WO2023202626A1 - 稠和哒嗪酮化合物作为prmt5抑制剂 - Google Patents

稠和哒嗪酮化合物作为prmt5抑制剂 Download PDF

Info

Publication number
WO2023202626A1
WO2023202626A1 PCT/CN2023/089249 CN2023089249W WO2023202626A1 WO 2023202626 A1 WO2023202626 A1 WO 2023202626A1 CN 2023089249 W CN2023089249 W CN 2023089249W WO 2023202626 A1 WO2023202626 A1 WO 2023202626A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
haloalkyl
cycloalkyl
halogen
membered heterocyclyl
Prior art date
Application number
PCT/CN2023/089249
Other languages
English (en)
French (fr)
Inventor
杨旭
王虎庭
石磊
刘磊
孙广龙
王晶晶
高蓓蓉
孟庆华
王建浩
Original Assignee
北京望实智慧科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 北京望实智慧科技有限公司 filed Critical 北京望实智慧科技有限公司
Publication of WO2023202626A1 publication Critical patent/WO2023202626A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic 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
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/5025Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with heterocyclic ring systems
    • 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
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/26Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings condensed with carbocyclic rings or ring systems
    • C07D237/30Phthalazines
    • C07D237/32Phthalazines with oxygen atoms directly attached to carbon atoms of the nitrogen-containing ring
    • 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
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • the present invention relates to a new class of PRMT5 inhibitors, or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates or solvates thereof.
  • the present invention also relates to methods for preparing the compounds, pharmaceutical compositions containing the compounds, and the effects of the compounds in preventing and treating PRMT5-mediated diseases such as cancer.
  • Protein arginine methyltransferase 5 belongs to type II PRMT. It is a type of S-adenosylmethionine (SAM)-dependent methyltransferase and is mainly responsible for converting the methyl group of SAM. Symmetrically transferred to the guanidine nitrogen atom at the end of an arginine residue in histones or other proteins.
  • SAM S-adenosylmethionine
  • PRMT5 forms a complex with MEP50 (methylosome protein 50) to recognize the substrate and localize it. At the same time, the complex morphology is also required for PRMT5 to catalyze the methyl transfer activity of histone 2A and histone 4.
  • PRMT5 is a general transcriptional repressor that can regulate the process of gene transcription and protein modification. At the same time, PRMT5 plays an important role in the proliferation, differentiation, and apoptosis of tumor cells and is a highly potential tumor treatment target. However, PRMT5 is also an essential gene for normal cells. PRMT5 knockout and siRNA knockdown studies have shown that inhibiting the activity of PRMT5 in normal tissues may cause many toxic side effects such as thrombocytopenia, infertility, skeletal muscle loss, and cardiac hypertrophy.
  • MTAP methylthioadenosine phosphorylase
  • PRMT5 inhibitors under investigation include SAM competitive inhibitors, substrate competitive inhibitors, SAM and substrate dual competitive inhibitors, and MTA synergistic inhibitors.
  • Non-MTA synergistic PRMT5 inhibitors indiscriminately inhibit the activity of PRMT5, and dose-limiting toxic side effects such as thrombocytopenia, anemia, and neutropenia have been found.
  • MTA synergistic inhibitors target the PRMT5/MTA complex and are expected to inhibit MTAP-deficient tumor cells while eliminating the impact on MTAP WT cells, which can improve the treatment window.
  • the present invention uses the PRMT5/MTA complex as a target and develops a new class of small molecule inhibitors that can be used to treat various cancers.
  • the compound of the present invention targets the PRMT5/MTA complex, is an MTA synergistic PRMT5 inhibitor, has excellent PRMT5/MTA inhibitory activity, and has obvious selectivity for the PRMT5/SAM complex.
  • the compounds of the present invention have good ADMET (absorption-distribution-metabolism-excretion-toxicity) properties.
  • the invention provides compounds of formula (I), or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers thereof Isomers, prodrugs, polymorphs, hydrates or solvates:
  • Z 1 , Z 2 and Z 3 are independently selected from N, CD or CH, and at least one of them is N;
  • Ring A is phenyl or 5-6 membered heteroaryl
  • R 1 is selected from H, D, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, which is optionally replaced by 1, 2, 3, Substituted with 4, 5, 6, 7, 8, 9 or 10 R substituents;
  • R 2 is selected from H, D, -C 0-6 alkylene-halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene -NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 3-8 cycloalkyl or 4-7 membered heterocyclyl;
  • n 0, 1, 2 or 3;
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C(O)OR a , C(O)NR b R c , S(O)R a , S(O) 2 R a , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aromatic group or 5-10 membered heteroaryl, preferably H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl; or two on the same or different atoms R' and the atom form a C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, which is optionally replaced by 1 or more D, Halogen, C
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, or R b and R c and the nitrogen atoms to which they are connected form a 4-7 membered heterocyclyl;
  • Each group in rings A, R 1 , R 2 , R, R', R a , R b and R c may be optionally substituted by D until completely deuterated.
  • the invention provides a compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph, hydrate or solvent thereof Compound:
  • Z 1 , Z 2 and Z 3 are independently selected from N, CD or CH, and at least one of them is N;
  • Ring A is phenyl or 5-6 membered heteroaryl
  • R 1 is selected from H, D, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, which is optionally replaced by 1, 2, 3, Substituted with 4, 5, 6, 7, 8, 9 or 10 R substituents;
  • R 2 is selected from H, D, -C 0-6 alkylene-halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene -NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 3-8 cycloalkyl or 4-7 membered heterocyclyl;
  • n 0, 1, 2 or 3;
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C(O)OR a , C(O)NR b R c , S(O)R a , S(O) 2 R a , C 1-6 alkyl, C 1-6 haloalkyl, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, preferably H, D , halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl; or two R' on the same or different atoms form a C 3-8 cycloalkyl with the atom Or 4-7 membered heterocyclyl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, or R b and R c and the nitrogen atoms to which they are connected form a 4-7 membered heterocyclyl;
  • Each group in rings A, R 1 , R 2 , R, R', R a , R b and R c may be optionally substituted by D until completely deuterated.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention, and optionally a pharmaceutically acceptable excipient, such as a carrier, adjuvant or vehicle.
  • the invention provides a pharmaceutical composition containing a compound of the invention and a pharmaceutically acceptable excipient, which also contains other therapeutic agents, such as selected from the following target drugs/cell activity modulators, including CDK4/ 6 inhibitors, MAT2A inhibitors, MAPK1/MAPK3 inhibitors, Type I PRMT inhibitors, EGFR inhibitors, SHP2 inhibitors, pan-KRAS inhibitors, KRASG12C inhibitors, RAF inhibitors, MEK inhibitors, ERK inhibitors, Bcl -2 inhibitors, SOS1 inhibitors, PARP inhibitors, MALT1 inhibitors, MALT2 inhibitors, BTK inhibitors, PI3K inhibitors, AKT inhibitors, FGFR inhibitors, DNA methyltransferase (DNMT) inhibitors, EZH1/ 2 inhibitors, EZH2 inhibitors, Menin-MLL inhibitors, IDH1 inhibitors, IDH2 inhibitors, IDH1/2 inhibitors, chemotherapy drugs, radiotherapy, STING agonists or immune checkpoint inhibitors/modul
  • the invention provides pharmaceutical compositions containing a compound of the invention and a pharmaceutically acceptable excipient, further comprising other therapeutic agents.
  • the invention provides the use of a compound of the invention for the preparation of a medicament for the treatment and/or prevention of PRMT5-mediated diseases.
  • the invention provides a method of treating and/or preventing a PRMT5-mediated disease in a subject, comprising administering to said subject a compound of the invention or a pharmaceutical composition of the invention.
  • the invention provides a compound of the invention or a pharmaceutical composition of the invention for use in the treatment and/or prevention of PRMT5-mediated diseases.
  • the present invention is used to treat and/or prevent cancer.
  • the present invention is used for the treatment and/or prevention of the following cancers: Heart: sarcomas (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyomas, fibromas, lipomas and teratomas.
  • bronchial carcinoma squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma
  • alveolar (bronchiolar) carcinoma bronchial adenoma, sarcoma, lymphoma, enchondromatous hamartoma, mesothelial carcinoma Tumors
  • gastrointestinal tract esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastric Secretinoma, carcinoid tumor, hemangioma), small intestine (adenocarcinoma, lymphoma, carcinoid tumor), Kaposi's sarcoma, leiomyoma, hemangioma
  • the PRMT5-mediated disease of the invention is selected from the following cancers: malignant peripheral nerve sheath tumors, interstitial skin cancer, glioblastoma multiforme, pancreatic cancer, cholangiocarcinoma, prostate cancer, breast cancer, brain cancer, skin cancer, cervical cancer, bladder cancer, astrocytoma, colorectal cancer, endometrial cancer, Esophageal cancer, gastric cancer, thymoma, head and neck cancer, hepatocellular carcinoma, laryngeal cancer, lung squamous cell carcinoma, lung adenocarcinoma, oral cancer, ovarian cancer, renal and thyroid cancer, and sarcoma.
  • the PRMT5-mediated disease of the invention is selected from MTAP-related cancers, such as hepatocellular carcinoma, breast cancer, skin cancer, bladder cancer, liver cancer, pancreatic cancer or head and neck cancer.
  • MTAP-related cancers such as hepatocellular carcinoma, breast cancer, skin cancer, bladder cancer, liver cancer, pancreatic cancer or head and neck cancer.
  • C 1-6 alkyl includes C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1-6 , C 1-5 , C 1-4 , C 1-3 , C 1 -2 , C 2-6 , C 2-5 , C 2-4 , C 2-3 , C 3-6 , C 3-5, C 3-4 , C 4-6 , C 4-5 and C 5 -6 alkyl.
  • C 1-6 alkyl refers to a straight or branched chain saturated hydrocarbon group having 1 to 6 carbon atoms. In some embodiments, C 1-4 alkyl and C 1-2 alkyl are preferred. Examples of C 1-6 alkyl groups include: methyl (C 1 ), ethyl (C 2 ), n-propyl (C 3 ), isopropyl (C 3 ), n-butyl (C 4 ), tert-butyl base (C 4 ), sec-butyl (C 4 ), isobutyl (C 4 ), n-pentyl (C 5 ), 3-pentyl (C 5 ), pentyl (C 5 ), neopentyl ( C 5 ), 3-methyl-2-butyl (C 5 ), tert-pentyl (C 5 ) and n-hexyl (C 6 ).
  • C 1-6 alkyl also includes heteroalkyl groups in which one or more (e.g., 1, 2, 3, or 4) carbon atoms are replaced by heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, Phosphorus) substitution.
  • Alkyl groups may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • alkyl abbreviations include: Me(-CH 3 ), Et(-CH 2 CH 3 ), iPr(-CH(CH 3 ) 2 ), nPr(-CH 2 CH 2 CH 3 ), n-Bu(-CH 2 CH 2 CH 2 CH 3 ) or i-Bu(-CH 2 CH(CH 3 ) 2 ).
  • C 2-6 alkenyl refers to a straight or branched chain hydrocarbon group having 2 to 6 carbon atoms and at least one carbon-carbon double bond. In some embodiments, C 2-4 alkenyl is preferred. Examples of C 2-6 alkenyl groups include: vinyl (C 2 ), 1-propenyl (C 3 ), 2-propenyl (C 3 ), 1-butenyl (C 4 ), 2-butenyl (C 4 ), butadienyl (C 4 ), pentenyl (C 5 ), pentadienyl (C 5 ), hexenyl (C 6 ), etc.
  • C 2-6 alkenyl also includes heteroalkenyl groups in which one or more (e.g., 1, 2, 3, or 4) carbon atoms are replaced by heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, Phosphorus) substitution.
  • Alkenyl groups may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 2-6 alkynyl refers to a straight or branched chain hydrocarbon group having 2 to 6 carbon atoms, at least one carbon-carbon triple bond, and optionally one or more carbon-carbon double bonds. In some embodiments, C 2-4 alkynyl is preferred. Examples of C 2-6 alkynyl groups include, but are not limited to: ethynyl (C 2 ), 1-propynyl (C 3 ), 2-propynyl (C 3 ), 1-butynyl (C 4 ), 2-Butynyl (C 4 ), pentynyl (C 5 ), hexynyl (C 6 ), etc.
  • C 2-6 alkynyl also includes heteroalkynyl groups in which one or more (e.g., 1, 2, 3, or 4) carbon atoms are replaced by heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, Phosphorus) substitution.
  • An alkynyl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 1-6 alkylene refers to a divalent group formed by removing another hydrogen of C 1-6 alkyl, and may be substituted or unsubstituted. In some embodiments, C 1-4 alkylene, C 2-4 alkylene, and C 1-3 alkylene are preferred.
  • the unsubstituted alkylene group includes, but is not limited to: methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), propylene (-CH 2 CH 2 CH 2 -), ethylene Base (-CH 2 CH 2 CH 2 CH 2 -), pentylene (- CH 2 CH 2 CH 2 CH 2 -), hexylene (-CH 2 CH 2 CH 2 CH 2 CH 2 CH 2 -), etc.
  • alkylene groups substituted by one or more alkyl (methyl) include, but are not limited to: substituted methylene (-CH(CH 3 )- , -C(CH 3 ) 2 -), substituted ethylene (-CH(CH 3 )CH 2 -, -CH 2 CH(CH 3 )-, -C(CH 3 ) 2 CH 2 -, -CH 2 C(CH 3 ) 2- ), substituted propylene (-CH(CH 3 )CH 2 CH 2 -, -CH 2 CH(CH 3 )CH 2 -, -CH 2 CH 2 CH(CH 3 ) -, -C(CH 3 ) 2 CH 2 CH 2 -, -CH 2 C(CH 3 ) 2 CH 2 -, -CH 2 CH 2 C(CH 3 ) 2 -), etc.
  • C 0-6 alkylene means a chemical bond and the above-mentioned “C 1-6 alkylene”
  • C 0-4 alkylene means a chemical bond and the above-mentioned "C 1-4 alkylene”.
  • Halo or "halogen” refers to fluorine (F), chlorine (Cl), bromine (Br) and iodine (I).
  • C 1-6 haloalkyl refers to the above-mentioned "C 1-6 alkyl” which is substituted by one or more halogen groups.
  • C 1-4 haloalkyl is particularly preferred, with C 1-2 haloalkyl being more preferred.
  • Exemplary haloalkyl groups include, but are not limited to: -CF 3 , -CH 2 F, -CHF 2 , -CHFCH 2 F, -CH 2 CHF 2 , -CF 2 CF 3 , -CCl 3 , -CH 2 Cl , -CHCl 2 , 2,2,2-trifluoro-1,1-dimethyl-ethyl, etc.
  • Haloalkyl groups may be substituted at any available point of attachment, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 3-10 cycloalkyl refers to a non-aromatic cyclic hydrocarbon group having 3 to 10 ring carbon atoms and zero heteroatoms. In some embodiments, C 3-8 cycloalkyl, C 3-7 cycloalkyl and C 3-6 cycloalkyl are particularly preferred, with C 5-6 cycloalkyl being more preferred. Cycloalkyl also includes ring systems in which the above-described cycloalkyl ring is fused with one or more aryl or heteroaryl groups, wherein the point of attachment is on the cycloalkyl ring, and in such cases the number of carbons continues as indicated The number of carbons in a cycloalkyl system.
  • Exemplary cycloalkyl groups include, but are not limited to: cyclopropyl (C 3 ), cyclopropenyl (C 3 ), cyclobutyl (C 4 ), cyclobutenyl (C 4 ), cyclopentyl ( C 5 ), cyclopentenyl (C 5 ), cyclohexyl (C 6 ), cyclohexenyl (C 6 ), cyclohexadienyl (C 6 ), cycloheptyl (C 7 ), cycloheptene group (C 7 ), cycloheptadienyl (C 7 ), cycloheptadienyl (C 7 ), etc.
  • a cycloalkyl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • 3-12 membered heterocyclyl refers to a saturated or unsaturated group of 3 to 12 membered non-aromatic ring system having ring carbon atoms and 1 to 5 ring heteroatoms, wherein each heteroatom is independently selected from Nitrogen, oxygen, sulfur, boron, phosphorus and silicon.
  • the point of attachment may be a carbon or nitrogen atom as long as the valency permits.
  • 4-10 membered heterocyclyl is preferred, which is a 4-10 membered non-aromatic ring system having ring carbon atoms and 1 to 5 ring heteroatoms; in some embodiments, 3-8 membered is preferred Heterocyclyl, which is a 3- to 8-membered non-aromatic ring system having ring carbon atoms and 1 to 4 ring heteroatoms; preferably a 4-8-membered heterocyclyl, which is a 3- to 8-membered non-aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms.
  • 4- to 8-membered non-aromatic ring system of atoms preferably 4-7-membered heterocyclyl, which is a 4- to 7-membered non-aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms; preferably 4-6-membered heterocyclyl Cyclic group, which is a 4- to 6-membered non-aromatic ring system with ring carbon atoms and 1 to 3 ring heteroatoms; more preferably, a 5-6-membered heterocyclyl group, which is a 4- to 6-membered non-aromatic ring system with ring carbon atoms and 1 to 3 ring heteroatoms.
  • Heterocyclyl also includes ring systems in which the above-described heterocyclyl ring is fused with one or more cycloalkyl groups, wherein the point of attachment is on the cycloalkyl ring, or in which the above-described heterocyclyl ring is fused with one or more aryl groups or Heteroaryl fused ring systems wherein the point of attachment is on the heterocyclyl ring; and in such cases, the number of ring members continues to represent the number of ring members in the heterocyclyl ring system.
  • Exemplary 3-membered heterocyclyl groups containing one heteroatom include, but are not limited to: aziridinyl, oxirinyl, and thiorenyl.
  • Exemplary 4-membered heterocyclyl groups containing one heteroatom include, but are not limited to: azetidinyl, oxetanyl, and thietanyl.
  • Exemplary 5-membered heterocyclyl groups containing one heteroatom include, but are not limited to: tetrahydrofuryl, dihydrofuryl, tetrahydrothienyl, dihydrothienyl, pyrrolidinyl, dihydropyrrolyl and pyrrolyl-2, 5-diketone.
  • Exemplary 5-membered heterocyclyl groups containing two heteroatoms include, but are not limited to: pyrazolidinyl, dioxol Alkyl, oxasulfuranyl, disulfuranyl and oxazolidin-2-one.
  • Exemplary 5-membered heterocyclyl groups containing three heteroatoms include, but are not limited to: triazolinyl, oxadiazolinyl, and thiadiazolinyl.
  • Exemplary 6-membered heterocyclyl groups containing one heteroatom include, but are not limited to: piperidinyl, tetrahydropyranyl, dihydropyridyl, and thianyl.
  • Exemplary 6-membered heterocyclyl groups containing two heteroatoms include, but are not limited to: piperazinyl, morpholinyl, dithianyl, and dioxanyl.
  • Exemplary 6-membered heterocyclyl groups containing three heteroatoms include, but are not limited to: hexahydrotriazinyl (triazinanyl).
  • Exemplary 7-membered heterocyclyl groups containing one heteroatom include, but are not limited to: azepanyl, oxpanyl, and thipanyl.
  • Exemplary 5-membered heterocyclyl fused to a C6 aryl ring include, but are not limited to: indolyl, isoindolyl , dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolinone, etc.
  • Exemplary 6-membered heterocyclyl fused to a C6 aryl ring include, but are not limited to: tetrahydroquinolyl, tetrahydroisoquinolyl, etc.
  • Heterocyclyl also includes the above-mentioned heterocyclyl sharing one or two atoms with a cycloalkyl, heterocyclyl, aryl or heteroaryl to form a bridged ring or spiro ring. As long as the valency allows, the shared atoms can be carbon or Nitrogen atom. Heterocyclyl also includes the above-mentioned heterocyclyl and heterocyclyl groups may be optionally substituted by one or more substituents, for example, by 1 to 5 substituents, 1 to 3 substituents or 1 substituent.
  • C 6-10 aryl refers to a monocyclic or polycyclic (e.g., bicyclic) 4n+2 aromatic ring system having 6-10 ring carbon atoms and zero heteroatoms (e.g., having a Shared 6 or 10 ⁇ electrons) group.
  • an aryl group has six ring carbon atoms ("C 6 aryl”; e.g., phenyl).
  • an aryl group has ten ring carbon atoms ("C 10 aryl”; eg, naphthyl, eg, 1-naphthyl and 2-naphthyl).
  • Aryl also includes ring systems in which the aryl ring described above is fused to one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on said aryl ring, in which case the number of carbon atoms continues to indicate The number of carbon atoms in the aryl ring system.
  • Aryl groups may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • 5-14 membered heteroaryl refers to a 5-14 membered monocyclic or bicyclic 4n+2 aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms (e.g., having a 6, 10 or 14 ⁇ electrons), wherein each heteroatom is independently selected from nitrogen, oxygen and sulfur.
  • the point of attachment may be a carbon or nitrogen atom as long as the valency permits.
  • Heteroaryl bicyclic systems may include one or more heteroatoms in one or both rings.
  • Heteroaryl also includes ring systems in which the heteroaryl ring described above is fused to one or more cycloalkyl or heterocyclyl groups, and the point of attachment is on the heteroaryl ring, in which case the carbon atom Number continues to represent the number of carbon atoms in the heteroaryl ring system.
  • 5-10 membered heteroaryl groups are preferred, which are 5-10 membered monocyclic or bicyclic 4n+2 aromatic ring systems having ring carbon atoms and 1-4 ring heteroatoms.
  • 5-6 membered heteroaryl groups are particularly preferred, which are 5-6 membered monocyclic or bicyclic 4n+2 aromatic ring systems having ring carbon atoms and 1-4 ring heteroatoms.
  • Exemplary 5-membered heteroaryl groups containing one heteroatom include, but are not limited to: pyrrolyl, furyl, and thienyl.
  • Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to: imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl and isothiazolyl.
  • Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to: triazolyl, oxadiazolyl (eg, 1,2,4-oxadiazolyl), and thiadiazolyl.
  • Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to: tetrazolyl.
  • Exemplary 6-membered heteroaryl groups containing one heteroatom include But not limited to: pyridyl or pyridone group.
  • Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to: pyridazinyl, pyrimidinyl, and pyrazinyl.
  • Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to, triazinyl and tetrazinyl, respectively.
  • Exemplary 7-membered heteroaryl groups containing one heteroatom include, but are not limited to: azepantrienyl, oxetapyltrienyl, and thioheptantrienyl.
  • Exemplary 5,6-bicyclic heteroaryl groups include, but are not limited to: indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothienyl, isobenzothienyl, benzofuranyl , benzisofuryl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzoxadiazolyl, benzothiazolyl, benzisothiazolyl, benzothiadiazolyl, Indazinyl and purinyl.
  • Exemplary 6,6-bicyclic heteroaryl groups include, but are not limited to: naphthyridinyl, pyridinyl, quinolinyl, isoquinolinyl, quinolinyl, quinoxalinyl, phthalazinyl, and quinazolinyl .
  • Heteroaryl groups may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • the divalent groups formed by removing another hydrogen from the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl groups defined above are collectively referred to as "subunits".
  • Ring-forming groups such as cycloalkyl, heterocyclyl, aryl and heteroaryl are collectively referred to as "cyclic groups”.
  • Alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, etc. are defined herein as optionally substituted groups.
  • Each R aa is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, or two R aa groups are combined to form heterocyclyl or Heteroaryl rings, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl group is independently replaced by 0, 1, 2, 3, 4 or 5 R dd groups group replacement;
  • Each R cc is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or two R cc groups are combined to form a heterocycle or heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently replaced by 0, 1, 2, 3, 4 or 5 R dd group substitution;
  • Each R ee is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocyclyl, and heteroaryl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl Alkyl, heterocyclyl, aryl and heteroaryl are independently substituted by 0, 1, 2, 3, 4 or 5 R gg groups;
  • Each R ff is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, or two R ff groups combine to form a heterocyclyl or a heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently replaced by 0, 1, 2, 3, 4 or 5 R gg group substitution;
  • cancer includes, but is not limited to, the following cancers: Heart: sarcomas (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyomas, fibromas, lipomas and teratomas; Lung: bronchial carcinoma (squamous cell carcinoma) cystic cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, enchondromatous hamartoma, mesothelioma; gastrointestinal tract: esophagus (squamous cell carcinoma) cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (duct
  • cancer includes, but is not limited to, the following cancers: malignant peripheral nerve sheath tumors, mesothelioma, glioblastoma multiforme, pancreatic cancer, cholangiocarcinoma, prostate cancer, breast cancer, brain cancer , skin cancer, cervical cancer, bladder cancer, astrocytoma, colorectal cancer, endometrial cancer, esophageal cancer, gastric cancer, thymoma, head and neck cancer, hepatocellular carcinoma, laryngeal cancer, lung squamous cell carcinoma, lung adenocarcinoma, Oral, ovarian, renal and thyroid cancers and sarcomas.
  • cancer includes, but is not limited to, the following cancers: hepatocellular carcinoma, breast cancer, skin cancer, bladder cancer, Liver, pancreatic, or head and neck cancer.
  • treatment refers to reversing, alleviating, inhibiting the progression of, or preventing the disorder or condition to which the term applies, or one or more symptoms of such disorder or condition.
  • noun “treat” refers to the action of the verb treat, as just defined.
  • the term "pharmaceutically acceptable salts” means those carboxylate salts and amino acid addition salts of the compounds of the present invention which are suitable for contact with patient tissue within the scope of reliable medical judgment and will not produce undue toxicity, Irritation effects, allergic reactions, etc., commensurate with a reasonable benefit/risk ratio, are effective for their intended use, including (where possible) zwitterionic forms of the compounds of the invention.
  • Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali metal and alkaline earth metal hydroxides or organic amines.
  • metals used as cations are sodium, potassium, magnesium, calcium, etc.
  • suitable amines are N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine and procaine.
  • Base addition salts of acidic compounds may be prepared in conventional manner by contacting the free acid form with a sufficient amount of the desired base to form the salt.
  • the free acid can be regenerated by contacting the salt form with the acid and isolating the free acid in the usual manner.
  • the free acid forms differ somewhat from their respective salt forms in certain physical properties, such as solubility in polar solvents, but for the purposes of this invention the salts are nevertheless equivalent to their respective free acids.
  • the salts may be sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, nitrates, phosphates, monohydrogen phosphates, dihydrogen phosphates, metaphosphates, pyrophosphates prepared from inorganic acids Salt, chloride, bromide, iodide, acids such as hydrochloric acid, nitric acid, sulfuric acid, hydrobromic acid, hydroiodic acid, phosphoric acid, etc.
  • Representative salts include: hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurate Acid, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthoate, Methanesulfonate, glucoheptonate, lactobionate, lauryl sulfonate and isethionate, etc.
  • Salts may also be prepared from organic acids, such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxyalkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, and the like.
  • organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxyalkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, and the like.
  • Representative salts include acetate, propionate, octanoate, isobutyrate, oxalate, malonate, succinate, suberate, sebacate, fumarate, malonate Lenate, mandelate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, naphthoate, benzenesulfonate, toluenesulfonate, phenylbenzoate Acid, citrate, lactate, maleate, tartrate, methanesulfonate, etc.
  • Pharmaceutically acceptable salts may include alkali and alkaline earth metal based cations such as sodium, lithium, potassium, calcium, magnesium, etc., as well as non-toxic ammonium, quaternary ammonium and amine cations including, but not limited to, ammonium, tetramethyl Ammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, etc. Also covered are salts of amino acids, such as arginates, gluconates, galacturonates, etc. (see, for example, Berge S.M. et al., "Pharmaceutical Salts," J. Pharm. Sci., 1977; 66:1- 19, incorporated by reference).
  • Subjects for administration include, but are not limited to: humans (i.e., males or females of any age group, e.g., pediatric subjects (e.g., infants, children, adolescents) or adult subjects (e.g., young Adults, middle-aged adults or older adults) and/or non-human animals, e.g., mammals, e.g., primates (e.g., cynomolgus monkeys, rhesus monkeys), cattle, pigs, horses, sheep , goats, rodents, cats and/or dogs.
  • the subject is human.
  • the subject is a non-human animal.
  • the terms "person,”"patient,” and “subject” are used interchangeably herein.
  • treatment includes an action in a subject suffering from a specific disease, disorder or condition that reduces the severity of the disease, disorder or condition, or delays or slows down the disease, disorder or the development of a condition ("therapeutic treatment”), and also includes effects that occur before a subject begins to suffer from a specific disease, disorder or condition ("preventive treatment").
  • an "effective amount" of a compound is an amount sufficient to elicit a target biological response.
  • the effective amount of a compound of the present invention may vary depending on factors such as, for example, the biological target, the pharmacokinetics of the compound, the disease being treated, the mode of administration, and the condition of the subject. Age health conditions and symptoms.
  • the effective amount includes a therapeutically effective amount and a preventive effective amount.
  • a "therapeutically effective amount" of a compound as used herein is an amount sufficient to provide a therapeutic benefit in treating a disease, disorder, or condition, or to cause one or more symptoms associated with the disease, disorder, or condition The amount to delay or minimize.
  • a therapeutically effective amount of a compound is that amount of therapeutic agent that, when used alone or in combination with other therapies, provides a therapeutic benefit in the treatment of a disease, disorder, or condition.
  • the term "therapeutically effective amount” may include an amount that improves overall treatment, reduces or avoids symptoms or causes of a disease or disorder, or enhances the therapeutic effect of other therapeutic agents.
  • a prophylactically effective amount of a compound as used herein is an amount sufficient to prevent a disease, disorder or condition, or to prevent one or more symptoms associated with a disease, disorder or condition, or to prevent a disease , the amount of recurrence of a disorder or condition.
  • a prophylactically effective amount of a compound is that amount of therapeutic agent that, when used alone or in combination with other agents, provides a prophylactic benefit in preventing a disease, disorder, or condition.
  • the term “prophylactically effective amount” may include an amount that improves overall prophylaxis, or an amount that enhances the prophylactic effect of other prophylactic agents.
  • Combination and related terms refer to the simultaneous or sequential administration of a compound of the invention and another therapeutic agent.
  • the compounds of the present invention may be administered simultaneously or sequentially with other therapeutic agents in separate unit dosage forms, or with other therapeutic agents in a single unit dosage form.
  • Figures 1 and 2 show the results of in vivo pharmacodynamic studies of the LU99 CDX model.
  • the "compounds of the present invention” refer to the following compounds of formula (I), formula (II) (including sub-general formulas, such as formula (I-1) to formula (IV-1), etc.), their pharmaceutical Acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates or solvates.
  • the invention provides a compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph, hydrate or Solvates:
  • Z 1 , Z 2 and Z 3 are independently selected from N, CD or CH, and at least one of them is N;
  • Ring A is phenyl or 5-6 membered heteroaryl
  • R 1 is selected from H, D, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, which is optionally replaced by 1, 2, 3, Substituted with 4, 5, 6, 7, 8, 9 or 10 R substituents;
  • R 2 is selected from H, D, -C 0-6 alkylene-halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene -NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 3-8 cycloalkyl or 4-7 membered heterocyclyl;
  • n 0, 1, 2 or 3;
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C(O)OR a , C(O)NR b R c , S(O)R a , S(O) 2 R a , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aromatic group or 5-10 membered heteroaryl, preferably H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl; or two on the same or different atoms R' and the atom form a C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, which is optionally replaced by 1 or more D, Halogen, C
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, or R b and R c and the nitrogen atoms to which they are connected form a 4-7 membered heterocyclyl;
  • Each group in rings A, R 1 , R 2 , R, R', R a , R b and R c may be optionally substituted by D until completely deuterated.
  • the present invention relates to a compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph, hydrate or Solvates:
  • Z 1 , Z 2 and Z 3 are independently selected from N, CD or CH, and at least one of them is N;
  • Ring A is phenyl or 5-6 membered heteroaryl
  • R 1 is selected from H, D, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, which is optionally replaced by 1, 2, 3, Substituted with 4, 5, 6, 7, 8, 9 or 10 R substituents;
  • R 2 is selected from H, D, -C 0-6 alkylene-halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene -NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 3-8 cycloalkyl or 4-7 membered heterocyclyl;
  • n 0, 1, 2 or 3;
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C(O)OR a , C(O)NR b R c , S(O)R a , S(O) 2 R a , C 1-6 alkyl, C 1-6 haloalkyl, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, preferably H, D , halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl; or two R' on the same or different atoms form a C 3-8 cycloalkyl with the atom Or 4-7 membered heterocyclyl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, or R b and R c and the nitrogen atoms to which they are connected form a 4-7 membered heterocyclyl;
  • Each group in rings A, R 1 , R 2 , R, R', R a , R b and R c may be optionally substituted by D until completely deuterated.
  • Z 1 is N, and Z 2 and Z 3 are independently selected from CD or CH; in another embodiment, Z 2 is N, and Z 1 and Z 3 are independently selected from CD. Or CH; In another specific embodiment, Z 3 is N, and Z 1 and Z 2 are independently selected from CD or CH; In another specific embodiment, Z 1 and Z 2 are N, and Z 3 is selected from from CD or CH; in another embodiment, Z 1 and Z 3 are N, and Z 2 is selected from CD or CH; in another embodiment, Z 2 and Z 3 are N, and Z 1 is selected from From CD or CH; in another specific embodiment, Z 1 , Z 2 and Z 3 are all N.
  • Ring A is phenyl; in another embodiment, Ring A is 5-6 membered heteroaryl, preferably 5-membered heteroaryl.
  • Ring A is selected from
  • Ring A is selected from preferred More preferably
  • R 1 is selected from H, D, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl; in another specific In an embodiment, R 1 is selected from C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl; in another specific embodiment, R 1 is selected from From 4-7 membered heterocyclyl, C 6-10 aryl or 5-6 membered heteroaryl; in another specific embodiment, R 1 is selected from phenyl, pyrrolidinyl, pyrazolidinyl, piperidine group or pyridonyl; in another embodiment, R 1 is H; in another embodiment, R 1 is D; in another embodiment, R 1 is C 3-10 cycloalkyl ; In another specific embodiment, R 1 is a 4-10 membered heterocyclyl group; In another specific embodiment, R 1 is a C 6-10 aryl group; In another specific embodiment, R
  • R 1 is substituted with 1 R substituent; in the above specific embodiments, R 1 is substituted with 2 R substituents; in the above specific embodiments, R 1 is substituted with 3 R substituents ; In the above specific embodiment, R 1 is substituted by 4 R substituents; In the above specific embodiment, R 1 is substituted by 5 R substituents; In the above specific embodiment, R 1 is substituted by 6 R substituents Substituted; in the above specific embodiments, R 1 is substituted by 7 R substituents; in the above specific embodiments, R 1 is substituted by 8 R substituents; in the above specific embodiments, R 1 is substituted by 9 R substituents group substitution; in the above specific embodiments, R 1 is substituted with 10 R substituents.
  • R is substituted by 1 R' substituent; in the above embodiment, R is substituted by 2 R' substituents; in the above embodiment, R is substituted by 3 R' substituents ; In the above specific embodiments, R is substituted by 4 R' substituents;
  • R' is H; in another more specific embodiment, R' is D; in another more specific embodiment, R' is halogen; in another more specific implementation In another more specific embodiment, R' is CN; in another more specific embodiment, R' is OR a ; in another more specific embodiment, R' is NR b R c ; in another more specific embodiment in, R' is C(O)OR a ; in another more specific embodiment, R' is C(O)NR b R c ; in another more specific embodiment, R' is S(O )R a ; in another more specific embodiment, R' is S(O) 2 R a ; in another more specific embodiment, R' is C 1-6 alkyl; in another more specific embodiment In an embodiment, R' is C 1-6 haloalkyl; in another more specific embodiment, R' is C 2-6 alkenyl; in another more specific embodiment, R' is C 2 -6 alkynyl; in another more specific embodiment, R' is C 3-10 cycloalkyl
  • R 2 is selected from H, D, -C 0-6 alkylene-halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 3-8 cycloalkyl or 4-7 membered heterocyclyl; in another embodiment
  • R 2 is selected from H, halogen, CN, -C 0-6 alkylene -OR a , -C 0-6 alkylene -NR b R c , C 1-6 alkyl, C 1-6 haloalkyl group or C 3-8 cycloalkyl; in another specific embodiment, R 2 is selected from H, -C 0-6 alkylene-OR a , C 1-6 alkyl, C 1-6 haloalkyl or C 3-6 cycloalkyl; in another specific embodiment, R 2 is selected from H, -
  • any technical solution or any combination thereof in any of the above specific embodiments may be combined with any technical solution or any combination thereof in other specific embodiments.
  • any technical solution of ring A or any combination thereof can be combined with any technical solution of Z 1 , Z 2 , Z 3 , R 1 , R 2 and m, etc., or any combination thereof.
  • the present invention is intended to include combinations of all these technical solutions, and due to space limitations, they will not be listed one by one.
  • the invention provides compounds of formula (I), or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates thereof substance or solvate:
  • Z 1 , Z 2 and Z 3 are independently selected from N, CD or CH, and at least one of them is N;
  • Ring A is phenyl or 5-6 membered heteroaryl
  • R 1 is selected from H, D, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, which is optionally replaced by 1, 2, 3, Substituted with 4, 5, 6, 7, 8, 9 or 10 R substituents;
  • R 2 is selected from H, D, -C 0-6 alkylene-halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene -NR b R c , C 1-6 Alkyl, C 1-6 haloalkyl, C 3-8 cycloalkyl or 4-7 membered heterocyclyl;
  • n 0, 1, 2 or 3;
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C(O)OR a , C(O)NR b R c , S(O)R a , S(O) 2 R a , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aromatic group or 5-10 membered heteroaryl, preferably H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl; or two on the same or different atoms R' and the atom form a C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, which is optionally replaced by 1 or more D, Halogen, C
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, or R b and R c and the nitrogen atoms to which they are connected form a 4-7 membered heterocyclyl;
  • Each group in rings A, R 1 , R 2 , R, R', R a , R b and R c may be optionally substituted by D until completely deuterated.
  • the invention provides compounds of formula (I), or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates thereof substance or solvate:
  • Z 1 , Z 2 and Z 3 are independently selected from N, CD or CH, and at least one of them is N;
  • Ring A is phenyl or 5-6 membered heteroaryl
  • R 1 is selected from H, D, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, which is optionally replaced by 1, 2, 3, Substituted with 4, 5, 6, 7, 8, 9 or 10 R substituents;
  • R 2 is selected from H, D, -C 0-6 alkylene-halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene -NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 3-8 cycloalkyl or 4-7 membered heterocyclyl;
  • n 0, 1, 2 or 3;
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C(O)OR a , C(O)NR b R c , S(O)R a , S(O) 2 R a , C 1-6 alkyl, C 1-6 haloalkyl, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, preferably H, D , halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl; or two R' on the same or different atoms form a C 3-8 cycloalkyl with the atom Or 4-7 membered heterocyclyl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, or R b and R c and the nitrogen atoms to which they are connected form a 4-7 membered heterocyclyl;
  • Each group in rings A, R 1 , R 2 , R, R', R a , R b and R c may be optionally substituted by D until completely deuterated.
  • the present invention provides compounds of the above formula (I), or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, Hydrate or solvate, in which ring A is a 5-6 membered heteroaryl group;
  • R1 and R2 are as defined above.
  • the present invention provides compounds of the above formula (I), or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, Hydrate or solvate, wherein R 2 is selected from H, halogen, CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene -NR b R c , C 1-6 alkyl , C 1-6 haloalkyl or C 3-8 cycloalkyl; preferably selected from H, -C 0-6 alkylene-OR a , C 1-6 alkyl, C 1-6 haloalkyl or C 3- 6 cycloalkyl; more preferably selected from H, F, Cl, Br, CH 2 CH 2 OH, Me, Et, Pr, iPr, CF 3 or cyclopropyl; more preferably C 1-6 alkyl or C 1 -6 Haloalkyl, especially Me.
  • the present invention provides compounds of the above formula (I), or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, Hydrate or solvate, wherein R 1 is selected from C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl; preferably selected from 4-7 membered Heterocyclyl, C 6-10 aryl or 5-6 membered heteroaryl; more preferably selected from phenyl, pyrrolidinyl, pyrazolidinyl, piperidyl or pyridonyl.
  • R is selected from halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1 -6 haloalkyl; or two R on the
  • the present invention provides compounds of the above formula (I), or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, Hydrate or solvate, wherein R' is selected from H, D, halogen, CN, OR a , NR b R c , C(O)OR a , C(O)NR b R c , S(O)R a , S(O ) 2 R a , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, 4-10 membered hetero Ring group, C 6-10 aryl or 5-10 membered heteroaryl; preferably, R' is selected from H, D, halogen, OR a , NR b R c , C 1-6 alkyl, C 1-6 Haloalkyl, C 2-6 alkenyl, C 2
  • the present invention provides compounds of the above formula (I), or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, Hydrates or solvates having the following general formula:
  • Y 1 is selected from N or CR Y1 ;
  • Y 2 is selected from N or CR Y2 ;
  • Y 3 is selected from N or CR Y3 ;
  • Y4 is selected from N or CR Y4 ;
  • R N is selected from H, C 1-6 alkyl or C 1-6 haloalkyl
  • R 1a , R 2a , R 3a , R 4a , R 5a , R 1b , R 1b ', R 2b , R 2b ', R 3b and R 4b have the same definitions as R above;
  • R Y1 , R Y2 , R Y3 and R Y4 have the same definitions as R'above;
  • the invention provides the above-described compounds, or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates or solvents thereof compound, which has the following general formula:
  • R 2 is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 3-8 cycloalkyl or 4-7 membered heterocyclyl;
  • R 1a is selected from H, D, halogen, CN, OR a or NR b R c ;
  • R 2a is selected from H, D, halogen or CN;
  • R 3a is selected from H or D
  • R 4a is selected from OR a or NR b R c ;
  • R 5a is selected from H, D or CN
  • R 3a , R 4a and the carbon atoms to which they are connected form a 4-6 membered heterocyclyl group or a 5-6 membered heteroaryl group, which is optionally substituted by 1, 2, 3 or 4 R'substituents;
  • R' is selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; or two R' on the same or different atoms and the atom form a C 3-8 cycloalkyl or 4-7 One-membered heterocyclyl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, or R b and R c and the nitrogen atoms to which they are connected form a 4-7 membered heterocyclyl.
  • the present invention provides compounds of the above formula (III-1), (III-2) or (III-3), or pharmaceutically acceptable salts, isotopic variants, tautomers thereof body, stereoisomer, prodrug, polymorph, hydrate or solvate, wherein,
  • R 2 is selected from H, C 1-6 alkyl or C 1-6 haloalkyl
  • R 1a is halogen, preferably F
  • R 2a is selected from H, D or halogen
  • R 3a is H or D
  • R 4a is OR a ;
  • R 5a is CN
  • R 3a , R 4a and the carbon atoms to which they are attached form a furyl or dihydrofuryl group, which is optionally substituted by 1, 2, 3 or 4 R'substituents;
  • R' is selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; or two R' on the same or different atoms and the atom form a C 3-6 cycloalkyl or 5-6 One-membered heterocyclyl;
  • R a is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 cycloalkyl or 4-7 membered heterocyclyl.
  • the present invention provides compounds of the above formula (III-1), (III-2) or (III-3), or pharmaceutically acceptable salts, isotopic variants, tautomers thereof body, stereoisomer, prodrug, polymorph, hydrate or solvate, wherein,
  • R 2 is selected from H, C 1-4 alkyl or C 1-4 haloalkyl
  • R 1a is halogen, preferably F
  • R 2a is selected from H, D or halogen
  • R 3a is H or D
  • R 4a is OR a ;
  • R 5a is CN
  • R 3a , R 4a and the carbon atoms to which they are connected form a dihydrofuranyl group, which is substituted by 1, 2, 3 or 4 R'substituents;
  • R' is selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; or two R' on the same or different atoms and the atom form a C 3-6 cycloalkyl or 5-6 One-membered heterocyclyl, preferably cyclopropyl;
  • R a is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl or 5-6 membered heterocyclyl.
  • the present invention provides compounds of the above formula (III-1), (III-2) or (III-3), or pharmaceutically acceptable salts, isotopic variants, tautomers thereof body, stereoisomer, prodrug, polymorph, hydrate or solvate, wherein,
  • R 2 is Me
  • R 1a is F
  • R 2a is H or Cl
  • R 3a , R 4a and the carbon atoms to which they are connected form a dihydrofuranyl group, which is substituted by 1, 2, 3 or 4 R'substituents;
  • R 5a is CN
  • R' is selected from H or Me, or two R' on the same or different atoms form a cyclopropyl group with the atom.
  • the invention provides the above-described compounds, or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates or solvents thereof compound, which has the following general formula:
  • R 2 is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 3-8 cycloalkyl or 4-7 membered heterocyclyl;
  • R 3b and R 4b are independently selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl; or R 3b , R 4b and the carbon to which they are attached
  • the atoms form C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, which are optionally replaced by 1, Substituted with 2, 3 or 4 R'substituents;
  • R N is selected from H, C 1-6 alkyl or C 1-6 haloalkyl
  • R 2b and R 2b ' are independently selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl; or R 2b and R 2b on the same or different atoms R 2b ' and the carbon atoms to which they are connected form a C 3-6 cycloalkyl or 4-7 membered heterocyclyl; or R 2b ' and R 3b and the carbon atoms to which they are connected form a C 6-10 aryl or 5-10 metaheteroaryl;
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl; or two R' on the same or different atoms form with the atom C 3-8 cycloalkyl or 4-7 membered heterocyclyl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, or R b and R c and the nitrogen atoms to which they are connected form a 4-7 membered heterocyclyl.
  • the present invention provides the compound of the above formula (IV-1), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • R 2 is selected from H, C 1-6 alkyl or C 1-6 haloalkyl
  • R 3b and R 4b are independently selected from H, D, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl; or R 3b , R 4b and the carbon atoms to which they are connected form C 3 -6 cycloalkyl, 4-7 membered heterocyclyl, phenyl or 5-6 membered heteroaryl, optionally substituted by 1, 2, 3 or 4 R'substituents;
  • R N is selected from H, C 1-6 alkyl or C 1-6 haloalkyl
  • R 2b and R 2b ' are independently selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl; or R 2b and R 2b on the same or different atoms R 2b ' and the carbon atoms they are connected to form a C 3-6 cycloalkyl group; or R 2b ' and R 3b and the carbon atoms they are connected to form a phenyl or 5-6 membered heteroaryl group;
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, or R b and R c and the nitrogen atoms to which they are connected form a 4-7 membered heterocyclyl.
  • the invention provides the above-described compounds, or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates or solvents thereof compound, which has the following general formula:
  • X is selected from NR N or CR 2b R 2b ';
  • R 2 is selected from H, C 1-6 alkyl or C 1-6 haloalkyl
  • Y 1 is selected from N or CR Y1 ;
  • Y 2 is selected from N or CR Y2 ;
  • Y 3 is selected from N or CR Y3 ;
  • Y4 is selected from N or CR Y4 ;
  • R N is selected from H, C 1-6 alkyl or C 1-6 haloalkyl
  • R 2b and R 2b ' are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; or R 2b and R 2b ' and the carbon atoms to which they are connected form a C 3-6 cycloalkyl;
  • R Y1 , R Y2 , R Y3 and R Y4 are independently selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl or 4-7 membered heterocyclyl, or R b and R c and the nitrogen atom to which they are connected form a 4-7 membered heterocyclyl.
  • the present invention provides the compound of the above formula (IV-2), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • X is selected from NR N or CR 2b R 2b ';
  • R 2 is selected from H, C 1-4 alkyl or C 1-4 haloalkyl
  • Y 1 is selected from N or CR Y1 ;
  • Y 2 is selected from N or CR Y2 ;
  • Y 3 is selected from N or CR Y3 ;
  • Y4 is selected from N or CR Y4 ;
  • R N is selected from H, C 1-4 alkyl or C 1-4 haloalkyl
  • R 2b and R 2b ' are independently selected from H, D, C 1-4 alkyl or C 1-4 haloalkyl; or R 2b and R 2b ' and the carbon atoms to which they are connected form a C 3-6 cycloalkyl;
  • R Y1 , R Y2 , R Y3 and R Y4 are independently selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl or C 1-6 haloalkyl, or R b and R c and the nitrogen atoms to which they are connected form a 5-6 membered heterocyclyl group.
  • the present invention provides the compound of the above formula (IV-2), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • X is selected from NR N or CR 2b R 2b ';
  • R 2 is Me
  • Y 1 is selected from N or CR Y1 ;
  • Y 2 is selected from N or CR Y2 ;
  • Y 3 is selected from N or CR Y3 ;
  • Y4 is selected from N or CR Y4 ;
  • R N is selected from H or Me
  • R 2b and R 2b ' are independently selected from H or Me, or R 2b and R 2b ' and the carbon atom to which they are attached form a cyclopropyl group;
  • R Y1 , R Y2 , R Y3 and R Y4 are selected from H, F, CN, Me, CF 3 or OMe.
  • the invention provides the above-described compounds, or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates or solvents thereof compound, which has the following general formula:
  • R 2 is selected from H, C 1-6 alkyl or C 1-6 haloalkyl
  • R 2b and R 2b ' are independently selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl;
  • R 3b is selected from H, D, halogen or CN;
  • R 4b is selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl;
  • R 2b ' and R 3b and the carbon atoms to which they are connected form phenyl or 5-6 membered heteroaryl;
  • R 3b , R 4b and the carbon atoms to which they are connected form a C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, which is optionally replaced by 1 , 2, 3 or 4 R' substituents substituted;
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl; or two R' on the same or different atoms form with the atom C 3-8 cycloalkyl or 4-7 membered heterocyclyl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, or R b and R c and the nitrogen atoms to which they are connected form a 4-7 membered heterocyclyl.
  • the present invention provides the compound of the above formula (IV-3), or a pharmaceutically acceptable salt, isotope change thereof isomer, tautomer, stereoisomer, prodrug, polymorph, hydrate or solvate, wherein,
  • R 2 is selected from H, C 1-4 alkyl or C 1-4 haloalkyl
  • R 2b and R 2b ' are independently selected from H, D, halogen, CN, OR a , NR b R c , C 1-4 alkyl or C 1-4 haloalkyl;
  • R 3b is selected from H or D
  • R 4b is selected from H, D, OR a , NR b R c , C 1-4 alkyl or C 1-4 haloalkyl;
  • R 2b ' and R 3b and the carbon atom to which they are attached form a phenyl group
  • R 3b , R 4b and the carbon atoms to which they are connected form C 3-6 cycloalkyl, 5-6 membered heterocyclyl, phenyl or 5-6 membered heteroaryl, which is optionally replaced by 1, 2, 3 Or substituted by 4 R'substituents;
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl or C 1-6 haloalkyl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl or C 1-6 haloalkyl.
  • the present invention provides the compound of the above formula (IV-3), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • R 2 is Me
  • R 2b is selected from H, D, F, Cl, Br, I, Me or CF 3 ;
  • R 2b ' is selected from H, D, F, Cl, Br, I, Me, CF 3 or OMe;
  • R 3b is H or D
  • R 4b is selected from H, D, Me, OMe or O-cyclopropyl
  • R 3b , R 4b and the carbon atoms to which they are connected form phenyl, cyclopentenyl or thienyl;
  • R 2b ', R 3b and the carbon atom to which they are attached form a phenyl group.
  • the invention provides the above-described compounds, or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates or solvents thereof compound, which has the following general formula:
  • R 1 and R 2 are as defined in any one of claims 1 and 3-6;
  • R 1 is a 4-7 membered heterocyclyl group, which is optionally substituted by 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 R substituents;
  • R 2 is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C(O)OR a , C(O)NR b R c , S(O)R a , S(O) 2 R a , C 1-6 alkyl, C 1-6 haloalkyl, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl , 4-7 membered heterocyclic group, C 6-10 aryl group or 5-10 membered heteroaryl group.
  • the present invention provides the compound of formula (V) above, or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph, Hydrate or solvate, wherein,
  • R 1 is a 5-6 membered heterocyclyl group, which is optionally substituted by 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 R substituents;
  • R 2 is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, 5-6 membered heterocyclyl, Phenyl or 5-6 membered heteroaryl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl Or 4-7 membered heterocyclyl.
  • the present invention provides the compound of formula (V) above, or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph, Hydrate or solvate, wherein,
  • R 1 is selected from pyrrolidinyl or piperidinyl, which is optionally substituted by 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 R substituents;
  • R 2 is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • R' is selected from H, D, halogen, CN, C 1-6 alkyl, C 1-6 haloalkyl, phenyl or 5-6 membered heteroaryl.
  • the invention provides the above-described compounds, or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates or solvents thereof compound, which has the following general formula:
  • R 2 is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • R 3b , R 4b and the carbon atoms to which they are connected form a C 6-10 aryl group or a 5-10 membered heteroaryl group, which is optionally substituted by 1, 2, 3 or 4 R'substituents;
  • R 2b and R 2b ' are independently selected from H, D, halogen, CN, C 1-6 alkyl or C 1-6 haloalkyl;
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C(O)OR a , C(O)NR b R c , S(O)R a , S(O) 2 R a , C 1-6 alkyl, C 1-6 haloalkyl, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl , 4-7 membered heterocyclic group, C 6-10 aryl group or 5-10 membered heteroaryl group.
  • the present invention provides the compound of the above formula (VI-1), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • R 2 is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • R 3b , R 4b and the carbon atoms to which they are attached form a phenyl group, which is optionally substituted by 1, 2, 3 or 4 R'substituents;
  • R 2b and R 2b ' are independently selected from H, D, halogen, CN, C 1-6 alkyl or C 1-6 haloalkyl;
  • R' is selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, 5-6 membered heterocyclyl, Phenyl or 5-6 membered heteroaryl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl.
  • the present invention provides the compound of the above formula (VI-1), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • X is selected from CR 2b R 2b ' or CR 2b R 2b '-CR 2b R 2b ';
  • R 2 is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • R 1b , R 1b ' and the carbon atoms to which they are connected form C O;
  • R 3b , R 4b and the carbon atoms to which they are attached form a phenyl group, which is optionally substituted by 1, 2, 3 or 4 R'substituents;
  • R 2b and R 2b ' are independently selected from H or D;
  • R' is selected from H, D, halogen, CN, C 1-6 alkyl, C 1-6 haloalkyl, phenyl or 5-6 membered heteroaryl.
  • the invention provides the above-described compounds, or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates or solvents thereof compound, which has the following general formula:
  • X is CR 2b R 2b ';
  • R 2 is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • Y 1 is selected from N or CR Y1 ;
  • Y 2 is selected from N or CR Y2 ;
  • Y 3 is selected from N or CR Y3 ;
  • Y4 is selected from N or CR Y4 ;
  • R 2b and R 2b ' are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl;
  • R Y1 , R Y2 , R Y3 and R Y4 are independently selected from H, D, halogen, CN, OR a , NR b R c , C(O)OR a , C(O)NR b R c , S(O )R a , S(O) 2 R a , C 1-6 alkyl, C 1-6 haloalkyl, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5 -10-membered heteroaryl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl , 4-7 membered heterocyclic group, C 6-10 aryl group or 5-10 membered heteroaryl group.
  • the present invention provides the compound of the above formula (VI-2), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • X is CR 2b R 2b ';
  • R 2 is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • Y 1 is selected from N or CR Y1 ;
  • Y 2 is selected from N or CR Y2 ;
  • Y 3 is selected from N or CR Y3 ;
  • Y4 is selected from N or CR Y4 ;
  • R 2b and R 2b ' are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl;
  • R Y1 , R Y2 , R Y3 and R Y4 are independently selected from R' is selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, 5-6 membered heterocyclyl, phenyl or 5-6 membered heteroaryl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl Or 4-7 membered heterocyclyl.
  • the present invention provides the compound of the above formula (VI-2), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • X is CR 2b R 2b ';
  • R 2 is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • R 1b , R 1b ' and the carbon atoms to which they are connected form C O;
  • Y 1 is CR Y1 ;
  • Y 2 is CR Y2 ;
  • Y 3 is CR Y3 ;
  • Y 4 is CR Y4 ;
  • R 2b and R 2b ' are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl;
  • R Y1 , R Y2 , R Y3 and R Y4 are independently selected from H, D, halogen, CN, C 1-6 alkyl, C 1-6 haloalkyl, phenyl or 5-6 membered heteroaryl.
  • the invention provides the above-described compounds, or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates or solvents thereof compound, which has the following general formula:
  • X is CR 2b R 2b ';
  • R 2 is selected from C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl or 4-7 membered heterocyclyl;
  • Y 1 is CR Y1 ;
  • Y 2 is CR Y2 ;
  • Y 3 is CR Y3 ;
  • Y 4 is CR Y4 ;
  • R 2b and R 2b ' are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl;
  • RY1 , RY2 , RY3 and RY4 are independently selected from H, D, halogen, CN, OR a , NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkene base, C 2-6 alkynyl, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl Or 4-7 membered heterocyclyl.
  • the present invention provides the compound of the above formula (VI-2), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • X is CR 2b R 2b ';
  • R 2 is Me, Et or cyclopropyl
  • R 1b , R 1b ' and the carbon atoms to which they are connected form C O;
  • Y 1 is CR Y1 ;
  • Y 2 is CR Y2 ;
  • Y 3 is CR Y3 ;
  • Y 4 is CR Y4 ;
  • R 2b and R 2b ' are independently selected from H or D;
  • R Y1 is selected from H, D, Cl, F, CN, Me, CF 3 or OMe;
  • R Y2 is selected from H, D, Me, Et, iPr, CF 3 , cyclopropyl, OMe, OEt, OiPr, OCF 3 , O-cyclopropyl, prop-1-en-2-yl or phenyl;
  • R Y3 is selected from H, D, Cl, F, Me, Et, iPr, CF 3 , cyclopropyl, prop-1-en-2-yl, OMe or O-cyclopropyl;
  • R Y4 is selected from H, D or Me
  • the present invention provides the compound of the above formula (VI-2), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • X is CR 2b R 2b ';
  • R 2 is selected from C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl or 4-7 membered heterocyclyl;
  • Y 1 is CR Y1 ;
  • Y 2 is CR Y2 ;
  • Y 3 is CR Y3 ;
  • Y 4 is CR Y4 ;
  • R 2b and R 2b ' are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl;
  • R Y1 , R Y2 , R Y3 and R Y4 are independently selected from H, D, halogen, OR a , NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkene base, C 2-6 alkynyl, C 3-10 cycloalkyl, 4-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl;
  • R Y1 and R Y3 are not H or D;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl Or 4-7 membered heterocyclyl.
  • the present invention provides the compound of the above formula (VI-2), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • X is CR 2b R 2b ';
  • R 2 is selected from C 1-6 alkyl, C 1-6 haloalkyl or C 3-6 cycloalkyl;
  • Y 1 is CR Y1 ;
  • Y 2 is CR Y2 ;
  • Y 3 is CR Y3 ;
  • Y 4 is CR Y4 ;
  • R 2b and R 2b ' are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl;
  • R Y1 and R Y3 are independently selected from H, D, halogen, OR a , NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl , C 3-6 cycloalkyl or 4-7 membered heterocyclyl;
  • R Y2 and R Y4 are independently selected from H, D, OR a , NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R Y1 and R Y3 are not H or D;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl Or 4-7 membered heterocyclyl.
  • the present invention provides the compound of the above formula (VI-2), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • X is CR 2b R 2b ';
  • R 2 is Me, Et or cyclopropyl
  • R 1b , R 1b ' and the carbon atoms to which they are connected form C O;
  • Y 1 is CR Y1 ;
  • Y 2 is CR Y2 ;
  • Y 3 is CR Y3 ;
  • Y 4 is CR Y4 ;
  • R 2b and R 2b ' are independently selected from H or D;
  • R Y1 is selected from H, D, Cl, F, Me, CF 3 or OMe;
  • R Y2 is selected from H, D, Et, iPr, OiPr, O-cyclopropyl or prop-1-en-2-yl;
  • R Y3 is selected from H, D, Cl, F, Me, Et, iPr, CF 3 , cyclopropyl, prop-1-en-2-yl, OMe or O-cyclopropyl;
  • R Y4 is selected from H or D
  • At least one of R Y1 and R Y3 is not H or D.
  • the present invention provides the compound of the above formula (VI-2), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • X is CR 2b R 2b ';
  • R 2 is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • Y 1 is CR Y1 ;
  • Y 2 is CR Y2 ;
  • Y 3 is CR Y3 ;
  • Y 4 is CR Y4 ;
  • R 2b and R 2b ' are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl;
  • R Y1 and R Y3 are independently selected from H, D, halogen, OR a , NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl or 4-7 membered hetero Ring group;
  • R Y2 and R Y4 are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl;
  • R Y1 and R Y3 are not H or D;
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl Or 4-7 membered heterocyclyl.
  • the present invention provides the compound of the above formula (VI-2), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph thereof form, hydrate or solvate, wherein,
  • X is CR 2b R 2b ';
  • R 2 is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • R 1b , R 1b ' and the carbon atoms to which they are connected form C O;
  • Y 1 is CR Y1 ;
  • Y 2 is CR Y2 ;
  • Y 3 is CR Y3 ;
  • Y 4 is CR Y4 ;
  • R 2b and R 2b ' are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl;
  • R Y1 is selected from H, D, halogen, C 1-6 alkyl or C 1-6 haloalkyl;
  • R Y2 is selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl
  • R Y3 is selected from H, D, halogen, OR a , C 1-6 alkyl, C 1-6 haloalkyl or C 3-6 cycloalkyl;
  • R Y4 is selected from H or D
  • R Y1 and R Y3 are not H or D;
  • R a is selected from H, C 1-6 alkyl or C 1-6 haloalkyl.
  • the present invention provides the compound of the above formula (VI-2), or a pharmaceutically acceptable salt, isotope change thereof isomer, tautomer, stereoisomer, prodrug, polymorph, hydrate or solvate, wherein,
  • X is CR 2b R 2b ';
  • R 2 is Me
  • R 1b , R 1b ' and the carbon atoms to which they are connected form C O;
  • Y 1 is CR Y1 ;
  • Y 2 is CR Y2 ;
  • Y 3 is CR Y3 ;
  • Y 4 is CR Y4 ;
  • R 2b and R 2b ' are independently selected from H or D;
  • R Y1 is selected from H, D, Cl, F, Me or CF 3 ;
  • R Y2 is selected from H, D or Et;
  • R Y3 is selected from H, D, Cl, F, Me, Et, OMe or cyclopropyl
  • R Y4 is selected from H or D
  • At least one of R Y1 and R Y3 is not H or D.
  • the invention provides the above-described compounds, or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates or solvents thereof compound, which has the following general formula:
  • R 2 is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • R 1a is selected from H, D, halogen, CN, OR a or NR b R c ;
  • R 2a is selected from H, D, halogen or CN;
  • R 3a is selected from H or D
  • R 4a is selected from OR a or NR b R c ;
  • R 5a is selected from H, D or CN
  • R 3a , R 4a and the carbon atoms to which they are connected form a 4-6 membered heterocyclyl group or a 5-6 membered heteroaryl group, which is optionally substituted by 1, 2, 3 or 4 R'substituents;
  • R' is selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl
  • R a , R b and R c are independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 4-7 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, or R b and R c and the nitrogen atoms to which they are connected form a 4-7 membered heterocyclyl.
  • the present invention provides the above-mentioned compound of formula (VII), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph, Hydrate or solvate, wherein,
  • R 2 is selected from C 1-6 alkyl or C 1-6 haloalkyl
  • R 1a is halogen, preferably F
  • R 2a is selected from H, D or halogen
  • R 3a is H or D
  • R 4a is OR a ;
  • R 5a is CN
  • R 3a , R 4a and the carbon atoms to which they are attached form a furyl or dihydrofuryl group, which is optionally substituted by 1, 2, 3 or 4 R'substituents;
  • R' is selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl
  • R a is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 cycloalkyl or 4-7 membered heterocyclyl.
  • the present invention provides the above-mentioned compound of formula (VII), or a pharmaceutically acceptable salt, isotopic variant, tautomer, stereoisomer, prodrug, polymorph, Hydrate or solvate, wherein,
  • R 2 is selected from C 1-4 alkyl or C 1-4 haloalkyl
  • R 1a is halogen, preferably F
  • R 2a is selected from H, D or halogen
  • R 3a is H or D
  • R 4a is OR a ;
  • R 5a is CN
  • R a is selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl or 5-6 membered heterocyclyl .
  • the invention provides compounds, or pharmaceutically acceptable salts, isotopic variants, tautomers, stereoisomers, prodrugs, polymorphs, hydrates or solvates thereof Object, wherein said compound is selected from Table 1 and Table 2 below.
  • the compounds of the present invention may contain one or more asymmetric centers and thus may exist in multiple stereoisomeric forms, for example, enantiomeric and/or diastereomeric forms.
  • the compounds of the present invention may be individual enantiomers, diastereomers, or geometric isomers (e.g., cis and trans isomers), or may be in the form of mixtures of stereoisomers, Includes racemic mixtures and mixtures enriched in one or more stereoisomers.
  • the isomers may be separated from the mixture by methods known to those skilled in the art, including chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts; or the preferred isomers may be separated by Prepared by asymmetric synthesis.
  • HPLC high pressure liquid chromatography
  • Tautomers are functional group isomers produced by the rapid movement of an atom in a molecule between two positions.
  • a tautomer is a special functional group isomer.
  • a pair of tautomers can interact with each other. conversion, but usually one of the more stable isomers is its main form of existence. The most important examples are the enol and keto tautomers.
  • solvate refers to a form of a compound or a salt thereof that is combined with a solvent, usually formed by a solvolysis reaction. This physical association may include hydrogen bonding.
  • solvents include water, methanol, ethanol, acetic acid, DMSO, THF, ether, etc.
  • Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric and non-stoichiometric solvates. In some cases, the solvate will be capable of isolating, for example, when one or more solvent molecules are incorporated into the crystal lattice of the crystalline solid.
  • “Solvate” includes both solution solvates and isolable solvates. Representative solvates include hydrates, ethanolates, and methoxides.
  • hydrate refers to a compound combined with water. Typically, the ratio of the number of water molecules contained in a hydrate of a compound to the number of molecules of the compound in the hydrate is determined.
  • a hydrate of a compound may be represented, for example, by the general formula R ⁇ x H 2 O, where R is the compound and x is a number greater than zero.
  • a given compound may form more than one hydrate type, including, for example, monohydrate (x is 1), lower hydrate (x is a number greater than 0 and less than 1, for example, hemihydrate (R ⁇ 0.5H 2 O)) and polyhydrates (x is a number greater than 1, for example, dihydrate (R ⁇ 2H 2 O) and hexahydrate (R ⁇ 6H 2 O)).
  • monohydrate x is 1
  • lower hydrate x is a number greater than 0 and less than 1, for example, hemihydrate (R ⁇ 0.5H 2 O)
  • polyhydrates x is a number greater than 1, for example, dihydrate (R ⁇ 2H 2 O) and hexahydrate (R ⁇ 6H 2 O)
  • the compounds of the invention may be in amorphous or crystalline forms (polymorphs). Furthermore, the compounds of the present invention may exist in one or more crystalline forms. Accordingly, the present invention includes within its scope all amorphous or crystalline forms of the compounds of the invention.
  • polymorph refers to a crystalline form of a compound (or a salt, hydrate or solvate thereof) in a specific crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms often have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optoelectronic properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature, and other factors can lead to the dominance of one crystalline form. Various polymorphs of the compounds can be prepared by crystallization under different conditions.
  • the present invention also includes isotopically labeled compounds (isotopic variants) which are identical to those described in formula (I), except that one or more atoms are surrounded by atoms having an atomic mass or mass number different from that common in nature. replaced.
  • isotopes that may be incorporated into the compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, 18 respectively. O, 17 O, 31 P, 32 P, 35 S, 18 F and 36 Cl.
  • the isotope-labeled compounds of formula (A) of the present invention and their prodrugs can generally be prepared by replacing non-isotopes with readily available isotope-labeled reagents when performing the following processes and/or the processes disclosed in the Examples and Preparation Examples. Labeled reagents.
  • prodrugs are also included within the context of the present invention.
  • the term "prodrug” as used herein refers to a compound that is converted in the body to its active form having a medical effect, for example, by hydrolysis in the blood.
  • Pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems, Vol. 14 of A.C.S. Symposium Series, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and D. Fleisher, S. Ramon and H. Barbra "Improved oral drug delivery: solubility limitations overcome by the use of prodrugs", Advanced Drug Delivery Reviews (1996) 19 (2) 115-130, each introduced This article serves as a reference.
  • a prodrug is any covalently bonded compound of the invention that releases the parent compound in the body when administered to a patient.
  • Prodrugs are typically prepared by modifying functional groups in a manner such that the modification can be cleaved by conventional manipulations or in vivo to yield the parent compound.
  • Prodrugs include, for example, compounds of the invention in which a hydroxyl, amino or thiol group is bonded to any group that can be cleaved to form a hydroxyl, amino or thiol group when administered to a patient.
  • representative examples of prodrugs include, but are not limited to, acetate/amide, formate/amide and benzoate/amide derivatives of the hydroxyl, thiol and amino functionality of the compound of formula (A).
  • esters such as methyl ester, ethyl ester, etc. can be used.
  • the ester itself may be reactive and/or hydrolyzable under human body conditions.
  • Suitable pharmaceutically acceptable in vivo hydrolyzable ester groups include those that readily break down in the human body to release the parent acid or salt thereof.
  • the present invention also provides pharmaceutical preparations, comprising a therapeutically effective amount of a compound of formula (A) or a therapeutically acceptable salt thereof and a pharmaceutically acceptable carrier, diluent or excipient thereof. All these forms belong to the invention.
  • the invention provides pharmaceutical compositions comprising a compound of the invention (also referred to as an "active ingredient") and a pharmaceutically acceptable excipient.
  • the pharmaceutical compositions comprise an effective amount of a compound of the invention.
  • the pharmaceutical compositions comprise a therapeutically effective amount of a compound of the invention.
  • the pharmaceutical compositions comprise a prophylactically effective amount of a compound of the invention.
  • compositions of the present invention refer to non-toxic carriers, adjuvants or vehicles that do not destroy the pharmacological activity of the compounds with which they are formulated.
  • Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of the present invention include, but are not limited to, ion exchangers, aluminum oxide, aluminum stearate, lecithin, serum proteins (such as human serum albumin) protein), buffer substances (such as phosphate), glycine, sorbic acid, potassium sorbate, partial glyceride mixture of saturated vegetable fatty acids, water, salt or electrolyte (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate , sodium chloride, zinc salt, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylate, wax, polyethylene-polyoxypropylene- Block polymers, polyethylene glycols, and
  • Suitable formulations for administering the compounds of the invention will be apparent to those of ordinary skill in the art and include, for example, tablets, pills, capsules, suppositories, lozenges, lozenges, solutions (especially injections (subcutaneous, intravenous solution for intramuscular administration) and infusion (injection)), elixir, syrup, cachet, emulsion, inhalation or dispersible powder.
  • the content of one or more pharmaceutically active compounds should range from 0.1 to 90% by weight, preferably from 0.5 to 50% by weight of the composition as a whole, ie an amount sufficient to achieve the dosage ranges specified below. If necessary, the specified dose may be administered several times per day.
  • kits eg, pharmaceutical packaging.
  • Kits provided may include a compound of the invention, other therapeutic agents, and first and second containers (e.g., vials, ampoules, bottles, syringes, and/or dispersible packaging or other) containing the compounds of the invention, other therapeutic agents. suitable container).
  • provided kits may also optionally include a third container containing pharmaceutical excipients for diluting or suspending the compounds of the invention and/or other therapeutic agents.
  • the compound of the invention and the other therapeutic agent provided in the first container and the second container are combined to form a unit dosage form.
  • parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intraarticular administration, intraarterial administration, intrasynovial administration, and intrasternal administration.
  • intracerebrospinal membrane drug administration intralesional drug administration, and intracranial injection or infusion techniques.
  • an effective amount of a compound provided herein is administered.
  • the amount of compound actually administered can be determined by the physician depending on the circumstances, including the condition being treated, the route of administration chosen, the compound actually administered, the age, weight and response of the individual patient, the severity of the patient's symptoms, etc. .
  • a compound provided herein is administered to a subject at risk of developing the condition, typically on the advice of and under the supervision of a physician, at dosage levels as described above.
  • Subjects at risk of developing a particular condition generally include subjects with a family history of the condition or those who have been determined by genetic testing or screening to be particularly susceptible to developing the condition.
  • compositions provided herein can also be administered over a long period of time ("chronic administration").
  • Long-term administration refers to the administration of a compound or pharmaceutical composition thereof over a long period of time, for example, 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or administration may be continued indefinitely, For example, the remainder of the subject's life.
  • chronic administration is intended to provide a constant level of the compound in the blood over an extended period of time, eg, within a therapeutic window.
  • a pharmaceutical composition may be administered as a bolus injection, eg, in order to increase the concentration of the compound in the blood to an effective level.
  • the bolus dose depends on the target systemic levels of the active ingredient through the body, e.g., an intramuscular or subcutaneous bolus dose provides a slow release of the active ingredient, whereas a bolus dose delivered directly into the vein (e.g., via an IV drip) ) can be delivered more quickly, allowing the concentration of active ingredients in the blood to quickly increase to effective levels.
  • the pharmaceutical composition may be administered as a continuous infusion, for example, by IV infusion, thereby providing a steady-state concentration of the active ingredient in the subject's body. Additionally, in other embodiments, a bolus dose of the pharmaceutical composition may be administered first, followed by a continuous infusion.
  • Oral compositions may take the form of bulk liquid solutions or suspensions, or bulk powders. More typically, however, the compositions are provided in unit dosage form to facilitate precise dosing.
  • dosage unit form refers to physically discrete units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active material suitable to produce the desired therapeutic effect in association with a suitable pharmaceutical excipient.
  • Typical unit dosage forms include prefilled, premeasured ampoules or syringes for liquid compositions, or pills, tablets, capsules, and the like in the case of solid compositions.
  • the compound will generally be a minor component (from about 0.1 to about 50% by weight, or preferably from about 1 to about 40% by weight), with the remainder being various components useful in forming the desired administration form. carriers or excipients and processing aids.
  • a typical regimen is one to five oral doses per day, especially two to four oral doses, typically three oral doses.
  • each dose provides from about 0.01 to about 20 mg/kg of a compound of the invention, with preferred doses each providing from about 0.1 to about 10 mg/kg, especially from about 1 to about 5 mg/kg.
  • a transdermal dose is generally selected in an amount of about 0.01 to about 20% by weight, preferably about 0.1 to about 20% by weight, preferably about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
  • Injectable dose levels range from about 0.1 mg/kg/hour to at least 10 mg/kg/hour from about 1 to about 120 hours, especially from 24 to 96 hours. To achieve adequate steady state levels, a preload bolus of about 0.1 mg/kg to about 10 mg/kg or more may also be given. For human patients weighing 40 to 80 kg, the maximum total dose should not exceed approximately 2 g/day.
  • Liquid forms suitable for oral administration may include suitable aqueous or non-aqueous carriers as well as buffering agents, suspending and dispersing agents, coloring agents, flavoring agents, and the like.
  • Solid forms may include, for example, any of the following components, or compounds of similar nature: binders, for example, microcrystalline cellulose, tragacanth, or gelatin; excipients, for example, starch or lactose, disintegrants, For example, alginic acid, Primogel or corn starch; lubricant, such as magnesium stearate; glidant, such as colloidal silicon dioxide; sweetener, such as sucrose or saccharin; or flavoring agent Flavors such as peppermint, methyl salicylate or orange flavoring.
  • binders for example, microcrystalline cellulose, tragacanth, or gelatin
  • excipients for example, starch or lactose, disintegrants, For example, alginic acid, Primogel or corn starch
  • Injectable compositions are typically based on injectable sterile saline or phosphate buffered saline, or other injectable excipients known in the art. As stated previously, in such compositions the active compound is typically a minor component, often about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
  • Transdermal compositions are typically formulated as topical ointments or creams containing the active ingredients.
  • the active ingredients When formulated as an ointment, the active ingredients are typically combined with a paraffin or water-miscible ointment base.
  • the active ingredient may be formulated as a cream with, for example, an oil-in-water cream base.
  • Such transdermal formulations are well known in the art and often include other ingredients for promoting stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and components are included within the scope provided by this invention.
  • transdermal administration may be achieved using reservoir or porous membrane types, or a variety of solid matrix patches.
  • compositions for oral administration, injection or topical administration are merely representative.
  • Other materials and processing techniques are described in Part 8 of Remington's Pharmaceutical Sciences, 17th edition, 1985, Mack Publishing Company, Easton, Pennsylvania, which article is incorporated by reference.
  • the compounds of the present invention may also be administered in sustained release form or from a sustained release drug delivery system.
  • sustained release materials can be found in Remington's Pharmaceutical Sciences.
  • the invention also relates to pharmaceutically acceptable formulations of the compounds of the invention.
  • the formulation includes water.
  • the formulation contains a cyclodextrin derivative.
  • the most common cyclodextrins are ⁇ -, ⁇ - and ⁇ -cyclodextrins consisting of 6, 7 and 8 ⁇ -1,4-linked glucose units respectively, optionally including a or multiple substituents including, but not limited to: methylated, hydroxyalkylated, acylated, and sulfoalkyl ether substitutions.
  • the cyclodextrin is a sulfoalkyl ether beta-cyclodextrin, for example, sulfobutyl ether beta-cyclodextrin, also known as Captisol. See, for example, U.S. 5,376,645.
  • the formulation includes hexapropyl-beta-cyclodextrin (eg, in water, 10-50%).
  • MTA-synergistic PRMT5 inhibitors can provide therapeutic benefits to a large number of tumor patients.
  • the compounds of the present invention exert therapeutic effects by negatively regulating the activity of MTA-bound PRMT5 in tumor cells, especially against MTAP-deficient cells or various MTAP-related tumor cells.
  • MTA synergistic PRMT5 inhibitors of the invention can treat a variety of cancers, including but not limited to tumor types, such as malignant peripheral nerve sheath tumors, mesothelioma, glioblastoma multiforme, Pancreatic cancer, bile duct cancer, prostate cancer, breast cancer, brain cancer, skin cancer, cervical cancer, bladder cancer, astrocytoma, colorectal cancer, endometrial cancer, esophageal cancer, stomach cancer, thymoma, head and neck cancer, liver Cell carcinoma, laryngeal cancer, lung squamous cell carcinoma, lung adenocarcinoma, oral cancer, ovarian cancer, renal cancer and thyroid cancer, and sarcoma.
  • tumor types such as malignant peripheral nerve sheath tumors, mesothelioma, glioblastoma multiforme, Pancreatic cancer, bile duct cancer, prostate cancer, breast cancer, brain cancer, skin cancer, cervical cancer, bladder cancer,
  • these compounds are useful in the treatment of: Heart: sarcomas (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma, etc.), myxoma, rhabdomyomas, fibromas, lipomas and teratomas; Lung: bronchial carcinoma ( Squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma, etc.), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; gastrointestinal tract: esophagus (Squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma, etc.), stomach (tumor, lymphoma, leiomyosarcoma, etc.), pancreas (ductal adenodeno
  • the MTA synergistic PRMT5 inhibitor of the present invention can be combined with other drugs to treat cancer, and contains at least one target drug/cell activity regulator, including CDK4/6 inhibitors, MAT2A inhibitors, MAPK1/MAPK3 inhibitors, Type I PRMT inhibitor, EGFR inhibitor, SHP2 inhibitor, pan-KRAS inhibitor, KRASG12C inhibitor, RAF inhibitor, MEK inhibitor, ERK inhibitor, Bcl-2 inhibitor, SOS1 inhibitor, PARP inhibitor, MALT1 inhibitor Agent, MALT2 inhibitor, BTK inhibitor, PI3K inhibitor, AKT inhibitor, FGFR inhibitor, DNA methyltransferase (DNMT) inhibitor, EZH1/2 inhibitor, EZH2 inhibitor, Menin-MLL inhibitor, IDH1 Inhibitors, IDH2 inhibitors, IDH1/2 inhibitors, chemotherapy drugs, radiotherapy, STING agonists or immune checkpoint inhibitors/modulators, etc.
  • target drug/cell activity regulator including CDK4/6 inhibitors, MAT2A inhibitors, MAPK1/MAPK3 inhibitor
  • n-butyllithium (2.4M, 21.2mL) was slowly added dropwise to a -78°C tetrahydrofuran (80mL) solution containing 2,2,6,6-tetramethylpiperidine (7.48g). After that, the temperature of the reaction solution was slowly raised to 0°C, and then cooled to -78°C. A solution of 6-chloronicotinic acid (2g) in tetrahydrofuran (10 mL) was slowly added dropwise to the above reaction solution, and stirred at this temperature for 90 minutes.
  • Tetrakis(triphenylphosphine)palladium (141 mg), 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2- (1H-pyrazole) (507 mg) and potassium carbonate (1.1 g) were added to a mixed solution of 2,6-dichloronicotinic acid methyl ester (500 mg) in tetrahydrofuran (15 mL) and water (3 mL). Under nitrogen protection, stir at 80°C for 5 hours.
  • Tributyl(1-ethoxyvinyl)tin (3.9g) and bis(triphenylphosphine)palladium dichloride (0.63g) were added to ethyl 2,4-dichloropyrimidine-5-carboxylate ( 2g) in a solution of N,N-dimethylformamide (20 mL), and stirred at 70°C for 2 hours under nitrogen protection.
  • the reaction solution was poured into ice-water (100 mL) and extracted with ethyl acetate (50 mL*3).
  • the combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure.
  • N-bromosuccinimide (453.3 mg) was added to a solution of 3-2 (0.7 g) in tetrahydrofuran (7.5 mL) and water (0.5 mL), and the mixture was stirred at room temperature for 16 hours.
  • MS: M+H + 352.8,354.7.
  • MS: M+H + 311.
  • Methyl 2-bromomethylbenzoate (2.36g) and potassium carbonate (2.84g) were added to a solution of 1-methyl-5-aminopyrazole (1g) in N,N-dimethylformamide (10mL) medium, stir at room temperature for 2 hours.
  • MS: M+H + 176.
  • n-butyllithium (2.4M, 0.65mL) was slowly added dropwise to a solution of 6-9 (300mg) in anhydrous tetrahydrofuran (5mL). solution, stir at -78°C for 0.5 hours, add tributyltin chloride (1g) dropwise, slowly rise to room temperature, and stir for 2 hours.
  • n-butyllithium (2.8mL, 1M) was slowly added dropwise to a solution of 5-bromo-3-methylisothiazole (1.0g) in tetrahydrofuran (50mL), and the temperature was controlled to be lower than -70°C and stirred 10 minutes.
  • Methyl 2,6-dichloronicotinate (724.0 mg) and bis(triphenylphosphine)palladium dichloride (235.0 mg) were added to N,N-dimethylmethyl of compound 8-1 (1.3 g).
  • amide (15 mL) solution and stirred at 80°C for 2 hours. Cool to room temperature, add water (100 mL), extract with ethyl acetate (50 mL ⁇ 3), wash the organic phase with saturated sodium chloride solution (50 mL), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • Tributyl(1-ethoxyethylene)tin (961.0 mg) and tetrakis(triphenylphosphine)palladium (236.0 mg) were added to compound 8-2 (550.0 mg) in N,N-dimethylformamide (15 mL) solution and stirred at 100°C for 2 hours. Cool to room temperature, add water (100 mL), extract with ethyl acetate (50 mL ⁇ 3), wash the organic phase with saturated sodium chloride solution (50 mL), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • Tributyl(1-ethoxyethylene)tin (3.9g) and bistriphenylphosphine palladium dichloride (0.6g) were added to ethyl 2,4-dichloro-5-pyrimidinecarboxylate (2.0g) of N,N-dimethylformamide (20 mL) and stirred at 70°C for 16 hours.
  • the reaction solution was poured into ice water (100 mL), and extracted with ethyl acetate (50 mL ⁇ 3). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure.
  • 9-1 (200.0 mg), tetraphenylphosphine palladium (84.0 mg) and sodium carbonate (155.0 mg) were added to the solution of 9-2 (298.0 mg) in dioxane (4 mL) and water (1 mL), Stir at 100°C for 2 hours under nitrogen protection. Cool to room temperature, add water (15 mL), extract with ethyl acetate (10 mL ⁇ 3), wash the organic phase with saturated sodium chloride solution (10 mL), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • Azidotrimethylsilane (71.4 mg) was added to a solution of potassium carbonate (150.0 mg) and 9-4 (150.0 mg) in acetonitrile (2 mL) at 0° C., and the mixture was stirred at 0° C. for 1 hour. It was raised to room temperature, filtered, and the filtrate was concentrated under reduced pressure to obtain a white solid 9-5 (70.0 mg). The crude product was directly used in the next reaction without purification. LC-MS m/z(ESI):447.1[M+H] + .
  • Triphenylphosphine (63.3 mg) was added to 9-6 (50.0 mg) in tetrahydrofuran (1.5 mL) and water (0.3 mL), and the mixture was stirred at 50° C. for 2 hours. Cool to room temperature. Concentrate under reduced pressure, and the residue is subjected to high performance liquid chromatography (column: -Xbridge-C18 150 x 19mm, 5um; mobile phase: [H 2 O (0.05% NH 3 )-ACN]; B%: 2%-20%, 20 min) to obtain white solid 9 (11.0 mg, yield 35.1%). LC-MS m/z(ESI):389.2[M+H] + .
  • Methyl 2-bromomethylbenzoate (1.5g) and potassium carbonate (3.0g) were added to N,N-dimethylformate of compound 3-methyliso-4-aminothiazole hydrochloride (1.0g).
  • amide solution (20 mL) stirred at 80°C for 5 hours. Cool to room temperature, add water (50 mL), extract with ethyl acetate (50 mL ⁇ 3), wash the organic phase with saturated sodium chloride solution (50 mL), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • n-butyllithium (1.8 mL, 2.5 M) was slowly dropped into the tetrahydrofuran solution (20 mL) of 10-1 (1.0 g), and the temperature was controlled to be lower than -70° C. and stirred for 10 minutes.
  • a solution (10 mL) of tributyltin chloride (2.1 g) in tetrahydrofuran was slowly added to the reaction solution, and the temperature was controlled to be lower than -70°C and stirred for 2 hours.
  • Tributyl(1-ethoxyethylene)tin (2.0g) and bis(triphenylphosphine)palladium dichloride (200.0mg) were added to ethyl 2,4-dichloro-5-pyrimidinecarboxylate (1.0 g) in N,N-dimethylformamide solution (30 mL), stir at 80°C for 2 hours. Cool to room temperature, add water (50 mL), extract with ethyl acetate (50 mL ⁇ 3), wash the organic phase with saturated sodium chloride solution (50 mL), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • Tributyl(1-ethoxyethylene)tin (1.2g) and tetrakis(triphenylphosphine)palladium (200.0mg) were added to N,N-dimethylformamide of compound 12-1 (400.0mg) (5 mL) solution and stirred at 100°C for 2 hours. Cool to room temperature, concentrate under reduced pressure, and wash the residue with diethyl ether (30 mL) to obtain black oil 12-2 (600.0 mg). The crude product is directly used in the next reaction. LC-MS m/z(ESI):288.2[M+H] + .
  • Methyl 2,6-dichloronicotinate (700.0 mg) and bis(triphenylphosphine)palladium dichloride (200.0 mg) were added to N,N-dimethylmethane of compound 10-2 (800.0 mg).
  • amide solution (15 mL), stirred at 80°C for 2 hours. Cool to room temperature, add water (50 mL), extract with ethyl acetate (50 mL ⁇ 3), wash the organic phase with saturated sodium chloride solution (50 mL), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • Tributyl(1-ethoxyethylene)tin 500.0 mg
  • tetrakis(triphenylphosphine)palladium (120.0 mg) were added to N,N-dimethylformamide of compound 13-1 (300.0 mg) (10 mL), stir at 100°C for 2 hours. Cool to room temperature, add water (50 mL), extract with ethyl acetate (50 mL ⁇ 3), wash the organic phase with saturated sodium chloride solution (50 mL), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • Water (50 mL) was added, extracted with ethyl acetate (50 mL ⁇ 3), the organic phase was washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure.
  • Tributyl(1-ethoxyethylene)tin (16.0g) and bis(triphenylphosphine)palladium dichloride (3.1g) were added to methyl 2-chloro-6-methoxynicotinic acid (8.0 g) in N,N-dimethylformamide (200 mL) solution, stir at 100°C for 5 hours. Cool to room temperature, add water (200 mL), extract with ethyl acetate (200 mL ⁇ 3), wash the organic phase with saturated sodium chloride solution (200 mL), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • Methyl 2-(2-bromoethyl)benzoate (1.0g) and potassium carbonate (1.5g) were added to the compound 3-methyl-4-aminoisothiazole hydrochloride (500mg) in N,N-di into methylformamide solution (20 mL) and stirred at 80°C for 5 hours. Cool to room temperature, add water (50 mL), extract with ethyl acetate (50 mL ⁇ 3), wash the organic phase with saturated sodium chloride solution (50 mL), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • n-butyllithium (0.4 mL, 2.5 M) was slowly added dropwise to the tetrahydrofuran solution (20 mL) of 14-9 (200.0 mg), and the temperature was controlled to be lower than -70°C and stirred for 10 minutes.
  • Example 11 or 14 The compounds listed below were synthesized through the same synthesis strategy of Example 11 or 14:
  • test concentration of the test compound and positive control compound is starting from 10 ⁇ M and diluted 3 times to 10 concentrations. Dilute the test compound and positive control with DMSO to 100 ⁇ the concentration to be tested (i.e. 1mM), then dilute the test compound 3 times to 10 concentrations, and then dilute it 20 times with 1 ⁇ reaction solution (at this time DMSO (concentration is 5%), and finally add 5 ⁇ L of each compound to the 384-well reaction plate for testing, and set up multiple wells for testing for each compound. Transfer 5 ⁇ L of 5% DMSO to the Max and Min wells.
  • step (7) Add 10 ⁇ L of the mixed solution of step (6) at 2.5 times the final concentration to start the reaction.
  • Inhibition% 100-(RLU-Mean(NC))/(Mean(PC)-Mean(NC))*100
  • RLU Luminescence value of the sample
  • Dose-response curves were fitted using GraphPad Prism 5 software, and IC50 was calculated using the ‘log[inhibitor]vs.response-variable slope’ program.
  • test concentration of the test compound and positive control compound is starting from 10 ⁇ M and diluted 3 times to 10 concentrations. Dilute the test compound and positive control with DMSO to 100 ⁇ the concentration to be tested (i.e. 1mM), then dilute the test compound 3 times to 10 concentrations, and then dilute it 20 times with 1 ⁇ reaction solution (at this time DMSO (concentration is 5%), and finally add 5 ⁇ L of each compound to the 384-well reaction plate for testing, and set up multiple wells for testing for each compound. Transfer 5 ⁇ L of 5% DMSO to the Max and Min wells.
  • step (7) Add 10 ⁇ L of the mixed solution of step (6) at 2.5 times the final concentration to start the reaction.
  • Inhibition% 100-(RLU-Mean(NC))/(Mean(PC)-Mean(NC))*100
  • RLU Luminescence value of the sample
  • Dose-response curves were fitted using GraphPad Prism 5 software, and IC50 was calculated using the ‘log[inhibitor]vs.response-variable slope’ procedure.
  • the compound of the present invention is an MTA-synergized PRMT5 inhibitor with significant inhibitory effect.
  • Caco-2 cells were purchased from the American Type Culture Collection (ATCC).
  • FBS medium was purchased from Sigma, DMEM was purchased from Corning Company (Cambridge, MA), non-essential amino acids (NEAA), Hank’s balanced salt solution (HBSS) and trypsin/EDTA were purchased from Thermo Fisher. Penicillin and streptomycin were purchased from Soleba.
  • HTS-96-well Transwell plate and other sterile consumables were purchased from Corning Company.
  • Millicell resistance measurement system was purchased from Millipore. Vision was purchased from Nexcelom Bioscience. Infinite200PRO microplate reader was purchased from Tecan. The MTS2/4 orbital shaker was purchased from IKA Labortechnik.
  • Caco-2 was cultured in cell culture flasks.
  • the incubator was set to 37°C, 5% CO 2 , and guaranteed relative humidity of 95%. Cells that reach 70-90% confluence can be used to seed Transwells.
  • the process of replacing the culture medium is as follows. Separate the Transwell chamber from the receiving plate. Discard the medium in the receiving plate first and then discard the medium in the Transwell chamber. Finally, add 75 ⁇ L of fresh culture medium to each chamber, and add 25 mL of fresh culture to the receiving plate. base.
  • Caco-2 After 14-18 days of culture, Caco-2 is fully confluent and completes differentiation. At this time, it can be applied to penetration testing.
  • V A is the volume of the receiving end solution (Ap ⁇ Bl is 0.235mL, Bl ⁇ Ap is 0.075mL), Area is the Transwell-96-well plate membrane area (0.143cm 2 ); time is the incubation time (unit: s ), [drug] acceptor is the drug concentration at the receiving end (relative value, use the corresponding Area Ratio to participate in the calculation), [drug] initial, donor is the initial drug concentration at the dosing end (relative value, use the corresponding Area Ratio to participate in the calculation) .
  • the efflux rate is calculated using the following formula:
  • P app (BA) is the apparent permeability coefficient from the basal end to the top
  • P app (AB) is the apparent permeability coefficient from the top to the basal end.
  • MRTX1719 has the following structure:
  • Liver cells are stored in liquid nitrogen. See the table below for detailed information.
  • Hepatocyte recovery solution includes Williams’ Medium E, Isotonic Percoll, DPBS, GlutaMAX, HEPES, FBS, human recombinant insulin and dexamethasone. Mix 49.5mL Williams’ Medium E and 0.5mL GlutaMAX as incubation solution.
  • the compound of the present invention is metabolically stable in mouse liver cells in vitro.
  • HCT116 MTAP Deletion cells were from Pharmaron (China); HCT116 cells were from ATCC (USA); McCoy's 5A medium was purchased from Invitrogen (USA); penicillin-streptomycin and fetal calf serum were purchased from Gibco (USA); The 384-well plate was purchased from PerkinElmer Company (USA); Cell-Titer Glo reagent was purchased from Promega Company (USA).
  • Cells were cultured in strict accordance with ATCC requirements. Transfer the cell culture medium (McCoy's 5A medium + 10% fetal bovine serum + 1% penicillin + streptomycin) to a 15 mL centrifuge tube, and centrifuge at 1000 rpm for 5 minutes. Remove the supernatant, resuspend the cells in complete culture medium, inoculate them into a culture dish at the required density, and place them in an incubator at 37°C, 95% humid air, and 5% carbon dioxide for culture. Depending on the cell growth, every 2-3 Replenish the culture medium once a day or perform passage.
  • McCoy's 5A medium + 10% fetal bovine serum + 1% penicillin + streptomycin to a 15 mL centrifuge tube, and centrifuge at 1000 rpm for 5 minutes. Remove the supernatant, resuspend the cells in complete culture medium, inoculate them into a culture dish at the required density, and place them in an incubator at 37°C,
  • the compounds of the present invention also have selectivity against mutant HCT116 cells.
  • the GI 50 of compounds 71 and 72 against HCT116 MTAP Deletion is ⁇ 0.1 ⁇ M, but the GI 50 against HCT116 is 1-10 ⁇ M, and the selectivity is more than 10 times.
  • HCT116 MTAP Deletion cells are from Horizon; HCT116 cells are from ATCC; RPMI-1640 culture medium was purchased from ATCC Company (USA); penicillin-streptomycin was purchased from Gibco Company (USA); fetal bovine serum was purchased from ExCell Company; 96-well The plate was purchased from Corning Company (USA); Symmetric Di-Methyl Arginine Motif [sdme-RG] antibody was purchased from CST (USA); IRDye 800CW Goat anti-Rabbit IgG secondary antibody was purchased from LI-COR (USA), and CellTag 700stain was purchased from LI-COR (USA).
  • Cells were cultured in strict accordance with ATCC requirements. Transfer the cell culture medium (RPMI-1640 medium + 10% serum + 1% penicillin + streptomycin) to a 15 mL centrifuge tube, and centrifuge at 1000 rpm for 5 min. Remove the supernatant, resuspend the cells in complete culture medium, inoculate them into a culture dish at the required density, and place them in an incubator at 37°C, 95% humid air, and 5% carbon dioxide for culture. Depending on the cell growth, every 2-3 Replenish the culture medium once a day or perform passage.
  • RPMI-1640 medium + 10% serum + 1% penicillin + streptomycin to a 15 mL centrifuge tube, and centrifuge at 1000 rpm for 5 min. Remove the supernatant, resuspend the cells in complete culture medium, inoculate them into a culture dish at the required density, and place them in an incubator at 37°C, 95% humid air, and 5% carbon dioxide for culture. Depending on the
  • inhibition rate percentage 100 - (signal value drug treatment group - signal value background value ) / (signal value 0.5% DMSO treatment group - signal value background value ) ⁇ 100.
  • LU99 cells were obtained from JCRB (Japan); RPMI-1640 and 0.25% trypsin-EDTA were purchased from Gibco (USA); fetal bovine serum was purchased from Dongling (China); PBS was purchased from Hyclone (USA); penicillin -Streptomycin was purchased from Melun Biotech (China); Matrigel was purchased from Corning (USA).
  • mice BALB/c Nude mice, female, 6-8 weeks old, weighing 18-22 grams. The animals were purchased from Beijing Vitong Lever Experimental Animal Technology Co., Ltd. The mice were raised in an SPF-level environment. Each cage has separate ventilation and exhaust, and all animals have free access to standard certified commercial laboratory diet and free drinking water.
  • the dosage of compound 72 is 30mpk, PO, once daily (QD) ⁇ 3 weeks. 6 mice per group.
  • Tumor diameter was measured twice weekly using vernier calipers.
  • the tumor inhibitory effect of the compound was evaluated by the tumor growth inhibition rate TGI (%).
  • TGI (%) [(1-(average tumor volume at the end of treatment in a certain treatment group - average tumor volume at the beginning of treatment in the treatment group))/(average tumor volume at the end of treatment in the solvent control group - start of treatment in the solvent control group) average tumor volume)] ⁇ 100%.

Abstract

本发明提供了一类式(I)所示的PRMT5抑制剂,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物。本发明还提供了所述化合物的制备方法、包含所述化合物的药物组合物,以及所述化合物在预防和治疗癌症中的作用。

Description

稠和哒嗪酮化合物作为PRMT5抑制剂 技术领域
本发明涉及一类新的PRMT5抑制剂,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物。本发明还涉及所述化合物的制备方法、包含所述化合物的药物组合物,以及所述化合物在预防和治疗PRMT5介导的疾病如癌症中的作用。
背景技术
蛋白质精氨酸甲基转移酶5(Protein arginine methyltransferase 5,PRMT5)属于二型PRMT,是一类S-腺苷甲硫氨酸(SAM)依赖性甲基转移酶,主要负责将SAM的甲基对称性地转移到组蛋白或其他蛋白质的精氨酸残基末端的胍基氮原子上。在体内,PRMT5与MEP50(methylosome protein 50)形成复合物,才能识别底物并进行定位,同时复合物形态也是PRMT5催化组蛋白2A和组蛋白4甲基转移活性所必需的。PRMT5是一种通用的转录抑制因子,可以调控基因转录和蛋白修饰的过程。同时,PRMT5在肿瘤细胞的增殖、分化、凋亡中发挥着重要功能,是极具潜力的肿瘤治疗靶点。然而,PRMT5也是正常细胞所必需的基因,PRMT5敲除和siRNA敲降研究表明,在正常组织中抑制PRMT5的活性可能引起血小板减少、***、骨骼肌损失、心肌肥厚等诸多毒副作用。
2016年,多个独立研究团队报道MTAP(甲硫腺苷磷酸化酶)缺失型肿瘤细胞增加对PRMT5活性的依赖。MTAP的缺失会上调细胞内MTA(甲硫腺苷)的水平,而MTA同时也是天然的PRMT5抑制剂。因此,MTAP缺失型肿瘤细胞会增加对PRMT5活性的依赖,这为靶向MTAP null肿瘤细胞,即靶向PRMT5/MTA复合物,同时不影响MTAP WT细胞提供了可能。
在研PRMT5抑制剂的作用机理包括SAM竞争性抑制剂、底物竞争性抑制剂、SAM和底物双重竞争性抑制剂及MTA协同的抑制剂。非MTA协同的PRMT5抑制剂无差别的抑制PRMT5的活性,已发现剂量限制性的血小板减少、贫血、中性粒细胞减少等毒副作用。其中,MTA协同的抑制剂靶向PRMT5/MTA复合物,有望在抑制MTAP缺失型肿瘤细胞的同时消除对MTAP WT细胞的影响,可以改善治疗窗口。
因此,开发靶向PRMT5/MTA复合物的PRMT5抑制剂将具有重要的临床应用价值,本领域对于该抑制剂存在需求。
发明内容
本发明以PRMT5/MTA复合物作为靶点,研发了一类新的小分子抑制剂,可用于治疗各种癌症。
本发明化合物靶向PRMT5/MTA复合物,是MTA协同的PRMT5抑制剂,具有优异的PRMT5/MTA抑制活性,并对PRMT5/SAM复合物具有明显的选择性。同时,本发明化合物具有良好的ADMET(吸收-分布-代谢-***-毒性)性质。
在一个方面,本发明提供了式(I)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立 体异构体、前药、多晶型、水合物或溶剂合物:
其中:
Z1、Z2和Z3独立地选自N、CD或CH,并且其中至少一个为N;
环A为苯基或5-6元杂芳基;
R1选自H、D、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
R2选自H、D、-C0-6亚烷基-卤素、-C0-6亚烷基-CN、-C0-6亚烷基-ORa、-C0-6亚烷基-NRbRc、C1-6烷基、C1-6卤代烷基、C3-8环烷基或4-7元杂环基;
m=0、1、2或3;
其中R选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O、C=S、C3-8环烷基或4-7元杂环基;或者相邻原子上的两个R与所述原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3或4个R’取代基取代;
R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,优选H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1个或多个D、卤素、C1-6烷基或C1-6卤代烷基取代;
Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基;
其中环A、R1、R2、R、R’、Ra、Rb和Rc中的各基团可任选地被D取代,直至完全氘代。
在另一个方面,本发明提供了式(I)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物:
其中:
Z1、Z2和Z3独立地选自N、CD或CH,并且其中至少一个为N;
环A为苯基或5-6元杂芳基;
R1选自H、D、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
R2选自H、D、-C0-6亚烷基-卤素、-C0-6亚烷基-CN、-C0-6亚烷基-ORa、-C0-6亚烷基-NRbRc、C1-6烷基、C1-6卤代烷基、C3-8环烷基或4-7元杂环基;
m=0、1、2或3;
其中R选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O、C=S、C3-8环烷基或4-7元杂环基;或者相邻原子上的两个R与所述原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3或4个R’取代基取代;
R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,优选H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基或4-7元杂环基;
Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基;
其中环A、R1、R2、R、R’、Ra、Rb和Rc中的各基团可任选地被D取代,直至完全氘代。
在另一个方面,本发明提供了一种药物组合物,所述药物组合物含有本发明化合物,和任选地药学上可接受的赋形剂,例如载体、佐剂或媒介物。
在另一个方面,本发明提供了含有本发明化合物和药学上可接受的赋形剂的药物组合物,其还含有其它治疗剂,例如选自以下靶点药物/细胞活性调节剂,包括CDK4/6抑制剂,MAT2A抑制剂,MAPK1/MAPK3抑制剂,Type I PRMT抑制剂,EGFR抑制剂,SHP2抑制剂,泛KRAS抑制剂,KRASG12C抑制剂,RAF抑制剂,MEK抑制剂,ERK抑制剂,Bcl-2抑制剂,SOS1抑制剂,PARP抑制剂,MALT1抑制剂,MALT2抑制剂,BTK抑制剂,PI3K抑制剂,AKT抑制剂,FGFR抑制剂,DNA甲基转移酶(DNMT)抑制剂,EZH1/2抑制剂,EZH2抑制剂,Menin-MLL抑制剂,IDH1抑制剂,IDH2抑制剂,IDH1/2抑制剂,化疗药物,放射疗法,STING激动剂或免疫检查点抑制剂/调节剂。
在另一个方面,本发明提供了含有本发明化合物和药学上可接受的赋形剂的药物组合物,其还含有其它治疗剂。
在另一个方面,本发明提供了本发明化合物在制备用于治疗和/或预防PRMT5介导的疾病的药物中的用途。
在另一个方面,本发明提供了在受试者中治疗和/或预防PRMT5介导的疾病的方法,包括向所述受试者给药本发明化合物或本发明药物组合物。
在另一个方面,本发明提供了本发明化合物或本发明药物组合物,其用于治疗和/或预防PRMT5介导的疾病。
在具体实施方案中,本发明用于治疗和/或预防癌症。在另一具体实施方案中,本发明用于治疗和/或预防以下癌症:心脏:肉瘤(血管肉瘤、纤维肉瘤、横纹肌肉瘤、脂肪肉瘤)、粘液瘤、横纹肌瘤、纤维瘤、脂肪瘤和畸胎瘤;肺:支气管癌(鳞状细胞、未分化小细胞、未分化大细胞、腺癌)、肺泡(细支气管)癌、支气管腺瘤、肉瘤、淋巴瘤、软骨瘤错构瘤、间皮瘤;胃肠道:食管(鳞状细胞癌、腺癌、平滑肌肉瘤、淋巴瘤)、胃(癌、淋巴瘤、平滑肌肉瘤)、胰腺(导管腺癌、胰岛素瘤、胰高血糖素瘤、胃泌素瘤、类癌瘤、血管瘤)、小肠(腺癌、淋巴瘤、类癌瘤)、卡波西肉瘤、平滑肌瘤、血管瘤、脂肪瘤、神经纤维瘤、纤维瘤)、大肠(腺癌、管状腺瘤、绒毛状腺瘤、错构瘤、平滑肌瘤);泌尿生殖道:肾脏(腺癌、威尔姆氏瘤(肾母细胞瘤)、淋巴瘤、白血病)、膀胱和尿道(鳞状细胞癌、移行细胞癌、腺癌)、***(腺癌、肉瘤)、睾丸(***瘤、畸胎瘤、胚胎癌、畸胎癌)、绒毛膜癌、肉瘤、***癌、纤维瘤、纤维腺瘤、腺瘤样肿瘤、脂肪瘤);肝脏:肝细胞瘤(肝细胞癌)、胆管癌、肝母细胞瘤、血管肉瘤、肝细胞腺瘤、血管瘤;胆道:胆囊癌、壶腹癌、胆管癌;骨:成骨肉瘤(osteosarcoma)、纤维肉瘤、恶性纤维组织细胞瘤、软骨肉瘤、尤文氏肉瘤、恶性淋巴瘤(网状细胞肉瘤)、多发性骨髓瘤、恶性巨细胞瘤脊索瘤、骨慢性外生骨疣(osteocartilaginous exostoses)、良性软骨瘤、软骨母细胞瘤、纤维软骨瘤、类骨质骨瘤和巨细胞瘤;神经***:颅骨(骨瘤、血管瘤、肉芽肿、黄色瘤、变形性骨炎)、脑膜(脑膜瘤、脑膜肉瘤、神经胶质瘤)、脑(星形细胞瘤、髓母细胞瘤、神经胶质瘤、室管膜瘤、生殖细胞瘤(松果体瘤)、多形性胶质母细胞瘤、少突胶质细胞瘤、神经鞘瘤、视网膜母细胞瘤、先天性肿瘤)、脊髓神经纤维瘤、脑膜瘤、神经胶质瘤、肉瘤);妇科:子宫(子宫内膜癌(浆液性囊腺癌、粘液性囊腺癌、未分类癌)、颗粒鞘细胞瘤、支持***瘤、无性细胞瘤、恶性畸胎瘤)、外阴(鳞状细胞癌、上皮内癌、腺癌、纤维肉瘤、黑色素瘤)、***(透明细胞癌、鳞状细胞癌、葡萄样肉瘤(胚胎性横纹肌肉瘤)、输卵管(癌);血液学:血液(髓性白血病(急性和慢性)、急性淋巴细胞白血病、慢性淋巴细胞性白血病、骨髓增生性疾病、多发性骨髓瘤、骨髓增生异常综合征)、霍奇金病、非霍奇金淋巴瘤(恶性淋巴瘤);皮肤:恶性黑色素瘤、基底细胞癌、鳞状细胞癌、卡波西肉瘤、痣发育不良痣、脂肪瘤、血管瘤、皮肤纤维瘤、瘢痕疙瘩、牛皮癣;和肾上腺:成神经细胞瘤;尤其是MTAP相关癌症,例如肝细胞癌、乳腺癌、皮肤癌、膀胱癌、肝癌、胰腺癌或头颈癌。
在另一具体实施方案中,本发明所述PRMT5介导的疾病选自以下癌症:恶性周围神经鞘瘤、间 皮瘤、多形性胶质母细胞瘤、胰腺癌、胆管癌、***癌、乳腺癌、脑癌、皮肤癌、***、膀胱癌、星形细胞瘤、结肠直肠癌、子宫内膜癌、食道癌、胃癌、胸腺瘤、头颈癌、肝细胞癌、喉癌、肺鳞癌、肺腺癌、口腔癌、卵巢癌、肾癌和甲状腺癌以及肉瘤。
在另一具体实施方案中,本发明所述PRMT5介导的疾病选自MTAP相关癌症,例如肝细胞癌、乳腺癌、皮肤癌、膀胱癌、肝癌、胰腺癌或头颈癌。
定义
化学定义
下面更详细地描述具体官能团和化学术语的定义。
当列出数值范围时,既定包括每个值和在所述范围内的子范围。例如“C1-6烷基”包括C1、C2、C3、C4、C5、C6、C1-6、C1-5、C1-4、C1-3、C1-2、C2-6、C2-5、C2-4、C2-3、C3-6、C3-5、C3-4、C4-6、C4-5和C5-6烷基。
“C1-6烷基”是指具有1至6个碳原子的直链或支链饱和烃基团。在一些实施方案中,C1-4烷基和C1-2烷基是优选的。C1-6烷基的例子包括:甲基(C1)、乙基(C2)、正丙基(C3)、异丙基(C3)、正丁基(C4)、叔丁基(C4)、仲丁基(C4)、异丁基(C4)、正戊基(C5)、3-戊基(C5)、戊基(C5)、新戊基(C5)、3-甲基-2-丁基(C5)、叔戊基(C5)和正己基(C6)。术语“C1-6烷基”还包括杂烷基,其中一或多个(例如,1、2、3或4个)碳原子被杂原子(例如,氧、硫、氮、硼、硅、磷)替代。烷基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。常规烷基缩写包括:Me(-CH3)、Et(-CH2CH3)、iPr(-CH(CH3)2)、nPr(-CH2CH2CH3)、n-Bu(-CH2CH2CH2CH3)或i-Bu(-CH2CH(CH3)2)。
“C2-6烯基”是指具有2至6个碳原子和至少一个碳碳双键的直链或支链烃基团。在一些实施方案中,C2-4烯基是优选的。C2-6烯基的例子包括:乙烯基(C2)、1-丙烯基(C3)、2-丙烯基(C3)、1-丁烯基(C4)、2-丁烯基(C4)、丁二烯基(C4)、戊烯基(C5)、戊二烯基(C5)、己烯基(C6),等等。术语“C2-6烯基”还包括杂烯基,其中一或多个(例如,1、2、3或4个)碳原子被杂原子(例如,氧、硫、氮、硼、硅、磷)替代。烯基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“C2-6炔基”是指具有2至6个碳原子、至少一个碳-碳叁键以及任选地一个或多个碳-碳双键的直链或支链烃基团。在一些实施方案中,C2-4炔基是优选的。C2-6炔基的例子包括但不限于:乙炔基(C2)、1-丙炔基(C3)、2-丙炔基(C3)、1-丁炔基(C4)、2-丁炔基(C4),戊炔基(C5)、己炔基(C6),等等。术语“C2- 6炔基”还包括杂炔基,其中一或多个(例如,1、2、3或4个)碳原子被杂原子(例如,氧、硫、氮、硼、硅、磷)替代。炔基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“C1-6亚烷基”是指除去C1-6烷基的另一个氢而形成的二价基团,并且可以是取代或未取代的。在一些实施方案中,C1-4亚烷基、C2-4亚烷基和C1-3亚烷基是优选的。未取代的所述亚烷基包括但不限于:亚甲基(-CH2-)、亚乙基(-CH2CH2-)、亚丙基(-CH2CH2CH2-)、亚丁基(-CH2CH2CH2CH2-)、亚戊基(- CH2CH2CH2CH2CH2-)、亚己基(-CH2CH2CH2CH2CH2CH2-),等等。示例性的取代的所述亚烷基,例如,被一个或多个烷基(甲基)取代的所述亚烷基,包括但不限于:取代的亚甲基(-CH(CH3)-、-C(CH3)2-)、取代的亚乙基(-CH(CH3)CH2-、-CH2CH(CH3)-、-C(CH3)2CH2-、-CH2C(CH3)2-)、取代的亚丙基(-CH(CH3)CH2CH2-、-CH2CH(CH3)CH2-、-CH2CH2CH(CH3)-、-C(CH3)2CH2CH2-、-CH2C(CH3)2CH2-、-CH2CH2C(CH3)2-),等等。
“C0-6亚烷基”是指化学键以及上述“C1-6亚烷基”,“C0-4亚烷基”是指化学键以及上述“C1-4亚烷基”。
“卤代”或“卤素”是指氟(F)、氯(Cl)、溴(Br)和碘(I)。
因此,“C1-6卤代烷基”是指上述“C1-6烷基”,其被一个或多个卤素基团取代。在一些实施方案中,C1-4卤代烷基是特别优选的,更优选C1-2卤代烷基。示例性的所述卤代烷基包括但不限于:-CF3、-CH2F、-CHF2、-CHFCH2F、-CH2CHF2、-CF2CF3、-CCl3、-CH2Cl、-CHCl2、2,2,2-三氟-1,1-二甲基-乙基,等等。卤代烷基基团可以在任何可用的连接点上被取代,例如,1至5个取代基、1至3个取代基或1个取代基。
“C3-10环烷基”是指具有3至10个环碳原子和零个杂原子的非芳香环烃基团。在一些实施方案中,C3-8环烷基、C3-7环烷基和C3-6环烷基是特别优选的,更优选C5-6环烷基。环烷基还包括其中上述环烷基环与一个或多个芳基或杂芳基稠合的环体系,其中连接点在环烷基环上,且在这样的情况中,碳的数目继续表示环烷基体系中的碳的数目。示例性的所述环烷基包括但不限于:环丙基(C3)、环丙烯基(C3)、环丁基(C4)、环丁烯基(C4)、环戊基(C5)、环戊烯基(C5)、环己基(C6)、环己烯基(C6)、环已二烯基(C6)、环庚基(C7)、环庚烯基(C7)、环庚二烯基(C7)、环庚三烯基(C7),等等。环烷基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“3-12元杂环基”是指具有环碳原子和1至5个环杂原子的3至12元非芳香环系的饱和或不饱和基团,其中,每个杂原子独立地选自氮、氧、硫、硼、磷和硅。在包含一个或多个氮原子的杂环基中,只要化合价允许,连接点可为碳或氮原子。在一些实施方案中,优选4-10元杂环基,其为具有环碳原子和1至5个环杂原子的4至10元非芳香环系;在一些实施方案中,优选3-8元杂环基,其为具有环碳原子和1至4个环杂原子的3至8元非芳香环系;优选4-8元杂环基,其为具有环碳原子和1至3个环杂原子的4至8元非芳香环系;优选4-7元杂环基,其为具有环碳原子和1至3个环杂原子的4至7元非芳香环系;优选4-6元杂环基,其为具有环碳原子和1至3个环杂原子的4至6元非芳香环系;更优选5-6元杂环基,其为具有环碳原子和1至3个环杂原子的5至6元非芳香环系。杂环基还包括其中上述杂环基环与一个或多个环烷基稠合的环体系,其中连接点在环烷基环上,或其中上述杂环基环与一个或多个芳基或杂芳基稠合的环体系,其中连接点在杂环基环上;且在这样的情况下,环成员的数目继续表示在杂环基环体系中环成员的数目。示例性的包含一个杂原子的3元杂环基包括但不限于:氮杂环丙烷基、氧杂环丙烷基、硫杂环丙烷基(thiorenyl)。示例性的含有一个杂原子的4元杂环基包括但不限于:氮杂环丁烷基、氧杂环丁烷基和硫杂环丁烷基。示例性的含有一个杂原子的5元杂环基包括但不限于:四氢呋喃基、二氢呋喃基、四氢噻吩基、二氢噻吩基、吡咯烷基、二氢吡咯基和吡咯基-2,5-二酮。示例性的包含两个杂原子的5元杂环基包括但不限于:吡唑烷基、二氧杂环戊 烷基、氧硫杂环戊烷基(oxasulfuranyl)、二硫杂环戊烷基(disulfuranyl)和噁唑烷-2-酮。示例性的包含三个杂原子的5元杂环基包括但不限于:***啉基、噁二唑啉基和噻二唑啉基。示例性的包含一个杂原子的6元杂环基包括但不限于:哌啶基、四氢吡喃基、二氢吡啶基和硫杂环己烷基(thianyl)。示例性的包含两个杂原子的6元杂环基包括但不限于:哌嗪基、吗啉基、二硫杂环己烷基、二噁烷基。示例性的包含三个杂原子的6元杂环基包括但不限于:六氢三嗪基(triazinanyl)。示例性的含有一个杂原子的7元杂环基包括但不限于:氮杂环庚烷基、氧杂环庚烷基和硫杂环庚烷基。示例性的与C6芳基环稠合的5元杂环基(在本文中也称作5,6-双环杂环基)包括但不限于:二氢吲哚基、异二氢吲哚基、二氢苯并呋喃基、二氢苯并噻吩基、苯并噁唑啉酮基,等等。示例性的与C6芳基环稠合的6元杂环基(本文还指的是6,6-双环杂环基)包括但不限于:四氢喹啉基、四氢异喹啉基,等等。杂环基还包括上述杂环基与一个环烷基、杂环基、芳基或杂芳基共享一个或两个原子,形成桥环或螺环,只要化合价允许,共享的原子可为碳或氮原子。杂环基还包括上述杂环基与杂环基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“C6-10芳基”是指具有6-10个环碳原子和零个杂原子的单环或多环的(例如,双环)4n+2芳族环体系(例如,具有以环状排列共享的6或10个π电子)的基团。在一些实施方案中,芳基具有六个环碳原子(“C6芳基”;例如,苯基)。在一些实施方案中,芳基具有十个环碳原子(“C10芳基”;例如,萘基,例如,1-萘基和2-萘基)。芳基还包括其中上述芳基环与一个或多个环烷基或杂环基稠合的环***,而且连接点在所述芳基环上,在这种情况下,碳原子的数目继续表示所述芳基环***中的碳原子数目。芳基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“5-14元杂芳基”是指具有环碳原子和1-4个环杂原子的5-14元单环或双环的4n+2芳族环体系(例如,具有以环状排列共享的6、10或14个π电子)的基团,其中每个杂原子独立地选自氮、氧和硫。在含有一个或多个氮原子的杂芳基中,只要化合价允许,连接点可以是碳或氮原子。杂芳基双环***在一个或两个环中可以包括一个或多个杂原子。杂芳基还包括其中上述杂芳基环与一个或多个环烷基或杂环基稠合的环***,而且连接点在所述杂芳基环上,在这种情况下,碳原子的数目继续表示所述杂芳基环***中的碳原子数目。在一些实施方案中,5-10元杂芳基是优选的,其为具有环碳原子和1-4个环杂原子的5-10元单环或双环的4n+2芳族环体系。在另一些实施方案中,5-6元杂芳基是特别优选的,其为具有环碳原子和1-4个环杂原子的5-6元单环或双环的4n+2芳族环体系。示例性的含有一个杂原子的5元杂芳基包括但不限于:吡咯基、呋喃基和噻吩基。示例性的含有两个杂原子的5元杂芳基包括但不限于:咪唑基、吡唑基、噁唑基、异噁唑基、噻唑基和异噻唑基。示例性的含有三个杂原子的5元杂芳基包括但不限于:***基、噁二唑基(例如,1,2,4-噁二唑基)和噻二唑基。示例性的含有四个杂原子的5元杂芳基包括但不限于:四唑基。示例性的含有一个杂原子的6元杂芳基包括 但不限于:吡啶基或吡啶酮基。示例性的含有两个杂原子的6元杂芳基包括但不限于:哒嗪基、嘧啶基和吡嗪基。示例性的含有三个或四个杂原子的6元杂芳基分别包括但不限于:三嗪基和四嗪基。示例性的含有一个杂原子的7元杂芳基包括但不限于:氮杂环庚三烯基、氧杂环庚三烯基和硫杂环庚三烯基。示例性的5,6-双环杂芳基包括但不限于:吲哚基、异吲哚基、吲唑基、苯并***基、苯并噻吩基、异苯并噻吩基、苯并呋喃基、苯并异呋喃基、苯并咪唑基、苯并噁唑基、苯并异噁唑基、苯并噁二唑基、苯并噻唑基、苯并异噻唑基、苯并噻二唑基、茚嗪基和嘌呤基。示例性的6,6-双环杂芳基包括但不限于:萘啶基、喋啶基、喹啉基、异喹啉基、噌琳基、喹喔啉基、酞嗪基和喹唑啉基。杂芳基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
上文定义的烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基等基团除去另一个氢而形成的二价基团统称为“亚基”。环烷基、杂环基、芳基和杂芳基等成环的基团统称为“环基”。
本文定义的烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基等为任选取代的基团。
示例性的碳原子上的取代基包括但不局限于:卤素、-CN、-NO2、-N3、-SO2H、-SO3H、-OH、-ORaa、-ON(Rbb)2、-N(Rbb)2、-N(Rbb)3 +X-、-N(ORcc)Rbb、-SH、-SRaa、-SSRcc、-C(=O)Raa、-CO2H、-CHO、-C(ORcc)2、-CO2Raa、-OC(=O)Raa、-OCO2Raa、-C(=O)N(Rbb)2、-OC(=O)N(Rbb)2、-NRbbC(=O)Raa、-NRbbCO2Raa、-NRbbC(=O)N(Rbb)2、-C(=NRbb)Raa、-C(=NRbb)ORaa、-OC(=NRbb)Raa、-OC(=NRbb)ORaa、-C(=NRbb)N(Rbb)2、-OC(=NRbb)N(Rbb)2、-NRbbC(=NRbb)N(Rbb)2、-C(=O)NRbbSO2Raa、-NRbbSO2Raa、-SO2N(Rbb)2、-SO2Raa、-SO2ORaa、-OSO2Raa、-S(=O)Raa、-OS(=O)Raa、-Si(Raa)3、-OSi(Raa)3、-C(=S)N(Rbb)2、-C(=O)SRaa、-C(=S)SRaa、-SC(=S)SRaa、-SC(=O)SRaa、-OC(=O)SRaa、-SC(=O)ORaa、-SC(=O)Raa、-P(=O)2Raa、-OP(=O)2Raa、-P(=O)(Raa)2、-OP(=O)(Raa)2、-OP(=O)(ORcc)2、-P(=O)2N(Rbb)2、-OP(=O)2N(Rbb)2、-P(=O)(NRbb)2、-OP(=O)(NRbb)2、-NRbbP(=O)(ORcc)2、-NRbbP(=O)(NRbb)2、-P(Rcc)2、-P(Rcc)3、-OP(Rcc)2、-OP(Rcc)3、-B(Raa)2、-B(ORcc)2、-BRaa(ORcc)、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个Rdd基团取代;
或者在碳原子上的两个偕氢被基团=O、=S、=NN(Rbb)2、=NNRbbC(=O)Raa、=NNRbbC(=O)ORaa、=NNRbbS(=O)2Raa、=NRbb或=NORcc取代;
Raa的每个独立地选自烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个Raa基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个Rdd基团取代;
Rbb的每个独立地选自:氢、-OH、-ORaa、-N(Rcc)2、-CN、-C(=O)Raa、-C(=O)N(Rcc)2、-CO2Raa、-SO2Raa、-C(=NRcc)ORaa、-C(=NRcc)N(Rcc)2、-SO2N(Rcc)2、-SO2Rcc、-SO2ORcc、-SORaa、-C(=S)N(Rcc)2、-C(=O)SRcc、-C(=S)SRcc、-P(=O)2Raa、-P(=O)(Raa)2、-P(=O)2N(Rcc)2、-P(=O)(NRcc)2、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个Rbb基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个Rdd基团取代;
Rcc的每个独立地选自氢、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个Rcc基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个Rdd基团取代;
Rdd的每个独立地选自:卤素、-CN、-NO2、-N3、-SO2H、-SO3H、-OH、-ORee、-ON(Rff)2、-N(Rff)2,、-N(Rff)3 +X-、-N(ORee)Rff、-SH、-SRee、-SSRee、-C(=O)Ree、-CO2H、-CO2Ree、-OC(=O)Ree、-OCO2Ree、-C(=O)N(Rff)2、-OC(=O)N(Rff)2、-NRffC(=O)Ree、-NRffCO2Ree、-NRffC(=O)N(Rff)2、-C(=NRff)ORee、-OC(=NRff)Ree、-OC(=NRff)ORee、-C(=NRff)N(Rff)2、-OC(=NRff)N(Rff)2、-NRffC(=NRff)N(Rff)2、-NRffSO2Ree、-SO2N(Rff)2、-SO2Ree、-SO2ORee、-OSO2Ree、-S(=O)Ree、-Si(Ree)3、-OSi(Ree)3、-C(=S)N(Rff)2、-C(=O)SRee、-C(=S)SRee、-SC(=S)SRee、-P(=O)2Ree、-P(=O)(Ree)2、-OP(=O)(Ree)2、-OP(=O)(ORee)2、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个Rgg基团取代,或者两个偕Rdd取代基可结合以形成=O或=S;
Ree的每个独立地选自烷基、卤代烷基、烯基、炔基、环烷基、芳基、杂环基和杂芳基,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个Rgg基团取代;
Rff的每个独立地选自氢、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个Rff基团结合形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个Rgg基团取代;
Rgg的每个独立地是:卤素、-CN、-NO2、-N3、-SO2H、-SO3H、-OH、-OC1-6烷基、-ON(C1-6烷基)2、-N(C1-6烷基)2、-N(C1-6烷基)3 +X-、-NH(C1-6烷基)2 +X-、-NH2(C1-6烷基)+X-、-NH3 +X-、-N(OC1-6烷基)(C1-6烷基)、-N(OH)(C1-6烷基)、-NH(OH)、-SH、-SC1-6烷基、-SS(C1-6烷基)、-C(=O)(C1-6烷基)、-CO2H、-CO2(C1-6烷基)、-OC(=O)(C1-6烷基)、-OCO2(C1-6烷基)、-C(=O)NH2、-C(=O)N(C1-6烷基)2、-OC(=O)NH(C1-6烷基)、-NHC(=O)(C1-6烷基)、-N(C1-6烷基)C(=O)(C1-6烷基)、-NHCO2(C1-6烷基)、-NHC(=O)N(C1-6烷基)2、-NHC(=O)NH(C1-6烷基)、-NHC(=O)NH2、-C(=NH)O(C1-6烷基)、-OC(=NH)(C1- 6烷基)、-OC(=NH)OC1-6烷基、-C(=NH)N(C1-6烷基)2、-C(=NH)NH(C1-6烷基)、-C(=NH)NH2、-OC(=NH)N(C1-6烷基)2、-OC(NH)NH(C1-6烷基)、-OC(NH)NH2、-NHC(NH)N(C1-6烷基)2、-NHC(=NH)NH2、-NHSO2(C1-6烷基)、-SO2N(C1-6烷基)2、-SO2NH(C1-6烷基)、-SO2NH2、-SO2C1-6烷基、-SO2OC1-6烷基、-OSO2C1-6烷基、-SOC1-6烷基、-Si(C1-6烷基)3、-OSi(C1-6烷基)3、-C(=S)N(C1-6烷基)2、C(=S)NH(C1-6烷基)、C(=S)NH2、-C(=O)S(C1-6烷基)、-C(=S)SC1-6烷基、-SC(=S)SC1-6烷基、-P(=O)2(C1-6烷基)、-P(=O)(C1- 6烷基)2、-OP(=O)(C1-6烷基)2、-OP(=O)(OC1-6烷基)2、C1-6烷基、C1-6卤代烷基、C2-C6烯基、C2-C6炔基、C3-C7环烷基、C6-C10芳基、C3-C7杂环基、C5-C10杂芳基;或者两个偕Rgg取代基可结合形成=O或=S;其中,X-为反离子。
示例性的氮原子上取代基包括但不局限于:氢、-OH、-ORaa、-N(Rcc)2、-CN、-C(=O)Raa、-C(=O)N(Rcc)2、-CO2Raa、-SO2Raa、-C(=NRbb)Raa、-C(=NRcc)ORaa、-C(=NRcc)N(Rcc)2、-SO2N(Rcc)2、-SO2Rcc、-SO2ORcc、-SORaa、-C(=S)N(Rcc)2、-C(=O)SRcc、-C(=S)SRcc、-P(=O)2Raa、-P(=O)(Raa)2、-P(=O)2N(Rcc)2、 -P(=O)(NRcc)2、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者连接至氮原子的两个Rcc基团结合形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个Rdd基团取代,且其中Raa、Rbb、Rcc和Rdd如上所述。
其它定义
术语“癌症”包括但不限于下列癌症:心脏:肉瘤(血管肉瘤、纤维肉瘤、横纹肌肉瘤、脂肪肉瘤)、粘液瘤、横纹肌瘤、纤维瘤、脂肪瘤和畸胎瘤;肺:支气管癌(鳞状细胞、未分化小细胞、未分化大细胞、腺癌)、肺泡(细支气管)癌、支气管腺瘤、肉瘤、淋巴瘤、软骨瘤错构瘤、间皮瘤;胃肠道:食管(鳞状细胞癌、腺癌、平滑肌肉瘤、淋巴瘤)、胃(癌、淋巴瘤、平滑肌肉瘤)、胰腺(导管腺癌、胰岛素瘤、胰高血糖素瘤、胃泌素瘤、类癌瘤、血管瘤)、小肠(腺癌、淋巴瘤、类癌瘤)、卡波西肉瘤、平滑肌瘤、血管瘤、脂肪瘤、神经纤维瘤、纤维瘤)、大肠(腺癌、管状腺瘤、绒毛状腺瘤、错构瘤、平滑肌瘤);泌尿生殖道:肾脏(腺癌、威尔姆氏瘤(肾母细胞瘤)、淋巴瘤、白血病)、膀胱和尿道(鳞状细胞癌、移行细胞癌、腺癌)、***(腺癌、肉瘤)、睾丸(***瘤、畸胎瘤、胚胎癌、畸胎癌)、绒毛膜癌、肉瘤、***癌、纤维瘤、纤维腺瘤、腺瘤样肿瘤、脂肪瘤);肝脏:肝细胞瘤(肝细胞癌)、胆管癌、肝母细胞瘤、血管肉瘤、肝细胞腺瘤、血管瘤;胆道:胆囊癌、壶腹癌、胆管癌;骨:成骨肉瘤(osteosarcoma)、纤维肉瘤、恶性纤维组织细胞瘤、软骨肉瘤、尤文氏肉瘤、恶性淋巴瘤(网状细胞肉瘤)、多发性骨髓瘤、恶性巨细胞瘤脊索瘤、骨慢性外生骨疣(osteocartilaginous exostoses)、良性软骨瘤、软骨母细胞瘤、纤维软骨瘤、类骨质骨瘤和巨细胞瘤;神经***:颅骨(骨瘤、血管瘤、肉芽肿、黄色瘤、变形性骨炎)、脑膜(脑膜瘤、脑膜肉瘤、神经胶质瘤)、脑(星形细胞瘤、髓母细胞瘤、神经胶质瘤、室管膜瘤、生殖细胞瘤(松果体瘤)、多形性胶质母细胞瘤、少突胶质细胞瘤、神经鞘瘤、视网膜母细胞瘤、先天性肿瘤)、脊髓神经纤维瘤、脑膜瘤、神经胶质瘤、肉瘤);妇科:子宫(子宫内膜癌(浆液性囊腺癌、粘液性囊腺癌、未分类癌)、颗粒鞘细胞瘤、支持***瘤、无性细胞瘤、恶性畸胎瘤)、外阴(鳞状细胞癌、上皮内癌、腺癌、纤维肉瘤、黑色素瘤)、***(透明细胞癌、鳞状细胞癌、葡萄样肉瘤(胚胎性横纹肌肉瘤)、输卵管(癌);血液学:血液(髓性白血病(急性和慢性)、急性淋巴细胞白血病、慢性淋巴细胞性白血病、骨髓增生性疾病、多发性骨髓瘤、骨髓增生异常综合征)、霍奇金病、非霍奇金淋巴瘤(恶性淋巴瘤);皮肤:恶性黑色素瘤、基底细胞癌、鳞状细胞癌、卡波西肉瘤、痣发育不良痣、脂肪瘤、血管瘤、皮肤纤维瘤、瘢痕疙瘩、牛皮癣;和肾上腺:成神经细胞瘤;尤其是MTAP相关癌症,例如肝细胞癌、乳腺癌、皮肤癌、膀胱癌、肝癌、胰腺癌或头颈癌。
在一个实施方案中,术语“癌症”包括但不限于下列癌症:恶性周围神经鞘瘤、间皮瘤、多形性胶质母细胞瘤、胰腺癌、胆管癌、***癌、乳腺癌、脑癌、皮肤癌、***、膀胱癌、星形细胞瘤、结肠直肠癌、子宫内膜癌、食道癌、胃癌、胸腺瘤、头颈癌、肝细胞癌、喉癌、肺鳞癌、肺腺癌、口腔癌、卵巢癌、肾癌和甲状腺癌以及肉瘤。
在个一个实施方案中,术语“癌症”包括但不限于下列癌症:肝细胞癌、乳腺癌、皮肤癌、膀胱癌、 肝癌、胰腺癌或头颈癌。
本文所用的术语“治疗”涉及逆转、减轻、抑制该术语适用的障碍或病症的进展或者预防之,或者这类障碍或病症的一种或多种症状。本文所用的名词“治疗”涉及动词治疗的动作,后者是如刚才所定义的。
本文所用的术语“药学上可接受的盐”表示本发明化合物的那些羧酸盐、氨基酸加成盐,它们在可靠的医学判断范围内适用于与患者组织接触,不会产生不恰当的毒性、刺激作用、***反应等,与合理的益处/风险比相称,就它们的预期应用而言是有效的,包括(可能的话)本发明化合物的两性离子形式。
药学上可接受的碱加成盐是与金属或胺生成的,例如碱金属与碱土金属氢氧化物或有机胺。用作阳离子的金属的实例有钠、钾、镁、钙等。适合的胺的实例有N,N'-二苄基乙二胺、氯普鲁卡因、胆碱、二乙醇胺、乙二胺、N-甲基葡糖胺和普鲁卡因。
酸性化合物的碱加成盐可以这样制备,按照常规方式使游离酸形式与足量所需的碱接触,生成盐。按照常规方式使盐形式与酸接触,再分离游离酸,可以使游离酸再生。游离酸形式在某些物理性质上多少不同于它们各自的盐形式,例如在极性溶剂中的溶解度,但是出于本发明的目的,盐还是等价于它们各自的游离酸。
盐可以是从无机酸制备的硫酸盐、焦硫酸盐、硫酸氢盐、亚硫酸盐、亚硫酸氢盐、硝酸盐、磷酸盐、磷酸一氢盐、磷酸二氢盐、偏磷酸盐、焦磷酸盐、氯化物、溴化物、碘化物,酸例如盐酸、硝酸、硫酸、氢溴酸、氢碘酸、磷酸等。代表性盐包括:氢溴酸盐、盐酸盐、硫酸盐、硫酸氢盐、硝酸盐、乙酸盐、草酸盐、戊酸盐、油酸盐、棕榈酸盐、硬脂酸盐、月桂酸盐、硼酸盐、苯甲酸盐、乳酸盐、磷酸盐、甲苯磺酸盐、柠檬酸盐、马来酸盐、富马酸盐、琥珀酸盐、酒石酸盐、萘甲酸盐、甲磺酸盐、葡庚糖酸盐、乳糖酸盐、月桂基磺酸盐和羟乙磺酸盐等。盐也可以是从有机酸制备的,例如脂肪族一元与二元羧酸、苯基取代的烷酸、羟基烷酸、烷二酸、芳香族酸、脂肪族与芳香族磺酸等。代表性盐包括乙酸盐、丙酸盐、辛酸盐、异丁酸盐、草酸盐、丙二酸盐、琥珀酸盐、辛二酸盐、癸二酸盐、富马酸盐、马来酸盐、扁桃酸盐、苯甲酸盐、氯苯甲酸盆、甲基苯甲酸盐、二硝基苯甲酸盐、萘甲酸盐、苯磺酸盐、甲苯磺酸盐、苯乙酸盐、柠檬酸盐、乳酸盐、马来酸盐、酒石酸盐、甲磺酸盐等。药学上可接受的盐可以包括基于碱金属与碱土金属的阳离子,例如钠、锂、钾、钙、镁等,以及无毒的铵、季铵和胺阳离子,包括但不限于铵、四甲基铵、四乙基铵、甲胺、二甲胺、三甲胺、三乙胺、乙胺等。还涵盖氨基酸的盐,例如精氨酸盐、葡糖酸盐、半乳糖醛酸盐等(例如参见Berge S.M.et al.,"Pharmaceutical Salts,”J.Pharm.Sci.,1977;66:1-19,引入此作为参考)。
给药的“受试者”包括但不限于:人(即,任何年龄组的男性或女性,例如,儿科受试者(例如,婴儿、儿童、青少年)或成人受试者(例如,年轻的成人、中年的成人或年长的成人))和/或非人的动物,例如,哺乳动物,例如,灵长类(例如,食蟹猴、恒河猴)、牛、猪、马、绵羊、山羊、啮齿动物、猫和/或狗。在一些实施方案中,受试者是人。在一些实施方案中,受试者是非人动物。本文可互换使用术语“人”、“患者”和“受试者”。
“疾病”、“障碍”和“病症”在本文中可互换地使用。
除非另作说明,否则,本文使用的术语“治疗”包括受试者患有具体疾病、障碍或病症时所发生的作用,它降低疾病、障碍或病症的严重程度,或延迟或减缓疾病、障碍或病症的发展(“治疗性治疗”),还包括在受试者开始患有具体疾病、障碍或病症之前发生的作用(“预防性治疗”)。
通常,化合物的“有效量”是指足以引起目标生物反应的数量。正如本领域普通技术人员所理解的那样,本发明化合物的有效量可以根据下列因素而改变:例如,生物学目标、化合物的药代动力学、所治疗的疾病、给药模式以及受试者的年龄健康情况和症状。有效量包括治疗有效量和预防有效量。
除非另作说明,否则,本文使用的化合物的“治疗有效量”是在治疗疾病、障碍或病症的过程中足以提供治疗益处的量,或使与疾病、障碍或病症有关的一或多种症状延迟或最小化的量。化合物的治疗有效量是指单独使用或与其它疗法联用时,治疗剂的量,它在治疗疾病、障碍或病症的过程中提供治疗益处。术语“治疗有效量”可以包括改善总体治疗、降低或避免疾病或病症的症状或病因、或增强其它治疗剂的治疗效果的量。
除非另作说明,否则,本文使用的化合物的“预防有效量”是足以预防疾病、障碍或病症的量,或足以预防与疾病、障碍或病症有关的一或多种症状的量,或防止疾病、障碍或病症复发的量。化合物的预防有效量是指单独使用或与其它药剂联用时,治疗剂的量,它在预防疾病、障碍或病症的过程中提供预防益处。术语“预防有效量”可以包括改善总体预防的量,或增强其它预防药剂的预防效果的量。
“组合”以及相关术语是指同时或依次给药本发明化合物和其它治疗剂。例如,本发明化合物可以与其它治疗剂以分开的单位剂型同时或依次给药,或与其它治疗剂一起在单一单位剂型中同时给药。
附图说明
图1和2示出了LU99 CDX模型体内药效学研究的结果。
具体实施方案
本文中,“本发明化合物”指的是以下的式(I)、式(II)等化合物(包括子通式,例如式(I-1)至式(IV-1)等)、其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物。
本文中,化合物使用标准的命名法命名。具有非对称中心的化合物,应该明白(除非另有说明)所有的光学异构体及其混合物均包含在内。此外,除非另有规定,本发明所包括的所有异构体化合物与碳碳双键可能以Z和E的形式出现。在不同的互变异构形式存在的化合物,一个所述化合物并不局限于任何特定的互变异构体,而是旨在涵盖所有的互变异构形式。
在一个实施方案中,本发明提供了式(I)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物:
其中:
Z1、Z2和Z3独立地选自N、CD或CH,并且其中至少一个为N;
环A为苯基或5-6元杂芳基;
R1选自H、D、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
R2选自H、D、-C0-6亚烷基-卤素、-C0-6亚烷基-CN、-C0-6亚烷基-ORa、-C0-6亚烷基-NRbRc、C1-6烷基、C1-6卤代烷基、C3-8环烷基或4-7元杂环基;
m=0、1、2或3;
其中R选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O、C=S、C3-8环烷基或4-7元杂环基;或者相邻原子上的两个R与所述原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3或4个R’取代基取代;
R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,优选H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1个或多个D、卤素、C1-6烷基或C1-6卤代烷基取代;
Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基;
其中环A、R1、R2、R、R’、Ra、Rb和Rc中的各基团可任选地被D取代,直至完全氘代。
在另一个实施方案中,本发明涉及式(I)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物:
其中:
Z1、Z2和Z3独立地选自N、CD或CH,并且其中至少一个为N;
环A为苯基或5-6元杂芳基;
R1选自H、D、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
R2选自H、D、-C0-6亚烷基-卤素、-C0-6亚烷基-CN、-C0-6亚烷基-ORa、-C0-6亚烷基-NRbRc、C1-6烷基、C1-6卤代烷基、C3-8环烷基或4-7元杂环基;
m=0、1、2或3;
其中R选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O、C=S、C3-8环烷基或4-7元杂环基;或者相邻原子上的两个R与所述原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3或4个R’取代基取代;
R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,优选H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基或4-7元杂环基;
Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基;
其中环A、R1、R2、R、R’、Ra、Rb和Rc中的各基团可任选地被D取代,直至完全氘代。
Z1、Z2和Z3
在一个具体实施方式中,Z1为N,且Z2和Z3独立地选自CD或CH;在另一个具体实施方式中,Z2为N,且Z1和Z3独立地选自CD或CH;在另一个具体实施方式中,Z3为N,且Z1和Z2独立地选自CD或CH;在另一个具体实施方式中,Z1和Z2为N,且Z3选自CD或CH;在另一个具体实施方式中,Z1和Z3为N,且Z2选自CD或CH;在另一个具体实施方式中,Z2和Z3为N,且Z1选自CD或CH;在另一个具体实施方式中,Z1、Z2和Z3均为N。
在一个更具体的实施方式中,选自以下:
环A
在一个具体实施方式中,环A为苯基;在另一个具体实施方式中,环A为5-6元杂芳基,优选5元杂芳基。
在一个更具体的实施方式中,环A选自
在另一个更具体的实施方式中,环A选自 优选更优选为
R1、R和R’
在一个具体实施方式中,R1选自H、D、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;在另一个具体实施方式中,R1选自C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基;在另一个具体实施方式中,R1选自4-7元杂环基、C6-10芳基或5-6元杂芳基;在另一个具体实施方式中,R1选自苯基、吡咯烷基、吡唑烷基、哌啶基或吡啶酮基;在另一个具体实施方式中,R1为H;在另一个具体实施方式中,R1为D;在另一个具体实施方式中,R1为C3-10环烷基;在另一个具体实施方式中,R1为4-10元杂环基;在另一个具体实施方式中,R1为C6-10芳基;在另一个具体实施方式中,R1为5-10元杂芳基。
在上述具体实施方式中,R1被1个R取代基取代;在上述具体实施方式中,R1被2个R取代基取代;在上述具体实施方式中,R1被3个R取代基取代;在上述具体实施方式中,R1被4个R取代基取代;在上述具体实施方式中,R1被5个R取代基取代;在上述具体实施方式中,R1被6个R取代基取代;在上述具体实施方式中,R1被7个R取代基取代;在上述具体实施方式中,R1被8个R取代基取代;在上述具体实施方式中,R1被9个R取代基取代;在上述具体实施方式中,R1被10个R取代基取代。
在更具体的实施方式中,R选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;在 另一更具体的实施方式中,R选自卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;在另一更具体的实施方式中,R为H;在另一更具体的实施方式中,R为D;在另一更具体的实施方式中,R为卤素;在另一更具体的实施方式中,R为CN;在另一更具体的实施方式中,R为ORa;在另一更具体的实施方式中,R为NRbRc;在另一更具体的实施方式中,R为C1-6烷基;在另一更具体的实施方式中,R为C1-6卤代烷基;在另一更具体的实施方式中,相同碳原子上的两个R与该碳原子形成C=O;在另一更具体的实施方式中,相同碳原子上的两个R与该碳原子形成C=S;在另一更具体的实施方式中,相同碳原子上的两个R与该碳原子形成C3-8环烷基,优选C3-6环烷基;在另一更具体的实施方式中,相同碳原子上的两个R与该碳原子形成4-7元杂环基;在另一更具体的实施方式中,相邻原子上的两个R与所述原子形成C3-8环烷基,优选C3-6环烷基;在另一更具体的实施方式中,相邻原子上的两个R与所述原子形成4-7元杂环基,优选5-6元杂环基;在另一更具体的实施方式中,相邻原子上的两个R与所述原子形成C6-10芳基,优选苯基;在另一更具体的实施方式中,相邻原子上的两个R与所述原子形成5-10元杂芳基,优选5-6元杂芳基;在另一更具体的实施方式中,相邻原子上的两个R与所述原子形成环戊烯基、苯基、噻吩基或吡啶基。
在上述具体实施方式中,R被1个R’取代基取代;在上述具体实施方式中,R被2个R’取代基取代;在上述具体实施方式中,R被3个R’取代基取代;在上述具体实施方式中,R被4个R’取代基取代;
在更具体的实施方式中,R’为H;在另一更具体的实施方式中,R’为D;在另一更具体的实施方式中,R’为卤素;在另一更具体的实施方式中,R’为CN;在另一更具体的实施方式中,R’为ORa;在另一更具体的实施方式中,R’为NRbRc;在另一更具体的实施方式中,R’为C(O)ORa;在另一更具体的实施方式中,R’为C(O)NRbRc;在另一更具体的实施方式中,R’为S(O)Ra;在另一更具体的实施方式中,R’为S(O)2Ra;在另一更具体的实施方式中,R’为C1-6烷基;在另一更具体的实施方式中,R’为C1-6卤代烷基;在另一更具体的实施方式中,R’为C2-6烯基;在另一更具体的实施方式中,R’为C2-6炔基;在另一更具体的实施方式中,R’为C3-10环烷基;在另一更具体的实施方式中,R’为4-10元杂环基;在另一更具体的实施方式中,R’为C6-10芳基;在另一更具体的实施方式中,R’为5-10元杂芳基;在另一更具体的实施方式中,相同或不同原子上的两个R’与所述原子形成任选地被1个或多个D、卤素、C1-6烷基或C1-6卤代烷基取代的C3-8环烷基(优选C3-6环烷基);在另一更具体的实施方式中,相同或不同原子上的两个R’与所述原子形成任选地被1个或多个D、卤素、C1-6烷基或C1-6卤代烷基取代的4-7元杂环基(优选5-6元杂环基);在另一更具体的实施方式中,R’为任选地被1个或多个D、卤素、C1-6烷基或C1-6卤代烷基取代的C6-10芳基(优选苯基);在另一更具体的实施方式中,R’为任选地被1个或多个D、卤素、C1-6烷基或C1-6卤代烷基取代的5-10元杂芳基(优选5-6元杂芳基)。
R2
在一个具体实施方式中,R2选自H、D、-C0-6亚烷基-卤素、-C0-6亚烷基-CN、-C0-6亚烷基-ORa、 -C0-6亚烷基-NRbRc、C1-6烷基、C1-6卤代烷基、C3-8环烷基或4-7元杂环基;在另一个具体实施方式中,R2选自H、卤素、CN、-C0-6亚烷基-ORa、-C0-6亚烷基-NRbRc、C1-6烷基、C1-6卤代烷基或C3-8环烷基;在另一个具体实施方式中,R2选自H、-C0-6亚烷基-ORa、C1-6烷基、C1-6卤代烷基或C3-6环烷基;在另一个具体实施方式中,R2选自H、F、Cl、Br、CH2CH2OH、Me、Et、Pr、iPr、CF3或环丙基;在另一个具体实施方式中,R2选自C1-6烷基或C1-6卤代烷基,优选Me;在另一个具体实施方式中,R2为H;在另一个具体实施方式中,R2为D;在另一个具体实施方式中,R2为-C0-6亚烷基-卤素,优选卤素;在另一个具体实施方式中,R2为-C0-6亚烷基-CN,优选CN;在另一个具体实施方式中,R2为-C0- 6亚烷基-ORa,优选-ORa;在另一个具体实施方式中,R2为-C0-6亚烷基-NRbRc,优选NRbRc;在另一个具体实施方式中,R2为C1-6烷基;在另一个具体实施方式中,R2为C1-6卤代烷基;在另一个具体实施方式中,R2为C3-8环烷基;在另一个具体实施方式中,R2为4-7元杂环基。
m
在一个具体实施方式中,m=0;在另一个具体实施方式中,m=1;在另一个具体实施方式中,m=2;在另一个具体实施方式中,m=3。
以上任一具体实施方案中的任一技术方案或其任意组合,可以与其它具体实施方案中的任一技术方案或其任意组合进行组合。例如,环A的任一技术方案或其任意组合,可以与Z1、Z2、Z3、R1、R2和m等的任一技术方案或其任意组合进行组合。本发明旨在包括所有这些技术方案的组合,限于篇幅,不再一一列出。
在更具体的实施方案中,本发明提供了式(I)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物:
其中:
Z1、Z2和Z3独立地选自N、CD或CH,并且其中至少一个为N;
环A为苯基或5-6元杂芳基;
R1选自H、D、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
R2选自H、D、-C0-6亚烷基-卤素、-C0-6亚烷基-CN、-C0-6亚烷基-ORa、-C0-6亚烷基-NRbRc、C1-6 烷基、C1-6卤代烷基、C3-8环烷基或4-7元杂环基;
m=0、1、2或3;
其中R选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O、C=S、C3-8环烷基或4-7元杂环基;或者相邻原子上的两个R与所述原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3或4个R’取代基取代;
R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,优选H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1个或多个D、卤素、C1-6烷基或C1-6卤代烷基取代;
Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基;
其中环A、R1、R2、R、R’、Ra、Rb和Rc中的各基团可任选地被D取代,直至完全氘代。
在更具体的实施方案中,本发明提供了式(I)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物:
其中:
Z1、Z2和Z3独立地选自N、CD或CH,并且其中至少一个为N;
环A为苯基或5-6元杂芳基;
R1选自H、D、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
R2选自H、D、-C0-6亚烷基-卤素、-C0-6亚烷基-CN、-C0-6亚烷基-ORa、-C0-6亚烷基-NRbRc、C1-6烷基、C1-6卤代烷基、C3-8环烷基或4-7元杂环基;
m=0、1、2或3;
其中R选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O、C=S、C3-8环烷基或4-7元杂环基;或者相邻原子上的两个R与所述原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3或4个R’取代基取 代;
R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,优选H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基或4-7元杂环基;
Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基;
其中环A、R1、R2、R、R’、Ra、Rb和Rc中的各基团可任选地被D取代,直至完全氘代。
在更具体的实施方案中,本发明提供了上述式(I)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中环A为5-6元杂芳基;
优选选自以下:
更优选选自以下:
更优选为
其中R1和R2如上文所定义。
在更具体的实施方案中,本发明提供了上述式(I)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中R2选自H、卤素、CN、-C0-6亚烷基-ORa、-C0-6亚烷基-NRbRc、C1-6烷基、C1-6卤代烷基或C3-8环烷基;优选选自H、-C0-6亚烷基-ORa、C1-6烷基、C1-6卤代烷基或C3-6环烷基;更优选选自H、F、Cl、Br、CH2CH2OH、Me、Et、Pr、iPr、CF3或环丙基;更优选为C1-6烷基或C1-6卤代烷基,尤其Me。
在更具体的实施方案中,本发明提供了上述式(I)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中R1选自C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基;优选选自4-7元杂环基、C6-10芳基或5-6元杂芳基;更优选选自苯基、吡咯烷基、吡唑烷基、哌啶基或吡啶酮基。
在更具体的实施方案中,本发明提供了上述式(I)化合物,或其药学上可接受的盐、同位素变体、 互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中R选自卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O、C=S或C3-6环烷基;或者相邻原子上的两个R与所述原子形成C3-6环烷基、5-6元杂环基、苯基或5-6元杂芳基,例如环戊烯基、苯基、噻吩基或吡啶基。
在更具体的实施方案中,本发明提供了上述式(I)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3- 10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;优选地,R’选自H、D、卤素、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;优选地,R’选自H、D、卤素、CN、C1-6烷基、C1-6卤代烷基、C6-10芳基或5-10元杂芳基;优选地,R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-6环烷基或5-6元杂环基。
在更具体的实施方案中,本发明提供了上述式(I)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:

其中,
表示单键或双键;
X选自NRN、CR2bR2b’、CR2bR2b’-CR2bR2b’或CR2b=CR2b’;
Y1选自N或CRY1
Y2选自N或CRY2
Y3选自N或CRY3
Y4选自N或CRY4
其中RN选自H、C1-6烷基或C1-6卤代烷基;
R1a、R2a、R3a、R4a、R5a、R1b、R1b’、R2b、R2b’、R3b和R4b具有上文中与R相同的定义;
RY1、RY2、RY3和RY4具有上文中与R’相同的定义;
其他基团如上文所定义。
在更具体的实施方案中,本发明提供了上述化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
其中,
R2选自H、C1-6烷基、C1-6卤代烷基、C3-8环烷基或4-7元杂环基;
R1a选自H、D、卤素、CN、ORa或NRbRc
R2a选自H、D、卤素或CN;
R3a选自H或D;
R4a选自ORa或NRbRc
R5a选自H、D或CN;
或者,R3a、R4a以及它们连接的碳原子形成4-6元杂环基或5-6元杂芳基,其任选地被1、2、3或4个R’取代基取代;
其中R’选自H、D、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基或4-7元杂环基;
Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基。
在更具体的实施方案中,本发明提供了上述式(III-1)、(III-2)或(III-3)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
R2选自H、C1-6烷基或C1-6卤代烷基;
R1a为卤素,优选F;
R2a选自H、D或卤素;
R3a为H或D;
R4a为ORa
R5a为CN;
或者,R3a、R4a以及它们连接的碳原子形成呋喃基或二氢呋喃基,其任选地被1、2、3或4个R’取代基取代;
其中R’选自H、D、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-6环烷基或5-6元杂环基;
Ra选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-7环烷基或4-7元杂环基。
在更具体的实施方案中,本发明提供了上述式(III-1)、(III-2)或(III-3)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
R2选自H、C1-4烷基或C1-4卤代烷基;
R1a为卤素,优选F;
R2a选自H、D或卤素;
R3a为H或D;
R4a为ORa
R5a为CN;
或者,R3a、R4a以及它们连接的碳原子形成二氢呋喃基,其被1、2、3或4个R’取代基取代;
其中R’选自H、D、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-6环烷基或5-6元杂环基,优选环丙基;
Ra选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-6环烷基或5-6元杂环基。
在更具体的实施方案中,本发明提供了上述式(III-1)、(III-2)或(III-3)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
R2为Me;
R1a为F;
R2a为H或Cl;
R3a、R4a以及它们连接的碳原子形成二氢呋喃基,其被1、2、3或4个R’取代基取代;
R5a为CN;
其中R’选自H或Me,或者相同或不同原子上的两个R’与所述原子形成环丙基。
在更具体的实施方案中,本发明提供了上述化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
其中,
表示单键或双键;
X选自NRN、CR2bR2b’、CR2bR2b’-CR2bR2b’或CR2b=CR2b’;
R2选自H、C1-6烷基、C1-6卤代烷基、C3-8环烷基或4-7元杂环基;
R1b和R1b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
R3b和R4b独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者R3b、R4b以及它们连接的碳原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、 2、3或4个R’取代基取代;
其中RN选自H、C1-6烷基或C1-6卤代烷基;
R2b和R2b’独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的R2b和R2b’以及它们连接的碳原子形成C3-6环烷基或4-7元杂环基;或者R2b’和R3b以及它们连接的碳原子形成C6-10芳基或5-10元杂芳基;
R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基或4-7元杂环基;
Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基。
在更具体的实施方案中,本发明提供了上述式(IV-1)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
表示单键或双键;
X选自NRN、CR2bR2b’或CR2b=CR2b’;
R2选自H、C1-6烷基或C1-6卤代烷基;
R1b和R1b’独立地选自H、D、C1-4烷基或C1-4卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
R3b和R4b独立地选自H、D、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者R3b、R4b以及它们连接的碳原子形成C3-6环烷基、4-7元杂环基、苯基或5-6元杂芳基,其任选地被1、2、3或4个R’取代基取代;
其中RN选自H、C1-6烷基或C1-6卤代烷基;
R2b和R2b’独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的R2b和R2b’以及它们连接的碳原子形成C3-6环烷基;或者R2b’和R3b以及它们连接的碳原子形成苯基或5-6元杂芳基;
R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;
Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基。
在更具体的实施方案中,本发明提供了上述化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
其中,
X选自NRN或CR2bR2b’;
R2选自H、C1-6烷基或C1-6卤代烷基;
R1b和R1b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
Y1选自N或CRY1
Y2选自N或CRY2
Y3选自N或CRY3
Y4选自N或CRY4
其中RN选自H、C1-6烷基或C1-6卤代烷基;
R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;或者R2b和R2b’以及它们连接的碳原子形成C3-6环烷基;
RY1、RY2、RY3和RY4独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;
Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基。
在更具体的实施方案中,本发明提供了上述式(IV-2)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
X选自NRN或CR2bR2b’;
R2选自H、C1-4烷基或C1-4卤代烷基;
R1b和R1b’独立地选自H、D、C1-4烷基或C1-4卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O;
Y1选自N或CRY1
Y2选自N或CRY2
Y3选自N或CRY3
Y4选自N或CRY4
其中RN选自H、C1-4烷基或C1-4卤代烷基;
R2b和R2b’独立地选自H、D、C1-4烷基或C1-4卤代烷基;或者R2b和R2b’以及它们连接的碳原子形成C3-6环烷基;
RY1、RY2、RY3和RY4独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;
Ra、Rb和Rc独立地选自H、C1-6烷基或C1-6卤代烷基,或者Rb和Rc以及它们连接的氮原子形成5-6元杂环基。
在更具体的实施方案中,本发明提供了上述式(IV-2)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
X选自NRN或CR2bR2b’;
R2为Me;
R1b和R1b’独立地选自H、D、C1-4烷基或C1-4卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O;
Y1选自N或CRY1
Y2选自N或CRY2
Y3选自N或CRY3
Y4选自N或CRY4
其中RN选自H或Me;
R2b和R2b’独立地选自H或Me,或者R2b和R2b’以及它们连接的碳原子形成环丙基;
RY1、RY2、RY3和RY4选自H、F、CN、Me、CF3或OMe。
在更具体的实施方案中,本发明提供了上述化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
其中,
表示单键或双键;
R2选自H、C1-6烷基或C1-6卤代烷基;
R1b和R1b’独立地选自H、C1-6烷基或C1-6卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
R2b和R2b’独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;
R3b选自H、D、卤素或CN;
R4b选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;
或者R2b’和R3b以及它们连接的碳原子形成苯基或5-6元杂芳基;
或者R3b、R4b以及它们连接的碳原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3或4个R’取代基取代;
R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基或4-7元杂环基;
Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基。
在更具体的实施方案中,本发明提供了上述式(IV-3)化合物,或其药学上可接受的盐、同位素变 体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
表示单键或双键;
R2选自H、C1-4烷基或C1-4卤代烷基;
R1b和R1b’独立地选自H、D、C1-4烷基或C1-4卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O;
R2b和R2b’独立地选自H、D、卤素、CN、ORa、NRbRc、C1-4烷基或C1-4卤代烷基;
R3b选自H或D;
R4b选自H、D、ORa、NRbRc、C1-4烷基或C1-4卤代烷基;
或者R2b’和R3b以及它们连接的碳原子形成苯基;
或者R3b、R4b以及它们连接的碳原子形成C3-6环烷基、5-6元杂环基、苯基或5-6元杂芳基,其任选地被1、2、3或4个R’取代基取代;
R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;
Ra、Rb和Rc独立地选自H、C1-6烷基或C1-6卤代烷基。
在更具体的实施方案中,本发明提供了上述式(IV-3)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
表示单键或双键;
R2为Me;
R1b和R1b’独立地选自H、D、C1-4烷基或C1-4卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O;
R2b选自H、D、F、Cl、Br、I、Me或CF3
R2b’选自H、D、F、Cl、Br、I、Me、CF3或OMe;
R3b为H或D;
R4b选自H、D、Me、OMe或O-环丙基;
或者,R3b、R4b以及它们连接的碳原子形成苯基、环戊烯基或噻吩基;
或者,R2b’、R3b以及它们连接的碳原子形成苯基。
在更具体的实施方案中,本发明提供了上述化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
其中:
R1和R2如权利要求1和3-6中任一项所定义;
优选地,
R1为4-7元杂环基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
R2选自C1-6烷基或C1-6卤代烷基;
其中R选自H、D、卤素、CN、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O或C=S;或者相邻原子上的两个R与所述原子形成苯基或5-6元杂芳基,其任选地被1、2、3或4个R’取代基取代;
R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;
其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基。
在更具体的实施方案中,本发明提供了上述式(V)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
R1为5-6元杂环基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
R2选自C1-6烷基或C1-6卤代烷基;
其中R选自H、D、卤素、CN、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O或C=S;或者相邻原子上的两个R与所述原子形成苯基,其任选地被1、2、3或4个R’取代基取代;
R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C3-6环烷基、5-6元杂环基、苯基或5-6元杂芳基;
其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基。
在更具体的实施方案中,本发明提供了上述式(V)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
R1选自吡咯烷基或哌啶基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
R2选自C1-6烷基或C1-6卤代烷基;
其中R选自H、D、卤素、CN、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O;或者相邻原子上的两个R与所述原子形成苯基,其任选地被1、2、3或4个R’取代基取代;
R’选自H、D、卤素、CN、C1-6烷基、C1-6卤代烷基、苯基或5-6元杂芳基。
在更具体的实施方案中,本发明提供了上述化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
其中:
表示单键或双键;
X选自CR2bR2b’、CR2bR2b’-CR2bR2b’或CR2b=CR2b’;
R2选自C1-6烷基或C1-6卤代烷基;
R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
R3b、R4b以及它们连接的碳原子形成C6-10芳基或5-10元杂芳基,其任选地被1、2、3或4个R’取代基取代;
R2b和R2b’独立地选自H、D、卤素、CN、C1-6烷基或C1-6卤代烷基;
R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;
其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基。
在更具体的实施方案中,本发明提供了上述式(VI-1)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
表示双键;
X选自CR2bR2b’、CR2bR2b’-CR2bR2b’或CR2b=CR2b’;
R2选自C1-6烷基或C1-6卤代烷基;
R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
R3b、R4b以及它们连接的碳原子形成苯基,其任选地被1、2、3或4个R’取代基取代;
R2b和R2b’独立地选自H、D、卤素、CN、C1-6烷基或C1-6卤代烷基;
R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C3-6环烷基、5-6元杂环基、苯基或5-6元杂芳基;
其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基或C2-6炔基。
在更具体的实施方案中,本发明提供了上述式(VI-1)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
表示双键;
X选自CR2bR2b’或CR2bR2b’-CR2bR2b’;
R2选自C1-6烷基或C1-6卤代烷基;
R1b、R1b’以及它们连接的碳原子形成C=O;
R3b、R4b以及它们连接的碳原子形成苯基,其任选地被1、2、3或4个R’取代基取代;
R2b和R2b’独立地选自H或D;
R’选自H、D、卤素、CN、C1-6烷基、C1-6卤代烷基、苯基或5-6元杂芳基。
在更具体的实施方案中,本发明提供了上述化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
其中:
X为CR2bR2b’;
R2选自C1-6烷基或C1-6卤代烷基;
R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
Y1选自N或CRY1
Y2选自N或CRY2
Y3选自N或CRY3
Y4选自N或CRY4
R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
RY1、RY2、RY3和RY4独立地选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;
其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基。
在更具体的实施方案中,本发明提供了上述式(VI-2)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
X为CR2bR2b’;
R2选自C1-6烷基或C1-6卤代烷基;
R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
Y1选自N或CRY1
Y2选自N或CRY2
Y3选自N或CRY3
Y4选自N或CRY4
R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
RY1、RY2、RY3和RY4独立地选自R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C3-6环烷基、5-6元杂环基、苯基或5-6元杂芳基;
其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基。
在更具体的实施方案中,本发明提供了上述式(VI-2)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
X为CR2bR2b’;
R2选自C1-6烷基或C1-6卤代烷基;
R1b、R1b’以及它们连接的碳原子形成C=O;
Y1为CRY1
Y2为CRY2
Y3为CRY3
Y4为CRY4
R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
RY1、RY2、RY3和RY4独立地选自H、D、卤素、CN、C1-6烷基、C1-6卤代烷基、苯基或5-6元杂芳基。
在更具体的实施方案中,本发明提供了上述化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
其中:
X为CR2bR2b’;
R2选自C1-6烷基、C1-6卤代烷基、C3-6环烷基或4-7元杂环基;
R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
Y1为CRY1
Y2为CRY2
Y3为CRY3
Y4为CRY4
R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
RY1、RY2、RY3和RY4独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C2- 6烯基、C2-6炔基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;
或者RY1和RY2、RY2和RY3、RY3和RY4,以及他们连接的原子一起形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1个或多个D、卤素、C1-6烷基或C1-6卤代烷基取代;
其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基。
在更具体的实施方案中,本发明提供了上述式(VI-2)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
X为CR2bR2b’;
R2为Me、Et或环丙基;
R1b、R1b’以及它们连接的碳原子形成C=O;
Y1为CRY1
Y2为CRY2
Y3为CRY3
Y4为CRY4
R2b和R2b’独立地选自H或D;
RY1选自H、D、Cl、F、CN、Me、CF3或OMe;
RY2选自H、D、Me、Et、iPr、CF3、环丙基、OMe、OEt、OiPr、OCF3、O-环丙基、丙-1-烯-2-基或苯基;
RY3选自H、D、Cl、F、Me、Et、iPr、CF3、环丙基、丙-1-烯-2-基、OMe或O-环丙基;
RY4选自H、D或Me;
或者RY1和RY2、RY2和RY3、RY3和RY4,以及他们连接的原子一起形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1个或多个D、卤素、C1-6烷基或C1-6卤代烷基取代。
在更具体的实施方案中,本发明提供了上述式(VI-2)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
X为CR2bR2b’;
R2选自C1-6烷基、C1-6卤代烷基、C3-6环烷基或4-7元杂环基;
R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
Y1为CRY1
Y2为CRY2
Y3为CRY3
Y4为CRY4
R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
RY1、RY2、RY3和RY4独立地选自H、D、卤素、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C2-6烯 基、C2-6炔基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;
其中RY1和RY3中至少一个不为H或D;
其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基。
在更具体的实施方案中,本发明提供了上述式(VI-2)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
X为CR2bR2b’;
R2选自C1-6烷基、C1-6卤代烷基或C3-6环烷基;
R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
Y1为CRY1
Y2为CRY2
Y3为CRY3
Y4为CRY4
R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
RY1和RY3独立地选自H、D、卤素、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-6环烷基或4-7元杂环基;
RY2和RY4独立地选自H、D、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C2-6烯基或C2-6炔基;
其中RY1和RY3中至少一个不为H或D;
其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基。
在更具体的实施方案中,本发明提供了上述式(VI-2)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
X为CR2bR2b’;
R2为Me、Et或环丙基;
R1b、R1b’以及它们连接的碳原子形成C=O;
Y1为CRY1
Y2为CRY2
Y3为CRY3
Y4为CRY4
R2b和R2b’独立地选自H或D;
RY1选自H、D、Cl、F、Me、CF3或OMe;
RY2选自H、D、Et、iPr、OiPr、O-环丙基或丙-1-烯-2-基;
RY3选自H、D、Cl、F、Me、Et、iPr、CF3、环丙基、丙-1-烯-2-基、OMe或O-环丙基;
RY4选自H或D;
其中RY1和RY3中至少一个不为H或D。
在更具体的实施方案中,本发明提供了上述式(VI-2)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
X为CR2bR2b’;
R2选自C1-6烷基或C1-6卤代烷基;
R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
Y1为CRY1
Y2为CRY2
Y3为CRY3
Y4为CRY4
R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
RY1和RY3独立地选自H、D、卤素、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C3-6环烷基或4-7元杂环基;
RY2和RY4独立地选自H、D、C1-6烷基或C1-6卤代烷基;
其中RY1和RY3中至少一个不为H或D;
其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基。
在更具体的实施方案中,本发明提供了上述式(VI-2)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
X为CR2bR2b’;
R2选自C1-6烷基或C1-6卤代烷基;
R1b、R1b’以及它们连接的碳原子形成C=O;
Y1为CRY1
Y2为CRY2
Y3为CRY3
Y4为CRY4
R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
RY1选自H、D、卤素、C1-6烷基或C1-6卤代烷基;
RY2选自H、D、C1-6烷基或C1-6卤代烷基;
RY3选自H、D、卤素、ORa、C1-6烷基、C1-6卤代烷基或C3-6环烷基;
RY4选自H或D;
其中RY1和RY3中至少一个不为H或D;
其中Ra选自H、C1-6烷基或C1-6卤代烷基。
在更具体的实施方案中,本发明提供了上述式(VI-2)化合物,或其药学上可接受的盐、同位素变 体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
X为CR2bR2b’;
R2为Me;
R1b、R1b’以及它们连接的碳原子形成C=O;
Y1为CRY1
Y2为CRY2
Y3为CRY3
Y4为CRY4
R2b和R2b’独立地选自H或D;
RY1选自H、D、Cl、F、Me或CF3
RY2选自H、D或Et;
RY3选自H、D、Cl、F、Me、Et、OMe或环丙基;
RY4选自H或D;
其中RY1和RY3中至少一个不为H或D。
在更具体的实施方案中,本发明提供了上述化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
其中,
R2选自C1-6烷基或C1-6卤代烷基;
R1a选自H、D、卤素、CN、ORa或NRbRc
R2a选自H、D、卤素或CN;
R3a选自H或D;
R4a选自ORa或NRbRc
R5a选自H、D或CN;
或者,R3a、R4a以及它们连接的碳原子形成4-6元杂环基或5-6元杂芳基,其任选地被1、2、3或4个R’取代基取代;
其中R’选自H、D、C1-6烷基或C1-6卤代烷基;
Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基。
在更具体的实施方案中,本发明提供了上述式(VII)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
R2选自C1-6烷基或C1-6卤代烷基;
R1a为卤素,优选F;
R2a选自H、D或卤素;
R3a为H或D;
R4a为ORa
R5a为CN;
或者,R3a、R4a以及它们连接的碳原子形成呋喃基或二氢呋喃基,其任选地被1、2、3或4个R’取代基取代;
其中R’选自H、D、C1-6烷基或C1-6卤代烷基;
Ra选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-7环烷基或4-7元杂环基。
在更具体的实施方案中,本发明提供了上述式(VII)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
R2选自C1-4烷基或C1-4卤代烷基;
R1a为卤素,优选F;
R2a选自H、D或卤素;
R3a为H或D;
R4a为ORa
R5a为CN;
其中Ra选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-6环烷基或5-6元杂环基。
在更具体的实施方案中,本发明提供了化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中所述化合物选自以下表1和表2。
表1:本发明的代表性化合物

















表2:优选的本发明的代表性化合物






本发明化合物可包括一个或多个不对称中心,且因此可以存在多种立体异构体形式,例如,对映异构体和/或非对映异构体形式。例如,本发明化合物可为单独的对映异构体、非对映异构体或几何异构体(例如顺式和反式异构体),或者可为立体异构体的混合物的形式,包括外消旋体混合物和富含一种或多种立体异构体的混合物。异构体可通过本领域技术人员已知的方法从混合物中分离,所述方法包括:手性高压液相色谱法(HPLC)以及手性盐的形成和结晶;或者优选的异构体可通过不对称合成来制备。
本发明化合物可以互变异构体形式存在。互变异构体为因分子中某一原子在两个位置迅速移动而产生的官能团异构体,互变异构体是一种特殊的官能团异构体,一对互变异构体可以互相转换,但通常以比较稳定的一种异构体为其主要的存在形式。最主要的例子为烯醇式和酮式互变异构体。
本领域技术人员将理解,有机化合物可以与溶剂形成复合物,其在该溶剂中发生反应或从该溶剂中沉淀或结晶出来。这些复合物称为“溶剂合物”。当溶剂是水时,复合物称为“水合物”。本发明涵盖了本发明化合物的所有溶剂合物。
术语“溶剂合物”是指通常由溶剂分解反应形成的与溶剂相结合的化合物或其盐的形式。这个物理缔合可包括氢键键合。常规溶剂包括包括水、甲醇、乙醇、乙酸、DMSO、THF、***等。本文所述的化合物可制备成,例如,结晶形式,且可被溶剂化。合适的溶剂合物包括药学上可接受的溶剂合物且进一步包括化学计量的溶剂合物和非化学计量的溶剂合物。在一些情况下,所述溶剂合物将能够分离,例如,当一或多个溶剂分子掺入结晶固体的晶格中时。“溶剂合物”包括溶液状态的溶剂合物和可分离的溶剂合物。代表性的溶剂合物包括水合物、乙醇合物和甲醇合物。
术语“水合物”是指与水相结合的化合物。通常,包含在化合物的水合物中的水分子数与该水合物中该化合物分子数的比率确定。因此,化合物的水合物可用例如通式R×x H2O代表,其中R是该化合物,和x是大于0的数。给定化合物可形成超过一种水合物类型,包括,例如,单水合物(x为1)、低级水合物(x是大于0且小于1的数,例如,半水合物(R×0.5H2O))和多水合物(x为大于1的数,例如,二水合物(R×2H2O)和六水合物(R×6H2O))。
本发明化合物可以是无定形或结晶形式(多晶型)。此外,本发明化合物可以以一种或多种结晶形式存在。因此,本发明在其范围内包括本发明化合物的所有无定形或结晶形式。术语“多晶型物”是指特定晶体堆积排列的化合物的结晶形式(或其盐、水合物或溶剂合物)。所有的多晶型物具有相同的元素组成。不同的结晶形式通常具有不同的X射线衍射图、红外光谱、熔点、密度、硬度、晶体形状、光电性质、稳定性和溶解度。重结晶溶剂、结晶速率、贮存温度和其他因素可导致一种结晶形式占优。化合物的各种多晶型物可在不同的条件下通过结晶制备。
本发明还包括同位素标记的化合物(同位素变体),它们等同于式(I)所述的那些,但一个或多个原子被原子质量或质量数不同于自然界常见的原子质量或质量数的原子所代替。可以引入本发明化合物中的同位素的实例包括氢、碳、氮、氧、磷、硫、氟和氯的同位素,分别例如2H、3H、13C、11C、14C、15N、18O、17O、31P、32P、35S、18F和36Cl。含有上述同位素和/或其它原子的其它同位素的本发明化合物、其前体药物和所述化合物或所述前体药物的药学上可接受的盐都属于本发明的范围。某些同位素标记的本发明化合物、例如引入放射性同位素(例如3H和14C)的那些可用于药物和/或底物组织分布测定。氚、即3H和碳-14、即14C同位素是特别优选的,因为它们容易制备和检测。进而,被更重的同位素取代,例如氘、即2H,由于代谢稳定性更高可以提供治疗上的益处,例如延长体内半衰期或减少剂量需求,因而在有些情况下可能是优选的。同位素标记的本发明式(A)化合物及其前体药物一般可以这样制备,在进行下述流程和/或实施例与制备例所公开的工艺时,用容易得到的同位素标记的试剂代替非同位素标记的试剂。
此外,前药也包括在本发明的上下文内。本文所用的术语“前药”是指在体内通过例如在血液中水解转变成其具有医学效应的活性形式的化合物。药学上可接受的前药描述于T.Higuchi和V.Stella,Prodrugs as Novel Delivery Systems,A.C.S.Symposium Series的Vol.14,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987,以及D.Fleisher、S.Ramon和H.Barbra“Improved oral drug delivery:solubility limitations overcome by the use of prodrugs”,Advanced Drug Delivery Reviews(1996)19(2)115-130,每篇引入本文作为参考。
前药为任何共价键合的本发明化合物,当将这种前药给予患者时,其在体内释放母体化合物。通常通过修饰官能团来制备前药,修饰是以使得该修饰可以通过常规操作或在体内裂解产生母体化合物的方式进行的。前药包括,例如,其中羟基、氨基或巯基与任意基团键合的本发明化合物,当将其给予患者时,可以裂解形成羟基、氨基或巯基。因此,前药的代表性实例包括(但不限于)式(A)化合物的羟基、巯基和氨基官能团的乙酸酯/酰胺、甲酸酯/酰胺和苯甲酸酯/酰胺衍生物。另外,在羧酸(-COOH)的情况下,可以使用酯,例如甲酯、乙酯等。酯本身可以是有活性的和/或可以在人体体内条件下水解。合适的药学上可接受的体内可水解的酯基包括容易在人体中分解而释放母体酸或其盐的那些基团。
本发明还提供药物制剂,包含治疗有效量的式(A)化合物或其治疗学上可接受的盐和其药学上可接受的载体、稀释剂或赋形剂。所有这些形式都属于本发明。
药物组合物和试剂盒
在另一方面,本发明提供了药物组合物,其包含本发明化合物(还称为“活性组分”)和药学上可接受的赋形剂。在一些实施方案中,所述药物组合物包含有效量的本发明化合物。在一些实施方案中,所述药物组合物包含治疗有效量的本发明化合物。在一些实施方案中,所述药物组合物包含预防有效量的本发明化合物。
用于本发明的药学上可接受的赋形剂是指不会破坏一起调配的化合物的药理学活性的无毒载剂、佐剂或媒剂。可以用于本发明组合物中的药学上可接受的载剂、佐剂或媒剂包括(但不限于)离子交换剂、氧化铝、硬脂酸铝、卵磷脂、血清蛋白(如人类血清白蛋白)、缓冲物质(如磷酸盐)、甘氨酸、山梨酸、山梨酸钾、饱和植物脂肪酸的偏甘油酯混合物、水、盐或电解质(如硫酸鱼精蛋白)、磷酸氢二钠、磷酸氢钾、氯化钠、锌盐、硅胶、三硅酸镁、聚乙烯吡咯烷酮、基于纤维素的物质、聚乙二醇、羧甲基纤维素钠、聚丙烯酸酯、蜡、聚乙烯-聚氧丙烯-嵌段聚合物、聚乙二醇以及羊毛脂。
用于给予本发明化合物的合适制剂将对于本领域普通技术人员而言是显而易见的,并且包括例如片剂、丸剂、胶囊、栓剂、锭剂、糖锭剂、溶液(特别是注射(皮下、静脉内、肌内)和输注(注射剂)用溶液)、酏剂、糖浆、扁囊剂、乳液、吸入剂或可分散粉剂。一种或多种药物活性化合物的含量的范围应该是作为整体的组合物的0.1至90wt%、优选0.5至50wt%,即,其量足以实现以下指定的剂量范围。如有必要,指定的剂量可每天给药若干次。
本发明还包括试剂盒(例如,药物包装)。所提供的试剂盒可以包括本发明化合物、其它治疗剂,以及含有本发明化合物、其它治疗剂的第一和第二容器(例如,小瓶、安瓿瓶、瓶、注射器和/或可分散包装或其它合适的容器)。在一些实施方案中,提供的试剂盒还可以任选包括第三容器,其含有用于稀释或悬浮本发明化合物和/或其它治疗剂的药用赋形剂。在一些实施方案中,提供在第一容器和第二容器中的本发明化合物和其它治疗剂组合形成一个单位剂型。
给药
本发明提供的药物组合物可以通过许多途径给药,包括但不限于:口服给药、肠胃外给药、吸入给药、局部给药、直肠给药、鼻腔给药、口腔给药、***给药、通过植入剂给药或其它给药方式。例 如,本文使用的肠胃外给药包括皮下给药、皮内给药、静脉内给药、肌肉内给药、关节内给药、动脉内给药、滑膜腔内给药、胸骨内给药、脑脊髓膜内给药、病灶内给药、和颅内的注射或输液技术。
通常,给予有效量的本文所提供的化合物。按照有关情况,包括所治疗的病症、选择的给药途径、实际给予的化合物、个体患者的年龄、体重和响应、患者症状的严重程度,等等,可以由医生确定实际上给予的化合物的量。
当用于预防本发明所述病症时,给予处于形成所述病症危险之中的受试者本文所提供的化合物,典型地基于医生的建议并在医生监督下给药,剂量水平如上所述。处于形成具体病症的危险之中的受试者,通常包括具有所述病症的家族史的受试者,或通过遗传试验或筛选确定尤其对形成所述病症敏感的那些受试者。
还可以长期给予本文所提供的药物组合物(“长期给药”)。长期给药是指在长时间内给予化合物或其药物组合物,例如,3个月、6个月、1年、2年、3年、5年等等,或者可无限期地持续给药,例如,受试者的余生。在一些实施方案中,长期给药意欲在长时间内在血液中提供所述化合物的恒定水平,例如,在治疗窗内。
可以使用各种给药方法,进一步递送本发明的药物组合物。例如,在一些实施方案中,可以推注给药药物组合物,例如,为了使化合物在血液中的浓度提高至有效水平。推注剂量取决于通过身体的活性组分的目标全身性水平,例如,肌内或皮下的推注剂量使活性组分缓慢释放,而直接递送至静脉的推注(例如,通过IV静脉滴注)能够更加快速地递送,使得活性组分在血液中的浓度快速升高至有效水平。在其它实施方案中,可以以持续输液形式给予药物组合物,例如,通过IV静脉滴注,从而在受试者身体中提供稳态浓度的活性组分。此外,在其它实施方案中,可以首先给予推注剂量的药物组合物,而后持续输液。
口服组合物可以采用散装液体溶液或混悬剂或散装粉剂形式。然而,更通常,为了便于精确地剂量给药,以单位剂量形式提供所述组合物。术语“单位剂型”是指适合作为人类患者及其它哺乳动物的单元剂量的物理离散单位,每个单位包含预定数量的、适于产生所需要的治疗效果的活性物质与合适药学赋形剂。典型的单位剂量形式包括液体组合物的预装填的、预先测量的安瓿或注射器,或者在固体组合物情况下的丸剂、片剂、胶囊剂等。在这种组合物中,所述化合物通常为较少的组分(约0.1至约50重量%,或优选约1至约40重量%),剩余部分为对于形成所需给药形式有用的各种载体或赋形剂以及加工助剂。
对于口服剂量,代表性的方案是,每天一个至五个口服剂量,尤其是两个至四个口服剂量,典型地是三个口服剂量。使用这些剂量给药模式,每个剂量提供大约0.01至大约20mg/kg的本发明化合物,优选的剂量各自提供大约0.1至大约10mg/kg,尤其是大约1至大约5mg/kg。
为了提供与使用注射剂量类似的血液水平,或比使用注射剂量更低的血液水平,通常选择透皮剂量,数量为大约0.01至大约20%重量,优选大约0.1至大约20%重量,优选大约0.1至大约10%重量,且更优选大约0.5至大约15%重量。
从大约1至大约120小时,尤其是24至96小时,注射剂量水平在大约0.1mg/kg/小时至至少10mg/kg/小时的范围。为了获得足够的稳定状态水平,还可以给予大约0.1mg/kg至大约10mg/kg或更多的预载推注。对于40至80kg的人类患者来说,最大总剂量不能超过大约2g/天。
适于口服给药的液体形式可包括合适的水性或非水载体以及缓冲剂、悬浮剂和分散剂、着色剂、调味剂,等等。固体形式可包括,例如,任何下列组份,或具有类似性质的化合物:粘合剂,例如,微晶纤维素、黄蓍胶或明胶;赋形剂,例如,淀粉或乳糖,崩解剂,例如,褐藻酸、Primogel或玉米淀粉;润滑剂,例如,硬脂酸镁;助流剂,例如,胶体二氧化硅;甜味剂,例如,蔗糖或糖精;或调 味剂,例如,薄荷、水杨酸甲酯或橙味调味剂。
可注射的组合物典型地基于可注射用的无菌盐水或磷酸盐缓冲盐水,或本领域中已知的其它可注射的赋形剂。如前所述,在这种组合物中,活性化合物典型地为较少的组分,经常为约0.05至10%重量,剩余部分为可注射的赋形剂等。
典型地将透皮组合物配制为含有活性组分的局部软膏剂或乳膏剂。当配制为软膏剂时,活性组分典型地与石蜡或可与水混溶的软膏基质组合。或者,活性组分可与例如水包油型乳膏基质一起配制为乳膏剂。这种透皮制剂是本领域中公知的,且通常包括用于提升活性组分或制剂的稳定的皮肤渗透的其它组份。所有这种已知的透皮制剂和组份包括在本发明提供的范围内。
本发明化合物还可通过经皮装置给予。因此,经皮给药可使用贮存器(reservoir)或多孔膜类型、或者多种固体基质的贴剂实现。
用于口服给予、注射或局部给予的组合物的上述组份仅仅是代表性的。其它材料以及加工技术等阐述于Remington's Pharmaceutical Sciences,17th edition,1985,Mack Publishing Company,Easton,Pennsylvania的第8部分中,本文以引用的方式引入该文献。
本发明化合物还可以以持续释放形式给予,或从持续释放给药***中给予。代表性的持续释放材料的描述可在Remington's Pharmaceutical Sciences中找到。
本发明还涉及本发明化合物的药学上可接受的制剂。在一个实施方案中,所述制剂包含水。在另一个实施方案中,所述制剂包含环糊精衍生物。最常见的环糊精为分别由6、7和8个α-1,4-连接的葡萄糖单元组成的α-、β-和γ-环糊精,其在连接的糖部分上任选包括一个或多个取代基,其包括但不限于:甲基化的、羟基烷基化的、酰化的和磺烷基醚取代。在一些实施方案中,所述环糊精为磺烷基醚β-环糊精,例如,磺丁基醚β-环糊精,也称作Captisol。参见,例如,U.S.5,376,645。在一些实施方案中,所述制剂包括六丙基-β-环糊精(例如,在水中,10-50%)。
适应症
在MTAP缺失的肿瘤,开发MTA协同的PRMT5抑制剂可以为大量的肿瘤病人提供治疗获益。本发明中的化合物通过负调节肿瘤细胞内MTA结合的PRMT5的活性发挥治疗作用,尤其是对MTAP缺陷的细胞,或MTAP相关的各种肿瘤细胞。
在一些实施方案中,本发明所述的MTA协同的PRMT5抑制剂可以治疗多种癌症,包括但不限于肿瘤类型,例如恶性周围神经鞘瘤、间皮瘤、多形性胶质母细胞瘤、胰腺癌、胆管癌、***癌、乳腺癌、脑癌、皮肤癌、***、膀胱癌、星形细胞瘤、结肠直肠癌、子宫内膜癌、食道癌、胃癌、胸腺瘤、头颈癌、肝细胞癌、喉癌、肺鳞癌、肺腺癌、口腔癌、卵巢癌、肾癌和甲状腺癌以及肉瘤。
更具体地说,这些化合物可用于治疗:心脏:肉瘤(血管肉瘤、纤维肉瘤、横纹肌肉瘤、脂肪肉瘤等)、粘液瘤、横纹肌瘤、纤维瘤、脂肪瘤和畸胎瘤;肺:支气管癌(鳞状细胞、未分化小细胞、未分化大细胞、腺癌等)、肺泡(细支气管)癌、支气管腺瘤、肉瘤、淋巴瘤、软骨瘤错构瘤、间皮瘤;胃肠道:食管(鳞状细胞癌、腺癌、平滑肌肉瘤、淋巴瘤等)、胃(肿瘤、淋巴瘤、平滑肌肉瘤等)、胰腺(导管腺癌、胰岛素瘤、胰高血糖素瘤、胃泌素瘤、类癌瘤、血管瘤等)、小肠(腺癌、淋巴瘤、类癌瘤等)、卡波西肉瘤、平滑肌瘤、血管瘤、脂肪瘤、神经纤维瘤、纤维瘤等)、大肠(腺癌、管状腺瘤、绒毛状腺瘤、错构瘤、平滑肌瘤等);泌尿生殖道:肾脏(腺癌、肾母细胞瘤等)、膀胱和尿道(鳞状细胞癌,移行细胞癌,腺癌等),***(腺癌,肉瘤等),睾丸(***瘤,畸胎瘤,胚胎癌,畸胎癌,绒癌、肉瘤、***癌、纤维瘤、纤维腺瘤、腺瘤样肿瘤、脂肪瘤等);肝脏:肝细胞瘤、胆管癌、肝母细胞瘤、血管肉瘤、肝细胞腺瘤、血管瘤;胆道:胆囊癌、壶腹癌、胆管癌等;骨:成骨肉瘤、纤维肉瘤、恶性纤维组织细胞瘤、软骨肉瘤、尤文肉瘤、恶性淋巴瘤(网状细胞肉瘤等)、 多发性骨髓瘤、恶性巨细胞瘤脊索瘤、骨软骨瘤(骨软骨外生骨疣)、良性软骨瘤、软骨母细胞瘤、软骨粘液纤维瘤、骨样骨瘤和骨巨细胞瘤;神经***:颅骨(骨瘤、血管瘤、肉芽肿、黄色瘤、变形性骨炎等)、脑膜(脑膜瘤、脑膜肉瘤、神经胶质瘤等)、脑(星形细胞瘤、髓母细胞瘤、神经胶质瘤、室管膜瘤、生殖细胞瘤(松果体瘤等)、多形性胶质母细胞瘤、少突胶质细胞瘤、神经鞘瘤、视网膜母细胞瘤、先天性肿瘤等)、脊髓神经纤维瘤、脑膜瘤、神经胶质瘤,肉瘤等);妇科:子宫(子宫内膜癌等)、宫颈(***、瘤前宫颈发育不良等)、卵巢(卵巢癌、浆液性囊腺癌、粘液性囊腺癌、未分类癌等)、颗粒鞘细胞瘤、支持***瘤、无性细胞瘤、恶性畸胎瘤等)、外阴(鳞状细胞癌、上皮内癌、腺癌、纤维肉瘤、黑色素瘤等)、***(透明细胞癌、鳞状细胞癌、葡萄状肉瘤(胚胎性横纹肌肉瘤等)、输卵管癌等;血液学:血液(髓性白血病(急性和慢性),急性淋巴细胞白血病、慢性淋巴细胞白血病、弥漫性大B细胞淋巴瘤、套细胞淋巴瘤(MCL),滤泡性淋巴瘤、骨髓增生性疾病、多发性骨髓瘤、骨髓增生异常综合征等)、霍奇金病、非霍奇金淋巴瘤(恶性淋巴瘤)等;皮肤:恶性黑色素瘤、基底细胞癌、鳞状细胞癌、卡波西肉瘤、痣发育不良痣、脂肪瘤、血管瘤、皮肤纤维瘤、瘢痕瘤,牛皮癣等;肾上腺:神经母细胞瘤等。
联合用药
本发明所述的MTA协同的PRMT5抑制剂可以与其他药物联合治疗癌症,至少包含一种靶点药物/细胞活性调节剂,包括CDK4/6抑制剂,MAT2A抑制剂,MAPK1/MAPK3抑制剂,Type I PRMT抑制剂,EGFR抑制剂,SHP2抑制剂,泛KRAS抑制剂,KRASG12C抑制剂,RAF抑制剂,MEK抑制剂,ERK抑制剂,Bcl-2抑制剂,SOS1抑制剂,PARP抑制剂,MALT1抑制剂,MALT2抑制剂,BTK抑制剂,PI3K抑制剂,AKT抑制剂,FGFR抑制剂,DNA甲基转移酶(DNMT)抑制剂,EZH1/2抑制剂,EZH2抑制剂,Menin-MLL抑制剂,IDH1抑制剂,IDH2抑制剂,IDH1/2抑制剂,化疗药物,放射疗法,STING激动剂或免疫检查点抑制剂/调节剂等。
实施例
本文所用的原料或试剂为可购买到的或由本领域通常已知的合成方法制备。
实施例
1.实施例1的合成
1.1中间体1-1的合成
将N,O-二甲羟胺盐酸盐(8.81g),1-(3-二甲基氨基丙基)-3-乙基碳二亚胺(EDCI)(17.3g)和吡啶(30mL)加入到2-苄氧基乙酸(10g)的二氯甲烷(200mL)溶液中,室温搅拌2小时。二氯甲烷(100mL)稀释,1M盐酸(100mL)洗涤,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析(硅胶,石油醚/乙酸乙酯=20/1-1/1)纯化,得无色油状物1-1(11g,收率78.5%)。MS:M+H+=210。
1.2中间体1-2的合成
氮气保护下,将正丁基锂(2.4M,21.2mL)缓慢滴加到含有2,2,6,6-四甲基哌啶(7.48g)的-78℃四氢呋喃(80mL)溶液中,滴毕,反应液缓慢升温至0℃,然后再降温至-78℃。将6-氯烟酸(2g)的四氢呋喃(10mL)溶液缓慢滴加至上述反应液,并在该温度下搅拌90分钟。升温至-60℃搅拌30分钟,并在该温度下加入1-1(11g),然后在-30℃~-20℃搅拌3.5小时。加入10%的柠檬酸水溶液淬灭,乙酸乙酯(100mL)萃取。有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得淡黄色油状物粗品1-2(2g),直接用于下一步。MS:M+H+=306。
1.3中间体1-3的合成
0℃下,将三甲基硅烷基重氮甲烷(91.48g)加入到1-2(2g)的四氢呋喃(20mL)溶液中,室温搅拌2小时。加入水(50mL)淬灭,乙酸乙酯(50mL)萃取,有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,抽滤,滤液减压浓缩。残余物用柱层析(硅胶,石油醚/乙酸乙酯=10/1-1/1)纯化,得白色固体1-3(680mg,收率30.8%)。MS:M+H+=320。
1.4中间体1-4的合成
将1-甲基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-吡唑(487mg),无水碳酸钠(451mg),四(三苯基膦)钯(247mg)加入到1-3(680mg)的二氧六环和水(5:1,8mL)溶液中,氮气保护下100℃搅拌2小时。冷却至室温,减压浓缩,得棕色固体化合物粗品1-4(1g),直接用于下一步反应。MS:M+H+=352。
1.5中间体1-5的合成
将水合肼(1mL)加入到1-4(1g)的无水乙醇(10mL)溶液中,70℃搅拌1小时。冷却至室温,减压浓缩,残余物用C18柱(乙腈/水=40/1-1/1)纯化,得白色固体1-5(720mg,收率71.4%)。MS:M+H+=348。
1.6中间体1-6的合成
-10℃下,将三氯化硼(9mL)滴加到1-5(720mg)的二氯甲烷(7mL)溶液中,搅拌1小时。加入冰的碳酸氢钠饱和水溶液(10mL),室温搅拌0.5小时,减压浓缩,残余物经高效液相制备纯化(柱:Gemini 5u C18 150 x 21.2mm;流动相:[水(FA)-ACN];B%:5%-50%,50min),得白色固体1-6的甲酸盐(300mg,收率56.2%)。MS:M+H+=258。1H NMR(400MHz,DMSO-d6)δppm 12.72(s,1H),9.33(d,J=0.8Hz,1H),8.45(s,1H),8.13(d,J=0.6Hz,1H),8.04(d,J=0.9Hz,1H),5.52(t,J=6.1Hz,1H),4.68(d,J=5.9Hz,2H),3.89(s,3H).
1.7中间体1-7的合成
偶氮二甲酸二异丙酯(313mg)加入到1-6(200mg)和三苯基膦(407mg),邻苯二甲酰亚胺(227mg)的二氯甲烷(3mL)溶液中,室温搅拌1小时。减压浓缩,得到白色固体化合物粗品1-7(150mg),直接用于下一步反应。MS:M+H+=387。
1.8化合物1的合成
水合肼(3mL)加入到粗品1-7(150mg)的无水乙醇(2mL)溶液中,70℃搅拌1小时。冷却至室温,减压浓缩,残余物经高效液相制备纯化(柱:Gemini 5u C18 150 x 21.2mm;流动相:[水(FA)-ACN];B%:5%-50%,50min),得白色固体1的甲酸盐(3.6mg,收率7.6%)。MS:M+H+=257.1。1H NMR(400MHz,CD3OD-d4)δppm 9.38(d,J=0.8Hz,1H),8.38(s,1H),8.28(s,1H),8.14(d,J=0.5Hz,1H),7.86(d,J=0.8Hz,1H),4.45(s,2H),3.89(s,3H).
2.实施例2的合成
2.1中间体2-1的合成
将四(三苯基膦)钯(141mg),1-甲基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-吡唑(507mg)和碳酸钾(1.1g)加入到2,6-二氯烟酸甲酯(500mg)的四氢呋喃(15mL)和水(3mL)混合溶液中。氮气保护下,80℃搅拌5小时。冷却至室温,加入水(30mL),乙酸乙酯(20mL*3)萃取,合并的有机相用饱和氯化钠溶液(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析(硅胶,石油醚/乙酸乙酯=1/1-5/1)纯化,得黄色油状化合物2-1(380mg,收率62.3%)。MS:M+H+=252.0,253.9
2.2中间体2-2的合成
将三丁基(1-乙氧基乙烯基)锡(454mg)和四(三苯基膦)钯(67.9mg)加入到2-1(250mg)的N,N-二甲基甲酰胺(10mL)溶液中,氮气保护下,100℃搅拌1小时。冷却至室温,加水(50mL),乙酸乙酯(20 mL*3)萃取,合并的有机相用饱和氯化钠溶液(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析(硅胶,石油醚/乙酸乙酯=20/1-1/1)纯化,得白色固体2-2(100mg,收率35%)。MS:M+H+=288.1。
2.3中间体2-3的合成
将N-溴代丁二酰亚胺(74mg)加入2-2(100mg)的四氢呋喃(5mL)和水(0.3mL)溶液中,室温下搅拌1小时。减压浓缩,加入乙酸乙酯(30mL),无水硫酸钠干燥,过滤,滤液减压浓缩,得到黄色固体2-3(100mg),直接用于下一步反应。MS:M+H+=338.1,340.0。
2.4中间体2-4的合成
0℃下,将叠氮钠(24mg)加入2-3(100mg)的N,N-二甲基甲酰胺(10mL)溶液中,0℃搅拌30分钟。加水(50mL),乙酸乙酯(20mL*3)萃取,合并的有机相用饱和氯化钠溶液(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用柱层析(硅胶,二氯甲烷/甲醇=100/0–40/1)纯化,得到白色固体2-4(71mg,收率84.3%)。MS:M+H+=301.1。
2.5中间体2-5的合成
将水合肼(135mg)加入到2-4(71mg)的乙醇(5mL)溶液中。70℃搅拌3小时。冷却至室温,减压浓缩,得到黄色固体2-5(60mg),直接用于下一步反应。MS:M+H+=283.1。
2.6化合物2的合成
10%Pd/C(60mg),2-5(60mg)和甲醇(10mL)混合液在1个大气压氢气环境下室温搅拌3小时。减压过滤,用热的二甲基亚砜淋洗,滤液经高效液相色谱纯化(柱:Gemini 5u C18 150 x 21.2mm;流动相:ACN-H2O(0.1%FA);B%:10%-40%;15min),得类白色固体2的甲酸盐(1.7mg,收率2.9%)。MS:M+H+=257.2,1HNMR(400MHz,DMSO-d6)δppm 8.63(d,J=8.5Hz,1H),8.49(s,1H),8.28(s,1H),8.13(d,J=8.5Hz,1H),4.67(s,2H),4.02(s,3H).
3.实施例3的合成
3.1中间体3-1的合成
将三丁基(1-乙氧基乙烯基)锡(3.9g)和双(三苯基膦)二氯化钯(0.63g)加入到2,4-二氯嘧啶-5-甲酸乙酯(2g)的N,N-二甲基甲酰胺(20mL)溶液中,氮气保护下70℃搅拌2小时。反应液倒入冰-水(100mL),乙酸乙酯(50mL*3)萃取,合并的有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析(硅胶,二氯甲烷/甲醇=10/1)纯化,得黄色固体3-1(1.7g,收率70%)。MS:M+H+=256.9。1HNMR(400MHz,CDCl3)δppm 8.70(s,1H),5.38(d,J=2.8Hz,1H),4.61(d,J=2.9Hz,1H),4.36(q,J=7.2Hz,2H),3.90(q,J=7.0Hz,2H),1.37(t,J=7.2Hz,3H),1.33(t,J=7.0Hz,3H).
3.2中间体3-2的合成
将1-甲基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-吡唑(1.47g),四(三苯基膦)钯(0.54g)和碳酸钾(1.3g)加入到3-1(1.2g)的二氧六环(10mL)和水(2.5mL)溶液中,氮气保护下100℃搅拌2小时。冷却至室温,加水(50mL),乙酸乙酯(50mL*3)萃取,合并的有机相用饱和氯化钠溶液(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析(硅胶,石油醚/乙酸乙酯=20/1-3/1)纯化,得黄色固体3-2(1g,收率65.96%)。MS:M+H+=302.9。
3.3中间体3-3的合成
将N-溴代丁二酰亚胺(453.3mg)加入到3-2(0.7g)的四氢呋喃(7.5mL)和水(0.5mL)溶液中,室温搅拌16小时。加水(50mL),乙酸乙酯(50mL*3)萃取,合并的有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析(硅胶,石油醚/乙酸乙酯=100/0-4/1)纯化,得黄色固体3-3(0.6g,收率69.7%)。MS:M+H+=352.8,354.7。
3.4中间体3-4的合成
0℃下,将叠氮化钠(82.82mg)加入到3-3(300mg)的N,N-二甲基甲酰胺(5mL)溶液中,0℃搅拌1小时。加水(30mL),乙酸乙酯(30mL*3)萃取,合并的有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到黄色固体3-4(200mg),直接用于下一步反应。MS:M+H+=316.0。
3.5中间体3-5的合成
将水合肼(63.51mg)加入到3-4(200mg)的乙醇(5mL)溶液中。50℃搅拌3小时,加水(20mL),乙酸乙酯(40mL*3)萃取,有机相用饱和氯化钠溶液(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得黄色固体3-5(100mg)直接用于下一步反应。MS:M+H+=284.1。
3.6化合物3的合成
将三苯基膦(37.57mg)加入到3-5(50mg)的四氢呋喃(2mL)和水(0.4mL)混合物中,室温搅拌16小时。减压浓缩,残余物经高效液相色谱纯化(柱:-Xbridge-C18 150 x 19mm,5um;流动相:[水(0.05%FA)-ACN];B%:2%-20%,20min),得类白色固体3的甲酸盐(15mg,收率35%)。MS:M+H+=258.2.1H NMR(400MHz,DMSO-d6)δppm 9.54(s,1H),8.62(s,1H),8.20(d,J=0.7Hz,1H),4.06(s,2H),3.95(s,3H).
4.实施例4的合成
4.1中间体4-1的合成
0℃氮气保护下,将浓硫酸(2.5mL)滴加到3-溴-5-氯吡啶-2-羧酸(5g)的甲醇(50mL)溶液中,70℃搅拌16小时。冷却至室温,倒入冰-水混合物(200mL)中,乙酸乙酯(50mL*3)萃取,合并的有机相用饱和食盐水(100mL x 2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得白色固体4-1(5g,收率94%)。MS:M+H+=252。
4.2中间体4-2的合成
将三丁基(1-乙氧基乙烯基)锡(1.5g)和二(三苯基膦)二氯化钯(0.55g)加入到4-1(1g)的N,N-二甲基甲酰胺(10mL)溶液中,氮气保护下100℃搅拌2小时。冷却至室温,减压浓缩,残余物用柱层析(硅胶,石油醚/乙酸乙酯=100/1-30/1)纯化,得白色固体4-2(0.7g,收率73%)。MS:M+H+=242。
4.3中间体4-3的合成
室温下,将N-溴代丁二酰亚胺(391mg)加入到4-2(530mg)的四氢呋喃和水(15:1,10mL)混合溶液中,室温搅拌2小时。减压浓缩,残余物加入乙酸乙酯(30mL),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用柱层析(硅胶,石油醚/乙酸乙酯=100/1-4/1)纯化,得白色固体4-3(500mg,收率77%)。MS:M+H+=291.9,294。
4.4中间体4-4的合成
冰-水浴下,将叠氮基三甲基硅烷(395mg)和碳酸钾(395mg)加入到4-3(500mg)的无水乙腈(10mL)溶液中,室温搅拌1小时。减压过滤,滤饼用乙酸乙酯(20mL)淋洗,滤液减压浓缩,残余物用柱层析(硅胶,石油醚/乙酸乙酯=100/1-4/1)纯化,得白色固体4-4(260mg,收率59%)。MS:M+H+=255。
4.5中间体4-5的合成
室温下,将水合肼(102mg)加入到4-4(260mg)的乙醇(25mL)溶液中,80℃搅拌2小时。冷却至室温,减压浓缩,残余物用柱层析(硅胶,二氯甲烷/甲醇=100/0-100/2)纯化,得白色固体4-5(150mg,收率62%)。MS:M+H+=237。
4.6化合物4-6的合成
将三苯基膦(250mg)和二碳酸二叔丁酯(277mg)加入到4-5(150mg)的四氢呋喃(6mL)和水(2mL)混合溶液中,80℃搅拌1小时。冷却至室温,减压浓缩,残余物用柱层析(硅胶,二氯甲烷/甲醇=100/0 -100/2)纯化,得白色固体4-6(50mg,收率37%)。MS:M+H+=311。
4.7化合物4-7的合成
将1-甲基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-吡唑(34mg),无水碳酸钾(27mg),1,1'-二(二苯膦基)二茂铁二氯化钯(20mg)加入到4-6(40mg)的二氧六环和水(4:1,2mL)混合溶液中,氮气保护下100℃搅拌1小时。冷却至室温,减压浓缩,残余物加入水(10mL)和乙酸乙酯(10mL),减压过滤得灰色固体粗品4-7(30mg),直接进行下一步。MS:M+H+=357。
4.8化合物4的合成
0℃下,将三氟乙酸(1mL)加入到4-7(30mg)的二氯甲烷(3mL)溶液中,0℃下搅拌1小时。减压浓缩,残余物经高效液相色谱纯化(柱:Gemini 5u C18 150 x 21.2mm;流动相:[水(0.1%TFA)-ACN];B%:10%-50%,15min),得白色固体4的三氟乙酸盐(14mg,收率66%)。MS:M+H+=256.9.1H NMR(400MHz,DMSO-d6)δppm 13.09(s,1H),9.39(d,J=2.0Hz,1H),8.58(s,1H),8.48(d,J=2.1Hz,1H),8.42(s,2H),8.28(d,J=0.5Hz,1H),4.50(d,J=5.3Hz,2H),3.94(s,3H).
5.实施例5的合成
5.1中间体5-1的合成
将1-甲基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-吡唑(1.1g),[1,1'-双(二苯基膦)二茂铁]二氯化钯(338mg)和碳酸氢钠(811mg)加入到4,6-二氯哒嗪-3-羧酸甲酯(1g)的1,4-二氧六环(15mL)和水(3mL)溶液中,氮气保护下100℃搅拌2小时。冷却至室温,加入水(50mL),乙酸乙酯(20mL*3)萃取,合并的有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析(硅胶,石油醚/乙酸乙酯=1/1-5/1),纯化得黄色固体5-1(500mg,收率41.3%)。MS:M+H+=253.1,255.1
5.2中间体5-2的合成
将三丁基(1-乙氧基乙烯基)锡(1.1g)和双(三苯基膦)二氯化钯(139.2mg)加入到5-1(500mg)的N,N-二甲基甲酰胺(10mL)溶液中,氮气保护下80℃搅拌1小时。冷却至室温,加水(60mL),乙酸乙酯(30mL*3)萃取,合并的有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析(硅胶,石油醚/乙酸乙酯=20/1-1/1),纯化得白色固体5-2(230mg,收率40.2%)。MS:M+H+=289.2。
5.3中间体5-3的合成
将N-溴代丁二酰亚胺(62mg)加入5-2(100mg)的四氢呋喃(5mL)和水(0.3mL)溶液中。室温搅拌1小时,减压浓缩,残余物加入乙酸乙酯(60mL),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用 柱层析(硅胶,石油醚/乙酸乙酯=100/1-4/1)纯化,得到黄色固体5-3(86mg,收率73.5%)。MS:M+H+=339.1,341.1。
5.4中间体5-4的合成
0℃下,将碳酸钾(70mg)和叠氮基三甲基硅烷(47mg)加入5-3(86mg)的乙腈(5mL)溶液中,0℃搅拌半小时。减压过滤,乙酸乙酯(20mL)淋洗,滤液减压浓缩,残余物用柱层析(硅胶,石油醚/乙酸乙酯=100/1-3/1)纯化,得到白色固体5-4(30mg,收率36.2)。MS:M+H+=302.1。
5.5中间体5-5的合成
将水合肼(46mg)加入5-4(30mg)的乙醇(5mL)溶液中。70℃搅拌3小时。减压浓缩,得到黄色固体5-5(27mg),直接用于下一步反应。MS:M+H+=284.1。
5.6化合物5的合成
将三苯基膦(60mg)加入5-5(27mg)的四氢呋喃(3mL)和水(1mL)溶液中,80℃搅拌5小时。减压浓缩,残余物经高效液相色谱纯化(柱:Gemini-C18 150 x 21.2mm,5um;流动相:ACN--H2O(0.1%FA);B%:2%-10%,20min),得类白色固体5的甲酸盐(5.2mg,收率4.1%)。MS:M+H+=258.1.1H NMR(400MHz,MeOD-d6)δppm 8.52(s,1H),8.30(s,1H),8.25(s,1H),4.58(s,2H),4.00(s,3H).
6.实施例6的合成
6.1中间体6-1的合成
-78℃氮气保护下,将2,2,6,6-四甲基哌啶(13.5g)缓慢滴加到正丁基锂(2.4M,53mL)的四氢呋喃(100mL)溶液中,滴毕,缓慢升温至0℃。降温至-78℃,滴加6-氯烟酸(5g)的四氢呋喃(10mL)溶液,搅拌90分钟。升温至-60℃搅拌30分钟,缓慢滴加N-甲氧基-N-甲基乙酰胺(13.1g)的四氢呋喃(20mL)溶液,-30℃~-20℃搅拌3.5小时。加入10%的柠檬酸水溶液(500mL)淬灭,乙酸乙酯(100mL*3)萃取。合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得淡黄色油状物粗品6-1(7g),直接用于下一步反应。MS:M+H+=199.9。
6.2中间体6-2的合成
0℃下,将三甲基硅烷基重氮甲烷(1M,10mL)加入到6-1(7g)的四氢呋喃(50mL)溶液中,室温搅拌1小时。加入水(200mL)淬灭,用乙酸乙酯(50mL*3)萃取,合并的有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用柱层析(硅胶,石油醚/乙酸乙酯=100/1-8/1)纯化,得白色固体6-2(2.7g,收率40%,共两步)。MS:M+H+=214。
6.3中间体6-3的合成
将N-碘代丁二酰亚胺(3g)和对甲苯磺酸(0.3g)加入到6-2(1.8g)的乙酸乙酯(80mL)溶液中,80℃搅拌24小时。冷却至室温,加水(200mL)洗涤,水相用乙酸乙酯(50mL*3)萃取,合并的有机相用饱和氯化钠溶液(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析(硅胶,石油醚/乙酸乙酯=100/0-10/1)纯化,得类白色固体6-3(2.6g,收率90%)。MS:M+H+=338。
6.4中间体6-4的合成
0℃下,将叠氮钠(290mg)加入到6-3(1.5g)的N,N-二甲基甲酰胺(10mL)溶液中,0℃搅拌30分钟。加水(100mL)稀释,乙酸乙酯(30mL*3)萃取,合并的有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析(硅胶,石油醚/乙酸乙酯=100/0-8/1)纯化,得白色固体6-4(0.7g,收率52%)。MS:M+H+=299,301。
6.5中间体6-5的合成
将水合肼(128mg)加入到6-4(0.6g)的乙醇(5mL)溶液中,80℃搅拌1小时。冷却至室温,减压浓缩,残余物用柱层析(硅胶,二氯甲烷/甲醇=100/0-50/1)纯化,得白色固体6-5(0.35g,收率62%)。MS:M+H+=281,283。
6.6中间体6-6的合成
将三苯基膦(489mg)和二碳酸二叔丁酯(540mg)加入到6-5(350mg)的四氢呋喃(15mL)和水(3mL)混合溶液中,80℃搅拌1小时。冷却至室温,减压浓缩,残余物用柱层析(硅胶,二氯甲烷/甲醇=100/0-30/1)纯化,得白色固体6-6(110mg,收率24%)。MS:M+H+=355,357。
6.7中间体6-7的合成
将2-溴甲基苯甲酸甲酯(2.36g)和碳酸钾(2.84g)加入到1-甲基-5-氨基吡唑(1g)的N,N-二甲基甲酰胺(10mL)溶液中,室温搅拌2小时。加入水(200mL),乙酸乙酯(100mL*3)萃取,合并的有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析(硅胶,石油醚/乙酸乙酯=5/1-1/1)纯化,得白色固体化合物6-7(1.3g,收率52.4%)。MS:M+H+=176。
6.8中间体6-8的合成
将碳酸钾(1.69g)加入到6-7(1.3g)的N,N-二甲基甲酰胺(15mL)溶液中,80℃搅拌2小时。冷却至室温,加水(150mL),乙酸乙酯(100mL*3)萃取,合并的有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析(硅胶,二氯甲烷/甲醇=20/1-1/1)纯化,得无色油状物6-8(570mg,收率39.3%)。MS:M+H+=214。
6.9中间体6-9的合成
将N-溴代丁二酰亚胺(91.8mg)加入到6-8(100mg)的乙腈(5mL)溶液中,40℃搅拌2小时。冷却至室温,加水(50mL),乙酸乙酯(50mL*3)萃取,合并的有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用柱层析(硅胶,石油醚/乙酸乙酯=10/1-1/1)纯化,得白色固体6-9(123mg,产率:85.3%)。MS:M+H+=292。
6.10中间体6-10的合成
-78℃氮气保护下,将正丁基锂(2.4M,0.65mL)缓慢滴加到6-9(300mg)的无水四氢呋喃(5mL)溶 液中,-78℃搅拌0.5小时,滴加三丁基氯化锡(1g),缓慢升至室温,搅拌2小时。降至0℃,加入饱和氯化铵水溶液(1mL)淬灭,加水(30mL)稀释,乙酸乙酯(20mL*3)萃取,合并的有机相用饱和氯化钠溶液(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用柱层析(硅胶,石油醚/乙酸乙酯=100/0-3/1)纯化,得到黄色液体6-10(130mg,收率25%)。MS:M+H+=504。
6.11中间体6-11的合成
将6-10(30mg)和四(三苯基膦)钯(20mg)加入到6-6(100mg)的N,N-二甲基甲酰胺(1mL)溶液中,氮气保护下110℃搅拌4小时。冷却至室温,减压浓缩,残余物用薄层层析板(二氯甲烷/甲醇=20/1)纯化,得黄色固体6-11(10mg,收率62.67%)。MS:M+H+=488。
6.12化合物6的合成
0℃下,将三氟乙酸(0.5mL)加入到6-11(10mg)的二氯甲烷(1mL)溶液中,0℃搅拌1小时。减压浓缩,残余物经高效液相色谱纯化(柱:AZZOTA C18 100A 10um;流动相:[水(0.1%NH3)-ACN];B%:10%-35%,17min),得白色固体6(1.5mg,收率18%)。MS:M+H+=388.2.1HNMR(400MHz,MeOD-d4)δppm 9.16(s,1H),8.39(s,1H),7.94(s,1H),7.90(d,J=7.7Hz,1H),7.80-7.76(m,1H),7.70(d,J=7.8Hz,1H),7.62(t,J=7.3Hz,1H),4.24(s,2H),3.86(s,3H).
7.实施例7的合成
7.1化合物7的合成
10%钯炭(18.78mg),3-5(50mg)和甲醇(5mL)混合溶液在1个大气压氢气环境下室温搅拌16小时。减压浓缩,二氯亚砜热溶解,过滤,有机相经高效液相色谱纯化(column:Gemini-C18 150 x 21.2mm,5um;mobile phase:[H2O(0.1%TFA)-ACN];B%:2%-100%,20min)得类白色固体7的三氟乙酸盐(20mg,收率40%)。LC-MS m/z(ESI):260.2[M+H]+.1H NMR(400MHz,DMSO-d6)δppm 12.76(s,1H),8.66(s,1H),8.29(s,1H),8.14(s,3H),7.99(s,1H),4.38(s,2H),4.03(d,J=5.5Hz,2H),3.89(s,3H).
8.实施例8的合成
8.1中间体8-1的合成
在-78℃,将正丁基锂(2.8mL,1M)缓慢滴加到5-溴-3-甲基异噻唑(1.0g)的四氢呋喃(50mL)溶液中,控制温度低于-70℃搅拌10分钟。将三丁基氯化锡(2.4g)的四氢呋喃(10mL)溶液缓慢加到反应液中,控制温度低于-70℃搅拌2小时。升至0℃,加入饱和氯化铵溶液(5mL)淬灭,加入水(100mL),乙酸乙酯(100mL×3)萃取,有机相用饱和氯化钠溶液(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=5/1)纯化得黄色油状物8-1(1.6g,收率66.0%)。
8.2中间体8-2的合成
将2,6-二氯烟酸甲酯(724.0mg)和二(三苯基膦)二氯化钯(235.0mg)加入到化合物8-1(1.3g)的N,N-二甲基甲酰胺(15mL)溶液中,在80℃搅拌2小时。冷却至室温,加入水(100mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=2/1)纯化得黄色固体8-2(550.0mg,收率57.0%)。LC-MS m/z(ESI):269.0[M+H]+
8.3中间体8-3的合成
将三丁基(1-乙氧基乙烯)锡(961.0mg)和四(三苯基膦)钯(236.0mg)加入到化合物8-2(550.0mg)的N,N-二甲基甲酰胺(15mL)溶液中,在100℃搅拌2小时。冷却至室温,加入水(100mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=5/1)纯化得黄色固体8-3(500.0mg,收率80.0%)。LC-MS m/z(ESI):305.1[M+H]+
8.4中间体8-4的合成
将N-溴代丁二酰亚胺(351.0mg)加入到化合物8-3(500.0mg)的四氢呋喃和水(10mL,V/V=15:1)混合溶液中,室温搅拌30分钟。加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=2/1)纯化得黄色固体8-4(510.0mg,收率87.0%)。LC-MS m/z(ESI):354.9[M+H]+
8.5中间体8-5的合成
将叠氮基三甲基硅烷(324.0mg)和碳酸钾(389.0mg)加入到化合物8-4(200.0mg)的乙腈(20mL)溶液中,在40℃搅拌16小时。冷却至室温,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=2/1)纯化得黄色固体8-5(120.0mg,收率67.0%)。LC-MS m/z(ESI):318.9[M+H]+
8.6中间体8-6的合成
将水合肼(15.0mg)加入到化合物8-5(120.0mg)的乙醇(5mL)溶液中,在70℃搅拌3小时。冷却至室温,减压浓缩。残余物经硅胶柱层析(洗脱剂:二氯甲烷/甲醇=10/1)纯化得白色固体8-6(70.0mg,收率75.0%)。LC-MS m/z(ESI):300.0[M+H]+
8.7化合物8的合成
将三苯基膦(88.0mg)加入到化合物8-6(70.0mg)的四氢呋喃和水(5mL,V/V=5:1)混合溶液中,室温搅拌2小时。反应液减压浓缩。残余物经高效液相色谱(柱:Gemini 5u C18 150 x 21.2mm;mobile phase:ACN--H2O(0.05%NH4OH);B%:2%-10%,11min)纯化得白色固体8(23.8mg,产率43.0%)。LC-MS m/z(ESI):274.1[M+H]+1H NMR(400MHz,CDCl3)δppm 8.71(d,J=8.4Hz,1H),8.40(d,J=8.4Hz,1H),8.28(s,1H),8.10(s,1H),4.19(s,2H),2.52(s,3H).
9.实施例9的合成
9.1中间体9-1的合成
将三丁基(1-乙氧基乙烯)锡(3.9g)和双三苯基磷二氯化钯(0.6g)加入到2,4-二氯-5-嘧啶甲酸乙酯(2.0g)的N,N-二甲基甲酰胺(20mL)溶液中,70℃搅拌16小时。反应液倒入冰水(100mL),乙酸乙酯(50mL×3)萃取,合并的有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:二氯甲烷/甲醇=0-10/1)纯化得黄色固体9-1(1.7g,收率70%)。LC-MS m/z(ESI):256.9[M+H]+1H NMR(400MHz,CDCl3)δppm 8.70(s,1H),5.38(d,J=2.8Hz,1H),4.61(d,J=2.9Hz,1H),4.36(q,J=7.2Hz,2H),3.90(q,J=7.0Hz,2H),1.37(t,J=7.2Hz,3H),1.33(t,J=7.0Hz,3H).
9.2中间体9-2的合成
将6-9(0.6g),醋酸钯(46.0mg),2-二环己基膦-2,6-二甲氧基-联苯(168.0mg)和三乙胺(1.0g)加入到二氧六环(10mL)溶液中,室温慢慢加入频那醇硼烷(1.0g)。氮气保护下70℃搅拌2小时。冷却至室温,加水(30mL),乙酸乙酯(20mL×3)萃取,有机相用饱和氯化钠溶液(10mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=1/1)纯化得黄色固体9-2(0.3g,收率43.0%)。LC-MS m/z(ESI):340.1[M+H]+
9.3中间体9-3的合成
将9-1(200.0mg),四三苯基膦钯(84.0mg)和碳酸钠(155.0mg)加入9-2(298.0mg)的二氧六环(4mL)和水(1mL)溶液中,氮气保护下100℃搅2小时。冷却至室温,加水(15mL),乙酸乙酯(10mL×3)萃取,有机相用饱和氯化钠溶液(10mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=2/1)纯化得棕色油状物9-3(190.0mg,收率60.0%)。LC-MS m/z(ESI):434.2[M+H]+
9.4中间体9-4的合成
将N-溴代丁二酰亚胺(85.0mg)加入到9-3(190.0mg)的四氢呋喃(3mL)和水(1mL)溶液中,室温搅拌1小时。加水(10mL),乙酸乙酯(10mL×3)萃取,有机相用饱和氯化钠溶液(10mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=3/1)纯化得黄色固体9-4(150.0mg,收率64.0%)。LC-MS m/z(ESI):483.9,485.9[M+H]+
9.5中间体9-5的合成
0℃下将叠氮基三甲基硅烷(71.4mg)加入到碳酸钾(150.0mg)和9-4(150.0mg)的乙腈(2mL)溶液中,0℃搅拌1小时。升至室温,过滤,滤液减压浓缩得到白色固体9-5(70.0mg),粗品未经纯化直接用于下一步反应。LC-MS m/z(ESI):447.1[M+H]+
9.6中间体9-6的合成
将水合肼(34.0mg)加入到9-5(70.0mg)的乙醇(2mL)溶液中。室温搅拌2小时,加水(8mL),过滤得白色固体9-6(50.0mg),粗品未经纯化直接用于下一步反应。LC-MS m/z(ESI):415.1[M+H]+
9.7化合物9的合成
将三苯基膦(63.3mg)加入到9-6(50.0mg)的四氢呋喃(1.5mL)和水(0.3mL)中,50℃搅拌2小时。冷却至室温。减压浓缩,残余物经高效液相色谱(column:-Xbridge-C18 150 x 19mm,5um;mobile phase:[H2O(0.05%NH3)-ACN];B%:2%-20%,20min)纯化得白色固体9(11.0mg,收率35.1%)。LC-MS m/z(ESI):389.2[M+H]+1H NMR(400MHz,DMSO-d6)δppm 9.49(s,1H),8.38(s,1H),7.87(d,J=8Hz,1H),7.81-7.74(m,2H),7.63(t,J=8Hz,1H),5.15(s,1H),4.96(s,1H),3.83(s,3H),3.27(d,J=8Hz,2H)。
10.实施例10的合成
10.1中间体10-1的合成
将2-溴甲基苯甲酸甲酯(1.5g)和碳酸钾(3.0g)加入到化合物3-甲基异-4-氨基噻唑盐酸盐(1.0g)的N,N-二甲基甲酰胺溶液(20mL)中,80℃搅拌5小时。冷却至室温,加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=3/1)纯化得白色固体10-1(1.3g,收率81.1%)。LC-MS m/z(ESI):231.1[M+H]+
10.2中间体10-2的合成
在-78℃下,将正丁基锂(1.8mL,2.5M)缓慢滴加到10-1(1.0g)的四氢呋喃溶液(20mL)中,控制温度低于-70℃搅拌10分钟。将三丁基氯化锡(2.1g)的四氢呋喃溶液(10mL)缓慢加到反应液中,控制温度低于-70℃搅拌2小时。升至0℃,加入饱和氯化铵溶液(5mL)淬灭,加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=4/1)纯化得白色固体10-2(800.0mg,收率38.2%)。LC-MS m/z(ESI):520.9[M+H]+
10.3中间体10-3的合成
将三丁基(1-乙氧基乙烯)锡(2.0g)和二(三苯基膦)二氯化钯(200.0mg)加入到2,4-二氯-5-嘧啶甲酸乙酯(1.0g)的N,N-二甲基甲酰胺溶液(30mL)中,80℃搅拌2小时。冷却至室温,加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=8/1)纯化得黄色固体10-3(400.0mg,收率34.1%)。LC-MS m/z(ESI):257.1[M+H]+
10.4中间体10-4的合成
将化合物10-2(600.0mg)和二氯[1,1'-双(耳叔丁基膦)二茂铁钯(II)(100.0mg)加入到化合物10-3(400.0mg)的N,N-二甲基甲酰胺(10mL)中,100℃搅拌2小时。冷却至室温,加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=1/1)纯化得黄色固体10-4(110.0mg,收率16.1%)。LC-MS m/z(ESI):451.1[M+H]+
10.5中间体10-5的合成
将N-溴代丁二酰亚胺(80.0mg)加入到化合物10-4(100.0mg)的四氢呋喃和水(5mL,V/V=15:1)混合溶剂中,室温搅拌30分钟。加入水(20mL),乙酸乙酯(30mL×3)萃取,有机相用饱和氯化钠溶液(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=1/1)纯化得黄色固体10-5(100.0mg,收率90.2%)。LC-MS m/z(ESI):501.0,503.0[M+H]+
10.6中间体10-6的合成
将叠氮基三甲基硅烷(100.0mg)和碳酸钾(50.0mg)加入到化合物10-5(100.0mg)的乙腈溶液(5mL)中,40℃搅拌16小时。冷却至室温,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=2/1)纯化得黄色固体10-6(50.0mg,收率54.2%)。LC-MS m/z(ESI):463.9[M+H]+
10.7中间体10-7的合成
将水合肼(50.0mg)加入到化合物10-6(50.0mg)的乙醇溶液(5mL)中,70℃搅拌3小时。冷却至室温,减压浓缩。残余物经硅胶柱层析(洗脱剂:二氯甲烷/甲醇=10/1)纯化得白色固体10-7(25.0mg,收率54.2%)。LC-MS m/z(ESI):431.9[M+H]+
10.8化合物10的合成
将三苯基膦(30.0mg)加入到化合物10-7(25.0mg)的四氢呋喃和水(5mL,V/V=5:1)混合溶剂中,室温搅拌2小时。反应液减压浓缩。残余物经高压液相制备(柱:Gemini 5u C18 150 x 21.2mm;mobile phase:ACN--H2O(0.05%TFA);B%:5%-45%,30min)纯化得白色固体10的三氟乙酸盐(1.4mg,产率6.4%)。LC-MS m/z(ESI):406.1[M+H]+1H NMR(400MHz,MeOD-d4)δppm 9.53(s,1H),7.83(d,J=7.6Hz,1H),7.73-7.68(m,1H),7.63(d,J=7.6Hz,1H),7.57(t,J=7.2Hz,1H),4.51(s,2H),3.75(s,2H),2.38(s,3H).
11.实施例11的合成
11.1中间体11-1的合成
将异吲哚啉-1-酮(1.0g),三-(二亚苯-BASE丙酮)二钯(344.0mg),4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(435.0mg)和碳酸铯(7.0g)加入到3-溴-4-碘吡啶(3.0g)的1,4-二氧六环(30mL)溶液中,氮气保护下70℃搅拌16小时。冷却至室温,加入水(30mL),乙酸乙酯(20mL×3)萃取,合并的有机相用饱和氯化钠溶液(10mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=5/1-1/1)纯化得黄色固体11-1(600.0mg,收率27.6%)。LC-MS m/z(ESI):288.9,290.9[M+H]+
11.2中间体11-2的合成
将六丁基二锡(12.3g),三环己基膦(0.8g),氯化锂(3.0g),三(二亚苄基丙酮)二钯(1.3g)加入到化合物6-6(5.0g)的1,4-二氧六环(20mL)溶液中,在100℃下搅拌16小时。冷却至室温,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=2/1)纯化得黄色油状物11-2(4.5g,收率71.4%)。LC-MS m/z(ESI):467.2[M+H]+
11.3中间体11-3的合成
将11-2(306.0mg)和四三苯基膦钯(50.0mg)加入到11-1(500.0mg)的N,N-二甲基甲酰胺(10mL)溶液中,110℃搅拌16小时。冷却至室温,加水(10mL),乙酸乙酯(20mL×3)萃取,合并的有机相用饱和氯化钠溶液(10mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:二氯甲烷/甲醇=20/1)纯化得黄色固体11-3(100.0mg,收率59.5%)。LC-MS m/z(ESI):485.0[M+H]+
11.4化合物11的合成
冰浴下,将三氟乙酸(1mL)加入11-3(100.0mg)的二氯甲烷(2mL)溶液中。室温搅拌1小时,饱和碳酸钠溶液调节pH=8,二氯甲烷(10mL×3)萃取,减压浓缩,残余物经高效液相色谱(柱:Xbridge-C18 150×19mm,5um;流动相:ACN--H2O(0.1%FA);B%:5%-10%,20min)得白色固体11的甲酸盐(35.0mg,收率44.3%)。LC-MS m/z(ESI):385.0[M+H]+1H NMR(400MHz,DMSO-d6)δppm12.76(s,1H),9.26(s,1H),9.00(s,1H),8.80(d,J=5.6Hz,1H),8.16(s,1H),7.79(d,J=5.6Hz,1H),7.67-7.66(m,2H),7.56–7.55(m,1H),7.49-7.44(m,1H),5.10(s,2H),3.80(s,2H).
12.实施例12的合成
12.1中间体12-1的合成
将2,6-二氯烟酸甲酯(400.0mg),碳酸钾(538.0mg)和四(三苯基膦)钯(200.0mg)加入到3-甲基-5-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1H-吡唑(1.3g)的四氢呋喃(10mL)和水(10mL)的混合溶液中,氮气保护下80℃搅拌3小时。冷却至室温,加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:二氯甲烷/甲醇=50/1)纯化得黄色固体12-1(400.0mg,收率81.6%)。LC-MS m/z(ESI):251.9[M+H]+
12.2中间体12-2的合成
将三丁基(1-乙氧基乙烯)锡(1.2g)和四(三苯基膦)钯(200.0mg)加入到化合物12-1(400.0mg)的N,N-二甲基甲酰胺(5mL)溶液中,100℃搅拌2小时。冷却至室温,减压浓缩,残余物用***(30mL)洗涤得黑色油状物12-2(600.0mg),粗品直接用于下一步反应。LC-MS m/z(ESI):288.2[M+H]+
12.3中间体12-3的合成
将N-溴代丁二酰亚胺(356.0mg)加入到化合物12-2(600.0mg)的四氢呋喃和水(10mL,V/V=15:1)混合溶液中,室温搅拌30分钟。加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=2/1)纯化得白色固体12-3(270.0mg,两步收率48.4%)。LC-MS m/z(ESI):337.9,339.9[M+H]+
12.4中间体12-4的合成
冰浴氮气氛围下,将叠氮基三甲基硅烷(177.0mg)和碳酸钾(212.0mg)加入到化合物12-3(260.0mg)的无水乙腈(20mL)溶液中,40℃搅拌16小时。冷却至室温,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=1/1)纯化得白色固12-4(130.0mg,收率56.5%)。LC-MS m/z(ESI):301.1[M+H]+
12.5中间体12-5的合成
将水合肼(50.0mg)加入到化合物12-4(130.0mg)的乙醇(5mL)溶液中,70℃搅拌1小时。冷却至室温,减压过滤,干燥得白色固体12-5(100.0mg),粗品直接用于下一步反应。LC-MS m/z(ESI):283.1[M+H]+
12.6化合物12的合成
将三苯基膦(150.0mg)加入到化合物12-5(100.0mg)的四氢呋喃和水(5mL,V/V=3:1)混合溶液中,室温搅拌5小时。加入***(30mL),减压过滤,滤饼用乙酸乙酯(10mL)和***(10mL)淋 洗,干燥后得白色固体12(46.5mg,产率46.8%)。LC-MS m/z(ESI):257.0[M+H]+1H NMR(400MHz,DMSO-d6)δppm 13.11(s,1H),8.56(d,J=8.0Hz,1H),8.30(d,J=8.0Hz,1H),6.80(s,1H),4.08(s,2H),2.32(s,3H).
13.实施例13的合成
13.1中间体13-1的合成
将2,6-二氯烟酸甲酯(700.0mg)和二(三苯基膦)二氯化钯(200.0mg)加入到化合物10-2(800.0mg)的N,N-二甲基甲酰胺溶液(15mL)中,80℃搅拌2小时。冷却至室温,加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=1/1)纯化得黄色固体13-1(300.0mg,收率49.1%)。LC-MS m/z(ESI):400.0[M+H]+
13.2中间体13-2的合成
将三丁基(1-乙氧基乙烯)锡(500.0mg)和四(三苯基膦)钯(120.0mg)加入到化合物13-1(300.0mg)的N,N-二甲基甲酰胺(10mL)中,100℃搅拌2小时。冷却至室温,加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=5/1)纯化得黄色固体13-2(300.0mg,收率92.1%)。LC-MS m/z(ESI):436.2[M+H]+
13.3中间体13-3的合成
将N-溴代丁二酰亚胺(200.0mg)加入到化合物13-2(300.0mg)的四氢呋喃和水(10mL,V/V=15:1)混合溶剂中,室温搅拌30分钟。加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=2/1)纯化得黄色固体13-3(200.0mg,收率60.3%)。LC-MS m/z(ESI):485.9,487.8[M+H]+
13.4中间体13-4的合成
将叠氮基三甲基硅烷(324.0mg)和碳酸钾(389.0mg)加入到化合物13-3(200.0mg)的乙腈溶液(10mL)中,40℃搅拌16小时。冷却至室温,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=2/1)纯化得黄色固体13-4(100.0mg,收率54.1%)。LC-MS m/z(ESI):448.9[M+H]+
13.5中间体13-5的合成
将水合肼(150.0mg)加入到化合物13-4(100.0mg)的乙醇溶液(5mL)中,70℃搅拌3小时。冷却至室温,减压浓缩。残余物经硅胶柱层析(洗脱剂:二氯甲烷/甲醇=10/1)纯化得白色固体13- 5(80.0mg,收率84.1%)。LC-MS m/z(ESI):300.0[M+H]+
13.6化合物13的合成
将三苯基膦(80.0mg)加入到化合物13-5(80.0mg)的四氢呋喃和水(5mL,V/V=5:1)混合溶剂中,室温搅拌2小时。反应液减压浓缩。残余物经高压液相色谱(柱:Gemini 5u C18 150 x 21.2mm;mobile phase:ACN--H2O(0.05%TFA);B%:5%-45%,30min)纯化得白色固体13的三氟乙酸盐(15.0mg,产率21%)。LC-MS m/z(ESI):405.0[M+H]+1H NMR(400MHz,DMSO-d6)δppm 13.23(s,1H),8.67(d,J=8.4Hz,1H),8.33(s,2H),7.86-7.82(m,2H),7.80-7.69(m,2H),7.62(t,J=7.6Hz,1H),4.91(s,2H),4.32(s,2H),2.37(s,3H).
14.实施例14的合成
14.1中间体14-1的合成
将三丁基(1-乙氧基乙烯)锡(16.0g)和二(三苯基膦)二氯化钯(3.1g)加入到甲基2-氯-6-甲氧基烟酸(8.0g)的N,N-二甲基甲酰胺(200mL)溶液中,100℃搅拌5小时。冷却至室温,加入水(200mL),乙酸乙酯(200mL×3)萃取,有机相用饱和氯化钠溶液(200mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=10/1)纯化得无色液体14-1(8.0g,收率85.1%)。LC-MS m/z(ESI):238.2[M+H]+
14.2中间体14-2的合成
将N-溴代丁二酰亚胺(9.0g)加入到化合物14-1(8.0g)的四氢呋喃和水(100mL,V/V=15:1)混合溶剂中,室温搅拌30分钟。加入水(100mL),乙酸乙酯(100mL×3)萃取,有机相用饱和氯化钠溶液(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=8/1)纯化得白色固体14-2(8.4g,收率87.1%)。LC-MS m/z(ESI):288.0,290.0[M+H]+
14.3中间体14-3的合成
将叠氮基三甲基硅烷(10.0g)和碳酸钾(7.0g)加入到化合物14-2(8.3g)的乙腈(100mL)溶液中,40℃搅拌5小时。冷却至室温,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=8/1)纯化得黄色固体14-3(6.5g,收率89.1%)。LC-MS m/z(ESI):251.1[M+H]+
14.4中间体14-4的合成
将水合肼(6.5g)加入到化合物14-3(6.5g)的乙醇(100mL)溶液中,70℃搅拌3小时。冷却至室温,减压浓缩。残余物经硅胶柱层析(洗脱剂:二氯甲烷/甲醇=10/1)纯化得白色固体14-4(5.8 g,收率86.3%)。LC-MS m/z(ESI):233.1[M+H]+
14.5中间体14-5的合成
在0℃下,依次将氢化钠(900.0mg,60%)和2-(三甲基硅烷基)乙氧甲基氯(8.0g)加入到化合物14-4(5.5g)的N,N-二甲基甲酰胺(50mL)溶液中,0℃搅拌1小时。加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=4/1)纯化得黄色固体14-5(4.5g,收率52.1%)。LC-MS m/z(ESI):385.1[M+Na]+
14.6中间体14-6的合成
将三苯基膦(3.5g)加入到化合物14-5(4.5g)的四氢呋喃和水(50mL,V/V=5:1)混合溶液中,室温搅拌2小时。反应液减压浓缩。再将二碳酸二叔丁酯(5.0g)加入到上述粗产物的四氢呋喃溶液中,室温搅拌1小时。反应液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=4/1)纯化得白色固体14-6(3.0g,收率55.1%)。LC-MS m/z(ESI):459.2[M+Na]+
14.7中间体14-7的合成
将碘化锂(1.0g)加入到化合物14-6(1.0g)的N,N-二甲基甲酰胺(10mL)溶液中,150℃搅拌4小时。冷却至室温,加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=4/1)纯化得黄色固体14-7(480.0mg,收率46.2%)。LC-MS m/z(ESI):445.2[M+Na]+
14.8中间体14-8的合成
在0℃下,将吡啶(50.0mg)和三氟甲磺酸酐(200.0mg)加入到化合物14-7(220.0mg)的二氯甲烷(5mL)溶液中,0℃搅拌2小时。加入水(10mL),二氯甲烷(10mL×3)萃取,有机相用饱和氯化钠溶液(10mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=4/1)纯化得白色固体14-8(180.0mg,收率62.2%)。LC-MS m/z(ESI):577.0[M+Na]+
14.9中间体14-9的合成
将2-(2-溴乙基)苯甲酸甲酯(1.0g)和碳酸钾(1.5g)加入到化合物3-甲基-4-氨基异噻唑盐酸盐(500mg)的N,N-二甲基甲酰胺溶液(20mL)中,80℃搅拌5小时。冷却至室温,加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=3/1)纯化得白色固体14-9(600.0mg,收率71.2%)。LC-MS m/z(ESI):[M+H]+
14.10中间体14-10的合成
在-78℃下,将正丁基锂(0.4mL,2.5M)缓慢滴加到14-9(200.0mg)的四氢呋喃溶液(20mL)中,控制温度低于-70℃搅拌10分钟。将三丁基氯化锡(500.0mg)的四氢呋喃溶液(2mL)缓慢加到反应液中,控制温度低于-70℃搅拌2小时。升至0℃,加入饱和氯化铵溶液(2mL)淬灭,加入水(50mL),乙酸乙酯(50mL×3)萃取,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=4/1)纯化得白色固体14-10(100.0mg,收率23.2%)。LC-MS m/z(ESI):535.0[M+H]+
14.11中间体14-11的合成
将化合物14-8(100.0mg)和甲磺酸[正丁基二(1-金刚烷基)膦](2-氨基-1,1'-联苯-2-基)钯(II)(20.0mg)加入到化合物14-10(90.0mg)的N,N-二甲基甲酰胺溶液(2mL)中,80℃搅拌2小时。冷却至室温,加入乙酸乙酯(30mL),水(20mL×3)萃取,有机相用饱和氯化钠溶液(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经硅胶柱层析(洗脱剂:二氯甲烷/甲醇=30/1)纯化得白色固体 14-11(60.0mg,收率57%)。LC-MS m/z(ESI):649.2[M+H]+
LC-MS m/z(ESI):385.1[M+Na]+
14.12化合物14的合成
将化合物14-11(60.0mg)的二氯甲烷和三氟乙酸(3mL,V/V=5:1)混合溶液室温搅拌2小时。反应液减压浓缩。将氨水(1mL)加入到上述粗产物的二氧六环(3mL)溶液中,50℃搅拌2小时,反应液减压浓缩。残余物经高压液相色谱(柱:Gemini 5u C18 150 x 21.2mm;mobile phase:ACN--H2O(0.05%TFA);B%:5%-50%,30min)纯化得白色固体14的三氟乙酸盐(4.1mg,产率11.2%)。LC-MS m/z(ESI):419.0[M+H]+1H NMR(400MHz,MeOD-d4)δppm 8.57(d,J=8.4Hz,1H),8.32(d,J=7.6Hz,1H),8.25(d,J=8.4Hz,1H),7.54-7.49(m,1H),7.42(t,J=7.2Hz,1H),7.31(d,J=7.6Hz,1H),4.53(s,2H),4.29(t,J=6.0Hz,2H),3.00(t,J=5.8Hz,2H),2.24(s,3H).
如下列表化合物,通过实施例11或14相同合成策略进行合成:














实验例1:在MTA存在时用放射性同位素Flash Plate技术测定本发明化合物的PRMT5:MEP50酶抑制活性
(1)配制1×反应缓冲液(50mM Tris 8.0(Sigma,Cat.No.93362),0.01%Tween-20(Sigma,Cat.No.P9416),1mM DTT(Sigma,Cat.No.D9779),50mM NaCl(Sigma,Cat.No.S9888)。
(2)受试化合物和阳性对照化合物测试浓度为10μM起始,3倍稀释10个浓度。将受试化合物以及阳性对照以DMSO稀释至待测浓度的100×(即1mM),然后受试化合物以此进行3倍稀释10个浓度,接下来以1×反应溶液稀释20倍(此时DMSO浓度为5%),最后各加入5μL至384孔反应板中待测,每个化合物设置复孔测试。Max孔和Min孔中转移5μL的5%DMSO。
(3)用1×反应缓冲液配制2.5倍终浓度的酶溶液。
(4)在化合物孔和阳性对照孔分别加10μL的2.5倍终浓度的酶溶液;在阴性对照孔中加10μL的1×反应缓冲液。
(5)1000rpm离心60秒,振荡混匀后室温孵育15分钟。
(6)用1×反应缓冲液配制2.5倍终浓度的3H-SAM,H4肽和MTA的混合溶液。
(7)加入10μL的2.5倍终浓度的步骤(6)的混合溶液,起始反应。
(8)将384孔板1000rpm离心60秒,振荡混匀后室温孵育60分钟。
(9)以1×反应缓冲液配制终止液(200uM SAM(Sigma,Cat.No.A7007))。
(10)加入5μL终止液,1000rpm离心60秒,将各孔所有溶液转至FlashPlate,室温孵育60分钟。
(11)用MicroBeta2读数。
(12)计算公式
抑制%=100-(RLU-Mean(NC))/(Mean(PC)-Mean(NC))*100
RLU:样品的发光值;
Mean(NC):阴性对照的平均发光值;
Mean(PC):阳性对照的平均发光值;
使用GraphPad Prism 5软件拟合剂量-效应曲线,通过‘log[inhibitor]vs.response-variable slope’程序计算IC50。
实验例2:不存在MTA时用放射性同位素Flash Plate技术测定本发明化合物的PRMT5:MEP50酶抑制活性
(1)配制1×反应缓冲液(50mM Tris 8.0(Sigma,Cat.No.93362),0.01%Tween-20(Sigma,Cat.No.P9416),1mM DTT(Sigma,Cat.No.D9779),50mM NaCl(Sigma,Cat.No.S9888)。
(2)受试化合物和阳性对照化合物测试浓度为10μM起始,3倍稀释10个浓度。将受试化合物以及阳性对照以DMSO稀释至待测浓度的100×(即1mM),然后受试化合物以此进行3倍稀释10个浓度,接下来以1×反应溶液稀释20倍(此时DMSO浓度为5%),最后各加入5μL至384孔反应板中待测,每个化合物设置复孔测试。Max孔和Min孔中转移5μL的5%DMSO。
(3)用1×反应缓冲液配制2.5倍终浓度的酶溶液。
(4)在化合物孔和阳性对照孔分别加10μL的2.5倍终浓度的酶溶液;在阴性对照孔中加10μL的1×反应缓冲液。
(5)1000rpm离心60秒,振荡混匀后室温孵育15分钟。
(6)用1×反应缓冲液配制2.5倍终浓度的3H-SAM和H4肽的混合溶液。
(7)加入10μL的2.5倍终浓度的步骤(6)的混合溶液,起始反应。
(8)将384孔板1000rpm离心60秒,振荡混匀后室温孵育60分钟。
(9)以1×反应缓冲液配制终止液(200uM SAM(Sigma,Cat.No.A7007))。
(10)加入5μL终止液,1000rpm离心60秒,将各孔所有溶液转至FlashPlate,室温孵育60分钟。
(11)用MicroBeta2读数。
(12)计算公式
抑制%=100-(RLU-Mean(NC))/(Mean(PC)-Mean(NC))*100
RLU:样品的发光值;
Mean(NC):阴性对照的平均发光值;
Mean(PC):阳性对照的平均发光值;
使用GraphPad Prism 5软件拟合剂量-效应曲线,通过‘log[inhibitor]vs.response-variable slope’程序计算IC50。
表3化合物对PRMT5抑制作用测试结果

实验结论:本发明化合物是MTA协同的PRMT5抑制剂,抑制作用显著。
实验例3:利用Caco-2细胞模型评价化合物的外排比
3.1仪器设备及材料
(1)Caco-2细胞购自美国典型菌种保藏中心(ATCC)。
(2)对照药***购自Sigma公司(St.Louis,MO)。地高辛购自MCE。米诺地尔购自中检所。
(3)FBS培养基购自Sigma,DMEM购自Corning公司(Cambridge,MA),非必需氨基酸(NEAA),Hank’s平衡盐溶液(HBSS)和胰蛋白酶/EDTA均购自赛默飞世尔。青霉素、链霉素购自索莱宝。
(4)HTS-96孔Transwell板和其他无菌耗材购自Corning公司。
(5)Millicell电阻测定***购自Millipore。Vision购自Nexcelom Bioscience。Infinite200PRO酶标仪购自Tecan。MTS2/4 orbital摇床购自IKA Labortechnik。
3.2试验设计
3.2.1细胞培养和种板
(1)使用含L-谷氨酰胺的高糖(4.5g/L)DMEM培养基,添加10%胎牛血清、0.1mg/mL链霉素和100单位的青霉素用于细胞培养。
(2)Caco-2培养于细胞培养瓶。培养箱设置为37℃、5%CO2、保证相对湿度95%。细胞汇合度达到70‐90%时可用于接种Transwell。
(3)细胞接种前,向Transwell上室每孔中加入50μL细胞培养基,下层培养板内加入25mL细胞培养基。将培养板置于37℃、5%CO2培养箱内孵育1小时后可用于接种细胞。
(4)细胞消化后,吸取细胞混悬液转移至圆底离心管,于120g离心5分钟。
(5)使用培养基重悬细胞,终浓度为6.86×105个细胞/mL。将细胞悬液以50μL每孔加入到96孔Transwell培养板上室中,最终接种密度为2.4×105个细胞/cm2。
(6)接种后48小时开始换液,培养14-18天,隔一天换一次培养基。
(7)更换培养基过程如下,将Transwell小室与接收板分开,先弃掉接收板中培养基然后再弃掉Transwell小室培养基,最后每个小室加入75μL新鲜培养基,接收板加入25mL新鲜培养基。
3.2.2细胞单层膜完整性的评价
(1)Caco-2经过14-18天培养后,完全汇合并完成分化。此时,可应用于穿透试验。
(2)用电阻仪(Millipore,USA)测量单层膜电阻,记录每孔电阻。
(3)测定结束后,将Transwell培养板放回培养箱。
(4)电阻值的计算:测定电阻值(ohms)×膜面积(cm2)=TEER值(ohm·cm2),若TEER值<230 ohms·cm2,则该孔不能用于穿透试验。
3.2.3溶液配制
(1)配制1L缓冲液(HBSS,10mM HEPES,pH 7.4),分别称取2.38g HEPES,0.35g碳酸氢钠,加900mL纯水让其溶解,然后加100mL 10×HBSS搅拌均匀,调PH至7.4,最后过滤。
(2)配制受试物的DMSO储备液。配制对照药的1mM DMSO储备液。用缓冲液稀释得到5μM工作液。受试物用缓冲液稀释液得到5μM工作液。最终体系的DMSO含量为0.5%。
(3)制备给药端溶液:
(4)制备接收端溶液:
3.2.4药物穿透试验
(1)从培养箱中取出Caco-2Transwell培养板。使用缓冲液缓冲液润洗细胞单层膜两次,37℃条件下孵育30分钟。
(2)测定化合物由顶端到基底端的转运速率时,向***式培养板每孔(顶端)加入125μL测试化合物及对照药溶液,测试化合物和对照药溶液接收板(基底端)每孔加入235μL HBSS(10mM HEPES,pH 7.4)缓冲液。从顶端溶液中转移50μL样品加到200μL含内标的乙腈(100nM阿普***,200nM拉贝洛尔,200nM咖啡因和2μM酪洛芬)作为0分钟顶端给药样品进行检测。
(3)测定化合物由基底端到顶端的转运速率时,向接收板(基底端)每孔加入285μL测试化合物及对照药溶液,向测试化合物溶液***式培养板每孔(顶端)加入75μL HBSS(10mM HEPES,pH 7.4)缓冲液,向对照药溶液***式培养板每孔(顶端)加入75μL HBSS(10mM HEPES,pH 7.4)缓冲液。从基底端溶液中转移50μL样品加到200μL含内标的乙腈(100nM阿普***,200nM拉贝洛尔,200nM咖啡因和2μM酪洛芬)作为0分钟基底端给药样品进行检测。
(4)37℃CO2培养箱中孵育2小时。
(5)转运实验结束后,从加药端(Ap→Bl方向取顶端,Bl→Ap方向取基底端)转移50μL样品到加到200μL含内标的乙腈(100nM阿普***,200nM拉贝洛尔,200nM咖啡因和2μM酪洛芬)。从接收端(Ap→Bl方向取基底端,Bl→Ap方向取顶端)转移50μL样品到加到200μL含内标的乙腈(100nM阿普***,200nM拉贝洛尔,200nM咖啡因和2μM酪洛芬)。1000rpm涡旋10分钟。每个时间点的淬灭样品,3220g离心30分钟。转移每个样品的上清液100μL到一个96孔上样盘中,同时在相对应的孔中加入100μL纯水。样品分析盘1000rpm涡旋2分钟然后进行LC/MS/MS分析。所有孵育样品都做双平行。
(6)两个小时转运实验结束后测量荧光值,用水准备10mm荧光黄储备液,并且用转运缓冲溶液稀释至100μM。Transwell小室(顶端)加入100μL荧光黄溶液,基底端加入300μL转运缓冲溶液,37℃,CO2培养箱中孵育30分钟。从顶端与基底端直接取出80μL溶液(使用基底外侧孔)并且转移到新的96孔板中。用酶标仪测量细胞荧光值(检测膜完整性),激发波长为485nm,发射波长为530nm。
3.3数据分析
数据计算均使用Excel进行。峰面积由离子色谱结果计算得出。化合物的表观渗透系数(Papp,单位:cm/s×10-6)用以下公式计算得出:
公式中:VA为接收端溶液的体积(Ap→Bl是0.235mL,Bl→Ap是0.075mL),Area为Transwell-96孔板膜面积(0.143cm2);time为孵育时间(单位:s),[drug]acceptor为接收端药物浓度(相对值,用对应的Area Ratio参与计算),[drug]initial,donor为加药端初始加药浓度(相对值,用对应的Area Ratio参与计算)。
外排率使用以下的公式计算得出:
公式中:Papp(B-A)为由基底端到顶端的表观渗透系数;Papp(A-B)为由顶端到基底端的表观渗透系数。
表4化合物的外排比率
实验结论:相对于现有化合物,本发明化合物外排作用显著降低。
其中MRTX1719具有以下结构:
实验例4:化合物肝细胞代谢稳定性研究
4.1材料
肝细胞保存于液氮中,具体信息见下表。
4.2试验设计
4.2.1化合物工作液的制备
将受试物和对照药维拉帕米粉末用DSMO配置成高浓度储备液10mM,使用前用50%乙腈/水稀释到100μM的工作液,受试物和维拉帕米的终浓度为1μM。
4.2.2肝细胞的制备
(1)肝细胞复苏液包括Williams’Medium E,Isotonic Percoll,DPBS,GlutaMAX,HEPES,FBS,人重组胰岛素和***。混合49.5mL Williams’Medium E和0.5mL GlutaMAX作为孵育液。
(2)将肝细胞复苏液和孵育液于使用前置于37℃水浴中至少预热15分钟。取一管超低温保存的肝细胞,确保肝细胞在复苏之前仍处于低温冰冻状态。将肝细胞迅速置于37℃水浴中并轻摇直至所有冰晶全部分散,喷洒75%乙醇后转移至生物安全柜中。
(3)将肝细胞小管的内容物倾入盛有50mL复苏培养基的离心管中,将其于100g离心10分钟。 离心后,吸出复苏培养基并加入足量孵育培养基得到细胞密度约1.0×106个细胞/mL的细胞混悬液。
(4)用Cellometer Vision对肝细胞进行计数及确定活细胞密度,肝细胞成活率必须大于75%。利用孵育培养基稀释肝细胞混悬液至活细胞密度为0.5×106个细胞/mL。
(5)将一部分密度为0.5×106个细胞/mL的肝细胞混悬液开水煮沸灭活5分钟作为阴性对照。细胞灭活后,便于考察非细胞酶系介导的底物转化。
4.2.3试验方法
(1)转移198μL活细胞或者灭活细胞的混悬液到96孔深孔板,将深孔板置于涡旋上于孵箱中预热10分钟。活细胞进行双平行孵育,灭活细胞进行单平行孵育。
(2)每孔加入2μL 100μM受试物或维拉帕米进行反应起始,将深孔板放回孵箱涡旋器上。
(3)孵育样品,分别于0、15、30、60、90和120分钟,取25μL混悬液,加入150μL含内标的乙腈(200nM阿普***、200nM拉贝洛尔、2μM酮洛芬、200nM咖啡因)终止反应。涡旋10分钟,于3220g、4℃条件离心30分钟,离心完成后取100μL上清液和100μL超纯水混匀用于UPLC-MS/MS分析检测。
4.3数据分析
所有的数据计算均通过Microsoft Excel软件进行。通过提取离子图谱检测峰面积。通过对母药消除百分比的自然对数与时间进行线性拟合,检测母药的体外半衰期(t1/2)。
体外半衰期(t1/2)通过斜率计算:
in vitro t1/2=0.693/k
表5化合物的肝细胞代谢稳定性
实验结论:本发明化合物体外小鼠肝细胞代谢稳定。
实验例5:细胞抗增殖实验方法
5.1材料与细胞
HCT116MTAP Deletion细胞来源于Pharmaron(中国);HCT116细胞来源于ATCC(美国);McCoy's5A培养基购于Invitrogen公司(美国);青霉素-链霉素和胎牛血清购于Gibco公司(美国);384孔板购于PerkinElmer公司(美国);Cell-Titer Glo试剂购于Promega公司(美国)。
5.2化合物储存
化合物溶解:所有化合物使用二甲基亚砜(DMSO)溶解后储存。在室温干燥的条件下短期保存3个月或者-20℃长期保存。
5.3细胞培养及传代
细胞严格按照ATCC要求进行培养。将细胞培养基(McCoy's 5A培养基+10%胎牛血清+1%青霉素+链霉素)转移至15mL离心管中,1000rpm,离心5min。去除上清,用完全培养基重悬细胞,按所需密度接种于培养皿中,置于37℃,95%的潮湿空气,5%二氧化碳的培养箱中培养,视细胞生长情况每2-3天补一次培养液或进行传代。
5.4细胞增殖活性检测
细胞在培养皿中的汇合度达到90%左右时,用PBS清洗细胞两次后,使用胰蛋白酶将细胞处理成单细胞,进行细胞计数,每孔40μl接种于384孔板(50个细胞/孔),置于37℃,5%二氧化碳条件下平衡10-15min。利用Echo550将化合物打入384孔板中,设置day0_cell和阴性对照(0.1%DMSO),每个化合物10个梯度,3倍稀释,双复孔,DMSO含量0.1%。将384孔板置于细胞培养箱中培养10天。
后将40μl细胞增殖检测试剂(CTG)加入到384孔板中并震板混匀,后将384孔板置于37℃,5%二氧化碳的条件下避光孵育30分钟。通过多功能酶标仪Envision进行读数。化合物抑制率百分比的计算公式如下:抑制率百分比=100-(信号值药物处理组-信号值day0_cell)/(信号值阴性对照组-信号值day0_cell)×100。根据不同药物浓度及其所对应的抑制率,使用GraphPad 8.0软件进行GI50曲线绘制,分析数据,得出最终GI50值。结果见表6,其中GI50≤0.1μM为A,0.1μM<GI50≤1μM为B,1μM<GI50≤10μM为C。
表6本发明化合物细胞抗增殖活性的测试结果
另外,本发明化合物还具有针对突变型HCT116细胞的选择性,例如化合物71和72对HCT116MTAP Deletion的GI50为≤0.1μM,但对HCT116的GI50为1-10μM,选择性超过10倍。
实验例6细胞SDMA In-cell western实验
6.1材料与细胞
HCT116MTAP Deletion细胞来源于Horizon;HCT116细胞来源于ATCC;RPMI-1640培养基购于ATCC公司(美国);青霉素-链霉素购于Gibco公司(美国);胎牛血清购于ExCell公司;96孔板购于Corning公司(美国);Symmetric Di-Methyl Arginine Motif[sdme-RG]抗体购于CST(美国);IRDye 800CW Goat anti-Rabbit IgG二抗购于LI-COR(美国),CellTag 700stain购于LI-COR(美国)。
6.2化合物储存
化合物溶解:所有化合物使用二甲基亚砜(DMSO)溶解后储存。-20℃长期保存。
6.3细胞培养及传代
细胞严格按照ATCC要求进行培养。将细胞培养基(RPMI-1640培养基+10%血清+1%青霉素+链霉素)转移至15mL离心管中,1000rpm,离心5min。去除上清,用完全培养基重悬细胞,按所需密度接种于培养皿中,置于37℃,95%的潮湿空气,5%二氧化碳的培养箱中培养,视细胞生长情况每2-3天补一次培养液或进行传代。
6.4 SDMA In-cell western实验
细胞在培养皿中的汇合度达到90%左右时,用PBS清洗细胞两次后,用胰酶消化细胞,每孔100μL接种于96孔板(800/孔),在37℃,5%CO2培养箱中培养过夜。将50μL 3x终浓度的化合物加入细胞培养板中,每个化合物10个梯度,3倍稀释,双复孔,DMSO终浓度0.5%,在37℃,5%CO2培养箱中培养96小时。
每孔加入150μL 8%多聚甲醛,常温孵育15分钟。弃上清,PBS洗板,每孔加入200μL新制备的1x Permeabilization buffer,室温孵育30分钟。弃上清,每孔加入200μL 1x Blocking buffer,室温孵育2小时。弃上清,每孔加入50μL一抗(CST,1x Incubation buffer 1:500稀释),4℃过夜。弃上清,每孔加入250μL 1x Washing buffer,清洗三次。每孔加入50μL二抗(IRDye 800CW Goat anti-Rabbit IgG和CellTag 700染料,Incubation buffer 1:800和1:500稀释),室温孵育1小时。弃上清,1x Washing buffer洗板。使用Licor Odyssey CLX检测信号。化合物抑制率百分比的计算公式如下:抑制率百分比=100-(信号值药物处理组-信号值背景值)/(信号值0.5%DMSO处理组-信号值背景值)×100。根据不同药物浓度及其所对应的抑制率,使用GraphPad 8.0软件进行IC50曲线绘制,分析数据,得出最终IC50值。结果见表7,其中IC50≤0.1μM为A,0.1μM<IC50≤1μM为B,1μM<IC50≤10μM为C。
表7细胞SDMA抑制活性
实验例7 LU99 CDX模型体内药效学研究
7.1实验材料:LU99细胞来源于JCRB(日本);RPMI-1640和0.25%胰酶-EDTA购自Gibco(美国);胎牛血清购自东岭(中国);PBS购自Hyclone(美国);青霉素-链霉素购于美仑生物(中国);Matrigel基质胶购于Corning(美国)。
7.2动物信息:BALB/c Nude小鼠,雌性,6-8周龄,体重18-22克,动物购自北京维通利华实验动物技术有限公司,将小鼠饲养在SPF级环境中,每个笼位单独送排风,所有动物都可以自由获取标准认证的商业实验室饮食和自由饮水。
7.3实验方法:
7.3.1细胞培养:人大细胞肺癌细胞LU99体外单层培养,培养条件为RPMI-1640培养基中加10%胎牛血清,37℃5%CO2孵箱培养。一周两次用胰酶-EDTA进行常规消化处理传代。当细胞数量到达要求时,收取细胞,计数。
7.3.2细胞接种:将0.2mL(5×106个)LU99细胞(加基质胶,体积比为1:1)细胞皮下接种于每只小鼠的右后背,肿瘤平均体积达到约150mm3时,依据肿瘤体积和动物体重采用随机分层分组方法开始分组给药。
7.3.3给药:化合物72的给药剂量为30mpk,PO,每天一次给药(QD)×3周。每组6只小鼠。
7.3.4肿瘤测量和实验指标:
每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:V=0.5a×b2,a和b分别表示肿瘤的长径和短径。每周两次测量小鼠体重。
化合物的抑瘤疗效用肿瘤生长抑制率TGI(%)来评价。
TGI(%)=[(1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积))/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)]×100%。
实验结果见图1和图2。实验过程中无小鼠发病或死亡。

Claims (50)

  1. 式(I)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物:
    其中:
    Z1、Z2和Z3独立地选自N、CD或CH,并且其中至少一个为N;
    环A为苯基或5-6元杂芳基;
    R1选自H、D、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
    R2选自H、D、-C0-6亚烷基-卤素、-C0-6亚烷基-CN、-C0-6亚烷基-ORa、-C0-6亚烷基-NRbRc、C1-6烷基、C1-6卤代烷基、C3-8环烷基或4-7元杂环基;
    m=0、1、2或3;
    其中R选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O、C=S、C3-8环烷基或4-7元杂环基;或者相邻原子上的两个R与所述原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3或4个R’取代基取代;
    R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,优选H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1个或多个D、卤素、C1-6烷基或C1-6卤代烷基取代;
    Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基;
    其中环A、R1、R2、R、R’、Ra、Rb和Rc中的各基团可任选地被D取代,直至完全氘代。
  2. 权利要求1的式(I)化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物:
    其中:
    Z1、Z2和Z3独立地选自N、CD或CH,并且其中至少一个为N;
    环A为苯基或5-6元杂芳基;
    R1选自H、D、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
    R2选自H、D、-C0-6亚烷基-卤素、-C0-6亚烷基-CN、-C0-6亚烷基-ORa、-C0-6亚烷基-NRbRc、C1-6烷基、C1-6卤代烷基、C3-8环烷基或4-7元杂环基;
    m=0、1、2或3;
    其中R选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O、C=S、C3-8环烷基或4-7元杂环基;或者相邻原子上的两个R与所述原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3或4个R’取代基取代;
    R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基,优选H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基或4-7元杂环基;
    Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基;
    其中环A、R1、R2、R、R’、Ra、Rb和Rc中的各基团可任选地被D取代,直至完全氘代。
  3. 权利要求1或2的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中环A为5-6元杂芳基;
    优选选自以下:
    更优选选自以下:
    更优选为
    其中R1和R2如权利要求2所定义。
  4. 权利要求1-3中任一项的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中R2选自H、卤素、CN、-C0-6亚烷基-ORa、-C0-6亚烷基-NRbRc、C1-6烷基、C1-6卤代烷基或C3-8环烷基;优选选自H、-C0-6亚烷基-ORa、C1-6烷基、C1-6卤代烷基或C3-6环烷基;更优选选自H、F、Cl、Br、CH2CH2OH、Me、Et、Pr、iPr、CF3或环丙基;更优选为C1-6烷基或C1-6卤代烷基,尤其Me。
  5. 权利要求1-4中任一项的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异 构体、前药、多晶型、水合物或溶剂合物,其中R1选自C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基;优选选自4-7元杂环基、C6-10芳基或5-6元杂芳基;更优选选自苯基、吡咯烷基、吡唑烷基、哌啶基或吡啶酮基。
  6. 权利要求1-5中任一项的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中R选自卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O、C=S或C3-6环烷基;或者相邻原子上的两个R与所述原子形成C3-6环烷基、5-6元杂环基、苯基或5-6元杂芳基,例如环戊烯基、苯基、噻吩基或吡啶基。
  7. 权利要求1-6中任一项的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;优选地,R’选自H、D、卤素、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;优选地,R’选自H、D、卤素、CN、C1-6烷基、C1-6卤代烷基、C6-10芳基或5-10元杂芳基;优选地,R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-6环烷基或5-6元杂环基。
  8. 权利要求1-7中任一项的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:

    其中,
    表示单键或双键;
    X选自NRN、CR2bR2b’、CR2bR2b’-CR2bR2b’或CR2b=CR2b’;
    Y1选自N或CRY1
    Y2选自N或CRY2
    Y3选自N或CRY3
    Y4选自N或CRY4
    其中RN选自H、C1-6烷基或C1-6卤代烷基;
    R1a、R2a、R3a、R4a、R5a、R1b、R1b’、R2b、R2b’、R3b和R4b具有权利要求1或2中与R相同的定义;
    RY1、RY2、RY3和RY4具有上文中与R’相同的定义;
    其他基团如权利要求1-7中任一项所定义。
  9. 权利要求8的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
    其中,
    R2选自H、C1-6烷基、C1-6卤代烷基、C3-8环烷基或4-7元杂环基;
    R1a选自H、D、卤素、CN、ORa或NRbRc
    R2a选自H、D、卤素或CN;
    R3a选自H或D;
    R4a选自ORa或NRbRc
    R5a选自H、D或CN;
    或者,R3a、R4a以及它们连接的碳原子形成4-6元杂环基或5-6元杂芳基,其任选地被1、2、3或4个R’取代基取代;
    其中R’选自H、D、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基或4-7元杂环基;
    Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基。
  10. 权利要求9的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    R2选自H、C1-6烷基或C1-6卤代烷基;
    R1a为卤素,优选F;
    R2a选自H、D或卤素;
    R3a为H或D;
    R4a为ORa
    R5a为CN;
    或者,R3a、R4a以及它们连接的碳原子形成呋喃基或二氢呋喃基,其任选地被1、2、3或4个R’取代基取代;
    其中R’选自H、D、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-6环烷基或5-6元杂环基;
    Ra选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-7环烷基或4-7元杂环基。
  11. 权利要求9的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    R2选自H、C1-4烷基或C1-4卤代烷基;
    R1a为卤素,优选F;
    R2a选自H、D或卤素;
    R3a为H或D;
    R4a为ORa
    R5a为CN;
    或者,R3a、R4a以及它们连接的碳原子形成二氢呋喃基,其被1、2、3或4个R’取代基取代;
    其中R’选自H、D、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-6环烷基或5-6元杂环基,优选环丙基;
    Ra选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-6环烷基或5-6元杂环基。
  12. 权利要求9的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    R2为Me;
    R1a为F;
    R2a为H或Cl;
    R3a、R4a以及它们连接的碳原子形成二氢呋喃基,其被1、2、3或4个R’取代基取代;
    R5a为CN;
    其中R’选自H或Me,或者相同或不同原子上的两个R’与所述原子形成环丙基。
  13. 权利要求8的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
    其中,
    表示单键或双键;
    X选自NRN、CR2bR2b’、CR2bR2b’-CR2bR2b’或CR2b=CR2b’;
    R2选自H、C1-6烷基、C1-6卤代烷基、C3-8环烷基或4-7元杂环基;
    R1b和R1b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
    R3b和R4b独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者R3b、R4b以及它们连接的碳原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3或4个R’取代基取代;
    其中RN选自H、C1-6烷基或C1-6卤代烷基;
    R2b和R2b’独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的R2b和R2b’以及它们连接的碳原子形成C3-6环烷基或4-7元杂环基;或者R2b’和R3b以及它们连接的碳原子形成C6-10芳基或5-10元杂芳基;
    R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基或4-7元杂环基;
    Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基。
  14. 权利要求13的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    表示单键或双键;
    X选自NRN、CR2bR2b’或CR2b=CR2b’;
    R2选自H、C1-6烷基或C1-6卤代烷基;
    R1b和R1b’独立地选自H、D、C1-4烷基或C1-4卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
    R3b和R4b独立地选自H、D、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者R3b、R4b以及它们连接的碳原子形成C3-6环烷基、4-7元杂环基、苯基或5-6元杂芳基,其任选地被1、2、3或4个R’取代基取代;
    其中RN选自H、C1-6烷基或C1-6卤代烷基;
    R2b和R2b’独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的R2b和R2b’以及它们连接的碳原子形成C3-6环烷基;或者R2b’和R3b以及它们连接的碳原子形成苯基或5-6元杂芳基;
    R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;
    Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基。
  15. 权利要求8的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
    其中,
    X选自NRN或CR2bR2b’;
    R2选自H、C1-6烷基或C1-6卤代烷基;
    R1b和R1b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
    Y1选自N或CRY1
    Y2选自N或CRY2
    Y3选自N或CRY3
    Y4选自N或CRY4
    其中RN选自H、C1-6烷基或C1-6卤代烷基;
    R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;或者R2b和R2b’以及它们连接的碳原子形成C3-6环烷基;
    RY1、RY2、RY3和RY4独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;
    Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基。
  16. 权利要求15的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    X选自NRN或CR2bR2b’;
    R2选自H、C1-4烷基或C1-4卤代烷基;
    R1b和R1b’独立地选自H、D、C1-4烷基或C1-4卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O;
    Y1选自N或CRY1
    Y2选自N或CRY2
    Y3选自N或CRY3
    Y4选自N或CRY4
    其中RN选自H、C1-4烷基或C1-4卤代烷基;
    R2b和R2b’独立地选自H、D、C1-4烷基或C1-4卤代烷基;或者R2b和R2b’以及它们连接的碳原子形成C3-6环烷基;
    RY1、RY2、RY3和RY4独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;
    Ra、Rb和Rc独立地选自H、C1-6烷基或C1-6卤代烷基,或者Rb和Rc以及它们连接的氮原子形成5-6元杂环基。
  17. 权利要求15的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    X选自NRN或CR2bR2b’;
    R2为Me;
    R1b和R1b’独立地选自H、D、C1-4烷基或C1-4卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O;
    Y1选自N或CRY1
    Y2选自N或CRY2
    Y3选自N或CRY3
    Y4选自N或CRY4
    其中RN选自H或Me;
    R2b和R2b’独立地选自H或Me,或者R2b和R2b’以及它们连接的碳原子形成环丙基;
    RY1、RY2、RY3和RY4选自H、F、CN、Me、CF3或OMe。
  18. 权利要求8的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
    其中,
    表示单键或双键;
    R2选自H、C1-6烷基或C1-6卤代烷基;
    R1b和R1b’独立地选自H、C1-6烷基或C1-6卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成 C=O或C=S;
    R2b和R2b’独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;
    R3b选自H、D、卤素或CN;
    R4b选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;
    或者R2b’和R3b以及它们连接的碳原子形成苯基或5-6元杂芳基;
    或者R3b、R4b以及它们连接的碳原子形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1、2、3或4个R’取代基取代;
    R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;或者相同或不同原子上的两个R’与所述原子形成C3-8环烷基或4-7元杂环基;
    Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基。
  19. 权利要求18的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    表示单键或双键;
    R2选自H、C1-4烷基或C1-4卤代烷基;
    R1b和R1b’独立地选自H、D、C1-4烷基或C1-4卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O;
    R2b和R2b’独立地选自H、D、卤素、CN、ORa、NRbRc、C1-4烷基或C1-4卤代烷基;
    R3b选自H或D;
    R4b选自H、D、ORa、NRbRc、C1-4烷基或C1-4卤代烷基;
    或者R2b’和R3b以及它们连接的碳原子形成苯基;
    或者R3b、R4b以及它们连接的碳原子形成C3-6环烷基、5-6元杂环基、苯基或5-6元杂芳基,其任选地被1、2、3或4个R’取代基取代;
    R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基或C1-6卤代烷基;
    Ra、Rb和Rc独立地选自H、C1-6烷基或C1-6卤代烷基。
  20. 权利要求18的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    表示单键或双键;
    R2为Me;
    R1b和R1b’独立地选自H、D、C1-4烷基或C1-4卤代烷基;或者R1b、R1b’以及它们连接的碳原子形成C=O;
    R2b选自H、D、F、Cl、Br、I、Me或CF3
    R2b’选自H、D、F、Cl、Br、I、Me、CF3或OMe;
    R3b为H或D;
    R4b选自H、D、Me、OMe或O-环丙基;
    或者,R3b、R4b以及它们连接的碳原子形成苯基、环戊烯基或噻吩基;
    或者,R2b’、R3b以及它们连接的碳原子形成苯基。
  21. 权利要求8的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
    其中:
    R1和R2如权利要求1和3-6中任一项所定义;
    优选地,
    R1为4-7元杂环基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
    R2选自C1-6烷基或C1-6卤代烷基;
    其中R选自H、D、卤素、CN、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O或C=S;或者相邻原子上的两个R与所述原子形成苯基或5-6元杂芳基,其任选地被1、2、3或4个R’取代基取代;
    R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;
    其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基。
  22. 权利要求21的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    R1为5-6元杂环基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
    R2选自C1-6烷基或C1-6卤代烷基;
    其中R选自H、D、卤素、CN、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O或C=S;或者相邻原子上的两个R与所述原子形成苯基,其任选地被1、2、3或4个R’取代基取代;
    R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C3-6环烷基、5-6元杂环基、苯基或5-6元杂芳基;
    其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基。
  23. 权利要求21的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    R1选自吡咯烷基或哌啶基,其任选地被1、2、3、4、5、6、7、8、9或10个R取代基取代;
    R2选自C1-6烷基或C1-6卤代烷基;
    其中R选自H、D、卤素、CN、C1-6烷基或C1-6卤代烷基;或者相同碳原子上的两个R与该碳原子形成C=O;或者相邻原子上的两个R与所述原子形成苯基,其任选地被1、2、3或4个R’取代基取代;
    R’选自H、D、卤素、CN、C1-6烷基、C1-6卤代烷基、苯基或5-6元杂芳基。
  24. 权利要求8的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
    其中:
    表示单键或双键;
    X选自CR2bR2b’、CR2bR2b’-CR2bR2b’或CR2b=CR2b’;
    R2选自C1-6烷基或C1-6卤代烷基;
    R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
    R3b、R4b以及它们连接的碳原子形成C6-10芳基或5-10元杂芳基,其任选地被1、2、3或4个R’取代基取代;
    R2b和R2b’独立地选自H、D、卤素、CN、C1-6烷基或C1-6卤代烷基;
    R’选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;
    其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基。
  25. 权利要求24的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    表示双键;
    X选自CR2bR2b’、CR2bR2b’-CR2bR2b’或CR2b=CR2b’;
    R2选自C1-6烷基或C1-6卤代烷基;
    R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
    R3b、R4b以及它们连接的碳原子形成苯基,其任选地被1、2、3或4个R’取代基取代;
    R2b和R2b’独立地选自H、D、卤素、CN、C1-6烷基或C1-6卤代烷基;
    R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C3-6环烷基、5-6元杂环基、苯基或5-6元杂芳基;
    其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基或C2-6炔基。
  26. 权利要求24的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    表示双键;
    X选自CR2bR2b’或CR2bR2b’-CR2bR2b’;
    R2选自C1-6烷基或C1-6卤代烷基;
    R1b、R1b’以及它们连接的碳原子形成C=O;
    R3b、R4b以及它们连接的碳原子形成苯基,其任选地被1、2、3或4个R’取代基取代;
    R2b和R2b’独立地选自H或D;
    R’选自H、D、卤素、CN、C1-6烷基、C1-6卤代烷基、苯基或5-6元杂芳基。
  27. 权利要求8的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前 药、多晶型、水合物或溶剂合物,其具有以下通式:
    其中:
    X为CR2bR2b’;
    R2选自C1-6烷基或C1-6卤代烷基;
    R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
    Y1选自N或CRY1
    Y2选自N或CRY2
    Y3选自N或CRY3
    Y4选自N或CRY4
    R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
    RY1、RY2、RY3和RY4独立地选自H、D、卤素、CN、ORa、NRbRc、C(O)ORa、C(O)NRbRc、S(O)Ra、S(O)2Ra、C1-6烷基、C1-6卤代烷基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;
    其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基。
  28. 权利要求27的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    X为CR2bR2b’;
    R2选自C1-6烷基或C1-6卤代烷基;
    R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
    Y1选自N或CRY1
    Y2选自N或CRY2
    Y3选自N或CRY3
    Y4选自N或CRY4
    R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
    RY1、RY2、RY3和RY4独立地选自R’选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C3-6环烷基、5-6元杂环基、苯基或5-6元杂芳基;
    其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基。
  29. 权利要求27的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    X为CR2bR2b’;
    R2选自C1-6烷基或C1-6卤代烷基;
    R1b、R1b’以及它们连接的碳原子形成C=O;
    Y1为CRY1
    Y2为CRY2
    Y3为CRY3
    Y4为CRY4
    R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
    RY1、RY2、RY3和RY4独立地选自H、D、卤素、CN、C1-6烷基、C1-6卤代烷基、苯基或5-6元杂芳基。
  30. 权利要求8的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
    其中:
    X为CR2bR2b’;
    R2选自C1-6烷基、C1-6卤代烷基、C3-6环烷基或4-7元杂环基;
    R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
    Y1为CRY1
    Y2为CRY2
    Y3为CRY3
    Y4为CRY4
    R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
    RY1、RY2、RY3和RY4独立地选自H、D、卤素、CN、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C2- 6烯基、C2-6炔基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;
    或者RY1和RY2、RY2和RY3、RY3和RY4,以及他们连接的原子一起形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1个或多个D、卤素、C1-6烷基或C1-6卤代烷基取代;
    其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基。
  31. 权利要求30的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    X为CR2bR2b’;
    R2为Me、Et或环丙基;
    R1b、R1b’以及它们连接的碳原子形成C=O;
    Y1为CRY1
    Y2为CRY2
    Y3为CRY3
    Y4为CRY4
    R2b和R2b’独立地选自H或D;
    RY1选自H、D、Cl、F、CN、Me、CF3或OMe;
    RY2选自H、D、Me、Et、iPr、CF3、环丙基、OMe、OEt、OiPr、OCF3、O-环丙基、丙-1-烯-2-基或苯基;
    RY3选自H、D、Cl、F、Me、Et、iPr、CF3、环丙基、丙-1-烯-2-基、OMe或O-环丙基;
    RY4选自H、D或Me;
    或者RY1和RY2、RY2和RY3、RY3和RY4,以及他们连接的原子一起形成C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,其任选地被1个或多个D、卤素、C1-6烷基或C1-6卤代烷基取代。
  32. 权利要求30的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    X为CR2bR2b’;
    R2选自C1-6烷基、C1-6卤代烷基、C3-6环烷基或4-7元杂环基;
    R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
    Y1为CRY1
    Y2为CRY2
    Y3为CRY3
    Y4为CRY4
    R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
    RY1、RY2、RY3和RY4独立地选自H、D、卤素、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-10环烷基、4-10元杂环基、C6-10芳基或5-10元杂芳基;
    其中RY1和RY3中至少一个不为H或D;
    其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基。
  33. 权利要求32的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    X为CR2bR2b’;
    R2选自C1-6烷基、C1-6卤代烷基或C3-6环烷基;
    R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
    Y1为CRY1
    Y2为CRY2
    Y3为CRY3
    Y4为CRY4
    R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
    RY1和RY3独立地选自H、D、卤素、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-6环烷基或4-7元杂环基;
    RY2和RY4独立地选自H、D、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C2-6烯基或C2-6炔基;
    其中RY1和RY3中至少一个不为H或D;
    其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基。
  34. 权利要求32的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前 药、多晶型、水合物或溶剂合物,其中,
    X为CR2bR2b’;
    R2为Me、Et或环丙基;
    R1b、R1b’以及它们连接的碳原子形成C=O;
    Y1为CRY1
    Y2为CRY2
    Y3为CRY3
    Y4为CRY4
    R2b和R2b’独立地选自H或D;
    RY1选自H、D、Cl、F、Me、CF3或OMe;
    RY2选自H、D、Et、iPr、OiPr、O-环丙基或丙-1-烯-2-基;
    RY3选自H、D、Cl、F、Me、Et、iPr、CF3、环丙基、丙-1-烯-2-基、OMe或O-环丙基;
    RY4选自H或D;
    其中RY1和RY3中至少一个不为H或D。
  35. 权利要求32的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    X为CR2bR2b’;
    R2选自C1-6烷基或C1-6卤代烷基;
    R1b、R1b’以及它们连接的碳原子形成C=O或C=S;
    Y1为CRY1
    Y2为CRY2
    Y3为CRY3
    Y4为CRY4
    R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
    RY1和RY3独立地选自H、D、卤素、ORa、NRbRc、C1-6烷基、C1-6卤代烷基、C3-6环烷基或4-7元杂环基;
    RY2和RY4独立地选自H、D、C1-6烷基或C1-6卤代烷基;
    其中RY1和RY3中至少一个不为H或D;
    其中Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基或4-7元杂环基。
  36. 权利要求35的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    X为CR2bR2b’;
    R2选自C1-6烷基或C1-6卤代烷基;
    R1b、R1b’以及它们连接的碳原子形成C=O;
    Y1为CRY1
    Y2为CRY2
    Y3为CRY3
    Y4为CRY4
    R2b和R2b’独立地选自H、D、C1-6烷基或C1-6卤代烷基;
    RY1选自H、D、卤素、C1-6烷基或C1-6卤代烷基;
    RY2选自H、D、C1-6烷基或C1-6卤代烷基;
    RY3选自H、D、卤素、ORa、C1-6烷基、C1-6卤代烷基或C3-6环烷基;
    RY4选自H或D;
    其中RY1和RY3中至少一个不为H或D;
    其中Ra选自H、C1-6烷基或C1-6卤代烷基。
  37. 权利要求35的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    X为CR2bR2b’;
    R2为Me;
    R1b、R1b’以及它们连接的碳原子形成C=O;
    Y1为CRY1
    Y2为CRY2
    Y3为CRY3
    Y4为CRY4
    R2b和R2b’独立地选自H或D;
    RY1选自H、D、Cl、F、Me或CF3
    RY2选自H、D或Et;
    RY3选自H、D、Cl、F、Me、Et、OMe或环丙基;
    RY4选自H或D;
    其中RY1和RY3中至少一个不为H或D。
  38. 权利要求8的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其具有以下通式:
    其中,
    R2选自C1-6烷基或C1-6卤代烷基;
    R1a选自H、D、卤素、CN、ORa或NRbRc
    R2a选自H、D、卤素或CN;
    R3a选自H或D;
    R4a选自ORa或NRbRc
    R5a选自H、D或CN;
    或者,R3a、R4a以及它们连接的碳原子形成4-6元杂环基或5-6元杂芳基,其任选地被1、2、3或4个R’取代基取代;
    其中R’选自H、D、C1-6烷基或C1-6卤代烷基;
    Ra、Rb和Rc独立地选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-8环烷基、4-7元杂环基、C6-10芳基或5-10元杂芳基,或者Rb和Rc以及它们连接的氮原子形成4-7元杂环基。
  39. 权利要求38的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    R2选自C1-6烷基或C1-6卤代烷基;
    R1a为卤素,优选F;
    R2a选自H、D或卤素;
    R3a为H或D;
    R4a为ORa
    R5a为CN;
    或者,R3a、R4a以及它们连接的碳原子形成呋喃基或二氢呋喃基,其任选地被1、2、3或4个R’取代基取代;
    其中R’选自H、D、C1-6烷基或C1-6卤代烷基;
    Ra选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-7环烷基或4-7元杂环基。
  40. 权利要求38的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中,
    R2选自C1-4烷基或C1-4卤代烷基;
    R1a为卤素,优选F;
    R2a选自H、D或卤素;
    R3a为H或D;
    R4a为ORa
    R5a为CN;
    其中Ra选自H、C1-6烷基、C1-6卤代烷基、C2-6烯基、C2-6炔基、C3-6环烷基或5-6元杂环基。
  41. 化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,其中所述化合物选自表1或表2。
  42. 药物组合物,其包含权利要求1-41中任一项的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,和药学上可接受的载体、佐剂或媒介物,任选地其它治疗剂。
  43. 权利要求42的药物组合物,其中所述其它治疗剂选自以下靶点药物/细胞活性调节剂,包括CDK4/6抑制剂,MAT2A抑制剂,MAPK1/MAPK3抑制剂,Type I PRMT抑制剂,EGFR抑制剂,SHP2抑制剂,泛KRAS抑制剂,KRASG12C抑制剂,RAF抑制剂,MEK抑制剂,ERK抑制剂,Bcl-2抑制剂,SOS1抑制剂,PARP抑制剂,MALT1抑制剂,MALT2抑制剂,BTK抑制剂,PI3K抑制剂,AKT抑制剂,FGFR抑制剂,DNA甲基转移酶(DNMT)抑制剂,EZH1/2抑制剂,EZH2抑制剂,Menin-MLL抑制剂,IDH1抑制剂,IDH2抑制剂,IDH1/2抑制剂,化疗药物,放射疗法,STING激动剂或免疫检查点抑制剂/调节剂。
  44. 权利要求1-41中任一项的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物在制备用于治疗或预防PRMT5介导的疾病的药物中的用途。
  45. 一种在受试者中治疗或预防PRMT5介导的疾病的方法,包括向所述受试者给药权利要求1-41中任一项的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶 型、水合物或溶剂合物,或权利要求42或43的药物组合物。
  46. 权利要求1-41中任一项的化合物,或其药学上可接受的盐、同位素变体、互变异构体、立体异构体、前药、多晶型、水合物或溶剂合物,或权利要求42或43的药物组合物,其用于治疗或预防PRMT5介导的疾病。
  47. 权利要求44的用途或权利要求45的方法或权利要求46的化合物或药物组合物的用途,其中所述疾病为癌症。
  48. 权利要求44的用途或权利要求45的方法或权利要求46的化合物或药物组合物的用途,其中所述PRMT5介导的疾病选自以下癌症:心脏:肉瘤(血管肉瘤、纤维肉瘤、横纹肌肉瘤、脂肪肉瘤)、粘液瘤、横纹肌瘤、纤维瘤、脂肪瘤和畸胎瘤;肺:支气管癌(鳞状细胞、未分化小细胞、未分化大细胞、腺癌)、肺泡(细支气管)癌、支气管腺瘤、肉瘤、淋巴瘤、软骨瘤错构瘤、间皮瘤;胃肠道:食管(鳞状细胞癌、腺癌、平滑肌肉瘤、淋巴瘤)、胃(癌、淋巴瘤、平滑肌肉瘤)、胰腺(导管腺癌、胰岛素瘤、胰高血糖素瘤、胃泌素瘤、类癌瘤、血管瘤)、小肠(腺癌、淋巴瘤、类癌瘤)、卡波西肉瘤、平滑肌瘤、血管瘤、脂肪瘤、神经纤维瘤、纤维瘤)、大肠(腺癌、管状腺瘤、绒毛状腺瘤、错构瘤、平滑肌瘤);泌尿生殖道:肾脏(腺癌、威尔姆氏瘤(肾母细胞瘤)、淋巴瘤、白血病)、膀胱和尿道(鳞状细胞癌、移行细胞癌、腺癌)、***(腺癌、肉瘤)、睾丸(***瘤、畸胎瘤、胚胎癌、畸胎癌)、绒毛膜癌、肉瘤、***癌、纤维瘤、纤维腺瘤、腺瘤样肿瘤、脂肪瘤);肝脏:肝细胞瘤(肝细胞癌)、胆管癌、肝母细胞瘤、血管肉瘤、肝细胞腺瘤、血管瘤;胆道:胆囊癌、壶腹癌、胆管癌;骨:成骨肉瘤(osteosarcoma)、纤维肉瘤、恶性纤维组织细胞瘤、软骨肉瘤、尤文氏肉瘤、恶性淋巴瘤(网状细胞肉瘤)、多发性骨髓瘤、恶性巨细胞瘤脊索瘤、骨慢性外生骨疣(osteocartilaginous exostoses)、良性软骨瘤、软骨母细胞瘤、纤维软骨瘤、类骨质骨瘤和巨细胞瘤;神经***:颅骨(骨瘤、血管瘤、肉芽肿、黄色瘤、变形性骨炎)、脑膜(脑膜瘤、脑膜肉瘤、神经胶质瘤)、脑(星形细胞瘤、髓母细胞瘤、神经胶质瘤、室管膜瘤、生殖细胞瘤(松果体瘤)、多形性胶质母细胞瘤、少突胶质细胞瘤、神经鞘瘤、视网膜母细胞瘤、先天性肿瘤)、脊髓神经纤维瘤、脑膜瘤、神经胶质瘤、肉瘤);妇科:子宫(子宫内膜癌(浆液性囊腺癌、粘液性囊腺癌、未分类癌)、颗粒鞘细胞瘤、支持***瘤、无性细胞瘤、恶性畸胎瘤)、外阴(鳞状细胞癌、上皮内癌、腺癌、纤维肉瘤、黑色素瘤)、***(透明细胞癌、鳞状细胞癌、葡萄样肉瘤(胚胎性横纹肌肉瘤)、输卵管(癌);血液学:血液(髓性白血病(急性和慢性)、急性淋巴细胞白血病、慢性淋巴细胞性白血病、骨髓增生性疾病、多发性骨髓瘤、骨髓增生异常综合征)、霍奇金病、非霍奇金淋巴瘤(恶性淋巴瘤);皮肤:恶性黑色素瘤、基底细胞癌、鳞状细胞癌、卡波西肉瘤、痣发育不良痣、脂肪瘤、血管瘤、皮肤纤维瘤、瘢痕疙瘩、牛皮癣;和肾上腺:成神经细胞瘤。
  49. 权利要求44的用途或权利要求45的方法或权利要求46的化合物或药物组合物的用途,其中所述PRMT5介导的疾病选自以下癌症:恶性周围神经鞘瘤、间皮瘤、多形性胶质母细胞瘤、胰腺癌、胆管癌、***癌、乳腺癌、脑癌、皮肤癌、***、膀胱癌、星形细胞瘤、结肠直肠癌、子宫内膜癌、食道癌、胃癌、胸腺瘤、头颈癌、肝细胞癌、喉癌、肺鳞癌、肺腺癌、口腔癌、卵巢癌、肾癌和甲状腺癌以及肉瘤。
  50. 权利要求44的用途或权利要求45的方法或权利要求46的化合物或药物组合物的用途,其中所述PRMT5介导的疾病选自MTAP相关癌症,例如肝细胞癌、乳腺癌、皮肤癌、膀胱癌、肝癌、胰腺癌或头颈癌。
PCT/CN2023/089249 2022-04-22 2023-04-19 稠和哒嗪酮化合物作为prmt5抑制剂 WO2023202626A1 (zh)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202210430110 2022-04-22
CN202210430110.6 2022-04-22
CN202211438072 2022-11-16
CN202211438072.5 2022-11-16

Publications (1)

Publication Number Publication Date
WO2023202626A1 true WO2023202626A1 (zh) 2023-10-26

Family

ID=88419277

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/089249 WO2023202626A1 (zh) 2022-04-22 2023-04-19 稠和哒嗪酮化合物作为prmt5抑制剂

Country Status (1)

Country Link
WO (1) WO2023202626A1 (zh)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021050915A1 (en) * 2019-09-12 2021-03-18 Mirati Therapeutics, Inc. Mta-cooperative prmt5 inhibitors
WO2022192745A1 (en) * 2021-03-11 2022-09-15 Mirati Therapeutics, Inc. Mta-cooperative prmt5 inhibitors
WO2022216648A1 (en) * 2021-04-08 2022-10-13 Mirati Therapeutics, Inc. Combination therapies using prmt5 inhibitors for the treatment of cancer
WO2022256806A1 (en) * 2021-06-02 2022-12-08 Ideaya Biosciences, Inc. Combination therapy comprising a mat2a inhibitor and a type ii prmt inhibitor
WO2023059798A1 (en) * 2021-10-06 2023-04-13 Mirati Therapeutics, Inc. Methods for separation of enantiomers
WO2023059795A1 (en) * 2021-10-06 2023-04-13 Mirati Therapeutics, Inc. Combination therapies using prmt5 inhibitors for the treatment of cancer

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021050915A1 (en) * 2019-09-12 2021-03-18 Mirati Therapeutics, Inc. Mta-cooperative prmt5 inhibitors
WO2022192745A1 (en) * 2021-03-11 2022-09-15 Mirati Therapeutics, Inc. Mta-cooperative prmt5 inhibitors
WO2022216648A1 (en) * 2021-04-08 2022-10-13 Mirati Therapeutics, Inc. Combination therapies using prmt5 inhibitors for the treatment of cancer
WO2022256806A1 (en) * 2021-06-02 2022-12-08 Ideaya Biosciences, Inc. Combination therapy comprising a mat2a inhibitor and a type ii prmt inhibitor
WO2023059798A1 (en) * 2021-10-06 2023-04-13 Mirati Therapeutics, Inc. Methods for separation of enantiomers
WO2023059795A1 (en) * 2021-10-06 2023-04-13 Mirati Therapeutics, Inc. Combination therapies using prmt5 inhibitors for the treatment of cancer

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SMITH CHRISTOPHER R ET AL.: "Design and Evaluation of Achiral, Non-atropisomeric 4- (aminomethyl) Phthalazin-1(2H)-one Derivatives as Novel PRMTS/MTA Inhibitors", BIOORGANIC MEDICINAL CHEMISTRY, vol. 71, 26 July 2022 (2022-07-26), pages 116947, XP087167033, DOI: 10.1016/j.bmc.2022.116947 *

Similar Documents

Publication Publication Date Title
TWI765908B (zh) 苯並咪唑類化合物激酶抑制劑及其製備方法和應用
CN111107848B (zh) 一种含有酰胺类衍生物的药物组合物及其制备方法和应用
WO2021057877A1 (zh) 取代的芳香稠合环衍生物及其组合物及用途
WO2020034988A1 (zh) 凋亡信号调节激酶1抑制剂的盐及其晶型
BRPI0718803B1 (pt) composto para inibir a progressão mitótica, composição farmacêutica e método in vitro para inibir a atividade de aurora cinase em uma célula
JP7092405B2 (ja) キナーゼ活性を阻害するためのジ(ヘテロ)アリール大環状化合物
CN109627263B (zh) 用于抑制激酶活性的二苯氨基嘧啶类化合物
TW202208375A (zh) 4-胺基-5-(6-(4-甲基哌-1-基)-1h-苯并[d]咪唑-2-基)噻吩并[2,3-b]吡啶-6(7h)-酮之鹽及晶形
WO2020020385A1 (zh) 含三并环类衍生物抑制剂、其制备方法和应用
CN115244053B (zh) 靶向蛋白降解化合物及其制备方法和应用
WO2023202626A1 (zh) 稠和哒嗪酮化合物作为prmt5抑制剂
CN116462685A (zh) 杂环化合物、包含其的药物组合物及其抗肿瘤应用
UA121315C2 (uk) Макроциклічні інгібітори кінази rip2
EP3715350A1 (en) Arylphosphine oxides for inhibiting kinase activity
WO2024037607A1 (zh) Prmt5抑制剂
WO2021057796A1 (zh) 取代的稠合三环衍生物及其组合物及用途
WO2024022433A1 (zh) Prmt5抑制剂
WO2024037608A1 (zh) Prmt5抑制剂
WO2024061345A1 (zh) Hpk1抑制剂、制备方法及其用途
JP7323218B2 (ja) 置換された縮合芳香環誘導体、その組成物、およびそれらの使用
WO2023207556A1 (zh) Prmt5-mta抑制剂
WO2023134688A1 (zh) 一种rock抑制剂的盐及盐的晶型、组合物和药物用途
WO2021164697A1 (zh) 取代的酰胺衍生物及其组合物及用途
WO2023216910A1 (zh) 取代的双环杂芳基化合物作为usp1抑制剂
CN117986248A (zh) Prmt5抑制剂

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: 23791281

Country of ref document: EP

Kind code of ref document: A1