 6.941 mmol) and XPHOS-Pd-G 2 (0.55 g, 0.694 mmol) were added, and stirred at 100° C. for 12 h under N 2 atmosphere. Prep-TLC purification gave compound 76e (1.35 g, 5.64 mmol, 81%). LC-MS: ESI [M+H] + = 240.2.

第五步:将化合物76e(1.35g,5.64mmol)溶于甲苯(30mL)中,加入DMF(0.02mL),25℃下滴加SOCl2(0.81g,6.771mmol),25℃搅1h。减压浓缩得到化合物76f(1.3g,crude)。LC-MS:ESI[M+H]+=258.2。Step 5: Compound 76e (1.35 g, 5.64 mmol) was dissolved in toluene (30 mL), DMF (0.02 mL) was added, SOCl 2 (0.81 g, 6.771 mmol) was added dropwise at 25°C, and stirred at 25°C for 1 h. The mixture was concentrated under reduced pressure to obtain compound 76f (1.3 g, crude). LC-MS: ESI [M+H] + = 258.2.

第六步:将化合物76f(0.045g,0.179mmol)和化合物int-29(0.05g,0.179mmol)溶于乙腈(20mL)中,加入DIEA(0.12g,0.894mmol),碘化钾(0.03g,0.179mmol),于80℃下搅拌2h。减压浓缩后反相制备,得到化合物76(45mg,0.095mmol,53.22%)。LC-MS:ESI[M+H]+=473.2。1H NMR(400MHz,DMSO)δ12.08(s,1H),8.60(q,J=4.5Hz,1H),7.95(d,J=7.8Hz,1H),7.88(d,J=7.7Hz,1H),7.61(s,1H),7.46(d,J=8.1Hz,1H),7.28–7.20(m,1H),5.84(s,1H),3.70(d,J=43.0Hz,2H),3.13(s,3H),2.80(d,J=4.8Hz,3H),2.70(s,2H),2.42(s,3H)。Step 6: Compound 76f (0.045 g, 0.179 mmol) and compound int-29 (0.05 g, 0.179 mmol) were dissolved in acetonitrile (20 mL), DIEA (0.12 g, 0.894 mmol) and potassium iodide (0.03 g, 0.179 mmol) were added, and stirred at 80°C for 2 h. After concentration under reduced pressure, the mixture was prepared by reverse phase to obtain compound 76 (45 mg, 0.095 mmol, 53.22%). LC-MS: ESI [M+H] + = 473.2. 1H NMR (400MHz, DMSO) δ12.08(s,1H),8.60(q,J=4.5Hz,1H),7.95(d,J=7.8Hz,1H),7.88(d,J=7.7Hz,1H),7.61(s,1H),7.46(d,J=8 .1Hz,1H),7.28–7.20(m,1H),5.84(s,1H),3.70(d,J=43.0Hz,2H),3.13(s,3H),2.80(d,J=4.8Hz,3H),2.70(s,2H),2.42(s,3H).

实施例77-实施例90、实施例96、98~99和实施例100~107的化合物77~90、96、98~99和100~107的制备方法参考实施例76。The preparation methods of compounds 77-90, 96, 98-99 and 100-107 of Examples 77-90, 96, 98-99 and 100-107 can be referred to Example 76.

表13化合物77~90、96、98~99和100~107








Table 13 Compounds 77-90, 96, 98-99 and 100-107








实施例91:5-(4-((8-氟-3-(甲硫基)-2-氧代-1,2-二氢-1,6-萘啶-7-基)甲基)哌嗪-1-基)-N,6-二甲基吡啶酰胺
Example 91: 5-(4-((8-fluoro-3-(methylthio)-2-oxo-1,2-dihydro-1,6-naphthyridin-7-yl)methyl)piperazin-1-yl)-N,6-dimethylpicolinamide

第一步:将化合物91a(2g,12.774mmol)溶于DMF(20mL)和ACN(20mL),加入selectfluor(5.43g,15.329mmol),N2置换3次,于80℃下搅拌2h。反应完成后,加入饱和碳酸氢钠溶液(30mL)淬灭反应。EA萃取5*30mL,无水硫酸钠干燥,减压浓缩,柱层析(EA/PE=1/3)分离纯化得到化合物91b(0.9g,5.15mmol,40.3%),LC-MS:ESI[M+H]+=175.2。Step 1: Compound 91a (2 g, 12.774 mmol) was dissolved in DMF (20 mL) and ACN (20 mL), selectfluor (5.43 g, 15.329 mmol) was added, N2 was replaced 3 times, and the mixture was stirred at 80°C for 2 h. After the reaction was completed, saturated sodium bicarbonate solution (30 mL) was added to quench the reaction. EA was extracted with 5*30 mL, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and separated and purified by column chromatography (EA/PE=1/3) to obtain compound 91b (0.9 g, 5.15 mmol, 40.3%), LC-MS: ESI[M+H] + =175.2.

第二步:将化合物91b(0.9g,5.15mmol)溶于THF(30mL)中,化合物2-甲硫基乙酸乙酯(0.83g,6.18mmol),N2置换,-78℃下滴加LiHMDS(25.7mL,25.7mmol),于-78℃下搅拌1h。后升至25℃搅拌12h。冰水浴下将反应液缓慢倒入饱和氯化铵溶液(100mL)中淬灭反应,过滤收集滤饼,为目标化合物91c(2.8g,crude)。LC-MS:ESI[M+H]+=245.2。Step 2: Dissolve compound 91b (0.9 g, 5.15 mmol) in THF (30 mL), compound ethyl 2-methylthioacetate (0.83 g, 6.18 mmol), replace with N 2 , add LiHMDS (25.7 mL, 25.7 mmol) dropwise at -78 °C, and stir at -78 °C for 1 h. Then raise the temperature to 25 °C and stir for 12 h. Slowly pour the reaction solution into saturated ammonium chloride solution (100 mL) under ice-water bath to quench the reaction, and collect the filter cake by filtration to obtain the target compound 91c (2.8 g, crude). LC-MS: ESI [M+H] + = 245.2.

第三步:将化合物91c(2.3g,9.4mmol)溶于dioxane(50mL)中,加入三丁基锡甲醇(3.32g,10.34mmol),XPHOS-Pd-G2(0.74g,0.94mmol),N2氛围下,于100℃下搅拌12h。Prep-TLC纯化得到化合物91d(0.8g,0.94mmol,35.2%)。LC-MS:ESI[M+H]+=241.2。Step 3: Compound 91c (2.3 g, 9.4 mmol) was dissolved in dioxane (50 mL), tributyltin methanol (3.32 g, 10.34 mmol) and XPHOS-Pd-G 2 (0.74 g, 0.94 mmol) were added, and stirred at 100° C. for 12 h under N 2 atmosphere. Prep-TLC purification gave compound 91d (0.8 g, 0.94 mmol, 35.2%). LC-MS: ESI [M+H] + = 241.2.

第四步:将化合物91d(0.8g,3.33mmol)溶于甲苯(30mL)中,加入DMF(0.05mL),25℃下滴加SOCl2(0.40g,3.33mmol),100℃搅拌4h。减压浓缩Prep-TLC纯化得到化合物91e(0.35g,1.35mmol,40.6%)。LC-MS:ESI[M+H]+=259.2。Step 4: Compound 91d (0.8 g, 3.33 mmol) was dissolved in toluene (30 mL), DMF (0.05 mL) was added, SOCl 2 (0.40 g, 3.33 mmol) was added dropwise at 25°C, and stirred at 100°C for 4 h. The mixture was concentrated under reduced pressure and purified by Prep-TLC to obtain compound 91e (0.35 g, 1.35 mmol, 40.6%). LC-MS: ESI [M+H] + = 259.2.

第五步:将化合物91e(30mg,0.116mmol)和化合物int-11(27.63mg,0.116mmol)溶于乙腈(20mL)中,加入DIEA(44.97mg,0.348mmol),碘化钾(38.50mg,0.232mmol),于80℃下搅拌2h。减压浓缩后反相制备,得到化合物91(45mg,0.095mmol,53.22%)。LC-MS:ESI[M+H]+=457.2。1H NMR(400MHz,DMSO)δ12.57(s,1H),8.65(s,1H),8.41(q,J=4.7Hz,1H),7.83–7.66(m,2H),7.45(d,J=8.3Hz,1H),3.79(s,2H),2.91(s,4H),2.79(d,J=4.9Hz,3H),2.70–2.60(m,4H),2.47(s,3H),2.45(s,3H)。 Step 5: Compound 91e (30 mg, 0.116 mmol) and compound int-11 (27.63 mg, 0.116 mmol) were dissolved in acetonitrile (20 mL), DIEA (44.97 mg, 0.348 mmol) and potassium iodide (38.50 mg, 0.232 mmol) were added, and stirred at 80°C for 2 h. After concentration under reduced pressure, the mixture was prepared by reverse phase to obtain compound 91 (45 mg, 0.095 mmol, 53.22%). LC-MS: ESI [M+H] + = 457.2. 1H NMR (400MHz, DMSO) δ12.57(s,1H),8.65(s,1H),8.41(q,J=4.7Hz,1H),7.83–7.66(m,2H),7.45(d,J=8. 3Hz,1H),3.79(s,2H),2.91(s,4H),2.79(d,J=4.9Hz,3H),2.70–2.60(m,4H),2.47(s,3H),2.45(s,3H).

实施例92-实施例95的化合物92~95、实施例97的化合物97和实施例108的化合物108的制备方法参考实施例91。The preparation methods of compounds 92-95 of Examples 92-95, compound 97 of Example 97 and compound 108 of Example 108 were prepared with reference to Example 91.

表14化合物92~95、97和108

Table 14 Compounds 92-95, 97 and 108

生物活性测试:Biological activity test:

1、PARP-1酶测定1. PARP-1 enzyme assay

实验材料:PARP1蛋白(BPS,Cat.No.80501),PARP2蛋白(BPS,Cat.No.80502),PARP5A蛋白(BPS,Cat.No.80504),Biotin-NAD+(R&D,Cat.No.6573),Strep-HRP (Thermo Pierce,Cat.No.21127),NAD+(TCI,Cat.No.D0919-5G),定量增强化学荧光HRP底物试剂盒(Thermo Pierce,Cat.No.15159),组蛋白(Active Motif,Cat.No.81167),Activited DNA(Genscript,Cat.No.L05182-01&02&03),anti-rabbit IgG,HRP-linked Antibody(CST,Cat.No.7074P2),anti-Poly/Mono-ADP Ribose(E6F6A)Rabbit mAb(CST,Cat.No.83732S),SuperSignal ELISA Femto Substrate(THERMO PIERCE,Cat.No.37074)。Experimental materials: PARP1 protein (BPS, Cat. No. 80501), PARP2 protein (BPS, Cat. No. 80502), PARP5A protein (BPS, Cat. No. 80504), Biotin-NAD+ (R&D, Cat. No. 6573), Strep-HRP (Thermo Pierce, Cat. No. 21127), NAD+ (TCI, Cat. No. D0919-5G), quantitative enhanced chemiluminescence HRP substrate kit (Thermo Pierce, Cat. No. 15159), histone (Active Motif, Cat. No. 81167), activated DNA (Genscript, Cat. No. L05182-01&02&03), anti-rabbit IgG, HRP-linked Antibody (CST, Cat. No. 7074P2), anti-Poly/Mono-ADP Ribose (E6F6A) Rabbit mAb (CST, Cat. No. 83732S), SuperSignal ELISA Femto Substrate (THERMO PIERCE, Cat. No. 37074).

1.1 PARP1酶测定1.1 PARP1 enzyme assay

1.1.1缓冲液的配制:PBST:1X PBS,0.05% Tween-20,封闭液:1X PBS,0.05%Tween-20,5% BSA,反应缓冲液:50mM Tris-HCl(pH7.5),0.005% Tween-20,0.01%BSA。1.1.1 Preparation of buffer: PBST: 1X PBS, 0.05% Tween-20, blocking solution: 1X PBS, 0.05% Tween-20, 5% BSA, reaction buffer: 50mM Tris-HCl (pH7.5), 0.005% Tween-20, 0.01% BSA.

1.1.2包被:用1xPBS配制50ng/mL的Histone包被液,转移25uL包被液至384孔反应板中,4℃包被过夜。1.1.2 Coating: Prepare 50 ng/mL Histone coating solution with 1xPBS, transfer 25uL of coating solution to a 384-well reaction plate, and coat overnight at 4°C.

1.1.3洗涤:包被结束后弃掉包被液,用PBST溶液进行洗涤,方法为转移50uLPBST至384孔反应板,静置5分钟,弃掉洗液,重新加满,重复洗涤3次,最后拍干反应板等待下一步封闭。1.1.3 Washing: After coating, discard the coating solution and wash with PBST solution. The method is to transfer 50uL PBST to a 384-well reaction plate, let it stand for 5 minutes, discard the washing solution, refill it, repeat the washing 3 times, and finally pat the reaction plate dry and wait for the next step of blocking.

1.1.4封闭:转移50uL封闭液至384孔反应板,静置1小时。1.1.4 Blocking: Transfer 50uL of blocking solution to a 384-well reaction plate and let stand for 1 hour.

洗涤:封闭结束后弃掉封闭液,用PBST溶液按照步骤2方法洗涤3次,最后拍干反应板。Washing: After blocking, discard the blocking solution and wash three times with PBST solution according to step 2, and finally pat the reaction plate dry.

1.1.5配制1000倍的化合物,转移1uL化合物至199uL反应缓冲液的96孔板中,混匀,将混匀后的化合物转移5uL至384孔反应板中。1.1.5 Prepare 1000 times the compound, transfer 1uL of the compound to 199uL of reaction buffer in a 96-well plate, mix well, and transfer 5uL of the mixed compound to a 384-well reaction plate.

1.1.6用反应缓冲液配制25/10倍PARP1-DNA溶液,转移10uLPARP1-DNA溶液至384孔反应板中,对于阴性对照孔,转移10uLDNA溶液,PARP1终浓度为0.02nM,DNA终浓度为0.8nM。1.1.6 Prepare 25/10 times PARP1-DNA solution with reaction buffer, transfer 10uL PARP1-DNA solution to the 384-well reaction plate. For the negative control well, transfer 10uL DNA solution. The final concentration of PARP1 is 0.02nM and the final concentration of DNA is 0.8nM.

1.1.7用反应缓冲液配制25/10倍NAD+溶液,转移10uLNAD+溶液至384孔反应板中,NAD+终浓度为3.5uM,室温孵育60分钟。1.1.7 Prepare 25/10 times NAD+ solution with reaction buffer, transfer 10uL NAD+ solution to a 384-well reaction plate, the final NAD+ concentration is 3.5uM, and incubate at room temperature for 60 minutes.

1.1.8用反应缓冲液配制25/10倍NAD+溶液,转移10uLNAD+溶液至384孔反应板中,NAD+终浓度为3.5uM,室温孵育60分钟。1.1.8 Prepare 25/10 times NAD+ solution with reaction buffer, transfer 10uL NAD+ solution to a 384-well reaction plate, the final NAD+ concentration is 3.5uM, and incubate at room temperature for 60 minutes.

1.1.9洗涤:反应结束后弃掉反应液,用PBST溶液按照步骤2方法洗涤3次,最后拍干反应板。1.1.9 Washing: After the reaction is completed, discard the reaction solution and wash three times with PBST solution according to step 2, and finally pat the reaction plate dry.

1.1.10用封闭液稀释2000倍一抗(anti-Poly/Mono-ADP Ribose Rabbit mAb),加入20uL一抗,室温孵育1.5小时。1.1.10 Dilute the primary antibody (anti-Poly/Mono-ADP Ribose Rabbit mAb) 2000 times with blocking solution, add 20uL primary antibody, and incubate at room temperature for 1.5 hours.

1.1.11洗涤:弃掉一抗,用PBST溶液按照步骤2方法洗涤3次,最后拍干反 应板。1.1.11 Washing: Discard the primary antibody and wash three times with PBST solution according to step 2. Finally, pat dry. Should board.

1.1.12用封闭液稀释2000倍二抗(anti-rabbit IgG,HRP-linked Antibody),加入20uL二抗,室温孵育1小时。1.1.12 Dilute the secondary antibody (anti-rabbit IgG, HRP-linked Antibody) 2000 times with blocking solution, add 20uL secondary antibody, and incubate at room temperature for 1 hour.

1.1.13洗涤:弃掉二抗,用PBST溶液按照步骤2方法洗涤3次,最后拍干反应板。1.1.13 Washing: Discard the secondary antibody and wash three times with PBST solution according to step 2, and finally pat the reaction plate dry.

1.1.14显色:1:1混合Femto-ECL Substrate A和Femto-ECLSubstrate B,转移25uL至384反应板中。1.1.14 Color development: Mix Femto-ECL Substrate A and Femto-ECL Substrate B in a 1:1 ratio and transfer 25uL to a 384 reaction plate.

1.1.15读数:用Envision读取化学发光数值RLU。1.1.15 Reading: Use Envision to read the chemiluminescence value RLU.

测试结果如下表15:The test results are shown in Table 15 below:

表15 PARP-1酶测试结果

Table 15 PARP-1 enzyme test results

结论:本发明化合物对PARP1具有显著的抑制作用。Conclusion: The compounds of the present invention have a significant inhibitory effect on PARP1.

2、细胞抗增殖活性测试:2. Cell anti-proliferation activity test:

BRCA突变细胞MDA-MB-436细胞采用DMEM培养基培养,含10%胎牛血清、100U/mL的青霉素、100μg/mL链霉素,置于37℃的5%饱和CO2孵箱中培养。当细胞生长至80%融合度时,收集细胞,300g离心10min,以1200个/孔铺96孔板。24h后,加入不同终浓度PARPi(0、0.01、0.1、1、10、100、1000nM),继续培养72h。对细胞进行换液处理(重新加入相同终浓度PARPi),继续培养96h。取出96孔板,采用CCK8法检测450nM波长下OD值,并统计细胞抑制率:抑制率%=1-(给药组平均OD值-Blank组平均OD值)/(Control组平均OD值-Blank组平均OD值)*100%。BRCA mutant MDA-MB-436 cells were cultured in DMEM medium containing 10% fetal bovine serum, 100U/mL penicillin, and 100μg/mL streptomycin, and cultured in a 5% saturated CO2 incubator at 37°C. When the cells grew to 80% confluence, the cells were collected, centrifuged at 300g for 10min, and plated on a 96-well plate at 1200 cells/well. After 24h, different final concentrations of PARPi (0, 0.01, 0.1, 1, 10, 100, 1000nM) were added and cultured for 72h. The cells were treated with a replacement medium (PARPi was added again at the same final concentration) and cultured for 96h. The 96-well plate was taken out, and the OD value at a wavelength of 450nM was detected by CCK8 method, and the cell inhibition rate was calculated: inhibition rate % = 1-(mean OD value of the drug group-mean OD value of the Blank group)/(mean OD value of the Control group-mean OD value of the Blank group)*100%.

BRCA野生型细胞DLD-1细胞采用RPMI-1640培养基培养,含10%胎牛血清、100U/mL的青霉素、100μg/mL链霉素,置于37℃的5%饱和CO2孵箱中培养。当细胞生长至80%融合度时,收集细胞,300g离心10min,以1000个/孔铺96孔板。24h后,加入不同终浓度PARPi(0、1、10μM),继续培养72h。对细胞进行换液处理(重新加入相同终浓度PARPi),继续培养96h。取出96孔板,采用CCK8法检测450nM波长下OD值,并统计细胞抑制率:BRCA wild-type cells DLD-1 cells were cultured in RPMI-1640 medium containing 10% fetal bovine serum, 100U/mL penicillin, and 100μg/mL streptomycin, and cultured in a 5% saturated CO2 incubator at 37°C. When the cells grew to 80% confluence, the cells were collected, centrifuged at 300g for 10min, and plated at 1000 cells/well in a 96-well plate. After 24h, different final concentrations of PARPi (0, 1, 10μM) were added and cultured for another 72h. The cells were treated with a change of medium (PARPi was re-added at the same final concentration) and cultured for another 96h. The 96-well plate was taken out, and the OD value at a wavelength of 450nM was detected by CCK8 method, and the cell inhibition rate was calculated:

抑制率%=1-(给药组平均OD值-Blank组平均OD值)/(Control组平均OD值-Blank组平均OD值)*100%。 Inhibition rate % = 1-(average OD value of the drug administration group-average OD value of the Blank group)/(average OD value of the Control group-average OD value of the Blank group)*100%.

测试结果如下表16:The test results are shown in Table 16 below:

表16细胞抗增殖活性测试结果

Table 16 Cell antiproliferative activity test results

结论:本发明化合物对BRCA突变MDA-MB-436细胞具有显著抑制用,对BRCA野生型DLD-1细胞无明显抑制作用,表明本发明化合物特异性抑制同源重组缺陷肿瘤细胞,且本发明如2、11、17、21、28、59等大量优选化合物,活性明显优于参考化合物AZD5305和AZD9574。Conclusion: The compounds of the present invention have significant inhibitory effect on BRCA mutant MDA-MB-436 cells, but have no significant inhibitory effect on BRCA wild-type DLD-1 cells, indicating that the compounds of the present invention specifically inhibit homologous recombination-deficient tumor cells, and a large number of preferred compounds of the present invention, such as 2, 11, 17, 21, 28, 59, etc., have significantly better activity than the reference compounds AZD5305 and AZD9574.

3、化合物对hERG钾离子通道的抑制实验3. Inhibition experiment of compounds on hERG potassium channel

细胞培养和处理:稳定表达hERG的CHO细胞培养于细胞培养瓶中,置于37℃,5%CO2的培养箱培养。待细胞密度生长至60~80%,吸走细胞培养液,用PBS洗一遍后加入Detachin消化。待消化完全后加入培养液中和,然后离心,吸走上清液,再加入培养液重悬,调节细胞密度为2~5×106/mL备用。Cell culture and treatment: CHO cells stably expressing hERG were cultured in a cell culture flask and placed in an incubator at 37°C and 5% CO 2. When the cell density grew to 60-80%, the cell culture medium was removed, and the cells were washed once with PBS and digested with Detachin. After complete digestion, the cells were neutralized with culture medium, centrifuged, the supernatant was removed, and the cells were resuspended with culture medium. The cell density was adjusted to 2-5×10 6 /mL for later use.

化合物准备:将化合物母液用100% DMSO进行稀释,即取10μL化合物母液加入到20μL DMSO中,3倍连续稀释至6个浓度。分别取4μL的6个浓度的化合物,加入到396μL细胞外液中,即100倍稀释得到6个中间浓度。再分别取80μL的6个中间浓度化合物,加入到320μL细胞外液中,即5倍稀释至需要测试的最终浓度。最高测试浓度为40μM,依次分别为40,13.33,4.44,1.48,0.49和0.16μM共6个浓度。最终测试浓度中的DMSO含量不超过0.2%,此浓度的DMSO对hERG钾通道没有影响。化合物准备由Bravo仪器完成整个稀释过程。Compound preparation: Dilute the compound stock solution with 100% DMSO, that is, take 10μL of the compound stock solution and add it to 20μL DMSO, and dilute it 3 times continuously to 6 concentrations. Take 4μL of the 6 concentrations of the compound respectively and add it to 396μL of extracellular fluid, that is, dilute it 100 times to get 6 intermediate concentrations. Then take 80μL of the 6 intermediate concentration compounds respectively and add them to 320μL of extracellular fluid, that is, dilute it 5 times to the final concentration to be tested. The highest test concentration is 40μM, and there are 6 concentrations of 40, 13.33, 4.44, 1.48, 0.49 and 0.16μM respectively. The DMSO content in the final test concentration does not exceed 0.2%, and this concentration of DMSO has no effect on the hERG potassium channel. The compound preparation is completed by the Bravo instrument throughout the dilution process.

电生理记录过程:单细胞高阻抗封接和全细胞模式形成过程全部由Qpatch仪器自动完成,在获得全细胞记录模式后,细胞钳制在-80毫伏,在给予一个5秒的+40毫伏去极化刺激前,先给予一个50毫秒的-50毫伏前置电压,然后复极化到-50毫伏维持5秒,再回到-80毫伏。每15秒施加此电压刺激,记录2分钟后给予细胞外液记录5分钟,然后开始给药过程,化合物浓度从最低测试浓度开始,每个测试浓度给予2.5分钟,连续给完所有浓度后,给予阳性对照化合物3μM Cisapride。每个浓度至少测试3个细胞(n≥3)。Electrophysiological recording process: The single-cell high-impedance sealing and whole-cell pattern formation process are all automatically completed by the Qpatch instrument. After obtaining the whole-cell recording mode, the cell is clamped at -80 mV. Before giving a 5-second +40 mV depolarizing stimulus, a 50-millisecond -50 mV pre-voltage is given, and then repolarizes to -50 mV for 5 seconds, and then returns to -80 mV. This voltage stimulus is applied every 15 seconds. After recording for 2 minutes, the extracellular solution is given for 5 minutes, and then the drug administration process begins. The compound concentration starts from the lowest test concentration, and each test concentration is given for 2.5 minutes. After all concentrations are given continuously, the positive control compound 3μM Cisapride is given. At least 3 cells (n≥3) are tested for each concentration.

数据处理:数据分析处理采用GraphPad Prism 5.0和Excel软件。化合物IC50使用GraphPad Prism 5软件通过以下方程拟合计算得出: Data processing: Data analysis and processing were performed using GraphPad Prism 5.0 and Excel software. The IC50 of the compound was calculated using GraphPad Prism 5 software by fitting the following equation:

Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))

其中,X为供试品检测浓度的Log值,Y为对应浓度下抑制百分率,Bottom和Top分别为最小和最大抑制百分率。Among them, X is the Log value of the test sample detection concentration, Y is the inhibition percentage at the corresponding concentration, and Bottom and Top are the minimum and maximum inhibition percentages, respectively.

表17化合物对hERG钾离子通道抑制结果
Table 17 Inhibition results of compounds on hERG potassium channels

结论:本发明化合物11、12、17、31、47、50、64、73、89和95等对hERG钾离子通道抑制作用较弱。Conclusion: Compounds 11, 12, 17, 31, 47, 50, 64, 73, 89 and 95 of the present invention have weak inhibitory effects on hERG potassium ion channels.

4、化合物体内脑血分布评价4. Evaluation of the brain and blood distribution of compounds in vivo

实验目的:获得化合物的脑血分布。Experimental purpose: To obtain the brain-blood distribution of the compound.

实验方案:通过监测化合物在小鼠脑和血浆中的含量,考察化合物的脑血分布。Experimental plan: The brain-blood distribution of the compound was investigated by monitoring the content of the compound in the mouse brain and plasma.

实验步骤:称量化合物,加少量DMSO,再入加注射用氯化钠溶液,配成1mg·mL-1的化合物溶液,待给药用。小鼠,雄鼠,按10 mg·kg-1口服给药,给药后1 h、6 h分别采集全血和全脑(n=3)。全血离心3500 rpm 15 min,收集上清血浆。称取离心管重量为M1,装入全脑的离心管重量为M2,加水匀浆后离心管重量为M3,取出30μL匀浆后离心管重量为M4。取30μL血浆和30μL脑匀浆于离心管中,加入120μL含20 ng·ml-1内标SAHA的乙腈沉淀,涡旋30s,于13000 rpm离心15 min,取上清装进样瓶待测。Experimental steps: Weigh the compound, add a small amount of DMSO, and then add sodium chloride solution for injection to make a 1mg·mL -1 compound solution for administration. Mice, male mice, were orally administered at 10 mg·kg -1. Whole blood and whole brain were collected 1 h and 6 h after administration (n=3). Whole blood was centrifuged at 3500 rpm for 15 min, and the supernatant plasma was collected. The weight of the centrifuge tube was weighed as M1, the weight of the centrifuge tube containing the whole brain was weighed as M2, the weight of the centrifuge tube after adding water homogenate was weighed as M3, and the weight of the centrifuge tube after taking out 30μL of homogenate was weighed as M4. Take 30μL of plasma and 30μL of brain homogenate in a centrifuge tube, add 120μL of acetonitrile containing 20 ng·ml -1 internal standard SAHA to precipitate, vortex for 30s, centrifuge at 13000 rpm for 15 min, and take the supernatant into the injection bottle for testing.

标准曲线范围:10~10000 ng·ml-1Standard curve range: 10~10000 ng·ml -1 .

脑中药物含量=实测值×0.03×(M3-M1)/[(M2-M1)×(M3-M4)]。Drug content in brain = measured value × 0.03 × (M3-M1)/[(M2-M1) × (M3-M4)].

化合物脑血分布结果如下表18所示:The results of the brain blood distribution of the compound are shown in Table 18 below:

表18化合物给药后小鼠脑血分布测试结果
Table 18 Results of the brain blood distribution test in mice after compound administration

结论:本发明化合物10、11、12、17、22、27、31、42、47、50和60等在1h和6h血浆浓度和脑组织浓度显著高于AZD9574。因此,化合物10、11、12、17、22、27、31、42、47、50和60等具有脑渗透的潜力。Conclusion: The plasma concentration and brain tissue concentration of compounds 10, 11, 12, 17, 22, 27, 31, 42, 47, 50 and 60 of the present invention are significantly higher than those of AZD9574 at 1h and 6h. Therefore, compounds 10, 11, 12, 17, 22, 27, 31, 42, 47, 50 and 60 have the potential for brain penetration.

5、化合物在Balb/c小鼠体内的药代动力学评价5. Pharmacokinetic evaluation of the compound in Balb/c mice

实验目的:了解化合物的药代动力学情况。Experimental purpose: To understand the pharmacokinetics of the compound.

实验依据:化学药物非临床药代动力学研究技术指导原则,2014年。Experimental basis: Technical guidelines for non-clinical pharmacokinetic studies of chemical drugs, 2014.

实验方案:通过Balb/c小鼠静脉给药(1mg·kg-1)和灌胃给药(1mg·kg-1),考察化合物的药代动力学情况。Experimental plan: The pharmacokinetics of the compound were investigated by intravenous administration (1 mg·kg-1) and oral administration (1 mg·kg-1) to Balb/c mice.

样品配制:称量0.2mg左右的化合物,加10μLDMSO溶解,再加注射用氯化钠溶液,配成0.1mg·mL-1的化合物溶液,待给药用。Sample preparation: weigh about 0.2 mg of the compound, add 10 μL DMSO to dissolve, and then add sodium chloride solution for injection to make a 0.1 mg·mL-1 compound solution for administration.

样品采集:6只Balb/c小鼠(成都达硕实验动物有限公司,许可证号:SCXK(川)2020-030),雄性,3只按1mg·kg-1静脉给药(IV)、3只按1mg·kg-1灌胃给药(PO),给药后5min、15min、30min、1h、2h、4h、6h、8h、10h、24h和48h采集约0.05mL血液,将收集的血液3500rpm化合物离心15min,收集上清血浆,-40℃冻存待测。以LC-MS/MS分析方法定量分析血药浓度,计算药代动力学参数,如达峰时间(Cmax),药时曲线下面积(AUC(0-t)),半衰期(T1/2),清除率(CL),组织分部(Vdss),生物 利用度(F)等。Sample collection: 6 male Balb/c mice (Chengdu Dashuo Experimental Animal Co., Ltd., license number: SCXK (Chuan) 2020-030), 3 mice were intravenously administered (IV) at 1 mg·kg-1, and 3 mice were gavaged (PO) at 1 mg·kg-1. About 0.05 mL of blood was collected at 5min, 15min, 30min, 1h, 2h, 4h, 6h, 8h, 10h, 24h and 48h after administration. The collected blood was centrifuged at 3500rpm for 15min, and the supernatant plasma was collected and frozen at -40℃ for testing. The blood drug concentration was quantitatively analyzed by LC-MS/MS analysis method, and pharmacokinetic parameters such as peak time (Cmax), area under the drug-time curve (AUC(0-t)), half-life (T 1/2 ), clearance (CL), tissue distribution (Vdss), biological Utilization (F), etc.

表19化合物在Balb/c小鼠体内的药代动力学测试结果
Table 19 Pharmacokinetic test results of compounds in Balb/c mice

结论:本发明化合物在Balb/c小鼠体内具有良好的药代动力学性质。Conclusion: The compounds of the present invention have good pharmacokinetic properties in Balb/c mice.

6、化合物47和64对乳腺癌MDA-MB-436裸小鼠皮下移植瘤模型的体内药效学研究6. In vivo pharmacodynamic study of compounds 47 and 64 on breast cancer MDA-MB-436 subcutaneous transplanted tumor model in nude mice

(1)主要仪器、器械(1) Main instruments and equipment

CO2细胞培养箱:Yamato品牌,型号IP610;生物安全柜:苏州安泰品牌,型号BSC-1304ⅡA2;常温离心机:Thermo Scientific品牌,型号SORVALLST 16;CO2 cell culture box: Yamato brand, model IP610; biological safety cabinet: Suzhou Antai brand, model BSC-1304ⅡA2; normal temperature centrifuge: Thermo Scientific brand, model SORVALLST 16;

数码倒置显微镜:Olympus品牌,型号CKX3-SLP;恒温水浴箱:上海跃进医疗器械有限公司,型号HSW-420;细胞计数板:上海求精生化试剂仪器有限公司,型号XB.K.25;液氮罐:Thermo品牌,型号CY50935-70;冰箱:青岛海尔品牌,型号BCD-601WDGX;医用低温冰箱:美国Thermo品牌,型号ULTS1651;Digital inverted microscope: Olympus brand, model CKX3-SLP; constant temperature water bath: Shanghai Yuejin Medical Instrument Co., Ltd., model HSW-420; cell counting plate: Shanghai Qiujing Biochemical Reagent Instrument Co., Ltd., model XB.K.25; liquid nitrogen tank: Thermo brand, model CY50935-70; refrigerator: Qingdao Haier brand, model BCD-601WDGX; medical low-temperature refrigerator: American Thermo brand, model ULTS1651;

纯水仪:美国Millipore品牌,型号F7PNO9748;立式高压灭菌锅:Yamato品牌,型号DKN812C;1mL一次性使用无菌注射器:上海康德莱企业发展集团股份有限公司;1mL一次性使用无菌胰岛素注射器:BD品牌;体重秤:上海衡平仪器仪表厂,型号JY2002;分析天枰:SARTORIUS品牌,型号BCE95I-1CEU;Water purifier: American Millipore brand, model F7PNO9748; vertical high pressure sterilizer: Yamato brand, model DKN812C; 1mL disposable sterile syringe: Shanghai Kangdeli Enterprise Development Group Co., Ltd.; 1mL disposable sterile insulin syringe: BD brand; weight scale: Shanghai Hengping Instrument Factory, model JY2002; analytical balance: SARTORIUS brand, model BCE95I-1CEU;

温湿度计:武强温湿表制造中心,量制冀字30260102;小型离心机:美国Thermo品牌,型号SORVALL LEGEND MICRO 17Centrifuge;迷你混合仪:Yeasen品牌,型号ES-VM25;金属浴:SCILOGEX品牌,型号SCL120-S。 Thermometer and hygrometer: Wuqiang Thermohygrometer Manufacturing Center, measurement and manufacture number: Jizi 30260102; small centrifuge: American Thermo brand, model: SORVALL LEGEND MICRO 17Centrifuge; mini mixer: Yeasen brand, model: ES-VM25; metal bath: SCILOGEX brand, model: SCL120-S.

(2)主要软件和数据处理系统(2) Main software and data processing systems

Graphpad Prism,版本6.0,Graphpad软件有限公司,Graphpad Prism软件用于整理、量化数据整理制作柱状统计图。Graphpad Prism, version 6.0, Graphpad Software Ltd., Graphpad Prism software is used to organize and quantify data and to create bar graphs.

(3)实验动物(3) Experimental animals

品系:NOD/SCID,等级:SPF级,来源:北京维通利华生物科技股份有限公司。Strain: NOD/SCID, Grade: SPF, Source: Beijing Weitonglihua Biotechnology Co., Ltd.

(4)造模方法(4) Modeling method

肿瘤细胞培养及准备:常规肿瘤细胞株的传代与培养,培养基中含有10%胎牛血清、100U/mL的青霉素、100μg/mL链霉素。当生长至80%融合度时,收集细胞,300g离心10min。预冷PBS洗涤3次,300g离心10min收集细胞,PBS重悬细胞,血球计数板进行细胞计数,调整细胞浓度,细胞收集完成后,细胞悬液置于冰上预冷。取100μL细胞悬液于小鼠背侧腋下皮下注射接种,在肿瘤体积达到200-300mm3时开始分组治疗。Tumor cell culture and preparation: For routine tumor cell line passage and culture, the culture medium contains 10% fetal bovine serum, 100U/mL penicillin, and 100μg/mL streptomycin. When the cells grow to 80% confluence, collect the cells and centrifuge at 300g for 10min. Wash with pre-cooled PBS three times, collect the cells by centrifugation at 300g for 10min, resuspend the cells in PBS, count the cells with a hemocytometer, adjust the cell concentration, and pre-cool the cell suspension on ice after cell collection. Take 100μL of the cell suspension and inject it subcutaneously in the dorsal armpit of the mouse. Start group treatment when the tumor volume reaches 200-300mm3 .

(5)观察及检测指标(5) Observation and testing indicators

一般状态观察:观察动物:所有计划观察的存活实验动物;观察时间:1天2次;观察内容:包括但不限于一般表现、行为状态、眼睛、口腔、鼻口部、耳、毛发及粪便、尿、生殖器等毒性症状,若出现异常,需进行详细描述。General status observation: Observation animals: All surviving experimental animals planned for observation; Observation time: Twice a day; Observation content: including but not limited to general manifestations, behavioral status, eyes, mouth, nose and mouth, ears, hair, feces, urine, genitals and other toxic symptoms. If any abnormalities are found, detailed descriptions are required.

(6)体重(6) Weight

测定动物:所有计划测定的存活实验动物;测定时间:造模前称量所有动物体重用于试验分组,造模后每2天称量1次小鼠体重,实时记录小鼠体重变化。有效判定标准:皮下移植瘤模型肿瘤体积(TV)、相对肿瘤体积(RTV)和相对肿瘤增殖率。每2天用游标卡尺测肿瘤的长径和短径,动态观察受试药抗肿瘤的效应。肿瘤体积(tumor volume,TV)的计算公式为:TV(mm3)=a×b2×0.5;其中a、b分别表示长径和短径。Animals to be measured: All surviving experimental animals planned for measurement; Measurement time: Weigh all animals before modeling for experimental grouping, weigh mice once every 2 days after modeling, and record the weight changes of mice in real time. Effective judgment criteria: Tumor volume (TV), relative tumor volume (RTV) and relative tumor proliferation rate of subcutaneous transplanted tumor model. Use a vernier caliper to measure the long and short diameters of the tumor every 2 days, and dynamically observe the anti-tumor effect of the test drug. The calculation formula of tumor volume (TV) is: TV (mm3) = a×b2×0.5; where a and b represent the long diameter and short diameter, respectively.

根据测量的结果计算出相对肿瘤体积(relative tumor volume,RTV),计算公式为:RTV=Vt/V0,其中V0为每一组在分笼给药时(即d0)测量所得TV,Vt为该组每一次测量时的TV。抗肿瘤活性的评价指标为相对肿瘤增值率T/C(%),计算公式如下:
T/C(%)=TRTV/CRTV×100%;
The relative tumor volume (RTV) was calculated based on the measurement results. The calculation formula was: RTV = Vt/V0, where V0 was the TV measured for each group at the time of cage administration (i.e., d0), and Vt was the TV for each measurement of the group. The evaluation index for antitumor activity was the relative tumor growth rate T/C (%), and the calculation formula was as follows:
T/C(%)=TRTV/CRTV×100%;

TRTV:治疗组相对肿瘤体积;CRTV:阴性对照组相对肿瘤体积。疗效评价标准:T/C(%)>60为无效;T/C(%)≤60,并经统计学分析P<0.05为有效。肿瘤重量测定及抑瘤率(%)的计算于治疗终点时处死动物,解剖剥离瘤块,称瘤重,并照相。按下式计算抑瘤率(%):TRTV: relative tumor volume of treatment group; CRTV: relative tumor volume of negative control group. Efficacy evaluation criteria: T/C (%)>60 is ineffective; T/C (%)≤60, and P<0.05 after statistical analysis is effective. Tumor weight determination and calculation of tumor inhibition rate (%) At the end of treatment, the animals were killed, the tumor was dissected and removed, the tumor weight was weighed, and photographed. The tumor inhibition rate (%) was calculated according to the following formula:

抑瘤率(肿瘤生长抑制率,%)=(阴性对照组平均瘤重(g)-给药组平均瘤重(g))/阴性对照组平均瘤重(g)×100%; Tumor inhibition rate (tumor growth inhibition rate, %) = (average tumor weight of negative control group (g) - average tumor weight of drug administration group (g)) / average tumor weight of negative control group (g) × 100%;

有效判定标准:抑瘤率(即肿瘤生长抑制率)<40%为无效;抑瘤率≥40%并经统计学处理P<0.05为有效。皮下瘤模型综合判定标准:上述2项有效判定标准(相对肿瘤增殖率和抑瘤率)中,有一项符合有效标准时判为有效。Effective judgment criteria: Tumor inhibition rate (i.e. tumor growth inhibition rate) < 40% is ineffective; tumor inhibition rate ≥ 40% and P < 0.05 after statistical analysis is effective. Subcutaneous tumor model comprehensive judgment criteria: It is judged to be effective when one of the above two effective judgment criteria (relative tumor proliferation rate and tumor inhibition rate) meets the effective criteria.

(7)数据采集和分析(7) Data collection and analysis

设施内的所有原始数据根据试验方案和研究机构的相关规范手动收集或用数据采集系统收集,手动收集的数据可以转录到Excel等软件中用来分析和报告。各组数据均为计量资料,表格中各组动物实验数据采用均值±标准差(Mean±SD)表示,图片中实验数据以均数±标准误(Mean±SEM.)描述,用EXCEL和graphpad prism软件进行t检验分析及生存分析。All raw data in the facility are collected manually or using a data acquisition system according to the experimental protocol and relevant regulations of the research institution. The manually collected data can be transcribed into software such as Excel for analysis and reporting. The data of each group are quantitative data. The experimental data of each group of animals in the table are expressed as mean ± standard deviation (Mean ± SD), and the experimental data in the picture are described as mean ± standard error (Mean ± SEM.). t-test analysis and survival analysis were performed using EXCEL and graphpad prism software.

实验过程:实验用雌性NOD/SCID小鼠30只,随机分为空白对照组、化合物47(0.3mg/kg)剂量组、化合物64(0.3mg/kg)剂量组、参考化合物AZD5305 0.3mg/kg和参考化合物Olaparib 100mg/kg剂量组,共5组,每组6只,均采用口服给药,1天1次。同时每2天称量体重并用游标卡尺测量肿瘤的长度和宽度,给药30天后将荷瘤小鼠麻醉处死,剥离肿瘤组织,称重并拍照,计算肿瘤抑制率。Experimental process: 30 female NOD/SCID mice were randomly divided into blank control group, compound 47 (0.3 mg/kg) dose group, compound 64 (0.3 mg/kg) dose group, reference compound AZD5305 0.3 mg/kg and reference compound Olaparib 100 mg/kg dose group, a total of 5 groups, 6 mice in each group, all were orally administered once a day. At the same time, the body weight was weighed every 2 days and the length and width of the tumor were measured with a vernier caliper. After 30 days of administration, the tumor-bearing mice were anesthetized and killed, the tumor tissue was peeled off, weighed and photographed, and the tumor inhibition rate was calculated.

结论:实验结果如图1和图2所示。细胞接种后,各给药组连续给药30天,与空白对照组比较,各给药组(AZD5305 0.3mg/kg、化合物47 0.3mg/kg、化合物64 0.3mg/kg、Olaparib 100mg/kg)对人乳腺癌细胞MM436细胞系NOD/SCID小鼠皮下瘤模型肿瘤生长有明显抑制效果。化合物47 0.3mg/kg剂量组和化合物64 0.3mg/kg剂量组的抗肿瘤效果显著优于参考化合物Olaparib 100mg/kg剂量组,与AZD5305 0.3mg/kg剂量组效果相当;另外,给药30天后,化合物64 0.3mg/kg和AZD5305 0.3mg/kg分别有1只小鼠肿瘤完全消退,显示了优异的抗肿瘤疗效。Conclusion: The experimental results are shown in Figures 1 and 2. After cell inoculation, each drug-treated group was continuously administered for 30 days. Compared with the blank control group, each drug-treated group (AZD5305 0.3 mg/kg, compound 47 0.3 mg/kg, compound 64 0.3 mg/kg, Olaparib 100 mg/kg) had a significant inhibitory effect on the growth of human breast cancer cell MM436 cell line NOD/SCID mouse subcutaneous tumor model. The anti-tumor effects of the compound 47 0.3 mg/kg dose group and the compound 64 0.3 mg/kg dose group were significantly better than the reference compound Olaparib 100 mg/kg dose group, and were equivalent to the AZD5305 0.3 mg/kg dose group; in addition, after 30 days of administration, the tumors of one mouse were completely regressed in the compound 64 0.3 mg/kg and AZD5305 0.3 mg/kg dose groups, respectively, showing excellent anti-tumor efficacy.

7、血液学毒性测定7. Hematological toxicity assay

CD34+造血干祖细胞(hmPB34-P-SC)在37℃、5% CO2的条件下于RPMI 1640完全培养基中培养过夜,培养基中添加了25ng/mL IL-3,25ng/mL IL-6,25ng/ml SCF和10%的胎牛血清。第二天,细胞重新悬浮,并以每孔1000细胞的浓度在96孔板中接种,并同时加入不同浓度药物处理,3个复孔。培养5天后,每孔中活细胞的数量使用CellTiter-Glo 2.0试剂盒测定确定,并通过酶标仪检测。CD34+ hematopoietic stem cells (hmPB34-P-SC) were cultured overnight in RPMI 1640 complete medium supplemented with 25ng/mL IL-3, 25ng/mL IL-6, 25ng/ml SCF and 10% fetal bovine serum at 37°C and 5% CO2. The next day, the cells were resuspended and seeded in 96-well plates at a concentration of 1000 cells per well, and different concentrations of drugs were added at the same time, with 3 replicates. After 5 days of culture, the number of live cells in each well was determined using the CellTiter-Glo 2.0 kit and detected by a microplate reader.

表20化合物对CD34+造血干祖细胞hmPB34-P-SC抑制活性测试结果

Table 20 Compounds' inhibitory activity test results on CD34+ hematopoietic stem cell hmPB34-P-SC

结论:本发明化合物对CD34+造血干祖细胞hmPB34-P-SC抑制作用较弱,尤其是化合物76、83、89和95等对hmPB34-P-SC抑制活性IC50值大于1000nM,显著优于阳性药物Olaparib。Conclusion: The compounds of the present invention have weak inhibitory effects on CD34+ hematopoietic stem progenitor cells hmPB34-P-SC, especially compounds 76, 83, 89 and 95, whose IC 50 values for hmPB34-P-SC inhibitory activity are greater than 1000nM, which are significantly better than the positive drug Olaparib.

8、化合物体内脑脊液与血浆分布评价8. Evaluation of the distribution of compounds in cerebrospinal fluid and plasma

实验步骤:称量化合物加少量DMSO,再入加注射用氯化钠溶液,配成5mg·mL-1的化合物溶液,待给药用。大鼠,雄鼠,按5mg·kg-1静脉给药,给药后0.25h、2h分别采集脑脊液和全血(n=1)。全血离心3500rpm 15min,收集上清血浆。取10μL血浆和10μL脑脊液于离心管中,加入40μL含20ng·ml-1内标SAHA的乙腈沉淀,涡旋30s,于13000rpm离心15min,取上清装进样瓶待测。标准曲线范围:1~1000ng·ml-1Experimental steps: Weigh the compound and add a small amount of DMSO, then add sodium chloride solution for injection to make a 5mg·mL -1 compound solution for administration. Male rats were intravenously administered at 5mg·kg -1. Cerebrospinal fluid and whole blood were collected at 0.25h and 2h after administration (n=1). Whole blood was centrifuged at 3500rpm for 15min, and the supernatant plasma was collected. 10μL plasma and 10μL cerebrospinal fluid were placed in a centrifuge tube, and 40μL acetonitrile containing 20ng·ml -1 internal standard SAHA was added for precipitation, vortexed for 30s, centrifuged at 13000rpm for 15min, and the supernatant was placed in a sample bottle for testing. Standard curve range: 1~1000ng·ml -1 .

表21化合物给药后大鼠脑积液与血浆分布测试结果
Table 21 Results of the test on the distribution of the compound in rat cerebrospinal fluid and plasma after administration

结论:本发明部分化合物具有较好的脑渗透潜力,尤其是化合物81、83、88、89、90、95、96和101等在大鼠脑脊液中具有较高的分布浓度。Conclusion: Some compounds of the present invention have good brain penetration potential, especially compounds 81, 83, 88, 89, 90, 95, 96 and 101, which have higher distribution concentrations in rat cerebrospinal fluid.

9、化合物11对乳腺癌MDA-MB-436裸小鼠皮下移植瘤模型的体内药效学研究9. In vivo pharmacodynamic study of compound 11 on breast cancer MDA-MB-436 subcutaneous transplanted tumor model in nude mice

实验过程:实验用雌性NOD/SCID小鼠18只,随机分为空白对照组、化合物AZD9574(3mg/kg)组和化合物11(1mg/kg)组,共3组,每组6只,均采用口服给药,1天1次。同时每2天称量体重并用游标卡尺测量肿瘤的长度和宽度,给药30天后将荷瘤小鼠麻醉处死,剥离肿瘤组织,称重并拍照,计算肿瘤抑制率。化合物AZD9574参照专利WO2021260092实施例20的合成方法合成获得。Experimental process: 18 female NOD/SCID mice were randomly divided into a blank control group, a compound AZD9574 (3 mg/kg) group, and a compound 11 (1 mg/kg) group, a total of 3 groups, 6 mice in each group, all of which were orally administered once a day. At the same time, the body weight was weighed every 2 days and the length and width of the tumor were measured with a vernier caliper. After 30 days of administration, the tumor-bearing mice were anesthetized and killed, the tumor tissue was peeled off, weighed and photographed, and the tumor inhibition rate was calculated. Compound AZD9574 was synthesized by referring to the synthesis method of Example 20 of patent WO2021260092.

结论:实验结果如图3所示。细胞接种后,各给药组连续给药30天,与空白对照 组比较,化合物AZD9574(3mg/kg)组和化合物11(1mg/kg)组对人乳腺癌MDA-MB-436细胞系NOD/SCID小鼠皮下瘤模型肿瘤生长有明显抑制效果,化合物11 1mg/kg组给药抗肿瘤效果优于AZD9574 3mg/kg组。Conclusion: The experimental results are shown in Figure 3. After cell inoculation, each drug group was given drug for 30 consecutive days, compared with the blank control. Compared with the control group, the compound AZD9574 (3 mg/kg) group and the compound 11 (1 mg/kg) group had a significant inhibitory effect on the tumor growth of the human breast cancer MDA-MB-436 cell line NOD/SCID mouse subcutaneous tumor model. The anti-tumor effect of the compound 11 1 mg/kg group was better than that of the AZD9574 3 mg/kg group.

10、化合物11对乳腺癌MDA-MB-436裸小鼠颅内模型的药效学研究10. Pharmacodynamic study of compound 11 on the intracranial model of breast cancer MDA-MB-436 nude mice

实验过程:MDA-MB-436-Luc细胞(购自于American Type Culture Collection公司)在DMEM培养基中培养,培养基中含有10%胎牛血清、100U/mL的青霉素、100μg/mL链霉素。当生长至80%融合度时,收集细胞,300g离心10min。预冷PBS洗涤3次,300g离心10min,PBS重悬细胞,血球计数板进行细胞计数,调整细胞浓度到2.5×107个/mL,细胞收集完成后,细胞悬液置于冰上预冷。取5μL细胞悬液于小鼠颅内(外侧2mm、前侧1mm、深3mm)注射接种,接种10天后通过活体成像结果开始分组治疗。实验用雌性NOD/SCID小鼠18只,随机分为空白对照组、化合物11 3mg/kg剂量组,参考化合物AZD9574 3mg/kg组,共3组,每组6只。各组均采用口服给药,1天1次,同时每2天1次称量体重,每7天1次测量小鼠脑部荧光强度,给药42天后停药观察,统计小鼠脑部荧光强度变化和小鼠生存率。Experimental process: MDA-MB-436-Luc cells (purchased from American Type Culture Collection) were cultured in DMEM medium containing 10% fetal bovine serum, 100U/mL penicillin, and 100μg/mL streptomycin. When grown to 80% confluence, the cells were collected and centrifuged at 300g for 10min. Washed 3 times with pre-cooled PBS, centrifuged at 300g for 10min, resuspended in PBS, counted with a hemocytometer, and the cell concentration was adjusted to 2.5×10 7 /mL. After cell collection, the cell suspension was pre-cooled on ice. Take 5μL of the cell suspension and inject it into the mouse's cranium (2mm outside, 1mm in front, 3mm deep). Ten days after inoculation, group treatment began based on the results of in vivo imaging. Eighteen female NOD/SCID mice were used in the experiment and randomly divided into a blank control group, a compound 11 3mg/kg dose group, and a reference compound AZD9574 3mg/kg group, for a total of 3 groups, with 6 mice in each group. Each group was orally administered once a day. The body weight was measured every 2 days and the fluorescence intensity of the mouse brain was measured every 7 days. The drug was stopped after 42 days of administration, and the changes in the fluorescence intensity of the mouse brain and the survival rate of the mice were counted.

结论:实验结果如图4~6所示。实验动物接种10天后进行实验分组,各给药组连续给药42天,与空白对照组比较,各给药组(AZD9574 3mg/kg、化合物11 3mg/kg)对人乳腺癌MDA-MB-436细胞系NOD/SCID小鼠颅内模型肿瘤生长有明显抑制效果,各给药组的荧光强度结果明显小于空白对照组,化合物11 3mg/kg组荧光强度弱于AZD9574 3mg/kg组,具有更强的颅内抗肿瘤效果;观察终点时,空白对照组、AZD95743mg/kg组、化合物11 3mg/kg组中位生存期分别为NA、37.5天、61.5天;观察终点时,化合物11的生存率明显优于参考化合物AZD9574,且化合物11的体重降低弱于参考化合物AZD9574,说明化合物11具有更优的颅内肿瘤治疗效果。 Conclusion: The experimental results are shown in Figures 4 to 6. The experimental animals were divided into experimental groups 10 days after inoculation, and each drug-treated group was continuously administered for 42 days. Compared with the blank control group, each drug-treated group (AZD9574 3 mg/kg, compound 11 3 mg/kg) had a significant inhibitory effect on the growth of intracranial tumors in NOD/SCID mice of human breast cancer MDA-MB-436 cell line. The fluorescence intensity results of each drug-treated group were significantly lower than that of the blank control group. The fluorescence intensity of the compound 11 3 mg/kg group was weaker than that of the AZD9574 3 mg/kg group, and it had a stronger intracranial anti-tumor effect. At the end of the observation, the median survival time of the blank control group, AZD9574 3 mg/kg group, and compound 11 3 mg/kg group was NA, 37.5 days, and 61.5 days, respectively. At the end of the observation, the survival rate of compound 11 was significantly better than that of the reference compound AZD9574, and the weight loss of compound 11 was weaker than that of the reference compound AZD9574, indicating that compound 11 has a better therapeutic effect on intracranial tumors.

Claims (29)

式Ⅰ的化合物或其药学上可接受的形式,其特征在于:所述式Ⅰ结构如下:
The compound of formula I or a pharmaceutically acceptable form thereof, characterized in that: the structure of formula I is as follows:
其中,in, X选自N或者C,且当X选自N时,表示单键,当X选自C时,表示双键;X is selected from N or C, and when X is selected from N, represents a single bond, when X is selected from C, represents a double bond; X1选自N或者C(R8a),X2选自N或者C(R8b),X3选自N或者C(R8c),且X1、X2和X3中至多有一个选自N; X1 is selected from N or C( R8a ), X2 is selected from N or C( R8b ), X3 is selected from N or C( R8c ), and at most one of X1 , X2 and X3 is selected from N; R1选自C1~8烷硫基、C1~8卤代烷硫基、C1~8氘代烷硫基、C1~8卤代烷氧基、C1~8卤代烷基或者C2~8烯基; R1 is selected from C1-8 alkylthio, C1-8 halogenated alkylthio, C1-8 deuterated alkylthio, C1-8 halogenated alkoxy, C1-8 halogenated alkyl or C2-8 alkenyl; R2选自氢、氟、氯、C1-4烷基、C1-4氟代烷基或者C1-4氘代烷基; R2 is selected from hydrogen, fluorine, chlorine, C1-4 alkyl, C1-4 fluoroalkyl or C1-4 deuterated alkyl; R3、R4独立地选自氢、氘、氟、C1-4烷基、C1-4氟代烷基或者C1-4氘代烷基;R 3 and R 4 are independently selected from hydrogen, deuterium, fluorine, C 1-4 alkyl, C 1-4 fluoroalkyl or C 1-4 deuterated alkyl; 或者,R3、R4与它们所连接的原子一起形成3-6元烷基环;Alternatively, R 3 , R 4 , together with the atoms to which they are attached, form a 3-6 membered alkyl ring; R5选自氢、氟、氯、氰基、C1-4烷基、C1-4氟代烷基或者C1-4氘代烷基;R 5 is selected from hydrogen, fluorine, chlorine, cyano, C 1-4 alkyl, C 1-4 fluoroalkyl or C 1-4 deuterated alkyl; 或者,R4、R5与它们所连接的原子一起形成5-7元烷基杂环;R4、R5与它们所连接的原子相连成环时,所述5-7元烷基杂环中,除主环上的X外,还含有1个O杂原子;Alternatively, R 4 , R 5 and the atoms to which they are connected together form a 5-7 membered alkyl heterocycle; when R 4 , R 5 and the atoms to which they are connected are connected to form a ring, the 5-7 membered alkyl heterocycle contains 1 O heteroatom in addition to X on the main ring; R3、R4与它们所连接的原子,和R4、R5与它们所连接的原子,两者不会同时成环;R 3 , R 4 and the atoms to which they are attached, and R 4 , R 5 and the atoms to which they are attached, will not form a ring at the same time; R6选自-C(O)-NH-R6a;R6a选自C1-4烷基、C1-4氟代烷基、C1-4氘代烷基或者3-6元环烷基;R 6 is selected from -C(O)-NH-R 6a ; R 6a is selected from C 1-4 alkyl, C 1-4 fluoroalkyl, C 1-4 deuterated alkyl or 3-6 membered cycloalkyl; R7选自氢、氟、氯、氰基、C1-4烷基、C1-4氟代烷基或者C1-4氘代烷基; R7 is selected from hydrogen, fluorine, chlorine, cyano, C1-4 alkyl, C1-4 fluoroalkyl or C1-4 deuterated alkyl; R8a、R8b、R8c独立地选自氢、氟、氯、C1-4烷基、C1-4氘代烷基、C1-4氟代烷基;R 8a , R 8b , and R 8c are independently selected from hydrogen, fluorine, chlorine, C 1-4 alkyl, C 1-4 deuterated alkyl, and C 1-4 fluoroalkyl; 所述药学上可接受的形式选自药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、氮氧化物、同位素标记物、代谢物或前药。The pharmaceutically acceptable form is selected from pharmaceutically acceptable salts, esters, stereoisomers, tautomers, polymorphs, solvates, N-oxides, isotopically labeled substances, metabolites or prodrugs.
根据权利要求1所述的化合物,其特征在于:The compound according to claim 1, characterized in that: X1选自N,X2选自C(R8b),X3选自C(R8c);或者X1选自C(R8a),X2选自N, X3选自C(R8c);或者X1选自C(R8a),X2选自者C(R8b),X3选自C(R8c)。 X1 is selected from N, X2 is selected from C( R8b ), and X3 is selected from C( R8c ); or X1 is selected from C( R8a ), X2 is selected from N, X3 is selected from C( R8c ); or X1 is selected from C( R8a ), X2 is selected from C( R8b ), and X3 is selected from C( R8c ). 根据权利要求1或2所述的化合物,其特征在于:The compound according to claim 1 or 2, characterized in that: R1选自C1~4烷硫基、C1~4卤代烷硫基、C1~4氘代烷硫基、C1~4卤代烷氧基、C1~4卤代烷基、C2~4烯基;R1中,所述卤代的卤选自F、Cl、Br;R 1 is selected from C 1-4 alkylthio, C 1-4 haloalkylthio, C 1-4 deuterated alkylthio, C 1-4 haloalkoxy, C 1-4 haloalkyl, C 2-4 alkenyl; in R 1 , the halogen is selected from F, Cl, Br; 优选的,R1选自甲硫基、卤代甲硫基、氘代甲硫基、卤代甲氧基、卤代甲基、卤代乙基或者乙烯基;R1中,所述卤代的卤选自F、Cl;Preferably, R 1 is selected from methylthio, halogenated methylthio, deuterated methylthio, halogenated methoxy, halogenated methyl, halogenated ethyl or vinyl; in R 1 , the halogenated halogen is selected from F and Cl; 更优选的,R1选自-S-CH3、-S-CF2H、-S-CF3、-S-CF2Cl、-S-CD3、-O-CF2H、-O-CF3、-O-CF2Cl、-CF2-CH3或者-CH=CH2More preferably, R 1 is selected from -S-CH 3 , -S-CF 2 H, -S-CF 3 , -S-CF 2 Cl, -S-CD 3 , -O-CF 2 H, -O-CF 3 , -O-CF 2 Cl, -CF 2 -CH 3 or -CH=CH 2 . 根据权利要求1~3任一项所述的化合物,其特征在于:至少满足下列的一项:The compound according to any one of claims 1 to 3, characterized in that at least one of the following is satisfied: R2选自氢、氟、氯、甲基、氟代甲基或者氘代甲基; R2 is selected from hydrogen, fluorine, chlorine, methyl, fluoromethyl or deuterated methyl; R3、R4独立地选自氢、氘、氟、甲基、氟代甲基或者氘代甲基;R 3 and R 4 are independently selected from hydrogen, deuterium, fluorine, methyl, fluoromethyl or deuterated methyl; R5选自氢、氟、氯、氰基、甲基、氟代甲基或者氘代甲基;R 5 is selected from hydrogen, fluorine, chlorine, cyano, methyl, fluoromethyl or deuterated methyl; R7选自氢、氟、氯、氰基、甲基、氟代甲基或者氘代甲基;R 7 is selected from hydrogen, fluorine, chlorine, cyano, methyl, fluoromethyl or deuterated methyl; R8a、R8a、R8c独立地选自氢、氟、氯、甲基、氟代甲基或者氘代甲基。R 8a , R 8a , R 8c are independently selected from hydrogen, fluorine, chlorine, methyl, fluoromethyl or deuterated methyl. 根据权利要求1~4任一项所述的化合物,其特征在于:The compound according to any one of claims 1 to 4, characterized in that: R6a选自甲基、氟代甲基、氘代甲基或者环丙基;R 6a is selected from methyl, fluoromethyl, deuterated methyl or cyclopropyl; 优选的,R6a选自甲基、-CF3、-CD3或者环丙基。Preferably, R 6a is selected from methyl, -CF 3 , -CD 3 or cyclopropyl. 根据权利要求1~5任一项所述的化合物,其特征在于:结构单元选自:

The compound according to any one of claims 1 to 5, characterized in that: Selected from:

根据权利要求1~5任一项所述的化合物,其特征在于:当R4、R5不与它们所连接的原子成环时,结构单元选自: The compound according to any one of claims 1 to 5, characterized in that: when R 4 and R 5 do not form a ring with the atoms to which they are connected, the structural unit Selected from: 根据权利要求1~7任一项所述的化合物,其特征在于:当R4、R5不与它们所连接的原子成环时,结构单元选自: The compound according to any one of claims 1 to 7, characterized in that when R 4 and R 5 do not form a ring with the atoms to which they are connected, the structural unit Selected from: 根据权利要求1~8任一项所述的化合物,其特征在于:当R4、R5与它们 所连接的原子成环时,结构单元选自: The compound according to any one of claims 1 to 8, characterized in that: when R 4 , R 5 and When the atoms connected form a ring, the structural unit Selected from: 优选的,当R4、R5与它们所连接的原子成环时,结构单元选自:
Preferably, when R 4 , R 5 and the atoms to which they are connected form a ring, the structural unit Selected from:
根据权利要求1~8任一项所述的化合物,其特征在于:将式Ⅰ中结构单元替换为 The compound according to any one of claims 1 to 8, characterized in that: the structural unit in formula I Replace with 根据权利要求1~10任一项所述的化合物,其特征在于:所述化合物选自:




The compound according to any one of claims 1 to 10, characterized in that the compound is selected from:




根据权利要求1或2所述的化合物,其特征在于:The compound according to claim 1 or 2, characterized in that: R1选自-S-CH3、-S-CF2H、-S-CF3、-S-CF2Cl、-S-CD3、-O-CF2H、-O-CF3、-O-CF2Cl、-CF2-CH3、-CH=CH2、-S-CFH2或者-O-CFH2R 1 is selected from -S-CH 3 , -S-CF 2 H, -S-CF 3 , -S-CF 2 Cl, -S-CD 3 , -O-CF 2 H, -O-CF 3 , -O-CF 2 Cl, -CF 2 -CH 3 , -CH=CH 2 , -S-CFH 2 or -O-CFH 2 . 根据权利要求1~2、4~5或12任一项所述的化合物,其特征在于:结构单元选自:

The compound according to any one of claims 1 to 2, 4 to 5 or 12, characterized in that: the structural unit Selected from:

根据权利要求1~2、4~5、7~8或12~13任一项所述的化合物,其特征在于:将式Ⅰ中结构单元替换为 The compound according to any one of claims 1 to 2, 4 to 5, 7 to 8 or 12 to 13, characterized in that: the structural unit in formula I Replace with 根据权利要求1~2、4~5、7~10或12~14任一项所述的化合物,其特征 在于:所述化合物选自:
The compound according to any one of claims 1 to 2, 4 to 5, 7 to 10 or 12 to 14, characterized in that In that: the compound is selected from:
根据权利要求1~2、4~5、7或12~13任一项所述的化合物,其特征在于:当R4、R5不与它们所连接的原子成环时,结构单元选自: The compound according to any one of claims 1 to 2, 4 to 5, 7 or 12 to 13, characterized in that when R 4 and R 5 do not form a ring with the atoms to which they are connected, the structural unit Selected from: 根据权利要求1~2、4~5、7或12~13或16任一项所述的化合物,其特征在于:当R4、R5与它们所连接的原子成环时,结构单元选自: The compound according to any one of claims 1 to 2, 4 to 5, 7 or 12 to 13 or 16, characterized in that when R 4 and R 5 form a ring with the atoms to which they are connected, the structural unit Selected from: 优选的,当R4、R5与它们所连接的原子成环时,结构单元选自:
Preferably, when R 4 , R 5 and the atoms to which they are connected form a ring, the structural unit Selected from:
根据权利要求1~2、4~5、7或12~13或16任一项所述的化合物,其特征在于:将式Ⅰ中结构单元替换为 The compound according to any one of claims 1 to 2, 4 to 5, 7 or 12 to 13 or 16, characterized in that: the structural unit in formula I Replace with 根据权利要求1~2、4~5、7、10、12~14或18任一项所述的化合物,其特征在于:将式Ⅰ中结构单元替换为 The compound according to any one of claims 1 to 2, 4 to 5, 7, 10, 12 to 14 or 18, characterized in that: the structural unit in formula I Replace with 根据权利要求1~2、4~5或12~13任一项所述的化合物,其特征在于:将式Ⅰ中结构单元替换为 R9选自H。The compound according to any one of claims 1 to 2, 4 to 5 or 12 to 13, characterized in that: the structural unit in formula I Replace with R9 is selected from H. 根据权利要求1~2、4~5、7、10、12~14或16~20任一项所述的化合物,其特征在于:所述化合物选自:

The compound according to any one of claims 1 to 2, 4 to 5, 7, 10, 12 to 14 or 16 to 20, characterized in that the compound is selected from:

药物组合物,其特征在于:其是以权利要求11、15或21任一项所述的 化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、氮氧化物、同位素标记物、代谢物或前药为活性成分,辅以药学上可接受的载体。The pharmaceutical composition is characterized in that: it is a pharmaceutical composition according to any one of claims 11, 15 or 21 The compound or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, nitrogen oxide, isotope label, metabolite or prodrug is an active ingredient, assisted by a pharmaceutically acceptable carrier. 权利要求11、15或21任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、氮氧化物、同位素标记物、代谢物或前药,及权利要求22所述的药物组合物,在制备用于预防和/或治疗PARP1酶相关疾病的药物中发挥用途。The compound according to any one of claims 11, 15 or 21, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, nitrogen oxide, isotope label, metabolite or prodrug thereof, and the pharmaceutical composition according to claim 22, are used in the preparation of a medicament for preventing and/or treating PARP1 enzyme-related diseases. 根据权利要求23所述的用途,其特征在于:所述PARP1酶相关疾病为肿瘤类病症。The use according to claim 23 is characterized in that the PARP1 enzyme-related disease is a tumor-related disease. 根据权利要求24所述的用途,其特征在于:所述肿瘤类病症缺乏HR依赖性DNA DSB修复途径。The use according to claim 24 is characterized in that the tumor-like disease lacks the HR-dependent DNA DSB repair pathway. 根据权利要求24或25所述的用途,其特征在于:所述肿瘤类病症包含一种或多种癌细胞,所述癌细胞相对于正常细胞具有降低的或缺失的通过HR修复DNA DSB的能力。The use according to claim 24 or 25 is characterized in that the tumor-like disease comprises one or more cancer cells, and the cancer cells have reduced or absent ability to repair DNA DSB through HR compared to normal cells. 根据权利要求26所述的用途,其特征在于:所述癌细胞具有BRCA1或BRCA2缺陷表型。The use according to claim 26, characterized in that the cancer cells have a BRCA1 or BRCA2 defective phenotype. 根据权利要求24~27任一项所述的用途,其特征在于:所述肿瘤类病症为乳腺癌、卵巢癌、原发性腹膜癌、胰腺癌、前列腺癌、血液癌、胃肠道癌、胶质母细胞瘤、肺癌、脑膜瘤、垂体瘤、颅咽管瘤、神经鞘瘤、胶质瘤、室管膜瘤、原始神经外胚层肿瘤、中枢神经系统淋巴瘤、生殖细胞肿瘤、转移瘤、脑癌或中枢神经系统癌。The use according to any one of claims 24 to 27 is characterized in that the tumor-related disease is breast cancer, ovarian cancer, primary peritoneal cancer, pancreatic cancer, prostate cancer, blood cancer, gastrointestinal cancer, glioblastoma, lung cancer, meningioma, pituitary tumor, craniopharyngioma, schwannoma, glioma, ependymoma, primitive neuroectodermal tumor, central nervous system lymphoma, germ cell tumor, metastasis, brain cancer or central nervous system cancer. 权利要求11、15或21任一项所述的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、氮氧化物、同位素标记物、代谢物或前药,及权利要求22所述的药物组合物,在制备PARP1抑制剂中的用途。 Use of the compound according to any one of claims 11, 15 or 21 or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, nitrogen oxide, isotope label, metabolite or prodrug, and the pharmaceutical composition according to claim 22 in the preparation of a PARP1 inhibitor.
PCT/CN2024/129351 2023-11-03 2024-11-01 Parp1-targeting compound and use thereof Pending WO2025092973A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN202311457218 2023-11-03
CN202311457218.5 2023-11-03
CN202410377779.2 2024-03-29
CN202410377779 2024-03-29
CN202411152648 2024-08-21
CN202411152648.0 2024-08-21

Publications (1)

Publication Number Publication Date
WO2025092973A1 true WO2025092973A1 (en) 2025-05-08

Family

ID=95582732

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2024/129351 Pending WO2025092973A1 (en) 2023-11-03 2024-11-01 Parp1-targeting compound and use thereof

Country Status (1)

Country Link
WO (1) WO2025092973A1 (en)

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114144413A (en) * 2019-07-19 2022-03-04 阿斯利康(瑞典)有限公司 PARP1 inhibitors
WO2022225934A1 (en) * 2021-04-19 2022-10-27 Xinthera, Inc. Parp1 inhibitors and uses thereof
WO2022228387A1 (en) * 2021-04-26 2022-11-03 Fochon Biosciences, Ltd. Compounds as parp inhibitors
WO2022261777A1 (en) * 2021-06-16 2022-12-22 Repare Therapeutics Inc. Use of atr inhibitors in combination with parp inhibitors for treating cancer
CN115768760A (en) * 2020-06-25 2023-03-07 阿斯利康(瑞典)有限公司 Quinoxaline derivatives as anti-cancer agents
WO2023046149A1 (en) * 2021-09-26 2023-03-30 张文燕 Quinoxaline compound and medicinal use thereof
WO2023051807A1 (en) * 2021-09-30 2023-04-06 海思科医药集团股份有限公司 Bicyclic derivative parp inhibitor and use thereof
CN116425744A (en) * 2022-01-13 2023-07-14 优领医药科技(香港)有限公司 Piperazine-containing ring derivative, pharmaceutically acceptable salt thereof, and preparation method and application thereof
WO2023138541A1 (en) * 2022-01-20 2023-07-27 微境生物医药科技(上海)有限公司 Picolinamide parp inhibitor, and preparation method therefor and medical use thereof

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114144413A (en) * 2019-07-19 2022-03-04 阿斯利康(瑞典)有限公司 PARP1 inhibitors
CN115768760A (en) * 2020-06-25 2023-03-07 阿斯利康(瑞典)有限公司 Quinoxaline derivatives as anti-cancer agents
WO2022225934A1 (en) * 2021-04-19 2022-10-27 Xinthera, Inc. Parp1 inhibitors and uses thereof
WO2022228387A1 (en) * 2021-04-26 2022-11-03 Fochon Biosciences, Ltd. Compounds as parp inhibitors
WO2022261777A1 (en) * 2021-06-16 2022-12-22 Repare Therapeutics Inc. Use of atr inhibitors in combination with parp inhibitors for treating cancer
WO2023046149A1 (en) * 2021-09-26 2023-03-30 张文燕 Quinoxaline compound and medicinal use thereof
WO2023051807A1 (en) * 2021-09-30 2023-04-06 海思科医药集团股份有限公司 Bicyclic derivative parp inhibitor and use thereof
CN116425744A (en) * 2022-01-13 2023-07-14 优领医药科技(香港)有限公司 Piperazine-containing ring derivative, pharmaceutically acceptable salt thereof, and preparation method and application thereof
WO2023138541A1 (en) * 2022-01-20 2023-07-27 微境生物医药科技(上海)有限公司 Picolinamide parp inhibitor, and preparation method therefor and medical use thereof

Similar Documents

Publication Publication Date Title
RU2656591C2 (en) Protein tyrosine kinase modulators and methods of use
CN111902417B (en) A kind of diaryl macrocyclic compound, pharmaceutical composition and use thereof
CN107286077B (en) A selective C-KIT kinase inhibitor
CN108834412B (en) Pyrimidines and variants thereof, and uses thereof
ES2753386T3 (en) 2-Hydroxy-1- {4 - [(4-phenyl) phenyl] carbonyl} piperazin-1-yl} ethane-1-one derivatives and related compounds as fatty acid synthase inhibitors (FASN) for the treatment of cancer
CN110612285B (en) Amide derivatives as Nav1.7 and Nav1.8 blockers
TWI896726B (en) Egfr inhibitor, preparation method therefor and application thereof
CN101641339A (en) Compounds for the prevention and treatment of cardiovascular disease
WO2015158310A1 (en) Tyrosine kinase inhibitor and uses thereof
CN109790166A (en) Imidazopyridine compounds for the treatment of cancer
CN106573906A (en) Piperidine-dione derivatives
CN104936945A (en) Pyridic ketone derivatives, method of preparing same, and pharmaceutical application thereof
WO2023066363A1 (en) Parp-1 degradation agent and use thereof
CN105294654A (en) ALK kinase inhibitor as well as preparation method and application thereof
TW201605805A (en) Quinazolinone and isoquinolinone derivative
CN116496318A (en) A kind of heterocyclic derivative and its composition and pharmaceutical application
CN113248497B (en) Fused ring derivatives useful as FGFR4 inhibitors
WO2020200160A1 (en) Quinolyl-containing compound and pharmaceutical composition, and use thereof
WO2024093956A1 (en) Polycyclic poly(adp-ribose) polymerase selective inhibitor
CN103936762B (en) Morpholine quinolines, Preparation Method And The Use
CN115867542B (en) Novel benzimidazole compound
CN114026090B (en) Compound for inhibiting EGFR kinase and preparation method and application thereof
WO2025092973A1 (en) Parp1-targeting compound and use thereof
WO2025051242A1 (en) Ras inhibitor
TW202404605A (en) Pharmaceutical composition containing SOS1 inhibitor

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: 24884970

Country of ref document: EP

Kind code of ref document: A1