WO2024094171A1 - 取代的氨基嘧啶类化合物、其制备方法及其在医药上的应用 - Google Patents

取代的氨基嘧啶类化合物、其制备方法及其在医药上的应用 Download PDF

Info

Publication number
WO2024094171A1
WO2024094171A1 PCT/CN2023/129593 CN2023129593W WO2024094171A1 WO 2024094171 A1 WO2024094171 A1 WO 2024094171A1 CN 2023129593 W CN2023129593 W CN 2023129593W WO 2024094171 A1 WO2024094171 A1 WO 2024094171A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
cycloalkyl
alkylene
alkoxy
Prior art date
Application number
PCT/CN2023/129593
Other languages
English (en)
French (fr)
Inventor
贾敏强
王伟民
吴婷
韩吉慧
贺峰
Original Assignee
江苏恒瑞医药股份有限公司
上海恒瑞医药有限公司
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 江苏恒瑞医药股份有限公司, 上海恒瑞医药有限公司 filed Critical 江苏恒瑞医药股份有限公司
Publication of WO2024094171A1 publication Critical patent/WO2024094171A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic 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 three hetero rings
    • C07D498/18Bridged systems

Definitions

  • the present disclosure belongs to the field of medicine, and relates to a substituted aminopyrimidine compound represented by the general formula (X), a preparation method thereof, a pharmaceutical composition containing the compound, and the use thereof as a therapeutic agent, in particular, the use thereof as an EGFR inhibitor and/or degrader and the use thereof in the preparation of a drug for treating and/or preventing EGFR-mediated or dependent diseases or conditions.
  • a substituted aminopyrimidine compound represented by the general formula (X) a preparation method thereof, a pharmaceutical composition containing the compound, and the use thereof as a therapeutic agent, in particular, the use thereof as an EGFR inhibitor and/or degrader and the use thereof in the preparation of a drug for treating and/or preventing EGFR-mediated or dependent diseases or conditions.
  • PROTAC Protein-Proteolysis Targeting Chimera
  • PROTAC is a hybrid bifunctional small molecule compound. Its structure contains two different ligands: one is the ubiquitin ligase E3 ligand, and the other is the ligand that binds to the target protein. The two ligands are connected by a connecting arm.
  • PROTAC forms a target protein-PROTAC-E3 ternary complex by bringing the target protein and the ubiquitin ligase E3 in the cell closer.
  • PROTAC has shown unique advantages: 1. PROTAC does not need to bind to the target protein for a long time and with high intensity, and the process of degrading the target protein is similar to a catalytic reaction, which can be cyclically bound and degraded. Thereby reducing the systemic exposure of the drug and reducing the occurrence of toxic side effects. 2. After the target protein is degraded, it needs to be resynthesized to restore its function.
  • PROTAC also has therapeutic potential for targets that are currently considered undruggable, such as transcription factors, scaffold proteins, and regulatory proteins.
  • Lung cancer is one of the most common malignant tumors. According to statistics, the cancer with the highest incidence rate in the world in 2018 is lung cancer, with 2.094 million new lung cancer patients. In 2018, the number of lung cancer cases in China was 868,000, with an incidence rate of 0.062%, accounting for 41.4% of the global number of cases.
  • Non-small cell lung cancer is the most common type of lung cancer, accounting for about 85% of the total number of lung cancers.
  • Epidermal growth factor receptor (EGFR) belongs to the family of cell surface receptors with tyrosine kinase activity. EGFR mutation is the most common driver gene for non-small cell lung cancer. About 40% of Chinese non-small cell lung cancer patients have EGFR mutations, while 11-16% of patients in Western countries have EGFR mutations. About 90% of EGFR mutations are deletions in exon 19 (Del19 mutations) and L858R point mutations in exon 21.
  • EGFR small molecule inhibitors have been approved for marketing and have been successfully used in the treatment of non-small cell lung cancer with EGFR mutations, becoming one of the main treatments for patients with advanced non-small cell lung cancer.
  • the first-generation EGFR tyrosine kinase inhibitors represented by gefitinib and erlotinib, are reversibly binding targeted drugs that inhibit activation by competing with ATP for binding to the EGFR kinase domain.
  • gefitinib and erlotinib are reversibly binding targeted drugs that inhibit activation by competing with ATP for binding to the EGFR kinase domain.
  • most patients will develop drug resistance after 10-12 months of treatment, and about 50% of patients develop drug resistance due to the secondary mutation of T790M.
  • the second-generation EGFR tyrosine kinase inhibitors represented by afatinib, are irreversible Targeted drugs, but they cannot solve the problem of T790M mutation resistance, and the lack of selectivity for wild-type EGFR makes the compound more toxic.
  • the emergence of the third-generation EGFR tyrosine kinase inhibitor osimertinib overcomes the resistance caused by EGFR T790M gene mutation, and has weak inhibition of wild-type EGFR and good selectivity. It has achieved great success in clinical practice, but new resistance will occur after 9-14 months of use. Studies have revealed that 6-26% of patients have C797X or other EGFR-dependent gene mutations (JAMA Oncol. 2018, 4, 1527-1534; Br J Cancer. 2019 Oct; 121 (9): 725-737.).
  • EGFR inhibitors for EGFR C797S gene mutations in the market, so it is necessary to develop new treatments for non-small cell lung cancer.
  • PROTAC technology can degrade EGFR, which can more effectively inhibit the EGFR signaling pathway and may become a potential treatment for non-small cell lung cancer.
  • PROTAC compounds targeting EGFR protein degradation include WO2017185036A1, WO2018119441A1, WO2019121562A1, WO2019149922A1, WO2021121261A1, WO2021127561A1, WO2021039622A1, WO2022055181A1, WO2022012623A1, and WO2022068849A1.
  • the purpose of the present disclosure is to provide a compound represented by the general formula (X), or a pharmaceutically acceptable salt thereof:
  • R 1 is selected from -P(O)R 10 R 11 , -S(O) r R 12 , -S(O) r N(R 13 R 14 ), -N(R 15 )S(O) r R 16 and R 9 ;
  • each R 9 is the same or different and is independently selected from a deuterium atom, a halogen, an alkyl, a haloalkyl, a deuterated alkyl, an alkoxy, a haloalkoxy, a deuterated alkoxy, a hydroxyl, an alkenyl, an alkynyl, a cyano, a nitro, a NR 20 R 21 , a C(O)NR 20 R 21 , a NR 20 C(O)R 24 , an -alkylene-NR 20 R 21 , an -alkylene-C(O)NR 20 R 21 , a C(O)R 24 , a C(O)OR 24 , a cycloalkyl, a cycloalkylalkyl, a heterocyclyl, a heterocyclylalkyl, a cycloalkyloxy, a heterocyclyloxy, an aryl and a heteroaryl;
  • R 9 and the carbon atom to which they are attached together form a cycloalkyl, heterocyclyl, aryl or heteroaryl group optionally substituted by one or more R 01 ;
  • L 1 is connected to a phenyl group, or L 1 is connected to a ring formed by two R 9s ;
  • R4 and R5 are the same or different and are each independently selected from a hydrogen atom, a deuterium atom, a halogen, an alkyl group, a haloalkyl group, a deuterated alkyl group, an alkoxy group, a haloalkoxy group, a deuterated alkoxy group, a hydroxyl group, an alkenyl group , an alkynyl group, a cyano group, a nitro group, a NR22R23 group, a C(O) NR22R23 group, a NR22C (O ) R24 group, a C(O) R24 group, a C(O) OR24 group , a cycloalkyl group, a heterocyclyl group, a heterocyclylalkyl group, a heterocyclyloxy group, an aryl group and a heteroaryl group; and the alkyl group, alkoxy group, alkenyl group, alkyny
  • Ring A is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • Each RA is the same or different and is independently selected from a deuterium atom, an oxo group, a halogen, an alkyl group, a haloalkyl group, an alkoxy group, a haloalkoxy group, a hydroxyl group, an alkenyl group, an alkynyl group, a cyano group, a nitro group, a NR25R26 group, a C(O) NR25R26 group , a cycloalkyl group, a heterocyclyl group, a heterocyclylalkyl group, a heterocyclyloxy group, an aryl group and a heteroaryl group; the alkyl group, alkoxy group, alkenyl group, alkynyl group, cycloalkyl group, a heterocyclyl group, a heterocyclylalkyl group, a heterocyclyloxy group, an aryl group and a heteroaryl group; the alkyl group,
  • L1 and L3 are the same or different and are each independently selected from a bond, O, S, NR L , S(O) r , C(O), C(O) NR L , NR L C(O), S(O) r NR L , NR L C(O) NR L , (CR L1 R L2 ) m , alkenyl and alkynyl; the alkenyl is optionally substituted with one or more R 01 ;
  • L2 is -(CR L3 R L4 ) n1 -R L5 -(CR L6 R L7 ) n2 -;
  • RL5 is selected from the group consisting of a bond, O, S, NR L ′, S(O), S(O) 2 , C(O), C(O)NR L ′, NR L ′C(O), and NR L ′C(O)NR L ′;
  • Each of RL1 , RL2 , RL3 , RL4 , RL6 and RL7 is the same or different and is independently selected from a hydrogen atom, a deuterium atom, a halogen, an alkyl group, a haloalkyl group, an alkoxy group, a haloalkoxy group, a hydroxyl group, an alkenyl group, an alkynyl group, a cyano group, a cycloalkyl group, a heterocyclic group, an aryl group and a heteroaryl group; the alkyl group, alkoxy group, alkenyl group, alkynyl group, cycloalkyl group, heterocyclic group, aryl group and heteroaryl group are independently substituted by one or more R01 ;
  • RL1 , RL2 and the carbon atoms to which they are connected together form a cycloalkyl or heterocyclic group
  • RL3 , RL4 and the carbon atoms to which they are connected together form a cycloalkyl or heterocyclic group
  • RL6 , RL7 and the carbon atoms to which they are connected together form a cycloalkyl or heterocyclic group
  • the cycloalkyl or heterocyclic group is independently optionally substituted by one or more R01 ;
  • R10 , R11 , R12 , R13 , R14 , R15 and R16 are the same or different and are each independently selected from a hydrogen atom, a deuterium atom, an alkyl group, a halogenated alkyl group, a deuterated alkyl group, an alkoxy group, a halogenated alkoxy group, a deuterated alkoxy group, an alkenyl group, an alkynyl group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the alkenyl group, the alkynyl group, the cycloalkyl group and the heterocyclic group are each independently optionally substituted by one or more R01 ;
  • Each of R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , RL and RL ' is the same or different and is independently selected from a hydrogen atom, a deuterium atom, an alkyl group, a haloalkyl group, an alkoxy group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the cycloalkyl group and the heterocyclic group are independently optionally substituted by one or more R 01 ;
  • R13 , R14 and the nitrogen atom to which they are connected together form a heterocyclic group; or NR15 and the carbon atom to which it is connected, R9 and the carbon atom to which it is connected together form a nitrogen-containing heterocyclic group or a nitrogen-containing heteroaryl group; or R20 , R21 and the nitrogen atom to which they are connected together form a heterocyclic group; or R22 , R23 and the nitrogen atom to which they are connected together form a heterocyclic group; or R25 , R26 and the nitrogen atom to which they are connected together form a heterocyclic group; the heterocyclic group, the nitrogen-containing heterocyclic group or the nitrogen-containing heteroaryl group are each independently optionally substituted by one or more R01 ;
  • Ra and Rb are the same or different and are each independently selected from a hydrogen atom, an alkyl group, a haloalkyl group, a deuterated alkyl group, an alkoxy group, a haloalkoxy group, a deuterated alkoxy group, a cycloalkyl group, and a heterocyclic group;
  • each R 01 is the same or different and is independently selected from a deuterium atom, an oxo group, a halogen, an alkyl group, an alkoxy group, a haloalkyl group, a haloalkoxy group, a hydroxyl group, a hydroxyalkyl group, a hydroxyalkoxy group, a cyano group, an alkenyl group, an alkynyl group, an amino group, an -NHalkyl group, an -N(alkyl) 2 group, an -alkylene-amino group, an -alkylene-NHalkyl group, an -alkylene-N(alkyl) 2 group , an amide group, a nitro group, a cycloalkyl group, a heterocyclyl group, a cycloalkylalkyl group, a heterocyclylalkyl group, a cycloalkyloxy group, a heterocyclyloxy group, an
  • Z1 is N or CR Z1 ;
  • Z2 is N or CR Z2 ;
  • Z3 is N or CR Z3 ;
  • R Z1 , R Z2 and R Z3 are the same or different and are each independently selected from a hydrogen atom, a deuterium atom, a halogen, an alkyl group, a haloalkyl group, a deuterated alkyl group, an alkoxy group, a haloalkoxy group, a deuterated alkoxy group, a hydroxyl group, an alkenyl group, an alkynyl group, a cyano group, a nitro group, a C(O)R 30 , a C(O)OR 30 , a NR 31 R 32 , a C(O)NR 31 R 32 , a NR 31 C(O)R 30 , a cycloalkyl group, a cycloalkyloxy group, a heterocyclyl group, a heterocyclyloxy group, an aryl group and a heteroaryl group; the alkyl group, alkoxy group, alkenyl group, alky
  • Each R 30 is the same or different and is independently selected from a hydrogen atom, an alkyl group, an alkenyl group, an alkynyl group, a cycloalkyl group, a heterocyclic group, an aryl group and a heteroaryl group; the alkyl group, the alkenyl group, the alkynyl group, the cycloalkyl group, the heterocyclic group, the aryl group and the heteroaryl group are independently optionally substituted by one or more R 02 ;
  • Each R 31 and R 32 are the same or different and are independently selected from hydrogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl and heterocyclic groups; the alkyl, alkoxy, cycloalkyl and heterocyclic groups are independently substituted by one or more R 02 ;
  • each R 02 is the same or different and is independently selected from a deuterium atom, an oxo group, a halogen, an alkyl group, an alkoxy group, a haloalkyl group, a haloalkoxy group, a hydroxyl group, a hydroxyalkyl group, a hydroxyalkoxy group, a cyano group, an alkenyl group, an alkynyl group, an amino group, -NHalkyl group, -N(alkyl) 2 , an -alkylene-amino group, an -alkylene-NHalkyl group, an -alkylene-N(alkyl) 2 group, an amide group, a nitro group, a cycloalkyl group, a heterocyclyl group, a cycloalkylalkyl group, a heterocyclylalkyl group, a cycloalkyloxy group, a heterocyclyloxy group, an aryl
  • J1 , J2 , J3 , J4 , J5 and J6 are the same or different and are each independently selected from a bond, O, S, -O-alkylene-, -alkylene-O-, C(O), -C(O)-alkylene-, -alkylene-C(O)-, C(O)N( RJ ), N( RJ )C(O), N( RJ )C(O)N( RJ ), S(O) v , S(O) vN ( RJ ), N( RJ )S(O) v , N( RJ ), -N( RJ )-alkylene-, -alkylene-N( RJ )-, alkylene, alkenyl, alkynyl, cycloalkyl, heterocyclyl, -alkylene-heterocyclyl-, -heterocyclyl-alkylene-, aryl and heteroaryl; the
  • Each R J is the same or different and is independently selected from a hydrogen atom, an alkyl group, an alkoxy group, a haloalkyl group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the cycloalkyl group and the heterocyclic group are independently substituted by one or more substituents selected from R 03 ;
  • each R 03 is the same or different and is independently selected from a deuterium atom, an oxo group, a halogen, an alkyl group, an alkoxy group, a haloalkyl group, a haloalkoxy group, a hydroxyl group, a hydroxyalkyl group, a hydroxyalkoxy group, a cyano group, an alkenyl group, an alkynyl group, an amino group, -NHalkyl group, -N(alkyl) 2 , an -alkylene-amino group, an -alkylene-NHalkyl group, an -alkylene-N(alkyl) 2 , an amide group, a nitro group, a cycloalkyl group, a heterocyclyl group, a cycloalkylalkyl group, a heterocyclylalkyl group, a cycloalkyloxy group, a heterocyclyloxy group, an aryl
  • E is selected from:
  • Ring D is aryl or heteroaryl
  • W is selected from a bond, O, S, S(O) r , NR 6 , C(O), C(O)NR 6 , NR 6 C(O) and (CR Y2 R Y2 ) s4 ;
  • Q 1 , Q 2 , Q 3 , Q 4 and Q 5 is a carbon atom and is connected to J 6 , and the others are each independently selected from N, CH and CR q ;
  • Each R q is the same or different and is independently selected from a deuterium atom, a halogen, an alkyl, a haloalkyl, a deuterated alkyl, an alkoxy, a haloalkoxy, a deuterated alkoxy, a hydroxyl, an alkenyl, an alkynyl, a cyano, a NR c R d , an alkylene NR c R d , a C(O)alkyl, a C(O)NR c R d , a cycloalkyl, a heterocyclyl, an aryl and a heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, alkylene, cycloalkyl, a heterocyclyl, an aryl and a heteroaryl are each independently optionally substituted with one or more R 04 ;
  • Each R c and R d are the same or different and are independently selected from hydrogen, halogen, alkyl, haloalkyl, deuterated alkyl, alkoxy, haloalkoxy, deuterated alkoxy, cycloalkyl and heterocyclic group; or R c , R d and the nitrogen atom to which they are attached together form a heterocyclic group, and the heterocyclic group is independently optionally substituted by one or more R 04 ;
  • Y2 is C( RY2 ) 2 or C(O);
  • Each R Y2 is the same or different and is independently selected from a hydrogen atom, a deuterium atom, an alkyl group, a haloalkyl group, an alkoxy group, a haloalkoxy group, an alkenyl group, an alkynyl group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the alkenyl group, the alkynyl group, the cycloalkyl group and the heterocyclic group are independently optionally substituted by one or more R O4 ;
  • R Y2 together with the carbon atom to which they are connected form a cycloalkyl or heterocyclic group; the cycloalkyl and heterocyclic groups are each independently optionally substituted by one or more R 04 ;
  • R Y1 and R Y3 are the same or different and are each independently selected from a hydrogen atom, a deuterium atom, an alkyl group, a haloalkyl group, an alkoxy group, a haloalkoxy group, an alkenyl group, an alkynyl group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the alkenyl group, the alkynyl group, the cycloalkyl group and the heterocyclic group are each independently optionally substituted by one or more R O4 ;
  • R 6 , R 7 and R 8 are the same or different and are each independently selected from a hydrogen atom, an alkyl group, a halogenated alkyl group, a deuterated alkyl group, an alkoxy group, a halogenated alkoxy group, a deuterated alkoxy group, a cycloalkyl group and a heterocyclic group;
  • each R 04 is the same or different and is independently selected from a deuterium atom, an oxo group, a halogen, an alkyl group, an alkoxy group, a haloalkyl group, a haloalkoxy group, a hydroxyl group, a hydroxyalkyl group, a hydroxyalkoxy group, a cyano group, an alkenyl group, an alkynyl group, an amino group, -NHalkyl group, -N(alkyl) 2 , an -alkylene-amino group, an -alkylene-NHalkyl group, an -alkylene-N(alkyl) 2 , an amide group, a nitro group, a cycloalkyl group, a heterocyclyl group, a cycloalkylalkyl group, a heterocyclylalkyl group, a cycloalkyloxy group, a heterocyclyloxy group, an aryl
  • n 0, 1, 2, 3, 4, 5 or 6;
  • n1, n2 and m are each independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
  • the purpose of the present disclosure is to provide a compound represented by the general formula (X), or a pharmaceutically acceptable salt thereof:
  • E is a ligand that binds to cerebellin E3 ubiquitin ligase
  • R1 , R9 , y , Ra , Rb , R4, R5 , Z1 , Z2 , Z3 , L1 , L2 , Ring A, L3 , RA , n, J1 to J6 are as defined above.
  • the purpose of the present disclosure is to provide a compound represented by general formula (I), or a pharmaceutically acceptable salt thereof:
  • R1 is selected from -P(O) R10R11 , -S(O ) rR12 , -S(O) rN ( R13R14 ) and -N( R15 )S(O) rR16 ;
  • R2 and R3 are the same or different and are each independently selected from a hydrogen atom, a deuterium atom, a halogen, an alkyl group, a haloalkyl group, a deuterated alkyl group, an alkoxy group, a haloalkoxy group, a deuterated alkoxy group, a hydroxyl group, an alkenyl group, an alkynyl group , a cyano group, a nitro group, a NR20R21 group, a C(O)NR20R21 group, a NR20C(O)R24 group , a -alkylene group-NR20R21 group, a -alkylene group-C(O)NR20R21 group , a C ( O) R24 group, a C(O) OR24 group, a cycloalkyl group, a cycloalkylalkyl group, a heterocyclyl group, a heterocyclylalkyl
  • R4 and R5 are the same or different and are each independently selected from a hydrogen atom, a deuterium atom, a halogen, an alkyl group, a haloalkyl group, a deuterated alkyl group, an alkoxy group, a haloalkoxy group, a deuterated alkoxy group, a hydroxyl group, an alkenyl group , an alkynyl group, a cyano group, a nitro group, NR22R23 , C(O )NR22R23 , NR22C (O) R24 , C(O) R24 , C(O) OR24 , a cycloalkyl group, a heterocyclic group, a heterocyclic alkyl group, a heterocyclic oxy group, an aryl group and a heteroaryl group; the alkyl group, alkoxy group, alkenyl group, alkynyl group, R 01 is optionally substituted with one or more R
  • Ring A is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • Each RA is the same or different and is independently selected from a deuterium atom, an oxo group, a halogen, an alkyl group, a haloalkyl group, an alkoxy group, a haloalkoxy group, a hydroxyl group, an alkenyl group, an alkynyl group, a cyano group, a nitro group, a NR25R26 group, a C(O) NR25R26 group , a cycloalkyl group, a heterocyclyl group, a heterocyclylalkyl group, a heterocyclyloxy group, an aryl group and a heteroaryl group; the alkyl group, alkoxy group, alkenyl group, alkynyl group, cycloalkyl group, a heterocyclyl group, a heterocyclylalkyl group, a heterocyclyloxy group, an aryl group and a heteroaryl group; the alkyl group,
  • L1 and L3 are the same or different and are each independently selected from a bond, O, S, NR L , S(O) r , C(O), C(O) NR L , NR L C(O), S(O) r NR L , NR L C(O) NR L , (CR L1 R L2 ) m , alkenyl and alkynyl; the alkenyl is optionally substituted with one or more R 01 ;
  • L2 is -(CR L3 R L4 ) n1 -R L5 -(CR L6 R L7 ) n2 -;
  • RL5 is selected from the group consisting of a bond, O, S, NR L ′, S(O), S(O) 2 , C(O), C(O)NR L ′, NR L ′C(O), and NR L ′C(O)NR L ′;
  • Each of RL1 , RL2 , RL3 , RL4 , RL6 and RL7 is the same or different and is independently selected from a hydrogen atom, a deuterium atom, a halogen, an alkyl group, a haloalkyl group, an alkoxy group, a haloalkoxy group, a hydroxyl group, an alkenyl group, an alkynyl group, a cyano group, a cycloalkyl group, a heterocyclic group, an aryl group and a heteroaryl group; the alkyl group, alkoxy group, alkenyl group, alkynyl group, cycloalkyl group, heterocyclic group, aryl group and heteroaryl group are independently substituted by one or more R01 ;
  • RL1 , RL2 and the carbon atoms to which they are connected together form a cycloalkyl or heterocyclic group
  • RL3 , RL4 and the carbon atoms to which they are connected together form a cycloalkyl or heterocyclic group
  • RL6 , RL7 and the carbon atoms to which they are connected together form a cycloalkyl or heterocyclic group
  • the cycloalkyl or heterocyclic group is independently optionally substituted by one or more R01 ;
  • R10 , R11 , R12 , R13 , R14 , R15 and R16 are the same or different and are each independently selected from a hydrogen atom, a deuterium atom, an alkyl group, a halogenated alkyl group, a deuterated alkyl group, an alkoxy group, a halogenated alkoxy group, a deuterated alkoxy group, an alkenyl group, an alkynyl group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the alkenyl group, the alkynyl group, the cycloalkyl group and the heterocyclic group are each independently optionally substituted by one or more R01 ;
  • Each of R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , RL and RL ' is the same or different and is independently selected from a hydrogen atom, a deuterium atom, an alkyl group, a haloalkyl group, an alkoxy group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the cycloalkyl group and the heterocyclic group are independently optionally substituted by one or more R 01 ;
  • Ra and Rb are the same or different and are each independently selected from a hydrogen atom, an alkyl group, a haloalkyl group, a deuterated alkyl group, an alkoxy group, a haloalkoxy group, a deuterated alkoxy group, a cycloalkyl group, and a heterocyclic group;
  • each R 01 is the same or different and is independently selected from a deuterium atom, an oxo group, a halogen, an alkyl group, an alkoxy group, a haloalkyl group, a haloalkoxy group, a hydroxyl group, a hydroxyalkyl group, a hydroxyalkoxy group, a cyano group, an alkenyl group, an alkynyl group, an amino group, an -NHalkyl group, an -N(alkyl) 2 group, an -alkylene-amino group, an -alkylene-NHalkyl group, an -alkylene-N(alkyl) 2 group , an amide group, a nitro group, a cycloalkyl group, a heterocyclyl group, a cycloalkylalkyl group, a heterocyclylalkyl group, a cycloalkyloxy group, a heterocyclyloxy group, an
  • Z1 is N or CR Z1 ;
  • Z2 is N or CR Z2 ;
  • Z3 is N or CR Z3 ;
  • R Z1 , R Z2 and R Z3 are the same or different and are each independently selected from a hydrogen atom, a deuterium atom, a halogen, an alkyl group, a haloalkyl group, a deuterated alkyl group, an alkoxy group, a haloalkoxy group, a deuterated alkoxy group, a hydroxyl group, an alkenyl group, an alkynyl group, a cyano group, a nitro group, a C(O)R 30 , a C(O)OR 30 , a NR 31 R 32 , a C(O)NR 31 R 32 , a NR 31 C(O)R 30 , a cycloalkyl group, a cycloalkyloxy group, a heterocyclyl group, a heterocyclyloxy group, an aryl group and a heteroaryl group; the alkyl group, alkoxy group, alkenyl group, alky
  • Each R 30 is the same or different and is independently selected from a hydrogen atom, an alkyl group, an alkenyl group, an alkynyl group, a cycloalkyl group, a heterocyclic group, an aryl group and a heteroaryl group; the alkyl group, the alkenyl group, the alkynyl group, the cycloalkyl group, the heterocyclic group, the aryl group and the heteroaryl group are independently optionally substituted by one or more R 02 ;
  • Each R 31 and R 32 are the same or different and are independently selected from hydrogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl and heterocyclic groups; the alkyl, alkoxy, cycloalkyl and heterocyclic groups are independently substituted by one or more R 02 ;
  • each R 02 is the same or different and is independently selected from a deuterium atom, an oxo group, a halogen, an alkyl group, an alkoxy group, a haloalkyl group, a haloalkoxy group, a hydroxyl group, a hydroxyalkyl group, a hydroxyalkoxy group, a cyano group, an alkenyl group, an alkynyl group, an amino group, -NHalkyl group, -N(alkyl) 2 , an -alkylene-amino group, an -alkylene-NHalkyl group, an -alkylene-N(alkyl) 2 group, an amide group, a nitro group, a cycloalkyl group, a heterocyclyl group, a cycloalkylalkyl group, a heterocyclylalkyl group, a cycloalkyloxy group, a heterocyclyloxy group, an aryl
  • J1 , J2 , J3 , J4 , J5 and J6 are the same or different and are each independently selected from a bond, O, S, -O-alkylene-, -alkylene-O-, C(O), -C(O)-alkylene-, -alkylene-C(O)-, C(O)N( RJ ), N( RJ )C(O), N( RJ )C(O)N( RJ ), S(O) v , S(O) vN ( RJ ), N( RJ )S(O) v , N( RJ ), -N( RJ )-alkylene-, -alkylene-N( RJ )-, alkylene, alkenyl, alkynyl, cycloalkyl, heterocyclyl, -alkylene-heterocyclyl-, -heterocyclyl- Alkylene-, aryl and heteroaryl; the
  • Each R J is the same or different and is independently selected from a hydrogen atom, an alkyl group, an alkoxy group, a haloalkyl group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the cycloalkyl group and the heterocyclic group are independently substituted by one or more substituents selected from R 03 ;
  • each R 03 is the same or different and is independently selected from a deuterium atom, an oxo group, a halogen, an alkyl group, an alkoxy group, a haloalkyl group, a haloalkoxy group, a hydroxyl group, a hydroxyalkyl group, a hydroxyalkoxy group, a cyano group, an alkenyl group, an alkynyl group, an amino group, -NHalkyl group, -N(alkyl) 2 , an -alkylene-amino group, an -alkylene-NHalkyl group, an -alkylene-N(alkyl) 2 , an amide group, a nitro group, a cycloalkyl group, a heterocyclyl group, a cycloalkylalkyl group, a heterocyclylalkyl group, a cycloalkyloxy group, a heterocyclyloxy group, an aryl
  • E is selected from:
  • One of Q 1 , Q 2 , Q 3 , Q 4 and Q 5 is a carbon atom and is connected to J 6 , and the others are each independently selected from N or CR Q ;
  • Each R Q is the same or different and is independently selected from a hydrogen atom, a deuterium atom, a halogen, an alkyl group, a haloalkyl group, a deuterated alkyl group, an alkoxy group, a haloalkoxy group, a deuterated alkoxy group, a hydroxyl group, an alkenyl group, an alkynyl group, a cyano group, a NR c R d , an alkylene group NR c R d , a C(O)alkyl group, a C(O)NR c R d , a cycloalkyl group, a heterocyclic group, an aryl group and a heteroaryl group; the alkyl group, the alkoxy group, the alkenyl group, the alkynyl group, the alkylene group, the cycloalkyl group, the heterocyclic group, the aryl group and a heteroaryl group
  • Each R c and R d are the same or different and are independently selected from hydrogen, halogen, alkyl, haloalkyl, deuterated alkyl, alkoxy, haloalkoxy, deuterated alkoxy, cycloalkyl and heterocyclic group; or R c , R d and the nitrogen atom to which they are attached together form a heterocyclic group, and the heterocyclic group is independently optionally substituted by one or more R 04 ;
  • Y2 is C( RY2 ) 2 or C(O);
  • Y1 is N or CR Y1 ;
  • Y3 is N or CR Y3 ;
  • Each R Y2 is the same or different and is independently selected from a hydrogen atom, a deuterium atom, an alkyl group, a haloalkyl group, an alkoxy group, a haloalkoxy group, an alkenyl group, an alkynyl group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the alkenyl group, the alkynyl group, the cycloalkyl group and the heterocyclic group are independently optionally substituted by one or more R O4 ;
  • R Y2 together with the carbon atom to which they are connected form a cycloalkyl or heterocyclic group; the cycloalkyl and heterocyclic groups are each independently optionally substituted by one or more R 04 ;
  • R Y1 and R Y3 are the same or different and are each independently selected from a hydrogen atom, a deuterium atom, an alkyl group, a haloalkyl group, an alkoxy group, a haloalkoxy group, an alkenyl group, an alkynyl group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the alkenyl group, the alkynyl group, the cycloalkyl group and the heterocyclic group are each independently optionally substituted by one or more R O4 ;
  • R 6 , R 7 and R 8 are the same or different and are each independently selected from a hydrogen atom, an alkyl group, a halogenated alkyl group, a deuterated alkyl group, an alkoxy group, a halogenated alkoxy group, a deuterated alkoxy group, a cycloalkyl group and a heterocyclic group;
  • each R 04 is the same or different and is independently selected from a deuterium atom, an oxo group, a halogen, an alkyl group, an alkoxy group, a haloalkyl group, a haloalkoxy group, a hydroxyl group, a hydroxyalkyl group, a hydroxyalkoxy group, a cyano group, an alkenyl group, an alkynyl group, an amino group, an -NHalkyl group, an -N(alkyl) 2 group, an -alkylene-amino group, an -alkylene-NHalkyl group, an -alkylene-N(alkyl) 2 group , an amide group, a nitro group, a cycloalkyl group, a heterocyclyl group, a cycloalkylalkyl group, a heterocyclylalkyl group, a cycloalkyloxy group, a heterocyclyloxy group, an
  • n 0, 1, 2, 3, 4, 5 or 6;
  • r 0, 1, or 2;
  • v 0, 1, or 2;
  • n1, n2 and m are each independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
  • the purpose of the present disclosure is to provide a compound represented by general formula (I), or a pharmaceutically acceptable salt thereof:
  • R 1 is selected from -P(O)R 10 R 11 , -SO 2 R 12 , -SO 2 N(R 13 R 14 ) and -N(R 15 )SO 2 R 16 ;
  • R2 and R3 are the same or different and are each independently selected from a hydrogen atom, a halogen, an alkyl, a haloalkyl , an alkoxy , a haloalkoxy, a hydroxyl, an alkenyl, an alkynyl, a cyano, a nitro, a NR20R21 , a C ( O) NR20R21 , a NR20C(O) R24 , an -alkylene - NR20R21 , an -alkylene-C(O ) NR20R21, a C(O) R24 , a C (O) OR24 , a cycloalkyl, a cycloalkylalkyl, a heterocyclic group, a heterocyclic group, a cycloalkyloxy, a heterocyclic groupoxy, an aryl and a heteroaryl group; and R 01 is optionally substituted with one or more R
  • R4 and R5 are the same or different and are each independently selected from a hydrogen atom, a halogen, an alkyl, a haloalkyl, an alkoxy , a haloalkoxy, a hydroxyl, an alkenyl, an alkynyl, a cyano, a nitro, a NR22R23 , a C(O) NR22R23 , a NR22C (O) R24 , a C(O) R24 , a C(O) OR24 , a cycloalkyl, a heterocyclyl, a heterocyclylalkyl, a heterocyclyloxy, an aryl and a heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, a heterocyclyl, a heterocyclylalkyl, a heterocyclyloxy, an aryl and a heteroaryl; the alkyl
  • Ring A is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • Each RA is the same or different and is independently selected from oxo, halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxy, alkenyl, alkynyl, cyano , nitro, NR25R26, C(O)NR25R26 , cycloalkyl , heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl are each independently optionally substituted with one or more R01 ;
  • L1 and L3 are the same or different and are each independently selected from a bond, O, S, NR L , S(O) r , C(O), C(O) NR L , NR L C(O), S(O) r NR L , NR L C(O) NR L , (CR L1 R L2 ) m , alkenyl and alkynyl; the alkenyl is optionally substituted with one or more R 01 ;
  • L2 is -(CR L3 R L4 ) n1 -R L5 -(CR L6 R L7 ) n2 -;
  • RL5 is selected from the group consisting of a bond, O, S, NR L ′, S(O), S(O) 2 , C(O), C(O)NR L ′, NR L ′C(O), and NR L ′C(O)NR L ′;
  • Each of RL1 , RL2 , RL3 , RL4 , RL6 and RL7 is the same or different and is independently selected from hydrogen, halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxy, alkenyl, alkynyl, cyano, cycloalkyl, heterocyclic, aryl and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclic, aryl and heteroaryl are independently substituted with one or more RL1 ;
  • RL1 , RL2 and the carbon atoms to which they are connected together form a cycloalkyl or heterocyclic group
  • RL3 , RL4 and the carbon atoms to which they are connected together form a cycloalkyl or heterocyclic group
  • RL6 , RL7 and the carbon atoms to which they are connected together form a cycloalkyl or heterocyclic group
  • the cycloalkyl or heterocyclic group is independently optionally substituted by one or more R01 ;
  • R10 , R11 , R12 , R13 , R14 , R15 and R16 are the same or different and are each independently selected from a hydrogen atom, an alkyl group, a halogenated alkyl group, an alkoxy group, a halogenated alkoxy group, an alkenyl group, an alkynyl group, a cycloalkyl group and a heterocyclic group; Alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl and heterocyclyl are each independently optionally substituted with one or more R 01 ;
  • Each of R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , RL and RL ' is the same or different and is independently selected from a hydrogen atom, an alkyl group, a haloalkyl group, an alkoxy group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the cycloalkyl group and the heterocyclic group are independently optionally substituted by one or more R 01 ;
  • R13 , R14 and the nitrogen atom to which they are connected together form a heterocyclic group; or NR15 and the carbon atom to which it is connected, R2 and the carbon atom to which it is connected together form a nitrogen-containing heterocyclic group or a nitrogen-containing heteroaryl group; or R20 , R21 and the nitrogen atom to which they are connected together form a heterocyclic group; or R22 , R23 and the nitrogen atom to which they are connected together form a heterocyclic group; or R25 , R26 and the nitrogen atom to which they are connected together form a heterocyclic group; the heterocyclic group, the nitrogen-containing heterocyclic group or the nitrogen-containing heteroaryl group are each independently optionally substituted by one or more R01 ;
  • Ra and Rb are the same or different and are each independently selected from a hydrogen atom, an alkyl group, a haloalkyl group, a cycloalkyl group, and a heterocyclic group;
  • each R 01 is the same or different and is independently selected from oxo, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, hydroxyalkoxy, cyano, alkenyl, alkynyl, amino, -NHalkyl, -N(alkyl) 2 , -alkylene-amino, -alkylene-NHalkyl, -alkylene-N(alkyl) 2 , amido, nitro, cycloalkyl, heterocyclyl, cycloalkylalkyl, heterocyclylalkyl, cycloalkyloxy, heterocyclyloxy, aryl and heteroaryl;
  • Z1 is N or CR Z1 ;
  • Z2 is N or CR Z2 ;
  • Z3 is N or CR Z3 ;
  • R Z1 , R Z2 and R Z3 are the same or different and are each independently selected from a hydrogen atom, a halogen, an alkyl, a haloalkyl, an alkoxy, a haloalkoxy, a hydroxyl, an alkenyl, an alkynyl, a cyano, a nitro, C(O)R 30 , C(O)OR 30 , NR 31 R 32 , C(O)NR 31 R 32 , NR 31 C(O)R 30 , a cycloalkyl, a cycloalkyloxy, a heterocyclyl, a heterocyclyloxy, an aryl and a heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkyloxy, a heterocyclyl, a heterocyclyloxy, an aryl and a heteroaryl; the alkyl, alkoxy, alken
  • Each R 30 is the same or different and is independently selected from a hydrogen atom, an alkyl group, an alkenyl group, an alkynyl group, a cycloalkyl group, a heterocyclic group, an aryl group and a heteroaryl group; the alkyl group, the alkenyl group, the alkynyl group, the cycloalkyl group, the heterocyclic group, the aryl group and the heteroaryl group are independently optionally substituted by one or more R 02 ;
  • Each R 31 and R 32 are the same or different and are independently selected from hydrogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl and heterocyclic groups; the alkyl, alkoxy, cycloalkyl and heterocyclic groups are independently substituted by one or more R 02 ;
  • Each R 02 is the same or different and is independently selected from oxo, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, hydroxyalkoxy, cyano, alkenyl, alkynyl, amino, -NHalkyl, -N(alkyl) 2 , -alkylene-amino, -alkylene-NHalkyl, -alkylene-N(alkyl) 2 , amide, nitro, cycloalkyl, heterocyclyl, cycloalkylalkyl, heterocyclylalkyl, cycloalkyloxy, heterocyclyloxy, aryl, and heteroaryl;
  • J1 , J2 , J3 , J4 , J5 and J6 are the same or different and are each independently selected from a bond, O, S, -O-alkylene-, -alkylene-O-, -C(O)-, -C(O)-alkylene-, -alkylene-C(O)-, -C(O)N(R J )-, -N(R J )C(O)-, -N(R J )C(O)N(R J )-, -S(O) v -, -S(O) v N(R J )-, -N(R J )S(O) v -, -N(R J )-, -N(R J )-alkylene-, -alkylene-N(R J ) )-, alkylene, alkenyl, alkynyl, cycloalkyl, heterocyclyl, -alkylene
  • Each R J is the same or different and is independently selected from a hydrogen atom, an alkyl group, an alkoxy group, a haloalkyl group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the cycloalkyl group and the heterocyclic group are independently substituted by one or more substituents selected from R 03 ;
  • each R 03 is the same or different and is independently selected from oxo, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, hydroxyalkoxy, cyano, alkenyl, alkynyl, amino, -NHalkyl, -N(alkyl) 2 , -alkylene-amino, -alkylene-NHalkyl, -alkylene-N(alkyl) 2 , amido, nitro, cycloalkyl, heterocyclyl, cycloalkylalkyl, heterocyclylalkyl, cycloalkyloxy, heterocyclyloxy, aryl and heteroaryl;
  • E is selected from:
  • One of Q 1 , Q 2 , Q 3 , Q 4 and Q 5 is a carbon atom and is connected to J 6 , and the others are each independently selected from N or CR Q ;
  • Each R Q is the same or different and is independently selected from hydrogen, halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxy, alkenyl, alkynyl, cyano, NR c R d , alkylene NR c R d , C(O)alkyl, C(O)NR c R d , cycloalkyl, heterocyclic, aryl and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, alkylene, cycloalkyl, heterocyclic, aryl and heteroaryl are each independently optionally substituted with one or more R 04 ;
  • R c and R d are the same or different and are independently selected from hydrogen, halogen, alkyl, cycloalkyl and heterocyclic groups; or R c , R d and the nitrogen atom to which they are connected together form a heterocyclic group, and the heterocyclic group is independently optionally substituted by one or more R 04 ;
  • Y2 is C( RY2 ) 2 or C(O);
  • Y1 is N or CR Y1 ;
  • Y3 is N or CR Y3 ;
  • Each R Y2 is the same or different and is independently selected from a hydrogen atom, an alkyl group, a haloalkyl group, an alkoxy group, a haloalkoxy group, an alkenyl group, an alkynyl group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the alkenyl group, the alkynyl group, the cycloalkyl group and the heterocyclic group are independently optionally substituted by one or more R 04 ;
  • R Y2 together with the carbon atom to which they are connected form a cycloalkyl or heterocyclic group; the cycloalkyl and heterocyclic groups are each independently optionally substituted by one or more R 04 ;
  • R Y1 and R Y3 are the same or different and are each independently selected from a hydrogen atom, an alkyl group, a haloalkyl group, an alkoxy group, a haloalkoxy group, an alkenyl group, an alkynyl group, a cycloalkyl group and a heterocyclic group; the alkyl group, the alkoxy group, the alkenyl group, the alkynyl group, the cycloalkyl group and the heterocyclic group are each independently optionally substituted by one or more R 04 ;
  • R 6 , R 7 and R 8 are the same or different and are each independently selected from a hydrogen atom, an alkyl group, a haloalkyl group, a cycloalkyl group and a heterocyclic group;
  • each R 04 is the same or different and is independently selected from oxo, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, hydroxyalkoxy, cyano, alkenyl, alkynyl, amino, -NHalkyl, -N(alkyl) 2 , -alkylene-amino, -alkylene-NHalkyl, -alkylene-N(alkyl) 2 , amido, nitro, cycloalkyl, heterocyclyl, cycloalkylalkyl, heterocyclylalkyl, cycloalkyloxy, heterocyclyloxy, aryl and heteroaryl;
  • n 0, 1, 2, 3, 4, 5 or 6;
  • r is 1 or 2;
  • v 1 or 2;
  • n1, n2 and m are each independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
  • the compound represented by the general formula (X) or (I) or a pharmaceutically acceptable salt thereof wherein ring A is phenyl or a 5- or 6-membered heteroaryl; preferably a 5- or 6-membered heteroaryl; more preferably a 5-membered heteroaryl; further preferably a pyrazolyl; most preferably
  • the compound represented by the general formula (X) or (I) or a pharmaceutically acceptable salt thereof wherein for Each RA is the same or different and is independently selected from halogen, C1-6 alkyl and C1-6 haloalkyl, and n is 0, 1 or 2; more preferably
  • the compound represented by the general formula (X) or (I) or a pharmaceutically acceptable salt thereof wherein for n3 is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10; n is 0, 1 or 2; RL3 , RL4 and RA are as defined in the general formula (I); preferably, for Each RL3 and RL4 are the same or different and are each independently selected from a hydrogen atom, a halogen, a C1-6 alkyl group and a C1-6 haloalkyl group, each RA is the same or different and is each independently selected from a halogen, a C1-6 alkyl group and a C1-6 haloalkyl group, n is 0, 1 or 2; n3 is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10;
  • n3 is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10; the bond with & is connected to the ring where Z 1 is located.
  • the compound represented by the general formula (X) or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (X') or a pharmaceutically acceptable salt thereof,
  • n 0, 1 or 2;
  • R 1 , R 9 , y, R 4 , R 5 , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , RA , J 1 to J 6 and E are as defined in the general formula (X).
  • the compound represented by the general formula (X) or (X') or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (X'-1) or a pharmaceutically acceptable salt thereof,
  • ring J 2 ' is a nitrogen-containing heterocyclic group; j is 0, 1, 2, 3, 4, 5 or 6;
  • R1 , R9 , y, R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , J1 , R03 , J4 , J5 , J6 and E are as defined in the general formula (X).
  • the compound represented by the general formula (X) or (X') or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (X'-2) or a pharmaceutically acceptable salt thereof,
  • ring J 3 ' is a nitrogen-containing heterocyclic group; j is 0, 1, 2, 3, 4, 5 or 6;
  • R1 , R9 , y, R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , J1 , R03 , J4 , J5 , J6 and E are as defined in the general formula (X).
  • E is selected from: s is 0, 1, 2 or 3, Y 1 , Y 2 , R 6 , R 7 , R 8 and R q are as defined in the general formula (X);
  • E is selected from: s is 0, 1, 2 or 3, Y 1 , Y 2 , R 6 , R 7 , R 8 and R q are as defined in the general formula (X);
  • E is selected from:
  • E is selected from:
  • Each R q is the same or different and is independently selected from halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxy, alkenyl, alkynyl, cyano, NR c R d , alkylene NR c R d , C(O)alkyl, C(O)NR c R d , cycloalkyl, heterocyclyl, aryl and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, alkylene, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more R 04 ; s is 0, 1, 2 or 3; Y 1 , Y 2 , R 6 , R 7 , R c , R d and R 04 are as defined in the general formula (I);
  • E is selected from: Y 1 is N or CH; Y 2 is CH 2 or C(O); each R q is the same or different and is independently selected from halogen, C 1-6 alkyl and C 1-6 haloalkyl; R 6 is a hydrogen atom or C 1-6 alkyl; R 7 is C 1-6 alkyl; s is 0, 1, 2 or 3;
  • E is selected from: Y 1 is N or CH; Y 2 is CH 2 or C(O); each R q is the same or different and is independently selected from halogen, C 1-6 alkyl and C 1-6 haloalkyl; R 6 is a hydrogen atom, R 7 is a C 1-6 alkyl, and R 8 is a C 1-6 alkyl; s is 0, 1, 2 or 3;
  • E is selected from:
  • E is selected from: R6 and R7 are the same or different and are each independently a hydrogen atom or a C1-6 alkyl group; each Rq is the same or different and is each independently selected from halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxy, alkenyl, alkynyl, cyano , NRcRd , alkyleneNRcRd , C(O) alkyl , C(O) NRcRd , cycloalkyl , heterocyclyl, aryl and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, alkylene, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted by one or more R04 ; s is 0, 1, 2 or 3; Rc , Rd and R04 are as defined in the general formula (I);
  • E is selected from R6 and R7 are the same or different and are each independently a hydrogen atom or a C1-6 alkyl group;
  • E is selected from
  • E is selected from:
  • the compound represented by the general formula (X) or (I) or a pharmaceutically acceptable salt thereof wherein Q 2 is a carbon atom and is connected to J 6 , Q 1 , Q 3 , Q 4 and Q 5 are all CR Q , and R Q is as defined in the general formula (I); in some embodiments, Q 2 is a carbon atom and is connected to J 6 , Q 1 , Q 3 , Q 4 and Q 5 are the same or different, and each independently is CH or CF; preferably, Q 2 is a carbon atom and is connected to J 6 , and Q 1 , Q 3 , Q 4 and Q 5 are all CH.
  • the compound represented by the general formula (X) or a pharmaceutically acceptable salt thereof wherein one of Q1 or Q2 is a carbon atom and is connected to J6 , the other as well as Q3 and Q4 are all CH or CRq , and Rq is as defined in the general formula (X); in some embodiments, Q2 is a carbon atom and is connected to J6 , Q1 , Q3 and Q4 are all CH or CRq , and Rq is as defined in the general formula (X); in some embodiments, Q2 is a carbon atom and is connected to J6 , Q1 , Q3 and Q4 are the same or different, and each independently is CH or CF.
  • each R Q is the same or different and is independently selected from a hydrogen atom, a halogen, a C 1-6 alkyl, a C 1-6 alkoxy, a C 1-6 haloalkyl, a C 1-6 haloalkoxy, a C 2-6 alkenyl, a C 2-6 alkynyl and a cyano group; preferably, each R Q is a hydrogen atom.
  • the compound represented by the general formula (I) or (X) or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (I'-1), (I'-2) or (I'-3) or a pharmaceutically acceptable salt thereof,
  • each R q is the same or different and is independently selected from halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxy, alkenyl, alkynyl, cyano, NR c R d , alkylene NR c R d , C(O)alkyl, C(O)NR c R d , cycloalkyl, heterocyclyl, aryl and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, alkylene, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more R 04 ;
  • R1 to R7 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , Rc , Rd , R04 , J1 to J6 , Y1 and Y2 are as defined in the general formula (I).
  • the compound represented by the general formula (I), (I'-3), (X) or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (I'-3-1) or (I'-3-2) or a pharmaceutically acceptable salt thereof,
  • R1 to R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 to J6 , Rq , Y2 and s are as defined in the general formula (I'-3).
  • the compound represented by the general formula (I), (I'-1), (I'-2), (X) or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (II) or (II') or a pharmaceutically acceptable salt thereof,
  • Each R q is the same or different and is independently selected from halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxy, alkenyl, alkynyl, cyano, NR c R d , alkylene NR c R d , C(O)alkyl, C(O)NR c R d , cycloalkyl, heterocyclyl, aryl and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, alkylene, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more R 04 ;
  • v1 is 0, 1, 2, 3, 4, 5, or 6
  • v2 is 0, 1, 2, 3, 4, 5, or 6;
  • R1 to R7 , RA , Ra , Rb , Rc , Rd, RZ1 , RZ2 , L1 , L2 , J2 , J6 , R04 and Y1 are as defined in the general formula (I).
  • X1 and X2 are each independently selected from CRb1 , CRb1Rb2 , N, NRb3 , O and S, each RB is the same or different and is each independently selected from oxo, halogen, C1-6 alkyl, C1-6 alkoxy, C1-6 haloalkyl , halogenated C1-6 alkoxy, cyano, C2-6 alkenyl, C2-6 alkynyl , 3- to 10-membered cycloalkyl and 3- to 10-membered heterocyclyl;
  • R b1 and R b2 are the same or different and are each independently selected from oxo, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, hydroxyalkoxy, cyano, alkenyl, alkynyl, amino, -NHalkyl, -N(alkyl) 2 , amide, nitro, cycloalkyl, heterocyclyl, cycloalkylalkyl, heterocyclylalkyl, cycloalkyloxy, heterocyclyloxy, aryl and heteroaryl;
  • R b3 is selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl and heterocyclyl; t is 0, 1, 2 or 3; u is 0, 1 or 2, the bond with ** is connected to L 1 , and R 1 and R 2 are as defined in the general formula (I);
  • R 1 is -P(O)R 10 R 11 or -N(R 15 )SO 2 R 16 ;
  • R 10 , R 11 , R 15 and R 16 are the same or different and are each independently a hydrogen atom or a C 1-6 alkyl group;
  • X 1 is CH, CR b1 or N, each RB and R b1 are the same or different and are each independently selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl and 3 to 6 membered cycloalkyl;
  • t is 0, 1 or 2; the bond with ** is connected to L 1 ;
  • R 1 is -P(O)R 10 R 11 or -N(R 15 )SO 2 R 16 ;
  • R 10 , R 11 , R 15 and R 16 are the same or different and are each independently a hydrogen atom or a C 1-6 alkyl group; the bond with ** is connected to L 1 ;
  • ring B is selected from a 5- or 6-membered heterocyclic group, a phenyl group, and a 5- or 6-membered heteroaryl group; preferably selected from a phenyl group, a pyridyl group, and a pyrazinyl group.
  • Selected from In some embodiments, for The bonds with ** are connected to L1 .
  • each R b1 and R b2 are the same or different and are each independently selected from a hydrogen atom, a halogen, a C 1-6 alkyl, a C 1-6 alkoxy, a C 1-6 haloalkyl, a halosubstituted C 1-6 alkoxy, a cyano group, a C 2-6 alkenyl group, a C 2-6 alkynyl group, a 3- to 10-membered cycloalkyl group and a 3- to 10-membered heterocyclic group.
  • the compound represented by the general formula (X) or a pharmaceutically acceptable salt thereof wherein for y, R 1 and R 9 are as defined in Formula (X); in some embodiments, for R 9a is a hydrogen atom or R 9 , R 1 and R 9 are as defined in the general formula (X); the bond with ** is connected to L 1 .
  • the compound represented by the general formula (I), (I'-1), (I'-2), (X), (X') or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (III) or (III') or a pharmaceutically acceptable salt thereof,
  • Ring J 2 ' is a nitrogen-containing heterocyclic group, and the nitrogen-containing heterocyclic group is optionally substituted by one or more R 03 ;
  • Each R q is the same or different and is independently selected from halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxy, alkenyl, alkynyl, cyano, NR c R d , alkylene NR c R d , C(O)alkyl, C(O)NR c R d , cycloalkyl, heterocyclyl, aryl and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, alkylene, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more R 04 ;
  • R1 , R2 , R4 to R7 , RA , Ra , Rb , Rc , Rd, RZ1 , RZ2 , L1 , L2 , J1 , J5 , J6 , R03 , R04 and Y1 are as defined in the general formula (I).
  • the compound represented by the general formula (I), (I'-1), (I'-2), (X), (X') or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (IV) or (IV') or a pharmaceutically acceptable salt thereof,
  • Ring J 6 ' is a nitrogen-containing heterocyclic group, and the nitrogen-containing heterocyclic group is optionally substituted by one or more R 03 ;
  • Each R q is the same or different and is independently selected from halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxy, alkenyl, alkynyl, cyano, NR c R d , alkylene NR c R d , C(O)alkyl, C(O)NR c R d , cycloalkyl, heterocyclyl, aryl and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, alkylene, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more R 04 ;
  • p 0, 1, 2, 3, 4, 5 or 6;
  • R1 , R2 , R4 to R7 , RA , Ra , Rb , Rc , Rd, RZ1 , RZ2 , L1 , L2 , J1 , J2 , R03 , R04 and Y1 are as defined in the general formula (I).
  • the compound represented by the general formula (I), (I'-3), (X), (X'), (X'-1) or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (V-1) or a pharmaceutically acceptable salt thereof,
  • ring J 2 ' is a nitrogen-containing heterocyclic group
  • R1 , R2 , R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , J1 , R03 , J4 , J5 , J6 , Rq , and Y2 are as defined in the general formula (I'-3).
  • the compound represented by the general formula (I), (I'-3), (X), (X'), (X'-2) or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (V-2) or a pharmaceutically acceptable salt thereof,
  • ring J 3 ' is a nitrogen-containing heterocyclic group
  • R 1 , R 2 , R 4 , R 5 , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , RA , J 1 , R 03 , J 4 , J 5 , J 6 , R q and Y 2 are as defined in the general formula (I'-3).
  • the compound represented by the general formula (I), (I'-3), (I'-3-1), (I'-3-2), (V-1), (X), (X'), (X'-1) or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (V-1-1) or (V-1-2) or a pharmaceutically acceptable salt thereof,
  • R 1 , R 2 , R 4 , R 5 , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , RA , n, J 1 , ring J 2 ′, R 03 , j, q, J 4 , J 5 , J 6 , R q , Y 2 and s are as defined in the general formula (V-1).
  • the compound represented by the general formula (I), (I'-3), (I'-3-1), (I'-3-2), (V-2), (X), (X'), (X'-2) or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (V-2-1) or (V-2-2) or a pharmaceutically acceptable salt thereof,
  • R 1 , R 2 , R 4 , R 5 , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , RA , n, J 1 , q, ring J 3 ′, R 03 , j, J 4 , J 5 , J 6 , R q , Y 2 and s are as defined in the general formula (V-2).
  • the compound represented by the general formula (X), (X'), (X'-2) or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (X'-3) or a pharmaceutically acceptable salt thereof,
  • ring J 3 ' is a nitrogen-containing heterocyclic group; j is 0, 1, 2, 3, 4, 5 or 6;
  • R1 , R9 , Y, R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , J1 , R03 , J4 , J5 , J6 , Rq , Y1 and R7 are as defined in the general formula (X).
  • the compounds represented by the general formula (X'-1), (III), (III'), (V-1), (V-1-1), (V-1-2) or their pharmaceutically acceptable salts wherein ring J 2 ' is a 3- to 12-membered nitrogen-containing heterocyclic group; in some embodiments, ring J 2 ' is a 5- or 6-membered monocyclic heterocyclic group containing at least one nitrogen atom or a 7- to 14-membered spiroheterocyclic group containing at least one nitrogen atom; in some embodiments, ring J 2 ' is selected from piperidinyl, piperazinyl and diazaspiro[3.5]nonane.
  • the compounds represented by the general formula (V-2), (V-2-1), (V-2-2), (X'-2), (X'-3) or their pharmaceutically acceptable salts wherein ring J 3 ' is a 3- to 12-membered nitrogen-containing heterocyclic group; in some embodiments, ring J 3 ' is a 3- to 10-membered nitrogen-containing heterocyclic group; in some embodiments, ring J 3 ' is a piperidinyl or piperazinyl; in some embodiments, ring J 3 ' is selected from
  • the compounds represented by the general formula (V-1), (V-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X'-1), (X'-2), (X'-3) or their pharmaceutically acceptable salts wherein each R 03 is the same or different and is independently selected from oxo, halogen, hydroxyl and C 1-6 alkyl; and/or j is 0, 1 or 2.
  • the compounds represented by the general formula (I), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X') or their pharmaceutically acceptable salts are wherein Y 2 is CH 2 or C(O); in some embodiments, Y 2 is CH 2 ; in some embodiments, Y 2 is C(O).
  • L 3 is a bond or a C 1-10 alkylene group; preferably a bond.
  • the compound represented by the general formula (X) or (I) or a pharmaceutically acceptable salt thereof wherein L 1 is O, L 2 is a C 1-10 alkylene group, and L 3 is a bond; preferably, L 1 is O, L 2 is selected from (CH 2 ) 3 , (CH 2 ) 4 and (CH 2 ) 5 , and L 3 is a bond.
  • J 1 is a bond or a 3- to 12-membered heterocyclyl; the 3- to 12-membered heterocyclyl is optionally substituted with one or more selected from oxo, halogen, hydroxyl, and C 1-6 alkyl; in some embodiments, J 1 is selected from a bond, piperidinyl, and piperazinyl, the piperidinyl and piperazinyl are each independently optionally substituted with one or more selected from oxo, halogen, hydroxyl, and C 1-6 alkyl; in some embodiments, J 1 is selected from a bond, piperidinyl, and piperazinyl.
  • J2 is a 3- to 12-membered cycloalkyl or 3- to 12-membered heterocyclyl optionally substituted by one or more R03 , R03 is selected from oxo, hydroxyl, halogen, C1-6 alkyl and C1-6 haloalkyl; More preferably, J2 is a 4- to 7-membered monocyclic heterocyclyl or 7- to 14-membered spiroheterocyclyl optionally substituted by one or more R03 , R03 is selected from hydroxyl, halogen, C1-6 alkyl and C1-6 haloalkyl; Further preferably, J2 is a 5- or 6-membered heterocyclyl optionally substituted by one or more R03 , R03 is selected from hydroxyl, halogen, C1-6 alkyl and C1-6 haloalkyl; Most preferably, J2 is a 1,4-piperidinyl or 1,4-piperazinyl optionally substituted by one or more R03 , R03
  • J2 is selected from The above ring is optionally substituted by one or more substituents selected from oxo, halogen, hydroxyl, C 1-6 alkyl and C 1-6 haloalkyl; preferably, J 2 is selected from More preferably, J2 is In some embodiments, J2 is selected from The key with * is connected to J 3 ;
  • J2 is selected from piperidinyl, piperazinyl and diazaspiro[3.5]nonane; the piperidinyl, piperazinyl and diazaspiro[3.5]nonane are each independently optionally substituted by one or more selected from oxo, halogen, hydroxyl and C1-6 alkyl; in some embodiments, J2 is piperidinyl or piperazinyl; the piperidinyl and piperazinyl are each independently optionally substituted by one or more selected from oxo, halogen, hydroxyl and C1-6 alkyl.
  • the compounds represented by the general formula (III), (III'), (V-1), (V-1-1), (V-1-2), (X'-1) or pharmaceutically acceptable salts thereof wherein ring J 2 ' is a 4- to 7-membered nitrogen-containing monocyclic heterocyclic group or a 7- to 14-membered nitrogen-containing spiro heterocyclic group optionally substituted by one or more R 03 , and R 03 is selected from hydroxyl, halogen, C 1-6 alkyl and C 1-6 haloalkyl; preferably, ring J 2 ' is a 5- or 6-membered nitrogen-containing heterocyclic group optionally substituted by one or more R 03 , and R 03 is selected from hydroxyl, halogen, C 1-6 alkyl and C 1-6 haloalkyl; more preferably, ring J 2 ' is a 1,4-piperidinyl or 1,4-piperazinyl group optionally substituted by one or more R 03 , and
  • ring J 2 ' is selected from The bonds with * are connected to C(O).
  • the compounds represented by the general formula (I), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) or pharmaceutically acceptable salts thereof, wherein J 4 is selected from a bond, O, S, -OC 1-6 alkylene-, -C 1-6 alkylene-O-, C(O), -C(O)-C 1-6 alkylene-, -C 1-6 alkylene-C(O)-, C(O)N(R J ), N(R J )C(O), N(R J ), -N(R J )-C 1-6 alkylene-, -C 1-6 alkylene-N(R J )-, C R 03 and R 04 are each independently substituted with one or more R 03, and R
  • the compounds represented by the general formula (I), (II), (III), (II'), (III'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) or pharmaceutically acceptable salts thereof, wherein J 6 is selected from a bond, O, S, -OC 1-6 alkylene-, -C 1-6 alkylene-O-, C(O), -C(O)-C 1-6 alkylene-, -C 1-6 alkylene-C(O)-, C(O)N(R J ), N(R J )C(O), N(R J ), -N(R J )-C 1-6 alkylene-, -C R 03 ′ is as defined in the general formula ( I ); preferably,
  • J 6 is a 5- or 6-membered heterocyclic group optionally substituted by one or more R 03 ', R 03 ' is selected from hydroxyl, halogen, C 1-6 alkyl, C 1-6 halogenated alkyl, C 1-6 ...alkyl, C 1- and C 1-6 haloalkyl; further preferably, J 6 is 1,4-piperidinyl or 1,4-piperazinyl optionally substituted by one or more R 03 ', R 03 ' is selected from hydroxyl, halogen, C 1-6 alkyl and C 1-6 haloalkyl; most preferably selected from In some embodiments, J is selected from a bond, and N(R J ), R J is a hydrogen atom or a methyl group; in some embodiments, J 6 is selected from a bond, and NH, connected from left to right;
  • J is selected from a bond, NH,
  • J 6 is N(R J ) or a 3- to 12-membered heterocyclic group, wherein the 3- to 12-membered heterocyclic group is optionally substituted by one or more selected from halogen, hydroxyl and C 1-6 alkyl;
  • R J is a hydrogen atom or C 1-6 alkyl;
  • J 6 is selected from a bond, N(R J ) and a 3- to 12-membered heterocyclic group, wherein the 3- to 12-membered heterocyclic group is optionally substituted by one or more selected from halogen, hydroxyl and C 1-6 alkyl;
  • R J is a hydrogen atom or C 1-6 alkyl;
  • J 6 is selected from a bond, N(R J ), piperidinyl and piperazinyl, and the piperidinyl and piperazinyl are each independently substituted with one or more selected from oxo, halogen, hydroxyl and C 1-6 alkyl, and R J is a hydrogen atom or a methyl group;
  • J 6 is selected from N(R J ), piperidinyl and piperazinyl, and the piperidinyl and piperazinyl are each independently substituted with one or more selected from oxo, halogen, hydroxyl and C 1-6 alkyl, and R J is a hydrogen atom or a methyl group;
  • J 6 is selected from a bond, O, N(R J ) and C 1-6 alkylene, R J is as defined in the general formula (I); in some embodiments, J 6 is selected from a bond, O, N(R J ) and (CH 2 ) q , q is 0, 1, 2, 3 or 4, R J is a hydrogen atom or a C 1-6 alkyl group.
  • the compound represented by the general formula (IV) or (IV') or a pharmaceutically acceptable salt thereof wherein ring J 6 ' is a 5- or 6-membered nitrogen-containing heterocyclic group optionally substituted by one or more R 03 ', R 03 ' is selected from hydroxyl, halogen, C 1-6 alkyl and C 1-6 haloalkyl; more preferably, ring J 6 ' is a 1,4-piperidinyl or 1,4-piperazinyl group optionally substituted by one or more R 03 ', R 03 ' is selected from hydroxyl, halogen, C 1-6 alkyl and C 1-6 haloalkyl; most preferably Selected from In some embodiments, ring J 6 ' is selected from The bonds with # are connected to the benzene ring.
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from
  • -J 4 -J 5 -J 6 - is selected from -C(O)-C 1-6 alkylene-3 to 8-membered heterocyclyl-, -C(O)-3 to 8-membered heterocyclyl-, -3 to 12-membered heterocyclyl-, -3 to 12-membered heterocyclyl-C 0-6 alkylene-3 to 12-membered heterocyclyl- and -C(O)-C 1-6 alkylene-N(R J )-;
  • R J is a hydrogen atom or a C 1-6 alkyl group; the 3 to 8-membered heterocyclyl and the 3 to 12-membered heterocyclyl are each independently optionally substituted by one or more selected from oxo, halogen, hydroxyl and C 1-6 alkyl;
  • R J is a hydrogen atom or a C 1-6 alkyl group; in some embodiments, -J 4 -J 5 -J 6 - is selected from -C(O
  • -J 4 -J 5 -J 6 - is a bond or a 3 to 12 membered heterocyclyl optionally substituted with one or more selected from halogen, hydroxy and C 1-6 alkyl; in some embodiments, -J 4 -J 5 -J 6 - is a bond; in some embodiments, -J 4 -J 5 -J 6 - is a 3 to 12 membered heterocyclyl optionally substituted with one or more selected from halogen, hydroxy and C 1-6 alkyl.
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from -heterocyclyl-C(O)-C 1-6 alkylene-heterocyclyl-, -heterocyclyl-C 1-6 alkylene-C(O)-heterocyclyl-, -heterocyclyl-C 1-6 alkylene-heterocyclyl-, -heterocyclyl-C 0-6 alkylene-heterocyclyl-C 0-6 alkylene-heterocyclyl- and -heterocyclyl-C 0-6 alkylene-heterocyclyl-C(O)-C 1-6 alkylene-N(R J )-;
  • the heterocyclyl is optionally substituted with one or more substituents selected from R 03 ;
  • R 03 and R J are as defined in the general formula (I);
  • -J1 - J2 - J3 - J4 - J5 - J6- is selected from -3 to 8-membered heterocyclyl-C(O) -C1-6 alkylene-3 to 8-membered heterocyclyl-, -3 to 8-membered heterocyclyl- C1-6 alkylene-C(O)-3 to 8-membered heterocyclyl-, -3 to 12-membered heterocyclyl- C1-6 alkylene-3 to 12-membered heterocyclyl-, -3 to 12-membered heterocyclyl- C0-6 alkylene-3 to 12-membered heterocyclyl-, and -3 to 12-membered heterocyclyl -C0-6 alkylene -3 to 12-membered heterocyclyl-C(O) -C1-6 alkylene-N( RJ )-; the 3 to 8-membered heterocyclyl and the 3 to 12-membered heterocyclyl are each independently selected from oxo, halogen,
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from X, Y, U, V, S and T are the same or different and are each independently CR 05 or N;
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from X, Y, U, V, S and T are the same or different and are each independently selected from CH, CR 03 and N; u, q, w, x, k, l and q1 are the same or different and are each independently 0, 1, 2, 3, 4, 5 or 6; a, b, c, e and f are the same or different and are each independently 0, 1, 2 or 3; d is 1, 2 or 3; g and h are the same or different and are each independently 0, 1 or 2; each R J3 and R J5 are the same or different and are each independently a hydrogen atom or R 03 ; R 03 and R J are as defined in the general formula (I);
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from X, Y, U, V, S and T are the same or different and are each independently CR 05 or N;
  • each R J3 is the same or different and is each independently a hydrogen atom or a C 1-6 alkyl group
  • each R J5 is the same or different and is each independently a hydrogen atom or a C 1-6 alkyl group
  • each R 03 is the same or different and is each independently selected from an oxo group, a halogen, a hydroxyl group and a C 1-6 alkyl group
  • each R 05 is the same or different and is each independently selected from a hydrogen atom, a halogen, a hydroxyl group and a C 1-6 alkyl group
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from q, u and w are the same or different and are each independently 0, 1, 2, 3, 4, 5 or 6; each R 03 is the same or different and is each independently selected from oxo, halogen, hydroxyl and C 1-6 alkyl; g and h are each independently 0, 1 or 2; q1 is 0, 1, 2, 3, 4, 5 or 6;
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is q, u and w are the same or different and are each independently 0, 1, 2, 3, 4, 5 or 6; each R 03 is the same or different and is each independently selected from halogen, hydroxyl and C 1-6 alkyl; g and h are each independently 0, 1 or 2;
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from q, u, x and w are the same or different and are each independently 0, 1, 2, 3, 4, 5 or 6; each R 03 is the same or different and is each independently selected from oxo, halogen, hydroxyl and C 1-6 alkyl;
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from In some embodiments, -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from
  • J2 is a 4- to 7-membered monocyclic heterocyclic group or a 7- to 14-membered spiro heterocyclic group optionally substituted by one or more R03 , R03 is selected from hydroxyl, halogen, C1-6 alkyl and C1-6 haloalkyl; J3 is a bond or C1-6 alkylene; J4 is a bond or C(O); J5 is a bond or C1-6 alkylene; J6 is N( RJ ) or a 3- to 12-membered heterocyclic group, wherein the 3- to 12-membered heterocyclic group is optionally substituted by one or more of the following: halogen, hydroxyl and C1-6 alkyl; RJ is a hydrogen atom or a C1-6 alkyl;
  • J1 is selected from a bond, piperidinyl and piperazinyl, and the piperidinyl and piperazinyl are each independently substituted by one or more selected from oxo, halogen, hydroxyl and C1-6 alkyl;
  • J2 is a 4-7 membered monocyclic heterocyclyl or a 7-14 membered spiroheterocyclyl optionally substituted by one or more R03 , R03 is selected from hydroxyl, halogen, C1-6 alkyl and C1-6 haloalkyl;
  • J3 is a bond or C1-6 alkylene;
  • J4 is a bond or C(O);
  • J5 is a bond or C1-6 alkylene;
  • J6 is selected from a bond, N( RJ ) and a 4-7 membered heterocyclyl, and the 4-7 membered heterocyclyl is optionally substituted by one or more selected from oxo, halogen, hydroxyl and C1-6 alkyl
  • Selected from q, u, x and w are the same or different and are each independently 0, 1, 2, 3, 4, 5 or 6; each R 03 is the same or different and is each independently selected from oxo, halogen, hydroxyl and C 1-6 alkyl; k is 0, 1, 2, 3, 4, 5 or 6;
  • Selected from q, u, x and w are the same or different and are each independently 0, 1, 2, 3, 4, 5 or 6; each R 03 is the same or different and is each independently selected from oxo, halogen, hydroxyl and C 1-6 alkyl;
  • Selected from q, u, x and w are the same or different and are each independently 0, 1, 2, 3, 4, 5 or 6; g and h are each independently 0, 1 or 2; each R 03 is the same or different and is each independently selected from oxo, halogen, hydroxyl and C 1-6 alkyl; q1 is 1, 2, 3, 4, 5 or 6;
  • a, b, e and f are the same or different and are each independently 0 or 1; and/or c is 1 or 2; and/or d is 1 or 2; and/or g is 0 or 1; and/or h is 0 or 1.
  • u, x and w are the same or different and are each independently 0, 1 or 2.
  • i 2 or 3.
  • X is N; and/or at least one of U and V is N; and/or at least one of S and T is N.
  • each R 05 is the same or different, and is independently a hydrogen atom or a hydroxyl group.
  • q is 0 or 1
  • k is 0 or 1.
  • the compounds represented by the general formula (I), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X'-1), (X'-2), (X'-3) or their pharmaceutically acceptable salts wherein q is 1, 2, 3, 4 or 5; in some embodiments, q is 1, 2 or 3; in some embodiments, q is 1; in some embodiments, q is 0.
  • q1 is 1.
  • R J5 is C 1-6 alkyl; in some embodiments, R J5 is methyl, and/or l is 1.
  • R J3 is a hydrogen atom
  • R 1 is -P(O)R 10 R 11 or -N(R 15 )SO 2 R 16 ;
  • R 10 , R 11 , R 15 and R 16 are the same or different and are each independently selected from a hydrogen atom, a C 1-6 alkyl group and a 3- to 6-membered cycloalkyl group;
  • R 1 is -N(R 15 )SO 2 R 16 ;
  • R 15 and R 16 are the same or different and are each independently selected from a hydrogen atom, a C 1-6 alkyl group and a 3- to 6-membered cycloalkyl group;
  • R 1 is -N(R 15 )SO 2 R 16 ;
  • R 15 is selected from a hydrogen atom, a C 1-6 alkyl group and a 3- to 6-membered cycloalkyl group;
  • R 16 is a C 1-6 alkyl group; in some embodiments, R 1 is -N(CH 3 )SO 2 CH 3 or -N
  • the compounds represented by the general formula (I), (II), (II'), (I'-1), (I'-2), (I'-3), (I'-3-1), (I'-3-2) or their pharmaceutically acceptable salts wherein R3 is connected to L1 ; specifically, R3 is a bond and is connected to L1 .
  • the compounds represented by the general formula (I) to (IV), (II') to (IV') or their pharmaceutically acceptable salts wherein R Z3 is selected from hydrogen atom, halogen, C 1-6 alkyl and C 1-6 alkoxy; preferably a hydrogen atom.
  • the compounds represented by the general formula (I), (II') to (IV'), (I'-2), (X), (X'), (X'-1), (X'-2), (X'-3) or their pharmaceutically acceptable salts wherein R7 is selected from a hydrogen atom, a C1-6 alkyl group and a C1-6 haloalkyl group; preferably, R7 is a C1-6 alkyl group; more preferably, a methyl group.
  • R 8 is C 1-6 alkyl; preferably methyl.
  • each R q is the same or different and is independently selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, hydroxyl, cyano and 3 to 6 membered cycloalkyl; in some embodiments, each R q is the same or different and is independently selected from halogen, C 1-6 alkyl and C 1-6 haloalkyl; in some embodiments, each R q is the same or different and is independently selected from halogen, C 1-6 alkyl and C 1-6 haloalkyl; in some embodiments, each R q is the same or different and is independently selected from halogen, C 1-6 alkyl and C 1-6 haloalkyl; in some embodiments, each R q is the same or different and is the same or different and is
  • each R q is the same or different and is each independently selected from halogen, C 1-6 alkyl and C 1-6 haloalkyl; and/or s is 0 or 1.
  • each R 9 is the same or different and is independently selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, hydroxyl, cyano and 3 to 6 membered cycloalkyl; in some embodiments, each R 9 is the same or different and is independently selected from halogen, C 1-6 alkyl and C 1-6 haloalkyl; in some embodiments, each R 9 is the same or different and is independently selected from halogen; in some embodiments, R 9 is F;
  • each R 9 is the same or different, and is each independently selected from halogen, C 1-6 alkyl, and C 1-6 haloalkyl; and/or y is 0 or 1.
  • R 9a is selected from a hydrogen atom, a halogen, a C 1-6 alkyl group, and a C 1-6 haloalkyl group; in some embodiments, R 9a is a hydrogen atom or a halogen; in some embodiments, R 9a is a hydrogen atom or F.
  • each R 01, R 02, R 03 and R 04 are the same or different and are each independently selected from oxo, halogen, hydroxyl, cyano, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 haloalkyl, C 1-6 haloalkoxy, C 1-6 hydroxyalkyl and 3 to 6 membered cycloalkyl; preferably, each R 01 , R 02 , R 03 and R 04 are the same or different and are each independently selected from oxo, halogen, hydroxyl, cyano, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 haloalkyl, C 1-6 haloalkoxy, C 1-6 hydroxyalkyl and 3 to 6 membered cycloalkyl; preferably, each R 01 , R 02 , R 03 and R 04 are the same or different and are each independently selected from oxo, halogen, hydroxy
  • each R 03 is the same or different and is independently selected from oxo, halogen, hydroxyl, C 1-6 alkyl and C 1-6 haloalkyl; preferably, each R 03 is the same or different and is independently selected from halogen, hydroxyl and C 1-6 alkyl; more preferably, each R 03 is the same or different and is independently selected from fluorine atom or hydroxyl; in some embodiments, each R 03 is the same or different and is independently selected from halogen, hydroxyl, C 1-6 alkyl and C
  • each RL1 , RL2 , RL3 , RL4 , RL6 and RL7 are the same or different and are each independently a hydrogen atom or a C1-6 alkyl group; in some embodiments, each of RL1 , RL2 , RL3 , RL4 , RL6 and RL7 is a hydrogen atom.
  • the compounds represented by the general formula (I), (II'), (III'), (IV'), (I'-2), (X), (X'), (X'-1), (X'-2), (X'-3) or their pharmaceutically acceptable salts wherein Y1 is CH or N; in some embodiments, Y1 is N; in some embodiments, Y1 is CH.
  • Y 3 is CH or N.
  • the compound represented by the general formula (II) or (II’) or a pharmaceutically acceptable salt thereof wherein v1 is 0 or 1; and v2 is 0 or 1.
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein R 1 is -N(R 15 )SO 2 R 16 ; R 15 and R 16 are the same or different and are independently selected from a hydrogen atom, a C 1-6 alkyl group and a 3 to 6-membered cycloalkyl group; R 2 is a hydrogen atom; R 3 is connected to L 1 ; L 1 is O, L 2 is (CH 2 ) n3 , n3 is 1, 2, 3, 4, 5, 6, 7 or 8; Ring A is n is 0; L 3 is a bond; R 4 is a hydrogen atom; R5 is halogen; Ra and Rb are both hydrogen atoms; Z1 is CRZ1 , RZ1 is a hydrogen atom or a C1-6 alkoxy group; Z2 is CRZ2 , RZ2 is a hydrogen atom or a halogen; Z3 is CH; -J1 - J2 -
  • E is selected from: Y1 is N or CH; Y2 is CH2 or C(O); each Rq is the same or different and is independently selected from halogen, C1-6 alkyl and C1-6 haloalkyl; R6 is a hydrogen atom or a C1-6 alkyl; R7 is a C1-6 alkyl; s is 0, 1, 2 or 3.
  • the compound represented by the general formula (II) or a pharmaceutically acceptable salt thereof wherein R 1 is -N(R 15 )SO 2 R 16 ; R 15 and R 16 are the same or different and are independently a hydrogen atom or a C 1-6 alkyl group; R 2 is a hydrogen atom; R 3 is connected to L 1 ; L 1 is O, and L 2 is a C 1-10 alkylene group; R 4 is a hydrogen atom; R 5 is a halogen; Ra and R b are both hydrogen atoms; R Z1 is a hydrogen atom or a C 1-6 alkoxy group; R Z2 is a hydrogen atom or a halogen; J 2 is a 1,4-piperidinyl or 1,4-piperazinyl group optionally substituted by one or more R 03 , R 03 is selected from hydroxyl, halogen, C 1-6 alkyl and C 1-6 haloalkyl; J 6 is a 1,4-piperid
  • the compound represented by the general formula (II') or a pharmaceutically acceptable salt thereof wherein R 1 is -N(R 15 )SO 2 R 16 ; R 15 and R 16 are the same or different and are independently a hydrogen atom or a C 1-6 alkyl group; R 2 is a hydrogen atom; R 3 is connected to L 1 ; L 1 is O, and L 2 is a C 1-10 alkylene group; R 4 is a hydrogen atom; R 5 is a halogen; Ra and R b are both hydrogen atoms; R Z1 is a hydrogen atom or a C 1-6 alkoxy group; R Z2 is a hydrogen atom or a halogen; J 2 is a 1,4-piperidinyl or 1,4-piperazinyl group optionally substituted by one or more R 03 , and R 03 is selected from hydroxyl, halogen, C 1-6 alkyl and C 1-6 haloalkyl; J 6 is a 1,4-N(R 15 )SO 2 R
  • the compound represented by the general formula (V-1) or a pharmaceutically acceptable salt thereof wherein R 1 is -N(R 15 )SO 2 R 16 ; R 15 and R 16 are the same or different and are independently selected from a hydrogen atom, a C 1-6 alkyl group and a 3- to 6-membered cycloalkyl group; R 2 is a hydrogen atom; R 4 is a hydrogen atom; R 5 is a halogen; Ra and R b are both hydrogen atoms; R Z1 is a hydrogen atom or a C 1-6 alkoxy group; R Z2 is a hydrogen atom or a halogen; L 1 is O, L 2 is (CH 2 ) n3 , n3 is 1, 2, 3, 4, 5, 6, 7 or 8; n is 0; J 1 is selected from a bond, a piperidinyl group and a piperazinyl group, and the piperidinyl group and the piperazinyl group are each independently optional
  • the compound represented by the general formula (V-2) or a pharmaceutically acceptable salt thereof wherein R 1 is -N(R 15 )SO 2 R 16 ; R 15 and R 16 are the same or different and are independently selected from a hydrogen atom, a C 1-6 alkyl group and a 3- to 6-membered cycloalkyl group; R 2 is a hydrogen atom; R 4 is a hydrogen atom; R 5 is a halogen; Ra and R b are both hydrogen atoms; R Z1 is a hydrogen atom or a C 1-6 alkoxy group; R Z2 is a hydrogen atom or a halogen; L 1 is O, L 2 is (CH 2 ) n3 , n3 is 1, 2, 3, 4, 5, 6, 7 or 8; n is 0; J 1 is selected from a bond, a piperidinyl group and a piperazinyl group, and the piperidinyl group and the piperazinyl group are each independently optional
  • R 2 is a hydrogen atom;
  • R 4 is a hydrogen atom;
  • R 5 is a halogen;
  • Ra and R b are both hydrogen atoms;
  • R Z1 is selected from a hydrogen atom, a methoxy group and a deuterated methoxy group;
  • R Z2 is a hydrogen atom or a halogen;
  • L 1 is O, and L 2 is selected from (CH 2 ) 3 , (CH 2 ) 4 and (CH 2 ) 5 ;
  • n is 0;
  • each R q is the same or different and is independently selected from a halogen, a C 1-6 alkyl group and a C 1-6 haloalkyl group;
  • Y 2 is CH 2 or C(O);
  • s is 0, 1 or 2;
  • R 2 is a hydrogen atom;
  • R 4 is a hydrogen atom;
  • R 5 is a halogen;
  • Ra and R b are both hydrogen atoms;
  • R Z1 is selected from a hydrogen atom, a methoxy group and a deuterated methoxy group;
  • R Z2 is a hydrogen atom or a halogen;
  • L 1 is O, and L 2 is selected from (CH 2 ) 3 , (CH 2 ) 4 and (CH 2 ) 5 ;
  • n is 0;
  • J 1 is selected from a bond, a piperidinyl group and a piperazinyl group, and the piperidinyl group and the piperazinyl group are each independently optionally substituted by one or more selected from an oxo group, a halogen group, a hydroxyl group and a C 1-6 alkyl group;
  • q is 1;
  • Ring J 2 ' is selected from a piperidinyl group,
  • the compound represented by the general formula (X') or a pharmaceutically acceptable salt thereof wherein R 1 is -P(O)R 10 R 11 or -N(R 15 )SO 2 R 16 ; R 10 , R 11 , R 15 and R 16 are the same or different and are each independently selected from a hydrogen atom, a methyl group, an ethyl group and a cyclopropyl group; each R 9 is the same or different and is each independently selected from a halogen, a C 1-6 alkyl group and a C 1-6 haloalkyl group; y is 0 or 1; Ra and R b are both hydrogen atoms; R 4 is a hydrogen atom; R 5 is a halogen; R Z1 is selected from a hydrogen atom, a methoxy group and a deuterated methoxy group; R Z2 is a hydrogen atom or a halogen; L 1 is O, and L 2 is selected from (CH 2 )
  • the compound represented by the general formula (X') or a pharmaceutically acceptable salt thereof wherein R 1 is -P(O)R 10 R 11 or -N(R 15 )SO 2 R 16 ; R 10 , R 11 , R 15 and R 16 are the same or different and are each independently selected from a hydrogen atom, a methyl group, an ethyl group and a cyclopropyl group; each R 9 is the same or different and is each independently a halogen; y is 0 or 1; Ra and R b are both hydrogen atoms; R 4 is a hydrogen atom; R 5 is a halogen; R Z1 is selected from a hydrogen atom; R Z2 is a hydrogen atom or a halogen; L 1 is O, L 2 is selected from (CH 2 ) 3 , (CH 2 ) 4 and (CH 2 ) 5 ; n is 0;
  • q, u and w are the same or different and are each independently 0, 1, 2, 3, 4, 5 or 6; each R 03 is the same or different and is each independently selected from oxo, halogen, hydroxyl and C 1-6 alkyl; g and h are each independently 0, 1 or 2; q1 is 0, 1, 2, 3, 4, 5 or 6;
  • E is selected from: Y1 is N or CH; Y2 is CH2 or C(O); each Rq is the same or different and is independently selected from halogen, C1-6 alkyl and C1-6 haloalkyl; R6 is a hydrogen atom, R7 is a C1-6 alkyl, and R8 is a C1-6 alkyl; s is 0, 1, 2 or 3.
  • the compound represented by the general formula (X') or a pharmaceutically acceptable salt thereof wherein R 1 is -P(O)R 10 R 11 or -N(R 15 )SO 2 R 16 ; R 10 , R 11 , R 15 and R 16 are the same or different and are each independently selected from a hydrogen atom, a C 1-6 alkyl group and a 3 to 6 membered cycloalkyl group; each R 9 is the same or different and is each independently halogen; y is 0 or 1; Ra and R b are both hydrogen atoms; R 4 is a hydrogen atom, and R 5 is a halogen or a C 1-6 haloalkyl group; R Z1 is selected from a hydrogen atom, a methoxy group and a deuterated methoxy group; R Z2 is a hydrogen atom or a halogen; -L 1 -L 2 - is -OC 1-10 alkylene-; n is 0;
  • Typical compounds of the present disclosure include, but are not limited to:
  • Another aspect of the present disclosure relates to a compound represented by general formula (Xa) or a salt thereof:
  • R J ′ is selected from the group consisting of a hydrogen atom, a hydroxyl group, a halogen, an alkyl group, an alkoxy group, a haloalkyl group, a haloalkoxy group, C(O)OR aa , -alkylene-C(O)OR aa , NR bb R cc , -alkylene-NR bb R cc , -C(O)R aa , -alkylene-C(O)R aa , S(O)OR aa and S(O) 2 OR aa ;
  • Raa is each independently selected from a hydrogen atom, an alkyl group, a haloalkyl group, a haloalkoxy group, a cycloalkyl group, a heterocyclic group, a heterocyclic alkyl group, a heterocyclic oxy group, an aryl group, and a heteroaryl group;
  • Rbb and Rcc are each independently selected from a hydrogen atom, an alkyl group, a cycloalkyl group, a heterocyclic group, and an amino protecting group, and the amino protecting group is preferably Boc;
  • R1 , R9 , y, R4 , R5 , Ra , Rb , Z1 , Z2 , Z3 , L1 , L2 , Ring A, RA , n, L3 , J1 and J2 are as defined in the general formula (X).
  • Another aspect of the present disclosure relates to a compound represented by general formula (Ia) or a salt thereof:
  • R J ′ is selected from the group consisting of a hydrogen atom, a hydroxyl group, a halogen, an alkyl group, an alkoxy group, a haloalkyl group, a haloalkoxy group, C(O)OR aa , -alkylene-C(O)OR aa , NR bb R cc , -alkylene-NR bb R cc , -C(O)R aa , -alkylene-C(O)R aa , S(O)OR aa and S(O) 2 OR aa ;
  • Raa is each independently selected from a hydrogen atom, an alkyl group, a haloalkyl group, a haloalkoxy group, a cycloalkyl group, a heterocyclic group, a heterocyclic alkyl group, a heterocyclic oxy group, an aryl group, and a heteroaryl group;
  • Rbb and Rcc are each independently selected from a hydrogen atom, an alkyl group, a cycloalkyl group, a heterocyclic group, and an amino protecting group, and the amino protecting group is preferably Boc;
  • Ring A, R1 to R5 , RA , Ra , Rb , Z1 , Z2 , Z3 , L1 , L2 , L3 , J1 , J2 and n are as defined in the general formula (I).
  • the compound represented by the general formula (Xa) or (Ia) or a salt thereof wherein R J ' is selected from the group consisting of hydrogen atom, C(O)OR aa , -C 1-6 alkylene-C(O)OR aa , -C(O)R aa and -C 1-6 alkylene-C(O)R aa , and R aa is a hydrogen atom or a C 1-6 alkyl group; in some embodiments, R J ' is selected from the group consisting of hydrogen atom, C(O)O tert-butyl group, -CH 2 -C(O)O tert-butyl group, -CH 2 -C(O)OH, -C(O)H and -CH 2 -C(O)H.
  • Another aspect of the present disclosure relates to a compound represented by the general formula (I'-3A), (I'-3-1A) or (I'-3-2A) or a salt thereof:
  • RL is selected from -OH, halogen and -O-alkyl
  • R1 to R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 to J6 , Rq , Y2 and s are as defined in the general formula (I'-3), (I'-3-1) or (I'-3-2).
  • Another aspect of the present disclosure relates to a compound represented by the general formula (IIIa) or (IVa) or a salt thereof:
  • R W is a hydrogen atom or an amino protecting group, and the amino protecting group is preferably Boc;
  • Ring J 2 ' is a nitrogen-containing heterocyclic group, and the nitrogen-containing heterocyclic group is optionally substituted by one or more R 03 ;
  • R1 , R2 , R4 , R5 , RA , Ra , Rb , RZ1 , RZ2 , L1 , L2 , J1 and J2 are as defined in the general formula (I).
  • Another aspect of the present disclosure relates to a compound represented by the general formula (V-1A) or a salt thereof:
  • R W is a hydrogen atom or an amino protecting group, and the amino protecting group is preferably Boc;
  • Ring J 2 ' is a nitrogen-containing heterocyclic group
  • j 0, 1, 2, 3, 4, 5, or 6;
  • R1 , R2 , R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 and R03 are as defined in the general formula (V-1).
  • Another aspect of the present disclosure relates to a compound represented by the general formula (V-2A) or a salt thereof:
  • q-1 is 0, 1, 2, 3, 4 or 5;
  • R 1 , R 2 , R 4 , R 5 , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , RA , n and J 1 are as defined in the general formula (V-2).
  • V-1A a compound represented by the general formula (V-1A), (V-1-1A) or (V-1-2A) or a salt thereof:
  • RL is selected from -OH, halogen and -O-alkyl
  • R1 , R2 , R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , ring J2 ', R03 , j, q, J4 , J5 , J6 , Rq , Y2 and s are as defined in the general formula (V-1), (V-1-1) or (V-1-2).
  • V-2A a compound represented by the general formula (V-2A), (V-2-1A) or (V-2-2A) or a salt thereof:
  • RL is selected from -OH, halogen and -O-alkyl
  • R1 , R2 , R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , q, ring J3 ', R03 , j, J4 , J5 , J6 , Rq , Y2 and s are as defined in the general formula (V-2), (V-2-1) or (V-2-2).
  • Another aspect of the present disclosure relates to a compound represented by the general formula (X'-1a) or (X'-2a) or a salt thereof:
  • ring J 2 ' is a nitrogen-containing heterocyclic group; j is 0, 1, 2, 3, 4, 5 or 6;
  • R1 , R9 , y, R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , J1 and R03 are as defined in the general formula (X).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by the above general formula (I'-3-1) or (I'-3-2) or a pharmaceutically acceptable salt thereof, the method comprising:
  • the compound represented by the general formula (I'-3-1A) or a salt thereof undergoes an intramolecular ring-closing reaction to obtain a compound represented by the general formula (I'-3-1) or a pharmaceutically acceptable salt thereof;
  • the compound represented by the general formula (I'-3-2A) or a salt thereof undergoes an intramolecular ring-closing reaction to obtain a compound represented by the general formula (I'-3-2) or a pharmaceutically acceptable salt thereof;
  • RL is selected from -OH, halogen and -O-alkyl
  • R1 to R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 to J6 , Rq , Y2 and s are as defined in the general formula (I'-3-1) or (I'-3-2).
  • Another aspect of the present disclosure relates to a method for preparing the compounds represented by the above general formulae (I'-3-1) and (I'-3-2) or pharmaceutically acceptable salts thereof, the method comprising:
  • R1 to R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 to J6 , Rq , Y2 and s are as defined in the general formula (I'-3-1) or (I'-3-2).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by the above general formula (III) or (III') or a pharmaceutically acceptable salt thereof, the method comprising:
  • the compound represented by the general formula (IIIa) or its salt (preferably hydrochloride) and the compound represented by the general formula (III'b) or its salt (preferably hydrochloride) undergo condensation reaction to obtain the compound represented by the general formula (III') or its pharmaceutically acceptable salt;
  • R W is a hydrogen atom
  • Ring J 2 ′, R 1 , R 2 , R 4 to R 7 , RA , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , J 1 , J 5 , J 6 , R q , s and n are as defined in the general formula (III), and Y 1 is as defined in the general formula (III′).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by the above general formula (IV) or (IV') or a pharmaceutically acceptable salt thereof, the method comprising:
  • the compound represented by the general formula (IVa) or its salt (preferably hydrochloride) and the compound represented by the general formula (IVb) or its salt (preferably hydrochloride) undergo condensation reaction to obtain the compound represented by the general formula (IV) or its pharmaceutically acceptable salt;
  • the compound represented by the general formula (IVa) or its salt (preferably hydrochloride) and the compound represented by the general formula (IV'b) or its salt (preferably hydrochloride) undergo condensation reaction to obtain the compound represented by the general formula (IV') or its pharmaceutically acceptable salt;
  • Another aspect of the present disclosure relates to a method for preparing the compound represented by the above general formula (V-1) or a pharmaceutically acceptable salt thereof, the method comprising:
  • the compound represented by the general formula (V-1A) or its salt undergoes a reductive amination reaction with the compound represented by the general formula (V-1B) or its salt to obtain the compound represented by the general formula (V-1) or its pharmaceutically acceptable salt;
  • R W is a hydrogen atom
  • R1 , R2 , R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , ring J2 ', R03 , j, q, J4 , J5 , J6 , Rq , Y2 and s are as defined in the general formula (V-1).
  • Another aspect of the present disclosure relates to a method for preparing the compound represented by the above general formula (V-2) or a pharmaceutically acceptable salt thereof, the method comprising:
  • the compound represented by the general formula (V-2A) or its salt undergoes a reductive amination reaction with the compound represented by the general formula (V-2B) or its salt (preferably hydrochloride) to obtain the compound represented by the general formula (V-2) or its pharmaceutically acceptable salt;
  • R 1 , R 2 , R 4 , R 5 , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , RA , n, J 1 , q, ring J 3 ′, R 03 , j, J 4 , J 5 , J 6 , R q , Y 2 and s are as defined in the general formula (V-2).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by the above general formula (V-1-1) or (V-1-2) or a pharmaceutically acceptable salt thereof, the method comprising:
  • the compound represented by the general formula (V-1-1A) or a salt thereof undergoes an intramolecular ring-closing reaction to obtain a compound represented by the general formula (V-1-1) or a pharmaceutically acceptable salt thereof;
  • the compound represented by the general formula (V-1-2A) or a salt thereof undergoes an intramolecular ring-closing reaction to obtain a compound represented by the general formula (V-1-2) or a pharmaceutically acceptable salt thereof;
  • RL is selected from -OH, halogen and -O-alkyl
  • R1 , R2 , R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , ring J2 ', R03 , j, q, J4 , J5 , J6 , Rq , Y2 and s are as defined in the general formula (V-1-1) or (V-1-2).
  • Another aspect of the present disclosure relates to a method for preparing the compounds represented by the above general formulae (V-1-1) and (V-1-2) or pharmaceutically acceptable salts thereof, the method comprising:
  • R 1 , R 2 , R 4 , R 5 , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , RA , n, J 1 , ring J 2 ', R 03 , j, q, J 4 , J 5 , J 6 , R q , Y 2 and s are as defined in the general formula (V-1-1) or (V-1-2).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by the above general formula (V-2-1) or (V-2-2) or a pharmaceutically acceptable salt thereof, the method comprising:
  • the compound represented by the general formula (V-2-1A) or a salt thereof undergoes an intramolecular ring-closing reaction to obtain a compound represented by the general formula (V-2-1) or a pharmaceutically acceptable salt thereof;
  • the compound represented by the general formula (V-2-2A) or a salt thereof undergoes an intramolecular ring-closing reaction to obtain a compound represented by the general formula (V-2-2) or a pharmaceutically acceptable salt thereof;
  • RL is selected from -OH, halogen and -O-alkyl
  • R1 , R2 , R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , q, ring J3 ', R03 , j, J4 , J5 , J6 , Rq , Y2 and s are as defined in the general formula (V-2-1) or (V-2-2).
  • Another aspect of the present disclosure relates to a method for preparing the compounds represented by the above general formulae (V-2-1) and (V-2-2) or pharmaceutically acceptable salts thereof, the method comprising:
  • R 1 , R 2 , R 4 , R 5 , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , RA , n, J 1 , q, ring J 3 ′, R 03 , j, J 4 , J 5 , J 6 , R q , Y 2 and s are as defined in the general formula (V-2-1) or (V-2-2).
  • Another aspect of the present disclosure relates to a method for preparing the compound represented by the above general formula (X'-1) or a pharmaceutically acceptable salt thereof, the method comprising:
  • the compound represented by the general formula (X'-1a) or its salt undergoes a reductive amination reaction with the compound represented by the general formula (X'-1b) or its salt to obtain the compound represented by the general formula (X'-1) or its pharmaceutically acceptable salt;
  • q is 1, 2, 3, 4, 5 or 6; q-1 is 0, 1, 2, 3, 4 or 5;
  • R1 , R9 , y, R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , Ring J2 ', R03 , j, J4 , J5 , J6 and E are as defined in the general formula (X'-1).
  • Another aspect of the present disclosure relates to a method for preparing the compound represented by the above general formula (X'-2) or a pharmaceutically acceptable salt thereof, the method comprising:
  • q is 1, 2, 3, 4, 5 or 6; q-1 is 0, 1, 2, 3, 4 or 5;
  • R1 , R9 , y, R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , Ring J3 ', R03 , j, J4 , J5 , J6 and E are as defined in the general formula (X'-2).
  • Another aspect of the present disclosure relates to a method for preparing the compound represented by the above general formula (X'-3) or a pharmaceutically acceptable salt thereof, the method comprising:
  • q is 1, 2, 3, 4, 5 or 6; q-1 is 0, 1, 2, 3, 4 or 5;
  • R1 , R9 , y, R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , Ring J3 ', R03 , j, J4 , J5 , J6 , Y1 , R7 , Rq and s are as defined in the general formula (X'-3).
  • the method for preparing the compounds represented by the general formula (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2) or their pharmaceutically acceptable salts wherein RL is -OC 1-6 alkyl; in some embodiments, RL is tert-butoxy.
  • the method for preparing the compound represented by the general formula (V-1-1), (V-1-2) or a pharmaceutically acceptable salt thereof wherein when the preparation method adopts a reductive amination reaction, q-1 is 0, 1, 2, 3, 4 or 5, and q is 1, 2, 3, 4, 5 or 6; in some embodiments, q-1 is 0, and q is 1.
  • compositions which contains compounds of the general formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) and shown in Table A, or pharmaceutically acceptable salts thereof, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the present disclosure further relates to the use of compounds or pharmaceutically acceptable salts thereof as shown in formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) and Table A, or pharmaceutical compositions containing the same, in the preparation of drugs for regulating EGFR ubiquitination and degradation.
  • the present disclosure further relates to the use of compounds or pharmaceutically acceptable salts thereof as shown in formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) and Table A, or pharmaceutical compositions comprising the same, in the preparation of medicaments for treating and/or preventing diseases or conditions mediated by or dependent on EGFR.
  • the present disclosure further relates to the use of compounds or pharmaceutically acceptable salts thereof as shown in formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) and Table A in the preparation of EGFR inhibitors and/or degraders, or pharmaceutical compositions comprising the same.
  • the present disclosure further relates to compounds of formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) and compounds shown in Table A or pharmaceutically acceptable salts thereof, or pharmaceutical compositions containing the same in the preparation of Use in a drug for treating and/or preventing cancer; preferably, the cancer is selected from squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, liver cancer, kidney cancer, bladder cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, head and neck cancer, nasopharyngeal cancer, oral cancer, salivary gland cancer, kidney cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, gastric cancer, leukemia,
  • the present disclosure further relates to compounds of formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) and compounds shown in Table A or pharmaceutically acceptable salts thereof, or pharmaceutical compositions comprising the same in the preparation of a pharmaceutical composition for treating and/or preventing a disease
  • the invention relates to a method for treating a disease or condition; preferably, the disease or condition is cancer or a tumor; preferably, the cancer is selected from squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, liver cancer, kidney cancer, bladder cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, head and neck cancer, nasopharyngeal cancer, oral cancer,
  • the present disclosure also relates to a method for regulating the ubiquitination and degradation of EGFR protein in a subject, which comprises administering to a patient in need thereof a therapeutically effective amount of the above-mentioned general formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) or a compound shown in Table A or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
  • the present disclosure also relates to a method for treating and/or preventing EGFR-mediated or dependent diseases or conditions, which comprises administering to a patient in need thereof a therapeutically effective amount of the above-mentioned general formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) or a compound shown in Table A or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
  • the present disclosure also relates to a method for inhibiting and/or degrading EGFR protein, which comprises administering to a patient in need thereof a therapeutically effective amount of the compounds of the above-mentioned general formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) or shown in Table A or their pharmaceutically acceptable salts, or a pharmaceutical composition comprising the same.
  • a method for inhibiting and/or degrading EGFR protein which comprises administering to a patient in need thereof a therapeutically effective amount of the compounds of the above-mentioned general formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1
  • the present disclosure also relates to a method for treating and/or preventing cancer, comprising administering to a patient in need thereof a therapeutically effective amount of the above-mentioned general formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) or the compounds shown in Table A a compound or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same; preferably, the cancer is selected from squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, liver cancer, kidney cancer, bladder cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, head and neck cancer, nasopharyngeal cancer, oral cancer, salivary gland cancer, kidney cancer
  • the present disclosure further relates to a compound or a pharmaceutically acceptable salt thereof as shown in the above-mentioned general formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) or Table A, or a pharmaceutical composition comprising the same, which is used as a drug.
  • the present disclosure further relates to a compound or a pharmaceutically acceptable salt thereof as shown in the above-mentioned general formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) or Table A, or a pharmaceutical composition comprising the same, which is used as a drug for regulating the ubiquitination and degradation of EGFR protein in a subject.
  • the present disclosure further relates to a compound or a pharmaceutically acceptable salt thereof as shown in the above-mentioned general formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) or Table A, which is used as an EGFR inhibitor and/or degrader.
  • the present disclosure further relates to a compound or a pharmaceutically acceptable salt thereof as shown in the above-mentioned general formula (I) to (IV), (II’) to (IV’), (I’-1), (I’-2), (I’-3), (V-1), (V-2), (I’-3-1), (I’-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X’), (X’-1), (X’-2), (X’-3) or Table A, or a pharmaceutical composition comprising the same, which is used as a drug for inhibiting EGFR activity and/or degrading EGFR protein.
  • the present disclosure further relates to a compound or a pharmaceutically acceptable salt thereof as shown in the above-mentioned general formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) or Table A, or a pharmaceutical composition comprising the same, which is used as a drug for treating and/or preventing diseases or conditions mediated by or dependent on EGFR.
  • the present disclosure further relates to compounds or pharmaceutically acceptable salts thereof as shown in the above-mentioned general formulas (I) to (IV), (II’) to (IV’), (I’-1), (I’-2), (I’-3), (V-1), (V-2), (I’-3-1), (I’-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X’), (X’-1), (X’-2), (X’-3) or Table A, for use in regulating ubiquitination and degradation of EGFR protein in a subject.
  • the present disclosure further relates to the above-mentioned general formulas (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) or a compound shown in Table A or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for use in inhibiting EGFR activity and/or degrading EGFR protein.
  • the present disclosure further relates to compounds or pharmaceutically acceptable salts thereof as shown in the above-mentioned general formula (I) to (IV), (II’) to (IV’), (I’-1), (I’-2), (I’-3), (V-1), (V-2), (I’-3-1), (I’-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X’), (X’-1), (X’-2), (X’-3) or Table A, for treating and/or preventing diseases or conditions mediated by or dependent on EGFR.
  • the present disclosure further relates to a compound of the above-mentioned general formula (I) to (IV), (II') to (IV'), (I'-1), (I'-2), (I'-3), (V-1), (V-2), (I'-3-1), (I'-3-2), (V-1-1), (V-1-2), (V-2-1), (V-2-2), (X), (X'), (X'-1), (X'-2), (X'-3) or a compound shown in Table A or a pharmaceutically acceptable salt thereof, or a drug containing the same A composition for treating and/or preventing cancer; preferably, the cancer is selected from squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, liver cancer, kidney cancer, bladder cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, head and neck cancer, nasopharyngeal cancer, oral cancer, salivary gland cancer, kidney cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, gastric
  • the EGFR-mediated or dependent disease or condition described in the present disclosure is cancer; the disease or condition is preferably selected from squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, liver cancer, kidney cancer, bladder cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, head and neck cancer, nasopharyngeal cancer, oral cancer, salivary gland cancer, kidney cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, gastric cancer, leukemia, lymphoma, glioma, neuroblastoma, melanoma, sarcoma, endometrial cancer, testicular cancer and thyroid cancer; further preferably lung cancer; more preferably non-small cell lung cancer.
  • Lung cancer in the present disclosure includes small cell lung cancer and non-small cell lung cancer; preferably non-small cell lung cancer.
  • the cancer described in the present disclosure preferably has an EGFR protein with L858R mutation.
  • the cancer described in the present disclosure is preferably an EGFR protein with a 19del mutation.
  • the cancer described in the present disclosure is preferably an EGFR protein with T790M mutation.
  • the cancer described in the present disclosure preferably has an EGFR protein with a C797X mutation.
  • the cancer described in the present disclosure preferably has EGFR protein with L858R and T790M mutations.
  • the cancer described in the present disclosure preferably has EGFR protein with 19del and T790M mutations.
  • the cancer described in the present disclosure preferably has EGFR protein with L858R and C797X mutations.
  • the cancer described in the present disclosure preferably has EGFR protein with 19del and C797X mutations.
  • the cancer described in the present disclosure preferably has EGFR protein with T790M and C797X mutations.
  • the cancer described in the present disclosure preferably has EGFR protein with L858R, T790M and C797X mutations.
  • the cancer described in the present disclosure preferably has EGFR protein with 19del, T790M and C797X mutations.
  • the C797X mutation described in the present disclosure is preferably a C797S mutation; wherein X represents any amino acid including S.
  • the active compound may be formulated for administration by any appropriate route, preferably in a unit dosage form, or in a form in which a patient can self-administer a single dose.
  • the unit dosage form of the disclosed compound or composition may be tablets, capsules, cachets, bottled liquids, powders, granules, lozenges, suppositories, reconstituted powders or liquid preparations.
  • suitable unit doses may be in the range of 0.1 to 1000 mg.
  • the pharmaceutical composition of the present disclosure may contain one or more excipients in addition to the active compound, and the excipients are selected from the following ingredients: fillers (diluents), binders, wetting agents, disintegrants or excipients, etc.
  • the composition may contain 0.1 to 99% by weight of the active compound.
  • the unit dose of the pharmaceutical composition is 0.001 mg-1000 mg.
  • the pharmaceutical composition contains 0.01-99.99% of the aforementioned compound or its pharmaceutically acceptable salt or its isotope substitution, based on the total weight of the composition. In certain embodiments, the pharmaceutical composition contains 0.1-99.9% of the aforementioned compound or its pharmaceutically acceptable salt or its isotope substitution. In certain embodiments, the pharmaceutical composition contains 0.5%-99.5% of the aforementioned compound or its pharmaceutically acceptable salt or its isotope substitution. In certain embodiments, the pharmaceutical composition contains 1%-99% of the aforementioned compound or its pharmaceutically acceptable salt or its isotope substitution. In certain embodiments, the pharmaceutical composition contains 2%-98% of the aforementioned compound or its pharmaceutically acceptable salt or its isotope substitution.
  • the pharmaceutical composition contains 0.01%-99.99% of a pharmaceutically acceptable excipient based on the total weight of the composition. In certain embodiments, the pharmaceutical composition contains 0.1%-99.9% of a pharmaceutically acceptable excipient. In certain embodiments, the pharmaceutical composition contains 0.5%-99.5% of a pharmaceutically acceptable excipient. In certain embodiments, the pharmaceutical composition contains 1%-99% of a pharmaceutically acceptable excipient. In certain embodiments, the pharmaceutical composition contains 2%-98% of a pharmaceutically acceptable excipient.
  • the pharmaceutical composition containing the active ingredient can be in a form suitable for oral administration, such as tablets, lozenges, pastilles, water or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Oral compositions can be prepared according to any method known in the art for preparing pharmaceutical compositions, and such compositions can contain one or more ingredients selected from the following: sweeteners, flavoring agents, coloring agents and preservatives to provide pleasing and palatable pharmaceutical preparations.
  • Tablets contain active ingredients and non-toxic pharmaceutically acceptable excipients suitable for preparing tablets for mixing. These excipients can be inert excipients, granulating agents, disintegrants, binders and lubricants. These tablets can be uncoated or can be coated by known techniques that mask the taste of the drug or delay disintegration and absorption in the gastrointestinal tract, thereby providing a sustained release effect over a long period of time.
  • Oral preparations may also be provided in soft gelatin capsules wherein the active ingredient is mixed with an inert solid diluent or wherein the active ingredient is mixed with a water-soluble carrier or an oily vehicle.
  • Aqueous suspensions contain the active substance and excipients suitable for preparing aqueous suspensions for mixing. Such excipients are suspending agents, dispersants or wetting agents. Aqueous suspensions may also contain one or more preservatives, one or more coloring agents, one or more flavoring agents and one or more sweetening agents.
  • Oil suspensions can be prepared by suspending the active ingredient in a vegetable oil or a mineral oil.
  • the oil suspension may contain a thickener.
  • the above-mentioned sweeteners and flavoring agents may be added to provide a palatable preparation. These compositions may be preserved by adding an antioxidant.
  • the pharmaceutical composition of the present disclosure may also be in the form of an oil-in-water emulsion.
  • the oil phase may be a vegetable oil, a mineral oil or a mixture thereof.
  • a suitable emulsifier may be a naturally occurring phospholipid, and the emulsion may also contain a sweetener, a flavoring agent, a preservative and an antioxidant.
  • Such preparations may also contain a demulcent, a preservative, a coloring agent and an antioxidant.
  • the pharmaceutical composition of the present disclosure may be in the form of a sterile injectable aqueous solution.
  • Acceptable solvents or solvents that may be used are water, Ringer's solution, and isotonic sodium chloride solution.
  • the sterile injectable preparation may be a sterile injectable water-in-oil microemulsion in which the active ingredient is dissolved in the oil phase, and the injectable solution or microemulsion may be injected into the patient's bloodstream by local mass injection.
  • a continuous intravenous drug delivery device may be used.
  • An example of such a device is the Deltec CADD-PLUS.TM.5400 intravenous injection pump.
  • compositions of the present disclosure can be in the form of sterile injection water or oil suspension for intramuscular and subcutaneous administration.
  • the suspension can be prepared with suitable dispersants or wetting agents and suspending agents according to known techniques.
  • Sterile injection preparations can also be sterile injection solutions or suspensions prepared in parenteral acceptable nontoxic diluents or solvents.
  • sterile fixed oils can be conveniently used as solvents or suspension media. For this purpose, any blended fixed oils can be used.
  • fatty acids can also be used to prepare injections.
  • the disclosed compounds may be administered in the form of suppositories for rectal administration.
  • These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid in the rectum and will therefore melt in the rectum to release the drug.
  • the dosage of a drug depends on a variety of factors, including but not limited to the following factors: the activity of the specific compound used, the age of the patient, the weight of the patient, the health status of the patient, the behavior of the patient, the diet of the patient, the time of administration, the mode of administration, the rate of excretion, the combination of drugs, the severity of the disease, etc.; in addition, the best treatment method such as the mode of treatment, the daily dosage of the compound or the type of pharmaceutically acceptable salt can be verified according to traditional treatment regimens.
  • alkyl refers to a saturated straight or branched aliphatic hydrocarbon group having 1 to 20 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) carbon atoms (i.e., C1-20 alkyl).
  • the alkyl group is preferably an alkyl group having 1 to 12 carbon atoms (i.e., C1-10 alkyl), and preferably an alkyl group having 1 to 6 carbon atoms (i.e., C1-6 alkyl).
  • Non-limiting examples include: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2- Dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5-methylhexyl,
  • the alkyl group may be substituted or unsubstituted, and when substituted, it may be substituted at any available attachment point, and the substituent is preferably selected from one or more of D atoms, halogen, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • alkylene refers to a divalent alkyl group, wherein the alkyl group is as defined above, and has 1 to 20 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) carbon atoms (i.e., C1-20 alkylene).
  • the alkylene group is preferably an alkylene group having 1 to 10 carbon atoms (i.e., C1-10 alkylene), preferably an alkylene group having 1 to 8 carbon atoms (i.e., C1-8 alkylene), more preferably an alkylene group having 2 to 7 carbon atoms (i.e., C2-7 alkylene) or an alkylene group having 1 to 6 carbon atoms (i.e., C1-6 alkylene).
  • Non-limiting examples include: -CH2- , -CH( CH3 )-, -C( CH3 ) 2- , -CH2CH2-, -CH( CH2CH3 )-, -CH2CH ( CH3 ) - , -CH2C ( CH3 ) 2- , -CH2CH2CH2- , -CH2CH2CH2- , -CH2CH2CH2CH2- , etc.
  • the alkylene group may be substituted or unsubstituted, and when substituted, it may be substituted at any available point of attachment, and the substituents are preferably selected from one or more of D atoms, halogen, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • C 0-6 alkylene refers to a divalent alkyl group having 0 to 6 (e.g., 0, 1, 2, 3, 4, 5 or 6) carbon atoms, C 0 being a bond; thus, C 0-6 alkylene includes a bond and C 1-6 alkylene; C 1-6 alkylene is as defined above.
  • alkenyl refers to an alkyl group containing at least one carbon-carbon double bond in the molecule, wherein the definition of alkyl is as described above, and it has 2 to 12 (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12) carbon atoms (i.e., C2-12 alkenyl).
  • the alkenyl group preferably has an alkenyl group of 2 to 6 carbon atoms (i.e., C2-6 alkenyl).
  • Non-limiting examples include: vinyl, propenyl, isopropenyl, butenyl, etc.
  • the alkenyl group can be substituted or unsubstituted, and when substituted, it can be substituted at any available point of attachment, and the substituent is preferably selected from one or more of D atoms, alkoxy, halogen, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • alkynyl refers to an alkyl group containing at least one carbon-carbon triple bond in the molecule, wherein alkyl is as defined above and has 2 to 12 (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12) carbon atoms.
  • the alkynyl group is preferably an alkynyl group having 2 to 6 carbon atoms (i.e., C 2-12 alkynyl).
  • the alkynyl group preferably has an alkynyl group having 2 to 6 carbon atoms (i.e., C 2-6 alkynyl).
  • Non-limiting examples include: ethynyl, propynyl, butynyl, pentynyl, hexynyl, etc.
  • the alkynyl group may be substituted or unsubstituted, and when substituted, it may be substituted at any available point of attachment, and the substituent group is preferably selected from one or more of a D atom, an alkoxy group, a halogen, a haloalkyl group, a haloalkoxy group, a cycloalkyloxy group, a heterocyclyloxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, a heterocyclyl group, an aryl group, and a heteroaryl group.
  • alkoxy refers to -O-(alkyl), wherein alkyl is as defined above. Non-limiting examples include: methoxy, ethoxy, propoxy and butoxy, etc. Alkoxy can be substituted or unsubstituted, and when substituted, it can be substituted at any usable point of attachment, and the substituent is preferably selected from one or more of D atoms, halogen, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic all-carbon ring (i.e., monocyclic cycloalkyl) or polycyclic ring system (i.e., polycyclic cycloalkyl) having 3 to 20 (e.g., 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) ring atoms (i.e., 3 to 20-membered cycloalkyl).
  • the cycloalkyl is preferably a cycloalkyl having 3 to 12 ring atoms (i.e., 3 to 12-membered cycloalkyl) or a cycloalkyl having 3 to 10 ring atoms (i.e., 3 to 10-membered cycloalkyl), more preferably a cycloalkyl having 3 to 8 ring atoms (i.e., 3 to 8-membered cycloalkyl), most preferably a cycloalkyl having 3 to 6 ring atoms (i.e., 3 to 6-membered cycloalkyl), a cycloalkyl having 4 to 7 ring atoms (i.e., 4 to 7-membered cycloalkyl) or a cycloalkyl having 5 or 6 ring atoms (i.e., 5 or 6-membered cycloalkyl); most preferably a cycloalkyl having 5 or 6 ring
  • Non-limiting examples of the monocyclic cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl and cyclooctyl.
  • the polycyclic cycloalkyl group includes: spirocycloalkyl group, fused cycloalkyl group and bridged cycloalkyl group.
  • spirocycloalkyl refers to a polycyclic system in which one carbon atom (called spiro atom) is shared between the rings, and the rings may contain one or more double bonds, or the rings may contain one or more heteroatoms selected from nitrogen, oxygen and sulfur (the nitrogen may be optionally oxidized, i.e., to form nitrogen oxides; the sulfur may be optionally oxidized, i.e., to form sulfoxides or sulfones, but not including -OO-, -OS- or -SS-), provided that at least one all-carbon ring is contained and the point of attachment is on the all-carbon ring, and it has 5 to 20 (e.g., 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) ring atoms (i.e., 5 to 20-membered spirocycloalkyl).
  • nitrogen may be optionally oxidized, i.e., to form nitrogen oxides
  • sulfur may be optionally
  • the spirocycloalkyl preferably has 6 to 14 ring atoms (i.e., 6 to 14-membered spirocycloalkyl), and more preferably has 7 to 10 ring atoms (i.e., 7 to 10-membered spirocycloalkyl).
  • the spirocycloalkyl includes monospirocycloalkyl and polyspirocycloalkyl (such as bispirocycloalkyl, etc.), preferably monospirocycloalkyl or bispirocycloalkyl, more preferably 3-yuan/4-yuan, 3-yuan/5-yuan, 3-yuan/6-yuan, 4-yuan/4-yuan, 4-yuan/5-yuan, 4-yuan/6-yuan, 5-yuan/3-yuan, 5-yuan/4-yuan, 5-yuan/5-yuan, 5-yuan/6-yuan, 5-yuan/7-yuan, 6-yuan/3-yuan, 6-yuan/4-yuan, 6-yuan/5-yuan, 6-yuan/6-yuan, 6-yuan/7-yuan, 7-yuan/5-yuan or 7-yuan/6-yuan monospiro
  • connection point can be at any position
  • fused cycloalkyl refers to a polycyclic system in which two adjacent carbon atoms are shared between the rings, which is a monocyclic cycloalkyl fused to one or more monocyclic cycloalkyls, or a monocyclic cycloalkyl fused to one or more heterocyclyls, aryls or heteroaryls, wherein the point of attachment is on the monocyclic cycloalkyl, which may contain one or more double bonds within the ring, and has 5 to 20 (e.g., 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) ring atoms (i.e., 5 to 20-membered fused cycloalkyl).
  • the fused cycloalkyl preferably has 6 to 14 ring atoms (i.e., 6 to 14-membered fused cycloalkyl), and more preferably has 7 to 10 ring atoms (i.e., 7 to 10-membered fused cycloalkyl).
  • the condensed cycloalkyl includes bicyclic condensed cycloalkyl and polycyclic condensed cycloalkyl (such as tricyclic condensed cycloalkyl, tetracyclic condensed cycloalkyl, etc.), preferably bicyclic condensed cycloalkyl or tricyclic condensed cycloalkyl, more preferably 3 yuan/4 yuan, 3 yuan/5 yuan, 3 yuan/6 yuan, 4 yuan/4 yuan, 4 yuan/5 yuan, 4 yuan/6 yuan, 5 yuan/3 yuan, 5 yuan/4 yuan, 5 yuan/5 yuan, 5 yuan/6 yuan, 5 yuan/7 yuan, 6 yuan/3 yuan, 6 yuan/4 yuan, 6 yuan/5 yu
  • connection point can be at any position
  • bridged cycloalkyl refers to a full-carbon polycyclic system that shares two carbon atoms that are not directly connected between the rings, which may contain one or more double bonds in the ring and has 5 to 20 (e.g., 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) carbon atoms (i.e., 5 to 20-membered bridged cycloalkyl).
  • the bridged cycloalkyl preferably has a bridged cycloalkyl of 6 to 14 carbon atoms (i.e., 6 to 14-membered bridged cycloalkyl), and more preferably has a bridged cycloalkyl of 7 to 10 carbon atoms (i.e., 7 to 10-membered bridged cycloalkyl).
  • the bridged cycloalkyl includes bicyclic bridged cycloalkyl and polycyclic bridged cycloalkyl (e.g., tricyclic bridged cycloalkyl, tetracyclic bridged cycloalkyl, etc.), preferably bicyclic bridged cycloalkyl or tricyclic bridged cycloalkyl.
  • Non-limiting examples include:
  • connection point can be at any position.
  • the cycloalkyl group may be substituted or unsubstituted. When substituted, it may be substituted at any available point of attachment, and the substituents are preferably selected from one or more of D atoms, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxyl, hydroxyalkyl, oxo, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic heterocycle (i.e., a monocyclic heterocyclyl) or a polycyclic heterocyclic ring system (i.e., a polycyclic heterocyclyl), which contains at least one (e.g., 1, 2, 3 or 4) heteroatoms selected from nitrogen, oxygen and sulfur (the nitrogen may be optionally oxidized, i.e., to form nitrogen oxides; the sulfur may be optionally oxidized, i.e., to form sulfoxides or sulfones, but does not include -O-O-, -O-S- or -S-S-), and has 3 to 20 (e.g., 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) ring atoms (i.e., a 3- to 20-membered heterocyclyl).
  • the heterocyclic group is preferably a heterocyclic group having 3 to 12 ring atoms (i.e., a 3- to 12-membered heterocyclic group) or a heterocyclic group having 3 to 10 ring atoms (i.e., a 3- to 10-membered heterocyclic group); further preferably, a heterocyclic group having 3 to 8 ring atoms (i.e., a 3- to 8-membered heterocyclic group); more preferably, a heterocyclic group having 3 to 6 ring atoms (i.e., a 3- to 6-membered heterocyclic group), a heterocyclic group having 4 to 7 ring atoms (i.e., a 4- to 7-membered heterocyclic group) or a heterocyclic group having 5 or 6 ring atoms (i.e., a 5- or 6-membered heterocyclic group); and most preferably, a heterocyclic group having 5 or 6 ring atoms.
  • Non-limiting examples of the monocyclic heterocyclic group include pyrrolidinyl, tetrahydropyranyl, 1,2,3,6-tetrahydropyridinyl, piperidinyl, piperazinyl, azetidinyl, morpholinyl, thiomorpholinyl and homopiperazinyl.
  • the polycyclic heterocyclic group includes a spiro heterocyclic group, a fused heterocyclic group and a bridged heterocyclic group.
  • spiroheterocyclyl refers to a polycyclic heterocyclic ring system in which one atom (called a spiro atom) is shared between the rings, and the rings may contain one or more double bonds, and the rings may contain at least one (e.g., 1, 2, 3, or 4) heteroatoms selected from nitrogen, oxygen, and sulfur (the nitrogen may be optionally oxidized, i.e., to form nitrogen oxides; the sulfur may be optionally oxidized, i.e., to form sulfoxides or sulfones, but not including -OO-, -OS-, or -SS-), provided that at least one monocyclic heterocyclic group is contained and the point of attachment is on the monocyclic heterocyclic group, and it has 5 to 20 (e.g., 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20) ring atoms (i.e., a 5- to 20-membered spiroheterocyclyl).
  • the spiroheterocyclyl is preferably a spiroheterocyclyl having 6 to 14 ring atoms (i.e., a 6- to 14-membered spiroheterocyclyl), and more preferably a spiroheterocyclyl having 7 to 11 ring atoms (i.e., a 7- to 11-membered spiroheterocyclyl).
  • the spiro heterocyclic group includes a monospiro heterocyclic group and a polyspiro heterocyclic group (such as a bispiro heterocyclic group, etc.), preferably a monospiro heterocyclic group or a bispiro heterocyclic group, more preferably a 3-membered/4-membered, 3-membered/5-membered, 3-membered/6-membered, 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/3-membered, 5-membered/4-membered, 5-membered/5-membered, 5-membered/6-membered, 5-membered/7-membered, 6-membered/3-membered, 6-membered/4-membered 1-, 6-, 5-, 6-, 6-, 6-, 6-, 6-, 6-, 6-, 7-, 7-, 5-, or 7-membered monospiro heterocyclic group.
  • Non-limiting examples include
  • fused heterocyclyl refers to a polycyclic heterocyclic ring system which shares two adjacent atoms between the rings, which may contain one or more double bonds within the ring, and which contains at least one (e.g., 1, 2, 3 or 4) heteroatoms selected from nitrogen, oxygen and sulfur (the nitrogen may be optionally oxidized, i.e., to form nitrogen oxides; the sulfur may be optionally oxidized, i.e., to form sulfoxides or sulfones, but does not include -OO-, -OS- or -SS-), which is a monocyclic heterocyclyl fused to one or more monocyclic heterocyclyls, or a monocyclic heterocyclyl fused to one or more of cycloalkyl, aryl or heteroaryl, wherein the point of attachment is on the monocyclic heterocyclyl, and has 5 to 20 (e.g., 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) ring
  • the fused heterocyclic group preferably has a fused heterocyclic group of 6 to 14 ring atoms (i.e., a 6 to 14-membered fused heterocyclic group), and more preferably has a fused heterocyclic group of 7 to 10 ring atoms (i.e., a 7 to 10-membered fused heterocyclic group).
  • the fused heterocyclic group includes bicyclic and polycyclic fused heterocyclic groups (such as tricyclic fused heterocyclic groups, tetracyclic fused heterocyclic groups, etc.), preferably a bicyclic fused heterocyclic group or a tricyclic fused heterocyclic group, more preferably a 3-yuan/4-yuan, 3-yuan/5-yuan, 3-yuan/6-yuan, 4-yuan/4-yuan, 4-yuan/5-yuan, 4-yuan/6-yuan, 5-yuan/3-yuan, 5-yuan/4-yuan, 5-yuan/5-yuan, 5-yuan/6-yuan, 5-yuan/7-yuan, 6-yuan/3-yuan, 6-yuan/4-yuan, 6-yuan/5-yuan, 6-yuan/6-yuan, 6-yuan/7-yuan, 6-yuan/3-yuan, 6-yu
  • bridged heterocyclic group refers to a polycyclic heterocyclic ring system that shares two atoms that are not directly connected between the rings, which may contain one or more double bonds in the ring, and which contains at least one (e.g., 1, 2, 3, or 4) heteroatoms selected from nitrogen, oxygen, and sulfur in the ring (the nitrogen may be optionally oxidized, i.e., to form nitrogen oxides; the sulfur may be optionally oxidized, i.e., to form sulfoxides or sulfones, but does not include -O-O-, -O-S-, or -S-S-), and has 5 to 20 (e.g., 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20) ring atoms (i.e., 5 to 20-membered bridged heterocyclic groups).
  • the bridged heterocyclic group is preferably a bridged heterocyclic group having 6 to 14 ring atoms (i.e., 6 to 14-membered bridged heterocyclic groups), and more preferably a bridged heterocyclic group having 7 to 10 ring atoms (i.e., 7 to 10-membered bridged heterocyclic groups). According to the number of constituent rings, it can be divided into bicyclic bridged heterocyclic groups and polycyclic bridged heterocyclic groups (such as tricyclic bridged heterocyclic groups, tetracyclic bridged heterocyclic groups, etc.), preferably bicyclic bridged heterocyclic groups or tricyclic bridged heterocyclic groups.
  • Non-limiting examples include:
  • the heterocyclic group may be substituted or unsubstituted. When substituted, it may be substituted at any available point of attachment, and the substituents are preferably selected from one or more of D atoms, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclic groupoxy, hydroxyl, hydroxyalkyl, oxo, cyano, amino, nitro, cycloalkyl, heterocyclic group, aryl and heteroaryl.
  • aryl refers to a monocyclic all-carbon aromatic ring (i.e., monocyclic aromatic group) or a polycyclic aromatic ring system (i.e., polycyclic aromatic group) having a conjugated ⁇ electron system, which has 6 to 20 (e.g., 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) ring atoms (i.e., 6 to 20-membered aromatic group). (i.e., 6 to 14-membered aromatic group).
  • the aromatic group is preferably an aromatic group having 6 to 14 ring atoms (i.e., 6 to 14-membered aromatic group) or an aromatic group having 6 to 10 ring atoms (i.e., 6 to 10-membered aromatic group).
  • the monocyclic aromatic group is, for example, phenyl.
  • Non-limiting examples of the polycyclic aromatic group include: naphthyl, anthracenyl, phenanthrenyl, etc.
  • the polycyclic aromatic group also includes a phenyl group fused with one or more heterocyclic groups or cycloalkyl groups, or a naphthyl group fused with one of the heterocyclic groups or cycloalkyl groups. or multiple fused, where the point of attachment is on the phenyl or naphthyl group, and in this case, the number of ring atoms continues to represent the number of ring atoms in the polycyclic aromatic ring system, non-limiting examples include:
  • the aryl group may be substituted or unsubstituted. When substituted, it may be substituted at any available point of attachment, and the substituents are preferably selected from one or more of D atoms, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxyl, hydroxyalkyl, oxo, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • heteroaryl refers to a monocyclic heteroaromatic ring (i.e., a monocyclic heteroaryl) or a polycyclic heteroaromatic ring system (i.e., a polycyclic heteroaryl) having a conjugated ⁇ electron system, which contains at least one (e.g., 1, 2, 3, or 4) heteroatoms selected from nitrogen, oxygen, and sulfur in the ring (the nitrogen may be optionally oxidized, i.e., to form nitrogen oxides; the sulfur may be optionally oxidized, i.e., to form sulfoxides or sulfones, but does not include -O-O-, -O-S-, or -S-S-), and has 5 to 14 (e.g., 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14) ring atoms (i.e., a 5- to 14-membered heteroaryl).
  • a monocyclic heteroaromatic ring i.e., a monocyclic hetero
  • the heteroaryl is preferably a heteroaryl having 5 to 10 ring atoms (i.e., a 5- to 10-membered heteroaryl), and more preferably a heteroaryl having 5 or 6 ring atoms (i.e., a 5- or 6-membered heteroaryl).
  • the monocyclic heteroaryl group includes, but is not limited to, furanyl, thienyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiadiazolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furazanyl, pyrrolyl, N-alkylpyrrolyl, pyridyl, pyrimidinyl, pyridonyl, N-alkylpyridone (e.g. etc.), pyrazinyl, pyridazinyl, etc.
  • the polycyclic heteroaryl non-limiting examples include: indolyl, indazolyl, quinolyl, isoquinolyl, quinoxalinyl, phthalazinyl, benzimidazolyl, benzothiophenyl, quinazolinyl, benzothiazolyl, carbazolyl, etc.
  • the polycyclic heteroaryl also includes a monocyclic heteroaryl fused with one or more aromatic groups, wherein the connection point is on the aromatic ring, and in this case, the number of ring atoms continues to represent the number of ring atoms in the polycyclic heteroaromatic ring system.
  • the polycyclic heteroaryl also includes a monocyclic heteroaryl fused with one or more cycloalkyl or heterocyclic groups, wherein the connection point is on the monocyclic heteroaromatic ring, and in this case, the number of ring atoms continues to represent the number of ring atoms in the polycyclic heteroaromatic ring system.
  • Non-limiting examples include:
  • the heteroaryl group may be substituted or unsubstituted. When substituted, it may be substituted at any available point of attachment, and the substituent is preferably selected from one or more of a deuterium atom, a halogen, an alkyl group, an alkoxy group, a haloalkyl group, a haloalkoxy group, a cycloalkyloxy group, a heterocyclyloxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, a heterocyclyl group, an aryl group, and a heteroaryl group.
  • cycloalkylalkyl refers to an alkyl group substituted with one or more cycloalkyl groups, wherein cycloalkyl and alkyl are as defined above.
  • heterocyclylalkyl refers to an alkyl group substituted by one or more heterocyclyl groups, wherein heterocyclyl and alkyl are as defined above.
  • alkoxyalkyl refers to an alkyl group substituted by one or more alkoxy groups, wherein alkoxy and alkyl are as defined above.
  • cycloalkyloxy refers to an -O-cycloalkyl group, wherein cycloalkyl is as defined above.
  • heterocyclyloxy refers to an -O-heterocyclyl group wherein the heterocyclyl group is as defined above.
  • haloalkyl refers to an alkyl group substituted with one or more halogens, wherein alkyl is as defined above.
  • haloalkoxy refers to an alkoxy group substituted with one or more halogens, wherein alkoxy is as defined above.
  • hydroxyalkyl refers to an alkyl group substituted with one or more hydroxy groups, wherein alkyl is as defined above.
  • hydroxyalkoxy refers to an alkoxy group substituted with one or more hydroxy groups, wherein alkoxy is as defined above.
  • deuterated alkyl refers to an alkyl group substituted with one or more deuterium atoms, wherein alkyl is as defined above.
  • deuterated methyl refers to a methyl group substituted with one or more deuterium atoms; such as CD 3 , CHD 2 and CH 2 D; preferably CD 3 .
  • deuterated alkoxy refers to an alkoxy group substituted with one or more deuterium atoms, such as deuterated methoxy (OCD 3 , OCHD 2 and OCH 2 D, preferably OCD 3 ).
  • halogen refers to fluorine, chlorine, bromine or iodine.
  • hydroxy refers to -OH.
  • amino refers to -NH2 .
  • cyano refers to -CN.
  • nitro refers to -NO2 .
  • TBS refers to tert-butyldimethylsilyl.
  • Piperidinyl is preferably 1,4-piperidinyl. 1,4-piperidinyl refers to
  • Piperazinyl is preferably 1,4-piperazinyl.
  • 1,4-piperazinyl refers to
  • ubiquitin ligase refers to a family of proteins that promote ubiquitin transfer to specific substrate proteins and target substrate proteins for degradation.
  • cerebellin is an E3 ubiquitin ligase protein that causes ubiquitin to be attached to lysine on a target protein alone or in combination with an E2 ubiquitin conjugating enzyme and then targets specific protein substrates for degradation by the proteasome. Therefore, E3 ubiquitin ligases alone or in combination with E2 ubiquitin conjugating enzymes are the cause of ubiquitin transfer to the target protein.
  • ubiquitin ligases participate in polyubiquitination so that a second ubiquitin is attached to a first ubiquitin, a third ubiquitin is attached to a second ubiquitin, and so on.
  • Polyubiquitination marks proteins for degradation by the proteasome.
  • monoubiquitination in which only a single ubiquitin is added to the substrate molecule by a ubiquitin ligase.
  • Monoubiquitinated proteins are not targeted to the proteasome for degradation, but may instead change in their cellular location or function, such as by binding to other proteins with a domain that can bind ubiquitin.
  • Lys48 on the ubiquitin chain. This is the lysine used to make polyubiquitin, which is recognized by the proteasome.
  • Cereblon (CRBN) E3 ubiquitin ligase is one of the most widely used E3 ubiquitin ligases.
  • CRBN-type E3 ubiquitin ligase ligands include thalidomide and its analogs pomalidomide and lenalidomide, all of which have a phthalimide backbone.
  • Cereblon (CRBN) forms a functional E3 ubiquitin ligase complex with damaged DNA binding protein 1 (DDB1) and Cullin 4A, in which CRBN acts as a substrate receptor. After thalidomide and other degrader molecules bind to CRBN, they induce CRBN to recognize new substrate proteins, causing them to be ubiquitinated and further recognized and degraded by the proteasome.
  • DDB1 DNA binding protein 1
  • Cullin 4A Cullin 4A
  • the ligand binding to cerebellin E3 ubiquitin ligase is a small molecule compound that binds to cerebellin E3 ubiquitin ligase.
  • the ligand of E3 ubiquitin ligase recruits E3 ubiquitin ligase to the vicinity of the target protein, and by getting closer to the target protein, the target protein is ubiquitinated and marked. The marked target protein will be degraded by the proteasome system in the body.
  • amino protecting group refers to a group that is easily removed and introduced on an amino group in order to keep the amino group unchanged when other parts of the molecule are reacted.
  • Non-limiting examples include: (trimethylsilyl)ethoxymethyl, tetramethylsilyl
  • the following are examples of the compounds: hydropyranyl, tert-butyloxycarbonyl (Boc), benzyloxycarbonyl (Cbz), methyloxycarbonyl (Fmoc), allyloxycarbonyl (Alloc), trimethylsilylethoxycarbonyl (Teoc), methoxycarbonyl, ethoxycarbonyl, phthaloyl (Pht), p-toluenesulfonyl (Tos), trifluoroacetyl (Tfa), trityl (Trt), 2,4-dimethoxybenzyl (DMB), acetyl, benzyl, allyl, p-methoxybenzy
  • stereoisomer refers to isomers with identical structures but different arrangements of atoms in space. It includes cis and trans (or Z and E) isomers, (-)- and (+)-isomers, (R)- and (S)-enantiomers, diastereomers, (D)- and (L)-isomers, tautomers, atropisomers, conformers and mixtures thereof (such as racemates, mixtures of diastereomers). Substituents in the disclosed compounds may have additional asymmetric atoms. All of these stereoisomers and their mixtures are included within the scope of the present disclosure.
  • Optically active (-)- and (+)-isomers, (R)- and (S)-enantiomers and (D)- and (L)-isomers may be prepared by chiral synthesis, chiral reagents or other conventional techniques.
  • An isomer of a compound disclosed herein can be prepared by asymmetric synthesis or chiral auxiliary, or, when the molecule contains a basic functional group (such as an amino group) or an acidic functional group (such as a carboxyl group), a diastereoisomer salt is formed with an appropriate optically active acid or base, and then the diastereoisomers are separated by conventional methods known in the art to obtain pure isomers.
  • the separation of enantiomers and diastereoisomers is usually completed by chromatography.
  • the bond Indicates that the configuration is not specified, that is, if there are chiral isomers in the chemical structure, the bond Can be or or include both and For all carbon-carbon double bonds, even if only one configuration is named, both the Z and E configurations are included.
  • tautomer or tautomeric form refers to a structural isomer that exists in equilibrium and is easily converted from one isomeric form to another isomeric form. It includes all possible tautomers, i.e., in the form of a single isomer or in the form of a mixture of any proportions of the tautomers. Non-limiting examples include: keto-enol, imine-enamine, lactam-lactim, etc. Examples of lactam-lactim equilibrium are shown below:
  • isotopic derivatives refers to a compound in which at least one atom is replaced by an atom having the same atomic number but different atomic masses.
  • isotopes that can be introduced into compounds of the present disclosure include stable and radioactive isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine and iodine, such as 2 H (deuterium, D), 3 H (tritium, T), 11 C, 13 C, 14 C, 15 N, 17 O, 18 O, 32 P, 33 P, 33 S, 34 S, 35 S, 36 S, 18 F, 36 Cl, 82 Br, 123 I, 124 I, 125 I, 129 I and 131 I , etc., preferably deuterium.
  • deuterated drugs Compared with non-deuterated drugs, deuterated drugs have the advantages of reducing toxic side effects, increasing drug stability, enhancing therapeutic effects, and extending drug biological half-life. All isotopic composition changes of the compounds disclosed herein, whether radioactive or not, are included in the scope of the present disclosure.
  • Each available hydrogen atom connected to a carbon atom can be independently replaced by a deuterium atom, wherein the replacement of deuterium can be partial or complete, and partial deuterium replacement means that at least one hydrogen is replaced by at least one deuterium.
  • deuterium When a position is specifically designated as deuterium (D), the position is understood to have an abundance of deuterium (ie, at least 15% deuterium incorporation) that is at least 1000 times greater than the natural abundance of deuterium, which is 0.015%.
  • Examples of compounds having a greater than natural abundance of deuterium may be at least 1000 times more abundant in deuterium (i.e., at least 15% deuterium incorporation), at least 2000 times more abundant in deuterium (i.e., at least 30% deuterium incorporation), at least 3000 times more abundant in deuterium (i.e., at least 45% deuterium incorporation), at least 3340 times more abundant in deuterium (i.e., at least 50.1% deuterium incorporation), at least 3500 times more abundant in deuterium (i.e., at least 52.5% deuterium incorporation), at least 4000 times more abundant in deuterium (i.e., at least 60% deuterium incorporation), at least 4500 times more abundant in deuterium (i
  • deuterium is incorporated at least 5000 times more abundant in deuterium (i.e., at least 75% deuterium incorporation), at least 5500 times more abundant in deuterium (i.e., at least 82.5% deuterium incorporation), at least 6000 times more abundant in deuterium (i.e., at least 90% deuterium incorporation), at least 6333.3 times more abundant in deuterium (i.e., at least 95% deuterium incorporation), at least 6466.7 times more abundant in deuterium (i.e., at least 97% deuterium incorporation), at least 6600 times more abundant in deuterium (i.e., at least 99% deuterium incorporation), at least 6633.3 times more abundant in deuterium (i.e., at least 99.5% deuterium incorporation), or more.
  • C 1-6 alkyl optionally substituted by halogen or cyano includes the case where the alkyl is substituted by halogen or cyano and the case where the alkyl is not substituted by halogen or cyano.
  • substitution refers to one or more hydrogen atoms in a group, preferably 1 to 6, more preferably 1 to 3 hydrogen atoms, which are replaced independently of each other by a corresponding number of substituents.
  • substitutions by experiment or theory) without undue effort.
  • an amino or hydroxyl group with free hydrogen may be unstable when combined with a carbon atom with an unsaturated bond (such as an alkene).
  • “Pharmaceutical composition” means a mixture containing one or more compounds described herein or their pharmaceutically acceptable salts and other chemical components, as well as other components such as pharmaceutically acceptable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration to an organism, facilitate the absorption of the active ingredients, and thus exert biological activity.
  • “Pharmaceutically acceptable salts” refer to salts of the compounds of the present disclosure, which may be selected from inorganic or organic salts. Such salts are safe and effective when used in mammals and have the desired biological activity. They may be prepared separately during the final isolation and purification of the compound, or by reacting a suitable group with a suitable base or acid.
  • Bases commonly used to form pharmaceutically acceptable salts include inorganic bases, such as sodium hydroxide and potassium hydroxide, and organic bases, such as ammonia. Acids commonly used to form pharmaceutically acceptable salts include inorganic acids and organic acids.
  • the term "therapeutically effective amount” refers to an amount of the drug or agent sufficient to achieve or at least partially achieve the desired effect.
  • the determination of a therapeutically effective amount varies from person to person, depending on the age and general condition of the recipient and on the specific active substance, and the appropriate therapeutically effective amount in each case can be determined by a person skilled in the art based on routine experiments.
  • pharmaceutically acceptable refers to those compounds, materials, compositions and/or dosage forms that are, within the scope of sound medical judgment, suitable for contact with patient tissues without excessive toxicity, irritation, allergic response, or other problems or complications, commensurate with a reasonable benefit/risk ratio, and effective for the intended use.
  • the method for preparing the compound represented by the general formula (III) or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • the compound represented by the general formula (IIIa) or its salt (preferably hydrochloride) and the compound represented by the general formula (IIIb) or its salt (preferably hydrochloride) undergo condensation reaction in the presence of a condensation agent under alkaline conditions to obtain the compound represented by the general formula (III) or its pharmaceutically acceptable salt;
  • R W is a hydrogen atom
  • Ring J 2 ′, R 1 , R 2 , R 4 to R 6 , RA , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , J 1 , J 5 , J 6 , R q , s and n are as defined in the general formula (III).
  • the method for preparing the compound represented by the general formula (III') or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • the compound represented by the general formula (IIIa) or its salt (preferably hydrochloride) and the compound represented by the general formula (III'b) or its salt (preferably hydrochloride) undergo condensation reaction in the presence of a condensation agent under alkaline conditions to obtain the compound represented by the general formula (III') or its pharmaceutically acceptable salt;
  • R W is a hydrogen atom
  • Ring J2 ', R1 , R2 , R4 , R5 , R7 , RA , Ra , Rb , RZ1 , RZ2 , L1 , L2 , J1 , J5 , J6 , Rq , s, n and Y1 are as defined in the general formula (III').
  • the method for preparing the compound represented by the general formula (IV) or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • the compound represented by the general formula (IVa) or its salt (preferably hydrochloride) and the compound represented by the general formula (IVb) or its salt (preferably hydrochloride) undergo condensation reaction in the presence of a condensation agent under alkaline conditions to obtain the compound represented by the general formula (IV) or its pharmaceutically acceptable salt;
  • Ring J 6 ′, R 1 , R 2 , R 4 to R 6 , RA , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , J 1 , J 2 , R q , p, s and n are as defined in the general formula (IV).
  • the method for preparing the compound represented by the general formula (IV') or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • the compound represented by the general formula (IVa) or its salt (preferably hydrochloride) and the compound represented by the general formula (IV'b) or its salt (preferably hydrochloride) undergo condensation reaction in the presence of a condensation agent under alkaline conditions to obtain the compound represented by the general formula (IV') or its pharmaceutically acceptable salt;
  • Ring J 6 ′, R 1 , R 2 , R 4 , R 5 , R 7 , RA , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , J 1 , J 2 , R q , p , s , n and Y 1 are as defined in the general formula (IV′).
  • the method for preparing the compound represented by the general formula (V-1) or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • R W is a hydrogen atom
  • R 1 , R 2 , R 4 , R 5 , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , RA , n, J 1 , ring J 2 ′, R 03 , j, q, J 4 , J 5 , J 6 , R q , Y 2 and s are as defined in the general formula (V-1).
  • the method for preparing the compound represented by the general formula (V-2) or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • R 1 , R 2 , R 4 , R 5 , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , RA , n, J 1 , q, ring J 3 ′, R 03 , j, J 4 , J 5 , J 6 , R q , Y 2 and s are as defined in the general formula (V-2).
  • the method for preparing the compound represented by the general formula (I'-3-1) or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • the compound represented by the general formula (I'-3-1A) or a salt thereof undergoes an intramolecular ring-closing reaction to obtain a compound represented by the general formula (I'-3-1) or a pharmaceutically acceptable salt thereof;
  • RL is selected from -OH, halogen and -O-alkyl
  • R1 to R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 to J6 , Rq , Y2 and s are as defined in the general formula (I'-3-1).
  • the method for preparing the compound represented by the general formula (I'-3-2) or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • the compound represented by the general formula (I'-3-2A) or a salt thereof undergoes an intramolecular ring-closing reaction to obtain a compound represented by the general formula (I'-3-2) or a pharmaceutically acceptable salt thereof;
  • RL is selected from -OH, halogen and -O-alkyl
  • R1 to R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 to J6 , Rq , Y2 and s are as defined in the general formula (I'-3-2).
  • the present invention discloses a method for preparing a compound represented by general formula (I'-3-1) and (I'-3-2) or a pharmaceutically acceptable salt thereof, comprising the following steps:
  • the compound represented by the general formula (I'-3) or its salt is subjected to chiral resolution to obtain the compound represented by the general formula (I'-3-1) and (I'-3-2) or its pharmaceutically acceptable salt;
  • R1 to R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 to J6 , Rq , Y2 and s are as defined in the general formula (I'-3-1) or (I'-3-2).
  • the method for preparing the compound represented by the general formula (V-1-1) or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • the compound represented by the general formula (V-1-1A) or a salt thereof undergoes an intramolecular ring-closing reaction to obtain a compound represented by the general formula (V-1-1) or a pharmaceutically acceptable salt thereof;
  • RL is selected from -OH, halogen and -O-alkyl
  • R1 , R2 , R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , ring J2 ', R03 , j, q, J4 , J5 , J6 , Rq , Y2 and s are as defined in the general formula (V-1-1).
  • the method for preparing the compound represented by the general formula (V-1-2) or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • the compound represented by the general formula (V-1-2A) or a salt thereof undergoes an intramolecular ring-closing reaction to obtain a compound represented by the general formula (V-1-2) or a pharmaceutically acceptable salt thereof;
  • RL is selected from -OH, halogen and -O-alkyl
  • R1 , R2 , R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , ring J2 ', R03 , j, q, J4 , J5 , J6 , Rq , Y2 and s are as defined in the general formula (V-1-2).
  • the present invention discloses a method for preparing a compound represented by general formula (V-1-1) and (V-1-2) or a pharmaceutically acceptable salt thereof, comprising the following steps:
  • the compound represented by the general formula (V-1) or its salt is subjected to chiral resolution to obtain the compound represented by the general formula (V-1-1) and (V-1-2) or its pharmaceutically acceptable salt;
  • R 1 , R 2 , R 4 , R 5 , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , RA , n, J 1 , ring J 2 ', R 03 , j, q, J 4 , J 5 , J 6 , R q , Y 2 and s are as defined in the general formula (V-1-1) or (V-1-2).
  • the method for preparing the compound represented by the general formula (V-2-1) or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • the compound represented by the general formula (V-2-1A) or a salt thereof undergoes an intramolecular ring-closing reaction to obtain a compound represented by the general formula (V-2-1) or a pharmaceutically acceptable salt thereof;
  • RL is selected from -OH, halogen and -O-alkyl
  • R1 , R2 , R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , q, ring J3 ', R03 , j, J4 , J5 , J6 , Rq , Y2 and s are as defined in the general formula (V-2-1).
  • the method for preparing the compound represented by the general formula (V-2-2) or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • the compound represented by the general formula (V-2-2A) or a salt thereof undergoes an intramolecular ring-closing reaction to obtain a compound represented by the general formula (V-2-2) or a pharmaceutically acceptable salt thereof;
  • RL is selected from -OH, halogen and -O-alkyl
  • R1 , R2 , R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , q, ring J3 ', R03 , j, J4 , J5 , J6 , Rq , Y2 and s are as defined in the general formula (V-2-2).
  • the present invention discloses a method for preparing a compound represented by general formula (V-2-1) and (V-2-2) or a pharmaceutically acceptable salt thereof, comprising the following steps:
  • the compound represented by the general formula (V-2) or its salt is subjected to chiral resolution to obtain the compound represented by the general formula (V-2-1) and (V-2-2) or its pharmaceutically acceptable salt;
  • R 1 , R 2 , R 4 , R 5 , Ra , R b , R Z1 , R Z2 , L 1 , L 2 , RA , n, J 1 , q, ring J 3 ′, R 03 , j, J 4 , J 5 , J 6 , R q , Y 2 and s are as defined in the general formula (V-2-1) or (V-2-2).
  • the method for preparing the compound represented by the general formula (X'-1) or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • q is 1, 2, 3, 4, 5 or 6; q-1 is 0, 1, 2, 3, 4 or 5;
  • R1 , R9 , y, R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , Ring J2 ', R03 , j, J4 , J5 , J6 and E are as defined in the general formula (X'-1).
  • the method for preparing the compound represented by the general formula (X'-2) or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • q is 1, 2, 3, 4, 5 or 6; q-1 is 0, 1, 2, 3, 4 or 5;
  • R1 , R9 , y, R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , Ring J3 ', R03 , j, J4 , J5 , J6 and E are as defined in the general formula (X'-2).
  • the method for preparing the compound represented by the general formula (X'-3) or a pharmaceutically acceptable salt thereof disclosed herein comprises the following steps:
  • q is 1, 2, 3, 4, 5 or 6; q-1 is 0, 1, 2, 3, 4 or 5;
  • R1 , R9 , y, R4 , R5 , Ra , Rb , RZ1 , RZ2 , L1 , L2 , RA , n, J1 , Ring J3 ', R03 , j, J4 , J5 , J6 , Y1 , R7 , Rq and s are as defined in the general formula (X'-3).
  • the reagents providing alkaline conditions in the above condensation reaction include organic bases and inorganic bases.
  • the organic bases include but are not limited to triethylamine, pyridine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, sodium acetate, potassium acetate, sodium tert-butoxide, potassium tert-butoxide or 1,8-diazabicycloundec-7-ene.
  • the inorganic bases include but are not limited to sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide and potassium hydroxide; preferably N,N-diisopropylethylamine.
  • the condensing agent described in the above reaction includes but is not limited to 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, N,N'-dicyclohexylcarbodiimide, N,N'-diisopropylcarbodiimide, O-benzotriazole-N,N,N',N'-tetramethyluronium tetrafluoroborate, 1-hydroxybenzotriazole, 1-hydroxy-7-azobenzotriazole, O-benzotriazole-N,N,N',N'- Tetramethyluronium hexafluorophosphate, O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (HATU), 2-(7-benzotriazolyl oxide)-N,N,N′,N′-tetramethyluronium hexafluoro
  • the reagents providing acidic conditions include but are not limited to hydrogen chloride, a 1,4-dioxane solution of hydrogen chloride, a 1,4-dioxane solution of hydrochloric acid, trifluoroacetic acid, formic acid, acetic acid, glacial acetic acid, hydrochloric acid, concentrated sulfuric acid, methanesulfonic acid, nitric acid, phosphoric acid, p-toluenesulfonic acid, titanium tetrachloride, Me3SiCl and TMSOTf; preferably acetic acid.
  • the acidic reagent can be an acidic reagent added directly; and/or an alkaline salt reagent is added, and the acid generated during the reaction of the alkaline salt and the acid introduced in the form of the acidic salt of the raw material.
  • the alkaline salt includes but is not limited to sodium acetate and anhydrous sodium acetate, preferably anhydrous sodium acetate.
  • the reducing agent includes but is not limited to sodium triacetoxyborohydride, sodium borohydride, lithium borohydride, sodium acetate borohydride, sodium cyanoborohydride and sodium acetylborohydride, preferably sodium triacetoxyborohydride or sodium cyanoborohydride; preferably sodium cyanoborohydride.
  • the intramolecular ring-closing reaction principle of the above-mentioned synthesis schemes six, six-1, seven, seven-1, eight and eight-1 is an amide condensation reaction and/or an amine ester exchange reaction, preferably an amine ester exchange reaction, and the amine ester exchange reaction is optionally carried out in the presence of a catalyst, and the catalyst is preferably benzenesulfonic acid.
  • the reaction in the above steps is preferably carried out in a solvent, and the solvent used includes but is not limited to: pyridine, ethylene glycol dimethyl ether, acetic acid, methanol, ethanol, acetonitrile, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1,4-dioxane, water, N,N-dimethylformamide, N,N-dimethylacetamide, 1,2-dibromoethane and a mixture thereof.
  • the solvent used includes but is not limited to: pyridine, ethylene glycol dimethyl ether, acetic acid, methanol, ethanol, acetonitrile, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • NMR shifts ( ⁇ ) are given in units of 10 -6 (ppm).
  • NMR measurements were performed using a Bruker AVANCE-400 NMR spectrometer or a Bruker AVANCE NEO 500M, with deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD) as the measuring solvent, and tetramethylsilane (TMS) as the internal standard.
  • DMSO-d 6 deuterated dimethyl sulfoxide
  • CDCl 3 deuterated chloroform
  • CD 3 OD deuterated methanol
  • TMS tetramethylsilane
  • MS was determined using an Agilent 1200/1290 DAD-6110/6120 Quadrupole MS LC-MS/MS instrument (manufacturer: Agilent, MS model: 6110/6120 Quadrupole MS).
  • HPLC High performance liquid chromatography
  • Chiral HPLC analysis was performed using an Agilent 1260 DAD high performance liquid chromatograph.
  • HPLC preparation was performed using Waters 2545-2767, Waters 2767-SQ Detecor2, Shimadzu LC-20AP and Gilson GX-281 preparative chromatographs.
  • the thin layer chromatography silica gel plate uses Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate.
  • the silica gel plate used in thin layer chromatography (TLC) adopts a specification of 0.15mm-0.2mm, and the specification used for thin layer chromatography separation and purification products is 0.4mm-0.5mm.
  • Silica gel column chromatography generally uses Yantai Huanghai Silica Gel 200-300 mesh silica gel as the carrier.
  • the average kinase inhibition rate and IC50 value were determined using NovoStar microplate reader (BMG, Germany).
  • the known starting materials disclosed in the present invention can be synthesized by methods known in the art, or can be purchased from companies such as ABCR GmbH & Co. KG, Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc, Bid Pharmaceuticals, and Darui Chemicals.
  • Argon atmosphere or nitrogen atmosphere means that the reaction bottle is connected to an argon or nitrogen balloon with a capacity of about 1L.
  • Hydrogen atmosphere means that the reaction bottle is connected to a hydrogen balloon with a capacity of about 1L.
  • the pressurized hydrogenation reaction uses a Parr 3916EKX hydrogenator and a Clear Blue QL-500 hydrogen generator or a HC2-SS hydrogenator.
  • the hydrogenation reaction is usually carried out by evacuating the vacuum, filling with hydrogen, and repeating the operation three times.
  • Microwave reactions were performed using a CEM Discover-S 908860 microwave reactor.
  • the solution refers to an aqueous solution.
  • reaction temperature is room temperature, 20°C to 30°C.
  • reaction solution was cooled to room temperature, the reaction solution was filtered through diatomaceous earth, the filter cake was washed with ethyl acetate (50 mL ⁇ 3), the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with eluent system B to obtain the title compound 1c (500 mg, yield: 18%).
  • N,N-dimethylformamide 3 mL
  • compound 1h 60 mg, 0.082 mmol
  • ( ⁇ )-2-(4-(4-((2,6-dioxopiperidin-3-yl)amino)phenyl)piperidin-1-yl)acetic acid hydrochloride 1i 31 mg, 0.082 mmol, prepared by the known method "General Step E on page 400 of the specification in WO2021127561A1"
  • O-(7-azabenzotriazole-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate 37 mg, 0.098 mmol
  • N,N-diisopropylethylamine 63 mg, 0.49 mmol
  • reaction solution was filtered and purified by HPLC preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, acetonitrile gradient: 55%-67%, flow rate: 30 mL/min) to give the title compound 1 (racemate, 40 mg, yield: 48%).
  • N,N-dimethylformamide 3 mL
  • compound 2b 40 mg, 0.055 mmol
  • compound 1i 22 mg, 0.058 mmol
  • O-(7-azabenzotriazole-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate 32 mg, 0.083 mmol
  • N,N-diisopropylethylamine 43 mg, 0.083 mmol
  • reaction solution was filtered and purified by high performance liquid preparative chromatography (Waters 2545, elution system: 10mmol/L ammonium bicarbonate aqueous solution and acetonitrile, acetonitrile gradient: 55%-60%, flow rate: 30mL/min) to obtain the title compound 2 (racemate, 30mg, yield: 53%).
  • N,N-dimethylformamide (3 mL), compound 3b (35 mg, 0.049 mmol), compound 1i (21 mg, 0.054 mmol), O-(7-azabenzotriazole-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (21 mg, 0.054 mmol) were added to a 25 mL single-mouth bottle in sequence.
  • N,N-diisopropylethylamine 32 mg, 0.25 mmol was slowly added under ice bath and reacted for 20 minutes.
  • reaction solution was filtered and purified by high performance liquid preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, acetonitrile gradient: 48%-58%, flow rate: 30 mL/min) to obtain the title compound 3 (racemate, 22 mg, yield: 43%).
  • N,N-dimethylformamide 3 mL
  • compound 1h 53 mg, 0.072 mmol
  • 2-(4-(3-(2,4-dioxotetrahydropyrimidine-1(2H)-yl)-1-methyl-1H-indazol-6-yl)-3,3-difluoropiperidin-1-yl)acetic acid hydrochloride 4a 28 mg, 0.061 mmol, prepared by the known method "page 401 of the specification in WO2021127561A1"
  • O-(7-azabenzotriazole-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate 27 mg, 0.072 mmol
  • N,N-diisopropylethylamine 46 mg, 0.36 mmol
  • reaction solution was filtered and purified by HPLC preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, acetonitrile gradient: 50%-65%, flow rate: 30 mL/min) to give the title compound 4 (racemate, 25 mg, yield: 32%).
  • N,N-dimethylformamide (3 mL), compound 3b (35 mg, 0.049 mmol), compound 4a (24 mg, 0.052 mmol), O-(7-azabenzotriazole-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (21 mg, 0.054 mmol) were added to a 25 mL three-necked flask in sequence.
  • N,N-diisopropylethylamine 32 mg, 0.25 mmol was slowly added under ice bath and reacted for 20 minutes.
  • reaction solution was filtered and purified by high performance liquid preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, acetonitrile gradient: 47%-53%, flow rate: 30 mL/min) to obtain the title compound 5 (racemic, 25 mg, yield: 46%).
  • N,N-dimethylformamide (3 mL), compound 6g (64 mg, 0.089 mmol), compound 4a (41 mg, 0.089 mmol), O-(7-azabenzotriazole-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (33 mg, 0.087 mmol) were added to a 25 mL three-necked flask in sequence.
  • N,N-diisopropylethylamine 34 mg, 0.26 mmol was slowly added under ice bath and reacted for 20 minutes.
  • reaction solution was filtered and purified by high performance liquid preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, acetonitrile gradient: 40%-60%, flow rate: 30 mL/min) to obtain the title compound 6 (racemic body, 35 mg, yield: 36%).
  • N,N-dimethylformamide (3 mL), compound 7b (32 mg, 0.047 mmol), compound 4a (21 mg, 0.047 mmol), O-(7-azabenzotriazole-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (17.7 mg, 0.047 mmol) were added to a 25 mL three-necked flask in sequence.
  • N,N-diisopropylethylamine (12 mg, 0.093 mmol) was slowly added under ice bath and reacted for 20 minutes.
  • reaction solution was filtered and purified by high performance liquid chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, acetonitrile
  • the title compound 7 (racemate, 10 mg, yield: 19%) was obtained by evaporation of 4% to 62% gradient, flow rate: 30 mL/min).
  • N,N-dimethylformamide (3mL), compound 8g (33mg, 0.044mmol), 1-(6-(3,3-difluoropiperidin-4-yl)-1-methyl-1H-indazol-3-yl)dihydropyrimidine-2,4(1H,3H)-dione hydrochloride 8h (18mg, 0.045mmol, prepared by the known method "WO2021127561A1, page 383 of the specification”), O-(7-azabenzotriazole-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (23mg, 0.060mmol) were added in sequence.
  • N,N-diisopropylethylamine 35mg, 0.27mmol was slowly added and reacted for 30 minutes.
  • the reaction solution was filtered and purified by HPLC preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, acetonitrile gradient: 45%-65%, flow rate: 30 mL/min) to give the title compound 8 (racemic form, 11 mg, yield: 23%).
  • reaction solution was filtered through diatomaceous earth, the filter cake was washed with ethyl acetate (30 mL), the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with eluent system B to obtain the title compound 10b (1.20 g, yield: 94%).
  • reaction solution was filtered and purified by high performance liquid preparative chromatography (Waters-2545, elution system: 10 mmol/L aqueous solution of ammonium bicarbonate and acetonitrile, acetonitrile gradient: 45%-65%, flow rate: 30 mL/min) to obtain the title compound 10 (racemic mixture, 25 mg, yield: 32%).
  • reaction solution After the reaction solution is cooled to room temperature, the reaction solution is filtered through diatomaceous earth, the filtrate is concentrated under reduced pressure, ethyl acetate (30 mL) is added to the resulting residue, the insoluble matter is filtered off, the filtrate is concentrated under reduced pressure, and vacuum dried to obtain the crude product title compound 11c (1.0 g, yield: 88%), which is directly used in the next step without purification.
  • reaction solution was filtered, and purified by high performance liquid preparative chromatography (Waters-2545, elution system: 10 mmol/L aqueous solution of ammonium bicarbonate and acetonitrile, acetonitrile gradient: 58%-68%, flow rate: 30 mL/min) to obtain the title compound 11 (racemic mixture, 18 mg, yield: 22%).
  • N,N-dimethylformamide (2 mL), compound 11f (60 mg, 0.073 mmol), (2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-L-alanine 12a (25 mg, 0.072 mmol, prepared by the method disclosed in patent application "Compound 235 on page 419 of the specification of WO2020051235A1", and O-(7-azabenzotriazole-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (41 mg, 0.11 mmol) were added in sequence, and N,N-diisopropylethylamine (29 mg, 0.22 mmol) was slowly added under ice bath, and the reaction was carried out for 20 minutes.
  • reaction solution was filtered and purified by HPLC preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, acetonitrile gradient: 40%-85%, flow rate: 30 mL/min) to obtain the title compound 12 (a mixture of 2 diastereoisomers, 20 mg, yield: 25%).
  • reaction solution was cooled to room temperature, the reaction solution was filtered with diatomaceous earth, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with eluent system A to obtain the title compound 13b (1.12 g, yield: 95%).
  • reaction solution was filtered and purified by high performance liquid preparative chromatography (Waters-2545, elution system: 10 mmol/L aqueous solution of ammonium bicarbonate and acetonitrile, acetonitrile gradient: 55%-85%, flow rate: 30 mL/min) to obtain the title compound 13e (60 mg, yield: 19%).

Landscapes

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

Abstract

一种取代的氨基嘧啶类化合物、其制备方法及含有该类化合物的药物组合物以及其作为治疗剂的用途,特别是作为EGFR抑制剂和/或降解剂的用途和在制备用于治疗和/或预防由EGFR介导的或依赖性的疾病或病症的药物中的用途。

Description

取代的氨基嘧啶类化合物、其制备方法及其在医药上的应用 技术领域
本公开属于医药领域,涉及一种通式(X)所示的取代的氨基嘧啶类化合物、其制备方法、含有该化合物的药物组合物以及其作为治疗剂的用途,特别是作为EGFR抑制剂和/或降解剂的用途和在制备用于治疗和/或预防由EGFR介导的或依赖性的疾病或病症的药物中的用途。
背景技术
PROTAC(Proteolysis Targeting Chimera)是一种杂合双功能小分子化合物。其结构中含有两种不同配体:一个是泛素连接酶E3配体,另一个是与靶蛋白结合配体,两个配体之间由连接臂相连。PROTAC通过将靶蛋白和细胞内的泛素连接酶E3拉近,形成靶蛋白-PROTAC-E3三元复合物,接着E3泛素连接酶给靶蛋白标记泛素化蛋白标签,随后启动细胞内强大的泛素化—蛋白酶体系统,特异性地降解靶蛋白,进而达到抑制相应蛋白信号通路的作用。与传统的小分子抑制剂相比,PROTAC展现出了独特的优势:1、PROTAC不需要与目标靶蛋白长时间和高强度的结合,并且降解靶蛋白过程类似于催化反应,可循环结合,降解靶蛋白。从而降低药物的系统暴露量,减少毒副作用的发生。2、靶蛋白被降解后需要重新合成才能恢复功能,因此降解靶蛋白比抑制其活性显示出更加高效、持久的抗肿瘤作用,且不易发生因靶蛋白突变而产生的耐药性。3、对于目前认为不可成药的靶点,例如转录因子、支架蛋白和调控蛋白等,PROTAC也具有治疗潜力。
肺癌是最为常见的恶性肿瘤之一。据统计2018年全球发病率最高的癌症是肺癌,有209.4万人为新发肺癌患者。中国2018年肺癌发病人数是86.8万人,发病率是0.062%,发病人数占全球的41.4%。非小细胞肺癌是最常见的肺癌类型,发病率约占肺癌总数的85%左右。表皮生长因子受体(Epidermal growth factor receptor,EGFR)属于具有酪氨酸激酶活性的细胞表面受体家族。EGFR突变是最常见的非小细胞肺癌驱动基因,大约40%的中国非小细胞肺癌患者存在EGFR突变,而11-16%的西方国家病人存在EGFR突变。EGFR的突变形式大约90%是19号外显子的缺失(Del19突变)和21号外显子的L858R点突变。
多个EGFR小分子抑制剂已经被批准上市,成功应用于具有EGFR突变的非小细胞肺癌的治疗,成为晚期非小细胞肺癌病人的主要治疗手段之一。第一代EGFR酪氨酸激酶抑制剂以吉非替尼和厄洛替尼为代表是可逆结合的靶向性药物,通过与ATP竞争结合EGFR激酶的结构域,达到抑制其活化的效果。但是大多数病人在经过10-12月的治疗后,会产生耐药,大约50%的病人耐药是由于产生了T790M的二次突变。第二代EGFR酪氨酸激酶抑制剂以阿法替尼为代表是不可逆 的靶向药物,但是不能够解决T790M突变耐药问题,并且由于对于野生型EGFR缺少选择性使得化合物毒性较大。第三代EGFR酪氨酸激酶抑制剂奥西替尼的出现克服了由于EGFR T790M基因突变导致的耐药,且对野生型EGFR的抑制较弱、选择性好,在临床上取得了巨大成功,但是使用9-14个月之后会产生新的耐药,研究揭示6-26%的病人产生了C797X或其它EGFR依赖的基因突变(JAMA Oncol.2018,4,1527-1534;Br J Cancer.2019Oct;121(9):725-737.)。
目前在市场上尚缺少针对EGFR C797S基因突变的单独用药有效的EGFR抑制剂,因此有必要开发新的用于非小细胞肺癌的治疗手段。不同于EGFR抑制剂,利用PROTAC技术可以降解EGFR,能够更加有效地抑制EGFR信号通路,有可能成为有潜力的非小细胞肺癌治疗方法。已公开的EGFR蛋白靶向降解PROTAC化合物的专利申请包括WO2017185036A1、WO2018119441A1、WO2019121562A1、WO2019149922A1、WO2021121261A1、WO2021127561A1、WO2021039622A1、WO2022055181A1、WO2022012623A1、WO2022068849A1。
发明内容
本公开的目的是提供一种通式(X)所示的化合物,或其可药用的盐:
其中,
R1选自-P(O)R10R11、-S(O)rR12、-S(O)rN(R13R14)、-N(R15)S(O)rR16和R9
各个R9相同或不同,且各自独立地选自氘原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、羟基、烯基、炔基、氰基、硝基、NR20R21、C(O)NR20R21、NR20C(O)R24、-亚烷基-NR20R21、-亚烷基-C(O)NR20R21、C(O)R24、C(O)OR24、环烷基、环烷基烷基、杂环基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、环烷基烷基、杂环基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
或,两个R9及与其相连的碳原子一起形成任选被一个或多个R01所取代的环烷基、杂环基、芳基或杂芳基;
L1与苯基连接,或L1连接至两个R9形成的环上;
R4和R5相同或不同,且各自独立地选自氢原子、氘原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、羟基、烯基、炔基、氰基、硝基、NR22R23、C(O)NR22R23、NR22C(O)R24、C(O)R24、C(O)OR24、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
或,R4、R5及与其相连的碳原子一起形成任选被一个或多个R01所取代的环烷基、杂环基、芳基或杂芳基;
环A选自环烷基、杂环基、芳基和杂芳基;
各个RA相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、硝基、NR25R26、C(O)NR25R26、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
L1和L3相同或不同,且各自独立地选自键、O、S、NRL、S(O)r、C(O)、C(O)NRL、NRLC(O)、S(O)rNRL、NRLC(O)NRL、(CRL1RL2)m、烯基和炔基;所述的烯基任选被一个或多个R01所取代;
L2为-(CRL3RL4)n1-RL5-(CRL6RL7)n2-;
RL5选自键、O、S、NRL’、S(O)、S(O)2、C(O)、C(O)NRL’、NRL’C(O)和NRL’C(O)NRL’;
各个RL1、RL2、RL3、RL4、RL6和RL7相同或不同,且各自独立地选自氢原子、氘原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
或,RL1、RL2及与其相连的碳原子一起形成环烷基或杂环基,或,RL3、RL4及与其相连的碳原子一起形成环烷基或杂环基,或,RL6、RL7及与其相连的碳原子一起形成环烷基或杂环基;所述的环烷基或杂环基各自独立地任选被一个或多个R01所取代;
或,RL3、RL4及与其相连的两个相邻碳原子一起形成C=C或C≡C,或RL6、RL7及与其相连的两个相邻碳原子一起形成C=C或C≡C;
R10、R11、R12、R13、R14、R15和R16相同或不同,且各自独立地选自氢原子、氘原子、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R01所取代;
各个R20、R21、R22、R23、R24、R25、R26、RL和RL’相同或不同,且各自独立地选自氢原子、氘原子、烷基、卤代烷基、烷氧基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个R01所取代;
或,R13、R14及与其相连的氮原子一起形成杂环基;或NR15及与其相连的碳原子、R9及与其相连的碳原子一起形成含氮杂环基或含氮杂芳基;或R20、R21及与其相连的氮原子一起形成杂环基;或R22、R23及与其相连的氮原子一起形成杂环基;或R25、R26及与其相连的氮原子一起形成杂环基;所述的杂环基、含氮杂环基或含氮杂芳基各自独立地任选被一个或多个R01所取代;
Ra和Rb相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、环烷基、和杂环基;
各个R01相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
Z1为N或CRZ1
Z2为N或CRZ2
Z3为N或CRZ3
RZ1、RZ2和RZ3相同或不同,且各自独立地选自氢原子、氘原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、羟基、烯基、炔基、氰基、硝基、C(O)R30、C(O)OR30、NR31R32、C(O)NR31R32、NR31C(O)R30、环烷基、环烷基氧基、杂环基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、环烷基氧基、杂环基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R02所取代;
各个R30相同或不同,且各自独立地选自氢原子、烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基;所述的烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R02所取代;
各个R31和R32相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个R02所取代;
各个R02相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
J1、J2、J3、J4、J5和J6相同或不同,且各自独立地选自键、O、S、-O-亚烷基-、-亚烷基-O-、C(O)、-C(O)-亚烷基-、-亚烷基-C(O)-、C(O)N(RJ)、N(RJ)C(O)、N(RJ)C(O)N(RJ)、S(O)v、S(O)vN(RJ)、N(RJ)S(O)v、N(RJ)、-N(RJ)-亚烷基-、-亚烷基-N(RJ)-、亚烷基、烯基、炔基、环烷基、杂环基、-亚烷基-杂环基-、-杂环基-亚烷基-、芳基和杂芳基;所述的亚烷基、烯基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个选自R03的取代基所取代;
各个RJ相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个选自R03的取代基所取代;
各个R03相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
E选自:
为单键或者双键;
环D为芳基或杂芳基;
W选自键、O、S、S(O)r、NR6、C(O)、C(O)NR6、NR6C(O)和(CRY2RY2)s4
Q1、Q2、Q3、Q4和Q5中的一者为碳原子且与J6连接,其它各自独立地选自N、CH和CRq
各个Rq相同或不同,且各自独立地选自氘原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、羟基、烯基、炔基、氰基、NRcRd、亚烷基NRcRd、C(O)烷基、C(O)NRcRd、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R04所取代;
或两个Rq及与其相连的环原子一起形成环烷基或杂环基;
各个Rc和Rd相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、环烷基和杂环基;或Rc、Rd及与其相连的氮原子一起形成杂环基,所述的杂环基各自独立地任选被一个或多个R04所取代;
Y2为C(RY2)2或C(O);
Y1为N或CRY1;Y3为N或CRY3;Y4选自键、CH2或C(O);
各个RY2相同或不同,且各自独立地选自氢原子、氘原子、烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R04所取代;
或,两个RY2与其相连的碳原子一起形成环烷基或杂环基;所述的环烷基和杂环基各自独立地任选被一个或多个R04所取代;
RY1和RY3相同或不同,且各自独立地选自氢原子、氘原子、烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R04所取代;
R6、R7和R8相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、环烷基和杂环基;
各个R04相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
n为0、1、2、3、4、5或6;
r为0、1或2;v为0、1或2;
y为0、1、2或3;s1为0、1、2、3、4、5或6;
s2为0、1、2、3或4;s3为0、1、2、3或4;s4为0、1、2、3或4;且,
n1、n2和m各自独立地为0、1、2、3、4、5、6、7、8、9或10。
本公开的目的是提供一种通式(X)所示的化合物,或其可药用的盐:
其中:
E为结合至小脑蛋白E3泛素连接酶的配体;
R1、R9、y、Ra、Rb、R4、R5、Z1、Z2、Z3、L1、L2、环A、L3、RA、n、J1至J6如上所定义。
本公开的目的是提供一种通式(I)所示的化合物,或其可药用的盐:
其中,
R1选自-P(O)R10R11、-S(O)rR12、-S(O)rN(R13R14)和-N(R15)S(O)rR16
R2和R3相同或不同,且各自独立地选自氢原子、氘原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、羟基、烯基、炔基、氰基、硝基、NR20R21、C(O)NR20R21、NR20C(O)R24、-亚烷基-NR20R21、-亚烷基-C(O)NR20R21、C(O)R24、C(O)OR24、环烷基、环烷基烷基、杂环基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、环烷基烷基、杂环基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
或,R2、R3及与其相连的碳原子一起形成任选被一个或多个R01所取代的环烷基、杂环基、芳基或杂芳基;或,R3与L1连接;
R4和R5相同或不同,且各自独立地选自氢原子、氘原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、羟基、烯基、炔基、氰基、硝基、NR22R23、C(O)NR22R23、NR22C(O)R24、C(O)R24、C(O)OR24、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔 基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
或,R4、R5及与其相连的碳原子一起形成任选被一个或多个R01所取代的环烷基、杂环基、芳基或杂芳基;
环A选自环烷基、杂环基、芳基和杂芳基;
各个RA相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、硝基、NR25R26、C(O)NR25R26、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
L1和L3相同或不同,且各自独立地选自键、O、S、NRL、S(O)r、C(O)、C(O)NRL、NRLC(O)、S(O)rNRL、NRLC(O)NRL、(CRL1RL2)m、烯基和炔基;所述的烯基任选被一个或多个R01所取代;
L2为-(CRL3RL4)n1-RL5-(CRL6RL7)n2-;
RL5选自键、O、S、NRL’、S(O)、S(O)2、C(O)、C(O)NRL’、NRL’C(O)和NRL’C(O)NRL’;
各个RL1、RL2、RL3、RL4、RL6和RL7相同或不同,且各自独立地选自氢原子、氘原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
或,RL1、RL2及与其相连的碳原子一起形成环烷基或杂环基,或,RL3、RL4及与其相连的碳原子一起形成环烷基或杂环基,或,RL6、RL7及与其相连的碳原子一起形成环烷基或杂环基;所述的环烷基或杂环基各自独立地任选被一个或多个R01所取代;
或,RL3、RL4及与其相连的两个相邻碳原子一起形成C=C或C≡C,或RL6、RL7及与其相连的两个相邻碳原子一起形成C=C或C≡C;
R10、R11、R12、R13、R14、R15和R16相同或不同,且各自独立地选自氢原子、氘原子、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R01所取代;
各个R20、R21、R22、R23、R24、R25、R26、RL和RL’相同或不同,且各自独立地选自氢原子、氘原子、烷基、卤代烷基、烷氧基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个R01所取代;
或,R13、R14及与其相连的氮原子一起形成杂环基;或NR15及与其相连的碳原子、R2及与其相连的碳原子一起形成含氮杂环基或含氮杂芳基;或R20、R21及 与其相连的氮原子一起形成杂环基;或R22、R23及与其相连的氮原子一起形成杂环基;或R25、R26及与其相连的氮原子一起形成杂环基;所述的杂环基、含氮杂环基或含氮杂芳基各自独立地任选被一个或多个R01所取代;
Ra和Rb相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、环烷基、和杂环基;
各个R01相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
Z1为N或CRZ1
Z2为N或CRZ2
Z3为N或CRZ3
RZ1、RZ2和RZ3相同或不同,且各自独立地选自氢原子、氘原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、羟基、烯基、炔基、氰基、硝基、C(O)R30、C(O)OR30、NR31R32、C(O)NR31R32、NR31C(O)R30、环烷基、环烷基氧基、杂环基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、环烷基氧基、杂环基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R02所取代;
各个R30相同或不同,且各自独立地选自氢原子、烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基;所述的烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R02所取代;
各个R31和R32相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个R02所取代;
各个R02相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
J1、J2、J3、J4、J5和J6相同或不同,且各自独立地选自键、O、S、-O-亚烷基-、-亚烷基-O-、C(O)、-C(O)-亚烷基-、-亚烷基-C(O)-、C(O)N(RJ)、N(RJ)C(O)、N(RJ)C(O)N(RJ)、S(O)v、S(O)vN(RJ)、N(RJ)S(O)v、N(RJ)、-N(RJ)-亚烷基-、-亚烷基-N(RJ)-、亚烷基、烯基、炔基、环烷基、杂环基、-亚烷基-杂环基-、-杂环基- 亚烷基-、芳基和杂芳基;所述的亚烷基、烯基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个选自R03的取代基所取代;
各个RJ相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个选自R03的取代基所取代;
各个R03相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
E选自:
为单键或者双键;
Q1、Q2、Q3、Q4和Q5中的一者为碳原子且与J6连接,其它各自独立地选自N或CRQ
各个RQ相同或不同,且各自独立地选自氢原子、氘原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、羟基、烯基、炔基、氰基、NRcRd、亚烷基NRcRd、C(O)烷基、C(O)NRcRd、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R04所取代;
各个Rc和Rd相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、环烷基和杂环基;或Rc、Rd及与其相连的氮原子一起形成杂环基,所述的杂环基各自独立地任选被一个或多个R04所取代;
Y2为C(RY2)2或C(O);
Y1为N或CRY1;Y3为N或CRY3
各个RY2相同或不同,且各自独立地选自氢原子、氘原子、烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R04所取代;
或,两个RY2与其相连的碳原子一起形成环烷基或杂环基;所述的环烷基和杂环基各自独立地任选被一个或多个R04所取代;
RY1和RY3相同或不同,且各自独立地选自氢原子、氘原子、烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R04所取代;
R6、R7和R8相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、环烷基和杂环基;
各个R04相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
n为0、1、2、3、4、5或6;
r为0、1或2;
v为0、1或2;且,
n1、n2和m各自独立地为0、1、2、3、4、5、6、7、8、9或10。
本公开的目的是提供一种通式(I)所示的化合物,或其可药用的盐:
其中:
R1选自-P(O)R10R11、-SO2R12、-SO2N(R13R14)和-N(R15)SO2R16
R2和R3相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、硝基、NR20R21、C(O)NR20R21、NR20C(O)R24、-亚烷基-NR20R21、-亚烷基-C(O)NR20R21、C(O)R24、C(O)OR24、环烷基、环烷基烷基、杂环基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、环烷基烷基、杂环 基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
或,R2、R3及与其相连的碳原子一起形成任选被一个或多个R01所取代的环烷基、杂环基、芳基或杂芳基;或,R3与L1连接;
R4和R5相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、硝基、NR22R23、C(O)NR22R23、NR22C(O)R24、C(O)R24、C(O)OR24、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
或,R4、R5及与其相连的碳原子一起形成任选被一个或多个R01所取代的环烷基、杂环基、芳基或杂芳基;
环A选自环烷基、杂环基、芳基和杂芳基;
各个RA相同或不同,且各自独立地选自氧代基、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、硝基、NR25R26、C(O)NR25R26、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
L1和L3相同或不同,且各自独立地选自键、O、S、NRL、S(O)r、C(O)、C(O)NRL、NRLC(O)、S(O)rNRL、NRLC(O)NRL、(CRL1RL2)m、烯基和炔基;所述的烯基任选被一个或多个R01所取代;
L2为-(CRL3RL4)n1-RL5-(CRL6RL7)n2-;
RL5选自键、O、S、NRL’、S(O)、S(O)2、C(O)、C(O)NRL’、NRL’C(O)和NRL’C(O)NRL’;
各个RL1、RL2、RL3、RL4、RL6和RL7相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
或,RL1、RL2及与其相连的碳原子一起形成环烷基或杂环基,或,RL3、RL4及与其相连的碳原子一起形成环烷基或杂环基,或,RL6、RL7及与其相连的碳原子一起形成环烷基或杂环基;所述的环烷基或杂环基各自独立地任选被一个或多个R01所取代;
或,RL3、RL4及与其相连的两个相邻碳原子一起形成C=C或C≡C,或RL6、RL7及与其相连的两个相邻碳原子一起形成C=C或C≡C;
R10、R11、R12、R13、R14、R15和R16相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、环烷基和杂环基;所述的 烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R01所取代;
各个R20、R21、R22、R23、R24、R25、R26、RL和RL’相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、烷氧基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个R01所取代;
或,R13、R14及与其相连的氮原子一起形成杂环基;或NR15及与其相连的碳原子、R2及与其相连的碳原子一起形成含氮杂环基或含氮杂芳基;或R20、R21及与其相连的氮原子一起形成杂环基;或R22、R23及与其相连的氮原子一起形成杂环基;或R25、R26及与其相连的氮原子一起形成杂环基;所述的杂环基、含氮杂环基或含氮杂芳基各自独立地任选被一个或多个R01所取代;
Ra和Rb相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、环烷基、和杂环基;
各个R01相同或不同,且各自独立地选自氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
Z1为N或CRZ1
Z2为N或CRZ2
Z3为N或CRZ3
RZ1、RZ2和RZ3相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、硝基、C(O)R30、C(O)OR30、NR31R32、C(O)NR31R32、NR31C(O)R30、环烷基、环烷基氧基、杂环基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、环烷基氧基、杂环基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R02所取代;
各个R30相同或不同,且各自独立地选自氢原子、烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基;所述的烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R02所取代;
各个R31和R32相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个R02所取代;
各个R02相同或不同,且各自独立地选自氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、 硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
J1、J2、J3、J4、J5和J6相同或不同,且各自独立地选自键、O、S、-O-亚烷基-、-亚烷基-O-、-C(O)-、-C(O)-亚烷基-、-亚烷基-C(O)-、-C(O)N(RJ)-、-N(RJ)C(O)-、-N(RJ)C(O)N(RJ)-、-S(O)v-、-S(O)vN(RJ)-、-N(RJ)S(O)v-、-N(RJ)-、-N(RJ)-亚烷基-、-亚烷基-N(RJ)-、亚烷基、烯基、炔基、环烷基、杂环基、-亚烷基-杂环基-、-杂环基-亚烷基-、芳基和杂芳基;所述的亚烷基、烯基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个选自R03的取代基所取代;
各个RJ相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个选自R03的取代基所取代;
各个R03相同或不同,且各自独立地选自氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
E选自:
为单键或者双键;
Q1、Q2、Q3、Q4和Q5中的一者为碳原子且与J6连接,其它各自独立地选自N或CRQ
各个RQ相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、NRcRd、亚烷基NRcRd、C(O)烷基、C(O)NRcRd、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R04所取代;
各个Rc和Rd相同或不同,且各自独立地选自氢原子、卤素、烷基、环烷基和杂环基;或Rc、Rd及与其相连的氮原子一起形成杂环基,所述的杂环基各自独立地任选被一个或多个R04所取代;
Y2为C(RY2)2或C(O);
Y1为N或CRY1;Y3为N或CRY3
各个RY2相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R04所取代;
或,两个RY2与其相连的碳原子一起形成环烷基或杂环基;所述的环烷基和杂环基各自独立地任选被一个或多个R04所取代;
RY1和RY3相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R04所取代;
R6、R7和R8相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、环烷基和杂环基;
各个R04相同或不同,且各自独立地选自氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
n为0、1、2、3、4、5或6;
r为1或2;
v为1或2;且,
n1、n2和m各自独立地为0、1、2、3、4、5、6、7、8、9或10。
在本公开的一些实施方案中,所述的通式(X)、(I)所示的化合物或其可药用的盐,其中环A为苯基或5或6元杂芳基;优选为5或6元杂芳基;更优选为5元杂芳基;进一步优选为吡唑基;最优选为
在本公开的一些实施方案中,所述的通式(X)、(I)所示的化合物或其可药用的盐,其中各个RA相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基,n为0、1或2;更优选为
在本公开的一些实施方案中,所述的通式(X)、(I)所示的化合物或其可药用的盐,其中n3为1、2、3、4、5、6、7、8、9或10;n为0、1或2;RL3、RL4和RA如通式(I)中所定义;优选地,各个RL3和RL4相同或不同,且各自独立地选自氢原子、卤素、C1-6烷基和C1-6卤代烷基,各个RA相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基,n为0、1或2;n3为1、2、3、4、5、6、7、8、9或10;
更优选地,n3为1、2、3、4、5、6、7、8、9或10;带&的键与Z1所在的环连接。
在本公开的一些实施方案中,所述的通式(X)所示的化合物或其可药用的盐,其为通式(X’)所示的化合物或其可药用的盐,
其中,n为0、1或2;
R1、R9、y、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、J1至J6和E如通式(X)中所定义。
在本公开的一些实施方案中,所述的通式(X)、(X’)所示的化合物或其可药用的盐,其为通式(X’-1)所示的化合物或其可药用的盐,
其中,环J2’为含氮杂环基;j为0、1、2、3、4、5或6;
q为0、1、2、3、4、5或6;n为0、1或2;
R1、R9、y、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、J1、R03、J4、J5、J6和E如通式(X)中所定义。
在本公开的一些实施方案中,所述的通式(X)、(X’)所示的化合物或其可药用的盐,其为通式(X’-2)所示的化合物或其可药用的盐,
其中,环J3’为含氮杂环基;j为0、1、2、3、4、5或6;
q为0、1、2、3、4、5或6;n为0、1或2;
R1、R9、y、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、J1、R03、J4、J5、J6和E如通式(X)中所定义。
本公开的一些实施方案中,所述的通式(I)、(X)、(X’)、(X’-1)、(X’-2)所示的化合物或其可药用的盐,其中,E选自: s为0、1、2或3,Rq、R6、Y1、R7、Y2和R8如通式(X)中所定义;
在一些实施方案中,E选自: s为0、1、2或3,Y1、Y2、R6、R7、R8和Rq如通式(X)中所定义;
在一些实施方案中,E选自: s为0、1、2或3,Y1、Y2、R6、R7、R8和Rq如通式(X)中所定义;
在一些实施方案中,E选自:
在一些实施方案中,E选自:各个Rq相同或不同,且各自独立地选自卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、NRcRd、亚烷基NRcRd、C(O)烷基、C(O)NRcRd、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R04所取代;s为0、1、2或3;Y1、Y2、R6、R7、Rc、Rd和R04如通式(I)中所定义;
在一些实施方案中,E选自:Y1为N或CH;Y2为CH2或C(O);各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;R6为氢原子或C1-6烷基;R7为C1-6烷基;s为0、1、2或3;
在一些实施方案中,E选自: Y1为N或CH;Y2为CH2或C(O);各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;R6为氢原子,R7为C1-6烷基,R8为C1-6烷基;s为0、1、2或3;
在一些实施方案中,E选自:
在一些实施方案中,E选自: R6和R7相同或不同,且各自独立地为氢原子或C1-6烷基;各个Rq相同或不同,且各自独立地选自卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、NRcRd、亚烷基NRcRd、C(O)烷基、C(O)NRcRd、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R04所取代;s为0、1、2或3;Rc、Rd和R04如通式(I)中所定义;
在一些实施方案中,E选自 R6和R7相同或不同,且各自独立地为氢原子或C1-6烷基;
在一些实施方案中,E选自
在一些实施方案中,E选自:
在本公开的一些实施方案中,所述的通式(X)、(I)所示的化合物或其可药用的盐,其中Q2为碳原子且与J6连接,Q1、Q3、Q4和Q5均为CRQ,RQ如通式(I)中所定义;在一些实施方案中,Q2为碳原子且与J6连接,Q1、Q3、Q4和Q5相同或不同,且各自独立地为CH或CF;优选地,Q2为碳原子且与J6连接,Q1、Q3、Q4和Q5均为CH。
在本公开的一些实施方案中,所述的通式(X)所示的化合物或其可药用的盐,其中Q1或Q2中的一者碳原子且与J6连接,另一者以及Q3和Q4均为CH或CRq,Rq如通式(X)中所定义;在一些实施方案中,Q2为碳原子且与J6连接,Q1、Q3和Q4均为CH或CRq,Rq如通式(X)中所定义;在一些实施方案中,Q2为碳原子且与J6连接,Q1、Q3和Q4相同或不同,且各自独立地为CH或CF。
在本公开的一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中各个RQ相同或不同,且各自独立地选自氢原子、卤素、C1-6烷基、C1-6烷氧基、C1-6卤代烷基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基和氰基;优选地,各个RQ均为氢原子。
在本公开的一些实施方案中,所述的通式(I)、(X)所示的化合物或其可药用的盐,其为通式(I’-1)、(I’-2)或(I’-3)所示的化合物或其可药用的盐,

其中,各个Rq相同或不同,且各自独立地选自卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、NRcRd、亚烷基NRcRd、C(O)烷基、C(O)NRcRd、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R04所取代;
n为0、1或2;s为0、1、2或3;
R1至R7、Ra、Rb、RZ1、RZ2、L1、L2、RA、Rc、Rd、R04、J1至J6、Y1和Y2如通式(I)中所定义。
在本公开的一些实施方案中,所述的通式(I)、(I’-3)、(X)所示的化合物或其可药用的盐,其为通式(I’-3-1)或(I’-3-2)所示的化合物或其可药用的盐,
其中,R1至R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1至J6、Rq、Y2和s如通式(I’-3)中所定义。
在本公开的一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(X)所示的化合物或其可药用的盐,其为通式(II)或(II’)所示的化合物或其可药用的盐,
其中,
各个Rq相同或不同,且各自独立地选自卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、NRcRd、亚烷基NRcRd、C(O)烷基、C(O)NRcRd、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R04所取代;
n为0、1或2;s为0、1、2或3;
v1为0、1、2、3、4、5或6;v2为0、1、2、3、4、5或6;
R1至R7、RA、Ra、Rb、Rc、Rd、RZ1、RZ2、L1、L2、J2、J6、R04和Y1如通式(I)中所定义。
本公开的一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(I’-3)、(I’-3-1)、(I’-3-2)、(II)、(II’)所示的化合物或其可药用的盐,其中, 环B选自环烷基、杂环基、芳基和杂芳基;各个RB相同或不同, 且各自独立地选自氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;t为0、1、2、3、4或5;带**的键与L1连接,R1、R2如通式(I)中所定义;
优选地,为单键或双键,X1和X2各自独立地选自CRb1、CRb1Rb2、N、NRb3、O和S,各个RB相同或不同,且各自独立地选自氧代基、卤素、C1-6烷基、C1-6烷氧基、C1-6卤代烷基、卤代C1-6烷氧基、氰基、C2-6烯基、C2-6炔基、3至10元环烷基和3至10元杂环基;
各个Rb1和Rb2相同或不同,且各自独立地选自氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;Rb3选自氢原子、烷基、卤代烷基、烯基、炔基、环烷基和杂环基;t为0、1、2或3;u为0、1或2,带**的键与L1连接,R1、R2如通式(I)中所定义;
更优选地,R1为-P(O)R10R11或-N(R15)SO2R16;R10、R11、R15和R16相同或不同,且各自独立地为氢原子或C1-6烷基;X1为CH、CRb1或N,各个RB和Rb1相同或不同,且各自独立地选自卤素、C1-6烷基、C1-6卤代烷基和3至6元环烷基;t为0、1或2;带**的键与L1连接;
进一步优选地,R1为-P(O)R10R11或-N(R15)SO2R16;R10、R11、R15和R16相同或不同,且各自独立地为氢原子或C1-6烷基;带**的键与L1连接;
最优选为带**的键与L1连接。
本公开的一些实施方案中,所述式(B2)中,环B选自5或6元杂环基、苯基和5或6元杂芳基;优选选自苯基、吡啶基和吡嗪基。
本公开的一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(I’-3)、(I’-3-1)、(I’-3-2)、(II)、(II’)所示的化合物或其可药用的盐,其中,R1和R2如通式(I)中所定义;在一些实施方案中,R1为-P(O)R10R11或-N(R15)SO2R16;R10、R11、R15和R16相同或不同,且各自独立地选自氢原子、C1-6烷基和3至6元环烷基;
在一些实施方案中,选自 在一些实施方案中,带**的键与L1连接。
本公开的一些实施方案中,所述式(B2)、(B3)中,各个Rb1和Rb2相同或不同,且各自独立地选自氢原子、卤素、C1-6烷基、C1-6烷氧基、C1-6卤代烷基、卤代C1-6烷氧基、氰基、C2-6烯基、C2-6炔基、3至10元环烷基和3至10元杂环基。
本公开的一些实施方案中,所述的通式(X)所示的化合物或其可药用的盐,其中y、R1和R9如通式(X)中所定义;在一些实施方案中,R9a为氢原子或R9,R1和R9如通式(X)中所定义;带**的键与L1连接。
本公开的一些实施方案中,所述的通式(X’)、(X’-1)、(X’-2)所示的化合物或其可药用的盐,其中R9a为氢原子或R9,R1和R9如通式(X)中所定义;带**的键与L1连接。
在本公开的一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(X)、(X’)所示的化合物或其可药用的盐,其为通式(III)或(III’)所示的化合物或其可药用的盐,
其中,
环J2’为含氮杂环基,所述含氮杂环基任选地被一个或多个R03所取代;
各个Rq相同或不同,且各自独立地选自卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、NRcRd、亚烷基NRcRd、C(O)烷基、C(O)NRcRd、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R04所取代;
n为0、1或2;s为0、1、2或3;
R1、R2、R4至R7、RA、Ra、Rb、Rc、Rd、RZ1、RZ2、L1、L2、J1、J5、J6、R03、R04和Y1如通式(I)中所定义。
在本公开的一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(X)、(X’)所示的化合物或其可药用的盐,其为通式(IV)或(IV’)所示的化合物或其可药用的盐,
其中,
环J6’为含氮杂环基,所述含氮杂环基任选地被一个或多个R03所取代;
各个Rq相同或不同,且各自独立地选自卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、NRcRd、亚烷基NRcRd、C(O)烷基、C(O)NRcRd、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R04所取代;
n为0、1或2;s为0、1、2或3;
p为0、1、2、3、4、5或6;
R1、R2、R4至R7、RA、Ra、Rb、Rc、Rd、RZ1、RZ2、L1、L2、J1、J2、R03、R04和Y1如通式(I)中所定义。
在本公开的一些实施方案中,所述的通式(I)、(I’-3)、(X)、(X’)、(X’-1)所示的化合物或其可药用的盐,其为通式(V-1)所示的化合物或其可药用的盐,
其中,环J2’为含氮杂环基;
q为0、1、2、3、4、5或6;j为0、1、2、3、4、5或6;
s为0、1、2或3;n为0、1或2;
R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、J1、R03、J4、J5、J6、Rq、和Y2如通式(I’-3)中所定义。
在本公开的一些实施方案中,所述的通式(I)、(I’-3)、(X)、(X’)、(X’-2)所示的化合物或其可药用的盐,其为通式(V-2)所示的化合物或其可药用的盐,
其中,环J3’为含氮杂环基;
q为0、1、2、3、4、5或6;j为0、1、2、3、4、5或6;
s为0、1、2或3;n为0、1或2;
R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、J1、R03、J4、J5、J6、Rq和Y2如通式(I’-3)中所定义。
在本公开的一些实施方案中,所述的通式(I)、(I’-3)、(I’-3-1)、(I’-3-2)、(V-1)、(X)、(X’)、(X’-1)所示的化合物或其可药用的盐,其为通式(V-1-1)或(V-1-2)所示的化合物或其可药用的盐,
其中,R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J2’、R03、j、q、J4、J5、J6、Rq、Y2和s如通式(V-1)中所定义。
在本公开的一些实施方案中,所述的通式(I)、(I’-3)、(I’-3-1)、(I’-3-2)、(V-2)、(X)、(X’)、(X’-2)所示的化合物或其可药用的盐,其为通式(V-2-1)或(V-2-2)所示的化合物或其可药用的盐,
其中,R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、q、环J3’、R03、j、J4、J5、J6、Rq、Y2和s如通式(V-2)中所定义。
在本公开的一些实施方案中,所述的通式(X)、(X’)、(X’-2)所示的化合物或其可药用的盐,其为通式(X’-3)所示的化合物或其可药用的盐,
其中,环J3’为含氮杂环基;j为0、1、2、3、4、5或6;
q为0、1、2、3、4、5或6;n为0、1或2;
R1、R9、y、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、J1、R03、J4、J5、J6、Rq、Y1和R7如通式(X)中所定义。
在本公开的一些实施方案中,所述的通式(I’-2)所示的化合物或其可药用的盐,其中选自Rq、R7和s如通式(I’-2)中所定义。
在本公开的一些实施方案中,所述的通式(III’)、(IV’)、(X’-3)所示的化合物或其可药用的盐,其中选自 Rq、R7和s如通式(X)中所定义;在一些实施方案中,选自
在一些实施方案中,选自
在本公开的一些实施方案中,所述的通式(X’-1)、(III)、(III’)、(V-1)、(V-1-1)、(V-1-2)所示的化合物或其可药用的盐,其中环J2’为3至12元含氮杂环基;在一些实施方案中,环J2’为5或6元至少含一个氮原子的单环杂环基或7至14元至少含一个氮原子的螺杂环基;在一些实施方案中,环J2’选自哌啶基、哌嗪基和二氮杂螺[3.5]壬烷。
在本公开的一些实施方案中,所述的通式(V-2)、(V-2-1)、(V-2-2)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中环J3’为3至12元含氮杂环基;在一些实施方案中,环J3’为3至10元含氮杂环基;在一些实施方案中,环J3’为哌啶基或哌嗪基;在一些实施方案中,环J3’选自
在本公开的一些实施方案中,所述的通式(V-1)、(V-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;和/或j为0、1或2。
在本公开的一些实施方案中,所述的通式(I)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)所示的化合物或其可药用 的盐,其中Y2为CH2或C(O);在一些实施方案中,Y2为CH2;在一些实施方案中,Y2为C(O)。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)、(III’)、(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中L1为键或氧原子;优选为氧原子。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)、(III’)、(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中L2为(CRL3RL4)n3,n3为0、1、2、3、4、5、6、7、8、9或10,RL3和RL4相同或不同,且各自独立地选自氢原子、卤素、C1-6烷基和C1-6卤代烷基;优选地,L2为键或C1-10亚烷基;进一步优选为C1-10亚烷基;更优选地,L2为(CH2)n3,n3为1、2、3、4、5、6、7或8;最优选选自(CH2)3、(CH2)4和(CH2)5
在本公开的一些实施方案中,所述的通式(X)、(I)所示的化合物或其可药用的盐,其中L3为键或C1-10亚烷基;优选为键。
在本公开的一些实施方案中,所述的通式(X)、(I)所示的化合物或其可药用的盐,其中L1为O,L2为C1-10亚烷基,L3为键;优选地,L1为O,L2选自(CH2)3、(CH2)4和(CH2)5,L3为键。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)、(III’)、(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中-L1-L2-为-O-C1-10亚烷基-;优选地,-L1-L2-为-O-(CH2)n3-;n3为1、2、3、4、5、6、7或8;更优选为-O-(CH2)3-、-O-(CH2)4-或-O-(CH2)5-。
本公开的一些实施方案中,所述的通式(I)至(IV)、(I’-1)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中,
在本公开一些实施方案中,所述的通式(I)、(III)、(IV)、(III’)、(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中J1选自键、O、S、-O-C1-6亚烷基-、-C1-6亚烷基-O-、C(O)、-C(O)-C1-6亚烷基-、-C1-6亚烷基-C(O)-、C(O)N(RJ)、N(RJ)C(O)、N(RJ)、-N(RJ)-C1-6亚烷基-、-C1-6亚烷基-N(RJ)-、C1-6亚烷基、C2-6烯基、C2-6炔基、3至12元环烷基、3至12元杂环基、-C1-6亚烷基-3至12元杂环基、-3至12元杂环基-C1-6亚烷基-、6至10元芳基和5至14元杂芳基; 所述的C1-6亚烷基、C2-6烯基、3至12元环烷基、3至12元杂环基、6至10元芳基和5至14元杂芳基各自独立地任选被1个或多个R03所取代,R03和RJ如通式(I)所定义;优选地,J1为键;
在一些实施方案中,J1为键或3至12元杂环基;所述的3至12元杂环基任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;在一些实施方案中,J1选自键、哌啶基和哌嗪基,所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;在一些实施方案中,J1选自键、哌啶基和哌嗪基。
在本公开一些实施方案中,所述的通式(I)、(II)、(II’)、(IV)、(IV’)、(I’-1)、(I’-2)、(I’-3)、(I’-3-1)、(I’-3-2)、(X)、(X’)所示的化合物或其可药用的盐,其中J2选自键、O、S、-O-C1-6亚烷基-、-C1-6亚烷基-O-、C(O)、-C(O)-C1-6亚烷基-、-C1-6亚烷基-C(O)-、C(O)N(RJ)、N(RJ)C(O)、N(RJ)、-N(RJ)-C1-6亚烷基-、-C1-6亚烷基-N(RJ)-、C1-6亚烷基、C2-6烯基、C2-6炔基、3至12元环烷基、3至12元杂环基、-C1-6亚烷基-3至12元杂环基、-3至12元杂环基-C1-6亚烷基-、6至10元芳基和5至14元杂芳基;所述的C1-6亚烷基、C2-6烯基、3至12元环烷基、3至12元杂环基、6至10元芳基和5至14元杂芳基各自独立地任选被1个或多个R03所取代,R03和RJ如通式(I)所定义;
优选地,J2为任选被一个或多个R03所取代的3至12元环烷基或3至12元杂环基,R03选自氧代基、羟基、卤素、C1-6烷基和C1-6卤代烷基;更优选地,J2为任选被一个或多个R03所取代的4至7元单环杂环基或7至14元螺杂环基,R03选自羟基、卤素、C1-6烷基和C1-6卤代烷基;进一步优选地,J2为任选被一个或多个R03所取代的5或6元杂环基,R03选自羟基、卤素、C1-6烷基和C1-6卤代烷基;最优选地,J2为任选被一个或多个R03所取代的1,4-哌啶基或1,4-哌嗪基,R03选自羟基、卤素、C1-6烷基和C1-6卤代烷基;
在一些实施方案中,J2选自 上述的环任选被1个或多个选自氧代基、卤素、羟基、C1-6烷基和C1-6卤代烷基的取代基所取代;优选地,J2选自更优选地,J2 在一些实施方案中,J2选自其中带*的键与J3连接;
在一些实施方案中,J2选自哌啶基、哌嗪基和二氮杂螺[3.5]壬烷;所述的哌啶基、哌嗪基和二氮杂螺[3.5]壬烷各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;在一些实施方案中,J2为哌啶基或哌嗪基;所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代。
在本公开一些实施方案中,所述的通式(III)、(III’)、(V-1)、(V-1-1)、(V-1-2)、(X’-1)所示的化合物或其可药用的盐,其中环J2’为任选被1或多个R03所取代的4至7元含氮单环杂环基或7至14元含氮螺杂环基,R03选自羟基、卤素、C1-6烷基和C1-6卤代烷基;优选地,环J2’为任选被1或多个R03所取代的5或6元含氮杂环基,R03选自羟基、卤素、C1-6烷基和C1-6卤代烷基;更优选地,环J2’为任选被1或多个R03所取代的1,4-哌啶基或1,4-哌嗪基,R03选自羟基、卤素、C1-6烷基和C1-6卤代烷基;
最优选为在一些实施方案中,环J2’选自 其中带*的键与C(O)连接。
在本公开一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(I’-3)、(I’-3-1)、(I’-3-2)、(X)、(X’)所示的化合物或其可药用的盐,其中J3选自键、O、S、-O-C1-6亚烷基-、-C1-6亚烷基-O-、C(O)、-C(O)-C1-6亚烷基-、-C1-6亚烷基-C(O)-、C(O)N(RJ)、N(RJ)C(O)、N(RJ)、-N(RJ)-C1-6亚烷基-、-C1-6亚烷基-N(RJ)-、C1-6亚烷基、C2-6烯基、C2-6炔基、3至12元环烷基、3至12元杂环基、-C1-6亚烷基-3至12元杂环基、-3至12元杂环基-C1-6亚烷基-、6至10元芳基和5至14元杂芳基;所述的C1-6亚烷基、C2-6烯基、3至12元环烷基、3至12元杂环基、6至10元芳基和5至14元杂芳基各自独立地任选被1个或多个R03所取代,R03和RJ如通式(I)所定义;优选地,J3为键或C1-6亚烷基;优选为键或CH2;在一些实施方案中,J3选自键、O、 -C(O)-C1-6亚烷基-、-C1-6亚烷基-C(O)-和N(RJ),RJ如通式(I)所定义;在一些实施方案中,J3选自键、O、-C(O)-CH2-、-CH2-C(O)-、-C(O)-CH(CH3)-和N(CH3)。
在本公开一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中J4选自键、O、S、-O-C1-6亚烷基-、-C1-6亚烷基-O-、C(O)、-C(O)-C1-6亚烷基-、-C1-6亚烷基-C(O)-、C(O)N(RJ)、N(RJ)C(O)、N(RJ)、-N(RJ)-C1-6亚烷基-、-C1-6亚烷基-N(RJ)-、C1-6亚烷基、C2-6烯基、C2-6炔基、3至12元环烷基、3至12元杂环基、-C1-6亚烷基-3至12元杂环基、-3至12元杂环基-C1-6亚烷基-、6至10元芳基和5至14元杂芳基;所述的C1-6亚烷基、C2-6烯基、3至12元环烷基、3至12元杂环基、6至10元芳基和5至14元杂芳基各自独立地任选被1个或多个R03所取代,R03和RJ如通式(I)所定义;优选地,J4为键或C(O);更优选地,J4为C(O);在一些实施方案中,J4为键。
在本公开一些实施方案中,所述的通式(I)、(III)、(III’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中J5选自键、O、S、-O-C1-6亚烷基-、-C1-6亚烷基-O-、C(O)、-C(O)-C1-6亚烷基-、-C1-6亚烷基-C(O)-、C(O)N(RJ)、N(RJ)C(O)、N(RJ)、-N(RJ)-C1-6亚烷基-、-C1-6亚烷基-N(RJ)-、C1-6亚烷基、C2-6烯基、C2-6炔基、3至12元环烷基、3至12元杂环基、-C1-6亚烷基-3至12元杂环基、-3至12元杂环基-C1-6亚烷基-、6至10元芳基和5至14元杂芳基;所述的C1-6亚烷基、C2-6烯基、3至12元环烷基、3至12元杂环基、6至10元芳基和5至14元杂芳基各自独立地任选被1个或多个R03所取代,R03和RJ如通式(I)所定义;优选地,J5为键或C1-6亚烷基;更优选为键或CH2;在一些实施方案中,J5为键或亚乙基;在一些实施方案中,J5为键。
在本公开一些实施方案中,所述的通式(I)、(II)、(III)、(II’)、(III’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中J6选自键、O、S、-O-C1-6亚烷基-、-C1-6亚烷基-O-、C(O)、-C(O)-C1-6亚烷基-、-C1-6亚烷基-C(O)-、C(O)N(RJ)、N(RJ)C(O)、N(RJ)、-N(RJ)-C1-6亚烷基-、-C1-6亚烷基-N(RJ)-、C1-6亚烷基、C2-6烯基、C2-6炔基、3至12元环烷基、3至12元杂环基、-C1-6亚烷基-3至12元杂环基、-3至12元杂环基-C1-6亚烷基-、6至10元芳基和5至14元杂芳基;所述的C1-6亚烷基、C2-6烯基、3至12元环烷基、3至12元杂环基、6至10元芳基和5至14元杂芳基各自独立地任选被1个或多个R03所取代,R03和RJ如通式(I)所定义;优选地,J6为任选被1个或多个R03’所取代的3至12元环烷基或3至12元杂环基,R03’选自氧代基、羟基、卤素、C1-6烷基和C1-6卤代烷基;更优选地,J6为任选被1或多个R03’所取代的5或6元杂环基,R03’选自羟基、卤素、C1-6烷 基和C1-6卤代烷基;进一步优选地,J6为任选被1或多个R03’所取代的1,4-哌啶基或1,4-哌嗪基,R03’选自羟基、卤素、C1-6烷基和C1-6卤代烷基;最优选选自 在一些实施方案中,J6选自键、 和N(RJ),RJ为氢原子或甲基;在一些实施方案中,J6选自键、 和NH,自左向右连接;
在一些实施方案中,J6选自键、 NH、
在一些实施方案中,J6为N(RJ)或3至12元杂环基,所述的3至12元杂环基任选被选自卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或C1-6烷基;在一些实施方案中,J6选自键、N(RJ)和3至12元杂环基,所述的3至12元杂环基任选被选自卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或C1-6烷基;
在一些实施方案中,J6选自键、N(RJ)、哌啶基和哌嗪基,所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代,RJ为氢原子或甲基;在一些实施方案中,J6选自N(RJ)、哌啶基和哌嗪基,所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代,RJ为氢原子或甲基;
在一些实施方案中,J6选自键、O、N(RJ)和C1-6亚烷基,RJ如通式(I)中所定义;在一些实施方案中,J6选自键、O、N(RJ)和(CH2)q,q为0、1、2、3或4,RJ为氢原子或C1-6烷基。
在本公开一些实施方案中,所述的通式(IV)或(IV’)所示的化合物或其可药用的盐,其中环J6’为任选被1或多个R03’所取代的5或6元含氮杂环基,R03’选自羟基、卤素、C1-6烷基和C1-6卤代烷基;更优选地,环J6’为任选被1或多个R03’所取代的1,4-哌啶基或1,4-哌嗪基,R03’选自羟基、卤素、C1-6烷基和C1-6卤代烷基;最优选 选自在一些实施方案中,环J6’选自带#的键与苯环连接。
在本公开一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(I’-3)、(I’-3-1)、(I’-3-2)所示的化合物或其可药用的盐,其中-J3-J4-J5-为-C(O)-C1-6亚烷基-或-C1-6亚烷基-C(O)-;优选为-CO-CH2-或-CH2-CO-。
在本公开的一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(I’-3)、(I’-3-1)、(I’-3-2)所示的化合物或其可药用的盐,其中-J1-J2-J3-J4-J5-J6-为J2和J6相同或不同,且各自独立地为环烷基或杂环基,所述的环烷基或杂环基各自独立地任选被一个或多个R03所取代;v1为0、1、2、3、4、5或6,v2为0、1、2、3、4、5或6,R03如通式(I)中所定义;优选地,-J1-J2-J3-J4-J5-J6-为J2和J6相同或不同,且各自独立地为1,4-哌啶基或1,4-哌嗪基,所述的1,4-哌啶基或1,4-哌嗪基各自独立地任选被一个或多个选自羟基、卤素、C1-6烷基和C1-6卤代烷基的取代基所取代;v1为0、1、2、3、4、5或6,v2为0、1、2、3、4、5或6;
最优选地,-J1-J2-J3-J4-J5-J6-选自
在本公开的一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中-J4-J5-J6-选自键、-C(O)-C1-6亚烷基-3至8元杂环基-、-C(O)-3至8元杂环基-、-3至12元杂环基-、-3至12元杂环基-C0-6亚烷基-3至12元杂环基-和-C(O)-C1-6亚烷基-N(RJ)-;RJ为氢原子或C1-6烷基;所述的3至8元杂环基和3至12元杂环基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或C1-6烷基;在一些实施方案中,-J4-J5-J6-选自键、-C(O)-C1-6亚烷基-哌啶基、哌啶基、哌嗪基和-C(O)-C1-6亚烷基-N(RJ)-;所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或甲基;
在一些实施方案中,-J4-J5-J6-选自-C(O)-C1-6亚烷基-3至8元杂环基-、-C(O)-3至8元杂环基-、-3至12元杂环基-、-3至12元杂环基-C0-6亚烷基-3至12元杂环基-和-C(O)-C1-6亚烷基-N(RJ)-;RJ为氢原子或C1-6烷基;所述的3至8元杂环基和3至12元杂环基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或C1-6烷基;在一些实施方案中,-J4-J5-J6-选自-C(O)-C1-6亚烷基-哌啶基、哌啶基、哌嗪基和-C(O)-C1-6亚烷基-N(RJ)-;所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或甲基;
在一些实施方案中,-J4-J5-J6-为键或任选被选自卤素、羟基和C1-6烷基中的一个或多个所取代的3至12元杂环基;在一些实施方案中,-J4-J5-J6-为键;在一些实施方案中,-J4-J5-J6-为任选被选自卤素、羟基和C1-6烷基中的一个或多个所取代的3至12元杂环基。
在本公开的一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(I’-3)、(I’-3-1)、(I’-3-2)、(X)、(X’)所示的化合物或其可药用的盐,其中-J1-J2-J3-J4-J5-J6-为X、Y、U和V相同或不同,且各自独立地为CR05或N;q、u和w相同或不同,且各自独立地为0、1、2、3、4、5或6;a、b、c、e和f相同或不同,且各自独立地为0、1、2或3;d为1、2或3;i为1、2或3;各个R05相同或不同,且各自独立地为氢原子或R03;J6和R03如通式(I)中所定义;
在一些实施方式中,-J1-J2-J3-J4-J5-J6-选自-杂环基-C(O)-C1-6亚烷基-杂环基-、-杂环基-C1-6亚烷基-C(O)-杂环基-、-杂环基-C1-6亚烷基-杂环基-、-杂环基-C0-6亚烷基-杂环基-C0-6亚烷基-杂环基-和-杂环基-C0-6亚烷基-杂环基-C(O)-C1-6亚烷基-N(RJ)-;所述的杂环基任选被一个或多个选自R03的取代基所取代;R03和RJ如通式(I)中所定义;
在一些实施方式中,-J1-J2-J3-J4-J5-J6-选自-3至8元杂环基-C(O)-C1-6亚烷基-3至8元杂环基-、-3至8元杂环基-C1-6亚烷基-C(O)-3至8元杂环基-、-3至12元杂环基-C1-6亚烷基-3至12元杂环基-、-3至12元杂环基-C0-6亚烷基-3至12元杂环基-C0-6亚烷基-3至12元杂环基-和-3至12元杂环基-C0-6亚烷基-3至12元杂环基-C(O)-C1-6亚烷基-N(RJ)-;所述的3至8元杂环基和3至12元杂环基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或C1-6烷基;
在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自 X、Y、U、V、S和T相同或不同,且各自独立地为CR05或N;
q、u、x、w、k和l相同或不同,且各自独立地为0、1、2、3、4、5或6;a、b、c、e和f相同或不同,且各自独立地为0、1、2或3;d为1、2或3;g和h相同或不同,且各自独立地为0、1或2;各个RJ2、RJ3和RJ5相同或不同,且各自独立地为氢原子或C1-6烷基;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;各个R05相同或不同,且各自独立地选自氢原子、卤素、羟基和C1-6烷基;
在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自 X、Y、U、V、S和T相同或不同,且各自独立地选自CH、CR03和N;u、q、w、x、k、l和q1相同或不同,且各自独立地为0、1、2、3、4、5或6;a、b、c、e和f相同或不同,且各自独立地为0、1、2或3;d为1、2或3;g和h相同或不同,且各自独立地为0、1或2;各个RJ3和RJ5相同或不同,且各自独立地为氢原子或R03;R03和RJ如通式(I)中所定义;
在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自 X、Y、U、V、S和T相同或不同,且各自独立地为CR05或N;
q、u、x、w、k和l相同或不同,且各自独立地为0、1、2、3、4、5或6;a、b、c、e和f相同或不同,且各自独立地为0、1、2或3;d为1、2或3;g和h相同或不同,且各自独立地为0、1或2;各个RJ3相同或不同,且各自独立地为氢原子或C1-6烷基;各个RJ5相同或不同,且各自独立地为氢原子或C1-6烷基;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;各个R05相同或不同,且各自独立地选自氢原子、卤素、羟基和C1-6烷基;
在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自 q、u和w相同或不同,且各自独立地为0、1、2、3、4、5或6;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;g和h各自独立地为0、1或2;q1为0、1、2、3、4、5或6;
在一些实施方案中,-J1-J2-J3-J4-J5-J6-为q、u和w相同或不同,且各自独立地为0、1、2、3、4、5或6;各个R03相同或不同,且各自独立地选自卤素、羟基和C1-6烷基;g和h各自独立地为0、1或2;
在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自 q、u、x和w相同或不同,且各自独立地为0、1、2、3、4、5或6;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;
在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自
在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自 在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自
在本公开的一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(I’-3)、(I’-3-1)、(I’-3-2)、(X)、(X’)所示的化合物或其可药用的盐,其中J1选自键、哌啶基和哌嗪基,所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷 基中的一个或多个所取代;J2为任选被一个或多个R03所取代的4至7元单环杂环基或7至14元螺杂环基,R03选自羟基、卤素、C1-6烷基和C1-6卤代烷基;J3为键或C1-6亚烷基;J4为键或C(O);J5为键或C1-6亚烷基;J6为N(RJ)或3至12元杂环基,所述的3至12元杂环基任选被选自卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或C1-6烷基;
在一些实施方案中,J1选自键、哌啶基和哌嗪基,所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;J2为任选被一个或多个R03所取代的4至7元单环杂环基或7至14元螺杂环基,R03选自羟基、卤素、C1-6烷基和C1-6卤代烷基;J3为键或C1-6亚烷基;J4为键或C(O);J5为键或C1-6亚烷基;J6选自键、N(RJ)和4至7元杂环基,所述的4至7元杂环基任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或甲基。
在本公开的一些实施方案中,所述的通式(V-1)、(V-1-1)、(V-1-2)、(X’-1)所示的化合物或其可药用的盐,其中选自 X、U、V、S和T相同或不同,且各自独立地为CR05或N;
q、u、x、w、k和l相同或不同,且各自独立地为0、1、2、3、4、5或6;a、b、c、e和f相同或不同,且各自独立地为0、1、2或3;d为1、2或3;g和h相同或不同,且各自独立地为0、1或2;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;各个R05相同或不同,且各自独立地选自氢原子、卤素、羟基和C1-6烷基;
在一些实施方案中,选自 q、u、x和w相同或不同,且各自独立地为0、1、2、3、4、5或6;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;k为0、1、2、3、4、5或6;
在一些实施方案中,选自
在一些实施方案中,选自
在一些实施方案中,选自
在本公开的一些实施方案中,所述的通式(V-2)、(V-2-1)、(V-2-2)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中选自 X、Y、V和T相同或不同,且各自独立地为CR05或N;
q、u、x、w、k和l相同或不同,且各自独立地为0、1、2、3、4、5或6;a、b、c、e和f相同或不同,且各自独立地为0、1、2或3;d为1、2或3;g和h相同或不同,且各自独立地为0、1或2;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;各个R05相同或不同,且各自独立地选自氢原子、卤素、羟基和C1-6烷基;
在一些实施方案中,选自 q、u、x和w相同或不同,且各自独立地为0、1、2、3、4、5或6;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;
在一些实施方案中,选自 q、u、x和w相同或不同,且各自独立地为0、1、2、3、4、5或6;g和h各自独立地为0、1或2;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;q1为1、2、3、4、5或6;
在一些实施方案中,选自
在一些实施方案中,选自
在一些实施方案中,选自
在本公开的一些实施方案中,a、b、e和f相同或不同,且各自独立地为0或1;和/或c为1或2;和/或d为1或2;和/或g为0或1;和/或h为0或1。
在本公开的一些实施方案中,u、x和w相同或不同,且各自独立地为0、1或2。
在本公开的一些实施方案中,i为2或3。
在本公开的一些实施方案中,X为N;和/或U与V中至少一者为N;和/或S与T中至少一者为N。
在本公开的一些实施方案中,各个R05相同或不同,且各自独立地为氢原子或羟基。
在本公开的一些实施方案中,q为0或1,且k为0或1。
在本公开的一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中q为1、2、3、4或5;在一些实施方案中,q为1、2或3;在一些实施方案中,q为1;在一些实施方案中,q为0。
在本公开的一些实施方案中,q1为1。
在本公开的一些实施方案中,其中RJ5为C1-6烷基;在一些实施方案中,RJ5为甲基,和/或l为1。
在本公开的一些实施方案中,其中RJ3为氢原子。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(B1)至(B5)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中R1为-P(O)R10R11或-N(R15)SO2R16;R10、R11、R15和R16如通式(I)中所定义;优选 地,R1为-P(O)R10R11或-N(R15)SO2R16;R10、R11、R15和R16相同或不同,且各自独立地为氢原子或C1-6烷基;更优选地,R1为-N(R15)SO2R16;R15和R16相同或不同,且各自独立地为氢原子或C1-6烷基;最优选地,R1为-N(CH3)SO2CH3
在一些实施方案中,R1为-P(O)R10R11或-N(R15)SO2R16;R10、R11、R15和R16相同或不同,且各自独立地选自氢原子、C1-6烷基和3至6元环烷基;在一些实施方案中,R1为-N(R15)SO2R16;R15和R16相同或不同,且各自独立地选自氢原子、C1-6烷基和3至6元环烷基;在一些实施方案中,R1为-N(R15)SO2R16;R15选自氢原子、C1-6烷基和3至6元环烷基;R16为C1-6烷基;在一些实施方案中,R1为-N(CH3)SO2CH3或-N(环丙基)SO2CH3;在一些实施方案中,R1选自-N(H)SO2CH3、-N(CH3)SO2CH3、-N(环丙基)SO2CH3和-P(O)(CH3)2
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)或(B1)、(I’-1)、(I’-2)、(I’-3)、(I’-3-1)、(I’-3-2)所示的化合物或其可药用的盐,其中R2和R3相同或不同,且各自独立地为氢原子或C1-6烷基;优选地,R2和R3均为氢原子。
在本公开的一些实施方案中,所述的通式(I)、(II)、(II’)、(I’-1)、(I’-2)、(I’-3)、(I’-3-1)、(I’-3-2)所示的化合物或其可药用的盐,其中R3与L1连接;具体的,R3为键且与L1连接。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中R4为氢原子或C1-6烷基;优选为氢原子。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中R5选自氢原子、卤素和C1-6烷基;在一些实施方案中,R5为卤素;在一些实施方案中,R5为溴原子;在一些实施方案中,R5为氯原子;在一些实施方案中,R5为卤素或C1-6卤代烷基;在一些实施方案中,R5选自Cl、Br或CF3
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中R4为氢原子,且R5为卤素或C1-6卤代烷基;在一些实施方案中,R4为氢原子,且R5为卤素。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中Ra和Rb相同或不同,且各自独立地选自氢原子或C1-6烷基;优选地,Ra和Rb均为氢原子。
在本公开的一些实施方案中,所述的通式(X)、(I)所示的化合物或其可药用的盐,其中Z1为CRZ1,RZ1如通式(I)中所定义;优选地,Z1为CRZ1,RZ1为氢原子或C1-6烷氧基;更优选地,Z1为CH或C-OCH3;在一些实施方案中,Z1为CRZ1,RZ1选自氢原子、C1-6烷氧基和氘代C1-6烷氧基;在一些实施方案中,Z1选自CH、C-OCH3和-OCD3
在一些实施方案中,所述的通式(X)、(I)所示的化合物或其可药用的盐,其中Z2为CRZ2,RZ2如通式(I)中所定义;优选地,Z2为CRZ2,RZ2为氢原子或卤素;更优选地,Z2为CH或C-F。
在本公开的一些实施方案中,所述的通式(X)、(I)所示的化合物或其可药用的盐,其中Z3为CH。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中RZ1选自氢原子、卤素、羟基、氰基、C1-6烷基、C1-6烷氧基、C1-6卤代烷基、C1-6卤代烷氧基、C1-6羟烷基、C1-6羟烷氧基、3至10元环烷基、3至10元环烷基氧基和3至10元杂环基氧基;优选地,RZ1选自氢原子、C1-6烷氧基、C1-6卤代烷氧基和C1-6羟烷氧基;更优选地,RZ1为氢原子或C1-6烷氧基;更优选为氢原子或甲氧基;在一些实施方案中,RZ1选自氢原子、C1-6烷氧基、C1-6卤代烷氧基、C1-6氘代烷氧基和C1-6羟烷氧基;在一些实施方案中,RZ1选自氢原子、甲氧基和氘代甲氧基。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中RZ2选自氢原子、卤素、羟基、氰基、C1-6烷基、C1-6烷氧基、C1-6卤代烷基、C1-6卤代烷氧基、C1-6羟烷基、C1-6羟烷氧基、3至10元环烷基、3至10元环烷基氧基和3至10元杂环基氧基;优选地,RZ2选自氢原子、卤素、C1-6烷基和C1-6卤代烷基;更优选地,RZ2为氢原子或卤素;最优选为氢原子或氟原子。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)所示的化合物或其可药用的盐,其中RZ3选自氢原子、卤素、C1-6烷基和C1-6烷氧基;优选为氢原子。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(I’-1)所示的化合物或其可药用的盐,其中R6为氢原子或C1-6烷基;优选为氢原子。
在本公开的一些实施方案中,所述的通式(I)、(II’)至(IV’)、(I’-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中R7选自氢原子、C1-6烷基和C1-6卤代烷基;优选地,R7为C1-6烷基;更优选为甲基。
在本公开的一些实施方案中,R8为C1-6烷基;优选为甲基。
在本公开的一些实施方案中,所述的通式(II)、(III)、(IV)、(II’)、(III’)、(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基、C1-6卤代烷基、C1-6烷氧基、C1-6卤代烷氧基、羟基、氰基和3至6元环烷基;在一些实施方案中,各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;在一些实施方案中,各个Rq相同或不同,且各自独立地为卤素;在一些实施方案中,Rq为F;
在一些实施方案中,各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;和/或s为0或1。
在本公开的一些实施方案中,所述的通式(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中各个R9相同或不同,且各自独立地选自卤素、C1-6烷基、C1-6卤代烷基、C1-6烷氧基、C1-6卤代烷氧基、羟基、氰基和3至6元环烷基;在一些实施方案中,各个R9相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;在一些实施方案中,各个R9相同或不同,且各自独立地为卤素;在一些实施方案中,R9为F;
在一些实施方案中,各个R9相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;和/或y为0或1。
在本公开的一些实施方案中,R9a选自氢原子、卤素、C1-6烷基和C1-6卤代烷基;在一些实施方案中,R9a为氢原子或卤素;在一些实施方案中,R9a为氢原子或F。
在本公开的一些实施方案中,所述的通式(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中y为0或1。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中R10、R11、R12、R13、R14、R15和R16相同或不同,且各自独立地选自氢原子、C1-6烷基和3至6元环烷基;优选地,R10、R11、R12、R13、R14、R15和R16相同或不同,且各自独立地为氢原子或C1-6烷基;更优选地,R10、R11、R12、R13、R14、R15和R16相同或不同,且各自独立地为氢原子或甲基。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(B1)至(B5)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中R15和R16相同或不同,且各自独立地选自氢原子、C1-6烷基和3至6元环烷基;在一些实施方案中,R15选自氢原子、C1-6烷基和3至6元环烷基;R16为C1-6烷基 或3至6元环烷基;在一些实施方案中,R15选自氢原子、甲基和环丙基,R16为甲基。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中R10、R11、R12、R13、R14、R15和R16相同或不同,且各自独立地选自氢原子、甲基、乙基和环丙基。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中R10、R11、R15和R16相同或不同,且各自独立地选自氢原子、甲基、乙基和环丙基;在一些实施方案中,R10、R11和R16为甲基,R15选自氢原子、甲基和环丙基。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中各个R20、R21、R22、R23、R24、R25、R26、RL和RL’相同或不同,且各自独立地为氢原子或C1-6烷基。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中各个R01、R02、R03和R04相同或不同,且各自独立地选自氧代基、卤素、羟基、氰基、C1-6烷基、C1-6烷氧基、C1-6卤代烷基、C1-6卤代烷氧基、C1-6羟烷基和3至6元环烷基;优选地,各个R01、R02、R03和R04相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基、C1-6烷基和C1-6卤代烷基;优选地,各个R03相同或不同,且各自独立地选自卤素、羟基和C1-6烷基;更优选地,各个R03相同或不同,且各自独立地为氟原子或羟基;在一些实施方案中,各个R03相同或不同,且各自独立地选自卤素、羟基、C1-6烷基和C1-6羟烷基;在一些实施方案中,各个R03相同或不同,且各自独立地选自F、羟基、甲基和羟甲基。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中各个RL1、RL2、 RL3、RL4、RL6和RL7相同或不同,且各自独立地为氢原子或C1-6烷基;在一些实施方案中,各个RL1、RL2、RL3、RL4、RL6和RL7均为氢原子。
在本公开的一些实施方案中,所述的通式(I)、(II’)、(III’)、(IV’)、(I’-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中Y1为CH或N;在一些实施方案中,Y1为N;在一些实施方案中,Y1为CH。
在本公开的一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中Y3为CH或N。
在本公开的一些实施方案中,所述的通式(I)、(II)、(III)、(IV)、(II’)、(III’)、(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中n为0、1或2,优选为0。
在本公开的一些实施方案中,所述的通式(II)、(III)、(IV)、(II’)、(III’)、(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中s为0、1或2,在一些实施方案中,s为0;在一些实施方案中,s为1。
在本公开的一些实施方案中,所述的通式(X)、(I)所示的化合物或其可药用的盐,其中m为1、2、3、4、5、6、7或8,优选为3、4、5或6。
在本公开的一些实施方案中,所述的通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中RL5为键,n1为0、1、2、3或4,n2为0、1、2、3或4。
在本公开的一些实施方案中,所述的通式(II)、(II’)所示的化合物或其可药用的盐,其中v1为0或1;且v2为0或1。
在本公开的一些实施方案中,所述的通式(IV)、(IV’)所示的化合物或其可药用的盐,其中p为1、2、3、4、5或6,优选为1。
在本公开的一些实施方案中,所述的通式(I)、(II)、(III)、(IV)、(II’)、(III’)、(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)所示的化合物或其可药用的盐,其中n3为3、4或5。
在本公开的一些实施方案中,所述的通式(X)、(X’)所示的化合物或其可药用的盐,其中s1为0、1或2。
在本公开的一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中R1为-N(R15)SO2R16;R15和R16相同或不同,且各自独立地选自氢原子、C1-6烷基和3至6元环烷基;R2为氢原子;R3与L1连接;L1为O,L2为(CH2)n3,n3为1、2、3、4、5、6、7或8;环A为n为0;L3为键;R4为氢原子; R5为卤素;Ra和Rb均为氢原子;Z1为CRZ1,RZ1为氢原子或C1-6烷氧基;Z2为CRZ2,RZ2为氢原子或卤素;Z3为CH;-J1-J2-J3-J4-J5-J6-选自 q、u、x和w相同或不同,且各自独立地为0、1、2、3、4、5或6;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;
E选自:Y1为N或CH;Y2为CH2或C(O);各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;R6为氢原子或C1-6烷基;R7为C1-6烷基;s为0、1、2或3。
在本公开的一些实施方案中,所述的通式(II)所示的化合物或其可药用的盐,其中R1为-N(R15)SO2R16;R15和R16相同或不同,且各自独立地为氢原子或C1-6烷基;R2为氢原子;R3与L1连接;L1为O,L2为C1-10亚烷基;R4为氢原子;R5为卤素;Ra和Rb均为氢原子;RZ1为氢原子或C1-6烷氧基;RZ2为氢原子或卤素;J2为任选被一个或多个R03所取代的1,4-哌啶基或1,4-哌嗪基,R03选自羟基、卤素、C1-6烷基和C1-6卤代烷基;J6为任选被1或多个R03’所取代的1,4-哌啶基或1,4-哌嗪基,R03’选自羟基、卤素、C1-6烷基和C1-6卤代烷基;v1为0、1、2、3、4、5或6,v2为0、1、2、3、4、5或6;R6为氢原子,s为0,n为0。
在本公开的一些实施方案中,所述的通式(II’)所示的化合物或其可药用的盐,其中R1为-N(R15)SO2R16;R15和R16相同或不同,且各自独立地为氢原子或C1-6烷基;R2为氢原子;R3与L1连接;L1为O,L2为C1-10亚烷基;R4为氢原子;R5为卤素;Ra和Rb均为氢原子;RZ1为氢原子或C1-6烷氧基;RZ2为氢原子或卤素;J2为任选被一个或多个R03所取代的1,4-哌啶基或1,4-哌嗪基,R03选自羟基、卤素、C1-6烷基和C1-6卤代烷基;J6为任选被1或多个R03’所取代的1,4-哌啶基或1,4-哌 嗪基,R03’选自羟基、卤素、C1-6烷基和C1-6卤代烷基;v1为0、1、2、3、4、5或6,v2为0、1、2、3、4、5或6;R7为C1-6烷基;Y1为N;s为0,n为0。
在本公开的一些实施方案中,所述的通式(V-1)所示的化合物或其可药用的盐,其中R1为-N(R15)SO2R16;R15和R16相同或不同,且各自独立地选自氢原子、C1-6烷基和3至6元环烷基;R2为氢原子;R4为氢原子;R5为卤素;Ra和Rb均为氢原子;RZ1为氢原子或C1-6烷氧基;RZ2为氢原子或卤素;L1为O,L2为(CH2)n3,n3为1、2、3、4、5、6、7或8;n为0;J1选自键、哌啶基和哌嗪基,所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;环J2’选自哌啶基、哌嗪基和二氮杂螺[3.5]壬烷;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;和/或j为0、1或2;q为1;-J4-J5-J6-选自-C(O)-C1-6亚烷基-哌啶基、哌啶基、哌嗪基和-C(O)-C1-6亚烷基-N(RJ)-;所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或甲基;各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;s为0、1或2;Y2为CH2或C(O)。
在本公开的一些实施方案中,所述的通式(V-2)所示的化合物或其可药用的盐,其中R1为-N(R15)SO2R16;R15和R16相同或不同,且各自独立地选自氢原子、C1-6烷基和3至6元环烷基;R2为氢原子;R4为氢原子;R5为卤素;Ra和Rb均为氢原子;RZ1为氢原子或C1-6烷氧基;RZ2为氢原子或卤素;L1为O,L2为(CH2)n3,n3为1、2、3、4、5、6、7或8;n为0;J1选自键、哌啶基和哌嗪基,所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;q为1;环J3’为哌啶基或哌嗪基;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;和/或j为0、1或2;-J4-J5-J6-选自-C(O)-C1-6亚烷基-哌啶基、哌啶基、哌嗪基和-C(O)-C1-6亚烷基-N(RJ)-;所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或甲基;各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;Y2为CH2或C(O);s为0、1或2。
在本公开的一些实施方案中,所述的通式(V-2)、(V-2-1)或(V-2-2)所示的化合物或其可药用的盐,其中R1为-P(O)R10R11或-N(R15)SO2R16;R10、R11、R15和R16相同或不同,且各自独立地选自氢原子、甲基、乙基和环丙基;
R2为氢原子;R4为氢原子;R5为卤素;Ra和Rb均为氢原子;RZ1选自氢原子、甲氧基和氘代甲氧基;RZ2为氢原子或卤素;L1为O,L2选自(CH2)3、(CH2)4和(CH2)5;n为0;各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;Y2为CH2或C(O);s为0、1或2;
选自
在本公开的一些实施方案中,所述的通式(V-1)、(V-2)、(V-1-1)、(V-1-2)、(V-2-1)或(V-2-2)所示的化合物或其可药用的盐,其中R1为-P(O)R10R11或-N(R15)SO2R16;R10、R11、R15和R16相同或不同,且各自独立地选自氢原子、甲基、乙基和环丙基;
R2为氢原子;R4为氢原子;R5为卤素;Ra和Rb均为氢原子;RZ1选自氢原子、甲氧基和氘代甲氧基;RZ2为氢原子或卤素;L1为O,L2选自(CH2)3、(CH2)4和(CH2)5;n为0;J1选自键、哌啶基和哌嗪基,所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;q为1;环J2’选自哌啶基、哌嗪基和二氮杂螺[3.5]壬烷;环J3’为哌啶基或哌嗪基;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;j为0、1或2;-J4-J5-J6-选自键、-C(O)-C1-6亚烷基-哌啶基、哌啶基、哌嗪基和-C(O)-C1-6亚烷基-N(RJ)-;所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或甲基;各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;Y2为CH2或C(O);s为0、1或2。
在本公开的一些实施方案中,所述的通式(X’)所示的化合物或其可药用的盐,其中R1为-P(O)R10R11或-N(R15)SO2R16;R10、R11、R15和R16相同或不同,且各自独立地选自氢原子、甲基、乙基和环丙基;各个R9相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;y为0或1;Ra和Rb均为氢原子;R4为氢原子;R5为卤素;RZ1选自氢原子、甲氧基和氘代甲氧基;RZ2为氢原子或卤素;L1为O,L2选自(CH2)3、(CH2)4和(CH2)5;n为0;
-J1-J2-J3-J4-J5-J6-选自 E选自:
在本公开的一些实施方案中,所述的通式(X’)所示的化合物或其可药用的盐,其中R1为-P(O)R10R11或-N(R15)SO2R16;R10、R11、R15和R16相同或不同,且各自独立地选自氢原子、甲基、乙基和环丙基;各个R9相同或不同,且各自独立地为卤素;y为0或1;Ra和Rb均为氢原子;R4为氢原子;R5为卤素;RZ1选自氢原 子、甲氧基和氘代甲氧基;RZ2为氢原子或卤素;L1为O,L2选自(CH2)3、(CH2)4和(CH2)5;n为0;
-J1-J2-J3-J4-J5-J6-选自 q、u和w相同或不同,且各自独立地为0、1、2、3、4、5或6;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;g和h各自独立地为0、1或2;q1为0、1、2、3、4、5或6;
E选自:Y1为N或CH;Y2为CH2或C(O);各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;R6为氢原子,R7为C1-6烷基,R8为C1-6烷基;s为0、1、2或3。
在本公开的一些实施方案中,所述的通式(X’)所示的化合物或其可药用的盐,其中R1为-P(O)R10R11或-N(R15)SO2R16;R10、R11、R15和R16相同或不同,且各自独立地选自氢原子、C1-6烷基和3至6元环烷基;各个R9相同或不同,且各自独立地为卤素;y为0或1;Ra和Rb均为氢原子;R4为氢原子,且R5为卤素或C1-6卤代烷基;RZ1选自氢原子、甲氧基和氘代甲氧基;RZ2为氢原子或卤素;-L1-L2-为-O-C1-10亚烷基-;n为0;-J1-J2-J3-J4-J5-J6-选自 E选自: s为0、1、2或3,各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;R6为氢原子;R7为C1-6烷基;R8为C1-6烷基;Y1为CH或N;Y2为CH2或C(O)。
表A本公开的典型化合物包括但不限于:




















本公开的另一方面涉及一种通式(Xa)所示的化合物或其盐:
其中,
RJ’选自氢原子、羟基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、C(O)ORaa、-亚烷基-C(O)ORaa、NRbbRcc、-亚烷基-NRbbRcc、-C(O)Raa、-亚烷基-C(O)Raa、S(O)ORaa和S(O)2ORaa
Raa各自独立地选自氢原子、烷基、卤代烷基、卤代烷氧基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;Rbb和Rcc各自独立地选自氢原子、烷基、环烷基、杂环基和氨基保护基,所述氨基保护基优选为Boc;
R1、R9、y、R4、R5、Ra、Rb、Z1、Z2、Z3、L1、L2、环A、RA、n、L3、J1和J2如通式(X)中所定义。
本公开的另一方面涉及一种通式(Ia)所示的化合物或其盐:
其中,
RJ’选自氢原子、羟基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、C(O)ORaa、-亚烷基-C(O)ORaa、NRbbRcc、-亚烷基-NRbbRcc、-C(O)Raa、-亚烷基-C(O)Raa、S(O)ORaa和S(O)2ORaa
Raa各自独立地选自氢原子、烷基、卤代烷基、卤代烷氧基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;Rbb和Rcc各自独立地选自氢原子、烷基、环烷基、杂环基和氨基保护基,所述氨基保护基优选为Boc;
环A、R1至R5、RA、Ra、Rb、Z1、Z2、Z3、L1、L2、L3、J1、J2和n如通式(I)中所定义。
本公开的一些实施方案中,所述通式(Xa)、(Ia)所示的化合物或其盐,其中RJ’选自氢原子、C(O)ORaa、-C1-6亚烷基-C(O)ORaa、-C(O)Raa和-C1-6亚烷基-C(O)Raa,Raa为氢原子或C1-6烷基;在一些实施方案中,RJ’选自氢原子、C(O)O叔丁基、-CH2-C(O)O叔丁基、-CH2-C(O)OH、-C(O)H和-CH2-C(O)H。
本公开的另一方面涉及通式(I’-3A)、(I’-3-1A)或(I’-3-2A)所示的化合物或其盐:
其中,RL选自-OH、卤素和-O-烷基;
R1至R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1至J6、Rq、Y2和s如通式(I’-3)、(I’-3-1)或(I’-3-2)中所定义。
本公开的另一方面涉及通式(IIIa)或(IVa)所示的化合物或其盐:
其中,
RW为氢原子或氨基保护基,所述氨基保护基优选为Boc;
环J2’为含氮杂环基,所述含氮杂环基任选地被一个或多个R03所取代;
n为0、1或2;p为0、1、2、3、4、5或6;
R1、R2、R4、R5、RA、Ra、Rb、RZ1、RZ2、L1、L2、J1和J2如通式(I)中所定义。
本公开的另一方面涉及通式(V-1A)所示的化合物或其盐:
其中,RW为氢原子或氨基保护基,所述氨基保护基优选为Boc;
环J2’为含氮杂环基;
j为0、1、2、3、4、5或6;
R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1和R03如通式(V-1)中所定义。
本公开的另一方面涉及通式(V-2A)所示的化合物或其盐:
其中,q-1为0、1、2、3、4或5;
R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n和J1如通式(V-2)中所定义。
本公开的另一方面涉及通式(V-1A)、(V-1-1A)或(V-1-2A)所示的化合物或其盐:
其中,RL选自-OH、卤素和-O-烷基;
R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J2’、R03、j、q、J4、J5、J6、Rq、Y2和s如通式(V-1)、(V-1-1)或(V-1-2)中所定义。
本公开的另一方面涉及通式(V-2A)、(V-2-1A)或(V-2-2A)所示的化合物或其盐:

其中,RL选自-OH、卤素和-O-烷基;
R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、q、环J3’、R03、j、J4、J5、J6、Rq、Y2和s如通式(V-2)、(V-2-1)或(V-2-2)中所定义。
本公开的另一方面涉及一种通式(X’-1a)或(X’-2a)所示的化合物或其盐:
其中,环J2’为含氮杂环基;j为0、1、2、3、4、5或6;
q-1为0、1、2、3、4或5;n为0、1或2;
R1、R9、y、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、J1和R03如通式(X)中所定义。
在本公开的一些实施方案中,所述的通式(I’-3A)、(I’-3-1A)、(I’-3-2A)、(V-1A)、(V-1-1A)、(V-1-2A)、(V-2A)、(V-2-1A)、(V-2-2A)所示的化合物或其可药用的盐,其中RL为-O-C1-6烷基;在一些实施方案中,RL为叔丁氧基。
表B本公开的典型中间体化合物或其盐包括但不限于:













本公开另一方面涉及一种制备上述通式(I’-3-1)或(I’-3-2)所示的化合物或其可药用的盐的方法,该方法包括:

通式(I’-3-1A)所示的化合物或其盐发生分子内关环反应得到通式(I’-3-1)所示的化合物或其可药用的盐;
通式(I’-3-2A)所示的化合物或其盐发生分子内关环反应得到通式(I’-3-2)所示的化合物或其可药用的盐;
其中,RL选自-OH、卤素和-O-烷基;
R1至R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1至J6、Rq、Y2和s如通式(I’-3-1)或(I’-3-2)中所定义。
本公开另一方面涉及一种制备上述通式(I’-3-1)和(I’-3-2)所示的化合物或其可药用的盐的方法,该方法包括:
通式(I’-3)所示的化合物或其盐经拆分得到通式(I’-3-1)和(I’-3-2)所示的化合物或其可药用的盐;
其中,R1至R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1至J6、Rq、Y2和s如通式(I’-3-1)或(I’-3-2)中所定义。
本公开另一方面涉及一种制备上述通式(III)或(III’)所示的化合物或其可药用的盐的方法,该方法包括:

通式(IIIa)所示的化合物或其盐(优选盐酸盐)与通式(IIIb)所示的化合物或其盐(优选盐酸盐),发生缩合反应得到通式(III)所示的化合物或其可药用的盐;
通式(IIIa)所示的化合物或其盐(优选盐酸盐)与通式(III’b)所示的化合物或其盐(优选盐酸盐),发生缩合反应得到通式(III’)所示的化合物或其可药用的盐;
其中,RW为氢原子;
环J2’、R1、R2、R4至R7、RA、Ra、Rb、RZ1、RZ2、L1、L2、J1、J5、J6、Rq、s和n如通式(III)中所定义,Y1如通式(III’)中所定义。
本公开另一方面涉及一种制备上述通式(IV)或(IV’)所示的化合物或其可药用的盐的方法,该方法包括:

通式(IVa)所示的化合物或其盐(优选盐酸盐)与通式(IVb)所示的化合物或其盐(优选盐酸盐),发生缩合反应得到通式(IV)所示的化合物或其可药用的盐;
通式(IVa)所示的化合物或其盐(优选盐酸盐)与通式(IV’b)所示的化合物或其盐(优选盐酸盐),发生缩合反应得到通式(IV’)所示的化合物或其可药用的盐;
其中,环J6’、R1、R2、R4至R7、RA、Ra、Rb、RZ1、RZ2、L1、L2、J1、J2、Rq、p、s和n如通式(IV)中所定义,Y1如通式(IV’)中所定义。
本公开另一方面涉及一种制备上述通式(V-1)所示的化合物或其可药用的盐的方法,该方法包括:
通式(V-1A)所示的化合物或其盐(优选三氟乙酸盐或盐酸盐)与通式(V-1B)所示的化合物或其盐发生还原胺化反应得到通式(V-1)所示的化合物或其可药用的盐;
其中,RW为氢原子;
R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J2’、R03、j、q、J4、J5、J6、Rq、Y2和s如通式(V-1)中所定义。
本公开另一方面涉及一种制备上述通式(V-2)所示的化合物或其可药用的盐的方法,该方法包括:
通式(V-2A)所示的化合物或其盐与通式(V-2B)所示的化合物或其盐(优选盐酸盐)发生还原胺化反应得到通式(V-2)所示的化合物或其可药用的盐;
其中,R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、q、环J3’、R03、j、J4、J5、J6、Rq、Y2和s如通式(V-2)中所定义。
本公开另一方面涉及一种制备上述通式(V-1-1)或(V-1-2)所示的化合物或其可药用的盐的方法,该方法包括:

通式(V-1-1A)所示的化合物或其盐发生分子内关环反应得到通式(V-1-1)所示的化合物或其可药用的盐;
通式(V-1-2A)所示的化合物或其盐发生分子内关环反应得到通式(V-1-2)所示的化合物或其可药用的盐;
其中,RL选自-OH、卤素和-O-烷基;
R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J2’、R03、j、q、J4、J5、J6、Rq、Y2和s如通式(V-1-1)或(V-1-2)中所定义。
本公开另一方面涉及一种制备上述通式(V-1-1)和(V-1-2)所示的化合物或其可药用的盐的方法,该方法包括:
通式(V-1)所示的化合物或其盐经拆分得到通式(V-1-1)和(V-1-2)所示的化合物或其可药用的盐;
其中,R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J2’、R03、j、q、J4、J5、J6、Rq、Y2和s如通式(V-1-1)或(V-1-2)中所定义。
本公开另一方面涉及一种制备上述通式(V-2-1)或(V-2-2)所示的化合物或其可药用的盐的方法,该方法包括:
通式(V-2-1A)所示的化合物或其盐发生分子内关环反应得到通式(V-2-1)所示的化合物或其可药用的盐;
通式(V-2-2A)所示的化合物或其盐发生分子内关环反应得到通式(V-2-2)所示的化合物或其可药用的盐;
其中,RL选自-OH、卤素和-O-烷基;
R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、q、环J3’、R03、j、J4、J5、J6、Rq、Y2和s如通式(V-2-1)或(V-2-2)中所定义。
本公开另一方面涉及一种制备上述通式(V-2-1)和(V-2-2)所示的化合物或其可药用的盐的方法,该方法包括:
通式(V-2)所示的化合物或其盐经拆分得到通式(V-2-1)和(V-2-2)所示的化合物或其可药用的盐;
其中,R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、q、环J3’、R03、j、J4、J5、J6、Rq、Y2和s如通式(V-2-1)或(V-2-2)中所定义。
本公开另一方面涉及一种制备上述通式(X’-1)所示的化合物或其可药用的盐的方法,该方法包括:
通式(X’-1a)所示的化合物或其盐(优选三氟乙酸盐或盐酸盐,更优选为盐酸盐)与通式(X’-1b)所示的化合物或其盐发生还原胺化反应得到通式(X’-1)所示的化合物或其可药用的盐;其中,
q为1、2、3、4、5或6;q-1为0、1、2、3、4或5;
R1、R9、y、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J2’、R03、j、J4、J5、J6和E如通式(X’-1)中所定义。
本公开另一方面涉及一种制备上述通式(X’-2)所示的化合物或其可药用的盐的方法,该方法包括:
通式(X’-2a)所示的化合物或其盐与通式(X’-2b)所示的化合物或其盐(优选盐酸盐)发生还原胺化反应得到通式(X’-2)所示的化合物或其可药用的盐;其中,
q为1、2、3、4、5或6;q-1为0、1、2、3、4或5;
R1、R9、y、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J3’、R03、j、J4、J5、J6和E如通式(X’-2)中所定义。
本公开另一方面涉及一种制备上述通式(X’-3)所示的化合物或其可药用的盐的方法,该方法包括:
通式(X’-2a)所示的化合物或其盐与通式(X’-3b)所示的化合物或其盐(优选盐酸盐)发生还原胺化反应得到通式(X’-3)所示的化合物或其可药用的盐;其中,
q为1、2、3、4、5或6;q-1为0、1、2、3、4或5;
R1、R9、y、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J3’、R03、j、J4、J5、J6、Y1、R7、Rq和s如通式(X’-3)中所定义。
在本公开的一些实施方案中,所述的通式(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)所示的化合物或其可药用的盐的制备方法,其中RL为-O-C1-6烷基;在一些实施方案中,RL为叔丁氧基。
在本公开的一些实施方案中,所述的通式(V-1-1)、(V-1-2)所示的化合物或其可药用的盐的制备方法,其中当所述制备方法采用还原胺化反应时,q-1为0、1、2、3、4或5,且q为1、2、3、4、5或6;在一些实施方案中,q-1为0,且q为1。
本公开的另一方面涉及一种药物组合物,所述药物组合物含有本公开通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)以及表A所示的化合物或其可药用的盐,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
本公开进一步涉及通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)以及表A所示的化合物或其可药用的盐,或包含其的药物组合物在制备用于调节EGFR泛素化和降解的药物中的用途。
本公开进一步涉及通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)以及表A所示的化合物或其可药用的盐,或包含其的药物组合物在制备用于治疗和/或预防由EGFR介导的或依赖性的疾病或病症的药物中的用途。
本公开进一步涉及通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)以及表A所示的化合物或其可药用的盐,或包含其的药物组合物在制备EGFR抑制剂和/或降解剂的用途。
本公开进一步涉及通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)以及表A所示的化合物或其可药用的盐,或包含其的药物组合物在制备用于治疗和/或预防癌症的药物中的用途;优选地,所述的癌症选自鳞状细胞癌、基底细胞癌、腺癌、肝癌、肾癌、膀胱癌、乳腺癌、宫颈癌、结直肠癌、食管癌、头颈癌、鼻咽癌、口腔癌、唾液腺癌、肾癌、肺癌、卵巢癌、胰腺癌、前列腺癌、胃癌、白血病、淋巴瘤、神经胶质瘤、神经母细胞瘤、黑素瘤、肉瘤、子宫内膜癌、睾丸癌和甲状腺癌;进一步优选为肺癌;更优选为非小细胞肺癌。
本公开进一步涉及通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)以及表A所示的化合物或其可药用的盐,或包含其的药物组合物在制备用于治疗和/或预防疾病或病症的药物中的用途;优选地,所述疾病或病症为癌症或肿瘤;优选地,所述的癌症选自鳞状细胞癌、基底细胞癌、腺癌、肝癌、肾癌、膀胱癌、乳腺癌、宫颈癌、结直肠癌、食管癌、头颈癌、鼻咽癌、口腔癌、唾液腺癌、肾癌、肺癌、卵巢癌、胰腺癌、前列腺癌、胃癌、白血病、淋巴瘤、神经胶质瘤、神经母细胞瘤、黑素瘤、肉瘤、子宫内膜癌、睾丸癌和甲状腺癌;进一步优选为肺癌;更优选为非小细胞肺癌。
本公开还涉及一种调节受试者体内的EGFR蛋白泛素化和降解的方法,其包括给予所需患者治疗有效量的上述通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)或表A所示的化合物或其可药用的盐,或包含其的药物组合物。
本公开还涉及一种治疗和/或预防由EGFR介导的或依赖性的疾病或病症的方法,其包括给予所需患者治疗有效量的上述通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)或表A所示的化合物或其可药用的盐,或包含其的药物组合物。
本公开还涉及一种抑制和/或降解EGFR蛋白的方法,其包括给予所需患者治疗有效量的上述通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)或表A所示的化合物或其可药用的盐,或包含其的药物组合物。
本公开还涉及一种治疗和/或预防癌症的方法,其包括给予所需患者治疗有效量的上述通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)或表A所示的化合物或其可药用的盐,或包含其的药物组合物;优选地,所述的癌症选自鳞状细胞癌、基底细胞癌、腺癌、肝癌、肾癌、膀胱癌、乳腺癌、宫颈癌、结直肠癌、食管癌、头颈癌、鼻咽癌、口腔癌、唾液腺癌、肾癌、肺癌、卵巢癌、胰腺癌、前列腺癌、胃癌、白血病、淋巴瘤、神经胶质瘤、神经母细胞瘤、黑素瘤、肉瘤、子宫内膜癌、睾丸癌和甲状腺癌;进一步优选为肺癌;更优选为非小细胞肺癌。
本公开进一步涉及一种上述通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)或表A所示的化合物或其可药用的盐,或包含其的药物组合物,其用作药物。
本公开进一步涉及一种上述通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)或表A所示的化合物或其可药用的盐,或包含其的药物组合物,其用作调节受试者体内的EGFR蛋白泛素化和降解的药物。
本公开进一步涉及一种上述通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)或表A所示的化合物或其可药用的盐,或包含其的药物组合物,其用作EGFR抑制剂和/或降解剂。
本公开进一步涉及一种上述通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)或表A所示的化合物或其可药用的盐,或包含其的药物组合物,其用作抑制EGFR活性和/或降解EGFR蛋白的药物。
本公开进一步涉及一种上述通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)或表A所示的化合物或其可药用的盐,或包含其的药物组合物,其用作治疗和/或预防由EGFR介导的或依赖性的疾病或病症的药物。
本公开进一步涉及上述通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)或表A所示的化合物或其可药用的盐,或包含其的药物组合物,其用于调节受试者体内的EGFR蛋白泛素化和降解。
本公开进一步涉及上述通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、 (X’-2)、(X’-3)或表A所示的化合物或其可药用的盐,或包含其的药物组合物,其用于抑制EGFR活性和/或降解EGFR蛋白。
本公开进一步涉及上述通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)或表A所示的化合物或其可药用的盐,或包含其的药物组合物,其用于治疗和/或预防由EGFR介导的或依赖性的疾病或病症。
本公开进一步涉及一种上述通式(I)至(IV)、(II’)至(IV’)、(I’-1)、(I’-2)、(I’-3)、(V-1)、(V-2)、(I’-3-1)、(I’-3-2)、(V-1-1)、(V-1-2)、(V-2-1)、(V-2-2)、(X)、(X’)、(X’-1)、(X’-2)、(X’-3)或表A所示的化合物或其可药用的盐,或包含其的药物组合物,其用于治疗和/或预防癌症;优选地,所述的癌症选自鳞状细胞癌、基底细胞癌、腺癌、肝癌、肾癌、膀胱癌、乳腺癌、宫颈癌、结直肠癌、食管癌、头颈癌、鼻咽癌、口腔癌、唾液腺癌、肾癌、肺癌、卵巢癌、胰腺癌、前列腺癌、胃癌、白血病、淋巴瘤、神经胶质瘤、神经母细胞瘤、黑素瘤、肉瘤、子宫内膜癌、睾丸癌和甲状腺癌;进一步优选为肺癌;更优选为非小细胞肺癌。
优选地,本公开中所述的由EGFR介导的或依赖性的疾病或病症为癌症;所述的疾病或病症优选选自鳞状细胞癌、基底细胞癌、腺癌、肝癌、肾癌、膀胱癌、乳腺癌、宫颈癌、结直肠癌、食管癌、头颈癌、鼻咽癌、口腔癌、唾液腺癌、肾癌、肺癌、卵巢癌、胰腺癌、前列腺癌、胃癌、白血病、淋巴瘤、神经胶质瘤、神经母细胞瘤、黑素瘤、肉瘤、子宫内膜癌、睾丸癌和甲状腺癌;进一步优选为肺癌;更优选为非小细胞肺癌。
本公开中的肺癌包括小细胞肺癌和非小细胞肺癌;优选为非小细胞肺癌。
本公开所述的癌症优选为具有L858R突变的EGFR蛋白。
本公开所述的癌症优选为具有19del突变的EGFR蛋白。
本公开所述的癌症优选为具有T790M突变的EGFR蛋白。
本公开所述的癌症优选为具有C797X突变的EGFR蛋白。
本公开所述的癌症优选为具有L858R和T790M突变的EGFR蛋白。
本公开所述的癌症优选为具有19del和T790M突变的EGFR蛋白。
本公开所述的癌症优选为具有L858R和C797X突变的EGFR蛋白。
本公开所述的癌症优选为具有19del和C797X突变的EGFR蛋白。
本公开所述的癌症优选为具有T790M和C797X突变的EGFR蛋白。
本公开所述的癌症优选为具有L858R、T790M和C797X突变的EGFR蛋白。
本公开所述的癌症优选为具有19del、T790M和C797X突变的EGFR蛋白。
本公开所述的C797X突变优选为C797S突变;其中X代表包括S的任何氨基酸。
可将活性化合物制成适合于通过任何适当途径给药的形式,活性化合物优选是以单位剂量的方式,或者是以患者可以以单剂自我给药的方式。本公开化合物或组合物的单位剂量的表达方式可以是片剂、胶囊、扁囊剂、瓶装药水、药粉、颗粒剂、锭剂、栓剂、再生药粉或液体制剂。
作为一般性指导,合适的单位剂量可以是0.1~1000mg。
本公开的药物组合物除活性化合物外,可含有一种或多种辅料,所述辅料选自以下成分:填充剂(稀释剂)、粘合剂、润湿剂、崩解剂或赋形剂等。根据给药方法的不同,组合物可含有0.1至99重量%的活性化合物。
在一些实施方案中,所述的药物组合物的单位剂量为0.001mg-1000mg。
在某些实施方案中,基于组合物的总重量,所述的药物组合物含有0.01-99.99%的前述化合物或其可药用的盐或其同位素取代物。在某些实施方案中,所述的药物组合物含有0.1-99.9%的前述化合物或其可药用的盐或其同位素取代物。在某些实施方案中,所述的药物组合物含有0.5%-99.5%的前述化合物或其可药用的盐或其同位素取代物。在某些实施方案中,所述的药物组合物含有1%-99%的前述化合物或其可药用的盐或其同位素取代物。在某些实施方案中,所述的药物组合物含有2%-98%的前述化合物或其可药用的盐或其同位素取代物。
在某些实施方案中,基于组合物的总重量,所述的药物组合物含有0.01%-99.99%的药学上可接受的赋形剂。在某些实施方案中,所述的药物组合物含有0.1%-99.9%的药学上可接受的赋形剂。在某些实施方案中,所述的药物组合物含有0.5%-99.5%的药学上可接受的赋形剂。在某些实施方案中,所述的药物组合物含有1%-99%的药学上可接受的赋形剂。在某些实施方案中,所述的药物组合物含有2%-98%的药学上可接受的赋形剂。
含活性成分的药物组合物可以是适用于口服的形式,例如片剂、糖锭剂、锭剂、水或油混悬液、可分散粉末或颗粒、乳液、硬或软胶囊,或糖浆剂或酏剂。可按照本领域任何已知制备药用组合物的方法制备口服组合物,此类组合物可含有一种或多种选自以下的成分:甜味剂、矫味剂、着色剂和防腐剂,以提供悦目和可口的药用制剂。片剂含有活性成分和用于混合的适宜制备片剂的无毒的可药用的赋形剂。这些赋形剂可以是惰性赋形剂、造粒剂、崩解剂、粘合剂和润滑剂。这些片剂可以不包衣或可通过掩盖药物的味道或在胃肠道中延迟崩解和吸收,因而在较长时间内提供缓释作用的已知技术将其包衣。
也可用其中活性成分与惰性固体稀释剂或其中活性成分与水溶性载体或油溶媒混合的软明胶胶囊提供口服制剂。
水混悬液含有活性物质和用于混合的适宜制备水混悬液的赋形剂。此类赋形剂是悬浮剂、分散剂或湿润剂。水混悬液也可以含有一种或多种防腐剂、一种或多种着色剂、一种或多种矫味剂和一种或多种甜味剂。
油混悬液可通过使活性成分悬浮于植物油或矿物油配制而成。油悬浮液可含有增稠剂。可加入上述的甜味剂和矫味剂,以提供可口的制剂。可通过加入抗氧化剂保存这些组合物。
本公开的药物组合物也可以是水包油乳剂的形式。油相可以是植物油、矿物油或其混合物。适宜的乳化剂可以是天然产生的磷脂,乳剂也可以含有甜味剂、矫味剂、防腐剂和抗氧剂。此类制剂也可含有缓和剂、防腐剂、着色剂和抗氧剂。
本公开的药物组合物可以是无菌注射水溶液形式。可以使用的可接受的溶媒或溶剂有水、林格氏液和等渗氯化钠溶液。无菌注射制剂可以是其中活性成分溶于油相的无菌注射水包油微乳,可通过局部大量注射将注射液或微乳注入患者的血流中。或者,最好按可保持本公开化合物恒定循环浓度的方式给予溶液和微乳。为保持这种恒定浓度,可使用连续静脉内递药装置。这种装置的实例是Deltec CADD-PLUS.TM.5400型静脉注射泵。
本公开的药物组合物可以是用于肌内和皮下给药的无菌注射水或油混悬液的形式。可按已知技术,用适宜的分散剂或湿润剂和悬浮剂配制该混悬液。无菌注射制剂也可以是在肠胃外可接受的无毒稀释剂或溶剂中制备的无菌注射溶液或混悬液。此外,可方便地用无菌固定油作为溶剂或悬浮介质。为此目的,可使用任何调和固定油。此外,脂肪酸也可以制备注射剂。
可按用于直肠给药的栓剂形式给予本公开化合物。可通过将药物与在普通温度下为固体但在直肠中为液体,因而在直肠中会溶化而释放药物的适宜的无刺激性赋形剂混合来制备这些药物组合物。
如本领域技术人员所熟知的,药物的给药剂量依赖于多种因素,包括但并非限定于以下因素:所用具体化合物的活性、患者的年龄、患者的体重、患者的健康状况、患者的行为、患者的饮食、给药时间、给药方式、排泄的速率、药物的组合、疾病的严重性等;另外,最佳的治疗方式如治疗的模式、化合物的日用量或可药用的盐的种类可以根据传统的治疗方案来验证。
术语说明
除非有相反陈述,在说明书和权利要求书中使用的术语具有下述含义。
术语“烷基”指饱和的直链或带有支链的脂肪族烃基,其具有1至20个(例如1、2、3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)碳原子(即C1-20烷基)。所述烷基优选具有1至12个碳原子的烷基(即C1-10烷基),优选具有1至6个碳原子的烷基(即C1-6烷基)。非限制性的实例包括:甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2- 二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基、正壬基、2-甲基-2-乙基己基、2-甲基-3-乙基己基、2,2-二乙基戊基、正癸基、3,3-二乙基己基、2,2-二乙基己基,及其各种支链异构体等。烷基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点被取代,取代基优选选自D原子、卤素、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“亚烷基”指二价烷基,其中烷基如上所定义,其具有1至20个(例如1、2、3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)碳原子(即C1-20亚烷基)。所述亚烷基优选具有1至10个碳原子的亚烷基(即C1-10亚烷基),优选具有1至8个碳原子的亚烷基(即C1-8亚烷基),更优选具有2至7个碳原子的亚烷基(即C2-7亚烷基)或具有1至6个碳原子的亚烷基(即C1-6亚烷基)。非限制性的实例包括:-CH2-、-CH(CH3)-、-C(CH3)2-、-CH2CH2-、-CH(CH2CH3)-、-CH2CH(CH3)-、-CH2C(CH3)2-、-CH2CH2CH2-、-CH2CH2CH2CH2-等。亚烷基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点被取代,取代基优选选自D原子、卤素、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“C0-6亚烷基”指二价烷基,其具有0至6个(例如0、1、2、3、4、5或6)碳原子,C0即为键;因此C0-6亚烷基包括键和C1-6亚烷基;C1-6亚烷基如上所定义。
术语“烯基”指分子中含有至少一个碳碳双键的烷基,其中烷基的定义如上所述,其具有2至12个(例如2、3、4、5、6、7、8、9、10、11或12个)碳原子(即C2-12烯基)。所述烯基优选具有2至6个碳原子的烯基(即C2-6烯基)。非限制性的实例包括:乙烯基、丙烯基、异丙烯基、丁烯基等。烯基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点被取代,取代基优选选自D原子、烷氧基、卤素、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“炔基”指分子中含有至少一个碳碳三键的烷基,其中烷基的定义如上所述,其具有2至12个(例如2、3、4、5、6、7、8、9、10、11或12个)碳原 子(即C2-12炔基)。所述炔基优选具有2至6个碳原子的炔基(即C2-6炔基)。非限制性的实例包括:乙炔基、丙炔基、丁炔基、戊炔基、己炔基等。炔基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点被取代,取代基优选选自D原子、烷氧基、卤素、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“烷氧基”指-O-(烷基),其中烷基的定义如上所述。非限制性的实例包括:甲氧基、乙氧基、丙氧基和丁氧基等。烷氧基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点被取代,取代基优选选自D原子、卤素、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“环烷基”指饱和或部分不饱和的单环全碳环(即单环环烷基)或多环系统(即多环环烷基),其具有3至20个(例如3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)环原子(即3至20元环烷基)。所述环烷基优选具有3至12个环原子的环烷基(即3至12元环烷基)或具有3至10个环原子的环烷基(即3至10元环烷基),更优选具有3至8个环原子的环烷基(即3至8元环烷基),最优选具有3至6个环原子的环烷基(即3至6元环烷基)、4至7个环原子的环烷基(即4至7元环烷基)或5或6个环原子的环烷基(即5或6元环烷基);最优选具有5或6个环原子的环烷基。
所述的单环环烷基,非限制性的实例包括:环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基和环辛基等。
所述的多环环烷基包括:螺环烷基、稠环烷基和桥环烷基。
术语“螺环烷基”指环之间共用一个碳原子(称螺原子)的多环系统,其环内可以含有一个或多个双键,或其环内可以含有一个或多个选自氮、氧和硫的杂原子(所述的氮可任选被氧化,即形成氮氧化物;所述的硫可任选被氧代,即形成亚砜或砜,但不包括-O-O-、-O-S-或-S-S-),条件是至少含有一个全碳环且连接点在该全碳环上,其具有5至20个(例如5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)环原子(即5至20元螺环烷基)。所述螺环烷基优选具有6至14个环原子的螺环烷基(即6至14元螺环烷基),更优选具有7至10个环原子的螺环烷基(即7至10元螺环烷基)。所述螺环烷基包括单螺环烷基和多螺环烷基(如双螺环烷基等),优选单螺环烷基或双螺环烷基,更优选3元/4元、3元/5元、3元/6元、4元/4元、4元/5元、4元/6元、5元/3元、5元/4元、5元/5元、5元/6元、5元/7元、6元/3元、6元/4元、6元/5元、6元/6元、6元/7元、7元/5元或7元/6元单螺环烷基。非限制性的实例包括:
其连接点可在任意位置;
等。
术语“稠环烷基”指环之间共享毗邻的两个碳原子的多环系统,其为单环环烷基与一个或多个单环环烷基稠合,或者单环环烷基与杂环基、芳基或杂芳基中的一个或多个稠合,其中连接点在单环环烷基上,其环内可以含有一个或多个双键,且具有5至20个(例如5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)环原子(即5至20元稠环烷基)。所述稠环烷基优选具有6至14个环原子的稠环烷基(即6至14元稠环烷基),更优选具有7至10个环原子的稠环烷基(即7至10元稠环烷基)。所述稠环烷基包括双环稠环烷基和多环稠环烷基(如三环稠环烷基、四环稠环烷基等),优选双环稠环烷基或三环稠环烷基,更优选3元/4元、3元/5元、3元/6元、4元/4元、4元/5元、4元/6元、5元/3元、5元/4元、5元/5元、5元/6元、5元/7元、6元/3元、6元/4元、6元/5元、6元/6元、6元/7元、7元/5元或7元/6元双环稠环烷基。非限制性的实例包括:
其连接点可在任意位置;
等。
术语“桥环烷基”指环之间共用两个不直接连接的碳原子的全碳多环系统,其环内可以含有一个或多个双键,且具有5至20个(例如5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)碳原子(即5至20元桥环烷基)。所述桥环烷基优选具有6至14个碳原子的桥环烷基(即6至14元桥环烷基),更优选具有7至10个碳原子的桥环烷基(即7至10元桥环烷基)。所述桥环烷基包括双环桥环烷基和多环桥环烷基(如三环桥环烷基、四环桥环烷基等),优选双环桥环烷基或三环桥环烷基。非限制性的实例包括:
其连接点可在任意位置。
环烷基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点被取代,取代基优选选自D原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氧代基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“杂环基”指饱和或部分不饱和的单环杂环(即单环杂环基)或多环杂环系统(即多环杂环基),其环内至少含有一个(例如1、2、3或4个)选自氮、氧和硫的杂原子(所述的氮可任选被氧化,即形成氮氧化物;所述的硫可任选被氧代,即形成亚砜或砜,但不包括-O-O-、-O-S-或-S-S-),且具有3至20个(例如3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)环原子(即3至20元杂环基)。所述杂环基优选具有3至12个环原子的杂环基(即3至12元杂环基)或具有3至10个环原子的杂环基(即3至10元杂环基);进一步优选具有3至8个环原子的杂环基(即3至8元杂环基);更优选具有3至6个环原子的杂环基(即3至6元杂环基)、4至7个环原子的杂环基(即4至7元杂环基)或5或6个环原子的杂环基(即5或6元杂环基);最优选具有5或6个环原子的杂环基。
所述的单环杂环基,非限制性的实例包括:吡咯烷基、四氢吡喃基、1,2,3,6-四氢吡啶基、哌啶基、哌嗪基、氮杂环丁烷基、吗啉基、硫代吗啉基和高哌嗪基等。
所述的多环杂环基包括螺杂环基、稠杂环基和桥杂环基。
术语“螺杂环基”指环之间共用一个原子(称螺原子)的多环杂环系统,其环内可以含有一个或多个双键,且其环内至少含有一个(例如1、2、3或4个)选自氮、氧和硫的杂原子(所述的氮可任选被氧化,即形成氮氧化物;所述的硫可任选被氧代,即形成亚砜或砜,但不包括-O-O-、-O-S-或-S-S-),条件是至少含有一个单环杂环基且连接点在该单环杂环基上,其具有5至20个(例如5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)环原子(即5至20元螺杂环基)。所述螺杂环基优选具有6至14个环原子的螺杂环基(即6至14元螺杂环基),更优选具有7至11个环原子的螺杂环基(即7至11元螺杂环基)。所述螺杂环基包括单螺杂环基和多螺杂环基(如双螺杂环基等),优选单螺杂环基或双螺杂环基,更优选3元/4元、3元/5元、3元/6元、4元/4元、4元/5元、4元/6元、5元/3元、5元/4元、5元/5元、5元/6元、5元/7元、6元/3元、6元/4 元、6元/5元、6元/6元、6元/7元、7元/5元或7元/6元单螺杂环基。非限制性的实例包括:
等。
术语“稠杂环基”指环之间共享毗邻的两个原子的多环杂环系统,其环内可以含有一个或多个双键,且其环内至少含有一个(例如1、2、3或4个)选自氮、氧和硫的杂原子(所述的氮可任选被氧化,即形成氮氧化物;所述的硫可任选被氧代,即形成亚砜或砜,但不包括-O-O-、-O-S-或-S-S-),其为单环杂环基与一个或多个单环杂环基稠合,或者单环杂环基与环烷基、芳基或杂芳基中的一个或多个稠合,其中连接点在单环杂环基上,且具有5至20个(例如5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)环原子(即5至20元稠杂环基)。所述稠杂环基优选具有6至14个环原子的稠杂环基(即6至14元稠杂环基),更优选具有7至10个环原子的稠杂环基(即7至10元稠杂环基)。所述稠杂环基包括双环和多环稠杂环基(如三环稠杂环基、四环稠杂环基等),优选双环稠杂环基或三环稠杂环基,更优选3元/4元、3元/5元、3元/6元、4元/4元、4元/5元、4元/6元、5元/3元、5元/4元、5元/5元、5元/6元、5元/7元、6元/3元、6元/4元、6元/5元、6元/6元、6元/7元、7元/5元或7元/6元双环稠杂环基。非限制性的实例包括:
等。
术语“桥杂环基”指环之间共用两个不直接连接的原子的多环杂环系统,其环内可以含有一个或多个双键,并且其环内至少含有一个(例如1、2、3或4个)选自氮、氧和硫的杂原子(所述的氮可任选被氧化,即形成氮氧化物;所述的硫可任选被氧代,即形成亚砜或砜,但不包括-O-O-、-O-S-或-S-S-),其具有5至20个(例如5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)环原子(即5至20元桥杂环基)。所述桥杂环基优选具有6至14个环原子的桥杂环基(即6至14元桥杂环基),更优选具有7至10个环原子的桥杂环基(即7至10元桥杂环基)。根据组成环的数目可以分为双环桥杂环基和多环桥杂环基(如三环桥杂环基、四环桥杂环基等),优选双环桥杂环基或三环桥杂环基。非限制性的实例包括:
等。
杂环基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点被取代,取代基优选选自D原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氧代基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“芳基”指具有共轭的π电子体系的单环全碳芳环(即单环芳基)或多环芳环系统(即多环芳基),其具有6至20个(例如6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)环原子(即6至20元芳基)。(即6至14元芳基)。所述芳基优选具有6至14个环原子的芳基(即6至14元芳基)或具有6至10个环原子的芳基(即6至10元芳基)。所述的单环芳基,例如苯基。所述的多环芳基,非限制性的实例包括:萘基、蒽基、菲基等。所述多环芳基还包括苯基与杂环基或环烷基中的一个或多个稠合,或萘基与杂环基或环烷基中的一个 或多个稠合,其中连接点在苯基或萘基上,并且在这种情况下,环原子个数继续表示多环芳环系统中的环原子个数,非限制性的实例包括:
等。
芳基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点被取代,取代基优选选自D原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氧代基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“杂芳基”指具有共轭的π电子体系的单环杂芳环(即单环杂芳基)或多环杂芳环系统(即多环杂芳基),其环内至少含有一个(例如1、2、3或4个)选自氮、氧和硫的杂原子(所述的氮可任选被氧化,即形成氮氧化物;所述的硫可任选被氧代,即形成亚砜或砜,但不包括-O-O-、-O-S-或-S-S-),其具有5至14个(例如5、6、7、8、9、10、11、12、13或14个)环原子(即5至14元杂芳基)。所述杂芳基优选具有5至10个环原子的杂芳基(即5至10元杂芳基),更优选具有5或6个环原子的杂芳基(即5或6元杂芳基)。
所述的单环杂芳基,非限制性的实例包括:呋喃基、噻吩基、噻唑基、异噻唑基、噁唑基、异噁唑基、噁二唑基、噻二唑基、咪唑基、吡唑基、三唑基、四唑基、呋咱基、吡咯基、N-烷基吡咯基、吡啶基、嘧啶基、吡啶酮基、N-烷基吡啶酮(如等)、吡嗪基、哒嗪基等。
所述的多环杂芳基,非限制性的实例包括:吲哚基、吲唑基、喹啉基、异喹啉基、喹喔啉基、酞嗪基、苯并咪唑基、苯并噻吩基、喹唑啉基、苯并噻唑基、咔唑基等。所述多环杂芳基还包括单环杂芳基与一个或多个芳基稠合,其中连接点在芳香环上,并且在这种情况下,环原子个数继续表示多环杂芳环系统中的环原子个数。所述多环杂芳基还包括单环杂芳基与环烷基或杂环基中的一个或多个稠合,其中连接点在单环杂芳环上,并且在这种情况下,环原子个数继续表示多环杂芳环系统中的环原子个数。非限制性的实例包括:
等。
杂芳基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点被取代,取代基优选选自氘原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“环烷基烷基”指烷基被一个或多个环烷基取代,其中环烷基、烷基如上所定义。
术语“杂环基烷基”指烷基被一个或多个杂环基取代,其中杂环基、烷基如上所定义。
术语“烷氧基烷基”指烷基被一个或多个烷氧基取代,其中烷氧基、烷基如上所定义。
术语“环烷基氧基”指-O-环烷基,其中环烷基如上所定义。
术语“杂环基氧基”指-O-杂环基,其中杂环基如上所定义。
术语“卤代烷基”指烷基被一个或多个卤素取代,其中烷基如上所定义。
术语“卤代烷氧基”指烷氧基被一个或多个卤素取代,其中烷氧基如上所定义。
术语“羟烷基”指烷基被一个或多个羟基取代,其中烷基如上所定义。
术语“羟烷氧基”指烷氧基被一个或多个羟基取代,其中烷氧基如上所定义。
术语“氘代烷基”指烷基被一个或多个氘原子取代,其中烷基如上所定义。
术语“氘代甲基”指甲基被一个或多个氘原子取代;如CD3、CHD2和CH2D;优选CD3
术语“氘代烷氧基”指烷氧基被一个或多个氘原子取代,如氘代甲氧基(OCD3、OCHD2和OCH2D,优选OCD3)。
术语“卤素”指氟、氯、溴或碘。
术语“羟基”指-OH。
术语“氨基”指-NH2
术语“氰基”指-CN。
术语“硝基”指-NO2
术语“氧代”或“氧代基”指“=O”。
术语“羰基”指C=O。
TBS指叔丁基二甲基硅基。
术语“酰胺基”指-CONH2
哌啶基优选1,4-哌啶基,1,4-哌啶基指
哌嗪基优选1,4-哌嗪基,1,4-哌嗪基指
术语“泛素连接酶”是指促进泛素向特异性底物蛋白质转移、靶向底物蛋白质用于降解的蛋白质家族。例如,小脑蛋白是单独或与E2泛素结合酶组合导致泛素连接到靶标蛋白质上的赖氨酸并且随后靶向特异性蛋白质底物用于通过蛋白酶体降解的E3泛素连接酶蛋白质。因此,E3泛素连接酶单独或与E2泛素结合酶复合是泛素向标靶蛋白质转移的原因。一般来说,泛素连接酶参与聚泛素化,以便第二泛素连接到第一泛素,第三泛素连接到第二泛素,等等。聚泛素化标记蛋白质用于通过蛋白酶体降解。然而,存在一些限于单泛素化的泛素化事件,其中仅单一泛素通过泛素连接酶添加到底物分子。单泛素化蛋白质不被靶向到蛋白酶体用于降解,但可能反而在其细胞位置或功能方面改变,例如经由结合具有能够结合泛素的结构域的其它蛋白质。让事情更复杂的是,泛素上的不同赖氨酸可以由E3靶向以制备链。最常见赖氨酸是泛素链上的Lys48。这是用以制备聚泛素的赖氨酸,其由蛋白酶体识别。
小脑蛋白(Cereblon,CRBN)E3泛素连接酶是使用最为广泛的E3泛素连接酶之一。目前已知的CRBN类型的E3泛素连接酶配体(CRBN配体)包括沙利度胺及其类似物泊马度胺和来那度胺,他们都具有邻苯二甲酰亚胺骨架。Cereblon(CRBN)与受损的DNA结合蛋白1(DDB1)和Cullin 4A形成功能性E3泛素连接酶复合物,在复合物中CRBN作为底物受体而发挥作用,沙利度胺等降解剂分子与CRBN结合后会诱导CRBN识别新的底物蛋白使其被泛素化并且进一步被蛋白酶体识别而降解。
结合小脑蛋白E3泛素连接酶的配体为结合至小脑蛋白E3泛素连接酶的小分子化合物。E3泛素连接酶的配体招募E3泛素连接酶到目标蛋白附近,通过与目标蛋白拉近距离,进而对目标蛋白进行泛素化标记,经标记后的目标蛋白会被体内的蛋白酶体系统进行降解。
术语“氨基保护基”是指为了使分子其它部位进行反应时氨基保持不变,在氨基上引入的易于脱去的基团。非限制性的实例包括:(三甲基硅)乙氧基甲基、四 氢吡喃基、叔丁氧羰基(Boc)、苄氧羰基(Cbz)、笏甲氧羰基(Fmoc)、烯丙氧羰基(Alloc)、三甲基硅乙氧羰基(Teoc)、甲氧羰基、乙氧羰基、邻苯二甲酰基(Pht)、对甲苯磺酰基(Tos)、三氟乙酰基(Tfa)、三苯甲基(Trt)、2,4-二甲氧基苄基(DMB)、乙酰基、苄基、烯丙基、对甲氧苄基等。
本公开化合物可以存在特定的立体异构体形式。术语“立体异构体”是指结构相同但原子在空间中的排列不同的异构体。其包括顺式和反式(或Z和E)异构体、(-)-和(+)-异构体、(R)-和(S)-对映异构体、非对映异构体、(D)-和(L)-异构体、互变异构体、阻转异构体、构象异构体及其混合物(如外消旋体、非对映异构体的混合物)。本公开化合物中的取代基可以存在另外的不对称原子。所有这些立体异构体以及它们的混合物,均包括在本公开的范围内。可以通过手性合成、手性试剂或者其他常规技术制备光学活性的(-)-和(+)-异构体、(R)-和(S)-对映异构体以及(D)-和(L)-异构体。本公开某化合物的一种异构体,可以通过不对称合成或者手性助剂来制备,或者,当分子中含有碱性官能团(如氨基)或酸性官能团(如羧基)时,与适当的光学活性的酸或碱形成非对映异构体的盐,然后通过本领域所公知的常规方法进行非对映异构体拆分,得到纯的异构体。此外,对映异构体和非对映异构体的分离通常是通过色谱法完成。
本公开所述化合物的化学结构中,键表示未指定构型,即如果化学结构中存在手性异构体,键可以为或者同时包含两种构型。对于所有的碳-碳双键,即使仅命名了一个构型,Z型和E型均包括在内。
本公开的化合物可以以不同的互变异构体形式存在,并且所有这样的形式包含在本公开的范围内。术语“互变异构体”或“互变异构体形式”是指平衡存在并且容易从一种异构形式转化为另一种异构形式的结构异构体。其包括所有可能的互变异构体,即以单一异构体的形式或以所述互变异构体的任意比例的混合物的形式存在。非限制性的实例包括:酮-烯醇、亚胺-烯胺、内酰胺-内酰亚胺等。内酰胺-内酰亚胺平衡实例如下所示:
如当提及吡唑基时,应理解为包括如下两种结构中的任何一种或两种互变异构体的混合物:
所有的互变异构形式在本公开的范围内,且化合物的命名不排除任何互变异构体。
本公开的化合物包括其化合物的所有合适的同位素衍生物。术语“同位素衍生物”是指至少一个原子被具有相同原子序数但原子质量不同的原子替代的化合物。可引入到本公开化合物中的同位素的实例包括氢、碳、氮、氧、磷、硫、氟、氯、溴和碘等的稳定和放射性的同位素,例如分别为2H(氘,D)、3H(氚,T)、11C、13C、14C、15N、17O、18O、32p、33p、33S、34S、35S、36S、18F、36Cl、82Br、123I、124I、125I、129I和131I等,优选氘。
相比于未氘代药物,氘代药物有降低毒副作用、增加药物稳定性、增强疗效、延长药物生物半衰期等优势。本公开的化合物的所有同位素组成的变换,无论放射性与否,都包括在本公开的范围之内。与碳原子连接的各个可用的氢原子可独立地被氘原子替换,其中氘的替换可以是部分或完全的,部分氘的替换是指至少一个氢被至少一个氘替换。
当一个位置被特别地指定为氘(D)时,该位置应理解为具有大于氘的天然丰度(其为0.015%)至少1000倍的丰度的氘(即至少15%的氘掺入)。示例中化合物的具有大于氘的天然丰度可以是至少1000倍的丰度的氘(即至少15%的氘掺入)、至少2000倍的丰度的氘(即至少30%的氘掺入)、至少3000倍的丰度的氘(即至少45%的氘掺入)、至少3340倍的丰度的氘(即至少50.1%的氘掺入)、至少3500倍的丰度的氘(即至少52.5%的氘掺入)、至少4000倍的丰度的氘(即至少60%的氘掺入)、至少4500倍的丰度的氘(即至少67.5%的氘掺入)、至少5000倍的丰度的氘(即至少75%的氘掺入)、至少5500倍的丰度的氘(即至少82.5%的氘掺入)、至少6000倍的丰度的氘(即至少90%的氘掺入)、至少6333.3倍的丰度的氘(即至少95%的氘掺入)、至少6466.7倍的丰度的氘(即至少97%的氘掺入)、至少6600倍的丰度的氘(即至少99%的氘掺入)、至少6633.3倍的丰度的氘(即至少99.5%的氘掺入)或更高丰度的氘。
“任选的”或“任选”是指随后所描述的事件或环境可以但不必然发生,其包括该事件或环境发生或不发生两种情形。例如“任选被卤素或者氰基取代的C1-6烷基”包括烷基被卤素或者氰基取代的情形和烷基不被卤素或氰基取代的情形。
“取代”或“取代的”指基团中的一个或多个氢原子,优选1至6个,更优选1至3个氢原子彼此独立地被相应数目的取代基取代。本领域技术人员能够在不付出过多努力的情况下(通过实验或理论)确定可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和键的碳原子(如烯)结合时可能是不稳定的。
“药物组合物”表示含有一种或多种本文所述化合物或其可药用的盐与其他化学组分的混合物,以及其他组分例如药学上可接受的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
“可药用的盐”是指本公开化合物的盐,可选自无机盐或有机盐。这类盐用于哺乳动物体内时具有安全性和有效性,且具有应有的生物活性。可以在化合物的最终分离和纯化过程中,或通过使合适的基团与合适的碱或酸反应来单独制备。通常用于形成药学上可接受的盐的碱包括无机碱,例如氢氧化钠和氢氧化钾,以及有机碱,例如氨。通常用于形成药学上可接受的盐的酸包括无机酸以及有机酸。
针对药物或药理学活性剂而言,术语“治疗有效量”是指足以达到或至少部分达到预期效果的药物或药剂的用量。治疗有效量的确定因人而异,取决于受体的年龄和一般情况,也取决于具体的活性物质,个案中合适的治疗有效量可以由本领域技术人员根据常规试验确定。
本文所用的术语“药学上可接受的”是指这些化合物、材料、组合物和/或剂型,在合理的医学判断范围内,适用于与患者组织接触而没有过度毒性、刺激性、过敏反应或其他问题或并发症,具有合理的获益/风险比,并且对预期的用途是有效。
本文所使用的,单数形式的“一个”、“一种”和“该”包括复数引用,反之亦然,除非上下文另外明确指出。
当将术语“约”应用于诸如pH、浓度、温度等的参数时,表明该参数可以变化±10%,并且有时更优选地在±5%之内。如本领域技术人员将理解的,当参数不是关键时,通常仅出于说明目的给出数字,而不是限制。
本公开化合物的合成方法
为了完成本公开的目的,本公开采用如下技术方案:
方案一
本公开通式(III)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(IIIa)所示的化合物或其盐(优选盐酸盐)与通式(IIIb)所示的化合物或其盐(优选盐酸盐),在碱性条件下缩合剂存在下发生缩合反应得到通式(III)所示的化合物或其可药用的盐;
RW为氢原子;
环J2’、R1、R2、R4至R6、RA、Ra、Rb、RZ1、RZ2、L1、L2、J1、J5、J6、Rq、s和n如通式(III)中所定义。
方案二
本公开通式(III’)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(IIIa)所示的化合物或其盐(优选盐酸盐)与通式(III’b)所示的化合物或其盐(优选盐酸盐),在碱性条件下缩合剂存在下发生缩合反应得到通式(III’)所示的化合物或其可药用的盐;
RW为氢原子;
环J2’、R1、R2、R4、R5、R7、RA、Ra、Rb、RZ1、RZ2、L1、L2、J1、J5、J6、Rq、s、n和Y1如通式(III’)中所定义。
方案三
本公开通式(IV)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(IVa)所示的化合物或其盐(优选盐酸盐)与通式(IVb)所示的化合物或其盐(优选盐酸盐),在碱性条件下缩合剂存在下发生缩合反应得到通式(IV)所示的化合物或其可药用的盐;
环J6’、R1、R2、R4至R6、RA、Ra、Rb、RZ1、RZ2、L1、L2、J1、J2、Rq、p、s和n如通式(IV)中所定义。
方案四
本公开通式(IV’)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(IVa)所示的化合物或其盐(优选盐酸盐)与通式(IV’b)所示的化合物或其盐(优选盐酸盐),在碱性条件下缩合剂存在下发生缩合反应得到通式(IV’)所示的化合物或其可药用的盐;
环J6’、R1、R2、R4、R5、R7、RA、Ra、Rb、RZ1、RZ2、L1、L2、J1、J2、Rq、p、s、n和Y1如通式(IV’)中所定义。
方案五-1
本公开通式(V-1)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(V-1A)所示的化合物或其盐(优选三氟乙酸盐或盐酸盐)与通式(V-1B)所示的化合物或其盐在酸性条件下发生还原胺化反应得到通式(V-1)所示的化合物或其可药用的盐;
RW为氢原子;
其中,R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J2’、R03、j、q、J4、J5、J6、Rq、Y2和s如通式(V-1)中所定义。
方案五-2
本公开通式(V-2)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(V-2A)所示的化合物或其盐与通式(V-2B)所示的化合物或其盐(优选盐酸盐)在酸性条件下发生还原胺化反应得到通式(V-2)所示的化合物或其可药用的盐;
其中,R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、q、环J3’、R03、j、J4、J5、J6、Rq、Y2和s如通式(V-2)中所定义。
方案六
本公开通式(I’-3-1)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(I’-3-1A)所示的化合物或其盐发生分子内关环反应得到通式(I’-3-1)所示的化合物或其可药用的盐;
其中,RL选自-OH、卤素和-O-烷基;
R1至R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1至J6、Rq、Y2和s如通式(I’-3-1)中所定义。
方案六-1
本公开通式(I’-3-2)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(I’-3-2A)所示的化合物或其盐发生分子内关环反应得到通式(I’-3-2)所示的化合物或其可药用的盐;
其中,RL选自-OH、卤素和-O-烷基;
R1至R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1至J6、Rq、Y2和s如通式(I’-3-2)中所定义。
方案六-2
本公开通式(I’-3-1)和(I’-3-2)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(I’-3)所示的化合物或其盐经手性拆分得到通式(I’-3-1)和(I’-3-2)所示的化合物或其可药用的盐;
其中,R1至R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1至J6、Rq、Y2和s如通式(I’-3-1)或(I’-3-2)中所定义。
方案七
本公开通式(V-1-1)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(V-1-1A)所示的化合物或其盐发生分子内关环反应得到通式(V-1-1)所示的化合物或其可药用的盐;
其中,RL选自-OH、卤素和-O-烷基;
R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J2’、R03、j、q、J4、J5、J6、Rq、Y2和s如通式(V-1-1)中所定义。
方案七-1
本公开通式(V-1-2)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(V-1-2A)所示的化合物或其盐发生分子内关环反应得到通式(V-1-2)所示的化合物或其可药用的盐;
其中,RL选自-OH、卤素和-O-烷基;
R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J2’、R03、j、q、J4、J5、J6、Rq、Y2和s如通式(V-1-2)中所定义。
方案七-2
本公开通式(V-1-1)和(V-1-2)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(V-1)所示的化合物或其盐经手性拆分得到通式(V-1-1)和(V-1-2)所示的化合物或其可药用的盐;
其中,R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J2’、R03、j、q、J4、J5、J6、Rq、Y2和s如通式(V-1-1)或(V-1-2)中所定义。
方案八
本公开通式(V-2-1)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(V-2-1A)所示的化合物或其盐发生分子内关环反应得到通式(V-2-1)所示的化合物或其可药用的盐;
其中,RL选自-OH、卤素和-O-烷基;
R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、q、环J3’、R03、j、J4、J5、J6、Rq、Y2和s如通式(V-2-1)中所定义。
方案八-1
本公开通式(V-2-2)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(V-2-2A)所示的化合物或其盐发生分子内关环反应得到通式(V-2-2)所示的化合物或其可药用的盐;
其中,RL选自-OH、卤素和-O-烷基;
R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、q、环J3’、R03、j、J4、J5、J6、Rq、Y2和s如通式(V-2-2)中所定义。
方案八-2
本公开通式(V-2-1)和(V-2-2)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(V-2)所示的化合物或其盐经手性拆分得到通式(V-2-1)和(V-2-2)所示的化合物或其可药用的盐;
其中,R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、q、环J3’、R03、j、J4、J5、J6、Rq、Y2和s如通式(V-2-1)或(V-2-2)中所定义。
方案九
本公开通式(X’-1)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(X’-1a)所示的化合物或其盐(优选三氟乙酸盐或盐酸盐)与通式(X’-1b)所示的化合物或其盐在酸性条件下发生还原胺化反应得到通式(X’-1)所示的化合物或其可药用的盐;其中,
q为1、2、3、4、5或6;q-1为0、1、2、3、4或5;
R1、R9、y、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J2’、R03、j、J4、J5、J6和E如通式(X’-1)中所定义。
方案十
本公开通式(X’-2)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(X’-2a)所示的化合物或其盐与通式(X’-2b)所示的化合物或其盐(优选盐酸盐)在酸性条件下发生还原胺化反应得到通式(X’-2)所示的化合物或其可药用的盐;其中,
q为1、2、3、4、5或6;q-1为0、1、2、3、4或5;
R1、R9、y、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J3’、R03、j、J4、J5、J6和E如通式(X’-2)中所定义。
方案十一
本公开通式(X’-3)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
通式(X’-2a)所示的化合物或其盐与通式(X’-3b)所示的化合物或其盐(优选盐酸盐)在酸性条件下发生还原胺化反应得到通式(X’-3)所示的化合物或其可药用的盐;其中,
q为1、2、3、4、5或6;q-1为0、1、2、3、4或5;
R1、R9、y、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J3’、R03、j、J4、J5、J6、Y1、R7、Rq和s如通式(X’-3)中所定义。
以上缩合反应中提供碱性条件的试剂包括有机碱和无机碱类,所述的有机碱类包括但不限于三乙胺、吡啶、N,N-二异丙基乙胺、正丁基锂、二异丙基氨基锂、醋酸钠、醋酸钾、叔丁醇钠、叔丁醇钾或1,8-二氮杂二环十一碳-7-烯,所述的无机碱类包括但不限于氢化钠、磷酸钾、碳酸钠、碳酸钾、碳酸铯、氢氧化钠、氢氧化锂和氢氧化钾;优选为N,N-二异丙基乙胺。
上述反应中所述的缩合剂包括但不限于1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐、N,N'-二环己基碳化二亚胺、N,N'-二异丙基碳二酰亚胺、O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸酯、1-羟基苯并三唑、1-羟基-7-偶氮苯并三氮唑、O-苯并三氮唑-N,N,N',N'-四甲脲六氟磷酸酯、O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(HATU)、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、苯并三氮唑-1-基氧基三(二甲基氨基)磷鎓六氟磷酸盐或六氟磷酸苯并三唑-1-基-氧基三吡咯烷基磷;优选为HATU。
上述合成方案中,提供酸性条件的试剂包括但不限于氯化氢、氯化氢的1,4-二氧六环溶液、盐酸的1,4-二氧六环溶液、三氟醋酸、甲酸、乙酸、冰乙酸、盐酸、浓硫酸、甲磺酸、硝酸、磷酸、对苯甲磺酸、四氯化钛、Me3SiCl和TMSOTf;优选为乙酸。
具体的,酸性试剂可以是直接加入的酸试剂;和/或加入碱性盐试剂,由该碱性盐与原料的酸性盐形式带入的酸在反应过程中生成的酸。所述的碱性盐包括但不限于乙酸钠、无水乙酸钠,优选无水乙酸钠。
上述合成方案中,所述的还原剂包括但不限于三乙酰氧基硼氢化钠、硼氢化钠、硼氢化锂、醋酸硼氢化钠、氰基硼氢化钠和乙酰硼氢化钠等,优选三乙酰氧基硼氢化钠或氰基硼氢化钠;优选氰基硼氢化钠。
上述合成方案六、六-1、七、七-1、八和八-1的分子内关环反应原理为酰胺缩合反应和/或胺酯交换反应,优选为胺酯交换反应,所述胺酯交换反应任选在催化剂存在下进行,所述催化剂优选为苯磺酸。
上述步骤的反应优选在溶剂中进行,所用的溶剂包括但不限于:吡啶、乙二醇二甲醚、醋酸、甲醇、乙醇、乙腈、正丁醇、甲苯、四氢呋喃、二氯甲烷、石油醚、乙酸乙酯、正己烷、二甲基亚砜、1,4-二氧六环、水、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、1,2-二溴乙烷及其混合物。
具体实施方式
以下结合实施例用于进一步描述本公开,但这些实施例并非限制着本公开的范围。
实施例
化合物的结构是通过核磁共振(NMR)或/和质谱(MS)来确定的。NMR位移(δ)以10-6(ppm)的单位给出。NMR的测定是用Bruker AVANCE-400核磁仪或Bruker AVANCE NEO 500M,测定溶剂为氘代二甲基亚砜(DMSO-d6)、氘代氯仿(CDCl3)、氘代甲醇(CD3OD),内标为四甲基硅烷(TMS)。
MS的测定用Agilent 1200/1290 DAD-6110/6120 Quadrupole MS液质联用仪(生产商:Agilent,MS型号:6110/6120 Quadrupole MS)。
waters ACQuity UPLC-QD/SQD(生产商:waters,MS型号:waters ACQuity Qda Detector/waters SQ Detector)。
THERMO Ultimate 3000-Q Exactive(生产商:THERMO,MS型号:THERMO Q Exactive)。
高效液相色谱法(HPLC)分析使用Agilent HPLC 1200DAD、Agilent HPLC 1200VWD和Waters HPLC e2695-2489高效液相色谱仪。
手性HPLC分析测定使用Agilent 1260 DAD高效液相色谱仪。
高效液相制备使用Waters 2545-2767、Waters 2767-SQ Detecor2、Shimadzu LC-20AP和Gilson GX-281制备型色谱仪。
手性制备使用Shimadzu LC-20AP制备型色谱仪。
CombiFlash快速制备仪使用Combiflash Rf200(TELEDYNE ISCO)。
薄层层析硅胶板使用烟台黄海HSGF254或青岛GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm~0.2mm,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm。
硅胶柱色谱法一般使用烟台黄海硅胶200~300目硅胶为载体。
激酶平均抑制率及IC50值的测定用NovoStar酶标仪(德国BMG公司)。
本公开的已知的起始原料可以采用或按照本领域已知的方法来合成,或可购买自ABCR GmbH&Co.KG、Acros Organics、Aldrich Chemical Company、韶远化学科技(Accela ChemBio Inc)、毕得医药、达瑞化学品等公司。
实施例中无特殊说明,反应均能够在氩气氛或氮气氛下进行。
氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
氢气氛是指反应瓶连接一个约1L容积的氢气气球。
加压氢化反应使用Parr 3916EKX型氢化仪和清蓝QL-500型氢气发生器或HC2-SS型氢化仪。
氢化反应通常抽真空,充入氢气,反复操作3次。
微波反应使用CEM Discover-S 908860型微波反应器。
实施例中无特殊说明,溶液是指水溶液。
实施例中无特殊说明,反应的温度为室温,为20℃~30℃。
实施例1
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(4-((2,6-二氧代哌啶-3-基)氨基)苯基)哌啶-1-基)乙酰基)哌嗪-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺1

第一步
4-(2-溴-5-甲氧基-4-硝基苯基)哌嗪-1-羧酸叔丁酯1b
称取1-溴-2-氟-4-甲氧基-5-硝基苯1a(2g,8.00mmol,毕得医药)、哌嗪-1-羧酸叔丁酯(1.64g,8.80mmol,韶远化学科技(上海)有限公司)到50mL茄瓶中,加入N,N-二甲基甲酰胺(25mL)和无水碳酸钾(2.76g,20.0mmol),然后85℃反应16小时。待反应液冷却至室温,向体系中加入水(30mL),乙酸乙酯萃取(30mL×3),合并有机相,用饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥。过滤、减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化残余物,得到标题化合物1b(3.30g,产率:99%)。
MS m/z(ESI):416.1[M+1]。
第二步
4-(5-甲氧基-4-硝基-2-(1H-吡唑-4-基)苯基)哌嗪-1-羧酸叔丁酯1c
称取化合物1b(2.8g,6.73mmol)、4-吡唑硼酸频哪醇酯(1.70g,8.74mmol,韶远化学科技(上海)有限公司)到100mL茄形瓶中,依次加入1,1′-双(二苯基膦)二茂 铁二氯化钯(492mg,0.67mmol)、碳酸钠(2.14g,20.18mmol)、1,4-二氧六环和水的混合溶剂(48mL,V/V=4/1),升温至105℃反应16小时。待反应液冷却到室温,反应液用硅藻土过滤,滤饼用乙酸乙酯洗涤(50mL×3),滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物1c(500mg,产率:18%)。
MS m/z(ESI):404.1[M+1]。
第三步
4-(4-氨基-5-甲氧基-2-(1H-吡唑-4-基)苯基)哌嗪-1-羧酸叔丁酯1d
将化合物1c(500mg,1.24mmol)溶于甲醇(10mL),加入10%干钯碳(150mg,1.24mmol,韶远化学科技(上海)有限公司),氢气置换三次后,氢气氛下反应16小时。反应液经硅藻土过滤,滤液减压浓缩,得到粗产品标题化合物1d(462mg,产率:99%),不经纯化直接用于下一步反应。
MS m/z(ESI):374.2[M+1]。
第四步
4-(4-((5-溴-4-((4-羟基-2-(N-甲基甲磺酰氨基)苯基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-(1H-吡唑-4-基)苯基)哌嗪-1-羧酸叔丁酯1f
将N-(2-((5-溴-2-氯嘧啶-4-基)氨基)-5-羟基苯基)-N-甲基甲磺酰胺1e(420mg,1.03mmol,采用专利申请“WO2021216440中说明书第70页的实施例A29”公开的方法制备而得)、化合物1d(442mg,1.18mmol)、三氟乙酸(470mg,4.12mmol)加入到异丙醇(40mL)中,加热升温至90℃反应16小时。冷却至室温,减压除去溶剂,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物1f(350mg,产率:46%)。
MS m/z(ESI):744.1[M+1]。
第五步
(Z)-4-(45-溴-24-甲氧基-62-(N-甲基甲磺酰氨基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌嗪-1-羧酸叔丁酯1g
称取化合物1f(150mg,0.20mmol)、1,4-二溴丁烷(87mg,0.40mmol)到50mL茄形瓶中,加入N,N-二甲基甲酰胺(2.5mL)、碳酸钾(111mg,0.81mmol)、四丁基碘化铵(37mg,0.101mmol),反应16小时。向体系中加入水(25mL),乙酸乙酯萃取(25mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL),无水硫酸钠干燥。过滤,减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物1g(76mg,产率:47%)。
MS m/z(ESI):798.2[M+1]。
第六步
(Z)-N-(45-溴-24-甲氧基-26-(哌嗪-1-基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺盐酸盐1h
将化合物1g(66mg,0.083mmol)溶于二氯甲烷(2mL),加入4M氯化氢的1,4-二氧六环溶液(2mL),反应2小时,减压除去溶剂,真空干燥,得标题化合物1h(60mg,产率:99%)。
MS m/z(ESI):698.1[M+1]。
第七步
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(4-((2,6-二氧代哌啶-3-基)氨基)苯基)哌啶-1-基)乙酰基)哌嗪-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂
-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺1
向25mL三口瓶中依次加入N,N-二甲基甲酰胺(3mL)、化合物1h(60mg,0.082mmol)、(±)-2-(4-(4-((2,6-二氧代哌啶-3-基)氨基)苯基)哌啶-1-基)乙酸盐酸盐1i(31mg,0.082mmol,采用公知的方法“WO2021127561A1中说明书第400页通用步骤E”制备得到)、O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(37mg,0.098mmol),冰浴下,缓慢加入N,N-二异丙基乙胺(63mg,0.49mmol),反应20分钟。反应液过滤,经高效液相制备色谱法纯化(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-67%,流速:30mL/min),得到标题化合物1(外消旋体,40mg,产率:48%)。
MS m/z(ESI):1025.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.75(s,1H),8.36(s,1H),8.24-8.02(m,2H),7.93(s,1H),7.79-7.64(m,2H),7.25(s,1H),7.05(s,1H),6.94(d,2H),6.76(s,1H),6.60(d,2H),6.12(dd,1H),5.65(d,1H),4.30-4.20(m,3H),3.84(s,3H),3.80(t,2H),3.77-3.55(m,4H),3.19(s,3H),3.17(s,1H),3.08(s,3H),2.95-2.88(m,2H),2.80(s,2H),2.77-2.69(m,3H),2.59(t,1H),2.56(t,1H),2.37-2.28(m,1H),2.14-2.01(m,5H),1.91-1.78(m,1H),1.73-1.52(m,6H)。
实施例2
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(4-((2,6-二氧代哌啶-3-基)氨基)苯基)哌啶-1-基)乙酰基)哌嗪-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十蕃-62-基)-N-甲基甲磺酰胺2

第一步
(Z)-4-(45-溴-24-甲氧基-62-(N-甲基甲磺酰氨基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十蕃-26-基)哌嗪-1-羧酸叔丁酯2a
称取化合物1f(100mg,0.13mmol)、1,3-二溴丙烷(41mg,0.20mmol)到50mL茄形瓶中,加入N,N-二甲基甲酰胺(2.5mL)、碳酸钾(74mg,0.54mmol)、四丁基碘化铵(25mg,0.067mmol),反应16小时。向体系中加入水(25mL),用乙酸乙酯萃取(25mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL),无水硫酸钠干燥。过滤,减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物2a(44mg,产率:42%)。
MS m/z(ESI):784.1[M+1]。
第二步
(Z)-N-(45-溴-24-甲氧基-26-(哌嗪-1-基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十蕃-62-基)-N-甲基甲磺酰胺盐酸盐2b
将化合物2a(44mg,0.056mmol)溶于二氯甲烷(2mL),加入4M氯化氢的1,4-二氧六环溶液(2mL),反应2小时,减压除去溶剂,真空干燥,得到标题化合物2b(40mg,产率:99%)。
MS m/z(ESI):684.1[M+1]。
第三步
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(4-((2,6-二氧代哌啶-3-基)氨基)苯基)哌啶-1-基)乙酰基)哌嗪-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十蕃-62-基)-N-甲基甲磺酰胺2
向25mL三口瓶中依次加入N,N-二甲基甲酰胺(3mL)、化合物2b(40mg,0.055mmol)、化合物1i(22mg,0.058mmol)、O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(32mg,0.083mmol),冰浴下,缓慢加入N,N-二异丙基乙胺(43mg, 0.33mmol),反应20分钟。反应液过滤,经高效液相制备色谱法纯化(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-60%,流速:30mL/min),得到标题化合物2(外消旋体,30mg,产率:53%)。
MS m/z(ESI):1011.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.74(s,1H),8.18(s,1H),8.08(s,1H),7.80(s,1H),7.72(d,2H),7.61(d,2H),7.12(d,1H),6.94(d,2H),6.75(s,1H),6.59(d,2H),5.98(d,1H),5.64(d,1H),4.30-4.19(m,3H),4.14(t,2H),3.85(s,3H),3.69-3.61(m,2H),3.59-3.47(m,2H),3.17(s,3H),3.15(s,2H),3.05(s,3H),2.90(d,2H),2.83-2.68(m,5H),2.57(dt,1H),2.35-2.20(m,3H),2.13-2.03(m,3H),1.85(qd,1H),1.68(d,2H),1.61-1.50(m,2H)。
实施例3
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(4-((2,6-二氧代哌啶-3-基)氨基)苯基)哌啶-1-基)乙酰基)哌嗪-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十二蕃-62-基)-N-甲基甲磺酰胺3
第一步
(Z)-4-(45-溴-24-甲氧基-62-(N-甲基甲磺酰氨基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十二蕃-26-基)哌嗪-1-羧酸叔丁酯3a
称取化合物1f(140mg,0.19mmol)、1,5-二溴戊烷(86mg,0.38mmol)到25mL茄形瓶中,加入N,N-二甲基甲酰胺(3mL)、碳酸钾(104mg,0.75mmol)、四丁基碘化铵(35mg,0.094mmol),然后40℃反应16小时。停止加热,向体系中加入水(8mL),用乙酸乙酯萃取(15mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物3a(80mg,产率:52%)。
MS m/z(ESI):812.1[M+1]。
第二步
(Z)-N-(45-溴-24-甲氧基-26-(哌嗪-1-基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十二蕃-62-基)-N-甲基甲磺酰胺盐酸盐3b
将化合物3a(80mg,0.098mmol)溶于二氯甲烷(2mL),加入三氟乙酸(0.5mL),反应1小时,向体系中加入饱和碳酸氢钠溶液(5mL),用二氯甲烷萃取(10mL×3),合并有机相,用饱和氯化钠溶液洗涤(15mL),无水硫酸钠干燥,过滤,滤液减压浓缩,得标题化合物3b(70mg,产率:99%)。
MS m/z(ESI):712.1[M+1]。
第三步
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(4-((2,6-二氧代哌啶-3-基)氨基)苯基)哌啶-1-基)乙酰基)哌嗪-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十二蕃-62-基)-N-甲基甲磺酰胺3
向25mL单口瓶中依次加入N,N-二甲基甲酰胺(3mL)、化合物3b(35mg,0.049mmol)、化合物1i(21mg,0.054mmol)、O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(21mg,0.054mmol),冰浴下,缓慢加入N,N-二异丙基乙胺(32mg,0.25mmol),反应20分钟。反应液过滤,经高效液相制备色谱法纯化(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:48%-58%,流速:30mL/min),得到标题化合物3(外消旋体,22mg,产率:43%)。
MS m/z(ESI):1039.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.73(s,1H),8.20-8.11(m,3H),7.86(s,1H),7.80(s,1H),7.74(d,1H),7.26(s,1H),7.09(s,1H),6.93(d,2H),6.77(s,1H),6.58(d,2H),6.23(dd,1H),5.63(d,1H),4.25-4.18(m,1H),4.15-4.06(m,2H),3.93(t,2H),3.82(s,3H),3.79-3.70(m,2H),3.70-3.60(m,2H),3.21(s,3H),3.19(s,2H),3.09(s,3H),2.96-2.89(m,2H),2.88-2.83(m,2H),2.82-2.76(m,2H),2.75-2.66(m,1H),2.56(dt,1H),2.37-2.28(m,1H),2.13-2.05(m,3H),1.90-1.78(m,3H),1.69(d,2H),1.63-1.53(m,2H),1.52-1.43(m,2H),1.32-1.20(m,2H)。
实施例4
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶-1-基)乙酰基)哌嗪-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂 -4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺4
向25mL三口瓶中依次加入N,N-二甲基甲酰胺(3mL)、化合物1h(53mg,0.072mmol)、2-(4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶-1-基)乙酸盐酸盐4a(28mg,0.061mmol,采用公知的方法“WO2021127561A1中说明书第401页”制备得到)、O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(27mg,0.072mmol),冰浴下,缓慢加入N,N-二异丙基乙胺(46mg,0.36mmol),反应20分钟。反应液过滤,经高效液相制备色谱法纯化(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:50%-65%%,流速:30mL/min),得到标题化合物4(外消旋体,25mg,产率:32%)。
MS m/z(ESI):1101.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.55(s,1H),8.37(s,1H),8.19(s,1H),8.09(s,1H),7.92(s,1H),7.77(s,1H),7.71(d,1H),7.58(d,1H),7.53(s,1H),7.26(s,1H),7.08(d,1H),7.06(s,1H),6.76(s,1H),6.12(dd,1H),4.29-4.22(m,2H),3.98(s,3H),3.92(t,2H),3.84(s,3H),3.80(t,2H),3.74-3.57(m,3H),3.41(s,2H),3.28-3.19(m,2H),3.18(s,3H),3.07(s,3H),3.01(d,1H),2.84-2.74(m,6H),2.69-2.62(m,1H),2.56-2.43(m,2H),2.28-2.18(m,1H),2.10-2.01(m,2H),1.86(d,1H),1.68-1.59(m,2H)。
实施例5
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶-1-基)乙酰基)哌嗪-1基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十二蕃-62-基)-N-甲基甲磺酰胺5
向25mL三口瓶中依次加入N,N-二甲基甲酰胺(3mL)、化合物3b(35mg,0.049mmol)、化合物4a(24mg,0.052mmol)、O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(21mg,0.054mmol),冰浴下,缓慢加入N,N-二异丙基乙胺(32mg,0.25mmol),反应20分钟。反应液过滤,经高效液相制备色谱法纯化(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:47%-53%,流速:30mL/min),得到标题化合物5(外消旋体,25mg,产率:46%)。
MS m/z(ESI):1115.3[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.54(s,1H),8.17(s,2H),8.09(s,1H),7.92(s,1H),7.80(s,1H),7.76(d,1H),7.58(d,1H),7.53(s,1H),7.29(s,1H),7.12(s,1H),7.08(d,1H),6.80(s,1H),6.24(dd,1H),4.18-4.10(m,2H),3.97(s,3H),3.96-3.90(m,4H),3.83(s,3H),3.79-3.69(m,3H),3.48-3.41(m,3H),3.29-3.24(m,2H),3.21(s,3H),3.10(s,3H),3.03(d,1H),2.93-2.86(m,3H),2.85-2.79(m,2H),2.76(t,2H),2.70-2.63(m,1H),2.31-2.20(m,1H),1.93-1.83(m,3H),1.53-1.44(m,2H),1.33-1.26(m,2H)。
实施例6
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶-1-基)乙酰基)哌嗪-1-基)-25-氟-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂十一蕃-62-基)-N-甲基甲基磺酰胺6
第一步
4-(2-溴-6-氟-4-硝基苯基)哌嗪-1-羧酸叔丁酯6b
称取1-溴-2,3-二氟-5-硝基苯6a(2g,8.4mmol)、哌嗪-1-羧酸叔丁酯(1.56g,8.40mmol)到50mL茄形瓶中,加入乙腈(40mL)、无水碳酸钾(2.32g,16.8mmol),然后85℃反应16小时。停止加热,待反应冷却至室温,向体系中加入水(30mL),用乙酸乙酯萃取(30mL×3),合并有机相,用饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物6b(2.78g,产率:82%)。
MS m/z(ESI):402.1[M-1]。
第二步
4-(2-氟-4-硝基-6-(1H-吡唑-4-基)苯基)哌嗪-1-羧酸叔丁酯6c
称取化合物6b(1g,2.47mmol)、4-吡唑硼酸频哪醇酯(720mg,3.71mmol)到100mL茄形瓶中,依次加入1,1′-双(二苯基膦)二茂铁二氯化钯(181mg,0.25mmol),磷酸钾(12.6g,38.67mmol),加入1,4-二氧六环和水的混合溶剂(12.5mL,V/V=4/1),然后在105℃反应16小时。停止加热,待反应液冷却到室温,反应液用硅藻 土过滤,滤饼用乙酸乙酯洗涤(15mL×3),滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物6c(470mg,产率:49%)。
MS m/z(ESI):392.3[M+1]。
第三步
4-(4-氨基-2-氟-6-(1H-吡唑-4-基)苯基)哌嗪-1-羧酸叔丁酯6d
将化合物6c(470mg,1.20mmol)溶于甲醇(10mL),加入10%干钯碳(128mg,1.20mmol),氢气置换三次后,氢气氛下反应4小时。滤液减压浓缩,得到粗产品标题化合物6d(430mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):362.2[M+1]。
第四步
4-(4-((5-溴-4-((4-羟基-2-(N-甲基甲磺酰氨基)苯基)氨基)嘧啶-2-基)氨基)-2-氟-6-(1H-吡唑-4-基)苯基)哌嗪-1-羧酸叔丁酯6e
将化合物1e(489mg,1.20mmol)、化合物6d(434mg,1.20mmol)、三氟乙酸(640mg,5.61mmol)加入到异丙醇(40mL)中,加热升温至90℃反应16小时。停止加热,待反应冷却至室温,减压除去溶剂,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物6e(150mg,产率:17%)。
MS m/z(ESI):676.2[M-55]。
第五步
(Z)-4-(45-溴-25-氟-62-(N-甲基甲磺酰氨基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌嗪-1-羧酸叔丁酯6f
称取化合物6e(145mg,0.20mmol)、1,4-二溴丁烷(86mg,0.40mmol)到50mL茄形瓶中,加入N,N-二甲基甲酰胺(5mL)、碳酸钾(109mg,0.79mmol)、四丁基碘化铵(37mg,0.10mmol),然后50℃反应10小时。停止加热,向体系中加入水(8mL),用乙酸乙酯萃取(15mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL),无水硫酸钠干燥。过滤,减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物6f(70mg,产率:45%)。
MS m/z(ESI):786.5[M+1]。
第六步
(Z)-N-(45-溴-25-氟-26-(哌嗪-1-基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺盐酸盐6g
将化合物6f(70mg,0.089mmol)溶于二氯甲烷(2mL),加入4M氯化氢的1,4-二氧六环溶液(2mL),反应2小时,减压除去溶剂,真空干燥,得标题化合物6g(64mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):684.2[M-1]。
第七步
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶-1-基)乙酰基)哌嗪-1-基)-25-氟-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂十一蕃-62-基)-N-甲基甲基磺酰胺6
向25mL三口瓶中依次加入N,N-二甲基甲酰胺(3mL)、化合物6g(64mg,0.089mmol)、化合物4a(41mg,0.089mmol)、O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(33mg,0.087mmol),冰浴下,缓慢加入N,N-二异丙基乙胺(34mg,0.26mmol),反应20分钟。反应液过滤,经高效液相制备色谱法纯化(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:40%-60%,流速:30mL/min),得到标题化合物6(外消旋体,35mg,产率:36%)。
MS m/z(ESI):1089.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.56(s,1H),9.52(s,1H),8.25(d,2H),8.07(s,1H),7.91(d,1H),7.59(dd,2H),7.53(s,1H),7.11-7.05(m,2H),7.03(s,1H),6.77(d,1H),6.26(dd,1H),4.37-4.20(m,3H),3.99(s,3H),3.92(t,2H),3.79-3.68(m,2H),3.57-3.24(m,6H),3.21(s,3H),3.09(s,3H),3.05-2.79(m,5H),2.76(t,2H),2.73-2.59(m,2H),2.30-2.18(m,1H),2.14-2.04(m,2H),2.03-1.95(m,1H),1.93-1.81(m,1H),1.74-1.68(m,2H)。
实施例7
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶-1-基)乙酰基)哌嗪-1-基)-25-氟-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十二蕃-62-基)-N-甲基甲磺酰胺7

第一步
(Z)-4-(45-溴-25-氟-62-(N-甲基甲磺酰氨基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十二蕃-26-基)-哌嗪-1-羧酸叔丁酯7a
称取化合物6e(150mg,0.20mmol)、1,5-二溴戊烷(71mg,0.31mmol)到25mL茄形瓶中,加入N,N-二甲基甲酰胺(3mL)、碳酸钾(113mg,0.82mmol)、四丁基碘化铵(38mg,0.10mmol),然后50℃反应8小时。停止加热,向体系中加入水(8mL),用乙酸乙酯萃取(15mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL),无水硫酸钠干燥。过滤,减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物7a(40mg,产率:24%)。
MS m/z(ESI):800.4[M+1]。
第二步
(Z)-N-(45-溴-25-氟-26-(哌嗪-1-基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十二蕃-62-基)-N-甲基甲磺酰胺盐酸盐7b
将化合物7a(40mg,0.050mmol)溶于二氯甲烷(2mL),加入4M氯化氢的1,4-二氧六环溶液(2mL),反应2小时,减压除去溶剂,真空干燥,得标题化合物7b(32mg,产率:91%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):700.3[M+1]。
第三步
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶-1-基)乙酰基)哌嗪-1-基)-25-氟-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十二蕃-62-基)-N-甲基甲磺酰胺7
向25mL三口瓶中依次加入N,N-二甲基甲酰胺(3mL)、化合物7b(32mg,0.047mmol)、化合物4a(21mg,0.047mmol)、O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(17.7mg,0.047mmol),冰浴下,缓慢加入N,N-二异丙基乙胺(12mg,0.093mmol),反应20分钟。反应液过滤,经高效液相制备色谱法纯化(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈 的梯度:42%-62%,流速:30mL/min),得到标题化合物7(外消旋体,10mg,产率:19%)。
MS m/z(ESI):1103.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.56(s,1H),9.48(s,1H),8.37(s,1H),8.25(s,1H),8.09(s,1H),7.96(s,1H),7.69(d,1H),7.60(d,1H),7.53(s,1H),7.14(d,1H),7.09(d,1H),7.04(s,1H),6.73(d,1H),6.34(dd,1H),4.44-4.30(m,1H),4.17-4.10(m,2H),3.99(s,3H),3.98-3.94(m,2H),3.92(t,2H),3.60-3.28(m,5H),3.25(s,3H),3.24-3.19(m,1H),3.13(s,3H),3.09-2.80(m,6H),2.76(t,2H),2.73-2.63(m,1H),2.48-2.43(m,1H),2.30-2.18(m,1H),1.95-1.82(m,3H),1.54-1.46(m,2H),1.38-1.30(m,2H)。
实施例8
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶-1-基)-2-氧代乙基)-4-羟基哌啶-1-基)-25-氟-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺8

第一步
1-(2-溴-6-氟-4-硝基苯基)哌啶-4-酮8a
称取化合物6a(400mg,1.68mmol)、4-哌啶酮盐酸盐(221mg,1.63mmol,阿达玛斯试剂有限公司)到50mL茄形瓶中,加入N,N-二甲基甲酰胺(5mL)、二异丙基乙胺(660mg,5.11mmol),然后100℃反应16小时。待反应液冷却至室温,向体系中加入水(15mL),乙酸乙酯萃取(20mL×2),合并有机相,用饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥。过滤,减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物8a(518mg,产率:97%)。
MS m/z(ESI):317.0[M+1]。
第二步
2-(1-(2-溴-6-氟-4-硝基苯基)-4-羟基哌啶-4-基)乙酸叔丁酯8b
称取乙酸叔丁酯(220mg,1.89mmol,阿达玛斯试剂有限公司)到50mL茄形瓶中,加入四氢呋喃(15mL),降温到-78℃,氮气氛下加入二异丙基氨基锂(2M,1.6mL),反应1小时。加入化合物8a(500mg,1.58mmol),反应30分钟。加入水(20mL)淬灭反应,用乙酸乙酯萃取(15mL×3),合并有机相,用饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物8b(680mg,产率:99%)。
MS m/z(ESI):433.0[M+1]。
第三步
2-(1-(2-氟-4-硝基-6-(1H-吡唑-4-基)苯基)-4-羟基哌啶-4-基)乙酸叔丁酯8c
称取化合物8b(675mg,1.56mmol)、4-吡唑硼酸频哪醇酯(333mg,1.72mmol)到100mL茄形瓶中,加入1,1′-双(二苯基膦)二茂铁二氯化钯(228mg,0.31mmol)、碳酸钾(430mg,3.11mmol)、1,4-二氧六环和水的混合溶剂(9mL,V/V=8/1),然后100℃反应8小时。待反应冷却到室温,反应液用硅藻土过滤,滤饼用乙酸乙酯洗涤(15mL×3),滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物8c(120mg,产率:18%)。
MS m/z(ESI):421.5[M+1]。
第四步
2-(1-(4-氨基-2-氟-6-(1H-吡唑-4-基)苯基)-4-羟基哌啶-4-基)乙酸叔丁酯8d
将化合物8c(115mg,0.27mmol)溶于甲醇(10mL),加入10%干钯碳(20mg,0.19mmol),氢气置换三次后,氢气氛下反应5小时。反应液经硅藻土过滤,滤液减压浓缩,得到粗产品标题化合物8d(101mg,产率:95%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):391.2[M+1]。
第五步
2-(1-(4-((5-溴-4-((4-羟基-2-(N-甲基甲磺酰氨基)苯基)氨基)嘧啶-2-基)氨基)-2-氟-6-(1H-吡唑-4-基)苯基)-4-羟基哌啶-4-基)乙酸叔丁酯8e
将化合物1e(102mg,0.25mmol)、化合物8d(93mg,0.24mmol)、三氟乙酸(55mg,0.48mmol)加入到正丁醇(10mL)中,加热升温至90℃反应16小时。待反应液冷却至室温,减压除去溶剂,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物8e(96mg,产率:53%)。
MS m/z(ESI):759.0[M-1]。
第六步
(Z)-(2-(1-(45-溴-25-氟-62-(N-甲基甲磺酰氨基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)-4-羟基哌啶-4-基)乙酸叔丁酯8f
称取化合物8e(90mg,0.12mmol)、1,4-二溴丁烷(32mg,0.15mmol)到50mL茄形瓶中,加入N,N-二甲基甲酰胺(5mL)、碳酸钾(66mg,0.48mmol)、四丁基碘化铵(23mg,0.62mmol),然后50℃反应16小时。待反应液冷却至室温,向体系中加入水(8mL),乙酸乙酯萃取(15mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL),无水硫酸钠干燥,过滤,减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物8f(60mg,产率:62%)。
MS m/z(ESI):815.1[M+1]。
第七步
(Z)-(2-(1-(45-溴-25-氟-62-(N-甲基甲磺酰胺基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)-4-羟基哌啶-4-基)乙酸8g
将化合物8f(40mg,0.050mmol)溶于二氯甲烷(10mL),加入4M氯化氢的1,4-二氧六环溶液(2mL),反应搅拌16小时。减压除去溶剂,真空干燥,得粗产品标题化合物8g(33mg,产率:89%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):759.0[M+1]。
第八步
(Z)-N-(45-溴-26-(4-(2-(4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶-1-基)-2-氧代乙基)-4-羟基哌啶-1-基)-25-氟-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺8
向25mL三口瓶中依次加入N,N-二甲基甲酰胺(3mL)、化合物8g(33mg,0.044mmol)、1-(6-(3,3-二氟哌啶-4-基)-1-甲基-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮盐酸盐8h(18mg,0.045mmol,采用公知的方法“WO2021127561A1中说明书第383页”制备得到)、O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(23mg,0.060mmol),冰水浴条件下,缓慢加入N,N-二异丙基乙胺(35mg,0.27mmol),反应30分钟。反应液过滤,经高效液相制备色谱法纯化(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:45%-65%,流速:30mL/min),得到标题化合物8(外消旋体,11mg,产率:23%)。
MS m/z(ESI):1104.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.56(s,1H),9.47(s,1H),8.23(s,2H),8.05(s,1H),7.88(t,1H),7.63-7.50(m,3H),7.10-6.99(m,3H),6.74(d,1H),6.24(dd,1H),4.87(t,1H),4.68(d,1H),4.64-4.58(m,1H),4.29-4.19(m,2H),3.99(d,3H),3.92(t,2H),3.72(s,2H),3.65-3.53(m,2H),3.40-3.34(m,3H),3.21(s,3H),3.08(s,3H),2.76(t,3H),2.60-2.53(m,3H),2.11-2.03(m,2H),1.97-1.92(m,1H),1.88-1.78(m,3H),1.74-1.67(m,2H),1.62-1.52(m,2H)。
实施例9
(±)-(Z)-N-(45-溴-26-(4-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)甲基)哌嗪-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺9
将化合物1h(50mg,0.086mmol)溶于二氯甲烷和甲醇的混合溶剂(5mL,V/V=1/1)中,加入(±)-1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-甲醛9a(39mg,0.11mmol,采用专利申请“WO2021077010A1中说明书第488页的化合物188”公开的方法制备而得),乙酸(13mg,0.21mmol),反应1小时。加入氰基硼氢化钠(13mg,0.21mmol),反应16小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:40%-60%,流速:30mL/min),得到标题化合物9(外消旋体,25mg,产率:33%)。
MS m/z(ESI):1051.1[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.07(s,1H),8.23(s,1H),8.19(s,1H),8.08(s,1H),7.88(s,1H),7.75(s,1H),7.72(d,1H),7.65(d,1H),7.31(d,1H),7.26-7.21(m,2H),7.06(d,1H),6.76(s,1H),6.10(dd,1H),5.06(dd,1H),4.24(t,2H),4.08-4.00(m,2H),3.84(s,3H),3.79(t,2H),3.17(s,3H),3.07(s,3H),2.96(t,2H),2.92-2.83(m,1H),2.82-2.74(m,4H),2.65-2.35(m,6H),2.23-2.13(m,2H),2.07-1.95(m,3H),1.89-1.75(m,3H),1.67-1.60(m,2H),1.19-1.10(m,2H)。
实施例10
(±)-(Z)-N-(45-溴-26-(2-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)甲基)-2,7-二氮杂螺[3.5]壬烷-7-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺10

第一步
7-(2-溴-5-甲氧基-4-硝基苯基)-2,7-二氮杂螺[3.5]壬烷-2-羧酸叔丁酯10a
称取化合物1a(2.0g,7.99mmol)、2,7-二氮杂螺[3.5]壬烷-2-羧酸叔丁酯(1.81g,7.99mmol,上海毕得医药)到100mL茄形瓶中,加入N,N-二甲基甲酰胺(20mL)、碳酸钾(2.20g,15.99mmol),然后90℃反应12小时。待反应冷却至室温,向体系中加入水(30mL),乙酸乙酯萃取(30mL×3),合并有机相,用饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥。过滤,减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物10a(3.0g,产率:87%)。
MS m/z(ESI):456.0[M+1]。
第二步
7-(5-甲氧基-4-硝基-2-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯基)-2,7-二氮杂螺[3.5]壬烷-2-羧酸叔丁酯10b
称取化合物10a(1.1g,2.40mmol)、1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-硼酸频哪醇酯(972mg,3.50mmol,上海毕得医药)到100mL茄形瓶中,依次加入1,1′-双(二苯基膦)二茂铁二氯化钯(175mg,0.24mmol)、碳酸钠(767mg,7.23mmol)、1,4-二 氧六环和水的混合溶剂(30mL,V/V=5/1),升温至100℃反应16小时。待反应冷却到室温,反应液用硅藻土过滤,滤饼用乙酸乙酯洗涤(30mL),滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物10b(1.20g,产率:94%)。
MS m/z(ESI):528.2[M+1]。
第三步
7-(4-氨基-5-甲氧基-2-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯基)-2,7-二氮杂螺[3.5]壬烷-2-羧酸叔丁酯10c
将化合物10b(990mg,1.87mmol)溶于甲醇(25mL),加入10%湿钯碳(含55%质量分数水,594mg,2.44mmol,韶远化学科技),氢气置换三次后,氢气气氛下反应16小时。反应液经硅藻土过滤,滤液减压浓缩,得到粗产品标题化合物10c(943mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):498.2[M+1]。
第四步
7-(4-((5-溴-4-((4-羟基-2-(N-甲基甲磺酰胺基)苯基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-(1H-吡唑-4-基)苯基)-2,7-二氮杂螺[3.5]壬烷-2-羧酸叔丁酯10d
将化合物1e(600mg,1.47mmol)、化合物10c(915mg,1.83mmol)、三氟乙酸(671mg,5.88mmol)加入到异丙醇(60mL)中,升温至90℃反应16小时。待反应液冷却至室温,减压除去溶剂,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物10d(0.5g,产率:43%)。
MS m/z(ESI):784.2[M+1]。
第五步
(Z)-7-(45-溴-24-甲氧基-62-(N-甲基甲磺酰胺基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)-2,7-二氮杂螺[3.5]壬烷-2-羧酸叔丁酯10e
称取化合物10d(500mg,0.60mmol)、1,4-二溴丁烷(151mg,0.75mmol)到50mL茄形瓶中,加入N,N-二甲基甲酰胺(8mL)、碳酸钾(350mg,2.51mmol)、四丁基碘化铵(117mg,0.31mmol),反应16小时。向体系中加入水(10mL),用乙酸乙酯萃取(15mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物10e(150mg,产率:28%)。
MS m/z(ESI):838.2[M+1]。
第六步
(Z)-N-(45-溴-24-甲氧基-26-(2,7-二氮杂螺[3.5]壬烷-7-基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺三氟乙酸盐10f
将化合物10e(60mg,0.072mmol)溶于二氯甲烷(2.4mL),加入三氟乙酸(0.6mL),反应1小时,向体系中加入饱和碳酸氢钠溶液(20mL),用二氯甲烷萃取(15mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(20mL),无水硫酸钠干燥。过滤,滤液减压浓缩,得标题化合物粗产物10f(52mg,产率:98%)。
MS m/z(ESI):738.2[M+1]。
第七步
(±)-(Z)-N-(45-溴-26-(2-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)甲基)-2,7-二氮杂螺[3.5]壬烷-7-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺10
将化合物10f(52mg,0.070mmol)溶于二氯甲烷和甲醇的混合溶剂中(5mL,V/V=1/1),加入乙酸(13mg,0.21mmol)。加入化合物9a(31mg,0.084mmol),反应1小时。加入氰基硼氢化钠(13mg,0.21mmol),反应16小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:45%-65%,流速:30mL/min),得到标题化合物10(外消旋体混合物,25mg,产率:32%)。
MS m/z(ESI):1091.1[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.05(s,1H),8.24(s,1H),8.18(s,1H),8.07(s,1H),7.87(s,1H),7.78-7.68(m,2H),7.64(d,1H),7.29(d,1H),7.24(s,1H),7.21(dd,1H),7.05(d,1H),6.73(s,1H),6.10(dd,1H),5.05(dd,1H),4.22(t,2H),4.02(d,2H),3.84(s,3H),3.78(t,2H),3.17(s,3H),3.07(s,3H),2.99-2.83(m,7H),2.72-2.52(m,6H),2.30(d,2H),2.07-1.95(m,3H),1.84-1.68(m,6H),1.67-1.60(m,3H),1.19-1.07(m,2H)。
实施例11
(±)-(Z)-N-(45-溴-26-(4-(4-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)甲基)哌嗪-1-基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺11

第一步
4-(1-(2-溴-5-甲氧基-4-硝基苯基)哌啶-4-基)哌嗪-1-羧酸叔丁酯11a
称取化合物1a(3g,12.00mmol)、4-(哌啶-4-基)哌嗪-1-羧酸叔丁酯(3.24g,12.03mmol,上海毕得医药)到100mL茄形瓶中,加入N,N-二甲基甲酰胺(30mL)、N,N-二异丙基乙胺(3.10g,23.99mmol),然后90℃反应12小时。待反应冷却至室温,向反应液中加入水(30mL),用乙酸乙酯萃取(30mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥,过滤,滤液减压浓缩,用正己烷和乙酸乙酯的混合溶剂(20mL,V/V=5/1)洗涤,过滤,收集滤饼,真空干燥,得到粗产品标题化合物11a(5.0g,产率:83%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):497.0[M-1]。
第二步
4-(1-(5-甲氧基-4-硝基-2-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯基)哌啶-4-基)哌嗪-1-羧酸叔丁酯11b
称取化合物11a(1.0g,2.00mmol)、1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-硼酸频哪醇酯(835mg,3.00mmol)到100mL茄形瓶中,依次加入1,1′-双(二苯基膦)二茂铁二氯化钯(146mg,0.20mmol)、碳酸钠(637mg,6.0mmol)、1,4-二氧六环和水的混合溶剂(20mL,V/V=5/1),升温至100℃反应16小时。待反应冷却到室温,反应液用硅藻土过滤,滤饼用乙酸乙酯洗涤(30mL),滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物11b(940mg,产率:99%)。
MS m/z(ESI):571.2[M+1]。
第三步
4-(1-(4-氨基-5-甲氧基-2-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯基)哌啶-4-基)哌嗪-1-羧酸叔丁酯11c
称取化合物11b(1.0g,1.75mmol)到100mL单口瓶中,加入乙醇和水的混合溶剂(36mL,V/V=5/1),然后加入铁粉(980mg,17.55mmol)和氯化铵(1.87g,34.96mmol)。升温到60℃反应16小时。待反应液冷却到室温,反应液用硅藻土过滤,滤液减压浓缩,向所得残余物加入乙酸乙酯(30mL),过滤除去不溶物,滤液减压浓缩,真空干燥,得到粗产品标题化合物11c(1.0g,产率:88%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):541.3[M+1]。
第四步
4-(1-(4-((5-溴-4-((4-羟基-2-(N-甲基甲磺酰胺基)苯基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-(1H-吡唑-4-基)苯基)哌啶-4-基)哌嗪-1-羧酸叔丁酯11d
将化合物1e(700mg,1.72mmol)、化合物11c(930mg,1.73mmol)、三氟乙酸(395mg,3.46mmol)加入到异丙醇(60mL)中,升温至90℃反应16小时。待反应液冷却至室温,减压除去溶剂,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物11d(1.3g,产率:91%)。
MS m/z(ESI):827.1[M+1]。
第五步
(Z)-4-(1-(45-溴-24-甲氧基-62-(N-甲基甲磺酰胺基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)哌嗪-1-羧酸叔丁酯11e
称取化合物11d(500mg,0.60mmol)、1,4-二溴丁烷(130mg,0.25mmol)到50mL茄形瓶中,加入N,N-二甲基甲酰胺(5mL)、碳酸钾(250mg,1.81mmol)、四丁基碘化铵(45mg,0.12mmol),反应16小时。向反应液中加入水(10mL),用乙酸乙酯萃取(15mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(20mL),无水硫 酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物11e(109mg,产率:20%)。
MS m/z(ESI):878.7[M-1]。
第六步
(Z)-N-(45-溴-24-甲氧基-26-(4-(哌嗪-1-基)哌啶-1-基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺二盐酸盐11f
将化合物11e(109mg,0.12mmol)溶于二氯甲烷(1mL),加入4M氯化氢的1,4-二氧六环溶液(1mL),反应1小时。减压除去溶剂,真空干燥,得到粗产品标题化合物11f(100mg,产率:95%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):781.1[M+1]。
第七步
(±)-(Z)-N-(45-溴-26-(4-(4-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)甲基)哌嗪-1-基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺11
将化合物11f(60mg,0.70mmol)溶于二氯甲烷和甲醇的混合溶剂中(2mL,V/V=1/1),加入无水乙酸钠(57mg,0.70mmol),反应10分钟。加入化合物9a(38mg,0.10mmol),反应1小时。加入氰基硼氢化钠(8mg,0.14mmol),反应30分钟。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:58%-68%,流速:30mL/min),得到标题化合物11(外消旋体混合物,18mg,产率:22%)。
MS m/z(ESI):1134.1[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.07(s,1H),8.23(s,1H),8.19(s,1H),8.08(s,1H),7.88(s,1H),7.75(s,1H),7.72(d,1H),7.65(d,1H),7.31(d,1H),7.26-7.21(m,2H),7.06(d,1H),6.76(s,1H),6.10(dd,1H),5.06(dd,1H),4.30-4.18(m,2H),4.04(d,2H),3.84(s,3H),3.82-3.75(m,2H),3.17(s,3H),3.07(s,3H),3.05-2.83(m,6H),2.65-2.35(m,8H),2.23-2.13(m,2H),2.07-1.95(m,6H),1.89-1.75(m,6H),1.67-1.55(m,3H),1.43-1.36(m,1H),1.19-1.10(m,2H)。
实施例12
N-((Z)-45-溴-26-(4-(4-((2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-4-基)-L-丙氨酰基)哌嗪-1-基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺12
向25mL单口瓶中依次加入N,N-二甲基甲酰胺(2mL)、化合物11f(60mg,0.073mmol)、(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-4-基)-L-丙氨酸12a(25mg,0.072mmol,采用专利申请“WO2020051235A1中说明书第419页的化合物235”公开的方法制备而得)、O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(41mg,0.11mmol),冰浴下,缓慢加入N,N-二异丙基乙胺(29mg,0.22mmol),反应20分钟。反应液过滤,经高效液相制备色谱法纯化(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:40%-85%,流速:30mL/min),得到标题化合物12(2个非对应异构体的混合物,20mg,产率:25%)。
MS m/z(ESI):1108.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.10(s,1H),8.23(s,1H),8.19(s,1H),8.08(s,1H),7.88(s,1H),7.77-7.68(m,2H),7.62(t,1H),7.25(s,1H),7.23(d,1H),7.18-7.14(m,1H),7.10-7.04(m,2H),6.76(s,1H),6.10(d,1H),5.07(dd,1H),4.93-4.86(m,1H),4.27-4.18(m,2H),3.84(s,3H),3.82-3.76(m,2H),3.74-3.26(m,6H),3.17(s,3H),3.07(s,3H),3.02(d,2H),2.93-2.84(m,1H),2.68-2.55(m,4H),2.36-2.20(m,2H),2.08-1.96(m,4H),1.84-1.77(m,2H),1.67-1.41(m,4H),1.29(d,3H)。
实施例13
(±)-(Z)-N-(45-溴-26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺13

第一步
1-(2-溴-5-甲氧基-4-硝基苯基)-4-(二甲氧基甲基)哌啶13a
称取化合物1a(3.2g,12.80mmol)、4-(二甲氧基甲基)哌啶(2.45g,15.39mmol,江苏艾康生物医药)到100mL茄形瓶中,加入N,N-二甲基甲酰胺(30mL)、无水碳酸钾(3.54g,25.61mmol),然后升温到90℃反应12小时。待反应冷却至室温,向反应液中加入水(30mL),用乙酸乙酯萃取(30mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物13a(3.8g,产率:76%)。
MS m/z(ESI):389.0[M+1]。
第二步
4-(二甲氧基甲基)-1-(5-甲氧基-4-硝基-2-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯基)哌啶13b
称取化合物13a(1.0g,2.57mmol)、1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-硼酸频哪醇酯(715mg,2.57mmol)到100mL茄形瓶中,依次加入1,1′-双(二苯基膦)二茂铁二氯化钯(188mg,0.26mmol)、碳酸钠(817mg,7.71mmol)、1,4-二氧六环和水的混合溶剂(18mL,V/V=5/1),升温至100℃反应12小时。待反应液冷却到室温,反应液用硅藻土过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物13b(1.12g,产率:95%)。
MS m/z(ESI):461.2[M+1]。
第三步
4-(4-(二甲氧基甲基)哌啶-1-基)-2-甲氧基-5-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯胺13c
称取化合物13b(1.12g,2.43mmol)到100mL单口瓶中,加入甲醇(10mL),然后加入10%湿钯碳(含55%质量分数水,259mg,0.24mmol)。氢气置换三次,氢气氛下反应12小时。反应液用硅藻土过滤,滤液浓缩,真空干燥,得到粗产品标题化合物13c(1g,产率:96%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):431.1[M+1]。
第四步
N-(2-((5-溴-2-((4-(4-(二甲氧基甲基)哌啶-1-基)-2-甲氧基-5-(1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)-5-羟基苯基)-N-甲基甲磺酰胺13d
将化合物1e(474mg,1.16mmol)、化合物13c(500mg,1.16mmol)、三氟乙酸(265mg,2.32mmol)加入到甲醇(5mL)中,升温至90℃反应12小时。待反应液冷却至室温,减压除去溶剂,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物13d(500mg,产率:60%)。
MS m/z(ESI):717.1[M+1]。
第五步
(Z)-N-(45-溴-26-(4-(二甲氧基甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺13e
称取化合物13d(300mg,0.42mmol)、1,4-二溴丁烷(91mg,0.42mmol)到50mL茄形瓶中,加入N,N-二甲基甲酰胺(5mL)、碳酸钾(174mg,1.26mmol)、四丁基碘化铵(42mg,0.13mmol),反应16小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-85%,流速:30mL/min),得到标题化合物13e(60mg,产率:19%)。
MS m/z(ESI):771.2[M+1]。
第六步
(Z)-N-(45-溴-26-(4-甲酰基哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺13f
称取化合物13e(60mg,0.08mmol)到50mL茄形瓶中,加入四氢呋喃(3mL),然后加入2M硫酸溶液(0.2mL),在30℃反应3小时。向体系中饱和碳酸氢钠溶液(5mL),用乙酸乙酯萃取(15mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(20mL),无水硫酸钠干燥,过滤,减压浓缩,得到粗产品标题化合物13f(56mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):725.1[M+1]。
第七步
(±)-(Z)-N-(45-溴-26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺13
将化合物13f(56mg,0.08mmol)溶于4mL二氯甲烷和甲醇的混合溶剂中(V/V=3/1),加入无水乙酸钠(63mg,0.80mmol),反应10分钟。加入(±)-2-(2,6-二氧代哌啶-3-基)-5-(哌嗪-1-基)异吲哚啉-1,3-二酮盐酸盐13g(41mg,0.11mmol,采用公知的方法“WO20211077010A1中说明书第398页实施例104”制备得到),反应1小时。加入氰基硼氢化钠(9mg,0.16mmol),反应30分钟。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-75%,流速:30mL/min),得到标题化合物13(外消旋体混合物,60mg,产率:74%)。
MS m/z(ESI):1051.2[M+1]。
1H NMR(500MHz,DMSO-d6):11.07(s,1H),8.22(s,1H),8.19(s,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.72(d,1H),7.68(d,1H),7.34(d,1H),7.28-7.21(m,2H),7.06(d,1H),6.77(s,1H),6.11(dd,1H),5.07(dd,1H),4.26-4.18(m,2H),3.85(s,3H),3.82-3.74(m,2H),3.49-3.41(m,4H),3.17(s,3H),3.07(s,3H),3.02-2.95(m,2H),2.92-2.83(m,1H),2.65-2.44(m,8H),2.28(d,2H),2.06-1.95(m,3H),1.79-1.71(m,2H),1.68-1.57(m,3H),1.35-1.27(m,2H)。
实施例13-1
N-((Z)-45-溴-26-(4-((4-(2-((S)-2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺13-1

第一步
(S)-5-氨基-4-(5-氟-1,3-二氧代异吲哚啉-2-基)-5-氧代戊酸叔丁酯13-1b
称取(S)-4,5-二氨基-5-氧代戊酸叔丁酯13-1a(500mg,3.01mmol,上海毕得医药)、4-氟邻苯二甲酸酐(500mg,3.01mmol,江苏艾康生物医药有限公司)到50mL茄形瓶中,加入5mL乙酸乙酯。冰水浴冷至10℃,滴加N,N′-羰基二咪唑(735mg,4.53mmol)的N,N-二甲基甲酰胺溶液(5mL),滴完后恢复至室温反应1.5小时。向反应液中加入水(30mL),用乙酸乙酯萃取(30mL×3),合并有机相,有机相依次用饱和碳酸氢钠溶液(30mL),饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥。过滤,滤液减压浓缩,得到标题化合物13-1b(930mg,产率:88%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):295.0[M-55]。
第二步
(S)-4-(2-(1-氨基-5-(叔丁氧基)-1,5-二氧代戊-2-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-羧酸苄酯13-1c
称取化合物13-1b(900mg,2.57mmol)和哌嗪-1-羧酸苄酯(800mg,3.63mmol,韶远(科技)上海有限公司)到50mL茄形瓶中,加入二甲基亚砜(10mL)和N,N-二异丙基乙胺(1.3g,10.06mmol),升温至100℃反应5小时。待反应液冷却到室温,将反应液倒入50mL冰水中,用乙酸乙酯萃取(30mL×3),合并有机相,有机相用饱和氯化钠溶液(30mL×2)洗涤,无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物13-1c(860mg,产率:57%)。
MS m/z(ESI):495.7[M-55]。
第三步
(S)-5-氨基-4-(1,3-二氧代-5-(哌嗪-1-基)异吲哚啉-2-基)-5-氧代戊酸叔丁酯13-1d
称取化合物13-1c(800mg,1.45mmol)到100mL单口瓶中,加入乙酸乙酯(20mL),然后加入10%湿钯碳(含55%质量分数的水,155mg,0.15mmol)。氢气置换三次,氢气气氛下反应12小时。反应液用硅藻土过滤,滤液浓缩,真空干燥,得到粗产品标题化合物13-1d(480mg,产率:75%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):417.2[M+1]。
第四步
(4S)-5-氨基-4-(5-(4-((1-((Z)-45-溴-24-甲氧基-62-(N-甲基甲磺酰胺基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)哌嗪-1-基)-1,3-二氧代异吲哚啉-2-基)-5-氧代戊酸叔丁酯13-1e
向25mL单口瓶中加入化合物13f(375mg,0.51mmol)、化合物13-1d(315mg,0.64mmol)、二氯甲烷(9mL)和甲醇(3mL),搅拌均匀后加入冰乙酸(95mg,1.58mmol),反应1小时。加入氰基硼氢化钠(63mg,1.05mmol),反应1小时。加入饱和碳酸氢钠溶液(10mL),用乙酸乙酯萃取(30mL×3),合并有机相,有机相用饱和氯化钠溶液(30mL×2)洗涤,无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物13-1e(360mg,产率:61%)。
MS m/z(ESI):1125.2[M+1]。
第五步
N-((Z)-45-溴-26-(4-((4-(2-((S)-2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺13-1
称取化合物13-1e(360mg,0.32mmol)和乙腈(40mL)到100mL茄形瓶中,加入苯磺酸(305mg,1.93mmol),升温至85℃反应16小时。反应液浓缩,残余物经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:40%-65%,流速:30mL/min),得到标题化合物13-1(180mg,产率:53%)。
MS m/z(ESI):1051.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.08(s,1H),8.22(s,1H),8.19(s,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.72(d,1H),7.68(d,1H),7.34(d,1H),7.28-7.21(m,2H),7.06(d,1H),6.77(s,1H),6.11(dd,1H),5.07(dd,1H),4.26-4.18(m,2H),3.85(s,3H),3.82-3.74(m,2H),3.49-3.41(m,4H),3.17(s,3H),3.07(s,3H),3.02-2.95(m,2H),2.92-2.83(m,1H),2.65-2.44(m,8H),2.28(d,2H),2.06-1.95(m,3H),1.79-1.71(m,2H),1.68-1.57(m,3H),1.35-1.27(m,2H)。
实施例13-1和13-2
N-((Z)-45-溴-26-(4-((4-(2-((S)-2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺13-1
N-((Z)-45-溴-26-(4-((4-(2-((R)-2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺13-2
将化合物13(50mg)经手性拆分(分离条件:Gilson-281,色谱柱:CHIRALPAK IE(20×250mM),流动相:乙腈/乙醇溶液=40/60(v/v),流速20mL/min)得到标题化合物13-1(10mg)和标题化合物13-2(10mg)。
化合物13-1:
MS m/z(ESI):1051.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.08(s,1H),8.22(s,1H),8.19(s,1H),8.08(s,1H),7.89(s,1H),7.74(s,1H),7.72(d,1H),7.68(d,1H),7.34(d,1H),7.28-7.21(m,2H),7.06(d,1H),6.77(s,1H),6.11(dd,1H),5.07(dd,1H),4.26-4.18(m,2H),3.85(s,3H),3.82-3.74(m,2H),3.49-3.41(m,4H),3.17(s,3H),3.07(s,3H),3.02-2.95(m,2H), 2.92-2.83(m,1H),2.65-2.44(m,8H),2.28(d,2H),2.06-1.95(m,3H),1.79-1.71(m,2H),1.68-1.57(m,3H),1.35-1.27(m,2H)。
手性HPLC分析方法:保留时间10.38分钟(Agilent1260 DAD,色谱柱:CHIRALPAK IE(4.6×150mM),5μm;流动相:乙腈/乙醇(含0.1%二乙胺)=40/60(v/v),流速1mL/min)。
化合物13-2:
MS m/z(ESI):1051.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),8.22(s,1H),8.19(s,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.72(d,1H),7.68(d,1H),7.34(d,1H),7.28-7.21(m,2H),7.06(d,1H),6.77(s,1H),6.11(dd,1H),5.07(dd,1H),4.26-4.18(m,2H),3.85(s,3H),3.82-3.74(m,2H),3.49-3.41(m,4H),3.17(s,3H),3.07(s,3H),3.02-2.95(m,2H),2.92-2.83(m,1H),2.65-2.44(m,8H),2.28(d,2H),2.06-1.95(m,3H),1.79-1.71(m,2H),1.68-1.57(m,3H),1.35-1.27(m,2H)。
手性HPLC分析方法:保留时间17.52分钟(Agilent1260 DAD,色谱柱:CHIRALPAK IE(4.6×150mM),5μm;流动相:乙腈/乙醇(含0.1%二乙胺)=40/60(v/v),流速1mL/min)。
实施例14
(±)-(Z)-N-(45-溴-26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-环丙基甲磺酰胺14

第一步
N-环丙基甲磺酰胺14b
将环丙胺14a(250mg,4.38mmol,韶远化学科技)溶于二氯甲烷(10mL)中,加入N,N-二异丙基乙胺(565mg,4.38mmol)、吡啶(346mg,4.38mmol)。在冰浴条件下加入甲基磺酰氯(500mg,4.38mmol),反应12小时。待反应完全后,减压除去溶剂,真空干燥,得到粗产品标题化合物14b(590mg,产率:99%),粗产品不经纯化直接用于下一步反应。
第二步
N-环丙基-N-(5-甲氧基-2-硝基苯基)甲磺酰胺14d
将化合物14b(440mg,3.25mmol)和2-氟-4-甲氧基-1-硝基苯14c(557mg,3.25mmol,上海毕得医药)溶于乙腈(10mL)中,加入碳酸铯(3.19g,9.79mmol),升温到90℃反应12小时。待反应液冷却到室温,减压浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系B纯化得到标题化合物14d(600mg,产率:64%)。
第三步
N-(2-氨基-5-甲氧基苯基)-N-环丙基甲磺酰胺14e
将化合物14d(600mg,2.09mmol)溶于甲醇(10mL)中,加入10%湿钯碳(含55%质量分数水,223mg,0.21mmol)。氢气置换三次,氢气气氛下反应12小时。 反应液用硅藻土过滤,滤液浓缩,真空干燥,得到粗产品标题化合物14e(526mg,产率:98%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):257.1[M+1]。
第四步
N-(2-((5-溴-2-氯嘧啶-4-基)氨基)-5-甲氧基苯基)-N-环丙基甲磺酰胺14g
将化合物14e(400mg,1.56mmol)和5-溴-2,4-二氯嘧啶14f(356mg,1.56mmol,上海毕得医药)溶于异丙醇(10mL)中,加入N,N-二异丙基乙胺(1.03g,7.97mmol),升温到100℃反应12小时。反应液减压浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物14g(400mg,产率:57%)。
MS m/z(ESI):447.0[M+1]。
第五步
N-(2-((5-溴-2-氯嘧啶-4-基)氨基)-5-羟基苯基)-N-环丙基甲磺酰胺14h
将化合物14g(300mg,0.67mmol)溶于二氯甲烷(3mL)中,加入1M三溴化硼的二氯甲烷溶液(3.36mL,3.36mmol),反应12小时。反应液用饱和碳酸氢钠溶液淬灭,用乙酸乙酯萃取(10mL×3),合并有机相,有机相用饱和氯化钠溶液(15mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物14h(280mg,产率:96%)。
MS m/z(ESI):432.8[M+1]。
第六步
N-(2-((5-溴-2-((4-(4-(二甲氧基甲基)哌啶-1-基)-2-甲氧基-5-(1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)-5-羟基苯基)-N-环丙基甲磺酰胺14i
将化合物13c(500mg,1.16mmol)和化合物14h(504mg,1.16mmol)溶于甲醇(10mL),加入三氟乙酸(397mg,3.48mmol),90℃反应12小时。反应液减压浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物14i(320mg,产率:37%)。
MS m/z(ESI):743.0[M+1]。
第七步
(Z)-N-(45-溴-26-(4-(二甲氧基甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-环丙基甲磺酰胺14j
将化合物14i(100mg,0.13mmol)溶于N,N-二甲基甲酰胺(3mL),加入1,4-二溴丁烷(35mg,0.16mmol)、无水碳酸钾(556mg,0.40mmol)、四丁基碘化铵(13mg,0.04mmol),反应30分钟。然后升温至60℃反应2小时,补加1,4-二溴丁烷(25mg,0.12mmol),升温到80℃反应7小时。反应液冷却至室温,加入水(10mL),用乙酸乙酯萃取(10mL×3),合并有机相,有机相依次用水(15mL×2)和饱和氯化钠溶液 (15mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物14j(47mg,产率:44%)。
MS m/z(ESI):797.2[M+1]。
第八步
(Z)-N-(45-溴-26-(4-甲酰基哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-环丙基甲磺酰胺14k
将化合物14j(47mg,0.059mmol)溶于四氢呋喃(5mL),加入2M硫酸(0.1mL),30℃反应1小时。将反应液过滤,收集滤饼,滤饼烘干,得到粗产品标题化合物14k(42mg,产率:94%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):751.2[M+1]。
第九步
(±)-(Z)-N-(45-溴-26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-环丙基甲磺酰胺14
称取化合物13g(30mg,0.079mmol)到25mL茄形瓶中,加入4mL二氯甲烷和甲醇的混合溶剂(V/V=3/1),加入无水乙酸钠(48mg,0.59mmol),反应15分钟。加入化合物14k(42mg,0.056mmol),反应30分钟,加入氰基硼氢化钠(7mg,0.12mmol),反应1小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:57%-63%,流速:30mL/min),得到标题化合物14(外消旋体混合物,10mg,产率:18%)。
MS m/z(ESI):1075.3[M-1]。
1H NMR(500MHz,DMSO-d6)δ11.08(s,1H),8.24(s,1H),8.18(s,1H),7.86(s,1H),7.79(d,2H),7.75(d,1H),7.68(d,1H),7.34(d,1H),7.29-7.22(m,2H),7.12(d,1H),6.78(s,1H),6.03(dd,1H),5.07(dd,1H),4.30-4.17(m,2H),3.96-3.88(m,1H),3.85(s,3H),3.71-3.63(m,1H),3.48-3.40(m,4H),3.31(s,2H),3.27-3.21(m,1H),3.17(s,3H),3.07-2.83(m,3H),2.68-2.41(m,5H),2.30-2.23(m,2H),2.21-2.11(m,1H),2.05-1.89(m,3H),1.83-1.57(m,5H),1.40-1.20(m,2H),1.02-0.90(m,2H),0.60-0.53(m,1H),0.23-0.16(m,1H)。
实施例15
(±)-(Z)-N-(45-溴-26-(4-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)甲基)哌嗪-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-环丙基甲磺酰胺15
第一步
4-(5-甲氧基-4-硝基-2-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯基)哌嗪-1-羧酸叔丁酯15a
将化合物1b(3.26g,7.83mmol)溶于50mL 1,4-二氧六环和水的混合溶剂(V/V=4/1),加入1-(四氢-2H-吡喃-2-基)-1H-吡唑4-硼酸频哪醇酯(2.61g,9.38mmol)、1,1'-双二苯基膦二茂铁二氯化钯(573mg,0.78mmol)、碳酸钠(2.49g,23.5mmol)。氮气置换三次,升温至90℃反应16小时。待反应冷却到室温,用硅藻土过滤反应液,滤液减压浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系B纯化得到标题化合物15a(2.7g,产率:70%)。
MS m/z(ESI):488.2[M+1]。
第二步
4-(4-氨基-5-甲氧基-2-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯基)哌嗪-1-羧酸叔丁酯15b
将化合物15a(2.7g,5.54mmol)溶于甲醇(50mL),加入10%湿钯碳(含55%质量分数水,671mg,5.55mmol),氢气置换三次,氢气气氛下反应16小时。反应液用硅藻土过滤,滤液减压浓缩,真空干燥,得到粗产品标题化合物15b(1.8g,产率:74%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):458.2[M+1]。
第三步
4-(4-((5-溴-4-((2-(N-环丙基甲磺酰胺基)-4-羟基苯基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-(1H-吡唑-4-基)苯基)哌嗪-1-羧酸叔丁酯15c
将化合物15b(500mg,1.09mmol)和化合物14h(536mg,1.23mmol)溶于异丙醇(10mL),然后加入三氟乙酸(530mg,4.65mmol)。升温到90℃反应16小时。反应液减压浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物15c(370mg,产率:44%)。
MS m/z(ESI):770.0[M+1]。
第四步
(Z)-4-(45-溴-62-(N-环丙基甲磺酰胺基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌嗪-1-羧酸叔丁酯15d
将化合物15c(210mg,0.27mmol)溶于N,N-二甲基甲酰胺(5mL),加入1,4-二溴丁烷(126mg,0.58mmol)、无水碳酸钾(113mg,0.82mmol)和四丁基碘化铵(27mg,0.82mmol)。反应30分钟,升温至60℃,反应2小时,再升温到80℃反应16小时。待反应液冷却到室温,加水(5mL),用乙酸乙酯萃取(10mL×3),合并有机相,有机相用饱和氯化钠溶液(15mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物15d(47mg,产率:21%)。
MS m/z(ESI):824.2[M+1]。
第五步
(Z)-N-(45-溴-24-甲氧基-26-(哌嗪-1-基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-环丙基甲磺酰胺三氟乙酸盐15e
将化合物15d(47mg,0.058mmol)溶于二氯甲烷(2mL),加入三氟乙酸(0.5mL),反应1小时。反应液减压浓缩,真空干燥,得到粗产品标题化合物15e(48mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):724.1[M+1]。
第六步
(±)-(Z)-N-(45-溴-26-(4-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)甲基)哌嗪-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-环丙基甲磺酰胺15
称取化合物15e(47mg,0.056mmol)到25mL茄形瓶中,加入4mL二氯甲烷和甲醇的混合溶剂(V/V=3/1),加入无水乙酸钠(50mg,0.61mmol),反应15分钟。然后加入化合物9a(62mg,0.17mmol)和乙酸(10mg,0.17mmol),反应30分钟。加入氰基硼氢化钠(7mg,0.11mmol),反应1小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:62%-68%,流速:30mL/min),得到标题化合物15(外消旋体混合物,20mg,产率:33%)。
MS m/z(ESI):1077.3[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.07(s,1H),8.26(s,1H),8.19(s 1H),7.86(s,1H),7.82-7.74(m,3H),7.66(d,1H),7.32-7.20(m,3H),7.12(d,1H),6.81(s,1H),6.03(dd,1H),5.06(dd,1H),4.28-4.19(m,2H),4.05(d,2H),3.96-3.91(m,1H),3.86(s,3H),3.73-3.62(m,1H),3.27-3.19(m,1H),3.18(s,3H),2.96(t,2H),2.92-2.72(m,5H),2.65-2.53(m,3H),2.24-2.17(m,3H),2.03-1.97(m,4H),1.88-1.77(m,3H),1.70-1.60(m,2H),1.49-1.41(m,1H),1.20-1.12(m,2H),1.03-0.88(m,2H),0.60-0.52(m,1H),0.23-0.15(m,1H)。
实施例16
(±)-(Z)-N-(45-氯-26-(4-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)甲基)哌嗪-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-环丙基甲磺酰胺16
采用实施例15的合成路线,将中间体14h替换为N-环丙基-N-(2-((2,5-二氯嘧啶-4-基)氨基)-5-羟基苯基)甲磺酰胺(参考实施例14中间体14h的合成路线,将化合物14f替换为2,4,5-三氯嘧啶(上海毕得医药)制备而得),经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:62%-67%,流速:30mL/min),得到标题化合物16(外消旋体混合物,25mg,产率:27%)。
MS m/z(ESI):1033.3[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.07(s,1H),8.24(s,1H),8.13(s,1H),7.88(s,1H),7.80(d,2H),7.72(d,1H),7.65(d,1H),7.31(s,1H),7.27(s,1H),7.23(d,1H),7.12(d,1H),6.80(s,1H),6.06(d,1H),5.06(dd,1H),4.29-4.16(m,2H),4.05(d,2H),3.96-3.89(m,1H),3.86(s,3H),3.73-3.64(m,1H),3.27-3.20(m,1H),3.17(s,3H),2.97(t,2H),2.92-2.72(m,5H),2.65-2.53(m,3H),2.28-2.11(m,3H),2.05-1.90(m,4H),1.88-1.77(m,3H),1.69-1.60(m,2H),1.49-1.41(m,1H),1.20-1.10(m,2H),0.97-0.88(m,2H),0.60-0.52(m,1H),0.23-0.15(m,1H)。
实施例17
(±)-(Z)-N-(45-溴-26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)甲磺酰胺17
第一步
4-(3-氟-4-硝基苯氧基)丁酸乙酯17b
将3-氟-4-硝基苯酚17a(1.0g,6.37mmol,上海毕得医药)溶于20mL N,N-二甲基甲酰胺,加入碳酸铯(4.2g,12.89mmol)和4-溴丁酸乙酯(1.3g,6.66mmol),升温到60℃反应16小时。反应液过滤除去不溶物,滤液浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物17b(980mg,产率:57%)。
MS m/z(ESI):289.1[M+18]。
第二步
4-(3-(甲基磺酰胺基)-4-硝基苯氧基)丁酸乙酯17c
向50mL单口瓶中加入碳酸铯(2.95g,9.05mmol)和乙腈(30mL),然后加入甲磺酰胺(520mg,5.47mmol)和化合物17b(980mg,3.61mmol)。升温至80℃反应12小时。反应液过滤,滤液浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物17c(1.25g,产率:100%)。
MS m/z(ESI):364.1[M+18]。
第三步
4-(3-(N-(4-甲氧基苄基)甲基磺酰胺基)-4-硝基苯氧基)丁酸乙酯17d
将化合物17c(1.0g,2.89mmol)溶于15mL N,N-二甲基甲酰胺,加入无水碳酸钾(800mg,5.79mmol)和4-甲氧基苄氯(600mg,3.83mmol),升温至80℃反应12小时。反应液过滤,滤液浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物17d(1.34g,产率:99%)。
MS m/z(ESI):484.1[M+18]。
第四步
N-(5-(4-羟基丁氧基)-2-硝基苯基)-N-(4-甲氧基苄基)甲磺酰胺17e
将化合物17d(730mg,1.56mmol)溶于15mL四氢呋喃,降温至-30℃,氮气氛下滴加2.5M氢化铝锂的四氢呋喃溶液(0.65mL),-30℃反应1小时。向反应液中加入十水合硫酸钠淬灭反应,然后加入无水硫酸钠干燥30分钟,过滤,滤液浓缩,真空干燥,得到粗产品标题化合物17e(650mg,产率:98%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):442.1[M+18]。
第五步
N-(2-氨基-5-(4-羟基丁氧基)苯基)-N-(4-甲氧基苄基)甲磺酰胺17f
向100mL单口瓶中加入化合物17e(630mg,1.48mmol)、乙醇(15mL)、兰尼镍(100mg,1.70mmol)和水合肼(400mg,7.99mmol),升温至80℃反应3小时。待反应液冷却到室温,浓缩反应液,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物17f(480mg,产率:82%)。
MS m/z(ESI):395.1[M+1]。
第六步
N-(2-((5-溴-2-氯嘧啶-4-基)氨基)-5-(4-羟基丁氧基)苯基)-N-(4-甲氧基苄基)甲磺酰胺17g
向50mL单口瓶中加入化合物17f(280mg,1.23mmol)和5-溴-2,4-二氯嘧啶(480mg,1.22mmol),然后加入20mL异丙醇和N,N-二异丙基乙胺(790mg,6.11mmol),升温至90℃反应16小时。待反应液冷却到室温,浓缩反应液,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物17g(360mg,产率:50%)。
MS m/z(ESI):585.0[M+1]。
第七步
N-(2-((5-溴-2-((4-(4-(二甲氧基甲基)哌啶-1-基)-2-甲氧基-5-(1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)-5-((5-羟基戊基)氧基)苯基)-N-(4-甲氧基苄基)甲磺酰胺17h
向50mL单口瓶中加入化合物17g(246mg,0.42mmol)和化合物13c(600mg,1.39mmol),然后加入15mL甲醇和三氟乙酸(490mg,4.30mmol),氮气氛下升温至90℃反应16小时。待反应液冷却到室温,浓缩反应液,加入乙酸乙酯(50mL),用饱和碳酸氢钠溶液(30mL)洗涤,分离出有机相,浓缩有机相,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物17h(230mg,产率:61%)。
MS m/z(ESI):895.1[M+1]。
第八步
(Z)-N-(45-溴-26-(4-(二甲氧基甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-(4-甲氧基苄基)甲磺酰胺17i
向25mL单口瓶中加入化合物17h(220mg,0.25mmol)、甲苯(14mL)和氰基亚甲基三正丁基膦(240mg,0.99mmol),氮气氛下升温至120℃反应16小时。待反应液冷却到室温,浓缩反应液,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物17i(215mg,产率:100%)。
MS m/z(ESI):877.1[M+1]。
第九步
(Z)-N-(45-溴-26-(4-甲酰基哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)甲磺酰胺17j
向25mL单口瓶中加入化合物17i(100mg,0.11mmol)和三氟乙酸(3mL),然后滴加甲烷磺酸(50mg,0.52mmol),反应2小时。减压除去溶剂,加入乙酸乙酯(15mL),用饱和碳酸氢钠(15mL)洗涤,分出有机相,有机相用无水硫酸钠干燥,过滤,滤液浓缩,真空干燥,得到粗产品标题化合物17j(40mg,产率:49%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):711.0[M+1]。
第十步
(±)-(Z)-N-(45-溴-26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)甲磺酰胺17
将化合物13g(60mg,0.16mmol)溶于3.5mL二氯甲烷和甲醇的混合溶剂(V/V=1/1)中,加入无水乙酸钠(60mg,0.73mmol),反应5分钟。加入化合物17j(40mg,0.056mmol),反应60分钟。然后加入三乙酰氧基硼氢化钠(24mg,0.11mmol),反应30分钟。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-75%,流速:30mL/min),得到标题化合物17(外消旋体,10mg,产率:17%)。
MS m/z(ESI):1037.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.08(s,1H),9.16(s,1H),8.24(s,1H),8.16(d,2H),7.78(s,1H),7.70-7.65(m,2H),7.56(d,1H),7.34(d,1H),7.26(d,1H),7.16(s,1H),6.78-6.75(m,2H),6.17-6.15(m,1H),5.07(dd,1H),4.25-4.20(m,2H),3.84(s,3H),3.71(t,2H),3.48-3.41(m,4H),2.99-2.83(m,6H),2.65-2.45(m,8H),2.27(d,2H),2.06-1.96(m,3H),1.78-1.58(m,5H),1.33-1.25(m,2H)。
实施例18
(±)-(Z)-N-(45-溴-26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-(甲氧基-d3)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺18

第一步
1-溴-2-氟-4-(甲氧基-d3)-5-硝基苯18b
称取4-溴-5-氟-2-硝基苯酚18a(0.65g,2.75mmol,上海毕得医药)到50mL烧瓶中,加入12mL N,N-二甲基甲酰胺、无水碳酸钾(456mg,3.29mmol)和氘代碘甲烷(456mg,3.14mmol),然后升温到45℃反应12小时。待反应冷却至室温,过滤,用N,N-二甲基甲酰胺(2mL×2)洗涤滤饼,滤饼真空干燥,得到粗产品标题化合物18b(696mg,产率:100%),粗产品不经纯化直接用于下一步反应。
第二步
1-(2-溴-5-(甲氧基-d3)-4-硝基苯基)-4-(二甲氧基甲基)哌啶18c
将化合物18b(696mg,2.75mmol)的N,N-二甲基甲酰胺(16mL)溶液、4-(二甲氧基甲基)哌啶(460mg,2.89mmol)加入到50mL茄形瓶中,加入无水碳酸钾(570mg,4.12mmol),然后升温到88℃反应4小时。待反应冷却至室温,向反应液中加入水(30mL),用乙酸乙酯萃取(40mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥,过滤,滤液减压浓缩,得到标题化合物18c(1.0g,产率:92%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):392.0[M+1]。
第三步
4-(二甲氧基甲基)-1-(5-(甲氧基-d3)-4-硝基-2-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯基)哌啶18d
称取化合物18c(1.0g,2.55mmol)、1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-硼酸频哪醇酯(1.0g,3.59mmol)到100mL茄形瓶中,加入1,1′-双(二苯基膦)二茂铁二氯 化钯(180mg,0.25mmol)、无水碳酸钠(540mg,5.09mmol)、24mL 1,4-二氧六环和水的混合溶剂(V/V=5/1),氮气置换三次,升温至100℃反应12小时。待反应液冷却到室温,反应液用硅藻土过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物18d(1.0g,产率:84%)。
MS m/z(ESI):464.2[M+1]。
第四步
4-(4-(二甲氧基甲基)哌啶-1-基)-2-(甲氧基-d3)-5-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯胺18e
称取化合物18d(1.0g,2.16mmol)到100mL单口瓶中,加入30mL甲醇,然后加入10%湿钯碳(含55%质量分数的水,220mg,0.21mmol)。氢气置换三次,氢气气氛下反应12小时。反应液用硅藻土过滤,滤液浓缩,真空干燥,得到粗产品标题化合物18e(880mg,产率:94%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):434.4[M+1]。
第五步
N-(2-((5-溴-2-((4-(4-(二甲氧基甲基)哌啶-1-基)-2-(甲氧基-d3)-5-(1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)-5-羟基苯基)-N-甲基甲磺酰胺18f
称取化合物1e(400mg,0.98mmol)和化合物18e(460mg,1.06mmol)到50mL单口瓶中,加入15mL甲醇和三氟乙酸(200mg,1.75mmol),升温至90℃反应20小时。待反应冷却至室温,减压除去溶剂,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物18f(480mg,产率:67%)。
MS m/z(ESI):720.2[M+1]。
第六步
(Z)-N-(45-溴-26-(4-(二甲氧基甲基)哌啶-1-基)-24-(甲氧基-d3)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺18g
称取化合物18f(240mg,0.33mmol)、1,4-二溴丁烷(80mg,0.37mmol)到50mL茄形瓶中,加入15mL二甲基亚砜、无水碳酸钾(138mg,1.00mmol)和四丁基碘化铵(37mg,0.10mmol),升温至50℃反应16小时。待反应冷却至室温,向反应液中加入水(30mL),用乙酸乙酯萃取(40mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物18g(111mg,产率:43%)。
MS m/z(ESI):774.3[M+1]。
第七步
(Z)-N-(45-溴-26-(4-甲酰基哌啶-1-基)-24-(甲氧基-d3)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺18h
称取化合物18g(111mg,0.14mmol)到50mL茄形瓶中,加入5mL四氢呋喃,然后加入2M硫酸溶液(0.36mL),升温到45℃反应3小时。向反应液中加入饱和碳酸氢钠溶液(5mL),用乙酸乙酯萃取(25mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(20mL),无水硫酸钠干燥,过滤、减压浓缩,得到粗产品标题化合物18h(104mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):728.2[M+1]。
第八步
(±)-(Z)-N-(45-溴-26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-(甲氧基-d3)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺18
将化合物18h(100mg,0.11mmol)溶于8mL二氯甲烷和甲醇的混合溶剂中(V/V=3/1),加入无水乙酸钠(73mg,0.89mmol),反应10分钟。加入化合物13g(46mg,0.13mmol)和乙酸(20mg,0.33mmol),反应1小时。加入氰基硼氢化钠(15mg,0.23mmol),反应1小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:45%-80%,流速:30mL/min),得到标题化合物18(外消旋体,60mg,产率:52%)。
MS m/z(ESI):1054.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.09(s,1H),8.22(s,1H),8.20(s,1H),8.09(s,1H),7.90(s,1H),7.81-7.66(m,3H),7.54-7.21(m,3H),7.06(d,1H),6.76(s,1H),6.11(dd,1H),5.09(dd,1H),4.32-4.12(m,3H),3.89-3.74(m,2H),3.68-3.57(m,1H),3.51-3.35(m,3H),3.25-3.13(m,5H),3.08(s,3H),3.04-2.95(m,2H),2.94-2.84(m,1H),2.66-2.45(m,8H),2.06-1.98(m,3H),1.85-1.71(m,2H),1.69-1.58(m,2H),1.49-1.26(m,2H)。
实施例19
(±)-(Z)-N-(45-氯-26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺19
采用实施例13的合成路线,将第四步原料中间体1e替换为N-(2-((2,5-二氯嘧啶-4-基)氨基)-5-羟基苯基)-N-甲基甲磺酰胺(采用专利申请“WO2021216440中说明书第56页的实施例A2”公开的方法制备而得),经高效液相制备色谱法纯化 (Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-80%,流速:30mL/min),制得标题化合物19(外消旋体,25mg,产率:17%)。
MS m/z(ESI):1007.2[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.08(s,1H),8.21(s,1H),8.15(s,1H),8.12(s,1H),7.90(s,1H),7.74-7.66(m,3H),7.34(d,1H),7.27-7.23(m,2H),7.05(d,1H),6.77(s,1H),6.13(dd,1H),5.07(dd,1H),4.27-4.19(m,2H),3.85(s,3H),3.79(t,2H),3.51-3.41(m,4H),3.16(s,3H),3.07(s,3H),3.01-2.94(m,2H),2.93-2.83(m,1H),2.67-2.55(m,8H),2.27(d,2H),2.06-1.97(m,3H),1.78-1.71(m,2H),1.67-1.58(m3H),1.35-1.25(m,2H)。
实施例19-1
N-((Z)-(45-氯-26-(4-((4-(2-((S)-2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺19-1
采用实施例13-1的手性合成路线,将第四步原料化合物13f替换为(Z)-N-(45-氯-26-(4-甲酰基哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺(采用实施例13中合成路线,将第四步原料化合物1e替换为N-(2-((2,5-二氯嘧啶-4-基)氨基)-5-羟基苯基)-N-甲基甲磺酰胺(采用专利申请“WO2021216440中说明书第56页的实施例A2”公开的方法制备而得),经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:53%-73%,流速:30mL/min),得到标题化合物19-1(48mg,产率:41%)。
MS m/z(ESI):1007.2[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.08(s,1H),8.21(s,1H),8.15(s,1H),8.12(s,1H),7.90(s,1H),7.74-7.66(m,3H),7.34(s,1H),7.27-7.23(m,2H),7.05(d,1H),6.77(s,1H),6.13(dd,1H),5.07(dd,1H),4.27-4.19(m,2H),3.85(s,3H),3.79(t,2H),3.51-3.41(m,4H),3.16(s,3H),3.07(s,3H),3.01-2.94(m,2H),2.93-2.83(m,1H),2.67-2.45(m,8H),2.27(d,2H),2.06-1.97(m,3H),1.78-1.71(m,2H),1.67-1.58(m3H),1.37-1.25(m,2H)。
实施例20
(±)-(Z)-5-(4-((1-(45-溴-62-(二甲基磷酰基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)哌嗪-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮20
第一步
(2-((5-溴-2-氯嘧啶-4-基)氨基)-5-甲氧基苯基)二甲基氧化膦20b
将5-溴-2,4-二氯嘧啶(1.8g,7.90mmol,上海毕得医药)、(2-氨基-5-甲氧基苯基)二甲基氧化膦20a(1.58g,7.93mmol,采用专利申请“WO2021216440中说明书第60页的实施例A18”公开的方法制备而得)溶于异丙醇(40mL)中,加入N,N-二异丙基乙胺(2mL),加热回流反应23小时。待反应液冷却到室温,减压浓缩除去溶剂,然后加入饱和碳酸氢钠溶液(40mL)和乙酸乙酯(40mL),分离出有机相,水相用乙酸乙酯萃取(30mL×3),合并有机相,有机相用无水硫酸钠干燥,过滤,滤液浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物20b(2.0g,产率:65%)。
MS m/z(ESI):388.0[M-1]。
第二步
(2-((5-溴-2-氯嘧啶-4-基)氨基)-5-羟基苯基)二甲基氧化膦20c
将化合物20b(1.0g,2.56mmol)溶于50mL二氯甲烷,降温至0℃,氮气氛下滴加1M三溴化硼的二氯甲烷溶液(10.2mL,10.2mmol,上海阿达玛斯试剂有限公司),滴加完后恢复室温反应12小时。向反应液中缓慢滴加冰水淬灭反应,用饱和碳酸氢钠溶液调节反应液pH到7,用二氯甲烷萃取(50mL×2),合并有机相,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物20c(743mg,产率:77%)。
MS m/z(ESI):376.0[M+1]。
第三步
(2-((5-溴-2-((4-(4-(二甲氧基甲基)哌啶-1-基)-2-甲氧基-5-(1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)-5-羟基苯基)二甲基氧化膦20d
将化合物20c(200mg,0.53mmol)和化合物13c(241mg,0.56mmol)溶于20mL甲醇,加入三氟乙酸(126mg,1.11mmol),升温到90℃反应12小时。待反应液冷却到室温,浓缩反应液,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物20d(66mg,产率:18%)。
MS m/z(ESI):686.0[M+1]。
第四步
(Z)-(45-溴-26-(4-(二甲氧基甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)二甲基氧化膦20e
将化合物20d(66mg,0.096mmol)和1,4-二溴丁烷(32mg,0.15mmol)溶于3mL二甲基亚砜,加入无水碳酸钾(28mg,0.20mmol)和四丁基碘化铵(8mg,0.024mmol),升温到60℃反应12小时。待反应液冷却到室温,过滤,滤液浓缩,真空干燥,得到粗产品标题化合物20e(73mg,产率:100%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):738.1[M-1]。
第五步
(Z)-1-(45-溴-62-(二甲基磷酰基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-甲醛20f
将化合物20e(73mg,0.099mmol)溶于4mL四氢呋喃,加入2M稀硫酸(0.5mL),反应4小时。用饱和碳酸氢钠溶液调节反应液pH到7,用乙酸乙酯(8mL×3)萃取,合并有机相,有机相用饱和氯化钠溶液洗涤(8mL),无水硫酸钠干燥,过滤, 浓缩,真空干燥,得到粗产品标题化合物20f(70mg,产率:68%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):694.3[M+1]。
第六步
(±)-(Z)-5-(4-((1-(45-溴-62-(二甲基磷酰基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)哌嗪-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮20
将化合物13g(40mg,0.11mmol)溶于6mL二氯甲烷和甲醇的混合溶剂(V/V=1/1)中,加入无水乙酸钠(50mg,0.61mmol),反应10分钟。然后加入化合物20f(70mg,0.10mmol),反应1小时。加入氰基硼氢化钠(15mg,0.25mmol),反应30分钟。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:45%-63%,流速:30mL/min),得到标题化合物20(外消旋体,14mg,产率:14%)。
MS m/z(ESI):1020.1[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.08(s,1H),10.49(s,1H),8.26(s,1H),8.18(s,1H),8.02(dd,1H),7.89(s,1H),7.77(s,1H),7.68(d,1H),7.37(s,1H),7.34(d,1H),7.26(dd,1H),6.98(dd,1H),6.79(s,1H),6.12(dd,1H),5.07(dd,1H),4.26-4.20(m,2H),3.84(s,3H),3.80-3.74(m,2H),3.49-3.41(m,4H),3.20-3.13(m,1H),3.00(d,2H),2.92-2.83(m,1H),2.65-2.45(m,8H),2.27(d,2H),2.05-1.97(m,3H),1.78-1.70(m,7H),1.67-1.52(m,3H),1.35-1.26(m,2H)。
实施例20-1
5-(4-((1-((Z)-45-溴-62-(二甲基磷酰基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)哌嗪-1-基)-2-((S)-2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮20-1
采用实施例13-1的手性合成路线,将第四步中间体13f替换成化合物20f,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-70%,流速:30mL/min),得到标题化合物20-1(66mg,产率:50%)。
MS m/z(ESI):1020.2[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.07(s,1H),10.50(s,1H),8.26(s,1H),8.18(s,1H),8.02(dd,1H),7.89(s,1H),7.77(s,1H),7.68(d,1H),7.37(s,1H),7.34(d,1H),7.26(dd,1H),6.99(dd,1H),6.79(s,1H),6.12(dd,1H),5.07(dd,1H),4.28-4.19(m,2H),3.84(s,3H),3.77(t,2H),3.51-3.40(m,4H),3.00(d,2H),2.93-2.83(m,1H),2.65-2.52(m,5H),2.27(d,2H),2.05-1.96(m,4H),1.79-1.70(m,8H),1.70-1.53(m,4H),1.36-1.26(m,3H)。
实施例21
(±)-(Z)-5-(4-((1-(45-氯-62-(二甲基磷酰基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)哌嗪-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮21
采用实施例20的合成路线,将路线中第一步的合成原料5-溴-2,4-二氯嘧啶替换为2,4,5-三氯嘧啶,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:45%-65%,流速:30mL/min),得到标题化合物21(外消旋体,10mg,产率:10%)。
MS m/z(ESI):976.7[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.09(s,1H),10.82(s,1H),8.27(s,1H),8.17(dd,1H),8.11(s,1H),7.89(s,1H),7.81(s,1H),7.77-7.65(m,1H),7.47(s,1H),7.40-7.18(m,2H),7.01(dd,1H),6.78(s,1H),6.08(d,1H),5.08(d,1H),4.26-4.18(m,2H),3.89-3.78(m,4H),3.49-3.39(m,2H),3.20-3.12(m,4H),3.04(d,2H),2.93-2.83(m,1H),2.65-2.53(m,6H),2.32-2.22(m,1H),2.06-1.95(m,4H),1.83-1.70(m,7H),1.61-1.50(m,4H),1.35-1.26(m,3H)。
实施例22
(±)-(Z)-N-(45-溴-26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十二蕃-62-基)-N-甲基甲磺酰胺22
采用实施例13的合成路线,第五步的原料1,4-二溴丁烷替换为1,5-二溴戊烷,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:50%-80%,流速:30mL/min),得到标题化合物22(外消旋体,43mg,产率:46%)。
MS m/z(ESI):1065.1[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.08(s,1H),8.18(s,1H),8.09(s,1H),8.00(s,1H),7.92(s,1H),7.83(d,1H),7.75(s,1H),7.69(d,1H),7.35(s,2H),7.27(d,1H),7.13(d,1H),6.81(s,1H),6.23(dd,1H),5.07(dd,1H),4.08(t,2H),3.94(t,2H),3.83(s,3H),3.55-3.41(m,4H),3.20(s,3H),3.10(s,3H),3.05(d,2H),2.95-2.83(m,1H),2.70-2.50(m,7H),2.31(d,2H),2.06-1.96(m,1H),1.92-1.75(m,4H),1.74-1.61(m,1H),1.53-1.42(m,2H),1.41-1.31(m,2H),1.30-1.17(m,3H)。
实施例23
(±)-(Z)-N-(45-溴-26-(4-((4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺23
将(±)-3-(1-甲基-6-(哌嗪-1-基)-1H-吲唑-3-基)哌啶-2,6-二酮盐酸盐23a(采用专利申请“WO2023083194 A1说明书第323页的实施例106”公开的方法制备而得)(30mg,0.082mmol)溶于4mL二氯甲烷和甲醇的混合溶剂中(V/V=1/1),加 入无水乙酸钠(57mg,0.69mmol),反应10分钟。加入化合物13f(50mg,0.069mmol),反应1小时。加入三乙酰氧基硼氢化钠(44mg,0.21mmol),反应1小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:60%-72%,流速:30mL/min),得到标题化合物23(外消旋体,13mg,产率:18%)。
MS m/z(ESI):1036.4[M+1]。
1H NMR(500MHz,DMSO-d6):δ10.85(s,1H),8.23(s,1H),8.19(s,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.73(d,1H),7.50(d,1H),7.25(s,1H),7.06(d,1H),6.92(dd,1H),6.84(d,1H),6.78(s,1H),6.11(dd,1H),4.28-4.20(m,3H),3.89(s,3H),3.85(s,3H),3.79(t,2H),3.26-3.20(m,4H),3.17(s,3H),3.07(s,3H),2.99(d,2H),2.65-2.52(m,8H),2.33-2.25(m,3H),2.19-2.12(m,1H),2.06-1.97(m,2H),1.75(d,2H),1.69-1.59(m,3H),1.35-1.26(m,2H)。
实施例23-1和23-2
N-((Z)-45-溴-26-(4-((4-(3-((S)-2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺23-1
N-((Z)-45-溴-26-(4-((4-(3-((R)-2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺23-2

化合物23(120mg)经手性拆分(分离条件:Gilson-281,色谱柱:CHIRALPAK IE(20×250mM),流动相:乙腈/乙醇=40/60(v/v),流速20mL/min)得到标题化合物(50mg,50mg)。
单一构型化合物(50mg)(较短保留时间,也记为23-1A:23-1和23-2中具有较短保留时间的化合物):
MS m/z(ESI):1036.2[M+1]。
1H NMR(500MHz,DMSO-d6):δ10.85(s,1H),8.23(s,1H),8.19(s,1H),8.07(s,1H),7.89(s,1H),7.75(s,1H),7.73(d,1H),7.50(d,1H),7.25(s,1H),7.06(d,1H),6.92(dd,1H),6.84(d,1H),6.78(s,1H),6.11(dd,1H),4.28-4.20(m,3H),3.89(s,3H),3.85(s,3H),3.79(t,2H),3.26-3.20(m,4H),3.17(s,3H),3.07(s,3H),2.99(d,2H),2.65-2.52(m,8H),2.33-2.25(m,3H),2.19-2.12(m,1H),2.06-1.97(m,2H),1.75(d,2H),1.69-1.59(m,3H),1.35-1.26(m,2H)。
手性HPLC分析方法:保留时间6.93分钟(Agilent1260 DAD,色谱柱:CHIRALPAK IE(4.6×150mM),5μm;流动相:乙腈/乙醇(含0.1%二乙胺)=40/60(v/v),流速1.0mL/min)。
单一构型化合物(50mg)(较长保留时间,也记为23-2A:23-1和23-2中具有较长保留时间的化合物):
MS m/z(ESI):1036.3[M+1]。
1H NMR(500MHz,DMSO-d6):δ10.85(s,1H),8.23(s,1H),8.19(s,1H),8.07(s,1H),7.89(s,1H),7.75(s,1H),7.73(d,1H),7.50(d,1H),7.25(s,1H),7.06(d,1H),6.92(dd,1H),6.84(d,1H),6.78(s,1H),6.11(dd,1H),4.28-4.20(m,3H),3.89(s,3H),3.85(s,3H),3.79(t,2H),3.26-3.20(m,4H),3.17(s,3H),3.07(s,3H),2.99(d,2H),2.65-2.52(m,8H),2.33-2.25(m,3H),2.19-2.12(m,1H),2.06-1.97(m,2H),1.75(d,2H),1.69-1.59(m,3H),1.35-1.26(m,2H)。
手性HPLC分析方法:保留时间11.37分钟(Agilent1260 DAD,色谱柱:CHIRALPAK IE(4.6×150mM),5μm;流动相:乙腈/乙醇(含0.1%二乙胺)=40/60(v/v),流速1.0mL/min)。
实施例24
(±)-(Z)-N-(45-溴-26-(4-((4-(3-(2,6-二氧代哌啶-3-基)-5-氟-1-甲基-1H-吲唑-6-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺24
第一步
3-(2,6-双(苄氧基)吡啶-3-基)-6-溴-5-氟-1-甲基-1H-吲唑24c
称取6-溴-5-氟-3-碘-1-甲基-1H-引唑24a(3.7g,10.42mmol,采用专利申请“WO2016057834 A1说明书第443页的实施例386”公开的方法制备而得)、2,6-双(苄氧基)吡啶-3-硼酸频哪醇酯24b(4.79g,11.47mmol,上海皓鸿生物医药有限公司)到100mL茄形瓶中,依次加入1,1′-双(二苯基膦)二茂铁二氯化钯二氯甲烷络合物(845mg,1.04mmol,安耐吉化学)、碳酸钠(2.76g,26.06mmol)、1,4-二氧六环和水的混合溶剂72mL(V/V=5/1),升温至90℃反应12小时。待反应液冷却到室温,将反应液倒入水(100mL)中,用乙酸乙酯萃取(100mL×3),合并有机相,有机相用 饱和氯化钠溶液(100mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物24c(4.1g,产率:76%)。
MS m/z(ESI):518.0[M+1]。
第二步
4-(3-(2,6-双(苄氧基)吡啶-3-基)-5-氟-1-甲基-1H-吲唑-6-基)哌嗪-1-羧酸叔丁酯24d
称取化合物24c(2.5g,4.82mmol)、哌嗪-1-羧酸叔丁酯(1.17g,6.27mmol)到100mL茄形瓶中,依次加入三(二亚苄基丙酮)二钯(442mg,0.48mmol)、1,1'-联萘-2,2'-双二苯膦(601mg,0.96mmol)、叔丁醇钠(927mg,9.65mmol)、甲苯(50mL),升温至100℃反应12小时。待反应液冷却到室温,将反应液倒入水(100mL)中,用乙酸乙酯萃取(100mL×3),合并有机相,有机相用饱和氯化钠溶液(100mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物24d(2.78g,产率:92%)。
MS m/z(ESI):624.3[M+1]。
第三步
(±)-4-(3-(2,6-二氧代哌啶-3-基)-5-氟-1-甲基-1H-吲唑-6-基)哌嗪-1-羧酸叔丁酯24e
化合物24d(500mg,0.80mmol)溶于25mL四氢呋喃和甲醇的混合溶剂(V/V=4/1)中,加入10%湿钯碳(含55%质量分数水,500mg,4.71mmol)和20%氢氧化钯碳(含50%质量分数水,500mg,3.59mmol,韶远(科技)上海有限公司)。氢气置换三次,氢气气氛下反应6小时。反应液经硅藻土过滤,滤液浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系A纯化,得到标题化合物24e(100mg,产率:28%)。
MS m/z(ESI):446.2[M+1]。
第四步
(±)-3-(5-氟-1-甲基-6-(哌嗪-1-基)-1H-吲唑-3-基)哌啶-2,6二酮盐酸盐24f
将化合物24e(300mg,0.67mmol)溶于二氯甲烷(6mL),加入4M氯化氢的1,4-二氧六环溶液(6mL),反应2小时。减压除去溶剂,真空干燥,得到粗产品标题化合物24f(252mg,99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):346.1[M+1]。
第五步
(±)-(Z)-N-(45-溴-26-(4-((4-(3-(2,6-二氧代哌啶-3-基)-5-氟-1-甲基-1H-吲唑-6-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺24
将化合物24f(120mg,0.31mmol)溶于10mL二氯甲烷和甲醇的混合溶剂(V/V=1/1)中,加入无水乙酸钠(113mg,1.38mmol),反应10分钟。加入化合物 13f(200mg,0.28mmol),反应1小时。加入三乙酰氧基硼氢化钠(180mg,0.85mmol),反应1小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:60%-70%,流速:30mL/min),得到标题化合物24(外消旋体,6mg,产率:10%)。
MS m/z(ESI):1054.5[M+1]。
1H NMR(500MHz,DMSO-d6):δ10.86(s,1H),8.23(s,1H),8.19(d,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.72(d,1H),7.46(d,1H),7.25(s,1H),7.11(d,1H),7.06(d,1H),6.78(s,1H),6.11(dd,1H),4.31-4.19(m,3H),3.94(s,3H),3.85(s,3H),3.81-3.75(m,2H),3.17(s,3H),3.13-3.04(m,7H),2.99(d,2H),2.69-2.53(m,8H),2.39-2.26(m,3H),2.18-2.10(m,1H),2.08-1.96(m,2H),1.75(d,2H),1.69-1.59(m,3H),1.36-1.26(m,2H)。
实施例24-1和24-2
N-((Z)-45-溴-26-(4-((4-(3-((S)-2,6-二氧代哌啶-3-基)-5-氟-1-甲基-1H-吲唑-6-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺24-1
N-((Z)-45-溴-26-(4-((4-(3-((R)-2,6-二氧代哌啶-3-基)-5-氟-1-甲基-1H-吲唑-6-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺24-2

将化合物24(30mg)经手性拆分(分离条件:Gilson-281,色谱柱:CHIRALPAK IE(20×250mM),流动相:乙腈/乙醇=40/60(v/v),流速20mL/min),得到标题化合物(10mg,10mg)。
单一构型化合物(10mg)(较短保留时间):
MS m/z(ESI):1054.6[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.86(s,1H),8.23(s,1H),8.19(d,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.72(d,1H),7.46(d,1H),7.25(s,1H),7.11(d,1H),7.06(d,1H),6.78(s,1H),6.11(dd,1H),4.31-4.19(m,3H),3.94(s,3H),3.85(s,3H),3.81-3.75(m,2H),3.17(s,3H),3.13-3.04(m,7H),2.99(d,2H),2.69-2.53(m,8H),2.39-2.26(m,3H),2.18-2.10(m,1H),2.08-1.96(m,2H),1.75(d,2H),1.69-1.59(m,3H),1.36-1.26(m,2H)。
手性HPLC分析方法:保留时间5.82分钟(Agilent1260 DAD,色谱柱:CHIRALPAK IE(4.6×150mM),5μm;流动相:乙腈/乙醇(含0.1%二乙胺)=40/60(v/v),流速1.0mL/min)。
单一构型化合物(10mg)(较长保留时间):
MS m/z(ESI):1054.5[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.86(s,1H),8.23(s,1H),8.19(d,1H),8.07(s,1H),7.89(s,1H),7.75(s,1H),7.72(d,1H),7.46(d,1H),7.25(s,1H),7.11(d,1H),7.06(d,1H),6.78(s,1H),6.11(dd,1H),4.31-4.19(m,3H),3.94(s,3H),3.85(s,3H),3.81-3.75(m,2H),3.17(s,3H),3.13-3.04(m,7H),2.99(d,2H),2.69-2.53(m,8H),2.39-2.26(m,3H),2.18-2.10(m,1H),2.08-1.96(m,2H),1.75(d,2H),1.69-1.59(m,3H),1.36-1.26(m,2H)。
手性HPLC分析方法:保留时间7.39分钟(Agilent1260 DAD,色谱柱:CHIRALPAK IE(4.6×150mM),5μm;流动相:乙腈/乙醇(含0.1%二乙胺)=40/60(v/v),流速1.0mL/min)。
实施例25
(±)-(Z)-N-(45-溴-26-(4-((4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌啶-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺25
采用实施例13的方法,将第七步中的化合物13g替换为(±)-3-(1-甲基-6-(哌啶-4-基)-1H-吲唑-3-基)哌啶-2,6-二酮盐酸盐(采用公知的方法“WO2023083194A1中说明书第211页中间体B47”制备得到)。经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:70%-90%,流速:30mL/min),制得标题化合物25(外消旋体,17mg,产率:30%)。
MS m/z(ESI):1035.4[M+1]。
1H NMR(500MHz,DMSO-d6):δ10.87(s,1H),8.23(s,1H),8.19(s,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.72(d,1H),7.60(d,1H),7.44(s,1H),7.25(s,1H),7.06(d,1H),7.04(d,1H),6.77(s,1H),6.11(dd,1H),4.32(dd,1H),4.26-4.20(m,2H),3.96(s,3H),3.84(s,3H),3.79(t,2H),3.17(s,3H),3.07(s,3H),2.99(d,4H),2.67-2.53(m,5H),2.37-2.31(m,1H),2.26(d,2H),2.19-2.12(m,1H),2.07-1.95(m,4H),1.83-1.71(m,6H),1.67-1.57(m,3H),1.33-1.26(m,2H)。
实施例26
(Z)-N-(45-溴-26-(4-((4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺26
采用实施例13的方法,将第七步中的化合物13g替换为1-(1-甲基-6-(哌嗪-1-基)-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮盐酸盐(采用公知的方法 “WO2023083194 A1中说明书第310页实施例89”制备得到)。经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-75%,流速:30mL/min),制得标题化合物26(12mg,产率:36%)。
MS m/z(ESI):1037.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.50(s,1H),8.23(s,1H),8.19(d,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.73(d,1H),7.45(d,1H),7.25(s,1H),7.06(d,1H),6.92(d,1H),6.82(s,1H),6.78(s,1H),6.11(dd,1H),4.27-4.21(m,2H),3.91-3.89(m,1H),3.88(s,3H),3.85(s,3H),3.79(t,2H),3.27-3.21(m,4H),3.17(s,3H),3.07(s,3H),2.99(d,2H),2.73(t,2H),2.59-2.53(m,5H),2.29(d,2H),2.08-1.96(m,4H),1.80-1.71(m,2H),1.68-1.59(m,3H),1.36-1.26(m,2H)。
实施例27
(Z)-N-(45-溴-26-(4-((4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)哌啶-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺27
采用实施例13的方法,将第七步中的化合物13g替换为1-(1-甲基-6-(哌啶-4-基)-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮盐酸盐(采用公知的方法“WO2023083194 A1中说明书第210页中间体B46”制备得到)。经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-75%,流速:30mL/min),制得标题化合物27(17mg,产率:34%)。
MS m/z(ESI):1036.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.53(s,1H),8.23(s,1H),8.19(s,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.73(d,1H),7.54(d,1H),7.45(s,1H),7.26(s,1H),7.06(d,1H),7.04(d,1H),6.78(s,1H),6.11(dd,1H),4.26-4.21(m,2H),3.96(s,3H),3.90(t,2H),3.85(s,3H),3.79(t,2H),3.17(s,3H),3.07(s,3H),2.99(d,4H),2.75(t,2H),2.65-2.62(m,2H),2.37-2.35(m,1H),2.26(d,2H),2.07-1.97(m,5H),1.84-1.72(m,6H),1.67-1.59(m,2H),1.33-1.27(m,2H)。
实施例28
(±)-(Z)-N-(45-溴-26-(4-(4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺28
采用实施例13的方法,将第七步中的化合物13g替换为(±)-1-(6-(3,3-二氟哌啶-4-基)-1-甲基-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮盐酸盐(采用专利申请“WO2022235945 A1中说明书第180页”公开的方法制备而得)。经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:60%-95%,流速:30mL/min),制得标题化合物28(外消旋体,16mg,产率:27%)。
MS m/z(ESI):1072.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.56(s,1H),8.23(s,1H),8.19(s,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.73(d,1H),7.59(d,1H),7.56(s,1H),7.26(s,1H),7.09(d,1H),7.06(d,1H),6.78(s,1H),6.11(dd,1H),4.28-4.20(m,2H),3.99(s,3H),3.92(t,2H),3.85(s,3H),3.79(t,2H),3.26-3.20(m,1H),3.17(s,3H),3.07(s,3H),3.05-2.95(m,3H),2.76(t,2H),2.61-2.52(m,3H),2.45-2.33(m,3H),2.24(t,1H),2.08-1.95(m,3H),1.89-1.83(m,1H),1.75(t,2H),1.68-1.59(m,3H),1.36-1.27(m,2H)。
实施例29
(±)-(Z)-N-(45-溴-26-(4-((4-(1-(2,6-二氧代哌啶-3-基)-3-甲基-2-氧代-2,3-二氢-1H-苯并[d]咪唑-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺29
采用实施例13的方法,将第七步中的化合物13g替换为(±)-3-(3-甲基-2-氧代-5-(哌嗪-1-基)-2,3-二氢-1H-苯并[d]咪唑-1-基)哌啶-2,6-二酮盐酸盐(采用专利申请“WO2019133531A1中说明书第1150页中间体PN”公开的方法制备而得)。经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:50%-80%,流速:30mL/min),制得标题化合物29(外消旋体,27mg,产率:33%)。
MS m/z(ESI):1052.3[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.06(s,1H),8.22(s,1H),8.19(s,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.72(d,1H),7.25(s,1H),7.06(d,1H),6.94(d,1H),6.84(d,1H),6.77(s,1H),6.63(dd,1H),6.11(dd,1H),5.29(dd,1H),4.26-4.20(m,2H),3.85(s,3H),3.79(t,2H),3.30(s,3H),3.17(s,3H),3.12-3.08(m,3H),3.07(s,3H),2.98(d,2H),2.89(ddd,1H),2.73-2.52(m,8H),2.28(d,2H),2.06-1.95(m,4H),1.74(d,2H),1.68-1.58(m,3H),1.35-1.27(m,2H)。
实施例30
(±)-(Z)-N-(45-溴-26-(4-((4-(1-(2,6-二氧代哌啶-3-基)-3-甲基-2-氧代-2,3-二氢-1H-苯并[d]咪唑-5-基)哌啶-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺30
采用实施例13的方法,将第七步中的化合物13g替换为(±)-3-(3-甲基-2-氧代-5-(哌啶-4-基)-2,3-二氢-1H-苯并[d]咪唑-1-基)哌啶-2,6-二酮盐酸盐(采用专利申请“WO2023283610-A1中说明书第119页的”公开的方法制备而得)。经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:50%-70%,流速:30mL/min),制得标题化合物30(外消旋体,18mg,产率:25%)。
MS m/z(ESI):1051.5[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.08(s,1H),8.23(s,1H),8.19(s,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.73(d,1H),7.25(s,1H),7.10(s,1H),7.06(d,1H),7.00(d,1H),6.91(d,1H),6.77(s,1H),6.11(dd,1H),5.33(dd,1H),4.27-4.19(m,2H),3.84(s,3H),3.79(t,2H),3.17(s,3H),3.07(s,3H),2.98(t,4H),2.94-2.84(m,2H), 2.75-2.52(m,6H),2.25(d,2H),2.08-1.95(m,6H),1.78-1.69(m,6H),1.67-1.56(m,3H),1.34-1.26(m,2H)。
实施例31
(±)-(Z)-1-(6-(1-((1-(45-溴-62-(二甲基磷酰基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)-3,3-二氟哌啶-4-基)-1-甲基-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮31
第一步
(±)-1-(6-(3,3-二氟哌啶-4-基)-1-甲基-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮盐酸盐31b
将(±)-4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶-1-羧酸叔丁酯31a(100mg,0.22mmol,采用专利申请“WO2022235945A1中说明书第180页”公开的方法制备而得)溶于4mL二氯甲烷,加入4M氯化氢的1,4-二氧六环溶液(2mL),反应1小时。减压除去溶剂,真空干燥,得到粗产品标题化合物31b(外消旋体,86mg,99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):364.4[M+1]。
第二步
(±)-(Z)-1-(6-(1-((1-(45-溴-62-(二甲基磷酰基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)-3,3-二氟哌啶-4-基)-1-甲基-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮31
将化合物31b(41mg,0.10mmol)溶于6mL二氯甲烷和甲醇的混合溶剂中(V/V=3/1),加入无水乙酸钠(67mg,0.82mmol),反应10分钟。加入化合物20f(70mg,0.10mmol)和乙酸(19mg,0.32mmol),反应1小时。加入氰基硼氢化钠(13mg,0.22mmol),反应1小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545, 洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:50%-80%,流速:30mL/min),得到标题化合物31(外消旋体,47mg,产率:45%)。
MS m/z(ESI):1041.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.56(s,1H),10.50(s,1H),8.27(s,1H),8.18(s,1H),8.03(dd,1H),7.90(s,1H),7.78(s,1H),7.59(d,1H),7.56(s,1H),7.38(s,1H),7.09(d,1H),6.99(dd,1H),6.79(s,1H),6.12(dd,1H),4.28-4.20(m,2H),3.99(s,3H),3.92(t,2H),3.84(s,3H),3.78(t,2H),3.27-3.15(m,2H),3.02(d,3H),2.76(t,2H),2.64-2.52(m,2H),2.45-2.33(m,3H),2.30-2.18(m,2H),2.05-1.97(m,2H),1.90-1.83(m,1H),1.81-1.76(m,2H),1.74(d,6H),1.69-1.54(m,3H),1.38-1.27(m,2H)。
实施例31-1和31-2
1-(6-((4S)-1-((1-((Z)-45-溴-62-(二甲基磷酰基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)-3,3-二氟哌啶-4-基)-1-甲基-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮31-1
1-(6-((4R)-1-((1-((Z)-45-溴-62-(二甲基磷酰基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)-3,3-二氟哌啶-4-基)-1-甲基-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮31-2

第一步
(S)-4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶基-1-羧酸叔丁酯31a-1
(R)-4-(3-(2,4-二氧代四氢嘧啶-1(2H)-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶基-1-羧酸叔丁酯31a-2
将化合物31a(1.0g)经手性拆分(分离条件:Gilson-281 Prep system,色谱柱:CHIRALPAK IG(20×250mm),流动相:正己烷/乙醇=40/60(v/v),流速20mL/min)得到标题化合物31a-2(400mg)和31a-1(450mg)。
标题化合物31a-2:(400mg)单一构型化合物(较短保留时间):
MS m/z(ESI):464.2[M+1]。
手性HPLC分析方法:保留时间14.32分钟(Agilent1260 DAD,色谱柱:CHIRALPAK IG 150×4.6mm,5μm;流动相:正己烷/乙醇(含0.1%二乙胺)=40/60(v/v),流速1mL/min);
标题化合物31a-1:(450mg)单一构型化合物(较长保留时间):
MS m/z(ESI):464.2[M+1]。
手性HPLC分析方法:保留时间20.11分钟(Agilent1260 DAD,色谱柱:CHIRALPAK IG 150×4.6mm,5μm;流动相:正己烷/乙醇(含0.1%二乙胺)=40/60(v/v),流速1mL/min)。
第二步
1-(6-((4S)-1-((1-((Z)-45-溴-62-(二甲基磷酰基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)-3,3-二氟哌啶-4-基)-1-甲基-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮31-1
1-(6-((4R)-1-((1-((Z)-45-溴-62-(二甲基磷酰基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)-3,3-二氟哌啶-4-基)-1-甲基-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮31-2
按照化合物31的合成路线,将化合物31a替换为化合物31a-2(100mg),得到标题化合物31-2(61mg,产率:47%)。
MS m/z(ESI):1041.2[M+1]。
1H NMR(500MHz,DMSO-d6):δ10.56(s,1H),10.50(s,1H),8.27(s,1H),8.18(s,1H),8.03(dd,1H),7.90(s,1H),7.78(s,1H),7.59(d,1H),7.56(s,1H),7.38(s,1H),7.09(d,1H),6.99(dd,1H),6.79(s,1H),6.12(dd,1H),4.28-4.20(m,2H),3.99(s,3H),3.92(t,2H),3.84(s,3H),3.78(t,2H),3.27-3.15(m,2H),3.02(d,3H),2.76(t,2H),2.64-2.52(m,2H),2.45-2.33(m,3H),2.30-2.18(m,2H),2.05-1.97(m,2H),1.90-1.83(m,1H),1.81-1.76(m,2H),1.74(d,6H),1.69-1.54(m,3H),1.38-1.27(m,2H)。
按照化合物31的合成路线,将31a替换为拆分后的化合物31a-1(100mg),得到标题化合物31-1(52mg,产率:41%)。
MS m/z(ESI):1041.2[M+1]。
1H NMR(500MHz,DMSO-d6):δ10.55(s,1H),10.49(s,1H),8.26(s,1H),8.18(s,1H),8.03(dd,1H),7.89(s,1H),7.77(s,1H),7.59(d,1H),7.56(s,1H),7.38(s,1H),7.09(d,1H),6.99(dd,1H),6.79(s,1H),6.12(dd,1H),4.28-4.20(m,2H),3.99(s,3H),3.92(t,2H),3.84(s,3H),3.78(t,2H),3.27-3.15(m,2H),3.02(d,3H),2.76(t,2H),2.64-2.52(m,2H),2.45-2.33(m,3H),2.30-2.18(m,2H),2.05-1.97(m,2H),1.90-1.83(m,1H),1.81-1.76(m,2H),1.74(d,6H),1.69-1.54(m,3H),1.38-1.27(m,2H)。
实施例32
N-((Z)-45-溴-26-(4-(((3R)-4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)-3-甲基哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺32

第一步
(R)-4-(3-(2,6-双(苄氧基)吡啶-3-基)-1-甲基-1H-吲唑-6-基)-3-甲基哌嗪-1-羧酸叔丁酯32b
称取3-(2,6-双(苄氧基)吡啶-3-基)-6-溴-1-甲基-1H-吲唑32a(300mg,0.60mmol,采用专利申请“WO2021127586A1中说明书第270页中间体9”公开的方法制备而得)、(R)-3-甲基哌嗪-1-羧酸叔丁酯(180mg,0.90mmol,韶远科技(上海)有限公司)到50mL茄形瓶中,依次加入甲磺酸(2-二环己基膦基-2’,6’-二异丙氧基-1,1’-联苯基)(2-氨基-1,1’-联苯-2-基)钯(II)(188mg,0.26mmol,阿达玛斯试剂)、2-二环己基膦-2’,6’-二异丙氧基-1,1’-联苯(56mg,0.12mmol,阿达玛斯试剂)、无水碳酸钾(166mg,1.20mmol)、1,4-二氧六环(10mL),氮气置换三次,升温至100℃反应12小时。待反应液冷却到室温,反应液用硅藻土过滤,滤液减压浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系B纯化,得到标题化合物32b(130mg,产率:35%)。
MS m/z(ESI):620.4[M+1]。
第二步
(3R)-4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)-3-甲基哌嗪-1-羧酸叔丁酯32c
将化合物32b(130mg,0.21mmol)溶于6mL四氢呋喃和甲醇的混合溶剂(V/V=1/1)中,加入10%湿钯碳(含55%质量分数水,22mg,0.21mmol)和20%氢氧化钯碳(含50%质量分数水,30mg,0.21mmol)。氢气置换三次,氢气气氛下升温至60℃反应20小时。反应液经硅藻土过滤,滤液浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物32c(一对非对映异构体混合物,比例1:1,60mg,产率:65%)。
MS m/z(ESI):442.5[M+1]。
第三步
3-(1-甲基-6-((R)-2-甲基哌嗪-1-基)-1H-吲唑-3-基)哌啶-2,6-二酮盐酸盐32d
将化合物32c(60mg,0.14mmol)溶于二氯甲烷(2mL),加入4M氯化氢的1,4-二氧六环溶液(1mL),反应1小时。减压除去溶剂,真空干燥,得到粗产品标题化合物32d(一对非对映异构体混合物,比例1:1,30mg,58%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):342.3[M+1]。
第四步
N-((Z)-45-溴-26-(4-(((3R)-4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)-3-甲基哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺32
将化合物32d(25mg,0.066mmol)溶于4mL二氯甲烷和甲醇的混合溶剂中(V/V=3/1),加入无水乙酸钠(22.6mg,0.28mmol),反应10分钟。加入化合物13f(40mg,0.055mmol),反应1小时。加入三乙酰氧基硼氢化钠(35mg,0.17mmol),反应1小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:45%-85%,流速:30mL/min),得到标题化合物32(一对非对映异构体混合物,比例1:1,10mg,产率:17%)。
MS m/z(ESI):1050.5[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.85(s,1H),8.24(s,1H),8.19(s,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.73(d,1H),7.49(d,1H),7.26(s,1H),7.06(d,1H),6.90(d,1H),6.78(s,2H),6.11(dd,1H),4.28-4.20(m,3H),4.16-4.09(m,1H),3.88(s,3H),3.85(s,3H),3.82-3.75(m,2H),3.17(s,3H),3.11-3.04(m,4H),3.02-2.95(m,2H),2.88(d,1H),2.73(d,1H),2.65-2.52(m,4H),2.35-2.25(m,3H),2.23-2.12(m,3H),2.09-1.95(m,3H),1.77(dd,2H),1.68-1.58(m,3H),1.37-1.28(m,2H),1.04(d,3H)。
实施例33
(±)-(Z)-N-(45-溴-26-(4-((4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)-4,7-二氮杂螺[2.5]辛-7-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺33

第一步
4-(3-(2,6-双(苄氧基)吡啶-3-基)-1-甲基-1H-吲唑-6-基)-4,7-二氮杂螺[2.5]辛烷-7-羧酸叔丁酯33a
称取化合物32a(300mg,0.60mmol)、4,7-二氮杂螺[2.5]辛烷-7-羧酸叔丁酯(191mg,0.90mmol,上海毕得医药)到微波管中,依次加入二(三叔丁基膦)钯(31mg,0.06mmol,韶远科技(上海)有限公司)、叔丁醇钠(58mg,0.60mmol)、二甲基亚砜(3mL)。氮气鼓吹2分钟,微波升温至120℃反应1小时。待反应冷却到室温,加水,用乙酸乙酯萃取(10mL×3),合并有机相,减压浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物33a(206mg,产率:56%)。
MS m/z(ESI):632.8[M+1]。
第二步
(±)-4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)-4,7-二氮杂螺[2.5]辛烷-7-羧酸叔丁酯33b
将化合物33a(206mg,0.33mmol)溶于6mL四氢呋喃和甲醇的混合溶剂(V/V=1/1)中,加入10%湿钯碳(含55%质量分数水,35mg,0.33mmol)和20%氢氧化钯碳(含50%质量分数水,46mg,0.33mmol)。氢气置换三次,氢气气氛下升温至60℃反应20小时。反应液经硅藻土过滤,滤液浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物33b(100mg,产率:68%)。
MS m/z(ESI):454.3[M+1]。
第三步
(±)-3-(1-甲基-6-(4,7-二氮杂螺[2.5]辛-4-基)-1H-吲唑-3-基)哌啶-2,6-二酮盐酸盐33c
将化合物33b(100mg,0.22mmol)溶于2mL二氯甲烷,加入4M氯化氢的1,4-二氧六环溶液(1mL),反应1小时。减压除去溶剂,真空干燥,得到粗产品标题化合物33c(70mg,81%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):354.2[M+1]。
第四步
(±)-(Z)-N-(45-溴-26-(4-((4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)-4,7-二氮杂螺[2.5]辛-7-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺33
将化合物33c(32mg,0.083mmol)溶于6mL二氯甲烷和甲醇的混合溶剂中(V/V=1/1),加入无水乙酸钠(45.2mg,0.55mmol),反应10分钟。加入化合物13f(40mg,0.055mmol),反应1小时。加入三乙酰氧基硼氢化钠(35mg,0.17mmol),反应1小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:60%-70%,流速:30mL/min),得到标题化合物33(外消旋体,6mg,产率:10%)。
MS m/z(ESI):1062.3[M+1]。
1H NMR(500MHz,DMSO-d6):δ10.84(s,1H),8.21(d,1H),8.18(s,1H),8.07(s,1H),7.88(d,1H),7.76-7.69(m,2H),7.48(dd,1H),7.25(d,1H),7.05(s,1H),6.96-6.88(m,2H),6.76(d,1H),6.10(d,1H),4.28-4.19(m,3H),3.87(d,3H),3.84(d,3H),3.81-3.74(m,2H),3.25-3.20(m,2H),3.17(s,3H),3.07(s,3H),3.01-2.92(m,2H),2.66-2.53(m,6H),2.33-2.25(m,2H),2.20-2.11(m,2H),2.06-1.94(m,5H),1.79-1.53(m,4H),1.49-1.41(m,2H),1.33-1.26(m,2H),0.85(t,2H)。
实施例34
N-((Z)-45-溴-26-(4-(((1R,4R)-5-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)-2,5-二氮杂双环[2.2.1]庚-2-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺34
采用实施例33的方法,将第一步中的4,7-二氮杂螺[2.5]辛烷-7-羧酸叔丁酯替换为(1R,4R)-2,5-二氮杂双环[2.2.1]庚烷-2-羧酸叔丁酯(上海毕得医药)。经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:30%-85%,流速:30mL/min),制得标题化合物34(一对非对映体混合物,比例1:1,23mg,产率:32%)。
MS m/z(ESI):1048.2[M+1]。
1H NMR(500MHz,DMSO-d6):δ10.84(s,1H),8.18(d,2H),8.06(s,1H),7.86(s,1H),7.73(s,1H),7.70(d,1H),7.44(d,1H),7.22(s,1H),7.04(d,1H),6.73(s,1H),6.57(dd,1H),6.45(d,1H),6.08(dd,1H),4.41(s,1H),4.22(dd,1H),4.20-4.15(m,2H),3.84(s,3H),3.82(s,3H),3.76(t,2H),3.53(s,1H),3.41(d,2H),3.25(dd,1H),3.16(s,3H),3.06(s,3H),2.98-2.88(m,4H),2.65-2.59(m,2H),2.48-2.42(m,2H),2.40(d,2H),2.32-2.24(m,1H),2.19-2.11(m,1H),2.04-1.94(m,2H),1.89-1.79(m,2H),1.75-1.56(m,4H),1.40-1.32(m,2H)。
实施例35
N-((Z)-45-溴-26-(4-(((1S,4S)-5-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6基)-2,5-二氮杂双环[2.2.1]庚-2-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-yl)-N-甲基甲磺酰胺35
采用实施例33的方法,将第一步4,7-二氮杂螺[2.5]辛烷-7-羧酸叔丁酯替换为(1S,4S)-2,5-二氮杂双环[2.2.1]庚烷-2-羧酸叔丁酯(上海毕得医药)。经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:52%-72%,流速:30mL/min),制得标题化合物35(一对非对映体混合物,比例1:1,8mg,产率:28%)。
MS m/z(ESI):1048.2[M+1]。
1H NMR(500MHz,DMSO-d6):δ10.84(s,1H),8.18(s,2H),8.07(s,1H),7.87(s,1H),7.78-7.68(m,2H),7.46(s,1H),7.23(s,1H),7.04(d,1H),6.73(s,1H),6.68-6.55(m,1H),6.46(s,1H),6.08(d,1H),4.52-4.38(m,1H),4.29-4.13(m,3H),3.90-3.84(m,3H),3.83(s,3H),3.79-3.73(m,2H),3.63-3.49(m,4H),3.16(s,3H),3.07(s,3H),3.01-2.87(m,3H),2.66-2.58(m,3H),2.43-2.35(m,1H),2.33-2.24(m,1H),2.20-2.12(m,1H),2.06-1.95(m,4H),1.92-1.70(m,4H),1.64-1.56(m,2H),1.50-1.42(m,1H),1.41-1.32(m,2H)。
实施例36
(±)-(Z)-N-(45-溴-26-(4-((4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)-1,4-二氮杂环庚-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺36
采用实施例32的方法,将第一步中的(R)-3-甲基哌嗪-1-羧酸叔丁酯替换为1,4-二氮杂环庚烷-1-羧酸叔丁酯(上海毕得医药)。经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:60%-80%,流速:30mL/min),制得标题化合物36(外消旋体,26mg,产率:29%)。
MS m/z(ESI):1050.5[M+1]。
1H NMR(500MHz,DMSO-d6):δ10.83(s,1H),8.20(d,2H),8.07(s,1H),7.88(s,1H),7.74(s,1H),7.71(d,1H),7.43(d,1H),7.24(s,1H),7.05(d,1H),6.75(s,1H),6.71(d,1H),6.52(s,1H),6.09(dd,1H),4.25-4.16(m,3H),3.84(s,6H),3.80-3.73(m,2H),3.63-3.53(m,5H),3.16(s,3H),3.07(s,3H),2.94(d,2H),2.80-2.73(m,2H),2.62-2.52(m,6H),2.37(d,2H),2.31-2.22(m,1H),2.18-2.10(m,1H),2.05-1.97(m,2H),1.94-1.86(m,2H),1.72-1.65(m,2H),1.63-1.58(m,2H),1.54-1.42(m,2H)。
实施例37
(±)-(Z)-5-(4-((1-(45-溴-62-(二甲基磷酰基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十二蕃-26-基)哌啶-4-基)甲基)哌嗪-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮37
采用实施例20的方法,将第四步中的1,4-二溴丁烷替换为1,5-二溴戊烷(上海毕得医药)。经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-70%,流速:30mL/min),制得标题化合物37(外消旋体,40mg,产率:20%)。
MS m/z(ESI):1034.4[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.07(s,1H),10.80(s,1H),8.21(dd,1H),8.16(s,1H),7.97(s,1H),7.94(s,1H),7.74(s,1H),7.68(d,1H),7.52(s,1H),7.35(d,1H),7.26(dd,1H),7.01(dd,1H),6.83(s,1H),6.26(dd,1H),5.07(dd,1H),4.12-4.04(m,2H),3.93(t,2H),3.83(s,3H),3.50-3.42(m,4H),3.40-3.34(m,3H),3.07(d,2H), 2.93-2.82(m,2H),2.68-2.55(m,5H),2.30(d,2H),2.05-1.98(m,1H),1.86-1.81(m,3H),1.78(d,6H),1.72-1.64(m,1H),1.47-1.32(m,4H),1.29-1.21(m,2H)。
实施例38
(±)-(Z)-N-(45-溴-26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-65-氟-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺38
采用实施例13的合成路线,将第四步原料化合物1e替换为N-(2-((5-溴-2-氯嘧啶-4-基)氨基)-4-氟-5-羟基苯基)-N-甲基甲磺酰胺(采用专利申请“KR102524856B1中说明书第27页中间体22”公开的方法制备而得),经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:65%-70%,流速:30mL/min),得到标题化合物38(外消旋体,13mg,产率:12%)。
MS m/z(ESI):1069.1[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.08(s,1H),8.22(s,1H),8.09(s,1H),8.08(s,1H),7.94(s,1H),7.75(s,1H),7.68(d,1H),7.43(d,1H),7.34(d,1H),7.26(d,1H),7.23(d,1H),6.96(s,1H),6.79(s,1H),5.07(dd,1H),4.22-4.15(m,1H),4.06(t,2H),3.86(s,3H),3.48-3.43(m,4H),3.39-3.26(m,3H),3.20(s,3H),3.09(s,3H),2.96(d,2H),2.92-2.83(m,1H),2.64-2.53(m,4H),2.28(d,2H),2.05-1.93(m,4H),1.81-1.71(m,4H),1.68-1.58(m,2H),1.35-1.26(m,2H)。
实施例39
(±)-(Z)-N-(45-溴-26-(4-((4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)-3,3-二氟哌啶-1-基)甲基)哌啶-1-基)-65-氟-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺39
采用实施例13的合成路线,将第四步中原料化合物1e替换为N-(2-((5-溴-2-氯嘧啶-4-基)氨基)-4-氟-5-羟基苯基)-N-甲基甲磺酰胺(采用专利申请“KR102524856B1中说明书第27页中间体22”公开的方法制备而得),以及将该合成路线中第七步的化合物13g替换为(±)-1-(6-(3,3-二氟哌啶-4-基)-1-甲基-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮盐酸盐(采用专利申请“WO2022235945 A1中说明书第180页”公开的方法制备而得),经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:60%-65%,流速:30mL/min),得到标题化合物39(外消旋体,23mg,产率:20%)。
MS m/z(ESI):1090.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.56(s,1H),8.22(s,1H),8.10(s,2H),7.95(s,1H),7.75(s,1H),7.59(d,1H),7.56(s,1H),7.43(d,1H),7.24(d,1H),7.09(d,1H),6.97(s,1H),6.79(s,1H),4.24-4.15(m,2H),4.10-4.03(m,2H),3.99(s,3H),3.92(t,2H),3.86(s,3H),3.26-3.14(m,4H),3.10(s,3H),3.03(d,1H),2.97(d,2H),2.76(t,2H),2.60-2.53(m,2H),2.45-2.34(m,3H),2.30-2.19(m,2H),2.03-1.92(m,3H),1.89-1.83(m,1H),1.82-1.71(m,4H),1.69-1.59(m,1H),1.37-1.27(m,2H)。
实施例40
(±)-(Z)-1-(6-(1-((1-(45-溴-62-(二甲基磷酰基)-65-氟-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)-3,3-二氟哌啶-4-基)-1-甲基-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮40

第一步
(2-((5-溴-2-氯嘧啶-4-基)氨基)-4-氟-5-甲氧基苯基)二甲基氧化膦40b
将5-溴-2,4-二氯嘧啶(1.6g,7.02mmol,上海毕得医药)、(2-氨基-4-氟-5-甲氧基苯基)二甲基氧化膦40a(1.53g,7.05mmol,采用专利申请“WO2023281417A1中说明书第44页中间体A14”公开的方法制备而得)溶于30mL异丙醇,加入5mL二异丙基乙胺,升温到90℃反应12小时。待反应液冷却到室温,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物40b(1.5g,产率:52%)。
MS m/z(ESI):407.9[M+1]。
第二步
(2-((5-溴-2-氯嘧啶-4-基)氨基)-4-氟-5-羟基苯基)二甲基氧化膦40c
将化合物40b(1.5g,3.67mmol)溶于20mL二氯甲烷,冰浴下滴加1M三溴化硼的二氯甲烷溶液(7.3mL),反应4小时。冰浴下,缓慢滴加甲醇淬灭反应,浓缩反应液,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物40c(900mg,产率:62%)。
MS m/z(ESI):391.9[M-1]。
第三步
(2-((5-溴-2-((4-(4-(二甲氧基甲基)哌啶-1-基)-2-甲氧基-5-(1H-吡唑-4-基)苯基)氨基)嘧啶-4-基)氨基)-4-氟-5-羟基苯基)二甲基氧化膦40d
将化合物40c(500mg,1.27mmol)和化合物13c(546mg,1.27mmol)溶于50mL甲醇,加入三氟乙酸(288mg,2.53mmol),氮气氛下,升温到80℃反应16小时。待反应液冷却至室温,减压除去溶剂,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物40d(610mg,产率:68%)。
MS m/z(ESI):704.1[M+1]。
第四步
(Z)-(45-溴-26-(4-(二甲氧基甲基)哌啶-1-基)-65-氟-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)二甲基氧化膦40e
将化合物40d(300mg,0.43mmol)、1,4-二溴丁烷(96mg,0.44mmol)、无水碳酸钾(118mg,0.85mmol)和四丁基碘化铵(15mg,0.045mmol)加入到25mL单口瓶中,加入4mL二甲基亚砜,反应1小时,然后升温到60℃反应12小时。减压除去溶剂,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物40e(197mg,产率:61%)。
MS m/z(ESI):756.1[M-1]。
第五步
(Z)-1-(45-溴-62-(二甲基磷酰基)-65-氟-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-甲醛40f
将化合物40e(190mg,0.25mmol)溶于6mL四氢呋喃,加入2M硫酸溶液(0.38mL),反应12小时。用饱和碳酸氢钠溶液调节反应液pH到7,用乙酸乙酯萃取(10mL×2),合并有机相,有机相用无水硫酸钠干燥,过滤,滤液浓缩,得到粗产品标题化合物40f(80mg,产率:45%),粗产品不经纯化直接投入下一步反应。
MS m/z(ESI):710.1[M-1]。
第六步
(±)-(Z)-1-(6-(1-((1-(45-溴-62-(二甲基磷酰基)-65-氟-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)-3,3-二氟哌啶-4-基)-1-甲基-1H-吲唑-3-基)二氢嘧啶-2,4(1H,3H)-二酮40
将化合物8h(35mg,0.088mmol)溶于6mL二氯甲烷和甲醇的混合溶剂(V/V=1/1)中,加入无水乙酸钠(43mg,0.052mmol),反应10分钟。然后加入化合物40f(60mg,0.084mmol),反应1小时。加入氰基硼氢化钠(11mg,0.18mmol),反应30分钟。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:50%-70%,流速:30mL/min),得到标题化合物40(外消旋体,13mg,产率:15%)。
MS m/z(ESI):1059.3[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.98(s,1H),10.57(s,1H),8.27(s,1H),8.05(s,1H),7.97(s,1H),7.86(d,1H),7.63(s,1H),7.54(s,1H),7.27-7.17(m,2H),7.14-7.06 (m,2H),6.80(s,1H),4.21-4.14(m,2H),4.13-4.08(m,2H),4.00(s,3H),3.92(t,2H),3.88(s,3H),3.03(d,2H),2.76(t,2H),2.65-2.50(m,12H),1.96-1.88(m,2H),1.80(d,6H),1.77-1.73(m,1H),1.68-1.60(m,2H),1.47-1.33(m,3H)。
实施例41
(±)-(Z)-5-(4-((1-(45-溴-62-(二甲基磷酰基)-65-氟-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)哌嗪-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮41
采用实施例40的合成路线,将第六步中间体8h替换为化合物13g,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:50%-70%,流速:30mL/min),得到标题化合物41(外消旋体,26mg,产率:22%)。
MS m/z(ESI):1038.2[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.07(s,1H),10.84(s,1H),8.23(s,1H),8.05(s,1H),7.97(s,1H),7.89-7.77(m,2H),7.68(d,1H),7.34(s,1H),7.31-7.21(m,2H),7.09(s,1H),6.80(s,1H),5.07(dd,1H),4.23-4.03(m,4H),3.87(s,3H),3.52-3.42(m,4H),3.05-2.93(m,2H),2.88(t,1H),2.66-2.52(m,8H),2.32-2.23(m,2H),2.06-1.98(m,1H),1.94-1.85(m,2H),1.84-1.71(m,8H),1.69-1.54(m,3H),1.47-1.25(m,2H)。
实施例42
(±)-(Z)-N-(45-氯-26-(4-((4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺42
采用实施例23的合成路线,将原料化合物13f替换为(Z)-N-(45-氯-26-(4-甲酰基哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂 -2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺(参考实施例13中合成路线,将第四步原料化合物1e替换为N-(2-((2,5-二氯嘧啶-4-基)氨基)-5-羟基苯基)-N-甲基甲磺酰胺(采用专利申请“WO2021216440中说明书第56页的实施例A2”公开的方法制备而得)),经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:50%-70%,流速:30mL/min),制得标题化合物42(外消旋体,60mg,产率:44%)。
MS m/z(ESI):992.3[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.85(s,1H),8.21(s,1H),8.15(s,1H),8.12(s,1H),7.90(s,1H),7.73(s,1H),7.71(d,1H),7.50(d,1H),7.26(s,1H),7.06(d,1H),6.92(dd,1H),6.84(d,1H),6.78(s,1H),6.14(dd,1H),4.29-4.20(m,3H),3.89(s,3H),3.85(s,3H),3.79(t,2H),3.26-3.19(m,4H),3.16(s,3H),3.07(s,3H),2.98(d,2H),2.65-2.51(m,8H),2.34-2.25(m,3H),2.19-2.12(m,1H),2.08-2.00(m,2H),1.75(d,2H),1.68-1.59(m,3H),1.35-1.26(m,2H)。
实施例43
(±)-(Z)-N-(45-溴-26-(4-((4-(3-(2,6-二氧代哌啶-3-基)-7-氟-1-甲基-1H-吲唑-6-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺43

第一步
6-溴-7-氟-3-碘-1-甲基-1H-吲唑43b
将6-溴-7-氟-3-碘-1H-吲唑43a(3.96g,11.62mmol,采用专利申请“WO2020210828 A1说明书第141页的实施例23”公开的方法制备而得)溶于30mL N,N-二甲基甲酰胺中,冰浴下加入氢化钠(540mg,14.09mmol),待不冒泡后加入碘甲烷(2.47g,17.40mmol),反应30分钟。向反应液中加入水(100mL),用乙酸乙酯萃取(100mL×3),合并有机相,有机相用饱和氯化钠溶液(100mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物43b(2.93g,产率:71%)。
MS m/z(ESI):354.8[M+1]。
第二步
3-(2,6-双(苄氧基)吡啶-3-基)-6-溴-7-氟-1-甲基-1H-吲唑43c
称取化合物43b(2.9g,8.17mmol)、化合物24b(3.41g,8.17mmol)到100mL茄形瓶中,依次加入1,1′-双(二苯基膦)二茂铁二氯化钯二氯甲烷络合物(598mg,0.82mmol)、无水碳酸钠(2.60g,24.53mmol)、48mL 1,4-二氧六环和水的混合溶剂(V/V=5/1),氮气置换三次,升温至90℃反应12小时。待反应液冷却到室温,将反应液倒入水(100mL)中,用乙酸乙酯萃取(100mL×3),合并有机相,有机相用饱和氯化钠溶液(100mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物43c(2.2g,产率:52%)。
MS m/z(ESI):518.1[M+1]。
第三步
4-(3-(2,6-双(苄氧基)吡啶-3-基)-7-氟-1-甲基-1H-吲唑-6-基)哌嗪-1-羧酸叔丁酯43d
称取化合物43c(1.1g,2.12mmol)、哌嗪-1-羧酸叔丁酯(593mg,3.18mmol)到100mL茄形瓶中,依次加入三(二亚苄基丙酮)二钯(198mg,0.21mmol)、1,1'-联萘-2,2'-双二苯膦(264mg,0.42mmol)、叔丁醇钠(408mg,4.24mmol)、甲苯(30mL),氮气置换三次,升温至100℃反应12小时。待反应液冷却到室温,将反应液倒入水(100mL)中,用乙酸乙酯萃取(100mL×3),合并有机相,有机相用饱和氯化钠溶液(100mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物43d(1.17g,产率:88%)。
MS m/z(ESI):624.3[M+1]。
第四步
(±)-4-(3-(2,6-二氧代哌啶-3-基)-7-氟-1-甲基-1H-吲唑-6-基)哌嗪-1-羧酸叔丁酯43e
将化合物43d(1.17g,1.88mmol)溶于30mL四氢呋喃和甲醇的混合溶剂(V/V=1/1)中,加入10%湿钯碳(含55%质量分数水,200mg,1.88mmol)和20%氢氧化钯碳(含55%质量分数水,260mg,1.85mmol)。氢气气氛下升温到60℃反应12小时。反应液经硅藻土过滤,滤液浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系A纯化,得到标题化合物43e(700mg,产率:84%)。
MS m/z(ESI):446.7[M+1]。
第五步
(±)-3-(7-氟-1-甲基-6-(哌嗪-1-基)-1H-吲唑-3-基)哌啶-2,6-二酮盐酸盐43f
将化合物43e(700mg,1.57mmol)溶于4mL二氯甲烷,加入4M氯化氢的1,4-二氧六环溶液(2mL),反应2小时。减压除去溶剂,真空干燥,得到粗产品标题化合物43f(500mg,98%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):346.3[M+1]。
第六步
(±)-(Z)-N-(45-溴-26-(4-((4-(3-(2,6-二氧代哌啶-3-基)-7-氟-1-甲基-1H-吲唑-6-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺43
将化合物43f(50mg,0.131mmol)溶于6mL二氯甲烷和甲醇的混合溶剂(V/V=1/1)中,加入无水乙酸钠(34mg,0.41mmol),反应10分钟。加入化合物13f(60mg,0.083mmol),反应1小时。加入三乙酰氧基硼氢化钠(53mg,0.25mmol),反应1小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:40%-85%,流速:30mL/min),得到标题化合物43(外消旋体,35mg,产率:40%)。
MS m/z(ESI):1054.6[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.90(s,1H),8.23(s,1H),8.18(s,1H),8.08(s,1H),7.89(s,1H),7.75(s,1H),7.72(d,1H),7.40(d,1H),7.25(s,1H),7.06(d,1H),6.92(dd,1H),6.77(s,1H),6.11(dd,1H),4.32(dd,1H),4.27-4.20(m,2H),4.07(s,3H),3.84(s,3H),3.78(t,2H),3.17(s,3H),3.14-3.08(m,4H),3.07(s,3H),2.98(d,2H),2.72-2.52(m,8H),2.38-2.25(m,3H),2.20-2.11(m,1H),2.07-2.00(m,2H),1.79-1.70(m,2H),1.68-1.58(m,3H),1.36-1.25(m,2H)。
实施例44
N-((10S,Z)-45-溴-26-(4-((4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌嗪-1-基)甲基)哌啶-1-yl)-24-甲氧基-10-甲基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺44
采用实施例13的合成路线,将第五步原料化合物1,4-二溴丁烷替换为(S)-2-甲基丁烷-1,4-二基二甲烷磺酸酯(采用公知的方法“Journal of Medicinal Chemistry,2018,61(17),7917-7928”制备而得),和将第七步原料化合物13g替换为化合物23a,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:45%-75%,流速:30mL/min),制得标题化合物44(一对非对映异构体混合物,比例1:1,11mg,产率:24%)。
MS m/z(ESI):1050.2[M+1]
1H NMR(500MHz,DMSO-d6)δ10.85(s,1H),8.23(s,1H),8.19(s,1H),8.07(s,1H),7.94(s,1H),7.75-7.70(m,2H),7.50(d,1H),7.22(s,1H),7.07(d,1H),6.92(d,1H),6.84(s,1H),6.78(s,1H),6.12(dd,1H),4.26(dd,1H),4.22(t,1H),4.19-4.14(m,1H),3.89(s,3H),3.85(s,3H),3.26-3.20(m,4H),3.18(s,3H),3.08(s,3H),3.03-2.92(m,2H),2.65-2.40(m,9H),2.33-2.25(m,2H),2.19-2.12(m,1H),2.03-1.95(m,1H),1.82-1.70(m,3H),1.68-1.60(m,2H),1.59-1.53(m,1H),1.49-1.42(m,1H),1.40-1.32(m,2H),1.08(d,3H)。
实施例45
(±)-(Z)-N-(45-溴-26-(4-((4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-10,10-二甲基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺45
采用实施例13的合成路线,将第五步原料化合物1,4-二溴丁烷替换为2,2-二甲基丁烷-1,4-二基二甲烷磺酸酯(采用公知的方法“Organic Letters,2019,21(23),9729-9733”制备而得),以及将第七步原料化合物13g替换为化合物23a,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:60%-80%,流速:30mL/min),制得标题化合物45(外消旋体,11mg,产率:34%)。
MS m/z(ESI):1064.3[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.85(s,1H),8.17(s,1H),8.14(s,1H),8.04(s,1H),7.91(s,1H),7.53(s,1H),7.50(d,1H),7.27(d,1H),6.96(d,1H),6.92(dd,1H),6.84(d,1H),6.77(s,1H),6.67(s,1H),6.28(dd,1H),4.25(dd,1H),4.23-4.18(m,2H),3.89(s,3H),3.85(s,3H),3.27(s,2H),3.25-3.21(m,4H),3.20(s,3H),3.07(s,3H),2.92-2.86(m,2H),2.65-2.53(m,7H),2.33-2.26(m,3H),2.20-2.12(m,1H),2.06-1.96(m,3H),1.74(d,2H),1.68-1.58(m,1H),1.36-1.28(m,2H),1.06(s,6H)。
实施例46
(±)-(Z)-5-(4-((1-(62-(二甲基磷酰基)-24-甲氧基-45-(三氟甲基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)哌嗪-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮46

第一步
(5-甲氧基-2-((2-(甲基硫基)-5-(三氟甲基)嘧啶-4-基)氨基)苯基)二甲基氧化膦46b
将4-氯-2-(甲基硫基)-5-(三氟甲基)嘧啶46a(800mg,3.50mmol,采用专利申请“WO2020231808 A1说明书第131页的中间体L-I-2”公开的方法制备而得)、化合物20a(700mg,3.51mmol)溶于25mL异丙醇,加入2mL二异丙基乙胺,升温至90℃反应16小时。减压除去溶剂,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物46b(170mg,12%)。
MS m/z(ESI):392.6[M+1]。
第二步
(2-((2-氯-5-(三氟甲基)嘧啶-4-基)氨基)-5-甲氧基苯基)二甲基氧化膦46c
将化合物46b(170mg,0.43mmol)溶于15mL二氯甲烷,于0℃加入磺酰氯(580mg,4.30mmol),0℃反应2小时。加入饱和碳酸氢钠溶液调节反应液pH到8,用二氯甲烷(15ml×2)萃取,合并有机相,有机相用无水硫酸钠干燥,过滤,滤液浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物46c(70mg,收率:42%)。
MS m/z(ESI):380.3[M+1]。
第三步
(2-((2-氯-5-(三氟甲基)嘧啶-4-基)氨基)-5-羟基苯基)二甲基氧化膦46d
将化合物46c(70mg,0.18mmol)加入到5mL二氯甲烷中,于0℃加入2M三溴化硼的二氯甲烷溶液(0.5mL),升至室温反应12小时。冰浴冷却,缓慢加入饱和碳酸氢钠溶液淬灭反应,用5mL氯仿和异丙醇(V/V=3/1)混合溶液萃取,有机 相减压浓缩,加入2mL乙酸乙酯和正己烷(V/V=1/1)的混合溶剂,过滤,用0.5mL甲醇洗涤滤饼,收集滤饼,真空干燥,得到粗产品标题化合物46d(67mg,收率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):366.3[M+1]。
第四步
(2-((2-((4-(4-(二甲氧基甲基)哌啶-1-基)-2-甲氧基-5-(1H-吡唑-4-基)苯基)氨基)-5-(三氟甲基)嘧啶-4-基)氨基)5-羟基苯基)二甲基氧化膦46e
将化合物46d(68mg,0.19mmol)和化合物13c(100mg,0.23mmol)溶于10mL甲醇,加入三氟乙酸(85mg,0.75mmol),升温至85℃反应16小时。减压除去溶剂,加入30mL二氯甲烷,加入饱和碳酸氢钠溶液调节反应液pH到7,有机相减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物46e(46mg,收率:36%)。
MS m/z(ESI):676.4[M+1]。
第五步
(Z)-(26-(4-(二甲氧基甲基)哌啶-1-基)-24-甲氧基-45-(三氟甲基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)二甲基氧化膦46f
将化合物46e(40mg,0.059mmol)溶于3mL二甲基亚砜,加入四丁基碘化铵(7mg,0.019mmol)、无水碳酸钾(25mg,0.18mmol)和1,4-二溴丁烷(13mg,0.06mmol),反应1小时,然后升温至55℃反应16小时。加入水(15mL),用乙酸乙酯(20mL×3)萃取,合并有机相,有机相用水(15mL×2)洗涤,饱和氯化钠溶液(15mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,真空干燥,得到粗产品标题化合物46f(35mg,收率:81%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):730.5[M+1]。
第六步
(Z)-1-(62-(二甲基磷酰基)-24-甲氧基-45-(三氟甲基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-甲醛46g
将化合物46f(35mg,0.048mmol)溶于3mL四氢呋喃,加入2M硫酸溶液(0.1mL),升温至55℃反应1小时。降温到0℃,加入饱和碳酸氢钠溶液调节反应液pH到7,用乙酸乙酯(10mL)萃取,有机相用无水硫酸钠干燥,过滤,滤液浓缩,得到粗产品标题化合物46g(33mg,收率:100%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):684.2[M+1]。
第七步
(±)-(Z)-5-(4-((1-(62-(二甲基磷酰基)-24-甲氧基-45-(三氟甲基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)哌嗪-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮46
将化合物13g(25mg,0.066mmol)加入到3.5mL甲醇和二氯甲烷的混合溶剂(V/V=1/6)中,加入无水乙酸钠(30mg,0.37mmol)反应30分钟,然后加入化合物46g(33mg,0.048mmol),反应1小时。加入三乙酰氧基硼氢化钠(21mg,0.099mmol)反应1小时。反应液浓缩,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:35%-95%,流速:30mL/min),得到标题化合物46(外消旋体,6mg,收率:12%)。
MS m/z(ESI):1008.7[M-1]。
1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),10.35(s,1H),8.33(s,1H),8.27(s,1H),8.24(s,1H),7.91(dd,1H),7.86(s,1H),7.68(d,1H),7.33(d,2H),7.26(d,1H),6.96(dd,1H),6.79(s,1H),6.14(d,1H),5.06(dd,1H),4.27-4.20(m,2H),3.84(s,3H),3.72(t,2H),3.47-3.42(m,4H),3.00(d,2H),2.88(td,1H),2.65-2.51(m,7H),2.27(d,2H),2.06-1.95(m,4H),1.79-1.74(m,2H),1.71(d,6H),1.66-1.58(m,3H),1.36-1.28(m,2H)。
实施例47
(±)-(Z)-N-(26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-45-(三氟甲基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺47
采用实施例46的合成路线,将第一步原料化合物20a替换为N-(2-氨基-5-甲氧基苯基)-N-甲基甲磺酰胺47a(采用专利申请“WO2021216440A1中说明书第56页实施例A2”公开的方法制备而得),经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-75%,流速:30mL/min),得到标题化合物47(外消旋体,28mg,产率:23%)。
MS m/z(ESI):1041.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.08(s,1H),8.33(s,1H),8.24(s,1H),8.19(s,1H),8.01(s,1H),7.85(s,1H),7.68(d,1H),7.63(d,1H),7.34(s,1H),7.26(d,1H),7.22(s,1H),7.04(d,1H),6.78(s,1H),6.13(dd,1H),5.07(dd,1H),4.27-4.21(m,2H),3.85(s,3H),3.76(t,2H),3.49-3.41(m,4H),3.15(s,3H),3.05(s,3H),2.99(d,2H), 2.92-2.83(m,1H),2.65-2.51(m,8H),2.28(d,2H),2.07-1.98(m,3H),1.79-1.72(m,2H),1.70-1.58(m,3H),1.36-1.26(m,2H)。
实施例48
(±)-(Z)-N-(45-溴-26-(4-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)氧基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺48
第一步
4-((1-(2-溴-5-甲氧基-4-硝基苯基)哌啶-4-基)氧基)哌啶-1-羧酸叔丁酯48b
将化合物1a(900mg,3.60mmol)和4-(哌啶-4-基氧基)哌啶-1-羧酸叔丁酯48a(1.33g,4.68mmol,采用公知的方法“WO2021077010A1中说明书第432页实施例170”制备得到)溶于20mL N,N-二甲基甲酰胺,加入无水碳酸钾(1.49g,10.78 mmol),升温到90℃反应过夜。反应液中加入水(30mL),用乙酸乙酯(30mL×3)萃取,合并有机相,用水(30mL×2)和饱和氯化钠溶液(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物48b(1.64g,产率:88%)。
MS m/z(ESI):458.1[M-55]。
第二步
4-((1-(5-甲氧基-4-硝基-2-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯基)哌啶-4-基)氧基)哌啶-1-羧酸叔丁酯48c
将化合物48b(910mg,1.77mmol)溶于1,4-二氧六环(20mL),加入1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-硼酸频哪醇酯(493mg,1.77mmol)、1,1'-双二苯基膦二茂铁二氯化钯(129mg,0.18mmol)、无水碳酸钠(562mg,5.30mmol),氮气置换三次。升温到100℃反应12小时。反应液经硅藻土过滤,用乙酸乙酯(20mL×2)萃取,合并有机相,用水(30mL)和饱和氯化钠溶液(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物48c(900mg,产率:87%)。
MS m/z(ESI):586.9[M+1]。
第三步
4-((1-(4-氨基-5-甲氧基-2-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯基)哌啶-4-基)氧基)哌啶-1-羧酸叔丁酯48d
将化合物48c(900mg,1.54mmol)溶于20mL甲醇,加入10%湿钯碳(含有55%质量分数水,350mg,1.64mmol),氢气氛围下反应18小时。反应液经硅藻土过滤,滤液减压浓缩,真空干燥,得到粗产品标题化合物48d(726mg,产率:85%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):556.3[M+1]。
第四步
4-((1-(4-((5-溴-4-((4-羟基-2-(N-甲基甲磺酰胺基)苯基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-(1H-吡唑-4-基)苯基)哌啶4-基)氧基)哌啶-1-羧酸叔丁酯48e
将化合物48d(710mg,1.28mmol)和化合物1e(520mg,1.28mmol)溶于20mL异丙醇,加入三氟乙酸(436mg,3.82mmol),90℃反应12小时。反应液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物48e(835mg,产率:78%)。
MS m/z(ESI):842.3[M+1]。
第五步
(Z)-4-((1-(45-溴-24-甲氧基-62-(N-甲基甲基磺酰胺基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)氧基)哌啶-1-羧酸叔丁酯48f
将化合物48e(356mg,0.42mmol)溶于30mL二甲亚砜,加入无水碳酸钾(175mg,1.27mmol)、四丁基碘化铵(42mg,0.13mmol)和1,4-二溴丁烷(137mg,0.63mmol),反应1小时,60℃反应12小时。向反应液中加入水(30mL),用乙酸乙酯(30mL×3)萃取,合并有机相,用水(30mL×2)和饱和氯化钠溶液(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物48f(350mg,产率:92%)。
MS m/z(ESI):896.5[M+1]。
第六步
(Z)-N-(45-溴-24-甲氧基-26-(4-(哌啶-4-基氧基)哌啶-1-基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺盐酸盐48g
将化合物48f(350mg,0.39mmol)溶于4mL二氯甲烷,加入4M氯化氢的1,4-二氧六环溶液(2mL),反应3小时。反应液减压浓缩,真空干燥,得到粗产品标题化合物48g(300mg,产率:92%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):796.3[M+1]。
第七步
(±)-(Z)-N-(45-溴-26-(4-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)氧基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺48
将化合物48g(108mg,0.13mmol)、2-(2,6-二氧代哌啶-3-基)-5-氟异吲哚啉-1,3-二酮48h(43mg,0.16mmol)溶于3mL N-甲基吡咯烷酮,加入N,N-二异丙基乙胺(84mg,0.65mmol),140℃微波反应2小时。反应液冷却至室温,经高效液相制备色谱法纯化(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:50%-95%,流速:30mL/min),得到标题化合物48(外消旋体,5mg,产率:4%)。
MS m/z(ESI):1052.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.08(s,1H),8.27(s,1H),8.19(s,1H),8.08(s,1H),7.87(s,1H),7.76(s,1H),7.72(d,1H),7.66(d,1H),7.34(d,1H),7.28-7.23(m,2H),7.05(d,1H),6.76(s,1H),6.10(dd,1H),5.07(dd,1H),4.27-4.20(m,2H),3.84(s,3H),3.82-3.72(m,2H),3.17(s,3H),3.07(s,3H),2.98-2.84(m,3H),2.66-2.56(m,7H),2.08-1.98(m,4H),1.94-1.86(m,4H),1.66-1.56(m,4H),1.54-1.41(m,4H)。
实施例49
(±)-(Z)-N-(45-溴-26-(4-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)(甲基)氨基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺49
采用实施例48的合成路线,将第一步原料化合物48a替换为4-(甲基(哌啶-4-基)氨基)哌啶-1-羧酸叔丁酯(采用公知的方法“WO2021058017A1中说明书第64页实施例16”制备得到),经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:35%-80%,流速:30mL/min),得到标题化合物49(外消旋体,18mg,产率:7%)。
MS m/z(ESI):1065.3[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),8.26(s,1H),8.18(s,1H),8.08(s,1H),7.87(s,1H),7.75(s,1H),7.73(d,1H),7.66(d,1H),7.32(d,1H),7.27-7.22(m,2H),7.05(d,1H),6.76(s,1H),6.10(dd,1H),5.06(dd,1H),4.27-4.19(m,2H),4.08(d,2H),3.84(s,3H),3.79(t,2H),3.17(s,3H),3.07(s,3H),3.04-2.95(m,3H),2.93-2.83(m,2H),2.64-2.53(m,6H),2.20(s,3H),2.09-1.95(m,4H),1.80-1.69(m,4H),1.67-1.59(m,3H),1.56-1.42(m,2H)。
实施例50
(±)-(Z)-N-(45-氯-26-(4-(4-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)甲基)哌嗪-1-基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺50
采用实施例11的合成路线,将第四步原料化合物1e替换为N-(2-((2,5-二氯嘧啶-4-基)氨基)-5-羟基苯基)-N-甲基甲磺酰胺(采用专利申请“WO2021216440中说明书第56页的实施例A2”公开的方法制备而得),经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-75%,流速:30mL/min),制得标题化合物50(外消旋体,18mg,产率:36%)。
MS m/z(ESI):1088.5[M-1]。
1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),8.21(s,1H),8.15(s,1H),8.12(s,1H),7.89(s,1H),7.75-7.68(m,2H),7.65(d,1H),7.30(s,1H),7.25(s,1H),7.22(d,1H),7.05(d,1H),6.76(s,1H),6.13(dd,1H),5.06(dd,1H),4.23(t,2H),4.03(d,2H),3.84(s,3H),3.79(t,2H),3.16(s,3H),3.07(s,3H),3.04-2.83(m,5H),2.62-2.53(m,4H),2.43-2.28(m,4H),2.22-2.10(m,3H),2.08-1.95(m,4H),1.87-1.73(m,5H),1.67-1.60(m,2H),1.59-1.42(m,3H),1.36-1.26(m,2H),1.18-1.08(d,2H)。
实施例51
(±)-(Z)-N-(45-溴-26-(4-((4-(4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)哌啶-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺51
采用实施例13的合成路线,将第七步原料化合物13g替换为2-(2,6-二氧代哌啶-3-基)-5-(4-(哌啶-4-基)哌嗪-1-基)异吲哚啉-1,3-二酮盐酸盐(采用专利申请“WO2018119441A1中说明书第483页实施例307”公开的方法制备而得),经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-75%,流速:30mL/min),得到标题化合物51(外消旋体,13mg,产率:24%)。
MS m/z(ESI):1134.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),8.21(s,1H),8.19(s,1H),8.07(s,1H),7.88(s,1H),7.74(s,1H),7.72(d,1H),7.67(d,1H),7.32(s,1H),7.27-7.22(m,2H),7.06(d,1H),6.76(s,1H),6.10(dd,1H),5.07(dd,1H),4.27-4.19(m,2H),3.84(s,3H),3.78(t,2H),3.46-3.38(m,4H),3.17(s,3H),3.07(s,3H),2.96(d,2H),2.92-2.83(m,3H),2.65-2.58(m,4H),2.57-2.51(m,4H),2.25-2.14(m,3H),2.06-1.97(m,3H),1.86(t,2H),1.79-1.73(m,2H),1.70(d,2H),1.63(t,2H),1.59-1.50(m,1H),1.48-1.37(m,2H),1.32-1.26(m,2H)。
实施例52
(±)-(Z)-N-(45-溴-26-(4-((4-(1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺52
第一步
4-(1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)哌嗪-1-羧酸叔丁酯52a
将化合物48h(150mg,0.54mmol)、4-(4-哌啶基)哌嗪-1-羧酸叔丁酯(150mg,0.56mmol)溶于5mL N-甲基吡咯烷酮,加入N,N-二异丙基乙胺(210mg,1.62mmol),140℃微波反应2小时。向反应液中加入水(20mL),用乙酸乙酯萃取(20mL×3),合并有机相,用水洗涤(30mL×2),无水硫酸钠干燥,过滤,滤液浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物52a(250mg,产率:88%)。
MS m/z(ESI):526.2[M+1]。
第二步
2-(2,6-二氧代哌啶-3-基)-5-(4-(哌嗪-1-基)哌啶-1-基)异吲哚啉-1,3-二酮二盐酸盐52b
将化合物52a(300mg,0.57mmol)溶于5mL二氯甲烷,加入4M氯化氢的1,4-二氧六环溶液(3mL),反应4小时。反应液减压浓缩,真空干燥,得到粗产品标题化合物52b(200mg,产率:70%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):426.2[M+1]。
第三步
(±)-(Z)-N-(45-溴-26-(4-((4-(1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌啶-4-基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺52
将化合物52b(27mg,0.054mmol)溶于4mL二氯甲烷和甲醇的混合溶剂中(V/V=3/1),加入无水乙酸钠(24mg,0.29mmol),反应10分钟。加入化合物13f(35mg,0.048mmol)和醋酸(11.4mg,0.19mmol),反应1小时。加入氰基硼氢化钠(6mg,0.1mmol),反应30分钟。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-75%,流速:30mL/min),得到标题化合物52(外消旋体,13mg,产率:24%)。
MS m/z(ESI):1134.3[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),8.20(s,1H),8.18(s,1H),8.07(s,1H),7.88(s,1H),7.74(s,1H),7.72(d,1H),7.65(d,1H),7.31(d,1H),7.26-7.21(m,2H),7.05(d,1H),6.76(s,1H),6.10(dd,1H),5.06(dd,1H),4.27-4.17(m,2H),4.05(d,2H),3.84(s,3H),3.78(t,2H),3.17(s,3H),3.07(s,3H),3.00-2.83(m,5H),2.25-2.26(m,13H),2.17(d,2H),2.07-1.96(m,3H),1.84(d,2H),1.69(d,2H),1.63(t,2H),1.58-1.51(m,1H),1.49-1.38(m,2H),1.32-1.24(m,2H)。
实施例53
(±)-(Z)-N-(45-溴-26-(4-((1'-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)-[4,4'-联哌啶]-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺53
采用实施例52的合成路线,将第一步原料化合物4-(4-哌啶基)哌嗪-1-羧酸叔丁酯替换为[4,4'-联哌啶]-1-羧酸叔丁酯(上海毕得医药),经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:70%-90%,流速:30mL/min),得到标题化合物53(外消旋体,20mg,产率:36%)。
MS m/z(ESI):1131.6[M-1]。
1H NMR(500MHz,DMSO-d6):δ11.07(s,1H),8.20(s,1H),8.18(s,1H),8.07(s,1H),7.88(s,1H),7.74(s,1H),7.72(d,1H),7.64(d,1H),7.30(s,1H),7.26-7.18(m,2H),7.05(d,1H),6.76(s,1H),6.10(dd,1H),5.05(dd,1H),4.25-4.19(m,2H),4.07(d,2H),3.84(s,3H),3.78(m,2H),3.17(s,3H),3.07(s,3H),3.00-2.82(m,6H),2.65-2.53(m,3H),2.15(d,2H),2.06-1.94(m,4H),1.83-1.51(m,11H),1.38-1.12(m,7H),1.08-0.99(m,2H)。
实施例54
(±)-(Z)-5-(4-((1-(45-氯-62-(二甲基磷酰基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十二蕃-26-基)哌啶-4-基)甲基)哌嗪-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮54
采用实施例20的合成路线,将第一步合成原料14f替换为2,4,5-三氯嘧啶,和将第四步合成原料1,4-二溴丁烷替换为1,5-二溴戊烷。经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:35%-65%,流速:30mL/min),制得标题化合物54(外消旋体,8mg,产率:10%)。
MS m/z(ESI):990.7[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.11-11.06(m,2H),8.33(dd,1H),8.08(s,1H),7.96(d,1H),7.95(s,1H),7.73(s,1H),7.68(d,1H),7.62(s,1H),7.35(s,1H),7.26(d,1H),7.04(dd,1H),6.83(s,1H),6.24(dd,1H),5.07(dd,1H),4.12-4.05(m,2H),3.94(t,2H),3.83(s,3H),3.52-3.42(m,4H),3.08(d,2H),2.93-2.83(m,1H),2.68-2.52(m,9H),2.30(d,2H),2.05-1.95(m,2H),1.85-1.74(m,9H),1.73-1.63(m,1H),1.49-1.32(m,5H)。
实施例55
(±)-(Z)-4-(4-((1-(45-溴-24-甲氧基-62-(N-甲基甲磺酰胺基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)哌啶-4-基)甲基)哌嗪-1-基)-N-(2,6-二氧代哌啶-3-基)-2-氟苯甲酰胺55
将(±)-N-(2,6-二氧代哌啶-3-基)-2-氟-4-(哌嗪-1-基)苯甲酰胺盐酸盐55a(采用专利申请“WO2023017446 A1说明书第863页的实施例5”公开的方法制备而得)(40 mg,0.11mmol)溶于4mL二氯甲烷和甲醇的混合溶剂中(V/V=1/1),加入无水乙酸钠(68mg,0.83mmol),反应10分钟。加入化合物13f(60mg,0.083mmol),反应1小时。加入三乙酰氧基硼氢化钠(53mg,0.25mmol),反应1小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-80%,流速:30mL/min),得到标题化合物55(外消旋体,28mg,产率:32%)。
MS m/z(ESI):1043.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.84(s,1H),8.22(s,1H),8.19(s,1H),8.08(s,1H),8.05(t,1H),7.89(s,1H),7.75(s,1H),7.72(d,1H),7.63(t,1H),7.25(s,1H),7.06(d,1H),6.86-6.73(m,3H),6.11(dd,1H),4.76-4.69(m,1H),4.23(t,2H),3.85(s,3H),3.79(t,2H),3.31-3.25(m,4H),3.17(s,3H),3.07(s,3H),2.98(d,2H),2.76(td,1H),2.65-2.52(m,6H),2.31-2.23(m,2H),2.17-1.95(m,5H),1.74(d,3H),1.68-1.58(m,3H),1.35-1.27(m,1H)。
实施例56
(±)-(Z)-N-(45-溴-26-(4-(4-(2-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)乙基)哌嗪-1-基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺56
第一步
(E)-3-(2,6-双(苄氧基)吡啶-3-基)-6-(2-乙氧基乙烯基)-1-甲基-1H-吲唑56a
将化合物32a(1.0g,2.00mmol)和(E)-2-(2-乙氧基乙烯基)-4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷(594mg,3.00mmol,韶远科技有限公司)溶于26mL 1,4-二氧六环和水的混合溶剂中(V/V=10/3),加入四三苯基膦钯(231mg,0.20mmol)和无水碳酸钠(635mg,5.99mmol)。氮气置换三次,于100℃反应16小时。加水(15mL),用乙酸乙酯(20mL×3)萃取,合并有机相,有机相用饱和氯化钠溶液(30mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系B纯化得到标题化合物56a(785mg,产率:80%)。
MS m/z(ESI):492.2[M+1]。
第二步
3-(2,6-双(苄氧基)吡啶-3-基)-6-(2,2-二乙氧基乙基)-1-甲基-1H-吲唑56b
将化合物56a(485mg,0.99mmol)溶于30mL无水乙醇,加入对甲苯磺酸一水合物(94mg,0.49mmol),于80℃反应1小时。待反应液冷却到室温,减压浓缩至反应液剩余5mL,加入饱和碳酸氢钠溶液调节反应液pH到7,用乙酸乙酯萃取(10mL×3),合并有机相,有机相用饱和氯化钠溶液(15mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系B纯化得到标题化合物56b(460mg,产率:87%)。
MS m/z(ESI):538.3[M+1]。
第三步
3-(6-(2,2-二乙氧基乙基)-1-甲基-1H-吲唑-3-基)哌啶-2,6-二酮56c
将化合物56b(460mg,0.86mmol)溶于60mL四氢呋喃和甲醇的混合溶剂(V/V=1/1)中,加入10%湿钯碳(含55%质量分数水,91mg,0.86mmol)和20%氢氧化钯碳(含55%质量分数水,120mg,0.85mmol)。于氢气氛围下60℃反应12小时。反应液用硅藻土过滤,滤液浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物56c(246mg,产率:80%)。
MS m/z(ESI):360.2[M+1]。
第四步
2-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)乙醛56d
将化合物56c(60mg,0.17mmol)溶于2mL四氢呋喃,加入2M硫酸溶液(0.8mL),反应3小时。向反应液中加入饱和碳酸氢钠溶液调节反应液pH到7,用乙酸乙酯萃取(5mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(10mL),无水硫酸钠干燥,过滤,滤液浓缩,真空干燥,得粗产品标题化合物56d(35mg,产率:73%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):286.1[M+1]。
第五步
(±)-(Z)-N-(45-溴-26-(4-(4-(2-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)乙基)哌嗪-1-基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺56
将化合物11f(50mg,0.058mmol)和化合物56d(35mg,0.12mmol)溶于5mL二氯甲烷和甲醇的混合溶剂中(V/V=4/1),加入无水乙酸钠(15mg,0.18mmol)和乙酸(7mg,0.12mmol)反应12小时。加入氰基硼氢化钠(8mg,0.13mmol),反应1.5小时。反应液过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:45%-65%,流速:30mL/min),得到标题化合物56(外消旋体,19mg,产率:29%)。
MS m/z(ESI):1048.4[M-1]。
1H NMR(500MHz,DMSO-d6)δ10.88(s,1H),8.23(s,1H),8.19(s,1H),8.08(s,1H),7.87(s,1H),7.75(s,1H),7.73(d,1H),7.60(d,1H),7.43(s,1H),7.25(s,1H),7.06(d,1H),7.01(d,1H),6.76(s,1H),6.10(dd,1H),4.33(dd,1H),4.23(t,2H),3.96(s,3H),3.84(s,3H),3.79(t,2H),3.17(s,3H),3.07(s,3H),3.01(d,2H),2.87(t,2H),2.72-2.29(m,11H),2.22-2.13(m,2H),2.08-1.96(m,3H),1.81(d,2H),1.68-1.59(m,2H),1.57-1.42(m,3H),1.32-1.24(m,2H)。
实施例57
(±)-(Z)-N-(45-溴-26-(4-(4-(4-(2,6-二氧代哌啶-3-基)-3,5-二氟苯乙基)哌嗪-1-基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺57
将化合物11f(50mg,0.059mmol)溶于3.5mL二氯甲烷和甲醇(V/V=6/1)的混合溶剂中,加入无水乙酸钠(50mg,0.61mmol),反应10分钟,加入(±)-2-(4-(2,6-二氧代哌啶-3-基)-3,5-二氟苯基)乙醛57a(采用专利申请“WO2022227032A1中说明书第19页的实施例3”公开的方法制备而得)(20mg,0.075mmol),继续反应10分钟,加入氰基硼氢化钠(7mg,0.12mmol),反应10分钟。反应液减压浓缩,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和 乙腈,乙腈的梯度:50%-95%,流速:30mL/min),得到标题化合物57(外消旋体,23mg,收率:36%)。
MS m/z(ESI):1032.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.95(s,1H),8.22(s,1H),8.19(s,1H),8.08(s,1H),7.87(s,1H),7.77-7.70(m,2H),7.25(s,1H),7.08-7.00(m,3H),6.75(s,1H),6.10(dd,1H),4.27-4.16(m,3H),3.84(s,3H),3.79(t,2H),3.17(s,3H),3.07(s,3H),3.01(d,2H),2.85-2.72(m,3H),2.62-2.39(m,12H),2.26-1.96(m,6H),1.86-1.77(m,2H),1.67-1.60(m,2H),1.58-1.44(m,2H)。
实施例58
(±)-(Z)-N-(45-溴-26-(4-(2-(4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)乙基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺58
采用实施例13的合成路线,将第一步原料化合物4-(2,2-二甲氧基甲基)哌啶替换为4-(2,2-二甲氧基乙基)哌啶(采用公知的方法“WO2020201080A1中说明书第63页实施例2”制备得到),经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:50%-70%,流速:30mL/min),得到标题化合物58(外消旋体,18mg,产率:7%)。
MS m/z(ESI):1065.3[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.08(s,1H),8.22(s,1H),8.19(s,1H),8.07(s,1H),7.88(s,1H),7.74(s,1H),7.73(d,1H),7.69(d,1H),7.36(s,1H),7.30-7.23(m,2H),7.06(d,1H),6.76(s,1H),6.10(dd,1H),5.07(dd,1H),4.26-4.19(m,2H),3.82(s,3H),3.81-3.76(m,2H),3.51-3.39(m,4H),3.17(s,3H),3.07(s,3H),2.96(d,2H),2.92-2.83(m 1H),2.65-2.32(m,7H),2.08-1.95(m,4H),1.71(d,2H),1.68-1.60(m,2H),1.56-1.43(m,3H),1.40-1.27(m,4H)。
实施例59
(±)-(Z)-N-(45-溴-26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)-4-羟基哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺59
第一步
4-((1-((苄氧基)羰基)-4-羟基哌啶-4-基)甲基)哌嗪-1-羧酸叔丁酯59b
将哌嗪-1-羧酸叔丁酯(186mg,1.00mmol)和1-氧杂-6-氮杂螺[2.5]辛烷-6-羧酸苄酯59a(370mg,1.50mmol,上海毕得医药)溶于3mL乙醇,加入N,N-二异丙基乙胺(258mg,2.00mmol),升温到80℃反应2小时。反应液减压浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系B纯化得到标题化合物59b(380mg,产率:87%)。
MS m/z(ESI):434.2[M+1]。
第二步
4-((4-羟基哌啶-4-基)甲基)哌嗪-1-羧酸叔丁酯59c
称取化合物59b(380mg,0.88mmol)到50mL单口瓶中,加入5mL甲醇,然后加入10%湿钯碳(含55%质量分数水,76mg,0.63mmol)。氢气氛下反应2小时。 反应液用硅藻土过滤,滤液浓缩,真空干燥,得到粗产品标题化合物59c(262mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):300.2[M+1]。
第三步
4-((1-(2-溴-5-甲氧基-4-硝基苯基)-4-羟基哌啶-4-基)甲基)哌嗪-1-羧酸叔丁酯59d
将化合物59c(263mg,0.88mmol)溶于16mL二甲基亚砜,加入化合物1a(220mg,0.88mmol)和N,N-二异丙基乙胺(284mg,2.19mmol),升温到90℃反应16小时。待反应液冷却至室温,向反应液中加入水(30mL),用乙酸乙酯萃取(40mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥。过滤、滤液减压浓缩,所得残余物用硅胶柱色谱法以洗脱剂体系B纯化得到标题化合物59d(340mg,产率:72%)。
MS m/z(ESI):527.1[M-1]。
第四步
4-((4-羟基-1-(5-甲氧基-4-硝基-2-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯基)哌啶-4-基)甲基)哌嗪-1-羧酸叔丁酯59e
称取化合物59d(340mg,0.64mmol)、1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-硼酸频哪醇酯(268mg,0.96mmol)到100mL茄形瓶中,加入1,1′-双(二苯基膦)二茂铁二氯化钯(70mg,0.096mmol)、无水碳酸钠(204mg,1.93mmol)、12mL 1,4-二氧六环和水的混合溶剂(V/V=5/1),氮气置换三次,升温至90℃反应4小时。待反应冷液却到室温,用硅藻土过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物59e(320mg,产率:82%)。
MS m/z(ESI):599.3[M-1]。
第五步
4-((1-(4-氨基-5-甲氧基-2-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-4-基)苯基)-4-羟基哌啶-4-基)甲基)哌嗪-1-羧酸叔丁酯59f
称取化合物59e(320mg,0.53mmol)到100mL单口瓶中,加入10mL甲醇,然后加入10%湿钯碳(含55%质量分数水,128mg,1.06mmol)。氢气气氛下反应12小时。反应液用硅藻土过滤,滤液浓缩,真空干燥,得到粗产品标题化合物59f(304mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):571.3[M+1]。
第六步
4-((1-(4-((5-溴-4-((4-羟基-2-(N-甲基甲磺酰胺基)苯基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-(1H-吡唑-4-基)苯基)-4-羟基哌啶-4-基)甲基)哌嗪-1-羧酸叔丁酯59g
将化合物1e(190mg,0.47mmol)、化合物59f(306mg,0.54mmol)、三氟乙酸(159mg,1.40mmol)加入到20mL异丙醇,升温至90℃反应12小时。待反应液 冷却至室温,向反应液中加入饱和碳酸氢钠溶液(15mL),用乙酸乙酯萃取(20mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物59g(210mg,产率:52%)。
MS m/z(ESI):857.3[M+1]。
第七步
(Z)-4-((1-(45-溴-24-甲氧基-62-(N-甲基甲磺酰胺基)-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-26-基)-4-羟基哌啶-4-基)甲基)哌嗪-1-羧酸叔丁酯59h
称取化合物59g(119mg,0.14mmol)、1,4-二溴丁烷(46mg,0.22mmol)到50mL单口瓶中,加入6mL二甲基亚砜、无水碳酸钾(67mg,0.49mmol)和四丁基碘化铵(17mg,0.046mmol),升温至60℃反应3小时。待反应液冷却至室温,向反应液中加入水(10mL),用乙酸乙酯萃取(15mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(20mL),无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物59h(67mg,产率:52%)。
MS m/z(ESI):911.3[M+1]。
第八步
(Z)-N-(45-溴-26-(4-羟基-4-(哌嗪-1-基甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺二三氟乙酸盐59i
将化合物59h(67mg,0.073mmol)溶于1.5mL二氯甲烷,加入0.5mL三氟乙酸,反应1小时。减压除去溶剂,向反应液中加入饱和碳酸氢钠溶液(5mL),用二氯甲烷萃取(8mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(15mL),无水硫酸钠干燥。过滤,滤液减压浓缩,得到粗产品标题化合物59i(59mg,产率:78%),粗产品直接用于下一步反应。
MS m/z(ESI):811.3[M+1]。
第九步
(±)-(Z)-N-(45-溴-26-(4-((4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)哌嗪-1-基)甲基)-4-羟基哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺59
称取化合物59i(59mg,0.057mmol)到5mL微波反应管中,加入3mL N-甲基吡咯烷酮、化合物48h(30mg,0.11mmol)和N,N-二异丙基乙胺(28mg,0.22mmol),然后升温到140℃微波反应3小时。待反应液冷却至室温,过滤,经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙 腈,乙腈的梯度:57%-63%,流速:30mL/min),得到标题化合物59(外消旋体,24mg,产率:40%)。
MS m/z(ESI):1067.3[M+1]。
1H NMR(500MHz,DMSO)δ10.89(s,1H),8.26(s,1H),8.18(s,1H),8.07(s,1H),7.87(s,1H),7.80-7.70(m,2H),7.68(d,1H),7.34(d,1H),7.30-7.24(m,2H),7.04(s,1H),6.78(s,1H),6.09(dd,1H),5.05(dd,1H),4.24-4.17(m,2H),4.15-4.08(m,1H),3.85(s,3H),3.79(t,2H),3.51-3.41(m,4H),3.17(s,3H),3.07(s,3H),2.91-2.81(m,3H),2.76-2.66(m,6H),2.64-2.53(m,2H),2.39(s,2H),2.05-1.95(m,3H),1.75-1.68(m,2H),1.65-1.53(m,4H)。
实施例60
N-((Z)-45-溴-26-(4-(((2S)-4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)-2-(羟甲基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺60
采用实施例52的合成路线,将第一步原料化合物4-(4-哌啶基)哌嗪-1-羧酸叔丁酯替换为(S)-2-(羟甲基)哌嗪-1-羧酸叔丁酯(上海毕得医药),经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:55%-75%,流速:30mL/min),得到标题化合物60(一对非对映异构体混合物,比例1:1,20mg,产率:22%)。
MS m/z(ESI):1081.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.09(s,1H),8.23(s,1H),8.19(s,1H),8.08(s,1H),7.88(s,1H),7.76(s,1H),7.73(d,1H),7.68(d,1H),7.29(s,1H),7.26(s,1H),7.20(d,1H),7.06(d,1H),6.77(s,1H),6.10(dd,1H),5.07(dd,1H),4.72(s,1H),4.23(t,2H),3.84(s,3H),3.78(t,2H),3.72-3.56(m,4H),3.31-3.21(m,2H),3.17(s,3H),3.07(s,3H),3.01-2.94(m,2H),2.93-2.83(m,2H),2.72-2.54(m,6H),2.45-2.38(m1H),2.30-2.22(m,1H),2.09-1.96(m,3H),1.78(d,1H),1.73-1.50(m,4H),1.35-1.25(m,2H)。
实施例61
N-((Z)-45-溴-26-(4-(((2R)-4-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)-2-(羟甲基)哌嗪-1-基)甲基)哌啶-1-基)-24-甲氧基-11H-7-氧杂-3,5-二氮杂-4(2,4)-嘧啶杂-1(4,1)-吡唑杂-2(1,3),6(1,4)-二苯杂环十一蕃-62-基)-N-甲基甲磺酰胺61
采用实施例52的合成路线,将第一步原料化合物4-(4-哌啶基)哌嗪-1-羧酸叔丁酯替换为(R)-2-(羟甲基)哌嗪-1-羧酸叔丁酯(上海毕得医药),经高效液相制备色谱法纯化(Waters-2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:60%-80%,流速:30mL/min),得到标题化合物61(一对非对映异构体混合物,比例1:1,29mg,产率:24%)。
MS m/z(ESI):1081.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.09(s,1H),8.23(s,1H),8.22(s,1H),8.17(s,1H),7.90(s,1H),7.86(s,1H),7.77(d,1H),7.72(d,1H),7.49(s,1H),7.37(d,1H),7.26(s,1H),7.06(d,1H),6.76(s,1H),6.13(dd,1H),5.74(s,1H),5.10(dd,1H),4.33-4.20(m,3H),4.15(d,1H),4.02(d,1H),3.86(s,3H),3.83-3.76(m,3H),3.63(d,1H),3.55-3.40(m,5H),3.18(s,3H),3.08(s,3H),3.05-2.97(m,2H),2.95-2.84(m,1H),2.65-2.53(m,5H),2.10-1.91(m,5H),1.76-1.69(m,1H),1.68-1.60(m,2H),1.54-1.37(m,2H)。
生物学评价
测试例1、本公开化合物对H1975/EGFR L858R-T790M-C797S、BaF3/EGFR L858R-T790M-C797S、BaF3/EGFR L858R-C797S、BaF3/EGFR Del19-C797S、BaF3/EGFR Del19-T790M-C797S、H1975/EGFR L858R-T790M、HCC827/EGFR Del19和A431/EGFR WT细胞的增殖抑制测定
1)实验材料与仪器
H1975/EGFR L858R-T790M-C797S为内部构建的稳转细胞株Pool,在NCI-H1975细胞(ATCC CRL-5908)中通过慢病毒感染导入pCDH-CMV-T790M-C797S-L858R基因,然后通过嘌呤霉素筛选获得。
BaF3/EGFR L858R-T790M-C797S(也即BaF3/LTC,KC-0122)购于康源博创,BaF3/EGFR L858R-C797S(也即BaF3/LC,CBP73047)、BaF3/EGFR Del19-C797S(也即BaF3DC,CBP73172)、BaF3/EGFR Del19-T790M-C797S(也即BAF3/DTC,CBP73173)购于南京科佰,H1975/EGFR L858R-T790M(也即H1975/LT,CRL-5908)、HCC827/EGFR Del19(也即HCC827/D,CRL-2868)、A431/EGFR WT(CRL-1555)购于ATCC。
DMEM培养基(Corning,10-013-CV)
RPMI1640培养基(美仑,PWL015)
嘌呤霉素(10mg/ml)(ThermoFisher,A11138-03)
0.25%胰蛋白酶-EDTA(1×)(Gibco,25200-072)
青霉素-链霉素(Gibco,15140-122)
DPBS(1×)(Gibco,14190-144)
FBS(Gibco,10091148)
GlutaMAX(100×)(Gibco,35050-061)
T75 Flask(TITAN,02055020)
384细胞培养板(白色不透明384孔微孔板,无菌和组织培养处理)(PE,6007680)
96孔圆底配药板(未处理)(JET BIOFIL,TCP-002-096)
CellTiter-Glo缓冲液(Promega,G756B)
CellTiter-Glo底物(Promega,G755B)
自动细胞计数仪(Countstar,IC1000)
离心机(Thermo,75004524)
恒温培养箱(Thermo,I160)
EnVision多模式读板仪(PerkinElmer,EnVision 2105)
2)实验方法
2.1、贴壁细胞铺板(第0天)
a、显微镜下观察细胞状态,确保细胞的融合度在~90%。
b、弃去细胞的上清,PBS润洗一次,倒去PBS。加入适量胰蛋白酶消化细胞,37℃静置5分钟。
c、用等体积含10%FBS的培养基终止消化,收集细胞悬液。300g,离心3分钟。用适量新鲜培养基悬浮细胞。
d、取重悬的细胞悬液20μL进行计数,确保细胞活率大于90%。
e、A431/WT、H1975/LTC、H1975/LT和HCC827/D:根据细胞计数结果,用含10%FBS的DMEM或含10%FBS的1640培养基(H1975/LTC另加2μg/ml嘌呤霉素)稀释细胞至相应铺板密度,分别按照每孔2000、300、300、600个细胞的密度将细胞接种于384孔培养板中,每孔20μL。(不同化合物根据可作相应调整)
f、将细胞培养板放在37℃,5%二氧化碳的培养箱中培养过夜。
2.2、悬浮细胞铺板(第1天)
a、显微镜下观察细胞状态。
b、将细胞悬液收集至15mL离心管中,300g,离心4分钟。用适量新鲜培养基悬浮细胞。
c、取重悬的细胞悬液20μL进行细胞计数,确保细胞活率大于90%。
d、BaF3/LTC、BaF3/LC、BaF3/DTC、BaF3/DC:根据细胞计数结果,用含10%FBS的1640培养基稀释细胞至相应铺板密度,按照每孔400个细胞的密度将细胞接种于384孔培养板中,每孔20μL。
2.3、加药(第1天)
a、化合物起始浓度为10mM或3.3mM,用DMSO将每个化合物3倍梯度稀释成10个浓度点(不同化合物最高浓度根据IC50的不同可作相应调整)。例如,在96孔圆底配药板中,将3μL化合物依次梯度稀释至6μL DMSO中。
b、将每个化合物各浓度点按500倍稀释到相应体积1640细胞培液中(H1975/LTC培液:含2μg/ml嘌呤霉素)。
c、向每个细胞板20μL/孔的细胞上清中依次加入20μL上述稀释好的化合物溶液。
d、将加完药的细胞板放置在37℃,5%二氧化碳的培养箱培养。
2.4、CTG检测(第4天)
a、使用前将CellTiter-Glo缓冲液和冻干的CellTiter-Glo底物放置平衡到室温,两者混合充分混匀配制成100mL的CellTiter-Glo试剂(或将已混好的CellTiter-Glo试剂从-20℃拿出平衡到室温)。
b、将待检测的板子从培养箱中取出,平衡至室温,每孔加入20μL CellTiter-Glo试剂。
c、振荡混匀2分钟使细胞充分裂解。
d、室温放置28分钟待信号稳定后在EnVision上进行检测发光信号值(RLU)。
2.5、IC50值计算
a、通过下列公式计算化合物各浓度的抑制率,使用XLfit根据化合物的对数浓度和抑制率进行曲线拟合并计算IC50值。
b、抑制率(%)=100-100×(RLU化合物-RLU空白对照)/(RLU溶媒对照-RLU空白对照)
本公开化合物生物活性由上述分析所得,计算所得的IC50如下表1和表2:
表1本公开化合物对H1975/EGFR L858R-T790M-C797S细胞增殖抑制活性
表2、本公开化合物对BaF3/LTC、BAF3/LC、BaF3/DTC、BaF3/DC、H1975/LT、HCC827/D和A431/WT细胞的增殖抑制活性

结论:本公开化合物对H1975/LTC、BaF3/LTC、BAF3/LC、BaF3/DTC、BaF3/DC、H1975/LT和HCC827/D细胞具有明显的选择性增殖抑制活性。
测试例2、本公开化合物对BaF3/EGFR L858R-T790M-C797S细胞总EGFR蛋白降解的测定
1.实验材料与仪器:
BaF3/EGFR L858R-T790M-C797S(康源博创,KC-0122)
1640培养基(美仑,PWL015)
青霉素-链霉素(Gibco,15140-122)
DPBS(1×)(Gibco,14190-144)
FBS(Gibco,10091148)
T75 Flask(TITAN,02055020)
96孔细胞培养板(Corning,3788)
96孔圆底配药板(未处理)(JET BIOFIL,TCP-002-096)
1.2mL 96孔深孔板(透明,无菌,方形孔,V形底)(TITAN,02089063)
总EGFR细胞试剂盒(PerkinElmer,64NG1PEG)
自动细胞计数仪(Countstar,IC1000)
离心机(Thermo,75004524)恒温培养箱(Thermo,I160)
微孔板混匀仪(Thermo,88880024)
EnVision多模式读板仪(PerkinElmer,EnVision 2105)
2.实验方法
2.1悬浮细胞铺板(第1天):
a、显微镜下观察细胞状态。
b、将细胞悬液收集至15mL离心管中,300g,离心4分钟。用适量新鲜培养基悬浮细胞。
c、取重悬的细胞悬液20μL进行细胞计数,确保细胞活率大于90%。
d、根据细胞计数结果,用含10%FBS的1640培养基稀释细胞至相应铺板密度,按照每孔10000-20000个细胞的密度将细胞接种于96孔培养板中,每孔25μL。
2.2加药(第1天)
a.化合物起始浓度为10mM,用DMSO将每个化合物5倍梯度稀释成8个浓度点。例如,在96孔圆底配药板中,将3μL化合物依次梯度稀释至12μL DMSO中。
b.将每个化合物各浓度点按167倍稀释到相应体积1640细胞培液中。
c.向每个细胞板25μL/孔的细胞悬液中依次加入5μL上述稀释好的化合物溶液。
d.将加完药的细胞板放置在37℃,5%二氧化碳的培养箱中培养6小时。
2.3收样及孵育抗体(第1天)
a.将96孔板拿出,按照总EGFR细胞试剂盒说明书所示每孔加入10uL(4×)*
补充裂解液(1份体积的封闭缓冲液和24份体积的4×裂解缓冲液),贴好透明薄膜,盖上板盖,短暂甩板,室温震荡裂解60min(800rpm)。
b.将裂解后的液体转移16μL至384孔板中,加入4μL预混好的抗体(d2抗体:Eu抗体=1:1预混),贴好铝箔膜,盖上板盖,室温震荡1分钟,短暂甩板,室温孵育过夜。
2.4 HTRF检测(第2天)
a.将待检测的384孔板从培养箱中取出,室温震荡1分钟,短暂甩板。
b.在EnVision上读取665nm和620nm两个不同波长的荧光发射信号值。
2.5DC50值和最大降解率计算
a.通过下列公式计算化合物各浓度的总EGFR降解率,其中最大值(Max)为化合物各浓度点降解总EGFR的最大降解率。使用XLfit根据化合物的对数浓度和降解率进行曲线拟合并计算DC50值。
b.计算每孔受体和供体发射信号的比率:Ratio=Signal 665nm/Signal 620nm×104
c.降解率(%)=100-100*(Ratio化合物-Ratio空白对照)/(Ratio溶媒对照-Ratio空白对照)
本公开化合物总EGFR降解活性由上述分析所得,计算所得的DC50如下表3:
表3、本公开化合物对BaF3/LTC细胞总EGFR蛋白降解活性
结论:本公开化合物对BaF3/LTC细胞总EGFR蛋白具有明显的降解活性。
测试例3、药代动力学评价
一、C57小鼠试验
1、摘要
以C57小鼠为受试动物,应用LC/MS/MS法测定了C57小鼠灌胃(i.g.)给予实施例化合物后不同时刻血浆中的药物浓度,研究本公开化合物在C57小鼠体内的药代动力学行为,评价其药动学特征。
2、试验方案
2.1、试验药品
化合物13、20、23、23-1A、23-2A、31和31-1。
2.2、试验动物
C57小鼠63只,雌性,平均分成7组,每组9只,由维通利华实验动物技术有限公司提供,生产许可证SCXK(浙)2019-0001,SCXK(京)2021-0006。
2.3、药物配制
分别称取一定量的受试化合物,加5%DMSO+5%吐温80+90%生理盐水,配制成0.1mg/mL淡黄色澄明溶液。
2.4、给药
灌胃给药组:给药剂量为2.0mg/kg,给药体积为20mL/kg。
3、操作
于给药前及给药后0.25、0.5、1.0、2.0、4.0、6.0、8.0、11.0、24.0小时,由眼眶采血0.1mL,置EDTA-K2抗凝试管中,10000rpm离心1分钟(4℃),1小时内分离血浆,-20℃或-80℃保存待测。采血至离心过程在冰浴条件下操作。给药后2小时进食。
测定不同的化合物给药后C57小鼠血浆中的待测化合物含量:取给药后各时刻的C57小鼠血浆样品20-50μL(因化合物不同而不同),140-450μL含内标(甲苯磺丁脲(100ng/mL),因化合物不同而不同)的乙腈溶液,涡旋混合,并在3700rpm下离心10分钟。取上清液进行LC/MS/MS分析。
4、药代动力学参数结果
表4、本公开化合物在C57小鼠体内的药代动力学参数
结论:本公开化合物在C57小鼠体内血药浓度高,暴露量高,清除率低,具有药代动力学优势。

Claims (27)

  1. 一种通式(X)所示的化合物,或其可药用的盐:
    其中:
    R1选自-P(O)R10R11、-S(O)rR12、-S(O)rN(R13R14)、-N(R15)S(O)rR16和R9
    各个R9相同或不同,且各自独立地选自氘原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、羟基、烯基、炔基、氰基、硝基、NR20R21、C(O)NR20R21、NR20C(O)R24、-亚烷基-NR20R21、-亚烷基-C(O)NR20R21、C(O)R24、C(O)OR24、环烷基、环烷基烷基、杂环基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、环烷基烷基、杂环基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
    或,两个R9及与其相连的碳原子一起形成任选被一个或多个R01所取代的环烷基、杂环基、芳基或杂芳基;
    L1与苯基连接,或L1连接至两个R9形成的环上;
    R4和R5相同或不同,且各自独立地选自氢原子、氘原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、羟基、烯基、炔基、氰基、硝基、NR22R23、C(O)NR22R23、NR22C(O)R24、C(O)R24、C(O)OR24、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
    或,R4、R5及与其相连的碳原子一起形成任选被一个或多个R01所取代的环烷基、杂环基、芳基或杂芳基;
    环A选自环烷基、杂环基、芳基和杂芳基;
    各个RA相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、硝基、NR25R26、C(O)NR25R26、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧 基、烯基、炔基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
    L1和L3相同或不同,且各自独立地选自键、O、S、NRL、S(O)r、C(O)、C(O)NRL、NRLC(O)、S(O)rNRL、NRLC(O)NRL、(CRL1RL2)m、烯基和炔基;所述的烯基任选被一个或多个R01所取代;
    L2为-(CRL3RL4)n1-RL5-(CRL6RL7)n2-;
    RL5选自键、O、S、NRL’、S(O)、S(O)2、C(O)、C(O)NRL’、NRL’C(O)和NRL’C(O)NRL’;
    各个RL1、RL2、RL3、RL4、RL6和RL7相同或不同,且各自独立地选自氢原子、氘原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
    或,RL1、RL2及与其相连的碳原子一起形成环烷基或杂环基,或,RL3、RL4及与其相连的碳原子一起形成环烷基或杂环基,或,RL6、RL7及与其相连的碳原子一起形成环烷基或杂环基;所述的环烷基或杂环基各自独立地任选被一个或多个R01所取代;
    或,RL3、RL4及与其相连的两个相邻碳原子一起形成C=C或C≡C,或RL6、RL7及与其相连的两个相邻碳原子一起形成C=C或C≡C;
    R10、R11、R12、R13、R14、R15和R16相同或不同,且各自独立地选自氢原子、氘原子、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R01所取代;
    各个R20、R21、R22、R23、R24、R25、R26、RL和RL’相同或不同,且各自独立地选自氢原子、氘原子、烷基、卤代烷基、烷氧基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个R01所取代;
    或,R13、R14及与其相连的氮原子一起形成杂环基;或NR15及与其相连的碳原子、R9及与其相连的碳原子一起形成含氮杂环基或含氮杂芳基;或R20、R21及与其相连的氮原子一起形成杂环基;或R22、R23及与其相连的氮原子一起形成杂环基;或R25、R26及与其相连的氮原子一起形成杂环基;所述的杂环基、含氮杂环基或含氮杂芳基各自独立地任选被一个或多个R01所取代;
    Ra和Rb相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、环烷基、和杂环基;
    各个R01相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、 酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
    Z1为N或CRZ1
    Z2为N或CRZ2
    Z3为N或CRZ3
    RZ1、RZ2和RZ3相同或不同,且各自独立地选自氢原子、氘原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、羟基、烯基、炔基、氰基、硝基、C(O)R30、C(O)OR30、NR31R32、C(O)NR31R32、NR31C(O)R30、环烷基、环烷基氧基、杂环基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、环烷基氧基、杂环基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R02所取代;
    各个R30相同或不同,且各自独立地选自氢原子、烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基;所述的烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R02所取代;
    各个R31和R32相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个R02所取代;
    各个R02相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
    J1、J2、J3、J4、J5和J6相同或不同,且各自独立地选自键、O、S、-O-亚烷基-、-亚烷基-O-、C(O)、-C(O)-亚烷基-、-亚烷基-C(O)-、C(O)N(RJ)、N(RJ)C(O)、N(RJ)C(O)N(RJ)、S(O)v、S(O)vN(RJ)、N(RJ)S(O)v、N(RJ)、-N(RJ)-亚烷基-、-亚烷基-N(RJ)-、亚烷基、烯基、炔基、环烷基、杂环基、-亚烷基-杂环基-、-杂环基-亚烷基-、芳基和杂芳基;所述的亚烷基、烯基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个选自R03的取代基所取代;
    各个RJ相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个选自R03的取代基所取代;
    各个R03相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、 酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
    E选自:
    为单键或者双键;
    环D为芳基或杂芳基;
    W选自键、O、S、S(O)r、NR6、C(O)、C(O)NR6、NR6C(O)和(CRY2RY2)s4
    Q1、Q2、Q3、Q4和Q5中的一者为碳原子且与J6连接,其它各自独立地选自N、CH和CRq
    各个Rq相同或不同,且各自独立地选自氘原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、羟基、烯基、炔基、氰基、NRcRd、亚烷基NRcRd、C(O)烷基、C(O)NRcRd、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R04所取代;
    或两个Rq及与其相连的环原子一起形成环烷基或杂环基;
    各个Rc和Rd相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、环烷基和杂环基;或Rc、Rd及与其相连的氮原子一起形成杂环基,所述的杂环基各自独立地任选被一个或多个R04所取代;
    Y2为C(RY2)2或C(O);
    Y1为N或CRY1;Y3为N或CRY3;Y4选自键、CH2或C(O);
    各个RY2相同或不同,且各自独立地选自氢原子、氘原子、烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R04所取代;
    或,两个RY2与其相连的碳原子一起形成环烷基或杂环基;所述的环烷基和杂环基各自独立地任选被一个或多个R04所取代;
    RY1和RY3相同或不同,且各自独立地选自氢原子、氘原子、烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R04所取代;
    R6、R7和R8相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、氘代烷基、烷氧基、卤代烷氧基、氘代烷氧基、环烷基和杂环基;
    各个R04相同或不同,且各自独立地选自氘原子、氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
    n为0、1、2、3、4、5或6;
    r为0、1或2;v为0、1或2;
    y为0、1、2或3;s1为0、1、2、3、4、5或6;
    s2为0、1、2、3或4;s3为0、1、2、3或4;s4为0、1、2、3或4;且,
    n1、n2和m各自独立地为0、1、2、3、4、5、6、7、8、9或10。
  2. 根据权利要求1所述的通式(X)所示的化合物,或其可药用的盐,其为通式(I)所示的化合物,或其可药用的盐:
    其中:
    R1选自-P(O)R10R11、-SO2R12、-SO2N(R13R14)和-N(R15)SO2R16
    R2和R3相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、硝基、NR20R21、C(O)NR20R21、 NR20C(O)R24、-亚烷基-NR20R21、-亚烷基-C(O)NR20R21、C(O)R24、C(O)OR24、环烷基、环烷基烷基、杂环基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、环烷基烷基、杂环基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
    或,R2、R3及与其相连的碳原子一起形成任选被一个或多个R01所取代的环烷基、杂环基、芳基或杂芳基;或,R3与L1连接;
    R4和R5相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、硝基、NR22R23、C(O)NR22R23、NR22C(O)R24、C(O)R24、C(O)OR24、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
    或,R4、R5及与其相连的碳原子一起形成任选被一个或多个R01所取代的环烷基、杂环基、芳基或杂芳基;
    环A选自环烷基、杂环基、芳基和杂芳基;
    各个RA相同或不同,且各自独立地选自氧代基、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、硝基、NR25R26、C(O)NR25R26、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
    L1和L3相同或不同,且各自独立地选自键、O、S、NRL、S(O)r、C(O)、C(O)NRL、NRLC(O)、S(O)rNRL、NRLC(O)NRL、(CRL1RL2)m、烯基和炔基;所述的烯基任选被一个或多个R01所取代;
    L2为-(CRL3RL4)n1-RL5-(CRL6RL7)n2-;
    RL5选自键、O、S、NRL’、S(O)、S(O)2、C(O)、C(O)NRL’、NRL’C(O)和NRL’C(O)NRL’;
    各个RL1、RL2、RL3、RL4、RL6和RL7相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R01所取代;
    或,RL1、RL2及与其相连的碳原子一起形成环烷基或杂环基,或,RL3、RL4及与其相连的碳原子一起形成环烷基或杂环基,或,RL6、RL7及与其相连的碳原子一起形成环烷基或杂环基;所述的环烷基或杂环基各自独立地任选被一个或多个R01所取代;
    或,RL3、RL4及与其相连的两个相邻碳原子一起形成C=C或C≡C,或RL6、RL7及与其相连的两个相邻碳原子一起形成C=C或C≡C;
    R10、R11、R12、R13、R14、R15和R16相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R01所取代;
    各个R20、R21、R22、R23、R24、R25、R26、RL和RL’相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、烷氧基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个R01所取代;
    或,R13、R14及与其相连的氮原子一起形成杂环基;或NR15及与其相连的碳原子、R2及与其相连的碳原子一起形成含氮杂环基或含氮杂芳基;或R20、R21及与其相连的氮原子一起形成杂环基;或R22、R23及与其相连的氮原子一起形成杂环基;或R25、R26及与其相连的氮原子一起形成杂环基;所述的杂环基、含氮杂环基或含氮杂芳基各自独立地任选被一个或多个R01所取代;
    Ra和Rb相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、环烷基、和杂环基;
    各个R01相同或不同,且各自独立地选自氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
    Z1为N或CRZ1
    Z2为N或CRZ2
    Z3为N或CRZ3
    RZ1、RZ2和RZ3相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、硝基、C(O)R30、C(O)OR30、NR31R32、C(O)NR31R32、NR31C(O)R30、环烷基、环烷基氧基、杂环基、杂环基氧基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、环烷基、环烷基氧基、杂环基、杂环基氧基、芳基和杂芳基各自独立地任选被一个或多个R02所取代;
    各个R30相同或不同,且各自独立地选自氢原子、烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基;所述的烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R02所取代;
    各个R31和R32相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个R02所取代;
    各个R02相同或不同,且各自独立地选自氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
    J1、J2、J3、J4、J5和J6相同或不同,且各自独立地选自键、O、S、-O-亚烷基-、-亚烷基-O-、-C(O)-、-C(O)-亚烷基-、-亚烷基-C(O)-、-C(O)N(RJ)-、-N(RJ)C(O)-、-N(RJ)C(O)N(RJ)-、-S(O)v-、-S(O)vN(RJ)-、-N(RJ)S(O)v-、-N(RJ)-、-N(RJ)-亚烷基-、-亚烷基-N(RJ)-、亚烷基、烯基、炔基、环烷基、杂环基、-亚烷基-杂环基-、-杂环基-亚烷基-、芳基和杂芳基;所述的亚烷基、烯基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个选自R03的取代基所取代;
    各个RJ相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、环烷基和杂环基;所述的烷基、烷氧基、环烷基和杂环基各自独立地任选被一个或多个选自R03的取代基所取代;
    各个R03相同或不同,且各自独立地选自氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
    E选自:
    为单键或者双键;
    Q1、Q2、Q3、Q4和Q5中的一者为碳原子且与J6连接,其它各自独立地选自N或CRQ
    各个RQ相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、烯基、炔基、氰基、NRcRd、亚烷基NRcRd、C(O)烷基、 C(O)NRcRd、环烷基、杂环基、芳基和杂芳基;所述的烷基、烷氧基、烯基、炔基、亚烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R04所取代;
    各个Rc和Rd相同或不同,且各自独立地选自氢原子、卤素、烷基、环烷基和杂环基;或Rc、Rd及与其相连的氮原子一起形成杂环基,所述的杂环基各自独立地任选被一个或多个R04所取代;
    Y2为C(RY2)2或C(O);
    Y1为N或CRY1;Y3为N或CRY3
    各个RY2相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R04所取代;
    或,两个RY2与其相连的碳原子一起形成环烷基或杂环基;所述的环烷基和杂环基各自独立地任选被一个或多个R04所取代;
    RY1和RY3相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、环烷基和杂环基;所述的烷基、烷氧基、烯基、炔基、环烷基和杂环基各自独立地任选被一个或多个R04所取代;
    R6、R7和R8相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、环烷基和杂环基;
    各个R04相同或不同,且各自独立地选自氧代基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、羟烷氧基、氰基、烯基、炔基、氨基、-NH烷基、-N(烷基)2、-亚烷基-氨基、-亚烷基-NH烷基、-亚烷基-N(烷基)2、酰胺基、硝基、环烷基、杂环基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基、芳基和杂芳基;
    n为0、1、2、3、4、5或6;
    r为1或2;
    v为1或2;且,
    n1、n2和m各自独立地为0、1、2、3、4、5、6、7、8、9或10。
  3. 根据权利要求1或2所述的通式(X)所示的化合物,或其可药用的盐,其中:环A为5或6元杂芳基;优选为吡唑基。
  4. 根据权利要求1至3任一所述的通式(X)所示的化合物,或其可药用的盐,其中:E选自: s为0、1、2或3,Rq、R6、Y1、R7、Y2和R8如权利要求1中所定义;优选地,E选自:s为0、1、2或3,Y1、Y2、R6、R7、R8和Rq如权利要求1中所定义;更优选地,E选自:Y1为N或CH;Y2为CH2或C(O);各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;R6为氢原子或C1-6烷基;R7为C1-6烷基;s为0、1、2或3。
  5. 根据权利要求1至3任一所述的通式(X)所示的化合物,或其可药用的盐,其为通式(I’-1)、(I’-2)或(I’-3)所示的化合物,或其可药用的盐:

    其中,s为0、1、2或3;n为0、1或2;
    R1至R7、Ra、Rb、RZ1、RZ2、L1、L2、RA、J1至J6、Rq、Y1和Y2如权利要求1中所定义。
  6. 根据权利要求1至5任一所述的通式(X)所示的化合物,或其可药用的盐,其为通式(V-1)所示的化合物,或其可药用的盐:
    其中,环J2’为含氮杂环基;
    q为0、1、2、3、4、5或6;j为0、1、2、3、4、5或6;
    s为0、1、2或3;n为0、1或2;
    R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、J1、R03、J4、J5、J6、Rq和Y2如权利要求1中所定义。
  7. 根据权利要求1至5任一所述的通式(X)所示的化合物,或其可药用的盐,其为通式(V-2)所示的化合物,或其可药用的盐:
    其中,环J3’为含氮杂环基;
    q为0、1、2、3、4、5或6;j为0、1、2、3、4、5或6;
    s为0、1、2或3;n为0、1或2;
    R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、J1、R03、J4、J5、J6、Rq和Y2如权利要求1中所定义。
  8. 根据权利要求1至5任一所述的通式(X)所示的化合物,或其可药用的盐,其中:-J1-J2-J3-J4-J5-J6-选自 X、Y、U、V、S和T相同或不同,且各自独立地选自CH、CR03和N;u、q、w、x、k、l和q1相同或不同,且各自独立地为0、1、2、3、4、5或6;a、b、c、e和f相同或不同,且各自独立地为0、1、2或3;d为1、2或3;g和h相同或不同,且各自独立地为0、1或2;各个RJ3和RJ5相同或不同,且各自独立地为氢原子或R03;R03和RJ如权利要求1中所定义;
    优选地,-J1-J2-J3-J4-J5-J6-选自 X、Y、U、V、S和T相同或不同,且各自独立地为CR05或N;q、u、x、w、k和l相同或不同,且各自独立地为0、1、2、3、4、5或6;a、b、c、e和f相同或不同,且各自独立地为0、1、2或3;d为1、2或3;g和h相同或不同,且各自独立地为0、1或2;各个RJ3相同或不同,且各自独立地为氢原子或C1-6烷基;各个RJ5相同或不同,且各自独立地为氢原子或C1-6烷基;各个R03相同或不同,且各自独立地选自氧代基、卤素、羟基和C1-6烷基;各个R05相同或不同,且各自独立地选自氢原子、卤素、羟基和C1-6烷基。
  9. 根据权利要求1至5任一所述的通式(X)所示的化合物,或其可药用的盐,其中:J1选自键、哌啶基和哌嗪基,所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;J2为任选被一个或多个R03所取代的4至7元单环杂环基或7至14元螺杂环基,R03选自羟基、卤素、C1-6烷基和C1-6卤代烷基;J3为键或C1-6亚烷基;J4为键或C(O);J5为键或C1-6亚烷基;J6为N(RJ)或3至12元杂环基,所述的3至12元杂环基任选被选自卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或甲基。
  10. 根据权利要求1至7任一所述的通式(X)所示的化合物,或其可药用的盐,其中:-J4-J5-J6-选自键、-C(O)-C1-6亚烷基-哌啶基、哌啶基、哌嗪基和-C(O)-C1-6亚烷基-N(RJ)-;所述的哌啶基和哌嗪基各自独立地任选被选自氧代基、卤素、羟基和C1-6烷基中的一个或多个所取代;RJ为氢原子或甲基;优选为键。
  11. 根据权利要求1至10任一所述的通式(X)所示的化合物,或其可药用的盐,其中:-L1-L2-为-O-C1-10亚烷基-;优选地,-L1-L2-为-O-(CH2)n3-;n3为1、2、3、4、5、6、7或8。
  12. 根据权利要求1至11任一所述的通式(X)所示的化合物,或其可药用的盐,其中,R1为-P(O)R10R11或-N(R15)SO2R16;R10、R11、R15和R16相同或不同,且各自独立地选自氢原子、C1-6烷基和3至6元环烷基;优选地,R1为-P(O)R10R11或-N(R15)SO2R16;R10、R11、R15和R16相同或不同,且各自独立地为氢原子或C1-6烷基。
  13. 根据权利要求1至12任一所述的通式(X)所示的化合物,或其可药用的盐,其中,R4为氢原子,且R5为卤素或C1-6卤代烷基;优选地,R4为氢原子,且R5为卤素。
  14. 根据权利要求1、3至13任一所述的通式(X)所示的化合物,或其可药用的盐,其中,RZ1选自氢原子、C1-6烷氧基、C1-6卤代烷氧基、C1-6氘代烷氧基和C1-6羟烷氧基;优选地,RZ1为氢原子或C1-6烷氧基。
  15. 根据权利要求1至14任一所述的通式(X)所示的化合物,或其可药用的盐,其中,RZ2选自氢原子、卤素、C1-6烷基和C1-6卤代烷基;优选地,RZ2为氢原子或卤素。
  16. 根据权利要求4至15任一所述的通式(X)所示的化合物,或其可药用的盐,其中,各个Rq相同或不同,且各自独立地选自卤素、C1-6烷基和C1-6卤代烷基;和/或s为0或1。
  17. 根据权利要求1至16任一所述的通式(X)所示的化合物,或其可药用的盐,其中,
  18. 根据权利要求1至17中任一项所述的通式(X)所示的化合物,或其可药用的盐,其选自以下任一化合物:















  19. 一种通式(Xa)所示的化合物或其盐:
    其中,
    RJ’选自氢原子、羟基、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、C(O)ORaa、-亚烷基-C(O)ORaa、NRbbRcc、-亚烷基-NRbbRcc、-C(O)Raa、-亚烷基-C(O)Raa、S(O)ORaa和S(O)2ORaa
    Raa各自独立地选自氢原子、烷基、卤代烷基、卤代烷氧基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;Rbb和Rcc各自独立地选自氢原子、烷基、环烷基、杂环基和氨基保护基,所述氨基保护基优选为Boc;
    环A、R1、R9、y、R4、R5、RA、Ra、Rb、Z1、Z2、Z3、L1、L2、L3、J1、J2和n如权利要求1中所定义。
  20. 化合物或其盐,其选自以下结构:






  21. 一种制备通式(X’-3)所示的化合物或其可药用的盐的方法,该方法包括:
    通式(X’-2a)所示的化合物或其盐与通式(X’-3b)所示的化合物或其盐(优选盐酸盐)发生还原胺化反应得到通式(X’-3)所示的化合物或其可药用的盐;其中,
    环J3’为含氮杂环基,j为0、1、2、3、4、5或6;
    q为1、2、3、4、5或6;q-1为0、1、2、3、4或5;
    n为0、1或2;s为0、1、2或3;
    R1、R9、y、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、J1、R03、J4、J5、J6、Y1、R7和Rq如权利要求1中所定义。
  22. 一种制备根据权利要求6所述的通式(V-1)所示的化合物或其可药用的盐的方法,该方法包括:
    通式(V-1A)所示的化合物或其盐(优选三氟乙酸盐或盐酸盐)与通式(V-1B)所示的化合物或其盐发生还原胺化反应得到通式(V-1)所示的化合物或其可药用的盐;
    其中,RW为氢原子;
    R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、环J2’、R03、j、q、J4、J5、J6、Rq、Y2和s如权利要求6中所定义。
  23. 一种制备根据权利要求7所述的通式(V-2)所示的化合物或其可药用的盐的方法,该方法包括:
    通式(V-2A)所示的化合物或其盐与通式(V-2B)所示的化合物或其盐(优选盐酸盐)发生还原胺化反应得到通式(V-2)所示的化合物或其可药用的盐;
    其中,R1、R2、R4、R5、Ra、Rb、RZ1、RZ2、L1、L2、RA、n、J1、q、环J3’、R03、j、J4、J5、J6、Rq、Y2和s如权利要求7中所定义。
  24. 一种药物组合物,所述药物组合物含有根据权利要求1至18中任一项所述的通式(X)所示的化合物或其可药用的盐,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
  25. 根据权利要求1至18中任一项所述的通式(X)所示的化合物或其可药用的盐或根据权利要求24所述的药物组合物在制备用于调节EGFR泛素化和降解的药物中的用途。
  26. 根据权利要求1至18中任一项所述的通式(X)所示的化合物或其可药用的盐或根据权利要求24所述的药物组合物在制备用于治疗和/或预防由EGFR介导的或依赖性的疾病或病症的药物中的用途。
  27. 根据权利要求1至18中任一项所述的通式(X)所示的化合物或其可药用的盐或根据权利要求24所述的药物组合物在制备用于治疗和/或预防疾病或病症的药物中的用途;优选地,所述疾病或病症为癌症或肿瘤;优选地,所述的癌症选 自鳞状细胞癌、基底细胞癌、腺癌、肝癌、肾癌、膀胱癌、乳腺癌、宫颈癌、结直肠癌、食管癌、头颈癌、鼻咽癌、口腔癌、唾液腺癌、肾癌、肺癌、卵巢癌、胰腺癌、前列腺癌、胃癌、白血病、淋巴瘤、神经胶质瘤、神经母细胞瘤、黑素瘤、肉瘤、子宫内膜癌、睾丸癌和甲状腺癌;进一步优选为肺癌;更优选为非小细胞肺癌。
PCT/CN2023/129593 2022-11-04 2023-11-03 取代的氨基嘧啶类化合物、其制备方法及其在医药上的应用 WO2024094171A1 (zh)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
CN202211377713.0 2022-11-04
CN202211377713 2022-11-04
CN202310310648.8 2023-03-23
CN202310310648 2023-03-23
CN202310568080.X 2023-05-19
CN202310568080 2023-05-19
CN202310965426 2023-08-02
CN202310965426.X 2023-08-02

Publications (1)

Publication Number Publication Date
WO2024094171A1 true WO2024094171A1 (zh) 2024-05-10

Family

ID=90929810

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/129593 WO2024094171A1 (zh) 2022-11-04 2023-11-03 取代的氨基嘧啶类化合物、其制备方法及其在医药上的应用

Country Status (1)

Country Link
WO (1) WO2024094171A1 (zh)

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004026881A1 (de) * 2002-08-21 2004-04-01 Schering Aktiengesellschaft Makrozyklische pyrimidine, deren herstellung und verwendung als arzneimittel
WO2009112439A1 (en) * 2008-03-10 2009-09-17 Janssen Pharmaceutica Nv 4-aryl-2-anilino-pyrimidines as plk kinase inhibitors
WO2009132202A2 (en) * 2008-04-24 2009-10-29 Incyte Corporation Macrocyclic compounds and their use as kinase inhibitors
WO2012125603A1 (en) * 2011-03-16 2012-09-20 Cephalon, Inc. Macrocyclic compounds as alk, fak and jak2 inhibitors
CN106164075A (zh) * 2014-04-03 2016-11-23 詹森药业有限公司 大环嘧啶衍生物
WO2017060322A2 (en) * 2015-10-10 2017-04-13 Bayer Pharma Aktiengesellschaft Ptefb-inhibitor-adc
WO2021168074A1 (en) * 2020-02-18 2021-08-26 Theseus Pharmaceuticals, Inc. Macrocyclic compounds and uses thereof
CN113527335A (zh) * 2020-04-15 2021-10-22 南京圣和药业股份有限公司 作为egfr抑制剂的大环类化合物及其应用
WO2021238817A1 (zh) * 2020-05-29 2021-12-02 百极弘烨(广东)医药科技有限公司 大环jak抑制剂及其应用
CN114656482A (zh) * 2020-12-23 2022-06-24 南京圣和药业股份有限公司 作为egfr抑制剂的大环杂环类化合物及其应用

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004026881A1 (de) * 2002-08-21 2004-04-01 Schering Aktiengesellschaft Makrozyklische pyrimidine, deren herstellung und verwendung als arzneimittel
CN1675225A (zh) * 2002-08-21 2005-09-28 舍林股份公司 大环嘧啶化合物、其制备方法以及作为药物的应用
WO2009112439A1 (en) * 2008-03-10 2009-09-17 Janssen Pharmaceutica Nv 4-aryl-2-anilino-pyrimidines as plk kinase inhibitors
WO2009132202A2 (en) * 2008-04-24 2009-10-29 Incyte Corporation Macrocyclic compounds and their use as kinase inhibitors
WO2012125603A1 (en) * 2011-03-16 2012-09-20 Cephalon, Inc. Macrocyclic compounds as alk, fak and jak2 inhibitors
CN106164075A (zh) * 2014-04-03 2016-11-23 詹森药业有限公司 大环嘧啶衍生物
WO2017060322A2 (en) * 2015-10-10 2017-04-13 Bayer Pharma Aktiengesellschaft Ptefb-inhibitor-adc
WO2021168074A1 (en) * 2020-02-18 2021-08-26 Theseus Pharmaceuticals, Inc. Macrocyclic compounds and uses thereof
CN113527335A (zh) * 2020-04-15 2021-10-22 南京圣和药业股份有限公司 作为egfr抑制剂的大环类化合物及其应用
WO2021238817A1 (zh) * 2020-05-29 2021-12-02 百极弘烨(广东)医药科技有限公司 大环jak抑制剂及其应用
CN113735856A (zh) * 2020-05-29 2021-12-03 百极弘烨(南通)医药科技有限公司 大环jak抑制剂及其应用
CN114656482A (zh) * 2020-12-23 2022-06-24 南京圣和药业股份有限公司 作为egfr抑制剂的大环杂环类化合物及其应用

Similar Documents

Publication Publication Date Title
TWI751155B (zh) 經胺取代之芳基或雜芳基化合物
CN109219604B (zh) 四氢异喹啉雌激素受体调节剂及其用途
WO2020073949A1 (zh) 含氮杂芳类衍生物调节剂、其制备方法和应用
WO2021031952A1 (zh) 氧代六元环并嘧啶类化合物,其制法与医药上的用途
KR20180132125A (ko) Mdm2 단백질 분해제
WO2023030385A1 (zh) 嘧啶并环类化合物及其制法和用途
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
CN113166103A (zh) Egfr抑制剂及其应用
WO2022268051A1 (zh) 稠合四环类化合物、其制备方法及其在医药上的应用
WO2021190417A1 (zh) 新型氨基嘧啶类egfr抑制剂
CN115956077A (zh) 可做为tlr9抑制剂的经取代杂芳基化合物
CN115867549A (zh) 1H-吡咯并[3,2-c]吡啶和1H-吡咯并[2,3-c]吡啶衍生物作为TLR9抑制剂用于治疗纤维化
JP2023538619A (ja) 線維症の処置のためのtlr9阻害剤としての1h-ベンゾ[d]イミダゾール誘導体
TW202130631A (zh) 3—(5—甲氧基—1—側氧基異吲哚啉—2—基)哌啶—2,6—二酮衍生物及其用途
WO2022247816A1 (zh) 含氮杂环类化合物、其制备方法及其在医药上的应用
WO2022194269A1 (zh) 新型egfr降解剂
WO2022111526A1 (zh) 一种苯环衍生物及其组合物和药学上的应用
WO2022134641A1 (zh) 芳香杂环类化合物、药物组合物及其应用
CN114127080A (zh) 作为激酶抑制剂的杂环化合物、包括该杂环化合物的组合物、及其使用方法
WO2023016518A1 (zh) 一种杂环衍生物及其组合物和药学上的应用
WO2022228543A1 (zh) 桥环类化合物、其制备方法及其在医药上的应用
WO2021139756A1 (zh) 三环四氢异喹啉类衍生物、其制备方法及其在医药上的应用
WO2021136354A1 (zh) 联苯类衍生物抑制剂、其制备方法和应用
CN112771032B (zh) 作为第四代egfr抑制剂的嘧啶吡唑类化合物
WO2023217232A1 (zh) 驱动蛋白kif18a抑制剂及其应用