WO2023072270A1 - Protac化合物、含其的药物组合物及其制备方法和应用 - Google Patents

Protac化合物、含其的药物组合物及其制备方法和应用 Download PDF

Info

Publication number
WO2023072270A1
WO2023072270A1 PCT/CN2022/128411 CN2022128411W WO2023072270A1 WO 2023072270 A1 WO2023072270 A1 WO 2023072270A1 CN 2022128411 W CN2022128411 W CN 2022128411W WO 2023072270 A1 WO2023072270 A1 WO 2023072270A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
membered
cycloalkyl
ring
amino
Prior art date
Application number
PCT/CN2022/128411
Other languages
English (en)
French (fr)
Inventor
吕志俭
白海云
高安慧
苏明波
钟利
Original Assignee
百极弘烨(南通)医药科技有限公司
百极弘烨(广东)医药科技有限公司
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 百极弘烨(南通)医药科技有限公司, 百极弘烨(广东)医药科技有限公司 filed Critical 百极弘烨(南通)医药科技有限公司
Publication of WO2023072270A1 publication Critical patent/WO2023072270A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the invention relates to a PROTAC compound, a pharmaceutical composition containing it, a preparation method and application thereof.
  • Bruton's tyrosine kinase is a member of the Tec family of non-receptor protein tyrosine kinases, and is a key regulator in the B cell antigen receptor (BCR) signaling pathway, distributed in lymphoid tissue, bone marrow and spleen.
  • BCR B cell antigen receptor
  • BTK X-linked agammaglobulinemia
  • CRBN a brain-associated protein with ionic protease activity, interacts with DNA damage-binding protein-1 (DDB1), arrestin 4 (Cullin4, CUL4A) and regulatory factor Cullins1 (ROC1) to form a functional E3 ubiquitin ligase complex CRBN-CRL4, which recognizes specific proteins through substrate receptors (SRs), prompts ubiquitin to specifically bind to substrate proteins, and marks proteins that need to be degraded, thereby initiating the degradation process.
  • DDB1 DNA damage-binding protein-1
  • Cullin4, CUL4A arrestin 4
  • ROC1 regulatory factor Cullins1
  • the target of miridamine antineoplastic drugs is CRBN, which regulates its downstream signaling by inhibiting the self-ubiquitination of CRBN and preventing CRBN from forming CRBN-CRL4 (an E3 ubiquitin ligase) together with DDB1, CUL4A and ROC1 Exercising the adjustment function.
  • PROTAC proteolysis targeting chimera
  • PROTAC proteolysis targeting chimera
  • BTK (C481S) mutation causes resistance to covalent BTK inhibitors such as ibrutinib (Woyach, et al.; Blood, 120(6):1175-1184, 2012.), and this degradation method can also effectively Targets BTK(C481S) mutation.
  • WO2020239103, CN110845500, US11028088, etc. have demonstrated PROTAC compounds with excellent properties.
  • the BTK-PROTAC molecule contains a structural group of amine compounds, it may degrade the substrate proteins Ikaros (IKZF1) and Aiolos (IKZF3) of amine compounds, causing various toxicities such as thrombocytopenia and neutropenia. side effect.
  • the purpose of the present invention is to provide a PROTAC compound with good efficacy, good bioavailability, safety, and ability to inhibit or degrade BTK protein, for treating BTK-related diseases such as tumors or autoimmune diseases, or inflammatory diseases.
  • BTK-related diseases such as tumors or autoimmune diseases, or inflammatory diseases.
  • the unique novel E3 ubiquitin ligase ligand in this class of PROTAC compounds can weaken the substrate degradation effect caused by immunomodulatory imide drug (IMiD), thereby reducing the activity of IMiD.
  • the first aspect of the present invention provides a compound represented by formula I, its pharmaceutically acceptable salt, its cis-trans isomers, its chiral isomers, its enantiomers, its diastereomers Isomers, their isotopic derivatives, their prodrugs, their solvates or their hydrates,
  • Z 1 is selected from: N or CR 1 ;
  • Z 2 is selected from: N or CR 1 ;
  • Z 3 is selected from: N or CR 7 ;
  • Z 4 is selected from: N or CR 7 ;
  • Z is selected from: N or CH;
  • W 1 is selected from: -O-, -S-, -NR b -, -CONR b -, -NR b CO-, -(CH 2 ) n NR b - or -(CH 2 ) n NR b CO-;
  • A is selected from: bond, C3-C20 cycloalkyl, 3-20 membered heterocyclic group, C6-C10 aryl, 5-15 membered heteroaryl, -(CH 2 ) p R c ; wherein, R c is selected From: C3-C20 cycloalkyl, 3-20 membered heterocyclic group, C6-C10 aryl group, 5-15 membered heteroaryl group, wherein, the heterocyclic group and heteroaryl group contain 1 to 4 members selected from Heteroatoms of O, S, and N; wherein, the C3-C20 cycloalkyl, 3-20 membered heterocyclyl, C6-C10 aryl, and 5-15 membered heteroaryl are optionally replaced by 1-4 R a replaces;
  • AL 1 , AL 2 and AL 3 are each independently selected from: bond, -O-, -S-, -NR' b -, -(CH 2 ) n' -, -CO-, -(CH 2 ) n' NR' b -, -CONR' b -, -NR' b CO-, -(CH 2 ) n' NR' b CO-;
  • a 2 and A 3 are each independently selected from: bond, C3-C20 cycloalkyl, 3-20 membered heterocyclyl, C6-C10 aryl, 5-15 membered heteroaryl, wherein said heterocyclyl ,
  • the heteroaryl group contains 1 to 4 heteroatoms selected from O, S, and N; wherein, the C3-C20 cycloalkyl, 3-20 membered heterocyclic group, C6-C10 aryl group, 5-15 membered heteroatom Aryl is optionally substituted by 1-4 R a ;
  • W is selected from: bond, -O-, -S-, -NR" b- , -( CH2 ) n" -, -CONR" b- , -NR" bCO- , -( CH2 ) n" NR b CO-;
  • R b , R' b and R" b are each independently selected from: H, C1-C6 alkyl, C1-C6 alkoxy, C3-C6 cycloalkyl, 4-6 membered heterocyclic group, C6-C10 aromatic Base, 5-10 membered heteroaryl;
  • R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are each independently selected from: H, halogen, cyano, hydroxyl, amino, carboxyl, amido, ester, C1-C6 alkyl, C1- C6 alkoxy, C3-C6 cycloalkyl, 4-6 membered heterocyclic group, C6-C10 aryl, 5-15 membered heteroaryl; wherein, the C1-C6 alkyl, C1-C6 alkoxy , C3-C6 cycloalkyl, 4-6 membered heterocyclyl, C6-C10 aryl, 5-15 membered heteroaryl are optionally substituted by 1-4 Ra;
  • R are each independently selected from: H, halogen, cyano, hydroxyl, amino, carboxyl, amido, ester, C1-C6 alkyl, C1-C6 alkoxy, C3-C6 cycloalkyl, 4-6 Membered heterocyclic group, C6-C10 aryl group, 5-15 membered heteroaryl group; wherein, the C1-C6 alkyl group, C1-C6 alkoxy group, C3-C6 cycloalkyl group, 4-6 membered heterocyclic group , C6-C10 aryl, 5-15 membered heteroaryl are optionally substituted by 1-4 Ra;
  • two adjacent R 7 and the ring atoms connected to it together form a C3-C20 cycloalkyl group, a 3-20 membered heterocyclic group, a C6-C10 aryl group, a 5-15 membered heteroaryl group, preferably C5 -C6 cycloalkyl, 5-6 membered heterocyclyl, phenyl, 5-6 membered heteroaryl; wherein, the cycloalkyl, heterocyclyl, aryl, phenyl, heteroaryl are optionally 1-4 Ra substitutions;
  • ring A 4 is ring A 4 , wherein any one of R 7 adjacent to W 2 , W 2 and the ring atoms connected to them form a ring, and ring A 4 is selected from: 8-16 membered bicyclic, tricyclic or polycyclic ring systems, preferably It is a 9-14 membered bicyclic or tricyclic ring system; the system is optionally substituted by 1-4 R a ; optionally, among the remaining R 7 , the adjacent two R 7 and the ring atom connected thereto Together form C3-C20 cycloalkyl, 3-20 membered heterocyclic group, C6-C10 aryl, 5-15 membered heteroaryl, preferably C5-C6 cycloalkyl, 5-6 membered heterocyclic group, benzene Base, 5-6 membered heteroaryl; wherein, the cycloalkyl, heterocyclyl, aryl, phenyl, heteroaryl are optionally substituted by 1-4 R;
  • ring A 5 is ring A 5 , wherein any one of R 7 adjacent to AL 3 , AL 3 and the ring atoms connected to them form a ring, and ring A 5 is selected from: 8-16 membered bicyclic, tricyclic or polycyclic ring systems, preferably It is a 9-14 membered bicyclic or tricyclic ring system; the system is optionally substituted by 1-4 R a ; optionally, among the remaining R 7 , the adjacent two R 7 and the ring atom connected thereto Together form C3-C20 cycloalkyl, 3-20 membered heterocyclic group, C6-C10 aryl, 5-15 membered heteroaryl, preferably C5-C6 cycloalkyl, 5-6 membered heterocyclic group, benzene Base, 5-6 membered heteroaryl; wherein, the cycloalkyl, heterocyclyl, aryl, phenyl, heteroaryl are optionally substituted by 1-4 R;
  • ring A 6 is ring A 6 , wherein any R 7 adjacent to W 2 , W 2 and the ring atoms connected to them form a ring, any R 7 adjacent to AL 3 , AL 3 and the ring atoms connected to them Forming a ring, ring A is selected from: 8-16 membered tricyclic or polycyclic ring systems, preferably 9-14 membered tricyclic systems; said system is optionally substituted by 1-4 R ; optionally, In the remaining R 7 , two adjacent R 7 and the ring atoms connected to it together form a C3-C20 cycloalkyl group, a 3-20 membered heterocyclic group, a C6-C10 aryl group, and a 5-15 membered heteroaryl group, preferably C5-C6 cycloalkyl, 5-6 membered heterocyclic group, phenyl, 5-6 membered heteroaryl; wherein, the cycloalkyl, heterocyclic group, aryl, phen
  • R is selected from: H, C1-C6 alkyl,
  • H in the above -CH 2 - is optionally substituted by R a ;
  • p, n, n' and n" are each independently selected from: 1, 2, 3, 4 or 5;
  • n 1 , n 2 , n 3 , n 4 , n 5 , n 6 and n 7 are each independently selected from: 0, 1, 2, 3, 4 or 5;
  • the limiting conditions are: at most one of Z 1 and Z 2 is N; at most one of A 1 , A 2 and A 3 is a key;
  • R 7 is one or more of H, halogen, cyano, hydroxyl, amino, carboxyl, C1-C4 alkyl, C1-C4 alkoxy and C3-C6 cycloalkyl
  • R 8 is H or C1- When C4 alkoxy group, A 1 is not 5-6 membered heterocyclic group and 5-6 membered heteroaryl group.
  • the compound has the structure shown in formula II-IV
  • n 13 is selected from: 0, 1, 2, 3 or 4;
  • W 1 , W 2 , A 1 , A 2 , A 3 , Ring A 4 , Ring A 5 , Ring A 6 , Z 1 , Z 2 , Z 8 , AL 1 , AL 2 , AL 3 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 8 , n 1 , n 2 , n 3 , n 4 , n 5 and n 6 are as defined above.
  • each R 13 is independently selected from: H, F, Cl, Br, I, OH, NH 2 , CN, OMe, Me, CH 2 OH, CO 2 H, CF 3 , CHF 2 or CONH 2 .
  • A is selected from: bond, C4-C7 cycloalkyl, 5-membered oxetane, 6-membered oxetane, 5-membered azacyclane, 6-membered azacyclane, 7 Azacycloalkyl, oxoazepanyl, phenyl, pyrazolyl, pyridyl, pyrazinyl, benzyl, C7-C15 spirocyclyl, C6-C15 acyclyl, C5-C15 Bridged ring group, C5-C15 heterospirocyclyl, C4-C15 heterocyclyl, C5-C15 heterobridged ring group; wherein, the C5-C15 heterospirocyclyl, C4-C15 heterospirocyclyl, C5 The -C15 heterobridged ring group contains 1 to 4 heteroatoms selected from O, S, N; preferably, A is selected from: 4-membere
  • A is selected from: wherein, the above-mentioned groups are optionally substituted by 1-4 R a , and the definition of R a is as above.
  • part K is selected from:
  • part K is not
  • both AL 1 and AL 2 are bonds.
  • AL 1 , AL 2 and AL 3 are all bonds.
  • each of the above groups is connected to part K.
  • both AL 1 and AL 2 are bonds, and the compound has the structure shown in formula II-1,
  • a 1 , A 2 , A 3 , R 3 , n 3 and partK are as defined above.
  • part K is selected from
  • both AL 1 and AL 2 are bonds, and the compound has the structure shown in formula II-2,
  • a 1 , A 2 , A 3 , R 7 , n 7 , R 3 and n 3 are as defined above.
  • a 1 is Preferably, the right end of each of the above groups is connected to A2 .
  • A2 is Preferably, the right end of each of the above groups is connected to A3 .
  • A3 is or key.
  • the right end of each of the above groups is connected to part K.
  • each of the above groups is connected to part K.
  • R 3-1 , R 3-2 , R 3-3 , R 3-4 and R 3-5 are each independently selected from H, halogen (fluorine, chlorine, bromine or iodine), hydroxyl, amino, C1- C6 alkyl (such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl), C1-C6 alkoxy (such as methoxy, ethoxy, n-propyl oxy, isopropoxy, n-butoxy, isobutoxy or tert-butoxy) or C1-C6 alkylamino (e.g. methylamino, ethylamino, n-propylamino, isopropylamino, n-butylamino, isobutylamino or tert-butylamino).
  • C1- C6 alkyl such as methyl, ethyl
  • R 3-1 is selected from halogen, such as fluorine, chlorine, bromine or iodine, preferably fluorine.
  • R 3-2 is selected from C1-C6 alkoxy, preferably methoxy.
  • R 3-3 is selected from H.
  • R 3-4 is selected from H.
  • R 3-5 is selected from H.
  • R 3-1 is selected from fluorine
  • R 3-2 is selected from methoxy
  • R 3-3 is selected from H
  • R 3-4 is selected from H
  • R 3-5 is selected from H.
  • the compound has the structure shown in formula V
  • A is selected from: bond, 3-20 membered cycloalkyl group, 3 membered heterocyclic group, 4 membered heterocyclic group, 7-20 membered heterocyclic group, C6-C10 aryl group, 7-15 membered heteroaryl group; wherein , the heterocyclic group and heteroaryl group contain 1 to 4 heteroatoms selected from O, S, N; wherein, the 3-20 membered cycloalkyl, 3 membered heterocyclic group, 4 membered heterocyclic group , 7-20 membered heterocyclyl, C6-C10 aryl, 7-15 membered heteroaryl are optionally substituted by 1-4 R a ;
  • W 1 , A 1 , A 2 , A 3 , Z 1 , Z 2 , AL 1 , AL 2 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , n 1 , n 2 , n 3 , n 4 , n 5 , n 6 and n 7 are as defined above;
  • A is selected from: bond, C4-C7 cycloalkyl, 7-membered azacycloalkyl, oxoazepanyl, benzyl, C7-C15 spirocyclyl, C6-C15 acyclyl , C5-C15 bridged ring group, C7-C15 heterospirocyclyl, C4 heterocyclyl, C7-C15 heterocyclyl, C7-C15 heterobridged ring group; wherein, the C7-C15 heterospirocyclyl , C4 heterocyclic group, C7-C15 heterocyclic group, C7-C15 heterocyclic ring group contains 1 to 4 heteroatoms selected from O, S, N; preferably, A is selected from: 4-membered aza Cycloalkyl, 7-membered azacycloalkyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, spir
  • A1 is selected from: Wherein, the above-mentioned groups are optionally substituted by 1-4 R a , and the definition of R a is as above.
  • each R 7 is independently selected from: -H, -F, -Cl, -Br, -I, -OH, -NH 2 , -CN, -OMe, -Me, -CH 2 OH , -CO 2 H, -CF 3 , -CHF 2 , -CONH 2 ; preferably, each R 7 is independently selected from: -H, -F, -OMe, -Me, -CF 3 or -CHF 2 .
  • ring A is selected from:
  • ring A is selected from:
  • ring A is selected from:
  • R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are each independently selected from: H, F, Cl, Br, I, OH, NH 2 , CN, OMe, Me , CH2OH , CO2H , CF3 , CHF2 or CONH2 .
  • Z1 is CH.
  • Z 2 is CH.
  • W 1 is selected from: -O-, -S-, -NH-, -CONH-, -NHCO- or -CH 2 NHCO-.
  • W 2 is selected from: -O-, -S-, -NH-, -CH 2 -, -CONH-, -NHCO-, -CH 2 NHCO- or a bond.
  • AL 1 , AL 2 and AL 3 are each independently selected from: -O-, -S-, -NH-, -CH 2 -, -CO-, -CH 2 NH-, -CH 2 N(CH 3 )-, -CH 2 CH 2 -, -CONH-, -NHCO-, -CH 2 NHCO- or a bond.
  • a 2 and A 3 are each independently selected from: bond, 3-12 membered heterocyclic group, C3-C12 cycloalkyl group, 5-15 membered heteroaryl group, 5-10 membered aryl group or Bond, wherein, described heterocyclyl, heteroaryl contain 1 to 4 heteroatoms selected from O, S, N; preferably, A 2 and A 3 are selected from: 4-membered azacycloalkyl, 5 Azacycloalkyl, 6-membered azacycloalkyl, 7-membered azacycloalkyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, tetrahydrofuranyl, tetrahydropyranyl, morpholinyl , piperidinyl, piperazinyl, pyrrolyl, spiro[3.3]heptanyl, 2-azaspiro[3.3]heptanyl, 2,6-diazaspir
  • A2 and A3 are each independently selected from: Wherein, the above-mentioned groups are optionally substituted by 1-4 R a , and the definition of R a is as above.
  • part L is selected from:
  • W 1 , W 2 , A 1 , A 2 , A 3 , Ring A 4 , Ring A 5 , Ring A 6 , Z 1 , Z 2 , Z 3 , Z 4 , Z 8 , Al 1 , AL 2 , AL 3 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , n 1 , n 2 , n 3 , n 4 , n 5 , n 6 and n 7 is the group corresponding to each specific compound in the examples.
  • the compound has the structure shown in formula VI-1 or VI-2,
  • R 7-1 , R 7-2 and R 7-3 are each independently selected from H, halogen (fluorine, chlorine, bromine or iodine), hydroxyl, amino, C1-C6 alkyl (such as methyl, ethyl, n- propyl, isopropyl, n-butyl, isobutyl or tert-butyl), C1-C6 alkoxy (e.g.
  • R 3-1 , R 3-2 , R 3-3 , R 3-4 and R 3-5 are each independently selected from H, halogen (fluorine, chlorine, bromine or iodine), hydroxyl, amino, C1-C6 alkane (e.g. methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl), C1-C6 alkoxy (e.g.
  • Z 1 , Z 2 , W 1 , R 1 , R 2 , R 4 , R 5 , R 6 , n 1 , n 2 , n 4 , n 5 , n 6 are as defined above;
  • R 7-1 , R 7-2 , R 7-3 , R 3-1 , R 3-2 , R 3-3 , R 3-4 and R 3-5 are not H at the same time.
  • the compound has the structure shown in formula VI-3 or VI-4,
  • R 7-1 , R 7-2 , R 7-3 , R 3-1 , R 3-2 , R 3-3 , R 3-4 and R 3-5 are as defined above;
  • R 7-1 , R 7-2 , R 7-3 , R 3-1 , R 3-2 , R 3-3 , R 3-4 and R 3-5 are not H at the same time.
  • R 7-1 is selected from halogen, such as fluorine, chlorine, bromine or iodine, preferably fluorine.
  • R 7-2 is selected from H.
  • R 7-3 is selected from H.
  • R 7-1 is selected from fluorine
  • R 7-2 is selected from H
  • R 7-3 is selected from H.
  • R 3-1 is selected from halogen, such as fluorine, chlorine, bromine or iodine, preferably fluorine.
  • R 3-2 is selected from C1-C6 alkoxy, preferably methoxy.
  • R 3-3 is selected from H.
  • R 3-4 is selected from H.
  • R 3-5 is selected from H.
  • R 3-1 is selected from fluorine
  • R 3-2 is selected from methoxy
  • R 3-3 is selected from H
  • R 3-4 is selected from H
  • R 3-5 is selected from H.
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from the compounds shown in the examples.
  • the second aspect of the present invention provides a pharmaceutical composition, which comprises the compound described in the first aspect, its pharmaceutically acceptable salt, its cis-trans isomers, its chiral isomers, its enantiomers Constructs, diastereoisomers, isotopic derivatives, prodrugs, solvates or hydrates thereof, and one or more pharmaceutically acceptable carriers or excipients.
  • the pharmaceutical composition further includes other therapeutic agents.
  • a preparation method of a pharmaceutical composition comprising the step of: mixing a pharmaceutically acceptable carrier or excipient with the compound of the present invention, its pharmaceutically acceptable salt, or its cis-trans isomerization isomers, enantiomers, diastereomers, isotopic derivatives, prodrugs, solvates or hydrates thereof to form a pharmaceutical composition.
  • the third aspect of the present invention provides a compound as described in the first aspect, its pharmaceutically acceptable salt, its cis-trans isomers, its chiral isomers, its enantiomers, its non- Use of the enantiomer, its isotope derivative, its prodrug, its solvate or its hydrate, or the pharmaceutical composition as described in the second aspect in the preparation of a medicament for treating BTK-mediated diseases.
  • the disease is selected from: autoimmune disease, inflammatory disease or tumor;
  • the autoimmune disease is preferably selected from: lupus, multiple sclerosis, amyotrophic lateral sclerosis, rheumatoid arthritis, psoriasis, complications caused by organ transplantation, diabetes, asthma, atopic dermatitis , autoimmune thyroid disease, ulcerative colitis, Crohn's disease, Alzheimer's disease, leukemia or lymphoma;
  • the inflammatory disease is preferably selected from the group consisting of keratitis, rhinitis, stomatitis, mumps, pharyngitis, tonsillitis, tracheitis, bronchitis, pneumonia, myocarditis, gastritis, gastroenteritis, cholecystitis or appendicitis;
  • the tumor is preferably selected from the group consisting of: small lymphocytic lymphoma, acute lymphoblastic leukemia, chronic lymphocytic leukemia, acute myeloid leukemia, chronic myelogenous leukemia, acute promyelocytic leukemia, chronic myeloid leukemia, diffuse large B cell Lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, Waldenstrom macroglobulinemia, follicular lymphoma, multiple myeloma, or mantle cell lymphoma.
  • small lymphocytic lymphoma acute lymphoblastic leukemia, chronic lymphocytic leukemia, acute myeloid leukemia, chronic myelogenous leukemia, acute promyelocytic leukemia, chronic myeloid leukemia, diffuse large B cell Lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, Waldenstrom macroglobulinemia, follicular lymph
  • the fourth aspect of the present invention provides a compound as described in the first aspect, its pharmaceutically acceptable salt, its cis-trans isomers, its chiral isomers, its enantiomers, its non- Use of an enantiomer, its isotopic derivative, its prodrug, its solvate or its hydrate, or the pharmaceutical composition as described in the second aspect in the preparation of a drug for inhibiting or regulating the activity of BTK protein kinase .
  • the BTK protein kinase is preferably non-mutated BTK protein kinase or mutated BTK protein kinase; the mutated BTK protein kinase is preferably C481S mutated BTK protein kinase.
  • the term "about” when used in reference to a specifically recited value means that the value may vary by no more than 1% from the recited value.
  • the expression “about 100” includes all values between 99 and 101 and in between (eg, 99.1, 99.2, 99.3, 99.4, etc.).
  • the term “comprises” or “includes (comprising)” can be open, semi-closed and closed. In other words, the term also includes “consisting essentially of”, or “consisting of”.
  • alkyl includes straight or branched chain alkyl groups.
  • C 1 -C 6 alkyl represents a straight-chain or branched alkyl group having 1-6 (eg 1, 2, 3, 4, 5 or 6) carbon atoms, such as methyl, ethyl, propyl , isopropyl, butyl, isobutyl, tert-butyl, etc.
  • cycloalkyl refers to a cyclic alkyl group containing a specific number of C atoms, such as "C3-C20 cycloalkyl” refers to a group having 3-20 (eg 3, 4, 5, 6, 7 , 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) carbon atoms. It is preferably a C3-C12 cycloalkyl group, more preferably a C3-C6 cycloalkyl group. Cycloalkyl groups can be monocyclic, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or similar groups.
  • cycloalkyl group can also be fused to an aryl group, a heteroaryl group, or a heterocyclyl group, wherein the ring connected to the parent structure is a cycloalkyl group, such as wait.
  • cycloalkyl is intended to include substituted cycloalkyl groups.
  • C1-C6 alkoxy refers to a straight or branched alkoxy group having 1-6 carbon atoms (eg 1, 2, 3, 4, 5 or 6); it has Formula C1-C6 alkyl-O- or -C1-C5 alkyl-O-C1-C5 alkyl (eg, -CH 2 -O-CH 2 CH 3 , -CH 2 -O-(CH 2 ) 2 CH 3. -CH 2 CH 2 -O-CH 2 CH 3 ) structure, preferably C1-C6 alkyl-O-, for example, methoxy, ethoxy, propoxy, isopropoxy, butoxy base, isobutoxy or tert-butoxy, etc.
  • C1-C6 alkyl-O- for example, methoxy, ethoxy, propoxy, isopropoxy, butoxy base, isobutoxy or tert-butoxy, etc.
  • heterocyclyl refers to a saturated or partially saturated cyclic group having N, S and O heteroatoms (including but not limited to monocyclic rings such as 3-7 members, 6-11 members Bicycle, or 8-16 membered tricyclic ring system),
  • 3-20 membered heterocyclic group means having 3-20 (such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) atoms and wherein 1-4 (eg 1, 2 or 3) atoms are heteroatoms selected from the group consisting of N, S and O or partially saturated cyclic groups.
  • it may be monocyclic or bicyclic, for example bridged or spiro.
  • the heterocyclic group is preferably a 4-12-membered heterocyclic group, more preferably a 4-8-membered or 4-6-membered heterocyclic group. Specific examples may be oxetane, azetidine, tetrahydro-2H-pyranyl, piperidyl, piperazinyl, tetrahydrofuranyl, morpholinyl, pyrrolidinyl and the like.
  • the heterocyclyl may be fused to a heteroaryl, aryl or cycloalkyl ring, wherein the ring bonded to the parent structure is a heterocyclyl such as wait.
  • aryl refers to an aromatic ring group that does not contain heteroatoms in the ring
  • C6-C10 aryl refers to an aromatic ring group that does not contain heteroatoms in the ring and has 6 to 10 (such as 6, 7, 8 , 9, 10) carbon atom aromatic ring group
  • the aryl group can be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring connected to the parent structure is an aryl ring.
  • phenyl ie six-membered aryl
  • naphthyl etc.
  • the six-membered aryl is also intended to include six-membered aryl and 5-6 membered cycloalkyl (such as ) and six-membered aryl and 5-6-membered heterocyclic group (such as wait).
  • the C6-C12 aryl group is preferably a C6-C10 aryl group.
  • Aryl groups can be optionally substituted or unsubstituted.
  • heteroaryl refers to a cyclic aromatic group having 1-4 (eg 1, 2 or 3) atoms are heteroatoms selected from the group consisting of N, S and O, "5-15 membered heteroatoms "Aryl” means having 5-15 (eg 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15) atoms and wherein 1-4 (eg 1, 2 or 3) atoms ) atom is a cyclic aromatic group selected from the heteroatoms of N, S and O in the following group.
  • the heteroaryl group is preferably 5-12 membered heteroaryl group, preferably 5-10 membered, more preferably 5-6 membered, which may be a single ring or a condensed ring.
  • heteroaryl ring may be fused to an aryl, heterocyclyl or cycloalkyl ring, wherein the ring bonded to the parent structure is a heteroaryl ring.
  • Heteroaryl groups can be optionally substituted or unsubstituted.
  • the substituents are preferably one or more of the following groups independently selected from alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, alkylthio , alkylamino, halogen, amino, nitro, hydroxyl, mercapto, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkylthio, oxo, amido, sulfonamide, Formyl, formamide, carboxyl and carboxylate groups, etc.
  • halogen refers to F, Cl, Br and I. More preferably, halogen is selected from F, Cl or Br.
  • amino refers to -NH2 .
  • ester group refers to a group with the structure -COOR, wherein R represents hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl radical or substituted aryl, heterocycle or substituted heterocycle.
  • ester groups include, but are not limited to: -COOCH 3 , -COOCH 2 CH 3 , -COOCH 2 CH 2 CH 3 , -COOCH 2 CH(CH 3 ) 2 .
  • amide refers to a group having the structure -CONRR', where R and R' can independently represent hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl, or Substituted cycloalkenyl, aryl or substituted aryl, heterocycle or substituted heterocycle, as defined above. R and R' can be the same or different in the dialkylamine moiety. Examples of amide groups include, but are not limited to: -CONH 2 , -CONHCH 3 , -CON(CH 3 ) 2 , and the like.
  • sulfonamido refers to a group with the structure -SO 2 NRR' or RSO 2 NR'-, where R and R' can independently represent hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted Cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heterocycle or substituted heterocycle, as defined above. R and R' can be the same or different in the dialkylamine moiety.
  • sulfonamide groups include, but are not limited to: -SO 2 NH 2 , -SO 2 NHCH 3 , -SO 2 N(CH 3 ) 2 , CH 3 SO 2 NH—, CH 3 SO 2 NCH 3 —, and the like.
  • substituted means that one or more hydrogen atoms on a specific group are replaced by a specific substituent.
  • the specific substituents are the corresponding substituents described above, or the substituents appearing in each embodiment.
  • a substituted group may have a substituent selected from a specific group at any substitutable position of the group, and the substituents may be the same or different at each position.
  • substituents contemplated by this invention are those that are stable or chemically feasible.
  • substituted or unsubstituted the groups described in the present invention can be substituted by substituents selected from the following groups: D, halogen, cyano, nitro, hydroxyl , amino, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, 3-12 membered heterocyclyl, C3-C12 cycloalkyl, 5-10 membered heteroaryl , C6-C10 aryl.
  • substituents selected from the following groups: D, halogen, cyano, nitro, hydroxyl , amino, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, 3-12 membered heterocyclyl, C3-C12 cycloalkyl, 5-10 membered heteroaryl , C6-C10 aryl.
  • linking group -L1- listed in the present invention does not specify its linking direction
  • its linking direction can be connected in the same direction as the reading sequence from left to right, or in the opposite direction to the above direction.
  • the connection direction of the linking groups listed in the present invention is connected in the same direction as the reading order from left to right.
  • An example is as follows, The linking group -L1- is -CD-, if -CD- connects ring A and ring B in the same direction as the reading order from left to right to form If -CD- connects ring A and ring B in the direction opposite to the above direction to form
  • the structural formulas described herein are intended to include all stereoisomers (such as cis-trans isomers, enantiomers, diastereomers and geometric isomers (or conformers)): R, S configurations containing asymmetric centers, (Z), (E) isomers of double bonds, cis-trans isomers of naphthenes, etc. Accordingly, individual stereochemical isomers of the compounds of the present invention or mixtures thereof as enantiomers, diastereomers or geometric isomers (or conformers) are within the scope of the present invention.
  • solvate refers to a complex in which a compound represented by formula I coordinates with solvent molecules to form a specific ratio.
  • the compound of the present invention refers to the compound represented by formula I, and also includes stereoisomers, pharmaceutically acceptable salts, prodrugs or solvates of the compound represented by formula I.
  • the compounds of the present invention may contain cis-trans isomerism, one or more chiral carbon atoms, and thus may give rise to cis-trans isomers, chiral isomers, enantiomers, diastereoisomers and others Combined Stereoisomeric Forms.
  • cis-trans isomers Organic molecules containing this type of isomerism, such as alkenes, azo compounds, alicyclic hydrocarbons, etc., are regarded as cis-trans isomers, and cis means that the same ligand is in the adjacent position.
  • cis or “cis- "Indicates; trans means that the same ligand is in the diagonal position, generally expressed by "anti” or "trans-”.
  • Each chiral carbon atom can be defined as (R)- or (S)- based on stereochemistry.
  • the present invention is intended to include all possible isomers, as well as their racemates and optically pure forms.
  • the preparation of the compounds of the present invention can select racemates, cis-trans isomers, chiral isomers, diastereomers or enantiomers as raw materials or intermediates.
  • Optically active isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, such as crystallization and chiral chromatography.
  • the invention also includes isotopically labeled compounds (ie, isotopically derivatives) equivalent to the original compounds disclosed herein. In practice, however, substitution of one or more atoms by an atom with a different atomic mass or mass number usually occurs.
  • isotopes in the isotopic derivatives of the present invention include hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine isotopes, such as 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl. Isotopic derivatives of the compounds of the present invention are within the protection scope of the present invention.
  • 3 H-labeled compounds and 14 C-labeled compounds are useful in tissue distribution experiments of drugs and substrates.
  • Tritium (ie 3 H) and carbon-14 (ie 14 C) labeled compounds are relatively easy to prepare and detect, and are the first choice among isotopes.
  • substitution of heavier isotopes such as deuterium, ie 2 H may be preferred in some cases due to its good metabolic stability, which has advantages in certain therapies, such as increased half-life in vivo or reduced dosage.
  • Isotopically-labeled compounds can be prepared in general manner by substituting a readily available isotopically-labeled reagent for a non-isotopically-labeled reagent, using the schemes disclosed in the Examples.
  • pharmaceutically acceptable salt includes pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to a salt formed with an inorganic or organic acid that retains the biological effectiveness of the free base without other side effects.
  • Inorganic acid salts include but not limited to hydrochloride, hydrobromide, sulfate, nitrate, phosphate, etc.
  • organic acid salts include but not limited to formate, acetate, 2,2-dichloroacetate , Trifluoroacetate, Propionate, Caproate, Caprylate, Caprate, Undecylenate, Glycolate, Gluconate, Lactate, Sebacate, Hexanoate glutarate, malonate, oxalate, maleate, succinate, fumarate, tartrate, citrate, palmitate, stearate, oleate , cinnamate, laurate, malate, glutamate, pyroglutamate, aspartate, benzoate, mesylate, benzenesulfonate, p
  • “Pharmaceutically acceptable base addition salt” refers to a salt formed with an inorganic base or an organic base that can maintain the biological effectiveness of the free acid without other side effects.
  • Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like.
  • Preferred inorganic salts are ammonium, sodium, potassium, calcium and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, those of primary, secondary, and tertiary amines, substituted amines, including natural substituted amines, cyclic amines, and basic ion exchange resins , such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, triethanolamine, dimethylethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, bicyclic Hexylamine, Lysine, Arginine, Histidine, Caffeine, Procaine, Choline, Betaine, Ethylenediamine, Glucosamine, Methylglucamine, Theobromine, Purine, Piperazine, Piperazine Pyridine, N-ethylpiperidine, polyamine resin, etc.
  • Preferred organic bases include isopropylamine, diethylamine, ethanolamine, trimethylamine,
  • a specific stereoisomer of a compound of the present invention can be prepared by asymmetric synthesis, or derivatized with a chiral auxiliary, and the resulting stereo mixture is separated, and then the chiral auxiliary is removed to obtain a pure cis-trans monomers, chiral monomers or mixed stereomonomers.
  • the molecule contains a cis-trans isomerism center, it can be purified by column chromatography (normal phase silica gel column or reverse phase HPLC preparation) to obtain a pure cis product or trans product; in addition, if the molecule contains a basic functional group , such as amino acids, or acidic functional groups, such as carboxyl groups, can form diastereoisomeric salts with suitable optically active acids or bases, and then be separated by conventional means such as separation crystallization or chromatography, and then pure Enantiomer.
  • column chromatography normal phase silica gel column or reverse phase HPLC preparation
  • the compounds of the present invention may be substituted with any number of substituents or functional groups to broaden their scope.
  • substituted refers to replacing a hydrogen radical with a designated structural substituent.
  • substituents may be the same or different for each position.
  • substitution includes all permissible organic compound substitutions. Broadly speaking, the permissible substituents include acyclic, cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic organic compounds.
  • heteroatom nitrogen may have hydrogen substituents or any permissible organic compound as described above to complement its valence.
  • stable herein means having a compound that is stable, detectable for a sufficient period of time to maintain the structural integrity of the compound, preferably active for a sufficient period of time, and is used herein for the above purposes.
  • Metabolites of compounds represented by formula I and their pharmaceutically acceptable salts, and prodrugs that can be transformed into compounds represented by formula I and their pharmaceutically acceptable salts in vivo are also included in the protection scope of the present invention.
  • each reaction is usually carried out in an inert solvent at room temperature to reflux temperature (such as 0°C-150°C, preferably 10°C-100°C).
  • the reaction time is usually 0.1-60 hours, preferably 0.5-48 hours.
  • the preparation of the compound of the present invention comprises the steps of:
  • compound (I-3) reacts with compound (I-4) to obtain compound (I-5);
  • compound (I-5) is reacted with compound (I-6) to obtain compound (I-7);
  • reaction solvent reaction temperature, reaction time, catalyst, etc.
  • reaction time reaction time, catalyst, etc.
  • compositions and methods of administration are provided.
  • the pharmaceutical composition can be used to prevent and/or treat (stabilize, alleviate or cure) BTK kinase-related diseases (autoimmune diseases, inflammatory diseases or tumors, etc.).
  • the pharmaceutical composition of the present invention comprises the compound of the present invention and pharmaceutically acceptable excipients or carriers in a safe and effective amount range.
  • safe and effective amount refers to: the amount of the compound is enough to obviously improve the condition without producing serious side effects.
  • the pharmaceutical composition contains 1-2000 mg of the compound of the present invention per dose, more preferably 10-200 mg of the compound of the present invention per dose.
  • the "one dose” is a capsule or tablet.
  • “Pharmaceutically acceptable carrier” refers to: one or more compatible solid or liquid fillers or gel substances, which are suitable for human use, and must have sufficient purity and low enough toxicity. "Compatibility” herein means that the components of the composition can be blended with the compound of the present invention and with each other without significantly reducing the efficacy of the compound.
  • Examples of pharmaceutically acceptable carrier parts include cellulose and derivatives thereof (such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid , magnesium stearate), calcium sulfate, vegetable oil (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (such as ), wetting agent (such as sodium lauryl sulfate), coloring agent, flavoring agent, stabilizer, antioxidant, preservative, pyrogen-free water, etc.
  • cellulose and derivatives thereof such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.
  • gelatin such as talc
  • solid lubricants such as stearic acid , magnesium stearate
  • calcium sulfate such
  • the administration mode of the compound or pharmaceutical composition of the present invention is not particularly limited, and representative administration modes include (but not limited to): oral, parenteral (intravenous, intramuscular or subcutaneous).
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is admixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with: (a) fillers or extenders, for example, Starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders such as hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, For example, glycerol; (d) disintegrants, such as agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow agents, such as paraffin; (f) Absorption accelerators such as quaternary ammonium compounds; (g) wetting agents such as cetyl alcohol and glyceryl mono
  • Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared with coatings and shell materials, such as enteric coatings and others well known in the art. They may contain opacifying agents and, in such compositions, the release of the active compound or compounds may be in a certain part of the alimentary canal in a delayed manner.
  • coatings and shell materials such as enteric coatings and others well known in the art. They may contain opacifying agents and, in such compositions, the release of the active compound or compounds may be in a certain part of the alimentary canal in a delayed manner.
  • Examples of usable embedding components are polymeric substances and waxy substances.
  • the active compounds can also be in microencapsulated form, if desired, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures.
  • liquid dosage forms may contain inert diluents conventionally used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or mixtures of these substances, etc.
  • inert diluents conventionally used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and
  • compositions can also contain adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • Suspensions in addition to the active compounds, may contain suspending agents, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
  • suspending agents for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
  • compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Suitable aqueous and non-aqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols, and suitable mixtures thereof.
  • the compounds of the invention may be administered alone or in combination with other pharmaceutically acceptable compounds such as anti-HBV agents.
  • the pharmaceutical composition When administered in combination, the pharmaceutical composition also includes one or more (2, 3, 4, or more) other pharmaceutically acceptable compounds.
  • One or more (2, 3, 4, or more) of the other pharmaceutically acceptable compounds can be used simultaneously, separately or sequentially with the compound of the present invention for the prevention and/or treatment of BTK , especially those associated with BTK mutations.
  • a safe and effective amount of the compound of the present invention is applied to a mammal (such as a human) in need of treatment, wherein the dosage is a pharmaceutically effective dosage when administered, for a person with a body weight of 60kg, the daily
  • the dosage is generally 1-2000 mg, preferably 20-500 mg.
  • factors such as the route of administration and the health status of the patient should also be considered for the specific dosage, which are within the skill of skilled physicians.
  • the compound of the present invention has novel structure and excellent BTK and BTK (C481S) kinase inhibitor effect;
  • the compound of the present invention has good drug efficacy, good bioavailability, is safer, and can inhibit and degrade BTK and BTK (C481S) protein.
  • the compound of the present invention contains a unique E3 ubiquitin ligase ligand, which can weaken the substrate degradation effect caused by immunomodulatory imine drugs (IMiD), thereby reducing the activity of IMiD.
  • IMD immunomodulatory imine drugs
  • PTLC or TLC (thin layer chromatography) preparations were performed on 20 x 20 cm plates (500 micron thick silica gel); silica gel chromatography was performed on a Biotage flash chromatography system.
  • 1 H NMR hydrogen spectrum
  • Bruker AscendTM400 spectrometer 400MHz, 298°K
  • the chemical shift (ppm) of residual proton in deuterated reagent is given as a reference: the ⁇ (chemical shift) of CDCl3 is 7.26 ppm, the ⁇ of CD 3 OD is 3.31 ppm, and the ⁇ of DMSO-d6 is 2.50 ppm.
  • HPLC high performance liquid chromatography
  • MS mass spectrometry
  • MS mass range 150-750 amu; positive ion electrospray ionization.
  • MS mass range 150-750 amu; positive ion electrospray ionization.
  • MS mass range 150-750 amu; positive ion electrospray ionization.
  • Step 1 tert-butyl 6-(tosyloxy)-2-azaspiro[3.3]heptane-2-carboxylate
  • Step 2 6-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-2-azaspiro[3.3]heptane tert-butyl alkane-2-carboxylate
  • Step 3 3-(4-phenoxyphenyl)-1-(2-azaspiro[3.3]heptan-6-yl)-1H-pyrazolo[3,4-pyrimidin-4-amine
  • Step 4 3-(6-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-2-azaspiro[ 3.3] Heptane-2-yl)azetidine-1-carboxylate tert-butyl ester
  • Step 5 1-(2-(azetidin-3-yl)-2-azaspiro[3.3]heptan-6-yl)-3-(4-phenoxyphenyl)-1H- Pyrazolo[3,4-d]pyrimidin-4-amine
  • Step 6 5-(3-(6-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-2-nitrogen Heterospiro[3.3]heptane-2-yl)azetidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-di ketone
  • reaction solution was semi-preparatively purified and freeze-dried to obtain 5-(3-(6-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4 -d]pyrimidin-1-yl)-2-azaspiro[3.3]heptane-2-yl)azetidin-1-yl)-2-(2,6-dioxopiperidine-3- base) isoindoline-1,3-dione (5 mg, yield 16%).
  • LCMS (ESI): [M+H] + 710.4; HPLC: purity 96%.
  • Step 3 1-(2-(azetidine-1-carboxylate tert-butyl-3-yl)octahydrocyclopentyl-3-(4-phenoxyphenyl)-1H-pyrazolo [3,4-d]pyrimidin-4-amine
  • the reaction solution was poured into water, extracted with ethyl acetate, separated, the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated to obtain a crude product.
  • Step 4 1-(2-(azetidin-3-yl)octahydrocyclopentyl-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidine -4-amine
  • Step 5 3- ⁇ 3-[5- ⁇ 4-Amino-3-[4-(phenyloxy)phenyl]pyrazolo[3,4-d]pyrimidin-1-yl ⁇ octahydrocyclopenta And[1,2-c]pyrrol-2-yl]azetidin-1-yl ⁇ azetidin-1-carboxylate-2-methylpropan-2-yl ester
  • Step 6 1-[2-[1-(azetidin-3-yl)azetidin-3-yl]octahydrocyclopenta[1,2-c]pyrrol-5-yl]-3 -[4-(Phenyloxy)phenyl]pyrazolo[3,4-d]pyrimidin-4-amine
  • Step 7 Methyl 5-(3-(5-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazol[3,4-d]pyrimidin-1-yl)hexahydrocyclo Pentane[c]pyrrol-2(1H)-yl)-[1,3'-diazetidine]-1'-yl)picolinate
  • Step 8 5-(3-(5-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazol[3,4-d]pyrimidin-1-yl)hexahydrocyclopentane [c]pyrrol-2(1H)-yl)-[1,3'-diazetidine]-1'-yl)picolinic acid
  • Step 9 5-(3-(5-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)hexahydrocyclopenta [c] Pyrrol-2(1H)-yl)-(1,3'-diazetidine]-1'-yl)-N-(2,6-dioxopiperidin-3-yl)pyridineamide
  • reaction solution was diluted with water, extracted with ethyl acetate, and separated.
  • organic phase was dried over anhydrous sodium sulfate, filtered, concentrated, and purified by preparative thin-layer chromatography (developing solvent: 5% methanol/dichloromethane) to obtain 5-(3-(5-(4-amino-3-(4 -phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)hexahydrocyclopenta[c]pyrrol-2(1H)-yl)-(1,3'-di Azetidine]-1'-yl)-N-(2,6-dioxopiperidin-3-yl)pyridineamide, white solid (27 mg, yield 25%).
  • Step 1 4-(4-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazine-1 -Benzyl carboxylate
  • Step 2 3-(4-phenoxyphenyl)-1-(4-(piperazin-1-yl)cyclohexyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine
  • Step 3 tert-butyl 3-(4-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl) ring Hexyl)piperazin-1-yl)azetidine-1-carboxylate
  • Step 4 1-(4-(4-(azetidin-3-yl)piperazin-1-yl)cyclohexyl)-3-(4-phenoxyphenyl)-1H-pyrazolo [3,4-d]pyrimidin-4-amine
  • Step 5 5-(3-(4-(4-(4-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl) ring Hexyl)piperazin-1-yl)azetidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)-6-fluoroisoindoline-1,3 - dione
  • reaction solution was poured into water, extracted with ethyl acetate, separated, the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated to obtain a crude product, purified by prep.TLC to obtain 5-(3-(4-(4-(4-amino -3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl )-2-(2,6-dioxopiperidin-3-yl)-6-fluoroisoindoline-1,3-dione (58 mg, yield 33%).
  • Step 1 Benzyl 4-(4-Hydroxycyclohexyl)piperazine-1-carboxylate
  • Step 2 Benzyl 4-(4-(tosyloxy)cyclohexyl)piperazine-1-carboxylate
  • Step 3 Benzyl 4-(4-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d] Pyrimidin-1-yl)cyclohexyl)piperazine-1-carboxylate
  • Step 4 3-(4-(2-Fluoro-3-methoxyphenoxy)phenyl)-1-(4-(piperazin-1-yl)cyclohexyl)-1H-pyrazolo[3 ,4-d]pyrimidin-4-amine
  • Step 5 tert-butyl 3-(4-(4-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3, 4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)azetidine-1-carboxylate
  • Step 6 1-(4-(azetidin-3-yl)piperazin-1-yl)cyclohexyl)-3-(4-(2-fluoro-3-methoxyphenoxy)benzene base)-1H-pyrazolo[3,4-d]pyrimidin-4-amine
  • Step 7 5-(3-(4-(4-(4-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)1H-pyrazolo[3,4 -d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)-2-(2,6-dioxopyridine-3-yl)isoindole -1,3-dione
  • reaction solution was poured into water, extracted with ethyl acetate, separated, the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated to obtain a crude product, purified by prep.TLC to obtain 5-(3-(4-(4-(4-amino -3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl )azetidin-1-yl)-2-(2,6-dioxopyridine-3-yl)isoindole-1,3-dione (39 mg, yield 21%).
  • Step 1 Benzyl 4-(4-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3,4-d] Pyrimidin-1-yl)cyclohexyl)piperazine-1-carboxylate
  • Step 2 3-(4-(2-Fluoro-3-methoxyphenoxy)phenyl)-1-(4-(piperazin-1-yl)cyclohexyl)-1H-pyrazolo[3 ,4-d]pyrimidin-4-amine
  • Step 3 tert-butyl 3-(4-(4-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3, 4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)azetidine-1-carboxylate
  • Step 4 1-(4-(azetidin-3-yl)piperazin-1-yl)cyclohexyl)-3-(4-(2-fluoro-3-methoxyphenoxy)benzene base)-1H-pyrazolo[3,4-d]pyrimidin-4-amine
  • Step 5 5-(3-(4-(4-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3, 4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)picolinate methyl ester
  • Step 6 5-(3-(4-(4-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)1H-pyrazolo[3,4 -d] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) azetidin-1-yl) picolinic acid
  • Step 7 5-(3-(4-(4-(4-amino-3-(4-(2-fluoro-3-methoxyphenoxy)phenyl)-1H-pyrazolo[3, 4-d]pyrimidin-1-yl)cyclohexyl))piperazin-1-yl)azetidin-1-yl)-N-(2,6-dioxopiperidin-3-yl)pyridine Amide
  • Step 1 Methyl 5-(3-(4-(4-(4-amino-3-(4-amino-3-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazol[3,4-d ]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)picolinate
  • Step 2 5-(3-(4-(4-(4-amino-3-(4-amino-3-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidine -1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)pyridinecarboxylic acid
  • Step 3 5-(3-(4-(4-(4-amino-3-(4-amino-3-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidine -1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)-N-(2,6-dioxypiperidin-3-yl)pyridineamide
  • reaction solution is diluted with water, extracted with dichloromethane, and separated.
  • organic phase is washed with anhydrous sodium sulfate After drying, it was filtered, concentrated, and purified by preparative thin-layer chromatography (developing solvent: 10% methanol/dichloromethane) to obtain 5-(3-(4-(4-(4-amino-3-(4-amino-3 -(4-phenoxyphenyl)-1H-pyrazol[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)-N -(2,6-Dioxopiperidin-3-yl)pyridine amide, white solid (64 mg, yield 54%).
  • Cis-4-aminocyclohexanol hydrochloride (4.0g), N-benzyl-2-chloro-N-(2-chloroethyl)ethylamine hydrochloride (8.5g) and NaHCO 3 (13.3g ) was dissolved in ethanol (60 mL), and the mixture was stirred at 90° C. for 16 h. The mixture was concentrated under reduced pressure, and the residue was extracted with water (200 mL) and ethyl acetate (150 mL ⁇ 3).
  • Step 3 tert-butyl 4-((1s,4s)-4-hydroxycyclohexyl)piperazine-1-carboxylate
  • Step 4 tert-butyl 4-((1s,4s)-4-(methylsulfonyloxy)cyclohexyl)piperazine-1-carboxylate
  • Step 5 4-((1r,4r)-4-(4-Amino-3-bromo-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazine-1-carboxy tert-butyl acid
  • Step 6 4-((1r,4r)-4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl) ring Hexyl)piperazine-1-carboxylate tert-butyl ester
  • Step 7 3-(4-((1r,4r)-4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidine-1- Base) cyclohexyl) piperazin-1-yl) azetidine-1-carboxylate tert-butyl ester
  • Step 8 4-(3-(4-((1r,4r)-4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidine -1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)-5-fluorobenzene-1,2-dicarboxylic acid dimethyl ester
  • the crude product was dissolved in 20 mL of DMSO, dimethyl 4,5-difluorobenzene-1,2-dicarboxylate (405 mg) (see WO2018/172423 for the synthesis method) and DIPEA (603 mg) were added, and the reaction was stirred at 85°C for 6 h .
  • the reaction mixture was cooled to room temperature, diluted with ethyl acetate (200 mL), and washed with water (50 mL ⁇ 3).
  • Step 9 4-(3-(4-((1r,4r)-4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidine -1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)-5-fluorobenzene-1,2-dicarboxylic acid
  • Step 10 5-(3-(4-((1r,4r)-4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidine -1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)-6-fluoro-2-((S)-2,6-dioxopiperidin-3-yl) Isoindoline-1,3-dione
  • Step 1 5-(3-(4-((1r,4r)-4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidine -1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)-6-fluoro-2-((R)-2,6-dioxopiperidin-3-yl) Isoindoline-1,3-dione
  • Step 3 tert-butyl 4-((1r,4r)-4-hydroxycyclohexyl)piperazine-1-carboxylate
  • Step 4 tert-butyl 4-((1r,4r)-4-(methylsulfonyloxy)cyclohexyl)piperazine-1-carboxylate
  • Step 5 4-((1s,4s)-4-(4-Amino-3-bromo-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazine-1-carboxy tert-butyl acid
  • Step 6 4-((1s,4s)-4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl) ring Hexyl)piperazine-1-carboxylate tert-butyl ester
  • Step 7 3-(4-((1s,4s)-4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidine-1- Base) cyclohexyl) piperazin-1-yl) azetidine-1-carboxylate tert-butyl ester
  • Step 8 4-(3-(4-((1s,4s)-4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidine -1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)-5-fluorobenzene-1,2-dicarboxylic acid dimethyl ester
  • the crude product was dissolved in 10 mL of DMSO, dimethyl 4,5-difluorobenzene-1,2-dicarboxylate (230 mg) (see WO2018/172423 for the synthesis method) and DIPEA (335 mg) were added, and the reaction was stirred at 85°C for 6 h .
  • the reaction mixture was cooled to room temperature, diluted with ethyl acetate (100 mL), and washed with water (50 mL ⁇ 3).
  • Step 9 4-(3-(4-((1s,4s)-4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidine -1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)-5-fluorobenzene-1,2-dicarboxylic acid
  • Step 10 5-(3-(4-((1s,4s)-4-(4-amino-3-(4-phenoxyphenyl)-1H pyrazolo[3,4-d]pyrimidine- 1-yl)cyclohexyl)piperazin-1-yl]azidine-1-yl]-2-((S)-2,6-dioxypiperidin-3-yl)-6-fluoroisoindole -1,3-dione
  • Step 1 Dimethyl phthalate 4-(3-(4-((1r,4r)-4-(4-amino-3-(4-phenoxyphenyl)-1H pyrazolo[3 ,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl]azidine-1-yl
  • the crude product was dissolved in 20 mL of DMSO, dimethyl 4-fluorophthalate (373 mg) and DIPEA (603 mg) were added, and the reaction was stirred at 85° C. for 6 h.
  • the reaction mixture was cooled to room temperature , diluted with ethyl acetate (200mL), and washed with water (50mL ⁇ 3).
  • Step 2 4-(3-(4-((1r,4r)-4-(4-amino-3-(4-phenoxyphenyl)-1H pyrazolo[3,4-d]pyrimidine- 1-yl)cyclohexyl)piperazin-1-yl-azacyclo-1-yl]phthalic acid
  • Dimethyl phthalate 4-(3-(4-((1r,4r)-4-(4-amino-3-(4-phenoxyphenyl)-1H pyrazolo[3,4- d] pyrimidin-1-yl)cyclohexyl)piperazin-1-yl]azidine-1-yl (500mg) was dissolved in 6mL 1,4-dioxane, added 6mL methanol and 2mL sodium hydroxide solution (5M), stirred and reacted at 75°C for 2h. Cooled to room temperature, adjusted pH to 5.0 with 2M dilute hydrochloric acid, and concentrated under reduced pressure to obtain a crude product.
  • Step 3 5-(3-(4-((1r,4r)-4-(4-amino-3-(4-phenoxyphenyl)-1H pyrazolo[3,4-d]pyrimidine- 1-yl)cyclohexyl)piperazin-1-yl]azidine-1-yl]-2-((S)-2,6-dioxopiperidin-3-yl)isoindole-1,3 - dione
  • Step 1 5-(3-(4-((1r,4r)-4-(4-amino-3-(4-phenoxyphenyl)-1H pyrazolo[3,4-d]pyrimidine- 1-yl) cyclohexyl) piperazin-1-yl] azadin-1-yl] -2-( -2,6-dioxopiperidin-3-yl)isoindole-1,3-dione
  • Step 1 Dimethyl phthalate 4-(3-(4-((1s,4s)-4-(4-amino-3-(4-phenoxyphenyl)-1H pyrazolo[3 ,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl]azidine-1-yl
  • the crude product was dissolved in 10 mL DMSO, dimethyl 4-fluorophthalate (212 mg) and DIPEA (335 mg) were added, and the reaction was stirred at 85° C. for 6 h.
  • the reaction mixture was cooled to room temperature, diluted with ethyl acetate (100 mL), and washed with water (50 mL ⁇ 3).
  • Step 2 4-(3-(4-((1s,4s)-4-(4-amino-3-(4-phenoxyphenyl)-1H pyrazolo[3,4-d]pyrimidine- 1-yl)cyclohexyl)piperazin-1-yl-azacyclo-1-yl]phthalic acid
  • Step 3 5-(3-(4-((1s,4s)-4-(4-amino-3-(4-phenoxyphenyl)-1H pyrazolo[3,4-d]pyrimidine- 1-yl)cyclohexyl)piperazin-1-yl]azidine-1-yl]-2-((S)-2,6-dioxopiperidin-3-yl)isoindole-1,3 - dione
  • the activity detection of BTK (WT) and its mutant protein BTK (C481S) uses the homogeneous time-resolved fluorescent HTRF TK-s as the substrate, and BTK can catalyze the biotin-labeled polypeptide substrate TK-s with the participation of ATP Phosphorylation modification.
  • the Eu-labeled TK-s-specific phosphorylated antibody reacts with the substrate through the antigen-antibody reaction, and at the same time, the streptavidin-labeled acceptor XL665 interacts specifically with biotin, so that the Eu-labeled donor can interact with streptavidin
  • the Hetin-labeled receptors are spatially close to each other.
  • the Eu-labeled donor can emit energy at a wavelength of 620nm.
  • the biomolecular interaction causes the fluorescent groups of the donor and the acceptor to approach, part of the energy resonance is transferred to the acceptor XL665, making it excited , emits emission light with a wavelength of 665nm.
  • the emission at 665 nm is only due to donor-induced FRET.
  • the excitation wavelength was set to 320nm in Envision TM , and the fluorescence values were read at emission wavelengths of 620nm and 665nm. By detecting the change of the fluorescence ratio of 665nm and 620nm per unit time, the initial velocity of BTK reaction was calculated.
  • the final volume of the reaction was 10 ⁇ L, and the specific reaction system was 2% DMSO, 0.5ng/ ⁇ L BTK, 1 ⁇ M TK-s, 80 ⁇ M ATP, 5 mM MgCl 2 , 1 mM DTT, 20 nM SEB, 1 ⁇ kinase buffer (kinase buffer).
  • the experimental results are shown in Table 1
  • the compound synthesized by using the technology of the invention has a certain inhibitory effect on BTK and BTK (C481) kinase.
  • BTK (C481S) cells to detect the degradation ability of the compound on BTK (WT) and BTK (C481S) protein, Mino human mantle cell lymphoma cell line, purchased from ATCC, culture medium RPMI-1640+ 15% FBS + 1% double antibody; HEK293 OE BTK (C481S) human embryonic kidney cells 293 were transfected with BTK C481S plasmid to form overexpressed BTK C481S cells, DMEM + 10% FBS + 1% double antibody, added Mino to 6-well plate Cells and HEK293 OE BTK (C481S) stably transfected cell line suspension were left at 37°C for 0.5h, and compounds of different concentrations were added to the cell suspension, and treated at 37°C, 5% CO 2 , and saturated humidity for 24h.
  • BTK degradation rate
  • BTK% (1-(BTK administration / ⁇ -actin)/(BTK vehicle / ⁇ -actin)) ⁇ 100%
  • BTK administration is the expression level of BTK in Mino cells at different doses of the compound
  • BTK vehicle is the expression level of BTK in Mino cells in the control group.
  • the maximum concentration of the compound in this experimental system is 1000nM
  • D max is the maximum degradation value in the experimental concentration system.
  • a ⁇ 10nM; 10nM ⁇ B ⁇ 1000nM; 1000nM ⁇ C; '-' means that the degradation rate of the compound is less than 31% within the range of the test system, and 'ND' means that the data of this item has not been tested.
  • the IKZF1 protein is one of the target protein substrates for the degradation of amine compounds.
  • the IMiD activity of the compound is judged by the IKZF1 protein degradation experiment.
  • the Mino cell samples degraded by the above BTK protein were detected, and the IKZF1 (CST, Cat.14859S) protein was simultaneously detected by Western blot method.
  • IKZF1% (1-(IKZF1 administration / ⁇ -actin)/(IKZF1 vehicle / ⁇ -actin)) ⁇ 100%
  • IKZF1 administration is the expression level of IKZF1 in Mino cells at different doses of the compound
  • IKZF1 vehicle is the expression level of IKZF1 in Mino cells in the control group.
  • the maximum concentration of the compound in this experimental system is 1000nM
  • D max is the maximum degradation value in the experimental concentration system
  • use Graphpad5.0 software according to the formula Y 100/(1+10 ⁇ ((LogIC 50 -X)*HillSlope)) DC 50 values were fitted.
  • the compound of the present invention has better degradation activity to BTK (WT), BTK (C481S), and while retaining better degradation activity to BTK, BTK (C481S), it has better degradation activity to IKZF1
  • the degradation activity of the compound is weak, such as the compounds shown in Examples 20 and 25-39, especially the compounds shown in Examples 29 and 33.
  • the BTK target-sensitive cell OCI-LY10 was used to evaluate the inhibition of cell proliferation caused by BTK degradation, and the IMiD activity-sensitive cell MM.1S was used to evaluate the compound's inhibitory effect on cell proliferation caused by IMiD activity.
  • CCK8 CellTiter 96 Aqueous Non-Radioactive Cell Proliferation Kit
  • cell viability rate (%) (compound well value-medium well value)/(DMSO well value-medium well value) ⁇ 100%
  • the compound of the present invention has better proliferation inhibitory activity on BTK-sensitive OCI-LY10 cells, and has weaker inhibitory activity on IMiD-sensitive MM.1S, such as Examples 12, 28, Compounds shown in 29, 32, 34, 35, 36, 38 and 39.
  • the Liver Microsomal Stability Assay uses extracted microsomes from liver cells to study the metabolic stability of compounds. Incubate with different species of liver microsomes (final concentration 0.33mg/mL) in reaction buffer 0.1M Tris, 5mM MgCl 2 , 0.005%BSA, pH7.4 and 1 ⁇ M final concentration test compound at 37°C 10min. The reaction was then initiated by adding a final concentration of 1 mM NADPH. Samples were taken at 0, 7, 17, 30 and 60 minutes, and 4°C cold methanol was added to terminate the reaction. After collecting samples by centrifugation at 4000 rpm for 5 minutes, the drug content in the samples was analyzed by LC-MS/MS. Log (compound remaining percentage) and incubation time were linearly regressed to calculate the slope-k, and the relevant metabolic stability parameters were calculated by the following formula:
  • LW liver weight(g)(per species)
  • HBF hepatic blood flow(mL/min)(per species)
  • mice were used for each compound to carry out the test, and the oral administration dose of mice was 10mg/kg, and the vehicle was: 10% solutol, 10% DMSO, 80% normal saline, after administration, respectively, at 5min, 15min, 30min, Blood was collected at 8 time points: 1h, 2h, 4h, 6h, and 24h.
  • the compound of the present application has a good inhibitory effect on BTK and BTK (C481) kinase. (C481S) has a better degradation rate.
  • F-substituted part K (especially ) and the connection base is The compound of the present invention has excellent BTK, BTK (C481S) degradation activity, while the degradation activity to IKZF1 is weak, and has good proliferation inhibitory activity to BTK-sensitive OCI-LY10 cells, and to IMiD-sensitive MM.1S The inhibitory activity is weak.
  • part K containing F substitutions (especially ) and the connection base is The compound of the present invention has good stability and excellent pharmacokinetic properties for animal (especially human) liver microsomal metabolism.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (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)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

一种PROTAC化合物及其制备方法及应用,所述PROTAC化合物具有式(I)所示结构,其可用于治疗与BTK相关疾病如肿瘤或自身免疫系统疾病。

Description

PROTAC化合物、含其的药物组合物及其制备方法和应用 技术领域
本发明涉及一种PROTAC化合物、含其的药物组合物及其制备方法和应用。
背景技术
布鲁顿酪氨酸蛋白激酶(BTK,Bruton's tyrosine kinase)是非受体蛋白酪氨酸激酶Tec家族的成员,是B细胞抗原受体(BCR)信号通路中的关键调节因子,分布在淋巴组织、骨髓以及脾脏中。BCR信号通路的异常激活会引起下游肿瘤细胞的增殖、分化以及血管生成等信号通路的激活,BTK突变导致X-连锁无丙种球蛋白血症(XLA),其中B细胞成熟受损,导致免疫球蛋白生成减少(Hendriks,et al.;Expert Opin Ther Targets 15;1002-1021,2011)。BTK在B细胞信号传导和功能中的核心作用使其成为B细胞恶性肿瘤以及自身免疫和炎症性疾病的一个有吸引力的治疗靶点。
CRBN是一种具有离子蛋白酶活性的大脑相关蛋白,与DNA损伤结合蛋白-1(DDB1),滞蛋白4(Cullin4,CUL4A)和调节因子Cullins1(ROC1)相互作用形成功能性E3泛素连接酶复合体(CRBN-CRL4),该复合体通过底物受体(substrate receptor,SRs)识别特定的蛋白,促使泛素特异性结合到底物蛋白上,将需要降解的蛋白标记,从而启动降解过程。该复合体主要通过调节DNA修复、复制和转录作用参与细胞周期调节、胚胎发育和癌变等过程。度胺类抗肿瘤药的靶点是CRBN,通过抑制CRBN的自泛素化并且阻止CRBN与DDB1、CUL4A和ROC1一起形成CRBN-CRL4(一种E3泛素连接酶),从而调节其下游的信号发挥调节功能。
PROTAC(proteolysis targeting chimera)分子是一类能够同时结合靶蛋白和E3泛素连接酶的双功能化合物,此类化合物能够诱导靶蛋白被细胞的蛋白酶体识别,引起靶蛋白的降解,能够有效地降低靶蛋白在细胞中的含量。降解BTK蛋白可通过有效阻断BCR信号而对B细胞功能产生显著影响。去除BTK将消除BTK激酶活性以及BTK的任何蛋白质相互作用或支架功能。通过异源双功能小分子将BTK招募到泛素连接酶CRBN,从而促进BTK的泛素化和蛋白酶体降解,可以实现BTK的特异性降解。BTK(C481S)突变引起依鲁替尼等共价BTK抑制剂的耐药(Woyach,et al.;Blood,120(6):1175-1184,2012.),而这种降解方法也可以有效地靶向BTK(C481S)突变。WO2020239103、CN110845500、US11028088等展现了具有优异性能的PROTAC化合物。
由于BTK-PROTAC分子中含有度胺类化合物结构基团,可能会降解度胺类化合物的底物蛋白Ikaros(IKZF1)和Aiolos(IKZF3)等,引起血小板减少和中性粒细胞减少等多种毒副作用。
发明内容
本发明目的是提供一种药效好、生物利用度好、更安全、能抑制或降解BTK蛋白的PROTAC化合物,用于治疗与BTK相关疾病如肿瘤或自身免疫系统疾病,或炎症疾病。该类PROTAC化合物中独特新型的E3泛素连接酶配体能减弱免疫调节性亚胺药物(immunomodulatory imide drug,IMiD)引起的底物降解作用,从而降低IMiD活性。
本发明的第一方面,提供一种式I所示的化合物、其药学上可接受的盐、其顺反异构体,其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,
Figure PCTCN2022128411-appb-000001
其中,
Z 1选自:N或CR 1
Z 2选自:N或CR 1
Z 3选自:N或CR 7
Z 4选自:N或CR 7
Z 8选自:N或CH;
W 1选自:-O-、-S-、-NR b-、-CONR b-、-NR bCO-、-(CH 2) nNR b-或-(CH 2) nNR bCO-;
A 1选自:键、C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基、-(CH 2) pR c;其中,R c选自:C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基,其中,所述的杂环基、杂芳基含有1到4个选自O、S、N的杂原子;其中,所述C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基任选地被1-4个R a取代;
AL 1、AL 2和AL 3各自独立地选自:键、-O-、-S-、-NR' b-、-(CH 2) n'-、-CO-、-(CH 2) n'NR' b-、-CONR' b-、-NR' bCO-、-(CH 2) n'NR' bCO-;
A 2和A 3各自独立地选自:键、C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基,其中,所述的杂环基、杂芳基含有1到4个选自O、S、N的杂原子;其中,所述C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基任选地被1-4个R a取代;
W 2选自:键、-O-、-S-、-NR” b-、-(CH 2) n”-、-CONR” b-、-NR” bCO-、-(CH 2) n”NR bCO-;
R b、R' b和R” b各自独立地选自:H、C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6 元杂环基、C6-C10芳基、5-10元杂芳基;
R 1、R 2、R 3、R 4、R 5和R 6各自独立地选自:H、卤素、氰基、羟基、氨基、羧基、酰胺基、酯基、C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-15元杂芳基;其中,所述C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-15元杂芳基任选地被1-4个Ra取代;
R 7各自独立地选自:H、卤素、氰基、羟基、氨基、羧基、酰胺基、酯基、C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-15元杂芳基;其中,所述C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-15元杂芳基任选地被1-4个Ra取代;
或者,相邻的两个R 7以及与其连接的环原子共同形成C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基,优选地为C5-C6环烷基、5-6元杂环基、苯基、5-6元杂芳基;其中,所述环烷基、杂环基、芳基、苯基、杂芳基任选地被1-4个Ra取代;
或者,
Figure PCTCN2022128411-appb-000002
为环A 4,其中,任意一个与W 2相邻的R 7、W 2以及与它们连接的环原子形成环,环A 4选自:8-16元双环、三环或多环体系,优选地为9-14元的双环或三环体系;所述体系任选地被1-4个R a取代;任选地,其余R 7中,相邻的两个R 7以及与其连接的环原子共同形成C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基,优选地为C5-C6环烷基、5-6元杂环基、苯基、5-6元杂芳基;其中,所述环烷基、杂环基、芳基、苯基、杂芳基任选地被1-4个Ra取代;
或者,
Figure PCTCN2022128411-appb-000003
为环A 5,其中,任意一个与AL 3相邻的R 7、AL 3以及与它们连接的环原子形成环,环A 5选自:8-16元双环、三环或多环体系,优选地为9-14元的双环或三环体系;所述体系任选地被1-4个R a取代;任选地,其余R 7中,相邻的两个R 7以及与其连接的环原子共同形成C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基,优选地为C5-C6环烷基、5-6元杂环基、苯基、5-6元杂芳基;其中,所述环烷基、杂环基、芳基、苯基、杂芳基任选地被1-4个Ra取代;
或者,
Figure PCTCN2022128411-appb-000004
为环A 6,其中,任意一个与W 2相邻的R 7、W 2以及与它们连接的环原子形成环,任意一个与AL 3相邻的R 7、AL 3以及与它们连接的环原子形成环,环A 6选自:8-16元三环或多环体系,优选地为9-14元三环体系;所述体系任选地被1-4个R a取代;任选地,其余R 7中,相邻的两个R 7以及与其连接的环原子共同形成C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基,优选地为C5-C6环烷基、5-6元杂环基、苯基、5-6元杂芳基;其中,所述环烷基、杂环基、芳基、苯基、杂芳基任选地被1-4个Ra取代;
R 8选自:H、C1-C6烷基、
Figure PCTCN2022128411-appb-000005
Figure PCTCN2022128411-appb-000006
上述-CH 2-中的H任选地被R a取代;
R a选自:卤素、氰基、羟基、氨基、羧基、酰胺基、酯基、氧代基(=O)、C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-10元杂芳基;其中,所述C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-10元杂芳基任选地进一步被1-4个选自下组的基团取代:卤素、氰基、羟基、氨基、羧基、酰胺基、酯基、氧代基(=O)、C1-C6烷基、C1-C6烷氧基;
p、n、n'和n”各自独立地选自:1、2、3、4或5;
n 1、n 2、n 3、n 4、n 5、n 6和n 7各自独立地选自:0、1、2、3、4或5;
限定条件是:Z 1和Z 2中最多有一个为N;A 1、A 2和A 3中最多有一个为键;
当part K为
Figure PCTCN2022128411-appb-000007
且R 7为H、卤素、氰基、羟基、氨基、羧基、C1-C4烷基、C1-C4烷氧基和C3-C6环烷基中的一个或多个,R 8为H或C1-C4烷氧基时,A 1不为5-6元杂环基和5-6元杂芳基。
在另一优选例中,所述化合物具有式II-IV所示的结构
Figure PCTCN2022128411-appb-000008
Figure PCTCN2022128411-appb-000009
其中,
R 13独立地选自:卤素、氰基、羟基、氨基、羧基、酰胺基、酯基、氧代基(=O)、C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-10元杂芳基;其中,所述C1-C6烷基、C1-C6烷氧基、C1-C6烷基、C1-C6烷基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-10元杂芳基任选进一步被1-4个选自下组的基团取代:卤素、氰基、羟基、氨基、羧基、酰胺基、酯基、氧代基(=O)、C1-C6烷基、C1-C6烷氧基;
n 13选自:0、1、2、3或4;
W 1、W 2、A 1、A 2、A 3、环A 4、环A 5、环A 6、Z 1、Z 2、Z 8、AL 1、AL 2、AL 3、R 1、R 2、R 3、R 4、R 5、R 6、R 8、n 1、n 2、n 3、n 4、n 5和n 6的定义如上所述。
在另一优选例中,R 13各自独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、OMe、Me、CH 2OH、CO 2H、CF 3、CHF 2或CONH 2
在另一优选例中,A 1选自:键、C4-C7环烷基、5元氧杂环烷、6元氧杂环烷、5元氮杂环烷、6元氮杂环烷、7元氮杂环烷基、氧代氮杂环庚烷基、苯基、吡唑基、吡啶基、吡嗪基、苄基、C7-C15螺环基、C6-C15并环基、C5-C15桥环基、C5-C15杂螺环基、C4-C15杂并环基、C5-C15杂桥环基;其中,所述的C5-C15杂螺环基、C4-C15杂并环基、C5-C15杂桥环基含有1到4个选自O、S、N的杂原子;优选地,A 1选自:4元氮杂环烷基、5元氮杂环烷基、6元氮杂环烷基、7元氮杂环烷基、环丁基、环戊基、环己基、环庚基、四氢呋喃基、四氢吡喃基、吗啡啉基、哌啶基、哌嗪基、吡咯烷基、 螺环[3.3]庚烷基、2-氮杂螺环[3.3]庚烷基、2,6-二氮螺环[3.3]庚烷基、环戊烷并环戊烷基、环戊烷并四氢吡咯基、2-氮杂双环[2.2.1]庚烷基、6-氮杂螺环[3.4]辛烷基、2-氮杂螺环[3.4]辛烷基、7-氮杂螺环[3.5]壬烷基、2-氮杂螺环[4.4]壬烷基、2-氮杂螺环[3.5]壬烷基、双环[1.1.1]戊烷基、苯基、吡啶基、吡嗪基、吡唑基、苯并四氢吡咯基、苯并哌啶基、苯并吗啡啉基、苯并哌嗪基、吡啶并四氢吡咯基、吡啶并哌啶基、吡啶并吗啡啉基、吡啶并哌嗪基、吡嗪并四氢吡咯基、吡嗪并哌啶基、吡嗪并吗啡啉基、吡嗪并哌嗪基、2,7-二氮螺环[4.4]壬烷基、2,7-二氮螺环[3.5]壬烷基、2,6-二氮螺环[3.4]辛烷基或键。
在另一优选例中,A 1选自:
Figure PCTCN2022128411-appb-000010
Figure PCTCN2022128411-appb-000011
Figure PCTCN2022128411-appb-000012
其中,上述基团任选地被1-4个R a取代,R a的定义如上所述。
在另一优选例中,part K选自:
Figure PCTCN2022128411-appb-000013
Figure PCTCN2022128411-appb-000014
在另一优选例中,part K不为
Figure PCTCN2022128411-appb-000015
在另一优选例中,AL 1及AL 2均为键。
在另一优选例中,AL 1、AL 2及AL 3均为键。
在另一优选例中,
Figure PCTCN2022128411-appb-000016
选自
Figure PCTCN2022128411-appb-000017
Figure PCTCN2022128411-appb-000018
Figure PCTCN2022128411-appb-000019
优选地,上述各基团的右端与part K连接。
在另一优选例中,AL 1及AL 2均为键,且所述化合物具有式II-1所示的结构,
Figure PCTCN2022128411-appb-000020
其中,A 1、A 2、A 3、R 3、n 3和partK的定义如上所述。
在另一优选例中,part K选自
Figure PCTCN2022128411-appb-000021
Figure PCTCN2022128411-appb-000022
Figure PCTCN2022128411-appb-000023
在另一优选例中,AL 1及AL 2均为键,且所述化合物具有式II-2所示的结构,
Figure PCTCN2022128411-appb-000024
其中,A 1、A 2、A 3、R 7、n 7、R 3和n 3的定义如上所述。
在另一优选例中,A 1
Figure PCTCN2022128411-appb-000025
Figure PCTCN2022128411-appb-000026
优选地,上述各基团的右端与A 2连接。
在另一优选例中,A 2
Figure PCTCN2022128411-appb-000027
Figure PCTCN2022128411-appb-000028
优选地,上述各基团的右端与A 3连接。
在另一优选例中,A 3
Figure PCTCN2022128411-appb-000029
或键。优选地,上述各基团的右端与part K连接。
在另一优选例中,
Figure PCTCN2022128411-appb-000030
为:
Figure PCTCN2022128411-appb-000031
Figure PCTCN2022128411-appb-000032
Figure PCTCN2022128411-appb-000033
优选地,上述各基团的右端与part K连接。
在另一优选例中,
Figure PCTCN2022128411-appb-000034
Figure PCTCN2022128411-appb-000035
其中,R 3-1、R 3-2、R 3-3、R 3-4和R 3-5各自独立地选自H、卤素(氟、氯、溴或碘)、羟基、氨基、C1-C6烷基(例如甲基、乙基、正丙基、异丙基、正丁基、异丁基或叔丁基)、C1-C6烷氧基(例如甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基或叔丁氧基)或C1-C6烷基氨基(例如甲基氨基、乙基氨基、正丙基氨基、异丙基氨基、正丁基氨基、异丁基氨基或叔丁基氨基)。
在另一优选例中,R 3-1选自卤素,例如氟、氯、溴或碘,优选为氟。
在另一优选例中,R 3-2选自C1-C6烷氧基,优选为甲氧基。
在另一优选例中,R 3-3选自H。
在另一优选例中,R 3-4选自H。
在另一优选例中,R 3-5选自H。
在另一优选例中,R 3-1选自氟,R 3-2选自甲氧基,R 3-3选自H,R 3-4选自H,R 3-5选自H。
在另一优选例中,所述化合物具有式V所示的结构
Figure PCTCN2022128411-appb-000036
其中,
A 1选自:键、3-20元环烷基、3元杂环基、4元杂环基、7-20元杂环基、C6-C10芳基、7-15元杂芳基;其中,所述的杂环基、杂芳基含有1到4个选自O、S、N的杂原子;其中,所述3-20元环烷基、3元杂环基、4元杂环基、7-20元杂环基、C6-C10芳基、7-15元杂芳基任选地被1-4个R a取代;
W 1、A 1、A 2、A 3、Z 1、Z 2、AL 1、AL 2、R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、n 1、n 2、n 3、n 4、n 5、n 6和n 7的定义如上所述;
特别地,A 1选自:键、C4-C7环烷基、7元氮杂环烷基、氧代氮杂环庚烷基、苄基、C7-C15螺环基、C6-C15并环基、C5-C15桥环基、C7-C15杂螺环基、C4杂并环基、C7-C15杂并环基、C7-C15杂桥环基;其中,所述的C7-C15杂螺环基、C4杂并环基、C7-C15杂并环基、C7-C15杂桥环基含有1到4个选自O、S、N的杂原子;优选地,A 1选自:4元氮杂环烷基、7元氮杂环烷基、环丁基、环戊基、环己基、环庚基、螺环[3.3]庚烷基、2-氮杂螺环[3.3]庚烷基、2,6-二氮螺环[3.3]庚烷基、环戊烷并环戊烷基、环戊烷并四氢吡咯基、2-氮杂双环[2.2.1]庚烷基、6-氮杂螺环[3.4]辛烷基、2-氮杂螺环[3.4]辛烷基、7-氮杂螺环[3.5]壬烷基、2-氮杂螺环[4.4]壬烷基、2-氮杂螺环[3.5]壬烷基、双环[1.1.1]戊烷基、苯并四氢吡咯基、苯并哌啶基、苯并吗啡啉基、苯并哌嗪基、吡啶并四氢吡咯基、吡啶并哌啶基、吡啶并吗啡啉基、吡啶并哌嗪基、吡嗪并四氢吡咯基、吡嗪并哌啶基、吡嗪并吗啡啉基、吡嗪并哌嗪基、2,7-二氮螺环[4.4]壬烷基、2,7-二氮螺环[3.5]壬烷基、2,6-二氮螺环[3.4]辛烷基或键;
更特别地,A 1选自:
Figure PCTCN2022128411-appb-000037
Figure PCTCN2022128411-appb-000038
Figure PCTCN2022128411-appb-000039
其中,上述基团任选地被1-4个R a取代,R a的定义如上所述。
在另一优选例中,R 7各自独立地选自:-H、-F、-Cl、-Br、-I、-OH、-NH 2、-CN、-OMe、-Me、-CH 2OH、-CO 2H、-CF 3、-CHF 2、-CONH 2;优选地,R 7各自独立地选自:-H、-F、-OMe、-Me、-CF 3或-CHF 2
在另一优选例中,当与其它取代基成环时,R 7各自独立地选自:-NHCO-、-N(Me)N=、-OCO-、-O-CH 2-、-O-、-N(CF 3)N=、-N=CH-CH=、-N(Me)CO-、-N(CF 3)CO-。
在另一优选例中,相邻的两个R 7以及与其连接的环原子共同形成环时,
Figure PCTCN2022128411-appb-000040
选自:
Figure PCTCN2022128411-appb-000041
在另一优选例中,环A 4选自:
Figure PCTCN2022128411-appb-000042
Figure PCTCN2022128411-appb-000043
在另一优选例中,环A 5选自:
Figure PCTCN2022128411-appb-000044
在另一优选例中,环A 6选自:
Figure PCTCN2022128411-appb-000045
在另一优选例中,R 1、R 2、R 3、R 4、R 5和R 6各自独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、OMe、Me、CH 2OH、CO 2H、CF 3、CHF 2或CONH 2
在另一优选例中,Z 1为CH。
在另一优选例中,Z 2为CH。
在另一优选例中,W 1选自:-O-、-S-、-NH-、-CONH-、-NHCO-或-CH 2NHCO-。
在另一优选例中,W 2选自:-O-、-S-、-NH-、-CH 2-、-CONH-、-NHCO-、-CH 2NHCO-或键。
在另一优选例中,AL 1、AL 2和AL 3各自独立地选自:-O-、-S-、-NH-、-CH 2-、-CO-、-CH 2NH-、-CH 2N(CH 3)-、-CH 2CH 2-、-CONH-、-NHCO-、-CH 2NHCO-或键。
在另一优选例中,A 2和A 3各自独立地选自:键、3-12元杂环基、C3-C12环烷基、5-15元杂芳基、5-10元芳基或键,其中,所述的杂环基、杂芳基含有1到4个选自O、S、N的杂原子;优选地,A 2和A 3选自:4元氮杂环烷基、5元氮杂环烷基、6元氮杂环烷基、7元氮杂环烷基、环丁基、环戊基、环己基、环庚基、四氢呋喃基、四氢吡喃基、吗啡啉基、哌啶基、哌嗪基、吡咯环基、螺环[3.3]庚烷基、2-氮杂螺环[3.3]庚烷基、2,6-二氮螺环[3.3]庚烷基、环戊烷并环戊烷基、环戊烷并四氢吡咯基、2-氮杂双环[2.2.1]庚烷基、6-氮杂螺环[3.4]辛烷基、2-氮杂螺环[3.4]辛烷基、7-氮杂螺环[3.5]壬烷基、2-氮杂螺环[4.4]壬烷基、2-氮杂螺环[3.5]壬烷基、双环[1.1.1]戊烷基、苯基、吡啶基、吡嗪基、吡唑基、苯并四氢吡咯基、苯并哌啶基、苯并吗啡啉基、苯并哌嗪基、吡啶并四氢吡咯基、吡啶并哌啶基、吡啶并吗啡啉基、吡啶并哌嗪基、吡嗪并四氢吡咯基、吡嗪并 哌啶基、吡嗪并吗啡啉基、吡嗪并哌嗪基、2,7-二氮螺环[4.4]壬烷基、2,7-二氮螺环[3.5]壬烷基、2,6-二氮螺环[3.4]辛烷基或键。
在另一优选例中,A 2和A 3各自独立地选自:
Figure PCTCN2022128411-appb-000046
Figure PCTCN2022128411-appb-000047
Figure PCTCN2022128411-appb-000048
其中,上述基团任选地被1-4个R a取代,R a的定义如上所述。
在另一优选例中,
Figure PCTCN2022128411-appb-000049
Figure PCTCN2022128411-appb-000050
在另一优选例中,
Figure PCTCN2022128411-appb-000051
选自:
Figure PCTCN2022128411-appb-000052
Figure PCTCN2022128411-appb-000053
在另一优选例中,part L选自:
Figure PCTCN2022128411-appb-000054
Figure PCTCN2022128411-appb-000055
Figure PCTCN2022128411-appb-000056
在另一优选例中,W 1、W 2、A 1、A 2、A 3、环A 4、环A 5、环A 6、Z 1、Z 2、Z 3、Z 4、Z 8、AL 1、AL 2、AL 3、R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、n 1、n 2、n 3、n 4、n 5、n 6和n 7为实施例中各具体化合物所对应基团。
在另一优选例中,所述化合物具有式VI-1或VI-2所示的结构,
Figure PCTCN2022128411-appb-000057
其中,
R 7-1、R 7-2和R 7-3各自独立地选自H、卤素(氟、氯、溴或碘)、羟基、氨基、C1-C6烷基(例如甲基、乙基、正丙基、异丙基、正丁基、异丁基或叔丁基)、C1-C6烷氧基(例如甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基或叔丁氧基)或C1-C6烷基氨基(例如甲基氨基、乙基氨基、正丙基氨基、异丙基氨基、正丁基氨基、异丁基氨基或叔丁基氨基);
R 3-1、R 3-2、R 3-3、R 3-4和R 3-5各自独立地选自H、卤素(氟、氯、溴或碘)、羟基、氨基、C1-C6烷基(例如甲基、乙基、正丙基、异丙基、正丁基、异丁基或叔丁基)、C1-C6烷氧基(例如甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基或叔丁氧基)或C1-C6烷基氨基(例如甲基氨基、乙基氨基、正丙基氨基、异丙基氨基、正丁基氨基、异丁基氨基或叔丁基氨基);
Z 1、Z 2、W 1、R 1、R 2、R 4、R 5、R 6、n 1、n 2、n 4、n 5、n 6的定义如上所述;
条件是:R 7-1、R 7-2、R 7-3、R 3-1、R 3-2、R 3-3、R 3-4和R 3-5不同时为H。
在另一优选例中,所述化合物具有式VI-3或VI-4所示的结构,
Figure PCTCN2022128411-appb-000058
其中,
R 7-1、R 7-2、R 7-3、R 3-1、R 3-2、R 3-3、R 3-4和R 3-5定义如上所述;
条件是:R 7-1、R 7-2、R 7-3、R 3-1、R 3-2、R 3-3、R 3-4和R 3-5不同时为H。
在另一优选例中,R 7-1选自卤素,例如氟、氯、溴或碘,优选为氟。
在另一优选例中,R 7-2选自H。
在另一优选例中,R 7-3选自H。
在另一优选例中,R 7-1选自氟,R 7-2选自H,R 7-3选自H。
在另一优选例中,R 3-1选自卤素,例如氟、氯、溴或碘,优选为氟。
在另一优选例中,R 3-2选自C1-C6烷氧基,优选为甲氧基。
在另一优选例中,R 3-3选自H。
在另一优选例中,R 3-4选自H。
在另一优选例中,R 3-5选自H。
在另一优选例中,R 3-1选自氟,R 3-2选自甲氧基,R 3-3选自H,R 3-4选自H,R 3-5选自H。
在另一优选例中,所述化合物选自:
Figure PCTCN2022128411-appb-000059
Figure PCTCN2022128411-appb-000060
Figure PCTCN2022128411-appb-000061
Figure PCTCN2022128411-appb-000062
Figure PCTCN2022128411-appb-000063
Figure PCTCN2022128411-appb-000064
在另一优选例中,所述化合物选自:
Figure PCTCN2022128411-appb-000065
Figure PCTCN2022128411-appb-000066
Figure PCTCN2022128411-appb-000067
Figure PCTCN2022128411-appb-000068
Figure PCTCN2022128411-appb-000069
Figure PCTCN2022128411-appb-000070
Figure PCTCN2022128411-appb-000071
Figure PCTCN2022128411-appb-000072
Figure PCTCN2022128411-appb-000073
在另一优选例中,所述化合物选自实施例中所示化合物。
本发明的第二方面,提供一种药物组合物,其包含第一方面所述的化合物、其药学上可接受的盐、其顺反异构体,其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,和一种或多种药学上可接受的载体或赋形剂。
在另一优选例中,所述药物组合物还包括其他治疗剂。
在另一优选例中,提供一种药物组合物的制备方法,包括步骤:将药学上可接受的载体或赋形剂,与本发明化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物进行混合,从而形成药物组合物。
本发明的第三方面,提供一种如第一方面所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,或如第二方面所述的药物组合物在制备治疗BTK介导的疾病的药物中的用途。
在另一优选例中,所述疾病选自:自体免疫疾病、炎性疾病或肿瘤;
其中,所述自体免疫疾病优选选自:狼疮、多发性硬化、肌肉缩性侧索硬化、类风湿性关节炎、银屑病、因器官移植导致的并发症、糖尿病、哮喘、特应性皮炎、自身免疫性甲状腺病、溃疡性结肠炎、克罗恩病、阿尔茨海默病、白血病或淋巴瘤;
所述炎性疾病优选选自:角膜炎、鼻炎、口腔炎、腮腺炎、咽炎、扁桃体炎、气管炎、支气管炎、肺炎、心肌炎、胃炎、肠胃炎、胆囊炎或阑尾炎;
所述肿瘤优选选自:小淋巴细胞淋巴瘤、急性淋巴细胞白血病、慢性淋巴细胞白血病、急性骨髓性白血病、慢性骨髓性白血病、急性早幼粒细胞白血病、慢性粒细胞白血病、弥漫性大B细胞淋巴瘤、血管内大B细胞淋巴瘤、原发性渗出性淋巴瘤、华氏巨球蛋白血症、滤泡性淋巴瘤、多发性骨髓瘤或套细胞淋巴瘤。
本发明的第四方面,提供一种如第一方面所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,或如第二方面所述的药物组合物在制备用于抑制或调节BTK蛋白激酶活性的药物中的用途。
其中,所述BTK蛋白激酶优选为非突变的BTK蛋白激酶或突变的BTK蛋白激酶;所述突变的BTK蛋白激酶优选为C481S突变的BTK蛋白激酶。
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。
具体实施方式
本发明人通过广泛而深入的研究,首次发现一种新的PROTAC化合物,其药效好、生物利用度好、更安全、能抑制并降解BTK蛋白,并可以更少地引发或不引发IMiD降解作用。在此基础上完成了本发明。
本文中,除非特别指出,所用术语具有本领域技术人员公知的一般含义。
当通过从左向右书写的常规化学式描述取代基时,该取代基也同样包括从右向左书写结构式时所得到的在化学上等同的取代基。
如本文所用,在提到具体列举的数值中使用时,术语“约”意指该值可以从列举的值变动不多于1%。例如,如本文所用,表述“约100”包括99和101和之间的全部值(例如,99.1、99.2、99.3、99.4等)。
如本文所用,术语“含有”或“包括(包含)”可以是开放式、半封闭式和封闭式的。换言之,所述术语也包括“基本上由…构成”、或“由…构成”。
如本文所用,术语“烷基”包括直链或支链的烷基。例如C 1-C 6烷基表示具有1-6个(例如1、2、3、4、5或6个)碳原子的直链或支链的烷基,例如甲基、乙基、丙基、异丙基、丁基、异丁基、叔丁基等。
如本文所用,术语“环烷基”是指包含特定数目的C原子的环状烷基,如“C3-C20环烷基”指具有3-20个(例如3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)碳原子的环烷基。优选地为C3-C12环烷基,更优选地为C3-C6环烷基。环烷基可以是单环,例如环丙基、环丁基、环戊基、环己基、或类似基团。也可 以是双环形式,例如桥环或螺环形式。所述环烷基也可以稠合于芳基、杂芳基、杂环基上,其中与母体结构连接在一起的环为环烷基,如
Figure PCTCN2022128411-appb-000074
等。本文中,环烷基意在包含取代环烷基。
如本文所用,术语“C1-C6烷氧基”是指具有1-6个碳原子(例如1、2、3、4、5或6个)的直链或支链的烷氧基;其具有式C1-C6烷基-O-或-C1-C5烷基-O-C1-C5烷基(如,-CH 2-O-CH 2CH 3、-CH 2-O-(CH 2) 2CH 3、-CH 2CH 2-O-CH 2CH 3)结构,优选地为C1-C6烷基-O-,例如,甲氧基、乙氧基、丙氧基、异丙氧基、丁氧基、异丁氧基或叔丁氧基等。
如本文所用,“杂环基(heterocyclyl)”是指具有N、S和O的杂原子的饱和或部分饱和的环状基团(包含但不限于如3-7元单环,6-11元双环,或8-16元三环系统),“3-20元杂环基”是指具有3-20个(例如3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)原子的且其中1-4个(例如1、2或3个)原子为选自下组N、S和O的杂原子的饱和或部分饱和的环状基团。优选地其可以是单环,也可以是双环形式,例如桥环或螺环形式。杂环基优选4-12元杂环基,更优选4-8元或4-6元杂环基。具体的实例可以为氧杂环丁烷、氮杂环丁烷、四氢-2H-吡喃基、哌啶基、哌嗪基、四氢呋喃基、吗啉基和吡咯烷基等。所述杂环基可以稠合于杂芳基、芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基,如
Figure PCTCN2022128411-appb-000075
等。
如本文所用,“芳基”是指环上不含杂原子的芳香族环基,“C6-C10芳基”是指在环上不含杂原子的具有6至10个(例如6、7、8、9、10个)碳原子的芳香族环基,所述芳基可以稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为芳基环。如苯基(即六元芳基)、萘基等,其中六元芳基还意在包含六元芳基并5-6元环烷基(如
Figure PCTCN2022128411-appb-000076
)和六元芳基并5-6元杂环基(如
Figure PCTCN2022128411-appb-000077
等)。C6-C12芳基优选C6-C10芳基。芳基可以是任选取代的或未取代的。
如本文所用,“杂芳基”指具有1-4个(例如1、2或3个)原子为选自下组N、S和O的杂原子的环状芳香基,“5-15元杂芳基”指具有5-15个(例如5、6、7、8、9、10、11、12、13、14或15个)原子的且其中1-4个(例如1、2或3个)原子为选自下组N、S和O的杂原子的环状芳香基团。杂芳基优选5-12元杂芳基,优选5-10元,更优选地5-6元,其可以是单环,也可以是稠环形式。具体的实例可以为吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基、吡咯基、吡唑基、咪唑基、(1,2,3)-三唑基以及(1,2,4)-三唑基、四唑基、呋喃基、噻吩基、异恶唑基、噻唑基、恶唑基等。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环。杂芳基可以是任选取代的或未取代的。当被取代时,取代基优选为一个或多个以下基团,其独立地选自 烷基、氘代烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、烷硫基、烷基氨基、卤素、氨基、硝基、羟基、巯基、氰基、环烷基、杂环基、芳基、杂芳基、环烷硫基、氧代基、酰胺基、磺酰胺基、甲酰基、甲酰胺基、羧基和羧酸酯基等。
如本文所用,“卤素”指F、Cl、Br和I。更佳地,卤素选自F、Cl或Br。
本文中,“氨基”是指-NH 2
本文中,“羧基”是指-COOH。
术语“酯基”是指带有结构-COOR的基团,其中R代表氢、烷基或取代的烷基、环烷基或取代的环烷基、环烯基或取代的环烯基、芳基或取代的芳基、杂环或取代的杂环。酯基的实例包括但不限:-COOCH 3、-COOCH 2CH 3、-COOCH 2CH 2CH 3、-COOCH 2CH(CH 3) 2
术语“酰胺基”是指带有结构-CONRR'的基团,其中R和R'可以独立地代表氢、烷基或取代的烷基、环烷基或取代的环烷基、环烯基或取代的环烯基、芳基或取代的芳基、杂环或取代的杂环,如上文所定义。R和R'在二烷基胺片段中可以相同或不同。酰胺基的实例包括但不限于:-CONH 2、-CONHCH 3、-CON(CH 3) 2等。
术语“磺酰胺基”是指带有结构-SO 2NRR'或RSO 2NR'-的基团,其中R和R'可以独立地代表氢、烷基或取代的烷基、环烷基或取代的环烷基、环烯基或取代的环烯基、芳基或取代的芳基、杂环或取代的杂环,如上文所定义。R和R'在二烷基胺片段中可以相同或不同。磺酰胺基的实例包括但不限:-SO 2NH 2、-SO 2NHCH 3、-SO 2N(CH 3) 2、CH 3SO 2NH-、CH 3SO 2NCH 3-等。
本文中,术语“取代”指特定的基团上的一个或多个氢原子被特定的取代基所取代。特定的取代基为在前文中相应描述的取代基,或各实施例中所出现的取代基。除非特别说明,某个取代的基团可以在该基团的任何可取代的位点上具有一个选自特定组的取代基,所述的取代基在各个位置上可以是相同或不同的。本领域技术人员应理解,本发明所预期的取代基的组合是那些稳定的或化学上可实现的组合。
本发明所述的基团除非特别说明是“取代的或未取代的”,否则本发明的基团均可被选自下组的取代基所取代:D、卤素、氰基、硝基、羟基、氨基、C1-C6烷基、C2-C6烯基、C2-C6炔基、C1-C6烷氧基、3-12元杂环基、C3-C12环烷基、5-10元杂芳基、C6-C10芳基。
本文中“任选地”指的是随后描述的事件或状况可能但不是必须出现的,并且该描述包括所述事件或状况发生的情况,以及所述事件或状况不发生的情况。
本文中,术语“多个”独立指2、3、4、5个或大于5个的正整数。
本发明所列举的连接基团-L1-没有指明其连接方向时,其连接方向可以按与从左往右的读取顺序相同的方向进行连接,也可以按与上述方向相反的方向进行连接。例如,本发明所列举的连接基团的连接方向按与从左往右的读取顺序相同的方向进行连接。举例说明如下,
Figure PCTCN2022128411-appb-000078
中连接基团-L1-为-C-D-,若-C-D-按与从左往右的读取 顺序相同的方向连接环A和环B构成
Figure PCTCN2022128411-appb-000079
若-C-D-按与上述方向相反的方向连接环A和环B构成
Figure PCTCN2022128411-appb-000080
除非特别说明,本发明所描述的结构式意在包括所有立体异构体(如顺反异构体、对映异构、非对映异构和几何异构体(或构象异构体)):含有不对称中心的R、S构型,双键的(Z)、(E)异构体,环烷烃的顺反异构体等。因此,本发明的化合物的单个立体化学异构体或其对映异构体、非对映异构体或几何异构体(或构象异构体)的混合物都属于本发明的范围。
如本文所用,术语“溶剂化物”是指式I所示的化合物与溶剂分子配位形成特定比例的配合物。
活性成分
如本文所用,“本发明的化合物”指式I所示的化合物,并且还包括式I所示化合物的立体异构体、药学上可接受的盐、前药或溶剂化物。
本发明的化合物可能含有顺反异构,一个或多个手性碳原子,且因此可产生顺反异构体、手性异构体、对映异构体、非对映异构体及其它组合的立体异构形式。顺反异构是指化合物分子中由于具有自由旋转的限制因素,使各个基团在空间的排列方式不同而出现的非对映异构现象。这种限制因素一般是有机化合物结构中出现如C=C双键、C=N双键、C=S双键、N=N双键、杂环或环烷烃等不能自由旋转的官能团所引起的。含有此类异构的有机分子如烯烃、偶氮化合物、脂环烃等被视作顺反异构体,顺式指同种配位体处于相邻位置,一般用“顺”或“cis-”表示;反式指同种配体处于对角位置,一般用“反”或“trans-”表示。每个手性碳原子可以基于立体化学而被定义为(R)-或(S)-。本发明旨在包括所有可能的异构体,及其外消旋体和光学纯形式。本发明的化合物的制备可以选择外消旋体、顺反异构体、手性异构体、非对映异构体或对映异构体作为原料或中间体。光学活性的异构体可以使用手性合成子或手性试剂来制备,或者使用常规技术进行拆分,例如采用结晶以及手性色谱等方法。
制备/分离个别光学异构体(也即,顺反异构体和手性异构体)的常规技术包括由合适的顺反前体或光学纯前体的手性合成,或者使用例如手性高效液相色谱法拆分消旋体(或盐或衍生物的消旋体),例如可参见Gerald Gübitz and Martin G.Schmid (Eds.),Chiral Separations,Methods and Protocols,Methods in Molecular Biology,Vol.243,2004;A.M.Stalcup,Chiral Separations,Annu.Rev.Anal.Chem.3:341-63,2010;Fumiss et al.(eds.),VOGEL’S ENCYCLOPEDIA OF PRACTICAL ORGANIC CHEMISTRY 5.sup.TH ED.,Longman Scientific and Technical Ltd.,Essex,1991,809-816;Heller,Acc.Chem.Res.1990,23,128。
本发明还包括同位素标记的化合物(也即,同位素衍生物),等同于原始化合物在此公开。不过实际上对一个或多个的原子被与其原子量或质量序数不同的原子取代通常会出现。本发明的同位素衍生物中的同位素的例子包括氢、碳、氮、氧、磷、硫、氟和氯同位素,分别如 2H、 3H、 13C、 11C、 14C、 15N、 18O、 17O、 31P、 32P、 35S、 18F和 36Cl。本发明的化合物的同位素衍生物都在本发明的保护范围之内。本文中, 3H标记的化合物和 14C标记的化合物,在药物和底物的组织分布实验中是有用的。氚(即 3H)和碳-14(即 14C)标记的化合物的制备和检测比较容易,是同位素中的首选。此外,较重同位素取代如氘,即 2H,由于其很好的代谢稳定性在某些疗法中有优势,例如在体内增加半衰期或减少用量,因此,在某些情况下可以优先考虑。同位素标记的化合物可以用一般的方法,通过用易得的同位素标记试剂替换为非同位素的试剂,用披露在示例中的方案可以制备。
本文中,术语“药学上可接受的盐”包括药学上可接受的酸加成盐和药学上可接受的碱加成盐。
“药学上可接受的酸加成盐”是指能够保留游离碱的生物有效性而无其它副作用的,与无机酸或有机酸所形成的盐。无机酸盐包括但不限于盐酸盐、氢溴酸盐、硫酸盐、硝酸盐、磷酸盐等;有机酸盐包括但不限于甲酸盐、乙酸盐、2,2-二氯乙酸盐、三氟乙酸盐、丙酸盐、己酸盐、辛酸盐、癸酸盐、十一碳烯酸盐、乙醇酸盐、葡糖酸盐、乳酸盐、癸二酸盐、己二酸盐、戊二酸盐、丙二酸盐、草酸盐、马来酸盐、琥珀酸盐、富马酸盐、酒石酸盐、柠檬酸盐、棕榈酸盐、硬脂酸盐、油酸盐、肉桂酸盐、月桂酸盐、苹果酸盐、谷氨酸盐、焦谷氨酸盐、天冬氨酸盐、苯甲酸盐、甲磺酸盐、苯磺酸盐、对甲苯磺酸盐、海藻酸盐、抗坏血酸盐、水杨酸盐、4-氨基水杨酸盐、萘二磺酸盐等。这些盐可通过本专业已知的方法制备。
“药学上可接受的碱加成盐”是指能够保持游离酸的生物有效性而无其它副作用的、与无机碱或有机碱所形成的盐。衍生自无机碱的盐包括但不限于钠盐、钾盐、锂盐、铵盐、钙盐、镁盐、铁盐、锌盐、铜盐、锰盐、铝盐等。优选的无机盐为铵盐、钠盐、钾盐、钙盐及镁盐。衍生自有机碱的盐包括但不限于以下的盐:伯胺类、仲胺类及叔胺类,被取代的胺类,包括天然的被取代胺类、环状胺类及碱性离子交换树脂,例如氨、异丙胺、三甲胺、二乙胺、三乙胺、三丙胺、乙醇胺、二乙醇胺、三乙醇胺、二甲基乙醇胺、2-二甲氨基乙醇、2-二乙氨基乙醇、二环己胺、赖氨酸、精氨酸、组氨酸、咖啡因、普鲁卡因、胆碱、甜菜碱、乙二胺、葡萄糖胺、甲基葡萄糖胺、可可碱、嘌呤、哌嗪、哌啶、N-乙基哌啶、聚胺树脂等。优选的有机碱包括异丙胺、二乙胺、乙醇胺、三甲胺、二环己基胺、胆碱及咖啡因。这些盐可通过本专业已知的方法制备。
如果要设计一个本发明的化合物特定的立体异构体的合成,它可以不对称合成制备,或用手性辅剂衍生化,将所产生的立体混合物分离,再除去手性辅剂而得到 纯的顺反单体,手性单体或混合的立体单体。如果分子中含顺反异构中心,可通过柱层析纯化(正相硅胶柱或反相高效液相制备)得到纯的顺式产物或反式产物;另外,如果分子中含有一个碱性官能团,如氨基酸,或酸性官能团,如羧基,可以用合适的光学活性的酸或碱的与之形成非对映异构体盐,再通过分离结晶或色谱等常规手段分离,然后就得到了纯的对映体。
如本文所述,本发明的化合物可与任何数量取代基或官能团取而扩大其包涵范围。通常,术语“取代”是指用指定结构取代基,代替氢自由基。当特定结构中的多个位置被多个特定的取代基取代时,取代基每一个位置可以是相同或不同。本文中所使用的术语“取代”包括所有允许有机化合物取代。从广义上讲,允许的取代基包括非环状的、环状的、支链的非支链的、碳环的和杂环的,芳环的和非芳环的有机化合物。本文中,如杂原子氮可以有氢取代基或任何允许的上文所述的有机化合物来补充其价态。此外,本发明是无意以任何方式限制允许取代有机化合物。本发明认为取代基和可变基团的组合以稳定化合物形式在疾病的治疗上是很好的。此处术语“稳定”是指具有稳定的化合物,在足够长的时间内检测足以维持化合物结构的完整性,最好是在足够长的时间内都在效,本文在此用于上述目的。
式I所示化合物及其药学可接受的盐的代谢产物,以及可以在体内转变为式I所示化合物及其药学可接受的盐的前药,也包含在本发明的保护范围之内。
化合物的制备方法
以下方案中描述了制备式I所示化合物的方法。在某些情况下,可以改变执行反应方案的步骤的顺序,以促进反应或避免不需要的副反应产物。本发明的化合物还可以任选将在本说明书中描述的或本领域已知的各种合成方法组合起来而方便的制得,这样的组合可由本发明所属领域的技术人员容易的进行。
通常,在制备流程中,各反应通常在惰性溶剂中,在室温至回流温度(如0℃-150℃,优选10℃-100℃)下进行。反应时间通常为0.1小时-60小时,较佳地为0.5-48小时。
优选地,本发明的化合物的制备包括步骤:
Figure PCTCN2022128411-appb-000081
式中,W 1、W 2、环A 1、环A 2、环A 3、环A 4、环A 5、环A 6、Z 1、Z 2、Z 3、Z 4、Z 8、AL 1、AL 2、AL 3、R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、n 1、n 2、n 3、n 4、n 5、n 6和n 7的定义如上所述;
s1)在合适的碱性条件下(如但不限于碳酸铯等),选择合适的溶剂(如但不限于DMF等),在合适的温度下(如但不限于75℃),化合物(I-1)与化合物(I-2)(如但不限于含OMs,Cl,Br等的环状化合物)反应,得到化合物(I-3);
s2)在合适的条件下(如但不限于还原胺化反应和取代反应),化合物(I-3)与化合物(I-4)反应,得到化合物(I-5);
s3)在合适的条件下(如但不限于还原胺化反应,取代反应和酰胺缩合反应),化合物(I-5)与化合物(I-6)反应,得到化合物(I-7);
s4)在合适的条件下(如但不限于还原胺化反应,取代反应和酰胺缩合反应),化合物(I-7)与化合物(I-8)反应,得到化合物(I)。
以上各反应步骤中,反应溶剂、反应温度、反应时间、催化剂等可以根据具体的反应物进行选择。
原料和中间体从商业来源购买,或由已知步骤制备,或使用本领域熟知的方法制备。
药物组合物和施用方法
由于本发明化合物具有优异的BTK激酶抑制活性,因此本发明化合物及其各种晶型,药学上可接受的无机或有机盐,水合物或溶剂合物,以及含有本发明化合物为主要活性成分的药物组合物可用于预防和/或治疗(稳定、减轻或治愈)BTK激酶相关疾病(自体免疫疾病、炎性疾病或肿瘤等)。
本发明的药物组合物包含安全有效量范围内的本发明化合物及药学上可以接受的赋形剂或载体。其中“安全有效量”指的是:化合物的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有1-2000mg本发明化合物/剂,更佳地,含有10-200mg本发明化合物/剂。较佳地,所述的“一剂”为一个胶囊或药片。
“药学上可接受的载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份能和本发明化合物以及它们之间相互掺和,而不明显降低化合物的药效。药学上可以接受的载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如
Figure PCTCN2022128411-appb-000082
)、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。
本发明化合物或药物组合物的施用方式没有特别限制,代表性的施用方式包括(但并不限于):口服、肠胃外(静脉内、肌肉内或皮下)。
用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。在这些固体剂型中,活性化合物与至少一种常规惰性赋形剂(或载体)混合,如柠檬酸钠或磷酸二钙,或与下述成分混合:(a)填料或增容剂,例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸;(b)粘合剂,例如,羟甲基纤维素、藻酸盐、明胶、聚乙烯基吡咯烷酮、蔗糖和阿拉伯胶;(c)保湿剂,例如,甘油;(d)崩解剂,例如,琼脂、碳酸钙、马铃薯淀粉或木薯淀粉、藻酸、某些复合硅酸盐、和碳酸钠;(e)缓溶剂,例如石蜡;(f)吸收加速剂,例如,季胺化合物;(g)润湿剂,例如鲸蜡醇和单硬脂酸甘油酯;(h)吸附剂,例如,高岭土;和(i)润滑剂,例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、十二烷基硫酸钠,或其混合物。胶囊剂、片剂和丸剂中,剂型也可包含缓冲剂。
固体剂型如片剂、糖丸、胶囊剂、丸剂和颗粒剂可采用包衣和壳材制备,如肠衣和其它本领域公知的材料。它们可包含不透明剂,并且,这种组合物中活性化合物或化合物的释放可以延迟的方式在消化道内的某一部分中释放。可采用的包埋组分的实例是聚合物质和蜡类物质。必要时,活性化合物也可与上述赋形剂中的一种或多种形成微胶囊形式。
用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。除了活性化合物外,液体剂型可包含本领域中常规采用的惰性稀释剂,如水或其它溶剂,增溶剂和乳化剂,例知,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、丙二醇、1,3-丁二醇、二甲基甲酰胺以及油,特别是棉籽油、花生油、玉米胚油、橄榄油、蓖麻油和芝麻油或这些物质的混合物等。
除了这些惰性稀释剂外,组合物也可包含助剂,如润湿剂、乳化剂和悬浮剂、甜味剂、矫味剂和香料。
除了活性化合物外,悬浮液可包含悬浮剂,例如,乙氧基化异十八烷醇、聚氧乙烯山梨醇和脱水山梨醇酯、微晶纤维素、甲醇铝和琼脂或这些物质的混合物等。
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、分散液、悬浮液或乳液,和用于重新溶解成无菌的可注射溶液或分散液的无菌粉末。适宜的含水和非水载体、稀释剂、溶剂或赋形剂包括水、乙醇、多元醇及其适宜的混合物。
本发明化合物可以单独给药,或者与其他药学上可接受的化合物(例如抗-HBV剂)联合给药。
联合给药时,所述药物组合物还包括与一种或多种(2种,3种,4种,或更多种)其他药学上可接受的化合物。该其他药学上可接受的化合物中的一种或多种(2种,3种,4种,或更多种)可与本发明的化合物同时、分开或顺序地用于预防和/或治疗BTK,尤其是BTK突变的相关疾病。
使用药物组合物时,是将安全有效量的本发明化合物适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1-2000mg,优选20-500mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。
本发明的主要优点在于:
1.本发明的化合物结构新颖且具有优异的BTK及BTK(C481S)激酶抑制剂作用;
2.本发明的化合物药效好、生物利用度好、更安全、能抑制并降解BTK及BTK(C481S)蛋白。
3.本发明的化合物含独特的E3泛素连接酶配体,能减弱免疫调节性亚胺药物(IMiD)引起的底物降解作用,从而降低IMiD活性。
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。
本文中,所有的温度均为℃,除非另有说明。
本文中,对收率而言的百分比均为质量百分比。
本文中,所有的份数均为体积份,所有的百分比均为体积百分比,除非另有说明。
本文中,PTLC或TLC(薄层色谱)的制备是在20×20cm的板(500微米厚的硅胶)上进行;硅胶层析法用Biotage快速色谱系统。
本文中, 1H NMR(氢谱)采用Bruker AscendTM400光谱仪,400MHz,298°K,并且将残余质子在氘化试剂中的化学位移(ppm)给出参考:CDCl 3的δ(化学位移)为7.26ppm,CD 3OD的δ为3.31ppm,DMSO-d6的δ为2.50ppm。
本文中,LCMS(液相色谱质谱联用)测试中,液相色谱采用安捷伦科技1200系列或6120四极谱仪;对于液相色谱,流动相为乙腈(A)和水(B)和0.01%甲酸,洗脱剂梯度:6.0分钟5-95%A,5.0分钟60-95%A,5.0分钟80-100%A和10分钟85-100%A,采用SBC1850毫米×4.6毫米×2.7微米的毛细管柱;质谱(MS)通过电喷雾离子质谱法(ESI)进行测定。
本文中,高效液相色谱(HPLC)-质谱(MS)分析条件:
LC1柱:SB-C18 50mm×4.6mm×2.7μm;
温度:50℃;
洗脱液:5:95至95:5的乙腈/水(上述比值为体积比)+0.01%甲酸,6分钟;
流量:1.5mL/min,注射5μL;
检测:PDA检测器,200-600nm;
MS:质量范围150-750amu;正离子电喷雾电离。
LC2柱:SB-C18 50mm×4.6mm×2.7μm;
温度:50℃;
洗脱液:5:95至95:5的乙腈/水(上述比值为体积比)+0.05%TFA(三氟乙酸)梯度,超过3.00分钟;
流量:1.5mL/min,注射5μL;
检测:PDA检测器,200-600nm;
MS:质量范围150-750amu;正离子电喷雾电离。
LC3柱:SB-C18 50mm×4.6mm×2.7μm;
温度:50℃;
洗脱液:10:90至98:2的乙腈/水(上述比值为体积比)+0.05%TFA梯度,超过3.75分钟;
流速:1.0mL/min,注射10μL;
检测:PDA检测器,200-600nm;
MS:质量范围150-750amu;正离子电喷雾电离。
本文中,各缩写所代表的意思如下所示:
AcOH=乙酸;Alk为烷基;AR为芳基;Boc=叔丁氧羰基;CH 2Cl 2=二氯甲烷; DBU=1,8-二氮杂双环[5.4.0]十一-7-烯;DCM=二氯甲烷;DEAD=偶氮二甲酸二乙酯;DMF=N,N-二甲基甲酰胺;DMSO=二甲基亚砜;EA=乙酸乙酯;Et=乙基;EtOAc=乙酸乙酯;EtOH=乙醇;HOAc=乙酸;LiOH=氢氧化锂;Me=甲基;MeCN=乙腈;MeOH=甲醇;MgSO 4=硫酸镁;NaCl=氯化钠;NaOH=氢氧化钠;Na 2SO 4=硫酸钠;PE=石油醚;Ph=苯基;PG=保护基;TFA=三氟乙酸;THF=四氢呋喃;Ts=对甲苯磺酰基;
rt=室温;h=小时;min=分钟;bs=宽峰;s=单峰;d=双峰;dd=双二重峰;t=三重峰;m=多重。
实施例1 5-(3-(6-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-2-氮杂螺环[3.3]庚烷-2-基)氮杂丁烷-1-基)-2-(2,6-二氧哌啶-3-基)异吲哚啉-1,3-二酮
Figure PCTCN2022128411-appb-000083
合成路线:
Figure PCTCN2022128411-appb-000084
步骤1:6-(甲苯磺酰氧基)-2-氮杂螺[3.3]庚烷-2-羧酸叔丁酯
6-羟基-2-氮杂螺[3.3]庚烷-2-羧酸叔丁酯(200mg)加入二氯甲烷(5mL)中,再加入三乙胺(190mg)和DMAP(23mg),室温搅拌5分钟,再加入对甲苯磺酰氯(215mg),室温搅拌16小时。TLC确认反应完全后,加入二氯甲烷和去离子水,萃取分层,收集有机层,浓缩二氯甲烷,残余物经柱层析纯化,得到白色固体6-(甲苯磺酰氧基)-2-氮杂螺[3.3]庚烷-2-羧酸叔丁酯(344mg,收率100%)。 1H NMR(400MHz,CDCl 3)δ7.79(d,J=8.3Hz,2H),7.36(d,J=8.3Hz,2H),4.71(p,J=7.2Hz,1H),3.86(s,4H),2.55–2.48(m,2H),2.48(s,3H),2.36–2.29(m,2H),1.43(s,9H)。
步骤2:6-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-2-氮杂螺[3.3]庚烷-2-羧酸叔丁酯
6-(甲苯磺酰氧基)-2-氮杂螺[3.3]庚烷-2-羧酸叔丁酯(344mg)、3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(284mg)和碳酸铯(611mg)加入N,N-二甲基甲酰胺(5mL)中,升温至60℃,搅拌反应16小时。LCMS确认反应完全。加入乙酸乙酯和去离子水,萃取分层,收集有机层,无水硫酸钠干燥,浓缩,残余物经柱层析纯化,得到类白色固体6-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-2-氮杂螺[3.3]庚烷-2-羧酸叔丁酯(410mg,收率87.8%)。 1H NMR(400MHz,CDCl 3)δ8.37(s,1H),7.67(d,J=8.4Hz,2H),7.42(t,J=7.8Hz,2H),7.20(dd,J=12.3,7.9Hz,3H),7.11(d,J=8.5Hz,2H),5.81(s,2H),5.41–5.28(m,1H),4.17–4.08(m,2H),4.03(s,2H),3.07–2.99(m,2H),2.82–2.71(m,2H),1.47(s,9H)。
步骤3:3-(4-苯氧基苯基)-1-(2-氮杂螺[3.3]庚烷-6-基)-1H-吡唑并[3,4-嘧啶-4-胺
6-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-2-氮杂螺[3.3]庚烷-2-羧酸叔丁酯(200mg)加入甲酸(3mL)中,室温搅拌1小时后,LCMS确认反应完全。浓缩甲酸,加入饱和碳酸氢钠水溶液和乙酸乙酯,萃取分层,收集有机层,无水硫酸钠干燥,浓缩乙酸乙酯,得到黄色油状物3-(4-苯氧基苯基)-1-(2-氮杂螺[3.3]庚烷-6-基)-1H-吡唑并[3,4-嘧啶-4-胺(95mg,收率59.4%)。 1H NMR(400MHz,CDCl 3)δ11.76(s,1H),9.83(s,2H),8.25(s,1H),7.59(d,J=8.7Hz,2H),7.49–7.41(m,2H),7.25(t,J=7.4Hz,1H),7.20(d,J=8.7Hz,1H),7.13(dd,J=8.6,1.0Hz,2H),6.29(s,1H),5.42–5.32(m,1H),4.26(s,4H),3.13–2.96(m,4H)。
步骤4:3-(6-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-2-氮杂螺[3.3]庚烷-2-基)氮杂环丁烷-1-羧酸叔丁酯
3-(4-苯氧基苯基)-1-(2-氮杂螺[3.3]庚烷-6-基)-1H-吡唑并[3,4-嘧啶-4-胺(95mg)和3-N-Boc-氮杂环丁酮(49mg)和乙酸(76mg)加入1,2-二氯乙烷(5mL)中,65℃搅拌3h,降温至室温后,再加入三乙酰氧基硼氢化钠(101mg,0.48mmol),室温搅拌16h。LCMS确认反应完全。加入饱和碳酸氢钠水溶液和二氯甲烷,萃取分层,收集有机层,无水硫酸钠干燥,浓缩二氯甲烷,残余物经柱层析纯化得到黄色油状物3-(6-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-2-氮杂螺[3.3]庚烷-2-基)氮杂环丁烷-1-羧酸叔丁酯(89mg,收率67.4%)。
步骤5:1-(2-(氮杂环丁烷-3-基)-2-氮杂螺[3.3]庚烷-6-基)-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
3-(6-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-2-氮杂螺[3.3]庚烷-2-基)氮杂环丁烷-1-羧酸叔丁酯(59mg)加入甲酸(3mL)中,室温搅拌1h后,LCMS确认反应完全。浓缩甲酸,加入饱和碳酸氢钠水溶液和二氯甲烷,萃取分层,收集有机层,无水硫酸钠干燥,浓缩二氯甲烷,得到浅黄色油状物1-(2-(氮杂环丁烷-3-基)-2-氮杂螺[3.3]庚烷-6-基)-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(20mg,收率41.4%)。
步骤6:5-(3-(6-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-2-氮杂螺[3.3]庚烷-2-基)氮杂环丁烷-1-基)-2-(2,6-二氧哌啶-3-基)异二氢吲哚-1,3-二酮
1-(2-(氮杂环丁烷-3-基)-2-氮杂螺[3.3]庚烷-6-基)-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(A2-6,20mg,0.04mmol)、2-(2,6-二氧哌啶-3-基)-5-氟异吲哚啉-1,3-二酮(12mg,0.04mmol)、N,N-二异丙基乙胺(15.5mg,0.12mmol)加入DMSO中,80℃搅拌1h,LCMS显示几乎反应完全。反应液经半制备纯化,冻干后,得到黄色固体状的5-(3-(6-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-2-氮杂螺环[3.3]庚烷-2-基)氮杂丁烷-1-基)-2-(2,6-二氧哌啶-3-基)异吲哚啉-1,3-二酮(5mg,收率16%)。LCMS(ESI):[M+H] +=710.4;HPLC:纯度96%. 1H NMR(400MHz,DMSO)δ11.08(s,1H),10.94(s,1H),8.29(s,1H),7.70(dd,J=17.9,8.5Hz,2H),7.45(dd,J=8.5,7.5Hz,2H),7.32–7.11(m,4H),6.94(d,J=1.9Hz,1H),6.78(dd,J=8.3,2.0Hz,1H),5.35(dt,J=15.9,8.1Hz,1H),5.08(dd,J=12.9,5.4Hz,1H),4.42(d,J=28.3Hz,3H),4.33–4.22(m,3H),4.07(d,J=9.4Hz,3H),3.01–2.81(m,4H),2.60(dd,J=34.6,17.3Hz,2H),2.08–1.95(m,1H)
参考合成化合物1的方法,使用合成化合物1类似的反应,制备了以下化合物:
实施例2 5-(3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)-(1,3':1',3”-三唑烷基)-1”-基)-2-(2,6-二氧哌啶-3-基)异吲哚啉-1,3-二酮
Figure PCTCN2022128411-appb-000085
黄色固体(2.10mg)。LCMS(ESI):[M+H] +=725.5;HPLC:纯度99%. 1H NMR(400MHz,DMSO-d 6)δppm 11.08(s,1H),8.31(s,1H),7.73-7.68(m,3H),7.48-7.44(m,2H),7.23-7.12(m,5H),6.88(m,1H),6.74-6.71(m,1H),5.82(br s,2H),5.10-5.05(m,2H),4.40-3.10(m,10H),2.94-2.84(m,2H),2.64-2.50(m,4H),2.04-2.01(m,2H).
实施例3 5-(3-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己烷-1-羰基)哌嗪-1-基)氮杂环丁烷-1-基)-2-(2,6-二氧哌啶-3-基)异吲哚啉-1,3-二酮
Figure PCTCN2022128411-appb-000086
黄色固体(2.13mg)。LCMS(ESI):[M+H] +=809.4;HPLC:纯度95%. 1H NMR(400MHz,DMSO-d 6)δppm 11.10(s,1H),8.35-8.26(s,1H),7.75-7.55(m,3H), 7.50-7.44(m,2H),7.25-7.02(m,5H),6.89(m,1H),6.75-6.71(m,1H),5.41-5.29(m,1H),5.20-5.03(m,1H),4.90-4.75(m,2H),4.40-4.10(m,4H),3.34-2.74(m,4H),2.70-2.50(m,5H),2.50-2.25(m,4H),2.10-1.60(m,8H).
实施例4 1-(4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)哌啶-1-羰基)环丁基)(甲基)氨基)甲基)哌啶-1-羰基)苯基)二氢嘧啶-2,4(1H,3H)-二酮
Figure PCTCN2022128411-appb-000087
白色固体(5.50mg)。LCMS(ESI):[M+H] +=811.7;HPLC:纯度99%. 1H NMR(400MHz,DMSO-d 6)δppm 10.50(s,1H),8.38(s,1H),7.75-7.55(m,2H),7.50-7.25(m,5H),7.23-7.02(m,6H),5.55-4.90(m,2H),4.61-4.29(m,2H),4.00-2.20(m,24H),2.18-1.90(m,8H).
实施例5 5-(3-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-2-(2,6-二氧哌啶-3-基)异吲哚啉-1,3-二酮
Figure PCTCN2022128411-appb-000088
黄色固体(0.4mg)。LCMS(ESI):[M+H] +=781.5;HPLC:纯度96%. 1H NMR(400MHz,DMSO)δ11.07(s,1H),8.24(s,1H),7.66(d,J=8.0Hz,3H),7.44(t,J=8.0Hz,2H),7.23–7.09(m,4H),6.80(s,1H),6.66(d,J=8.0Hz,1H),5.06(m,1H),4.66(m,1H),4.10(m,2H),3.86(m,2H),3.29(m,1H),2.87(m,1H),2.70-2.25(m,8H),2.04(m,6H),1.51(m,2H)。
实施例6 6-(3-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)哌啶-1-基)双环[1.1.1]戊烷-1-羰基)-2-(2,6-二氧哌啶-3-基)-6,7-二氢吡咯[3,4-f]异吲哚-1,3(2H,5H)-二酮
Figure PCTCN2022128411-appb-000089
白色固体(0.7mg)。LCMS(ESI):[M+H] +=778.4;HPLC:纯度98%.
实施例7 5-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)六氢环戊烷[c]吡咯-2(1H)-基)-1,3'-二氮杂丁烷-1'-基)-2-(2,6-二氧哌啶-3-基)异吲哚啉-1,3-二酮
Figure PCTCN2022128411-appb-000090
黄色粉末(2.35mg)。LCMS(ESI):[M+H] +=779.5;HPLC:纯度99%. 1H NMR(400MHz,DMSO-d 6)δppm 11.08(s,1H),8.31(s,1H),7.73-7.68(m,3H),7.48-7.44(m,2H),7.23-7.12(m,5H),6.88(m,1H),6.74-6.71(m,1H),5.60-5.40(m,2H),5.20-5.05(m,2H),4.40-3.50(m,10H),3.24-2.84(m,5H),2.64-2.30(m,5H),2.14-1.91(m,4H).
实施例8 5-(3-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)氮杂环-1-基)-1,3'-二氮杂环丙烷-1'-基)-2-(2,6-二氧哌啶-3-基)异吲哚啉-1,3-二酮
Figure PCTCN2022128411-appb-000091
黄色固体(2.06mg)。LCMS(ESI):[M+H] +=767.5;HPLC:纯度96%. 1H NMR(400MHz,DMSO-d 6)δppm 11.06(s,1H),8.24(s,1H),7.75-7.60(m,3H),7.48-7.44(m,2H),7.23-7.12(m,5H),6.80(m,1H),6.74-6.65(m,1H),5.10-4.90(m,2H),4.07-4.03(m,2H),3.83-3.80(m,2H),3.65(br s,1H),3.44(br s,2H),3.20(br s,1H),3.00-2.80(m,3H),2.64-2.50(m,3H),2.22-2.16(m,2H),2.15-1.87(m,5H),1.75-1.60(m,1H).
实施例9 2-(2,6-二氧哌啶-3-基)-6-(1-(4-(3-(4-(2-氟-3-甲氧基苯氧基)苯基)-7-甲氧基-1H-吡唑并[4,3-c]吡啶-1-基)环己烷-1-羰基)氮杂丁-3-基)-6,7-二氢吡咯[3,4-f]异吲哚-1,3(2H,5H)-二酮
Figure PCTCN2022128411-appb-000092
黄色固体(8.00mg)。LCMS(ESI):[M+H] +=828.60;1H NMR(400MHz,DMSO-d6)δ11.13(s,1H),9.36(s,1H),8.25(s,1H),8.11–8.00(m,2H),7.91(s,2H),7.23–7.08(m,4H),6.85(m,,1H),5.15(m,2H),4.41(m,6H),4.18(s,3H),4.08(d,J=7.8Hz,2H),3.97(d,J=7.8Hz,2H),2.98–2.85(m,1H),2.70–2.55(m,2H),2.47–2.30(m,2H),2.11(m,6H),1.90(d,J=13.5Hz,2H),1.62(d,J=13.8Hz,2H)。
实施例10 3-((4-(3-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)哌啶-1-基)-(1,3'-二氮杂氮基)-1'-基)-3-氟苯基)氨基)哌啶-2,6-二酮
Figure PCTCN2022128411-appb-000093
黄色固体(1.25mg)。LCMS(ESI):[M+H] +=717.4;HPLC:纯度94%.
实施例11 6-(3-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)哌啶-1-基)环丁烷-1-羰基)-2-(2,6-二氧哌啶-3-基)-6,7-二氢吡咯[3,4-f]异吲哚-1,3(2H,5H)-二酮
Figure PCTCN2022128411-appb-000094
类白色固体(5.9mg)。LCMS(ESI):[M+H] +=766.7;HPLC:纯度97%. 1H NMR(400MHz,DMSO-d6)δppm 11.13(s,1H),9.00(s,1H),9.75-9.30(m,2H),8.32(s,1H),8.00-7.90(m,2H),7.73-7.60(m,2H),7.51-7.40(m,2H),7.25-7.10(m,5H),5.20-4.60(m,6H),4.20(s,1H),3.80-3.50(m,3H),3.25-3.10(m,3H),3.00-2.85(m,1H),2.70-2.00(m, 8H).
实施例12 5-(3-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)哌啶-1-基)-1,3'-二氮杂丁烷-1'-基)-N-(2,6-二氧哌啶-3-基)吡啶酰胺
Figure PCTCN2022128411-appb-000095
类白色固体(5.9mg)。LCMS(ESI):[M+H] +=728.0;HPLC:纯度95%. 1H NMR(400MHz,DMSO-d6)δppm 10.80(s,1H),8.70(d,J=8.0Hz,1H),8.32(s,1H),7.95-7.80(m,2H),7.67(d,J=8.0Hz,2H),7.44(t,J=8.0Hz,2H),7.30-6.85(m,6H),5.20-3.50(m,15H),3.25-3.10(m,3H),3.40-3.30(m,2H),2.80-1.90(m,8H).
实施例13 5-(1'-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)-[3,3'-二氮杂环丁烷]-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮
Figure PCTCN2022128411-appb-000096
黄色固体(51.3mg)。LCMS(ESI):[M+H] +=752.5;HPLC:纯度99.3%. 1H NMR(400MHz,DMSO-d6)δppm 11.07(s,1H),8.31(d,J=3.0Hz,1H),7.71-7.63(m,3H),7.45(t,J=7.8Hz,2H),7.23-7.12(m,5H),6.80(d,J=2.0Hz,1H),6.70-6.64(m,1H),5.06(dd,J=12.9,5.3Hz,1H),4.97-4.83(m,1H),4.79-4.67(m,1H),4.35-4.05(m,5H),3.98(d,J=8.2Hz,2H),3.83(d,J=5.5Hz,2H),3.53-3.31(m,1H),3.28-3.12(m,1H),3.06(s,1H),2.89(s,1H),2.70-2.54(m,1H),2.32-2.19(m,1H),2.17-1.78(m,8H),1.52-1.35(m,1H).
实施例14 5-(3-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)六氢环戊[c]吡咯-2(1H)-基)氮杂环丁-1-基)吡咯烷-1-基)-N-(2,6-二氧哌啶-3-基)吡啶酰胺
Figure PCTCN2022128411-appb-000097
类白色固体(11.9mg)。LCMS(ESI):[M+H] +=768.7;HPLC:纯度93%. 1H NMR(400MHz,DMSO-d6)δppm 10.80(s,1H),8.70(d,J=8.0Hz,1H),8.31(s,1H),8.00-7.80(m,2H),7.70(d,J=8.0Hz,2H),7.45(t,J=8.0Hz,2H),7.30-7.00(m,5H),5.60-5.40(m,1H),4.75(m,2H),4.10-1.90(m,25H).
实施例15 3-((4-(3-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)六氢环戊[c]吡咯-2(1H)-基)氮杂环丁-1-基)吡咯烷-1-基)-3-氟苯基)氨基哌啶-2,6-二酮
Figure PCTCN2022128411-appb-000098
类白色固体(0.9mg)。LCMS(ESI):[M+H] +=757.7;HPLC:纯度85%.
实施例16 5-(3-(3-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)六氢环戊烷[c]吡咯-2(1H))-基)氮杂环丁烷-1-基)甲基)吡咯烷-1-基)-N-(2,6-二氧代哌啶-3-基)吡啶酰胺
Figure PCTCN2022128411-appb-000099
类白色固体(11.9mg)。LCMS(ESI):[M+H] +=782.8;HPLC:纯度96%. 1H NMR(400MHz,DMSO-d6)δppm 10.84(s,1H),8.63(d,J=8.0Hz,1H),8.25(s,1H),7.92(s,1H),7.84(d,J=8.0Hz,1H),7.67(d,J=8.0Hz,2H),7.44(t,J=8.0Hz,2H),7.30-7.10(m,5H),6.98(m,1H),5.38(m,1H),4.74(m,1H),3.60-3.10(m,8H),3.10-2.80(m,4H),2.80-2.60(m,3H),2.40-2.25(m,5H),2.25-2.00(m,4H),1.91(m,2H),1.88(m,1H).
实施例17 5-(3-(5-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)六氢环戊[c]吡咯-2(1H)-基)-(1,3'-二氮杂丁烷基)-1'-基)-2-(2,6-二氧哌啶-3-基)-6-氟异吲哚啉-1,3-二酮
Figure PCTCN2022128411-appb-000100
类白色固体(20.1mg)。LCMS(ESI):[M+H] +=844.9;HPLC:纯度96%. 1H NMR(400MHz,DMSO-d6)δppm 11.10(s,1H),8.25(s,1H),7.70-7.55(m,3H),7.25-7.00(m,4H),6.90(d,J=8.0Hz,1H),6.80(t,J=8.0Hz,1H),5.40(m,1H),5.10(m,1H),4.25-4.20(m,2H),3.90(m,2H),3.80(s,3H),3.60(m,1H),3.40(m,2H),3.20(m,2H),3.00-2.75(m,3H),2.70-2.30(m,6H),2.10-1.90(m,4H).
实施例18 5-(3-((5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)六氢环戊[c]吡咯-2(1H)-基)甲基)吡咯烷-1-基)-N-(2,6-二氧哌啶-3-基)吡啶酰胺
Figure PCTCN2022128411-appb-000101
类白色固体(0.9mg)。LCMS(ESI):[M+H] +=727.2;HPLC:纯度91%.
实施例19 5-(3-((4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)甲基)吡咯烷-1-基)-2-(2,6-二氧哌啶-3-基)异吲哚-1,3-二酮
Figure PCTCN2022128411-appb-000102
类白色固体(20.9mg)。LCMS(ESI):[M+H] +=809.1;HPLC:纯度93%. 1H NMR(400MHz,DMSO-d6)δppm 11.10(s,1H),8.25(s,1H),7.70-7.55(m,3H),7.50-7.40(m,2H),7.25-7.00(m,5H),6.90(m,1H),6.80(m,1H),5.10(m,1H),4.75(m,1H),3.75-2.50 (m,16H),2.25-1.90(m,10H),1.75(m,3H),1.48(m,2H).
实施例20 5-(3-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己烷-1-羰基)哌嗪-1-基)氮杂丁-1-基)-N-(2,6-二氧哌啶-3-基)吡啶酰胺
Figure PCTCN2022128411-appb-000103
类白色固体(25.1mg)。LCMS(ESI):[M+H] +=832.2;HPLC:纯度95%. 1H NMR(400MHz,DMSO-d6)δppm 10.85(s,1H),8.73(d,J=8.0H Z,1H),8.40(s,1H),7.89(m,2H),7.65(d,J=8.0Hz,2H),7.20-7.17(m,3H),7.15(t,J=8.0H Z,1H),7.02(d,J=8.0H Z,1H),6.84(t,J=8.0Hz,1H),4.80(m,1H),4.70(m,1H),4.50-3.50(m,9H),3.20(m,4H),3.00(m,1H),2.75(m,2H),2.30(m,2H),2.20(m,2H),2.00(m,3H),1.80(m,2H),1.75(m,2H).
实施例21 N-(2,6-二氧哌啶-3-基)-5-(3-(3-(4-(2-氟-3-甲氧基苯氧基)苯基)-7-甲氧基-1H-吡唑并[4,3-c]吡啶-1-基)哌啶-1-基)-(1,3'-二氮杂丁]-1'-基)吡啶酰胺
Figure PCTCN2022128411-appb-000104
类白色固体(21.9mg)。LCMS(ESI):[M+H] +=790.5;HPLC:纯度96%. 1H NMR(400MHz,DMSO-d6)δppm 10.83(s,1H),9.00(s,1H),8.68(d,J=8.2Hz,1H),8.10(s,1H),8.02(d,J=8.7Hz,2H),7.83(dd,J=12.3,5.6Hz,2H),7.20(td,J=8.4,1.8Hz,1H),7.16–7.06(m,3H),6.91(dd,J=8.6,2.7Hz,1H),6.84(t,J=7.1Hz,1H),4.99(d,J=11.5Hz,1H),4.81–4.66(m,1H),4.06(s,3H),4.01(t,J=7.6Hz,2H),3.90(s,3H),3.79(dd,J=8.2,4.3Hz,2H),3.66(s,1H),3.44(s,2H),3.00(s,3H),2.89(d,J=7.6Hz,2H),2.83–2.73(m,1H),2.53(d,J=10.4Hz,1H),2.24–2.12(m,3H),2.01(m,5H).
实施例22 6-((3-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)哌啶-1-基)环丁基)甲基)-2-(2,6-二氧哌啶-3-基)-6,7-二氢吡咯[3,4-f]异吲哚-1,3(2H,5H)-二酮
Figure PCTCN2022128411-appb-000105
白色固体(0.9mg)。LCMS(ESI):[M+H] +=751.8;HPLC:纯度92%.
实施例23 5-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己基)甲基)哌嗪-1-基)-N-(2,6-二氧哌啶-3-基)吡啶酰胺
Figure PCTCN2022128411-appb-000106
类白色固体(0.9mg)。LCMS(ESI):[M+H] +=715.5;HPLC:纯度96%.
实施例24 N-(2,6-二氧代哌啶-3-基)-5-(5-(3-(4-(2-氟-3-甲氧基苯氧基)苯基)-7-甲氧基-1H-吡唑并[4,3-c]吡啶-1-基)六氢环戊烷-2(1H)基)-[1,3'-联氮杂环丁烷]-1'-基)吡啶-2(1H)基吡啶-1'-基)吡啶甲酰胺
Figure PCTCN2022128411-appb-000107
类白色固体(20.8mg)。LCMS(ESI):[M+H] +=816.5;HPLC:纯度95.6%. 1H NMR(400MHz,DMSO-d6)δppm 10.86(s,1H),8.99(s,1H),8.69(d,J=8.2Hz,1H),8.09(s,1H),8.01(d,J=8.7Hz,2H),7.85(d,J=8.6Hz,1H),7.81(d,J=2.3Hz,1H), 7.23-7.15(m,1H),7.15-7.04(m,3H),6.90(dd,J=8.6,2.5Hz,1H),6.82(t,J=7.6Hz,1H),5.82-5.68(m,1H),4.81-4.68(m,1H),4.06(s,3H),4.02(t,J=7.5Hz,2H),3.90(s,3H),3.79(dd,J=7.7,4.4Hz,2H),3.67(s,1H),3.60(t,J=6.1Hz,1H),3.34(s,1H),3.08(s,3H),2.88-2.74(m,3H),2.53(d,J=10.4Hz,1H),2.41-2.28(m,4H),2.18(ddd,J=26.3,13.3,4.6Hz,1H),2.06-1.88(m,3H),1.82-1.68(m,1H),1.30-1.22(m,1H).
实施例25 5-(3-(5-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑[3,4-d]嘧啶-1-基)六氢环戊[c]吡咯-2(1H)-基)-(1,3'-二氮杂丁烷]-1'-基)-N-(2,6-二氧哌啶-3-基)吡啶酰胺
Figure PCTCN2022128411-appb-000108
类白色固体(20.1mg)。LCMS(ESI):[M+H] +=801.7;HPLC:纯度96%. 1H NMR(400MHz,DMSO-d6)δppm 10.84(s,1H),8.68(d,J=8.0Hz,1H),8.25(s,1H),7.83(t,J=8.7Hz,2H),7.65(d,J=8.0Hz,2H),7.25-7.0(m,4H),6.91(d,J=8.0Hz,1H),6.84(t,J=8.0Hz,1H),5.50-5.30(m,1H),4.81-4.68(m,1H),4.06(s,3H),4.03(m,2H),3.90(s,3H),3.76(m,1H),3.48-3.30(m,3H),3.02(m,3H),2.75(m,3H),2.53(m,2H),2.40-2.25(m,4H),2.25(m,1H),2.03(m,1H),1.90(m,2H).
实施例26 5-(4-(1-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己基)氮杂环糊精-3-基)哌嗪-1-基)-N-(2,6-二氧哌啶-3-基)吡啶酰胺
Figure PCTCN2022128411-appb-000109
类白色固体(20.8mg)。LCMS(ESI):[M+H] +=755.8;HPLC:纯度93%. 1H NMR(400MHz,DMSO-d6)δppm 10.86(s,1H),8.74(d,J=8.0Hz,1H),8.37(s,2H),7.89(d,J=8.7Hz,1H),7.50-7.35(m,3H),7.25-7.10(m,5H),5.00-4.00(m,7H),3.51-3.30(m,6H),2.80-2.50(m,4H),2.40-1.80(m,12H).
实施例27 4-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)六氢环戊烷-2(1H)基)-[1,3'-联氮杂环丁烷]-1'-基)-N-(2,6-二氧代哌啶-3-基)-2-甲氧基苯甲酰胺
Figure PCTCN2022128411-appb-000110
类白色固体(20.7mg)。LCMS(ESI):[M+H] +=782.9;HPLC:纯度92%. 1H NMR(400MHz,DMSO-d6)δppm 10.86(s,1H),8.40(d,J=8.0Hz,1H),8.25(s,1H),8.22(m,1H),7.77(d,J=8.0Hz,1H),7.66(d,J=8.0Hz,2H),7.44(t,J=8.0Hz,2H),7.25-7.00(m,5H),6.10(m,1H),6.03(s,1H),5.41(m,1H),4.69(m,1H),3.97(m,2H),3.88(s,3H),3.80-3.30(m,6H),3.14(m,4H),2.90-2.50(m,4H),2.48-2.30(m,3H),2.25-1.90(m,5H).
实施例28 5-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)六氢环戊[c]吡咯-2(1H)-基)-(1,3'-二氮杂丁烷]-1'-基)-N-(2,6-二氧哌啶-3-基)吡啶酰胺
Figure PCTCN2022128411-appb-000111
合成路线:
Figure PCTCN2022128411-appb-000112
步骤1:5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)六氢环戊[c]吡咯-2(1H)-羧酸叔丁酯
4-甲基苯磺酸-2-{[(2-甲基丙-2-基)氧基]羰基}八氢吡咯并[3,4-a][5]轮烯-5-基酯(2.82g),3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(2.24g),碳酸铯(4.82g)加入N-N-二甲基甲酰胺(30mL)中,室温搅拌5分钟,升温到80℃反应搅拌4小时。LCMS确认反应物消失后,将反应液倒入水中,乙酸乙酯萃取,分液,有机相用无水硫酸钠干燥后过滤,浓缩得到粗品。粗品经反相半制备纯化得到白色固体5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)六氢环戊[c]吡咯-2(1H)-羧酸叔丁酯2.8g,(79%),LCMS(ESI):[M+H] +=512.9。
步骤2:1-(八氢环戊[c]吡咯-5-基)-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)六氢环戊[c]吡咯-2(1H)-羧酸叔丁酯(2.80g)加入二氯甲烷(10mL)中,然后加入三氟乙酸(2mL),室温搅拌3小时。TLC确认反应完全后,浓缩,得到类黄色固体(2.30g)。该固体直接用于下一步。
步骤3:1-(2-(氮杂环丁烷-1-羧酸叔丁酯-3-基)八氢环戊基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
1-(八氢环戊[c]吡咯-5-基)-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺加入二氯乙烷(20mL)中,加入三乙胺(1.12g).室温搅拌10分钟后加入2mL冰醋酸和N-叔丁氧羰基-4-哌啶酮(1.98g),室温搅拌1小时后加入醋酸硼氢化钠(2.36g g),升温到65℃,在该温度下继续搅拌4小时,TLC监测直至反应不再继续。将反应液倒入水中,乙酸乙酯萃取,分液,有机相用无水硫酸钠干燥后过滤,浓缩得到粗品。该粗品经快速硅胶柱(流动相:0-10%甲醇/二氯甲烷)纯化得到类黄色固体1-(2-(氮杂环丁烷-1-羧酸叔丁酯-3-基)八氢环戊基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(2.60g,收率82%),LCMS(ESI):[M+H] +=568.5。
步骤4:1-(2-(氮杂环丁烷-3-基)八氢环戊基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
1-(2-(氮杂环丁烷-1-羧酸叔丁酯-3-基)八氢环戊基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(2.60g)加入二氯甲烷(10mL)中,然后加入三氟乙酸(2mL),室温搅拌3小时。TLC确认反应完全后,浓缩得到类黄色固体1-(2-(氮杂环丁烷-3-基)八氢环戊基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(2.20g)。该粗品固体直接用于下一步。
步骤5:3-{3-[5-{4-氨基-3-[4-(苯基氧基)苯基]吡唑并[3,4-d]嘧啶-1-基}八氢环戊并[1,2-c]吡咯-2-基]氮杂环丁-1-基}氮杂环丁烷-1-甲酸-2-甲基丙-2-基酯
1-(2-(氮杂环丁烷-3-基)八氢环戊基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(1.60g)加入二氯乙烷(10mL)中,加入三乙胺(0.69g)。室温搅拌10分钟后加入2mL 冰醋酸和1-叔丁氧羰基-3-氮杂环丁酮(1.21g),室温搅拌过夜,然后加入醋酸硼氢化钠(1.45g),升温到65℃,在该温度下继续搅拌4小时,TLC监测反应完全。将反应液倒入水中,乙酸乙酯萃取,分液,有机相用无水硫酸钠干燥后过滤,浓缩得到粗品,粗品经快速硅胶柱(流动相:0-10%甲醇/二氯甲烷)纯化得到类黄色固体3-{3-[5-{4-氨基-3-[4-(苯基氧基)苯基]吡唑并[3,4-d]嘧啶-1-基}八氢环戊并[1,2-c]吡咯-2-基]氮杂环丁-1-基}氮杂环丁烷-1-甲酸-2-甲基丙-2-基酯(1.60g,收率75%),LCMS(ESI):[M+H] +=623.7。
步骤6:1-[2-[1-(氮杂环丁-3-基)氮杂环丁-3-基]八氢环戊并[1,2-c]吡咯-5-基]-3-[4-(苯基氧基)苯基]吡唑并[3,4-d]嘧啶-4-胺
3-{3-[5-{4-氨基-3-[4-(苯基氧基)苯基]吡唑并[3,4-d]嘧啶-1-基}八氢环戊并[1,2-c]吡咯-2-基]氮杂环丁-1-基}氮杂环丁烷-1-甲酸-2-甲基丙-2-基酯(1.60g)加入二氯甲烷(15mL)中,然后加入三氟乙酸(3mL),室温搅拌10小时。TLC确认反应完全后,浓缩二氯甲烷,残余物经反相半制备柱纯化,冻干后得到淡黄色固体1-[2-[1-(氮杂环丁-3-基)氮杂环丁-3-基]八氢环戊并[1,2-c]吡咯-5-基]-3-[4-(苯基氧基)苯基]吡唑并[3,4-d]嘧啶-4-胺(1.10g,收率82%)。LCMS(ESI):[M+H] +=523.5。
步骤7:甲基5-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)六氢环戊烷[c]吡咯-2(1H)-基)-[1,3'-联氮杂环丁烷]-1'-基)吡啶甲酸酯
1-[2-[1-(氮杂环丁-3-基)氮杂环丁-3-基]八氢环戊并[1,2-c]吡咯-5-基]-3-[4-(苯基氧基)苯基]吡唑并[3,4-d]嘧啶-4-胺(250mg)溶于二甲基亚砜(25mL)中,加入5-氟吡啶甲酸甲酯(148mg),碳酸氢钠(200mg),室温搅拌5分钟,后升温至80℃,并在该温度下继续搅拌16小时.TLC监测反应完全,反应液加水稀释,乙酸乙酯萃取,分液。有机相以无水硫酸钠干燥后过滤,浓缩,后经制备薄层色谱(展开剂:10%甲醇/二氯甲烷)纯化得甲基5-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)六氢环戊烷[c]吡咯-2(1H)-基)-[1,3'-联氮杂环丁烷]-1'-基)吡啶甲酸酯,无色油(100mg,收率31%)。LCMS(ESI):[M+H] +=658.6。
步骤8:5-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)六氢环戊烷[c]吡咯-2(1H)-基)-[1,3'-联氮杂环丁烷]-1'-基)吡啶甲酸
甲基5-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)六氢环戊烷[c]吡咯-2(1H)-基)-[1,3'-联氮杂环丁烷]-1'-基)吡啶甲酸酯(100mg),溶于四氢呋喃(12mL),加入水(3mL),氢氧化锂一水合物(25mg),于室温下搅拌16小时。反应完全后,加入2M稀盐酸调节PH至1-2,浓缩后经反相(流动相:5%-45%乙腈/水(+1‰TFA))纯化,冻干得5-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)六氢环戊烷[c]吡咯-2(1H)-基)-[1,3'-联氮杂环丁烷]-1'-基)吡啶甲酸,无色油(92mg,收率94%)。LCMS(ESI):[M+H] +=644.9。
步骤9:5-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)六氢环戊[c] 吡咯-2(1H)-基)-(1,3'-二氮杂丁烷]-1'-基)-N-(2,6-二氧哌啶-3-基)吡啶酰胺
5-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)六氢环戊烷[c]吡咯-2(1H)-基)-[1,3'-联氮杂环丁烷]-1'-基)吡啶甲酸(92mg),3-氨基-2,6-哌啶二酮盐酸盐(70mg),溶于N,N-二甲基甲酰胺(15mL),室温搅拌5分钟,加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(82mg),后滴加N,N-二异丙基乙胺(150mg),室温搅拌2小时。反应完全后,反应液加水稀释,乙酸乙酯萃取,分液。有机相以无水硫酸钠干燥后过滤,浓缩,后经制备薄层色谱(展开剂:5%甲醇/二氯甲烷)纯化得5-(3-(5-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)六氢环戊[c]吡咯-2(1H)-基)-(1,3'-二氮杂丁烷]-1'-基)-N-(2,6-二氧哌啶-3-基)吡啶酰胺,白色固体(27mg,收率25%)。LCMS(ESI):[M+H] +=754.2;HPLC:纯度96%. 1H NMR(400MHz,DMSO-d 6)δppm 10.83(s,1H),8.67(d,1H),8.26(s,1H),7.86–7.79(m,2H),7.66(d,2H),7.48–7.41(m,2H),7.25–7.09(m,5H),6.91(dd,1H),5.39(dt,1H),4.74(ddd,1H),4.03(t,2H),3.80(dd,2H),3.66(s,1H),3.39(s,2H),3.08(s,3H),2.88–2.72(m,3H),2.53(d,3H),2.36(dd,5H),2.18(tt,2H),2.05–1.97(m,1H),1.90(dd,2H).
实施例29 5-(3-(4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-2-(2,6-二氧代哌啶-3-基)-6-氟异二氢吲哚-1,3-二酮
Figure PCTCN2022128411-appb-000113
合成路线:
Figure PCTCN2022128411-appb-000114
步骤1:4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸苄酯
3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(1.2g),4-(4-(甲苯磺酰氧基)环己基) 哌嗪-1-羧酸苄酯(1.88g)和碳酸铯(2.6g)加入N,N-二甲基甲酰胺,60℃反应过夜。LCMS显示产物占25%。过滤,滤液反相柱纯化,冻干后得到4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸苄酯(755mg,收率31%)。LCMS(ESI):[M+H] +=604.5。
步骤2:3-(4-苯氧基苯基)-1-(4-(哌嗪-1-基)环己基)-1H-吡唑并[3,4-d]嘧啶-4-胺
4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸苄酯(755mg)加入二氯甲烷和甲醇(1:1)的混合溶剂中,加入Pd/C,置换H2三次,室温搅拌3小时,TLC显示反应完全。过滤,浓缩,得到3-(4-苯氧基苯基)-1-(4-(哌嗪-1-基)环己基)-1H-吡唑并[3,4-d]嘧啶-4-胺(400mg,收率68%)。LCMS(ESI):[M+H] +=470.0。
步骤3:叔丁基3-(4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-羧酸盐
3-(4-苯氧基苯基)-1-(4-(哌嗪-1-基)环己基)-1H-吡唑并[3,4-d]嘧啶-4-胺(400mg),1-Boc-3-氮杂环丁酮(175mg)和醋酸(271mg)加入1,2-二氯乙烷中。升温至65℃,搅拌1小时,再加入醋酸硼氢化钠(362mg),关闭加热,搅拌1小时,LCMS显示基本反应完全。倒入水中,二氯甲烷萃取,浓缩,残余物经反相柱纯化,冻干后得到叔丁基3-(4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-羧酸盐(274mg,收率52%)。LCMS(ESI):[M+H] +=625.6。
步骤4:1-(4-(4-(氮杂环丁烷-3-基)哌嗪-1-基)环己基)-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
叔丁基3-(4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-羧酸盐(137mg)加入二氯甲烷中,加入三氟醋酸(1mL),室温搅拌1小时,TLC显示反应完全。浓缩,加入饱和碳酸氢钠水溶液中和,二氯甲烷萃取,浓缩得到1-(4-(4-(氮杂环丁烷-3-基)哌嗪-1-基)环己基)-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(115mg)。LCMS(ESI):[M+H] +=525.0。
步骤5:5-(3-(4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-2-(2,6-二氧代哌啶-3-基)-6-氟异二氢吲哚-1,3-二酮
1-(4-(4-(氮杂环丁烷-3-基)哌嗪-1-基)环己基)-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(115mg),2-(2,6-二氧代哌啶-3-基)-5,6-二氟异二氢吲哚-1,3-二酮(65mg)和N,N-二异丙基乙胺(85mg)加入二甲基亚砜中,升温至90℃,搅拌1小时,LCMS显示基本反应完全。将反应液倒入水中,乙酸乙酯萃取,分液,有机相用无水硫酸钠干燥后过滤,浓缩得到粗品,prep.TLC纯化得到5-(3-(4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-2-(2,6-二氧代哌啶-3-基)-6-氟异二氢吲哚-1,3-二酮(58mg,收率33%)。LCMS(ESI):[M+H] +=799.1; 1H NMR(400MHz,DMSO)δ11.09(s,1H),8.24(s,1H),7.63(dd,3H),7.44(t,2H),7.23–7.09(m,4H),6.93(d,1H),5.07(dd,1H),4.66(s,1H),4.23(s,2H),4.00(s,2H),2.87(dd, 1H),2.58(m,4H),2.39(s,4H),2.04(m,6H),1.51(s,2H)。
实施例30 5-(3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-2-(2,6-二氧吡哌啶-3-基)异吲哚-1,3-二酮
Figure PCTCN2022128411-appb-000115
合成路线:
Figure PCTCN2022128411-appb-000116
步骤1:4-(4-羟基环己基)哌嗪-1-羧酸苄酯
4-羟基环己酮(2g),N-Cbz-哌嗪(3.8g)和醋酸(1.58g)加入1,2-二氯乙烷中,升温至65℃,搅拌1小时,再加入醋酸硼氢化钠(7.4g),关闭加热,搅拌2小时,LCMS显示基本反应完全。加入饱和碳酸氢钠水溶液中和,二氯甲烷萃取,有机相用无水硫酸钠干燥后过滤,浓缩得到4-(4-羟基环己基)哌嗪-1-羧酸苄酯(5.57g)。该粗品直接用于下一步。LCMS(ESI):[M+H] +=319.2。
步骤2:4-(4-(甲苯磺酰氧基)环己基)哌嗪-1-羧酸苄酯
4-(4-羟基环己基)哌嗪-1-羧酸苄酯(5.57g)加入二氯甲烷中,冰水浴,再加入三乙胺(3.55g)和4-二甲氨基吡啶(428mg),搅拌5分钟。分批加入对甲苯磺酰氯(4g),控温<5℃。室温搅拌过夜。LCMS显示基本反应完全。将反应液倒入水中,二氯甲烷萃取,分液,二氯甲烷层浓缩,残余物经反相柱纯化,冻干后得到1.86g橙黄色油状物4-(4-(甲苯磺酰氧基)环己基)哌嗪-1-羧酸苄酯(1.86g,收率22.5%)。LCMS(ESI):[M+H] +=473.0。
步骤3:苄基4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸盐
3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(1g),4-(4-(甲苯磺 酰氧基)环己基)哌嗪-1-羧酸苄酯(1.34g)和碳酸钾(1.18g)加入N,N-二甲基甲酰胺,90℃封管反应过夜。LCMS显示产物占30%,主峰是3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺。过滤,滤液反相柱纯化,冻干后得到棕色固体苄基4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸盐(328mg,收率17.7%)。LCMS(ESI):[M+H] +=651.8。
步骤4:3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1-(4-(哌嗪-1-基)环己基)-1H-吡唑并[3,4-d]嘧啶-4-胺
苄基4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸盐(251mg)加入二氯甲烷和甲醇(1:1)的混合溶剂中,加入Pd/C,置换H 2三次,室温搅拌3小时,TLC显示反应完全。过滤,浓缩,得到3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1-(4-(哌嗪-1-基)环己基)-1H-吡唑并[3,4-d]嘧啶-4-胺(190mg,收率73%)。LCMS(ESI):[M+H] +=517.9。
步骤5:叔丁基3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-羧酸盐
3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1-(4-(哌嗪-1-基)环己基)-1H-吡唑并[3,4-d]嘧啶-4-胺(190mg),1-Boc-3-氮杂环丁酮(69mg)和醋酸(117mg)加入1,2-二氯乙烷中。升温至65℃,搅拌1小时,再加入醋酸硼氢化钠(156mg),关闭加热,搅拌1小时,LCMS显示基本反应完全。倒入水中,二氯甲烷萃取,浓缩,残余物经反相柱纯化,冻干后得到叔丁基3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-羧酸盐(148mg,收率60%)。LCMS(ESI):[M+H] +=673.0。
步骤6:1-(4-(氮杂环丁烷-3-基)哌嗪-1-基)环己基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
叔丁基3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-羧酸盐(148mg)加入二氯甲烷中,加入三氟醋酸(1mL),室温搅拌1小时,TLC显示反应完全。浓缩,加入饱和碳酸氢钠水溶液中和,二氯甲烷萃取,浓缩得到1-(4-(氮杂环丁烷-3-基)哌嗪-1-基)环己基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(126mg)。LCMS(ESI):[M+H] +=573.0。
步骤7:5-(3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-2-(2,6-二氧吡哌啶-3-基)异吲哚-1,3-二酮
1-(4-(氮杂环丁烷-3-基)哌嗪-1-基)环己基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(126mg),4-氟沙度利安(61mg)和N,N-二异丙基乙胺(85mg)加入二甲基亚砜,升温至90℃,搅拌1小时,LCMS显示基本反应完全。将反应液倒 入水中,乙酸乙酯萃取,分液,有机相用无水硫酸钠干燥后过滤,浓缩得到粗品,prep.TLC纯化得到5-(3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-2-(2,6-二氧吡哌啶-3-基)异吲哚-1,3-二酮(39mg,收率21%)。LCMS(ESI):[M+H] +=829.4;1H NMR(400MHz,DMSO)δ11.07(s,1H),8.23(s,1H),7.65(m,3H),7.24–7.06(m,4H),6.82(m,2H),6.71–6.62(m,1H),5.06(m,1H),4.65(s,1H),4.10(m,2H),3.90(s,3H),3.88–3.83(m,2H),3.30(s,2H),2.96–2.82(m,2H),2.66–2.54(m,4H),2.40(s,4H),2.00(m,6H),1.51(s,2H)。
实施例31 5-(3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基))哌嗪-1-基)氮杂环丁烷-1-基)-N-(2,6-二氧代哌啶-3-基)吡啶酰胺
Figure PCTCN2022128411-appb-000117
合成路线:
Figure PCTCN2022128411-appb-000118
步骤1:苄基4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸盐
3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(500mg),4-(4-(甲苯磺酰氧基)环己基)哌嗪-1-羧酸苄酯(672mg)和碳酸钾(590mg)加入N,N-二甲基甲酰胺,90℃封管反应过夜。LCMS显示产物占30%,主峰是3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺。过滤,滤液反相柱纯化,冻干后得到棕色固体苄基4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸盐(251mg,收率27%)。LCMS(ESI):[M+H] +=651.8。
步骤2:3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1-(4-(哌嗪-1-基)环己基)-1H-吡唑并[3,4-d]嘧啶-4-胺
苄基4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸盐(251mg)加入二氯甲烷和甲醇(1:1)的混合溶剂中,加入Pd/C,置换H 2三次,室温搅拌3小时,TLC显示反应完全。过滤,浓缩,得到3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1-(4-(哌嗪-1-基)环己基)-1H-吡唑并[3,4-d]嘧啶-4-胺(217mg,收率100%)。LCMS(ESI):[M+H] +=517.9。
步骤3:叔丁基3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-羧酸盐
3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1-(4-(哌嗪-1-基)环己基)-1H-吡唑并[3,4-d]嘧啶-4-胺(217mg),1-Boc-3-氮杂环丁酮(79mg)和醋酸(134mg)加入1,2-二氯乙烷中。升温至65℃,搅拌1小时,再加入醋酸硼氢化钠(178mg),关闭加热,搅拌1小时,LCMS显示基本反应完全。倒入水中,二氯甲烷萃取,浓缩,残余物经反相柱纯化,冻干后得到叔丁基3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-羧酸盐(249mg,收率88%)。LCMS(ESI):[M+H] +=673.0。
步骤4:1-(4-(氮杂环丁烷-3-基)哌嗪-1-基)环己基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺
叔丁基3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-羧酸盐(249mg)加入二氯甲烷中,加入三氟醋酸(2mL),室温搅拌1小时,TLC显示反应完全。浓缩,加入饱和碳酸氢钠水溶液中和,二氯甲烷萃取,浓缩得到1-(4-(氮杂环丁烷-3-基)哌嗪-1-基)环己基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(212mg,收率100%)。LCMS(ESI):[M+H] +=573.0。
步骤5:5-(3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)吡啶甲酸甲酯
1-(4-(氮杂环丁烷-3-基)哌嗪-1-基)环己基)-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(212mg),5-氟吡啶甲酸甲酯(58mg)和碳酸氢钠(62mg)加入二甲基亚砜中,80℃搅拌6小时,LCMS显示反应完全。过滤,滤液反相柱纯化,冻干后得到甲基5-(3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)吡啶甲酸酯(260mg,收率99%)。LCMS(ESI):[M+H] +=708.0。
步骤6:5-(3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)吡啶甲酸
5-(3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基) 环己基)哌嗪-1-基)氮杂环丁烷-1-基)吡啶甲酸甲酯(260mg)加入四氢呋喃和水中,加入氢氧化锂,40℃反应2小时,LCMS显示反应完全。浓缩,除掉四氢呋喃,加入3N盐酸调pH至弱酸性,产物水溶性较好,不易萃取。经反相柱纯化,冻干后得到5-(3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)吡啶甲酸(150mg,收率58.8%)。LCMS(ESI):[M+H] +=694.0。
步骤7:5-(3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基))哌嗪-1-基)氮杂环丁烷-1-基)-N-(2,6-二氧代哌啶-3-基)吡啶酰胺
5-(3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)吡啶甲酸(150mg)和3-氨基-2,6-二哌啶二酮(35mg)加入N,N-二甲基甲酰胺中,加入N,N-二异丙基乙胺(56mg)和HATU(82mg),室温搅拌2小时,LCMS显示反应完全。经反相柱纯化,再prep.TLC纯化得到5-(3-(4-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基))哌嗪-1-基)氮杂环丁烷-1-基)-N-(2,6-二氧代哌啶-3-基)吡啶酰胺(25mg,收率14%)。LCMS(ESI):[M+H] +=804.3; 1H NMR(400MHz,DMSO)δ10.84(s,1H),8.68(d,1H),8.23(s,1H),7.83(m,2H),7.64(m,2H),7.23–7.05(m,4H),6.91(m,1H),6.83(m,1H),4.74(m,1H),4.66(s,1H),4.08(m,2H),3.90(s,3H),3.80(s,2H),3.30(s,1H),2.81(m,1H),2.61(m,4H),2.36(m,4H),2.24–2.11(m,2H),2.01(s,6H),1.51(s,2H)。
实施例32 5-(3-(4-(4-(4-氨基-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-N-(2,6-二氧哌啶-3-基)吡啶酰胺
Figure PCTCN2022128411-appb-000119
合成路线:
Figure PCTCN2022128411-appb-000120
步骤1:甲基5-(3-(4-(4-(4-氨基-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)吡啶甲酸酯
(4-(4-(氮杂环丁烷-3-基)哌嗪-1-基l)环己基)-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-4-胺(110mg)溶于二甲基亚砜(25mL)中,加入5-氟吡啶甲酸甲酯(65mg),碳酸氢钠(88mg),室温搅拌5分钟,后升温至80℃,并在该温度下继续搅拌16小时。TLC监测反应完全,反应液加水稀释搅拌,白色固体析出,过滤烘干得甲基5-(3-(4-(4-(4-氨基-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)吡啶甲酸酯,白色固体(130mg,收率93%),LCMS(ESI):[M+H] +=660.0。
步骤2:5-(3-(4-(4-(4-氨基-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)吡啶甲酸
甲基5-(3-(4-(4-(4-氨基-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)吡啶甲酸酯(130mg),溶于四氢呋喃(20mL),加入水(5mL),氢氧化锂一水合物(33mg),于室温下搅拌16小时。反应完全后,加入2M稀盐酸调节PH至1-2,反应液浓缩后反相(流动相:5%-45%乙腈/水(+1‰TFA))纯化,冻干得5-(3-(4-(4-(4-氨基-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)吡啶甲酸,无色油(100mg,收率78%),LCMS(ESI):[M+H] +=646.0。
步骤3:5-(3-(4-(4-(4-氨基-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-N-(2,6-二氧哌啶-3-基)吡啶酰胺
5-(3-(4-(4-(4-氨基-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)吡啶甲酸(100mg),3-氨基-2,6-哌啶二酮盐酸盐(76mg),溶于N,N-二甲基甲酰胺(15mL),加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(88mg),室温搅拌5分钟,后滴加N,N-二异丙基乙胺(150mg),室温搅拌2小时。反应完全后,反应液加水稀释,二氯甲烷萃取,分液。有机相以无水硫酸钠干燥后过滤,浓缩,后经制备薄层色谱(展开剂:10%甲醇/二氯甲烷)纯化得5-(3-(4-(4-(4-氨基-3-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-N-(2,6-二氧哌啶-3-基)吡啶酰胺,白色固体(64mg,收率54%)。LCMS(ESI):[M+H] +=756.3;HPLC:纯度99%.1H NMR(400MHz,DMSO-d6)δppm 10.85(s,1H),8.69(m,1H),8.24(s,1H),7.90–7.77(m,2H),7.65(d,2H),7.49–7.39(m,2H),7.23–7.09(m,5H),6.90(m,1H),4.81–4.60(m,2H),4.07(m,2H),3.80(m,2H),2.79(m,1H),2.60(m,5H),2.41(s,4H),2.18(m,1H),2.00(m,7H),1.51(m,2H),1.24(m,1H).
实施例33 5-(3-(4-(4-氨基-3-(4-(2-氟-3-甲氧基苯氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂丁-1-基)-2-(2,6-二氧哌啶-3-基)-6-氟异吲哚-1,3-二酮
Figure PCTCN2022128411-appb-000121
类白色固体(0.9mg)。LCMS(ESI):[M+H] +=847.7;HPLC:纯度91%。
实施例34和35 5-(3-(反式-4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-2-(2,6-二氧代哌啶-3-基)-6-氟异二氢吲哚-1,3-二酮和5-(3-(顺式-4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-2-(2,6-二氧代哌啶-3-基)-6-氟异二氢吲哚-1,3-二酮
Figure PCTCN2022128411-appb-000122
1-(4-(4-(氮杂环丁烷-3-基)哌嗪-1-基)环己基)-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-4-胺(2.0g),2-(2,6-二氧代哌啶-3-基)-5,6-二氟异二氢吲哚-1,3-二酮(1.13g)和N,N-二异丙基乙胺(1.47g)加入二甲基亚砜100mL中,升温至90℃,搅拌1小时,LCMS显示基本反应完全。将反应液倒入1000mL水中,有固体析出,过滤得4.2g。该固体经柱层析分离(淋洗剂DCM/MeOH=100/1 50/1 33/1 25/1),收取DCM/MeOH=25/1(600mL)洗脱液,浓缩得5-(3-(反式-4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-2-(2,6-二氧代哌啶-3-基)-6-氟 异二氢吲哚-1,3-二酮(730mg白色固体),LCMS(ESI):[M+H] +=799.8; 1H NMR(400MHz,DMSO)δ11.09(s,1H),8.24(s,1H),7.63(m,3H),7.44(m,2H),7.23–7.09(m,4H),6.93(d,J=7.6Hz,1H),5.07(m,1H),4.66(m,1H),4.23(m,2H),4.01(m,2H),2.87(m,1H),2.65-2.30(m,8H),2.04(m,6H),1.51(m,2H);收取淋洗剂(DCM/MeOH=20/1,1200mL)洗脱液,浓缩得到5-(3-(顺式-4-(4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁烷-1-基)-2-(2,6-二氧代哌啶-3-基)-6-氟异二氢吲哚-1,3-二酮(270mg白色固体),LCMS(ESI):[M+H] +=799.8; 1H NMR(400MHz,DMSO)δ11.09(s,1H),8.24(s,1H),7.63(m,3H),7.44(t,J=7.9Hz,2H),7.23–7.09(m,4H),6.93(d,J=7.6Hz,1H),5.07(m,1H),4.66(m,1H),4.23(m,2H),4.00(m,2H),2.87(m,1H),2.65-2.30(m,8H),2.04(m,6H),1.51(m,2H)。
实施例36 5-(3-(4-((1R,4R)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((S)-2,6-二氧哌啶-3-基)-6-氟异吲哚-1,3-二酮
Figure PCTCN2022128411-appb-000123
合成路线:
Figure PCTCN2022128411-appb-000124
步骤1:(1s,4s)-4-(4-苄基哌嗪-1-基)环己醇
顺式-4-氨基环己醇盐酸盐(4.0g)、N-苄基-2-氯-N-(2-氯乙基)乙胺盐酸盐(8.5g)和NaHCO 3(13.3g)溶于乙醇(60mL)中,混合物在90℃下搅拌16h。混合物减压浓缩,将残留物用水(200mL)和乙酸乙酯(150mL×3)萃取。有机相用无水硫酸钠干燥,浓缩,所得粗品用硅胶柱色谱纯化(DCM/甲醇(v/v)=10/1),得(1s,4s)-4-(4-苄基哌嗪-1-基)环己醇,呈浅黄色油状物(4.5g,收率62%)。LCMS(ESI):[M+H] +=275.3。
步骤2:(1s,4s)-4-(哌嗪-1-基)环己醇
(1s,4s)-4-(4-苄基哌嗪-1-基)环己醇(4.5g)、醋酸(1mL)和Pd/C(10%,450mg)溶于甲醇(30mL)中,室温在H 2(1atm)下搅拌16h。过滤混合物,浓缩滤液,得粗品(1s,4s)-4-(哌嗪-1-基)环己醇,呈浅黄色油状物(3.5g)。LCMS(ESI):[M+H] +=185.1。
步骤3:4-((1s,4s)-4-羟基环己基)哌嗪-1-羧酸叔丁酯
将粗品(1s,4s)-4-(哌嗪-1-基)环己醇(3.5g)、(Boc) 2O(4.6g)和DIPEA(4.2g)溶于THF(40mL)中的混合物在室温下搅拌4h。后将混合物浓缩,用水(150mL)稀释,DCM(150mL×2)萃取。有机相用无水Na 2SO 4干燥,浓缩,后粗品经硅胶柱色谱纯化(DCM/甲醇(v/v)=10/1),得4-((1s,4s)-4-羟基环己基)哌嗪-1-羧酸叔丁酯,无色油状物(3.8g,收率81%)。LCMS(ESI):[M+H] +=285.2。
步骤4:4-((1s,4s)-4-(甲基磺酰氧基)环己基)哌嗪-1-羧酸叔丁酯
在0℃下向4-((1s,4s)-4-羟基环己基)哌嗪-1-羧酸叔丁酯(3.8g)和DIPEA(2.6g)溶于DCM(40mL)中的混合物中逐滴添加MsCl(1.8g)。反应混合液室温下搅拌2h。将反应液用水(150mL)洗涤,水相DCM(100mL×2)萃取,合并有机相用无水硫酸钠干燥,过滤并浓缩,得到粗品4-((1s,4s)-4-(甲基磺酰氧基)环己基)哌嗪-1-羧酸叔丁酯,棕色油状(4.0g)。LCMS(ESI):[M+H] +=363.2。
步骤5:4-((1r,4r)-4-(4-氨基-3-溴-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸叔丁酯
3-溴-1H-吡唑并[3,4-d]嘧啶-4-胺(2.2g),4-((1s,4s)-4-(甲基磺酰氧基)环己基)哌嗪-1-羧酸叔丁酯(4.0g)和Cs 2CO 3(6.7g)溶于DMF(50mL)中,在100℃下搅拌16h。将反应混合物冷却至室温,用乙酸乙酯(200mL)稀释,并用水(50mL×4)洗涤。有机相经无水硫酸钠干燥,过滤,浓缩后粗品用硅胶柱色谱纯化(DCM/甲醇(v/v)=10/1),得4-((1r,4r)-4-(4-氨基-3-溴-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸叔丁酯,浅褐色固体(970mg,收率19%)。LCMS(ESI):[M+H] +=480.2。
步骤6:4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸叔丁酯
4-((1r,4r)-4-(4-氨基-3-溴-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸叔丁酯(970mg)、4,4,5,5-四甲基-2-(4-苯氧基苯基)-1,3,2-二氧杂硼杂环戊烷(897mg)、Pd(PPh 3) 2Cl 2(70mg)和K 3PO 4(1.0g)溶于1,4-二氧六环(8mL)和H 2O(2mL)中,氮气保护,反应液在80℃下搅拌16h。将反应液冷却至室温,过滤,将滤液用水(100mL)稀释并用乙酸乙酯(100mL×3)萃取。将合并的有机相经无水硫酸钠干燥,浓缩,所得粗品用硅胶柱色谱纯化(DCM/甲醇(v/v)=10/1),得4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸叔丁酯,米色固体(880mg,收率76%)。LCMS(ESI):[M+H] +=570.4。
步骤7:3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-羧酸叔丁酯
向4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸叔丁酯(880mg)溶于DCM(15mL)中,所得混合溶液中加入三氟醋酸(3mL),在室温下搅拌2h。将反应液减压浓缩,得棕色油状粗品。该粗品溶于二氯乙烷(30mL),加入三乙胺(0.4mL),室温搅拌10min后加入1-Boc-3-氮杂环丁酮(317mg),滴加醋酸(0.6mL),于65℃搅拌1h。将反应液冷却至室温后,加入醋酸硼氢化钠(655mg,3.1mmol),加热至65℃继续搅拌2h。反应完全后冷却至室温,加入碳酸氢钠水溶液(100mL)洗涤,DCM(100mL×2)萃取,分液。有机相以无水硫酸钠干燥后过滤,浓缩,经制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1)得3-(4-((1r,4r)-4-(4- 氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-羧酸叔丁酯,米色固体(730mg,收率75%)。LCMS(ESI):[M+H] +=625.4[M+H] +
步骤8:4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸二甲酯
3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-羧酸叔丁酯(730mg)溶于DCM(15mL)中,所得反应溶液中加入三氟醋酸(3mL),在室温下搅拌2h。将反应液浓缩,得棕色油状粗品。该粗品溶解于20mL DMSO中,加入4,5-二氟苯-1,2-二甲酸二甲酯(405mg)(合成方法见WO2018/172423)和DIPEA(603mg),于85℃下搅拌反应6h。将反应混合物冷却至室温,用乙酸乙酯(200mL)稀释,并用水(50mL×3)洗涤。有机相经无水硫酸钠干燥,过滤,浓缩,后经制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1)得4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸二甲酯,无色透明油状物(610mg,收率71%)。LCMS(ESI):[M+H] +=735.3。
步骤9:4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸
4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸二甲酯(610mg)溶于6mL 1,4-二氧六环中,加入6mL甲醇及2mL氢氧化钠溶液(5M),于75℃下搅拌反应2h。冷却至室温,以2M稀盐酸调节PH至5.0,减压浓缩得粗品。该粗品经制备HPLC纯化(C18,流动相:乙腈/水(含0.1%TFA)=5%~35%),冻干得4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸,白色固体(450mg,收率76%)。LCMS(ESI):[M+H] +=707.2。
步骤10:5-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-6-氟-2-((S)-2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮
将上述4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸(124mg)溶解在4mL N,N-二甲基甲酰胺中,加入N-甲基咪唑(86mg),再加入四甲基氯代脲六氟磷酸酯(123mg),室温搅拌30min,加入(S)-3-氨基哌啶-2,6-二酮盐酸盐(29mg)(合成方法见Heterocycles 2015,91,764-781),继续室温搅拌6h。反应液中加入60mL水,以二氯甲烷/甲醇(v/v)=10/1(20mL×3)萃取,有机相用10mL饱和氯化钠溶液洗涤,无水硫酸钠干燥,减压浓缩后粗品用制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1),得5-(3-(4-((1R,4R)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-6-氟-2-((S)-2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮,黄色固体(38mg,收率27%)。LCMS(ESI):[M+H] +=799.8;HPLC:纯度96.8%. 1H NMR(400MHz,DMSO-d 6)δ11.08(s,1H),8.24(s,1H),7.65(d,J=8.7Hz,2H),7.60(d,J=11.1Hz,1H),7.44(t,J=7.9Hz,2H),7.23–7.09(m,5H),6.93(d,J=7.6Hz,1H),5.07(dd,J=12.8,5.4Hz,1H),4.74–4.60(m,1H),4.31–4.18(m,2H),4.06–3.93(m,2H),3.32–3.15(m,4H),2.95–2.81(m,1H),2.78–2.53(m,5H),2.50–2.31(m,4H),2.12–1.93 (m,6H),1.62–1.45(m,2H).
实施例37 5-(3-(4-((1R,4R)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((R)-2,6-二氧哌啶-3-基)-6-氟异吲哚-1,3-二酮
Figure PCTCN2022128411-appb-000125
合成路线:
Figure PCTCN2022128411-appb-000126
步骤1:5-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-6-氟-2-((R)-2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮
将4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸(130mg)溶解在4mL N,N-二甲基甲酰胺中,加入N-甲基咪唑(90mg),再加入四甲基氯代脲六氟磷酸酯(129mg),室温搅拌30min,加入(R)-3-氨基哌啶-2,6-二酮盐酸盐(30mg)(合成方法见Heterocycles2015,91,764-781),继续室温搅拌16h。反应液中加入60mL水,以二氯甲烷/甲醇(v/v)=10/1(20mL×3)萃取,有机相用10mL饱和氯化钠溶液洗涤,无水硫酸钠干燥,减压浓缩后粗品用制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1),得5-(3-(4-((1R,4R)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-6-氟-2-((R)-2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮,黄色固体(63mg,收率43%)。LCMS(ESI):[M+H] +=799.7;HPLC:纯度97.9%. 1H NMR(400MHz,DMSO-d 6)δ11.09(s,1H),8.23(s,1H),7.65(d,J=8.6Hz,2H),7.60(d,J=11.2Hz,1H),7.44(t,J=7.9Hz,2H),7.24–7.08(m,5H),6.92(d,J=7.6Hz,1H),5.07(dd,J=12.8,5.4Hz,1H),4.71–4.60(m,1H),4.30–4.18(m,2H),4.05–3.94(m,2H),3.34–3.24(m,5H),2.94–2.83(m,1H),2.68–2.53(m,5H),2.49–2.32(m,4H),2.11–1.90(m,6H),1.58–1.42(m,2H).
实施例38 5-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((S)-2,6-二氧哌啶-3-基)-6-氟异吲哚-1,3-二酮
Figure PCTCN2022128411-appb-000127
合成路线:
Figure PCTCN2022128411-appb-000128
步骤1:(1r,4r)-4-(4-苄基哌嗪-1-基)环己醇
反式-4-氨基环己醇盐酸盐(20.0g)、N-苄基-2-氯-N-(2-氯乙基)乙胺盐酸盐(42.5g)和NaHCO 3(66.5g)溶于乙醇(300mL)中,在90℃下搅拌16h。减压浓缩,将残留物用水(400mL)稀释,乙酸乙酯(400mL×3)萃取。有机相用无水硫酸钠干燥,浓缩,所得粗品用硅胶柱色谱分离提纯(DCM/甲醇(v/v)=10/1),得(1r,4r)-4-(4-苄基哌嗪-1-基)环己醇,浅黄色油状物(12.7g,收率35%)。LCMS(ESI):[M+H] +=275.3。
步骤2:(1r,4r)-4-(哌嗪-1-基)环己醇
(1r,4r)-4-(4-苄基哌嗪-1-基)环己醇(12.7g)、醋酸(3mL)和Pd/C(10%,1.27g)溶于甲醇(100mL)中,室温在H 2(1atm)下搅拌16h。过滤混合物,浓缩滤液,得粗品(1r,4r)-4-(哌嗪-1-基)环己醇,浅黄色油状物(18.7g)。LCMS(ESI):[M+H] +=185.1。
步骤3:4-((1r,4r)-4-羟基环己基)哌嗪-1-羧酸叔丁酯
粗品(1r,4r)-4-(哌嗪-1-基)环己醇(18.7g)、(Boc) 2O(28.9g)和DIPEA(26g)溶于THF(100mL)中,室温下搅拌4h。将混合物浓缩,用水(300mL)稀释,DCM(300mL×2)萃取。有机相用无水硫酸钠干燥,浓缩,所得粗品用硅胶柱色谱分离纯化(DCM/甲醇(v/v)=10/1),得4-((1r,4r)-4-羟基环己基)哌嗪-1-羧酸叔丁酯,无色油状物(10g,收率35%)。LCMS(ESI):[M+H] +=285.2。
步骤4:4-((1r,4r)-4-(甲基磺酰氧基)环己基)哌嗪-1-羧酸叔丁酯
0℃下向4-((1r,4r)-4-羟基环己基)哌嗪-1-羧酸叔丁酯(3g)和DIPEA(2.0g)于DCM(40mL)中的混合物中逐滴添加MsCl(1.4g)。室温下搅拌2h。反应液加入水(150mL),用DCM(100mL×2)萃取,合并有机相用无水硫酸钠干燥,过滤并浓缩,得到粗品4-((1r,4r)-4-(甲基磺酰氧基)环己基)哌嗪-1-羧酸叔丁酯,棕色油状物(4.0g)。LCMS(ESI):[M+H] +=363.2。
步骤5:4-((1s,4s)-4-(4-氨基-3-溴-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸叔丁酯
3-溴-1H-吡唑并[3,4-d]嘧啶-4-胺(2.2g)、4-((1r,4r)-4-(甲基磺酰氧基)环己基)哌嗪-1-羧酸叔丁酯(4.0g)和Cs 2CO 3(6.7g)溶于DMF(50mL)中,在100℃搅拌16h。将反应混合物冷却至室温,用乙酸乙酯(200mL)稀释,并用水(50mL×4)洗涤。有机相经无水硫酸钠干燥,过滤,浓缩,所得粗品经制备HPLC纯化(C18,流动相:乙腈/水(含0.1%TFA)=15%~40%),冻干得4-((1s,4s)-4-(4-氨基-3-溴-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸叔丁酯,浅褐色固体(1.3g,收率26%)。LCMS(ESI):[M+H] +=480.2。
步骤6:4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸叔丁酯
将4-((1s,4s)-4-(4-氨基-3-溴-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸叔丁酯(1.3g)、4,4,5,5-四甲基-2-(4-苯氧基苯基)-1,3,2-二氧杂硼杂环戊烷(1.2g)、Pd(PPh 3) 2Cl 2(91mg)和K 3PO 4(1.43g)溶于1,4-二氧六环(8mL)和H 2O(2mL)中,反应混合物在80℃下搅拌16h。将反应液冷却至室温,后过滤,将滤液用水(100mL)稀释并用乙酸乙酯(100mL×3)萃取。合并的有机相经无水硫酸钠干燥,浓缩,粗品用硅胶柱色谱分离提纯(DCM/甲醇(v/v)=10/1),得4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸叔丁酯,米色固体(417mg,收率27%)。LCMS(ESI):[M+H] +=570.4。
步骤7:3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-羧酸叔丁酯
4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-羧酸叔丁酯(417mg)溶于DCM(10mL)中,反应液中加入三氟醋酸(3mL),在室温下搅拌2h。将反应液减压浓缩,得棕色油状粗品。该粗品溶于二氯乙烷(30mL),加入三乙胺(0.2mL),室温搅拌10min,加入1-Boc-3-氮杂环丁酮(159mg),滴加醋酸(0.3mL),于65℃搅拌1h。将反应液冷却至室温后,加入醋酸硼氢化钠(311mg),加热至65℃继续搅拌2h。反应完全后冷却至室温,加入碳酸氢钠水溶液(50mL)洗涤,DCM(50mL×2)萃取,分液。有机相以无水硫酸钠干燥后过滤,浓缩,后经制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1)得3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧 基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-羧酸叔丁酯,呈米色固体(420mg,收率92%)。LCMS(ESI):[M+H] +=625.4。
步骤8:4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸二甲酯
3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-羧酸叔丁酯(420mg)溶于DCM(10mL)中,所得反应液中加入三氟醋酸(3mL),在室温下搅拌2h。后将反应液真空浓缩,得棕色油状粗品。该粗品溶解于10mL DMSO中,加入4,5-二氟苯-1,2-二甲酸二甲酯(230mg)(合成方法见WO2018/172423)和DIPEA(335mg),于85℃下搅拌反应6h。将反应混合物冷却至室温,用乙酸乙酯(100mL)稀释,并用水(50mL×3)洗涤。有机相经无水硫酸钠干燥,过滤,浓缩,后经制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1)得4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸二甲酯,无色透明油状物(490mg,收率99%)。LCMS(ESI):[M+H] +=735.3。
步骤9:4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸
将4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸二甲酯(490mg)溶于5mL 1,4-二氧六环中,加入5mL甲醇和1.5mL氢氧化钠溶液(5M),反应混合物于75℃下搅拌反应2h。冷却至室温,用2M稀盐酸调节PH至5.0,减压浓缩得粗品。该粗品经制备HPLC纯化(C18,流动相:乙腈/水(含0.1%TFA)=5%~35%),冻干得4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸,白色固体(264mg,收率56%)。LCMS(ESI):[M+H] +=707.2。
步骤10:5-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((S)-2,6-二氧哌啶-3-基)-6-氟异吲哚-1,3-二酮
将上述4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸(90mg)溶解在4mL N,N-二甲基甲酰胺中,加入N-甲基咪唑(62mg),再加入四甲基氯代脲六氟磷酸酯(90mg),室温搅拌30min,加入(S)-3-氨基哌啶-2,6-二酮盐酸盐(21mg)(合成方法见Heterocycles2015,91,764-781),继续室温搅拌2.5h。反应液中加入30mL水,以二氯甲烷/甲醇(v/v)=10/1(20mL×3)萃取,有机相用10mL饱和氯化钠溶液洗涤,无水硫酸钠干燥,减压浓缩后粗品用制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1),得5-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((S)-2,6-二氧哌啶-3-基)-6-氟异吲哚-1,3-二酮,黄色固体(16mg,收率16%)。LCMS(ESI):[M+H] +=799.9;HPLC:纯度99.2%。 1H NMR(400MHz,DMSO-d 6)δ11.09(s,1H),8.24(s,1H),7.63(d,J=8.3Hz,2H),7.60(d,J=11.2Hz,1H),7.44(t,J=7.8Hz,2H),7.27–7.09(m,4H),6.93(d,J=7.4Hz,1H),5.08(dd,J=12.8Hz,5.4Hz,1H),4.74–4.60(m,1H),4.31–4.18(m,2H),4.06–3.93(m,2H),,3.29– 3.11(m,4H),2.95–2.81(m,1H),2.72–2.56(m,5H),2.50–2.31(m,4H),2.12–1.93(m,6H),1.62–1.45(m,2H).
实施例39 5-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-(
Figure PCTCN2022128411-appb-000129
-2,6-二氧哌啶-3-基)-6-氟异吲哚-1,3-二酮
Figure PCTCN2022128411-appb-000130
合成路线:
Figure PCTCN2022128411-appb-000131
步骤:将4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-基)-5-氟苯-1,2-二甲酸(90mg)溶解在4mL N,N-二甲基甲酰胺中,加入N-甲基咪唑(62mg),再加入四甲基氯代脲六氟磷酸酯(90mg),室温搅拌30min,加入(R)-3-氨基哌啶-2,6-二酮盐酸盐(21mg)(合成方法见Heterocycles 2015,91,764-781),继续室温搅拌2.5h。后反应液中加入30mL水,以二氯甲烷/甲醇(v/v)=10/1(20mL×3)萃取,有机相用10mL饱和氯化钠溶液洗涤,无水硫酸钠干燥,减压浓缩后粗品用制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1),得5-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((R)-2,6-二氧哌啶-3-基)-6-氟异吲哚-1,3-二酮,黄色固体(16mg,收率16%)。LCMS(ESI):[M+H] +=799.9;HPLC:纯度99.5%。 1H NMR(400MHz,DMSO-d 6)δ11.09(s,1H),8.24(s,1H),7.65(d,J=8.3Hz,2H),7.61(d,J=11.2Hz,1H),7.44(t,J=7.8Hz,2H),7.27–7.09(m,4H),6.93(d,J=7.4Hz,1H),5.08(dd,J=12.8Hz,5.4Hz,1H),4.74–4.60(m,1H),4.31–4.18(m,2H),4.06–3.93(m,2H),3.29–3.11(m,4H),2.95–2.81(m,1H),2.72–2.56(m,5H),2.50–2.31(m,4H),2.12–1.93(m,6H),1.62–1.45(m,2H).
实施例40 5-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((S)-2,6-二氧哌啶-3-基)异吲哚-1,3-二酮
Figure PCTCN2022128411-appb-000132
合成路线:
Figure PCTCN2022128411-appb-000133
步骤1:邻苯二甲酸二甲酯4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基
3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂环-1-羧酸叔丁酯(730mg)溶于DCM(15mL)中,所得反应溶液中加入三氟醋酸(3mL),在室温下搅拌2h。将反应液浓缩,得棕色油状粗品。该粗品溶解于20mL DMSO中,加入4-氟邻苯二甲酸二甲酯(373mg)和DIPEA(603mg),于85℃下搅拌反应6h。将反应混合物冷却至室温,用乙酸乙酯(200mL)稀释,并用水(50mL×3)洗涤。有机相经无水硫酸钠干燥,过滤,浓缩,后经制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1)得邻苯二甲酸二甲酯4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基,无色透明油状物(500mg,收率60%)。LCMS(ESI):[M+H] +=717.3。
步骤2:4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基-氮杂环-1-基]邻苯二甲酸
邻苯二甲酸二甲酯4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基(500mg)溶于6mL 1,4-二氧六环中,加入6mL甲醇及2mL氢氧化钠溶液(5M),于75℃下搅拌反应2h。冷却至室温,以2M稀盐酸调节PH至5.0,减压浓缩得粗品。该粗品经制备HPLC纯化(C18,流动相:乙腈/水(含0.1%TFA)=5%~35%),冻干得4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基-氮杂环-1-基]邻苯二甲酸,白色固体(300mg,收率62%)。LCMS(ESI):[M+H] +=689.3。
步骤3:5-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((S)-2,6-二氧哌啶-3-基)异吲哚-1,3-二酮
将上述4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基) 环己基)哌嗪-1-基-氮杂环-1-基]邻苯二甲酸(121mg)溶解在4mL N,N-二甲基甲酰胺中,加入N-甲基咪唑(86mg),再加入四甲基氯代脲六氟磷酸酯(123mg),室温搅拌30min,加入(S)-3-氨基哌啶-2,6-二酮盐酸盐(29mg)(合成方法见Heterocycles 2015,91,764-781),继续室温搅拌6h。反应液中加入60mL水,以二氯甲烷/甲醇(v/v)=10/1(20mL×3)萃取,有机相用10mL饱和氯化钠溶液洗涤,无水硫酸钠干燥,减压浓缩后粗品用制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1),得5-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((S)-2,6-二氧哌啶-3-基)异吲哚-1,3-二酮,黄色固体(30mg,收率27%)。LCMS(ESI):[M+H] +=781.4;HPLC:纯度96.0%. 1H NMR(400MHz,DMSO)δ11.07(s,1H),8.23(s,1H),7.65(d,J=8.0Hz,3H),7.43(t,J=8.0Hz,2H),7.23–7.09(m,4H),6.80(s,1H),6.66(d,J=8.0Hz,1H),5.06(m,1H),4.66(m,1H),4.10(m,2H),3.86(m,2H),3.29(m,1H),2.87(m,1H),2.70-2.25(m,8H),2.04(m,6H),1.51(m,2H)。
实施例41 5-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((R)-2,6-二氧哌啶-3-基)异吲哚-1,3-二酮
Figure PCTCN2022128411-appb-000134
合成路线:
Figure PCTCN2022128411-appb-000135
步骤1:5-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-(
Figure PCTCN2022128411-appb-000136
-2,6-二氧哌啶-3-基)异吲哚-1,3-二酮
4-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基-氮杂环-1-基]邻苯二甲酸(121mg)溶解在4mL N,N-二甲基甲酰胺中,加入N-甲基咪唑(86mg),再加入四甲基氯代脲六氟磷酸酯(123mg),室温搅拌30min,加入(S)-3-氨基哌啶-2,6-二酮盐酸盐(29mg)(合成方法见Heterocycles 2015,91,764-781),继续室温搅拌6h。反应液中加入60mL水,以二氯甲烷/甲醇(v/v)=10/1(20mL×3)萃取,有机相用10mL饱和氯化钠溶液洗涤,无水硫酸钠干燥,减压浓缩后粗品用制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1),得5-(3-(4-((1r,4r)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((R)-2,6-二氧哌啶-3-基)异吲哚-1,3-二酮(27mg,收率20%)。LCMS(ESI):[M+H] +=781.4;HPLC:纯度96.6%. 1H NMR(400MHz,DMSO)δ11.07(s,1H),8.24(s,1H),7.66(d,J=8.0Hz,3H),7.44(t,J=8.0Hz,2H),7.23–7.09(m,4H),6.80(s,1H), 6.66(d,J=8.0Hz,1H),5.06(m,1H),4.66(m,1H),4.10(m,2H),3.86(m,2H),3.29(m,1H),2.87(m,1H),2.70-2.25(m,8H),2.04(m,6H),1.51(m,2H)。
实施例42 5-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((S)-2,6-二氧哌啶-3-基)异吲哚-1,3-二酮
Figure PCTCN2022128411-appb-000137
合成路线:
Figure PCTCN2022128411-appb-000138
步骤1:邻苯二甲酸二甲酯4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基
3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基)氮杂环丁-1-羧酸叔丁酯(420mg)溶于DCM(10mL)中,所得反应液中加入三氟醋酸(3mL),在室温下搅拌2h。后将反应液真空浓缩,得棕色油状粗品。该粗品溶解于10mL DMSO中,加入4-氟邻苯二甲酸二甲酯(212mg)和DIPEA(335mg),于85℃下搅拌反应6h。将反应混合物冷却至室温,用乙酸乙酯(100mL)稀释,并用水(50mL×3)洗涤。有机相经无水硫酸钠干燥,过滤,浓缩,后经制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1)得邻苯二甲酸二甲酯4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基,无色透明油状物(370mg,收率77%)。LCMS(ESI):[M+H] +=717.3。
步骤2:4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基-氮杂环-1-基]邻苯二甲酸
将邻苯二甲酸二甲酯4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基(370mg)溶于5mL 1,4-二氧六环中,加入5mL甲醇和1.5mL氢氧化钠溶液(5M),反应混合物于75℃下搅拌反应2h。冷却至室温,用2M稀盐酸调节PH至5.0,减压浓缩得粗品。该粗品经制备HPLC纯化(C18,流 动相:乙腈/水(含0.1%TFA)=5%~35%),冻干得4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基-氮杂环-1-基]邻苯二甲酸,白色固体(200mg,收率56%)。LCMS(ESI):[M+H] +=689.3。
步骤3:5-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((S)-2,6-二氧哌啶-3-基)异吲哚-1,3-二酮
将上述4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基-氮杂环-1-基]邻苯二甲酸(90mg)溶解在4mL N,N-二甲基甲酰胺中,加入N-甲基咪唑(62mg),再加入四甲基氯代脲六氟磷酸酯(90mg),室温搅拌30min,加入(S)-3-氨基哌啶-2,6-二酮盐酸盐(22mg)(合成方法见Heterocycles 2015,91,764-781),继续室温搅拌2.5h。反应液中加入30mL水,以二氯甲烷/甲醇(v/v)=10/1(20mL×3)萃取,有机相用10mL饱和氯化钠溶液洗涤,无水硫酸钠干燥,减压浓缩后粗品用制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1),得5-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((S)-2,6-二氧哌啶-3-基)异吲哚-1,3-二酮,黄色固体(18mg,收率18%)。LCMS(ESI):[M+H] +=781.4;HPLC:纯度97.3%。 1H NMR(400MHz,DMSO)δ11.07(s,1H),8.24(s,1H),7.66(d,J=8.0Hz,3H),7.44(t,J=8.0Hz,2H),7.23–7.09(m,4H),6.80(s,1H),6.66(d,J=8.0Hz,1H),5.06(m,1H),4.66(m,1H),4.10(m,2H),3.86(m,2H),3.29(m,1H),2.87(m,1H),2.70-2.25(m,8H),2.04(m,6H),1.51(m,2H)。
实施例43 5-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((R)-2,6-二氧哌啶-3-基)异吲哚-1,3-二酮
Figure PCTCN2022128411-appb-000139
合成路线:
Figure PCTCN2022128411-appb-000140
步骤:将4-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基-氮杂环-1-基]邻苯二甲酸(90mg)溶解在4mL N,N-二甲基甲酰胺中,加入N-甲基咪唑(62mg),再加入四甲基氯代脲六氟磷酸酯(90mg),室温搅拌30min,加入(R)-3-氨基哌啶-2,6-二酮盐酸盐(22mg)(合成方法见Heterocycles 2015,91,764-781),继续室温搅拌2.5h。后反应液中加入30mL水,以二氯甲烷/甲醇(v/v)=10/1(20mL×3)萃取,有机相用10mL饱和氯化钠溶液洗涤, 无水硫酸钠干燥,减压浓缩后粗品用制备薄层色谱纯化(展开剂:DCM/甲醇(v/v)=15/1),得5-(3-(4-((1s,4s)-4-(4-氨基-3-(4-苯氧基苯基)-1H吡唑并[3,4-d]嘧啶-1-基)环己基)哌嗪-1-基]氮杂啶-1-基]-2-((R)-2,6-二氧哌啶-3-基)异吲哚-1,3-二酮,黄色固体(18mg,收率18%)。LCMS(ESI):[M+H] +=781.5;HPLC:纯度96.5%。 1H NMR(400MHz,DMSO)δ11.07(s,1H),8.24(s,1H),7.66(d,J=8.0Hz,3H),7.44(t,J=8.0Hz,2H),7.23–7.09(m,4H),6.80(s,1H),6.66(d,J=8.0Hz,1H),5.06(m,1H),4.66(m,1H),4.10(m,2H),3.86(m,2H),3.29(m,1H),2.87(m,1H),2.70-2.25(m,8H),2.04(m,6H),1.51(m,2H)。
效果例1
1.1分子实验
BTK(WT)及其突变蛋白BTK(C481S)的活性检测,以均相时间分辨荧光HTRF的TK-s为底物,BTK在ATP的参与下可催化生物素标记的多肽底物TK-s发生磷酸化修饰。Eu标记的TK-s特异性磷酸化抗体与底物通过抗原抗体反应,同时链霉亲和素标记的受体XL665与生物素特异性相互作用,从而使得Eu标记的供体能够与链霉亲和素标记的受体在空间上相互靠近。在320nm激发下,Eu标记的供体可发射出620nm波长的能量,当生物分子相互作用导致供体和受体的荧光基团接近时,部分能量共振转移到受体XL665上,使其受到激发,发出665nm波长的发射光。665nm的发射光仅由供体引起的FRET产生。TK-s底物经BTK磷酸化后,在Envision TM设置激发波长320nm,在发射波长620nm和665nm处读取荧光值。通过检测单位时间内665nm和620nm的荧光比值的变化,经计算得到BTK反应初速度。
将化合物和酶缓冲液加入384反应板(ProxiPlateTM-384 Plus,PerkinElmer)中,室温孵育半小时后,加入底物缓冲液启动反应,室温孵育一个小时后,加入XL665和抗体继续孵育一个小时,最终用EnvisionTM检测荧光信号665nm和620nm的比值。同时设不加酶的空白对照组、以DMSO替代化合物的溶剂对照组。反应终体积为10μL,具体反应体系为2%DMSO,0.5ng/μL BTK,1μM TK-s,80μM ATP,5mM MgCl 2,1mM DTT,20nM SEB,1×kinase buffer(激酶缓冲液)。实验结果如表1所示
表1:化合物对BTK和BTK(C481S)激酶的抑制活性
Figure PCTCN2022128411-appb-000141
Figure PCTCN2022128411-appb-000142
效果:运用本发明技术所合成的化合物对BTK、以及BTK(C481)激酶有一定的抑制作用。
1.2细胞实验
1.2.1 BTK蛋白降解试验
分别利用Mino和HEK293过转BTK(C481S)稳转细胞检测化合物对BTK(WT) 和BTK(C481S)蛋白的降解能力,Mino人套细胞淋巴瘤细胞株,购自ATCC,培养基RPMI-1640+15%FBS+1%双抗;HEK293 OE BTK(C481S)人胚胎肾细胞293过转BTK C481S质粒形成过表达BTK C481S细胞,DMEM+10%FBS+1%双抗,向6孔板内加入Mino细胞和HEK293 OE BTK(C481S)稳转细胞株悬液37℃静置0.5h,将不同浓度的化合物加入细胞悬液,37℃,5%CO 2,饱和湿度处理24h,培养结束后,收集细胞,加入1×loading(Biorad,Cat.1610747)。通过蛋白质印迹法(Western Blot)检测BTK(CST,Cat.8547S)和β-actin(CST,Cat.4970S)的表达,并计算BTK相对于内参β-actin的表达量,实验结果如表2所示。根据以下公式计算获得给药组BTK蛋白相对于溶媒组BTK蛋白的降解率(BTK%):
BTK%=(1-(BTK 给药/β-actin)/(BTK 溶媒/β-actin))×100%
BTK 给药为化合物不同剂量下Mino细胞中BTK的表达量,BTK 溶媒为对照组Mino细胞中BTK的表达量。本实验体系的化合物最高浓度为1000nM,D max为实验浓度体系内的降解最大值。并使用Graphpad5.0软件根据公式Y=100/(1+10^((LogIC 50-X)*HillSlope))拟合出DC 50值。
表2:化合物对BTK(WT)以及BTK(C481S)降解
Figure PCTCN2022128411-appb-000143
Figure PCTCN2022128411-appb-000144
备注:A<10nM;10nM<B<1000nM;1000nM<C;‘-’表示在测试体系范围内化合物的降解率小于31%,‘ND’表示该项数据未测。
1.2.2细胞水平IKZF1降解实验
IKZF1蛋白是度胺类化合物降解的靶蛋白底物之一,通过IKZF1蛋白降解实验来判断化合物的IMiD活性强弱。检测上述BTK蛋白降解的Mino细胞样品,利用蛋白质免疫印迹方法同步检测IKZF1(CST,Cat.14859S)蛋白。根据以下公式计算获得给药组BTK蛋白相对于溶媒组BTK蛋白的降解率(BTK%):
IKZF1%=(1-(IKZF1 给药/β-actin)/(IKZF1 溶媒/β-actin))×100%
IKZF1 给药为化合物不同剂量下Mino细胞中IKZF1的表达量,IKZF1 溶媒为对照组Mino细胞中IKZF1的表达量。本实验体系的化合物最高浓度为1000nM,D max为实验浓度体系内的降解最大值,并使用Graphpad5.0软件根据公式Y=100/(1+10^((LogIC 50-X)*HillSlope))拟合出DC 50值。
表3:化合物对IKZF1的降解作用
Figure PCTCN2022128411-appb-000145
Figure PCTCN2022128411-appb-000146
备注:A<10nM;10nM<B<1000nM;1000nM<C。‘-’表示在测试体系范围内化 合物的降解率小于31%,‘ND’表示该项数据未测试。
表4:部分化合物对IKZF1的降解作用
Figure PCTCN2022128411-appb-000147
效果:由表1-4可知,本发明的化合物对BTK(WT)、BTK(C481S)有较好的降解活性,且在保留对BTK、BTK(C481S)具有较好降解活性的同时,对IKZF1的降解活性较弱,例如实施例20和25-39所示的化合物,尤其是实施例29和33所示的化合物。
1.2.3细胞增殖抑制实验
利用BTK靶点敏感细胞OCI-LY10来评价BTK降解导致的细胞增殖抑制作用,利用IMiD活性敏感细胞MM.1S评估化合物对IMiD活性导致的细胞增殖抑制作用。将OCI-LY10和MM.1S细胞分别收集后,离心,去上清,用2mL完全培养基重悬,计数后调整细胞密度为7.40×10 6个/mL;以80μL孔接种于96孔板中,即20000个/孔细胞;将稀释好的化合物以20μL/孔加入到细胞96孔板中,最终起始最高浓度为20μM(DMSO终浓度为0.2%),将OCI-LY10细胞板在37℃,5%CO 2培养箱中培养3天后;MM.1S细胞板在37℃,5%CO 2培养箱中培养7天后,使用CellTiter 96 Aqueous Non-Radioactive细胞增殖试剂盒(CCK8)进行检测。每孔加入10μL CCK8试剂,轻拍板子让其混匀,放入培养箱中孵育,通过酶标仪检测450nm和650nm的吸光度,直到 DMSO处理组吸光度数值(OD 450-OD 650)达到0.6-0.8时读板检测。利用细胞活性率的计算公式:细胞活性率(%)=(化合物孔数值-培养基孔数值)/(DMSO孔数值-培养基孔数值)×100%,使用Graphpad Prism 5.0软件根据公式Y=100/(1+10^((LogIC 50-X)*HillSlope))拟合出IC 50值。
表5:化合物对OCI-LY10和MM.1S的增殖抑制活性
Figure PCTCN2022128411-appb-000148
Figure PCTCN2022128411-appb-000149
备注:‘ND’表示该项数据未测
效果:由表1-5可知,本发明的化合物对BTK敏感的OCI-LY10细胞有较好的增殖抑制活性,且对IMiD敏感的MM.1S的抑制活性较弱,例如实施例12,28,29,32,34,35,36,38和39所示的化合物。
1.3化合物的药代动力学分析
1.3.1化合物肝微粒体稳定性实验
肝微粒体稳定性分析是采用提取的肝脏细胞的微粒体来研究化合物的代谢稳定性。在反应缓冲液0.1M Tris,5mM MgCl 2,0.005%BSA,pH7.4与1μM终浓度测试化合物中分别与不同种属的肝微粒体(终浓度为0.33mg/mL)在37℃条件下孵育10min。随后加入终浓度1mM NADPH启动反应。分别在0、7、17、30和60min采样,加入4℃冷甲醇终止反应,4000rpm,5min离心收集样品后,用LC-MS/MS分析样品中药物含量。将Log(compound remaining percetage)与孵育时间进行线性回归,算出斜率-k,并通过以下公式计算相关代谢稳定性参数:
T half=-0.693/k
Intrinsic clearance:
Figure PCTCN2022128411-appb-000150
In vivo clearance:
Figure PCTCN2022128411-appb-000151
Hepatic clearance:
Figure PCTCN2022128411-appb-000152
Metabolic bioavailability:
Figure PCTCN2022128411-appb-000153
With P: microsomal protein concentration(mg/mL)
Houston: Houston factor(45 mg of microsomal protein/g liver)
LW: liver weight(g)(per species)
HBF: hepatic blood flow(mL/min)(per species)
fu: unbound fraction(fu=1 is generally used)
表6化合物在不同种属肝微粒体中代谢稳定性
Figure PCTCN2022128411-appb-000154
Figure PCTCN2022128411-appb-000155
备注:代谢稳定性判定标准:MF%>70%,代谢稳定性较好;30%<MF%<70%,代谢稳定性中等;MF%<30%,代谢稳定性较差。
效果:由表6可知,本发明的部分化合物对小鼠肝微粒体代谢稳定性较好,如实施例13、17、25、28、29、32等所示;部分化合物对比格犬肝微粒体代谢稳定性较好,如实施例17、25、28、29等化合物;部分化合物对人肝微粒体代谢稳定性较好,如实施例29等化合物。
1.3.2.小鼠药代动力学测试
本试验通过单次口服给予受试化合物于ICR小鼠,测试不同时间点的小鼠血浆中受试化合物的浓度,评价受试化合物在小鼠体内口服代谢特征。每个化合物采用3只ICR小鼠开展试验,小鼠口服给药剂量为10mg/kg,溶媒为:10%solutol,10%DMSO,80%生理盐水,给药后分别于5min,15min,30min,1h,2h,4h,6h,24h共8个时间点采血。每个时间点每只动物经眼球后静脉丛取血50μL,置EDTA抗凝试管中(EDTA:血=1:10),弹匀,置冰上,30min内离心,12000g 4℃离心3min,取上清,-20℃保存。用LC-MS/MS分析样品中药物含量。
表7:化合物口服吸收的药代动力学参数
Figure PCTCN2022128411-appb-000156
Figure PCTCN2022128411-appb-000157
结论:运用本发明技术合成的化合物在小鼠体内具有一定的口服吸收。
讨论
综上所述,可以看出本申请化合物对BTK、以及BTK(C481)激酶具有较好的抑制作用,在Mino细胞和HEK293过转BTK(C481S)稳转细胞中,对BTK(WT)以及BTK(C481S)具有较好的降解率。特别值得注意的是,相较于不含F取代的part K和其他连接基的化合物,含有F取代的part K(特别是
Figure PCTCN2022128411-appb-000158
)和连接基为
Figure PCTCN2022128411-appb-000159
的本发明化合物具有优异的BTK、BTK(C481S)降解活性,同时对IKZF1的降解活性较弱,并且对BTK敏感的OCI-LY10细胞有较好的增殖抑制活性,且对IMiD敏感的MM.1S的抑制活性较弱。此外,含有F取代的part K(特别是
Figure PCTCN2022128411-appb-000160
)和连接基为
Figure PCTCN2022128411-appb-000161
的本发明化合物对动物(尤其是人)肝微粒体代谢具有较好稳定性以及优异的药代动力学性质。
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。

Claims (17)

  1. 一种式I所示的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,
    Figure PCTCN2022128411-appb-100001
    其中,
    Z 1选自:N或CR 1
    Z 2选自:N或CR 1
    Z 3选自:N或CR 7
    Z 4选自:N或CR 7
    Z 8选自:N或CH;
    W 1选自:-O-、-S-、-NR b-、-CONR b-、-NR bCO-、-(CH 2) nNR b-或-(CH 2) nNR bCO-;
    A 1选自:键、C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基、-(CH 2) pR c;其中,R c选自:C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基,其中,所述的杂环基、杂芳基含有1到4个选自O、S、N的杂原子;其中,所述C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基任选地被1-4个R a取代;
    AL 1、AL 2和AL 3各自独立地选自:键、-O-、-S-、-NR' b-、-(CH 2) n'-、-CO-、-(CH 2) n'NR' b-、-CONR' b-、-NR' bCO-、-(CH 2) n'NR' bCO-;
    A 2和A 3各自独立地选自:键、C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基,其中,所述的杂环基、杂芳基含有1到4个选自O、S、N的杂原子;其中,所述C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基任选地被1-4个R a取代;
    W 2选自:键、-O-、-S-、-NR” b-、-(CH 2) n”-、-CONR” b-、-NR” bCO-、-(CH 2) n”NR” bCO-;
    R b、R' b和R” b各自独立地选自:H、C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-10元杂芳基;
    R 1、R 2、R 3、R 4、R 5和R 6各自独立地选自:H、卤素、氰基、羟基、氨基、羧基、酰胺基、酯基、C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-15元杂芳基;其中,所述C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环 基、C6-C10芳基、5-15元杂芳基任选地被1-4个R a取代;
    R 7各自独立地选自:H、卤素、氰基、羟基、氨基、羧基、酰胺基、酯基、C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-15元杂芳基;其中,所述C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-15元杂芳基任选地被1-4个Ra取代;优选地,R 7各自独立地选自:-H、-F、-Cl、-Br、-I、-OH、-NH 2、-CN、-OMe、-Me、-CH 2OH、-CO 2H、-CF 3、-CHF 2、-CONH 2;更优选地,R 7各自独立地选自:-H、-F、-OMe、-Me、-CF 3或-CHF 2
    或者,相邻的两个R 7以及与其连接的环原子共同形成C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基,优选地为C5-C6环烷基、5-6元杂环基、苯基、5-6元杂芳基;其中,所述环烷基、杂环基、芳基、苯基、杂芳基任选地被1-4个R a取代;
    或者,
    Figure PCTCN2022128411-appb-100002
    为环A 4,其中,任意一个与W 2相邻的R 7、W 2以及与它们连接的环原子形成环,环A 4选自:8-16元双环、三环或多环体系,优选地为9-14元的双环或三环体系;所述体系任选地被1-4个R a取代;任选地,其余R 7中,相邻的两个R 7以及与其连接的环原子共同形成C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基,优选地为C5-C6环烷基、5-6元杂环基、苯基、5-6元杂芳基;其中,所述环烷基、杂环基、芳基、苯基、杂芳基任选地被1-4个R a取代;
    或者,
    Figure PCTCN2022128411-appb-100003
    为环A 5,其中,任意一个与AL 3相邻的R 7、AL 3以及与它们连接的环原子形成环,环A 5选自:8-16元双环、三环或多环体系,优选地为9-14元的双环或三环体系;所述体系任选地被1-4个R a取代;任选地,其余R 7中,相邻的两个R 7以及与其连接的环原子共同形成C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基,优选地为C5-C6环烷基、5-6元杂环基、苯基、5-6元杂芳基;其中,所述环烷基、杂环基、芳基、苯基、杂芳基任选地被1-4个R a取代;
    或者,
    Figure PCTCN2022128411-appb-100004
    为环A 6,其中,任意一个与W 2相邻的R 7、W 2以及与它们连接的环原子形成环,任意一个与AL 3相邻的R 7、AL 3以及与它们连接的环原子形成环,环A 6选自:8-16元三环或多环体系,优选地为9-14元三环体系;所述体系任选地被1-4个R a取代;任选地,其余R 7中,相邻的两个R 7以及与其连接的环原子共同形成C3-C20环烷基、3-20元杂环基、C6-C10芳基、5-15元杂芳基,优选地为C5-C6环烷基、5-6元杂环基、苯基、5-6元杂芳基;其中,所述环烷基、杂环基、芳基、苯基、杂芳基任选地被1-4个R a取代;
    R 8选自:H、C1-C6烷基、
    Figure PCTCN2022128411-appb-100005
    Figure PCTCN2022128411-appb-100006
    上述-CH 2-中的H任选地被R a取代;
    R a选自:卤素、氰基、羟基、氨基、羧基、酰胺基、酯基、氧代基(=O)、C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-10元杂芳基;其中,所述C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-10元杂芳基任选地进一步被1-4个选自下组的基团取代:卤素、氰基、羟基、氨基、羧基、酰胺基、酯基、氧代基(=O)、C1-C6烷基、C1-C6烷氧基;
    p、n、n'和n”各自独立地选自:1、2、3、4或5;
    n 1、n 2、n 3、n 4、n 5、n 6和n 7各自独立地选自:0、1、2、3、4或5。
  2. 如权利要求1所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,所述化合物具有式II-IV所示的结构
    Figure PCTCN2022128411-appb-100007
    Figure PCTCN2022128411-appb-100008
    其中,
    R 13独立地选自:卤素、氰基、羟基、氨基、羧基、酰胺基、酯基、氧代基(=O)、C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-10元杂芳基;其中,所述C1-C6烷基、C1-C6烷氧基、C1-C6烷基、C1-C6烷基、C3-C6环烷基、4-6元杂环基、C6-C10芳基、5-10元杂芳基任选进一步被1-4个选自下组的基团取代:卤素、氰基、羟基、氨基、羧基、酰胺基、酯基、氧代基(=O)、C1-C6烷基、C1-C6烷氧基;
    n 13选自:0、1、2、3或4;
    W 1、W 2、A 1、A 2、A 3、环A 4、环A 5、环A 6、Z 1、Z 2、Z 8、AL 1、AL 2、AL 3、R 1、R 2、R 3、R 4、R 5、R 6、R 8、n 1、n 2、n 3、n 4、n 5和n 6的定义如权利要求1中所述;
    特别地,R 13各自独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、OMe、Me、CH 2OH、CO 2H、CF 3、CHF 2或CONH 2
  3. 如权利要求1或2所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,
    A 1选自:键、C4-C7环烷基、5元氧杂环烷、6元氧杂环烷、5元氮杂环烷、6元氮杂环烷、7元氮杂环烷基、氧代氮杂环庚烷基、苯基、吡唑基、吡啶基、吡嗪基、苄基、C7-C15螺环基、C6-C15并环基、C5-C15桥环基、C5-C15杂螺环基、C4-C15杂并环基、C5-C15杂桥环基;其中,所述的C5-C15杂螺环基、C4-C15杂并环基、C5-C15杂桥环基含有1到4个选自O、S、N的杂原子;优选地,A1选自:4元氮杂环烷基、5元氮杂环烷基、6元氮杂环烷基、7元氮杂环烷基、环丁基、环戊基、环己基、环庚基、四氢呋喃基、四氢吡喃基、吗啡啉基、哌啶基、哌嗪基、吡咯烷基、螺环[3.3]庚烷基、2-氮杂螺环[3.3]庚烷基、2,6-二氮螺环[3.3]庚烷基、环戊烷并环戊烷基、环戊烷并四氢吡咯基、2-氮杂双环[2.2.1]庚烷基、6-氮杂螺环[3.4]辛烷基、2-氮杂螺环[3.4]辛烷基、7-氮杂螺环[3.5]壬烷基、2-氮杂螺环[4.4]壬烷基、2-氮杂螺环[3.5]壬烷基、双环[1.1.1]戊烷基、苯基、吡啶基、吡嗪基、吡唑基、苯并四氢吡咯基、苯并哌啶基、苯并吗啡 啉基、苯并哌嗪基、吡啶并四氢吡咯基、吡啶并哌啶基、吡啶并吗啡啉基、吡啶并哌嗪基、吡嗪并四氢吡咯基、吡嗪并哌啶基、吡嗪并吗啡啉基、吡嗪并哌嗪基、2,7-二氮螺环[4.4]壬烷基、2,7-二氮螺环[3.5]壬烷基、2,6-二氮螺环[3.4]辛烷基或键;更优选地,A 1选自:
    Figure PCTCN2022128411-appb-100009
    Figure PCTCN2022128411-appb-100010
    Figure PCTCN2022128411-appb-100011
    其中,上述基团任选地被1-4个R a取代,R a的定义如权利要求1中所述;
    和/或,part K选自:
    Figure PCTCN2022128411-appb-100012
    Figure PCTCN2022128411-appb-100013
  4. 如权利要求1所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,所述化合物具有式V-1或V-2所示的结构
    Figure PCTCN2022128411-appb-100014
    Figure PCTCN2022128411-appb-100015
    其中,
    A 1选自:键、3-20元环烷基、3元杂环基、4元杂环基、7-20元杂环基、C6-C10芳基、7-15元杂芳基;其中,所述的杂环基、杂芳基含有1到4个选自O、S、N的杂原子;其中,所述3-20元环烷基、3元杂环基、4元杂环基、7-20元杂环基、C6-C10芳基、7-15元杂芳基任选地被1-4个R a取代;
    W 1、A 1、A 2、A 3、Z 1、Z 2、AL 1、AL 2、R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、n 1、n 2、n 3、n 4、n 5、n 6和n 7的定义如权利要求1中所述;
    特别地,A 1选自:键、C4-C7环烷基、7元氮杂环烷基、氧代氮杂环庚烷基、苄基、C7-C15螺环基、C6-C15并环基、C5-C15桥环基、C7-C15杂螺环基、C4杂并环基、C7-C15杂并环基、C7-C15杂桥环基;其中,所述的C7-C15杂螺环基、C4杂并环基、C7-C15杂并环基、C7-C15杂桥环基含有1到4个选自O、S、N的杂原子;优选地,A 1选自:4元氮杂环烷基、7元氮杂环烷基、环丁基、环戊基、环己基、环庚基、螺环[3.3]庚烷基、2-氮杂螺环[3.3]庚烷基、2,6-二氮螺环[3.3]庚烷基、环戊烷并环戊烷基、环戊烷并四氢吡咯基、2-氮杂双环[2.2.1]庚烷基、6-氮杂螺环[3.4]辛烷基、2-氮杂螺环[3.4]辛烷基、7-氮杂螺环[3.5]壬烷基、2-氮杂螺环[4.4]壬烷基、2-氮杂螺环[3.5]壬烷基、双环[1.1.1]戊烷基、苯并四氢吡咯基、苯并哌啶基、苯并吗啡啉基、苯并哌嗪基、吡啶并四氢吡咯基、吡啶并哌啶基、吡啶并吗啡啉基、吡啶并哌嗪基、吡嗪并四氢吡咯基、吡嗪并哌啶基、吡嗪并吗啡啉基、吡嗪并哌嗪基、2,7-二氮螺环[4.4]壬烷基、2,7-二氮螺环[3.5]壬烷基、2,6-二氮螺环[3.4]辛烷基或键;
    更特别地,A 1选自:
    Figure PCTCN2022128411-appb-100016
    Figure PCTCN2022128411-appb-100017
    Figure PCTCN2022128411-appb-100018
    其中,上述基团任选地被1-4个R a取代,R a的定义如权利要求1所述。
  5. 如权利要求1-4中任一项所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,所述化合物满足下述条件中的一个或多个,
    (1)相邻的两个R 7以及与其连接的环原子共同形成环时,
    Figure PCTCN2022128411-appb-100019
    选自:
    Figure PCTCN2022128411-appb-100020
    Figure PCTCN2022128411-appb-100021
    (2)环A 4选自:
    Figure PCTCN2022128411-appb-100022
    Figure PCTCN2022128411-appb-100023
    (3)环A 5选自:
    Figure PCTCN2022128411-appb-100024
    (4)环A 6选自:
    Figure PCTCN2022128411-appb-100025
    (5)R 1、R 2、R 3、R 4、R 5和R 6各自独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、OMe、Me、CH 2OH、CO 2H、CF 3、CHF 2或CONH 2
    (6)Z 1为CH;
    (7)Z 2为CH;
    (8)W 1选自:-O-、-S-、-NH-、-CONH-、-NHCO-或-CH 2NHCO-;
    (9)W 2选自:-O-、-S-、-NH-、-CH 2-、-CONH-、-NHCO-、-CH 2NHCO-或键;
    (10)AL 1、AL 2和AL 3各自独立地选自:-O-、-S-、-NH-、-CH 2-、-CO-、-CH 2NH-、-CH 2N(CH 3)-、-CH 2CH 2-、-CONH-、-NHCO-、-CH 2NHCO-或键;
    (11)A 2和A 3各自独立地选自:键、3-12元杂环基、C3-C12环烷基、5-15元杂芳基、5-10元芳基或键,其中,所述的杂环基、杂芳基含有1到4个选自O、S、N的杂原子;优选地,A 2和A 3选自:4元氮杂环烷基、5元氮杂环烷基、6元氮杂环烷基、7元氮杂环烷基、环丁 基、环戊基、环己基、环庚基、四氢呋喃基、四氢吡喃基、吗啡啉基、哌啶基、哌嗪基、吡咯环基、螺环[3.3]庚烷基、2-氮杂螺环[3.3]庚烷基、2,6-二氮螺环[3.3]庚烷基、环戊烷并环戊烷基、环戊烷并四氢吡咯基、2-氮杂双环[2.2.1]庚烷基、6-氮杂螺环[3.4]辛烷基、2-氮杂螺环[3.4]辛烷基、7-氮杂螺环[3.5]壬烷基、2-氮杂螺环[4.4]壬烷基、2-氮杂螺环[3.5]壬烷基、双环[1.1.1]戊烷基、苯基、吡啶基、吡嗪基、吡唑基、苯并四氢吡咯基、苯并哌啶基、苯并吗啡啉基、苯并哌嗪基、吡啶并四氢吡咯基、吡啶并哌啶基、吡啶并吗啡啉基、吡啶并哌嗪基、吡嗪并四氢吡咯基、吡嗪并哌啶基、吡嗪并吗啡啉基、吡嗪并哌嗪基、2,7-二氮螺环[4.4]壬烷基、2,7-二氮螺环[3.5]壬烷基、2,6-二氮螺环[3.4]辛烷基或键;更优选地,A 2和A 3各自独立地选自:
    Figure PCTCN2022128411-appb-100026
    Figure PCTCN2022128411-appb-100027
    其中,上述基团任选地被1-4个R a取代,R a的定义如权利要求1中所述;
    其中,特别地,
    Figure PCTCN2022128411-appb-100028
    Figure PCTCN2022128411-appb-100029
    或者,
    Figure PCTCN2022128411-appb-100030
    选自:
    Figure PCTCN2022128411-appb-100031
    Figure PCTCN2022128411-appb-100032
  6. 如权利要求1-5中任一项所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,part L选自:
    Figure PCTCN2022128411-appb-100033
    Figure PCTCN2022128411-appb-100034
  7. 如权利要求1-3中任一项所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,
    AL 1及AL 2均为键;
    或者,AL 1、AL 2及AL 3均为键;
    或者,
    Figure PCTCN2022128411-appb-100035
    选自
    Figure PCTCN2022128411-appb-100036
    Figure PCTCN2022128411-appb-100037
    Figure PCTCN2022128411-appb-100038
    Figure PCTCN2022128411-appb-100039
    优选地,上述各基团的右端与part K连接。
  8. 如权利要求1所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,AL 1及AL 2均为键,且所述化合物具有式II-1所示的结构,
    Figure PCTCN2022128411-appb-100040
    其中,A 1、A 2、A 3、R 3、n 3和partK的定义如权利要求1-3中所述;
    优选地,part K选自
    Figure PCTCN2022128411-appb-100041
    Figure PCTCN2022128411-appb-100042
  9. 如权利要求8所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,AL 1及AL 2均为键,且所述化合物具有式II-2所示的结构,
    Figure PCTCN2022128411-appb-100043
    其中,A 1、A 2、A 3、R 7、n 7、R 3和n 3的定义如权利要求1-3中所述。
  10. 如权利要求8或9所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,
    A 1
    Figure PCTCN2022128411-appb-100044
    优选地,上述各基团的右端与A 2连接;
    或者,A 2
    Figure PCTCN2022128411-appb-100045
    Figure PCTCN2022128411-appb-100046
    优选地,上述各基团的右端与A 3连接;
    或者,A 3
    Figure PCTCN2022128411-appb-100047
    或键;优选地,上述各基团的右端与part K连接;
    或者,
    Figure PCTCN2022128411-appb-100048
    为:
    Figure PCTCN2022128411-appb-100049
    Figure PCTCN2022128411-appb-100050
    Figure PCTCN2022128411-appb-100051
    优选地,上述各基团的右端与part K连接;
    或者,
    Figure PCTCN2022128411-appb-100052
    Figure PCTCN2022128411-appb-100053
    其中,R 3-1、R 3-2、R 3-3、R 3-4和R 3-5各自独立地选自H、卤素(氟、氯、溴或碘)、羟基、氨基、C1-C6烷基(例如甲基、 乙基、正丙基、异丙基、正丁基、异丁基或叔丁基)、C1-C6烷氧基(例如甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基或叔丁氧基)或C1-C6烷基氨基(例如甲基氨基、乙基氨基、正丙基氨基、异丙基氨基、正丁基氨基、异丁基氨基或叔丁基氨基);
    或者,R 3-1选自卤素,例如氟、氯、溴或碘,优选为氟;
    或者,R 3-2选自C1-C6烷氧基,优选为甲氧基;
    或者,R 3-3选自H;
    或者,R 3-4选自H;
    或者,R 3-5选自H;
    或者,R 3-1选自氟,R 3-2选自甲氧基,R 3-3选自H,R 3-4选自H,R 3-5选自H。
  11. 如权利要求1所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,所述化合物具有式VI-1或VI-2所示的结构,
    Figure PCTCN2022128411-appb-100054
    其中,
    R 7-1、R 7-2和R 7-3各自独立地选自H、卤素(氟、氯、溴或碘)、羟基、氨基、C1-C6烷基(例如甲基、乙基、正丙基、异丙基、正丁基、异丁基或叔丁基)、C1-C6烷氧基(例如甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基或叔丁氧基)或C1-C6烷基氨基(例如甲基氨基、乙基氨基、正丙基氨基、异 丙基氨基、正丁基氨基、异丁基氨基或叔丁基氨基);
    R 3-1、R 3-2、R 3-3、R 3-4和R 3-5各自独立地选自H、卤素(氟、氯、溴或碘)、羟基、氨基、C1-C6烷基(例如甲基、乙基、正丙基、异丙基、正丁基、异丁基或叔丁基)、C1-C6烷氧基(例如甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基或叔丁氧基)或C1-C6烷基氨基(例如甲基氨基、乙基氨基、正丙基氨基、异丙基氨基、正丁基氨基、异丁基氨基或叔丁基氨基);
    Z 1、Z 2、W 1、R 1、R 2、R 4、R 5、R 6、n 1、n 2、n 4、n 5、n 6的定义如权利要求1或4中所述;
    条件是:R 7-1、R 7-2、R 7-3、R 3-1、R 3-2、R 3-3、R 3-4和R 3-5不同时为H。
  12. 如权利要求11所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,所述化合物具有式VI-3或VI-4所示的结构,
    Figure PCTCN2022128411-appb-100055
    其中,
    R 7-1、R 7-2、R 7-3、R 3-1、R 3-2、R 3-3、R 3-4和R 3-5定义如权利要求11中所述;
    条件是:R 7-1、R 7-2、R 7-3、R 3-1、R 3-2、R 3-3、R 3-4和R 3-5不同时为H。
  13. 如权利要求11或12所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,
    R 7-1选自卤素,例如氟、氯、溴或碘,优选为氟;
    或者,R 7-2选自H;
    或者,R 7-3选自H;
    或者,R 7-1选自氟,R 7-2选自H,R 7-3选自H;
    或者,R 3-1选自卤素,例如氟、氯、溴或碘,优选为氟;
    或者,R 3-2选自C1-C6烷氧基,优选为甲氧基;
    或者,R 3-3选自H;
    或者,R 3-4选自H;
    或者,R 3-5选自H;
    或者,R 3-1选自氟,R 3-2选自甲氧基,R 3-3选自H,R 3-4选自H,R 3-5选自H。
  14. 如权利要求1所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,其特征在于,所述化合物选自:
    Figure PCTCN2022128411-appb-100056
    Figure PCTCN2022128411-appb-100057
    Figure PCTCN2022128411-appb-100058
    Figure PCTCN2022128411-appb-100059
    Figure PCTCN2022128411-appb-100060
    Figure PCTCN2022128411-appb-100061
    Figure PCTCN2022128411-appb-100062
    Figure PCTCN2022128411-appb-100063
    Figure PCTCN2022128411-appb-100064
    Figure PCTCN2022128411-appb-100065
    Figure PCTCN2022128411-appb-100066
    Figure PCTCN2022128411-appb-100067
    Figure PCTCN2022128411-appb-100068
    Figure PCTCN2022128411-appb-100069
  15. 一种药物组合物,其包含权利要求1-14中任一项所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,和一种或多种药学上可接受的载体或赋形剂。
  16. 一种如权利要求1-14中任一项所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,或如权利要求15所述的药物组合物在制备治疗BTK介导的疾病的药物中的用途;
    优选地,所述疾病选自:自体免疫疾病、炎性疾病或肿瘤;
    所述自体免疫疾病优选选自:狼疮、多发性硬化、肌肉缩性侧索硬化、类风湿性关节炎、银屑病、因器官移植导致的并发症、糖尿病、哮喘、特应性皮炎、自身免疫性甲状腺病、溃疡性结肠炎、克罗恩病、阿尔茨海默病、白血病或淋巴瘤;
    所述炎性疾病优选选自:角膜炎、鼻炎、口腔炎、腮腺炎、咽炎、扁桃体炎、气管炎、支气管炎、肺炎、心肌炎、胃炎、肠胃炎、胆囊炎或阑尾炎;
    所述肿瘤优选选自:小淋巴细胞淋巴瘤、急性淋巴细胞白血病、慢性淋巴细胞白血病、急性骨髓性白血病、慢性骨髓性白血病、急性早幼粒细胞白血病、慢性粒细胞白血病、弥漫性大B细胞淋巴瘤、血管内大B细胞淋巴瘤、原发性渗出性淋巴瘤、华氏巨球蛋白血症、滤泡性淋巴瘤、多发性骨髓瘤或套细胞淋巴瘤。
  17. 一种如权利要求1-14中任一项所述的化合物、其药学上可接受的盐、其顺反异构体、其手性异构体、其对映异构体、其非对映异构体、其同位素衍生物、其前药、其溶剂化物或其水合物,或如权利要求15所述的药物组合物在制备用于抑制或调节BTK蛋白激酶活性的药物中的用途;
    其中,所述BTK蛋白激酶优选为非突变的BTK蛋白激酶或突变的BTK蛋白激酶; 所述突变的BTK蛋白激酶优选为C481S突变的BTK蛋白激酶。
PCT/CN2022/128411 2021-10-29 2022-10-28 Protac化合物、含其的药物组合物及其制备方法和应用 WO2023072270A1 (zh)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202111272891.2 2021-10-29
CN202111272891 2021-10-29
CN202210126407.3 2022-02-10
CN202210126407 2022-02-10

Publications (1)

Publication Number Publication Date
WO2023072270A1 true WO2023072270A1 (zh) 2023-05-04

Family

ID=86159076

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/128411 WO2023072270A1 (zh) 2021-10-29 2022-10-28 Protac化合物、含其的药物组合物及其制备方法和应用

Country Status (1)

Country Link
WO (1) WO2023072270A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20230137175A1 (en) * 2021-08-17 2023-05-04 Endotarget Inc. Compounds and methods for the targeted degradation of bruton's tyrosine kinase
WO2024012570A1 (zh) * 2022-07-15 2024-01-18 西藏海思科制药有限公司 一种含氮杂环衍生物及其组合物和药学上的应用

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112010858A (zh) * 2019-05-31 2020-12-01 四川海思科制药有限公司 一种btk抑制剂及其制备方法和药学上的应用
WO2020239103A1 (zh) * 2019-05-31 2020-12-03 四川海思科制药有限公司 一种btk抑制剂环衍生物及其制备方法和药学上的应用
WO2021068380A1 (zh) * 2019-10-09 2021-04-15 清华大学 一种靶向btk蛋白降解化合物的制备及其在治疗自身免疫系统疾病与肿瘤中的应用
WO2022007824A1 (zh) * 2020-07-07 2022-01-13 四川海思科制药有限公司 一种具有降解btk激酶的化合物及其制备方法和药学上的应用
CN113999233A (zh) * 2020-07-28 2022-02-01 四川海思科制药有限公司 一种btk抑制剂环衍生物及其制备方法和药学上的应用
CN114507235A (zh) * 2020-11-17 2022-05-17 海思科医药集团股份有限公司 一种具有降解btk激酶的化合物及其制备方法和药学上的应用
CN115304606A (zh) * 2021-06-21 2022-11-08 清华大学 一种同时靶向btk和gspt1蛋白的降解剂
WO2022253250A1 (zh) * 2021-06-01 2022-12-08 正大天晴药业集团股份有限公司 含有并环或螺环的布鲁顿酪氨酸激酶降解剂

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112010858A (zh) * 2019-05-31 2020-12-01 四川海思科制药有限公司 一种btk抑制剂及其制备方法和药学上的应用
WO2020239103A1 (zh) * 2019-05-31 2020-12-03 四川海思科制药有限公司 一种btk抑制剂环衍生物及其制备方法和药学上的应用
WO2021068380A1 (zh) * 2019-10-09 2021-04-15 清华大学 一种靶向btk蛋白降解化合物的制备及其在治疗自身免疫系统疾病与肿瘤中的应用
WO2022007824A1 (zh) * 2020-07-07 2022-01-13 四川海思科制药有限公司 一种具有降解btk激酶的化合物及其制备方法和药学上的应用
CN113999233A (zh) * 2020-07-28 2022-02-01 四川海思科制药有限公司 一种btk抑制剂环衍生物及其制备方法和药学上的应用
CN114507235A (zh) * 2020-11-17 2022-05-17 海思科医药集团股份有限公司 一种具有降解btk激酶的化合物及其制备方法和药学上的应用
WO2022253250A1 (zh) * 2021-06-01 2022-12-08 正大天晴药业集团股份有限公司 含有并环或螺环的布鲁顿酪氨酸激酶降解剂
CN115304606A (zh) * 2021-06-21 2022-11-08 清华大学 一种同时靶向btk和gspt1蛋白的降解剂

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20230137175A1 (en) * 2021-08-17 2023-05-04 Endotarget Inc. Compounds and methods for the targeted degradation of bruton's tyrosine kinase
WO2024012570A1 (zh) * 2022-07-15 2024-01-18 西藏海思科制药有限公司 一种含氮杂环衍生物及其组合物和药学上的应用

Similar Documents

Publication Publication Date Title
ES2866152T3 (es) Derivados de tirosina amida como inhibidores de la Rho-quinasa
ES2768996T3 (es) Pirrolopirimidinas y pirazolopirimidinas utilizadas como inhibidores de la proteasa específica de ubiquitina 7
JP7073359B2 (ja) ユビキチン特異的プロテアーゼ7の阻害剤としてのピペリジン誘導体
WO2019158019A1 (zh) 嘧啶并环化合物及其制备方法和应用
EP2545045B1 (en) Piperidin-4-yl azetidine derivatives as jak1 inhibitors
EP3712151A1 (en) Spiro aromatic ring compound and application thereof
WO2023072270A1 (zh) Protac化合物、含其的药物组合物及其制备方法和应用
WO2020239103A1 (zh) 一种btk抑制剂环衍生物及其制备方法和药学上的应用
KR20220004100A (ko) 안드로겐 수용체를 표적 분해하는 이중 기능성의 키메라 헤테로 고리 화합물 및 이의 용도
BR112020019264A2 (pt) Compostos para tratar doença de huntington
BR112020011979A2 (pt) compostos de indol substituídos por amida úteis como inibidores de tlr
CN111819176A (zh) 4-氮杂吲哚化合物
EP3947375B1 (en) Imidazolonylquinoline compounds and therapeutic uses thereof
WO2022134641A1 (zh) 芳香杂环类化合物、药物组合物及其应用
WO2022117075A1 (zh) 氮杂并环化合物、其制备方法及其用途
WO2022253309A1 (zh) 取代的杂环化合物及其应用
WO2023227080A1 (zh) 一种protac化合物、含其的药物组合物及其制备方法和应用
WO2024044570A1 (en) Compounds and methods for modulating her2
WO2024017372A1 (zh) 一种吲哚酮衍生物及其应用
WO2024083256A1 (zh) pan-KRAS降解剂及其制备方法和应用
CN115557946A (zh) 杂环内酰胺类化合物,包含其的药物组合物及其用途
WO2024084448A2 (en) Fused bicyclic compounds and their use as mer and axl inhibitors
WO2024067691A1 (zh) 含氮杂环类化合物及其医药用途
WO2023046030A1 (zh) 一种egfr小分子抑制剂、含其的药物组合物及其用途
TW202409039A (zh) 依莫帕米結合蛋白抑制劑及其用途

Legal Events

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

Ref document number: 22886145

Country of ref document: EP

Kind code of ref document: A1