WO2023072273A1 - 作为cbl-b抑制剂的并环化合物 - Google Patents

作为cbl-b抑制剂的并环化合物 Download PDF

Info

Publication number
WO2023072273A1
WO2023072273A1 PCT/CN2022/128426 CN2022128426W WO2023072273A1 WO 2023072273 A1 WO2023072273 A1 WO 2023072273A1 CN 2022128426 W CN2022128426 W CN 2022128426W WO 2023072273 A1 WO2023072273 A1 WO 2023072273A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
cycloalkyl
methyl
optionally substituted
membered
Prior art date
Application number
PCT/CN2022/128426
Other languages
English (en)
French (fr)
Inventor
麦万笋
刘晓武
施文强
邓志雄
祝伟
李正涛
邹昊
Original Assignee
先声再明医药有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 先声再明医药有限公司 filed Critical 先声再明医药有限公司
Publication of WO2023072273A1 publication Critical patent/WO2023072273A1/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
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • Patent application No. 202111271704.9 submitted to the State Intellectual Property Office of China on October 29, 2021;
  • Patent application No. 202210103242.8 submitted to the State Intellectual Property Office of China on January 27, 2022;
  • Patent application No. 202210899553.X filed with the State Intellectual Property Office of China on July 28, 2022;
  • Patent application No. 202211063507.2 submitted to the State Intellectual Property Office of China on September 1, 2022.
  • the present application relates to a compound or a stereoisomer or a pharmaceutically acceptable salt thereof as a Cbl-b inhibitor, a preparation method thereof, a pharmaceutical composition containing the compound or a stereoisomer or a pharmaceutically acceptable salt thereof, And the use of said compound or its stereoisomer or its pharmaceutically acceptable salt or said pharmaceutical composition in preventing or treating diseases or diseases mediated by Cbl-b.
  • Intracellular signal cascades are usually regulated by protein phosphorylation, and gene expression and regulation are affected by epigenetics, and the proteins that regulate these signals are themselves subject to the "production-degradation" balance of intracellular proteins regulation to maintain cellular homeostasis.
  • protein degradation mainly through two pathways: lysosomal degradation pathway and ubiquitin-mediated proteasomal degradation pathway.
  • the ubiquitin-mediated pathway is a specific protein degradation pathway subject to strict spatiotemporal regulation.
  • the ubiquitin system widely exists in eukaryotes and is a precise regulatory system for intracellular protein degradation.
  • Ubiquitin (Ub) is a highly conserved small protein present in most eukaryotic cells.
  • the ubiquitin system consists of ubiquitin, 26S proteasome, and various enzymes (E1, E2, E3, and deubiquitinase, etc.).
  • the ubiquitination of protein is through the E1-E2-E3 level involving ubiquitin activating enzyme E1 (ubactivating enzyme, E1), ubiquitin conjugating enzyme E2 (Ub conjugating enzyme, E2), ubiquitin ligase E3 (Ub ligase, E3)
  • E1 activating enzyme
  • E2 Ub conjugating enzyme
  • E3 ubiquitin ligase
  • the cascade reaction is completed, and then the ubiquitinated protein is degraded by the 26S proteasome (A patent review of the ubiquitin ligase system:2015-201 Expert Opin Ther Pat.2018,28(12):919–937).
  • the human genome encodes approximately 35 E2-conjugating enzymes and more than 500 E3 ligases.
  • Casitas B-lineage lymphoma proto-oncogene-b (Cbl-b) is an E3 ligase that negatively regulates T cell activation.
  • Cbl-b belongs to the Cbl family, which includes c-Cbl, Cbl-b and Cbl-3 (Cbl:many adaptations to regulate protein tyrosine kinases.Nat.Rev.Mol.Cell Biol.2001,2(4):294– 307).
  • Cbl protein mainly negatively regulates T cell activation, growth factors (such as epidermal growth factor receptor (EGFR), c-KIT, platelet-derived growth factor receptor (PDGFR) and non-receptor tyrosine kinase signaling (such as Src family kinases and Zap70) (Regulating the regulator:Negative regulation of Cbl ubiquitin ligases.Trends Biochem Sci,2006,31(2):79–88; The Cbl family proteins:Ring leaders in regulation of cell signaling.J Cell Physiol.2006,2 09 :21–43).
  • growth factors such as epidermal growth factor receptor (EGFR), c-KIT, platelet-derived growth factor receptor (PDGFR) and non-receptor tyrosine kinase signaling (such as Src family kinases and Zap70)
  • EGFR epidermal growth factor receptor
  • c-KIT platelet-derived growth factor receptor
  • PDGFR plate
  • Cbl protein is related to human cancer (Germline CBL mutations cause developmental abnormalities and predispose to juvenile myelomonocytic leukemia. Nat Genet, 2010,42(9):794–800).
  • Cbl-b plays a key role in establishing T cell activation thresholds and controlling peripheral T cell tolerance. Accumulating evidence indicates that Cbl-b also regulates innate immune responses and plays a role in host defense against pathogens. Play an important role.
  • Cbl-b gene knockout mice show severe autoimmune diseases (Negative regulation of lymphocyte activation and autoimmunity by the molecular adapter Cbl-b.Nature,2000,403:211–216). Therefore, Cbl -b can serve as an important immunomodulatory therapeutic target.
  • the application relates to a compound of formula (I) or a stereoisomer or a pharmaceutically acceptable salt thereof,
  • a 1 -A 2 -A 3 said A 1 and A 2 are independently selected from C(R 11 ) n or N, A 3 is selected from CR 11a R 11b , NR 12 , O or S;
  • n is selected from 0 or 1;
  • R 11a , R 11b , R 11c , R 11 , R 12 are independently selected from H, halogen, OH, CN, NH 2 , NH(C 1 -C 6 alkyl), N(C 1 -C 6 alkyl ) 2 , C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 6 cycloalkyl or C 3 -C 6 cycloalkyloxy, wherein the C 1 -C 6 alk Base, C 1 -C 6 alkoxy, C 3 -C 6 cycloalkyl or C 3 -C 6 cycloalkyloxy are optionally substituted by R 11d ;
  • Q ring is selected from phenyl, 5-6 membered heteroaryl or 5-7 membered heterocyclic group, the phenyl, 5-6 membered heteroaryl or 5-7 membered heterocyclic group is optionally substituted by R 10 ;
  • Y 1 , Y 2 , Y 3 and Y 4 are independently selected from CR b or N;
  • R b is selected from H, halogen, OH, CN, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 alkylthio, NH 2 , NH(C 1 -C 6 alkyl ), N(C 1 -C 6 alkyl) 2 , NHC(O)(C 1 -C 6 alkyl), NHC(O)-O(C 1 -C 6 alkyl), N(C 1 -C 6 alkyl) C(O)-O(C 1 -C 6 alkyl), NHS(O) 2 (C 1 -C 6 alkyl), C 3 -C 6 cycloalkyl, C 3 -C 6 ring Alkyl-O-, C 3 -C 6 cycloalkyl-NH-, N(C 3 -C 6 cycloalkyl) 2 , NHC(O)-C 3 -C 6 cycloalkyl, NHS(O) 2 -C 3
  • R b and the C atoms connected to them jointly form a C 3 -C 6 cycloalkenyl group, a phenyl group, a 4-7 membered heterocyclic group or a 5-6 membered heteroaryl group, and the C 3 -C 6 cycloalkenyl group , phenyl, 4-7 membered heterocyclyl or 5-6 membered heteroaryl are optionally substituted by R 2a ;
  • R 4 , R 5 , R 7 , R 8 and R 9 are independently selected from H, halogen, OH, C 1 -C 6 alkyl or C 1 -C 6 alkoxy, and the C 1 -C 6 alkane or C 1 -C 6 alkoxy is optionally substituted by R 4a ;
  • R 8 , R 9 and their connected atoms together form a C 3 -C 6 cycloalkyl or a 4-7 membered heterocyclic group, and the C 3 -C 6 cycloalkyl or 4-7 membered heterocyclic group is optionally further substituted by R 8a ;
  • R 4 , R 5 and their connected atoms together form a C 3 -C 6 cycloalkyl or a 4-7 membered heterocyclic group, and the C 3 -C 6 cycloalkyl or 4-7 membered heterocyclic group is optionally is further substituted by R 8a ; or when p is taken from 2, two R 4 and the atoms connected to them together form a C 3 -C 6 cycloalkyl or a 4-7 membered heterocyclic group, the C 3 -C 6 ring Alkyl or 4-7 membered heterocyclyl is optionally further substituted by R 8a ;
  • Each R 6a is independently selected from H, C 1 -C 6 alkyl, phenyl, 4-7 membered heterocyclic group or 5-6 membered heteroaryl, said C 1 -C 6 alkyl, phenyl, A 4-7 membered heterocyclic group or a 5-6 membered heteroaryl group is optionally further substituted by R 6b , or two R 6a on one N atom form a 4-7 membered heterocyclic group or a 5-6 Member heteroaryl, the 4-7 member heterocyclic group or 5-6 member heteroaryl is optionally further substituted by R 6b ;
  • R 1 and R 2 are independently selected from H, halogen, CN, NH 2 , NH(C 1 -C 6 alkyl), N(C 1 -C 6 alkyl) 2 , C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 10 cycloalkyl or 4-10 membered heterocyclic group, wherein said C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 10 Cycloalkyl or 4-10 membered heterocyclyl is optionally substituted by R 1a ,
  • the group is optionally substituted by R 1d ;
  • W is selected from (CR 13 R 14 ) k W 1 ; said W 1 is selected from 5-10 membered heteroaryl or 4-10 membered heterocyclic group, said 5-10 membered heteroaryl, 4-10 membered heterocyclic group
  • the ring group is optionally substituted by R 15 ;
  • R 13 and R 14 are independently selected from H, halogen, OH, C 1 -C 6 alkyl or C 1 -C 6 alkoxy;
  • R 15 is selected from halogen, OH, NH 2 , NH(C 1 -C 6 alkyl), N(C 1 -C 6 alkyl) 2 , C 1 -C 6 alkyl, C 3 -C 10 cycloalkyl or 4-7 membered heterocyclyl, wherein The C 1 -C 6 alkyl, C 3 -C 10 cycloalkyl or 4-7 membered heterocyclic group is optionally substituted by R 15a ;
  • the group is optionally substituted by R 13a ;
  • R 8a , R 13a and R 1d are independently selected from halogen, OH, CN, C 1 -C 6 alkyl or C 1 -C 6 alkoxy, said C 1 -C 6 alkyl or C 1 -C 6 Alkoxy is optionally further substituted by halogen;
  • k is selected from 0 or 1;
  • p is selected from 0, 1 or 2.
  • R b is selected from H, halogen, OH, CN, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, NH 2 , NH(C 1 -C 6 alkyl), N (C 1 -C 6 alkyl) 2 , NHC(O)(C 1 -C 6 alkyl), NHC(O)-O(C 1 -C 6 alkyl), N(C 1 -C 6 alkyl )C(O)-O(C 1 -C 6 alkyl), NHS(O) 2 (C 1 -C 6 alkyl), C 3 -C 6 cycloalkyl, C 3 -C 6 cycloalkyl- O-, C 3 -C 6 cycloalkyl-NH-, N(C 3 -C 6 cycloalkyl) 2 , NHC(O)-C 3 -C 6 cycloalkyl, NHS(O) 2 -C 3 -C 6 cycloalkyl,
  • C CA 3
  • a 3 is selected from CR 11a R 11b , NR 12 , O or S
  • a 3 is selected from CR 11c or N.
  • selected from A 1 -C A 3 , wherein A 1 is selected from C or N, and A 3 is selected from CR 11c or N.
  • selected from A 1 -C A 3 , wherein A 1 is selected from N, and A 3 is selected from CR 11c or N.
  • selected from C ⁇ CA 3 and said A 3 is selected from CR 11a R 11b , NR 12 , O or S.
  • selected from C ⁇ CA 3 and said A 3 is selected from NR 12 , O or S.
  • selected from C ⁇ CA 3 and said A 3 is selected from O.
  • a 1 -A 2 -A 3 selected from A 1 -A 2 -A 3 , wherein A 1 and A 2 are independently selected from C(R 11 ) n or N, and A 3 is selected from NR 12 , O or S.
  • a 1 -A 2 -A 3 selected from A 1 -A 2 -A 3 , wherein A 1 is selected from N, A 2 is selected from C(R 11 ) n , and A 3 is selected from NR 12 .
  • R 11a , R 11b , R 11c , R 11 , R 12 are independently selected from H, halogen, OH, NH 2 , NH(C 1 -C 6 alkyl), N(C 1 - C 6 alkyl) 2 , C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 6 cycloalkyl or C 3 -C 6 cycloalkyloxy, wherein said C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 6 cycloalkyl or C 3 -C 6 cycloalkyloxy is optionally substituted by R 11d .
  • R 11a , R 11b , R 11c , R 11 , R 12 are independently selected from H, halogen, OH, NH 2 , C 1 -C 6 alkyl or C 3 -C 6 cycloalkyl , the C 1 -C 6 alkyl or C 3 -C 6 cycloalkyl is optionally substituted by R 11d .
  • R 11a , R 11b , R 11c , R 11 , R 12 are independently selected from H, halogen or C 1 -C 6 alkyl.
  • R 11a , R 11b , R 11c , R 11 , R 12 are independently selected from H, F or methyl.
  • R 11d is selected from halogen, OH, NH 2 , C 1 -C 6 alkyl, or halogenated C 1 -C 6 alkyl.
  • n is selected from zero.
  • R 11c is selected from H, halogen, or C 1 -C 6 alkyl optionally substituted with R 11d .
  • R 11c is selected from H or halo.
  • R 11c is selected from F.
  • R 12 is selected from H, halogen, OH, C 1 -C 6 alkyl or C 3 -C 6 cycloalkyl, said C 1 -C 6 alkyl or C 3 -C 6 cycloalkane is optionally substituted by R 11d .
  • R 12 is selected from H or C 1 -C 3 alkyl.
  • R 12 is selected from H or methyl.
  • R 12 is selected from methyl.
  • selected from A 1 -C A 3 , wherein A 1 is selected from N, and A 3 is selected from N.
  • ring Q is selected from the group consisting of phenyl, pyridyl, pyrimidinyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isoxazolyl, Thiazolyl or a 5-7 membered heterocyclic group containing 1 or 2 N as heteroatoms.
  • ring Q is selected from the following groups optionally substituted by R 10 : phenyl,
  • ring Q is selected from the following groups optionally substituted by R 10 : phenyl,
  • ring Q is selected from the following groups optionally substituted by R 10 : phenyl,
  • the Q ring is selected from the group optionally substituted by R 10
  • ring Q is selected from phenyl optionally substituted with R 10 .
  • the NH 2 , C 1 -C 6 alkyl, C 2 -C 4 alkynyl, C 1 -C 6 alkoxy or C 3 -C 6 cycloalkyl are optionally substituted by R 10a .
  • R 10 is selected from halogen, NH 2 , CN, C 1 -C 6 alkyl, C 1 -C 6 alkoxy or C 3 -C 6 cycloalkyl, said NH 2 or C 1 -C 6 alkyl is optionally substituted by R 10a .
  • R 10 is selected from halogen, C 1 -C 6 alkyl optionally substituted with halogen, C 1 -C 6 alkoxy, or C 3 -C 6 cycloalkyl.
  • R 10a is selected from halogen, OH, NH 2 , C 1 -C 6 alkyl, halogenated C 1 -C 6 alkyl, C 1 -C 6 alkoxy, or halogenated C 1 -C 6 alkoxy.
  • R 10a is selected from halogen, OH, C 1 -C 6 alkyl, or halogenated C 1 -C 6 alkyl. In some embodiments, R 10a is selected from halogen or C 1 -C 6 alkyl.
  • R 10a is selected from F or CH 3 .
  • R 10 is selected from F, Cl, methyl, Br, ethyl, CF 3 , methoxy, or cyclopropyl.
  • Y 1 , Y 2 and Y 4 are independently selected from CR b or N, and Y 3 is selected from CR b .
  • Y 1 , Y 2 are independently selected from CR b or N, and Y 3 and Y 4 are independently selected from CR b .
  • Y 1 , Y 2 , Y 3 and Y 4 are all CR b .
  • Y 1 and Y 2 are both N, and Y 3 and Y 4 are independently selected from CR b .
  • Y 1 is N and Y 2 , Y 3 and Y 4 are independently selected from CR b .
  • Y 1 , Y 2 , and Y 3 are all CR b , and Y 4 is N.
  • Y 1 , Y 2 and Y 3 are all CH and Y 4 is CR b ; alternatively, Y 1 is N, Y 2 and Y 3 are both CH and Y 4 is CR b ; or, Y 1 , Y 2 , Y 3 are all CH and Y 4 is N.
  • R b is selected from H, halogen, CN, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 alkylthio, NH 2 , NH(C 1 - C 6 alkyl), C 3 -C 6 cycloalkyl, C 3 -C 6 cycloalkyl-O-, C 3 -C 6 cycloalkyl-NH-, 4-7 membered heterocyclyl, 4-7 Membered heterocyclyloxy, 4-7 membered heterocyclyl-NH-, 5-10 membered heteroaryl, 5-10 membered heteroaryloxy or 5-10 membered heteroaryl-NH-, wherein the C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 alkylthio, C 3 -C 6 cycloalkyl, 4-7 membered heterocyclic group or 5-10 membered heteroaryl The group is optionally substituted by R
  • R b is selected from H, halogen, OH, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 alkylthio, NH 2 , NH(C 1 - C 6 alkyl), N(C 1 -C 6 alkyl) 2 , C 3 -C 6 cycloalkyl-O-, C 3 -C 6 cycloalkyl-NH-, 4-7 membered heterocyclyl- O-, 4-7 membered heterocyclyl-NH- or 5-10 membered heteroaryl, wherein the C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 alkylthio A group, a C 3 -C 6 cycloalkyl group, a 4-7 membered heterocyclyl group or a 5-10 membered heteroaryl group are optionally substituted by R 2a .
  • R b is selected from H, halogen, OH, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, NH 2 , NH(C 1 -C 6 alkyl), N(C 1 -C 6 alkyl) 2 , C 3 -C 6 cycloalkyl-O-, C 3 -C 6 cycloalkyl-NH-, 4-7 membered heterocyclyl-O-, 4-7 membered heterocycle Group -NH- or 5-10 membered heteroaryl, wherein said C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 6 cycloalkyl, 4-7 membered heterocyclic Or 5-10 membered heteroaryl is optionally substituted by R 2a .
  • R b is selected from H, halogen, CN, or optionally substituted by R 2a from methyl, ethoxy, NHCH 3 , NHEt, NH(i-Pr), pyrazolyl, Cyclopropyl-O-, cyclobutyl-NH-, oxetanyl-O-, cyclopropyl, SCH 3 .
  • R b is selected from H, halogen, or the following groups optionally substituted by R 2a : methyl, ethoxy, NHCH 3 , NHEt, NH(i-Pr), pyrazolyl, cyclopropyl -O-, cyclobutyl-NH- or oxetanyl-O-.
  • R 2a is selected from halogen, OH, or C 1 -C 3 alkyl.
  • R 2a is selected from F, OH or methyl.
  • R b is selected from H, F, CN, CH 3 , CF 3 , OEt, NHCH 3 , NHEt, NH(i-Pr), SCH 3 ,
  • R b is selected from H, F, CF 3 , OEt, NHCH 3 , NHEt, NH(i-Pr),
  • R b is selected from H, F, CF 3 , CN, CH 3 , OEt, NHEt or SCH3 . In some embodiments, Rb is selected from H or CF3 .
  • R b , R 1 and their respective connected atoms and bonds together form a C 3 -C 6 cycloalkenyl or a 4-7 membered heterocyclyl, the C 3 -C 6 cycloalkyl or 4 -7-membered heterocyclyl is optionally substituted by R 1d .
  • R b , R 1 , and the atoms and bonds to which they are each attached together form a C 3 -C 6 cycloalkenyl optionally substituted with R 1d .
  • R 1d is selected from halogen, OH, C 1 -C 3 alkyl, or halo C 1 -C 3 alkyl.
  • R b , R 1 , and the atoms and bonds to which they are each attached together form cyclopentenyl.
  • X is selected from Or C 1 -C 6 alkyl optionally substituted by Re .
  • X is selected from
  • X is selected from
  • Ring B is selected from 5-10 membered nitrogen-containing heteroaryl, 4-7 membered monocyclic nitrogen-containing heterocyclyl, or 6-10 membered nitrogen-containing heterocyclyl optionally substituted by R.
  • ring B is selected from the following groups optionally substituted by R : tetrahydropyrrolyl, piperidinyl, piperazinyl, morpholinyl,
  • ring B is selected from the following groups optionally substituted by R : tetrahydropyrrolyl, piperidinyl, piperazinyl, morpholinyl,
  • ring B is selected from the following groups optionally substituted by R : tetrahydropyrrolyl, piperidinyl, piperazinyl, morpholinyl,
  • ring B is selected from the following groups optionally substituted by R : tetrahydropyrrolyl, piperidinyl, piperazinyl, morpholinyl,
  • R 3a is selected from F.
  • R 4a is selected from halogen, OH, or C 1 -C 3 alkoxy.
  • R 4a is selected from F or OH.
  • R 8a is selected from halogen, OH, or C 1 -C 3 alkyl.
  • p is selected from 1.
  • p is selected from zero.
  • p is selected from 2, and two R 4 and the atoms they are attached to together form a C 3 -C 6 cycloalkyl optionally substituted with R 8a .
  • p is selected from 2, and two R 4 and the atoms to which they are attached together form cyclopropyl or cyclobutyl.
  • ring D is selected from the following groups optionally substituted by R 6 : C 3 -C 6 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered monocyclic nitrogen-containing heterocyclyl, or 6-10 membered nitrogen-containing heterocyclic group, and the ring D is connected to L through a non-N atom.
  • ring D is selected from the following groups optionally substituted by R: cyclopropyl , cyclobutyl, cyclopentyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, Pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, triazolyl, thiazolyl, isothiazolyl or pyridyl.
  • ring D is selected from the following groups optionally substituted by R: cyclobutyl , cyclopentyl, piperidinyl, pyridinyl,
  • R6 is selected from F or methyl.
  • L is selected from a bond, -NR 7 -, -NR 7 CR 8 R 9 -, -O-, or -CR 8 R 9 -.
  • L is selected from a bond, -NR 7 -, -NR 7 CH 2 -, -O-, or -CR 8 R 9 -.
  • R 7 , R 8 and R 9 are independently selected from H, halogen, C 1 -C 3 alkyl, or OH.
  • R 7 , R 8 , and R 9 are independently selected from H, halogen, or C 1 -C 3 alkyl (eg, methyl).
  • R7 , R8 , and R9 are independently selected from H or halogen (eg, F).
  • R7 is selected from H.
  • R 8 , R 9 are selected from H, methyl or F.
  • L is selected from a bond, -NH-, -NHCH2- , -NHCH( CH3 )-, -O-, -C(F) 2- , or -CH2- .
  • L is selected from a bond, -NH-, -NHCH2- , -O-, -C(F) 2- , or -CH2- .
  • X is selected from C 1 -C 6 alkyl optionally substituted with Re .
  • R e is selected from halogen, OH, C 1 -C 3 alkyl, NH 2 , NH(C 1 -C 3 alkyl), N(C 1 -C 3 alkyl) 2 or C 1 -C 3 alkoxy, wherein said C 1 -C 3 alkyl or C 1 -C 3 alkoxy is further optionally substituted by R f .
  • R e is selected from C 1 -C 3 alkyl, N(C 1 -C 3 alkyl) 2 or C 1 -C 3 alkoxy, wherein said C 1 -C 3 alkyl Or C 1 -C 3 alkoxy is further optionally substituted by R f .
  • R f is selected from halogen, OH, or N(C 1 -C 6 alkyl) 2 .
  • R f is selected from N(CH 3 ) 2 .
  • Re is selected from N(CH 3 ) 2 , CH 2 N(CH 3 ) 2 or
  • X is selected from
  • X is selected from the following groups:
  • X is selected from the following groups:
  • X is selected from the following groups:
  • X is selected from the following groups:
  • X is selected from
  • X is selected from
  • X is selected from
  • R 1 and R 2 together form a C 3 -C 8 cycloalkyl group or a 4-10 membered heterocyclic group, and the C 3 -C 8 cycloalkyl group or a 4-10 membered heterocyclic group Cyclic is optionally substituted by R 1b .
  • R 1 and R 2 together form a C 3 -C 6 cycloalkyl or a 4-7 membered heterocyclyl, and the C 3 -C 6 cycloalkyl or 4-7 membered heterocyclyl Cyclic is optionally substituted by R 1b .
  • R 1 , R 2 and the atoms to which they are attached together form a group optionally substituted by R 1b : cyclobutyl, spiro[2,3]hexyl, or oxetanyl.
  • R 1b is selected from halogen, CN, C 1 -C 3 alkyl or C 1 -C 3 alkoxy, and the C 1 -C 3 alkyl or C 1 -C 3 alkoxy is any is replaced by R 1c .
  • R 1c is selected from halogen, OH or CN.
  • R 1c is selected from CN.
  • R 1b is selected from F, CN, methyl, methoxy, or CH 2 CN.
  • R 1 and R 2 are independently selected from H, halogen, CN, NH 2 , NH(C 1 -C 6 alkyl), N(C 1 -C 6 alkyl) 2 , C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 8 cycloalkyl or 4-7 membered heterocyclic group, the C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 8 cycloalkyl or 4-7 membered heterocyclic group is optionally substituted by R 1a , and the C 3 -C 8 cycloalkyl or 4-7 membered heterocyclic group may be a spiro ring, bridged ring or in ring form.
  • R and R are independently selected from H, halogen, CN, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, or C 3 -C 6 cycloalkyl, the C 1 -C 3 alkyl, C 1 -C 3 alkoxy or C 3 -C 6 cycloalkyl is optionally substituted by R 1a .
  • R 1 and R 2 are independently selected from H, C 1 -C 3 alkyl or C 3 -C 6 cycloalkyl, said C 1 -C 3 alkyl or C 3 -C 6 cycloalkane The group is optionally substituted by R 1a .
  • R 1 and R 2 are independently selected from H or C 1 -C 3 alkyl optionally substituted with R 1a .
  • R 1 and R 2 are independently selected from H, F, methyl, CF 3 , cyclopropyl, cyclobutyl, or Or R 1 , R 2 and the atoms connected to them jointly form the following group:
  • R 1 and R 2 are independently selected from H, methyl or cyclobutyl, or R 1 , R 2 and the atoms to which they are attached together form the following group:
  • R 1 and R 2 are independently selected from H or methyl, or R 1 , R 2 and the atoms they are connected to together form the following group:
  • R and R are independently selected from H, F, cyclopropyl, methyl, or Or R 1 , R 2 and the atoms connected to them jointly form the following group:
  • R 1 , R 2 and the atoms to which they are attached together form the following group:
  • R 1 , R 2 and the atoms to which they are attached together form the following group:
  • R and R are independently selected from H, methyl, or cyclobutyl.
  • R and R are independently selected from H or methyl.
  • W is selected from -(CR 13 R 14 )W 1 .
  • R 13 , R 14 are independently selected from H, halogen, OH or methyl.
  • both R 13 and R 14 are H.
  • R 1 and R 13 together with the atoms and bonds to which they are attached form a C 3 -C 6 cycloalkyl optionally substituted by R 13a .
  • R 13a is selected from C 1 -C 3 alkyl.
  • R 13a is selected from methyl.
  • R and R together with the atoms and bonds to which they are attached form
  • W is selected from W 1 .
  • W is selected from 5-10 membered heteroaryl or 6-10 membered heterocyclyl optionally substituted by R.
  • W is selected from 5-10 membered heteroaryl optionally substituted by R.
  • W is selected from 5-membered heteroaryl or 8-membered heterocyclyl optionally substituted by R.
  • W is selected from 5-membered heteroaryl optionally substituted with R.
  • W is selected from the following groups optionally substituted by R : pyrrolyl, thienyl, furyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, thiadiazolyl, tri Azazolyl, oxazolyl, isoxazolyl, oxadiazolyl or 6,7-dihydro-5H-pyrrolo[2,1-c][1,2,4]triazolyl.
  • W is selected from the following groups optionally substituted by R : pyrrolyl, thienyl, furyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, thiadiazolyl, tri Azazolyl, oxazolyl, isoxazolyl or oxadiazolyl.
  • W is selected from the group optionally substituted by R from the group consisting of triazolyl , oxazolyl, isoxazolyl, oxadiazolyl, or 6,7-dihydro-5H-pyrrole And[2,1-c][1,2,4]triazolyl.
  • W 1 is selected from triazolyl, oxazolyl, isoxazolyl, or oxadiazolyl, optionally substituted by R 15 .
  • R 15 is selected from halogen, OH, NH 2 , C 1 -C 3 alkyl or C 3 -C 6 cycloalkyl, said C 1 -C 3 alkyl or C 3 -C 6 ring Alkyl is optionally substituted by R 15a .
  • R 15 is selected from OH, C 1 -C 3 alkyl or C 3 -C 6 cycloalkyl, the C 1 -C 3 alkyl or C 3 -C 6 cycloalkyl optionally replaced by R 15a is substituted.
  • R 15 is selected from methyl or cyclopropyl optionally substituted with R 15a .
  • R 15a is selected from halogen, OH or CN.
  • R 15a is selected from F.
  • R 15 is selected from methyl, CHF 2 or cyclopropyl.
  • W is selected from the following groups:
  • W is selected from the following groups:
  • W is selected from
  • the compound of formula (I) or its stereoisomer or pharmaceutically acceptable salt thereof of the present application is selected from the compound of formula (II) or its stereoisomer or its pharmaceutically acceptable salt:
  • Y 1 , Y 2 , Y 3 , Y 4 , X, Q, W, R 1 and R 2 are as defined above.
  • the compound of formula (I) or its stereoisomer or pharmaceutically acceptable salt thereof of the present application is selected from the compound of formula (III) or its stereoisomer or its pharmaceutically acceptable salt:
  • Z 1 , Z 2 , Z 3 are independently selected from CH, CR 10 or N; R 10 , Y 1 , Y 2 , Y 3 , Y 4 , X, W, R 1 and R 2 are as defined above.
  • the compound of formula (I) or its stereoisomer or pharmaceutically acceptable salt thereof of the present application is selected from the compound of formula (IV) or its stereoisomer or its pharmaceutically acceptable salt:
  • R 10 , Y 1 , Y 2 , Y 3 , Y 4 , X, W, R 1 and R 2 are as defined above.
  • the compound of formula (I) or its stereoisomer or pharmaceutically acceptable salt thereof of the present application is selected from the compound of formula (V) or its stereoisomer or its pharmaceutically acceptable salt:
  • Z 1 , Z 2 , Z 3 are independently selected from CH, CR 10 or N;
  • a 3 is selected from CR 11a R 11b , NR 12 , O or S;
  • R 10 , R 11a , R 11b , R 12 , Y 1 , Y 2 , Y 3 , Y 4 , X, W, R 1 and R 2 are as defined above.
  • the compound of formula (I) or its stereoisomer or pharmaceutically acceptable salt thereof of the present application is selected from the compound of formula (VI) or its stereoisomer or its pharmaceutically acceptable salt:
  • Z 1 is selected from CH, CR 10 or N
  • Z 2 is selected from CH 2 , CHR 10 , NH or NR 10 , O or S
  • R 10 , Y 1 , Y 2 , Y 3 , Y 4 , X, W , R 1 and R 2 are as defined above.
  • the compound of formula (I) of the present application or its stereoisomer or its pharmaceutically acceptable salt is selected from the following compounds or its pharmaceutically acceptable salt:
  • the application provides a pharmaceutical composition, which comprises the compound of formula (I), compound of formula (II), compound of formula (III), compound of formula (IV), compound of formula (V), compound of formula (VI) A compound or its stereoisomer or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable auxiliary material.
  • the present application provides a method for treating a disease or condition mediated by Cbl-b in a mammal, comprising administering a therapeutically effective amount of a compound of formula (I), formula (II) to a mammal in need of the treatment, preferably a human being ), a compound of formula (III), a compound of formula (IV), a compound of formula (V), a compound of formula (VI) or a stereoisomer or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
  • the present application provides a method for treating or preventing tumors or autoimmune diseases in mammals, comprising administering a therapeutically effective amount of a compound of formula (I), formula (II) to a mammal in need of the treatment or prevention, preferably a human ), a compound of formula (III), a compound of formula (IV), a compound of formula (V), a compound of formula (VI) or a stereoisomer or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
  • the application provides a compound of formula (I), a compound of formula (II), a compound of formula (III), a compound of formula (IV), a compound of formula (V), a compound of formula (VI) or its stereoisomer or its Use of a pharmaceutically acceptable salt, or a pharmaceutical composition thereof, in the preparation of a medicament for preventing or treating a disease or disease mediated by Cbl-b.
  • the application provides a compound of formula (I), a compound of formula (II), a compound of formula (III), a compound of formula (IV), a compound of formula (V), a compound of formula (VI) or its stereoisomer or its Use of pharmaceutically acceptable salts or pharmaceutical compositions thereof in the preparation of drugs for preventing or treating tumors or autoimmune diseases.
  • the application provides a compound of formula (I), a compound of formula (II), a compound of formula (III), a compound of formula (IV), a compound of formula (V), a compound of formula (VI) or its stereoisomer or its Use of a pharmaceutically acceptable salt, or a pharmaceutical composition thereof, in preventing or treating diseases or conditions mediated by Cbl-b.
  • the application provides a compound of formula (I), a compound of formula (II), a compound of formula (III), a compound of formula (IV), a compound of formula (V), a compound of formula (VI) or its stereoisomer or its Use of pharmaceutically acceptable salts or pharmaceutical compositions thereof in preventing or treating tumors or autoimmune diseases.
  • the application provides compounds of formula (I), compounds of formula (II), compounds of formula (III) and compounds of formula (IV) for preventing or treating mammals, preferably human diseases or diseases mediated by Cbl-b ) compound, compound of formula (V), compound of formula (VI) or its stereoisomer or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
  • the present application provides compounds of formula (I), compounds of formula (II), compounds of formula (III), compounds of formula (IV), compounds of formula (V) compound, compound of formula (VI) or its stereoisomer or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
  • the disease or condition mediated by Cbl-b is selected from tumors or autoimmune diseases.
  • tautomer refers to isomers of functional groups resulting from the rapid movement of an atom in a molecule between two positions. Compounds of the present application may exhibit tautomerism. Tautomeric compounds can exist in two or more interconvertible species. Tautomers generally exist in equilibrium and attempts to isolate a single tautomer usually result in a mixture whose physicochemical properties are consistent with the mixture of compounds. The position of equilibrium depends on the chemical properties within the molecule. For example, in many aliphatic aldehydes and ketones such as acetaldehyde, the keto form predominates; in phenols, the enol form predominates. This application encompasses all tautomeric forms of the compounds.
  • stereoisomer refers to isomers resulting from differences in the arrangement of atoms in a molecule in space, including cis-trans isomers, enantiomers and diastereomers.
  • the compounds of the present application may have asymmetric atoms such as carbon atoms, sulfur atoms, nitrogen atoms, phosphorus atoms or asymmetric double bonds, so the compounds of the present application may exist in specific geometric or stereoisomer forms.
  • Specific geometric or stereoisomeric forms may be cis and trans isomers, E and Z geometric isomers, (-)- and (+)-enantiomers, (R)- and (S )-enantiomers, diastereomers, (D)-isomers, (L)-isomers, and racemic or other mixtures thereof, such as enantiomers or diastereomers Enriched mixtures, all of the above isomers and mixtures thereof fall within the definition of the compounds of the present application.
  • asymmetric carbon atoms there may be additional asymmetric carbon atoms, asymmetric sulfur atoms, asymmetric nitrogen atoms or asymmetric phosphorus atoms in substituents such as alkyl groups, and these isomers and their mixtures involved in all substituents are also included in Within the definition of the compounds of the present application.
  • the compounds containing asymmetric atoms of the present application can be isolated in optically pure form or racemic form, optically active form can be resolved from racemic mixture, or synthesized by using chiral raw materials or chiral reagents .
  • substituted means that any one or more hydrogen atoms on the specified atom are replaced by a substituent, as long as the valence of the specified atom is normal and the substituted compound is stable.
  • ethyl is “optionally” substituted with halogen , meaning that the ethyl group can be unsubstituted ( CH2CH3 ), monosubstituted ( CH2CH2F , CH2CH2Cl , etc.), polysubstituted ( CHFCH2F , CH2CHF2 , CHFCH2Cl , CH2CHCl2 , etc. ) or fully substituted ( CF2CF3 , CF2CCl3 , CCl2CCl3 , etc.) . It will be appreciated by those skilled in the art that for any group containing one or more substituents, no sterically impossible and/or synthetically impossible substitution or substitution pattern is introduced.
  • any variable eg R 1a , R 2a
  • its definition is independent at each occurrence. For example, if a group is substituted by 2 R 1a , each R 1a has independent options.
  • linking group When the number of a linking group is 0, such as -(CR 13 R 14 ) 0 -, it means that the linking group is a bond.
  • linking group mentioned herein does not indicate its linking direction
  • its linking direction is arbitrary.
  • L in is selected from “-NR 7 CH 2 -”
  • L can be connected to ring D from left to right to form “ring D-NR 7 CH 2 -”
  • the direction of linking ring D constitutes "ring D-CH 2 NR 7 -”.
  • Cm - Cn herein refers to having an integer number of carbon atoms in the range of mn.
  • C 1 -C 6 means that the group can have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms.
  • alkyl refers to a hydrocarbon group having the general formula C n H 2n+1 , and the alkyl group may be straight or branched.
  • C 1 -C 6 alkyl is understood to mean a straight-chain or branched saturated monovalent hydrocarbon radical having 1, 2, 3, 4, 5 or 6 carbon atoms.
  • alkyl group examples include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, 2- Methylbutyl, 1-methylbutyl, 1-ethylpropyl, 1,2-dimethylpropyl, neopentyl, 1,1-dimethylpropyl, 4-methylpentyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 2-ethylbutyl, 1-ethylbutyl, 3,3-dimethylbutyl, 2,2-di Methylbutyl, 1,1-dimethylbutyl, 2,3-dimethylbutyl, 1,3-dimethylbutyl or 1,2-dimethylbutyl, etc.
  • C 1 -C 3 alkyl is understood to mean a linear or branched saturated monovalent hydrocarbon radical having 1, 2 or 3 carbon atoms.
  • the "C 1 -C 6 alkyl group” may include “C 1 -C 3 alkyl group”.
  • alkoxy refers to a monovalent group produced by the loss of a hydrogen atom on a hydroxyl group of a straight-chain or branched alcohol, which can be understood as “alkyloxy” or “alkyl-O-”.
  • C 1 -C 6 alkoxy can be understood as “C 1 -C 6 alkyloxy” or “C 1 -C 6 alkyl-O-”; the term “C 1 -C 3 alkoxy” Can be understood as “C 1 -C 3 alkyloxy” or "C 1 -C 3 alkyl-O-”.
  • the "C 1 -C 6 alkoxy” may further include "C 1 -C 3 alkoxy”.
  • alkenyl refers to a linear or branched monovalent unsaturated aliphatic hydrocarbon group consisting of carbon atoms and hydrogen atoms and having at least one double bond.
  • C 2 -C 4 alkenyl is understood to mean a linear or branched unsaturated monovalent hydrocarbon group containing one or more double bonds and having 2, 3 or 4 carbon atoms, "C 2 -C 4 alkenyl” is preferably C 2 or C 3 alkenyl. It is understood that where the alkenyl group contains more than one double bond, the double bonds may be separated from each other or conjugated.
  • alkenyl group examples include, but are not limited to, vinyl, allyl, (E)-2-methylvinyl, (Z)-2-methylvinyl, (E)-but-2-enyl , (Z)-but-2-enyl, (E)-but-1-enyl, (Z)-but-1-enyl, isopropenyl, 2-methylprop-2-enyl, 1 -Methylprop-2-enyl, 2-methylprop-1-enyl, (E)-1-methylprop-1-enyl or (Z)-1-methylprop-1-enyl wait.
  • alkynyl refers to a linear or branched monovalent unsaturated aliphatic hydrocarbon group consisting of carbon atoms and hydrogen atoms and having at least one triple bond.
  • C 2 -C 4 alkynyl is understood to mean a straight-chain or branched unsaturated monovalent hydrocarbon radical comprising one or more triple bonds and having 2, 3 or 4 carbon atoms.
  • Examples of “C 2 -C 4 alkynyl” include, but are not limited to, ethynyl (-C ⁇ CH), propynyl (-C ⁇ CCH 3 , -CH 2 C ⁇ CH), but-1-ynyl, but -2-ynyl or but-3-ynyl.
  • C 2 -C 4 alkynyl may include “C 2 -C 3 alkynyl", examples of “C 2 -C 3 alkynyl” include ethynyl (-C ⁇ CH), prop-1-ynyl (-C ⁇ CCH 3 ), prop-2-ynyl (propargyl).
  • cycloalkyl refers to a fully saturated carbocyclic ring in the form of a monocyclic ring, a double ring, a bridged ring, or a spiro ring. Unless otherwise indicated, the carbocycle is typically a 3 to 10 membered ring.
  • C 3 -C 10 cycloalkyl is understood to mean a saturated monovalent monocyclic, fused, spiro or bridged ring having 3, 4, 5, 6, 7, 8, 9 or 10 carbons atom.
  • cycloalkyl examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, norbornyl (bicyclo[2.2 .1] heptyl), bicyclo [2.2.2] octyl, adamantyl, spiro [4.5] decanyl, etc.
  • C 3 -C 10 cycloalkyl may include “C 3 -C 8 cycloalkyl", “C 3 -C 8 cycloalkyl” may include “C 3 -C 6 cycloalkyl”, “C 3 -C 6 cycloalkyl” may include “C 3 -C 4 cycloalkyl”.
  • C 3 -C 6 cycloalkyl can be understood as meaning a saturated monovalent monocyclic or bicyclic hydrocarbon ring having 3, 4, 5 or 6 carbon atoms, specific examples include but are not limited to cyclopropyl, cyclo Butyl, cyclopentyl or cyclohexyl, etc.
  • cycloalkyloxy can be understood as “cycloalkyl-O-”.
  • cycloalkenyl refers to a non-aromatic monocyclic or polycyclic hydrocarbon group containing at least one carbon-carbon double bond.
  • C 3 -C 6 cycloalkenyl refers to a non-aromatic cyclic hydrocarbon group having 3, 4, 5 or 6 carbon atoms as ring atoms and containing at least one carbon-carbon double bond.
  • Specific examples of C 3 -C 6 cycloalkenyl include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, and the like.
  • 4-10 membered heterocyclic group refers to a heterocyclic group with 4, 5, 6, 7, 8, 9 or 10 ring atoms, and its ring atoms contain 1, 2, 3, 4 or 5 independent selected from the heteroatoms or heteroatom groups described above.
  • the term "4-10 membered nitrogen-containing heterocyclic group” refers to a 4, 5, 6, 7, 8, 9 or 10-membered heterocyclic group containing at least one N atom in its ring atom, "4-10 membered nitrogen-containing "Heterocyclic group” includes "6-10 membered nitrogen-containing heterocyclic group”.
  • 4-7 membered monocyclic nitrogen-containing heterocyclic group refers to a 4, 5, 6 or 7-membered heterocyclic group in the form of a monocyclic ring containing at least one N atom.
  • “4-10 membered heterocyclic group” includes “6-10 membered heterocyclic group”, “6-10 membered heterocyclic group” further includes “6-7 membered heterocyclic group”, wherein, the specific Examples include, but are not limited to, azetidinyl, thietanyl, or oxetanyl; specific examples of 5-membered heterocyclic groups include, but are not limited to, tetrahydrofuranyl, dioxolyl, pyrrole Alkyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl, 4,5-dihydrooxazolyl or 2,5-dihydro-1H-pyrrolyl; specific examples of 6-membered heterocyclic groups include, but are not limited to
  • the heterocyclic group can also be a bicyclic group, wherein, specific examples of the 5,5-membered bicyclic group include, but are not limited to, hexahydrocyclopenta[c]pyrrol-2(1H)-yl; 5,6-membered bicyclic group Specific examples include, but are not limited to, hexahydropyrrolo[1,2-a]pyrazin-2(1H)-yl, 5,6,7,8-tetrahydro-[1,2,4]triazolo[4 ,3-a]pyrazinyl or 5,6,7,8-tetrahydroimidazo[1,5-a]pyrazinyl.
  • the heterocyclic group may be a benzo-fused ring group of the above-mentioned 4-7 membered heterocyclic group, specific examples include but not limited to dihydroisoquinolyl and the like.
  • “4-10 membered heterocyclic group” may include “5-10 membered heterocyclic group”, “4-7 membered heterocyclic group”, “5-7 membered heterocyclic group”, “5-6 membered heterocyclic group” , “6-8 membered heterocyclyl", “4-10 membered heterocycloalkyl”, “5-10 membered heterocycloalkyl", “4-7 membered heterocycloalkyl”, “5-6 membered heterocycloalkyl”Cycloalkyl”,”6-8 membered heterocyclyl", etc.
  • "4-7 membered heterocyclyl” may further include "4-6 membered heterocyclyl", "5-7 membered heterocyclyl", “5-6 membered heterocyclyl”, “4-7 membered heterocycloal
  • heterocyclyloxy can be understood as “heterocyclyl-O-”.
  • 4-10 membered heterocycloalkyl refers to a heterocycloalkyl group with 4, 5, 6, 7, 8, 9 or 10 ring atoms, and its ring atoms contain 1, 2, 3, 4 or 5 independently selected from the heteroatoms or heteroatom groups described above.
  • 4-membered heterocycloalkyl includes “4-7 membered heterocycloalkyl", wherein specific examples of 4-membered heterocycloalkyl include, but are not limited to, azetidinyl, oxetanyl or thibutanyl; Specific examples of 5-membered heterocycloalkyl groups include, but are not limited to, tetrahydrofuranyl, tetrahydrothiophenyl, pyrrolidinyl, isoxazolidinyl, oxazolidinyl, isothiazolidinyl, thiazolidinyl, imidazolidinyl, or tetrahydrofuranyl.
  • 6-membered heterocycloalkyl include, but are not limited to, piperidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, morpholinyl, piperazinyl, 1,4-thiaxanyl , 1,4-dioxanyl, thiomorpholinyl, 1,3-dithianyl, or 1,4-dithianyl; specific examples of 7-membered heterocycloalkyl include, but are not limited to, aza Cycloheptyl, oxepanyl or thiepanyl.
  • aryl refers to an all-carbon monocyclic or fused polycyclic aromatic ring group having a conjugated ⁇ -electron system.
  • Aryl groups can have 6-14 carbon atoms or 6-10 carbon atoms.
  • C 6 -C 10 aryl is understood as a monovalent aromatic monocyclic or bicyclic group having 6 to 10 carbon atoms.
  • rings having 6 carbon atoms such as phenyl; or rings having 10 carbon atoms (“C 10 aryl”), such as naphthyl.
  • aryloxy is understood to mean “aryl-O-”.
  • heteroaryl refers to an aromatic monocyclic or fused polycyclic ring system, which contains at least one ring atom selected from N, O, and S, and the remaining ring atoms are C aromatic ring groups.
  • heteroaryl refers to include monovalent monocyclic or bicyclic aromatic ring systems having 5, 6, 7, 8, 9 or 10 ring atoms, in particular 5 or 6 or 9 or 10 ring atoms, and it contains 1, 2, 3, 4 or 5, preferably 1, 2 or 3 heteroatoms independently selected from N, O and S.
  • heteroaryl is selected from thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl or thiazolyl Diazolyl, etc.
  • benzo derivatives such as benzofuryl, benzothienyl, benzothiazolyl, benzoxazolyl, benzisoxazolyl, benzimidazolyl, benzotriazole base, indazolyl, indolyl or isoindolyl, etc.; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, or triazinyl, etc., and their benzo derivatives, such as quinolinyl, quinazole Linyl or isoquinolinyl, etc.; or azocinyl, indolizinyl, purinyl, etc.
  • 5-6 membered heteroaryl refers to an aromatic ring system having 5 or 6 ring atoms, and which contains 1, 2 or 3, preferably 1 or 2, heteroatoms independently selected from N, O and S .
  • 6-membered heteroaryl refers to an aromatic ring system having 6 ring atoms and which contains 1, 2 or 3, preferably 1 or 2 N as heteroatoms.
  • 5-10 membered heteroaryl includes “5-6 membered heteroaryl”.
  • heteroaryloxy is understood to mean “heteroaryl-O-”.
  • halo or halogen refers to fluorine (F), chlorine (Cl), bromine (Br) or iodine (I).
  • hydroxyl refers to a -OH group.
  • cyano refers to a -CN group.
  • mercapto refers to a -SH group.
  • amino refers to a -NH2 group.
  • nitro refers to a -NO2 group.
  • terapéuticaally effective amount means (i) treating or preventing a particular disease, condition or disorder, (ii) alleviating, ameliorating or eliminating one or more symptoms of a particular disease, condition or disorder, or (iii) preventing or delaying The amount of a compound of the application for the onset of one or more symptoms of a particular disease, condition or disorder described herein.
  • the amount of a compound of the present application that constitutes a “therapeutically effective amount” will vary depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but can be routinely determined by a person skilled in the art according to its own knowledge and this disclosure.
  • pharmaceutically acceptable refers to those compounds, materials, compositions and/or dosage forms which, within the scope of sound medical judgment, are suitable for use in contact with human and animal tissues without excessive Toxicity, irritation, allergic reaction, or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt refers to salts of pharmaceutically acceptable acids or bases, including salts formed between compounds and inorganic or organic acids, and salts formed between compounds and inorganic or organic bases.
  • pharmaceutical composition refers to a mixture of one or more compounds of the present application or their stereoisomers or salts thereof and pharmaceutically acceptable auxiliary materials.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound of the present application to an organism.
  • pharmaceutically acceptable excipients refers to those excipients that have no obvious stimulating effect on the organism and will not impair the biological activity and performance of the active compound. Suitable excipients are well known to those skilled in the art, such as carbohydrates, waxes, water-soluble and/or water-swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like.
  • the present application also includes isotopically labeled compounds of the present application that are identical to those described herein, but wherein one or more atoms are replaced by an atom having an atomic mass or mass number different from that normally found in nature.
  • isotopes that may be incorporated into the compounds of the present application include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 123 I, 125 I and 36 Cl, etc.
  • Certain isotopically labeled compounds of the present application are useful in compound and/or substrate tissue distribution assays.
  • Tritiated (ie3H ) and carbon-14 (ie14C ) isotopes are especially preferred for their ease of preparation and detectability.
  • Positron-emitting isotopes such as 15 O, 13 N, 11 C, and 18 F, can be used in positron emission tomography (PET) studies to determine substrate occupancy.
  • Isotopically labeled compounds of the present application can generally be prepared by following procedures similar to those disclosed in the Schemes and/or Examples below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • the pharmaceutical composition of the present application can be prepared by combining the compound of the present application with suitable pharmaceutically acceptable auxiliary materials, for example, it can be formulated into solid, semi-solid, liquid or gaseous preparations, such as tablets, pills, capsules, powders , granules, ointments, emulsions, suspensions, suppositories, injections, inhalants, gels, microspheres and aerosols, etc.
  • Typical routes for administering a compound of the present application or a stereoisomer thereof or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof include, but are not limited to, oral, rectal, topical, inhalation, parenteral, sublingual, intravaginal, intranasal, Intraocular, intraperitoneal, intramuscular, subcutaneous, intravenous administration.
  • the pharmaceutical composition of the present application can be produced by methods well known in the art, such as conventional mixing methods, dissolving methods, granulating methods, emulsifying methods, freeze-drying methods and the like.
  • the pharmaceutical composition is in oral form.
  • the pharmaceutical compositions can be formulated by mixing the active compounds with pharmaceutically acceptable excipients well known in the art. These excipients enable the compounds of the present application to be formulated into tablets, pills, lozenges, dragees, capsules, liquids, gels, slurries, suspensions, etc., for oral administration to patients.
  • Solid oral compositions can be prepared by conventional methods of mixing, filling or tabletting. It can be obtained, for example, by mixing the active compound with solid excipients, optionally milling the resulting mixture, adding other suitable excipients if desired, and processing the mixture into granules to obtain tablets Or the core of the sugar coating.
  • Suitable auxiliary materials include but are not limited to: binders, diluents, disintegrants, lubricants, glidants or flavoring agents, etc.
  • the pharmaceutical composition may also be adapted for parenteral administration as a suitable unit dosage form of sterile solutions, suspensions or lyophilized products.
  • the daily dosage is 0.01 mg/kg to 200 mg/kg body weight, preferably 0.05 mg/kg to 50 mg/kg body weight, more preferably 0.1 mg/kg to 30 mg /kg body weight, in single or divided doses.
  • the compounds of the present application can be prepared by various synthetic methods well known to those skilled in the art, including the specific examples listed below, the embodiments formed by combining them with other chemical synthesis methods, and the methods well known to those skilled in the art In an equivalent alternative, preferred embodiments include but are not limited to the examples of the present application.
  • some compounds of the general formula (III) of the present application can be prepared by those skilled in the field of organic synthesis through the following synthetic route 1:
  • X' is halogen or OTf
  • X 2 is halogen
  • CG 1 and CG 2 are independently selected from B(OR) 2 , BF 3 K, SnR' 4 , ZnX 2
  • the R is selected from H or C 1 -C 6 alkyl
  • R' is selected from C 1 -C 6 alkyl
  • X 2 is halogen
  • Z 1 , Z 2 , Z 3 , Y 1 , Y 2 , Y 3 , Y 4 , W, R 1 , R 2 , R 4 and ring B are as defined in the general formula (III).
  • some compounds of the general formula (III) of the present application can be prepared by those skilled in the field of organic synthesis through the following synthetic route 2:
  • X' is halogen or OTf;
  • X" is halogen;
  • CG 1 is independently selected from B(OR) 2 , BF 3 K, SnR' 4 , ZnX",
  • the R is selected from H or C 1 -C 6 alkyl, R' is selected from C 1 -C 6 alkyl,
  • X" is halogen;
  • Z 1 , Z 2 , Z 3 , Y 1 , Y 2 , Y 3 , Y 4 , W, R 1 , R 2 , R 4 and ring B are as defined in the general formula (III).
  • some compounds of the general formula (VI) of the present application can be prepared by those skilled in the field of organic synthesis through the following synthetic route 3:
  • X' is halogen or OTf
  • Z 1 , Z 2 , Y 1 , Y 2 , Y 3 , Y 4 , W, R 1 , R 2 , R 4 and ring B are as defined in general formula (VI).
  • some compounds of the general formula (V) of the present application can be prepared by those skilled in the field of organic synthesis through the following synthetic route 4:
  • X' is halogen or OTf
  • X" is halogen
  • CG 1 is independently selected from B(OR) 2 , BF 3 K, SnR' 4 , ZnX"
  • the R is selected from H or C 1 -C 6 alkyl
  • R' is selected from C 1 -C 6 alkyl
  • Z 1 , Z 2 , Z 3 , X, Y 1 , Y 2 , Y 3 , Y 4 , W, R 1 , R 2 , R 4 and ring B are as defined in the general formula (V).
  • ratios indicated for mixed solvents are volume mixing ratios. Unless otherwise stated, % means wt%.
  • NMR nuclear magnetic resonance
  • MS mass spectroscopy
  • the following eluents can be mixed eluents formed by two or more solvents, and the ratio is the volume ratio of each solvent.
  • “0-10% methanol/dichloromethane” means that in the gradient elution process, mixed eluents
  • the volume ratio of methanol and dichloromethane in the mixture is 0:100 ⁇ 10:90.
  • B 2 Pin 2 pinacol diboronate; Pd(dppf)Cl 2 : [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride; NIS: N-iodosuccinyl imine; DCM: dichloromethane; Pd(PPh 3 ) 4 : tetrakistriphenylphosphine palladium; THF: tetrahydrofuran; XphosPdG 2 : chloro(2-dicyclohexylphosphino-2',4',6'-tri Isopropyl-1,1'-biphenyl)[2-(2'-amino-1,1'-biphenyl)]palladium(II); Xphos: 2-dicyclohexylphosphine-2',4',6'-triisopropylbiphenyl; SFC: supercritical fluid chromatography; (Boc) 2
  • Step 1 4-Methyl-3-(3-Methyl-1-(3-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl) Preparation of phenyl)cyclobutyl)-4H-1,2,4-triazole (1B)
  • Step 5 3-[3-[3-Methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl]phenyl]-7-[[ Preparation of (3S)-3-methyl-1-piperidinyl]methyl]pyrido[1,2-a]pyrimidin-4-one (Compound 1)
  • Example 2 3-[3-[3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl]phenyl]-7-[ [(3S)-3-methyl-1-piperidinyl]methyl]-9-(trifluoromethyl)pyrido[1,2-a]pyrimidin-4-one (compound 2), 3-( 3-((1S,3R)-3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)phenyl)-7-(( (S)-3-methylpiperidin-1-yl)methyl)-9-(trifluoromethyl)-4H-pyrido[1,2-a]pyrimidin-4-one and 3-(3- ((1R,3S)-3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)phenyl)-7-(((S Preparation of )-3-methylpiperidin-1-yl)methyl
  • Step 3 7-[[(3S)-3-Methyl-1-piperidinyl]methyl]-9-(trifluoromethyl)pyrido[1,2-a]pyrimidin-4-one (2E ) preparation
  • Step 5 3-[3-[3-Methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl]phenyl]-7-[[ (3S)-3-methyl-1-piperidinyl]methyl]-9-(trifluoromethyl)pyrido[1,2-a]pyrimidin-4-one (compound 2), 3-(3 -((1S,3R)-3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)phenyl)-7-((( S)-3-methylpiperidin-1-yl)methyl)-9-(trifluoromethyl)-4H-pyrido[1,2-a]pyrimidin-4-one and 3-(3-( (1R,3S)-3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)phenyl)-7-(((S) Preparation of -3-methylpiperidin-1-y
  • Step 2 Preparation of 1-(5-bromo-2-hydroxy-3-methylphenyl)-3-(dimethylamino)prop-2-en-1-one (58B)
  • N,N-Dimethylformamide dimethyl acetal (4.16 g, 34.92 mmol) was added to a sealed tube containing a solution of 58A (3 g, 13.3 mmol) in anhydrous toluene (60 mL) at room temperature. The reaction solution was then warmed to 115°C and stirred for 16 hours. After the reaction, the reaction liquid was cooled to room temperature, water (60 mL) was added, and ethyl acetate extracted (50 mL ⁇ 3).
  • Step 4 6-Bromo-8-methyl-3-(3-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl) Preparation of phenyl)-4H-chromen-4-one (58D)
  • Step 5 (S)-8-Methyl-3-(3-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl) Preparation of phenyl)-6-((3-methylpiperidin-1-yl)methyl)-4H-chromen-4-one (compound 58)
  • the organic phase is dried with anhydrous sodium sulfate, filtered, and distilled under reduced pressure to obtain the crude product, and the crude product is used for high performance liquid chromatography (chromatographic column: Welch Xtimate C18; column length 150mm, inner diameter 30mm, particle diameter 5 ⁇ m; mobile phase A: water (containing 0.225% formic acid); mobile phase B: acetonitrile; gradient: mobile phase B from 24% to 54% in 7 minutes, then eluted with 100% mobile phase B for 3 minutes; flow rate: 25 mL/min) for purification.
  • the title compound 58 was obtained (43.25 mg, 34%).
  • Step 1 Preparation of tert-butyl (4-formylthiazol-2-yl)carbamate (59A)
  • Step 4 Ethyl 2-(3-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)phenyl)acetate (59D) preparation of
  • Step 5 3-(Dimethylamino)-2-(3-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)benzene base) preparation of ethyl acrylate (59E)
  • Step 6 (S)-6-(3-(3-Methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)phenyl)-2 Preparation of -((3-methylpiperidin-1-yl)methyl)-5H-thiazolo[3,2-a]pyrimidin-5-one (compound 59)
  • Step 1 5-(9-methyl-3-(3-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl) Phenyl)-4-oxo-4H-pyrido[1,2-a]pyrimidin-7-yl)-3,6-1,2,3,6-tetrahydropyridine-1-carboxylic acid tert-butyl ester
  • (60A) 5-(9-methyl-3-(3-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl) Phenyl)-4-oxo-4H-pyrido[1,2-a]pyrimidin-7-yl)-3,6-1,2,3,6-tetrahydropyridine-1-carboxylic acid tert-butyl ester
  • Step 2 3-(9-methyl-3-(3-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl) Preparation of tert-butyl phenyl)-4-oxo-4H-pyrido[1,2-a]pyrimidin-7-yl]piperidine-1-carboxylate (60B)
  • Step 3 9-methyl-3-(3-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)phenyl) Preparation of -7-(piperidin-3-yl)-4H-pyrido[1,2-a]pyrimidin-4-one (compound 60)
  • Step 4 Preparation of 2-(2-(3-bromophenyl)-2-cyclobutylpropionyl)-N-methylthiosemicarbazide (61D)
  • Step 5 5-(1-(3-Bromophenyl)-1-cyclobutylethyl)-4-methyl-4H-1,2,4-triazole-3-thiol (61E) preparation
  • 61D (1.5g, 4.05mmol) was dissolved in aqueous sodium hydroxide solution (1.5M, 30mL), heated to 50°C, and stirred for 12 hours. After the reaction was complete, the reaction solution was cooled to room temperature, adjusted to pH 3 with 1M aqueous hydrochloric acid, extracted with ethyl acetate, dried with anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain the crude product of the title compound 61E (1.3 g). used directly in the next reaction.
  • aqueous sodium hydroxide solution 1.5M, 30mL
  • Step 6 Preparation of 3-(1-(3-bromophenyl)-1-cyclobutylethyl)-4-methyl-4H-1,2,4-triazole (61F)
  • Step 7 3-(1-Cyclobutyl-1-(3-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl ) ethyl)-4-methyl-4H-1,2,4-triazole (61G) preparation
  • Step 8 3-(3-(1-Cyclobutyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)ethyl)phenyl)-7-(( Preparation of (S)-3-methylpiperidin-1-yl)methyl)-9-(trifluoromethyl)-4H-pyrido[1,2-a]pyrimidin-4-one (compound 61)
  • 63B (480 mg, 1.78 mmol) was added to a mixture of water (5 mL) and tetrahydrofuran (5 mL), and lithium hydroxide (299.34 mg, 7.13 mmol) was added, and stirred at room temperature for 3 hours. After the reaction, tetrahydrofuran was removed by rotary evaporation, extracted three times with ethyl acetate, the aqueous phase was adjusted to pH 3-4 with 1M hydrochloric acid, extracted with ethyl acetate, and the organic phase was concentrated to dryness under reduced pressure to obtain the solid title compound 63C (215 mg).
  • 63D (110 mg, 321.40 ⁇ mol) was added to aqueous sodium hydroxide (1M, 3 mL). The reaction was stirred at room temperature for 12 hours. After the reaction, adjust the pH to 3-4 with 1M hydrochloric acid. It was extracted with ethyl acetate, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to dryness under reduced pressure to obtain the title product 63E (77 mg) as a solid.
  • Step 7 3-[Cyclopropyl(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)methyl]-4- Preparation of Methyl-4H-1,2,4 Triazole (63G)
  • Step 8 3-(3-(Cyclopropyl(4-methyl-4H-1,2,4-triazol-3-yl)methyl)phenyl)-7-(((S)-3- Preparation of methylpiperidin-1-yl)methyl)-9-trifluoromethyl-4H-pyrido[1,2-a]pyrimidin-4-one (compound 63)
  • the crude product is prepared with a high performance liquid chromatography column (column: Phenomenex C18, column length 80mm, inner diameter 40mm, particle diameter 3 ⁇ m; mobile phase A: water (0.1% ammonia water), mobile phase B: acetonitrile; gradient: mobile phase B takes 8 minutes from 51% to 81%, and then eluted with 100% mobile phase B for 4 minutes; flow rate: 30mL/min) separation,
  • the title compound 63 was obtained (3.49 mg, yield: 6%).
  • Step 4 3-(3-(Cyclopropyl(4-methyl-4H-1,2,4-triazol-3-yl)methyl)phenyl)-8-methyl-6-((( Preparation of S)-3-methylpiperidin-1-yl)methyl)-4H-chromen-4-one (compound 64)
  • Step 2 Preparation of methyl 1-(5-bromopyridin-3-yl)-3-methylcyclobutane-1-carboxylate (65B)
  • Step 4 Preparation of 2-(1-(5-bromopyridin-3-yl)-3-methylcyclobutane-1-carbonyl)-N-methylhydrazine-1-thioamide (65D)
  • 65D (1.5 g, 4.20 mmol) was dissolved in aqueous sodium hydroxide solution (1.0 M, 25 mL), nitrogen was replaced three times, and the mixture was stirred at room temperature for 12 hours.
  • the reaction solution was adjusted to pH 3 with 1M hydrochloric acid aqueous solution, extracted with ethyl acetate (30mL ⁇ 3), the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated to dryness under reduced pressure to obtain the crude product of the title compound 65E (1.3g). used directly in the next reaction.
  • Step 6 Preparation of 3-bromo-5-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)pyridine (65F)
  • Step 7 3-(3-Methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)-5-(4,4,5,5- Preparation of tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (65G)
  • Step 8 6-Bromo-8-methyl-3-(5-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl) Preparation of pyridin-3-yl)-4H-chromen-4-one (65H)
  • the preparation method is the same as compound 58D. Starting from 65G (20 mg, 56.46 ⁇ mol), the title compound 65H (15 mg, yield: 57%) was finally obtained.
  • Step 9 (S)-8-Methyl-3-(5-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl) Preparation of pyridin-3-yl)-6-((3-methylpiperidin-1-yl)methyl)-4H-chromen-4-one (compound 65)
  • Step 1 to Step 3 Preparation of 1-(3-bromo-5-(trifluoromethyl)phenyl)-3-methylcyclobutanecarboxylic acid (66C)
  • step 1 to step 2 is the same as that of compound 65B, and the preparation method of step 3 is the same as that of 63C.
  • 2-(3-bromo-5-(trifluoromethyl)phenyl)acetic acid (1 g, 3.53 mmol)
  • the title compound 66C (170 mg, yield: 14%) was finally obtained through three-step reactions.
  • Step 4 to Step 7 4-Methyl-3-(3-methyl-1-(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2 Preparation of -yl)-5-(trifluoromethyl)phenyl)cyclobutyl)-4H-1,2,4-triazole (66G)
  • Step 8 (S)-8-Methyl-3-(3-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl) Preparation of -5-trifluoromethylphenyl)-6-((3-methylpiperidin-1-yl)methyl)-4H-chromene-4-one (compound 66)
  • Step 1 to Step 4 4-Methyl-3-(2-(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl )propane-2-yl)-4H-1,2,4-triazole (67D) preparation
  • step 4-step 7 of Example 10 the difference is that 66C is replaced by 2-(3-bromophenyl)-2-methylpropionic acid (250 mg, 1.03 mmol), and the title compound 67D ( 116 mg, yield: 35%).
  • Step 5 (S)-8-Methyl-3-(3-(2-(4-Methyl-4H-1,2,4-triazol-3-yl)propan-2-yl)phenyl) - Preparation of 6-((3-methylpiperidin-1-yl)methyl)-4H-chromen-4-one (67)
  • Step 1 Preparation of cis-1-(3-bromophenyl)-3-hydroxycyclobutanecarboxylic acid (68A)
  • Step 2 Preparation of cis-2-(1-(3-bromophenyl)-3-hydroxycyclobutanecarbonyl)-N-methylhydrazinocarbothioamide (68B)
  • 68A (10.1g, 37.25mmol) was dissolved in dichloromethane (100mL), and DIPEA (14.44g, 111.76mmol), T 3 P (17.78g, 55.88mmol) and 1-amino-3 -Methylthiourea (4.70g, 44.71mmol), the reaction solution was stirred overnight at room temperature. Quenched with water (100 mL), a white precipitate was precipitated, filtered, and the filter cake was collected to obtain the title compound 68B (10.32 g, yield: 77%) as a white solid.
  • Step 3 cis-3-(3-bromophenyl)-3-(5-mercapto-4-methyl-4H-1,2,4-triazol-3-yl)cyclobutanol (68C) preparation
  • 68B (10.32g, 28.81mmol) was dissolved in sodium hydroxide solution (1M, 173.87mL), replaced with nitrogen three times, and stirred overnight at room temperature.
  • the pH of the reaction solution was adjusted to 3 with 2M hydrochloric acid, a white precipitate was precipitated, filtered, and the filter cake was collected to obtain a white solid.
  • the filtrate was extracted with ethyl acetate (100 mL ⁇ 3), the organic phase was dried with anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation to obtain a white solid.
  • the title compound 68C (8.66 g, yield: 88%) was obtained in total as a white solid.
  • Step 4 Preparation of cis-3-(3-bromophenyl)-3-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutanol (68D)
  • the reaction solution was quenched with saturated sodium sulfite aqueous solution, extracted with dichloromethane (100mL ⁇ 3), a white precipitate was precipitated, filtered, the filter cake was dissolved in methanol (200mL), stirred at 60°C for half an hour, filtered, and the filtrate was concentrated to dryness under reduced pressure
  • the title compound 68D was obtained as a white solid (2.39 g, yield: 30%).
  • Step 5 Methanesulfonic acid-cis-3-(3-bromophenyl)-3-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl ester (68E) preparation
  • reaction solution was quenched with water (50mL), extracted with dichloromethane (50mL ⁇ 2), the organic phase was dried with anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation to obtain a crude product, which was subjected to normal phase column chromatography (0-11 % methanol/dichloromethane) to obtain the title compound 68E (2.18 g, yield: 73%).
  • Step 6 trans-3-(3-bromophenyl)-3-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutanecarbonitrile (68F) and cis-3 Preparation of -(3-bromophenyl)-3-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutanecarbonitrile (68G)
  • Step 7 to Step 8 trans-3-(3-(6-bromo-8-methyl-4-oxo-4H-chromen-3-yl)phenyl)-3-(4-methyl- Preparation of 4H-1,2,4-triazol-3-yl)cyclobutanecarbonitrile (68J)
  • Step 9 trans-3-(4-methyl-4H-1,2,4-triazol-3-yl)-3-(3-(8-methyl-6-(((S)-3 Preparation of -methylpiperidin-1-yl)methyl)-4-oxo-4H-chromen-3-yl)phenyl)cyclobutanecarbonitrile (compound 68)
  • Step 1 Preparation of 3-(trans-1-(3-bromophenyl)-3-fluorocyclobutyl)-4-methyl-4H-1,2,4-triazole (69A)
  • Step 2 to Step 3 6-Bromo-3-(3-(trans-3-fluoro-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl) Preparation of phenyl)-8-methyl-4H-chromen-4-one (69C)
  • step 7-step 8 of Example 9 Referring to the synthesis method of step 7-step 8 of Example 9, except that 65F was replaced by 69A (50 mg, 160.26 ⁇ mol), the title compound 69C (15 mg, yield: 20%) was obtained in the same way.
  • Step 1 Preparation of ethyl 3-(3-bromophenyl)oxetane-3-carboxylate (70A)
  • Step 3 Preparation of 2-(3-(3-bromophenyl)oxetane-3-carbonyl)-N-methylhydrazine-1-thioamide (70C)
  • Step 4 5-(3-(3-Bromophenyl)oxetan-3-yl)-4-methyl-4H-1,2,4-triazole-3-thiol (70D) preparation
  • Step 5 Preparation of 3-(3-(3-bromophenyl)oxetan-3-yl)-4-methyl-4H-1,2,4-triazole (70E)
  • Step 6 4-Methyl-3-(3-(3-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)oxa Preparation of cyclobutan-3-yl)-4H-1,2,4-triazole (70F)
  • Step 7 6-Bromo-8-methyl-3-(3-(3-(4-methyl-4H-1,2,4-triazol-3-yl)oxetan-3-yl ) phenyl) -4H-chromen-4-ketone (70G) preparation
  • Step 8 8-Methyl-3-(3-(2-(4-Methyl-4H-1,2,4-triazol-3-yl)oxetan-2-yl)phenyl) - Preparation of 6-(((S)-3-methylpiperidin-1-yl)methyl)-4H-chromen-4-one (compound 70)
  • the organic phase is dried with anhydrous sodium sulfate, filtered, and distilled under reduced pressure to obtain the crude product, and the crude product is used for high performance liquid chromatography (chromatographic column: Welch Xtimate C18; column length 150mm, inner diameter 30mm, particle diameter 5 ⁇ m; mobile phase A: water (containing 0.225% ammonia water); mobile phase B: acetonitrile; gradient: mobile phase B from 5% to 95% in 18 minutes; flow rate: 25 mL/min) for purification.
  • the title compound 70 (4.1 mg, 19%) was obtained.
  • Step 1 Preparation of methyl 2-(2-bromoethoxy)-2-(3-bromophenyl)acetate (71A)
  • Dissolve 71B (695.0 mg, 2.56 mmol) in acetonitrile (10.0 mL) at room temperature, add aqueous sodium hydroxide solution (10.0 mL, 3.0 M), and react at 65°C for 1.0 hour. After the reaction, the temperature was lowered to 0° C., a saturated aqueous citric acid solution was added to adjust the pH to acidic, and the mixture was extracted three times with dichloromethane. The organic phase was dried over anhydrous sodium sulfate, filtered, and distilled under reduced pressure to obtain the title crude product 71C (594.0 mg, yield: 90%), which was directly used in the next reaction without further purification.
  • 71C (594.0mg, 2.31mmol), 4-methylthiosemicarbazide (292.0mg, 2.77mmol), HATU (1.05g, 2.77mmol), potassium phosphate (1.05g, 2.77mmol) and DIPEA (896.0mg, 6.93 mmol, 1.21 mL) was added to DMF (6.0 mL), and reacted at room temperature for 1.0 hour. After the reaction was completed, water (60.0 mL) was added and extracted with dichloromethane (6.0 mL ⁇ 3). The extracted organic phase was dried over anhydrous sodium sulfate, filtered, and distilled under reduced pressure to obtain the title product 71D (720.0 mg, yield: 90%), which was directly used in the next reaction without further purification.
  • Step 5 5-(2-(3-Bromophenyl)oxetan-2-yl)-4-methyl-4H-1,2,4-triazole-3-thiol (71E) preparation
  • 71D (720.0 mg, 2.09 mmol) was added into an aqueous solution of sodium hydroxide (10.0 mL, 3.0 M), and the reaction solution was reacted at 80° C. for 1.0 hour. After the reaction, the temperature of the reaction liquid was lowered to 0° C., a saturated aqueous citric acid solution was added to adjust the pH to acidic, and the mixture was extracted three times with dichloromethane. The extracted organic phase was dried over anhydrous sodium sulfate, filtered, and distilled under reduced pressure to obtain the title product 71E (614.0 mg, yield: 90%), which was directly used in the next reaction without further purification.
  • 71E (614.0 mg, 1.88 mmol) was dissolved in dichloromethane (6.0 mL) at room temperature, and glacial acetic acid (0.5 mL) and hydrogen peroxide (3.0 mL) were slowly added dropwise to the reaction solution successively. The reaction solution was reacted at room temperature for 1.0 hour. After the reaction was completed, water (60.0 mL) was added and extracted with dichloromethane (6.0 mL ⁇ 3). The extracted organic phase was dried over anhydrous sodium sulfate, filtered, and distilled under reduced pressure to obtain the title product 71F (500.0 mg, yield: 90%), which was directly used in the next reaction without further purification.
  • Step 7 4-Methyl-3-(2-(3-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)oxa Preparation of cyclobutan-2-yl)-4H-1,2,4-triazole (71G)
  • Step 8 8-Methyl-3-(3-(2-(4-Methyl-4H-1,2,4-triazol-3-yl)oxetan-2-yl)phenyl) - Preparation of 6-(((S)-3-methylpiperidin-1-yl)methyl)-4H-chromen-4-one (compound 71)
  • the crude product is prepared through a high performance liquid chromatography column (column: Welch Xtimate C18, column length 150mm, inner diameter 30mm, particle diameter 5 ⁇ m; mobile phase A: water (0.225 % ammonia water), mobile phase B: acetonitrile; gradient: mobile phase B from 5% to 95% in 24 minutes) to obtain the title compound 71 (6.86 mg, yield: 37%).
  • Step 2 8-((Dimethylamino)methyl)-9-methyl-3-(3-(3-methyl-1-(4-methyl-4H-1,2,4-triazole Preparation of -3-yl)cyclobutyl)phenyl)-4H-pyrido[1,2-a]pyrimidin-4-one (compound 72)
  • Step 1 Preparation of 3-(3-bromophenyl)-3-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutan-1-one (73A)
  • Step 2 Preparation of 3-(1-(3-bromophenyl)-3,3-difluorocyclobutyl)-4-methyl-4H-1,2,4-triazole (73B)
  • Step 3 3-(3,3-Difluoro-1-(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzene Base) cyclobutyl) -4-methyl-4H-1,2,4-triazole (73C) preparation
  • Step 4 (S)-3-(3-(3,3-difluoro-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)phenyl) - Preparation of 8-methyl-6-((3-methylpiperidin-1-yl)methyl)-4H-chromen-4-one (compound 73)
  • Step 4 6-Bromo-8-cyclopropyl-3-(3-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl ) phenyl) -4H-chromen-4 ketone (74D) preparation
  • Step 5 (S)-8-Cyclopropyl-3-(3-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl ) phenyl)-6-((3-methylpiperidin-1-yl)methyl)-4H-chromen-4-one (compound 74)
  • the organic phase was dried with anhydrous sodium sulfate, filtered, and distilled under reduced pressure to obtain the crude product, which was prepared through a high performance liquid chromatography column (column: Welch Xtimate C18, column length 150mm, inner diameter 30mm, particle diameter 5 ⁇ m; mobile phase A: water (containing 0.225% ammonia water), mobile phase B: acetonitrile; gradient: mobile phase B from 5% to 95% took 24 minutes, flow rate: 24mL/min) to separate the title compound 74 (5.5mg, yield: 25.8%) .
  • Step 1 Preparation of (S)-1-(2-hydroxy-5-((3-methylpiperidin-1-yl)methyl)phenyl)ethan-1-one (75A)
  • Step 2 Preparation of (S)-1-(2-hydroxy-3-iodo-5-((3-methylpiperidin-1-yl)methyl)phenyl)ethan-1-one (75B)
  • 75A (0.95g, 3.84mmol) was dissolved in acetonitrile (50mL), then p-toluenesulfonic acid monohydrate (1.1g, 5.76mmol) and NIS (1.3g, 5.76mmol) were added, and the mixture was refluxed overnight under nitrogen protection.
  • Step 5 Preparation of (S)-8-cyclopropyl-3-iodo-6-((3-methylpiperidin-1-yl)methyl)-4H-chromen-4-one (75E)
  • Step 6 (S)-8-Cyclopropyl-3-(5-(3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl )Pyridin-3-yl)-6-((3-methylpiperidin-1-yl)methyl)-4H-chromene-4-one (compound 75) preparation
  • step 4 of Example 17 the difference is that 73C in step 4 was replaced by 65G (10.96 mg, 30.95 ⁇ mol), and 64C was replaced by 75E (13.10 mg, 30.95 ⁇ mol), and the title compound 75 (6.8 mg, yield: 42%).
  • n-butyllithium (190.56mg, 2.97mmol, 0.518mL) was added to a DME solution (20mL) of 4-methyl-1,2,4-triazole (247.01mg, 2.97mmol) ) mixture, stirred for 1 hour, m-bromobenzaldehyde (500mg, 2.7mmol) was added and reacted for 1h, then the temperature was slowly raised to 0°C, and stirred for 1h.
  • Step 4 3-((3-Bromophenyl)(cyclopropyl)fluoromethyl)-4-methyl-4H-1,2,4-triazole (120D)
  • Step 5 3-(Cyclopropylfluoro(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)methyl) -4-Methyl-4H-1,2,4-triazole (120E)
  • Step 6 3-(3-(Cyclopropylfluoro(4-methyl-4H-1,2,4-triazol-3-yl)methyl)phenyl)-8-methyl-6-(( S)-3-methylpiperidin-1-yl)methyl)-4H-chromene-4-one (compound 120)
  • Step 7 3-(3-((S)-Cyclopropylfluoro(4-methyl-4H-1,2,4-triazol-3-yl)methyl)phenyl)-8-methyl- 6-((S)-3-methylpiperidin-1-yl)methyl)-4H-chromen-4-one and 3-(3-((R)-cyclopropylfluoro(4-methyl -4H-1,2,4-triazol-3-yl)methyl)phenyl)-8-methyl-6-(((S)-3-methylpiperidin-1-yl)methyl) -4H-chromen-4-one
  • Example 21 7-fluoro-8-methyl-3-(3-(cis-3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl) Cyclobutyl)phenyl)-6-(((s)-3-methylpiperidin-1-yl)methyl)-4H-chromene-4-one (compound 121)
  • Step 1 cis-3-(1-(3-bromophenyl)-3-methylcyclobutyl)-4-methyl-4H-1,2,4-triazole (1A-P1) preparation
  • Step 2 4-Methyl-3-(cis-3-methyl-1-(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane -2-yl)phenyl)cyclobutyl)-4H-1,2,4-triazole (1B-P1)
  • Pd(PPh 3 ) 2 Cl 2 (826.77 mg, 1.18 mmol) was added to 121A (6.9 g), tributyl(1-ethoxyethylene)tin (15.340 g, 42.48 mmol, 14.35 mL) and In 1,4-dioxane (100mL), nitrogen was replaced three times, the mixture was heated to 100°C and stirred for 12 hours, after the reaction was completed, the reaction was returned to room temperature, and aqueous hydrochloric acid (2M, 13mL) was added and stirred for 1 hour.
  • reaction solution was fully quenched with an excess of sodium metabisulfite aqueous solution at 0°C (ice-water bath), dichloromethane (50mL ⁇ 3) Extraction, the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and purified by normal phase column chromatography (eluted with pure PE) to obtain the title compound 121C (2 g).
  • Step 4 Preparation of 1-(5-bromo-4-fluoro-2-hydroxy-3-methylphenyl)-3-(dimethylamino)-prop-2-en-1-one (121D)
  • Step 7 6-Bromo-7-fluoro-8-methyl-3-(3-(cis-3-methyl-1-(4-methyl-4H-1,2,4-triazole-3 -yl)cyclobutyl)phenyl)-4H-chromen-4-one (121G)
  • Step 8 7-Fluoro-8-methyl-3-(3-(cis-3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl) ring Butyl)phenyl)-6-(((s)-3-methylpiperidin-1-yl)methyl)-4H-chromene-4-one (compound 121)
  • Step 1 1-Methyl-N-((trifluoro- ⁇ 4 -boryl)methyl)cyclobutan-1-amine potassium salt (122A)
  • Step 2 7-Fluoro-8-methyl-3-(3-(cis-3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl) ring Butyl)phenyl)-6-(((1-methylcyclobutyl)amino)methyl)-4H-chromen-4-one (compound 122)
  • a mixture of 121G (10 mg, 20.73 ⁇ mol), 122A (23.12 mg), XphosPdG2 (2.95 mg, 3.76 ⁇ mol), Xphos (3.58 mg, 7.52 ⁇ mol) and potassium carbonate (10.39 mg, 75.17 ⁇ mol) was evacuated and filled with argon , water (0.5 mL) and 1,4-dioxane (2 mL) were added to the mixture and heated to 100° C. for 1 hour. After the reaction was completed, it was cooled to room temperature, and water (10 mL) and ethyl acetate (10 mL ⁇ 3) were added for extraction.
  • Step 3 1-(2-Hydroxy-3-methyl-5-(1-((1-methylcyclobutyl)amino)ethyl)phenyl)ethan-1-one (123C)
  • Step 4 3-(Dimethylamino)-1-(2-hydroxy-3-methyl-5-(1-((1-methylcyclobutyl)amino)ethyl)phenyl)propan-2- En-1-one (123D)
  • Step 5 3-Iodo-8-methyl-6-(1-((1-methylcyclobutyl)amino)ethyl)-4H-chromen-4-one (123E)
  • Step 6 8-Methyl-3-(3-(cis-3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)benzene Base)-6-(1-((1-methylcyclobutyl)amino)ethyl)-4H-chromen-4-one (compound 123)
  • Step 1 Preparation of (S)-7-fluoro-8-methyl-6-((3-methylpiperidin-1-yl)methyl)-4H-chromen-4-one (124A)
  • the crude product is prepared through a high performance liquid chromatography column (chromatographic column: Welch Xtimate C18; column length 150mm, inner diameter 30mm, particle diameter 5 ⁇ m; mobile phase A: water ( Contains 0.225% NH 3 ), mobile phase B: acetonitrile; gradient: mobile phase B from 5% to 95% in 18 minutes) to obtain the title compound 124A (150 mg).
  • chromatographic column Welch Xtimate C18; column length 150mm, inner diameter 30mm, particle diameter 5 ⁇ m; mobile phase A: water ( Contains 0.225% NH 3 ), mobile phase B: acetonitrile; gradient: mobile phase B from 5% to 95% in 18 minutes
  • Step 2 (S,E)-1-(4-fluoro-2-hydroxy-3-methyl-5-((3-methylpiperidin-1-yl)methyl)phenyl)-3-( Preparation of pyrrolidin-1-yl)prop-2-en-1-one (124B)
  • Step 3 (S)-7-Fluoro-3-iodo-8-methyl-6-((3-methylpiperidin-1-yl)methyl)-4H-chromen-4-one (124C) preparation of
  • the crude product is prepared through a high performance liquid chromatography column (chromatographic column: Welch Xtimate C18; column length 150mm, internal diameter 30mm, particle diameter 5 ⁇ m; mobile phase A: water (containing 0.225 % NH 3 ), mobile phase B: acetonitrile; gradient: mobile phase B from 5% to 95% in 18 minutes) to obtain the title compound 124C (80 mg).
  • chromatographic column Welch Xtimate C18; column length 150mm, internal diameter 30mm, particle diameter 5 ⁇ m; mobile phase A: water (containing 0.225 % NH 3 ), mobile phase B: acetonitrile; gradient: mobile phase B from 5% to 95% in 18 minutes
  • Step 4 3-(3-(Cyclopropyl(4-methyl-4H-1,2,4-triazol-3-yl)methyl)phenyl)-7-fluoro-8-methyl-6 Preparation of -(((S)-3-methylpiperidin-1-yl)methyl)-4H-chromen-4-one (compound 124)
  • the crude product is prepared through a high performance liquid chromatography column (chromatographic column: Welch Xtimate C18; column length 150mm, inner diameter 30mm, particle diameter 5 ⁇ m; mobile phase A: water ( Contains 0.225% NH 3 ), mobile phase B: acetonitrile; gradient: mobile phase B from 5% to 95% in 18 minutes) to obtain the title compound 124 (15 mg).
  • chromatographic column Welch Xtimate C18; column length 150mm, inner diameter 30mm, particle diameter 5 ⁇ m; mobile phase A: water ( Contains 0.225% NH 3 ), mobile phase B: acetonitrile; gradient: mobile phase B from 5% to 95% in 18 minutes
  • Example 25 8-Methyl-3-(3-(cis-3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl) Preparation of phenyl)-6-((1-methylcyclobutyl)amino)methyl)-4H-pyrano[2,3-c]pyridin-4-one (compound 125)
  • Step 4 Preparation of 2-methyl-6-(((1-methylcyclobutyl)amino)methyl)pyridin-3-ol (compound 125D)
  • Step 5 Preparation of 4-bromo-2-methyl-6-(((1-methylcyclobutyl)amino)methyl)pyridin-3-ol (Compound 125E)
  • Step 6 Preparation of 1-(3-hydroxy-2-methyl-6-(((1-methylcyclobutyl)amino)methyl)pyridin-4-yl)ethan-1-one (125F)
  • 125E (37.0 mg, 0.132 mmol), tributyl(1-ethoxyethylene) tin (95.5 mg, 0.264 mmol) were added to toluene (1.0 mL), and nitrogen was replaced. Under nitrogen atmosphere, bistriphenylphosphine palladium dichloride (9.25 mg, 13.2 ⁇ mol) was added, and the resulting mixture was heated to 100° C. under nitrogen protection and stirred for 16 hours.
  • Step 7 3-(Dimethylamino)-1-(3-hydroxy-2-methyl-6-((1-methylcyclobutyl)amino)methyl)pyridin-4-yl)propan-2- Preparation of en-1-one (125G)
  • 125F (29.0 mg, 117.0 ⁇ mol) was added to N,N-dimethylformamide dimethyl acetal (1.0 mL), and the reaction solution was heated to 100° C. and stirred for 1 hour. After the reaction, the reaction solution was directly distilled under reduced pressure to obtain the title compound 125G crude product (35.0 mg), which was directly used in the next reaction without further purification.
  • Step 8 3-iodo-8-methyl-6-(((1-methylcyclobutyl)amino)methyl)-4H-pyrano[2,3-c]pyridin-4-one (125H ) preparation
  • Step 9 8-Methyl-3-(3-(cis-3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)benzene Preparation of -6-((1-methylcyclobutyl)amino)methyl)-4H-pyrano[2,3-c]pyridin-4-one (compound 125)
  • 125H (30.0 mg, 77.9 ⁇ mol), 1B-P1 (33.0 mg, 93.5 ⁇ mol) and potassium carbonate (21.5 mg, 156.0 ⁇ mol) were added to a mixture of water (0.4 mL) and 1,4-dioxane (2.0 mL) in the mixture and replaced with nitrogen. Under nitrogen atmosphere, Pd(dppf)Cl 2 (6.0 mg, 8.0 ⁇ mol) was added, and the resulting mixture was heated to 100° C. under nitrogen protection, and stirred for 1.0 hour.
  • Step 1 Preparation of 1-(6-amino-5-(trifluoromethyl)pyridin-3-yl)ethan-1-one (126A)
  • Step 2 Preparation of 5-(1-((1-methylcyclobutyl)amino)ethyl)-3-(trifluoromethyl)pyridin-2-amine (126B)
  • Step 5 3-(3-(cis-3-methyl-1-(4-methyl-4H-1,2,4-triazol-3-yl)cyclobutyl)phenyl)-7- Preparation of (1-((1-methylcyclobutyl)amino)ethyl)-9-(trifluoromethyl)-4H-pyrido[1,2-a]pyrimidin-4-one (compound 126)
  • Example 27 8-methyl-6-(((S)-3-methylpiperidin-1-yl)methyl)-3-(3-(1,2,2,2-tetrafluoro-1 Preparation of -(4-methyl-4H-1,2,4-triazol-3-yl)ethyl)phenyl)-4H-chromen-4-one (compound 128)
  • reaction solution was transferred to room temperature for 0.5 hour reaction. After the reaction was completed, the temperature of the reaction solution was cooled to 0° C. in an ice bath, and then saturated aqueous ammonium chloride solution was added and stirred for ten minutes. After stirring, it was extracted three times with ethyl acetate (60.0 mL), the obtained organic phase was dried and concentrated, and the crude product was washed twice with petroleum ether (60.0 mL) to obtain the title compound 128A (250.0 mg).
  • Step 2 Preparation of 3-(1-(3-bromophenyl)-1,2,2,2-tetrafluoroethyl)-4-methyl-4H-1,2,4-triazole (128B)
  • Dissolve 128A (100.0mg, 0.3mmol) in dichloromethane (3.0mL), cool the reaction solution to 0°C, add DAST (0.079mL, 0.6mmol) dropwise to the reaction solution, after the dropwise addition, react The solution was transferred to room temperature for 0.5 hours. After the reaction, the temperature of the reaction solution was cooled to 0° C. in an ice bath, and then a saturated aqueous sodium bicarbonate solution was added and stirred for ten minutes.
  • Step 3 4-Methyl-3-(1,2,2,2-tetrafluoro-1-(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborin Preparation of Heterocyclopentane-2-yl)phenyl)ethyl)-4H-1,2,4-triazole (128C)
  • Step 4 8-Methyl-6-(((S)-3-methylpiperidin-1-yl)methyl)-3-(3-(1,2,2,2-tetrafluoro-1- Preparation of (4-methyl-4H-1,2,4-triazol-3-yl)ethyl)phenyl)-4H-chromen-4-one (compound 128)
  • Example 28 3-(3-(Cyclopropylfluoro(4-methyl-4H-1,2,4-triazol-3-yl)methyl)phenyl)-6-(((R)- 4,4-difluoro-3-methylpiperidin-1-yl)methyl)-8-methyl-4H-chromen-4-one (compound 129)
  • 6-bromo-8-methyl-4H-chromen-4-one 500 mg, 2.09 mmol
  • Pd( PPh3 ) 4 (241.68 mg, 209.15 ⁇ mol)
  • (tributyltin)methanol 2.01g, 6.27mmol
  • dioxane 5mL
  • 129A (100 mg, 525.78 ⁇ mol) was dissolved in DCM (2 mL), thionyl chloride (125.10 mg, 1.05 mmol, 76.38 ⁇ L) was dropped into the mixture and stirred at room temperature for 3 hours. After the reaction was completed, the reaction solution was concentrated to obtain the crude product of the title compound 129B (109.7 mg), which was directly used in the next step without purification.
  • Step 4 (R)-1-(5-((4,4-Difluoro-3-methylpiperidin-1-yl)methyl)-2-hydroxy-3-methylphenyl)-3- (Pyrrolidin-1-yl)prop-2-en-1-one (129D)
  • Step 5 (R)-6-((4,4-Difluoro-3-methylpiperidin-1-yl)methyl)-3-iodo-8-methyl-4H-chromen-4-one (129E)
  • Step 6 3-(3-(Cyclopropylfluoro(4-methyl-4H-1,2,4-triazol-3-yl)methyl)phenyl)-6-(((R)-4 ,4-difluoro-3-methylpiperidin-1-yl)methyl)-8-methyl-4H-chromene-4-one (129)
  • Eu-Ubquitin (Cisbio) was incubated with UbcH5B (ENZO), E1 (ENZO) at 37°C for 4 hours to prepare Eu-Ubquitin-UbcH5B.
  • Eu-Ubquitin-UbcH5B is aliquoted and stored at -80°C.
  • Cbl-b activity assays were performed in 384-well plates (Perkin Elmer).

Landscapes

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

Abstract

本申请描述了作为Cbl-b抑制剂的由通式(I)表示的并环化合物或其立体异构体或其药学上可接受的盐,包含所述通式(I)化合物或其立体异构体或其药学上可接受的盐的药物组合物,以及所述通式(I)化合物或其立体异构体或其药学上可接受的盐或所述药物组合物在预防或治疗由Cbl-b介导的疾病或病症中的用途。

Description

作为Cbl-b抑制剂的并环化合物
相关申请的交叉引用
本申请要求以下4件中国发明专利申请的权益和优先权,在此将它们的全部内容以援引的方式整体并入本文中:
2021年10月29日向中国国家知识产权局提交的第202111271704.9号专利申请;
2022年01月27日向中国国家知识产权局提交的第202210103242.8号专利申请;
2022年07月28日向中国国家知识产权局提交的第202210899553.X号专利申请;以及
2022年09月01日向中国国家知识产权局提交的第202211063507.2号专利申请。
技术领域
本申请涉及作为Cbl-b抑制剂的并环化合物或其立体异构体或其可药用盐、其制备方法、含有该化合物或其立体异构体或其可药用盐的药物组合物、以及所述化合物或其立体异构体或其可药用盐或所述药物组合物在预防或治疗由Cbl-b介导的疾病或病症中的用途。
背景技术
细胞内的信号级联传导通常由蛋白磷酸化进行调控,基因的表达和调控则受到表观遗传学的影响,而调控这些信号的蛋白质,其本身也受到细胞内蛋白质的“生成-降解”平衡的调控,以维持细胞的自稳态。目前所知蛋白质降解主要通过两种途径:溶酶体降解途径和泛素介导的蛋白酶体降解途径。其中泛素介导的途径是一个受到严格的时空调控的特异性蛋白质降解途径。泛素系统广泛存在于真核生物中,是精确的胞内蛋白质降解调控系统。泛素(ubiquitin,Ub)是一种存在于大多数真核细胞中的高度保守的小蛋白质。它的主要功能是标记需要分解掉的蛋白质,使其水解。泛素系统由泛素、26S蛋白酶体、多种酶(E1、E2、E3及去泛素酶等)组成。蛋白的泛素化通过泛素激活酶E1(ubactivating enzyme,E1)、泛素结合酶E2(Ub conjugating enzyme,E2)、泛素连接酶E3(Ub ligase,E3)参与的E1-E2-E3级联反应完成,随后泛素化的蛋白被26S蛋白酶体降解(A patent review of the ubiquitin ligase system:2015-201 Expert Opin Ther Pat.2018,28(12):919–937)。
人类基因组大约编码35个E2结合酶以及超过500个E3连接酶。Casitas B谱系淋巴瘤原癌基因-b(Casitas B-lineage lymphoma proto-oncogene-b,Cbl-b)是一个负反馈调节T细胞激活的E3连接酶。Cbl-b属于Cbl家族,该家族包括c-Cbl、Cbl-b以及Cbl-3(Cbl:many adaptations to regulate protein tyrosine kinases.Nat.Rev.Mol.Cell Biol.2001,2(4):294–307)。Cbl蛋白主要负调控T细胞活化、生长因子(例如表皮生长因子受体(EGFR)、c-KIT、血小板衍生生长因子受体(PDGFR)和非受体型酪氨酸激酶信号(如Src家族激酶和Zap70)(Regulating the regulator:Negative regulation of Cbl ubiquitin ligases.Trends Biochem Sci,2006,31(2):79–88;The Cbl family proteins:Ring leaders in regulation of cell signaling.J Cell Physiol.2006,209:21–43)。研究发现Cbl蛋白的功能性失活与人类癌症有关(Germline CBL mutations cause developmental abnormalities and predispose to juvenile myelomonocytic leukemia.Nat Genet,2010,42(9):794–800)。在三种Cbl蛋白中,Cbl-b在建立T细胞激活阈值和控制外周T细胞耐受性方面起着关键作用,越来越多的证据表明Cbl-b还调节先天免疫反应,并在宿主防御病原体中起重要作用。Cbl-b基因敲除小鼠显示出严重的自身免疫性疾病(Negative regulation of lymphocyte activation and autoimmunity by the molecular adaptor Cbl-b.Nature,2000,403:211–216)。因此,Cbl-b可以作为一个重要的免疫调节治疗靶点。
发明内容
本申请涉及式(I)化合物或其立体异构体或其药学上可接受的盐,
Figure PCTCN2022128426-appb-000001
其中,
Figure PCTCN2022128426-appb-000002
选自以下情况中的任一种:i)C=C-A 3,所述A 3选自CR 11aR 11b、NR 12、O或S;ii)A 1-C=A 3,所述A 1选自C或N,A 3选自CR 11c或N;iii)A 1-A 2-A 3,所述A 1和A 2独立地选自C(R 11) n或N,A 3选自CR 11aR 11b、NR 12、O或S;
n选自0或1;
R 11a、R 11b、R 11c、R 11、R 12彼此独立地选自H、卤素、OH、CN、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 6环烷基或C 3-C 6环烷基氧基,其中所述的C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 6环烷基或C 3-C 6环烷基氧基任选被R 11d取代;
Q环选自苯基、5-6元杂芳基或5-7元杂环基,所述苯基、5-6元杂芳基或5-7元杂环基任选被R 10取代;
R 10选自卤素、=O、OH、NH 2、NO 2、CN、C 1-C 6烷基、C 2-C 4烯基、C 2-C 4炔基、C 1-C 6烷氧基、C 3-C 6环烷基、C 3-C 6环烷基氧基、C 3-C 6环烷基-NH-、4-7元杂环基、4-7元杂环基氧基或4-7元杂环基-NH-,所述NH 2、C 1-C 6烷基、C 2-C 4烯基、C 2-C 4炔基、C 1-C 6烷氧基、C 3-C 6环烷基、C 3-C 6环烷基氧基、C 3-C 6环烷基-NH-、4-7元杂环基、4-7元杂环基氧基或4-7元杂环基-NH-任选被R 10a取代;
Y 1、Y 2、Y 3和Y 4独立选自CR b或N;
X选自卤素、CN、OH、COOH、CONH 2、C 1-C 6烷基、C 1-C 6烷氧基、
Figure PCTCN2022128426-appb-000003
其中C 1-C 6烷基或C 1-C 6烷氧基任选被R e取代,环B选自任选被R 3取代的以下基团:4-10元含氮杂环基或5-10元含氮杂芳基,环D选自任选被R 6取代的以下基团:C 3-C 10环烷基、4-10元杂环基、苯基或5-10元杂芳基,并且环D以非N原子与L相连,L选自键、-NR 7-、-NR 7CR 8R 9-、-O-、-C(=O)-、-C(=O)NH-或-CR 8R 9-;
R b选自H、卤素、OH、CN、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6烷硫基、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、NHC(O)(C 1-C 6烷基)、NHC(O)-O(C 1-C 6烷基)、N(C 1-C 6烷基)C(O)-O(C 1-C 6烷基)、NHS(O) 2(C 1-C 6烷基)、C 3-C 6环烷基、C 3-C 6环烷基-O-、C 3-C 6环烷基-NH-、N(C 3-C 6环烷基) 2、NHC(O)-C 3-C 6环烷基、NHS(O) 2-C 3-C 6环烷基、4-7元杂环基、4-7元杂环基氧基、4-7元杂环基-NH-、N(4-7元杂环基) 2、NHC(O)-4-7元杂环基、NHS(O) 2-4-7元杂环基、C 6-C 10芳基、C 6-C 10芳基氧基、C 6-C 10芳基-NH-、N(C 6-C 10芳基) 2、NHC(O)-C 6-C 10芳基、NHS(O) 2-C 6-C 10芳基、5-10元杂芳基、5-10元杂芳基氧基或5-10元杂芳基-NH-、N(5-10元杂芳基) 2、NHC(O)-5-10元杂芳基、NHS(O) 2-5-10元杂芳基,其中所述的C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6烷硫基、C 3-C 6环烷基、4-7元杂环基、C 6-C 10芳基或5-10元杂芳基任选被R 2a取代;
或者两个R b与其相连的C原子共同形成C 3-C 6环烯基、苯基、4-7元杂环基或5-6元杂芳基,所述C 3-C 6环烯基、苯基、4-7元杂环基或5-6元杂芳基任选被R 2a取代;
R 4、R 5、R 7、R 8和R 9彼此独立地选自H、卤素、OH、C 1-C 6烷基或C 1-C 6烷氧基,所述C 1-C 6烷基或C 1-C 6烷氧基任选被R 4a取代;
或者R 8、R 9及其相连的原子一起形成C 3-C 6环烷基或4-7元杂环基,所述C 3-C 6环烷基或4-7元杂环基任选进一步被R 8a取代;
或者R 4、R 5及其相连的原子一起形成C 3-C 6环烷基或4-7元杂环基,所述C 3-C 6环烷基或4-7元杂环基任选进一步被R 8a取代;或者当p取自2时,两个R 4及其相连的原子一起形成C 3-C 6环烷基或4-7元杂环基,所述C 3-C 6环烷基或4-7元杂环基任选进一步被R 8a取代;
或者R 4、R 5一起形成=O;
R 3和R 6独立地选自卤素、CN、=O、OH、NO 2、C 1-C 6烷基、OR 6a、SR 6a、N(R 6a) 2、S(O) 2R 6a、S(O) 2N(R 6a) 2、S(O)R 6a、S(O)N(R 6a) 2、C(O)R 6a、C(O)OR 6a、C(O)N(R 6a) 2、C(O)N(R 6a)OR 6a、OC(O)R 6a、OC(O)N(R 6a) 2、N(R 6a)C(O)OR 6a、N(R 6a)C(O)R 6a、N(R 6a)C(O)N(R 6a) 2、N(R 6a)C(NR 6a)N(R 6a) 2、N(R 6a)S(O) 2N(R 6a) 2、N(R 6a)S(O) 2R 6a、C 3-C 10环烷基、4-7元杂环基、6-10元芳基或5-10元杂芳基,其中C 1-C 6烷基、C 3-C 10环烷基、4-7元杂环基、C 6-C 10元芳基或5-10元杂芳基任选进一步被R 3a取代;
每一个R 6a独立地选自H、C 1-C 6烷基、苯基、4-7元杂环基或5-6元杂芳基,所述C 1-C 6烷基、苯基、4-7元杂环基或5-6元杂芳基任选进一步被R 6b取代,或者一个N原子上的2个R 6a与其相连的N一起形成4-7元杂环基或5-6元杂芳基,所述4-7元杂环基或5-6元杂芳基任选进一步被R 6b取代;
R 3a、R 4a、R 6b和R e独立选自卤素、OH、CN、=O、C 1-C 6烷基、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、COOH或C 1-C 6烷氧基,其中所述的C 1-C 6烷基或C 1-C 6烷氧基进一步任选被R f取代;
R f选自卤素、OH、=O、NH 2、NH(C 1-C 6烷基)或N(C 1-C 6烷基) 2
R 11d、R 2a和R 10a独立选自卤素、OH、CN、=O、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 1-C 6烷基、卤代C 1-C 6烷基、C 1-C 6烷氧基或卤代C 1-C 6烷氧基;
R 1和R 2独立选自H、卤素、CN、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 10环烷基或4-10元杂环基,其中所述的C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 10环烷基或4-10元杂环基任选被R 1a取代,
或者R 1、R 2与其连接的原子共同形成C 3-C 10环烷基或4-10元杂环基,所述C 3-C 10环烷基或4-10元杂环基任选被R 1b取代;
或者R 1、R b及其各自相连的原子和键共同形成C 3-C 6环烯基或4-7元杂环基,所述C 3-C 6环烯基或4-7元杂环基任选被R 1d取代;
R 1a和R 1b独立选自卤素、OH、CN、=O、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 1-C 6烷基或C 1-C 6烷氧基,所述C 1-C 6烷基或C 1-C 6烷氧基任选进一步被R 1c取代;
R 1c选自卤素、OH、CN、=O、NH 2或COOH;
W选自(CR 13R 14) kW 1;所述W 1选自5-10元杂芳基或4-10元杂环基,所述5-10元杂芳基、4-10元杂环基任选被R 15取代;R 13、R 14独立选自H、卤素、OH、C 1-C 6烷基或C 1-C 6烷氧基;R 15选自卤素、OH、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 1-C 6烷基、C 3-C 10环烷基或4-7元杂环基,其中所述的C 1-C 6烷基、C 3-C 10环烷基或4-7元杂环基任选被R 15a取代;
或者R 1与R 13及其各自相连的原子和键共同形成C 3-C 6环烷基或4-7元杂环基,所述C 3-C 6环烷基或4-7元杂环基任选被R 13a取代;
R 8a、R 13a和R 1d独立地选自卤素、OH、CN、C 1-C 6烷基或C 1-C 6烷氧基,所述C 1-C 6烷基或C 1-C 6烷氧基任选进一步被卤素取代;R 15a选自卤素、=O、OH、CN或C 1-C 6烷基;
k选自0或1;
p选自0、1或2。
在一些实施方案中,环D选自任选被R 6取代的以下基团:C 3-C 10环烷基、4-10元杂环基、苯基或5-10元杂芳基,并且环D以非N原子与L相连,L选自键、-NR 7-、-NR 7CH 2-、 -O-、-C(=O)-、-C(=O)NH-或-CR 8R 9-。
在一些实施方案中,R b选自H、卤素、OH、CN、C 1-C 6烷基、C 1-C 6烷氧基、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、NHC(O)(C 1-C 6烷基)、NHC(O)-O(C 1-C 6烷基)、N(C 1-C 6烷基)C(O)-O(C 1-C 6烷基)、NHS(O) 2(C 1-C 6烷基)、C 3-C 6环烷基、C 3-C 6环烷基-O-、C 3-C 6环烷基-NH-、N(C 3-C 6环烷基) 2、NHC(O)-C 3-C 6环烷基、NHS(O) 2-C 3-C 6环烷基、4-7元杂环基、4-7元杂环基氧基、4-7元杂环基-NH-、N(4-7元杂环基) 2、NHC(O)-4-7元杂环基、NHS(O) 2-4-7元杂环基、C 6-C 10芳基、C 6-C 10芳基氧基、C 6-C 10芳基-NH-、N(C 6-C 10芳基) 2、NHC(O)-C 6-C 10芳基、NHS(O) 2-C 6-C 10芳基、5-10元杂芳基、5-10元杂芳基氧基或5-10元杂芳基-NH-、N(5-10元杂芳基) 2、NHC(O)-5-10元杂芳基、NHS(O) 2-5-10元杂芳基,其中所述的C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 6环烷基、4-7元杂环基、C 6-C 10芳基或5-10元杂芳基任选被R 2a取代;或者两个R b与其相连的C原子共同形成C 3-C 6环烯基、苯基、4-7元杂环基或5-6元杂芳基,所述C 3-C 6环烯基、苯基、4-7元杂环基或5-6元杂芳基任选被R 2a取代。
在一些实施方案中,
Figure PCTCN2022128426-appb-000004
选自:i)C=C-A 3,所述A 3选自CR 11aR 11b、NR 12、O或S;或ii)A 1-C=A 3,所述A 1选自C或N,A 3选自CR 11c或N。
在一些实施方案中,
Figure PCTCN2022128426-appb-000005
选自A 1-C=A 3,所述A 1选自C或N,A 3选自CR 11c或N。
在一些实施方案中,
Figure PCTCN2022128426-appb-000006
选自A 1-C=A 3,所述A 1选自N,A 3选自CR 11c或N。
在一些实施方案中,
Figure PCTCN2022128426-appb-000007
选自C=C-A 3,所述A 3选自CR 11aR 11b、NR 12、O或S。
在一些实施方案中,
Figure PCTCN2022128426-appb-000008
选自C=C-A 3,所述A 3选自NR 12、O或S。
在一些实施方案中,
Figure PCTCN2022128426-appb-000009
选自C=C-A 3,所述A 3选自O。
在一些实施方案中,
Figure PCTCN2022128426-appb-000010
选自A 1-A 2-A 3,所述A 1和A 2独立地选自C(R 11) n或N,A 3选自NR 12、O或S。
在一些实施方案中,
Figure PCTCN2022128426-appb-000011
选自A 1-A 2-A 3,所述A 1选自N,A 2选自C(R 11) n,A 3选自NR 12
在一些实施方案中,R 11a、R 11b、R 11c、R 11、R 12彼此独立地选自H、卤素、OH、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 6环烷基或C 3-C 6环烷基氧基,其中所述的C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 6环烷基或C 3-C 6环烷基氧基任选被R 11d取代。
在一些实施方案中,R 11a、R 11b、R 11c、R 11、R 12彼此独立地选自H、卤素、OH、NH 2、C 1-C 6烷基或C 3-C 6环烷基,所述C 1-C 6烷基或C 3-C 6环烷基任选被R 11d取代。在一些实施方案中,R 11a、R 11b、R 11c、R 11、R 12彼此独立地选自H、卤素或C 1-C 6烷基。在一些实施方案中,R 11a、R 11b、R 11c、R 11、R 12彼此独立地选自H、F或甲基。
在一些实施方案中,R 11d选自卤素、OH、NH 2、C 1-C 6烷基或卤代C 1-C 6烷基。
在一些实施方案中,n选自0。
在一些实施方案中,R 11c选自H、卤素或C 1-C 6烷基,所述C 1-C 6烷基任选被R 11d取代。
在一些实施方案中,R 11c选自H或卤素。
在一些实施方案中,R 11c选自F。
在一些实施方案中,R 12选自H、卤素、OH、C 1-C 6烷基或C 3-C 6环烷基,所述C 1-C 6烷 基或C 3-C 6环烷基任选被R 11d取代。
在一些实施方案中,R 12选自H或C 1-C 3烷基。
在一些实施方案中,R 12选自H或甲基。
在一些实施方案中,R 12选自甲基。
在一些实施方案中,
Figure PCTCN2022128426-appb-000012
选自以下情况中的一种:i)C=C-A 3,所述A 3选自NH、N-CH 3、O或S;ii)A 1-C=A 3,所述A 1选自N,A 3选自N或C-F;iii)A 1-A 2-A 3,所述A 1选自N,A 2选自C,A 3选自N-CH 3
在一些实施方案中,
Figure PCTCN2022128426-appb-000013
选自以下情况中的一种:i)C=C-A 3,所述A 3选自N-CH 3、O或S;ii)A 1-C=A 3,所述A 1选自N,A 3选自N或C-F;iii)A 1-A 2-A 3,所述A 1选自N,A 2选自C,A 3选自N-CH 3
在一些实施方案中,
Figure PCTCN2022128426-appb-000014
选自A 1-C=A 3,所述A 1选自N,A 3选自N。
在一些实施方案中,Q环选自任选被R 10取代的以下基团:苯基、吡啶基、嘧啶基、吡唑基、咪唑基、噁唑基、异噁唑基、噻唑基、异噻唑基或5-7元杂环基,所述5-7元杂环基包含1或2个N作为杂原子。
在一些实施方案中,Q环选自任选被R 10取代的以下基团:苯基、
Figure PCTCN2022128426-appb-000015
Figure PCTCN2022128426-appb-000016
在一些实施方案中,Q环选自任选被R 10取代的以下基团:苯基、
Figure PCTCN2022128426-appb-000017
Figure PCTCN2022128426-appb-000018
在一些实施方案中,Q环选自任选被R 10取代的以下基团:苯基、
Figure PCTCN2022128426-appb-000019
Figure PCTCN2022128426-appb-000020
在一些实施方案中,Q环选自任选被R 10取代的
Figure PCTCN2022128426-appb-000021
在一些实施方案中,Q环选自任选被R 10取代的苯基。
在一些实施方案中,R 10选自卤素、=O、OH、NH 2、CN、C 1-C 6烷基、C 2-C 4烯基、C 2-C 4炔基、C 1-C 6烷氧基、C 3-C 6环烷基、C 3-C 6环烷基-O-、C 3-C 6环烷基-NH-、4-7元杂环基、4-7元杂环基氧基或4-7元杂环基-NH-,所述NH 2、C 1-C 6烷基、C 2-C 4烯基、C 2-C 4炔基、C 1-C 6烷氧基或C 3-C 6环烷基任选被R 10a取代。
在一些实施方案中,R 10选自卤素、=O、NH 2、CN、C 1-C 6烷基、C 2-C 4炔基、C 1-C 6烷氧基、C 3-C 6环烷基、C 3-C 6环烷基-O-或C 3-C 6环烷基-NH-,所述NH 2、C 1-C 6烷基、C 2-C 4炔基、C 1-C 6烷氧基或C 3-C 6环烷基任选被R 10a取代。在一些实施方案中,R 10选自卤素、=O、NH 2、C 1-C 6烷基、C 2-C 4炔基、C 1-C 6烷氧基、C 3-C 6环烷基、C 3-C 6环烷基-O-或C 3-C 6环烷基-NH-,所述NH 2、C 1-C 6烷基、C 2-C 4炔基、C 1-C 6烷氧基或C 3-C 6环烷基任选被R 10a取代。
在一些实施方案中,R 10选自卤素、=O、NH 2、C 1-C 6烷基、C 2-C 4炔基、C 1-C 6烷氧基、C 3-C 6环烷基,所述NH 2、C 1-C 6烷基、C 2-C 4炔基、C 1-C 6烷氧基或C 3-C 6环烷基任选被R 10a取代。
在一些实施方案中,R 10选自卤素、NH 2、CN、C 1-C 6烷基、C 1-C 6烷氧基或C 3-C 6环烷基,所述NH 2或C 1-C 6烷基任选被R 10a取代。在一些实施方案中,R 10选自卤素、任选地被卤素取代的C 1-C 6烷基、C 1-C 6烷氧基或C 3-C 6环烷基。
在一些实施方案中,R 10a选自卤素、OH、NH 2、C 1-C 6烷基、卤代C 1-C 6烷基、C 1-C 6烷氧基或卤代C 1-C 6烷氧基。
在一些实施方案中,R 10a选自卤素、OH、C 1-C 6烷基或卤代C 1-C 6烷基。在一些实施方案中,R 10a选自卤素或C 1-C 6烷基。
在一些实施方案中,R 10a选自F或CH 3
在一些实施方案中,R 10选自=O、F、Cl、Br、CN、甲基、乙基、乙炔基、CF 3、CHF 2、甲氧基、N(CH 3) 2、环丙基、环丙基-O-或环丙基-NH-。
在一些实施方案中,R 10选自=O、F、Cl、乙炔基、CF 3、CHF 2、甲氧基、N(CH 3) 2、环丙基、环丙基-O-或环丙基-NH-。
在一些实施方案中,R 10选自=O、F、Cl、Br、CN、甲基、乙基、乙炔基、CF 3、甲氧基、N(CH 3) 2或环丙基。
在一些实施方案中,R 10选自=O、F、Cl、乙炔基、CF 3、甲氧基、N(CH 3) 2或环丙基。
在一些实施方案中,R 10选自F、Cl、甲基、Br、乙基、CF 3、甲氧基或环丙基。
在一些实施方案中,
Figure PCTCN2022128426-appb-000022
选自
Figure PCTCN2022128426-appb-000023
Figure PCTCN2022128426-appb-000024
Figure PCTCN2022128426-appb-000025
在一些实施方案中,
Figure PCTCN2022128426-appb-000026
选自
Figure PCTCN2022128426-appb-000027
Figure PCTCN2022128426-appb-000028
在一些实施方案中,
Figure PCTCN2022128426-appb-000029
选自
Figure PCTCN2022128426-appb-000030
Figure PCTCN2022128426-appb-000031
Figure PCTCN2022128426-appb-000032
在一些实施方案中,
Figure PCTCN2022128426-appb-000033
选自
Figure PCTCN2022128426-appb-000034
Figure PCTCN2022128426-appb-000035
在一些实施方案中,
Figure PCTCN2022128426-appb-000036
选自
Figure PCTCN2022128426-appb-000037
Figure PCTCN2022128426-appb-000038
在一些实施方案中,
Figure PCTCN2022128426-appb-000039
选自
Figure PCTCN2022128426-appb-000040
Figure PCTCN2022128426-appb-000041
在一些实施方案中,
Figure PCTCN2022128426-appb-000042
选自
Figure PCTCN2022128426-appb-000043
Figure PCTCN2022128426-appb-000044
Figure PCTCN2022128426-appb-000045
在一些实施方案中,
Figure PCTCN2022128426-appb-000046
选自
Figure PCTCN2022128426-appb-000047
Figure PCTCN2022128426-appb-000048
在一些实施方案中,
Figure PCTCN2022128426-appb-000049
选自
Figure PCTCN2022128426-appb-000050
Figure PCTCN2022128426-appb-000051
在一些实施方案中,
Figure PCTCN2022128426-appb-000052
选自
Figure PCTCN2022128426-appb-000053
在一些实施方案中,
Figure PCTCN2022128426-appb-000054
选自
Figure PCTCN2022128426-appb-000055
在一些实施方案中,
Figure PCTCN2022128426-appb-000056
选自
Figure PCTCN2022128426-appb-000057
在一些实施方案中,Y 1、Y 2和Y 4独立选自CR b或N,Y 3选自CR b
在一些实施方案中,Y 1、Y 2独立选自CR b或N,Y 3和Y 4独立选自CR b
在一些实施方案中,Y 1、Y 2、Y 3和Y 4均为CR b
在一些实施方案中,Y 1和Y 2均为N,Y 3和Y 4独立选自CR b
在一些实施方案中,Y 1为N,Y 2、Y 3和Y 4独立选自CR b
在一些实施方案中,Y 1、Y 2、Y 3均为CR b、Y 4为N。
在一些实施方案中,Y 1、Y 2和Y 3均为CH以及Y 4为CR b;或者,Y 1为N,Y 2和Y 3均为CH以及Y 4为CR b;或者,Y 1、Y 2、Y 3均为CH以及Y 4为N。
在一些实施方案中,R b选自H、卤素、CN、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6烷硫基、NH 2、NH(C 1-C 6烷基)、C 3-C 6环烷基、C 3-C 6环烷基-O-、C 3-C 6环烷基-NH-、4-7元杂环基、4-7元杂环基氧基、4-7元杂环基-NH-、5-10元杂芳基、5-10元杂芳基氧基或5-10元杂芳基-NH-,其中所述的C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6烷硫基、C 3-C 6环烷基、4-7元杂环基或5-10元杂芳基任选被R 2a取代。
在一些实施方案中,R b选自H、卤素、OH、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6烷硫基、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 3-C 6环烷基-O-、C 3-C 6环烷基-NH-、4-7元杂环基-O-、4-7元杂环基-NH-或5-10元杂芳基,其中所述的C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6烷硫基、C 3-C 6环烷基、4-7元杂环基或5-10元杂芳基任选被R 2a取代。
在一些实施方案中,R b选自H、卤素、OH、C 1-C 6烷基、C 1-C 6烷氧基、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 3-C 6环烷基-O-、C 3-C 6环烷基-NH-、4-7元杂环基-O-、4-7元杂环基-NH-或5-10元杂芳基,其中所述的C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 6环烷基、4-7元杂环基或5-10元杂芳基任选被R 2a取代。
在一些实施方案中,R b选自H、卤素、CN或任选被R 2a取代的以下基团:甲基、乙氧基、NHCH 3、NHEt、NH(i-Pr)、吡唑基、环丙基-O-、环丁基-NH-、氧杂环丁基-O-、环丙基、SCH 3
在一些实施方案中,R b选自H、卤素或任选被R 2a取代的以下基团:甲基、乙氧基、NHCH 3、NHEt、NH(i-Pr)、吡唑基、环丙基-O-、环丁基-NH-或氧杂环丁基-O-。
在一些实施方案中,R 2a选自卤素、OH、=O、C 1-C 3烷基或C 1-C 3烷氧基。
在一些实施方案中,R 2a选自卤素、OH或C 1-C 3烷基。
在一些实施方案中,R 2a选自F、OH或甲基。
在一些实施方案中,R b选自H、F、CN、CH 3、CF 3、OEt、
Figure PCTCN2022128426-appb-000058
Figure PCTCN2022128426-appb-000059
NHCH 3、NHEt、NH(i-Pr)、SCH 3
Figure PCTCN2022128426-appb-000060
在一些实施方案中,R b选自H、F、CF 3、OEt、
Figure PCTCN2022128426-appb-000061
NHCH 3、NHEt、NH(i-Pr)、
Figure PCTCN2022128426-appb-000062
在一些实施方案中,R b选自H、F、CF 3、CN、CH 3
Figure PCTCN2022128426-appb-000063
OEt、NHEt或SCH 3。在一些实施方案中,R b选自H或CF 3
在一些实施方案中,R b、R 1及其各自相连的原子和键共同形成C 3-C 6环烯基或4-7元杂环基,所述C 3-C 6环烷基或4-7元杂环基任选被R 1d取代。
在一些实施方案中,R b、R 1及其各自相连的原子和键共同形成任选被R 1d取代的C 3-C 6环 烯基。
在一些实施方案中,R 1d选自卤素、OH、C 1-C 3烷基或卤代C 1-C 3烷基。
在一些实施方案中,R b、R 1及其各自相连的原子和键共同形成环戊烯基。
在一些实施方案中,
Figure PCTCN2022128426-appb-000064
选自
Figure PCTCN2022128426-appb-000065
在一些实施方案中,
Figure PCTCN2022128426-appb-000066
选自
Figure PCTCN2022128426-appb-000067
Figure PCTCN2022128426-appb-000068
在一些实施方案中,
Figure PCTCN2022128426-appb-000069
选自
Figure PCTCN2022128426-appb-000070
Figure PCTCN2022128426-appb-000071
在一些实施方案中,
Figure PCTCN2022128426-appb-000072
选自
Figure PCTCN2022128426-appb-000073
在一些实施方案中,X选自
Figure PCTCN2022128426-appb-000074
或任选被R e取代的C 1-C 6烷基。
在一些实施方案中,X选自
Figure PCTCN2022128426-appb-000075
在一些实施方案中,X选自
Figure PCTCN2022128426-appb-000076
在一些实施方案中,环B选自任选被R 3取代的5-10元含氮杂芳基、4-7元单环含氮杂环基或6-10元含氮杂环基。
在一些实施方案中,环B选自任选被R 3取代的以下基团:四氢吡咯基、哌啶基、哌嗪基、吗啉基、
Figure PCTCN2022128426-appb-000077
Figure PCTCN2022128426-appb-000078
在一些实施方案中,环B选自任选被R 3取代的以下基团:四氢吡咯基、哌啶基、哌嗪基、吗啉基、
Figure PCTCN2022128426-appb-000079
Figure PCTCN2022128426-appb-000080
在一些实施方案中,环B选自任选被R 3取代的以下基团:四氢吡咯基、哌啶基、哌嗪基、吗啉基、
Figure PCTCN2022128426-appb-000081
Figure PCTCN2022128426-appb-000082
在一些实施方案中,环B选自任选被R 3取代的以下基团:四氢吡咯基、哌啶基、哌嗪基、吗啉基、
Figure PCTCN2022128426-appb-000083
Figure PCTCN2022128426-appb-000084
在一些实施方案中,R 3选自卤素、OH、=O、CN、C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 6环烷基、C 6-C 10芳基或5-10元杂芳基,所述C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 6环烷基、C 6-C 10芳基或5-10元杂芳基任选进一步被R 3a取代。
在一些实施方案中,R 3选自卤素、OH、=O、C 1-C 3烷基、C 3-C 6环烷基或苯基,所述C 1-C 3烷基、C 3-C 6环烷基或苯基任选进一步被R 3a取代。
在一些实施方案中,R 3a选自卤素、OH、=O或C 1-C 3烷氧基。
在一些实施方案中,R 3a选自F。
在一些实施方案中,R 3选自=O、OH、F、甲基、异丙基、CF 3、环丙基或苯基。
在一些实施方案中,R 3选自=O、OH、F、甲基、CF 3、环丙基或苯基。
在一些实施方案中,R 4、R 5独立选自H、卤素、OH或任选被R 4a取代的C 1-C 3烷基;或者R 4、R 5及其相连的原子一起形成任选被R 8a取代的C 3-C 6环烷基;或者R 4、R 5一起形成=O;或者当p取自2时,两个R 4及其相连的原子一起形成任选被R 8a取代的C 3-C 6环烷基。
在一些实施方案中,R 4、R 5独立选自H、卤素、OH或任选被R 4a取代的C 1-C 3烷基;或者R 4、R 5及其相连的原子一起形成任选被R 8a取代的C 3-C 6环烷基;或者R 4、R 5一起形成=O。
在一些实施方案中,R 4a选自卤素、OH或C 1-C 3烷氧基。
在一些实施方案中,R 4a选自F或OH。
在一些实施方案中,R 8a选自卤素、OH或C 1-C 3烷基。
在一些实施方案中,R 4、R 5独立选自H、甲基、羟甲基或CF 3,或者R 4、R 5及其相连的原子一起形成环丙基,或者R 4、R 5一起形成=O。
在一些实施方案中,p选自1。
在一些实施方案中,p选自0。
在一些实施方案中,p选自2,且两个R 4及其相连的原子一起形成任选被R 8a取代的C 3-C 6环烷基。
在一些实施方案中,p选自2,且两个R 4及其相连的原子一起形成环丙基或环丁基。
在一些实施方案中,
Figure PCTCN2022128426-appb-000085
选自如下基团:
Figure PCTCN2022128426-appb-000086
Figure PCTCN2022128426-appb-000087
在一些实施方案中,
Figure PCTCN2022128426-appb-000088
选自如下基团:
Figure PCTCN2022128426-appb-000089
Figure PCTCN2022128426-appb-000090
Figure PCTCN2022128426-appb-000091
在一些实施方案中,
Figure PCTCN2022128426-appb-000092
选自如下基团:
Figure PCTCN2022128426-appb-000093
Figure PCTCN2022128426-appb-000094
在一些实施方案中,
Figure PCTCN2022128426-appb-000095
选自如下基团:
Figure PCTCN2022128426-appb-000096
Figure PCTCN2022128426-appb-000097
Figure PCTCN2022128426-appb-000098
在一些实施方案中,
Figure PCTCN2022128426-appb-000099
选自如下基团:
Figure PCTCN2022128426-appb-000100
Figure PCTCN2022128426-appb-000101
在一些实施方案中,环D选自任选被R 6取代的以下基团:C 3-C 6环烷基、5-6元杂芳基、4-7元单环含氮杂环基或6-10元含氮杂环基,并且环D以非N原子与L相连。
在一些实施方案中,环D选自任选被R 6取代的以下基团:环丙基、环丁基、环戊基、四氢吡咯基、哌啶基、哌嗪基、
Figure PCTCN2022128426-appb-000102
吡唑基、咪唑基、噁唑基、异噁唑基、三唑基、噻唑基、异噻唑基或吡啶基。
在一些实施方案中,环D选自任选被R 6取代的以下基团:环丁基、环戊基、哌啶基、吡啶基、
Figure PCTCN2022128426-appb-000103
在一些实施方案中,R 6选自卤素、OH、CN、=O或任选被R 3a取代的C 1-C 3烷基。
在一些实施方案中,R 6选自卤素、=O、OH或C 1-C 3烷基。
在一些实施方案中,R 6选自F或甲基。
在一些实施方案中,L选自键、-NR 7-、-NR 7CR 8R 9-、-O-或-CR 8R 9-。
在一些实施方案中,L选自键、-NR 7-、-NR 7CH 2-、-O-或-CR 8R 9-。
在一些实施方案中,R 7、R 8和R 9独立选自H、卤素、C 1-C 3烷基或OH。
在一些实施方案中,R 7、R 8和R 9独立选自H、卤素或C 1-C 3烷基(例如,甲基)。
在一些实施方案中,R 7、R 8和R 9独立选自H或卤素(例如,F)。
在一些实施方案中,R 7选自H。
在一些实施方案中,R 8、R 9选自H、甲基或F。
在一些实施方案中,L选自键、-NH-、-NHCH 2-、-NHCH(CH 3)-、-O-、-C(F) 2-或-CH 2-。
在一些实施方案中,L选自键、-NH-、-NHCH 2-、-O-、-C(F) 2-或-CH 2-。
在一些实施方案中,
Figure PCTCN2022128426-appb-000104
选自如下基团:
Figure PCTCN2022128426-appb-000105
Figure PCTCN2022128426-appb-000106
在一些实施方案中,
Figure PCTCN2022128426-appb-000107
选自如下基团:
Figure PCTCN2022128426-appb-000108
Figure PCTCN2022128426-appb-000109
在一些实施方案中,
Figure PCTCN2022128426-appb-000110
选自如下基团:
Figure PCTCN2022128426-appb-000111
Figure PCTCN2022128426-appb-000112
在一些实施方案中,
Figure PCTCN2022128426-appb-000113
选自
Figure PCTCN2022128426-appb-000114
在一些实施方案中,
Figure PCTCN2022128426-appb-000115
选自
Figure PCTCN2022128426-appb-000116
在一些实施方案中,X选自任选被R e取代的C 1-C 6烷基。
在一些实施方案中,R e选自卤素、OH、C 1-C 3烷基、NH 2、NH(C 1-C 3烷基)、N(C 1-C 3烷基) 2或C 1-C 3烷氧基,其中所述的C 1-C 3烷基或C 1-C 3烷氧基进一步任选被R f取代。
在一些实施方案中,R e选自C 1-C 3烷基、N(C 1-C 3烷基) 2或C 1-C 3烷氧基,其中所述的C 1-C 3烷基或C 1-C 3烷氧基进一步任选被R f取代。
在一些实施方案中,R f选自卤素、OH或N(C 1-C 6烷基) 2
在一些实施方案中,R f选自N(CH 3) 2
在一些实施方案中,R e选自N(CH 3) 2、CH 2N(CH 3) 2
Figure PCTCN2022128426-appb-000117
在一些实施方案中,X选自
Figure PCTCN2022128426-appb-000118
在一些实施方案中,X选自如下基团:
Figure PCTCN2022128426-appb-000119
Figure PCTCN2022128426-appb-000120
在一些实施方案中,X选自如下基团:
Figure PCTCN2022128426-appb-000121
Figure PCTCN2022128426-appb-000122
在一些实施方案中,X选自如下基团:
Figure PCTCN2022128426-appb-000123
Figure PCTCN2022128426-appb-000124
Figure PCTCN2022128426-appb-000125
在一些实施方案中,X选自如下基团:
Figure PCTCN2022128426-appb-000126
Figure PCTCN2022128426-appb-000127
Figure PCTCN2022128426-appb-000128
在一些实施方案中,X选自
Figure PCTCN2022128426-appb-000129
在一些实施方案中,X选自
Figure PCTCN2022128426-appb-000130
在一些实施方案中,X选自
Figure PCTCN2022128426-appb-000131
在一些实施方案中,R 1、R 2与其连接的原子共同形成C 3-C 8环烷基或4-10元杂环基,所述C 3-C 8环烷基或4-10元杂环基任选被R 1b取代。
在一些实施方案中,R 1、R 2与其连接的原子共同形成C 3-C 6环烷基或4-7元杂环基,所述C 3-C 6环烷基或4-7元杂环基任选被R 1b取代。
在一些实施方案中,R 1、R 2与其连接的原子共同形成任选被R 1b取代的如下基团:环丁基、螺[2,3]己基或氧杂环丁基。
在一些实施方案中,R 1b选自卤素、OH、CN、=O、NH 2、C 1-C 3烷基或C 1-C 3烷氧基,所述C 1-C 3烷基或C 1-C 3烷氧基任选被R 1c取代。
在一些实施方案中,R 1b选自卤素、CN、C 1-C 3烷基或C 1-C 3烷氧基,所述C 1-C 3烷基或C 1-C 3烷氧基任选被R 1c取代。
在一些实施方案中,R 1c选自卤素、OH或CN。
在一些实施方案中,R 1c选自CN。
在一些实施方案中,R 1b选自F、CN、甲基、甲氧基或CH 2CN。
在一些实施方案中,R 1和R 2独立选自H、卤素、CN、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 8环烷基或4-7元杂环基,所述C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 8环烷基或4-7元杂环基任选被R 1a取代,且所述C 3-C 8环烷基或4-7元杂环基可以是螺环、桥环或并环形式的。
在一些实施方案中,R 1和R 2独立选自H、卤素、CN、C 1-C 3烷基、C 1-C 3烷氧基或C 3-C 6环烷基,所述C 1-C 3烷基、C 1-C 3烷氧基或C 3-C 6环烷基任选被R 1a取代。
在一些实施方案中,R 1和R 2独立选自H、C 1-C 3烷基或C 3-C 6环烷基,所述C 1-C 3烷基或C 3-C 6环烷基任选被R 1a取代。
在一些实施方案中,R 1和R 2独立选自H或C 1-C 3烷基,所述C 1-C 3烷基任选被R 1a取代。
在一些实施方案中,R 1a选自卤素、OH、CN、=O、NH 2、C 1-C 3烷基或C 1-C 3烷氧基。
在一些实施方案中,R 1和R 2独立选自H、F、甲基、CF 3、环丙基、环丁基或
Figure PCTCN2022128426-appb-000132
或者R 1、R 2与其连接的原子共同形成如下基团:
Figure PCTCN2022128426-appb-000133
Figure PCTCN2022128426-appb-000134
在一些实施方案中,R 1和R 2独立选自H、甲基或环丁基,或者R 1、R 2与其连接的原子共同形成如下基团:
Figure PCTCN2022128426-appb-000135
在一些实施方案中,R 1和R 2独立选自H或甲基,或者R 1、R 2与其连接的原子共同形成如下基团:
Figure PCTCN2022128426-appb-000136
在一些实施方案中,R 1和R 2独立选自H、F、环丙基、甲基、或
Figure PCTCN2022128426-appb-000137
或者R 1、R 2与其连接的原子共同形成如下基团:
Figure PCTCN2022128426-appb-000138
在一些实施方案中,R 1、R 2与其连接的原子共同形成如下基团:
Figure PCTCN2022128426-appb-000139
Figure PCTCN2022128426-appb-000140
在一些实施方案中,R 1、R 2与其连接的原子共同形成如下基团:
Figure PCTCN2022128426-appb-000141
在一些实施方案中,R 1和R 2独立选自H、甲基或环丁基。
在一些实施方案中,R 1和R 2独立选自H或甲基。
在一些实施方案中,W选自-(CR 13R 14)W 1
在一些实施方案中,R 13、R 14独立选自H、卤素、OH或甲基。
在一些实施方案中,R 13、R 14均为H。
在一些实施方案中,R 1与R 13及其各自相连的原子和键共同形成任选被R 13a取代的C 3-C 6环烷基。
在一些实施方案中,R 13a选自C 1-C 3烷基。
在一些实施方案中,R 13a选自甲基。
在一些实施方案中,R 1与R 13及其各自相连的原子和键共同形成
Figure PCTCN2022128426-appb-000142
在一些实施方案中,W选自W 1
在一些实施方案中,W 1选自任选被R 15取代的5-10元杂芳基或6-10元杂环基。
在一些实施方案中,W 1选自任选被R 15取代的5-10元杂芳基。
在一些实施方案中,W 1选自任选被R 15取代的5元杂芳基或8元杂环基。
在一些实施方案中,W 1选自任选被R 15取代的5元杂芳基。
在一些实施方案中,W 1选自任选被R 15取代的如下基团:吡咯基、噻吩基、呋喃基、吡唑基、咪唑基、噻唑基、异噻唑基、噻二唑基、三氮唑基、噁唑基、异噁唑基、噁二唑基或6,7-二氢-5H-吡咯并[2,1-c][1,2,4]三唑基。
在一些实施方案中,W 1选自任选被R 15取代的如下基团:吡咯基、噻吩基、呋喃基、吡唑基、咪唑基、噻唑基、异噻唑基、噻二唑基、三氮唑基、噁唑基、异噁唑基或噁二唑基。
在一些实施方案中,W 1选自任选被R 15取代的如下基团:三氮唑基、噁唑基、异噁唑基、噁二唑基或6,7-二氢-5H-吡咯并[2,1-c][1,2,4]三唑基。
在一些实施方案中,W 1选自任选被R 15取代的如下基团:三氮唑基、噁唑基、异噁唑基或噁二唑基。
在一些实施方案中,R 15选自卤素、OH、NH 2、C 1-C 3烷基或C 3-C 6环烷基,所述C 1-C 3烷基或C 3-C 6环烷基任选被R 15a取代。
在一些实施方案中,R 15选自OH、C 1-C 3烷基或C 3-C 6环烷基,所述C 1-C 3烷基或C 3-C 6环烷基任选被R 15a取代。
在一些实施方案中,R 15选自甲基或环丙基,所述甲基或环丙基任选被R 15a取代。
在一些实施方案中,R 15a选自卤素、OH或CN。
在一些实施方案中,R 15a选自F。
在一些实施方案中,R 15选自甲基、CHF 2或环丙基。
在一些实施方案中,W 1选自如下基团:
Figure PCTCN2022128426-appb-000143
Figure PCTCN2022128426-appb-000144
在一些实施方案中,W 1选自如下基团:
Figure PCTCN2022128426-appb-000145
Figure PCTCN2022128426-appb-000146
在一些实施方案中,W选自
Figure PCTCN2022128426-appb-000147
在一些实施方案中,本申请的式(I)化合物或其立体异构体或其药学上可接受的盐选自式(II)化合物或其立体异构体或其药学上可接受的盐:
Figure PCTCN2022128426-appb-000148
其中,Y 1、Y 2、Y 3、Y 4、X、Q、W、R 1和R 2如上文所定义。
在一些实施方案中,本申请的式(I)化合物或其立体异构体或其药学上可接受的盐选自式(III)化合物或其立体异构体或其药学上可接受的盐:
Figure PCTCN2022128426-appb-000149
其中,Z 1、Z 2、Z 3独立地选自CH、CR 10或N;R 10、Y 1、Y 2、Y 3、Y 4、X、W、R 1和R 2如上文所定义。
在一些实施方案中,本申请的式(I)化合物或其立体异构体或其药学上可接受的盐选自式(IV)化合物或其立体异构体或其药学上可接受的盐:
Figure PCTCN2022128426-appb-000150
其中R 10、Y 1、Y 2、Y 3、Y 4、X、W、R 1和R 2如上文所定义。
在一些实施方案中,本申请的式(I)化合物或其立体异构体或其药学上可接受的盐选自式(V)化合物或其立体异构体或其药学上可接受的盐:
Figure PCTCN2022128426-appb-000151
其中,Z 1、Z 2、Z 3独立地选自CH、CR 10或N;A 3选自CR 11aR 11b、NR 12、O或S;R 10、R 11a、R 11b、R 12、Y 1、Y 2、Y 3、Y 4、X、W、R 1和R 2如上文所定义。
在一些实施方案中,本申请的式(I)化合物或其立体异构体或其药学上可接受的盐选自式(VI)化合物或其立体异构体或其药学上可接受的盐:
Figure PCTCN2022128426-appb-000152
其中,Z 1选自CH、CR 10或N;Z 2选自CH 2、CHR 10、NH或NR 10、O或S;R 10、Y 1、Y 2、Y 3、Y 4、X、W、R 1和R 2如上文所定义。
在一些实施方案中,本申请的式(I)化合物或其立体异构体或其药学上可接受的盐选自以下化合物或其药学上可接受的盐:
Figure PCTCN2022128426-appb-000153
Figure PCTCN2022128426-appb-000154
Figure PCTCN2022128426-appb-000155
Figure PCTCN2022128426-appb-000156
Figure PCTCN2022128426-appb-000157
Figure PCTCN2022128426-appb-000158
Figure PCTCN2022128426-appb-000159
Figure PCTCN2022128426-appb-000160
Figure PCTCN2022128426-appb-000161
Figure PCTCN2022128426-appb-000162
另一方面,本申请提供药物组合物,其包含本申请的式(I)化合物、式(II)化合物、式(III)化合物、式(IV)化合物、式(V)化合物、式(VI)化合物或其立体异构体或其药学上可接受的盐和药学上可接受的辅料。
另一方面,本申请提供治疗哺乳动物的由Cbl-b介导的疾病或病症的方法,包括对需要该治疗的哺乳动物,优选人类,给予治疗有效量的式(I)化合物、式(II)化合物、式(III)化合物、式(IV)化合物、式(V)化合物、式(VI)化合物或其立体异构体或其药学上可接受的盐、或其药物组合物。
另一方面,本申请提供治疗或预防哺乳动物的肿瘤或自身免疫性疾病的方法,包括对需要该治疗或预防的哺乳动物,优选人类,给予治疗有效量的式(I)化合物、式(II)化合物、式(III)化合物、式(IV)化合物、式(V)化合物、式(VI)化合物或其立体异构体或其药学上可接受的盐、或其药物组合物。
另一方面,本申请提供式(I)化合物、式(II)化合物、式(III)化合物、式(IV)化合物、式(V)化合物、式(VI)化合物或其立体异构体或其药学上可接受的盐、或其药物组合物在制备预防或者治疗由Cbl-b介导的疾病或病症的药物中的用途。
另一方面,本申请提供式(I)化合物、式(II)化合物、式(III)化合物、式(IV)化合物、式(V)化合物、式(VI)化合物或其立体异构体或其药学上可接受的盐、或其药物组合物在制备预防或者治疗肿瘤或自身免疫性疾病的药物中的用途。
另一方面,本申请提供式(I)化合物、式(II)化合物、式(III)化合物、式(IV)化合物、式(V)化合物、式(VI)化合物或其立体异构体或其药学上可接受的盐、或其药物组合物在预防或者治疗由Cbl-b介导的疾病或病症中的用途。
另一方面,本申请提供式(I)化合物、式(II)化合物、式(III)化合物、式(IV)化合物、式(V)化合物、式(VI)化合物或其立体异构体或其药学上可接受的盐、或其药物组合物在预防或者治疗肿瘤或自身免疫性疾病中的用途。
另一方面,本申请提供用于预防或者治疗哺乳动物,优选人类的由Cbl-b介导的疾病或病症的式(I)化合物、式(II)化合物、式(III)化合物、式(IV)化合物、式(V)化合物、式(VI)化合物或其立体异构体或其药学上可接受的盐、或其药物组合物。
另一方面,本申请提供用于预防或者治疗哺乳动物,优选人类的肿瘤或自身免疫性疾病的式(I)化合物、式(II)化合物、式(III)化合物、式(IV)化合物、式(V)化合物、式(VI)化合物或其立体异构体或其药学上可接受的盐、或其药物组合物。
在一些实施方案中,由Cbl-b介导的疾病或病症选自肿瘤或自身免疫性疾病。
术语定义和说明
除非另有说明,本申请中所用的术语具有下列含义,本申请中记载的基团和术语定义, 包括其作为实例的定义、示例性的定义、优选的定义、表格中记载的定义、实施例中具体化合物的定义等,可以彼此之间任意组合和结合。一个特定的术语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照本领域普通的含义去理解。当本文中出现商品名时,意在指代其对应的商品或其活性成分。
本文中
Figure PCTCN2022128426-appb-000163
表示连接位点。
本文中,由实线和虚线描绘的键
Figure PCTCN2022128426-appb-000164
表示单键或双键。
本文中消旋体或者对映体纯的化合物的图示法来自Maehr,J.Chem.Ed.1985,62:114-120。除非另有说明,用楔形键和虚楔键
Figure PCTCN2022128426-appb-000165
表示一个立体中心的绝对构型,用黑实键和虚键
Figure PCTCN2022128426-appb-000166
表示一个立体中心的相对构型(如脂环化合物的顺反构型)。
术语“互变异构体”是指因分子中某一原子在两个位置迅速移动而产生的官能团异构体。本申请化合物可表现出互变异构现象。互变异构的化合物可以存在两种或多种可相互转化的种类。互变异构体一般以平衡形式存在,尝试分离单一互变异构体时通常产生一种混合物,其理化性质与化合物的混合物是一致的。平衡的位置取决于分子内的化学特性。例如,在很多脂族醛和酮如乙醛中,酮型占优势;而在酚中,烯醇型占优势。本申请包含化合物的所有互变异构形式。
术语“立体异构体”是指由分子中原子在空间上排列方式不同所产生的异构体,包括顺反异构体、对映异构体和非对映异构体。
本申请的化合物可以具有不对称原子如碳原子、硫原子、氮原子、磷原子或不对称双键,因此本申请的化合物可以存在特定的几何或立体异构体形式。特定的几何或立体异构体形式可以是顺式和反式异构体、E型和Z型几何异构体、(-)-和(+)-对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,以及其外消旋混合物或其它混合物,例如对映异构体或非对映体富集的混合物,以上所有这些异构体以及它们的混合物都属于本申请化合物的定义范围之内。烷基等取代基中可存在另外的不对称碳原子、不对称硫原子、不对称氮原子或不对称磷原子,所有取代基中涉及到的这些异构体以及它们的混合物,也均包括在本申请化合物的定义范围之内。本申请的含有不对称原子的化合物可以以光学活性纯的形式或外消旋形式被分离出来,光学活性纯的形式可以从外消旋混合物拆分,或通过使用手性原料或手性试剂合成。
术语“被取代”是指特定原子上的任意一个或多个氢原子被取代基取代,只要特定原子的价态是正常的并且取代后的化合物是稳定的。当取代基为氧代(即=O)时,意味着两个氢原子被取代,氧代不会发生在芳香基上。
术语“任选”或“任选地”是指随后描述的事件或情况可以发生或不发生,该描述包括发生所述事件或情况和不发生所述事件或情况。例如,乙基“任选”被卤素取代,是指乙基可以是未被取代的(CH 2CH 3)、单取代的(CH 2CH 2F、CH 2CH 2Cl等)、多取代的(CHFCH 2F、CH 2CHF 2、CHFCH 2Cl、CH 2CHCl 2等)或完全被取代的(CF 2CF 3、CF 2CCl 3、CCl 2CCl 3等)。本领域技术人员可理解,对于包含一个或多个取代基的任何基团,不会引入任何在空间上不可能存在和/或不能合成的取代或取代模式。
当任何变量(例如R 1a、R 2a)在化合物的组成或结构中出现一次以上时,其在每一种情况下的定义都是独立的。例如,如果一个基团被2个R 1a所取代,则每个R 1a都有独立的选项。
当一个连接基团的数量为0时,比如-(CR 13R 14) 0-,表示该连接基团为键。
当其中一个变量选自化学键或不存在时,表示其连接的两个基团直接相连,比如
Figure PCTCN2022128426-appb-000167
中L代表键时表示该结构实际上是
Figure PCTCN2022128426-appb-000168
当本文中涉及到的连接基团若没有指明其连接方向,则其连接方向是任意的。例如当结构单元
Figure PCTCN2022128426-appb-000169
中的L选自“-NR 7CH 2-”时,此时L既可以按照与从左到右的方向连接环D构成“环D-NR 7CH 2-”,也可以按照从右到左的方向连接环D构成“环D-CH 2NR 7-”。
本文中的C m-C n是指具有m-n范围中的整数个碳原子。例如“C 1-C 6”是指该基团可具有1个碳原子、2个碳原子、3个碳原子、4个碳原子、5个碳原子或6个碳原子。
术语“烷基”是指通式为C nH 2n+1的烃基,该烷基可以是直链或支链的。术语“C 1-C 6烷基”应理解为表示具有1、2、3、4、5或6个碳原子的直链或支链饱和一价烃基。所述烷基的具体实例包括但不限于甲基、乙基、丙基、丁基、戊基、己基、异丙基、异丁基、仲丁基、叔丁基、异戊基、2-甲基丁基、1-甲基丁基、1-乙基丙基、1,2-二甲基丙基、新戊基、1,1-二甲基丙基、4-甲基戊基、3-甲基戊基、2-甲基戊基、1-甲基戊基、2-乙基丁基、1-乙基丁基、3,3-二甲基丁基、2,2-二甲基丁基、1,1-二甲基丁基、2,3-二甲基丁基、1,3-二甲基丁基或1,2-二甲基丁基等。术语“C 1-C 3烷基”可理解为表示具有1、2或3个碳原子的直链或支链饱和一价烃基。所述“C 1-C 6烷基”可以包含“C 1-C 3烷基”。
术语“烷氧基”是指直链或支链醇类失去羟基上的氢原子产生的一价基团,可理解为“烷基氧基”或“烷基-O-”。术语“C 1-C 6烷氧基”可理解为“C 1-C 6烷基氧基”或“C 1-C 6烷基-O-”;术语“C 1-C 3烷氧基”可理解为“C 1-C 3烷基氧基”或“C 1-C 3烷基-O-”。所述“C 1-C 6烷氧基”可以进一步包含“C 1-C 3烷氧基”。
术语“烯基”是指由碳原子和氢原子组成的直链或支链的且具有至少一个双键的一价不饱和脂肪族烃基。术语“C 2-C 4烯基”应理解为表示直链或支链的不饱和一价烃基,其包含一个或多个双键并且具有2、3或4个碳原子,“C 2-C 4烯基”优选C 2或C 3烯基。应理解,在所述烯基包含多于一个双键的情况下,所述双键可相互分离或共轭。所述烯基的具体实例包括但不限于乙烯基、烯丙基、(E)-2-甲基乙烯基、(Z)-2-甲基乙烯基、(E)-丁-2-烯基、(Z)-丁-2-烯基、(E)-丁-1-烯基、(Z)-丁-1-烯基、异丙烯基、2-甲基丙-2-烯基、1-甲基丙-2-烯基、2-甲基丙-1-烯基、(E)-1-甲基丙-1-烯基或(Z)-1-甲基丙-1-烯基等。
术语“炔基”是指由碳原子和氢原子组成的直链或支链的具有至少一个三键的一价不饱和脂肪族烃基。术语“C 2-C 4炔基”可理解为表示直链或支链的不饱和一价烃基,其包含一个或多个三键并且具有2、3或4个碳原子。“C 2-C 4炔基”的实例包括但不限于乙炔基(-C≡CH)、丙炔基(-C≡CCH 3、-CH 2C≡CH)、丁-1-炔基、丁-2-炔基或丁-3-炔基。“C 2-C 4炔基”可以包含“C 2-C 3炔基”,“C 2-C 3炔基”实例包括乙炔基(-C≡CH)、丙-1-炔基(-C≡CCH 3)、丙-2-炔基(炔丙基)。
术语“环烷基”是指完全饱和的且以单环、并环、桥环或螺环等形式存在的碳环。除非另有指示,该碳环通常为3至10元环。术语“C 3-C 10环烷基”应理解为表示饱和的一价单环、并环、螺环或桥环,其具有3、4、5、6、7、8、9或10个碳原子。所述环烷基的具体实例包括但不限于环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环壬基、环癸基,降冰片基(双环[2.2.1]庚基)、双环[2.2.2]辛基、金刚烷基、螺[4.5]癸烷基等。术语“C 3-C 10环烷基”可以包含“C 3-C 8环烷基”,“C 3-C 8环烷基”可以包含“C 3-C 6环烷基”,“C 3-C 6环烷基”可以包含“C 3-C 4环烷基”。术语“C 3-C 6环烷基”可理解为表示饱和的一价单环或双环烃环,其具有3、4、5或6个碳原子,具体实例包括但不限于环丙基、环丁基、环戊基或环己基等。
术语“环烷基氧基”可理解为“环烷基-O-”。
术语“环烯基”是指含有至少一个碳-碳双键的非芳族单环或多环烃基。“C 3-C 6环烯基”是指具有3、4、5或6个碳原子作为环原子且包含至少一个碳-碳双键的非芳族环状烃基。C 3-C 6环烯基的具体实例包括但不限于环丙烯基、环丁烯基、环戊烯基、环戊二烯基、环己烯基、环己二烯基等。
术语“杂环基”是指完全饱和的或部分饱和的(整体上不是具有芳香性的杂芳族)一价单环、并环、螺环或桥环基团,其环原子中含有1、2、3、4或5个杂原子或杂原子团(即含有杂原 子的原子团),所述“杂原子或杂原子团”包括但不限于氮原子(N)、氧原子(O)、硫原子(S)、磷原子(P)、硼原子(B)、-S(=O) 2-、-S(=O)-以及任选被取代的-NH-、-S(=O)(=NH)-、-C(=O)NH-、-C(=NH)-、-S(=O) 2NH-、S(=O)NH-或-NHC(=O)NH-等。术语“4-10元杂环基”是指环原子数目为4、5、6、7、8、9或10的杂环基,且其环原子中含有1、2、3、4或5个独立选自上文所述的杂原子或杂原子团。术语“4-10元含氮杂环基”是指其环原子中至少含1个N原子的4、5、6、7、8、9或10元杂环基,“4-10元含氮杂环基”包括“6-10元含氮杂环基”。术语“4-7元单环含氮杂环基”是指单环形式的且环原子中至少含1个N原子的4、5、6或7元杂环基。“4-10元杂环基”包括“6-10元杂环基”,“6-10元杂环基”进一步包括“6-7元杂环基”,其中,4元杂环基的具体实例包括但不限于氮杂环丁烷基、硫杂环丁烷基或氧杂环丁烷基;5元杂环基的具体实例包括但不限于四氢呋喃基、二氧杂环戊烯基、吡咯烷基、咪唑烷基、吡唑烷基、吡咯啉基、4,5-二氢噁唑基或2,5-二氢-1H-吡咯基;6元杂环基的具体实例包括但不限于四氢吡喃基、哌啶基、吗啉基、二噻烷基、硫代吗啉基、哌嗪基、三噻烷基、四氢吡啶基或4H-[1,3,4]噻二嗪基;7元杂环基的具体实例包括但不限于二氮杂环庚烷基;8元杂环基的具体实例包括但不限于6,7-二氢-5H-吡咯并[2,1-c][1,2,4]三唑基。所述杂环基还可以是双环基,其中,5,5元双环基的具体实例包括但不限于六氢环戊并[c]吡咯-2(1H)-基;5,6元双环基的具体实例包括但不限于六氢吡咯并[1,2-a]吡嗪-2(1H)-基、5,6,7,8-四氢-[1,2,4]三唑并[4,3-a]吡嗪基或5,6,7,8-四氢咪唑并[1,5-a]吡嗪基。任选地,所述杂环基可以是上述4-7元杂环基的苯并稠合环基,具体实例包括但不限于二氢异喹啉基等。“4-10元杂环基”可以包含“5-10元杂环基”、“4-7元杂环基”、“5-7元杂环基”、“5-6元杂环基”、“6-8元杂环基”、“4-10元杂环烷基”、“5-10元杂环烷基”、“4-7元杂环烷基”、“5-6元杂环烷基”、“6-8元杂环烷基”等范围,“4-7元杂环基”进一步可以包含“4-6元杂环基”、“5-7元杂环基”、“5-6元杂环基”、“4-7元杂环烷基”、“4-6元杂环烷基”、“5-6元杂环烷基”等范围。本申请中尽管有些双环类杂环基部分地含有一个苯环或一个杂芳环,但所述杂环基整体上仍是无芳香性的。
术语“杂环基氧基”可理解为“杂环基-O-”。
术语“杂环烷基”是指完全饱和的且以单环、并环、桥环或螺环等形式存在的一价环状基团,其环的环原子中含有1、2、3、4或5个杂原子或杂原子团(即含有杂原子的原子团),所述“杂原子或杂原子团”包括但不限于氮原子(N)、氧原子(O)、硫原子(S)、磷原子(P)、硼原子(B)、-S(=O) 2-、-S(=O)-以及任选被取代的-NH-、-S(=O)(=NH)-、-C(=O)NH-、-C(=NH)-、-S(=O) 2NH-、S(=O)NH-或-NHC(=O)NH-等。术语“4-10元杂环烷基”是指环原子数目为4、5、6、7、8、9或10的杂环烷基,且其环原子中含有1、2、3、4或5个独立选自上文所述的杂原子或杂原子团。“4-10元杂环烷基”包括“4-7元杂环烷基”,其中,4元杂环烷基的具体实例包括但不限于吖丁啶基、噁丁环基或噻丁环基;5元杂环烷基的具体实例包括但不限于四氢呋喃基、四氢噻吩基、吡咯烷基、异噁唑烷基、噁唑烷基、异噻唑烷基、噻唑烷基、咪唑烷基或四氢吡唑基;6元杂环烷基的具体实例包括但不限于哌啶基、四氢吡喃基、四氢噻喃基、吗啉基、哌嗪基、1,4-噻噁烷基、1,4-二氧六环基、硫代吗啉基、1,3-二噻烷基或1,4-二噻烷基;7元杂环烷基的具体实例包括但不限于氮杂环庚烷基、氧杂环庚烷基或硫杂环庚烷基。
术语“芳基”是指具有共轭的π电子体系的全碳单环或稠合多环的芳香环基团。芳基可以具有6-14个碳原子或6-10个碳原子。术语“C 6-C 10芳基”应理解为具有6~10个碳原子的一价芳香性的单环或双环基团。特别是具有6个碳原子的环(“C 6芳基”),例如苯基;或者具有10个碳原子的环(“C 10芳基”),例如萘基。
术语“芳基氧基”可理解为“芳基-O-”。
术语“杂芳基”是指具有芳香性的单环或稠合多环体系,其中含有至少一个选自N、O、S的环原子,其余环原子为C的芳香环基。术语“5-10元杂芳基”应理解为包括这样的一价单环或双环芳族环系:其具有5、6、7、8、9或10个环原子,特别是5或6或9或10个环原子,且其包含1、2、3、4或5个,优选1、2或3个独立选自N、O和S的杂原子。特别地,杂芳基选自噻吩基、呋喃基、吡咯基、噁唑基、噻唑基、咪唑基、吡唑基、异噁唑基、异噻唑 基、噁二唑基、三唑基或噻二唑基等以及它们的苯并衍生物,例如苯并呋喃基、苯并噻吩基、苯并噻唑基、苯并噁唑基、苯并异噁唑基、苯并咪唑基、苯并三唑基、吲唑基、吲哚基或异吲哚基等;或吡啶基、哒嗪基、嘧啶基、吡嗪基或三嗪基等以及它们的苯并衍生物,例如喹啉基、喹唑啉基或异喹啉基等;或吖辛因基、吲嗪基、嘌呤基等以及它们的苯并衍生物;或噌啉基、酞嗪基、喹唑啉基、喹喔啉基、萘啶基、蝶啶基、咔唑基、吖啶基、吩嗪基、吩噻嗪基或吩噁嗪基等。术语“5-10元含氮杂芳基”是指其环原子中至少含1个N原子的5、6、7、8、9或10元杂芳基,“5-10元含氮杂芳基”包含“5-6元含氮杂芳基”。术语“5-6元杂芳基”指具有5或6个环原子的芳族环系,且其包含1、2或3个,优选1或2个独立选自N、O和S的杂原子。术语“6元杂芳基”指具有6个环原子的芳族环系,且其包含1、2或3个,优选1或2个N作为杂原子。术语“5-10元杂芳基”包含“5-6元杂芳基”。
术语“杂芳基氧基”可理解为“杂芳基-O-”。
术语“卤”或“卤素”是指氟(F)、氯(Cl)、溴(Br)或碘(I)。
术语“羟基”是指-OH基团。
术语“氰基”是指-CN基团。
术语“巯基”是指-SH基团。
术语“氨基”是指-NH 2基团。
术语“硝基”是指-NO 2基团。
术语“治疗有效量”意指(i)治疗或预防特定疾病、病况或病症,(ii)减轻、改善或消除特定疾病、病况或病症的一种或多种症状,或(iii)预防或延迟本文中所述的特定疾病、病况或病症的一种或多种症状发作的本申请化合物的用量。构成“治疗有效量”的本申请化合物的量取决于该化合物、疾病状态及其严重性、给药方式以及待被治疗的哺乳动物的年龄而改变,但可例行性地由本领域技术人员根据其自身的知识及本公开内容而确定。
术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”是指药学上可接受的酸或碱的盐,包括化合物与无机酸或有机酸形成的盐,以及化合物与无机碱或有机碱形成的盐。
术语“药物组合物”是指一种或多种本申请的化合物或其立体异构体或其盐与药学上可接受的辅料组成的混合物。药物组合物的目的是有利于对有机体给予本申请的化合物。
术语“药学上可接受的辅料”是指对有机体无明显刺激作用,而且不会损害该活性化合物的生物活性及性能的那些辅料。合适的辅料是本领域技术人员熟知的,例如碳水化合物、蜡、水溶性和/或水可膨胀的聚合物、亲水性或疏水性材料、明胶、油、溶剂、水等。
词语“包括(comprise)”或“包含(comprise)”及其英文变体例如comprises或comprising应理解为开放的、非排他性的意义,即“包括但不限于”。
本申请还包括与本文中记载的那些相同的,但一个或多个原子被原子量或质量数不同于自然中通常发现的原子量或质量数的原子置换的同位素标记的本申请化合物。可结合到本申请化合物的同位素的实例包括氢、碳、氮、氧、磷、硫、氟、碘和氯的同位素,诸如分别为 2H、 3H、 11C、 13C、 14C、 13N、 15N、 15O、 17O、 18O、 31P、 32P、 35S、 18F、 123I、 125I和 36Cl等。
某些同位素标记的本申请化合物(例如用 3H及 14C标记)可用于化合物和/或底物组织分布分析中。氚化(即 3H)和碳-14(即 14C)同位素对于由于它们易于制备和可检测性是尤其优选的。正电子发射同位素,诸如 15O、 13N、 11C和 18F可用于正电子发射断层扫描(PET)研究以测定底物占有率。通常可以通过与公开于下文的方案和/或实施例中的那些类似的下列程序,通过同位素标记试剂取代未经同位素标记的试剂来制备同位素标记的本申请化合物。
本申请的药物组合物可通过将本申请的化合物与适宜的药学上可接受的辅料组合而制备,例如可配制成固态、半固态、液态或气态制剂,如片剂、丸剂、胶囊剂、粉剂、颗粒剂、膏剂、乳剂、悬浮剂、栓剂、注射剂、吸入剂、凝胶剂、微球及气溶胶等。
给予本申请化合物或其立体异构体或其药学上可接受的盐或其药物组合物的典型途径包括但不限于口服、直肠、局部、吸入、肠胃外、舌下、阴道内、鼻内、眼内、腹膜内、肌内、皮下、静脉内给药。
本申请的药物组合物可以采用本领域众所周知的方法制造,如常规的混合法、溶解法、制粒法、乳化法、冷冻干燥法等。
在一些实施方案中,药物组合物是口服形式。对于口服给药,可以通过将活性化合物与本领域熟知的药学上可接受的辅料混合,来配制该药物组合物。这些辅料能使本申请的化合物被配制成片剂、丸剂、锭剂、糖衣剂、胶囊剂、液体、凝胶剂、浆剂、悬浮剂等,用于对患者的口服给药。
可以通过常规的混合、填充或压片方法来制备固体口服组合物。例如,可通过下述方法获得:将所述的活性化合物与固体辅料混合,任选地碾磨所得的混合物,如果需要则加入其它合适的辅料,然后将该混合物加工成颗粒,得到了片剂或糖衣剂的核心。适合的辅料包括但不限于:粘合剂、稀释剂、崩解剂、润滑剂、助流剂或矫味剂等。
药物组合物还可适用于肠胃外给药,如合适的单位剂型的无菌溶液剂、混悬剂或冻干产品。
本文所述的通式Ⅰ化合物的所有施用方法中,每天给药的剂量为0.01mg/kg到200mg/kg体重,优选为0.05mg/kg到50mg/kg体重,更优选0.1mg/kg到30mg/kg体重,以单独或分开剂量的形式。
本申请的化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施例、其与其它化学合成方法的结合所形成的实施方案以及本领域技术上人员所熟知的等同替换方式,优选的实施方案包括但不限于本申请的实施例。
本申请具体实施例的化学反应是在合适的溶剂中完成的,所述的溶剂须适合于本申请的化学变化及其所需的试剂和物料。为了获得本申请的化合物,有时需要本领域技术人员在已有实施例的基础上对合成步骤或者反应流程进行修改或选择。
在一些实施方案中,本申请通式(III)的部分化合物可以由有机合成领域技术人员通过以下合成路线1来制备:
Figure PCTCN2022128426-appb-000170
合成路线1
其中:X’为卤素或OTf;X 2为卤素;CG 1和CG 2独立地选自B(OR) 2、BF 3K、SnR’ 4、ZnX 2
Figure PCTCN2022128426-appb-000171
所述R选自H或C 1-C 6烷基,R’选自C 1-C 6烷基,X 2为卤素;Z 1、Z 2、Z 3、Y 1、Y 2、Y 3、Y 4、W、R 1、R 2、R 4和环B如通式(III)中所定义。
在一些实施方案中,本申请通式(III)的部分化合物可以由有机合成领域技术人员通过以下合成路线2来制备:
Figure PCTCN2022128426-appb-000172
合成路线2
其中:X’为卤素或OTf;X”为卤素;CG 1独立地选自B(OR) 2、BF 3K、SnR’ 4、ZnX”、
Figure PCTCN2022128426-appb-000173
所述R选自H或C 1-C 6烷基,R’选自C 1-C 6烷基,X”为卤素;Z 1、Z 2、Z 3、Y 1、Y 2、Y 3、Y 4、W、R 1、R 2、R 4和环B如通式(III)中所定义。
在一些实施方案中,本申请通式(VI)的部分化合物可以由有机合成领域技术人员通过以下合成路线3来制备:
Figure PCTCN2022128426-appb-000174
合成路线3
其中:X’为卤素或OTf;Z 1、Z 2、Y 1、Y 2、Y 3、Y 4、W、R 1、R 2、R 4和环B如通式(VI)中所定义。
在一些实施方案中,本申请通式(V)的部分化合物可以由有机合成领域技术人员通过以下合成路线4来制备:
Figure PCTCN2022128426-appb-000175
合成路线4
其中:X’为卤素或OTf;X”为卤素;CG 1独立地选自B(OR) 2、BF 3K、SnR’ 4、ZnX”、
Figure PCTCN2022128426-appb-000176
所述R选自H或C 1-C 6烷基,R’选自C 1-C 6烷基;Z 1、Z 2、Z 3、X、Y 1、Y 2、Y 3、Y 4、W、R 1、R 2、R 4和环B如通式(V)中所定义。
实施例
下面通过实施例对发明进行详细描述,但并不意味着对本申请的任何不利限制。本文已经详细地描述了发明,其中也公开了其具体实施方案,对本领域的技术人员而言,在不脱离本申请精神和范围的情况下针对本申请具体实施方案进行各种改变和改进将是显而易见的。本申请所使用的所有试剂是市售的,无需进一步纯化即可使用。
除非另作说明,混合溶剂表示的比例是体积混合比例。除非另作说明,否则,%是指wt%。
化合物经手工或
Figure PCTCN2022128426-appb-000177
软件命名,市售化合物采用供应商目录名称。
化合物的结构是通过核磁共振(NMR)和/或质谱(MS)来确定的。NMR位移的单位为10 - 6(ppm)。NMR测定的溶剂为氘代二甲基亚砜、氘代氯仿、氘代甲醇等,内标为四甲基硅烷(TMS);“IC 50”指半数抑制浓度,指达到最大抑制效果一半时的浓度。
下文的洗脱剂可由两种或多种溶剂形成混合洗脱剂,其比值为各溶剂的体积比,如“0~10% 甲醇/二氯甲烷”表示梯度洗脱过程中,混合洗脱剂中的甲醇与二氯甲烷的体积用量比为0:100~10:90。
术语或缩写说明:
B 2Pin 2:联硼酸频哪醇酯;Pd(dppf)Cl 2:[1,1'-双(二苯基膦)二茂铁]二氯化钯;NIS:N-碘代丁二酰亚胺;DCM:二氯甲烷;Pd(PPh 3) 4:四三苯基膦钯;THF:四氢呋喃;XphosPdG 2:氯(2-二环己基膦基-2',4',6'-三异丙基-1,1'-联苯基)[2-(2'-氨基-1,1'-联苯)]钯(II);Xphos:2-二环己基膦-2',4',6'-三异丙基联苯;SFC:超临界流体色谱;(Boc) 2O:二叔丁基二碳酸酯;DAMP:N,N-二甲基吡啶-4-胺;TFA:三氟乙酸;Pd(dba) 2:双二亚苄基丙酮钯;P(tBu) 3:三叔丁基膦;AcOH:醋酸;Bredereck’s regent:1-叔丁氧基-N,N,N',N'-四甲基甲二胺;dioxane:1,4-二氧六环;t-BuOK:叔丁醇钾;LDA:二异丙基氨基锂;HATU:2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯;DIPEA:N,N-二异丙基乙胺;DMF-DMA:N,N-二甲基甲酰胺二甲基缩醛;T 3P:丙基磷酸酐;TEA:三乙胺;DMSO:二甲亚砜;DAST:二乙胺基三氟化硫;PPh 3:三苯基膦;Ziram:二甲基二硫代氨基甲酸锌;DEAD:偶氮二甲酸二乙酯;(Rh(OAc) 2) 2:二聚醋酸铑;D-M's reagent:戴斯马丁氧化剂;XtalFluor-E:(二乙氨基)二氟锍鎓四氟硼酸盐;Pd(OAc) 2:醋酸钯;P(Cy) 3:三环己基膦;DIBAL-H:二异丁基氢化铝;DME:乙二醇二甲醚;CAN:硝酸铈铵;IPA:异丙醇;DEA:二乙醇胺;DMF:N,N-二甲基甲酰胺;DBU:1,8-二氮杂二环[5.4.0]十一碳-7-烯;MsCl:甲烷磺酰氯;NBS:N-溴代琥珀酰亚胺;DCE:二氯乙烷;DIEA:N,N-二异丙基乙胺;pyrrolidine:四氢吡咯;pyridine:吡啶;DHP:3,4-二氢吡喃;PPTS:吡啶对甲苯磺酸盐
实施例1:3-[3-[3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基]苯基]-7-[[(3S)-3-甲基-1-哌啶基]甲基]吡啶并[1,2-a]嘧啶-4-酮(化合物1)的制备
Figure PCTCN2022128426-appb-000178
合成路线:
Figure PCTCN2022128426-appb-000179
步骤1:4-甲基-3-(3-甲基-1-(3-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯基)环丁基)-4H-1,2,4-三氮唑(1B)的制备
将3-(1-(3-溴苯基)-3-甲基环丁基)-4-甲基-4H-1,2,4-三氮唑(400mg,1.31mmol)、B 2Pin 2(498mg,1.96mmol)溶于1,4-二氧六环(40mL)中,然后加入醋酸钾(385mg,3.92mmol)和Pd(dppf)Cl 2(96mg,0.13mmol),抽换氮气3次。混合物在氮气保护下加热至110℃,并搅拌12小时。反应液冷却至室温,过滤,滤饼用乙酸乙酯洗涤,把滤液蒸干得到标题化合物1B粗品,该粗品直接用于下一步反应。
MS m/z(ESI):354.1[M+H].
步骤2:7-溴-3-碘-4H-吡啶并[1,2-a]嘧啶-4-酮(1D)的制备
将7-溴-4H-吡啶并[1,2-a]嘧啶-4-酮(250mg,1.11mmol)溶于二氯甲烷(20mL)中,加入N-碘代丁二酰亚胺(375mg,1.67mmol)。所得溶液室温下搅拌过夜,然后加入饱和亚硫酸钠水溶液除掉过量的N-碘代丁二酰亚胺,混合物用乙酸乙酯萃取,有机相用无水硫酸镁干燥,过滤,蒸干溶液后得到标题化合物1D的粗品,该粗品直接用于下一步反应。
MS m/z(ESI):351.1/353.1[M+H].
步骤3:3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基)苯基)-7-乙烯基-4H-吡啶并[1,2-a]嘧啶-4-酮(1F)的制备
往1B(200mg,0.57mmol)、1D(302mg,0.86mmol)、碳酸钾(472mg,3.42mmol)以及Pd(PPh 3) 4(132mg,0.11mmol)的混合物中加入1,4-二氧六环(15mL)和水(5mL),抽换氮气3次,然后在氮气保护下加热至80℃,搅拌3小时。反应液冷却至室温,加入乙烯三氟硼酸钾(131mg,0.86mmol),混合物加热至100℃,并搅拌3小时。反应液冷却至室温,旋蒸除去溶剂,残留物通过反相柱色谱法进行纯化(色谱柱:
Figure PCTCN2022128426-appb-000180
快速硅胶柱;流动相:乙腈/水=60:40),得到标题化合物1F(180mg)。
MS m/z(ESI):398.1[M+H]。
步骤4:3-[3-[3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基]苯基]-4-氧代-吡啶并[1,2-a]嘧啶-7-甲醛(1G)的制备
将1F(180mg,0.45mmol)溶于四氢呋喃(5mL),然后加入二水合锇酸钾和0.5mol/L高碘酸钠水溶液(5mL),混合物在室温下搅拌1小时。反应完毕后滤去固体,滤饼用乙酸乙酯洗涤,滤液分去水层,有机相用饱和亚硫酸钠水溶液洗涤,无水硫酸镁干燥,过滤,蒸干,得标题化合物1G粗品(160mg),粗品直接用于下一步反应。
MS m/z(ESI):400.2[M+H].
步骤5:3-[3-[3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基]苯基]-7-[[(3S)-3-甲基-1-哌啶基]甲基]吡啶并[1,2-a]嘧啶-4-酮(化合物1)的制备
向1G(90mg,0.23mmol)的二氯甲烷(10mL)溶液中按顺序加入(S)-3-甲基哌啶盐酸盐(46mg,0.34mmol)、醋酸钾(44mg,0.45mmol)以及醋酸硼氢化钠(57mg,0.27mmol)。所得混合物在氮气保护下搅拌过夜。反应液用甲醇淬灭,蒸干溶剂,残留物进行正相柱色谱法(甲醇:二氯甲烷=1:8),然后再进行反相柱色谱法纯化(色谱柱:
Figure PCTCN2022128426-appb-000181
快速硅胶柱;流动相:乙腈/水=60:40),得到标题化合物1(9.1mg)。
MS m/z(ESI):483.4[M+H].
1H NMR(400MHz,DMSO-d 6)δ8.99(s,1H),8.60–8.59(m,1H),8.37–8.29(m,1H),7.94(dd,J=9.1,1.7Hz,1H),7.89(brs,1H),7.74(d,J=9.0Hz,1H),7.68–7.62(m,1H),7.47–7.40(m,1H),7.28(d,J=7.9Hz,1H),3.63–3.53(m,2H),3.28–3.22(m,3H),2.94–2.82(m,2H),2.76–2.67(m,2H),2.60–2.54(m,2H),2.36–2.23(m,1H),1.94(t,J=10.8Hz,1H),1.70–1.56(m,4H),1.51–1.41(m,1H),1.11–1.08(m,3H),0.91–0.77(m,4H).
实施例2:3-[3-[3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基]苯基]-7-[[(3S)-3-甲基-1-哌啶基]甲基]-9-(三氟甲基)吡啶并[1,2-a]嘧啶-4-酮(化合物2)、3-(3-((1S,3R)-3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基)苯基)-7-(((S)-3-甲基哌啶-1-基)甲基)-9-(三氟甲基)-4H-吡啶并[1,2-a]嘧啶-4-酮和3-(3-((1R,3S)-3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基)苯基)-7-(((S)-3-甲基哌啶-1-基)甲基)-9-(三氟甲基)-4H-吡啶并[1,2-a]嘧啶-4-酮的制备
Figure PCTCN2022128426-appb-000182
合成路线:
Figure PCTCN2022128426-appb-000183
步骤1:(S)-三氟(3-甲基哌啶-1-基)甲基硼酸钾(2B)的制备
往(S)-3-甲基哌啶盐酸盐(2A,540mg,3.98mmol)、(溴甲基)三氟硼酸钾(960mg,4.78mmol)、碳酸钾(605mg,4.38mmol)以及碘化钾(67mg,0.40mmol)中加入无水四氢呋喃(10mL),抽换氮气3次。混合物在氮气保护下加热至回流并搅拌12小时。反应完毕后冷却至室温,加入100mL丙酮,过滤,滤饼用丙酮洗涤,滤液蒸干得到标题化合物2B粗品420mg,该粗品直接用于下一步反应。
MS m/z(ESI):162.1[M-KF+H] +
步骤2:7-溴-9-(三氟甲基)-4H-吡啶并[1,2-a]嘧啶-4-酮(2D)的制备
往5-溴-3-(三氟甲基)吡啶-2-胺(2C,8.00g,33.2mmol)以及5-(甲氧基亚甲基)-2,2-二甲基-1,3-二氧六环-4,6-二酮(7.42g,39.8mmol)的混合物中加入1,2-二氯苯(100mL)中,混合物加热 至110℃,搅拌4小时,然后温度升至200℃,继续搅拌3小时。反应完毕后降至室温,经正相柱色谱法(石油醚:乙酸乙酯=3:1)分离纯化,得到标题化合物2D(4.9g)。
MS m/z(ESI):293.1/295.1[M+H] +
步骤3:7-[[(3S)-3-甲基-1-哌啶基]甲基]-9-(三氟甲基)吡啶并[1,2-a]嘧啶-4-酮(2E)的制备
往2D(410mg,1.40mmol)、2B(460mg,2.10mmol)、XphosPdG 2(110mg,0.14mmol)、2-二环己基膦-2',4',6'-三异丙基联苯(110mg,0.14mmol)以及碳酸钾(133mg,4.20mmol)的混合物中加入1,4-二氧六环(20mL)和水(5mL),抽换氮气3次,然后在氮气保护下加热至100℃,搅拌4小时。反应完毕后冷却至室温,减压浓缩,残留物经反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000184
快速硅胶柱;流动相:乙腈/水=60:40)分离纯化,得到标题化合物2E(210mg)。
MS m/z(ESI):326.1[M+H] +
步骤4:3-碘-7-[[(3S)-3-甲基-1-哌啶基]甲基]-9-(三氟甲基)吡啶并[1,2-a]嘧啶-4-酮(2F)的制备
将2E(210mg,0.66mmol)溶于乙腈(10ml),加入碘(197mg,0.77mmol)以及硝酸铈铵(342mg,0.65mmol),混合物在室温下搅拌12小时。反应完毕后过量的碘用饱和亚硫酸钠水溶液淬灭,二氯甲烷萃取,有机相用无水硫酸镁干燥,过滤,蒸干,得标题化合物2F粗品(240mg),粗品直接用于下一步反应。
MS m/z(ESI):452.0[M+H] +
步骤5:3-[3-[3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基]苯基]-7-[[(3S)-3-甲基-1-哌啶基]甲基]-9-(三氟甲基)吡啶并[1,2-a]嘧啶-4-酮(化合物2)、3-(3-((1S,3R)-3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基)苯基)-7-(((S)-3-甲基哌啶-1-基)甲基)-9-(三氟甲基)-4H-吡啶并[1,2-a]嘧啶-4-酮和3-(3-((1R,3S)-3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基)苯基)-7-(((S)-3-甲基哌啶-1-基)甲基)-9-(三氟甲基)-4H-吡啶并[1,2-a]嘧啶-4-酮的制备
向2F(240mg,0.53mmol)、1B(188mg,0.53mmol)以及碳酸钾(221mg,1.60mmol)的二氧六环/水(25mL,4:1)溶液中,加入Pd(dppf)Cl 2(39mg,0.53mmol),所得混合物在氮气保护下加热至80℃,搅拌4小时。反应液冷却至室温,旋蒸除去溶剂,残留物先直接进行正相柱色谱法(甲醇:二氯甲烷=1:10),然后再进行反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000185
快速硅胶柱;流动相:乙腈/水=75:25)分离纯化,得到标题化合物2(130mg)。
MS m/z(ESI):551.0[M+H] +
将化合物2进行SFC(仪器:CAS-SH-ANA-SFC-G(Agilent 1260 with DAD detector);柱子:ChiralPak AS-3柱长150mm,内径4.6mm,粒径3μm;流动相A:CO 2,流动相B:IPA(含0.05%DEA);梯度:流动相B从5%到40%用时4.5分钟,然后用5%流动相B洗脱1.5分钟;流速:2.5mL/min;柱温:40℃;自动背压调节器(ABPR):100bar)分离,得异构体标题化合物2-P1(t R=4.52min,70mg)和标题化合物2-P2(t R=4.69min,15mg)。
2-P1
MS m/z(ESI):551.2[M+H] +
1H NMR(400MHz,CDCl 3)δ9.17(s,1H),8.58(s,1H),8.25(br.s.,1H),7.97(s,1H),7.88(s,1H),7.63(d,J=8.0Hz,1H),7.45(t,J=8.0Hz,1H),7.32(d,J=8.0Hz,1H),3.56(br.s.,2H),3.25(s,3H),2.99-2.84(m,2H),2.84-2.62(m,5H),2.10-1.94(m,1H),1.78-1.53(m,6H),1.19-1.10(m,3H),1.02-0.91(m,1H),0.90-0.82(m,3H)。
2-P2
MS m/z(ESI):551.2[M+H] +
1H NMR(400MHz,CDCl 3)δ9.16(s,1H),8.59-8.55(m,1H),8.25(s,1H),8.03(s,1H),7.71(s,1H),7.62-7.57(m,1H),7.45-7.39(m,1H),7.23-7.16(m,1H),3.56(s,2H),3.31(s,3H),3.26-3.16(m,2H),2.83-2.69(m,2H),2.65-2.54(m,1H),2.41-2.31(m,2H),2.06-1.97(m,1H),1.71-1.54(m,5H),1.39-1.23(m,2H),1.18-1.11(m,3H),0.89-0.85(m,3H)。
实施例3:(S)-8-甲基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基)苯基)-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物58)的制备
Figure PCTCN2022128426-appb-000186
合成路线:
Figure PCTCN2022128426-appb-000187
步骤1:1-(5-溴-2-羟基-3-甲基苯基)乙酮(58A)的制备
将1-(2-羟基-3-甲基苯基)乙酮(2g,13.32mmol)溶于无水二氯甲烷(20mL)中。在0℃冰浴下,将溶于无水二氯甲烷(20mL)的液溴(3.62g,22.64mmol,1.24mL)缓慢滴加到溶液中。保持反应在0℃冰浴下搅拌1小时。反应结束后,待反应液温度上升到室温,过量液溴用饱和偏重亚硫酸钠水溶液淬灭,用二氯甲烷萃取反应液。有机相用无水硫酸钠干燥,过滤并减压蒸馏旋干得到粗产品。粗产品用正相柱色谱法(石油醚:乙酸乙酯=100:1)进行纯化,得到标题化合物58A(3g)。
MS m/z(ESI):228.8/230.8[M+H] +
步骤2:1-(5-溴-2-羟基-3-甲基苯基)-3-(二甲氨基)丙-2-烯-1-酮(58B)的制备
在室温下,将N,N-二甲基甲酰胺二甲基缩醛(4.16g,34.92mmol)加入到盛有58A(3g,13.3mmol)的无水甲苯溶液(60mL)的封管中。然后将反应液升温至115℃并搅拌16小时。反应结束后,将反应液冷却至室温,加入水(60mL),乙酸乙酯萃取(50mL×3)。有机相用无水硫酸钠干燥,过滤,并减压蒸馏得到粗产品;将粗产品用正相柱色谱法(石油醚:乙酸乙酯=73:27)进行纯化,得到标题产物58B(640mg)。
MS m/z(ESI):284.9/285.7[M+H] +
步骤3:6-溴-3碘-8-甲基-4H-色烯-4-酮(58C)的制备
在室温下,将吡啶(353.54mg,4.47mmol,360.05μL)与碘(1.13g,4.47mmol)加入58B(635mg,2.23mmol)的氯仿溶液(10mL)中。反应混合物在室温下反应16小时。反应结束后,过量的碘用饱和亚硫酸钠水溶液淬灭,并用二氯甲烷萃取。有机相用无水硫酸钠进行干燥,过滤,并减压蒸馏得到标题粗产品58C(732mg)直接用于下一步反应。
MS m/z(ESI):364.7/366.7[M+H] +
步骤4:6-溴-8-甲基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-4H-色烯-4-酮(58D)的制备
将58C(300mg,822.01μmol)、1B(232.31mg,657.60μmol)和磷酸钾(261.73mg,1.23mmol)加入到1,4-二氧六环(10mL)与水(1mL)的混合液中,并置换氮气;在氮气氛围下,将Pd(dppf)Cl 2(6.01mg,8.22μmol)加入反应液中。投料结束后,将反应升温至100℃并在氮气氛围下搅拌16 小时。反应结束后,将反应液冷却至室温,加入水(15mL)并用乙酸乙酯萃取。萃取所得有机相用无水硫酸钠干燥,过滤,减压蒸馏得到粗产品,并经过正相柱色谱法(石油醚:乙酸乙酯=0:100)纯化,得到标题产物58D(240mg)。
MS m/z(ESI):464.0/466.0[M+H] +
步骤5:(S)-8-甲基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物58)的制备
将58D(120mg,258.42μmol)、2B(79.27mg,361.79μmol)和碳酸钾(107.15mg,775.27μmol)加入水(1mL)与1,4-二氧六环(10mL)的混合液中,并置换氮气。在氮气氛围下,将XphosPdG 2(20.33mg,25.84μmol)与Xphos(24.64mg,51.68μmol)加入溶液中,并将反应升温至90℃搅拌16小时。反应结束后,将反应冷却至室温,加入水(10mL)和乙酸乙酯萃取(10mL×3)。有机相用无水硫酸钠干燥,过滤,减压蒸馏得到粗产品,粗产品用高效液相色谱法(色谱柱:Welch Xtimate C18;柱长150mm,内径30mm,粒径5μm;流动相A:水(含0.225%甲酸);流动相B:乙腈;梯度:流动相B从24%到54%,用时7分钟,然后用100%流动相B洗脱3分钟;流速:25mL/min)进行纯化。得到标题化合物58(43.25mg,34%)。
MS m/z(ESI):497.2[M+H] +
1H NMR(400MHz,CDCl 3)8.61-8.05(m,1H),8.04-7.94(m,2H),7.75(br.s.,1H),7.62(s,1H),7.46-7.35(m,3H),3.81-3.66(m,2H),3.34-3.24(m,3H),3.22-3.13(m,1H),3.05-2.97(m,1H),2.96-2.85(m,3H),2.73-2.64(m,2H),2.52(s,3H),2.39-2.27(m,1H),2.14-2.00(m,1H),1.91-1.62(m,5H),1.19-1.10(m,3H),0.89-0.82(m,3H).
实施例4:(S)-6-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-2-((3-甲基哌啶-1-基)甲基)-5H-噻唑并[3,2-a]嘧啶-5-酮(化合物59)的制备
Figure PCTCN2022128426-appb-000188
步骤1:(4-甲酰基噻唑-2-基)氨基甲酸叔丁酯(59A)的制备
将2-氨基噻唑-4-甲醛(400mg,3.12mmol)、三乙胺(948mg,9.36mmol)以及(Boc) 2O(817mg,3.75mmol)溶于无水二氯甲烷(20mL)中,然后加入DMAP(76mg,0.62mmol),反应液在室温下搅拌2小时。反应结束后,减压蒸馏旋干得到粗产品。粗产品用正相柱色谱法(石油醚:乙酸乙酯=1:1)进行纯化,得到标题化合物59A(620mg)。
MS m/z(ESI):229.1[M+H] +.
步骤2:(S)-5-((3-甲基哌啶-1-基)甲基)噻唑-2-胺(59B)的制备
将59A(240mg,1.05mmol)、三乙胺(319mg,3.15mmol)以及(S)-3-甲基哌啶盐酸盐(214mg,1.58mmol)溶于无水二氯甲烷(20mL)中,然后加入乙酸硼氢化钠(334mg,1.58mmol),反应液在室温下搅拌过夜。反应结束后,用饱和氯化铵水溶液淬灭,二氯甲烷萃取,有机相减压蒸馏旋干得到粗产品。粗产品加入三氟乙酸(5mL)并搅拌1小时,减压浓缩除去三氟乙酸,残留物用饱和碳酸氢钠溶液中和,二氯甲烷萃取,有机相减压旋干得到粗品,粗品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000189
快速硅胶柱;流动相:乙腈/水=75:25)进行纯化,得到标题化合物59B(150mg)。
MS m/z(ESI):212.1[M+H] +.
步骤3:2-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)丙二酸二乙酯(59C)的制备
向1A(1g,3.27mmol)、丙二酸二乙酯(10.5mg,65.3mmol)、碳酸钾(1.4g,9.8mmol)和碳酸氢钠(0.82g,9.8mmol)的混合物中加入双二亚苄基丙酮钯(188mg,0.33mmol)和三叔丁基膦(132mg,0.65mmol)并抽换氮气3次,氮气保护下160℃封管反应18小时。反应结束后降至室温,过滤,滤饼用乙酸乙酯洗两次,滤液减压蒸干。残留物用正相柱色谱法(二氯甲烷:甲醇=15:1)纯化,得到标题化合物59C(1.2g)。
MS m/z(ESI):386.1[M+H] +
步骤4:2-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)乙酸乙酯(59D)的制备
向59C(1.2g,3.11mmol)中加入10mL 6N浓盐酸和10mL冰醋酸,所得溶液加热至110℃并搅拌2小时。反应结束后降至室温,减压蒸干。残留物溶于乙醇中(20mL),然后加入0.2mL浓硫酸,反应液回流2小时,然后降至室温,减压蒸干,残留物用饱和碳酸氢钠溶液中和,二氯甲烷萃取,有机相减压蒸干,残留物用正相柱色谱法(二氯甲烷:甲醇=15:1)纯化,得到标题化合物59D(680mg)。
MS m/z(ESI):314.2[M+H] +
步骤5:3-(二甲氨基)-2-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)丙烯酸乙酯(59E)的制备
把59D(640mg,2.04mmol)和1-叔丁氧基-N,N,N',N'-四甲基甲二胺(1.1g,6.13mmol)溶于30mL无水四氢呋喃,所得溶液氮气保护下回流12小时。反应结束后降至室温,减压蒸去低沸点物质,真空干燥后得标题化合物59E(780mg),粗品直接进行下一步反应。
MS m/z(ESI):369.1[M+H] +
步骤6:(S)-6-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-2-((3-甲基哌啶-1-基)甲基)-5H-噻唑并[3,2-a]嘧啶-5-酮(化合物59)的制备
把59E(104mg,0.28mmol)和59B(50mg,0.24mmol)溶于5mL冰醋酸,反应液回流12小时。反应结束后降至室温,减压浓缩,粗品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000190
快速硅胶柱;流动相:乙腈/水=75:25)纯化,得到标题化合物59(6mg)。
MS m/z(ESI):489.5[M+H] +
1H NMR(400MHz,CDCl 3)8.31-8.21(m,2H),8.14(s,1H),7.73(s,1H),7.61-7.49(m,1H),7.47-7.36(m,1H),7.32-7.24(m,1H),4.24-3.98(m,1H),3.68(s,3H),3.24-3.16(m,4H),2.93-2.73(m,4H),2.05-1.94(m,1H),1.81-1.53(m,5H),1.52-1.38(m,1H),1.08(s,3H),0.86-0.81(m,3H)
实施例5:9-甲基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基)苯基)-7-(哌啶-3- 基)-4H-吡啶并[1,2-a]嘧啶-4-酮(化合物60)的制备
Figure PCTCN2022128426-appb-000191
步骤1:5-(9-甲基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基)苯基)-4-氧代-4H-吡啶并[1,2-a]嘧啶-7-基)-3,6-1,2,3,6-四氢吡啶-1-羧酸叔丁酯(60A)的制备
7-溴-9-甲基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基)苯基)-4H-吡啶并[1,2-a]嘧啶-4-酮的合成过程与实施例3步骤4类似,不同的是把58C(300mg,822.01μmol)替换成7-溴-3-碘-4H-吡啶并[1,2-a]嘧啶-4-酮(288mg,822.01μmol),同法制备。
将5-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-3,6-二氢吡啶-1(2H)-羧酸叔丁酯(43.95mg,142.13μmol)、7-溴-9-甲基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基)苯基)-4H-吡啶并[1,2-a]嘧啶-4-酮(60mg,129.21μmol)和碳酸钾(53.57mg,387.63μmol)加入二氧六环(3mL)和水(0.6mL)的混合液中,置换氮气。然后把Pd(dppf)Cl 2(9.48mg,12.92μmol)加入反应液中,置换氮气。将反应升温至100℃并在氮气保护下搅拌16小时。反应结束后,反应液冷却至室温,加入水(15mL)并用乙酸乙酯萃取。有机相用无水硫酸钠干燥,过滤,减压蒸馏得到粗产品,粗产品经过正相柱色谱法(石油醚:乙酸乙酯=1:1)纯化,得到标题产物60A(90mg)。
MS m/z(ESI):567.2[M+H] +
步骤2:3-(9-甲基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基)苯基)-4-氧代-4H-吡啶并[1,2-a]嘧啶-7-基]哌啶-1-羧酸叔丁酯(60B)的制备
向60A(90mg,158.82μmol)中加入乙酸乙酯(5mL)和湿钯碳(19.29mg,15.88μmol,10%纯度),置换三次氢气,25℃搅拌12小时。反应结束后,过滤,浓缩,得到粗品标题产物60B(40mg),该粗品直接用于下一步反应。
MS m/z(ESI):569.2[M+H] +
步骤3:9-甲基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基)苯基)-7-(哌啶-3-基)-4H-吡啶并[1,2-a]嘧啶-4-酮(化合物60)的制备
往60B(35mg,61.54μmol)中加入盐酸/二氧六环(1mL,4mol/L),室温下搅拌3小时。反应结束后,旋蒸除去溶剂得到粗产品,粗产品用制备高效液相色谱柱(柱子:Phenomenex Gemini C18,柱长75mm,内径40mm,粒径3μm;流动相A:水(0.225%FA),流动相B:乙腈;流速:25mL/min,梯度:流动相B从13%到43%用时7分钟,然后用100%流动相B洗脱2分钟;流速:25mL/min)分离,得到标题化合物60(4.2mg)。
MS m/z(ESI):469.1[M+H] +
1H NMR(400MHz,DMSO-d 6)8.99-8.85(m,1H),8.51(d,J=4.8Hz,1H),8.20-7.98(m,1H),7.88-7.66(m,1H),7.60-7.52(m,2H),7.48-7.40(m,1H),7.39-7.35(m,0.5H),7.25-7.20(m,0.5H),3.73-3.38(m,2H),3.35-3.26(m,3H),3.24-3.12(m,2H),2.96-2.90(m,2H),2.72-2.67(m,3H),2.57(s,3H),2.43-1.93(m,4H),1.86-1.71(m,1H),1.19-1.09(m,3H).
实施例6:3-(3-(1-环丁基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)乙基)苯基)-7-(((S)-3-甲基哌啶-1-基)甲基)-9-(三氟甲基)-4H-吡啶并[1,2-a]嘧啶-4-酮(化合物61)的制备
Figure PCTCN2022128426-appb-000192
步骤1:2-(3-溴苯基)-2-环丁基乙酸甲酯(61A)的制备
在0℃(冰水浴)条件下,向2-(3-溴苯基)乙酸甲酯(5g,21.83mmol)的无水DMF(50mL)溶液中加入叔丁醇钾(3.18g,28.38mmol),混合物在0℃(冰水浴)搅拌0.5小时,在0℃(冰水浴)条件下向反应液中加入环丁基溴(3.54g,26.19mmol)。反应液恢复至室温,在室温下搅拌12小时。反应完毕后用饱和氯化铵水溶液淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,过滤,减压浓缩,得到无色油状粗品,粗品经正相柱色谱法(石油醚:乙酸乙酯=10:1)纯化,得到标题化合物61A(4.5g)。
MS m/z(ESI):283.16/284.9[M+H] +.
步骤2:2-(3-溴苯基)-2-环丁基丙酸甲酯(61B)的制备
将2-(3-溴苯基)-2-环丁基乙酸甲酯(4.5g,15.89mmol)溶于无水四氢呋喃(10mL),抽换氮气3次,混合物在-78℃(干冰/乙醇)条件下冷却,向混合物中滴加二异丙基氨基锂(13mL,26mmol,2M),混合物在-78℃(干冰/乙醇)氮气保护下,搅拌1小时,再向混合物中滴加碘甲烷(22.80g,160.63mmol,10mL)。反应液恢复至室温,在氮气保护下,搅拌3小时。反应完毕后用饱和氯化铵水溶液淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,过滤,减压浓缩得粗品,粗品经正相柱色谱法(石油醚:乙酸乙酯=10:1)分离纯化,得到标题化合物61B(4.1g)。
MS m/z(ESI):297.19/296.9[M+H] +
步骤3:2-(3-溴苯基)-2-环丁基丙酸(61C)的制备
往61B(2g,6.73mmol)的水(3mL)和四氢呋喃(20mL)的混合溶液中加入氢氧化钠(1.35g,33.65mmol),所得混合物加热至80℃,搅拌12小时。反应液冷却至室温,用2M的盐酸水溶液调节pH至5,乙酸乙酯萃取,有机相用无水硫酸钠干燥,过滤,减压浓缩,得标题化合物61C粗品(1.8g),粗品直接用于下一步反应。
MS m/z(ESI):283.16/283.8[M+H] +
步骤4:2-(2-(3-溴苯基)-2-环丁基丙酰基)-N-甲基氨基硫脲(61D)的制备
向61C(1.8g,6.06mmol)、4-甲基-3-硫代氨基脲(1.8g,6.06mmol)、DIPEA(764.33mg,7.27mmol)和无水DMF(15mL)的混合物中加入HATU(1.35g,33.65mmol),室温搅拌12小时。反应完毕后反应液用水淬灭,乙酸乙酯萃取,有机相用无水硫酸钠干燥,过滤,减压浓缩得粗品,粗品经正相柱色谱法(石油醚:乙酸乙酯=1:1)分离纯化,得到标题化合物61D(2g)。
MS m/z(ESI):370.31/371.9[M+H] +
步骤5:5-(1-(3-溴苯基)-1-环丁基乙基)-4-甲基-4H-1,2,4-三氮唑-3-硫醇(61E)的制备
将61D(1.5g,4.05mmol)溶于氢氧化钠水溶液(1.5M,30mL)中,加热至50℃,搅拌12小 时。反应完毕后反应液冷却至室温,用1M的盐酸水溶液调节pH至3,乙酸乙酯萃取,有机相用无水硫酸钠干燥,过滤,减压浓缩,得到标题化合物61E粗品(1.3g),粗品直接用于下一步反应。
MS m/z(ESI):352.29/351.9[M+H] +
步骤6:3-(1-(3-溴苯基)-1-环丁基乙基)-4-甲基-4H-1,2,4-三氮唑(61F)的制备
在0℃(冰水浴)条件下,向61E(1.3g,3.69mmol)、乙酸(6mL,3.69mmol)和二氯甲烷(20mL)的混合物中缓慢滴加过氧化氢溶液(2.060g,18.17mmol,纯度:30%)。反应液恢复至室温,在室温下搅拌12小时。反应完毕后用饱和硫代硫酸钠水溶液淬灭,二氯甲烷萃取,有机相用无水硫酸钠干燥,过滤,减压浓缩得粗品,粗品经正相柱色谱法(石油醚:乙酸乙酯=0:1)分离纯化,得到标题化合物61F(900mg)。
MS m/z(ESI):320.23/321.9[M+H] +.
步骤7:3-(1-环丁基-1-(3-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)苯基)乙基)-4-甲基-4H-1,2,4-三氮唑(61G)的制备
向61F(900mg,2.81mmol)、B 2Pin 2(1.07g,4.22mmol)、乙酸钾(551mg,5.61mmol)和无水二氧六环(30mL)的混合液中,加入Pd(dppf)Cl 2·DCM(39mg,0.53mmol),所得混合物在氮气保护下加热至110℃,搅拌12小时。反应完毕后冷却至室温,过滤,减压浓缩,残留物经正相柱色谱法(石油醚:乙酸乙酯=0:1)纯化,得到标题化合物61G(540mg)。
MS m/z(ESI):367.29/368.5[M+H] +.
步骤8:3-(3-(1-环丁基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)乙基)苯基)-7-(((S)-3-甲基哌啶-1-基)甲基)-9-(三氟甲基)-4H-吡啶并[1,2-a]嘧啶-4-酮(化合物61)的制备
向61G(200mg,544.53μmol)、2F(246.00mg,545.18μmol)、碳酸钠(170.00mg,1.60mmol)、水(0.4mL)和无水二氧六环(2mL)的混合液中,加入Pd(dppf)Cl 2(40mg,54.67μmol),所得混合物在氮气保护下加热至100℃,搅拌12小时。反应完毕后冷却至室温,过滤,减压浓缩得到粗产品,粗产品经制备高效液相色谱柱(柱子:Welch Xtimate C18柱长150mm,内径30mm,粒径5μm;流动相A:水(0.225%FA),流动相B:乙腈;梯度:流动相B从28%到58%用时7分钟,然后用100%流动相B洗脱3.2分钟;流速:25mL/min)分离,得标题化合物61(116mg)。
MS m/z(ESI):564.64/565.1[M+H] +
1H NMR(400MHz,CDCl 3)9.19(s,1H),8.55(s,1H),8.38(s,1H),8.06(s,1H),7.62(d,J=7.6Hz,1H),7.52(s,1H),7.47-7.41(m,1H),7.10(d,J=8.0Hz,1H),3.80-3.68(m,2H),3.51-3.40(m,1H),3.13(s,3H),3.06-2.81(m,4H),2.27-2.11(m,1H),2.08-1.78(m,8H),1.76-1.58(m,4H),1.04-0.93(m,1H),0.89(d,J=6.4Hz,3H).
实施例7:3-(3-(环丙基(4-甲基-4H-1,2,4-三唑-3-基)甲基)苯基)-7-(((S)-3-甲基哌啶-1-基)甲基)-9-三氟甲基-4H-吡啶并[1,2-a]嘧啶-4-酮(化合物63)的制备
Figure PCTCN2022128426-appb-000193
Figure PCTCN2022128426-appb-000194
步骤1:2-(3-溴苯基)-2-重氮乙酸甲酯(63A)的制备
将2-(3-溴苯基)乙酸甲酯(5g,21.83mmol)和1,8-二氮杂二环[5.4.0]十一碳-7-烯(9.97g,65.48mmol,9.77mL)加入乙腈(49.98mL)中,降温到0℃,在氮气保护下滴加4-乙酰氨基苯磺酰叠氮(6.29g,26.19mmol)。将反应液升温至25℃并在氮气保护下搅拌3小时。反应结束后,加入饱和氯化铵水溶液(20mL)并用乙酸乙酯萃取3次。萃取所得有机相用无水硫酸钠干燥,过滤,减压蒸馏得到粗产品,粗产品经过正相柱色谱法(石油醚:乙酸乙酯=10:1)纯化,得到固体标题化合物63A(5.5g,收率:94%)。
MS m/z:254.8,256.8[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ7.77(s,1H),7.45(d,J=7.0Hz,1H),7.42-7.34(m,2H),3.81(s,3H).
步骤2:2-(3-溴苯基)-2-环丙基乙酸甲酯(63B)的制备
往二氯甲烷(20mL)中加入环丙基硼酸(1.35g,15.68mmol)、63A(2g,7.84mmol)、碳酸钠(831.5mg,7.84mmol),置换三次氮气,在光反应仪(450nm)中25℃下搅拌12小时。反应结束后,减压蒸馏得到粗产品,粗产品经过正相柱色谱法(石油醚:乙酸乙酯=10:1)纯化,得到固体标题化合物63B(1.2g,收率:28%)。
MS m/z(ESI):269.01[M+H] +&271.01[M+H] +.
步骤3:2-(3-溴苯基)-2-环丙基乙酸(63C)的制备
将63B(480mg,1.78mmol)加到水(5mL)和四氢呋喃(5mL)的混合液中,再加入氢氧化锂(299.34mg,7.13mmol),室温下搅拌3小时。反应结束后,旋蒸除掉四氢呋喃,用乙酸乙酯萃取3次,水相用1M的盐酸调pH到3~4,再用乙酸乙酯萃取,有机相减压浓缩至干得到固体标题化合物63C(215mg)。
MS m/z(ESI):255.01[M+H] +&257.01[M+H] +.
步骤4:1-[(2-(3-溴苯基)-2-环丙基乙酰基)氨基]-3-甲基硫脲(63D)的制备
向DMF(2mL)中加入DIPEA(519.28mg,4.02mmol,699.83μL)、63C(205mg,803.58μmol)、4-甲基氨基硫脲(211.27mg,2.01mmol)和HATU(460.09mg,1.21mmol)。在室温下搅拌6个小时。反应结束后,用水稀释,再用乙酸乙酯萃取3次。有机相用无水硫酸钠干燥,过滤,滤液减压浓缩至干,得到粗产品。粗产品经过正相柱色谱法(石油醚:乙酸乙酯=0:1)纯化,得到固体标题产物63D(0.11g,收率:8%)。
MS m/z(ESI):342.02[M+H] +&344.02[M+H] +.
步骤5:5-[(3-溴苯基)环丙基甲基]-4-甲基-4H-1,2,4-三氮唑-3-硫醇(63E)的制备
将63D(110mg,321.40μmol)加入到氢氧化钠水溶液(1M,3mL)中。反应液在室温下搅拌12个小时。反应结束后,用1M的盐酸调pH到3~4。用乙酸乙酯萃取,有机相用饱和食盐水洗,无水硫酸钠干燥,过滤,滤液减压浓缩至干,得到固体标题产物63E(77mg)。
步骤6:3-[(3-溴苯基)环丙基甲基]-4-甲基-4H-1,2,4-三氮唑(63F)的制备
将63E(77mg,237.48μmol)加入到二氯甲烷(2mL)和醋酸(285.22mg,4.75mmol)中,冷却至0℃。再逐滴加入双氧水(40.39mg,391.84μmol,纯度33%),恢复至室温下搅拌12个小时。反应结束后,加入饱和亚硫酸氢钠和饱和碳酸氢钠水溶液,调pH到8左右。用二氯甲烷萃取,有机相用饱和亚硫酸氢钠水溶液洗,无水硫酸钠干燥,过滤,滤液减压浓缩至干,得到固体标题产物63F(62mg,收率:90%)。
MS m/z(ESI):292.04[M+H] +&294.04[M+H] +.
步骤7:3-[环丙基(3-(4,4,5,5-四甲基-1,3,2-二噁硼戊环-2-基)苯基)甲基]-4-甲基-4H-1,2,4三氮唑(63G)的制备
将63F(57mg,195.09μmol)、B 2Pin 2(148.62mg,585.27μmol)和醋酸钾(45.04mg,585.27μmol)加到二氧六环(2mL)中,置换氮气,加入Pd(dppf)Cl 2·DCM(15.93mg,19.51μmol)。反应在90℃下搅拌两个小时。反应结束后,反应液用水稀释,然后用乙酸乙酯萃取3次,合并有机相用无水硫酸钠干燥,过滤,滤液减压浓缩至干,得到粗产品,粗产品经过正相柱色谱法(石油醚:乙酸乙酯=10:1)纯化,得到固体标题产物63G(66mg,收率:100%)。
步骤8:3-(3-(环丙基(4-甲基-4H-1,2,4-三唑-3-基)甲基)苯基)-7-(((S)-3-甲基哌啶-1-基)甲基)-9-三氟甲基-4H-吡啶并[1,2-a]嘧啶-4-酮(化合物63)的制备
将63G(60mg,176.87μmol)、2F(66.51mg,147.39μmol)和碳酸钠(46.86mg,442.17μmol)加到1,4-二氧六环(1mL)和水(0.2mL)中,置换三次氮气,加入Pd(dppf)Cl 2(10.78mg,14.74μmol)。反应在100℃下搅拌三个小时。待反应结束后,反应液冷却至室温,过滤,滤液旋蒸除去溶剂得到粗产品,粗产品用制备高效液相色谱柱(柱子:Phenomenex C18,柱长80mm,内径40mm,粒径3μm;流动相A:水(0.1%氨水),流动相B:乙腈;梯度:流动相B从51%到81%用时8分钟,然后用100%流动相B洗脱4分钟;流速:30mL/min)分离,得标题化合物63(3.49mg,收率:6%)。
MS m/z(ESI):537.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ9.12(s,1H),8.62(s,1H),8.37-8.28(m,2H),7.79-7.74(m,1H),7.68(m,1H),7.42(m,1H),7.29(m,1H),3.67-3.54(m,3H),3.41-3.39(m,3H),2.79(s,2H),1.97(s,1H),1.72-1.40(m,6H),0.90-0.77(m,4H),0.67-0.50(m,2H),0.41-0.30(m,2H).
实施例8:3-(3-(环丙基(4-甲基-4H-1,2,4-三唑-3-基)甲基)苯基)-8-甲基-6-(((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物64)的制备
Figure PCTCN2022128426-appb-000195
Figure PCTCN2022128426-appb-000196
步骤1:(S)-2-羟基-3-甲基-5-(3-甲基哌啶-1-基)甲基苯乙酮(64A)的制备
将5-溴-2-羟基-3-甲基苯乙酮(1g,4.37mmol)、2B(1.43g,6.55mmol)、XphosPd G 2(343.04mg,436.55μmol)、碳酸钾(1.21g,8.73mmol)和Xphos(416.22mg,873.09μmol)称入反应管中,抽真空置换氩气,用注射器加入水(4mL)和二氧六环(12mL),加热至100℃反应1小时,冷却至室温,反应液直接用反相柱纯化(色谱柱:
Figure PCTCN2022128426-appb-000197
快速硅胶柱;流动相:乙腈/水=75:25)后冻干,得标题化合物64A(330mg,收率:29%)。
MS m/z(ESI):262.17[M+H] +
步骤2:(S)-3-二甲氨基-1-(2-羟基-3-甲基-5-((3-甲基哌啶-1-基)甲基)苯基)丙-2-烯-1-酮(64B)的制备
将64A(330mg,1.26mmol)溶解入DMF-DMA(6mL)中,加热至90℃,反应8小时。冷却至室温,浓缩并用氯仿共沸三次,得64B粗品(399mg)直接用于下步。
MS m/z(ESI):317.22[M+H] +
步骤3:(S)-3-碘-8-甲基-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(64C)的制备
将64B(399mg,1.26mmol)溶解入氯仿(4mL)中,冰水浴下依次加入碘(640.06mg,2.52mmol)和吡啶(199.48mg,2.52mmol,203.15μL),于室温反应1小时,饱和亚硫酸氢钠水溶液(1mL)淬灭,过滤,滤液浓缩得粗品,粗品经反相柱(色谱柱:
Figure PCTCN2022128426-appb-000198
快速硅胶柱;流动相:乙腈/水=7:3)纯化并冻干,得标题化合物64C(152mg,收率:30%)。
步骤4:3-(3-(环丙基(4-甲基-4H-1,2,4-三唑-3-基)甲基)苯基)-8-甲基-6-(((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物64)的制备
于一干燥反应管中,加入63G(32.02mg,94.40μmol)、64C(25mg,62.93μmol)、Pd(dppf)Cl 2(4.57mg,6.29μmol)和碳酸钾(8.70mg,62.93μmol),抽真空置换氮气,注射器加入水(0.5mL)和1,4-二氧六环(2mL),加热至100℃反应1小时,冷却至室温,用反相柱纯化(色谱柱:
Figure PCTCN2022128426-appb-000199
快速硅胶柱;流动相:乙腈/水=8:2)并冻干,得标题化合物64(10.5mg,收率:35%)。
MS m/z(ESI):483.27[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.57(s,1H),8.35(s,1H),7.87(s,1H),7.61(s,1H),7.52(s,1H),7.47-7.38(m,2H),7.31(d,J=7.4Hz,1H),3.61–3.51(m,3H),3.42(s,3H),2.72(s,2H),2.45(s,3H),2.10-1.75(m,2H),1.64–1.48(m,5H),0.85-0.76(m,4H),0.66–0.50(m,2H),0.40-0.30(m,2H).
实施例9:(S)-8-甲基-3-(5-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)吡啶-3-基)-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物65)的制备
Figure PCTCN2022128426-appb-000200
步骤1:2-(5-溴吡啶-3-基)乙酸甲酯(65A)的制备
将2-(5-溴吡啶-3-基)乙酸(2.4g,11.11mmol)溶于甲醇(5mL)中,慢慢滴加二氯亚砜(524.16mg,4.41mmol),混合物加热至80℃回流,搅拌12小时。反应液降至室温,向反应液中加入乙酸乙酯(20mL)和水(10mL),分液,有机相用饱和碳酸氢钠水溶液(20mL×3)和饱和食盐水洗涤,有机相用无水硫酸钠干燥,过滤,减压浓缩至干,得到标题化合物粗品65A(2.27g,收率:86%),粗品直接用于下一步反应。
MS m/z(ESI):230.06/231.9[M+H] +
1H NMR(400MHz,CDCl 3)δ8.67-8.55(m,1H),8.48-8.38(m,1H),7.86-7.78(m,1H),3.74(s,3H),3.63(s,2H)
步骤2:1-(5-溴吡啶-3-基)-3-甲基环丁烷-1-羧酸甲酯(65B)的制备
在0℃(冰水浴)条件下,将氢化钠(790mg,19.75mmol,纯度:60%)加入到65A(2.27g,9.87mmol)和无水DMF(15mL)中,混合物在0℃(冰水浴)搅拌0.5小时。在0℃(冰水浴)条件下向混合物中加入1,3-二溴-2-甲基丙烷(3,2.56g,11.84mmol),反应液在0℃(冰水浴)搅拌1.5小时。用饱和氯化铵水溶液淬灭反应,乙酸乙酯萃取,有机相用无水硫酸钠干燥,过滤,减压浓缩至干,得到目标粗品化合物,粗品进行正相柱色谱法(石油醚:乙酸乙酯=10:1)纯化,得到标题化合物65B(1.3g,收率:46%)。
MS m/z(ESI):284.15/285.9[M+H] +.
1H NMR(400MHz,CDCl 3)δ8.61-8.56(m,2H),7.88-7.83(m,1H),3.66(s,3H),2.72- 2.61(m,2H),2.54-2.44(m,2H),2.12-1.99(m,1H),1.12(d,J=4.0Hz,3H)
步骤3:1-(5-溴吡啶-3-基)-3-甲基环丁烷-1-碳酰肼(65C)的制备
往65B(1.3g,4.58mmol)和乙醇(15mL)中加入水合肼(12.340g,209.53mmol,纯度:85%),混合物加热至85℃,搅拌12小时。反应液降至室温,混合物直接蒸干,得到标题化合物粗品65C(1.3g),粗品直接用于下一步反应。
MS m/z(ESI):284.15/285.7[M+H] +.
步骤4:2-(1-(5-溴吡啶-3-基)-3-甲基环丁烷-1-羰基)-N-甲基肼-1-硫代酰胺(65D)的制备
往65C(1.3g,4.58mmol)和无水四氢呋喃(15mL)中加入异硫氰酸甲酯(1.00g,13.73mmol),混合物加热至80℃,搅拌3小时。反应液降至室温,加入乙酸乙酯(20mL),混合物在冰水浴中搅拌1小时,过滤,滤饼用乙酸乙酯(20mL×2)洗涤,将滤饼收集干燥,得到标题化合物粗品65D(1.5g),粗品直接用于下一步反应。
MS m/z(ESI):357.27/359.0[M+H] +.
步骤5:5-(1-(3-溴吡啶-3-基)-3-甲基环丁基)-4-甲基-4H-1,2,4-三氮唑-3-硫醇(65E)的制备
将65D(1.5g,4.20mmol)溶于氢氧化钠水溶液(1.0M,25mL)中,置换三次氮气,混合物在室温下搅拌12小时。反应液用1M的盐酸水溶液调节pH至3,乙酸乙酯(30mL×3)萃取,有机相用无水硫酸钠干燥,过滤,减压浓缩至干,得到标题化合物65E粗品(1.3g),粗品直接用于下一步反应。
MS m/z(ESI):339.25/341.0[M+H] +.
步骤6:3-溴-5-(3-甲基-1-(4-甲基-4H-1,2,4-三氮唑-3-基)环丁基)吡啶(65F)的制备
在0℃(冰水浴)条件下,将过氧化氢(9.2g,81.15mmol,纯度:30%)和冰乙酸(177.53mg,2.96mmol,3mL)缓慢滴加到65E(1.3g,3.83mmol)和二氯甲烷(15mL)的混合物中。反应液恢复至室温,在室温下搅拌12小时。用饱和硫代硫酸钠水溶液淬灭,二氯甲烷(20mL×3)萃取,有机相用无水硫酸钠干燥,过滤,旋干,得到目标粗品化合物,粗品用制备高效液相色谱柱(柱子:Welch Xtimate C18柱长150mm,内径30mm,粒径5μm;流动相A:水(0.1%氨水+0.1%碳酸氢铵),流动相B:乙腈;梯度:流动相B从20%到50%用时8分钟,然后用100%流动相B洗脱2.5分钟;流速:30mL/min)分离,得标题化合物65F(521mg,收率:45%)。
MS m/z(ESI):307.2/308.8[M+H] +.
1H NMR(400MHz,CDCl 3)δ8.62-8.56(m,2H),8.01(s,1H),7.79-7.75(m,1H),3.24(s,3H),2.91-2.80(m,2H),2.75-2.66(m,3H),1.18-1.14(m,3H).
步骤7:3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)-5-(4,4,5,5-四甲基-1,3,2-二噁硼戊环-2-基)吡啶(65G)的制备
参照实施例6步骤7的合成方法,不同的是将61F替换为65F(30mg,97.66μmol),同法制得标题化合物65G(20mg,收率:58%)。
MS m/z(ESI):355.0[M+H] +.
步骤8:6-溴-8-甲基-3-(5-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)吡啶-3-基)-4H-色烯-4-酮(65H)的制备
制备方法同化合物58D。从65G(20mg,56.46μmol)出发,最终得到标题化合物65H(15mg,收率:57%)。
MS m/z(ESI):465.1[M+H] +.
步骤9:(S)-8-甲基-3-(5-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)吡啶-3-基)-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物65)的制备
制备方法同化合物58。65H(15mg,32.23μmol)与2B(10.59mg,48.35μmol)在90℃下反应,最终得到标题化合物65(6.8mg,收率:42%)。
MS m/z(ESI):498.28[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.78(d,J=5.7Hz,1H),8.70–8.65(m,1H),8.55(d,J=2.4Hz,1H),8.35(s,1H),8.05(t,J=2.2Hz,1H),7.89(d,J=2.5Hz,1H),7.66(s,1H),3.54(s,2H), 3.28(s,3H),2.96(d,J=3.6Hz,2H),2.78–2.70(m,2H),2.61(d,J=6.6Hz,3H),2.49(s,3H)1.90(t,J=11.3Hz,1H),1.69–1.49(m,5H),1.12(d,J=5.1Hz,3H),0.83(d,J=5.3Hz,4H).
实施例10:(S)-8-甲基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)-5-三氟甲基苯基)-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物66)的制备
Figure PCTCN2022128426-appb-000201
步骤1至步骤3:1-(3-溴-5-(三氟甲基)苯基)-3-甲基环丁羧酸(66C)的制备
步骤1至步骤2制备方法同化合物65B的制备方法,步骤3制备方法同63C的制备方法。从2-(3-溴-5-(三氟甲基)苯基)乙酸(1g,3.53mmol)出发,经三步反应,最终得到标题化合物66C(170mg,收率:14%)。
MS m/z(ESI):335.0/337.0[M-H] -
步骤4至步骤7:4-甲基-3-(3-甲基-1-(3-(4,4,5,5-四甲基-1,3,2-二噁硼戊环-2-基)-5-(三氟甲基)苯基)环丁基)-4H-1,2,4-三唑(66G)的制备
制备方法同化合物63G的制备方法。从66C(270mg,0.80mmol)出发,经四步反应,最终得到标题化合物66G(33.77mg,收率:10%)。
MS m/z(ESI):422.2[M+H] +
步骤8:(S)-8-甲基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)-5-三氟甲基苯基)-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物66)的制备
制备方法同化合物63的制备方法。64C(18mg,45.31μmol)与66G(19.09mg,45.31μmol)反应,最终得到标题化合物66(11mg,收率:43%)。
MS m/z(ESI):565.25[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.80(s,1H),8.34(s,1H),8.0-7.75(m,2H),7.75-7.5(m,2H),7.5-7.36(s,1H),3.54(s,2H),3.28(s,3H),2.96(s,2H),2.8-2.7(m,4H),2.6(s,3H),2.4(m,1H),1.90 (s,1H),1.75-1.5(m,5H),1.26(s,3H),0.83(m,4H).
实施例11:(S)-8-甲基-3-(3-(2-(4-甲基-4H-1,2,4-三唑-3-基)丙烷-2-基)苯基)-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物67)的制备
Figure PCTCN2022128426-appb-000202
步骤1至步骤4:4-甲基-3-(2-(3-(4,4,5,5-四甲基-1,3,2-二噁硼戊环-2-基)苯基)丙烷-2-基)-4H-1,2,4-三唑(67D)的制备
参照实施例10步骤4-步骤7的合成方法,不同的是将66C替换为2-(3-溴苯基)-2-甲基丙酸(250mg,1.03mmol),同法制得标题化合物67D(116mg,收率:35%)。
MS m/z(ESI):328.2[M+H] +
步骤5:(S)-8-甲基-3-(3-(2-(4-甲基-4H-1,2,4-三唑-3-基)丙烷-2-基)苯基)-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(67)的制备
制备方法同化合物63的制备方法。64C(15mg,37.76μmol)与67D(12.36mg,37.76μmol)反应,最终得到标题化合物67(6mg,收率:34%)。
MS m/z(ESI):471.27[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.63(s,1H),8.38(s,1H),7.87(s,1H),7.63(s,1H),7.54–7.36(m,3H),7.17(dt,J=7.6,1.7Hz,1H),3.53(s,2H),3.15(s,3H),2.71(d,J=13.6Hz,2H),2.50(s,3H),1.91(d,J=12.6Hz,1H),1.77(s,6H),1.72–1.42(m,5H),0.83(d,J=5.3Hz,4H).
实施例12:(1S,3R)-3-(4-甲基-4H-1,2,4-三唑-3-基)-3-(3-(8-甲基-6-(((S)-3-甲基哌啶-1-基)甲基)-4-氧代-4H-色烯-3-基)苯基)环丁甲腈(化合物68)的制备
Figure PCTCN2022128426-appb-000203
Figure PCTCN2022128426-appb-000204
步骤1:顺式-1-(3-溴苯基)-3-羟基环丁烷羧酸(68A)的制备
将2-(3-溴苯基)乙酸(20g,93.00mmol)溶于四氢呋喃(200mL)中,在0℃(冰水浴)下缓慢加入异丙基氯化镁(2M,103.93mL),恢复到室温搅拌1小时。再加入环氧氯丙烷(16.01g,172.99mmol,13.53mL),室温搅拌3小时。再将异丙基氯化镁(2M,103.93mL)滴加入上述反应液中,室温搅拌过夜。反应结束后用2M HCl溶液调节pH至1~2,再用二氯甲烷(500mL×3)萃取。合并有机相,有机相用无水硫酸钠干燥,过滤,旋蒸除去溶剂,得到粗产品,粗产品用正相柱色谱法(0-50%乙酸乙酯/石油醚)纯化,得到标题化合物68A(10.1g,收率:40%)。
1H NMR(400MHz,DMSO-d 6)δ12.43(br.s.,1H),7.53-7.49(m,1H),7.48-7.42(m,1H),7.40-7.26(m,2H),5.31-5.10(m,1H),3.92-3.80(m,1H),2.78-2.69(m,2H),2.54-2.51(m,2H)。
步骤2:顺式-2-(1-(3-溴苯基)-3-羟基环丁烷羰基)-N-甲基肼基硫代甲酰胺(68B)的制备
将68A(10.1g,37.25mmol)溶于二氯甲烷(100mL)中,在搅拌下,依次加入DIPEA(14.44g,111.76mmol)、T 3P(17.78g,55.88mmol)和1-氨基-3-甲基硫脲(4.70g,44.71mmol),反应液室温搅拌过夜。用水(100mL)淬灭,析出白色沉淀,过滤,滤饼收集得白色固体标题化合物68B(10.32g,收率:77%)。
MS m/z(ESI):358.3/359.7[M+H] +.
步骤3:顺式-3-(3-溴苯基)-3-(5-巯基-4-甲基-4H-1,2,4-三唑-3-基)环丁醇(68C)的制备
68B(10.32g,28.81mmol)溶于氢氧化钠溶液(1M,173.87mL)中,置换氮气三次,室温搅拌过夜。反应液用2M盐酸调节pH到3,析出白色沉淀,过滤,滤饼收集得白色固体。滤液用乙酸乙酯(100mL×3)萃取,有机相用无水硫酸钠干燥,过滤,旋蒸除去溶剂得白色固体,前后总共得白色固体标题化合物68C(8.66g,收率:88%)。
MS m/z(ESI):340.2/341.8[M+H] +.
步骤4:顺式-3-(3-溴苯基)-3-(4-甲基-4H-1,2,4-三唑-3-基)环丁醇(68D)的制备
68C(8.66g,25.45mmol)溶于二氯甲烷(100mL)中,加入冰醋酸(38.21g,636.32mmol),0℃(冰水浴)下缓慢滴加30%的双氧水(37.71g,332.59mmol)。滴加完毕后反应液恢复到室温,反应2小时。反应液用饱和亚硫酸钠水溶液淬灭,二氯甲烷(100mL×3)萃取,析出白色沉淀,过滤,滤饼溶于甲醇(200mL),在60℃下搅拌半小时,过滤,滤液减压浓缩至干得白色固体标题化合物68D(2.39g,收率:30%)。
MS m/z(ESI):308.2/309.7[M+H] +.
步骤5:甲磺酸-顺式-3-(3-溴苯基)-3-(4-甲基-4H-1,2,4-三唑-3-基)环丁酯(68E)的制备
68D(2.39g,7.76mmol)溶于二氯甲烷(30mL)中,加入三乙胺(9.42g,93.06mmol),混合物冷却至0℃(冰水浴),加入甲烷磺酰氯(11.33g,98.91mmol),室温反应2小时。反应液用水(50mL)淬灭,二氯甲烷萃取(50mL×2),有机相用无水硫酸钠干燥,过滤,旋蒸除去溶剂得粗产品,粗产品用正相柱色谱法(0-11%甲醇/二氯甲烷)分离,得到标题化合物68E(2.18g,收率:73%)。
MS m/z(ESI):386.3/387.8[M+H] +.
步骤6:反式-3-(3-溴苯基)-3-(4-甲基-4H-1,2,4-三唑-3-基)环丁甲腈(68F)和顺式-3-(3-溴苯基)-3-(4-甲基-4H-1,2,4-三唑-3-基)环丁甲腈(68G)的制备
68E(2.11g,5.46mmol)溶于二甲亚砜(50mL),依次加入碳酸钾(1.51g,10.93mmol)和氰化钾(1.74g,26.72mmol),反应液在120℃、氮气保护下搅拌32小时。反应液冷却到室温后倒入乙酸乙酯(80mL)中,水(80mL×2)洗,有机相用无水硫酸钠干燥,过滤,旋蒸除去溶剂得粗产品,经正相柱色谱法(0-20%甲醇/二氯甲烷)分离,得到黄色油状物(616mg),该黄色油状物再经反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000205
快速硅胶柱;洗脱剂:15%到30%(v/v)乙腈/水)纯化,得到精制的产品,通过干冰/乙醇浴(-78℃)冻干得标题化合物68F(218mg,收率:13%)和标题化合物68G(124mg,收率:7%)。
化合物68F:
MS m/z(ESI):317.2/318.8[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.43(s,1H),7.53-7.48(m,1H),7.44-7.40(m,1H),7.37-7.32(m,1H),7.23-7.19(m,1H),3.63-3.36(m,1H),3.33-3.30(m,2H),3.17(s,3H),3.06-2.96(m,2H)。
化合物68G:
MS m/z(ESI):317.2/318.8[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.38(s,1H),7.54-7.47(m,2H),7.37-7.32(m,1H),7.29-7.24(m,1H),3.62-3.52(m,1H),3.29-3.22(m,2H),3.19-3.16(m,3H),3.15-3.07(m,2H)。
步骤7至步骤8:反式-3-(3-(6-溴-8-甲基-4-氧代-4H-色烯-3-基)苯基)-3-(4-甲基-4H-1,2,4-三唑-3-基)环丁甲腈(68J)的制备
参照实施例9步骤7-步骤8的合成方法,不同的是将65F替换为68F(17.6mg,55.52μmol),同法制得标题化合物68J(15mg,收率:57%)。
MS m/z(ESI):475.1[M+H] +
步骤9:反式-3-(4-甲基-4H-1,2,4-三唑-3-基)-3-(3-(8-甲基-6-(((S)-3-甲基哌啶-1-基)甲基)-4-氧代-4H-色烯-3-基)苯基)环丁甲腈(化合物68)的制备
参照实施例9步骤9的合成方法,不同的是将65H替换为68J(15mg,31.65μmol),与2B(10.37mg,47.33μmol)反应,最终得到标题化合物68(5.1mg,收率:32%)。
MS m/z(ESI):508.26[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.67(s,1H),8.45(s,1H),7.89(d,J=2.1Hz,1H),7.69–7.62(m,1H),7.55–7.37(m,3H),7.25–7.17(m,1H),3.53(s,2H),3.46–3.37(m,4H),3.28(s,3H),3.09–3.05(m,1H),2.78–2.70(m,2H),2.51(s,3H),1.89(d,J=11.6Hz,1H),1.67–1.47(m,5H),0.83(d,J=5.3Hz,4H).
实施例13:3-(3-((1R,3S)-3-氟-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-8-甲基-6-(((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物69)的制备
Figure PCTCN2022128426-appb-000206
步骤1:3-(反式-1-(3-溴苯基)-3-氟环丁基)-4-甲基-4H-1,2,4-三唑(69A)的制备
顺式-3-(3-溴苯基)-3-(4-甲基-4H-1,2,4-三唑-3-基)环丁醇(100mg,324.49μmol)溶解入二氯甲烷(5mL),冷却至0℃,氩气环境下,滴加二乙胺基三氟化硫(78.46mg,486.74μmol),搅拌1小时,饱和NaHCO 3水溶液(1mL)淬灭,浓缩,反相柱纯化(色谱柱:
Figure PCTCN2022128426-appb-000207
快速硅胶柱;流动相:乙腈/水=7:3),得标题化合物69A(51mg,收率:51%)。
MS m/z(ESI):310.03/312.03[M+H] +.
步骤2至步骤3:6-溴-3-(3-(反式-3-氟-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-8-甲基-4H-色烯-4-酮(69C)的制备
参照实施例9步骤7-步骤8的合成方法,不同的是将65F替换为69A(50mg,160.26μmol),同法制得标题化合物69C(15mg,收率:20%)。
MS m/z(ESI):468.1[M+H] +.
步骤4:3-(3-(反式-3-氟-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-8-甲基-6-(((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物69)的制备
参照实施例9步骤9的合成方法,不同的是将65H替换为69C(15mg,32.03μmol),与2B(10.53mg,48.04μmol)反应,最终得到标题化合物69(2.2mg,收率:14%)。
MS m/z(ESI):501.26[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.66(s,1H),8.43(s,1H),7.88(d,J=2.1Hz,1H),7.64(d,J=2.1Hz,1H),7.53–7.47(m,3H),7.20(dt,J=7.4,1.8Hz,1H),5.20–5.01(m,1H),3.51(d,J=14.7Hz,5H),2.91–2.81(m,2H),2.74(t,J=10.7Hz,2H),2.50(s,3H),2.05–1.87(m,2H),1.69–1.49(m,6H),0.83(d,J=5.3Hz,4H).
实施例14:8-甲基-3-(3-(2-(4-甲基-4H-1,2,4-三唑-3-基)氧杂环丁烷-2-基)苯基)-6-(((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物70)的制备
Figure PCTCN2022128426-appb-000208
步骤1:3-(3-溴苯基)氧杂环丁烷-3-甲酸乙酯(70A)的制备
将2-(3-溴苯基)-3-羟基-2-(羟甲基)丙酸乙酯(3.0g,10.38mmol)和三苯基膦(5.44g,20.75mmol)溶于无水二氧六环(30.0mL)中。在0℃冰浴和氩气流吹扫下,将二甲基二硫代氨基甲酸锌(2.71g,15.56mmol)和DEAD(3.61g,20.75mmol)依次滴加反应液中。反应液缓慢升温至室温并搅拌16小时。TLC监测原料消失后,结束反应,过滤并减压蒸馏至干得到粗产品标题化合物70A(2.81g,收率:99.9%)。粗产品无需进一步纯化,直接用于下一步反应。
步骤2:3-(3-溴苯基)氧杂环丁烷-3-羧酸(70B)的制备
在室温下,将70A(2.81g,10.38mmol)溶于乙腈(30.0mL)中,加入氢氧化钠的水溶液(30.0mL,3.0M),反应液在65℃下反应1.0小时。反应结束后,将反应液降温至0℃,加入饱和的柠檬酸水溶液并调节pH至酸性,用二氯甲烷萃取三次。有机相用无水硫酸钠进行干燥,过滤,并减压蒸馏得到粗产品,粗品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000209
快速硅胶柱;流动相:乙腈/水=20:80)纯化,得到标题化合物70B(750.0mg,收率:29.6%)。
MS m/z(ESI):255.0/257.0[M-H] -.
步骤3:2-(3-(3-溴苯基)氧杂环丁烷-3-羰基)-N-甲基肼-1-硫代酰胺(70C)的制备
将70B(750.0mg,2.92mmol)、4-甲基氨基硫脲(368.16mg,3.50mmol)、HATU、磷酸钾(1.32g,3.5mmol)和DIPEA(1.3g,8.75mmol,1.52mL)加入到DMF(8.0mL)中,反应在室温下反应1.0小时。反应结束后,加入水(60.0mL)并用二氯甲烷(6.0mL×3)萃取。萃取所得有机相用无水硫酸钠干燥,过滤,减压蒸馏得到标题产物70C(900.0mg,收率:90%),无需进一步纯化,直接用于下一步反应。
MS m/z(ESI):344.0/346.0[M+H] +
步骤4:5-(3-(3-溴苯基)氧杂环丁烷-3-基)-4-甲基-4H-1,2,4-三唑-3-硫醇(70D)的制备
将70C(900.0mg,2.61mmol)加入氢氧化钠的水溶液(10.0mL,3.0M)中,反应在80℃下反应1.0小时。反应结束后,将反应液降温至0℃,加入饱和的柠檬酸水溶液并调节pH至酸性,用二氯甲烷萃取三次。萃取所得有机相用无水硫酸钠干燥,过滤,减压蒸馏得到标题产物 70D(750.0mg,收率:87.9%),无需进一步纯化,直接用于下一步反应。
MS m/z(ESI):326.0/328.0[M+H] +
步骤5:3-(3-(3-溴苯基)氧杂环丁烷-3-基)-4-甲基-4H-1,2,4-三唑(70E)的制备
室温下,将70D(750.0mg,2.31mmol)溶于二氯甲烷(6.0mL)中,向反应液中依次缓慢滴加冰醋酸(0.5mL)和过氧化氢(9.0mL)。反应液在室温下反应1.0小时。反应结束后,加入水(60.0mL)并用二氯甲烷(6.0mL×3)萃取。萃取所得有机相用无水硫酸钠干燥,过滤,减压蒸馏得到标题产物70E(600.0mg,收率:88.7%),无需进一步纯化,直接用于下一步反应。
MS m/z(ESI):294.0/296.0[M+H] +
步骤6:4-甲基-3-(3-(3-(4,4,5,5-四甲基-1,3,2-二噁硼戊环-2-基)苯基)氧杂环丁烷-3-基)-4H-1,2,4-三唑(70F)的制备
向70E(100mg,0.34mmol)、B 2Pin 2(129.53mg,0.5mmol)、乙酸钾(100mg,1.02mmol)和无水二氧六环(3.0mL)的混合液中,加入Pd(dppf)Cl 2·DCM(17.2mg,0.068mmol),所得混合物在氮气保护下加热至100℃,搅拌12小时。反应完毕后冷却至室温,过滤,减压浓缩,残留物经正相柱色谱法(石油醚:乙酸乙酯=0:1)纯化,得到标题化合70F(80.0mg,收率:69%)。
MS m/z(ESI):342.0[M+H] +.
步骤7:6-溴-8-甲基-3-(3-(3-(4-甲基-4H-1,2,4-三唑-3-基)氧杂环丁烷-3-基)苯基)-4H-色烯-4-酮(70G)的制备
将70F(20.0mg,58.61μmol)、58C(21.39mg,58.61μmol)和碳酸钾(16.1mg,117.23μmol)加入水(0.2mL)与1,4-二氧六环(1.0mL)的混合液中,并置换氮气。在氮气氛围下,加入Pd(dppf)Cl 2(8.51mg,11.72μmol),所得混合物在氮气保护下加热至100℃,搅拌1.0小时。反应完毕后冷却至室温,过滤,减压浓缩得到粗产品,粗品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000210
快速硅胶柱;流动相:乙腈/水=60:40)纯化,得到标题化合物70G(20.0mg,收率:75.5%)。
MS m/z(ESI):452.0/454.0[M+H] +.
步骤8:8-甲基-3-(3-(2-(4-甲基-4H-1,2,4-三唑-3-基)氧杂环丁烷-2-基)苯基)-6-(((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物70)的制备
将70G(20mg,44.22μmol)、2B(29.0mg,132.66μmol)和碳酸钾(18.3mg,132.66μmol)加入水(0.2mL)与1,4-二氧六环(1.0mL)的混合液中,并置换氮气。在氮气氛围下,将XphosPd G 2(6.96mg,8.84μmol)与Xphos(8.43mg,17.69μmol)加入溶液中,并将反应液升温至100℃搅拌1.0小时。反应结束后,将反应液冷却至室温,加入水(10mL)和乙酸乙酯(10mL×3)萃取。有机相用无水硫酸钠干燥,过滤,减压蒸馏得到粗产品,粗产品用高效液相色谱法(色谱柱:Welch Xtimate C18;柱长150mm,内径30mm,粒径5μm;流动相A:水(含0.225%氨水);流动相B:乙腈;梯度:流动相B从5%到95%用时18分钟;流速:25mL/min)进行纯化。得到标题化合物70(4.1mg,19%)。
MS m/z(ESI):485.42[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.63(s,1H),8.48(s,1H),7.85(s,1H),7.61(s,1H),7.53(s,3H),7.28(s,1H),5.39(s,2H),5.10(s,2H),3.50(s,2H),3.29(s,3H),2.70(s,2H),1.57(s,4H),1.23(s,3H),0.79(s,3H).
实施例15:8-甲基-3-(3-(2-(4-甲基-4H-1,2,4-三唑-3-基)氧杂环丁烷-2-基)苯基)-6-(((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物71)的制备
Figure PCTCN2022128426-appb-000211
Figure PCTCN2022128426-appb-000212
步骤1:2-(2-溴乙氧基)-2-(3-溴苯基)乙酸甲酯(71A)的制备
将63A(1.0g,3.92mmol)溶于无水二氯甲烷(5.0mL)中。在0℃冰浴下,将溶液滴加到溶有溴乙醇(499mg,3.92mmol)和二聚醋酸铑(173mg,0.392mmol)的二氯甲烷(5.0mL)混合物中。反应液缓慢升温至室温并搅拌1小时。TLC监测反应完全后,过滤,滤液减压蒸馏得到粗产品。粗产品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000213
快速硅胶柱;流动相:乙腈/水=55:45)纯化,得到标题化合物71A(1.10g,收率:79.7%)。
步骤2:2-(3-溴苯基)氧杂环丁烷-2-羧酸甲酯(71B)的制备
将NaH(102.0mg,2.84mmol,60%分散在矿物油中)加入到封管中,置换4次氩气保护,在冰浴下,加入DMF(1.0mL),然后将71A(1.0g,2.84mmol)溶于DMF(9.0mL)并缓慢滴加到封管中,滴加完毕后,将反应液缓慢升温至室温并搅拌2.0小时。反应结束后,将反应液滴加到冰水(100.0mL)中,用乙酸乙酯萃取(10.0mL×3)。有机相用无水硫酸钠干燥,过滤,减压蒸馏得到标题产物71B(695.0mg,收率:90%),无需进一步纯化,直接用于下一步反应。
步骤3:2-(3-溴苯基)氧杂环丁烷-2-羧酸(71C)的制备
在室温下,将71B(695.0mg,2.56mmol)溶于乙腈(10.0mL)中,加入氢氧化钠的水溶液(10.0mL,3.0M),65℃下反应1.0小时。反应结束后,降温至0℃,加入饱和的柠檬酸水溶液并调节pH至酸性,用二氯甲烷萃取三次。有机相用无水硫酸钠进行干燥,过滤,并减压蒸馏得到标题粗产品71C(594.0mg,收率:90%),无需进一步纯化,直接用于下一步反应。
MS m/z(ESI):255.0/257.0[M-H] -.
步骤4:2-(2-(3-溴苯基)氧杂环丁烷-2-羰基)-N-甲基肼-1-硫代酰胺(71D)的制备
将71C(594.0mg,2.31mmol)、4-甲基氨基硫脲(292.0mg,2.77mmol)、HATU(1.05g,2.77mmol)、磷酸钾(1.05g,2.77mmol)和DIPEA(896.0mg,6.93mmol,1.21mL)加入到DMF(6.0mL)中,室温下反应1.0小时。反应结束后,加入水(60.0mL)并用二氯甲烷(6.0mL×3)萃取。萃取所得有机相用无水硫酸钠干燥,过滤,减压蒸馏得到标题产物71D(720.0mg,收率:90%),无需进一步纯化,直接用于下一步反应。
MS m/z(ESI):344.0/346.0[M+H] +
步骤5:5-(2-(3-溴苯基)氧杂环丁烷-2-基)-4-甲基-4H-1,2,4-三唑-3-硫醇(71E)的制备
将71D(720.0mg,2.09mmol)加入氢氧化钠的水溶液(10.0mL,3.0M)中,反应液在80℃下反应1.0小时。反应结束后,将反应液降温至0℃,加入饱和的柠檬酸水溶液并调节pH至酸性, 用二氯甲烷萃取三次。萃取所得有机相用无水硫酸钠干燥,过滤,减压蒸馏得到标题产物71E(614.0mg,收率:90%),无需进一步纯化,直接用于下一步反应。
MS m/z(ESI):326.0/328.0[M+H] +
步骤6:3-(2-(3-溴苯基)氧杂环丁烷-2-基)-4-甲基-4H-1,2,4-三唑(71F)的制备
在室温下,将71E(614.0mg,1.88mmol)溶于二氯甲烷(6.0mL)中,向反应液中依次缓慢滴加冰醋酸(0.5mL)和过氧化氢(3.0mL)。反应液在室温下反应1.0小时。反应结束后,加入水(60.0mL)并用二氯甲烷(6.0mL×3)萃取。萃取所得有机相用无水硫酸钠干燥,过滤,减压蒸馏得到标题产物71F(500.0mg,收率:90%),无需进一步纯化,直接用于下一步反应。
MS m/z(ESI):294.0/296.0[M+H] +
步骤7:4-甲基-3-(2-(3-(4,4,5,5-四甲基-1,3,2-二噁硼戊环-2-基)苯基)氧杂环丁烷-2-基)-4H-1,2,4-三唑(71G)的制备
向71F(100mg,0.34mmol)、B 2Pin 2(129.53mg,0.5mmol)、乙酸钾(100.0mg,1.02mmol)和无水二氧六环(3.0mL)的混合液中,加入Pd(dppf)Cl 2·DCM(17.2mg,0.068mmol),所得混合物在氮气保护下加热至100℃,搅拌12小时。反应完毕后,反应混合物冷却至室温,过滤,减压浓缩,残留物经正相柱色谱法(石油醚:乙酸乙酯=0:1)纯化,得到标题化合71G(80.0mg,收率:68%)。
MS m/z(ESI):342.0[M+H] +.
步骤8:8-甲基-3-(3-(2-(4-甲基-4H-1,2,4-三唑-3-基)氧杂环丁烷-2-基)苯基)-6-(((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物71)的制备
将71G(25.7mg,75.52μmol)、64C(15.0mg,37.76μmol)和碳酸钾(15.63mg,113.28μmol)加入水(0.3mL)与1,4-二氧六环(1.5mL)的混合液中,并置换氮气。在氮气氛围下,加入Pd(dppf)Cl 2(5.48mg,7.55μmol),所得混合物在氮气保护下加热至100℃,搅拌1.0小时。反应完毕后冷却至室温,过滤,减压浓缩得到粗产品,粗产品经制备高效液相色谱柱(柱子:Welch Xtimate C18,柱长150mm,内径30mm,粒径5μm;流动相A:水(0.225%氨水),流动相B:乙腈;梯度:流动相B从5%到95%用时24分钟)分离,得标题化合物71(6.86mg,收率:37%)。
MS m/z(ESI):485.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ8.62(s,1H),8.46(s,1H),7.85(s,1H),7.59(d,J=8.1Hz,2H),7.51(q,J=8.3,7.6Hz,2H),7.29(d,J=6.7Hz,1H),4.68(dt,J=22.3,7.5Hz,2H),3.88(s,1H),3.51(s,2H),3.29(s,3H),2.93–2.86(m,1H),2.70(s,2H),2.46(s,3H),1.92–1.81(m,1H),1.66–1.54(m,4H),1.45(d,J=11.8Hz,1H),0.84(s,1H),0.79(d,J=4.6Hz,3H).
实施例16:8-((二甲基氨基)甲基)-9-甲基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-4H-吡啶并[1,2-a]嘧啶-4-酮(化合物72)的制备
Figure PCTCN2022128426-appb-000214
Figure PCTCN2022128426-appb-000215
步骤1:4-((二甲氨基)甲基)-3-甲基吡啶-2-胺(72A)的制备
将4-溴-3-甲基吡啶-2-胺(1g,5.35mmol)、((二甲氨基)甲基)三氟硼酸钾(1.06g,6.42mmol)、XPhos Pd G2(421mg,535μmol)、碳酸钾(2.21g,16.05mmol)和Xphos(511mg,1.07mmol)加入反应管中,抽真空置换氩气,用注射器加入水(8mL)和二氧六环(30mL),加热至100℃反应12小时,冷却至室温,反应液直接用反相柱纯化(色谱柱:
Figure PCTCN2022128426-appb-000216
快速硅胶柱;流动相:乙腈/水=60:40)后冻干得标题化合物72A(410mg,收率:46%)。
MS m/z(ESI):166.1[M+H] +
步骤2:8-((二甲基氨基)甲基)-9-甲基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-4H-吡啶并[1,2-a]嘧啶-4-酮(化合物72)的制备
将59E(89.2mg,242.08μmol)和72A(40.0mg,242.08μmol)加入冰醋酸(3mL)中,将反应液升温至110℃搅拌2小时。反应结束后,将反应液蒸馏得到粗产品,粗产品用高效液相色谱法(色谱柱:Welch Xtimate C18;柱长150mm,内径30mm,粒径5μm;流动相A:水(含0.225%氨水);流动相B:乙腈;梯度:流动相B从5%到95%用时24分钟,流速:24mL/min)进行纯化,得到标题化合物72(20.0mg,18.6%)。
MS m/z(ESI):443.2[M+H] +.
1H NMR(400MHz,DMSO-d6)δ9.00(s,1H),8.64(s,1H),8.31(d,J=30.4Hz,1H),7.87–7.63(m,2H),7.44(d,J=7.6Hz,2H),7.12(s,1H),3.59(s,1H),3.27(s,3H),3.22(s,2H),3.12(s,1H),2.87(s,1H),2.57(s,3H),2.27(s,6H),1.17(d,J=49.7Hz,1H),1.09(s,3H).
实施例17:(S)-3-(3-(3,3-二氟-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-8-甲基-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物73)的制备
Figure PCTCN2022128426-appb-000217
Figure PCTCN2022128426-appb-000218
步骤1:3-(3-溴苯基)-3-(4-甲基-4H-1,2,4-三唑-3-基)环丁-1-酮(73A)的制备
将(1S,3S)-3-(3-溴苯基)-3-(4-甲基-4H-1,2,4-三唑-3-基)环丁醇(500.0mg,1.62mmol)溶于无水二氯甲烷(20mL)中。在0℃冰浴下,将戴斯马丁氧化剂(1.38g,3.24mmol)分批加入到溶液中。加入完成后,反应液温度缓慢上升到室温并搅拌2.0小时。反应结束后,过滤掉反应残渣,用二氯甲烷润洗,有机相用无水硫酸钠干燥,过滤并减压蒸馏至干得到粗产品。粗品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000219
快速硅胶柱;流动相:乙腈/水=45:55)纯化,得到标题化合物73A(400.0mg,收率:80%)。
MS m/z(ESI):306/308[M+H] +.
步骤2:3-(1-(3-溴苯基)-3,3-二氟环丁基)-4-甲基-4H-1,2,4-三唑(73B)的制备
在冰浴下,将三乙胺氟化氢(663.0mg,3.92mmol)加入二氯甲烷(5.0mL)中,置换4次氩气气体保护,然后在氩气流的吹扫下向溶液中依次加入XtalFluor-E(897.5mg,3.92mmol)和73A(400.0mg,1.31mmol)。然后将反应液缓慢升温至室温并搅拌2.0小时。反应结束后,将反应液冷却至0℃,加入饱和氯化铵水溶液(60mL)淬灭反应,二氯甲烷(50mL×3)萃取。有机相用无水硫酸钠干燥,过滤并减压蒸馏至干得到粗产品。粗品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000220
快速硅胶柱;流动相:乙腈/水=60:40)纯化,得到标题化合物73B(20.0mg,收率:9.3%)。
MS m/z(ESI):328.0/330.0[M+H] +
步骤3:3-(3,3-二氟-1-(3-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)苯基)环丁基)-4-甲基-4H-1,2,4-三唑(73C)的制备
向73B(10.0mg,30.47μmol)、B 2Pin 2(9.92mg,36.57μmol)、乙酸钾(8.96mg,91.42μmol)和无水二氧六环(1.0mL)的混合液中,加入Pd(dppf)Cl 2·DCM(4.42mg,6.09μmol),所得混合物在氮气保护下加热至100℃,搅拌12小时。反应完毕后冷却至室温,过滤,减压浓缩,残留物经正相柱色谱法(石油醚:乙酸乙酯=0:1)纯化,得到标题化合物73C(10.0mg,收率:87%)。
MS m/z(ESI):376.1[M+H] +.
步骤4:(S)-3-(3-(3,3-二氟-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-8-甲基-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物73)的制备
将73C(10.0mg,26.65μmol)、64C(10.59mg,26.65μmol)和碳酸钾(7.36mg,53.3μmol)加入水(0.2mL)与1,4-二氧六环(1.0mL)的混合液中,并置换氮气。在氮气氛围下,加入Pd(dppf)Cl 2(3.87mg,5.33μmol),所得混合物在氮气保护下加热至100℃,搅拌1.0小时。反应完毕后冷却至室温,过滤,减压浓缩得到粗产品,粗产品经制备高效液相色谱柱(柱子:Welch Xtimate C18,柱长150mm,内径30mm,粒径5μm;流动相A:水(含0.225%氨水),流动相B:乙腈;梯度:流动相B从5%到95%用时18分钟)分离,得标题化合物73(2.21mg,收率:16%)。
MS m/z(ESI):519.46[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ8.62(s,1H),8.40(s,1H),7.86(s,1H),7.61(s,1H),7.57(s,1H),7.53–7.43(m,2H),7.30(d,J=7.5Hz,1H),3.72(s,2H),3.50(s,2H),3.45(d,J=13.0Hz,1H),3.31(s,3H),2.71(s,2H),1.87(s,1H),1.57(s,3H),1.23(s,2H),0.79(s,3H).
实施例18:(S)-8-环丙基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物74)的制备
Figure PCTCN2022128426-appb-000221
步骤1:6-溴-8-碘-4H-色烯-4-酮(74A)的制备
将1-(5-溴-2-羟基-3-碘苯基)乙基-1-酮(341mg,1.00mmol)溶解入DMF-DMA(6mL)中,加热至90℃,反应2小时。冷却至室温,浓缩并用氯仿共沸三次,得一棕色胶状物。将此胶状物溶于二氯甲烷(20mL),加入4mL浓盐酸,室温搅拌3小时。混合物用水稀释,分液,水相用二氯甲烷萃取3次,有机相用饱和碳酸氢钠水溶液洗涤,MgSO 4干燥,过滤,减压浓缩至干得粗品标题化合物74A(310mg,收率:89%)。
MS m/z(ESI):351.0/353.0[M+H] +
步骤2:6-溴-8-环丙基-4H-色烯-4-酮(74B)的制备
将74A(300.0mg,854.86μmol)、环丙基硼酸(73.43mg,854.86μmol)和磷酸钾(544.36mg,2.56mmol)加入水(1.6mL)与甲苯(8.0mL)的混合液中,并置换氮气。在氮气氛围下,将醋酸钯(38.38mg,170.97μmol)与三环己基膦(95.89mg,341.94μmol)加入溶液中,并将反应体系升温至90℃并搅拌16小时。粗产品经反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000222
快速硅胶柱;流动相:乙腈/水=55:45)纯化,得到标题化合物74B(141.0mg,收率:62.2%)。
MS m/z(ESI):265.0/267.0[M+H] +
步骤3:6-溴-8-环丙基-3-碘-4H-色烯-4-酮(74C)的制备
在室温下,依次将74B(141.0mg,531.87μmol)、硝酸铈铵(282.0mg,531.87μmol)和碘 (162.0mg,638.24μmol)加入到乙腈(3.0mL)中。然后将反应液升温至80℃并搅拌16小时。反应结束后,将反应液冷却至室温,加入水(60mL),乙酸乙酯(50mL×3)萃取。有机相用无水硫酸钠干燥,过滤,并减压蒸馏得到粗产品;将粗产品用正相柱色谱法(石油醚:乙酸乙酯=90:10)进行纯化,得到标题产物74C(50.0mg,收率:24%)。
MS m/z(ESI):391.0/393.0[M+H] +
步骤4:6-溴-8-环丙基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-4H-色烯-4酮(74D)的制备
将74C(50.0mg,127.88μmol)、1B(45.1mg,127.88μmol)和碳酸钾(35.29mg,255.76μmol)加入水(0.2mL)与1,4-二氧六环(1.0mL)的混合液中,并置换氮气。在氮气氛围下,加入Pd(dppf)Cl 2(18.56mg,25.58μmol),所得混合物在氮气保护下加热至85℃,搅拌1.0小时。粗产品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000223
快速硅胶柱;流动相:乙腈/水=50:50)纯化,得到标题化合物74D(20.0mg,收率:32.0%)。
MS m/z(ESI):490/492[M+H] +.
步骤5:(S)-8-环丙基-3-(3-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物74)的制备
将74D(20mg,40.78μmol)、2B(26.81mg,122.35μmol)和碳酸钾(16.88mg,122.35μmol)加入水(0.2mL)与1,4-二氧六环(1.0mL)的混合液中,并置换氮气。在氮气氛围下,将XphosPd G 2(6.42mg,8.16μmol)与Xphos(7.78mg,16.31μmol)加入溶液中,并将反应液升温至100℃搅拌16小时。反应结束后,将反应液冷却至室温,加入水(10mL)和乙酸乙酯(10mL×3)萃取。有机相用无水硫酸钠干燥,过滤,减压蒸馏得到粗产品,粗产品经制备高效液相色谱柱(柱子:Welch Xtimate C18,柱长150mm,内径30mm,粒径5μm;流动相A:水(含0.225%氨水),流动相B:乙腈;梯度:流动相B从5%到95%用时24分钟,流速:24mL/min)分离,得标题化合物74(5.5mg,收率:25.8%)。
MS m/z(ESI):523.54[M+H] +
1H NMR(400MHz,DMSO-d6)δ8.63(s,1H),8.32(d,J=30.9Hz,1H),7.85(s,1H),7.64(s,1H),7.44(d,J=12.4Hz,2H),7.30(s,2H),3.51(s,1H),3.28(s,1H),3.22(s,3H),2.87(s,1H),2.67(s,1H),2.54(s,2H),2.35(s,1H),1.58(s,4H),1.07(s,5H),0.81(s,8H).
实施例19:(S)-8-环丙基-3-(5-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)吡啶-3-基)-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物75)的制备
Figure PCTCN2022128426-appb-000224
步骤1:(S)-1-(2-羟基-5-((3-甲基哌啶-1-基)甲基)苯基)乙烷-1-酮(75A)的制备
将1-(5-溴-2-羟基苯基)乙烷-1-酮(1.2g,5.58mmol)、2B(1.3g,6.14mmol)和碳酸钾(2.3g,16.74mmol)加入水(15mL)与1,4-二氧六环(60mL)的混合液中,并置换氮气。在氮气氛围下,将XphosPd G2(438.5mg,558.03μmol)与Xphos(532.0mg,1.12mmol)加入溶液中,并将反应升温至100℃搅拌16小时。反应结束后,将反应冷却至室温,加入水(50mL)和乙酸乙酯(50mL×3)萃取。有机相用无水硫酸钠干燥,过滤,减压蒸馏得到粗产品,粗产品经用正相柱色谱法分离(二氯甲烷/[甲醇(0.2M NH 3)]=10:1),得标题化合物75A(950mg,收率:69%)。
MS m/z(ESI):248.3[M+H] +
步骤2:(S)-1-(2-羟基-3-碘-5-((3-甲基哌啶-1-基)甲基)苯基)乙烷-1-酮(75B)的制备
将75A(0.95g,3.84mmol)溶于乙腈(50mL)中,然后加入一水合对甲苯磺酸(1.1g,5.76mmol)与NIS(1.3g,5.76mmol),混合物在氮气保护下回流过夜。反应液降至室温,用饱和亚硫酸钠水溶液淬灭,二氯甲烷萃取,减压蒸除溶剂,残留物用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000225
快速硅胶柱;流动相:乙腈/水=60:40)纯化,得到标题化合物75B(750.0mg,收率:52%)。
MS m/z(ESI):374.3[M+H] +
步骤3:(S)-8-碘-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(75C)的制备
将75B(0.75g,2.01mmol)与DMF-DMA的混合物加热至100℃,搅拌1小时后冷却至室温,减压蒸除低沸物,残留物用二氯甲烷溶解,然后加入浓盐酸,混合物搅拌2小时,冰水浴下用4N NaOH(a.q.)中和至中性,再加入饱和碳酸氢钠水溶液调至碱性,二氯甲烷萃取3次,减压浓缩至干,残留物用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000226
快速硅胶柱;流动相:乙腈/水=60:40)纯化,得到标题化合物75C(700.0mg,收率:91%)。
MS m/z(ESI):384.2[M+H] +
步骤4:(S)-8-环丙基-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(75D)的制备
往75C(700mg,1.83mmol)、环丙基三氟硼酸钾(541mg,3.65mmol)和碳酸钾(757mg,5.48mmol)的混合物中加入水(5mL)与甲苯(20mL),混合物置换氮气。在氮气氛围下,加入Pd(dppf)Cl 2(134mg,183μmol),所得混合物在氮气保护下加热至100℃,搅拌4小时。反应完毕后冷却至室温,分去水相,减压浓缩得到粗产品,残留物用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000227
快速硅胶柱;流动相:乙腈/水=60:40)分离,得到标题化合物75D(440.0mg,收率:81%)。
MS m/z(ESI):298.0[M+H] +
步骤5:(S)-8-环丙基-3-碘-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(75E)的制备
将75D(150mg,504.39μmol)溶于甲醇(10mL),然后加入二氢吡咯(102mg,1.43mmol),所得溶液回流1小时,冷却,减压浓缩至干,残留物用二氯甲烷(10mL)溶解,加入吡啶(120mg,1.51mmol)和碘(384mg,1.51mmol),搅拌2小时,反应完后用饱和亚硫酸钠水溶液淬灭,二氯甲烷萃取3次,无水硫酸镁干燥,减压浓缩至干,得到标题化合物75E粗品(200.0mg,收率:94%)。
MS m/z(ESI):424.0[M+H] +
步骤6:(S)-8-环丙基-3-(5-(3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)吡啶-3-基)-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物75)的制备
参照实施例17步骤4的合成方法,不同的是将步骤4中的73C替换为65G(10.96mg,30.95μmol),64C替换为75E(13.10mg,30.95μmol),同法制得标题化合物75(6.8mg,收率:42%)。
MS m/z(ESI):524.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.78(s,1H),8.67(s,1H),8.52(s,1H),8.32(s,1H),8.03(s,1H),7.84(s,1H),7.32(s,1H),3.56(s,2H),3.50(s,3H),2.93-2.9(m,2H),2.72-2.65(m,2H),2.58-2.56(m,2H),2.39-2.30(m,2H),1.90-1.80(m,1H),1.65-1.40(m,5H),1.10-1.00(s,5H),0.85-0.75(m,6H).
实施例20:3-(3-(环丙基氟(4-甲基-4H-1,2,4-三唑-3-基)甲基)苯基)-8-甲基-6-((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物120)
合成路线:
Figure PCTCN2022128426-appb-000228
步骤1:(3-溴苯基)(4-甲基-4H-1,2,4-三唑-3-基)甲醇(120A)
在-50℃氩气保护下,将正丁基锂(190.56mg、2.97mmol、0.518mL)加入4-甲基-1,2,4-三唑(247.01mg、2.97mmol)的DME溶液(20mL)混合物中,搅拌1小时,加入间溴苯甲醛(500mg、2.7mmol)并反应1h,随后缓慢升温至0℃,搅拌1h。反应完毕后用饱和NH 4Cl水溶液淬灭,乙酸乙酯(100mL)萃取,有机相无水硫酸钠干燥,过滤、浓缩,粗品经反相色谱柱(
Figure PCTCN2022128426-appb-000229
快速硅胶柱;流动相:乙腈/水=60:40)纯化,得标题化合物120A(189mg)。
MS m/z(ESI):268/270[M+H] +
步骤2:(3-溴苯基)(4-甲基-4H-1,2,4-三唑-3-基)甲酮(120B)
将120A(189mg,704.94μmol)溶于DCM(10mL)中,加入Dess-Martin试剂(1.23g,1.41mmol),室温搅拌4小时。经饱和Na 2SO 3水溶液淬灭,过滤、分离,乙酸乙酯(100mL)萃取,有机相无水硫酸钠干燥,过滤、浓缩,粗品经反相色谱柱(
Figure PCTCN2022128426-appb-000230
快速硅胶柱;流动相:乙腈/水=80:20)纯化得到标题化合物120B(162mg)。
MS m/z(ESI):266/268[M+H] +
步骤3:(3-溴苯基)(环丙基)(4-甲基-4H-1,2,4-三唑-3-基)甲醇(120C)
将120B(162mg,608.81μmol)溶于THF(2mL)中,在0℃氩气条件下向混合物中加入环丙基溴化镁(132.67mg,913.21μmol,0.456mL),加热至室温并搅拌1小时。经饱和NH 4Cl水溶液淬灭,浓缩经硅胶柱色谱法纯化得到标题化合物120C(102mg,330.98μmol,收率54.37%)。
MS m/z(ESI):308/310[M+H] +
步骤4:3-((3-溴苯基)(环丙基)氟甲基)-4-甲基-4H-1,2,4-三唑(120D)
将120C(102mg,330.98μmol)溶于DCM(2mL)中,冰浴冷却,用注射器加入DAST(106.70mg,661.97μmol),然后搅拌1小时。饱和NaHCO 3水溶液淬灭,经反相色谱柱(
Figure PCTCN2022128426-appb-000231
快速硅胶柱;流动相:乙腈/水=80:20)纯化,得标题化合物120D(51mg)。
MS m/z(ESI):310/312[M+H] +
步骤5:3-(环丙基氟(3-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)苯基)甲基)-4-甲基-4H-1,2,4-三唑(120E)
将120D(38mg,122.52μmol)、Pd(dppf)Cl 2(8.89mg,12.25μmol)、醋酸钾(12.02mg,122.52μmol)和B 2Pin 2(46.67mg,183.8μmol)混合,抽真空并充入氩气,然后加入1,4-二氧六环(2mL),将混合物加热至100℃,反应5小时。冷却至室温,过滤得到120E的粗品,无需纯化直接进行下一步。
MS m/z(ESI):358.2[M+H] +
步骤6:3-(3-(环丙基氟(4-甲基-4H-1,2,4-三唑-3-基)甲基)苯基)-8-甲基-6-((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物120)
向反应管中加入64C(35mg,88.11μmol)、120E(44.06mg,123.35μmol)、Pd(dppf)Cl 2(6.39mg,8.81μmol)和K 2CO 3(24.35mg,176.21μmol),抽真空并充入氩气,然后用注射器加入水(0.5mL)和二氧六环(2mL),加热至100℃,反应1小时。反应完毕后冷却至室温,经反相色谱柱(
Figure PCTCN2022128426-appb-000232
快速硅胶柱;流动相:乙腈/水=85:15)纯化,得到标题产物120(30mg)。
MS m/z(ESI):501.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.65(s,1H),8.55(s,1H),7.86(s,1H),7.70–7.50(m,3H),7.42(s,1H),7.19(d,J=7.7Hz,1H),3.51(s,2H),3.31(s,3H),2.80-2.66(m,2H),2.48(s,3H),2.20-2.00(m,1H),1.95-1.80(m,1H),1.69–1.42(m,5H),0.90-0.75(m,4H),0.75-0.60(m,2H),0.60-0.48(m,1H),0.48-0.40(s,1H).
步骤7:3-(3-((S)-环丙基氟(4-甲基-4H-1,2,4-三唑-3-基)甲基)苯基)-8-甲基-6-((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮和3-(3-((R)-环丙基氟(4-甲基-4H-1,2,4-三唑-3-基)甲基)苯基)-8-甲基-6-(((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮
将化合物120(27mg)经过超临界流体色谱法纯化分离(色谱柱:OD-H;柱长150mm,内径4.6mm;流动相A:IPA(含0.05%DEA),流动相B:超临界二氧化碳;梯度:流动相B从60%到60%;流速:2.5mL/min;)得到标题化合物120-P1(12mg)与标题化合物120-P2(8mg)。120-P1保留时间:1.673min
MS m/z(ESI):501.3[M+H] +
120-P2保留时间:2.097min
MS m/z(ESI):501.3[M+H] +
实施例21:7-氟-8-甲基-3-(3-(顺式-3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-6-(((s)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物121)
合成路线:
Figure PCTCN2022128426-appb-000233
步骤1:顺式-3-(1-(3-溴苯基)-3-甲基环丁基)-4-甲基-4H-1,2,4-三氮唑(1A-P1)的制备
将1A(2g,6.54mmol)进行制备分离纯化(色谱柱:Welch Xtimate C18柱长150mm,内径30mm,粒径5μm;流动相A:水(含0.225%NH 3),流动相B:乙腈;梯度:流动相B从5%到95%用时18分钟),得标题化合物1A-P1(610mg,收率:31%)。
MS m/z(ESI):;305.7,307.7[M+H] +
1H NMR(400MHz,CDCl3)δ7.97(s,1H),7.54-7.51(m,1H),7.41-7.37(m,1H),7.25-7.18(m,2H),3.18(s,3H),2.85-2.75(m,2H),2.70-2.58(m,3H),1.13(d,J=4.0Hz,3H).
步骤2:4-甲基-3-(顺式-3-甲基-1-(3-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)苯基)环丁基)-4H-1,2,4-三唑(1B-P1)
1A-P1(100mg,326.58μmol)、Pd(dppf)Cl 2(23.70mg,32.66μmol)和醋酸钾(32.05mg,326.58μmol)的混合物抽真空并充入氩气,然后加入二氧六环(2mL),将混合物加热至100℃,反应5小时。反应完毕后冷却至室温,过滤得到标题化合物1B-P1的粗品,直接用于下一步。MS m/z(ESI):354.2[M+H] +
步骤1:6-溴-3-氟-2-甲基苯酚(121A)的制备
将异丙胺(1.76g,29.73mmol)缓慢加到起始原料3-氟-2-甲基苯酚(5g,29.73mmol)和二氯甲烷(80mL)中,将混合物冷却在-78℃(干冰/乙醇)浴中将NBS(5.29g,29.73mmol)缓慢分批加到混合物中,混合物在-78℃(干冰/乙醇)浴搅拌0.5小时,反应恢复到室温,加入盐酸水溶液(2M,100mL)并搅拌10分钟,将有机相分离并旋干得到白色固体,向固体中加入石油醚(100mL)并搅拌10分钟,将混合物过滤,滤液旋干得标题化合物121A粗产品(6.9g),无需纯化直接用于下一步反应。
1H NMR(400MHz,CDCl 3)δ7.27-7.22(m,1H),6.60-6.55(m,1H),5.65(s,1H),2.23-2.21(m,3H).
步骤2:1-(4-氟-2-羟基-3-甲基苯基)乙-1-酮(121B)的制备
在氮气流下将Pd(PPh 3) 2Cl 2(826.77mg,1.18mmol)加到121A(6.9g),三丁基(1-乙氧基乙烯)锡(15.340g,42.48mmol,14.35mL)和1,4-二氧六环(100mL)中,置换氮气三次,将混合物加热至100℃并搅拌12小时,反应完毕后反应恢复到室温,加入盐酸水溶液(2M,13mL)搅拌1小时。反应液用饱和氟化钾水溶液充分淬灭,乙酸乙酯(100mL×3)萃取,有机相用无水硫酸钠干燥,过滤,浓缩,经正相柱色谱法(纯PE洗脱)纯化,得到标题化合物121B(3.5g)。MS m/z(ESI):168.16/168.7[M+H] +.
1H NMR(400MHz,CDCl 3)δ12.92-12.89(m,1H),7.65-7.55(m,1H),6.64-6.57(m,1H),2.61(s,3H),2.18-2.15(m,3H)
步骤3:1-(5-溴-4-氟-2-羟基-3-甲基苯基)乙-1-酮(121C)的制备
在0℃(冰水浴)条件下,往121B(3g,17.84mmol)的无水DCM(30mL)溶液中缓慢滴加液溴(4.37g,27.32mmol,1.50mL)和无水DCM(10mL)的混合溶液,滴加完毕后反应液在0℃(冰水浴)搅拌1.5小时,反应完毕后在0℃(冰水浴)条件下反应液用过量的偏重亚硫酸钠水溶液充分淬灭,二氯甲烷(50mL×3)萃取,有机相用无水硫酸钠干燥,过滤,减压浓缩,经正相柱色谱法(纯PE洗脱)纯化,得到标题化合物121C(2g)。
1H NMR(400MHz,CDCl 3)δ12.83-12.72(m,1H),7.84-7.75(m,1H),2.62(s,3H),2.22-2.16(m,3H)
步骤4:1-(5-溴-4-氟-2-羟基-3-甲基苯基)-3-(二甲基氨基)-丙-2-烯-1-酮(121D)的制备
将N,N-二甲基甲酰胺二甲基缩醛(3.38g,28.33mmol)加到121C(2g,8.10mmol)和甲苯(30mL)中,将混合物加热至115℃并搅拌12小时。反应完毕后恢复到室温,加水(20mL)淬灭,乙酸乙酯(30mL×3)萃取,有机相用无水硫酸钠干燥,过滤,减压浓缩,经正相柱色谱法(石油醚:乙酸乙酯=5:1)纯化,得到标题化合物121D(1g)。
MS m/z(ESI):302.14/303.8[M+H] +.
1H NMR(400MHz,CDCl 3)δ7.90(d,J=12.0Hz,1H),7.71(d,J=8.0Hz,1H),5.62(d,J=12.0Hz,1H),3.22(s,3H),3.11-2.93(m,3H),2.27-2.11(m,3H)
步骤5:6-溴-7-氟-8-甲基-4H-色烯-4-酮(121E)的制备
将121D(1g,3.31mmol)溶于乙酸酐(20mL)中,加热至140℃回流搅拌12小时。反应完毕后恢复到室温,加水(20mL)、乙酸乙酯(30mL×3)萃取,有机相用无水硫酸钠干燥,过滤,减压旋浓缩,粗品经正相柱色谱法(石油醚:乙酸乙酯=5:1)纯化,得到标题化合物121E(760mg)。MS m/z(ESI):257.06/258.9[M+H] +.
1H NMR(400MHz,CDCl 3)δ8.30(d,J=8.0Hz,1H),7.91(d,J=8.0Hz,1H),6.37-6.34(m,1H),2.44-2.41(m,3H)
步骤6:6-溴-7-氟-3-碘-8-甲基-4H-色烯-4-酮(121F)的制备
将121E(256mg,1.0mmol)、四氢吡咯(142mg,2.0mmol)加入甲醇(2mL)中,混合物加热至60℃,搅拌1小时。反应完毕后冷却至室温,减压浓缩得到粗产品。将粗品、碘单质(279.18mg,1.1mmol)、吡啶(237.0mg,3.0mmol)加入氯仿(2mL)中,所得混合物在常温下搅拌1小时。反应完毕后冷却至室温,减压浓缩得到粗产品,粗产品经正相快速柱色谱法(PE:EA=3:1)分离,得标题化合物121F(271.1mg,收率:71.0%)。
MS m/z(ESI):383.1[M+H] +
步骤7:6-溴-7-氟-8-甲基-3-(3-(顺式-3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-4H-色烯-4-酮(121G)
向反应管中加入121F(55mg,143.62μmol)、1B-P1(50.74mg,143.62μmol)、Pd(dppf)Cl 2(10.42mg,14.36μmol)和碳酸钾(39.70mg,287.24μmol),抽真空并充入氩气,然后用注射器加入水(0.5mL)和1,4-二氧六环(2mL),加热至100℃,反应1小时。反应完毕后冷却至室温,经反相色谱柱(
Figure PCTCN2022128426-appb-000234
快速硅胶柱;流动相:乙腈/水=80:20)纯化,得到标题产物121G(44mg)。
MS m/z(ESI):482.1/484.1[M+H] +
步骤8:7-氟-8-甲基-3-(3-(顺式-3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-6-(((s)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物121)
向反应管中加入121G(10mg,20.73μmol),2B(6.81mg,31.10μmol)、碳酸钾(5.73mg,41.46μmol)、XPhos Pd G2(1.63mg,2.07μmol)和XPhos(1.97mg,4.14μmol)抽真空并充入氩气,然后用注射器加入水(0.5mL)和1,4-二氧六环(2mL),加热至100℃,反应1小时。反应完毕后冷却至室温,经反相色谱柱(
Figure PCTCN2022128426-appb-000235
快速硅胶柱;流动相:乙腈/水=85:15)纯化,得到标题产物121(2mg)。
MS m/z(ESI):515.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.65(s,1H),8.29(s,1H),8.00(s,1H),7.62(s,1H),7.45(s,2H),7.30(s,1H),3.59(s,2H),3.22(s,3H),2.81(d,J=48.5Hz,5H),2.37(s,3H),1.91(s,1H),1.67–1.37(m,6H),1.23(s,1H),1.15-1.0(m,3H),0.9-0.75(m,4H).
实施例22:7-氟-8-甲基-3-(3-(顺式-3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-6-(((1-甲基环丁基)氨基)甲基)-4H-色烯-4-酮(化合物122)
合成路线:
Figure PCTCN2022128426-appb-000236
步骤1:1-甲基-N-((三氟-λ 4-硼烷基)甲基)环丁-1-胺钾盐(122A)
将1-甲基环丁胺盐酸盐(100mg,822.31μmol,HCl)溶于叔丁醇(2mL)中,并向混合物中加入THF(0.5mL)、(溴甲基)三氟硼酸钾(165.15mg,822.31μmol)和碳酸钾(340.95mg,2.47mmol),加热至80℃并搅拌6h。反应完毕后浓缩反应液并将残渣溶于丙酮(2mL)中,搅拌30分钟,过滤并浓缩,得到标题化合物122A的粗品(168mg),无需纯化直接用于下一步。
MS m/z(ESI):148.1[M-KF+H] +
步骤2:7-氟-8-甲基-3-(3-(顺式-3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-6-(((1-甲基环丁基)氨基)甲基)-4H-色烯-4-酮(化合物122)
121G(10mg,20.73μmol)、122A(23.12mg)、XphosPdG2(2.95mg,3.76μmol)、Xphos(3.58mg,7.52μmol)和碳酸钾(10.39mg,75.17μmol)的混合物抽真空并充入氩气,向混合物中加入水(0.5mL)和1,4-二氧六环(2mL),加热至100℃持续1小时。反应完毕后冷却至室温,加入水(10mL)和乙酸乙酯(10mL×3)萃取。有机相用无水硫酸钠干燥,过滤,浓缩,残余物经反相色谱柱(
Figure PCTCN2022128426-appb-000237
快速硅胶柱;流动相:乙腈/水=80:20)纯化得到标题化合物122(5mg)MS m/z(ESI):501.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.64(s,1H),8.31(s,1H),8.14(d,J=8.2Hz,1H),7.62(s,1H),7.44(d,J=4.8Hz,2H),7.30(s,1H),3.74(s,2H),3.23(s,3H),3.16(s,1H),2.90-2.84(m,2H),2.37(s,3H),2.00-1.95(m,2H),1.76–1.68(m,4H),1.31–1.23(m,6H),1.08(d,J=4.7Hz,3H).
实施例23:8-甲基-3-(3-(顺式-3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-6-(1-((1-甲基环丁基)氨基)乙基)-4H-色烯-4-酮(化合物123)
合成路线:
Figure PCTCN2022128426-appb-000238
步骤1:2-甲基-4-(1-((1-甲基环丁基)氨基)乙基)苯酚(123A)
将1-甲基环丁胺(971.74mg,7.99mmol,HCl)、1-(4-羟基-3-甲基苯基)乙酮(300mg,2.00mmol)、四异丙醇钛(2.27g,7.99mmol)、三乙胺(1.01g,9.99mmol,1.39mL)和DCE(10mL)的混合物加热至80℃过夜。反应完毕后冷却至室温,然后用冰浴冷却,加入MeOH(2mL)和NaBH 4(377.89mg,9.99mmol),搅拌2小时。反应完毕后用水淬灭,硅藻土过滤,乙酸乙酯萃取(5mL×3),有机相用无水硫酸钠干燥,过滤,浓缩,粗品经反相色谱柱(
Figure PCTCN2022128426-appb-000239
快速硅胶柱;流动相:乙腈/水=50:50)纯化,得标题化合物123A(270mg)。
MS m/z(ESI):220.2[M+H] +
步骤2:2-溴-6-甲基-4-(1-((1-甲基环丁基)氨基)乙基)苯酚(123B)
将123A(270mg,1.23mmol)溶于DCM(4.66mL)中,通过干燥乙醇浴冷却至-78℃,依次加入DIEA(249.14mg,2.46mmol,335.77μL)和NBS(328.67mg,1.85mmol),保持-78℃反应2小时。加入HCl水溶液(1M)淬灭,加入水(10mL)和DCM(10mL×3)萃取。有机相用无水硫酸钠干燥,过滤,浓缩,残余物通过反相色谱柱(
Figure PCTCN2022128426-appb-000240
快速硅胶柱;流动相:乙腈/水=50:50)纯化,得到标题产物123B(170mg)。
MS m/z(ESI):298.1/300.1[M+H] +
步骤3:1-(2-羟基-3-甲基-5-(1-((1-甲基环丁基)氨基)乙基)苯基)乙-1-酮(123C)
123B(50mg,167.66μmol)和Pd(PPh 3) 2Cl 2(19.37mg,16.77μmol)的混合物抽真空并充入氩气,然后向混合物中加入三正丁基(1-乙氧基乙烯基)锡(121.10mg,335.33μmol)和二氧六环(2mL),加热至100℃过夜。冷却至室温,加入1M HCl水溶液(1mL)并搅拌1小时。反应液用饱和氟化钾水溶液充分淬灭,乙酸乙酯(100mL X 3)萃取,有机相用无水硫酸钠干燥,过滤,浓缩,再经反相色谱柱(
Figure PCTCN2022128426-appb-000241
快速硅胶柱;流动相:乙腈/水=60:40)纯化,得到标题产物123C(22mg)。
MS m/z(ESI):262.2[M+H] +
步骤4:3-(二甲氨基)-1-(2-羟基-3-甲基-5-(1-((1-甲基环丁基)氨基)乙基)苯基)丙-2-烯-1-酮(123D)
将123C(27mg,103.31μmol)溶于DMF-DMA(3mL)中,加热至80℃,反应1小时。反应完毕后冷却至室温,浓缩得到标题产物123D粗品(32mg),无需纯化直接进行下一步。
MS m/z(ESI):317.2[M+H] +
步骤5:3-碘-8-甲基-6-(1-((1-甲基环丁基)氨基)乙基)-4H-色烯-4-酮(123E)
将123D(32mg,101.13μmol,)溶于DCM(5mL)中,依次加入I 2(35.29mg,139.05μmol)和吡啶(11.00mg,139.05μmol,11.20μL),搅拌1小时,再经反相色谱柱(
Figure PCTCN2022128426-appb-000242
快速硅胶柱;流动相:乙腈/水=80:20)纯化,得标题产物123E(13mg)。
MS m/z(ESI):398.1[M+H] +
步骤6:8-甲基-3-(3-(顺式-3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-6-(1-((1-甲基环丁基)氨基)乙基)-4H-色烯-4-酮(化合物123)
向反应管中加入123E(13mg,32.73μmol)、1B-P1(17.34mg,49.09μmol)、Pd(dppf)Cl 2(2.37mg,3.27μmol)和碳酸钾(9.05mg,65.45μmol),抽真空并充入氩气,然后用注射器加入水(0.5mL)和二氧六环(2mL),加热至100℃反应1小时。反应完毕后冷却至室温,过滤,减压浓缩,再经反相色谱柱(
Figure PCTCN2022128426-appb-000243
快速硅胶柱;流动相:乙腈/水=85:15)纯化,得到标题产物123(3.7mg)。
MS m/z(ESI):497.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.61(s,1H),8.29(s,1H),8.00(s,1H),7.77(s,1H),7.63(s,1H),7.50–7.40(m,2H),7.30(s,1H),4.00(s,1H),3.23(s,3H),2.87(s,2H),2.54(d,J=6.7Hz,3H),2.48(s,3H),1.85-1.71(s,2H),1.60-1.47(s,2H),1.42–1.21(m,6H),1.20-1.05(m,6H).
实施例24:3-(3-(环丙基(4-甲基-4H-1,2,4-三唑-3-基)甲基)苯基)-7-氟-8-甲基-6-(((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物124)的制备
Figure PCTCN2022128426-appb-000244
步骤1:(S)-7-氟-8-甲基-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(124A)的制备
将中间体121E(200mg,778.04μmol)、2B(339.40mg,1.56mmol)、XPhos Pd G2(61.22mg,77.80μmol)、XPhos(74.18mg,155.61μmol)和碳酸钾(214.74mg,1.56mmol)加入水(0.4mL)与1,4-二氧六环(2.0mL)的混合液中,并置换氮气。所得混合物在氮气保护下加热至100℃,搅拌12小时。反应完毕后冷却至室温,过滤,减压浓缩得到粗产品,粗产品经制备高效液相色谱柱(色谱柱:Welch Xtimate C18;柱长150mm,内径30mm,粒径5μm;流动相A:水(含0.225%NH 3),流动相B:乙腈;梯度:流动相B从5%到95%用时18分钟)分离,得标题化合物124A(150mg)。
MS m/z(ESI):290.2[M+H] +
步骤2:(S,E)-1-(4-氟-2-羟基-3-甲基-5-((3-甲基哌啶-1-基)甲基)苯基)-3-(吡咯烷-1-基)丙-2-烯-1-酮(124B)的制备
将124A(150mg,518.41μmol)、四氢吡咯(36.87mg,518.41μmol)加入甲醇(2mL)中,所得混合物加热至60℃,搅拌1小时。反应完毕后冷却至室温,减压浓缩得到标题产物124B(180.0mg)粗产品,无需进一步纯化直接用于下一步。
MS m/z(ESI):361.2[M+H] +
步骤3:(S)-7-氟-3-碘-8-甲基-6-((3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(124C)的制备
将124B(180mg,499.36μmol)、碘单质(253.48mg,998.71μmol)、吡啶(79.00mg,998.71μmol,80.45μL)加入氯仿(2mL)中,所得混合物在常温下搅拌1小时。反应完毕后冷却至室温,减压浓缩得到粗产品,粗产品经制备高效液相色谱柱(色谱柱:Welch Xtimate C18;柱长150mm,内径30mm,粒径5μm;流动相A:水(含0.225%NH 3),流动相B:乙腈;梯度:流动相B从5%到95%用时18分钟)分离,得标题化合物124C(80mg)。
MS m/z(ESI):416.1[M+H] +
步骤4:3-(3-(环丙基(4-甲基-4H-1,2,4-三唑-3-基)甲基)苯基)-7-氟-8-甲基-6-(((S)-3-甲基哌啶-1-基)甲基)-4H-色烯-4-酮(化合物124)的制备
将124C(60mg,144.49μmol)、63G(49.02mg,144.49μmol)、Pd(dppf)Cl 2(10.57mg,14.45μmol)和碳酸钾(39.88mg,288.99μmol)加入水(0.4mL)与1,4-二氧六环(2.0mL)的混合液中,并置换氮气。所得混合物在氮气保护下加热至100℃,搅拌2小时。反应完毕后冷却至室温,过滤,减压浓缩得到粗产品,粗产品经制备高效液相色谱柱(色谱柱:Welch Xtimate C18;柱长150mm,内径30mm,粒径5μm;流动相A:水(含0.225%NH 3),流动相B:乙腈;梯度:流动相B从5%到95%用时18分钟)分离,得标题化合物124(15mg)。
MS m/z(ESI):501.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.59(s,1H),8.35(s,1H),8.00(d,J=9.1Hz,1H),7.51(s,1H),7.48–7.36(m,2H),7.31(d,J=7.7Hz,1H),3.59(s,2H),3.55(d,J=8.9Hz,1H),3.41(s,3H), 2.37(s,3H),2.34–2.31(m,2H),1.98–1.87(m,2H),1.72–1.55(m,5H),0.88–0.84(m,1H),0.81(d,J=5.7Hz,3H),0.65–0.50(m,4H).
实施例25:8-甲基-3-(3-(顺式-3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-6-((1-甲基环丁基)氨基)甲基)-4H-吡喃并[2,3-c]吡啶-4-酮(化合物125)的制备
合成路线:
Figure PCTCN2022128426-appb-000245
步骤1:6-溴-2-甲基-3-((四氢-2H-吡喃-2-基)氧基)吡啶(125A)的制备
将起始原料6-溴-2-甲基吡啶-3-醇(100.0mg,0.53mmol)、DHP(134.21mg,1.60mmol)加入到四氢呋喃(2.0mL)中,然后加入PPTS(13.37mg,53.19μmol),混合物加热至70℃,搅拌16小时。粗产品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000246
快速硅胶柱;流动相:乙腈/水=80:20)纯化,得到标题化合物125A(82.0mg)。
MS m/z(ESI):186.96/188.96[M+H-THP] +
步骤2:6-甲基-5-((四氢-2H-吡喃-2-基)氧基)吡啶醛(125B)的制备
将125A(82.0mg,0.3mmol)加入到四氢呋喃(3.0mL)中,置换氮气并降温至-78℃,然后加入正丁基锂(0.12mL,0.3mmol),混合物搅拌0.5小时后,然后加入DMF(22.02mg,0.3mmol),在-78℃下搅拌1h。反应结束后,用饱和氯化铵水溶液淬灭反应,乙酸乙酯萃取(5mL×3)。有机相用无水硫酸钠干燥,过滤,并减压蒸馏得到粗产品。将粗产品用正相柱色谱法(石油醚:乙酸乙酯=90:10)纯化,得到标题化合物125B(53.0mg)。
MS m/z(ESI):272.0[M+H] +
步骤3:1-甲基-N-((6-甲基-5-((四氢-2H-吡喃-2-基)氧基)吡啶-2-基甲基)环丁-1-胺(化合物125C)的制备
将125B(53.0mg,0.24mmol)、1-甲基环丁胺盐酸盐(87.39mg,0.72mmol)和钛酸四异丙酯(272.33mg,0.96mmol)加入到DCE(2.0mL)中,然后加入三乙胺(121.2mg,1.2mmol),所得混合物加热至80℃,搅拌16小时。TLC监控原料反应完全后,向反应液加入甲醇(2.0mL),然后加入硼氢化钠(45.31mg,1.2mmol),在室温下搅拌3h。反应结束后,用水淬灭反应,乙酸乙酯萃取(5mL×3)。有机相用无水硫酸钠干燥,过滤,并减压蒸馏得到粗产品。粗产品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000247
快速硅胶柱;流动相:乙腈/水=65:35)纯化,得到标题化合物125C(48.0mg)。
MS m/z(ESI):207.2[M+H-THP] +
步骤4:2-甲基-6-(((1-甲基环丁基)氨基)甲基)吡啶-3-醇(化合物125D)的制备
在室温下,将125C(48.0mg,μmol)加入到甲醇(1.0mL)中,彻底溶清后加入盐酸二氧六环(4.0M,2.0mL)溶液。然后将反应液加热至45℃搅拌5小时。反应结束后,将反应液直接减压蒸馏得到标题化合物125D(34.0mg)粗产品,粗品无需进一步纯化。
MS m/z(ESI):207.14[M+H]+。
步骤5:4-溴-2-甲基-6-(((1-甲基环丁基)氨基)甲基)吡啶-3-醇(化合物125E)的制备
将125D(34.0mg,165.0μmol)、二异丙胺(33.4mg,330.0μmol)加入到二氯甲烷(1.0mL)中,并置换氮气并降温至-72℃,然后加入NBS(32.3mg,181.5μmol),在-72℃下搅拌2小时。反应结束后,用水淬灭反应,二氯甲烷萃取(5mL×3)。有机相用无水硫酸钠干燥,过滤,并减压蒸馏得到粗产品,粗产品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000248
快速硅胶柱;流动相:乙腈/水=50:50)纯化,得到标题化合物125E(37.0mg)。
MS m/z(ESI):285.05/287.05[M+H] +.
步骤6:1-(3-羟基-2-甲基-6-(((1-甲基环丁基)氨基)甲基)吡啶-4-基)乙-1-酮(125F)的制备
将125E(37.0mg,0.132mmol)、三丁基(1-乙氧基乙烯)锡(95.5mg,0.264mmol)加入到甲苯(1.0mL)中,并置换氮气。在氮气氛围下,加入双三苯基膦二氯化钯(9.25mg,13.2μmol),所得混合物在氮气保护下加热至100℃,搅拌16小时。粗产品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000249
快速硅胶柱;流动相:乙腈/水=70:30)纯化,将浓缩得到的中间体在盐酸二氧六环溶液(4.0M,mL)中室温搅拌1小时,反应液浓缩得到标题化合物125F(29.0mg)。
MS m/z(ESI):249.15[M+H] +
步骤7:3-(二甲氨基)-1-(3-羟基-2-甲基-6-((1-甲基环丁基)氨基)甲基)吡啶-4-基)丙-2-烯-1-酮(125G)的制备
将125F(29.0mg,117.0μmol)加入N,N-二甲基甲酰胺二甲基缩醛(1.0mL)中,将反应液加热至100℃搅拌1小时。反应结束后,将反应液直接减压蒸馏得到得到标题化合物125G粗产品(35.0mg),粗品无需进一步纯化,直接用于下一步反应。
MS m/z(ESI):304.19[M+H] +
步骤8:3-碘-8-甲基-6-(((1-甲基环丁基)氨基)甲基)-4H-吡喃并[2,3-c]吡啶-4-酮(125H)的制备
将125G(35.0mg,0.115mmol)和碘(58.7mg,0.23mmol)加入到氯仿(2.0mL)中,然后加入吡啶(0.3mL),室温下搅拌1小时。反应完毕后饱和亚硫酸氢钠水溶液(1mL)淬灭,过滤,滤液浓缩得粗品,粗产品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000250
快速硅胶柱;流动相:乙腈/水=80:20)纯化,得到标题化合物125H(30.0mg)。
MS m/z(ESI):385.03[M+H] +
步骤9:8-甲基-3-(3-(顺式-3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-6-((1-甲基环丁基)氨基)甲基)-4H-吡喃并[2,3-c]吡啶-4-酮(化合物125)的制备
将125H(30.0mg,77.9μmol)、1B-P1(33.0mg,93.5μmol)和碳酸钾(21.5mg,156.0μmol)加入水(0.4mL)与1,4-二氧六环(2.0mL)的混合液中,并置换氮气。在氮气氛围下,加入Pd(dppf)Cl 2(6.0mg,8.0μmol),所得混合物在氮气保护下加热至100℃,搅拌1.0小时。反应结束后,反应液冷却至室温,过滤,浓缩,粗产品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000251
快速硅胶柱;流动相:乙腈/水=70:30)纯化,得到标题化合物125(7.1mg)。
MS m/z(ESI):484.26[M+H] +.
实施例26:3-(3-(顺式-3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-7-(1-((1-甲基环丁基)氨基)乙基)-9-(三氟甲基)-4H-吡啶并[1,2-a]嘧啶-4-酮(化合物126)的制备
合成路线:
Figure PCTCN2022128426-appb-000252
步骤1:1-(6-氨基-5-(三氟甲基)吡啶-3-基)乙-1-酮(126A)的制备
将5-溴-3-(三氟甲基)吡啶-2-胺(300.0mg,1.24mmol)、三丁基(1-乙氧基乙烯)锡(674.32mg,1.87mmol)加入到甲苯(10.0mL)中,并置换氮气。在氮气氛围下,加入双三苯基膦二氯化钯(87.37mg,124.48μmol),所得混合物在氮气保护下加热至100℃,搅拌16.0小时。反应结束后,用水淬灭反应,乙酸乙酯萃取(5mL×3)。有机相用无水硫酸钠干燥,过滤,浓缩,粗产品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000253
快速硅胶柱;流动相:乙腈/水=70:30)纯化,将浓缩得到的中间体用盐酸二氧六环溶液(4.0M,5.0mL)在室温下搅拌1.0小时,反应液浓缩后得到标题化合物126A(180.0mg)。
MS m/z(ESI):205.15[M+H] +
步骤2:5-(1-((1-甲基环丁基)氨基)乙基)-3-(三氟甲基)吡啶-2-胺(126B)的制备
将126A(180.0mg,0.88mmol)、1-甲基环丁胺盐酸盐(321.67mg,2.65mmol)和钛酸四乙酯(1.0g,3.53mmol)加入到DCE(4.0mL)中,然后加入三乙胺(446.1mg,4.41mmol),所得混合物加热至80℃,搅拌16.0小时。TLC监控原料消失后,向反应液加入甲醇(4.0mL),然后加入硼氢化钠(116.79mg,4.41mmol),在室温下搅拌3.0h。反应结束后,用水淬灭,硅藻土过滤,乙酸乙酯萃取(5mL×3),有机相用无水硫酸钠干燥,过滤,浓缩,粗产品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000254
快速硅胶柱;流动相:乙腈/水=65:35)纯化,得到标题化合物126B(180.0mg)。
MS m/z(ESI):274.3[M+H] +
步骤3:7-(1-((1-甲基环丁基)氨基)乙基)-9-(三氟甲基)-4H-吡啶并[1,2-a]嘧啶-4-酮(化合物126C)的制备
将126B(180.0mg,658.63μmol)和5-(甲氧亚甲基)-2,2-二甲基-1,3-二氧杂环已烷-4,6-二酮(122.61mg,658.63μmol)加入到二苯醚(3.0mL)中,升温至120℃搅拌1.0小时。TLC监控原料消失后,将反应升温至210℃搅拌1.0小时。随后将反应冷却至室温,所得粗产品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000255
快速硅胶柱;流动相:乙腈/水=60:40)纯化,得到标题化合物126C(20.0mg)。
MS m/z(ESI):326.33[M+H] +
步骤4:3-碘-7-(1-((1-甲基环丁基)氨基)乙基)-9-(三氟甲基)-4H-吡啶并[1,2-a]嘧啶-4-酮(化合物126D)的制备
在室温下,依次将126C(20.0mg,61.48μmol)、硝酸铈铵(32.58mg,561.48μmol)和碘(15.6mg,61.48μmol)加入到乙腈溶液(3.0mL)中。然后将反应液搅拌6小时。反应结束后,加入饱和的亚硫酸钠水溶液(10mL),二氯甲烷萃取(50mL×3)。有机相用无水硫酸钠干燥,过滤,并减压蒸馏得到粗产品。将粗产品经反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000256
快速硅胶柱;流动相:乙腈/水=50:50)纯化得到标题化合物126D(10.0mg)。
MS m/z(ESI):452.22[M+H] +
步骤5:3-(3-(顺式-3-甲基-1-(4-甲基-4H-1,2,4-三唑-3-基)环丁基)苯基)-7-(1-((1-甲基环丁基)氨基)乙基)-9-(三氟甲基)-4H-吡啶并[1,2-a]嘧啶-4-酮(化合物126)的制备
将126D(10.0mg,22.16μmol)、1B-P1(11.74mg,33.24μmol)和碳酸钾(6.12mg,44.32μmol)加入水(0.4mL)与1,4-二氧六环(2.0mL)的混合液中,并置换氮气。在氮气氛围下,加入Pd(dppf)Cl 2(1.62mg,2.22μmol),所得混合物在氮气保护下加热至100℃,搅拌1.0小时。反应完毕后将所得粗产品用反相柱色谱法(色谱柱:
Figure PCTCN2022128426-appb-000257
快速硅胶柱;流动相:乙腈/水=50:50)纯化得到标题化合物126(2.4mg)。
MS m/z(ESI):551.64[M+H] +.
1H NMR(400MHz,DMSO-d6)δ9.29(s,1H),8.68(s,1H),8.61(s,1H),8.31(s,1H),7.93(s,1H),7.69(d,J=7.7Hz,1H),7.48(t,J=7.8Hz,1H),7.30(d,J=7.8Hz,1H),4.18(d,J=6.2Hz,1H),3.25(s,3H),2.90(s,2H),2.57(d,J=6.5Hz,3H),1.91(t,J=9.7Hz,1H),1.77(t,J=9.4Hz,2H),1.59(d,J=6.9Hz,2H),1.50(s,1H),1.34(d,J=6.7Hz,3H),1.15(s,3H),1.11(d,J=4.7Hz,3H).
实施例27:8-甲基-6-(((S)-3-甲基哌啶-1-基)甲基)-3-(3-(1,2,2,2-四氟-1-(4-甲基-4H-1,2,4-三唑-3-基)乙基)苯基)-4H-色烯-4-酮(化合物128)的制备
合成路线:
Figure PCTCN2022128426-appb-000258
步骤1:1-(3-溴苯基)-2,2,2-三氟-1-(4-甲基-4H-1,2,4-三唑-3-基)乙-1-醇(128A)的制备
将4-甲基-4H-1,2,4-三唑(102.46mg,1.23mmol)溶于四氢呋喃(10.0mL)中,将反应液降温至-60℃后,向反应液中滴加正丁基锂的正己烷溶液(2.5M,0.492mL,1.23mmol),滴加完毕后,反应液继续保持-60℃搅拌1.0小时。然后滴加1-(3-溴苯基)-2,2,2-三氟乙-1-酮(312.0mg,1.23mmol)溶于四氢呋喃(1.5mL)的溶液。滴加完毕后,将反应液转移至室温反应0.5小时。反应结束后,将反应液冰浴降温至0℃,然后加入饱和的氯化铵水溶液并搅拌十分钟。搅拌完毕后,用乙酸乙酯(60.0mL)萃取三次,所得的有机相干燥并浓缩,粗品用石油醚(60.0mL)洗涤2次,得到标题化合物128A(250.0mg)。
MS m/z(ESI):336.0/338.0[M+H]+.
步骤2:3-(1-(3-溴苯基)-1,2,2,2-四氟乙基)-4-甲基-4H-1,2,4-三唑(128B)的制备
将128A(100.0mg,0.3mmol)溶于二氯甲烷(3.0mL)中,将反应液降温至0℃后,向反应液中滴加DAST(0.079mL,0.6mmol),滴加完毕后,反应液转移至室温反应0.5小时。反应结束后,将反应液冰浴降温至0℃,然后加入饱和的碳酸氢钠水溶液并搅拌十分钟。搅拌完毕后,用二氯甲烷(30.0mL)萃取三次,所得有机相用无水硫酸钠干燥,过滤并浓缩,所得粗品经正相柱色谱法(二氯甲烷:甲醇=95:5)纯化,得到标题化合物128B(60.0mg)。
MS m/z(ESI):338.0/340.0[M+H] +.
步骤3:4-甲基-3-(1,2,2,2-四氟-1-(3-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)苯基)乙基)-4H-1,2,4-三唑(128C)的制备
向128B(60.0mg,177.46μmol)、联硼酸频那醇酯(67.61mg,266.19μmol)、乙酸钾(52.17mg,532.39μmol)和无水二氧六环(2.0mL)的混合液中,加入Pd(dppf)Cl 212.88mg,17.75μmol),所得混合物在氮气保护下加热至100℃,搅拌1小时。反应完毕后冷却至室温,过滤,减压浓缩,残留物经正相柱色谱法(石油醚:乙酸乙酯=0:1)纯化得到标题化合物128C(60.0mg)。
MS m/z(ESI):386.1[M+H] +.
步骤4:8-甲基-6-(((S)-3-甲基哌啶-1-基)甲基)-3-(3-(1,2,2,2-四氟-1-(4-甲基-4H-1,2,4-三唑-3-基)乙基)苯基)-4H-色烯-4-酮(化合物128)的制备
将128C(60.0mg,155.78μmol)、64C(41.26mg,103.85μmol)和碳酸钾(28.66mg,207.7μmol)加入水(0.2mL)与1,4-二氧六环(1.0mL)的混合液中,并置换氮气。在氮气氛围下,加入Pd(dppf)Cl 2(7.6mg,10.39μmol),所得混合物在氮气保护下加热至100℃,搅拌1.0小时。反应完毕后冷却至室温,过滤,减压浓缩得到粗品,粗品经制备高效液相色谱柱(色谱柱:Welch Xtimate C18;柱长150mm,内径30mm,粒径5μm;流动相A:水(含0.225%NH 3),流动相B:乙腈;梯度:流动相B从5%到95%用时18分钟)分离,得标题化合物128(25.0mg)。
MS m/z(ESI):529.3[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ8.78(s,1H),8.71(s,1H),7.87(s,1H),7.81(d,J=7.7Hz,1H),7.66(t,J=8.5Hz,3H),7.41(d,J=7.8Hz,1H),3.52(s,2H),2.73(t,J=11.0Hz,2H),2.50(s,3H),1.89(t,J=10.7Hz,1H),1.63(dd,J=29.5,10.5Hz,4H),1.48(q,J=12.2Hz,1H),0.87(s,1H),0.82(d,J=5.0Hz,3H).
实施例28:3-(3-(环丙基氟(4-甲基-4H-1,2,4-三唑-3-基)甲基)苯基)-6-(((R)-4,4-二氟-3-甲基哌啶-1-基)甲基)-8-甲基-4H-色烯-4-酮(化合物129)
合成路线:
Figure PCTCN2022128426-appb-000259
步骤1:6-(羟甲基)-8-甲基-4H-色烯-4-酮(129A)
参考实施例18中74A的合成步骤,用58A代替1-(5-溴-2-羟基-3-碘苯基)乙基-1-酮,合成6-溴-8-甲基-4H-色烯-4-酮。MS m/z(ESI):239.0,241.0[M+H] +
向干燥的反应管中加入6-溴-8-甲基-4H-色烯-4-酮(500mg,2.09mmol)、Pd(PPh 3) 4(241.68mg,209.15μmol)、(三丁基锡)甲醇(2.01g,6.27mmol)和二氧六环(5mL),抽真空并充入氩气,加热至80℃,反应8小时。反应完毕后冷却至室温,经反相色谱柱(
Figure PCTCN2022128426-appb-000260
快速硅胶柱;流动相:乙腈/水=60:40)纯化,得标题产物129A(180mg)。
MS m/z(ESI):191[M+H] +
步骤2:(6-(氯甲基)-8-甲基-4H-色烯-4-酮(129B)
将129A(100mg,525.78μmol)溶于DCM(2mL)中,将二氯亚砜(125.10mg,1.05mmol,76.38μL)滴入混合物中,在室温下搅拌3小时。反应完毕后浓缩反应液得到标题化合物129B的粗品(109.7mg),无需纯化直接用于下一步。
MS m/z(ESI):209[M+H] +
步骤3:(R)-6-((4,4-二氟-3-甲基哌啶-1-基)甲基)-8-甲基-4H-色烯-4-酮(129C)
将(3R)-4,4-二氟-3-甲基哌啶(65mg,480.93μmol)和129B(100.34mg,480.93μmol)的混合物溶于DMF(2mL)中,向混合物中加入KI(7.98mg,48.09μmol)和K 2CO 3(132.93mg,961.86μmol),在室温下搅拌6小时。反应完全后加入水和乙酸乙酯萃取,饱和食盐水洗涤,有机相用无水硫酸钠干燥,过滤、浓缩,残余物经反相色谱柱(
Figure PCTCN2022128426-appb-000261
快速硅胶柱;流动相:乙腈/水=75:25)纯化,得到标题产物129C(71mg)。
MS m/z(ESI):308.1[M+H] +
步骤4:(R)-1-(5-((4,4-二氟-3-甲基哌啶-1-基)甲基)-2-羟基-3-甲基苯基)-3-(吡咯烷-1-基)丙-2-烯-1-酮(129D)
将129C(71mg,231.02μmol)溶于甲醇(2mL)中,加入吡咯烷(49.29mg,693.06μmol)溶解,并加热至50℃,搅拌1小时。反应完毕后冷却至室温,反应液浓缩得到标题化合物129D粗品(87mg),无需纯化直接用于下一步。
MS m/z(ESI):379.2[M+H] +
步骤5:(R)-6-((4,4-二氟-3-甲基哌啶-1-基)甲基)-3-碘-8-甲基-4H-色烯-4-酮(129E)
将129D(87mg,229.88μmol)溶于DCM(5mL)中,向混合物中加入I 2(116.69mg,459.76μmol)和吡啶(36.37mg,459.76μmol,37.04μL),搅拌1小时。反应完毕后反应液浓缩,经反相色谱柱(
Figure PCTCN2022128426-appb-000262
快速硅胶柱;流动相:乙腈/水=80:20)纯化,得到标题产物129E(60mg)。
MS m/z(ESI):434[M+H] +
步骤6:3-(3-(环丙基氟(4-甲基-4H-1,2,4-三唑-3-基)甲基)苯基)-6-(((R)-4,4-二氟-3-甲基哌啶-1-基)甲基)-8-甲基-4H-色烯-4-酮(129)
向反应管中加入129E(50mg,115.41μmol)、120E(57.72mg,161.58μmol)、Pd(dppf)Cl 2(8.37mg,11.54μmol)和K 2CO 3(31.90mg,230.82μmol),抽真空并充入氩气,然后用注射器加入水(0.5mL)和二氧六环(2mL),加热至100℃反应1小时。冷却至室温,过滤,浓缩,经反相色谱柱(
Figure PCTCN2022128426-appb-000263
快速硅胶柱;流动相:乙腈/水=90:10)纯化得到标题产物129(18mg)。
MS m/z(ESI):537.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ8.64(s,1H),8.54(s,1H),7.88(s,1H),7.64(s,1H),7.60-7.46(m,2H),7.42(s,1H),7.19(d,J=7.7Hz,1H),3.61(s,2H),3.32(s,3H),2.81-2.65(m,2H),2.48(s,3H),2.31–2.21(m,1H),2.15-1.95(m,4H),1.30-1.20(m,1H),0.91(d,J=6.5Hz,3H),0.75-0.6(m,2H),0.58–0.50(m,1H),0.48-0.38(m,1H).
参考上述实施例的合成方法以及前述通用合成路线1、合成路线2、合成路线3或合成路线4,合成如下化合物,其结构和质谱数据为:
Figure PCTCN2022128426-appb-000264
Figure PCTCN2022128426-appb-000265
Figure PCTCN2022128426-appb-000266
Figure PCTCN2022128426-appb-000267
Figure PCTCN2022128426-appb-000268
Figure PCTCN2022128426-appb-000269
Figure PCTCN2022128426-appb-000270
Figure PCTCN2022128426-appb-000271
Figure PCTCN2022128426-appb-000272
生物学活性及相关性质测试例
测试例1:Cbl-b活性实验
实验目的:测试化合物对Cbl-b蛋白与UbcH5B-Ub相互作用抑制活性
实验方法:Eu-Ubquitin(Cisbio)与UbcH5B(ENZO),E1(ENZO)在37℃孵育4小时制备Eu-Ubquitin-UbcH5B。Eu-Ubquitin-UbcH5B分装,于-80℃保存。Cbl-b活性实验在384孔板(Perkin Elmer)中进行。100nL 3倍梯度稀释化合物(终浓度为10μM-0.5nM,起始浓度10μM,进行3倍稀释,10个点,第10个点是0.5nM)与5μL 50nM Biotin-Cbl-b蛋白(Sigma)在室温孵育1小时,反应缓冲液为50mM HEPES pH 7.0(Gibco),100mM NaCl(Sigma),0.01%Triton X-100(Sigma),0.01%BSA(Sigma)以及l mM DTT(Invitrogen)。在反应板中加入5μL Src混合液(40nM Src(R&D),2mM ATP(Sigma),10mM MgCl 2(Sigma)),室温孵育3小时。在反应板中加入10μL检测液(12.5nM Strepdividin-XL665(Cisbio),500nM Eu-Ubquitin-UbcH5B,120nM EDTA(Invitrogen),0.004%BSA(Sigma)),室温孵育过夜,在Envision(Perkin Elmer)上读取HTRF信号(665nm/615nm)。使用IDBS XLfit计算IC 50
实验结果如表1所示。
表1
化合物编号 IC 50(nM)
1 76.6
2 40.3
2-P1 27.1
2-P2 30.5
3 295.9
4 267.9
5 449.6
8 366.2
9 130.1
10 42.9
11 31.2
12 63.9
13 31.9
14 29.3
17 167.5
58 9.7
59 43.1
62 23.6
63 41.6
64 38.4
65 17.0
66 88.9
67 96.4
68 17.8
69 15.0
71 203.2
73 64.9
74 19.1
75 7.4
88 32.7
89 41.6
120 17.5
120-P1 212.4
120-P2 7.8
121 10.1
122 12.3
123 5.4
124 19.7
126 11.1
127 13.3
128 28.6
129 17.8
测试例2:Jurkat T激活实验
实验目的:测试化合物对Jurkat T细胞IL-2释放激活作用
实验方法:96孔细胞板(Corning)使用2μg/mL Anti-Human CD3 Clone OKT3(BD)37℃包被4小时。3倍梯度稀释化合物(终浓度为10μM-4.6nM,起始浓度10μM,进行3倍稀释,8个点,第8个点是4.6nM)与220μL 1.11x10 6/mL Jurkat T细胞(ATCC)孵育1小时,加入5μL 45μg/mL Anti-Human CD28 Clone CD28.2(BD),混合均匀,转移100μL至前述CD3包被细胞板中,37℃细胞培养箱培养48小时,收上清使用IL-2ELISA试剂盒(BD)检测IL-2释放量。使用Prism分析EC 50
实验结果如表2所示。
表2
化合物编号 EC 50(nM)
2 634.2
2-P1 425.9
2-P2 400.8
58 112.1
62 524.9
65 265.0
68 456.4
69 552.3
120 687.9
121 250.5
122 554.6
123 53.6
124 382.6
126 80.5
127 208.6
128 279.3

Claims (26)

  1. 式(I)化合物或其立体异构体或其药学上可接受的盐:
    Figure PCTCN2022128426-appb-100001
    其中,
    Figure PCTCN2022128426-appb-100002
    选自以下情况中的任一种:i)C=C-A 3,所述A 3选自CR 11aR 11b、NR 12、O或S;ii)A 1-C=A 3,所述A 1选自C或N,A 3选自CR 11c或N;iii)A 1-A 2-A 3,所述A 1和A 2独立地选自C(R 11) n或N,A 3选自CR 11aR 11b、NR 12、O或S;
    n选自0或1;
    R 11a、R 11b、R 11c、R 11、R 12彼此独立地选自H、卤素、OH、CN、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 6环烷基或C 3-C 6环烷基氧基,其中所述的C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 6环烷基或C 3-C 6环烷基氧基任选被R 11d取代;
    Q环选自苯基、5-6元杂芳基或5-7元杂环基,所述苯基、5-6元杂芳基或5-7元杂环基任选被R 10取代;
    R 10选自卤素、=O、OH、NH 2、NO 2、CN、C 1-C 6烷基、C 2-C 4烯基、C 2-C 4炔基、C 1-C 6烷氧基、C 3-C 6环烷基、C 3-C 6环烷基氧基、C 3-C 6环烷基-NH-、4-7元杂环基、4-7元杂环基氧基或4-7元杂环基-NH-,所述NH 2、C 1-C 6烷基、C 2-C 4烯基、C 2-C 4炔基、C 1-C 6烷氧基、C 3-C 6环烷基、C 3-C 6环烷基氧基、C 3-C 6环烷基-NH-、4-7元杂环基、4-7元杂环基氧基或4-7元杂环基-NH-任选被R 10a取代;
    Y 1、Y 2、Y 3和Y 4独立选自CR b或N;
    X选自卤素、CN、OH、COOH、CONH 2、C 1-C 6烷基、C 1-C 6烷氧基、
    Figure PCTCN2022128426-appb-100003
    其中C 1-C 6烷基或C 1-C 6烷氧基任选被R e取代,环B选自任选被R 3取代的以下基团:4-10元含氮杂环基或5-10元含氮杂芳基,环D选自任选被R 6取代的以下基团:C 3-C 10环烷基、4-10元杂环基、苯基或5-10元杂芳基,并且环D以非N原子与L相连,L选自键、-NR 7-、-NR 7CR 8R 9-、-O-、-C(=O)-、-C(=O)NH-或-CR 8R 9-;
    R b选自H、卤素、OH、CN、C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6烷硫基、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、NHC(O)(C 1-C 6烷基)、NHC(O)-O(C 1-C 6烷基)、N(C 1-C 6烷基)C(O)-O(C 1-C 6烷基)、NHS(O) 2(C 1-C 6烷基)、C 3-C 6环烷基、C 3-C 6环烷基-O-、C 3-C 6环烷基-NH-、N(C 3-C 6环烷基) 2、NHC(O)-C 3-C 6环烷基、NHS(O) 2-C 3-C 6环烷基、4-7元杂环基、4-7元杂环基氧基、4-7元杂环基-NH-、N(4-7元杂环基) 2、NHC(O)-4-7元杂环基、NHS(O) 2-4-7元杂环基、C 6-C 10芳基、C 6-C 10芳基氧基、C 6-C 10芳基-NH-、N(C 6-C 10芳基) 2、NHC(O)-C 6-C 10芳基、NHS(O) 2-C 6-C 10芳基、5-10元杂芳基、5-10元杂芳基氧基或5-10元杂芳基-NH-、N(5-10元杂芳基) 2、NHC(O)-5-10元杂芳基、NHS(O) 2-5-10元杂芳基,其中所述的C 1-C 6烷基、C 1-C 6烷氧基、C 1-C 6烷硫基、C 3-C 6环烷基、4-7元杂环基、C 6-C 10芳基或5-10元杂芳基任选被R 2a取代;
    或者两个R b与其相连的C原子共同形成C 3-C 6环烯基、苯基、4-7元杂环基或5-6元杂芳基,所述C 3-C 6环烯基、苯基、4-7元杂环基或5-6元杂芳基任选被R 2a取代;
    R 4、R 5、R 7、R 8和R 9彼此独立地选自H、卤素、OH、C 1-C 6烷基或C 1-C 6烷氧基,所述C 1-C 6烷基或C 1-C 6烷氧基任选被R 4a取代;
    或者R 8、R 9及其相连的原子一起形成C 3-C 6环烷基或4-7元杂环基,所述C 3-C 6环烷基或4-7元杂环基任选进一步被R 8a取代;
    或者R 4、R 5及其相连的原子一起形成C 3-C 6环烷基或4-7元杂环基,所述C 3-C 6环烷基或4-7元杂环基任选进一步被R 8a取代;或者当p取自2时,两个R 4及其相连的原子一起形成C 3-C 6环烷基或4-7元杂环基,所述C 3-C 6环烷基或4-7元杂环基任选进一步被R 8a取代;
    或者R 4、R 5一起形成=O;
    R 3和R 6独立地选自卤素、CN、=O、OH、NO 2、C 1-C 6烷基、OR 6a、SR 6a、N(R 6a) 2、S(O) 2R 6a、S(O) 2N(R 6a) 2、S(O)R 6a、S(O)N(R 6a) 2、C(O)R 6a、C(O)OR 6a、C(O)N(R 6a) 2、C(O)N(R 6a)OR 6a、OC(O)R 6a、OC(O)N(R 6a) 2、N(R 6a)C(O)OR 6a、N(R 6a)C(O)R 6a、N(R 6a)C(O)N(R 6a) 2、N(R 6a)C(NR 6a)N(R 6a) 2、N(R 6a)S(O) 2N(R 6a) 2、N(R 6a)S(O) 2R 6a、C 3-C 10环烷基、4-7元杂环基、6-10元芳基或5-10元杂芳基,其中C 1-C 6烷基、C 3-C 10环烷基、4-7元杂环基、C 6-C 10元芳基或5-10元杂芳基任选进一步被R 3a取代;
    每一个R 6a独立地选自H、C 1-C 6烷基、苯基、4-7元杂环基或5-6元杂芳基,所述C 1-C 6烷基、苯基、4-7元杂环基或5-6元杂芳基任选进一步被R 6b取代,或者一个N原子上的2个R 6a与其相连的N一起形成4-7元杂环基或5-6元杂芳基,所述4-7元杂环基或5-6元杂芳基任选进一步被R 6b取代;
    R 3a、R 4a、R 6b和R e独立选自卤素、OH、CN、=O、C 1-C 6烷基、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、COOH或C 1-C 6烷氧基,其中所述的C 1-C 6烷基或C 1-C 6烷氧基进一步任选被R f取代;
    R f选自卤素、OH、=O、NH 2、NH(C 1-C 6烷基)或N(C 1-C 6烷基) 2
    R 11d、R 2a和R 10a独立选自卤素、OH、CN、=O、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 1-C 6烷基、卤代C 1-C 6烷基、C 1-C 6烷氧基或卤代C 1-C 6烷氧基;
    R 1和R 2独立选自H、卤素、CN、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 10环烷基或4-10元杂环基,其中所述的C 1-C 6烷基、C 1-C 6烷氧基、C 3-C 10环烷基或4-10元杂环基任选被R 1a取代,
    或者R 1、R 2与其连接的原子共同形成C 3-C 10环烷基或4-10元杂环基,所述C 3-C 10环烷基或4-10元杂环基任选被R 1b取代;
    或者R 1、R b及其各自相连的原子和键共同形成C 3-C 6环烯基或4-7元杂环基,所述C 3-C 6环烯基或4-7元杂环基任选被R 1d取代;
    R 1a和R 1b独立选自卤素、OH、CN、=O、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 1-C 6烷基或C 1-C 6烷氧基,所述C 1-C 6烷基或C 1-C 6烷氧基任选进一步被R 1c取代;
    R 1c选自卤素、OH、CN、=O、NH 2或COOH;
    W选自(CR 13R 14) kW 1;所述W 1选自5-10元杂芳基或4-10元杂环基,所述5-10元杂芳基、4-10元杂环基任选被R 15取代;R 13、R 14独立选自H、卤素、OH、C 1-C 6烷基或C 1-C 6烷氧基;R 15选自卤素、OH、NH 2、NH(C 1-C 6烷基)、N(C 1-C 6烷基) 2、C 1-C 6烷基、C 3-C 10环烷基或4-7元杂环基,其中所述的C 1-C 6烷基、C 3-C 10环烷基或4-7元杂环基任选被R 15a取代;
    或者R 1与R 13及其各自相连的原子和键共同形成C 3-C 6环烷基或4-7元杂环基,所述C 3-C 6环烷基或4-7元杂环基任选被R 13a取代;
    R 8a、R 13a和R 1d独立地选自卤素、OH、CN、C 1-C 6烷基或C 1-C 6烷氧基,所述C 1-C 6烷基或C 1-C 6烷氧基任选进一步被卤素取代;R 15a选自卤素、=O、OH、CN或C 1-C 6烷基;
    k选自0或1;
    p选自0、1或2。
  2. 根据权利要求1所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,
    Figure PCTCN2022128426-appb-100004
    选自A 1-C=A 3,所述A 1选自C或N,A 3选自CR 11c或N;或者所述A 1选自N,A 3选自CR 11c或N;或者所述A 1选自N,A 3选自N或C-F;或者所述A 1选自N,A 3选自N。
  3. 根据权利要求1所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,
    Figure PCTCN2022128426-appb-100005
    选自C=C-A 3,所述A 3选自CR 11aR 11b、NR 12、O或S;或者所述A 3选自NR 12、O或S;或者所述A 3选自NH、N-CH 3、O或S;或者所述A 3选自O。
  4. 根据权利要求1至3中任一项所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,Q环选自任选被R 10取代的以下基团:苯基、吡啶基、嘧啶基、吡唑基、咪唑基、噁唑基、异噁唑基、噻唑基、异噻唑基或5-7元杂环基,所述5-7元杂环基包含1或2个N作为杂原子;或者,
    Q环选自任选被R 10取代的以下基团:苯基、
    Figure PCTCN2022128426-appb-100006
    Figure PCTCN2022128426-appb-100007
    或者,
    Q环选自任选被R 10取代的以下基团:苯基、
    Figure PCTCN2022128426-appb-100008
    Figure PCTCN2022128426-appb-100009
    或者,
    Q环选自任选被R 10取代的
    Figure PCTCN2022128426-appb-100010
    或者,
    Q环选自任选被R 10取代的苯基。
  5. 根据权利要求1-4中任一项所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,R 10选自卤素、=O、NH 2、CN、C 1-C 6烷基、C 2-C 4炔基、C 1-C 6烷氧基、C 3-C 6环烷基、C 3-C 6环烷基-O-或C 3-C 6环烷基-NH-,所述NH 2、C 1-C 6烷基、C 2-C 4炔基、C 1-C 6烷氧基或C 3-C 6环烷基任选被R 10a取代;或者,
    R 10选自卤素、=O、NH 2、C 1-C 6烷基、C 2-C 4炔基、C 1-C 6烷氧基、C 3-C 6环烷基、C 3-C 6环烷基-O-或C 3-C 6环烷基-NH-,所述NH 2、C 1-C 6烷基、C 2-C 4炔基、C 1-C 6烷氧基或C 3-C 6环烷基任选被R 10a取代;或者,
    R 10选自卤素、=O、NH 2、C 1-C 6烷基、C 2-C 4炔基、C 1-C 6烷氧基、C 3-C 6环烷基,所述NH 2、C 1-C 6烷基、C 2-C 4炔基、C 1-C 6烷氧基或C 3-C 6环烷基任选被R 10a取代;或者,
    R 10选自卤素、NH 2、CN、C 1-C 6烷基、C 1-C 6烷氧基或C 3-C 6环烷基,所述NH 2或C 1-C 6烷基任选被R 10a取代;或者,
    R 10选自卤素、任选地被卤素取代的C 1-C 6烷基、C 1-C 6烷氧基或C 3-C 6环烷基。
  6. 根据权利要求1-5中任一项所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,R 10a选自卤素、OH、NH 2、C 1-C 6烷基、卤代C 1-C 6烷基、C 1-C 6烷氧基或卤代C 1-C 6烷氧基;或者,
    R 10a选自卤素、OH、C 1-C 6烷基或卤代C 1-C 6烷基;或者,
    R 10a选自卤素或C 1-C 6烷基;或者,
    R 10a选自F或CH 3
  7. 根据权利要求1-6中任一项所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,Y 1、Y 2和Y 4独立选自CR b或N,Y 3选自CR b;或者,
    Y 1、Y 2独立选自CR b或N,Y 3和Y 4独立选自CR b;或者,
    Y 1、Y 2、Y 3和Y 4均为CR b;或者,
    Y 1和Y 2均为N,Y 3和Y 4独立选自CR b;或者,
    Y 1为N,Y 2、Y 3和Y 4独立选自CR b;或者,
    Y 1、Y 2、Y 3均为CR b、Y 4为N;或者,
    Y 1、Y 2和Y 3均为CH以及Y 4为CR b;或者,
    Y 1为N,Y 2和Y 3均为CH以及Y 4为CR b;或者,
    Y 1、Y 2、Y 3均为CH以及Y 4为N。
  8. 根据权利要求1-7中任一项所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,X为
    Figure PCTCN2022128426-appb-100011
    其中,环B选自任选被R 3取代的5-10元含氮杂芳基、4-7元单环含氮杂环基或6-10元含氮杂环基;或者,
    环B选自任选被R 3取代的以下基团:四氢吡咯基、哌啶基、哌嗪基、吗啉基、
    Figure PCTCN2022128426-appb-100012
    Figure PCTCN2022128426-appb-100013
    Figure PCTCN2022128426-appb-100014
    或者,
    环B选自任选被R 3取代的以下基团:四氢吡咯基、哌啶基、哌嗪基、吗啉基、
    Figure PCTCN2022128426-appb-100015
    Figure PCTCN2022128426-appb-100016
    Figure PCTCN2022128426-appb-100017
    或者,
    环B选自任选被R 3取代的以下基团:四氢吡咯基、哌啶基、哌嗪基、吗啉基、
    Figure PCTCN2022128426-appb-100018
    Figure PCTCN2022128426-appb-100019
    Figure PCTCN2022128426-appb-100020
    或者,
    环B选自任选被R 3取代的以下基团:四氢吡咯基、哌啶基、哌嗪基、吗啉基、
    Figure PCTCN2022128426-appb-100021
    Figure PCTCN2022128426-appb-100022
  9. 根据权利要求1-8中任一项所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,R 3选自卤素、OH、=O、C 1-C 3烷基、C 3-C 6环烷基或苯基,所述C 1-C 3烷基、C 3-C 6环烷基或苯基任选进一步被R 3a取代;或者,R 3选自=O、OH、F、甲基、异丙基、CF 3、环丙基或苯基;或者,R 3选自=O、OH、F、甲基、CF 3、环丙基或苯基。
  10. 根据权利要求1-9中任一项所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,R 4、R 5独立选自H、卤素、OH或任选被R 4a取代的C 1-C 3烷基;或者R 4、R 5及其相连的原子一起形成任选被R 8a取代的C 3-C 6环烷基;或者R 4、R 5一起形成=O;或者,
    R 4、R 5独立选自H、甲基、羟甲基或CF 3,或者R 4、R 5及其相连的原子一起形成环丙基,或者R 4、R 5一起形成=O。
  11. 根据权利要求1-10中任一项所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,
    Figure PCTCN2022128426-appb-100023
    选自如下基团:
    Figure PCTCN2022128426-appb-100024
    Figure PCTCN2022128426-appb-100025
    Figure PCTCN2022128426-appb-100026
    或者,
    Figure PCTCN2022128426-appb-100027
    选自如下基团:
    Figure PCTCN2022128426-appb-100028
    Figure PCTCN2022128426-appb-100029
    或者,
    Figure PCTCN2022128426-appb-100030
    选自如下基团:
    Figure PCTCN2022128426-appb-100031
    Figure PCTCN2022128426-appb-100032
  12. 根据权利要求1-7中任一项所述的式(I)化合物或其立体异构体其药学上可接受的盐,其中X为
    Figure PCTCN2022128426-appb-100033
    其中环D选自任选被R 6取代的以下基团:C 3-C 6环烷基、5-6元杂芳基、4-7元单环含氮杂环基或6-10元含氮杂环基,并且环D以非N原子与L相连;或者,
    环D选自任选被R 6取代的以下基团:环丙基、环丁基、环戊基、四氢吡咯基、哌啶基、哌嗪基、
    Figure PCTCN2022128426-appb-100034
    吡唑基、咪唑基、噁唑基、异噁唑基、三唑基、噻唑基、异噻唑基或吡啶基;或者,
    环D选自任选被R 6取代的以下基团:环丁基、环戊基、哌啶基、吡啶基、
    Figure PCTCN2022128426-appb-100035
    Figure PCTCN2022128426-appb-100036
  13. 根据权利要求1-7或12中任一项所述的式(I)化合物或其立体异构体其药学上可接受的盐,其中R 6选自卤素、OH、CN、=O或任选被R 3a取代的C 1-C 3烷基;或者,R 6选自卤素、=O、OH或C 1-C 3烷基;或者,R 6选自F或甲基。
  14. 根据权利要求1-7或12-13中任一项所述的式(I)化合物或其立体异构体其药学上可接受的盐,其中L选自键、-NR 7-、-NR 7CR 8R 9-、-O-或-CR 8R 9-;或者,
    L选自键、-NR 7-、-NR 7CH 2-、-O-或-CR 8R 9-;或者,
    L选自键、-NH-、-NHCH 2-、-NHCH(CH 3)-、-O-、-C(F) 2-或-CH 2-;或者,
    L选自键、-NH-、-NHCH 2-、-O-、-C(F) 2-或-CH 2-。
  15. 根据权利要求1-7或12-14中任一项所述的式(I)化合物或其立体异构体其药学上可接受的盐,其中
    Figure PCTCN2022128426-appb-100037
    选自如下基团:
    Figure PCTCN2022128426-appb-100038
    Figure PCTCN2022128426-appb-100039
    或者,
    Figure PCTCN2022128426-appb-100040
    选自如下基团:
    Figure PCTCN2022128426-appb-100041
    Figure PCTCN2022128426-appb-100042
    或者,
    Figure PCTCN2022128426-appb-100043
    选自如下基团:
    Figure PCTCN2022128426-appb-100044
    Figure PCTCN2022128426-appb-100045
    或者,
    Figure PCTCN2022128426-appb-100046
    选自
    Figure PCTCN2022128426-appb-100047
    或者,
    Figure PCTCN2022128426-appb-100048
    选自
    Figure PCTCN2022128426-appb-100049
  16. 根据权利要求1-15中任一项所述的式(I)化合物或其立体异构体或其药学上可接受的盐,R 1、R 2与其连接的原子共同形成C 3-C 6环烷基或4-7元杂环基,所述C 3-C 6环烷基或4-7元杂环基任选被R 1b取代;或者,
    R 1、R 2与其连接的原子共同形成任选被R 1b取代的如下基团:环丁基、螺[2,3]己基或氧杂环丁基;或者,
    R 1和R 2独立选自H、卤素、CN、C 1-C 3烷基、C 1-C 3烷氧基或C 3-C 6环烷基,所述C 1-C 3烷基、C 1-C 3烷氧基或C 3-C 6环烷基任选被R 1a取代;或者,
    R 1和R 2独立选自H、C 1-C 3烷基或C 3-C 6环烷基,所述C 1-C 3烷基或C 3-C 6环烷基任选被R 1a取代;或者,
    R 1和R 2独立选自H或C 1-C 3烷基,所述C 1-C 3烷基任选被R 1a取代;或者,
    R 1和R 2独立选自H、F、环丙基、甲基、或
    Figure PCTCN2022128426-appb-100050
    或者
    R 1、R 2与其连接的原子共同形成如下基团:
    Figure PCTCN2022128426-appb-100051
  17. 根据权利要求1-16中任一项所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,W 1选自任选被R 15取代的5-10元杂芳基或6-10元杂环基;或者,
    W 1选自任选被R 15取代的5-10元杂芳基;或者,
    W 1选自任选被R 15取代的5元杂芳基或8元杂环基;或者,
    W 1选自任选被R 15取代的如下基团:吡咯基、噻吩基、呋喃基、吡唑基、咪唑基、噻唑基、异噻唑基、噻二唑基、三氮唑基、噁唑基、异噁唑基、噁二唑基或6,7-二氢-5H-吡咯并[2,1-c][1,2,4]三唑基;或者,
    W 1选自任选被R 15取代的如下基团:吡咯基、噻吩基、呋喃基、吡唑基、咪唑基、噻唑基、异噻唑基、噻二唑基、三氮唑基、噁唑基、异噁唑基或噁二唑基;或者,
    W 1选自任选被R 15取代的如下基团:三氮唑基、噁唑基、异噁唑基、噁二唑基或6,7-二氢-5H-吡咯并[2,1-c][1,2,4]三唑基;或者,
    W 1选自任选被R 15取代的如下基团:三氮唑基、噁唑基、异噁唑基或噁二唑基。
  18. 根据权利要求1-17中任一项所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,R 15选自卤素、OH、NH 2、C 1-C 3烷基或C 3-C 6环烷基,所述C 1-C 3烷基或C 3-C 6环烷基任选被R 15a取代;或者,
    R 15选自OH、C 1-C 3烷基或C 3-C 6环烷基,所述C 1-C 3烷基或C 3-C 6环烷基任选被R 15a取代;或者,
    R 15选自甲基或环丙基,所述甲基或环丙基任选被R 15a取代;或者,
    R 15选自甲基、CHF 2或环丙基。
  19. 根据权利要求1所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,所述式(I)化合物或其立体异构体或其药学上可接受的盐选自式(II)化合物或其立体异构体或其药学上可接受的盐:
    Figure PCTCN2022128426-appb-100052
    其中,Y 1、Y 2、Y 3、Y 4、X、Q、W、R 1和R 2如权利要求1中所定义。
  20. 根据权利要求1所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,所述式(I)化合物或其立体异构体或其药学上可接受的盐选自式(III)化合物或其立体异构体或其药学上可接受的盐:
    Figure PCTCN2022128426-appb-100053
    其中,Z 1、Z 2、Z 3独立地选自CH、CR 10或N;R 10、Y 1、Y 2、Y 3、Y 4、X、W、R 1和R 2如权利要求1中所定义。
  21. 根据权利要求1所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,所述式(I)化合物或其立体异构体或其药学上可接受的盐选自式(V)化合物或其立体异构体或其药学上可接受的盐:
    Figure PCTCN2022128426-appb-100054
    其中,Z 1、Z 2、Z 3独立地选自CH、CR 10或N;A 3选自CR 11aR 11b、NR 12、O或S;R 10、R 11a、R 11b、R 12、Y 1、Y 2、Y 3、Y 4、X、W、R 1和R 2如权利要求1中所定义。
  22. 根据权利要求1所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中,所述式(I)化合物或其立体异构体或其药学上可接受的盐选自式(VI)化合物或其立体异构体或其药学上可接受的盐:
    Figure PCTCN2022128426-appb-100055
    其中,Z 1选自CH、CR 10或N;Z 2选自CR 10、N、O或S;R 10、Y 1、Y 2、Y 3、Y 4、X、W、R 1和R 2如权利要求1中所定义。
  23. 根据权利要求1所述的式(I)化合物或其立体异构体或其药学上可接受的盐,其中所述化合物具有选自如下之一的结构或其药学上可接受的盐:
    Figure PCTCN2022128426-appb-100056
    Figure PCTCN2022128426-appb-100057
    Figure PCTCN2022128426-appb-100058
    Figure PCTCN2022128426-appb-100059
    Figure PCTCN2022128426-appb-100060
    Figure PCTCN2022128426-appb-100061
    Figure PCTCN2022128426-appb-100062
    Figure PCTCN2022128426-appb-100063
    Figure PCTCN2022128426-appb-100064
  24. 一种药物组合物,所述药物组合物包含权利要求1-23任一项所述的化合物或其立体异构体或其药学上可接受的盐,以及药学上可接受的辅料。
  25. 用于预防或治疗由Cbl-b介导的疾病或病症的方法,其包括向有需要的个体施用权利要求1-23任一项所述的化合物或其立体异构体或其药学上可接受的盐或者权利要求24所述的药物组合物,所述由Cbl-b介导的疾病或病症优选肿瘤或自身免疫性疾病。
  26. 用于预防或治疗由Cbl-b介导的疾病或病症的权利要求1-23任一项所述的化合物或其立体异构体或其药学上可接受的盐或者权利要求24所述的药物组合物,所述由Cbl-b介导的疾病或病症优选肿瘤或自身免疫性疾病。
PCT/CN2022/128426 2021-10-29 2022-10-28 作为cbl-b抑制剂的并环化合物 WO2023072273A1 (zh)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
CN202111271704 2021-10-29
CN202111271704.9 2021-10-29
CN202210103242 2022-01-27
CN202210103242.8 2022-01-27
CN202210899553.X 2022-07-28
CN202210899553 2022-07-28
CN202211063507 2022-09-01
CN202211063507.2 2022-09-01

Publications (1)

Publication Number Publication Date
WO2023072273A1 true WO2023072273A1 (zh) 2023-05-04

Family

ID=86159070

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/128426 WO2023072273A1 (zh) 2021-10-29 2022-10-28 作为cbl-b抑制剂的并环化合物

Country Status (1)

Country Link
WO (1) WO2023072273A1 (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024017201A1 (en) * 2022-07-18 2024-01-25 Insilico Medicine Ip Limited Cbl-b inhibitors and methods of uses thereof
WO2024020034A1 (en) * 2022-07-20 2024-01-25 Arcus Biosciences, Inc. Cbl-b inhibitors and methods of use thereof
WO2024105563A1 (en) * 2022-11-16 2024-05-23 Pfizer Inc. Substituted bicyclic pyridone derivatives

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1861590A (zh) * 2006-06-14 2006-11-15 浙江大学 类黄酮衍生物及其制备方法和用途
WO2009053799A1 (en) * 2007-10-24 2009-04-30 Glenmark Pharmaceuticals, S.A. Novel cannabinoid receptor ligands, pharmaceutical compositions containing them, and process for their preparation
CN101591370A (zh) * 2008-05-27 2009-12-02 中国农业科学院兰州畜牧与兽药研究所 一类葛根素衍生物的合成方法
WO2019148005A1 (en) * 2018-01-26 2019-08-01 Nurix Therapeutics, Inc. Inhibitors of cbl-b and methods of use thereof
WO2020210508A1 (en) * 2019-04-09 2020-10-15 Nurix Therapeutics, Inc. 3-substituted piperidine compounds for cbl-b inhibition, and use of a cbl-b inhibitor in combination with a cancer vaccine and/or oncolytic virus
WO2020264398A1 (en) * 2019-06-26 2020-12-30 Nurix Therapeutics, Inc. Substituted benzyl-triazole compounds for cbl-b inhibition, and further uses thereof
WO2022169997A1 (en) * 2021-02-03 2022-08-11 Genentech, Inc. Lactams as cbl-b inhibitors
WO2022217276A1 (en) * 2021-04-09 2022-10-13 Nimbus Clio, Inc. Cbl-b modulators and uses thereof
WO2022221704A1 (en) * 2021-04-16 2022-10-20 Hotspot Therapeutics, Inc. Compounds, compositions and methods of treating cancer

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1861590A (zh) * 2006-06-14 2006-11-15 浙江大学 类黄酮衍生物及其制备方法和用途
WO2009053799A1 (en) * 2007-10-24 2009-04-30 Glenmark Pharmaceuticals, S.A. Novel cannabinoid receptor ligands, pharmaceutical compositions containing them, and process for their preparation
CN101591370A (zh) * 2008-05-27 2009-12-02 中国农业科学院兰州畜牧与兽药研究所 一类葛根素衍生物的合成方法
WO2019148005A1 (en) * 2018-01-26 2019-08-01 Nurix Therapeutics, Inc. Inhibitors of cbl-b and methods of use thereof
WO2020210508A1 (en) * 2019-04-09 2020-10-15 Nurix Therapeutics, Inc. 3-substituted piperidine compounds for cbl-b inhibition, and use of a cbl-b inhibitor in combination with a cancer vaccine and/or oncolytic virus
WO2020264398A1 (en) * 2019-06-26 2020-12-30 Nurix Therapeutics, Inc. Substituted benzyl-triazole compounds for cbl-b inhibition, and further uses thereof
WO2022169997A1 (en) * 2021-02-03 2022-08-11 Genentech, Inc. Lactams as cbl-b inhibitors
WO2022217276A1 (en) * 2021-04-09 2022-10-13 Nimbus Clio, Inc. Cbl-b modulators and uses thereof
WO2022221704A1 (en) * 2021-04-16 2022-10-20 Hotspot Therapeutics, Inc. Compounds, compositions and methods of treating cancer

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
JI, QING'E ET AL.: "Synthesis of Isoflavone Compounds as Cardiovascular System Drug", ACTA PHARMACEUTICA SINICA, vol. 24, no. 12, 31 December 1989 (1989-12-31), XP009545903, ISSN: 0513-4870 *
YUAN JIN-WEI, YANG LIANG-RU, MAI WEN-PENG, MAO PU, WANG FAN, XUE YING, XIAO YONG-MEI, QU LING-BO: "Ultrasound-assisted regioselective synthesis of aminomethylated daidzein derivatives via Mannich reaction", ZEITSCHRIFT FUR NATURFORSCHUNG - SECTION B JOURNAL OF CHEMICAL SCIENCES, vol. 70, no. 10, 1 October 2015 (2015-10-01), DE , pages 727 - 734, XP093061977, ISSN: 0932-0776, DOI: 10.1515/znb-2015-0068 *
ZHU, HUIQING ET AL.: "Initial Research on Synthesis of 7-Methoxy-4/-Hydroxy-3/-Substituted Aminomethyl Isoflavones and Biological Activities Thereof", CHINESE JOURNAL OF PHARMACEUTICALS, vol. 21, no. 9, 31 December 1990 (1990-12-31), XP009545904, ISSN: 1001-8255 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024017201A1 (en) * 2022-07-18 2024-01-25 Insilico Medicine Ip Limited Cbl-b inhibitors and methods of uses thereof
WO2024020034A1 (en) * 2022-07-20 2024-01-25 Arcus Biosciences, Inc. Cbl-b inhibitors and methods of use thereof
WO2024105563A1 (en) * 2022-11-16 2024-05-23 Pfizer Inc. Substituted bicyclic pyridone derivatives

Similar Documents

Publication Publication Date Title
WO2023072273A1 (zh) 作为cbl-b抑制剂的并环化合物
CN107873031B (zh) 作为tnf活性调节剂的苯并噁嗪酮衍生物及其类似物
CN107253963B (zh) 吡啶酮和氮杂吡啶酮化合物及使用方法
CN111433200A (zh) 用于治疗疾病的新化合物及其药物组合物
TWI704149B (zh) 抑制瞬態電位受器a1離子通道
CN107820494B (zh) 核受体调节剂
CN115850268A (zh) 作为cftr增效剂的吡咯并嘧啶
TW201512173A (zh) 炔基醇及其使用方法
CN114728962A (zh) 血浆激肽释放酶抑制剂及其用途
MX2012011132A (es) Compuestos antagonistas selectivos de receptor nk-3 novedosos, composicion farmaceutica y metodos para utilizarse en trastornos transmitidos por receptores nk-3.
WO2021143680A1 (zh) 杂芳基类衍生物及其制备方法和用途
TW201607948A (zh) 5-氯-2-二氟甲氧基苯基吡唑并嘧啶化合物、組合物及其使用方法
WO2020108613A1 (zh) 杂芳类衍生物调节剂、其制备方法和应用
CN102459272A (zh) 对P110δ具有选择性的为PI3K抑制剂的二环嘧啶化合物和使用方法
WO2016169421A1 (zh) 咪唑并异吲哚类衍生物、其制备方法及其在医药上的应用
TW201722958A (zh) 化學化合物
CN111499634A (zh) 一种喹唑啉化合物及其在医药上的应用
WO2022170952A1 (zh) 一种作为sos1抑制剂的多环哒嗪酮类衍生物、其制备方法及用途
WO2023036330A1 (zh) 作为Cbl-b抑制剂的三环类化合物
WO2023198199A1 (zh) 膜缔合酪氨酸和苏氨酸激酶抑制剂
CN112513021A (zh) RORγ拮抗剂及其在药物中的应用
CN114466850A (zh) [1,2,4]三唑并[1,5-c]喹唑啉-5-胺
TW201806949A (zh) 經取代5,6,7,8-四氫[1,2,4]三唑并[4,3-a]吡啶-3(2h)-酮及2,5,6,7-四氫-3h-吡咯并[2,1-c][1,2,4]三唑-3-酮及其用途
WO2023202623A1 (zh) Polq抑制剂化合物及其应用
WO2023131277A1 (zh) Nlrp3炎症小体抑制剂及其应用

Legal Events

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

Ref document number: 22886148

Country of ref document: EP

Kind code of ref document: A1