WO2024027712A1 - 含氮杂环类化合物、其制备方法及其应用 - Google Patents

含氮杂环类化合物、其制备方法及其应用 Download PDF

Info

Publication number
WO2024027712A1
WO2024027712A1 PCT/CN2023/110566 CN2023110566W WO2024027712A1 WO 2024027712 A1 WO2024027712 A1 WO 2024027712A1 CN 2023110566 W CN2023110566 W CN 2023110566W WO 2024027712 A1 WO2024027712 A1 WO 2024027712A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
group
independently
membered
ring
Prior art date
Application number
PCT/CN2023/110566
Other languages
English (en)
French (fr)
Inventor
许大强
郑永勇
党奎峰
薛小超
何旭芝
王桂敏
Original Assignee
索智生物科技(浙江)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 索智生物科技(浙江)有限公司 filed Critical 索智生物科技(浙江)有限公司
Publication of WO2024027712A1 publication Critical patent/WO2024027712A1/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
    • 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
    • A61K31/437Heterocyclic 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 five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D471/12Heterocyclic 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 three hetero rings
    • C07D471/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/056Ortho-condensed systems with two or more oxygen atoms as ring hetero atoms in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • C07D491/14Ortho-condensed systems
    • C07D491/147Ortho-condensed systems the condensed system containing one ring with oxygen as ring hetero atom and two rings with nitrogen as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems

Definitions

  • the present invention relates to nitrogen-containing heterocyclic compounds, their preparation methods and their applications.
  • TLRs Toll-like receptors
  • TLRs Toll like receptors
  • PAMPs pathogen-associated molecular patterns
  • TLR7/8/9 are mainly highly expressed in DC cells and B cells, TLR7/8 mainly recognizes ssRNA, and TLR9 mainly recognizes CpG-DNA.
  • TLR7/8/9 is activated after binding its ligand, and binds to the adapter protein MyD88 in the cytoplasm, activating the NF- ⁇ B and IRF pathways, activating DC cells, and producing type I interferon and various other inflammatory cytokines.
  • B cells TLR7/8/9 combines with nucleic acid substances and plays an important role in the production of antinuclear antibodies by B cells, and the type I interferon secreted by DC cells also promotes this autoimmune B cell Further proliferation and activation of cells cause a series of inflammatory reactions.
  • Lupus erythematosus is a chronic and relapsing autoimmune connective tissue disease that, in addition to causing damage to the patient's skin, can also affect multiple internal organs and connective tissues of the patient. According to statistics from the Lupus Foundation of America, more than 1.5 million people in the United States are suffering from the disease so far, and there are at least more than 5 million patients worldwide.
  • Lupus erythematosus can be divided into systemic lupus erythematosus (SLE), discoid lupus erythematosus, subacute cutaneous lupus erythematosus, and deep lupus erythematosus.
  • SLE systemic lupus erythematosus
  • discoid lupus erythematosus subacute cutaneous lupus erythematosus
  • deep lupus erythematosus Among them, SLE is the most common, accounting for about 70% of all patients. It is also the most severe type of lupus erythematosus. Its clinical manifestations include a series of symptoms such as pain, rash, fatigue, fever, joint swelling, etc. It is easy to relapse and damage various organs. , more than half of SLE patients will suffer permanent organ damage, so SLE is also known as the "immortal cancer”.
  • mice overexpressing TLR7 can exacerbate autoimmune diseases and autoinflammation (Santiago-Raber ML, et al., J Immunol., 2008, 181:1556-1562), and functional inhibition of TLR7/9 can alleviate B6-Fasl p r and BXSB and other pathological manifestations of lupus mice (Dwight H. Kono, et al., PNAS, 2009, 106(29): 12061-12066).
  • TLR8 is related to rheumatoid arthritis (Enevold C., et al., J Rheumatol., 2010, 37, 905-910).
  • TLR9 knockout mice alone showed worsening of disease symptoms, leading to some questions about the relationship of TLR9 signaling to autoimmune diseases.
  • Compounds that modulate TLR7 and/or TLR8 activity, and methods of using these compounds, may therefore provide substantial therapeutic benefit to a variety of autoimmune patients.
  • Patent documents WO2018005586A1, WO2018026620A1, WO2018047081A1, WO2018049089A1, WO2019028301A1, WO2020086503A1, WO2021067326A1, WO2022022489A1 published and reported a series of TLR7/8 inhibitors . As potential therapeutic targets for autoimmune diseases, it is necessary to develop more TLR7/8 inhibitors.
  • the technical problem to be solved by the present invention is that the existing TLR7/8 inhibitors have a single structure. Therefore, the present invention provides a nitrogen-containing heterocyclic compound, its preparation method and its application. This type of compound has good inhibitory activity against TLR7/8.
  • the present invention provides a compound represented by formula II, a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of a pharmaceutically acceptable salt thereof:
  • R 1 is
  • R a is independently C 1 to C 6 alkyl or C 3 to C 6 cycloalkyl
  • R a1 , R a2 , R a3 and R a4 are independently hydrogen, C 1 to C 3 alkyl, halogen or C 1 to C 3 alkyl substituted by 1, 2 or 3 halogens. base;
  • U is independently N or CH
  • Ring A is a 3 to 10-membered heterocarbocyclic ring or a 3 to 10-membered carbocyclic ring;
  • R 21 , R 22 and R 23 are independently hydroxyl, cyano, oxo, halogen or C 1 to C 6 alkyl;
  • R 24 is C 1 to C 6 alkyl or (R 241 )(R 241a )N- , R 241 and R 241a are independently hydrogen or C 1 to C 6 alkyl;
  • R 25 is C 1 to C 6 alkyl;
  • R 26 is independently C 1 to C 6 alkyl;
  • R 271 is independently C 1 to C 6 alkyl;
  • the number of heteroatoms is 1, 2 or 3, and the heteroatoms are selected from one or more of N, O and S;
  • the number of heteroatoms is independently 1, 2 or 3, and the heteroatoms are independently selected from one or more of N, O and S;
  • the number of heteroatoms is independently 1, 2, 3 or 4, and the heteroatoms are independently selected from one or more of N, O and S;
  • R 3 is C 1 to C 6 alkyl or C 3 to C 6 cycloalkyl
  • R A is hydrogen or halogen
  • V is CR 4 or N
  • R 4 is hydrogen, C 1 to C 3 alkyl, C 3 to C 6 cycloalkyl or halogen.
  • the compound represented by Formula II a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of a pharmaceutically acceptable salt thereof:
  • R 1 is
  • R a is independently C 1 to C 6 alkyl or C 3 to C 6 cycloalkyl
  • R a1 , R a2 , R a3 and R a4 are independently hydrogen, C 1 to C 3 alkyl, halogen or C 1 to C 3 alkyl substituted by 1, 2 or 3 halogens;
  • U is independently N or CH
  • Ring A is a 3-10 membered heterocarbon ring
  • R 21 , R 22 and R 23 are independently hydroxyl, cyano, oxo, halogen or C 1 to C 6 alkyl;
  • R 24 is C 1 to C 6 alkyl or (R 241 )(R 241a )N- , R 241 and R 241a are independently hydrogen or C 1 to C 6 alkyl;
  • R 25 is C 1 to C 6 alkyl;
  • R 26 is independently C 1 to C 6 alkyl;
  • R 271 is independently C 1 to C 6 alkyl;
  • the number of heteroatoms is 1, 2 or 3, and the heteroatoms are selected from one or more of N, O and S;
  • the number of heteroatoms is independently 1, 2 or 3, and the heteroatoms are independently selected from one or more of N, O and S;
  • the number of heteroatoms is independently 1, 2, 3 or 4, and the heteroatoms are independently selected from one or more of N, O and S;
  • R 3 is C 1 to C 6 alkyl or C 3 to C 6 cycloalkyl
  • R A is hydrogen or halogen
  • V is CR 4 or N
  • R 4 is hydrogen, C 1 to C 3 alkyl, C 3 to C 6 cycloalkyl or halogen.
  • R 1 is
  • R a is independently C 1 to C 6 alkyl or C 3 to C 6 cycloalkyl
  • R a1 , R a2 , R a3 and R a4 are independently hydrogen, C 1 to C 3 alkyl, halogen or C 1 to C 3 alkyl substituted by 1, 2 or 3 halogens;
  • U is independently N or CH
  • Ring A is a 3-10 membered heterocarbon ring
  • R 21 , R 22 and R 23 are independently hydroxyl, cyano, oxo, halogen or C 1 to C 6 alkyl;
  • R 24 is C 1 to C 6 alkyl or (R 241 )(R 241a )N- , R 241 and R 241a are independently hydrogen or C 1 to C 6 alkyl;
  • R 25 is C 1 to C 6 alkyl;
  • R 26 is independently C 1 to C 6 alkyl;
  • the number of heteroatoms is 1, 2 or 3, and the heteroatoms are selected from one or more types of N, O and S;
  • the number of heteroatoms is independently 1, 2 or 3, and the heteroatoms are independently selected from one or more of N, O and S;
  • the number of heteroatoms is independently 1, 2, 3 or 4, and the heteroatoms are independently selected from one or more of N, O and S;
  • R 3 is C 1 to C 6 alkyl or C 3 to C 6 cycloalkyl
  • R 4 is hydrogen, C 1 to C 3 alkyl, C 3 to C 6 cycloalkyl or halogen.
  • the C 1 to C 6 alkyl group can be independently a C 1 to C 3 alkyl group; and can also be independently a methyl, ethyl, n-propyl or isopropyl group, For example, methyl.
  • the C 3 to C 6 cycloalkyl group can be independently a C 3 to C 4 cycloalkyl group; and can also be independently a cyclopropyl group.
  • the C 1 -C 3 alkyl group in R a1 , R a2 , R a3 or R a4 , can be independently methyl.
  • the C 1 to C 3 alkyl group in the C 1 to C 3 alkyl group substituted by 1, 2 or 3 halogens can be Independently methyl.
  • the halogen in R a1 , R a2 , R a3 or R a4 , the halogen can independently be fluorine.
  • the 3- to 10-membered heterocarbocyclic ring may be a 6- to 8-membered heterocarbocyclic ring.
  • the 3- to 10-membered heterocarbocyclic ring can be a monocyclic ring or a bicyclic ring; the bicyclic ring can be a spiro ring or a bridged ring, such as a bridged ring.
  • the number of heteroatoms in the 3- to 10-membered heterocarbocyclic ring may be 1 or 2.
  • the heteroatom in ring A, in the 3-10 membered heterocarbocyclic ring, the heteroatom may be N.
  • the 3-10 membered heterocarbocyclic ring is, for example,
  • the 3-10-membered carbocyclic ring is a 3-6-membered carbocyclic ring, such as cyclopropyl ring, cyclobutyl ring, cyclopentyl ring or cyclohexyl ring,
  • the carbon atom marked with * means that when it is a chiral carbon atom, it is S configuration, R configuration or a mixture thereof; the carbon atom marked # means that when it is a chiral carbon atom, it is S configuration, R configuration or a mixture thereof.
  • the C 1 to C 6 alkylene group in L 1 or L 2 , can be independently a C 1 to C 4 alkylene group; it can also be independently a methylene group, an ethylene group or a
  • the q end is connected to R2 .
  • the C 1 to C 6 alkyl group can be a C 1 to C 3 alkyl group; it can also be a methyl, ethyl, n-propyl or isopropyl group.
  • the C 1 to C 6 alkyl group in the C 1 to C 6 alkyl group substituted by 1, 2 or 3 R 23 can be a C 1 to C 3 alkyl group. ; It can also be methyl, ethyl, n-propyl or isopropyl.
  • the 3- to 10-membered cycloalkyl group in R 2 , can be a 3- to 6-membered cycloalkyl group; it can also be a 3 to 4-membered cycloalkyl group, such as cyclopropyl or cyclobutyl, and Such as cyclopropyl.
  • the 3-10-membered cycloalkyl group in the 3-10-membered cycloalkyl group substituted by 1, 2 or 3 R 22 can be a 3-6-membered cycloalkyl group. ; It can also be a 3 to 4-membered cycloalkyl group, such as cyclopropyl or cyclobutyl, or cyclopropyl.
  • the 3- to 10-membered heterocycloalkyl group in R 2 , can be a monocyclic ring or a bicyclic ring; the bicyclic ring can be a spiro ring or a bridged ring, for example, the 3- to 10-membered heterocycloalkyl group is Single ring.
  • the number of heteroatoms in the 3- to 10-membered heterocycloalkyl group may be 1 or 2.
  • the heteroatom in R 2 , in the 3-10 membered heterocycloalkyl group, can be independently N or O.
  • the 3- to 10-membered heterocycloalkyl group can be a 3- to 6-membered heterocycloalkyl group; it can also be a 4 to 6-membered heterocycloalkyl group; it can also be
  • the 3-10-membered heterocycloalkyl group in the 3-10-membered heterocycloalkyl group substituted by 1, 2 or 3 R 21 can be monocyclic or bicyclic;
  • the bicyclic ring may be a spiro ring or a bridged ring, for example, the 3- to 10-membered heterocycloalkyl group may be a monocyclic ring.
  • the number of heteroatoms in the 3-10-membered heterocycloalkyl group can be For 1 or 2 pieces.
  • the heteroatom in the 3-10-membered heterocycloalkyl group can be independently is N or S.
  • the 3-10-membered heterocycloalkyl group in the 3-10-membered heterocycloalkyl group substituted by 1, 2 or 3 R 21 can be a 3-6-membered heterocycloalkyl group.
  • the 3 to 10 membered heterocycle in the 3 to 10 membered heterocycloalkyl and the 3 to 10 membered heterocycloalkyl substituted by 1, 2 or 3 R 21 Alkyl groups are each independently
  • the number of heteroatoms in the 5- to 6-membered heteroaryl group may be 2.
  • the number of heteroatoms in the 5- to 6-membered heteroaryl group may be 3 or 4.
  • the heteroatom in the 5- to 6-membered heteroaryl group may be N.
  • the 5- to 6-membered heteroaryl group in the 5- to 6-membered heteroaryl group substituted by 1, 2 or 3 R 26 can be
  • the 5- to 6-membered heteroaryl group and the 5- to 6-membered heteroaryl group in the 5- to 6-membered heteroaryl group substituted by 1, 2 or 3 R 26 are each independently for
  • the C 1 to C 6 alkyl group can be independently a C 1 to C 3 alkyl group; it can also be independently a methyl, ethyl, n-propyl or isopropyl group, For example, methyl.
  • the halogen in R 23 , can independently be fluorine.
  • the C 1 to C 6 alkyl group can be a C 1 to C 3 alkyl group; it can also be a methyl, ethyl, n-propyl or isopropyl group, such as methyl.
  • the C 1 to C 6 alkyl group in R 241 or R 241a , can be independently a C 1 to C 3 alkyl group; it can also be independently a methyl, ethyl, n-propyl or iso propyl.
  • the C 1 to C 6 alkyl group can be a C 1 to C 3 alkyl group; it can also be a methyl group, an ethyl group, an n-propyl group or an isopropyl group, such as a methyl group.
  • the C 1 to C 6 alkyl group can be independently a C 1 to C 3 alkyl group; it can also be independently a methyl, ethyl, n-propyl or isopropyl group, For example, methyl.
  • the C 1 to C 6 alkyl group can be independently a C 1 to C 3 alkyl group; it can also be independently a methyl group, an ethyl group, an n-propyl group or an iso-propyl group.
  • Propyl such as methyl.
  • the C 3 to C 6 cycloalkyl group can be independently a C 3 to C 4 cycloalkyl group; and can also be independently a cyclopropyl group.
  • the halogen in R 27 or R 27a , can be F.
  • the C 1 to C 6 alkyl group in the C 1 to C 6 alkyl group substituted by 1, 2 or 3 halogens can be independently C 1 to C 3 Alkyl; it can also be independently ethyl.
  • the C 1 to C 6 alkyl group can be independently a C 1 to C 3 alkyl group; it can also be independently a methyl, ethyl, n-propyl or isopropyl group, For example, methyl.
  • the C 1 to C 6 alkyl group can be a C 1 to C 3 alkyl group; it can also be a methyl, ethyl, n-propyl or isopropyl group, such as isopropyl .
  • the C 3 to C 6 cycloalkyl group can be a C 3 to C 4 cycloalkyl group; it can also be a cyclopropyl group.
  • the halogen in R A , can be fluorine, chlorine or bromine, or fluorine.
  • the C 1 to C 3 alkyl group may be methyl, ethyl, n-propyl or isopropyl.
  • the C 3 to C 6 cycloalkyl group can be a C 3 to C 4 cycloalkyl group; it can also be a cyclopropyl group.
  • the halogen in R 4 , can be fluorine.
  • R 1 is
  • R a is independently C 1 to C 6 alkyl or C 3-6 cycloalkyl
  • R a1 , R a2 , R a3 and R a4 are independently hydrogen, C 1 to C 3 alkyl, halogen or C 1 to C 3 alkyl substituted by 1, 2 or 3 halogens;
  • U is independently N or CH
  • R 1 is For example
  • Ring A is a 3-10 membered heterocarbocyclic ring.
  • ring A is a 3-10 membered carbocyclic ring.
  • R 21 , R 22 and R 23 are independently hydroxyl, cyano, oxo, halogen or C 1 to C 6 alkyl;
  • R 24 is C 1 to C 6 alkyl or (R 241 ) (R 241a ) N-, R 241 and R 241a are independently hydrogen or C 1 to C 6 alkyl;
  • R 25 is C 1 to C 6 alkyl;
  • R 26 is independently C 1 to C 6 alkyl;
  • R 271 is independently C 1 to C 6 alkyl.
  • R 3 is C 1 to C 6 alkyl or C 3-6 cycloalkyl.
  • R 4 is hydrogen, C 1 -C 3 alkyl, C 3-6 cycloalkyl or halogen.
  • RA is hydrogen or halogen
  • RA is halogen
  • R4 is hydrogen or halogen.
  • R4 is hydrogen
  • R 1 is
  • R a is C 1 to C 6 alkyl
  • R a1 , R a2 , R a3 and R a4 are independently hydrogen or C 1 to C 3 alkyl
  • U is independently N or CH
  • Ring A is a 3 to 10-membered heterocarbocyclic ring or a 3 to 10-membered carbocyclic ring;
  • R 21 and R 22 are independently hydroxyl, oxo, cyano, halogen or C 1 to C 6 alkyl;
  • R 24 is C 1 to C 6 alkyl or (R 241 ) (R 241a ) N-, R 241 and R 241a are independently hydrogen;
  • R 25 is C 1 to C 6 alkyl;
  • R 26 is independently C 1 to C 6 alkyl;
  • the number of heteroatoms is 1 or 2, and the heteroatom is N;
  • the number of heteroatoms is independently 1 or 2, and the heteroatoms are independently selected from one or more of N, O and S;
  • the number of heteroatoms is 2 to 4, and the heteroatoms are independently selected from one or more of N, O and S;
  • V is CR 4 or N
  • R A is hydrogen or halogen
  • R 3 is C 1 to C 6 alkyl
  • R 4 is hydrogen
  • R 1 is
  • R a is methyl
  • R a1 , R a2 , R a3 and R a4 are independently hydrogen or C 1 to C 3 alkyl
  • U is independently N or CH
  • Ring A is a 3-10 membered heterocarbon ring
  • R 21 and R 22 are independently hydroxyl, oxo, cyano, halogen or C 1 to C 6 alkyl;
  • R 24 is C 1 to C 6 alkyl or (R 241 ) (R 241a ) N-, R 241 and R 241a are independently hydrogen;
  • R 25 is C 1 to C 6 alkyl;
  • R 26 is independently C 1 to C 6 alkyl;
  • the number of heteroatoms is 1 or 2, and the heteroatom is N;
  • the number of heteroatoms is independently 1 or 2, and the heteroatoms are independently selected from one or more of N, O and S;
  • the number of heteroatoms is 2, and the heteroatoms are independently selected from one or more of N, O and S;
  • V is CR 4 or N
  • R A is hydrogen or halogen
  • R 3 is C 1 to C 6 alkyl
  • R 4 is hydrogen
  • R 1 is
  • R a is C 1 to C 6 alkyl
  • R a1 , R a2 , R a3 and R a4 are independently hydrogen or C 1 to C 3 alkyl
  • U is independently N or CH
  • Ring A is a 3-10 membered heterocarbon ring
  • R 21 and R 22 are independently oxo, cyano, halogen or C 1 to C 6 alkyl;
  • R 24 is C 1 to C 6 alkyl or (R 241 ) (R 241a ) N-, R 241 and R 241a is independently hydrogen;
  • R 25 is C 1 to C 6 alkyl;
  • R 26 is independently C 1 to C 6 alkyl;
  • the number of heteroatoms is 1, and the heteroatom is N;
  • the number of heteroatoms is independently 1 or 2, and the heteroatoms are independently selected from one or more of N, O and S;
  • the number of heteroatoms is 2, and the heteroatoms are independently selected from one or more of N, O and S;
  • R 3 is C 1 to C 6 alkyl
  • R 4 is hydrogen
  • R 1 is
  • R 1 is
  • R 1 is
  • R a is C 1 -C 6 alkyl, such as methyl.
  • R a1 , R a2 , R a3 and R a4 are independently hydrogen or C 1 -C 3 alkyl.
  • R 21 and R 22 are independently hydroxyl, oxo, cyano, halogen or C 1 to C 6 alkyl;
  • R 24 is C 1 to C 6 alkyl or (R 241 ) (R 241a )N-, R 241 and R 241a are independently hydrogen;
  • R 25 is C 1 to C 6 alkyl;
  • R 26 is independently C 1 to C 6 alkyl;
  • R 21 and R 22 are independently oxo, cyano, halogen or C 1 to C 6 alkyl;
  • R 24 is C 1 to C 6 alkyl or (R 241 ) (R 241a ) N-, R 241 and R 241a are independently hydrogen;
  • R 25 is C 1 to C 6 alkyl;
  • R 26 is independently C 1 to C 6 alkyl;
  • the number of heteroatoms in the 5- to 6-membered heteroaryl group is 2, 3 or 4, and the heteroatoms are independently selected from one or more of N, O and S.
  • V is CR 4
  • another example is N.
  • R A is, for example, hydrogen, or halogen.
  • R 3 is C 1 -C 6 alkyl.
  • R 4 is H.
  • R 1 , in, R a can be C 1 to C 6 alkyl or C 3-6 cycloalkyl;
  • R a1 and R a2 can be independently hydrogen or halogen;
  • R a3 can be hydrogen or halogen;
  • R a4 can be hydrogen, C 1 to C 3 alkyl or trifluoromethyl.
  • R 1 , in, R a can be C 1 ⁇ C 3 alkyl or C 3-6 cycloalkyl;
  • R a1 and R a2 can be independently hydrogen or fluorine;
  • R a3 can be hydrogen or fluorine;
  • R a4 can be hydrogen, methyl or tris Fluoromethyl.
  • R 1 , in, R a can be methyl, isopropyl or cyclopropyl;
  • R a1 and R a2 can be independently hydrogen or fluorine;
  • R a3 can be hydrogen or fluorine;
  • R a4 can be hydrogen, methyl or trifluoromethyl.
  • R 1 the can be
  • R 1 , X, Y and Z may be independently -C(R a1 )(R a2 )-, -O- or -S-; R a1 and R a2 may be independently hydrogen.
  • R 1 can be
  • R 1 can be
  • R a1 , R a2 and R a3 can be independently hydrogen, C 1 to C 3 alkyl, Trifluoromethyl or halogen.
  • R 1 can be
  • R 1 can be
  • R a1 and R a2 can be independently hydrogen or halogen;
  • R a3 can be hydrogen;
  • R a4 can be hydrogen, C 1 to C 3 alkyl or trifluoromethyl.
  • R a1 and R a2 can be independently hydrogen or fluorine;
  • R a3 can be hydrogen;
  • R a4 can be hydrogen, methyl or trifluoromethyl.
  • R 1 can be
  • R 1 can be
  • R 1 can be
  • R 1 can be
  • R 1 can be
  • R 1 can be
  • R a in R 1 , R a can be a C 1 to C 3 alkyl group, or it can be a methyl group, an ethyl group, an n-propyl group or an isopropyl group, or it can also be a methyl group.
  • R 1 can be
  • L1 can be a single bond.
  • L2 can be a single bond.
  • Ring A can be a 6- to 8-membered heterocarbocyclic ring.
  • the number of heteroatoms in ring A can be 1 or 2, and the heteroatoms can be N.
  • Ring A can be a 6- to 8-membered heterocarbocyclic ring, in which the number of heteroatoms can be independently 1 or 2, and the heteroatoms can be N.
  • ring A can be The s end is connected to L 1 .
  • ring A can be The s end is connected to L 1 .
  • R 2 can be hydrogen, 3 to 10 membered heterocycloalkyl, 3 to 10 membered heterocycloalkyl substituted by 1, 2 or 3 R 21 , substituted by 1, 2 or 3 R 21 3 to 10-membered cycloalkyl substituted by 3 R 22 or C 1 to C 6 alkyl substituted by 1, 2 or 3 R 23 ;
  • R 21 can be oxo
  • R 22 can be cyano
  • R 23 can be halogen.
  • R 2 can be hydrogen, a 3- to 6-membered heterocycloalkyl group, a 3- to 6-membered heterocycloalkyl group substituted by 1, 2 or 3 R 21 , a 3- to 6-membered heterocycloalkyl group substituted by 1, 2 or 3 R 21 3 to 4-membered cycloalkyl substituted by 3 R 22 or C 1 to C 6 alkyl substituted by 1, 2 or 3 R 23 ;
  • R 21 can be oxo
  • R 22 can be cyano
  • R 23 can be halogen.
  • R 2 can be hydrogen, 4-6 membered heterocycloalkyl, 4-6 membered heterocycloalkyl substituted by 1, 2 or 3 R 21 , substituted by 1, 2 or 3 R 21 A 4-membered cycloalkyl group substituted by 3 R 22 or a C 1 to C 3 alkyl group substituted by 1, 2 or 3 R 23 ;
  • R 21 can be oxo,
  • R 22 can be cyano, and
  • R 23 can be For halogen.
  • R 2 can be hydrogen
  • R 2 can be hydroxyl
  • R 21 can be hydroxyl, oxo or halogen;
  • R 24 can be C 1 to C 6 alkyl;
  • R 25 can be C 1 to C 6 alkyl;
  • R 26 can be C 1 to C 6 alkyl;
  • R 21 can be hydroxyl, oxo or halogen;
  • R 24 can be C 1 ⁇ C 3 alkyl;
  • R 25 can be C 1 ⁇ C 3 alkyl;
  • R 26 can be C 1 ⁇ C 3 alkyl;
  • R 21 can be hydroxyl, oxo or halogen;
  • R 24 can be C 1 ⁇ C 3 alkyl;
  • R 25 can be C 1 ⁇ C 3 alkyl;
  • R 26 can be C 1 ⁇ C 3 alkyl group;
  • R 271 can be C 1 to C 3 alkyl base.
  • R 21 can be hydroxyl, oxo or halogen;
  • R 24 can be methyl;
  • R 25 can be methyl;
  • R 26 can be methyl;
  • R 2 can be cyano, trifluoromethyl
  • R 2 can be cyano, trifluoromethyl
  • R2 can be hydrogen, hydroxyl, cyano, trifluoromethyl,
  • R2 can be hydrogen, hydroxyl, cyano, trifluoromethyl,
  • R 2 is hydrogen, hydroxyl, cyano,
  • L 1 can be a single bond
  • L 2 can be a single bond
  • R 2 can be hydrogen, 3 to 10-membered heterocycloalkyl, 3 to 10-membered heterocycloalkyl substituted by 1, 2 or 3
  • R 21 R _ _ _ 21 may be oxo
  • R 22 may be cyano
  • R 23 may be halogen.
  • L 1 can be a single bond
  • L 2 can be a single bond
  • R 2 can be hydrogen, 3 to 6-membered heterocycloalkyl, 3 to 3 substituted by 1, 2 or 3
  • R 21 R _ _ _ 21 may be oxo
  • R 22 may be cyano
  • R 23 may be halogen.
  • L 1 can be a single bond
  • L 2 can be a single bond
  • R 2 can be hydrogen, 4-6 membered heterocycloalkyl, 4-6 substituted by 1, 2 or 3 R 21 6-membered heterocycloalkyl, 4-membered cycloalkyl substituted by 1, 2 or 3 R 22 or C 1 to C 3 alkyl substituted by 1 , 2 or 3 R 23
  • R 21 can is oxo
  • R 22 may be cyano
  • R 23 may be halogen.
  • L 1 can be a single bond
  • L 2 can be a single bond
  • R 2 can be hydrogen
  • L 1 can be a single bond
  • R 21 can be C 1 to C 6 alkyl or Oxo
  • R 22 can be cyano
  • R 24 can be C 1 to C 6 alkyl, (R 241 ) (R 241a ) N-, R 241 and R 241a can be independently hydrogen or C 1 to C 6 alkyl group
  • R 25 can be a C 1 to C 6 alkyl group
  • R 26 can be a C 1 to C 6 alkyl group.
  • L 1 can be a single bond
  • R 21 can be C 1 to C 3 alkyl or oxo
  • R 22 can be cyano
  • R 24 can be C 1 to C 3 alkyl, (R 241 ) (R 241a ) N-, R 241 and R 241a can be independently hydrogen or C 1 to C 3 alkyl
  • R 25 can be a C 1 to C 3 alkyl group
  • R 26 can be a C 1 to C 3 alkyl group
  • L 1 can be a single bond
  • R 21 can be C 1 to C 3 alkyl or oxo
  • R 22 can be is cyano
  • R 24 can be C 1 to C 3 alkyl, (R 241 ) (R 241a ) N-, R 241 and R 241a can be independently hydrogen or C 1 to C 3 alkyl
  • R 25 can be C 1 to C 3 alkyl
  • R 26 can be C 1 to C 3 alkyl.
  • L 1 can be a single bond
  • R 21 can be methyl or oxo
  • R 22 can be cyano
  • R 24 may be methyl, (R 241 )(R 241a )N-, R 241 and R 241a may be independently hydrogen or methyl
  • R 25 may be methyl
  • R 26 may be methyl.
  • L 1 can be a single bond
  • R 2 can be hydroxyl
  • R 21 can be hydroxyl, oxo or halogen;
  • R 24 can be C 1 ⁇ C 6 alkyl;
  • R 25 can be C 1 ⁇ C 6 alky
  • R 21 can be hydroxyl, oxo or halogen;
  • R 24 can be C 1 ⁇ C 3 alkyl;
  • R 25 can be C 1 ⁇ C 3 alky
  • R 21 can be hydroxyl, oxo or halogen;
  • R 24 can be C 1 ⁇ C 3 alkyl;
  • R 25 can be C 1 ⁇ C 3 alkyl
  • R 21 can be hydroxyl, oxo or halogen;
  • R 24 can be methyl;
  • R 25 can be Methyl;
  • R 26 can be
  • R 2 can be cyano, trifluoromethyl
  • L 1 single bond, methylene, ethylene or -(C O)-or
  • the q end is connected to R2 .
  • R 21 can be C 1 to C 6 alkyl, oxo or halogen;
  • R 24 can be C 1 to C 6 alkyl or (R 241 ) (R 241a ) N-, R 241 and R 241a can be independently hydrogen or C 1 to C 6 alkyl;
  • R 27 and R 27a can be independently hydrogen or C 1 to C 6 alkyl.
  • R 2 can be hydrogen or 3-10 membered heterocycloalkyl.
  • R 2 can be hydrogen or 3-6 membered heterocycloalkyl.
  • R 2 can be hydrogen or 4-6 membered heterocycloalkyl.
  • R2 can be hydrogen or
  • R 21 can be C 1 ⁇ C 6 alkyl;
  • R 24 can be C 1 ⁇ C 6 alkyl or (R 241 ) (R 241a ) N-,
  • R 241 and R 241a can be independently hydrogen or C 1 ⁇ C 6 alkyl.
  • R 21 can be C 1 ⁇ C 3 alkyl;
  • R 24 can be C 1 ⁇ C 3 alkyl or (R 241 ) (R 241a ) N-,
  • R 241 and R 241a can independently be hydrogen or C 1 ⁇ C 3 alkyl.
  • R 21 may be methyl;
  • R 24 may be methyl or (R 241 )(R 241a )N-, and
  • R 241 and R 241a may independently be hydrogen or methyl.
  • R 2 can be hydroxyl
  • R 2 can be a 3-10 membered heterocycloalkyl group, a 3-10 membered heterocycloalkyl group substituted by 1, 2 or 3 R 21 or (R 27 ) (R 27a ) N -; R 21 may be oxo or halogen; R 27 and R 27a may independently be hydrogen or C 1 to C 6 alkyl.
  • R 2 can be a 3-6 membered heterocycloalkyl group, a 3-6 membered heterocycloalkyl group substituted by 1, 2 or 3 R 21 or (R 27 ) (R 27a ) N -; R 21 may be oxo or halogen; R 27 and R 27a may independently be hydrogen or C 1 to C 3 alkyl.
  • R 2 can be a 4-6 membered heterocycloalkyl group, a 4-6 membered heterocycloalkyl group substituted by 1, 2 or 3 R 21 or (R 27 ) (R 27a ) N -; R 21 may be oxo or halogen; R 27 and R 27a may independently be hydrogen or methyl.
  • R 2 can be
  • R 2 can be hydrogen, hydroxyl
  • L 1 can be a single bond
  • L 2 can be a single bond
  • R 2 can be hydrogen or a 3-10-membered heterocycloalkyl group.
  • L 1 can be a single bond
  • L 2 can be a single bond
  • R 2 can be hydrogen or a 3- to 6-membered heterocycloalkyl group.
  • L 1 can be a single bond
  • L 2 can be a single bond
  • R 2 can be hydrogen or a 4- to 6-membered heterocycloalkyl group.
  • L 1 can be a single bond
  • L 2 can be a single bond
  • R 2 can be hydrogen or
  • L 1 can be a single bond
  • R 2 can be hydroxyl, 5-6 membered heteroaryl, or 1
  • One, two or three R 21- substituted 3-10-membered heterocycloalkyl or R 24 -S( O) 2 -
  • R 21 can be C 1 ⁇ C 6 alkyl
  • R 24 can be C 1 ⁇ C 6 alkyl or (R 241 )(R 241a )N-, R 241 and R 241a may independently be hydrogen or C 1 to C 6 alkyl.
  • L 1 can be a single bond
  • R 2 can be hydroxyl, 5-6 membered heteroaryl, or 1
  • One, two or three R 21- substituted 3-6-membered heterocycloalkyl or R 24 -S( O) 2 -
  • R 21 can be C 1 ⁇ C 3 alkyl
  • R 24 can be C 1 ⁇ C 3 alkyl or (R 241 )(R 241a )N-, R 241 and R 241a may independently be hydrogen or C 1 to C 3 alkyl.
  • L 1 can be a single bond
  • R 21 can be methyl
  • R 24 can be methyl or (R 241 ) (R 241a )N-, R 241 and R 241a may independently be hydrogen or methyl.
  • L 1 can be a single bond
  • R 2 can be hydroxyl
  • R 2 can be a 3 to 10-membered heterocycloalkyl group, consisting of 1, 2, or 3
  • R 21 can be oxo or halogen;
  • R 27 and R 27a can independently be hydrogen or C 1 to C 6 alkyl.
  • R 2 can be a 3 to 6-membered heterocycloalkyl group, consisting of 1 , 3 to 6-membered heterocycloalkyl substituted by 2 or 3 R 21 or (R 27 ) (R 27a ) N-;
  • R 21 can be oxo or halogen;
  • R 27 and R 27a can independently be hydrogen or C 1 to C 3 alkyl.
  • R 2 can be a 4 to 6-membered heterocycloalkyl group, consisting of 1 , 4-6 membered heterocycloalkyl substituted by 2 or 3 R 21 or (R 27 ) (R 27a ) N-;
  • R 21 can be oxo or halogen;
  • R 27 and R 27a can independently be hydrogen or methyl.
  • R 2 can be
  • R 3 can be C 1 -C 3 alkyl or C 3-6 cycloalkyl.
  • R3 can be isopropyl or cyclopropyl.
  • R 4 can be hydrogen or methyl.
  • the compound represented by formula II can be any one of the following compounds:
  • the compound represented by formula II is A stereoisomer that peaks first under the following conditions: Chromatographic column: Ultimate XB-C18, mobile phase: a mixed solution of acetonitrile and water containing 0.1v% formic acid; the elution gradient is: acetonitrile in the mobile phase within 0-3 minutes The volume percentage of acetonitrile in the mobile phase is 10%. Within 3-16 min, the volume percentage of acetonitrile in the mobile phase rises from 25% to 45%.
  • the volume percentage of acetonitrile in the mobile phase is 95%; preferably, it is as follows A stereoisomer with a retention time of 4.03 min under the conditions, the conditions are: chromatographic column: Ultimate XB-C18, 21.2*250mm, 10 ⁇ m; mobile phase: a mixed solution of acetonitrile and water containing 0.1v% formic acid; mobile phase : A mixed solution of acetonitrile and water containing 0.1v% formic acid; the elution gradient is: within 0-3min, the volume percentage of acetonitrile in the mobile phase is 10%, within 3-16min, the volume percentage of acetonitrile in the mobile phase increases from 25% to 45%, within 16-20min, the volume percentage of acetonitrile in the mobile phase is 95%; flow rate: 20ml/min; column temperature: 30°C.
  • the compound represented by formula II is The peak emerges after the following conditions
  • One stereoisomer Chromatographic column: Ultimate - Within 16 minutes, the volume percentage of acetonitrile in the mobile phase increases from 25% to 45%. Within 16-20 minutes, the volume percentage of acetonitrile in the mobile phase is 95%; preferably, the retention time is 4.10 minutes under the following conditions
  • a stereoisomer the conditions are: chromatographic column: Ultimate XB-C18, 21.2*250mm, 10 ⁇ m; mobile phase: a mixed solution of acetonitrile and water containing 0.1v% formic acid; mobile phase: acetonitrile containing 0.1v% formic acid Mix solution with water; the elution gradient is: within 0-3min, the volume percentage of acetonitrile in the mobile phase is 10%, within 3-16min, the volume percentage of acetonitrile in the mobile phase increases from 25% to 45%, within 16-20min, The volume percentage of acetonitrile in the mobile
  • the present invention also provides a pharmaceutical composition, which includes substance X and pharmaceutical excipients;
  • the substance X is the above-mentioned compound represented by formula II, a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of a pharmaceutically acceptable salt thereof.
  • the invention also provides the use of substance X in preparing TLR7 and/or TLR8 inhibitors
  • the substance X is the above-mentioned compound represented by formula II, a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of a pharmaceutically acceptable salt thereof.
  • the TLR7 and/or TLR8 inhibitor may be selective relative to TLR9.
  • the TLR7 and/or TLR8 inhibitors can be used in vitro.
  • the present invention also provides an application of substance X in the preparation of medicines
  • the substance X is the above-mentioned compound represented by formula II, a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of a pharmaceutically acceptable salt thereof;
  • the drugs are drugs used to treat diseases related to TLR7 and/or TLR8.
  • the disease related to TLR7 and/or TLR8 may be an autoimmune disease, or may be Sjogren's syndrome, lupus erythematosus, multiple sclerosis, rheumatoid arthritis, systemic sclerosis or Psoriasis.
  • the lupus erythematosus may be systemic lupus erythematosus (SLE), discoid lupus erythematosus or subacute cutaneous lupus erythematosus.
  • SLE systemic lupus erythematosus
  • discoid lupus erythematosus or subacute cutaneous lupus erythematosus.
  • the present invention also provides the use of substance X in the preparation of medicines for treating autoimmune diseases
  • the substance X is the above-mentioned compound represented by formula II, a pharmaceutically acceptable salt thereof, a solvate thereof or a solvate of a pharmaceutically acceptable salt thereof;
  • the autoimmune disease may be Sjögren's syndrome, lupus erythematosus, multiple sclerosis, rheumatoid arthritis, systemic sclerosis or psoriasis.
  • the lupus erythematosus may be systemic lupus erythematosus (SLE), discoid lupus erythematosus or subacute cutaneous lupus erythematosus.
  • SLE systemic lupus erythematosus
  • discoid lupus erythematosus or subacute cutaneous lupus erythematosus.
  • pharmaceutically acceptable refers to those compounds, materials, compositions and/or dosage forms that are found in reliable medical To the extent judged to be suitable for use in contact with human and animal tissue without undue toxicity, irritation, allergic reactions or other problems or complications and commensurate with a reasonable benefit/risk ratio.
  • salts refers to salts of compounds of the present invention prepared from compounds having specific substituents found in the present invention and relatively non-toxic acids or bases.
  • base addition salts can be obtained by contacting the free form of such compounds with a sufficient amount of base in pure solution or in a suitable inert solvent.
  • Pharmaceutically acceptable base addition salts include sodium, potassium calcium, ammonium, organic amine or magnesium salts or similar salts.
  • base addition salts can be obtained by contacting the free form of such compounds with a sufficient amount of acid in neat solution or in a suitable inert solvent.
  • Examples of pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, bicarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid , hydrogen sulfate, hydriodic acid, phosphorous acid, etc., and organic acid salts, including acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid , fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, methanesulfonic acid and other similar acids; also includes amino acids (such as arginine, etc.) salts, and salts of organic acids such as glucuronic acid.
  • Certain specific compounds of the present invention contain both basic and acidic functional groups
  • the pharmaceutically acceptable salts of the present invention can be synthesized by conventional chemical methods from parent compounds containing acid groups or bases.
  • such salts are prepared by reacting the free acid or base form of these compounds with a stoichiometric amount of the appropriate base or acid in water or an organic solvent or a mixture of the two.
  • the "-" at the end of a group means that the group is connected to the rest of the molecule through that site.
  • the number of atoms in a ring is usually defined as the ring membership.
  • a "3-7 membered ring” refers to a “ring” with 3-7 atoms arranged around it.
  • halogen refers to fluorine, chlorine, bromine and iodine.
  • alkyl refers to a linear or branched, saturated, monovalent hydrocarbon group having the specified number of carbon atoms (eg, C 1 -C 6 ).
  • Alkyl groups include, but are not limited to: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, etc.
  • alkylene is a divalent group connected to the rest of the molecule by two single bonds and is otherwise defined in the same manner as the term "alkyl”.
  • cycloalkyl refers to a cyclic, saturated, monovalent hydrocarbon group having a specified number of carbon atoms (for example, C 3 -C 10 ), which is a monocyclic or bicyclic system, and the bicyclic ring can be Spiro ring or bridged ring, the spiro ring refers to one carbon atom shared between single rings, and the bridged ring refers to two or more carbon atoms shared between single rings.
  • Cycloalkyl groups include, but are not limited to: wait.
  • Carbocycle meets any of the following conditions, and the rest of the definitions are the same as the term “cycloalkyl”: 1. It is connected to the rest of the molecule through more than two single bonds; 2. It shares two atoms and one root with the rest of the molecule. bond; the term “carbocycle” refers to a saturated ring.
  • heterocycloalkyl refers to a group having a specified number of ring atoms (e.g., 3-10 members) and a specified number of heteroatoms (e.g., 1, 2 or 3), a cyclic, saturated monovalent group of specified heteroatom species (one or more of N, O and S), which is a single ring system or a bicyclic system, and the bicyclic ring can be It is a spiro ring or a bridged ring.
  • the spiro ring means that the single rings share one carbon atom
  • the bridged ring means that the single rings share more than two carbon atoms.
  • Heterocycloalkyl groups are attached to the rest of the molecule through carbon atoms or heteroatoms. Heterocycloalkyl groups include, but are not limited to: wait.
  • heterocarbocycle is connected to the rest of the molecule by more than two single bonds, and the rest of the definition is the same as the term “heterocycloalkyl”.
  • heterocarbocycle refers to a saturated ring.
  • 5-6 membered heteroaryl by itself or in combination with other terms refers to a cyclic group with a conjugated ⁇ electron system consisting of 5 to 6 ring atoms, with 1, 2, 3 or 4 ring atoms. are heteroatoms independently selected from O, S and N, and the remainder are carbon atoms.
  • the 5-6 membered heteroaryl group can be connected to the rest of the molecule through a heteroatom or a carbon atom, and the 5-6 membered heteroaryl group includes 5-membered and 6-membered heteroaryl groups and the like.
  • Examples of the 5-6 membered heteroaryl include but are not limited to pyrrolyl (including N-pyrrolyl, 2-pyrrolyl and 3-pyrrolyl, etc.), pyrazolyl (including 2-pyrazolyl and 3-pyrrolyl).
  • azolyl group, etc. imidazolyl group (including N-imidazolyl, 2-imidazolyl, 4-imidazolyl and 5-imidazolyl, etc.), oxazolyl (including 2-oxazolyl, 4-oxazolyl and 5-oxazolyl) Oxazolyl, etc.), triazolyl (1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl, 1H-1,2,4-triazolyl and 4H-1, 2,4-triazolyl), tetrazolyl, isoxazolyl (including 3-isoxazolyl, 4-isoxazolyl and 5-isoxazolyl, etc.), thiazolyl (including 2-thiazolyl , 4-thiazolyl and 5-thiazolyl, etc.), furyl (including 2-furyl and 3-furyl, etc.), thienyl (including 2-thienyl and 3-thienyl, etc.), pyridyl
  • treatment refers to the administration of one or more pharmaceutical substances, in particular compounds of formula (I) according to the invention and or pharmaceutically acceptable salts thereof, to an individual suffering from a disease or having symptoms of said disease, with To cure, alleviate, alleviate, alter, treat, ameliorate, ameliorate or affect the disease or the symptoms of the disease.
  • the terms “treat,” “contact,” and “react” refer to the addition or mixing of two or more reagents under appropriate conditions to produce the indicated and or desired products. It is understood that , the reaction leading to the product shown and/or desired may not necessarily result directly from the combination of the two reagents initially added, i.e. there may be one or more intermediates generated in the mixture that ultimately lead to Formation of the indicated and or desired products.
  • a "patient” is defined as any warm-blooded animal, such as, but not limited to, a mouse, guinea pig, dog, horse or human, preferably the patient is a human.
  • the reagents and raw materials used in the present invention are all commercially available.
  • the positive and progressive effect of the present invention is that this type of compound has good inhibitory activity on TLR7/8.
  • the reverse-phase preparation and purification conditions in the examples are: Preparation column: Ultimate ) 10v% (0-3min), 25-45v% (3-16min), 95v% (16-20min), flow rate 20mL/min, column temperature 30°C, refer to this for the reverse-phase preparation and purification steps in Example 1-76 condition.
  • Step B 4-(3-isopropyl-1H-indol-5-yl)-5,6-dihydropyridine-1(2H)-carboxylic acid tert-butyl ester (compound 1.3)
  • Step C tert-butyl 4-(3-isopropyl-1H-indol-5-yl)piperidine-1-carboxylate (compound 1.4)
  • Step D 4-(2-Bromo-3-isopropyl-1H-indol-5-yl)piperidine-1-carboxylic acid tert-butyl ester (compound 1.5)
  • Step E 4-(3-isopropyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole -5-yl)piperidine-1-carboxylic acid tert-butyl ester (compound 1.6)
  • Step F 2-((trifluoromethyl)sulfonyl)oxy)cyclopent-1-en-1-carboxylic acid methyl ester (compound 1.8)
  • 2-Oxocyclopentane-1-carboxylic acid methyl ester (10 g, 70.35 mmol) and N,N-diisopropylethylamine (13.64 g, 105.53 mmol) were dissolved in dichloromethane (100 mL).
  • Trifluoromethanesulfonic anhydride 21.83g, 77.38mmol was dissolved in dichloromethane (50mL), and the solution was slowly dropped into the reaction solution at -25°C. After the dripping is completed, the reaction solution continues to stir and react at -25-0°C for 2 hours.
  • Step G Methyl 2-(trimethylsilyl)ethynyl)cyclopent-1-ene-1-carboxylate (Compound 1.9)
  • Step I 2-ethylcyclopent-1-ene-1-carboxamide (compound 1.11)
  • Step K 2-Methyl-2,5,6,7-tetrahydro-1H-cyclopenta[c]pyridin-1-one (compound 1.13)
  • Step L 4-bromo-2-methyl-2,5,6,7-tetrahydro-1H-cyclopenta[c]pyridin-1-one (compound 1.14)
  • Step M tert-Butyl 4-(3-isopropyl-2-(2-methyl-1-oxy-2,5,6,7-tetrahydro-1H-cyclopentane[c]pyridine-4 -yl)-1H-indol-5-yl)piperidine-1-carboxylate (compound 1.15)
  • reaction solution was stirred and reacted at 85°C for 2 hours.
  • the reaction solution was concentrated under reduced pressure and purified by column chromatography (DCM/MeOH (5%)) to obtain product 1.15 (270 mg, crude product yield: 108%).
  • Step N 4-(3-isopropyl-5-(piperidin-4-yl)-1H-indol-2-yl)-2-methyl-2,5,6,7-tetrahydro-1H -Cyclopent[c]pyridin-1-one (compound 1)
  • Step A Ethyl 2-(4-(3-isopropyl-2-(2-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopenta[c]pyridine -4-yl)-1H-indol-5-yl)piperidin-1-yl)acetate (compound 5.1)
  • Step B 2-(4-(3-isopropyl-2-(2-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopentadiene[c]pyridine- 4-yl)-1H-indol-5-yl)piperidin-1-yl)acetic acid (compound 5.2)
  • Step C 4-(5-(1-(2-(1,1-Dioxidethiomorpholino)-2-oxoethyl)piperidin-4-yl)-3-isopropyl-1H-indole Indo-2-yl)-2-methyl-2,5,6,7-tetrahydro-1H-cyclopenta[c]pyridin-1-one (compound 5)
  • Step B 2-(4-(3-isopropyl-2-(2-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopentadiene[c]pyridine- 4-yl)-1H-indol-5-yl)piperidin-1-yl)ethanesulfonamide (compound 9)
  • Step A tert-butyl (2-(4-(3-isopropyl-2-(2-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopentadienyl[c ]pyridin-4-yl)-1H-indol-5-yl)piperidin-1-yl)-2-oxoethyl)(methyl)carbamate (compound 12.1)
  • Step B 4-(3-isopropyl-5-(1-(methylglycyl)piperidin-4-yl)-1H-indol-2-yl)-2-methyl-2,5 ,6,7-tetrahydro-1H-cyclopenta[c] Pyridin-1-one (compound 12)
  • Step A 4-nitrophenyl 4-(3-isopropyl-2-(2-methyl-1-oxy-2,5,6,7-tetrahydro-1H-cyclopentane [c] Pyridin-4-yl)-1H-indol-5-yl)piperidine-1-carboxylate (compound 15.1)
  • Step B 4-(3-isopropyl-5-(1-(4-methylpiperazine-1-carbonyl)piperidin-4-yl)-1H-indol-2-yl)-2-methyl Base-2,5,6,7-tetrahydro-1H-cyclopentane[c]pyridin-1-one (compound 15)
  • Step B (E)-5-bromo-4-(2-(dimethylamino)ethenyl)-1-methyl-3-nitropyridin-2(1H)-one (Compound 16.3)
  • Step C 4-bromo-6-methyl-1,6-dihydro-7H-pyrrole[2,3-c]pyridin-7-one (compound 16.4)
  • Step D 4-(3-isopropyl-2-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrole[2,3-c]pyridin-4-yl)-1H -indole-5-yl)piper tert-butylpyridine-1-carboxylate (compound 16.5)
  • Step E 4-(3-isopropyl-5-(piperidin-4-yl)-1H-indol-2-yl)-6-methyl-1,6-dihydro-7H-pyrrole [2 ,3-c]pyridin-7-one (compound 16.6)
  • Step F 2-(4-(3-isopropyl-2-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrole[2,3-c]pyridin-4-yl )-1H-indol-5-yl)piperidin-1-yl)acetamide (compound 16)
  • Step B 3,4-bis(methoxycarbonyl)pyridine 1-oxide (compound 17.3)
  • Step E 4-Chloro-1,3-dihydrofuran[3,4-c]pyridine (Compound 17.6)
  • Step F N-(2,4-dimethoxybenzyl)-1,3-dihydrofuro[3,4-c]pyridin-4-amine (Compound 17.7)
  • Step H 7-bromo-1,3-dihydrofuran[3,4-c]pyridin-4-amine (compound 17.9)
  • Step I 6-bromo-7,9-dihydrofura[3,4-c][1,2,4]triazolo[1,5-a]pyridine (compound 17.10)
  • Step J tert-Butyl 4-(2-(7,9-dihydrofuran[3,4-c][1,2,4]triazolyl[1,5-a]pyridin-6-yl)- 3-isopropyl-1H-indol-5-yl)piperidine-1-carboxylate (compound 17.11)
  • Step K 6-(3-isopropyl-5-(piperidin-4-yl)-1H-indol-2-yl)-7,9-dihydrofuran[3,4-c][1, 2,4]triazolyl[1,5-a]pyridine (compound 17.12)
  • Step L 2-(4-(2-(7,9-dihydrofuran[3,4-c][1,2,4]triazolyl[1,5-a]pyridin-6-yl)- 3-isopropyl-1H-indol-5-yl)piperidin-1-yl)acetamide (compound 17)
  • Step A Ethyl 2-(4-(3-isopropyl-2-(2-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopentyl[c]pyridine -4-yl)-1H-indol-5-yl)piperidin-1-yl)acetate (compound 22.1)
  • Step B 2-(4-(3-isopropyl-2-(2-methyl-1-oxo-2,5,6,7-tetrahydro-1H-cyclopentyl[c]pyridine-4 -yl)-1H-indol-5-yl)piperidin-1-yl)acetic acid (compound 22.2)
  • Step C 4-(5-(1-(2-(3,3-difluoropyrrolidin-1-yl)-2-oxoethyl)piperidin-4-yl)-3-isopropyl-1H -indol-2-yl)-2-methyl-2,5,6,7-tetrahydro-1H-cyclopentyl[c]pyridin-1-one (compound 22)
  • Step B 2-(2-Methyl-2H-tetrazol-5-yl)acetic acid (Compound 23.3)
  • Step C 4-(3-isopropyl-5-(1-(2-(2-methyl-2H-tetrazol-5-yl)acetyl)piperidin-4-yl)-1H-indole -2-yl)-2-methyl-2,5,6,7-tetrahydro-1H-cyclopentyl[c]pyridin-1-one (compound 23)
  • Step A 4-bromo-1,6-dimethyl-1,6-dihydro-7H-pyrrolo[2,3-c]pyridin-7-one (compound 35.2)
  • Step B tert-Butyl 4-(2-(1,6-dimethyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin-4-yl)- 3-isopropyl-1H-indol-5-yl)piperidine-1-carboxylate (compound 35.3)
  • Step C 4-(3-isopropyl-5-(piperidin-4-yl)-1H-indol-2-yl)-1,6-dimethyl-1,6-dihydro-7H- Pyrrolo[2,3-c]pyridin-7-one (compound 35.4)
  • Step D 2-(4-(2-(1,6-dimethyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin-4-yl)- 3-isopropyl-1H-indol-5-yl)piperidin-1-yl)acetamide (compound 35)
  • Step A tert-butyl(2-(4-(2-(7,9-dihydrofuran[3,4-c][1,2,4]triazolo[1,5-a]pyridine-6 -(yl)-3-isopropyl)-1H-indol-5-yl)piperidin-1-yl)-2-oxoethyl)(methyl)carbamate (compound 37.1)
  • Step B 1-(4-(2-(7,9-dihydrofuran[3,4-c][1,2,4]triazolo[1,5-a]pyridin-6-yl)- 3-isopropyl-1H-indol-5-yl)piperidin-1-yl)-2-(methylamino)ethane-1-one (compound 37)
  • Step A 4-nitrophenyl 4-(2-(7,9-dihydrofuran[3,4-c][1,2,4]triazolo[1,5-a]pyridine-6- yl)-3-isopropyl-1H-indol-5-yl)piperidine-1-carboxylate (compound 38.1)
  • Step B (4-(2-(7,9-dihydrofuran[3,4-c][1,2,4]triazolo[1,5-a]pyridin-6-yl)-3- Isopropyl-1H-indol-5-yl)piperidin-1-yl)(4-methylpiperazin-1-yl)methanone (compound 38)
  • Step B 2-(4-(2-(7,9-dihydrofuran[3,4-c][1,2,4]triazole[1,5-a]pyridin-6-yl)-3 -Isopropyl-1H-indol-5-yl)piperidin-1-yl)ethane-1-sulfonamide (compound 39)
  • Step A Ethyl 2-(4-(2-(7,9-dihydrofuran[3,4-c][1,2,4]triazolo[1,5-a]pyridin-6-yl )-3-isopropyl-1H-indol-5-yl)piperidin-1-yl)acetate (compound 41.1)
  • Step B 2-(4-(2-(7,9-dihydrofuran[3,4-c][1,2,4]triazolo[1,5-a]pyridin-6-yl)- 3-Isopropyl-1H-indol-5-yl)piperidin-1-yl)acetic acid (Compound 41.2)
  • Step C N-cyclopropyl-2-(4-(2-(7,9-dihydrofuran[3,4-c][1,2,4]triazolo[1,5-a]pyridine -6-yl)-3-isopropyl)-1H-indol-5-yl)piperidin-1-yl)acetamide (compound 41)
  • Dissolve compound 41.2 (50 mg, 0.11 mmol) in 3 mL DMF, add HATU (62.7 mg, 0.165 mmol) and triethylamine (33.4 mg, 0.33 mmol) solutions, stir the reaction solution at room temperature for 30 minutes, and then add dropwise Cyclopropylamine (9.4 mg, 0.165 mmol), the reaction solution was stirred at room temperature for 1 hour.
  • reaction solution was adjusted to a neutral pH with ammonium chloride aqueous solution, extracted with ethyl acetate (10mL ⁇ 3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and reversed phase preparation and purification to obtain Compound 41 (10.1 mg, yield: 18.4%), LCMS ESI (+) m/z: 499.3 (M+1).
  • Example 1 use 4-(2-(7,9-dihydrofuran[3,4-c][1,2,4]triazole[1,5-a]pyridin-6-yl)-3 -Isopropyl-1H-indol-5-yl) piperidine-1-carboxylic acid tert-butyl ester and 4M hydrochloric acid methanol solution as raw materials.
  • Example 1 4-bromo-6-methyl-1-toluenesulfonyl-1,6-dihydro-7H-pyrazolo[3,4-c]pyridin-7-one and 2-(4 -(3-isopropyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indol-5-yl ) piperidin-1-yl) acetamide as raw material.
  • Example 2 4-(4-fluoro-3-isopropyl-5-(piperazin-1-yl)-1H-pyrrolo[2,3-c]pyridin-2-yl)-2- It is prepared from methyl-2,5,6,7-tetrahydro-1H-cyclopentane[c]pyridin-1-one and 2-bromoacetamide.
  • Example 2 use 6-(4-fluoro-3-isopropyl-5-(piperidin-4-yl)-1H-pyrrolo[2,3-c]pyridin-2-yl)-7, It is prepared from 9-dihydrofuran[3,4-c][1,2,4]triazolo[1,5-a]pyridine and 2-bromoacetamide.
  • tert-butyl 2-(4-(2-(8,9-dihydro-7H-cyclopenta[c][1,2,4]triazolo[1,5-a]pyridine -6-yl)-3-isopropyl-1H-indol-5-yl)piperidin-1-yl)acetate and a dioxane solution of hydrochloric acid are used as raw materials.
  • Example 2 use 6-(4-fluoro-3-isopropyl-5-(piperazin-1-yl)-1H-pyrrolo[2,3-c]pyridin-2-yl)-7, It is prepared from 9-dihydrofuran[3,4-c][1,2,4]triazolo[1,5-a]pyridine and 2-bromoacetamide.
  • Step A 3-isopropyl-5-(1,4-dioxaspiro[4.5]dec-8-yl)-2-(4,4,5,5-tetramethyl-1,3,2 -Dioxaborane-2-yl)-1H-indole (Compound 72.2)
  • Step B 6-(3-isopropyl-5-(1,4-dioxaspirin[4.5]dec-8-yl)-1H-indol-2-yl)-7,9-dihydrofurano [3,4-c][1,2,4]triazolo[1,5-a]pyridine (compound 72.3)
  • Step C 4-(2-(7,9-dihydrofurfur[3,4-c][1,2,4]triazolo[1,5-a]pyridin-6-yl)-3-iso Propyl-1H-indol-5-yl)cyclohexan-1-one (Compound 72.4)
  • Step D 6-(5-(4-(3,3-difluoroazetidin-1-yl)cyclohexyl)-3-isopropyl-1H-indol-2-yl)-7, 9-Dihydrofuro[3,4-c][1,2,4]triazolo[1,5-a]pyridine (compound 72)
  • Step A 4-(3-isopropyl-5-(1,4-dioxaspiro[4.5]dec-8-yl)-1H-indol-2-yl)-2-methyl-2, 5,6,7-Tetrahydro-1H-cyclopentyl[c]pyridin-1-one (Compound 73.1)
  • Step B 4-(3-isopropyl-5-(4-oxocyclohexyl)-1H-indol-2-yl)-2-methyl-2,5,6,7-tetrahydro-1H -Cyclopentane[c]pyridine -1-one (compound 73.2)
  • Step C 4-(5-(4-(3,3-difluoroazetidin-1-yl)cyclohexyl)-3-isopropyl-1H-indol-2-yl)-2- Methyl-2,5,6,7-tetrahydro-1H-cyclopentyl[c]pyridin-1-one (compound 73)
  • Step A (S)-4-(5-(4-(3-fluoropyrrolidin-1-yl)cyclohexyl)-3-isopropyl-1H-indol-2-yl)-2-methyl -2,5,6,7-tetrahydro-1H-cyclopentyl[c]pyridin-1-one (compound 74)
  • Example 75 Example 75, Example 76
  • * marks the carbon atom as S configuration, R configuration or their mixture
  • # marks the carbon atom as S configuration, R configuration or their mixture.
  • Step A (S)-6-(5-(4-(3-fluoropyrrolidin-1-yl)cyclohexyl)-3-isopropyl-1H-indol-2-yl)-7,9- Dihydrofura[3,4-c][1,2,4]triazolo[1,5-a]pyridine
  • reaction solution was extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and purified by reverse phase preparation (preparation column: Ultimate XB-C18, 21.2*250mm, 10 ⁇ m; elution phase: mobile phase 0.1% FA in H2O /ACN, elution gradient (ratio of ACN) 10% (0-3min), 25-45% (3-16min), 95% (16-20min)) to obtain product 75 (retention time is 4.03min, 4.2mg, Yield: 18%) and product 76 (retention time 4.10 min, 3.3 mg, yield: 14%).
  • compound 75 is one of the stereoisomers.
  • Compound 76 is Another stereoisomer in .
  • Neomycin (Normocin TM , InvivoGen)
  • the purpose of this experiment is to detect the antagonistic activity of compounds in HEK-Blue TM hTLR7 cells through HEK alkaline phosphatase assay medium.
  • HEK-Blue hTLR7 cells were cultured in complete medium (containing 10% inactivated fetal bovine serum, 100ug/mL neomycin, 100ug/mL bleomycin, 10ug/mL blasticidin, 0.5% penicillin-streptomycin DMEM high-glucose medium containing phytozoic acid solution). Take cells that grow well and have a cell density of 80-90%, remove the growth medium supernatant, wash the cells once with preheated PBS, add preheated PBS, and culture and digest the cells at 37°C. Add complete culture medium to terminate digestion, transfer the cell suspension to a 15 mL centrifuge tube, and centrifuge to remove the supernatant.
  • complete medium containing 10% inactivated fetal bovine serum, 100ug/mL neomycin, 100ug/mL bleomycin, 10ug/mL blasticidin, 0.5% penicillin-streptomycin DMEM high-glucose medium containing
  • Add detection medium disperse the cells and count them to prepare a cell suspension with a concentration of 3.3 ⁇ 10 5 /mL. Add the cell suspension to the 384-well cell culture plate, 30uL/well, and the number of cells in each well is 10,000 cells/well.
  • the agonist R848 was prepared into a stock solution using 100% DMSO, with a concentration of 40mM; the test compound was prepared into a stock solution using 100% DMSO, with a concentration of 10mM.
  • the dispensing plate is shaken and mixed on a shaker (320 rpm) and set aside.
  • Centrifuge the preparation plate, and use ECHO to add the test solution to the 384-well cell culture plate containing the cell suspension add the test compound to the well containing the cell suspension, 50nL/well, and the final concentration of the compound is 1000, 333.3, 111.1, 37, 12.3, 4.1, 1.3, 0.45, 0.15, 0.05nM, and add the agonist R848, 50nL/well, as the compound group; add 100% DMSO to the well containing the cell suspension, 50nL/well, and Add agonist R848, 50 nL/well, as a positive control group; add 100% DMSO to the wells containing cell suspension, 100 nL/well, as a negative control group.
  • the final concentration of agonist R848 in each well was 0.8 uM, and the final concentration of DMSO in each well was 0.33%.
  • the cell plate was then placed in a 37°C, 5% CO2 incubator for 16 hours. Then the cell plate was taken out and centrifuged at 1000 rpm for 1 minute, and the optical density value OD 620 of each well at 620 nm was read using a microplate reader.
  • the software draws the measurement effect curve of the test compound and calculates the concentration of the compound when the inhibition rate reaches 50%, that is, the antagonistic activity IC50 value, see Table 1.
  • Neomycin (Normocin TM , InvivoGen)
  • the purpose of this experiment is to detect the antagonistic activity of compounds in HEK-Blue TM hTLR8 cells through HEK alkaline phosphatase assay medium.
  • HEK-Blue hTLR8 cells were cultured in complete medium (containing 10% inactivated fetal calf serum, 100ug/mL neomycin, 100ug/mL bleomycin, 30ug/mL blasticidin, 0.5% penicillin-streptomycin DMEM high-glucose medium containing phytozoic acid solution). Take cells that grow well and have a cell density of 80-90%, remove the growth medium supernatant, wash the cells once with preheated PBS, add preheated PBS, and culture and digest the cells at 37°C. Add complete culture medium to terminate digestion, transfer the cell suspension to a 15 mL centrifuge tube, and centrifuge to remove the supernatant.
  • complete medium containing 10% inactivated fetal calf serum, 100ug/mL neomycin, 100ug/mL bleomycin, 30ug/mL blasticidin, 0.5% penicillin-streptomycin DMEM high-glucose medium
  • Add detection medium disperse the cells and count them to prepare a cell suspension with a concentration of 3.3 ⁇ 10 5 /mL. Add the cell suspension to the 384-well cell culture plate, 30uL/well, and the number of cells in each well is 10,000 cells/well.
  • the agonist R848 was prepared into a stock solution using 100% DMSO, with a concentration of 40mM; the test compound was prepared into a stock solution using 100% DMSO, with a concentration of 10mM.
  • the dispensing plate is shaken and mixed on a shaker (320 rpm) and set aside.
  • Centrifuge the preparation plate, and use ECHO to add the test solution to the 384-well cell culture plate containing the cell suspension add the test compound to the well containing the cell suspension, 50nL/well, and the final concentration of the compound is 1000, 333.3, 111.1, 37, 12.3, 4.1, 1.3, 0.45, 0.15, 0.05nM, and add the agonist R848, 50nL/well, as a compound group; add 100% DMSO to the cells containing Add 50 nL/well of the suspension to the wells containing the cell suspension, and add the agonist R848, 50 nL/well, as a positive control group; add 100% DMSO to the wells containing the cell suspension, 100 nL/well, as a negative control group.
  • the final concentration of agonist R848 in each well was 3uM and the final concentration of DMSO in each well was 0.33%.
  • the cell plate was then placed in a 37°C, 5% CO2 incubator for 16 hours. Then the cell plate was taken out and centrifuged at 1000 rpm for 1 minute, and the optical density value OD 620 of each well at 620 nm was read using a microplate reader.
  • the software draws the measurement effect curve of the test compound and calculates the concentration of the compound when the inhibition rate reaches 50%, that is, the antagonistic activity IC50 value, see Table 1.
  • Neomycin (Normocin TM , InvivoGen)
  • the purpose of this experiment is to detect the antagonistic activity of compounds in HEK-Blue TM hTLR9 cells through HEK alkaline phosphatase assay medium.
  • HEK-Blue hTLR9 cells were cultured in complete medium (containing 10% inactivated fetal calf serum, 100ug/mL neomycin, 100ug/mL Bleomycin, 10ug/mL blasticidin, 0.5% penicillin-streptomycin solution in DMEM high glucose medium). Take cells that grow well and have a cell density of 80-90%, remove the growth medium supernatant, wash the cells once with preheated PBS, add preheated PBS, and culture and digest the cells at 37°C. Add complete culture medium to terminate digestion, transfer the cell suspension to a 15 mL centrifuge tube, and centrifuge to remove the supernatant.
  • complete medium containing 10% inactivated fetal calf serum, 100ug/mL neomycin, 100ug/mL Bleomycin, 10ug/mL blasticidin, 0.5% penicillin-streptomycin solution in DMEM high glucose medium.
  • Add detection medium disperse the cells and count them to prepare a cell suspension with a concentration of 5 ⁇ 10 5 /mL. Add the cell suspension to the 384-well cell culture plate, 30uL/well, and the number of cells in each well is 15,000 cells/well.
  • the dispensing plate is shaken and mixed on the shaker (320rpm) (except ODN2006) and set aside.
  • the final concentration of agonist ODN2006 in each well was 1 uM, and the final concentration of DMSO in each well was 0.167%.
  • the cell plate was then placed in a 37°C, 5% CO2 incubator for 16 hours. Then the cell plate was taken out and centrifuged at 1000 rpm for 1 minute, and the optical density value OD 620 of each well at 620 nm was read using a microplate reader.
  • the software draws the measurement effect curve of the test compound and calculates the compound concentration when the inhibition rate reaches 50%, that is, the antagonistic activity IC 50 value, see Table 1.
  • the IC 50 values of the controls Enpatoran and Afimetoran were also measured.
  • the compound of the present invention has obvious antagonistic effect on HEK-Blue-hTLR7/8 cells, but has no obvious antagonistic effect on HEK-Blue hTLR9 cells.
  • mice BALB/c female mice, Beijing Vitong Lihua Experimental Animal Technology Co., Ltd., 8 weeks old.
  • mice were randomly divided into blank group, control group and experimental group, with 6 animals in each group. After the animals were divided into groups, the corresponding compounds of the present invention (0.3 mg/kg) and the control Afimetoran (0.3 mg/kg) were administered; 0.5 h after administration, Gardiquimod 2 mg/kg was injected intraperitoneally. The endpoint of the experiment was reached 1.5 hours after administration of Gardiquimod. At the end of the experiment, the mice were anesthetized, blood was drawn from the heart into an anticoagulant tube, and plasma was collected for IL-6 cytokine detection. The IL-6 inhibition rate was calculated based on the IL-6 plasma concentrations in the control group and experimental group.
  • Inhibition rate (Avg(Vehicle)-Avg(Group))/(Avg(vehicle)-Avg(NC))*100%.
  • Avg (Vehicle) plasma IL-6 of the control group
  • Avg (Group) plasma IL-6 of the experimental group
  • Avg (NC) plasma IL-6 of the blank group.
  • the compound of the present invention has obvious inhibitory effect on IL-6.
  • the rapidly activated potassium channel encoded by the human ether-a-go-go-related gene is an important ion channel involved in the formation of the third phase repolarization of myocardial action potential.
  • Drugs that block hERG channels can lead to prolonged cardiac repolarization, and the electrocardiogram shows a prolonged QT interval, which is called long QT syndrome.
  • Drug-induced delayed ventricular repolarization may in some cases lead to a fatal arrhythmia - torsades de pointes Ventricular tachycardia.
  • This experiment used whole-cell patch clamp technology to study the inhibitory effect of compounds on hERG potassium channels and evaluate their risk of inducing ventricular repolarization toxicity.
  • the HEK293 cell line stably expressing the hERG potassium channel was used.
  • the hERG potassium channel cells were purchased from Creacell (catalog number: A-0320).
  • the HEK293 cell line stably expressing the hERG potassium channel was cultured in DMEM medium containing 10% fetal calf serum and 0.8 mg/mL G418 at a culture temperature of 37°C and a carbon dioxide concentration of 5%.
  • Cell passage Remove the old culture medium and wash once with PBS, then add 2mL TrypLE TM Express solution and incubate at 37°C for about 1 minute. When the cells detach from the bottom of the dish, add approximately 5 mL of complete culture medium preheated at 37°C. Gently pipet the cell suspension with a pipette to separate the aggregated cells. Transfer the cell suspension to a sterile centrifuge tube and centrifuge at 1000 rpm for 5 min to collect the cells. For expansion or maintenance culture, cells are seeded in a 10cm cell culture dish, and the number of cells seeded in each cell culture dish is 6 ⁇ 10 5 cells (final volume: 10mL).
  • cell density must not exceed 80%.
  • cells were separated with TrypLE TM Express before the test, culture medium was added to terminate digestion and centrifuged, cells were resuspended and counted, the cell density was adjusted to 2-3 ⁇ 10 6 cells/mL, and the cells were gently mixed with a room temperature balance shaker. 15-20 minutes on the machine for testing.
  • Electrophysiological testing was performed using fully automated patch clamp QPatch 48 X (Sophion) equipment. Place the prepared cells on the centrifuge of the Qpatch workbench, wash the cells using multiple centrifugation/suspension methods, and replace the cell culture medium with extracellular fluid. Take out an MTP-96 board and place it in the MTP source position. Take out the QPlate chip, and then place the QPlate at the Qplate source position. The robotic arm scans the MTP-96 board and QPlate chip barcode and grabs it to the measurement station. Draw intracellular and external fluids from the liquid pool and add them to the intracellular liquid pool, cell and test substance pool of the QPlate chip respectively. At the measuring station, all measuring points on the QPlate undergo initial quality control.
  • the quality control process includes drawing cell suspension from the cell container of the centrifuge, and positioning the cells onto the chip wells through a pressure controller to establish a high-resistance seal to form a whole-cell recording mode. Once a stable control current baseline is obtained, the test substance can be pipetted from the MTP-96 plate and applied to the cells according to the concentration gradient.
  • the voltage stimulation protocol for whole-cell patch clamp recording of whole-cell hERG potassium currents is as follows: when whole-cell sealing is formed, the cell membrane voltage is clamped at -80mV. The clamping voltage is depolarized from -80mV to -50mV for 0.5s (as leakage current detection), then stepped to 30mV for 2.5s, and then quickly returned to -50mV for 4s to stimulate the tail current of the hERG channel. Repeat data collection every 10 seconds to observe the effect of the drug on the hERG tail current. A -50mV stimulus of 0.5s was used as leakage current detection. Test data is collected by Qpatch and stored in the connected service station.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (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

本发明公开了含氮杂环类化合物、其制备方法及其应用。本发明提供了一种式(II)所示的化合物。该类化合物对TLR7/8具有较好的抑制活性;

Description

含氮杂环类化合物、其制备方法及其应用
本申请要求申请日为2022年08月01日的中国专利申请202210917037.5的优先权。本申请引用上述中国专利申请的全文。本申请要求申请日为2022年12月26日的中国专利申请202211680354.6的优先权。本申请引用上述中国专利申请的全文。
技术领域
本发明涉及含氮杂环类化合物、其制备方法及其应用。
背景技术
自身免疫疾病是一系列慢性全身性炎症性疾病,其特征是免疫系统失调,最终导致对自身抗原的耐受性下降。尽管这些疾病的确切病因和发病机制仍不清楚,但先天和适应性免疫系统的异常过程已证明参与这些疾病的发生。研究表明Toll样受体(Toll-like receptors,TLRs)在各种自身免疫性疾病中起着重要作用,包括干燥综合征,系统性红斑狼疮,多发性硬化症,类风湿性关节炎,系统性硬化症和牛皮癣等(Liu Y.,等人,Clin Rev Allergy Immunol.,2014,47,136-147;Thwaites R.,等人,Front Immunol.,2014,5)。
Toll样受体(Toll like receptors,TLR)是一类进化保守的跨膜先天免疫受体,它们参与保护人体健康的第一线防御,对于识别病原体相关分子模式(PAMP)起重要作用(Kawai,T.,等人,Nature Immunol.,2010,11,373-384)。TLR表达于各种免疫细胞,根据其表达部位的不同可分为两类:表达于细胞膜的TLR(TLR1/2/4/5/6)和表达于内体膜的TLR(TLR3/7/8/9),分别识别PAMP中的不同成分和分子。其中,TLR7/8/9主要在DC细胞和B细胞中高表达,TLR7/8主要识别ssRNA,TLR9主要识别CpG-DNA。TLR7/8/9结合其配体后被激活,在胞质中与接头蛋白MyD88相结合,启动NF-κB和IRF通路,活化DC细胞,产生I型干扰素和其它多种炎性细胞因子。在B细胞中,TLR7/8/9和核酸类物质相结合后,在B细胞产生抗核抗体的过程中起重要作用,并且DC细胞分泌的I型干扰素还会促进这种自身免疫性B细胞的进一步增殖和活化,从而引起一系列的炎症反应。
红斑狼疮是一种慢性且易反复发作的自身免疫性结缔组织疾病,除了对患者皮肤造成损害之外,还会累及患者的多种内脏器官和结缔组织。据美国狼疮基金会的统计数据显示,截至目前美国已有超150万人患有该疾病,而在全球范围至少有超500万名患者。
红斑狼疮可分为系统性红斑狼疮(SLE)、盘状红斑狼疮、亚急性皮肤型红斑狼疮以及深部红斑狼疮等类型。其中,SLE最为常见,约占所有患者的70%左右,同时也是红斑狼疮中最为严重的类型,临床表现包括疼痛、皮疹、疲劳、发热、关节肿胀等一系列症状,容易复发病变损伤各种器官,超一半的SLE患者会出现永久性器官损伤,因此SLE还有“不死癌症”之称。
针对系统性红斑狼疮,传统的治疗手段主要是使用免疫抑制剂、糖皮质激素以及抗疟药物等,但绝大多数都难以治愈且对患者造成很大伤害,目前仍然缺乏有效的根治手段。
因此,迫切需要改善更大比例的患者群体的新疗法,并且可以长期、安全使用。在系统性红斑狼 疮(SLE)患者的PBMC中发现了TLR7/9以及I型干扰素的表达被明显上调的现象(Beverly D.LC等人,Mol Immunol.,2014,61:38-43)。据报道过度表达TLR7的小鼠可以加剧自身免疫疾病和自身炎症(Santiago-Raber ML,等人,J Immunol.,2008,181:1556-1562),功能性抑制TLR7/9可以缓解B6-Fasl p r和BXSB等狼疮小鼠的病理表现(Dwight H.Kono,等人,PNAS,2009,106(29):12061-12066)。此外,有报道TLR8与类风湿关节炎有关(Enevold C.,等人,J Rheumatol.,2010,37,905-910)。但令人惊讶的是,单独的TLR9基因敲除小鼠表现出疾病症状的恶化,导致一些关于TLR9信号传导与自身免疫性疾病的关系的问题。因此调节TLR7和/TLR8活性的化合物以及使用这些化合物的方法可为多种自身免疫性患者提供实质的治疗益处。
专利文献WO2018005586A1、WO2018026620A1、WO2018047081A1、WO2018049089A1、WO2019028301A1、WO2020086503A1、WO2021067326A1、WO2022022489A1公布报道了一系列TLR7/8抑制剂。作为潜在治疗自身免疫疾病靶点,有必要开发更多TLR7/8抑制剂。
发明内容
本发明所要解决的技术问题现有的TLR7/8抑制剂结构单一,为此,本发明提供了一种含氮杂环类化合物、其制备方法及其应用。该类化合物对TLR7/8具有较好的抑制活性。
本发明提供了一种如式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物:
其中,R1
Ra独立地为C1~C6烷基或C3~C6环烷基;
X、Y、Z和W独立地为-C(Ra1)(Ra2)-、=CRa3-、-C(=O)-、-NRa4-、=N-、-O-或-S-;
Ra1、Ra2、Ra3和Ra4独立地为氢、C1~C3烷基、卤素或被1个、2个或3个卤素取代的C1~C3烷 基;
U独立地为N或CH;
为单键或者双键,相邻的两个不同时为双键;
环A为3~10元杂碳环或3~10元碳环;
L1为单键、C1~C6亚烷基或-(C=O)-;
L2为单键、C1~C6亚烷基或-(C=O)-;
R2为氢、羟基、氰基、C1~C6烷基、3~10元环烷基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、被1个、2个或3个R21取代的3~10元杂环烷基、被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R23取代的C1~C6烷基或被1个、2个或3个R26取代的5~6元杂芳基;
R21、R22和R23独立地为羟基、氰基、氧代、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢或C1~C6烷基;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C1~C6烷基、C3~C6环烷基、R271-(C=O)-或被1个、2个或3个卤素取代的C1~C6烷基,R271独立地为C1~C6烷基;
所述的3~10元杂碳环中,杂原子的数量为1个、2个或3个,杂原子选自N、O和S中的一种或多种;
所述的3~10元杂环烷基中,杂原子的数量独立地为1个、2个或3个,杂原子独立地选自N、O和S中的一种或多种;
所述的5~6元杂芳基中,杂原子的数量独立地为1个、2个、3个或4个,杂原子独立地选自N、O和S中的一种或多种;
R3为C1~C6烷基或C3~C6环烷基;
RA为氢或卤素;
V为C-R4或N,R4为氢、C1~C3烷基、C3~C6环烷基或卤素。
在某一方案中,所述如式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物:
其中,R1
Ra独立地为C1~C6烷基或C3~C6环烷基;
X、Y、Z和W独立地为-C(Ra1)(Ra2)-、=CRa3-、-C(=O)-、-NRa4-、=N-、-O-或-S-;
Ra1、Ra2、Ra3和Ra4独立地为氢、C1~C3烷基、卤素或被1个、2个或3个卤素取代的C1~C3烷基;
U独立地为N或CH;
为单键或者双键,相邻的两个不同时为双键;
环A为3~10元杂碳环;
L1为单键、C1~C6亚烷基或-(C=O)-;
L2为单键、C1~C6亚烷基或-(C=O)-;
R2为氢、羟基、氰基、C1~C6烷基、3~10元环烷基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、被1个、2个或3个R21取代的3~10元杂环烷基、被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R23取代的C1~C6烷基或被1个、2个或3个R26取代的5~6元杂芳基;
R21、R22和R23独立地为羟基、氰基、氧代、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢或C1~C6烷基;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C1~C6烷基、C3~C6环烷基、R271-(C=O)-或被1个、2个或3个卤素取代的C1~C6烷基,R271独立地为C1~C6烷基;
所述的3~10元杂碳环中,杂原子的数量为1个、2个或3个,杂原子选自N、O和S中的一种或多种;
所述的3~10元杂环烷基中,杂原子的数量独立地为1个、2个或3个,杂原子独立地选自N、O和S中的一种或多种;
所述的5~6元杂芳基中,杂原子的数量独立地为1个、2个、3个或4个,杂原子独立地选自N、O和S中的一种或多种;
R3为C1~C6烷基或C3~C6环烷基;
RA为氢或卤素;
V为C-R4或N,R4为氢、C1~C3烷基、C3~C6环烷基或卤素。
在某一方案中,所述的如式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物里,所述的如式II所示的化合物中某些基团的定义如下所述,其余基团的定义如其他任一方案所述(以下简称“在某一方案中”):
其中,R1
Ra独立地为C1~C6烷基或C3~C6环烷基;
X、Y、Z和W独立地为-C(Ra1)(Ra2)-、=CRa3-、-C(=O)-、-NRa4-、=N-、-O-或-S-;
Ra1、Ra2、Ra3和Ra4独立地为氢、C1~C3烷基、卤素或被1个、2个或3个卤素取代的C1~C3烷基;
U独立地为N或CH;
为单键或者双键,相邻的两个不同时为双键;
环A为3~10元杂碳环;
L1为单键、C1~C6亚烷基或-(C=O)-;
L2为单键、C1~C6亚烷基或-(C=O)-;
R2为氢、羟基、氰基、C1~C6烷基、3~10元环烷基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、被1个、2个或3个R21取代的3~10元杂环烷基、被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R23取代的C1~C6烷基或被1个、2个或3个R26取代的5~6元杂芳基;
R21、R22和R23独立地为羟基、氰基、氧代、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢或C1~C6烷基;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C1~C6烷基、C3~C6环烷基或R271-(C=O)-,R271独立地为C1~C6烷基;
所述的3~10元杂碳环中,杂原子的数量为1个、2个或3个,杂原子选自N、O和S中的一种或多种;
所述的3~10元杂环烷基中,杂原子的数量独立地为1个、2个或3个,杂原子独立地选自N、O和S中的一种或多种;
所述的5~6元杂芳基中,杂原子的数量独立地为1个、2个、3个或4个,杂原子独立地选自N、O和S中的一种或多种;
R3为C1~C6烷基或C3~C6环烷基;
R4为氢、C1~C3烷基、C3~C6环烷基或卤素。
在某一方案中,Ra中,所述C1~C6烷基可独立地为C1~C3烷基;又可独立地为甲基、乙基、正丙基或异丙基,例如甲基。
在某一方案中,Ra中,所述C3~C6环烷基可独立地为C3~C4环烷基;又可独立地为环丙基。
在某一方案中,Ra1、Ra2、Ra3或Ra4中,所述C1~C3烷基可独立地为甲基。
在某一方案中,Ra1、Ra2、Ra3或Ra4中,所述被1个、2个或3个卤素取代的C1~C3烷基中的C1~C3烷基可独立地为甲基。
在某一方案中,Ra1、Ra2、Ra3或Ra4中,所述卤素可独立地为氟。
在某一方案中,环A中,所述3~10元杂碳环可为6~8元杂碳环。
在某一方案中,环A中,所述3~10元杂碳环可为单环或双环;所述双环可为螺环或桥环,例如桥环。
在某一方案中,环A中,所述3~10元杂碳环中,杂原子的数量可为1个或2个。
在某一方案中,环A中,所述3~10元杂碳环中,所述杂原子可为N。
在某一方案中,环A中,所述3~10元杂碳环例如为
在某一方案中,环A中,所述3~10元碳环为3~6元碳环,例如环丙烷基环、环丁烷基环、环戊烷基环或环己烷基环,例如其中*标记碳原子表示当为手性碳原子时,为S构型、R构型或其混合物;#标记碳原子表示当为手性碳原子时,为S构型、R构型或其混合物,例如
在某一方案中,L1或L2中,所述C1~C6亚烷基可独立地为C1~C4亚烷基;又可独立地为亚甲基、亚乙基或所述q端与R2相连接。
在某一方案中,R2中,所述C1~C6烷基可为C1~C3烷基;又可为甲基、乙基、正丙基或异丙基。
在某一方案中,R2中,所述被1个、2个或3个R23取代的C1~C6烷基中的C1~C6烷基可为C1~C3烷基;又可为甲基、乙基、正丙基或异丙基。
在某一方案中,R2中,所述3~10元环烷基可为3~6元环烷基;又可为3~4元环烷基,例如环丙基或环丁基,又如环丙基。
在某一方案中,R2中,所述被1个、2个或3个R22取代的3~10元环烷基中的3~10元环烷基可为3~6元环烷基;又可为3~4元环烷基,例如环丙基或环丁基,又如环丙基。
在某一方案中,R2中,所述3~10元杂环烷基可为单环或双环;所述双环可为螺环或桥环,例如所述3~10元杂环烷基为单环。在某一方案中,R2中,所述3~10元杂环烷基中,杂原子的数量可为1个或2个。
在某一方案中,R2中,所述3~10元杂环烷基中,杂原子可独立地为N或O。
在某一方案中,R2中,所述3~10元杂环烷基可为3~6元杂环烷基;又可为4~6元杂环烷基;又可为
在某一方案中,R2中,所述被1个、2个或3个R21取代的3~10元杂环烷基中的3~10元杂环烷基可为单环或双环;所述双环可为螺环或桥环,例如所述3~10元杂环烷基为单环。
在某一方案中,R2中,所述被1个、2个或3个R21取代的3~10元杂环烷基中的3~10元杂环烷基中,杂原子的数量可为1个或2个。
在某一方案中,R2中,所述被1个、2个或3个R21取代的3~10元杂环烷基中的3~10元杂环烷基中,杂原子可独立地为N或S。
在某一方案中,R2中,所述被1个、2个或3个R21取代的3~10元杂环烷基中的3~10元杂环烷基可为3~6元杂环烷基;又可为4~6元杂环烷基;又可为
在某一方案中,R2中,所述3~10元杂环烷基和被1个、2个或3个R21取代的3~10元杂环烷基中的3~10元杂环烷基各自独立地为
在某一方案中,R2中,所述5~6元杂芳基,杂原子的数量可为2个。
在某一方案中,R2中,所述5~6元杂芳基可为
在某一方案中,R2中,所述被1个、2个或3个R26取代的5~6元杂芳基中的5~6元杂芳基中,杂原子的数量可为3个或4个。
在某一方案中,R2中,所述被1个、2个或3个R26取代的5~6元杂芳基中的5~6元杂芳基中,杂原子可为N。
在某一方案中,R2中,所述被1个、2个或3个R26取代的5~6元杂芳基中的5~6元杂芳基可为
在某一方案中,R2中,所述5~6元杂芳基和被1个、2个或3个R26取代的5~6元杂芳基中的5~6元杂芳基各自独立地为
在某一方案中,R21中,所述C1~C6烷基可独立地为C1~C3烷基;又可独立地为甲基、乙基、正丙基或异丙基,例如甲基。
在某一方案中,R23中,所述卤素可独立地为氟。
在某一方案中,R24中,所述C1~C6烷基可为C1~C3烷基;又可为甲基、乙基、正丙基或异丙基,例如甲基。
在某一方案中,R241或R241a中,所述C1~C6烷基可独立地为C1~C3烷基;又可独立地为甲基、乙基、正丙基或异丙基。
在某一方案中,R25中,所述C1~C6烷基可为C1~C3烷基;又可为甲基、乙基、正丙基或异丙基,例如甲基。
在某一方案中,R26中,所述C1~C6烷基可独立地为C1~C3烷基;又可独立地为甲基、乙基、正丙基或异丙基,例如甲基。
在某一方案中,R27或R27a中,所述C1~C6烷基可独立地为C1~C3烷基;又可独立地为甲基、乙基、正丙基或异丙基,例如甲基。
在某一方案中,R27或R27a中,所述C3~C6环烷基可独立地为C3~C4环烷基;又可独立地为环丙基。
在某一方案中,R27或R27a中,所述卤素可为F。
在某一方案中,R27或R27a中,所述被1个、2个或3个卤素取代的C1~C6烷基中C1~C6烷基可独立地为C1~C3烷基;又可独立地为乙基。
在某一方案中,R271中,所述C1~C6烷基可独立地为C1~C3烷基;又可独立地为甲基、乙基、正丙基或异丙基,例如甲基。
在某一方案中,R3中,所述C1~C6烷基可为C1~C3烷基;又可为甲基、乙基、正丙基或异丙基,例如异丙基。
在某一方案中,R3中,所述C3~C6环烷基可为C3~C4环烷基;又可为环丙基。
在某一方案中,RA中,所述卤素可为氟、氯或溴,又可为氟。
在某一方案中,R4中,所述C1~C3烷基可为甲基、乙基、正丙基或异丙基。
在某一方案中,R4中,所述C3~C6环烷基可为C3~C4环烷基;又可为环丙基。
在某一方案中,R4中,所述卤素可为氟。
在某一方案中,R1
其中,Ra独立地为C1~C6烷基或C3-6环烷基;
X、Y、Z和W独立地为-C(Ra1)(Ra2)-、=CRa3-、-C(=O)-、-NRa4-、=N-、-O-或-S-;
Ra1、Ra2、Ra3和Ra4独立地为氢、C1~C3烷基、卤素或被1个、2个或3个卤素取代的C1~C3烷基;
U独立地为N或CH;
为单键或者双键,相邻的两个不同时为双键。
在某一方案中,R1例如
在某一方案中,环A为3~10元杂碳环。
在某一方案中,环A为3~10元碳环。
在某一方案中,L1为单键、C1~C6亚烷基或-(C=O)-。
在某一方案中,L2为单键、C1~C6亚烷基或-(C=O)-。
在某一方案中,R2为氢、羟基、氰基、C1~C6烷基、3~10元环烷基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、被1个、2个或3个R21取代的3~10元杂环烷基、被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R23取代的C1~C6烷基或被1个、2个或3个R26取代的5~6元杂芳基;
其中,R21、R22和R23独立地为羟基、氰基、氧代、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢或C1~C6烷基;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C1~C6烷基、C3~C6环烷基或R271-(C=O)-,R271独立地为C1~C6烷基。在某一方案中,R3为C1~C6烷基或C3-6环烷基。
在某一方案中,R4为氢、C1~C3烷基、C3-6环烷基或卤素。
在某一方案中,RA为氢或卤素。
在某一方案中,RA为卤素。
在某一方案中,R4为氢或卤素。
在某一方案中,R4为氢。
在某一方案中,R1
Ra为C1~C6烷基;
X、Y和Z独立地为-C(Ra1)(Ra2)-、=CRa3-、-NRa4-、-O-、-S-或=N-;
Ra1、Ra2、Ra3和Ra4独立地为氢或C1~C3烷基;
U独立地为N或CH;
为单键或者双键,相邻的两个不同时为双键;
环A为3~10元杂碳环或3~10元碳环;
L1为单键、C1~C6亚烷基或-(C=O)-;
L2为单键、C1~C6亚烷基或-(C=O)-;
R2为氢、羟基、氰基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R26取代的5~6元杂芳基或被1个、2个或3个R21取代的3~10元杂环烷基;
R21和R22独立地为羟基、氧代、氰基、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C3~C6环烷基、C1~C6烷基、被1个、2个或3个卤素取代的C1~C6烷基或R271-(C=O)-,R271独立地为C1~C6烷基;
所述的3~10元杂碳环中,杂原子的数量为1个或2个,杂原子为N;
所述的3~10元杂环烷基中,杂原子的数量独立地为1个或2个,杂原子独立地选自N、O和S中的一种或多种;
所述的5~6元杂芳基中,杂原子的数量为2个至4个,杂原子独立地选自N、O和S中的一种或多种;
V为C-R4或N;
RA为氢或卤素;
R3为C1~C6烷基;
R4为氢。
在某一方案中,R1
Ra为甲基;
X、Y和Z独立地为-C(Ra1)(Ra2)-、=CRa3-、-NRa4-、-O-、-S-、=N-、或-C(=O)-;
Ra1、Ra2、Ra3和Ra4独立地为氢或C1~C3烷基;
U独立地为N或CH;
为单键或者双键,相邻的两个不同时为双键;
环A为3~10元杂碳环;
L1为单键、C1~C6亚烷基或-(C=O)-;
L2为单键、C1~C6亚烷基或-(C=O)-;
R2为氢、羟基、氰基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R26取代的5~6元杂芳基或被1个、2个或3个R21取代的3~10元杂环烷基;
R21和R22独立地为羟基、氧代、氰基、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C3~C6环烷基、C1~C6烷基、被1个、2个或3个卤素取代的C1~C6烷基或R271-(C=O)-,R271独立地为C1~C6烷基;
所述的3~10元杂碳环中,杂原子的数量为1个或2个,杂原子为N;
所述的3~10元杂环烷基中,杂原子的数量独立地为1个或2个,杂原子独立地选自N、O和S中的一种或多种;
所述的5~6元杂芳基中,杂原子的数量为2个,杂原子独立地选自N、O和S中的一种或多种;
V为C-R4或N;
RA为氢或卤素;
R3为C1~C6烷基;
R4为氢。
在某一方案中,R1
Ra为C1~C6烷基;
X、Y和Z独立地为-C(Ra1)(Ra2)-、=CRa3-、-NRa4-或-O-;
Ra1、Ra2、Ra3和Ra4独立地为氢或C1~C3烷基;
U独立地为N或CH;
为单键或者双键,相邻的两个不同时为双键;
环A为3~10元杂碳环;
L1为单键、C1~C6亚烷基或-(C=O)-;
L2为单键、C1~C6亚烷基或-(C=O)-;
R2为氢、羟基、氰基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、 被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R26取代的5~6元杂芳基或被1个、2个或3个R21取代的3~10元杂环烷基;
R21和R22独立地为氧代、氰基、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C3~C6环烷基、C1~C6烷基或R271-(C=O)-,R271独立地为C1~C6烷基;
所述的3~10元杂碳环中,杂原子的数量为1个,杂原子为N;
所述的3~10元杂环烷基中,杂原子的数量独立地为1个或2个,杂原子独立地选自N、O和S中的一种或多种;
所述的5~6元杂芳基中,杂原子的数量为2个,杂原子独立地选自N、O和S中的一种或多种;
R3为C1~C6烷基;
R4为氢。
在某一方案中,R1例如,R1
又如,R1
在某一方案中,Ra为C1~C6烷基,例如甲基。
在某一方案中,X、Y和Z独立地为-C(Ra1)(Ra2)-、=CRa3-、-NRa4-、-O-、-S-或=N-;
例如,X、Y和Z独立地为-C(Ra1)(Ra2)-、=CRa3-、-NRa4-、-O-、-S-、=N-或-C(=O)-;
又如,X、Y和Z独立地为-C(Ra1)(Ra2)-、=CRa3-、-NRa4-或-O-。
在某一方案中,Ra1、Ra2、Ra3和Ra4独立地为氢或C1~C3烷基。
在某一方案中,R2为氢、羟基、氰基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R26取代的5~6元杂芳基或被1个、2个或3个R21取代的3~10元杂环烷基。
在某一方案中,R21和R22独立地为羟基、氧代、氰基、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独 立地为氢、C3~C6环烷基、C1~C6烷基、被1个、2个或3个卤素取代的C1~C6烷基或R271-(C=O)-,R271独立地为C1~C6烷基;
例如,R21和R22独立地为氧代、氰基、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C3~C6环烷基、C1~C6烷基或R271-(C=O)-,R271独立地为C1~C6烷基。
在某一方案中,所述的5~6元杂芳基中,杂原子的数量为2、3或4个,杂原子独立地选自N、O和S中的一种或多种。
在某一方案中,V为C-R4,又如为N。
在某一方案中,RA例如为氢,又如为卤素。
在某一方案中,R3为C1~C6烷基。
在某一方案中,R4为H。
在某一方案中,R1中,中,Ra可为C1~C6烷基或C3-6环烷基;X、Y和Z可独立地为-C(Ra1)(Ra2)-、=CRa3-、-C(=O)-、-NRa4-、=N-、-O-或-S-;Ra1和Ra2可独立地为氢或卤素;Ra3可为氢或卤素;Ra4可为氢、C1~C3烷基或三氟甲基。
在某一方案中,R1中,中,Ra可为C1~C3烷基或C3-6环烷基;X、Y和Z可独立地为-C(Ra1)(Ra2)-、=CRa3-、-C(=O)-、-NRa4-、=N-或-O-;Ra1和Ra2可独立地为氢或氟;Ra3可为氢或氟;Ra4可为氢、甲基或三氟甲基。
在某一方案中,R1中,中,Ra可为甲基、异丙基或环丙基;X、Y和Z可独立地为-C(Ra1)(Ra2)-、=CRa3-、-C(=O)-、-NRa4-、=N-或-O-;Ra1和Ra2可独立地为氢或氟;Ra3可为氢或氟;Ra4可为氢、甲基或三氟甲基。
在某一方案中,R1中,所述可为
在某一方案中,R1中,中,X、Y和Z可独立地为-C(Ra1)(Ra2)-、=N-、-O-或-S-;Ra1和Ra2可独立地为氢。
在某一方案中,R1中,中,X、Y和Z可独立地为-C(Ra1)(Ra2)-、-O-或-S-;Ra1和Ra2可独立地为氢。
在某一方案中,R1中,可为
在某一方案中,R1中,中,X、Y和Z可独立地为=CRa3-或-NRa4-;Ra3和Ra4可独立地为氢、C1~C3烷基、三氟甲基或卤素。
在某一方案中,R1中,可为
在某一方案中,R1中,中,X、Y和Z可独立地为-C(Ra1)(Ra2)-或=CRa3-;Ra1、Ra2和Ra3可独立地为氢、C1~C3烷基、三氟甲基或卤素。
在某一方案中,R1中,可为
在某一方案中,R1中,中,X、Y和Z可独立地为=CRa3-或-NRa4-;Ra3和Ra4可独立地为氢、C1~C3烷基、三氟甲基或卤素。
在某一方案中,R1中,可为
在某一方案中,R1中,中,X、Y和Z可独立地为-C(Ra1)(Ra2)-、=CRa3-、-C(=O)-、-NRa4-、=N-、-O-或-S-;Ra1和Ra2可独立地为氢或卤素;Ra3可为氢;Ra4可为氢、C1~C3烷基或三氟甲基。
在某一方案中,R1中,中,X、Y和Z可独立地为-C(Ra1)(Ra2)-、=CRa3-、-C(=O)-、-NRa4-、=N-、-O-或-S-;Ra1和Ra2可独立地为氢或氟;Ra3可为氢;Ra4可为氢、甲基或三氟甲基。
在某一方案中,R1中,可为
在某一方案中,R1中,中,X、Y、Z和W可独立地为-C(Ra1)(Ra2)-、=N-、-O-或-S-;Ra1和Ra2可独立地为氢;Ra可为C1~C3烷基。
在某一方案中,R1中,中,X、Y、Z和W可独立地为-C(Ra1)(Ra2)-、=N-、-O-或-S-;Ra1和Ra2可独立地为氢;Ra可为甲基。
在某一方案中,R1中,可为
在某一方案中,R1可为
在某一方案中,R1可为
在某一方案中,R1可为
在某一方案中,R1中,可为
在某一方案中,R1中,Ra可为C1~C3烷基,又可为甲基、乙基、正丙基或异丙基,还可为甲基。
在某一方案中,R1中,可为
在某一方案中,L1可为C1~C6亚烷基或-(C=O)-。
在某一方案中,L1可为单键。
在某一方案中,L1为单键、亚甲基、亚乙基、-(C=O)-或所述q端与R2相连接;
在某一方案中,L2可为C1~C6亚烷基或-(C=O)-。
在某一方案中,L2可为单键。
在某一方案中,L2为单键、亚甲基、亚乙基、-(C=O)-或所述q端与R2相连接;
在某一方案中,L1和L2不同时为C1~C6亚烷基,且L1和L2不同时为-(C=O)-。
在某一方案中,环A可为6~8元杂碳环。
在某一方案中,环A中杂原子的数量可为1个或2个,杂原子可为N。
在某一方案中,环A可为6~8元杂碳环,其中杂原子的数量可独立地为1个或2个,杂原子可为N。
在某一方案中,环A可为其中s端与L1相连接。
在某一方案中,环A可为其中s端与L1相连接。
在某一方案中,R2可为氢、3~10元杂环烷基、被1个、2个或3个R21取代的3~10元杂环烷基、被1个、2个或3个R22取代的3~10元环烷基或被1个、2个或3个R23取代的C1~C6烷基;R21可为氧代,R22可为氰基,R23可为卤素。
在某一方案中,R2可为氢、3~6元杂环烷基、被1个、2个或3个R21取代的3~6元杂环烷基、被1个、2个或3个R22取代的3~4元环烷基或被1个、2个或3个R23取代的C1~C6烷基;R21可为氧代,R22可为氰基,R23可为卤素。
在某一方案中,R2可为氢、4~6元杂环烷基、被1个、2个或3个R21取代的4~6元杂环烷基、被1个、2个或3个R22取代的4元环烷基或被1个、2个或3个R23取代的C1~C3烷基;R21可为氧代,R22可为氰基,R23可为卤素。
在某一方案中,R2可为氢、
在某一方案中,R2可为羟基、被1个、2个或3个R21取代的3~10元杂环烷基、被1个、2个或3个R22取代的3~10元环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基或被1个、2个或3个R26取代的5~6元杂芳基;R21可为C1~C6烷基或氧代;R22可为氰基;R24可为C1~C6烷基、(R241)(R241a)N-,R241和R241a可独立地为氢或C1~C6烷基;R25可为C1~C6烷基;R26可为C1~C6烷基。
在某一方案中,R2可为羟基、被1个、2个或3个R21取代的3~6元杂环烷基、被1个、2个或3个R22取代的3~6元环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基或被1个、2个或3个R26取代的5~6元杂芳基;R21可为C1~C3烷基或氧代;R22可为氰基;R24可为C1~C3烷基、(R241)(R241a)N-,R241和R241a可独立地为氢或C1~C3烷基;R25可为C1~C3烷基;R26可为C1~C3烷基。
在某一方案中,R2可为羟基、被1个、2个或3个R21取代的5~6元杂环烷基、被1个、2个或3个R22取代的3元环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5元杂芳基或被1个、2个或3个R26取代的5元杂芳基;R21可为C1~C3烷基或氧代;R22可为氰基;R24可为C1~C3烷基、(R241)(R241a)N-,R241和R241a可独立地为氢或C1~C3烷基;R25可为C1~C3烷基;R26可为C1~C3烷基。
在某一方案中,R2可为羟基、被1个、2个或3个R21取代的5~6元杂环烷基、被1个、2个或3个R22取代的3元环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5元杂芳基或被1个、2个或3个R26取代的5元杂芳基;R21可为甲基或氧代;R22可为氰基;R24可为甲基、(R241)(R241a)N-,R241和R241a可独立地为氢或甲基;R25可为甲基;R26可为甲基。
在某一方案中,R2可为羟基、
在某一方案中,R2可为氰基、三氟甲基、3~10元杂环烷基、被1个、2个或3个R21取代的3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、被1个、2个或3个R26取代的5~6元杂芳基或(R27)(R27a)N-;R21可为羟基、氧代或卤素;R24可为C1~C6烷基;R25可为C1~C6烷基;R26可为C1~C6烷基;R27和R27a可独立地为氢、C1~C6烷基、C3~C6环烷基或R271-(C=O)-,R271可为C1~C6烷基。
在某一方案中,R2可为氰基、三氟甲基、3~6元杂环烷基、被1个、2个或3个R21取代的3~6元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、被1个、2个或3个R26取代的5~6元杂芳基或(R27)(R27a)N-;R21可为羟基、氧代或卤素;R24可为C1~C3烷基;R25可为C1~C3烷基;R26可为C1~C3烷基;R27和R27a可独立地为氢、C1~C3烷基、C3~C6环烷基或R271-(C=O)-,R271可为C1~C3烷基。
在某一方案中,R2可为氰基、三氟甲基、4~6元杂环烷基、被1个、2个或3个R21取代的4~6元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、被1个、2个或3个R26取代的5元杂芳基或(R27)(R27a)N-;R21可为羟基、氧代或卤素;R24可为C1~C3烷基;R25可为C1~C3烷基;R26可为C1~C3烷基;R27和R27a可独立地为氢、C1~C3烷基、C3~C6环烷基或R271-(C=O)-,R271可为C1~C3烷基。
在某一方案中,R2可为氰基、三氟甲基、4~6元杂环烷基、被1个、2个或3个R21取代的4~6元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、被1个、2个或3个R26取代的5元杂芳基或(R27)(R27a)N-;R21可为羟基、氧代或卤素;R24可为甲基;R25可为甲基;R26可为甲基;R27和R27a可独立地为氢、甲基、异丙基、环丙基或R271-(C=O)-,R271可为甲基。
在某一方案中,R2可为氰基、三氟甲基、
在某一方案中,R2可为氰基、三氟甲基、
在某一方案中,R2可为氢、羟基、氰基、三氟甲基、
在某一方案中,R2可为氢、羟基、氰基、三氟甲基、
在某一方案中,R2为氢、羟基、氰基、
在某一方案中,L1可为单键;L2可为单键;R2可为氢、3~10元杂环烷基、被1个、2个或3个R21取代的3~10元杂环烷基、被1个、2个或3个R22取代的3~10元环烷基或被1个、2个或3个R23取代的C1~C6烷基;R21可为氧代,R22可为氰基,R23可为卤素。
在某一方案中,L1可为单键;L2可为单键;R2可为氢、3~6元杂环烷基、被1个、2个或3个R21取代的3~6元杂环烷基、被1个、2个或3个R22取代的3~4元环烷基或被1个、2个或3个R23取代的C1~C6烷基;R21可为氧代,R22可为氰基,R23可为卤素。
在某一方案中,L1可为单键;L2可为单键;R2可为氢、4~6元杂环烷基、被1个、2个或3个R21取代的4~6元杂环烷基、被1个、2个或3个R22取代的4元环烷基或被1个、2个或3个R23取代的C1~C3烷基;R21可为氧代,R22可为氰基,R23可为卤素。
在某一方案中,L1可为单键;L2可为单键;R2可为氢、
在某一方案中,L1可为单键;L2可为C1~C6亚烷基或-(C=O)-;R2可为羟基、被1个、2个或3个R21取代的3~10元杂环烷基、被1个、2个或3个R22取代的3~10元环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基或被1个、2个或3个R26取代的5~6元杂芳基;R21可为C1~C6烷基或氧代;R22可为氰基;R24可为C1~C6烷基、(R241)(R241a)N-,R241和R241a可独立地为氢或C1~C6烷基;R25可为C1~C6烷基;R26可为C1~C6烷基。
在某一方案中,L1可为单键;L2可为C1~C6亚烷基或-(C=O)-;R2可为羟基、被1个、2个或3个R21取代的3~6元杂环烷基、被1个、2个或3个R22取代的3~6元环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基或被1个、2个或3个R26取代的5~6元杂芳基;R21可为C1~C3烷基或氧代; R22可为氰基;R24可为C1~C3烷基、(R241)(R241a)N-,R241和R241a可独立地为氢或C1~C3烷基;R25可为C1~C3烷基;R26可为C1~C3烷基。
在某一方案中,L1可为单键;L2可为C1~C6亚烷基或-(C=O)-;R2可为羟基、被1个、2个或3个R21取代的5~6元杂环烷基、被1个、2个或3个R22取代的3元环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5元杂芳基或被1个、2个或3个R26取代的5元杂芳基;R21可为C1~C3烷基或氧代;R22可为氰基;R24可为C1~C3烷基、(R241)(R241a)N-,R241和R241a可独立地为氢或C1~C3烷基;R25可为C1~C3烷基;R26可为C1~C3烷基。
在某一方案中,L1可为单键;L2可为C1~C6亚烷基或-(C=O)-;R2可为羟基、被1个、2个或3个R21取代的5~6元杂环烷基、被1个、2个或3个R22取代的3元环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5元杂芳基或被1个、2个或3个R26取代的5元杂芳基;R21可为甲基或氧代;R22可为氰基;R24可为甲基、(R241)(R241a)N-,R241和R241a可独立地为氢或甲基;R25可为甲基;R26可为甲基。
在某一方案中,L1可为单键;L2可为C1~C6亚烷基或-(C=O)-;R2可为羟基、
在某一方案中,L1可为C1~C6亚烷基或-(C=O)-;L2可为C1~C6亚烷基或-(C=O)-,其中,L1和L2不同时为C1~C6亚烷基,且L1和L2不同时为-(C=O)-;R2可为氰基、三氟甲基、3~10元杂环烷基、被1个、2个或3个R21取代的3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、被1个、2个或3个R26取代的5~6元杂芳基或(R27)(R27a)N-;R21可为羟基、氧代或卤素;R24可为C1~C6烷基;R25可为C1~C6烷基;R26可为C1~C6烷基;R27和R27a可独立地为氢、C1~C6烷基、C3~C6环烷基或R271-(C=O)-,R271可为C1~C6烷基。
在某一方案中,L1可为C1~C6亚烷基或-(C=O)-;L2可为C1~C6亚烷基或-(C=O)-,其中,L1和L2不同时为C1~C6亚烷基,且L1和L2不同时为-(C=O)-;R2可为氰基、三氟甲基、3~6元杂环烷基、被1个、2个或3个R21取代的3~6元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、被1个、2个或3个R26取代的5~6元杂芳基或(R27)(R27a)N-;R21可为羟基、氧代或卤素;R24可为C1~C3烷基;R25可为C1~C3烷基;R26可为C1~C3烷基;R27和R27a可独立地为氢、C1~C3烷基、C3~C6环烷基或R271-(C=O)-,R271可为C1~C3烷基。
在某一方案中,L1可为C1~C6亚烷基或-(C=O)-;L2可为C1~C6亚烷基或-(C=O)-,其中,L1和L2不同时为C1~C6亚烷基,且L1和L2不同时为-(C=O)-;R2可为氰基、三氟甲基、4~6元杂环烷基、被1个、2个或3个R21取代的4~6元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、被1个、2个或3个R26取代的5元杂芳基或(R27)(R27a)N-;R21可为羟基、氧代或卤素;R24可为C1~C3烷基;R25可为C1~C3烷基;R26可为C1~C3烷基;R27和R27a可独立地为氢、C1~C3烷基、C3~C6环烷基或R271-(C=O)-,R271可为C1~C3烷基。
在某一方案中,L1可为C1~C6亚烷基或-(C=O)-;L2可为C1~C6亚烷基或-(C=O)-,其中,L1和L2 不同时为C1~C6亚烷基,且L1和L2不同时为-(C=O)-;R2可为氰基、三氟甲基、4~6元杂环烷基、被1个、2个或3个R21取代的4~6元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、被1个、2个或3个R26取代的5元杂芳基或(R27)(R27a)N-;R21可为羟基、氧代或卤素;R24可为甲基;R25可为甲基;R26可为甲基;R27和R27a可独立地为氢、甲基、异丙基、环丙基或R271-(C=O)-,R271可为甲基。
在某一方案中,L1可为C1~C6亚烷基或-(C=O)-;L2可为C1~C6亚烷基或-(C=O)-,其中,L1和L2不同时为C1~C6亚烷基,且L1和L2不同时为-(C=O)-;R2可为氰基、三氟甲基、
在某一方案中,L1为单键、亚甲基、亚乙基或-(C=O)-或所述q端与L2相连接。
在某一方案中,L1单键、亚甲基、亚乙基或-(C=O)-或所述q端与R2相连接。
在某一方案中,可为下述结构中的任意一种

在某一方案中,可为下述结构中的任意一种
在某一方案中,可为下述结构中的任意一种
在某一方案中,为H、
在某一方案中,R2可为氢、羟基、3~10元杂环烷基、5~6元杂芳基、被1个、2个或3个R21取代的3~10元杂环烷基、R24-S(=O)2-或(R27)(R27a)N-;R21可为C1~C6烷基、氧代或卤素;R24可为C1~C6烷基或(R241)(R241a)N-,R241和R241a可独立地为氢或C1~C6烷基;R27和R27a可独立地为氢或C1~C6烷基。
在某一方案中,R2可为氢或3~10元杂环烷基。
在某一方案中,R2可为氢或3~6元杂环烷基。
在某一方案中,R2可为氢或4~6元杂环烷基。
在某一方案中,R2可为氢或
在某一方案中,R2可为羟基、5~6元杂芳基、被1个、2个或3个R21取代的3~10元杂环烷基或R24-S(=O)2-;R21可为C1~C6烷基;R24可为C1~C6烷基或(R241)(R241a)N-,R241和R241a可独立地为氢或C1~C6烷基。
在某一方案中,R2可为羟基、5~6元杂芳基、被1个、2个或3个R21取代的3~6元杂环烷基或R24-S(=O)2-;R21可为C1~C3烷基;R24可为C1~C3烷基或(R241)(R241a)N-,R241和R241a可独立地为氢或C1~C3烷基。
在某一方案中,R2可为羟基、5元杂芳基、被1个、2个或3个R21取代的5~6元杂环烷基或R24- S(=O)2-;R21可为甲基;R24可为甲基或(R241)(R241a)N-,R241和R241a可独立地为氢或甲基。
在某一方案中,R2可为羟基、
在某一方案中,R2可为3~10元杂环烷基、被1个、2个或3个R21取代的3~10元杂环烷基或(R27)(R27a)N-;R21可为氧代或卤素;R27和R27a可独立地为氢或C1~C6烷基。
在某一方案中,R2可为3~6元杂环烷基、被1个、2个或3个R21取代的3~6元杂环烷基或(R27)(R27a)N-;R21可为氧代或卤素;R27和R27a可独立地为氢或C1~C3烷基。
在某一方案中,R2可为4~6元杂环烷基、被1个、2个或3个R21取代的4~6元杂环烷基或(R27)(R27a)N-;R21可为氧代或卤素;R27和R27a可独立地为氢或甲基。
在某一方案中,R2可为
在某一方案中,R2可为氢、羟基、
在某一方案中,L1可为单键;L2可为单键;R2可为氢或3~10元杂环烷基。
在某一方案中,L1可为单键;L2可为单键;R2可为氢或3~6元杂环烷基。
在某一方案中,L1可为单键;L2可为单键;R2可为氢或4~6元杂环烷基。
在某一方案中,L1可为单键;L2可为单键;R2可为氢或
在某一方案中,L1可为单键;L2可为C1~C6亚烷基或-(C=O)-;R2可为羟基、5~6元杂芳基、被1个、2个或3个R21取代的3~10元杂环烷基或R24-S(=O)2-;R21可为C1~C6烷基;R24可为C1~C6烷基或(R241)(R241a)N-,R241和R241a可独立地为氢或C1~C6烷基。
在某一方案中,L1可为单键;L2可为C1~C6亚烷基或-(C=O)-;R2可为羟基、5~6元杂芳基、被1个、2个或3个R21取代的3~6元杂环烷基或R24-S(=O)2-;R21可为C1~C3烷基;R24可为C1~C3烷基或(R241)(R241a)N-,R241和R241a可独立地为氢或C1~C3烷基。
在某一方案中,L1可为单键;L2可为C1~C6亚烷基或-(C=O)-;R2可为羟基、5元杂芳基、被1个、2个或3个R21取代的5~6元杂环烷基或R24-S(=O)2-;R21可为甲基;R24可为甲基或(R241)(R241a)N-,R241和R241a可独立地为氢或甲基。
在某一方案中,L1可为单键;L2可为C1~C6亚烷基或-(C=O)-;R2可为羟基、
在某一方案中,L1可为C1~C6亚烷基或-(C=O)-;L2可为C1~C6亚烷基或-(C=O)-,其中,L1和L2 不同时为C1~C6亚烷基,且L1和L2不同时为-(C=O)-;R2可为3~10元杂环烷基、被1个、2个或3个R21取代的3~10元杂环烷基或(R27)(R27a)N-;R21可为氧代或卤素;R27和R27a可独立地为氢或C1~C6烷基。
在某一方案中,L1可为C1~C6亚烷基或-(C=O)-;L2可为C1~C6亚烷基或-(C=O)-,其中,L1和L2不同时为C1~C6亚烷基,且L1和L2不同时为-(C=O)-;R2可为3~6元杂环烷基、被1个、2个或3个R21取代的3~6元杂环烷基或(R27)(R27a)N-;R21可为氧代或卤素;R27和R27a可独立地为氢或C1~C3烷基。
在某一方案中,L1可为C1~C6亚烷基或-(C=O)-;L2可为C1~C6亚烷基或-(C=O)-,其中,L1和L2不同时为C1~C6亚烷基,且L1和L2不同时为-(C=O)-;R2可为4~6元杂环烷基、被1个、2个或3个R21取代的4~6元杂环烷基或(R27)(R27a)N-;R21可为氧代或卤素;R27和R27a可独立地为氢或甲基。
在某一方案中,L1可为C1~C6亚烷基或-(C=O)-;L2可为C1~C6亚烷基或-(C=O)-,其中,L1和L2不同时为C1~C6亚烷基,且L1和L2不同时为-(C=O)-;R2可为
在某一方案中,R3可为C1~C3烷基或C3-6环烷基。
在某一方案中,R3可为异丙基或环丙基。
在某一方案中,R4可为氢或甲基。
在某一方案中,所述式II所示的化合物可为下述化合物中的任意一种:










某一方案中,所述式II所示的化合物为中在以下条件先出峰的一个立体异构体:色谱柱:Ultimate XB-C18,流动相:含0.1v%甲酸的乙腈和水混合溶液;洗脱梯度为:0-3min内流动相中乙腈的体积百分比为10%,3-16min内,流动相中乙腈的体积百分比由25%上升至45%,16-20min内,流动相中乙腈的体积百分比为95%;较佳的,为在下述条件下保留时间为4.03min的一个立体异构体,所述条件为:色谱柱:Ultimate XB-C18,21.2*250mm,10μm;流动相:含0.1v%甲酸的乙腈和水混合溶液;流动相:含0.1v%甲酸的乙腈和水混合溶液;洗脱梯度为:0-3min内流动相中乙腈的体积百分比为10%,3-16min内,流动相中乙腈的体积百分比由25%上升至45%,16-20min内,流动相中乙腈的体积百分比为95%;流速:20ml/min;柱温:30℃。
某一方案中,所述式II所示的化合物为中在以下条件后出峰的 一个立体异构体:色谱柱:Ultimate XB-C18,流动相:含0.1v%甲酸的乙腈和水混合溶液;洗脱梯度为:0-3min内流动相中乙腈的体积百分比为10%,3-16min内,流动相中乙腈的体积百分比由25%上升至45%,16-20min内,流动相中乙腈的体积百分比为95%;较佳的,为在下述条件下保留时间为4.10min的一个立体异构体,所述条件为:色谱柱:Ultimate XB-C18,21.2*250mm,10μm;流动相:含0.1v%甲酸的乙腈和水混合溶液;流动相:含0.1v%甲酸的乙腈和水混合溶液;洗脱梯度为:0-3min内流动相中乙腈的体积百分比为10%,3-16min内,流动相中乙腈的体积百分比由25%上升至45%,16-20min内,流动相中乙腈的体积百分比为95%;流速:20ml/min;柱温:30℃。
本发明还提供了一种药物组合物,其包括物质X和药用辅料;
所述的物质X为上述的如式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物。
本发明还提供了一种物质X在制备TLR7和/或TLR8抑制剂中的应用;
所述的物质X为上述的如式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物。
在某一方案中,相对于TLR9,所述的TLR7和/或TLR8抑制剂可具有选择性。
在某一方案中,所述的TLR7和/或TLR8抑制剂可在体外使用。
本发明还提供了一种物质X在制备药物中的应用;
所述的物质X为上述的如式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物;
所述的药物为用于治疗与TLR7和/或TLR8相关疾病的药物。
在某一方案中,所述的与TLR7和/或TLR8相关疾病可为自身免疫性疾病,又可为干燥综合征、红斑狼疮、多发性硬化症、类风湿性关节炎、系统性硬化症或牛皮癣。
在某一方案中,所述的红斑狼疮可为系统性红斑狼疮(SLE)、盘状红斑狼疮或亚急性皮肤型红斑狼疮。
本发明还提供了一种物质X在制备用于治疗自身免疫性疾病的药物中的应用;
所述的物质X为上述的如式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物;
在某一方案中,所述的自身免疫性疾病可为干燥综合征、红斑狼疮、多发性硬化症、类风湿性关节炎、系统性硬化症或牛皮癣。
在某一方案中,所述的红斑狼疮可为系统性红斑狼疮(SLE)、盘状红斑狼疮或亚急性皮肤型红斑狼疮。
定义
除非另有说明,本发明所用的下列术语旨在具有下列定义。一个特定的术语在没有特别定义的情况下不应该被认为是不清楚的,而应该按照普通的含义去理解。
术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学 判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其他问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”是指本发明化合物的盐,由本发明发现的具有特定取代基的化合物与相对无毒的酸或碱制备。当本发明的化合物中含有相对酸性的功能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的碱与这类化合物中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钠、钾钙、铵、有机胺或镁盐或类似的盐。当本发明的化合物中含有相对碱性的官能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的酸与这类化合物中的游离形式接触的方式获得碱加成盐。药学上可接受的酸加成盐包括的实例包括无机酸盐,所述无机酸包括例如盐酸、氢溴酸、硝酸、碳酸、碳酸氢根、磷酸、磷酸一氢根、磷酸二氢根、硫酸、硫酸氢根、氢碘酸、亚磷酸等,以及有机酸盐,所述有机酸包括如乙酸、丙酸、异丁酸、马来酸、丙二酸、苯甲酸、琥珀酸、辛二酸、反丁烯二酸、乳酸、扁桃酸、邻苯二甲酸、苯磺酸、对甲苯磺酸、柠檬酸、酒石酸和甲磺酸等类似的酸;还包括氨基酸(如精氨酸等)的盐,以及如葡糖醛酸等有机酸的盐。本发明的某些特定的化合物含有碱性和酸性的官能团,从而可以被转换为任一碱或酸加成盐。
本发明的药学上可接受的盐可由含有酸根或碱基的母体化合物通过常规的化学方法合成。一般情况下,这样的盐的制备方法是:在水或有机溶剂或两者的混合物中,经由游离酸或碱形式的这些化合物与化学计量的适当的碱或酸反应来制备。
是指基团通过该位点与化合物其他部分连接。
基团末端的“-”是指该基团通过该位点与分子其余部分相连。例如,CH3-C(=O)-是指乙酰基。
环上原子的数目通常被定义为环的元数,例如,“3-7元环”是指环绕排列3-7个原子的“环”。
术语“卤素”是指氟、氯、溴和碘。
术语“烷基”是指具有指定碳原子数(例如,C1-C6)的、直链或支链的、饱和的一价烃基。烷基包括但不限于:甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基等。
术语“亚烷基”为二价基团,其通过两个单键与分子其余部分相连,其余定义同术语“烷基”。
术语“环烷基”是指具有指定碳原子数(例如,C3-C10)的、环状的、饱和的一价烃基,其为其为单环体系或双环体系,所述双环可为螺环或桥环,所述螺环是指单环之间共用一个碳原子,所述桥环是指单环之间共用两个以上碳原子。环烷基包括但不限于: 等。
术语“碳环”满足下述任一条件,其余定义同术语“环烷基”:1、通过两个以上的单键与分子其余部分相连;2、与分子其余部分共用两个原子和一根键;术语“碳环”为饱和的环。
术语“杂环烷基”是指具有指定环原子数(例如,3-10元)的、指定杂原子数(例如,1个、2个 或3个)的、指定杂原子种类(N、O和S中的一种或多种)的、环状的、饱和的一价基团,其为单环体系或双环体系,所述双环可为螺环或桥环,所述螺环是指单环之间共用一个碳原子,所述桥环是指单环之间共用两个以上碳原子。杂环烷基通过碳原子或杂原子与分子其余部分相连。杂环烷基包括但不限于: 等。
术语“杂碳环”通过两个以上的单键与分子其余部分相连,其余定义同术语“杂环烷基”。术语“杂碳环”为饱和的环。
术语“5-6元杂芳基”本身或者与其他术语联合分别表示由5至6个环原子组成的具有共轭π电子体系的环状基团,其1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子。5-6元杂芳基可通过杂原子或碳原子连接到分子的其余部分,所述5-6元杂芳基包括5元和6元杂芳基等。所述5-6元杂芳基的实例包括但不限于吡咯基(包括N-吡咯基、2-吡咯基和3-吡咯基等)、吡唑基(包括2-吡唑基和3-吡唑基等)、咪唑基(包括N-咪唑基、2-咪唑基、4-咪唑基和5-咪唑基等)、噁唑基(包括2-噁唑基、4-噁唑基和5-噁唑基等)、三唑基(1H-1,2,3-三唑基、2H-1,2,3-三唑基、1H-1,2,4-三唑基和4H-1,2,4-三唑基)、四唑基、异噁唑基(包括3-异噁唑基、4-异噁唑基和5-异噁唑基等)、噻唑基(包括2-噻唑基、4-噻唑基和5-噻唑基等)、呋喃基(包括2-呋喃基和3-呋喃基等)、噻吩基(包括2-噻吩基和3-噻吩基等)、吡啶基(包括2-吡啶基、3-吡啶基和4吡啶基等)、吡嗪基、嘧啶基(包括2-嘧啶基和4-嘧啶基等)等。
术语“治疗”指给患有疾病或者具有所述疾病的症状的个体施用一种或多种药物物质、特别是本发明所述的式(I)化合物和或其药学上可接受的盐,用以治愈、缓解、减轻、改变、医治、改善、改进或影响所述疾病或者所述疾病的症状。
当涉及化学反应时,术语“处理”、“接触”和“反应”指在适当的条件下加入或混合两种或更多种试剂,以产生所示的和或所需的产物应当理解的是,产生所示的和/或所需的产物的反应可能不一定直接来自最初加入的两种试剂的组合,即,在混合物中可能存在生成的一个或多个中间体,这些中间体最终导致了所示的和或所需的产物的形成。
如本发明所用,“患者”定义为任何温血动物,例如不限于小鼠、豚鼠、狗、马或人,所述患者最好是人。
本发明所用的未具体定义的技术和科学术语具有本发明所属领域的技术人员通常理解的含义。
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:该类化合物对TLR7/8具有较好的抑制活性。
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。
如无特殊说明,实施例中反相制备纯化条件为:制备柱:Ultimate XB-C18,21.2*250mm,10μm;洗脱相:流动相0.1v%FAin H2O/ACN,洗脱梯度(ACN的比例)10v%(0-3min),25-45v%(3-16min),95v%(16-20min),流速20mL/min,柱温30℃,实施例1-76中反相制备纯化步骤参考此条件。
实施例1
4-(3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊[c]吡啶-1-酮
步骤A:5-溴-3-异丙基-1H-吲哚(化合物1.2)
将三乙基硅烷(8.72g,76.53mmol),三氟乙酸(8.88g,76.53mmol)溶解于80mL甲苯中,室温下加入5-溴-1H-吲哚(5.0g,25.51mmol),丙酮(3.70g,63.78mmol)的甲苯溶液(30mL),70℃搅拌2.5小时。反应完全后,向反应液中加NaHCO3溶液淬灭,乙酸乙酯萃取,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析(EA/PE(0-10%))纯化得到产物1.2(5.0g,产率:83.3%)。LCMS ESI(+)m/z:238.0(M+1)。
步骤B:4-(3-异丙基-1H-吲哚-5-基)-5,6-二氢吡啶-1(2H)-羧酸叔丁酯(化合物1.3)
将1.2(3.91g,16.50mmol)溶解于60mL和10mL水中,加入4-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-5,6-二氢吡啶-1(2H)-羧酸叔丁酯(5.36g,17.32mmol),Pd(dppf)Cl2(0.61g,0.82mmol)和碳酸铯(16.14g,49.50mmol),反应液在80℃下搅拌5小时。反应完全后,将反应液过滤,滤液中加乙酸乙酯萃取,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析(EA/PE(0-10%))纯化得到产物1.3(3.39g,产率:60%)。LCMS ESI(+)m/z:341.2(M+1)。
步骤C:4-(3-异丙基-1H-吲哚-5-基)哌啶-1-羧酸叔丁酯(化合物1.4)
将1.3(3.39g,83.35mmol)溶解于60mL乙酸乙酯中,加入钯碳(0.848g),反应液在室温下搅拌5小时。将反应液过滤,乙酸乙酯萃取,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析(EA/PE(0-10%))纯化得到产物1.4(2.70g,产率:79%)。LCMS ESI(+)m/z:343.2(M+1)。
步骤D:4-(2-溴-3-异丙基-1H-吲哚-5-基)哌啶-1-羧酸叔丁酯(化合物1.5)
将1.4(2.70g,7.89mmol)溶解于75mL二氯乙烷中,冰水浴下滴加NBS(1.33g,7.50mmol)的二氯乙烷溶液(75mL),反应液在0-10℃下搅拌0.5小时。将反应液加Na2SO3溶液淬灭,二氯甲烷萃取,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析(EA/PE(0-10%))纯化得到产物1.5(2.45g,产率:73%)。LCMS ESI(+)m/z:421/423(M+1)。
步骤E:4-(3-异丙基-2-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吲哚-5-基)哌啶-1-羧酸叔丁酯(化合物1.6)
将1.5(1.68g,4.0mmol)溶解于33mL 1,4-二氧六环中,加入二苯甲腈二氯化钯(46mg,0.12mmol),S-Phos(197mg,0.48mmol),三乙胺(1.21g,12.0mmol),反应液在85℃下搅拌3小时。将反应液旋干,乙酸乙酯萃取,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析(EA/PE(0-7%))纯化得到产物1.6(0.534g,产率:29%)。LCMS ESI(+)m/z:469.2(M+1)。
步骤F:2-((三氟甲基)磺酰基)氧基)环戊-1-烯-1-羧酸甲酯(化合物1.8)
将2-氧环戊烷-1-羧酸甲酯(10g,70.35mmol)和N,N-二异丙基乙胺(13.64g,105.53mmol)溶于二氯甲烷(100mL)中。三氟甲磺酸酐(21.83g,77.38mmol)溶于二氯甲烷(50mL)中,在-25℃下将该溶液缓慢滴至反应液中。滴毕,反应液继续在-25-0℃下搅拌反应2小时。反应液用饱和碳酸氢钠溶液淬灭,二氯甲烷萃取2次,合并有机相,有机相食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,柱层析(PE/EA(4%))纯化得到产物1.8(16.0g,产率:83%)。LCMS ESI(+)m/z:275(M+1)。
步骤G:2-(三甲基硅基)乙炔基)环戊-1-烯-1-羧酸甲酯(化合物1.9)
将化合物1.8(16.0g,58.36mmol)、三甲基乙炔基硅(11.46g,116.72mmol)、双三苯基膦二氯化钯(4.10g,5.84mmol)、碘化亚铜(1.11g,5.84mmol)和三乙胺(17.70g,175.09mmol)溶于DMF(200mL)中。反应体系用氩气置换3次。反应液60℃下搅拌反应1小时。反应液用乙酸乙酯和水萃取两次。合并的有机相用饱和食盐水洗涤并用无水硫酸钠干燥,过滤,减压浓缩,柱层析(PE/EA(4%))纯化得到产物1.9(8.0g,产率:61.7%)。LCMS ESI(+)m/z:223(M+1)。
步骤H:2-乙基环戊-1-烯-1-羧酸(化合物1.10)
将化合物1.9(8g,36.01mmol)溶解于甲醇(80mL)和四氢呋喃(80mL)中,加入氢氧化锂 溶液(3.02g,72.02mmol,72ml)),反应液在50℃下搅拌反应2小时。反应液减压浓缩,柱层析(DCM/MeOH(10%))纯化得到产物1.10(4.8g,产率:98%)。LCMS ESI(+)m/z:137(M+1)。
步骤I:2-乙基环戊-1-烯-1-甲酰胺(化合物1.11)
将化合物1.10(2.72g,20.0mmol)和N,N-二异丙基乙胺(7.75g,60.0mmol)溶解在N,N-二甲基甲酰胺(50mL)中。室温下加入HATU(11.4g,30.0mmol),反应液在室温下搅拌0.5小时,继续往反应液中加入氯化铵(3.21g,60.0mmol),室温下搅拌过夜。向反应液中加入水,用乙酸乙酯萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析(PE/EA(40%))纯化得到产物1.11(2.45g,产率:90%)。LCMS ESI(+)m/z:136(M+1)。
步骤J:2,5,6,7-四氢-1H-环戊[c]吡啶-1-酮(化合物1.12)
将化合物1.11(2.45g,18.13mmol)溶解在二甲胺甲醇溶液(2M,12mL)中。反应液在100℃微波搅拌反应2小时。反应液减压浓缩,柱层析(PE/EA(4%))纯化得到产物1.12(1.65g,产率:67.3%)。LCMS ESI(+)m/z:136(M+1)。
步骤K:2-甲基-2,5,6,7-四氢-1H-环戊[c]吡啶-1-酮(化合物1.13)
将化合物1.12(675mg,5.0mmol)溶解在N,N-二甲基甲酰胺(10mL)中。0℃下将氢化钠(240mg,60%,6.0mmol)缓慢加至反应液中搅拌反应30分钟。然后将碘甲烷滴至反应液中继续在室温下搅拌反应2小时。反应液用氯化铵溶液淬灭,乙酸乙酯萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析(DCM/MeOH(10%))纯化得到产物1.13(650mg,产率:88%)。LCMS ESI(+)m/z:150(M+1)。
步骤L:4-溴-2-甲基-2,5,6,7-四氢-1H-环戊[c]吡啶-1-酮(化合物1.14)
将化合物1.13(639mg,4.28mmol)溶解在乙腈(10mL)中。室温下将NBS(800mg,4.50mmol)加至反应液中搅拌反应3小时。反应液减压浓缩,柱层析(DCM/MeOH(5%))纯化得到产物1.14(980mg,产率:100%)。LCMS ESI(+)m/z:228/230(M+1)。
步骤M:叔丁基4-(3-异丙基-2-(2-甲基-1-氧基-2,5,6,7-四氢-1H-环戊烷[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-羧酸盐(化合物1.15)
将化合物1.14(120mg,0.52mmol)、4-(3-异丙基-2-(4,4,5,5-四甲基-1,3,2-二氧苯甲醛-2-基)-1H-吲哚-5-基)哌啶-1-羧酸叔丁酯(240mg,0.51mmol)、磷酸钾(326mg,1.53mmol)、[1,1'-双(二苯基膦)二茂铁]二氯化钯(37mg,0.051mmol)溶解在1,4-二氧六环(10mL)和水(2mL)中。反应体系氩气置换3次。反应液85℃下搅拌反应2小时。反应液减压浓缩,柱层析(DCM/MeOH(5%))纯化得到产物1.15(270mg,粗品产率:108%)。LCMS ESI(+)m/z:490(M+1)。
步骤N:4-(3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊[c]吡啶-1-酮(化合物1)
将化合物1.15(270mg,0.55mmol)溶解在无水甲醇(3mL)和二氯甲烷(1mL)中。室温下将氯化氢-二氧六环溶液(1mL)滴至反应体系中。反应液室温下下搅拌反应1小时。反应液减压浓缩,柱层析(DCM/MeOH/NH3·H2O(10:1:0.1))纯化得到产物1(156mg,产率:73%)。LCMS ESI(+)m/z:390(M+1)。1H NMR(400MHz,DMSO)δ10.77(s,1H),8.65(s,1H),7.56(s,1H),7.49(s,1H),7.25(d,J =8.3Hz,1H),6.94(dd,J=8.4,1.2Hz,1H),3.51(s,3H),3.37(s,2H),3.05–2.93(m,3H),2.88(ddd,J=15.6,10.4,5.6Hz,1H),2.72(dd,J=16.0,8.0Hz,4H),1.94(qd,J=15.2,5.6Hz,6H),1.35(d,J=7.2Hz,6H).
实施例2
2-(4-(3-异丙基-2-(2-甲基-1-氧基-2,5,6,7-四氢-1H-环戊烷[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺
将化合物1(40mg,0.10mmol)和DBU(61mg,0.40mmol)溶于1,2-二氯乙烷(3mL)中,然后将2-溴乙酰胺(17mg,0.12mmol)加至反应体系中。反应液继续在室温下下搅拌反应18小时。反应液用二氯甲烷萃取2次,合并有机相,有机相食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,反相制备纯化得到产物2(20mg,产率:44%)。LCMS ESI(+)m/z:447(M+1),224(M/2+1)。1H NMR(400MHz,DMSO)δ10.68(s,1H),7.55(s,1H),7.49(s,1H),7.26(s,1H),7.21(d,J=8.4Hz,1H),7.14(s,1H),6.97(dd,J=8.4,1.2Hz,1H),3.50(s,3H),2.99(d,J=7.2Hz,1H),2.95(d,J=12.8Hz,2H),2.90(s,2H),2.75–2.68(m,5H),2.20(dd,J=11.2,8.8Hz,2H),2.03–1.93(m,2H),1.81(dt,J=25.2,10.0Hz,4H),1.35(d,J=7.2Hz,6H).
实施例3
2-(4-(3-异丙基-2-(2-甲基-1-氧基-2,5,6,7-四氢-1H-环戊[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)-N-甲基乙酰胺
将化合物1(15mg,0.04mmol)和2-氯-N-甲基乙酰胺(13mg,0.12mmol)在氮气保护下溶解于5mL THF中,加入DIEA(52mg,0.4mmol),反应液在70℃下搅拌过夜。向反应液中加入水20mL,用乙酸乙酯(25mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤, 减压浓缩,用反相制备纯化得到产物3(8mg,产率:44%)。LCMS ESI(+)m/z:461.2(M+1)。1H NMR(400MHz,DMSO-d6)δ10.69(s,1H),7.72(q,J=4.8Hz,1H),7.55(s,1H),7.49(s,1H),7.21(d,J=8.3Hz,1H),6.97(d,J=8.3Hz,1H),3.50(s,3H),3.01–2.96(m,1H),2.92(s,3H),2.89(s,1H),2.72(q,J=7.7Hz,4H),2.65(d,J=4.7Hz,3H),2.56–2.52(m,1H),2.18(td,J=11.5,2.9Hz,2H),1.98(q,J=7.5Hz,2H),1.80(dtd,J=22.4,12.8,3.7Hz,4H),1.35(d,J=7.0Hz,6H).
实施例4
2-(4-(3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊二烯[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)-N,N-二甲基乙酰胺
将化合物1(40mg,0.103mmol)溶解于2mL THF和1mL DMF中,加入2-氯-N,N-二甲基乙酰胺(19mg,0.154mmol),TEA(104mg,1.03mmol),反应液在室温下搅拌过夜。将反应液旋干,用二氯甲烷萃取,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,用反相制备纯化得到产物4(14.5mg,产率:30%)。LCMS ESI(+)m/z:475.3(M+1)。1H NMR(400MHz,DMSO)δ10.70(s,1H),7.55(s,1H),7.49(s,1H),7.22(d,J=8.3Hz,1H),6.96(dd,J=8.4,1.3Hz,1H),3.50(s,3H),3.45(s,2H),3.11(d,J=9.3Hz,2H),3.03(d,J=9.2Hz,3H),2.98(dd,J=14.1,7.0Hz,1H),2.85(s,3H),2.72(dd,J=16.0,8.0Hz,4H),2.61(dd,J=16.8,9.4Hz,1H),2.41(s,2H),2.03–1.92(m,2H),1.82(s,4H),1.34(d,J=7.0Hz,6H).
实施例5
4-(5-(1-(2-(1,1-二氧化硫代吗啉代)-2-氧代乙基)哌啶-4-基)-3-异丙基-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊[c]吡啶-1-酮
步骤A:乙基2-(4-(3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊a[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)乙酸酯(化合物5.1)
将化合物1(50mg,0.118mmol)溶于3mL二氯甲烷中,加入DIEA(109mg,0.846mmol)和2-溴乙酸乙酯(24mg,0.141mmol),用氩气置换三次,反应液在室温条件下搅拌过夜。向反应液中加入水5mL,用二氯甲烷(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到化合物5.1(65mg,产率:99.3%)。LCMS ESI(+)m/z:476.3(M+1)。
步骤B:2-(4-(3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊二烯[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)乙酸(化合物5.2)
将化合物5.1(65mg,0.126mmol)溶于THF(1mL)和MeOH(1mL)中,加入NaOH(100mg)和1.5mL水,反应液在室温条件下搅拌2小时。向反应液中加入盐酸溶液至中性,用iPrOH:DCM=1:3萃取三次,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩得到化合物5.2(60mg,产率:106%)。LCMS ESI(+)m/z:448.2(M+1)。
步骤C:4-(5-(1-(2-(1,1-二氧化硫代吗啉代)-2-氧代乙基)哌啶-4-基)-3-异丙基-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊[c]吡啶-1-酮(化合物5)
将化合物5.2(30mg,0.067mmol)溶解在3mL乙腈中,加入HATU(38.4mg,0.101mmol),TEA(20.3mg,0.201mmol)和硫代吗啉1,1-二氧化物(13.6mg,0.101mmol)反应液室温条件下搅拌过夜,用水淬灭,用二氯甲烷(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,用反相制备纯化得到化合物5(5.2mg,产率:13.7%)。LCMS ESI(+)m/z:565.2(M+1)。1H NMR(400MHz,DMSO)δ10.68(s,1H),7.55(s,1H),7.47(s,1H),7.21(d,J=8.3Hz,1H),6.96(d,J=8.5Hz,1H),4.01(s,2H),3.87(s,2H),3.50(s,3H),3.29–3.27(m,2H),3.25(s,2H),3.13(s,2H),2.97(dd,J=14.0,7.2Hz,3H),2.72(dd,J=15.6,7.8Hz,4H),2.55(s,1H),2.13(t,J=10.7Hz,2H),1.98(dd,J=15.0,7.6Hz,2H),1.80(d,J=10.6Hz,2H),1.70(t,J=10.5Hz,2H),1.34(d,J=7.0Hz,6H).
实施例6
4-(3-异丙基-5-(1-(恶唑-2-基甲基)哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊[c]吡啶-1-酮
将化合物1(40mg,0.1mmol)溶解于5mL DCM中,依次加入三乙胺(40mg,0.4mmol),噁唑-2-甲醛(39mg,0.4mmol),醋酸(32mg,0.2mmol)以及醋酸硼氢化钠(85mg,0.4mmol),反应液在室温下搅拌2小时。向反应液中加入饱和氯化铵水溶液15mL,用二氯甲烷(10mL X 3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到产物6(26mg,产率:56%)。LCMS ESI(+)m/z:471.3(M+1)。1H NMR(400MHz,CDCl3)δ8.07(s,1H),7.69(s,1H),7.60(s,1H),7.30(d,J=8.4Hz,1H),7.14(d,J=5.0Hz,2H),7.09(dd,J=8.4,1.3Hz,1H),3.89(s,2H),3.57(s,3H),3.18(s,2H),3.01(dt,J=14.1,7.0Hz,1H),2.88(t,J=7.4Hz,2H),2.73(t,J=7.5Hz,2H),2.64(t,J=11.9Hz,1H),2.42(s,2H),2.06–2.01(m,2H),1.95(d,J=11.7Hz,2H),1.71(s,2H),1.38(d,J=7.1Hz,6H).
实施例7
4-(3-异丙基-5-(1-(噻唑-2-基甲基)哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊二烯[c]吡啶-1-酮
将化合物1(30mg,0.077mmol)溶解在5mL二氯甲烷溶液中,加入三乙胺(31mg,0.31mmol)、2-醛基噻唑(35mg,0.31mmol)、乙酸(9.2mg,0.15mmol)、三乙酰氧基硼氢化钠(65.4mg,0.31,mmol),室温下搅拌过夜。旋干溶剂,反相制备纯化得到化合物7(18mg,产率:48%)。LCMS ESI(+)m/z:487.3(M+1).1H NMR(400MHz,DMSO)δ10.68(s,1H),7.72(d,J=3.3Hz,1H),7.66(d,J=3.3Hz,1H),7.56(s,1H),7.49(s,1H),7.21(d,J=8.3Hz,1H),6.97(dd,J=8.4,1.4Hz,1H),3.87(s,2H),3.50(s,3H),3.02(d,J=13.1Hz,2H),3.00–2.94(m,1H),2.72(d,J=7.5Hz,4H),2.63–2.54(m,1H),2.26(dd,J=11.5,8.8Hz,2H),2.02–1.93(m,2H),1.84–1.69(m,4H),1.34(d,J=7.0Hz,6H).
实施例8
4-(3-异丙基-5-(1-(2-(甲基磺酰基)乙基)哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊烷[c]吡啶-1-酮
将化合物1(40mg,0.094mmol)溶解于3mL二氯乙烷中,加入1-溴-2-(甲基磺酰基)乙烷(21mg,0.113mmol),DBU(57mg,0.37mmol),反应液在室温下搅拌过夜。将反应液用二氯甲烷萃取,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,用反相制备纯化得到产物8(38mg,产率:82%)。LCMS ESI(+)m/z:496.2(M+1)。
LCMS ESI(+)m/z:496.2(M+1)。1H NMR(400MHz,DMSO)δ10.68(s,1H),7.55(s,1H),7.47(s,1H),7.20(d,J=8.3Hz,1H),6.95(d,J=8.4Hz,1H),3.50(s,3H),3.31(s,2H),3.07(s,3H),3.04–2.93(m,3H),2.78–2.68(m,6H),2.56(dd,J=9.5,5.9Hz,1H),2.10(t,J=10.6Hz,2H),2.01–1.93(m,2H),1.79(d,J=11.3Hz,2H),1.68(dt,J=12.0,9.0Hz,2H),1.34(d,J=7.0Hz,6H).
实施例9
2-(4-(3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊二烯[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)乙磺酰胺
步骤A:2-氯乙磺酰胺(化合物9.2)
将2-氯乙磺酰氯(100mg,0.61mmol)溶解于5mL THF中,加入氨水(0.5mL),三乙胺(186mg,1.83mmol),反应液在室温下搅拌过夜。将反应液直接用于下一步反应。LCMS ESI(+)m/z:144(M+1)。
步骤B:2-(4-(3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊二烯[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)乙磺酰胺(化合物9)
将9.2(87mg,0.61mmol)溶解于5mL THF中,加入4-(3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊二烯[c]吡啶-1-酮(40mg,0.103mmol),反应液在室温下搅拌过夜。将反应液用乙酸乙酯萃取,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,用反相制备纯化得到产物9(12mg,产率:24%)。LCMS ESI(+)m/z:497(M+1)。1H NMR(400MHz,DMSO)δ10.67(s,1H),7.55(s,1H),7.47(s,1H),7.20(d,J=8.3Hz,1H),6.95(dd,J=8.4,1.3Hz,1H),6.79(s,2H),3.50(s,3H),3.21–3.17(m,2H),3.02–2.93(m,3H),2.73(dt,J=20.8,7.9Hz,6H),2.59–2.52(m,1H),2.11(dt,J=16.8,8.5Hz,2H),2.03–1.92(m,2H),1.83–1.66(m,4H),1.34(d,J=7.0Hz,6H).
实施例10
4-(3-异丙基-5-(1-(氧杂环丁烷-3-基)哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊二烯[c]吡啶-1-酮
将化合物1(30mg,0.077mmol)溶解在5mL二氯甲烷溶液中,加入三乙胺(31mg,0.31mmol)、3-氧杂环丁酮(22.2mg,0.31mmol)、乙酸(9.2mg,0.15mmol)、三乙酰氧基硼氢化钠(65.4mg,0.31,mmol),室温下搅拌过夜。旋干溶剂,反相制备纯化得到化合物10(22mg,产率:64%)。LCMS ESI(+)m/z:446.3(M+1).1H NMR(400MHz,DMSO)δ10.68(s,1H),7.55(s,1H),7.49(s,1H),7.21(d,J=8.3Hz,1H),6.96(dd,J=8.4,1.2Hz,1H),4.56(t,J=6.5Hz,2H),4.46(t,J=6.1Hz,2H),3.50(s,3H),3.42–3.39(m,1H),2.98(dt,J=14.0,7.0Hz,1H),2.81(d,J=10.9Hz,2H),2.72(q,J=7.3Hz,4H),2.55(t,J=4.1Hz,1H),1.98(dd,J=14.9,7.5Hz,2H),1.88(dd,J=11.3,9.2Hz,2H),1.74(dt,J=12.0,7.7Hz,4H),1.34(d,J=7.0Hz,6H).
实施例11
4-(5-(1-(二甲基甘氨酸)哌啶-4-基)-3-异丙基-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊[c]吡啶-1-酮
将二甲基甘氨酸(10mg,0.10mmol)、三乙胺(50mg,0.50mmol)溶于DMF(2mL)中,室温下加入HATU(38mg,0.10mmol)加至反应体系中搅拌反应0.5小时。然后加入化合物1(40mg,0.10mmol)继续在室温下搅拌反应18小时。反应液用乙酸乙酯萃取2次,合并有机相,有机相食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,反相制备纯化得到产物11(25mg,产率:53%)。LCMS ESI(+)m/z:475(M+1),238(M/2+1)。1H NMR(400MHz,DMSO)δ10.69(s,1H),7.55(s,1H),7.47(s,1H),7.21(d,J=8.4Hz,1H),6.94(dd,J=8.4,1.2Hz,1H),4.53(d,J=12.4Hz,1H),4.17(d,J=12.4Hz,1H),3.50(s,3H),3.20–3.12(m,2H),3.08(d,J=13.6Hz,1H),2.98(dt,J=14.0,6.9Hz,1H),2.82(t,J=12.0Hz,1H),2.75–2.69(m,3H),2.67–2.59(m,1H),2.22(s,6H),1.98(dq,J=15.2,7.6Hz,3H),1.83(d,J=12.4Hz,2H),1.72–1.60(m,1H),1.48(dd,J=12.8,4.0Hz,1H),1.34(d,J=7.2Hz,6H).
实施例12
4-(3-异丙基-5-(1-(甲基甘氨酰)哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊[c]吡啶-1-酮
步骤A:叔丁基(2-(4-(3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊二[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)-2-氧代乙基)(甲基)氨基甲酸酯(化合物12.1)
将化合物1(30mg,0.077mmol)溶解到5mL DMF中,加入HATU(44mg,0.115mmol),DIPEA(30mg,0.231mmol)和N-(叔丁氧羰基)-N-甲基甘氨酸(17.5mg,0.092mmol),反应液在室温搅拌2小时。向反应液中加入水,用乙酸乙酯萃取,有机相用饱和食盐水洗,无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到化合物12.1(37mg,产率:86%),LCMS ESI(+)m/z:561.3(M+1)。
步骤B:4-(3-异丙基-5-(1-(甲基甘氨酰)哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊[c] 吡啶-1-酮(化合物12)
将化合物12.1(37mg,0.066mmol)溶解在3mL甲醇中,加入4M盐酸甲醇溶液3mL,反应液在室温搅拌1.5小时。将反应液减压浓缩,剩余物用反相制备纯化得到产物12(20mg,产率:66%)。LCMS ESI(+)m/z:461.3(M+1)。1H NMR(400MHz,DMSO-d6)δ10.77(s,1H),8.92(s,1H),7.56(s,1H),7.48(s,1H),7.23(d,J=8.3Hz,1H),6.95(dd,J=8.4,1.2Hz,1H),4.54(s,1H),4.15(dt,J=15.8,5.5Hz,1H),4.05(dd,J=16.2,6.6Hz,1H),3.77(d,J=13.3Hz,1H),3.51(s,3H),3.18(t,J=12.0Hz,1H),2.98(p,J=7.0Hz,1H),2.94–2.84(m,1H),2.83–2.65(m,5H),2.58(t,J=5.4Hz,3H),1.97(p,J=7.6Hz,2H),1.88(d,J=13.1Hz,2H),1.76–1.63(m,1H),1.58–1.43(m,1H),1.34(d,J=7.0Hz,6H).
实施例13
4-(5-(1-(2-羟基-2-甲基丙基)哌啶-4-基)-3-异丙基-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊[c]吡啶-1-酮
将化合物1(15mg,0.04mmol)和1-氯-2-甲基丙烷-2-醇(217mg,2.0mmol)在氮气保护下溶解于5mL DMF中,加入DIEA(52mg,0.4mmol),反应液在120℃下搅拌过夜。向反应液中加入水20mL,用乙酸乙酯(25mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,用反相制备纯化得到产物13(2.5mg,产率:14%)。LCMS ESI(+)m/z:462.3(M+1)。1H NMR(400MHz,DMSO-d6)δ10.67(s,1H),7.55(s,1H),7.47(d,J=1.5Hz,1H),7.20(d,J=8.3Hz,1H),6.95(dd,J=8.4,1.5Hz,1H),3.50(s,3H),3.10–2.94(m,4H),2.72(q,J=7.8Hz,4H),2.30–2.20(m,4H),1.98(q,J=7.5Hz,2H),1.73(td,J=9.2,7.8,3.2Hz,4H),1.34(d,J=7.0Hz,6H),1.11(s,6H).
实施例14
4-(3-异丙基-5-(1-(2-吗啉代乙酰基)哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊二烯[c]吡啶-1-酮
将化合物1(70mg)溶解在10mL NMP溶液中,加入三乙胺(58mg,0.58mmol)、氯乙酰氯(33mg,0.29mmol),室温下搅拌1小时。加入DBU(147mg,0.78mmol)、吗啉(50mg,0.58mmol),室温下搅拌过夜。旋干溶剂,反相制备纯化得到化合物14(40mg,产率:40%)。LCMS ESI(+)m/z:517.3(M+1).1H NMR(400MHz,DMSO)δ10.70(s,1H),7.55(s,1H),7.47(s,1H),7.21(d,J=8.3Hz,1H),6.94(dd,J=8.4,1.2Hz,1H),4.52(d,J=12.6Hz,1H),4.17(d,J=13.0Hz,1H),3.59(t,J=4.4Hz,4H),3.50(s,3H),3.28(s,1H),3.08(d,J=13.3Hz,2H),2.98(dt,J=14.1,7.0Hz,1H),2.84(dd,J=16.0,8.0Hz,1H),2.72(d,J=7.7Hz,4H),2.62(d,J=10.8Hz,1H),2.43(d,J=2.8Hz,4H),2.04–1.92(m,2H),1.83(t,J=10.9Hz,2H),1.71(dd,J=12.3,3.5Hz,1H),1.46(dd,J=12.6,3.8Hz,1H),1.34(d,J=7.0Hz,6H).
实施例15
4-(3-异丙基-5-(1-(4-甲基哌嗪-1-羰基)哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊烷[c]吡啶-1-酮
步骤A:4-硝基苯基4-(3-异丙基-2-(2-甲基-1-氧基-2,5,6,7-四氢-1H-环戊烷[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-羧酸酯(化合物15.1)
将化合物1(50mg,0.12mmol)溶于无水四氢呋喃(2mL)中,在0℃下加入DIPEA(31mg,0.24mmol)和4-硝基苯氧基酰氯(36mg,0.18mmol)中,在室温下反应2小时。反应完成后,向反应液滴加饱和碳酸氢钠溶液淬灭反应,然后用DCM(20mL*3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱色谱纯化得到产物15.1(60mg,产率:90%)。LCMS ESI(+)m/z:555(M+1)。
步骤B:4-(3-异丙基-5-(1-(4-甲基哌嗪-1-羰基)哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊烷[c]吡啶-1-酮(化合物15)
将化合物15.1(35mg,0.06mmol)和1-甲基哌嗪(30mg,0.30mmol),碳酸钾(17mg,0.12mmol)加入到DMF(4mL)中,在150℃的微波下反应1.5小时。反应完成后,向反应液加入10mL水,然后用乙酸乙酯(20mL*3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤, 减压浓缩,反相制备纯化得到产物15(19mg,产率:41%)。LCMS ESI(+)m/z:516(M+1)。1H NMR(400MHz,d6-DMSO)δ10.69(s,1H),8.14(s,1H),7.55(s,1H),7.47(s,1H),7.21(d,J=8.3Hz,1H),6.95(dd,J=8.4,1.1Hz,1H),3.71(d,J=12.9Hz,2H),3.50(s,3H),3.17(s,4H),2.98(dt,J=14.1,7.0Hz,1H),2.85(t,J=11.9Hz,2H),2.72(dd,J=15.9,7.6Hz,5H),2.35(s,4H),2.21(s,3H),2.06–1.90(m,2H),1.79(d,J=11.0Hz,2H),1.71–1.54(m,2H),1.34(d,J=7.0Hz,6H).
实施例16
2-(4-(3-异丙基-2-(6-甲基-7-氧代-6,7-二氢-1H-吡咯[2,3-c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺
步骤A:5-溴-1,4-二甲基-3-硝基吡啶-2(1H)-酮(化合物16.2)
将5-溴-4-甲基-3-硝基吡啶-2(1H)-酮(500mg,1.28mmol)溶解于7mL DMF中,加入氢化钠(103mg,2.58mmol),反应液在室温下搅拌0.5小时,再加入碘甲烷(366mg,2.58mmol),反应液在室温下搅拌2小时。向反应液中加入水15mL,用乙酸乙酯(10mL X 3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到产物16.2(333mg,产率:63%)。LCMS ESI(+)m/z:247/249(M+1)。
步骤B:(E)-5-溴-4-(2-(二甲氨基)乙烯基)-1-甲基-3-硝基吡啶-2(1H)-酮(化合物16.3)
将化合物16.2(333mg,1.35mmol)用10mL DMF溶解,加入DMF-DMA(0.6mL),反应液在80℃下搅拌4小时。将反应液减压浓缩得到产物16.3(406mg,产率:100%)。LCMS ESI(+)m/z:302/304(M+1)。
步骤C:4-溴-6-甲基-1,6-二氢-7H-吡咯[2,3-c]吡啶-7-酮(化合物16.4)
将化合物16.3(406mg,1.35mmol)溶解于10mL醋酸中,加入铁粉(378mg,6.75mmol),反应液在110℃下搅拌过夜。反应液过滤,减压浓缩,加入水(10mL),用碳酸氢钠溶液调pH=7,加乙酸乙酯(3 x 35mL)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到产物16.4(118mg,产率:39%)。LCMS ESI(+)m/z:227/229(M+1)。
步骤D:4-(3-异丙基-2-(6-甲基-7-氧代-6,7-二氢-1H-吡咯[2,3-c]吡啶-4-基)-1H-吲哚-5-基)哌 啶-1-羧酸叔丁酯(化合物16.5)
将化合物16.4(50mg,0.22mmol)和4-(3-异丙基-2-(4,4,5,5-四甲基-1,3,2-二氧苯甲醛-2-基)-1H-吲哚-5-基)哌啶-1-羧酸叔丁酯(155mg,0.33mmol)溶解于5mL二氧六环和1mL水中,加入磷酸钾(140mg,0.66mmol)和PdCl2(dppf)(32mg,0.04mmol),反应液在85℃下搅拌2小时。反应液倒入水(10mL)中,二氯甲烷(3 x 35mL)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到产物16.5(105mg,产率:98%)。LCMS ESI(+)m/z:489.3(M+1)。
步骤E:4-(3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-6-甲基-1,6-二氢-7H-吡咯[2,3-c]吡啶-7-酮(化合物16.6)
将化合物16.5(105mg,0.22mmol)溶解于3mL甲醇,滴加HCl/MeOH(1mL),反应液在室温下搅拌3小时。反应液减压浓缩,得到产物16.6(85mg,产率:100%)。LCMS ESI(+)m/z:389.2(M+1)。
步骤F:2-(4-(3-异丙基-2-(6-甲基-7-氧代-6,7-二氢-1H-吡咯[2,3-c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺(化合物16)
将化合物16.6(47mg,0.12mmol)溶解于3mL二氯乙烷,滴加DBU(0.1mL)和2-溴乙酰胺(19mg,0.14mmol),反应液在室温下搅拌4.5小时。将反应液加入水(10mL)中,二氯甲烷(3 x 25mL)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到产物16(5.7mg,产率:10%)。LCMS ESI(+)m/z:446.2(M+1)。1H NMR(400MHz,DMSO)δ12.12(s,1H),10.78(s,1H),8.20(s,1H),7.52(s,1H),7.33(t,J=2.6Hz,1H),7.24(d,J=8.3Hz,2H),7.21(s,1H),7.13(s,1H),6.97(d,J=8.3Hz,1H),6.19(s,1H),3.59(s,3H),3.13(dt,J=13.1,6.6Hz,2H),2.94(d,J=11.0Hz,2H),2.89(s,2H),2.19(t,J=10.0Hz,2H),1.83(dd,J=22.6,12.8Hz,4H),1.37(d,J=7.0Hz,6H).
实施例17
2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑基[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酰胺
步骤A:吡啶-3,4-二甲酸二甲酯(化合物17.2)
将吡啶-3,4-二羧酸(2.00g,11.97mmol)加入到20mL氯化亚砜中,反应液在80℃反应过夜。减压浓缩,在0℃下加入到10mL甲醇中,反应液升温至室温,搅拌0.5小时。减压浓缩,将固体用乙酸乙酯溶解,用饱和碳酸氢钠溶液洗涤两次,有机相用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到产物17.2(2.00g,产率:85%),LCMS ESI(+)m/z:196.1(M+1)。
步骤B:3,4-双(甲氧羰基)吡啶1-氧化物(化合物17.3)
将化合物17.2(500mg,2.56mmol)溶解在7mL乙腈中,在0℃下加入尿素(308mg,5.13mmol)和30%过氧化氢(0.5mL,5.13mmol),缓慢加入三氟乙酸酐(1.08g,5.13mmol),反应液在室温下搅拌过夜。加入水稀释,用二氯甲烷(15mLx2)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到产物17.3(500mg,产率:93%)。LCMS ESI(+)m/z:212.0(M+1)。
步骤C:2-氯吡啶-3,4-二甲酸二甲酯(化合物17.4)
将化合物17.3(500mg,2.37mmol)加入到6mL三氯氧磷中,氮气保护下,110℃反应过夜。将反应液减压浓缩,剩余物滴加到水中,用碳酸氢钠溶液调节pH至8,用乙酸乙酯(15mLx2)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到产物17.4(200mg,产率:37%)。LCMS ESI(+)m/z:230.0(M+1)。
步骤D:(2-氯吡啶-3,4-二基)二甲醇(化合物17.5)
将化合物17.4(155mg,0.68mmol)溶解于5mL乙醇中,室温搅拌,分批加入硼氢化钠(154mg,4.06mmol),反应液在室温下搅拌1小时。反应结束后,用甲酸调节pH至5,减压浓缩,柱层析纯化得到产物17.5(100mg,产率:85%)。LCMS ESI(+)m/z:174.0(M+1)。
步骤E:4-氯-1,3-二氢呋喃[3,4-c]吡啶(化合物17.6)
将化合物17.5(100mg,0.58mmol)加入到5mL无水二氯甲烷中,然后依次加入二氧化锰(100mg,1.16mmol)、三乙基硅烷(1mL)和三氟乙酸(2mL),反应液在室温下搅拌过夜。将反应液通过铺有硅藻土的抽滤漏斗,二氯甲烷冲洗,减压浓缩,柱层析纯化得到产物17.6(75mg,产率:83%)。LCMS ESI(+)m/z:156.0(M+1)。
步骤F:N-(2,4-二甲氧基苄基)-1,3-二氢呋喃并[3,4-c]吡啶-4-胺(化合物17.7)
将化合物17.6(75mg,0.48mmol)和2,4-二甲氧基苯甲胺(162mg,0.97mmol)溶解于5mL无水甲苯中,然后依次加入Pd2(dba)3(44mg,0.05mmol)、BINAP(60mg,0.10mmol)和t-BuONa(184mg,1.92mmol)。氮气保护下,100℃反应过夜。反应结束后,减压浓缩,柱层析纯化得到产物17.7(70mg,产率:51%)。LCMS ESI(+)m/z:287.1(M+1)。
步骤G:1,3-二氢呋喃并[3,4-c]吡啶-4-胺(化合物17.8)
将化合物17.7(35mg,0.12mmol)加入到2mL三氟乙酸中,反应液在室温下搅拌1小时。反应结束后,减压浓缩,用乙酸乙酯溶解固体,用饱和碳酸氢钠溶液洗涤两次,有机相用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到产物17.8(15mg,产率:90%),LCMS ESI(+)m/z:137.1(M+1)。
步骤H:7-溴-1,3-二氢呋喃[3,4-c]吡啶-4-胺(化合物17.9)
将化合物17.8(15mg,0.11mmol)溶解于3mL乙腈中,再加入NBS(21mg,0.12mmol),室温反应2小时。反应结束后,减压浓缩,柱层析纯化得到产物17.9(10mg,产率:42%)。LCMS ESI(+)m/z:215.0,217.0(M+1)。
步骤I:6-溴-7,9-二氢呋喃并[3,4-c][1,2,4]三唑并[1,5-a]吡啶(化合物17.10)
将化合物17.9(10mg,0.05mmol)溶解于3mL DMF中,再加入DMF-DMA(18mg,0.15mmol),反应液在85℃搅拌2.5小时。然后减压浓缩,将固体溶解于3mL甲醇中,加入吡啶(5mg,0.06mmol)和羟胺-0-磺酸(7mg,0.06mmol),反应液在室温下搅拌过夜。反应结束后,减压浓缩,加入磷酸钾溶液和乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到产物17.10(7mg,产率:59%),LCMS ESI(+)m/z:240.0(M+1)。
步骤J:叔丁基4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑基[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-羧酸盐(化合物17.11)
将化合物17.10(7mg,0.029mmol)和4-(3-异丙基-2-(4,4,5,5-四甲基-1,3,2-二氧苯甲醛-2-基)-1H-吲哚-5-基)哌啶-1-羧酸叔丁酯(14mg,0.029mmol)加入到3mL 1,4-二氧六环和0.6mL水的混合液中,然后加入磷酸钾(18mg,0.087mmol)和Pd(dppf)Cl2(4mg,0.006mmol),氮气保护下,85℃反应2小时。用水淬灭反应,用乙酸乙酯(5mLX2)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到产物17.11(6mg,产率:41%),LCMS ESI(+)m/z:502.3(M+1)。1H NMR(400MHz,MeOD)δ8.73(s,1H),8.47(s,1H),7.59(s,1H),7.31(d,J=8.4Hz,1H),7.05(d,J=8.3Hz,1H),5.45(s,2H),5.18(s,2H),4.21(d,J=13.0Hz,2H),3.11(dt,J=14.0,7.0Hz,1H),2.89(d,J=19.0Hz,2H),2.77(t,J=12.1Hz,1H),1.85(d,J=12.3Hz,2H),1.66(td,J=12.6,3.8Hz,2H), 1.49(s,9H),1.44(d,J=7.0Hz,6H).
步骤K:6-(3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-7,9-二氢呋喃[3,4-c][1,2,4]三唑基[1,5-a]吡啶(化合物17.12)
将化合物17.11(6mg,0.012mmol)加入到1mL甲醇中,在冰浴中加入1mL 4M氯化氢的甲醇溶液,反应液在室温下反应2小时。减压浓缩,得到粗产物17.12(4mg),LCMS ESI(+)m/z:402.1(M+1)。
步骤L:2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑基[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酰胺(化合物17)
将化合物17.12(4mg,0.01mmol)溶解于3mL DCE中,再加入2-溴代乙酰胺(2mg,0.01mmol)和DBU(6mg,0.04mmol),反应液在室温下搅拌3小时。反应结束后,加入水和乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到产物17(2.3mg,产率:50%),LCMS ESI(+)m/z:459.2(M+1)。
1H NMR(400MHz,MeOD)δ8.75(s,1H),8.49(s,1H),7.64(s,1H),7.32(d,J=8.4Hz,1H),7.10(dd,J=8.5,1.5Hz,1H),5.47(t,J=3.0Hz,2H),5.21–5.16(m,2H),4.59(s,2H),3.38(s,1H),3.26(s,1H),3.12(dt,J=17.6,7.0Hz,1H),2.81–2.71(m,1H),2.64(t,J=9.8Hz,2H),2.08-1.96(m,4H),1.45(d,J=7.0Hz,6H).
实施例18
4-(5-(1-(2-(3,3-二氟吡咯烷-1-基)乙酰基)哌啶-4-基)-3-异丙基-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊二烯[c]吡啶-1-酮
将化合物1(50mg)溶解在10mL NMP溶液中,加入三乙胺(40mg,0.38mmol)、氯乙酰氯(22mg,0.19mmol),室温下搅拌1小时。加入DBU(98mg,0.64mmol)、3,3-二氟吡咯烷(55mg,0.38mmol),室温下搅拌过夜。旋干溶剂,反相制备纯化得到化合物18(14mg,产率:20%)。LCMS ESI(+)m/z:537.3(M+1).1H NMR(400MHz,DMSO)δ10.70(s,1H),7.55(s,1H),7.47(s,1H),7.21(d,J=8.3Hz,1H),6.94(dd,J=8.4,1.3Hz,1H),4.52(d,J=13.0Hz,1H),4.03(d,J=13.1Hz,1H),3.50(s,3H),3.14–2.93(m,4H),2.87–2.78(m,3H),2.78–2.58(m,6H),2.28(ddd,J=22.6,9.4,4.7Hz,3H),1.98(dd,J=14.8,7.4Hz,2H),1.82(d,J=11.3Hz,2H),1.66(dd,J=21.3,12.1Hz,1H),1.54–1.43(m,1H),1.33(d,J=7.0Hz,6H).
实施例19
4-(3-异丙基-5-(1-(2-(甲磺酰基)乙酰基)哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊烷[c]吡啶-1-酮
具体反应方程式如下所示:
将2-(甲磺酰基)乙酸(17mg,0.12mmol)溶解于5mL DMF中,依次加入DIPEA(39mg,0.3mmol)和HATU(57mg,0.15mmol),反应液在室温下搅拌0.5小时,再加入化合物1(40mg,0.1mmol),室温搅拌过夜。向反应液中加入水15mL,用二氯甲烷(10mL X 3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反向制备纯化得到产物19(17.9mg,产率:35%)。LCMS ESI(+)m/z:510(M+1)。1H NMR(400MHz,DMSO)δ10.70(s,1H),7.55(s,1H),7.47(s,1H),7.22(d,J=8.3Hz,1H),6.94(d,J=8.4Hz,1H),4.56(d,J=14.5Hz,1H),4.49(t,J=9.6Hz,2H),4.12(d,J=13.1Hz,1H),3.50(s,3H),3.22–3.10(m,5H),3.01–2.96(m,1H),2.86(t,J=12.0Hz,1H),2.72(t,J=8.0Hz,4H),2.00–1.95(m,2H),1.84(s,2H),1.77–1.68(m,1H),1.54–1.47(m,1H),1.34(d,J=7.0Hz,6H).
实施例20
4-(4-(3-异丙基-2-(2-甲基-1-氧-2,5,6,7-四氢-1H-环戊基[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)-4-氧代丁腈
具体反应方程式如下所示:
将3-氰基丙酸(12mg,0.12mmol)溶解于5mL DMF中,依次加入DIPEA(39mg,0.3mmol)和HATU(57mg,0.15mmol),反应液在室温下搅拌0.5小时,再加入化合物1(40mg,0.1mmol),室温搅拌过夜。向反应液中加入水15mL,用二氯甲烷(10mL X 3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反向制备纯化得到产物20(24mg,产率:51%)。LCMS ESI(+)m/z:471(M+1)。1H NMR(400MHz,DMSO)δ10.70(s,1H),7.55(s,1H),7.48(s,1H),7.21(d,J=8.3Hz,1H),6.95(dd,J=8.4,1.3Hz,1H),4.56(d,J=13.3Hz,1H),3.94(d,J=13.5Hz,1H),3.50(s,3H),3.12(t,J=12.0Hz,1H),3.01–2.94(m,1H),2.88–2.78(m,2H),2.78–2.73(m,2H),2.70(s,2H),2.67(dd,J=3.6,1.9Hz,2H),2.64(d,J=6.6Hz,2H),2.02–1.93(m,2H),1.83(d,J=12.3Hz,2H),1.67(dt,J=12.4,8.9Hz,1H),1.50(dt,J=12.3,8.7Hz,1H),1.33(d,J=7.0Hz,6H).
实施例21
N-(2-(4-(3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊二[c]吡啶-4-基)-1H-吲哚)-5-基)哌啶-1-基)-2-氧乙基)甲磺酰胺
具体反应方程式如下所示:
将化合物1(40mg,0.1mmol)溶于4mL DMF中,加入(甲基磺酰基)甘氨酸(18.4mg,0.12mmol),EDCI(28.8mg,0.15mmol),DMAP(18.3mg,0.15mmol)和DIPEA(25.8mg,0.2mmol),反应液在室温条件下搅拌2小时。向反应液中加入水5mL,用乙酸乙酯(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到化合物21(7.6mg,产率:14.5%)。LCMS ESI(+)m/z:525.2(M+1)。
1H NMR(400MHz,DMSO)δ10.70(s,1H),7.55(s,1H),7.48(s,1H),7.21(d,J=8.3Hz,1H),7.02(s,1H),6.97–6.92(m,1H),4.53(d,J=13.0Hz,1H),4.11–3.80(m,3H),3.50(s,3H),3.13(t,J=12.5Hz, 1H),3.02–2.94(m,4H),2.84(t,J=12.0Hz,1H),2.76–2.65(m,5H),2.03–1.92(m,2H),1.84(d,J=12.2Hz,2H),1.68(d,J=13.1Hz,1H),1.52(d,J=8.8Hz,1H),1.34(d,J=7.0Hz,6H).
实施例22
4-(5-(1-(2-(3,3-二氟吡咯烷-1-基)-2-氧乙基)哌啶-4-基)-3-异丙基-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊基[c]吡啶-1-酮
具体反应方程式如下所示:
步骤A:乙基2-(4-(3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊基[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)醋酸盐(化合物22.1)
将化合物1(100mg,0.26mmol)和2-溴乙酸乙酯(43.4mg,0.26mmol)溶解于10mL DCE中,加入DBU(79mg,0.52mmol),反应液在室温下搅拌1小时。向反应液中加入饱和氯化铵溶液20mL,用乙酸乙酯(25mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到产物22.1(120mg,产率:98.4%)。LCMS ESI(+)m/z:476.3(M+1)。
步骤B:2-(4-(3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊基[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)乙酸(化合物22.2)
将化合物22.1(120mg,0.25mmol)溶解于10mL乙醇中,加入10%NaOH水溶液2mL,反应液在室温下搅拌1小时。向反应液中加入稀盐酸溶液20mL,用乙酸乙酯(25mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到产物22.2(102mg,产率:91.3%)。LCMS ESI(+)m/z:448.3(M+1)。
步骤C:4-(5-(1-(2-(3,3-二氟吡咯烷-1-基)-2-氧乙基)哌啶-4-基)-3-异丙基-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊基[c]吡啶-1-酮(化合物22)
将化合物22.2(102mg,0.23mmol)和3,3-二氟吡咯烷(24.6mg,0.23mmol)溶解于10mL DMF中,加入HATU(129.2mg,0.34mmol)和DIEA(59.3mg,0.46mmol),反应液在室温下搅 拌1小时。向反应液中加入饱和氯化铵溶液20mL,用乙酸乙酯(25mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,用反相制备纯化得到产物22(28mg,产率:22.9%)。LCMS ESI(+)m/z:537.3(M+1)。1H NMR(400MHz,DMSO-d6)δ10.68(s,1H),7.55(s,1H),7.47(d,J=1.5Hz,1H),7.21(d,J=8.3Hz,1H),6.96(dd,J=8.4,1.6Hz,1H),4.08(t,J=13.1Hz,1H),3.84(t,J=7.4Hz,1H),3.72(t,J=13.4Hz,1H),3.53(t,J=7.5Hz,1H),3.50(s,3H),3.16(d,J=19.2Hz,2H),3.02–2.91(m,3H),2.72(q,J=7.8Hz,4H),2.41(ddd,J=28.4,14.1,6.9Hz,3H),2.24–2.13(m,2H),1.97(p,J=7.6Hz,2H),1.82–1.63(m,4H),1.34(d,J=7.0Hz,6H).
实施例23
4-(3-异丙基-5-(1-(2-(2-甲基-2H-四唑-5-基)乙酰基)哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊基[c]吡啶-1-酮
具体反应方程式如下所示:
步骤A:2-(2-甲基-2H-四唑-5-基)乙酸乙酯(化合物23.2)
将化合物23.1(1.0g,6.41mmol)溶解到40mL乙腈中,然后加入碘甲烷(2.27g,16.02mmol)和碳酸钾(2.21mg,16.02mmol),在80℃下搅拌过夜,LCMS检测反应完全,用乙酸乙酯(100mLX2)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到产物23.2(190mg,产率:17%)。LCMS ESI(+)m/z:171.0(M+1)。
步骤B:2-(2-甲基-2H-四唑-5-基)乙酸(化合物23.3)
将化合物23.2(180mg,1.05mmol)溶解到到3mL甲醇和1mL水中,然后加入氢氧化钠(127mg,3.17mmol),在室温下反应2小时。直接旋干,加入DCM:MeOH=10:1溶剂20mL溶解,过滤,减压浓缩,油泵拉干,得到粗产物23.3(300mg)。LCMS ESI(+)m/z:143.1(M+1)。
步骤C:4-(3-异丙基-5-(1-(2-(2-甲基-2H-四唑-5-基)乙酰基)哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊基[c]吡啶-1-酮(化合物23)
将化合物23.3(30mg,0.206mmol)和4-(3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊基[c]吡啶-1-酮(40mg,0.103mmol)溶解在4mL DMF中,然后加入DMAP(25mg,0.206mmol),DIPEA(66mg,0.515mmol)和EDCI(39mg,0.206mmol),反应液室温下搅拌过夜。LCMS监测反应50%。用反相制备纯化得到产物23(14.2mg,产率:13%)。LCMS ESI(+)m/z:514.3(M+1)。1H NMR(400MHz,DMSO)δ10.70(s,1H),7.56(s,1H),7.49(s,1H),7.22(d,J=8.3Hz,1H),6.96(d,J=8.4Hz,1H),4.53(d,J=12.6Hz,1H),4.34(s,3H),4.12(d,J=16.5Hz,3H),3.50(s,3H),3.20(s,1H),2.98(d,J=7.0Hz,1H),2.85(t,J=12.1Hz,1H),2.75–2.66(m,5H),1.98(dd,J=14.9,7.5Hz,2H),1.84(d,J=12.6Hz,2H),1.69(dd,J=12.4,3.5Hz,1H),1.52(dd,J=12.7,4.0Hz,1H),1.35(d,J=7.0Hz,6H).
实施例24
N-环丙基-2-(4-(3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺
具体反应方程式如下所示:
将化合物22.2(50mg,0.112mmol)溶于3mL DMF中,加入HATU(63.9mg,0.168mmol)和TEA(34mg,0.336mmol),反应液在室温条件下搅拌30分钟,在冰浴条件下加入环丙胺(9.6mg,0.168mmol),反应液在室温条件下搅拌1小时。向反应液中加入水5mL,用二氯甲烷(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到化合物24(7.4mg,产率:13.6%)。LCMS ESI(+)m/z:487.2(M+1)。1H NMR(400MHz,DMSO)δ10.68(s,1H),7.74(d,J=3.6Hz,1H),7.55(s,1H),7.48(s,1H),7.20(d,J=8.3Hz,1H),6.97(d,J=8.4Hz,1H),3.50(s,3H),3.03–2.95(m,1H),2.90(d,J=12.5Hz,4H),2.72(dd,J=15.4,7.7Hz,4H),2.68–2.61(m,2H),2.17(t,J=9.7Hz,2H),2.03–1.92(m,2H),1.86–1.69(m,4H),1.34(d,J=7.0Hz,6H),0.70–0.56(m,2H),0.54–0.41(m,2H).
实施例25
N-(2-(4-(3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊二[c]吡啶-4-基)-1H-吲哚)-5-基)哌啶-1-基)-2-氧代乙基)乙酰胺
具体反应方程式如下所示:
将化合物1(30mg,0.077mmol)溶于4mL DMF中,加入乙酰甘氨酸(18mg,0.154mmol),EDCI(29.5mg,0.154mmol),DMAP(18.8mg,0.154mmol)和DIPEA(50mg,0.385mmol),反应液在室温条件下搅拌过夜。向反应液中加入水5mL,用二氯甲烷(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到化合物25(15.4mg,产率:41.0%)。LCMS ESI(+)m/z:489.3(M+1)。1H NMR(400MHz,DMSO)δ10.70(s,1H),7.96(t,J=5.2Hz,1H),7.52(d,J=8.4Hz,2H),7.21(d,J=8.4Hz,1H),6.95(d,J=9.5Hz,1H),4.53(d,J=13.5Hz,1H),4.08–3.82(m,3H),3.50(s,3H),3.12(t,J=12.3Hz,1H),3.04–2.93(m,1H),2.83(t,J=12.1Hz,1H),2.75–2.67(m,5H),2.03–1.93(m,2H),1.92–1.78(m,5H),1.73–1.59(m,1H),1.58–1.43(m,1H),1.34(d,J=7.0Hz,6H).
实施例26
2-(4-溴苯基)乙烷-1-醇-2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-N-甲基乙酰胺
具体反应方程式如下所示:
将化合物17.12(25mg,0.0623mmol)溶于3mL四氢呋喃中,加入2-氯-N-甲基乙酰胺(20.1mg,0.187mmol),DIEA(80.6mg,0.623mmol),K2CO3(17.2mg,0.124mmol)和KI(20.6mg,0.124mmol),反应液在60℃条件下搅拌过夜。向反应液中加入水5mL,用乙酸乙酯(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到化合物26(4.1mg,产率:13.9%)。LCMS ESI(+)m/z:473.3(M+1)。1H NMR(400MHz,MeOD)δ8.75(s,1H),8.49(s,1H),7.64(s,1H),7.32(d,J=8.4Hz,1H),7.11(s,1H),5.46(s,2H),5.19(d,J=3.1Hz,2H),3.36(s,2H),3.24(d,J=11.3Hz,2H),3.12(dt,J=14.1,7.0Hz,1H),2.81(s,3H),2.75(t,J=11.4Hz,1H),2.64(t,J=10.6Hz,2H),2.11–1.93(m,4H),1.45(d,J=7.0Hz,6H).
实施例27
4-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-4-氧代丁腈
具体反应方程式如下所示:
将3-氰基丙酸(7mg,0.07mmol)溶解于5mL DMF中,依次加入DIPEA(23mg,0.18mmol)和HATU(34mg,0.09mmol),反应液在室温下搅拌0.5小时,再加入化合物17.12(25mg,0.06mmol),室温搅拌过夜。向反应液中加入水15mL,用二氯甲烷(10mL X 3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反向制备纯化得到产物27(13.2mg,产率:45%)。LCMS ESI(+)m/z:483.2(M+1)。1H NMR(400MHz,DMSO)δ11.01(s,1H),8.92(s,1H),8.61(s,1H),7.56(s,1H),7.29(d,J=8.4Hz,1H),7.03(dd,J=8.4,1.1Hz,1H),5.42(s,2H),5.12(t,J=3.0Hz,2H),4.57(d,J=12.9Hz,1H),3.95(d,J=13.0Hz,1H),3.16–3.01(m,2H),2.89–2.67(m,4H),2.65(d,J=6.6Hz,2H),1.84(d,J=12.1Hz,2H),1.75–1.65(m,1H),1.57–1.48(m,1H),1.38(d,J=7.0Hz,6H).
实施例28
N-(2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-2-氧乙基)甲磺酰胺
具体反应方程式如下所示:
将化合物17.12(25mg,0.0623mmol)溶于3mL DMF中,加入(甲基磺酰基)甘氨酸(19mg,0.125mmol),EDCI(24mg,0.125mmol),DMAP(15.3mg,0.125mmol)和DIPEA(24.2mg,0.188mmol),反应液在室温条件下搅拌2小时。向反应液中加入水5mL,用乙酸乙酯(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到化合物28(7.1mg,产率:21.3%)。LCMS ESI(+)m/z:537.2(M+1)。
1H NMR(400MHz,DMSO)δ11.01(s,1H),8.92(s,1H),8.61(s,1H),7.57(s,1H),7.30(d,J=8.4Hz,1H),7.03(t,J=6.9Hz,2H),5.42(s,2H),5.13(s,2H),4.55(d,J=12.0Hz,1H),4.05–3.88(m,3H),3.30(s,1H),3.14(t,J=12.3Hz,1H),3.06(dd,J=14.1,7.1Hz,1H),2.97(s,3H),2.87(t,J=11.8Hz,1H),1.85(d,J=11.6Hz,2H),1.77–1.66(m,1H),1.54(dd,J=22.0,12.7Hz,1H),1.38(d,J=7.0Hz,6H).
实施例29
4-(5-(1-(环丙基甘氨酰)哌啶-4-基)-3-异丙基-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊基[c]吡啶-1-酮
具体反应方程式如下所示:
将化合物1(50mg,0.12mmol)溶于5mL NMP中,加入三乙胺(38.96mg,0.38mmol),氯乙酰氯(21.74mg,0.19mmol),反应液在0℃反应0.5小时,然后升温至室温反应4小时,原料消失,然后加入DBU(97.63mg,0.64mmol),环丙氨(10.99mg,0.19mmol),反应液室温过夜。向反应液中加入氯化铵5mL,用乙酸乙酯(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到化合物29(2.1mg,产率:4.0%)。LCMS ESI(+)m/z:487.3 1H NMR(400MHz,CDCl3)δ7.83(s,1H),7.57(s,1H),7.31(d,J=8.5Hz,1H),7.17(s,1H),7.03(s,1H),4.80(d,J=13.4Hz,1H),3.88(d,J=13.5Hz,1H),3.80–3.64(m,2H),3.60(s,3H),3.20(t,J=12.3Hz,1H),3.04(p,J=6.9Hz,1H),2.91(t,J=7.4Hz,3H),2.81–2.67(m,3H),2.40(s,1H),2.12–1.95(m,7H),1.40(d,J=7.1Hz,6H),0.70(s,2H),0.58(d,J=6.3Hz,2H).
实施例30
1-(4-(3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊基[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)甲基)环丙烷-1-甲腈
具体反应方程式如下所示:
将化合物1(10mg,0.025mmol)溶于乙腈5mL中,加入1-(溴甲基)环丙烷-1-甲腈(4.1mg,0.025mmol),DIPEA(6.63mg,0.051mmol),反应液在80℃反应过夜,向反应液中加入氯化铵5mL,用乙酸乙酯(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到化合物30(3mg,产率:25%)。LCMS ESI(+)m/z:469.3.1H NMR(400MHz,DMSO)δ10.68(s,1H),7.56(s,1H),7.48(s,1H),7.21(d,J=8.3Hz,1H),6.97(dd,J=8.4,1.6Hz,1H),3.50(s,3H),3.12(d,J=11.3Hz,2H),3.04–2.93(m,1H),2.72(q,J=7.8Hz,4H),2.52(s,1H),2.45(s,2H),2.12(td,J=11.4,2.9Hz,2H),1.96(q,J=7.8Hz,2H),1.86–1.66(m,4H),1.34(d,J=7.0Hz,6H),1.30–1.21(m,2H),0.99–0.88(m,2H).
实施例31
2-(3,3-二氟吡咯烷-1-基)-1-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酮
具体反应方程式如下所示:
将化合物17.12(20mg,0.05mmol)加入到3mL N-甲基吡咯烷酮中,加入三乙胺(25mg,0.25mmol),在冰浴中加入氯乙酰氯(8mg,0.08mmol),反应液在室温下反应1.5小时。再加入DBU(38mg,0.25mmol)和3,3-二氟吡咯烷盐酸盐(11mg,0.08mmol),反应液在室温下搅拌过夜。反应结束后,加入水和乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到产物31(11.7mg,产率:43%),LCMS ESI(+)m/z:549.3(M+1)。1H NMR(400MHz,DMSO)δ11.01(s,1H),8.91(s,1H),8.61(s,1H),7.55(s,1H),7.30(d,J=8.4Hz,1H),7.03(d,J=8.5Hz,1H),5.42(s,2H),5.13(s,2H),4.53(d,J=12.0Hz,1H),4.04(d,J=13.0Hz,1H),3.51(d,J=14.2Hz,1H),3.30(d,J=6.7Hz,1H),3.14–2.95(m,4H),2.83(dt,J=6.7,4.8Hz,2H),2.66(dd,J=19.4,10.6Hz,2H),2.24(ddd,J=28.0,13.8,6.3Hz,2H),1.84(d,J=11.6Hz,2H),1.68(dt,J=12.0,9.0Hz,1H),1.50(dt,J=12.3,8.6Hz,1H),1.38(d,J=7.0Hz,6H).
实施例32
6-(3-异丙基-5-(1-(2-(甲基磺酰基)乙基)哌啶-4-基)-1H-吲哚-2-基)-7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶
具体反应方程式如下所示:
将化合物17.12(30mg,0.07mmol)溶解于2mL二氯乙烷中,加入1-溴-2-(甲基磺酰基)乙烷(16mg,0.08mmol),DBU(42mg,0.28mmol),反应液在室温下搅拌过夜。将反应液用二氯甲烷萃取,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,用反相制备纯化得到产物32(16.1mg,产率:46%),LCMS ESI(+)m/z:508.2(M+1)。1H NMR(400MHz,DMSO)δ10.99(s,1H),8.92(s,1H),8.61(s,1H),7.55(s,1H),7.29(d,J=8.4Hz,1H),7.04(d,J=8.5Hz,1H),5.42(s,2H),5.13(s,2H),3.10–2.99(m,6H),2.76(s,2H),2.59(s,1H),2.52(s,2H),2.12(s,2H),1.86–1.64(m,4H),1.38(d,J=7.0Hz,6H).
实施例33
6-(3-异丙基-5-(1-(氧杂环丁烷-3-基)哌啶-4-基)-1H-吲哚-2-基)-7,9-二氢呋喃[3,4-c][1,2,4]三唑[1,5-a]吡啶
具体反应方程式如下所示:
将化合物17.12(25mg,0.06mmol)加入到3mL二氯甲烷中,加入3-氧杂环丁酮(18mg,0.25mmol),乙酸(7mg,0.13mmol)和三乙酰氧基硼氢化钠(53mg,0.25mmol),反应液在室温下搅拌过夜。反应结束后,加入水淬灭反应,用乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到产物33(10.4mg,产率:37%),LCMS ESI(+)m/z:458.2(M+1)。1H NMR(400MHz,DMSO)δ11.00(s,1H),8.91(s,1H),8.61(s,1H),7.57(s,1H),7.29(d,J=8.4Hz,1H),7.05(dd,J=8.4,1.3Hz,1H),5.42(d,J=2.9Hz,2H),5.13(t,J=3.1Hz,2H),4.56(t,J=6.5Hz,2H),4.47(t,J=6.1Hz,2H),3.05(dt,J=14.0,7.0Hz,1H),2.82(d,J=11.0Hz,2H),2.59(ddd,J=11.4,9.8,4.1Hz,1H),2.53(d,J=1.9Hz,1H),1.88(dt,J=10.9,5.6Hz,2H),1.84–1.68(m,4H),1.39(d,J=7.0Hz,6H).
实施例34
2-(环丙基氨基)-1-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酮
具体反应方程式如下所示:
将化合物17.12(35mg,0.09mmol)加入到4mL N-甲基吡咯烷酮中,加入三乙胺(44mg,0.44mmol),在冰浴中加入氯乙酰氯(15mg,0.13mmol),反应液在室温下反应1.5小时。再加入DBU(66mg,0.44mmol)和环丙胺(7mg,0.13mmol),反应液在室温下搅拌过夜。反应结束后,加入水和乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到产物34(8.3mg,产率:19%),LCMS ESI(+)m/z:499.3(M+1)。1H NMR(400MHz,DMSO)δ11.02(s,1H),8.92(s,1H),8.62(s,1H),7.57(s,1H),7.30(d,J=8.4Hz,1H),7.04(dd,J=8.4,1.3Hz,1H),5.43(s,2H),5.13(t,J=3.1Hz,2H),4.58(d,J=12.4Hz,1H),3.95(d,J=13.2Hz,1H),3.47(d,J=15.1Hz,2H),3.06(dd,J=18.4,11.3Hz,2H),2.87(tt,J=11.6,3.1Hz,1H),2.67(dd,J=7.9,6.2Hz,1H),2.53(d,J=1.9Hz,1H),2.18(ddd,J=10.0,6.7,3.6Hz,1H),1.85(d,J=11.5Hz,2H),1.67(ddd,J=16.5,13.3,4.3Hz,1H),1.53(ddd,J=25.1,12.6,4.0Hz,1H),1.39(d,J=7.0Hz,6H),0.42–0.32(m,2H),0.31–0.23(m,2H).
实施例35
2-(4-(2-(1,6-二甲基-7-氧代-6,7-二氢-1H-吡咯并[2,3-c]吡啶-4-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酰胺
具体反应方程式如下所示:
步骤A:4-溴-1,6-二甲基-1,6-二氢-7H-吡咯并[2,3-c]吡啶-7-酮(化合物35.2)
将NaH(27mg,0.664mmol)置于100mL三口瓶中,用氩气置换三次,在冰浴条件下滴加4-溴-6-甲基-1,6-二氢-7H-吡咯并[2,3-c]吡啶-7-酮(100mg,0.443mmol)的DMF(5mL)溶液,反应液在室温条件下搅拌30分钟,然后加入CH3I(94.2mg,0.664mmol),反应液在室温条件下搅拌1小时。向反应液中加入氯化铵的水溶液5mL,用乙酸乙酯(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到化合物35.2(87mg,产率:81.8%),LCMS ESI(+)m/z:241.0(M+1)。
步骤B:叔丁基4-(2-(1,6-二甲基-7-氧代-6,7-二氢-1H-吡咯并[2,3-c]吡啶-4-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-羧酸盐(化合物35.3)
将化合物35.2(67mg,0.28mmol)溶于5mL 1,4-二氧六环溶液和1mL水中,加入4-(2-(((12-氮杂亚甲基)膦基)碘硼烷基)-3-异丙基-1H-吲哚-5-基)哌啶-1-羧酸叔丁酯(196.6mg,0.42mmol),K3PO4(178.3mg,0.84mmol),Pd(dppf)Cl2(41mg,0.056mmol),反应液用氩气置换三次,在85℃条件下搅拌2小时。向反应液中加入水5mL,用乙酸乙酯(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到化合物35.3(141mg,产率:100%),LCMS ESI(+)m/z:503.3(M+1)。
步骤C:4-(3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-1,6-二甲基-1,6-二氢-7H-吡咯并[2,3-c]吡啶-7-酮(化合物35.4)
在化合物35.3(141mg,0.28mmol)中滴加盐酸的甲醇溶液(5mL),反应液在室温条件下搅拌1小时。反应液用NaOH水溶液调至中性,用iPrOH:DCM=1:3(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩得到化合物35.4(113mg,产率:100%),LCMS ESI(+)m/z:403.2(M+1)。
步骤D:2-(4-(2-(1,6-二甲基-7-氧代-6,7-二氢-1H-吡咯并[2,3-c]吡啶-4-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酰胺(化合物35)
将化合物35.4(113mg,0.28mmol)溶于5mL DCE中,加入2-溴乙酰胺(47mg,0.336mmol)和DBU(170mg,1.12mmol),反应液在室温条件下搅拌3小时。向反应液中加入水5mL,用乙酸乙酯(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到化合物35(22.5mg,产率:17.5%),LCMS ESI(+)m/z:460.3(M+1)。1H NMR(400MHz)δ10.76(s,1H),7.52(s,1H),7.32(d,J=2.8Hz,1H),7.26(s,1H),7.24(d, J=8.3Hz,1H),7.19(s,1H),7.14(s,1H),6.97(dd,J=8.4,1.4Hz,1H),6.10(d,J=2.8Hz,1H),4.11(s,3H),3.54(s,3H),3.14–3.05(m,1H),3.03–2.84(m,4H),2.52(s,1H),2.20(s,2H),1.91–1.72(m,4H),1.36(d,J=7.0Hz,6H).
实施例36
1-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-2-吗啉乙酮
具体反应方程式如下所示:
将化合物17.12(20mg,0.05mmol)加入到3mL N-甲基吡咯烷酮中,加入三乙胺(25mg,0.25mmol),在冰浴中加入氯乙酰氯(8mg,0.08mmol),反应液在室温下反应1.5小时。再加入DBU(38mg,0.25mmol)和吗啉(7mg,0.08mmol),反应液在室温下搅拌过夜。反应结束后,加入水和乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到产物36(11.5mg,产率:44%),LCMS ESI(+)m/z:529.3(M+1)。1H NMR(400MHz,DMSO)δ11.02(s,1H),8.92(s,1H),8.61(s,1H),7.55(s,1H),7.30(d,J=8.4Hz,1H),7.02(dd,J=8.4,1.3Hz,1H),5.42(t,J=2.8Hz,2H),5.13(t,J=3.1Hz,2H),4.53(d,J=12.5Hz,1H),4.18(d,J=13.1Hz,1H),3.60(t,J=4.5Hz,4H),3.31(d,J=13.5Hz,2H),3.11–3.03(m,2H),2.87(t,J=12.0Hz,1H),2.69–2.61(m,1H),2.44(d,J=3.3Hz,4H),1.85(t,J=10.9Hz,2H),1.74(td,J=12.2,3.4Hz,1H),1.48(ddd,J=25.3,12.6,3.7Hz,1H),1.38(d,J=7.0Hz,6H).
实施例37
1-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-2-(甲氨基)乙烷-1-酮
具体反应方程式如下所示:
步骤A:叔丁基(2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基)-1H-吲哚-5-基)哌啶-1-基)-2-氧代乙基)(甲基)氨基甲酸酯(化合物37.1)
将化合物17.12(30mg,0.075mmol)溶解到5mL DMF中,加入HATU(42mg,0.112mmol),DIPEA(29mg,0.224mmol)和N-(叔丁氧羰基)-N-甲基甘氨酸(18.4mg,0.097mmol),反应液在室温搅拌2小时。向反应液中加入水,用乙酸乙酯萃取,有机相用饱和食盐水洗,无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到化合物37.1(36mg,产率:85%),LCMS ESI(+)m/z:573.3(M+1)。
步骤B:1-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-2-(甲氨基)乙烷-1-酮(化合物37)
将化合物37.1(36mg,0.063mmol)溶解在3mL甲醇中,加入4M盐酸甲醇溶液3mL,反应液在室温搅拌2小时。将反应液减压浓缩,剩余物用反相制备纯化得到产物37(3.4mg,产率:12%),LCMS ESI(+)m/z:473.3(M+1)。1H NMR(400MHz,DMSO-d6)δ11.04(s,1H),8.92(s,1H),8.61(s,1H),7.56(s,1H),7.30(d,J=8.4Hz,1H),7.03(d,J=9.6Hz,1H),5.42(s,2H),5.13(s,2H),4.56(d,J=12.5Hz,1H),3.90(d,J=13.4Hz,1H),3.62–3.49(m,2H),3.15–2.99(m,2H),2.87(t,J=13.7Hz,1H),2.75–2.66(m,1H),2.52(d,J=1.9Hz,1H),2.38(s,3H),1.85(d,J=11.8Hz,2H),1.75–1.62(m,1H),1.53(d,J=7.8Hz,1H),1.38(d,J=7.0Hz,6H).
实施例38
(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)(4-甲基哌嗪-1-基)甲酮
具体反应方程式如下所示:
步骤A:4-硝基苯基4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-羧酸盐(化合物38.1)
将化合物17.12(40mg,0.09mmol)溶于无水DCM(4mL)中,在0℃下加入DIPEA(24mg,0.18mmol)和4-硝基苯氧基酰氯(28mg,0.14mmol)中,在室温下反应2小时。反应完成后,向反 应液滴加饱和碳酸氢钠溶液淬灭反应,然后用DCM(20mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱色谱纯化得到产物38.1(43mg,产率:84%),LCMS ESI(+)m/z:567.2(M+1)。
步骤B:(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)(4-甲基哌嗪-1-基)甲酮(化合物38)
将38.1(43mg,0.076mmol)和1-甲基哌嗪(38mg,0.379mmol),碳酸钾(21mg,0.152mmol)加入到DMF(4mL)中,在150℃的微波下反应1.5小时。反应完成后,向反应液加入10mL水,然后用EA(20mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到产物38(14mg,产率:35%),LCMS ESI(+)m/z:528.3(M+1)。1H NMR(400MHz,MeOD)δ8.75(s,1H),8.49(s,1H),7.60(s,1H),7.31(d,J=8.4Hz,1H),7.08(dd,J=8.5,1.5Hz,1H),5.46(t,J=3.0Hz,2H),5.21–5.14(m,2H),3.88(d,J=13.1Hz,2H),3.44–3.34(m,4H),3.16–3.06(m,1H),3.01(dd,J=12.6,10.7Hz,2H),2.83(ddd,J=12.0,8.4,3.5Hz,1H),2.68(s,4H),2.47(s,3H),1.91(d,J=10.8Hz,2H),1.77(qd,J=12.7,3.9Hz,2H),1.45(d,J=7.1Hz,6H).
实施例39
2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙烷-1-磺酰胺
具体反应方程式如下所示:
步骤A:2-氯乙磺酰胺(化合物39.2)
将2-氯乙磺酰氯(100mg,0.61mmol)溶解于5mL THF中,加入氨水(0.5mL)反应液在室温下搅拌过夜。将反应液直接用于下一步反应,LCMS ESI(+)m/z:144.0(M+1)。
步骤B:2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙烷-1-磺酰胺(化合物39)
将化合物39.2(87mg,0.61mmol),三乙胺(186mg,1.83mmol)溶解于5mL THF中,加入化合物17.12(25mg,0.06mmol),反应液在室温下搅拌过夜。将反应液用乙酸乙酯萃取,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,用反相制备纯化得到产物39(3.5mg,产率:12%),LCMS ESI(+)m/z:509.2(M+1)。1H NMR(400MHz,DMSO)δ10.99(s,1H),8.91(s,1H), 8.61(s,1H),7.56(s,1H),7.29(d,J=8.4Hz,1H),7.04(d,J=7.6Hz,1H),6.80(s,2H),5.42(s,2H),5.13(s,2H),3.22–3.17(m,2H),3.05(dd,J=14.0,7.2Hz,1H),2.99(d,J=11.2Hz,2H),2.81–2.73(m,2H),2.59(t,J=8.0Hz,1H),2.13(t,J=11.2Hz,2H),2.04–1.95(m,2H),1.79(d,J=12.0Hz,2H),1.38(d,J=7.2Hz,6H).
实施例40
1-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-2-甲基丙烷-2-醇
将化合物17.12(40mg,0.1mmol)和1-氯-2-甲基丙烷-2-醇(1.1g,10.0mmol),在氮气保护下溶解于3mL DMF中,加入DIEA(52mg,0.4mmol),反应液在120℃下搅拌2小时。向反应液中加入水20mL,用乙酸乙酯(25mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,用反相制备纯化得到产物40(5.5mg,产率:12%),LCMS ESI(+)m/z:474.3(M+1)。1H NMR(400MHz,DMSO-d6)δ10.99(s,1H),8.92(s,1H),8.61(s,1H),7.55(d,J=1.5Hz,1H),7.29(d,J=8.3Hz,1H),7.04(dd,J=8.5,1.5Hz,1H),5.42(t,J=3.3Hz,2H),5.13(t,J=3.4Hz,2H),3.14–2.99(m,4H),2.30(d,J=6.2Hz,4H),1.80–1.69(m,4H),1.38(d,J=7.0Hz,6H),1.12(s,6H).
实施例41
N-环丙基-2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺
具体反应方程式如下所示:
步骤A:乙基2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸酯(化合物41.1)
将化合物17.12(50mg,0.125mmol)溶于3mL DCM中,加入2-溴乙酸乙酯(31.3mg,0.87mmol)和DIPEA(97mg,0.75mmol),反应液在室温条件下搅拌2小时。向反应液中加入水5mL,用乙酸乙酯(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,柱层析纯化得到化合物41.1(49mg,产率:80.4%),LCMS ESI(+)m/z:488.3(M+1)。
步骤B:2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸(化合物41.2)
将化合物41.1(49mg,0.10mmol)溶于1mL甲醇和1mL四氢呋喃中,加入氢氧化钠(10mg,0.25mmol)的水(1.5mL)溶液,反应液在室温条件下搅拌1小时。反应液用盐酸水溶液调节PH至中性,用iPrOH:DCM=1:3(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,得到化合物41.2(50mg,产率:100%),LCMS ESI(+)m/z:460.2(M+1)。
步骤C:N-环丙基-2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺(化合物41)
将化合物41.2(50mg,0.11mmol)溶于3mL DMF中,加入HATU(62.7mg,0.165mmol)和三乙胺(33.4mg,0.33mmol)溶液,反应液在室温条件下搅拌30分钟,然后滴加环丙胺(9.4mg,0.165mmol),反应液在室温条件下搅拌1小时。反应液用氯化铵水溶液调节PH至中性,用乙酸乙酯(10mLX3)萃取,合并有机相,用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到化合物41(10.1mg,产率:18.4%),LCMS ESI(+)m/z:499.3(M+1)。1H NMR(400MHz,DMSO)δ10.99(s,1H),8.92(s,1H),8.61(s,1H),7.74(d,J=3.9Hz,1H),7.56(s,1H),7.29(d,J=8.4Hz,1H),7.05(d,J=8.5Hz,1H),5.42(s,2H),5.13(s,2H),3.08–3.01(m,1H),2.90(d,J=9.8Hz,4H),2.68–2.66(m,1H),2.52(d,J=1.2Hz,1H),2.17(dd,J=11.0,8.0Hz,2H),1.83–1.72(m,4H),1.39(d,J=7.0Hz,6H),0.62(td,J=7.0,4.8Hz,2H),0.54–0.45(m,2H).
实施例42
(S)-1-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑基[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-2-(3-氟吡咯烷-1-基)乙酮
具体反应方程式如下所示:
将化合物17.12(14mg,0.03mmol)加入到2mL N-甲基吡咯烷酮中,加入三乙胺(18mg,0.17mmol),在冰浴中加入氯乙酰氯(6mg,0.05mmol),反应液在室温下反应1.5小时,再加入DBU(27mg,0.17mmol)和3-(S)-氟吡咯烷盐酸盐(7mg,0.05mmol),反应液在室温下搅拌过夜。反应结束后,加入水和乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到化合物42(4.8mg,产率:26%),LCMS ESI(+)m/z:531.2(M+1)。1H NMR(400MHz,DMSO)δ11.02(s,1H),8.91(s,1H),8.61(s,1H),7.55(s,1H),7.30(d,J=8.4Hz,1H),7.03(d,J=8.5Hz,1H),5.42(s,2H),5.13(t,J=3.1Hz,2H),4.54(d,J=12.0Hz,1H),4.09(t,J=11.5Hz,1H),3.12(dd,J=18.8,6.6Hz,2H),3.03(dd,J=14.1,6.9Hz,2H),2.96(dd,J=7.9,6.1Hz,2H),2.91–2.81(m,2H),2.70–2.67(m,1H),2.53(d,J=1.9Hz,2H),2.25–2.09(m,1H),1.97(ddd,J=28.8,15.1,7.5Hz,1H),1.84(d,J=12.3Hz,2H),1.78–1.63(m,1H),1.54–1.45(m,1H),1.38(d,J=7.0Hz,6H).
实施例43
6-(3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶
参照实施例1,以4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-甲酸叔丁酯和4M盐酸的甲醇溶液为原料制得。LCMS ESI(+)m/z:402.2。1H NMR(400MHz,MeOD)δ8.92(s,1H),8.87(s,1H),7.67(s,1H),7.38(d,J=8.3Hz,1H),7.14(d,J=8.3Hz,1H),5.51(s,2H),5.25(s,2H),3.54(d,J=11.3Hz,2H),3.20(t,J=12.1Hz,2H),3.14(s,1H),3.02(t,J=10.9Hz,1H),2.14(d,J=13.6Hz,2H),2.04(dd,J=23.3,11.7Hz,2H),1.47(d,J=6.2Hz,6H).
实施例44
2-(4-(2-(8,9-二氢-7H-环戊烷[c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酰胺
参照实施例1,以2-(4-(3-异丙基-2-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺和6-溴-8,9-二氢-7H-环戊烷[c][1,2,4]三唑并[1,5-a]吡啶为原料制得。LCMS ESI(+)m/z:457.2。1H NMR(400MHz,DMSO)δ10.78(s,1H),7.58–7.48(m,2H),7.32–7.21(m,2H),7.16(d,J=3.2Hz,1H),7.00(dd,J=8.4,1.5Hz,1H),3.17(s,2H),2.98–2.93(m,5H),2.81(t,J=7.6Hz,2H),2.67(dt,J=3.6,1.8Hz,1H),2.33(dt,J=3.6,1.8Hz,1H),2.23(dd,J=12.3,9.2Hz,2H),2.03(q,J=7.7Hz,2H),1.88–1.74(m,4H),1.34(d,J=7.0Hz,6H).
实施例45
2-(4-(3-异丙基-2-(6-甲基-7-氧代-6,7-二氢-1H-吡唑并[3,4-c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺
参照实施例1,以4-溴-6-甲基-1-甲苯磺酰基-1,6-二氢-7H-吡唑并[3,4-c]吡啶-7-酮和2-(4-(3-异丙基-2-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺为原料制得。LCMS ESI(+)m/z:447.2。1H NMR(400MHz,DMSO)δ13.6(s,1H),11.6(s,1H),8.38(d,J=4.2Hz,1H),7.62(s,1H),7.54(s,1H),7.30(d,J=8.3Hz,2H),7.19(s,2H),3.67(s,3H),3.29(s,2H),3.14–2.88(m,5H),2.29(m,1H),1.86–1.76(m,4H),1.35(d,J=7.0Hz,6H).
实施例46
2-(4-(3-异丙基-2-(7-甲氧基-2,3-二氢苯并呋喃-4-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺
参照实施例2,以3-异丙基-2-(7-甲氧基-2,3-二氢苯并呋喃-4-基)-5-(哌啶-4-基)-1H-吲哚盐酸盐和2-溴乙酰胺为原料制得。LCMS ESI(+)m/z:448.3。1H NMR(400MHz,DMSO)δ10.67(s,1H),7.50(s,1H),7.25(s,1H),7.21(d,J=8.3Hz,1H),7.13(s,1H),6.95(t,J=8.3Hz,2H),6.78(d,J=8.3Hz,1H),4.55(t,J=8.7Hz,2H),3.81(s,3H),3.16(t,J=8.7Hz,2H),3.07(dd,J=14.0,7.0Hz,1H),2.94(d,J=10.8 Hz,2H),2.89(s,2H),2.19(t,J=11.2Hz,3H),1.82(dd,J=23.9,12.5Hz,4H),1.35(d,J=7.0Hz,6H).
实施例47
2-(4-(4-氟-3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊烷[c]吡啶-4-基)-1H-吡咯并[2,3-c]吡啶-5-基)哌嗪-1-基)乙酰胺
参照实施例2,以4-(4-氟-3-异丙基-5-(哌嗪-1-基)-1H-吡咯并[2,3-c]吡啶-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊烷[c]吡啶-1-酮和2-溴乙酰胺为原料制得。LCMS ESI(+)m/z:467.2。1H NMR(400MHz,DMSO)δ11.34(d,J=2.2Hz,1H),8.21(d,J=1.6Hz,1H),7.68(s,1H),7.18(dd,J=28.4,3.2Hz,2H),3.51(s,3H),3.22(t,J=4.7Hz,4H),2.99(dq,J=10.2,3.5Hz,1H),2.94(s,2H),2.72(dt,J=13.8,7.6Hz,4H),2.63(t,J=4.8Hz,4H),1.99(p,J=7.6Hz,2H),1.26(dd,J=7.0,1.2Hz,6H).
实施例48
1-((4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)甲基)环丙烷-1-甲腈
参照实施例30,以6-(3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶盐酸盐和1-(溴甲基)环丙烷-1-甲腈为原料制得。LCMS ESI(+)m/z:481.2。1H NMR(400MHz,DMSO)δ11.08(s,1H),8.93(s,1H),8.62(s,1H),7.59(s,1H),7.33(d,J=6.2Hz,1H),7.06(d,J=8.4Hz,1H),5.43(s,2H),5.13(t,J=3.0Hz,2H),3.48(s,2H),3.17(s,2H),3.05(dd,J=14.0,7.0Hz,1H),2.90(s,1H),2.58(s,2H),2.15(s,2H),2.03–1.96(m,1H),1.82(s,1H),1.51(s,2H),1.39(d,J=7.0Hz,6H),1.32(d,J=15.0Hz,2H).
实施例49
2-(4-(3-异丙基-2-(6-甲基-7-氧代-2,3,6,7-四氢呋喃[2,3-c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺
参照实施例2,以4-(3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-6-甲基-2,3-二氢呋喃[2,3-c]吡啶 -7(6H)-酮和2-溴代乙酰胺为原料制得。LCMS ESI(+)m/z:449.2。1H NMR(400MHz,DMSO)δ10.73(s,1H),7.50(s,1H),7.31(s,1H),7.26(s,1H),7.22(d,J=8.3Hz,1H),7.14(s,1H),6.98(d,J=8.4Hz,1H),4.55(t,J=9.3Hz,2H),3.54(s,3H),3.07(dt,J=9.1,8.2Hz,4H),2.94(d,J=11.0Hz,2H),2.90(s,2H),2.19(td,J=10.9,2.2Hz,2H),1.81(ddd,J=25.1,17.3,7.0Hz,4H),1.37(d,J=7.0Hz,6H).
实施例50
2-(4-(3-异丙基-2-(5-甲基-4-氧代-1,3,4,5-四氢呋喃[3,4-c]吡啶-7-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺
参照实施例1,以7-溴-5-甲基-3,5-二氢呋喃[3,4-c]吡啶-4(1H)-酮和2-(4-(3-异丙基-2-(4,4,5,5-四甲基-1,3-二氧杂环戊-2-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺为原料制得。LCMS ESI(+)m/z:449.3。1H NMR(400MHz,DMSO)δ10.72(s,1H),7.72(s,1H),7.50(s,1H),7.26–7.19(m,2H),7.13(s,1H),7.00–6.95(m,1H),4.93(d,J=3.5Hz,4H),3.56(s,3H),3.03(dd,J=14.0,7.0Hz,1H),2.95–2.87(m,4H),2.56(s,1H),2.17(s,2H),1.78(d,J=14.7Hz,4H),1.36(d,J=7.0Hz,6H).
实施例51
2-(4-(3-异丙基-2-(2-异丙基-1-氧代-2,5,6,7-四氢-1H-环戊[c]吡啶-)4-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺
参照实施例1,以2-(4-(3-异丙基-2-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺和4-溴-2-异丙基-2,5,6,7-四氢-1H-环戊[c]吡啶-1-酮为原料制得。LCMS ESI(+)m/z:475.3。1H NMR(400MHz,DMSO)δ10.71(s,1H),7.49(s,1H),7.43(s,1H),7.25(s,1H),7.21(d,J=8.3Hz,1H),7.13(s,1H),6.97(d,J=8.3Hz,1H),5.15(dt,J=13.6,6.8Hz,1H),3.01–2.91(m,4H),2.89(s,2H),2.74(t,J=7.5Hz,4H),2.19(t,J=10.0Hz,2H),2.01–1.95(m,2H),1.79(dd,J=24.5,10.1Hz,4H),1.35(d,J=7.0Hz,6H),1.31(d,J=6.8Hz,6H).
实施例52
2-(4-(3-异丙基-2-(2-甲基-1-氧代-1,2,5,6,7,8-六氢异喹啉-4-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺
参照实施例1,以4-溴-2-甲基-5,6,7,8-四氢异喹啉-1(2H)-酮和2-(4-(3-异丙基-2-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺为原料制得。LCMS ESI(+)m/z:461.3。1H NMR(400MHz,DMSO)δ10.66(s,1H),7.48(s,2H),7.24(s,1H),7.19(d,J=8.3Hz,1H),7.13(s,1H),6.96(d,J=7.8Hz,1H),3.46(s,3H),2.95–2.87(m,5H),2.45–2.36(m,4H),2.16(d,J=10.8Hz,2H),2.08(s,1H),1.86–1.74(m,4H),1.60(s,4H),1.31(d,J=5.4Hz,6H).
实施例53
2-(4-(6-氟-3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊烷[c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺
参照实施例2,以4-(6-氟-3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊烷[c]吡啶-1-酮和2-溴乙酰胺为原料制得。LCMS ESI(+)m/z:465.2。1H NMR(400MHz,DMSO)δ10.80(s,1H),7.57(s,1H),7.51(d,J=7.3Hz,1H),7.27(s,1H),7.14(d,J=3.2Hz,1H),7.00(d,J=11.3Hz,1H),3.50(s,3H),3.01–2.93(m,3H),2.91(s,2H),2.80(ddt,J=11.9,8.2,3.7Hz,1H),2.71(q,J=8.1Hz,4H),2.21(td,J=11.5,2.5Hz,2H),1.97(p,J=7.6Hz,2H),1.87(qd,J=12.3,3.6Hz,2H),1.75(dd,J=13.1,3.6Hz,2H),1.34(d,J=7.0Hz,6H).
实施例54
2-(5-(3-异丙基-2-(2-甲基-1-氧代-2,5,6,7-四氢-1H-环戊烷[c]吡啶-4-基)-1H-吲哚-5-基)六氢环戊[c]吡咯-2(1H)-基)乙酰胺
参照实施例2,以2-溴乙酰胺和4-(3-异丙基-5-(八氢环戊烷[c]吡咯-5-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊烷[c]吡啶-1-酮为原料制得。LCMS ESI(+)m/z:473.3。1HNMR(400MHz,DMSO) δ10.66(s,1H),7.56(s,1H),7.48(s,1H),7.20(d,J=8.4Hz,1H),7.18–7.09(m,2H),7.03(d,J=8.2Hz,1H),3.50(s,3H),3.03–2.92(m,4H),2.77–2.66(m,6H),2.61(q,J=7.0Hz,2H),2.32(dd,J=9.0,5.8Hz,2H),2.22(dt,J=12.4,6.3Hz,2H),2.03–1.89(m,2H),1.52(td,J=11.9,7.9Hz,2H),1.33(d,J=7.0Hz,6H).
实施例55
1-(3,3-二氟吡咯烷-1-基)-2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2),4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙烷-1-酮
参照实施例5,以2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸和3,3-二氟吡咯烷为原料制得。LCMS ESI(+)m/z:549.3。1H NMR(400MHz,MeOD)δ8.76(s,1H),8.50(s,1H),7.64(s,1H),7.33(d,J=8.4Hz,1H),7.11(dd,J=8.4,1.3Hz,1H),5.48(t,J=3.0Hz,2H),5.22–5.18(m,2H),4.59(s,2H),4.03(t,J=12.6Hz,1H),3.85(dd,J=14.7,7.4Hz,2H),3.72(t,J=7.6Hz,1H),3.58(s,1H),3.50(s,1H),3.13(dd,J=8.7,5.4Hz,1H),2.75(dd,J=10.0,5.3Hz,1H),2.61–2.33(m,4H),1.99(s,4H),1.46(d,J=7.0Hz,6H).
实施例56
(S)-2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-1-(3-氟吡咯烷-1-基)乙烷-1-酮
参照实施例5,以2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸和(S)-3-氟吡咯烷为原料制得。LCMS ESI(+)m/z:531.3。1H NMR(400MHz,MeOD)δ8.75(s,1H),8.49(s,1H),7.63(s,1H),7.31(d,J=8.4Hz,1H),7.09(d,J=8.4Hz,1H),5.46(d,J=2.9Hz,2H),5.32(dd,J=52.8,25.7Hz,1H),5.19(t,J=3.2Hz,2H),3.99–3.66(m,3H),3.57–3.33(m,3H),3.24(t,J=11.6Hz,2H),3.11(dd,J=14.1,7.0Hz,1H),2.77–2.63(m,1H),2.49(d,J=11.4Hz,2H),2.38–2.07(m,2H),1.97(d,J=23.2Hz,4H),1.45(d,J=7.0Hz,6H).
实施例57
1-(3,3-二氟氮杂环丁烷-1-基)-2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2),4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙烷-1-酮
参照实施例5,以2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸和3,3-二氟氮杂环丁烷为原料制得。LCMS ESI(+)m/z:535.3。1H NMR(400MHz,MeOD)δ8.75(s,1H),8.49(s,1H),7.61(s,1H),7.31(d,J=8.4Hz,1H),7.09(dd,J=8.4,1.3Hz,1H),5.46(t,J=3.0Hz,2H),5.19(t,J=3.3Hz,2H),4.72(t,J=12.0Hz,2H),4.58(s,1H),4.37(t,J=12.1Hz,2H),3.27(s,1H),3.12(dd,J=9.0,4.8Hz,3H),2.66(dd,J=13.0,7.8Hz,1H),2.44–2.29(m,2H),2.01–1.86(m,4H),1.45(d,J=7.0Hz,6H).
实施例58
2-(4-溴苯基)乙烷-1-醇(S)-1-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-)a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-2-(3-羟基吡咯烷-1-基)乙烷-1-酮
参照实施例14,以2-氯-1-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙烷-1-酮和R-吡咯烷-3-醇为原料制得。LCMS ESI(+)m/z:529.3。1H NMR(400MHz,DMSO)δ11.01(s,1H),8.92(s,1H),8.61(s,1H),7.55(s,1H),7.29(d,J=8.4Hz,1H),7.02(d,J=8.4Hz,1H),5.42(s,2H),5.13(d,J=3.0Hz,2H),4.53(d,J=12.6Hz,1H),4.23–4.11(m,2H),3.43(s,1H),3.28(d,J=9.2Hz,2H),3.06(dt,J=14.1,9.9Hz,3H),2.83(dt,J=13.2,10.5Hz,2H),2.63(d,J=10.4Hz,1H),2.42–2.38(m,1H),1.98(dd,J=12.9,7.2Hz,1H),1.83(d,J=12.4Hz,2H),1.80–1.41(m,4H),1.38(d,J=7.0Hz,6H).
实施例59
2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-1- (3-氟氮杂环丁烷-1-基)乙烷-1-酮
参照实施例5,以2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸和3-氟氮杂环丁烷为原料制得。LCMS ESI(+)m/z:517.3。1H NMR(400MHz,MeOD)δ8.75(s,1H),8.49(s,1H),7.62(s,1H),7.31(d,J=8.4Hz,1H),7.09(dd,J=8.4,1.4Hz,1H),5.46(d,J=3.3Hz,2H),5.37–5.23(m,1H),5.20–5.16(m,2H),4.70–4.54(m,3H),4.41–4.28(m,2H),4.12–4.03(m,1H),3.19(d,J=11.6Hz,2H),3.11(dd,J=14.1,7.1Hz,1H),2.74–2.65(m,1H),2.46(s,2H),1.96(t,J=10.3Hz,4H),1.45(d,J=7.0Hz,6H).
实施例60
(S)-2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-1-(3-羟基吡咯烷-1-基)乙烷-1-酮
参照实施例5,以2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸和(S)-吡咯烷-3-醇为原料制得。LCMS ESI(+)m/z:529.3。1H NMR(400MHz,MeOD)δ8.75(s,1H),8.49(s,1H),7.65(s,1H),7.33(d,J=8.4Hz,1H),7.11(d,J=8.3Hz,1H),5.47(s,2H),5.20–5.17(m,2H),4.64–4.43(m,3H),3.73(d,J=4.2Hz,1H),3.65(dd,J=9.0,4.3Hz,2H),3.60–3.50(m,2H),3.47(d,J=8.9Hz,2H),3.13(dd,J=14.0,7.0Hz,1H),2.84(d,J=5.3Hz,3H),2.06(dd,J=29.2,9.1Hz,5H),1.45(d,J=7.0Hz,6H).
实施例61
(R)-2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-1-(3-羟基吡咯烷-1-基)乙烷-1-酮
参照实施例5,以2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸和(R)-吡咯烷-3-醇为原料制得。LCMS ESI(+)m/z:529.3。1H NMR(400MHz,MeOD)δ8.75(s,1H),8.50(d,J=10.3Hz,1H),7.63(s,1H),7.32(d,J=8.4Hz,1H),7.10(d,J=8.4Hz,1H),5.46(s,2H),5.20–5.17(m,2H),4.95(s,1H),4.58(s,1H),4.46(d,J=24.8Hz,1H),3.68(dd,J=8.6,4.6Hz,2H),3.59(d,J=8.5Hz,1H),3.52(s,2H),3.33(s,1H),3.11(dd,J=13.9,6.7Hz,1H),2.68(dd,J=55.1,7.3Hz,3H),2.04(ddd,J=26.3,11.3,6.6Hz,6H),1.45(d,J=7.0Hz,6H).
实施例62
(R)-2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-1-(3-氟吡咯烷-1-基)乙烷-1-酮
参照实施例5,以2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸和(R)-3-氟吡咯烷为原料制得。LCMS ESI(+)m/z:531.3。1H NMR(400MHz,MeOD)δ8.75(s,1H),8.49(s,1H),7.66(s,1H),7.35(d,J=8.4Hz,1H),7.12(d,J=8.5Hz,1H),5.47(t,J=3.0Hz,2H),5.40–5.24(m,1H),5.20–5.15(m,2H),4.04–3.89(m,2H),3.89–3.68(m,3H),3.59(tdd,J=16.6,11.2,5.6Hz,4H),3.14(dd,J=14.1,7.0Hz,1H),3.04(s,2H),2.95–2.88(m,1H),2.37–2.26(m,1H),2.13(d,J=19.1Hz,4H),1.46(d,J=7.0Hz,6H).
实施例63
2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-1-(吡咯烷-1-基)乙烷-1-酮
参照实施例5,以2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸和吡咯烷为原料制得。LCMS ESI(+)m/z:513.3。1H NMR(400MHz,MeOD)δ8.75(s,1H),8.49(s,1H),7.64(s,1H),7.33(d,J=8.4Hz,1H),7.10(d,J=8.5Hz,1H),5.46(s,2H),5.18(t,J=3.0Hz,2H),3.65(s,2H),3.54(t,J=6.8Hz,2H),3.48(t,J=6.9Hz,2H),3.41–3.36(m,2H),3.12(dt,J=14.0,7.0Hz,1H),2.85–2.64(m,3H),2.13–1.98(m,6H),1.92(dd,J=13.5,6.7Hz,2H),1.45(d,J=7.0Hz,6H).
实施例64
2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-N-异丙基乙酰胺
参照实施例5,以2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸和异丙胺为原料制得。LCMS ESI(+)m/z:501.3。1H NMR(400MHz,MeOD)δ8.65(s,1H),8.39(s,1H),7.53(s,1H),7.21(s,1H),7.00(d,J=8.4Hz,1H),5.37(s,2H),5.09(s,2H),4.00–3.90(m,1H),3.10(s,2H),3.01(dd,J=13.8,6.9Hz,3H),2.59(s,1H),2.39(s,2H),1.85(s,4H),1.35(d,J=7.0Hz,6H),1.10(d,J=6.4Hz,6H).
实施例65
2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)-N-(2,2,2-三氟乙基)乙酰胺
参照实施例5,以2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸和2,2,2-三氟乙烷-1-胺为原料制得。LCMS ESI(+)m/z:541.3。1H NMR(400MHz,MeOD)δ8.75(s,1H),8.49(s,1H),7.63(s,1H),7.31(d,J=8.4Hz,1H),7.10(dd,J=8.4,1.4Hz,1H),5.46(t,J=3.0Hz,2H),5.23–5.15(m,2H),4.02–3.98(m,1H),3.95(d,J=9.3Hz,1H),3.22(s,2H),3.14–3.05(m,3H),2.71–2.61(m,1H),2.42(t,J=10.4Hz,2H),1.96(dt,J=27.5,8.2Hz,4H),1.45(d,J=7.0Hz,6H).
实施例66
2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-4-氟-3-异丙基-1H-吡咯并[2,3-c]吡啶-5-基)哌啶-1-基)乙酰胺
参照实施例2,以6-(4-氟-3-异丙基-5-(哌啶-4-基)-1H-吡咯并[2,3-c]吡啶-2-基)-7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶和2-溴乙酰胺为原料制得。LCMS ESI(+)m/z:478.2。1H NMR(400MHz,MeOD)δ8.96(s,1H),8.39(s,1H),8.17(s,1H),5.41(s,2H),4.63(s,2H),3.29(s,2H),2.89(m,1H),2.78(m,1H),2.51-2.31(m,4H),1.74-1.49(m,4H),1.41(d,J=7.0Hz,6H).
实施例67
2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸
参照实施例2,以叔丁基2-(4-(2-(8,9-二氢-7H-环戊[c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)哌啶-1-基)乙酸酯和盐酸的二氧六环溶液为原料制得。LCMS ESI(+)m/z:460.2。1H NMR(400MHz,MeOD)δ8.76(s,1H),8.49(s,1H),7.67(s,1H),7.35(d,J=8.4Hz,1H),7.11(d,J=8.4Hz,1H),5.47(t,J=3.0Hz,2H),5.18(t,J=3.2Hz,2H),3.74(d,J=11.3Hz,2H),3.66(s,2H),3.13(dt,J=14.0,7.0Hz,3H),2.97(s,1H),2.16(d,J=14.9Hz,4H),1.46(d,J=7.0Hz,6H).
实施例68
2-(4-(3-异丙基-2-(6-甲基-7-氧代-6,7-二氢噻吩并[2,3-c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)乙酰胺
参照实施例2,以4-(3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-6-甲基二烯基[2,3-c]吡啶-7(6H)-酮和2-溴乙酰胺为原料制得。LCMS ESI(+)m/z:463.2。1H NMR(400MHz,DMSO)δ10.92(s,1H),8.05(d,J=5.2Hz,1H),7.62(s,1H),7.54(s,1H),7.25(d,J=8.3Hz,2H),7.14(s,1H),7.08(d,J=5.2Hz,1H),7.01(dd,J=8.4,1.3Hz,1H),3.63(s,3H),3.04–2.88(m,6H),2.19(t,J=10.0Hz,2H),1.86–1.76(m,4H),1.35(d,J=7.0Hz,6H).
实施例69
2-(4-(2-(7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-4-氟-3-异丙基-1H-吡咯并[2,3-c]吡啶-5-基)哌嗪-1-基)乙酰胺
参照实施例2,以6-(4-氟-3-异丙基-5-(哌嗪-1-基)-1H-吡咯并[2,3-c]吡啶-2-基)-7,9-二氢呋喃[3,4-c][1,2,4]三唑并[1,5-a]吡啶和2-溴乙酰胺为原料制得。LCMS ESI(+)m/z:479.2。1H NMR(400MHz,MeOD)δ8.96(s,1H),8.36(s,1H),8.16(s,1H),5.42(s,2H),4.65(s,2H),3.32(t,11.3Hz,4H),3.29(s,2H),2.78(m,1H),2.56(t,J=11.2Hz,4H),1.40(d,J=7.2Hz,6H).
实施例70
2-(4-(3-异丙基-2-(6-甲基-7-氧代-6,7-二氢-1H-吡咯并[2,3-c]吡啶-4-基)-1H-吲哚-5-基)哌啶-1-基)-N-甲基乙酰胺
参照实施例3,以4-(3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-6-甲基-1H-吡咯并[2,3-c]吡啶-7(6H)-酮和2-氯-N-甲基乙酰胺为原料制得。LCMS ESI(+)m/z:460.2。1H NMR(400MHz,DMSO)δ12.13(s,1H),10.79(s,1H),7.73(q,J=4.6Hz,1H),7.52(d,J=1.5Hz,1H),7.33(t,J=2.7Hz,1H),7.28–7.19(m,2H),6.97(dd,J=8.4,1.6Hz,1H),6.19(t,J=2.4Hz,1H),3.59(s,3H),3.16–3.10(m,1H),2.91(d,J=12.6Hz,4H),2.65(d,J=4.7Hz,3H),2.55(dq,J=10.6,3.3Hz,1H),2.18(td,J=11.3,2.8Hz,2H),1.91–1.72(m,4H),1.37(d,J=7.0Hz,6H).
实施例71
4-(3-异丙基-5-(1-(氧杂环丁烷-3-基)哌啶-4-基)-1H-吲哚-2-基)-6-甲基-1H-吡咯并[2,3-c]吡啶-7(6H)-酮
参照实施例6,以4-(3-异丙基-5-(哌啶-4-基)-1H-吲哚-2-基)-6-甲基-1H-吡咯并[2,3-c]吡啶-7(6H)-酮和氧杂环丁烷-3-酮为原料制得。LCMS ESI(+)m/z:445.2。1H NMR(400MHz,DMSO)δ12.13(s,1H),10.78(s,1H),7.52(d,J=1.5Hz,1H),7.33(t,J=2.7Hz,1H),7.27–7.19(m,2H),6.97(dd,J=8.4,1.6Hz,1H),6.19(t,J=2.3Hz,1H),4.56(t,J=6.4Hz,2H),4.46(t,J=6.1Hz,2H),3.59(s,3H),3.41(s,1H),3.16–3.10(m,1H),2.82(dt,J=11.6,3.0Hz,2H),2.57(tt,J=11.2,4.1Hz,1H),1.88(td,J=11.2,2.6Hz,2H),1.76(dtd,J=28.0,12.5,3.6Hz,4H),1.37(d,J=7.0Hz,6H).
实施例72
6-(5-(4-(3,3-二氟氮杂环丁烷-1-基)环己基)-3-异丙基-1H-吲哚-2-基)-7,9-二氢呋喃并[3,4- c][1,2,4]三唑并[1,5-a]吡啶
步骤A:3-异丙基-5-(1,4-二氧杂螺[4.5]癸-8-基)-2-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-1H-吲哚(化合物72.2)
将化合物2-溴-3-异丙基-5-(1,4-二氧杂螺[4.5]癸-8-基)-1H-吲哚(225mg,0.6mmol)、4,4,5,5-四甲基-1,3,2-二氧杂硼烷(768mg,6mmol)、双(苄腈)氯化钯(II)(23g,0.06mmol)、2-二环己基膦基-2′,6′-二甲氧基联苯基(30mg,0.072mmol)和三乙胺(182mg,1.8mmol)溶解在1,4二氧六环(10ml)中。反应体系氩气置换3次。反应液85℃下搅拌反应3小时。反应液减压浓缩,柱层析(MeOH/DCM(2%))纯化得到产物72.2(213mg,产率:83%)。LCMS ESI(+)m/z:426(M+1)。
步骤B:6-(3-异丙基-5-(1,4-二氧阿司匹林[4.5]癸-8-基)-1H-吲哚-2-基)-7,9-二氢呋喃并[3,4-c][1,2,4]三唑并[1,5-a]吡啶(化合物72.3)
将化合物72.2(128mg,0.3mmol),6-溴-7,9-二氢氟[3,4-c][1,2,4]三唑并[1,5-a]吡啶(72mg,0.3mmol),1,1'-双二苯基膦二茂铁二氯化钯(22mg,0.03mmol),磷酸钾(190mg,0.9mmol)溶于1,4-二氧六环(5ml)和水(1ml)中。反应体系氩气置换3次。反应液80℃下搅拌反应16小时。反应液过滤,滤液减压浓缩,柱层析(MeOH/DCM(2%))纯化得到产物72.3(34mg,产率:25%)。LCMS ESI(+)m/z:459(M+1)。
步骤C:4-(2-(7,9-二氢糠[3,4-c][1,2,4]三唑并[1,5-a]吡啶-6-基)-3-异丙基-1H-吲哚-5-基)环己烷-1-酮(化合物72.4)
将化合物72.3(34mg,0.074mmol)溶于二氯甲烷(4ml)中。室温下将三氟乙酸(1ml)滴至反应体系中。反应液室温下下搅拌反应2小时。反应液用二氯甲烷萃取,无水硫酸钠干燥,过滤,减压浓缩,得到产物72.4(30mg,产率:98%)。LCMS ESI(+)m/z:415(M+1)。
步骤D:6-(5-(4-(3,3-二氟氮杂环丁烷-1-基)环己基)-3-异丙基-1H-吲哚-2-基)-7,9-二氢呋喃并[3,4-c][1,2,4]三唑并[1,5-a]吡啶(化合物72)
将化合物72.4(30mg,0.072mmol)、3,3-二氟氮杂环丁烷盐酸盐(93mg,0.72mmol)、醋酸(5drops)溶于DMF(3ml)和四氢呋喃(3ml)中。反应液室温下搅拌反应1小时。然后在0℃下往反应液中缓慢加入氰基硼氢化钠(23mg,0.36mmol),反应液继续在室温下搅拌反应1小 时。反应液用乙酸乙酯萃取,无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到产物72(7.32mg,产率:21%)。
LCMS ESI(+)m/z:492(M+1)。
1H NMR(400MHz,DMSO)δ10.98(s,1H),8.91(s,1H),8.61(s,1H),7.54(s,1H),7.27(d,J=8.4Hz,1H),7.01(dd,J=8.4,1.2Hz,1H),5.42(s,2H),5.13(t,J=3.2Hz,2H),3.57(t,J=10.8Hz,4H),3.04(dt,J=14.0,7.2Hz,1H),2.57(d,J=12.4Hz,1H),2.22(s,1H),1.86(d,J=11.2Hz,4H),1.52(dd,J=23.2,12.4Hz,2H),1.38(d,J=7.2Hz,6H),1.15(dd,J=22.8,11.4Hz,2H).
实施例73
4-(5-(4-(3,3-二氟氮杂环丁烷-1-基)环己基)-3-异丙基-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊烷[c]吡啶-1-酮
步骤A:4-(3-异丙基-5-(1,4-二氧杂螺[4.5]癸-8-基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊基[c]吡啶-1-酮(化合物73.1)
将化合物72.1(250mg,0.66mmol),2-甲基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)-2,5,6,7-四氢-1H-环戊二烯[c]吡啶-1-酮(364mg,1.32mmol)、碳酸钾(274mg,1.98mmol)、三(二亚苄基丙酮)二钯(60mg,0.065mmol)和2-二环己基膦-2',4',6'-三异丙基联苯(63mg,0.13mmol)溶解在1,4-二氧六环(12ml)和水(2ml)中。反应体系氩气置换3次。反应液80℃下搅拌反应4小时。反应液减压浓缩,柱层析(PE/EA(50%))纯化得到产物73.1(156mg,产率:53%)。LCMS ESI(+)m/z:447(M+1)。
步骤B:4-(3-异丙基-5-(4-氧代环己基)-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊烷[c]吡啶 -1-酮(化合物73.2)
将化合物73.1(156mg,0.35mmol)溶于二氯甲烷(6ml)中。室温下将三氟乙酸(2ml)滴至反应体系中。反应液室温下下搅拌反应16小时。反应液用二氯甲烷萃取,无水硫酸钠干燥,过滤,减压浓缩,得到产物73.2(120mg,产率:85%)。LCMS ESI(+)m/z:403(M+1)。
步骤C:4-(5-(4-(3,3-二氟氮杂环丁烷-1-基)环己基)-3-异丙基-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊基[c]吡啶-1-酮(化合物73)
将化合物73.2(40mg,0.1mmol)、3,3-二氟氮杂环丁烷盐酸盐(129mg,1.0mmol)、醋酸(5drops)溶于DMF(3ml)和四氢呋喃(3ml)中。反应液室温下搅拌反应1小时。然后在0℃下往反应液中缓慢加入氰基硼氢化钠(31mg,0.5mmol),反应液继续在室温下搅拌反应1小时。反应液用乙酸乙酯萃取,无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到产物73(5.70mg,产率:12%)。LCMS ESI(+)m/z:480(M+1)。
1H NMR(400MHz,DMSO)δ10.66(s,1H),7.55(s,1H),7.46(s,1H),7.19(d,J=8.4Hz,1H),6.93(dd,J=8.4,1.6Hz,1H),3.56(t,J=12.4Hz,4H),3.50(s,3H),3.02–2.93(m,1H),2.71(dd,J=16.0,8.0Hz,4H),2.54(s,1H),2.20(t,J=11.2Hz,1H),2.01–1.93(m,2H),1.84(d,J=10.8Hz,4H),1.50(dd,J=23.2,12.4Hz,2H),1.33(d,J=7.2Hz,6H),1.14(dd,J=22.0,11.2Hz,2H).
实施例74
(S)-4-(5-(4-(3-氟吡咯烷-1-基)环己基)-3-异丙基-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊基[c]吡啶-1-酮
步骤A:(S)-4-(5-(4-(3-氟吡咯烷-1-基)环己基)-3-异丙基-1H-吲哚-2-基)-2-甲基-2,5,6,7-四氢-1H-环戊基[c]吡啶-1-酮(化合物74)
将化合物73.2(40mg,0.1mmol)、(3S)-3-氟吡咯烷盐酸盐(129mg,1.0mmol)、醋酸(5drops)溶于DMF(3ml)和四氢呋喃(3ml)中。反应液室温下搅拌反应1小时。然后在0℃下往反应液中缓慢加入氰基硼氢化钠(31mg,0.5mmol),反应液继续在室温下搅拌反应1小时。反应 液用乙酸乙酯萃取,无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化得到产物74(9.71mg,产率:20%)。LCMS ESI(+)m/z:476(M+1)。
1H NMR(400MHz,DMSO)δ10.66(d,J=5.6Hz,1H),8.14(s,1H),7.55(s,1H),7.46(d,J=5.6Hz,1H),7.20(dd,J=8.4,4.4Hz,1H),6.94(d,J=8.4Hz,1H),5.25(d,J=56.4Hz,1H),3.50(s,3H),2.98(dt,J=14.4,7.2Hz,2H),2.85(s,1H),2.72(dd,J=16.0,8.0Hz,4H),2.62(s,1H),2.56(d,J=16.4Hz,1H),2.44–2.36(m,1H),2.25–1.82(m,9H),1.65–1.49(m,3H),1.40(s,1H),1.33(dd,J=7.2,1.2Hz,6H).
实施例75、实施例76
其中*标记碳原子为S构型、R构型或其混合物;#标记碳原子为S构型、R构型或其混合物
(S)-6-(5-(4-(3-氟吡咯烷-1-基)环己基)-3-异丙基-1H-吲哚-2-基)-7,9-二氢呋喃并[3,4-c][1,2,4]三唑并[1,5-a]吡啶
步骤A:(S)-6-(5-(4-(3-氟吡咯烷-1-基)环己基)-3-异丙基-1H-吲哚-2-基)-7,9-二氢呋喃并[3,4-c][1,2,4]三唑并[1,5-a]吡啶
将化合物72.4(20mg,0.048mmol)、(3S)-3-氟吡咯烷盐酸盐(62mg,0.48mmol)、醋酸(5drops)溶于DMF(3ml)和四氢呋喃(3ml)中。反应液室温下搅拌反应1小时。然后在0℃下往反应液中缓慢加入氰基硼氢化钠(15mg,0.24mmol),反应液继续在室温下搅拌反应1小时。反应液用乙酸乙酯萃取,无水硫酸钠干燥,过滤,减压浓缩,反相制备纯化(制备柱:Ultimate XB-C18,21.2*250mm,10μm;洗脱相:流动相0.1%FA in H2O/ACN,洗脱梯度(ACN的比例)10%(0-3min),25-45%(3-16min),95%(16-20min))得到产物75(保留时间是4.03min,4.2mg,产率:18%)和产物76(保留时间是4.10min,3.3mg,产率:14%)。LCMS ESI(+)m/z:488(M+1)。
其中,化合物75为中的一个立体异构体。
化合物76为中的另一个立体异构体。
75:1H NMR(400MHz,DMSO)δ10.98(s,1H),8.92(s,1H),8.62(s,1H),8.15(s,1H),7.55(s,1H),7.28(d,J=8.4Hz,1H),7.03(d,J=7.6Hz,1H),5.43(s,2H),5.35–5.16(m,1H),5.13(s,2H),3.06(dd,J=14.0,7.2Hz,2H),2.94(d,J=7.2Hz,2H),2.83(dd,J=11.6,4.8Hz,1H),2.75(dd,J=10.4,3.2Hz,1H),2.61–2.58(m,1H),2.31–2.23(m,1H),2.22–2.11(m,1H),2.07(d,J=12.4Hz,2H),2.03–1.83(m,4H),1.57(dd,J=24.4,12.0Hz,2H),1.39(d,J=7.2Hz,6H).
76:1H NMR(400MHz,DMSO)δ10.97(s,1H),8.91(s,1H),8.61(s,1H),8.14(s,1H),7.54(s,1H),7.30(d,J=8.4Hz,1H),7.05(d,J=8.4Hz,1H),5.42(s,2H),5.29(d,J=54.8Hz,1H),5.13(s,2H),3.05(dt,J=14.0,7.2Hz,2H),2.94(d,J=16.8Hz,2H),2.71(s,1H),2.18(d,J=20.8Hz,2H),2.08–1.85(m,6H),1.63(t,J=15.2Hz,4H),1.38(d,J=7.2Hz,6H).
效果实施例一、本申请化合物细胞水平药效评价
1、本申请化合物对人TLR7激活通路的拮抗作用
1.1实验材料及仪器
1.1.1材料和试剂
1)HEK-Blue hTLR7细胞(InvivoGen)
2)HEK碱性磷酸酶检测培养基(HEK-BlueTM Detection,InvivoGen)
3)DMEM高糖培养基(DMEM,Gibco)
4)杀稻瘟菌素(Blasticidin,InvivoGen)
5)博来霉素(ZeocinTM,InvivoGen)
6)新霉素(NormocinTM,InvivoGen)
7)青霉素-链霉素溶液(PS,Penicillin-Streptomycin,Gibco)
8)胎牛血清(FBS,Gibco)
9)磷酸盐缓冲液(PBS,Gibco)
10)蕾西莫特(R848,MCE)
11)DMSO(Sigma)
1.1.2耗材和仪器
1)384-孔平底细胞培养板(Corning)
2)震板仪(QILINBEIER)
3)15毫升离心管(Corning)
4)离心机(Eppendorf)
5)CO2恒温细胞培养箱(Thermo Scientific)
6)显微镜(OLYMPUS)
7)自动细胞计数器(CountessII,Gibco)
8)酶标仪(Multiplate reader,PerkinElmer)
9)声波液体处理器(ECHO,LABCYTE)
1.2实验步骤
本实验的目的是通过HEK碱性磷酸酶检测培养基检测化合物在HEK-BlueTM hTLR7细胞中的拮抗活性。
HEK-Blue hTLR7细胞培养于完全培养基(含10%灭活胎牛血清,100ug/mL新霉素,100ug/mL博来霉素,10ug/mL杀稻瘟菌素,0.5%青霉素-链霉素溶液的DMEM高糖培养基)中。取生长良好,细胞密度为80-90%的细胞,移除生长培养基上清,使用预热的PBS清洗细胞一次,再加入预热的PBS,并将细胞至于37℃培养消化。加入完全培养基终止消化,将细胞悬液转移至15mL离心管,离心去除上清。加入检测培养基,分散细胞并计数,制备浓度为3.3×105/mL细胞悬液。将细胞悬液加至384孔细胞培养板中,30uL/孔,每孔细胞数为10000个/孔。
使用100%DMSO将激动剂R848制成储配液,浓度为40mM;将受试化合物使用100%DMSO制备为储备液,浓度为10mM。在96孔配药板上将受试化合物的储备液用100%DMSO配置测试浓度并做3倍梯度稀释至600、200、66.67、22.22、7.41、2.47、0.82、0.27、0.09、0.03uM;阴性对照孔为100%DMSO;用100%DMSO将R848的40mM储备液稀释至480uM。配药板在震板仪(320rpm)上震荡混匀,备用。
将配药板离心,使用ECHO添加测试溶液至含有细胞悬液的384孔细胞培养板中:将受试化合物加入到含有细胞悬液的孔中,50nL/孔,化合物终浓度依次为1000、333.3、111.1、37、12.3、4.1、1.3、0.45、0.15、0.05nM,并加入激动剂R848,50nL/孔,作为化合物组;将100%DMSO加入到含有细胞悬液的孔中,50nL/孔,并加入激动剂R848,50nL/孔,作为阳性对照组;将100%DMSO加入到含有细胞悬液的孔中,100nL/孔,作为阴性对照组。激动剂R848在各孔中的终浓度为0.8uM,DMSO在各孔中的终浓度为0.33%。而后将细胞板放置于37℃,5%CO2培养箱培养16小时。随后取出细胞板并于1000rpm离心1分钟,使用酶标仪读取每孔在620nm处的光密度值OD620
使用以下公式计算受试化合物的拮抗活性:抑制率(%)=100-(OD620化合物-OD620阳性对照均值)/(OD620阴性对照均值-OD620阳性对照均值)×100,用Graphpad Prism软件绘制受试化合物的计量作用曲线,并计算抑制率达到50%时的化合物浓度,即拮抗活性IC50值,见表1。
2、本申请化合物对人TLR8激活通路的拮抗作用
2.1实验材料及仪器
2.1.1材料和试剂
1)HEK-Blue hTLR8细胞(InvivoGen)
2)HEK碱性磷酸酶检测培养基(HEK-BlueTM Detection,InvivoGen)
3)DMEM高糖培养基(DMEM,Gibco)
4)杀稻瘟菌素(Blasticidin,InvivoGen)
5)博来霉素(ZeocinTM,InvivoGen)
6)新霉素(NormocinTM,InvivoGen)
7)青霉素-链霉素溶液(PS,Penicillin-Streptomycin,Gibco)
8)胎牛血清(FBS,Gibco)
9)磷酸盐缓冲液(PBS,Gibco)
10)蕾西莫特(R848,MCE)
11)DMSO(Sigma)
2.1.2耗材和仪器
1)384-孔平底细胞培养板(Corning)
2)震板仪(QILINBEIER)
3)15毫升离心管(Corning)
4)离心机(Eppendorf)
5)CO2恒温细胞培养箱(Thermo Scientific)
6)显微镜(OLYMPUS)
7)自动细胞计数器(CountessII,Gibco)
8)酶标仪(Multiplate reader,PerkinElmer)
9)声波液体处理器(ECHO,LABCYTE)
2.2实验步骤
本实验的目的是通过HEK碱性磷酸酶检测培养基检测化合物在HEK-BlueTM hTLR8细胞中的拮抗活性。
HEK-Blue hTLR8细胞培养于完全培养基(含10%灭活胎牛血清,100ug/mL新霉素,100ug/mL博来霉素,30ug/mL杀稻瘟菌素,0.5%青霉素-链霉素溶液的DMEM高糖培养基)中。取生长良好,细胞密度为80-90%的细胞,移除生长培养基上清,使用预热的PBS清洗细胞一次,再加入预热的PBS,并将细胞至于37℃培养消化。加入完全培养基终止消化,将细胞悬液转移至15mL离心管,离心去除上清。加入检测培养基,分散细胞并计数,制备浓度为3.3×105/mL细胞悬液。将细胞悬液加至384孔细胞培养板中,30uL/孔,每孔细胞数为10000个/孔。
使用100%DMSO将激动剂R848制成储配液,浓度为40mM;将受试化合物使用100%DMSO制备为储备液,浓度为10mM。在96孔配药板上将受试化合物的储备液用100%DMSO配置测试浓度并做3倍梯度稀释至600、200、66.67、22.22、7.41、2.47、0.82、0.27、0.09、0.03uM;阴性对照孔为100%DMSO;用100%DMSO将R848的40mM储备液稀释至1.8mM。配药板在震板仪(320rpm)上震荡混匀,备用。
将配药板离心,使用ECHO添加测试溶液至含有细胞悬液的384孔细胞培养板中:将受试化合物加入到含有细胞悬液的孔中,50nL/孔,化合物终浓度依次为1000、333.3、111.1、37、12.3、4.1、1.3、0.45、0.15、0.05nM,并加入激动剂R848,50nL/孔,作为化合物组;将100%DMSO加入到含有细胞 悬液的孔中,50nL/孔,并加入激动剂R848,50nL/孔,作为阳性对照组;将100%DMSO加入到含有细胞悬液的孔中,100nL/孔,作为阴性对照组。激动剂R848在各孔中的终浓度为3uM,DMSO在各孔中的终浓度为0.33%。而后将细胞板放置于37℃,5%CO2培养箱培养16小时。随后取出细胞板并于1000rpm离心1分钟,使用酶标仪读取每孔在620nm处的光密度值OD620
使用以下公式计算受试化合物的拮抗活性:抑制率(%)=100-(OD620化合物-OD620阳性对照均值)/(OD620阴性对照均值-OD620阳性对照均值)×100,用Graphpad Prism软件绘制受试化合物的计量作用曲线,并计算抑制率达到50%时的化合物浓度,即拮抗活性IC50值,见表1。
3、本申请化合物对人TLR9激活通路的拮抗作用
3.1实验材料及仪器
3.1.1材料和试剂
1)HEK-Blue hTLR9细胞(InvivoGen)
2)HEK碱性磷酸酶检测培养基(HEK-BlueTM Detection,InvivoGen)
3)DMEM高糖培养基(DMEM,Gibco)
4)杀稻瘟菌素(Blasticidin,InvivoGen)
5)博来霉素(ZeocinTM,InvivoGen)
6)新霉素(NormocinTM,InvivoGen)
7)青霉素-链霉素溶液(PS,Penicillin-Streptomycin,Gibco)
8)胎牛血清(FBS,Gibco)
9)磷酸盐缓冲液(PBS,Gibco)
10)CpG ODN2006(InvivoGen)
11)DMSO(Sigma)
3.1.2耗材和仪器
1)384-孔平底细胞培养板(Corning)
2)震板仪(QILINBEIER)
3)15毫升离心管(Corning)
4)离心机(Eppendorf)
5)CO2恒温细胞培养箱(Thermo Scientific)
6)显微镜(OLYMPUS)
7)自动细胞计数器(CountessII,Gibco)
8)酶标仪(Multiplate reader,PerkinElmer)
9)声波液体处理器(ECHO,LABCYTE)
3.2实验步骤
本实验的目的是通过HEK碱性磷酸酶检测培养基检测化合物在HEK-BlueTM hTLR9细胞中的拮抗活性。
HEK-Blue hTLR9细胞培养于完全培养基(含10%灭活胎牛血清,100ug/mL新霉素,100ug/mL 博来霉素,10ug/mL杀稻瘟菌素,0.5%青霉素-链霉素溶液的DMEM高糖培养基)中。取生长良好,细胞密度为80-90%的细胞,移除生长培养基上清,使用预热的PBS清洗细胞一次,再加入预热的PBS,并将细胞至于37℃培养消化。加入完全培养基终止消化,将细胞悬液转移至15mL离心管,离心去除上清。加入检测培养基,分散细胞并计数,制备浓度为5×105/mL细胞悬液。将细胞悬液加至384孔细胞培养板中,30uL/孔,每孔细胞数为15000个/孔。
使用试剂盒自带的无菌水将激动剂ODN2006制成储配液,浓度为300uM;将受试化合物使用100%DMSO制备为储备液,浓度为30mM。在96孔配药板上将受试化合物的储备液用100%DMSO配置测试浓度并做3倍梯度稀释至30000、10000、3333.33、1111.11、370.37、123.46、41.15、13.72、4.57、1.52uM;阴性对照孔为100%DMSO。配药板在震板仪(320rpm)上震荡混匀(ODN2006除外),备用。
将配药板离心,使用ECHO添加测试溶液至含有细胞悬液的384孔细胞培养板中:将受试化合物加入到含有细胞悬液的孔中,50nL/孔,化合物终浓度依次为50000、16666.67、5555.56、1851.85、617.28、205.76、68.59、22.86、7.62、2.54nM,并加入激动剂ODN2006储备液,100nL/孔,作为化合物组;将100%DMSO加入到含有细胞悬液的孔中,50nL/孔,并加入激动剂ODN2006,100nL/孔,作为阳性对照组;将细胞悬液孔作为阴性对照组。激动剂ODN2006在各孔中的终浓度为1uM,DMSO在各孔中的终浓度为0.167%。而后将细胞板放置于37℃,5%CO2培养箱培养16小时。随后取出细胞板并于1000rpm离心1分钟,使用酶标仪读取每孔在620nm处的光密度值OD620
使用以下公式计算受试化合物的拮抗活性:抑制率(%)=100-(OD620化合物-OD620阳性对照均值)/(OD620阴性对照均值-OD620阳性对照均值)×100,用Graphpad Prism软件绘制受试化合物的计量作用曲线,并计算抑制率达到50%时的化合物浓度,即拮抗活性IC50值,见表1。
其中,除测量实施例化合物外,也测量了对照物Enpatoran和Afimetoran的IC50值。
表1本发明化合物在HEK-Blue hTLR7/8/9细胞中的活性


结论:本发明化合物对HEK-Blue-hTLR7/8细胞有明显拮抗作用,同时对HEK-Blue hTLR9细胞无明显拮抗作用。
效果实施例二、本申请化合物对动物体内IL-6抑制实验
实验材料:化合物溶液的配制:Gardiquimod加入Saline涡旋、超声至均匀,配置成终浓度为0.2mg/ml的Gardiquimod澄清溶液。受试化合物用5%DMSO、3.7%HCl、PEG400、Saline涡旋、超声至均匀,配置成终浓度为0.03mg/ml的受试物澄清溶液(终浓度含5%DMSO+1%(3.7%HCL)+40%PEG400+54%Saline)。
实验动物:BALB/c雌鼠,北京维通利华实验动物技术有限公司,8周龄。
实验方法:动物随机分为空白组、对照组和实验组,每组6只。动物分组后,给予相应的本发明化合物(0.3mg/kg)及对照物Afimetoran(0.3mg/kg);给药后0.5h,腹腔注射Gardiquimod 2mg/kg。Gardiquimod给药后1.5h达到实验终点。实验终点,麻醉小鼠,心脏取血至抗凝管中,采集血浆,进行IL-6细胞因子检测。通过对照组和实验组IL-6血浆浓度,计算IL-6抑制率。计算公式:抑制率=(Avg(Vehicle)-Avg(Group))/(Avg(vehicle)-Avg(NC))*100%。其中:Avg(Vehicle):对照组血浆IL-6;Avg(Group):实验组血浆IL-6;Avg(NC):空白组血浆IL-6。
表2部分发明化合物动物体内IL-6抑制实验
结论:本发明化合物对IL-6有明显的抑制作用。
效果实施例三、本申请化合物对hERG通道电流的影响
人类ether-a-go-go-related基因(hERG)编码的快速激活钾通道是参与心肌动作电位3期复极的形成的重要离子通道。药物阻断hERG通道能够导致心脏复极延长,心电图表现为QT间期延长,称为长QT间期综合征。药物引起的心室延迟复极在某些情况下可能引发致命性心律失常-尖端扭转型 室性心动过速。
本实验采用了全细胞膜片钳技术来研究化合物对hERG钾通道的抑制作用,并评价其引发心室复极毒性的风险。
细胞培养
采用稳定表达hERG钾通道的HEK293细胞系,hERG钾通道细胞购于Creacell公司(货号:A-0320)。
hERG钾通道稳定表达的HEK293细胞系在含有10%胎牛血清及0.8mg/mL G418的DMEM培养基中培养,培养温度为37℃,二氧化碳浓度为5%。
细胞传代:除去旧培养基并用PBS洗一次,然后加入2mL TrypLETM Express溶液,37℃孵育1min左右。当细胞从皿底脱离,加入约5mL 37℃预热的完全培养基。将细胞悬液用吸管轻轻吹打使聚集的细胞分离。将细胞悬液转移至无菌的离心管中,1000rpm离心5min收集细胞。扩增或维持培养,将细胞接种于10cm细胞培养皿,每个细胞培养皿接种细胞量为6×105cells(最终体积:10mL)。
维持细胞的电生理活性,细胞密度必须不能超过80%。
膜片钳检测,试验之前细胞用TrypLETM Express分离,加入培养基终止消化后离心,重悬细胞计数,调整细胞密度为2-3×106cells/mL,然后用室温平衡摇床轻混细胞15-20min上机进行检测。
膜片钳检测
使用全自动膜片钳QPatch 48 X(Sophion)设备进行电生理检测。将制备好的细胞放置于Qpatch工作台的离心机上,使用多次离心/悬浮方法清洗细胞,将细胞培养基置换为细胞外液。取出一个MTP-96板,然后放置到MTP source的位置。取出QPlate芯片,随后将QPlate放置在Qplate source位置。机械手臂扫描MTP-96板以及QPlate芯片条形码,并抓取至测量站。从液体池中吸取细胞内、外液分别加到QPlate芯片的细胞内液池、细胞与受试物池中。在测量站,QPlate上所有测量位点都要经过初始质量控制。质量控制流程包括从离心机的细胞容器中吸取细胞悬液,以及通过压力控制器将细胞定位到芯片孔上,建立高阻封接,形成全细胞记录模式。一旦获得稳定的对照电流基线,即可从受试物MTP-96板中按照浓度梯度吸取受试物施加到细胞上。
全细胞膜片钳记录全细胞hERG钾电流的电压刺激方案如下:当形成全细胞封接后细胞膜电压钳制于-80mV。钳制电压由-80mV除极至-50mV维持0.5s(作为漏电流检测),然后阶跃至30mV维持2.5s,再迅速恢复至-50mV维持4s可以激发出hERG通道的尾电流。每隔10s重复采集数据,观察药物对hERG尾电流的作用。以0.5s的-50mV刺激作为漏电流检测。试验数据由Qpatch进行采集并储存于连接的服务站中。
数据分析
首先将每一个药物浓度作用后的电流和空白对照电流标准化然后计算每一个药物浓度对应的抑制率对每一个浓度计算平均数和标准误,并用以下的方程计算每种化合物的半抑制浓度:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))用以上方程对剂量依赖效应进行非线性拟合,其中C代表受试物浓度,IC50为半抑制浓度,h代表希尔系数。曲线拟合以及IC50的计算利用 Graphpad软件完成。部分化合物测试结果如表3所示:
表3部分实施例化合物对hERG电流的半抑制浓度(IC50)
结果表明:本发明化合物对hERG通道抑制作用较弱。

Claims (18)

  1. 一种如式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物:
    其中,R1
    Ra独立地为C1~C6烷基或C3~C6环烷基;
    X、Y、Z和W独立地为-C(Ra1)(Ra2)-、=CRa3-、-C(=O)-、-NRa4-、=N-、-O-或-S-;
    Ra1、Ra2、Ra3和Ra4独立地为氢、C1~C3烷基、卤素或被1个、2个或3个卤素取代的C1~C3烷基;
    U独立地为N或CH;
    为单键或者双键,相邻的两个不同时为双键;
    环A为3~10元杂碳环或3~10元碳环;
    L1为单键、C1~C6亚烷基或-(C=O)-;
    L2为单键、C1~C6亚烷基或-(C=O)-;
    R2为氢、羟基、氰基、C1~C6烷基、3~10元环烷基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、被1个、2个或3个R21取代的3~10元杂环烷基、被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R23取代的C1~C6烷基或被1个、2个或3个R26取代的5~6元杂芳基;
    R21、R22和R23独立地为羟基、氰基、氧代、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢或C1~C6烷基;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C1~C6烷基、C3~C6环烷基、R271-(C=O)-或被1个、2个或3个卤素取代的C1~C6烷基,R271 独立地为C1~C6烷基;
    所述的3~10元杂碳环中,杂原子的数量为1个、2个或3个,杂原子选自N、O和S中的一种或多种;
    所述的3~10元杂环烷基中,杂原子的数量独立地为1个、2个或3个,杂原子独立地选自N、O和S中的一种或多种;
    所述的5~6元杂芳基中,杂原子的数量独立地为1个、2个、3个或4个,杂原子独立地选自N、O和S中的一种或多种;
    R3为C1~C6烷基或C3~C6环烷基;
    RA为氢或卤素;
    V为C-R4或N,R4为氢、C1~C3烷基、C3~C6环烷基或卤素。
  2. 如权利要求1所述如式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物,其特征在于,所述式II所示的化合物中,环A为3~10元杂碳环。
  3. 如权利要求1所述如式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物,其特征在于,所述式II所示的化合物为如式I所示的化合物;
    其中,R1
    Ra独立地为C1~C6烷基或C3~C6环烷基;
    X、Y、Z和W独立地为-C(Ra1)(Ra2)-、=CRa3-、-C(=O)-、-NRa4-、=N-、-O-或-S-;
    Ra1、Ra2、Ra3和Ra4独立地为氢、C1~C3烷基、卤素或被1个、2个或3个卤素取代的C1~C3烷基;
    U独立地为N或CH;
    为单键或者双键,相邻的两个不同时为双键;
    环A为3~10元杂碳环;
    L1为单键、C1~C6亚烷基或-(C=O)-;
    L2为单键、C1~C6亚烷基或-(C=O)-;
    R2为氢、羟基、氰基、C1~C6烷基、3~10元环烷基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、被1个、2个或3个R21取代的3~10元杂环烷基、被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R23取代的C1~C6烷基或被1个、2个或3个R26取代的5~6元杂芳基;
    R21、R22和R23独立地为羟基、氰基、氧代、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢或C1~C6烷基;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C1~C6烷基、C3~C6环烷基或R271-(C=O)-,R271独立地为C1~C6烷基;
    所述的3~10元杂碳环中,杂原子的数量为1个、2个或3个,杂原子选自N、O和S中的一种或多种;
    所述的3~10元杂环烷基中,杂原子的数量独立地为1个、2个或3个,杂原子独立地选自N、O和S中的一种或多种;
    所述的5~6元杂芳基中,杂原子的数量独立地为1个、2个、3个或4个,杂原子独立地选自N、O和S中的一种或多种;
    R3为C1~C6烷基或C3~C6环烷基;
    R4为氢、C1~C3烷基、C3~C6环烷基或卤素。
  4. 如权利要求1-3中任一项所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物,其特征在于,所述的式II所示的化合物满足下列条件中的一个或多个:
    a)环A中,所述3~10元杂碳环为6~8元杂碳环;
    b)环A中,所述3~10元杂碳环为单环或双环,所述双环为螺环或桥环;
    c)环A中,所述3~10元杂碳环中,杂原子的数量为1个或2个;
    d)环A中,所述3~10元杂碳环中,所述杂原子为N;
    e)L1中,所述C1~C6亚烷基为C1~C4亚烷基;
    f)L2中,所述C1~C6亚烷基为C1~C4亚烷基;
    g)L1和L2不同时为C1~C6亚烷基,且L1和L2不同时为-(C=O)-;
    h)R2中,所述C1~C6烷基为C1~C3烷基;
    i)R2中,所述被1个、2个或3个R23取代的C1~C6烷基中的C1~C6烷基为C1~C3烷基;
    j)R2中,所述3~10元环烷基为3~6元环烷基;
    k)R2中,所述被1个、2个或3个R22取代的3~10元环烷基中的3~10元环烷基为3~6元环烷基;
    l)R2中,所述3~10元杂环烷基为单环或双环,所述双环为螺环或桥环;
    m)R2中,所述3~10元杂环烷基中,杂原子的数量为1个或2个;
    n)R2中,所述3~10元杂环烷基中,杂原子为N和/或O;
    o)R2中,所述3~10元杂环烷基为3~6元杂环烷基;
    p)R2中,所述被1个、2个或3个R21取代的3~10元杂环烷基中的3~10元杂环烷基为单环或双环,所述双环为螺环或桥环;
    q)R2中,所述被1个、2个或3个R21取代的3~10元杂环烷基中的3~10元杂环烷基中,杂原子的数量为1个或2个;
    r)R2中,所述被1个、2个或3个R21取代的3~10元杂环烷基中的3~10元杂环烷基中,杂原子为N和/或S;
    s)R2中,所述被1个、2个或3个R21取代的3~10元杂环烷基中的3~10元杂环烷基为3~6元杂环烷基;
    t)R2中,所述5~6元杂芳基,杂原子的数量为2个;
    u)R2中,所述被1个、2个或3个R26取代的5~6元杂芳基中的5~6元杂芳基中,杂原子的数量为3个或4个;
    v)R2中,所述被1个、2个或3个R26取代的5~6元杂芳基中的5~6元杂芳基中,杂原子为N;
    w)R3中,所述C1~C6烷基为C1~C3烷基;
    x)R3中,所述C3~C6环烷基为C3~C4环烷基;
    y)R4中,所述C1~C3烷基为甲基、乙基、正丙基或异丙基;
    z)R4中,所述C3~C6环烷基为C3~C4环烷基;
    aa)R4中,所述卤素为氟。
  5. 如权利要求4所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物,其特征在于,所述的式II所示的化合物满足下列条件中的一个或多个:
    a)L1中,所述C1~C6亚烷基为亚甲基、亚乙基或所述q端与L2相连接;
    b)L2中,所述C1~C6亚烷基为亚甲基、亚乙基或所述q端与R2相连接;
    c)R2中,所述C1~C6烷基为甲基、乙基、正丙基或异丙基;
    d)R2中,所述被1个、2个或3个R23取代的C1~C6烷基中的C1~C6烷基为甲基、乙基、正丙基或异丙基;
    e)R2中,所述3~10元环烷基为3~4元环烷基;
    f)R2中,所述被1个、2个或3个R22取代的3~10元环烷基中的3~10元环烷基为3~4元环烷基,例如环丙基或环丁基;
    g)R2中,所述3~10元杂环烷基为4~6元杂环烷基;
    h)R2中,所述被1个、2个或3个R21取代的3~10元杂环烷基中的3~10元杂环烷基为4~6元杂环烷基;
    i)R2中,所述5~6元杂芳基为
    j)R2中,所述被1个、2个或3个R26取代的5~6元杂芳基中的5~6元杂芳基为
    k)R3中,所述C1~C6烷基为甲基、乙基、正丙基或异丙基;
    l)R3中,所述C3~C6环烷基为环丙基;
    m)R4中,所述C3~C6环烷基为环丙基。
  6. 如权利要求5所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物,其特征在于,所述的式II所示的化合物满足下列条件中的一个或多个:
    a)R2中,所述3~10元环烷基为环丙基或环丁基;
    b)R2中,所述3~10元杂环烷基为
    c)R2中,所述被1个、2个或3个R21取代的3~10元杂环烷基中的3~10元杂环烷基为
  7. 如权利要求1-3中任一项所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物,其特征在于,所述的式II所示的化合物满足下列条件中的一个或多个:
    a)Ra中,所述C1~C6烷基为C1~C3烷基;
    b)Ra中,所述C3~C6环烷基为C3~C4环烷基;
    c)Ra1、Ra2、Ra3或Ra4中,所述C1~C3烷基为甲基;
    d)Ra1、Ra2、Ra3或Ra4中,所述被1个、2个或3个卤素取代的C1~C3烷基中的C1~C3烷基为甲基;
    e)Ra1、Ra2、Ra3或Ra4中,所述卤素为氟;
    f)R21中,所述C1~C6烷基为C1~C3烷基;
    g)R23中,所述卤素为氟;
    h)R24中,所述C1~C6烷基为C1~C3烷基;
    i)R241或R241a中,所述C1~C6烷基为C1~C3烷基;
    j)R25中,所述C1~C6烷基为C1~C3烷基;
    k)R26中,所述C1~C6烷基为C1~C3烷基;
    l)R27或R27a中,所述C1~C6烷基为C1~C3烷基;
    m)R27或R27a中,所述C3~C6环烷基为C3~C4环烷基;
    n)R271中,所述C1~C6烷基为C1~C3烷基。
  8. 如权利要求7所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物,其特征在于,所述的式II所示的化合物满足下列条件中的一个或多个:
    a)Ra中,所述C1~C6烷基为甲基、乙基、正丙基或异丙基;
    b)Ra中,所述C3~C6环烷基为环丙基;
    c)R21中,所述C1~C6烷基为甲基、乙基、正丙基或异丙基;
    d)R24中,所述C1~C6烷基为甲基、乙基、正丙基或异丙基;
    e)R241或R241a中,所述C1~C6烷基为甲基、乙基、正丙基或异丙基;
    f)R25中,所述C1~C6烷基为甲基、乙基、正丙基或异丙基;
    g)R26中,所述C1~C6烷基为甲基、乙基、正丙基或异丙基;
    h)R27或R27a中,所述C1~C6烷基为甲基、乙基、正丙基或异丙基;
    i)R27或R27a中,所述C3~C6环烷基为环丙基;
    j)R271中,所述C1~C6烷基为甲基、乙基、正丙基或异丙基。
  9. 如权利要求1-3中任一项所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物,其特征在于,所述的式II所示的化合物满足下列条件中的一个或多个:
    a)Ra中,所述C1~C6烷基为甲基;
    b)环A中,所述3~10元杂碳环为单环或双环;所述双环为桥环;
    优选地,环A中,所述3~10元杂碳环为
    c)环A中,所述3~10元碳环为3~6元碳环,例如环丙烷基环、环丁烷基环、环戊烷基环或环己烷基环,例如
    d)R2中,所述3~10元环烷基和被1个、2个或3个R22取代的3~10元环烷基中的3~10元环烷基各自独立地为环丙基;
    e)R2中,所述3~10元杂环烷基为单环;
    f)R2中,所述被1个、2个或3个R21取代的3~10元杂环烷基中的3~10元杂环烷基为单环;
    优选地,R2中,所述3~10元杂环烷基和被1个、2个或3个R21取代的3~10元杂环烷基中的3~10元杂环烷基各自独立地为
    g)R2中,所述5~6元杂芳基和被1个、2个或3个R26取代的5~6元杂芳基中的5~6元杂芳基各自独立地为
    h)R21、R24、R25、R26、R27、R27a、R271和R 3中,所述C1~C6烷基各自独立地为甲基;
    i)R27或R27a中,所述卤素为F;
    j)R27或R27a中,所述被1个、2个或3个卤素取代的C1~C6烷基中C1~C6烷基可独立地为C1~C3烷基;又可独立地为乙基;
    k)RA中,所述卤素为氟、氯或溴,例如为氟;
    l)R3中,所述C1~C6烷基为异丙基。
  10. 如权利要求1-3中任一项所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物,其特征在于,所述的式II所示的化合物满足下列条件中的一个或多个:
    a)R1
    例如,R1为又如,R1
    b)Ra为C1~C6烷基,例如甲基;
    c)X、Y和Z独立地为-C(Ra1)(Ra2)-、=CRa3-、-NRa4-、-O-、-S-或=N-;
    例如,X、Y和Z独立地为-C(Ra1)(Ra2)-、=CRa3-、-NRa4-、-O-、-S-、=N-或-C(=O)-;
    又如,X、Y和Z独立地为-C(Ra1)(Ra2)-、=CRa3-、-NRa4-或-O-;
    d)Ra1、Ra2、Ra3和Ra4独立地为氢或C1~C3烷基;
    e)R2为氢、羟基、氰基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R26取代的5~6元杂芳基或被1个、2个或3个R21取代的3~10元杂环烷基;
    f)R21和R22独立地为羟基、氧代、氰基、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C3~C6环烷基、C1~C6烷基、被1个、2个或3个卤素取代的C1~C6烷基或R271-(C=O)-,R271独立地为C1~C6烷基;
    例如,R21和R22独立地为氧代、氰基、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C3~C6环烷基、C1~C6烷基或R271-(C=O)-,R271独立地为C1~C6烷基;
    g)所述的5~6元杂芳基中,杂原子的数量为2、3或4个,杂原子独立地选自N、O和S中的一种或多种;
    h)R3为C1~C6烷基;
    i)R4为H。
  11. 如权利要求1-3中任一项所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物,其特征在于,所述的式II所示的化合物满足下列条件中的一个或多个:
    a)R1中,中,Ra为C1~C6烷基或C3-6环烷基;X、Y和Z可独立地为-C(Ra1)(Ra2)-、=CRa3-、-C(=O)-、-NRa4-、=N-、-O-或-S-;Ra1和Ra2可独立地为氢或卤素;Ra3可为氢或卤素;Ra4可为氢、C1~C3烷基或三氟甲基;
    b)R1中,中,X、Y和Z独立地为-C(Ra1)(Ra2)-、=N-、-O-或-S-;Ra1和Ra2可独立地为氢;
    c)R1中,中,X、Y和Z独立地为=CRa3-或-NRa4-;Ra3和Ra4可独立地为氢、C1~C3烷基、三氟甲基或卤素;
    d)R1中,中,X、Y和Z独立地为-C(Ra1)(Ra2)-或=CRa3-;Ra1、Ra2和Ra3可独立地为氢、C1~C3烷基、三氟甲基或卤素;
    e)R1中,中,X、Y和Z独立地为=CRa3-或-NRa4-;Ra3和Ra4可独立地为氢、C1~C3烷基、三氟甲基或卤素;
    f)R1中,中,X、Y和Z独立地为-C(Ra1)(Ra2)-、=CRa3-、-C(=O)-、-NRa4-、=N-、-O-或-S-;Ra1和Ra2可独立地为氢或卤素;Ra3可为氢;Ra4可为氢、C1~C3烷基或三氟甲基;
    g)R1中,中,X、Y、Z和W独立地为-C(Ra1)(Ra2)-、=N-、-O-或-S-;Ra1和Ra2可独立地为氢;Ra可为C1~C3烷基;
    h)L1为单键;L2可为单键;R2可为氢、3~10元杂环烷基、被1个、2个或3个R21取代的3~10元杂环烷基、被1个、2个或3个R22取代的3~10元环烷基或被1个、2个或3个R23取代的C1~C6烷基;R21可为氧代,R22可为氰基,R23可为卤素;
    或者,L1为单键;L2可为C1~C6亚烷基或-(C=O)-;R2可为羟基、被1个、2个或3个R21取代的3~10元杂环烷基、被1个、2个或3个R22取代的3~10元环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基或被1个、2个或3个R26取代的5~6元杂芳基;R21可为C1~C6烷基或氧代;R22可为氰基;R24可为C1~C6烷基、(R241)(R241a)N-,R241和R241a可独立地为氢或C1~C6烷基;R25可为C1~C6烷基;R26可为C1~C6烷基;
    或者,L1为C1~C6亚烷基或-(C=O)-;L2可为C1~C6亚烷基或-(C=O)-,其中,L1和L2不同时为C1~C6亚烷基,且L1和L2不同时为-(C=O)-;R2可为氰基、三氟甲基、3~10元杂环烷基、被1个、2个或3个R21取代的3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、被1个、2个或3个R26取代的5~6元杂芳基或(R27)(R27a)N-;R21可为羟基、氧代或卤素;R24可为C1~C6烷基;R25可为C1~C6烷基;R26可为C1~C6烷基;R27和R27a可独立地为氢、C1~C6烷基、C3~C6环烷基或R271-(C=O)-,R271可为C1~C6烷基;
    或者,L1为单键;L2可为单键;R2可为氢或3~10元杂环烷基;
    或者,L1为单键;L2可为C1~C6亚烷基或-(C=O)-;R2可为羟基、5~6元杂芳基、被1个、2个或3个R21取代的3~10元杂环烷基或R24-S(=O)2-;R21可为C1~C6烷基;R24可为C1~C6烷基或(R241)(R241a)N-,R241和R241a可独立地为氢或C1~C6烷基;
    或者,L1可为C1~C6亚烷基或-(C=O)-;L2可为C1~C6亚烷基或-(C=O)-,其中,L1和L2不同时为C1~C6亚烷基,且L1和L2不同时为-(C=O)-;R2可为3~10元杂环烷基、被1个、2个或3个R21取代的3~10元杂环烷基或(R27)(R27a)N-;R21可为氧代或卤素;R27和R27a可独立地为氢或C1~C6烷基。
  12. 如权利要求1-3中任一项所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物,其特征在于,所述的式II所示的化合物满足下列条件中的一个或多个:
    a)R1 优选地,R1为
    b)环A为其中s端与L1相连接;优选地,环A为其中s端与L1相连接;
    c)L1为单键、亚甲基、亚乙基或-(C=O)-或所述q端与L2相连接;
    d)L2为单键、亚甲基、亚乙基或-(C=O)-或所述q端与R2相连接;
    e)R2为氢、羟基、氰基、三氟甲基、 优选地,R2为氢、羟基、氰基、 进一步优选地,为H、
    f)R3为异丙基或环丙基;
    g)R4为氢或甲基。
  13. 如权利要求1-3中任一项所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物,其特征在于所述式II所示的化合物为如下任一方案:
    方案一:所述式II所示的化合物为式I所示化合物,其中
    R1
    Ra为C1~C6烷基;
    X、Y和Z独立地为-C(Ra1)(Ra2)-、=CRa3-、-NRa4-或-O-;
    Ra1、Ra2、Ra3和Ra4独立地为氢或C1~C3烷基;
    U独立地为N或CH;
    为单键或者双键,相邻的两个不同时为双键;
    环A为3~10元杂碳环;
    L1为单键、C1~C6亚烷基或-(C=O)-;
    L2为单键、C1~C6亚烷基或-(C=O)-;
    R2为氢、羟基、氰基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R26取代的5~6元杂芳基或被1个、2个或3个R21取代的3~10元杂环烷基;
    R21和R22独立地为氧代、氰基、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C3~C6环烷基、C1~C6烷基或R271-(C=O)-,R271独立地为C1~C6烷基;
    所述的3~10元杂碳环中,杂原子的数量为1个,杂原子为N;
    所述的3~10元杂环烷基中,杂原子的数量独立地为1个或2个,杂原子独立地选自N、O和S中的一种或多种;
    所述的5~6元杂芳基中,杂原子的数量为2个,杂原子独立地选自N、O和S中的一种或多种;
    R3为C1~C6烷基;
    R4为氢;
    方案二:
    R1
    Ra为C1~C6烷基;
    X、Y和Z独立地为-C(Ra1)(Ra2)-、=CRa3-、-NRa4-、-O-、-S-或=N-;
    Ra1、Ra2、Ra3和Ra4独立地为氢或C1~C3烷基;
    U独立地为N或CH;
    为单键或者双键,相邻的两个不同时为双键;
    环A为3~10元杂碳环或3~10元碳环;
    L1为单键、C1~C6亚烷基或-(C=O)-;
    L2为单键、C1~C6亚烷基或-(C=O)-;
    R2为氢、羟基、氰基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R26取代的5~6元杂芳基或被1 个、2个或3个R21取代的3~10元杂环烷基;
    R21和R22独立地为羟基、氧代、氰基、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C3~C6环烷基、C1~C6烷基、被1个、2个或3个卤素取代的C1~C6烷基或R271-(C=O)-,R271独立地为C1~C6烷基;
    所述的3~10元杂碳环中,杂原子的数量为1个或2个,杂原子为N;
    所述的3~10元杂环烷基中,杂原子的数量独立地为1个或2个,杂原子独立地选自N、O和S中的一种或多种;
    所述的5~6元杂芳基中,杂原子的数量为2个至4个,杂原子独立地选自N、O和S中的一种或多种;
    V为C-R4或N;
    RA为氢或卤素;
    R3为C1~C6烷基;
    R4为氢;
    方案三:R1
    Ra为甲基;
    X、Y和Z独立地为-C(Ra1)(Ra2)-、=CRa3-、-NRa4-、-O-、-S-、=N-、或-C(=O)-;
    Ra1、Ra2、Ra3和Ra4独立地为氢或C1~C3烷基;
    U独立地为N或CH;
    为单键或者双键,相邻的两个不同时为双键;
    环A为3~10元杂碳环;
    L1为单键、C1~C6亚烷基或-(C=O)-;
    L2为单键、C1~C6亚烷基或-(C=O)-;
    R2为氢、羟基、氰基、3~10元杂环烷基、R24-S(=O)2-、R25-S(=O)2-(NH)-、5~6元杂芳基、(R27)(R27a)N-、被1个、2个或3个R22取代的3~10元环烷基、被1个、2个或3个R26取代的5~6元杂芳基或被1个、2个或3个R21取代的3~10元杂环烷基;
    R21和R22独立地为羟基、氧代、氰基、卤素或C1~C6烷基;R24为C1~C6烷基或(R241)(R241a)N-,R241和R241a独立地为氢;R25为C1~C6烷基;R26独立地为C1~C6烷基;R27和R27a独立地为氢、C3~C6环烷基、C1~C6烷基、被1个、2个或3个卤素取代的C1~C6烷基或R271-(C=O)-,R271独立地为C1~C6烷基;
    所述的3~10元杂碳环中,杂原子的数量为1个或2个,杂原子为N;
    所述的3~10元杂环烷基中,杂原子的数量独立地为1个或2个,杂原子独立地选自N、O和S中的一种或多种;
    所述的5~6元杂芳基中,杂原子的数量为2个,杂原子独立地选自N、O和S中的一种或多种;
    V为C-R4或N;
    RA为氢或卤素;
    R3为C1~C6烷基;
    R4为氢。
  14. 如权利要求1-3中任一项所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物,其特征在于,所述的式II所示的化合物为如下任一结构:










  15. 一种药物组合物,其包括物质X和药用辅料;
    所述的物质X为如权利要求1~14中任一项所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物。
  16. 一种物质X在制备TLR7和/或TLR8抑制剂中的应用;
    所述的物质X为如权利要求1~14中任一项所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物。
  17. 一种物质X在制备药物中的应用;
    所述的物质X为如权利要求1~14中任一项所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物;
    所述的药物为用于治疗与TLR7和/或TLR8相关疾病的药物。
  18. 一种物质X在制备用于治疗自身免疫性疾病的药物中的应用;
    所述的物质X为如权利要求1~14中任一项所述的式II所示的化合物、其药学上可接受的盐、其溶剂合物或其药学上可接受的盐的溶剂合物。
PCT/CN2023/110566 2022-08-01 2023-08-01 含氮杂环类化合物、其制备方法及其应用 WO2024027712A1 (zh)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202210917037 2022-08-01
CN202210917037.5 2022-08-01
CN202211680354 2022-12-26
CN202211680354.6 2022-12-26

Publications (1)

Publication Number Publication Date
WO2024027712A1 true WO2024027712A1 (zh) 2024-02-08

Family

ID=89667864

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/110566 WO2024027712A1 (zh) 2022-08-01 2023-08-01 含氮杂环类化合物、其制备方法及其应用

Country Status (2)

Country Link
CN (1) CN117486861A (zh)
WO (1) WO2024027712A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060235037A1 (en) * 2005-04-15 2006-10-19 Purandare Ashok V Heterocyclic inhibitors of protein arginine methyl transferases
CN101163675A (zh) * 2005-04-19 2008-04-16 协和发酵工业株式会社 含氮杂环化合物
US20200062758A1 (en) * 2016-11-11 2020-02-27 Hepo Pharmaceutical Co., Ltd Nitrogen-Containing Heterocyclic Compound, Preparation Method, Intermediate, Pharmaceutical Composition and use
CN111448190A (zh) * 2017-11-14 2020-07-24 百时美施贵宝公司 取代的吲哚化合物
CN114591339A (zh) * 2022-05-10 2022-06-07 上海维申医药有限公司 一类Toll样受体抑制剂及其制备和应用

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060235037A1 (en) * 2005-04-15 2006-10-19 Purandare Ashok V Heterocyclic inhibitors of protein arginine methyl transferases
CN101163675A (zh) * 2005-04-19 2008-04-16 协和发酵工业株式会社 含氮杂环化合物
US20200062758A1 (en) * 2016-11-11 2020-02-27 Hepo Pharmaceutical Co., Ltd Nitrogen-Containing Heterocyclic Compound, Preparation Method, Intermediate, Pharmaceutical Composition and use
CN111448190A (zh) * 2017-11-14 2020-07-24 百时美施贵宝公司 取代的吲哚化合物
CN114591339A (zh) * 2022-05-10 2022-06-07 上海维申医药有限公司 一类Toll样受体抑制剂及其制备和应用

Also Published As

Publication number Publication date
CN117486861A (zh) 2024-02-02

Similar Documents

Publication Publication Date Title
TWI712591B (zh) 作為hdac抑制劑之3-烷基-4-醯胺基-雙環[4,5,0]羥肟酸
WO2021219072A1 (zh) 作为kras抑制剂的杂环化合物的制备及其应用方法
WO2020108590A1 (zh) 嘧啶并五元氮杂环类衍生物、其制备方法及其在医药上的应用
DK2855476T3 (en) TETRAHYDROPYRAZOLOPYRIMIDINE COMPOUNDS
CN113166139A (zh) 作为HPK1抑制剂的吡咯并[2,3-b]吡啶及其用途
KR20220027879A (ko) 질소 함유 헤테로사이클릭 유도체의 조절제, 이의 제조방법 및 용도
TW202116773A (zh) 作為HPK1抑制劑之吡咯并[2,3-b]吡及其用途
WO2020238791A1 (zh) 氢化吡啶并嘧啶类衍生物、其制备方法及其在医药上的应用
JP2022532758A (ja) 二環系誘導体を含む阻害剤、その製造方法及び使用
CN103038233A (zh) 吡啶酮和氮杂吡啶酮化合物及使用方法
WO2017071516A1 (zh) 一种蛋白激酶抑制剂及其制备方法和医药用途
WO2021115457A9 (zh) 吡唑并[1,5-a]吡啶类化合物及其制备方法和应用
CN114341127A (zh) 作为hpk1抑制剂的氨基吡嗪化合物及其用途
WO2021190417A1 (zh) 新型氨基嘧啶类egfr抑制剂
CN113423708A (zh) 作为tlr7激动剂的咪唑并[2,1-f][1,2,4]三嗪-4-胺衍生物
WO2022247816A1 (zh) 含氮杂环类化合物、其制备方法及其在医药上的应用
KR20230035036A (ko) 이중 키나제-브로모도메인 억제제
WO2022166920A1 (zh) 一种吡咯并哒嗪类化合物及其制备方法和用途
KR20220140710A (ko) 트리아졸로피리다진 유도체, 이의 제조 방법, 약물 조성물 및 용도
WO2023016518A1 (zh) 一种杂环衍生物及其组合物和药学上的应用
CN112839941B (zh) 作为TGF-βR1抑制剂的化合物及其应用
WO2023041049A1 (zh) 作为sos1抑制剂的杂环化合物及其用途
WO2023165581A1 (zh) 一种吡啶类衍生物及其用途
WO2024027712A1 (zh) 含氮杂环类化合物、其制备方法及其应用
TWI779487B (zh) 二氫萘啶酮類化合物,其製法與醫藥上的用途

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: 23849418

Country of ref document: EP

Kind code of ref document: A1