WO2024027762A1 - 一种稠环化合物、其制备方法及其应用 - Google Patents

一种稠环化合物、其制备方法及其应用 Download PDF

Info

Publication number
WO2024027762A1
WO2024027762A1 PCT/CN2023/110759 CN2023110759W WO2024027762A1 WO 2024027762 A1 WO2024027762 A1 WO 2024027762A1 CN 2023110759 W CN2023110759 W CN 2023110759W WO 2024027762 A1 WO2024027762 A1 WO 2024027762A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
alkyl
independently
cycloalkyl
group
Prior art date
Application number
PCT/CN2023/110759
Other languages
English (en)
French (fr)
Inventor
罗会兵
周华勇
Original Assignee
上海艾力斯医药科技股份有限公司
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 上海艾力斯医药科技股份有限公司 filed Critical 上海艾力斯医药科技股份有限公司
Publication of WO2024027762A1 publication Critical patent/WO2024027762A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the invention relates to a fused ring compound, its preparation method and its application.
  • RAS protein is a guanine trinucleotide phosphate (GTP)-binding protein with a molecular weight of 21 kDa located on the cell membrane and composed of 188 or 189 amino acids.
  • GTP guanine trinucleotide phosphate
  • the activity of RAS protein is regulated by binding to GTP or guanine dinucleotide phosphate (GDP). When bound to GDP, it is in an "inactive" state, and when bound to GTP, it is in an “activated” state.
  • GTP guanine trinucleotide phosphate
  • GTPase activating proteins can enhance the GTP hydrolysis function of RAS protein, while guanine nucleotide exchange Factors (GEF) can catalyze the exchange of nucleotides (GTP for GDP).
  • GEF guanine nucleotide exchange Factors
  • SOS1 Non of Sevenless 1
  • RAS can promote RAS to release GDP and combine with GTP, thereby activating RAS.
  • RAS After RAS is activated, it can activate multiple downstream signaling pathways, including MAPK signaling pathway and PI3K signaling pathway. These signaling pathways play an important role in promoting cell differentiation, proliferation, and survival.
  • RAS mutations are genetic drivers of many cancers and are found in 20%-30% of human tumors, such as lung, colorectal, and pancreatic cancers.
  • the RAS gene family includes KRAS, NRAS and HRAS.
  • KRAS mutations exist in a variety of tumors, such as lung adenocarcinoma (32%), colorectal cancer (41%), and pancreatic cancer (86%).
  • KRAS mutations are the most common G12 mutation in codon 12, for example, Among lung adenocarcinomas, colorectal cancers and pancreatic cancers with KRAS mutations, G12 mutations account for 85%, 68% and 91% respectively; the frequency of HRAS mutations and NRAS mutations is relatively low and mainly occurs in melanoma, leukemia and thyroid cancer. among other types of cancer.
  • RAS protein abnormal activation of RAS protein (such as gene mutation, amplification and overexpression, etc.) is also closely related to resistance to some anti-tumor drugs, such as EGFR monoclonal antibodies and EGFR small molecule inhibitors. Therefore, RAS-related signaling pathways have become important anti-tumor targets.
  • the guanine nucleotide exchange factor of the RAS family involved in cancer-related signaling pathways is mainly SOS1. Reducing the expression of SOS1 can significantly inhibit the proliferation and survival of KRAS-mutated cancer cells. Since SOS1 is a common node for multiple activations of RAS signaling pathways, and almost all growth factor receptors activate RAS signaling pathways through SOS1, SOS1 inhibitors have the potential to become broad-spectrum anticancer drugs. The signaling pathways activated by SOS1 also play an important role in other mutated types of cancer.
  • SOS1 can interact with the adapter protein Grb2 to form a SOS1/Grb2 complex, which binds to activated receptor tyrosine kinases (such as EGFR, HER2, Erbb4, TRKA, TRKB, TRKC, RET and AXL, etc.).
  • SOS1 can also be recruited to phosphorylated cell surface receptors, such as T cell receptors, B cell receptors, and monocyte colony-stimulating factor receptors.
  • the localization of SOS1 on the cell membrane allows SOS1 to better promote the activation of RAS family proteins and activate downstream signaling pathways.
  • SOS1 is also involved in the activation of other GTP hydrolases, such as RAC1, etc.
  • RAC1 is also related to the pathogenesis of various human cancers and other diseases.
  • SOS1 mutations have been found in lung adenocarcinoma, embryonal rhabdomyosarcoma, and cutaneous granulosa cell tumors, and SOS1 overexpression has been found in bladder cancer and prostate cancer.
  • inherited SOS1 mutations are also associated with the pathogenesis of RAS lesions, including Noonan syndrome and cardio-facial-cutaneous syndrome. Syndrome and type 1 hereditary gingival fibroma, etc.
  • the technical problem to be solved by the present invention is the defects of existing SOS1 inhibitors such as single structure.
  • the present invention provides a fused ring compound, its preparation method and its application. This type of compound has good inhibitory activity against SOS1.
  • the present invention provides a fused ring compound shown in formula I, its stereoisomer or its pharmaceutically acceptable salt
  • Ring A is C 6-12 aryl, 5-10 membered heteroaryl, C 3-7 cycloalkyl, C 3-7 cycloalkenyl or 3-7 membered heterocyclyl;
  • R 1a , R 1b , R 1c , R 1d , R 1e , R 1f , R 1g , R 1h and R 1i is independently deuterium, hydroxyl, halogen, -N(R 7 ) 2 , -SR 9 , Nitro, cyano, C 1-6 alkyl, C 1-6 alkyl-O-, C 2-6 alkenyl, C 2-6 alkynyl, C 1 substituted by one or more R 1-a -6 alkyl, C 1-6 alkyl-O- substituted by one or more R 1-b , C 2-6 alkenyl substituted by one or more R 1-c , substituted by one or more R 1-d substituted C 2-6 alkynyl, C 3-7 cycloalkyl, C 3-7 cycloalkyl -O-, 3-7-membered heterocyclyl, 3-7-membered heterocyclyl-O-, C 3-7 cycloalkyl substituted by one or more R
  • Each R 1-a , R 1-b , R 1-c , R 1-d , R 1 -e , R 1-f , R 1-g , R 1-h and R 1-i is each independently Deuterium, hydroxyl, halogen, -N(R 7 ) 2 , -SR 9 , nitro, cyano, C 1-6 alkyl, C 1-6 alkyl-O-, C 2-6 alkenyl, C 2 -6 alkynyl, C 3-7 cycloalkyl, C 3-7 cycloalkenyl or 3-7 membered heterocyclyl;
  • Ring D is C 6-12 aryl, 5-6 membered heteroaryl, C 3-7 cycloalkyl, C 3-7 cycloalkenyl or 3-7 membered heterocyclyl;
  • R 2 is absent, hydrogen, deuterium, hydroxyl, halogen, amino, cyano, oxo, nitro, C 1-6 alkyl, C 1-6 alkyl-O-, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 cycloalkyl, C 3-7 cycloalkenyl or 3-7 membered heterocyclyl;
  • R 3 is hydrogen, deuterium, C 1-6 alkyl, C 1-6 alkyl substituted by one or more R 3a , C 2-6 alkenyl, C 2-6 substituted by one or more R 3b Alkenyl, C 2-6 alkynyl, C 2-6 alkynyl substituted by one or more R 3c , C 3-7 cycloalkyl, C 3-7 cycloalkyl substituted by one or more R 3d , 3-7 membered heterocyclyl, 3-7 membered heterocyclyl substituted by one or more R 3e , C 6-12 aryl, C 6-12 aryl substituted by one or more R 3f , 5 -10-membered heteroaryl, 5-10-membered heteroaryl substituted by one or more R 3g , C 3-7 cycloalkenyl or C 3-7 cycloalkenyl substituted by one or more R 3h ; when When there are multiple substituents, they may be the same or different;
  • R 3a , R 3b , R 3c , R 3d , R 3e , R 3f , R 3g and R 3h is independently deuterium, hydroxyl, halogen, -N(R 7 ) 2 , cyano, nitro, C 1-6 alkyl, C 1-6 alkyl substituted by one or more R 3-a , C 1-6 alkyl-O-, C 1-6 alkyl substituted by one or more R 3-b Base -O-, C 2-6 alkenyl, C 2-6 alkynyl, C 2-6 alkenyl substituted by one or more R 3-c , C 2 substituted by one or more R 3-d -6 alkynyl, C 3-7 cycloalkyl, C 3-7 cycloalkyl substituted by one or more R 3-e , 3-7 membered heterocyclyl, substituted by one or more R 3-f 3-7 membered heterocyclyl, C 6-12 aryl, C 6-12 aryl substituted by
  • X is a connecting bond, -O-, -(C 1 -C 4 alkylene)-, -O-(C 1 -C 4 alkylene)-, -(C 2 -C 4 alkenylene)- or -(C 2 -C 6 alkynylene)-;
  • R 4 is hydrogen, C 1-6 alkyl, C 1-6 alkyl-O-, cyano group, halogen, hydroxyl, -N(R 7 ) 2 , 3-7 membered heterocyclic group, substituted by one or more R 4a- substituted 3-7-membered heterocyclyl, C 3-7 cycloalkyl, or C 3-7 cycloalkyl substituted by one or more R 4b ; when there are multiple substituents, they may be the same or different;
  • Each R 4a and R 4b is independently hydrogen, deuterium, hydroxyl, halogen, amino, cyano, nitro, amino protecting group, C 1-6 alkyl, or C 1- substituted with one or more deuterium 6 alkyl;
  • R 5 is absent, oxo, hydrogen, deuterium, hydroxyl, halogen, amino, cyano, nitro, C 1-6 alkyl, C 1-6 alkyl substituted by one or more R 5a , C 1 -6 alkyl-O-, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 cycloalkyl, C 3-7 cycloalkyl substituted by one or more R 5b , 3-7 Member heterocyclyl, 3-7-membered heterocyclyl substituted by one or more R 5c , C 3-7 cycloalkenyl or C 3-7 cycloalkenyl substituted by one or more R 5d ; when substituent When there are multiple, they are the same or different;
  • Each R 5a , R 5b and R 5c is independently deuterium, C 1-6 alkyl, hydroxyl, halogen, amino or cyano;
  • Each R 7 is independently hydrogen, C 1-6 alkyl, C 1-6 alkyl substituted with one or more R 7b , C 3-7 cycloalkyl, C 3-7 cycloalkenyl, or amino protected group, or two R 7 together with the attached nitrogen atom form a 3-7-membered heterocyclyl group or a 3-7-membered heterocyclyl group substituted by one or more R 7a ; when there are multiple substituents, they are the same or different;
  • Each R 7a and R 7b is independently deuterium, C 1-6 alkyl, halogen, hydroxy, amino, cyano, C 1-6 alkyl-O-, C 2-6 alkenyl, or C 2-6 Alkynyl;
  • Each R 8 and R 9 is independently hydrogen, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-7 cycloalkyl, C 3-7 cycloalkenyl, or 3-7 membered heterocyclyl;
  • R 10 is hydrogen, deuterium, hydroxyl, halogen, amino, cyano, C 1-6 alkyl-O-, C 2-6 alkenyl, C 2-6 alkynyl or C 1-6 alkyl;
  • R 11 is cyano, C 1-6 alkyl or C 1-6 alkyl substituted by one or more R 11a ; when there are multiple substituents, they are the same or different;
  • Each R 11a is independently deuterium, halogen, or hydroxyl
  • n 0, 1, 2, 3, 4 or 5;
  • p 1, 2, 3, 4, 5 or 6;
  • the 3-7-membered heterocyclic group in the 3-7-membered heterocyclic group and the 3-7-membered heterocyclic group substituted by a substituent is each independently "the heteroatom species is independently selected from N, O and S, and the heteroatom The number of 3-7-membered heterocyclic groups is 1, 2 or 3";
  • the 5-10-membered heteroaryl group in the 5-10-membered heteroaryl group and the 5-10-membered heteroaryl group substituted by a substituent is each independently "the heteroatom species is independently selected from N, O and S, and the heteroatom The number of 5-10 membered heteroaryl groups is 1, 2 or 3";
  • the 5-6-membered heteroaryl group is "a 5-6-membered heteroaryl group whose heteroatom species is independently selected from N, O and S, and the number of heteroatoms is 1, 2 or 3";
  • the amino protecting group is tert-butoxycarbonyl, benzyloxycarbonyl, fluorenylmethoxycarbonyl, allyloxycarbonyl, phthaloyl, benzyl, p-methoxybenzyl, trityl or p-toluene sulfonyl;
  • the fused ring compound shown in formula I is or mixtures thereof.
  • fused ring compound shown in formula I its stereoisomer or its pharmaceutically acceptable salt, certain groups have the following definitions, and the definitions of unmentioned groups are as follows: described in any aspect of the invention (the content of this paragraph is hereinafter referred to as "in a certain aspect").
  • R 4 is C 1-6 alkyl, C 1-6 alkyl-O-, cyano, halogen, hydroxyl, -N(R 7 ) 2 , 3- 7-membered heterocyclyl, 3-7-membered heterocyclyl substituted by one or more R 4a , C 3-7 cycloalkyl or C 3-7 cycloalkyl substituted by one or more R 4b ; when substituted When there are multiple bases, they can be the same or different.
  • R 6 is oxo;
  • L is a connecting bond, it means that R 3 is directly connected to the D ring;
  • R 3 is One or more R 3e substituted 3-7-membered heterocyclic groups; when there are multiple substituents, they may be the same or different; each R 3e is independently deuterium, hydroxyl, halogen, -N(R 7 ) 2 , Cyano, nitro, C 1-6 alkyl, C 1-6 alkyl substituted by one or more R 3-a , C 1-6 alkyl-O-, substituted by one or more R 3-b Substituted C 1-6 alkyl-O-, C 2
  • R 6 is oxo;
  • L is a connecting bond; when L is a connecting bond, it means that R 3 is directly connected to the D ring;
  • R 3 is a 3-7-membered heterogeneous compound substituted by one or more R 3e Ring group; when there are multiple substituents, they may be the same or different;
  • -CH 2 -CH CH-CH 2 -CH 3
  • -CH CH-CH(CH 3 ) 2
  • the C 2-6 alkynyl groups in the 6- alkynyl group are each independently ethynyl (-C ⁇ CH), propynyl (-C ⁇ C-CH 3 ), -C ⁇ C-
  • C 3-7 cycloalkyl in ring A, C 3-7 cycloalkyl in R 1 , C 3-7 cycloalkyl in the C 3-7 cycloalkyl substituted by a substituent The two R 1 and the connected ring atom together form a C 3-7 cycloalkyl group. The two R 1 and the connected ring atom together form C 3 in the C 3-7 cycloalkyl substituted by a substituent.
  • Ring group specifically, when R 3 is a 4-7-membered monocyclic heterocyclyl group, the 4-7-membered monocyclic heterocyclyl group can be piperidinyl, tetrahydropyranyl, piperazinyl, aza cyclobutyl, morpholinyl or pyrrolidinyl; when R 3 is a 5-7-membered bridged heterocyclyl, the 5-7-membered bridged heterocyclyl can be When R 3 is a 5-7 membered spirocyclic heterocyclyl, the 5-7 membered spirocyclic heterocyclyl can be For example
  • R 4 the number of heteroatoms of the 3-7-membered heterocyclyl in the 3-7-membered heterocyclyl or the 3-7-membered heterocyclyl substituted by one or more R 4a Independently 1, 2 or 3; the heteroatom species of the 3-7-membered heterocyclic group in the 3-7-membered heterocyclic group and the 3-7-membered heterocyclic group substituted by one or more R 4a Independently selected from one or more of N, O and S; the 3-7-membered heterocyclic group, the 3-7-membered heterocyclic group substituted by one or more R 4a
  • the groups are each independently "a 3-7-membered heterocyclyl group with heteroatom species independently selected from N, O and S, and the number of heteroatoms is 1, 2 or 3"; further, the 3-7-membered heterocyclic group The number of heteroatoms of the 3-7-membered heterocyclic group in the heterocyclyl group or the 3-7-membered
  • the two R 7 together with the attached nitrogen atom form a 3-7 membered heterocyclyl group, a 3-7 membered heterocyclyl group substituted by one or more R 7a
  • the number of heteroatoms of the 3-7-membered heterocyclic group is independently 1 or 2; the two R 7 together with the connected nitrogen atoms form a 3-7-membered heterocyclic group, which is substituted by one or more R 7a
  • the heteroatom species of the 3-7-membered heterocyclic group in the 3-7-membered heterocyclic group are independently selected from one or two types of N and O; the two R 7 together with the connected nitrogen atom form 3
  • the 3-7-membered heterocyclyl groups in the 7-membered heterocyclyl group and the 3-7-membered heterocyclyl group substituted by one or more R 7a are each independently "a type of heteroatom species independently selected from N and O species.” Or two kinds, 4-7 membered heterocyclic groups with 1 or 2 heteroatom
  • the 3-7-membered heterocyclic group is "a 6-membered heterocyclic group with N heteroatom type and 1 or 2 heteroatoms", for example, 1,2 -Dihydropyridyl, another example is "c" means that the atom is located in the meta position of the N atom of the B ring.
  • R 3c , R 3d , R 3e , R 3f , R 3g and R 3h are each independently It is phenyl or naphthyl; for example: phenyl.
  • Ring A, R 1 , R 1a , R 1b , R 1c , R 1d , R 1e , R 1f , R 1g , R 1h , R 1i , R 3 , R 3a , R 3b , R 3c , R 3d , R 3e , R 3f , R 3g and R 3h , the 5-10-membered heteroaryl group in the 5-10-membered heteroaryl group and the 5-10-membered heteroaryl group substituted by a substituent are each independently Ground is "a 5- to 8-membered heteroaryl group with 1, 2, or 3 heteroatoms, and the heteroatom species are independently selected from N, O, and S.”
  • the -(C 1 -C 4 alkylene)-, -O-(C 1 -C 4 alkylene)- in -(C 1 -C 4 alkylene)- )- are each independently methylene, ethylene, n-propylene or isopropylene, for example: -CH 2 -, -CH 2 CH 2 -, -CH(CH 3 )-, -CH 2 CH 2 CH 2 -, -CH(CH 3 )CH 2 -, -CH(CH 2 CH 3 )- or -C(CH 3 ) 2 -, preferably -CH 2 -, -CH 2 CH 2 -, -CH( CH 3 )- or -CH 2 CH 2 CH 2 -; further, "-O-(C 1 -C 4 alkylene)-" in "C 1 -C 4 alkylene” is connected to R 4 .
  • -CH CH-CH 2 -; further, the -(C 2 -C 4 alkenylene)- can be The f end is connected to R 4 .
  • the -(C 2 -C 6Alkynylene )- can be -C ⁇ C-*, -C ⁇ C-CH 2 -* or -C ⁇ CC(CH 3 ) 2 -*, where the * end is connected to R 4 .
  • Ring A is C 6-12 aryl; preferably, Ring A is phenyl.
  • each R 1 is independently halogen, hydroxyl, -N(R 7 ) 2 , C 1-6 alkyl, cyano, C 1-6 alkyl-O-, C 2-6 alkene group, C 2-6 alkynyl, C 1-6 alkyl substituted by one or more R 1a or C 1-6 alkyl-O- substituted by one or more R 1b ; preferably, each R 1 is independently halogen, C 1-6 alkyl, cyano or C 1-6 alkyl substituted by one or more R 1a ; when there are multiple substituents, they are the same or different.
  • each R 1a and R 1b is independently halogen; preferably, each R 1a and R 1b is independently F or Cl.
  • n 0, 1, 2 or 3; preferably, m is 2.
  • R 2 is hydrogen, hydroxyl, halogen, amino, cyano, C 1-6 alkyl or C 1-6 alkyl-O-; preferably, R 2 is hydrogen.
  • L is the connecting key
  • R 3 is C 3-7 cycloalkyl, C 3-7 cycloalkyl substituted by one or more R 3d , 3-7 membered heterocyclyl, substituted by one or more R 3e 3-7 membered heterocyclyl, C 3-7 cycloalkenyl or C 3-7 cycloalkenyl substituted by one or more R 3h ; preferably, R 3 is C 3-7 cycloalkyl, substituted by one Or C 3-7 cycloalkyl substituted by R 3d , 3-7 membered heterocyclyl substituted by one or more R 3e , C 3-7 cycloalkenyl or C substituted by one or more R 3h 3-7 cycloalkenyl; more preferably, R 3 is a 3-7-membered heterocyclyl substituted by one or more R 3e ; when there are multiple substituents, they are the same or different.
  • each R 3-a , R 3-b and R 3-j are each independently deuterium, hydroxyl, halogen or C 3-7 cycloalkyl; further, each R 3-a and Each R 3-b is independently deuterium, hydroxyl, halogen or C 3-7 cycloalkyl; further, each R 3-b is independently deuterium, hydroxyl or halogen.
  • X is a connecting bond, -(C 1 -C 4 alkylene)-, -O-(C 1 -C 4 alkylene)-, -(C 2 -C 4 alkenylene) - or -(C 2 -C 6 alkynylene)-; preferably, "C 1 -C 4 alkylene” in -O-(C 1 -C 4 alkylene)- is connected to R 4 ; preferably Where , _ _ _ _ _ (C 2 -C 6 alkynylene)-; preferably, "C 1 -C 4 alkylene” in -O-(C 1 -C 4 alkylene)- is connected to R 4 .
  • R 4 is hydrogen, C 1-6 alkyl, C 1-6 alkyl-O-, cyano, halogen, hydroxyl, -N(R 7 ) 2 , 3-7 membered heterocyclyl , 3-7 membered heterocyclyl substituted by one or more R 4a , C 3-7 cycloalkyl or C 3-7 cycloalkyl substituted by one or more R 4b ;
  • R 4 is hydrogen , C 1-6 alkyl, cyano group, hydroxyl, -N(R 7 ) 2 , 3-7 membered heterocyclic group, 3-7 membered heterocyclic group substituted by one or more R 4a or substituted by one or more R 4a C 3-7 cycloalkyl substituted by R 4b ; more preferably, R 4 is hydrogen, C 1-6 alkyl, cyano, hydroxyl, -N(R 7 ) 2 , substituted by one or more R 4a 3-7 membered heterocycl
  • each R 4a and R 4b is independently hydroxyl, halogen, amino, C 1-6 alkyl, or C 1-6 alkyl substituted with one or more deuterium; preferably, each R 4a and R 4b are each independently amino, C 1-6 alkyl, or C 1-6 alkyl substituted by one or more deuterium; more preferably, each R 4a and R 4b are each independently C 1 -6 alkyl or amino ; further preferably, each R 4a is independently C 1-6 alkyl.
  • R 5 is hydrogen, hydroxyl, halogen, amino, cyano or C 1-6 alkyl; preferably, R 5 is hydrogen or C 1-6 alkyl; more preferably, R 5 is C 1-6 alkyl.
  • each R 7 is independently hydrogen, C 1-6 alkyl, or C 1-6 alkyl substituted with one or more R 7b , or two R 7 together with the attached nitrogen atom Forming a 3-7-membered heterocyclyl group or a 3-7-membered heterocyclyl group substituted by one or more R 7a ; further, each R 7 is independently hydrogen, C 1-6 alkyl or substituted by one or more R 7a R 7b substituted C 1-6 alkyl, or two R 7 together with the attached nitrogen atom form 3- 7-membered heterocyclyl; further, each R 7 is independently hydrogen or C 1-6 alkyl; when there are multiple substituents, they may be the same or different.
  • each R 7b is independently deuterium, halogen, hydroxyl or cyano; preferably, each R 7b is independently deuterium or hydroxyl; more preferably, each R 7b is independently deuterium.
  • each R 9 is independently hydrogen or C 1-6 alkyl; preferably, each R 9 is independently C 1-6 alkyl.
  • Ring D is a 3-7 membered heterocyclyl.
  • R 10 is C 1-6 alkyl; preferably, R 10 is methyl.
  • R 11 is C 1-6 alkyl; preferably, R 11 is methyl.
  • Ring A is C 6-12 aryl
  • Each R 1 is independently halogen, hydroxyl, -N(R 7 ) 2 , C 1-6 alkyl, cyano, C 1-6 alkyl-O-, C 2-6 alkenyl, C 2-6 Alkynyl, C 1-6 alkyl substituted by one or more R 1a or C 1-6 alkyl-O- substituted by one or more R 1b ; when the substituents are multiple, they are the same or different;
  • Each R 1a and R 1b is independently halogen
  • n 0, 1, 2 or 3;
  • R 2 is hydrogen, hydroxyl, halogen, amino, cyano, C 1-6 alkyl or C 1-6 alkyl-O-;
  • R 3 is C 3-7 cycloalkyl, C 3-7 cycloalkyl substituted by one or more R 3d , 3-7 membered heterocyclyl, 3-7 membered heterocyclic group substituted by one or more R 3e Ring group, C 3-7 cycloalkenyl or C 3-7 cycloalkenyl substituted by one or more R 3h ; when there are multiple substituents, they may be the same or different;
  • Each R 3-a , R 3-b and R 3-j is independently deuterium, hydroxyl, halogen or C 3-7 cycloalkyl;
  • X is a connecting bond, -O-, -(C 1 -C 4 alkylene)-, -O-(C 1 -C 4 alkylene)-, -(C 2 -C 4 alkenylene)- or -(C 2 -C 4 alkynylene)-; preferably, "C 1 -C 4 alkylene" in -O-(C 1 -C 4 alkylene)- is connected to R 4 ;
  • R 4 is hydrogen, C 1-6 alkyl, C 1-6 alkyl-O-, cyano group, halogen, hydroxyl, -N(R 7 ) 2 , 3-7 membered heterocyclic group, substituted by one or more R 4a- substituted 3-7-membered heterocyclyl, C 3-7 cycloalkyl, or C 3-7 cycloalkyl substituted by one or more R 4b ; when there are multiple substituents, they may be the same or different;
  • Each R 4a and R 4b is independently hydroxyl, halogen, amino, C 1-6 alkyl, or C 1-6 alkyl substituted with one or more deuterium;
  • R 5 is hydrogen, hydroxyl, halogen, amino, cyano or C 1-6 alkyl
  • Each R 7 is independently hydrogen, C 1-6 alkyl, or C 1-6 alkyl substituted by one or more R 7b , or two R 7 together with the attached nitrogen atom form a 3-7 membered heterogeneous A cyclic group or a 3-7-membered heterocyclic group substituted by one or more R 7a ; when there are multiple substituents, they are the same or different;
  • Each R 7a and R 7b is independently deuterium, halogen, hydroxyl or cyano;
  • Each R 9 is independently hydrogen or C 1-6 alkyl
  • Ring D is a 3-7 membered heterocyclic group
  • R 10 is C 1-6 alkyl
  • R 11 is C 1-6 alkyl
  • the 3-7-membered heterocyclic group in the 3-7-membered heterocyclic group and the 3-7-membered heterocyclic group substituted by a substituent is each independently "the heteroatom species is independently selected from N, O and S, and the heteroatom The number of 3-7-membered heterocyclic groups is 1, 2 or 3.”
  • Ring A is C 6-12 aryl
  • Each R 1 is independently halogen, hydroxyl, -N(R 7 ) 2 , C 1-6 alkyl, cyano, C 1-6 alkyl-O-, C 2-6 alkenyl, C 2-6 Alkynyl, C 1-6 alkyl substituted by one or more R 1a or C 1-6 alkyl-O- substituted by one or more R 1b ; when the substituents are multiple, they are the same or different;
  • Each R 1a and R 1b is independently halogen
  • n 0, 1, 2 or 3;
  • R 2 is hydrogen, hydroxyl, halogen, amino, cyano, C 1-6 alkyl or C 1-6 alkyl-O-;
  • R 3 is C 3-7 cycloalkyl, C 3-7 cycloalkyl substituted by one or more R 3d , 3-7 membered heterocyclyl, 3-7 membered heterocyclic group substituted by one or more R 3e Ring group, C 3-7 cycloalkenyl or C 3-7 cycloalkenyl substituted by one or more R 3h ; when there are multiple substituents, they may be the same or different;
  • Each R 3-a and R 3-b is independently deuterium, hydroxyl or halogen
  • X is a connecting bond, -(C 1 -C 4 alkylene)-, -O-(C 1 -C 4 alkylene)-, -(C 2 -C 4 alkenylene)- or -(C 2 -C 4 alkynylene)-; preferably, "C 1 -C 4 alkylene" in -O-(C 1 -C 4 alkylene)- is connected to R 4 ;
  • R 4 is hydrogen, C 1-6 alkyl, C 1-6 alkyl-O-, cyano group, halogen, hydroxyl, -N(R 7 ) 2 , 3-7 membered heterocyclic group, substituted by one or more R 4a- substituted 3-7-membered heterocyclyl, C 3-7 cycloalkyl, or C 3-7 cycloalkyl substituted by one or more R 4b ; when there are multiple substituents, they may be the same or different;
  • Each R 4a and R 4b is independently hydroxyl, halogen, amino, C 1-6 alkyl, or C 1-6 alkyl substituted with one or more deuterium;
  • R 5 is hydrogen, hydroxyl, halogen, amino, cyano or C 1-6 alkyl
  • Each R 7 is independently hydrogen or C 1-6 alkyl
  • Each R 9 is independently hydrogen or C 1-6 alkyl
  • Ring D is a 3-7 membered heterocyclic group
  • R 10 is C 1-6 alkyl
  • R 11 is C 1-6 alkyl.
  • Ring A is C 6-12 aryl
  • Each R 1 is independently halogen, hydroxyl, -N(R 7 ) 2 , C 1-6 alkyl, cyano, C 1-6 alkyl-O-, C 2-6 alkenyl, C 2-6 Alkynyl, C 1-6 alkyl substituted by one or more R 1a or C 1-6 alkyl-O- substituted by one or more R 1b ; when the substituents are multiple, they are the same or different;
  • Each R 1a and R 1b is independently halogen
  • n 0, 1, 2 or 3;
  • R 2 is hydrogen, hydroxyl, halogen, amino, cyano, C 1-6 alkyl or C 1-6 alkyl-O-;
  • R 3 is C 3-7 cycloalkyl, C 3-7 cycloalkyl substituted by one or more R 3d , 3-7 membered heterocyclyl, 3-7 membered heterocyclic group substituted by one or more R 3e Ring group, C 3-7 cycloalkenyl or C 3-7 cycloalkenyl substituted by one or more R 3h ; when there are multiple substituents, they may be the same or different;
  • Each R 3-a , R 3-b and R 3-j is independently deuterium, hydroxyl, halogen or C 3-7 cycloalkyl;
  • X is a connecting bond, -O-, -(C 1 -C 4 alkylene)-, -O-(C 1 -C 4 alkylene)-, -(C 2 -C 4 alkenylene)- or -(C 2 -C 6 alkynylene)-; preferably, "C 1 -C 4 alkylene" in -O-(C 1 -C 4 alkylene)- is connected to R 4 ;
  • R 4 is hydrogen, C 1-6 alkyl, C 1-6 alkyl-O-, cyano group, halogen, hydroxyl, -N(R 7 ) 2 , 3-7 membered heterocyclic group, substituted by one or more R 4a- substituted 3-7-membered heterocyclyl, C 3-7 cycloalkyl, or C 3-7 cycloalkyl substituted by one or more R 4b ; when there are multiple substituents, they may be the same or different;
  • Each R 4a and R 4b is independently hydroxyl, halogen, amino, C 1-6 alkyl, or C 1-6 alkyl substituted with one or more deuterium;
  • R 5 is hydrogen, hydroxyl, halogen, amino, cyano or C 1-6 alkyl
  • Each R 7 is independently hydrogen, C 1-6 alkyl, or C 1-6 alkyl substituted by one or more R 7b , or two R 7 together with the attached nitrogen atom form a 3-7 membered heterogeneous A cyclic group or a 3-7-membered heterocyclic group substituted by one or more R 7a ; when there are multiple substituents, they are the same or different;
  • Each R 7a and R 7b is independently deuterium, halogen, hydroxyl or cyano;
  • Each R 9 is independently hydrogen or C 1-6 alkyl
  • Ring D is a 3-7 membered heterocyclic group
  • R 10 is C 1-6 alkyl
  • R 11 is C 1-6 alkyl
  • the 3-7-membered heterocyclic group in the 3-7-membered heterocyclic group and the 3-7-membered heterocyclic group substituted by a substituent is each independently "the heteroatom species is independently selected from N, O and S, and the heteroatom The number of 3-7-membered heterocyclic groups is 1, 2 or 3.”
  • Ring A is C 6-12 aryl
  • Each R 1 is independently halogen, hydroxyl, -N(R 7 ) 2 , C 1-6 alkyl, cyano, C 1-6 alkyl-O-, C 2-6 alkenyl, C 2-6 Alkynyl, by C 1-6 alkyl substituted by one or more R 1a or C 1-6 alkyl-O- substituted by one or more R 1b ; when the substituents are multiple, they are the same or different;
  • Each R 1a and R 1b is independently halogen
  • n 0, 1, 2 or 3;
  • R 2 is hydrogen, hydroxyl, halogen, amino, cyano, C 1-6 alkyl or C 1-6 alkyl-O-;
  • R 3 is C 3-7 cycloalkyl, C 3-7 cycloalkyl substituted by one or more R 3d , 3-7 membered heterocyclyl substituted by one or more R 3e , C 3-7 ring Alkenyl or C 3-7 cycloalkenyl substituted by one or more R 3h ; when there are multiple substituents, they are the same or different;
  • Each R 3-a , R 3-b and R 3-j is independently deuterium, hydroxyl, halogen or C 3-7 cycloalkyl;
  • X is a connecting bond, -O-, -(C 1 -C 4 alkylene)-, -O-(C 1 -C 4 alkylene)-, -(C 2 -C 4 alkenylene)- or -(C 2 -C 6 alkynylene)-; preferably, "C 1 -C 4 alkylene" in -O-(C 1 -C 4 alkylene)- is connected to R 4 ;
  • R 4 is hydrogen, C 1-6 alkyl, C 1-6 alkyl-O-, cyano group, halogen, hydroxyl, -N(R 7 ) 2 , 3-7 membered heterocyclic group, substituted by one or more R 4a- substituted 3-7-membered heterocyclyl, C 3-7 cycloalkyl, or C 3-7 cycloalkyl substituted by one or more R 4b ; when there are multiple substituents, they may be the same or different;
  • Each R 4a and R 4b is independently hydroxyl, halogen, amino, C 1-6 alkyl, or C 1-6 alkyl substituted with one or more deuterium;
  • R 5 is hydrogen, hydroxyl, halogen, amino, cyano or C 1-6 alkyl
  • Each R 7 is independently hydrogen, C 1-6 alkyl, or C 1-6 alkyl substituted by one or more R 7b , or two R 7 together with the attached nitrogen atom form a 3-7 membered heterogeneous A cyclic group or a 3-7-membered heterocyclic group substituted by one or more R 7a ; when there are multiple substituents, they are the same or different;
  • Each R 7a and R 7b is independently deuterium, halogen, hydroxyl or cyano;
  • Each R 9 is independently hydrogen or C 1-6 alkyl
  • Ring D is a 3-7 membered heterocyclic group
  • R 10 is C 1-6 alkyl
  • R 11 is C 1-6 alkyl
  • the 3-7-membered heterocyclic group in the 3-7-membered heterocyclic group and the 3-7-membered heterocyclic group substituted by a substituent is each independently "the heteroatom species is independently selected from N, O and S, and the heteroatom The number of 3-7-membered heterocyclic groups is 1, 2 or 3.”
  • Ring A is C 6-12 aryl
  • Each R 1 is independently halogen, hydroxyl, -N(R 7 ) 2 , C 1-6 alkyl, cyano, C 1-6 alkyl-O-, C 2-6 alkenyl, C 2-6 Alkynyl, C 1-6 alkyl substituted by one or more R 1a or C 1-6 alkyl-O- substituted by one or more R 1b ; when the substituent is multiple, the same or different;
  • Each R 1a and R 1b is independently halogen
  • n 0, 1, 2 or 3;
  • R 2 is hydrogen, hydroxyl, halogen, amino, cyano, C 1-6 alkyl or C 1-6 alkyl-O-;
  • R 3 is C 3-7 cycloalkyl, C 3-7 cycloalkyl substituted by one or more R 3d , 3-7 membered heterocyclyl, 3-7 membered heterocyclic group substituted by one or more R 3e Ring group, C 3-7 cycloalkenyl or C 3-7 cycloalkenyl substituted by one or more R 3h ; when there are multiple substituents, they may be the same or different;
  • Each R 3-a , R 3-b and R 3-j is independently deuterium, hydroxyl, halogen or C 3-7 cycloalkyl;
  • X is a connecting bond, -O-, -(C 1 -C 4 alkylene)-, -O-(C 1 -C 4 alkylene)-, -(C 2 -C 4 alkenylene)- or -(C 2 -C 6 alkynylene)-; preferably, "C 1 -C 4 alkylene" in -O-(C 1 -C 4 alkylene)- is connected to R 4 ;
  • R 4 is -N(R 7 ) 2 , 3-7 membered heterocyclyl, 3-7 membered heterocyclyl substituted by one or more R 4a , C 3-7 cycloalkyl or substituted by one or more R 4b substituted C 3-7 cycloalkyl; when there are multiple substituents, they are the same or different;
  • Each R 4a and R 4b is independently hydroxyl, halogen, amino, C 1-6 alkyl, or C 1-6 alkyl substituted with one or more deuterium;
  • R 5 is hydrogen, hydroxyl, halogen, amino, cyano or C 1-6 alkyl
  • Each R 7 is independently hydrogen, C 1-6 alkyl, or C 1-6 alkyl substituted by one or more R 7b , or two R 7 together with the attached nitrogen atom form a 3-7 membered heterogeneous A cyclic group or a 3-7-membered heterocyclic group substituted by one or more R 7a ; when there are multiple substituents, they are the same or different;
  • Each R 7a and R 7b is independently deuterium, halogen, hydroxyl or cyano;
  • Each R 9 is independently hydrogen or C 1-6 alkyl
  • Ring D is a 3-7 membered heterocyclic group
  • R 10 is C 1-6 alkyl
  • R 11 is C 1-6 alkyl
  • the 3-7-membered heterocyclic group in the 3-7-membered heterocyclic group and the 3-7-membered heterocyclic group substituted by a substituent is each independently "the heteroatom species is independently selected from N, O and S, and the heteroatom The number of 3-7-membered heterocyclic groups is 1, 2 or 3.”
  • Ring A is C 6-12 aryl
  • Each R 1 is independently halogen, cyano, C 1-6 alkyl or C 1-6 alkyl substituted by one or more R 1a ; when the substituents are multiple, they are the same or different;
  • Each R 1a is independently halogen
  • n 2;
  • R 2 is hydrogen
  • R 3 is a 3-7-membered heterocyclic group substituted by one or more R 3e ; when there are multiple substituents, they are the same or different;
  • Each R 3-b is independently deuterium, hydroxyl, or halogen
  • X is a connecting bond, -O-, -(C 1 -C 4 alkylene)-, -O-(C 1 -C 4 alkylene)-, -(C 2 -C 4 alkenylene)- or -(C 2 -C 6 alkynylene)-;
  • the "C 1 -C 4 alkylene" in "-O-(C 1 -C 4 alkylene)-" is connected to R 4 ;
  • R 4 is hydrogen, C 1-6 alkyl, cyano group, hydroxyl, -N(R 7 ) 2 , 3-7 membered heterocyclyl, 3-7 membered heterocyclyl substituted by one or more R 4a or C 3-7 cycloalkyl substituted by one or more R 4b ; when there are multiple substituents, they are the same or different;
  • Each R 4a and R 4b is independently amino, C 1-6 alkyl, or C 1-6 alkyl substituted with one or more deuterium;
  • R 5 is hydrogen or C 1-6 alkyl
  • Each R 7 is independently hydrogen, C 1-6 alkyl, or C 1-6 alkyl substituted by one or more R 7b , or two R 7 together with the attached nitrogen atom form a 3-7 membered heterogeneous Ring group; when there are multiple substituents, they may be the same or different;
  • Each R 7b is independently deuterium or hydroxyl
  • Each R 9 is independently C 1-6 alkyl
  • Ring D is a 3-7 membered heterocyclic group
  • R 10 is C 1-6 alkyl
  • R 11 is C 1-6 alkyl
  • the 3-7-membered heterocyclic group in the 3-7-membered heterocyclic group and the 3-7-membered heterocyclic group substituted by a substituent is each independently "the heteroatom species is independently selected from N, O and S, and the heteroatom The number of 3-7-membered heterocyclic groups is 1, 2 or 3.”
  • Ring A is C 6-12 aryl
  • Each R 1 is independently halogen, cyano, C 1-6 alkyl or C 1-6 alkyl substituted by one or more R 1a ; when the substituents are multiple, they are the same or different;
  • Each R 1a is independently halogen
  • n 2;
  • R 2 is hydrogen
  • R 3 is a 3-7-membered heterocyclic group substituted by one or more R 3e ; when there are multiple substituents, they are the same or different;
  • Each R 3-b is independently deuterium or halogen
  • X is a connecting bond, -(C 1 -C 4 alkylene)-, -O-(C 1 -C 4 alkylene)-, -(C 2 -C 4 alkenylene)- or -(C 2 -C 6 alkynylene)-;
  • C 1 -C 4 alkylene in “-O-(C 1 -C 4 alkylene)-" is connected to R 4 ;
  • R 4 is hydrogen, C 1-6 alkyl, cyano group, hydroxyl, -N(R 7 ) 2 , 3-7 membered heterocyclyl substituted by one or more R 4a or substituted by one or more R 4b C 3-7 cycloalkyl; when there are multiple substituents, they may be the same or different;
  • Each R 4a and R 4b is independently amino, C 1-6 alkyl, or C 1-6 alkyl substituted with one or more deuterium;
  • R 5 is hydrogen or C 1-6 alkyl
  • Each R 7 is independently hydrogen or C 1-6 alkyl
  • Each R 9 is independently C 1-6 alkyl
  • Ring D is a 3-7 membered heterocyclic group
  • R 10 is C 1-6 alkyl
  • R 11 is C 1-6 alkyl.
  • Ring A is C 6-12 aryl, for example
  • Each R 1 is independently halogen, cyano, C 1-6 alkyl or C 1-6 alkyl substituted by one or more R 1a ; when the substituents are multiple, they are the same or different;
  • Each R 1a is independently halogen
  • n 2;
  • R 2 is hydrogen
  • R 3 is a 3-7-membered heterocyclic group substituted by one or more R 3e ; when there are multiple substituents, they are the same or different;
  • Each R 3-b is independently deuterium, hydroxyl, or halogen
  • X is -(C 1 -C 4 alkylene)-, -O-, -O-(C 1 -C 4 alkylene)-, -(C 2 -C 4 alkenylene)- or -(C 2 -C 6 alkynylene)-;
  • "-O-(C 1 -C 4 alkylene)-" in "C 1 -C 4 alkylene” is connected to R 4 ;
  • R 4 is -N(R 7 ) 2 , a 3-7-membered heterocyclyl group or a 3-7-membered heterocyclyl group substituted by one or more R 4a ; when there are multiple substituents, they may be the same or different;
  • Each R 4a is independently C 1-6 alkyl
  • R 5 is hydrogen or C 1-6 alkyl
  • Each R 7 is independently hydrogen, C 1-6 alkyl, or C 1-6 alkyl substituted by one or more R 7b , or two R 7 together with the attached nitrogen atom form a 3-7 membered heterogeneous Ring group; when there are multiple substituents, they may be the same or different;
  • Each R 7b is independently deuterium or hydroxyl
  • Each R 9 is independently C 1-6 alkyl
  • Ring D is a 3-7 membered heterocyclic group
  • R 10 is methyl
  • R 11 is methyl
  • the 3-7-membered heterocyclic group in the 3-7-membered heterocyclic group and the 3-7-membered heterocyclic group substituted by a substituent is each independently "the heteroatom species is independently selected from N, O and S, and the heteroatom The number of 3-7-membered heterocyclic groups is 1, 2 or 3.”
  • Ring A is C 6-12 aryl
  • Each R 1 is independently halogen, cyano, C 1-6 alkyl or C 1-6 alkyl substituted by one or more R 1a ; when the substituents are multiple, they are the same or different;
  • Each R 1a is independently halogen
  • n 2;
  • R 2 is hydrogen
  • R 3 is a 3-7-membered heterocyclic group substituted by one or more R 3e ; when there are multiple substituents, they are the same or different;
  • Each R 3-b is independently deuterium
  • X is -(C 1 -C 4 alkylene)-, -O-(C 1 -C 4 alkylene)-, -(C 2 -C 4 alkylene)- or -(C 2 -C 6 Alkynylene)-;
  • "-O-(C 1 -C 4 alkylene)-" in "C 1 -C 4 alkylene” is connected to R 4 ;
  • R 4 is -N(R 7 ) 2 or a 3-7-membered heterocyclic group substituted by one or more R 4a ; when there are multiple substituents, they are the same or different;
  • Each R 4a is independently C 1-6 alkyl
  • R 5 is hydrogen or C 1-6 alkyl
  • Each R 7 is independently hydrogen or C 1-6 alkyl
  • Each R 9 is independently C 1-6 alkyl
  • Ring D is a 3-7 membered heterocyclic group
  • R 10 is methyl
  • R 11 is methyl
  • Ring A is
  • each R 1 is independently halogen, cyano, C 1-6 alkyl or C 1-6 alkyl substituted by one or more R 1a .
  • R 1 is F, Cl, -CHF 2 , -CF 3 , cyano, -CH 3 or -CF 2 CH 3 , preferably F, -CHF 2 , -CH 3 or cyano.
  • R2 is hydrogen
  • R 5 is hydrogen or C 1-6 alkyl, for example: R 5 is hydrogen or methyl.
  • _ alkenylene)-or-(C 2 - C 6 alkynylene)-, "-O-(C 1 -C 4 alkylene)-" in "C 1 -C 4 alkylene” is connected to R 4 ; for example: connecting bond, Another example: connection key, Among them, the f end is connected to R 4 .
  • ring D when ring D is a 3-7-membered heterocyclyl group, ring D can be 1,2-dihydropyridyl, for example "c" means that the atom is located in the meta position of the N atom of the B ring.
  • R 3 when R 3 is a C 3-7 cycloalkyl group or a C 3-7 cycloalkyl group substituted by one or more R 3d , the R 3 can be
  • R 3 when R 3 is a 3-7-membered heterocyclyl group or a 3-7-membered heterocyclyl group substituted by one or more R 3e , the R 3 can be It can also be For example Another example Another example preferred
  • R 4 can be hydrogen, methyl, methoxy, cyano, halogen, hydroxyl, -NH 2 , -NHCH 3 , -N(CH 3 ) 2 , -N(CH 2 CH 3 ) 2 , -N(CD 3 ) 2 , Also hydrogen, methyl, methoxy, cyano, halogen, hydroxyl, -NH 2 , -NHCH 3 , -N(CH 3 ) 2 ,
  • R 10 is methyl
  • R 11 is methyl
  • the compound represented by formula I is any of the following compounds:
  • the present invention also provides a fused ring compound as shown in formula I as mentioned above, its stereoisomer or its pharmaceutically acceptable salt preparation method, wherein, when for "c" means that when the atom is located in the meta position of the N atom of the B ring, the preparation method includes the following steps: intermediate I-A1 and raw material N-phenylbis(trifluoromethanesulfonyl)imide or trifluoromethanesulfonic anhydride The reaction obtains intermediate I-A2, which then undergoes a coupling reaction with raw material I-A3 to obtain intermediate I-A4. I-A4 then undergoes a substitution reaction or palladium-catalyzed Buchwald coupling reaction with raw material I-A5. Obtain the compound of general formula IA;
  • substitution reaction between I-A4 and I-A5 occurs, it is carried out under the action of a base, including but not limited to cesium carbonate, sodium carbonate, potassium carbonate, triethylamine, diisopropylethylamine, etc.;
  • the catalyst includes but is not limited to RuPhos Pd G3, BrettPhos Pd G3, XPhos Pd G3, XantPhos Pd G3, RuPhos Pd G4 , BrettPhos Pd G4, etc.
  • the base includes but is not limited to cesium carbonate, sodium carbonate, potassium phosphate, sodium carbonate, potassium tert-butoxide, sodium tert-butoxide, etc.;
  • RG is hydrogen, -B(OH) 2 , -ZnBr or -ZnI.
  • the present invention also provides a compound represented by formula I-A1:
  • L, R 2 , R 3 , R 5 and R 10 are as described in any of the above solutions.
  • the compound represented by formula I-A1 is any of the following compounds:
  • the invention also provides a compound represented by formula I-A2:
  • L, R 2 , R 3 , R 5 and R 10 are as described in any of the above solutions.
  • the compound represented by formula I-A2 is any of the following compounds:
  • the invention also provides any of the following compounds:
  • the invention provides a pharmaceutical composition, which comprises:
  • substance Q which is a fused ring compound represented by formula I, its stereoisomer or a pharmaceutically acceptable salt thereof, and
  • the present invention provides the use of substance Q or the pharmaceutical composition as described above in the preparation of SOS1 inhibitors.
  • the substance Q is a fused ring compound shown in formula I, its stereoisomer or its pharmaceutically acceptable Take that with a grain of salt.
  • the SOS1 inhibitor can be used in mammalian organisms; it can also be used in vitro, mainly for experimental purposes, for example: as a standard sample or control sample to provide comparison, or according to the field
  • a kit is prepared using conventional methods to provide rapid detection of the effect of compounds on inhibiting SOS1.
  • the present invention provides the use of substance Q or the pharmaceutical composition as mentioned above in the preparation of medicines;
  • the substance Q is the fused ring compound shown in formula I as mentioned above, its stereoisomer or Its pharmaceutically acceptable salt;
  • the drug can be a drug for treating and/or preventing diseases related to SOS1 activity or expression level.
  • the disease related to SOS1 activity or expression is selected from non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, colon cancer, thyroid cancer, melanoma tumor, embryonal rhabdomyosarcoma, cutaneous granular cell tumor, liver cancer, rectal cancer, bladder cancer, throat cancer, breast cancer, prostate cancer, glioblastoma, ovarian cancer, head and neck squamous cell carcinoma, cervical cancer, esophageal cancer, Kidney cancer, skin cancer, lymphoma, gastric cancer, cholangiocarcinoma, uterine cancer, endometrial cancer, urothelial cancer, acute myeloid leukemia, myelofibrosis, B-cell lymphoma, monocytic leukemia, splenomegaly Hypereosinophilia, eosinophilic syndrome, and multiple myeloid carcinoma, as well as diseases
  • the present invention provides the use of substance Q or the pharmaceutical composition as mentioned above in the preparation of medicines;
  • the substance Q is the fused ring compound shown in formula I as mentioned above, its stereoisomer or Its pharmaceutically acceptable salt;
  • the drug is a drug for treating and/or preventing the following types of diseases;
  • the disease is selected from non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, colon cancer Cancer, thyroid cancer, melanoma, embryonal rhabdomyosarcoma, cutaneous granular cell tumor, liver cancer, rectal cancer, bladder cancer, throat cancer, breast cancer, prostate cancer, glioblastoma, ovarian cancer, head and neck squamous cell carcinoma, Cervical cancer, esophageal cancer, kidney cancer, skin cancer, lymphoma, gastric cancer, cholangiocarcinoma, uterine cancer, endometrial cancer, urothelial cancer, acute my
  • the invention provides a method for inhibiting SOS1, which includes administering to a patient (e.g., a human) a therapeutically effective amount of substance Q or a pharmaceutical composition as described above;
  • the substance Q is the fused ring compound shown in formula I as mentioned above, its stereoisomer or its pharmaceutically acceptable salt.
  • the present invention provides a method for treating and/preventing diseases, which includes administering to a patient (such as a human) a therapeutically effective amount of substance Q or a pharmaceutical composition as described above;
  • the substance Q is the fused ring compound shown in formula I as mentioned above, its stereoisomer or its pharmaceutically acceptable salt;
  • the disease is a disease related to SOS1 activity or expression level.
  • the disease related to SOS1 activity or expression is selected from the group consisting of non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, and colon cancer.
  • thyroid cancer thyroid cancer, melanoma, embryonal rhabdomyosarcoma, cutaneous granular cell tumor, liver cancer, rectal cancer, bladder cancer, throat cancer, breast cancer, prostate cancer, glioblastoma, ovarian cancer, head and neck squamous cell carcinoma, cervical cancer Cancer, esophageal cancer, kidney cancer, skin cancer, lymphoma, gastric cancer, cholangiocarcinoma, uterine cancer, endometrial cancer, urothelial cancer, acute myeloid leukemia, myelofibrosis, B-cell lymphoma, monocytic leukemia , splenomegaly, eosinophilic syndrome and multiple myeloma, as well as diseases related to SOS1 genetic mutations, including but not limited to neurofibromatosis type 1, Noonan syndrome, multiple lentigines Noonan syndrome, capillary malformation-arteriovenous malformation syndrome, cardio-facial-cutaneous syndrome, Costello syndrome
  • halogen refers to fluorine, chlorine, bromine or iodine.
  • alkyl refers to a saturated straight or branched chain hydrocarbon group consisting only of carbon atoms and hydrogen atoms and having a specified number of carbon atoms (e.g., C 1-10 , preferably C 1-6 , more preferably C 1-4 ) .
  • Alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, isobutyl, sec-butyl, n-pentyl or n-hexyl, etc.
  • alkylene means a saturated divalent hydrocarbyl group obtained by removing two hydrogen atoms from a saturated straight or branched chain hydrocarbon group; that is, one of the hydrogens in the alkyl group is substituted, as defined above.
  • alkylene groups include methylene (-CH 2 -), ethylene ⁇ including -CH 2 CH 2 - or -CH(CH 3 )- ⁇ , n-propylene ⁇ including -CH 2 CH 2 CH 2 - ⁇ , isopropylene ⁇ including -CH(CH 3 )CH 2 -, -CH(CH 2 CH 3 )- or -C(CH 3 ) 2 - ⁇ , etc.
  • alkenylene refers to an unsaturated divalent group obtained by removing two hydrogen atoms from a linear or branched alkene molecule; that is, one of the hydrogens in the alkenyl group is substituted, and the alkenyl group is as defined above.
  • alkynyl refers to a straight or branched hydrocarbon chain radical having at least one triple bond, which consists only of carbon atoms and hydrogen atoms, and has a specified number of carbon atoms (e.g., C 2-10 , preferably C 2-6 , more preferably C 2-4 ) and is connected to the rest of the molecule through a single bond; for example, alkynyl groups include but are not limited to ethynyl (-C ⁇ CH), propynyl (-C ⁇ C-CH 3 ), - C ⁇ C-CH 2 -CH 3 , -CH 2 C ⁇ C-CH 3 , -C ⁇ C-CH 2 -CH 2 -CH 3 , -CH 2 C ⁇ C-CH 2 -CH 3 , -C ⁇ C-CH 2 -CH 2 -CH 3 , -C ⁇ C-CH 2 -CH 2 -CH(CH 3 ) 2 , -C ⁇ CC(CH 3 ) 3 ,
  • alkynylene refers to an unsaturated divalent group obtained by removing two hydrogen atoms from a straight-chain or branched alkyne molecule; that is, one of the hydrogens in the alkynyl group is substituted.
  • the alkynyl group is as defined above. narrate.
  • alkynylene groups include, but are not limited to, ethynylene, propynylene, pentynylene, and the like, such as -C ⁇ C-, -CH 2 -C ⁇ CH, -C ⁇ C-CH 2 -, -C ⁇ C-CH 2 -CH 2 -, -C ⁇ C-CH(CH 3 )-CH 2 -, -C ⁇ CC(CH 3 ) 2 -.
  • C 1-6 alkyl-O- C 1-6 alkyl is as defined above.
  • cycloalkyl means a saturated monocyclic or polycyclic (eg, bicyclic, tricyclic or multicyclic bridged, fused (fused) or spirocyclic ring system) carbocyclic substituent, and which may be Any suitable carbon atom is connected to the rest of the molecule by a single bond; for example, having 3 to 15 ring carbon atoms, preferably 3 to 10 ring carbon atoms, more preferably 3 to 7 ring carbon atoms; for example, C 3- 7 Cycloalkyl includes but is not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or wait.
  • cycloalkenyl means a monocyclic or polycyclic (such as bicyclic, tricyclic or more cyclic bridged, fused (fused) or spirocyclic ring system having at least one double bond (such as a carbon-carbon double bond) ) of a non-aromatic cyclic hydrocarbon group, and it can be connected to the rest of the molecule by a single bond via any suitable carbon atom; for example, it has 3 to 15 ring carbon atoms, preferably 3 to 10 ring carbon atoms, more preferably Having 3 to 7 ring carbon atoms; for example, C 3-7 cycloalkenyl includes but is not limited to cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl or cycloheptenyl.
  • Monocyclic or polycyclic such as bicyclic, tricyclic or more bridged rings, fused rings (fused rings) or spiro ring systems
  • heterocyclic hydrocarbon groups preferably containing 1 to 3 heterocyclic groups selected from N, O and S
  • the heterocyclyl group may
  • aryl refers to an aromatic group consisting of a conjugated hydrocarbon ring system composed of carbon atoms that satisfies the 4n+2 rule, and each ring is aromatic. In one embodiment, “aryl” refers to an aromatic group having 6 to 18 (preferably 6 to 12) carbon atoms. Examples of aryl groups include, but are not limited to, phenyl or naphthyl, and the like.
  • heteroaryl means a 5-20-membered (preferably 5-12-membered, more preferably 5-10-membered) ring having 2 to 15 carbon atoms and 1 to 5 heteroatoms selected from nitrogen, oxygen and sulfur. More preferably, it is a 5- to 8-membered, most preferably a 5- to 6-membered) conjugated ring system group.
  • the heteroaryl group may be a monocyclic, bicyclic, tricyclic or multicyclic ring system, and may also be fused with a cycloalkyl, cycloalkenyl or heterocyclyl group as defined above.
  • heteroaryl refers to an aromatic group containing heteroatoms, and each ring is aromatic; preferably the heteroatom species is independently selected from N, O and S, and the number of heteroatoms is 1 , 2 or 3 5-10 membered heteroaryl groups or 5-6 membered heteroaryl groups.
  • heteroaryl groups include, but are not limited to, thienyl, imidazolyl, pyrazolyl, thiazolyl, oxazolyl, diazolyl, oxadiazolyl, isoxazolyl, pyridyl, pyrimidinyl, pyrazinyl, Pyridazinyl, benzimidazolyl (for example ), benzopyrazolyl, indolyl, furyl, pyrrolyl, triazolyl, tetrazolyl, triazinyl, indolizinyl, isoxazolyl, thiadiazolyl, isoindolyl, indazole Base, isoindazolyl, purinyl, quinolyl, isoquinolinyl, diazonaphthyl, naphthyridinyl, quinoxalinyl, pteridinyl, carbazolyl, carbolinyl, phenanth
  • cycloalkyl-O- cycloalkyl is as defined above.
  • cycloalkenyl-O- cycloalkenyl is as defined above.
  • heterocyclyl-O- heterocyclyl is as defined above.
  • the "-" at the end of a group means that the group is connected to other fragments in the molecule through this site.
  • one or more means 1, 2, 3, 4, 5, 6, 7, 8, 9 or more.
  • the present invention adopts traditional methods of mass spectrometry and elemental analysis, and each step and condition can refer to the conventional operating steps and conditions in the art.
  • this invention employs standard nomenclature and standard laboratory procedures and techniques of analytical chemistry, synthetic organic chemistry, and optics. In some cases, standard techniques are used for chemical synthesis, chemical analysis.
  • heteroatom species are independently selected from N, O and S, and the number of heteroatoms is 1, 2 or 3
  • the heteroatom species are independently selected from N, O and S
  • the heteroatom species is independently selected from one or more of N, O and S.
  • the number of heteroatoms is 1 or 2
  • the heteroatom species are independently selected from N and O
  • the heteroatom species are independently selected from N and O
  • the heteroatom species are independently selected from N and O
  • the fused ring compound represented by Formula I of the present invention may contain one or more chiral centers and exist in different optically active forms.
  • a compound contains enantiomers when it contains a chiral center.
  • the present invention includes both isomers and mixtures of isomers, such as racemic mixtures. Enantiomers can be resolved by methods known in the art, such as crystallization and chiral chromatography. When the fused ring compound represented by formula I contains more than one chiral center, diastereomers may exist.
  • the present invention includes resolved optically pure specific isomers as well as mixtures of diastereoisomers. Diastereomers can be resolved by methods known in the art, such as crystallization and preparative chromatography.
  • stereoisomer includes conformational isomers and configurational isomers, wherein configurational isomers mainly include cis-trans isomers and optical isomers.
  • the compounds described in the present invention may exist in the form of stereoisomers, and therefore encompass all possible stereoisomer forms, including but not limited to cis-trans isomers, enantiomers, diastereomers, Atropisomers, etc., the compounds of the present invention can also be in the form of any combination or any mixture of the aforementioned stereoisomers, such as meso, racemate, equal mixtures of atropisomers, etc. exist.
  • a single enantiomer, a single diastereomer or a mixture thereof, or a single atropisomer or a mixture thereof When the compound described in the present invention contains an olefin double bond, unless otherwise specified, it includes cis isomers and trans isomers, and any combination thereof.
  • Atropisomers of the present invention are stereoisomers with axial or planar chirality resulting from restricted intramolecular rotation.
  • the present invention provides compounds represented by the above-mentioned types of structures, or their cis-trans isomers, meso, racemates, enantiomers, diastereomers, and atropisomers. isomers, tautomers or mixtures thereof, where "mixtures thereof” include any of the aforementioned stereoisomers (such as cis-trans isomers, enantiomers, diastereomers, Any form of mixture between atropisomers), tautomers and/or mixtures (meso, racemate), such as mixtures of cis and trans isomers, enantiomers and A mixture of diastereoisomers, a mixture of diastereomers, a mixture of atropisomers, or a mixture of cis-trans isomers and racemates, enantiomers and diastereomers
  • the fused ring compounds shown in Formula I, their stereoisomers or their pharmaceutically acceptable salts are intended to encompass the fused ring compounds shown in Formula I any isotopically labeled (or "radiolabeled") variant of a substance, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  • a variant may be a fused ring compound as shown in Formula I, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof in which one or more atoms have an atomic mass or mass number different from that normally found in nature. Obtained by atomic substitution of atomic mass or mass number.
  • the radionuclide used will depend on the specific application of the radiolabeled variant. For example, for in vitro receptor labeling and competition assays, 3 H or 14 C are often useful. For radiography applications, 11 C or 18 F are often useful.
  • isotopic variants of the compounds of the invention in particular those in which one or more radioactive isotopes have been incorporated, can be advantageous, for example, to investigate the mechanism of action or the distribution of the active ingredient in the body; due to the relative ease of preparation and Detectability, compounds labeled with 3H or 14C isotopes are particularly suitable for this purpose.
  • the incorporation of isotopes such as deuterium can produce specific therapeutic benefits due to the greater metabolic stability of the compound, such as extending the half-life in the body or reducing the required effective dose; therefore, such modifications of the compounds of the invention can also be used in some This situation constitutes a preferred embodiment of the present invention.
  • Isotopic variants of the compounds of the present invention can be prepared by methods known to those skilled in the art, for example by the methods described below and in the operating examples, by using corresponding isotopically modified specific reagents and/or starting compounds. .
  • a "pharmaceutical composition” refers to a formulation comprising a compound of the invention and a vehicle generally accepted in the art for delivering a biologically active compound to a mammal, such as a human.
  • the medium includes a pharmaceutically acceptable carrier.
  • the purpose of pharmaceutical compositions is to facilitate administration to organisms and facilitate the absorption of active ingredients to exert biological activity.
  • pharmaceutically acceptable refers to substances (such as pharmaceutical excipients) that do not affect the biological activity or properties of the compounds of the invention and are relatively non-toxic, that is, the substances can be administered to individuals without causing adverse effects. Biologically react or interact in an adverse manner with any component contained in the composition.
  • pharmaceutical excipients refer to the excipients and additives used in the production of drugs and formulation of prescriptions. They are, in addition to active ingredients, included in pharmaceutical preparations. all substances in . See the Pharmacopoeia of the People's Republic of China (2015 Edition), Part Four, or Handbook of Pharmaceutical Excipients (Raymond C Rowe, 2009 Sixth Edition). Excipients are mainly used to provide a safe, stable and functional pharmaceutical composition. They can also provide a method to enable the active ingredients to dissolve at a desired rate after administration, or promote the activity of the composition after administration. Ingredients are absorbed effectively.
  • the pharmaceutical excipients may be inert fillers, or provide certain functions, such as stabilizing the overall pH value of the composition or preventing degradation of the active ingredients of the composition.
  • the pharmaceutical excipients may include one or more of the following excipients: binders, suspending agents, emulsifiers, diluents, fillers, granulating agents, adhesives, disintegrants, lubricants, and anti-adhesion agents. Agents, glidants, wetting agents, gelling agents, absorption delaying agents, dissolution inhibitors, enhancers, adsorbents, buffers, chelating agents, preservatives, colorants, flavorings and sweeteners.
  • compositions of the present invention may be prepared according to the disclosure using any method known to those skilled in the art. For example, conventional mixing, dissolving, granulating, emulsifying, grinding, encapsulating, embedding or freeze-drying processes.
  • the fused ring compound represented by Formula I, its stereoisomer or its pharmaceutically acceptable salt can be administered in any form of a pharmaceutical composition.
  • compositions may be prepared according to methods well known in the pharmaceutical art and may be administered by a variety of routes, depending on the desired local or systemic treatment and the area to be treated. Administration may be topical (including epidermal and transdermal, ocular and mucosal, including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powder or aerosol, including by nebulizer; intratracheal or intranasal) , oral (solid and liquid formulations) or parenteral forms of administration.
  • Examples of solid oral dosage forms include, but are not limited to, powders, capsules, caplets, softgels, and tablets.
  • Examples of liquid formulations for oral or mucosal administration include, but are not limited to, suspensions, emulsions, elixirs, and solutions.
  • Examples of topical formulations include, but are not limited to, emulsions, gels, ointments, creams, patches, pastes, foams, lotions, drops, or serum formulations.
  • Examples of formulations for parenteral administration include, but are not limited to, injectable solutions, dry formulations which may be dissolved or suspended in a pharmaceutically acceptable carrier, injectable suspensions, and injectable emulsions.
  • compositions and preparations for topical administration may include transdermal patches, salves, lotions, ointments, gels, drops, suppositories, sprays, liquids and powders.
  • suitable formulations of the pharmaceutical composition include, but are not limited to, eye drops and other ophthalmic formulations; aerosols: such as nasal sprays or inhalants.
  • Oral administration may include dosage forms formulated for once daily or twice daily (BID) administration.
  • Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal, intramuscular, or injection or infusion; or intracranial, such as intrathecal or intraventricular administration. Parenteral administration may be in the form of a single bolus dose, or may be by continuous infusion pump. Conventional pharmaceutical carriers, water, powdered or oily bases, thickening agents, and the like may be necessary or desirable.
  • Pharmaceutical compositions including the present invention may also be in controlled or delayed release dosage forms (eg liposomes or microspheres).
  • treatment refers to therapeutic therapy or palliative measures.
  • treatment means: (1) alleviating the disease or one or more biological manifestations of the condition, (2) interfering with (a) one or more points in the biological cascade that causes or causes the condition or (b) ) one or more biological manifestations of a condition, (3) amelioration of one or more symptoms, effects, or side effects associated with the condition, or one or more symptoms, effects, or side effects associated with the condition or its treatment, or (4) slow the progression of a condition or one or more biological manifestations of a condition.
  • Treatment may also refer to prolonging survival compared to expected survival without treatment.
  • prevention refers to the reduction of the risk of acquiring or developing a disease or disorder.
  • terapéuticaally effective amount refers to an amount of a compound sufficient to effectively treat the disease or condition described herein when administered to a patient.
  • the “therapeutically effective amount” will vary depending on the compound, the condition and its severity, and the age of the patient to be treated, but can be adjusted as necessary by one skilled in the art.
  • patient refers to any animal, preferably a mammal, and most preferably a human, to which a compound or composition is or has been administered in accordance with embodiments of the present invention.
  • mammal includes any mammal. Examples of mammals include, but are not limited to, cattle, horses, sheep, pigs, cats, dogs, mice, rats, rabbits, guinea pigs, monkeys, humans, etc., with humans being the most preferred.
  • pharmaceutically acceptable salt refers to a salt obtained by reacting a compound with a pharmaceutically acceptable (relatively nontoxic, safe, and suitable for use by a patient) acid or base.
  • base addition salts can be obtained by contacting the free form of the compound with a sufficient amount of a pharmaceutically acceptable base in a suitable inert solvent.
  • Pharmaceutically acceptable base addition salts include, but are not limited to, sodium salts, potassium salts, calcium salts, aluminum salts, magnesium salts, bismuth salts, ammonium salts, etc.
  • acid addition salts can be obtained by contacting the free form of the compound with a sufficient amount of a pharmaceutically acceptable acid in a suitable inert solvent.
  • a pharmaceutically acceptable acid include inorganic acids and organic acids.
  • the reagents and raw materials used in the present invention are all commercially available.
  • the positive progressive effect of the present invention is that: the fused ring compound represented by Formula I of the present invention has good inhibitory activity against SOS1; It is expected to treat and/or prevent diseases related to SOS1 activity or expression.
  • the structure of the compound of the present invention is determined by nuclear magnetic resonance (NMR) or/and mass spectrometry (MS). NMR chemical shifts ( ⁇ ) are given in parts per million (ppm).
  • the NMR was measured using a Bruker AVANCE-400 NMR instrument.
  • the measurement solvent was deuterated dimethyl sulfoxide (DMSO-d 6 ) or deuterated chloroform (CDCl 3 ), and the internal standard was tetramethylsilane (TMS). ).
  • the mass spectrometry was measured using an Agilent 1260-6125B single quadrupole liquid mass spectrometer using an electrospray ion source (ESI).
  • the thin layer chromatography chromatography silica gel plate uses Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate.
  • the specifications used are 0.15mm ⁇ 0.20mm.
  • the specifications used for the thin layer chromatography chromatography are 0.4mm ⁇ 0.5mm.
  • Preparative high-performance liquid chromatography used an AutoPurification LC preparation system equipped with an ACQUITY QDa mass spectrometer detector produced by Waters.
  • the preparative column was SunFire C18 5 ⁇ m 19x250mm OBD preparative column.
  • the mobile phase used varying gradients of water (containing 0.1% formic acid)-acetonitrile to elute the compounds.
  • Boc tert-butoxycarbonyl
  • TIPS triisopropylsilyl
  • Bn benzyl
  • DMSO dimethyl sulfoxide
  • Tf trifluoromethanesulfonyl
  • Ms methanesulfonyl.
  • 6-Chloro-2-methylpyridin-3-ol (10 g, 69.6 mmol) was dissolved in acetonitrile (100 mL), and N-bromosuccinimide (13.6 g, 76.6 mmol) was added. The reaction was stirred at room temperature for 1.5 hours. The reaction was concentrated under reduced pressure, diluted with saturated aqueous sodium sulfite solution (200 mL) and saturated aqueous sodium chloride solution (200 mL), and extracted with ethyl acetate (200 mL x 2). The organic phase was washed with saturated sodium chloride solution (300 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • Step 2 tert-Butyl (S)-2-((4-bromo-6-chloro-2-methylpyridin-3-yl)oxy)methyl)pyrrolidine-1-carboxylate
  • Step 3 tert-Butyl (S)-2-((6-chloro-2-methyl-4-(methylamino)pyridin-3-yl)oxy)methyl)pyrrolidine-1-carboxylate
  • Step 4 tert-Butyl(S)-2-((6-chloro-5-iodo-2-methyl-4-(methylamino)pyridin-3-yl)oxy)methyl)pyrrolidine-1- Carboxylate
  • Step 5 tert-Butyl(S)-2-((6-chloro-5-(3-ethoxy-3-oxoprop-1-en-1-yl)-2-methyl-4-( Methylamino)pyridin-3-yl)oxy)methyl)pyrrolidine-1-carboxylate
  • Step 6 tert-Butyl(S)-2-((5-chloro-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridin-8-yl)oxy (Methyl)pyrrolidine-1-carboxylate
  • Step 7 tert-Butyl (S)-2-((3-bromo-5-chloro-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridine-8 -yl)oxy)methyl)pyrrolidine-1-carboxylate
  • Step 8 tert-Butyl(S)-2-((3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-chloro-1,7-dimethyl-2-oxo- 1,2-Dihydro-1,6-naphthyridin-8-yl)oxy)methyl)pyrrolidine-1-carboxylate
  • Step 9 tert-Butyl (S)-2-((3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-((R)-1-(3-(difluoromethyl) )-2-Fluorophenyl)ethyl)amino)-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridin-8-yl)oxy)methyl )pyrrolidine-1-carboxylate
  • Step 10 3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-((R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethyl) Amino)-1,7-dimethyl-8-((S)-pyrrolidin-2-yl)methoxy)-1,6-naphthyridin-2(1H)-one
  • Step 11 3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-((R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethyl) Amino)-1,7-dimethyl-8-((S)-1-methylpyrrolidin-2-yl)methoxy)-1,6-naphthyridin-2(1H)-one (Compound 1 )
  • Step 12 (3-(1-acetyl-4-fluoropiperidin-4-yl)-5-((R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethyl )Amino)-1,7-dimethyl-8-((S)-1-methylpyrrolidin-2-yl)methoxy)-1,6-naphthyridin-2(1H)-one (Compound 59)
  • Step 13 (3-(1-acetyl-4-methoxypiperidin-4-yl)-5-((R)-1-(3-(difluoromethyl)-2-fluorophenyl) Ethyl)amino)-1,7-dimethyl-8-((S)-1-methylpyrrolidin-2-yl)methoxy)-1,6-naphthyridin-2(1H)-one (Compound 2)
  • Compound 2 was prepared by referring to the method of steps 2 to 13 in Example 1, wherein the raw material (S)-(-)-1-tert-butoxycarbonyl-2-pyrrolidinemethanol in step 2 was replaced with (R)-( -)-1-tert-butoxycarbonyl-2-pyrrolidinemethanol.
  • Step 1 tert-Butyl (2-((4-bromo-6-chloro-2-methylpyridin-3-yl)oxy)ethyl)(methyl)carbamate
  • Step 2 tert-Butyl (2-((6-chloro-2-methyl-4-(methylaminopyridin-3-yl)oxy)ethyl)(methyl)carbamate
  • Step 3 tert-Butyl (2-((6-chloro-5-iodo-yl-4-(methylaminopyridin-3-yl)oxy)ethyl)(methyl)carbamate
  • tert-Butyl (2-((6-chloro-2-methyl-4-(methylaminopyridin-3-yl)oxy)ethyl)(methyl)carbamate (28.3g, 84.6mmol) Dissolve in acetic acid (300mL), add N-iodosuccinimide (28.5g, 126.9mmol), stir at room temperature for 20 hours. Add saturated sodium sulfite aqueous solution (200mL) to dilute, add saturated sodium bicarbonate aqueous solution (200mL) to neutralize , add ethyl acetate (300mL) for extraction.
  • Step 4 3-(5-(2-((tert-butoxycarbonyl)(methyl)amino)ethoxy)-2-chloro-6-methyl-4-(methylamino)pyridin-3-yl )Ethyl acrylate
  • Step 5 tert-Butyl(2-((5-chloro-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridin-8-yl)oxy)ethyl (Methyl)carbamate
  • Step 6 tert-butyl (2-((3-bromo-5-chloro-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridin-8-yl) Oxy)ethyl(methyl)carbamate
  • Step 7 tert-Butyl (2-((3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-chloro-1,7-dimethyl-2-oxo-1,2 -Dihydro-1,6-naphthyridin-8-yl)oxy)ethyl(methyl)carbamate
  • Step 8 tert-Butyl(R)-(2-((3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)- 2-Fluorophenyl)ethyl)amino)-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridin-8-yl)oxy)ethyl(methyl base) carbamate
  • Step 9 (R)-3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl )amino)-1,7-dimethyl-8-(2-(methylamino)ethoxy)-1,6-naphthyridin-2(1H)-one
  • Step 10 (R)-3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl )amino)-8-(2-(dimethylamino)ethoxy)-1,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Step 11 (R)-3-(1-acetyl-4-fluoropiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl )amino)-8-(2-(dimethylamino)ethoxy)-1,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Step 12 (R)-3-(1-acetyl-4-methoxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-8-(2-(dimethylamino)ethoxy)-1,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • step 11 The residue obtained in step 11 was dissolved in methanol (15 mL) and a solution of sodium methoxide in methanol (455 mg, 2.44 mmol, 30% wtin MeOH) was added. The reaction solution was stirred at 40° C. for 24 hours, concentrated under reduced pressure, and the residue was purified by preparative HPLC to obtain compound 4 as a white solid (25 mg, yield 19%).
  • Step 5 8-(benzyloxy)-5-chloro-1,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Step 6 8-(benzyloxy)-3-bromo-5-chloro-1,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Step 7 3-(1-acetyl-4-hydroxypiperidin-4-yl)-8-(benzyloxy)-5-chloro-1,7-dimethyl-1,6-naphthyridin-2 (1H)-ketone
  • Step 8 (R)-3-(1-acetyl-4-hydroxypiperidin-4-yl)-8-(benzyloxy)-5-((1-(3-(difluoromethyl)- 2-Fluorophenyl)ethyl)amino)-1,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • reaction solution was cooled to room temperature, water and ethyl acetate were added to extract and separate the layers.
  • the organic phase was washed once with 10% citric acid solution (300 mL) and saturated brine (100 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • Step 9 (R)-3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl )Amino)-8-hydroxy-1,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Step 10 (R)-3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl )Amino)-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridin-8-yl triflate
  • Step 11 (R)-3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl )Amino)-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridine-8-carbonitrile
  • Step 1 (R)-3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl )Amino)-1,7-dimethyl-8-((triisopropylsilyl)ethynyl)-1,6-naphthyridin-2(1H)-one
  • Step 2 (R)-3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl )Amino)-8-ethynyl-1,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Step 1 (R)-3-(1-acetyl-4-fluoropiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl )Amino)-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridine-8-carbonitrile
  • Step 2 (R)-3-(1-acetyl-4-methoxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridine-8-carbonitrile
  • Step 1 tert-Butyl(R)-(4-(3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2 -Fluorophenyl)ethyl)amino)-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridin-8-yl)-2-methylbutan-3 -Alkyn-2-yl)carbamate
  • Step 2 (R)-3-(1-acetyl-4-hydroxypiperidin-4-yl)-8-(3-amino-3-methylbut-1-yn-1-yl)-5- ((1-(3-(Difluoromethyl)-2-fluorophenyl)ethyl)amino)-1,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Step 1 tert-Butyl(S)-2-((3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-(((R)-1-(3-(difluoromethyl) (yl)-2-fluorophenyl)ethyl)amino)-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridin-8-yl)ethynyl)pyrrole Alkane-1-carboxylate
  • Step 2 3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-(((R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethyl )Amino)-1,7-dimethyl-8-((S)-pyrrolidin-2-ylethynyl)-1,6-naphthyridin-2(1H)-one
  • Step 3 3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-(((R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethyl )Amino)-1,7-dimethyl-8-(((S)-1-methylpyrrolidin-2-yl)ethynyl)-1,6-naphthyridin-2(1H)-one
  • Step 1 3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-chloro-8-hydroxy-1,7-dimethyl-1,6-naphthyridin-2(1H)- ketone
  • Step 2 3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-chloro-1,7-dimethyl-2-oxo-1,2-dihydro-1,6- Naphthyridin-8-yl triflate
  • Step 3 tert-butyl(4-(3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-chloro-1,7-dimethyl-2-oxo-1,2- Dihydro-1,6-naphthyridin-8-yl)-2-methylbut-3-yn-2-yl)carbamate
  • Step 4 tert-butyl (4-(3-(1-acetyl-4-hydroxypiperidin-4-yl)-5-chloro-1,7-dimethyl-2-oxo-1,2- Dihydro-1,6-naphthyridin-8-yl)-2-methylbut-3-yn-2-yl)carbamate
  • Step 5 (R)-3-(1-((3-(1-acetyl-4-hydroxypiperidin-4-yl)-8-(3-amino-3-methylbutan-1-yne- 1-yl)-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridin-5-yl)amino)ethyl)-2-methylbenzonitrile
  • Compound 17 was prepared by referring to steps 3 to 5 of Example 15, wherein the raw material (2-methylbut-3-yn-2-yl)carbamic acid tert-butyl ester in step 3 was replaced with (1-ethynylcyclopropyl tert-butyl carbamate, the raw material (R)-3-(1-aminoethyl)-2-methylbenzonitrile in step 4 is replaced by (R)-1-(3-(difluoromethyl) )-2-fluorophenyl)ethane-1-amine.
  • Step 1 3-(1-acetyl-4-methoxypiperidin-4-yl)-8-(benzyloxy)-5-chloro-1,7-dimethyl-1,6-naphthyridine -2(1H)-ketone
  • Step 2 3-(1-acetyl-4-methoxypiperidin-4-yl)-5-chloro-8-hydroxy-1,7-dimethyl-1,6-naphthyridin-2(1H )-ketone
  • Step 3 3-(1-acetyl-4-methoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-2-oxo-1,2-dihydro-1, 6-naphthyridin-8-yl trifluoromethanesulfonate
  • Step 4 3-(1-acetyl-4-methoxypiperidin-4-yl)-5-chloro-8-(3-(dimethylamino)prop-1-yn-1-yl)-1 ,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Step 5 (R)-3-(1-acetyl-4-methoxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-8-(3-(dimethylamino)prop-1-yn-1-yl)-1,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Step 1 3-(1-acetyl-4-(methoxy-d 3 )piperidin-4-yl)-8-(benzyloxy)-5-chloro-1,7-dimethyl-1 ,6-Naphthyridin-2(1H)-one
  • Step 2 3-(1-acetyl-4-(methoxy-d 3 )piperidin-4-yl)-5-chloro-8-hydroxy-1,7-dimethyl-1,6-naphthalene Dino-2(1H)-one
  • Step 3 3-(1-acetyl-4-(methoxy-d 3 )piperidin-4-yl)-5-chloro-1,7-dimethyl-2-oxo-1,2- Dihydro-1,6-naphthyridin-8-yl triflate
  • Step 4 tert-butyl(4-(3-(1-acetyl-4-(methoxy-d 3 )piperidin-4-yl)-5-chloro-1,7-dimethyl-2- Oxo-1,2-dihydro-1,6-naphthyridin-8-yl)-2-methylbut-3-yn-2-yl)carbamate
  • Step 5 tert-butyl(R)-(4-(3-(1-acetyl-4-(methoxy-d 3 )piperidin-4-yl)-5-((1-(3-( Difluoromethyl)-2-fluorophenyl)ethyl)amino)-1,7-dimethyl-2-oxo-1,2-dihydro-1,6-naphthyridin-8-yl)- 2-Methylbut-3-yn-2-yl)carbamate
  • Step 6 (R)-3-(1-acetyl-4-(methoxy-d 3 )piperidin-4-yl)-8-(3-amino-3-methylbut-1-yne- 1-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl)amino)-1,7-dimethyl-1,6-naphthyridine-2( 1H)-ketone
  • Compound 34 was prepared with reference to step 5 of Example 17, wherein the raw material (R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethane-1-amine was replaced with (R)-3-( 1-Aminoethyl)-2-methylbenzonitrile.
  • Step 1 3-(1-acetyl-4-ethoxypiperidin-4-yl)-8-(benzyloxy)-5-chloro-1,7-dimethyl-1,6-naphthyridine -2(1H)-ketone
  • Step 2 3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-8-hydroxy-1,7-dimethyl-1,6-naphthyridin-2(1H )-ketone
  • Step 3 3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-2-oxo-1,2-dihydro-1, 6-naphthyridin-8-yl trifluoromethanesulfonate
  • Step 4 3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-8-(3-(dimethylamino)prop-1-yn-1-yl)-1 ,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Step 5 (R)-3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl) Ethyl)amino)-8-(3-(dimethylamino)prop-1-yn-1-yl)-1,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Compound 65 was prepared with reference to Example 36, in which the raw material (R)-3-(1-aminoethyl)-2-fluorobenzonitrile was replaced by (R)-3-(1-aminoethyl)-2-methyl benzonitrile.
  • Step 1 (R)-3-(1-acetyl-4-fluoropiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl )amino)-8-(3-(dimethylamino)prop-1-yn-1-yl)-1,7-dimethyl-1,6-naphthyridin-2(1H)-one (compound 106)
  • Step 2 (R)-3-(1-acetyl-4-((2-hydroxyethyl)amino)piperidin-4-yl)-5-((1-(3-(difluoromethyl)) -2-Fluorophenyl)ethyl)amino)-8-(3-(dimethylamino)prop-1-yn-1-yl)-1,7-dimethyl-1,6-naphthyridine-2 (1H)-ketone
  • Compound 70 was prepared according to the method of Example 40, in which the raw material 2,2,2-trifluoroethanol was replaced with ethylene glycol. ESI-MS m/z: 628.3[M+1] + .
  • Compound 72 was prepared according to the method of Example 40, in which the raw material 2,2,2-trifluoroethanol was replaced with isopropyl alcohol.
  • Step 1 3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-8-(3-(diethylamino)prop-1-yn-1-yl)-1 ,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Step 2 (R)-3-(1-acetyl-4-ethoxypiperidin-4-yl)-8-(3-(diethylamino)prop-1-yn-1-yl)-5 -((1-(3-(Difluoromethyl)-2-fluorophenyl)ethyl)amino)-1,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Step 1 3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-8-(3-(pyrrolidin-1-yl)propanol) -1-yn-1-yl)-1,6-naphthyridin-2(1H)-one
  • Step 2 (R)-3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-1,7-dimethyl -8-(3-(pyrrolidin-1-yl)prop-1-yn-1-yl)-1,6-naphthyridin-2(1H)-one
  • Step 1 3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-8-(3-hydroxyprop-1-yn-1-yl)-1,7-di Methyl-1,6-naphthyridin-2(1H)-one
  • Step 2 3-(3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-2-oxo-1,2-dihydro -1,6-Naphthyridin-8-yl)prop-2-yn-1-yl methanesulfonate
  • Step 3 3-(1-acetyl-4-ethoxypiperidin-4-yl)-8-(3-(bis(methyl-d 3 )amino)prop-1-yn-1-yl) -5-Chloro-1,7-dimethyl-1,6-naphthyridin-2(1H)-one
  • Step 4 (R)-3-(1-acetyl-4-ethoxypiperidin-4-yl)-8-(3-(bis(methyl-d 3 )amino)propan-1-yne- 1-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl)amino)-1,7-dimethyl-1,6-naphthyridine-2( 1H)-ketone
  • Compound 76 was prepared with reference to step 4 of Example 47, wherein the starting material (R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethane-1-amine was replaced with (R)-3- (1-Aminoethyl)-2-fluorobenzonitrile.
  • Compound 77 was prepared with reference to step 4 of Example 47, wherein the starting material (R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethane-1-amine was replaced with (R)-3- (1-Aminoethyl)-2-methylbenzonitrile.
  • Step 1 tert-Butyl 4-((3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-2-oxo-1, 2-Dihydro-1,6-naphthyridin-8-yl)oxy)piperidine-1-carboxylate
  • Step 2 3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-8-(piperidin-4-yloxy)-1 ,6-Naphthyridin-2(1H)-one
  • Step 3 3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-8-((1-methylpiperidin-4-yl) )oxy)-1,6-naphthyridin-2(1H)-one
  • Step 4 (R)-3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-1,7-dimethyl-8-((1-methylpiperidin-4-yl)oxy)-1,6-naphthyridin-2(1H)-one
  • Step 1 tert-Butyl 3-((3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-2-oxo-1, 2-Dihydro-1,6-naphthyridin-8-yl)oxy)azetidine-1-carboxylate
  • Step 2 3-(1-acetyl-4-ethoxypiperidin-4-yl)-8-(azetidin-3-yloxy)-5-chloro-1,7-dimethyl Base-1,6-naphthyridin-2(1H)-one
  • Step 3 3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-8-((1-methylazetidine- 3-yl)oxy)-1,6-naphthyridin-2(1H)-one
  • step 2 Dissolve the crude product obtained in step 2 in tetrahydrofuran (10 mL), add 30% formaldehyde aqueous solution (0.5 mL) and sodium triacetoxyborohydride (1.0 g, 4.74 mmol), and stir at room temperature for 1 hour.
  • the reactant was diluted with water and extracted with ethyl acetate.
  • the organic phase was washed with saturated sodium bicarbonate solution and saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • Step 4 (R)-3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-1,7-dimethyl-8-((1-methylazetidin-3-yl)oxy)-1,6-naphthyridin-2(1H)-one
  • Compound 80 was prepared with reference to step 4 of Example 51, wherein the starting material (R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethane-1-amine was replaced with (R)-3- (1-Aminoethyl)-2-fluorobenzonitrile.
  • Step 1 tert-Butyl(S)-2-(((3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-2- Oxo-1,2-dihydro-1,6-naphthyridin-8-yl)oxy)methyl)pyrrolidine-1-carboxylate
  • Step 2 (S)-3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-8-(pyrrolidin-2-ylmethane) Oxy)-1,6-naphthyridin-2(1H)-one
  • Step 3 (S)-3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-8-((1-methylpyrrolidine -2-yl)methoxy)-1,6-naphthyridin-2(1H)-one
  • Step 4 3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-(((R)-1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-1,7-dimethyl-8-(((S)-1-methylpyrrolidin-2-yl)methoxy)-1,6-naphthyridin-2(1H)- ketone
  • Compound 82 was prepared with reference to step 4 of Example 53, wherein the starting material (R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethane-1-amine was replaced with (R)-3- (1-Aminoethyl)-2-fluorobenzonitrile.
  • Step 1 tert-Butyl(R)-3-((3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-2-oxy Generation-1,2-dihydro-1,6-naphthyridin-8-yl)oxy)pyrrolidine-1-carboxylate
  • Step 2 (R)-3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-8-(pyrrolidin-3-yloxy base)-1,6-naphthyridin-2(1H)-one
  • Step 3 (R)-3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-8-((1-methylpyrrolidine -3-yl)oxy)-1,6-naphthyridin-2(1H)-one
  • Step 4 3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-(((R)-1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-1,7-dimethyl-8-(((R)-1-methylpyrrolidin-3-yl)oxy)-1,6-naphthyridin-2(1H)-one
  • Compound 84 was prepared according to the method of Example 55, in which the raw material (S)-3-hydroxypyrrolidine-1-carboxylic acid tert-butyl ester in step 1 was replaced with (R)-3-hydroxypyrrolidine-1-carboxylic acid. Tert-butyl ester.
  • Step 1 3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-chloro-1,7-dimethyl-8-(2-(pyrrolidin-1-yl)ethyl) Oxy)-1,6-naphthyridin-2(1H)-one
  • Step 2 (R)-3-(1-acetyl-4-ethoxypiperidin-4-yl)-5-((1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-1,7-dimethyl-8-(2-(pyrrolidin-1-yl)ethoxy)-1,6-naphthyridin-2(1H)-one
  • Compound 86 was prepared according to step 2 of Example 57, wherein the starting material (R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethane-1-amine was replaced It is (R)-3-(1-aminoethyl)-2-fluorobenzonitrile.
  • Compound 87 was prepared according to the method of Example 53, in which the raw material (S)-1-tert-butoxycarbonyl-2-pyrrolidinemethanol in step 1 was replaced with (R)-1-tert-butoxycarbonyl-2-pyrrolidine. Methanol. ESI-MS m/z: 644.3[M+1] + .
  • Compound 88 was prepared according to the method of Example 53, in which the raw material (S)-1-tert-butoxycarbonyl-2-pyrrolidinemethanol in step 1 was replaced with (R)-1-tert-butoxycarbonyl-2-pyrrolidine. Methanol, the raw material (R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethane-1-amine in step 4 is replaced with (R)-3-(1-aminoethyl) -2-Fluorobenzonitrile.
  • Example A Inhibitory effects of compounds on cell growth of NCI-H358 and MIA PaCa-2 tumor cell lines under 3D culture conditions use.
  • NCI-H358 was purchased from Shanghai Dijin Biotechnology Co., Ltd.
  • MIA PaCa-2 was purchased from Shanghai Dio Biotechnology Co., Ltd.
  • Cells in the logarithmic growth phase were inoculated into ultra-low adsorption 96-well plates (4000, 2000 cells/well, 180 ⁇ l/well for NCI-H358 and MIA PaCa-2 cells respectively) and cultured at 37°C and 5% CO2. , the cells were aggregated to form microspheres, and a gradient dilution of the test compound was added after 1 day.
  • the details are as follows: Take the compound stock solution (10mM) previously dissolved in DMSO, dilute it 4 times to 10 gradient concentrations, and use culture medium to dilute it to 10 times the target concentration in another 96-well plate, and then Add 20 ⁇ l/well of the compound solution to the 96-well plate in which cells are seeded to reach the target concentration (10000, 2500, 625, 156, 39, 10, 2.5, 0.6, 0.15, 0.04nM). Set up 3 duplicate wells for each concentration and set up a blank control.
  • CellTiter- 3D reagent luciferase ATP bioluminescence detection reagent used to detect 3D cell microspheres, purchased from Promega, Cat. No. G9683
  • shake for 10 minutes incubate at room temperature for 20 minutes, and then detect the fluorescence intensity (light collection time is 100ms).
  • cell activity inhibition rate (%) [(luminescence intensity 6 days containing cell culture medium control group - luminescence intensity 6 days compound group )/(luminescence intensity 6 days containing cells) Medium control group – luminescence intensity 6 days cell-free medium control group )] ⁇ 100%.
  • Data were analyzed using GraphPad Prism 8.3 software, and nonlinear S-curve regression was used to fit the data to obtain a dose-response curve, from which the IC 50 value was calculated. The results are shown in Table 1.
  • BI-3406 is an SOS1 inhibitor reported in the literature (Cancer Discovery 2021 (11) 142-157), and its structure is:
  • Example B In vivo pharmacokinetic test in rats
  • SD rats were used as test animals to study the pharmacokinetic behavior of the compounds of the present invention administered orally and intravenously in rats.
  • Oral administration group 3 female SD rats were fasted for 12 hours and then administered the test compound by gavage (dose was 5 mg/kg, administration volume was 10 mL/kg, vehicle was 10% DMSO/10% Solutol/80% normal saline) ).
  • the sampling time points were 0.25h, 0.5h, 1h, 2h, 3h, 4h, 6h, 8h and 24h after administration.
  • Solutol refers to polyethylene glycol (15)-hydroxystearate.
  • Intravenous administration group 3 female SD rats were given test compounds intravenously after fasting for 12 hours (dose: 1 mg/kg, administration volume: 5 mL/kg, vehicle: 10% DMSO/10% Solutol/80% normal saline) ).
  • the sampling time points were 5min, 0.25h, 0.5h, 1h, 2h, 4h, 8h and 24h after administration.
  • 0.2 mL of venous blood was taken from the retroocular venous plexus of the rat, placed in a K 2 -EDTA (dipotassium ethylenediaminetetraacetate) test tube, centrifuged at 6800g for 6 minutes at 4°C, and the plasma was separated at -80 Store at °C.
  • K 2 -EDTA dipotassium ethylenediaminetetraacetate
  • AUC plasma exposure
  • Cmax maximum plasma concentration
  • V apparent volume of distribution
  • CL clearance
  • F oral bioavailability.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (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

本发明公开了一种稠环化合物、其制备方法及其应用。本发明提供了一种如式(I)所示的稠环化合物、其立体异构体或其药学上可接受的盐。本发明的如式I所示的稠环化合物对SOS1具有较好地抑制活性;有望治疗和/或预防与SOS1活性或表达量相关的疾病;

Description

一种稠环化合物、其制备方法及其应用
本申请要求申请日为2022/8/5的中国专利申请2022109369139、申请日为2022/10/21的中国专利申请202211294374X、申请日为2022/11/30的中国专利申请2022115292859和申请日为2023/7/24的中国专利申请2023109135728的优先权。本申请引用上述中国专利申请的全文。
技术领域
本发明涉及一种稠环化合物、其制备方法及其应用。
背景技术
RAS蛋白是一种位于细胞膜上的分子量为21kDa的鸟嘌呤三核苷酸磷酸(GTP)结合蛋白,由188或189个氨基酸构成。RAS蛋白的活性则是通过与GTP或鸟嘌呤二核苷酸磷酸(GDP)的结合进行调节,当与GDP结合时,处于“失活”状态,当与GTP结合时,处于“激活”状态。RAS蛋白本身有比较微弱的GTP酶水解功能和较慢的核苷酸交换速率,结合GTP酶激活蛋白(GTPase activating proteins,GAP),可以增强RAS蛋白的GTP水解功能,而鸟嘌呤核苷酸交换因子(GEF)可以催化核苷酸的交换(GTP交换GDP)。SOS1(Son of Sevenless 1)是一种RAS特异的GEF,SOS1与RAS结合可以促进RAS释放GDP而与GTP结合,从而激活RAS。RAS被激活后,可以激活多条下游信号通路,包括MAPK信号通路、PI3K信号通路,这些信号通路在促进细胞分化、增殖、生存方面具有重要作用。RAS突变是许多癌症的基因驱动因素,20%-30%的人类肿瘤中都存在RAS突变,例如肺癌、结直肠癌和胰腺癌。
RAS基因家族包括KRAS、NRAS和HRAS。KRAS突变存在于多种肿瘤中,如肺腺癌(32%)、结直肠癌(41%)、胰腺癌(86%),KRAS突变又以第12位密码子的G12突变最为常见,例如,在KRAS突变的肺腺癌、结直肠癌及胰腺癌中,G12突变又分别占85%、68%及91%;HRAS突变和NRAS突变频率相对较低,主要发生于黑色素瘤、白血病和甲状腺癌等癌症种类中。另外,RAS蛋白的异常激活(如基因突变、扩增和过表达等)和一些抗肿瘤药物的耐药性也紧密相关,如EGFR单抗和EGFR小分子抑制剂等。因此,RAS相关信号通路成为重要的抗肿瘤靶标。
参与癌症相关的信号通路的RAS家族的鸟嘌呤核苷酸交换因子主要为SOS1,降低SOS1的表达可以显著抑制KRAS突变的癌细胞的增殖和存活。由于SOS1是多条激活RAS信号通路的共有节点,几乎所有的生长因子受体是通过SOS1来启动RAS信号通路,因此SOS1抑制剂有潜力成为广谱抗癌药物。SOS1参与激活的信号通路在其他突变类型的癌症中也起着重要的作用。SOS1可以和接头蛋白Grb2相互作用,形成SOS1/Grb2复合体,结合到活化的受体酪氨酸激酶上(如EGFR、HER2、Erbb4、TRKA、TRKB、TRKC、RET和AXL等)。SOS1也可以被招募到磷酸化的细胞表面受体上,如T细胞受体、B细胞受体和单核细胞集落刺激因子受体等。SOS1在细胞膜上的定位使得SOS1可以更好地促进RAS家族蛋白活化,激活下游信号通路。SOS1还参与了其他GTP水解酶的活化,如RAC1等,RAC1也和多种人类癌症和其他疾病的发病机制有关。除此之外,SOS1突变被发现存在于肺腺癌、胚胎性横纹肌肉瘤和皮肤颗粒细胞肿瘤中,SOS1的过表达被发现存在于膀胱癌和前列腺癌中。除癌症以外,遗传性的SOS1突变也和RAS病变的发病机制相关,包括努南氏综合征、心-面-皮肤综 合征以及一型遗传性牙龈纤维瘤等。
目前尚无SOS1抑制剂批准上市,因此需要开发新的疗效好的SOS1抑制剂来满足临床需求。
发明内容
本发明要解决的技术问题是现有的SOS1抑制剂结构单一等缺陷,本发明提供了一种稠环化合物、其制备方法及其应用。该类化合物对SOS1具有较好地抑制活性。
本发明提供了一种如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐;
其中:
环A为C6-12芳基、5-10元杂芳基、C3-7环烷基、C3-7环烯基或3-7元杂环基;
每个R1独立地为氘、羟基、卤素、-N(R7)2、-SR9、氰基、氧代(=O)、硝基、C1-6烷基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1a取代的C1-6烷基、被一个或多个R1b取代的C1-6烷基-O-、被一个或多个R1c取代的C2-6烯基、被一个或多个R1d取代的C2-6炔基、C3-7环烷基、被一个或多个R1e取代的C3-7环烷基、3-7元杂环基、被一个或多个R1f取代的3-7元杂环基、C6-12芳基、被一个或多个R1g取代的C6-12芳基、5-10元杂芳基、被一个或多个R1h取代的5-10元杂芳基、C3-7环烯基、被一个或多个R1i取代的C3-7环烯基、-C(=O)-O-R9、-O-C(=O)-R9、-C(=O)-N(R7)2、-S(O)2-R9、-NR8-S(O)2-R9、-NR8-S(O)2-N(R7)2、-S(O)2-N(R7)2、-S(O)-N(R7)2、-S(O)-R9、-NR8-S(O)-R9、-NR8-S(O)-N(R7)2、-C(=O)-R9、-NR8C(=O)-R9或-NR8C(=O)-O-R9,或两个R1与所相连的环原子一起形成C3-7环烷基、被一个或多个R1e取代的C3-7环烷基、3-7元杂环基、被一个或多个R1f取代的3-7元杂环基、C3-7环烯基或被一个或多个R1i取代的C3-7环烯基;当取代基为多个时,相同或不同;
每个R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h和R1i各自独立地为氘、羟基、卤素、-N(R7)2、-SR9、硝基、氰基、C1-6烷基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1-a取代的C1-6烷基、被一个或多个R1-b取代的C1-6烷基-O-、被一个或多个R1-c取代的C2-6烯基、被一个或多个R1-d取代的C2- 6炔基、C3-7环烷基、C3-7环烷基-O-、3-7元杂环基、3-7元杂环基-O-、被一个或多个R1-e取代的C3-7环烷基、被一个或多个R1-e取代的C3-7环烷基-O-、被一个或多个R1-f取代的3-7元杂环基、被一个或多个R1-f取代的3-7元杂环基-O-、C6-12芳基、被一个或多个R1-g取代的C6-12芳基、5-10元杂芳基、被一个或多个R1-h取代的5-10元杂芳基、C3-7环烯基、C3-7环烯基-O-、被一个或多个R1-i取代的C3-7环烯基、被一个或多个R1-i取代的C3-7环烯基-O-、-C(=O)-O-R9、-O-C(=O)-R9、-C(=O)-N(R7)2、-S(O)2-R9、-NR8-S(O)2-R9、-NR8-S(O)2-N(R7)2、-S(O)2-N(R7)2、-S(O)-N(R7)2、-S(O)-R9、-NR8-S(O)-R9、-NR8- S(O)-N(R7)2、-C(=O)-R9、-NR8C(=O)-R9或-NR8C(=O)-O-R9;当取代基为多个时,相同或不同;
每个R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h和R1-i各自独立地为氘、羟基、卤素、-N(R7)2、-SR9、硝基、氰基、C1-6烷基、C1-6烷基-O-、C2-6烯基、C2-6炔基、C3-7环烷基、C3-7环烯基或3-7元杂环基;
环D为C6-12芳基、5-6元杂芳基、C3-7环烷基、C3-7环烯基或3-7元杂环基;
R2为不存在、氢、氘、羟基、卤素、氨基、氰基、氧代、硝基、C1-6烷基、C1-6烷基-O-、C2-6烯基、C2-6炔基、C3-7环烷基、C3-7环烯基或3-7元杂环基;
L为连接键、-C(=O)-、-C(=O)O-、-C(=O)NR8-、-NR8-、-S-、-O-、-S(O)-、-S(O)2-或-(CH2)p-;当L为连接键时,表示R3直接与D环连接;
R3为氢、氘、C1-6烷基、被一个或多个R3a取代的C1-6烷基、C2-6烯基、被一个或多个R3b取代的C2-6烯基、C2-6炔基、被一个或多个R3c取代的C2-6炔基、C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、3-7元杂环基、被一个或多个R3e取代的3-7元杂环基、C6-12芳基、被一个或多个R3f取代的C6-12芳基、5-10元杂芳基、被一个或多个R3g取代的5-10元杂芳基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;当取代基为多个时,相同或不同;
每个R3a、R3b、R3c、R3d、R3e、R3f、R3g和R3h各自独立地为氘、羟基、卤素、-N(R7)2、氰基、硝基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R3-c取代的C2-6烯基、被一个或多个R3-d取代的C2-6炔基、C3-7环烷基、被一个或多个R3-e取代的C3-7环烷基、3-7元杂环基、被一个或多个R3-f取代的3-7元杂环基、C6-12芳基、被一个或多个R3-g取代的C6-12芳基、5-10元杂芳基、被一个或多个R3-h取代的5-10元杂芳基、C3-7环烯基、被一个或多个R3-i取代的C3-7环烯基、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、-SR9、-C(=O)-O-R9、-O-C(=O)-R9、-C(=O)-N(R7)2、-S(O)2-R9、-NR8-S(O)2-R9、-NR8-S(O)2-N(R7)2、-S(O)2-N(R7)2、-S(O)-N(R7)2、-S(O)-R9、-NR8-S(O)-R9、-NR8-S(O)-N(R7)2、-C(=O)-R9、-NR8C(=O)-R9或-NR8C(=O)-O-R9,或两个R3d、两个R3e、两个R3f、两个R3g或两个R3h与所相连的环原子一起形成C3-7环烷基、被一个或多个R3-e取代的C3-7环烷基、3-7元杂环基、被一个或多个R3-f取代的3-7元杂环基、C3-7环烯基或被一个或多个R3-i取代的C3-7环烯基;当取代基为多个时,相同或不同;
每个R3-a、R3-b、R3-c、R3-d、R3-e、R3-f、R3-g、R3-h、R3-i和R3-j各自独立地为氘、羟基、卤素、-N(R7)2、硝基、氰基、C1-6烷基、C1-6烷基-O-、C3-7环烷基、C3-7环烯基、3-7元杂环基、-SR9、-C(=O)-O-R9、-O-C(=O)-R9、-C(=O)-N(R7)2、-S(O)2-R9、-NR8-S(O)2-R9、-NR8-S(O)2-N(R7)2、-S(O)2-N(R7)2、-C(=O)-R9、-NR8C(=O)-R9或-NR8C(=O)-O-R9
X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;
R4为氢、C1-6烷基、C1-6烷基-O-、氰基、卤素、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
每个R4a和R4b各自独立地为氢、氘、羟基、卤素、氨基、氰基、硝基、氨基保护基团、C1-6烷基或被一个或多个氘取代的C1-6烷基;
R5为不存在、氧代、氢、氘、羟基、卤素、氨基、氰基、硝基、C1-6烷基、被一个或多个R5a取代的C1-6烷基、C1-6烷基-O-、C2-6烯基、C2-6炔基、C3-7环烷基、被一个或多个R5b取代的C3-7环烷基、3-7元杂环基、被一个或多个R5c取代的3-7元杂环基、C3-7环烯基或被一个或多个R5d取代的C3-7环烯基;当取代基为多个时,相同或不同;
每个R5a、R5b和R5c各自独立地为氘、C1-6烷基、羟基、卤素、氨基或氰基;
R6为氢、氘、羟基、卤素、氨基、氰基、氧代(=O)、C1-6烷基、C1-6烷基-O-、C2-6烯基、C2-6炔基或不存在;
每个R7独立地为氢、C1-6烷基、被一个或多个R7b取代的C1-6烷基、C3-7环烷基、C3-7环烯基或氨基保护基团,或两个R7与所相连的氮原子一起形成3-7元杂环基或被一个或多个R7a取代的3-7元杂环基;当取代基为多个时,相同或不同;
每个R7a和R7b各自独立地为氘、C1-6烷基、卤素、羟基、氨基、氰基、C1-6烷基-O-、C2-6烯基或C2-6炔基;
每个R8和R9各自独立地为氢、C1-6烷基、C2-6烯基、C2-6炔基、C3-7环烷基、C3-7环烯基或3-7元杂环基;
R10为氢、氘、羟基、卤素、氨基、氰基、C1-6烷基-O-、C2-6烯基、C2-6炔基或C1-6烷基;
R11为氰基、C1-6烷基或被一个或多个R11a取代的C1-6烷基;当取代基为多个时,相同或不同;
每个R11a独立地为氘、卤素或羟基;
m为0、1、2、3、4或5;
p为1、2、3、4、5或6;
所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”;
所述5-10元杂芳基和被取代基取代的5-10元杂芳基中的5-10元杂芳基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的5-10元杂芳基”;
所述5-6元杂芳基为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的5-6元杂芳基”;
所述的氨基保护基团为叔丁氧羰基、苄氧羰基、芴甲氧羰基、烯丙氧羰基、邻苯二甲酰基、苄基、对甲氧基苄基、三苯甲基或对甲苯磺酰基;
当*的碳原子具有手性时,如式I所示的稠环化合物为 或其混合物。
在某一方案中,所述如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,某些基团具有如下定义,未提及的基团的定义如本发明中任一方案所述(本段内容以下简称为“在某一方案中”)。
在某一方案中,当X为连接键时,R4为C1-6烷基、C1-6烷基-O-、氰基、卤素、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同。
在某一方案中,X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;当X为连接键时,R4为氰基、
在某一方案中,R6为氧代;L为连接键、-C(=O)-、-C(=O)O-、-C(=O)NR8-、-NR8-、-O-、-S-、-S(O)-、-S(O)2-或-(CH2)p-;当L为连接键时,表示R3直接与D环连接;R3为被一个或多个R3e取代的3-7元杂环基;当取代基为多个时,相同或不同;每个R3e各自独立地为氘、羟基、卤素、-N(R7)2、氰基、硝基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1- 6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R3-c取代的C2-6烯基、被一个或多个R3-d取代的C2-6炔基、C3-7环烷基、被一个或多个R3-e取代的C3-7环烷基、3-7元杂环基、被一个或多个R3-f取代的3-7元杂环基、C6-12芳基、被一个或多个R3-g取代的C6-12芳基、5-10元杂芳基、被一个或多个R3-h取代的5-10元杂芳基、C3-7环烯基、被一个或多个R3-i取代的C3-7环烯基、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、-SR9、-C(=O)-O-R9、-O-C(=O)-R9、-C(=O)-N(R7)2、-S(O)2-R9、-NR8-S(O)2-R9、-NR8-S(O)2-N(R7)2、-S(O)2-N(R7)2、-S(O)-N(R7)2、-S(O)-R9、-NR8-S(O)-R9、-NR8-S(O)-N(R7)2、-C(=O)-R9、-NR8C(=O)-R9或-NR8C(=O)-O-R9;当取代基为多个时,相同或不同。
在某一方案中,R6为氧代;L为连接键;当L为连接键时,表示R3直接与D环连接;R3为被一个或多个R3e取代的3-7元杂环基;当取代基为多个时,相同或不同;每个R3e各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、C2-6炔基或-C(=O)-R9;当取代基为多个时,相同或不同。
在某一方案中,不为氢。
在某一方案中,R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i、R2、R3、R3a、R3b、R3c、R3d、R3e、R3f、R3g、R3h、R3-a、R3-b、R3-c、R3-d、R3- e、R3-f、R3-g、R3-h、R3-i、R3-j、R4、R4a、R4b、R5、R5a、R5b、R5c、R6、R7、R7a、R7b、R8、R9、R10和R11中,所述C1-6烷基、被取代基取代的C1-6烷基中的C1-6烷基各自独立地为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基,例如为甲基或乙基。
在某一方案中,R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i、R2、R3a、R3b、R3c、R3d、R3e、R3f、R3g、R3h、R3-a、R3-b、R3-c、R3-d、R3-e、R3- f、R3-g、R3-h、R3-i、R3-j、R4、R4a、R4b、R5、R5a、R5b、R5c、R6、R7a、R7b、R10和R11a中,所述卤素各自独立地为F、Cl、Br或I,例如为F。
在某一方案中,R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i、R2、R3a、R3b、R3c、R3d、R3e、R3f、R3g、R3h、R3-a、R3-b、R3-c、R3-d、R3-e、R3- f、R3-g、R3-h、R3-i、R3-j、R4、R5、R6、R7a、R7b和R10中,所述C1-6烷基-O-、被取代基取代的C1-6烷基-O-中的C1-6烷基-O-各自独立地为甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基、仲丁氧基或叔丁氧基,例如为甲氧基。
在某一方案中,R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i、R2、R3、R3a、R3b、R3c、R3d、R3e、R3f、R3g、R3h、R5、R6、R7a、R7b、R8、R9和R10中,所述C2-6烯基、被取代基取代的C2-6烯基中的C2-6烯基各自独立地为乙烯基(-CH=CH2)、丙烯基(-CH=CH-CH3)、烯丙基(-CH2-CH=CH2)、异丙烯基(-C(CH3)=CH2)、-CH=CH-CH2-CH3、-CH2-CH=CH-CH3、-CH=CH-CH2-CH2-CH3、-CH2-CH=CH-CH2-CH3、-CH=CH-CH(CH3)2、-CH2-CH=C(CH3)2或-CH=CH-(CH2)3-CH3
在某一方案中,R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i、R2、R3、R3a、R3b、R3c、R3d、R3e、R3f、R3g、R3h、R5、R6、R7a、R7b、R8、R9和R10中,所述C2-6炔基、被取代基取代的C2-6炔基中的C2-6炔基各自独立地为乙炔基(-C≡CH)、丙炔基(-C≡C-CH3)、-C≡C-CH2-CH3、-CH2C≡C-CH3、-C≡C-CH2-CH2-CH3、-CH2C≡C-CH2-CH3、-C≡C-CH2-CH2-CH2-CH3、-C≡C-CH2-CH(CH3)2、-C≡C-C(CH3)3、-C≡C-CH(CH3)2或-CH2C≡C-CH2-CH2-CH3
在某一方案中,环A中的C3-7环烷基,R1中的C3-7环烷基、被取代基取代的C3-7环烷基中的C3- 7环烷基,两个R1与所相连的环原子一起形成C3-7环烷基、两个R1与所相连的环原子一起形成被取代基取代的C3-7环烷基中的C3-7环烷基,R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h和R1i中的C3-7环烷基、被取代基取代的C3-7环烷基中的C3-7环烷基、C3-7环烷基-O-中的C3-7环烷基、被取代基取代的C3- 7环烷基-O-中的C3-7环烷基,环D、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i和R2中的C3-7环烷基,R3中的C3-7环烷基、被取代基取代的C3-7环烷基中的C3-7环烷基,R3a、R3b、R3c、R3d、R3e、R3f、R3g和R3h中的C3-7环烷基、被取代基取代的C3-7环烷基中的C3-7环烷基、C3-7环烷基-O-中的C3- 7环烷基、被取代基取代的C3-7环烷基-O-中的C3-7环烷基,两个R3d、两个R3e、两个R3f、两个R3g或两个R3h与所相连的环原子一起形成C3-7环烷基,两个R3d、两个R3e、两个R3f、两个R3g或两个R3h与所相连的环原子一起形成被取代基取代的C3-7环烷基中的C3-7环烷基,R3-a、R3-b、R3-c、R3-d、R3-e、R3-f、R3-g、R3-h、R3-i和R3-j中的C3-7环烷基,R4中的C3-7环烷基、被取代基取代的C3-7环烷基中的C3- 7环烷基,R5中的C3-7环烷基、被取代基取代的C3-7环烷基中的C3-7环烷基,R7中的C3-7环烷基,R8和R9中的C3-7环烷基,各自独立地为环丙基、环丁基、环戊基、环己基、环庚基或例如为环丙基。
在某一方案中,环A中的C3-7环烯基,R1中的C3-7环烯基、被取代基取代的C3-7环烯基中的C3- 7环烯基、两个R1与所相连的环原子一起形成C3-7环烯基、两个R1与所相连的环原子一起形成被取代基取代的C3-7环烯基中的C3-7环烯基,R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h和R1i中的C3-7环烯基、被取代基取代的C3-7环烯基中的C3-7环烯基、C3-7环烯基-O-中的C3-7环烯基、被取代基取代的C3- 7环烯基-O-中的C3-7环烯基,环D、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i和R2中的C3-7环烯基,R3中的C3-7环烯基、被取代基取代的C3-7环烯基中的C3-7环烯基,R3a、R3b、R3c、R3d、R3e、R3f、R3g和R3h中的C3-7环烯基、被取代基取代的C3-7环烯基中的C3-7环烯基,两个R3d、两个R3e、两个R3f、两个R3g或两个R3h与所相连的环原子一起形成C3-7环烯基,两个R3d、两个R3e、两个R3f、两个R3g或两个R3h与所相连的环原子一起形成被取代基取代的C3-7环烯基中的C3-7环烯基,R3-a、R3- b、R3-c、R3-d、R3-e、R3-f、R3-g、R3-h、R3-i和R3-j中的C3-7环烯基,R5中的C3-7环烯基、被取代基取代的C3-7环烯基中的C3-7环烯基,R7、R8和R9中的C3-7环烯基,所述C3-7环烯基各自独立地为环丙烯基、环丁烯基、环戊烯基、环己烯基或环庚烯基。
在某一方案中,环A、环D、R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R1-a、R1-b、R1- c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i、R2、R3、R3a、R3b、R3c、R3d、R3e、R3f、R3g、R3h、R3-a、R3-b、R3-c、R3-d、R3-e、R3-f、R3-g、R3-h、R3-i、R3-j、R4、R5、R7、R8和R9中,所述3-7元杂环基、被取代基取代的3-7元杂环基中的3-7元杂环基、3-7元杂环基-O-中的3-7元杂环基、被取代基取代的3-7元杂环基-O-中的3-7元杂环基各自独立地为饱和或部分不饱和的非芳香性的单环或多环(例如双环、三环或更多环的桥环、并环(稠环)或螺环体系)杂环基,具体地,单环杂环基可为吡咯烷基、四氢吡喃基、哌啶基、吗啉基、硫代吗啉基、高哌嗪基、哌嗪基、氮杂环丁基、1,2-二氢吡啶基、四氢吡啶基等。
在某一方案中,R3中,所述3-7元杂环基、被一个或多个R3e取代的3-7元杂环基中的3-7元杂环基的杂原子个数独立为1个或2个;所述3-7元杂环基、被一个或多个R3e取代的3-7元杂环基中的3-7元杂环基的杂原子种类独立选自N和O中的一种或两种;所述3-7元杂环基、被一个或多个R3e取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N和O中的一种或两种,杂原子个数为1个或2个的3-7元杂环基”;进一步地,所述3-7元杂环基、被一个或多个R3e取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子个数为1个或2个,杂原子种类独立选自N和O”的4-7元杂环基;所述的4-7元杂环基可为4-7元单环杂环基、5-7元桥环杂环基或5-7元螺环杂环基;具体地,当R3为4-7元单环杂环基时,所述4-7元单环杂环基可为哌啶基、四氢吡喃基、哌嗪基、氮杂环丁基、吗啉基或吡咯烷基;当R3为5-7元桥环杂环基时,所述5-7元桥环杂环基可为当R3为5-7元螺环杂环基时,所述5-7元螺环杂环基 可为例如为
在某一方案中,R4中,所述3-7元杂环基、被一个或多个R4a取代的3-7元杂环基中的3-7元杂环基的杂原子个数独立为1个、2个或3个;所述3-7元杂环基、被一个或多个R4a取代的3-7元杂环基中的3-7元杂环基的杂原子种类独立选自N、O和S中的一种或多种;所述3-7元杂环基、被一个或多个R4a取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”;进一步地,所述3-7元杂环基、被一个或多个R4a取代的3-7元杂环基中的3-7元杂环基的杂原子个数独立为1个或2个;所述3-7元杂环基、被一个或多个R4a取代的3-7元杂环基中的3-7元杂环基的杂原子种类独立选自N和O中的一种或两种;所述3-7元杂环基、被一个或多个R4a取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N和O中的一种或两种,杂原子个数为1个或2个的4-7元杂环基”;例如氮杂环丁基、吡咯烷基、哌啶基、哌嗪基、四氢吡啶基或吗啉基;又例如 再例如
在某一方案中,R7中,所述两个R7与所相连的氮原子一起形成3-7元杂环基、被一个或多个R7a取代的3-7元杂环基中的3-7元杂环基的杂原子个数独立为1个或2个;所述两个R7与所相连的氮原子一起形成3-7元杂环基、被一个或多个R7a取代的3-7元杂环基中的3-7元杂环基的杂原子种类独立选自N和O中的一种或两种;所述两个R7与所相连的氮原子一起形成3-7元杂环基、被一个或多个R7a取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N和O种的一种或两种,杂原子个数为1个或2个的4-7元杂环基”;例如:吡咯烷基,又例如
在某一方案中,环D中,所述3-7元杂环基为“杂原子种类为N,杂原子个数为1个或2个的6元杂环基”,例如为1,2-二氢吡啶基,又例如为“c”表示该原子位于B环N原子的间位。
在某一方案中,环A、环D、R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R3、R3a、R3b、 R3c、R3d、R3e、R3f、R3g和R3h中,所述C6-12芳基、被取代基取代的C6-12芳基中的C6-12芳基各自独立地为苯基或萘基;例如:苯基。
在某一方案中,环A、R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R3、R3a、R3b、R3c、R3d、R3e、R3f、R3g和R3h中,所述5-10元杂芳基、被取代基取代的5-10元杂芳基中的5-10元杂芳基各自独立地为“杂原子个数为1个、2个或3个,杂原子种类独立选自N、O和S的5-8元杂芳基”。
在某一方案中,X中,所述-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-中的-(C1-C4亚烷基)-各自独立地为亚甲基、亚乙基、亚正丙基或亚异丙基,例如:-CH2-、-CH2CH2-、-CH(CH3)-、-CH2CH2CH2-、-CH(CH3)CH2-、-CH(CH2CH3)-或-C(CH3)2-,优选-CH2-、-CH2CH2-、-CH(CH3)-或-CH2CH2CH2-;进一步地,“-O-(C1-C4亚烷基)-”中“C1-C4亚烷基”与R4相连。
在某一方案中,X中,所述-(C2-C4亚烯基)-独立地为亚乙烯基、亚丙烯基、亚烯丙基或亚丁烯基,例如-CH=CH-、-CH=CH-CH2-;进一步地,所述-(C2-C4亚烯基)-可为 其中f端与R4相连。
在某一方案中,X中,所述-(C2-C6亚炔基)-独立地为亚乙炔基、亚丙炔基、亚炔丙基、亚丁炔基或亚戊炔基,例如-C≡C-、-CH2-C≡CH、-C≡C-CH2-、-C≡C-CH2-CH2-、-C≡C-CH(CH3)-CH2-、-C≡C-C(CH3)2-,优选为-C≡C-、-C≡C-CH2-或-C≡C-C(CH3)2-;进一步地,所述-(C2-C6亚炔基)-可为-C≡C-*、-C≡C-CH2-*或-C≡C-C(CH3)2-*,其中*端与R4相连。
在某一方案中,环A为C6-12芳基;优选地,环A为苯基。
在某一方案中,每个R1独立地为卤素、羟基、-N(R7)2、C1-6烷基、氰基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1a取代的C1-6烷基或被一个或多个R1b取代的C1-6烷基-O-;优选地,每个R1独立地为卤素、C1-6烷基、氰基或被一个或多个R1a取代的C1-6烷基;当取代基为多个时,相同或不同。
在某一方案中,每个R1a和R1b各自独立地为卤素;优选地,每个R1a和R1b各自独立地为F或Cl。
在某一方案中,m为0、1、2或3;优选地,m为2。
在某一方案中,R2为氢、羟基、卤素、氨基、氰基、C1-6烷基或C1-6烷基-O-;优选地,R2为氢。
在某一方案中,L为连接键。
在某一方案中,R3为C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、3-7元杂环基、被一个或多个R3e取代的3-7元杂环基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;优选地,R3为C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、被一个或多个R3e取代的3-7元杂环基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;更优选地,R3为被一个或多个R3e取代的3-7元杂环基;当取代基为多个时,相同或不同。
在某一方案中,每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一 个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、C2-6炔基或-C(=O)-R9;进一步地,每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C2-6炔基或-C(=O)-R9;较佳地,每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-或-C(=O)-R9;优选地,每个R3d、R3e和R3h各自独立地为羟基、-N(R7)2、C1-6烷基-O-、被一个或多个R3- b取代的C1-6烷基-O-、C3-7环烷基-O-或-C(=O)-R9;进一步优选地,每个R3d、R3e和R3h各自独立地为羟基、卤素、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3- j取代的C3-7环烷基-O-或-C(=O)-R9;更优选地,每个R3d、R3e和R3h各自独立地为羟基、卤素、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-或-C(=O)-R9;当取代基为多个时,相同或不同。
在某一方案中,每个R3-a、R3-b和R3-j各自独立地为氘、羟基、卤素或C3-7环烷基;进一步地,每个R3-a和R3-b各自独立地为氘、羟基、卤素或C3-7环烷基;更进一步地,每个R3-b独立地为氘、羟基或卤素。
在某一方案中,X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;进一步地,X为连接键、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;优选地,X为-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连。
在某一方案中,R4为氢、C1-6烷基、C1-6烷基-O-、氰基、卤素、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;优选地,R4为氢、C1-6烷基、氰基、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基或被一个或多个R4b取代的C3-7环烷基;更优选地,R4为氢、C1-6烷基、氰基、羟基、-N(R7)2、被一个或多个R4a取代的3-7元杂环基或被一个或多个R4b取代的C3-7环烷基;进一步优选地,R4为-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;最优选地,R4为-N(R7)2、3-7元杂环基或被一个或多个R4a取代的3-7元杂环基;当取代基为多个时,相同或不同。
在某一方案中,每个R4a和R4b各自独立地为羟基、卤素、氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;优选地,每个R4a和R4b各自独立地为氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;更优选地,每个R4a和R4b各自独立地为C1-6烷基或氨基;进一步优选地,每个R4a独立地为C1- 6烷基。
在某一方案中,R5为氢、羟基、卤素、氨基、氰基或C1-6烷基;优选地,R5为氢或C1-6烷基;更优选地,R5为C1-6烷基。
在某一方案中,R6为氧代(=O)。
在某一方案中,每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基或被一个或多个R7a取代的3-7元杂环基;进一步地,每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3- 7元杂环基;更进一步地,每个R7独立地为氢或C1-6烷基;当取代基为多个时,相同或不同。
在某一方案中,每个R7b独立地为氘、卤素、羟基或氰基;优选地,每个R7b独立地为氘或羟基;更优选地,每个R7b独立地为氘。
在某一方案中,每个R9独立地为氢或C1-6烷基;优选地,每个R9独立地为C1-6烷基。
在某一方案中,环D为3-7元杂环基。
在某一方案中,R10为C1-6烷基;优选地,R10为甲基。
在某一方案中,R11为C1-6烷基;优选地,R11为甲基。
在某一方案中,所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中,
环A为C6-12芳基;
每个R1独立地为卤素、羟基、-N(R7)2、C1-6烷基、氰基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1a取代的C1-6烷基或被一个或多个R1b取代的C1-6烷基-O-;当取代基为多个时,相同或不同;
每个R1a和R1b各自独立地为卤素;
m为0、1、2或3;
R2为氢、羟基、卤素、氨基、氰基、C1-6烷基或C1-6烷基-O-;
L为连接键;
R3为C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、3-7元杂环基、被一个或多个R3e取代的3-7元杂环基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;当取代基为多个时,相同或不同;
每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、C2-6炔基或-C(=O)-R9;当取代基为多个时,相同或不同;
每个R3-a、R3-b和R3-j各自独立地为氘、羟基、卤素或C3-7环烷基;
X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C4亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;
R4为氢、C1-6烷基、C1-6烷基-O-、氰基、卤素、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
每个R4a和R4b各自独立地为羟基、卤素、氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;
R5为氢、羟基、卤素、氨基、氰基或C1-6烷基;
R6为氧代(=O);
每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基或被一个或多个R7a取代的3-7元杂环基;当取代基为多个时,相同或不同;
每个R7a和R7b独立地为氘、卤素、羟基或氰基;
每个R9独立地为氢或C1-6烷基;
环D为3-7元杂环基;
R10为C1-6烷基;
R11为C1-6烷基;
其中,不为氢;
所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”。
在某一方案中,所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中,
环A为C6-12芳基;
每个R1独立地为卤素、羟基、-N(R7)2、C1-6烷基、氰基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1a取代的C1-6烷基或被一个或多个R1b取代的C1-6烷基-O-;当取代基为多个时,相同或不同;
每个R1a和R1b各自独立地为卤素;
m为0、1、2或3;
R2为氢、羟基、卤素、氨基、氰基、C1-6烷基或C1-6烷基-O-;
L为连接键;
R3为C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、3-7元杂环基、被一个或多个R3e取代的3-7元杂环基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;当取代基为多个时,相同或不同;
每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C2-6炔基或-C(=O)-R9;当取代基为多个时,相同或不同;
每个R3-a和R3-b各自独立地为氘、羟基或卤素;
X为连接键、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C4亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;
R4为氢、C1-6烷基、C1-6烷基-O-、氰基、卤素、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
每个R4a和R4b各自独立地为羟基、卤素、氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;
R5为氢、羟基、卤素、氨基、氰基或C1-6烷基;
R6为氧代(=O);
每个R7独立地为氢或C1-6烷基;
每个R9独立地为氢或C1-6烷基;
环D为3-7元杂环基;
R10为C1-6烷基;
R11为C1-6烷基。
在某一方案中,所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中,
环A为C6-12芳基;
每个R1独立地为卤素、羟基、-N(R7)2、C1-6烷基、氰基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1a取代的C1-6烷基或被一个或多个R1b取代的C1-6烷基-O-;当取代基为多个时,相同或不同;
每个R1a和R1b各自独立地为卤素;
m为0、1、2或3;
R2为氢、羟基、卤素、氨基、氰基、C1-6烷基或C1-6烷基-O-;
L为连接键;
R3为C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、3-7元杂环基、被一个或多个R3e取代的3-7元杂环基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;当取代基为多个时,相同或不同;
每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、C2-6炔基或-C(=O)-R9;当取代基为多个时,相同或不同;
每个R3-a、R3-b和R3-j各自独立地为氘、羟基、卤素或C3-7环烷基;
X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;
R4为氢、C1-6烷基、C1-6烷基-O-、氰基、卤素、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
每个R4a和R4b各自独立地为羟基、卤素、氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;
R5为氢、羟基、卤素、氨基、氰基或C1-6烷基;
R6为氧代(=O);
每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基或被一个或多个R7a取代的3-7元杂环基;当取代基为多个时,相同或不同;
每个R7a和R7b独立地为氘、卤素、羟基或氰基;
每个R9独立地为氢或C1-6烷基;
环D为3-7元杂环基;
R10为C1-6烷基;
R11为C1-6烷基;
其中,不为氢;
所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”。
在某一方案中,所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中,
环A为C6-12芳基;
每个R1独立地为卤素、羟基、-N(R7)2、C1-6烷基、氰基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被 一个或多个R1a取代的C1-6烷基或被一个或多个R1b取代的C1-6烷基-O-;当取代基为多个时,相同或不同;
每个R1a和R1b各自独立地为卤素;
m为0、1、2或3;
R2为氢、羟基、卤素、氨基、氰基、C1-6烷基或C1-6烷基-O-;
L为连接键;
R3为C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、被一个或多个R3e取代的3-7元杂环基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;当取代基为多个时,相同或不同;
每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、C2-6炔基或-C(=O)-R9;当取代基为多个时,相同或不同;
每个R3-a、R3-b和R3-j各自独立地为氘、羟基、卤素或C3-7环烷基;
X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;
R4为氢、C1-6烷基、C1-6烷基-O-、氰基、卤素、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
每个R4a和R4b各自独立地为羟基、卤素、氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;
R5为氢、羟基、卤素、氨基、氰基或C1-6烷基;
R6为氧代(=O);
每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基或被一个或多个R7a取代的3-7元杂环基;当取代基为多个时,相同或不同;
每个R7a和R7b独立地为氘、卤素、羟基或氰基;
每个R9独立地为氢或C1-6烷基;
环D为3-7元杂环基;
R10为C1-6烷基;
R11为C1-6烷基;
其中,不为氢;
所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”。
在某一方案中,所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中,
环A为C6-12芳基;
每个R1独立地为卤素、羟基、-N(R7)2、C1-6烷基、氰基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1a取代的C1-6烷基或被一个或多个R1b取代的C1-6烷基-O-;当取代基为多个时,相同或 不同;
每个R1a和R1b各自独立地为卤素;
m为0、1、2或3;
R2为氢、羟基、卤素、氨基、氰基、C1-6烷基或C1-6烷基-O-;
L为连接键;
R3为C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、3-7元杂环基、被一个或多个R3e取代的3-7元杂环基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;当取代基为多个时,相同或不同;
每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、C2-6炔基或-C(=O)-R9;当取代基为多个时,相同或不同;
每个R3-a、R3-b和R3-j各自独立地为氘、羟基、卤素或C3-7环烷基;
X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;
R4为-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
每个R4a和R4b各自独立地为羟基、卤素、氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;
R5为氢、羟基、卤素、氨基、氰基或C1-6烷基;
R6为氧代(=O);
每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基或被一个或多个R7a取代的3-7元杂环基;当取代基为多个时,相同或不同;
每个R7a和R7b独立地为氘、卤素、羟基或氰基;
每个R9独立地为氢或C1-6烷基;
环D为3-7元杂环基;
R10为C1-6烷基;
R11为C1-6烷基;
所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”。
在某一方案中,所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中,
环A为C6-12芳基;
每个R1独立地为卤素、氰基、C1-6烷基或被一个或多个R1a取代的C1-6烷基;当取代基为多个时,相同或不同;
每个R1a独立地为卤素;
m为2;
R2为氢;
L为连接键;
R3为被一个或多个R3e取代的3-7元杂环基;当取代基为多个时,相同或不同;
每个R3e独立地为羟基、卤素、-N(R7)2、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3- 7环烷基-O-或-C(=O)-R9;当取代基为多个时,相同或不同;
每个R3-b独立地为氘、羟基或卤素;
X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,“-O-(C1-C4亚烷基)-”中“C1-C4亚烷基”与R4相连;
R4为氢、C1-6烷基、氰基、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
每个R4a和R4b各自独立地为氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;
R5为氢或C1-6烷基;
R6为氧代(=O);
每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基;当取代基为多个时,相同或不同;
每个R7b独立地为氘或羟基;
每个R9独立地为C1-6烷基;
环D为3-7元杂环基;
R10为C1-6烷基;
R11为C1-6烷基;
其中,不为氢;
所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”。
在某一方案中,所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中,
环A为C6-12芳基;
每个R1独立地为卤素、氰基、C1-6烷基或被一个或多个R1a取代的C1-6烷基;当取代基为多个时,相同或不同;
每个R1a独立地为卤素;
m为2;
R2为氢;
L为连接键;
R3为被一个或多个R3e取代的3-7元杂环基;当取代基为多个时,相同或不同;
每个R3e独立地为羟基、卤素、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-或-C(=O)-R9;当取代基为多个时,相同或不同;
每个R3-b独立地为氘或卤素;
X为连接键、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,“-O-(C1-C4亚烷基)-”中“C1-C4亚烷基”与R4相连;
R4为氢、C1-6烷基、氰基、羟基、-N(R7)2、被一个或多个R4a取代的3-7元杂环基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
每个R4a和R4b各自独立地为氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;
R5为氢或C1-6烷基;
R6为氧代(=O);
每个R7独立地为氢或C1-6烷基;
每个R9独立地为C1-6烷基;
环D为3-7元杂环基;
R10为C1-6烷基;
R11为C1-6烷基。
在某一方案中,所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中,
环A为C6-12芳基,例如
每个R1独立地为卤素、氰基、C1-6烷基或被一个或多个R1a取代的C1-6烷基;当取代基为多个时,相同或不同;
每个R1a独立地为卤素;
m为2;
R2为氢;
L为连接键;
R3为被一个或多个R3e取代的3-7元杂环基;当取代基为多个时,相同或不同;
每个R3e独立地为羟基、-N(R7)2、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-或-C(=O)-R9;当取代基为多个时,相同或不同;
每个R3-b独立地为氘、羟基或卤素;
X为-(C1-C4亚烷基)-、-O-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,“-O-(C1-C4亚烷基)-”中“C1-C4亚烷基”与R4相连;
R4为-N(R7)2、3-7元杂环基或被一个或多个R4a取代的3-7元杂环基;当取代基为多个时,相同或不同;
每个R4a独立地为C1-6烷基;
R5为氢或C1-6烷基;
R6为氧代(=O);
每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基;当取代基为多个时,相同或不同;
每个R7b独立地为氘或羟基;
每个R9独立地为C1-6烷基;
环D为3-7元杂环基;
R10为甲基;
R11为甲基;
所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”。
在某一方案中,所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中,
环A为C6-12芳基;
每个R1独立地为卤素、氰基、C1-6烷基或被一个或多个R1a取代的C1-6烷基;当取代基为多个时,相同或不同;
每个R1a独立地为卤素;
m为2;
R2为氢;
L为连接键;
R3为被一个或多个R3e取代的3-7元杂环基;当取代基为多个时,相同或不同;
每个R3e独立地为羟基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-或-C(=O)-R9;当取代基为多个时,相同或不同;
每个R3-b独立地为氘;
X为-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,“-O-(C1-C4亚烷基)-”中“C1-C4亚烷基”与R4相连;
R4为-N(R7)2或被一个或多个R4a取代的3-7元杂环基;当取代基为多个时,相同或不同;
每个R4a独立地为C1-6烷基;
R5为氢或C1-6烷基;
R6为氧代(=O);
每个R7独立地为氢或C1-6烷基;
每个R9独立地为C1-6烷基;
环D为3-7元杂环基;
R10为甲基;
R11为甲基。
在某一方案中,环A为
在某一方案中,每个R1独立地为卤素、氰基、C1-6烷基或被一个或多个R1a取代的C1-6烷基,当取代基为多个时,相同或不同;例如:R1为F、Cl、-CHF2、-CF3、氰基、-CH3或-CF2CH3,优选为F、-CHF2、-CH3或氰基。
在某一方案中,R2为氢。
在某一方案中,R5为氢或C1-6烷基,例如:R5为氢或甲基。
在某一方案中,X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2- C6亚炔基)-,“-O-(C1-C4亚烷基)-”中“C1-C4亚烷基”与R4相连;例如:连接键、 又例如:连接键、 其中,f端与R4相连。
在某一方案中,可为又可为 还可为 例如又例如
在某一方案中,当环D为3-7元杂环基时,环D可为1,2-二氢吡啶基,例如为 “c”表示该原子位于B环N原子的间位。
在某一方案中,“c”表示该原子位于B环N原子的间位。
在某一方案中,可为 又可为 例如:
在某一方案中,当R3为C3-7环烷基或被一个或多个R3d取代的C3-7环烷基时,所述的R3可为
在某一方案中,当R3为3-7元杂环基或被一个或多个R3e取代的3-7元杂环基时,所述的R3可为 又可为 例如 又例如 再例如优选
在某一方案中,可为 又可为 例如 又例如 再例如 优选
在某一方案中,R4可为氢、甲基、甲氧基、氰基、卤素、羟基、-NH2、-NHCH3、-N(CH3)2、-N(CH2CH3)2、-N(CD3)2 又为氢、甲基、甲氧基、氰基、卤素、羟基、-NH2、-NHCH3、-N(CH3)2
在某一方案中,可为甲基、甲氧基、氰基、卤素、 又可为氢、甲基、甲氧基、氰基、卤素、 例如甲基、氰基、 优选 还优选
在某一方案中,所述如式I所示的稠环化合物为
在某一方案中,R10为甲基。
在某一方案中,R11为甲基。
在某一方案中,当R4为氰基时,
在某一方案中,所述式I所示的化合物为以下任一化合物:
























本发明还提供了一种如前所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐 的制备方法,其中,当“c”表示该原子位于B环N原子的间位时,其制备方法包括以下步骤:中间体I-A1与原料N-苯基双(三氟甲烷磺酰)亚胺或三氟甲磺酸酐反应得到中间体I-A2,I-A2再与原料I-A3发生偶联反应,得到中间体I-A4,I-A4再与原料I-A5发生取代反应或钯催化的Buchwald偶联反应,得到通式I-A化合物;
若I-A4与I-A5发生取代反应,其是在碱作用下进行,所述碱包括但不限于碳酸铯、碳酸钠、碳酸钾、三乙胺、二异丙基乙胺等;
若I-A4与I-A5发生Buchwald偶联反应,其是在催化剂、碱存在下进行,所述催化剂包括但不限于RuPhos Pd G3、BrettPhos Pd G3、XPhos Pd G3、XantPhos Pd G3、RuPhos Pd G4、BrettPhos Pd G4等,所述碱包括但不限于碳酸铯、碳酸钠、磷酸钾、碳酸钠、叔丁醇钾、叔丁醇钠等;
其中:环A、m、X、L、R1、R2、R3、R4、R5、R10和R11的定义如上任一方案所述;
RG为氢、-B(OH)2-ZnBr或-ZnI。
本发明还提供了一种如式I-A1所示的化合物:
其中,L、R2、R3、R5和R10的定义如上任一方案所述。
在某一方案中,所述如式I-A1所示的化合物为如下任一化合物:
本发明还提供了一种如式I-A2所示的化合物:
其中,L、R2、R3、R5和R10的定义如上任一方案所述。
在某一方案中,所述如式I-A2所示的化合物为如下任一化合物:
本发明还提供了如下任一化合物:


本发明提供了一种药物组合物,所述药物组合物包含:
(1)(治疗有效量的)物质Q,所述物质Q为式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,及
(2)药学可接受的辅料。
本发明提供了一种物质Q或如前所述的药物组合物在制备SOS1抑制剂中的应用,所述物质Q为式I所示的稠环化合物、其立体异构体或其药学上可接受的盐。
所述的应用中,较佳地,所述的SOS1抑制剂可用于哺乳动物生物体内;也可用于生物体外,主要作为实验用途,例如:作为标准样或对照样提供比对,或按照本领域常规方法制成试剂盒,为化合物抑制SOS1的效果提供快速检测。
本发明提供了一种物质Q或如前所述的药物组合物在制备药物中的应用;所述物质Q为如前所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐;所述药物可为治疗和/或预防与SOS1活性或表达量相关的疾病的药物。
所述的应用中,较佳地,所述与SOS1活性或表达量相关的疾病选自非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、结肠癌、甲状腺癌、黑色素瘤、胚胎性横纹肌肉瘤、皮肤颗粒细胞肿瘤、肝癌、直肠癌、膀胱癌、咽喉癌、乳腺癌、前列腺癌、神经胶质细胞瘤、卵巢癌、头颈部鳞癌、宫颈癌、食管癌、肾癌、皮肤癌、淋巴瘤、胃癌、胆管癌、子宫癌、子宫内膜癌、尿路上皮癌、急性髓系白血病、骨髓纤维化、B细胞淋巴瘤、单核细胞白血病、脾大性红细胞增多、嗜酸性白细胞增多综合征和多发性骨髓癌,以及和SOS1遗传性突变相关的疾病,包括但不限于一型神经纤维瘤、努南氏综合征、多发雀斑样恁型努南氏综合征、毛细血管畸形—动静脉畸形综合征、心-面-皮肤综合征、克斯提洛氏综合征、雷吉士综合征和一型遗传性牙龈纤维瘤。
本发明提供了一种物质Q或如前所述的药物组合物在制备药物中的应用;所述物质Q为如前所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐;所述药物为治疗和/或预防以下各类疾病的药物;所述疾病选自非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、结肠癌、甲状腺癌、黑色素瘤、胚胎性横纹肌肉瘤、皮肤颗粒细胞肿瘤、肝癌、直肠癌、膀胱癌、咽喉癌、乳腺癌、前列腺癌、神经胶质细胞瘤、卵巢癌、头颈部鳞癌、宫颈癌、食管癌、肾癌、皮肤癌、淋巴瘤、胃癌、胆管癌、子宫癌、子宫内膜癌、尿路上皮癌、急性髓系白血病、骨髓纤维化、B细胞淋巴瘤、单核细胞白血病、脾大性红细胞增多、嗜酸性白细胞增多综合征和多发性骨髓癌,以及一型神经纤维瘤、努南氏综合征、多发雀斑样恁型努南氏综合征、毛细血管畸形—动静脉畸形综合征、心-面-皮肤综合征、克斯提洛氏综合征、雷吉士综合征和一型遗传性牙龈纤维瘤。
本发明提供了一种抑制SOS1的方法,其包括向患者(例如人类)施用治疗有效量的物质Q或如前所述的药物组合物;
所述物质Q为如前所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐。
本发明提供了一种治疗和/预防疾病的方法,其包括向患者(例如人类)施用治疗有效量的物质Q或如前所述的药物组合物;
所述物质Q为如前所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐;
所述疾病为与SOS1活性或表达量相关的疾病。
所述治疗和/预防疾病的方法中,较佳地,所述与SOS1活性或表达量相关的疾病选自非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、结肠癌、甲状腺癌、黑色素瘤、胚胎性横纹肌肉瘤、皮肤颗粒细胞肿瘤、肝癌、直肠癌、膀胱癌、咽喉癌、乳腺癌、前列腺癌、神经胶质细胞瘤、卵巢癌、头颈部鳞癌、宫颈癌、食管癌、肾癌、皮肤癌、淋巴瘤、胃癌、胆管癌、子宫癌、子宫内膜癌、尿路上皮癌、急性髓系白血病、骨髓纤维化、B细胞淋巴瘤、单核细胞白血病、脾大性红细胞增多、嗜酸性白细胞增多综合征和多发性骨髓癌,以及和SOS1遗传性突变相关的疾病,包括但不限于一型神经纤维瘤、努南氏综合征、多发雀斑样恁型努南氏综合征、毛细血管畸形—动静脉畸形综合征、心-面-皮肤综合征、克斯提洛氏综合征、雷吉士综合征和一型遗传性牙龈纤维瘤。
如无特别说明,本发明所用术语具有如下含义:
术语“卤素”是指氟、氯、溴或碘。
术语“烷基”是指仅由碳原子和氢原子组成、具有指定的碳原子数(例如C1-10,优选C1-6,更优选C1-4)的饱和直链或支链烃基。烷基包括但不限于甲基、乙基、正丙基、异丙基、正丁基、叔丁基、异丁基、仲丁基、正戊基或正己基等。
术语“亚烷基”表示从饱和的直链或支链烃基中去掉两个氢原子所得到的饱和的二价烃基基团;即烷基中的一个氢被取代,烷基的定义如上所述。亚烷基基团的实例包括亚甲基(-CH2-),亚乙基{包括-CH2CH2-或-CH(CH3)-},亚正丙基{包括-CH2CH2CH2-},亚异丙基{包括-CH(CH3)CH2-、-CH(CH2CH3)-或-C(CH3)2-}等。
术语“烯基”是指具有至少一个双键的直链或支链的烃链基,仅由碳原子和氢原子组成、具有指定碳原子数(例如C2-10,优选C2-6,更优选C2-4)且通过单键与分子的其余部分连接,其包括顺反异构或Z-E异构;例如,烯基包括但不限于乙烯基(-CH=CH2)、丙烯基(-CH=CH-CH3)、烯丙基(-CH2- CH=CH2)、异丙烯基(-C(CH3)=CH2)、-CH=CH-CH2-CH3、-CH2-CH=CH-CH3、-CH=CH-CH2-CH2-CH3、-CH2-CH=CH-CH2-CH3、-CH=CH-CH(CH3)2、-CH2-CH=C(CH3)2或-CH=CH-(CH2)3-CH3等。
术语“亚烯基”表示从直链或支链烯烃分子中去掉两个氢原子所得到的不饱和的二价基团;即烯基中的一个氢被取代,烯基的定义如上所述。亚烯基基团的实例包括但不限于-CH=CH-、-CH=CH-CH2-等。
术语“炔基”是指具有至少一个三键的直链或支链的烃链基,其仅由碳原子和氢原子组成、具有指定碳原子数(例如C2-10,优选C2-6,更优选C2-4)且通过单键与分子的其余部分连接;例如,炔基包括但不限于乙炔基(-C≡CH)、丙炔基(-C≡C-CH3)、-C≡C-CH2-CH3、-CH2C≡C-CH3、-C≡C-CH2-CH2-CH3、-CH2C≡C-CH2-CH3、-C≡C-CH2-CH2-CH2-CH3、-C≡C-CH2-CH(CH3)2、-C≡C-C(CH3)3、-C≡C-CH(CH3)2或-CH2C≡C-CH2-CH2-CH3等。
术语“亚炔基”是指从直链或支链炔烃分子中去掉两个氢原子所得到的不饱和的二价基团;即炔基中的一个氢被取代,炔基的定义如上所述。亚炔基基团的实例包括但不限于亚乙炔基、亚丙炔基、亚戊炔基等,例如-C≡C-、-CH2-C≡CH、-C≡C-CH2-、-C≡C-CH2-CH2-、-C≡C-CH(CH3)-CH2-、-C≡C-C(CH3)2-。
术语“C1-6烷基-O-”中,C1-6烷基的定义如前所述。
术语“环烷基”意指饱和的单环或多环(例如双环、三环或更多环的桥环、并环(稠环)或螺环体系)的碳环取代基,且其可经由任何适宜的碳原子通过单键与分子的其余部分连接;例如具有3至15个环碳原子,优选具有3至10个环碳原子,更优选具有3至7个环碳原子;例如C3-7环烷基包括但不限于环丙基、环丁基、环戊基、环己基、环庚基或等。
术语“环烯基”意指具有至少一个双键(如碳碳双键)的单环或多环(例如双环、三环或更多环的桥环、并环(稠环)或螺环体系)的非芳香性的环烃基,且其可经由任何适宜的碳原子通过单键与分子的其余部分连接;例如具有3至15个环碳原子,优选具有3至10个环碳原子,更优选具有3至7个环碳原子;例如C3-7环烯基包括但不限于环丙烯基、环丁烯基、环戊烯基、环己烯基或环庚烯基。
术语“杂环基”是指由2至14个(优选2至6个)碳原子以及1至6个选自N、O、S、S(=O)和S(=O)2的杂原子或杂原子基团组成的稳定的3-20元(优选3-10元,进一步优选3-7元,更进一步优选4-7元,最优选6元)的饱和或部分不饱和的非芳香性的单环或多环(例如双环、三环或更多环的桥环、并环(稠环)或螺环体系)杂环烃基;优选含有1至3个选自N、O和S的杂原子的3-7元杂环基,进一步优选“杂原子个数为1个或2个,杂原子种类独立选自N和O”的4-7元杂环基,所述的4-7元杂环基可为4-7元单环杂环基、5-7元桥环杂环基或5-7元螺环杂环基;3-7元杂环基的实例包括但不限于氮杂环丙基、环氧乙烷基、环氧丙烷基、硫杂环丁烷基、四氢噻吩基、噻唑烷基、异噻唑烷基、噁唑烷基、异噁唑烷基、吡唑烷基、哌啶基、四氢吡喃基、哌嗪基、四氢吡啶基、氮杂环丁基、吡咯烷基、吗啉基、2-氧杂-5-氮杂双环[2.2.1]庚基、2H-吡喃基、氧氮杂卓基、二氮杂卓基、硫氮杂卓基、氮杂双环庚基、噻恶烷基、二氢呋喃基、咪唑啉基、咪唑烷基、硫杂环己基、三嗪烷基、二硫杂环己基、二硫杂环戊基、二氧杂环己基、2,6-二氮杂螺[3.3]庚基、1,2-二氢吡啶基、高哌嗪基、硫代吗啉基、 噻喃基、四氢呋喃基、4H-吡喃基或二氢吡咯基,例如:
术语“芳基”是指由碳原子组成的满足4n+2规则的共轭烃环体系的芳香基团,每个环均具有芳香性。在某一方案中,“芳基”是指具有6至18个(优选6-12个)碳原子的芳香基团。芳基的实例包括但不限于苯基或萘基等。
术语“杂芳基”意指环内具有2至15个碳原子和1至5个选自氮、氧和硫的杂原子的5-20元(优选5-12元,进一步优选5-10元,更进一步优选5-8元,最优选5-6元)共轭环系基团。除非本说明书中另外特别指明,否则杂芳基可为单环、双环、三环或更多环的环体系,还可以与上文所定义的环烷基、环烯基或杂环基稠合,条件是杂芳基经由芳香环上的原子通过单键与分子的其余部分连接。在某一方案中,术语“杂芳基”是指含有杂原子的芳香基团,每个环均具有芳香性;优选杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的5-10元杂芳基或5-6元杂芳基。杂芳基的实例包括但不限于噻吩基、咪唑基、吡唑基、噻唑基、噁唑基、二唑基、噁二唑基、异噁唑基、吡啶基、嘧啶基、吡嗪基、哒嗪基、苯并咪唑基(例如为)、苯并吡唑基、吲哚基、呋喃基、吡咯基、三唑基、四唑基、三嗪基、吲嗪基、异唑基、噻二唑基、异吲哚基、吲唑基、异吲唑基、嘌呤基、喹啉基、异喹啉基、二氮萘基、萘啶基、喹噁啉基、蝶啶基、咔唑基、咔啉基、菲啶基、菲咯啉基、吖啶基、吩嗪基、异噻唑基、苯并噻唑基、苯并异噻唑基、苯并噻吩基、噁三唑基、噌啉基、喹唑啉基、中氮茚基、邻二氮杂菲基、异噁唑基、吩噁嗪基、吩噻嗪基、苯并恶唑基或苯并异唑基。
术语“环烷基-O-”中,环烷基的定义如前所述。
术语“环烯基-O-”中,环烯基的定义如前所述。
术语“杂环基-O-”中,杂环基的定义如前所述。
基团末端的“-”是指该基团通过该位点与分子中的其他片段连接。
应该理解,在本发明中使用的单数形式,如“一种”,包括复数指代,除非另有规定。
术语“一个或多个”是指即1、2、3、4、5、6、7、8、9或更多。
除非另有说明,本发明采用质谱、元素分析的传统方法,各步骤和条件可参照本领域常规的操作步骤和条件。
除非另有指明,本发明采用分析化学、有机合成化学和光学的标准命名及标准实验室步骤和技术。在某些情况下,标准技术被用于化学合成、化学分析。
另外,需要说明的是,除非以其他方式明确指出,在本发明中所采用的描述方式“…独立地为”应做广义理解,是指所描述的各个个体之间是相互独立的,可以独立地为相同或不同的具体基团。更详细地,描述方式“…独立地为”既可以是指在不同基团中,相同符号之间所表达的具体选项之间互相不影响;也可以表示在相同的基团中,相同符号之间所表达的具体选项之间互相不影响。
应该理解,在本发明中使用的“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个”中“杂原子种类独立选自N、O和S”可以理解为“杂原子种类独立选自N、O和S中的一种或多种”。在本发明中使用的“杂原子个数为1个或2个,杂原子种类独立选自N和O”中“杂原子种类独立选自N和O”可以理解为“杂原子种类独立选自N和O中的一种或两种”。
本领域技术人员可以理解,根据本领域中使用的惯例,本发明描述基团的结构式中所使用的是指,相应的基团R通过该位点与化合物中的其它片段、基团进行连接。
除非另有规定,本文使用的所有技术术语和科学术语具有要求保护主题所属领域的标准含义。倘若对于某术语存在多个定义,则以本文定义为准。
如本文中所使用的,本发明式I所示的稠环化合物可以含有一个或多个手性中心,并以不同的光学活性形式存在。当化合物含有一个手性中心时,化合物包含对映异构体。本发明包括这两种异构体和异构体的混合物,如外消旋混合物。对映异构体可以通过本专业已知的方法拆分,例如结晶以及手性色谱等方法。当式I所示的稠环化合物含有多于一个手性中心时,可以存在非对映异构体。本发明包括拆分过的光学纯的特定异构体以及非对映异构体的混合物。非对映异构体可由本专业已知方法拆分,比如结晶以及制备色谱。术语“立体异构体”包括构象异构体和构型异构体,其中构型异构体主要包括顺反异构体和旋光异构体。本发明所述化合物可以以立体异构体的形式存在,并因此涵盖所有可能的立体异构体形式,包括但不限于顺反异构体、对映异构体、非对映异构体、阻转异构体等,本发明所述化合物也可以以前述的立体异构体的任何组合或任何混合物,例如内消旋体、外消旋体、阻转异构体的等量混合物等形式存在。例如单一对映异构体,单一非对映异构体或以上的混合物,或单一阻转异构体或其混合物。当本发明所述的化合物含有烯烃双键时,除非特别说明,否则其包括顺式异构体和反式异构体,以及其任何组合。本发明的阻转异构体为基于分子内旋转受限制而产生的轴向或平面手性的立体异构体。
如前所述,本发明提供了上述各类结构所示化合物,或其顺反异构体、内消旋体、外消旋体、对映异构体、非对映异构体、阻转异构体、互变异构体或其混合物形式,其中“其混合物形式”包括前述的任一立体异构体(例如顺反异构体、对映异构体、非对映异构体、阻转异构体)、互变异构体和/或混合物(内消旋体、外消旋体)之间的任意形式的混合,例如顺反异构体的混合物,对映异构体和非对映异构体的混合物,非对映异构体的混合物,阻转异构体的混合物,或顺反异构体和外消旋体的混合,对映异构体和非对映异构体混合物的混合、顺反异构体和互变异构体的混合物、阻转异构体与非对映异构体混合物的混合等。
如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐意图涵盖如式I所示的稠环化合 物、其立体异构体或其药学上可接受的盐的任何同位素标记的(或“放射性标记的”)变体。这种变体可以是如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中一个或多个原子被原子质量或质量数不同于通常在自然界中所发现的原子质量或质量数的原子置换而得到。所使用的放射性核素将取决于该放射性标记的变体的具体应用。举例来说,对于体外受体标记和竞争测定,3H或14C常常是有用的。对于放射成像应用,11C或18F常常是有用的。
本发明化合物的特定同位素变体,特别是其中已经结合一种或多种放射性同位素的同位素变体,可有益于例如考察作用机制或在体内的活性组份分布;由于相对容易的可制备性和可检测性,标记有3H或14C同位素的化合物特别适用于此目的。另外,纳入同位素如氘,由于该化合物具有更好的代谢稳定性,例如延长体内的半衰期或降低所需的有效剂量,可产生特别的治疗益处;因此本发明化合物的这种修饰还可在一些情况下构成本发明的优选实施方案。本发明化合物的同位素变体可通过本领域技术人员已知的方法,例如通过以下描述的方法及操作实施例中描述的方法,通过使用相应的同位素修饰的特定试剂和/或起始化合物来制备。
在本发明中,“药物组合物”是指包含本发明化合物与本领域通常接受的用于将生物活性化合物输送至哺乳动物(例如人)的介质的制剂。该介质包括药学上可接受的载体。药物组合物的目的是促进生物体的给药,利于活性成分的吸收进而发挥生物活性。
在本发明中,“药学上可接受的”是指不影响本发明化合物的生物活性或性质的物质(如药用辅料),并且相对无毒,即该物质可施用于个体而不造成不良的生物反应或以不良方式与组合物中包含的任意组分相互作用。
术语“药用辅料”、“药学可接受的辅料”或“药学上可接受的载体”是指生产药品和调配处方时使用的赋形剂和附加剂,是除活性成分以外,包含在药物制剂中的所有物质。可参见中华人民共和国药典(2015年版)四部、或、Handbook of Pharmaceutical Excipients(Raymond C Rowe,2009 Sixth Edition)。辅料主要用于提供一个安全、稳定和功能性的药物组合物,还可以提供方法,使受试者接受给药后活性成分以所期望速率溶出,或促进受试者接受组合物给药后活性成分得到有效吸收。所述的药用辅料可以是惰性填充剂,或者提供某种功能,例如稳定该组合物的整体pH值或防止组合物活性成分的降解。所述的药用辅料可以包括下列辅料中的一种或多种:粘合剂、助悬剂、乳化剂、稀释剂、填充剂、成粒剂、胶粘剂、崩解剂、润滑剂、抗粘着剂、助流剂、润湿剂、胶凝剂、吸收延迟剂、溶解抑制剂、增强剂、吸附剂、缓冲剂、螯合剂、防腐剂、着色剂、矫味剂和甜味剂。
本发明的药物组合物可根据公开的内容使用本领域技术人员已知的任何方法来制备。例如,常规混合、溶解、造粒、乳化、磨细、包封、包埋或冻干工艺。
当用作药物时,所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐可以以药物组合物的任何形式给药。这些组合物可根据药学领域熟知的方法制备,可以各种途径施用,视需要局部或系统性治疗和要治疗的区域而定。给予可以是局部(包括表皮和透皮,眼部和粘膜,包括鼻内,阴道和直肠递送),肺(例如,通过粉末或气溶胶吸入或吹入,包括通过喷雾器;气管内或鼻内),口服(固体和液体制剂)或胃肠外给予形式。固体口服制剂的实例包括但不限于粉末、胶囊、囊片、软胶囊剂和片剂。口服或粘膜给药的液体制剂实例包括但不限于悬浮液、乳液、酏剂和溶液。局部用制剂的实例包括但不限于乳剂、凝胶剂、软膏剂、乳膏剂、贴剂、糊剂、泡沫剂、洗剂、滴剂或血清制剂。 胃肠外给药的制剂实例包括但不限于注射用溶液、可以溶解或悬浮在药学上可接受载体中的干制剂、注射用悬浮液和注射用乳剂。外用给药的药物组合物和制剂可包括透皮贴片、油膏剂、乳液、软膏剂、凝胶、滴剂、栓剂、喷剂、液体和粉末。所述的药物组合物的其它合适制剂的实例包括但不限于滴眼液和其他眼科制剂;气雾剂:如鼻腔喷雾剂或吸入剂。口服给药可以包括配制为每日一次或每日两次(BID)给药的剂型。胃肠外给药包括静脉内、动脉内、皮下、腹膜内肌肉内或注射或输液;或颅内如鞘内或心室内给药。胃肠外给药可以单次推注剂量形式,或可以是通过连续灌注泵。常规药学载体、水、粉末或油状基底、增稠剂等可能是必须或需要的。包括本发明的药物组合物还可以是控释或延迟释放剂型(例如脂质体或微球)。
术语“治疗”指治疗性疗法或缓解性措施。涉及具体病症时,治疗指:(1)缓解疾病或者病症的一种或多种生物学表现,(2)干扰(a)导致或引起病症的生物级联中的一个或多个点或(b)病症的一种或多种生物学表现,(3)改善与病症相关的一种或多种症状、影响或副作用,或者与病症或其治疗相关的一种或多种症状、影响或副作用,或(4)减缓病症或者病症的一种或多种生物学表现发展。“治疗”也可以指与不接受治疗的期望存活相比延长生存期。
术语“预防”是指获得或发生疾病或障碍的风险降低。
术语“治疗有效量”是指在给予患者时足以有效治疗本文所述的疾病或病症的化合物的量。“治疗有效量”将根据化合物、病症及其严重度、以及欲治疗患者的年龄而变化,但可由本领域技术人员根据需要进行调整。
术语“患者”是指根据本发明的实施例,即将或已经接受了该化合物或组合物给药的任何动物,哺乳动物为优,人类最优。术语“哺乳动物”包括任何哺乳动物。哺乳动物的实例包括但不限于牛、马、羊、猪、猫、狗、小鼠、大鼠、家兔、豚鼠、猴、人等,以人类为最优。
除非另外说明,应当应用本文所使用的下列定义。出于本发明的目的,化学元素与元素周期表CAS版,和《化学和物理手册》,第75版,1994一致。此外,有机化学一般原理可参考"Organic Chemistry",Thomas Sorrell,University Science Books,Sausalito:1999,和"March's Advanced Organic Chemistry”by Michael B.Smith and Jerry March,John Wiley&Sons,New York:2007中的描述,其全部内容通过引用并入本文。
术语“药学上可接受的盐”是指化合物与药学上可接受的(相对无毒、安全、适合于患者使用)酸或碱反应得到的盐。当化合物中含有相对酸性的官能团时,可以通过在合适的惰性溶剂中用足量的药学上可接受的碱与化合物的游离形式接触的方式获得碱加成盐。药学上可接受的碱加成盐包括但不限于钠盐、钾盐、钙盐、铝盐、镁盐、铋盐、铵盐等。当化合物中含有相对碱性的官能团时,可以通过在合适的惰性溶剂中用足量的药学上可接受的酸与化合物的游离形式接触的方式获得酸加成盐。所述药学上可接受的酸包括无机酸和有机酸。具体可参见Berge et al.,"Pharmaceutical Salts",Journal of Pharmaceutical Science 66:1-19(1977)、或、Handbook of Pharmaceutical Salts:Properties,Selection,and Use(P.Heinrich Stahl and Camille G.Wermuth,ed.,Wiley-VCH,2002)。
在不违背本领域常识的基础上,上述各优选条件,可任意组合,即得本发明各较佳实例。
本发明所用试剂和原料均市售可得。
本发明的积极进步效果在于:本发明的如式I所示的稠环化合物对SOS1具有较好地抑制活性; 有望治疗和/或预防与SOS1活性或表达量相关的疾病。
具体实施方式
下面通过实施例的方式进一步说明本发明,但并不因此将本发明限制在所述的实施例范围之中。下列实施例中未注明具体条件的实验方法,按照常规方法和条件,或按照商品说明书选择。
本发明的化合物结构是通过核磁共振(NMR)或/和质谱(MS)来确定的。核磁共振化学位移(δ)以百万分之一(ppm)的单位给出。核磁共振的测定是用布鲁克(Bruker)AVANCE-400核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6)或氘代氯仿(CDCl3),内标为四甲基硅烷(TMS)。质谱的测定是用安捷伦(Agilent)1260-6125B单四极杆液质联用仪,采用的是电喷雾离子源(ESI)。
对于硅胶柱层析,使用的是拜泰齐(Biotage)Selekt快速制备色谱仪以及适当规格的拜泰齐生产的BK-SIL硅胶预填充柱或艾杰尔(Agela)生产的Claricep Flash硅胶预填充柱。
薄层层析色谱硅胶板使用烟台黄海HSGF254或青岛GF254硅胶板,采用的规格是0.15mm~0.20mm,制备薄层层析色谱采用的规格是0.4mm~0.5mm。
制备高效液相色谱(制备HPLC)使用的是沃特世(Waters)公司生产的配备ACQUITY QDa质谱检测器的AutoPurification LC制备系统。制备色谱柱用的是SunFire C18 5μm 19x250mm OBD制备柱。流动相使用不同梯度的水(含0.1%甲酸)-乙腈来洗脱化合物。
本发明中英文缩写如下所示,
Boc:叔丁氧羰基;TIPS:三异丙基硅基;Bn:苄基;DMSO:二甲基亚砜;Tf:三氟甲磺酰基;Ms:甲磺酰基。
实施例1
3-(1-乙酰基-4-羟基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((S)-1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮(化合物1)和3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((S)-1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮(化合物2)
步骤1:4-溴-6-氯-2-甲基吡啶-3-醇
将6-氯-2-甲基吡啶-3-醇(10g,69.6mmol)溶于乙腈(100mL),加入N-溴代丁二酰亚胺(13.6g,76.6mmol)。室温搅拌反应1.5小时。反应物减压浓缩,加入饱和亚硫酸钠水溶液(200mL)和饱和氯化钠水溶液(200mL)稀释,加乙酸乙酯(200mL x 2)萃取。有机相再用饱和氯化钠溶液(300mL)洗涤,无水硫酸钠干燥,减压浓缩,残留物用乙醇(80mL)打浆处理,得到产物4-溴-6-氯-2-甲基吡啶-3-醇,黄色固体(14.0g,收率93%)。ESI-MS m/z:221.9[M+H]+1H NMR(400MHz,DMSO-d6)δ9.92(s,1H),7.78(s,1H),2.40(s,3H).
步骤2:叔丁基(S)-2-((4-溴-6-氯-2-甲基吡啶-3-基)氧基)甲基)吡咯烷-1-羧酸酯
将4-溴-6-氯-2-甲基吡啶-3-醇(2.5g,11.2mmol)和(S)-(-)-1-叔丁氧羰基-2-吡咯烷甲醇(2.7g,13.5mmol)溶于四氢呋喃(50mL),氮气保护,0℃下加入三苯基膦(3.5g,13.5mmol)和偶氮二甲酸二异丙酯(2.7g,13.5mmol)。反应升至室温搅拌2小时。加入水(100mL)稀释,用乙酸乙酯(200mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(石油醚/乙酸乙酯=15:1,体积比)得到产物叔丁基(S)-2-((4-溴-6-氯-2-甲基吡啶-3-基)氧基)甲基)吡咯烷-1-羧酸酯,无色油状物(4.7g,收率86%)。ESI-MS m/z:405.1[M+H]+1H NMR(400MHz,CDCl3)δ7.38(s,1H),4.12(s,1H),4.05-3.75(m,2H),3.42(t,J=6.7Hz,2H),2.52(s,3H),2.36-2.22(m,1H),2.14-1.85(m,3H),1.46(s,9H).
步骤3:叔丁基(S)-2-((6-氯-2-甲基-4-(甲氨基)吡啶-3-基)氧基)甲基)吡咯烷-1-羧酸酯
将叔丁基(S)-2-((4-溴-6-氯-2-甲基吡啶-3-基)氧基)甲基)吡咯烷-1-羧酸酯(5.0g,12.3mmol)溶于二甲亚砜(20mL),加入23%甲胺水溶液(30mL),105℃搅拌反应72小时。反应物冷却至室温,加入水(150mL)稀释,加乙酸乙酯(300mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,得到产物叔丁基(S)-2-((6-氯-2-甲基-4-(甲氨基)吡啶-3-基)氧基)甲基)吡咯烷-1-羧酸酯,无色油状物(4.4g),粗品直接用于下一步反应。ESI-MS m/z:356.2[M+H]+
步骤4:叔丁基(S)-2-((6-氯-5-碘-2-甲基-4-(甲氨基)吡啶-3-基)氧基)甲基)吡咯烷-1-羧酸酯
将叔丁基(S)-2-((6-氯-2-甲基-4-(甲氨基)吡啶-3-基)氧基)甲基)吡咯烷-1-羧酸酯(4.4g, 12.4mmol)溶于乙酸(20mL),加入N-碘代丁二酰亚胺(3.0g,13.3mmol),室温搅拌1.5小时。加入饱和亚硫酸钠水溶液(100mL)稀释,加入饱和碳酸氢钠水溶液中和,加乙酸乙酯(300mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(石油醚/乙酸乙酯=6:1,体积比)得到产物叔丁基(S)-2-((6-氯-5-碘-2-甲基-4-(甲氨基)吡啶-3-基)氧基)甲基)吡咯烷-1-羧酸酯,黄色油状物(4.6g,收率79%)。ESI-MS m/z:482.1[M+H]+1H NMR(400MHz,DMSO-d6)δ5.44(brs,1H),4.01(s,1H),3.72(s,1H),3.63(t,J=11.2Hz,1H),3.27(s,2H),3.08(s,3H),2.24(s,3H),2.03(brs,2H),1.94(s,1H),1.89-1.77(m,1H),1.38(s,9H).
步骤5:叔丁基(S)-2-((6-氯-5-(3-乙氧基-3-氧代丙-1-烯-1-基)-2-甲基-4-(甲氨基)吡啶-3-基)氧基)甲基)吡咯烷-1-羧酸酯
将叔丁基(S)-2-((6-氯-5-碘-2-甲基-4-(甲氨基)吡啶-3-基)氧基)甲基)吡咯烷-1-羧酸酯(3.2g,6.65mmol),醋酸钯(150mg,0.67mmol),三(邻甲基苯基)磷(405mg,1.33mmol),三乙胺(1.3g,13.3mmol)和丙烯酸乙酯(2.0g,20.0mmol),溶于N,N-二甲基甲酰胺(30mL)。氮气保护,90℃搅拌反应4小时。反应物冷却至室温后,加入水(300mL)稀释,加乙酸乙酯(500mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(石油醚/乙酸乙酯=5:1,体积比)得到产物叔丁基(S)-2-((6-氯-5-(3-乙氧基-3-氧代丙-1-烯-1-基)-2-甲基-4-(甲氨基)吡啶-3-基)氧基)甲基)吡咯烷-1-羧酸酯,黄色油状物(2.4g,收率80%)。ESI-MS m/z:454.2[M+H]+1H NMR(400MHz,CDCl3)δ7.75(d,J=16.4Hz,1H),6.13(d,J=16.4Hz,1H),4.27(q,J=7.2Hz,2H),4.09(s,1H),3.95-3.80(m,2H),3.54-3.35(m,2H),2.90(d,J=4.4Hz,3H),2.39(s,3H),2.08(s,2H),2.03-1.95(m,1H),1.94-1.83(m,1H),1.47(s,9H),1.34(t,J=7.2Hz,3H).
步骤6:叔丁基(S)-2-((5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲基)吡咯烷-1-羧酸酯
将叔丁基(S)-2-((6-氯-5-(3-乙氧基-3-氧代丙-1-烯-1-基)-2-甲基-4-(甲氨基)吡啶-3-基)氧基)甲基)吡咯烷-1-羧酸酯(2.4g,5.30mmol)溶于乙醇(20mL),加入甲硫醇钠(408mg,5.83mmol),室温搅拌0.5小时。反应物减压浓缩,向残留物中加入水(100mL)稀释,加乙酸乙酯(150 mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,得到产物叔丁基(S)-2-((5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲基)吡咯烷-1-羧酸酯,黄色油状物(2.0g,收率92%)。ESI-MS m/z:408.2[[M+H]+1H NMR(400MHz,CDCl3)δ8.03(d,J=9.8Hz,1H),6.75(d,J=9.8Hz,1H),4.18(s,1H),3.94(s,3H),3.73(d,J=61.2Hz,2H),3.40(s,2H),2.60(s,3H),2.14(brs,2H),2.02-1.86(m,2H),1.45(s,9H)
步骤7:叔丁基(S)-2-((3-溴-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲基)吡咯烷-1-羧酸酯
将叔丁基(S)-2-((5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲基)吡咯烷-1-羧酸酯(2.0g,4.9mmol)溶于乙酸(30mL),加入N-溴代丁二酰亚胺(1.8g,9.83mmol),反应60℃下搅拌5小时。加入饱和亚硫酸钠水溶液(100mL)稀释,加乙酸乙酯(150mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(石油醚/乙酸乙酯=4:1,体积比)得到产物叔丁基(S)-2-((3-溴-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲基)吡咯烷-1-羧酸酯,黄色固体(600mg,收率25%)。ESI-MS m/z:486.1[M+H]+1H NMR(400MHz,CDCl3)δ8.48(s,1H),4.20(s,1H),4.04(s,3H),3.89-3.73(m,2H),3.50-3.37(m,2H),2.59(s,3H),2.16(brs,2H),2.06-1.92(m,2H),1.47(s,9H).
步骤8:叔丁基(S)-2-((3-(1-乙酰基-4-羟基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲基)吡咯烷-1-羧酸酯
将叔丁基(S)-2-((3-溴-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲基)吡咯烷-1-羧酸酯(600mg,1.23mmol)和1-乙酰哌啶-4-酮(696mg,4.94mmol)溶于四氢呋喃(35mL),氮气保护,于-30℃下滴加碘化钐(49.4mL,4.94mmol,0.1M四氢呋喃溶液)。反应-30℃下搅拌0.5小时。-30℃下加入饱和氯化铵水溶液(100mL)稀释,加乙酸乙酯(300mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(石油醚/乙酸乙酯=1:15,体积比)得到产物叔丁基(S)-2-((3-(1-乙酰基-4-羟基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲 基)吡咯烷-1-羧酸酯,黄色油状物(550mg,收率81%)。ESI-MS m/z:549.2[M+H]+
步骤9:叔丁基(S)-2-((3-(1-乙酰基-4-羟基哌啶-4-基)-5-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲基)吡咯烷-1-羧酸酯
将叔丁基(S)-2-((3-(1-乙酰基-4-羟基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲基)吡咯烷-1-羧酸酯(550mg,1.0mmol),甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(91mg,0.1mmol),(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺(570mg,3.0mmol)和叔丁醇钠(288mg,3.0mmol)溶于甲苯中(15mL),氮气保护,100℃搅拌反应2小时。冷却至室温后,减压浓缩,残留物硅胶柱层析(二氯甲烷/甲醇=25:1,体积比)得到产物叔丁基(S)-2-((3-(1-乙酰基-4-羟基哌啶-4-基)-5-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲基)吡咯烷-1-羧酸酯,黄色油状物(350mg,收率50%)。ESI-MS m/z:702.4[M+H]+1H NMR(400MHz,CDCl3)δ7.67-7.57(m,1H),7.55-7.40(m,2H),7.15(td,J=7.6,2.0Hz,1H),6.92(t,J=55.2,1H),5.84(brs,1H),5.59(brs,1H),5.55-5.34(m,1H),4.61(d,J=13.2Hz,1H),3.91(d,J=7.2Hz,3H),3.76-3.56(m,4H),3.52-3.40(m,1H),3.36(d,J=7.8Hz,2H),3.17(td,J=12.8,2.8Hz,1H),2.33(s,3H),2.28-2.18(m,1H),2.13(d,J=2.8Hz,3H),2.09-2.05(m,1H),1.94(td,J=12.8,4.4Hz,3H),1.84-1.74(m,1H),1.62(d,J=7.0Hz,3H),1.44(s,9H).
步骤10:3-(1-乙酰基-4-羟基哌啶-4-基)-5-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((S)-吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮
将叔丁基(S)-2-((3-(1-乙酰基-4-羟基哌啶-4-基)-5-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲基)吡咯烷-1-羧酸酯(350mg,0.50mmol)溶于二氯甲烷(3mL),加入三氟乙酸(1mL),室温搅拌1小时。减压浓缩,得到3-(1-乙酰基-4-羟基哌啶-4-基)-5-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((S)-吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮粗品,直接用于下一步反应。ESI-MS m/z:602.3[M+H]+
步骤11:3-(1-乙酰基-4-羟基哌啶-4-基)-5-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((S)-1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮(化合物1)
将步骤9得到的3-(1-乙酰基-4-羟基哌啶-4-基)-5-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((S)-吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮粗品溶于四氢呋喃(6mL),加入30%甲醛水溶液(3mL),加入三乙酰氧基硼氢化钠(358mg,0.997mmol),室温下搅拌0.5小时。加入饱和碳酸氢钠水溶液(100mL)稀释,加乙酸乙酯(100mL x 3)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=13:1,体积比)得到化合物1,黄色固体(170mg,收率55%)。ESI-MS m/z:616.4[M+H]+1H NMR(400MHz,CDCl3)δ7.56(d,J=3.2Hz,1H),7.51(q,J=7.6Hz,1H),7.45(t,J=7.6Hz,1H),7.16(t,J=7.6Hz,1H),6.92(t,J=55.2Hz,1H),5.77(d,J=5.6Hz,1H),5.59(p,J=6.8Hz,1H),5.31(t,J=7.6Hz,1H),4.62(d,J=13.2Hz,1H),3.92(s,3H),3.71(d,J=8.5Hz,2H),3.57(brs,1H),3.26(brs,1H),3.17(td,J=12.4,2.4Hz,1H),2.81(s,1H),2.57(s, 3H),2.39(s,3H),2.23(dt,J=13.2,2.4Hz,1H),2.13(d,J=2.4Hz,3H),2.06(dd,J=13.2,2.4Hz,2H),1.94(td,J=12.8,4.8Hz,2H),1.87-1.72(m,4H),1.63(d,J=7.0Hz,3H).
步骤12:(3-(1-乙酰基-4-氟哌啶-4-基)-5-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((S)-1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮(化合物59)
将3-(1-乙酰基-4-羟基哌啶-4-基)-5-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((S)-1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮(化合物1,150mg,0.244mmol)溶于二氯甲烷(5mL),氮气保护,0℃下加入二乙氨基三氟化硫(78mg,0.487mmol)。反应升至室温搅拌2小时。加入水(20mL)稀释,加乙酸乙酯(50mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,得到(3-(1-乙酰基-4-氟哌啶-4-基)-5-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((S)-1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮粗品,直接用于下一步反应。ESI-MS m/z:618.3[M+H]+
步骤13:(3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((S)-1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮(化合物2)
将步骤12得到的(3-(1-乙酰基-4-氟哌啶-4-基)-5-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((S)-1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮粗品溶于甲醇(15mL), 加入30%甲醇钠的甲醇溶液(455mg,2.44mmol)。反应液40℃下搅拌24小时,减压浓缩,残留物通过制备HPLC纯化得到化合物2,白色固体(26mg,收率17%)。ESI-MS m/z:630.3[M+H]+1H NMR(400MHz,CDCl3)δ7.70(s,1H),7.51(q,J=6.4Hz,1H),7.45(t,J=7.2Hz,1H),7.16(t,J=7.6Hz,1H),6.91(t,J=55.1Hz,1H),5.59(p,J=7.2Hz,1H),5.20(d,J=7.2Hz,1H),4.58(d,J=12.0Hz,1H),3.87(s,3H),3.76-3.63(m,2H),3.56-3.40(m,2H),3.27(s,3H),3.12(t,J=7.2Hz,1H),2.93(q,J=10.0Hz,1H),2.72-2.49(m,4H),2.47(d,J=2.0Hz,3H),2.37(d,J=2.0Hz,3H),2.30(q,J=9.2Hz,1H),2.13(s,3H),2.10-2.00(m,2H),1.95-1.85(m,1H),1.84-1.67(m,3H),1.64(d,J=7.0Hz,3H).13C NMR(101MHz,CDCl3)δ168.91,162.27,152.21,149.77,149.75,140.14,133.22,132.87,132.75,131.01,129.76,129.29,125.20,124.21,113.16,110.87,110.80,101.91,101.89,64.83,57.64,50.31,46.58,46.49,42.32,41.77,37.31,33.91,32.87,32.18,31.70,30.98,29.14,22.66,21.89,21.46,20.10.
实施例2
3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((R)-1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮(化合物3)
化合物2参考实施例1中步骤2~步骤13的方法制备,其中将步骤2中的原料(S)-(-)-1-叔丁氧羰基-2-吡咯烷甲醇替换为(R)-(-)-1-叔丁氧羰基-2-吡咯烷甲醇。ESI-MS m/z:630.3[M+H]+
实施例3
(R)-3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(2-(二甲氨基)乙氧基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物4)
步骤1:叔丁基(2-((4-溴-6-氯-2-甲基吡啶-3-基)氧基)乙基)(甲基)氨基甲酸酯
将4-溴-6-氯-2-甲基吡啶-3-醇(21g,95mmol)和叔丁基(2-羟乙基)(甲基)氨基甲酸酯(18.2g,104.5mmol)溶于四氢呋喃(400mL),氮气保护0℃下加入三苯基膦(30g,114mmol)和偶氮二甲酸二异丙酯(23g,114mmol)。反应升至室温搅拌1小时。加入水(400mL)稀释,加乙酸乙酯(800mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(石油醚/乙酸乙酯=15:1,体积比)得到产物,无色油状物(32g,收率89%)。ESI-MS m/z:379.1[M+1]+1H NMR(400MHz,CDCl3)δ7.38(s,1H),4.01(d,J=16.7Hz,2H),3.62(s,2H),3.03(s,3H),2.49(s,3H),1.45(s,9H).
步骤2:叔丁基(2-((6-氯-2-甲基-4-(甲氨基吡啶-3-基)氧基)乙基)(甲基)氨基甲酸酯
将叔丁基(2-((4-溴-6-氯-2-甲基吡啶-3-基)氧基)乙基)(甲基)氨基甲酸酯(32g,84.6mmol)溶于二甲亚砜(80mL),加入甲胺水溶液(80mL,23%),120℃封管搅拌16小时。冷却至室温后,加入水(300mL)稀释,加乙酸乙酯(600mL)萃取。有机相用无水硫酸钠干燥,减压浓缩得到产物,无色油状物(28.3g,粗品)。ESI-MS m/z:330.1[M+1]+1H NMR(400MHz,CDCl3)δ6.32(s,1H),3.87(s,2H),3.55(t,J=4.9Hz,2H),2.98(s,3H),2.82(d,J=5.1Hz,3H),2.33(s,3H),1.45(s,9H).
步骤3:叔丁基(2-((6-氯-5-碘-基-4-(甲氨基吡啶-3-基)氧基)乙基)(甲基)氨基甲酸酯
将叔丁基(2-((6-氯-2-甲基-4-(甲氨基吡啶-3-基)氧基)乙基)(甲基)氨基甲酸酯(28.3g,84.6mmol)溶于乙酸(300mL),加入N-碘代丁二酰亚胺(28.5g,126.9mmol),室温搅拌20小时。加入饱和亚硫酸钠水溶液(200mL)稀释,加入饱和碳酸氢钠水溶液(200mL)中和,加乙酸乙酯(300mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(石油醚/乙酸乙酯=6:1,体积比)得到产物,黄色油状物(37.3g,收率96%)。ESI-MS m/z:456.1[M+1]+1H NMR(400MHz,CDCl3)δ4.62(s,1H),3.88–3.75(m,2H),3.57(s,2H),3.16(s,3H),2.99(s,3H),2.35(s,3H),1.45(s,9H).
步骤4:3-(5-(2-((叔丁氧基羰基)(甲基)氨基)乙氧基)-2-氯-6-甲基-4-(甲氨基)吡啶-3-基)丙烯酸乙酯
将叔丁基(2-((6-氯-5-碘-基-4-(甲氨基吡啶-3-基)氧基)乙基)(甲基)氨基甲酸酯(28.1g,61.7mmol),醋酸钯(1.38mg,6.17mmol),三(邻甲基苯基)磷(3.74mg,12.3mmol),三乙胺(18.7g,185.1mmol)和丙烯酸乙酯(30.8g,308.8mmol),溶于N,N-二甲基甲酰胺(300mL)。氮气保护,100℃搅拌反应16小时。冷却至室温后,加入水(900mL)稀释,加乙酸乙酯(900mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(石油醚/乙酸乙酯=5:1,体积比)得到产物,黄色油状物(24g,收率91%)。ESI-MS m/z:428.1[M+1]+1H NMR(400MHz,CDCl3)δ7.74(d,J=16.2Hz,1H),6.12(d,J=16.3Hz,1H),4.26(q,J=7.1Hz,2H),3.89(s,2H),3.58(t,J=5.0Hz,2H),3.00(s,3H),2.89(d,J=5.3Hz,3H),2.37(s,3H),1.46(s,9H),1.33(t,J=7.1Hz,3H).
步骤5:叔丁基(2-((5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)乙基(甲基)氨基甲酸酯
将3-(5-(2-((叔丁氧基羰基)(甲基)氨基)乙氧基)-2-氯-6-甲基-4-(甲氨基)吡啶-3-基)丙烯酸乙酯(24g,56.2mmol)溶于乙醇(250mL),加入甲硫醇钠(4.73g,67.4mmol),室温搅拌1小时。旋干溶剂,加入水(500mL)稀释,加乙酸乙酯(600mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,得到产物,黄色油状物(14.3g,收率67%)。ESI-MS m/z:382.1[[M+1]+1H NMR(400MHz,CDCl3)δ8.03(d,J=9.8Hz,1H),6.74(d,J=9.8Hz,1H),3.93(s,3H),3.85(d,J=18.7Hz,2H),3.64(d,J=5.4Hz,2H),3.03(s,3H),2.56(s,3H),1.47(d,J=3.7Hz,9H).
步骤6:叔丁基(2-((3-溴-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)乙基(甲基)氨基甲酸酯
将叔丁基(2-((5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)乙基(甲基)氨基甲酸酯(6.3g,16.5mmol)溶于乙酸(120mL),加入二溴海因(14.2g,49.6mmol),反应75℃下搅拌5小时。加入饱和亚硫酸钠水溶液(200mL)稀释,加乙酸乙酯(400mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(石油醚/乙酸乙酯=4:1,体积比)得到产物,黄色固体(6.5mg,收率85%)。ESI-MS m/z:460.1[M+1]+1H NMR(400MHz,CDCl3)δ8.46(s,1H),3.99(s,3H),3.84(d,J=21.1Hz,2H),3.64(d,J=5.5Hz,2H),3.03(s,3H),2.55(s,3H),1.47(s,9H).
步骤7:叔丁基(2-((3-(1-乙酰基-4-羟基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)乙基(甲基)氨基甲酸酯
将叔丁基(2-((3-溴-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)乙基(甲基)氨基甲酸酯(600mg,1.3mmol)和1-乙酰哌啶-4-酮(737mg,5.22mmol)溶于四氢呋喃(15mL),氮气保护,于-40℃下滴加碘化钐(52mL,52mmol,0.1M in THF)。反应-40℃下搅拌0.5小时。-40℃下加入饱和氯化铵水溶液(100mL)稀释,加乙酸乙酯(300mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(石油醚/乙酸乙酯=1:15,体积比)得到产物,黄色油状物(500mg,收率73.7%)。ESI-MS m/z:523.1[M+1]+1H NMR(400MHz,CDCl3)δ7.93(s,1H),5.27(s,1H),4.62(d,J=10.4Hz,1H),3.97(s,3H),3.92–3.82(m,2H),3.76(dd,J=14.3,7.8Hz,2H),3.65(t,J=5.4Hz,2H),3.19(d,J=18.2Hz,1H),3.04(s,3H),2.58(s,3H),2.14(d,J=9.9Hz,5H),1.87(td,J=12.9,5.1Hz,2H),1.48(s,9H).
步骤8:叔丁基(R)-(2-((3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)乙基(甲基)氨基甲酸酯
将叔丁基(2-((3-(1-乙酰基-4-羟基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)乙基(甲基)氨基甲酸酯(500mg,0.957mmol),甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(96mg,0.096mmol),(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺(271mg,1.436mmol)和叔丁醇钠(275mg,2.87mmol)溶于甲苯中(10mL),氮气保护,90℃搅拌反应3小时。冷却至室温后,减压浓缩,残留物硅胶柱层析(二氯甲烷/甲醇=25:1,体积比)得到产物,黄色油状物(400mg,收率62%)。ESI-MS m/z:676.1[M+1]+1H NMR(400MHz,CDCl3)δ7.71(s,1H),7.55–7.40(m,2H),7.16(t,J=7.6Hz,1H),6.93(t,J=55.1Hz,1H),5.89(s,1H),5.62(dd,J=17.5,11.8Hz,2H),4.61(d,J=11.1Hz,1H),3.91(s,3H),3.71(d,J=8.9Hz,4H),3.56(t,J=5.2Hz,2H),3.24–3.08(m,1H),3.00(s,3H),2.32(d,J=0.9Hz,3H),2.23(d,J=13.3Hz,1H),2.13(d,J=5.4Hz,3H),2.05(d,J=12.9Hz,1H),1.98–1.78(m,2H),1.62(d,J=6.7Hz,3H),1.46(d,J=8.7Hz,9H).
步骤9:(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(2-(甲氨基)乙氧基)-1,6-萘啶-2(1H)-酮
将叔丁基(R)-(2-((3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)乙基(甲基)氨基甲酸酯(180mg,0.267mmol)溶于二氯甲烷(3mL),加入三氟乙酸(1mL),室温搅拌0.5小时。减压浓缩,残留物直接用于下一步反应。ESI-MS m/z:576.3[M+1]+
步骤10:(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(2-(二甲氨基)乙氧基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
将步骤9得到的残留物溶于四氢呋喃(5mL),加入甲醛水溶液(1.5mL,30%)和三乙酰氧基硼氢化钠(566mg,2.67mmol),室温下搅拌0.5小时。加入饱和碳酸氢钠水溶液(10mL)稀释,加乙酸乙酯(50mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(二氯甲烷/甲醇=13:1,体积比)得到产物,黄色固体(130mg)。ESI-MS m/z:590.1[M+1]+1H NMR(400MHz,CDCl3)δ7.69(d,J=3.7Hz,1H),7.58–7.41(m,2H),7.16(dd,J=7.6,6.3Hz,1H),6.93(t,J=55.1Hz,1H),5.88(s,1H),5.61(d,J=7.3Hz,2H),4.61(d,J=12.3Hz,1H),3.92(s,3H),3.79–3.68(m,4H),3.17(td,J=12.7,2.5Hz,1H),2.81(s,2H),2.43(s,6H),2.37(d,J=0.7Hz,3H),2.26–2.19(m,1H),2.13(d,J=5.1Hz,3H),2.05(d,J=12.4Hz,1H),1.99–1.89(m,1H),1.87–1.75(m,1H),1.62(d,J=6.0Hz,3H).
步骤11:(R)-3-(1-乙酰基-4-氟哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(2-(二甲氨基)乙氧基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
将(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(2-(二甲氨基)乙氧基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(130mg,0.22mmol)溶于二氯甲烷(5mL),氮气保护,0℃下 加入二乙氨基三氟化硫(70mg,0.447mmol)。反应升至室温搅拌1小时。加入水(20mL)稀释,加乙酸乙酯(50mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物直接用于下一步反应。ESI-MS m/z:592.3[M+1]+
步骤12:(R)-3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(2-(二甲氨基)乙氧基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
将步骤11得到的残留物溶于甲醇(15mL),加入甲醇钠的甲醇溶液(455mg,2.44mmol,30%w.t.in MeOH)。反应液40℃下搅拌24小时,减压浓缩,残留物制备HPLC纯化得到化合物4,白色固体(25mg,收率19%)。ESI-MS m/z:604.1[M+1]+1H NMR(400MHz,CDCl3)δ7.69(s,1H),7.55–7.42(m,2H),7.17(t,J=7.7Hz,1H),6.91(t,J=55.1Hz,1H),5.68–5.51(m,1H),5.20(d,J=6.8Hz,1H),4.58(d,J=11.0Hz,1H),4.15(s,2H),3.85(s,3H),3.73(dd,J=18.4,12.4Hz,3H),3.54–3.40(m,1H),3.27(s,3H),2.99–2.86(m,1H),2.82(t,J=6.0Hz,2H),2.42(s,6H),2.37(d,J=1.6Hz,3H),2.13(s,3H),2.04(d,J=14.1Hz,1H),1.90(dd,J=18.5,6.9Hz,1H),1.64(d,J=7.0Hz,3H).
实施例4
(R)-3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(2-(甲氨基)乙氧基)-1,6-萘啶-2(1H)-酮(化合物5)
由中间体(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(2-(甲氨基)乙氧基)-1,6-萘啶-2(1H)-酮(实施例3步骤9制备得到)出发,参照实施例3的步骤11和步骤12制备得到化合物5。ESI-MS m/z:590.3[M+1]+1H NMR(400MHz,CDCl3)δ8.38(s,1H),7.69(s,1H),7.46(dd,J=18.7,11.7Hz,2H),7.17(s,1H),6.90(t,J=55.1Hz,1H),5.66–5.54(m,1H),5.25(s,1H),4.57(d,J=11.5Hz,1H),3.83(s,5H),3.70(d,J=13.8Hz,1H),3.48(dd,J=23.0,10.3Hz,1H),3.27(s,3H),3.16(d,J=4.8Hz,2H),2.99–2.86(m,1H),2.64(d,J=13.0Hz,3H),2.51(ddd,J=31.6,13.8,4.5Hz,2H),2.37(d,J=1.3Hz,3H),2.13(s,3H),2.08–1.98(m,1H),1.89(t,J=11.2Hz,1H),1.64(d,J=7.0Hz,3H).
实施例5
(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-甲腈(化合物6)
步骤1:3-(苄氧基)-4-溴-6-氯-2-甲基吡啶
将4-溴-6-氯-2-甲基吡啶-3-醇(14.0g,62.9mmol)溶于N,N-二甲基甲酰胺(50mL),加入溴化苄(14.0g,81.8mmol),反应室温下搅拌16小时。加入水(300mL)稀释,加甲基叔丁基醚(250mL x 2)萃取。有机相用饱和氯化钠水溶液(300mL)洗涤,无水硫酸钠干燥,减压浓缩,残留物用乙醇打浆处理,得到产物,白色固体(17.5g,收率89%)。ESI-MS m/z:312.0[M+H]+
步骤2:3-(苄氧基)-6-氯-N,2-二甲基吡啶-4-胺
将3-(苄氧基)-4-溴-6-氯-2-甲基吡啶(17.5g,56.0mmol)溶于二甲基亚砜(150mL),加入25%甲胺水溶液(80mL),于105℃下搅拌反应5小时。冷却至室温后,加入饱和氯化铵水溶液(250mL) 稀释,加乙酸乙酯(300mL x 2)萃取。有机相饱和氯化钠水溶液(300mL)洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(石油醚/乙酸乙酯=6:1,体积比)得到产物,白色固体(8.0g,收率54%)。ESI-MS m/z:263.1[M+H]+
步骤3:3-(苄氧基)-6-氯-5-碘-N,2-二甲基吡啶-4-胺
将3-(苄氧基)-6-氯-N,2-二甲基吡啶-4-胺(8.0g,30.5mmol)溶于乙酸(40mL),加入N-碘代丁二酰亚胺(8.2g,36.6mmol),室温搅拌6小时。加入饱和亚硫酸钠水溶液(200mL)稀释,加入饱和碳酸氢钠水溶液(400mL)中和,加乙酸乙酯(200mL x 2)萃取。有机相饱和氯化钠水溶液(300mL)洗涤,无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(石油醚/乙酸乙酯=10:1,体积比)得到产物,白色固体(8.3g,收率73%)。ESI-MS m/z:389.0[M+H]+
步骤4:3-(5-(苄氧基)-2-氯-6-甲基-4-(甲氨基)吡啶-3-基)丙烯酸乙酯
将3-(苄氧基)-6-氯-5-碘-N,2-二甲基吡啶-4-胺(8.3g,21.4mmol),醋酸钯(480mg,2.14mmol),三(邻甲基苯基)磷(1.3g,4.27mmol),三乙胺(4.3g,42.7mmol)和丙烯酸乙酯(6.4g,64.1mmol),溶于N,N-二甲基甲酰胺(120mL)。氮气保护,95℃搅拌反应6小时。冷却至室温后,加入水(500mL)稀释,加乙酸乙酯(300mL x 2)萃取。有机相饱和氯化钠水溶液(300mL)洗涤,无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(石油醚/乙酸乙酯=6:1,体积比)得到产物,白色固体(5.8g,收率75%)。ESI-MS m/z:361.1[M+H]+
步骤5:8-(苄氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮
3-(5-(苄氧基)-2-氯-6-甲基-4-(甲氨基)吡啶-3-基)丙烯酸乙酯(4g,11.1mmol)溶于无水乙醇(40mL),加入甲硫醇钠(0.77g,11.1mmol),室温下搅拌2小时,TLC显示反应完全。反应液加入水和乙酸乙酯,分出有机层,水相再用乙酸乙酯萃取,合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(石油醚/乙酸乙酯=5:1,体积比)得到产物(2.7g,收率77%)。ESI-MS m/z:315[M+H]+1H NMR(400MHz,CDCl3)δ8.05(d,J=9.8Hz,1H),7.46-7.35(m,5H),6.75(d,J=9.8Hz,1H),4.79(s,2H),3.89(s,3H),2.59(s,3H).
步骤6:8-(苄氧基)-3-溴-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮
将8-(苄氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮(8.0g,25.4mmol)溶于N,N-二甲基甲酰胺(120mL),加入二溴海因(36.3g,127mmol),反应70℃下避光搅拌5小时。加入饱和亚硫酸钠水溶液(200mL)稀释,加乙酸乙酯(400mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(石油醚/乙酸乙酯=5:1,体积比)得到产物,淡黄色固体(7g,收率70%)。ESI-MS m/z:393.1[M+1]+1H NMR(400MHz,CDCl3)δ8.48(s,1H),7.47-7.29(m,5H),4.79(s,2H),3.96(s,3H),2.58(s,3H).
步骤7:3-(1-乙酰基-4-羟基哌啶-4-基)-8-(苄氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮
向2L三口烧瓶中加入8-(苄氧基)-3-bromo-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮(7.5g,19.03mmol)、四氢呋喃(200mL)以及1-乙酰-4-哌啶酮(10.7g,76.10mmol)使其完全溶溶解,氮气保护,反应液冷却至-40℃,滴加0.1mol/L二碘化钐四氢呋喃溶液(500mL),加入完毕后继续在-40℃反应0.5小时。加入饱和氯化铵(300mL)淬灭反应,再用乙酸乙酯(400mL)萃取,有机相用饱和食盐水(300mL)洗涤,无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(石油醚/乙酸乙酯=3:1,体积比),得灰白色固体3-(1-乙酰基-4-羟基哌啶-4-基)-8-(苄氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮(6.6g,收率76%)。ESI-MS m/z:456.3[M+H]+1H NMR(400MHz,CDCl3)δ7.94(s,1H),7.45–7.36(m,5H),5.35-5.2(m,1H),4.80(s,2H),4.68–4.52(m,1H),3.92(s,3H),3.80=-3.60(m,1H),3.33–3.10(m,1H),2.61(s,3H),2.18(s,1H),2.16(s,3H),1.92-1.81(m,2H),1.29–1.23(m,2H).
步骤8:(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-8-(苄氧基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
向250mL圆底烧瓶中加入3-(1-乙酰基-4-羟基哌啶-4-基)-8-(苄氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮(9.6g,21.03mmol)、甲苯(100mL),叔丁醇钠(6.0g,63.10mmol),甲烷磺酸(2-二环己 基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(Brettphos Pd G3,1.9g,2.10mmol)以及(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺(6.0g,31.64mmol),氮气保护,在80℃下搅拌反应1小时。反应液冷却至室温,加入水和乙酸乙酯萃取分层,有机相依次用10%柠檬酸溶液(300mL)、饱和食盐水(100mL)各洗涤一次,无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(石油醚/乙酸乙酯=3:1,体积比),得淡黄色固体(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-8-(苄氧基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(8.8g,收率70%)。ESI-MS m/z:609.2[M+H]+1H NMR(400MHz,CDCl3)δ7.64-7.55(m,2H),7.50-7.44(m,1H),7.40-7.35(m,4H),7.23-7.16(m,1H),7.16–7.08(m,1H),7.07–6.95(m,1H),6.86(dd,J=55.0,3.4Hz,1H),5.82-5.60(m,J=50.7Hz,2H),4.73-4.53(m,3H),3.88(s,3H),3.73-3.64(m,2H),3.16(t,J=12.7Hz,1H),2.42(s,2H),2.33–2.25(m,3H),2.13(d,J=7.9Hz,3H),2.06-1.98(m,1H),1.75-1.60(m,4H).
步骤9:(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮
向250mL圆底烧瓶中加入(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-8-(苄氧基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(2.0g,3.23mmol)、甲醇(50mL),10%钯炭(1.5g),氢气氛围下,室温搅拌反应1小时。反应液过滤,减压浓缩,残留物硅胶柱层析(石油醚/乙酸乙酯=3:1,体积比),得淡黄色固体(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮(1.4g,收率80%)。ESI-MS m/z:519.3[M+H]+1H NMR(400MHz,CDCl3)δ7.66(s,1H),7.52(q,J=7.8Hz,1H),7.43(t,J=7.2Hz,1H),7.17-7.11(m,1H),6.91(t,J=55.1Hz,1H),5.93(s,1H),5.63-5.51(m,1H),5.49-5.3(m,1H),4.51(d,J=13.4Hz,1H),3.95(s,3H),3.71–3.59(m,2H),3.10(td,J=12.8,2.9Hz,1H),2.35(s,2H),2.23–2.12(m,1H),2.08(d,J=2.2Hz,3H),1.99(d,J=12.7Hz,2H),1.90–1.75(m,3H),1.61(d,J=6.9Hz,3H).
步骤10:(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯
向(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮(300mg,0.58mmol)、N,N-二异丙基乙胺(150mg,1.16mmol)和4-二甲氨基吡啶(71mg,0.58mmol)的二氯甲烷(15mL)溶液中加入N-苯基双(三氟甲烷磺酰)亚胺(270mg,0.75mmol),室温搅拌反应1.5小时,加入二氯甲烷和水萃取分层,有机相用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,残留物经硅胶柱层析(甲醇/二氯甲烷=20:1,体积比)纯化得到产物,黄色固体产物(340mg,收率90%)。ESI-MS m/z:651[M+H]+
步骤11:(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-甲腈
将(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(38mg,0.058mmol)溶于干燥N,N-二甲基甲酰胺中(5mL),加入四(三苯基膦)钯(14mg,0.012mmol),氰化锌(68mg,0.58mmol),氮气保护,反应液于110℃下搅拌1小时,冷却至室温,硅藻土抽滤,乙酸乙酯(20ml)洗涤,再加乙酸乙酯(100mL)萃取,加水洗涤(20ml),饱和食盐水(20ml)洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=20:1,体积比)得到粗品,将粗品再通过制备HPLC纯化得到化合物6,白色固体(4mg,收率13%)。ESI-MS m/z:528.2[M+H]+1H NMR(400MHz,CDCl3)δ8.08(d,J=8.6Hz,1H),7.57–7.45(m,2H),7.36–7.28(m,1H),7.20(t,J=7.6Hz,1H),6.93(t,J=55.0Hz,1H),6.00–5.73(m,2H),4.63–4.50(m,1H),4.07(s,3H),3.78–3.64(m,2H),3.23–3.10(m,1H),2.62(s,3H),2.24–2.16(m,1H),2.10(d,J=33.5Hz,3H),2.04–1.80(m,3H),1.67–1.62(m,3H).
实施例6
(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7,8-三甲基-1,6-萘啶-2(1H)-酮(化合物7)
将(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(66mg,0.1mmol)溶于1,4-二氧六环中(5mL),加入四(三苯基膦)钯(11mg,0.01mmol),磷酸钾(106mg,0.5mmol),氮气保护,再加入三甲基环三硼氧烷(0.14ml,1mmol),反应液于100℃下搅拌2小时,冷却至室温,硅藻土抽滤,乙酸乙酯(20ml)洗涤,再加乙酸乙酯(100mL)萃取,加水洗涤(20ml),饱和食盐水(20ml)洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=20:1)得到粗品,将粗品再通过制备HPLC纯化得到化合物7,白色固体(2.5mg,收率4.8%)。ESI-MS m/z:517.2[M+H]+1H NMR(400MHz,CDCl3)δ7.59(d,J=3.8Hz,1H),7.58–7.50(m,1H),7.47(t,J=7.2Hz,1H),7.22–7.15(m,1H),6.95(t,J=55.1Hz,1H),5.91(s,1H),5.69–5.60(m,1H),5.34(s,1H),4.68–4.60(m,1H),3.76–3.72(m,2H),3.69(s,3H),3.25–3.15(m,1H),2.38(s,3H),2.33(s,3H),2.29–2.22(m,1H),2.16(d,J=3.4Hz,3H),2.12–2.06(m,1H),2.00–1.91(m,1H),1.82–1.76(m,1H),1.66(d,J=7.0Hz,3H).
实施例7
(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-乙炔基-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物8)
步骤1:(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((三异丙基硅基)乙炔基)-1,6-萘啶-2(1H)-酮
向(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(40mg,0.06mmol)和三异丙基硅基乙炔(110mg,0.6mmol)的四氢呋喃(5mL)溶液中依次加入四-三苯基膦钯(10mg,0.009mmol),碘化亚铜(5mg,0.03mmol)三乙胺(1mL),氩气置换3次后,在60℃搅拌反应4小时后,过滤,滤液减压浓缩得粗品,粗品经硅胶柱层析(甲醇/二氯甲烷=20:1,体积比)得到黄色固体产物(30mg)。ESI-MS m/z:683.4[M+H]+
步骤2:(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-乙炔基-1,7-二甲基-1,6-萘啶-2(1H)-酮
向(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((三异丙基硅基)乙炔基)-1,6-萘啶-2(1H)-酮(30mg,0.04mmol)的四氢呋喃(15mL)溶液中加入四丁基氟化铵的四氢呋喃溶液(0.15mL,1M),在室温下搅拌16小时后,加乙酸乙酯(60mL)和水(50mL),分出有机相,水洗两次(30mL×2),减压浓缩得到粗品产物,粗品通过制备HPLC纯化得到化合物8,白色固体(15mg)。ESI-MS m/z:527.2[M+1]+1H NMR(400MHz,DMSO-d6)δ8.49–8.41(m,1H),8.07(d,J=7.1Hz,1H),7.66(t,J=7.4Hz,1H),7.46(t,J=7.1Hz,1H),7.37–7.08(m,3H),5.73–5.64(m,1H),5.49(s,1H),4.59(s,1H),4.39–4.30(m,1H),3.90(s,3H),3.71(d,J=11.8Hz,1H),3.50–3.44(m,1H),2.92(t,J=12.8Hz,1H),2.38(s,3H),2.35-2.29(m,1H),2.03(s,3H),1.57(d,J=7.1Hz,3H),1.50(d,J=13.1Hz,1H),1.26–1.20(m,1H).
实施例8
(R)-3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-甲腈(化合物9)
步骤1:(R)-3-(1-乙酰基-4-氟哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-甲腈
将化合物6(70mg,0.13mmol)溶于干燥二氯甲烷中(8mL),氮气保护,冷却至0℃,加入二乙胺基三氟化硫(42mg,0.26mmol),反应液于室温下搅拌2小时,加水(20ml)淬灭反应,再加乙酸乙酯(100mL x 2)萃取,饱和食盐水(20ml)洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=20:1,体积比)得到产物,黄色固体(47mg,收率68%)。ESI-MS m/z:530.2[M+H]+。
步骤2:(R)-3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-甲腈
将(R)-3-(1-乙酰基-4-氟哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-甲腈(47mg,0.089mmol)溶于无水甲醇中(5mL),氮气保护,加入甲醇钠的甲醇溶液(5N 0.36ml,1.78mmol),反应液于室温下搅拌12小时,加水(20ml)淬灭反应,再加乙酸乙酯(100mL x 2)萃取,饱和食盐水(20ml)洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=20:1)得到粗品,将粗品通过制备HPLC纯化得到化合物9,白色固体(13mg,收率27%)。ESI-MS m/z:542.2[M+H]+。1H NMR(400MHz,CDCl3)δ7.71(s,1H),7.49(t,J=7.2Hz,2H),7.20(t,J=7.7Hz,1H),6.89(t,J=55.0Hz,1H),6.05(d,J=5.7Hz,1H),5.77–5.68(m,1H),4.58(d,J=12.1Hz,1H),3.99(s,3H),3.75–3.66(m,1H),3.54–3.42(m,1H),3.25(s,3H),2.98–2.86(m,1H),2.61(s,3H),2.58–2.36(m,2H),2.12(s,3H),2.09–2.01(m,1H),1.93–1.83(m,1H),1.68(d,J=7.2Hz,3H).
实施例9
(R)-3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-乙炔基-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物10)
由化合物8出发,参照实施例8的步骤1和步骤2制备得到化合物10。ESI-MS m/z:541.2[M+1]+
实施例10
(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-8-(3-氨基-3-甲基丁-1-炔-1-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物11)
步骤1:叔丁基(R)-(4-(3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)-2-甲基丁-3-炔-2-基)氨基甲酸酯
将(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(250mg,0.38mmol)溶于干燥四氢呋喃中(10mL),加入四(三苯基膦)钯(87mg,0.076mmol),(2-甲基丁-3-炔-2-基)氨基甲酸叔丁酯(695mg,3.8mmol),碘化亚铜(50mg,0.27mmol),三乙胺(2mL),氮气保护,反应液于60℃下搅拌3小时,冷却至室温,硅藻土抽滤,乙酸乙酯(20ml)洗涤,再加乙酸乙酯(100mL x 2)萃取,加水洗涤(20ml),饱和食盐水(20ml)洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=20:1,体积比)得到产物,淡黄色固体(59mg,收率22%)。ESI-MS m/z:684.3[M+H]+
步骤2:(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-8-(3-氨基-3-甲基丁-1-炔-1-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
将叔丁基(R)-(4-(3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)-2-甲基丁-3-炔-2-基)氨基甲酸酯(59mg,0.086mmol)溶于二氯甲烷中(5mL),加入三氟乙酸(2mL),反应液于室温下搅拌2小时,加二氯甲烷(20ml)稀释,加水洗涤(20ml),饱和食盐水(20ml)洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=20:1,体积比)得到化合物11,淡白色固体(20mg,收率40%)。ESI-MS m/z:584.3[M+H]+
实施例11
(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-羟基-3-甲基丁-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物12)
由中间体(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(实施例5步骤10制备得到)和原料2-甲基-3-丁炔-2-醇出发,参照实施例10的步骤1制备得到化合物12。ESI-MS m/z:585.3[M+1]+1H NMR(400MHz,DMSO-d6)δ8.42(d,J=1.8Hz,1H),8.00(d,J=7.1Hz,1H),7.66(t,J=7.5Hz,1H),7.46(t,J=7.0Hz,1H),7.37–7.07(m,3H),5.75–5.64(m,1H),5.51(s,1H),5.37(s,1H),4.39–4.29(m,1H),3.89(s,3H),3.74–3.67(m,1H),3.50–3.42(m,1H),2.93(t,J=12.7Hz,1H),2.36(s,3H),2.03(s,3H),1.63–1.58(m, 1H),1.56(d,J=7.1Hz,3H),1.53–1.48(m,1H),1.44(s,6H),1.06(t,J=7.2Hz,1H).
实施例12
3-(1-乙酰基-4-羟基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((S)-1-甲基吡咯烷-2-基)乙炔基)-1,6-萘啶-2(1H)-酮(化合物13)
步骤1:叔丁基(S)-2-((3-(1-乙酰基-4-羟基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)乙炔基)吡咯烷-1-羧酸酯
将(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(150mg,0.23mmol)和叔丁基(S)-2-乙炔基吡咯烷-1-羧酸酯(180mg,13.5mmol)、碘化亚铜(13mg,0.07mmol)、四三苯基膦钯(27mg,0.02mmol)、和三乙胺(3.5mL)溶于四氢呋喃(15mL)。氮气置换后,加热60℃搅拌反应10小时。通过硅藻土滤除固体,滤液减压浓缩,残留物通过硅胶柱层析纯化(二氯甲烷/甲醇=100:3,体积比)得到产物。黄色油状物(100mg,63%)。ESI-MS m/z:696.3[M+1]+
步骤2:3-(1-乙酰基-4-羟基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((S)-吡咯烷-2-基乙炔基)-1,6-萘啶-2(1H)-酮
将叔丁基(S)-2-((3-(1-乙酰基-4-羟基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)乙炔基)吡咯烷-1-羧酸酯(100mg,0.1mmol)溶于二氯甲烷(3mL),加入三氟乙酸(0.5mL),室温下反应1小时。减压浓缩,残留物通过硅胶柱层析纯化(二氯甲烷/甲醇=100:6,体积比)得到产物,黄色油状物(77mg),直接用于下一步反应。ESI-MS m/z:596.2[M+1]+
步骤3:3-(1-乙酰基-4-羟基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((S)-1-甲基吡咯烷-2-基)乙炔基)-1,6-萘啶-2(1H)-酮
将步骤2得到产物粗品溶于四氢呋喃(10mL),加入30%甲醛水溶液(3mL),加入三乙酰氧基硼氢化钠(360mg,1mmol),室温下搅拌0.5小时。加入饱和碳酸氢钠水溶液(30mL)稀释,加乙酸乙酯(100mL x 3)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物通过制备HPLC纯化得到化合物13,黄色固体产物(5mg)。ESI-MS m/z:610.3[M+H]+
实施例13
3-(1-乙酰基-4-羟基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((R)-1-甲基吡咯烷-2-基)乙炔基)-1,6-萘啶-2(1H)-酮(化合物14)
由中间体(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(实施例5步骤10制备得到)和原料叔丁基(R)-2-乙炔基吡咯烷-1-羧酸酯出发,参照实施例12制备得到化合物14。ESI-MS m/z:610.3[M+1]+。
实施例14
(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物15)
由中间体(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(实施例5步骤10制备得到)和原料N,N-二甲基炔丙胺出发,参照实施例10的步骤1制备得到化合物15。ESI-MS m/z:584.3[M+1]+
实施例15
(R)-3-(1-((3-(1-乙酰基-4-羟基哌啶-4-基)-8-(3-氨基-3-甲基丁-1-炔-1-基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-5-基)氨基)乙基)-2-甲基苯甲腈(化合物16)
步骤1:3-(1-乙酰基-4-羟基哌啶-4-基)-5-氯-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-羟基哌啶-4-基)-5-氯-1,7-二甲基-8-(苄氧基)-1,6-萘啶-2(1H)-酮(2.4g,5.27mmol)溶于二氯甲烷(24mL),氮气保护,0℃下加入三氯化硼(53mL,52.7mmol)。反应升至室温搅拌1小时。将反应液滴加至冷的甲醇(200mL)中,减压浓缩,残留物溶于甲醇(50mL)中,加入氨的饱和甲醇溶液(3mL),减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=20:1,体积比)得到产物,白色泡沫状固体(1g,收率52%)。ESI-MS m/z:366.1[M+H]+
步骤2:3-(1-乙酰基-4-羟基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯
将3-(1-乙酰基-4-羟基哌啶-4-基)-5-氯-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮(1g,2.7mmol)溶于二氯甲烷(20mL),氮气保护,0℃下加入三乙胺(1.1g,10.8mmol),4-二甲氨基吡啶(330mg,2.7mmol)和苯基双(三氟甲烷磺酰)亚胺(1.17g,3.28mmol)。0℃下反应搅拌0.5小时。减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=99:1,体积比)得到产物(1g,收率72%)。ESI-MS m/z:498.1[M+H]+1H NMR(400MHz,CDCl3)δ7.96(s,1H),4.62(m,1H),3.81(s,3H),3.79–3.50(m,2H),3.40-3.00(m,1H),2.69(s,3H),2.16(s,3H),1.90(td,J=13.0,5.0Hz,2H).
步骤3:叔丁基(4-(3-(1-乙酰基-4-羟基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)-2-甲基丁-3-炔-2-基)氨基甲酸酯
将3-(1-乙酰基-4-羟基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(395mg,6.65mmol),叔丁基(2-甲基丁-3-炔-2-基)氨基甲酸酯(160mg,0.87mmol),碘化亚铜(45mg,0.24mmol),四三苯基膦钯(91mg,0.08mmol)和三乙胺(1mL)溶于四氢呋喃(10mL)。氮气保护,40℃搅拌反应10分钟。反应物冷却至室温后,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=20:1,体积比)得到产物(420mg,收率99%)。ESI-MS m/z:531.2[M+H]+1H NMR(400MHz,CDCl3)δ7.96(s,1H),4.58(t,J=20.8Hz,1H),4.14(s,3H),3.75–3.67(m,2H),3.16(td,J=12.8,2.3Hz,1H),2.74(s,3H),2.15(s,3H),1.95–1.75(m,4H),1.68(s,6H),1.46(s,9H).
步骤4:叔丁基(4-(3-(1-乙酰基-4-羟基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)-2-甲基丁-3-炔-2-基)氨基甲酸酯
将叔丁基(4-(3-(1-乙酰基-4-羟基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)-2-甲基丁-3-炔-2-基)氨基甲酸酯(360mg,0.68mmol),(R)-3-(1-氨基乙基)-2-甲基苯甲腈(326mg,2mmol),甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(Brettphos Pd G3,61mg,0.067mmol)和叔丁醇钠(136mg,1.4mmol)溶于甲苯(10mL)。氮气保护,80℃搅拌反应50分钟。反应物冷却至室温后,加入水(50mL)稀释,加乙酸乙酯(50mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,得到产物(300mg,收率67%)。ESI-MS m/z:654.4[M+H]+
步骤5:(R)-3-(1-((3-(1-乙酰基-4-羟基哌啶-4-基)-8-(3-氨基-3-甲基丁-1-炔-1-基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-5-基)氨基)乙基)-2-甲基苯甲腈
将叔丁基(R)-(4-(3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-氰基-2-甲基苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)-2-甲基丁-3-炔-2-基)氨基甲酸酯(25mg,0.038mmol)溶于二氯甲烷(3mL),加入三氟乙酸(1mL),室温搅拌1小时。减压浓缩,残留物通过制备HPLC纯化得到化合物16,白色固体(11mg,收率52%)。ESI-MS m/z:555.3[M+H]+1H NMR(400MHz,CD3OD)δ8.24(d,J=27.6Hz,1H),7.69(t,J=7.1Hz,1H),7.49(t,J=14.6Hz,1H),7.31–7.21(m,1H),4.00(s,3H),3.85(d,J=11.8Hz,1H),3.70-3.60(m,1H),3.2-3.08(m,2H),2.74(s,3H),2.47(s,3H),2.42-2.32(m,3H),2.20(s,3H),1.92–1.88(m,1H),1.86(s,3H),1.85-1.75(m,1H),1.72(s,6H),1.57(d,J=7.0Hz,3H).
实施例16
(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-8-((1-氨基环丙基)乙炔基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物17)
化合物17参照实施例15的步骤3~步骤5制备,其中将步骤3中的原料(2-甲基丁-3-炔-2-基)氨基甲酸叔丁酯替换为(1-乙炔基环丙基)氨基甲酸叔丁酯,步骤4中的原料(R)-3-(1-氨基乙基)-2-甲基苯甲腈替换为(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺。ESI-MS m/z:582.3[M+1]+
实施例17
(R)-3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙- 1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物18)
步骤1:3-(1-乙酰基-4-甲氧基哌啶-4-基)-8-(苄氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-羟基哌啶-4-基)-8-(苄氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮(5g,10.96mmol)溶于N,N-二甲基甲酰胺(50mL),在0℃下加入钠氢(2.19g,54.8mmol),搅拌5分钟加入碘甲烷(6.2g,43.84mmol),0℃搅拌反应1小时。加水淬灭反应,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=99:1,体积比)得到产物,黄色固体(4.0g,收率78%)。ESI-MS m/z:470.1[M+1]+1H NMR(400MHz,CDCl3)δ8.13(s,1H),7.40(s,5H),4.79(s,2H),4.58(d,J=12.9Hz,1H),3.88(s,3H),3.70(dd,J=11.1,4.7Hz,1H),3.49(ddd,J=13.0,9.1,3.7Hz,1H),3.28(s,3H),2.94(dd,J=14.6,9.5Hz,1H),2.59(s,3H),2.55–2.38(m,2H),2.13(s,3H),2.09–1.93(m,2H).
步骤2:3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-氯-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-甲氧基哌啶-4-基)-8-(苄氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮(4.0g,8.51mmol),溶于甲醇(40mL),加入二氧化铂(800mg),在氢气球氛围下,室温下氢化反应30分钟。反应物通过硅藻土过滤,滤液减压浓缩,残留物通过硅胶柱层析二氯甲烷/甲醇=95:5,体积比)得到产物,白色固体(2.6g,收率81%)。ESI-MS m/z:380.1[M+1]+1H NMR(400MHz,DMSO-d6)δ9.44(s,1H),7.93(s,1H),4.30(d,J=12.7Hz,1H),3.86(s,3H),3.68(d,J=1.8Hz,1H),3.33–3.26(m,1H),3.18(s,3H),2.76(dd,J=13.3,11.3Hz,1H),2.44(s,3H),2.30(dq,J=8.3,4.7Hz,2H),2.02(s,4H),1.89– 1.76(m,2H).
步骤3:3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯
将3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-氯-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮(2.0g,5.26mmol),4-二甲氨基吡啶(641mg,5.26mmol),三乙胺(2.12mg,21mmol)溶于二氯甲烷(40mL),0℃下加入苯基双(三氟甲烷磺酰)亚胺(2.25g,6.31mmol),0℃下继续反应30分钟。反应物减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=99:1,体积比)得到产物,白色固体(2.2g,收率90%)。ESI-MS m/z:512.1[M+1]+1H NMR(400MHz,CDCl3)δ8.14(s,1H),4.59(d,J=10.9Hz,1H),3.75(s,3H),3.70(s,1H),3.51(d,J=11.0Hz,1H),3.30(s,3H),2.93(dd,J=21.1,9.3Hz,1H),2.68(s,3H),2.48(d,J=12.7Hz,2H),2.15(s,3H),2.02–1.90(m,2H).
步骤4:3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-氯-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(510mg,1.0mmol),N,N-二甲基炔丙胺(250mg,3.0mmol),碘化亚铜(57mg,0.3mmol),四三苯基磷钯(115mg,0.1mmol)溶于四氢呋喃(5mL)和三乙胺(1mL),氮气保护,55℃下反应30分钟。反应物减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=99:1,体积比)得到产物,白色固体(370mg,收率83%)。ESI-MS m/z:445.1[M+1]+1H NMR(400MHz,CDCl3)δ8.14(s,1H),4.60–4.50(m,1H),4.08(s,3H),3.72(s,2H),3.68(d,J=2.1Hz,1H),3.48(td,J=13.1,2.5Hz,1H),3.26(s,3H),2.99–2.88(m,1H),2.75(s,3H),2.46–2.36(m,9H),2.12(s,3H),2.05–1.92(m,2H).
步骤5:(R)-3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-氯-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(175mg,0.394mmol),甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(35mg,0.039mmol),(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺(223mg,1.18mmol)和叔丁醇钠(75mg,0.78mmol)溶于1,4-二氧六环(5mL),氮气保护,80℃搅拌反应1小时。冷却至室温后,加水稀释,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,残留物通过制备HPLC纯化得到化合物18,白色固体(100mg,收率42%)。ESI-MS m/z:598.1[[M+1]+1H NMR(400MHz,DMSO-d6)δ8.19(d,J=1.4Hz,1H),8.01(d,J=7.0Hz,1H),7.64(s,1H),7.45(t,J=7.0Hz,1H),7.24(dt,J=64.5,41.7Hz,2H),5.68(p,J=7.0Hz,1H),4.32(d,J=11.4Hz,1H),3.87(s,3H),3.70(d,J=13.7Hz,1H),3.48(s,2H),3.42–3.29(m,3H),3.10(d,J=2.9Hz,3H),2.82(t,J=12.4Hz,1H),2.38(s,3H),2.28–2.11(m,8H),2.11–1.95(m,4H),1.57(d,J=7.1Hz,3H).
实施例18
3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((S)-1-甲基吡咯烷-2-基)乙炔基)-1,6-萘啶-2(1H)-酮(化合物19)
由化合物13出发,参照实施例8的步骤1和步骤2制备得到化合物19。ESI-MS m/z:624.3[M+1]+1H NMR(400MHz,CD3OD)δ8.17(s,1H),7.56(t,J=7.2Hz,1H),7.42(t,J=6.8Hz,1H),7.18(t,J=7.7 Hz,1H),6.99(t,J=54.9Hz,1H),5.71(q,J=6.8Hz,1H),4.44(d,J=11.9Hz,1H),4.01(s,3H),3.81(d,J=13.4Hz,1H),3.53(dd,J=17.4,10.2Hz,2H),3.16(s,3H),3.09–2.91(m,2H),2.55–2.46(m,4H),2.45(s,3H),2.43–2.15(m,6H),2.14(s,3H),2.06–1.81(m,3H),1.63(d,J=7.1Hz,3H).
实施例19
3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((R)-1-甲基吡咯烷-2-基)乙炔基)-1,6-萘啶-2(1H)-酮(化合物20)
由化合物14出发,参照实施例8的步骤1和步骤2制备得到化合物20。ESI-MS m/z:624.3[M+1]+
实施例20
(R)-3-(1-乙酰基-4-甲氧基哌啶-4-基)-8-(3-氨基-3-甲基丁-1-炔-1-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物21)
由化合物11出发,参照实施例8的步骤1和步骤2制备得到化合物20。ESI-MS m/z:598.3[M+1]+
实施例21
(R)-3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-8-(3-氨基-3-甲基丁-1-炔-1-基)-5-((1-(3-(二氟甲基)-2-氟 苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物22)
步骤1:3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-8-(苄氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-羟基哌啶-4-基)-8-(苄氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮(1.1g,2.41mmol)溶于N,N-二甲基甲酰胺(50mL),氮气保护下在0℃下加入氢化钠(290mg,7.25mmol,60%),0℃下搅拌10分钟,加入氘代碘甲烷(1.05g,7.25mmol),缓慢升至室温,搅拌反应12小时。加入水(100mL)稀释,加乙酸乙酯(100mL)萃取,饱和氯化钠溶液洗涤有机相。有机相用无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=99:1,体积比)得到产物3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-5-氯-1,7-二甲基-8-(苄氧基)-1,6-萘啶-2(1H)-酮,黄色固体((800mg,收率70%)。ESI-MS m/z:473.1[M+1]+1H NMR(400MHz,CDCl3)δ8.13(s,1H),7.46–7.34(m,5H),4.80(s,2H),4.70-4.50(m,1H),3.88(s,3H),3.80-3.60(m,1H),3.60-3.40(m,1H),3.05-2.90(m,1H),2.59(s,3H),2.55-2.40(m,2H),2.15(s,3H),2.10-1.90(m,2H).
步骤2:3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-5-氯-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-5-氯-1,7-二甲基-8-(苄氧基)-1,6-萘啶-2(1H)-酮(800mg,1.69mmol)溶于甲醇(20mL),加入二氧化铂(160mg),室温下用氢气球氢化反应30分钟。反应物通过硅藻土过滤,滤液减压浓缩得产物,白色固体(600mg,收率93%)。直接用于下一步反应。ESI-MS m/z:383.1[M+1]+
步骤3:3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯
将3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-5-氯-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮(660mg,1.72mmol),4-二甲氨基吡啶(210mg,1.72mmol),三乙胺(516mg,5.16mmol)溶于二氯甲烷(10mL),氮气保护下0℃下加入苯基双(三氟甲烷磺酰)亚胺(740mg,2mmol),0℃反应30分钟。低温浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=99:1,体积比)得到产物3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯,白色固体(740mg,收率83%)。ESI-MS m/z:515.1[M+1]+1H NMR(400MHz,CDCl3)δ8.14(s,1H),4.80-4.40(m,1H),3.90-3.50(m,4H),3.60–3.32(m,1H),3.10-2.80(m,1H),2.67(s,3H),2.55-2.45(m,2H),2.15(s,3H),2.05-1.85(m,2H).
步骤4:叔丁基(4-(3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)-2-甲基丁-3-炔-2-基)氨基甲酸酯
将3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(350mg,0.68mmol),叔丁基(2-甲基丁-3-炔-2-基)氨基甲酸酯(125mg,0.68mmol),碘化亚铜(39mg,0.2mmol),四三苯基膦钯(78mg,0.068mmol)和三乙胺(1mL)溶于四氢呋喃(10mL)。氮气保护,50℃搅拌反应2小时。反应物冷却至室温后,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=99:1,体积比)得到产物(300mg,收率81%)。ESI-MS m/z:548.2[M+H]+1H NMR(400MHz,CDCl3)δ8.13(s,1H),4.70-4.50(m,1H),4.08(s,3H),3.79–3.61(m,1H),3.60-3.4(m,1H),3.00-2.80(m,1H),2.73(s,3H),2.50-2.30(m,2H),2.13(s,3H),2.10-1.90(m,2H),1.67(s,6H),1.44(s,9H).
步骤5:叔丁基(R)-(4-(3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)-2-甲基丁-3-炔-2-基)氨基甲酸酯
将叔丁基(4-(3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)-2-甲基丁-3-炔-2-基)氨基甲酸酯(300mg,0.55mmol),(R)-1-(3-(二氟甲基)-2-氟苯基)-乙基)1-胺(342mg,1.8mmol),甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(Brettphos Pd G3,54mg,0.059mmol)和叔丁醇钠(120mg,1.25mmol)溶于甲苯(10mL)。氮气保护,80℃搅拌反应50分钟。反应物冷却至室温后,加入水(50mL)稀释,加乙酸乙酯(50mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,得到产物粗品(400mg)。直接用于下一步反应。ESI-MS m/z:701.4[M+H]+
步骤6:(R)-3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-8-(3-氨基-3-甲基丁-1-炔-1-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
将叔丁基(R)-(4-(3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)-2-甲基丁-3-炔-2-基)氨基甲酸酯(400mg,0.57mmol)溶于二氯甲烷(3mL),加入三氟乙酸(1mL),室温搅拌1小时。减压浓缩,残留通过制备HPLC纯化得到化合物22,白色固体(30mg,收率9%)。ESI-MS m/z:601.3[M+H]+1H NMR(400MHz,CD3OD)δ8.19(s,1H),7.56(t,J=7.4Hz,1H),7.43(t,J=6.9Hz,1H),7.19(t,J=7.7Hz,1H),6.99(t,J=54.9Hz,1H),5.72(q,J=6.9Hz,1H),4.44(d,J=13.3Hz,1H),3.98(s,3H),3.81(d,J=12.1Hz,1H),3.54(td,J=13.1,2.5Hz,1H),3.03(dd,J=12.8,10.3Hz,1H),2.46(s,3H),2.42–2.16(m,4H),2.14(s,3H),1.72(s, 6H),1.64(d,J=7.1Hz,3H).
实施例22
(R)-3-(1-((3-(1-乙酰基-4-甲氧基哌啶-4-基)-8-(3-氨基-3-甲基丁-1-炔-1-基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-5-基)氨基)乙基)-2-甲基苯甲腈(化合物23)
由化合物16出发,参照实施例8的步骤1和步骤2制备得到化合物23。ESI-MS m/z:569.3[M+1]+1H NMR(400MHz,CD3OD)δ8.18(s,1H),7.70(d,J=7.9Hz,1H),7.51(d,J=7.5Hz,1H),7.27(t,J=7.8Hz,1H),5.62(q,J=7.0Hz,1H),4.44(d,J=12.8Hz,1H),3.99(s,3H),3.81(d,J=13.6Hz,1H),3.62–3.45(m,1H),3.15(s,3H),3.10-3.00(m,1H),2.73(s,3H),2.47(s,3H),2.44–2.16(m,4H),2.14(s,3H),1.72(s,6H),1.57(d,J=7.0Hz,3H).
实施例23
(R)-3-(1-乙酰基-4-甲氧基哌啶-4-基)-8-((1-氨基环丙基)乙炔基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物24)
由化合物17出发,参照实施例8的步骤1和步骤2制备得到化合物24。ESI-MS m/z:596.3[M+1]+
实施例24
3-(1-乙酰基-4-羟基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((Z)-2- ((S)-1-甲基吡咯烷-2-基)乙烯基)-1,6-萘啶-2(1H)-酮(化合物25)
将化合物13(20mg,0.03mmol)溶于四氢呋喃(5mL),加入10%钯炭(5mg),室温下用氢气球氢化反应4小时。通过硅藻土过滤,滤液减压浓缩,残留物通过制备HPLC纯化得到化合物25,白色固体(1.74mg,收率9%)。ESI-MS m/z:612.2[M+H]+
实施例25
(R,Z)-3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-烯-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物26)
由化合物18出发,参照实施例24的方法得到化合物26。ESI-MS m/z:600.3[M+1]+
实施例26
3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((Z)-2-((S)-1-甲基吡咯烷-2-基)乙烯基)-1,6-萘啶-2(1H)-酮(化合物27)
由化合物19出发,参照实施例24的方法得到化合物27。ESI-MS m/z:626.3[M+1]+1H NMR(400MHz,CD3OD)δ8.21(s,1H),7.57(d,J=5.5Hz,1H),7.40(t,J=6.8Hz,1H),7.15(dd,J=14.5,6.8Hz,1H),7.13–6.81(m,2H),5.78-5.55(m,2H),4.44(d,J=11.3Hz,1H),3.81(d,J=12.9Hz,1H),3.67–3.43(m,2H),3.39(s,2H),3.16(s,3H),3.11–2.92(m,2H),2.63–1.90(m,14H),1.88–1.45(m,9H).
实施例27
3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((Z)-2-((R)-1-甲基吡咯烷-2-基)乙烯基)-1,6-萘啶-2(1H)-酮(化合物28)
由化合物20出发,参照实施例24的方法得到化合物28。ESI-MS m/z:626.3[M+1]+1H NMR(400MHz,CD3OD)δ8.22(s,1H),7.56(d,J=6.9Hz,1H),7.41(t,J=7.0Hz,1H),7.16(t,J=7.6Hz,1H),7.15-6.80(m,2H),5.66(dt,J=20.0,8.9Hz,2H),4.44(d,J=12.8Hz,1H),3.88–3.73(m,1H),3.65–3.46(m,2H),3.38(m,2H),3.16(s,3H),3.11–2.86(m,2H),2.69–1.90(m,14H),1.90–1.45(m,9H).
实施例28
3-(1-乙酰基-4-羟基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(2-((S)-1-甲基吡咯烷-2-基)乙基)-1,6-萘啶-2(1H)-酮(化合物29)
将化合物25(40mg,0.06mmol)溶于四氢呋喃(5mL),加入10%钯炭(10mg),在60℃下用氢气球氢化反应5小时。通过硅藻土过滤,滤液减压浓缩,残留物通过制备HPLC纯化得到化合物29。ESI-MS m/z:614.2[M+H]+
实施例29
(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物30)
由化合物15出发,参照实施例28的方法得到化合物30。ESI-MS m/z:588.3[M+1]+
实施例30
(R)-3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物31)
由化合物30出发,参照实施例8的步骤1和步骤2制备得到化合物31。ESI-MS m/z:602.3[M+1]+
实施例31
3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(2-((S)-1-甲基吡咯烷-2-基)乙基)-1,6-萘啶-2(1H)-酮(化合物32)
由化合物28出发,参照实施例28的方法得到化合物32。ESI-MS m/z:628.3[M+1]+
实施例32
3-(1-乙酰基-4-甲氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(2-((R)-1-甲基吡咯烷-2-基)乙基)-1,6-萘啶-2(1H)-酮(化合物33)
由化合物27出发,参照实施例28的方法得到化合物33。ESI-MS m/z:628.3[M+1]+
实施例33
(R)-3-(1-((3-(1-乙酰基-4-甲氧基哌啶-4-基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-5-基)氨基)乙基)-2-甲基苯甲腈(化合物34)
化合物34参照实施例17步骤5制备,其中将原料(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺替换为(R)-3-(1-氨基乙基)-2-甲基苯甲腈。ESI-MS m/z:569.3[M+1]+
实施例34
(R)-3-(1-((3-(1-乙酰基-4-(甲氧基-d3)哌啶-4-基)-8-(3-氨基-3-甲基丁-1-炔-1-基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-5-基)氨基)乙基)-2-甲基苯甲腈(化合物35)
化合物35参照实施例21步骤5和步骤6制备,其中将步骤5中的原料(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺替换为(R)-3-(1-氨基乙基)-2-甲基苯甲腈。ESI-MS m/z:572.3[M+1]+
实施例35
(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物63)
步骤1:3-(1-乙酰基-4-乙氧基哌啶-4-基)-8-(苄氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-羟基哌啶-4-基)-8-(苄氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮(8.8g,19.3mmol)溶于N,N-二甲基甲酰胺(100mL),在0℃下加入钠氢(3.86g,96.5mmol),搅拌5分钟后加入碘乙烷(9.0g,57.9mmol),0℃继续搅拌反应1小时。加水淬灭反应,再以乙酸乙酯萃取,饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=99:1,体积比)得到产物,黄色固体(8.0g,收率86%)。ESI-MS m/z:484.1[M+1]+1H NMR(400MHz,Chloroform-d)δ8.14(s,1H),7.39(s,5H),4.77(s,2H),4.57(ddt,J=13.2,4.8,2.2Hz,1H),3.86(s,3H),3.69(ddq,J=8.9,4.7,2.1 Hz,1H),3.50(td,J=13.0,2.7Hz,1H),3.45–3.36(m,2H),2.93(td,J=13.0,2.7Hz,1H),2.62–2.48(m,5H),2.12(s,3H),1.94–1.83(m,2H),1.36(t,J=6.9Hz,3H).
步骤2:3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-8-(苄氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮(8.0g,16.5mmol),溶于甲醇(160mL),加入二氧化铂(1.6g),在氢气球氛围下,室温下搅拌30分钟。反应物通过硅藻土过滤,滤液减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物,白色固体(6.0g,收率92%)。ESI-MS m/z:394.1[M+1]+1H NMR(400MHz,DMSO-d6)δ9.44(s,1H),7.95(s,1H),4.36–4.26(m,1H),3.86(s,3H),3.74–3.66(m,1H),3.39–3.28(m,4H),2.82–2.71(m,1H),2.43(s,3H),2.37(td,J=13.6,4.8Hz,1H),2.03(s,3H),1.79–1.68(m,2H),1.27(t,J=6.9Hz,3H).
步骤3:3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮(3.2g,8.14mmol)、4-二甲氨基吡啶(993mg,8.14mmol)和三乙胺(3.29g,32.56mmol)溶于二氯甲烷(60mL)。反应物冷却到0℃,加入苯基双(三氟甲烷磺酰)亚胺(3.49g,9.77mmol),0℃下继续反应30分钟。反应物减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=99:1,体积比)得到产物,白色固体(3.7g,收率87%)。ESI-MS m/z:526.1[M+1]+1H NMR(400MHz,Chloroform-d)δ8.18(s,1H),4.60(ddt,J=13.2,4.6,2.1Hz,1H),3.76(s,3H),3.71(dt,J=4.5,2.1Hz,1H),3.56–3.38(m,3H),2.96(td,J=13.0,2.8Hz,1H),2.63–2.48(m,2H),2.15(s,3H),1.89(ddq,J=30.9,14.2,2.7Hz,2H),1.38(t,J=6.9Hz,3H)
步骤4:3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(1.1g,2.09mmol)、N,N-二甲基炔丙胺(863mg,10.4mmol)、碘化亚铜(114mg,0.6mmol)、四三苯基磷钯(231mg,0.2mmol)、三乙胺(3mL)溶于四氢呋喃(15mL)。氮气保护,55℃下反应1小时。反应物减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=99:1,体积比)得到产物,白色固体(800mg,收率83.6%)。ESI-MS m/z:459.1[M+1]+1H NMR(400MHz,CDCl3)δ8.16(s,1H),4.62–4.50(m,1H),4.07(s,3H),3.68(ddt,J=13.2,4.5,2.2Hz,1H),3.61(s,2H),3.48(td,J=13.1,2.7Hz,1H),3.43–3.33(m,2H),2.98–2.85(m,1H),2.74(s,3H),2.51(dtd,J=18.7,13.4,4.8Hz,2H),2.40(s,6H),2.11(s,3H),1.95–1.79(m,2H),1.34(t,J=6.9Hz,3H).
步骤5:(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(600mg,1.31mmol)、甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(119mg,0.131mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺(743mg,3.93mmol)和叔丁醇钠(377mg,3.93mmol)溶于甲苯(12mL)。氮气保护,80℃搅拌反应1小时。反应物冷却至室温后,加水稀释,再以二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物,白色固体(500mg,收率62.5%)。ESI-MS m/z:612.1[M+1]+1H NMR(400MHz,DMSO-d6)δ8.17–8.11(m,1H),7.97(dd,J=7.1,2.3Hz,1H),7.65(q,J=6.7,6.1Hz,1H),7.45(t,J=7.1Hz,1H),7.36–7.04(m,2H),5.65(t,J=7.1Hz,1H),4.37–4.26(m, 1H),3.86(s,3H),3.69(d,J=13.2Hz,1H),3.47(s,2H),3.41–3.28(m,3H),3.21(dtt,J=13.5,8.9,4.4Hz,2H),2.84(tt,J=12.9,3.1Hz,1H),2.38(s,3H),2.20(s,6H),2.16–2.11(m,1H),2.02(s,4H),1.55(d,J=7.1Hz,3H),1.18(td,J=7.0,1.7Hz,3H).
实施例36
(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-5-基)氨基)乙基)-2-氟苯甲腈(化合物64)
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(400mg,0.873mmol)、甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(79mg,0.087mmol)、(R)-3-(1-氨乙基)-2-氟苯甲腈(427mg,2.62mmol)和叔丁醇钠(251mg,2.62mmol)溶于1,4-二氧六环(8mL)。氮气保护,80℃搅拌反应1小时。反应物冷却至室温后,加水稀释,再以二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物,类白色固体(310mg,收率60.6%)。ESI-MS m/z:587.1[M+1]+1H NMR(400MHz,DMSO-d6)δ8.13(s,1H),8.00(d,J=6.9Hz,1H),7.78(dt,J=18.6,6.0Hz,2H),7.34(t,J=7.7Hz,1H),5.57(t,J=7.0Hz,1H),4.32(d,J=12.5Hz,1H),3.86(s,3H),3.70(d,J=13.4Hz,1H),3.48(s,2H),3.42–3.33(m,3H),3.22(dq,J=15.4,7.8Hz,2H),2.84(d,J=11.0Hz,1H),2.38(s,3H),2.21(s,6H),2.12(d,J=15.9Hz,1H),2.03(s,4H),1.56(d,J=7.1Hz,3H),1.22–1.17(m,3H).
实施例37
(R)-3-(1-((3-(1-乙酰基-4-乙氧基哌啶-4-基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-5-基)氨基)乙基)-2-甲基苯甲腈(化合物65)
化合物65参照实施例36制备,其中将原料(R)-3-(1-氨乙基)-2-氟苯甲腈替换为(R)-3-(1-氨基乙基)-2-甲基苯甲腈。ESI-MS m/z:583.3[M+1]+
实施例38
(R)-3-(1-乙酰基-4-((2-羟乙基)氨基)哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物66)
步骤1:(R)-3-(1-乙酰基-4-氟哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物106)
将(R)-3-(1-乙酰基-4-羟基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物15,100mg,0.17mmol)溶于二氯甲烷(5mL)。氮气保护,0℃下加入二乙氨基三氟化硫(110mg,0.69mmol)。反应升至室温搅拌12小时。加入饱和碳酸氢钠水溶液淬灭反应,再用二氯甲烷萃取,饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,残留物直接用于下一步反应。ESI-MS m/z:586.3[M+1]+
步骤2:(R)-3-(1-乙酰基-4-((2-羟乙基)氨基)哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
将(R)-3-(1-乙酰基-4-氟哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(110mg,0.17mmol)、乙醇胺(207mg,3.4mmol)和三乙胺(0.5mL)溶于乙腈(5mL),90℃下搅拌16小时,反应物减压浓缩,残留物通过制备HPLC纯化得到产物,白色固体(60mg)。ESI-MS m/z:627.1[M+1]+1H NMR(400MHz,DMSO-d6)δ8.42–8.05(m,2H),7.74(q,J=7.7Hz,1H),7.45(t,J=7.1Hz,1H),7.38–7.05(m,2H),5.67(h,J=7.3Hz,1H),4.67(s,1H),4.19(d,J=10.2Hz,1H),3.87(d,J=2.7Hz,3H),3.46–3.27(m,7H),3.08(s,3H),2.47–2.34(m,4H),2.20(s,7H),2.00(s,3H),1.89–1.72(m,2H),1.59(t,J=6.7Hz,3H).
实施例39
(R)-3-(1-乙酰基-4-(乙氨基)哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物67)
将(R)-3-(1-乙酰基-4-氟哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(110mg,0.17mmol)、30%乙胺的乙醇溶液(2.5mL)和三乙胺(0.5mL)溶于乙醇(5mL),90℃下搅拌16小时,反应物减压浓缩,残留物通过制备HPLC纯化得到产物,白色固体(40mg)。ESI-MS m/z:611.1[M+1]+1H NMR(400MHz,DMSO-d6)δ8.19(s,1H),7.98(d,J=1.5Hz,1H),7.92(t,J=6.3Hz,1H),7.63(q,J=6.8Hz,1H),7.46(t,J=7.1Hz,1H),7.36–7.07(m,2H),5.66(p,J=7.3Hz,1H),4.14(t,J=12.0Hz,2H),3.90(s,3H),3.59(d,J=10.6Hz,2H),3.50(s,2H),3.15(p,J=9.4,7.8Hz,1H),2.37(d,J=1.6Hz,3H),2.22(s,9H),2.03–1.97(m,3H),1.86–1.72(m,2H),1.57(d,J=7.1Hz,3H),0.95(t,J=7.0Hz,3H).
实施例40
(R)-3-(1-乙酰基-4-(2,2,2-三氟乙氧基)哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物68)
将(R)-3-(1-乙酰基-4-氟哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(110mg,0.17mmol)加入到钠氢(136mg,3.4mmol)和2,2,2- 三氟乙醇(5mL)的反应体系中,50℃下搅拌16小时,反应物加水稀释,二氯甲烷萃取,有机相用无水硫酸钠干燥,减压浓缩,残留物通过制备HPLC纯化得到产物,白色固体(15mg,收率13.2%)。ESI-MS m/z:666.1[M+1]+1H NMR(400MHz,DMSO-d6)δ8.14(d,J=1.7Hz,1H),7.90(t,J=6.5Hz,1H),7.64(q,J=7.1Hz,1H),7.47(d,J=7.3Hz,1H),7.37–7.07(m,2H),5.72–5.59(m,1H),4.33(d,J=10.5Hz,1H),3.87(s,3H),3.83–3.70(m,3H),3.48(s,2H),3.41–3.31(m,2H),2.86(t,J=12.6Hz,1H),2.38(d,J=1.2Hz,4H),2.21(s,6H),2.03(s,5H),1.60–1.53(m,3H).
实施例41
(R)-3-(1-乙酰基-4-(2,2-二氟乙氧基)哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮)(化合物69)
化合物69参照实施例40的方法制备,其中将原料2,2,2-三氟乙醇替换为2,2-二氟乙醇。ESI-MS m/z:648.1[M+1]+1H NMR(400MHz,DMSO-d6)δ8.15(s,1H),7.90(t,J=5.9Hz,1H),7.63(q,J=6.7Hz,1H),7.46(t,J=7.1Hz,1H),7.37–7.06(m,2H),6.33–6.01(m,1H),5.65(p,J=7.0Hz,1H),4.31(d,J=11.8Hz,1H),3.87(s,3H),3.71(d,J=13.3Hz,1H),3.47(s,2H),3.37(t,J=12.4Hz,3H),2.87(t,J=12.4Hz,1H),2.37(s,4H),2.20(s,7H),2.13–2.05(m,2H),2.03(s,3H),1.56(d,J=7.1Hz,3H).
实施例42
(R)-3-(1-乙酰基-4-(2-羟基乙氧基)哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物70)
化合物70参照实施例40的方法制备,其中将原料2,2,2-三氟乙醇替换为乙二醇。ESI-MS m/z:628.3[M+1]+1H NMR(400MHz,Methanol-d4)δ8.71–8.63(m,1H),7.57(t,J=7.4Hz,1H),7.44(t,J=7.0Hz,1H),7.20(t,J=7.7Hz,1H),7.02(t,J=54.9Hz,1H),5.70(q,J=7.1Hz,1H),4.55–4.43(m,1H),4.02(s,3H),3.96–3.88(m,2H),3.84(d,J=13.7Hz,1H),3.67(s,2H),3.63–3.50(m,3H),3.11–2.97(m,1H),2.67–2.53(m,2H),2.46(d,J=4.4Hz,9H),2.17(s,3H),2.05–1.88(m,2H),1.62(d,J=7.1Hz,3H).
实施例43
(R)-3-(1-乙酰基-4-环丙氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮)(化合物71)
将(R)-3-(1-乙酰基-4-氟哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(260mg)、环丙醇(2mL)和碳酸铯(690mg,2.12mmol)溶于N,N-二甲基甲酰胺(5mL),50℃下搅拌16小时。反应物加水稀释,二氯甲烷萃取,有机相用无水硫酸钠干燥,减压浓缩,残留物通过制备HPLC纯化得到产物,白色固体(12mg)。ESI-MS m/z:624.3[M+1]+
实施例44
(R)-3-(1-乙酰基-4-异丙氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-(3-(二甲氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物72)
化合物72参照实施例40的方法制备,其中将原料2,2,2-三氟乙醇替换为异丙醇。ESI-MS m/z:626.3[M+1]+
实施例45
(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-8-(3-(二乙氨基)丙-1-炔-1-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物73)
步骤1:3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-(3-(二乙氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(250mg,0.48mmol)溶于四氢呋喃(10mL),加入N,N-二乙基炔丙胺(63mg,0.57mmol)、碘化亚酮(10mg,0.048mmol)、四三苯基膦钯(100mg,0.1mmol)和三乙胺(1mL),氮气保护,40℃下搅拌30分钟。加入水(20mL)稀释,加乙酸乙酯(50mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析纯化(二氯甲烷/甲醇=100~100:6梯度洗脱)得到产物(225mg,收率70%)。ESI-MS m/z:487.3[M+H]+1H NMR(400MHz,Chloroform-d)δ8.17(s,1H),4.59–4.52(m,1H),4.05(s,3H),3.97(s,1H),3.74–3.65(m,1H),3.55–3.33(m,4H),2.98–2.82(m,4H),2.73(s,3H),2.57–2.46(m,2H),2.12(s,3H),1.88(ddd,J=27.3,13.9,2.7Hz,2H),1.37(t,J=7.0Hz,9H).
步骤2:(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-8-(3-(二乙氨基)丙-1-炔-1-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
向50mL圆底烧瓶中加入3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-(3-(二乙氨基)丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(150mg,0.31mmol)、甲苯(6mL)、叔丁醇钠(89mg,0.93mmol)、甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(28mg,0.031mmol)和(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺(176mg,0.93mmol),氮气保护,90℃下反应1小时。反应液加入乙酸乙酯(100mL),用饱和食盐水(100mL)洗,无水硫酸钠干燥,减压浓缩,残留物通过制备HPLC纯化得到产物(30mg)。ESI-MS m/z:640.2[M+H]+1H NMR(400MHz,DMSO-d6)δ8.15(s,1H),7.98(d,J=7.2Hz,1H),7.64(t,J=6.3Hz,1H),7.46(t,J=7.3Hz,1H),7.36–7.08(m,2H),5.65(t,J=7.1Hz,1H),4.32(d,J=12.8Hz,1H),3.87(s,3H),3.71(d,J=13.4Hz,1H),3.65 (s,2H),3.25–3.18(m,4H),2.85(t,J=12.9Hz,1H),2.49–2.46(m,4H),2.37(s,3H),2.23–2.11(m,3H),2.03(s,3H),1.56(d,J=7.0Hz,3H),1.19(td,J=7.0,2.1Hz,3H),1.00(t,J=7.1Hz,6H).
实施例46
(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(3-(吡咯烷-1-基)丙-1-炔-1-基)-1,6-萘啶-2(1H)-酮(化合物74)
步骤1:3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-(3-(吡咯烷-1-基)丙-1-炔-1-基)-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(392mg,0.75mmol)溶于四氢呋喃(15mL),加入1-(丙-2-炔-1-基)吡咯烷(97mg,0.89mmol)、碘化亚酮(14mg,0.074mmol)、四三苯基膦钯(172mg,0.15mmol)和三乙胺(1.2mL),氮气保护,40℃下搅拌30分钟。反应物加入水(20mL)稀释,加乙酸乙酯(50mL)萃取。有机相用无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析纯化(二氯甲烷/甲醇=100~100:6)得到产物(210mg,收率58%)。ESI-MS m/z:485.3[M+H]+1H NMR(400MHz,Chloroform-d)δ8.16(s,1H),4.58–4.54(m,1H),4.07(s,3H),3.77(s,2H),3.71–3.66(m,2H),3.59(d,J=4.2Hz,1H),3.49(td,J=13.2,2.7Hz,2H),3.43–3.32(m,2H),2.93(td,J=13.1,2.8Hz,2H),2.74(d,J=4.1Hz,3H),2.59–2.46(m,3H),2.12(s,3H),1.90–1.81(m,4H),1.40–1.32(m,4H).
步骤2:(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基 -8-(3-(吡咯烷-1-基)丙-1-炔-1-基)-1,6-萘啶-2(1H)-酮
向50mL圆底烧瓶中加入3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-(3-(吡咯烷-1-基)丙-1-炔-1-基)-1,6-萘啶-2(1H)-酮(170mg,0.35mmol)、甲苯(6mL)、叔丁醇钠(100mg,1.05mmol)、甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(32mg,0.035mmol)和(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺(200mg,1.05mmol),氮气保护,90℃下反应1小时。反应液加入乙酸乙酯(150mL),用饱和食盐水(100mL)洗,无水硫酸钠干燥,减压浓缩,残留物通过制备HPLC纯化得到产物,淡黄色固体(68mg)。ESI-MS m/z:638.2[M+H]+1H NMR(400MHz,DMSO-d6)δ8.15(d,J=1.5Hz,1H),7.98(dd,J=7.0,2.2Hz,1H),7.65(q,J=6.6Hz,1H),7.46(t,J=7.1Hz,1H),7.38–7.06(m,2H),5.65(p,J=7.0Hz,1H),4.32(d,J=12.6Hz,1H),3.86(s,3H),3.69(dd,J=13.6,9.5Hz,1H),3.63(s,2H),3.42–3.31(m,2H),3.21(qd,J=8.9,4.0Hz,2H),2.85(ddd,J=13.3,10.0,3.3Hz,1H),2.54(d,J=6.1Hz,4H),2.37(s,3H),2.19(dtd,J=30.4,13.2,12.8,4.2Hz,3H),2.03(s,3H),1.69(q,J=3.4,3.0Hz,4H),1.56(d,J=7.0Hz,3H),1.19(td,J=7.0,2.0Hz,3H).
实施例47
(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-8-(3-(双(甲基-d3)氨基)丙-1-炔-1-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(化合物75)
步骤1:3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-(3-羟基丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基三氟甲磺酸酯(1.05g,2.0mmol)、丙炔氧基三甲基硅烷(1.28g,10.mmol)、碘化亚铜(114mg,0.6mmol)、四三苯基磷钯(231mg,0.2mmol)和三乙胺(3mL)溶于四氢呋喃(15mL),氮气保护,55℃下反应60分钟。反应物减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=99:1,体积比)得到产物,棕色固体(460mg,收率53%)。ESI-MS m/z:432.1[M+1]+1H NMR(400MHz,Chloroform-d)δ8.15(s,1H),4.59(s,2H),4.54(d,J=12.0Hz,1H),4.06(d,J=4.7Hz,3H),3.68(d,J=13.6Hz,1H),3.55–3.43(m,1H),3.38(p,J=7.9Hz,2H),2.97–2.87(m,1H),2.73(s,3H),2.51(ddt,J=17.9,13.4,6.5Hz,2H),2.12(s,3H),1.86(dd,J=26.9,13.9Hz,2H),1.35(s,3H).
步骤2:3-(3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)丙-2-炔-1-基甲磺酸酯
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-(3-羟基丙-1-炔-1-基)-1,7-二甲基-1,6-萘啶-2(1H)-酮(460mg,1.06mmol)溶于二氯甲烷(10mL),0℃下加入三乙胺(321mg,3.18mmol)和甲基磺酰氯(241mg,2.12mmol),继续0℃下搅拌反应30分钟。反应物加水稀释,再用二氯甲烷萃取,饱和碳酸氢钠洗涤,饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=98:2,体积比)得到产物,白色固体(300mg,收率55.6%)。ESI-MS m/z:510.1[M+1]+1H NMR(400MHz,Chloroform-d)δ8.17(s,1H),5.15(s,2H),4.57(d,J=14.0Hz,1H),4.04(s,3H),3.70(d,J=16.1Hz,1H),3.57–3.45(m,1H),3.39(t,J=7.2Hz,2H),3.12(s,3H),3.01–2.91(m,1H),2.75(s,3H),2.52(d,J=13.5Hz,2H),2.15(s,3H),1.97–1.80(m,2H),1.35(t,J=6.9Hz,3H).
步骤3:3-(1-乙酰基-4-乙氧基哌啶-4-基)-8-(3-(双(甲基-d3)氨基)丙-1-炔-1-基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)丙-2-炔-1-基甲磺酸酯(300mg,0.589mmol)和碳酸钾(243mg,1.76mmol)溶于N,N-二甲基乙酰胺(60mL),加入二甲基-d6-胺盐酸盐(102mg,1.17mmol),50℃下反应1小时。加水稀释,乙酸乙酯萃取,饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物,淡黄色固体(160mg,收率58.6%)。ESI-MS m/z:465.1[M+1]+1H NMR(400MHz,Chloroform-d)δ8.17(s,1H),4.63–4.53(m,1H),4.08(s,3H),3.73–3.64(m,1H),3.60(s,2H),3.49(td,J=13.1,2.6Hz,1H),3.39(qd,J=8.7,4.4Hz,2H),2.98–2.89(m,1H),2.75(s,3H),2.53(dtd,J=18.3,13.4,4.8Hz,2H),2.12(s,3H),1.94–1.86(m,2H),1.35(t,J=6.9Hz,3H).
步骤4:(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-8-(3-(双(甲基-d3)氨基)丙-1-炔-1-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-8-(3-(双(甲基-d3)氨基)丙-1-炔-1-基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮(160mg,0.344mmol)、甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(31mg,0.034mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺(195mg,1.03mmol)和叔丁醇钠(99mg,1.03mmol)溶于甲苯(5mL),氮气保护,80℃搅拌反应1小时。反应物冷却至室温后,加水稀释,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物,白色固体(170mg,收率80.5%)。ESI-MS m/z:618.1[M+1]+1H NMR(400MHz,DMSO-d6)δ8.14(s,1H),7.97(dd,J=7.2,2.3Hz,1H),7.65(q,J=6.4,6.0Hz,1H),7.46(t,J=7.1Hz,1H),7.36–7.06(m,2H),5.65(p,J=7.0Hz,1H),4.36–4.27(m,1H),3.86(s,3H),3.70 (d,J=13.3Hz,1H),3.47(s,2H),3.21(dtt,J=13.3,8.7,4.3Hz,2H),2.84(tt,J=12.8,3.1Hz,1H),2.38(s,3H),2.26–2.09(m,3H),2.02(s,5H),1.55(d,J=7.1Hz,3H),1.17(dd,J=6.9,1.9Hz,3H).
实施例48
(R)-3-(1-((3-(1-乙酰基-4-乙氧基哌啶-4-基)-8-(3-(双(甲基-d3)氨基)丙-1-炔-1-基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-5-基)氨基)乙基)-2-氟苯甲腈(化合物76)
化合物76参照实施例47的步骤4制备,其中将原料(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺替换为(R)-3-(1-氨乙基)-2-氟苯甲腈。ESI-MS m/z:593.3[M+1]+
实施例49
(R)-3-(1-((3-(1-乙酰基-4-乙氧基哌啶-4-基)-8-(3-(双(甲基-d3)氨基)丙-1-炔-1-基)-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-5-基)氨基)乙基)-2-甲基苯甲腈(化合物77)
化合物77参照实施例47的步骤4制备,其中将原料(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺替换为(R)-3-(1-氨基乙基)-2-甲基苯甲腈。ESI-MS m/z:589.4[M+1]+
实施例50
(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((1-甲基哌啶-4-基)氧)-1,6-萘啶-2(1H)-酮(化合物78)
步骤1:叔丁基4-((3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧)哌啶-1-羧酸酯
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮(393mg,1.0mmol)、N-叔丁氧羰基-4-羟基哌啶(519mg,3.0mmol)和三苯基膦(1.05g,4.0mmol)溶于四氢呋喃(10mL),氮气保护,0℃下加入偶氮二甲酸二异丙酯(909mg,4.5mmol),搅拌反应1小时。反应物加水稀释,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=99:2,体积比)得到产物,黄色固体(540mg,收率93%)。ESI-MS m/z:577.1[M+1]+1H NMR(400MHz,Chloroform-d)δ8.12(s,1H),4.56(d,J=13.0Hz,1H),4.19–4.07(m,2H),3.80(s,4H),3.75–3.62(m,1H),3.56–3.31(m,3H),2.99–2.86(m,1H),2.73–2.57(m,3H),2.55(m,4H),2.11(s,3H),1.87(dd,J=23.9,13.8Hz,4H),1.68–1.57(m,2H),1.44(s,9H),1.35(t,J=6.9Hz,3H).
步骤2:3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-(哌啶-4-基氧基)-1,6-萘啶-2(1H)-酮
将叔丁基4-((3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧)哌啶-1-羧酸酯(520mg,0.903mmol)溶于二氯甲烷(8mL),加入三氟乙酸(4mL),室温下搅拌15分钟。反应物减压浓缩,残留物直接用于下一步反应。ESI-MS m/z:477.1[M+1]+
步骤3:3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-((1-甲基哌啶-4-基)氧基)-1,6-萘啶-2(1H)-酮
将步骤2得到的产物粗品溶于四氢呋喃(10mL),加入30%甲醛水溶液(1mL)和三乙酰氧基硼氢化钠(1.82g,9.03mmol),室温下搅拌1小时,反应物加水稀释,乙酸乙酯萃取,有机相依次用饱和碳酸氢钠溶液和饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物,淡黄色固体(420mg)。ESI-MS m/z:491.1[M+1]+1H NMR(400MHz,Chloroform-d)δ8.11(s,1H),4.55(ddt,J=13.1,4.6,2.1Hz,1H),3.81(s,3H),3.75–3.63(m,2H),3.53–3.33(m,3H),2.99–2.80(m,3H),2.63–2.47(m,5H),2.26(s,3H),2.11(s,3H),1.97–1.81(m,8H),1.34(t,J=6.9Hz,3H).
步骤4:(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((1-甲基哌啶-4-基)氧)-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-((1-甲基哌啶-4-基)氧)-1,6-萘啶-2(1H)-酮(400mg,0.816mmol)、甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(74mg,0.081mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺(461mg,2.44mmol)和叔丁醇钠(234mg,2.44mmol)溶于甲苯(10mL),氮气保护,80℃搅拌反应1小时。反应物冷却至室温后,加水稀释,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物,白色固体(370mg,收率70%)。ESI-MS m/z:644.1[M+1]+1H NMR(400MHz,DMSO-d6)δ8.12(s,1H),7.67(q,J=6.1,5.2Hz,1H),7.51–7.39(m,2H),7.36–7.07(m,2H),5.56(t,J=7.1Hz,1H),4.41–4.26(m,1H),3.69(d,J=12.8Hz,1H),3.63(s,3H),3.47(dt,J=10.0,4.5Hz,2H),3.43–3.32(m,3H),3.28–3.16(m,2H),2.84(dddd,J=13.3,10.7,6.8,3.7Hz,1H),2.72–2.61(m,2H),2.20(s,3H),2.08(s,3H),2.02(s,3H),1.85–1.75(m,2H),1.72–1.54(m,4H),1.52(d,J=7.1Hz,3H),1.20(dd,J=7.0,1.6Hz,3H).
实施例51
(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((1-甲基氮杂环丁烷-3-基)氧)-1,6-萘啶-2(1H)-酮(化合物79)
步骤1:叔丁基3-((3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)氮杂环丁烷-1-羧酸酯
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮(393mg,1.0mmol)、N-叔丁氧羰基-3-羟基氮杂环丁烷(519mg,3.0mmol)和三苯基膦(1.05g,4.0mmol)溶于四氢呋喃(10mL),氮气保护,0℃下加入偶氮二甲酸二异丙酯(909mg,4.5mmol),搅拌反应1小时。反应物加水稀释,乙酸乙酯萃取,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=99:2,体积比)得到产物,黄色固体(280mg,收率51%)。ESI-MS m/z:549.1[M+1]+
步骤2:3-(1-乙酰基-4-乙氧基哌啶-4-基)-8-(氮杂环丁烷-3-基氧基)-5-氯-1,7-二甲基-1,6-萘啶-2(1H)-酮
将叔丁基3-((3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)氮杂环丁烷-1-羧酸酯(260mg,0.474mmol)溶于二氯甲烷(4mL),加入三氟乙酸(2mL),室温下搅拌15分钟。反应物减压浓缩,残留物直接用于下一步反应。ESI-MS m/z:449.1[M+1]+
步骤3:3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-((1-甲基氮杂环丁烷-3-基)氧基)-1,6-萘啶-2(1H)-酮
将步骤2得到的产物粗品溶于四氢呋喃(10mL),加入30%甲醛水溶液(0.5mL)和三乙酰氧基硼氢化钠(1.0g,4.74mmol),室温下搅拌1小时。反应物加水稀释,乙酸乙酯萃取,有机相用饱和碳酸氢钠溶液洗涤,饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物,淡黄色固体(180mg,收率82%)。ESI-MS m/z:463.1[M+1]+1H NMR(400MHz,Chloroform-d)δ8.12(s,1H),4.62–4.52(m,1H),4.31(p,J=6.1Hz,1H),3.80(s,3H),3.75–3.65(m,3H),3.53–3.34(m,3H),3.27(td,J=6.2,2.0Hz,2H),2.92(td,J=13.1,2.8Hz,1H),2.60–2.46(m,5H),2.41(s,3H),2.12(s,3H),1.95–1.80(m,2H),1.35(t,J=6.9Hz,3H).
步骤4:(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-((1-甲基氮杂环丁烷-3-基)氧基)-1,6-萘啶-2(1H)-酮
将3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-((1-甲基氮杂环丁烷-3-基)氧基)-1,6-萘啶-2(1H)-酮(165mg,0.357mmol)、甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(32mg,0.036mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺(202mg,1.07mmol)和叔丁醇钠(98mg,1.07mmol)溶于甲苯(5mL),氮气保护,80℃搅拌反应1小时。反应物冷却至室温后,加水稀释,二氯甲烷萃取,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物,白色固体(150mg,收率68%)。ESI-MS m/z:616.1[M+1]+1H NMR(400MHz,DMSO-d6)δ8.11(s,1H),7.67(q,J=6.5,5.7Hz,1H),7.54(dd,J=7.3,2.3Hz,1H),7.44(t,J=7.1Hz,1H),7.35–7.07(m,2H),5.56(p,J=7.1Hz,1H),4.31(d,J=12.6Hz,1H),4.17(p,J=5.9Hz,1H),3.69(d,J=14.3Hz,1H),3.62(s,3H),3.59–3.53(m,2H),3.36(tt,J=13.0,3.3Hz,2H),3.27–3.17(m,2H),3.09(d,J=7.0Hz,2H),2.85(ddt,J=13.9,10.0,4.1Hz,1H),2.26(s,3H),2.17(s,3H),2.03(s,5H),1.52(d,J=7.1Hz,3H),1.21–1.17(m,3H).
实施例52
(R)-3-(1-((3-(1-乙酰基-4-乙氧基哌啶-4-基)-1,7-二甲基-8-((1-甲基氮杂环丁烷-3-基)氧基)-2-氧代-1,2-二氢-1,6-萘啶-5-基)氨基)乙基)-2-氟苯甲腈(化合物80)
化合物80参照实施例51的步骤4制备,其中将原料(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺替换为(R)-3-(1-氨乙基)-2-氟苯甲腈。ESI-MS m/z:591.3[M+1]+
实施例53
3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((S)-1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮(化合物81)
步骤1:叔丁基(S)-2-(((3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲基)吡咯烷-1-羧酸酯
向50mL的圆底烧瓶中加入3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮(600mg,1.53mmol)、(S)-1-叔丁氧羰基-2-吡咯烷甲醇(920mg,4.58mmol)、无水四氢呋喃(10mL)以及三苯基膦(800mg,3.05mmol),氮气保护,反应液冷却至0℃,加入偶氮二甲酸二异丙酯(616mg,3.05mmol),0℃继续反应2小时。反应液中加入饱和氯化铵溶液(50mL),再用乙酸乙酯(50mL)萃取两次,合并有机相,用饱和食盐水(100mL)洗,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析纯化(二氯甲烷/甲醇=100~100:5)得到产物,黄色固体(526mg,60%)。ESI-MS m/z:577.3[M+H]+1H NMR(400MHz,Chloroform-d)δ8.12(d,J=3.5Hz,1H),5.01–4.93(m,1H),4.87–4.81(m,1H),4.54(s,2H),4.36–4.11(m,1H),3.90(s,3H),3.82(s,2H),3.40–3.29(m,5H),2.57(s,2H),2.47–2.28(m,2H),2.15(d,J=4.4Hz,3H),2.00–1.84(m,4H),1.44–1.42(m,4H),1.39–1.22(m,10H).
步骤2:(S)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-(吡咯烷-2-基甲氧基)-1,6-萘啶-2(1H)-酮
向50mL的圆底烧瓶中加入叔丁基(S)-2-(((3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)甲基)吡咯烷-1-羧酸酯(500mg,0.86mmol)、二氯甲烷(6mL)和三氟乙酸(3mL),室温反应1小时。反应物减压蒸干溶剂,得到粗产物,直接用于下一步反应。ESI-MS m/z:477.2[M+1]+
步骤3:(S)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-((1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮
将步骤2得到的产物粗品溶于四氢呋喃(10mL),加入30%甲醛水溶液(0.5mL)和三乙酰氧基硼氢化钠(936mg,4.47mmol),室温搅拌1小时。反应物中加入饱和碳酸氢钠溶液,再用二氯甲烷萃取,有机相用无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物(383mg,收率90%)。ESI-MS m/z:491.2[M+1]+1H NMR(400MHz,Chloroform-d)δ8.14(s,1H),4.62–4.53(m,1H),3.91(s,3H),3.81(s,1H),3.75–3.59(m,2H),3.50(td,J=13.1,2.6Hz,1H),3.45–3.36(m,2H),2.97–2.90(m,1H),2.77(s,1H),2.61(s,3H),2.59–2.45(m,5H),2.36–2.27(m,1H),2.13(s,3H),1.99–1.81(m,6H),1.36(t,J=6.9Hz,3H).
步骤4:3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((S)-1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮
向50mL圆底烧瓶中加入(S)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-((1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮(370mg,0.75mmol)、甲苯(8mL)、叔丁醇钠(217mg,2.26mmol)、甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2',4',6'-三异丙基-1,1'-联苯)(2'-氨基-1,1'-联苯-2-基)钯(II)(68mg,0.075mmol)和(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺(429mg,2.26mmol),氮气保护,90℃下反应1小时。反应液冷却至室温,加入乙酸乙酯(150mL),用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物(371mg,收率76%)。ESI-MS m/z:644.2[M+H]+1H NMR(400MHz,DMSO-d6)δ8.12(s,1H),7.66(d,J=8.9Hz,1H),7.53–7.41(m,2H),7.37–7.07(m,2H),5.58(p,J=7.1Hz,1H),4.32(d,J=12.5Hz,1H),3.75(s,3H),3.71(d,J=13.6Hz,1H),3.57(dd,J=9.2,5.3Hz,1H),3.46–3.40(m,2H),3.30–3.17(m,2H),2.95(dd,J=9.2,4.9Hz,1H),2.85(t,J=12.6Hz,1H),2.34(s,3H),2.31–2.26(m,1H),2.23(s,3H),2.23–2.11(m,3H),2.04(s,3H),1.99–1.93(m,1H),1.71–1.56(m,3H),1.53(d,J=7.1Hz,3H),1.27–1.17(m,5H).
实施例54
3-((R)-1-((3-(1-乙酰基-4-乙氧基哌啶-4-基)-1,7-二甲基-8-(((S)-1-甲基吡咯烷-2-基)甲氧基)-2-氧代-1,2-二氢-1,6-萘啶-5-基)氨基)乙基)-2-氟苯甲腈(化合物82)
化合物82参照实施例53的步骤4制备,其中将原料(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺替换为(R)-3-(1-氨乙基)-2-氟苯甲腈。ESI-MS m/z:619.3[M+1]+
实施例55
3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((R)-1-甲基吡咯烷-3-基)氧基)-1,6-萘啶-2(1H)-酮(化合物83)
步骤1:叔丁基(R)-3-((3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)吡咯烷-1-羧酸酯
向50mL的圆底烧瓶中加入3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮(250mg,0.64mmol)、(S)-3-羟基吡咯烷-1-羧酸叔丁酯(356mg,1.91mmol)、无水四氢呋喃(8mL)以及三苯基膦(833mg,3.18mmol),氮气保护,反应液冷却至0℃,滴加偶氮二甲酸二异丙酯(513mg,2.54mmol),加完后继续在0℃反应2小时。反应液中加入饱和氯化铵溶液(50mL),用乙酸乙酯(50mL)萃取两次,合并有机相,以饱和食盐水(100mL)洗,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析纯化(二氯甲烷/甲醇=100~100:5)得到产物。ESI-MS m/z:563.3[M+H]+1H NMR(400MHz,Chloroform-d)δ8.15(s,1H),4.59(s,1H),4.51–4.40(m,2H),3.81(s,3H),3.63(dd,J=34.3,8.9Hz,3H),3.55–3.20(m,6H),2.54(s,3H),2.16(s,3H),2.03–1.84(m,6H),1.47(s,9H),1.38(t,J=6.9Hz,3H).
步骤2:(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-(吡咯烷-3-基氧基)-1,6-萘啶-2(1H)-酮
向50mL的圆底烧瓶中加入叔丁基(R)-3-((3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-2-氧代-1,2-二氢-1,6-萘啶-8-基)氧基)吡咯烷-1-羧酸酯(430mg,0.76mmol)、二氯甲烷(6mL)和三氟乙酸(3mL),室温反应1小时。反应物减压蒸干溶剂,得到粗产物,直接用于下一步反应。ESI-MS m/z:463.2[M+1]+
步骤3:(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-((1-甲基吡咯烷-3-基)氧基)-1,6-萘啶-2(1H)-酮
将步骤2得到的产物粗品溶于四氢呋喃(10mL),加入30%甲醛水溶液(0.5mL)和三乙酰氧基硼氢化钠(812mg,3.83mmol),室温搅拌1小时。反应物中加入饱和碳酸氢钠溶液,再用二氯甲烷萃取,有机相用无水硫酸钠干燥,减压浓缩,残留物硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物(280mg,收率77%)。ESI-MS m/z:477.2[M+1]+1H NMR(400MHz,Chloroform-d)δ8.13(d,J=0.9Hz,1H),4.58(d,J=13.2Hz,1H),4.46(s,1H),3.85(s,3H),3.69(d,J=13.8Hz,1H),3.57–3.34(m,3H),3.03–2.79(m,3H),2.64–2.60(m,16H),2.59(s,3H),2.56–2.38(m,6H),2.13(s,3H),2.07(d,J=6.6Hz,1H),1.96–1.80(m,3H),1.36(t,J=6.9Hz,3H).
步骤4:3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((R)-1-甲基吡咯烷-3-基)氧基)-1,6-萘啶-2(1H)-酮
向50mL圆底烧瓶中加入(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-((1-甲基吡咯烷-3-基)氧基)-1,6-萘啶-2(1H)-酮(114mg,0.24mmol)、甲苯(8mL)、叔丁醇钠(69mg,2.26mmol)、甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2’,4’,6’-三异丙基-1,1’-联苯)(2’-氨基-1,1’-联苯-2-基)钯(II)(22mg,0.023mmol)和(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺(136mg,0.72mmol),氮气保护,90℃下反应1小时。反应液冷却至室温,加入乙酸乙酯(150mL),用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物(120mg,收率79%)。ESI-MS m/z:630.2[M+H]+1H NMR(400MHz,DMSO-d6)δ8.13(d,J=1.5Hz,1H),7.71–7.65(m,1H),7.54–7.41(m,2H),7.39–7.06(m,2H),5.57(p,J=7.0Hz,1H),4.32(d,J=12.7Hz,1H),4.26(td,J=5.4,2.5Hz,1H),3.72(s,1H),3.68(s,3H),3.37(qd,J=12.4,11.5,4.9Hz,2H),3.29–3.17(m,3H),2.88–2.79(m,2H),2.66(d,J=10.8Hz,1H),2.23(d,J=6.1Hz,6H),2.21–2.09(m,4H),2.04(s,3H),1.91(s,1H),1.83–1.74(m,1H),1.53(d,J=7.0Hz,3H),1.20(ddd,J=9.0,5.8,2.3Hz,3H).
实施例56
3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((S)-1-甲基吡咯烷-3-基)氧基)-1,6-萘啶-2(1H)-酮(化合物84)
化合物84参照实施例55的方法制备,其中将步骤1中的原料(S)-3-羟基吡咯烷-1-羧酸叔丁酯替换为(R)-3-羟基吡咯烷-1-羧酸叔丁酯。ESI-MS m/z:630.2[M+H]+1H NMR(400MHz,DMSO-d6)δ8.12(s,1H),7.68(q,J=6.8,6.4Hz,1H),7.53–7.42(m,2H),7.39–7.07(m,2H),5.58(p,J=7.0Hz,1H),4.33 (d,J=12.7Hz,1H),4.26(d,J=6.7Hz,1H),3.72(d,J=6.7Hz,1H),3.68(s,3H),3.40–3.34(m,4H),3.29–3.17(m,2H),2.90–2.80(m,2H),2.65–2.59(m,1H),2.23(d,J=5.2Hz,6H),2.19–2.07(m,3H),2.04(s,3H),1.87–1.78(m,1H),1.53(d,J=7.1Hz,3H),1.24–1.16(m,4H).
实施例57
(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(2-(吡咯烷-1-基)乙氧基)-1,6-萘啶-2(1H)-酮(化合物85)
步骤1:3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-(2-(吡咯烷-1-基)乙氧基)-1,6-萘啶-2(1H)-酮
向50mL的圆底烧瓶中加入3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-8-羟基-1,7-二甲基-1,6-萘啶-2(1H)-酮(100mg,0.25mmol)、N-(2-氯乙基)吡咯烷盐酸盐(43mg,0.25mmol)、N,N-二甲基甲酰胺(8mL)以及碳酸钾(833mg,3.18mmol),氮气保护,反应液60℃下反应2小时。向反应物中加入水(100mL)和乙酸乙酯(100mL),分出有机相,用饱和食盐水(100mL)洗,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析纯化(二氯甲烷/甲醇=100~100:5)得到产物,黄色固体(81mg,65%)。ESI-MS m/z:491.3[M+H]+1H NMR(400MHz,Chloroform-d)δ8.14(s,1H),4.58(d,J=13.0Hz,1H),4.09(s,2H),3.88(s,3H),3.70(d,J=13.1Hz,1H),3.57–3.34(m,4H),3.26(s,2H),2.99–2.88(m,1H),2.64(s,3H),2.61–2.46(m,3H),2.13(s,3H),2.02(s,5H),1.95–1.82(m,4H),1.36(t,J=6.9Hz,3H).
步骤2:(R)-3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(2-(吡咯烷-1-基)乙氧基)-1,6-萘啶-2(1H)-酮
向50mL圆底烧瓶中加入3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-氯-1,7-二甲基-8-(2-(吡咯烷-1-基)乙氧基)-1,6-萘啶-2(1H)-酮(80mg,0.16mmol)、甲苯(5mL)、叔丁醇钠(47mg,0.49mmol)、甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2’,4’,6’-三异丙基-1,1’-联苯)(2’-氨基-1,1’-联苯-2-基)钯(II)(15mg,0.016mmol)和(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺(93mg,0.49mmol),氮气保护,90℃下反应1小时。反应液加入乙酸乙酯(150mL),用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,减压浓缩,残留物通过硅胶柱层析(二氯甲烷/甲醇=95:5,体积比)得到产物(66mg,收率70%)。ESI-MS m/z:644.2[M+H]+1H NMR(400MHz,DMSO-d6)δ8.13(s,1H),7.68(s,1H),7.54–7.41(m,2H),7.38–7.07(m,2H),5.57(p,J=7.1Hz,1H),4.31(dd,J=11.6,8.4Hz,1H),3.73(s,3H),3.67(q,J=6.9,6.0Hz,3H),3.30–3.18(m,2H),2.85(ddd,J=13.2,10.0,3.8Hz,1H),2.71(t,J=5.9Hz,2H),2.43(q,J=3.8Hz,4H),2.32–2.24(m,1H),2.23(s,3H),2.21–2.08(m,1H),2.04(s,3H),1.65–1.61(m,4H),1.53(d,J=7.1Hz,3H),1.34(d,J=6.1Hz,1H),1.24–1.17(m,5H).
实施例58
(R)-3-(1-((3-(1-乙酰基-4-乙氧基哌啶-4-基)-1,7-二甲基-2-氧代-8-(2-(吡咯烷-1-基)乙氧基)-1,2-二氢-1,6-萘啶-5-基)氨基)乙基)-2-氟苯甲腈(化合物86)
化合物86参照实施例57的步骤2制备,其中将原料(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺替换 为(R)-3-(1-氨乙基)-2-氟苯甲腈。ESI-MS m/z:619.3[M+1]+
实施例59
3-(1-乙酰基-4-乙氧基哌啶-4-基)-5-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,7-二甲基-8-(((R)-1-甲基吡咯烷-2-基)甲氧基)-1,6-萘啶-2(1H)-酮(化合物87)
化合物87参照实施例53的方法制备,其中将步骤1中的原料(S)-1-叔丁氧羰基-2-吡咯烷甲醇替换为(R)-1-叔丁氧羰基-2-吡咯烷甲醇。ESI-MS m/z:644.3[M+1]+
实施例60
3-((R)-1-((3-(1-乙酰基-4-乙氧基哌啶-4-基)-1,7-二甲基-8-(((R)-1-甲基吡咯烷-2-基)甲氧基)-2-氧代-1,2-二氢-1,6-萘啶-5-基)氨基)乙基)-2-氟苯甲腈(化合物88)
化合物88参照实施例53的方法制备,其中将步骤1中的原料(S)-1-叔丁氧羰基-2-吡咯烷甲醇替换为(R)-1-叔丁氧羰基-2-吡咯烷甲醇,步骤4中的原料(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺替换为(R)-3-(1-氨乙基)-2-氟苯甲腈。ESI-MS m/z:619.3[M+1]+
生物测试实施例
实施例A:化合物对NCI-H358和MIA PaCa-2肿瘤细胞株在3D培养条件下的细胞生长抑制作 用。
细胞来源:NCI-H358购自上海迪津生物科技有限公司;MIA PaCa-2购自上海迪奥生物科技有限公司。
取处于对数生长期的细胞接种在超低吸附96孔板中(NCI-H358,MIA PaCa-2细胞分别为4000,2000个/孔,180μl/孔),于37℃、5%CO2培养,使细胞聚集形成微球,1天后加入梯度稀释的待测化合物。具体如下:取事先溶解在DMSO中的化合物储存液(10mM),倍比(4倍)稀释为10个梯度浓度,并用培养基在另一96孔板中稀释到目的浓度的10倍,然后在接种细胞的96孔板中加入化合物溶液20μl/孔,即到达目的浓度(10000、2500、625、156、39、10、2.5、0.6、0.15、0.04nM)。每个浓度设3个复孔,并设空白对照。放入37℃、5%CO2中继续培养6天后,每孔加入50μl CellTiter-3D试剂(用于检测3D细胞微球的荧光素酶ATP生物发光检测试剂,购自Promega,货号G9683),震荡10min,室温孵育20min后,检测荧光发光强度(收光时间为100ms)。计算各浓度化合物对3D细胞微球的活性抑制率,细胞活性抑制率(%)=[(发光强度6天含细胞培养基对照组-发光强度6天化合物组)/(发光强度6天含细胞培养基对照组–发光强度6天无细胞培养基对照组)]×100%。使用GraphPad Prism 8.3软件分析数据,利用非线性S曲线回归来拟合数据得出剂量-效应曲线,并由此计算IC50值。结果见表1。
表1

表中“--”表示未检测。
其中,BI-3406为文献报道的SOS1抑制剂(Cancer Discovery 2021(11)142-157),其结构为:
结果表明,本发明化合物对NCI-H358和MIA PaCa-2肿瘤细胞株表现出良好的细胞生长抑制作用。
实施例B:大鼠体内药代动力学测试
以SD大鼠为受试动物,研究了本发明化合物口服和静脉注射给药在大鼠体内的药代动力学行为。
口服给药组:雌性SD大鼠3只,禁食12h后灌胃给予测试化合物(剂量为5mg/kg,给药体积为10mL/kg,溶媒为10%DMSO/10%Solutol/80%生理盐水)。采样时间点为给药后0.25h,0.5h,1h,2h,3h,4h,6h,8h和24h。[Solutol是指聚乙二醇(15)-羟基硬脂酸酯。]
静脉给药组:雌性SD大鼠3只,禁食12h后静脉注射给予测试化合物(剂量为1mg/kg,给药体积为5mL/kg,溶媒为10%DMSO/10%Solutol/80%生理盐水)。采样时间点为给药后5min,0.25h,0.5h,1h,2h,4h,8h和24h。
在以上设定时间点经大鼠眼球后静脉丛取静脉血0.2mL,置于K2-EDTA(乙二胺四乙酸二钾)试管中,4℃下6800g离心6min,分离血浆,于-80℃下保存。
取血浆样品20μL,加入200μL甲醇(含10ng/mL的内标物维拉帕米),涡旋混合1min后18000g离心7min,转移200μL上清液至96孔板,以液相色谱-串联质谱法测定血浆中化合物的浓度,并用WinNonlin 7.0软件计算化合物的药代动力学主要参数,结果见表2。
表2

AUC:血浆暴露量;Cmax:最大血药浓度;V:表观分布容积;CL:清除率;F:口服生物利用度。
结果表明,本发明化合物的药代动力学性质,如血浆暴露量AUC和最大血药浓度Cmax都表现较好。

Claims (19)

  1. 一种如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,
    其中:
    环A为C6-12芳基、5-10元杂芳基、C3-7环烷基、C3-7环烯基或3-7元杂环基;
    每个R1独立地为氘、羟基、卤素、-N(R7)2、-SR9、氰基、氧代、硝基、C1-6烷基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1a取代的C1-6烷基、被一个或多个R1b取代的C1-6烷基-O-、被一个或多个R1c取代的C2-6烯基、被一个或多个R1d取代的C2-6炔基、C3-7环烷基、被一个或多个R1e取代的C3-7环烷基、3-7元杂环基、被一个或多个R1f取代的3-7元杂环基、C6-12芳基、被一个或多个R1g取代的C6-12芳基、5-10元杂芳基、被一个或多个R1h取代的5-10元杂芳基、C3-7环烯基、被一个或多个R1i取代的C3-7环烯基、-C(=O)-O-R9、-O-C(=O)-R9、-C(=O)-N(R7)2、-S(O)2-R9、-NR8-S(O)2-R9、-NR8-S(O)2-N(R7)2、-S(O)2-N(R7)2、-S(O)-N(R7)2、-S(O)-R9、-NR8-S(O)-R9、-NR8-S(O)-N(R7)2、-C(=O)-R9、-NR8C(=O)-R9或-NR8C(=O)-O-R9,或两个R1与所相连的环原子一起形成C3-7环烷基、被一个或多个R1e取代的C3-7环烷基、3-7元杂环基、被一个或多个R1f取代的3-7元杂环基、C3-7环烯基或被一个或多个R1i取代的C3-7环烯基;当取代基为多个时,相同或不同;
    每个R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h和R1i各自独立地为氘、羟基、卤素、-N(R7)2、-SR9、硝基、氰基、C1-6烷基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1-a取代的C1-6烷基、被一个或多个R1-b取代的C1-6烷基-O-、被一个或多个R1-c取代的C2-6烯基、被一个或多个R1-d取代的C2- 6炔基、C3-7环烷基、C3-7环烷基-O-、3-7元杂环基、3-7元杂环基-O-、被一个或多个R1-e取代的C3-7环烷基、被一个或多个R1-e取代的C3-7环烷基-O-、被一个或多个R1-f取代的3-7元杂环基、被一个或多个R1-f取代的3-7元杂环基-O-、C6-12芳基、被一个或多个R1-g取代的C6-12芳基、5-10元杂芳基、被一个或多个R1-h取代的5-10元杂芳基、C3-7环烯基、C3-7环烯基-O-、被一个或多个R1-i取代的C3-7环烯基、被一个或多个R1-i取代的C3-7环烯基-O-、-C(=O)-O-R9、-O-C(=O)-R9、-C(=O)-N(R7)2、-S(O)2-R9、-NR8-S(O)2-R9、-NR8-S(O)2-N(R7)2、-S(O)2-N(R7)2、-S(O)-N(R7)2、-S(O)-R9、-NR8-S(O)-R9、-NR8-S(O)-N(R7)2、-C(=O)-R9、-NR8C(=O)-R9或-NR8C(=O)-O-R9;当取代基为多个时,相同或不同;
    每个R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h和R1-i各自独立地为氘、羟基、卤素、-N(R7)2、-SR9、硝基、氰基、C1-6烷基、C1-6烷基-O-、C2-6烯基、C2-6炔基、C3-7环烷基、C3-7环烯基或3-7元杂环基;
    环D为C6-12芳基、5-6元杂芳基、C3-7环烷基、C3-7环烯基或3-7元杂环基;
    R2为不存在、氢、氘、羟基、卤素、氨基、氰基、氧代、硝基、C1-6烷基、C1-6烷基-O-、C2-6烯基、C2-6炔基、C3-7环烷基、C3-7环烯基或3-7元杂环基;
    L为连接键、-C(=O)-、-C(=O)O-、-C(=O)NR8-、-NR8-、-S-、-O-、-S(O)-、-S(O)2-或-(CH2)p-;当L为连接键时,表示R3直接与D环连接;
    R3为氢、氘、C1-6烷基、被一个或多个R3a取代的C1-6烷基、C2-6烯基、被一个或多个R3b取代的C2-6烯基、C2-6炔基、被一个或多个R3c取代的C2-6炔基、C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、3-7元杂环基、被一个或多个R3e取代的3-7元杂环基、C6-12芳基、被一个或多个R3f取代的C6-12芳基、5-10元杂芳基、被一个或多个R3g取代的5-10元杂芳基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;当取代基为多个时,相同或不同;
    每个R3a、R3b、R3c、R3d、R3e、R3f、R3g和R3h各自独立地为氘、羟基、卤素、-N(R7)2、氰基、硝基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R3-c取代的C2-6烯基、被一个或多个R3-d取代的C2-6炔基、C3-7环烷基、被一个或多个R3-e取代的C3-7环烷基、3-7元杂环基、被一个或多个R3-f取代的3-7元杂环基、C6-12芳基、被一个或多个R3-g取代的C6-12芳基、5-10元杂芳基、被一个或多个R3-h取代的5-10元杂芳基、C3-7环烯基、被一个或多个R3-i取代的C3-7环烯基、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、-SR9、-C(=O)-O-R9、-O-C(=O)-R9、-C(=O)-N(R7)2、-S(O)2-R9、-NR8-S(O)2-R9、-NR8-S(O)2-N(R7)2、-S(O)2-N(R7)2、-S(O)-N(R7)2、-S(O)-R9、-NR8-S(O)-R9、-NR8-S(O)-N(R7)2、-C(=O)-R9、-NR8C(=O)-R9或-NR8C(=O)-O-R9,或两个R3d、两个R3e、两个R3f、两个R3g或两个R3h与所相连的环原子一起形成C3-7环烷基、被一个或多个R3-e取代的C3-7环烷基、3-7元杂环基、被一个或多个R3-f取代的3-7元杂环基、C3-7环烯基或被一个或多个R3-i取代的C3-7环烯基;当取代基为多个时,相同或不同;
    每个R3-a、R3-b、R3-c、R3-d、R3-e、R3-f、R3-g、R3-h、R3-i和R3-j各自独立地为氘、羟基、卤素、-N(R7)2、硝基、氰基、C1-6烷基、C1-6烷基-O-、C3-7环烷基、C3-7环烯基、3-7元杂环基、-SR9、-C(=O)-O-R9、-O-C(=O)-R9、-C(=O)-N(R7)2、-S(O)2-R9、-NR8-S(O)2-R9、-NR8-S(O)2-N(R7)2、-S(O)2-N(R7)2、-C(=O)-R9、-NR8C(=O)-R9或-NR8C(=O)-O-R9
    X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;
    R4为氢、C1-6烷基、C1-6烷基-O-、氰基、卤素、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
    每个R4a和R4b各自独立地为氢、氘、羟基、卤素、氨基、氰基、硝基、氨基保护基团、C1-6烷基或被一个或多个氘取代的C1-6烷基;
    R5为不存在、氧代、氢、氘、羟基、卤素、氨基、氰基、硝基、C1-6烷基、被一个或多个R5a取代的C1-6烷基、C1-6烷基-O-、C2-6烯基、C2-6炔基、C3-7环烷基、被一个或多个R5b取代的C3-7环烷基、3-7元杂环基、被一个或多个R5c取代的3-7元杂环基、C3-7环烯基或被一个或多个R5d取代的C3-7环烯基;当取代基为多个时,相同或不同;
    每个R5a、R5b和R5c各自独立地为氘、C1-6烷基、羟基、卤素、氨基或氰基;
    R6为氢、氘、羟基、卤素、氨基、氰基、氧代、C1-6烷基、C1-6烷基-O-、C2-6烯基、C2-6炔基或不存在;
    每个R7独立地为氢、C1-6烷基、被一个或多个R7b取代的C1-6烷基、C3-7环烷基、C3-7环烯基或氨基保护基团,或两个R7与所相连的氮原子一起形成3-7元杂环基或被一个或多个R7a取代的3-7元杂环基;当取代基为多个时,相同或不同;
    每个R7a和R7b各自独立地为氘、C1-6烷基、卤素、羟基、氨基、氰基、C1-6烷基-O-、C2-6烯基或C2-6炔基;
    每个R8和R9各自独立地为氢、C1-6烷基、C2-6烯基、C2-6炔基、C3-7环烷基、C3-7环烯基或3-7元杂环基;
    R10为氢、氘、羟基、卤素、氨基、氰基、C1-6烷基-O-、C2-6烯基、C2-6炔基或C1-6烷基;
    R11为氰基、C1-6烷基或被一个或多个R11a取代的C1-6烷基;当取代基为多个时,相同或不同;
    每个R11a独立地为氘、卤素或羟基;
    m为0、1、2、3、4或5;
    p为1、2、3、4、5或6;
    所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”;
    所述5-10元杂芳基和被取代基取代的5-10元杂芳基中的5-10元杂芳基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的5-10元杂芳基”;
    所述5-6元杂芳基为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的5-6元杂芳基”;
    所述的氨基保护基团为叔丁氧羰基、苄氧羰基、芴甲氧羰基、烯丙氧羰基、邻苯二甲酰基、苄基、对甲氧基苄基、三苯甲基或对甲苯磺酰基;
    当*的碳原子具有手性时,如式I所示的稠环化合物为 或其混合物。
  2. 如权利要求1所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,其特征在于,当X为连接键时,R4为C1-6烷基、C1-6烷基-O-、氰基、卤素、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同。
  3. 如权利要求1所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,其特征在于,
    X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;当X为连接键时,R4为氰基、
  4. 如权利要求1所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,其特征在于,
    R6为氧代;L为连接键、-C(=O)-、-C(=O)O-、-C(=O)NR8-、-NR8-、-O-、-S-、-S(O)-、-S(O)2-或-(CH2)p-;当L为连接键时,表示R3直接与D环连接;R3为被一个或多个R3e取代的3-7元杂环基;当取代基为多个时,相同或不同;每个R3e各自独立地为氘、羟基、卤素、-N(R7)2、氰基、硝基、C1- 6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C2- 6烯基、C2-6炔基、被一个或多个R3-c取代的C2-6烯基、被一个或多个R3-d取代的C2-6炔基、C3-7环烷基、被一个或多个R3-e取代的C3-7环烷基、3-7元杂环基、被一个或多个R3-f取代的3-7元杂环基、C6-12芳基、被一个或多个R3-g取代的C6-12芳基、5-10元杂芳基、被一个或多个R3-h取代的5-10元杂芳基、C3-7环烯基、被一个或多个R3-i取代的C3-7环烯基、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、-SR9、-C(=O)-O-R9、-O-C(=O)-R9、-C(=O)-N(R7)2、-S(O)2-R9、-NR8-S(O)2-R9、-NR8-S(O)2-N(R7)2、-S(O)2-N(R7)2、-S(O)-N(R7)2、-S(O)-R9、-NR8-S(O)-R9、-NR8-S(O)-N(R7)2、-C(=O)-R9、-NR8C(=O)-R9或-NR8C(=O)-O-R9;当取代基为多个时,相同或不同;
    进一步地,
    R6为氧代;L为连接键;当L为连接键时,表示R3直接与D环连接;R3为被一个或多个R3e取代的3-7元杂环基;当取代基为多个时,相同或不同;每个R3e各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、C2-6炔基或-C(=O)-R9;当取代基为多个时,相同或不同。
  5. 如权利要求1-4中任一项所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,其特征在于,其满足以下条件中的一种或多种:
    (1)R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1- g、R1-h、R1-i、R2、R3、R3a、R3b、R3c、R3d、R3e、R3f、R3g、R3h、R3-a、R3-b、R3-c、R3-d、R3-e、R3-f、R3-g、R3-h、R3-i、R3-j、R4、R4a、R4b、R5、R5a、R5b、R5c、R6、R7、R7a、R7b、R8、R9、R10和R11中,所述C1-6烷基、被取代基取代的C1-6烷基中的C1-6烷基各自独立地为甲基、乙基、正丙基、异丙基、 正丁基、异丁基、仲丁基或叔丁基,例如为甲基或乙基;
    (2)R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i、R2、R3a、R3b、R3c、R3d、R3e、R3f、R3g、R3h、R3-a、R3-b、R3-c、R3-d、R3-e、R3-f、R3-g、R3- h、R3-i、R3-j、R4、R4a、R4b、R5、R5a、R5b、R5c、R6、R7a、R7b、R10和R11a中,所述卤素各自独立地为F、Cl、Br或I,例如为F;
    (3)在某一方案中,R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i、R2、R3a、R3b、R3c、R3d、R3e、R3f、R3g、R3h、R3-a、R3-b、R3-c、R3-d、R3- e、R3-f、R3-g、R3-h、R3-i、R3-j、R4、R5、R6、R7a、R7b和R10中,所述C1-6烷基-O-、被取代基取代的C1-6烷基-O-中的C1-6烷基-O-各自独立地为甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基、仲丁氧基或叔丁氧基,例如为甲氧基;
    (4)R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i、R2、R3、R3a、R3b、R3c、R3d、R3e、R3f、R3g、R3h、R5、R6、R7a、R7b、R8、R9和R10中,所述C2-6烯基、被取代基取代的C2-6烯基中的C2-6烯基各自独立地为-CH=CH2、-CH=CH-CH3、-CH2-CH=CH2、-C(CH3)=CH2、-CH=CH-CH2-CH3、-CH2-CH=CH-CH3、-CH=CH-CH2-CH2-CH3、-CH2-CH=CH-CH2-CH3、-CH=CH-CH(CH3)2、-CH2-CH=C(CH3)2或-CH=CH-(CH2)3-CH3
    (5)R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i、R2、R3、R3a、R3b、R3c、R3d、R3e、R3f、R3g、R3h、R5、R6、R7a、R7b、R8、R9和R10中,所述C2-6炔基、被取代基取代的C2-6炔基中的C2-6炔基各自独立地为-C≡CH、-C≡C-CH3、-C≡C-CH2-CH3、-CH2C≡C-CH3、-C≡C-CH2-CH2-CH3、-CH2C≡C-CH2-CH3、-C≡C-CH2-CH2-CH2-CH3、-C≡C-CH2-CH(CH3)2、-C≡C-C(CH3)3、-C≡C-CH(CH3)2或-CH2C≡C-CH2-CH2-CH3
    (6)环A中的C3-7环烷基,R1中的C3-7环烷基、被取代基取代的C3-7环烷基中的C3-7环烷基,两个R1与所相连的环原子一起形成C3-7环烷基、两个R1与所相连的环原子一起形成被取代基取代的C3-7环烷基中的C3-7环烷基,R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h和R1i中的C3-7环烷基、被取代基取代的C3-7环烷基中的C3-7环烷基、C3-7环烷基-O-中的C3-7环烷基、被取代基取代的C3-7环烷基-O-中的C3-7环烷基,环D、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i和R2中的C3-7环烷基,R3中的C3-7环烷基、被取代基取代的C3-7环烷基中的C3-7环烷基,R3a、R3b、R3c、R3d、R3e、R3f、R3g和R3h中的C3-7环烷基、被取代基取代的C3-7环烷基中的C3-7环烷基、C3-7环烷基-O-中的C3-7环烷基、被取代基取代的C3-7环烷基-O-中的C3-7环烷基,两个R3d、两个R3e、两个R3f、两个R3g或两个R3h与所相连的环原子一起形成C3-7环烷基,两个R3d、两个R3e、两个R3f、两个R3g或两个R3h与所相连的环原子一起形成被取代基取代的C3-7环烷基中的C3-7环烷基,R3-a、R3-b、R3-c、R3-d、R3-e、R3-f、R3- g、R3-h、R3-i和R3-j中的C3-7环烷基,R4中的C3-7环烷基、被取代基取代的C3-7环烷基中的C3-7环烷基,R5中的C3-7环烷基、被取代基取代的C3-7环烷基中的C3-7环烷基,R7中的C3-7环烷基,R8和R9中的C3-7环烷基,各自独立地为环丙基、环丁基、环戊基、环己基、环庚基或例如为环丙基;
    (7)环A中的C3-7环烯基,R1中的C3-7环烯基、被取代基取代的C3-7环烯基中的C3-7环烯基、两个R1与所相连的环原子一起形成C3-7环烯基、两个R1与所相连的环原子一起形成被取代基取代的C3-7环烯基中的C3-7环烯基,R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h和R1i中的C3-7环烯基、被取代基取代的C3-7环烯基中的C3-7环烯基、C3-7环烯基-O-中的C3-7环烯基、被取代基取代的C3-7环烯基-O-中的C3-7环烯基,环D、R1-a、R1-b、R1-c、R1-d、R1-e、R1-f、R1-g、R1-h、R1-i和R2中的C3-7环烯基,R3中的C3-7环烯基、被取代基取代的C3-7环烯基中的C3-7环烯基,R3a、R3b、R3c、R3d、R3e、R3f、R3g和R3h中的C3-7环烯基、被取代基取代的C3-7环烯基中的C3-7环烯基,两个R3d、两个R3e、两个R3f、两个R3g或两个R3h与所相连的环原子一起形成C3-7环烯基,两个R3d、两个R3e、两个R3f、两个R3g或两个R3h与所相连的环原子一起形成被取代基取代的C3-7环烯基中的C3-7环烯基,R3-a、R3-b、R3-c、R3-d、R3-e、R3-f、R3-g、R3-h、R3-i和R3-j中的C3-7环烯基,R5中的C3-7环烯基、被取代基取代的C3-7环烯基中的C3-7环烯基,R7、R8和R9中的C3-7环烯基,所述C3-7环烯基各自独立地为环丙烯基、环丁烯基、环戊烯基、环己烯基或环庚烯基;
    (8)环A、环D、R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R1-a、R1-b、R1-c、R1-d、R1- e、R1-f、R1-g、R1-h、R1-i、R2、R3、R3a、R3b、R3c、R3d、R3e、R3f、R3g、R3h、R3-a、R3-b、R3-c、R3-d、R3-e、R3-f、R3-g、R3-h、R3-i、R3-j、R4、R5、R7、R8和R9中,所述3-7元杂环基、被取代基取代的3-7元杂环基中的3-7元杂环基、3-7元杂环基-O-中的3-7元杂环基、被取代基取代的3-7元杂环基-O-中的3-7元杂环基各自独立地为饱和或部分不饱和的非芳香性的单环或多环杂环基,具体地,单环杂环基可为吡咯烷基、四氢吡喃基、哌啶基、吗啉基、硫代吗啉基、高哌嗪基、哌嗪基、氮杂环丁基、1,2-二氢吡啶基、四氢吡啶基等,多环可为桥环、稠环或螺环;
    (9)R3中,所述3-7元杂环基、被一个或多个R3e取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N和O中的一种或两种,杂原子个数为1个或2个的3-7元杂环基”;进一步地,所述3-7元杂环基、被一个或多个R3e取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子个数为1个或2个,杂原子种类独立选自N和O”的4-7元杂环基;所述的4-7元杂环基可为4-7元单环杂环基、5-7元桥环杂环基或5-7元螺环杂环基;具体地,当R3为4-7元单环杂环基时,所述4-7元单环杂环基可为哌啶基、四氢吡喃基、哌嗪基、氮杂环丁基、吗啉基或吡咯烷基;当R3为5-7元桥环杂环基时,所述5-7元桥环杂环基可为当R3为5-7元螺环杂环基时,所述5-7元螺环杂环基可为例如为
    (10)R4中,所述3-7元杂环基、被一个或多个R4a取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”;进一步地,所述3-7元杂环基、被一个或多个R4a取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N和O中的一种或两种,杂原子个数为1个或2个的4-7元杂环基”;例如氮杂环丁基、吡咯烷基、哌啶基、哌嗪基、四氢吡啶基或吗啉基;又例如 再例如
    (11)R7中,所述两个R7与所相连的氮原子一起形成3-7元杂环基、被一个或多个R7a取代的3-7元杂环基中的3-7元杂环基的杂原子个数独立为1个或2个;所述两个R7与所相连的氮原子一起形成3-7元杂环基、被一个或多个R7a取代的3-7元杂环基中的3-7元杂环基的杂原子种类独立选自N和O中的一种或两种;所述两个R7与所相连的氮原子一起形成3-7元杂环基、被一个或多个R7a取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N和O中的一种或两种,杂原子个数为1个或2个的4-7元杂环基”;例如:吡咯烷基,又例如
    (12)环D中,所述3-7元杂环基为“杂原子种类为N,杂原子个数为1个或2个的6元杂环基”,例如为1,2-二氢吡啶基,又例如为“c”表示该原子位于B环N原子的间位;
    (13)环A、环D、R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R3、R3a、R3b、R3c、R3d、R3e、R3f、R3g和R3h中,所述C6-12芳基、被取代基取代的C6-12芳基中的C6-12芳基各自独立地为苯基或萘基;例如:苯基;
    (14)环A、R1、R1a、R1b、R1c、R1d、R1e、R1f、R1g、R1h、R1i、R3、R3a、R3b、R3c、R3d、R3e、R3f、R3g和R3h中,所述5-10元杂芳基、被取代基取代的5-10元杂芳基中的5-10元杂芳基各自独立地为“杂原子个数为1个、2个或3个,杂原子种类独立选自N、O和S的5-8元杂芳基”;
    (15)X中,所述-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-中的-(C1-C4亚烷基)-各自独立地为亚甲基、亚乙基、亚正丙基或亚异丙基,例如:-CH2-、-CH2CH2-、-CH(CH3)-、-CH2CH2CH2-、-CH(CH3)CH2-、-CH(CH2CH3)-或-C(CH3)2-,优选-CH2-、-CH2CH2-、-CH(CH3)-或-CH2CH2CH2-;进一步地,“-O-(C1-C4亚烷基)-”中“C1-C4亚烷基”与R4相连;
    (16)X中,所述-(C2-C4亚烯基)-独立地为亚乙烯基、亚丙烯基、亚烯丙基或亚丁烯基,例如-CH=CH-、-CH=CH-CH2-;进一步地,所述-(C2-C4亚烯基)-可为 其中f端与R4相连;和
    (17)X中,所述-(C2-C6亚炔基)-独立地为亚乙炔基、亚丙炔基、亚炔丙基、亚丁炔基或亚戊炔基,例如-C≡C-、-CH2-C≡CH、-C≡C-CH2-、-C≡C-CH2-CH2-、-C≡C-CH(CH3)-CH2-、-C≡C-C(CH3)2-,优选为-C≡C-、-C≡C-CH2-或-C≡C-C(CH3)2-;进一步地,所述-(C2-C6亚炔基)-可为-C≡C-*、-C≡C-CH2-*或-C≡C-C(CH3)2-*,其中*端与R4相连。
  6. 如权利要求1-4中任一项所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,其特征在于,其满足以下条件中的一种或多种:
    (1)R3中,所述3-7元杂环基、被一个或多个R3e取代的3-7元杂环基中的3-7元杂环基的杂原子个数独立地为1个或2个;
    (2)R3中,所述3-7元杂环基、被一个或多个R3e取代的3-7元杂环基中的3-7元杂环基的杂原子种类独立选自N和O中的一种或两种;
    (3)R3中,所述3-7元杂环基、被一个或多个R3e取代的3-7元杂环基中的3-7元杂环基各自独立地为4-7元的杂环基;
    (4)R4中,所述3-7元杂环基、被一个或多个R4a取代的3-7元杂环基中的3-7元杂环基的杂原子个数独立地为1个、2个或3个;进一步地,所述3-7元杂环基、被一个或多个R4a取代的3-7元杂环基中的3-7元杂环基的杂原子个数独立地为1个或2个;
    (5)R4中,所述3-7元杂环基、被一个或多个R4a取代的3-7元杂环基中的3-7元杂环基的杂原子种类独立选自N、O和S中的一种或多种;进一步地,所述3-7元杂环基、被一个或多个R4a取代的3-7元杂环基中的3-7元杂环基的杂原子种类独立选自N和O中的一种或两种;
    (6)R4中,所述3-7元杂环基、被一个或多个R4a取代的3-7元杂环基中的3-7元杂环基各自独立地为4-7元的杂环基;
    (7)R7中,所述两个R7与所相连的氮原子一起形成3-7元杂环基、被一个或多个R7a取代的3-7元杂环基中的3-7元杂环基的杂原子个数独立为1个或2个;
    (8)R7中,所述两个R7与所相连的氮原子一起形成3-7元杂环基、被一个或多个R7a取代的3-7元杂环基中的3-7元杂环基的杂原子种类独立选自N和O中的一种或两种;
    (9)R7中,所述两个R7与所相连的氮原子一起形成3-7元杂环基、被一个或多个R7a取代的3- 7元杂环基中的3-7元杂环基各自独立地为4-7元杂环基;
    (10)环D中,所述3-7元杂环基的杂原子种类为N;
    (11)环D中,所述3-7元杂环基的杂原子个数为1个或2个;和
    (12)环D中,所述3-7元杂环基为6元杂环基。
  7. 如权利要求1-4中任一项所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,其特征在于,其满足以下条件中的一种或多种:
    (1)环A为C6-12芳基;优选地,环A为苯基;
    (2)每个R1独立地为卤素、羟基、-N(R7)2、C1-6烷基、氰基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1a取代的C1-6烷基或被一个或多个R1b取代的C1-6烷基-O-;优选地,每个R1独立地为卤素、C1-6烷基、氰基或被一个或多个R1a取代的C1-6烷基;当取代基为多个时,相同或不同;
    (3)每个R1a和R1b各自独立地为卤素;优选地,每个R1a和R1b各自独立地为F或Cl;
    (4)m为0、1、2或3;优选地,m为2;
    (5)R2为氢、羟基、卤素、氨基、氰基、C1-6烷基或C1-6烷基-O-;优选地,R2为氢;
    (6)L为连接键;
    (7)R3为C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、3-7元杂环基、被一个或多个R3e取代的3-7元杂环基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;优选地,R3为C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、被一个或多个R3e取代的3-7元杂环基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;更优选地,R3为被一个或多个R3e取代的3-7元杂环基;当取代基为多个时,相同或不同;
    (8)每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3- a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、C2-6炔基或-C(=O)-R9;进一步地,每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C2-6炔基或-C(=O)-R9;较佳地,每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-或-C(=O)-R9;优选地,每个R3d、R3e和R3h各自独立地为羟基、-N(R7)2、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-或-C(=O)-R9;进一步优选地,每个R3d、R3e和R3h各自独立地为羟基、卤素、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-或-C(=O)-R9;更优选地,每个R3d、R3e和R3h各自独立地为羟基、卤素、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-或-C(=O)-R9;当取代基为多个时,相同或不同;
    (9)每个R3-a、R3-b和R3-j各自独立地为氘、羟基、卤素或C3-7环烷基;进一步地,每个R3-a和R3-b各自独立地为氘、羟基、卤素或C3-7环烷基;更进一步地,每个R3-b独立地为氘、羟基或卤素;
    (10)X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;进一步地,X为连接键、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;优选地,X为-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6 亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;
    (11)R4为氢、C1-6烷基、C1-6烷基-O-、氰基、卤素、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;优选地,R4为氢、C1-6烷基、氰基、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基或被一个或多个R4b取代的C3-7环烷基;更优选地,R4为氢、C1-6烷基、氰基、羟基、-N(R7)2、被一个或多个R4a取代的3-7元杂环基或被一个或多个R4b取代的C3-7环烷基;进一步优选地,R4为-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;最优选地,R4为-N(R7)2、3-7元杂环基或被一个或多个R4a取代的3-7元杂环基;当取代基为多个时,相同或不同;
    (12)每个R4a和R4b各自独立地为羟基、卤素、氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;优选地,每个R4a和R4b各自独立地为氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;更优选地,每个R4a和R4b各自独立地为C1-6烷基或氨基;进一步优选地,每个R4a独立地为C1-6烷基;
    (13)R5为氢、羟基、卤素、氨基、氰基或C1-6烷基;优选地,R5为氢或C1-6烷基;更优选地,R5为C1-6烷基;
    (14)R6为氧代;
    (15)每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基或被一个或多个R7a取代的3-7元杂环基;进一步地,每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基;更进一步地,每个R7独立地为氢或C1-6烷基;当取代基为多个时,相同或不同;
    (16)每个R7b独立地为氘、卤素、羟基或氰基;优选地,每个R7b独立地为氘或羟基;更优选地,每个R7b独立地为氘;
    (17)每个R9独立地为氢或C1-6烷基;优选地,每个R9独立地为C1-6烷基;
    (18)环D为3-7元杂环基;
    (19)R10为C1-6烷基;优选地,R10为甲基;
    (20)R11为C1-6烷基;优选地,R11为甲基;和
    (21)不为氢。
  8. 如权利要求1-7中任一项所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,其特征在于,
    环A为C6-12芳基;
    每个R1独立地为卤素、羟基、-N(R7)2、C1-6烷基、氰基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1a取代的C1-6烷基或被一个或多个R1b取代的C1-6烷基-O-;当取代基为多个时,相同或不同;
    每个R1a和R1b各自独立地为卤素;
    m为0、1、2或3;
    R2为氢、羟基、卤素、氨基、氰基、C1-6烷基或C1-6烷基-O-;
    L为连接键;
    R3为C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、3-7元杂环基、被一个或多个R3e取代的3-7元杂环基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;当取代基为多个时,相同或不同;
    每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、C2-6炔基或-C(=O)-R9;当取代基为多个时,相同或不同;
    每个R3-a、R3-b和R3-j各自独立地为氘、羟基、卤素或C3-7环烷基;
    X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C4亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;
    R4为氢、C1-6烷基、C1-6烷基-O-、氰基、卤素、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
    每个R4a和R4b各自独立地为羟基、卤素、氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;
    R5为氢、羟基、卤素、氨基、氰基或C1-6烷基;
    R6为氧代;
    每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基或被一个或多个R7a取代的3-7元杂环基;当取代基为多个时,相同或不同;
    每个R7a和R7b各自独立地为氘、卤素、羟基或氰基;
    每个R9独立地为氢或C1-6烷基;
    环D为3-7元杂环基;
    R10为C1-6烷基;
    R11为C1-6烷基;
    所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”;
    进一步地,
    环A为C6-12芳基;
    每个R1独立地为卤素、氰基、C1-6烷基或被一个或多个R1a取代的C1-6烷基;当取代基为多个时,相同或不同;
    每个R1a独立地为卤素;
    m为2;
    R2为氢;
    L为连接键;
    R3为被一个或多个R3e取代的3-7元杂环基;当取代基为多个时,相同或不同;
    每个R3e独立地为羟基、卤素、-N(R7)2、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3- 7环烷基-O-或-C(=O)-R9;当取代基为多个时,相同或不同;
    每个R3-b独立地为氘、羟基或卤素;
    X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,“-O-(C1-C4亚烷基)-”中“C1-C4亚烷基”与R4相连;
    R4为氢、C1-6烷基、氰基、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
    每个R4a和R4b各自独立地为氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;
    R5为氢或C1-6烷基;
    R6为氧代;
    每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基;当取代基为多个时,相同或不同;
    每个R7b独立地为氘或羟基;
    每个R9独立地为C1-6烷基;
    环D为3-7元杂环基;
    R10为C1-6烷基;
    R11为C1-6烷基;
    所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”;
    更进一步,
    环A为C6-12芳基(例如,);
    每个R1独立地为卤素、氰基、C1-6烷基或被一个或多个R1a取代的C1-6烷基;当取代基为多个时,相同或不同;
    每个R1a独立地为卤素;
    m为2;
    R2为氢;
    L为连接键;
    R3为被一个或多个R3e取代的3-7元杂环基;当取代基为多个时,相同或不同;
    每个R3e独立地为羟基、-N(R7)2、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-或-C(=O)-R9;当取代基为多个时,相同或不同;
    每个R3-b独立地为氘、羟基或卤素;
    X为-(C1-C4亚烷基)-、-O-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,“-O-(C1-C4亚烷基)-”中“C1-C4亚烷基”与R4相连;
    R4为-N(R7)2、3-7元杂环基或被一个或多个R4a取代的3-7元杂环基;当取代基为多个时,相同或不同;
    每个R4a独立地为C1-6烷基;
    R5为氢或C1-6烷基;
    R6为氧代;
    每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基;当取代基为多个时,相同或不同;
    每个R7b独立地为氘或羟基;
    每个R9独立地为C1-6烷基;
    环D为3-7元杂环基;
    R10为甲基;
    R11为甲基;
    所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”。
  9. 如权利要求1-7中任一项所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,其特征在于,所述的如式I所示的稠环化合物为如下方案1、方案2、方案3或方案4:
    方案1:所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中,
    环A为C6-12芳基;
    每个R1独立地为卤素、羟基、-N(R7)2、C1-6烷基、氰基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1a取代的C1-6烷基或被一个或多个R1b取代的C1-6烷基-O-;当取代基为多个时,相同或不同;
    每个R1a和R1b各自独立地为卤素;
    m为0、1、2或3;
    R2为氢、羟基、卤素、氨基、氰基、C1-6烷基或C1-6烷基-O-;
    L为连接键;
    R3为C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、3-7元杂环基、被一个或多个R3e取代的3-7元杂环基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;当取代基为多个时,相同或不同;
    每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、C2-6炔基或-C(=O)-R9;当取代基为多个时,相同或不同;
    每个R3-a、R3-b和R3-j各自独立地为氘、羟基、卤素或C3-7环烷基;
    X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;
    R4为氢、C1-6烷基、C1-6烷基-O-、氰基、卤素、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
    每个R4a和R4b各自独立地为羟基、卤素、氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;
    R5为氢、羟基、卤素、氨基、氰基或C1-6烷基;
    R6为氧代(=O);
    每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基或被一个或多个R7a取代的3-7元杂环基;当取代基为多个时,相同或不同;
    每个R7a和R7b独立地为氘、卤素、羟基或氰基;
    每个R9独立地为氢或C1-6烷基;
    环D为3-7元杂环基;
    R10为C1-6烷基;
    R11为C1-6烷基;
    其中,不为氢;
    所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”;
    方案2:所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中,
    环A为C6-12芳基;
    每个R1独立地为卤素、氰基、C1-6烷基或被一个或多个R1a取代的C1-6烷基;当取代基为多个时,相同或不同;
    每个R1a独立地为卤素;
    m为2;
    R2为氢;
    L为连接键;
    R3为被一个或多个R3e取代的3-7元杂环基;当取代基为多个时,相同或不同;
    每个R3e独立地为羟基、卤素、-N(R7)2、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3- 7环烷基-O-或-C(=O)-R9;当取代基为多个时,相同或不同;
    每个R3-b独立地为氘、羟基或卤素;
    X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,“-O-(C1-C4亚烷基)-”中“C1-C4亚烷基”与R4相连;
    R4为氢、C1-6烷基、氰基、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
    每个R4a和R4b各自独立地为氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;
    R5为氢或C1-6烷基;
    R6为氧代(=O);
    每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基;当取代基为多个时,相同或不同;
    每个R7b独立地为氘或羟基;
    每个R9独立地为C1-6烷基;
    环D为3-7元杂环基;
    R10为C1-6烷基;
    R11为C1-6烷基;
    其中,不为氢;
    所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”;
    方案3:所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中,
    环A为C6-12芳基;
    每个R1独立地为卤素、羟基、-N(R7)2、C1-6烷基、氰基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1a取代的C1-6烷基或被一个或多个R1b取代的C1-6烷基-O-;当取代基为多个时,相同或不同;
    每个R1a和R1b各自独立地为卤素;
    m为0、1、2或3;
    R2为氢、羟基、卤素、氨基、氰基、C1-6烷基或C1-6烷基-O-;
    L为连接键;
    R3为C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、被一个或多个R3e取代的3-7元杂环基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;当取代基为多个时,相同或不同;
    每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、C2-6炔基或-C(=O)-R9;当取代基为多个时,相同或不同;
    每个R3-a、R3-b和R3-j各自独立地为氘、羟基、卤素或C3-7环烷基;
    X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;
    R4为氢、C1-6烷基、C1-6烷基-O-、氰基、卤素、羟基、-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
    每个R4a和R4b各自独立地为羟基、卤素、氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;
    R5为氢、羟基、卤素、氨基、氰基或C1-6烷基;
    R6为氧代(=O);
    每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基或被一个或多个R7a取代的3-7元杂环基;当取代基为多个时,相同或不同;
    每个R7a和R7b独立地为氘、卤素、羟基或氰基;
    每个R9独立地为氢或C1-6烷基;
    环D为3-7元杂环基;
    R10为C1-6烷基;
    R11为C1-6烷基;
    其中,不为氢;
    所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”;
    方案4:所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐中,
    环A为C6-12芳基;
    每个R1独立地为卤素、羟基、-N(R7)2、C1-6烷基、氰基、C1-6烷基-O-、C2-6烯基、C2-6炔基、被一个或多个R1a取代的C1-6烷基或被一个或多个R1b取代的C1-6烷基-O-;当取代基为多个时,相同或不同;
    每个R1a和R1b各自独立地为卤素;
    m为0、1、2或3;
    R2为氢、羟基、卤素、氨基、氰基、C1-6烷基或C1-6烷基-O-;
    L为连接键;
    R3为C3-7环烷基、被一个或多个R3d取代的C3-7环烷基、3-7元杂环基、被一个或多个R3e取代的3-7元杂环基、C3-7环烯基或被一个或多个R3h取代的C3-7环烯基;当取代基为多个时,相同或不同;
    每个R3d、R3e和R3h各自独立地为羟基、卤素、-N(R7)2、氰基、C1-6烷基、被一个或多个R3-a取代的C1-6烷基、C1-6烷基-O-、被一个或多个R3-b取代的C1-6烷基-O-、C3-7环烷基-O-、被一个或多个R3-j取代的C3-7环烷基-O-、C2-6炔基或-C(=O)-R9;当取代基为多个时,相同或不同;
    每个R3-a、R3-b和R3-j各自独立地为氘、羟基、卤素或C3-7环烷基;
    X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-;较佳地,-O-(C1-C4亚烷基)-中“C1-C4亚烷基”与R4相连;
    R4为-N(R7)2、3-7元杂环基、被一个或多个R4a取代的3-7元杂环基、C3-7环烷基或被一个或多个R4b取代的C3-7环烷基;当取代基为多个时,相同或不同;
    每个R4a和R4b各自独立地为羟基、卤素、氨基、C1-6烷基或被一个或多个氘取代的C1-6烷基;
    R5为氢、羟基、卤素、氨基、氰基或C1-6烷基;
    R6为氧代(=O);
    每个R7独立地为氢、C1-6烷基或被一个或多个R7b取代的C1-6烷基,或两个R7与所相连的氮原子一起形成3-7元杂环基或被一个或多个R7a取代的3-7元杂环基;当取代基为多个时,相同或不同;
    每个R7a和R7b独立地为氘、卤素、羟基或氰基;
    每个R9独立地为氢或C1-6烷基;
    环D为3-7元杂环基;
    R10为C1-6烷基;
    R11为C1-6烷基;
    所述3-7元杂环基和被取代基取代的3-7元杂环基中的3-7元杂环基各自独立地为“杂原子种类独 立选自N、O和S,杂原子个数为1个、2个或3个的3-7元杂环基”。
  10. 如权利要求1-7中任一项所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,其特征在于,其满足以下条件中的一种或多种:
    (1)环A为
    (2)每个R1独立地为卤素、氰基、C1-6烷基或被一个或多个R1a取代的C1-6烷基,当取代基为多个时,相同或不同;例如:R1为F、Cl、-CHF2、-CF3、氰基、-CH3或-CF2CH3,优选为F、-CHF2、-CH3或氰基;
    (3)R2为氢;
    (4)R5为氢或C1-6烷基,例如:R5为氢或甲基;
    (5)X为连接键、-O-、-(C1-C4亚烷基)-、-O-(C1-C4亚烷基)-、-(C2-C4亚烯基)-或-(C2-C6亚炔基)-,“-O-(C1-C4亚烷基)-”中“C1-C4亚烷基”与R4相连;例如:连接键、 又例如:连接键、 其中,f端与R4相连;
    (6)当环D为3-7元杂环基时,环D为1,2-二氢吡啶基,例如为“c”表示该原子位于B环N原子的间位;
    (7)当R3为C3-7环烷基或被一个或多个R3d取代的C3-7环烷基时,所述的R3
    (8)当R3为3-7元杂环基或被一个或多个R3e取代的3-7元杂环基时,所述的R3 又为 例如 又例如 再例如 优选
    (9)R4为氢、甲基、甲氧基、氰基、卤素、羟基、-NH2、-NHCH3、-N(CH3)2、-N(CD3)2、-N(CH2CH3)2 又为氢、甲基、甲氧基、氰基、卤素、羟基、-NH2、-NHCH3、-N(CH3)2
    (10)R10为甲基;
    (11)R11为甲基;
    (12)当R4为氰基时,
    较佳地,所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐满足以下条件中的一种或多种:
    (1A)又可为 还可为 例如又例如
    (1B) 又为 例如 又例如 再例如优选
    (1C)“c”表示该原子位于B环N原子的间位;
    (1D)为甲基、甲氧基、氰基、卤素、 又可为氢、甲基、甲氧基、氰基、卤素、 例如甲基、氰基、 优选 还优选
    更佳地,所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐满足以下条件:
    例如:
    具体地,所述如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐为
  11. 如权利要求1-10中任一项所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,其特征在于,所述的如式I所示的稠环化合物选自以下任一化合物:









  12. 一种如权利要求1-11中任一项所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐的制备方法,其特征在于,其中,当“c”表示该原子位于B环N原子的间位时,其制备方法包括以下步骤:中间体I-A1与原料N-苯基双(三氟甲烷磺酰)亚胺或三氟甲磺酸酐反应得到中间体I-A2,I-A2再与原料I-A3发生偶联反应,得到中间体I-A4,I-A4再与原料I-A5发生取代反应或钯催化的Buchwald偶联反应,得到通式I-A化合物;
    若I-A4与I-A5发生取代反应,其是在碱作用下进行,所述碱为碳酸铯、碳酸钠、碳酸钾、三乙胺或二异丙基乙胺;
    若I-A4与I-A5发生Buchwald偶联反应,其是在催化剂、碱存在下进行,所述催化剂为RuPhos Pd G3、BrettPhos Pd G3、XPhos Pd G3、XantPhos Pd G3、RuPhos Pd G4或BrettPhos Pd G4,所述碱为碳酸铯、碳酸钠、磷酸钾、碳酸钠、叔丁醇钾或叔丁醇钠;
    其中:环A、m、X、L、R1、R2、R3、R4、R5、R10和R11的定义如权利要求1-11中任一项所述;
    RG为氢、-B(OH)2-ZnBr或-ZnI。
  13. 一种如式I-A1所示的化合物、如式I-A2所示的化合物,其特征在于:
    其中:L、R2、R3、R5和R10的定义如权利要求1-11中任一项所述。
  14. 如权利要求13所述的化合物,其特征在于:
    (1)所述如式I-A1所示的化合物为如下任一化合物:
    (2)所述如式I-A2所示的化合物为如下任一化合物:
  15. 如下任一化合物:


  16. 一种药物组合物,其特征在于,其包含:
    (1)物质Q,所述物质Q为如权利要求1-11中任一项所述的式I所示的稠环化合物、其立体异构体或其药学上可接受的盐,及
    (2)药学可接受的辅料。
  17. 一种物质Q或如权利要求16所述的药物组合物在制备SOS1抑制剂中的应用,其特征在于, 所述物质Q为如权利要求1-11中任一项所述的式I所示的稠环化合物、其立体异构体或其药学上可接受的盐。
  18. 一种物质Q或如权利要求16所述的药物组合物在制备药物中的应用;其特征在于,所述物质Q为如权利要求1-11中任一项所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐;所述药物可为治疗和/或预防与SOS1活性或表达量相关的疾病的药物;
    较佳地,所述与SOS1活性或表达量相关的疾病选自非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、结肠癌、甲状腺癌、黑色素瘤、胚胎性横纹肌肉瘤、皮肤颗粒细胞肿瘤、肝癌、直肠癌、膀胱癌、咽喉癌、乳腺癌、前列腺癌、神经胶质细胞瘤、卵巢癌、头颈部鳞癌、宫颈癌、食管癌、肾癌、皮肤癌、淋巴瘤、胃癌、胆管癌、子宫癌、子宫内膜癌、尿路上皮癌、急性髓系白血病、骨髓纤维化、B细胞淋巴瘤、单核细胞白血病、脾大性红细胞增多、嗜酸性白细胞增多综合征和多发性骨髓癌,以及和SOS1遗传性突变相关的疾病,如一型神经纤维瘤、努南氏综合征、多发雀斑样恁型努南氏综合征、毛细血管畸形—动静脉畸形综合征、心-面-皮肤综合征、克斯提洛氏综合征、雷吉士综合征和一型遗传性牙龈纤维瘤。
  19. 一种物质Q或如权利要求16所述的药物组合物在制备药物中的应用;其特征在于,所述物质Q为如权利要求1-11中任一项所述的如式I所示的稠环化合物、其立体异构体或其药学上可接受的盐;所述药物为治疗和/或预防以下各类疾病的药物;所述疾病选自非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、结肠癌、甲状腺癌、黑色素瘤、胚胎性横纹肌肉瘤、皮肤颗粒细胞肿瘤、肝癌、直肠癌、膀胱癌、咽喉癌、乳腺癌、前列腺癌、神经胶质细胞瘤、卵巢癌、头颈部鳞癌、宫颈癌、食管癌、肾癌、皮肤癌、淋巴瘤、胃癌、胆管癌、子宫癌、子宫内膜癌、尿路上皮癌、急性髓系白血病、骨髓纤维化、B细胞淋巴瘤、单核细胞白血病、脾大性红细胞增多、嗜酸性白细胞增多综合征和多发性骨髓癌,以及一型神经纤维瘤、努南氏综合征、多发雀斑样恁型努南氏综合征、毛细血管畸形—动静脉畸形综合征、心-面-皮肤综合征、克斯提洛氏综合征、雷吉士综合征和一型遗传性牙龈纤维瘤。
PCT/CN2023/110759 2022-08-05 2023-08-02 一种稠环化合物、其制备方法及其应用 WO2024027762A1 (zh)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
CN202210936913.9 2022-08-05
CN202210936913 2022-08-05
CN202211294374 2022-10-21
CN202211294374.X 2022-10-21
CN202211529285 2022-11-30
CN202211529285.9 2022-11-30
CN202310913572 2023-07-24
CN202310913572.8 2023-07-24

Publications (1)

Publication Number Publication Date
WO2024027762A1 true WO2024027762A1 (zh) 2024-02-08

Family

ID=89848520

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/110759 WO2024027762A1 (zh) 2022-08-05 2023-08-02 一种稠环化合物、其制备方法及其应用

Country Status (1)

Country Link
WO (1) WO2024027762A1 (zh)

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140323478A1 (en) * 2013-04-30 2014-10-30 Afraxis Holdings, Inc. Serine/threonine kinase inhibitors
CN111372932A (zh) * 2017-12-21 2020-07-03 勃林格殷格翰国际有限公司 作为sos1抑制剂的新颖苄氨基取代吡啶并嘧啶酮及衍生物
CN113200981A (zh) * 2021-02-10 2021-08-03 杭州英创医药科技有限公司 作为sos1抑制剂的杂环化合物
WO2021173524A1 (en) * 2020-02-24 2021-09-02 Mirati Therapeutics, Inc. Sos1 inhibitors
WO2021249519A1 (zh) * 2020-06-11 2021-12-16 江苏恒瑞医药股份有限公司 吡啶酮并嘧啶类衍生物、其制备方法及其在医药上的应用
WO2021249475A1 (zh) * 2020-06-10 2021-12-16 江苏恒瑞医药股份有限公司 稠合喹唑啉类衍生物、其制备方法及其在医药上的应用
CN114075195A (zh) * 2020-08-21 2022-02-22 广东东阳光药业有限公司 嘧啶酮衍生物及其在药物中的应用
CN114456165A (zh) * 2022-02-14 2022-05-10 上海翰森生物医药科技有限公司 含氮并环类衍生物调节剂、其制备方法和应用
CN114685487A (zh) * 2020-12-27 2022-07-01 上海凌达生物医药有限公司 一类嘧啶并杂环类化合物、制备方法和用途
WO2022156792A1 (en) * 2021-01-25 2022-07-28 Guangdong Newopp Biopharmaceuticals Co., Ltd. Heterocyclic compounds as sos1 inhibitors
CN114835703A (zh) * 2021-02-02 2022-08-02 苏州泽璟生物制药股份有限公司 取代嘧啶并吡啶酮类抑制剂及其制备方法和应用
WO2022160931A1 (zh) * 2021-01-28 2022-08-04 浙江海正药业股份有限公司 吡啶并嘧啶类衍生物及其制备方法和用途
WO2022184116A1 (zh) * 2021-03-05 2022-09-09 江苏先声药业有限公司 新型sos1抑制剂及其制备方法和应用
CN115043842A (zh) * 2021-03-09 2022-09-13 苏州泽璟生物制药股份有限公司 胺基取代双环类抑制剂及其制备方法和应用

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140323478A1 (en) * 2013-04-30 2014-10-30 Afraxis Holdings, Inc. Serine/threonine kinase inhibitors
CN111372932A (zh) * 2017-12-21 2020-07-03 勃林格殷格翰国际有限公司 作为sos1抑制剂的新颖苄氨基取代吡啶并嘧啶酮及衍生物
WO2021173524A1 (en) * 2020-02-24 2021-09-02 Mirati Therapeutics, Inc. Sos1 inhibitors
WO2021249475A1 (zh) * 2020-06-10 2021-12-16 江苏恒瑞医药股份有限公司 稠合喹唑啉类衍生物、其制备方法及其在医药上的应用
WO2021249519A1 (zh) * 2020-06-11 2021-12-16 江苏恒瑞医药股份有限公司 吡啶酮并嘧啶类衍生物、其制备方法及其在医药上的应用
CN114075195A (zh) * 2020-08-21 2022-02-22 广东东阳光药业有限公司 嘧啶酮衍生物及其在药物中的应用
CN114685487A (zh) * 2020-12-27 2022-07-01 上海凌达生物医药有限公司 一类嘧啶并杂环类化合物、制备方法和用途
WO2022156792A1 (en) * 2021-01-25 2022-07-28 Guangdong Newopp Biopharmaceuticals Co., Ltd. Heterocyclic compounds as sos1 inhibitors
WO2022160931A1 (zh) * 2021-01-28 2022-08-04 浙江海正药业股份有限公司 吡啶并嘧啶类衍生物及其制备方法和用途
CN114835703A (zh) * 2021-02-02 2022-08-02 苏州泽璟生物制药股份有限公司 取代嘧啶并吡啶酮类抑制剂及其制备方法和应用
CN113200981A (zh) * 2021-02-10 2021-08-03 杭州英创医药科技有限公司 作为sos1抑制剂的杂环化合物
WO2022184116A1 (zh) * 2021-03-05 2022-09-09 江苏先声药业有限公司 新型sos1抑制剂及其制备方法和应用
CN115043842A (zh) * 2021-03-09 2022-09-13 苏州泽璟生物制药股份有限公司 胺基取代双环类抑制剂及其制备方法和应用
CN114456165A (zh) * 2022-02-14 2022-05-10 上海翰森生物医药科技有限公司 含氮并环类衍生物调节剂、其制备方法和应用

Similar Documents

Publication Publication Date Title
WO2021203768A1 (zh) 嘧啶并二环类衍生物、其制备方法及其在医药上的应用
WO2021068898A1 (zh) 新颖的kras g12c蛋白抑制剂及其制备方法和用途
CN111018861B (zh) 吡咯并嘧啶化合物
CN111393434B (zh) 取代的4-苯基哌啶及其制备和用途
TWI596095B (zh) Dihydropyridazine-3,5-dione derivatives
WO2021129653A1 (zh) 一种蛋白降解剂化合物的制备方法和应用
TW201522342A (zh) 用於治療寄生蟲疾病之化合物及組合物
WO2019157879A1 (zh) 作为trk抑制剂的杂环化合物
TWI810550B (zh) 3-氮雜二環烷基衍生物及包含其之醫藥組成物
WO2023274324A1 (zh) 一种含氮杂环化合物、其制备方法、中间体及应用
WO2016050165A1 (zh) 氮杂双环衍生物、其制法与医药上的用途
WO2023174175A1 (zh) Kif18a抑制剂
WO2021249475A1 (zh) 稠合喹唑啉类衍生物、其制备方法及其在医药上的应用
WO2021098811A1 (zh) 吡唑并杂芳基类衍生物、其制备方法及其在医药上的应用
WO2022194221A1 (zh) 呋喃稠环取代的戊二酰亚胺类化合物
WO2023134465A1 (zh) 一种含氮杂环化合物、其制备方法、中间体及应用
WO2023143312A1 (zh) 一种含氮杂环化合物、其制备方法及应用
WO2023041055A1 (zh) Kif18a抑制剂
WO2020020377A1 (zh) 用作fgfr4抑制剂的稠环衍生物
WO2023217230A1 (zh) 驱动蛋白kif18a抑制剂及其应用
WO2020259703A1 (zh) 一种吡唑酮并嘧啶类化合物、其制备方法及应用
WO2024027762A1 (zh) 一种稠环化合物、其制备方法及其应用
TW202333693A (zh) 苯并哌啶衍生物調節劑、其製備方法和應用
WO2022222911A1 (zh) 嘧啶酮化合物及其用途
WO2022171118A1 (zh) 一种具有抗肿瘤活性的化合物及其用途

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23849467

Country of ref document: EP

Kind code of ref document: A1