WO2024017251A1 - 抑制并诱导egfr降解的大环类化合物、其制备方法及其在医药上的应用 - Google Patents

抑制并诱导egfr降解的大环类化合物、其制备方法及其在医药上的应用 Download PDF

Info

Publication number
WO2024017251A1
WO2024017251A1 PCT/CN2023/107927 CN2023107927W WO2024017251A1 WO 2024017251 A1 WO2024017251 A1 WO 2024017251A1 CN 2023107927 W CN2023107927 W CN 2023107927W WO 2024017251 A1 WO2024017251 A1 WO 2024017251A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
general formula
alkyl
heterocyclyl
cycloalkyl
Prior art date
Application number
PCT/CN2023/107927
Other languages
English (en)
French (fr)
Inventor
贾敏强
池江涛
贺峰
陶维康
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 WO2024017251A1 publication Critical patent/WO2024017251A1/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
    • 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
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/08Bridged systems

Definitions

  • the present disclosure belongs to the field of medicine and relates to a macrocyclic compound represented by general formula (I), its preparation method, a pharmaceutical composition containing the compound and its use as a therapeutic agent, especially as an EGFR inhibitor and/or Use of degradation agents and in the preparation of medicaments for the treatment and/or prevention of EGFR-mediated or -dependent diseases or conditions.
  • a macrocyclic compound represented by general formula (I) its preparation method, a pharmaceutical composition containing the compound and its use as a therapeutic agent, especially as an EGFR inhibitor and/or Use of degradation agents and in the preparation of medicaments for the treatment and/or prevention of EGFR-mediated or -dependent diseases or conditions.
  • PROTAC Protein Engineering Targeting Chimera
  • PROTAC Proteolysis Targeting Chimera
  • Its structure contains two different ligands: one is a ubiquitin ligase E3 ligand, and the other is a ligand that binds to the target protein.
  • the two ligands are connected by a connecting arm.
  • PROTAC brings the target protein and the intracellular ubiquitin ligase E3 closer to form a target protein-PROTAC-E3 ternary complex, and then the E3 ubiquitin ligase marks the target protein with a ubiquitin protein tag, and then initiates a powerful intracellular
  • the ubiquitination-proteasome system specifically degrades target proteins, thereby inhibiting the corresponding protein signaling pathways.
  • PROTAC shows 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 and can be recycled and degraded target protein, 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. Therefore, degrading the target protein shows a more efficient and lasting anti-tumor effect than inhibiting its activity. And it is not easy to develop drug resistance due to target protein mutations; 3. PROTAC also has therapeutic potential for targets that are currently considered undruggable, such as transcription factors, scaffolding 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 was lung cancer, with 2.094 million new lung cancer patients. The number of lung cancer cases in China in 2018 was 868,000, with an incidence rate of 0.062%, accounting for 41.4% of the global total.
  • Non-small cell lung cancer is the most common type of lung cancer, with an incidence rate of approximately 85% of the total number of lung cancers.
  • Epidermal growth factor receptor (EGFR) belongs to a family of cell surface receptors with tyrosine kinase activity. EGFR mutation is the most common driver gene of non-small cell lung cancer. About 40% of Chinese non-small cell lung cancer patients have EGFR mutation, while 11-16% of patients in Western countries have EGFR mutation. About 90% of the mutant forms of EGFR are deletion of exon 19 (Del19 mutation) and L858R point mutation of 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 of EGFR tyrosine kinase inhibitors represented by gefitinib and erlotinib, are reversibly binding targeted drugs that compete with ATP to bind to the structural domain of EGFR kinase to inhibit its activation.
  • gefitinib and erlotinib are reversibly binding targeted drugs that compete with ATP to bind to the structural domain of EGFR kinase to inhibit its activation.
  • most patients will develop drug resistance after 10 to 12 months of treatment. About 50% of patients are drug resistant due to the secondary mutation of T790M.
  • the second generation EGFR tyrosine kinase inhibitor represented by afatinib
  • afatinib is Reversible targeted drugs, however, cannot solve the problem of T790M mutation resistance, and the lack of selectivity for wild-type EGFR makes the compounds more toxic.
  • the emergence of the third-generation EGFR tyrosine kinase inhibitor osimertinib has overcome the resistance caused by the EGFR T790M gene mutation, and has weak inhibition of wild-type EGFR and good selectivity. It has achieved great success in clinical practice. However, New resistance will develop after 9-14 months of use, and studies have revealed that 6-26% of patients develop C797X or other EGFR-dependent gene mutations.
  • PROTAC compounds targeting EGFR protein degradation include WO2017185036A1, WO2018119441A1, WO2019121562A1, WO2019149922A1, WO2021121261A1, WO2021127561A1, WO2021039622A1, WO202205518 1A1, WO2022012623A1, WO2022068849A1, etc.
  • R 1 is selected from hydrogen atom, halogen, alkyl group, alkoxy group, hydroxyl group, alkenyl group, alkynyl group, cyano group, -NR 21 R 22 , -alkylene group NR 21 R 22 , -C(O)NR 21 R 22. Cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, alkylene, cycloalkyl Alkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl are each independently optionally substituted by one or more R 01 ;
  • R 2 is selected from -P(O)R 23 R 24 , -SO 2 R 23 , -SO 2 N(R 23 R 24 ), -N(R 23 )SO 2 R 24 , halogen, alkyl, alkoxy and amino;
  • R 1 , R 2 and the carbon atom to which they are connected together form a cycloalkyl, heterocyclyl, aryl or heteroaryl group optionally substituted by one or more R 01 ;
  • R 3 is selected from hydrogen atom, halogen, alkyl group, alkoxy group, hydroxyl group, alkenyl group, alkynyl group, cyano group, -NR 31 R 32 , -alkylene group NR 31 R 32 , -C(O)NR 31 R 32. Cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heteroaryl Each of the cyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl groups is independently optionally substituted by one or more R 01 ;
  • R 4 is selected from hydrogen atom, halogen, alkyl, alkenyl, alkoxy and haloalkyl;
  • R 21 , R 22 , R 23 , R 24 , R 31 and R 32 are the same or different, and each is independently selected from a hydrogen atom, an alkyl group, an alkoxy group, an amino group, a cycloalkyl group, a heterocyclyl group, and a heterocyclyl group.
  • Alkyl, heterocyclyloxy, aryl and heteroaryl; the alkyl, alkoxy, amino, cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl are each independently optionally substituted by one or more R 01 ;
  • Each R 01 is the same or different, and each is independently selected from oxo, halogen, alkyl, alkoxy, haloalkyl, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl , heterocyclyloxy, aryl and heteroaryl;
  • Z 1 is N or CR Z1 ;
  • Z 2 is N or CR Z2 ;
  • Z 3 is N or CR Z3 ;
  • R Z1 , R Z2 and R Z3 are the same or different, and are each independently selected from hydrogen atom, halogen, alkyl group, alkoxy group, hydroxyl group, alkenyl group, alkynyl group, cyano group, -C(O)R c , - NR a R b , -C(O)NR a R b , -alkylene R c , cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl; so
  • the alkyl, alkoxy, alkenyl, alkynyl, alkylene, cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl groups are each independently optional. Replaced by one or more R 02 ;
  • R Z1 , R Z2 and the carbon atom to which they are connected together form a cycloalkyl, heterocyclyl, aryl or heteroaryl group optionally substituted by one or more R 02 ;
  • R a and R b are the same or different, and are each independently selected from a hydrogen atom, a halogen, an alkyl group, a hydroxyl group, an alkenyl group, an alkynyl group, an amino group, a cycloalkyl group, a heterocyclyl group, a heterocyclylalkyl group, an aryl group, and Heteroaryl; the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heterocyclylalkyl, aryl and heteroaryl groups are each independently optionally substituted by one or more R 02 ;
  • R c is selected from hydrogen atom, halogen, alkyl, alkoxy, hydroxyl, alkenyl, alkynyl, cyano, amino, cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aromatic base and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, amino, cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl Each independently optionally substituted by one or more R 02 ;
  • Each R 02 is the same or different, and each is independently selected from oxo, halogen, alkyl, alkoxy, haloalkyl, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl , heterocyclyloxy, aryl and heteroaryl;
  • L 1 is selected from O, S, NRL , C(O), C(O)NR L , NR L C(O), NR L C(O)NR L and (CR L1 R L2 ) m ;
  • L 2 , L 3 , L 4 , L 5 and L 6 are the same or different, and are each independently selected from bonds, O, S, NRL , C(O), C(O)NR L , NR L C(O ), NR L C(O)NR L and (CR L3 R L4 ) n ;
  • R L1 , R L2 , R L3 and R L4 are each independently selected from hydrogen atoms, halogens, alkyl groups, alkoxy groups and haloalkyl groups; or, R L3 , R L4 and the same carbon atom connected to them together form a cycloalkyl group or heterocyclyl;
  • R L is selected from hydrogen atoms, alkyl groups and haloalkyl groups
  • J 1 is selected from cycloalkyl, heterocyclyl, heterocyclyl-heterocyclyl, aryl, heteroaryl, O, C(O), S(O) r , (CR J1 R J2 ) p and N (R J ); the cycloalkyl, heterocyclyl, aryl and heteroaryl groups are each independently optionally substituted by one or more R 03 ;
  • J 2 , J 3 , J 4 and J 5 are the same or different, and are each independently selected from S, O(CR J3 R J4 ) q , (CR J3 R J4 ) q O, C(O)(CR J3 R J4 ) q , (CR J3 R J4 ) q C(O), C(O)N(R J ), N(R J )C(O), N(R J )C(O)N(R J ), S(O) r , S(O) r N(R J ), N(R J )S(O) r , N(R J )S(O) r N(R J ), (CR J3 R J4 ) q , N(R J )(CR J3 R J4 ) q , (CR J3 R J4 ) q N(R J ), alkenylene, alkynylene, cycloalkyl, heterocyclyl, alkylene-heterocycle
  • J 6 is selected from O, S, S(O) r , (CR J5 R J6 ) t , (CR J5 R J6 ) t N(R J ), N(R J )(CR J5 R J6 ) t , C( O)(CR J5 R J6 ) t , (CR J5 R J6 ) t C(O), C(O)N(R J ), N(R J )C(O), N(R J )C(O )N(R J ), alkenylene, alkynylene, cycloalkyl and heterocyclyl; the alkenylene, alkynylene, cycloalkyl and heterocyclyl are each independently optionally substituted by one or more Replaced by R 03 ;
  • R J is selected from hydrogen atoms, alkyl groups and haloalkyl groups
  • R J1 , R J2 , R J3 , R J4 , R J5 and R J6 are each independently selected from hydrogen atom, halogen, alkyl group, alkoxy group, hydroxyl group, cycloalkyl group, heterocyclyl group, heterocyclylalkyl group, Heterocyclyloxy, aryl and heteroaryl; each of the alkyl, alkoxy, cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl Independently optionally substituted by one or more R 03 ;
  • R J1 , R J2 and the same carbon atom connected to them together form a cycloalkyl or heterocyclyl group optionally substituted by one or more R 03 ; or, R J3 , R J4 and the same carbon atom connected to them Together, they form a cycloalkyl or heterocyclyl group optionally substituted by one or more R 03 ; or, R J5 , R J6 and the same carbon atom connected to them together form a cycloalkyl or heterocyclyl group optionally substituted by one or more R 03 Cycloalkyl or heterocyclyl;
  • R 03 is selected from oxo, halogen, alkyl, alkoxy, haloalkyl, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, heterocyclyloxy, aryl and heteroaryl;
  • Ring A 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 N or CR Q ;
  • RQ is the same or different, and each is independently selected from hydrogen atoms, halogen, alkyl, alkoxy, hydroxyl, alkenyl, alkynyl, cyano and -(CH 2 ) t1 NR d R e ; the alkane
  • the base, alkoxy group, alkenyl group and alkynyl group are each independently optionally substituted by one or more R 04 ;
  • R d and R e are each independently selected from hydrogen atoms, halogens, alkyl groups, cycloalkyl groups and heterocyclyl groups;
  • R Z 4 is C(R Z ) 2 or C(O);
  • R Z is the same or different, and each is independently selected from hydrogen atoms, halogens, alkyl groups and amino, or two R Z together with the carbon atoms to which they are attached form a cycloalkyl or heterocyclyl group;
  • Z 8 is N or CR Z8 ;
  • R Z4 and R Z7 are each independently selected from hydrogen atoms, alkyl groups and haloalkyl groups;
  • R Z8 is selected from hydrogen atoms, alkyl groups, alkoxy groups and haloalkyl groups
  • R 04 is selected from oxo, halogen, alkyl, alkoxy, haloalkyl, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, heterocyclyloxy, aryl and heteroaryl;
  • n, p, q, t and t1 are each independently 0, 1, 2, 3, 4, 5 or 6;
  • r is 1 or 2.
  • the purpose of this disclosure is to provide a compound represented by general formula (I), or a pharmaceutically acceptable salt thereof;
  • R 1 is selected from hydrogen atom, halogen, alkyl group, alkoxy group, hydroxyl group, alkenyl group, alkynyl group, cyano group, -NR 21 R 22 , -alkylene group NR 21 R 22 , -C(O)NR 21 R 22. Cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, alkylene, cycloalkyl Alkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl are each independently optionally substituted by one or more R 01 ;
  • R 2 is selected from -P(O)R 23 R 24 , -SO 2 R 23 , -SO 2 N(R 23 R 24 ), -N(R 23 )SO 2 R 24 , halogen, alkyl, alkoxy and amino;
  • R 1 , R 2 and the carbon atom to which they are connected together form a cycloalkyl, heterocyclyl, aryl or heteroaryl group optionally substituted by one or more R 01 ;
  • R 3 is selected from hydrogen atom, halogen, alkyl group, alkoxy group, hydroxyl group, alkenyl group, alkynyl group, cyano group, -NR 31 R 32 , -alkylene group NR 31 R 32 , -C(O)NR 31 R 32. Cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heteroaryl Each of the cyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl groups is independently optionally substituted by one or more R 01 ;
  • R 4 is selected from hydrogen atom, halogen, alkyl, alkenyl, alkoxy and haloalkyl;
  • R 3 , R 4 and the carbon atom to which they are connected together form a cycloalkyl, heterocyclyl, aryl or heteroaryl group optionally substituted by one or more R 01 ;
  • R 21 , R 22 , R 23 , R 24 , R 31 and R 32 are the same or different, and are each independently selected from alkyl, alkoxy, amino, cycloalkyl, heterocyclyl, heterocyclylalkyl, Heterocyclyloxy, aryl and heteroaryl; the alkyl, alkoxy, amino, cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl Each group is independently optionally substituted by one or more R 01 ;
  • Each R 01 is the same or different, and each is independently selected from oxo, halogen, alkyl, alkoxy, halogen.
  • Z 1 is N or CR Z1 ;
  • Z 2 is N or CR Z2 ;
  • Z 3 is N or CR Z3 ;
  • R Z1 , R Z2 and R Z3 are the same or different, and are each independently selected from hydrogen atom, halogen, alkyl group, alkoxy group, hydroxyl group, alkenyl group, alkynyl group, cyano group, -C(O)R c , - NR a R b , -C(O)NR a R b , -alkylene R c , cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl; so
  • the alkyl, alkoxy, alkenyl, alkynyl, alkylene, cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl groups are each independently optional. Replaced by one or more R 02 ;
  • R Z1 , R Z2 and the carbon atom to which they are connected together form a cycloalkyl, heterocyclyl, aryl or heteroaryl group optionally substituted by one or more R 02 ;
  • R a and R b are the same or different, and are each independently selected from a hydrogen atom, a halogen, an alkyl group, a hydroxyl group, an alkenyl group, an alkynyl group, an amino group, a cycloalkyl group, a heterocyclyl group, a heterocyclylalkyl group, an aryl group, and Heteroaryl; the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heterocyclylalkyl, aryl and heteroaryl groups are each independently optionally substituted by one or more R 02 ;
  • R c is selected from hydrogen atom, halogen, alkyl, alkoxy, hydroxyl, alkenyl, alkynyl, cyano, amino, cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aromatic base and heteroaryl; the alkyl, alkoxy, alkenyl, alkynyl, amino, cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl Each independently optionally substituted by one or more R 02 ;
  • Each R 02 is the same or different, and each is independently selected from oxo, halogen, alkyl, alkoxy, haloalkyl, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl , heterocyclyloxy, aryl and heteroaryl;
  • L 1 is selected from O, S, NRL , C(O), C(O)NR L , NR L C(O), NR L C(O)NR L and (CR L1 R L2 ) m ;
  • L 2 , L 3 , L 4 , L 5 and L 6 are the same or different, and are each independently selected from bonds, O, S, NRL , C(O), C(O)NR L , NR L C(O ), NR L C(O)NR L and (CR L3 R L4 ) n ;
  • R L1 , R L2 , R L3 and R L4 are each independently selected from hydrogen atoms, halogens, alkyl groups, alkoxy groups and haloalkyl groups; or, R L3 , R L4 and the same carbon atom connected to them together form a cycloalkyl group or heterocyclyl;
  • R L is selected from hydrogen atoms, alkyl groups and haloalkyl groups
  • J 1 is selected from cycloalkyl, heterocyclyl, heterocyclyl-heterocyclyl, aryl, heteroaryl, O, C(O), S(O) r , (CR J1 R J2 ) p and N( R J ); the cycloalkyl, heterocyclyl, aryl and heteroaryl groups are each independently optionally substituted by one or more R 03 ;
  • J 2 , J 3 , J 4 and J 5 are the same or different, and are each independently selected from S, O(CR J3 R J4 ) q , (CR J3 R J4 ) q O, C(O)(CR J3 R J4 ) q , (CR J3 R J4 ) q C(O), C(O)N(R J ), N(R J )C(O), N(R J )C(O)N(R J ), S(O) r , S(O) r N(R J ), N(R J )S(O) r , N(R J )S(O) r N(R J ), (CR J3 R J4 ) q , N(R J )(CR J3 R J4 ) q , (CR J3 R J4 ) q N(R J ), alkenylene, alkynylene, cycloalkyl, heterocyclyl, alkylene-heterocycle
  • J 6 is selected from O, S, S(O) r , (CR J5 R J6 ) t , (CR J5 R J6 ) t N(R J ), N(R J )(CR J5 R J6 ) t , C( O)(CR J5 R J6 ) t , (CR J5 R J6 ) t C(O), C(O)N(R J ), N(R J )C(O), N(R J )C(O )N(R J ), alkenylene, alkynylene, cycloalkyl and heterocyclyl; the alkenylene, alkynylene, cycloalkyl and heterocyclyl are each independently optionally substituted by one or more Replaced by R 03 ;
  • R J is selected from hydrogen atoms, alkyl groups and haloalkyl groups
  • R J1 , R J2 , R J3 , R J4 , R J5 and R J6 are each independently selected from hydrogen atom, halogen, alkyl group, alkoxy group, hydroxyl group, cycloalkyl group, heterocyclyl group, heterocyclylalkyl group, Heterocyclyloxy, aryl and heteroaryl; each of the alkyl, alkoxy, cycloalkyl, heterocyclyl, heterocyclylalkyl, heterocyclyloxy, aryl and heteroaryl Independently optionally substituted by one or more R 03 ;
  • R J1 , R J2 and the same carbon atom connected to them together form a cycloalkyl or heterocyclyl group optionally substituted by one or more R 03 ; or, R J3 , R J4 and the same carbon atom connected to them Together, they form a cycloalkyl or heterocyclyl group optionally substituted by one or more R 03 ; or, R J5 , R J6 and the same carbon atom connected to them together form a cycloalkyl or heterocyclyl group optionally substituted by one or more R 03 Cycloalkyl or heterocyclyl;
  • R 03 is selected from oxo, halogen, alkyl, alkoxy, haloalkyl, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, heterocyclyloxy, aryl and heteroaryl;
  • Ring A 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 N or CR Q ;
  • RQ is the same or different, and each is independently selected from hydrogen atoms, halogen, alkyl, alkoxy, hydroxyl, alkenyl, alkynyl, cyano and -(CH 2 ) t1 NR d R e ; the alkane
  • the base, alkoxy group, alkenyl group and alkynyl group are each independently optionally substituted by one or more R 04 ;
  • R d and R e are each independently selected from hydrogen atoms, halogens, alkyl groups, cycloalkyl groups and heterocyclyl groups;
  • Z 4 is C(R Z ) 2 or C(O);
  • R Z is the same or different, and each is independently selected from hydrogen atoms, halogens, alkyl groups and amino groups, or two R Z together with the carbon atoms to which they are connected form a ring.
  • Z 8 is N or CR Z8 ;
  • R Z4 and R Z7 are each independently selected from hydrogen atoms, alkyl groups and haloalkyl groups;
  • R Z8 is selected from hydrogen atoms, alkyl groups, alkoxy groups and haloalkyl groups
  • R 04 is selected from oxo, halogen, alkyl, alkoxy, haloalkyl, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, heterocyclyloxy, aryl and heteroaryl;
  • n, p, q, t and t1 are each independently 0, 1, 2, 3, 4, 5 or 6;
  • r is 1 or 2.
  • n5 is 1, 2, 3, 4, 5, 6 or 7;
  • R 1 , R 2 , R 3 , R 4 , R L3 , R L4 , L 5 , L 6 , R Z1 and R Z2 are as defined in general formula (I);
  • n2 is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10
  • n5 is 1, 2, 3, 4, 5, 6 or 7;
  • R L3 and R L4 are each independently selected from a hydrogen atom, a C 1-6 alkyl group and a C 1-6 alkoxy group;
  • R 1 , R 2 , R 3 , R 4 , R Z1 and R Z2 are as defined in general formula (I).
  • n3 and n4 are each independently 0, 1, 2, 3 or 4;
  • L 1 is selected from O, S and CH 2 ;
  • L 3 and L 6 are each independently selected from NH, C(O), C(O)NH, NHC(O), CH 2 and
  • R 1 , R 2 , R 3 , R 4 , R Z1 and R Z2 are as defined by general formula (I);
  • the compound represented by formula (Ia) or a pharmaceutically acceptable salt thereof wherein n3 is 1 or 2 and n4 is 1 or 2.
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein L 1 is O; L 2 , L 4 and L 5 are each independently selected from the group consisting of bonds , CH 2 , CH 2 CH 2 , C(CH 3 ) 2 and L 3 is selected from -C(O)NH-, -NHC(O)- and -CH 2 -; L 6 is selected from CH 2 , C(CH 3 ) 2 and In some implementations In the scheme, L 1 is O; L 2 , L 3 , L 4 , L 5 and L 6 are CH 2 respectively.
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein, -L 1 -L 2 -L 3 -L 4 -L 5 -L 6 - is -O-(CR L3 R L4 ) n2 - or -O-(CR L3 R L4 ) n5 -OC(O)-; in some embodiments, -L 1 -L 2 -L 3 -L 4 -L 5 -L 6 - is -O-(CR L3 R L4 ) n2 -; in some embodiments, -L 1 -L 2 -L 3 -L 4 -L 5 -L 6 - is -O-(CR L3 R L4 ) n5 -OC(O)-; n2 is 2, 3, 4, 5, 6, 7 or 8; n5 is 1, 2, 3, 4, 5, 6 or 7; R L3 and R L4 are independent is selected from hydrogen atom, C 1-6 alky
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein L 1 is O, -L 2 -L 3 -L 4 - is -(CH 2 ) n5 -, n5 is 2, 3 or 4.
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein L 1 is O, -L 2 -L 3 -L 4 - is C 1- 6 alkylene, L 5 is CH 2 or O, L 6 is CH 2 or C(O); in some embodiments, L 1 is O, -L 2 -L 3 -L 4 - is -(CH 2 ) 3 - or -(CH 2 ) 4 -, L 5 is CH 2 or O, L 6 is CH 2 or C(O); in some embodiments, L 1 is O, -L 2 -L 3 -L 4 - is -(CH 2 ) n5 -, n5 is 2, 3, 4, 5 or 6, L 5 is O, and L 6 is C(O).
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein -L 2 -L 3 -L 4 - is a C 1-6 alkylene group; In some embodiments, -L 2 -L 3 -L 4 - is -(CH 2 ) n5 -, n5 is 2, 3, 4, 5, or 6; in some embodiments, -L 2 -L 3 - L 4 - is -(CH 2 ) 3 - or -(CH 2 ) 4 -.
  • the compound represented by general formula (I) or a pharmaceutically acceptable salt thereof is a compound represented by general formula (I') or a pharmaceutically acceptable salt thereof:
  • Ring J 1' is a nitrogen-containing heterocyclic group; the nitrogen-containing heterocyclic group is optionally substituted by one or more R 03 ;
  • n5 is 1, 2, 3, 4, 5, 6 or 7;
  • Ring A, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , J 3 to J 6 and R 03 are as defined in general formula (I).
  • the compound represented by general formula (I) or a pharmaceutically acceptable salt thereof is a compound represented by general formula (IM) or a pharmaceutically acceptable salt thereof:
  • Ring C is a nitrogen-containing heterocyclic group
  • Rings A, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , J 1 , q, J 4 to J 6 and R 03 are as defined in general formula (I).
  • the compound represented by general formula (I) or a pharmaceutically acceptable salt thereof is a compound represented by general formula (IN) or a pharmaceutically acceptable salt thereof:
  • Ring B is a nitrogen-containing heterocyclic group
  • Ring A, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , J 1 , J 2 , q, J 4 to J 6 and R 03 are as in the general formula (I) defined.
  • Q 1 , Q 2 , Q 3 , Q 4 and Q 5 are each independently N or CR Q ;
  • R Q is selected from hydrogen atom, halogen, C 1-6 alkyl group, C 1-6 alkoxy group, hydroxyl group, C 2-6 alkenyl, C 2-6 alkynyl, cyano group, amino, NH C 1-6 alkyl and N(C 1-6 alkyl) 2 ; the C 1-6 alkyl, C 1- 6 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl and amino are each independently optionally substituted by one or more R 04 ;
  • R 04 is selected from oxo, halogen, C 1-6 alkyl Base, C 1-6 alkoxy group, C 1-6 haloalkyl group, hydroxyl group, C 1-6 hydroxyalkyl group, cyano group, amino group, nitro group, 3 to 8 membered cycloalkyl group, 4 to 12 membered heterocyclic group , 4 to
  • Ring A is selected from:
  • Z4 is CH2 or C(O); RZ4 is selected from hydrogen atom, C1-6 alkyl group and C1-6 haloalkyl group; RZ7 is hydrogen atom or C1-6 alkyl group; RQ are each independently Selected from hydrogen atom, halogen, C 1-6 alkyl group, C 1-6 haloalkyl group, C 1-6 alkoxy group, C 1-6 haloalkoxy group, cyano group and amino group.
  • Ring A is selected from: In some embodiments, Ring A is
  • Z 8 is CH or nitrogen atom;
  • Z 4 is CH 2 or C(O);
  • R Z4 is selected from hydrogen atom, C 1-6 alkyl group and C 1-6 haloalkyl group;
  • R Z7 is hydrogen atom or C 1-6 Alkyl group;
  • R Q is selected from hydrogen atom, halogen, C 1-6 alkyl group, C 1-6 haloalkyl group, C 1-6 alkoxy group, C 1-6 haloalkoxy group, cyano group and amino group;
  • Ring A is selected from: RQ is each independently selected from a hydrogen atom, halogen, C 1-6 alkyl group, C 1-6 haloalkyl group, C 1-6 haloalkoxy group and cyano group;
  • Ring A is selected from: RQ is each independently selected from hydrogen atom, halogen, C 1-6 alkyl group, C 1-6 haloalkyl group and C 1-6 haloalkoxy group;
  • Ring A is selected from: Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and R Z4 are as defined by general formula (I);
  • Ring A is selected from:
  • Ring A is selected from: Z 8 is CH or nitrogen atom; Z 4 is CH 2 or C(O); R Z7 is hydrogen atom or C 1-6 alkyl; R Z4 is C 1-6 alkyl; R Q is halogen or C 1- 6 alkyl; k is 0, 1, 2, 3 or 4; j is 0, 1, 2 or 3; s is 0, 1, 2 or 3;
  • Ring A is selected from:
  • Ring A is selected from:
  • Ring A is selected from:
  • Ring A is selected from: In some embodiments, Ring A is In some embodiments, Ring A is selected from:
  • the compound represented by general formula (I) or a pharmaceutically acceptable salt thereof is of general formula (II-1), (II-2) or (II-3) The compound shown, or a pharmaceutically acceptable salt thereof:
  • n2 is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10;
  • Ring J 1 ' is a 5- to 14-membered nitrogen-containing heterocyclic group
  • Ring J 3' is a 5- to 14-membered nitrogen-containing heterocyclic group, and the 5- to 14-membered nitrogen-containing heterocyclic group is optionally substituted by one or more members selected from halogen, hydroxyl, C 1-6 alkyl and C 1 Substituted with -6 haloalkyl substituents;
  • R J is a hydrogen atom or C 1-6 alkyl group; s is 0, 1 or 2;
  • R 1 , R 2 , R 3 , R 4 , R Z1 , R Z2 , R L3 , R L4 , J 1 , q, J 3 , J 4 , J 5 , J 6 , Z 4 , R Z4 , Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and R Q are as defined by general formula (I).
  • the compound represented by general formula (I), (I') or a pharmaceutically acceptable salt thereof is of general formula (I'-1) or (I'-2 ), or a pharmaceutically acceptable salt thereof:
  • Ring J 1' is a nitrogen-containing heterocyclic group; the nitrogen-containing heterocyclic group is optionally substituted by one or more R 03 ;
  • n5 is 1, 2, 3, 4, 5, 6 or 7; j is 0, 1 or 2; k is 0, 1 or 2;
  • R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , J 3 to J 6 , R Q , R Z4 , R Z7 , Z 8 and R 03 are as in the general formula (I) defined in.
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof is a compound represented by the general formula (II-3'), or a pharmaceutically acceptable salt thereof Salt:
  • Ring C is a nitrogen-containing heterocyclic group
  • R 1 , R 2 , R 3 , R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , J 1 , J 2 , R 03 , R J5 , R J6 , t, R J , R Q and Z 4 are as defined by general formula (I).
  • the compound represented by general formula (I) or (IM) or a pharmaceutically acceptable salt thereof is a compound represented by general formula (III-1), or a pharmaceutically acceptable salt thereof.
  • Medicinal salt :
  • Ring C is a nitrogen-containing heterocyclic group
  • n5 is 1, 2, 3, 4, 5, 6 or 7;
  • R 1 , R 2 , R 3 , R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , J 1 , R 03 , q, J 4 , J 5 , J 6 , R Q and Z 4 is as defined by general formula (I).
  • the compound represented by general formula (I) or (IN) or a pharmaceutically acceptable salt thereof is a compound represented by general formula (III-2), or a pharmaceutically acceptable salt thereof Medicinal salt:
  • Ring B is a nitrogen-containing heterocyclic group
  • n5 is 1, 2, 3, 4, 5, 6 or 7;
  • R 1 , R 2 , R 3 , R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , J 1 , J 2 , q, R 03 , J 4 , J 5 , J 6 , R Q and Z 4 are as defined by general formula (I).
  • Ring group in some embodiments, Ring B is selected from piperidinyl, piperazinyl, and diazaspiro[5.5]undecyl.
  • One of Q 3 and Q 4 is a carbon atom and is connected to J 6 , and the remaining two and Q 1 and Q 5 are the same or different, and each is independently N or CR Q ; in some embodiments, One of Q 2 , Q 3 , and Q 4 is a carbon atom and is connected to J 6 , and the remaining two and Q 1 and Q 5 are CR Q ; in some embodiments, Q 2 is a carbon atom and is connected to J 6 connection, Q 1 , Q 3 , Q 4 and Q 5 are all CR Q ; in the above scheme, R Q is as defined in the general formula (I);
  • 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; in some embodiments, Q 2 is J 6 is connected, Q 1 , Q 3 , Q 4 and Q 5 are all CH.
  • the general formula (I), (IM), (IN), (II-3), (II-3'), (III-1), (III-2) The compound shown or a pharmaceutically acceptable salt thereof, wherein J1 is a 5- to 7-membered monoheterocyclyl or a 7- to 14-membered spiroheterocyclyl optionally substituted by 1 or more R 03 ;
  • R 03 is selected from Oxo group , halogen, C 1-6 alkyl group, C 1-6 alkoxy group, C 1-6 haloalkyl group, hydroxyl group, C 1-6 hydroxyalkyl group, cyano group, amino group, nitro group, 3 to 8 yuan Cycloalkyl, 5 to 14 membered heterocyclyl, 5 to 14 membered heterocyclyloxy, 6 to 10 membered aryl and 5 to 14 membered heteroaryl;
  • J1 is a 5- to 7-membered monoheterocyclyl or a 7- to 14-membered spiroheterocyclyl optionally substituted by 1 or more R03 ;
  • R03 is selected from oxo, halogen, C 1-6 alkyl, C 1-6 haloalkyl and C 1-6 alkoxy;
  • J 1 is a 5- to 7-membered monoheterocyclyl optionally substituted by 1 or more R 03
  • R 03 is selected from oxo, halogen, C 1-6 alkyl, C 1-6 haloalkyl and C 1-6 alkoxy; in some embodiments, J 1 is a 5- to 7-membered monoheterocyclyl;
  • J 1 is a 7- to 14-membered spiroheterocyclyl optionally substituted by 1 or more R 03 ;
  • R 03 is selected from oxo, halogen, C 1-6 alkyl, C 1- 6 haloalkyl and C 1-6 alkoxy; in some embodiments, J 1 is 7 to 14 membered spiroheterocyclyl;
  • J 1 is a bond or a 3- to 12-membered heterocyclyl group; the 3- to 12-membered heterocyclyl group is optionally selected from one or more of halogen, hydroxyl, and C 1-6 alkyl. Substituted by substituents;
  • J1 is selected from: Preferably The * end is connected to J; in some embodiments, J is selected from the group consisting of bond, piperidinyl, and piperazinyl.
  • the compound represented by general formula (I) or a pharmaceutically acceptable salt thereof wherein J 2 , J 3 , J 4 and J 5 are the same or different, and are each independently selected from the group consisting of bonds, O, (CH 2 ) 1-6 , O(CH 2 ) 1-6 , (CH 2 ) 1-6 O, C(O), C(O)(CH 2 ) 1-6 , (CH 2 ) 1-6 C(O), N(R J ), N(R J )(CH 2 ) 1-6 , (CH 2 ) 1-6 N(R J ), 5 to 14-membered heterocyclyl, (CH 2 ) 1 -6 -5 to 7-membered heterocyclyl and 5 to 7-membered heterocyclyl -(CH 2 ) 1-6 ; the 5 to 14-membered heterocyclyl is optionally substituted by 1 or more R 03 ; R J is a hydrogen atom or C 1-6 alkyl group; each R 03 is the
  • J is selected from bond, O, C(O), NH, N(CH 3 ), CH 2 , C(CH 3 ) 2 , CH 2 CH 2 , CH 2 CH 2 CH 2 , CH 2 CH 2 CH 2 CH 2 , OCH 2 , CH 2 O , OCH 2 CH 2 , CH 2 CH 2 O, C(O)CH 2 , CH 2 C(O), C(O)CH 2 CH 2 , CH 2 CH 2 C(O), NHCH 2 , CH 2 NH , NHCH 2 CH 2 , CH 2 CH 2 NH, C(O)NHCH 2 , CH 2 C(O)NH, C(O)NHCH 2 CH 2 , CH 2 CH 2 C(O)NH, 1,4- Piperidylene-, (CH 2 ) 1-2 -1,4-piperidinylene, 1,4-piperidinylene-(CH 2 ) 1-2 , 1,4-piperazinylene, ( CH 2 ) 1-2 -1,4-piperazinylene, ( CH 2
  • J 2 is selected from bond, C(O), 3 to 12 membered heterocyclyl, and (CH 2 ) p , p is 0, 1, 2, 3, 4, 5, or 6; 3 to 12 membered heterocyclyl is optionally substituted with one or more selected from halogen, hydroxyl, and C 1-6 alkyl; in some embodiments, J 2 is selected from bond, C(O), CH 2 and a 3 to 10-membered heterocyclyl group, the 3 to 10-membered heterocyclyl group is optionally selected from one or more of halogen, hydroxyl and C 1-6 alkyl.
  • J2 is selected from bond, C(O), CH2 , piperidinyl, piperazinyl, and The piperidinyl and piperazinyl groups are each independently optionally substituted with one or more selected from halogen, hydroxyl and C 1-6 alkyl; in some embodiments, J 2 is selected from bond, C (O), CH2 , and 3 to 12 membered heterocyclyl; in some embodiments, J2 is selected from bond, C(O), CH2 , piperidinyl, piperazinyl,
  • the compound represented by the general formula (I'-2) or a pharmaceutically acceptable salt thereof wherein k is 0, 1 or 2; in some embodiments, k is 0 or 1; In some embodiments, k is 0.
  • the general formula (I), (II-1), (II-2), (I'), (I'-1), (I'-2) is represented by The compound or its pharmaceutically acceptable salt, wherein J 3 is selected from bond, O, (CH 2 ) 1-6 , C(O), N(R J ), 5 to 14-membered heterocyclyl, (CH 2 ) 1-6 - 5 to 7-membered heterocyclyl and 5 to 7-membered heterocyclyl -(CH 2 ) 1-6 , R J is a hydrogen atom or a C 1-6 alkyl group; each of the 5 to 14-membered heterocyclic groups is independently optionally substituted by one or more R 03 , and R 03 is selected from From oxo, halogen, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 haloalkyl and hydroxyl;
  • J is selected from bond, O, C(O), NH, N(CH 3 ), CH 2 , C(CH 3 ) 2 , CH 2 CH 2 , CH 2 CH 2 CH 2 , CH 2 CH 2 CH 2 CH 2 , OCH 2 , CH 2 O , OCH 2 CH 2 , CH 2 CH 2 O, C(O)CH 2 , CH 2 C(O), C(O)CH 2 CH 2 , CH 2 CH 2 C(O), NHCH 2 , CH 2 NH , NHCH 2 CH 2 , CH 2 CH 2 NH, C(O)NHCH 2 , CH 2 C(O)NH, C(O)NHCH 2 CH 2 , CH 2 CH 2 C(O)NH, 1,4- Piperidylene-, (CH 2 ) 1-2 -1,4-piperidinylene, 1,4-piperidinylene-(CH 2 ) 1-2 , 1,4-piperazinylene, ( CH 2 ) 1-2 -1,4-piperazinylene, ( CH 2
  • J3 is selected from ( CH2 ) q , 3 to 10 membered cycloalkyl, and 3 to 10 membered heterocyclyl, q is 0, 1, 2, 3, 4, 5, or 6; in some In the embodiment, J 3 is selected from CH 2 , piperidinyl and piperazinyl; each of the piperidinyl and piperazinyl groups is independently selected from one of halogen, hydroxyl and C 1-6 alkyl. or more substituted; in some embodiments, J 3 is selected from CH 2 , CH 2 CH 2 , piperidinyl, and piperazinyl; in some embodiments, J 3 is selected from CH 2 , piperidinyl, and piperazinyl. Azinyl.
  • J4 is selected from bond, O, C(O), NH, N(CH 3 ), CH 2 , C(CH 3 ) 2 , CH 2 CH 2 , CH 2 CH 2 CH 2 , CH 2 CH 2 CH 2 CH 2 , OCH 2 , CH 2 O , OCH 2 CH 2 , CH 2 CH 2 O, C(O)CH 2 , CH 2 C(O), C(O)CH 2 CH 2 , CH 2 CH 2 C(O), NHCH 2 , CH 2 NH , NHCH 2 CH 2 , CH 2 CH 2 NH, C(O)NHCH 2 , CH 2 C(O)NH, C(O)NHCH 2 CH 2 , CH 2 CH 2 C(O)NH, 1,4- Piperidylene-, (CH 2 ) 1-2 -1,4-piperidinylene, 1,4-piperidinylene-(CH 2 ) 1-2 , 1,4-piperazinylene, ( CH 2 ) 1-2 -1,4-piperazinylene, ( CH 2
  • J5 is selected from bond, O, C(O), NH, N(CH 3 ), CH 2 , C(CH 3 ) 2 , CH 2 CH 2 , CH 2 CH 2 CH 2 , CH 2 CH 2 CH 2 CH 2 , OCH 2 , CH 2 O , OCH 2 CH 2 , CH 2 CH 2 O, C(O)CH 2 , CH 2 C(O), C(O)CH 2 CH 2 , CH 2 CH 2 C(O), NHCH 2 , CH 2 NH, NHCH 2 CH 2 , CH 2 CH 2 NH, C(O)NHCH 2 , CH 2 C(O)NH, C(O)NHCH 2 CH 2 , CH 2 CH 2 C(O)NH, 1,4-piperidinylidene-, (CH 2 ) 1- 2-1,4 -piperidinylene, 1,4-piperidinylene-(CH 2 ) 1-2 , 1,4-piperazinylene, (CH 2 ) 1-2 -1,4-yliden
  • the general formula (I), (II-1), (II-2), (I'), (I'-1), (I'-2) is represented by The compound or a pharmaceutically acceptable salt thereof, wherein -J 3 -J 4 -J 5 - is -C 1-6 alkylene- or -4 to 7-membered heterocyclyl -C(O)-; preferably - CH 2 -or In some embodiments, -J 3 -J 4 -J 5 - is -C 1-6 alkylene-; preferably -CH 2 -.
  • the 8-membered cycloalkyl group and the 5- to 14-membered heterocyclyl group are each independently optionally substituted by one or more R 03 , R J is a hydrogen atom or a C 1-6 alkyl group; each R 03 is
  • J 6 is a 5- to 7-membered heterocyclyl group or a C 1-6 alkylene group N(R J ), and the 5- to 7-membered heterocyclyl group is optionally substituted by 1 or more members selected from halogen , hydroxyl, C 1-6 alkoxy, C 1-6 alkyl, C 1-6 haloalkyl and C 1-6 haloalkoxy substituents; R J is a hydrogen atom or C 1-6 alkyl ; In some embodiments, J 6 is selected from In some embodiments, J6 is selected from
  • J6 is selected from bond, O, 3 to 12 membered heterocyclyl, and C 1-6 alkylene N( RJ ), the 3 to 12 membered heterocyclyl optionally selected from Substituted with one or more substituents of halogen, hydroxyl, and C 1-6 alkyl;
  • R is a hydrogen atom or C 1-6 alkyl;
  • J is selected from bond, piperidinyl, Piperazinyl, ethylamino and diazaspiro[5.5]undecyl, the piperidinyl and piperazinyl are each independently selected from one of halogen, hydroxyl and C 1-6 alkyl or replaced by more than one;
  • J6 is selected from bond, 3 to 12 membered heterocyclyl optionally selected from halogen and C 1-6 alkylene N( RJ ) Substituted by one or more substituents in the hydroxyl group;
  • R J is a hydrogen atom or C 1-6 alkyl group;
  • J is selected from bond, piperidinyl,
  • J is selected from bond, piperidinyl,
  • J6 is selected from the group consisting of bond, piperidinyl and piperazinyl; each of the piperidinyl and piperazinyl groups is independently optionally substituted with one or more selected from halogen and hydroxyl; In some embodiments, J6 is selected from 5 to 7-membered heterocyclyl optionally substituted by 1 or more R03 ; each R03 is the same or different, and each is independently selected from halogen, hydroxyl, C1- 6 alkyl and C 1-6 haloalkyl; in some embodiments, J 6 is selected from a 6-membered nitrogen-containing heterocyclyl optionally substituted by 1 or more halogens; more preferably, it is selected from
  • each R 03 is the same or different , and each is independently selected from oxo, halogen, C 1-6 alkyl, hydroxyl, and cyclopropyl; in some embodiments, each R 03 is the same or different, and each is independently selected from hydroxyl, halogen, and C 1-6 alkyl; in some embodiments, each R 03 is the same or different, and each is independently hydroxyl or halogen; in some embodiments, R 03 is halogen; in some embodiments, R 03 is F; In some embodiments, each R 03 is the same or different, and each is independently selected from F, methyl, and hydroxyl.
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is -4 to 7-membered heterocyclyl-C(O)-C 1-6 alkylene-4 to 7-membered heterocyclyl-or -4 to 7-membered heterocyclyl-C 1-6 alkylene-4 to 7-membered heterocyclyl-C(O)-C 1-6 alkylene N(R J )-, each of the 4 to 7-membered heterocyclyl groups is independently optionally substituted by one or more oxo groups.
  • R J is a hydrogen atom or C 1-6 Alkyl; preferably selected from Most preferably, -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 Connect from left to right.
  • the compound represented by the general formula (II-1), (II-2), (I'), (I'-1), (I'-2) or its Medicinal salts of which It is a 4 to 7-membered nitrogen-containing heterocyclyl group-C(O)-C 1-6 alkylene group-4 to 7-membered heterocyclyl group, and the 4- to 7-membered nitrogen-containing heterocyclyl group and the 4- to 7-membered heterocyclic group
  • Each ring group is independently optionally substituted by one or more oxo groups, halogen, hydroxyl, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 haloalkyl and C 1-6 haloalkoxy. Substituted with substituents of the group; preferably selected from Connect from left to right.
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is X, Y, U, and V are the same or different, and are each independently CR 05 or N; q, u, and w 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 or 2; g is 1, 2, 3, 4 or 5; each R 05 is the same or different , and each is independently selected from hydrogen atoms, halogens, hydroxyl and alkyl groups; J 6 and R 03 are as defined in general formula (I);
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from -heterocyclyl-C(O)-alkylene-heterocyclyl-, -heterocyclyl -C 0-6 alkylene-heterocyclyl-C(O)-alkylene-N(R J )-, -heterocyclyl-heterocyclyl-alkylene-heterocyclyl-, -heterocycle -alkylene-heterocyclyl-, -heterocyclyl-alkylene-heterocyclyl-heterocyclyl-, and -heterocyclyl-alkylene-C(O)-heterocyclyl-, as described
  • the heterocyclic groups are each independently optionally substituted by one or more R 03 , R J is a hydrogen atom or a C 1-6 alkyl group; R 03 is as defined in general formula (
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from -3 to 10 membered heterocyclyl-C(O)-C 1-6 alkylene-3 to 10-membered heterocyclyl-, -3 to 10-membered heterocyclyl-C 0-6 alkylene -3 to 10-membered heterocyclyl-C(O)-C 1-6 alkylene -N(R J )-, -3 to 10-membered heterocyclyl-3 to 10-membered heterocyclyl-C 1-6 alkylene-3 to 10-membered heterocyclyl-, -3 to 10-membered heterocyclyl -C 1-6 alkylene-3 to 12-membered heterocyclyl-and-3 to 10-membered heterocyclyl-C 1-6 alkylene-3 to 10-membered heterocyclyl-3 to 10-membered heterocyclyl -, the 3 to 10-membered heterocyclyl and 3 to 12-membered heterocyclyl are
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from: Connect from left to right;
  • X, Y, U, V, S, and T are the same or different, and are each independently CR 05 or N; q, q1, q2, u, v, w, and t 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 or 2; each R J3 is the same or different, And each is independently a hydrogen atom or a C 1-6 alkyl group; each R J5 is the same or different, and each is independently a hydrogen atom or a C 1-6 alkyl group; each R 03 is the same or different, and each is independently selected From halogen, hydroxyl and C 1-6 alkyl; each R 05 is the same or different, and each is independently selected from hydrogen atom, halogen, hydroxyl and C 1-6 alkyl; provided that q and q1 are different at the same time and are 0;
  • -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; q, u, v, w and t 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 each is independently 0, 1, 2 or 3; d is 1 or 2; each R J3 is the same or different, and each is independently is a hydrogen atom or a C 1-6 alkyl group; each R J5 is the same or different, and each is independently a hydrogen atom or a C 1-6 alkyl group; each R 03 is the same or different, and each is independently selected from halogen, hydroxyl and C 1-6 alkyl; each R 05 is the same or different, and each is independently selected from hydrogen atoms, halogens, hydroxyl
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from: Connected from left to right; each q is the same or different, and each is independently 0, 1, 2, 3, 4, 5 or 6; each u is the same or different, and each is independently 0, 1, 2, 3, 4, 5 or 6; each q1 is the same or different, and each is independently 1, 2, 3, 4, 5 or 6; each R 03 is the same or different, and each is independently selected from halogen, hydroxyl and C 1-6 alkyl;
  • -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 - is selected from: Connect from left to right;
  • -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: Connect from left to right.
  • X, Y, U, V, S and T are the same or different and are each independently CR 05 or N; q, u, v and w are the same or different and are each independently 0, 1, 2, 3, 4 , 5 or 6; t is 0, 1, 2, 3, 4, 5 or 6; a, b, c, e and f are the same or different and each independently 0, 1, 2 or 3; d is 1 Or 2; each R J3 is a hydrogen atom; each R 03 is the same or different, and each is independently selected from halogen, hydroxyl and C 1-6 alkyl; each R 05 is the same or different, and each is independently selected from a hydrogen atom, Halogen, hydroxyl and C 1-6 alkyl; each R J5 is the same or different, and each is independently a hydrogen atom or C 1-6 alkyl;
  • the compound represented by the general formula (II-3') or a pharmaceutically acceptable salt thereof, in for X, Y and U are the same or different and are each independently CR 05 or N; q and t are the same or different and are each independently 0, 1, 2, 3, 4, 5 or 6; each R 05 is the same or Different, and each is independently selected from hydrogen atoms, halogens, hydroxyl and C 1-6 alkyl groups; R J3 and R J5 are as defined in general formula (I).
  • the compound represented by the general formula (I), (IM), (IN) or a pharmaceutically acceptable salt thereof wherein J 1 is selected from the group consisting of bond, piperidinyl and piperazine base; and/or J 2 is selected from bonds, C(O), CH 2 and 3 to 12-membered heterocyclyl groups, and the 3 to 12-membered heterocyclyl groups are optionally selected from halogen, hydroxyl and C 1-6 Substituted with one or more of the alkyl groups; and/or J 3 is selected from CH 2 , CH 2 CH 2 , piperidinyl and piperazinyl; and/or J 4 is a bond or C(O); and/or J 5 is a bond; and/or J 6 is selected from the group consisting of bonds, 3 to 12-membered heterocyclyl and C 1-6 alkylene N(R J ), and the 3 to 12-membered heterocyclyl is optionally selected from Substituted by one or more substituents among
  • J is selected from bond, piperidinyl and piperazinyl; and/or J is selected from bond, C(O), CH and 3 to 10 membered heterocyclyl, said 3 to 10 membered heterocyclyl
  • the 10-membered heterocyclic group is optionally substituted with one or more selected from halogen, hydroxyl and C 1-6 alkyl; and/or J 3 is selected from CH 2 , piperidinyl and piperazinyl; and/or J 4 is a bond or C(O); and/or J 5 is a bond; and/or J 6 is selected from the group consisting of bonds, 3 to 12-membered heterocyclyl and C 1-6 alkylene N(R J ),
  • the 3 to 12-membered heterocyclic group is optionally substituted with one or more substituents selected from halogen, hydroxyl and C 1-6 alkyl; R J is a hydrogen atom or C 1-6 alkyl.
  • u is 0, 1, or 2; and/or v is 0, 1, or 2; and/or w is 0, 1, or 2.
  • a, b, e, and f are the same or different, and each independently is 0 or 1; and/or c is 1 or 2; and/or g is 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.
  • X, Y, U, V, S, and T are not N at the same time.
  • each R 05 is the same or different, and each is independently a hydrogen atom or a hydroxyl group.
  • J 2 , J 3 , J 4 and J 5 are the same or different, and are each independently selected from bonds, O, C(O), NH, N(CH 3 ), CH 2 , C(CH 3 ) 2 , CH 2 CH 2 , CH 2 CH 2 CH 2 , CH 2 CH 2 CH 2 , OCH 2 , CH 2 O , OCH 2 CH 2 , CH 2 CH 2 O, C(O)CH 2 , CH 2 C(O), C(O)CH 2 CH 2 , CH 2 CH 2 C(O), NHCH 2 , CH 2 NH , NHCH 2 CH 2 , CH 2 CH 2 NH, C(O)NHCH 2 , CH 2 C(O)NH, C(O)NHCH 2 CH 2 , CH 2 CH 2 C(O)NH, 1,4- Piperidylene, (CH 2 ) 1-2 -1,4-piperidinylene, 1,4-piperidinylene-(CH 2 ) 1-2 , 1,4-piperazinylene, (CH 2
  • R 1 is selected from hydrogen atom, halogen and C 1-6 alkyl; preferably, R 1 is a hydrogen atom; in some embodiments, R 1 is C 1-6 alkyl; in some embodiments, R 1 is methyl; in some embodiments, R 1 is 3 to 8 membered cycloalkyl; in some embodiments, R 1 is cyclopropyl.
  • R 2 is -N( R 23 )SO 2 R 24 or -P(O)R 23 R 24 ;
  • R 23 and R 24 are each independently a hydrogen atom or a C 1-6 alkyl group; in some embodiments, R 2 is -N(R 23 )SO 2 R 24 , R 23 and R 24 are the same or different, and each is independently a hydrogen atom or a C 1-6 alkyl group; in some embodiments, R 2 is -N(R 23 )SO 2 R 24 , R 23 and R 24 are methyl respectively;
  • R 2 is -N(R 23 )SO 2 R 24 , R 23 and R 24 are as defined in Formula (I); in some embodiments, R 2 is -N(R 23 ) SO 2 R 24 , R 23 and R 24 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; in some embodiments, R 2 is -N( R 23 )SO 2 R 24 , R 23 and R 24 are the same or different, and each is independently methyl and cyclopropyl;
  • R 2 is -N(R 23 )SO 2 R 24 or -P(O)R 23 R 24 ;
  • R 23 and R 24 are the same or different, and each is independently selected from a hydrogen atom, C 1 -6 alkyl and 3 to 6 membered cycloalkyl; in some embodiments, R 2 is -N(R 23 )SO 2 R 24 or -P(O)R 23 R 24 ;
  • R 23 and R 24 are the same or Different, and each independently selected from hydrogen atoms, methyl and cyclopropyl groups;
  • R 2 is -P(O)R 23 R 24 , R 23 and R 24 are the same or different, and each independently is a hydrogen atom or a C 1-6 alkyl group; in some embodiments, R 2 is -P(O)R 23 R 24 , and R 23 and R 24 are respectively methyl.
  • R 3 is selected from halogen, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 haloalkyl or C 2-6 alkenyl; in some embodiments, R 3 is halogen or C 2-6 alkenyl; in some embodiments, R3 is bromine or vinyl; in some embodiments, R3 is halogen; in some embodiments, R3 is chlorine or bromine; in some embodiments, R3 is bromine.
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein Z 1 is CR Z1 ; R Z1 is selected from hydrogen atoms, C 1-6 alkoxy groups , C 1-6 alkyl and C 1-6 haloalkoxy; in some embodiments, Z 1 is CR Z1 and R Z1 is a hydrogen atom;
  • Z1 is CRZ1 ; RZ1 is selected from hydrogen atom, halogen, C 1-6 alkoxy, C 1-6 alkyl, C 1-6 haloalkoxy and 3 to 6 membered cycloalkyl group; in some embodiments, Z 1 is CR Z1 , R Z1 is a hydrogen atom or halogen; in some embodiments, Z 1 is CH or CF; in some embodiments, Z 1 is N.
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein Z 2 is CR Z2 ; R Z2 is selected from hydrogen atom, C 1-6 alkoxy group, C 1 -6 alkyl and C 1-6 haloalkoxy; in some embodiments, Z 2 is CR Z2 , R Z2 is C 1-6 alkoxy; in some embodiments, Z 2 is CR Z2 , R Z2 is a methoxy group; in some embodiments, Z 2 is CR Z2 , and R Z2 is a hydrogen atom or a C 1-6 alkoxy group; in some embodiments, Z 2 is CH or COCH 3 .
  • R Z1 is selected from hydrogen atoms, halogens, C 1-6 alkoxy, C 1-6 alkyl, C 1-6 haloalkoxy and 3 to 6 membered cycloalkyl; in some In embodiments, R Z1 is a hydrogen atom or halogen; in some embodiments, R Z1 is a hydrogen atom or F.
  • the compound represented by the general formula (I), (II-2), (I'), (I'-1), (IM), (IN) or the compound thereof can Pharmaceutically acceptable salts, wherein R Z4 is a hydrogen atom or a C 1-6 alkyl group; preferably a C 1-6 alkyl group; more preferably CH 3 .
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein R 1 is selected from hydrogen atoms, halogens and C 1-6 alkyl groups; R 2 is -N(R 23 )SO 2 R 24 or -P(O)R 23 R 24 ; R 23 and R 24 are each independently a hydrogen atom or a C 1-6 alkyl group; R 3 is selected from halogen, C 1-6 Alkyl, C 1-6 alkoxy, C 1-6 haloalkyl or C 2-6 alkenyl; R 4 is hydrogen; Z 1 is CR Z1 ; R Z1 is selected from hydrogen atom, C 1-6 alkoxy group , C 1-6 alkyl and C 1-6 haloalkoxy; Z 2 is CR Z2 ; R Z2 is selected from hydrogen atom, C 1-6 alkoxy, C 1-6 alkyl and C 1-6 haloalkoxy Base; Z 3 is CH; -L 1 -L 2 -
  • R J is a hydrogen atom or C 1-6 alkyl group
  • R 03 is the same or different, and each is independently selected from halogen, hydroxyl, C 1-6 alkoxy, C 1-6 alkyl, C 1-6 haloalkyl and C 1-6 haloalkoxy; ring A is selected since:
  • Z 8 is CH or nitrogen atom;
  • Z 4 is CH 2 or C(O);
  • R Z4 is selected from hydrogen atom, C 1-6 alkyl group and C 1-6 haloalkyl group;
  • R Z7 is hydrogen atom or C 1-6 Alkyl group;
  • R Q is selected from hydrogen atom, halogen, C 1-6 alkyl group, C 1-6 haloalkyl group, C 1-6 alkoxy group, C 1-6 haloalkoxy group, cyano group and amino group.
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein R 1 is a hydrogen atom; R 2 is -N(R 23 )SO 2 R 24 , R 23 and R 24 are each independently a hydrogen atom or a C 1-6 alkyl group; R 3 is a halogen or a C 2-6 alkenyl group; R 4 is a hydrogen atom; Z 1 is CR Z1 , and R Z1 is a hydrogen atom or Halogen; Z 2 is CR Z2 , R Z2 is a hydrogen atom or C 1-6 alkoxy group; Z 3 is CH; L 1 is O, -L 2 -L 3 -L 4 - is -(CH 2 ) 3 - Or -(CH 2 ) 4 -, L 5 is CH 2 or O, L 6 is CH 2 or C(O); -J 1 -J 2 -J 3 -J 4 -J 5 -J 6 -selected from Ring A
  • the compound represented by the general formula (I'-1) or a pharmaceutically acceptable salt thereof wherein R 1 is a hydrogen atom; R 2 is -N(R 23 )SO 2 R 24 , R 23 and R 24 are each independently a hydrogen atom or a C 1-6 alkyl group; R 3 is a halogen; R 4 is a hydrogen atom; R Z1 is a hydrogen atom or a halogen; R Z2 is a hydrogen atom or C 1 -6 alkoxy; R L3 and R L4 are both H; L 5 is CH 2 or O, L 6 is CH 2 or C(O); n5 is 3, 4 or 5; Ring J 1' is -J 3 -J 4 -J 5 - is -C 1-6 alkylene-; J 6 is selected from Z 8 is CH or nitrogen atom; R Z4 is C 1-6 alkyl; j is 0.
  • the compound represented by the general formula (I'-2) or a pharmaceutically acceptable salt thereof wherein R 1 is a hydrogen atom; R 2 is -N(R 23 )SO 2 R 24 , R 23 and R 24 are each independently a hydrogen atom or a C 1-6 alkyl group; R 3 is a halogen; R 4 is a hydrogen atom; R Z1 is a hydrogen atom or a halogen; R Z2 is a hydrogen atom or C 1 -6 alkoxy; R L3 and R L4 are both H; L 5 is CH 2 or O, L 6 is CH 2 or C(O); n5 is 3, 4 or 5; Ring J 1' is -J 3 -J 4 -J 5 - is -C 1-6 alkylene-; J 6 is selected from R Z7 is a hydrogen atom or C 1-6 alkyl group; k is 0.
  • the compound represented by the general formula (I'-2) or a pharmaceutically acceptable salt thereof wherein R 1 is a hydrogen atom; R 2 is -N(R 23 )SO 2 R 24 , R 23 and R 24 are each independently a hydrogen atom or a C 1-6 alkyl group; R 3 is a halogen; R 4 is a hydrogen atom; R Z1 is a hydrogen atom or a halogen; R Z2 is a hydrogen atom or C 1 -6 alkoxy; R L3 and R L4 are both H; L 5 is CH 2 or O, L 6 is CH 2 or C(O); n5 is 3 or 4; Ring J 1' is piperidinyl or pipera Azinyl; -J 3 -J 4 -J 5 - is -C 1-6 alkylene-; J 6 is selected from bond, piperidinyl, piperazinyl, ethylamino and diazaspiro[5.5]eleven Alkyl
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein R 1 is a hydrogen atom; R 2 is -N(R 23 )SO 2 R 24 , R 23 and R 24 are the same or different, and are each independently selected from hydrogen atoms, C 1-6 alkyl groups and 3 to 6-membered cycloalkyl groups; R 3 is halogen or C 2-6 alkenyl group; R 4 is hydrogen Atom; Z 1 is CR Z1 , R Z1 is hydrogen atom or halogen; Z 2 is CR Z2 , R Z2 is hydrogen atom or C 1-6 alkoxy group; Z 3 is CH; L 1 is O, -L 2 - L 3 -L 4 - is -(CH 2 ) 3 - or -(CH 2 ) 4 -, L 5 is CH 2 or O, L 6 is CH 2 or C(O); -J 1 -J 2 -J 3 -J 4 -J 5 -
  • Z 8 is CH or nitrogen atom;
  • Z 4 is CH 2 or C(O);
  • R Z7 is hydrogen atom or C 1-6 alkyl;
  • R Z4 is C 1-6 alkyl;
  • R Q is halogen or C 1- 6 alkyl;
  • k is 0, 1, 2, 3 or 4;
  • j is 0, 1, 2 or 3;
  • s is 0, 1, 2 or 3.
  • the compound represented by the general formula (III-1) or a pharmaceutically acceptable salt thereof wherein R 1 is a hydrogen atom; R 2 is -N(R 23 )SO 2 R 24 , R 23 and R 24 are the same or different, and are each independently selected from hydrogen atoms, C 1-6 alkyl and 3 to 6-membered cycloalkyl; R 3 is halogen or C 2-6 alkenyl; R 4 is a hydrogen atom; R Z1 is a hydrogen atom or halogen; R Z2 is a hydrogen atom or C 1-6 alkoxy group; R L3 and R L4 are both hydrogen atoms; L 5 is CH 2 or O, L 6 is CH 2 or C(O); n5 is 3 or 4; J 1 is selected from bond, piperidinyl and piperazinyl; Ring C is selected from piperidinyl, piperazinyl and u is 0, 1 or 2; each R 03 is the same or different, and each is independently selected from
  • the compound represented by the general formula (III-2) or a pharmaceutically acceptable salt thereof wherein R 1 is a hydrogen atom; R 2 is -N(R 23 )SO 2 R 24 , R 23 and R 24 are the same or different, and are each independently selected from hydrogen atoms, C 1-6 alkyl and 3 to 6-membered cycloalkyl; R 3 is halogen or C 2-6 alkenyl; R 4 is a hydrogen atom; R Z1 is a hydrogen atom or halogen; R Z2 is a hydrogen atom or C 1-6 alkoxy group; R L3 and R L4 are both hydrogen atoms; L 5 is CH 2 or O, L 6 is CH 2 or C(O); n5 is 3 or 4; J 1 is selected from bond, piperidyl and piperazinyl; J 2 is selected from bond, C(O), CH 2 , piperidyl, piperazinyl and The piperidinyl and piperazinyl groups are each independently
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein R 1 is a hydrogen atom; R 2 is -N(R 23 )SO 2 R 24 Or -P(O)R 23 R 24 ; R 23 and R 24 are the same or different, and are each independently selected from hydrogen atoms, methyl and cyclopropyl groups; R 3 is halogen or C 2-6 alkenyl; R 4 is a hydrogen atom; Z 1 is CR Z1 , R Z1 is a hydrogen atom or halogen; Z 2 is CR Z2 , R Z2 is a hydrogen atom or C 1-6 alkoxy group; Z 3 is CH; L 1 is O, -L 2 -L 3 -L 4 - is -(CH 2 ) 3 - or -(CH 2 ) 4 -, L 5 is CH 2 or O, L 6 is CH 2 or C(O); -J 1 -J 2 -J 3 -J 4
  • Ring A is selected from:
  • Typical compounds of the present disclosure include, but are not limited to:
  • Another aspect of the present disclosure relates to compounds represented by general formula (II-1A) or (II-2A) or salts thereof:
  • R A is a hydrogen atom or an amino protecting group; the amino protecting group is preferably Boc;
  • Ring J 1' is a 5 to 14-membered nitrogen-containing heterocyclic group
  • R 1 , R 2 , R 3 , R 4 , R Z1 , R Z2 , R L3 , R L4 and J 1 are as defined by general formula (I);
  • R 1 , R 2 , R 3 , R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , ring J 1' and n5 are as defined in the general formula (I').
  • Another aspect of the present disclosure relates to compounds represented by general formula (IMA) or (INA) or salts thereof:
  • Ring C is a nitrogen-containing heterocyclic group
  • n5 is 1, 2, 3, 4, 5, 6 or 7;
  • R 1 , R 2 , R 3 , R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , J 1 , J 2 and R 03 are as defined in general formula (I).
  • Another aspect of the present disclosure relates to a compound represented by general formula (II-3'A) or a salt thereof:
  • Ring C, R 1 , R 2 , R 3 , R 4 , R Z1 , R Z2 , R L3 , R L4 , n5, L 5 , L 6 , J 1 , J 2 , R 03 and u are as in the general formula (II -3') as defined in.
  • Another aspect of the present disclosure relates to a compound represented by general formula (II-3B) or a salt thereof:
  • J 5 is a C 1-6 alkylene group, and the C 1-6 alkylene group is substituted by 1 or more selected from halogen, hydroxyl, C 1-6 alkyl, C 1-6 haloalkyl and C 1-6 alkyl Substituted by an oxygen substituent;
  • Ring J 3' is a 5- to 14-membered nitrogen-containing heterocyclic group, and the 5- to 14-membered nitrogen-containing heterocyclic group is optionally substituted by one or more members selected from halogen, hydroxyl, C 1-6 alkyl and C 1 -6 substituted by haloalkyl;
  • J 4 is O, OC 1-6 alkylene, C 1-6 alkylene O, S, NH, NHC 1-6 alkylene, C 1-6 alkylene NH, C 1-6 alkylene , C(O), C(O)C 1-6 alkylene or C 1-6 alkylene C(O);
  • R J is a hydrogen atom or C 1-6 alkyl group;
  • Z 4 is CH 2 or C(O);
  • RQ is each independently selected from halogen, C 1-6 haloalkyl and cyano.
  • Typical intermediate compounds of the present disclosure or salts thereof include, but are not limited to:
  • Another aspect of the present disclosure relates to a method for preparing the compound represented by the above general formula (I') or a pharmaceutically acceptable salt thereof.
  • method which includes:
  • a compound represented by the general formula (I'A) or a salt thereof is subjected to a condensation reaction with a compound represented by the general formula (I'B) or a salt thereof to obtain a compound represented by the general formula (I') or its medicinal salt,
  • ring A, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , ring J 1' , J 3 to J 6 and n5 are as in the general formula (I') definition.
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by the above general formula (IM) or a pharmaceutically acceptable salt thereof, which method includes:
  • the compound represented by the general formula (IMA) or a salt thereof (preferably a hydrochloride or trifluoroacetate) reacts with the compound represented by the general formula (IMB) or a salt thereof to obtain the compound represented by the general formula (IM).
  • IMA general formula
  • IMB general formula
  • IM general formula
  • ring A, ring C, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , n5, L 5 , L 6 , J 1 , q, J 4 to J 6 , R 03 and u are as follows It is defined in the general formula (IM).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by the above general formula (IN) or a pharmaceutically acceptable salt thereof, which method includes:
  • the compound represented by the general formula (INA) or a salt thereof undergoes a reductive amination reaction with the compound represented by the general formula (INB) or a salt thereof (preferably a hydrochloride) to obtain a compound represented by the general formula (IN) or its salt.
  • medicinal salt a reductive amination reaction with the compound represented by the general formula (INB) or a salt thereof (preferably a hydrochloride) to obtain a compound represented by the general formula (IN) or its salt.
  • ring A, ring B, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , n5, L 5 , L 6 , J 1 , J 2 , q, J 4 to J 6 , R 03 and u are as defined in general formula (IN).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by the above general formula (II-1), (II-2) or (II-3) or a pharmaceutically acceptable salt thereof, which method includes:
  • the compound represented by the general formula (II-1A) or a salt thereof (preferably a hydrochloride) and the compound represented by the general formula (II-1B) or a salt thereof are subjected to a condensation reaction to obtain a compound represented by the general formula (II-1) compound or a pharmaceutically acceptable salt thereof,
  • the compound represented by the general formula (II-1A) or a salt thereof (preferably a hydrochloride) and the compound represented by the general formula (II-2B) or a salt thereof are subjected to a condensation reaction to obtain a compound represented by the general formula (II-2) compound or a pharmaceutically acceptable salt thereof,
  • the compound represented by the general formula (II-2A) or a salt thereof is subjected to a reductive amination reaction with the compound represented by the general formula (II-3B) or a salt thereof to obtain a compound represented by the general formula (II-3) or a pharmaceutically acceptable compound thereof salt used; among them,
  • R A is a hydrogen atom
  • R J is a hydrogen atom or a C 1-6 alkyl group
  • Ring J 1 ' is a 5 to 14-membered nitrogen-containing heterocyclic group
  • Ring J 3' is a 5- to 14-membered nitrogen-containing heterocyclic group, and the 5- to 14-membered nitrogen-containing heterocyclic group is optionally substituted by one or more members selected from halogen, hydroxyl, C 1-6 alkyl and C 1 -6 substituted by haloalkyl substituents; n2 is 2, 3, 4, 5, 6, 7 or 8; x is 0, 1, 2 or 3; s is 0, 1 or 2; q is 1, 2, 3 or 4;
  • R 1 , R 2 , R 3 , R 4 , R Z1 , R Z2 , R L3 , R L4 , J 1 , J 3 , J 4 , J 5 , J 6 , Z 4 , R Z4 , Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and R Q are as defined in general formula (I).
  • Another aspect of the present disclosure relates to a method for preparing the compound represented by the above general formula (II-3') or a pharmaceutically acceptable salt thereof, which method includes:
  • the compound represented by the general formula (II-3'A) or a salt thereof (preferably a hydrochloride) and the compound represented by the general formula (II-3'B) or a salt thereof are subjected to a condensation reaction to obtain the general formula (II-3 ') or a pharmaceutically acceptable salt thereof,
  • ring C, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , n5, L 5 , L 6 , J 1 , J 2 , R 03 , u, R J5 , R J6 , t, R J , R Q , Z 4 and s are as defined in general formula (II-3').
  • Another aspect of the present disclosure relates to a method for preparing the compound represented by the above general formula (I'-1) or a pharmaceutically acceptable salt thereof, which method includes:
  • the compound represented by the general formula (I'A) or a salt thereof (preferably a hydrochloride) and the compound represented by the general formula (II'-2B) or a salt thereof are subjected to a condensation reaction to obtain the compound represented by the general formula (I'-1). the compound shown or a pharmaceutically acceptable salt thereof,
  • R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , n5, ring J 1' , J 3 to J 6 , R Q , R Z4 , Z 8 and j are as follows It is defined in the general formula (I'-1).
  • Another aspect of the present disclosure relates to a method for preparing the compound represented by the above general formula (I'-2) or a pharmaceutically acceptable salt thereof, which method includes:
  • the compound represented by the general formula (I'A) or a salt thereof (preferably a hydrochloride) and the compound represented by the general formula (I'-2B) or a salt thereof are subjected to a condensation reaction to obtain the compound represented by the general formula (I'-2). the compound shown or a pharmaceutically acceptable salt thereof,
  • R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , n5, ring J 1' , J 3 to J 6 , R Q , R Z7 and k are as in the general formula ( As defined in I'-2).
  • Another aspect of the present disclosure relates to a method for preparing the compound represented by the above general formula (III-1) or a pharmaceutically acceptable salt thereof, which method includes:
  • ring C, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , n5, L 5 , L 6 , J 1 , R 03 , u, q, J 4 to J 6 , R Q , Z 4 and s are as defined in general formula (III-1).
  • Another aspect of the present disclosure relates to a method for preparing the compound represented by the above general formula (III-2) or a pharmaceutically acceptable salt thereof, which method includes:
  • the compound represented by the general formula (INA) or a salt thereof undergoes a reductive amination reaction with the compound represented by the general formula (III-2B) or a salt thereof (preferably a hydrochloride) to obtain a compound represented by the general formula (III-2) compound or a pharmaceutically acceptable salt thereof,
  • ring B, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , n5, L 5 , L 6 , J 1 , J 2 , q, R 03 , u, J 4 to J 6 , R Q , Z 4 and s are as defined in general formula (III-2).
  • Another aspect of the present disclosure relates to a pharmaceutical composition containing a therapeutically effective amount of general formula (I), general formula (II-1), general formula (II-2), general formula ( II-3), (I'), (I'-1), (I'-2), (IM), (IN), (II-3'), (III-1), (III-2) and a compound shown in Table A or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • general formula (I) general formula (II-1), general formula (II-2), general formula ( II-3), (I'), (I'-1), (I'-2), (IM), (IN), (II-3'), (III-1), (III-2) and a compound shown in Table A or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the present disclosure further relates to general formula (I), general formula (II-1), general formula (II-2), general formula (II-3), (I'), (I'-1), (I'- 2), (IM), (IN), (II-3'), (III-1), (III-2) and the compounds shown in Table A or pharmaceutically acceptable salts thereof, or pharmaceutical compositions containing them Use in the preparation of drugs for modulating EGFR ubiquitination and degradation.
  • the present disclosure further relates to general formula (I), general formula (II-1), general formula (II-2), general formula (II-3), (I'), (I'-1), (I'- 2), (IM), (IN), (II-3'), (III-1), (III-2) and the compounds shown in Table A or pharmaceutically acceptable salts thereof, or pharmaceutical compositions containing them Use in the preparation of a medicament for the treatment and/or prevention of EGFR-mediated or -dependent diseases or conditions.
  • the EGFR-mediated or dependent disease or condition is preferably selected from cancer.
  • the present disclosure further relates to general formula (I), general formula (II-1), general formula (II-2), general formula (II-3), (I'), (I'-1), (I'- 2), (IM), (IN), (II-3'), (III-1), (III-2) and the compounds shown in Table A or pharmaceutically acceptable salts thereof, or pharmaceutical compositions containing them
  • the cancer is selected from squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, liver cancer, kidney cancer, bladder cancer, breast cancer, and cervical cancer , colorectal cancer, esophageal cancer, head and neck cancer, nasopharyngeal cancer, oral cancer, salivary gland cancer, kidney cancer, liver cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, gastric cancer, leukemia, lymphoma, glioma, neurological mother Cytoma, melanoma, sarcoma, endometrial
  • the present disclosure also relates to a method for regulating EGFR protein ubiquitination and degradation in a subject, which includes administering the above general formula (I), general formula (II-1), general formula (II-2) to the patient in need , General formula (II-3), (I'), (I'-1), (I'-2), (IM), (IN), (II-3'), (III-1), ( III-2) or the compound shown in Table A or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition containing the same.
  • the present disclosure also relates to a method for inhibiting and/or degrading EGFR protein, which includes administering the above general formula (I), general formula (II-1), general formula (II-2), general formula (II- 3), (I'), (I'-1), (I'-2), (IM), (IN), (II-3'), (III-1), (III-2) or table
  • the present disclosure also relates to a method for treating and/or preventing EGFR-mediated or dependent diseases or conditions, which includes administering to a patient in need the above general formula (I), general formula (II-1), general formula ( II-2), general formula (II-3), (I'), (I'-1), (I'-2), (IM), (IN), (II-3'), (III- 1), (III-2) or the compound shown in Table A or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition containing the same.
  • the present disclosure also relates to a method for treating and/or preventing cancer, which includes administering the above general formula (I), general formula (II-1), general formula (II-2), general formula (II-3) to a patient in need ), (I'), (I'-1), (I'-2), (IM), (IN), (II-3'), (III-1), (III-2) or Table A
  • the present disclosure further relates to the above-mentioned general formula (I), general formula (II-1), general formula (II-2), general formula (II-3), (I'), (I'-1), ( I'-2), (IM), (IN), (II-3'), (III-1), (III-2) or compounds shown in Table A or pharmaceutically acceptable salts thereof, or compounds containing them A pharmaceutical composition for use as a medicament.
  • the present disclosure further relates to the above-mentioned general formula (I), general formula (II-1), general formula (II-2), general formula (II-3), (I'), (I'-1), ( I'-2), (IM), (IN), (II-3'), (III-1), (III-2) or compounds shown in Table A or pharmaceutically acceptable salts thereof, or compounds containing them
  • a pharmaceutical composition for use as a drug that regulates ubiquitination and degradation of EGFR protein in a subject
  • the present disclosure further relates to the above-mentioned general formula (I), general formula (II-1), general formula (II-2), general formula (II-3), (I'), (I'-1), ( I'-2), (IM), (IN), (II-3'), (III-1), (III-2) or compounds shown in Table A or pharmaceutically acceptable salts thereof, or compounds containing them
  • Pharmaceutical compositions for use as EGFR inhibitors and/or degraders Pharmaceutical compositions for use as EGFR inhibitors and/or degraders.
  • the present disclosure further relates to the above-mentioned general formula (I), general formula (II-1), general formula (II-2), general formula (II-3), (I'), (I'-1), ( I'-2), (IM), (IN), (II-3'), (III-1), (III-2) or compounds shown in Table A or pharmaceutically acceptable compounds thereof A salt, or a pharmaceutical composition containing the same, for use as a medicament for the treatment and/or prevention of EGFR-mediated or dependent diseases or conditions.
  • the present disclosure further relates to the above general formula (I), general formula (II-1), general formula (II-2), general formula (II-3), (I'), (I'-1), (I' -2), (IM), (IN), (II-3'), (III-1), (III-2) or a compound shown in Table A or a pharmaceutically acceptable salt thereof, or a medicine containing the same Compositions for modulating EGFR protein ubiquitination and degradation in a subject.
  • the present disclosure further relates to the above general formula (I), general formula (II-1), general formula (II-2), general formula (II-3), (I'), (I'-1), (I' -2), (IM), (IN), (II-3'), (III-1), (III-2) or a compound shown in Table A or a pharmaceutically acceptable salt thereof, or a medicine containing the same Compositions for inhibiting and/or degrading EGFR protein.
  • the present disclosure further relates to the above general formula (I), general formula (II-1), general formula (II-2), general formula (II-3), (I'), (I'-1), (I' -2), (IM), (IN), (II-3'), (III-1), (III-2) or a compound shown in Table A or a pharmaceutically acceptable salt thereof, or a medicine containing the same Compositions for the treatment and/or prevention of EGFR-mediated or -dependent diseases or conditions.
  • the present disclosure further relates to the above-mentioned general formula (I), general formula (II-1), general formula (II-2), general formula (II-3), (I'), (I'-1), ( I'-2), (IM), (IN), (II-3'), (III-1), (III-2) or compounds shown in Table A or pharmaceutically acceptable salts thereof, or compounds containing them
  • a pharmaceutical 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, liver cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, gastric cancer, leukemia, lymphoma, glioma, neuroblastoma Cytomas, melanomas, sarcomas, endometri
  • the EGFR-mediated or dependent disease or disorder described in the present disclosure is cancer; the disease or disorder is preferably selected from the group consisting of squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, liver cancer, renal cancer, Bladder cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, head and neck cancer, nasopharyngeal cancer, oral cancer, salivary gland cancer, kidney cancer, liver cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, gastric cancer, leukemia, lymphoma tumors, glioma, neuroblastoma, melanoma, sarcoma, endometrial cancer, testicular cancer, thyroid cancer, glioblastoma, brain metastases, solid tumors, oropharyngeal cancer, bronchial tumors and skin Cancer; more preferably lung cancer; more preferably non-small cell lung cancer.
  • the lung cancer is preferably small cell lung cancer or non-small cell lung cancer
  • the non-small cell lung cancer is preferably lung adenocarcinoma, lung squamous cell carcinoma or large cell lung cancer
  • the head and neck cancer is preferably head and neck squamous cell carcinoma
  • the esophageal cancer is preferably Esophageal squamous cell carcinoma
  • the glioblastoma is preferably glioblastoma multiforme.
  • the cancer described in this disclosure is preferably an EGFR protein with an L858R mutation.
  • the cancer described in this disclosure is preferably an EGFR protein with a 19del mutation.
  • the cancer described in this disclosure is preferably an EGFR protein with a T790M mutation.
  • the cancer described in this disclosure is preferably an EGFR protein with a C797X mutation.
  • the cancer described in this disclosure is preferably an EGFR protein with L858R and T790M mutations.
  • the cancer described in this disclosure is preferably an EGFR protein with 19del and T790M mutations.
  • the cancer described in this disclosure is preferably an EGFR protein with L858R and C797X mutations.
  • the cancer described in this disclosure is preferably an EGFR protein with 19del and C797X mutations.
  • the cancer described in this disclosure is preferably an EGFR protein with T790M and C797X mutations.
  • the cancers described in this disclosure are preferably EGFR proteins with L858R, T790M and C797X mutations.
  • Cancers described in this disclosure are preferably EGFR proteins with 19del, T790M and C797X mutations.
  • the C797X mutation described in the present disclosure is preferably a C797S mutation; where X represents any amino acid including S.
  • the active compounds may be formulated in a form suitable for administration by any suitable route, preferably in unit dose form, or in such form that the patient may self-administer in single doses.
  • the unit dosage form of a compound or composition of the present disclosure may be expressed as a tablet, capsule, cachet, bottled solution, powder, granule, lozenge, suppository, reconstituted powder or liquid preparation.
  • suitable unit doses may range from 0.1 to 1000 mg.
  • the pharmaceutical composition of the present disclosure may contain one or more auxiliary materials selected from the following ingredients: fillers (diluents), binders, wetting agents, disintegrants or excipients wait.
  • auxiliary materials selected from the following ingredients: fillers (diluents), binders, wetting agents, disintegrants or excipients wait.
  • the composition may contain from 0.1 to 99% by weight of active compound.
  • compositions containing the active ingredients may be in forms suitable for oral administration, such as tablets, dragees, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixir.
  • Oral compositions may be prepared according to any method known in the art for preparing pharmaceutical compositions, and such compositions may contain one or more ingredients selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preservatives, to provide medicinal preparations that are pleasing to the eye and palatable.
  • Tablets contain the active ingredient in admixture with nontoxic pharmaceutically acceptable excipients suitable for the manufacture of tablets.
  • excipients may be inert excipients, granulating agents, disintegrating agents, binders and lubricants. These tablets may be uncoated or may be coated by known techniques to mask the taste of the drug or to delay disintegration and absorption in the gastrointestinal tract, thereby providing sustained release over an extended period of time.
  • Oral formulations may also be presented in soft gelatin capsules in which the active ingredient is mixed with an inert solid diluent, or in which the active ingredient is mixed with a water-soluble carrier or oil vehicle.
  • Aqueous suspensions contain the active substances and excipients suitable for the preparation of aqueous suspensions for mixing. Such excipients are suspending, dispersing 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 formulated by suspending the active ingredient in vegetable or mineral oil.
  • Oil suspensions may contain thickening agents. Sweetening and flavoring agents as described above may be added to provide a palatable preparation. These compositions can be preserved by adding antioxidants.
  • compositions of the present disclosure may also be in the form of oil-in-water emulsions.
  • the oil phase can be vegetable oil, mineral oil or mixtures thereof.
  • Suitable emulsifiers may be naturally occurring phospholipids, and the emulsions may also contain sweeteners, flavoring agents, preservatives and antioxidants.
  • Such preparations may also contain demulcents, preservatives, coloring agents and antioxidants.
  • compositions of the present disclosure may be in the form of sterile injectable aqueous solutions.
  • Acceptable vehicles or solvents that may be used are water, Ringer's solution and isotonic sodium chloride solution.
  • Sterile injectable preparations may be sterile injectable oil-in-water microemulsions in which the active ingredient is dissolved in an oily phase.
  • the injectable solution or microemulsion may be injected into the patient's bloodstream by local mass injection.
  • solutions and microemulsions are preferably administered in a manner that maintains constant circulating concentrations of the compounds of the present disclosure.
  • continuous intravenous drug delivery devices can be used.
  • An example of such a device is the Deltec CADD-PLUS.TM.5400 intravenous pump.
  • compositions of the present disclosure may be in the form of sterile injectable aqueous or oily suspensions for intramuscular and subcutaneous administration.
  • the suspension may be formulated according to known techniques using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension prepared in a nontoxic parenterally acceptable diluent or solvent.
  • sterile fixed oil can be conveniently used as the solvent or suspending medium. For this purpose, any blend of fixed oils can be used.
  • fatty acids are also prepared as injectables.
  • the compounds of the present disclosure may be administered in the form of suppositories for rectal administration.
  • These pharmaceutical compositions may 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 dissolve in the rectum to release the drug.
  • the dosage of a drug depends on a variety of factors, including, but not limited to, the activity of the specific compound used, the patient's age, the patient's weight, the patient's health, and the patient's behavior. , patient's diet, administration time, administration method, excretion rate, combination of drugs, severity of disease, etc.; in addition, the best treatment method such as treatment mode, daily dosage of compounds or pharmaceutically acceptable salts Types can be verified based on traditional treatment regimens.
  • alkyl refers to a saturated linear or branched aliphatic hydrocarbon group having 1 to 20 (for example, 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. C 1-20 alkyl).
  • the alkyl group is preferably an alkyl group having 1 to 12 carbon atoms (ie, C 1-12 alkyl group), more preferably an alkyl group having 1 to 6 carbon atoms (ie, C 1-6 alkyl group).
  • 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
  • the alkyl group may be substituted or unsubstituted, and when substituted, it may be substituted at any available point of attachment.
  • the substituents are preferably selected from the group consisting of D atoms, halogen, alkoxy, haloalkyl, haloalkoxy, cyclic One or more of alkyloxy, 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., C 1-20 alkylene group).
  • the alkylene group is preferably an alkylene group having 1 to 12 carbon atoms (i.e., C 1-12 alkylene group), and more preferably an alkylene group having 1 to 6 carbon atoms (i.e., C 1-6 alkylene group ).
  • Non-limiting examples include: -CH 2 -, -CH(CH 3 )-, -C(CH 3 ) 2 -, -CH 2 CH 2 -, -CH(CH 2 CH 3 )-, -CH 2 CH (CH 3 )-, -CH 2 C(CH 3 ) 2 -, -CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 -, etc.
  • the alkylene group may be substituted or unsubstituted, and when substituted, it may be substituted at any available point of attachment.
  • the substituent is preferably selected from the group consisting of D atoms, halogen, alkoxy, haloalkyl, haloalkoxy, One or more of cycloalkyloxy, heterocyclyloxy, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • C 0-6 alkylene refers to an alkylene group having 0, 1, 2, 3, 4, 5 or 6 carbon atoms; specifically, a C 0-6 alkylene group includes bonds 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, where the alkyl group is as defined above and has 2 to 12 (for example, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12) carbon atoms (i.e. C 2-12 alkenyl).
  • the alkenyl group is preferably an alkenyl group having 2 to 6 carbon atoms (i.e., C 2-6 alkenyl group).
  • Non-limiting examples include: vinyl, propenyl, isopropenyl, butenyl, and the like.
  • the alkenyl group may be substituted or unsubstituted, and when substituted, it may be substituted at any available point of attachment.
  • the substituents are preferably selected from the group consisting of D atoms, alkoxy, halogen, haloalkyl, haloalkoxy, cyclic One or more of alkyloxy, heterocyclyloxy, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • alkenylene refers to a divalent alkenyl group, wherein alkenyl is as defined above and has 2 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 (ie, C 2-20 alkenylene).
  • the alkenylene group is preferably an alkenylene group having 2 to 12 carbon atoms (i.e., C 2-12 alkenylene group), and more preferably an alkenylene group having 2 to 6 carbon atoms (i.e., C 1-6 alkenylene group).
  • Alkenylene may be substituted or unsubstituted, and when substituted, it may be substituted at any available point of attachment.
  • the substituent is preferably selected from the group consisting of D atom, alkoxy, halogen, haloalkyl, haloalkoxy, One or more of 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, where the alkyl group is as defined above and has 2 to 12 (for example, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12) carbon atoms (i.e. C 2-12 alkynyl).
  • the alkynyl group is preferably an alkynyl group having 2 to 6 carbon atoms (ie, C 2-6 alkynyl group).
  • Non-limiting examples include: ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like.
  • Alkynyl can be Substituted or unsubstituted, when substituted, it can be substituted at any available point of attachment.
  • the substituent is preferably selected from D atom, alkoxy, halogen, haloalkyl, haloalkoxy, cycloalkyloxy , one or more of heterocyclyloxy, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • alkynylene refers to a divalent alkynyl group, wherein the alkynyl group is as defined above and has 2 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 (ie C 2-20 alkynylene).
  • the alkynylene group is preferably an alkynylene group having 2 to 12 carbon atoms (i.e., C 2-12 alkynylene group), and more preferably an alkynylene group having 2 to 6 carbon atoms (i.e., C 2-6 alkynylene group) ).
  • Non-limiting examples include: ethynylene, -CH 2 C ⁇ C-, -CH 2 CH 2 C ⁇ C-, etc.
  • the alkynylene group may be substituted or unsubstituted, and when substituted, it may be substituted at any available point of attachment.
  • the substituent is preferably selected from the group consisting of D atom, alkoxy, halogen, haloalkyl, haloalkoxy, One or more of cycloalkyloxy, heterocyclyloxy, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • alkoxy refers to -O-(alkyl), where alkyl is as defined above. Non-limiting examples include: methoxy, ethoxy, propoxy, butoxy, and the like. Alkoxy may be substituted or unsubstituted, and when substituted, it may be substituted at any available point of attachment.
  • the substituent is preferably selected from the group consisting of D atom, halogen, alkoxy, haloalkyl, haloalkoxy, One or more of 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 group preferably has 3 to 12 ring atoms (i.e., 3 to 12-membered cycloalkyl) or 3 to 10 ring atoms (i.e., 3 to 10-membered cycloalkyl), more Preferred are cycloalkyl groups having 3 to 8 ring atoms (ie, 3 to 8 membered cycloalkyl groups), most preferably cycloalkyl groups having 3 to 6 ring atoms (ie, 3 to 6 membered cycloalkyl groups), and more preferably Cycloalkyl group with 5 or 6 ring atoms (i.e. 5- or 6-membered cycloalkyl group).
  • Non-limiting examples of the monocyclic cycloalkyl include: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, and cycloheptyl , cycloheptatrienyl and cyclooctyl, etc.
  • 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 a spiro atom) is shared between the rings.
  • the ring may contain one or more double bonds, or the ring may contain one or more atoms selected from nitrogen, Heteroatoms of oxygen and sulfur (the nitrogen can be optionally oxidized, that is, to form nitrogen oxides; the sulfur can be optionally oxidized, that is, to form sulfoxide or sulfone, but does not include -OO-, -OS - or -SS-), the condition is that it contains at least one all-carbon ring and the connection point is on the all-carbon ring, which has 5 to 20 (for example, 5, 6, 7, 8, 9, 10, 11, 12, 13 , 14, 15, 16, 17, 18, 19 or 20) ring atoms (i.e.
  • the spirocycloalkyl group is preferably a spirocycloalkyl group having 6 to 14 ring atoms (i.e., a 6- to 14-membered spirocycloalkyl group), and more preferably a spirocycloalkyl group having 7 to 10 ring atoms (i.e., a 7 to 10-membered spirocycloalkyl group). membered spirocycloalkyl).
  • the spirocycloalkyl group includes a single spirocycloalkyl group and a polyspirocycloalkyl group (such as a double spirocycloalkyl group, etc.), preferably a single spirocycloalkyl group or a double spirocycloalkyl group, 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-membered/6-membere
  • 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 group fused with one or more monocyclic cycloalkyl groups, or a monocyclic cycloalkyl group fused with a heterocyclic cycloalkyl group.
  • One or more of the cyclic groups, aryl groups or heteroaryl groups are condensed, wherein the point of attachment is on a single-ring cycloalkyl group, which may contain one or more double bonds in the ring, and has 5 to 20 (for example, 5 , 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) ring atoms (i.e.
  • the fused ring alkyl group is preferably a fused ring alkyl group having 6 to 14 ring atoms (ie, a 6 to 14-membered fused ring alkyl group), and more preferably a fused ring alkyl group having 7 to 10 ring atoms (ie, a 7 to 10 membered ring alkyl group). fused ring alkyl group).
  • the fused cycloalkyl group includes bicyclic fused cycloalkyl and polycyclic fused cycloalkyl (such as tricyclic fused cycloalkyl, tetracyclic fused cycloalkyl, etc.), preferably bicyclic fused cycloalkyl or tricyclic fused 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 yuan, 6
  • bridged cycloalkyl refers to an all-carbon polycyclic ring system that shares two carbon atoms that are not directly connected between the rings.
  • the ring may contain one or more double bonds and has 5 to 20 (for example, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) carbon atoms (ie, 5 to 20 membered bridged cycloalkyl).
  • the bridged cycloalkyl group is preferably a bridged cycloalkyl group having 6 to 14 carbon atoms (i.e., a 6- to 14-membered bridged cycloalkyl group), and more preferably a bridged cycloalkyl group having 7 to 10 carbon atoms (i.e., a 7 to 10-membered bridged cycloalkyl group). bridged cycloalkyl).
  • the bridged cycloalkyl group includes bicyclic bridged cycloalkyl and polycyclic bridged cycloalkyl (such as tricyclic bridged cycloalkyl, tetracyclic bridged cycloalkyl, etc.), preferably bicyclic bridged cycloalkyl or tricyclic bridged cycloalkyl .
  • Non-limiting examples include:
  • Cycloalkyl may be substituted or unsubstituted, and when substituted, it may be substituted at any available point of attachment.
  • the substituents are preferably selected from the group consisting of D atoms, halogen, alkyl, alkoxy, haloalkyl, haloalkyl
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic heterocycle (i.e., monocyclic heterocyclyl) or a polycyclic heterocyclic system (i.e., polycyclic heterocyclyl), which contains at least one ring (such as 1, 2, 3 or 4) heteroatoms selected from nitrogen, oxygen and sulfur (the nitrogen can be optionally oxidized, that is, to form nitrogen oxides; the sulfur can be optionally oxogenated, that is, to form sulfoxide or sulfone, but not including -O-O-, -O-S- or -S-S-), and having 3 to 20 (e.g.
  • the heterocyclyl group preferably has a heterocyclyl group with 3 to 12 ring atoms (i.e., a 3 to 12-membered heterocyclyl group) or a heterocyclyl group with 3 to 10 ring atoms (i.e., a 3 to 10-membered heterocyclyl group); further Heterocyclyl groups having 3 to 8 ring atoms (i.e., 3 to 8-membered heterocyclyl groups) are preferred; heterocyclyl groups having 3 to 6 ring atoms (i.e., 3 to 6-membered heterocyclyl groups) or heterocyclyl groups having 4 to 6 ring atoms are more preferred.
  • Heterocyclyl groups with 7 ring atoms i.e., 4 to 7-membered heterocyclyl groups
  • most preferred are heterocyclyl groups with 5 or 6 ring atoms (i.e., 5- or 6-membered heterocyclyl groups).
  • Non-limiting examples of the monocyclic heterocyclyl include: pyrrolidinyl, tetrahydropyranyl, 1,2,3,6-tetrahydropyridyl, piperidinyl, piperazinyl, nitrogen heterocycle Butyl, morpholinyl, thiomorpholinyl and homopiperazinyl, etc.
  • the polycyclic heterocyclyl group includes spiroheterocyclyl group, fused heterocyclyl group and bridged heterocyclyl group.
  • spiroheterocyclyl refers to a polycyclic heterocyclic system in which the rings share one atom (called a spiro atom).
  • the ring may contain one or more double bonds, and the ring contains at least one (for example, 1, 2, 3 or 4) heteroatoms selected from nitrogen, oxygen and sulfur (the nitrogen can be optionally oxidized, that is, to form nitrogen oxides; the sulfur can be optionally oxo-substituted, that is, to form sulfoxide or sulfone, But excluding -OO-, -OS- or -SS-), provided that it contains at least one monocyclic heterocyclyl group and the point of attachment is on the monocyclic heterocyclyl group, which has 5 to 20 (for example, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) ring atoms (ie, 5 to 20 membered spiroheterocyclyl).
  • the spiroheterocyclyl group is preferably a spiroheterocyclyl group having 6 to 14 ring atoms (i.e., a 6- to 14-membered spiroheterocyclyl group), and more preferably a spiroheterocyclyl group having 7 to 14 ring atoms (i.e., a 7 to 14-membered spiroheterocyclyl group). membered spiroheterocyclyl) or a spiroheterocyclyl having 7 to 11 ring atoms (i.e., 7 to 11 membered spiroheterocyclyl).
  • the spiroheterocyclyl group includes a single spiroheterocyclyl group and a polyspiroheterocyclyl group (such as a double spiroheterocyclyl group, etc.), preferably a single spiroheterocyclyl group or a double spiroheterocyclyl group, more preferably 3-membered/4-membered, 3-membered or 3-membered spiroheterocyclyl.
  • Non-limiting examples include: wait.
  • fused heterocyclyl refers to a polycyclic heterocyclic system in which two adjacent atoms are shared between the rings.
  • the ring may contain one or more double bonds, and the ring contains at least one (for example, 1, 2, 3 or 4) heteroatoms selected from nitrogen, oxygen and sulfur (the nitrogen can be optionally oxidized, that is, to form nitrogen oxides; the sulfur can be optionally oxoated, that is, to form sulfoxide or sulfone, but not Including -OO-, -OS- or -SS-), which is a monocyclic heterocyclyl fused with one or more monocyclic heterocyclyl groups, or a monocyclic heterocyclyl group with a cycloalkyl, aryl or heteroaryl group.
  • the fused heterocyclyl group is preferably a fused heterocyclyl group having 6 to 14 ring atoms (i.e., a 6- to 14-membered fused heterocyclyl group), and more preferably a fused heterocyclyl group having 7 to 10 ring atoms (i.e., a 7 to 10-membered fused heterocyclyl group). fused heterocyclic group).
  • the fused heterocyclyl group includes bicyclic and polycyclic fused heterocyclyl groups (such as tricyclic fused heterocyclyl group, tetracyclic fused heterocyclyl group, etc.), preferably bicyclic fused heterocyclyl group or tricyclic fused heterocyclyl group, 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-membered/6-membered bicyclic fused heterocyclyl
  • bridged heterocyclyl refers to a polycyclic heterocyclic system that shares two atoms that are not directly connected between the rings.
  • the ring may contain one or more double bonds, and the ring contains at least one (for example, 1, 2, 3 or 4) heteroatoms selected from nitrogen, oxygen and sulfur (the nitrogen can be optionally oxidized, that is, to form nitrogen oxides; the sulfur can be optionally oxo-substituted, that is, to form sulfoxide or sulfone, but not including -OO-, -OS- or -SS-), which have 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.
  • the bridged heterocyclyl group is preferably a bridged heterocyclyl group having 6 to 14 ring atoms (i.e., a 6- to 14-membered bridged heterocyclyl group), and more preferably a bridged heterocyclyl group having 7 to 10 ring atoms (i.e., a 7 to 10-membered bridged heterocyclyl group). Yuan-bridged heterocyclyl).
  • bicyclic bridged heterocyclyl and polycyclic bridged heterocyclyl such as tricyclic bridged heterocyclyl, tetracyclic bridged heterocyclyl, etc.
  • bicyclic bridged heterocyclyl or tricyclic bridged heterocyclyl preferably bicyclic bridged heterocyclyl or tricyclic bridged heterocyclyl. base.
  • Non-limiting examples include:
  • Heterocyclyl may be substituted or unsubstituted. When substituted, it may be substituted at any available point of attachment.
  • the substituent is preferably selected from D atoms, halogen, alkyl, alkoxy, haloalkyl, haloalkyl.
  • aryl refers to a monocyclic all-carbon aromatic ring (i.e., a monocyclic aryl) or a polycyclic aromatic ring system (i.e., a polycyclic aryl) having a conjugated ⁇ electron system, having 6 to 14 (e.g., 6 , 7, 8, 9, 10, 11, 12, 13 or 14) ring atoms (i.e. 6 to 14 membered aryl group).
  • the aryl group is preferably an aryl group having 6 to 10 ring atoms (ie, 6 to 10 membered aryl group).
  • the monocyclic aryl group is, for example, phenyl.
  • Non-limiting examples of the polycyclic aromatic group include: naphthyl, anthracenyl, phenanthrenyl, etc.
  • the polycyclic aryl group also includes phenyl fused with one or more of heterocyclyl or cycloalkyl, or naphthyl fused with one or more of heterocyclyl or cycloalkyl, where the point of attachment on phenyl or naphthyl, and in this case, the number of ring atoms continues to mean the number of ring atoms in the polycyclic aromatic ring system, non-limiting examples include:
  • Aryl groups may be substituted or unsubstituted. When substituted, they may be substituted at any available point of attachment.
  • the substituents are preferably selected from D atoms, halogens, alkyl groups, alkoxy groups, haloalkyl groups, haloalkoxy groups. or Multiple.
  • heteroaryl refers to a monocyclic heteroaromatic ring (i.e., monocyclic heteroaryl) or a polycyclic heteroaromatic ring system (i.e., polycyclic heteroaryl) with a conjugated ⁇ electron system, which contains at least one (For example, 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 oxo-substituted, i.e.
  • the heteroaryl group is preferably a heteroaryl group having 5 to 10 ring atoms (i.e., a 5- to 10-membered heteroaryl group), and more preferably a heteroaryl group having 5 or 6 ring atoms (i.e., a 5- or 6-membered heteroaryl group).
  • Non-limiting examples of the monocyclic heteroaryl include: furyl, thienyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiadiazolyl, imidazolyl , pyrazolyl, triazolyl, tetrazolyl, furazyl, pyrrolyl, N-alkylpyrrolyl, pyridyl, pyrimidinyl, pyridonyl, N-alkylpyrrolyl ridinone (such as etc.), pyrazinyl, pyridazinyl, etc.
  • Non-limiting examples of the polycyclic heteroaryl include: indolyl, indazolyl, quinolyl, isoquinolyl, quinoxalinyl, phthalazinyl, benzimidazolyl, benzothiophene base, quinazolinyl, benzothiazolyl, carbazolyl, etc.
  • the polycyclic heteroaryl also includes a monocyclic heteroaryl fused with one or more aryl groups, wherein the point of attachment is on the aromatic ring, and in this case, the number of ring atoms continues to represent a polycyclic heteroaryl ring The number of ring atoms in the system.
  • the polycyclic heteroaryl also includes a monocyclic heteroaryl fused to one or more of cycloalkyl or heterocyclyl, wherein the point of attachment is on the monocyclic heteroaromatic ring, and in this case, the ring
  • the number of atoms continues to represent the number of ring atoms in the polycyclic heteroaromatic ring system.
  • Non-limiting examples include:
  • Heteroaryl groups may be substituted or unsubstituted. When substituted, they may be substituted at any available point of attachment.
  • the substituents are preferably selected from D atoms, halogens, alkyl groups, alkoxy groups, haloalkyl groups, haloalkyl groups.
  • cycloalkylalkyl refers to an alkyl group substituted by one or more cycloalkyl groups, where cycloalkyl and alkyl are as defined above.
  • heterocyclylalkyl refers to an alkyl group substituted by one or more heterocyclyl groups, where heterocyclyl and alkyl are as defined above.
  • cycloalkyloxy refers to -O-cycloalkyl, where cycloalkyl is as defined above.
  • heterocyclyloxy refers to -O-heterocyclyl, where heterocyclyl is as defined above.
  • alkoxyalkyl refers to an alkyl group substituted with one or more alkoxy groups, where alkoxy and alkyl are as defined above.
  • haloalkyl refers to an alkyl group substituted with one or more halogens, where alkyl is as defined above.
  • haloalkoxy refers to an alkoxy group substituted with one or more halogens, where alkoxy is as defined above.
  • hydroxyalkyl refers to an alkyl group substituted with one or more hydroxyl groups, where alkyl is as defined above.
  • 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.
  • Hydrogen or H is also a hydrogen atom.
  • amino protecting group refers to an easily removable group introduced on the amino group in order to keep the amino group unchanged when other parts of the molecule react.
  • Non-limiting examples include: (trimethylsilyl)ethoxymethyl, tetrahydropyranyl, tert-butoxycarbonyl (Boc), benzyloxycarbonyl (Cbz), watmethoxycarbonyl (Fmoc), allyl Oxycarbonyl (Alloc), trimethylsilylethoxycarbonyl (Teoc), methoxycarbonyl, ethoxycarbonyl, phthalyl (Pht), p-toluenesulfonyl (Tos), trifluoroacetyl (Tfa), Trityl (Trt), 2,4-dimethoxybenzyl (DMB), acetyl, benzyl, allyl, p-methoxybenzyl, etc.
  • stereoisomer refers to isomers that have the same structure but different arrangements of atoms in space. It includes cis and trans (or Z and E) isomers, (-)- and (+)-isomers, (R)- and (S)-enantiomers, diastereomers isomers, (D)- and (L)-isomers, tautomers, atropisomers, conformational isomers and their mixtures (e.g. racemates, mixtures of diastereomers) . Additional asymmetric atoms may be present for substituents in the compounds of the present disclosure.
  • Optically active (-)- and (+)-isomers, (R)- and (S)-enantiomers, and (D)- and (L)-isomer Optically active (-)- and (+)-isomers, (R)- and (S)-enantiomers, and (D)- and (L)-isomer.
  • An isomer of a certain compound of the present disclosure can be prepared through asymmetric synthesis or chiral auxiliaries, or when the molecule contains basic functional groups (such as amino) or acidic functional groups (such as carboxyl), and appropriate optical Reactive acids or bases form diastereomeric salts, and then the diastereoisomers are resolved by conventional methods known in the art to obtain pure isomers. Furthermore, the separation of enantiomers and diastereomers is usually accomplished by chromatography.
  • the bond Indicates that the configuration is not specified, i.e. if there are chiral isomers in the chemical structure, the bond can be or both and Two configurations. For all carbon-carbon double bonds, even if only one configuration is named, both the Z and E forms are included.
  • tautomer or tautomeric form
  • tautomer refers to a structural isomer that exists in equilibrium and is readily converted from one isomeric form to another. It includes all possible tautomers, either as single isomers or as mixtures of said tautomers in any proportion. Non-limiting examples include: keto-enol, imine-enamine, lactam-lactam, and the like. An example of a lactam-lactimide equilibrium is shown below:
  • the compounds of the present disclosure include all suitable isotopic derivatives of the compounds thereof.
  • isotopic derivative refers to a compound in which at least one atom is replaced by an atom with the same atomic number but a different atomic mass.
  • isotopes that may be incorporated into the compounds of the present disclosure include stable and radioactive isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine, iodine, and the like, such as 2 H (deuterium, D), respectively.
  • deuterated drugs Compared with non-deuterated drugs, deuterated drugs have the advantages of reducing side effects, increasing drug stability, enhancing efficacy, and extending the biological half-life of the drug. All variations in the isotopic composition of the compounds of the present disclosure, whether radioactive or not, are included within the scope of the present disclosure.
  • Each available hydrogen atom connected to a carbon atom can be independently replaced by a deuterium atom, where the replacement of deuterium can be partial or complete.
  • the replacement of partial deuterium means that at least one hydrogen is replaced by at least one deuterium.
  • deuterium D When a position is specifically designated as deuterium D, that 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 natural abundance greater than deuterium may be at least 1000 times the abundance of deuterium (i.e., at least 15% deuterium incorporation), at least 2000 times the abundance of deuterium (i.e., at least 30% deuterium incorporation) , at least 3000 times the abundance of deuterium (i.e. at least 45% deuterium incorporation), at least 3340 times the abundance of deuterium (i.e.
  • deuterium incorporation at least 50.1% deuterium incorporation
  • at least 3500 times the abundance of deuterium i.e. at least 50.1% deuterium incorporation) 52.5% deuterium incorporation
  • at least 4000 times the abundance of deuterium i.e. at least 60% deuterium incorporation
  • at least 4500 times the abundance of deuterium i.e.
  • deuterium incorporation at least 67.5% deuterium incorporation
  • at least 5000 times the abundance of deuterium Abundance of deuterium i.e., at least 75% deuterium incorporation
  • at least 6466.7 times the abundance of deuterium i.e., at least 97% deuterium incorporation
  • C 1-6 alkyl optionally substituted by halogen or cyano group includes the case where the alkyl group is substituted by halogen or cyano group and the case where the alkyl group is not substituted by halogen or cyano group.
  • Substituted or “substituted” means that one or more hydrogen atoms in a group, preferably 1 to 6, more preferably 1 to 3 hydrogen atoms, are independently substituted with a corresponding number of substituents.
  • a person skilled in the art will be able to determine possible or impossible substitutions without undue effort (either experimentally or theoretically).
  • an amino or hydroxyl group with a 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 a pharmaceutically acceptable salt thereof, together with other chemical components, as well as other ingredients such as pharmaceutically acceptable carriers and excipients.
  • the purpose of pharmaceutical compositions is to facilitate administration to living organisms and facilitate the absorption of active ingredients to exert biological activity.
  • “Pharmaceutically acceptable salts” refer to salts of compounds of the present disclosure, which may be selected from inorganic salts or organic salts. This type of salt is safe and effective when used in mammals, and has due biological activity. They can be prepared individually 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 as well as organic acids.
  • pharmaceutically acceptable refers to compounds, materials, compositions and/or dosage forms which, within the scope of reasonable medical judgment, are suitable for contact with patient tissue without undue toxicity, irritation, allergic reaction or other problems or complications, has a reasonable benefit/risk ratio, and is effective for the intended use.
  • the preparation method of the compound represented by the general formula (II-1) or a pharmaceutically acceptable salt thereof of the present disclosure includes the following steps:
  • the compound represented by the general formula (II-1A) or a salt thereof (preferably a hydrochloride) and the compound represented by the general formula (II-1B) or a salt thereof are subjected to a condensation reaction in the presence of a condensing agent under alkaline conditions. , obtain the compound represented by general formula (II-1) or a pharmaceutically acceptable salt thereof; wherein,
  • R A is a hydrogen atom
  • R 1 , R 2 , R 3 , R 4 , R Z1 , R Z2 , R L3 , R L4 , n2, ring J 1' , J 3 , J 4 , J 5 , J 6 , Q 1 , Q 2 , Q 3 , Q 4 and Q 5 are as defined in general formula (II-1).
  • the preparation method of the compound represented by the general formula (II-2) or a pharmaceutically acceptable salt thereof of the present disclosure includes the following steps:
  • the compound represented by the general formula (II-1A) or a salt thereof (preferably a hydrochloride) and the compound represented by the general formula (II-2B) or a salt thereof are subjected to a condensation reaction in the presence of a condensing agent under alkaline conditions. , obtain the compound represented by general formula (II-2) or a pharmaceutically acceptable salt thereof; wherein,
  • R A is a hydrogen atom
  • R 1 , R 2 , R 3 , R 4 , R Z1 , R Z2 , R L3 , R L4 , n2, ring J 1' , J 3 , J 4 , J 5 , J 6 , R Z4 , Q 1 , Q 2 , Q 3 and Q 4 are as defined in general formula (II-2).
  • the preparation method of the compound represented by the general formula (II-3) or a pharmaceutically acceptable salt thereof of the present disclosure includes the following steps:
  • R 1 , R 2 , R 3 , R 4 , R Z1 , R Z2 , R L3 , R L4 , J 1 , q, ring J 3' , J 4 , J 5 , Z 4 , R Q , R J , s , n2 and x are as defined in general formulas (II-3) and (II-2A).
  • the preparation method of the compound represented by the general formula (I') or a pharmaceutically acceptable salt thereof of the present disclosure includes the following steps:
  • the compound represented by the general formula (I'A) or a salt thereof (preferably the hydrochloride) and the compound represented by the general formula (I'B) or a salt thereof are subjected to a condensation reaction under alkaline conditions in the presence of a condensing agent.
  • ring A, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , ring J 1' , J 3 to J 6 and n5 are as in the general formula (I') definition.
  • the preparation method of the compound represented by the general formula (I'-1) or a pharmaceutically acceptable salt thereof of the present disclosure includes the following steps:
  • the compound represented by the general formula (I'A) or a salt thereof (preferably the hydrochloride) and the compound represented by the general formula (II'-2B) or a salt thereof are condensed under alkaline conditions in the presence of a condensing agent.
  • the reaction yields a compound represented by general formula (I'-1) or a pharmaceutically acceptable salt thereof,
  • R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , n5, ring J 1' , J 3 to J 6 , R Q , R Z4 , Z 8 and j are as follows It is defined in the general formula (I'-1).
  • the preparation method of the compound represented by the general formula (I'-2) or a pharmaceutically acceptable salt thereof of the present disclosure includes the following steps:
  • the compound represented by the general formula (I'A) or a salt thereof (preferably the hydrochloride) and the compound represented by the general formula (I'-2B) or a salt thereof are condensed under alkaline conditions in the presence of a condensing agent.
  • the reaction yields a compound represented by general formula (I'-2) or a pharmaceutically acceptable salt thereof,
  • R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , L 5 , L 6 , n5, ring J 1' , J 3 to J 6 , R Q , R Z7 and k are as in the general formula ( As defined in I'-2).
  • the preparation method of the compound represented by the general formula (IM) of the present disclosure or a pharmaceutically acceptable salt thereof includes the following steps:
  • the compound represented by the general formula (IMA) or a salt thereof (preferably a hydrochloride or trifluoroacetate) and the compound represented by the general formula (IMB) or a salt thereof are reduced under acidic conditions in the presence of a reducing agent.
  • the amination reaction yields the compound represented by the general formula (IM) or a pharmaceutically acceptable salt thereof,
  • ring A, ring C, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , n5, L 5 , L 6 , J 1 , q, J 4 to J 6 , R 03 and u are as follows It is defined in the general formula (IM).
  • the preparation method of the compound represented by the general formula (IN) of the present disclosure or a pharmaceutically acceptable salt thereof includes the following steps:
  • the compound represented by the general formula (INA) or its salt and the compound represented by the general formula (INB) or its salt (preferably the hydrochloride) undergo a reductive amination reaction under acidic conditions in the presence of a reducing agent to obtain the general formula ( The compound represented by IN) or a pharmaceutically acceptable salt thereof,
  • ring A, ring B, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , n5, L 5 , L 6 , J 1 , J 2 , q, J 4 to J 6 , R 03 and u are as defined in general formula (IN).
  • the preparation method of the compound represented by the general formula (II-3') or a pharmaceutically acceptable salt thereof of the present disclosure includes the following steps:
  • ring C, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , n5, L 5 , L 6 , J 1 , J 2 , R 03 , u, R J5 , R J6 , t, R J , R Q , Z 4 and s are as defined in general formula (II-3').
  • the preparation method of the compound represented by the general formula (III-1) or a pharmaceutically acceptable salt thereof of the present disclosure includes the following steps:
  • the compound represented by the general formula (IMA) or a salt thereof (preferably a hydrochloride or trifluoroacetate) and the compound represented by the general formula (III-1B) or a salt thereof occur under acidic conditions in the presence of a reducing agent
  • the reductive amination reaction obtains the compound represented by the general formula (III-1) or a pharmaceutically acceptable salt thereof,
  • ring C, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , n5, L 5 , L 6 , J 1 , R 03 , u, q, J 4 to J 6 , R Q , Z 4 and s are as defined in general formula (III-1).
  • the preparation method of the compound represented by the general formula (III-2) or a pharmaceutically acceptable salt thereof of the present disclosure includes the following steps:
  • ring B, R 1 to R 4 , R Z1 , R Z2 , R L3 , R L4 , n5, L 5 , L 6 , J 1 , J 2 , q, R 03 , u, J 4 to J 6 , R Q , Z 4 and s are as defined in general formula (III-2).
  • the reagents that provide alkaline conditions in the above condensation reaction include organic bases and inorganic bases.
  • the organic bases include but are not limited to triethylamine, pyridine, diisopropylethylamine (i.e. N,N-diisopropylethylamine).
  • 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 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'-tetramethylurea tetrafluoroborate, 1-hydroxybenzotriazole, 1-hydroxy-7-azobenzotriazole, O-benzotriazole-N,N,N',N'-tetramethylurea hexafluorophosphate, 2-(7-azobenzotriazole Azole)-N,N,N',N'-tetramethylurea hexafluorophosphate (HATU), 2-(7-oxybenzotriazole)-N,N,N',N'-tetramethylurea Methylurea hexafluor
  • reagents that provide weakly acidic conditions include but are not limited to acetic acid (acetic acid), Ti(i-PrO) 3 , BF 3 Et 2 O, TiCl 4 , etc.
  • reagents that provide acidic conditions include but are not limited to hydrogen chloride, hydrogen chloride solution in 1,4-dioxane, hydrochloric acid solution in 1,4-dioxane, trifluoroacetic acid, formic acid, acetic acid, and glacial acetic acid.
  • hydrochloric acid concentrated sulfuric acid, methanesulfonic acid, nitric acid, phosphoric acid, p-phenylsulfonic acid, titanium tetrachloride, Me 3 SiCl and TMSOTf; preferably acetic acid or glacial acetic acid.
  • the reagent providing acidic conditions can be an acid reagent added directly; and/or an alkaline salt reagent is added, and the acid brought in by the alkaline salt and the acidic salt form of the raw material is added during the reaction.
  • the alkaline salt includes but is not limited to sodium acetate, anhydrous sodium acetate, preferably anhydrous sodium acetate; the acidic salt of the raw material is preferably a hydrochloride or trifluoroacetate; specifically, when the raw material is in the form of an acidic salt
  • the way to provide acidic conditions can be: adding the reagent sodium acetate, and during the reaction process, sodium acetate and the acid in the raw material together form acetic acid.
  • the reducing agent includes but is not limited to sodium triacetoxyborohydride, sodium borohydride, lithium borohydride, sodium acetate borohydride, sodium cyanoborohydride, sodium acetylborohydride, etc., preferably triacetyl borohydride.
  • sodium oxyborohydride or sodium cyanoborohydride is preferred.
  • the reaction of the above steps is preferably carried out in a solvent.
  • the solvents used include but are not limited to: pyridine, 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-dibromo Ethane and its mixtures.
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) or/and mass spectrometry (MS). NMR shifts ( ⁇ ) are given in units of 10 -6 (ppm). NMR is measured using Bruker AVANCE-400 nuclear magnetic instrument or Bruker AVANCE NEO 500M.
  • the measurement solvents are deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), and deuterated methanol (CD 3 OD).
  • the internal standard is tetramethylsilane (TMS).
  • MS was measured using Agilent 1200/1290DAD-6110/6120Quadrupole MS liquid mass spectrometer (manufacturer: Agilent, MS model: 6110/6120Quadrupole MS).
  • HPLC High performance liquid chromatography
  • High-performance liquid phase preparation uses Waters 2545-2767, Waters 2767-SQ Detecor2, Shimadzu LC-20AP and Gilson GX-281 preparative chromatographs.
  • Chiral preparation uses Shimadzu LC-20AP preparative chromatograph.
  • CombiFlash rapid preparation instrument uses Combiflash Rf200 (TELEDYNE ISCO).
  • Thin layer chromatography silica gel plates use Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plates.
  • the specifications of silica gel plates used in thin layer chromatography (TLC) are 0.15mm ⁇ 0.2mm.
  • the specifications used for thin layer chromatography separation and purification products are 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 IC 50 value were measured using NovoStar microplate reader (BMG Company, Germany).
  • the known starting materials of the present disclosure can be synthesized by methods known in the art, or can be purchased from ABCR GmbH & Co. KG, Acros Organics, Aldrich Chemical Company, Shaoyuan Chemical Technology (Accela ChemBio Inc), Dari Chemicals, Bailingwei, Shanghai Bide Pharmaceutical and other companies.
  • Argon atmosphere or nitrogen atmosphere means that the reaction bottle is connected to an argon or nitrogen balloon with a volume of about 1L.
  • the hydrogen atmosphere refers to the reaction bottle connected to a hydrogen balloon with a volume of about 1L.
  • the pressurized hydrogenation reaction uses Parr 3916EKX hydrogenator and Clear Blue QL-500 hydrogen generator or HC2-SS hydrogenator.
  • the hydrogenation reaction is usually evacuated, filled with hydrogen, and repeated three times.
  • the microwave reaction uses CEM Discover-S 908860 microwave reactor.
  • the solution refers to an aqueous solution.
  • the reaction temperature is room temperature, which is 20°C to 30°C.
  • the reaction process in the embodiment is monitored by thin layer chromatography (TLC).
  • TLC thin layer chromatography
  • the developing agent used in the reaction, the column chromatography eluent system and the thin layer chromatography developing agent system used to purify the compound include: A: Two Methyl chloride/methanol system, B: n-hexane/ethyl acetate system, the volume ratio of the solvent is adjusted according to the polarity of the compound, and a small amount of alkaline or acidic reagents such as triethylamine and acetic acid can also be added for adjustment.
  • reaction solution was purified by high performance liquid phase preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 65%-80%, flow rate: 30 mL/min) to obtain the title compound 1 (racemic mixture, 15 mg, yield: 35%).
  • 4-(L-Alanyl)piperazine-1-carboxylic acid benzyl ester 5a (3.56g, 10.86mmol) was added in sequence to a 250mL single-neck bottle, using the compound 4j disclosed on page 42 of the specification in the patent application "WO2014057266" Prepared by the method), 2-(2,6-dioxopiperidin-3-yl)-4-fluoroindoline-1,3-dione 5b (2.5g, 9.05mmol, Shanghai Haohong Biopharmaceutical Technology Co., Ltd.), dimethyl sulfoxide (50 mL) and N, N-diisopropylethylamine (3.51 g, 27.15 mmol), and the temperature was raised to 100°C for 16 hours.
  • reaction solution was concentrated under reduced pressure, 20 mL of water was added, the pH of the solution was adjusted to about 8 with saturated sodium bicarbonate solution, extracted with ethyl acetate (50 mL ⁇ 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain The residue was purified by silica gel column chromatography using eluent system A to obtain 5 g of the title compound (900 mg, yield: 54%).
  • reaction solution was concentrated and filtered, and purified by high-performance liquid phase preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, acetonitrile gradient: 67%-82%, flow rate: 30 mL/min) to obtain Title compound 5 (diastereomeric mixture, 1:1, 27 mg, yield: 43%).
  • reaction solution was purified by high performance liquid phase preparative chromatography (Waters2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 50%-62%, flow rate: 30 mL/min) to obtain the title compound 6 ( Racemic mixture, 17 mg, yield: 44%).
  • reaction solution was purified by high performance liquid phase preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 53%-78%, flow rate: 30 mL/min) to obtain the title compound 7 (racemic mixture, 85 mg, yield: 37%).
  • reaction solution was purified by high performance liquid phase preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 53%-57%, flow rate: 30 mL/min) to obtain the title compound 9 (racemic mixture, 15 mg, yield: 19%).
  • reaction solution was purified by high performance liquid phase preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 50%-70%, flow rate: 30 mL/min) to obtain the title compound 10 (Four isomer mixture, 1:1:1:1, 15 mg, yield: 20%).
  • reaction solution was purified by high performance liquid phase preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 55%-59%, flow rate: 30 mL/min) to obtain the title compound 11 (Four isomer mixture, 1:1:1:1, 17 mg, yield: 23%).
  • reaction solution is concentrated, and the resulting residue is subjected to high performance liquid phase preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 40%-60%, flow rate: 30 mL/min) and purified to obtain the title compound 12f (60 mg, yield: 47%).
  • reaction solution was purified by high performance liquid phase preparative chromatography (Waters2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 45%-52%, flow rate: 30 mL/min) to obtain the title compound 12 ( Racemic mixture, 16 mg, yield: 23%).
  • reaction solution was purified by high performance liquid phase preparative chromatography (Waters2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 49%-54%, flow rate: 30 mL/min) to obtain the title compound 13 ( Racemic mixture, 25 mg, yield: 17%).
  • reaction solution was purified by high performance liquid phase preparative chromatography (Waters2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 49%-56%, flow rate: 30 mL/min) to obtain the title compound 14 ( Racemic mixture, 25 mg, yield: 18%).
  • reaction solution was purified by high performance liquid phase preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 55%-75%, flow rate: 30 mL/min) to obtain the title compound 17 (racemic mixture, 10 mg, yield: 17%).
  • Cyclopropylamine 18a (250 mg, 4.38 mmol, Shaoyuan Chemical Technology) was dissolved in dichloromethane (10 mL), and N, N-diisopropylethylamine (565 mg, 4.38 mmol) and pyridine (346 mg, 4.38 mmol) were added. . Add methylsulfonyl chloride (500 mg, 4.38 mmol) under ice bath conditions and react for 12 hours. After the reaction was completed, the solvent was removed under reduced pressure and dried under vacuum to obtain the crude product title compound 18b (590 mg, yield: 99%). The crude product was directly used in the next reaction without purification.
  • reaction solution was purified by high performance liquid phase preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 55%-75%, flow rate: 30 mL/min) to obtain the title compound 18 (racemic mixture, 15 mg, yield: 21%).
  • N,N-dimethylformamide (2mL), compound 15g (45mg, 0.054mmol), (2-(2,6-dioxopiperidin-3-yl)-1, 3-Dioxoisoindolin-4-yl)-L-alanine 19a (21 mg, 0.061 mmol, prepared by the method disclosed in the patent application "Compound 235 on page 419 of the specification in WO2020051235A1", not for mixture of enantiomers), O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethylurea hexafluorophosphate (41 mg, 0.11 mmol), ice Under the bath, slowly add N,N-diisopropylethylamine (30 mg, 0.078 mmol) and react for 20 minutes.
  • reaction solution was filtered and purified by high performance liquid phase preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 45%-65%, flow rate: 30 mL/min) to obtain the title Compound 19 (mixture of 2 diastereomers, ratio 1:1, 35 mg, yield: 60%).
  • N,N-dimethylformamide 4mL
  • compound 19a 150mg, 0.43mmol
  • 4-(dimethoxymethyl)piperidine 105mg, 0.66mmol
  • O- (7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethylurea hexafluorophosphate 248mg, 0.65mmol
  • N,N- Diisopropylethylamine 225 mg, 1.74 mmol
  • the reaction solution was concentrated, and the residue was purified by silica gel column chromatography using eluent system A to obtain the title compound 20a (a mixture of two diastereomers, ratio: 1:1, 210 mg, yield: 99%).
  • reaction solution was subjected to high performance liquid phase preparative chromatography (Waters 2545, elution system: 10 mmol/L ammonium bicarbonate aqueous solution and acetonitrile, gradient of acetonitrile: 49%-56%, flow rate: 30 mL/min) to obtain the title compound 20 (2 A mixture of diastereomers, ratio: 1:1, 48 mg, yield: 52%).
  • reaction solution was cooled to room temperature, filtered through diatomaceous earth, and the filtrate was concentrated to obtain The residue was purified by silica gel column chromatography using eluent system B to obtain the title compound 21b (2.6 g, yield: 91%).

Landscapes

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

Abstract

本公开涉及抑制并诱导EGFR降解的大环类化合物、其制备方法及其在医药上的应用。具体而言,本公开涉及一种通式(I)所示的大环类化合物、其制备方法及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。

Description

抑制并诱导EGFR降解的大环类化合物、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(I)所示的大环类化合物、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。
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等。
发明内容
本公开的目的在于提供一种通式(I)所示的化合物,或其可药用的盐:
其中:
R1选自氢原子、卤素、烷基、烷氧基、羟基、烯基、炔基、氰基、-NR21R22、-亚烷基NR21R22、-C(O)NR21R22、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、亚烷基、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;
R2选自-P(O)R23R24、-SO2R23、-SO2N(R23R24)、-N(R23)SO2R24、卤素、烷基、烷氧基和氨基;
或,R1、R2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;
R3选自氢原子、卤素、烷基、烷氧基、羟基、烯基、炔基、氰基、-NR31R32、-亚烷基NR31R32、-C(O)NR31R32、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的被一个或多个R01所取代;
R4选自氢原子、卤素、烷基、烯基、烷氧基和卤代烷基;
或,R3、R4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;
R21、R22、R23、R24、R31和R32相同或不同,且各自独立地选自氢原子、烷基、烷氧基、氨基、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、氨基、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的被一个或多个R01所取代;
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、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、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、烷氧基、羟基、烯基、炔基、氰基、-C(O)Rc、-NRaRb、-C(O)NRaRb、-亚烷基Rc、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、亚烷基、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R02所取代;
或,RZ1、RZ2及与其连接的碳原子一起形成任选被一个或多个R02所取代的环烷基、杂环基、芳基或杂芳基;
Ra和Rb相同或不同,且各自独立地选自氢原子、卤素、烷基、羟基、烯基、炔基、氨基、环烷基、杂环基、杂环基烷基、芳基和杂芳基;所述的烷基、烯基、炔基、环烷基、杂环基、杂环基烷基、芳基和杂芳基各自独立地任选被一个或多个R02所取代;
Rc选自氢原子、卤素、烷基、烷氧基、羟基、烯基、炔基、氰基、氨基、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、氨基、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、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基和杂芳基;
L1选自O、S、NRL、C(O)、C(O)NRL、NRLC(O)、NRLC(O)NRL和(CRL1RL2)m
L2、L3、L4、L5和L6相同或不同,且各自独立地选自键、O、S、NRL、C(O)、C(O)NRL、NRLC(O)、NRLC(O)NRL和(CRL3RL4)n
RL1、RL2、RL3和RL4各自独立地选自氢原子、卤素、烷基、烷氧基和卤代烷基;或,RL3、RL4及与其相连的同一碳原子一起形成环烷基或杂环基;
RL选自氢原子、烷基和卤代烷基;
J1选自环烷基、杂环基、杂环基-杂环基、芳基、杂芳基、O、C(O)、S(O)r、 (CRJ1RJ2)p和N(RJ);所述的环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R03所取代;
J2、J3、J4和J5相同或不同,且各自独立地选自S、O(CRJ3RJ4)q、(CRJ3RJ4)qO、C(O)(CRJ3RJ4)q、(CRJ3RJ4)qC(O)、C(O)N(RJ)、N(RJ)C(O)、N(RJ)C(O)N(RJ)、S(O)r、S(O)rN(RJ)、N(RJ)S(O)r、N(RJ)S(O)rN(RJ)、(CRJ3RJ4)q、N(RJ)(CRJ3RJ4)q、(CRJ3RJ4)qN(RJ)、亚烯基、亚炔基、环烷基、杂环基、亚烷基-杂环基、杂环基-亚烷基、芳基和杂芳基;所述的亚烯基、亚炔基、环烷基、亚烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R03所取代;
J6选自O、S、S(O)r、(CRJ5RJ6)t、(CRJ5RJ6)tN(RJ)、N(RJ)(CRJ5RJ6)t、C(O)(CRJ5RJ6)t、(CRJ5RJ6)tC(O)、C(O)N(RJ)、N(RJ)C(O)、N(RJ)C(O)N(RJ)、亚烯基、亚炔基、环烷基和杂环基;所述的亚烯基、亚炔基、环烷基和杂环基各自独立地任选被一个或多个R03所取代;
RJ选自氢原子、烷基和卤代烷基;
RJ1、RJ2、RJ3、RJ4、RJ5和RJ6各自独立地选自氢原子、卤素、烷基、烷氧基、羟基、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、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R03所取代;
或,RJ1、RJ2及与其相连的同一碳原子一起形成任选被一个或多个R03所取代的环烷基或杂环基;或,RJ3、RJ4及与其相连的同一碳原子一起形成任选被一个或多个R03所取代的环烷基或杂环基;或,RJ5、RJ6及与其相连的同一碳原子一起形成任选被一个或多个R03所取代的环烷基或杂环基;
R03选自氧代基、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基和杂芳基;
环A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,其它各自独立地为N或CRQ
RQ相同或不同,且各自独立地选自氢原子、卤素、烷基、烷氧基、羟基、烯基、炔基、氰基和-(CH2)t1NRdRe;所述的烷基、烷氧基、烯基和炔基各自独立地任选被一个或多个R04所取代;
Rd和Re各自独立地选自氢原子、卤素、烷基、环烷基和杂环基;
Z4为C(RZ)2或C(O);RZ相同或不同,且各自独立地选自氢原子、卤素、烷基 和氨基,或两个RZ与其相连的碳原子一起形成环烷基或杂环基;
Z8为N或CRZ8
RZ4和RZ7各自独立地选自氢原子、烷基和卤代烷基;
RZ8选自氢原子、烷基、烷氧基和卤代烷基;
R04选自氧代基、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基和杂芳基;
m、n、p、q、t和t1各自独立地为0、1、2、3、4、5或6;
r为1或2。
本公开的目的在于提供一种通式(I)所示的化合物,或其可药用的盐;
其中:
R1选自氢原子、卤素、烷基、烷氧基、羟基、烯基、炔基、氰基、-NR21R22、-亚烷基NR21R22、-C(O)NR21R22、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、亚烷基、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;
R2选自-P(O)R23R24、-SO2R23、-SO2N(R23R24)、-N(R23)SO2R24、卤素、烷基、烷氧基和氨基;
或,R1、R2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;
R3选自氢原子、卤素、烷基、烷氧基、羟基、烯基、炔基、氰基、-NR31R32、-亚烷基NR31R32、-C(O)NR31R32、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的被一个或多个R01所取代;
R4选自氢原子、卤素、烷基、烯基、烷氧基和卤代烷基;
或,R3、R4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;
R21、R22、R23、R24、R31和R32相同或不同,且各自独立地选自烷基、烷氧基、氨基、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、氨基、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的被一个或多个R01所取代;
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、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、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、烷氧基、羟基、烯基、炔基、氰基、-C(O)Rc、-NRaRb、-C(O)NRaRb、-亚烷基Rc、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、亚烷基、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R02所取代;
或,RZ1、RZ2及与其连接的碳原子一起形成任选被一个或多个R02所取代的环烷基、杂环基、芳基或杂芳基;
Ra和Rb相同或不同,且各自独立地选自氢原子、卤素、烷基、羟基、烯基、炔基、氨基、环烷基、杂环基、杂环基烷基、芳基和杂芳基;所述的烷基、烯基、炔基、环烷基、杂环基、杂环基烷基、芳基和杂芳基各自独立地任选被一个或多个R02所取代;
Rc选自氢原子、卤素、烷基、烷氧基、羟基、烯基、炔基、氰基、氨基、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、氨基、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、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基和杂芳基;
L1选自O、S、NRL、C(O)、C(O)NRL、NRLC(O)、NRLC(O)NRL和(CRL1RL2)m
L2、L3、L4、L5和L6相同或不同,且各自独立地选自键、O、S、NRL、C(O)、C(O)NRL、NRLC(O)、NRLC(O)NRL和(CRL3RL4)n
RL1、RL2、RL3和RL4各自独立地选自氢原子、卤素、烷基、烷氧基和卤代烷基;或,RL3、RL4及与其相连的同一碳原子一起形成环烷基或杂环基;
RL选自氢原子、烷基和卤代烷基;
J1选自环烷基、杂环基、杂环基-杂环基、芳基、杂芳基、O、C(O)、S(O)r、(CRJ1RJ2)p和N(RJ);所述的环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R03所取代;
J2、J3、J4和J5相同或不同,且各自独立地选自S、O(CRJ3RJ4)q、(CRJ3RJ4)qO、C(O)(CRJ3RJ4)q、(CRJ3RJ4)qC(O)、C(O)N(RJ)、N(RJ)C(O)、N(RJ)C(O)N(RJ)、S(O)r、S(O)rN(RJ)、N(RJ)S(O)r、N(RJ)S(O)rN(RJ)、(CRJ3RJ4)q、N(RJ)(CRJ3RJ4)q、(CRJ3RJ4)qN(RJ)、亚烯基、亚炔基、环烷基、杂环基、亚烷基-杂环基、杂环基-亚 烷基、芳基和杂芳基;所述的亚烯基、亚炔基、环烷基、亚烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R03所取代;
J6选自O、S、S(O)r、(CRJ5RJ6)t、(CRJ5RJ6)tN(RJ)、N(RJ)(CRJ5RJ6)t、C(O)(CRJ5RJ6)t、(CRJ5RJ6)tC(O)、C(O)N(RJ)、N(RJ)C(O)、N(RJ)C(O)N(RJ)、亚烯基、亚炔基、环烷基和杂环基;所述的亚烯基、亚炔基、环烷基和杂环基各自独立地任选被一个或多个R03所取代;
RJ选自氢原子、烷基和卤代烷基;
RJ1、RJ2、RJ3、RJ4、RJ5和RJ6各自独立地选自氢原子、卤素、烷基、烷氧基、羟基、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、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R03所取代;
或,RJ1、RJ2及与其相连的同一碳原子一起形成任选被一个或多个R03所取代的环烷基或杂环基;或,RJ3、RJ4及与其相连的同一碳原子一起形成任选被一个或多个R03所取代的环烷基或杂环基;或,RJ5、RJ6及与其相连的同一碳原子一起形成任选被一个或多个R03所取代的环烷基或杂环基;
R03选自氧代基、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基和杂芳基;
环A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,其它各自独立地为N或CRQ
RQ相同或不同,且各自独立地选自氢原子、卤素、烷基、烷氧基、羟基、烯基、炔基、氰基和-(CH2)t1NRdRe;所述的烷基、烷氧基、烯基和炔基各自独立地任选被一个或多个R04所取代;
Rd和Re各自独立地选自氢原子、卤素、烷基、环烷基和杂环基;
Z4为C(RZ)2或C(O);RZ相同或不同,且各自独立地选自氢原子、卤素、烷基和氨基,或两个RZ与其相连的碳原子一起形成环烷基或杂环基;
Z8为N或CRZ8
RZ4和RZ7各自独立地选自氢原子、烷基和卤代烷基;
RZ8选自氢原子、烷基、烷氧基和卤代烷基;
R04选自氧代基、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基和杂芳基;
m、n、p、q、t和t1各自独立地为0、1、2、3、4、5或6;
r为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:
n5为1、2、3、4、5、6或7;
R1、R2、R3、R4、RL3、RL4、L5、L6、RZ1和RZ2如通式(I)中所定义;
在一些实施方案中, 在一些实施方案中,在一些实施方案中,
n2为1、2、3、4、5、6、7、8、9或10;n5为1、2、3、4、5、6或7;
RL3和RL4各自独立地选自氢原子、C1-6烷基和C1-6烷氧基;
R1、R2、R3、R4、RZ1和RZ2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)所示的化合物或其可药用的盐,其中,
n3和n4各自独立地为0、1、2、3或4;
L1选自O、S和CH2
L3和L6各自独立地选自NH、C(O)、C(O)NH、NHC(O)、CH2
R1、R2、R3、R4、RZ1和RZ2如通式(I)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,所述的式(Ia)所示的化合物或其可药用的盐,其中,n3为1或2,n4为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,L1为O;L2、L4和L5各自独立地选自键、CH2、CH2CH2、C(CH3)2L3选自-C(O)NH-、-NHC(O)-和-CH2-;L6选自CH2、C(CH3)2在一些实施 方案中,L1为O;L2、L3、L4、L5和L6分别为CH2
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,-L1-L2-L3-L4-L5-L6-为-O-(CRL3RL4)n2-或-O-(CRL3RL4)n5-O-C(O)-;在一些实施方案中,-L1-L2-L3-L4-L5-L6-为-O-(CRL3RL4)n2-;在一些实施方案中,-L1-L2-L3-L4-L5-L6-为-O-(CRL3RL4)n5-O-C(O)-;n2为2、3、4、5、6、7或8;n5为1、2、3、4、5、6或7;RL3和RL4各自独立地选自氢原子、C1-6烷基和C1-6烷氧基。
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,L1为O,-L2-L3-L4-为-(CH2)n5-,n5为2、3或4。
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,L1为O,-L2-L3-L4-为C1-6亚烷基,L5为CH2或O,L6为CH2或C(O);在一些实施方案中,L1为O,-L2-L3-L4-为-(CH2)3-或-(CH2)4-,L5为CH2或O,L6为CH2或C(O);在一些实施方案中,L1为O,-L2-L3-L4-为-(CH2)n5-,n5为2、3、4、5或6,L5为O,L6为C(O)。
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,L1为O。
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,-L2-L3-L4-为C1-6亚烷基;在一些实施方案中,-L2-L3-L4-为-(CH2)n5-,n5为2、3、4、5或6;在一些实施方案中,-L2-L3-L4-为-(CH2)3-或-(CH2)4-。
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,其为通式(I’)所示的化合物或其可药用的盐:
其中,
环J1’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;
n5为1、2、3、4、5、6或7;
环A、R1至R4、RZ1、RZ2、RL3、RL4、L5、L6、J3至J6和R03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)所示的化合物或其可药用的盐,其为通式(IM)所示的化合物或其可药用的盐:
其中,
环C为含氮杂环基;
n5为1、2、3、4、5、6或7;u为0、1、2、3、4、5或6;
环A、R1至R4、RZ1、RZ2、RL3、RL4、L5、L6、J1、q、J4至J6和R03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)所示的化合物或其可药用的盐,其为通式(IN)所示的化合物或其可药用的盐:
其中,
环B为含氮杂环基;
n5为1、2、3、4、5、6或7;u为0、1、2、3、4、5或6;
环A、R1至R4、RZ1、RZ2、RL3、RL4、L5、L6、J1、J2、q、J4至J6和R03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’)、(IM)或(IN)所示的化合物或其可药用的盐,其中环A选自:

Q1、Q2、Q3、Q4和Q5各自独立地为N或CRQ;RQ选自氢原子、卤素、C1-6烷基、C1-6烷氧基、羟基、C2-6烯基、C2-6炔基、氰基、氨基、NH C1-6烷基和N(C1-6烷基)2;所述的C1-6烷基、C1-6烷氧基、C2-6烯基、C2-6炔基和氨基各自独立地任选被一个或多个R04所取代;R04选自氧代基、卤素、C1-6烷基、C1-6烷氧基、C1-6卤代烷基、羟基、C1-6羟烷基、氰基、氨基、硝基、3至8元环烷基、4至12元杂环基、4至12元杂环基氧基、6至10元芳基和5至14元杂芳基;RZ4如通式(I)中所定义;
在一些实施方案中,环A选自:
Z4为CH2或C(O);RZ4选自氢原子、C1-6烷基和C1-6卤代烷基;RZ7为氢原子或C1-6烷基;RQ各自独立地选自氢原子、卤素、C1-6烷基、C1-6卤代烷基、C1-6烷氧基、C1-6卤代烷氧基、氰基和氨基。
在本公开的一些实施方案中,所述的通式(I)、(I’)、(IM)或(IN)所示的化合物或其可药用的盐,其中环A选自:
在一些实施方案中,环A选自: 在一些实施方案中,环A为
Z8为CH或氮原子;Z4为CH2或C(O);RZ4选自氢原子、C1-6烷基和C1-6卤代烷基;RZ7为氢原子或C1-6烷基;RQ选自氢原子、卤素、C1-6烷基、C1-6卤代烷基、C1-6烷氧基、C1-6卤代烷氧基、氰基和氨基;
在一些实施方案中,环A选自: RQ各自独立地选自氢原子、卤素、C1-6烷基、C1-6卤代烷基、C1-6卤代烷氧基和氰基;
在一些实施方案中,环A选自:
RQ各自独立地选自氢原子、卤素、C1-6烷基、C1-6卤代烷基和C1-6卤代烷氧基;
在一些实施方案中,环A选自:
Q1、Q2、Q3、Q4、Q5和RZ4如通式(I)所定义;
在一些实施方案中,环A选自:
在一些实施方式中,所述的通式(I)、(I’)、(IM)或(IN)所示的化合物或其可药 用的盐,其中环A选自:
在一些实施方案中,环A选自:Z8为CH或氮原子;Z4为CH2或C(O);RZ7为氢原子或C1-6烷基;RZ4为C1-6烷基;RQ为卤素或C1-6烷基;k为0、1、2、3或4;j为0、1、2或3;s为0、1、2或3;
在一些实施方案中,环A选自:
在一些实施方案中,环A选自:
在一些实施方案中,环A选自:
在一些实施方案中,环A选自: 在一些实施方案中,环A为在一些实施方案中,环A选自
在本公开的一些实施方案中,所述的通式(I)、(I’)、(IM)、(IN)、(II-1)、(II-2)、(II-3)、(I’-1)、(I’-2)、(II-3’)、(III-1)或(III-2)所示的化合物或其可药用的盐,其中RQ为氢原子或卤素;在一些实施方案中,RQ为氢原子;在一些实施方案中,RQ为卤素,在一些实施方案中,RQ为氟。
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,其为通式(II-1)、(II-2)或(II-3)所示的化合物,或其可药用的盐:

其中,
n2为1、2、3、4、5、6、7、8、9或10;
环J1’为5至14元含氮杂环基;
环J3’为5至14元含氮杂环基,所述的5至14元含氮杂环基任选被1个或多个选自卤素、羟基、C1-6烷基和C1-6卤代烷基的取代基所取代;
RJ为氢原子或C1-6烷基;s为0、1或2;
R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、J1、q、J3、J4、J5、J6、Z4、RZ4、Q1、Q2、Q3、Q4、Q5和RQ如通式(I)所定义。
在本公开的一些实施方案中,所述的通式(I)、(I’)所示的化合物或其可药用的盐,其为通式(I’-1)或(I’-2)所示的化合物,或其可药用的盐:
其中,环J1’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;
n5为1、2、3、4、5、6或7;j为0、1或2;k为0、1或2;
R1至R4、RZ1、RZ2、RL3、RL4、L5、L6、J3至J6、RQ、RZ4、RZ7、Z8和R03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)所示的化合物或其可药用的盐,其为通式(II-3’)所示的化合物,或其可药用的盐:
其中,
环C为含氮杂环基;
n5为1、2、3、4、5、6或7;u为0、1、2、3、4、5或6;s为0、1、2或3;
R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、L5、L6、J1、J2、R03、RJ5、RJ6、t、RJ、RQ和Z4如通式(I)所定义。
在本公开的一些实施方案中,所述的通式(I)或(IM)所示的化合物或其可药用的盐,其为通式(III-1)所示的化合物,或其可药用的盐:
其中,
环C为含氮杂环基;
n5为1、2、3、4、5、6或7;
u为0、1、2、3、4、5或6;s为0、1、2或3;
R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、L5、L6、J1、R03、q、J4、J5、J6、RQ和Z4如通式(I)所定义。
在本公开的一些实施方案中,所述的通式(I)或(IN)所示的化合物或其可药用的盐,其为通式(III-2)所示的化合物,或其可药用的盐:
其中,
环B为含氮杂环基;
n5为1、2、3、4、5、6或7;
u为0、1、2、3、4、5或6;s为0、1、2或3;
R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、L5、L6、J1、J2、q、R03、J4、J5、J6、RQ和Z4如通式(I)所定义。
在本公开的一些实施方案中,所述的通式(I)、(IM)、(IN)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中Z4为CH2或C(O),RQ为氢原子或卤素;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(I)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中,L5为CH2或O,在一些实施方案中,L5为O。
在本公开的一些实施方案中,所述的通式(I)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中,L6为CH2或C(O);在一些实施方案中,L6为C(O)。
在一些实施方案中,所述的通式(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)或(III-2)所示的化合物或其可药用的盐,其中n5为2、3、4、5或6;在一些实施方案中,n5为3或4;在一些实施方案中,n5为4。
在一些实施方案中,所述的通式(I)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)或(III-2)所示的化合物或其可药用的盐,其中L5为CH2或O,L6为CH2或C(O);在一些实施方案中,-L5-L6-为-(CH2)2-或-O-C(O)-;在一些实施方案中,-L5-L6-为-(CH2)2-;在一些实施方案中,-L5-L6-为-O-C(O)-。
在本公开的一些实施方案中,所述的通式(I)、(I’)、(IM)、(IN)、(II-1)、(II-2)、 (II-3)、(I’-1)、(I’-2)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中t为1。
在本公开的一些实施方案中,所述的通式(I)、(IM)、(II-3’)、(III-1)所示的化合物或其可药用的盐,其中,环C为3至12元含氮杂环基;在一些实施方案中,环C选自哌啶基、哌嗪基、在一些实施方案中,环C选自哌啶基、哌嗪基和
在本公开的一些实施方案中,所述的通式(I)、(IN)、(III-2)所示的化合物或其可药用的盐,其中环B为3至12元含氮杂环基;在一些实施方案中,环B选自哌啶基、哌嗪基和二氮杂螺[5.5]十一烷基。
在本公开的一些实施方案中,所述的通式(I)、(IM)、(IN)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中u为0、1或2;在一些实施方案中,u为0;在一些实施方案中,u为1;在一些实施方案中,u为2。
在本公开的一些实施方案中,所述的通式(I)、(I’)、(IM)、(IN)、(II-3)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中Z4为C(O);在一些实施方案中,Z4为CH2
在本公开的一些实施方案中,所述的通式(I)、(I’)、(IM)、(IN)、(II-1)所示的化合物或其可药用的盐,其中Q2、Q3和Q4中的一者为碳原子且与J6连接,余下的两者以及Q1和Q5相同或不同,且各自独立地为N或CRQ;在一些实施方案中,Q2、Q3和Q4中的一者为碳原子且与J6连接,余下的两者以及Q1和Q5均为CRQ;在一些实施方案中,Q2为碳原子且与J6连接,Q1、Q3、Q4和Q5均为CRQ;以上方案中,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;在一些实施方案中,Q2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(I)、(I’)、(IM)、(IN)、(II-2)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和Q4均为CRQ;RQ如通式(I)中所定义;在一些实施方案中,Q2与J6连接,Q1、Q3和Q4均为CH。
在本公开的一些实施方案中,所述的通式(I)、(I’)、(IM)、(IN)、(II-3)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中s为0或1;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(I)、(I’)、(IM)、(IN)、(II-1)、(II-2)、(II-3)、(I’-1)、(I’-2)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中q为1、2、3或4;在一些实施方案中,q为0或1;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,所述的通式(I)、(IM)、(IN)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中q1为1、2、3或4;在一些实施方案中,q1为1。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)所示的化合物或其可药用的盐,其中n2为3、4、5、6或7;优选为5。
在本公开的一些实施方案中,所述的通式(I)、(IM)、(IN)、(II-3)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中J1为任选被1或多个R03所取代的5至7元单杂环基或7至14元螺杂环基;R03选自氧代基、卤素、C1-6烷基、C1-6烷氧基、C1-6卤代烷基、羟基、C1-6羟烷基、氰基、氨基、硝基、3至8元环烷基、5至14元杂环基、5至14元杂环基氧基、6至10元芳基和5至14元杂芳基;
在一些实施方案中,J1为任选被1或多个R03所取代的5至7元单杂环基或7至14元螺杂环基;R03选自氧代基、卤素、C1-6烷基、C1-6卤代烷基和C1-6烷氧基;优选地,J1为任选被1个或多个R03所取代的5至7元单杂环基,R03选自氧代基、卤素、C1-6烷基、C1-6卤代烷基和C1-6烷氧基;在一些实施方案中,J1为5至7元单杂环基;
在一些实施方案中,J1为任选被1或多个R03所取代的7至14元螺杂环基;R03选自氧代基、卤素、C1-6烷基、C1-6卤代烷基和C1-6烷氧基;在一些实施方案中,J1为7至14元螺杂环基;
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卤素、羟基和C1-6烷基中的一个或多个取代基所取代;
在一些实施方案中,J1选自:
优选为*端与J2连接;在一些实施方案中,J1选自键、哌啶基和哌嗪基。
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J2、J3、J4和J5相同或不同,且各自独立地选自键、O、(CH2)1-6、O(CH2)1-6、(CH2)1-6O、C(O)、C(O)(CH2)1-6、(CH2)1-6C(O)、N(RJ)、N(RJ)(CH2)1-6、(CH2)1-6N(RJ)、5至14元杂环基、(CH2)1-6-5至7元杂环基和5至7元杂环基-(CH2)1-6;所述的5至14元杂环基任选被1或多个R03所取代;RJ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、C1-6烷基、C1-6烷氧基、C1-6卤代烷基、羟基、C1-6羟烷基、氰基、氨基、硝基、3至8元环烷基、5至14元杂环基、5至14元杂环基氧基、6至10元芳基和5至14元杂芳基;在一些实施方案中,J2、J3、J4和J5相同或不同,且各自独立地选自键、O、(CH2)1-6、C(O)、N(RJ)、5至14元杂环基、(CH2)1-6-5至7元杂环基和5至7元杂环基-(CH2)1-6,RJ为氢原子或C1-6烷基;所述的5至14元杂环基各自独立地任选被一个或多个R03所取代,R03选自氧代基、卤素、C1-6烷基、C1-6烷氧基、C1-6卤代烷基和羟基;在一些实施方案中,J2、J3、J4和J5相同或不同,且各自独立地选自键、(CH2)q、C(O)和3至12元杂环基,所述3至12元杂环基任选被一个或多个R03所取代,R03和q如通式(I)中所定义;在一些实施方案中,J2、J3、J4和J5相同或不同,且各自独立地选自键、CH2、C(O)和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)、(IN)、(II-3’)或(III-2)所示的化合物或其可药用的盐,其中J2选自键、O、(CH2)1-6、C(O)、N(RJ)、5至14元杂环基、(CH2)1-6-5至7元杂环基和5至7元杂环基-(CH2)1-6,RJ为氢原子或C1-6烷基;所述的5至14元杂环基各自独立地任选被一个或多个R03所取代,R03选自氧代基、卤素、C1-6烷基、C1-6烷氧基、C1-6卤代烷基和羟基;优选为(CH2)1-6或C(O);更优选为CH2或C(O);
在一些实施方案中,J2选自键、O、C(O)、NH、N(CH3)、CH2、C(CH3)2、CH2CH2、CH2CH2CH2、CH2CH2CH2CH2、OCH2、CH2O、OCH2CH2、CH2CH2O、C(O)CH2、CH2C(O)、C(O)CH2CH2、CH2CH2C(O)、NHCH2、CH2NH、NHCH2CH2、CH2CH2NH、C(O)NHCH2、CH2C(O)NH、C(O)NHCH2CH2、CH2CH2C(O)NH、1,4-亚哌啶基-、(CH2)1-2-1,4-亚哌啶基、1,4-亚哌啶基-(CH2)1-2、1,4-亚哌嗪基、(CH2)1-2-1,4-亚哌嗪基和1,4-亚哌嗪基-(CH2)1-2;在一些实施方案中,J2选自键、C(O)、O、CH2、CH2CH2、NH和5至14元杂环基;
在一些实施方案中,J2选自键、C(O)、3至12元杂环基和(CH2)p,p为0、1、2、3、4、5或6;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;在一些实施方案中,J2选自键、C(O)、CH2和3至10元杂环基,所述的3至10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;在一些实施方案中,J2选自键、C(O)、CH2、哌啶基、哌嗪基和所述的哌啶基和哌嗪基各自独立地任选被选自卤素、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选自键、C(O)、CH2和3至12元杂环基;在一些实施方案中,J2选自键、C(O)、CH2、哌啶基、哌嗪基、
在本公开的一些实施方案中,所述的通式(I)、(I’)、(IM)、(IN)、(II-1)、(II-2)、(II-3)、(I’-1)、(I’-2)、(II-3’)、(III-1)或(III-2)所示的化合物或其可药用的盐,其中,RL3和RL4均为氢原子。
在一些实施方案中,所述的通式(I’)、(II-1)、(II-2)、(I’-1)、(I’-2)所示的化合物或其可药用的盐,其中环J1’为任选被1个或多个R03所取代的4至7元含氮杂环基,R03选自氧代基、卤素、羟基、C1-6烷基、C1-6卤代烷基和C1-6烷氧基;在一些实施方案中,环J1’为4至7元含氮杂环基;在一些实施方案中,环J1’为哌啶基或哌嗪基;在一些实施方案中,环J1’
在一些实施方案中,所述的通式(I’-1)所示的化合物或其可药用的盐,其中j为0或1;在一些实施方案中,j为0。
在一些实施方案中,所述的通式(I’-2)所示的化合物或其可药用的盐,其中k为0、1或2;在一些实施方案中,k为0或1;在一些实施方案中,k为0。
在一些实施方案中,所述的通式(I)、(I’)、(IM)、(IN)、(I’-1)所示的化合物或其可药用的盐,其中Z8为CH或氮原子。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(I’)、(I’-1)、(I’-2)所示的化合物或其可药用的盐,其中J3选自键、O、(CH2)1-6、C(O)、N(RJ)、5至14元杂环基、(CH2)1-6-5至7元杂环基和5至7元杂环基-(CH2)1-6,RJ为氢原子或C1-6烷基;所述的5至14元杂环基各自独立地任选被一个或多个R03所取代,R03选自氧代基、卤素、C1-6烷基、C1-6烷氧基、C1-6卤代烷基和羟基;
在一些实施方案中,J3选自键、O、C(O)、NH、N(CH3)、CH2、C(CH3)2、CH2CH2、CH2CH2CH2、CH2CH2CH2CH2、OCH2、CH2O、OCH2CH2、CH2CH2O、C(O)CH2、CH2C(O)、C(O)CH2CH2、CH2CH2C(O)、NHCH2、CH2NH、NHCH2CH2、CH2CH2NH、C(O)NHCH2、CH2C(O)NH、C(O)NHCH2CH2、CH2CH2C(O)NH、1,4-亚哌啶基-、(CH2)1-2-1,4-亚哌啶基、1,4-亚哌啶基-(CH2)1-2、1,4-亚哌嗪基、(CH2)1-2-1,4-亚哌嗪基和1,4-亚哌嗪基-(CH2)1-2;在一些实施方案中,J3选自键、C(O)、O、CH2、CH2CH2、NH、5至14元杂环基、苯基和5或6元杂芳基;在一些实施 方案中,J3为CH2
在一些实施方案中,J3选自(CH2)q、3至10元环烷基和3至10元杂环基,q为0、1、2、3、4、5或6;在一些实施方案中,J3选自CH2、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,J3选自CH2、CH2CH2、哌啶基和哌嗪基;在一些实施方案中,J3选自CH2、哌啶基和哌嗪基。
在本公开的一些实施方案中,所述的通式(II-3)或(II-3’)所示的化合物或其可药用的盐,其中环J3’为4至7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’
在本公开的一些实施方案中,所述的通式(I)、(IM)、(IN)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中J4选自键、O、(CH2)1-6、C(O)、N(RJ)、5至14元杂环基、(CH2)1-6-5至7元杂环基和5至7元杂环基-(CH2)1-6,RJ为氢原子或C1-6烷基;所述的5至14元杂环基各自独立地任选被一个或多个R03所取代,R03选自氧代基、卤素、C1-6烷基、C1-6烷氧基、C1-6卤代烷基和羟基;
在一些实施方案中,J4选自键、O、C(O)、NH、N(CH3)、CH2、C(CH3)2、CH2CH2、CH2CH2CH2、CH2CH2CH2CH2、OCH2、CH2O、OCH2CH2、CH2CH2O、C(O)CH2、CH2C(O)、C(O)CH2CH2、CH2CH2C(O)、NHCH2、CH2NH、NHCH2CH2、CH2CH2NH、C(O)NHCH2、CH2C(O)NH、C(O)NHCH2CH2、CH2CH2C(O)NH、1,4-亚哌啶基-、(CH2)1-2-1,4-亚哌啶基、1,4-亚哌啶基-(CH2)1-2、1,4-亚哌嗪基、(CH2)1-2-1,4-亚哌嗪基和1,4-亚哌嗪基-(CH2)1-2;在一些实施方案中,J4选自键、C(O)、O、CH2、CH2CH2、NH、5至14元杂环基、苯基和5或6元杂芳基;在一些实施方案中,J4为键或C(O)。
在本公开的一些实施方案中,所述的通式(I)、(IM)、(IN)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中J5选自键、O、(CH2)1-6、C(O)、N(RJ)、5至14元杂环基、(CH2)1-6-5至7元杂环基和5至7元杂环基-(CH2)1-6,RJ为氢原子或C1-6烷基;所述的5至14元杂环基各自独立地任选被一个或多个R03所取代,R03选自氧代基、卤素、C1-6烷基、C1-6烷氧基、C1-6卤代烷基和羟基;
在一些实施方案中,J5选自键、O、C(O)、NH、N(CH3)、CH2、C(CH3)2、CH2CH2、CH2CH2CH2、CH2CH2CH2CH2、OCH2、CH2O、OCH2CH2、CH2CH2O、 C(O)CH2、CH2C(O)、C(O)CH2CH2、CH2CH2C(O)、NHCH2、CH2NH、NHCH2CH2、CH2CH2NH、C(O)NHCH2、CH2C(O)NH、C(O)NHCH2CH2、CH2CH2C(O)NH、1,4-亚哌啶基-、(CH2)1-2-1,4-亚哌啶基、1,4-亚哌啶基-(CH2)1-2、1,4-亚哌嗪基、(CH2)1-2-1,4-亚哌嗪基和1,4-亚哌嗪基-(CH2)1-2;在一些实施方案中,J5选自键、C(O)、O、CH2、CH2CH2、NH、5至14元杂环基、苯基和5或6元杂芳基;在一些实施方案中,J5为键。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(I’)、(I’-1)、(I’-2)所示的化合物或其可药用的盐,其中-J3-J4-J5-为-C1-6亚烷基-或-4至7元杂环基-C(O)-;优选为-CH2-或在一些实施方案中,-J3-J4-J5-为-C1-6亚烷基-;优选为-CH2-。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中J6选自O、S、N(RJ)、C1-6亚烷基N(RJ)、C(O)C1-6亚烷基、C1-6亚烷基C(O)、C1-6亚烷基、C2-6亚烯基、C2-6亚炔基、3至8元环烷基和5至14元杂环基;所述的C1-6亚烷基、C2-6亚烯基、C2-6亚炔基、3至8元环烷基和5至14元杂环基各自独立地任选被一个或多个R03所取代,RJ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、C1-6烷基、C1-6烷氧基、C1-6卤代烷基、羟基、C1-6羟烷基、氰基、氨基、硝基、3至8元环烷基、5至14元杂环基、5至14元杂环基氧基、6至10元芳基和5至14元杂芳基;在一些实施方案中,J6选自N(RJ)、C1-6亚烷基N(RJ)、C2-6亚炔基和5至14元杂环基,所述的C1-6亚烷基、C2-6亚炔基和5至14元杂环基任选被1或多个R03所取代,RJ为氢原子或C1-6烷基,各个R03相同或不同,且各自独立地选自卤素、羟基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基和C1-6卤代烷氧基;
在一些实施方案中,J6为5至7元杂环基或C1-6亚烷基N(RJ),所述的5至7元杂环基任选被1或多个选自卤素、羟基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基和C1-6卤代烷氧基的取代基所取代;RJ为氢原子或C1-6烷基;在一些实施方案中,J6选自在一些实施方案中,J6选自
在一些实施方案中,J6选自键、O、3至12元杂环基和C1-6亚烷基N(RJ),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取代基所取代;RJ为氢原子或C1-6烷基;在一些实施方案中,J6选自键、哌啶基、哌嗪基、乙氨基和二氮杂螺[5.5]十一烷基,所述的哌啶基和哌嗪基各自独立地任选被选自卤素、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,J6选自键、3至12元杂环基和C1-6亚烷基N(RJ),所述的3至12元杂环基任选被选自卤素和羟基中的一个或多个取代基所取代;RJ为氢原子或C1-6烷基;
在一些实施方案中,J6选自键、哌啶基、
在一些实施方案中,J6选自键、哌啶基、
在一些实施方案中,J6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卤素和羟基中的一个或多个所取代;在一些实施方案中,J6选自任选被1或多个R03所取代的5至7元杂环基;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卤代烷基;在一些实施方案中,J6选自任选被1或多个卤素所取代的6元含氮杂环基;更优选选自
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)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、C1-6烷基、羟基和环丙基;在一些实施方案中,各个R03相同或不同,且各自独立地选自羟基、卤素和C1-6烷基;在一些实施方案中,各个R03相同或不同,且各自独立地为羟基或卤素;在一些实施方案中,R03为卤素;在一些实施方案中,R03为F;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、甲基和羟基。
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-J1-J2-J3-J4-J5-J6-为-4至7元杂环基-C(O)-C1-6亚烷基-4至7元杂环基-或-4至7元杂环基-C1-6亚烷基-4至7元杂环基-C(O)-C1-6亚烷基N(RJ)-,所述的4至7元杂环基各自独立地任选被一个或多个选自氧代基、卤素、羟基、C1-6烷基、C1-6烷氧基、C1-6卤代烷基和C1-6卤代烷氧基的取代基所取代,RJ为氢原子或C1-6烷基;优选地选自 最优选地,-J1-J2-J3-J4-J5-J6-选自:
在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自 自左向右连接。
在本公开的一些实施方案中,所述的通式(II-1)、(II-2)、(I’)、(I’-1)、(I’-2)所示的化合物或其可药用的盐,其中为4至7元含氮杂环基-C(O)-C1-6亚烷基-4至7元杂环基,所述的4至7元含氮杂环基和4至7元杂环基各自独立地任选被一个或多个选自氧代基、卤素、羟基、C1-6烷基、C1-6烷氧基、C1-6卤代烷基和C1-6卤代烷氧基的取代基所取代;优选选自 自左向右连接。
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-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或2;g为1、2、3、4或5;各个R05相同或不同,且各自独立地选自氢原子、卤素、羟基和烷基;J6和R03如通式(I)中所定义;
在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自-杂环基-C(O)-亚烷基-杂环基-、-杂环基-C0-6亚烷基-杂环基-C(O)-亚烷基-N(RJ)-、-杂环基-杂环基-亚烷基-杂环基-、-杂环基-亚烷基-杂环基-、-杂环基-亚烷基-杂环基-杂环基-和-杂环基-亚烷基-C(O)-杂环基-,所述的杂环基各自独立地任选被一个或多个R03所取代,RJ为氢原子或C1-6烷基;R03如通式(I)中所定义;
在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自-3至10元杂环基-C(O)-C1-6亚烷基-3至10元杂环基-、-3至10元杂环基-C0-6亚烷基-3至10元杂环基-C(O)-C1-6亚烷基 -N(RJ)-、-3至10元杂环基-3至10元杂环基-C1-6亚烷基-3至10元杂环基-、-3至10元杂环基-C1-6亚烷基-3至12元杂环基-和-3至10元杂环基-C1-6亚烷基-3至10元杂环基-3至10元杂环基-,所述的3至10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烷基中的一个或多的取代基所取代,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:
自左向右连接;
X、Y、U、V、S和T相同或不同,且各自独立地为CR05或N;q、q1、q2、u、v、w和t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;条件是,q与q1不同时为0;
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、v、w和t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:
自左向右连接;各个q相同或不同,且各自独立地为0、1、2、3、4、5或6;各个u相同或不同,且各自独立地为0、1、2、3、4、5或6;各个q1相同或不同,且各自独立地为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;
在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自:


自左向右连接;
q为0、1、2、3、4、5或6;u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;
在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自:
自左向右连接;
在一些实施方案中,-J1-J2-J3-J4-J5-J6-选自:
自左向右连接。
在本公开一些实施方案中,所述的通式(IM)、(III-1)所示的化合物或其可药用的盐,其中选自:
自左向右连接;
X、Y、U、V、S和T相同或不同,且各自独立地为CR05或N;q、u、v和w相同或不同,且各自独立地为0、1、2、3、4、5或6;t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或2;各个RJ3为氢原子;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;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;各个RJ5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,选自:
自左向右连接;各个q相同或不同,且各自独立地为0、1、2、3、4、5或6;各个u相同或不同,且各自独立地为0、1、2、3、4、5或6;q1为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;
在一些实施方案中,选自: 自左向右连接。
在本公开一些实施方案中,所述的通式(IN)、(III-2)所示的化合物或其可药用的盐,其中选自:
自左向右连接;X、Y、V、S和T相同或不同,且各自独立地为CR05或N;q、u、v和w相同或不同,且各自独立地为0、1、2、3、4、5或6;t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或2;RJ3为氢原子;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;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;各个RJ5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,选自:
自左向右连接;各个q相同或不同,且各自独立地为0、1、2、3、4、5或6;u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;
在一些实施方案中,选自: 自左向右连接。
在本公开一些实施方案中,所述的通式(II-3’)所示的化合物或其可药用的盐, 其中X、Y和U相同或不同,且各自独立地为CR05或N;q和t相同或不同,且各自独立地为0、1、2、3、4、5或6;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;RJ3和RJ5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)、(IM)、(IN)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;和/或J2选自键、C(O)、CH2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;和/或J3选自CH2、CH2CH2、哌啶基和哌嗪基;和/或J4为键或C(O);和/或J5为键;和/或J6选自键、3至12元杂环基和C1-6亚烷基N(RJ),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取代基所取代;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;和/或J2选自键、C(O)、CH2和3至10元杂环基,所述的3至10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;和/或J3选自CH2、哌啶基和哌嗪基;和/或J4为键或C(O);和/或J5为键;和/或J6选自键、3至12元杂环基和C1-6亚烷基N(RJ),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取代基所取代;RJ为氢原子或C1-6烷基。
在本公开的一些实施方案中,u为0、1或2;和/或v为0、1或2;和/或w为0、1或2。
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g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,X、Y、U、V、S和T不同时为N。
在本公开的一些实施方案中,所述的通式(I)、(IM)、(IN)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中,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,所述的通式(I)所示的化合物或其可药用的盐,其中,J1选自:
*端与J2连接;
J2、J3、J4和J5相同或不同,且各自独立地选自键、O、C(O)、NH、N(CH3)、CH2、C(CH3)2、CH2CH2、CH2CH2CH2、CH2CH2CH2CH2、OCH2、CH2O、OCH2CH2、CH2CH2O、C(O)CH2、CH2C(O)、C(O)CH2CH2、CH2CH2C(O)、NHCH2、CH2NH、NHCH2CH2、CH2CH2NH、C(O)NHCH2、CH2C(O)NH、C(O)NHCH2CH2、CH2CH2C(O)NH、1,4-亚哌啶基、(CH2)1-2-1,4-亚哌啶基、1,4-亚哌啶基-(CH2)1-2、1,4-亚哌嗪基、(CH2)1-2-1,4-亚哌嗪基和1,4-亚哌嗪基-(CH2)1-2
J6选自:键、O、S、NH、N(CH3)、-CH=CH-、亚乙炔基、
在本公开的一些实施方案中,在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中R1选自氢原子、卤素和C1-6烷基;优选地,R1为氢原子;在一些实施方案中,R1为C1-6烷基;在一些实施方案中,R1为甲基;在一些实施方案中,R1为3至8元环烷基;在一些实施方案中,R1为环丙基。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中R1为氢原子;和/或R4为氢原子。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中R2为-N(R23)SO2R24或者-P(O)R23R24;R23和R24各自独立地为氢原子或C1-6烷基;在一些实施方案中,R2为-N(R23)SO2R24,R23和R24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,R2为-N(R23)SO2R24,R23和R24分别为甲基;
在一些实施方案中,R2为-N(R23)SO2R24,R23和R24如通式(I)中所定义;在一些实施方案中,R2为-N(R23)SO2R24,R23和R24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,R2为-N(R23)SO2R24,R23和R24相同或不同,且各自独立地为甲基和环丙基;
在一些实施方案中,R2为-N(R23)SO2R24或者-P(O)R23R24;R23和R24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,R2为-N(R23)SO2R24或者-P(O)R23R24;R23和R24相同或不同,且各自独立地选自氢原子、甲基和环丙基;
在一些实施方案中,R2为-P(O)R23R24,R23和R24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,R2为-P(O)R23R24,R23和R24分别为甲基。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中R1、R2及与其相连的C原子一起形成任选的被一个或多个R01所取代的5至7元杂环基或5至14元杂芳基;各个R01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、羟基、C1-6羟烷基、氰基、氨基、硝基、3至8元环烷基、5至14元杂环基、5至14元杂环基氧基、6至10元芳基和5至14元杂芳基。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中R3选自卤素、C1-6烷基、C1-6烷氧基、C1-6卤代烷基或C2-6烯基;在一些实施方案中,R3为卤素或C2-6烯基;在一些实施方案中,R3为溴或乙烯基;在一些实施方案中,R3为卤素;在一些实施方案中,R3为氯或溴;在一些实施方案中,R3为溴。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中R4选自氢原子、卤素、C1-6烷基、C2-6烯基和C1-6卤代烷基;优选地,R4为氢原子。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中R3、R4及与其相连的碳原子一起形成任选的被一个或多个R01所取代的3至8元环烷基、5至7元杂环基或5至14元杂芳基;R01选自卤素、C1-6烷基、C1-6烷氧基、C1-6卤代烷基和C1-6卤代烷氧基。
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Z1为CRZ1;RZ1选自氢原子、C1-6烷氧基、C1-6烷基和C1-6卤代烷氧基;在一些实施方案中,Z1为CRZ1,RZ1为氢原子;
在一些实施方案中,Z1为CRZ1;RZ1选自氢原子、卤素、C1-6烷氧基、C1-6烷基、C1-6卤代烷氧基和3至6元环烷基;在一些实施方案中,Z1为CRZ1,RZ1为氢原子或卤素;在一些实施方案中,Z1为CH或CF;在一些实施方案中,Z1为N。
在一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中Z2为CRZ2;RZ2选自氢原子、C1-6烷氧基、C1-6烷基和C1-6卤代烷氧基;在一些实施方案中,Z2为CRZ2,RZ2为C1-6烷氧基;在一些实施方案中,Z2为CRZ2,RZ2为甲氧基;在一些实施方案中,Z2为CRZ2,RZ2为氢原子或C1-6烷氧基;在一些实施方案中,Z2为CH或COCH3
在一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中RZ1选自氢原子、卤素、C1-6烷氧基、C1-6烷基、C1-6卤代烷氧基和3至6元环烷基;在一些 实施方案中,RZ1为氢原子或卤素;在一些实施方案中,RZ1为氢原子或F。
在一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中RZ2选自氢原子、C1-6烷氧基、C1-6烷基和C1-6卤代烷氧基;在一些实施方案中,RZ2为C1-6烷氧基;在一些实施方案中,RZ2为甲氧基;在一些实施方案中,RZ2为氢原子或C1-6烷氧基;在一些实施方案中,RZ2为氢原子或甲氧基。
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Z3为CH。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中RZ1、RZ2及与其相连的碳子一起形成任选被一个或多个R02所取代的3至8元环烷基、3至8元杂环基或5至14元杂芳基;各个R02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、羟基、C1-6羟烷基、氰基、氨基、硝基、3至8元环烷基、5至14元杂环基、5至14元杂环基氧基、6至10元芳基和5至14元杂芳基。
在本公开的一些实施方案中,所述的通式(I)、(II-2)、(I’)、(I’-1)、(IM)、(IN)所示的化合物或其可药用的盐,其中RZ4为氢原子或C1-6烷基;优选为C1-6烷基;更优选为CH3
在本公开的一些实施方案中,所述的通式(I)、(I’)、(I’-2)、(IM)、(IN)所示的化合物或其可药用的盐,其中RZ7为氢原子或C1-6烷基;优选为氢原子。
在一些实施方案中,所述的通式(I)、(II-1)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中
在一些实施方案中,所述的通式(I)、(II-1)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中R23和R24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,R23和R24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,R23和R24相同或不同,且各自独立地为C1-6烷基;在一些实施方案中,R23和R24均为甲基;在一些实施方案中,R23和R24相同或不同,且各自独立地为甲基或环丙基。
在一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中R23选自氢 原子、C1-6烷基和3至6元环烷基,R24为C1-6烷基或3至6元环烷基;在一些实施方案中,R23为C1-6烷基或3至6元环烷基,R24为C1-6烷基;在一些实施方案中,R23为甲基或环丙基,R24为甲基。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中RJ1、RJ2、RJ3、RJ4、RJ5和RJ6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,RJ1、RJ2、RJ3、RJ4、RJ5和RJ6相同或不同,且各自独立地为氢原子。
在本公开的一些实施方案中,所述的通式(I)、(IM)、(IN)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中,RJ5为甲基。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中RJ5和RJ6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,RJ5和RJ6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)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中RJ为氢原子或C1-6烷基;在一些实施方案中,RJ为氢原子或甲基;在一些实施方案中,RJ为氢原子。
在本公开的一些实施方案中,所述的通式(I)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中R21、R22、R31、R32、Ra、Rb、RL、Rd和Re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)、(II-1)、(II-2)、(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)所示的化合物或其可药用的盐,其中Rc、RL1、RL2、RJ1、RJ2、RJ3和RJ4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)所示的化合物或其可药用的盐,其中,R1选自氢原子、卤素和C1-6烷基;R2为-N(R23)SO2R24或者-P(O)R23R24;R23和R24各自独立地为氢原子或C1-6烷基;R3选自卤素、C1-6烷基、C1-6烷氧基、C1-6卤代烷基或C2-6烯基;R4为氢;Z1为CRZ1;RZ1选自氢原子、C1-6烷氧基、C1-6烷基和C1-6卤代烷氧基;Z2为CRZ2;RZ2选自氢原子、C1-6烷氧基、C1-6烷基和C1-6卤代烷氧基;Z3为CH;-L1-L2-L3-L4-L5-L6-为-O-C(RL3RL4)n2-;n2为2、3、4、5、6、7或8;RL3和RL4各自独立地选自氢原子、C1-6烷基和C1-6烷氧基;J1为任选被1或多个R03取代的5至7元杂环基或7至14元螺杂环基;J2选自键、C(O)、O、CH2、CH2CH2、NH和5至14元杂环基;J3选自键、C(O)、O、CH2、CH2CH2、NH、5至14元杂环基、苯基和5或6元杂芳基;J4选自键、C(O)、O、CH2、CH2CH2、NH、5至14元杂环基、苯基和5或6元杂芳基;J5选自键、C(O)、O、CH2、CH2CH2、NH、5至14元杂环基、苯基和5或6元杂芳基;J6选自N(RJ)、C1-6亚烷基N(RJ)、 C2-6亚炔基和5至14元杂环基,所述的C1-6亚烷基、C2-6亚炔基和5至14元杂环基任选被1或多个R03所取代,RJ为氢原子或C1-6烷基;
各个R03相同或不同,且各自独立地选自卤素、羟基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基和C1-6卤代烷氧基;环A选自:
Z8为CH或氮原子;Z4为CH2或C(O);RZ4选自氢原子、C1-6烷基和C1-6卤代烷基;RZ7为氢原子或C1-6烷基;RQ选自氢原子、卤素、C1-6烷基、C1-6卤代烷基、C1-6烷氧基、C1-6卤代烷氧基、氰基和氨基。
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为氢原子;R2为-N(R23)SO2R24,R23和R24各自独立地为氢原子或C1-6烷基;R3为卤素或C2-6烯基;R4为氢原子;Z1为CRZ1,RZ1为氢原子或卤素;Z2为CRZ2,RZ2为氢原子或C1-6烷氧基;Z3为CH;L1为O,-L2-L3-L4-为-(CH2)3-或-(CH2)4-,L5为CH2或O,L6为CH2或C(O);-J1-J2-J3-J4-J5-J6-选自 环A选自:
在本公开的一些实施方案中,所述的通式(I’-1)所示的化合物或其可药用的盐,其中,R1为氢原子;R2为-N(R23)SO2R24,R23和R24各自独立地为氢原子或C1-6烷基;R3为卤素;R4为氢原子;RZ1为氢原子或卤素;RZ2为氢原子或C1-6烷氧基;RL3和RL4均为H;L5为CH2或O,L6为CH2或C(O);n5为3、4或5;环J1’-J3-J4-J5-为-C1-6亚烷基-;J6选自Z8为CH或氮原子;RZ4为C1-6烷基;j为0。
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,R1为氢原子;R2为-N(R23)SO2R24,R23和R24各自独立地为氢原子或C1-6烷基;R3为卤素;R4为氢原子;RZ1为氢原子或卤素;RZ2为氢原子或C1-6烷氧基; RL3和RL4均为H;L5为CH2或O,L6为CH2或C(O);n5为3、4或5;环J1’-J3-J4-J5-为-C1-6亚烷基-;J6选自RZ7为氢原子或C1-6烷基;k为0。
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,R1为氢原子;R2为-N(R23)SO2R24,R23和R24各自独立地为氢原子或C1-6烷基;R3为卤素;R4为氢原子;RZ1为氢原子或卤素;RZ2为氢原子或C1-6烷氧基;RL3和RL4均为H;L5为CH2或O,L6为CH2或C(O);n5为3或4;环J1’为哌啶基或哌嗪基;-J3-J4-J5-为-C1-6亚烷基-;J6选自键、哌啶基、哌嗪基、乙氨基和二氮杂螺[5.5]十一烷基,所述的哌啶基和哌嗪基各自独立地任选被选自卤素、羟基和C1-6烷基中的一个或多个所取代;RZ7为氢原子或C1-6烷基;RQ为卤素;k为0或1。
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为氢原子;R2为-N(R23)SO2R24,R23和R24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;R3为卤素或C2-6烯基;R4为氢原子;Z1为CRZ1,RZ1为氢原子或卤素;Z2为CRZ2,RZ2为氢原子或C1-6烷氧基;Z3为CH;L1为O,-L2-L3-L4-为-(CH2)3-或-(CH2)4-,L5为CH2或O,L6为CH2或C(O);-J1-J2-J3-J4-J5-J6-选自:
自左向右连接;
q为0、1、2、3、4、5或6;u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;且环A选自:
Z8为CH或氮原子;Z4为CH2或C(O);RZ7为氢原子或C1-6烷基;RZ4为C1-6烷基;RQ为卤素或C1-6烷基;k为0、1、2、3或4;j为0、1、2或3;s为0、1、2或3。
在本公开的一些实施方案中,所述的通式(III-1)所示的化合物或其可药用的盐,其中,R1为氢原子;R2为-N(R23)SO2R24,R23和R24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;R3为卤素或C2-6烯基;R4为氢原子;RZ1为氢原子或卤素;RZ2为氢原子或C1-6烷氧基;RL3和RL4均为氢原子;L5为CH2或O,L6为CH2或C(O);n5为3或4;J1选自键、哌啶基和哌嗪基;环C选自哌啶基、哌嗪基和u为0、1或2;各个R03相同或不同,且各自独立地选自羟基、卤素和C1-6烷基;q为1;J4为键或C(O);J5为键;J6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卤素和羟基中的一个或多个所取代;Z4为CH2或C(O);RQ为卤素;且s为0或1。
在本公开的一些实施方案中,所述的通式(III-2)所示的化合物或其可药用的盐,其中,R1为氢原子;R2为-N(R23)SO2R24,R23和R24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;R3为卤素或C2-6烯基;R4为氢原子;RZ1为氢原子或卤素;RZ2为氢原子或C1-6烷氧基;RL3和RL4均为氢原子;L5为CH2或O,L6为CH2或C(O);n5为3或4;J1选自键、哌啶基和哌嗪基;J2选自键、C(O)、CH2、哌啶基、哌嗪基和所述的哌啶基和哌嗪基各自独立地任选被选自卤素、羟基和C1-6烷基中的一个或多个所取代;q为1;环B选自哌啶基、哌嗪基和二氮杂螺[5.5]十一烷基;u为0、1或2;各个R03相同或不同,且各自独立地选自羟基、卤素和C1-6烷基;J4为键或C(O);J5为键;J6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卤素和羟基中的一个或多个所取代;Z4为CH2或C(O);RQ为卤素;且s为0或1。
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为氢原子;R2为-N(R23)SO2R24或者-P(O)R23R24;R23和R24相同或不同,且各自独立地选自氢原子、甲基和环丙基;R3为卤素或C2-6烯基;R4为氢原子;Z1为CRZ1,RZ1为氢原子或卤素;Z2为CRZ2,RZ2为氢原子或C1-6烷氧基;Z3为CH;L1为O,-L2-L3-L4-为-(CH2)3-或-(CH2)4-,L5为CH2或O,L6为CH2或C(O); -J1-J2-J3-J4-J5-J6-选自 自左向右连接;
环A选自:
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通式(II-1A)或(II-2A)所示的化合物或其盐:
其中,RA为氢原子或氨基保护基;所述氨基保护基优选为Boc;
环J1’为5至14元含氮杂环基;
x为0、1、2或3;n2为2、3、4、5、6、7或8;
R1、R2、R3、R4、RZ1、RZ2、RL3、RL4和J1如通式(I)所定义;
条件是,通式(II-1A)所示的化合物不是:
本公开的另一方面涉及通式(I’A)所示的化合物或其盐:
其中,R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、L5、L6、环J1’和n5如通式(I’)中所定义。
本公开的另一方面涉及通式(IMA)或(INA)所示的化合物或其盐:
其中,环C为含氮杂环基;
n5为1、2、3、4、5、6或7;
q-1为0、1、2、3、4或5;u为0、1、2、3、4、5或6;
R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、L5、L6、J1、J2和R03如通式(I)中所定义。
在本公开的一些实施方案中,所述通式(IMA)所示的化合物或其盐,其中环C上的NH为未被取代的NH。
本公开的另一方面涉及通式(II-3’A)所示的化合物或其盐:
其中,
环C、R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、n5、L5、L6、J1、J2、R03和u如通式(II-3’)中所定义。
本公开的另一方面涉及一种通式(II-3B)所示的化合物或其盐:
其中,
J5为C1-6亚烷基,所述C1-6亚烷基被1或多个选自卤素、羟基、C1-6烷基、C1-6卤代烷基和C1-6烷氧基的取代基所取代;
环J3’为5至14元含氮杂环基,所述的5至14元含氮杂环基任选被1个或多个选自卤素、羟基、C1-6烷基和C1-6卤代烷基所取代;
J4为O、OC1-6亚烷基、C1-6亚烷基O、S、NH、NHC1-6亚烷基、C1-6亚烷基NH、C1-6亚烷基、C(O)、C(O)C1-6亚烷基或C1-6亚烷基C(O);
RJ为氢原子或C1-6烷基;Z4为CH2或C(O);
s为0、1或2;RQ各自独立地选自卤素、C1-6卤代烷基和氰基。
本公开的一些实施方案中,所述通式(INA)或(III-1B)所示的化合物或其盐,其中q-1为0或1;在一些实施方案中,q-1为0。
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’)所示的化合物或其可药用的盐的方 法,该方法包括:
通式(I’A)所示的化合物或其盐(优选为盐酸盐)与通式(I’B)所示的化合物或其盐进行缩合反应得到通式(I’)所示的化合物或其可药用的盐,
其中,环A、R1至R4、RZ1、RZ2、RL3、RL4、L5、L6、环J1’、J3至J6和n5如通式(I’)中所定义。
本公开另一方面涉及一种制备上述通式(IM)所示的化合物或其可药用的盐的方法,该方法包括:
通式(IMA)所示的化合物或其盐(优选为盐酸盐或三氟乙酸盐)与通式(IMB)所示的化合物或其盐发生还原胺化反应得到通式(IM)所示的化合物或其可药用的盐,
其中,环A、环C、R1至R4、RZ1、RZ2、RL3、RL4、n5、L5、L6、J1、q、J4至J6、R03和u如通式(IM)中所定义。
本公开另一方面涉及一种制备上述通式(IN)所示的化合物或其可药用的盐的方法,该方法包括:
通式(INA)所示的化合物或其盐与通式(INB)所示的化合物或其盐(优选为盐酸盐)发生还原胺化反应得到通式(IN)所示的化合物或其可药用的盐,
其中,环A、环B、R1至R4、RZ1、RZ2、RL3、RL4、n5、L5、L6、J1、J2、q、J4至J6、R03和u如通式(IN)中所定义。
本公开另一方面涉及一种制备上述通式(II-1)、(II-2)或(II-3)所示的化合物或其可药用的盐的方法,该方法包括:
通式(II-1A)所示的化合物或其盐(优选为盐酸盐)与通式(II-1B)所示的化合物或其盐进行缩合反应得到通式(II-1)所示的化合物或其可药用的盐,
通式(II-1A)所示的化合物或其盐(优选为盐酸盐)与通式(II-2B)所示的化合物或其盐进行缩合反应得到通式(II-2)所示的化合物或其可药用的盐,
通式(II-2A)所示的化合物或其盐与通式(II-3B)所示的化合物或其盐进行还原胺化反应得到通式(II-3)所示的化合物或其可药用的盐;其中,
RA为氢原子;RJ为氢原子或C1-6烷基;环J1’为5至14元含氮杂环基;
环J3’为5至14元含氮杂环基,所述的5至14元含氮杂环基任选被1个或多个选自卤素、羟基、C1-6烷基和C1-6卤代烷基的取代基所取代;n2为2、3、4、5、 6、7或8;x为0、1、2或3;s为0、1或2;q为1、2、3或4;
R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、J1、J3、J4、J5、J6、Z4、RZ4、Q1、Q2、Q3、Q4、Q5和RQ如通式(I)中所定义。
本公开另一方面涉及一种制备上述通式(II-3’)所示的化合物或其可药用的盐的方法,该方法包括:
通式(II-3’A)所示的化合物或其盐(优选为盐酸盐)与通式(II-3’B)所示的化合物或其盐进行缩合反应得到通式(II-3’)所示的化合物或其可药用的盐,
其中,环C、R1至R4、RZ1、RZ2、RL3、RL4、n5、L5、L6、J1、J2、R03、u、RJ5、RJ6、t、RJ、RQ、Z4和s如通式(II-3’)中所定义。
本公开另一方面涉及一种制备上述通式(I’-1)所示的化合物或其可药用的盐的方法,该方法包括:
通式(I’A)所示的化合物或其盐(优选为盐酸盐)与通式(II’-2B)所示的化合物或其盐进行缩合反应得到通式(I’-1)所示的化合物或其可药用的盐,
其中,R1至R4、RZ1、RZ2、RL3、RL4、L5、L6、n5、环J1’、J3至J6、RQ、RZ4、Z8和j如通式(I’-1)中所定义。
本公开另一方面涉及一种制备上述通式(I’-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’A)所示的化合物或其盐(优选为盐酸盐)与通式(I’-2B)所示的化合物或其盐进行缩合反应得到通式(I’-2)所示的化合物或其可药用的盐,
其中,R1至R4、RZ1、RZ2、RL3、RL4、L5、L6、n5、环J1’、J3至J6、RQ、RZ7和k如通式(I’-2)中所定义。
本公开另一方面涉及一种制备上述通式(III-1)所示的化合物或其可药用的盐的方法,该方法包括:
通式(IMA)所示的化合物或其盐(优选为盐酸盐或三氟乙酸盐)与通式(III-1B)所示的化合物或其盐发生还原胺化反应得到通式(III-1)所示的化合物或其可药用的盐,
其中,环C、R1至R4、RZ1、RZ2、RL3、RL4、n5、L5、L6、J1、R03、u、q、J4至J6、RQ、Z4和s如通式(III-1)中所定义。
本公开另一方面涉及一种制备上述通式(III-2)所示的化合物或其可药用的盐的方法,该方法包括:
通式(INA)所示的化合物或其盐与通式(III-2B)所示的化合物或其盐(优选为盐酸盐)发生还原胺化反应得到通式(III-2)所示的化合物或其可药用的盐,
其中,环B、R1至R4、RZ1、RZ2、RL3、RL4、n5、L5、L6、J1、J2、q、R03、u、J4至J6、RQ、Z4和s如通式(III-2)中所定义。
在本公开一些实施方案中,所述通式(IM)、(IN)、(III-1)、(III-2)所示的化合物或其可药用盐的制备方法,其中q为1、2、3、4、5或6,q-1为0、1、2、3、4、或5。
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治疗有效量的本公开通式(I)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)以及表A所示的化合物或其可药用盐,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)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)以及表A所示的化合物或其可药用盐,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)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)以及表A所示的化合物或其可药用盐,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。
在本公开中,由EGFR介导的或依赖性的疾病或病症优选选自癌症。
本公开进一步涉及通式(I)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)以及表A所示的化合物或其可药用盐,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、肝癌、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、甲状腺癌、胶质母细胞瘤(Glioblastoma)、脑转移瘤(Brain Metastases)、实体瘤、口咽癌(Oropharyngeal Carcinoma)、支气管肿瘤(Bronchial Neoplasms)和皮肤癌;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上述通式(I)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)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上述通式(I)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)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上述通式(I)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)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上述通式(I)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)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、肝癌、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、甲状腺癌、胶质母细胞瘤、脑转移瘤、实体瘤、口咽癌、支气管肿瘤和皮肤癌;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)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)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)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)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)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)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)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)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)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)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)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)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,其用于抑制和/或降解EGFR蛋白。
本公开进一步涉及上述通式(I)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)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)、通式(II-1)、通式(II-2)、通式(II-3)、(I’)、(I’-1)、(I’-2)、(IM)、(IN)、(II-3’)、(III-1)、(III-2)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、肝癌、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、甲状腺癌、胶质母细胞瘤、脑转移瘤、实体瘤、口咽癌、支气管肿瘤和皮肤癌;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、肝癌、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、甲状腺癌、胶质母细胞瘤、脑转移瘤、实体瘤、口咽癌、支气管肿瘤和皮肤癌;进一步优选为肺癌;更优选为非小细胞肺癌。
所述肺癌优选小细胞肺癌或非小细胞肺癌,所述非小细胞肺癌优选肺腺癌、肺鳞癌或大细胞肺癌;所述头颈癌优选头颈部鳞状细胞癌;所述食管癌优选食管鳞状细胞癌;所述胶质母细胞瘤优选多形性胶质母细胞瘤。
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。
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C1-12烷基),更优选具有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12个碳原子的亚烷基(即C1-12亚烷基),更优选具有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”指具有0、1、2、3、4、5或6个碳原子的亚烷基;具体的,C0-6亚烷基包含键和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“亚烯基”指二价烯基,其中烯基如上所定义,其具有2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C2-20亚烯基)。所述亚烯基优选具有2至12个碳原子的亚烯基(即C2-12亚烯基),更优选具有2至6个碳原子的亚烯基(即C1-6亚烯基)。非限制性的实例包括:亚乙烯基、-CH2CH=C-、-CH2CH2CH=C-等。亚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“亚炔基”指二价炔基,其中炔基如上所定义,其具有2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C2-20亚炔基)。所述亚炔基优选具有2至12个碳原子的亚炔基(即C2-12亚炔基),更优选具有2至6个碳原子的亚炔基(即C2-6亚炔基)。非限制性的实例包括:亚乙炔基、-CH2C≡C-、-CH2CH2C≡C-等。亚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),更优选具有5或6个环原子的环烷基(即5或6元环烷基)。
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)或具有4至7个环原子的杂环基(即4至7元杂环基);最优选具有5或6个环原子的杂环基(即5或6元杂环基)。
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14个环原子的螺杂环基(即7至14元螺杂环基)或具有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14个(例如6、7、8、9、10、11、12、13或14个)环原子(即6至14元芳基)。所述芳基优选具有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、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“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“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“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“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。
1,4-亚哌啶基指1,4-亚哌嗪基指
氢或H也即氢原子。
需要说明的是,(CRJ1RJ2)p中,当p为0时,(CRJ1RJ2)p为键;同理,当q为0时,(CRJ3RJ4)q为键;当t为0时,(CRJ5RJ6)t为键。
需要说明的是,*表示连接位点。
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。
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(II-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(II-1A)所示的化合物或其盐(优选为盐酸盐)和通式(II-1B)所示的化合物或其盐,在碱性条件下缩合剂存在下,进行缩合反应,得到通式(II-1)所示的化合物或其可药用的盐;其中,
RA为氢原子;
R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、n2、环J1’、J3、J4、J5、J6、Q1、Q2、Q3、Q4和Q5如通式(II-1)中所定义。
方案二
本公开通式(II-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(II-1A)所示的化合物或其盐(优选为盐酸盐)和通式(II-2B)所示的化合物或其盐,在碱性条件下缩合剂存在下,进行缩合反应,得到通式(II-2)所示的化合物或其可药用的盐;其中,
RA为氢原子;
R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、n2、环J1’、J3、J4、J5、J6、RZ4、Q1、Q2、Q3和Q4如通式(II-2)中所定义。
方案三
本公开通式(II-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(II-2A)所示的化合物或其盐和通式(II-3B)所示的化合物或其盐(优选为盐酸盐),在弱酸性条件下,还原剂存在下发生还原胺化反应,得到通式(II-3)所示的化合物或其可药用的盐;其中,
R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、J1、q、环J3’、J4、J5、Z4、RQ、RJ、s、n2和x如通式(II-3)和(II-2A)中所定义。
方案四
本公开通式(I’)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’A)所示的化合物或其盐(优选盐酸盐)与通式(I’B)所示的化合物或其盐,在碱性条件下,缩合剂存在下,进行缩合反应得到通式(I’)所示的化合物或其可药用的盐,
其中,环A、R1至R4、RZ1、RZ2、RL3、RL4、L5、L6、环J1’、J3至J6和n5如通式(I’)中所定义。
方案五
本公开通式(I’-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’A)所示的化合物或其盐(优选盐酸盐)与通式(II’-2B)所示的化合物或其盐,在碱性条件下,缩合剂存在下,进行缩合反应得到通式(I’-1)所示的化合物或其可药用的盐,
其中,R1至R4、RZ1、RZ2、RL3、RL4、L5、L6、n5、环J1’、J3至J6、RQ、RZ4、Z8和j如通式(I’-1)中所定义。
方案六
本公开通式(I’-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’A)所示的化合物或其盐(优选盐酸盐)与通式(I’-2B)所示的化合物或其盐,在碱性条件下,缩合剂存在下,进行缩合反应得到通式(I’-2)所示的化合物或其可药用的盐,
其中,R1至R4、RZ1、RZ2、RL3、RL4、L5、L6、n5、环J1’、J3至J6、RQ、RZ7和k如通式(I’-2)中所定义。
方案七-1
本公开通式(IM)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(IMA)所示的化合物或其盐(优选为盐酸盐或三氟乙酸盐)与通式(IMB)所示的化合物或其盐在酸性条件下,还原剂存在下,发生还原胺化反应得到通式(IM)所示的化合物或其可药用的盐,
其中,环A、环C、R1至R4、RZ1、RZ2、RL3、RL4、n5、L5、L6、J1、q、J4至J6、R03和u如通式(IM)中所定义。
方案七-2
本公开通式(IN)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(INA)所示的化合物或其盐与通式(INB)所示的化合物或其盐(优选为盐酸盐)在酸性条件下,还原剂存在下发生还原胺化反应得到通式(IN)所示的化合物或其可药用的盐,
其中,环A、环B、R1至R4、RZ1、RZ2、RL3、RL4、n5、L5、L6、J1、J2、q、J4至J6、R03和u如通式(IN)中所定义。
方案八
本公开通式(II-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(II-3’A)所示的化合物或其盐(优选为盐酸盐)与通式(II-3’B)所示的化合物或其盐在碱性条件下,缩合剂存在下进行缩合反应得到通式(II-3’)所示的化合物 或其可药用的盐,
其中,环C、R1至R4、RZ1、RZ2、RL3、RL4、n5、L5、L6、J1、J2、R03、u、RJ5、RJ6、t、RJ、RQ、Z4和s如通式(II-3’)中所定义。
方案九-1
本公开通式(III-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(IMA)所示的化合物或其盐(优选为盐酸盐或三氟乙酸盐)与通式(III-1B)所示的化合物或其盐在酸性条件下,还原剂存在下发生还原胺化反应得到通式(III-1)所示的化合物或其可药用的盐,
其中,环C、R1至R4、RZ1、RZ2、RL3、RL4、n5、L5、L6、J1、R03、u、q、J4至J6、RQ、Z4和s如通式(III-1)中所定义。
方案九-2
本公开通式(III-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(INA)所示的化合物或其盐与通式(III-2B)所示的化合物或其盐(优选为盐酸盐)在酸性条件下,还原剂存在下发生还原胺化反应得到通式(III-2)所示的化合 物或其可药用的盐,
其中,环B、R1至R4、RZ1、RZ2、RL3、RL4、n5、L5、L6、J1、J2、q、R03、u、J4至J6、RQ、Z4和s如通式(III-2)中所定义。
以上缩合反应中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、二异丙基乙胺(也即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、2-(7-偶氮苯并三氮唑)-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,提供弱酸性条件的试剂包括但不限于醋酸(乙酸)、Ti(i-PrO)3和BF3Et2O、TiCl4等。
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;优选为乙酸或冰乙酸。
具体的,上述还原胺化反应中,提供酸性条件的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;原料的酸性盐优选为盐酸盐或三氟乙酸盐;具体的,当原料是酸性盐的形式时,则提供酸性条件的方式可以是:加入试剂乙酸钠,在反应过程中由乙酸钠与原料中的酸一起形成醋酸。
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。
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 1200/1290DAD-6110/6120Quadrupole MS液质联用仪(生产商:Agilent,MS型号:6110/6120Quadrupole 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 1260DAD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)、达瑞化学品、百灵威、上海毕得医药等公司。
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℃。
实施例中的反应进程的监测采用薄层色谱法(TLC),反应所使用的展开剂,纯化化合物采用的柱层析的洗脱剂体系和薄层色谱法的展开剂体系包括:A:二氯甲烷/甲醇体系,B:正己烷/乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的三乙胺和醋酸等碱性或酸性试剂进行调节。
实施例1
(±)-N-(35-溴-14-(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)-16-甲氧基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺1
第一步
4-(2-(5-羟基戊-1-炔-1-基)-5-甲氧基-4-硝基苯基)哌嗪-1-羧酸叔丁酯1b
将5-(2-氟-4-甲氧基-5-硝基苯基)戊-4-炔-1-醇1a(1.01g,3.99mmol,采用专利申请“WO2021216440中说明书第54页的实施例A1”公开的方法制备而得)溶于N,N-二甲基甲酰胺(11mL)中,加入无水碳酸钾(1.7g,12.30mmol),1-哌嗪羧酸叔丁酯(950mg,5.10mmol),升温到80℃反应12小时。反应液冷却至室温,加水 10mL,用氯仿/异丙醇(V/V=3/1,30mL×3)萃取,合并有机相,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B纯化得到标题化合物1b(1.18g,产率:70%)。
MS m/z(ESI):420.2[M+1]。
第二步
4-(4-氨基-2-(5-羟基戊基)-5-甲氧基苯基)哌嗪-1-羧酸叔丁酯1c
向50mL单口瓶中加入化合物1b(1.18g,2.81mmol)、甲醇(20mL),加入10%干钯碳(100mg,百灵威科技),氢气置换三次,在氢气氛下反应12小时。反应液经硅藻土过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物1c(0.79g,产率:71%)。
MS m/z(ESI):394.2[M+1]。
第三步
4-(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)-2-(5-羟基戊基)-5-甲氧基苯基)哌嗪-1-羧酸叔丁酯1e
向250mL单口瓶中加入化合物1c(600mg,1.52mmol)、N-(2-((5-溴-2-氯嘧啶-4-基)氨基)-5-羟基苯基)-N-甲基甲磺酰胺1d(740mg,1.82mmol,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)、异丙醇(120mL)和甲烷磺酸(64mg,0.67mmol),升温至90℃反应48小时。反应液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物1e(930mg,产率:80%)。
MS m/z(ESI):764.1[M+1]。
第四步
4-(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)-2-(5-溴戊基)-5-甲氧基苯基)哌嗪-1-羧酸叔丁酯1f
向100mL单口瓶中加入化合物1e(920mg,1.20mmol)、氯仿(25mL)、吡啶(7.8mL),搅拌使化合物溶解完全,加入三苯基膦(1.9g,7.24mmol)、四溴化碳(2.4g,7.24mmol),反应12小时。向反应液中加入水20mL,用氯仿/异丙醇(V/V=3/1,30mL×3)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩。所得残留物经高效液相制备色谱法(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:31%-46%,流速:30mL/min)分离,得到标题化合物1f(500mg,产率:50%)。
MS m/z(ESI):826.1[M+1]。
第五步
溴-16-甲氧基-52-(N-甲基甲磺酰胺基)-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-14-基)哌嗪-1-羧酸叔丁酯1g
向50mL单口瓶中加入化合物1f(100mg,0.121mmol)、N,N-二甲基甲酰胺(13mL)、碘化钾(6mg,0.036mmol)、无水碳酸钾(67mg,0.485mmol),反应12小时。 反应液过滤,滤液减压浓缩后真空干燥,得到粗品标题化合物1g(110mg,产率:100%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):746.1[M+1]。
第六步
N-(35-溴-16-甲氧基-14-(哌嗪-1-基)-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺盐酸盐1h
将化合物1g(90mg,0.121mmol)溶于二氯甲烷(1mL)中,加入4M氯化氢的1,4-二氧六环溶液(1mL),反应1小时。反应液减压浓缩,真空干燥,得到粗品标题化合物1h(87mg,产率:100%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):646.1[M+1]。
第七步
(±)-N-(35-溴-14-(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)-16-甲氧基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺1
向25mL单口瓶中加入化合物1h(30mg,0.044mmol)、(±)-2-(4-(4-((2,6-二氧代哌啶-3-基)氨基)苯基)哌啶-1-基)乙酸1i(20mg,0.052mmol,采用专利申请“WO2021127561中说明书第408页的实施例1”公开的方法制备而得)、N,N-二甲基甲酰胺(2mL),搅拌均匀后加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(20mg,0.053mmol),搅拌5分种后加入二异丙基乙胺(50mg,0.387mmol),继续反应1小时。反应液经高效液相制备色谱法(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%-80%,流速:30mL/min)纯化,得到标题化合物1(外消旋体混合物,15mg,产率:35%)。
MS m/z(ESI):973.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.76(s,1H),8.16(s,1H),8.06(s,1H),7.46(s,2H),7.35(d,1H),7.28(d,1H),7.09(dd,1H),6.94(d,2H),6.75(s,1H),6.59(d,2H),5.64(d,1H),4.36(t,2H),4.28-4.22(m,1H),3.78(s,3H),3.66(s,2H),3.54(s,2H),3.17(s,2H),3.09(s,3H),3.00(s,3H),2.91(d,2H),2.79-2.65(m,4H),2.64-2.53(m,2H),2.37-2.28(m,2H),2.15-2.05(m,4H),2.03-1.95(m,1H),1.90-1.82(m,1H),1.80-1.74(m,2H),1.69(d,2H),1.61-1.50(m,3H),1.33-1.26(m,2H)。
实施例2
(±)-N-(35-溴-14-(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)-16-甲氧基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺2
向25mL单口瓶中加入化合物1h(30mg,0.044mmol)、(±)-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)乙酸三氟乙酸盐2a(24mg,0.044mmol,采用专利申请“WO2021127561中说明书第581页的实施例70”公开的方法制备而得)、N,N-二甲基甲酰胺(2mL),搅拌均匀后加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(20mg,0.053mmol),搅拌5分钟,加入二异丙基乙胺(50mg,0.387mmol),反应2小时,反应液经高效液相制备色谱法(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%-80%,流速:30mL/min)纯化,得到标题化合物2(外消旋体混合物,16mg,产率:35%)。
MS m/z(ESI):1049.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.56(s,1H),8.16(s,1H),8.07(s,1H),7.59(d,1H),7.53(s,1H),7.47(s,2H),7.35(d,1H),7.28(d,1H),7.11-7.06(m,2H),6.77(s,1H),4.36(t,2H),3.98(s,3H),3.92(t,2H),3.79(s,3H),3.67-3.50(m,3H),3.42(s,2H),3.30-3.17(m,4H),3.09(s,3H),3.00(s,3H),2.82-2.62(m,7H),2.30-2.19(m,1H),2.15-2.09(m,2H),2.03-1.94(m,1H),1.90-1.82(m,1H),1.80-1.74(m,2H),1.59-1.51(m,2H),1.34-1.26(m,2H)。
实施例3
(±)-N-(14-(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)-16-甲氧基-35-乙烯基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺3

第一步
4-(16-甲氧基-52-(N-甲基甲磺酰胺基)-35-乙烯基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-14-基)哌嗪-1-羧酸叔丁酯3a
向25mL单口瓶中加入化合物1g(50mg,0.067mmol)、5.5mL 1,4-二氧六环和水的混合溶剂(V/V=10/1)、乙烯基硼酸频那醇酯(30mg,0.195mmol,上海毕得医药)、[1,1’-双(二苯基膦基)二茂铁]二氯化钯(II)(6mg,0.008mmol,上海毕得医药)、碳酸钠(60mg,0.566mmol),氮气置换3次,升温到110℃反应12小时,反应液减压浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物3a(46mg,产率:99%)。
MS m/z(ESI):694.3[M+1]。
第二步
N-(16-甲氧基-14-(哌嗪-1-基)-35-乙烯基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺3b
将化合物3a(46mg,0.066mmol)溶于二氯甲烷(1.5mL)中,加入4M氯化氢的1,4-二氧六环溶液(1.5mL),反应2小时。反应液减压浓缩,真空干燥,得到粗品标题化合物3b(42mg,产率:100%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):594.2[M+1]。
第三步
(±)-N-(14-(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)-16-甲氧基-35-乙烯基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺3
向25mL单口瓶中加入化合物3b(42mg,0.067mmol),化合物1i(33mg,0.086mmol),N,N-二甲基甲酰胺(2mL),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(33mg,0.087mmol),搅拌5分钟,加入二异丙基乙胺(60mg,0.464mmol),反应1小时,反应液经高效液相制备色谱法(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:54%-79%,流速:30mL/min)得到标题化合物3(外消旋体混合物,11mg,产率:18%)。
MS m/z(ESI):921.4[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.76(s,1H),8.21(d,2H),7.54(s,1H),7.39(s,1H),7.28(d,1H),7.21(d,1H),7.08(d,1H),6.94(d,2H),6.82(dd,1H),6.74(s,1H),6.59(d,2H),5.71-5.62(m,2H),5.19(d,1H),4.35(t,2H),4.28-4.21(m,1H),3.79(s,3H),3.65(s,2H),3.53(s,2H),3.17(s,2H),3.03(s,3H),2.94(s,3H),2.91(d,2H),2.77-2.61(m,5H),2.60-2.54(m,1H),2.37-2.27(m,1H),2.14-2.04(m,4H),2.03-1.95(m,1H),1.90-1.82(m,1H),1.78(s,2H),1.69(d,2H),1.63-1.52(m,4H),1.32-1.26(m,2H)。
实施例4
(±)-N-(14-(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)-16-甲氧基-35-乙烯基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺4
向25mL单口瓶中加入化合物3b(42mg,0.067mmol)、化合物2a(37mg,0.070mmol)、N,N-二甲基甲酰胺(2mL),搅拌均匀后加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(27mg,0.071mmol),搅拌5分钟,加入二异丙基乙胺(70mg,0.542mmol),反应2小时,反应液经高效液相制备色谱法(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%-78%,流速:30mL/min)纯化,得到标题化合物4(外消旋体混合物,16mg,产率:35%)。
MS m/z(ESI):997.3[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.56(s,1H),8.22(d,2H),7.59(d,1H),7.54(d,2H),7.40(s,1H),7.28(d,1H),7.21(d,1H),7.08(dd,2H),6.82(dd,1H),6.76(s,1H),5.67(d,1H),5.20(d,1H),4.35(t,2H),3.98(s,3H),3.92(t,2H),3.79(s,3H),3.66-3.50(m,4H),3.42(s,3H),3.31-3.16(m,3H),3.03(s,3H),2.95(s,3H),2.82-2.63(m,7H),2.29-2.20(m,1H),2.16-2.08(m,2H),1.89-1.75(m,3H),1.60-1.53(m,2H),1.33-1.25(m,2H)。
实施例5
N-(35-溴-14-(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) 哌嗪-1-基)甲基)哌啶-1-基)-16-甲氧基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺5
第一步
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)哌嗪-1-羧酸苄基酯5c
向250mL单口瓶中依次加入4-(L-丙氨酰)哌嗪-1-羧酸苄基酯5a(3.56g,10.86mmol,采用专利申请“WO2014057266中说明书第42页的化合物4j”公开的方法制备而得)、2-(2,6-二氧代哌啶-3-基)-4-氟吲哚啉-1,3-二酮5b(2.5g,9.05mmol,上海皓鸿生物医药科技有限公司)、二甲基亚砜(50mL)和N,N-二异丙基乙胺(3.51g,27.15mmol),升温到100℃反应16小时。将反应液缓慢加入到冰水中,过滤,收集滤饼,水洗(10mL×3),然后将滤饼溶于二氯甲烷中(100mL),用水洗(30mL×3),合并有机相,用无水硫酸钠干燥,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5c(非对映体混合物,1:1,4.3g,产率:87%)。
MS m/z(ESI):548.2[M+1]。
第二步
2-(2,6-二氧代哌啶-3-基)-4-(((S)-1-氧代-1-(哌嗪-1-基)丙-2-基)氨基)异吲哚啉-1,3-二酮5d
向500mL单口瓶中加入化合物5c(3.6g,6.57mmol)、四氢呋喃(100mL)、甲醇(100mL),搅拌溶解后,加入10%干钯碳(500mg,4.70mmol,百灵威科技),氢气置换三次,氢气氛下反应16小时,过滤除去钯碳,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,得到标题产物5d(非对映体混合物,1:1,2.1g,产率:77%)。
MS m/z(ESI):414.3[M+1]。
第三步
5-(2-(4-(二甲氧基甲基)哌啶-1-基)-4-甲氧基-5-硝基苯基)戊-4-炔-1-醇5e
将化合物1a(4.5g,17.77mmol)溶于N,N-二甲基甲酰胺(50mL),加入无水碳酸钾(7.40g,53.54mmol)、4-(二甲氧基甲基)哌啶(3.7g,23.24mmol),升温至80℃反应12小时。反应液冷却至室温,加入水50mL,用氯仿/异丙醇(V/V=3/1,150mL×2)萃取,合并有机相,用无水硫酸钠干燥,过滤,反应液浓缩,用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物5e(5.3g,产率:76%)。
MS m/z(ESI):393.2[M+1]。
第四步
5-(5-氨基-2-(4-(二甲氧基甲基)哌啶-1-基)-4-甲氧基苯基)戊-1-醇5f
向250mL单口瓶中依次加入化合物5e(3.5g,8.92mmol)、甲醇(100mL)和10%干钯碳(445mg,百灵威科技),氢气置换三次,氢气氛围下反应12小时。过滤,滤液浓缩,用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物5f(2.8g,产率:85%)。
MS m/z(ESI):367.3[M+1]。
第五步
N-(2-((5-溴-2-((4-(4-(二甲氧基甲基)哌啶-1-基)-5-(5-羟基戊基)-2-甲氧基苯基)氨基)嘧啶-4-基)氨基)-5-羟基苯基)-N-甲基甲磺酰胺5g
将化合物5f(0.82g,2.24mmol)、化合物1d(938mg,2.30mmol)加入到甲醇(40mL)中,加入甲烷磺酸(300mg,3.12mmol),升温到90℃反应12小时。反应液减压浓缩,加入水20mL,用饱和碳酸氢钠溶液调节溶液pH到8左右,用乙酸乙酯(50mL×3)萃取,合并有机相,用无水硫酸钠干燥,过滤,浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物5g(900mg,产率:54%)。
MS m/z(ESI):737.2[M+1]。
第六步
N-(2-((5-溴-2-((5-(5-溴戊基)-4-(4-(二甲氧基甲基)哌啶-1-基)-2-甲氧基苯基)氨基)嘧 啶-4-基)氨基)-5-羟基苯基)-N-甲基甲磺酰胺5h
将化合物5g(900mg,1.22mmol)溶于氯仿(25mL)中,依次加入吡啶(8mL)、三苯基膦(1.92g,7.32mmol)、四溴化碳(2.43g,7.33mmol),氮气氛围下反应12小时。加水30mL,用氯仿/异丙醇混合液萃取(V/V=3/1,50mL×3),有机相合并,用无水硫酸钠干燥,过滤,滤液浓缩,经高效液相制备色谱法(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:31%-46%,流速:30mL/min)分离,得到标题化合物5h(560mg,产率:57%)。
MS m/z(ESI):799.1[M+1]。
第七步
N-(35-溴-14-(4-(二甲氧基甲基)哌啶-1-基)-16-甲氧基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺5i
将化合物5h(550mg,0.687mmol)溶于N,N-二甲基甲酰胺(70mL)中,加入碘化钾(35mg,0.211mmol)、无水碳酸钾(380mg,2.75mmol),反应12小时。抽滤,反应液减压浓缩,真空干燥,得到粗品标题化合物5i(494mg,产率:100%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):719.2[M+1]。
第八步
N-(35-溴-14-(4-甲酰基哌啶-1-基)-16-甲氧基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺5j
将化合物5i(60mg,0.083mmol)加入到四氢呋喃(1.5mL)中,加入2M硫酸(0.12mL),升温到60℃反应1小时。反应液冷却到室温,用饱和碳酸氢钠溶液调节反应液pH至8左右,用乙酸乙酯(20mL×3)萃取,有机相合并后,用无水硫酸钠干燥,过滤,浓缩,得到粗品标题化合物5j(39mg,产率:69%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):673.1[M+1]。
第九步
N-(35-溴-14-(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)哌嗪-1-基)甲基)哌啶-1-基)-16-甲氧基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺5
将化合物5j(35mg,0.078mmol)加入到3.3mL二氯甲烷和甲醇的混合溶剂中(V/V=10/1),加入无水乙酸钠(37mg,0.451mmol),搅拌15分钟,然后加入化合物5d(39mg,0.056mmol),继续搅拌10分钟,加入三乙酰氧基硼氢化钠(25mg,0.12mmol),反应12小时。反应液浓缩过滤,经高效液相制备色谱法(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:67%-82%,流速:30mL/min)纯化,得到标题化合物5(非对映体混合物,1:1,27mg,产率:43%)。
MS m/z(ESI):1070.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.10(s,1H),8.15(s,1H),8.05(s,1H),7.61(t,1H),7.44(d,2H),7.34(d,1H),7.27(d,1H),7.21(d,1H),7.17-7.12(m,1H),7.07(t,2H),6.74(s,1H),5.06(dd,1H),4.88(t,1H),4.35(t,2H),3.80(s,3H),3.69-3.53(m,4H),3.46-3.42(m,2H),3.09(s,3H),2.99(s,3H),2.93-2.84(m,1H),2.81(d,2H),2.65-2.54(m,5H),2.44-2.35(m,3H),2.32-2.26(m,1H),2.21(d,2H),2.13-2.00(m,4H),1.81-1.73(m,4H),1.66-1.46(m,3H),1.28(d,3H)。
实施例6
(±)-N-(35-溴-14-(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)-15-氟-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺6
第一步
4-(2-溴-6-氟-4-硝基苯基)哌嗪-1-羧酸叔丁酯6b
将1-溴-2,3-二氟-5-硝基苯6a(1.0g,4.20mmol,上海毕得医药)溶于乙腈(20mL)中,加入无水碳酸钾(1.16g,8.39mmol)、哌嗪-1-羧酸叔丁酯(783mg,4.20mmol),升温到85℃反应16小时。反应液冷却至室温,过滤,滤液浓缩,用硅胶柱色谱法以洗脱剂体系B纯化,得到标题化合物6b(1.45g,产率:85%)。
MS m/z(ESI):347.9[M-55]。
第二步
4-(2-氟-6-(5-羟基戊-1-炔-1-基)-4-硝基苯基)哌嗪-1-羧酸叔丁酯6c
向50mL单口瓶中加入化合物6b(800mg,1.98mmol)、N,N-二甲基甲酰胺(12mL)、二(三苯基膦)二氯化钯(139mg,0.20mmol)、碘化亚铜(76mg,0.40mmol)、三苯基膦(78mg,0.30mmol),搅拌10分钟,然后加入4-戊炔-1-醇(249mg,2.96mmol)、二异丙基乙胺(1.02g,7.89mmol),升温至80℃反应16小时。反应液经硅藻土过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化,得到标题化合物6c(650mg,产率:81%)。
MS m/z(ESI):408.2[M+1]。
第三步
4-(4-氨基-2-氟-6-(5-羟基戊基)苯基)哌嗪-1-羧酸叔丁酯6d
向50mL单口瓶中加入化合物6c(650mg,1.60mmol)、甲醇(15mL),加入10%干钯碳(196mg,百灵威科技),氢气置换三次,在氢气氛下反应16小时。反应液经硅藻土过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物6d(500mg,产率:82%)。
MS m/z(ESI):382.8[M+1]。
第四步
4-(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)-2-氟-6-(5-羟基戊基)苯基)哌嗪-1-羧酸叔丁酯6e
向250mL单口瓶中加入化合物6d(100mg,0.26mmol)、化合物1d(128mg,0.31mmol)、异丙醇(3mL)和三氟乙酸(53mg,0.46mmol),升温至90℃反应16小时。反应液减压浓缩,所得残留物溶于二氯甲烷(8mL)中,加入三乙胺(106mg,1.04mmol)和二碳酸二叔丁酯(57mg,0.26mmol),反应1小时。反应液减压浓缩,所得残留物经硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物6e(135mg,产率:68%)。
MS m/z(ESI):752.4[M+1]。
第五步
4-(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)-2-(5-溴戊基)-6-氟苯基)哌嗪-1-羧酸叔丁酯6f
向100mL单口瓶中加入化合物6e(135mg,0.18mmol)、氯仿(10mL)、吡啶(3mL),搅拌使化合物溶解完全,加入三苯基膦(282mg,1.08mmol)、四溴化碳(356mg,1.07mmol),反应12小时。向反应液中加入水(20mL),用氯仿/异丙醇(V/V=3/1,30mL×3)萃取,合并有机相,有机相用无水硫酸钠干燥。过滤,滤液浓缩,所得残留物经高效液相制备色谱法(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:80%-89%,流速:30mL/min)分离,得到标题化合物6f(60mg,产率:41%)。
MS m/z(ESI):814.1[M+1]。
第六步
4-(35-溴-15-氟-52-(N-甲基甲磺酰胺基)-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-14-基)哌嗪-1-羧酸叔丁酯6g
向50mL单口瓶中加入化合物6f(60mg,0.074mmol)、N,N-二甲基甲酰胺(8mL)、碘化钾(4mg,0.024mmol)、无水碳酸钾(41mg,0.30mmol),反应16小时。反应液过滤,滤液减压浓缩,真空干燥,得到粗品标题化合物6g(54mg,产率:100%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):734.1[M+1]。
第七步
N-(35-溴-15-氟-14-(哌嗪-1-基)-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺盐酸盐6h
将化合物6g(54mg,0.074mmol)溶于二氯甲烷(2mL)中,加入4M氯化氢的1,4-二氧六环溶液(0.55mL),反应3小时。反应液减压浓缩,真空干燥,得到粗品标题化合物6h(49mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):634.1[M+1]。
第八步
(±)-N-(35-溴-14-(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)-15-氟-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺6
向25mL单口瓶中加入化合物6h(25mg,0.037mmol)、化合物2a(20mg,0.037mmol)、N,N-二甲基甲酰胺(2mL),搅拌均匀后加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(22mg,0.058mmol),搅拌5分种后加入二异丙基乙胺(39mg,0.301mmol),继续反应1小时。反应液经高效液相制备色谱法(Waters2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:50%-62%,流速:30mL/min)纯化,得到标题化合物6(外消旋体混合物,17mg,产率:44%)。
MS m/z(ESI):1037.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.54(s,1H),9.39(s,1H),8.19(s,1H),8.04(s,1H),7.59(d,1H),7.52(s,1H),7.39(d,1H),7.35(d,1H),7.28(d,1H),7.13-7.05(m,2H),6.71(d,1H),4.43-4.31(m,3H),4.07-3.96(m,4H),3.92(t,2H),3.54-3.26(m,2H),3.25-3.13(m,3H),3.11(s,3H),3.00(s,3H),2.95-2.83(m,2H),2.81-2.62(m,6H),2.47-2.42(m,1H),2.27-2.18(m,1H),2.16-2.05(m,2H),2.03-1.95(m,1H),1.91-1.74(m,3H),1.63-1.54(m,2H),1.37-1.28(m,2H)。
实施例7
(±)-N-(35-溴-14-(4-(2-(4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌啶-1-基)乙酰基)哌嗪-1-基)-16-甲氧基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺7
第一步
(±)-2-(4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌啶-1-基)乙酸 盐酸盐7b
将(±)-2-(4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌啶-1-基)乙酸叔丁酯7a(100mg,0.227mmol,采用专利申请“WO2021127586中说明书第220页的实施例8”公开的方法制备而得)溶于二氯甲烷(2mL)中,加入4M氯化氢的1,4-二氧六环溶液(3mL),30℃反应6小时。将反应液浓缩,真空干燥,得粗品标题化合物7b(外消旋体混合物,94mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):385.3[M+1]。
第二步
(±)-N-(35-溴-14-(4-(2-(4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌啶-1-基)乙酰基)哌嗪-1-基)-16-甲氧基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺7
向25mL单口瓶中加入化合物7b(88mg,0.21mmol)、化合物1h(215mg,0.25mmol)、N,N-二甲基甲酰胺(10mL),搅拌均匀后加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(174mg,0.46mmol)和二异丙基乙胺(89mg,0.69mmol),反应1小时。反应液经高效液相制备色谱法(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%-78%,流速:30mL/min)纯化,得到标题化合物7(外消旋体混合物,85mg,产率:37%)。
MS m/z(ESI):1012.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.91(s,1H),8.17(s,1H),8.10(s,1H),7.60(d,1H),7.46(s,2H),7.43(s,1H),7.35(dd,1H),7.29(d,1H),7.09(dd,1H),7.03(d,1H),6.77(s,1H),4.40-4.30(m,3H),3.96(s,3H),3.78(s,3H),3.71-3.63(m,2H),3.34(s,2H),3.22(s,2H),3.09(s,3H),3.03-2.95(m,4H),2.80-2.75(m,2H),2.72-2.56(m,7H),2.20-2.07(m,5H),1.85-1.71(m,6H),1.58-1.51(m,2H),1.34-1.25(m,2H)。
实施例8
(±)-N-(35-溴-14-(4-(2-(4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌啶-1-基)乙酰基)哌嗪-1-基)-15-氟-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺8
向25mL单口瓶中加入化合物6h(40mg,0.60mmol)、化合物7b(26mg,0.061mmol)、N,N-二甲基甲酰胺(2mL),搅拌均匀后加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(34mg,0.089mmol),搅拌5分钟,加入二异丙基乙胺(46mg,0.36mmol),反应16小时,反应液经高效液相制备色谱法(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)纯化,得到标题化合物8(外消旋体混合物,16mg,产率:27%)。
MS m/z(ESI):1000.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.88(s,1H),9.39(s,1H),8.20(s,1H),8.04(s,1H),7.61(d,1H),7.43-7.37(m,2H),7.35(d,1H),7.28(d,1H),7.10(dd,1H),7.02(d,1H),6.70(dd,1H),4.41-4.28(m,4H),4.18-4.07(m,1H),3.97(s,3H),3.27-3.23(m,1H),3.19-3.06(m,5H),3.00(s,3H),2.98-2.83(m,3H),2.80-2.56(m,7H),2.39-2.29(m,1H),2.21-2.06(m,5H),1.87-1.69(m,6H),1.63-1.54(m,2H),1.37-1.28(m,2H)。
实施例9
(±)-N-(35-溴-14-(4-(2-(4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌嗪-1-基)乙酰基)哌嗪-1-基)-16-甲氧基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺9

第一步
4-(1H-吲唑-6-基)哌嗪-1-羧酸叔丁酯9b
将6-溴-1H-吲唑9a(1g,5.08mmol,上海毕得医药)放入50mL封管,加入甲苯(10mL)、哌嗪-1-羧酸叔丁酯(1.13g,6.07mmol)、2-双环己基膦-2',6'-二异丙氧基联苯(237mg,0.51mmol)、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)(212mg,0.25mmol)、1M双-(三甲基硅基)胺锂的甲苯溶液(12.2mL,12.2mmol,阿达玛斯试剂),氮气鼓泡2分钟,升温至90℃反应12小时。反应液经硅藻土过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物9b(1.5g,产率:98%)。
MS m/z(ESI):303.2[M+1]。
第二步
4-(3-碘-1H-吲唑-6-基)哌嗪-1-羧酸叔丁酯9c
将化合物9b(1g,3.31mmol)溶于N,N-二甲基甲酰胺(10mL),加入氢氧化钾(403mg,7.18mmol)和碘(840mg,3.31mmol),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,依次用水(30mL×2)、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9c(1.04g,产率:73%)。
MS m/z(ESI):429.0[M+1]。
第三步
4-(3-碘-1-甲基-1H-吲唑-6-基)哌嗪-1-羧酸叔丁酯9d
将化合物9c(1.14g,2.66mmol)溶于N,N-二甲基甲酰胺(20mL),加入氢氧化钾(300mg,5.35mmol),冰浴冷却,加入碘甲烷(378mg,2.66mmol),恢复室温反应2小时。反应液中加入水(30mL),用乙酸乙酯(30mL×3)萃取,合并有机相,有机相用饱和氯化钠溶液(30mL×3)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9d(650mg,产率:55%)。
MS m/z(ESI):443.0[M+1]。
第四步
4-(3-(2,6-双(苄氧基)吡啶-3-基)-1-甲基-1H-吲唑-6-基)哌嗪-1-羧酸叔丁酯9f
将2,6-二苄氧基-3-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂戊烷-2-基)吡啶9e(1.1g,2.64mmol,采用专利申请“WO2021127586中说明书第198页的实施例6”公开的方法制备而得)和化合物9d(640mg,1.45mmol)溶于1,4-二氧六环(6mL),加入1,1'-双二苯基膦二茂铁二氯化钯(106mg,0.145mmol)、碳酸铯(1.18g,3.62mmol)和水(2mL)。氮气置换三次,升温到100℃反应2小时。反应液经硅藻土过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化得到标题化合物9f(500mg,产率:57%)。
MS m/z(ESI):606.6[M+1]。
第五步
(±)-4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌嗪-1-羧酸叔丁酯9g
将化合物9f(200mg,0.33mmol)溶于甲醇(5mL),加入10%干钯碳(100mg,0.94mmol)和20%湿氢氧化钯碳(含50%水,100mg,0.71mmol,韶远化学科技),氢气置换三次,氢气氛围下50℃反应18小时。反应液用硅藻土过滤,滤液浓缩,用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物9g(外消旋体混合物,113mg,产率:80%)。
MS m/z(ESI):428.2[M+1]。
第六步
(±)-3-(1-甲基-6-(哌嗪-1-基)-1H-吲唑-3-基)哌啶-2,6-二酮 盐酸盐9h
将化合物9g(113mg,0.264mmol)溶于二氯甲烷(1mL),加入4M氯化氢的1,4-二氧六环溶液(3mL),反应2小时。反应液减压浓缩,真空干燥,得到粗品标题化合物9h(外消旋体混合物,96mg,产率:100%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):328.1[M+1]。
第七步
(±)-2-(4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌嗪-1-基)乙酸叔丁酯9i
将化合物9h(96mg,0.293mmol)溶于N,N-二甲基甲酰胺(2mL)中,加入二异丙基乙胺(113mg,0.874mmol)、2-溴乙酸叔丁酯(86mg,0.441mmol),反应2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(15mL)洗涤,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A纯化,得到标题化合物9i(外消旋体混合物,130mg,产率:100%)。
MS m/z(ESI):442.1[M+1]。
第八步
(±)-2-(4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌嗪-1-基)乙酸 盐酸盐9j
将化合物9i(130mg,0.294mmol)溶于二氯甲烷(1mL)中,加入4M氯化氢的1,4-二氧六环溶液(3mL),30℃反应6小时。将反应液浓缩,真空干燥,得粗品标题化合物9j(外消旋体混合物,123mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):386.3[M+1]。
第九步
(±)-N-(35-溴-14-(4-(2-(4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌嗪-1-基)乙酰基)哌嗪-1-基)-16-甲氧基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺9
向25mL单口瓶中加入化合物9j(33mg,0.078mmol)、化合物1h(52mg,0.076mmol)、N,N-二甲基甲酰胺(2mL),搅拌均匀后加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(35mg,0.092mmol)、二异丙基乙胺(30mg,0.23mmol),反应1小时。反应液经高效液相制备色谱法(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%-57%,流速:30mL/min)纯化,得到标题化合物9(外消旋体混合物,15mg,产率:19%)。
MS m/z(ESI):1013.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.85(s,1H),8.16(s,1H),8.06(s,1H),7.50(d,1H),7.45(s,2H),7.34(d,1H),7.27(d,1H),7.08(dd,1H),6.92(d,1H),6.85(s,1H),6.75(s,1H),4.35(t,2H),4.26(dd,1H),3.89(s,3H),3.79(s,3H),3.70-3.50(m,4H),3.27-3.18(m,4H),3.09(s,3H),2.99(s,3H),2.79-2.57(m,10H),2.37-2.25(m,2H),2.20-2.05(m,3H),2.03-1.95(m,1H),1.81-1.72(m,2H),1.58-1.49(m,2H),1.33-1.26(m,2H)。
实施例10
(35-溴-14-(4-(2-(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)乙酰基)哌嗪-1-基)-16-甲氧基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺10
第一步
2-(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)乙酸 盐酸盐10b
将2-(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)乙酸叔丁酯10a(134mg,0.28mmol,采用专利申请“WO2021127586中说明书第336页的实施例4”公开的方法制备而得)溶于二氯甲烷(0.5mL)中,加入4M氯化氢的1,4-二氧六环溶液(5mL),30℃反应4小时。将反应液浓缩,真空干燥,得粗品标题化合物10b(四个异构体混合物,理论比例1:1:1:1,127mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):421.2[M+1]。
第二步
N-(35-溴-14-(4-(2-(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)乙酰基)哌嗪-1-基)-16-甲氧基-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺10
向25mL单口瓶中加入化合物10b(32mg,0.071mmol)、化合物1h(49mg,0.072mmol)、N,N-二甲基甲酰胺(2mL),搅拌均匀后加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(33mg,0.087mmol)和二异丙基乙胺(28mg,0.22mmol),反应1小时。反应液经高效液相制备色谱法(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)纯化,得到标题化合物10(四个异构体混合物,1:1:1:1,15mg,产率:20%)。
MS m/z(ESI):1048.3[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.89(s,1H),8.16(s,1H),8.07(s,1H),7.65(d,1H),7.53(s,1H),7.47(s,2H),7.35(d,1H),7.28(d,1H),7.12-7.06(m,2H),6.77(s,1H),4.40-4.32(m,3H),3.98(s,3H),3.79(s,3H),3.67-3.49(m,3H),3.42(s,2H),3.30(s,3H),3.26-3.16(m,1H),3.09(s,3H),3.04-2.97(m,4H),2.83-2.57(m,7H),2.40-2.32(m,1H),2.28-2.09(m,4H),1.90-1.73(m,3H),1.59-1.51(m,2H),1.35-1.26 (m,2H)。
实施例11
N-(35-溴-14-(4-(2-(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)乙酰基)哌嗪-1-基)-15-氟-6-氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十一蕃-52-基)-N-甲基甲磺酰胺11
向25mL单口瓶中加入化合物10b(33mg,0.072mmol)、化合物6h(60mg,0.072mmol)、N,N-二甲基甲酰胺(2mL),搅拌均匀后加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(33mg,0.087mmol)、二异丙基乙胺(28mg,0.217mmol),反应1小时。反应液经高效液相制备色谱法(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%-59%,流速:30mL/min)纯化,得到标题化合物11(四个异构体混合物,1:1:1:1,17mg,产率:23%)。
MS m/z(ESI):1036.4[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.92(s,1H),9.42(s,1H),8.20(s,1H),8.06(s,1H),7.65(d,1H),7.52(s,1H),7.42-7.25(m,3H),7.14-7.04(m,2H),6.76-6.65(m,1H),4.46-4.29(m,3H),3.99(s,3H),3.64-3.16(m,11H),3.11(s,3H),3.00(s,3H),2.82-2.58(m,8H),2.27-2.07(m,3H),2.03-1.95(m,1H),1.92-1.72(m,2H),1.64-1.54(m,2H),1.39-1.27(m,2H)。
实施例12
(±)-N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺12
第一步
4-(2-溴-5-甲氧基-4-硝基苯基)哌嗪-1-羧酸叔丁酯12b
将1-溴-2-氟-4-甲氧基-5-硝基苯12a(3g,12.0mmol,江苏艾康生物医药研发有限公司)溶于N,N-二甲基甲酰胺(30mL)中,加入无水碳酸钾(4.14g,30.0mmol)和哌嗪-1-羧酸叔丁酯(2.34g,12.6mmol),升温到90℃反应12小时。将反应液倒入冰水(50mL)中,过滤,收集滤饼,滤饼真空干燥,得到粗品标题化合物12b(4.9g,产率:98%),粗品不经纯化直接用于下一步反应。
MS m/z(ESI):360.1[M-55]。
第二步
4-(5-甲氧基-2-(甲氧基羰基)-4-硝基苯基)哌嗪-1-羧酸叔丁酯12c
称取化合物12b(3g,2.40mmol)、1,1'-双(二苯基膦)二茂铁二氯化钯(II)(527mg,0.72mmol)、三乙胺(7.3g,72.1mmol)到100mL单口瓶中,加入N,N-二甲 基甲酰胺(10mL)和甲醇(10mL)。一氧化碳置换三次,升温到85℃,一氧化碳气氛下反应12小时。待反应液冷却到室温后,加水(25mL),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,无水硫酸钠干燥,过滤,滤液浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系B纯化得到标题化合物12c(1.98g,产率:69%)。
MS m/z(ESI):396.1[M+1]。
第三步
4-(4-氨基-5-甲氧基-2-(甲氧基羰基)苯基)哌嗪-1-羧酸叔丁酯12d
将化合物12c(500mg,1.26mmol)溶于甲醇(15mL),加入10%干钯碳(134mg,1.26mmol)。氢气置换三次,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,得到粗品标题化合物12d(460mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):366.5[M+1]。
第四步
4-(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-(甲氧基羰基)苯基)哌嗪-1-羧酸叔丁酯12e
称取化合物12d(460mg,1.26mmol)、化合物1d(513mg,1.26mmol)到50mL单口瓶中,加入异丙醇(40mL)和三氟乙酸(287mg,2.51mmol),升温到90℃反应12小时。待反应液冷却到室温,减压浓缩除去溶剂,所得残留物用硅胶柱色谱法以洗脱剂体系B纯化得到标题化合物12e(130mg,产率:14%)。
MS m/z(ESI):736.1[M+1]。
第五步
5-((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)-2-(4-(叔丁氧羰基)哌嗪-1-基)-4-甲氧基苯甲酸12f
称取化合物12e(130mg,0.18mmol)到50mL单口瓶中,加入6mL甲醇、四氢呋喃和水的混合溶剂(V/V/V=1/1/1)。然后加入氢氧化钠(35mg,,0.86mmol),升温到60℃反应12小时。待反应液冷却到室温,加入1M盐酸溶液调pH到6-7之间,浓缩反应液,所得残留物经高效液相制备色谱法(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)纯化,得到标题化合物12f(60mg,产率:47%)。
MS m/z(ESI):722.3[M+1]。
第六步
4-(35-溴-16-甲氧基-52-(N-甲基甲磺酰胺基)-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-14-基)哌嗪-1-羧酸叔丁酯12g
将化合物12f(50mg,0.069mmol)溶于N,N-二甲基甲酰胺(2mL),加入1,4-二溴丁烷(30mg,0.139mmol)、无水碳酸钾(29mg,0.21mmol)和四丁基碘化铵(13 mg,0.35mmol),升温到60℃反应3小时。加水(5mL),用乙酸乙酯萃取(10mL×3),合并有机相,用饱和氯化钠溶液(15mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系B纯化得到标题化合物12g(50mg,产率:93%)。
MS m/z(ESI):776.3[M+1]。
第七步
N-(35-溴-16-甲氧基-12-氧代-14-(哌嗪-1-基)-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺盐酸盐12h
将化合物12g(50mg,0.064mmol)溶于二氯甲烷(3mL)中,加入4M氯化氢的1,4-二氧六环溶液(2mL),反应30分钟。反应液减压浓缩,真空干燥,得到粗品标题化合物12h(45mg,产率:98%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):676.2[M+1]。
第八步
(±)-N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮
杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺12
向25mL单口瓶中加入化合物12h(45mg,0.063mmol)、化合物2a(30mg,0.056mmol)、N,N-二甲基甲酰胺(2mL),搅拌均匀后加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(24mg,0.063mmol),搅拌5分种后加入二异丙基乙胺(24mg,0.19mmol),继续反应1小时。反应液经高效液相制备色谱法(Waters2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:45%-52%,流速:30mL/min)纯化,得到标题化合物12(外消旋体混合物,16mg,产率:23%)。
MS m/z(ESI):1079.5[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.56(s,1H),8.18(s,1H),7.99(s,1H),7.81(s,1H),7.76(s,1H),7.59(d,1H),7.53(s,1H),7.51(d,1H),7.14(d,1H),7.08(d,1H),6.85(dd,1H),6.73(s,1H),4.44(t,2H),4.30(t,2H),3.98(s,3H),3.92(t,2H),3.85(s,3H),3.68-3.17(m,8H),3.10(s,3H),3.03(s,3H),2.97-2.92(m,2H),2.90-2.85(m,2H),2.76(t,3H),2.32-1.83(m,2H),2.02-1.95(m,1H),1.91-1.83(m,3H),1.76-1.68(m,2H)。
实施例13
(±)-N-(35-溴-14-(4-(2-(4-(3-(2,6-二氧代哌啶-3-基)-1-甲基-1H-吲唑-6-基)哌啶-1-基)乙酰基)哌嗪-1-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺13
向25mL单口瓶中加入化合物12h(100mg,0.14mmol)、化合物7b(59mg,0.14mmol)、N,N-二甲基甲酰胺(3mL),搅拌均匀后加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(53mg,0.14mmol),搅拌5分种后加入二异丙基乙胺(54mg,0.42mmol),继续反应1小时。反应液经高效液相制备色谱法(Waters2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:49%-54%,流速:30mL/min)纯化,得到标题化合物13(外消旋体混合物,25mg,产率:17%)。
MS m/z(ESI):1042.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.91(s,1H),8.18(s,1H),7.99(s,1H),7.83(s,1H),7.76(s,1H),7.60(d,1H),7.51(d,1H),7.43(s,1H),7.15(d,1H),7.03(d,1H),6.84(dd,1H),6.73(s,1H),4.48-4.42(m,2H),4.35-4.26(m,3H),3.96(s,3H),3.84(s,3H),3.72-3.65(s,2H),3.58-3.52(m,2H),3.33(s,3H),3.24-3.20(m,1H),3.10(s,3H),3.03(s,3H),3.00-2.93(m,3H),2.89-2.84(m,2H),2.71-2.53(m,3H),2.40-2.27(m,2H),2.20-2.11(m,3H),1.89-1.67(m,6H)。
实施例14
(±)-N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺14

向25mL单口瓶中加入化合物12h(100mg,0.14mmol)、化合物1i(64mg,0.14mmol)、N,N-二甲基甲酰胺(3mL),搅拌均匀后加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(53mg,0.14mmol),搅拌5分种后加入二异丙基乙胺(54mg,0.42mmol),继续反应1小时。反应液经高效液相制备色谱法(Waters2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:49%-56%,流速:30mL/min)纯化,得到标题化合物14(外消旋体混合物,25mg,产率:18%)。
MS m/z(ESI):1003.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.80(s,1H),8.18(s,1H),7.99(s,1H),7.82(s,1H),7.76(s,1H),7.51(d,1H),7.15(d,1H),6.94(d,2H),6.84(dd,1H),6.71(s,1H),6.59(d,2H),5.68(d,1H),4.48-4.40(m,2H),4.33-4.22(m,3H),3.85(s,3H),3.70-3.62(m,2H),3.57-3.50(m,2H),3.33(s,3H),3.21-3.14(m,2H),3.10(s,3H),3.03(s,3H),2.98-2.82(m,5H),2.77-2.68(m,2H),2.62-2.53(m,2H),2.38-2.25(m,1H),2.15-2.03(m,2H),1.89-1.82(m,2H),1.74-1.64(m,3H),1.63-1.52(m,1H)。
实施例15
(±)-N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺15

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15a
称取化合物12a(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),然后于90℃反应12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15a(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-2-(甲氧基羰基)-4-硝基苯基)哌啶-4-基)哌嗪-1-羧酸叔丁酯15b
称取化合物15a(1.5g,3.00mmol)、1,1'-双(二苯基膦)二茂铁二氯化钯(II)(220mg,0.30mmol)、三乙胺(3.04g,30.0mmol)到100mL单口瓶中,加入N,N-二甲基甲酰胺和甲醇的混合溶剂(30mL,V/V=1/1)。一氧化碳置换三次,升温到85℃,一氧化碳气氛下反应12小时。待反应液冷却到室温后,加水(25mL),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(25mL),无水硫酸钠干燥,过滤,滤液浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标 题化合物15b(1.3g,产率:90%)。
MS m/z(ESI):479.2[M+1]。
第三步
4-(1-(4-氨基-5-甲氧基-2-(甲氧基羰基)苯基)哌啶-4-基)哌嗪-1-羧酸叔丁酯15c
将化合物15b(1.2g,2.51mmol)溶于甲醇(30mL),加入10%干钯碳(267mg,2.51mmol)。氢气置换三次,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15c(1.12g,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):449.2[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-(甲氧基羰基)苯基)哌啶-4-基)哌嗪-1-羧酸叔丁酯15d
称取化合物15c(1.12g,2.49mmol)、化合物1d(1.02g,2.50mmol)到100mL单口瓶中,加入异丙醇(120mL)和三氟乙酸(855mg,7.50mmol),升温到90℃反应12小时。待反应液冷却到室温,减压浓缩除去溶剂,所得残留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(2.0g,产率:98%)。
MS m/z(ESI):819.0[M+1]。
第五步
5-((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)-2-(4-(4-(叔丁氧基羰基)哌嗪-1-基)哌啶-1-基)-4-甲氧基苯甲酸15e
称取化合物15d(2.0g,2.44mmol)到250mL单口瓶中,加入甲醇、四氢呋喃和水的混合溶剂(30mL,V/V/V=1/1/1)。然后加入氢氧化钠(488mg,12.2mmol),升温到60℃反应12小时。待反应液冷却到室温,加入1M盐酸溶液调节反应液pH到6-7之间,浓缩反应液,所得残留物经高效液相制备色谱法(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)纯化,得到标题化合物15e(500mg,产率:25%)。
MS m/z(ESI):805.0[M+1]。
第六步
4-(1-(35-溴-16-甲氧基-52-(N-甲基甲磺酰胺基)-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-14-基)哌啶-4-基)哌嗪-1-羧酸叔丁酯15f
将化合物15e(350mg,0.43mmol)溶于N,N-二甲基甲酰胺(15mL),加入1,4-二溴丁烷(122mg,0.56mmol)、无水碳酸钾(181mg,1.31mmol)和四丁基碘化铵(49mg,0.13mmol),升温到60℃反应12小时。加水(5mL),用乙酸乙酯萃取(10mL×3),合并有机相,用饱和氯化钠溶液(15mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物15f(360mg,产 率:96%)。
MS m/z(ESI):859.2[M+1]。
第七步
N-(35-溴-16-甲氧基-12-氧代-14-(4-(哌嗪-1-基)哌啶-1-基)-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺 二盐酸盐15g
将化合物15f(370mg,0.43mmol)溶于二氯甲烷(8mL)中,加入4M氯化氢的1,4-二氧六环溶液(2.5mL),反应30分钟。反应液减压浓缩,真空干燥,得到粗产品标题化合物15g(350mg,产率:98%),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
(±)-N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺15
向25mL单口瓶中加入化合物15g(45mg,0.054mmol)、(±)-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15h(22mg,0.060mmol,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),用甲醇和二氯甲烷的混合溶剂(4mL,V/V=1/1)溶解,搅拌均匀后加入无水乙酸钠(50mg,0.61mmol)和乙酸(8mg,0.13mmol),反应30分钟。加入氰基硼氢化钠(11mg,0.18mmol),继续反应2小时。反应液经高效液相制备色谱法(Waters2545,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)纯化,得到标题化合物15(外消旋体混合物,30mg,产率:46%)。
MS m/z(ESI):1112.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),8.16(s,1H),7.95(s,1H),7.78(s,1H),7.70(d,1H),7.64(d,1H),7.51(d,1H),7.30(s,1H),7.22(dd,1H),7.13(d,1H),6.82(dd,1H),6.65(s,1H),5.06(dd,1H),4.44-4.36(m,2H),4.32-4.26(m,2H),4.08-3.98(m,2H),3.84(s,3H),3.30(s,4H),3.17-3.11(m,2H),3.10(s,3H),3.03(s,3H),2.96(t,2H),2.92-2.83(m,1H),2.68(t,2H),2.64-2.55(m,3H),2.40-2.29(m,3H),2.28-2.20(m,1H),2.15-2.09(m,2H),2.05-1.97(m,1H),1.89-1.67(m,9H),1.52-1.42(m,2H),1.19-1.07(m,2H)。
实施例16
(±)-N-(35-氯-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺16
采用实施例15的合成路线,将第四步原料化合物1d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16a(采用专利申请“WO2021216440中说明书第58页的实施例A2”公开的方法制备而得),得到标题化合物16(外消旋体混合物,18mg,产率:40%)。
MS m/z(ESI):1068.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),8.11-8.06(m,2H),7.76(s,1H),7.71(s,1H),7.65(d,1H),7.50(d,1H),7.30(d,1H),7.22(dd,1H),7.12(d,1H),6.83(dd,1H),6.65(s,1H),5.06(dd,1H),4.44-4.37(m,2H),4.33-4.26(m,2H),4.07-3.99(m,2H),3.84(s,3H),3.30(s,4H),3.19-3.11(m,2H),3.09(s,3H),3.02(s,3H),2.96(t,2H),2.92-2.83(m,1H),2.68(t,2H),2.64-2.54(m,3H),2.42-2.21(m,4H),2.17-2.09(m,2H),2.05-1.97(m,1H),1.89-1.67(m,9H),1.52-1.42(m,2H),1.19-1.08(m,2H)。
实施例17
(±)-N-(35-溴-14-(4-(4-((1-(2-(2,6-二氧代哌啶-3-基)-6-氟-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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺17
第一步
5-(4-(二甲氧基甲基)哌啶-1-基)-2-(2,6-二氧代哌啶-3-基)-6-氟异吲哚啉-1,3-二酮 17b
将2-(2,6-二氧代哌啶-3-基)-5,6-二氟异吲哚啉-1,3-二酮17a(800mg,2.72mmol,上海毕得医药)溶于N-甲基吡咯烷酮(10mL)中,加入碳酸氢钠(275mg,3.27mmol)、4-(二甲氧基甲基)哌啶(433mg,2.72mmol,韶远化学科技),升温到90℃反应12小时。待反应液冷却到室温后,减压浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物17b(600mg,产率:51%)。
MS m/z(ESI):434.1[M+1]。
第二步
1-(2-(2,6-二氧代哌啶-3-基)-6-氟-1,3-二氧代异吲哚啉-5-基)哌啶-4-甲醛17c
称取化合物17b(300mg,0.69mmol)到100mL单口瓶中,加入6mL四氢呋喃,再加入2M稀硫酸溶液(3mL),反应12小时。加入碳酸氢钠固体,调节反应液pH至7-8,加入无水硫酸钠干燥,过滤,滤液浓缩,真空干燥,得到粗产品标题化合物17c(260mg,产率:97%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):388.1[M+1]。
第三步
(±)-N-(35-溴-14-(4-(4-((1-(2-(2,6-二氧代哌啶-3-基)-6-氟-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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺17
向25mL单口瓶中加入化合物15g(40mg,0.048mmol)、化合物17c(21mg,0.054mmol),用甲醇和二氯甲烷的混合溶剂(2mL,V/V=1/1)溶解,搅拌均匀后加入无水乙酸钠(22mg,0.27mmol)和乙酸(6.6mg,0.11mmol),反应30分钟,加入氰基硼氢化钠(10mg,0.17mmol),继续反应2小时。反应液经高效液相制备色谱法(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)纯化,得到标题化合物17(外消旋体混合物,10mg,产率:17%)。
MS m/z(ESI):1130.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.08(s,1H),8.16(s,1H),7.95(s,1H),7.78(s,1H),7.73-7.67(m,2H),7.51(d,1H),7.43(d,1H),7.13(d,1H),6.82(dd,1H),6.65(s,1H),5.10(dd,1H),4.45-4.37(m,2H),4.33-4.25(m,2H),3.84(s,3H),3.64-3.57(m,2H),3.30(s,4H),3.18-3.12(m,2H),3.10(s,3H),3.03(s,3H),2.93-2.84(m,3H),2.72-2.57(m,6H),2.44-2.31(m,2H),2.28-2.21(m,1H),2.17(d,2H),2.06-2.00(m,1H),1.91-1.67(m,9H),1.52-1.42(m,2H),1.30-1.20(m,2H)。
实施例18
(±)-N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-环丙基甲磺酰胺18
第一步
N-环丙基甲磺酰胺18b
将环丙胺18a(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)和吡啶(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18b(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18d
将化合物18b(440mg,3.25mmol)和2-氟-4-甲氧基-1-硝基苯18c(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,所得残留物用硅胶柱色谱 法以洗脱剂体系B纯化得到标题化合物18d(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18e
将化合物18d(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,韶远化学科技)。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18e(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18g
将化合物18e(400mg,1.56mmol)和5-溴-2,4-二氯嘧啶18f(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,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物18g(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18h
将化合物18g(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,用饱和氯化钠溶液(15mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物18h(280mg,产率:96%)。
MS m/z(ESI):432.8[M+1]。
第六步
4-(1-(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-(甲氧基羰基)苯基)哌啶-4-基)哌嗪-1-羧酸叔丁酯18i
称取化合物18h(95mg,0.22mmol)、化合物15c(98mg,0.22mmol)到50mL单口瓶中,加入异丙醇(3mL)和三氟乙酸(75mg,0.66mmol),升温到100℃反应12小时。待反应液冷却到室温,减压浓缩除去溶剂,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物18i(135mg,产率:73%)。
MS m/z(ESI):845.2[M+1]。
第七步
5-((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)-2-(4-(4-(叔丁氧基羰基)哌嗪-1-基)哌啶-1-基)-4-甲氧基苯甲酸18j
称取化合物18i(135mg,0.16mmol)到50mL单口瓶中,加入6mL甲醇、四氢呋喃和水的混合溶剂(V/V/V=1/1/1)。然后加入氢氧化钠(40mg,1.00mmol), 升温到60℃反应12小时。待反应液冷却到室温,加入1M盐酸溶液调节反应液pH到6-7之间,浓缩反应液,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物18j(70mg,产率:53%)。
MS m/z(ESI):831.2[M+1]。
第八步
4-(1-(35-溴-52-(N-环丙基甲磺酰胺基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-14-基)哌啶-4-基)哌嗪-1-羧酸叔丁酯18k
将化合物18j(70mg,0.08mmol)溶于N,N-二甲基甲酰胺(2mL),加入1,4-二溴丁烷(19mg,0.09mmol)、无水碳酸钾(35mg,0.25mmol)和四丁基碘化铵(9mg,0.02mmol),升温到60℃反应4小时。加水(5mL),用乙酸乙酯萃取(10mL×3),合并有机相,用饱和氯化钠溶液(15mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物18k(56mg,产率:75%)。
MS m/z(ESI):885.0[M+1]。
第九步
N-(35-溴-16-甲氧基-12-氧代-14-(4-(哌嗪-1-基)哌啶-1-基)-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-环丙基甲磺酰胺 二三氟乙酸盐18l
将化合物18k(56mg,0.06mmol)溶于二氯甲烷(3mL)中,加入三氟乙酸(0.5mL),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18l(63mg,产率:98%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):785.0[M+1]。
第十步
(±)-N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-环丙基甲磺酰胺18
将化合物18l(63mg,0.062mmol)、无水乙酸钠(51mg,0.62mmol)溶于4mL二氯甲烷和甲醇的混合溶剂(V/V=3/1)中,搅拌10分钟,随后加入化合物15h(35mg,0.095mmol)和乙酸(12mg,0.20mmol),反应1小时,接着加入氰基硼氢化钠(9mg,0.15mmol),反应1小时。反应液经高效液相制备色谱法(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)纯化,得到标题化合物18(外消旋体混合物,15mg,产率:21%)。
MS m/z(ESI):1138.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),8.16(s,1H),7.80(s,1H),7.68(s,1H),7.65(d,1H),7.62(s,1H),7.50(d,1H),7.30(s,1H),7.22(d,1H),7.17(d,1H),6.80(dd,1H),6.67(s,1H),5.06(dd,1H),4.48-4.40(m,1H),4.37-4.24(m,3H), 4.09-3.99(m,2H),3.85(s,3H),3.21-3.10(m,6H),2.96(t,2H),2.91-2.83(m,1H),2.74-2.65(m,2H),2.62-2.50(m,6H),2.42-2.20(m,4H),2.16-2.09(m,2H),2.04-1.96(m,2H),1.94-1.74(m,8H),1.71-1.62(m,1H),1.54-1.42(m,2H),1.19-1.08(m,2H),0.83-0.72(m,2H),0.50-0.41(m,1H),0.13-0.05(m,1H)。
实施例19
N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺19
向25mL单口瓶中依次加入N,N-二甲基甲酰胺(2mL)、化合物15g(45mg,0.054mmol)、(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19a(21mg,0.061mmol,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,非对映异构体混合物)、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(30mg,0.078mmol),反应20分钟。反应液过滤,经高效液相制备色谱法(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)纯化,得到标题化合物19(2个非对映异构体的混合物,比例1:1,35mg,产率:60%)。
MS m/z(ESI):1086.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.10(s,1H),8.16(s,1H),7.95(s,1H),7.78(s,1H),7.70(s,1H),7.62(dd,1H),7.51(d,1H),7.22(d,1H),7.18-7.14(m,1H),7.13(d,1H),7.07(d,1H),6.82(dd,1H),6.66(s,1H),5.06(dd,1H),4.93-4.85(m,1H),4.45-4.36(m,2H),4.32-4.25(m,2H),3.84(s,3H),3.73-3.38(m,4H),3.19-3.13(m,2H),3.10(s,3H),3.03(s,3H),2.93-2.84(m,1H),2.69(t,2H),2.64-2.43(m,6H),2.38-2.30(m,1H),2.08-1.95(m,1H),1.91-1.78(m,4H),1.75-1.67(m,2H),1.55-1.43(m,2H),1.29(d,3H)。
实施例20
N-(35-溴-14-(4-((1-((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) 哌啶-4-基)甲基)哌嗪-1-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺20
第一步
4-(((S)-1-(4-(二甲氧基甲基)哌啶-1-基)-1-氧代丙-2-基)氨基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮20a
向25mL单口瓶中依次加入N,N-二甲基甲酰胺(4mL)、化合物19a(150mg,0.43mmol)、4-(二甲氧基甲基)哌啶(105mg,0.66mmol)、O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(248mg,0.65mmol),冰浴下,缓慢加入N,N-二异丙基乙胺(225mg,1.74mmol),反应1小时。浓缩反应液,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物20a(2个非对映异构体的混合物,比例:1:1,210mg,产率:99%)。
MS m/z(ESI):487.1[M+1]。
第二步
1-((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)哌啶-4-甲醛20b
称取化合物20a(210mg,0.43mmol)到100mL单口瓶中,加入10mL四氢呋喃,再加入2M稀硫酸溶液(2.2mL),升温到60℃反应1小时。待反应液冷却到室温后,加入碳酸氢钠固体,调节反应液pH至7-8,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洗涤(20mL),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20b(2个非对映异构体的混合物,比例:1:1,190mg,产率:100%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):441.1[M+1]。
第三步
N-(35-溴-14-(4-((1-((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)哌啶-4-基)甲基)哌嗪-1-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺20
将化合物12h(60mg,0.084mmol)溶于二氯甲烷和甲醇(9mL,V/V=1/1)的混合溶剂中,加入无水乙酸钠(56mg,0.68mmol),反应10分钟。加入化合物20b(38mg,0.086mmol)和乙酸(16mg,0.27mmol),反应20分钟。加入氰基硼氢化钠(15mg,0.25mmol),反应半小时。反应液经高效液相制备色谱法(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:49%-56%,流速:30mL/min)得到标题化合物20(2个非对映异构体的混合物,比例:1:1,48mg,产率:52%)。
MS m/z(ESI):1100.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.10(s,1H),8.17(s,1H),7.96(s,1H),7.79(s,1H),7.71(s,1H),7.63-7.58(m,1H),7.50(d,1H),7.25-7.15(m,2H),7.13(d,1H),7.06(d,1H),6.83(d,1H),6.70(d,1H),5.06(dd,1H),4.91-4.82(m,1H),4.45-4.34(m,3H),4.32-4.26(m,2H),4.12-4.02(m,1H),3.86(s,3H),3.29(s,4H),3.09(s,3H),3.03(s,3H),2.95-2.84(m,4H),2.70-2.35(m,7H),2.24-2.14(m,2H),2.06-2.00(m,1H),1.90-1.67(m,7H),1.28(t,3H)。
实施例21
(±)-N-(35-溴-14-(4-((9-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)-3,9-二氮杂螺[5.5]十一烷-3-基)甲基)哌啶-1-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺21

第一步
1-(2-溴-5-甲氧基-4-硝基苯基)-4-(二甲氧基甲基)哌啶21a
将化合物12a(2.65g,10.6mmol)和4-(二甲氧基甲基)哌啶(2.65g,16.6mmol)溶于50mL N,N-二甲基甲酰胺中,加入碳酸钾(2.93g,21.2mmol),升温到90℃反应12小时。向反应液中加入水(50mL),用乙酸乙酯萃取(50mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(50mL×2),无水硫酸钠干燥,过滤,滤液浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系B纯化得到标题化合物21a(3.5g,产率:85%)。
MS m/z(ESI):389.0[M+1]。
第二步
2-(4-(二甲氧基甲基)哌啶-1-基)-4-甲氧基-5-硝基苯甲酸甲酯21b
将化合物21a(3g,7.71mmol)溶于N,N-二甲基甲酰胺和甲醇的混合溶剂(30mL,V/V=1/2)中,氮气鼓泡5分钟,加入1,1'-双二苯基膦二茂铁二氯化钯(564mg,0.77mmol)、三乙胺(7.8g,77mmol)。一氧化碳气体置换三次,升温到85℃,一氧化碳氛围下反应18小时。待反应液冷却至室温,硅藻土过滤,滤液浓缩,所得 残留物用硅胶柱色谱法以洗脱剂体系B纯化得到标题化合物21b(2.6g,产率:91%)。
MS m/z(ESI):369.2[M+1]。
第三步
5-氨基-2-(4-(二甲氧基甲基)哌啶-1-基)-4-甲氧基苯甲酸甲酯21c
将化合物21b(2.6g,7.06mmol)溶于75mL甲醇、四氢呋喃的混合溶剂(V/V=2/1)中,加入10%干钯碳(751mg,7.06mmol),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,所得残留物用硅胶柱色谱法以洗脱剂体系B纯化得到标题化合物21c(1.3g,产率:54%)。
MS m/z(ESI):339.3[M+1]。
第四步
5-((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)-2-(4-(二甲氧基甲基)哌啶-1-基)-4-甲氧基苯甲酸甲酯21d
将化合物21c(500mg,1.45mmol)、化合物1d(602mg,1.47mmol)加入到甲醇(10mL)中,加入三氟乙酸(337mg,2.96mmol),升温90℃反应48小时。浓缩反应液,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物21d(455mg,产率:43%)。
MS m/z(ESI):709.1[M+1]。
第五步
5-((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)-2-(4-(二甲氧基甲基)哌啶-1-基)-4-甲氧基苯甲酸21e
将化合物21d(455mg,0.641mmol)加入15mL甲醇、四氢呋喃和水的混合溶剂(V/V/V=1/1/1)中。然后加入氢氧化钠(128mg,3.2mmol),升温到50℃反应12小时。待反应液冷却到室温,加入1M盐酸溶液调节反应液pH到6-7之间,浓缩反应液,所得残留物经高效液相制备色谱法(Waters 2545,洗脱体系:10mmol/L碳酸氢铵的水溶液和乙腈,乙腈的梯度:33%-53%,流速:30mL/min)纯化,得到标题化合物21e(100mg,产率:22%)。
MS m/z(ESI):695.0[M+1]。
第六步
N-(35-溴-14-(4-(二甲氧基甲基)哌啶-1-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺21f
将化合物21e(100mg,0.14mmol)溶于N,N-二甲基甲酰胺(5mL),加入1,4-二溴丁烷(37mg,0.17mmol)、碳酸钾(60mg,0.73mmol)、四丁基碘化铵(20mg,0.054mmol),升温到60℃反应12小时。待反应液冷却到室温,加水(25mL),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(25mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯 化得到标题化合物21f(90mg,产率:84%)。
MS m/z(ESI):749.1[M+1]。
第七步
N-(35-溴-14-(4-甲酰基哌啶-1-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺21g
将化合物21f(97mg,0.13mmol)溶于四氢呋喃(10mL),加入2M硫酸溶液(0.2mL),升温到30℃反应3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,有机相用饱和碳酸氢钠溶液洗涤,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21g(90mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):703.1[M+1]。
第八步
(±)-9-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)-3,9-二氮杂螺[5.5]十一
烷-3-羧酸叔丁酯21i
将(±)-2-(2,6-二氧代哌啶-3-基)-5-氟异吲哚啉-1,3-二酮21h(150mg,0.54mmol,上海毕得医药)溶于N-甲基吡咯烷酮(3mL),加入3,9-二氮杂螺[5.5]十一烷-3-羧酸叔丁酯(152mg,0.6mmol,南京药石科技股份有限公司)、N,N-二异丙基乙胺(10mg,1.62mmol),微波140℃反应2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,有机相用饱和碳酸氢钠溶液洗涤,无水硫酸钠干燥,过滤,滤液浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物21i(250mg,产率:90%)。
MS m/z(ESI):511.2[M+1]。
第九步
(±)-2-(2,6-二氧代哌啶-3-基)-5-(3,9-二氮杂螺[5.5]十一烷-3-基)异吲哚啉-1,3-二酮盐酸盐21j
将化合物21i(250mg,0.49mmol)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30分钟。反应液减压浓缩,真空干燥,得到粗产品标题化合物21j(200mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):411.6[M+1]。
第十步
(±)-N-(35-溴-14-(4-((9-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)-3,9-二氮杂螺[5.5]十一烷-3-基)甲基)哌啶-1-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮
杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺21
称取化合物21j(16mg,0.036mmol)到25mL茄形瓶中,加入4mL二氯甲烷、甲醇的混合溶剂(V/V=3/1),加入无水乙酸钠(12mg,0.15mmol),反应15分钟,加入化合物21g(20mg,0.028mmol),反应30分钟,加入氰基硼氢化钠(4mg,0.067 mmol),反应1小时。反应液过滤,经高效液相制备色谱法(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)纯化,得到标题化合物21(外消旋体混合物,10mg,产率:32%)。
MS m/z(ESI):1097.2[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.07(s,1H),8.16(s,1H),7.95(s 1H),7.77(s,1H),7.69(s,1H),7.65(d,1H),7.51(d,1H),7.30(d,1H),7.21(m,1H),7.13(d,1H),6.82(m,1H),6.66(s,1H),5.06(dd,1H),4.45-4.35(m,2H),4.32-4.25(m,2H),3.84(s,3H),3.51-3.42(m,4H),3.14-3.06(m,5H),3.03(s,3H),2.93-2.83(m,1H),2.67(t,2H),2.62-2.53(m,2H),2.41-2.13(m,5H),2.05-1.96(m,2H),1.89-1.81(m,2H),1.79-1.66(m,4H),1.65-1.40(m,9H),1.20-1.11(m,2H)。
实施例22
(±)-N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺22
采用实施例21的方法,将第八步原料3,9-二氮杂螺[5.5]十一烷-3-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),制得标题化合物22(外消旋体混合物,13mg,产率:27%)。
MS m/z(ESI):1111.4[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.07(s,1H),8.16(s,1H),7.96(s 1H),7.78(s,1H),7.70(s,1H),7.65(d,1H),7.51(d,1H),7.31(s,1H),7.23(d,1H),7.12(d,1H),6.82(m,1H),6.67(s,1H),5.06(dd,1H),4.42-4.36(m,2H),4.33-4.26(m,2H),4.13-4.05(m,2H),3.84(s,3H),3.15-3.06(m,5H),3.03(s,3H),2.95-2.83(m,4H),2.74-2.53(m,12H),2.05-1.95(m,2H),1.89-1.63(m,10H),1.50-1.14(m,5H)。
实施例23
(±)-N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺23
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23a
将化合物12a(1g,3.99mmol)溶于N,N-二甲基甲酰胺(20mL)中,加入无水碳酸钾(1.10g,7.95mmol)、2,7-二氮杂螺[3.5]壬烷-2-羧酸叔丁酯(906mg,4.00mmol),升温到90℃反应16小时。将反应液过滤,浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物23a(1.7g,产率:93%)。
MS m/z(ESI):400.0[M-55]。
第二步
7-(5-甲氧基-2-(甲氧基羰基)-4-硝基苯基)-2,7-二氮杂螺[3.5]壬烷-2-羧酸叔丁酯23b
称取化合物23a(600mg,1.21mmol)、1,1'-双(二苯基膦)二茂铁二氯化钯(II)(144mg,0.19mmol)、三乙胺(1.33g,13.1mmol)到100mL单口瓶中,加入N,N-二甲基甲酰胺(12mL)和甲醇(12mL)。一氧化碳置换三次,升温到85℃,一氧化碳 气氛下反应16小时。待反应液冷却到室温后,加水(25mL),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,无水硫酸钠干燥,过滤,滤液浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物23b(320mg,产率:55%)。
MS m/z(ESI):436.5[M+1]。
第三步
7-(4-氨基-5-甲氧基-2-(甲氧基羰基)苯基)-2,7-二氮杂螺[3.5]壬烷-2-羧酸叔丁酯23c
将化合物23b(340mg,0.78mmol)溶于甲醇(20mL),加入10%湿钯碳(含55%质量分数水,83mg,0.077mmol,韶远化学科技)。氢气置换三次,氢气氛下反应3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23c(280mg,产率:88%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):406.1[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-(甲氧基羰基)苯基)-2,7-二氮杂螺[3.5]壬烷-2-羧酸叔丁酯23d
称取化合物23c(280mg,0.69mmol)、化合物1d(253mg,0.62mmol)到50mL单口瓶中,加入正丁醇(12mL)和三氟乙酸(142mg,1.24mmol),升温到90℃反应16小时。待反应液冷却到室温,减压浓缩除去溶剂,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物23d(300mg,产率:55%)。
MS m/z(ESI):774.0[M-1]。
第五步
5-((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)-2-(2-(叔丁氧基羰基)-2,7-二氮杂螺[3.5]壬烷-7-基)-4-甲氧基苯甲酸23e
称取化合物23d(300mg,0.31mmol)到50mL单口瓶中,加入10mL甲醇。然后加入2M氢氧化钠溶液(1.55mL),升温到60℃反应16小时。待反应液冷却到室温,加入1M盐酸溶液调节反应液pH到4左右,加水(20mL),用二氯甲烷萃取(35mL×3),合并有机相,减压除去溶剂,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物23e(230mg,产率:97%)。
MS m/z(ESI):762.0[M+1]。
第六步
7-(35-溴-16-甲氧基-52-(N-甲基甲磺酰胺基)-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-14-基)-2,7-二氮杂螺[3.5]壬烷-2-羧酸叔丁酯23f
将化合物23e(220mg,0.28mmol)溶于N,N-二甲基甲酰胺(10mL),加入1,4-二溴丁烷(63mg,0.29mmol)、无水碳酸钾(120mg,0.86mmol)和四丁基碘化铵(15mg,0.086mmol),升温到60℃反应6小时。待反应液冷至室温,加水(15mL), 用乙酸乙酯萃取(20mL×3),合并有机相,用饱和氯化钠溶液(15mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物23f(108mg,产率:45%)。
MS m/z(ESI):816.1[M+1]。
第七步
N-(35-溴-16-甲氧基-12-氧代-14-(2,7-二氮杂螺[3.5]壬烷-7-基)-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺三氟乙酸盐23g
将化合物23f(60mg,0.073mmol)溶于二氯甲烷(2mL)中,冰浴下滴加三氟乙酸(600mg,5.26mmol),滴完后反应1小时。反应液减压浓缩,真空干燥,得到粗产品标题化合物23g(60mg,产率:99%),粗产品不经纯化直接用于下一步反应。
MS m/z(ESI):714.0[M-1]。
第八步
(±)-N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺23
向25mL单口瓶中加入化合物23g(60mg,0.073mmol)、二氯甲烷(3mL)和甲醇(1mL),搅拌均匀后加入无水乙酸钠(62mg,0.076mmol),搅拌10分钟后加入化合物15h(48mg,0.11mmol)和冰乙酸(15mg,0.25mmol),反应1小时,再加入氰基硼氢化钠(10mg,0.17mmol),反应1小时。反应液经高效液相制备色谱法(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%-85%,流速:30mL/min)纯化,得到标题化合物23(外消旋体混合物,50mg,产率:63%)。
MS m/z(ESI):1069.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),8.16(s,1H),7.96(s,1H),7.76(s,1H),7.70(s,1H),7.64(d,1H),7.50(d,1H),7.29(d,1H),7.21(dd,1H),7.13(d,1H),6.82(dd,1H),6.66(s,1H),5.06(dd,1H),4.44-4.34(m,2H),4.33-4.23(m,2H),4.01(d,2H),3.84(s,3H),3.09(s,3H),3.03(s,3H),2.98-2.83(m,7H),2.82-2.72(m,4H),2.62-2.52(m,2H),2.33-2.25(m,2H),2.05-1.95(m,2H),1.89-1.80(m,2H),1.79-1.65(m,7H),1.61-1.51(m,1H),1.20-1.09(m,2H)。
实施例24
(±)-N-(35-溴-14-(4-(2-(1-(4-((2,6-二氧代哌啶-3-基)氨基)-2-氟苯基)-4-羟基哌啶-4-基)乙酰基)哌嗪-1-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺24
向25mL单口瓶中依次加入N,N-二甲基甲酰胺(2mL)、化合物12h(30mg,0.042mmol)、(±)-2-(1-(4-((2,6-二氧代哌啶-3-基)氨基)-2-氟苯基)-4-羟基哌啶-4-基)乙酸 盐酸盐24a(24mg,0.058mmol,采用专利申请“WO2021127561A1中说明书第633页实施例83”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(16mg,0.042mmol),冰浴下,缓慢加入N,N-二异丙基乙胺(16mg,0.12mmol),反应20分钟。反应液过滤,经高效液相制备色谱法(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%-70%,流速:30mL/min)纯化,得到标题化合物24(外消旋体混合物,8mg,产率:18%)。
MS m/z(ESI):1037.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.77(s,1H),8.18(s,1H),7.98(s,1H),7.77(d,2H),7.50(d,1H),7.14(d,1H),6.91-6.79(m,2H),6.74(s,1H),6.49(dd,1H),6.41(d,1H),5.77(d,1H),4.83(s,1H),4.50-4.40(m,2H),4.34-4.21(m,3H),3.86(s,3H),3.67-3.55(m,4H),3.10(s,3H),3.03(s,3H),2.94-2.81(m,8H),2.77-2.68(m,1H),2.61-2.53(m,3H),2.13-2.04(m,1H),1.92-1.82(m,3H),1.80-1.61(m,6H)。
实施例25
(±)-N-(35-溴-14-(9-((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)-3,9-二氮杂螺[5.5]壬-3-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺25
采用实施例23的合成路线,将第一步原料2,7-二氮杂螺[3.5]壬烷-2-羧酸叔丁酯替换为3,9-二氮杂螺[5.5]壬烷-3-羧酸叔丁酯(上海毕得医药),经高效液相制备色谱法(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)纯化,得到标题化合物25(外消旋体混合物,9mg,产率:20%)。
MS m/z(ESI):1097.2[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.07(s,1H),8.16(s,1H),7.95(s,1H),7.77(s,1H),7.68(s,1H),7.65(d,1H),7.51(d,1H),7.30(d,1H),7.22(dd,1H),7.13(d,1H),6.82(dd,1H),6.71(s,1H),5.06(dd,1H),4.41-4.35(m,2H),4.29(t,2H),4.03(d,2H),3.84(s,3H),3.09(s,3H),3.02(s,3H),2.96(t,2H),2.89-2.82(m,4H),2.65-2.53(m,3H),2.38-2.27(m,4H),2.18-2.10(m,2H),2.04-1.96(m,2H),1.88-1.75(m,5H),1.74-1.66(m,2H),1.55-1.42(m,7H),1.18-1.08(m,2H)。
实施例26
(±)-N-(35-溴-14-(4-(4-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)氮杂环丁-3-基)甲基)哌嗪-1-基)哌啶-1-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺26
第一步
3-((4-(1-(35-溴-16-甲氧基-52-(N-甲基甲基磺酰胺基)-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-14-基)哌啶-4-基)哌嗪-1-基)甲基)氮杂环丁烷-1-羧酸叔丁酯26a
向25mL单口瓶中加入化合物15g(52.5mg,0.063mmol)、3-甲酰基氮杂环丁烷-1-羧酸叔丁酯(24mg,0.13mmol,上海毕得医药),加入15mL甲醇和二氯甲烷的混合溶剂(V/V=2/1),搅拌均匀后加入无水乙酸钠(52mg,0.63mmol)和乙酸(4mg,0.067mmol),反应30分钟。加入氰基硼氢化钠(8mg,0.13mmol),继续反 应2小时。减压除去溶剂,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物26a(58mg,产率:99%)。
MS m/z(ESI):928.2[M+1]。
第二步
N-(14-(4-(4-(氮杂环丁-3-基甲基)哌嗪-1-基)哌啶-1-基)-35-溴-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺26b
将化合物26a(58mg,0.062mmol)溶于2mL二氯甲烷,加入1mL三氟乙酸,反应16小时。反应液减压浓缩,真空干燥,所得残留物经高效液相制备色谱法(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)纯化,得到标题化合物26b(30mg,产率:58%)
MS m/z(ESI):828.2[M+1]。
第三步
(±)-N-(35-溴-14-(4-(4-((1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)氮杂环丁-3-基)甲基)哌嗪-1-基)哌啶-1-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺26
向25mL单口瓶中加入化合物26b(30mg,0.036mmol)和化合物21h(20mg,0.072mmol),加入4mL N-甲基吡咯烷酮,搅拌均匀后加入N,N-二异丙基乙胺(24mg,0.19mmol),微波140℃反应4小时。反应液经高效液相制备色谱法(Waters2545,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)纯化,得到标题化合物26(外消旋体混合物,12mg,产率:31%)。
MS m/z(ESI):1084.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),8.16(s,1H),7.96(s,1H),7.80-7.77(m,1H),7.70(s,1H),7.64(d,1H),7.53-7.48(m,1H),7.13(d,1H),6.82(dd,1H),6.77(d,1H),6.67-6.62(m,2H),5.05(dd,1H),4.41(t,2H),4.29(t,2H),4.13(t,2H),3.84(s,3H),3.71-3.65(m,2H),3.30-3.27(m,4H),3.18-3.11(m,2H),3.10(s,3H),3.03(s,3H),3.01-2.96(m,1H),2.92-2.82(m,2H),2.73-2.53(m,9H),2.04-1.96(m,2H),1.92-1.78(m,4H),1.74-1.67(m,2H),1.54-1.42(m,2H)。
实施例27
N-(35-溴-14-(4-(4-(((3S)-1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)吡咯烷-3-基)甲基)哌嗪-1-基)哌啶-1-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺27
采用实施例15的合成路线,将第八步原料化合物15h替换为(3R)-1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)吡咯烷-3-甲醛(采用专利申请“WO2022253250A1中说明书第60页的实施例27”公开的方法制备而得),经高效液相制备色谱法(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)纯化,得到标题化合物27(一对非对映异构体混合物,1:1,40mg,产率:48%)。
MS m/z(ESI):1098.4[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.06(s,1H),8.16(s,1H),7.95(s,1H),7.78(s,1H),7.70(s,1H),7.64(d,1H),7.51(d,1H),7.13(d,1H),6.89(d,1H),6.84-6.78(m,2H),6.65(s,1H),5.05(dd,1H),4.44-4.36(m,2H),4.29(t,2H),3.84(s,3H),3.57-3.45(m,2H),3.43-3.36(m,6H),3.18-3.11(m,3H),3.10(s,3H),3.02(s,3H),2.93-2.83(m,1H),2.68(t,2H),2.62-2.53(m,4H),2.33(d,2H),2.29-2.22(m,1H),2.16-2.07(m,1H),2.04-1.95(m,2H),1.90-1.79(m,4H),1.77-1.67(m,3H),1.52-1.42(m,3H)。
实施例28
N-(35-溴-14-(4-(4-(((3R)-1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)吡咯烷-3-基)甲基)哌嗪-1-基)哌啶-1-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺28
采用实施例15的合成路线,将第八步原料化合物15h替换为(3S)-1-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-5-基)吡咯烷-3-甲醛(采用专利申请“WO2022253250A1中说明书第59页的实施例26”公开的方法制备而得),经高效液相制备色谱法(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)纯化,得到标题化合物28(一对非对映异构体混合物,1:1,35mg,产率:38%)。
MS m/z(ESI):1098.4[M+1]。
1H NMR(500MHz,DMSO-d6):δ11.06(s,1H),8.16(d,1H),7.95(s,1H),7.79(s, 1H),7.70(s,1H),7.64(d,1H),7.51(d,1H),7.13(d,1H),6.89(s,1H),6.83-6.79(m,2H),6.65(s,1H),5.05(dd,1H),4.45-4.36(m,2H),4.29(t,2H),3.84(s,3H),3.58-3.38(m,8H),3.18-3.11(m,3H),3.09(s,3H),3.02(s,3H),2.93-2.83(m,1H),2.72-2.57(m,6H),2.37-2.30(m,2H),2.29-2.22(m,1H),2.16-2.09(m,1H),2.04-1.96(m,2H),1.90-1.79(m,4H),1.74-1.67(m,3H),1.52-1.40(m,3H)。
实施例29
(±)-N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺29
采用实施例21的合成路线,将第八步原料化合物3,9-二氮杂螺[5.5]十一烷-3-羧酸叔丁酯替换为4-(哌啶-4-基)哌嗪-1-羧酸叔丁酯,经高效液相制备色谱法(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)纯化,得到标题化合物29(外消旋体混合物,14mg,产率:29%)。
MS m/z(ESI):1112.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),8.16(s,1H),7.95(s,1H),7.77(s,1H),7.69(s,1H),7.65(d,1H),7.51(d,1H),7.32(d,1H),7.24(dd,1H),7.13(d,1H),6.82(dd,1H),6.65(s,1H),5.06(dd,1H),4.43-4.35(m,2H),4.28(t,2H),4.05(d,2H),3.84(s,3H),3.31-3.27(m,4H),3.09(s,3H),3.08-3.06(m,1H),3.02(s,3H),2.96(t,2H),2.92-2.83(m,1H),2.70-2.25(m,9H),2.18-2.10(m,2H),2.04-1.95(m,2H),1.88-1.80(m,4H),1.77-1.66(m,4H),1.62-1.52(m,1H),1.49-1.39(m,2H),1.20-1.10(m,2H)。
实施例30
(±)-N-(35-溴-14-(4-(4-((1-(2-(2,6-二氧代哌啶-3-基)-1-氧代异吲哚啉-5-基)哌啶-4-基)甲基)哌嗪-1-基)哌啶-1-基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺30
采用实施例15的合成路线,将第八步原料化合物15h替换为(±)-1-(2-(2,6-二氧代哌啶-3-基)-1-氧代异吲哚啉-5-基)哌啶-4-甲醛(采用专利申请“WO2022198112A1中说明书第437页化合物67”公开的方法制备而得),经高效液相制备色谱法(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)纯化,得到标题化合物30(外消旋体混合物,8mg,产率:19%)。
MS m/z(ESI):1098.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.94(s,1H),8.16(s,1H),7.95(s,1H),7.78(s,1H),7.70(s,1H),7.53-7.48(m,2H),7.13(d,1H),7.06-7.01(m,2H),6.82(dd,1H),6.66(s,1H),5.04(dd,1H),4.43-4.37(m,2H),4.34-4.26(m,3H),4.19(d,1H),3.90-3.81(m,4H),3.17-3.12(m,2H),3.10(s,3H),3.03(s,3H),2.94-2.85(m,1H),2.81(t,2H),2.71-2.21(m,11H),2.13(d,2H),2.03-1.93(m,3H),1.89-1.67(m,9H),1.52-1.42(m,3H),1.19-1.12(m,2H)。
实施例31
(±)-5-(4-((4-(1-(35-溴-52-(二甲基磷酰基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-14-基)哌啶-4-基)哌嗪-1-基)甲基)哌啶-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮31

第一步
(2-氨基-5-甲氧基苯基)二甲基氧化膦31b
将2-碘-4-甲氧基苯胺31a(3.5g,14.05mmol,韶远科技(上海)有限公司)和二甲基氧化膦(1.7g,21.78mmol,韶远科技(上海)有限公司)溶于20mL N,N-二甲基甲酰胺中,加入醋酸钯(158mg,0.70mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(813mg,1.41mmol)和磷酸钾(4.5g,21.20mmol)。氮气置换三次,升温到120℃反应36小时。待反应液冷却到室温,用硅藻土过滤反应液,滤液减压浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物31b(1.58g,产率:56%)。
MS m/z(ESI):200.0[M+1]。
第二步
(2-((5-溴-2-氯嘧啶-4-基)氨基)-5-甲氧基苯基)二甲基氧化膦31c
将化合物31b(1.58g,7.93mmol)和5-溴-2,4-二氯嘧啶(1.8g,7.90mmol)溶于42mL N,N-二异丙基乙胺和异丙醇的混合溶剂(V/V=1/20)中,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,向残余物加入饱和碳酸氢钠溶液(20mL),用乙酸乙酯萃取(30mL×3),合并有机相,无水硫酸钠干燥,减压除去溶剂, 所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物31c(2.0g,产率:65%)。
MS m/z(ESI):390.0[M+1]。
第三步
(2-((5-溴-2-氯嘧啶-4-基)氨基)-5-羟基苯基)二甲基氧化膦31d
将化合物31c(1.0g,2.56mmol)溶于50mL二氯甲烷,冰浴冷却到0℃,氮气保护下滴加1M三溴化硼的二氯甲烷溶液(2.57g,10.26mmol),自然升温至室温反应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(20mL×2)和乙酸乙酯(20mL×2)萃取。合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物31d(880mg,产率:91%)。
MS m/z(ESI):375.9[M+1]。
第四步
4-(1-(4-((5-溴-4-((2-(二甲基磷酰基)-4-羟基苯基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-(甲氧基羰基)苯基)哌啶-4-基)哌嗪-1-羧酸叔丁酯31e
称取化合物15c(300mg,0.56mmol)、化合物31d(210mg,0.56mmol)到100mL单口瓶中,加入异丙醇(40mL)和三氟乙酸(255mg,2.24mmol),升温到90℃反应12小时,然后升温到100℃反应24小时。待反应液冷却到室温,加入N,N-二异丙基乙胺中和三氟乙酸,减压浓缩除去溶剂,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物31e(200mg,产率:45%)。
MS m/z(ESI):788.1[M+1]。
第五步
5-((5-溴-4-((2-(二甲基磷酰基)-4-羟基苯基)氨基)嘧啶-2-基)氨基)-2-(4-(4-(叔丁氧基羰基)哌嗪-1-基)哌啶-1-基)-4-甲氧基苯甲酸31f
称取化合物31e(330mg,0.42mmol)到50mL单口瓶中,加入12mL甲醇、四氢呋喃和水的混合溶剂(V/V/V=1/1/1)。然后加入氢氧化钠(84mg,2.10mmol),升温到60℃反应12小时。待反应液冷却到室温,加入1M盐酸溶液调节反应液pH到6-7之间,浓缩反应液,所得残留物经高效液相制备色谱法(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%-55%,流速:30mL/min)纯化,得到标题化合物31f(40mg,产率:12%)。
MS m/z(ESI):774.1[M+1]。
第六步
4-(1-(35-溴-52-(二甲基磷酰基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-14-基)哌啶-4-基)哌嗪-1-羧酸叔丁酯31g
将化合物31f(20mg,0.026mmol)溶于1.5mL N,N-二甲基甲酰胺,加入1,4-二溴丁烷(9mg,0.042mmol)、无水碳酸钾(11mg,0.080mmol)和四丁基碘化铵(5 mg,0.014mmol),氮气氛下,反应16小时。加水(5mL),用乙酸乙酯萃取(10mL×3),合并有机相,用饱和氯化钠溶液(15mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩,所得残留物用硅胶柱色谱法以洗脱剂体系A纯化得到标题化合物31g(11.5mg,产率:54%)。
MS m/z(ESI):828.3[M+1]。
第七步
35-溴-52-(二甲基磷酰基)-16-甲氧基-14-(4-(哌嗪-1-基)哌啶-1-基)-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-12-酮二盐酸盐31h
将化合物31g(20mg,0.024mmol)溶于1.5mL二氯甲烷,加入4M氯化氢的1,4-二氧六环溶液(1.5mL),反应2小时。反应液减压浓缩,真空干燥,得到粗产品标题化合物31h(18mg,产率:93%),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
(±)-5-(4-((4-(1-(35-溴-52-(二甲基磷酰基)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-14-基)哌啶-4-基)哌嗪-1-基)甲基)哌啶-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮31
向25mL单口瓶中加入化合物31h(18mg,0.022mmol)、化合物15h(14mg,0.038mmol),加入甲醇和二氯甲烷的混合溶剂(4mL,V/V=1/3),搅拌均匀后加入无水乙酸钠(41mg,0.50mmol)和乙酸(5mg,0.083mmol),反应1小时。加入氰基硼氢化钠(3mg,0.050mmol),继续反应1.5小时。反应液经高效液相制备色谱法(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)纯化,得到标题化合物31(外消旋体混合物,3mg,产率:11%)。
MS m/z(ESI):1081.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),10.12(s,1H),8.15(s,1H),7.90(s,1H),7.75(dd,1H),7.68(s,1H),7.65(d,1H),7.30(d,1H),7.24-7.18(m,1H),7.08(dd,1H),6.86(dd,1H),6.65(s,1H),5.06(dd,1H),4.44-4.38(m,2H),4.26(t,2H),4.03(d,2H),3.83(s,3H),3.33-3.28(m,4H),3.19(d,2H),3.00-2.83(m,3H),2.70(t,2H),2.64-2.22(m,4H),2.12(d,2H),2.05-1.95(m,5H),1.87-1.74(m,6H),1.69(s,3H),1.66(s,3H),1.55-1.40(m,4H),1.17-1.08(m,2H)。
实施例32
(±)-N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺32
采用实施例15的合成路线,将第八步原料化合物15h替换为(±)-2-(4-(2,6-二氧代哌啶-3-基)-3,5-二氟苯基)乙醛(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),经高效液相制备色谱法(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)纯化,得到标题化合物32(外消旋体混合物,9mg,产率:24%)。
MS m/z(ESI):1010.6[M+1]。
1H NMR(500MHz,DMSO-d6):δ10.95(s,1H),8.17(s,1H),7.96(s,1H),7.78(s,1H),7.71(s,1H),7.50(d,1H),7.13(d,1H),7.04(d,2H),6.83(dd,1H),6.67(s,1H),4.45-4.37(m,2H),4.30(t,2H),4.20(dd,1H),3.85(s,3H),3.30-3.27(m,4H),3.20-3.12(m,2H),3.09(s,3H),3.03(s,3H),2.85-2.74(m,3H),2.72-2.35(m,10H),2.17-2.07(m,1H),2.04-1.95(m,2H),1.91-1.81(m,3H),1.74-1.67(m,2H),1.58-1.41(m,2H)。
实施例33
(±)-N-(35-溴-14-(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)-16-甲氧基-12-氧代-6,11-二氧杂-2,4-二氮杂-3(2,4)-嘧啶杂-1(1,3),5(1,4)-二苯杂环十二蕃-52-基)-N-甲基甲磺酰胺33
采用实施例21的合成路线,将第十步原料化合物21j替换为9h,经高效液相制备色谱法(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%-90%,流速:30mL/min)纯化,得到标题化合物33(外消旋体混合物,14mg,产率:22%)。
MS m/z(ESI):1014.3[M+1]。
1H NMR(500MHz,DMSO-d6):δ10.85(s,1H),8.16(s,1H),7.95(s,1H),7.78(s,1H),7.70(s,1H),7.50(t,2H),7.13(d,1H),6.92(d,1H),6.83(d,2H),6.68(s,1H),4.43-4.36(m,2H),4.32-4.23(m,3H),3.89(s,3H),3.85(s,3H),3.25-3.19(m,4H),3.15-3.12(m,1H),3.10(s,3H),3.03(s,3H),2.70(t,2H),2.65-2.58(m,2H),2.57-2.52(m,4H),2.34-2.21(m,3H),2.19-2.12(m,1H),2.03-1.95(m,1H),1.90-1.61(m,7H), 1.22-1.15(m,2H)。
生物学评价
测试例1、本公开化合物对H1975/EGFR L858R-T790M-C797S、H1975/EGFR L858R-T790M、HCC827/EGFR Del19、BaF3/EGFR L858R-T790M-C797S、BaF3/EGFR L858R-C797S、BaF3/EGFR L858R、BaF3/EGFR Del19-T790M-C797S、BaF3/EGFR Del19-C797S、BaF3/EGFR Del19-T790M、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;A431/EGFR WT(CRL-1555)、HCC827/EGFR Del19(CRL-2868);H1975/EGFR L858R-T790M(CRL-5908)购于ATCC;BaF3/EGFR L858R-T790M-C797S(KC-0122)购于康源博创;BaF3/EGFR L858R-C797S(CBP73047)、BaF3/EGFR L858R(CBP73040)、BaF3/EGFR Del19-T790M-C797S(CBP73173)、BaF3/EGFR Del19-T790M(CBP73046)、BaF3/EGFR Del19-C797S(CBP73172)购于南京科佰。
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)
T75Flask(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)
1.2mL 96孔深孔板(透明,无菌,方形孔,V形底)(TITAN,02089063)
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1640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、根据细胞计数结果,用含10%FBS的DMEM(A431/EGFR WT)或含10%FBS的1640培养基(H1975/EGFR L858R-T790M-C797S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(A431/EGFR WT)、600(HCC827/EGFR Del19)、300(H1975/EGFR L858R-T790M-C797S、H1975/EGFR L858R-T790M)个细胞的密度将细胞接种于384孔培养板中,每孔20μL。
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、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(BaF3/EGFR L858R-T790M-C797S、BaF3/EGFR L858R-C797S、BaF3/EGFR L858R、BaF3/EGFR Del19-T790M-C797S、BaF3/EGFR Del19-T790M、BaF3/EGFR Del19-C797S)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(A431/EGFR WT)或3.3mM(H1975/EGFR L858R-T790M-C797S、H1975/EGFR L858R-T790M、HCC827/EGFR Del19、BaF3/EGFR L858R-T790M-C797S、BaF3/EGFR L858R-C797S、BaF3/EGFR L858R、BaF3/EGFR Del19-T790M-C797S、BaF3/EGFR Del19-T790M、BaF3/EGFR Del19-C797S),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。
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值和最大抑制率计算
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所示。
表1、本公开化合物对H1975/LTC、H1975/LT和HCC827/D突变细胞的增殖抑制活性

表2、本公开化合物对BaF3/EGFR突变细胞和A431WT细胞的增殖抑制活性

结论:本公开化合物对H1975/EGFR LTC和LT突变细胞、HCC827/EGFR D突变细胞和BaF3/EGFR多种突变细胞具有明显的选择性增殖抑制活性。
EGFR基因突变简称:
“L858R_T790M_C797S”也即LTC,“Del19_T790M_C797S”也即DTC,“L858R_C797S”也即LC,“Del19_C797S”也即DC,“Del19_T790M”也即DT,“L858R”也即L,“Del19”也即D。
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、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10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+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。
2.5、DC50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,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R。
c.降解率(%)=100-100×(R化合物-R空白对照)/(R溶媒对照-R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值如下表3所示。
表3、本公开化合物对BaF3/EGFR LTC总EGFR蛋白降解DC50

结论:本公开化合物对BaF3/EGFR LTC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.)/静脉注射(i.v.)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1、7、15、16和18。
2.2、试验动物
C57小鼠90只,雌性,平均分成10组,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)2021-0011。分别静脉注射和灌胃给药。
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。
静脉注射给药组:给药剂量为1.0mg/kg,给药体积为10mL/kg。
3、操作
灌胃给药组: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℃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。
静脉注射给药组:于给药前及给药后5分钟,0.25、0.5、1.0、2.0、4.0、8.0、11.0、24小时采血,处理同灌胃给药组。
测定不同浓度的药物给药后,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50μL,25μL内标溶液(甲苯磺丁脲(100ng/mL)或喜树碱(1ug/ml)和450μL乙腈),涡旋混合,并在3700rpm下离心10分钟。取上清液2μL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 (25)

  1. 一种通式(I)所示的化合物,或其可药用的盐:
    其中:
    R1选自氢原子、卤素、烷基、烷氧基、羟基、烯基、炔基、氰基、-NR21R22、-亚烷基NR21R22、-C(O)NR21R22、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、亚烷基、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;
    R2选自-P(O)R23R24、-SO2R23、-SO2N(R23R24)、-N(R23)SO2R24、卤素、烷基、烷氧基和氨基;
    或,R1、R2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;
    R3选自氢原子、卤素、烷基、烷氧基、羟基、烯基、炔基、氰基、-NR31R32、-亚烷基NR31R32、-C(O)NR31R32、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的被一个或多个R01所取代;
    R4选自氢原子、卤素、烷基、烯基、烷氧基和卤代烷基;
    或,R3、R4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;
    R21、R22、R23、R24、R31和R32相同或不同,且各自独立地选自氢原子、烷基、烷氧基、氨基、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、氨基、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的被一个或多个R01所取代;
    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、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、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、烷氧基、 羟基、烯基、炔基、氰基、-C(O)Rc、-NRaRb、-C(O)NRaRb、-亚烷基Rc、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、亚烷基、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R02所取代;
    或,RZ1、RZ2及与其连接的碳原子一起形成任选被一个或多个R02所取代的环烷基、杂环基、芳基或杂芳基;
    Ra和Rb相同或不同,且各自独立地选自氢原子、卤素、烷基、羟基、烯基、炔基、氨基、环烷基、杂环基、杂环基烷基、芳基和杂芳基;所述的烷基、烯基、炔基、环烷基、杂环基、杂环基烷基、芳基和杂芳基各自独立地任选被一个或多个R02所取代;
    Rc选自氢原子、卤素、烷基、烷氧基、羟基、烯基、炔基、氰基、氨基、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、氨基、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、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基和杂芳基;
    L1选自O、S、NRL、C(O)、C(O)NRL、NRLC(O)、NRLC(O)NRL和(CRL1RL2)m
    L2、L3、L4、L5和L6相同或不同,且各自独立地选自键、O、S、NRL、C(O)、C(O)NRL、NRLC(O)、NRLC(O)NRL和(CRL3RL4)n
    RL1、RL2、RL3和RL4各自独立地选自氢原子、卤素、烷基、烷氧基和卤代烷基;或,RL3、RL4及与其相连的同一碳原子一起形成环烷基或杂环基;
    RL选自氢原子、烷基和卤代烷基;
    J1选自环烷基、杂环基、杂环基-杂环基、芳基、杂芳基、O、C(O)、S(O)r、(CRJ1RJ2)p和N(RJ);所述的环烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R03所取代;
    J2、J3、J4和J5相同或不同,且各自独立地选自S、O(CRJ3RJ4)q、(CRJ3RJ4)qO、C(O)(CRJ3RJ4)q、(CRJ3RJ4)qC(O)、C(O)N(RJ)、N(RJ)C(O)、N(RJ)C(O)N(RJ)、S(O)r、S(O)rN(RJ)、N(RJ)S(O)r、N(RJ)S(O)rN(RJ)、(CRJ3RJ4)q、N(RJ)(CRJ3RJ4)q、(CRJ3RJ4)qN(RJ)、亚烯基、亚炔基、环烷基、杂环基、亚烷基-杂环基、杂环基-亚烷基、芳基和杂芳基;所述的亚烯基、亚炔基、环烷基、亚烷基、杂环基、芳基和杂芳基各自独立地任选被一个或多个R03所取代;
    J6选自O、S、S(O)r、(CRJ5RJ6)t、(CRJ5RJ6)tN(RJ)、N(RJ)(CRJ5RJ6)t、C(O)(CRJ5RJ6)t、(CRJ5RJ6)tC(O)、C(O)N(RJ)、N(RJ)C(O)、N(RJ)C(O)N(RJ)、亚烯基、亚炔基、环烷基和杂环基;所述的亚烯基、亚炔基、环烷基和杂环基各自独立地任选被一个或多个R03所取代;
    RJ选自氢原子、烷基和卤代烷基;
    RJ1、RJ2、RJ3、RJ4、RJ5和RJ6各自独立地选自氢原子、卤素、烷基、烷氧基、羟基、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、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R03所取代;
    或,RJ1、RJ2及与其相连的同一碳原子一起形成任选被一个或多个R03所取代的环烷基或杂环基;或,RJ3、RJ4及与其相连的同一碳原子一起形成任选被一个或多个R03所取代的环烷基或杂环基;或,RJ5、RJ6及与其相连的同一碳原子一起形成任选被一个或多个R03所取代的环烷基或杂环基;
    R03选自氧代基、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基和杂芳基;
    环A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,其它各自独立地为N或CRQ
    RQ相同或不同,且各自独立地选自氢原子、卤素、烷基、烷氧基、羟基、烯基、炔基、氰基和-(CH2)t1NRdRe;所述的烷基、烷氧基、烯基和炔基各自独立地任选被一个或多个R04所取代;
    Rd和Re各自独立地选自氢原子、卤素、烷基、环烷基和杂环基;
    Z4为C(RZ)2或C(O);RZ相同或不同,且各自独立地选自氢原子、卤素、烷基和氨基,或两个RZ与其相连的碳原子一起形成环烷基或杂环基;
    Z8为N或CRZ8
    RZ4和RZ7各自独立地选自氢原子、烷基和卤代烷基;
    RZ8选自氢原子、烷基、烷氧基和卤代烷基;
    R04选自氧代基、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基和杂芳基;
    m、n、p、q、t和t1各自独立地为0、1、2、3、4、5或6;
    r为1或2。
  2. 根据权利要求1所述的通式(I)所示的化合物,或其可药用的盐,其中:
    n2为1、2、3、4、5、6、7、8、9或10;n5为1、2、3、4、5、6或7;
    RL3和RL4各自独立地选自氢原子、C1-6烷基和C1-6烷氧基;
    R1、R2、R3、R4、RZ1和RZ2如权利要求1中所定义。
  3. 根据权利要求1或2所述的通式(I)所示的化合物,或其可药用的盐,其中:环A选自:
    Z8为CH或氮原子;Z4为CH2或C(O);RZ7为氢原子或C1-6烷基;RZ4为C1-6烷基;RQ为卤素或C1-6烷基;k为0、1、2、3或4;j为0、1、2或3;s为0、1、2或3;
    优选地,环A选自:
  4. 根据权利要求1所述的通式(I)所示的化合物,或其可药用的盐,其为通式(II-1)、(II-2)或(II-3)所示的化合物,或其可药用的盐:

    其中,
    n2为1、2、3、4、5、6、7、8、9或10;
    环J1’为5至14元含氮杂环基;
    环J3’为5至14元含氮杂环基,所述的5至14元含氮杂环基任选被1个或多个选自卤素、羟基、C1-6烷基和C1-6卤代烷基的取代基所取代;
    RJ为氢原子或C1-6烷基;s为0、1或2;
    R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、J1、q、J3、J4、J5、J6、Z4、RZ4、Q1、Q2、Q3、Q4、Q5和RQ如权利要求1所定义。
  5. 根据权利要求1所述的通式(I)所示的化合物,或其可药用的盐,其为通式(I’-1)或(I’-2)所示的化合物,或其可药用的盐:

    其中,
    环J1’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;
    n5为1、2、3、4、5、6或7;
    j为0、1或2;k为0、1或2;
    R1至R4、RZ1、RZ2、RL3、RL4、L5、L6、J3至J6、RQ、RZ4、RZ7、Z8和R03如权利要求1中所定义。
  6. 根据权利要求1所述的通式(I)所示的化合物,或其可药用的盐,其为通式(III-1)所示的化合物,或其可药用的盐:
    其中,
    环C为含氮杂环基;
    n5为1、2、3、4、5、6或7;
    u为0、1、2、3、4、5或6;s为0、1、2或3;
    R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、L5、L6、J1、R03、q、J4、J5、J6、RQ和Z4如权利要求1中所定义。
  7. 根据权利要求1所述的通式(I)所示的化合物,或其可药用的盐,其为通式(III-2)所示的化合物,或其可药用的盐:
    其中,
    环B为含氮杂环基;
    n5为1、2、3、4、5、6或7;
    u为0、1、2、3、4、5或6;s为0、1、2或3;
    R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、L5、L6、J1、J2、q、R03、J4、J5、J6、RQ和Z4如权利要求1中所定义。
  8. 根据权利要求1至4或6、7中任一项所述的通式(I)所示的化合物,或其可药用的盐,其中Z4为C(O)。
  9. 根据权利要求1至3中任一项所述的通式(I)所示的化合物,或其可药用的盐,其中,-J1-J2-J3-J4-J5-J6-选自:
    自左向右连接;各个q相同或不同,且各自独立地为0、1、2、3、4、5或6;各个u相同或不同,且各自独立地为0、1、2、3、4、5或6;各个q1相同或不同,且各自独立地为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。
  10. 根据权利要求1至3中任一项所述的通式(I)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;和/或J2选自键、C(O)、CH2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;和/或J3选自CH2、CH2CH2、哌啶基和哌嗪基;和/或J4为键或C(O);和/或J5为键;和/或J6选自键、3至12元杂环基和C1-6亚烷基N(RJ),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取代基所取代;RJ为氢原子或C1-6烷基。
  11. 根据权利要求1至10中任一项所述的通式(I)所示的化合物,或其可药用的盐,其中,R1为氢原子;和/或R3为卤素或C2-6烯基;和/或R4为氢原子。
  12. 根据权利要求1至11中任一项所述的通式(I)所示的化合物,或其可药用的盐,其中,R2为-N(R23)SO2R24或者-P(O)R23R24;R23和R24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,R2为-N(R23)SO2R24,R23和R24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3和5至12中任一项所述的通式(I)所示的化合物,或其可药用的盐,其中,L5为CH2或O,L6为CH2或C(O);优选地,L5为O,L6为C(O)。
  14. 根据权利要求1至13中任一项所述的通式(I)所示的化合物,或其可药用的盐,其中,RZ1选自氢原子、卤素、C1-6烷氧基、C1-6烷基、C1-6卤代烷氧基和3至6元环烷基;优选地,RZ1为氢原子或卤素。
  15. 根据权利要求1至14中任一项所述的通式(I)所示的化合物,或其可药用的盐,其中,RZ2选自氢原子、C1-6烷氧基、C1-6烷基和C1-6卤代烷氧基;优选地,RZ2为氢原子或C1-6烷氧基。
  16. 根据权利要求1至15中任一项所述的通式(I)所示的化合物,或其可药用的盐,其中,
  17. 根据权利要求1至16中任一项所述的通式(I)所示的化合物,或其可药用的盐,其选自以下任一化合物:









  18. 一种通式(IMA)或(INA)所示的化合物或其盐:
    其中,
    环C为含氮杂环基;
    n5为1、2、3、4、5、6或7;
    q-1为0、1、2、3、4或5;u为0、1、2、3、4、5或6;
    R1、R2、R3、R4、RZ1、RZ2、RL3、RL4、L5、L6、J1、J2和R03如权利要求1中所定义。
  19. 化合物或其盐,其选自以下结构:


  20. 一种制备根据权利要求6所述的通式(III-1)所示的化合物或其可药用的盐的方法,该方法包括:
    通式(IMA)所示的化合物或其盐(优选为盐酸盐或三氟乙酸盐)与通式(III-1B)所示的化合物或其盐发生还原胺化反应得到通式(III-1)所示的化合物或其可药用的盐,
    其中,环C、R1至R4、RZ1、RZ2、RL3、RL4、n5、L5、L6、J1、R03、u、q、J4至J6、RQ、Z4和s如权利要求6中所定义。
  21. 一种制备根据权利要求7所述的通式(III-2)所示的化合物或其可药用的盐的方法,该方法包括:
    通式(INA)所示的化合物或其盐与通式(III-2B)所示的化合物或其盐(优选为盐酸盐)发生还原胺化反应得到通式(III-2)所示的化合物或其可药用的盐,
    其中,
    环B、R1至R4、RZ1、RZ2、RL3、RL4、n5、L5、L6、J1、J2、q、R03、u、J4至J6、RQ、Z4和s如权利要求7中所定义。
  22. 一种药物组合物,所述药物组合物含有根据权利要求1至17中任一项所述的通式(I)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。
  23. 根据权利要求1至17中任一项所述的通式(I)所示的化合物或其可药用的盐或根据权利要求22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。
  24. 根据权利要求1至17中任一项所述的通式(I)所示的化合物或其可药用的盐或根据权利要求22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。
  25. 根据权利要求1至17中任一项所述的通式(I)所示的化合物或其可药用的盐或根据权利要求22所述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、肝癌、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、甲状腺癌、胶质母细胞瘤、脑转移瘤、实体瘤、口咽癌、支气管肿瘤和皮肤癌;进一步优选 为肺癌;更优选为非小细胞肺癌。
PCT/CN2023/107927 2022-07-18 2023-07-18 抑制并诱导egfr降解的大环类化合物、其制备方法及其在医药上的应用 WO2024017251A1 (zh)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN202210841290 2022-07-18
CN202210841290.7 2022-07-18
CN202211664576.9 2022-12-23
CN202211664576 2022-12-23
CN202310265432.4 2023-03-17
CN202310265432 2023-03-17

Publications (1)

Publication Number Publication Date
WO2024017251A1 true WO2024017251A1 (zh) 2024-01-25

Family

ID=89617124

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/107927 WO2024017251A1 (zh) 2022-07-18 2023-07-18 抑制并诱导egfr降解的大环类化合物、其制备方法及其在医药上的应用

Country Status (2)

Country Link
TW (1) TW202404979A (zh)
WO (1) WO2024017251A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013126132A1 (en) * 2012-02-21 2013-08-29 Merck Patent Gmbh Cyclic diaminopyrimidine derivatives
WO2021163627A1 (en) * 2020-02-14 2021-08-19 Salk Institute For Biological Studies Macrocyclic ulk1/2 inhibitors
WO2021216440A1 (en) * 2020-04-20 2021-10-28 Tenova Pharmaceuticals Inc. Novel protein kinase inhibitors
CN114394974A (zh) * 2022-03-25 2022-04-26 中国药科大学 多取代三芳基大环化合物与应用
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 (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013126132A1 (en) * 2012-02-21 2013-08-29 Merck Patent Gmbh Cyclic diaminopyrimidine derivatives
WO2021163627A1 (en) * 2020-02-14 2021-08-19 Salk Institute For Biological Studies Macrocyclic ulk1/2 inhibitors
WO2021216440A1 (en) * 2020-04-20 2021-10-28 Tenova Pharmaceuticals Inc. Novel protein kinase inhibitors
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
CN114394974A (zh) * 2022-03-25 2022-04-26 中国药科大学 多取代三芳基大环化合物与应用

Also Published As

Publication number Publication date
TW202404979A (zh) 2024-02-01

Similar Documents

Publication Publication Date Title
WO2020073949A1 (zh) 含氮杂芳类衍生物调节剂、其制备方法和应用
WO2021218110A1 (zh) 一类苯并噻唑基联芳基类化合物、制备方法和用途
CN109219604B (zh) 四氢异喹啉雌激素受体调节剂及其用途
US20220119363A1 (en) 2-amino-n-phenyl-nicotinamides as nav1.8 inhibitors
WO2021000885A1 (zh) 喹唑啉酮类衍生物、其制备方法及其在医药上的应用
TW202014423A (zh) 吲哚類大環衍生物、其製備方法及其在醫藥上的應用
WO2022268051A1 (zh) 稠合四环类化合物、其制备方法及其在医药上的应用
JP2023539136A (ja) Tlr9阻害剤として有用な置換ヘテロアリール化合物
WO2022247816A1 (zh) 含氮杂环类化合物、其制备方法及其在医药上的应用
TW201917129A (zh) 含吡唑基的三并環類衍生物、其製備方法和應用
WO2022194269A1 (zh) 新型egfr降解剂
WO2022228543A1 (zh) 桥环类化合物、其制备方法及其在医药上的应用
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
JP2023538393A (ja) 線維症の処置のためのtlr9の阻害剤としてのイミダゾ[1,2-a]ピリジンおよび[1,2,4]トリアゾロ[1,5-a]ピリジン誘導体
WO2023093845A1 (zh) 一种用于雄激素受体蛋白靶向降解的嵌合体化合物、其制备方法及其在医药上的应用
WO2024017251A1 (zh) 抑制并诱导egfr降解的大环类化合物、其制备方法及其在医药上的应用
WO2023072297A1 (zh) 含氮的四环化合物、其制备方法及其在医药上的应用
WO2022268229A1 (zh) 蛋白抑制剂或降解剂、包含其的药物组合物及医药上的用途
KR20240019071A (ko) N2-페닐피리미딘-2,4-디아민 화합물, 및 그 제조 방법 및 사용 방법
WO2024094171A1 (zh) 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
WO2020114474A1 (zh) 一种杂环化合物、其应用及含其的药物组合物
TW202045493A (zh) 2-胺基嘧啶類衍生物、其製備方法及其在醫藥上的應用
CN117384165A (zh) 一种用于egfr蛋白靶向降解的嵌合体化合物、其制备方法及其在医药上的应用
CN118005655A (zh) 稠环类化合物、其制备方法及其在医药上的应用
WO2024022444A1 (zh) 稠环类化合物、其制备方法及其在医药上的应用

Legal Events

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

Ref document number: 23842314

Country of ref document: EP

Kind code of ref document: A1