WO2021218939A1 - 稠环化合物及其在医药上的应用 - Google Patents

稠环化合物及其在医药上的应用 Download PDF

Info

Publication number
WO2021218939A1
WO2021218939A1 PCT/CN2021/090082 CN2021090082W WO2021218939A1 WO 2021218939 A1 WO2021218939 A1 WO 2021218939A1 CN 2021090082 W CN2021090082 W CN 2021090082W WO 2021218939 A1 WO2021218939 A1 WO 2021218939A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkylene
compound
alkyl
cycloalkyl
aryl
Prior art date
Application number
PCT/CN2021/090082
Other languages
English (en)
French (fr)
Inventor
吴颢
陈小平
余军
路渊
何将旗
王维
湛波
李波燕
张运来
王冬
谢秀军
朱小惯
兰宏
王家炳
丁列明
Original Assignee
贝达药业股份有限公司
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 贝达药业股份有限公司 filed Critical 贝达药业股份有限公司
Priority to CN202180029506.3A priority Critical patent/CN115427414A/zh
Priority to US17/997,328 priority patent/US20230203055A1/en
Publication of WO2021218939A1 publication Critical patent/WO2021218939A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/052Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being six-membered
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D471/22Heterocyclic 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 systems contains four or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • 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
    • C07D487/22Heterocyclic 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 four or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • C07D491/14Ortho-condensed systems
    • C07D491/147Ortho-condensed systems the condensed system containing one ring with oxygen as ring hetero atom and two rings with nitrogen as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/22Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains four or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/22Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains four or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Definitions

  • the present invention relates to a new type of fused ring compound, which has cancer therapeutic activity.
  • the present invention also relates to preparation methods of these compounds and pharmaceutical compositions containing them.
  • RAS is the gene with the highest mutation rate in human tumors. About 20-30% of all tumors have RAS mutations, about 98% of pancreatic cancer, 52% of colon cancer, and 43% of multiple myeloma. And 32% of lung adenocarcinomas have RAS gene mutations.
  • the most common mutation mode of RAS is point mutation, which often occurs in codons 12, 13, and 61, among which codon 12 is the most common mutation.
  • KRAS-G12C mutations account for about 10-20% of KRAS mutations, and 14% of non-small cell lung cancers.
  • RAS protein is a low-molecular-weight guanosine triphosphate (GTP) binding protein with only one polypeptide chain.
  • GTP low-molecular-weight guanosine triphosphate
  • RAS includes two conformations: the active GTP binding conformation and the inactive GDP binding conformation. Under certain conditions, they can be transformed into each other to form the RAS cycle and regulate the activation of multiple downstream signaling pathways. The most important ones include the RAF-MEK-ERK and PI3K-AKT-mTOR signaling pathways.
  • RAS is called the "transmission of cellular signaling network”. Molecular switch”. Under normal circumstances, RAS is in an inactive state combined with GDP, and RAS is activated after receiving upstream signal stimulation, and the signal chain is only temporarily active. However, when RAS is mutated, the frequency of exchange between RAS and GDP/GTP is accelerated. RAS can bind to GTP for a long time, so that RAS and downstream signals are activated for a long time, and cell proliferation is out of control, leading to malignant transformation of cells.
  • KRAS G12C inhibitors AMG510 (WO2018217651A1) and MRTX849 (WO2019099524A1) have entered the late clinical stage.
  • the present invention provides a KRAS modulator with a novel structure.
  • the present invention provides a compound represented by general formula (I), its tautomers, deuterated compounds or pharmaceutical salts:
  • R 1 is each independently selected from H, amino, halogen, C 1-3 cyano, C 1-3 hydroxyalkyl, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy; or 2 R 1 and the connected atoms together form a C 3-6 cycloalkyl group or a 3-6 membered heterocyclic group;
  • R 2 is acryl or substituted acryl
  • Ring A is selected from C 3-10 cycloalkyl, 3-10 membered heterocyclic group, C 6-12 aryl, 5-12 membered heteroaryl;
  • X 3 is selected from C, CH or N;
  • X 4 is selected from CR 6 or N;
  • X 5 is selected from CR 7 or N;
  • X 6 is selected from CR 8 or N;
  • X 7 is selected from NR 9 or CHR 9 ;
  • R 6 or R 8 are independently selected from H, halogen, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 3-4 cycloalkyl, C 3-12 heterocycloalkane Group, C 2-3 alkenyl, C 2-3 alkynyl, aryl or heteroaryl;
  • R 7 is selected from H, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, -NH-C 1-6 alkyl, -N (C 1-6 alkyl) 2. Cyano or halogen;
  • R 9 is selected from C 1-6 alkyl, -C 0-3 alkylene-C 6-14 aryl, -C 0-3 alkylene-(5-14 membered heteroaryl), -C 0- 3 alkylene-C 3-14 cycloalkyl, -C 0-3 alkylene-(3-14 membered heterocycloalkyl), -OC 0-3 alkylene-C 6-14 aryl,- OC 0-3 alkylene-(5-14 membered heteroaryl), -OC 0-3 alkylene-C 3-14 cycloalkyl, -OC 0-3 alkylene-(3-14 membered hetero Cycloalkyl), -NH-C 1-6 alkyl, -N (C 1-6 alkyl) 2 , -NH-C 0-3 alkylene-C 6-14 aryl, -NH-C 0 -3 alkylene group - (5-14 membered heteroaryl), - NH-C 0-3 alkylene -
  • a or b is independently selected from 1, 2, 3 or 4;
  • formula (I) is selected from compounds of formula (I-1),
  • R 1 is each independently selected from H, amino, halogen, C 1-3 cyano, C 1-3 hydroxyalkyl or C 1-6 alkyl; or 2 R 1 and the connected atoms together form C 3-6 Cycloalkyl or 3-6 membered heterocyclic group;
  • R 2 is acryl or substituted acryl
  • Ring A is selected from C 3-10 cycloalkyl, 3-10 membered heterocyclic group, C 6-12 aryl, 5-12 membered heteroaryl;
  • X 3 is selected from C, CH or N;
  • X 4 is selected from CR 6 or N;
  • R 6 is independently selected from H, halogen, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 3-4 cycloalkyl, C 3-12 heterocycloalkyl, C 2-3 alkenyl, C 2-3 alkynyl, aryl or heteroaryl;
  • R 7 is selected from H, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, -NH-C 1-6 alkyl, -N (C 1-6 alkyl) 2. Cyano or halogen;
  • R 9 is selected from -C 0-3 alkylene-C 6-14 aryl, -C 0-3 alkylene-(5-14 membered heteroaryl), -C 0-3 alkylene-C 3 -14 cycloalkyl, -C 0-3 alkylene-(3-14 membered heterocycloalkyl), -OC 0-3 alkylene-C 6-14 aryl, -OC 0-3 alkylene -(5-14 membered heteroaryl), -OC 0-3 alkylene-C 3-14 cycloalkyl, -OC 0-3 alkylene-(3-14 membered heterocycloalkyl), -NH -C 1-6 alkyl, -N(C 1-6 alkyl) 2 , -NH-C 0-3 alkylene-C 6-14 aryl, -NH-C 0-3 alkylene-( 5-14 membered heteroaryl), - NH-C 0-3 alkylene -C 3-14 cyclo
  • a or b is independently selected from 1, 2, 3 or 4;
  • R 2 in formula (I) is selected from
  • the A ring in formula (I) is selected from
  • R 3 in formula (I) is selected from H, halogen, oxo, -OR a , -C 0-3 alkylene-N(R a ) 2 , C 1-6 haloalkyl, R a is independently H, C 1-6 alkyl, C 1-6 haloalkyl.
  • the substituent of X 3 in formula (I) is selected from C, CH or N.
  • X 4 in formula (I) is selected from CR 6 or N, and the substituent of R 6 is selected from halogen, C 1-3 alkyl, C 1-3 haloalkyl or C 2-3 alkenyl .
  • X 5 in formula (I) is selected from CR 7 or N, and the substituent of R 7 is selected from H or C 1-3 alkyl.
  • X 6 in formula (I) is selected from CR 8 or N, and the substituent of R 8 is selected from H, halogen or C 1-3 alkyl, preferably X 6 is N.
  • X 7 in formula (I) is selected from NR 9 or CHR 9 , preferably NR 9 .
  • R 9 in formula (I) is selected from -C 0-3 alkylene-C 6-14 aryl, -C 0-3 alkylene-(5-14 membered heteroaryl), -C 0-3 alkylene-C 3-14 cycloalkyl, -C 0-3 alkylene-(3-14 membered heterocycloalkyl), the -C 0-3 alkylene-C 6 -14 aryl, -C 0-3 alkylene-(5-14 membered heteroaryl), -C 0-3 alkylene-C 3-14 cycloalkyl, -C 0-3 alkylene- (3-14 membered heterocycloalkyl) unsubstituted or further selected from halogen, C 1-3 alkyl, C 1-3 alkenyl, C 1-3 haloalkyl, -(CH 2 ) 0-3 -S (R a) 5, C 1-3 alkoxy, C 3-6 cycloalkyl, C 1-3 al
  • formula (I) is selected from compounds of formula (Ia) to formula (Ie),
  • the present invention also provides a compound represented by general formula (II), its tautomer, deuterated product or pharmaceutical salt:
  • R 10 is selected from C 1-6 alkyl, -C 0-3 alkylene-C 6-14 aryl, -C 0-3 alkylene-(5-14 membered heteroaryl), -C 0- 3 alkylene-C 3-14 cycloalkyl, -C 0-3 alkylene-(3-14 membered heterocycloalkyl), -OC 0-3 alkylene-C 6-14 aryl,- OC 0-3 alkylene-(5-14 membered heteroaryl), -OC 0-3 alkylene-C 3-14 cycloalkyl, -OC 0-3 alkylene-(3-14 membered hetero Cycloalkyl), -NH-C 1-6 alkyl, -N(C 1-6 alkyl) 2 , -NH-C 0-3 alkylene-C 6-14 aryl, -NH-C 0 -3 alkylene group - (5-14 membered heteroaryl), - NH-C 0-3 alkylene -
  • R 11 is selected from H, amino, halogen, C 1-3 cyano, C 1-3 hydroxyalkyl, C 1-6 alkyl; preferably methyl or ethyl cyano;
  • R 12 is acryl or substituted acryl; preferably
  • R 13 is selected from H, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, -NH-C 1-6 alkyl, -N (C 1-6 alkyl) 2. Cyano or halogen; preferably H;
  • Ring B is selected from C 3-10 cycloalkyl, 3-10 membered heterocyclic group, C 6-12 aryl, 5-12 membered heteroaryl;
  • the substituent of X 8 is selected from CR 15 or N; preferably N, halogen or CH;
  • X 9 is selected from C, CH or N;
  • X 10 or X 11 are independently selected from -O-(CH 2 ) 0-2 -or CH 2;
  • c or d is independently selected from 1, 2, 3 or 4;
  • formula (II) is selected from compounds of formula (IIa) to (IId),
  • the present invention also provides a pharmaceutical composition, characterized in that the pharmaceutical composition comprises a therapeutically effective amount of at least one compound represented by formula (I) or formula (II) and at least one pharmaceutically acceptable Accessories.
  • the present invention further provides a pharmaceutical composition, characterized in that the therapeutically effective amount of at least one compound represented by formula (I) or formula (II) and pharmaceutically acceptable excipients have a mass percentage of 0.0001 :1-10.
  • the invention provides the application of the compound or pharmaceutical composition represented by the structural formula (I) or the formula (II) in the preparation of medicines.
  • the method for preparing the compound of formula (I) includes the following steps:
  • the compound of formula (IA) undergoes an acylation reaction with compound XR 2 under basic conditions to obtain a compound of formula (I), its tautomers, deuterated compounds or pharmaceutical salts, wherein X is halogen; R 1 -R 3.
  • X 1 -X 7 , a or b are as defined in formula (I).
  • the application is an application in the preparation of a medicine for treating and/or preventing cancer.
  • the application is an application for preparing a medicine for treating diseases mediated by KRAS G12C.
  • the disease is cancer.
  • the cancer is selected from breast cancer, multiple myeloma, bladder cancer, endometrial cancer, gastric cancer, cervical cancer, rhabdomyosarcoma, non-small cell lung cancer, small cell lung cancer, pleomorphic lung cancer, ovarian cancer, esophagus Carcinoma, melanoma, colorectal cancer, hepatocellular carcinoma, head and neck tumor, hepatobiliary cell carcinoma, myelodysplastic syndrome, malignant glioma, prostate cancer, thyroid cancer, Xuwang's cell tumor, lung squamous cell Carcinoma, lichenoid keratosis, synovial sarcoma, skin cancer, pancreatic cancer, testicular cancer or liposarcoma.
  • the present invention also provides a method for treating and/or preventing diseases, which comprises administering a therapeutically effective amount of at least any one of the compounds represented by structural formula (I) or formula (II) or a pharmaceutical composition containing the same to a subject.
  • the present invention also provides a method for treating and/or preventing diseases mediated by KRAS G12C, which comprises administering a therapeutically effective amount of at least any one of the compounds represented by structural formula (I) or formula (II) or containing it to a subject.
  • Pharmaceutical composition comprising
  • the present invention also provides a method for treating cancer, which comprises administering a therapeutically effective amount of at least any one of the compounds represented by structural formula (I) or formula (II) or a pharmaceutical composition containing the same to a subject.
  • the KRAS G12C-mediated disease is cancer.
  • the cancer is selected from breast cancer, multiple myeloma, bladder cancer, endometrial cancer, gastric cancer, cervical cancer, rhabdomyosarcoma, non-small cell lung cancer, small cell lung cancer, pleomorphic Lung cancer, ovarian cancer, esophageal cancer, melanoma, colorectal cancer, hepatocellular tumor, head and neck tumor, hepatobiliary cell carcinoma, myelodysplastic syndrome, malignant glioma, prostate cancer, thyroid cancer, Xuwang's cell Tumor, lung squamous cell carcinoma, lichenoid keratosis, synovial sarcoma, skin cancer, pancreatic cancer, testicular cancer or liposarcoma.
  • halogen used in the present invention refers to fluorine, chlorine, bromine, or iodine.
  • alkyl includes a linear or branched monovalent saturated hydrocarbon group.
  • alkyl includes methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 3-(2-methyl)butyl, 2 -Pentyl, 2-methylbutyl, neopentyl, n-hexyl, 2-hexyl, 2-methylpentyl, etc.
  • a "C1-8 alkyl group” in the "1-8” refers to a straight or branched chain radical form with 7 or 8 carbon atoms arranged group.
  • C 1-3 alkylene means methylene, 1,2-ethylene, 1,3-propylene or 1,2-isopropylidene.
  • Alkoxy refers to the oxyether form of the aforementioned linear or branched alkyl group, that is, -O-alkyl.
  • compositions comprising "a” pharmaceutically acceptable excipient can be interpreted to mean that the composition includes “one or more” pharmaceutically acceptable excipients.
  • aryl in the present invention, unless otherwise specified, refers to an unsubstituted or substituted monocyclic or condensed ring aromatic group including carbon ring atoms.
  • the aryl group is a 6 to 10 membered monocyclic or bicyclic aromatic ring group.
  • it is phenyl and naphthyl.
  • Most preferred is phenyl.
  • the aryl ring may be fused to a heteroaryl group, a heterocyclic group or a cycloalkyl group, wherein the ring connected to the parent structure is an aryl ring, and non-limiting examples include, but are not limited to, benzocyclopentyl.
  • heterocyclic group in the present invention, unless otherwise specified, refers to an unsubstituted or substituted 3-8 membered stable monomer consisting of carbon atoms and 1-3 heteroatoms selected from N, O or S Ring system, which is a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, which includes 3 to 20 carbon atoms, wherein nitrogen or sulfur heteroatoms can be selectively oxidized, and nitrogen heteroatoms can be selectively The ground is quaternized.
  • the heterocyclic group can be attached to any heteroatom or carbon atom to form a stable structure.
  • heterocyclic groups include, but are not limited to, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, oxopiperazinyl, oxopiperidinyl, tetrahydrofuranyl, dioxolane, Tetrahydroimidazolyl, tetrahydrothiazolyl, tetrahydrooxazolyl, tetrahydropyranyl, morpholinyl, thiomorpholinyl, thiomorpholinyl sulfoxide, thiomorpholinylsulfone and tetrahydro Oxadiazolyl.
  • heteroaryl in the present invention, unless otherwise specified, refers to an unsubstituted or substituted stable 5- or 6-membered monocyclic aromatic ring system or an unsubstituted or substituted 9- or 10-membered benzo Condensed heteroaromatic ring system or bicyclic heteroaromatic ring system, which consists of carbon atoms and 1-4 heteroatoms selected from N, O or S, and wherein the nitrogen or sulfur heteroatoms can be selectively Upon oxidation, the nitrogen heteroatoms can be selectively quaternized.
  • Heteroaryl groups can be attached to any heteroatom or carbon atom to form a stable structure.
  • heteroaryl groups include, but are not limited to, thienyl, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrazolyl, pyrrolyl, thiazolyl, thiadiazolyl, triazolyl, pyridyl, pyridyl Azinyl, indolyl, azaindolyl, indazolyl, benzimidazolyl, benzofuranyl, benzothienyl, benzisoxazolyl, benzothiazolyl, benzothiazolyl, benzene And thiadiazolyl, benzotriazolyl adenine, quinolinyl or isoquinolinyl.
  • the heteroaryl group may be fused on an aryl, heterocyclic or cycloalkyl ring, wherein the ring connected to the parent structure is a heteroaryl ring.
  • cycloalkyl refers to a cyclic saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent having 3-10 carbon atoms, for example, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl .
  • substituted means that one or more hydrogen atoms in the group are respectively replaced by the same or different substituents.
  • the substituents are independently selected from the group comprising -F, -Cl, -Br, -I, -OH, trifluoromethoxy, ethoxy, propoxy, isopropoxy, n-butoxy Group, isobutoxy, tert-butoxy, -SCH 3 , -SC 2 H 5 , formaldehyde, -C(OCH 3 ), cyano, nitro, -CF 3 , -OCF 3 , amino, dimethyl Groups of amino group, methylthio group, sulfonyl group and acetyl group.
  • substituted alkyl groups include, but are not limited to, 2,3-dihydroxypropyl, 2-aminoethyl, 2-hydroxyethyl, pentachloroethyl, trifluoromethyl, methoxymethyl, pentafluoroethyl , Phenylmethyl, Dioxenylmethyl and Piperazinylmethyl.
  • substituted alkoxy groups include, but are not limited to, 2-hydroxyethoxy, 2-fluoroethoxy, 2,2-difluoroethoxy, 2-methoxyethoxy, 2-aminoethoxy, 2,3-Dihydroxypropoxy, cyclopropylmethoxy, aminomethoxy, trifluoromethoxy, 2-diethylaminoethoxy, 2-ethoxycarbonylethoxy, 3- Hydroxypropoxy.
  • pharmaceutically acceptable salt refers to a salt prepared from a pharmaceutically acceptable non-toxic base or acid.
  • the corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic bases, including inorganic bases and organic bases.
  • Salts derived from inorganic bases include aluminum, ammonium, calcium, copper (high and low prices), ferric, ferrous, lithium, magnesium, manganese (high and low prices), potassium, sodium, zinc and the like. Particularly preferred are the salts of ammonium, calcium, magnesium, potassium, and sodium.
  • Non-toxic organic bases that can be derivatized into salts include primary, secondary and tertiary amines, as well as cyclic amines and amines containing substituents, such as naturally occurring and synthetic amines containing substituents.
  • non-toxic organic bases capable of forming salts, including ion exchange resins and arginine, betaine, caffeine, choline, N',N'-dibenzylethylenediamine, diethylamine, 2 -Diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, reduced glucosamine, glucosamine, histidine, haamine, isopropylamine , Lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resin, procaine, purine, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, etc.
  • the corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic acids, including inorganic acids and organic acids.
  • acids include, for example, acetic acid, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, citric acid, ethanesulfonic acid, isethionic acid, formic acid, fumaric acid, gluconic acid, glutamic acid, hydrobromic acid, Hydroiodic acid, perchloric acid, hydrochloric acid, isethionic acid, propionic acid, glycolic acid, lactic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, mucic acid, nitric acid, oxalic acid, pantothenic acid, phosphoric acid , Succinic acid, sulfuric acid, 2-naphthalenesulfonic acid, cyclohexylamine sulfonic acid, salicylic acid, saccharic acid, trifluor acid, acetic acid, benzen
  • the compound represented by formula (I) or formula (II) will be used as a medicine, it is preferable to use a certain purity, for example, at least 60% purity, more suitable purity is at least 75%, and particularly suitable purity is at least 98%. % (% is a weight ratio).
  • the prodrug of the compound of the present invention is included in the protection scope of the present invention.
  • the prodrug refers to a functional derivative that is easily converted into a desired compound in the body.
  • any pharmaceutically acceptable salt, ester, ester salt or other derivative of the compound of the present application can directly or indirectly provide the compound of the present application or its pharmaceutically active metabolite or Residues.
  • the compound of the present invention may contain one or more asymmetric centers, and may produce diastereomers and optical isomers from this.
  • the present invention includes all possible diastereomers and their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers and their pharmaceutically acceptable salts.
  • the present invention includes any possible solvates and polymorphs.
  • the type of solvent that forms the solvate is not particularly limited, as long as the solvent is pharmaceutically acceptable.
  • water, ethanol, propanol, acetone and similar solvents can be used.
  • composition refers to a product containing a specified amount of each specified ingredient, and any product produced directly or indirectly from a combination of specified amounts of each specified ingredient. Therefore, a pharmaceutical composition containing the compound of the present invention as an active ingredient and a method for preparing the compound of the present invention are also part of the present invention.
  • some crystalline forms of the compound may exist in polymorphs, and this polymorph is included in the present invention.
  • some compounds may form solvates with water (ie, hydrates) or common organic solvents, and such solvates also fall within the scope of the present invention.
  • the pharmaceutical composition provided by the present invention includes as an active ingredient a compound represented by formula (I) or formula (II) (or a pharmaceutically acceptable salt thereof), a pharmaceutically acceptable excipient and other optional Therapeutic components or excipients.
  • a pharmaceutical composition of the present invention includes oral, rectal, topical and A pharmaceutical composition for parenteral administration (including subcutaneous administration, intramuscular injection, and intravenous administration).
  • the pharmaceutical composition of the present invention can be conveniently prepared in a unit dosage form known in the art and prepared by any preparation method known in the pharmaceutical field.
  • the dosage level of the drug is about 0.01 mg/kg body weight to 150 mg/kg body weight per day, or 0.5 mg to 7 g per patient per day.
  • the effective treatment drug dosage level is 0.01 mg/kg body weight to 50 mg/kg body weight per day, or 0.5mg to 3.5g per patient per day.
  • the specific dosage level and treatment plan for any particular patient will depend on many factors, including the activity of the specific compound used, age, weight, overall health, gender, diet, time of administration, route of administration, excretion rate, drug combination The condition and the severity of the specific disease being treated.
  • Step A Compound I-1 is methylated to obtain compound I-2;
  • Step B Compound 1-2 undergoes substitution reaction with PMBNH 2 to obtain compound 1-3;
  • Step C Compound I-3 removes the PMB protecting group under the action of Pd/C;
  • Step D Compound I-4 converts the amino group into bromine under the action of CuBr 2;
  • Step E Compound I-5 is hydrolyzed under alkaline conditions such as LiOH to obtain compound I-6;
  • Step F Compound I-6 converts carboxyl group into amide under the action of CDI;
  • Step G Compound I-7 is introduced into R 9 group under the action of oxalyl chloride
  • Step H Compound I-8 is self-closing under the action of a strong base such as NaH to obtain compound I-9;
  • Step I Phosphorus oxychloride converts -OH of I-9 into -Cl with high temperature stirring to obtain compound I-10;
  • Step J Compound I-10 and Under alkaline conditions, such as DIEA, undergo substitution reaction to compound I-11;
  • Step K I-11 and Under the action of Pd catalyst such as Pd(dppf)Cl 2 , the A ring is introduced through Suzuki coupling to obtain compound I-12, n is selected from 1, 2, 3 or 4;
  • Pd catalyst such as Pd(dppf)Cl 2
  • Step L I-12 closes the ring by itself under alkaline conditions such as Cs 2 CO 3;
  • Step M Under acidic conditions, such as trifluoroacetic acid, etc., remove the Boc protecting group;
  • Step N Under weak alkaline conditions such as sodium bicarbonate, etc., the target compound I can be obtained by introducing the R 2 group.
  • Step O Synthesize II-1 through steps similar to I-12, and II-1 closes the ring by itself under alkaline conditions such as Cs 2 CO 3;
  • Step P Under acidic conditions, such as trifluoroacetic acid, etc., remove the Boc protecting group;
  • Step Q Under weakly alkaline conditions such as sodium bicarbonate, etc., the target compound I can be obtained by introducing the R 12 group.
  • CDI carbonyl diimidazole
  • DIPEA diisopropylethylamine
  • DMSO dimethyl sulfoxide
  • PE petroleum ether
  • THF Tetrahydrofuran
  • TFA trifluoroacetic acid
  • pre-HPLC Preparative high performance liquid chromatography
  • CDI N,N'-carbonyldiimidazole
  • the substrate M-7 (4.61g, 18.15mmol) was dissolved in THF (45mL), and then under nitrogen protection, oxalyl chloride (9.21g, 72.60mmol, 6.14mL) was added, and the reaction was refluxed under heating for 1hr, and then spin-dried under reduced pressure. Washed with DCM, concentrated again, and then added a solution of 2-isopropyl-4-methyl-3-pyridinamine (4.09 g, 27.22 mmol) in DCM (40 mL), and stirred at room temperature for 1 hr.
  • Example 1 Compound 1 ((S)-1-(4-acryloyl-2-methylpiperazin-1-yl)-11-fluoro-4-(2-isopropyl-4-methylpyridine- Synthesis of 3-yl)-4H-isochromene bridge [4,3-h]quinazolin-3(6H)-one)
  • reaction was completed, it was cooled to the chamber and added The reaction system was diluted with EA, and then the organic phase was washed with saturated sodium bicarbonate, and after washing with saturated brine, the organic phase was dried, spin-dried, and proceeded directly to the next reaction.
  • Example 2 Compound 2 ((S)-8-(4-acryloyl-2-methylpiperazin-1-yl)-12-fluoro-11-(2-isopropyl-4-methylpyridine- Synthesis of 3-yl)-5H-isochrome [3,4-g]quinazolin-10(11H)-one)
  • Example 3 Compound 3 ((S)-1-(acryloyl-2-methylpiperazin-1-yl)-10,11-difluoro-4-(2-isopropyl-4-methylpyridine) Synthesis of -3-yl)-4H-isochrome [4,3-h]quinazolin-3(6H)-one)
  • Example 4 Compound 4 ((S)-8-(acryloyl-2-methylpiperazin-1-yl)-1,12-difluoro-11-(2-isopropyl-4-methylpyridine) Synthesis of -3-yl)-5H-isochrome [3,4-g]quinazolin-10(11H)-one)
  • Example 5 Compound 5((S)-1-(acryloyl-2-methylpiperazin-1-yl)-10-chloro-11-fluoro-4-(2-isopropyl-4-methyl Synthesis of (pyridin-3-yl)-4H-isochrome[4,3-h]quinazolin-3(6H)-one)
  • Example 6 Compound 6((S)-8-(acryloyl-2-methylpiperazin-1-yl)-1-chloro-12-fluoro-11-(2-isopropyl-4-methyl Synthesis of (pyridin-3-yl)-5H-isochrome [3,4-g]quinazolin-10(11H)-one)
  • Example 7 Compound 7 (10-((S)-4-(acryloyl-2-methylpiperazin-1-yl)-8-fluoro-13-(2-isopropyl-4-methylpyridine) Synthesis of -3-yl)-2,3-dihydrobenzo[4,5]oxydipine[3,2-h]quinolin-12(13H)one)
  • Example 8 Compound 8 (9-((S)-4-(acryloyl-2-methylpiperazin-1-yl)-13-fluoro-12-(2-isopropyl-4-methylpyridine) -3-yl)-5,6-dihydrobenzo[4,5]oxydipine[2,3-h]quinolin-11(12H)one)
  • Example 9 Compound 9 ((S)-1-(acryloyl-2-methylpiperazin-1-yl)-11-fluoro-10-hydroxy-4-(2-isopropyl-4-methyl Synthesis of (pyridin-3-yl)-4H-isochrome[4,3-h]quinazolin-3(6H)-one)
  • Example 10 Compound 10 ((S)-8-(acryloyl-2-methylpiperazin-1-yl)-12-fluoro-1-hydroxy-11-(2-isopropyl-4-methyl Synthesis of (pyridin-3-yl)-5H-isochrome [3,4-g]quinazolin-10(11H)-one)
  • Example 11 Compound 11 ((S)-1-(acryloyl-2-methylpiperazin-1-yl)-11-fluoro-10-methoxy-4-(2-isopropyl-4- Synthesis of (methylpyridin-3-yl)-4H-isochrome[4,3-h]quinazolin-3(6H)-one)
  • Example 12 Compound 12 ((S)-8-(acryloyl-2-methylpiperazin-1-yl)-12-fluoro-1-methoxy-11-(2-isopropyl-4- Synthesis of methylpyridin-3-yl)-5H-isochrome [3,4-g]quinazolin-10(11H)-one)
  • Example 13 Compound (S)-1-(4-acryloyl-2-methylpiperazin-1-yl)-11-chloro-10-fluoro-4-(2-isopropyl-4-methyl Synthesis of pyridin-3-yl)-4H-isochrome[4,3-h]quinazolin-3(6H)-one
  • reaction solution was added to 600mL of cold saturated aqueous ammonium chloride solution, 500mL of ethyl acetate was added, the layers were separated, the organic phase was separated, the aqueous phase was extracted with ethyl acetate 200mL*1, the organic phases were combined, and saturated sodium chloride solution 2 *300mL washing, the organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. A light brown solid (12.19 g, 36.68 mmol, 87.92% yield) was obtained, which was used directly in the next step.
  • compound 13-2 (12.19g, 36.68mmol) was dissolved in DCM (122mL), and 4 drops of DMF were added dropwise; then cooled in an ice-water bath, and oxalyl chloride (13.97g, 110.03mmol, 9.31mL) was added dropwise DCM (14mL); then stirred and reacted for half an hour, the reaction solution was concentrated under reduced pressure, and DCM 20mL ⁇ 2 was used twice; ammonium hydroxide (15M, 684.66mL) was cooled with an ice water bath; the intermediate residue was added dropwise in dry THF (60mL), after dripping, let it rise to room temperature and react for 1hs.
  • Dissolve compound 13-4 (2000mg, 3.94mmol) in dimethoxyethane (20mL) at room temperature, add NaH (472.83mg, 11.82mmol, 60% purity) under nitrogen protection, and stir for 2 minutes at room temperature , Until the bubbles are reduced to a small amount, microwave reaction at 90°C for 10 minutes.
  • the organic phases are combined, dried over anhydrous sodium sulfate, filtered, and the filtrate is concentrated under reduced pressure. A brown-yellow solid (1.6 g, 3.75 mmol, 95.17% yield) was obtained, which was directly thrown into the step.
  • reaction solution was diluted with DCM 10ml, washed with saturated brine (3*15ml), the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by prep-HPLC to obtain compound 13 (18.9 mg, 32.14umol, 40.76% yield) as an off-white solid powder.
  • Example 14 Compound (S)-1-(4-acryloyl-2-methylpiperazin-1-yl)-10-fluoro-4-(2-isopropyl-4-methylpyridine-3- Yl)-11-vinyl-4H-isochrome[4,3-h]quinazolin-3(6H)-one
  • reaction solution was diluted with DCM 10ml, washed with saturated brine (3*15ml), the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by pre-HPLC. A pale yellow solid powder product (26.1 mg, 45.03umol, 44.72% yield) was obtained.
  • Example 15 Compound (S)-1-(4-acryloyl-2-methylpiperazin-1-yl)-10-fluoro-4-(2-isopropyl-4-methylpyridine-3- Yl)-11-methyl-4H-isochrome[4,3-h]quinazolin-3(6H)-one
  • reaction solution was diluted with DCM 10ml, washed with saturated brine (3*10ml), the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by pre-HPLC. A white solid powder product (11.5mg, 20.26umol, 23.46% yield) was obtained.
  • Example 16 Compound (S)-1-(4-acryloyl-2-methylpiperazin-1-yl)-10-fluoro-4-(2-isopropyl-4-methylpyridine-3- Yl)-11-(trifluoromethyl)-4H-isochrome[4,3-h]quinazolin-3(6H)-one
  • Example 17 Compound (S)-8-(4-acryloyl-2-methylpiperazin-1-yl)-1,2,12-trifluoro-11-(2-isopropyl-4-methyl Of pyridin-3-yl)-5H isochrome [3,4-g]quinazolin-10(11H)-one
  • Example 18 Compound S)-1-(4-acryloyl-2-methylpiperazin-1-yl)-11-chloro-9,10-difluoro-4-(2-isopropyl-4- Synthesis of methylpyridin-3-yl)-4H-isochrome[4,3-h]quinazolin-3(6H)-one
  • Example 19 Compound 10-((S)-4-acryloyl-2-methylpiperazin-1-yl)-8-chloro-7-fluoro-13-(2-isopropyl-4-methyl Synthesis of pyridin-3-yl)-2,3-dihydrobenzo[4,5]oxa[3,2-h]quinazolin-12(13H)-one
  • Example 20 Compound (S)-7-(4-acryloyl-2-methylpiperazin-1-yl)-5-chloro-10-(2-isopropyl-4-methylpyridine-3- Yl)-10H pyridine[3',2': 4,5]pyran[3,2-h]quinazolin-9(12H)-one
  • Example 21 Compound (S)-1-(4-acryloyl-2-methylpiperazin-1-yl)-13-fluoro-4-(2-isopropyl-4-methylpyridine-3- Yl)-4H-benzo[5,6]isochrome[4,3-h]quinazolin-3(6H)-one
  • Example 22 Compound (S)-10-(4-acryloyl-2-methylpiperazin-1-yl)-14-fluoro-13-(2-isopropyl-4-methylpyridine-3- Yl)-7H-benzo[5,6]isochrome[3,4-g]quinazolin-12(13H)-one
  • Example 23 Compound (S)-1-(4-acryloyl-2-methylpiperazin-1-yl)-11-chloro-4-(2-isopropyl-4-methylpyridine-3- Yl)-4H-pyridine[3',4': 4,5]pyrano[3,2-h]quinazolin-3(6H)-one
  • Example 24 Compound 10-((S)-4-acryloyl-2-methylpiperazin-1-yl)-8-chloro-13-(4,6-diethylpyrimidin-5-yl)- Synthesis of 7-fluoro-2,3-dihydrobenzo[4,5]epoxyheptane[3,2-h]quinazolin-12(13H)-one
  • Example 25 Compound 2-(1-acryloyl-4-(10,11-difluoro-4-(2-isopropyl-4-methylpyridin-3-yl)-3-oxo-3, Synthesis of 6-dihydro-4H-isochrome[4,3-h]quinazolin-1-yl)piperazin-2-yl)acetonitrile
  • Example 26 Compound 2-(1-acryloyl-4-(1,12-difluoro-11-(2-isopropyl-4-methylpyridin-3-yl)-10-oxy-10,11 -Dihydro-5H-isochrome [3,4-g]quinazolin-8-yl)piperazin-2-yl)acetonitrile
  • Example 27 Compound (S)-1-(4-acryloyl-2-methylpiperazin-1-yl)-11-chloro-9,10-difluoro-4-(3-isopropyl-6 Synthesis of ,7-Dihydro-5H-cyclopentyl[c]pyridin-4-yl)-4H-isochromatic[4,3-h]quinazolin-3(6H)-one
  • Example 28 Compound (S)-1-(4-acryloyl-2-methylpiperazin-1-yl)-11-chloro-10-fluoro-7-hydroxy-4-(2-isopropyl- Synthesis of 4-methylpyridin-3-yl)-4H-isochroma[4,3-h]quinazolin-3(6H)-one
  • Example 29 Compound (S)-1-(4-acryloyl-2-methylpiperazin-1-yl)-11-chloro-7,10-difluoro-4-(2-isopropyl-4 -Methylpyridin-3-yl)-4H-isochromatic [4,3-h]quinazolin-3(6H)-one
  • Example 30 Compound 10-((S)-4-acryloyl-2-methylpiperazin-1-yl)-8-chloro-6,7-difluoro-13-(2-isopropyl-4 -Methylpyridin-3-yl)-2,3-dihydrobenzo[4,5]oxa[3,2-h]quinazolin-12(13H)-one
  • Example 31 Compound 10-((S)-4-acryloyl-2-methylpiperazin-1-yl)-8-chloro-13-(3,5-diethylpyridin-4-yl)- Synthesis of 7-fluoro-2,3-dihydrobenzo[4,5]oxa[3,2-h]quinazolin-12(13H)-one
  • Example 32 Compound 10-((S)-4-acryloyl-2-methylpiperazin-1-yl)-8-chloro-13-(4,6-dicyclopropylpyrimidin-5-yl) Synthesis of -7-fluoro-2,3-dihydrobenzo[4,5]oxa[3,2-h]quinazolin-12(13H)-one
  • Example 33 Compound 1-(5-acryloyl-2,5-diazabicyclo[4.1.0]heptan-2-yl)-11-chloro-10-fluoro-4-(2-isopropyl Of 4-methylpyridin-3-yl)-4H-isochromenol[4,3-h]quinazolin-3(6H)-one
  • Example 34 Compound (S)-11-chloro-9,10-difluoro-1-(4-(2-fluoroacryloyl)-2-methylpiperazin-1-yl)-4-(2- Synthesis of isopropyl-4-methylpyridin-3-yl)-4H-isochromo[4,3-h]quinazolin-3(6H)-one
  • Examples 17-23 and 28-30 were synthesized by referring to the steps in Example 3.
  • the carboxylic acid was reduced to an aldehyde group and then to a hydroxyl group, and then the bromine atom was boronicated, using the common intermediate M-11 or 13-6
  • the target product was finally obtained by Suzuki coupling with the corresponding boronic ester, ring closure, deprotection and introduction of acryloyl group; in Examples 24, 27, 31 and 32, the chlorine was ethylated, isopropylated and cyclopropylated, respectively, Refer to the synthesis of M-11, and then Suzuki coupling with the corresponding boronic ester, ring closure, deprotection and introduction of acryloyl group to finally obtain the target product; Examples 25 and 26 were Boc protected and then referred to the synthesis of M-11, and then combined with the corresponding The boronic ester finally obtains the target product through Suzuki coupling, ring closure, deprotection and introduction of acryloyl group.
  • Example 35 Compound (S)-1-(4-acryloyl-2-methylpiperazin-1-yl)-11-chloro-9,10-difluoro-4-(2-isopropyl-4 -Methylpyridin-3-yl)-4H-isochromatic [4,3-h]quinazolin-3(6H)-one-6,6-d2
  • Example 36 Compound (S)-8-(4-acryloyl-2-methylpiperazin-1-yl)-11-(2-isopropyl-4-methylpyridin-3-yl)-5H -Synthesis of isochrome [3',4': 5,6]pyrido[2,3-d]pyrimidine-10(11H)-one
  • Example 37 Compound (S)-8-(4-acryloyl-2-methylpiperazin-1-yl)-1-hydroxy-11-(2-isopropyl-4-methylpyridine-3- Yl)-5H-isochrome [3',4': 5,6]pyrido[2,3-d]pyrimidine 10(11H)-one
  • Inhibition percentage (maximum-reading)/(maximum-minimum)*100.
  • the “maximum value” is the DMSO control; the “minimum value” indicates the cell-free control group.
  • Curve were fitted using Graphpad Prism software to obtain a value 50 IC.
  • Inhibition percentage (maximum-reading)/(maximum-minimum)*100.
  • the “maximum value” is the DMSO control; the “minimum value” indicates the cell-free control group.
  • Curve were fitted using Graphpad Prism software to obtain a value 50 IC.

Landscapes

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

Abstract

本发明涉及一种新型化合物(式 I ),其具有癌症治疗活性。本发明还涉及这些化合物的制备方法以及包含其的药物组合物。

Description

稠环化合物及其在医药上的应用 技术领域
本发明涉及一种新型稠环化合物,其具有癌症治疗活性。本发明还涉及这些化合物的制备方法以及包含其的药物组合物。
背景技术
临床数据显示,RAS是人类肿瘤中发生突变率最高的基因,所有肿瘤中,约20-30%有RAS突变,大约98%的胰腺癌,52%的结肠癌,43%的多发性骨髓瘤,及32%的肺腺癌中存在RAS基因突变。RAS最常见的突变方式是点突变,经常发生在12、13、61密码子,其中又以第12位密码子突变最常见。KRAS-G12C突变占KRAS突变的约10-20%,在非小细胞肺癌中占14%。RAS蛋白是一种只有一条多肽链的低分子量的三磷酸鸟苷(Guanosine triphosphate,GTP)结合蛋白,包括两种构象:有活性的GTP结合构象和无活性的GDP结合构象,这两种构象在一定条件下可以相互转化,构成RAS循环,调控多条下游信号通路的激活,其中最主要的包括RAF-MEK-ERK和PI3K-AKT-mTOR信号通路,RAS被称为细胞信号网络传递中的“分子开关”。正常情况下RAS为与GDP结合的非活化状态,而RAS接收上游信号刺激后被激活,信号链只有短暂的活性。可是当RAS发生突变后,RAS与GDP/GTP交换的频率被加快,RAS可与GTP长时间结合,使RAS及下游信号长期处于活化状态,细胞增殖失去控制,导致细胞恶性转化。
目前针对KRAS突变的药物研发是当前新药研究热点之一。KRAS G12C抑制剂AMG510(WO2018217651A1)和MRTX849(WO2019099524A1)已进入后期临床阶段。本发明提供一种新型结构的KRAS调节剂。
发明内容
本发明提供一种通式(I)所示的化合物、其互变异构体、氘代物或药用盐:
Figure PCTCN2021090082-appb-000001
其中,
R 1各自独立地选自H、氨基、卤素、C 1-3氰基、C 1-3羟基烷基、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基;或者2个R 1与相连的原子共同形成C 3-6环烷基或3-6元杂环基;
R 2为丙烯酰基或取代的丙烯酰基;
A环选自C 3-10环烷基、3-10元杂环基、C 6-12芳基、5-12元杂芳基;
X 1独立地选自-C(R 4) 1-2-(CH 2) 0-2-、-NR 4-(CH 2) 0-2-、-O-(CH 2) 0-2-、-S-(CH 2) 0-2-、
Figure PCTCN2021090082-appb-000002
Figure PCTCN2021090082-appb-000003
-CH=CH-或-N=CH-;
X 2独立地选自-C(R 5) 1-2-(CH 2) 0-2-、-NR 5-(CH 2) 0-2-、-O-(CH 2) 0-2-、-S-(CH 2) 0-2-、
Figure PCTCN2021090082-appb-000004
Figure PCTCN2021090082-appb-000005
-CH=CH-或-N=CH-;
X 3选自C、CH或N;
X 4选自CR 6或N;
X 5选自CR 7或N;
X 6选自CR 8或N;
X 7选自NR 9或CHR 9
R 6或R 8独立地选自H、卤素、C 1-3烷基、C 1-3卤代烷基、C 1-3烷氧基、C 3-4环烷基、C 3-12杂环烷基、C 2-3烯基、C 2-3炔基、芳基或杂芳基;
R 7选自H、羟基、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、-NH-C 1-6烷基、-N(C 1-6烷基) 2、氰基或卤素;
R 9选自C 1-6烷基、-C 0-3亚烷基-C 6-14芳基、-C 0-3亚烷基-(5-14元杂芳基)、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-O-C 0-3亚烷基-C 6-14芳基、-O-C 0-3亚烷基-(5-14元杂芳基)、-O-C 0-3亚烷基-C 3-14环烷基、-O-C 0-3亚烷基-(3-14元杂环烷基)、-NH-C 1-6烷基、-N(C 1-6烷基) 2、-NH-C 0-3亚烷基-C 6-14芳基、-NH-C 0-3亚烷基-(5-14元杂芳基)、-NH-C 0-3亚烷基-C 3-14环烷基、-NH-C 0-3亚烷基-(3-14元杂环烷基),R 9任选未取代或进一步被一个或多个R 9a取代基所取代;
R 3、R 4、R 5、R 9a各自独立地选自H、D、氧代、硝基、卤素、C 1-6烷基、C 1-6氰基、C 1-6卤代烷基、-C 0-3亚烷基-OR a、-C 0-3亚烷基-N(R a) 2、-C 0-3亚烷基-NR aC(=O)R a、-C 0-3亚烷基-NR aC(=O)OR a、-C 0-3亚烷基-NR aS(=O) 2R a、-C 0-3亚烷基-S(=O)R a、-C 0-3亚烷基-S(=O) 2R a、-C 0-3亚烷基-S(=O) 2N(R a) 2、-C 0-3亚烷基-SR a、-C 0-3亚烷基-S(R a) 5、-C 0-3亚烷基-C(=O)N(R a) 2、-C 0-3亚烷基-C(=O)R a、-C 0-3亚烷基-C(=O)OR a、C 2-6烯基、C 2-6炔基、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-C 0-3亚烷基-C 6-14芳基或-C 0-3亚烷基-(5-14元杂芳基),所述-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-C 0-3亚烷基-C 6-14芳基或-C 0-3亚烷基-(5-14元杂芳基)任选未取代或进一步被一个或多个R a取代基所取代,且每个R a独立地选自H、卤素、C 1-6烷基、C 1-6卤代烷基、C 3-14环烷基、3-14元杂环烷基、C 2-3烯基、C 2-3炔基、芳基或杂芳基;
a或b独立地选自1、2、3或4;
Figure PCTCN2021090082-appb-000006
为双键或单键。
一些实施方式中,式(I)选自式(I-1)化合物,
Figure PCTCN2021090082-appb-000007
其中,
R 1各自独立地选自H、氨基、卤素、C 1-3氰基、C 1-3羟基烷基或C 1-6烷基;或者2个R 1与相连的原子共同形成C 3-6环烷基或3-6元杂环基;
R 2为丙烯酰基或取代的丙烯酰基;
A环选自C 3-10环烷基、3-10元杂环基、C 6-12芳基、5-12元杂芳基;
X 1独立地选自-C(R 4) 2-(CH 2) 0-2-、-NR 4-(CH 2) 0-2-、-O-(CH 2) 0-2-、-S-(CH 2) 0-2-、
Figure PCTCN2021090082-appb-000008
Figure PCTCN2021090082-appb-000009
-C(=O)-(CH 2) 0-2-、-CH=CH-或-N=CH-,R 4独立地选自H、D、卤素或C 1-3烷基;
X 2独立地选自-C(R 5) 2-(CH 2) 0-2-、-NR 5-(CH 2) 0-2-、-O-(CH 2) 0-2-、-S-(CH 2) 0-2-、
Figure PCTCN2021090082-appb-000010
Figure PCTCN2021090082-appb-000011
-C(=O)-(CH 2) 0-2-、-CH=CH-或-N=CH-,R 5独立地选自H、D、卤素或C 1-3烷基;
X 3选自C、CH或N;
X 4选自CR 6或N;
R 6独立地选自H、卤素、C 1-3烷基、C 1-3卤代烷基、C 1-3烷氧基、C 3-4环烷基、C 3-12杂环烷基、C 2-3烯基、C 2-3炔基、芳基或杂芳基;
R 7选自H、羟基、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、-NH-C 1-6烷基、-N(C 1-6烷基) 2、氰基或卤素;
R 9选自-C 0-3亚烷基-C 6-14芳基、-C 0-3亚烷基-(5-14元杂芳基)、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-O-C 0-3亚烷基-C 6-14芳基、-O-C 0-3亚烷基-(5-14元杂芳基)、-O-C 0-3亚烷基-C 3-14环烷基、-O-C 0-3亚烷基-(3-14元杂环烷基)、-NH-C 1-6烷基、-N(C 1-6烷基) 2、-NH-C 0-3亚烷基-C 6-14芳基、-NH-C 0-3亚烷基-(5-14元杂芳基)、-NH-C 0-3亚烷基-C 3-14环烷基、-NH-C 0-3亚烷基-(3-14元杂环烷基),R 9任选未取代或进一步被1-4个R 9a取代基所取代;
R 3或R 9a各自独立地选自H、D、氧代、硝基、卤素、C 1-6烷基、C 1-6氰基、C 1-6卤代烷基、-C 0-3亚烷基-OR a、-C 0-3亚烷基-N(R a) 2、-C 0-3亚烷基-NR aC(=O)R a、-C 0-3亚烷基-NR aC(=O)OR a、-C 0-3亚烷基-NR aS(=O) 2R a、-C 0-3亚烷基-S(=O)R a、-C 0-3亚烷基-S(=O) 2R a、-C 0-3亚烷基-S(=O) 2N(R a) 2、-C 0-3亚烷基-SR a、-C 0-3亚烷基-S(R a) 5、-C 0-3亚烷基-C(=O)N(R a) 2、-C 0-3亚烷基-C(=O)R a、-C 0-3亚烷基-C(=O)OR a、C 2-6烯基、C 2-6炔基、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-C 0-3亚烷基-C 6-14芳基或-C 0-3亚烷基-(5-14元杂芳基),所述-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-C 0-3亚烷基-C 6-14芳基或-C 0-3亚烷基-(5-14元杂芳基)任选未取代或进一步被一个或多个R a取代基所取 代,且每个R a独立地选自H、卤素、C 1-6烷基、C 1-6卤代烷基、C 3-14环烷基、3-14元杂环烷基、C 2-3烯基、C 2-3炔基、芳基或杂芳基;
a或b独立地选自1、2、3或4;
Figure PCTCN2021090082-appb-000012
为双键或单键。
一些实施方式中,式(I)中的
Figure PCTCN2021090082-appb-000013
选自
Figure PCTCN2021090082-appb-000014
Figure PCTCN2021090082-appb-000015
一些实施方式中,式(I)中的R 2选自
Figure PCTCN2021090082-appb-000016
Figure PCTCN2021090082-appb-000017
一些实施方式中,式(I)中的A环选自
Figure PCTCN2021090082-appb-000018
Figure PCTCN2021090082-appb-000019
一些实施方式中,式(I)中的R 3选自H、卤素、氧代、-OR a、-C 0-3亚烷基-N(R a) 2、C 1-6卤代烷基,R a独立地为H、C 1-6烷基、C 1-6卤代烷基。
一些实施方式中,式(I)中的X 1选自-C(R 4) 2-(CH 2) 0-2-、-NH-(CH 2) 0-2-、-O-(CH 2) 0-2-、-C(O)-(CH 2) 0-2-、
Figure PCTCN2021090082-appb-000020
-CH=CH-或-N=CH-,R 4独立地选自H、D、卤素或C 1-3烷基。
一些实施方式中,式(I)中的X 2选自-C(R 5) 2-(CH 2) 0-2-、-NH-(CH 2) 0-2-、-O-(CH 2) 0-2-、-C(O)-(CH 2) 0-2-、
Figure PCTCN2021090082-appb-000021
-CH=CH-或-N=CH-,R 5独立地选自H、D、卤素或C 1-3烷基。
一些实施方式中,式(I)中的X 3的取代基选自C、CH或N。
一些实施方式中,式(I)中的X 4选自CR 6或N,且R 6的取代基选自卤素、C 1-3烷基、C 1-3卤代烷基或C 2-3烯基。
一些实施方式中,式(I)中的X 5选自CR 7或N,且R 7的取代基选自H或C 1-3烷基。
一些实施方式中,式(I)中的X 6选自CR 8或N,且R 8的取代基选自H、卤素或C 1-3烷基,优选X 6为N。
一些实施方式中,式(I)中的X 7选自NR 9或CHR 9,优选NR 9
一些实施方式中,式(I)中的R 9选自-C 0-3亚烷基-C 6-14芳基、-C 0-3亚烷基-(5-14元杂芳基)、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基),所述-C 0-3亚烷基-C 6-14芳基、-C 0-3亚烷基-(5-14元杂芳基)、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)未被取代或进一步被1-4个选自卤素、C 1-3烷基、C 1-3烯基、C 1-3卤代烷基、-(CH 2) 0-3-S(R a) 5、C 1-3烷氧基、C 3-6环烷基、C 1-3烷基取代的C 3-6环烷基、-C 0-3亚烷基-苯基、-C 0-3亚烷基-N(R a) 2的取代基所取代,且每个R a独立地选自H或C 1-6烷基;优选R 9选自
Figure PCTCN2021090082-appb-000022
Figure PCTCN2021090082-appb-000023
一些实施方式中,式(I)中的
Figure PCTCN2021090082-appb-000024
选自
Figure PCTCN2021090082-appb-000025
Figure PCTCN2021090082-appb-000026
Figure PCTCN2021090082-appb-000027
一些实施方式中,式(I)选自式(Ia)至式(Ie)化合物,
Figure PCTCN2021090082-appb-000028
其中,取代基R 1-R 9、a、b如式(I)所定义。
本发明还提供一种通式(II)所示的化合物、其互变异构体、氘代物或药用盐:
Figure PCTCN2021090082-appb-000029
Figure PCTCN2021090082-appb-000030
其中,
R 10选自C 1-6烷基、-C 0-3亚烷基-C 6-14芳基、-C 0-3亚烷基-(5-14元杂芳基)、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-O-C 0-3亚烷基-C 6-14芳基、-O-C 0-3亚烷基-(5-14元杂芳基)、-O-C 0-3亚烷基-C 3-14环烷基、-O-C 0-3亚烷基-(3-14元杂环烷基)、-NH-C 1-6烷基、-N(C 1-6烷基) 2、-NH-C 0-3亚烷基-C 6-14芳基、-NH-C 0-3亚烷基-(5-14元杂芳基)、-NH-C 0-3亚烷基-C 3-14环烷基、-NH-C 0-3亚烷基-(3-14元杂环烷基),R 10任选未取代或进一步被1-4个R 10a取代基所取代;优选R 10
Figure PCTCN2021090082-appb-000031
Figure PCTCN2021090082-appb-000032
R 11选自H、氨基、卤素、C 1-3氰基、C 1-3羟基烷基、C 1-6烷基;优选为甲基或乙氰基;
R 12为丙烯酰基或取代的丙烯酰基;优选为
Figure PCTCN2021090082-appb-000033
Figure PCTCN2021090082-appb-000034
R 13选自H、羟基、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、-NH-C 1-6烷基、-N(C 1-6烷基) 2、氰基或卤素;优选为H;
B环选自C 3-10环烷基、3-10元杂环基、C 6-12芳基、5-12元杂芳基;
X 8的取代基选自CR 15或N;优选为N、卤素或CH;
X 9选自C、CH或N;
X 10或X 11独立地选自-O-(CH 2) 0-2-或CH 2;
R 14、R 15或R 10a独立地选自氢、氧代、酰基、氰基、卤素、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-3-OR b、-(CH 2) 0-3-N(R b) 2、-(CH 2) 0-3-S(=O)R b、-(CH 2) 0-3-S(=O) 2R b、-(CH 2) 0-3-SR b、-(CH 2) 0-3-S(R a) 5、-(CH 2) 0-3-C(=O)N(R b) 2、C 2-3烯基、C 2-3炔基、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-C 0-3亚烷基-C 6-14芳基或-C 0-3亚烷基-(5-14元杂芳基),且每个R b独立地选自H、C 1-6烷基、C 1-6卤代烷基、C 3-14环烷基、3-14元杂环烷基、C 2-3烯基、C 2-3炔基、芳基或杂芳基;
c或d独立地选自1、2、3或4;
Figure PCTCN2021090082-appb-000035
为双键或单键。
一些实施方式中,式(II)选自式(IIa)至(IId)化合物,
Figure PCTCN2021090082-appb-000036
其中,取代基如式(II)所定义。
本发明还提供了一种药物组合物,其特征在于,所述药物组合物包含治疗有效量的至少一种式(I)或式(II)所示的化合物和至少一种药学上可接受的辅料。
本发明进一步提供了一种药物组合物,其特征在于,所述的治疗有效量的至少一种式(I)或式(II)所示的化合物和药学上可接受的辅料的质量百分比为0.0001:1-10。
本发明提供了结构式(I)或式(II)所示化合物或药物组合物在制备药物中的应用。
一些实施方式中,制备式(I)化合物的方法包括以下步骤:
Figure PCTCN2021090082-appb-000037
式(IA)化合物在碱性条件下与化合物X-R 2发生酰化反应,得到式(I)化合物、其互变异构体、氘代物或药用盐,其中,X为卤素;R 1-R 3、X 1-X 7、a或b如式(I)所定义。
本发明进一步提供了所述应用的优选技术方案:
作为优选,所述应用为制备治疗和/或预防癌症药物中的应用。
作为优选,所述应用为制备用于治疗由KRAS G12C介导的疾病的药物的应用。作为优选,所述疾病是癌症。
作为优选,所述癌症选自乳腺癌、多发性骨髓瘤、膀胱癌、子宫内膜癌、胃癌、宫颈癌、横纹肌肉瘤、非小细胞肺癌、小细胞肺癌、多形性肺癌、卵巢癌、食管癌、黑色素瘤、结肠直肠癌、肝细胞瘤、头颈部肿瘤、肝胆管细胞癌、骨髓增生异常综合征、恶性胶质瘤、前列腺癌、甲状腺癌、徐旺氏细胞瘤、肺鳞状细胞癌、苔藓样角化病、滑膜肉瘤、皮肤癌、胰腺癌、睾丸癌或脂肪肉瘤。
本发明还提供了一种治疗和/或预防疾病的方法,包括向治疗对象施用治疗有效量的至少任意一种结构式(I)或式(II)所示化合物或含其的药物组合物。
本发明还提供了一种治疗和/或预防由KRAS G12C介导的疾病的方法,包括向治疗对象施用治疗有效量的至少任意一种结构式(I)或式(II)所示化合物或含其的药物组合物。
本发明还提供了一种治疗癌症的方法,包括向治疗对象施用治疗有效量的至少任意一种结构式(I)或式(II)所示化合物或含其的药物组合物。
作为优选,在上述方法中,所述KRAS G12C介导的疾病是癌症。
作为优选,在上述方法中,所述的癌症选自乳腺癌、多发性骨髓瘤、膀胱癌、子宫内膜癌、胃癌、宫颈癌、横纹肌肉瘤、非小细胞肺癌、小细胞肺癌、多形性肺癌、卵巢癌、食管癌、黑色素瘤、结肠直肠癌、肝细胞瘤、头颈部肿瘤、肝胆管细胞癌、骨髓增生异常综合征、恶性胶质瘤、前列腺癌、甲状腺癌、徐旺氏细胞瘤、肺鳞状细胞癌、苔藓样角化病、滑膜肉瘤、皮肤癌、胰腺癌、睾丸癌或脂肪肉瘤。
除非另有说明,所述结构通式中使用的一般化学术语具有通常的含义。
例如,除非另有说明,本发明所用的术语“卤素”是指氟、氯、溴或碘。
在本发明中,除非另有说明,“烷基”包括直链或支链的一价饱和烃基。例如,烷基包括甲基、乙基、丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、3-(2-甲基)丁基、2-戊基、2-甲基丁基、新戊基、正己基、2-己基、2-甲基戊基等。类似的,“基 1-8烷基”中的“ 1-8”是指包含有1、2、3、4、5、6、7或8个碳原子的直链或支链形式排列的基团。
“C 1-3亚烷基”是指亚甲基、1,2-亚乙基、1,3-亚丙基或1,2-亚异丙基。
“烷氧基”是指前述的直链或支链烷基的氧醚形式,即-O-烷基。
在本发明中,“一”、“一个”、“该”、“至少一个”和“一个或多个”可互换使用。因此,例如,包含“一种”药学上可接受的赋形剂的组合物可以被解释为表示该组合物包括“一种或多种”药学上可接受的赋形剂。
术语“芳基”,在本发明中,除非另有说明,是指未取代或取代的包括碳环的原子的单环或稠环芳香基团。优选芳基为6到10元的单环或双环的芳香环基团。优选为苯基、萘基。最优选为苯基。所述芳基环可以稠合于杂芳基、杂环基或环烷基上,其中与母体结构连接在一起的环为芳基环,非限制性实例包括但不限于苯并环戊基。
术语“杂环基”,在本发明中,除非另有说明,是指由碳原子和1-3个选自N、O或S的杂原子组成的未取代或取代的3-8元稳定单环系统,其为饱和或部分不饱和单环或多环环状烃取代基,其包括3至20个碳原子,其中氮或硫杂原子可以选择性地被氧化,并且氮杂原子可以选择性地被季铵化。该杂环基可以被连接到任何的杂原子或碳原子上以形成稳定的结构。这些杂环基的实例包括但不限于氮杂环丁烷基、吡咯烷基、哌啶基、哌嗪基、氧代哌嗪基、氧代哌啶基、四氢呋喃基、二氧戊环基、四氢咪唑基、四氢噻唑基、四氢恶唑基、四氢吡喃基、吗啉基、硫代吗啉基、硫代吗啉基亚砜、硫代吗啉基砜基和四氢恶二唑基。
术语“杂芳基”,在本发明中,除非另有说明,是指未取代或取代的稳定的5元或6元单环芳族环系统或未取代或取代的9元或10元苯并稠合杂芳族环系统或双环杂芳族环系统,其由碳原子和1-4个选自N、O或S的杂原子组成,并且其中所述氮或硫杂原子可以选择性地被氧化,所述氮杂原子可以选择性地被季铵化。杂芳基可以连接在任何杂原子或碳原子上以形成稳定的结构。杂芳基的实例包括但不限于噻吩基、呋喃基、咪唑基、异恶唑基、恶唑基、吡唑基、吡咯基、噻唑基、噻二唑基、三唑基、吡啶基、哒嗪基、吲哚基、氮杂吲哚基、吲唑基、苯并咪唑基、苯并呋喃基、苯并噻吩基、苯并异恶唑基、苯并噻唑基、苯并噻唑基、苯并噻二唑基、苯并三唑基腺嘌呤、喹啉基或异喹啉基。所述杂芳基可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环。
术语“环烷基”是指具有3-10个碳原子的环状饱和或部分不饱和单环或多环环状烃取代基,例如,环丙基、环丁基、环戊基或环己基。
术语“取代的”是指基团中的一个或多个氢原子分别被相同的或者不同的取代基所取代。典型的取代基包括但不限于卤素(F、Cl、Br或I)、C 1-8烷基、C 3-12环烷基、-OR 1、-SR 1、=O、=S、-C(O)R 1、-C(S)R 1、=NR 1、-C(O)OR 1、-C(S)OR 1、-NR 1R 1、-C(O)NR 1R 1、氰基、硝基、-S(O) 2R 1、-O-S(O 2)OR 1、-O-S(O) 2R 1、-OP(O)(OR 1)(OR 2);其中R 1和R 2独立地选自-H、C 1-6烷基、C 1-6卤代烷基。在一些实施例中,取代基独立地选自包含-F、-Cl、-Br、-I、-OH、三氟甲氧基、乙氧基、丙氧基、异丙氧基、正丁氧基、异丁氧基、叔丁氧基、-SCH 3、-SC 2H 5、甲醛基、-C(OCH 3)、氰基、硝基、-CF 3、-OCF 3、氨基、二甲基氨基、甲硫基、磺酰基和乙酰基的基团。
取代烷基的实例包括但不限于2,3-二羟基丙基、2-氨基乙基、2-羟乙基、五氯乙基、三氟甲基、甲氧基甲基、五氟乙基、苯基甲基、二恶茂基甲基和哌嗪基甲基。
取代烷氧基的实例包括但不限于2-羟基乙氧基、2-氟乙氧基、2,2-二氟乙氧基、2-甲氧基乙氧基、2-氨基乙氧基、2,3-二羟基丙氧基、环丙基甲氧基、氨基甲氧基、三氟甲氧基、2-二乙基氨基乙氧基、2-乙氧基羰基乙氧基、3-羟基丙氧基。
术语“药学上可接受的盐”是指从药学上可接受的无毒的碱或酸制备的盐。当本发明提供的化合物是酸时,可以从药学上可接受的无毒的碱,包括无机碱和有机碱,方便地制得其相应的盐。从无机碱衍生的盐包括铝、铵、钙、铜(高价和低价)、三价铁、亚铁、锂、镁、锰(高价和低价)、钾、钠、锌之类的盐。特别优选铵、钙、镁、钾和钠的盐。药学上可接受的能够衍生成盐的无毒有机碱包括伯胺、仲胺和叔胺,也包括环胺及含有取代基的胺,如天然存在的和合成的含取代基的胺。能够成盐的其他药学上可接受的无毒有机碱,包括离子交换树脂以及精氨酸、甜菜碱、咖啡因、胆碱、N',N'-二苄乙二胺、二乙胺、2-二乙氨基乙醇、2-二甲胺基乙醇、乙醇胺、乙二胺、N-乙基吗啉、N-乙基哌啶、还原葡萄糖胺、氨基葡萄糖、组氨酸、哈胺、异丙胺、赖氨酸,甲基葡萄糖胺、吗啉、哌嗪、哌啶、多胺树脂、普鲁卡因、嘌呤、可可碱、三乙胺、三甲胺、三丙胺、氨丁三醇等。
当本发明提供的化合物是碱时,可以从药学上可接受的无毒的酸,包括无机酸和有机酸,方便制得其相应的盐。这样的酸包括,如,醋酸、苯磺酸、苯甲酸、樟脑磺酸、柠檬酸、乙磺酸、羟乙基磺酸、甲酸、富马酸、葡萄糖酸、谷氨酸、氢溴酸、氢碘酸、高氯酸、盐酸、羟乙磺酸、丙酸、乙醇酸、乳酸、马来酸、苹果酸、扁桃酸、甲磺酸、黏酸、硝酸、草酸、扑酸、泛酸、磷酸、琥珀酸、硫酸、2-萘磺酸、环己胺磺酸、水杨酸、糖精酸、三 氟乙酸、酒石酸和对甲苯磺酸等。较优地,柠檬酸、氢溴酸、甲酸、盐酸、马来酸、磷酸、硫酸和酒石酸。更优地,甲酸和盐酸。
由于式(I)或式(II)所示化合物将作为药物应用,较优地,使用一定纯度,例如,至少为60%纯度,比较合适的纯度为至少75%,特别合适地纯度为至少98%(%是重量比)。
本发明化合物的药物前体包含在本发明的保护范围内。通常,所述药物前体是指很容易在体内转化成所需化合物的功能性衍生物。例如,本申请化合物的任何药学上可接受的盐、酯、酯的盐或其它衍生物,其在向受体施用后能够直接或间接地提供本申请的化合物或其具有药学活性的代谢物或残基。
本发明所述化合物可能含有一个或多个不对称中心,并可能由此产生非对映异构体和光学异构体。本发明包括所有可能的非对映异构体及其外消旋混合物、其基本上纯的拆分对映异构体、所有可能的几何异构体及其药学上可接受的盐。
当式(I)或式(II)所示化合物用较重的同位素(例如氘)替代可能提供某些治疗优势,这是由于更大的代谢稳定性,例如增加体内半衰期或减少剂量要求。
当式(I)或式(II)所示化合物及其药学上可接受的盐存在溶剂化物或多晶型时,本发明包括任何可能的溶剂化物和多晶型。形成溶剂化物的溶剂类型没有特别的限定,只要该溶剂是药学上可以接受的。例如,水、乙醇、丙醇、丙酮等类似的溶剂都可以采用。
术语“组合物”,在本发明中,是指包括包含指定量的各指定成分的产品,以及直接或间接地由指定量的各指定成分的组合生产的任何产品。因此,含有本发明的化合物作为活性成分的药物组合物以及制备本发明化合物的方法也是本发明的一部分。此外,化合物的一些结晶形式可以多晶型存在,并且此多晶型包括在本发明中。另外,一些化合物可以与水(即水合物)或常见的有机溶剂形成溶剂化物,并且此类溶剂化物也落入本发明的范围内。
本发明提供的药物组合物包括作为活性组分的式(I)或式(II)所示化合物(或其药学上可接受的盐)、一种药学上可接受的赋形剂及其他可选的治疗组分或辅料。尽管任何给定的情况下,最适合的活性组分给药方式取决于接受给药的特定的主体、主体性质和病情严重程度,但是本发明的药物组合物包括适于口腔、直肠、局部和不经肠道(包括皮下给药、肌肉注射、静脉给药)给药的药物组合物。本发明的药物组合物可以方便地以本领域公知的单位剂型存在和药学领域公知的任何制备方法制备。
一般情况下,治疗上述所示的状况或不适,药物的剂量水平约为每天0.01mg/kg体重到150mg/kg体重,或者每个病人每天0.5mg到7g。例如,炎症、癌症、牛皮癣、过敏/哮 喘、免疫系统的疾病和不适、中枢神经系统(CNS)的疾病和不适,有效治疗的药物剂量水平为每天0.01mg/kg体重到50mg/kg体重,或者每个病人每天0.5mg到3.5g。
但是,可以理解,可能需要比上述那些更低或更高的剂量。任何特定病人的具体剂量水平和治疗方案将取决于多种因素,包括所用具体化合物的活性、年龄、体重、综合健康状况、性别、饮食、给药时间、给药途径、排泄率、药物联用的情况和接受治疗的特定疾病的严重程度。
合成方案
合成路线1
Figure PCTCN2021090082-appb-000038
步骤A:化合物I-1甲酯化得到化合物I-2;
步骤B:化合物I-2与PMBNH 2经过取代反应得到化合物I-3;
步骤C:化合物I-3在Pd/C作用下脱去PMB保护基;
步骤D:化合物I-4在CuBr 2作用下,将氨基转化为溴;
步骤E:化合物I-5在碱性条件如LiOH等作用下水解,得到化合物I-6;
步骤F:化合物I-6在CDI作用下,将羧基转化为酰胺;
步骤G:化合物I-7在草酰氯的作用下,引入R 9基团;
步骤H:化合物I-8在强碱如NaH等作用下自身关环得到化合物I-9;
步骤I:三氯氧磷高温搅拌将I-9的-OH转化为-Cl得到化合物I-10;
步骤J:化合物I-10与
Figure PCTCN2021090082-appb-000039
在碱性条件下如DIEA等经过取代反应的到化合物I-11;
步骤K:I-11与
Figure PCTCN2021090082-appb-000040
在Pd催化剂如Pd(dppf)Cl 2作用下经过Suzuki偶联得到引入A环,得到化合物I-12,n选自1、2、3或4;
步骤L:I-12在碱性条件下如Cs 2CO 3作用下自身关环;
步骤M:酸性条件下如三氟乙酸等,脱去Boc保护基;
步骤N:在弱碱性条件下如碳酸氢钠等,引入R 2基团,即可得到目标化合物I。
合成路线2
Figure PCTCN2021090082-appb-000041
步骤O:通过类似I-12的步骤合成II-1,II-1在碱性条件下如Cs 2CO 3作用下自身关环;
步骤P:酸性条件下如三氟乙酸等,脱去Boc保护基;
步骤Q:在弱碱性条件下如碳酸氢钠等,引入R 12基团,即可得到目标化合物I。
具体实施方式
为使上述内容更清楚、明确,本发明将用以下实施例来进一步阐述本发明的技术方案。以下实施例仅用于说明本发明的具体实施方式,以使本领域的技术人员能够理解本发明,但不用于限制本发明的保护范围。本发明的具体实施方式中,未作特别说明的技术手段或方法等为本领域的常规技术手段或方法等。
除非另有说明,本发明所有的一部分和百分比均按重量计算,所有温度均指摄氏度。
实施例中使用了下列缩略语:
CDI:羰基二咪唑;
DCM:二氯甲烷;
DIPEA:二异丙基乙胺;
DME:二甲醚;
DMF:N,N-二甲基甲酰胺;
DMSO:二甲亚砜;
EA:乙酸乙酯;
MeOH:甲醇;
PE:石油醚;
Pd/C:钯碳:
THF:四氢呋喃;
TFA:三氟乙酸;
pre-TLC:制备薄层层析硅胶板;
pre-HPLC:制备高效液相色谱;
PMBNH 2:4-甲氧基苄氨;
SOCl 2:二氯亚砜;
CDI:N,N'-羰基二咪唑
中间体M-11的合成
Figure PCTCN2021090082-appb-000042
步骤1:化合物M-2的合成
把商业可得的原料M-1(11.64g,60mmol)溶解在甲醇中(80mL),然后滴加入二氯亚砜(21.41g,180.00mmol,13.05mL);滴毕,保温55℃反应过夜。TLC确定原料转化完全。浓缩除去甲醇。加入乙酸乙酯120mL,再用饱和氯化钠水溶液40mL×2洗涤、分液。加入无水硫酸钠干燥。过滤,除干燥剂,浓缩至恒重,得到无色液体M-2(10.1g,48.53mmol,81%yield)。
步骤2:化合物M-3的合成
把化合物M-2(10.1g,48.53mmol)和4-甲氧基苄氨(9.99g,72.80mmol)溶解在DMSO(50mL),加入二异丙基乙基胺(25.09g,194.13mmol,33.81mL),加热至60℃,搅拌反应。搅拌反应3.5小时LC-MS原料转化完全。反应混合物冷却至室温后,加入到250g的冰水混合物中,搅拌10分钟。加入乙酸乙酯200mL×2萃取、分液;合并有机相。加入饱和氯化钠水溶液120mL×2,洗涤、分液。无水硫酸钠干燥,除干燥剂,浓缩得到白色固体15.82g,为产品粗品。用PE/EA=8/1混合物150mL,将粗品打浆;最终得到白色固体M-3(12.0g,36.52mmol,75%yield)。
步骤3:化合物M-4的合成
把上面得到的白色固体(12.0g,36.89mmol)溶解在甲醇(300mL),氮气置换,然后加入10%钯碳(1200mg),再氢气置换,保持氢气氛围,常温搅拌反应。搅拌反应4.5小时;LC-MS:原料转化完全。硅藻土助滤,除去钯碳,并用甲醇少许漂洗滤渣。浓缩滤液蒸干,加入二氯甲烷溶解,无水硫酸钠干燥;再次浓缩蒸干,得到类白色残留物。加入 PE/EA=30:1混合物180mL,将浓缩所得类白色残留物打浆。过滤,收集滤饼,抽干、称重:类白色固体粉末M-4被得到(7.24g,35.29mmol,96%yield)。ESI-MS m/z:206.04[M+H] +
步骤4:化合物M-5的合成
将化合物M-4(7.24g,35.29mmol)溶解在MeCN(80mL)。氮气保护下,开启搅拌,冷却至0℃,然后加入CuBr 2(11.82g,52.94mmol);搅拌2分钟,然后滴加入亚硝酸叔丁酯(5.46g,52.94mmol),搅拌10分钟;撤去冷浴,自然升室温,搅拌反应1小时。TLC分析,原料转换完全;可以进行后处理。反应混合物加入到400g冰水混合物中,然后用乙酸乙酯400mL萃取、分液(饱和氯化钠水溶液少许促进分层)。饱和氯化铵水溶液150mL×2洗涤有机相、分液。加入无水硫酸钠适量干燥有机相。过滤除干燥剂,浓缩得到黄棕色粘稠物12.5g。柱层析分离纯化(PE/EA=97/3),最终得到白色固体M-5(9.1g,33.83mmol,96%yield)。ESI-MS m/z:270.11[M+H] +
步骤5:化合物M-6的合成
把上面得到的白色固体M-5(6.0g,22.30mmol)加入到反应瓶中,然后加入甲醇50mL,然后室温下加入含有LiOH(587mg)的水溶液20mL搅拌反应,2小时反应结束。旋去大部分的甲醇,然后调pH=3,EA(100mL x 3)萃取,得到有机相后,加入无水硫酸钠适量干燥有机相。过滤除干燥剂,浓缩得到白色固体(5.6g,98%yield)。ESI-MS m/z:253.11[M-H] -
步骤6:化合物M-7的合成
把底物M-6(5.5g,21.57mmol)溶解在THF(80mL)中,然后加入DIEA(8.36g,64.71mmol,11.27mL),然后加入羰基二咪唑(4.19g,25.88mmol),加热到50℃下进行反应。检测反应。直到原料全部消失。然后冷却到室温,待用。把氨水120mL冷却到0℃以下,然后把上面的反应液,慢慢加入其中,加完后,自然升到室温,反应。检测反应,直到原料全部反应完全。加入EA(120mL)稀释后饱和食盐水洗两次,然后旋干,柱层析分离(PE/EA=2:1),得到白色固体(5.32g,20.94mmol,97%yield)。ESI-MS m/z:253.96[M+H] +
步骤7:化合物M-8的合成
把底物M-7(4.61g,18.15mmol)溶解在THF(45mL)中,然后氮气保护下,加入草酰氯(9.21g,72.60mmol,6.14mL),加热回流反应1hr,减压旋干,用DCM洗,再次浓缩,然后加入溶解有2-异丙基-4-甲基-3-吡啶胺(4.09g,27.22mmol)的DCM(40mL)溶液,室温下 搅拌反应1hr。反应结束后往体系中加入碳酸氢钠水溶液,然后加入EA(200mL)萃取,得到有机相后,干燥浓缩,柱层析分离(PE/EA=1:1),得到白色固体M-8(6.7g,15.57mmol,86%yield)。ESI-MS m/z:430.146[M+H] +
步骤8:化合物M-9的合成
把底物M-8(4.8g,11.16mmol)溶解在DME(60mL)中,然后室温下慢慢加入NaH(535.5mg),等到体系不再冒泡后,微波加热到90℃下反应10分钟,反应结束后,加入饱和碳酸氢钠溶液淬灭反应,然后加入EA(200mL),饱和食盐水洗有机相,得到有机相后,干燥浓缩,柱层析分离(PE/EA=1:1),得到白色固体M-9(3.96g,9.65mmol,86%yield)。ESI-MS m/z:410.14[M+H] +
步骤9:化合物M-10的合成
把底物M-10(2.1g,5.12mmol)溶解在MeCN(20mL)中,然后加入DIPEA(6.62g,51.19mmol,8.92mL),然后加入POCl 3(3.92g,25.60mmol,2.45mL),加热到60℃下反应。LCMS检测反应到原料全部消失。反应结束后,恢复室温后,减压浓缩,得到淡黄色油状物,用DCM带两次后,再次旋干,得到油状物直接进行下一步反应。ESI-MS m/z:428.14[M+H] +
步骤10:化合物M-11的合成
把上一步得到的化合物M-10溶解在10mL的DMF中,然后加入DIPEA(1.32g,10.22mmol,1.78mL),然后加入溶解在DMF(10mL)中的(3S)-3-甲基哌嗪-1-羧酸叔丁酯(1.53g,7.66mmol),室温下搅拌反应,等到底物完全反应后,然后加入EA(200mL),饱和食盐水洗有机相,得到有机相后,干燥浓缩,柱层析分离(PE/EA=1:1),得到白色固体M-11(2.9g,4.89mmol,96%yield)。ESI-MS m/z:592.14[M+H] +
实施例1:化合物1((S)-1-(4-丙烯酰基-2-甲基哌嗪-1-基)-11-氟-4-(2-异丙基-4-甲基吡啶-3-基)-4H-异铬烯桥[4,3-h]喹唑啉-3(6H)-酮)的合成
Figure PCTCN2021090082-appb-000043
步骤1:化合物1-1的合成
将化合物M-11(400mg,0.68mmol)加入到反应瓶中,然后加入硼酸(205mg,1.35mmol)和K 3PO 4(429.9mg,2.03mmol),往反应瓶中加入CPME和纯净水,然后用N 2鼓起排体系中的氧气。5分钟后,把催化剂Pd(dppf)Cl 2(74mg,0.1mmol)加入到反应瓶中,然后封好瓶塞,微波下反应(90℃,40min),反应结束后,冷却到室后,加入EA稀释反应体系,然后饱和碳酸氢钠洗有机相,饱和食盐水洗后,有机相干燥,旋干,直接进行下一步反应。
步骤2:化合物1-2的合成
把上面得到的化合物1-1(410mg,0.66mmol),溶解在DMF中,然后加入Cs 2CO 3(431mg,1.32mmol),微波下加热到125℃反应30分钟,反应结束后冷却到室温,然后加入EA稀释,饱和碳酸氢钠洗后,饱和食盐水洗,然后有机相干燥,浓缩,pre-TLC(EA)制备分离,得到白色泡沫状固体(169mg,42%)。ESI-MS m/z:600.04[M+H] +
步骤3:化合物1-3的合成
把上面得到的化合物1-2(169mg,0.28mmol)溶解在DCM(4mL),然后室温下加入CF 3COOH(1mL),保持室温反应30分钟,反应结束后,加入饱和碳酸氢钠水溶液淬灭反应,然后加入EA萃取水相,合并有机相后,干燥,浓缩后得到,直接用于下一步反应。
步骤4:化合物1的合成
把上面得到的化合物加入到干燥的DCM(4mL)中,热后冷却到0℃,往体系中慢慢加入稀释后的丙烯酰氯(25mg,0.26mmol),10分钟后,TLC显示底物反应完全,加入饱和碳酸氢钠淬灭反应,然后加入EA稀释反应体系,水相,饱和食盐水洗后,有机相干燥,浓缩后,pre-TLC制备纯化,得到白色固体化合物1(141mg,98%)。ESI-MS m/z: 554.33.[M+H] +1H NMR(500MHz,CDCl 3)δ8.49(d,J=4.9Hz,1H),8.02(d,J=7.7Hz,1H),7.41(t,J=7.7Hz,1H),7.36(t,J=7.4Hz,1H),7.18(d,J=11.9Hz,1H),7.05(d,J=7.7Hz,2H),6.60(dd,J=32.2,17.0Hz,1H),6.39(d,J=16.8Hz,1H),5.80(d,J=10.4Hz,1H),4.73(d,J=16.0Hz,1H),4.57-4.37(m,2H),2.16-2.06(m,3H),1.56-1.44(m,2H),1.41(d,J=6.7Hz,2H),1.21(t,J=6.3Hz,3H),1.12(d,J=6.8Hz,3H)。
实施例2:化合物2((S)-8-(4-丙烯酰基-2-甲基哌嗪-1-基)-12-氟-11-(2-异丙基-4-甲基吡啶-3-基)-5H-异色烯[3,4-g]喹唑啉-10(11H)-酮)的合成
Figure PCTCN2021090082-appb-000044
步骤1:化合物2-1的合成
把上面得到的化合物1-1(410mg,0.66mmol),溶解在DMF中,然后加入Cs 2CO 3(431mg,1.32mmol),微波下加热到125℃反应30分钟,反应结束后冷却到室温,然后加入EA稀释,饱和碳酸氢钠洗后,饱和食盐水洗,然后有机相干燥,浓缩,pre-TLC(EA)制备分离,得到白色泡沫状固体(170mg,42%)。ESI-MS m/z:600.04[M+H] +
步骤2:化合物2-2的合成
把上面得到的化合物2-1(169mg,0.28mmol)溶解在DCM(4mL),然后室温下加入CF 3COOH(1mL),保持室温反应30分钟,反应结束后,加入饱和碳酸氢钠水溶液淬灭反应,然后加入EA萃取水相,合并有机相后,干燥,浓缩后得到,直接用于下一步反应。
步骤3:化合物2的合成
把上面得到的化合物加入到干燥的DCM(4mL)中,热后冷却到0℃,往体系中慢慢加入稀释后的丙烯酰氯(25mg,0.26mmol),10分钟后,TLC显示底物反应完全,加入饱和碳酸氢钠淬灭反应,然后加入EA稀释反应体系,水相,饱和食盐水洗后,有机相干 燥,浓缩后,pre-TLC制备纯化,得到白色固体化合物2(115mg,84%)。ESI-MS m/z:554.33[M+H] +1H NMR(500MHz,CDCl 3)δ8.55(d,J=4.9Hz,1H),7.73(d,J=7.7Hz,1H),7.38(t,J=7.3Hz,1H),7.34(t,J=7.6Hz,1H),7.25-7.17(m,2H),7.11(d,J=4.9Hz,1H),6.60(dd,J=35.6,13.4Hz,1H),6.39(d,J=16.7Hz,1H),5.79(d,J=10.5Hz,1H),5.17-5.05(m,2H),2.15(d,J=6.5Hz,3H),1.25(dd,J=6.9,4.0Hz,4H),1.17(d,J=7.0Hz,4H).
实施例3:化合物3((S)-1-(丙烯酰基-2-甲基哌嗪-1-基)-10,11-二氟-4-(2-异丙基-4-甲基吡啶-3-基)-4H-异色烯[4,3-h]喹唑啉-3(6H)-酮)的合成
Figure PCTCN2021090082-appb-000045
步骤1:化合物3-1的合成
将化合物M-11(603mg,1.02mmol)加入到反应瓶中,然后加入硼酸(345.9mg,2.04mmol)和K 3PO 4(1.3g,6.11mmol),往反应瓶中加入CPME和纯净水,然后用N 2鼓起排体系中的氧气。5分钟后,把催化剂SPhos Pd G2(147mg,0.2mmol)加入到反应瓶中,然后封好瓶塞,微波下反应(90℃,40min),反应结束后,冷却到室后,加入EA稀释反应体系,然后饱和碳酸氢钠洗有机相,饱和食盐水洗后,有机相干燥,旋干,直接进行下一步反应。ESI-MS m/z:638.44[M+H] +.
步骤2:化合物3-2的合成
把上面得到的化合物3-1(535mg,0.84mmol),溶解在DMF中,然后加入Cs 2CO 3(820mg,2.52mmol),微波下加热到125℃反应30分钟,反应结束后冷却到室温,然后加入EA稀释,饱和碳酸氢钠洗后,饱和食盐水洗,然后有机相干燥,浓缩,pre-TLC(EA)制备分离,得到白色泡沫状固体(360mg,67%)。ESI-MS m/z:618.37[M+H] +
步骤3:化合物3-3的合成
把上面得到的化合物3-2(349mg,0.56mmol)溶解在DCM(6mL),然后室温下 加入CF 3COOH(2mL),保持室温反应30分钟,反应结束后,加入饱和碳酸氢钠水溶液淬灭反应,然后加入EA萃取水相,合并有机相后,干燥,浓缩后得到,直接用于下一步反应。
步骤4:化合物3的合成
把上面得到的化合物加入到干燥的DCM(4mL)中,热后冷却到0℃,往体系中慢慢加入稀释后的丙烯酰氯(51mg,0.56mmol),10分钟后,TLC显示底物反应完全,加入饱和碳酸氢钠淬灭反应,然后加入EA稀释反应体系,水相,饱和食盐水洗后,有机相干燥,浓缩后,pre-TLC制备纯化,得到白色固体化合物3(305mg,94%)。ESI-MS m/z:572.3[M+H] +1H NMR(500MHz,CDCl 3)δ8.49(d,J=4.9Hz,1H),7.38(td,J=7.9,4.8Hz,1H),7.24-7.13(m,2H),7.07(t,J=4.2Hz,1H),6.92(d,J=7.4Hz,1H),6.62(dt,J=38.5,14.2Hz,1H),6.40(d,J=16.8Hz,1H),5.80(d,J=10.5Hz,1H),4.52-4.38(m,3H),2.11(d,J=11.8Hz,3H),1.41(d,J=6.2Hz,2H),1.22(t,J=7.1Hz,3H),1.11(t,J=7.2Hz,3H).
实施例4:化合物4((S)-8-(丙烯酰基-2-甲基哌嗪-1-基)-1,12-二氟-11-(2-异丙基-4-甲基吡啶-3-基)-5H-异色烯[3,4-g]喹唑啉-10(11H)-酮)的合成
Figure PCTCN2021090082-appb-000046
步骤1:化合物4-1的合成
把上面得到的化合物3-1(535mg,0.84mmol),溶解在DMF中,然后加入Cs 2CO 3(820mg,2.52mmol),微波下加热到125℃反应30分钟,反应结束后冷却到室温,然后加入EA稀释,饱和碳酸氢钠洗后,饱和食盐水洗,然后有机相干燥,浓缩,pre-TLC(EA)制备分离,得到白色泡沫状固体(170mg,33%)。ESI-MS m/z:618.37[M+H] +
步骤2:化合物4-2的合成
把上面得到的化合物4-1(170mg,0.28mmol)溶解在DCM(6mL),然后室温下 加入CF 3COOH(2mL),保持室温反应30分钟,反应结束后,加入饱和碳酸氢钠水溶液淬灭反应,然后加入EA萃取水相,合并有机相后,干燥,浓缩后得到,直接用于下一步反应。
步骤3:化合物4的合成
把上面得到的化合物加入到干燥的DCM(4mL)中,热后冷却到0℃,往体系中慢慢加入稀释后的丙烯酰氯(25mg,0.28mmol),10分钟后,TLC显示底物反应完全,加入饱和碳酸氢钠淬灭反应,然后加入EA稀释反应体系,水相,饱和食盐水洗后,有机相干燥,浓缩后,pre-TLC制备纯化,得到白色固体化合物4(110mg,68%)。ESI-MS m/z:572.3[M+H] +1H NMR(500MHz,CDCl 3)δ8.54(d,J=4.9Hz,1H),7.40(td,J=7.9,4.8Hz,1H),7.24(s,1H),7.09(t,J=6.3Hz,3H),6.61(ddd,J=39.8,14.7,6.6Hz,1H),6.39(d,J=16.8Hz,1H),5.79(d,J=10.5Hz,1H),2.13(d,J=6.9Hz,3H),1.57-1.44(m,2H),1.39(d,J=6.7Hz,1H),1.25(t,J=5.9Hz,3H),1.18(q,J=6.8,6.4Hz,4H).
实施例5:化合物5((S)-1-(丙烯酰基-2-甲基哌嗪-1-基)-10-氯-11-氟-4-(2-异丙基-4-甲基吡啶-3-基)-4H-异色烯[4,3-h]喹唑啉-3(6H)-酮)的合成
Figure PCTCN2021090082-appb-000047
化合物5具体合成步骤参照实施例3。ESI-MS m/z:588.3[M+H] +1H NMR(500MHz,CDCl 3)δ8.49(d,J=4.9Hz,1H),7.49(d,J=8.0Hz,1H),7.31(t,J=7.8Hz,1H),7.21(d,J=10.0Hz,1H),7.05(dd,J=9.3,5.9Hz,2H),6.62(dt,J=37.2,13.9Hz,1H),6.40(d,J=16.8Hz,1H),5.80(d,J=10.4Hz,1H),4.50-4.35(m,2H),2.11(d,J=10.5Hz,3H).
实施例6:化合物6((S)-8-(丙烯酰基-2-甲基哌嗪-1-基)-1-氯-12-氟-11-(2-异丙基-4-甲基吡啶-3-基)-5H-异色烯[3,4-g]喹唑啉-10(11H)-酮)的合成
Figure PCTCN2021090082-appb-000048
化合物6具体合成步骤参照实施例4。ESI-MS m/z:588.3[M+H] +1H NMR(500MHz,CDCl 3)δ8.53(d,J=4.9Hz,1H),7.43(d,J=8.1Hz,1H),7.33(t,J=7.8Hz,1H),7.20(d,J=7.4Hz,1H),7.09(d,J=4.9Hz,1H),6.59(dd,J=31.6,16.4Hz,1H),6.39(d,J=16.8Hz,1H),5.79(d,J=10.5Hz,1H)。
实施例7:化合物7(10-((S)-4-(丙烯酰基-2-甲基哌嗪-1-基)-8-氟-13-(2-异丙基-4-甲基吡啶-3-基)-2,3-二氢苯并[4,5]氧地平[3,2-h]喹啉-12(13H)酮)的合成
Figure PCTCN2021090082-appb-000049
化合物7具体合成步骤参照实施例3。ESI-MS m/z:568.3[M+H] +1H NMR(500MHz,CDCl 3)δ8.47(d,J=4.9Hz,1H),7.54(t,J=5.7Hz,1H),7.44-7.30(m,3H),7.22(d,J=7.2Hz,1H),7.08(d,J=4.9Hz,1H),6.61(dd,J=38.6,12.8Hz,1H),6.40(d,J=16.7Hz,1H),5.80(d,J=10.5Hz,1H),2.18(d,J=3.5Hz,3H),1.23-1.04(m,6H)。
实施例8:化合物8(9-((S)-4-(丙烯酰基-2-甲基哌嗪-1-基)-13-氟-12-(2-异丙基-4-甲基吡啶-3-基)-5,6-二氢苯并[4,5]氧地平[2,3-h]喹啉-11(12H)酮)的合成
Figure PCTCN2021090082-appb-000050
化合物8具体合成步骤参照实施例4。ESI-MS m/z:568.3[M+H] +1H NMR(500MHz,CDCl 3)δ8.50(d,J=4.9Hz,1H),7.34(dq,J=22.7,7.8Hz,5H),7.07(d,J=4.9Hz,1H),6.73-6.51(m,1H),6.40(d,J=16.8Hz,1H),5.80(d,J=10.5Hz,1H)。
实施例9:化合物9((S)-1-(丙烯酰基-2-甲基哌嗪-1-基)-11-氟-10-羟基-4-(2-异丙基-4-甲基吡啶-3-基)-4H-异色烯[4,3-h]喹唑啉-3(6H)-酮)的合成
Figure PCTCN2021090082-appb-000051
步骤1:化合物9-4的合成
参照实施例3合成化合物9-3。把9-3(100mg,0.161mmol)溶解在DCM(5mL)中,然后冷却到0℃后,慢慢加入BBr 3(404mg,1.61mmol),维持在该温度下反应1小时,液质检测反应直到原料消耗完全,然后在该温度下,加入饱和碳酸氢钠水溶液淬灭反应,然后加入DCM稀释该反应后,水洗,有机相干燥后,旋干,直接进行下一步反应。(96mg,98%yield)。ESI-MS m/z:606.33[M+H] +
步骤2:化合物9的合成
把上面得到的化合物溶解在THF(3mL)中,然后加入6N的NaOH溶液0.2mL,室温下搅拌反应30分钟,底物反应完全,然后饱和碳酸氢钠中和体系中的强碱后,加入DCM萃取两次,然后合并有机相后,干燥,旋干,HPLC制备分离得到目标产物9(54mg,59%yield)。ESI-MS m/z:570.3[M+H] +1H NMR(500MHz,CDCl 3)δ8.52(d,J=5.0Hz,1H),7.22(q,J=11.5,9.7Hz,2H),7.12(d,J=5.0Hz,1H),7.02(d,J=8.2Hz,1H),6.64(d,J=7.4Hz,1H),6.43(d,J=16.7Hz,1H),5.83(d,J=10.5Hz,1H),5.30(s,2H),2.13(d,J=9.6Hz,3H),1.24(t,J=7.6Hz,3H),1.12(d,J=6.9Hz,3H).
实施例10:化合物10((S)-8-(丙烯酰基-2-甲基哌嗪-1-基)-12-氟-1-羟基-11-(2-异丙基-4-甲基吡啶-3-基)-5H-异色烯[3,4-g]喹唑啉-10(11H)-酮)的合成
Figure PCTCN2021090082-appb-000052
化合物10具体的合成步骤参照实施例9。ESI-MS m/z:570.3[M+H] +
实施例11:化合物11((S)-1-(丙烯酰基-2-甲基哌嗪-1-基)-11-氟-10-甲氧基-4-(2-异丙基-4- 甲基吡啶-3-基)-4H-异色烯[4,3-h]喹唑啉-3(6H)-酮)的合成
Figure PCTCN2021090082-appb-000053
化合物11具体的合成步骤参照实施例9。ESI-MS m/z:584.4[M+H] +
实施例12:化合物12((S)-8-(丙烯酰基-2-甲基哌嗪-1-基)-12-氟-1-甲氧基-11-(2-异丙基-4-甲基吡啶-3-基)-5H-异色烯[3,4-g]喹唑啉-10(11H)-酮)的合成
Figure PCTCN2021090082-appb-000054
化合物12具体的合成步骤参照实施例9。ESI-MS m/z:584.4[M+H] +
实施例13:化合物(S)-1-(4-丙烯酰基-2-甲基哌嗪-1-基)-11-氯-10-氟-4-(2-异丙基-4-甲基吡啶-3-基)-4H-异色烯[4,3-h]喹唑啉-3(6H)-酮的合成
Figure PCTCN2021090082-appb-000055
步骤1:化合物13-1的合成
在室温下,将化合物2-氨基-4-溴-3-氟苯甲酸(10g,42.73mmol)溶于DMF(30mL)中,加入NCS(6.85g,51.28mmol),70℃下反应3hs。反应液冷却至室温,加入150ml冰水中,析出浅黄色固体,过滤,滤饼用冰水洗(2*20ml),滤饼真空干燥(-0.1Mpa,50℃,4h)。得到黄色固体状化合物(11.2g,41.72mmol,97.63%yield)。ESI-MS m/z:267.9[M+H] +
步骤2:化合物13-2的合成
在室温下,将化合物13-1(11.2g,41.72mmol)溶解在MeCN(146mL)中,氮气保护下,开启搅拌,然后加入溴化铜(11.18g,50.06mmol),冰水浴降温,冷却至0℃,然后滴加入亚硝酸叔丁酯(6.45g,62.58mmol)的MeCN(52mL)溶液;滴毕,保温0-5℃搅拌30分钟,然后撤去冷浴,自然升至室温,搅拌反应3hs。反应液加入到600mL冷的饱和氯化铵水溶液中,加入乙酸乙酯500mL,分层,分出有机相,水相再用乙酸乙酯200mL*1萃取,合并有机相,饱和氯化钠溶液2*300mL洗涤,有机相无水硫酸钠干燥,过滤,滤液减压浓缩。得到浅棕色固体(12.19g,36.68mmol,87.92%yield),直接用于下一步。ESI-MS m/z:330.8[M+H] +
步骤3:化合物13-3的合成
在室温下,将化合物13-2(12.19g,36.68mmol)溶解在DCM(122mL)中,并滴加入4滴DMF;然后冰水浴冷却,滴加草酰氯(13.97g,110.03mmol,9.31mL)的DCM(14mL);然后搅拌反应半小时,反应液减压浓缩,并用DCM 20mL×2带2次;将氢氧化铵(15M,684.66mL)用冰水浴冷却;滴加中间残留物的干燥THF(60mL),滴毕,自然升室温反应1hs。加入乙酸乙酯300mL,饱和氯化钠水溶液300mL,分层,分出有机相;水相再用乙酸乙酯150mL萃取,分液;合并有机相,再用饱和氯化钠水溶液2*100mL洗涤;加入无水硫酸钠干燥;过滤,滤液减压浓缩。得到棕色固体(10.05g,30.33mmol,82.69%yield)。ESI-MS m/z:329.8[M+H] +
步骤4:化合物13-4的合成
在室温下,将化合物13-3(9.6g,28.97mmol)溶解在THF(192mL),氮气保护下,加入草酰氯(14.71g,115.88mmol,9.81mL)加热至70℃回流反应2hs;减压浓缩除去THF及草酰氯,并用无水DCM(15mL×2)浓缩蒸干。浓缩残留物用DCM(20ml)溶解,滴加含有2-异丙基-4-甲基吡啶-3-胺(4.35g,28.97mmol)的DCM(160mL)溶液,然后室温搅拌反应2小时。反应混合物用DCM(100mL)稀释后,依次用饱和氯化铵水溶液(100mL),饱和碳酸氢钠水溶液(100mL)洗涤、分液;再用饱和氯化钠水溶液100mL洗涤、分液;无水硫酸钠干燥,过滤,滤液减压浓缩,得到棕色固体化合物(13.3g,26.20mmol,90.44% yield),直接投下步。ESI-MS m/z:505.9[M+H] +
步骤5:化合物13-5的合成
在室温下,将化合物13-4(2000mg,3.94mmol)溶解在二甲氧基乙烷(20mL)中,氮气保护下,加入NaH(472.83mg,11.82mmol,60%purity),室温搅拌2分钟,直至气泡减少至微量,微波90℃反应10分钟。加入300ml冰水中,浓盐酸调PH至5-6,EA提取(3*200ml),水相无产品,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩。得到棕黄色固体(1.6g,3.75mmol,95.17%yield),直接投下步。ESI-MS m/z:426.0[M+H] +
步骤6:化合物13-6的合成
室温下,在反应瓶中加入化合物13-5(10.3g,24.14mmol),加入ACN(103mL)溶解,再加入POCl 3(7.40g,48.28mmol)和DIPEA(12.48g,96.56mmol,16.82mL),然后80℃反应1h。将溶剂旋干,加无水DCM(15ml*2)带两次除掉剩余POCl 3。得到褐色油状物产物(10.5g,23.59mmol,97.72%yield)。在反应瓶中加入上述褐色油状物(10.5g,23.59mmol),加入DMF(105mL)溶解,再加入(3S)-3-甲基哌嗪-1-羧酸叔丁酯(6.14g,30.67mmol)和DIPEA(9.15g,70.77mmol,12.33mL),20℃下反应0.5小时。加入500ml冰水中,加入300ml EA,分层,水相EA提取150ml*1,合并有机相,饱和食盐水洗(2*300ml),干燥,过滤,减压浓缩。得到棕黑色固体产物(13.0g,21.35mmol,90.50%yield),直接投下步。ESI-MS m/z:608.1[M+H] +
步骤7:化合物13-7的合成
在室温下,向反应瓶中加入化合物13-6(1000mg,1.64mmol),[2-氟-6-(羟甲基)苯基]硼酸(507.44mg,2.99mmol),加入CPME(12mL),搅拌溶解,加入K 3PO 4(950.72mg,4.48mmol),H 2O(1.5mL),SPhos Pd G2(161.15mg,223.94umol),氮气保护,110℃微波反应50分钟。用EA 20ml稀释,硅藻土过滤,滤液加水20ml,分层,有机相饱和食盐水洗(1*15ml),无水硫酸钠干燥,过滤,滤液减压浓缩。残留物过硅胶柱纯化(EA:PE=10%to 100%)。得到棕色固体产物(550mg,840.79umol,56.32%yield)。ESI-MS m/z:654.2[M+H] +
步骤8:化合物13-8的合成
向反应瓶中加入化合物13-7(550mg,840.79umol),加入DMF(11mL),搅拌溶解,加入Cs 2CO 3(1.10g,3.36mmol),氮气保护,100℃微波反应40分钟。硅藻土过滤,滤液加入50ml水中,加入EA30ml,分层,有机相饱和食盐水洗(2*20ml),无水硫酸钠干燥,过滤,减压浓缩。残留物过硅胶柱纯化(EA:PE=10%to 100%)。得到棕黄色固体产物(300mg,473.08umol,56.27%yield)。ESI-MS m/z:634.3[M+H] +
步骤9:化合物13的合成
向反应瓶中加入化合物13-8(50mg,78.85umol),加入DCM(0.7mL),搅拌溶解,加入TFA(539.41mg,4.73mmol,350.27uL),20℃反应0.5小时。反应液减压浓缩,并且用无水DCM(2*3ml)带2次,加入无水DCM(1mL),DIPEA(509.51mg,3.94mmol,686.67uL),降温至0℃,滴加丙烯酰氯(8.56mg,94.62umol)的0.25ml无水DCM溶液,0℃反应10分钟。反应液DCM10ml稀释,饱和食盐水洗(3*15ml),有机相无水硫酸钠干燥,过滤,减压浓缩。残留物经prep-HPLC纯化,得到类白色固体粉末状化合物13(18.9mg,32.14umol,40.76%yield)。该化合物的特征峰如下: 1H NMR(500MHz,CDCl 3)δ8.51(d,J=4.9Hz,1H),7.52(s,1H),7.40(td,J=7.9,4.9Hz,1H),7.22-7.14(m,1H),7.07(d,J=16.7Hz,1H),6.95(d,J=7.4Hz,1H),6.69-6.52(m,1H),6.40(d,J=16.7Hz,1H),5.80(d,J=10.6Hz,1H)。ESI-MS m/z:588.2[M+H] +
实施例14:化合物(S)-1-(4-丙烯酰基-2-甲基哌嗪-1-基)-10-氟-4-(2-异丙基-4-甲基吡啶-3-基)-11-乙烯基-4H-异色烯[4,3-h]喹唑啉-3(6H)-酮的合成
Figure PCTCN2021090082-appb-000056
步骤1:化合物14-1的合成
在室温下,向化合物13-8(75mg,118.27umol)的二氧六环(3mL)和水(0.4mL)中,加入乙烯基三氟硼酸钾(158.42mg,1.18mmol),K 3PO 4(301.25mg,1.42mmol),PdCl 2(dppf) 2DCM(19.32mg,23.65umol),氮气保护,140℃下微波反应1.5hs。加入20ml EA,10ml水,分层,有机相饱和食盐水洗(2*15ml),无水硫酸钠干燥,过滤,减压浓缩。残留物通过Prep-TLC纯化(EA)。得到棕色固体状产物(48mg,76.71umol,64.86%yield)。ESI-MS m/z:626.3[M+H] +
步骤2:化合物14的合成
反应瓶中加入化合物14-1(63mg,100.68umol),加入DCM(0.9mL),搅拌溶解,加入TFA(688.79mg,6.04mmol,448.72uL),20℃反应0.5小时。反应液减压浓缩,并且用无水DCM(2*3ml)带2次,加入无水DCM(1.0mL),DIPEA(650.61mg,5.03mmol,876.83uL),降温至0℃,滴加丙烯酰氯(10.94mg,120.82umol),0℃反应10分钟。反应液DCM  10ml稀释,饱和食盐水洗(3*15ml),有机相无水硫酸钠干燥,过滤,减压浓缩。残留物pre-HPLC纯化。得到淡黄色固体粉末状产物(26.1mg,45.03umol,44.72%yield)。该化合物的特征峰如下: 1H NMR(500MHz,CDCl 3)δ8.48(d,J=4.9Hz,1H),7.71(d,J=15.0Hz,1H),7.37(ddd,J=12.7,8.9,4.9Hz,1H),7.18-7.03(m,2H),6.94(d,J=7.4Hz,1H),6.68(dt,J=54.2,23.0Hz,2H),6.40(d,J=16.8Hz,1H),5.76(dd,J=33.6,14.2Hz,2H),5.32(d,J=10.7Hz,1H)。ESI-MS m/z:580.3[M+H] +
实施例15:化合物(S)-1-(4-丙烯酰基-2-甲基哌嗪-1-基)-10-氟-4-(2-异丙基-4-甲基吡啶-3-基)-11-甲基-4H-异色烯[4,3-h]喹唑啉-3(6H)-酮的合成
Figure PCTCN2021090082-appb-000057
步骤1:化合物15-1的合成
在室温下,向化合物13-8(75mg,118.27umol)的二氧六环(3.6mL)和水(0.45mL)中,加入三甲基环硼氧烷(296.93mg,1.18mmol,50%purity),K 3PO 4(251.04mg,1.18mmol),SPhos Pd G2(17.02mg,23.65umol),氮气保护,140℃下微波反应0.5hs。加入10mlEA,5ml水,分层,水相无产品弃,有机相饱和食盐水洗(2*10ml),无水硫酸钠干燥,过滤,减压浓缩。残留物经pre-TLC纯化(EA:PE=85%)。得到棕色固体状产物(41mg,66.81umol,49.60%yield)。ESI-MS m/z:614.3[M+H] +
步骤2:化合物15的合成
反应瓶中加入化合物15-1(53.00mg,86.36umol),加入DCM(770.00uL),搅拌溶解,加入TFA(590.80mg,5.18mmol,383.63uL),20℃反应0.5小时。反应液减压浓缩,并且用无水DCM(2*3ml)带2次,加入无水DCM(1.0mL),DIPEA(558.05mg,4.32mmol,752.09uL),降温至0℃,滴加丙烯酰氯(9.38mg,103.63umol),0℃反应10分钟。反应液DCM 10ml稀释,饱和食盐水洗(3*10ml),有机相无水硫酸钠干燥,过滤,减压浓缩。残留物pre-HPLC纯化。得到白色固体粉末状产物(11.5mg,20.26umol,23.46%yield)。该化合物的特征峰如下: 1H NMR(500MHz,CDCl 3)δ8.47(d,J=4.8Hz,1H),7.39-7.29(m,2H),7.18-7.10(m,1H),7.05(t,J=4.5Hz,1H),6.94(d,J=7.4Hz,1H),6.62(d,J=38.1Hz,1H),6.39(d,J=16.7Hz,1H),5.79(d,J=10.8Hz,1H)。ESI-MS m/z:568.3[M+H] +
实施例16:化合物(S)-1-(4-丙烯酰基-2-甲基哌嗪-1-基)-10-氟-4-(2-异丙基-4-甲基吡啶-3-基)-11-(三氟甲基)-4H-异色烯[4,3-h]喹唑啉-3(6H)-酮的合成
Figure PCTCN2021090082-appb-000058
步骤1:化合物16-1的合成
500ml三口烧瓶中加入2-氨基-4-溴-3-氟-5-碘-苯甲酸(14.0g,38.90mmol),CH 3CN(230mL),CuBr(6.70g,46.68mmol)搅拌溶解,氮气置换并保护,降温至0℃,滴加亚硝酸叔丁酯(6.02g,58.35mmol)与CH 3CN(230mL)配成的溶液,约30分钟滴入,升至25℃反应3小时。将反应液缓慢倒入冷的饱和NH 4Cl溶液250ml中,搅拌,待放热停止,加入EA 200ml。搅拌,静置,分层;水相再用EA 100ml萃取;合并有机相,饱和NaCl溶液100ml洗;硫酸钠干燥,过滤,脱溶剂得粗品。过柱(EA:PE=1:1.5)得黄色固体16-1(9.3g,21.94mmol,56.42%yield)。ESI-MS m/z:422.8[M+H] +
步骤2:化合物16-2的合成
100ml烧瓶中加入化合物16-1(6.3g,14.87mmol),DCM(60mL),搅拌溶解,加入DMF(54.33mg,743.28umol,57.55uL),冰水浴冷却,滴加草酰氯(5.66g,44.60mmol),室温搅拌30min,减压脱溶,并用DCM10ml*2次带除草酰氯,用THF 10ml溶解。滴加上述THF溶液至NH 3.H 2O(15M,277.49mL),滴完后25℃反应2小时。加入EA 80ml,饱和盐水50ml,搅拌,分层;水相EA 40ml再次萃取,合并有机相,饱和盐水50ml洗;硫酸钠干燥,过滤,脱溶剂,得粗品。过柱(PE:EA=1:1)得灰白色固体产品16-2(5.8g,13.72mmol,92.28%yield)。ESI-MS m/z:421.8[M+H] +
步骤3:化合物16-3的合成
500ml烧瓶中加入化合物16-2(8.0g,18.92mmol),THF(150mL),水浴下加入草酰氯(7.20g,56.76mmol),加热回流2小时,减压脱溶,DCM带10ml*2次。加入DCM(120mL), 2-异丙基-4-甲基吡啶-3-胺(2.84g,18.92mmol),25℃反应2小时。加DCM 45ml稀释;氯化氨饱和溶液洗,饱和小苏打水溶液洗,盐水洗;硫酸钠干燥,过滤,脱溶。过硅胶柱(EA:PE=1:2)得类白色固体16-3(4.0g,6.68mmol,35.29%yield)。ESI-MS m/z:597.9[M+H] +
步骤4:化合物16-4的合成
100ml烧瓶中加入化合物16-3(4.3g,7.18mmol),THF(60mL),NaH(1.44g,35.89mmol,60%purity),氮气置换并保护,加热至50℃保温反应2小时。降温,<20℃;将反应液倒入饱和氯化铵水溶液150ml中;加入EA 100ml萃取*2次;合并有机相,盐水洗,硫酸钠干燥,过滤,减压脱溶剂得粗品。用THF:PE=1:5约100ml过浆,得类白色固体产品16-4(3.3g,6.37mmol,88.73%yield)。ESI-MS m/z:517.9[M+H] +
步骤5:化合物16-5的合成
在室温下,25mL烧瓶中加入化合物16-4(2.1g,4.05mmol),CH 3CN(24mL),DIPEA(2.10g,16.21mmol,2.82mL),搅拌下加入POCl 3(1.86g,12.16mmol),加热至80℃反应1小时。降温<40℃,减压脱溶,DCM带除酸20ml*2次,得黄褐色固体产品16-5(4.0g,粗品)。ESI-MS m/z:535.9[M+H] +
步骤6:化合物16-6的合成
在室温下,25ml烧瓶中加入化合物16-5(4.0g,7.45mmol),DMF(15mL),叔丁基(3S)-3-甲基哌嗪-1-羧酸酯(1.94g,9.69mmol),DIPEA(3.85g,29.82mmol,5.19mL),25℃反应2小时。加EA 100ml,水30ml,搅拌,分层;水洗10ml*3次;盐水洗,硫酸钠干燥,脱溶剂得粗品;将粗品过硅胶柱(EA:PE=1:1)得黄色泡沫状固体产品16-6(4.0g,5.71mmol,76.61%yield)。ESI-MS m/z:700.1[M+H] +
步骤7:化合物16-7的合成
在室温下,100ml烧瓶中加入化合物16-6(1g,1.43mmol),DMF(18mL),CuI(67.98mg,356.95umol),2,2-二氟-2-氟磺酰乙酸甲酯(329.16mg,1.71mmol)氮气置换并保护,80℃反应2小时。降温,<30℃。加EA100ml,水20ml,搅拌,分层;水洗20ml*3次;盐水洗;硫酸钠干燥,过滤,脱溶得粗品。Pre-HPLC得产品,得白色固体产品16-7(0.23g,357.99umol,25.07%yield)。ESI-MS m/z:642.2[M+H] +
步骤8:化合物16-8的合成
在室温下,10ml烧瓶中加入化合物16-7(220mg,342.42umol),CPME(3mL),[2-氟-6-(羟甲基)苯基]硼酸(116.39mg,684.84umol),K 3PO 4(218.06mg,1.03mmol), SPhos-Pd-G2(36.96mg,51.36umol),H 2O(0.3mL),氮气置换并保护,加热至110℃,反应0.5小时。冷却至室温;分层,盐水洗;硫酸钠干燥,过滤;脱溶剂得粗品。将粗品通过Pre-TLC得白色固体产品16-8(52mg,75.61umol,21.23%yield)。ESI-MS m/z:688.3[M+H] +
步骤9:化合物16-9的合成
在室温下,10ml烧瓶中加入化合物16-8(43mg,62.53umol),DMF(5mL),Cs 2CO 3(101.69mg,312.64umol),氮气置换并保护,加热至65℃反应2小时。降温;加EA 30ml,水5ml,搅拌,分层;盐水洗5ml*3次;硫酸钠干燥,过滤,减压脱溶剂得粗品。将粗品通过Pre-TLC纯化(EA:PE=3:1)得白色泡沫状固体16-9(35mg,52.42umol,83.83%yield)。ESI-MS m/z:668.3[M+H] +
步骤10:化合物16-10的合成
10ml烧瓶中加入化合物16-9(35mg,52.42umol),EtOH(1mL),HCl/二氧六环(2M,1.31mL),20℃反应2小时。减压脱溶剂,得褐色油状液体16-10(32mg,crude)。ESI-MS m/z:568.2[M+H] +
步骤11:化合物16的合成
10ml烧瓶中加入化合物16-10(32mg,56.38umol),DCM(1mL),DIPEA(7.29mg,56.38umol,9.82uL),氮气置换并保护,降温至0℃,加入丙烯酰氯(10.21mg,112.76umol),继续0℃反应30min。加DCM10ml,盐水洗10ml;硫酸钠干燥,过滤;减压脱溶剂,得粗品。将粗品通过Pre-HPLC得白色固体产品16(7.6mg,12.23umol,21.69%yield)。该化合物的特征峰如下:NMR(500MHz,)δ8.51(d,J=4.9Hz),7.75(d,J=12.3Hz),7.45-7.34(m),7.12(dd,J=24.0,15.2Hz),6.94(d,J=7.4Hz),6.74-6.50(m),6.40(d,J=16.8Hz),5.80(d,J=10.5Hz)。ESI-MS m/z:622.2[M+H] +
实施例17:化合物(S)-8-(4-丙烯酰-2-甲基哌嗪-1-基)-1,2,12-三氟-11-(2-异丙基-4-甲基吡啶-3-基)-5H异色烯[3,4-g]喹唑啉-10(11H)-酮的合成
Figure PCTCN2021090082-appb-000059
化合物17具体合成步骤参照实施例3。ESI-MS m/z:590.2[M+H] +
实施例18:化合物S)-1-(4-丙烯酰-2-甲基哌嗪-1-基)-11-氯-9,10-二氟-4-(2-异丙基-4-甲基吡啶-3-基)-4H-异色烯[4,3-h]喹唑啉-3(6H)-酮的合成
Figure PCTCN2021090082-appb-000060
化合物18具体合成步骤参照实施例13。该化合物的特征峰如下: 1H NMR(500MHz,CDCl 3)δ8.49(d,J=4.7Hz,1H),7.53(s,1H),7.23(dd,J=16.6,8.4Hz,1H),7.07(s,1H),6.90(dd,J=7.6,3.9Hz,1H),6.77-6.50(m,1H),6.40(d,J=16.7Hz,1H),5.80(d,J=10.5Hz,1H)。ESI-MS m/z:606.2[M+H] +
实施例19:化合物10-((S)-4-丙烯酰-2-甲基哌嗪-1-基)-8-氯-7-氟-13-(2-异丙基-4-甲基吡啶-3-基)-2,3-二氢苯并[4,5]氧杂[3,2-h]喹唑啉-12(13H)-酮的合成
Figure PCTCN2021090082-appb-000061
化合物19具体合成步骤参照实施例3与实施例13。 1H NMR(500MHz,CDCl 3)δ8.44(d,J=4.9Hz,1H),7.67(s,1H),7.36(dd,J=13.3,7.9Hz,1H),7.09(dd,J=10.9,7.2Hz,2H),7.00(d,J=7.5Hz,1H),6.61(d,J=21.4Hz,1H),6.40(d,J=16.9Hz,1H),5.80(d,J=10.5Hz,1H);ESI-MS m/z:602.2[M+H] +
实施例20:化合物(S)-7-(4-丙烯酰基-2-甲基哌嗪-1-基)-5-氯-10-(2-异丙基-4-甲基吡啶-3-基)-10H吡啶[3',2':4,5]吡喃[3,2-h]喹唑啉-9(12H)-酮的合成
Figure PCTCN2021090082-appb-000062
化合物20具体合成步骤参照实施例13。该化合物的特征峰如下: 1H NMR(500MHz,CDCl 3)δ8.65(d,J=8.0Hz,1H),8.50(d,J=4.9Hz,2H),7.51(s,1H),7.35(dd,J=8.1,4.8Hz,1H),7.07(dd,J=10.0,6.1Hz,1H),6.58(s,1H),6.40(d,J=16.6Hz,1H),5.80(d,J=10.5Hz,1H)。ESI-MS m/z:571.2[M+H] +
实施例21:化合物(S)-1-(4-丙烯酰基-2-甲基哌嗪-1-基)-13-氟-4-(2-异丙基-4-甲基吡啶-3-基)-4H-苯并[5,6]异色烯[4,3-h]喹唑啉-3(6H)-酮的合成
Figure PCTCN2021090082-appb-000063
化合物21具体合成步骤参照实施例3。 1H NMR(500MHz,CDCl 3)δ8.49(d,J=4.9Hz,1H),7.97-7.84(m,3H),7.57(ddd,J=14.9,13.8,6.8Hz,2H),7.30(d,J=10.3Hz,1H),7.21(d,J=8.3Hz,1H),7.06(d,J=10.1Hz,1H),6.62(dd,J=34.9,10.8Hz,1H),6.41(d,J=16.6Hz,1H),5.81(dd,J=10.5,1.4Hz,1H),5.30(s,6H),4.62(q,J=13.1Hz,2H);ESI-MS m/z:604.3[M+H] +
实施例22:化合物(S)-10-(4-丙烯酰基-2-甲基哌嗪-1-基)-14-氟-13-(2-异丙基-4-甲基吡啶-3-基)-7H-苯并[5,6]异色烯[3,4-g]喹唑啉-12(13H)-酮的合成
Figure PCTCN2021090082-appb-000064
化合物22具体合成步骤参照实施例3。ESI-MS m/z:604.3[M+H] +
实施例23:化合物(S)-1-(4-丙烯酰基-2-甲基哌嗪-1-基)-11-氯-4-(2-异丙基-4-甲基吡啶-3-基)-4H-吡啶[3',4':4,5]吡喃并[3,2-h]喹唑啉-3(6H)-酮的合成
Figure PCTCN2021090082-appb-000065
化合物23具体合成步骤参照实施例13。该化合物的特征峰如下: 1H NMR(500MHz,CDCl 3)δ9.58(s,1H),8.59(d,J=4.8Hz,1H),8.50(d,J=4.9Hz,1H),7.54(s,1H),7.10-7.01(m,2H),6.58(s,1H),6.40(d,J=17.0Hz,1H),5.80(d,J=10.5Hz,1H)。ESI-MS m/z:571.2[M+H] +
实施例24:化合物10-((S)-4-丙烯酰基-2-甲基哌嗪-1-基)-8-氯-13-(4,6-二乙基嘧啶-5-基)-7-氟-2,3-二氢苯并[4,5]环氧庚烷[3,2-h]喹唑啉-12(13H)-酮的合成
Figure PCTCN2021090082-appb-000066
化合物24具体合成步骤参照实施例13。该化合物的特征峰如下: 1H NMR(500MHz,CDCl 3)δ9.06(d,J=12.0Hz,1H),7.39(dd,J=12.9,7.3Hz,1H),7.15-6.98(m,2H),6.67-6.47(m,1H),6.38(dd,J=21.9,16.6Hz,1H),5.81(d,J=10.5Hz,1H)。ESI-MS m/z:603.2[M+H] +
实施例25:化合物2-(1-丙烯酰基-4-(10,11-二氟-4-(2-异丙基-4-甲基吡啶-3-基)-3-氧代-3,6-二氢-4H-异色烯[4,3-h]喹唑啉-1-基)哌嗪-2-基)乙腈的合成
Figure PCTCN2021090082-appb-000067
化合物25具体合成步骤参照实施例3。 1H NMR(500MHz,CDCl 3)δ8.54(d,J=4.6Hz,1H),7.39(dd,J=12.6,7.7Hz,1H),7.33-7.28(m,1H),7.16(dd,J=20.3,11.3Hz,2H),6.93(d,J=7.3Hz,1H),6.68-6.57(m,1H),6.47-6.39(m,1H),5.87(d,J=10.4Hz,1H),4.56-4.36(m, 4H);ESI-MS m/z:597.2[M+H] +
实施例26:化合物2-(1-丙烯酰基-4-(1,12-二氟-11-(2-异丙基-4-甲基吡啶-3-基)-10-氧-10,11-二氢-5H-异色烯[3,4-g]喹唑啉-8-基)哌嗪-2-基)乙腈的合成
Figure PCTCN2021090082-appb-000068
化合物26具体合成步骤参照实施例3。ESI-MS m/z:597.2[M+H] +
实施例27:化合物(S)-1-(4-丙烯酰-2-甲基哌嗪-1-基)-11-氯-9,10-二氟-4-(3-异丙基-6,7-二氢-5H-环戊基[c]吡啶-4-基)-4H-异色[4,3-h]喹唑啉-3(6H)-酮的合成
Figure PCTCN2021090082-appb-000069
化合物27具体合成步骤参照实施例13。ESI-MS m/z:632.2[M+H] +
实施例28:化合物(S)-1-(4-丙烯酰-2-甲基哌嗪-1-基)-11-氯-10-氟-7-羟基-4-(2-异丙基-4-甲基吡啶-3-基)-4H-异色[4,3-h]喹唑啉-3(6H)-酮的合成
Figure PCTCN2021090082-appb-000070
化合物28具体合成步骤参照实施例3。ESI-MS m/z:604.2[M+H] +
实施例29:化合物(S)-1-(4-丙烯酰-2-甲基哌嗪-1-基)-11-氯-7,10-二氟-4-(2-异丙基-4-甲基吡啶-3-基)-4H-异色[4,3-h]喹唑啉-3(6H)-酮的合成
Figure PCTCN2021090082-appb-000071
化合物29具体合成步骤参照实施例3。ESI-MS m/z:606.2[M+H] +
实施例30:化合物10-((S)-4-丙烯酰-2-甲基哌嗪-1-基)-8-氯-6,7-二氟-13-(2-异丙基-4-甲基吡啶-3-基)-2,3-二氢苯并[4,5]氧杂[3,2-h]喹唑啉-12(13H)-酮的合成
Figure PCTCN2021090082-appb-000072
化合物30具体合成步骤参照实施例3。ESI-MS m/z:620.2[M+H] +
实施例31:化合物10-((S)-4-丙烯酰-2-甲基哌嗪-1-基)-8-氯-13-(3,5-二乙基吡啶-4-基)-7-氟-2,3-二氢苯并[4,5]氧杂[3,2-h]喹唑啉-12(13H)-酮的合成
Figure PCTCN2021090082-appb-000073
化合物31具体合成步骤参照实施例13。ESI-MS m/z:602.2[M+H] +
实施例32:化合物10-((S)-4-丙烯酰-2-甲基哌嗪-1-基)-8-氯-13-(4,6-二环丙基嘧啶-5-基)-7-氟-2,3-二氢苯并[4,5]氧杂[3,2-h]喹唑啉-12(13H)-酮的合成
Figure PCTCN2021090082-appb-000074
化合物32具体合成步骤参照实施例13。ESI-MS m/z:627.2[M+H] +
实施例33:化合物1-(5-丙烯酰-2,5-二氮杂二环[4.1.0]庚烷-2-基)-11-氯-10-氟-4-(2-异丙基-4-甲基吡啶-3-基)-4H-异色烯醇[4,3-h]喹唑啉-3(6H)-酮的合成
Figure PCTCN2021090082-appb-000075
化合物33具体合成步骤参照中间体M-11的合成和实施例3。ESI-MS m/z:586.2[M+H] +
实施例34:化合物(S)-11-氯-9,10-二氟-1-(4-(2-氟丙烯酰)-2-甲基哌嗪-1-基)-4-(2-异丙基-4-甲基吡啶-3-基)-4H-异色[4,3-h]喹唑啉-3(6H)-酮的合成
Figure PCTCN2021090082-appb-000076
化合物34具体合成步骤参照实施例3。ESI-MS m/z:624.2[M+H] +
实施例17-23与28-30参考实施例3的步骤进行合成,将羧酸还原为醛基进而还原为羟基,然后将溴原子进行硼酯化,用共用中间体M-11或13-6与相应硼酯通过Suzuki偶联、关环、脱保护和引入丙烯酰基最终得到目标产物;实施例24、27、31与32将氯分别进行乙基化、异丙基化与环丙基化,参照M-11的合成,然后与相应硼酯通过Suzuki偶联、关环、脱保护和引入丙烯酰基最终得到目标产物;实施例25和26进行Boc保护后参照M-11 的合成,然后与相应硼酯通过Suzuki偶联、关环、脱保护和引入丙烯酰基最终得到目标产物。
对应的合成中间体如下表:
Figure PCTCN2021090082-appb-000077
Figure PCTCN2021090082-appb-000078
实施例35:化合物(S)-1-(4-丙烯酰-2-甲基哌嗪-1-基)-11-氯-9,10-二氟-4-(2-异丙基-4-甲基吡啶-3-基)-4H-异色[4,3-h]喹唑啉-3(6H)-酮-6,6-d2的合成
Figure PCTCN2021090082-appb-000079
步骤1:化合物35-1的合成
在室温下,将2-溴-3,4-二氟苯甲酸(20.00g,84.39mmol)溶于甲醇(100.00mL)中,加入浓硫酸(4.52mL),升温至70℃反应8h。将反应液浓缩,浓缩物经过柱层析分离纯化得到目标产物35-1(18.2g,85.92%产率)。ESI-MS m/z:250.9[M+H] +
步骤2:化合物35-2的合成
在室温下,将化合物35-1(8.00g,31.87mmol),联硼酸频哪醇(9.60g),PdCl 2(dppf).CH 2Cl 2和醋酸钾溶于二氧六环,氮气置换,升温至100℃反应过夜。将反应混合物过滤后,向其中加入乙酸乙酯和水进行萃取,有机相用无水硫酸钠干燥后浓缩,浓缩物经硅胶柱纯化后,得到目标产物35-2。ESI-MS m/z:299.1[M+H] +
步骤3:化合物35-3的合成
在室温下,将化合物35-2(1.72g,5.77mmol)溶于四氢呋喃(8.60mL)中,25℃加入NaBD 4(0.8600g),再缓慢滴加CD 3OD(8.60mL),加毕,搅拌30min。蒸干反应夜,将蒸干的粗品用水溶解,用稀盐酸调节PH至酸性3,有白色固体析出,抽滤,滤饼即目标产物35-3(0.8700g,87.69%产率)。ESI-MS m/z:173.1[M+H] +
步骤4:化合物35-4的合成
在室温下,将化合物35-3(0.71g,4.11mmol),化合物13-6(1.00g,1.64mmol),sphos G2 Pd(0.12g)和磷酸钾(1.05g)溶于环戊基甲基醚(9.00mL)和水(1.00mL)的混合溶液中,氮气置换,微波升温至100℃反应25min。EA萃取反应液三次,饱和食盐水洗涤,无水硫酸镁干燥,蒸干,拌样柱层析分离纯化得粗产物,粗产物再经过薄层板分离纯化得目标产物35-4(0.11g,9.93%产率)。ESI-MS m/z:674.3[M+H] +
步骤5至步骤7的合成步骤参考实施例1。ESI-MS m/z:608.2[M+H] +
实施例36:化合物(S)-8-(4-丙烯酰基-2-甲基哌嗪-1-基)-11-(2-异丙基-4-甲基吡啶-3-基)-5H-异色烯[3',4':5,6]吡啶并[2,3-d]嘧啶-10(11H)-酮的合成
Figure PCTCN2021090082-appb-000080
步骤1与步骤2具体合成步骤参照实施例16。
步骤3:化合物36-3的合成
于250ml单口瓶中加入化合物36-2(2g,5.73mmol),ACN(40mL),DIEA(3.71g,28.67mmol,4.99mL),然后0℃滴加POCl 3(2.64g,17.20mmol),升温至80℃搅拌一小时。直接浓缩得到黑色固体粗品36-3(1.91g,5.20mmol,90.70%yield)。
步骤4:化合物36-4的合成
于250ml单口瓶中加入化合物36-3(1.9g,5.17mmol),DCM(40mL),DIEA(2.67g,20.70mmol,3.60mL),0℃下搅拌加入(3S)-3-甲基哌嗪-1-羧酸叔丁酯(1.35g,6.73mmol),然后恢复室温搅拌1小时。加入硅胶拌样过柱(DCM/MEOH=20:1),浓缩得到棕色固体状产物(2.2g,4.14mmol,80.07%yield)。
步骤5:化合物27-5的合成
于20ml微波管中加入[2-(羟甲基)苯基]硼酸(114.46mg,753.27umol),化合物36-4(200mg,376.63umol),SPHOS Pd G2(54.21mg,75.33umol),K 3PO 4(159.89mg,753.27umol),CPME(4mL),H 2O(0.4mL)置换氮气后,微波90℃反应50分钟。加入EA,有机相水洗,有机相用无水硫酸钠干燥,过滤,旋干。过硅胶柱纯化DCM/MEOH(15:1)得到淡黄色固体状产物(170mg,282.07umol,74.89%yield)。ESI-MS m/z:603.4[M+H] +
步骤6:化合物36-6的合成
于微波管中加入化合物36-5(150mg,248.88umol),Cs 2CO 3(162.18mg,497.76umol),DMF(3mL)微波100℃反应15分钟。加入EA,有机相水洗,有机相用无水硫酸钠干燥,过滤,浓缩,得到淡黄色固体状产物(130mg,223.10umol,89.64%yield)。ESI-MS m/z:583.4[M+H] +
步骤7:化合物36-7的合成
于25ml单口瓶中加入化合物36-6(130mg,223.10umol),DCM(6mL),常温搅拌下加入TFA(1mL),然后常温搅拌10分钟。直接浓缩,得到淡黄色油状产物(97mg,201.00umol,90.10%yield)。ESI-MS m/z:483.4[M+H] +
步骤8:化合物36的合成
于25ml单口瓶中加入化合物36-7(97mg,201.00umol),DCM(5mL),DIEA(77.94mg,603.01umol,105.03uL),0℃下缓慢滴加丙烯酰氯(18.19mg,201.00umol),然后搅拌10分钟。加入冰水,EA萃取,有机相水洗,有机相用无水硫酸钠干燥,过滤,旋干。通过Pre-HPLC纯化得到淡黄色固体状目标产物(58.8mg,109.57umol,54.51%yield)。该化合物的特征峰如下: 1H NMR(500MHz,CDCl 3)δ8.60(d,J=4.9Hz,1H),7.57-7.47(m,2H),7.41(t,J=7.5Hz,1H),7.29(t,J=7.8Hz,1H),7.14(dd,J=14.5,6.1Hz,2H),6.68-6.53(m,1H),6.40(d,J=16.9Hz,1H),5.80(d,J=10.6Hz,1H)。ESI-MS m/z:537.4[M+H] +
实施例37:化合物(S)-8-(4-丙烯酰基-2-甲基哌嗪-1-基)-1-羟基-11-(2-异丙基-4-甲基吡啶-3-基)-5H-异色烯[3',4':5,6]吡啶并[2,3-d]嘧啶10(11H)-酮的合成
Figure PCTCN2021090082-appb-000081
步骤1:化合物37-1的合成
于微波管中加入[2-(羟甲基)-6-甲氧基苯基]硼酸(137.08mg,753.27umol),化合物36-4(200mg,376.63umol),CPME(3mL),H 2O(0.5mL),SPHOS Pd G2(54.21mg,75.33umol),K 3PO 4(159.89mg,753.27umol),反应液置换氮气之后,90℃微波反应50分钟。加入EA和水,无水硫酸钠干燥,过滤旋干。通过Pre-TLC纯化得到棕色固体状目标产物37-1(211mg,333.48umol,88.54%yield)。ESI-MS m/z:633.46[M+H] +
步骤2:化合物37-2的合成
微波管中加入化合物37-1(210mg,331.90umol),DMF(4mL),Cs 2CO 3(324.42mg,995.70umol)然后100℃微波反应15分钟。加入EA,有机相水洗,有机相用无水硫酸钠干燥,过滤,浓缩得到淡黄色固体状目标产物37-2(191mg,311.73umol,93.92%yield)。ESI-MS m/z:613.43[M+H] +
步骤3:化合物37-3的合成
25ml单口瓶中加入化合物37-2(190mg,310.09umol),DCM(5mL),TFA(1mL),常温搅拌20分钟。直接浓缩得到棕色油体状目标产物37-3(149mg,290.67umol,93.74%yield)。ESI-MS m/z:513.35[M+H] +
步骤4:化合物37-4的合成
于25ml单口瓶中加入化合物37-3(141mg,275.07umol),DCM(5mL),DIEA(106.65mg,825.20umol,143.73uL),0℃搅拌下加入丙烯酰氯(24.90mg,275.07umol),搅拌反应五分钟。加入冰水,饱和碳酸氢钠溶液,淬灭,EA萃取,有机相无水硫酸钠干燥,过滤,旋干。通过Pre-TLC纯化得到淡黄色固体状目标产物37-4(211mg,333.48umol,88.54% yield)。ESI-MS m/z:567.40[M+H] +
步骤5:化合物37-5的合成
单口瓶中加入化合物37-4(50mg,88.24umol),二氯乙烷(10mL),0℃下滴加BBr 3(88.24mg,352.95umol),然后升温至60℃搅拌6小时。0摄氏度加入冰水,然后加入饱和碳酸氢钠溶液,EA萃取,干燥,过滤,旋干。直接浓缩得到淡红色固体状粗品目标产物37-5。ESI-MS m/z:633.00[M+H] +
步骤6:化合物37的合成
单口瓶中加入化合物37-5(27mg,42.62umol),THF(9mL),然后加入1ml的2N的氢氧化钠溶液,搅拌30分钟。加入饱和碳酸氢钠溶液,然后除去THF,EA萃取,有机相用无水硫酸钠干燥,过滤,旋干。通过Pre-TLC纯化得到棕色固体状目标产物37(13.0mg,23.52umol,55.20%yield)。ESI-MS m/z:553.3[M+H] +
下述的实施例采用上述方法合成,或使用相应中间体的类似方法合成。
Figure PCTCN2021090082-appb-000082
Figure PCTCN2021090082-appb-000083
Figure PCTCN2021090082-appb-000084
Figure PCTCN2021090082-appb-000085
Figure PCTCN2021090082-appb-000086
Figure PCTCN2021090082-appb-000087
药理实验
实施例1:细胞增殖抑制检测
将MIA PaCa-2细胞按600细胞、160μL/孔铺96孔超低吸附板。孵育隔夜后,配制梯度浓度的化合物溶液,分别向各孔细胞中加入40μL各浓度的待测化合物DMSO溶液,化合物终浓度为10000、2000、400、80、16、3.2、0.64、0.12、0.025、0nM(DMSO终浓 度均为0.25%)。37℃,5%CO 2孵育96h。向各孔中加入50μL Cell-titer Glo工作液,震荡混匀后室温孵育10min,多功能酶标仪读取Luminescence发光值,将发光值读数转换为抑制百分数:
抑制百分数=(最大值-读数)/(最大值-最小值)*100。
“最大值”为DMSO对照;“最小值”表示无细胞对照组。
用Graphpad Prism软件进行曲线拟合并得到IC 50值。
实施例化合物对MIA PaCa-2细胞抑制的IC 50数据参见表1。
实施例2:细胞增殖抑制检测
将H358细胞按2000细胞、190μL/孔铺96孔超低吸附板。孵育隔夜后,配制梯度浓度的化合物溶液,分别向各孔细胞中加入10μL各浓度的待测化合物DMSO溶液,化合物终浓度为10000、3333.3、1111.1、370.4、123.5、41.2、13.7、4.6、1.5、0nM(DMSO终浓度均为0.25%)。37℃,5%CO 2孵育96h。向各孔中加入50μL Cell-titer Glo工作液,震荡混匀后室温孵育10min,多功能酶标仪读取Luminescence发光值,将发光值读数转换为抑制百分数:
抑制百分数=(最大值-读数)/(最大值-最小值)*100。
“最大值”为DMSO对照;“最小值”表示无细胞对照组。
用Graphpad Prism软件进行曲线拟合并得到IC 50值。
实施例化合物对H358细胞抑制的IC 50数据参见表1。
表1
Figure PCTCN2021090082-appb-000088
Figure PCTCN2021090082-appb-000089
虽然本发明已通过其实施方式进行了全面的描述,但是值得注意的是,各种变化和修改对于本领域技术人员都是显而易见的。这样的变化和修改都应该包括在本发明所附权利要求的范围内。

Claims (24)

  1. 一种通式(I)所示的化合物、其互变异构体、氘代物或药用盐:
    Figure PCTCN2021090082-appb-100001
    其中,
    R 1各自独立地选自H、氨基、卤素、C 1-3氰基、C 1-3羟基烷基、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基;或者2个R 1与相连的原子共同形成C 3-6环烷基或3-6元杂环基;
    R 2为丙烯酰基或取代的丙烯酰基;
    A环选自C 3-10环烷基、3-10元杂环基、C 6-12芳基、5-12元杂芳基;
    X 1独立地选自-C(R 4) 1-2-(CH 2) 0-2-、-NR 4-(CH 2) 0-2-、-O-(CH 2) 0-2-、-S-(CH 2) 0-2-、
    Figure PCTCN2021090082-appb-100002
    Figure PCTCN2021090082-appb-100003
    -CH=CH-或-N=CH-;
    X 2独立地选自-C(R 5) 1-2-(CH 2) 0-2-、-NR 5-(CH 2) 0-2-、-O-(CH 2) 0-2-、-S-(CH 2) 0-2-、
    Figure PCTCN2021090082-appb-100004
    Figure PCTCN2021090082-appb-100005
    -CH=CH-或-N=CH-;
    X 3选自C、CH或N;
    X 4选自CR 6或N;
    X 5选自CR 7或N;
    X 6选自CR 8或N;
    X 7选自NR 9或CHR 9
    R 6或R 8独立地选自H、卤素、C 1-3烷基、C 1-3卤代烷基、C 1-3烷氧基、C 3-4环烷基、C 3-12杂环烷基、C 2-3烯基、C 2-3炔基、芳基或杂芳基;
    R 7选自H、羟基、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、-NH-C 1-6烷基、-N(C 1-6烷基) 2、氰基或卤素;
    R 9选自C 1-6烷基、-C 0-3亚烷基-C 6-14芳基、-C 0-3亚烷基-(5-14元杂芳基)、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-O-C 0-3亚烷基-C 6-14芳基、-O-C 0-3亚烷基-(5-14元杂芳基)、-O-C 0-3亚烷基-C 3-14环烷基、-O-C 0-3亚烷基-(3-14元杂环烷基)、-NH-C 1-6烷基、-N(C 1-6烷基) 2、-NH-C 0-3亚烷基-C 6-14芳基、-NH-C 0-3亚烷基-(5-14元杂芳基)、-NH-C 0-3亚烷基-C 3-14环烷基、-NH-C 0-3亚烷基-(3-14元杂环烷基),R 9任选未取代或进一步被一个或多个R 9a取代基所取代;
    R 3、R 4、R 5、R 9a各自独立地选自H、D、氧代、硝基、卤素、C 1-6烷基、C 1-6氰基、C 1-6卤代烷基、-C 0-3亚烷基-OR a、-C 0-3亚烷基-N(R a) 2、-C 0-3亚烷基-NR aC(=O)R a、-C 0-3亚烷基-NR aC(=O)OR a、-C 0-3亚烷基-NR aS(=O) 2R a、-C 0-3亚烷基-S(=O)R a、-C 0-3亚烷基-S(=O) 2R a、-C 0-3亚烷基-S(=O) 2N(R a) 2、-C 0-3亚烷基-SR a、-C 0-3亚烷基-S(R a) 5、-C 0-3亚烷基-C(=O)N(R a) 2、-C 0-3亚烷基-C(=O)R a、-C 0-3亚烷基-C(=O)OR a、C 2-6烯基、C 2-6炔基、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-C 0-3亚烷基-C 6-14芳基或-C 0-3亚烷基-(5-14元杂芳基),所述-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-C 0-3亚烷基-C 6-14芳基或-C 0-3亚烷基-(5-14元杂芳基)任选未取代或进一步被一个或多个R a取代基所取代,且每个R a独立地选自H、卤素、C 1-6烷基、C 1-6卤代烷基、C 3-14环烷基、3-14元杂环烷基、C 2-3烯基、C 2-3炔基、芳基或杂芳基;
    a或b独立地选自1、2、3或4;
    Figure PCTCN2021090082-appb-100006
    为双键或单键。
  2. 根据权利要求1所述的化合物、其互变异构体、氘代物或药用盐,其特征在于,选自式(I-1)化合物,
    Figure PCTCN2021090082-appb-100007
    其中,
    R 1各自独立地选自H、氨基、卤素、C 1-3氰基、C 1-3羟基烷基或C 1-6烷基;或者2个R 1与相连的原子共同形成C 3-6环烷基或3-6元杂环基;
    R 2为丙烯酰基或取代的丙烯酰基;
    A环选自C 3-10环烷基、3-10元杂环基、C 6-12芳基、5-12元杂芳基;
    X 1独立地选自-C(R 4) 2-(CH 2) 0-2-、-NR 4-(CH 2) 0-2-、-O-(CH 2) 0-2-、-S-(CH 2) 0-2-、
    Figure PCTCN2021090082-appb-100008
    Figure PCTCN2021090082-appb-100009
    -C(=O)-(CH 2) 0-2-、-CH=CH-或-N=CH-,R 4独立地选自H、D、卤素或C 1-3烷基;
    X 2独立地选自-C(R 5) 2-(CH 2) 0-2-、-NR 5-(CH 2) 0-2-、-O-(CH 2) 0-2-、-S-(CH 2) 0-2-、
    Figure PCTCN2021090082-appb-100010
    Figure PCTCN2021090082-appb-100011
    -C(=O)-(CH 2) 0-2-、-CH=CH-或-N=CH-,R 5独立地选自H、D、卤素或C 1-3烷基;
    X 3选自C、CH或N;
    X 4选自CR 6或N;
    R 6独立地选自H、卤素、C 1-3烷基、C 1-3卤代烷基、C 1-3烷氧基、C 3-4环烷基、C 3-12杂环烷基、C 2-3烯基、C 2-3炔基、芳基或杂芳基;
    R 7选自H、羟基、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、-NH-C 1-6烷基、-N(C 1-6烷基) 2、氰基或卤素;
    R 9选自-C 0-3亚烷基-C 6-14芳基、-C 0-3亚烷基-(5-14元杂芳基)、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-O-C 0-3亚烷基-C 6-14芳基、-O-C 0-3亚烷基-(5-14元杂芳基)、-O-C 0-3亚烷基-C 3-14环烷基、-O-C 0-3亚烷基-(3-14元杂环烷基)、-NH-C 1-6烷基、-N(C 1-6烷基) 2、-NH-C 0-3亚烷基-C 6-14芳基、-NH-C 0-3亚烷基-(5-14元杂芳基)、-NH-C 0-3亚烷基-C 3-14环烷基、-NH-C 0-3亚烷基-(3-14元杂环烷基),R 9任选未取代或进一步被1-4个R 9a取代基所取代;
    R 3或R 9a各自独立地选自H、D、氧代、硝基、卤素、C 1-6烷基、C 1-6氰基、C 1-6卤代烷基、-C 0-3亚烷基-OR a、-C 0-3亚烷基-N(R a) 2、-C 0-3亚烷基-NR aC(=O)R a、-C 0-3亚烷基-NR aC(=O)OR a、-C 0-3亚烷基-NR aS(=O) 2R a、-C 0-3亚烷基-S(=O)R a、-C 0-3亚烷基-S(=O) 2R a、-C 0-3亚烷基-S(=O) 2N(R a) 2、-C 0-3亚烷基-SR a、-C 0-3亚烷基-S(R a) 5、-C 0-3亚烷基-C(=O)N(R a) 2、-C 0-3亚烷基-C(=O)R a、-C 0-3亚烷基-C(=O)OR a、C 2-6烯基、C 2-6炔基、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-C 0-3亚烷基-C 6-14芳基或-C 0-3亚烷基-(5-14元杂芳基),所述-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-C 0-3亚烷基 -C 6-14芳基或-C 0-3亚烷基-(5-14元杂芳基)任选未取代或进一步被一个或多个R a取代基所取代,且每个R a独立地选自H、卤素、C 1-6烷基、C 1-6卤代烷基、C 3-14环烷基、3-14元杂环烷基、C 2-3烯基、C 2-3炔基、芳基或杂芳基;
    a或b独立地选自1、2、3或4;
    Figure PCTCN2021090082-appb-100012
    为双键或单键。
  3. 根据权利要求1或2所述的化合物、其互变异构体、氘代物或药用盐,其特征在于,所述
    Figure PCTCN2021090082-appb-100013
    选自
    Figure PCTCN2021090082-appb-100014
    Figure PCTCN2021090082-appb-100015
  4. 根据权利要求1-3任一项所述的化合物、其互变异构体或药用盐,其特征在于,所述R 2选自
    Figure PCTCN2021090082-appb-100016
    Figure PCTCN2021090082-appb-100017
  5. 根据权利要求1-4任一项所述的化合物、其互变异构体、氘代物或药用盐,其特征在于,所述A环选自
    Figure PCTCN2021090082-appb-100018
    Figure PCTCN2021090082-appb-100019
  6. 根据权利要求1-5任一项所述的化合物、其互变异构体或药用盐,其特征在于,所述R 3选自H、卤素、氧代、-OR a、-C 0-3亚烷基-N(R a) 2、C 1-6卤代烷基,R a独立地为H、C 1-6烷基、C 1-6卤代烷基。
  7. 根据权利要求1-6任一项所述的化合物、其互变异构体、氘代物或药用盐,其特征在于,所述X 1选自-C(R 4) 2-(CH 2) 0-2-、-NH-(CH 2) 0-2-、-O-(CH 2) 0-2-、-C(O)-(CH 2) 0-2-、
    Figure PCTCN2021090082-appb-100020
    -CH=CH-或-N=CH-,R 4独立地选自H、D、卤素或C 1-3烷基。
  8. 根据权利要求1-7任一项所述的化合物、其互变异构体、氘代物或药用盐,其特征在于,所述X 2选自-C(R 5) 2-(CH 2) 0-2-、-NH-(CH 2) 0-2-、-O-(CH 2) 0-2-、-C(O)-(CH 2) 0-2-、
    Figure PCTCN2021090082-appb-100021
    -CH=CH-或-N=CH-,R 5独立地选自H、D、卤素或C 1-3烷基。
  9. 根据权利要求1-8任一项所述的化合物、其互变异构体、氘代物或药用盐,其特征在于,所述X 3的取代基选自C、CH或N。
  10. 根据权利要求1-9所述的任一项化合物、其互变异构体、氘代物或药用盐,其特征在于,所述X 4选自CR 6或N,且R 6的取代基选自卤素、C 1-3烷基、C 1-3卤代烷基或C 2-3烯基。
  11. 根据权利要求1-10所述的任一项化合物、其互变异构体、氘代物或药用盐,其特征在于,所述X 5选自CR 7或N,且R 7的取代基选自H或C 1-3烷基。
  12. 根据权利要求1-11所述的任一项化合物、其互变异构体、氘代物或药用盐,其特征在于,所述X 6选自CR 8或N,且R 8的取代基选自H、卤素或C 1-3烷基,优选X 6为N。
  13. 根据权利要求1-12所述的任一项化合物、其互变异构体、氘代物或药用盐,其特征在于,所述X 7选自NR 9或CHR 9,优选NR 9
  14. 根据权利要求1-13所述的任一项化合物、其互变异构体、氘代物或药用盐,其特征在于,所述R 9选自-C 0-3亚烷基-C 6-14芳基、-C 0-3亚烷基-(5-14元杂芳基)、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基),所述-C 0-3亚烷基-C 6-14芳基、-C 0-3亚烷基-(5-14元杂芳基)、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)未被取代或进一步被1-4个选自卤素、C 1-3烷基、C 1-3烯基、C 1-3卤代烷基、-(CH 2) 0-3-S(R a) 5、C 1-3烷氧基、C 3-6环烷基、C 1-3烷基取代的C 3-6环烷基、-C 0-3亚烷基-苯基、-C 0-3亚烷基-N(R a) 2的取代基所取代,且每个R a独立地选自H或C 1-6烷基;优选R 9选自
    Figure PCTCN2021090082-appb-100022
    Figure PCTCN2021090082-appb-100023
    Figure PCTCN2021090082-appb-100024
  15. 根据权利要求1-14所述的任一项化合物、其互变异构体、氘代物或药用盐,其特征在于,所述
    Figure PCTCN2021090082-appb-100025
    选自
    Figure PCTCN2021090082-appb-100026
    Figure PCTCN2021090082-appb-100027
  16. 根据权利要求1-15中任一项所述的化合物、其互变异构体、氘代物或药用盐,其特征在于,选自式(Ia)至式(Ie)化合物,
    Figure PCTCN2021090082-appb-100028
    其中,取代基R 1-R 9、a、b如式(I)所定义。
  17. 一种化合物、其互变异构体、氘代物或药用盐,其中,所述化合物选自:
    Figure PCTCN2021090082-appb-100029
    Figure PCTCN2021090082-appb-100030
    Figure PCTCN2021090082-appb-100031
    Figure PCTCN2021090082-appb-100032
  18. 一种通式(II)所示的化合物、其互变异构体、氘代物或药用盐,
    Figure PCTCN2021090082-appb-100033
    其中,
    R 10选自C 1-6烷基、-C 0-3亚烷基-C 6-14芳基、-C 0-3亚烷基-(5-14元杂芳基)、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-O-C 0-3亚烷基-C 6-14芳基、-O-C 0-3亚烷基-(5-14元杂芳基)、-O-C 0-3亚烷基-C 3-14环烷基、-O-C 0-3亚烷基-(3-14元杂环烷基)、-NH-C 1-6烷基、-N(C 1-6烷基) 2、-NH-C 0-3亚烷基-C 6-14芳基、-NH-C 0-3亚烷基-(5-14元杂芳基)、-NH-C 0-3亚烷基-C 3-14环烷基、-NH-C 0-3亚烷基-(3-14元杂环烷基),R 10任选未取代或进一步被1-4个R 10a取代基所取代;优选R 10
    Figure PCTCN2021090082-appb-100034
    Figure PCTCN2021090082-appb-100035
    R 11选自H、氨基、卤素、C 1-3氰基、C 1-3羟基烷基、C 1-6烷基;优选为甲基或乙氰基;
    R 12为丙烯酰基或取代的丙烯酰基;优选为
    Figure PCTCN2021090082-appb-100036
    Figure PCTCN2021090082-appb-100037
    R 13选自H、羟基、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、-NH-C 1-6烷基、-N(C 1-6烷基) 2、氰基或卤素;优选为H;
    B环选自C 3-10环烷基、3-10元杂环基、C 6-12芳基、5-12元杂芳基;
    X 8的取代基选自CR 15或N;优选为N、卤素或CH;
    X 9选自C、CH或N;
    X 10或X 11独立地选自-O-(CH 2) 0-2-或CH 2;
    R 14、R 15或R 10a独立地选自氢、氧代、酰基、氰基、卤素、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-3-OR b、-(CH 2) 0-3-N(R b) 2、-(CH 2) 0-3-S(=O)R b、-(CH 2) 0-3-S(=O) 2R b、-(CH 2) 0-3-SR b、-(CH 2) 0-3-S(R a) 5、-(CH 2) 0-3-C(=O)N(R b) 2、C 2-3烯基、C 2-3炔基、-C 0-3亚烷基-C 3-14环烷基、-C 0-3亚烷基-(3-14元杂环烷基)、-C 0-3亚烷基-C 6-14芳基或-C 0-3亚烷基-(5-14元杂芳基),且每个R b独立地选自H、C 1-6烷基、C 1-6卤代烷基、C 3-14环烷基、3-14元杂环烷基、C 2-3烯基、C 2-3炔基、芳基或杂芳基;
    c或d独立地选自1、2、3或4;
    Figure PCTCN2021090082-appb-100038
    为双键或单键。
  19. 根据权利要求18中所述的化合物、其互变异构体、氘代物或药用盐,其特征在于,选自式(IIa)至(IId)化合物,
    Figure PCTCN2021090082-appb-100039
    其中,取代基如式(II)所定义。
  20. 一种化合物、其互变异构体、氘代物或药用盐,其中,所述化合物选自:
    Figure PCTCN2021090082-appb-100040
    Figure PCTCN2021090082-appb-100041
  21. 一种药物组合物,其特征在于,所述药物组合物含有治疗有效量的权利要求1-20任一项所述的化合物和至少一种药学上可接受的辅料。
  22. 权利要求1-20任一项所述的化合物或权利要求21所述的药物组合物在制备药物中的应用。
  23. 一种治疗和/或预防疾病的方法,包括向治疗对象施用治疗有效量的权利要求1-20任一项所述的化合物或权利要求21所述的药物组合物。
  24. 一种制备式(I)化合物的方法,该方法包括以下步骤:
    Figure PCTCN2021090082-appb-100042
    式(IA)化合物在碱性条件下与化合物X-R 2发生酰化反应,得到式(I)化合物、其互变异构体、氘代物或药用盐,其中,X为卤素;R 1-R 3、X 1-X 7、a或b如权利要求1中所定义。
PCT/CN2021/090082 2020-04-28 2021-04-27 稠环化合物及其在医药上的应用 WO2021218939A1 (zh)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202180029506.3A CN115427414A (zh) 2020-04-28 2021-04-27 稠环化合物及其在医药上的应用
US17/997,328 US20230203055A1 (en) 2020-04-28 2021-04-27 Fused ring compound and application thereof in medicine

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CNPCT/CN2020/087465 2020-04-28
CN2020087465 2020-04-28
CN202010844659.0 2020-08-20
CN202010844659 2020-08-20

Publications (1)

Publication Number Publication Date
WO2021218939A1 true WO2021218939A1 (zh) 2021-11-04

Family

ID=78373339

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/090082 WO2021218939A1 (zh) 2020-04-28 2021-04-27 稠环化合物及其在医药上的应用

Country Status (4)

Country Link
US (1) US20230203055A1 (zh)
CN (1) CN115427414A (zh)
TW (1) TW202144339A (zh)
WO (1) WO2021218939A1 (zh)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022235864A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors
WO2022235870A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors for the treatment of cancer
WO2023008462A1 (ja) 2021-07-27 2023-02-02 東レ株式会社 癌の治療及び/又は予防のための医薬品
WO2023114954A1 (en) 2021-12-17 2023-06-22 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
EP4227307A1 (en) 2022-02-11 2023-08-16 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109020901A (zh) * 2018-07-12 2018-12-18 长春海谱润斯科技有限公司 一种芳香胺类衍生物及其有机电致发光器件
WO2019099524A1 (en) * 2017-11-15 2019-05-23 Mirati Therapeutics, Inc. Kras g12c inhibitors
CN110234652A (zh) * 2016-12-22 2019-09-13 贝达药业股份有限公司 苯并咪唑衍生物及其制备方法和用途
CN110997668A (zh) * 2017-05-22 2020-04-10 美国安进公司 Kras g12c抑制剂及其使用方法

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110234652A (zh) * 2016-12-22 2019-09-13 贝达药业股份有限公司 苯并咪唑衍生物及其制备方法和用途
CN110997668A (zh) * 2017-05-22 2020-04-10 美国安进公司 Kras g12c抑制剂及其使用方法
WO2019099524A1 (en) * 2017-11-15 2019-05-23 Mirati Therapeutics, Inc. Kras g12c inhibitors
CN109020901A (zh) * 2018-07-12 2018-12-18 长春海谱润斯科技有限公司 一种芳香胺类衍生物及其有机电致发光器件

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022235864A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors
WO2022235870A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors for the treatment of cancer
WO2023008462A1 (ja) 2021-07-27 2023-02-02 東レ株式会社 癌の治療及び/又は予防のための医薬品
WO2023114954A1 (en) 2021-12-17 2023-06-22 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
EP4227307A1 (en) 2022-02-11 2023-08-16 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors

Also Published As

Publication number Publication date
CN115427414A (zh) 2022-12-02
US20230203055A1 (en) 2023-06-29
TW202144339A (zh) 2021-12-01

Similar Documents

Publication Publication Date Title
WO2021218939A1 (zh) 稠环化合物及其在医药上的应用
CN114761408B (zh) Kras g12c抑制剂及其在医药上的应用
JP6877407B2 (ja) Ntrk関連障害の治療に有用な化合物および組成物
WO2020259432A1 (zh) Kras-g12c抑制剂
WO2015127872A1 (zh) 2,4-二取代苯-1,5-二胺衍生物及其应用以及由其制备的药物组合物和药用组合物
WO2020216190A1 (zh) 一种喹唑啉化合物及其在医药上的应用
CN111499634B (zh) 一种喹唑啉化合物及其在医药上的应用
WO2020011246A1 (zh) 含苯环的化合物、其制备方法及应用
WO2017084640A1 (zh) 一种含氮杂环化合物及其制备方法与在抑制激酶活性中的应用
CN104736533B (zh) Vegfr3抑制剂
WO2022194066A1 (zh) Kras g12d抑制剂及其在医药上的应用
WO2016050165A1 (zh) 氮杂双环衍生物、其制法与医药上的用途
CN114685460A (zh) Kras g12c抑制剂及其在医药上的应用
WO2023098426A1 (zh) 一种7-(萘-1-基)吡啶并[4,3-d]嘧啶衍生物及其制备和应用
WO2022135590A1 (zh) 一类嘧啶并杂环类化合物、制备方法和用途
CN109384780A (zh) 一类四氢原小檗碱类化合物,其制备方法、用途及药物组合物
TW202210488A (zh) 吡嗪類衍生物及其在抑制shp2中的應用
WO2022166983A1 (zh) 杂芳基并哌啶类衍生物及其药物组合物和应用
WO2022105921A1 (zh) 一类嘧啶并杂环类化合物、制备方法和用途
WO2021164793A1 (zh) 用作激酶抑制剂的化合物及其应用
WO2023155927A1 (zh) Yap/taz与tead相互作用的抑制剂、其制备、药物组合物及应用
WO2023083269A1 (zh) 一种芳杂环类化合物及其应用
CN110461842A (zh) 作为tnf活性调节剂的稠合五环咪唑衍生物
TW202029963A (zh) 1,2,3,4-四氫喹㗁啉衍生物及其製備方法和應用
WO2020156162A1 (zh) Mek抑制剂及其在医药上的应用

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: 21795282

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21795282

Country of ref document: EP

Kind code of ref document: A1