WO2022258023A1 - 用作cdk激酶抑制剂的化合物及其应用 - Google Patents

用作cdk激酶抑制剂的化合物及其应用 Download PDF

Info

Publication number
WO2022258023A1
WO2022258023A1 PCT/CN2022/097927 CN2022097927W WO2022258023A1 WO 2022258023 A1 WO2022258023 A1 WO 2022258023A1 CN 2022097927 W CN2022097927 W CN 2022097927W WO 2022258023 A1 WO2022258023 A1 WO 2022258023A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
substituted
alkoxy
halogenated
Prior art date
Application number
PCT/CN2022/097927
Other languages
English (en)
French (fr)
Inventor
李钧
牛成山
陈明涛
梁阿朋
李美华
董胜利
吴豫生
Original Assignee
郑州同源康医药有限公司
浙江同源康医药股份有限公司
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 郑州同源康医药有限公司, 浙江同源康医药股份有限公司 filed Critical 郑州同源康医药有限公司
Priority to EP22819620.0A priority Critical patent/EP4353724A1/en
Priority to KR1020247000908A priority patent/KR20240021239A/ko
Priority to AU2022289202A priority patent/AU2022289202A1/en
Priority to CA3221997A priority patent/CA3221997A1/en
Priority to CN202280006672.6A priority patent/CN116456987A/zh
Publication of WO2022258023A1 publication Critical patent/WO2022258023A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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

  • the invention relates to the field of medical technology, in particular to a compound used as a CDK kinase inhibitor and its application in regulating CDK kinase activity or treating CDK-related diseases.
  • the cell cycle refers to the whole process that cells go through from the completion of one division to the end of the next division, which is divided into two stages: interphase and division phase.
  • the interphase is further divided into three phases, the early DNA synthesis phase (G1 phase), the DNA synthesis phase (S phase) and the late DNA synthesis phase (G2 phase); the division phase is the M phase.
  • Cyclins and cyclin-dependent kinases (CDKs) are the core molecules in the entire cell cycle regulation mechanism.
  • cyclin B/CDK1 CyclinB/CDK1
  • cyclin A/CDK2 CyclinA/CDK2
  • cyclin E/CDK2 CyclinE/CDK2
  • cell Cyclin D/CDK4 CyclinD/CDK4
  • Cyclin D/CDK6 Cyclin D/CDK6
  • Other functions of cyclin/CDK heterodynes include transcriptional regulation, DNA repair, differentiation and apoptosis.
  • CDKs cyclin-dependent kinases
  • the CDK4/6 inhibitors palbociclib, ribociclib, and abemaciclib target breast and other cancers; however, like other kinase inhibitors, their effects may Limited by the development of primary or acquired resistance over time. Therefore, PF-06873600 developed by Pfizer, as a new multi-target CDK kinase inhibitor, is expected to solve the phenomenon of drug resistance under existing conditions. PF-06873600 has also entered phase I clinical trials for the treatment of metastatic breast cancer.
  • Cyclin-dependent kinase (CDKs) inhibitors are the most promising field of cancer treatment, and the development of new compounds with CDK kinase inhibitory activity and better pharmacodynamics and pharmacokinetic properties has become an important factor in the development of new anti-tumor drugs. It is an important research project of drugs, and will eventually be used in the treatment of diseases such as human tumors.
  • the object of the present invention is to provide a compound represented by formula (I), its preparation method and its use in CDK2/4/6 kinase inhibitor drugs.
  • the first aspect of the present invention provides a compound of formula (I), or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof,
  • R 1 is selected from the following group: unsubstituted or 1 to 4 R 6 substituted C3-C10 cycloalkyl, unsubstituted or 1 to 4 R 6 substituted without or containing 1-3 selected from N, O, A 5-15-membered ring with a heteroatom of S, unsubstituted or 1 to 4 R 6 substituted 5-15-membered spiro rings that do not contain or contain 1-3 heteroatoms selected from N, O, and S, unsubstituted Substituted or 1 to 4 R 6 substituted 5-15-membered bridged rings that do not contain or contain 1-3 heteroatoms selected from N, O, and S, unsubstituted or 1 to 4 R 6 substituted 1- A 3-10 membered heterocycloalkyl group with 4 heteroatoms selected from N, O, S, unsubstituted or 1 to 4 R 6 substituted C6-C10 aryl, unsubstituted or 1 to 4 R 6 substituted A 3-10 membere
  • R 2 is selected from the group consisting of H, F, OH, C1-C4 alkyl, halogenated C1-C4 alkyl, C1-C4 alkoxy, halogenated C1-C4 alkoxy, and R 2 can be combined with The atoms are connected to form a spiro ring, a bridge ring, or a ring structure;
  • R 3a and R 3b are independently selected from the group consisting of H, F, OH, C1-C4 alkyl, halogenated C1-C4 alkyl, C1-C4 alkoxy, halogenated C1-C4 alkoxy;
  • each of the C1-C4 alkyl and halogenated C1-C4 alkyl in R 2 , R 3a and R 3b is optionally replaced by halogen, OH, C1-C4 alkoxy or halogenated C1-C4 alkoxy replace;
  • R is selected from the group consisting of H, halogen, C1 - C2 alkyl, halogenated C1-C2 alkyl, C1-C4 alkoxy, halogenated C1-C4 alkoxy;
  • Each R 7 is independently selected from the group consisting of C1-C4 alkyl, -L-(C3-C8 cycloalkyl), -L-(3- containing 1-4 heteroatoms selected from N, O, S 10-membered heterocycloalkyl), -L-(3-10-membered heteroaryl containing 1-5 heteroatoms selected from N, O, S), -L-(3-10-membered aryl), wherein
  • the C1-C4 alkyl, C3-C8 cycloalkyl, 3-10 membered heterocycloalkyl, 3-10 membered heteroaryl, 3-10 membered aryl are optionally replaced by 0 to 4 D, OH , Halogen, CN, C1-C4 Alkyl, Halogenated C1-C4 Alkyl, N(C1-C4 Alkyl) 2 , NHCO(C1-C4 Alkyl), SO 2 (C1-C4 Alkyl), CO 2 (C
  • R 8 and R 9 are independently selected from the following group: H, C1-C4 alkyl, halogenated C1-C4 alkyl, C3-C8 cycloalkyl, -L-(C3-C8 cycloalkyl), -L- (3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S), -L- (3-10 heterocycloalkyl containing 1-5 heteroatoms selected from N, O, S membered heteroaryl); or
  • Each R' and R" is independently selected from the group consisting of H, C1-C4 alkyl
  • Each L is independently a bond or a C1-C4 alkylene, the C1-C4 alkylene is optionally substituted by OH, C1-C4 alkoxy or halogenated C1-C4 alkoxy;
  • p is selected from the group consisting of 0, 1, 2, 3, 4;
  • r is selected from the group: 0, 1, 2, 3, 4.
  • R 1 is selected from the following group: C3-C10 cycloalkyl unsubstituted or substituted by 1 to 4 R 6 , unsubstituted or substituted by 1 to 4 R 6 without or containing 1-3 5-15 membered rings selected from N, O, S heteroatoms, unsubstituted or 1 to 4 R 6 substituted 5 containing or containing 1-3 heteroatoms selected from N, O, S -15 membered spiro ring, unsubstituted or 1 to 4 R 6 substituted 5-15 membered bridged rings without or containing 1 to 3 heteroatoms selected from N, O, S, unsubstituted or 1 to 4 R 6 substituted 3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S, unsubstituted or 1 to 4 R 6 substituted C6-C10 aryl, unsubstituted or 1 ⁇ 4 R 6 substituted 3-10 membered heteroaryl groups containing 1-5
  • R is selected from the group consisting of H, F ;
  • R 3a and R 3b are independently selected from the group consisting of H, F, OH, C1-C4 alkyl;
  • R 4 is selected from the following group: substituted or unsubstituted 3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S, -OR 4 ', -NR 4 'R 4 ", Wherein, R 4 ' and R 4 " are independently selected from the following group: H, substituted or unsubstituted C1-C6 alkyl, and the substitution refers to being substituted by 1-3 substituents selected from the group: deuterium, halogen , C1-C4 alkyl;
  • R is selected from the group consisting of H, C1 - C2 alkyl
  • Each R 7 is independently selected from the group consisting of C1-C4 alkyl, -L-(C3-C8 cycloalkyl), -L-(3- containing 1-4 heteroatoms selected from N, O, S 10-membered heterocycloalkyl), -L-(3-10-membered heteroaryl containing 1-5 heteroatoms selected from N, O, S), -L-(3-10-membered aryl), wherein
  • the C1-C4 alkyl, C3-C8 cycloalkyl, 3-10 membered heterocycloalkyl, 3-10 membered heteroaryl, 3-10 membered aryl are optionally replaced by 0 to 4 D, OH , Halogen, CN, C1-C4 Alkyl, Halogenated C1-C4 Alkyl, N(C1-C4 Alkyl) 2 , NHCO(C1-C4 Alkyl), SO 2 (C1-C4 Alkyl), CO 2 (C
  • R 8 and R 9 are independently selected from the following group: H, C1-C4 alkyl, halogenated C1-C4 alkyl, C3-C8 cycloalkyl, -L-(C3-C8 cycloalkyl), -L- (3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S), -L- (3-10 heterocycloalkyl containing 1-5 heteroatoms selected from N, O, S membered heteroaryl); or
  • Each R' and R" is independently selected from the group consisting of H, C1-C4 alkyl
  • Each L is independently a bond or a C1-C4 alkylene, the C1-C4 alkylene is optionally substituted by OH, C1-C4 alkoxy or halogenated C1-C4 alkoxy;
  • p is selected from the group consisting of 0, 1, 2, 3, 4;
  • r is selected from the group: 0, 1, 2, 3, 4.
  • R 1 is selected from the following group: unsubstituted or 1-4 R 6 substituted C3-C10 cycloalkyl, unsubstituted or 1-4 R 6 substituted containing 1-4 selected from 3-10 membered heterocycloalkyl with N, O, S heteroatoms, C6-C10 aryl unsubstituted or 1 to 4 R 6 substituted, unsubstituted or 1 to 4 R 6 substituted containing 1-5 A 3-10 membered heteroaryl group selected from N, O, S heteroatoms;
  • R is selected from the group consisting of H, F ;
  • R 3a and R 3b are independently selected from the group consisting of H, F, OH, C1-C4 alkyl;
  • R 4 is selected from the following group: substituted or unsubstituted 3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S, -OR 4 ', wherein R 4 ' is independently selected from From the following group: H, substituted or unsubstituted C1-C6 alkyl, said substitution refers to being substituted by 1-3 substituents selected from the following group: deuterium, halogen, C1-C4 alkyl;
  • R is selected from the group consisting of H, C1 - C2 alkyl
  • Each R 7 is independently selected from the group consisting of C1-C4 alkyl, -L-(C3-C8 cycloalkyl), -L-(3- containing 1-4 heteroatoms selected from N, O, S 10-membered heterocycloalkyl), -L-(3-10-membered heteroaryl containing 1-5 heteroatoms selected from N, O, S), -L-(3-10-membered aryl), wherein
  • the C1-C4 alkyl, C3-C8 cycloalkyl, 3-10 membered heterocycloalkyl, 3-10 membered heteroaryl, 3-10 membered aryl are optionally replaced by 0 to 4 D, OH , Halogen, CN, C1-C4 Alkyl, Halogenated C1-C4 Alkyl, N(C1-C4 Alkyl) 2 , NHCO(C1-C4 Alkyl), SO 2 (C1-C4 Alkyl), CO 2 (C
  • R 8 and R 9 are independently selected from the following group: H, C1-C4 alkyl, halogenated C1-C4 alkyl, C3-C8 cycloalkyl, -L-(C3-C8 cycloalkyl), -L- (3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S), -L- (3-10 heterocycloalkyl containing 1-5 heteroatoms selected from N, O, S membered heteroaryl); or
  • Each R' and R" is independently selected from the group consisting of H, C1-C4 alkyl
  • Each L is independently a bond or a C1-C4 alkylene, the C1-C4 alkylene is optionally substituted by OH, C1-C4 alkoxy or halogenated C1-C4 alkoxy;
  • p is selected from the group consisting of 0, 1, 2, 3, 4;
  • r is selected from the group: 0, 1, 2, 3, 4.
  • R 1 is selected from the following group: C3-C10 cycloalkyl unsubstituted or substituted by 1 to 4 R 6 , unsubstituted or substituted by 1 to 4 R 6 without or containing 1-3 5-15 membered rings selected from N, O, S heteroatoms, unsubstituted or 1 to 4 R 6 substituted 5 containing or containing 1-3 heteroatoms selected from N, O, S -15 membered spiro ring, unsubstituted or 1 to 4 R 6 substituted 5-15 membered bridged rings without or containing 1 to 3 heteroatoms selected from N, O, S, unsubstituted or 1 to 4 R 6 substituted 3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S, unsubstituted or 1 to 4 R 6 substituted C6-C10 aryl, unsubstituted or 1 ⁇ 4 R 6 substituted 3-10 membered heteroaryl groups containing 1-5
  • R 2 is selected from the group consisting of H, F, OH, C1-C4 alkyl, halogenated C1-C4 alkyl, C1-C4 alkoxy, halogenated C1-C4 alkoxy, and R 2 can be combined with The atoms are connected to form a spiro ring, a bridge ring, or a ring structure;
  • R 3a and R 3b are independently selected from the group consisting of H, F, OH, C1-C4 alkyl, halogenated C1-C4 alkyl, C1-C4 alkoxy, halogenated C1-C4 alkoxy;
  • each of the C1-C4 alkyl and halogenated C1-C4 alkyl in R 2 , R 3a and R 3b is optionally replaced by halogen, OH, C1-C4 alkoxy or halogenated C1-C4 alkoxy replace;
  • R is selected from the group consisting of H, halogen, C1 - C2 alkyl, halogenated C1-C2 alkyl, C1-C4 alkoxy, halogenated C1-C4 alkoxy;
  • Each R 7 is independently selected from the group consisting of C1-C4 alkyl, -L-(C3-C8 cycloalkyl), -L-(3- containing 1-4 heteroatoms selected from N, O, S 10-membered heterocycloalkyl), -L-(3-10-membered heteroaryl containing 1-5 heteroatoms selected from N, O, S), -L-(3-10-membered aryl), wherein
  • the C1-C4 alkyl, C3-C8 cycloalkyl, 3-10 membered heterocycloalkyl, 3-10 membered heteroaryl, 3-10 membered aryl are optionally replaced by 0 to 4 D, OH , halogen, CN, C1-C4 alkyl, halogenated C1-C4 alkyl, N(C1-C4 alkyl) 2 , NHCO(C1-C4 alkyl), SO2(C1-C4 alkyl), containing
  • R 8 and R 9 are independently selected from the following group: H, C1-C4 alkyl, halogenated C1-C4 alkyl, C3-C8 cycloalkyl, -L-(C3-C8 cycloalkyl), -L- (3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S), -L- (3-10 heterocycloalkyl containing 1-5 heteroatoms selected from N, O, S membered heteroaryl); or
  • Each R' and R" is independently selected from the group consisting of H, C1-C4 alkyl
  • Each L is independently a bond or a C1-C4 alkylene, the C1-C4 alkylene is optionally substituted by OH, C1-C4 alkoxy or halogenated C1-C4 alkoxy;
  • p is selected from the group consisting of 0, 1, 2, 3, 4;
  • r is selected from the group: 0, 1, 2, 3, 4.
  • the compound has the structure shown in formula (II) or formula (III):
  • the A ring is selected from the group consisting of 6-10 membered aryls substituted with 0-4 R 1 's, C3-C8 cycloalkyls substituted with 0-4 R 1 's, 0-4 R 1 's substituted A 5-10 membered heteroaryl group containing 1-5 heteroatoms selected from N, O, S;
  • R 2 ' is selected from the group consisting of H, deuterium
  • R 4 ' is selected from the group consisting of H, deuterium, OH;
  • R 41 and R 51 are independently selected from the group consisting of H, C1-C4 alkyl, halogenated C1-C4 alkyl, C3-C8 cycloalkyl, -L'-(C3-C8 cycloalkyl), -L '-(3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S), -L'-(containing 1-5 heteroatoms selected from N, O, S 3-10 membered heteroaryl), L' is a bond or a C1-C6 alkylene group; or
  • n is selected from the group: 0, 1, 2, 3, 4.
  • the compound has the structure shown in formula (II):
  • the A ring is selected from the group consisting of 6-10 membered aryls substituted with 0-4 R 1 's, C3-C8 cycloalkyls substituted with 0-4 R 1 's, 0-4 R 1 's substituted A 5-10 membered heteroaryl group containing 1-5 heteroatoms selected from N, O, S;
  • R 2 ' is selected from the group consisting of H, deuterium
  • R 41 and R 51 are independently selected from the group consisting of H, C1-C4 alkyl, halogenated C1-C4 alkyl, C3-C8 cycloalkyl, -L'-(C3-C8 cycloalkyl), -L '-(3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S), -L'-(containing 1-5 heteroatoms selected from N, O, S 3-10 membered heteroaryl), L' is a bond or a C1-C6 alkylene group; or
  • n is selected from the group: 0, 1, 2, 3, 4.
  • ring A is a 6-10 membered aryl group substituted by 0-4 R 1 's.
  • R 3 ' is C1-C4 alkyl.
  • ring A is phenyl
  • R 1 ' is selected from the group consisting of H, halogen, CN, C1-C4 alkoxy.
  • R 1 ' is F, OMe.
  • ring A is phenyl
  • n is 1
  • R 1 ' is selected from the following group: H, halogen, CN, C1-C4 alkoxy.
  • R 1 ' is F.
  • R 1 ' is F.
  • ring A is a C3-C8 cycloalkyl group substituted by 0-4 R 1 's.
  • the compound has the structure shown in formula (IV) or formula (V):
  • q is selected from the group consisting of 1, 2, 3, 4, 5, and 6.
  • ring A is a 5-10 membered heteroaryl group containing 0-4 R 1 ' substituted and containing 1-5 heteroatoms selected from N, O, and S.
  • ring A is selected from the following group: 6-10 membered aryl group with 0-4 R 1 ' substitution, C3-C8 cycloalkyl group with 0-4 R 1 ' substitution, 0 -4 R 1 'substituted 5-10 membered heteroaryl containing 1-5 heteroatoms selected from N, O, S;
  • Each R 1 ' is independently selected from the group consisting of deuterium, halogen, OH, CN, SO2R 31 , COR 31 , CO2R 31 , NR 41 R 51 , NHCOR 41 , CONR 41 R 51 , halogenated C1-C4 alkyl, C1 -C4 alkoxy, halogenated C1-C4 alkoxy, substituted or unsubstituted C3-C6 cycloalkyl, substituted or unsubstituted 3- containing 1-4 heteroatoms selected from N, O, S 6-membered heterocycloalkyl;
  • R 2 ' is selected from the group consisting of H, deuterium
  • R 4 ' is selected from the group consisting of H, deuterium
  • R 31 is C1-C4 alkyl
  • R 41 and R 51 are independently selected from the following group: H, C1-C4 alkyl;
  • n is selected from the group: 0, 1, 2, 3, 4.
  • ring A is selected from the following group: 6-10 membered aryl group with 0-4 R 1 ' substitution, C3-C8 cycloalkyl group with 0-4 R 1 ' substitution, 0 -4 R 1 'substituted 5-10 membered heteroaryl containing 1-5 heteroatoms selected from N, O, S;
  • Each R 1 ' is independently selected from the group consisting of deuterium, halogen, OH, CN, SO2R 31 , COR 31 , CO2R 31 , NR 41 R 51 , NHCOR 41 , CONR 41 R 51 , halogenated C1-C4 alkyl, C1 -C4 alkoxy, halogenated C1-C4 alkoxy, substituted or unsubstituted C3-C6 cycloalkyl, substituted or unsubstituted 3- containing 1-4 heteroatoms selected from N, O, S 6-membered heterocycloalkyl;
  • R 2 ' is selected from the group consisting of H, deuterium
  • R 31 is C1-C4 alkyl
  • R 41 and R 51 are independently selected from the following group: H, C1-C4 alkyl;
  • n is selected from the group: 0, 1, 2, 3, 4.
  • ring A is selected from the following group: 6-10 membered aryl group with 0-4 R 1 ' substitutions, C3-C8 cycloalkyl group with 0-4 R 1 'substitutions;
  • R 1 ' is selected from the group consisting of halogen, OH, C1-C4 alkoxy, halogenated C1-C4 alkoxy;
  • R 2 ' is selected from the group consisting of H, deuterium
  • n is selected from the group: 0, 1, 2, 3, 4.
  • the compound is selected from the following group:
  • the compound has the structure shown in formula (III):
  • R 2 ' is selected from the group consisting of H, deuterium
  • R 4 ' is selected from the group consisting of H, deuterium, OH;
  • R 2 ' is: H
  • R 3 ' is selected from the following group: substituted or unsubstituted C1-C4 alkyl, said substitution means being substituted by 1-3 substituents selected from the group: halogen, CN, C1-C4 alkoxy;
  • the compound is selected from the following group:
  • R 1 is selected from the following group: unsubstituted or 1 to 4 R 6 substituted C3-C10 cycloalkyl, unsubstituted or 1 to 4 R 6 substituted without or containing 1-3 selected from N, O, A 5-15-membered ring with a heteroatom of S, unsubstituted or 1 to 4 R 6 substituted 5-15-membered spiro rings that do not contain or contain 1-3 heteroatoms selected from N, O, and S, unsubstituted Substituted or 1 to 4 R 6 substituted 5-15-membered bridged rings that do not contain or contain 1-3 heteroatoms selected from N, O, and S, unsubstituted or 1 to 4 R 6 substituted 1- A 3-10 membered heterocycloalkyl group with 4 heteroatoms selected from N, O, S, unsubstituted or 1 to 4 R 6 substituted C6-C10 aryl, unsubstituted or 1 to 4 R 6 substituted A 3-10-member
  • R 2 is selected from the group consisting of H, F, OH, C1-C4 alkyl, halogenated C1-C4 alkyl, C1-C4 alkoxy, halogenated C1-C4 alkoxy, and R 2 can be combined with The atoms are connected to form a spiro ring, a bridge ring, or a ring structure;
  • R 3a and R 3b are independently selected from the group consisting of H, F, OH, C1-C4 alkyl, halogenated C1-C4 alkyl, C1-C4 alkoxy, halogenated C1-C4 alkoxy;
  • each of the C1-C4 alkyl and halogenated C1-C4 alkyl in R 2 , R 3a and R 3b is optionally replaced by halogen, OH, C1-C4 alkoxy or halogenated C1-C4 alkoxy replace;
  • R is selected from the group consisting of H, halogen, C1 - C2 alkyl, halogenated C1-C2 alkyl, C1-C4 alkoxy, halogenated C1-C4 alkoxy;
  • Each R 7 is independently selected from the group consisting of C1-C4 alkyl, -L-(C3-C8 cycloalkyl), -L-(3- containing 1-4 heteroatoms selected from N, O, S 10-membered heterocycloalkyl), -L-(3-10-membered heteroaryl containing 1-5 heteroatoms selected from N, O, S), -L-(3-10-membered aryl), wherein
  • the C1-C4 alkyl, C3-C8 cycloalkyl, 3-10 membered heterocycloalkyl, 3-10 membered heteroaryl, 3-10 membered aryl are optionally replaced by 0 to 4 OH, halogen , C1-C4 alkyl, C1-C4 alkoxy or halogenated C1-C4 alkoxy;
  • R 8 and R 9 are independently selected from the following group: H, C1-C4 alkyl, halogenated C1-C4 alkyl, C3-C8 cycloalkyl, -L-(C3-C8 cycloalkyl), -L- (3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S), -L- (3-10 heterocycloalkyl containing 1-5 heteroatoms selected from N, O, S membered heteroaryl); or
  • Each R' and R" is independently selected from the group consisting of H, C1-C4 alkyl
  • Each L is independently a bond or a C1-C4 alkylene, the C1-C4 alkylene is optionally substituted by OH, C1-C4 alkoxy or halogenated C1-C4 alkoxy;
  • p is selected from the group consisting of 0, 1, 2, 3, 4;
  • r is selected from the group: 0, 1, 2, 3, 4.
  • R 1 is selected from the following group: C3-C10 cycloalkyl unsubstituted or substituted by 1 to 4 R 6 , unsubstituted or substituted by 1 to 4 R 6 without or containing 1-3 5-15 membered rings selected from N, O, S heteroatoms, unsubstituted or 1 to 4 R 6 substituted 5 containing or containing 1-3 heteroatoms selected from N, O, S -15 membered spiro ring, unsubstituted or 1 to 4 R 6 substituted 5-15 membered bridged rings without or containing 1 to 3 heteroatoms selected from N, O, S, unsubstituted or 1 to 4 C6 -C10 aryl group substituted by R6, unsubstituted or 1-4 R6 - substituted 3-10 membered heteroaryl group containing 1-5 heteroatoms selected from N, O, S;
  • R is selected from the group consisting of H, F ;
  • R 3a and R 3b are independently selected from the group consisting of H, F, OH, C1-C4 alkyl;
  • each of the C1-C4 alkyl and halogenated C1-C4 alkyl in R 2 , R 3a and R 3b is optionally replaced by halogen, OH, C1-C4 alkoxy or halogenated C1-C4 alkoxy replace;
  • R 4 is selected from the following group: substituted or unsubstituted 3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S, -NR 4 'R 4 ", wherein, R 4 ' and R 4 "are independently selected from the following group: H, substituted or unsubstituted C1-C6 alkyl, said substitution means being substituted by 1-3 substituents selected from the group: deuterium, halogen, C1-C4 alkyl;
  • R is selected from the group consisting of H, C1 - C2 alkyl
  • Each R 7 is independently selected from the group consisting of C1-C4 alkyl, -L-(C3-C8 cycloalkyl), -L-(3- containing 1-4 heteroatoms selected from N, O, S 10-membered heterocycloalkyl), -L-(3-10-membered heteroaryl containing 1-5 heteroatoms selected from N, O, S), -L-(3-10-membered aryl), wherein
  • the C1-C4 alkyl, C3-C8 cycloalkyl, 3-10 membered heterocycloalkyl, 3-10 membered heteroaryl, 3-10 membered aryl are optionally replaced by 0 to 4 OH, halogen , C1-C4 alkyl, C1-C4 alkoxy or halogenated C1-C4 alkoxy;
  • R 8 and R 9 are independently selected from the following group: H, C1-C4 alkyl, halogenated C1-C4 alkyl, C3-C8 cycloalkyl, -L-(C3-C8 cycloalkyl), -L- (3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S), -L- (3-10 heterocycloalkyl containing 1-5 heteroatoms selected from N, O, S membered heteroaryl); or
  • Each R' and R" is independently selected from the group consisting of H, C1-C4 alkyl
  • Each L is independently a bond or a C1-C4 alkylene, the C1-C4 alkylene is optionally substituted by OH, C1-C4 alkoxy or halogenated C1-C4 alkoxy;
  • p is selected from the group consisting of 0, 1, 2, 3, 4;
  • r is selected from the group: 0, 1, 2, 3, 4.
  • R 1 is selected from the group consisting of:
  • R 4 is selected from the following group: Br, substituted or unsubstituted 3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S, substituted or unsubstituted C1-C4 alkoxy , substituted or unsubstituted C1-C4 alkylthio, substituted or unsubstituted C1-C4 alkylamino;
  • R 6 is as defined in claim 1 .
  • R is selected from the following group:
  • R is selected from the group consisting of substituted or unsubstituted 3-10 membered heterocycloalkyl containing 1-4 heteroatoms selected from N, O, S, substituted or unsubstituted C1-C4 alkoxy, substituted Or unsubstituted C1-C4 alkylamino;
  • R 6 is as defined in claim 1 .
  • R is selected from the group consisting of C1 - C4 alkyl, halogenated C1-C4 alkyl;
  • R 8 and R 9 are independently selected from the group consisting of H, C1-C4 alkyl, halogenated C1-C4 alkyl, C3-C8 cycloalkyl; or
  • R' and R" are independently selected from the group consisting of H, C1-C4 alkyl.
  • p is 0, and r is 2.
  • R 4 is selected from the following group: Br,
  • the compound is selected from the following group:
  • the pharmaceutically acceptable salt is an inorganic acid salt or an organic acid salt
  • the inorganic acid salt is selected from the group consisting of hydrochloride, hydrobromide, hydroiodide, sulfate, bisulfate, nitrate, phosphate, acid phosphate;
  • the organic acid salt is selected from the group consisting of formate, acetate, trifluoroacetate, propionate, pyruvate, glycolate, oxalate, malonate, fumarate Salt, maleate, lactate, malate, citrate, tartrate, methanesulfonate, ethanesulfonate, benzenesulfonate, salicylate, picrate, glutamate , Ascorbate, Camphorate, Camphorsulfonate, Camphorsulfonate.
  • the second aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the compound described in the first aspect of the present invention, or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof, and a pharmaceutically acceptable carrier.
  • the third aspect of the present invention provides a use of the compound described in the first aspect of the present invention, or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof, for the preparation of CDK2/4/ 6 kinase inhibitor drugs.
  • the fourth aspect of the present invention provides a use of the compound described in the first aspect of the present invention, or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof, for the preparation of CDK-regulating Kinase activity or drugs to treat CDK-associated diseases.
  • the CDK-related disease is cancer or tumor.
  • the cancer or tumor is selected from the group consisting of breast cancer, ovarian cancer, bladder cancer, uterine cancer, lung cancer, colorectal cancer, prostate cancer, pancreatic cancer, gastric cancer, thyroid cancer, esophagus cancer, kidney cancer Carcinoma, liver cancer, head and neck cancer glioblastoma, mantle cell lymphoma (MCL), chronic myeloid leukemia (CML) and acute myeloid leukemia (AML).
  • MCL mantle cell lymphoma
  • CML chronic myeloid leukemia
  • AML acute myeloid leukemia
  • the lung cancer is non-small cell lung cancer.
  • Figure 1 is the experimental results of the xMCF-7_Palbo-R xenograft tumor model.
  • Figure 2 is the experimental results of the OVCAR-3 xenograft tumor model.
  • Figure 3 is the experimental results of the MV4-11 xenograft tumor model.
  • the present inventors unexpectedly prepared a compound with excellent CDK kinase inhibitory activity and a preparation method thereof. On this basis, the inventors have completed the present invention.
  • halogen refers to F, Cl, Br or I.
  • C1-C6 alkyl refers to a linear or branched alkyl group including 1-6 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl group, tert-butyl group, neopentyl group, tertyl group, or similar groups.
  • C1-C4 alkyl “C1-C2 alkyl” have similar meanings.
  • C2-C6 alkenyl refers to a straight chain or branched alkenyl group with 2-6 carbon atoms containing a double bond, including non-limiting ethenyl, propenyl, butenyl , Isobutenyl, Pentenyl and Hexenyl etc.
  • C2-C6 alkynyl refers to a straight-chain or branched-chain alkynyl group with 2-6 carbon atoms containing a triple bond, including without limitation ethynyl, propynyl, butynyl, group, isobutynyl, pentynyl and hexynyl, etc.
  • C3-C8 cycloalkyl refers to a cyclic alkyl group having 3-8 carbon atoms in the ring, including without limitation cyclopropyl, cyclobutyl, cyclopentyl, cyclo Hexyl, cycloheptyl, cyclooctyl, etc.
  • C3-C10 cycloalkyl “C3-C6 cycloalkyl” have similar meanings.
  • C1-C6 alkoxy refers to a straight-chain or branched alkoxy group with 1-6 carbon atoms, including without limitation methoxy, ethoxy, propoxy, Isopropoxy and butoxy, etc. Preference is given to C1-C4 alkoxy.
  • heterocycloalkyl is a 4-8 membered heterocycloalkyl group containing 1, 2 or 3 heteroatoms selected from N, O, S, including (but not limited to) the following groups :
  • heterocycloalkyl group containing 1-4 heteroatoms selected from N, O, S has a similar meaning.
  • aromatic ring or “aryl” has the same meaning, preferably “C6-C10 aryl”.
  • C6-C10 aryl refers to an aromatic ring group having 6-10 carbon atoms without heteroatoms in the ring, such as phenyl, naphthyl and the like.
  • 3-10 membered aryl refers to an aromatic ring group having 3-10 carbon atoms without heteroatoms in the ring, such as phenyl, naphthyl and the like.
  • 6-10 membered aryl has a similar meaning.
  • heteroaryl has the same meaning and refers to a heteroaromatic group containing one to more heteroatoms.
  • heteroaromatic group containing one to more heteroatoms.
  • 3-10 membered heteroaryl refers to an aromatic heterocyclic ring containing 1 to 4 heteroatoms selected from oxygen, sulfur and nitrogen and 3 to 10 carbon atoms.
  • Non-limiting examples include: furyl, thienyl, pyridyl, pyrazolyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, imidazolyl, tetrazolyl, and the like.
  • the heteroaryl ring may be fused to an aryl, heterocyclyl or cycloalkyl ring, wherein the ring bonded to the parent structure is a heteroaryl ring.
  • Heteroaryl groups can be optionally substituted or unsubstituted.
  • halo refers to substitution by halogen.
  • substituted means that one or more hydrogen atoms on a specific group are replaced by a specific substituent.
  • the specific substituents are the corresponding substituents described above, or the substituents appearing in each embodiment.
  • a substituted group may have a substituent selected from a specific group at any substitutable position of the group, and the substituents may be the same or different at each position.
  • substituents contemplated by this invention are those that are stable or chemically feasible.
  • the substituents are for example (but not limited to): halogen, hydroxyl, carboxyl (-COOH), C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 3- to 12-membered heterocyclic group, aryl group, heteroaryl group, C1-C8 aldehyde group, C2-C10 acyl group, C2-C10 ester group, amino group, C1-C6 alkoxy group, C1-C10 sulfonyl group, etc.
  • the term 1-6 means 1, 2, 3, 4, 5 or 6. Other similar terms have similar meanings.
  • the present invention provides a compound of formula (I), or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof,
  • any one of R 1 ', R 2 ', ring A, R 31 , R 41 , R 51 , L', and n is independently the specific compound of the present invention The corresponding group in .
  • the compound is preferably the compound prepared in the embodiment of the present invention.
  • the term "pharmaceutically acceptable salt” refers to a salt of a compound of the present invention with an acid or a base which is suitable for use as a medicine.
  • Pharmaceutically acceptable salts include inorganic salts and organic salts.
  • a preferred class of salts are the salts of the compounds of the invention with acids.
  • Acids suitable for forming salts include, but are not limited to: inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid; formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, Fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, benzoic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, benzenesulfonic acid, naphthalenesulfonic acid and other organic acids; Amino acids such as amino acid, phenylalanine, aspartic acid, and glutamic acid.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid
  • salts of the compounds of the present invention with bases such as alkali metal salts (e.g. sodium or potassium salts), alkaline earth metal salts (e.g. magnesium or calcium salts), ammonium salts (e.g.
  • lower alkanolammonium salts and other pharmaceutically acceptable amine salts such as methylamine salts, ethylamine salts, propylamine salts, dimethylamine salts, trimethylamine salts, diethylamine salts, triethylamine salts, tert-butyl amine salts, ethylenediamine salts, hydroxyethylamine salts, dihydroxyethylamine salts, trihydroxyethylamine salts, and amine salts formed from morpholine, piperazine, and lysine, respectively.
  • methylamine salts such as methylamine salts, ethylamine salts, propylamine salts, dimethylamine salts, trimethylamine salts, diethylamine salts, triethylamine salts, tert-butyl amine salts, ethylenediamine salts, hydroxyethylamine salts, dihydroxyethylamine salts, trihydroxyethylamine salt
  • solvate refers to a complex in which a compound of the present invention coordinates with solvent molecules to form a specific ratio.
  • prodrug includes that it itself may be biologically active or inactive, and when taken in an appropriate manner, it undergoes metabolism or chemical reactions in the human body to convert into a class of compounds of formula (I), or A salt or solution of a compound of formula (I).
  • the prodrugs include (but are not limited to) carboxylates, carbonates, phosphates, nitrates, sulfates, sulfone esters, sulfoxide esters, amino compounds, carbamates, azo compounds of the compounds , phosphoramide, glucoside, ether, acetal and other forms.
  • the preparation method of the compound of formula (I) of the present invention is described in more detail below, but these specific methods do not constitute any limitation to the present invention.
  • the compounds of the present invention can also be conveniently prepared by optionally combining various synthetic methods described in the specification or known in the art, and such combinations can be easily performed by those skilled in the art to which the present invention belongs.
  • the preparation process of the compounds of the present invention is shown in the examples of the present invention, and the raw materials and reagents used therein can be purchased through commercial channels unless otherwise specified.
  • compositions and methods of administration are provided.
  • the compound of the present invention has excellent antitumor activity, the compound of the present invention and its various crystal forms, pharmaceutically acceptable inorganic or organic salts, hydrates or solvates, and drugs containing the compound of the present invention as the main active ingredient
  • the composition can be used for the treatment, prevention and alleviation of tumor-related diseases.
  • the pharmaceutical composition of the present invention comprises the compound of the present invention or a pharmacologically acceptable salt thereof within a safe and effective amount range and a pharmaceutically acceptable excipient or carrier.
  • safe and effective dose refers to: the amount of the compound is sufficient to obviously improve the condition without causing severe side effects.
  • the pharmaceutical composition contains 1-2000 mg of the compound of the present invention per dose, more preferably 10-1000 mg of the compound of the present invention per dose.
  • the "one dose” is a capsule or tablet.
  • “Pharmaceutically acceptable carrier” refers to: one or more compatible solid or liquid fillers or gel substances, which are suitable for human use, and must have sufficient purity and low enough toxicity. "Compatibility” herein means that the components of the composition can be blended with the compound of the present invention and with each other without significantly reducing the efficacy of the compound.
  • Examples of pharmaceutically acceptable carrier parts include cellulose and derivatives thereof (such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid , magnesium stearate), calcium sulfate, vegetable oil (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (such as ), wetting agent (such as sodium lauryl sulfate), coloring agent, flavoring agent, stabilizer, antioxidant, preservative, pyrogen-free water, etc.
  • cellulose and derivatives thereof such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.
  • gelatin such as talc
  • solid lubricants such as stearic acid , magnesium stearate
  • calcium sulfate such
  • the pharmaceutical composition is injection, capsule, tablet, pill, powder or granule.
  • the mode of administration of the compound or pharmaceutical composition of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical administration .
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is admixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with (a) fillers or extenders, for example, Starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders such as hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, For example, glycerol; (d) disintegrants, for example, agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow agents, such as paraffin; (f) Absorption accelerators such as quaternary ammonium compounds; (g) wetting agents such as cetyl alcohol and glyceryl mono
  • Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared with coatings and shell materials, such as enteric coatings and others well known in the art. They may contain opacifying agents and, in such compositions, the release of the active compound or compounds may be in a certain part of the alimentary canal in a delayed manner.
  • coatings and shell materials such as enteric coatings and others well known in the art. They may contain opacifying agents and, in such compositions, the release of the active compound or compounds may be in a certain part of the alimentary canal in a delayed manner.
  • Examples of usable embedding components are polymeric substances and waxy substances.
  • the active compounds can also be in microencapsulated form, if desired, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures.
  • liquid dosage forms may contain inert diluents conventionally used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or mixtures of these substances, etc.
  • inert diluents conventionally used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and
  • compositions can also contain adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • Suspensions in addition to the active compounds, may contain suspending agents, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
  • suspending agents for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
  • compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Suitable aqueous and non-aqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols, and suitable mixtures thereof.
  • Dosage forms for topical administration of a compound of this invention include ointments, powders, patches, sprays and inhalants.
  • the active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required, if necessary.
  • the compound of the present invention can be administered alone or in combination with other pharmaceutically acceptable compounds (such as antineoplastic drugs).
  • other pharmaceutically acceptable compounds such as antineoplastic drugs.
  • the treatment method of the present invention can be used alone or in combination with other treatment methods or drugs.
  • a safe and effective amount of the compound of the present invention is applied to a mammal (such as a human) in need of treatment, wherein the dosage is a pharmaceutically effective dosage when administered, and for a person with a body weight of 60kg, the daily
  • the dosage is usually 1-2000 mg, preferably 50-1000 mg.
  • factors such as the route of administration and the health status of the patient should also be considered for the specific dosage, which are within the skill of skilled physicians.
  • the present invention has the following main advantages:
  • the compound has excellent CDK kinase inhibitory activity
  • the compound has excellent cytostatic activity
  • the compound has excellent drug efficacy in vivo.
  • the starting material S1 is reduced by lithium aluminum hydride to obtain the intermediate S1-1, and then nucleophilicly substituted with various substituted primary amines to obtain the product S1-2, and then oxidized by manganese dioxide to obtain the aldehyde pyrimidine compound S1-3, and then Cyclization reaction with acetate containing different substituents, followed by oxidation to obtain sulfone-pyrimidinone intermediate S1-5, and reaction of sulfone-pyrimidinone intermediate S1-5 with substituted primary amine to obtain the final product.
  • Substituted sulfonyl chloride S2 is reacted with 4-Boc-aminopiperidine to obtain sulfonamide intermediate S2-1, and then deprotected to obtain intermediate S2-3.
  • Filter wash the filter cake with cold ethyl acetate, add the filter cake to water (150ml) and ethyl acetate (200mL), adjust the pH to 1 with 4N aqueous hydrochloric acid solution, separate the layers, and adjust the aqueous phase with 6N aqueous sodium hydroxide solution PH to 11, extracted with ethyl acetate (200mL x2), dried the organic phase with anhydrous sodium sulfate, filtered, concentrated, and repeated the above operations until the ee value was greater than 99%, and 8 g of the product with 99.3% ee was obtained.
  • Dissolve compound int2 (290mg, 2.6eq) in dry THF (5ml), replace nitrogen, then cool down to -70°C, add dropwise lithium dimethylsilylamide (1.0M, 2.06ml, 4.0eq), dropwise , kept at -70°C for 1h, then added dropwise a solution of compound 1 (137.8mg, 1.0eq) in tetrahydrofuran (3ml), and reacted overnight at 40°C.
  • TLC monitors that the reaction is complete, lower the temperature, add dropwise saturated ammonium chloride aqueous solution to the reaction system to quench, then add ethyl acetate (30ml x 3) for extraction, separate liquids, collect ethyl acetate and add anhydrous sodium sulfate to dry, filter and spin
  • the product (90mg) was obtained by drying on the column.
  • Tetrahydrofuran (10 mL) was added into a 100 mL three-necked flask, cooled to -5°C, and a solution of titanium tetrachloride (3.04 g, 16 mmol) in dichloromethane (5 mL) was added dropwise. After dropping and stirring for 10 minutes, add dropwise int1 (1.07g, 4mmol) and tetrahydrofuran (10mL) solution of diethyl malonate (1.28g, 8mmol) in the reaction solution, after dropping and stirring for 30 minutes, add dropwise pyridine ( 1.58g, 20mmol), after dropping, it was naturally raised to room temperature and stirred overnight. TLC monitored the completion of the reaction.
  • OXONE 160mg, 0.268mmol was added to a mixed solution of C4-5 (45mg, 0.107mmol) in tetrahydrofuran and water, and after the addition, it was naturally raised to room temperature for 2 hours. The completion of the reaction was monitored by TLC, diluted with water, extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, and concentrated to obtain 51 mg of crude product.
  • MCF7, T47D, OVCAR3 and HCC1806 cells were seeded in 96-well plates at 3000, 3000, 2500 and 2000 cells/well, respectively, and cultured overnight in a 37°C, 5% CO 2 incubator.
  • a) Prepare a gradient dilution solution of the test compound: Palbociclib, the compound of the example is 10 mM as the stock solution. Then take 2.5 ⁇ l of the stock solution and dissolve it in 497.5 ⁇ l of DMSO-free culture solution, and then carry out 3-fold serial gradient dilution with 0.1% DMSO culture solution, with a total of 9 concentrations.
  • concentration of the compound after dilution is as follows:
  • %Cell Survival 100% ⁇ (OD_Sample-OD_LCave)/(OD_HC-OD_LCave)
  • OD_Sample Cell readout for spiked compounds
  • IC50 (nM) values were calculated by concentration-response curve fitting.
  • A represents IC50 value ⁇ 50nM
  • B represents IC50 value > 50nM and ⁇ 200nM
  • C represents IC50 value > 200nM and ⁇ 1000nM
  • D represents IC50 value > 1000nM.
  • the patented compound has a good inhibitory effect on both palbociclib-sensitive and drug-resistant cells. Compared with PF-06873600, the patented compound has the same or even better activity, and is expected to be further developed into a drug-resistant Drugs for regulating CDK kinase activity or treating CDK-related diseases.
  • EMEM medium ATCC,30-2003
  • DMEM/F12 medium Gabco-11330-032
  • Microplate reader e.g.MD-SpectraMax ID5 or EnVision
  • the inhibition rate was calculated using the following formula:
  • the patented compound has a good inhibitory rate on palbociclib-resistant cell lines, which is better than PF-06873600.
  • A stands for IC50 value ⁇ 10nM
  • B stands for IC50 value > 10nM and ⁇ 100nM
  • C stands for IC50 value > 100nM and ⁇ 500nM
  • D stands for IC50 value > 500nM
  • 1 ⁇ 107xMCF-7/Palbo-R cells were suspended in 0.2mL of a mixture of Matrigel and mouse (1:1), and injected subcutaneously into 6-8-week-old Balb/c nube female mice .
  • mice were randomly assigned to 5 groups, each group consisted of 3 animals, and the doses used in each group were: (a) Vehicle; (b) 10mg/kg PF-06873600; (c) 20mg/kg C27; (d) 10mg /kg C3; (h) 20mg/kg C3.
  • Doses were administered by oral gavage twice daily for 20 days. Tumor volumes and animal body weights were measured twice a week until the end of the experiment. The day of dosing was defined as day 0. The measurement time points were the 0th day, the 3rd day, the 7th day, the 10th day, the 14th day, the 17th day and the 20th day. All mice were euthanized by cervical dislocation. At the end of the experiment, the tumors were collected and weighed to calculate the TGI value (see Table 4). The experimental results are shown in Figure 1.
  • OVCAR-3 cells were cultured in RPMI1640 medium containing 20% fetal bovine serum and 10 ⁇ g/mL Insalin. Cells in the exponential growth phase were collected, mixed with PBS and Matrigel 1:1, and resuspended to a suitable concentration for subcutaneous tumor inoculation in nude mice.
  • OVCAR-3 cells were suspended in a mixture of 0.2mL PBS and Matrigel (1:1), and injected subcutaneously into the right side of a 6-8 week old BALB/c nube female mouse. side back.
  • mice were randomly assigned to 6 groups, each group contained 5 animals, and the doses used in each group were: (a) Vehicle; (b) 60mg/kg Palbociclib; (c) 30mg/kg PF-06873600; (d) 30mg /kg C3; (e) 30 mg/kg C29; (f) 50 mg/kg C29.
  • Vehicle is DMSO/Solutol/Saline (5%/10%/85%), Palbociclib, PF-06873600, C3, C29 are dissolved in 6mg/mL, 3mg/mL, 3mg/mL, 3mg/mL and 5mg/mL respectively Vehicle.
  • mice in Vehicle 30mg/kg PF-06873600, 30mg/kg C3, 30mg/kg C29 and 50mg/kg C29 groups were administered twice a day by oral gavage for 21 days; mice in 60mg/kg Palbociclib group were administered daily
  • Dosing was given once by oral gavage for 21 days. Tumor volumes and animal body weights were measured twice a week until the end of the experiment. The day of dosing was defined as day 0. The measurement time points were the 0th day, the 3rd day, the 7th day, the 10th day, the 14th day, the 17th day and the 21st day. All mice were euthanized by cervical dislocation. At the end of the experiment, the tumors were collected and weighed to calculate the TGI value (see Table 5). The experimental results are shown in Figure 2.
  • MV-4-11 cells were cultured in IMEM medium containing 10% fetal bovine serum. Cells in the exponential growth phase were collected and resuspended in PBS to a suitable concentration for subcutaneous tumor inoculation in nude mice.
  • mice were randomly assigned to 5 groups, and each group contained 5 animals: (a) Vehicle; (b) 20mg/kg Palbociclib; (c) 10mg/kg PF-06873600; (d) 10mg/kg C3; (e) 20mg/kg C3.
  • Vehicle is DMSO/Solutol/Saline (5%/10%/85%), Palbociclib, PF-06873600, and TYK-00127 are dissolved in Vehicle at concentrations of 2mg/mL, 1mg/mL, 1mg/mL and 2mg/mL respectively .
  • mice in Vehicle 10mg/kg TY-3301, 10mg/kg C3, and 20mg/kg C3 were administered twice a day by oral gavage for 15 days; mice in 20mg/kg TY-3300 group were given oral gavage daily
  • the feeding method is administered once for 15 days.
  • Tumor volumes and animal body weights were measured twice a week until the end of the experiment.
  • the day of dosing was defined as day 0.
  • the measurement time points were the 0th day, the 3rd day, the 7th day, the 10th day, the 14th day and the 15th day. All mice were euthanized by cervical dislocation.
  • the tumors were collected and weighed to calculate the TGI value (see Table 6).
  • Table 6 The experimental results are shown in Figure 3.
  • the present invention provides a class of compounds with excellent kinase, cell, and in vivo efficacy and their synthesis methods. This type of compound is expected to be developed to solve the problem of drug resistance such as palbociclib in existing clinics, and bring new drugs to this type of patients. treatment plan.

Abstract

本发明涉及用作CDK激酶抑制剂的化合物及其应用。具体地,本发明化合物具有式(I)所示结构,其中各基团和取代基的定义如说明书中所述;本发明还公开了所述化合物的制备方法及其在调节CDK激酶活性或治疗CDK相关疾病方面的用途。

Description

用作CDK激酶抑制剂的化合物及其应用 技术领域
本发明涉及医药技术领域,具体涉及用作CDK激酶抑制剂的化合物,及其在调节CDK激酶活性或治疗CDK相关疾病方面的应用。
背景技术
细胞周期(cell cycle)是指细胞从一次***完成开始到下一次***结束所经历的全过程,分为间期与***期两个阶段。间期又分为三期即DNA合成前期(G1期)、DNA合成期(S期)与DNA合成后期(G2期);***期即M期。细胞周期蛋白(Cyclins)和细胞周期蛋白依赖性激酶(CDKs)是整个细胞周期调控机制中的核心分子。已经鉴定出至少16种哺乳动物细胞周期蛋白,其中细胞周期蛋白B/CDK1(CyclinB/CDK1)、细胞周期蛋白A/CDK2(CyclinA/CDK2)、细胞周期蛋白E/CDK2(CyclinE/CDK2)、细胞周期蛋白D/CDK4(CyclinD/CDK4)、细胞周期蛋白D/CDK6(CyclinD/CDK6)和可能的其他heterodynes是细胞周期进展的重要调节因子。细胞周期蛋白/CDK heterodynes的其他功能包括转录调节、DNA修复、分化和凋亡。
已证实细胞周期失调是人内癌症的一个共同特征,细胞周期蛋白依赖性激酶(CDKs)抑制剂对细胞周期的调控具有重要的作用。在癌症治疗领域有着广泛的应用前景。如CDK4/6抑制剂帕博西尼(palbociclib)、瑞博西尼(ribociclib)和玻玛西尼(abemaciclib)针对乳腺癌和其他癌症的治疗;但与其他激酶抑制剂一样,它们的作用可能随着时间的推移被原发性或获得性抗性的发展限制。因此,辉瑞公司(Pfizer)开发出的PF-06873600作为一种全新的多靶点CDK激酶抑制剂,将有望解决现有条件下出现的耐药现象。PF-06873600也已进入临床I期试验用于转移性乳腺癌的治疗。
细胞周期蛋白依赖性激酶(CDKs)抑制剂作为最具前景的癌症治疗领域,开发新的具有CDK激酶抑制活性的、具有更好药效学、药代动力学性能的化合物已成为开发新型抗肿瘤药物的重要研究项目,并最终用于人类肿瘤等疾病的治疗中。
发明内容
本发明的目的在于提供一种式(I)所示化合物及其制备方法和其在用于CDK2/4/6激酶抑制剂药物方面的用途。
本发明的第一方面,提供了式(Ⅰ)化合物、或其药学上可接受的盐、立体异构体、溶剂化物或前药,
Figure PCTCN2022097927-appb-000001
其中:
R 1选自下组:未取代或1~4个R 6取代的C3-C10环烷基、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元并环、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元螺环、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元桥环、未取代或1~4个R 6取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、未取代或1~4个R 6取代的C6-C10芳基、未取代或1~4个R 6取代的含1-5个选自N、O、S的杂原子的3-10元杂芳基、未取代或1~4个R 6取代的C1-C6 烷基;
R 2选自下组:H、F、OH、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基,且R 2可与环上的原子相连形成螺环、桥环、或并环结构;
R 3a和R 3b独立地选自下组:H、F、OH、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
其中,R 2、R 3a和R 3b中每个所述C1-C4烷基和卤代C1-C4烷基任选被卤素、OH、C1-C4烷氧基或卤代C1-C4烷氧基取代;
R 4选自下组:Br、取代或未取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、-OR 4'、-SR 4'、-NR 4'R 4",其中,R 4'和R 4"分别独立地选自下组:H、COR 7、取代或未取代的C1-C6烷基、取代或未取代的C3-C6环烷基、取代或未取代的含1-4个选自N、O、S的杂原子的3-6元杂环烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、OH、CN、=O、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
R 5选自下组:H、卤素、C1-C2烷基、卤代C1-C2烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
各R 6独立地选自下组:H、氘、羟基、卤素、氰基、=O、COR 7、CO 2R 7、CONR 8R 9、CO 2NR 8R 9、SO 2R 7、SO 2NR 8R 9、NR 8SO 2R 7、NHSO 2NR 8R 9
Figure PCTCN2022097927-appb-000002
NR 8R 9、R”’取代或未取代的含1-3个选自N、O、S的杂原子的3-10元杂环烷基、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、C3-C8环烷基、卤代C3-C8环烷基、C4-C10螺环、C3-C10并环、C4-C10桥环、硫代C1~C6烷基、C6-C10芳基、含1-5个选自N、O、S的杂原子的3-10元杂芳基;
R”’选自下组:=O、NR’R”、C1-C4烷基、卤代C1-C4烷基;
各R 7独立地选自下组:C1-C4烷基、-L-(C3-C8环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基)、-L-(3-10元芳基),其中所述的C1-C4烷基、C3-C8环烷基、3-10元杂环烷基、3-10元杂芳基、3-10元芳基任选的被0~4个D、OH、卤素、CN、C1-C4烷基、卤代C1-C4烷基、N(C1-C4烷基) 2、NHCO(C1-C4烷基)、SO 2(C1-C4烷基)、CO 2(C1-C4烷基)、含1-4个选自N、O、S的杂原子的3-6元杂环烷基、CO(C1-C4烷基)、C1-C4烷氧基或卤代C1-C4烷氧基取代;
R 8和R 9独立地选自下组:H、C1-C4烷基、卤代C1-C4烷基、C3-C8环烷基、-L-(C3-C8环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基);或者
R 8和R 9可以与其所连接的N一起形成取代或未取代的4-6元杂环基,所述取代指被选自下组的1-3个取代基取代:=O、NR’R”、C1-C6烷基;
各R’和R”独立地选自下组:H、C1-C4烷基;
各L独立地为键或C1-C4亚烷基,所述C1-C4亚烷基任选被OH、C1-C4烷氧基或卤代C1-C4烷氧基取代;
p选自下组:0、1、2、3、4;
r选自下组:0、1、2、3、4。
在另一优选例中,R 1选自下组:未取代或1~4个R 6取代的C3-C10环烷基、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元并环、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元螺环、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元桥环、未取代或1~4个R 6取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、未取代或1~4个R 6取代的C6-C10芳基、未取代或1~4个R 6取代的含1-5个选自N、O、S的杂原子的3-10元杂芳基;
R 2选自下组:H、F;
R 3a和R 3b独立地选自下组:H、F、OH、C1-C4烷基;
R 4选自下组:取代或未取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、-OR 4'、-NR 4'R 4",其中,R 4'和R 4"分别独立地选自下组:H、取代或未取代的C1-C6烷基所述取代指被选自下组的1-3个取代基取代:氘、卤素、C1-C4烷基;
R 5选自下组:H、C1-C2烷基;
各R 6独立地选自下组:H、氘、羟基、卤素、氰基、=O、COR 7、CO 2R 7、CONR 8R 9、SO 2R 7、SO 2NR 8R 9、NR 8SO 2R 7、NHSO 2NR 8R 9、NR 8R 9、R”’取代或未取代的含1-3个选自N、O、S的杂原子的3-10元杂环烷基、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、C3-C8环烷基、卤代C3-C8环烷基、C4-C10螺环、C3-C10并环、C4-C10桥环、硫代C1~C6烷基、C6-C10芳基、含1-5个选自N、O、S的杂原子的3-10元杂芳基;
R”’选自下组:=O、NR’R”、C1-C4烷基、卤代C1-C4烷基;
各R 7独立地选自下组:C1-C4烷基、-L-(C3-C8环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基)、-L-(3-10元芳基),其中所述的C1-C4烷基、C3-C8环烷基、3-10元杂环烷基、3-10元杂芳基、3-10元芳基任选的被0~4个D、OH、卤素、CN、C1-C4烷基、卤代C1-C4烷基、N(C1-C4烷基) 2、NHCO(C1-C4烷基)、SO 2(C1-C4烷基)、CO 2(C1-C4烷基)、含1-4个选自N、O、S的杂原子的3-6元杂环烷基、CO(C1-C4烷基)、C1-C4烷氧基或卤代C1-C4烷氧基取代;
R 8和R 9独立地选自下组:H、C1-C4烷基、卤代C1-C4烷基、C3-C8环烷基、-L-(C3-C8环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基);或者
R 8和R 9可以与其所连接的N一起形成取代或未取代的4-6元杂环基,所述取代指被选自下组的1-3个取代基取代:=O、NR’R”、C1-C6烷基;
各R’和R”独立地选自下组:H、C1-C4烷基;
各L独立地为键或C1-C4亚烷基,所述C1-C4亚烷基任选被OH、C1-C4烷氧基或卤代C1-C4烷氧基取代;
p选自下组:0、1、2、3、4;
r选自下组:0、1、2、3、4。
在另一优选例中,R 1选自下组:未取代或1~4个R 6取代的C3-C10环烷基、未取代或1~4个R 6取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、未取代或1~4个R 6取代的C6-C10芳基、未取代或1~4个R 6取代的含1-5个选自N、O、S的杂原子的3-10元杂芳基;
R 2选自下组:H、F;
R 3a和R 3b独立地选自下组:H、F、OH、C1-C4烷基;
R 4选自下组:取代或未取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、-OR 4',其中,R 4'独立地选自下组:H、取代或未取代的C1-C6烷基所述取代指被选自下组的1-3个取代基取代:氘、卤素、C1-C4烷基;
R 5选自下组:H、C1-C2烷基;
各R 6独立地选自下组:H、氘、羟基、卤素、氰基、=O、COR 7、CO 2R 7、CONR 8R 9、SO 2R 7、SO 2NR 8R 9、NR 8SO 2R 7、NHSO 2NR 8R 9、NR 8R 9、R”’取代或未取代的含1-3个选自N、O、S的杂原子的3-10元杂环烷基、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、C3-C8环烷基、卤代C3-C8环烷基、C4-C10螺环、C3-C10并环、C4-C10桥环、硫代C1~C6烷基、C6-C10芳基、含1-5个选自N、O、S的杂原子的3-10元杂芳基;
R”’选自下组:=O、NR’R”、C1-C4烷基、卤代C1-C4烷基;
各R 7独立地选自下组:C1-C4烷基、-L-(C3-C8环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基)、-L-(3-10元芳基),其中所述的C1-C4烷基、C3-C8环烷基、3-10元杂环烷基、3-10元杂芳基、3-10 元芳基任选的被0~4个D、OH、卤素、CN、C1-C4烷基、卤代C1-C4烷基、N(C1-C4烷基) 2、NHCO(C1-C4烷基)、SO 2(C1-C4烷基)、CO 2(C1-C4烷基)、含1-4个选自N、O、S的杂原子的3-6元杂环烷基、CO(C1-C4烷基)、C1-C4烷氧基或卤代C1-C4烷氧基取代;
R 8和R 9独立地选自下组:H、C1-C4烷基、卤代C1-C4烷基、C3-C8环烷基、-L-(C3-C8环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基);或者
R 8和R 9可以与其所连接的N一起形成取代或未取代的4-6元杂环基,所述取代指被选自下组的1-3个取代基取代:=O、NR’R”、C1-C6烷基;
各R’和R”独立地选自下组:H、C1-C4烷基;
各L独立地为键或C1-C4亚烷基,所述C1-C4亚烷基任选被OH、C1-C4烷氧基或卤代C1-C4烷氧基取代;
p选自下组:0、1、2、3、4;
r选自下组:0、1、2、3、4。
在另一优选例中,R 1选自下组:未取代或1~4个R 6取代的C3-C10环烷基、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元并环、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元螺环、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元桥环、未取代或1~4个R 6取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、未取代或1~4个R 6取代的C6-C10芳基、未取代或1~4个R 6取代的含1-5个选自N、O、S的杂原子的3-10元杂芳基、未取代或1~4个R 6取代的C1-C6烷基;
R 2选自下组:H、F、OH、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基,且R 2可与环上的原子相连形成螺环、桥环、或并环结构;
R 3a和R 3b独立地选自下组:H、F、OH、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
其中,R 2、R 3a和R 3b中每个所述C1-C4烷基和卤代C1-C4烷基任选被卤素、OH、C1-C4烷氧基或卤代C1-C4烷氧基取代;
R 4选自下组:Br、取代或未取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、-OR 4'、-SR 4'、-NR 4'R 4",其中,R 4'和R 4"分别独立地选自下组:H、COR 7、取代或未取代的C1-C6烷基、取代或未取代的C3-C6环烷基、取代或未取代的含1-4个选自N、O、S的杂原子的3-6元杂环烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、OH、CN、=O、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
R 5选自下组:H、卤素、C1-C2烷基、卤代C1-C2烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
各R 6独立地选自下组:H、氘、羟基、卤素、氰基、=O、COR 7、CO 2R 7、CONR 8R 9、CO 2NR 8R 9、SO 2R 7、SO 2NR 8R 9、NR 8SO 2R 7、NHSO 2NR 8R 9
Figure PCTCN2022097927-appb-000003
NR 8R 9、R”’取代或未取代的含1-3个选自N、O、S的杂原子的3-10元杂环烷基、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、C3-C8环烷基、卤代C3-C8环烷基、C4-C10螺环、C3-C10并环、C4-C10桥环、硫代C1~C6烷基、C6-C10芳基、含1-5个选自N、O、S的杂原子的3-10元杂芳基;
R”’选自下组:=O、NR’R”;
各R 7独立地选自下组:C1-C4烷基、-L-(C3-C8环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基)、-L-(3-10元芳基),其中所述的C1-C4烷基、C3-C8环烷基、3-10元杂环烷基、3-10元杂芳基、3-10元芳基任选的被0~4个D、OH、卤素、CN、C1-C4烷基、卤代C1-C4烷基、N(C1-C4烷基) 2、 NHCO(C1-C4烷基)、SO2(C1-C4烷基)、含1-4个选自N、O、S的杂原子的3-6元杂环烷基、CO(C1-C4烷基)、C1-C4烷氧基或卤代C1-C4烷氧基取代;
R 8和R 9独立地选自下组:H、C1-C4烷基、卤代C1-C4烷基、C3-C8环烷基、-L-(C3-C8环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基);或者
R 8和R 9可以与其所连接的N一起形成取代或未取代的4-6元杂环基,所述取代指被选自下组的1-3个取代基取代:=O、NR’R”、C1-C6烷基;
各R’和R”独立地选自下组:H、C1-C4烷基;
各L独立地为键或C1-C4亚烷基,所述C1-C4亚烷基任选被OH、C1-C4烷氧基或卤代C1-C4烷氧基取代;
p选自下组:0、1、2、3、4;
r选自下组:0、1、2、3、4。
在另一优选例中,所述化合物具有式(II)或式(Ⅲ)所示结构:
Figure PCTCN2022097927-appb-000004
其中:
A环选自下组:含0-4个R 1’取代的6-10元芳基、含0-4个R 1’取代的C3-C8环烷基、含0-4个R 1’取代的含1-5个选自N、O、S的杂原子的5-10元杂芳基;
各R 1’独立地选自下组:氘、卤素、OH、CN、SO 2R 31、COR 31、CO 2R 31、NR 41R 51、NHCOR 41、CONR 41R 51、OCONR 41R 51、NHCON R 41R 51、NHCOOR 41、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、取代或未取代的C3-C6环烷基、取代或未取代的含1-4个选自N、O、S的杂原子的3-6元杂环烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、OH、CN、=O、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
R 2’选自下组:H、氘;
R 3’选自下组:取代或未取代的C1-C4烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、OH、CN、=O、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
R 4’选自下组:H、氘、OH;R 31选自下组:H、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、取代或未取代的C3-C6环烷基、取代或未取代的含1-4个选自N、O、S的杂原子的3-6元杂环烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、OH、CN、=O、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
R 41和R 51独立地选自下组:H、C1-C4烷基、卤代C1-C4烷基、C3-C8环烷基、-L’-(C3-C8环烷基)、-L’-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L’-(含1-5个选自N、O、S的杂原子的3-10元杂芳基),L’为键或C1-C6亚烷基;或者
R 41和R 51可以与其所连接的N一起形成取代或未取代的4-6元杂环基,所述取代指被选自下组的1-3个取代基取代:卤素、OH、=O、C1-C6烷基;
n选自下组:0、1、2、3、4。
在另一优选例中,所述化合物具有式(II)所示结构:
Figure PCTCN2022097927-appb-000005
其中:
A环选自下组:含0-4个R 1’取代的6-10元芳基、含0-4个R 1’取代的C3-C8环烷基、含0-4个R 1’取代的含1-5个选自N、O、S的杂原子的5-10元杂芳基;
各R 1’独立地选自下组:氘、卤素、OH、CN、SO 2R 31、COR 31、CO 2R 31、NR 41R 51、NHCOR 41、CONR 41R 51、OCONR 41R 51、NHCON R 41R 51、NHCOOR 41、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、取代或未取代的C3-C6环烷基、取代或未取代的含1-4个选自N、O、S的杂原子的3-6元杂环烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、OH、CN、=O、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
R 2’选自下组:H、氘;
R 31选自下组:H、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、取代或未取代的C3-C6环烷基、取代或未取代的含1-4个选自N、O、S的杂原子的3-6元杂环烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、OH、CN、=O、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
R 41和R 51独立地选自下组:H、C1-C4烷基、卤代C1-C4烷基、C3-C8环烷基、-L’-(C3-C8环烷基)、-L’-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L’-(含1-5个选自N、O、S的杂原子的3-10元杂芳基),L’为键或C1-C6亚烷基;或者
R 41和R 51可以与其所连接的N一起形成取代或未取代的4-6元杂环基,所述取代指被选自下组的1-3个取代基取代:卤素、OH、=O、C1-C6烷基;
n选自下组:0、1、2、3、4。
在另一优选例中,A环为含0-4个R 1’取代的6-10元芳基。
在另一优选例中,R 3’为C1-C4烷基。
在另一优选例中,A环为苯基。
在另一优选例中,
Figure PCTCN2022097927-appb-000006
具有如下结构:
Figure PCTCN2022097927-appb-000007
在另一优选例中,
Figure PCTCN2022097927-appb-000008
中,R 1’选自下组:H、卤素、CN、C1-C4烷氧基。
在另一优选例中,
Figure PCTCN2022097927-appb-000009
中,R 1’为F、OMe。
在另一优选例中,
Figure PCTCN2022097927-appb-000010
中,A环为苯基,n为1,R 1’选自下组:H、卤素、CN、C1-C4烷氧基。
在另一优选例中,R 1’为F。
在另一优选例中,
Figure PCTCN2022097927-appb-000011
具有如下结构:
Figure PCTCN2022097927-appb-000012
在另一优选例中,
Figure PCTCN2022097927-appb-000013
中,R 1’为F。
在另一优选例中,A环为含0-4个R 1’取代的C3-C8环烷基。
在另一优选例中,所述化合物具有式(IV)或式(V)所示结构:
Figure PCTCN2022097927-appb-000014
其中,q选自下组:1、2、3、4、5、6。
在另一优选例中,A环为含0-4个R 1’取代的含1-5个选自N、O、S的杂原子的5-10元杂芳基。
在另一优选例中,A环选自下组:含0-4个R 1’取代的6-10元芳基、含0-4个R 1’取代的C3-C8环烷基、含0-4个R 1’取代的含1-5个选自N、O、S的杂原子的5-10元杂芳基;
各R 1’独立地选自下组:氘、卤素、OH、CN、SO2R 31、COR 31、CO2R 31、NR 41R 51、NHCOR 41、CONR 41R 51、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、取代或未取代的C3-C6环烷基、取代或未取代的含1-4个选自N、O、S的杂原子的3-6元杂环烷基;
R 2’选自下组:H、氘;
R 4’选自下组:H、氘;
R 31为C1-C4烷基;
R 41和R 51独立地选自下组:H、C1-C4烷基;
n选自下组:0、1、2、3、4。
在另一优选例中,A环选自下组:含0-4个R 1’取代的6-10元芳基、含0-4个R 1’取代的C3-C8环烷基、含0-4个R 1’取代的含1-5个选自N、O、S的杂原子的5-10元杂芳基;
各R 1’独立地选自下组:氘、卤素、OH、CN、SO2R 31、COR 31、CO2R 31、NR 41R 51、NHCOR 41、CONR 41R 51、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、取代或未取代的C3-C6环烷基、取代或未取代的含1-4个选自N、O、S的杂原子的3-6元杂环烷基;
R 2’选自下组:H、氘;
R 31为C1-C4烷基;
R 41和R 51独立地选自下组:H、C1-C4烷基;
n选自下组:0、1、2、3、4。
在另一优选例中,A环选自下组:含0-4个R 1’取代的6-10元芳基、含0-4个R 1’取代的 C3-C8环烷基;
R 1’选自下组:卤素、OH、C1-C4烷氧基、卤代C1-C4烷氧基;
R 2’选自下组:H、氘;
n选自下组:0、1、2、3、4。
在另一优选例中,所述化合物选自下组:
Figure PCTCN2022097927-appb-000015
Figure PCTCN2022097927-appb-000016
Figure PCTCN2022097927-appb-000017
Figure PCTCN2022097927-appb-000018
Figure PCTCN2022097927-appb-000019
Figure PCTCN2022097927-appb-000020
在另一优选例中,所述化合物具有式(III)所示结构:
Figure PCTCN2022097927-appb-000021
其中:
R 2’选自下组:H、氘;
R 3’选自下组:取代或未取代的C1-C4烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、OH、CN、=O、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
R 4’选自下组:H、氘、OH;
在另一优选例中,
R 2’为:H;
R 3’选自下组:取代或未取代的C1-C4烷基,所述取代指被选自下组的1-3个取代基取代:卤素、CN、C1-C4烷氧基;
在另一优选例中,所述化合物选自下组:
Figure PCTCN2022097927-appb-000022
在另一优选例中,
R 1选自下组:未取代或1~4个R 6取代的C3-C10环烷基、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元并环、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元螺环、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元桥环、未取代或1~4个R 6取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、未取代或1~4个R 6取代的C6-C10芳基、未取代或1~4个R 6取代的含1-5个选自N、O、S的杂原子的3-10元杂芳基、未取代或1~4个R 6取代的C1-C6烷基;
R 2选自下组:H、F、OH、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基,且R 2可与环上的原子相连形成螺环、桥环、或并环结构;
R 3a和R 3b独立地选自下组:H、F、OH、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
其中,R 2、R 3a和R 3b中每个所述C1-C4烷基和卤代C1-C4烷基任选被卤素、OH、C1-C4烷氧基或卤代C1-C4烷氧基取代;
R 4选自下组:Br、取代或未取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、-OR 4'、-SR 4'、-NR 4'R 4",其中,R 4'和R 4"分别独立地选自下组:H、COR 7、取代或未取代的C1-C6烷基、取代或未取代的C3-C6环烷基、取代或未取代的含1-4个选自N、O、S的杂原子的3-6元杂环烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、OH、CN、=O、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
R 5选自下组:H、卤素、C1-C2烷基、卤代C1-C2烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
各R 6独立地选自下组:H、氘、羟基、卤素、氰基、=O、COR 7、CO 2R 7、CONR 8R 9、CO 2NR 8R 9、SO 2R 7、SO 2NR 8R 9、NR 8SO 2R 7、NHSO 2NR 8R 9
Figure PCTCN2022097927-appb-000023
NR 8R 9、R”’取代或未取代的含1-3个选自N、O、S的杂原子的3-10元杂环烷基、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、C3-C8环烷基、卤代C3-C8环烷基、C4-C10螺环、C3-C10 并环、C4-C10桥环、硫代C1~C6烷基、C6-C10芳基、含1-5个选自N、O、S的杂原子的3-10元杂芳基;
R”’选自下组:=O、NR’R”;
各R 7独立地选自下组:C1-C4烷基、-L-(C3-C8环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基)、-L-(3-10元芳基),其中所述的C1-C4烷基、C3-C8环烷基、3-10元杂环烷基、3-10元杂芳基、3-10元芳基任选的被0~4个OH、卤素、C1-C4烷基、C1-C4烷氧基或卤代C1-C4烷氧基取代;
R 8和R 9独立地选自下组:H、C1-C4烷基、卤代C1-C4烷基、C3-C8环烷基、-L-(C3-C8环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基);或者
R 8和R 9可以与其所连接的N一起形成取代或未取代的4-6元杂环基,所述取代指被选自下组的1-3个取代基取代:=O、NR’R”、C1-C6烷基;
各R’和R”独立地选自下组:H、C1-C4烷基;
各L独立地为键或C1-C4亚烷基,所述C1-C4亚烷基任选被OH、C1-C4烷氧基或卤代C1-C4烷氧基取代;
p选自下组:0、1、2、3、4;
r选自下组:0、1、2、3、4。
在另一优选例中,R 1选自下组:未取代或1~4个R 6取代的C3-C10环烷基、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元并环、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元螺环、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元桥环、未取代或1~4个R 6取代的C6-C10芳基、未取代或1~4个R 6取代的含1-5个选自N、O、S的杂原子的3-10元杂芳基;
R 2选自下组:H、F;
R 3a和R 3b独立地选自下组:H、F、OH、C1-C4烷基;
其中,R 2、R 3a和R 3b中每个所述C1-C4烷基和卤代C1-C4烷基任选被卤素、OH、C1-C4烷氧基或卤代C1-C4烷氧基取代;
R 4选自下组:取代或未取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、-NR 4'R 4",其中,R 4'和R 4"分别独立地选自下组:H、取代或未取代的C1-C6烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、C1-C4烷基;
R 5选自下组:H、C1-C2烷基;
各R 6独立地选自下组:H、氘、羟基、卤素、氰基、=O、COR 7、CO 2R 7、CONR 8R 9、SO 2R 7、SO 2NR 8R 9、NR 8SO 2R 7、NHSO 2NR 8R 9、NR 8R 9、R”’取代或未取代的含1-3个选自N、O、S的杂原子的3-10元杂环烷基、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、C3-C8环烷基、卤代C3-C8环烷基、C4-C10螺环、C3-C10并环、C4-C10桥环、硫代C1~C6烷基、C6-C10芳基、含1-5个选自N、O、S的杂原子的3-10元杂芳基;
R”’选自下组:=O、NR’R”;
各R 7独立地选自下组:C1-C4烷基、-L-(C3-C8环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基)、-L-(3-10元芳基),其中所述的C1-C4烷基、C3-C8环烷基、3-10元杂环烷基、3-10元杂芳基、3-10元芳基任选的被0~4个OH、卤素、C1-C4烷基、C1-C4烷氧基或卤代C1-C4烷氧基取代;
R 8和R 9独立地选自下组:H、C1-C4烷基、卤代C1-C4烷基、C3-C8环烷基、-L-(C3-C8环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基);或者
R 8和R 9可以与其所连接的N一起形成取代或未取代的4-6元杂环基,所述取代指被选自下组的1-3个取代基取代:=O、NR’R”、C1-C6烷基;
各R’和R”独立地选自下组:H、C1-C4烷基;
各L独立地为键或C1-C4亚烷基,所述C1-C4亚烷基任选被OH、C1-C4烷氧基或卤代C1-C4烷氧基取代;
p选自下组:0、1、2、3、4;
r选自下组:0、1、2、3、4。
在另一优选例中,
R 1选自下组:
Figure PCTCN2022097927-appb-000024
Figure PCTCN2022097927-appb-000025
R 4选自下组:Br、取代或未取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、取代或未取代的C1-C4烷氧基、取代或未取代的C1-C4烷硫基、取代或未取代的C1-C4烷胺基;
R 6如权利要求1所定义。
在另一优选例中,R 1选自下组:
Figure PCTCN2022097927-appb-000026
Figure PCTCN2022097927-appb-000027
R 4选自下组:取代或未取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、取代或未取代的C1-C4烷氧基、取代或未取代的C1-C4烷胺基;
R 6如权利要求1所定义。
在另一优选例中,
各R 6独立地选自下组:H、氘、羟基、卤素、氰基、=O、COR 7、CO 2R 7、CONR 8R 9、CO 2NR 8R 9、SO 2R 7、SO 2NR 8R 9、NR 8SO 2R 7、NHSO 2NR 8R 9、NR 8R 9、R”’取代或未取代的含1-3个选自N、O、S的杂原子的3-10元杂环烷基、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、C3-C8环烷基、卤代C3-C8环烷基;
R”’选自下组:=O、NR’R”、C1-C4烷基、卤代C1-C4烷基;
R 7选自下组:C1-C4烷基、卤代C1-C4烷基;
R 8和R 9独立地选自下组:H、C1-C4烷基、卤代C1-C4烷基、C3-C8环烷基;或者
R 8和R 9可以与其所连接的N一起形成取代或未取代的4-6元杂环基,所述取代指被选自下组的1-3个取代基取代:=O、NR’R”、C1-C6烷基;
R’和R”独立地选自下组:H、C1-C4烷基。
在另一优选例中,p为0,且r为2。
在另一优选例中,R 4选自下组:Br、
Figure PCTCN2022097927-appb-000028
Figure PCTCN2022097927-appb-000029
Figure PCTCN2022097927-appb-000030
在另一优选例中,所述化合物选自下组:
Figure PCTCN2022097927-appb-000031
Figure PCTCN2022097927-appb-000032
Figure PCTCN2022097927-appb-000033
在另一优选例中,所述药学上可接受的盐为无机酸盐或有机酸盐;
所述无机酸盐选自下组:盐酸盐、氢溴酸盐、氢碘酸盐、硫酸盐、硫酸氢盐、硝酸盐、磷酸盐、酸式磷酸盐;
所述有机酸盐选自下组:甲酸盐、乙酸盐、三氟乙酸盐、丙酸盐、丙酮酸盐、羟乙酸盐、乙二酸盐、丙二酸盐、富马酸盐、马来酸盐、乳酸盐、苹果酸盐、柠檬酸盐、酒石酸盐、甲磺酸盐、乙磺酸盐、苯磺酸盐、水杨酸盐、苦味酸盐、谷氨酸盐、抗坏血酸盐、樟脑酸盐、樟脑磺盐、樟脑磺酸盐。
本发明的第二方面,提供了一种药物组合物,包含治疗有效量的本发明第一方面所述的化合物、或其药学上可接受的盐、立体异构体、溶剂化物或前药,以及药学上可接受的载体。
本发明的第三方面,提供了一种本发明第一方面所述的化合物、或其药学上可接受的盐、立体异构体、溶剂化物或前药的用途,用于制备CDK2/4/6激酶抑制剂药物。
本发明的第四方面,提供了一种本发明第一方面所述的化合物、或其药学上可接受的盐、立体异构体、溶剂化物或前药的用途,用于制备用于调节CDK激酶活性或治疗CDK相关疾病的药物。
在另一优选例中,所述的CDK相关疾病为癌症或肿瘤。
在另一优选例中,所述癌症或肿瘤选自下组:乳腺癌、卵巢癌、膀胱癌、子宫癌、肺癌、结直肠癌、***癌、胰腺癌、胃癌、甲状腺癌、食道癌、肾癌、肝癌、头颈癌胶质母细胞瘤、外套细胞淋巴瘤(MCL)、慢性髓细胞白血病(CML)和急性髓细胞白血病(AML)。
在另一优选例中,所述肺癌为非小细胞肺癌。
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。
附图说明
图1是xMCF-7_Palbo-R异种移植瘤模型的实验结果。
图2是OVCAR-3异种移植瘤模型的实验结果。
图3是MV4-11异种移植瘤模型的实验结果。
具体实施方式
本发明人经过长期而深入的研究,意外地制备了一种具有优异CDK激酶抑制活性的化合物及其制备方法。在此基础上,发明人完成了本发明。
术语
在本发明中,除非特别指出,所用术语具有本领域技术人员公知的一般含义。
在本发明中,术语“卤素”指F、Cl、Br或I。
在本发明中,“C1-C6烷基”是指包括1-6个碳原子的直链或支链的烷基,例如甲基、乙基、丙基、异丙基、丁基、异丁基、叔丁基、新戊基、特戊基、或类似基团。术语“C1-C4烷基”、“C1-C2烷基”具有类似含义。
在本发明中,术语“C2-C6烯基”是指具有2-6个碳原子的含有一个双键的直链或支链烯基,非限制性地包括乙烯基、丙烯基、丁烯基、异丁烯基、戊烯基和己烯基等。
在本发明中,术语“C2-C6炔基”是指具有2-6个碳原子的含有一个三键的直链或支链炔基,非限制性地包括乙炔基、丙炔基、丁炔基、异丁炔基、戊炔基和己炔基等。
在本发明中,术语“C3-C8环烷基”是指在环上具有3-8个碳原子的环状烷基,非限制性地包括环丙基、环丁基、环戊基、环己基、环庚基、环辛基等。术语“C3-C10环烷基”、“C3-C6环烷基”具有类似含义。
在本发明中,术语“C1-C6烷氧基”是指具有1-6个碳原子的直链或支链烷氧基,非限制性地包括甲氧基、乙氧基、丙氧基、异丙氧基和丁氧基等。优选为C1-C4烷氧基。
在本发明中,术语“杂环烷基”为含1、2或3个选自N、O、S的杂原子的4-8元杂环烷基,包括(但并不限于)如下基团:
Figure PCTCN2022097927-appb-000034
术语“含1-4个选自N、O、S的杂原子的3-10元杂环烷基”具有类似含义。
在本发明中,术语“芳环”或“芳基”具有相同的含义,优选为“C6-C10芳基”。术语“C6-C10芳基”是指在环上不含杂原子的具有6-10个碳原子的芳香族环基,如苯基、萘基等。类似地,术语“3-10元芳基”是指在环上不含杂原子的具有3-10个碳原子的芳香族环基,如苯基、萘基等。术语“6-10元芳基”具有类似含义。
在本发明中,术语“芳香杂环”或“杂芳基”具有相同的含义,指包含一个到多个杂原子的杂芳族基团。例如“3-10元杂芳基”是指含有1~4个选自氧、硫和氮中的杂原子以及3-10个碳原子的芳香杂环。非限制性例子包括:呋喃基、噻吩基、吡啶基、吡唑基、吡咯基、N-烷基吡咯基、嘧啶基、吡嗪基、咪唑基、四唑基等。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环。杂芳基可以是任选取代的或未取代的。
在本发明中,术语“卤代”是指被卤素取代。
在本发明中,术语“取代”指特定的基团上的一个或多个氢原子被特定的取代基所取代。特定的取代基为在前文中相应描述的取代基,或各实施例中所出现的取代基。除非特别说明,某个取代的基团可以在该基团的任何可取代的位点上具有一个选自特定组的取代基,所述的取代基在各个位置上可以是相同或不同的。本领域技术人员应理解,本发明所预期的取代基的组合是那些稳定的或化学上可实现的组合。所述取代基例如(但并不限于):卤素、羟基、羧基(-COOH)、C1-C6烷基、C2-C6烯基、C2-C6炔基、C3-C8环烷基、3-至12元杂环基、芳基、杂芳基、C1-C8醛基、C2-C10酰基、C2-C10酯基、氨基、C1-C6烷氧基、C1-C10磺酰基等。
在本发明中,术语1-6个指1、2、3、4、5或6个。其他类似术语具有类似含义。
化合物
本发明提供了式(Ⅰ)化合物、或其药学上可接受的盐、立体异构体、溶剂化物或前药,
Figure PCTCN2022097927-appb-000035
其中,各基团定义如上文所述。
在另一优选例中,所述的化合物中,R 1、R 2、R 3a、R 3b、R 4、R 5、R 6、R 7、R 8、R 9、L、R’、R”、R”’、p和r中任一个分别独立地为本发明所述具体化合物中所对应的基团。
更具体地,本发明提供了式(II)或(Ⅲ)化合物,
Figure PCTCN2022097927-appb-000036
其中,各基团定义如上文所述。
在另一优选例中,所述的化合物中,R 1’、R 2’、环A、R 31、R 41、R 51、L’、n中任一个分别独立地为本发明所述具体化合物中所对应的基团。
在另一优选例中,所述化合物优选为本发明实施例所制备化合物。
如本文所用,术语“药学上可接受的盐”指本发明化合物与酸或碱所形成的适合用作药物的盐。药学上可接受的盐包括无机盐和有机盐。一类优选的盐是本发明化合物与酸形成的盐。适合形成盐的酸包括但并不限于:盐酸、氢溴酸、氢氟酸、硫酸、硝酸、磷酸等无机酸;甲酸、乙酸、三氟乙酸、丙酸、草酸、丙二酸、琥珀酸、富马酸、马来酸、乳酸、苹果酸、酒石酸、柠檬酸、苦味酸、苯甲酸、甲磺酸、乙磺酸、对甲苯磺酸、苯磺酸、萘磺酸等有机酸;以及脯氨酸、苯丙氨酸、天冬氨酸、谷氨酸等氨基酸。
另一类优选的盐是本发明化合物与碱形成的盐,例如碱金属盐(例如钠盐或钾盐)、碱土金属盐(例如镁盐或钙盐)、铵盐(如低级的烷醇铵盐以及其它药学上可接受的胺盐),例如甲胺盐、乙胺盐、丙胺盐、二甲基胺盐、三甲基胺盐、二乙基胺盐、三乙基胺盐、叔丁基胺盐、乙二胺盐、羟乙胺盐、二羟乙胺盐、三羟乙胺盐,以及分别由吗啉、哌嗪、赖氨酸形成的胺盐。
术语“溶剂化物”指本发明化合物与溶剂分子配位形成特定比例的配合物。
术语“前药”包括其本身可以是具有生物学活性的或非活性的,当用适当的方法服用后,其在人体内进行代谢或化学反应而转变成式(I)的一类化合物,或式(I)的一个化合物所组成的盐或溶液。所述的前药包括(但不局限于)所述化合物的羧酸酯、碳酸酯、磷酸酯、硝酸酯、硫酸酯、砜酯、亚砜酯、氨基化合物、氨基甲酸盐、偶氮化合物、磷酰胺、葡萄糖苷、醚、乙缩醛等形式。
制备方法
下面更具体地描述本发明式(I)结构化合物的制备方法,但这些具体方法不对本发明构成任何限制。本发明化合物还可以任选将在本说明书中描述的或本领域已知的各种合成方法组合起来而方便地制得,这样的组合可由本发明所属领域的技术人员容易地进行。
典型地,本发明化合物的制备工艺流程如本发明实施例所示,其中所用原料和试剂如无特殊说明,均可通过商业途径购买。
药物组合物和施用方法
由于本发明化合物具有优异的抗肿瘤活性,因此本发明化合物及其各种晶型,药学上可接受的无机或有机盐,水合物或溶剂合物,以及含有本发明化合物为主要活性成分的药物组合物可用于治疗、预防以及缓解与肿瘤相关的疾病。
本发明的药物组合物包含安全有效量范围内的本发明化合物或其药理上可接受的盐及药理上可以接受的赋形剂或载体。其中“安全有效量”指的是:化合物的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有1-2000mg本发明化合物/剂,更佳地,含有10-1000mg本发明化合物/剂。较佳地,所述的“一剂”为一个胶囊或药片。
“药学上可以接受的载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份能和本发明的化合物以及它们之间相互掺和,而不明显降低化合物的药效。药学上可以接受的载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如
Figure PCTCN2022097927-appb-000037
)、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。
所述的药物组合物为注射剂、囊剂、片剂、丸剂、散剂或颗粒剂。
本发明化合物或药物组合物的施用方式没有特别限制,代表性的施用方式包括(但并不限于):口服、瘤内、直肠、肠胃外(静脉内、肌肉内或皮下)、和局部给药。
用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。在这些固体剂型中,活性化合物与至少一种常规惰性赋形剂(或载体)混合,如柠檬酸钠或磷酸二钙,或与下述成分混合:(a)填料或增容剂,例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸;(b)粘合剂,例如,羟甲基纤维素、藻酸盐、明胶、聚乙烯基吡咯烷酮、蔗糖和***胶;(c)保湿剂,例如,甘油;(d)崩解剂,例如,琼脂、碳酸钙、马铃薯淀粉或木薯淀粉、藻酸、某些复合硅酸盐、和碳酸钠;(e)缓溶剂,例如石蜡;(f)吸收加速剂,例如,季胺化合物;(g)润湿剂,例如鲸蜡醇和单硬脂酸甘油酯;(h)吸附剂,例如,高岭土;和(i)润滑剂,例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、十二烷基硫酸钠,或其混合物。胶囊剂、片剂和丸剂中,剂型也可包含缓冲剂。
固体剂型如片剂、糖丸、胶囊剂、丸剂和颗粒剂可采用包衣和壳材制备,如肠衣和其它本领域公知的材料。它们可包含不透明剂,并且,这种组合物中活性化合物或化合物的释放可以延迟的方式在消化道内的某一部分中释放。可采用的包埋组分的实例是聚合物质和蜡类物质。必要时,活性化合物也可与上述赋形剂中的一种或多种形成微胶囊形式。
用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。除了活性化合物外,液体剂型可包含本领域中常规采用的惰性稀释剂,如水或其它溶剂,增溶剂和乳化剂,例知,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、丙二醇、1,3-丁二醇、二甲基甲酰胺以及油,特别是棉籽油、花生油、玉米胚油、橄榄油、蓖麻油和芝麻油或这些物质的混合物等。
除了这些惰性稀释剂外,组合物也可包含助剂,如润湿剂、乳化剂和悬浮剂、甜味 剂、矫味剂和香料。
除了活性化合物外,悬浮液可包含悬浮剂,例如,乙氧基化异十八烷醇、聚氧乙烯山梨醇和脱水山梨醇酯、微晶纤维素、甲醇铝和琼脂或这些物质的混合物等。
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、分散液、悬浮液或乳液,和用于重新溶解成无菌的可注射溶液或分散液的无菌粉末。适宜的含水和非水载体、稀释剂、溶剂或赋形剂包括水、乙醇、多元醇及其适宜的混合物。
用于局部给药的本发明化合物的剂型包括软膏剂、散剂、贴剂、喷射剂和吸入剂。活性成分在无菌条件下与生理上可接受的载体及任何防腐剂、缓冲剂,或必要时可能需要的推进剂一起混合。
本发明化合物可以单独给药,或者与其他药学上可接受的其他化合物(如抗肿瘤药物)联合给药。
本发明治疗方法可以单独施用,或者与其它治疗手段或者治疗药物联用。
使用药物组合物时,是将安全有效量的本发明化合物适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1~2000mg,优选50~1000mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。
与现有技术相比,本发明具有以下主要优点:
(1)所述化合物具有优异的CDK激酶抑制活性;
(2)所述化合物具有优异的细胞抑制活性;
(3)所述化合物具有优异的体内药效。
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件如Sambrook等人,分子克隆:实验室手册(New York:Cold Spring Harbor Laboratory Press,1989)中所述的条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。
除非另行定义,文中所使用的所有专业与科学用语与本领域熟练人员所熟悉的意义相同。此外,任何与所记载内容相似或均等的方法及材料皆可应用于本发明方法中。文中所述的较佳实施方法与材料仅作示范之用。
实施例合成路线:
Figure PCTCN2022097927-appb-000038
起始物料S1经氢化铝锂还原得到中间体S1-1,后与各种取代的伯胺亲核取代得到产物S1-2,再经过二氧化锰氧化得醛基嘧啶化合物S1-3,再经过与含有不同取代基的乙酸酯发生环化反应,后再氧化得砜基嘧啶酮类中间体S1-5,砜基嘧啶酮中间体S1-5与取代的伯胺反应得终产物。
Figure PCTCN2022097927-appb-000039
取代的磺酰氯S2与4-Boc-氨基哌啶反应得磺酰胺中间体S2-1,后脱保护得中间体S2-3。
中间体的制备:
中间体int1的合成:
Figure PCTCN2022097927-appb-000040
1、int1-2的合成
于2000mL单口瓶中加入1-甲基环戊烯(57.0g,0.695mol)和二氯甲烷(1200mL),0℃下加入间氯过氧苯甲酸(211.0g,1.043mol),升到室温,反应16h。反应完毕,过滤,滤饼用二氯甲烷(100ml x2)洗两次,滤液用饱和的碳酸钠水溶液(300ml x3)洗三次,饱和的碳酸氢钠水溶液(300ml)洗两3次,饱和的氯化钠水溶液(300ml x2)洗两3次,无水硫酸钠干燥有机相,过滤,控温5℃以下减压蒸馏,得79g粗品,直接用于下一步反应。
2、int1-3的合成
于350mL封管中加入int1-2(40.0g,0.347mol),苄胺(74.0g,0.694mol)和乙醇(170mL),于90℃反应40h,冷却到室温,浓缩,加入水(100mL),乙酸乙酯(150mL x2)萃取两次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析得产物35g。1H NMR(400MHz,CDCl3)δ=7.36-7.31(m,4H),7.28-7.23(m,1H),3.91-3.85(m,1H),3.79-3.74(m,1H),2.86(dd,J=7.8,8.5Hz,1H),2.12-2.03(m,1H),1.75-1.53(m,5H),1.37-1.27(m,1H),1.22(s,3H)。
3、int1-4的合成
于2000mL单口瓶中加入int1-3(35.0g,0.17mol)和乙酸乙酯(350mL),加热至微微回流的条件下,滴加L-扁桃酸(13.0g,0.085mol)的乙酸乙酯(200mL)溶液,滴毕,自然降至室温,搅拌16h。过滤,用冷的乙酸乙酯洗滤饼,将滤饼加入水(150ml)和乙酸乙酯(200mL)中,用4N盐酸水溶液调PH至1,分液,水相用6N氢氧化钠水溶液调PH至11,乙酸乙酯(200mL x2)萃取,无水硫酸钠干燥有机相,过滤,浓缩,重复上述操作至ee值大于99%,得8g产物99.3%ee。1H NMR(400MHz,CDCl3)δ=7.38-7.30(m,4H),7.27-7.23(m,1H),3.94-3.75(m,2H),2.88(dd,J=7.8,8.4Hz,1H),2.16-2.03(m,1H),1.79-1.57(m,4H),1.53-1.39(m,2H),1.38-1.28(m,1H),1.25(s,3H)。手性HPLC分析方法在Agilent LC 1260使用
Figure PCTCN2022097927-appb-000041
5μm i-Amylose-1,4.6*250mm,S/N:H19-351585柱上进行,将该柱加热至30℃,在A(0.1%二乙胺/正己烷):B(0.1%二乙胺/异丙醇)=95%:5%的流动相(流速1ml/min,检测波长为254nm)下洗脱20min,产物的保留时间为7.659min
4、int1-5的合成
于1000mL单口瓶中加入int1-4(18.7g,0.091mol),Pd/C(2.0g,10%)和甲醇(350mL),氮气置换气体3次,之后氢气置换气体3次。室温反应16h。过滤,甲醇(30mL x3)洗滤 饼,浓缩,得产物10g,直接用于下一步反应。1H NMR(400MHz,CDCl3)δ=3.03(t,J=7.4Hz,1H),2.19-2.01(m,1H),1.83-1.58(m,4H),1.42(s,3H),1.35-1.25(m,1H),1.22(s,3H)。
5、int1-6的合成
于250mL单口瓶中加入[4-氯-2-(甲基硫基)嘧啶-5-基]甲醇(8.33g,43.83mmol)、int1-5(4.8g,41.74m mol)、乙醇(100mL)和N,N-二异丙基乙胺(16.15g,125.22mmol)。升温80℃反应16h。冷却到室温,浓缩,柱层析石油醚:乙酸乙酯=5:1~1:1,得产物10.5g。
6、int1的合成
于500mL单口瓶中加入int1-6(10.5g,39.03mmol),二氯甲烷(300mL)和二氧化锰(39.95g,390.3mmol)。室温搅拌16h。过滤,二氯甲烷(30mL x2)洗滤饼,浓缩有机相得产物9.3g。 1H NMR(400MHz,CDCl 3)δ=9.73(s,1H),8.66(br s,1H),8.35(s,1H),4.39(ddd,J=6.5,8.2,9.6Hz,1H),4.16(s,1H),2.57(s,3H),2.33-2.22(m,1H),2.03-1.92(m,1H),1.89-1.68(m,3H),1.68-1.56(m,1H),1.17(s,3H)。MS:268[M+H]+。
中间体int2的合成:
Figure PCTCN2022097927-appb-000042
于100mL三口瓶中加入int2-1(63g,0.379mol),二氯甲烷/水(1:1)(38mL)和二氟氢钾(29.6g,0.379mol),室温搅拌下滴加溴二氟(三甲基硅基)甲烷(154g,0.758mmol)室温下反应3h。冰浴下加入水(50ml),乙酸乙酯(30mL x2)萃取,合并有机相,无水硫酸钠干燥,过滤,浓缩有机相,柱层析得产物31.5g。
中间体int3的合成:
Figure PCTCN2022097927-appb-000043
1、int3-1的合成
将化合物int2(290mg,2.6eq)溶于干燥的THF(5ml),置换氮气,然后降温至-70℃,滴加二甲基硅基氨基锂(1.0M,2.06ml,4.0eq),滴毕,-70℃保温1h,然后滴加化合物1(137.8mg,1.0eq)的四氢呋喃(3ml)溶液,滴毕,40℃反应过夜。TLC监控反应完全,降温,向反应体系中滴加饱和氯化铵水溶液淬灭,然后加乙酸乙酯(30ml x 3)萃取,分液,收集乙酸乙酯相加无水硫酸钠干燥,过滤旋干过柱得产物(90mg)。
2、int3的合成:
将化合物int3-1(90mg,1.0eq)溶于四氢呋喃与水的混合液中,降温至0℃,然后加入Oxone(310mg,2.0eq),自然升至室温反应过夜,监控反应完成后,过滤,向滤液中加水,加乙酸乙酯萃取,干燥后旋干得化合物3(80mg)。
中间体int4的合成:
Figure PCTCN2022097927-appb-000044
1、int4-2的合成
于100mL单口瓶中加入int3-1(3.0g,13.33mmol)、4M HCl/Dioxane(30mL)和二氯甲烷 (30mL)。室温反应2h。浓缩反应液,得产物1.60g。
2、int4-3的合成
于100mL单口瓶加入int3-2(1.25g,10.0mmol),15%NaOH水溶液(10ml)和四氢呋喃(20mL)。0℃下滴加甲基磺酰氯(1.72g,15.0mmol),滴毕室温反应2h。加入水(30mL)、乙酸乙酯(20mL)萃取三次,合并有机相,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=5:1~1:1,得产物800mg。
3、int4-4的合成
于50mL单口瓶中加入int3-3(800mg,3.94mmol),氰基硼氢化钠(347mg,5.52mmol),冰醋酸(236mg,3.94mmol),对甲氧基苄胺(540mg,3.94mmol)和1,2-二氯乙烷(10mL)。室温反应3h。反应完毕,向反应液加入饱和碳酸氢钠水溶液,二氯甲烷萃取,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=5:1~0:1,得产物790mg。
4、int4的合成
于50mL单口瓶中加入int3-4(790mg,2.44mmol),Pd(OH) 2/C(80mg,10%),冰醋酸(146mg,2.44mmol)和甲醇(20mL),氮气置换气体3次,之后氢气置换气体三次。70℃反应16h。过滤,甲醇(50mL x3)洗滤饼,浓缩,得产物350mg。
中间体int5的合成:
Figure PCTCN2022097927-appb-000045
1、int5-2的合成
于100mL单口瓶中加入4-Boc-氨基哌啶(1.04g,1.0eq),二氯甲烷(10mL),N,N-二异丙基乙胺(1.05g,2.0eq),降温至0℃后滴加int5-1(1.0g,1.0eq)的二氯甲烷(2mL)溶液,自然升至室温反应3h。反应完毕,向反应液加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析二氯甲烷:甲醇=30:1,得产物1.59g。
2、int5-3的合成
于100mL单口瓶中加入int5-2(800mg,1.0eq),CD 3I(1.95g,6.0eq),K 2CO 3(1.24g,4.0eq),丙酮(10mL),65℃搅拌过夜。反应完毕,蒸干溶剂后,向反应体系中加水(15mL),加乙酸乙酯(15mL×2)萃取,无水硫酸钠干燥有机相,过滤,浓缩得产物792mg。
3、int5的合成
于100mL单口瓶中加入int5-3(720mg,1.0eq),二氯甲烷(10mL),然后室温加入盐酸/二氧六环(15mL)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得产物500mg。
中间体int6的合成:
Figure PCTCN2022097927-appb-000046
1、int6-2的合成
于500mL三口瓶中加入NaH(396.3mg,1.5eq),四氢呋喃(200mL),N 2保护,降温至0℃后加入苄硫醇(1.36g,1.0eq),搅拌10min,然后滴加C2-1(2.0g,1.05eq)的四氢呋喃(10mL)溶液,自然升至室温反应2h。反应完毕,降温至0℃,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=2:1,得2.4g。
2、int6-3的合成
于100mL单口瓶中加入int6-2(1.93g,1.0eq),二氯甲烷(20mL),降温至0℃后,向反应瓶中加水(8mL),加浓盐酸(2.0mL),然后加入次氯酸钠水溶液(10.4%,9mL), 室温反应10min,反应完成。向反应体系加水(20mL),加二氯甲烷(20mL×3)萃取,无水硫酸钠干燥有机相,过滤,将滤液降至0℃后,加入4-Boc-氨基哌啶(1.78g,1.0eq)与三乙胺(1.8g,2.0eq)的混合二氯甲烷溶液,rt反应2h。反应完成后向反应体系中加水,分液,无水硫酸钠干燥有机相,过滤,浓缩,柱层析二氯甲烷:甲醇=100:1,得1.2g。
3、int6的合成
于100mL单口瓶中加入C1-3(1.2g,1.0eq),二氯甲烷(10mL),然后室温加入盐酸/二氧六环(15mL)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得1.0g。
中间体int7的合成:
Figure PCTCN2022097927-appb-000047
1、Int7-2的合成:
于250mL单口瓶中加入Int7-1(11.9g,1.0eq),乙酸乙酯(12mL),然后室温加入HCl/dioxane(15mL)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得8.06g。
2、Int7-3的合成:
于250mL单口瓶中加入Int7-2(8.06g,1.0eq),二氯甲烷(80mL),三乙胺(12.08g,2.0eq),降温至0℃后滴加甲基磺酰氯(7.53g,1.1eq),自然升至室温反应2h。反应完毕,向反应液加入氯化铵水溶液,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,加入EA:PE约1:5打浆,过滤,收集滤饼得6.67g。
3、Int7-5的合成:
于250mL单口瓶中加入Int7-3(6.67g,1.0eq),干燥的四氢呋喃(110mL),叔丁基亚磺酰胺(5g,1.1eq),然后室温下加入钛酸四乙酯(14.5g,1.7eq),N2保护,70℃下搅拌过夜。点板监控反应完全,将反应液降温至0℃后,一次性加入NaBD4,室温反应3h。反应完全后,向反应体系加入水(80mL),有大量固体析出,过滤,残渣用EA洗涤5次后,合并滤液浓缩后用乙酸乙酯萃取3次,干燥乙酸乙酯相,过滤,柱层析二氯甲烷:甲醇=80:1得7.14g。
4、Int7的合成:
于100mL单口瓶中加入Int7-5(7.14g,1.0eq),二氯甲烷(10mL),然后室温加入15mL盐酸/二氧六环,室温反应4h。TCL监控反应完全,过滤,收集滤饼得5.88g。
中间体int8的合成:
Figure PCTCN2022097927-appb-000048
1、int8-2的合成
于100mL单口瓶中加入C3-1(4.8g,1.0eq),N,N-二甲基甲酰胺(50mL),硫代乙酸钾(8.2g,2.0eq),60℃反应过夜。反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=10:1~5:1,得4.1g。2、int8-3的合成
于100mL单口瓶中加入2M盐酸-乙腈溶液(25mL),降温至10℃以下后加入NCS(8.45g, 2.0eq),保温搅拌10min后,加入C3-2(4.1g,1.0eq)的乙腈溶液,室温反应2h。蒸走大部分乙腈后,向反应体系中加水,然后用乙酸乙酯萃取两次,加无水硫酸钠干燥乙酸乙酯相,浓缩后,加至4-Boc-氨基哌啶(4.5g,0.7eq)与三乙胺(6.4g,2.0eq)的二氯甲烷(20mL)溶液中,室温搅拌反应2h。反应完毕,向反应液加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析二氯甲烷:甲醇=30:1,得280mg。
3、int8的合成
于100mL单口瓶中加入C3-3(80mg,1.0eq),二氯甲烷(3mL),然后室温加入三氟乙酸(3mL)。室温反应2h。TLC监测反应完毕,浓缩后加乙酸乙酯超声,有固体析出,再次浓缩得62mg。
中间体int9的合成:
Figure PCTCN2022097927-appb-000049
1、int9的合成
于100mL单口瓶中加入C4-1(2.0g,1.0eq),乙醇(70mL),水合肼(8mL,10eq),雷尼镍(10mL),室温反应过夜。反应完成后,抽滤,加无水硫酸钠干燥滤液,再次抽滤后浓缩,得1.8g粗品。
中间体int10的合成:
Figure PCTCN2022097927-appb-000050
1、int10-2的合成
于50mL圆底烧瓶中加入int10-1(0.5g,2.94mmol),加入15mL二氯甲烷溶解搅拌,然后加入硫代乙酸钾(0.67g,5.88mmol),将反应移至60℃条件下搅拌4h。反应结束后,加入乙酸乙酯和饱和食盐水萃取有机相,无水硫酸钠干燥,减压浓缩,柱层析得2.36g粗产物。
2、int10-3的合成
向50mL的三口烧瓶中加入HCl:MeCN(v,5:1)2mL,降温至10℃以下,加入N-溴代丁二酰亚胺(619.2mg,4.64mmol),搅拌10-30min,然后滴加int10-2(382mg,2.32mmol)的乙腈溶液,滴毕,反应温度保持10℃左右,搅拌1-2h,反应结束后,加入二氯甲烷和饱和氯化铵水溶液萃取有机相,无水硫酸钠干燥,减压浓缩,柱层析得400mg粗产物。
3、int10-4的合成
于50mL单口瓶中加入int10-3(210mg,1.1mmol),加入6mL二氯甲烷搅拌溶解,然后加入4-Boc-氨基哌啶(109.6mg,0.55mmol),均匀搅拌后加入三乙胺(221mg,2.2mmol),室温 反应,搅拌过夜。待其充分反应后,将反应减压浓缩,加适量的石油醚、乙酸乙酯对其打浆得到319mg的白色固体粗产物。
4、int10的合成
于50mL的圆底烧瓶中加入int10(319mg,0.9mmol),加入二氯甲烷(5mL)对其搅拌溶解,将反应体系降低到0℃,然后滴加三氟乙酸(1mL),滴毕,反应2h,待其充分反应后,减压浓缩,加适量的石油醚、乙酸乙酯对其打浆得到160mg的白色固体粗产物。
中间体int11的合成:
Figure PCTCN2022097927-appb-000051
1、int11-2的合成
于100mL单口瓶中加入4-Boc-氨基哌啶(1.0g,1.0eq),二氯甲烷(10mL),三乙胺(1.01g,2.0eq),降温至0℃后滴加int11-1(1.01g,1.0eq)的二氯甲烷(2mL)溶液,自然升至室温反应2h。反应完毕,向反应液加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=5:1~1:1,得1.73g。
2、int11-3的合成
于100mL单口瓶中加入int11-2(600mg,1.0eq),二氯甲烷(30mL),氮气保护,0℃下滴加DAST(1.34g,5.0eq),滴毕,室温反应过夜。反应完毕,向反应体系中加入冰水,二氯甲烷萃取两次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=8:1~6:1,得360mg。
3、int11的合成
于100mL单口瓶中加入int11-3(480mg,1.0eq),二氯甲烷(17mL),然后室温加入盐酸/1,4-二氧六环(10mL)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得370mg。
中间体int12的合成:
Figure PCTCN2022097927-appb-000052
1、int12-2的合成
于100mL单口瓶中加入int11-2,然后加入二氯甲烷/甲醇(50mL/5mL),0℃下加入硼氢化钠(115mg,1.1eq),室温反应过夜。反应完毕,向反应体系加水淬灭,加二氯甲烷萃取两次,无水硫酸钠干燥有机相,浓缩得980mg。
2、int12-3的合成
于100mL单口瓶中加入C3-2(780mg,1.0eq),二氯甲烷(30mL),氮气保护,0℃下滴加DAST(381mg,1.1eq),滴毕,室温反应3小时。反应完毕,向反应体系中加入冰水,二氯甲烷萃取两次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=8:1~6:1,得120mg。
3、int12的合成
于100mL单口瓶中加入C3-3(160mg,1.0eq),二氯甲烷(5mL),然后室温加入盐酸/1,4-二氧六环(5mL)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得103mg。
中间体int13的合成:
Figure PCTCN2022097927-appb-000053
1、int13-2的合成
向氮气保护的250mL三口瓶中加入int13-1(6.8g,1.0eq),DPPA(25.0g,1.5eq),三乙胺(30.6g,5.0eq),1,4-二氧六环(68mL),100℃反应2小时后,降温至40℃~50℃,滴加苄醇(13.1g,2.0eq),滴毕,升温至100℃反应16小时。监控反应完全,降至室温后,向反应体系中加入饱和食盐水,用乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=10:1得9.8g。
2、int13-3的合成
向氮气保护的三口瓶中加入int13-2(9.8g,1.0eq)与超干四氢呋喃(98mL),0℃下滴加1.0M硼烷四氢呋喃(88.2mL),滴毕,保温1小时后,室温反应过夜。监控反应完成,降温至0℃向反应体系中加入水(19.6mL),10%氢氧化钠水溶液(68.7mL),然后滴加30%双氧水(49mL),室温反应5小时。监控反应完毕,向反应体系中加入乙酸乙酯萃取2次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=10:1得4.8g。
3、int13-4的合成
于100mL单口瓶中加入C5-3(3g,1.0eq),二氯甲烷(92mL),氮气保护,-20℃下滴加DAST(4.1g,2.0eq),滴毕,室温反应3小时。反应完毕,向反应体系中加入冰水,二氯甲烷萃取两次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=10:1,得580mg。
4、int13的合成
将C5-4(660mg,1.0eq)溶于甲醇(5mL),加入Pd/C(13.6mg,10%),氮气保护,置换氢气三次,室温反应过夜。监控反应完成,过滤加入盐酸/1,4-二氧六环(2mL),浓缩得360mg。
中间体int14的合成:
Figure PCTCN2022097927-appb-000054
1、int14-2的合成
向三口瓶中加入int14-1(1.86g,1.0eq),二氯甲烷(74mL),降温至0℃后分三批加入PCC(3.4g,2.0eq),氮气保护,室温反应48h。监控反应完毕,向反应体系中加入饱和食盐水,过滤,滤液用二氯甲烷萃取2次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=5:1得1.62g。
2、int14-3的合成
于100mL单口瓶中加入C6-2(1.62g,1.0eq),二氯甲烷(83mL),氮气保护,0℃下滴加DAST(5.5g,5.0eq),滴毕,室温反应过夜。反应完毕,向反应体系中加入冰水,二氯甲烷萃取两次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=10:1,得910mg。
3、int14的合成
将C6-3(910mg,1.0eq)溶于甲醇(5mL),加入Pd/C(91mg,10%),氮气保护,置换氢气三次,室温反应过夜。监控反应完成,过滤加入盐酸/1,4-二氧六环(5mL),浓缩得650mg。
中间体int15的合成:
Figure PCTCN2022097927-appb-000055
1、int15-2的合成
将乙二醛-1,1-二甲基乙缩醛(500mg,1.0eq),int15-1(1.48g,1.0eq),甲醇(10mL),置于 单口瓶中,室温反应2小时。监控反应完毕,向反应体系中加入N-Boc-反式-1,4-环己二胺(1.11g,1.1eq),乙酸(288mg,1.0eq),升温至75℃反应过夜。监控反应完成,向反应体系加碳酸氢钠水溶液,调PH>7,然后加水,加乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=2:1,得320mg。
2、int15的合成
于100mL单口瓶中加入C7-2(320mg,1.0eq),二氯甲烷(3mL),然后室温加入盐酸/1,4-二氧六环(5mL)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得240mg。
中间体int16的合成:
Figure PCTCN2022097927-appb-000056
1、int16-2的合成:
于250mL单口瓶中加入int16-1(2g,1.0eq),BH 3/THF(1M 34Ml 3eq),然后室温加入四氢呋喃(80mL)回流反应过夜。TLC监测反应完毕,降温至室温后滴加1M HCl 100ml后,用乙酸乙酯萃取5次后水相降温至零度,用饱和碳酸氢钠溶液调至中性后再次用乙酸乙酯萃取,有机相干燥浓缩得产品690mg。
2、int16-3的合成:
于100mL单口瓶中加入int16-2(690mg,1.0eq),二氯甲烷(30mL),三乙胺(1.27g,3eq),降温至0℃后滴加甲基磺酰氯(723mg,1.5eq),自然升至室温反应2h。反应完毕,向反应液加入氯化铵水溶液,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析二氯甲烷:甲醇=200:1-100:1得640mg。
3、int16的合成:
于100mL三口瓶中加入int16-3(640mg,1.0eq),乙醇(64mL),Pd/C(64mg)氢气氛围下50℃反应2h,反应完毕,过滤,浓缩,得165mg。
中间体int17的合成:
Figure PCTCN2022097927-appb-000057
1、int17-2的合成
于50mL单口瓶中加入3-{[(叔丁氧基)羰基]氨基}双环[1.1.1]戊烷-1-甲酸甲酯(560mg,2.32mmol)和甲醇(5mL),室温把水合肼滴加到反应混合液中,加毕,室温搅拌30分钟,加热至80℃反应2-3个小时。LC-MS显示反应完毕,减压浓缩的粗品,粗品用甲苯带水2次后,得白色固体(562mg)。未进一步纯化直接用于下一步。
2、int17-3的合成
于50mL单口瓶中把C8-1(562mg,2.32mmol)和TsOH(40mg,0.232mmol)加入到原甲酸三乙酯(8mL)中,加热至80℃反应4小时。LC-MS显示反应完毕,降至50℃,减压浓缩得粗品,粗品柱层析(PE-PE:EA=2:1)得白色固体465mg。
3、int17的合成
于50mL单口瓶加入C8-2(465mg,1.852mmol)和二氯甲烷(5mL),冰浴条件下降温至0℃,向反应液中滴加三氟醋酸(1mL),滴毕,自然升至室温反应4小时。TLC显示反应完毕,减压浓缩得粗品,粗品用乙酸乙酯(20mL)打浆,过滤得白色固体(347mg)。
实施例1
本发明合成的化合物:
Figure PCTCN2022097927-appb-000058
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000059
1、C1-1的合成
于50mL单口瓶中加入2-(氧杂环丁烷-3-基)乙酸乙酯(200mg,1.54mmol)和干燥四氢呋喃,-78℃下滴加LiHMDS(2.15mmol),滴毕-78℃反应30min,之后滴加int1(165mg,1.54mmol)的四氢呋喃溶液,滴毕缓慢升至室温反应16h。反应完毕,加入饱和氯化铵,用乙酸乙酯萃取,无水硫酸钠干燥有机相,过滤,浓缩,石油醚:乙酸乙酯=10:1~5:1过柱得60mg。
2、C1-2的合成
于50mL单口瓶中加入C2-1(55mg,0.16mol)、四氢呋喃(5mL)和水(1mL),降温0℃,分批加入OXONE(240mg,0.40mol)。室温反应2h,反应完毕,过滤,滤饼用EA洗两次,收集母液,浓缩,得50mg。
3、C1的合成
于50mL单口瓶中加入C2-2(50mg,0.12mmol),1-甲砜基-4-氨基哌啶(32mg,0.15mmol),N,N-二异丙基乙胺(81mg,0.3mmol)和二甲基亚砜(5mL)。升温60℃反应16h。反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,石油醚:乙酸乙酯=1:5制备板分离得产物15mg。 1H NMR(400MHz,CDCl 3)δ8.47(s,1H),7.46(d,J=1.4Hz,1H),5.74(t,J=8.5Hz,1H),5.35(s,2H),5.06(ddd,J=8.5,6.0,2.6Hz,2H),4.70(ddd,J=11.1,7.0,6.0Hz,2H),4.43–4.28(m,1H),3.98(s,1H),3.90–3.73(m,2H),3.00–2.86(m,2H),2.82(s,3H),2.30–2.13(m,3H),2.07–1.95(m,2H),1.95–1.76(m,2H),1.76–1.58(m,2H),1.13(s,3H)。
实施例2
本发明合成的化合物:
Figure PCTCN2022097927-appb-000060
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000061
1、C2的合成
于50mL单口瓶中加入int3(396mg,1.23mmol),int4(300mg,1.47mmol),N,N-二异丙基乙胺(475mg,3.68mmol)和二甲基亚砜(5mL)。升温60℃反应16h。反应完毕,向反应液加入水,EA萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=5:1~0:1,得产物370mg。
实施例3
本发明合成的化合物:
Figure PCTCN2022097927-appb-000062
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000063
C3的合成
将化合物int3(80mg)溶于DMSO,加入1-甲砜基-4-氨基哌啶(88.4mg,2.0eq),三乙胺(0.11ml,3.0eq),60℃下反应过夜。监控反应完成,向体系中加水,然后加乙酸乙酯萃取,合并有机相,无水硫酸钠干燥,减压浓缩,制备板分离得到产物(50mg)。 1H NMR(400MHz,CDCl 3)δ8.45(s,1H),7.37(s,1H),6.77(t,J=75.0Hz,1H),5.81(s,1H),5.44(m,1H),3.98(br,1H),3.82(t,J=12.1Hz,2H),2.99–2.87(m,2H),2.83(s,3H),2.74(br,1H),2.35–2.16(m,3H),2.04(d,J=7.1Hz,3H),1.93(t,J=9.5Hz,1H),1.82(dd,J=12.0,6.4Hz,1H),1.67(td,J=12.9,12.0,4.0Hz,2H),1.17(s,3H)。
实施例4
本发明合成的化合物:
Figure PCTCN2022097927-appb-000064
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000065
C4-1的合成
在100mL三口瓶中加入四氢呋喃(10mL),降温至-5℃,滴加四氯化钛(3.04g,16mmol)的二氯甲烷(5mL)溶液。滴毕搅拌10分钟,向反应液中滴加int1(1.07g,4mmol)和丙二酸二乙酯(1.28g,8mmol)的四氢呋喃(10mL)溶液,滴毕搅拌30分钟后,滴加吡啶(1.58g,20mmol),滴毕自然升至室温搅拌过夜。TLC监测反应完毕。加入水稀释,乙酸乙酯萃取,无水硫酸钠干燥,过滤,浓缩,柱层析石油醚:乙酸乙酯=5:1~3:1,得产物1.31g。
C4-2的合成
于50mL单口瓶加入C4-1(1.31g,3.20mmol)和四氢呋喃(15mL),室温滴加叔丁醇钾(36mg,0.32mmol)的四氢呋喃(1mL)溶液,室温搅拌15分钟。TLC监测反应完毕,柱层析石油醚:乙酸乙酯=3:1~1:1,得1.02g。
C4-3的合成
于50mL单口瓶加入C4-2(1.01g,2.78mmol),氢氧化锂(200mg,8.35mmol)四氢呋喃(10mL)和水(10mL),室温搅拌4小时。TLC监测反应完毕,加入水稀释,乙酸乙酯萃取,无水硫酸钠干燥,过滤,浓缩,得粗品0.98g。
C4-4的合成
于50mL单口瓶加入C4-3(335mg,1.00mmol),叠氮磷酸二苯酯(330mg,1.20mmol),三乙胺(122mg,1.20mmol)和叔丁醇(5mL),加热至78℃反应16小时。TLC监测反应完毕,加入水稀释,乙酸乙酯萃取,无水硫酸钠干燥,过滤,浓缩,柱层析石油醚:乙酸乙酯=4:1~2:1,得83mg。
C4-5的合成
0℃条件下,把氢化钠(5.6mg,0.139mmol)加入到C4-4(47mg,0.116mmol)的N,N-二甲基甲酰胺(3ml)溶液中,加毕自然升至室温反应30分钟,然后降温至0℃,滴加碘甲烷到反应液中,滴毕升至室温反应2小时。TLC监测反应完毕,加入水稀释,乙酸乙酯萃取,无水硫酸钠干燥,过滤,浓缩,柱层析得48mg。
C4-6的合成
0℃条件下,把OXONE(160mg,0.268mmol)加入到C4-5(45mg,0.107mmol)的四氢呋喃和水的混合溶液中,加毕自然升至室温反应2小时。TLC监测反应完毕,加入水稀释,乙酸乙酯萃取,无水硫酸钠干燥,过滤,浓缩,得粗品51mg。
C4-7的合成
于50mL单口瓶加入C4-6(51mg,0.113mmol),1-甲砜基-4-氨基哌啶(49mg,0.226mmol),二异丙基乙胺(59mg,0.452mmol)和二甲基亚砜(2mL),加热至60℃反应4小时。TLC监测反应完毕,加入水稀释,乙酸乙酯萃取,无水硫酸钠干燥,过滤,浓缩,柱层析得45mg。C4的合成
0℃条件下,把甲基磺酸(78mg,0.82mmol)滴加到C4-7(45mg,0.082mmol)的二氯甲烷(3mL)溶液中,滴毕,自然升至室温反应1小时。TLC监测反应完毕,加入水稀释,乙酸乙酯萃取,无水硫酸钠干燥,过滤,浓缩,柱层析得25mg。 1H NMR(400MHz,CDCl 3)δ8.39(s,1H),6.26(s,1H),5.88(s,1H),5.22(s,1H),4.98(s,1H),3.95(s,1H),3.78(s,2H),2.95(s,2H),2.89(d,J=4.3Hz,3H),2.83(s,3H),2.72(s,1H),2.26–2.13(m,3H),2.02(s,1H),1.93–1.78(m,3H),1.59(m,3H),1.34(s,3H).
实施例5
本发明合成的化合物:
Figure PCTCN2022097927-appb-000066
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000067
C5-2的合成
于100mL单口瓶中加入4-Boc-氨基哌啶(1.0g,1.0eq),二氯甲烷(10ml),三乙胺(1.01g,2.0eq),降温至0℃后滴加C1-1(1.28g,1.0eq)的二氯甲烷(2ml)溶液,自然升至室温反应2h。反应完毕,向反应液加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=5:1~1:1,得1.83g。
C5-3的合成
于100mL单口瓶中加入C5-2(1.83g,1.0eq),二氯甲烷(18ml),然后室温加入HCl/dioxane(10ml)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得1.7g。
C5的合成
于50mL单口瓶中加入C5-3(74.3mg,1.2eq)、int3(70mg,1.0eq)、二异丙基乙胺(70mg,3.0eq)、二甲基亚砜(3ml)。60℃反应过夜。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC得24mg。MS[M+1]:618.6  1H NMR(400MHz,CDCl 3)δ8.38(d,J=14.1Hz,1H),7.92(d,J=8.2Hz,2H),7.83(d,J=8.3Hz,2H),7.33(s,1H),6.75(t,J=75.1Hz,1H),5.76(t,J=8.4Hz,1H),5.64–5.27(m,1H),3.77(t,J=43.4Hz,3H),3.06–2.43(m,3H),2.36–2.09(m,4H),2.07–1.66(m,6H),1.14(s,3H).
实施例6
本发明合成的化合物:
Figure PCTCN2022097927-appb-000068
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000069
C6-2的合成
于100mL单口瓶中加入4-Boc-氨基哌啶(1.0g,1.0eq),二氯甲烷(10ml),三乙胺(1.01g,2.0eq),降温至0℃后滴加C2-1(1.058g,1.0eq)的二氯甲烷(2ml)溶液,自然升至室温反应2h。反应完毕,向反应液加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=5:1~1:1,得1.74g。
C6-3的合成
于100mL单口瓶中加入C6-2(1.74g,1.0eq),二氯甲烷(17ml),然后室温加入HCl/dioxane(10ml)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得1.5g。
C6的合成
于50mL单口瓶中加入C6-3(67mg,1.2eq)、int3(70mg,1.0eq)、二异丙基乙胺(70mg,3.0eq)、二甲基亚砜(3ml)。60℃反应过夜。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC得34mg。 1H NMR(400MHz,CDCl 3)δ8.43(s,1H),7.36(s,1H),6.82(t,J=75.3Hz,1H),5.73(s,1H),5.32(m,J=62.4,53.3Hz,2H),4.37–4.12(m,1H),3.80(d,J=10.9Hz,3H),2.93(m,J=25.6,14.7Hz,4H),2.51–2.02(m,7H),1.98–1.65(m,4H),1.26(s,J=6.6Hz,3H).
MS[M+1]:575.6
实施例7
本发明合成的化合物:
Figure PCTCN2022097927-appb-000070
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000071
C7-2的合成
于100mL单口瓶中加入4-Boc-氨基哌啶(1.0g,1.0eq),二氯甲烷(10ml),三乙胺(1.01g,2.0eq),降温至0℃后滴加C3-1(1.06g,1.0eq),自然升至室温反应2h。反应完毕,向反应液加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=5:1~1:1,得1.89g。
C7-3的合成
于100mL单口瓶中加入C7-2(1.74g,1.0eq),二氯甲烷(17ml),然后室温加入HCl/dioxane(10ml)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得1.51g。
C7的合成
于50mL单口瓶中加入C7-3(128.7mg,2.0eq)、int3(80mg,1.0eq)、二异丙基乙胺(105.7mg,4.0eq)、二甲基亚砜(4ml)。60℃反应过夜。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC得42mg。 1H NMR(400MHz,CDCl 3)δ8.42(s,1H),7.89(m,J=14.4,8.1Hz,1H),7.34(s,1H),7.06–6.95(m,2H),6.76(t,J=75.1Hz,1H),5.78(t,J=8.7Hz,1H),5.48(d,J=34.3Hz,1H),4.08–3.73(m,3H),3.03–2.62(m,3H),2.24(m,J=41.8,22.4,9.7Hz,3H),2.09–1.66(m,7H),1.14(s,3H).MS[M+1]:586.5。
实施例8
本发明合成的化合物:
Figure PCTCN2022097927-appb-000072
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000073
C8-2的合成
于100mL单口瓶中加入4-Boc-氨基哌啶(1.0g,1.0eq),二氯甲烷(10ml),三乙胺(1.01g,2.0eq),降温至0℃后滴加C4-1(1.0g,1.05eq)的二氯甲烷(2ml)溶液,自然升至室温反应2h。反应完毕,向反应液加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=5:1~1:1,得1.88g。
C8-3的合成
于100mL单口瓶中加入C8-2(1.88g,1.0eq),二氯甲烷(19ml),然后室温加入HCl/dioxane(13ml)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得1.44g。
C8的合成
于50mL单口瓶中加入C8-3(149.5mg,2.0eq)、int3(100mg,1.0eq)、二异丙基乙胺(105.7mg,4.0eq)、二甲基亚砜(5ml)。60℃反应过夜。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC得72mg。 1H NMR(400MHz,CDCl 3)δ8.40(s,1H),7.66(d,J=8.2Hz,2H),7.39–7.30(m,3H),6.75(t,J=75.1Hz,1H),5.76(t,J=8.5Hz,1H),5.45(d,J=54.7Hz,1H),3.94–3.57(m,3H),2.79–2.49(m,3H),2.46(s,3H),2.31–2.07(m,3H),2.06–1.67(m,7H),1.13(s,3H).MS[M+1]:564.6。
实施例9
本发明合成的化合物:
Figure PCTCN2022097927-appb-000074
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000075
C9-2的合成
于50mL单口瓶中加入4-Boc-氨基哌啶(300mg,1.0eq),二氯甲烷(6ml),三乙胺(303mg,2.0eq),降温至0℃后滴加C5-1(303mg,1.01eq)的二氯甲烷(2ml)溶液,自然升至室温反应2h。反应完毕,向反应液加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=5:1~1:1,得550mg。
C9-3的合成
于50mL单口瓶中加入C9-2(550mg,1.0eq),二氯甲烷(8ml),然后室温加入HCl/dioxane(6ml)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得443mg。
C9的合成
于50mL单口瓶中加入C9-3(93mg,1.5eq)、int3(80mg,1.0eq)、二异丙基乙胺(79.3mg,3.0eq)、二甲基亚砜(5ml)。60℃反应过夜。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC得39mg。 1H NMR(400MHz,CDCl 3)δ8.42(s,1H),8.08(d,J=7.8Hz,1H),7.90(d,J=7.5Hz,1H),7.75(dt,J=21.6,7.5Hz,2H),7.34(s,1H),6.76(t,J=75.1Hz,1H),5.79(t,J=8.5Hz,1H),5.51(s,1H),3.92(d,J=13.1Hz,3H),2.90(d,J=54.2Hz,2H),2.70(dd,J=19.2,8.6Hz,1H),2.24(m,J=36.2,20.3,9.9Hz,4H),2.07–1.68(m,6H),1.14(s,3H).
MS[M+1]:575.6。
实施例10
本发明合成的化合物:
Figure PCTCN2022097927-appb-000076
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000077
C10-2的合成
于50mL单口瓶中加入4-Boc-氨基哌啶(300mg,1.0eq),二氯甲烷(6ml),三乙胺(303mg,2.0eq),降温至0℃后滴加C6-1(313mg,1.01eq),自然升至室温反应2h。反应完毕,向反应液加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析PE:EA=5:1~1:1,得530mg。
C10-3的合成
于50mL单口瓶中加入C10-2(530mg,1.0eq),二氯甲烷(8ml),然后室温加入HCl/dioxane(6ml)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得380mg。
C10的合成
于50mL单口瓶中加入C10-3(94.5mg,1.5eq)、int3(80mg,1.0eq)、二异丙基乙胺(79.3mg,3.0eq)、二甲基亚砜(5ml)。60℃反应过夜。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC得38mg。 1H NMR(400MHz,CDCl 3)δ8.40(s,1H),7.46(t,J=8.0Hz,1H),7.40–7.30(m,2H),7.28(s,1H),7.15(dd,J=8.2,1.9Hz,1H),6.75(t,J=75.1Hz,1H),5.76(t,J=8.5Hz,1H),5.36(d,J=45.1Hz,1H),3.94–3.63(m,6H),2.71(s,3H),2.33–2.08(m,3H),2.07–1.65(m,7H),1.14(s,3H).MS[M+1]:580.6。
实施例11
本发明合成的化合物:
Figure PCTCN2022097927-appb-000078
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000079
C11-2的合成
于50mL单口瓶中加入4-Boc-氨基哌啶(300mg,1.0eq),DCM(6ml),TEA(303mg,2.0eq),降温至0℃后滴加C7-1(385.8mg,1.01eq)的DCM(3.0ml)溶液,自然升至室温反应2h。反应完毕,向反应液加入水,DCM萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析PE:EA=5:1~1:1,得500mg。
C11-3的合成
于50mL单口瓶中加入C7-2(500mg,1.0eq),DCM(8ml),然后室温加入HCl/dioxane(6ml)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得383mg。
C11的合成
于50mL单口瓶中加入C11-3(109.3mg,1.5eq)、INT(80mg,1.0eq)、DIPEA(79.3mg,3.0eq)、DMSO(5ml)。60℃反应过夜。LC-MS监测反应完毕,向反应液加入水,EA萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC得29mg。 1H NMR(400MHz,CDCl 3)δ8.40(s,1H),8.14(d,J=8.3Hz,2H),7.99(d,J=8.3Hz,2H),7.33(s,1H),6.75(t,J=75.1Hz,1H),5.77(t,J=8.5Hz,1H),5.47(s,1H),3.83(d,J=38.5Hz,3H),3.14(s,3H),2.66 (dt,J=41.4,21.1Hz,3H),2.24(m,J=37.0,20.3,9.8Hz,3H),2.07–1.67(m,7H),1.13(s,3H).MS[M+1]:628.8。
实施例12
本发明合成的化合物:
Figure PCTCN2022097927-appb-000080
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000081
C12-2的合成
于100mL单口瓶中加入4-Boc-氨基哌啶(1.0g,1.0eq),二氯甲烷(10ml),三乙胺(1.01g,2.0eq),降温至0℃后滴加C8-1(1.18g,1.0eq)的二氯甲烷(2ml)溶液,自然升至室温反应2h。反应完毕,向反应液加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=5:1~1:1,得1.64g。
C12-3的合成
于100mL单口瓶中加入C12-2(1.64g,1.0eq),二氯甲烷(16ml),然后室温加入HCl/dioxane(10ml)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得1.32g。
C12的合成
于50mL单口瓶中加入C12-3(120mg,2.0eq)、int3(80mg,1.0eq)、二异丙基乙胺(80mg,3.0eq)、二甲基亚砜(3ml)。60℃反应过夜。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC得42mg。 1H NMR(400MHz,CDCl 3)δ8.41(s,1H),7.80(m,J=7.9,5.0,2.4Hz,2H),7.34(s,1H),7.26–7.20(m,2H),6.75(t,J=75.0Hz,1H),5.76(t,J=8.5Hz,1H),5.40(s,1H),3.81(d,J=37.9Hz,3H),2.62(d,J=63.5Hz,3H),2.41–2.08(m,4H),2.08–1.64(m,6H),1.15(d,J=12.6Hz,3H).MS[M+1]:568.5。
实施例13
本发明合成的化合物:
Figure PCTCN2022097927-appb-000082
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000083
C13-2的合成
于100mL单口瓶中加入4-Boc-氨基哌啶(1.0g,1.0eq),二氯甲烷(10ml),三乙胺(1.01g,2.0eq),降温至0℃后滴加C9-1(1.0g,1.05eq)的二氯甲烷(2ml)溶液,自然升至室温反应2h。反应完毕,向反应液加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析PE:EA=5:1~1:1,得1.74g。
C13-3的合成
于100mL单口瓶中加入C9-2(1.74g,1.0eq),二氯甲烷(17ml),然后室温加入HCl/dioxane(10ml)。室温反应2h。TLC监测反应完毕,过滤,收集滤饼得1.31g。
C13的合成
于50mL单口瓶中加入C9-3(149.5mg,2.0eq)、int3(100mg,1.0eq)、二异丙基乙胺(105.7mg,4.0eq)、二甲基亚砜(5ml)。60℃反应过夜。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC得63mg。 1H NMR(400MHz,CDCl 3)δ8.43(s,1H),7.58(d,J=4.9Hz,2H),7.45(t,J=6.8Hz,2H),7.38(s,1H),6.74(t,J=75.2Hz,1H),5.87–5.40(m,2H),3.75(d,J=72.5Hz,3H),2.59(d,J=61.7Hz,3H),2.46(s,3H),2.21–1.94(m,4H),1.75(d,J=61.1Hz,6H),1.21(d,J=32.5Hz,3H).MS[M+1]:564.6。
实施例14
本发明合成的化合物:
Figure PCTCN2022097927-appb-000084
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000085
C14-2的合成
于50mL单口瓶中加入C14-1(500mg,2.4mmol),加入10mL二氯甲烷搅拌溶解,然后加入4-Boc-氨基哌啶(480mg,2.4mmol),均匀搅拌后加入三乙胺(485mg,4.8mmol),室温反应,搅拌过夜。待其充分反应后,将反应减压浓缩,加适量的石油醚/乙酸乙酯打浆得到942mg的白色固体产物。
C14-3的合成
于100mL的圆底烧瓶中加入C14-2(942mg,2.54mmol),加入二氯甲烷(20mL)对其搅拌溶解,然后滴加HCl/Dioxane(6mL,24mmol),滴毕,反应2h,TLC检测反应完全。减压浓缩,加适量的石油醚/乙酸乙酯打浆得到730mg产物。
C14的合成
于10mL封管中加入int3(60mg,0.154mmol),然后加入二甲基亚砜(3mL),搅拌均匀后,加入C14-3(94.4mg,0.308mmol),升温至60℃,搅拌过夜。将反应冷却至室温,加入适量的水和乙酸乙酯萃取有机相,无水硫酸钠干燥有机相,减压浓缩,柱层析(石油醚:乙酸乙酯=1:1),得61mg,然后对其进行打浆纯化得37mg。
实施例15
本发明合成的化合物:
Figure PCTCN2022097927-appb-000086
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000087
C15-2的合成:
于100mL单口瓶中加入C15-1(1g,4.7mmol),三乙胺(1.25mL,9.4mmol)和二氯甲烷(5mL),0℃下滴加4-Boc-氨基哌啶(0.94g,4.7mmol)的二氯甲烷(5mL)溶液,升至室温后反应2h。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,得1.73g。MS[M+1]:377
C15-3的合成:
于100mL单口瓶中加入C15-2(1.73g,4.6mmol)和二氯甲烷溶液(20mL),搅拌条件下室温滴加HCl/dioxane(10mL),滴毕反应2h。LC-MS监测反应完毕,浓缩反应液,加入少量乙酸乙酯和大量石油醚进行超声、洗涤,后过滤干燥得粗产品1.47g。MS[M+1]:277
C15的合成:
于50mL单口瓶中加入C15-3(131mg,0.42mmol),int3(80mg,0.21mmol),二异丙基乙胺(0.15mL,0.84mmol)和二甲基亚砜(3mL)。升温60℃反应16h。反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC(展开剂为二氯甲烷:甲醇=60:1)纯化得69mg。MS[M+1]:567 1H NMR(400MHz,CDCl 3)δ8.41(s,1H),7.32(d,J=7.3Hz,3H),7.08(t,J=8.6Hz,1H),6.76(s,1H),5.78(t,J=8.5Hz,1H),5.36(s,1H),3.76(m,3H),2.70(m,3H),2.17(d,J=11.7Hz,4H),2.01(d,J=11.5Hz,3H),1.80(m,3H),1.14(s,3H).
实施例16
本发明合成的化合物:
Figure PCTCN2022097927-appb-000088
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000089
C16-2的合成:
于100mL单口瓶中加入C16-1(883mg,5mmol),三乙胺(1.38mL,10mmol)和二氯甲烷(5mL),0℃下滴加4-Boc-氨基哌啶(1g,5mmol)的二氯甲烷(5mL)溶液,升至室温后反应2h。LC-MS监测反应完毕,向反应液加入水,EA萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,得1.64g。MS[M+1]:340
C16-3的合成:
于100mL单口瓶中加入C16-2(1.64g,4.8mmol)和二氯甲烷溶液(10mL),搅拌条件下室温滴加HCl/dioxane(10mL),滴毕反应2h。LC-MS监测反应完毕,浓缩反应液,加入少量乙酸乙酯和大量石油醚进行超声、洗涤,后过滤干燥得粗产品1.32g。MS[M+1]:240
C16的合成:
于50mL单口瓶中加入C16-3(143.5mg,0.52mmol),int3(100mg,0.26mmol),二异丙基乙胺(0.18mL,1.04mmol)和二甲基亚砜(3mL)。升温60℃反应16h。反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC(展开剂为二氯甲烷:甲醇=60:1)纯化得67mg。MS[M+1]:548 1H NMR(400MHz,CDCl 3)δ8.39 (s,1H),7.79(d,J=8.0Hz,2H),7.64(t,J=7.4Hz,1H),7.56(t,J=7.7Hz,2H),7.33(s,1H),6.75(s,1H),5.76(t,J=8.6Hz,1H),5.30(s,1H),3.79(d,J=34.0Hz,3H),2.70(m,3H),2.06(dd,J=62.3,10.2Hz,10H),1.13(s,3H).
实施例17
本发明合成的化合物:
Figure PCTCN2022097927-appb-000090
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000091
C17-2的合成:
于100mL单口瓶中加入C17-1(1g,5mmol),三乙胺(2mL,10mmol)和二氯甲烷(5mL),0℃下滴加4-Boc-氨基哌啶(1g,5mmol)的二氯甲烷(5mL)溶液,升至室温后反应2h。LC-MS监测反应完毕,向反应液加入水,EA萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,得1.8g。MS[M+1]:259
C17-3的合成:
于100mL单口瓶中加入C17-2(1.8g,5mmol)和二氯甲烷溶液(18mL),搅拌条件下室温滴加HCl/dioxane(10mL),滴毕反应2h。LC-MS监测反应完毕,浓缩反应液,加入少量乙酸乙酯和大量石油醚进行超声、洗涤,后过滤干燥得粗产品1.57g。MS[M+1]:259 C17的合成:
于50mL单口瓶中加入C17-3(118mg,0.40mmol),int3(77mg,0.20mmol),二异丙基乙胺(0.14mL,0.80mmol)和二甲基亚砜(3mL)。升温60℃反应16h。反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC(展开剂为二氯甲烷:甲醇=60:1)纯化得29mg。MS[M+1]:604  1H NMR(400MHz,CDCl 3)δ8.40(s,1H),7.62–7.46(m,3H),7.33(s,2H),6.75(s,1H),5.77(t,J=8.5Hz,1H),5.40(s,1H),3.82(d,J=45.3Hz,3H),2.70(s,3H),2.15(d,J=12.9Hz,3H),2.09–1.84(m,4H),1.78(s,3H),1.14(s,3H).
实施例18
本发明合成的化合物:
Figure PCTCN2022097927-appb-000092
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000093
C18-2的合成:
于100mL单口瓶中加入C18-1(1.06g,5mmol),三乙胺(1.38mL,10mmol)和二氯甲烷(5mL),0℃下滴加4-Boc-氨基哌啶(1g,5mmol)的二氯甲烷(5mL)溶液,升至室温后反应2h。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,得1.69g。MS[M+1]:377
C18-3的合成:
于100mL单口瓶中加入C18-2(1.69g,4.5mmol)和乙酸乙酯溶液(20mL),搅拌条件下室温滴加HCl/dioxane(10mL),滴毕反应2h。LC-MS监测反应完毕,浓缩反应液,加入少量乙酸乙酯和大量石油醚进行超声、洗涤,后过滤干燥得粗产品1.57g。MS[M+1]:277
C18的合成:
于50mL单口瓶中加入C18-3(162mg,0.52mmol),int(100mg,0.26mmol),二异丙基乙胺(0.18mL,1.04mmol)和二甲基亚砜(3mL)。升温60℃反应16h。反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC(展开剂为二氯甲烷:甲醇=60:1)纯化得55mg。MS[M+1]:567 1H NMR(400MHz,CDCl 3)δ8.34(s,1H),7.56(t,J=8.2Hz,1H),7.50(d,J=8.8Hz,1H),7.35–7.24(m,2H),6.75(s,1H),5.70(t,J=8.6Hz,1H),5.35(s,1H),3.74(d,J=53.6Hz,3H),2.63(s,3H),2.28–2.04(m,4H),2.03–1.89(m,3H),1.89–1.71(m,3H),1.07(s,3H).
实施例19
本发明合成的化合物:
Figure PCTCN2022097927-appb-000094
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000095
C19-2的合成:
于100mL单口瓶中加入C19-1(1g,4.7mmol),三乙胺(1.25mL,9.4mmol)和二氯甲烷(5mL),0℃下滴加4-Boc-氨基哌啶(0.94g,4.7mmol)的二氯甲烷(5mL)溶液,升至室温后反应2h。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,得2.35g。MS[M+1]:377
C19-3的合成:
于100mL单口瓶中加入C19-2(2.35g,6.2mmol)和二氯甲烷溶液(20mL),搅拌条件下室温滴加HCl/dioxane(10mL),滴毕反应2h。LC-MS监测反应完毕,浓缩反应液,加入少量乙酸乙酯和大量石油醚进行超声、洗涤,后过滤干燥得粗产品1.47g。MS[M+1]:277
C19的合成:
于50mL单口瓶中加入C19-3(162mg,0.52mmol),int3(100mg,0.26mmol),二异丙基乙胺(0.18mL,1.04mmol)和二甲基亚砜(3mL)。升温60℃反应16h。反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC(展开剂为二氯甲烷:甲醇=60:1)纯化得93mg。MS[M+1]:567 1H NMR(400MHz,CDCl 3)δ8.35(s,1H),7.48(m,1H),7.27(s,1H),6.98(t,J=8.9Hz,2H),6.75(s,1H),5.71(d,J=8.7Hz,1H),5.01(s,1H),3.88(d,J=11.9Hz,3H),2.87(s,2H),2.64(s,1H),2.11(d,J=10.3Hz,3H),1.96(d,J=14.7Hz,4H),1.72(s,3H),1.08(s,3H).
实施例20
本发明合成的化合物:
Figure PCTCN2022097927-appb-000096
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000097
C20-2的合成:
于100mL单口瓶中加入C20-1(0.5g,2.04mmol),三乙胺(0.54mL,4.08mmol)和二氯甲烷(5mL),0℃下滴加4-Boc-氨基哌啶(0.4g,2.04mmol)的二氯甲烷(5mL)溶液,升至室温后反应2h。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,得850mg。MS[M+1]:409
C20-3的合成:
于100mL单口瓶中加入C20-2(850mg,2.08mmol)和二氯甲烷溶液(10mL),搅拌条件下室温滴加HCl/dioxane(3.8mL),滴毕反应2h。LC-MS监测反应完毕,浓缩反应液,加入少量乙酸乙酯和大量石油醚进行超声、洗涤,后过滤干燥得粗产品692mg。MS[M+1]:345C20的合成:
于50mL单口瓶中加入C20-3(144.5mg,0.42mmol),int(80mg,0.21mmol),二异丙基乙胺(0.15mL,0.84mmol)和二甲基亚砜(2mL)。升温60℃反应16h。反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC(展开剂为二氯甲烷:甲醇=60:1)纯化得83mg。MS[M+1]:618 1H NMR(400MHz,CDCl 3)δ8.40(s,1H),8.04(s,1H),7.98(d,J=7.7Hz,1H),7.90(d,J=7.8Hz,1H),7.72(t,J=7.9Hz,1H),7.33(s,1H),6.76(t,J=75.1Hz,1H),5.76(t,J=8.6Hz,1H),5.30(s,1H),3.83(d,J=33.6Hz,3H),2.70(m,3H),2.33–2.10(m,3H),1.99(m,4H),1.89(m,3H),1.13(s,3H).
实施例21
本发明合成的化合物:
Figure PCTCN2022097927-appb-000098
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000099
C21-2的合成:
于100mL单口瓶中加入C21-1(0.5g,2.17mmol),三乙胺(0.58mL,4.34mmol)和二氯甲烷(5mL),0℃下滴加4-Boc-氨基哌啶(0.43g,2.17mmol)的二氯甲烷(5mL)溶液,升至室温后反应2h。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,得860mg。MS[M+1]:395
C21-3的合成:
于100mL单口瓶中加入C21-2(860mg,2.18mmol)和二氯甲烷溶液(10mL),搅拌条件下室温滴加HCl/dioxane(4mL),滴毕反应2h。LC-MS监测反应完毕,浓缩反应液,加入少量乙酸乙酯和大量石油醚进行超声、洗涤,后过滤干燥得粗产品692mg。MS[M+1]:331 C21的合成:
于50mL单口瓶中加入C21-3(139mg,0.42mmol),int(80mg,0.21mmol),二异丙基乙胺(0.15mL,0.84mmol)和二甲基亚砜(2mL)。升温60℃反应16h。反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC(展开剂为二氯甲烷:甲醇=60:1)纯化得46mg。MS[M+1]:604 1H NMR(400MHz,CDCl 3)δ8.42(s,1H),7.73(q,J=8.3Hz,1H),7.34(s,1H),7.12(m,J=9.3,6.0Hz,1H),6.76(s,J=75.1Hz,1H),5.79(t,J=8.4Hz,1H),5.50(s,1H),3.97(m,1H),3.86(d,J=14.0Hz,2H),2.90(m,2H),2.71(m,1H),2.38–2.11(m,4H),2.10–1.97(m,3H),1.80(m,3H),1.15(s,3H).
实施例22
本发明合成的化合物:
Figure PCTCN2022097927-appb-000100
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000101
C22-2的合成:
于100mL单口瓶中加入C22-1(2g,9mmol),三乙胺(2.4mL,18mmol)和二氯甲烷(10mL),0℃下滴加4-Boc-氨基哌啶(1.81g,9mmol)的二氯甲烷(10mL)溶液,升至室温后反应2h。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,得2.2g。MS[M+1]:386
C22-3的合成:
于100mL单口瓶中加入C22-2(1g,2.6mmol),甲醇(10mL)和Pd/C(0.14g 10%),在H2条件下室温搅拌12h。LC-MS监测反应完毕,滤出Pd/C,浓缩反应液,得产品857mg。MS[M+1]:356
C22-4的合成:
于100mL单口瓶中加入C22-3(750mg,2.11mmol)和N,N-二甲基甲酰胺(20mL),碘甲烷(2ml),0℃下滴加氢氧化钾溶液(0.3g,10mL),升至室温后加热至50℃,回流搅拌过夜。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,柱层析石油醚:乙酸乙酯=3:1,得80mg。MS[M+1]:384
C22-5的合成:
于100mL单口瓶中加入C22-4(634mg,1.65mmol)和二氯甲烷溶液(10mL),搅拌条件下室温滴加HCl/dioxane(3mL),滴毕反应2h。LC-MS监测反应完毕,浓缩反应液,加入少量乙酸乙酯和大量石油醚进行超声、洗涤,后过滤干燥得粗产品260mg。MS[M+1]:256C22的合成:
于50mL单口瓶中加入C22-5(166mg,0.42mmol),int3(100mg,0.26mmol),二异丙基乙胺(0.18mL,1.04mmol)和二甲基亚砜(3mL)。升温60℃反应16h。反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC(展开 剂为二氯甲烷:甲醇=60:1)纯化得15mg。MS[M+1]:593 1H NMR(400MHz,CDCl 3)δ8.39(s,1H),7.61(d,J=8.8Hz,2H),7.32(s,1H),7.03–6.88(m,1H),6.70(d,J=8.9Hz,2H),5.81–5.69(m,1H),5.15(m,1H),3.86(d,J=17.0Hz,3H),3.07(s,6H),2.72(s,3H),2.40–2.17(m,2H),2.04(m,1H),2.00(m,4H),1.89(m,3H),1.13(s,3H).
实施例23
本发明合成的化合物:
Figure PCTCN2022097927-appb-000102
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000103
C23-2的合成
于50mL单口瓶中加入化合物4-叔丁氧羰基氨基哌啶(500mg,1.00eq),C19-1(516mg,1.00eq)和二氯甲烷(5mL)。0℃下滴加三乙胺(504mg,1.00eq),升温至室温反应3h。TLC监测反应完毕,向反应液中加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,浓缩后产物加入石油醚/乙酸乙酯(10/1)打浆,过滤后得950mg。C23-3的合成
于100mL单口瓶中加入C23-2(900mg,1eq)和二氯甲烷(5mL),随后缓慢加入HCl/dioxane(5ml,3eq)。室温反应2h,TLC监测反应完毕,浓缩,浓缩后产物加入石油醚打浆,过滤后得700mg。
C23的合成
于50mL单口瓶中加入int3(100mg,1eq)、C23-3(139mg,2eq)、二异丙基乙胺(166mg,2eq)和二甲基亚砜(5mL)。60℃反应16h。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC纯化得21mg。 1H NMR(400MHz,CDCl 3)δ8.40(s,1H),7.70(d,J=8.5Hz,2H),7.63–7.51(m,2H),7.33(s,1H),6.75(t,J=75.1Hz,1H),5.76(t,J=8.6Hz,1H),5.43(s,1H),3.80(d,J=38.4Hz,3H),2.71(s,2H),2.52(s,1H),2.31–2.10(m,4H),2.00(q,J=10.1,9.4Hz,2H),1.94–1.77(m,2H),1.73–1.66(m,2H),1.37(s,9H),1.13(s,3H).MS[M+1]:606
实施例24
本发明合成的化合物:
Figure PCTCN2022097927-appb-000104
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000105
C24-2的合成
于50mL单口瓶中加入化合物4-叔丁氧羰基氨基哌啶(500mg,1.00eq),C20-1(516mg,1.00eq)和二氯甲烷(5mL)。0℃下滴加三乙胺(504mg,1.00eq),升温至室温反应3h。TLC监测反应完毕,向反应液中加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,浓缩后产物加入石油醚/乙酸乙酯(10/1)打浆,过滤后得900mg。C24-3的合成
于100mL单口瓶中加入C24-2(900mg,1eq)和二氯甲烷(5mL),随后缓慢加入HCl/dioxane(5ml,3eq)。室温反应2h,TLC监测反应完毕,浓缩,浓缩后产物加入石油醚打浆,过滤后得650mg。
C24的合成
于50mL单口瓶中加入int3(100mg,1eq)、C24-3(139mg,2eq)、二异丙基乙胺(166mg,2eq)和二甲基亚砜(5mL)。60℃反应16h。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC纯化得11mg。 1H NMR(400MHz,CDCl 3)δ8.40(s,1H),7.72(d,J=8.6Hz,2H),7.33(s,1H),7.02(d,J=8.6Hz,2H),6.75(t,J=75.1Hz,1H),5.76(t,J=8.5Hz,1H),5.42(s,1H),3.90(s,3H),3.87-3.62(m,3H),2.62(d,J=67.4Hz,3H),2.31-2.20(m,1H),2.13(d,J=12.8Hz,2H),2.01(dd,J=16.1,6.9Hz,3H),1.90(t,J=9.2Hz,1H),1.79(s,1H),1.26(t,J=7.2Hz,1H),1.14(s,3H),0.92-0.82(m,2H).
MS[M+1]:580
实施例25
本发明合成的化合物:
Figure PCTCN2022097927-appb-000106
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000107
C25-2的合成
于50mL单口瓶中加入化合物4-叔丁氧羰基氨基哌啶(1g,1.00eq),C21-1(1.02g,1.00eq)和二氯甲烷(20mL)。0℃下滴加三乙胺(1.04g,1.00eq),升温至室温反应3h。TLC监测反应完毕,向反应液中加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,浓缩后产物加入石油醚/乙酸乙酯(10/1)打浆,过滤后得2.03g。
C25-3的合成
于100mL单口瓶中加入C25-2(2.03g,1eq)和二氯甲烷(20mL),随后缓慢加入HCl/dioxane(15ml,3eq)。室温反应2h,TLC监测反应完毕,浓缩,浓缩后产物加入石油醚打浆,过滤后得1.64g。
C25的合成
于50mL单口瓶中加入int3(100mg,1eq)、C25-3(80mg,2eq)、二异丙基乙胺(66mg,2eq)和二甲基亚砜(5mL)。60℃反应16h。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC纯化得30mg。 1H NMR(400MHz,CDCl 3)δ8.42(s,1H),7.87(td,J=7.4,1.8Hz,1H),7.66–7.56(m,1H),7.39-7.29(m,2H),7.26-7.21(m,1H),6.76(t,J=75.1Hz,1H),5.78(t,J=8.5Hz,1H),5.43(s,1H),3.88(d,J=11.7Hz,3H),2.79(d,J=57.9Hz,3H),2.26(td,J=12.0,11.5,7.0Hz,1H),2.15(d,J=12.9Hz,2H),2.01(q,J=10.1,8.9Hz,3H),1.89(q,J=8.3,7.6Hz,1H),1.80(d,J=11.1Hz,1H),1.26(s,1H),1.14(s,3H),0.93-0.81(m,1H).MS[M+1]:568
实施例26
本发明合成的化合物:
Figure PCTCN2022097927-appb-000108
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000109
C26-2的合成
于50mL单口瓶中加入化合物4-叔丁氧羰基氨基哌啶(0.94g,1.00eq),C22-1(1g,1.00eq)和二氯甲烷(20mL)。0℃下滴加三乙胺(0.95g,1.00eq),升温至室温反应3h。TLC监测反应完毕,向反应液中加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,浓缩后产物加入石油醚/乙酸乙酯(10/1)打浆,过滤后得1.64g。
C26-3的合成
于100mL单口瓶中加入C26-2(1.64g,1eq)和二氯甲烷(20mL),随后缓慢加入HCl/dioxane(15ml,3eq)。室温反应2h,TLC监测反应完毕,浓缩,浓缩后产物加入石油醚打浆,过滤后得1.4g。
C26的合成
于50mL单口瓶中加入int3(100mg,1eq)、C26-3(85mg,2eq)、二异丙基乙胺(66mg,2eq)和二甲基亚砜(5mL)。60℃反应16h。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC纯化得35mg。 1H NMR(400MHz,CDCl 3)δ8.42(s,1H),7.65–7.53(m,1H),7.35(s,1H),7.32–7.28(m,1H),7.22(td,J=9.0,4.0Hz,1H),6.76(t,J=75.1Hz,1H),5.79(t,J=8.5Hz,1H),5.46(s,1H),4.06–3.78(m,2H),2.90(s,2H),2.72(s,1H),2.27(td,J=11.5,7.0Hz,1H),2.17(d,J=12.8Hz,2H),2.02(td,J=10.7,10.1,5.8Hz,3H),1.97–1.75(m,3H),1.27(q,J=8.0,7.5Hz,1H),1.14(s,3H),0.86(q,J=7.4,5.9Hz,1H).MS[M+1]:586
实施例27
本发明合成的化合物:
Figure PCTCN2022097927-appb-000110
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000111
C27-2的合成
于50mL单口瓶中加入化合物4-叔丁氧羰基氨基哌啶(0.56g,1.00eq),C23-1(0.52g,1.00eq)和二氯甲烷(10mL)。0℃下滴加三乙胺(0.57g,1.00eq),升温至室温反应3h。TLC监测反应完毕,向反应液中加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,浓缩后产物加入石油醚/乙酸乙酯(10/1)打浆,过滤后得0.86g。C27-3的合成
于100mL单口瓶中加入C27-2(0.86g,1eq)和二氯甲烷(10mL),随后缓慢加入HCl/dioxane(15ml,3eq)。室温反应2h,TLC监测反应完毕,浓缩,浓缩后产物加入石油醚打浆,过滤后得0.42g。
C27的合成
于50mL单口瓶中加入int3(50mg,1eq)、C27-3(62mg,2eq)、二异丙基乙胺(33mg,2eq)和二甲基亚砜(5mL)。60℃反应16h。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC纯化得18mg。 1H NMR(400MHz,CDCl 3)δ8.74(d,J=4.8Hz,1H),8.42(s,1H),8.02–7.89(m,2H),7.57–7.49(m,1H),7.35(d,J=10.2Hz,1H),6.76(t,J=75.1Hz,2H),5.78(t,J=8.6Hz,1H),5.44(s,1H),3.96(d,J=12.7Hz,3H),3.14–2.98(m,2H),2.73(s,1H),2.29–2.21(m,1H),2.14(d,J=11.6Hz,2H),2.01(p,J=8.8Hz,3H),1.95–1.87(m,1H),1.82(s,1H),1.17(d,J=2.9Hz,1H),1.14(s,3H).MS[M+1]:551
实施例28
本发明合成的化合物:
Figure PCTCN2022097927-appb-000112
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000113
C28-2的合成
于50mL单口瓶中加入化合物4-叔丁氧羰基氨基哌啶(1g,1.00eq),C24-1(1.15g,1.05eq)和二氯甲烷(10mL)。0℃下滴加三乙胺(1.01g,2.00eq),升温至室温反应3h。TLC监测反应完毕,向反应液中加入水,二氯甲烷萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,浓缩后产物加入石油醚/乙酸乙酯(10/1)打浆,过滤后得1.8g。
C28-3的合成
于100mL单口瓶中加入C28-2(1.8g,1eq)和二氯甲烷(15mL),随后缓慢加入HCl/dioxane(15ml,3eq)。室温反应2h,TLC监测反应完毕,浓缩,浓缩后产物加入石油醚打浆,过滤后得1.2g。
C28的合成
于50mL单口瓶中加入int3(100mg,1eq)、C28-3(164mg,2eq)、二异丙基乙胺(132mg,4eq)和二甲基亚砜(3mL)。60℃反应16h。LC-MS监测反应完毕,向反应液加入水,乙酸乙酯萃取三次,无水硫酸钠干燥有机相,过滤,浓缩,Prep-TLC纯化得60mg。 1H NMR(400MHz,CDCl 3)δ8.40(s,1H),7.68(d,J=8.1Hz,2H),7.37-7.33(m,2H),6.75(s,1H),5.76(t,J=8.6Hz,1H),5.46(s,1H),3.80(d,J=38.1Hz,3H),2.68(t,J=7.7Hz,3H),2.52(s,1H),2.29-2.19(m,1H),2.13(d,J=12.6Hz,2H),2.08-1.94(m,3H),1.92-1.76(m,2H),1.70(dt,J=15.0,7.4Hz,4H),1.26(q,J=5.1Hz,2H),1.13(s,3H),0.97(t,J=7.3Hz,3H),0.92-0.82(m,2H).MS[M+1]:592
实施例29
本发明合成的化合物:
Figure PCTCN2022097927-appb-000114
合成路线及实验过程如下:
Figure PCTCN2022097927-appb-000115
将化合物int3(80mg)溶于DMSO,加入int7(89mg,2.0eq),三乙胺(0.11ml,3.0eq),60℃下反应过夜。监控反应完成,向体系中加水,然后加乙酸乙酯萃取,合并有机相,无水硫酸钠干燥,减压浓缩,制备板分离得到产物(50mg)。 1H NMR(400MHz,CDCl 3)δ8.45(s,1H),7.36(s,1H),6.77(t,J=75.1Hz,1H),5.82(s,1H),5.57(d,J=55.6Hz,1H),3.89–3.73(m,2H),2.91(m,J=11.8,2.8Hz,2H),2.83(s,3H),2.74(s,1H),2.34–2.11(m,4H),2.10–1.98(m,2H),1.99–1.78(m,2H),1.78–1.58(m,2H),1.16(s,3H).MS[M+1]:488.5。
采用类似上述的合成方法合成下列表中化合物:
Figure PCTCN2022097927-appb-000116
Figure PCTCN2022097927-appb-000117
Figure PCTCN2022097927-appb-000118
Figure PCTCN2022097927-appb-000119
Figure PCTCN2022097927-appb-000120
Figure PCTCN2022097927-appb-000121
Figure PCTCN2022097927-appb-000122
Figure PCTCN2022097927-appb-000123
Figure PCTCN2022097927-appb-000124
细胞增殖抑制试验
一、实验材料
细胞系 供应商
MCF7 中国科学院上海细胞库
T47D 中国科学院上海细胞库
OVCAR3 北纳生物
HCC1806 北纳生物
对照化合物WO2018033815A1 PF-06873600、帕博西尼(Palbociclib)
Figure PCTCN2022097927-appb-000125
二、实验方案
将MCF7、T47D、OVCAR3和HCC1806细胞分别以3000、3000、2500、2000个细胞/孔接种在96孔板中,并在37℃,5%CO 2培养箱中培养过夜。
1、化合物稀释
a)配制受测化合物梯度稀释溶液:帕博西尼(Palbociclib)、实施例化合物以10mM为储液。然后取2.5μl储液溶解于497.5μl无DMSO培养液中,再以0.1%DMSO培养液进行3倍连续梯度稀释,共9个浓度,稀释后化合物浓度如下:
10000nM,3333.33nM,1111.11nM,370.34nM,123.45nM,41.15nM,13.72nM,4.57nM,1.52nM
b)充分混匀后分别取20μL培化合物溶液加入含有80μL细胞的细胞培养板中,每个浓度4个复孔。
c)将细胞转移至培养箱孵育7天,培养至第4天时用含相同浓度化合物的新鲜培养液进行换液。
2、MTT检测
a)取出细胞培养板在hood中加入5mg/ml MTT 10μL.
b)把细胞培养板放回培养箱继续孵育3小时
c)取出细胞培养板去除培养液,加入异丙醇(0.4mMHCl,0.4%NP40)100μL,室温摇床30分钟,570nm读板。
3、数据分析
使用如下公式计算存率(%Cell Survival):
%Cell Survival=100%×(OD_Sample-OD_LCave)/(OD_HC-OD_LCave)
OD_HC:0.1‰DMSO对照组细胞读数
OD_Sample:加入化合物的细胞读数
OD_LC:空白培养基读数
Analyed by Prizm:Dose-response-Inhibition-Log(inhibitor)vs response(three parameters for the best fit)
通过浓度-响应曲线拟合计算IC50(nM)值。
细胞增殖抑制试验结果见下表1。
表1
Figure PCTCN2022097927-appb-000126
Figure PCTCN2022097927-appb-000127
Figure PCTCN2022097927-appb-000128
Figure PCTCN2022097927-appb-000129
A代表IC50值≤50nM,B代表IC50值>50nM且≤200nM,C代表IC50值>200nM且≤1000nM,D代表IC50值>1000nM。
从上表可知本专利化合物对帕博西尼敏感型和耐药型细胞均有很好的抑制作用,与PF-06873600相比本专利化合物与之活性相当,乃至更优,有望进一步开发成为用于调节CDK激酶活性或治疗CDK相关疾病方面的药物。
MCF-7-Palbo-R细胞抗增殖实验
一、实验材料
MCF-7/palbo-R
EMEM培养基(ATCC,30-2003);DMEM/F12培养基(Gibco-11330-032)
胎牛血清(cellmax-SA211.02)
DMSO(SIGMA,D2650)
BrdU ELISA细胞增殖实验试剂盒(Roche,11647229001)
酶标仪(e.g.MD-SpectraMax ID5 or EnVision)
二、实验方案
1、细胞铺板
1)细胞计数,调整细胞悬浮液浓度
2)将MCF7/Palbo-R细胞悬浮液以90μl体积接种于96孔板中,使每孔细胞数为4000-5000个;
3)空白对照组加入90μl培养基;
2、化合物稀释
a)配制受测化合物梯度稀释溶液:帕博西尼(Palbociclib)、实施例化合物以10μM为储液。以0.1%DMSO培养液进行3倍连续梯度稀释,共9个浓度,稀释后化合物浓度如下:
10000nM,3333.33nM,1111.11nM,370.34nM,123.45nM,41.15nM,13.72nM,4.57nM,1.52nM
b)化合物处理72小时后:
1)用RPMI培养基稀释10mM的Brdu储备液,以20uL/孔加入96孔板,350转/分钟振摇10分钟。
2)将96孔板放回培养箱1或2小时。
3)去除96孔板中培养基,加入200ul/孔Fixation/Denaturation溶液,室温孵育30分钟
4)去除固定液,加入100ul过氧化物酶偶联抗Brdu抗体溶液(抗体稀释液1:100稀释制备),
在350转/分钟的转速下室温孵化1小时。
6)96孔板以300ul/孔PBS洗三次。
8)加入100ul过氧化物酶底物溶液,在室温下以350转/分振摇约10-20分钟,直到DMSO处理的细胞对照孔变为中蓝色。颜色太深会导致吸光度值超出读取范围。
9)加入25ul的1M硫酸终止反应。振摇以确保颜色均匀。
10)记录在450nm下的吸光度。
3、数据处理
使用如下公式计算抑制率:
IR(%)=(1–(RLU compound–RLU blank)/(RLU control–RLU blank))*100%.MCF-7-Palbo-R细胞抗增殖实验结果见表2
表2
Figure PCTCN2022097927-appb-000130
本专利化合物对帕博西尼耐药的细胞株具有很好的抑制率,且优于PF-06873600。
酶活性测试
准备化合物:
1、将待测化合物及帕博西尼配置成0.5nM的DMSO溶液;
2、用Echo550转移20nL储液至384孔板。使用DMSO做空白对照。
实验步骤:
1、按下表准备含有酶、底物、辅酶因子的1.3X酶溶液;
2、每孔中加入15uL的1.3X酶溶液,室温下孵育30min;
3、加入5uL的ATP溶液启动反应。最终每孔的体积为20uL;
4、室温下孵育150min后加入75uL的终止缓冲溶液终止实验;
5、使用EZ reader分析样品。
Figure PCTCN2022097927-appb-000131
数据分析:
使用下诉方程计算%抑制率
使用DMSO处理的为阳性对照(PC)
不加酶的为阴性对照(NC)
%抑制率=100-100*((CR PC-CR sample)/(CR PC-CR NC))
酶抑制实验结果见表3
表3
Figure PCTCN2022097927-appb-000132
Figure PCTCN2022097927-appb-000133
Figure PCTCN2022097927-appb-000134
A代表IC50值≤10nM,B代表IC50值>10nM且≤100nM,C代表IC50值>100nM且≤500nM,D代表IC50值>500nM,数据表明本专利化合物对CDK2、CDK4、CDK6均有较优的激酶抑制活性。
异种移植瘤实验:
xMCF-7/Palbo-R模型:
为了建立异种移植肿瘤模型,将1×107xMCF-7/Palbo-R细胞悬浮在0.2mL Matrigel和mouse(1:1)的混合液中,皮下注射到6-8周的Balb/c nube雌性小鼠。定期观察肿瘤生长情况,用游标卡尺测量(体积=1/2×(长×宽2))体积,待肿瘤生长至平均体积100~150mm3时,根据肿瘤大小和小鼠体重随机分组给药(接种后12天)。将小鼠随机分配到5组,每组包含3只动物,每组所用剂量分别为:(a)Vehicle;(b)10mg/kg PF-06873600;(c)20mg/kg C27;(d)10mg/kg C3;(h)20mg/kg C3。每天通过口服管饲法给药两次,持续20天。每周测量肿瘤体积和动物体重两次,并持续到试验结束。给药日定义为第0天。测量时间点为第0天、第3天、第7天、第10天、第14天、第17天和第20天。所有小鼠均采用颈椎脱臼法安乐死。实验结束时,收集肿瘤并称重计算TGI值(见表4)。实验结果如图1所示。
表4
化合物 剂量(mg/Kg) TGI
PF-06873600 10 36.2
C27 20 61.5
C3 10 64.4
C3 20 64.6
OVCAR-3模型:
1、细胞培养
OVCAR-3细胞培养在含20%胎牛血清和10μg/mL Insalin的RPMI1640培养液中。收集指数生长期的细胞,PBS与Matrigel 1:1混合后重悬细胞至适合浓度用于裸鼠皮下肿瘤接种。
2、动物造模
为了建立异种移植肿瘤模型,将1×10 7OVCAR-3细胞悬浮在0.2mL PBS和Matrigel(1:1)的混合液中,皮下注射到6-8周的BALB/c nube雌性小鼠于右侧背部。定期观察肿瘤生长情况,用游标卡尺测量(体积=1/2×(长×宽 2))体积,待肿瘤生长至平均体积100~150mm 3时,根据肿瘤大小和小鼠体重随机分组给药(接种后27天)。将小鼠随机分配到6组,每组包含5只动物,每组所用剂量分别为:(a)Vehicle;(b)60mg/kg Palbociclib;(c)30mg/kg PF-06873600;(d)30mg/kg C3;(e)30mg/kg C29;(f)50mg/kg C29。Vehicle是DMSO/Solutol/Saline(5%/10%/85%),Palbociclib,PF-06873600,C3,C29分别6mg/mL,3mg/mL,3mg/mL,3mg/mL和5mg/mL浓度溶解在Vehicle中。其中Vehicle,30mg/kg PF-06873600,30mg/kg C3,30mg/kg C29和50mg/kg C29组小鼠每天通过口服管饲法给药两次,持续21天;60mg/kg Palbociclib组小鼠每天通过口服管饲法给药一次,持续21天。每周测量肿瘤体积和动物体重两次,并持续到试验结束。给药日定义为第0天。测量时间点为第0天、第3天、第7天、第10天、第14天、第17天和第21天。所有小鼠均采用颈椎脱臼法安乐死。实验结束时,收集肿瘤并称重计算TGI值(见表5)。实验结果如图2所示。
表5
化合物 剂量(mg/Kg) TGI
Palbociclib 60 27.7
PF-06873600 30 52.6
C3 30 71.3
C29 30 69.0
C29 50 74.8
MV4-11模型:
1、细胞培养
MV-4-11细胞培养在含10%胎牛血清的IMEM培养液中。收集指数生长期的细胞,PBS重悬细胞至适合浓度用于裸鼠皮下肿瘤接种。
2、动物造模
为了建立异种移植肿瘤模型,将5×10 6MV4-11细胞悬浮在0.1mL PBS和Matrigel1:1的混合液中,皮下注射到6-8周的NOD/SCID小鼠于右侧背部。定期观察肿瘤生长情况,用游标卡尺测量(体积=1/2×(长×宽 2))体积,待肿瘤生长至平均体积100~150mm 3时,根据肿瘤大小和小鼠体重随机分组给药(接种后12天)。将小鼠随机分配到5组,每组包含5只动物:(a)Vehicle;(b)20mg/kg Palbociclib;(c)10mg/kg PF-06873600;(d)10mg/kg C3;(e)20mg/kg C3。Vehicle是DMSO/Solutol/Saline(5%/10%/85%),Palbociclib,PF-06873600,TYK-00127两组分别2mg/mL,1mg/mL,1mg/mL和2mg/mL浓度溶解在Vehicle中。其中Vehicle,10mg/kg TY-3301,10mg/kg C3,20mg/kg C3组小鼠每天通过口服管饲法给药两次,持续15天;20mg/kg TY-3300组小鼠每天通过口服管饲法给药一次,持续15天。每周测量肿瘤体积和动物体重两次,并持续到试验结束。给药日定义为第0天。测量时间点为第0天、第3天、第7天、第10天、第14天和第15天。所有小鼠均采用颈椎脱臼法安乐死。实验结束时,收集肿瘤并称重计算TGI值(见表6)。实验结果如图3所示。
表6
化合物 剂量(mg/Kg) TGI
Palbociclib 20 39.0
PF-06873600 10 60.9
C3 10 69.5
C3 20 95.3
动物模型的体内药效实验表明本专利中的化合物表现出了较好的肿瘤抑制效果,且实验效果远优于帕博西尼对照组,与PF-06873600相比同样表现出了较大的优势。
本发明提供了一类具有优异激酶、细胞、体内药效的化合物及其合成方法,该类化合物有望开发成为解决现有临床中帕博西尼等耐药的问题,为这类病人带来新的治疗方案。
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。

Claims (10)

  1. 式(Ⅰ)化合物、或其药学上可接受的盐、立体异构体、溶剂化物或前药,
    Figure PCTCN2022097927-appb-100001
    其中:
    R 1选自下组:未取代或1~4个R 6取代的C3-C10环烷基、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元并环、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元螺环、未取代或1~4个R 6取代的不含或含1-3个选自N、O、S的杂原子的5-15元桥环、未取代或1~4个R 6取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、未取代或1~4个R 6取代的C6-C10芳基、未取代或1~4个R 6取代的含1-5个选自N、O、S的杂原子的3-10元杂芳基、未取代或1~4个R 6取代的C1-C6烷基;
    R 2选自下组:H、F、OH、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基,且R 2可与环上的原子相连形成螺环、桥环、或并环结构;
    R 3a和R 3b独立地选自下组:H、F、OH、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
    其中,R 2、R 3a和R 3b中每个所述C1-C4烷基和卤代C1-C4烷基任选被卤素、OH、C1-C4烷氧基或卤代C1-C4烷氧基取代;
    R 4选自下组:Br、取代或未取代的含1-4个选自N、O、S的杂原子的3-10元杂环烷基、-OR 4'、-SR 4'、-NR 4'R 4",其中,R 4'和R 4"分别独立地选自下组:H、COR 7、取代或未取代的C1-C6烷基、取代或未取代的C3-C6环烷基、取代或未取代的含1-4个选自N、O、S的杂原子的3-6元杂环烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、OH、CN、=O、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
    R 5选自下组:H、卤素、C1-C2烷基、卤代C1-C2烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
    各R 6独立地选自下组:H、氘、羟基、卤素、氰基、=O、COR 7、CO 2R 7、CONR 8R 9、CO 2NR 8R 9、SO 2R 7、SO 2NR 8R 9、NR 8SO 2R 7、NHSO 2NR 8R 9
    Figure PCTCN2022097927-appb-100002
    NR 8R 9、R”’取代或未取代的含1-3个选自N、O、S的杂原子的3-10元杂环烷基、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、C3-C8环烷基、卤代C3-C8环烷基、C4-C10螺环、C3-C10并环、C4-C10桥环、硫代C1~C6烷基、C6-C10芳基、含1-5个选自N、O、S的杂原子的3-10元杂芳基;
    R”’选自下组:=O、NR’R”、C1-C4烷基、卤代C1-C4烷基;
    各R 7独立地选自下组:C1-C4烷基、-L-(C3-C8环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基)、-L-(3-10元芳基),其中所述的C1-C4烷基、C3-C8环烷基、3-10元杂环烷基、3-10元杂芳基、3-10元芳基任选的被0~4个D、OH、卤素、CN、C1-C4烷基、卤代C1-C4烷基、N(C1-C4烷基) 2、NHCO(C1-C4烷基)、SO 2(C1-C4烷基)、CO 2(C1-C4烷基)、含1-4个选自N、O、S的杂原子的3-6元杂环烷基、CO(C1-C4烷基)、C1-C4烷氧基或卤代C1-C4烷氧基取代;
    R 8和R 9独立地选自下组:H、C1-C4烷基、卤代C1-C4烷基、C3-C8环烷基、-L-(C3-C8 环烷基)、-L-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L-(含1-5个选自N、O、S的杂原子的3-10元杂芳基);或者
    R 8和R 9可以与其所连接的N一起形成取代或未取代的4-6元杂环基,所述取代指被选自下组的1-3个取代基取代:=O、NR’R”、C1-C6烷基;
    各R’和R”独立地选自下组:H、C1-C4烷基;
    各L独立地为键或C1-C4亚烷基,所述C1-C4亚烷基任选被OH、C1-C4烷氧基或卤代C1-C4烷氧基取代;
    p选自下组:0、1、2、3、4;
    r选自下组:0、1、2、3、4。
  2. 如权利要求1所述的化合物,其特征在于,所述化合物具有式(II)或式(Ⅲ)所示结构:
    Figure PCTCN2022097927-appb-100003
    其中:
    A环选自下组:含0-4个R 1’取代的6-10元芳基、含0-4个R 1’取代的C3-C8环烷基、含0-4个R 1’取代的含1-5个选自N、O、S的杂原子的5-10元杂芳基;
    各R 1’独立地选自下组:氘、卤素、OH、CN、SO 2R 31、COR 31、CO 2R 31、NR 41R 51、NHCOR 41、CONR 41R 51、OCONR 41R 51、NHCON R 41R 51、NHCOOR 41、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、取代或未取代的C3-C6环烷基、取代或未取代的含1-4个选自N、O、S的杂原子的3-6元杂环烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、OH、CN、=O、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
    R 2’选自下组:H、氘;
    R 3’选自下组:取代或未取代的C1-C4烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、OH、CN、=O、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
    R 4’选自下组:H、氘、OH;R 31选自下组:H、C1-C4烷基、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、取代或未取代的C3-C6环烷基、取代或未取代的含1-4个选自N、O、S的杂原子的3-6元杂环烷基,所述取代指被选自下组的1-3个取代基取代:氘、卤素、OH、CN、=O、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基;
    R 41和R 51独立地选自下组:H、C1-C4烷基、卤代C1-C4烷基、C3-C8环烷基、-L’-(C3-C8环烷基)、-L’-(含1-4个选自N、O、S的杂原子的3-10元杂环烷基)、-L’-(含1-5个选自N、O、S的杂原子的3-10元杂芳基),L’为键或C1-C6亚烷基;或者
    R 41和R 51可以与其所连接的N一起形成取代或未取代的4-6元杂环基,所述取代指被选自下组的1-3个取代基取代:卤素、OH、=O、C1-C6烷基;
    n选自下组:0、1、2、3、4。
  3. 如权利要求2所述化合物,其特征在于,A环为含0-4个R 1’取代的6-10元芳基。
  4. 如权利要求2所述化合物,其特征在于,A环为含0-4个R 1’取代的C3-C8环烷基。
  5. 如权利要求2所述的化合物,其特征在于,
    A环选自下组:含0-4个R 1’取代的6-10元芳基、含0-4个R 1’取代的C3-C8环烷基、含0-4个R 1’取代的含1-5个选自N、O、S的杂原子的5-10元杂芳基;
    各R 1’独立地选自下组:氘、卤素、OH、CN、SO2R 31、COR 31、CO2R 31、NR 41R 51、NHCOR 41、CONR 41R 51、卤代C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷氧基、取代或未取代的C3-C6环烷基、取代或未取代的含1-4个选自N、O、S的杂原子的3-6元杂环烷基;
    R 2’选自下组:H、氘;
    R 4’选自下组:H、氘;
    R 31为C1-C4烷基;
    R 41和R 51独立地选自下组:H、C1-C4烷基;
    n选自下组:0、1、2、3、4。
  6. 如权利要求2所述的化合物,其特征在于,
    A环选自下组:含0-4个R 1’取代的6-10元芳基、含0-4个R 1’取代的C3-C8环烷基;
    R 1’选自下组:卤素、OH、C1-C4烷氧基、卤代C1-C4烷氧基;
    R 2’选自下组:H、氘;
    n选自下组:0、1、2、3、4。
  7. 一种化合物,其特征在于,所述化合物选自下组:
    Figure PCTCN2022097927-appb-100004
    Figure PCTCN2022097927-appb-100005
    Figure PCTCN2022097927-appb-100006
    Figure PCTCN2022097927-appb-100007
    Figure PCTCN2022097927-appb-100008
    Figure PCTCN2022097927-appb-100009
  8. 一种药物组合物,其特征在于,包含治疗有效量的权利要求1所述的化合物、或其药学上可接受的盐、立体异构体、溶剂化物或前药,以及药学上可接受的载体。
  9. 一种权利要求1所述的化合物、或其药学上可接受的盐、立体异构体、溶剂化物或前药的用途,其特征在于,用于制备CDK2/4/6激酶抑制剂药物。
  10. 一种权利要求1所述的化合物、或其药学上可接受的盐、立体异构体、溶剂化物或前药的用途,其特征在于,用于制备用于调节CDK激酶活性或治疗CDK相关疾病的药物。
PCT/CN2022/097927 2021-06-09 2022-06-09 用作cdk激酶抑制剂的化合物及其应用 WO2022258023A1 (zh)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP22819620.0A EP4353724A1 (en) 2021-06-09 2022-06-09 Compound as cdk kinase inhibitor and use thereof
KR1020247000908A KR20240021239A (ko) 2021-06-09 2022-06-09 Cdk 키나아제 억제제로 사용되는 화합물 및 이의 용도
AU2022289202A AU2022289202A1 (en) 2021-06-09 2022-06-09 Compound as cdk kinase inhibitor and use thereof
CA3221997A CA3221997A1 (en) 2021-06-09 2022-06-09 Compound as cdk kinase inhibitor and use thereof
CN202280006672.6A CN116456987A (zh) 2021-06-09 2022-06-09 用作cdk激酶抑制剂的化合物及其应用

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202110644327 2021-06-09
CN202110644327.2 2021-06-09
CN202111164489.2 2021-09-30
CN202111164489 2021-09-30

Publications (1)

Publication Number Publication Date
WO2022258023A1 true WO2022258023A1 (zh) 2022-12-15

Family

ID=84425714

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/097927 WO2022258023A1 (zh) 2021-06-09 2022-06-09 用作cdk激酶抑制剂的化合物及其应用

Country Status (7)

Country Link
EP (1) EP4353724A1 (zh)
KR (1) KR20240021239A (zh)
CN (1) CN116456987A (zh)
AU (1) AU2022289202A1 (zh)
CA (1) CA3221997A1 (zh)
TW (1) TWI823420B (zh)
WO (1) WO2022258023A1 (zh)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180044344A1 (en) * 2016-08-15 2018-02-15 Pfizer Inc. CDK2/4/6 Inhibitors
WO2020065494A1 (en) * 2018-09-25 2020-04-02 Pfizer Inc. Synthesis of pyrido[2,3-d]pyrimidin-7(8h)-ones

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108191857B (zh) * 2017-01-24 2020-10-23 晟科药业(江苏)有限公司 6-取代的吡啶并[2,3-d]嘧啶类化合物作为蛋白激酶抑制剂

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180044344A1 (en) * 2016-08-15 2018-02-15 Pfizer Inc. CDK2/4/6 Inhibitors
WO2018033815A1 (en) 2016-08-15 2018-02-22 Pfizer Inc. Pyridopyrimdinone cdk2/4/6 inhibitors
WO2020065494A1 (en) * 2018-09-25 2020-04-02 Pfizer Inc. Synthesis of pyrido[2,3-d]pyrimidin-7(8h)-ones

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS

Also Published As

Publication number Publication date
CN116456987A (zh) 2023-07-18
EP4353724A1 (en) 2024-04-17
AU2022289202A1 (en) 2024-01-18
KR20240021239A (ko) 2024-02-16
TWI823420B (zh) 2023-11-21
TW202304912A (zh) 2023-02-01
CA3221997A1 (en) 2022-12-15

Similar Documents

Publication Publication Date Title
US9890168B2 (en) 2,4-disubstituted 7H-pyrrolo[2,3-d]pyrimidine derivative, preparation method and medicinal use thereof
TWI639602B (zh) 三環旋轉酶抑制劑
WO2015127872A1 (zh) 2,4-二取代苯-1,5-二胺衍生物及其应用以及由其制备的药物组合物和药用组合物
WO2015022926A1 (ja) 新規な縮合ピリミジン化合物又はその塩
US10085983B2 (en) Azabicyclo derivatives, process for preparation thereof and medical use thereof
EA023350B1 (ru) Противомикробные соединения, способы их получения и применение
WO2015144021A1 (zh) 取代氮杂环类衍生物、含其的药物组合物及其在抗肿瘤中的应用
WO2016192132A1 (zh) 作为alk抑制剂的嘧啶衍生物
WO2022135432A1 (zh) 作为egfr抑制剂的大环杂环类化合物及其应用
WO2022268230A1 (zh) 作为kif18a抑制剂的化合物
EP0776891B1 (en) Pyrrolylbenzimidazole derivatives
CN114685531A (zh) 四并环化合物及其药物组合物和应用
WO2023280283A1 (zh) 用作shp2抑制剂的化合物及其应用
TW201922709A (zh) 表皮生長因子受體抑制劑
WO2023078451A1 (zh) 用作cdk7激酶抑制剂的化合物及其应用
CN106279119B (zh) 一种新型激酶抑制剂的制备及其应用
WO2022258023A1 (zh) 用作cdk激酶抑制剂的化合物及其应用
US20150152133A1 (en) Acylation derivatives of paridis saponins i, preparation method therefor and application thereof
CN115322158A (zh) 作为krasg12c蛋白抑制剂的取代喹唑啉类化合物
CN112358435B (zh) 取代芳并杂环类化合物、制备方法和抑制ulk1及抗肿瘤用途
TWI794576B (zh) 一類含氟取代的苯并噻吩類化合物及其藥物組合物及應用
WO2022007841A1 (zh) 一种egfr抑制剂、其制备方法和在药学上的应用
CN111233774B (zh) 一种胺基嘧啶类化合物
CN113336774A (zh) 作为trk抑制剂的取代的手性二芳基大环化合物
CN116783183A (zh) 作为vhl抑制剂用于治疗贫血和癌症的1-(2-(4-环丙基-1h-1,2,3-***-1-基)乙酰基)-4-羟基-n-(苄基)吡咯烷-2-甲酰胺衍生物

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: 22819620

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202280006672.6

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 3221997

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2022289202

Country of ref document: AU

Ref document number: AU2022289202

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 20247000908

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2022819620

Country of ref document: EP

Ref document number: 1020247000908

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022289202

Country of ref document: AU

Date of ref document: 20220609

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2022819620

Country of ref document: EP

Effective date: 20240109