WO2024002318A1 - 哒嗪并吡啶酮衍生物及其用途 - Google Patents

哒嗪并吡啶酮衍生物及其用途 Download PDF

Info

Publication number
WO2024002318A1
WO2024002318A1 PCT/CN2023/104412 CN2023104412W WO2024002318A1 WO 2024002318 A1 WO2024002318 A1 WO 2024002318A1 CN 2023104412 W CN2023104412 W CN 2023104412W WO 2024002318 A1 WO2024002318 A1 WO 2024002318A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
halogen
methyl
substituted
group
Prior art date
Application number
PCT/CN2023/104412
Other languages
English (en)
French (fr)
Inventor
陈寿军
强晓明
宁德争
丁兆
Original Assignee
四川汇宇制药股份有限公司
四川汇宇海玥医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 四川汇宇制药股份有限公司, 四川汇宇海玥医药科技有限公司 filed Critical 四川汇宇制药股份有限公司
Publication of WO2024002318A1 publication Critical patent/WO2024002318A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/5025Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with heterocyclic ring systems
    • 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
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention relates to the field of medical technology, and in particular to a type of pyridazinopyridone derivatives used as SOS1 protein inhibitors and their uses.
  • SOS1 (son of sevenless homolog 1) protein is a regulatory protein widely expressed in cells. As a type of guanine nucleotide exchange factor for Ras or Rac1 protein, it plays a role in the intracellular Ras or Rac1 signal transduction pathway. Important regulatory role (Baltanás, FC; Zarich, N.; Rojas- JM; Santos, E. Biochim. Biophys. Acta. Rev. Cancer. 2020, 1874, 188445). The role of SOS1 protein in the Ras signal transduction pathway is to promote the release of GDP from Ras, bind to GTP, and convert the Ras protein from an inactive state to an active state.
  • RAS mutations KRAS, NRAS, HRAS are present in 90% of pancreatic cancers, 45% of colon cancers, and 35% of lung cancers.
  • NSCLC Non-small cell lung cancer
  • Ras proto-oncogene is the most common in NSCLC Mutated genes (Prior, L. et al, Cancer Res. 2012, 72 (10), 2457-2467; Li, L. et al, J Exp Clin Cancer Res. 2018, 37 (1), 178), among which, v-Ki-ras2Kirsten
  • the rat sarcoma viral oncogene (Kras) accounts for 90% of RAS mutations in lung adenocarcinoma (Hunter, JC; et al, Mol. Cancer Res. 2015, 13(9), 1325-35).
  • SOS1 plays an important regulatory role in many signal transduction pathways in cells. Studies have shown that inhibiting SOS1 activity has a potent inhibitory effect on cancer cell proliferation caused by all major Kras gene mutations (Kessler, D.; Gerlach, D.; Kraut, N., McConnell, D.B. Curr. Opin. Chem. Biol .2021,62,109-118). In addition, a clinical study in 62 patients with ovarian cancer showed that the expression of RAS mutations and SOS1 mutations in ovarian cancer tissues increased significantly, and the prognosis was related to shortened PFS of patients, suggesting that RAS and SOS1 targeted therapy are more effective in ovarian cancer.
  • SOS1 gene mutations have also been found in many other cancer cells, such as embryonal rhabdomyosarcoma, Sertoli cell testicular tumors, and cutaneous granulosa cell tumors (Denayer et al., Genes Chromosomes Cancer, 2010, 49(3):242-52) and lung adenocarcinoma (Cancer Genome AtIas Research Network., Nature 2014, 511(7511), 543-50).
  • SOS1 gene is involved in bladder cancer (Watanabe etal., IUBMB Life., 2000, 49(4), 317-20) and prostate cancer (Timofeeva et al., Int.J.Oncol., 2009, 35(4) ):751-60) are highly expressed.
  • BCR-ABL activates GRB2 through phosphorylation and recruits SOS1, thereby continuously activating the Ras/MAPK signaling pathway, leading to the malignant proliferation of hematopoietic stem cells. Therefore, SOS1 protein is also a potential new target for the treatment of chronic myelogenous leukemia.
  • SOS1 gene mutations are also closely related to some pathogenic rash diseases such as Noonan syndrome (NS), cardiofaciocutaneous syndrome (CFC) and type I hereditary gingival fibromatosis (Pierre et al. al., Biochem. Pharmacol., 2011, 82(9):1049-56).
  • NS Noonan syndrome
  • CFC cardiofaciocutaneous syndrome
  • type I hereditary gingival fibromatosis Pierre et al. al., Biochem. Pharmacol., 2011, 82(9):1049-56.
  • SOS1 inhibition is mechanistically similar to SHP2 inhibition (Nichols, RJ; et al. Nat Cell Biol. 2018, 20(9), 1064-1073), which indicates that SOS1 inhibition can also enhance the efficacy of KRAS G12C and MEK inhibitors .
  • Preliminary data show significant synergy between SOS1 and MEK inhibition in PDX models with multiple G12 and G13 KRAS mutations (Hofmann, MH; et al; Cancer Discov. 2020, 10.1158/2159-8290.CD-20- 0142.).
  • inhibiting SOS1 has the potential advantage of directly increasing the efficacy of KRAS G12C inhibitors by increasing the amount of mutant KRAS G12C (Hillig, R.C. ,et al,Proc Natl Acad Sci U S A.2019,116(7),2551-2560). Although further research is needed, inhibiting SOS1 has great potential for clinical application as a combination therapy strategy.
  • the main technical problem solved by the present invention is to provide a class of pyridazinopyridone derivatives that have a strong selective inhibitory effect on SOS1.
  • the present invention provides the compound represented by formula I, or its tautomer, stereoisomer, solvate, metabolite, isotope label, pharmaceutically acceptable salt, and co-crystal:
  • Ring A is selected from aryl and heteroaryl groups, which are optionally selected from halogen, alkyl, alkoxy, -OH, -CN, -NR 5 R 6 , -SF 5 , -SO 2 R 12 is substituted by a substituent, and the alkyl and alkoxy groups are optionally further substituted by a substituent selected from halogen and -NR 5 R 6 , where R 5 and R 6 are independently selected from H , alkyl, R 12 is alkyl and optionally substituted by halogen;
  • R a , R b , R c are independently selected from H and alkyl
  • R 2 is -XR 13 , X is selected from alkylene, a single bond, R 13 is selected from alkyl, cycloalkyl, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl, aryl, heteroaryl; in:
  • R 13 is an alkyl group
  • X is a single bond
  • R 13 is selected from aryl or heteroaryl, it is optionally substituted by a substituent selected from halogen, alkyl, alkoxy, -OH, -CN, -NR 14 R 15 , R 14 , R 15 Independently selected from H, alkyl;
  • R 13 is selected from cycloalkyl, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl, optionally selected from halogen, alkyl, -YR 16 is substituted with a substituent, and the alkyl group is optionally further substituted with a substituent selected from halogen, -OH, alkoxy, -NR 17 R 18 ; wherein:
  • Y is an alkylene group, and R 16 is selected from cycloalkyl, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl, aryl, and heteroaryl;
  • Y is selected from R 16 is selected from alkyl, alkoxy, -NR 17 R 18 , cycloalkyl, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl, aryl, heteroaryl;
  • R 17 and R 18 are independently selected from H and alkyl.
  • Ring A is selected from aryl and heteroaryl, and the aryl and heteroaryl are optionally substituted by halogen, alkyl, alkoxy, -OH, -CN, -NR 5 R 6
  • the alkyl and alkoxy groups are optionally further substituted by substituents selected from halogen and -NR 5 R 6 , where R 5 and R 6 are independently selected from H and alkyl.
  • Ring A is selected from a 6- to 12-membered aryl group and a 5- to 12-membered heteroaryl group.
  • the heteroaryl group contains heteroatoms selected from N, O, and S.
  • the substituents of the above groups are as defined before.
  • ring A is selected from a 6-membered aryl group and a 5- to 11-membered heteroaryl group.
  • the heteroaryl group contains 1 to 3 heteroatoms selected from N, O, and S.
  • the substituents of the above groups are as described above. definition.
  • ring A is selected from a 6-membered aryl group, a 6-membered aryl group and a 5-membered heteroaryl group.
  • the heteroaryl group contains 1 heteroatom selected from N, O, and S.
  • the substituents of the above groups are as above. defined.
  • Ring A is selected from phenyl, phenyl-thienyl, and the substituents of the above groups are as defined before.
  • Ring A is phenyl and its substituents are as defined before.
  • Ring A the substituents contained in Ring A are selected from halogen, C1-C6 alkyl, C1-C6 alkoxy, -OH, -CN, -NR 5 R 6 , -SF 5 , -SO 2 R 12 ,
  • the alkyl and alkoxy groups are optionally further substituted with substituents selected from halogen and -NR 5 R 6 , wherein R 5 and R 6 are independently selected from H, C1 to C6 alkyl, and R 12 is C1 ⁇ C6 alkyl and optionally substituted by halogen.
  • the substituent contained in ring A is selected from halogen, C1-C6 alkyl, -CN, -NR 5 R 6 , -SF 5 , -SO 2 R 12 , and the alkyl group is optionally further selected from Substituted from halogen, -NR 5 R 6 substituents, wherein R 5 and R 6 are independently selected from H, C1-C6 alkyl, R 12 is C1-C6 alkyl and optionally substituted by halogen.
  • the substituent contained in ring A is selected from -F, methyl, -F substituted methyl, -NR 5 R 6 substituted methyl, -CN, -NR 5 R 6 , -SF 5 , - SO 2 R 12 , wherein R 5 and R 6 are independently selected from H and methyl, and R 12 is -F substituted methyl.
  • the substituents contained in Ring A are selected from -F, -CH 3 , -CHF 2 , -CF 3 , -CH 2 NHCH 3 , -CN, -NH 2 , -SF 5 , -SO 2 CHF 2 .
  • Ring A is selected from halogen, alkyl, -CN, and the alkyl group is optionally further substituted with a substituent selected from halogen, -NR 5 R 6 , where R 5 , R 6 is independently selected from H, alkyl.
  • the substituent contained in Ring A is selected from halogen, C1-C6 alkyl, -CN, and the alkyl group is optionally further substituted with a substituent selected from halogen, -NR 5 R 6 , where R 5.
  • R 6 is independently selected from H and C1 ⁇ C6 alkyl.
  • the substituents contained in ring A are selected from -F, methyl, -F substituted methyl, -NR 5 R 6 substituted methyl, -CN, wherein R 5 and R 6 are independently selected from H, methyl.
  • Ring A contains said substituent selected from -F, -CH 3 , -CHF 2 , -CF 3 , -CH 2 NHCH 3 , -CN, -NH 2 .
  • Ring A is selected from:
  • Ring A is selected from:
  • R a , R b , and R c are independently selected from H and C1 to C6 alkyl groups.
  • R a , R b , and R c are independently selected from H and methyl.
  • R a is methyl
  • R b is H
  • R c is H
  • R 1 , R 3 and R 4 are independently selected from H, alkyl, alkoxy, -OH, -CN, -NR 7 R 8 , and the alkyl and alkoxy groups are optionally selected from halogen.
  • -NR 7 R 8 is substituted by the substituent, wherein R 7 and R 8 are independently selected from H and alkyl.
  • R 1 , R 3 , and R 4 are independently selected from H, C1 to C6 alkyl, C1 to C6 alkoxy, -OH, -CN, -NR 7 R 8 , and the alkyl and alkoxy groups can be Optionally, it is substituted by a substituent selected from halogen and -NR 7 R 8 , wherein R 7 and R 8 are independently selected from H and C1 to C6 alkyl.
  • R 1 , R 3 , and R 4 are independently selected from H, C1 to C6 alkyl, C1 to C6 alkoxy, -OH, and -CN.
  • R 1 , R 3 and R 4 are independently selected from H, methyl, methoxy, -OH, -CN.
  • R 1 is selected from H, methyl, methoxy, -CN.
  • R3 is H.
  • R 4 is selected from methyl, methoxy, -OH.
  • R 1 , R 3 and R 4 are independently selected from H, alkyl, alkoxy, -OH, -NR 7 R 8 , and the alkyl and alkoxy are optionally selected from halogen, -NR 7 R 8 is substituted by a substituent, wherein R 7 and R 8 are independently selected from H and alkyl.
  • R 1 , R 3 and R 4 are independently selected from H and alkyl.
  • R 1 , R 3 and R 4 are independently selected from H and C1 to C6 alkyl.
  • R 1 , R 3 and R 4 are independently selected from H and methyl.
  • R1 is H.
  • R3 is H.
  • R 4 is methyl.
  • R 1 and R 3 are independently selected from H, halogen, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 alkylthio, -OH, -CN, -NR 7 R 8 , and the alkyl
  • the radical and alkoxy group are optionally substituted with substituents selected from halogen and -NR 7 R 8 , where R 7 and R 8 are independently selected from H and C1 to C6 alkyl.
  • R 1 and R 3 are independently selected from H, C1 to C6 alkyl, C1 to C6 alkoxy, and -CN.
  • R 1 and R 3 are independently selected from H, methyl, methoxy, and -CN.
  • R 1 is selected from H, methyl, methoxy, -CN.
  • R 1 is selected from H, methyl, methoxy.
  • R3 is H.
  • R 4 is selected from C1 to C6 alkyl and -OH.
  • R 4 is halogen
  • R 4 is chlorine
  • R 13 is an alkyl group
  • X is a single bond
  • R 2 is -XR 13 , 9 R 10 , -NR 22 R 23 is substituted by the substituent, and R 9 , R 10 , R 22 , and R 23 are independently selected from C1 to C6 alkyl groups.
  • R2 is -XR13 , Substituted with N(CH 3 ) 2 and -N(CH 3 ) 2 substituents.
  • R 2 is selected from -CH 3 ,
  • R 2 is -XR 13 , 9 R 10 is substituted by a substituent, and R 9 and R 10 are independently selected from C1 to C6 alkyl groups.
  • R2 is -XR13 , Substituted with N(CH 3 ) 2 substituent.
  • R 2 is selected from -CH 3 ,
  • R 2 is -XR 13 , From heteroatoms of N, O, and S, the aryl and heteroaryl groups are optionally selected from halogen, C1 to C6 alkyl, C1 to C6 alkoxy, -OH, -CN, -NR 14 R 15 Substituted with substituents, R 14 and R 15 are independently selected from H and C1 to C6 alkyl.
  • R2 is -XR13 , S heteroatom, the phenyl and heteroaryl groups are optionally substituted with substituents selected from halogen, C1 ⁇ C6 alkyl, C1 ⁇ C6 alkoxy, -OH, -CN, -NR 14 R 15 , R 14 and R 15 are independently selected from H and C1 to C6 alkyl.
  • R 2 is -XR 13
  • X is a single bond
  • R 13 is pyridyl or pyrimidinyl
  • the pyridyl or pyrimidinyl is optionally substituted by a C1 to C6 alkoxy group.
  • R 2 is selected from
  • R 2 is -XR 13 , X is methylene and a single bond, R 13 is phenyl, 5-6 membered heteroaryl, and the heteroaryl contains 1 selected from N, O, S Heteroatom, the phenyl and heteroaryl groups are optionally substituted with substituents selected from halogen, C1 ⁇ C6 alkyl, C1 ⁇ C6 alkoxy, -OH, -CN, -NR 14 R 15 , R 14 and R 15 are independently selected from H and C1-C6 alkyl.
  • R 2 is -XR 13
  • X is a single bond
  • R 13 is a pyridyl group
  • the pyridyl group is optionally substituted by a C1 to C6 alkoxy group.
  • R 2 is
  • R 2 is -XR 13
  • X is selected from C1 to C3 alkylene, a single bond
  • R 13 is selected from 3 to 10 membered cycloalkyl, 3 to 10 membered heterocycloalkyl, 3 to 10 membered ring Alkenyl, 3-10 membered heterocyclic alkenyl, the heterocycloalkyl, heterocycloalkenyl contains heteroatoms selected from N, O, S, the cycloalkyl, heterocycloalkyl, cycloalkenyl,
  • the heterocyclenyl group is optionally selected from halogen, C1-C6 alkyl, -YR 16 is substituted with a substituent, and the C1 to C6 alkyl group is optionally further substituted with a substituent selected from halogen, -OH, C1 to C6 alkoxy group, and -NR 17 R 18 .
  • R 2 is -XR 13 .
  • the base contains heteroatoms selected from N, O, and S, and the cycloalkyl and heterocycloalkyl groups are optionally selected from halogen, C1 to C6 alkyl, -YR 16 is substituted with a substituent, and the C1 to C6 alkyl group is optionally further substituted with a substituent selected from halogen and -OH.
  • R 2 is -XR 13 . ⁇ 2 heteroatoms selected from N, O, S.
  • the cycloalkyl and heterocycloalkyl groups are optionally selected from halogen, C1 ⁇ C6 alkyl, -YR 16 is substituted with a substituent, and the C1 to C6 alkyl group is optionally further substituted with a substituent selected from halogen and -OH.
  • R 2 is -XR 13
  • X is selected from methylene, a single bond
  • R 13 is selected from cyclopropyl, cyclobutyl, cyclopentyl, bicyclo[1.1.1]pentyl, Cyclohexyl, adamantyl, azetidinyl, tetrahydrofuryl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, 1,4-ethylene piperidinyl, morpholinyl, the above groups are optionally selected from halogen, C1 ⁇ C6 alkyl, -YR 16 is substituted with a substituent, and the C1 to C6 alkyl group is optionally further substituted with a substituent selected from halogen and -OH.
  • R 2 is -XR 13
  • X is selected from methylene and a single bond
  • R 13 is selected from 3 to 6-membered cycloalkyl, 5 to 8-membered heterocycloalkyl
  • the heterocycloalkyl contains 1 ⁇ 2 heteroatoms selected from N, O, S.
  • the cycloalkyl and heterocycloalkyl groups are optionally selected from halogen, C1 ⁇ C6 alkyl, -YR 16 is substituted with a substituent, and the C1 to C6 alkyl group is optionally further substituted with a substituent selected from halogen and -OH.
  • R 2 is -XR 13
  • X is selected from methylene, a single bond
  • R 13 is selected from cyclopropyl, cyclobutyl, cyclopentyl, bicyclo[1.1.1]pentyl, Cyclohexyl, tetrahydrofuryl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydropyrrolyl, piperidinyl, 1,4-ethylenepiperidinyl, morpholinyl
  • the above groups are optional Selected from halogen, C1 ⁇ C6 alkyl, -YR 16 is substituted with a substituent, and the C1 to C6 alkyl group is optionally further substituted with a substituent selected from halogen and -OH.
  • R 2 is -XR 13 , group, -YR 16 substituent, and the C1 to C6 alkyl group is optionally further substituted by halogen.
  • R 2 is -XR 13
  • X is selected from methylene, a single bond
  • R 13 is selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, the above groups are optional It is substituted by a substituent selected from halogen, C1-C6 alkyl, and -YR 16 , and the C1-C6 alkyl is optionally further substituted by halogen.
  • R 17 and R 18 are independently selected from H and C1 to C6 alkyl.
  • R 17 and R 18 are independently selected from H and methyl.
  • R 17 and R 18 are both methyl.
  • R 2 is -XR 13
  • X is selected from methylene, a single bond
  • R 13 is selected from cyclopropyl, cyclobutyl, cyclopentyl, bicyclo[1.1.1]pentyl, Cyclohexyl, adamantyl, azetidinyl, tetrahydrofuryl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, 1,4-ethylene piperidinyl, morpholinyl, the above groups are optionally selected from -F, methyl, is substituted with a substituent, wherein the methyl group is optionally further substituted with a substituent selected from -F, -OH.
  • R2 is selected from:
  • R 2 is -XR 13
  • X is selected from methylene, a single bond
  • R 13 is selected from cyclopropyl, cyclobutyl, cyclopentyl, bicyclo[1.1.1]pentyl, Cyclohexyl, tetrahydrofuryl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydropyrrolyl, piperidinyl, 1,4-ethylenepiperidinyl, morpholinyl, the above groups are optional Selected from -F, methyl, is substituted with a substituent, wherein the methyl group is optionally further substituted with a substituent selected from -F, -OH.
  • R2 is selected from:
  • R 2 is -XR 13
  • X is a single bond
  • R 13 is selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the above groups are optionally selected from -F , substituted by a methyl substituent, wherein the methyl group is optionally further substituted by -F.
  • R2 is selected from:
  • Y is a C1 to C3 alkylene group
  • R 16 is selected from a 3 to 10 membered cycloalkyl group, a 3 to 10 membered heterocycloalkyl group, a 3 to 10 membered cycloalkenyl group, and a 3 to 10 membered heterocyclic alkenyl group.
  • 6-12-membered aryl group, 5-12-membered heteroaryl group, the heterocycloalkyl group, heterocycloalkenyl group, and heteroaryl group contain heteroatoms selected from N, O, and S.
  • Y is a C1-C3 alkylene group
  • R 16 is a 3-10 membered heterocycloalkyl group
  • the heterocycloalkyl group contains 1 heteroatom selected from N, O, and S.
  • Y is methylene
  • R 16 is a 5-membered heterocycloalkyl group
  • the heterocycloalkyl group contains 1 heteroatom selected from N, O, and S.
  • Y is methylene
  • R 16 is tetrahydropyrrolyl
  • R 2 is
  • Y is selected from R 16 is selected from C1 ⁇ C6 alkyl, C1 ⁇ C6 alkoxy, -NR 17 R 18 , 3 ⁇ 10 membered cycloalkyl, 3 ⁇ 10 membered heterocycloalkyl, 3 ⁇ 10 membered cycloalkenyl, 3 ⁇ 10-membered heterocycloalkenyl, 6-12-membered aryl, 5-12-membered heteroaryl, and the heterocycloalkyl, heterocycloalkenyl, and heteroaryl contain heteroatoms selected from N, O, and S.
  • Y is selected from R 16 is selected from C1 to C6 alkyl, C1 to C6 alkoxy, -NR 17 R 18 , 3 to 10 membered heterocycloalkyl, 6 to 12 membered aryl, and 5 to 12 membered heteroaryl.
  • Cycloalkyl and heteroaryl groups contain 1 to 2 heteroatoms selected from N, O, and S.
  • Y is selected from R 16 is selected from C1 to C6 alkyl, C1 to C6 alkoxy, -NR 17 R 18 , 5 to 6 membered heterocycloalkyl, 6 to 6 membered aryl, and 5 to 6 membered heteroaryl, and the heterocycloalkyl
  • the base and heteroaryl group contain 1 to 2 heteroatoms selected from N and O.
  • Y is selected from R 16 is selected from methyl, methoxy, -N(CH 3 ) 2 , morpholinyl, phenyl, and isoxazolyl.
  • R 2 is selected from
  • R 2 is selected from
  • R 2 is selected from 3 to 6-membered cycloalkyl, 3 to 6-membered heterocycloalkyl, the heterocycloalkyl contains 1 to 2 heteroatoms selected from N, O, and S, and the cycloalkyl
  • R 2 is selected from 3-6 membered cycloalkyl and 5-6 membered heterocycloalkyl.
  • the heterocycloalkyl contains 1 heteroatom selected from O and S.
  • R2 is selected from:
  • the compound is selected from one of the following:
  • compositions which contain the compounds, or their tautomers, stereoisomers, solvates, metabolites, isotope labels, pharmaceutically acceptable salts, co-crystals, and pharmaceutical compositions. acceptable excipients or auxiliary ingredients.
  • the invention provides the compound, or its tautomer, stereoisomer, solvate, metabolite, isotope label, pharmaceutically acceptable salt, co-crystal, or the pharmaceutical composition in Use in the preparation of SOS1 inhibitors.
  • the invention provides the compound, or its tautomer, stereoisomer, solvate, metabolite, isotope label, pharmaceutically acceptable salt, co-crystal, or the pharmaceutical composition in Use in the preparation of medicaments for the treatment of diseases mediated by SOS1.
  • the disease is selected from the group consisting of cancer and pathogenic rash diseases.
  • the cancer is selected from: non-small cell lung cancer, lung cancer, pancreatic cancer, ovarian cancer, bladder cancer, prostate cancer, chronic myelogenous leukemia, colorectal cancer, brain cancer, liver cancer, kidney cancer, gastric cancer, and breast cancer. ;
  • the pathogenic rash disease is selected from the group consisting of: Noonan syndrome, cardiofaciocutaneous syndrome, and type I hereditary gingival fibromatosis.
  • the invention provides the compound, or its tautomer, stereoisomer, solvate, metabolite, isotope label, pharmaceutically acceptable salt, co-crystal, or the pharmaceutical composition in Use in the preparation of drugs for treating diseases resulting in overexpression of SOS1 protein.
  • the invention provides the compound, or its tautomer, stereoisomer, solvate, metabolite, isotope label, pharmaceutically acceptable salt, co-crystal, or the pharmaceutical composition in Use in preparing drugs for treating diseases caused by overexpression of SOS1 protein.
  • the present invention also provides pharmaceutical compositions containing the compounds, or their tautomers, stereoisomers, solvates, metabolites, isotope markers, and pharmaceutically acceptable salts administered simultaneously or separately. , cocrystals, and RAS inhibitors.
  • the RAS inhibitor is a KRAS inhibitor.
  • the RAS inhibitor is a KRAS G12C inhibitor.
  • the RAS inhibitor is Adagrasib.
  • the present invention also provides the use of the pharmaceutical composition in preparing drugs for treating cancer.
  • the cancer is selected from non-small cell lung cancer and pancreatic cancer.
  • the present invention provides a method for treating diseases mediated by SOS1, which includes administering to a subject the compound, or its tautomer, stereoisomer, solvate, metabolite, isotope label, pharmaceutical acceptable salts, co-crystals, or steps of the pharmaceutical compositions described above.
  • the present invention provides a method for treating diseases that cause overexpression of SOS1 protein, which includes administering to a subject the compound, or its tautomer, stereoisomer, solvate, metabolite, isotope label, Pharmaceutically acceptable salts, co-crystals, or steps of the pharmaceutical compositions described.
  • the present invention provides a method for treating diseases caused by overexpression of SOS1 protein, which includes administering to a subject the compound, or its tautomer, stereoisomer, solvate, metabolite, isotope label, Pharmaceutically acceptable salts, co-crystals, or steps of the pharmaceutical compositions described.
  • the amount of drug administered to the subject is an effective amount.
  • Tautomers refer to functional group isomers resulting from the movement of an atom in a molecule between two positions, especially when there are mobile hydrogen atoms in the molecule, such as ketone formula and enol form Tautomers.
  • stereoisomers refer to isomers produced by the interconnection order of atoms or atomic groups in the molecule with the same order but different spatial arrangement, including cis-trans isomers, optical isomers, and conformational isomers.
  • the stereoisomers described in the present invention also include mixtures of two or more stereoisomers, such as mixtures of enantiomers and/or diastereomers in any ratio.
  • isotope label means that one or more atoms in the molecule are replaced by atoms with different atomic masses or mass numbers.
  • isotopes that may be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine and iodine, such as, but not limited to, 2 H, 3 H, 11 C, 13 C, 14 C respectively. , 13 N, 15 N, 15 O, 17 O, 18 O, 35 S, 18 F, 36 Cl, 123 I and 125 I.
  • Certain isotope-labeled compounds of the invention can be used for drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e.
  • a compound of the invention may be enriched in 1%, 2%, 5%, 10%, 25%, 50%, 75%, 90%, 95% or 99% of a specified isotope.
  • compounds of the invention substituted with heavier isotopes such as deuterium, ie, 2H , may provide certain therapeutic advantages.
  • the "pharmaceutically acceptable salt” mentioned in the present invention refers to the salt formed by the compound of the present invention and an acid or a base suitable for use as a medicine.
  • the above-mentioned acids and bases are generalized Lewis acids and bases.
  • Suitable salt-forming acids include inorganic acids, organic acids, and acidic amino acids.
  • Solvate as used herein means an association formed between one or more solvent molecules and a compound of the present invention suitable for use as a pharmaceutical.
  • form solvation Solvents include water and organic solvents.
  • ring refers to any covalently closed structure, including, for example, carbocyclic rings (such as aryl or cycloalkyl), heterocyclic rings (such as heteroaryl or heterocycloalkyl), aromatic groups (such as aryl or heteroaryl), non-aryl (such as cycloalkyl or heterocycloalkyl).
  • the "ring” mentioned in the present invention may be a single ring or a polycyclic ring (including a bicyclic ring), and may be a fused ring, a spiro ring or a bridged ring.
  • unit represents the number of ring atoms constituting the ring skeleton.
  • optionally substituted means that it can be substituted by one or more (including two) specified substituents, or it can be unsubstituted.
  • cycloalkyl refers to a saturated carbocyclic hydrocarbon group having a monocyclic or polycyclic ring.
  • 3--10-membered cycloalkyl refers to a saturated monocyclic or polycyclic hydrocarbon ring with 3-10 ring carbon atoms. Examples include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, bicyclo [1.1.1] Pentyl, cyclohexyl, adamantyl.
  • the structural formula of cyclopropane is The structural formula of cyclobutane is The structural formula of cyclopentane is The structural formula of bicyclo[1.1.1]pentane is The structural formula of cyclohexane is The structural formula of adamantane is As otherwise indicated, the cycloalkyl group is optionally substituted with one or more suitable substituents.
  • heterocycloalkyl refers to a saturated cyclic group having a monocyclic or polycyclic ring, in which at least one ring atom is a heteroatom and the remaining ring atoms are carbon. Heteroatoms include but are not limited to N, O, S, P, Si, etc., preferably N, O, S.
  • heterocycloalkyl refers to a saturated monocyclic or polycyclic group with 3-10 ring atoms, in which at least one ring atom is a heteroatom and the remaining ring atoms are carbon. Examples include but are not Limited to: As otherwise indicated, the heterocycloalkyl group is optionally substituted with one or more suitable substituents.
  • 3-10-membered cycloalkenyl refers to an unsaturated non-aromatic monocyclic or polycyclic carbocyclic hydrocarbon group with 3-10 ring carbon atoms. Examples include but are not limited to cyclopropenyl, cyclobutenyl, and cyclopentenyl. Alkenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, etc. As otherwise indicated, the cycloalkenyl group is optionally substituted with one or more suitable substituents.
  • heterocycloalkenyl refers to a non-aromatic monocyclic or polycyclic group containing at least one double bond, in which at least one ring atom is a heteroatom and the remaining ring atoms are carbon. Heteroatoms include but are not limited to N, O, S, P, Si, etc., preferably N, O, S. Examples of “3-10 membered heterocyclic alkenyl” include but are not limited to: As otherwise indicated, the heterocycloalkenyl group is optionally substituted with one or more suitable substituents.
  • aryl refers to an all-carbon monocyclic or polycyclic aromatic group with a conjugated ⁇ electron system, including monocyclic aryl (such as a 6-membered ring phenyl), condensed ring aryl (having an aromatic ring and an aromatic The rings share ring edges, such as naphthyl in a 10-membered ring) and biaryl (connected through single bonds to form an aryl-aryl structure, such as biphenyl in a 12-membered ring). As otherwise indicated, the aryl groups are optionally substituted with one or more suitable substituents.
  • heteroaryl refers to a monocyclic or polycyclic aromatic group with a conjugated ⁇ electron system, in which at least one ring atom is a heteroatom and the remaining ring atoms are carbon. Heteroatoms are preferably N, O, S.
  • the heteroaryl group described in the present invention includes monocyclic heteroaryl groups (such as 5-membered ring thienyl, 5-membered ring isoxazolyl, 6-membered ring pyridyl), condensed ring heteroaryl (with heteroaryl Rings and heteroaromatic rings share ring edges, or aromatic rings and heteroaromatic rings share ring edges, such as 9-membered benzothienyl), bisheteroaryl (connected through a single bond to form a heteroaryl-heteroaryl structure, Or an aryl-heteroaryl structure, such as an 11-membered phenyl-thienyl ring: ).
  • the heteroaryl groups are optionally substituted with one or more suitable substituents.
  • halogen includes F, Cl, Br or I.
  • alkyl refers to a straight or branched chain saturated aliphatic hydrocarbon group.
  • C1 ⁇ C6 alkyl refers to a straight-chain or branched saturated aliphatic hydrocarbon group with 1, 2, 3, 4, 5 or 6 carbon atoms. Examples include but are not limited to methyl, ethyl, n-propyl, Isopropyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, etc. As otherwise indicated, the alkyl groups are optionally substituted with one or more suitable substituents.
  • alkylene refers to a saturated divalent hydrocarbon group obtained by removing two hydrogen atoms from a saturated linear or branched aliphatic hydrocarbon group; that is, one of the hydrogens in the alkyl group is substituted, and the alkyl group is as defined above described.
  • Examples of "C1-C3 alkylene” include but are not limited to methylene (-CH 2 -), ethylene ⁇ including -CH 2 CH 2 - Or -CH(CH 3 )- ⁇ , isopropylene ⁇ including -CH(CH 3 )CH 2 - or -C(CH 3 ) 2 - ⁇ , etc.
  • alkoxy refers to an "alkyl” group as defined above connected through an oxygen atom, i.e. an "alkoxy” group may be defined as -OR, where R is an alkyl group as defined above.
  • Examples of "C1-C6 alkoxy” include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentyloxy base, n-hexyloxy group, etc.
  • the alkoxy groups are optionally substituted with one or more suitable substituents.
  • alkylthio refers to an "alkyl” group as defined above connected through a sulfur atom, i.e. an "alkylthio" group may be defined as -SR, where R is an alkyl group as defined above. As otherwise indicated, the alkylthio group is optionally substituted with one or more suitable substituents.
  • the pharmaceutically acceptable excipients of the present invention are the general term for all additional materials in medicines other than the main drug.
  • the excipients should have the following properties: (1) No toxic effects on the human body and almost no side effects; (2) Stable chemical properties , not easily affected by temperature, pH, storage time, etc.; (3) It has no incompatibility with the main drug and does not affect the efficacy and quality inspection of the main drug; (4) It does not interact with packaging materials.
  • the auxiliary materials in the present invention include, but are not limited to, fillers (diluents), lubricants (glidants or anti-adhesive agents), dispersants, wetting agents, adhesives, regulators, solubilizers, antioxidants, and bacteriostatic agents.
  • the administration mode of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative administration modes include (but are not limited to): oral, parenteral (intravenous, intramuscular or subcutaneous), and topical administration.
  • subject includes humans or non-human animals.
  • exemplary human subjects include subjects (referred to as patients) suffering from a disease, such as those described herein, or normal individuals.
  • Non-human animals include all vertebrate animals, such as non-mammals and mammals, such as non-human primates, domestic animals and/or domesticated animals.
  • an "effective amount” refers to an amount of a compound that, when administered, alleviates to a certain extent one or more symptoms of the disease being treated. Dosage regimens can be adjusted to provide the best desired response.
  • the beneficial effects of the present invention are: the present invention provides a series of compounds with obvious inhibitory effects on SOS1 protein, and provides new solutions for the treatment of diseases with SOS1 as the target, such as cancer, pathogenic rash diseases, etc. Compared with existing SOS1 inhibitors, the compounds of the present invention have significant improvements in activity, druggability, etc., especially better liver microsome stability, good druggability, and can be used to prepare drugs for the treatment of related diseases. Broad application prospects.
  • the structure of the compound of the present invention is determined by nuclear magnetic resonance (NMR) or/and liquid mass spectrometry (LC-MS). Chemical shifts ( ⁇ ) for NMR are given in units of parts per million (ppm). NMR was measured using an AVANCE NEO 400MHz Bruker instrument. The measurement solvents were deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), and deuterated methanol (CD 3 OD). The internal standard was tetramethylmethane. silane (TMS). MS was measured using an ISQ-EC Thermo Fisher LC-MS instrument. Prep-HPLC is a GX-281Gilson chromatograph.
  • the separation methods are: (Method 1) Sun Fire Prep C18 OBDTM 5 ⁇ m, 30 ⁇ 150mm Column, 0.04% HCl aqueous solution/acetonitrile; (Method 2) Sun Fire Prep C18 OBDTM 5 ⁇ m, 30 ⁇ 150mm Column , 0.06 % formic acid aqueous solution/acetonitrile; (Method 3) % aqueous trifluoroacetic acid/acetonitrile.
  • the starting materials in the embodiments of the present invention are known and can be purchased on the market, or can be synthesized according to methods known in the art.
  • reaction temperature is room temperature, which is 20°C to 30°C.
  • DIPEA N,N-diisopropylethylamine
  • HATU 2-(7-azobenzotriazole)-N,N,N',N'-tetramethylurea hexafluorophosphate
  • NBS N-bromosuccinimide
  • reaction solution was extracted with methyl tert-butyl ether (100 mL), the organic phases were combined, washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain 2-formyl- 3-Oxoglutarate dimethyl ester was directly used in the next reaction without further purification, with a yield of 98.6%; ESI-MS (m/z): 203.2[M+H] + .
  • the preparation method refers to Example 1.
  • reaction solution was cooled to room temperature, saturated aqueous ammonium chloride solution (40 mL) was added to quench the reaction, extracted with dichloromethane (50 mL ⁇ 2), the organic phases were combined, and washed with saturated brine (50 mL ⁇ 3). Dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • reaction solution was extracted with ethyl acetate (100 mL ⁇ 3), the organic phases were combined, washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain 2-formyl-3 -Dimethyl oxoglutarate, yield 99.9%; ESI-MS (m/z): 203.2 [M+H] + .
  • Tetrahydro-2H-pyran-4-amine (2.0g, 19.773mmol) and 2-formyl-3-oxoglutarate dimethyl ester (4.8g, 23.727mmol) were dissolved in methanol (120mL) and iced.
  • sodium methoxide (2.1g, 39.545mmol) under a water bath and react at 80°C for 6 hours.
  • the reaction solution was cooled to room temperature, concentrated under reduced pressure to remove most of the solvent, and saturated aqueous ammonium chloride solution (100 mL) was added to quench the reaction.
  • Extract 50mL 1N
  • saturated sodium bicarbonate aqueous solution adjust the reaction solution to a neutral pH
  • separate the organic phase extract the aqueous phase with ethyl acetate (50mL ⁇ 2)
  • combine the organic phases and use saturated salt Wash with water (50 mL), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • reaction solution was lowered to room temperature, filtered, the filter cake was rinsed with ethanol (5mL ⁇ 2), and dried under vacuum to obtain 4-hydroxy-1-methyl-6-(tetrahydro-2H-pyran-4-yl)pyrido [3,4-d]pyridazin-7(6H)-one, yield 88.3%; ESI-MS (m/z): 262.2[M+H] + .
  • Acetonitrile (10 mL) and 4-hydroxy-1-methyl-6-(tetrahydro-2H-pyran-4-yl)pyrido[3,4-d]pyridazin-7(6H)-one 200 mg ,0.765mmol
  • add phosphorus oxybromide 2.2g, 7.655mmol
  • cool the reaction solution to 0°C add saturated sodium bicarbonate aqueous solution to adjust the pH value of the reaction solution to neutral, extract with dichloromethane (20 mL ⁇ 3), combine the organic phases, and use saturated brine (20 mL).
  • the preparation method refers to Example 1.
  • the preparation method refers to Example 8.
  • the preparation method refers to Example 1.
  • the preparation method refers to Example 1.
  • reaction solution was cooled to room temperature, 30% potassium fluoride aqueous solution (100 mL) was added to quench the reaction, filtered, the filtrate was extracted with ethyl acetate (20 mL ⁇ 3), the organic phases were combined, and saturated brine (20 mL ⁇ 3) was added.
  • reaction solution is cooled to room temperature, filtered, and the filter cake is rinsed with a small amount of ethanol and dried under vacuum to obtain 6-cyclobutyl-4-hydroxy-1-methylpyrido[3,4-d]pyridazine- 7(6H)-ketone, yield 93.3%; ESI-MS (m/z): 232.1[M+H] + .
  • 6-Cyclobutyl-4-hydroxy-1-methylpyrido[3,4-d]pyridazin-7(6H)-one 500g, 2.165mmol
  • phosphorus oxybromide 1.2g, 4.330mmol
  • acetonitrile 50 mL
  • reaction solution was cooled to room temperature, quenched by slowly adding saturated sodium bicarbonate aqueous solution (50 mL) dropwise, extracted with dichloromethane (50 mL ⁇ 3), the organic phases were combined, washed with saturated brine (50 mL), and dried over anhydrous sodium sulfate.
  • step af of Example 12 For the preparation method, refer to step af of Example 12 to obtain 2-(4-(((R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethyl)amino)-1-methyl -7-Oxopyrido[3,4-d]pyridazine-6(7H)-yl)propionic acid methyl ester, yield 55.4%; ESI-MS (m/z): 435.4[M+H] + .
  • Step h): 2-(4-(((R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethyl)amino)-1-methyl-7-oxopyrido Preparation of [3,4-d]pyridazine-6(7H)-yl)-N,N-dimethylpropionamide
  • reaction solution is cooled to room temperature, filtered, and the filter cake is rinsed with ethanol (10 mL) and dried under vacuum to obtain 6-cyclopropyl-1-methylpyrido[3,4-d]pyridazine-4, 7(3H,6H)-diketone, yield 57.9%; ESI-MS (m/z): 218.2[M+H] + .
  • Example 28 Referring to the preparation method of Example 28 and using corresponding raw materials, the compounds in the following examples were prepared.
  • the aqueous phase is extracted with ethyl acetate (50 mL ⁇ 3).
  • the organic phases are combined and washed with saturated brine. (100 mL), washed with anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure.
  • the preparation method refers to step ai of Example 32.
  • the obtained crude product is purified by Prep-HPLC (separation method 2) to obtain (R)-4-((1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-6-(2-hydroxyethyl)-1-methylpyrido[3,4-d]pyridazine-7(6H)-onecarboxylate, yield 18.4%;
  • the preparation method refers to step ai of Example 32.
  • the crude product obtained is purified by Prep-HPLC (separation method 3) to obtain 4-(((R)-1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-6-(4-hydroxybut-2-yl)-1-methylpyrido[3,4-d]pyridazin-7(6H)-one, yield 26.7%;
  • the preparation method refers to step ai of Example 32.
  • the obtained crude product is purified by Prep-HPLC (separation method 3) to obtain (R)-4-((1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-6-(1-(hydroxymethyl)cyclopropyl)-1-methylpyrido[3,4-d]pyridazin-7(6H)-one, yield 23.7%;
  • Example 35 For the preparation method, refer to Example 35.
  • the crude product obtained is purified by Prep-HPLC (separation method 3) to obtain (R)-4-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl) Amino)-6-(1-(difluoromethyl)cyclopropyl)-1-methylpyrido[3,4-d]pyridazin-7(6H)-one, yield 15.7%;
  • Example 28 For the preparation method, refer to Example 28.
  • the crude product obtained is purified by Prep-HPLC (separation method 3) to obtain 4-(((R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethyl) Amino)-1-methyl-6-((tetrahydrofuran-3-yl)methyl)pyrido[3,4-d]pyridazin-7(6H)-one, yield 21.8%;
  • 1 H NMR 400MHz ,DMSO-d 6 ) ⁇ 9.28(s,1H),7.62-7.56(m,2H),7.50-7.47(m,1H),7.37-7.10(m,2H),6.56(s,1H),5.65 -5.59(m,1H),4.20-4.06(m,2H),3.88-3.83(m,1H),3.69-3.56(m,3H),2.88-2.78(m,1H),2.35(s,3H)
  • reaction solution was cooled to room temperature, added acetonitrile (100 mL), stirred at room temperature for 10 min, filtered, and the filter cake was rinsed with acetonitrile (10 mL) and dried under vacuum to obtain 6- (1-methylcyclopropyl)-2,3-dihydropyrido[3,4-d]pyridazine-1,4,7(6H)-trione, yield 68.3%; ESI-MS(m/z): 234.0[M+H] + .
  • 6-(1-Methylcyclopropyl)-2,3-dihydropyrido[3,4-d]pyridazine-1,4,7(6H)-trione 780mg, 3.348mmol was dissolved at room temperature.
  • phosphine oxybromide (1.9g, 6.696mmol) and acetonitrile (70mL) were added to the reaction bottle, and the temperature was raised to 105°C for 3 hours.
  • 1,4-Dibromo-6-(1-methylcyclopropyl)pyrido[3,4-d]pyridazin-7(6H)-one (205mg, 0.571mmol), DIPEA (369mg, 2.855mmol) ), (R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethane-1-amine hydrochloride (142 mg, 0.628 mmol) and 1,4-dioxane (5 mL ) into the microwave reaction bottle, react in the microwave reactor at 105°C for 1 hour, cool the reaction solution to room temperature, and concentrate under reduced pressure.
  • the preparation method refers to Example 45.
  • the crude product obtained is purified by Prep-HPLC (separation method 3) to obtain (R)-6-cyclopropyl-1-methyl-4-((1-(2-methyl-3- (Trifluoromethyl)phenyl)ethyl)amino)pyrido[3,4-d]pyridazin-7(6H)-one, yield 10.3%;
  • 6-Cyclopropyl-1,8-dimethylpyrido[3,4-d]pyridazine-4,7(3H,6H)-dione (300mg, 1.299mmol), phosphorus oxybromide (743mg , 2.597mmol) and acetonitrile (50mL) were added to the reaction bottle in sequence, and the temperature was raised to reflux under nitrogen protection and the reaction was stirred for 2 hours.
  • reaction solution was cooled to room temperature, saturated aqueous sodium bicarbonate solution (50 mL) was added to quench the reaction, extracted with dichloromethane (50 mL ⁇ 3), the organic phases were combined, washed with saturated brine (50 mL), and anhydrous. Dry over sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • Example 49 Referring to the preparation method of Example 49 and using corresponding raw materials, the compounds in the following examples were prepared.
  • the preparation method refers to Example 54.
  • the crude product obtained is purified by Prep-HPLC (separation method 3) to obtain (R)-6-(1-benzoylpiperidin-4-yl)-4-((1-(3- (Difluoromethyl)-2-fluorophenyl)ethyl)amino)-1-methylpyrido[3,4-d]pyridazin-7(6H)-one, yield 41.2%;
  • the preparation method refers to step af of Example 49.
  • the obtained crude product is purified by Prep-HPLC (separation method 3) to obtain 4-(((R)-1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-1-methyl-6-(quinuclidin-3-yl)pyrido[3,4-d]pyridazin-7(6H)-one, yield 10.4%;
  • 1 H NMR 400MHz, DMSO-d 6 ) ⁇ 9.03-8.91(m,1H),8.01-7.87(m,1H),7.62-7.53(m,1H),7.52-7.45(m,1H),7.37-7.12(m ,2H),6.61-6.57(m,1H),5.74-5.60(m,1H),5.07-4.90(m,1H),3.50-3.36(m,1H),3.19-3.07(m,1H),2.86 -2.65(
  • Example 56 Referring to the preparation method of Example 56 and using corresponding raw materials, the compounds in the following examples were prepared.
  • the preparation method refers to step af of Example 49.
  • the obtained crude product is purified by Prep-HPLC (separation method 3) to obtain (R)-4-((1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-6-(2-methoxypyridin-4-yl)-1-methylpyrido[3,4-d]pyridazin-7(6H)-one, yield 25.5%;
  • the preparation method refers to step af of Example 49.
  • the obtained crude product is purified by Prep-HPLC (separation method 3) to obtain (R)-4-((1-(3-(difluoromethyl)-2-fluorophenyl)) Ethyl)amino)-1-methyl-6-morpholino[3,4-d]pyridazin-7(6H)-one, yield 33.1%;
  • Step c): (R)-3-(4-(4-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl)amino)-1-methyl-7-oxo Preparation of pyrido[3,4-d]pyridazine-6(7H)-yl)piperidin-1-yl)-N,N-dimethyl-3-oxopropionamide
  • reaction solution was washed with saturated aqueous sodium bicarbonate solution (50 mL ⁇ 2) and saturated brine (30 mL) in sequence, dried over anhydrous sodium sulfate, and filtered.
  • (R)-4-(4-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl)amino)-1-methyl-7-oxopyrido Preparation of [3,4-d]pyridazine-6(7H)-yl)piperazine-1-carboxylic acid tert-butyl ester.
  • the preparation method refers to step af of Example 67, and is purified by Prep-HPLC (separation method 3) to obtain (R)-4-((1-(3-(difluoromethyl)-2-fluorophenyl)ethyl) )Amino)-1-methyl-6-(pyrimidin-5-yl)pyrido[3,4-d]pyridazin-7(6H)-one, yield 15.5%;
  • reaction solution was concentrated under reduced pressure, and saturated aqueous sodium bicarbonate solution (30 mL) and dichloromethane (30 mL) were added to the residue, stirred at room temperature for 5 min, left to separate, collect the organic phase, and extract the aqueous phase with dichloromethane (20 mL). ⁇ 2), combine the organic phases, dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
  • the preparation method refers to Example 68.
  • the crude product obtained is purified by Prep-HPLC (separation method 3) to obtain (R)-6-(1-acetyl azetidin-3-yl)-4-((1-(3) -(Difluoromethyl)-2-fluorophenyl)ethyl)amino)-1-methylpyrido[3,4-d]pyridazin-7(6H)-one; Yield 12.4%;
  • Example 76 For the preparation method, refer to Example 76.
  • reaction solution was cooled to room temperature, saturated aqueous ammonium chloride solution (20 mL) was added to quench the reaction, and extracted with dichloromethane (20 mL ⁇ 3). The organic phases were combined, washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure.
  • Example 78 Referring to the preparation method of Example 78 and using corresponding raw materials, the compounds in the following examples were prepared.
  • Example 82 Referring to the preparation method of Example 82 and using corresponding raw materials, the compounds in the following examples were prepared.
  • 6-Cyclopropyl-2,3-dihydropyrido[3,4-d]pyridazine-1,4,7(6H)-trione (8.8g, 68.493mmol) and acetonitrile (500mL) were added to the reaction In the bottle, add DIPEA (26.5g, 205.479mmol) and phosphorus oxychloride (31.5g, 205.479mmol) under stirring at room temperature, and raise the temperature to 100°C for 3 hours under nitrogen protection.
  • 1,4-Dichloro-6-cyclopropylpyrido[3,4-d]pyridazin-7(6H)-one 500mg, 1.953mmol
  • (R)-1-(3-(difluoro Methyl)-2-fluorophenyl)ethane-1-amine hydrochloride 439 mg, 1.953 mmol
  • 1,4-dioxane 10 mL
  • Example 86 Referring to the preparation method of Example 86 and using corresponding raw materials, the compounds in the following examples were prepared.
  • Example 93 Referring to the preparation method of Example 93 and using corresponding raw materials, the compounds in the following examples were prepared.
  • Test Example 1 SOS1 inhibitory activity measurement
  • the effect of SOS1 inhibitors on the interaction between SOS1 and KRAS protein was detected by homogeneous time-resolved fluorescence technology (HTRF) to evaluate its inhibitory level on SOS1 protein.
  • the protein and detection reagent used were KRAS-G12D/SOS1 BINDING ASSAY KIT (Cisbio).
  • the 2mM test compound stock solution was diluted 20 times (100 ⁇ M) with Diluent reagent, and then the Diluent reagent (5% DMSO) was used to perform a 5-fold concentration gradient. Dilute to obtain a total of 8 concentrations of working solution of the test compound.
  • Negative is the group without inhibitors
  • Blank is the group without enzyme
  • IC 50 by inhibition rate was calculated by GraphPad Prism software. Each compound was measured in 2 replicate wells. The IC 50 value of the compound of the present invention for inhibiting SOS1 activity is 3.7-308.0 nM.
  • the test data of some example compounds are as shown in Table 1:
  • the compound of the present invention has obvious inhibitory effect on SOS1 in vitro, can be used as an SOS1 inhibitor, and has broad application prospects in the fields of cancer, pathogenic rash diseases and other diseases mediated by SOS1 protein.
  • Metabolic stability affects the clearance rate, half-life and oral bioavailability of compounds in the body, and is one of the most important ADME pharmaceutical properties of compounds.
  • Liver microsomal stability assay is a common method to study metabolic stability.
  • the present invention uses rat liver microsome assay to examine the metabolic stability of compounds.
  • Preparation of compound stock solution Take appropriate amounts of the test compound and testosterone, prepare a 1mM stock solution with DMSO, and store it in a 4°C refrigerator.
  • NADPH working solution Weigh an appropriate amount of NADPH and use 16mM MgCl 2 to prepare a NADPH working solution with a concentration of 4.0mM.
  • rat liver microsome working solution Take an appropriate amount of rat liver microsome and dilute it with PBS to make a liver microsomal working solution of 1 mg/ml.
  • the T 1/2 of the compounds of the present invention was tested to be 9.8-129 min, and the CL (liver) was 14.3-45.4 ml/min/kg.
  • the stability test results of the compounds in some embodiments on rat liver microsomes are shown in Table 2:
  • MRTX0902 is a compound disclosed in WO2021127429 (Examples 12-10), which has been reported to have SOS1 inhibitory activity.
  • the plasma stability of compounds is also an important factor in the development of new drugs.
  • Good plasma stability is an important guarantee for good pharmacokinetics and pharmacodynamics.
  • the present invention incubates the compounds in the examples with the plasma of rats, mice, monkeys, dogs, and humans respectively at 37°C, and evaluates by measuring the sample concentrations at different incubation time points and calculating the corresponding remaining percentages. Stability of compounds in plasma of 5 species.
  • Compound intermediate concentration working solution Take an appropriate amount of the test compound and enalapril maleate 10mM stock solution, dilute it with DMSO to 1mM, and store it in a refrigerator at 4°C.
  • Blank plasma preparation Take appropriate amounts of frozen plasma from blank rats, mice, monkeys, dogs, and humans, and thaw them at 37°C.
  • Test Example 4 Measurement of liver drug metabolizing enzyme inhibitory activity
  • CYP450 The main site of drug metabolism is the liver, and the main component of the mixed-function oxidative enzyme system present in the liver is the CYP450 enzyme. This enzyme system causes most clinical drug interactions, which in turn leads to an increased incidence of adverse drug reactions.
  • CYP450 is a large supergene family composed of many isoenzymes. Among them, CYP3A4 is the main metabolic enzyme and participates in the metabolism of nearly half of the drugs in clinical practice. Similarly, CYP2C9 and CYP2C19 are also important metabolic enzymes and are clinically involved in the metabolism of a variety of drugs. Therefore, by testing the inhibitory activity of compounds on CYP450 enzymes in the early stage, the risk of drug-drug interactions can be judged and the safety of medication can be improved.
  • human liver microsomes are used as the CYP3A4 enzyme source, and the specific probe substrates of each CYP isoenzyme (CYP3A4 adopts two substrates, midazolam and testosterone, are CYP3A4-M and CYP3A4-, respectively. T represents), were incubated with a range of concentrations of the test compound in the presence of the cofactor NADPH.
  • Use LC-MS/MS to measure the production of metabolites of the probe substrate in the incubation system, calculate the IC 50 value of the test compound for each CYP450 enzyme subtype, and evaluate its inhibitory effect on each CYP450 enzyme subtype.
  • This experiment uses human pancreatic cancer MIA PaCa-2 cell subcutaneous xenograft tumor model to evaluate the anti-tumor activity of the compound.
  • TGI (%) [1-(average tumor volume at the end of administration in a certain treatment group - average tumor volume at the beginning of administration in this treatment group)/(average tumor volume at the end of administration in the vehicle control group) -The average tumor volume in the vehicle control group at the beginning of administration)] ⁇ 100%.
  • Body weight change (%) (average weight of animals at the end of administration in a certain treatment group - average weight of animals at the beginning of administration in this treatment group)/average weight of animals at the beginning of administration in this treatment group ⁇ 100 %.
  • This experiment uses human non-small cell lung cancer NCI-H358 cell subcutaneous xenograft tumor model to evaluate the anti-tumor activity of the compound.
  • TGI (%) [1-(average tumor volume at the end of administration in a certain treatment group - average tumor volume at the beginning of administration in this treatment group)/(average tumor volume at the end of administration in the vehicle control group) -The average tumor volume in the vehicle control group at the beginning of administration)] ⁇ 100%.
  • Body weight change (%) (average weight of animals at the end of administration in a certain treatment group - average weight of animals at the beginning of administration in this treatment group)/average weight of animals at the beginning of administration in this treatment group ⁇ 100 %.
  • the compounds of the present invention have obvious inhibitory effects on SOS1 in vitro, can be used as SOS1 inhibitors, have excellent anti-tumor effects in animals, and are effective in treating cancer and pathogenic rashes mediated by SOS1 protein. It has broad application prospects in fields such as diseases and other diseases.
  • the compound of the present invention compared with MRTX0902, has better liver microsomal metabolism stability and plasma stability, and has a better inhibitory effect on liver drug metabolism enzymes. It is weak, has significantly improved anti-tumor activity in vivo, and has good medicinal properties and clinical application prospects.

Landscapes

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

Abstract

涉及医药技术领域,特别是涉及一类用作SOS1蛋白抑制剂的哒嗪并吡啶酮衍生物及其用途。提供的化合物对SOS1蛋白活性具有明显的抑制作用,可作为SOS1蛋白抑制剂,成药性佳,可用以制备治疗由SOS1蛋白介导的癌症、病原性皮疹病等疾病的药物,具有广阔的应用前景。

Description

哒嗪并吡啶酮衍生物及其用途 技术领域
本发明涉及医药技术领域,特别是涉及一类用作SOS1蛋白抑制剂的哒嗪并吡啶酮衍生物及其用途。
背景技术
SOS1(son of sevenless homolog 1)蛋白是一种在细胞中广泛表达的调控蛋白,作为Ras或Rac1蛋白的一类鸟嘌呤核苷酸交换因子,在细胞内Ras或Rac1信号转导通路中起着重要的调控作用(Baltanás,F.C.;Zarich,N.;Rojas-J.M.;Santos,E.Biochim.Biophys.Acta.Rev.Cancer.2020,1874,188445)。SOS1蛋白在Ras信号转导途径中的作用是促进Ras释放GDP,结合GTP,使Ras蛋白由非活性状态转变为活性状态。
目前已知的RAS家族共有三个基因:KRAS,NRAS和HRAS。RAS酶的突变与肿瘤发生密切相关,在所有肿瘤中约25%检测到突变(de CastroJ.;Belda-Iniesta C.;Transl Lung Cancer Res.2013,2(2),142-51.),在不同类型的肿瘤中,RAS突变类型也不同。RAS突变(KRAS,NRAS,HRAS)存在于90%的胰腺癌,45%的结肠癌和35%的肺癌中。非小细胞肺癌(NSCLC)占所有肺癌病例的80%(Jemal,B.;et al,CA Cancer J.Clin.2011,61(2),69-90),Ras原癌基因是NSCLC中最常见的突变基因(Prior,L.et al,Cancer Res.2012,72(10),2457-2467;Li,L.et al,J Exp Clin Cancer Res.2018,37(1),178),其中,v-Ki-ras2Kirsten大鼠肉瘤病毒致癌基因(Kras)占肺腺癌RAS突变的90%(Hunter,J.C.;et al,Mol.Cancer Res.2015,13(9),1325-35)。
作为Kras的“心脏起搏器”,SOS1在细胞内许多信号转导通路中起着重要的调控作用。研究表明,抑制SOS1活性对于基于所有主要Kras基因突变所导致的癌细胞增殖具有有效的抑制作用(Kessler,D.;Gerlach,D.;Kraut,N.,McConnell,D.B.Curr.Opin.Chem.Biol.2021,62,109-118)。另外,一项在62例卵巢癌患者中的临床研究显示,RAS突变和SOS1突变在卵巢癌组织中表达显著上升,且预后上均与患者PFS缩短相关,提示RAS和SOS1靶向治疗在卵巢癌患者中有潜在价值。同时,SOS1基因突变也在其他很多癌细胞中发现例如胚胎横纹肌肉瘤、塞尔托利细胞睾丸肿瘤、皮肤颗粒细胞瘤(Denayer etal.,Genes Chromosomes Cancer,2010,49(3):242-52)和肺腺癌(Cancer Genome AtIas Research Network.,Nature 2014,511(7511),543-50)。另外,研究发现SOS1基因在膀胱癌(Watanabe etal.,IUBMB Life.,2000,49(4),317-20)及前列腺癌(Timofeeva et al.,Int.J.Oncol.,2009,35(4):751-60)中均有高表达。在慢性粒细胞白血病的发生发展过程中,BCR-ABL通过磷酸化活化GRB2、募集SOS1,从而持续性激活Ras/MAPK信号通路,导致造血干细胞的恶性增殖。因此,SOS1蛋白也是慢性粒细胞白血病治疗潜在的新靶标。除了癌症外,研究表明遗传性SOS1基因突变也与一些病原性皮疹病如努南综合症(NS),心面皮肤综合症(CFC)及I型遗传性牙龈纤维瘤病也密切相关(Pierre et al.,Biochem.Pharmacol.,2011,82(9):1049-56)。
SOS1抑制在机理上与SHP2抑制颇为相似(Nichols,R.J.;et al.Nat Cell Biol.2018,20(9),1064-1073),这表明SOS1抑制同样可以增强KRASG12C和MEK抑制剂的功效。初步数据显示,在多个G12和G13KRAS突变的PDX模型中,SOS1和MEK抑制之间存在显著的协同作用(Hofmann,M.H.;et al;Cancer Discov.2020,10.1158/2159-8290.CD-20-0142.)。除了抑制Kraswt的反馈激活外,由于KRASG12C变构抑制剂只能与KRASGDP结合,抑制SOS1具有通过增加突变体KRASG12C的数量来直接提高KRASG12C抑制剂的疗效的潜在优势(Hillig,R.C.,et al,Proc Natl Acad Sci U S A.2019,116(7),2551-2560)。虽然还有待进一步的研究,作为一种联合疗法的策略,抑制SOS1在临床上具有很大的应用潜力。
目前,全球范围内仍无针对SOS1靶点的药物上市,在研化合物均处于早期临床或临床前研究阶段。虽然有少数几个药研公司或研究机构对SOS1抑制剂均有相应研究并有相关专利公布,例如勃林格殷格翰公司公开了一类苄胺取代的喹唑啉衍生物用作SOS1抑制剂(US20190358230A1)以及拜耳制药公开了一类2-甲基-氮杂喹唑啉类化合 物用作SOS1抑制剂(WO2019201848A1),但现阶段已开发的SOS1抑制剂远未达到临床满足需求,开发新的SOS1抑制剂仍具有非常广阔的临床应用前景。
发明内容
本发明主要解决的技术问题是提供一类哒嗪并吡啶酮衍生物,对SOS1具有强效的选择性抑制作用。
为解决上述技术问题,本发明提供了式I所示的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶:
其中:
环A选自芳基、杂芳基,所述芳基、杂芳基可选地被选自卤素、烷基、烷氧基、-OH、-CN、-NR5R6、-SF5、-SO2R12的取代基所取代,所述烷基、烷氧基可选地进一步被选自卤素、-NR5R6的取代基所取代,其中,R5、R6独立选自H、烷基,R12为烷基且可选地被卤素所取代;
Ra、Rb、Rc独立选自H、烷基;
R1、R3、R4独立选自H、卤素、烷基、烯基、环烷基、环烯基、烷氧基、烷硫基、-OH、-CN、-NR7R8、-C(=O)H、-C(=O)NR19R20、-C(=O)OR21,所述烷基、烷氧基可选地被选自卤素、-NR7R8的取代基所取代,其中R7、R8、R19、R20、R21独立选自H、烷基;
R2为-X-R13,X选自亚烷基、一单键,R13选自烷基、环烷基、杂环烷基、环烯基、杂环烯基、芳基、杂芳基;其中:
当R13为烷基时,X为一单键,所述烷基可选地被选自-OH、烷氧基、卤素、-C(=O)NR9R10、-C(=O)OR11、-NR22R23的取代基所取代,R9、R10、R11、R22、R23独立选自H、烷基;
当R13选自芳基、杂芳基时,可选地被选自卤素、烷基、烷氧基、-OH、-CN、-NR14R15的取代基所取代,R14、R15独立选自H、烷基;
当R13选自环烷基、杂环烷基、环烯基、杂环烯基时,可选地被选自卤素、烷基、-Y-R16的取代基所取代,所述烷基可选地进一步被选自卤素、-OH、烷氧基、-NR17R18的取代基所取代;其中:
Y为亚烷基,R16选自环烷基、杂环烷基、环烯基、杂环烯基、芳基、杂芳基;
或者,Y选自R16选自烷基、烷氧基、-NR17R18、环烷基、杂环烷基、环烯基、杂环烯基、芳基、杂芳基;
R17、R18独立选自H、烷基。
进一步地,环A选自芳基、杂芳基,所述芳基、杂芳基可选地被选自卤素、烷基、烷氧基、-OH、-CN、-NR5R6的取代基所取代,所述烷基、烷氧基可选地进一步被选自卤素、-NR5R6的取代基所取代,其中R5、R6独立选自H、烷基。
进一步地,环A选自6~12元芳基、5~12元杂芳基,所述杂芳基含有选自N、O、S的杂原子,上述基团的取代基如前所定义。
优选地,环A选自6元芳基、5~11元杂芳基,所述杂芳基含有1~3个选自N、O、S的杂原子,上述基团的取代基如前所定义。
优选地,环A选自6元芳基、6元芳基-5元杂芳基,所述杂芳基含有1个选自N、O、S的杂原子,上述基团的取代基如前所定义。
优选地,环A选自苯基、苯基-噻吩基,上述基团的取代基如前所定义。
优选地,环A为苯基,其取代基如前所定义。
进一步地,环A含有的所述取代基选自卤素、C1~C6烷基、C1~C6烷氧基、-OH、-CN、-NR5R6、-SF5、-SO2R12,所述烷基、烷氧基可选地进一步被选自卤素、-NR5R6的取代基所取代,其中,R5、R6独立选自H、C1~C6烷基,R12为C1~C6烷基且可选地被卤素所取代。
优选地,环A含有的所述取代基选自卤素、C1~C6烷基、-CN、-NR5R6、-SF5、-SO2R12,所述烷基可选地进一步被选自卤素、-NR5R6的取代基所取代,其中,R5、R6独立选自H、C1~C6烷基,R12为C1~C6烷基且可选地被卤素所取代。
优选地,环A含有的所述取代基选自-F、甲基、-F取代的甲基、-NR5R6取代的甲基、-CN、-NR5R6、-SF5、-SO2R12,其中,R5、R6独立选自H、甲基,R12为-F取代的甲基。
优选地,环A含有的所述取代基选自-F、-CH3、-CHF2、-CF3、-CH2NHCH3、-CN、-NH2、-SF5、-SO2CHF2
进一步地,环A含有的所述取代基选自卤素、烷基、-CN,所述烷基可选地进一步被选自卤素、-NR5R6的取代基所取代,其中R5、R6独立选自H、烷基。
优选地,环A含有的所述取代基选自卤素、C1~C6烷基、-CN,所述烷基可选地进一步被选自卤素、-NR5R6的取代基所取代,其中R5、R6独立选自H、C1~C6烷基。
优选地,环A含有的所述取代基选自-F、甲基、-F取代的甲基、-NR5R6取代的甲基、-CN,其中R5、R6独立选自H、甲基。
优选地,环A含有的所述取代基选自-F、-CH3、-CHF2、-CF3、-CH2NHCH3、-CN、-NH2
进一步地,环A选自:
优选地,环A选自:
进一步地,Ra、Rb、Rc独立选自H、C1~C6烷基。
优选地,Ra、Rb、Rc独立选自H、甲基。
优选地,Ra为甲基,Rb为H,Rc为H。
进一步地,所述的化合物结构如式II所示:
其中各基团如前任意一项技术方案所定义。
进一步地,R1、R3、R4独立选自H、烷基、烷氧基、-OH、-CN、-NR7R8,所述烷基、烷氧基可选地被选自卤素、-NR7R8的取代基所取代,其中R7、R8独立选自H、烷基。
优选地,R1、R3、R4独立选自H、C1~C6烷基、C1~C6烷氧基、-OH、-CN、-NR7R8,所述烷基、烷氧基可选地被选自卤素、-NR7R8的取代基所取代,其中R7、R8独立选自H、C1~C6烷基。
优选地,R1、R3、R4独立选自H、C1~C6烷基、C1~C6烷氧基、-OH、-CN。
优选地,R1、R3、R4独立选自H、甲基、甲氧基、-OH、-CN。
优选地,R1选自H、甲基、甲氧基、-CN。优选地,R3为H。优选地,R4选自甲基、甲氧基、-OH。
进一步地,R1、R3、R4独立选自H、烷基、烷氧基、-OH、-NR7R8,所述烷基、烷氧基可选地被选自卤素、-NR7R8的取代基所取代,其中R7、R8独立选自H、烷基。
优选地,R1、R3、R4独立选自H、烷基。
优选地,R1、R3、R4独立选自H、C1~C6烷基。
优选地,R1、R3、R4独立选自H、甲基。
优选地,R1为H。优选地,R3为H。优选地,R4为甲基。
进一步地,R1、R3独立选自H、卤素、C1~C6烷基、C1~C6烷氧基、C1~C6烷硫基、-OH、-CN、-NR7R8,所述烷基、烷氧基可选地被选自卤素、-NR7R8的取代基所取代,其中R7、R8独立选自H、C1~C6烷基。
优选地,R1、R3独立选自H、C1~C6烷基、C1~C6烷氧基、-CN。
优选地,R1、R3独立选自H、甲基、甲氧基、-CN。
优选地,R1选自H、甲基、甲氧基、-CN。优选地,R1选自H、甲基、甲氧基。优选地,R3为H。
进一步地,R4选自H、C1~C6烷基、C2~C6烯基、3~10元环烷基、3~10元环烯基、C1~C6烷氧基、C1~C6烷硫基、-OH、-CN、-C(=O)H、-C(=O)NR19R20、-C(=O)OR21,所述烷基可选地被卤素所取代,其中R19、R20、R21独立选自H、C1~C6烷基。
优选地,R4选自C1~C6烷基、C2~C6烯基、3~5元环烷基、5元环烯基、C1~C6烷氧基、C1~C6烷硫基、-OH、-CN、-C(=O)H、-C(=O)NR19R20、-C(=O)OR21,所述烷基可选地被卤素所取代,其中R19、R20、R21独立选自H、C1~C6烷基。
优选地,R4选自C1~C6烷基、-OH。
优选地,R4选自甲基、乙基、二氟甲基、乙烯基、环丙烷基、环戊烯基、甲氧基、甲硫基、-OH、-CN、-C(=O)H、-C(=O)NH2、-C(=O)N(CH3)2、-C(=O)OH、-C(=O)OCH3
进一步地,R4为卤素。
优选地,R4为氯。
进一步地,当R13为烷基时,X为一单键,所述烷基可选地被选自-OH、烷氧基、卤素、-C(=O)NR9R10、-C(=O)OR11的取代基所取代,R9、R10、R11独立选自H、烷基。
进一步地,R2为-X-R13,X为一单键,R13为C1~C6烷基,所述C1~C6烷基可选地被选自-OH、C1~C6烷氧基、卤素、-C(=O)NR9R10、-C(=O)OR11、-NR22R23的取代基所取代,R9、R10、R11、R22、R23独立选自H、C1~C6烷基。
优选地,R2为-X-R13,X为一单键,R13为C1~C6烷基,所述C1~C6烷基可选地被选自-OH、卤素、-C(=O)NR9R10、-NR22R23的取代基所取代,R9、R10、R22、R23独立选自C1~C6烷基。
优选地,R2为-X-R13,X为一单键,R13为C1~C6烷基,所述C1~C6烷基可选地被选自-OH、-F、-C(=O)N(CH3)2、-N(CH3)2的取代基所取代。
优选地,R2选自-CH3
进一步地,R2为-X-R13,X为一单键,R13为C1~C6烷基,所述C1~C6烷基可选地被选自-OH、C1~C6烷氧基、卤素、-C(=O)NR9R10、-C(=O)OR11的取代基所取代,R9、R10、R11独立选自H、C1~C6烷基。
优选地,R2为-X-R13,X为一单键,R13为C1~C6烷基,所述C1~C6烷基可选地被选自-OH、卤素、-C(=O)NR9R10的取代基所取代,R9、R10独立选自C1~C6烷基。
优选地,R2为-X-R13,X为一单键,R13为C1~C6烷基,所述C1~C6烷基可选地被选自-OH、-F、-C(=O)N(CH3)2的取代基所取代。
优选地,R2选自-CH3
进一步地,R2为-X-R13,X选自C1~C3亚烷基、一单键,R13选自6~12元芳基、5~12元杂芳基,所述杂芳基含有选自N、O、S的杂原子,所述芳基、杂芳基可选地被选自卤素、C1~C6烷基、C1~C6烷氧基、-OH、-CN、-NR14R15的取代基所取代,R14、R15独立选自H、C1~C6烷基。
优选地,R2为-X-R13,X为亚甲基、一单键,R13为苯基、5~6元杂芳基,所述杂芳基含有1~2个选自N、O、S的杂原子,所述苯基、杂芳基可选地被选自卤素、C1~C6烷基、C1~C6烷氧基、-OH、-CN、-NR14R15的取代基所取代,R14、R15独立选自H、C1~C6烷基。
优选地,R2为-X-R13,X为一单键,R13为吡啶基、嘧啶基,所述吡啶基、嘧啶基可选地被C1~C6烷氧基所取代。
优选地,R2选自
进一步地,R2为-X-R13,X为亚甲基、一单键,R13为苯基、5~6元杂芳基,所述杂芳基含有1个选自N、O、S的杂原子,所述苯基、杂芳基可选地被选自卤素、C1~C6烷基、C1~C6烷氧基、-OH、-CN、-NR14R15的取代基所取代,R14、R15独立选自H、C1~C6烷基。
优选地,R2为-X-R13,X为一单键,R13为吡啶基,所述吡啶基可选地被C1~C6烷氧基所取代。
优选地,R2
进一步地,R2为-X-R13,X选自C1~C3亚烷基、一单键,R13选自3~10元环烷基、3~10元杂环烷基、3~10元环烯基、3~10元杂环烯基,所述杂环烷基、杂环烯基含有选自N、O、S的杂原子,所述环烷基、杂环烷基、环烯基、杂环烯基可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被选自卤素、-OH、C1~C6烷氧基、-NR17R18的取代基所取代。
优选地,R2为-X-R13,X选自C1~C3亚烷基、一单键,R13选自3~10元环烷基、3~10元杂环烷基,所述杂环烷基含有选自N、O、S的杂原子,所述环烷基、杂环烷基可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被选自卤素、-OH的取代基所取代。
优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自3~10元环烷基、4~8元杂环烷基,所述杂环烷基含有1~2个选自N、O、S的杂原子,所述环烷基、杂环烷基可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被选自卤素、-OH的取代基所取代。
优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自环丙烷基、环丁烷基、环戊烷基、二环[1.1.1]戊烷基、环己烷基、金刚烷基、氮杂环丁烷基、四氢呋喃基、四氢吡喃基、四氢噻喃基、四氢吡咯基、哌啶基、哌嗪基、1,4-亚乙基哌啶基、吗啉基,上述基团可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被选自卤素、-OH的取代基所取代。
优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自3~6元环烷基、5~8元杂环烷基,所述杂环烷基含有1~2个选自N、O、S的杂原子,所述环烷基、杂环烷基可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被选自卤素、-OH的取代基所取代。
优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自环丙烷基、环丁烷基、环戊烷基、二环[1.1.1]戊烷基、环己烷基、四氢呋喃基、四氢吡喃基、四氢噻喃基、四氢吡咯基、哌啶基、1,4-亚乙基哌啶基、吗啉基,上述基团可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被选自卤素、-OH的取代基所取代。
优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自3~6元环烷基,所述环烷基可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被卤素所取代。
优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自环丙烷基、环丁烷基、环戊烷基、环己烷基,上述基团可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被卤素所取代。
进一步地,R17、R18独立选自H、C1~C6烷基。
优选地,R17、R18独立选自H、甲基。
优选地,R17、R18均为甲基。
进一步地,R2为-X-R13,X选自亚甲基、一单键,R13选自环丙烷基、环丁烷基、环戊烷基、二环[1.1.1]戊烷基、环己烷基、金刚烷基、氮杂环丁烷基、四氢呋喃基、四氢吡喃基、四氢噻喃基、四氢吡咯基、哌啶基、哌嗪基、1,4-亚乙基哌啶基、吗啉基,上述基团可选地被选自-F、甲基、的取代基所取代,其中甲基可选地进一步被选自-F、-OH的取代基所取代。
优选地,R2选自:
优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自环丙烷基、环丁烷基、环戊烷基、二环[1.1.1]戊烷基、环己烷基、四氢呋喃基、四氢吡喃基、四氢噻喃基、四氢吡咯基、哌啶基、1,4-亚乙基哌啶基、吗啉基,上述基团可选地被选自-F、甲基、的取代基所取代,其中甲基可选地进一步被选自-F、-OH的取代基所取代。
优选地,R2选自:
进一步地,R2为-X-R13,X为一单键,R13选自环丙烷基、环丁烷基、环戊烷基、环己烷基,上述基团可选地被选自-F、甲基的取代基所取代,其中甲基可选地进一步被-F所取代。
优选地,R2选自:
进一步地,Y为C1~C3亚烷基,R16选自3~10元环烷基、3~10元杂环烷基、3~10元环烯基、3~10元杂环烯基、6~12元芳基、5~12元杂芳基,所述杂环烷基、杂环烯基、杂芳基含有选自N、O、S的杂原子。
优选地,Y为C1~C3亚烷基,R16为3~10元杂环烷基,所述杂环烷基含有1个选自N、O、S的杂原子。
优选地,Y为亚甲基,R16为5元杂环烷基,所述杂环烷基含有1个选自N、O、S的杂原子。
优选地,Y为亚甲基,R16为四氢吡咯基。
进一步地,R2
进一步地,Y选自R16选自C1~C6烷基、C1~C6烷氧基、-NR17R18、3~10元环烷基、3~10元杂环烷基、3~10元环烯基、3~10元杂环烯基、6~12元芳基、5~12元杂芳基,所述杂环烷基、杂环烯基、杂芳基含有选自N、O、S的杂原子。
优选地,Y选自R16选自C1~C6烷基、C1~C6烷氧基、-NR17R18、3~10元杂环烷基、6~12元芳基、5~12元杂芳基,所述杂环烷基、杂芳基含有1~2个选自N、O、S的杂原子。
优选地,Y选自R16选自C1~C6烷基、C1~C6烷氧基、-NR17R18、5~6元杂环烷基、6元芳基、5~6元杂芳基,所述杂环烷基、杂芳基含有1~2个选自N、O的杂原子。
优选地,Y选自R16选自甲基、甲氧基、-N(CH3)2、吗啉基、苯基、异噁唑基。
进一步地,R2选自
优选地,R2选自
进一步地,R2选自环烷基、杂环烷基、环烯基、杂环烯基,所述环烷基、杂环烷基、环烯基、杂环烯基可选地被选自卤素、烷基、卤素取代的烷基、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;或者,R2选自烷基,所述烷基可选地被选自卤素、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;其中,R9、R10、R11独立选自H、烷基。
优选地,R2选自3~10元环烷基、3~10元杂环烷基、3~10元环烯基、3~10元杂环烯基,所述杂环烷基、杂环烯基含有选自N、O、S的杂原子,所述环烷基、杂环烷基、环烯基、杂环烯基可选地被选自卤素、烷基、卤素取代的烷基、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;或者,R2选自C1~C6烷基,所述C1~C6烷基可选地被选自卤素、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;其中,R9、R10、R11独立选自H、烷基。
优选地,R2选自3~6元环烷基、3~6元杂环烷基、5~6元环烯基、5~6元杂环烯基,所述杂环烷基、杂环烯基含有选自N、O、S的杂原子,所述环烷基、杂环烷基、环烯基、杂环烯基可选地被选自卤素、烷基、卤素取代的烷基、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;或者,R2选自C1~C6烷基,所述C1~C6烷基可选地被选自卤素、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;其中,R9、R10、R11独立选自H、烷基。
优选地,R2选自3~6元环烷基、3~6元杂环烷基,所述杂环烷基含有1~2个选自N、O、S的杂原子,所述环烷基、杂环烷基可选地被选自卤素、C1~C6烷基、卤素取代的C1~C6烷基、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;或者,R2选自C1~C6烷基,所述C1~C6烷基可选地被选自卤素、-C(=O)NR9R10的取代基所取代;其中,R9、R10、R11独立选自C1~C6烷基。
优选地,R2选自3~6元环烷基、5~6元杂环烷基,所述杂环烷基含有1个选自O、S的杂原子,所述环烷基、杂环烷基可选地被选自卤素、C1~C3烷基、卤素取代的C1~C3烷基、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;或者,R2选自C1~C3烷基,所述C1~C3烷基可选地被选自卤素、-C(=O)NR9R10的取代基所取代;其中,R9、R10、R11独立选自C1~C3烷基。
优选地,R2选自环丙烷基、环丁烷基、环戊烷基、二环[1.1.1]戊烷基、环己烷基、四氢呋喃基、四氢吡喃基、四氢噻喃基,上述基团可选地被选自卤素、C1~C3烷基、卤素取代的C1~C3烷基、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;或者,R2选自甲基、乙基、丙基,上述基团可选地被选自卤素、-C(=O)NR9R10的取代基所取代;其中,R9、R10、R11独立选自C1~C3烷基。
优选地,R2选自环丙烷基、环丁烷基、环戊烷基、二环[1.1.1]戊烷基、环己烷基、四氢呋喃基、四氢吡喃基、四氢噻喃基,上述基团可选地被选自氟、甲基、三氟甲基、-C(=O)N(CH3)2、-C(=O)OCH3的取代基所取代;或者,R2选自甲基、乙基、丙基,上述基团可选地被选自氟、-C(=O)N(CH3)2的取代基所取代。
优选地,R2选自:
优选地,为如下构型:
进一步地,所述的化合物选自如下之一:



本发明提供了药物组合物,其含有所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,以及药学上可接受的辅料或者辅助性成分。
本发明提供了所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,或所述的药物组合物在制备SOS1抑制剂中的用途。
本发明提供了所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,或所述的药物组合物在制备用于治疗由SOS1介导的疾病的药物中的用途。
进一步地,所述疾病选自:癌症、病原性皮疹病。
进一步地,所述癌症选自:非小细胞肺癌、肺癌、胰腺癌、卵巢癌、膀胱癌、前列腺癌、慢性粒细胞白血病、结直肠癌、脑癌、肝癌、肾癌、胃癌、及乳腺癌;
所述病原性皮疹病选自:努南综合症、心面皮肤综合症、I型遗传性牙龈纤维瘤病。
本发明提供了所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,或所述的药物组合物在制备治疗致使SOS1蛋白过度表达的疾病的药物中的用途。
本发明提供了所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,或所述的药物组合物在制备治疗SOS1蛋白过度表达所致疾病的药物中的用途。
本发明还提供了药物组合物,含有同时或者分别给药的所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,以及RAS抑制剂。
进一步地,所述RAS抑制剂为KRAS抑制剂。
优选地,所述RAS抑制剂为KRAS G12C抑制剂。
优选地,所述RAS抑制剂为Adagrasib。
本发明还提供了所述的药物组合物在制备治疗癌症的药物中的用途。
进一步地,所述癌症选自非小细胞肺癌、胰腺癌。
本发明提供了治疗由SOS1介导的疾病的方法,其包括向受试者施用所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,或所述的药物组合物的步骤。
本发明提供了治疗致使SOS1蛋白过度表达的疾病的方法,其包括向受试者施用所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,或所述的药物组合物的步骤。
本发明提供了治疗SOS1蛋白过度表达所致疾病的方法,其包括向受试者施用所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,或所述的药物组合物的步骤。
在一实施方式中,向受试者施用的药物量为有效量。
术语定义:
在本发明中,除非另有说明,否则本文中使用的科学和技术名词具有本领域技术人员所通常理解的含义。并且,本文中所用的相关术语和实验室操作步骤均为相应领域内广泛使用的术语和常规步骤。同时,为了更好地理解本发明,下面提供相关术语的定义和解释。
本发明所述“互变异构体”是指因分子中某原子在两个位置移动而产生的官能团异构体,尤其是分子中存在机动的氢原子,例如酮式和烯醇式互变异构体。
本发明所述“立体异构体”是指分子中原子或原子团互相连接次序相同,但空间排列不同而产生的异构体,包括顺反异构体、旋光异构体、构象异构体。本发明所述立体异构体还包括两种或多种立体异构体的混合物形式,例如对映异构体和/或非对映异构体的任何比例的混合物。
本发明所述“同位素标记物”是指分子中一个或多个原子被具有不同原子质量或质量数的原子替代。可以掺入本发明化合物的同位素的实例包括氢、碳、氮、氧、磷、硫、氟、氯和碘的同位素,分别例如但不限于2H、3H、11C、13C、14C、13N、15N、15O、17O、18O、35S、18F、36Cl、123I和125I。某些同位素标记的本发明化合物可用于药物和/或底物组织分布研究,放射性同位素氚即3H和碳-14即14C因其易于掺入和方便的检测手段而特别可用于该目的,例如,本发明化合物可以富含1%、2%、5%、10%、25%、50%、75%、90%、95%或99%的指定同位素。此外,用较重的同位素例如氘即2H取代的本发明化合物可以提供某些治疗优势。
本发明所述“药学上可接受的盐”是指本发明化合物与酸或碱所形成的适合用作药物的盐。上述酸碱为广义的路易斯酸碱。适合形成盐的酸包括无机酸、有机酸、酸性氨基酸。
本发明所述“溶剂化物”表示一个或多个溶剂分子与本发明化合物所形成的适合用作药物的缔合物。形成溶剂化 物的溶剂包括水、有机溶剂。
本发明中,“环”是指任意的共价封闭结构,包括例如碳环(例如芳基或环烷基)、杂环(例如杂芳基或杂环烷基)、芳香基(如芳基或杂芳基)、非芳香基(如环烷基或杂环烷基)。本发明所述“环”可以是单环也可以是多环(包括双环),可以是稠环、螺环或桥环。
本发明中,“元”代表构成环骨架的环原子数目。
本发明中,“可选地被···取代”是指可以被一个或多个(包括两个)指定的取代基取代,也可以为非取代。
术语“环烷基”指具有单环或多环的饱和的碳环烃基。“3~10元环烷基”指具有3~10个环碳原子的饱和的单环或多环烃环,实例包括但不限于环丙烷基、环丁烷基、环戊烷基、二环[1.1.1]戊烷基、环己烷基、金刚烷基。例如,环丙烷的结构式为环丁烷的结构式为环戊烷的结构式为二环[1.1.1]戊烷的结构式为环己烷的结构式为金刚烷的结构式为如另外指明,所述环烷基可选地被一个或多个适合的取代基所取代。
术语“杂环烷基”指具有单环或多环的饱和的环状基团,且其中至少有一个环原子为杂原子,其余环原子为碳。杂原子包括但不限于N、O、S、P、Si等,优选为N、O、S。“3~10元杂环烷基”指具有3~10个环原子的饱和的单环或多环基团,且其中至少有一个环原子为杂原子,其余环原子为碳,实例包括但不限于: 如另外指明,所述杂环烷基可选地被一个或多个适合的取代基所取代。
术语“环烯基”指含有至少一个碳-碳双键(即C=C)的非芳族的单环或多环的碳环烃基。“3~10元环烯基”指具有3~10个环碳原子的不饱和非芳族的单环或多环碳环烃基,实例包括但不限于环丙烯基、环丁烯基、环戊烯基、环戊二烯基、环己烯基、环己二烯基等。如另外指明,所述环烯基可选地被一个或多个适合的取代基所取代。
术语“杂环烯基”指含有至少一个双键的非芳族的单环或多环基团,且其中至少有一个环原子为杂原子,其余环原子为碳。杂原子包括但不限于N、O、S、P、Si等,优选为N、O、S。“3~10元杂环烯基”的实例包括但不限于:如另外指明,所述杂环烯基可选地被一个或多个适合的取代基所取代。
术语“芳基”指具有共轭π电子系统的全碳单环或多环芳族基团,包括单环芳基(例如6元环的苯基)、稠环芳基(具有芳环和芳环共有环边,例如10元环的萘基)、联芳基(通过单键连接形成芳基-芳基结构,例如12元环的联苯基)。如另外指明,所述芳基可选地被一个或多个适合的取代基所取代。
术语“杂芳基”指具有共轭π电子系统的单环或多环芳族基团,且其中至少有一个环原子为杂原子,其余环原子为碳。杂原子优选为N、O、S。本发明所述的杂芳基,包括单环杂芳基(例如5元环的噻吩基、5元环的异噁唑基、6元环的吡啶基)、稠环杂芳基(具有杂芳环和杂芳环共有环边,或者芳环和杂芳环共有环边,例如9元环的苯并噻吩基)、联杂芳基(通过单键连接形成杂芳基-杂芳基结构,或者芳基-杂芳基结构,例如11元环的苯基-噻吩基:)。如另外指明,所述杂芳基可选地被一个或多个适合的取代基所取代。
术语“卤素”包括F、Cl、Br或I。
术语“烷基”指直链或支链的饱和脂肪族烃基。“C1~C6烷基”指具有1、2、3、4、5或6个碳原子的直链或支链的饱和脂肪族烃基,实例包括但不限于甲基、乙基、正丙基、异丙基、正丁基、仲丁基、叔丁基、正戊基、正己基等。如另外指明,所述烷基可选地被一个或多个适合的取代基所取代。
术语“亚烷基”指从饱和的直链或支链脂肪族烃基中去掉两个氢原子所得到的饱和的二价烃基基团;即烷基中的一个氢被取代,烷基的定义如上所述。“C1~C3亚烷基”的实例包括但不限于亚甲基(-CH2-),亚乙基{包括-CH2CH2- 或-CH(CH3)-},亚异丙基{包括-CH(CH3)CH2-或-C(CH3)2-}等。
术语“烷氧基”指通过氧原子连接至如上文所定义的“烷基”,即“烷氧基”基团可以定义为-OR,其中R是如上定义的烷基。“C1~C6烷氧基”的实例包括但不限于甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、仲丁氧基、叔丁氧基、正戊氧基、正己氧基等。如另外指明,所述烷氧基可选地被一个或多个适合的取代基所取代。
术语“烷硫基”指通过硫原子连接至如上文所定义的“烷基”,即“烷硫基”基团可以定义为-SR,其中R是如上定义的烷基。如另外指明,所述烷硫基可选地被一个或多个适合的取代基所取代。
术语“烯基”指含有至少一个碳-碳双键(即C=C)的直链或支链的脂肪烃基团。双键可以作为E或Z异构体存在。双键可以位于烃链的任何可能的位置。“C2~C6烯基”的实例包括但不限于乙烯基、1-丙烯基、2-丙烯基、1-、2-或3-丁烯基等。如另外指明,所述烯基可选地被一个或多个适合的取代基所取代。
本发明所述药学上可接受的辅料,是药物中除主药以外的一切附加材料的总称,辅料应当具备如下性质:(1)对人体无毒害作用,几无副作用;(2)化学性质稳定,不易受温度、pH、保存时间等的影响;(3)与主药无配伍禁忌,不影响主药的疗效和质量检查;(4)不与包装材料相互发生作用。本发明中辅料包括但不仅限于填充剂(稀释剂)、润滑剂(助流剂或抗粘着剂)、分散剂、湿润剂、粘合剂、调节剂、增溶剂、抗氧剂、抑菌剂、乳化剂、崩解剂等。本发明化合物或药物组合物的施用方式没有特别限制,代表性的施用方式包括(但并不限于):口服、肠胃外(静脉内、肌肉内或皮下)、和局部给药。
术语“受试者”包括人或非人动物。示例性人受试者包括患有疾病(如本文所述的疾病)的受试者(简称患者)或正常个体。“非人动物”包括所有脊椎动物,例如非哺乳动物和哺乳动物,例如非人灵长类、家畜和/或驯化动物。
如本文中所述“有效量”指被给药后会在一定程度上缓解所治疗疾病的一种或多种症状的化合物的量。可调整给药方案以提供最佳所需响应。
本发明的有益效果是:本发明提供了一系列对SOS1蛋白具有明显的抑制作用的化合物,为以SOS1为治疗的靶点的疾病如癌症、病原性皮疹病等的治疗提供新的方案。相较于现有的SOS1抑制剂,本发明化合物在活性、成药性等方面有明显改善,尤其是具有更好的肝微粒体稳定性,成药性质佳,可用于制备治疗相关疾病的药物,具有广阔的应用前景。
具体实施方式
下面对本发明的技术方案进行清楚、完整地描述,显然,所描述的实施例是本发明一部分实施例,而不是全部的实施例。基于本发明中的实施例,本领域普通技术人员在没有做出创造性劳动前提下所获得的所有其他实施例,都属于本发明保护的范围。
如本文使用的和除非另作说明,术语“包含”,“包括”,“具有”,“含有”,包括其语法上的等同形式,通常应当理解为开放式且非限制性的,例如,不排除其他未列举的要素或步骤。
本发明的化合物结构是通过核磁共振(NMR)或/和液质联用色谱(LC-MS)来确定的。NMR的化学位移(δ)以百万分之一(ppm)的单位给出。NMR的测定是用AVANCE NEO 400MHz Bruker仪器,测定溶剂为氘代二甲基亚砜(DMSO-d6)、氘代氯仿(CDCl3)、氘代甲醇(CD3OD),内标为四甲基硅烷(TMS)。MS的测定是用ISQ-EC Thermo Fisher LC-MS仪器。Prep-HPLC是GX-281Gilson色谱仪,分离方法有:(方法1)Sun Fire Prep C18 OBDTM 5μm,30×150mm Column,0.04%HCl水溶液/乙腈;(方法2)Sun Fire Prep C18 OBDTM 5μm,30×150mm Column,0.06%甲酸水溶液/乙腈;(方法3)Xbridge Prep C18 OBDTM 5μm,30×150mm Column,10mM NH4HCO3水溶液/乙腈;(方法4)SunFire Prep C18 OBDTM 5μm,30×150mm Column,0.02%三氟乙酸水溶液/乙腈。
本发明实施例中的起始原料是已知的,并且可以在市场上买到,或者可以按照本领域的已知方法合成得到。
本发明所使用的溶剂,若无特殊说明,是指可经市售获得。
实施例中若无特殊说明,反应的温度为室温,为20℃~30℃。
本发明中涉及的化学缩写简称具有以下意义:
DMF:N,N-二甲基甲酰胺
DMF-DMA:N,N-二甲基甲酰胺二甲基缩醛
DMSO:二甲基亚砜
DIPEA:N,N-二异丙基乙胺
HATU:2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯
NBS:N-溴代丁二酰亚胺
THF:四氢呋喃
Prep-HPLC:制备型高效液相色谱仪
实施例1
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
步骤a):2-甲酰基-3-氧代戊二酸二甲酯的制备
将3-氧代戊二酸二甲酯(10.0g,57.421mmol)和1-甲基四氢呋喃(100mL)加入反应瓶中,冰水浴下滴加DMF-DMA(6.8g,57.42mmol),室温搅拌反应2h。向反应液中加入4N的稀盐酸(30mL),维持室温下继续反应1h。反应结束后,反应液用甲基叔丁基醚萃取(100mL),合并有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得2-甲酰基-3-氧代戊二酸二甲酯,无需进一步纯化直接用于下一步反应,产率98.6%;ESI-MS(m/z):203.2[M+H]+
步骤b):4-羟基-1-(1-甲基环丙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备
将1-甲基环丙烷-1-胺盐酸盐(4.7g,43.925mmol)和2-甲酰基-3-氧代戊二酸二甲酯(8.9g,43.925mmol)用甲醇(120mL)溶解,冰水浴下加入甲醇钠(5.2g,96.635mmol),升温至80℃反应6h。反应结束后,将反应液冷却至室温,减压浓缩除去大部分溶剂,加饱和氯化铵水溶液(100mL)淬灭反应,用4N的盐酸将反应液调至pH=2~3,析出大量固体,过滤,滤饼用1N稀盐酸洗涤,收集滤饼,减压干燥,滤饼用甲基叔丁基醚和石油醚混合溶剂(80mL,v/v=4/1)打浆,过滤,滤饼减压干燥,得4-羟基-1-(1-甲基环丙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯,产率83.8%;ESI-MS(m/z):224.2[M+H]+
步骤c):1-(1-甲基环丙基)-6-氧-4-(三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-甲酸甲酯的制备
将4-羟基-1-(1-甲基环丙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(6.4g,28.662mmol)、吡啶(4.5g,57.324mmol)和1,2-二氯乙烷(60mL)加入反应瓶中,冰水浴下加入三氟甲磺酸酐(12.1g,42.993mmol),冰水浴下搅拌反应2min。加饱和氯化铵水溶液(50mL)淬灭反应,静置分液,水相用二氯甲烷萃取(100mL),合并有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=10/1到3/1),得1-(1-甲基环丙基)-6-氧-4-(三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-甲酸甲酯,产率91.2%;ESI-MS(m/z):356.2[M+H]+
步骤d):4-乙酰基-1-(1-甲基环丙基)-6-氧基-1,6-二氢吡啶-3-甲酸甲酯的制备
将1-(1-甲基环丙基)-6-氧-4-(三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-甲酸甲酯(9.0g,25.352mmol)、1,4-二氧六环(60mL)、三丁基(1-乙氧基乙烯基)锡(11.9g,32.958mmol)、三乙胺(3.3g,32.958mmol)和二(三苯基膦)二氯化钯(356mg,0.507mmol)依次加入反应瓶中,氮气置换三次,升温至100℃搅拌反应过夜。反应结束后,将反应液冷却至室温,减压蒸除溶剂,加入4N的稀盐酸调至pH=2-3,继续室温下搅拌1h,加入饱和氟化钾水溶液(80mL)淬灭反应,加入乙酸乙酯(100mL),继续搅拌30min,过滤,滤饼用乙酸乙酯(50mL)洗涤,滤液分液,有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=1/1),得4-乙酰基-1-(1-甲基环丙基)-6-氧基-1,6-二氢吡啶-3-甲酸甲酯,产率83.3%;ESI-MS(m/z):250.2[M+H]+
步骤e):4-羟基-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-乙酰基-1-(1-甲基环丙基)-6-氧基-1,6-二氢吡啶-3-甲酸甲酯(5.0g,20.059mmol)和无水乙醇(50mL)加 入反应瓶中,室温下加入水合肼(5mL,80%),80℃搅拌反应1h。将反应液冷却至室温,减压浓缩,向残余物加入乙酸乙酯(30mL),室温打浆1h,过滤,滤饼用乙酸乙酯淋洗(30mL),收集滤饼,减压干燥,得4-羟基-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率95.6%;ESI-MS(m/z):232.1[M+H]+
步骤f):4-溴-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-羟基-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(950mg,4.108mmol)、三溴氧磷(2.4g,8.720mmol)和乙腈(100mL)加入反应瓶中,升温至回流反应2h。将反应液冷却至室温,倾入饱和碳酸氢钠水溶液(200mL)中淬灭,用二氯甲烷萃取(200mL×2),合并有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得4-溴-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率82.7%;ESI-MS(m/z):294.0[M+H]+
步骤g):(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-溴-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(450mg,1.530mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙-1-胺盐酸盐(690mg,3.058mmol)、DIPEA(1.2g,9.285mmol)、氟化铯(0.9g,5.925mmol)和DMSO(10mL)加入反应瓶中,升温至80℃反应3h。将反应液冷却至室温,加入饱和氯化铵水溶液(40mL)淬灭反应,用二氯甲烷萃取(50mL×2),合并有机相,用饱和食盐水(50mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品经Prep-HPLC(方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率8.1%;1H NMR(400MHz,DMSO-d6)δ9.26(s,1H),7.74(d,J=6.8Hz,1H),7.61(t,J=7.2Hz,1H),7.48(t,J=7.2Hz,1H),7.39-7.09(m,2H),6.48(s,1H),5.66-5.59(m,1H),2.32(s,3H),1.58-1.53(m,6H),1.20-1.14(m,2H),1.10-1.04(m,2H);ESI-MS(m/z):403.0[M+H]+
实施例2
(R)-1-甲基-4-((1-(2-甲基-3-(三氟甲基)苯基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例1,所得粗品经硅胶柱层析纯化(洗脱剂:甲醇/二氯甲烷=5/95),再经Prep-HPLC(方法3)纯化,得(R)-1-甲基-4-((1-(2-甲基-3-(三氟甲基)苯基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率14.3%;1H NMR(400MHz,DMSO-d6)δ9.24(s,1H),7.76(d,J=6.8Hz,1H),7.73(d,J=7.6Hz,1H),7.54(d,J=7.6Hz,1H),7.35(t,J=7.6Hz,1H),6.46(s,1H),5.64-5.54(m,1H),2.53(s,3H),2.32(s,3H),1.54(s,3H),1.51(d,J=7.2Hz,3H),1.20-1.04(m,4H);ESI-MS(m/z):417.0[M+H]+
实施例3
(R)-2-甲基-3-(1-((1-甲基-6-(1-甲基环丙基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-4-基)氨基)乙基)苯甲腈的制备
制备方法参考实施例1,经Prep-HPLC(方法3)纯化,得(R)-2-甲基-3-(1-((1-甲基-6-(1-甲基环丙基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-4-基)氨基)乙基)苯甲腈,产率8.9%;1H NMR(400MHz,DMSO-d6)δ9.22(s,1H),7.79-7.70(m,2H),7.62(d,J=6.8Hz,1H),7.34(t,J=7.6Hz,1H),6.46(s,1H),5.54-5.45(m,1H),2.63(s,3H),2.32(s,3H),1.53(s,3H),1.50(d,J=7.2Hz,3H),1.19-1.04(m,4H);ESI-MS(m/z):374.0[M+H]+
实施例4
(R)-4-((1-(3-(二氟甲基)-2-甲基苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H))-酮的制备
制备方法参考实施例1,经Prep-HPLC(方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-甲基苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H))-酮,产率16.0%;1H NMR(400MHz,DMSO-d6)δ9.24(s,1H),7.71(d,J=6.8Hz,1H),7.60(d,J=7.6Hz,1H),7.38(d,J=7.6Hz,1H),7.28(t,J=7.6Hz,1H),7.21(t,J=56.0Hz,1H),6.46(s,1H),5.64-5.58(m,1H),2.46(s,3H),2.32(s,3H),1.53(s,3H),1.49(d,J=6.8Hz,3H),1.16-1.12(m,2H),1.08-1.04(m,2H);ESI-MS(m/z):399.0[M+H]+
实施例5
(R)-1-甲基4-((1-(5-(2-((甲基氨基)甲基)苯基)噻吩-2-基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
步骤a):(R)-4-((1-(5-溴噻吩-2-基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-溴-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(200mg,0.680mmol)、(R)-1-(5-溴噻吩-2-基)乙烷-1-胺盐酸盐(330mg,1.360mmol)、DIPEA(530mg,4.101mmol)、氟化铯(620g,4.082mmol)和DMSO(2mL)加入反应瓶中,升温至80℃反应3h。反应结束后,将反应液冷却至室温,加入饱和氯化铵水溶液(40mL)淬灭反应,用二氯甲烷萃取(50mL×2),合并有机相,用饱和食盐水(50mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品经硅胶柱层析纯化(洗脱剂:甲醇/二氯甲烷=4/96),得(R)-4-((1-(5-溴噻吩-2-基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率42.1%;ESI-MS(m/z):419.0[M+H]+
步骤b):(R)-1-甲基-4-((1-(5-(2-((甲基氨基)甲基)苯基)噻吩-2-基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-4-((1-(5-溴噻吩-2-基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(105mg,0.250mmol)、N-甲基-1-(2-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)苯基)甲胺(200mg,0.372mmol)、Pd(PPh3)4(120mg,0.104mmol)、碳酸铯(490mg,1.504mmol)、1,4-二氧六环(2mL)和水(0.5mL)加入到反应瓶中,100℃反应3h。将反应液冷却至室温,减压浓缩,所得粗品经硅胶柱层析纯化(洗脱剂:甲醇/二氯甲烷=15/85),再经Prep-HPLC(方法3)纯化,得(R)-1-甲基-4-((1-(5-(2-((甲基氨基)甲基)苯基)噻吩-2-基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率3.0%;1H NMR(400MHz,DMSO-d6)δ9.15(s,1H),7.77(d,J=8.0Hz,1H),7.49(d,J=7.2Hz,1H),7.37-7.22(m,4H),7.16-7.13(m,1H),7.09-7.06(m,1H),6.50(s,1H),5.87-5.77(m,1H),3.66(s,2H),2.39(s,3H),2.26(s,3H),1.68(d,J=6.8Hz,3H),1.50(s,3H),1.16-0.98(m,4H);ESI-MS(m/z):460.0[M+H]+
实施例6
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(四氢-2H-吡喃-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
步骤a):2-甲酰基-3-氧代戊二酸二甲酯的制备
将3-氧代戊二酸二甲酯(5.0g,28.711mmol)和1-甲基四氢呋喃(100mL)加入反应瓶中,冰水浴下滴加DMF-DMA(4.1g,34.454mmol),室温搅拌反应2h。向反应液中加入盐酸(100mL,4N),维持室温继续反应1h。反应结束后,反应液用乙酸乙酯萃取(100mL×3),合并有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得2-甲酰基-3-氧代戊二酸二甲酯,产率99.9%;ESI-MS(m/z):203.2[M+H]+
步骤b):4-羟基-6-氧代-1-(四氢-2H-吡喃-4-基)-1,6-二氢吡啶-3-甲酸甲酯的制备
将四氢-2H-吡喃-4-胺(2.0g,19.773mmol)和2-甲酰基-3-氧代戊二酸二甲酯(4.8g,23.727mmol)用甲醇(120mL)溶解,冰水浴下加入甲醇钠(2.1g,39.545mmol),80℃反应6h。将反应液冷却至室温,减压浓缩除去大部分溶剂,加饱和氯化铵水溶液(100mL)淬灭反应,用4N的盐酸将反应液调至pH=1~2,用乙酸乙酯萃取(100mL×3),合并有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=1/2),得4-羟基-6-氧代-1-(四氢-2H-吡喃-4-基)-1,6-二氢吡啶-3-甲酸甲酯,产率79.9%;ESI-MS(m/z):254.2[M+H]+
步骤c):6-氧代-1-(四氢-2H-吡喃-4-基)-4-(((三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-甲酸甲酯的制备
将4-羟基-6-氧代-1-(四氢-2H-吡喃-4-基)-1,6-二氢吡啶-3-甲酸甲酯(3.0g,11.846mmol)、吡啶(1.9g,23.692mmol)和1,2-二氯乙烷(60mL)加入反应瓶中,冰水浴下滴加三氟甲磺酸酐(6.7g,23.692mmol),维持冰水浴下搅拌反应2min。加水(100mL)淬灭反应,静置分取有机相,水相用二氯甲烷萃取(100mL×2),合并有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=3/1),得6-氧代-1-(四氢-2H-吡喃-4-基)-4-(((三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-甲酸甲酯,产率89.1%;ESI-MS(m/z):386.2[M+H]+
步骤d):4-乙酰基-6-氧代-1-(四氢-2H-吡喃-4-基)-1,6-二氢吡啶-3-甲酸甲酯的制备
将6-氧代-1-(四氢-2H-吡喃-4-基)-4-(((三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-甲酸甲酯(4.0g,10.381mmol)、1,4-二氧六环(80mL)、三丁基(1-乙氧基乙烯基)锡(4.9g,13.496mmol)、三乙胺(3.1g,31.143mmol)和二(三苯基膦)二氯化钯(364mg,0.519mmol)依次加入反应瓶中,置换氮气三次,维持100℃搅拌反应3h。将反应液冷却至室温,加入饱和氟化钾水溶液(30mL)淬灭反应,室温搅拌5min后过滤,滤饼用乙酸乙酯淋洗(30mL×2),滤液分液,水相用乙酸乙酯萃取(50mL×2),合并有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物加乙酸乙酯溶解(40mL),加入稀盐酸(40mL,1N)室温搅拌30min,再加饱和碳酸氢钠水溶液,将反应液调至pH值为中性,分取有机相,水相用乙酸乙酯萃取(50mL×2),合并有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=1/1),得4-乙酰基-6-氧代-1-(四氢-2H-吡喃-4-基)-1,6-二氢吡啶-3-甲酸甲酯,产率71.4%;ESI-MS(m/z):280.2[M+H]+
步骤e):4-羟基-1-甲基-6-(四氢-2H-吡喃-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-乙酰基-6-氧代-1-(四氢-2H-吡喃-4-基)-1,6-二氢吡啶-3-甲酸甲酯(2.0g,7.168mmol)和无水乙醇(40mL)加入反应瓶中,室温下加入水合肼(2mL,80%),80℃搅拌反应1h。将反应液降至室温,过滤,滤饼用乙醇淋洗(5mL×2),真空干燥得4-羟基-1-甲基-6-(四氢-2H-吡喃-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率88.3%;ESI-MS(m/z):262.2[M+H]+
步骤f):4-溴-1-甲基-6-(四氢-2H-吡喃-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将乙腈(10mL)和4-羟基-1-甲基-6-(四氢-2H-吡喃-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮(200mg,0.765mmol)加 入反应瓶中,室温下加入三溴氧磷(2.2g,7.655mmol),置换氮气三次,升温至80℃搅拌反应1小时。反应结束后,将反应液冷却至0℃,加入饱和碳酸氢钠水溶液将反应液调至pH值为中性,用二氯甲烷萃取(20mL×3),合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=15/1),得4-溴-1-甲基-6-(四氢-2H-吡喃-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率28.2%;ESI-MS(m/z):324.2[M+H]+
步骤g):(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(四氢-2H-吡喃-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-溴-1-甲基-6-(四氢-2H-吡喃-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮(70mg,0.216mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙-1-胺盐酸盐(97mg,0.432mmol)、氟化铯(197mg,1.296mmol)、DIPEA(167mg,1.296mmol)和DMF(4mL)加入反应瓶中,氮气置换三次,升温至80℃搅拌反应1h。反应结束后,将反应液冷却至室温,加水(10mL)淬灭反应,用二氯甲烷萃取(10mL×3),合并有机相,用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品经Prep-HPLC(方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(四氢-2H-吡喃-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率26.9%;1H NMR(400MHz,DMSO-d6)δ9.16(s,1H),7.79(d,J=6.8Hz,1H),7.60(t,J=7.2Hz,1H),7.48(t,J=7.6Hz,1H),7.27(t,J=7.6Hz,1H),7.24(t,J=54.0Hz,1H),6.57(s,1H),5.67-5.60(m,1H),5.27-5.18(m,1H),4.11-4.07(m,2H),3.56(t,J=11.6Hz,2H),2.34(s,3H),2.19-2.09(m,2H),1.85-1.82(m,2H),1.57(d,J=6.8Hz,3H);ESI-MS(m/z):433.0[M+H]+
实施例7
(R)-1-甲基-4-((1-(2-甲基-3-(三氟甲基)苯基)乙基)氨基)-6-(四氢-2H-吡喃-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例6的步骤g,经Prep-HPLC(方法3)纯化,得(R)-1-甲基-4-((1-(2-甲基-3-(三氟甲基)苯基)乙基)氨基)-6-(四氢-2H-吡喃-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率19.3%;1H NMR(400MHz,DMSO-d6)δ9.14(s,1H),7.83(d,J=6.8Hz,1H),7.71(t,J=7.2Hz,1H),7.54(t,J=8.0Hz,1H),7.36(t,J=7.6Hz,1H),6.56(s,1H),5.65-5.58(m,1H),5.26-5.18(m,1H),4.10-4.06(m,2H),3.55(t,J=11.6Hz,2H),2.54(s,3H),2.34(s,3H),2.20-2.08(m,2H),1.82(d,J=12.0Hz,2H),1.52(d,J=7.2Hz,3H);ESI-MS(m/z):447.0[M+H]+
实施例8
(R)-6-(1,1-二氧代四氢-2H-噻喃-4-基)-1-甲基-4-((1-(2-甲基-3-(三氟甲基)苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例1,所得粗品经硅胶柱层析纯化(洗脱剂:甲醇/二氯甲烷=6/94),再经Prep-HPLC(方法3)纯化,得(R)-6-(1,1-二氧代四氢-2H-噻喃-4-基)-1-甲基-4-((1-(2-甲基-3-(三氟甲基)苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率6.7%;1H NMR(400MHz,CD3OD-d6)δ9.13(s,1H),7.70(d,J=7.6Hz,1H),7.55-7.48(m,2H),7.32-7.25(m,1H),6.77(s,1H),5.75-5.65(m,1H),5.45-5.36(m,1H),3.57-3.48(m,2H),3.26-3.18(m,2H),2.77-2.67(m,2H),2.58(s,3H),2.45(s,3H),2.37-2.30(m,2H),1.61(d,J=6.8Hz,3H);ESI-MS(m/z):495.0[M+H]+
实施例9
(R)-3-(1-((6-(1,1-二;氧代四氢-2H-噻喃-4-基)-1-甲基-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-4-基)氨基)乙基)-2-甲基苄腈的制备
制备方法参考实施例8,所得粗品经硅胶柱层析纯化(洗脱剂:甲醇/二氯甲烷=6/94),再经Prep-HPLC(方法3)纯化,得(R)-3-(1-((6-(1,1-二氧代四氢-2H-噻喃-4-基)-1-甲基-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-4-基)氨基)乙基)-2-甲基苄腈,产率5.3%;1H NMR(400MHz,DMSO-d6)δ9.01(s,1H),7.96(d,J=6.4Hz,1H),7.72(d,J=7.2Hz,1H),7.63(d,J=7.6Hz,1H),7.35(t,J=7.6Hz,1H),6.59(s,1H),5.56-5.47(m,1H),5.35-5.26(m,1H),3.63-3.53(m,2H),3.28-3.22(m,2H),2.68-2.52(m,5H),2.34(s,3H),2.25-2.18(m,2H),1.53(d,J=7.2Hz,3H);ESI-MS(m/z):452.0[M+H]+
实施例10
(R)-4-((1-(3-(二氟甲基)-2-甲基苯基)乙基)氨基)-6-(1,1-二氧代四氢-2H-噻喃-4-基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例1,所得粗品经硅胶柱层析纯化(洗脱剂:甲醇/二氯甲烷=6/94),再经Prep-HPLC(方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-甲基苯基)乙基)氨基)-6-(1,1-二氧代四氢-2H-噻喃-4-基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率5.0%;1H NMR(400MHz,DMSO-d6)δ9.02(s,1H),7.92(d,J=6.8Hz,1H),7.59(d,J=7.6Hz,1H),7.39(d,J=7.6Hz,1H),7.35-7.07(m,2H),6.59(s,1H),5.68-5.60(m,1H),5.35-5.26(m,1H),3.63-3.52(m,2H),3.26-3.23(m,2H),2.68-2.54(m,2H),2.47(s,3H),2.34(s,3H),2.25-2.18(m,2H),1.52(d,J=7.2Hz,3H);ESI-MS(m/z):477.0[M+H]+
实施例11
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1,1-二氧代四氢-2H-噻喃-4-基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例1,所得粗品经硅胶柱层析纯化(洗脱剂:甲醇/二氯甲烷=4/96),再经Prep-HPLC(方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1,1-二氧代四氢-2H-噻喃-4-基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率7.3%;1H NMR(400MHz,DMSO-d6)δ9.05(s,1H),7.95(d,J=6.4Hz,1H),7.60(t,J=7.2Hz,1H),7.49(t,J=6.8Hz,1H),7.38-7.10(m,2H),6.62(s,1H),5.67-5.60(m,1H),5.38-5.27(m,1H),3.65-3.53(m,2H),3.30-3.24(m,2H),2.65-2.53(m,2H),2.35(s,3H),2.27-2.18(m,2H),1.59(d,J=7.2Hz,3H);ESI-MS(m/z):481.0[M+H]+
实施例12
(R)-6-环丁基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
步骤a):1-环丁基-4-羟基-6-氧代-1,6-二氢吡啶-3-羧酸甲酯的制备
将2-甲酰基-3-氧代戊二酸二甲酯(5.6g,27.716mmol)、环丁胺(2.4g,33.259mmol)、甲醇钠(3.5mg,60.975mmol)和甲醇(50mL)加入反应瓶中,升温至80℃搅拌反应2h。反应结束后,冷却至室温,加入饱和氯化铵水溶液(50mL)淬灭反应,以4N的盐酸将反应液调至pH=3-4,用二氯甲烷萃取(20mL×3),合并有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得1-环丁基-4-羟基-6-氧代-1,6-二氢吡啶-3-羧酸甲酯,无需纯化直接用于下一步,产率82.2%;ESI-MS(m/z):224.1[M+H]+
步骤b):1-环丁基-6-氧代-4-(((三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-羧酸甲酯的制备
将1-环丁基-4-羟基-6-氧代-1,6-二氢吡啶-3-羧酸甲酯(5.1g,22.870mmol)溶解在二氯甲烷(50mL)中,冰浴下加入吡啶(3.6g,45.740mmol),随后滴加三氟甲磺酸酐(9.7g,34.305mmol),滴毕,升温至室温搅拌20min。加水(50mL)淬灭反应,以二氯甲烷萃取(20mL×3),合并有机相,饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=3/1),得1-环丁基-6-氧代-4-(((三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-羧酸甲酯,产率60.5%;ESI-MS(m/z):356.1[M+H]+
步骤c):4-乙酰基-1-环丁基-6-氧代-1,6-二氢吡啶-3-羧酸甲酯的制备
将1-环丁基-6-氧代-4-(((三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-羧酸甲酯(4.9g,13.803mmol)、三丁基(1-乙氧基乙烯基)锡烷(6.0g,16.564mmol)、Pd(PPh3)Cl2(486mg,0.690mmol)、三乙胺(4.2g,42.409mmol)和1,4二氧六环(50mL)加入反应瓶中,氮气置换三次,升温至100℃搅拌反应5h。反应结束后,将反应液冷却至室温,加30%氟化钾水溶液(100mL)淬灭反应,过滤,滤液用乙酸乙酯萃取(20mL×3),合并有机相,用饱和食盐水(20mL×3)洗涤,有机相减压浓缩,加入氯化氢的乙酸乙酯(4M)溶液调至pH=2-3,室温搅拌30min,减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=20/1),得4-乙酰基-1-环丁基-6-氧代-1,6-二氢吡啶-3-羧酸甲酯,产率43.6%;ESI-MS(m/z):250.1[M+H]+
步骤d):6-环丁基-4-羟基-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-乙酰基-1-环丁基-6-氧代-1,6-二氢吡啶-3-羧酸甲酯(1.5g,6.024mmol),水合肼(1.1g,18.072mmol,80%)和乙醇(20mL)加入反应瓶中,升温至回流搅拌反应1h。反应结束后,反应液冷却至室温,过滤,滤饼用少量乙醇淋洗,真空干燥,得6-环丁基-4-羟基-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率93.3%;ESI-MS(m/z):232.1[M+H]+
步骤e):4-溴-6-环丁基-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将6-环丁基-4-羟基-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮(500g,2.165mmol)、三溴氧磷(1.2g,4.330mmol)和乙腈(50mL)依次加入反应瓶中,氮气保护下升温至105℃搅拌反应2h。将反应液冷却至室温,缓慢滴加饱和碳酸氢钠水溶液(50mL)淬灭,以二氯甲烷萃取(50mL×3),合并有机相,用饱和食盐水洗涤(50mL),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:乙酸乙酯/石油醚=4/1),得4-溴-6-环丁基-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率51.5%;ESI-MS(m/z):294.1[M+H]+
步骤f):(R)-6-环丁基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-溴-6-环丁基-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮(300mg,1.020mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙-1-胺盐酸盐(460mg,2.040mmol)、DIPEA(658mg,5.100mmol)和1,4-二氧六环(3mL)加入反应瓶中,100℃微波反应2h。反应结束后,冷却至室温,加入饱和氯化铵水溶液(20mL)淬灭反应,用乙酸乙酯萃取(20mL×3),合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(方法3)纯化,得(R)-6-环丁基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率23.5%; 1H NMR(400MHz,DMSO-d6)δ9.11(s,1H),7.81(d,J=6.8Hz,1H),7.62(t,J=7.6Hz,1H),7.48(t,J=7.2Hz,1H),7.27(t,J=7.2Hz,1H),7.27(t,J=52.0Hz,1H),6.50(s,1H),5.69-5.62(m,1H),5.22-5.13(m,1H),2.45(t,J=7.6Hz,4H),2.34(s,3H),1.93-1.84(m,2H),1.58(d,J=7.2Hz,3H);ESI-MS(m/z):403.0[M+H]+
实施例13
((R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基环丁基)吡啶并[3,4-d]哒嗪-7(6H))-酮的制备
制备方法参考实施例12,经Prep-HPLC(方法3)纯化,得((R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基环丁基)吡啶并[3,4-d]哒嗪-7(6H))-酮;1H NMR(400MHz,DMSO-d6)δ8.84(s,1H),7.72(d,J=6.8Hz,1H),7.60(t,J=7.6Hz,1H),7.48(t,J=7.2Hz,1H),7.27(d,J=7.6Hz,1H)7.24(t,J=54.4Hz,1H),6.44(s,1H),5.62(p,J=6.8Hz,1H),2.62-2.54(m,2H),2.43(t,J=9.6Hz,2H),2.33(s,3H),1.99-1.90(m,1H),1.82-1.75(m 1H),1.68(s,3H),1.56(d,J=7.2Hz,3H);产率6.7%;ESI-MS(m/z):417.2[M+H]+
实施例14
4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(四氢呋喃-3-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例12,经Prep-HPLC(方法3)纯化,得4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(四氢呋喃-3-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率24.7%;1H NMR(400MHz,DMSO-d6)δ8.94(d,J=7.6Hz,1H),7.81(t,J=7.2Hz,1H),7.60(t,J=7.6Hz,1H),7.48(t,J=7.2Hz,1H),7.26(t,J=7.6Hz,1H),7.26(t,J=56.0Hz,1H),6.57(s,1H),5.67-5.58(m,1H),5.56-5.48(m,1H),4.22-4.12(m,1H),4.03(d,J=5.6Hz,1H),4.01-3.87(m,1H),3.87-3.80(m,1H),2.35(s,3H),2.34-2.14(m,2H),1.57(d,J=7.0Hz,3H);ESI-MS(m/z):419.0[M+H]+
实施例15
(R)-4-((1-(3-氨基-5-(三氟甲基)苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
步骤a):(R)-1-甲基-6-(1-甲基环丙基)-4-((1-(3-硝基-5-(三氟甲基)苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-溴-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(200mg,0.680mmol)、(R)-1-(3-硝基-5-(三氟甲 基)苯基)乙基-1-胺盐酸盐(280mg,1.035mmol)、DIPEA(440mg,3.404mmol)和1,4-二氧六环(3mL)加入反应瓶中,100℃微波反应4h。反应结束后,冷却至室温,加入饱和氯化铵水溶液(20mL)淬灭反应,用乙酸乙酯萃取(20mL×3),合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品经硅胶柱层析纯化(洗脱剂:乙酸乙酯/石油醚=2/1),得(R)-1-甲基-6-(1-甲基环丙基)-4-((1-(3-硝基-5-(三氟甲基)苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率54.6%;ESI-MS(m/z):448.1[M+H]+
步骤b):(R)-4-((1-(3-氨基-5-(三氟甲基)苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-1-甲基-6-(1-甲基环丙基)-4-((1-(3-硝基-5-(三氟甲基)苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮(166mg,0.371mmol)、铁粉(100mg,1.790mmol)、甲醇(4mL)和饱和氯化铵水溶液(2mL)加入反应瓶中,90℃反应30min。反应结束后,将反应液冷却至室温,加入水(20mL)和二氯甲烷(20mL)剧烈搅拌5min,过滤,滤液分取有机相,水相用二氯甲烷萃取(30mL×2),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(方法3)纯化,得(R)-4-((1-(3-氨基-5-(三氟甲基)苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率29.1%;1H NMR(400MHz,DMSO-d6)δ9.21(s,1H),7.61(d,J=6.8Hz,1H),6.82(s,2H),6.68(s,1H),6.48(s,1H),5.49(s,2H),5.38-5.30(m,1H),2.34(s,3H),1.55-1.46(m,6H),1.17-1.11(m,2H),1.10-1.03(m,2H);ESI-MS(m/z):418.0[M+H]+
实施例16
(R)-6-环戊基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例12,经Prep-HPLC(方法3)纯化得(R)-6-环戊基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率25.3%;1H NMR(400MHz,DMSO-d6)δ9.08(s,1H),7.72(d,J=6.8Hz,1H),7.60(t,J=7.2Hz,1H),7.48(t,J=6.8Hz,1H),7.37-7.17(m,2H),6.54(s,1H),5.67-5.59(m,1H),5.27-5.17(m,1H),2.34(s,3H),2.20-2.07(m,2H),2.01-1.89(m,4H),1.77-1.67(m,2H),1.57(d,J=7.2Hz,3H);ESI-MS(m/z):417.0[M+H]+
实施例17
(1R,4,)-4-(4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)环已烷-1-甲酸甲酯三氟乙酸盐的制备
制备方法参考实施例12,经Prep-HPLC(方法4)纯化,得(1R,4r)-4-(4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)环己烷-1-甲酸甲酯三氟乙酸盐,产率17.6%;1H NMR(400MHz,DMSO-d6)δ10.06(s,1H),9.75(s,1H),7.73(t,J=7.6Hz,1H),7.61(t,J=7.2Hz,1H),7.40-7.31(m,1H),7.25(t,J=56.0Hz,1H),6.92(s,1H),5.55-5.48(m,1H),4.90-4.82(m,1H),3.63(s,3H),2.49-2.46(m,2H),2.45(s,3H),2.18-1.88(m,6H),1.73(d,J=6.8Hz,3H),1.65-1.54(m,2H);ESI-MS(m/z):489.0[M+H]+
实施例18
(R)-6-(双环[1.1.11戊-1-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例12,经Prep-HPLC(方法3)纯化,得(R)-6-(双环[1.1.1]戊-1-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率19.7%;1H NMR(400MHz,DMSO-d6)δ8.83(s,1H),7.80(d,J=6.8Hz,1H),7.60(t,J=7.6Hz,1H),7.47(d,J=7.2Hz,1H),7.27(t,J=7.2Hz,1H),7.24(t,J=56.0Hz,1H),6.47(s,1H),5.68-5.61(m,1H),2.75(s,1H),2.46(s,6H),2.33(s,3H),1.57(d,J=7.2Hz,3H);ESI-MS(m/z):415.0[M+H]+
实施例19
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-异丙基-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐的制备
制备方法参考实施例12,经Prep-HPLC(方法2)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-异丙 基-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐,产率27.9%;1H NMR(400MHz,DMSO-d6)δ9.14(s,1H),8.13(s,1H),7.80(s,1H),7.63-7.59(m,1H),7.51-7.47(m,1H),7.38-7.11(m,2H),6.55(s,1H),5.66-5.59(m,1H),5.33-5.23(m,1H),2.35(s,3H),1.58(d,J=7.2Hz,3H),1.49-1.47(m,6H);ESI-MS(m/z):391.2[M+H]+
实施例20
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,6-二甲基吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐的制备
制备方法参考实施例12,经Prep-HPLC(方法2)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,6-二甲基吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐,产率21.7%;1H NMR(400MHz,DMSO-d6)δ9.29(s,1H),8.14(s,1H),7.65-7.52(m,2H),7.48(t,J=7.2Hz,1H),7.26(t,J=7.6Hz,1H),7.26(t,J=52.0Hz,1H),6.53(s,1H),5.63-5.56(m,1H),3.69(s,3H),2.35(s,3H),1.54(d,J=7.2Hz,3H);ESI-MS(m/z):363.0[M+H]+
实施例21
(R)-6-环已基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例12,经Prep-HPLC(方法3)纯化,得(R)-6-环己基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率6.0%;1H NMR(400MHz,DMSO-d6)δ9.06(s,1H),7.72(d,J=6.8Hz,1H),7.60(t,J=7.2Hz,1H),7.48(t,J=6.8Hz,1H),7.38-7.10(m,2H),6.56(s,1H),5.69-6.59(t,J=6.8Hz,1H),4.96-4.85(m,1H),2.34(s,3H),1.99-1.72(m,8H),1.58(d,J=7.2Hz,3H),1.52-1.44(m,2H);ESI-MS(m/z):431.0[M+H]+
实施例22
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-(三氟甲基)环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例12,经Prep-HPLC(方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-(三氟甲基)环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率5.3%;1H NMR(400MHz,DMSO-d6)δ9.27(s,1H),7.84 (s,1H),7.58(t,J=7.2Hz,1H),7.49(t,J=6.8Hz,1H),7.37-7.10(m,2H),6.60(s,1H),5.61-5.50(m,1H),2.33(s,3H),1.88-1.67(m,2H),1.60-1.49(m,5H);ESI-MS(m/z):457.0[M+H]+
实施例23
(R)-6-(2,2-二氟乙基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参照实施例12,经Prep-HPLC(方法3)纯化,得(R)-6-(2,2-二氟乙基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮;产率18.2%;1H NMR(400MHz,DMSO-d6)δ9.27(s,1H),7.68(s,1H),7.59(t,J=7.2Hz,1H),7.49(t,J=7.2Hz,1H),7.28(t,J=7.6Hz,1H),7.23(t,J=54.4Hz,1H),6.64(s,1H),6.46((t,J=55.2Hz,1H),5.62(p,J=6.8Hz,1H),4.65-4.48(m,2H),2.36(s,3H),1.55(d,J=7.2Hz,3H);ESI-MS(m/z):413.1[M+H]+
实施例24
(R)-6-(4,4-二氟环己基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例12,经Prep-HPLC(方法3)纯化,得(R)-6-(4,4-二氟环己基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率23.7%;1H NMR(400MHz,DMSO-d6)δ9.02(s,1H),7.81(d,J=6.8Hz,1H),7.59(t,J=7.6Hz,1H),7.48(t,J=7.2Hz,1H),7.27(t,J=7.6Hz,1H),7.24(t,J=56.0Hz,1H),6.60(s,1H),5.67-5.60(m,1H),5.16-5.04(m,1H),2.35(s,3H),2.29-2.16(m,4H),2.07-1.93(m,4H),1.58(d,J=7.2Hz,3H);ESI-MS(m/z):467.0[M+H]+
实施例25
(R)-6-(3,3-二氟环丁基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮三氟乙酸盐的制备
制备方法参考实施例12,经Prep-HPLC(方法4)纯化,得(R)-6-(3,3-二氟环丁基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮三氟乙酸盐,产率31.2%;1H NMR(400MHz,DMSO-d6)δ13.75(s,1H),9.51(s,1H),9.30(s,1H),7.71-7.60(m,2H),7.39-7.12(m,2H),6.91(s,1H),5.58-5.51(m,1H),4.91-4.85(m,1H),3.36-3.17(m,4H),2.47(s,3H),1.70(d,J=6.4Hz,3H);ESI-MS(m/z):439.2[M+H]+
实施例26
2-(4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)-N,N-二甲基丙酚胺的制备
步骤a-f):2-(4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)丙酸甲酯
制备方法参考实施例12的步骤a-f,得2-(4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)丙酸甲酯,产率55.4%;ESI-MS(m/z):435.4[M+H]+
步骤g):2-(4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)丙酸的制备
将2-(4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)丙酸甲酯(280mg,0.645mmol)、氢氧化锂水溶液(4M,0.6mL)、甲醇(2mL)和THF(6mL)加入反应瓶中,室温反应0.5h。以1N的稀盐酸将反应液调至pH=2-3,加入饱和食盐水(20mL),用乙酸乙酯萃取(20mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经中压制备色谱(洗脱剂:乙腈/水=1/9)纯化,得2-(4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)丙酸,产率43.3%;ESI-MS(m/z):421.4[M+H]+
步骤h):2-(4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)-N,N-二甲基丙酰胺的制备
将2-(4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)丙酸(70mg,0.167mmol)、二甲胺盐酸盐(20mg,0.250mmol)、HATU(95mg,0.250mmol)、DIPEA(65mg,0.501mmol)和二氯甲烷(6mL)依次加入反应瓶中,室温搅拌反应30min。加入饱和碳酸氢钠水溶液(20mL)淬灭反应,用二 氯甲烷萃取(20mL×3),合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(方法3)纯化,得2-(4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)-N,N-二甲基丙酰胺,产率13.7%;1H NMR(400MHz,DMSO-d6)δ9.10(s,1H),7.96-7.92(m,1H),7.62-7.56(m,1H),7.52-7.46(m,1H),7.32-7.26(m,1H),7.24(t,J=56.0Hz,1H),6.53(s,1H),6.09-6.01(m,1H),5.72-5.62(m,1H),3.20(s,3H),2.88(s,3H),2.35(s,3H),1.72(d,J=7.2Hz,3H),1.58(t,J=6.8Hz,3H);ESI-MS(m/z):448.0[M+H]+
实施例27
(R)-1-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)-N,N-二甲基环丙烷-1-甲酚胺的制备
制备方法参考实施例26,经Prep-HPLC(方法3)纯化,得(R)-1-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)-N,N-二甲基环丙烷-1-甲酰胺;产率19.0%;1H NMR(400MHz,DMSO-d6)δ9.28(s,1H),7.82(d,J=6.8Hz,1H),7.59(t,J=7.6Hz,1H),7.48(t,J=7.2Hz,1H),7.27(t,J=7.6Hz,1H),7.24(t,J=54.4Hz,1H),6.53(s,1H),5.62(p,J=6.8Hz,1H),2.84(s,6H),2.34(s,3H),1.79-1.68(m,2H),1.63-1.50(m,5H);ESI-MS(m/z):460.2[M+H]+
实施例28
(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
步骤a):1-环丙基-4-羟基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备
将2-甲酰基-3-氧代戊二酸二甲酯(5.6g,27.723mmol)、环丙胺(1.7g,30.495mmol)、甲醇钠(1.5mg,60.975mmol)和甲醇(50mL)加入反应瓶中,80℃加热搅拌反应2h。反应结束后,冷却至室温,加入饱和氯化铵水溶液(50mL)淬灭反应,以4N的盐酸将反应液调至pH=3~4,用二氯甲烷萃取(20mL×3),合并有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得1-环丙基-4-羟基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯,无需纯化直接用于下一步,产率70.7%;ESI-MS(m/z):210.1[M+H]+
步骤b):1-环丙基-6-氧代-4-(((三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-甲酸甲酯的制备
将1-环丙基-4-羟基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(4.1g,19.617mmol)溶解在二氯甲烷(50mL)中,冰浴下加入吡啶(2.0g,25.502mmol),随后滴加三氟甲磺酸酐(7.2g,25.502mmol),滴加完成后,升温至室温搅拌20min。反应结束后,加水(50mL)淬灭反应,以二氯甲烷萃取(20mL×3),合并有机相,以饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=3/1)纯化,得1-环丙基-6-氧代-4-(((三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-甲酸甲酯,产率54.5%;ESI-MS(m/z):342.1[M+H]+
步骤c):4-乙酰基-1-环丙基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备
将1-环丙基-6-氧代-4-(((三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-甲酸甲酯(3.6g,10.557mmol)、三丁基(1-乙氧基乙烯基)锡(6.0g,16.575mmol)、Pd(PPh3)2Cl2(486mg,0.690mmol)、三乙胺(4.2g,42.409mmol)和1,4-二氧六环(50mL)加入反应瓶中,氮气置换三次,升温至100℃搅拌反应5h,反应液冷却至室温,加30%氟化钾水溶液(100mL)淬灭反应,过滤,滤液用乙酸乙酯萃取(20mL×3),合并有机相,用饱和食盐水(20mL×3)洗涤,有机相减压浓缩,加入氯化氢的乙酸乙酯(4M)溶液调至pH=2-3,室温搅拌30min,减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=2/1),得4-乙酰基-1-环丙基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯,产率84.0%;ESI-MS(m/z):236.1[M+H]+
步骤d):6-环丙基-1-甲基吡啶并[3,4-d]哒嗪-4,7(3H,6H)-二酮的制备
将4-乙酰基-1-环丙基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(2.1g,8.936mmol),水合肼(1.1g,18.072mmol,80%)和乙醇(20mL)加入反应瓶中,升温至90℃反应1h。反应结束后,反应液冷却至室温,过滤,滤饼用乙醇(10mL)淋洗,真空干燥,得6-环丙基-1-甲基吡啶并[3,4-d]哒嗪-4,7(3H,6H)-二酮,产率57.9%;ESI-MS(m/z):218.2[M+H]+
步骤e):4-溴-6-环丙基-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将6-环丙基-1-甲基吡啶并[3,4-d]哒嗪-4,7(3H,6H)-二酮(300mg,1.382mmol)、三溴氧磷(770mg,2.764mmol)和乙腈(30mL)依次加入反应瓶中,氮气保护下升温至回流搅拌反应2h。反应结束后,冷却至室温,缓慢滴加饱和碳酸氢钠水溶液(50mL)淬灭反应,以二氯甲烷萃取(50mL×3),合并有机相,用饱和食盐水洗涤(50mL),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:乙酸乙酯/石油醚=4/1),得4-溴-6-环丙基-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率42.6%;ESI-MS(m/z):280.1[M+H]+
步骤f):(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-溴-6-环丙基-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮(165mg,0.589mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙基-1-胺盐酸盐(172mg,0.766mmol)、DIPEA(228mg,1.767mmol)和1,4-二氧六环(3mL)加入反应瓶中,100℃微波反应2h。反应结束后,冷却至室温,加入饱和氯化铵水溶液(20mL)淬灭反应,用乙酸乙酯萃取(20mL×3),合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率22.1%;1H NMR(400MHz,DMSO-d6)δ9.02(s,1H),7.73(d,J=6.8Hz,1H),7.60(t,J=7.6Hz,1H),7.48(t,J=7.2Hz,1H),7.26(t,J=7.6Hz,1H),7.23(t,J=54.4Hz,1H),6.51(s,1H),5.63(p,J=7.2Hz,1H),3.66-3.60(m,1H),2.33(s,3H),1.55(d,J=7.2Hz,3H),1.21-1.08(m,4H);ESI-MS(m/z):389.2[M+H]+
参考实施例28的制备方法,以相应的原料,制得了以下实施例中的化合物。
实施例30
(R)-1-甲基-6-(1-甲基环丙基)-4-(1-(3-(五氟-λ6-磺酰基)苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐的制备
将4-溴-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(100mg,0.341mmol)、(R)-1-(3-(五氟-λ6-磺酰基)苯基)乙烷-1-胺盐酸盐(125mg,0.443mmol)、DIPEA(132mg,1.023mmol)和DMSO(8mL)依次加入微波反应瓶中,105℃下微波反应1.5h。反应结束后,冷却至室温,减压浓缩,残余物经Prep-HPLC(分离方法2)纯化,得(R)-1-甲基-6-(1-甲基环丙基)-4-(1-(3-(五氟-λ6-磺酰基)苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率25.7%;1H NMR(400MHz,DMSO-d6)δ9.21(s,1H),8.15(s,1H),7.90(s,1H),7.75-7.68(m,3H),7.55(t,J=8.0Hz,1H),6.48(s,1H),5.50-5.43(m,1H),2.33(s,3H),1.57(d,J=7.2Hz,3H),1.54(s,3H),1.17-1.12(m,2H),1.10-1.06(m,2H);ESI-MS(m/z):461.0[M+H]+
实施例31
(R)-4-((1-(3-((二氟甲基)磺酚基)-2-甲基苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮制备
步骤a):S-(3-溴-2-甲基苯基)乙酸巯酯的制备
将2-溴-6-碘甲苯(10.0g,33.681mmol)、硫代乙酸钾(4.6g,40.417mmol)、Xant-phos(2.3g,4.042mmol)、Pd2(dba)3(1.8g,2.021mmol)、甲苯(65mL)和乙腈(33mL)加入反应瓶中,氮气置换三次,升温至70℃搅拌反应2h。反应结束后,冷却至室温,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=20/1)纯化,得S-(3-溴-2-甲基苯基)乙酸巯酯,产率93.2%;ESI-MS(m/z):245.0[M+H]+
步骤b):3-溴-2-甲基苯硫酚的制备
将S-(3-溴-2-甲基苯基)乙酸巯酯(7.6g,31.101mmol)和甲醇(80mL)加入反应瓶中,加入氢氧化钾(2.6g,46.512mmol),室温搅拌30min,加水(150mL)稀释,用1N的盐酸调至pH=2~3,以乙酸乙酯(100mL×3)萃取,合并有机相,用饱和食盐水(200mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得3-溴-2-甲基苯硫酚;产率:95.8%;ESI-MS(m/z):203.0[M+H]+
步骤c):(3-溴-2-甲基苯基)(二氟甲基)硫醚的制备
将二氟溴乙酸钠(9.7g,49.483mmol)、碳酸钾(9.1g,65.998mmol)和乙腈(70mL)加入反应瓶中,加入3-溴-2-甲基苯硫酚(6.7g,32.999mmol),升温至100℃搅拌1h。反应结束后,冷却至室温,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=20/1)纯化,得(3-溴-2-甲基苯基)(二氟甲基)硫醚,产率73.0%;ESI-MS(m/z):252.9[M+H]+
步骤d):1-溴-3-((二氟甲基)亚磺酰基)-2-甲基苯的制备
将(3-溴-2-甲基苯基)(二氟甲基)硫醚(5.0g,19.755mmol)和二氯甲烷(60mL)加入反应瓶中,冰水浴下分批加入间氯过氧苯甲酸(10.2g,59.265mmol),升至室温搅拌过夜。反应结束后,加入饱和碳酸氢钠水溶液(100mL)淬灭反应,以二氯甲烷(100mL×3)萃取,合并有机相,以饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=10/1)纯化,得1-溴-3-((二氟甲基)亚磺酰基)-2-甲基苯,产率 33.8%;ESI-MS(m/z):268.9[M+H]+
步骤e):1-(3-((二氟甲基)亚磺酰基)-2-甲基苯)乙烷-1-酮的制备
将1-溴-3-((二氟甲基)亚磺酰基)-2-甲基苯(1.7g,6.317mmol)、三丁基(1-乙氧基乙烯)锡(3.0g,8.213mmol)、三乙胺(1.3g,12.634mmol)、二三苯基膦二氯化钯(222mg,0.316mmol)和1,4-二氧六环(180mL)加入反应瓶中,氮气置换三次,升温至100℃搅拌16h。反应结束后,将反应液冷却至室温,加入2N盐酸(80mL),室温搅拌1h。用乙酸乙酯(150mL×3)萃取,合并有机相,用饱和食盐水(200mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=10/1)纯化,得1-(3-((二氟甲基)亚磺酰基)-2-甲基苯)乙烷-1-酮,产率95.5%;ESI-MS(m/z):233.0[M+H]+
步骤f):1-(3-((二氟甲基)磺酰基)-2-甲基苯)乙烷-1-酮的制备
将1-(3-((二氟甲基)亚磺酰基)-2-甲基苯)乙烷-1-酮(1.2g,4.952mmol)、乙腈(10mL)、水(10mL)和氯仿(10mL)加入反应瓶中,冰浴下加入高碘酸钠(3.18g,14.855mmol)和三氯化钌(103mmol,0.495mmol),室温搅拌2h,加水(100mL)稀释,用二氯甲烷(80mL×3)萃取,合并有机相,以饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=5/1)得1-(3-((二氟甲基)磺酰基)-2-甲基苯)乙烷-1-酮,产率93.3%;ESI-MS(m/z):249.0[M+H]+
步骤g):(R,Z)-N-(1-(3-((二氟甲基)磺酰基)-2-甲基苯基)乙基)-2-甲基丙烷-2-亚磺酰胺的制备
将1-(3-((二氟甲基)磺酰基)-2-甲基苯)乙烷-1-酮(1.1g,4.431mmol)、(R)-(+)-叔丁基亚磺酰胺(698mg,5.760mmol)、钛酸四乙酯(2.3g,6.646mmol,65%)和四氢呋喃(30mL)加入反应瓶中,升温至80℃,搅拌过夜。反应结束后,冷却至室温,加水(50mL)和乙酸乙酯(100mL)室温搅拌5min,过滤,滤液分相,水相以乙酸乙酯(50mL×3)萃取,合并有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=3/1)纯化,得(R,Z)-N-(1-(3-((二氟甲基)磺酰基)-2-甲基苯基)乙基)-2-甲基丙烷-2-亚磺酰胺,产率98.3%;ESI-MS(m/z):352.1[M+H]+
步骤h):(S)-N-((R))-1-(3-((二氟甲基)磺酰基)-2-甲基苯基)乙基)-2-甲基丙烷-2-磺酰胺的制备
将(R,Z)-N-(1-(3-((二氟甲基)磺酰基)-2-甲基苯基)乙基)-2-甲基丙烷-2-亚磺酰胺(1.7g,4.837mmol)、四氢呋喃(30ml)和水(0.5mL)加入反应瓶中,冷却至-78℃,缓慢加入硼氢化钠(275mg,7.256mmol),加毕,升至室温继续搅拌1h,加入饱和氯化铵水溶液(50mL)淬灭反应,以乙酸乙酯(50mL×3)萃取,合并有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=3/1)纯化,得(S)-N-((R))-1-(3-((二氟甲基)磺酰基)-2-甲基苯基)乙基)-2-甲基丙烷-2-磺酰胺,产率46.7%;ESI-MS(m/z):354.1[M+H]+
步骤i):(R)-1-(3-((二氟甲基)磺酰基)-2-甲基苯基)乙烷-1-胺盐酸盐的制备
将(S)-N-((R))-1-(3-((二氟甲基)磺酰基)-2-甲基苯基)乙基)-2-甲基丙烷-2-磺酰胺(798mg,2.258mmol)、1,4-二氧六环(15mL)加入反应瓶中,加入氯化氢的1,4二氧六环溶液(7mL,4N),室温搅拌1h,减压浓缩,残余物加入乙酸乙酯和石油醚(20mL,V/V=1:10)的混合溶剂中,室温搅拌20min,过滤,滤饼干燥,得(R)-1-(3-((二氟甲基)磺酰基)-2-甲基苯基)乙烷-1-胺盐酸盐,产率92.9%;ESI-MS(m/z):250.1[M+H]+
步骤j):(R)-4-((1-(3-((二氟甲基)磺酰基)-2-甲基苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-溴-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(200mg,0.678mmol)、(R)-1-(3-((二氟甲基)磺酰基)-2-甲基苯基)乙烷-1-胺盐酸盐(291mg,1.019mmol)、DIPEA(438mg,3.390mmol)和1,4-二氧六环(8mL)加入反应瓶中,在微波反应器中升温至105℃,搅拌反应3h。反应结束后,将反应液冷却至室温,减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-((二氟甲基)磺酰基)-2-甲基苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率28.0%;1H NMR(400MHz,DMSO-d6)δ9.24(s,1H),7.95(d,J=7.6Hz,1H),7.88(d,J=6.8Hz,1H),7.81(d,J=6.4Hz,1H),7.53(t,J=8.0Hz,1H),7.33(t,J=52.0Hz,1H),6.47(s,1H),5.61(p,J=6.8Hz,1H),2.77(s,3H),2.32(s,3H),1.55-1.50(m,6H),1.20-1.06(m,4H);ESI-MS(m/z):463.1[M+H]+
实施例32
(R)-4-(1-(3-(二氟甲基)-2-氟苯基乙基)氨基)-1-甲基-6-(1-(吡咯烷-1-基甲基)环丙基)甲基)吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐的制备
步骤a):4-羟基-1-(1-(羟甲基)环丙基)甲基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备
将2-甲酰基-3-氧戊二酸二甲酯(10.0g,49.505mmol)、(1-(氨甲基)环丙基)甲醇(6.0g,59.406mmol)、甲醇钠(5.9g,108.911mmol)和甲醇(100mL)加入反应瓶中,升温至80℃搅拌反应2h。反应结束后,冷却至室温,加入饱和氯化铵水溶液(50mL)淬灭反应,以4N的盐酸将反应液调至pH=3-4,用二氯甲烷萃取(50mL×3),合并有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=1/1)纯化,得4-羟基-1-(1-(羟甲基)环丙基)甲基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯,产率47.9%;ESI-MS(m/z):254.2[M+H]+
步骤b):4-(苯甲酰氧基)-1-(1-(苯甲酰氧基)环丙基)甲基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备
将4-羟基-1-(1-(羟甲基)环丙基)甲基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(6.0g,23.715mmol)、二氯甲烷(60mL)和三乙胺(9.6g,94.862mmol)加入反应瓶中,滴加苯甲酰氯(10.0g,71.145mmol),加毕,室温搅拌反应16h。加水(100mL)淬灭反应,以二氯甲烷萃取(50mL×3),合并有机相,饱和食盐水洗(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=2/1)纯化,得4-(苯甲酰氧基)-1-(1-(苯甲酰氧基)环丙基)甲基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯,产率88.7%;ESI-MS(m/z):462.2[M+H]+
步骤c):1-((1-(苯甲酰氧基)甲基)环丙基)甲基)-4-羟基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备
将4-(苯甲酰氧基)-1-(1-(苯甲酰氧基)环丙基)甲基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(9.7g,21.041mmol)和甲醇(100mL)加入反应瓶中,冰水浴下加入碳酸钾(5.8g,42.082mmol),冰水浴搅拌反应10min。加入饱和氯化铵水溶液(50mL)淬灭反应,以4N的盐酸将反应液调至pH=3-4,用二氯甲烷萃取(20mL×3),合并有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=1/1)纯化,得1-((1-(苯甲酰氧基)甲基)环丙基)甲基)-4-羟基-6-氧代-1,6-二氢吡啶-3-羧酸甲酯,产率81.2%;ESI-MS(m/z):358.2[M+H]+
步骤d):1-((1-(苯甲酰氧基)甲基)环丙基)甲基)-6-氧代-4-((三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-甲酸甲酯的制备
将1-((1-(苯甲酰氧基)甲基)环丙基)甲基)-4-羟基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(6.1g,17.087mmol)和二氯甲烷(60mL)加入反应瓶中,冰水浴下加入吡啶(2.7g,34.174mmol),滴加三氟甲磺酸酐(7.2g,25.631mmol),冰水浴下继续搅拌反应20min。加水(60mL)淬灭反应,以二氯甲烷萃取(30mL×3),合并有机相,饱和食盐水洗(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=3/1)纯化,得1-((1-(苯甲酰氧基)甲基)环丙基)甲基)-6-氧代-4-((三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-甲酸甲酯,产率93.3%;ESI-MS(m/z):490.1[M+H]+
步骤e):4-乙酰基-1-(1-(苯甲酰氧基)甲基)环丙基)甲基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备
将1-((1-(苯甲酰氧基)甲基)环丙基)甲基)-6-氧代-4-((三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-甲酸甲酯(7.8g,16.155mmol)、三丁基(1-乙氧基乙烯基)锡(6.3g,14.650mmol)、Pd(PPh3)2Cl2(486mg,0.690mmol)、三乙胺(4.0g,40.388mmol)和1,4-二氧六环(80mL)加入反应瓶中,氮气置换三次,升温至100℃搅拌反应5h。反应结束后,冷却至室温,加KF水溶液(80mL)淬灭反应,过滤,滤液用乙酸乙酯萃取(30mL×3),合并有机相,减压浓缩,加入氯化氢的乙酸乙酯(30mL,4N)溶液,室温搅拌30min,减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=3/1)纯化,得4-乙酰基-1-(1-(苯甲酰氧基)甲基)环丙基)甲基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯,产率50.5%;ESI-MS(m/z):384.2[M+H]+
步骤f):(1-((1-甲基-4,7-二氧代-3,7-二氢吡啶并[3,4-d]哒嗪-6(4H)-基)甲基)环丙基)苯甲酸甲酯的制备
将4-乙酰基-1-(1-(苯甲酰氧基)甲基)环丙基)甲基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(3.1g,8.094mmol)、水合肼(917mg,24.282mmol,80%)和乙醇(30mL)加入反应瓶中,升温至90℃搅拌反应1h。反应结束后,冷却至室温,过滤,滤饼用乙醇(10mL)淋洗,真空干燥,得(1-((1-甲基-4,7-二氧代-3,7-二氢吡啶并[3,4-d]哒嗪-6(4H)-基)甲基)环丙基)苯甲酸甲酯,产率81.2%;ESI-MS(m/z):366.1[M+H]+
步骤g):(1-(4-溴-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)甲基)环丙基)苯甲酸甲酯的制备
将(1-((1-甲基-4,7-二氧代-3,7-二氢吡啶并[3,4-d]哒嗪-6(4H)-基)甲基)环丙基)苯甲酸甲酯(500mg,1.370mmol)、三溴氧磷(777mg,2.740mmol)和乙腈(50mL)依次加入反应瓶中,氮气保护下升温至105℃搅拌反应2h。反应结束后,冷却至室温,加饱和碳酸氢钠水溶液(50mL)淬灭反应,以二氯甲烷萃取(50mL×3),合并有机相,用饱和食盐水洗涤(50mL),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:乙酸乙酯/石油醚=4/1)纯化,得(1-(4-溴-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)甲基)环丙基)苯甲酸甲酯,产率68.2%;ESI-MS(m/z):428.1[M+H]+
步骤h):(R)-(1-((4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)甲基)环丙基)苯甲酸甲酯的制备
将(1-(4-溴-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)甲基)环丙基)苯甲酸甲酯(408mg,0.953mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙-1-胺盐酸盐(336mg,1.430mmol)、DIPEA(492mg,3.812mmol)和1,4-二氧六环(10mL)加入反应瓶中,100℃微波反应2h。反应结束后,冷却至室温,加入饱和氯化铵水溶液(20mL)淬灭反应,用乙酸乙酯萃取(20mL×3),合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:乙酸乙酯/石油醚=4/1)纯化,得(R)-(1-((4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)甲基)环丙基)苯甲酸甲酯,产率58.7%;ESI-MS(m/z):537.3[M+H]+
步骤i):(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(羟甲基)环丙基)甲基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-(1-((4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)甲基)环丙基)苯甲酸甲酯(300mg,0.560mmol)、一水合氢氧化锂(47mg,1.120mmol)和甲醇(50mL)加入反应瓶中,室温搅拌反应1h。反应结束后,加水(20mL)淬灭反应,用二氯甲烷萃取(20mL×3),合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(羟甲基)环丙基)甲基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,无需纯化直接用于下一步,产率98.7%;ESI-MS(m/z):433.1[M+H]+
步骤j):(R)-6-((1-(氯甲基)环丙基)甲基)-4-(1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(羟甲基)环丙基)甲基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮(230mg,0.532mmol)和氯仿(50mL)加入反应瓶中,滴加氯化亚砜(630mg,5.032mmol),加毕室温搅拌反应1h。反应结束后,减压浓缩,得(R)-6-((1-(氯甲基)环丙基)甲基)-4-(1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,无需纯化直接用于下一步,产率97.1%;ESI-MS(m/z):451.2[M+H]+
步骤k):(R)-4-(1-(3-(二氟甲基)-2-氟苯基乙基)氨基)-1-甲基-6-(1-(吡咯烷-1-基甲基)环丙基)甲基)吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐的制备
将(R)-6-((1-(氯甲基)环丙基)甲基)-4-(1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮(252mg,0.559mmol)、吡咯烷(252mg,1.112mmol)、DIPEA(216mg,1.677mmol)和乙腈(10mL)加入 反应瓶中,升温至80℃搅拌反应8h。反应结束后,冷却至室温,减压浓缩,残余物经Prep-HPLC(分离方法2)纯化,得(R)-4-(1-(3-(二氟甲基)-2-氟苯基乙基)氨基)-1-甲基-6-(1-(吡咯烷-1-基甲基)环丙基)甲基)吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐,产率17.5%;1H NMR(400MHz,DMSO-d6)δ9.23(s,1H),8.16(s,1H),7.60(t,J=7.6Hz,1H),7.54-7.45(m,2H),7.26(t,J=7.6Hz,1H),7.23(t,J=56.0Hz,1H),6.47(s,1H),5.63-5.57(m,1H),4.18(s,2H),2.42-2.38(m,4H),2.34(s,3H),2.24(s,2H),1.59-1.51(m,7H),0.87-0.77(m,2H),0.40-0.33(m,2H);ESI-MS(m/z):486.0[M+H]+
实施例33
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(2-羟乙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐的制备
制备方法参考实施例32的步骤a-i,所得粗品经Prep-HPLC(分离方法2)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(2-羟乙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐,产率18.4%;1H NMR(400MHz,DMSO-d6)δ13.7(1H),9.28(s,1H),8.14(1H),7.84(s,1H),7.60(t,J=7.6Hz,1H),7.50(t,J=7.2Hz,1H),7.28(t,J=7.6Hz,1H),7.24(d,J=54.4Hz,1H),6.57(s,1H),5.61(q,J=7.2Hz,1H),5.06(t,J=5.6Hz,1H),4.25-4.20(1H),4.16-4.10(1H),3.75(q,J=5.4Hz,2H),2.36(s,3H),1.56(d,J=7.2Hz,3H);ESI-MS(m/z):393.1[M+H]+
实施例34
4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(4-羟基丁-2-基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例32的步骤a-i,所得粗品经Prep-HPLC(分离方法3)纯化,得4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(4-羟基丁-2-基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率26.7%;1H NMR(400MHz,DMSO-d6)δ9.12(s,1H),7.70(d,J=6.8Hz,1H),7.59(t,J=7.2Hz,1H),7.48(t,J=7.2Hz,1H),7.27(t,J=7.6Hz,1H),7.24(t,J=56.0Hz,1H),6.53(s,1H),5.66-5.59(m,1H),5.33-5.25(m,1H),4.57(s,1H),3.44-3.39(m,2H),2.34(s,3H),2.06-2.00(m,2H),1.57(d,J=7.2Hz,3H),1.47(d,J=6.8Hz,3H));ESI-MS(m/z):421.0[M+H]+
实施例35
(R)-4-(1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-((1-(二氟甲基)环丙基)甲基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
步骤a):(R)-4-(1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)甲基)环丙烷-1-甲醛的制备
将(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(羟甲基)环丙基)甲基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮(200mg,0.463mmol)、戴斯-马丁氧化剂(295mg,0.695mmol)和二氯甲烷(10mL)加入反应瓶中,室温搅拌反应5h,加水(20mL)淬灭反应,以二氯甲烷萃取(20mL×3),合并有机相,以饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:二氯甲烷/甲醇=20/1)纯化,得(R)-4-(1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)甲基)环丙烷-1-甲醛,产率65.7%;ESI-MS(m/z):431.0[M+H]+
步骤b):(R)-4-(1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-((1-(二氟甲基)环丙基)甲基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-4-(1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)甲基)环丙烷-1-甲醛(130mg,0.302mmol)和二氯甲烷(10mL)加入反应瓶中,滴加二乙胺基三氟化硫(97mg,0.604mmol),滴毕,室温搅拌反应18h。反应结束后,加入饱和碳酸氢钠水溶液(20mL)淬灭反应,以二氯甲烷萃取(20mL×3),合并有机相,以饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-4-(1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-((1-(二氟甲基)环丙基)甲基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率14.7%;1H NMR(400MHz,DMSO-d6)δ9.20(s,1H),7.70-7.59(m,1H),7.56(d,J=7.6Hz,1H),7.48(t,J=7.2Hz,1H),7.27(t,J=7.6Hz,1H),7.23(t,J=52.0Hz,1H),6.57(s,1H),5.96(t,J=56.0Hz,1H),5.66-5.59(m,1H),4.43-4.25(m,2H),2.35(s,3H),1.56(d,J=7.2Hz,3H),1.01-0.98(m,2H),0.88-0.84(m,2H);ESI-MS(m/z):453.0[M+H]+
实施例36
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(羟甲基)环丙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例32的步骤a-i,所得粗品经Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(羟甲基)环丙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率23.7%;1H NMR(400MHz,DMSO-d6)δ9.16(s,1H),7.84(d,J=6.8Hz,1H),7.58(t,J=7.6Hz,1H),7.48(t,J=7.2Hz,1H),7.26(t,J=7.6Hz,1H),7.24(t,J=56.0Hz,1H),6.48(s,1H),5.67-5.61(m,1H),5.03(t,J=5.8Hz,1H),3.70(s,1H),3.58(s,1H),2.33(s,3H),1.56(d,J=7.2Hz,3H),1.24-1.10(m,4H);ESI-MS(m/z):419.0[M+H]+
实施例37
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(二氟甲基)环丙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例35,所得粗品经Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(二氟甲基)环丙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率15.7%;1H NMR(400MHz,DMSO-d6)δ9.20(s,1H),7.86(s,1H),7.58(t,J=7.2Hz,1H),7.48(t,J=7.2Hz,1H),7.27(t,J=7.6Hz,1H),7.24(t,J=56.0Hz,1H),6.58(s,1H),6.34(t,J=56.0Hz,1H),5.66-5.59(m,1H),2.34(s,3H),1.59-1.52(m,5H),1.50-1.38(m,2H);ESI-MS(m/z):439.0[M+H]+
实施例38
4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-((四氢呋喃-3-基)甲基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例28,所得粗品经Prep-HPLC(分离方法3)纯化,得4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-((四氢呋喃-3-基)甲基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率21.8%;1H NMR(400MHz,DMSO-d6)δ9.28(s,1H),7.62-7.56(m,2H),7.50-7.47(m,1H),7.37-7.10(m,2H),6.56(s,1H),5.65-5.59(m,1H),4.20-4.06(m,2H),3.88-3.83(m,1H),3.69-3.56(m,3H),2.88-2.78(m,1H),2.35(s,3H),2.01-1.91(m,1H),1.73-1.64(m,1H),1.56(d,J=7.2Hz,3H);ESI-MS(m/z):433.2[M+H]+
实施例39
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(氟甲基)环丙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(羟甲基)环丙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮(150mg,0.359mmol)和二氯甲烷(10mL)加入反应瓶中,冰水浴下滴加二乙胺基三氟化硫(97mg,0.604mmol),冰浴下搅拌反应20min。升至室温,加入饱和碳酸氢钠水溶液(20mL)淬灭反应,以二氯甲烷萃取(20mL×3),合并有机相,以饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(氟甲基)环丙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率13.2%;1H NMR(400MHz,DMSO-d6)δ9.24(s,1H),7.90(s,1H),7.59(t,J=7.6Hz,1H),7.49(t,J=7.2Hz,1H),7.27(t,J=7.6Hz,1H),7.24(t,J=56.0Hz,1H),6.57(s,1H),5.65-5.58(m,1H),4.78-4.52(m,2H),2.34(s,3H),1.57(d,J=7.2Hz,3H),1.37-1.32(m,4H);ESI-MS(m/z):421.0[M+H]+
实施例40
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-甲氧基-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐的制备
步骤a):(R)-8-溴-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
室温下将(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(2.5g,6.213mmol)和二氯甲烷(20mL)加入反应瓶中,滴加液溴(2.0g,12.426mmol),滴毕,室温搅拌反应3h。反应结束后,加饱和硫代硫酸钠水溶液(20mL)淬灭,以二氯甲烷(50mL)萃取,有机相以饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(石油醚/乙酸乙酯=1/2)纯化,得(R)-8-溴-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮;产率:43.5%;ESI-MS(m/z):481.1[M+H]+
步骤b):(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-甲氧基-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐的制备
室温下将(R)-8-溴-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(190mg,0.392mmol)和甲醇(5mL)加入反应瓶中,缓慢加入甲醇钠(32mg,0.588mmol),室温反应30min,加饱和氯化铵水溶液(10mL)淬灭,以乙酸乙酯(20mL×2)萃取,合并有机相,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法1)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-8-甲氧基-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐,产率47.4%;1H NMR(400MHz,DMSO-d6)δ13.46(s,1H),9.60(s,1H),7.59-7.76(m,2H),7.11-7.38(m,3H),5.49-5.53(m,1H)3.99(s,3H),2.61(s,3H),1.70-1.72(d,J=8.0Hz,3H),1.56(s,3H),1.09-1.26(m,4H);ESI-MS(m/z):433.1[M+H]+
实施例41
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,8-二甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐的制备
室温下将(R)-8-溴-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(230mg,0.483mmol)、1,4-二氧六环(4mL)、水(1mL)、甲基硼酸(58mg,0.967mmol)、Pd(dppf)Cl2(72mg,0.096mmol)和碳酸钾(200mg,1.449mmol)加入微波管中,氮气吹扫1min,100℃微波反应1.5h。反应结束后,冷却至室温,加饱和氯化铵水溶液(20mL)淬灭,以乙酸乙酯(20mL×2)萃取,合并有机相,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法1)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,8-二甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐,产率20.0%;1H NMR(400MHz,DMSO-d6)δ13.45(s,1H),9.81(s,1H),7.59-7.79(m,2H),7.11-7.38(m,3H),5.51-5.55(m,1H),2.71(s,3H),2.50(s,3H),1.71-1.73(d,J=8.0Hz,3H),1.56(s,3H),1.08-1.27(m,4H);ESI-MS(m/z):417.2[M+H]+
实施例42
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-8-腈的制备
室温下将(R)-8-溴-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(160mg,0.332mmol)、DMF(5mL)、氰化锌(59mg,0.659mmol)和四(三苯基膦)钯(7.7mg,6.661mmol)加入反应瓶中,氮气置换三次,升温至100℃反应1h。反应结束后,冷却至室温,加饱和氯化铵水溶液(10mL)淬灭,以乙酸乙酯(20mL×2)萃取,合并有机相,用饱和食盐水(30mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基环丙基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-8-腈,产率17.7%;1H NMR(400MHz,DMSO-d6)δ9.89(s,1H),7.55-7.73(m,2H),7.11-7.38(m,3H),5.57-5.62(m,1H),2.74(s,3H),1.66-1.68(d,J=8.0Hz,3H),1.59(s,3H),1.10-1.32(m,4H);ESI-MS(m/z):428.1[M+H]+
实施例43
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲氧基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
步骤a):4-氰基-1-(1-甲基环丙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备
室温下将1-(1-甲基环丙基)-6-氧代-4-(((三氟甲基)磺酰基)氧基)-1,6-二氢吡啶-3-甲酸甲酯(4.3g,12.113mmol)、氰化锌(2.2g,24.226mmol)、四三苯基膦钯(280mg,0.242mmol)和DMF(20mL)加入反应瓶中,氮气置换三次,升温至80℃反应1h。反应结束后,冷却过滤,滤液倒入水中(30mL),以乙酸乙酯萃取(50mL×2),合并有机相,以饱和食盐水洗涤(50mL×3)),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=1/1)纯化,得4-氰基-1-(1-甲基环丙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯,产率78.2%;ESI-MS(m/z):233.1[M+H]+
步骤b):1-(1-甲基环丙基)-6-氧代-1,6-二氢吡啶-3,4-二甲酸二甲酯的制备
室温下将4-氰基-1-(1-甲基环丙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(2.1g,9.052mmol)和6N的盐酸(10mL)加入反应瓶中,升温至回流反应5h,加入甲醇(40mL),继续回流反应过夜。反应结束后,冷却至室温,减压除去有机溶剂,加水(30mL)稀释,以乙酸乙酯(50mL×2)萃取,合并有机相,饱和碳酸氢钠水溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得1-(1-甲基环丙基)-6-氧代-1,6-二氢吡啶-3,4-二甲酸二甲酯,无需纯化直接用于下一步反应,产率75.1%,ESI-MS(m/z):266.0[M+H]+
步骤c):6-(1-甲基环丙基)-2,3-二氢吡啶并[3,4-d]哒嗪-1,4,7(6H)-三酮的制备
室温下将1-(1-甲基环丙基)-6-氧代-1,6-二氢吡啶-3,4-二甲酸二甲酯(1.6g,6.038mmol)和水合肼(10mL,80%)加入反应瓶中,升温至100℃反应过夜,将反应液冷却至室温,加入乙腈(100mL),室温搅拌10min,过滤,滤饼用乙腈(10mL)淋洗,真空干燥,得6-(1-甲基环丙基)-2,3-二氢吡啶并[3,4-d]哒嗪-1,4,7(6H)-三酮,产率68.3%; ESI-MS(m/z):234.0[M+H]+
步骤d):1,4-二溴-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
室温下将6-(1-甲基环丙基)-2,3-二氢吡啶并[3,4-d]哒嗪-1,4,7(6H)-三酮(780mg,3.348mmol)、三溴氧膦(1.9g,6.696mmol)和乙腈(70mL)加入反应瓶中,升温至105℃反应3h。反应结束后,冷却至室温,缓慢倾入饱和碳酸氢钠水溶液(200mL)中,充分搅拌,用二氯甲烷(100mL×2)萃取,合并有机相,饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=1/2),得1,4-二溴-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率47%;ESI-MS(m/z):358.0[M+H]+
步骤e):(R)-1-氯-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将1,4-二溴-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(205mg,0.571mmol)、DIPEA(369mg,2.855mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺盐酸盐(142mg,0.628mmol)和1,4-二氧六环(5mL)加入微波反应瓶中,在微波反应器中105℃反应1h,将反应液冷却至室温,减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=1/2)纯化,得(R)-1-氯-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率56.0%;ESI-MS(m/z):423.1[M+H]+
步骤f):(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲氧基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-1-氯-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(65mg,0.154mmol)、甲醇钠(13mg,0.231mmol)和甲醇加入到反应瓶中,升温至80℃搅拌2h。反应结束后,冷却至室温,加入二氯甲烷(30ml)稀释,倒入饱和氯化铵水溶液(50mL)中,以二氯甲烷萃取(30mL×2),合并有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲氧基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率35.0%;1H NMR(400MHz,DMSO-d6)δ9.21(s,1H),7.61(t,J=7.6Hz,1H),7.47(d,J=7.2Hz,2H),7.38-7.11(m,2H),6.48(s,1H),5.50(p,J=6.4Hz,1H),3.83(s,3H),1.55-1.52(m,6H),1.18-1.06(m,4H);ESI-MS(m/z):419.2[M+H]+
实施例44
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-羟基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-1-氯-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(65mg,0.154mmol)和醋酸(3mL)加入反应瓶中,升温至100℃搅拌反应30min。反应结束后,冷却至室温,减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-羟基-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮;产率33.0%;1H NMR(400MHz,DMSO-d6)δ11.14(s,1H),8.98(s,1H),7.61(t,J=7.6Hz,1H),7.48(t,J=7.2Hz,1H),7.29(t,J=7.6Hz,1H),7.22(t,J=54.4Hz,1H),6.99(d,J=6.8Hz,1H),6.80(s,1H),5.17(p,J=6.8Hz,1H),1.56-1.47(m,6H),1.20-1.03(m,4H);ESI-MS(m/z):405.2[M+H]+
实施例45
(R)-3-(1-((6-环丙基-1-甲基-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-4-基)氨基)乙基)-2-甲基苯甲腈的制备
将4-溴-6-环丙基-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮(100mg,0.357mmol)、DIPEA(231mg,1.786mmol)、(R)-3-(1-氨基乙基)-2-甲基苯甲腈盐酸盐(105mg,0.535mmol)和1,4-二氧六环(5mL)加入反应瓶中,在微波反应器中升温至105℃搅拌反应1h。反应结束后,冷却至室温,减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-3-(1-((6-环丙基-1-甲基-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-4-基)氨基)乙基)-2-甲基苯甲腈,产率31.7%;1H NMR(400MHz,DMSO-d6)δ8.99(s,1H),7.78(s,1H),7.72(d,J=7.6,1H),7.63(d,J=7.6,1H),7.34(t,J=7.6Hz,1H),6.50(s,1H),5.50(p,J=6.8Hz,1H),3.64-3.58(m,1H),2.62(s,3H),2.33(s,3H),1.50(d,J=7.2Hz,3H),1.18-1.06(m,4H);ESI-MS(m/z):360.2[M+H]+
实施例46
(R)-6-环丙基-1-甲基-4-((1-(2-甲基-3-(三氟甲基)苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例45,所得粗品经Prep-HPLC(分离方法3)纯化,得(R)-6-环丙基-1-甲基-4-((1-(2-甲基-3-(三氟甲基)苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率10.3%;1H NMR(400MHz,DMSO-d6)δ8.99(s,1H),7.78-7.70(m,2H),7.53(d,J=8.0Hz,1H),7.34(t,J=7.6Hz,1H),6.49(s,1H),5.67-5.56(m,1H),3.67-3.57(m,1H),2.53(s,3H),2.33(s,3H),1.50(d,J=6.8Hz,3H),1.20-1.06(m,4H);ESI-MS(m/z):403.0[M+H]+
实施例47
(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,8-二甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
步骤a):4-乙酰基-5-溴-1-环丙基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备
将4-乙酰基-1-环丙基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(3.4g,14.468mmol)、NBS(3.1g,17.362mmol)和醋酸(30mL)加入反应瓶中,室温搅拌反应24h。反应结束后,将反应液减压浓缩,加入饱和碳酸氢钠水溶液(50mL)淬灭反应,用二氯甲烷萃取(20mL×3),合并有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=3/1),得4-乙酰基-5-溴-1-环丙基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯,产率86.1%;ESI-MS(m/z):314.0[M+H]+
步骤b):4-乙酰基-1-环丙基-5-甲基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备
将4-乙酰基-5-溴-1-环丙基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(2.5g,7.962mmol)、甲基硼酸(704mg,11.943mmol)、K2CO3(2.2g,15.924mmol)、Pd(PPh3)4(460mg,0.398mmol)、1,4-二氧六环(30mL)和水(6mL)依次加入反应瓶中,氮气置换三次,100℃搅拌反应16h。反应结束后,冷却至室温,加水(50mL)淬灭反应,以乙酸乙酯萃取(20mL×3),合并有机相,以饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=3/1)纯化,得4-乙酰基-1-环丙基-5-甲基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯,产率33.0%;ESI-MS(m/z):250.1[M+H]+
步骤c):6-环丙基-1,8-二甲基吡啶并[3,4-d]哒嗪-4,7(3H,6H)-二酮的制备
将4-乙酰基-1-环丙基-5-甲基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(650mg,2.610mmol)、水合肼(0.5mL,80%)和乙醇(10mL)加入反应瓶中,升温至90℃搅拌反应1h。反应结束后,将反应液冷却至室温,过滤,滤饼用乙醇(2mL)淋洗,真空干燥,得6-环丙基-1,8-二甲基吡啶并[3,4-d]哒嗪-4,7(3H,6H)-二酮,产率92.3%;ESI-MS(m/z):232.1[M+H]+
步骤d):4-溴-6-环丙基-1,8-二甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将6-环丙基-1,8-二甲基吡啶并[3,4-d]哒嗪-4,7(3H,6H)-二酮(300mg,1.299mmol)、三溴氧磷(743mg,2.597mmol)和乙腈(50mL)依次加入反应瓶中,氮气保护下升温至回流搅拌反应2h。反应结束后,将反应液冷却至室温,加饱和碳酸氢钠水溶液(50mL)淬灭反应,以二氯甲烷萃取(50mL×3),合并有机相,用饱和食盐水洗涤(50mL),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:乙酸乙酯/石油醚=4/1),得4-溴-6-环丙基-1,8-二甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率49.7%;ESI-MS(m/z):294.1[M+H]+
步骤e):(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,8-二甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-溴-6-环丙基-1,8-二甲基吡啶并[3,4-d]哒嗪-7(6H)-酮(110mg,0.374mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙-1-胺盐酸盐(132mg,0.561mmol)、DIPEA(241mg,1.870mmol)和1,4-二氧六环(3mL)加入反应瓶中,100℃微波反应1h。加入饱和氯化铵水溶液(20mL)淬灭反应,用乙酸乙酯萃取(20mL×3),合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1,8-二甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率23.5%;1H NMR(400MHz,DMSO-d6)δ8.80(s,1H),7.63(d,J=6.8Hz,1H),7.57(t,J=7.6Hz,1H),7.47(t,J=7.2Hz,1H),7.25(t,J=7.6Hz,1H),7.24(t,J=54.0Hz,1H),5.64-5.57(m,1H),3.66-3.61(m,1H),2.61(s,3H),2.46(s,3H),1.55(d,J=7.2Hz,3H),1.16-1.13(m,2H),1.11-1.08(m,2H);ESI-MS(m/z):403.0[M+H]+
实施例48
(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-甲基苯基)乙基)氨基)-1,8-二甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例47步骤e,所得粗品经Prep-HPLC(分离方法3)纯化,得(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-甲基苯基)乙基)氨基)-1,8-二甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率13.5%;1H NMR(400MHz,DMSO-d6)δ8.78(s,1H),7.65-7.55(m,2H),7.36(t,J=7.6Hz,1H),7.26(t,J=7.6Hz,1H),7.21(t,J=56.0Hz,1H),5.63-5.56(m,1H),3.65-3.59(m,1H),2.61(s,3H),2.45(s,6H),1.48(d,J=6.8Hz,3H),1.17-1.05(m,4H);ESI-MS(m/z):399.0[M+H]+
实施例49
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(哌啶-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
步骤a-d):4-(1-甲基-4,7-二氧代-3,7-二氢吡啶并[3,4-d]哒嗪-6(4H)-基)哌啶-1-甲酸叔丁酯的制备
制备方法参考实施例28的步骤a-d。
步骤e):4-(1-甲基-7-氧代-4-(1H-1,2,4-三氮唑-1-基)吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-甲酸叔丁酯的制备
将4-(1-甲基-4,7-二氧代-3,7-二氢吡啶并[3,4-d]哒嗪-6(4H)-基)哌啶-1-甲酸叔丁酯(2.0g,5.549mmol)、1H-1,2,4-三氮唑(3.1g,44.394mmol)、乙腈(60mL)和三乙胺(9.0g,88.788mmol)加入反应瓶中,三氯氧磷(1.7g,11.098mmol)的乙腈(20mL)溶液滴加到反应液中,维持室温搅拌反应过夜。反应结束后,减压浓缩除去大部分溶剂,加水(100mL)室温打浆20min,过滤,滤饼用水(10mL×2)淋洗,真空干燥,得4-(1-甲基-7-氧代-4-(1H-1,2,4-三氮唑-1-基)吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-甲酸叔丁酯,产率90.1%;ESI-MS(m/z):412.2[M+H]+
步骤f):(R)-4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-甲酸叔丁酯的制备
将4-(1-甲基-7-氧代-4-(1H-1,2,4-三氮唑-1-基)吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-甲酸叔丁酯(2.2g,5.347mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺盐酸盐(1.7g,7.485mmol)和1,4-二氧六环(20mL)加入微波管中,维持110℃下微波反应4h。反应结束后,将反应液转移至圆底瓶中减压浓缩,残余物用硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=15/1),得(R)-4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-甲酸叔丁酯,产率88.6%;ESI-MS(m/z):518.2[M+H]+
步骤g):(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(哌啶-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-甲酸叔丁酯(100mg,0.193mmol)和四氢呋喃(2mL)加入反应瓶中,室温下向反应液中滴加氯化氢的1,4-二氧六环溶液(4M,2mL),维持室温反应0.5h。反应结束后,将反应液减压浓缩,残余物用二氯甲烷(4mL)溶解,加入三乙胺(1mL),减压浓缩,残余物用Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(哌啶-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率41.8%;1H NMR(400MHz,DMSO-d6)δ9.16(s,1H),7.86(d,J=6.8Hz,1H),7.58(t,J=7.2Hz,1H),7.48(t,J=7.2Hz,1H),7.27(t,J=7.6Hz,1H),7.24(t,J=54.4Hz,1H),6.55(s,1H),5.66-5.51(m,1H),5.03-4.95(m,1H),3.16-3.13(m,2H),2.70-2.64(m,2H),2.44-2.40(m,1H),2.34(s,3H),2.03-1.93(m,2H),1.83-1.75(m,2H),1.57(d,J=6.8Hz,3H);ESI-MS(m/z):432.0[M+H]+
参考实施例49的制备方法,以相应的原料,制得了以下实施例中的化合物。
实施例51
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-(吗啉-4-羰基)哌啶-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(哌啶-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐(100mg,0.214mmol)、二氯甲烷(2mL)和三乙胺(86mg,0.855mmol)加入反应瓶中,将吗啉-4-甲酰氯(320mg,2.137mmol)的二氯甲烷(3mL)溶液缓慢滴加至反应液中,维持室温搅拌反应1h。反应结束后,将反应液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-(吗啉-4-羰基)哌啶-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率21.2%;1H NMR(400MHz,DMSO-d6)δ9.05(s,1H),7.85(d,J=6.4Hz,1H),7.59(t,J=7.2Hz,1H),7.48(t,J=7.2Hz,1H),7.26(t,J=7.6Hz,1H),7.24(t,J=54.0Hz,1H),6.59(s,1H),5.66-5.59(m,1H),5.11-5.03(m,1H),3.89(d,J=12.8Hz,2H),3.60(t,J=4.8Hz,4H),3.17(t,J=4.8Hz,4H),3.02(d,J=12.4Hz,2H),2.34(s,3H),2.02-1.88(m,4H),1.57(d,J=6.8Hz,3H);ESI-MS(m/z):545.0[M+H]+
实施例52
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基哌啶-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(哌啶-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐(100mg,0.214mmol)、二氯甲烷(2mL)和三乙胺(86mg,0.855mmol)加入反应瓶中,将碘甲烷(33mg,0.235mmol)的二氯甲烷(2mL)溶液滴加至反应液中,维持室温搅拌反应1h。反应结束后,减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基哌啶-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率26.5%;1H NMR(400MHz,DMSO-d6)δ9.18(s,1H),7.81(d,J=6.8Hz,1H),7.58(t,J=7.6Hz,1H),7.48(t,J=7.2Hz,1H),7.27(t,J=7.6Hz,1H),7.24(t,J=54.4Hz,1H),6.56(s,1H),5.65-5.58(m,1H),4.97-4.90(m,1H),2.98(d,J=8.8Hz,2H),2.34(s,3H),2.26(s,3H),2.19-2.06(m,4H),1.83-1.80(m,2H),1.56(d,J=6.8Hz,3H);ESI-MS(m/z):446.0[M+H]+
实施例53
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(异噁唑-4-羰基)哌啶-4-基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(哌啶-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐(100mg,0.214mmol)、异噁唑-4-甲酸(48mg,0.428mmol)、HATU(163mg,0.428mmol)、二氯甲烷(2mL)和三乙胺(86mg,0.855mmol)加入反应瓶中,室温搅拌反应2h。反应结束后,减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(异噁唑-4-羰基)哌啶-4-基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率38.8%;1H NMR(400MHz,DMSO-d6)δ9.44(s,1H),9.06(s,1H),8.91(s,1H),7.72(s,1H),7.58(t,J=7.6Hz,1H),7.48(t,J=7.2Hz,1H),7.26(t,J=7.6Hz,1H),7.23(t,J=54.4Hz,1H),6.60(s,1H),5.66-5.60(m,1H),5.28-5.21(m,1H),4.81-4.74(m,1H),4.21-4.12(m,1H),3.51-3.42(m,1H),3.04-3.96(m,1H),2.35(s,3H),2.07-1.91(m,4H),1.56(d,J=6.8Hz,3H);ESI-MS(m/z):527.0[M+H]+
实施例54
(R)-6-(1-乙酰基哌啶-4-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶酮[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(哌啶-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐(100mg,0.214mmol)、乙酰氯(18mg,0.235mmol)、二氯甲烷(2mL)和三乙胺(86mg,0.855mmol)加入反应瓶中,室温搅拌反应1h。反应结束后,将反应液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-6-(1-乙酰基哌啶-4-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶酮[3,4-d]哒嗪-7(6H)-酮,产率22.7%;1H NMR(400MHz,DMSO-d6)δ9.05(s,1H),7.73(t,J=7.6Hz,1H),7.58(t,J=7.6Hz,1H),7.48(t,J=7.2Hz,1H),7.27(t,J=7.2Hz,1H),7.24(t,J=54.4Hz,1H),6.59(s,1H),5.66-5.59(m,1H),5.22-5.14(m,1H),4.67(t,J=13.2Hz,1H),4.07(t,J=13.6Hz,1H),3.30-3.24(m,1H),2.76-2.67(m,1H),2.35(s,3H),2.09(s,3H),1.97-1.88(m,4H),1.57(d,J=6.8Hz,3H);ESI-MS(m/z):474.0[M+H]+
实施例55
(R)-6-(1-苯甲酚基哌啶-4-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例54,所得粗品经Prep-HPLC(分离方法3)纯化,得(R)-6-(1-苯甲酰基哌啶-4-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率41.2%;1H NMR(400MHz,DMSO-d6)δ9.06(s,1H),7.43(t,J=6.8Hz,1H),7.61(t,J=7.2Hz,1H),7.50-7.46(m,6H),7.27(t,J=7.6Hz,1H),7.24(t,J=54.4Hz,1H),6.59(s,1H),5.67-5.60(m,1H),5.24-5.16(m,1H),4.87-4.73(m,1H),3.91-3.76(m,1H),3.44-3.33(m,1H),3.10-2.97(m,1H),2.35(s,3H),2.18-1.87(m,4H),1.59(d,J=7.2Hz,3H);ESI-MS(m/z):536.0[M+H]+
实施例56
4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(奎宁环-3-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例49的步骤a-f,所得粗品经Prep-HPLC(分离方法3)纯化,得4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(奎宁环-3-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率10.4%;1H NMR(400MHz,DMSO-d6)δ9.03-8.91(m,1H),8.01-7.87(m,1H),7.62-7.53(m,1H),7.52-7.45(m,1H),7.37-7.12(m,2H),6.61-6.57(m,1H),5.74-5.60(m,1H),5.07-4.90(m,1H),3.50-3.36(m,1H),3.19-3.07(m,1H),2.86-2.65(m,4H),2.38-2.34(m,3H),2.15-2.03(m,1H),1.82-1.69(m,2H),1.61-1.55(m,3H),1.49-1.35(m,2H);ESI-MS(m/z):458.0[M+H]+
参考实施例56的制备方法,以相应的原料,制得了以下实施例中的化合物。
实施例60
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(2-甲氧基吡啶-4-基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例49的步骤a-f,所得粗品经Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(2-甲氧基吡啶-4-基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率25.5%;1H NMR(400MHz,DMSO-d6)δ9.22(s,1H),8.40(d,J=5.6Hz,1H),7.65(d,J=6.8Hz,1H),7.59(t,J=7.8Hz,1H),7.48(t,J=7.2Hz,1H),7.30-7.24(m,2H),7.23(t,J=56.0Hz,1H),7.18(s,1H),6.66(s,1H),5.66-5.99(m,1H),3.96(s,3H),2.38(s,3H),1.51(d,J=7.2Hz,3H);ESI-MS(m/z):456.0[M+H]+
实施例61
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-吗啉并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例49的步骤a-f,所得粗品经Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-吗啉并[3,4-d]哒嗪-7(6H)-酮,产率33.1%;1H NMR(400MHz,DMSO-d6)δ9.29(s,1H),7.82(d,J=6.8Hz,1H),7.61(t,J=7.2Hz,1H),7.48(t,J=6.8Hz,1H),7.26(t,J=7.6Hz,1H),7.24(t,J=54.4Hz,1H),6.59(s,1H),5.65-5.58(m,1H),3.79-3.49(m,8H),2.34(s,3H),1.54(d,J=7.2Hz,3H);ESI-MS(m/z):434.0[M+H]+
实施例62
(R)-3-(4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-基)-N,柞二甲基-3-氧代丙酰胺的制备
步骤a):(R)-3-(4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-基)-3-氧代丙酸甲酯的制备
将(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(哌啶-4-基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐(100mg,0.214mmol)、3-甲氧基-3-氧代丙酸(51mg,0.428mmol)、HATU(163mg,0.428mmol)、二氯甲烷(5mL)和三乙胺(86mg,0.855mmol)加入反应瓶中,室温搅拌反应2h。反应结束后,减压浓缩,残余物经硅胶柱层析(二氯甲烷/甲醇=15/1)纯化,得(R)-3-(4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-基)-3-氧代丙酸甲酯,产率81.2%;ESI-MS(m/z):532.2[M+H]+
步骤b):(R)-3-(4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-基)-3-氧代丙酸的制备
将(R)-3-(4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-基)-3-氧代丙酸甲酯(92mg,0.174mmol)、一水合氢氧化锂(35mg,0.871mmol)、甲醇(1mL)和水(1mL)加入反应瓶中,室温搅拌反应1h。用稀盐酸(1N)将反应液调至pH=2~3,用二氯甲烷萃取(10mL×3),合并有机相,用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得(R)-3-(4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-基)-3-氧代丙酸,产率97.2%;ESI-MS(m/z):518.2[M+H]+。
步骤c):(R)-3-(4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-基)-N,N-二甲基-3-氧代丙酰胺的制备
将(R)-3-(4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-基)-3-氧代丙酸(88mg,0.169mmol)、二甲胺盐酸盐(28mg,0.338mmol)、HATU(129mg,0.338mmol)、二氯甲烷(5mL)和三乙胺(51mg,0.507mmol)加入反应瓶中,室温搅拌反应2h。反应结束后,将反应液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-3-(4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌啶-1-基)-N,N-二甲基-3-氧代丙酰胺,产率33.1%;1H NMR(400MHz,DMSO-d6)δ9.06(s,1H),7.83(d,J=6.4Hz,1H),7.59(t,J=7.2Hz,1H),7.48(t,J=7.2Hz,1H),7.27(t,J=8.0Hz,1H),7.24(t,J=54.0Hz,1H),6.59(s,1H),5.66-5.59(m,1H),5.22-5.12(m,1H),4.67(d,J=13.2Hz,1H),4.02(d,J=13.6Hz,1H),3.72(d,J=16.0Hz,1H),3.53(d,J=16.0Hz,1H),3.26-3.19(m,1H),2.96(s,3H),2.84(s,3H),2.81-2.73(m,1H),2.35(s,3H),2.07-1.85(m,4H),1.57(d,J=6.8Hz,3H);ESI-MS(m/z):545.0[M+H]+
实施例63
6-(1-乙酰基吡咯烷-3-基)-4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(吡咯烷-3-基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐(120mg,0.264mmol)、三乙胺(80mg,0.793mmol)和二氯甲烷(3mL)加入反应瓶中,室温下滴加乙酰氯(21mg,0.264mmol),滴毕,维持室温搅拌反应30min。反应结束后,向反应液中加水(30mL)稀释,用二氯甲烷(20mL×3)萃取,合并有机相,以饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得6-(1-乙酰基吡咯烷-3-基)-4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率12.3%;1H NMR(400MHz,DMSO-d6)δ9.13-9.06(m,1H),7.72(s,1H),7.61(t,J=7.6Hz,1H),7.49(t,J=7.2Hz,1H),7.27(t,J=7.6Hz,1H),7.24(t,J=54.4Hz,1H),6.60(d,J=7.6Hz,1H),5.62(q,J=6.8Hz,1H),5.56-5.42(m,1H),4.07-3.91(m,1H),3.87-3.76(m,1H),3.67-3.57(m,1H),3.52-3.39(m,1H),2.48-2.40(m,2H),2.35(s,3H),1.99(d,J=13.2Hz,3H),1.57(d,J=7.2Hz,3H);ESI-MS(m/z):460.2[M+H]+
实施例64
4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基吡咯烷-3-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(吡咯烷-3-基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐(200mg,0.441mmol)、三乙胺(133mg,1.322mmol)和二氯甲烷(4mL)加入反应瓶中,室温下向反应液中滴加碘甲烷(63mg,0.441mmol),维持室温搅拌反应1h。向反应液中加入水(30mL)稀释,用二氯甲烷(20mL×3)萃取,合并有机相,以饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-甲基吡咯烷-3-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率10.0%;1H NMR(400MHz,DMSO-d6)δ9.06(s,1H),7.72(d,J=6.8Hz,1H),7.60(d,J=5.2Hz,1H),7.48(t,J=7.2Hz,1H),7.28(d,J=7.6Hz,1H),7.24(t,J=54.4Hz,1H),6.53(s,1H),5.65-5.59(m,1H),5.49-5.38(m,1H),3.09-3.02(m,1H),2.90-2.83(m,1H),2.78-2.72(m,1H),2.47-2.41(m,1H),2.36(d,J=6.8Hz,3H),2.34(s,3H),2.35-2.14(m,1H),2.12-2.04(m,1H),1.57(d,J=7.2Hz,3H);ESI-MS(m/z):432.2[M+H]+
实施例65
4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(异噁唑-4-羰基)吡咯烷-3-基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(吡咯烷-3-基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐(120mg,0.264mmol)、4-异噁唑甲酸(45mg,0.396mmol)、HATU(151mg,0.396mmol)、三乙胺(80mg,0.793mmol)和二氯甲烷(5mL)加入反应瓶中,室温搅拌反应30min。向反应液中加水(40mL)稀释,用二氯甲烷(30mL×3)萃取,合并有机相,以饱和食盐水(80mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-(异噁唑-4-羰基)吡咯烷-3-基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮;产率11.8%;1H NMR(400MHz,DMSO-d6)δ9.55-9.46(m,1H),9.22-9.10(m,1H),8.99-8.89(m,1H),7.73(s,1H),7.61(t,J=7.2Hz,1H),7.49(t,J=7.2Hz,1H),7.27(t,J=7.2Hz,1H),7.23(t,J=54.4Hz,1H),6.62(s,1H),5.73-5.51(m,2H),4.27-4.09(m,1H),4.07-3.94(m,1H),3.93-3.81(m,1H),3.78-3.60(m,1H),2.63-2.53(m,1H),2.47-2.40(m,1H),2.36(s,3H),1.57(d,J=7.2Hz,3H);ESI-MS(m/z):513.2[M+H]+
实施例66
4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-(吗啉-4-羰基)吡咯烷-3-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将4-((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(吡咯烷-3-基)吡啶并[3,4-d]哒嗪-7(6H)-酮盐酸盐(120mg,0.264mmol)、三乙胺(80mg,0.793mmol)和二氯甲烷(3mL)加入反应瓶中,室温下,向反应液中滴加吗啉-4-甲酰氯(79mg,0.264mmol),维持室温搅拌反应30min。向反应液中加入水(30mL),用二氯甲烷(20mL×3)萃取,合并有机相,以饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得4-(((R)-1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(1-(吗啉-4-羰基)吡咯烷-3-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率15.2%;1H NMR(400MHz,DMSO-d6)δ9.05(d,J=9.2Hz,1H),7.76(s,1H),7.59(t,J=7.6Hz,1H),7.50(t,J=7.2Hz,1H),7.26(t,J=7.2Hz,1H),7.26(t,J=54.4Hz,1H),6.60(s,1H),5.65-5.54(m,1H),5.47-5.38(m,1H),3.85-3.79(m,1H),3.623.53(m,8H),3.23-3.16(m,4H),2.35(s,3H),2.34-2.30(s,1H),1.57(t,J=6.0Hz,3H);ESI-MS(m/z):531.2[M+H]+
实施例67
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(哌嗪-1-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
步骤a):4-(4-羟基-5-(甲氧基羰基)-2-氧代吡啶-1(2H)-基)哌嗪-1-甲酸叔丁酯的制备。
将2-甲酰基-3-氧代戊二酸二甲酯(5.5g,0.027mol)、4-氨基哌嗪-1-甲酸叔丁酯(6.0g,0.030mol)和甲醇 (150mL)依次加入反应瓶中,室温搅拌下缓慢加入甲醇钠(2.2g,0.040mol),氮气保护下升温至80℃反应1.5h。反应结束后,将反应液冷却至室温,加入2M盐酸调至pH=4-5,减压浓缩,向残余物中加入二氯甲烷(25mL)和甲醇(25mL),室温搅拌20min,过滤,滤饼用甲醇(10mL)淋洗,滤液减压浓缩,得4-(4-羟基-5-(甲氧基羰基)-2-氧代吡啶-1(2H)-基)哌嗪-1-甲酸叔丁酯,产率41.9%;ESI-MS(m/z):354.2[M+H]+
步骤b):4-(5-(甲氧基羰基)-2-氧代-4-(((三氟甲基)磺酰基)氧)吡啶-1(2H)-基)哌嗪-1-甲酸叔丁酯的制备。
将4-(4-羟基-5-(甲氧基羰基)-2-氧代吡啶-1(2H)-基)哌嗪-1-甲酸叔丁酯(4.0g,0.011mol)溶解在1,2-二氯乙烷(100mL)中,冰水浴下加入吡啶(1.8g,0.023mol),缓慢滴加三氟甲磺酸酐(6.4g,0.023mol),滴毕,升至室温搅拌反应5min。反应结束后,加水(50mL)淬灭反应,用二氯甲烷萃取(50mL×3),合并有机相,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=3/1),得4-(5-(甲氧基羰基)-2-氧代-4-(((三氟甲基)磺酰基)氧)吡啶-1(2H)-基)哌嗪-1-甲酸叔丁酯,产率45.5%;ESI-MS(m/z):486.1[M+H]+
步骤c):4-(4-乙酰基-5-(甲氧基羰基)-2-氧代吡啶-1(2H)-基)哌嗪-1-甲酸叔丁酯的制备。
将4-(5-(甲氧基羰基)-2-氧代-4-(((三氟甲基)磺酰基)氧)吡啶-1(2H)-基)哌嗪-1-甲酸叔丁酯(2.5g,5.150mmol)、三丁基(1-乙氧基乙烯基)锡(2.2g,6.180mmol)、三乙胺(1.6g,0.015mol)、双三苯基膦二氯化钯(289mg,0.412mmol)和1,4-二氧六环(50mL)依次加入反应瓶中,氮气置换3次,升温至100℃反应2h。反应结束后,冷却至室温,加入乙酸乙酯(50mL)和20%氟化钾水溶液(50mL),室温搅拌10min,过滤,滤饼用乙酸乙酯淋洗(30mL×3),滤液静置分层,收集有机相,用20%氟化钾水溶液(40mL)和饱和食盐水(40mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,将残余物溶于乙酸乙酯(30mL)中,加入氯化氢的乙酸乙酯溶液(10mL,4M),室温搅拌反应1h。将反应液依次用饱和碳酸氢钠水溶液(50mL×2)和饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=7/3),得4-(4-乙酰基-5-(甲氧基羰基)-2-氧代吡啶-1(2H)-基)哌嗪-1-甲酸叔丁酯,产率51.2%;ESI-MS(m/z):380.2[M+H]+
步骤d):4-(1-甲基-4,7-二氧-3,7-二氢吡啶并[3,4-d]哒嗪-6(4H)-基)哌嗪-1-甲酸叔丁酯的制备。
将4-(4-乙酰基-5-(甲氧基羰基)-2-氧代吡啶-1(2H)-基)哌嗪-1-甲酸叔丁酯(1.0g,2.636mmol)和乙醇(20mL)加入反应瓶中,室温搅拌下加入水合肼(660mg,10.543mmol,80%),氮气保护下升温至90℃反应1h。反应结束后,冷却至室温,加入甲基叔丁基醚(20mL),室温搅拌10min,过滤,滤饼减压干燥,得4-(1-甲基-4,7-二氧-3,7-二氢吡啶并[3,4-d]哒嗪-6(4H)-基)哌嗪-1-甲酸叔丁酯,产率81.9%;ESI-MS(m/z):362.2[M+H]+
步骤e):4-(1-甲基-7-氧代-4-(1H-1,2,4-三氮唑-1-基)吡啶并[3,4-d]哒嗪-6(7H)-基)哌嗪-1-甲酸叔丁酯的制备。
将4-(1-甲基-4,7-二氧-3,7-二氢吡啶并[3,4-d]哒嗪-6(4H)-基)哌嗪-1-甲酸叔丁酯(780mg,2.158mmol)、1,2,4-三氮唑(1.2g,17.264mmol)和乙腈(30mL)依次加入反应瓶中,氮气置换3次,加入三乙胺(3.5g,34.528mmol)和三氯氧磷(827mg,5.395mmol)的乙腈(3mL)溶液,氮气保护下室温搅拌反应3h。反应结束后,缓慢加入水(20mL)淬灭反应,二氯甲烷萃取(30mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=97/3),得4-(1-甲基-7-氧代-4-(1H-1,2,4-三氮唑-1-基)吡啶并[3,4-d]哒嗪-6(7H)-基)哌嗪-1-甲酸叔丁酯,产率39.3%;ESI-MS(m/z):413.2[M+H]+
步骤f):(R)-4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌嗪-1-甲酸叔丁酯的制备。
将4-(1-甲基-7-氧代-4-(1H-1,2,4-三氮唑-1-基)吡啶并[3,4-d]哒嗪-6(7H)-基)哌嗪-1-甲酸叔丁酯(350mg,0.848mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺盐酸盐(249mg,1.102mmol)和1,4-二氧六环(10mL)依次加入微波反应瓶中,置于微波反应器中升温至115℃反应6h。将反应液冷却至室温,减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=19:1),得(R)-4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌嗪-1-甲酸叔丁酯,产率79.7%;ESI-MS(m/z):533.2[M+H]+
步骤g):(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(哌嗪-1-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备。
将(R)-4-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)哌嗪-1-甲酸叔丁酯(100mg,0.188mmol)和氯化氢的乙酸乙酯溶液(5mL,2M)加入反应瓶中,室温搅拌反应1h。将反应液减压浓缩,残余物经Prep-HPLC纯化(分离方法3),得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(哌嗪-1-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率17.8%;1H NMR(400MHz,DMSO-d6)δ9.27(s,1H),7.80(d,J=6.8Hz,1H), 7.63-7.59(m,1H),7.50-7.46(m,1H),7.37-7.10(m,2H),6.57(s,1H),5.65-5.58(m,1H),3.28(s,2H),3.18-3.15(m,1H),2.88(s,3H),2.79-2.66(m,2H),2.34(s,4H),1.54(d,J=6.8Hz,3H);ESI-MS(m/z):433.2[M+H]+
实施例68
(R)-6-(4-乙酰基哌嗪-1-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐的制备
将(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(哌嗪-1-基)吡啶并[3,4-d]哒嗪-7(6H)-酮(100mg,0.231mmol)、三乙胺(117mg,1.156mmol)和二氯甲烷(5mL)加入反应瓶中,冰水浴下缓慢加入乙酰氯(22mg,0.277mmol),氮气保护下室温搅拌反应10min。反应结束后,用饱和氯化铵水溶液(10mL)淬灭反应,二氯甲烷萃取(20mL×2),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC纯化(分离方法2),得(R)-6-(4-乙酰基哌嗪-1-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐,产率30.1%;1H NMR(400MHz,DMSO-d6)δ9.32(s,1H),8.13(s,1H),7.90(s,1H),7.64-7.60(m,1H),7.51-7.48(m,1H),7.38-7.11(m,2H),6.62(s,1H),5.64-5.57(m,1H),3.55-3.46(m,2H),3.33(s,6H),2.35(s,3H),2.08(s,3H),1.55(d,J=7.2Hz,3H);ESI-MS(m/z):475.2[M+H]+
实施例69
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(4-(吗啉-4-羰基)哌嗪-1-基)吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐的制备
将(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(哌嗪-1-基)吡啶并[3,4-d]哒嗪-7(6H)-酮(100mg,0.231mmol)、三乙胺(117mg,1.156mmol)和二氯甲烷(5mL)加入反应瓶中,室温搅拌下加入吗啉-4-甲酰氯(42mg,0.277mmol),氮气保护下室温搅拌反应10min。反应结束后,用饱和氯化铵水溶液(10mL)淬灭反应,二氯甲烷萃取(20mL×2),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC纯化(分离方法2),得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(4-(吗啉-4-羰基)哌嗪-1-基)吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐,产率29.5%;1H NMR(400MHz,DMSO-d6)δ13.77(s,1H),9.48(s,1H),9.08(s,2H),7.68-7.64(m,1H),7.55-7.52(m,1H),7.38-7.11(m,2H),6.72(s,1H),5.62-5.55(m,1H),3.77(t,J=4.8Hz,4H),3.59(t,J=4.8Hz,4H),3.19(t,J=4.8Hz,4H),3.08(t,J=4.8Hz,4H),2.38(s,3H),1.59(d,J=6.8Hz,3H);ESI-MS(m/z):546.2[M+H]+
实施例70
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(嘧啶-5-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例67的步骤a-f,经Prep-HPLC(分离方法3)纯化,得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-6-(嘧啶-5-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率15.5%;1H NMR(400MHz,DMSO-d6)δ9.38(s,1H),9.31(s,1H),9.18(s,2H),7.64-7.56(m,2H),7.49(t,J=7.2Hz,1H),7.27(t,J=7.2Hz,1H),7.24(t,J=56.0Hz,1H),6.73(s,1H),5.67-5.60(m,1H),2.40(s,3H),1.52(d,J=7.2Hz,3H);ESI-MS(m/z):427.0[M+H]+
实施例71
(R)-6-(金刚烷-1-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
步骤a-e):6-(金刚烷-1-基)-1-甲基-4-(1H-1,2,4-三唑-1-基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例67的步骤a-e,得6-(金刚烷-1-基)-1-甲基-4-(1H-1,2,4-三唑-1-基)吡啶并[3,4-d]哒嗪-7(6H)-酮,ESI-MS(m/z):363.2[M+H]+
步骤f):(R)-6-(金刚烷-1-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将6-(金刚烷-1-基)-1-甲基-4-(1H-1,2,4-三唑-1-基)吡啶并[3,4-d]哒嗪-7(6H)-酮(309mg,0.853mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺盐酸盐(290mg,1.285mmol)和1,4-二氧六环(6mL)加入微波反应瓶中,于微波仪中升温至110℃反应5h。反应结束后,减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-6-(金刚烷-1-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率4.9%;1H NMR(400MHz,DMSO-d6)δ8.81(s,1H),7.83(s,1H),7.63(t,J=7.2Hz,1H),7.51-7.47(m,1H),7.38-7.11(m,2H),6.46(s,1H),5.67-5.60(m,1H),3.32(s,3H),2.42-2.20(m,9H),1.80-1.71(m,6H),1.57(d,J=7.3Hz,3H);ESI-MS(m/z):483.0[M+H]+
实施例72
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(2-(二甲氨基)乙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐的制备
步骤a):4-羟基-1-(2-羟基乙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备。
将2-甲酰基-3-氧代戊二酸二甲酯(5.5g,0.027mol)、2-氨基乙醇(1.8g,0.030mol)和甲醇(120mL)依次加入反应瓶中,室温搅拌下缓慢加入甲醇钠(2.2g,0.040mol),氮气保护下升温至80℃反应2h。反应结束后,冷却至室温,用浓盐酸调至pH=4-5,减压浓缩,向残余物中加入二氯甲烷(50mL),室温搅拌20min,过滤,滤饼用二氯甲烷(10mL)淋洗,滤液减压浓缩,得4-羟基-1-(2-羟基乙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯,产率78.2%;ESI-MS(m/z):214.1[M+H]+
步骤b):4-(苯甲酰氧基)-1-(2-(苯甲酰氧基)乙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备。
将4-羟基-1-(2-羟基乙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(4.5g,0.021mol)、三乙胺(10.6g,0.105mol)和二氯甲烷(100mL)加入反应瓶中,室温搅拌下加入苯甲酰氯(8.8g,0.063mol),氮气保护下室温搅拌反应10min。反应结束后,用饱和氯化铵水溶液(30mL)淬灭反应,二氯甲烷萃取(50mL×3),合并有机相,以饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=3/1),得4-(苯甲酰氧基)-1-(2-(苯甲酰氧基)乙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯,产率92.6%;ESI-MS(m/z):422.1[M+H]+
步骤c):1-(2-(苯甲酰氧基)乙基)-4-羟基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备。
将4-(苯甲酰氧基)-1-(2-(苯甲酰氧基)乙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(8.2g,0.019mol)和甲醇(200mL)加入反应瓶中,冰水浴下加入碳酸钾(4.0g,0.029mol),氮气保护下0℃反应2h。反应结束后,用2M的稀盐酸(30mL)淬灭,二氯甲烷萃取(80mL×3),合并有机相,以饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得1-(2-(苯甲酰氧基)乙基)-4-羟基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯,产率73.0%;ESI-MS(m/z):318.1[M+H]+
步骤d):1-(2-(苯甲酰氧基)乙基)-6-氧代-4-(((三氟甲基)磺酰基)氧)-1,6-二氢吡啶-3-甲酸甲酯的制备。
将1-(2-(苯甲酰氧基)乙基)-4-羟基-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(4.4g,0.014mol)溶解在1,2-二氯乙烷(100mL)中,冰水浴下加入吡啶(2.2g,0.028mol),缓慢滴加三氟甲磺酸酐(7.8g,0.028mol),加毕,升至室温搅拌反应5min,加水(50mL)淬灭反应,用二氯甲烷萃取(50mL×3),合并有机相,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=2/1),得1-(2-(苯甲酰氧基)乙基)-6-氧代-4-(((三氟甲基)磺酰基)氧)-1,6-二氢吡啶-3-甲酸甲酯,产率63.6%;ESI-MS(m/z):450.0[M+H]+
步骤e):4-乙酰基-1-(2-(苯甲酰氧基)乙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯的制备。
将1-(2-(苯甲酰氧基)乙基)-6-氧代-4-(((三氟甲基)磺酰基)氧)-1,6-二氢吡啶-3-甲酸甲酯(4.0g,8.902mmol)、三丁基(1-乙氧基乙烯基)锡(3.8g,10.682mmol)、三乙胺(2.7g,0.027mol),双三苯基膦二氯化钯(312mg,0.445mmol)和1,4-二氧六环(80mL)加入反应瓶中,氮气置换3次后升温至100℃反应2h。反应结束后,冷却至室温,加入乙酸乙酯(50mL)和20%氟化钾水溶液(50mL),室温搅拌10min,过滤,滤饼用乙酸乙酯淋洗(30mL×3),滤液静置分层,收集有机相,用20%氟化钾水溶液(40mL)和饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,向残余物中加入乙酸乙酯溶解(30mL),室温搅拌下加入氯化氢的乙酸乙酯溶液(15mL,4M),室温反应1h。将反应液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=1/1),得 4-乙酰基-1-(2-(苯甲酰氧基)乙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯,产率62.5%;ESI-MS(m/z):344.1[M+H]+
步骤f):2-(1-甲基-4,7-二氧代-3,7-二氢吡啶并[3,4-d]哒嗪-6(4H)-基)苯甲酸乙酯的制备
将4-乙酰基-1-(2-(苯甲酰氧基)乙基)-6-氧代-1,6-二氢吡啶-3-甲酸甲酯(1.9g,5.564mmol)和乙醇(40mL)加入反应瓶中,室温下加入水合肼(1.1g,0.022mol,80%),氮气保护下升温至90℃反应10min。反应结束后,冷却至室温,加入甲基叔丁基醚(40mL),室温搅拌10min,过滤,用甲基叔丁基醚淋洗(10mL×2),滤饼真空干燥,得2-(1-甲基-4,7-二氧代-3,7-二氢吡啶并[3,4-d]哒嗪-6(4H)-基)苯甲酸乙酯,产率83.9%;ESI-MS(m/z):326.1[M+H]+
步骤g):2-(4-溴-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)苯甲酸乙酯的制备。
将2-(1-甲基-4,7-二氧代-3,7-二氢吡啶并[3,4-d]哒嗪-6(4H)-基)苯甲酸乙酯(1.5g,4.668mmol)、三溴氧磷(2.7g,9.336mmol)和乙腈(40mL)加入反应瓶中,氮气保护下升温至105℃反应1h。将反应液冷却至室温,用饱和碳酸氢钠水溶液(40mL)淬灭反应,用二氯甲烷萃取(40mL×3),合并有机相,以饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=3/7),得2-(4-溴-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)苯甲酸乙酯,产率55.9%;ESI-MS(m/z):388.0[M+H]+
步骤h):(R)-2-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)苯甲酸乙酯的制备。
将2-(4-溴-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)苯甲酸乙酯(1.0g,2.609mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺盐酸盐(706mg,3.131mmol)、N,N-二异丙基乙胺(1.0g,7.827mmol)和1,4-二氧六环(15mL)加入微波反应瓶中,置于微波反应器中升温至100℃反应3h。将反应液冷却至室温,减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=19/1),得(R)-2-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)苯甲酸乙酯,产率64.8%;ESI-MS(m/z):497.2[M+H]+
步骤i):(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(2-羟基乙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备。
将(R)-2-(4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基-7-氧代吡啶并[3,4-d]哒嗪-6(7H)-基)苯甲酸乙酯(830mg,1.672mmol)、甲醇(10mL)和四氢呋喃(10mL)加入反应瓶中,室温搅拌下加入氢氧化锂水溶液(2.5mL,3M),室温搅拌反应20min。反应结束后,用1M稀盐酸调至pH=3-4,减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=19/1),得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(2-羟基乙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率88.1%;ESI-MS(m/z):393.1[M+H]+
步骤j):(R)-6-(2-氯乙基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备。
将(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(2-羟基乙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮(570mg,1.453mmol)和二氯甲烷(15mL)依次加入反应瓶中,室温搅拌下加入二氯亚砜(691mg,5.811mmol),氮气保护下室温搅拌反应4h。将反应液减压浓缩,向残余物中加入饱和碳酸氢钠水溶液(30mL)和二氯甲烷(30mL),室温搅拌5min,静置分层,收集有机相,水相用二氯甲烷萃取(20mL×2),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=97:3),得(R)-6-(2-氯乙基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率37.2%;ESI-MS(m/z):411.1[M+H]+
步骤k):(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(2-(二甲氨基)乙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐的制备。
将(R)-6-(2-氯乙基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮(100mg,0.243mmol)、N,N-二异丙基乙胺(315mg,2.434mmol)和四氢呋喃(5mL)加入玻璃恒容反应瓶中,室温搅拌下加入二甲胺四氢呋喃溶液(1.3mL,2M),升温至80℃搅拌反应过夜。反应结束后,冷却至室温,减压浓缩,残余物经Prep-HPLC纯化(分离方法2),得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(2-(二甲氨基)乙基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮甲酸盐,产率13.5%;1H NMR(400MHz,DMSO-d6)δ9.30(s,1H),8.13(s,1H),7.65-7.61(m,2H),7.51-7.47(m,1H),7.37-7.10(m,2H),6.55(s,1H),5.65-5.60(m,1H),4.35-4.22(m,2H),3.31(s,3H),2.95-2.80(m,2H),2.43-2.41(m,3H),2.35(s,3H),1.56(d,J=6.8Hz,3H);ESI-MS(m/z):420.2[M+H]+
实施例73
(R)-6-(1-乙酰氮杂环丁烷-3-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例68,所得粗品经Prep-HPLC(分离方法3)纯化,得(R)-6-(1-乙酰氮杂环丁烷-3-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮;产率12.4%;1H NMR(400MHz,DMSO-d6)δ9.11(d,J=7.6Hz,1H),7.85-7.78(m,1H),7.61(t,J=7.6Hz,1H),7.49(t,J=7.2Hz,1H),7.27(t,J=7.6Hz,1H),7.24(t,J=54.4Hz,1H),6.57(s,1H),5.65(p,J=6.4Hz,1H),5.39-5.29(m,1H),4.62(t,J=8.8Hz,1H),4.55-4.49(m,1H),4.43-4.39(m,1H),4.32(t,J=10.0Hz,1H),2.36(s,3H),1.83(s,3H),1.57(d,J=7.2Hz,3H);ESI-MS(m/z):446.2[M+H]+
实施例74
(R)-6-(氮杂环丁烷-3-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮三氟乙酸盐的制备
制备方法参考实施例67,经Prep-HPLC纯化(分离方法4)得(R)-6-(氮杂环丁烷-3-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮三氟乙酸盐;产率21.7%;1H NMR(400MHz,DMSO-d6)δ13.88(s,1H),9.33(s,1H),9.10(s,2H),7.59-7.68(m,2H),7.39-7.32(m,1H),7.24(t,J=54.0Hz,1H),6.93(s,1H),5.58(p,J=6.8Hz,1H),5.29(p,J=8.0Hz,1H),4.62-4.52(m,2H),4.42-4.33(m,2H),2.47(s,3H),1.67(d,J=6.7Hz,3H);ESI-MS(m/z):404.2[M+H]+
实施例75
(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-乙烯基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-1-氯-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮(250mg,0.613mmol)、乙烯三氟硼酸钾(164mg,1.226mmol)、碳酸铯(398mg,1.226mmol)、1,1'-双二苯基膦二茂铁二氯化钯(45mg,0.061mmol)、1,4-二氧六环(10mL)和水(2mL)依次加入到反应瓶中,氮气置换三次,升温至80℃搅拌反应2h。反应结束后,将反应液冷却至室温,加饱和氯化铵水溶液(20mL)淬灭反应,以二氯甲烷萃取(20mL×3),合并有机相,饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-乙烯基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率17.5%;1H NMR(400MHz,DMSO-d6)δ9.07(s,1H),8.00(d,J=6.8Hz,1H),7.62(t,J=7.6Hz,1H),7.49(t,J=7.2Hz,1H),7.27(t,J=7.2Hz,1H),7.25(t,J=52.0Hz,1H),7.13-7.09(m,1H),6.74(s,1H),6.09-6.04(m,1H),5.75-5.68(m,1H),5.38-5.55 (m,1H),3.67-3.61(m,1H),1.58(d,J=7.2Hz,3H),1.18-1.12(m,4H);ESI-MS(m/z):401.0[M+H]+
实施例76
(R)-1-(环戊-1-烯-1-基)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-1-氯-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮(250mg,0.613mmol)、2-(环戊-1-烯-1-基)-4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷(238mg,1.226mmol)、碳酸铯(398mg,1.226mmol)、1,1'-双二苯基膦二茂铁二氯化钯(45mg,0.061mmol)、1,4-二氧六环(10mL)和水(2mL)依次加入到反应瓶中,氮气置换三次,升温至80℃搅拌反应2h。将反应液冷却至室温,加水饱和氯化铵水溶液(20mL)淬灭反应,二氯甲烷萃取(20mL×3),合并有机相,饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-1-(环戊-1-烯-1-基)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率28.5%;1H NMR(400MHz,DMSO-d6)δ9.04(s,1H),7.92(d,J=6.8Hz,1H),7.62(t,J=7.6Hz,1H),7.49(t,J=7.2Hz,1H),7.27(t,J=7.6Hz,1H),7.24(t,J=52.0Hz,1H),6.68(s,1H),6.16(s,1H),5.73-5.66(m,1H),3.67-3.61(m,1H),2.68(d,J=7.8Hz,2H),2.56(d,J=7.8Hz,2H),1.92-1.85(m,2H),1.57(d,J=7.2Hz,3H),1.20-1.10(m,4H);ESI-MS(m/z):441.0[M+H]+
实施例77
(R)-1-(环戊-1-烯-1-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
制备方法参考实施例76,残余物物经Prep-HPLC(分离方法3)纯化,得(R)-1-(环戊-1-烯-1-基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率5.8%;1H NMR(400MHz,DMSO-d6)δ9.29(s,1H),7.93(d,J=6.8Hz,1H),7.63(t,J=7.6Hz,1H),7.49(t,J=6.8Hz,1H),7.38-7.11(m,2H),6.65(s,1H),6.15(s,1H),5.72-5.63(m,1H),2.72-2.64(m,2H),2.59-2.52(m,2H),1.92-1.85(m,2H),1.60-1.54(m,6H),1.21-1.07(m,4H);ESI-MS(m/z):455.0[M+H]+
实施例78
(R)-1-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-1-氯-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(100mg,0.237mmol)、环丙基硼酸(41mg,0.474mmol)、碳酸铯(154mg,0.474mmol)、1,1'-双二苯基膦二茂铁二氯化钯(17mg,0.024mmol)、甲苯(3mL)和水(1mL)依次加入反应瓶中,氮气置换三次,升温至100℃搅拌反应3h。 反应结束后,将反应液冷却至室温,加饱和氯化铵水溶液(20mL)淬灭反应,以二氯甲烷萃取(20mL×3)。合并有机相,以饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法2)纯化,得(R)-1-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率18.5%;1H NMR(400MHz,DMSO-d6)δ9.27(s,1H),7.74(d,J=6.8Hz,1H),7.62(t,J=7.6Hz,1H),7.48(t,J=7.6Hz,1H),7.26(t,J=7.6Hz,1H),7.24(t,J=52.0Hz,1H),6.79(s,1H),5.62-5.99(m,1H),2.21-2.24(m,1H),1.55(s,3H),1.54(s,3H),1.22-1.14(m,2H),1.11-1.03(m,2H),0.86-0.77(s,4H);ESI-MS(m/z):429.0[M+H]+
参考实施例78的制备方法,以相应的原料,制得了以下实施例中的化合物。
实施例80
(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲氧基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-1-氯-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮(65mg,0.245mmol)、甲醇钠(40mg,0.734mmol)和甲醇加入到反应瓶中,升温至80℃搅拌反应2h。反应结束后,冷却至室温,加入二氯甲烷(30mL)稀释,倒入饱和氯化铵水溶液中(50mL)淬灭,以二氯甲烷萃取(30mL×2),合并有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲氧基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率32.3%;1H NMR(400MHz,DMSO-d6)δ8.98(s,1H),7.60(t,J=7.6Hz,1H),7.48(t,J=6.8Hz,2H),7.27(d,J=7.6Hz,1H),7.24(s,J=53.6Hz,1H),6.51(s,1H),5.52(p,J=7.2Hz,1H),3.84(s,3H),3.65-3.59(m,1H),1.54(d,J=6.9Hz,3H),1.19-1.10(m,4H);ESI-MS(m/z):405.2[M+H]+
实施例81
(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-(甲硫基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-1-氯-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮(100mg,0.245mmol)、甲硫醇钠(25.8mg,0.368mmol)和甲醇(4mL)加入反应瓶中,室温下反应4h。加入饱和氯化铵水溶液(10mL)淬灭,用乙酸乙酯(20mL)萃取,有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-(甲硫基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率27.6%;1H NMR(400MHz,DMSO-d6)δ9.07(s,1H),7.78-7.76(d,J=8.0Hz,1H),7.65- 7.60(m,1H),7.51-7.48(m,1H),7.38-7.11(m,2H),6.38(s,1H),5.64-5.57(m,1H),3.65-3.59(m,1H),2.46(s,3H),1.57-1.55(d,J=8.0Hz,3H),1.17-1.10(m,4H);ESI-MS(m/z):421.0[M+H]+
实施例82
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-腈的制备
将(R)-1-氯-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)吡啶并[3,4-d]哒嗪-7(6H)-酮(440mg,1.043mmol)、氰化锌(73mg,0.626mmol)、三(二亚苄基丙酮)二钯(95mg,0.104mmol)、1,1'-双二苯基膦二茂铁二氯化钯(76mg,0.104mmol)和DMF(5mL)加入反应瓶中,氮气置换三次,升温至100℃搅拌反应3h。将反应液冷却至室温,加水(50mL)淬灭反应,以二氯甲烷萃取(20mL×3),合并有机相,以饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得((R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲腈,产率38.5%;1H NMR(400MHz,DMSO-d6)δ9.52(s,1H),8.91(d,J=6.8Hz,1H),7.67(t,J=7.6Hz,1H),7.53(t,J=7.2Hz,1H),7.31(t,J=7.6Hz,1H),7.25(t,J=56.0Hz,1H),6.28(s,1H),5.87-5.80(m,1H),1.63(d,J=7.2Hz,3H),1.55(s,3H),1.24-1.14(m,2H),1.14-1.06(m,2H);ESI-MS(m/z):414.0[M+H]+
参考实施例82的制备方法,以相应的原料,制得了以下实施例中的化合物。
实施例84
(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲醛的制备
将(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-1-甲基吡啶并[3,4-d]哒嗪-7(6H)-酮(100mg,0.257mmol)、二氧化硒(86mg,0.772mmol)和1,4-二氧六环(5mL)加入反应瓶中,氮气置换3次,升温至回流反应30min。反应结束后,冷却至室温,用饱和碳酸氢钠水溶液(20mL)淬灭反应,以二氯甲烷萃取(20mL×2),合并有机相,以饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC纯化(分离方法3),得(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲醛,产率19.2%;1H NMR(400MHz,DMSO-d6)δ9.83(s,1H),9.22(s,1H),8.96(d,J=6.8Hz,1H),7.70-7.66(m,1H),7.55-7.51(m,1H),7.47(s,1H),7.39-7.12(m,2H),5.99-5.92(m,1H),3.68-3.62(m,1H),1.65(d,J=7.2Hz,3H),1.20-1.10(m,4H);ESI-MS(m/z):403.1[M+H]+
实施例85
(R)-6-环丙基-1-(二氟甲基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲醛(100mg,0.248mmol)和二氯甲烷(5mL)加入反应瓶中,冰水浴下滴加二乙氨基三氟化硫(84mg,0.522mmol),氮气保护下维持0℃反应30min。反应结束后,用饱和碳酸氢钠水溶液(20mL)淬灭反应,以二氯甲烷萃取(20mL×2),合并有机相,以饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC纯化(分离方法3),得(R)-6-环丙基-1-(二氟甲基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率11.2%;1H NMR(400MHz,DMSO-d6)δ9.22(s,1H),8.51(s,1H),7.67-7.63(m,1H),7.53-7.50(m,1H),7.38-7.11(m,2H),7.05-6.78(m,1H),6.56(s,1H),5.79-5.72(m,1H),3.67-3.61(m,1H),1.60(d,J=7.2Hz,3H),1.21-1.13(m,4H);ESI-MS(m/z):425.1[M+H]+
实施例86
(R)-1-氯-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
步骤a):1,4-二氯-6-环丙基吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将6-环丙基-2,3-二氢吡啶并[3,4-d]哒嗪-1,4,7(6H)-三酮(8.8g,68.493mmol)和乙腈(500mL)加入反应瓶中,室温搅拌下加入DIPEA(26.5g,205.479mmol)和三氯氧磷(31.5g,205.479mmol),氮气保护下升温至100℃反应3h。反应结束后,冷却至室温,将反应液缓慢倒入冰块与碳酸氢钠的混合液中淬灭,二氯甲烷萃取(100mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=49/1),得1,4-二氯-6-环丙基吡啶并[3,4-d]哒嗪-7(6H)-酮,产率5.6%;ESI-MS(m/z):256.0[M+H]+
步骤b):(R)-1-氯-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮的制备
将1,4-二氯-6-环丙基吡啶并[3,4-d]哒嗪-7(6H)-酮(500mg,1.953mmol)、(R)-1-(3-(二氟甲基)-2-氟苯基)乙烷-1-胺盐酸盐(439mg,1.953mmol)和1,4-二氧六环(10mL)加入微波反应瓶中,于微波仪中升温至120℃反应2h。反应结束后,冷却至室温,减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-1-氯-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮,产率26.6%;1H NMR(400MHz,DMSO-d6)δ9.14(s,1H),8.14(d,J=6.8Hz,1H),7.63(t,J=7.6Hz,1H),7.50(t,J=7.2Hz,1H),7.28(t,J=7.6Hz,1H),7.24(t,J=56.0Hz,1H),6.51(s,1H),5.66-5.59(m,1H),3.66-3.60(m,1H),1.58(d,J=7.2Hz,3H),1.20-1.09(m,4H);ESI-MS(m/z):409.0[M+H]+
参考实施例86的制备方法,以相应的原料,制得了以下实施例中的化合物。
实施例88
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酸甲酯的制备
将(R)-1-氯-6-(1-甲基环丙基)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)吡啶并[3,4-d]哒嗪-7(6H)-酮(100mg,0.236mmol)、三乙胺(96mg,0.946mmol)、甲醇(3mL)和1,1'-双二苯基膦二茂铁二氯化钯(18mg,0.024mmol)加入反应瓶中,在室温搅拌下持续鼓入一氧化碳气体反应2h。反应结束后,减压浓缩,残余物用Prep-HPLC纯化(分离方法3),得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酸甲酯,产率34.1%;1H NMR(400MHz,DMSO-d6)δ9.41(s,1H),8.95(d,J=6.8Hz,1H),7.65(t,J=7.6Hz,1H),7.51(t,J=8.2Hz,1H),7.29(t,J=7.6Hz,1H),7.25(t,J=54.4Hz,1H),7.12(s,1H),5.84-5.78(m,1H),3.81(s,3H),1.62(d,J=6.8Hz,3H),1.55(s,3H),1.23-1.15(m,2H),1.12-1.06(m,2H);ESI-MS(m/z):447.0[M+H]+
实施例89
(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酸的制备
将(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酸甲酯(100mg,0.224mmol)、氢氧化锂(27mg,1.120mmol)、甲醇(2mL)和水(2mL)加入反应瓶中,室温反应1h。反应结束后,减压浓缩,残余物用Prep-HPLC纯化(分离方法3),得(R)-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-6-(1-甲基环丙基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酸,产率44.5%;1H NMR(400MHz,DMSO-d6)δ12.83(s,1H),9.41(s,1H),8.59(s,1H),7.66(t,J=7.6Hz,1H),7.51(t,J=7.2Hz,1H),7.31(s,1H),7.29(t,J=7.6Hz,1H),7.24(t,J=54.4Hz,1H),5.83-5.76(m,1H),1.62(d,J=6.8Hz,3H),1.56(s,3H),1.23-1.08(m,4H);ESI-MS(m/z):433.0[M+H]+
实施例90
(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酸的制备
将(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲醛(100mg,0.248mmol)、四氢呋喃(4mL)、叔丁醇(2mL)和水(2mL)加入反应瓶中,室温搅拌下依次加入亚氯酸钠(112mg,1.240mmol)和磷酸二氢钾(169mg,1.240mmol),氮气保护下室温搅拌反应1h。反应结束后,加入饱和食盐水(10mL),用二氯甲烷萃取(20mL×3),合并有机相,以饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC纯化(分离方法2),得(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酸,产率10.5%;1H NMR(400MHz,DMSO-d6)δ12.83(s,1H),9.17(s,1H),8.61(d,J=6.8Hz,1H),7.67-7.63(m,1H),7.53-7.49(m,1H),7.38-7.11(m,3H),5.85-5.78(m,1H),3.68-3.62(m,1H), 1.62(d,J=7.2Hz,3H),1.21-1.13(m,4H);ESI-MS(m/z):419.1[M+H]+
实施例91
(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酸甲酯的制备
将(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酸(120mg,0.287mmol)、HATU(164mg,0.431mmol)、三乙胺(58mg,0.574mmol)、甲醇(92mg,2.870mmol)和二氯甲烷(5mL)加入到反应瓶中,室温反应2h。反应结束后,加饱和氯化铵水溶液(40mL)淬灭反应,用二氯甲烷(30mL×3)萃取,合并有机相,以饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酸甲酯;产率18.5%;1H NMR(400MHz,DMSO-d6)δ9.16(s,1H),8.60(d,J=6.8Hz,1H),7.64(t,J=7.6Hz,1H),7.51(t,J=7.2Hz,1H),7.29(t,J=7.6Hz,1H),7.24(t,J=54.4Hz,1H),7.15(s,1H),5.81(p,J=7.2Hz,1H),3.81(s,3H),3.70-3.59(m,1H),1.61(d,J=7.2Hz,3H),1.21-1.08(m,4H);ESI-MS(m/z):433.1[M+H]+
实施例92
(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酚胺的制备
将(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酸甲酯(150mg,0.347mmol)和氨的甲醇溶液(6mL,7M)加入恒容反应瓶中,室温搅拌过夜。反应结束后,将反应液减压浓缩,残余物经Prep-HPLC(分离方法3)纯化,得(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酰胺;产率22.8%;1H NMR(400MHz,DMSO-d6)δ9.12(s,1H),8.40(d,J=6.8Hz,1H),8.00(s,1H),7.68-7.60(m,2H),7.50(t,J=7.2Hz,1H),7.31-7.25(m,2H),7.24(s,J=54.4Hz,1H),5.77(p,J=7.2Hz,1H),3.68-3.61(m,1H),1.60(d,J=7.2Hz,3H),1.19-1.10(m,4H);ESI-MS(m/z):418.2[M+H]+
实施例93
(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-N,柞二甲基-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酰胺的制备
将(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酸(100mg,0.239mmol)、HATU(136mg,0.359mmol)、三乙胺(73mg,0.717mmol)、二甲胺的四氢呋喃溶液(358μL,2M)和二氯甲烷(8mL)加入到反应瓶中,室温反应2h。反应结束后,加饱和氯化铵水溶液(40mL)淬灭反应,用二氯甲烷(30mL×3)萃取,合并有机相,以饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩, 残余物经Prep-HPLC(分离方法3)得(R)-6-环丙基-4-((1-(3-(二氟甲基)-2-氟苯基)乙基)氨基)-N,N-二甲基-7-氧代-6,7-二氢吡啶并[3,4-d]哒嗪-1-甲酰胺;产率31.0%;1H NMR(400MHz,DMSO-d6)δ9.15(s,1H),8.15(d,J=6.8Hz,1H),7.65(t,J=7.6Hz,1H),7.50(t,J=7.2Hz,1H),7.29(t,J=7.6Hz,1H),7.24(t,J=54.4Hz,1H),6.22(s,1H),5.74(p,J=7.2Hz,1H),3.68-3.60(m,1H),3.01(s,3H),2.81(s,3H),1.59(d,J=7.0Hz,3H),1.21-1.09(m,4H);ESI-MS(m/z):446.2[M+H]+
参考实施例93的制备方法,以相应的原料,制得了以下实施例中的化合物。
生物活性测试
试验例1:SOS1抑制活性测定
通过均相时间分辨荧光技术(HTRF)检测SOS1抑制剂对SOS1与KRAS蛋白之间相互作用的影响,从而评估其对SOS1蛋白的抑制水平。所用蛋白和检测试剂采用KRAS-G12D/SOS1 BINDING ASSAY KIT(Cisbio),首先将2mM受试化合物母液用Diluent试剂稀释20倍(100μM),再用Diluent试剂(5%DMSO)逐次进行5倍浓度梯度稀释,得到受试化合物工作液共8个浓度。在384孔板中,每孔依次加入4μL tag2-KRASG12D蛋白(含50μM GTP)、2μL受试化合物、4μL tag1-SOS1蛋白,双复孔,室温孵育15min。每孔依次加入Anti-tag1-Tb3+工作液和Anti-tag2-XL665工作液各5μL,在4℃孵育3h。将384孔板置于多功能酶标仪上读值,设置激发光波长为337nm,记录620nm和665nm的读值。数据结果以每孔665nm信号值与620nm信号值的比值呈现,即:Ratio=104×665nm信号值/620nm信号值。通过Ratio值计算抑制率:
%抑制率=[(Ratio阴性-Ratio化合物)/(Ratio阴性-RatioBlank)]×100
注:阴性为不加抑制剂组;Blank为不加酶组。
IC50通过抑制率由GraphPad Prism软件进行计算。每个化合物每次测定2个复孔。本发明化合物抑制SOS1活性的IC50值为3.7~308.0nM,部分实施例化合物测试数据如表1所示:
表1



从以上数据可以看出,本发明化合物在体外对SOS1具有明显的抑制作用,可用作SOS1抑制剂,在由SOS1蛋白介导的癌症、病原性皮疹病等疾病等领域具有广阔的应用前景。
试验例2:肝微粒体代谢稳定性测定
代谢稳定性影响着化合物在机体内的清除率、半衰期和口服生物利用度,是化合物最重要的ADME成药性质之一。肝微粒体稳定性测定是研究代谢稳定性的常用方法。本发明采用大鼠肝微粒体测定来考察化合物的代谢稳定性。
试验方法
a、溶液配制:
化合物储备液配制:取受试化合物和睾酮适量,用DMSO配制成1mM储备液,4℃冰箱中保存。
化合物工作液配制:分别取1mM的化合物储备液,然后用乙腈-水(v:v=1:1)稀释至100μM,即得。
NADPH工作液配制:称取适量的NADPH,用16mM的MgCl2配制成浓度为4.0mM的NADPH工作液。
大鼠肝微粒体工作液配制:取大鼠肝微粒体适量,用PBS稀释成1mg/ml的肝微粒体工作液。
b、样品孵育:
在孵育体系中依次加入48μL PBS,100μL大鼠肝微粒体工作液,2μL受试化合物工作液,混合均匀;在37℃预孵5min后,加入50μL NADPH启动反应。孵育相应时间点后,加入适量含内标的冰乙腈终止反应,涡旋混匀,离心取上清,进样LC-MS/MS检测化合物的代谢剩余量。
c、数据处理:
采用Excel等软件对T1/2和CL(liver)按以下公式进行数据处理。
默认fu(血液中的游离分数)等于1。
测试本发明化合物的T1/2为9.8~129min,CL(liver)为14.3~45.4ml/min/kg,部分实施例中的化合物对大鼠肝微粒体稳定性测试结果如表2所示:
表2

从表2数据可知,本发明部分实施例化合物与化合物MRTX0902相比,具有更好的代谢稳定性,具有很好的成药性质。MRTX0902为WO2021127429公开的化合物(实施例12-10),已被报道具有SOS1抑制活性。
试验例3:血浆稳定性试验
除了化合物的肝代谢稳定性,化合物的血浆稳定性也是新药研发的一个重要的影响因素。良好的血浆稳定性是具有良好的药动学和药效学的重要保障。本发明将实施例中的化合物分别与大鼠、小鼠、猴、犬、人的血浆在37℃条件下孵育,通过测定不同孵育时间点的样品浓度,计算相应的剩余百分比,以此来评价化合物在5种属血浆中的稳定性。
试验方法
a、溶液配制
化合物中间浓度工作液:取受试化合物和马来酸依那普利10mM储备液适量,用DMSO稀释至1mM,4℃冰箱中保存。
化合物工作液配制:分别取1mM的化合物中间浓度工作液适量,然后用乙腈-水(v:v=1:1)稀释至50μM,即得。
空白血浆准备:分别取适量的空白大鼠、小鼠、猴、犬、人的冻存血浆,在37℃进行解冻。
b、样品孵育:
取245μL空白大鼠血浆加入5μL化合物工作液,混合均匀;在37℃进行孵育,到达相应时间点后从孵育体系中取出血浆样品50μL,加入450μL冰乙腈(含内标)终止反应。涡旋混匀,离心取上清。
c、数据处理,采用LC-MS/MS检测化合物在各时间点内标归一化后的峰面积比,以0时间点化合物剩余量为100%,使用excel计算化合物在各个时间点的剩余量。
实验结果显示,本发明化合物与大鼠、小鼠、猴、犬、人5个种属血浆在37℃作用2h,剩余量均在100%左右,未发生明显变化,表明这类化合物具有很好的血浆稳定性,具有很好的成药性质。例如,以下表3列举了本发明2个示例性化合物的血浆稳定性测试数据:
表3

试验例4:肝脏药物代谢酶抑制活性测定
药物代谢的主要场所是肝脏,而肝脏中存在的混合功能氧化酶系的主要成分是CYP450酶,该酶系引起了临床上大多数药物的相互作用,进而导致药物不良反应发生率提高。CYP450是一组由许多同工酶组成的超基因大家族,其中CYP3A4作为主要代谢酶,参与了临床上将近一半药物的代谢。同样,CYP2C9、CYP2C19也作为重要代谢酶,临床上参与了多种药物代谢。因此,早期通过测试化合物对CYP450酶的抑制活性,可评判发生药物-药物相互作用的风险,提高用药安全性。
在本发明中,采用人肝微粒体作为CYP3A4酶源,将各个CYP同工酶的特异性探针底物(CYP3A4采用咪达唑仑和睾酮两个底物,分别以CYP3A4-M和CYP3A4-T表示),分别与一系列浓度的受试化合物在辅因子NADPH存在的条件下孵育。使用LC-MS/MS测定孵育体系中探针底物的代谢产物的生成量,计算受试化合物对各个CYP450酶亚型的IC50值,评价其对各个CYP450酶亚型的抑制作用。实验时,在孵育体系中依次加入49μL PBS,50μL探针底物,50μL人肝微粒体工作液,然后加入1μL各个浓度的受试化合物工作液,混合均匀;在37℃预孵5min后,加入50μL NADPH启动反应。孵育相应时间后,加入适量含内标的冰乙腈终止反应,涡旋混匀,离心取上清,进样LC-MS/MS检测探针底物的代谢产物的生成量。以0浓度点酶活性(以代谢产物生成量表征)为100%,计算不同受试化合物浓度下代谢产物的剩余酶活百分比。IC50通过剩余酶活由Graphpad Prism软件进行计算。
实验结果显示,本发明化合物对CYP2C9、CYP2C19、CYP3A4-M、CYP3A4-T的抑制活性明显弱于参照化合物MRTX0902,有利于降低药物-药物相互作用的风险。例如,以下表4列举了本发明3个示例性化合物和参照化合物MRTX0902的测试数据:
表4
试验例5:体内药效研究1
实验目的:本实验采用人胰腺癌MIA PaCa-2细胞皮下异种移植肿瘤模型评价化合物的抗肿瘤活性。
实验操作:选用Balb/c-nu裸鼠,雌性,6-8周,体重约18-22克。每只小鼠在右侧肩胛皮下接种0.1mL(5×106个)MIA PaCa-2细胞(加基质胶,体积比为1:1)。当平均肿瘤体积达到127mm3时开始给药。将受试化合物每日口服灌胃给药(给药体积10mL/kg),连续给药21天,给药剂量和给药频次如表5所示。肿瘤体积每周测量两次,体积以mm3计量,通过以下公式计算:V=0.5a×b2,其中a和b分别是肿瘤的长径和短径。化合物的抑瘤疗效用TGI(%)评价,TGI(%)反映肿瘤生长抑制率。TGI(%)的计算:TGI(%)=[1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)/(溶媒对照组给药结束时平均瘤体积-溶媒对照组开始给药时平均瘤体积)]×100%。体重变化(%)的计算:体重变化(%)=(某处理组给药结束时动物平均体重-该处理组开始给药时动物平均体重)/该处理组开始给药时动物平均体重×100%。
实验结果显示,本发明化合物在人胰腺癌MIA PaCa-2细胞皮下异种移植肿瘤模型上展现出较强的抑瘤活性,给药后瘤体积较参照化合物MRTX0902实验组明显更小,肿瘤生长抑制率TGI(%)显著提升。例如,以下表5列举了本发明2个示例性化合物和参照化合物MRTX0902在25mg/kg给药剂量下的实验数据:
表5
试验例6:体内药效研究11
实验目的:本实验采用人非小细胞肺癌NCI-H358细胞皮下异种移植肿瘤模型评价化合物的抗肿瘤活性。
实验操作:选用Balb/c-nu裸鼠,雌性,6-8周,体重约18-22克。每只小鼠在右侧肩胛皮下接种0.1mL(5×106个)NCI-H358细胞(加基质胶,体积比为1:1)。当平均肿瘤体积达到约170mm3时开始给药。将受试化合物每日口服灌胃给药(给药体积10mL/kg),连续给药21天,给药剂量和给药频次如表6所示。肿瘤体积每周测量两次,体积以mm3计量,通过以下公式计算:V=0.5a×b2,其中a和b分别是肿瘤的长径和短径。化合物的抑瘤疗效用TGI(%)评价,TGI(%)反映肿瘤生长抑制率。TGI(%)的计算:TGI(%)=[1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)/(溶媒对照组给药结束时平均瘤体积-溶媒对照组开始给药时平均瘤体积)]×100%。体重变化(%)的计算:体重变化(%)=(某处理组给药结束时动物平均体重-该处理组开始给药时动物平均体重)/该处理组开始给药时动物平均体重×100%。
实验结果显示,本发明化合物在人非小细胞肺癌NCI-H358细胞皮下异种移植肿瘤模型上展现出较强的抑瘤活性,给药后瘤体积较参照化合物MRTX0902实验组明显更小,肿瘤生长抑制率TGI(%)显著提升。例如,以下表6列举了本发明1个示例性化合物分别在25mg/kg、50mg/kg给药剂量下和参照化合物MRTX0902在25mg/kg给药剂量下的实验数据:
表6
从以上数据可以看出,本发明化合物在体外对SOS1具有明显的抑制作用,可用作SOS1抑制剂,在动物体内具有较优的抗肿瘤效果,在由SOS1蛋白介导的癌症、病原性皮疹病等疾病等领域具有广阔的应用前景。此外,本发明化合物与MRTX0902相比,具有更好的肝微粒体代谢稳定性和血浆稳定性,对肝脏药物代谢酶的抑制作用更 弱,且体内抗肿瘤活性显著提升,具有很好的成药性质和临床应用前景。
尽管已经示出和描述了本发明的实施例,对于本领域的普通技术人员而言,可以理解在不脱离本发明的原理和精神的情况下可以对这些实施例进行多种变化、修改、替换和变型,本发明的范围由所附权利要求及其等同物限定。

Claims (38)

  1. 式I所示的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶:
    其中:
    环A选自芳基、杂芳基,所述芳基、杂芳基可选地被选自卤素、烷基、烷氧基、-OH、-CN、-NR5R6、-SF5、-SO2R12的取代基所取代,所述烷基、烷氧基可选地进一步被选自卤素、-NR5R6的取代基所取代,其中,R5、R6独立选自H、烷基,R12为烷基且可选地被卤素所取代;
    Ra、Rb、Rc独立选自H、烷基;
    R1、R3、R4独立选自H、卤素、烷基、烯基、环烷基、环烯基、烷氧基、烷硫基、-OH、-CN、-NR7R8、-C(=O)H、-C(=O)NR19R20、-C(=O)OR21,所述烷基、烷氧基可选地被选自卤素、-NR7R8的取代基所取代,其中R7、R8、R19、R20、R21独立选自H、烷基;
    R2为-X-R13,X选自亚烷基、一单键,R13选自烷基、环烷基、杂环烷基、环烯基、杂环烯基、芳基、杂芳基;其中:
    当R13为烷基时,X为一单键,所述烷基可选地被选自-OH、烷氧基、卤素、-C(=O)NR9R10、-C(=O)OR11、-NR22R23的取代基所取代,R9、R10、R11、R22、R23独立选自H、烷基;
    当R13选自芳基、杂芳基时,可选地被选自卤素、烷基、烷氧基、-OH、-CN、-NR14R15的取代基所取代,R14、R15独立选自H、烷基;
    当R13选自环烷基、杂环烷基、环烯基、杂环烯基时,可选地被选自卤素、烷基、-Y-R16的取代基所取代,所述烷基可选地进一步被选自卤素、-OH、烷氧基、-NR17R18的取代基所取代;其中:
    Y为亚烷基,R16选自环烷基、杂环烷基、环烯基、杂环烯基、芳基、杂芳基;
    或者,Y选自R16选自烷基、烷氧基、-NR17R18、环烷基、杂环烷基、环烯基、杂环烯基、芳基、杂芳基;
    R17、R18独立选自H、烷基。
  2. 根据权利要求1所述的化合物,其特征在于,环A选自芳基、杂芳基,所述芳基、杂芳基可选地被选自卤素、烷基、烷氧基、-OH、-CN、-NR5R6的取代基所取代,所述烷基、烷氧基可选地进一步被选自卤素、-NR5R6的取代基所取代,其中R5、R6独立选自H、烷基。
  3. 根据权利要求1或2所述的化合物,其特征在于,环A选自6~12元芳基、5~12元杂芳基,所述杂芳基含有选自N、O、S的杂原子,上述基团的取代基如权利要求1或2所定义;
    优选地,环A选自6元芳基、5~11元杂芳基,所述杂芳基含有1~3个选自N、O、S的杂原子,上述基团的取代基如权利要求1或2所定义;
    优选地,环A选自6元芳基、6元芳基-5元杂芳基,所述杂芳基含有1个选自N、O、S的杂原子,上述基团的取代基如权利要求1或2所定义;
    优选地,环A选自苯基、苯基-噻吩基,上述基团的取代基如权利要求1或2所定义;
    优选地,环A为苯基,其取代基如权利要求1或2所定义。
  4. 根据权利要求1或3所述的化合物,其特征在于,环A含有的所述取代基选自卤素、C1~C6烷基、C1~C6烷氧基、-OH、-CN、-NR5R6、-SF5、-SO2R12,所述烷基、烷氧基可选地进一步被选自卤素、-NR5R6的取代基所取代,其中,R5、R6独立选自H、C1~C6烷基,R12为C1~C6烷基且可选地被卤素所取代;
    优选地,环A含有的所述取代基选自卤素、C1~C6烷基、-CN、-NR5R6、-SF5、-SO2R12,所述烷基可选地进一步被选自卤素、-NR5R6的取代基所取代,其中,R5、R6独立选自H、C1~C6烷基,R12为C1~C6烷基且可选地被卤素所取代;
    优选地,环A含有的所述取代基选自-F、甲基、-F取代的甲基、-NR5R6取代的甲基、-CN、-NR5R6、-SF5、-SO2R12,其中,R5、R6独立选自H、甲基,R12为-F取代的甲基;
    优选地,环A含有的所述取代基选自-F、-CH3、-CHF2、-CF3、-CH2NHCH3、-CN、-NH2、-SF5、-SO2CHF2
  5. 根据权利要求1~3任意一项所述的化合物,其特征在于,环A含有的所述取代基选自卤素、烷基、-CN,所述烷基可选地进一步被选自卤素、-NR5R6的取代基所取代,其中R5、R6独立选自H、烷基;
    优选地,所述取代基选自卤素、C1~C6烷基、-CN,所述烷基可选地进一步被选自卤素、-NR5R6的取代基所取代,其中R5、R6独立选自H、C1~C6烷基;
    优选地,所述取代基选自-F、甲基、-F取代的甲基、-NR5R6取代的甲基、-CN,其中R5、R6独立选自H、甲基;
    优选地,所述取代基选自-F、-CH3、-CHF2、-CF3、-CH2NHCH3、-CN、-NH2
  6. 根据权利要求1所述的化合物,其特征在于,环A选自:
    优选地,环A选自:
  7. 根据权利要求1~6任意一项所述的化合物,其特征在于,Ra、Rb、Rc独立选自H、C1~C6烷基;
    优选地,Ra、Rb、Rc独立选自H、甲基;
    优选地,Ra为甲基,Rb为H,Rc为H。
  8. 根据权利要求7所述的化合物,其特征在于,结构如式Ⅱ所示:
    其中各基团如权利要求1~6任意一项所定义。
  9. 根据权利要求1~8任意一项所述的化合物,其特征在于,R1、R3、R4独立选自H、烷基、烷氧基、-OH、-CN、-NR7R8,所述烷基、烷氧基可选地被选自卤素、-NR7R8的取代基所取代,其中R7、R8独立选自H、烷基;优选地,R1、R3、R4独立选自H、C1~C6烷基、C1~C6烷氧基、-OH、-CN、-NR7R8,所述烷基、烷氧基可选地被选自卤素、-NR7R8的取代基所取代,其中R7、R8独立选自H、C1~C6烷基;优选地,R1、R3、R4独立选自H、C1~C6烷基、C1~C6烷氧基、-OH、-CN;优选地,R1、R3、R4独立选自H、甲基、甲氧基、-OH、-CN;优选地,R1选自H、甲基、甲氧基、-CN;优选地,R3为H;优选地,R4选自甲基、甲氧基、-OH。
  10. 根据权利要求1~8任意一项所述的化合物,其特征在于,R1、R3、R4独立选自H、烷基、烷氧基、-OH、-NR7R8,所述烷基、烷氧基可选地被选自卤素、-NR7R8的取代基所取代,其中R7、R8独立选自H、烷基;优选地,R1、R3、R4独立选自H、烷基;优选地,R1、R3、R4独立选自H、C1~C6烷基;优选地,R1、R3、R4独立选自H、甲基;优选地,R1为H;优选地,R3为H;优选地,R4为甲基。
  11. 根据权利要求1~8任意一项所述的化合物,其特征在于,R1、R3独立选自H、卤素、C1~C6烷基、C1~C6烷氧基、C1~C6烷硫基、-OH、-CN、-NR7R8,所述烷基、烷氧基可选地被选自卤素、-NR7R8的取代基所取代,其中R7、R8独立选自H、C1~C6烷基;优选地,R1、R3独立选自H、C1~C6烷基、C1~C6烷氧基、-CN;优选地,R1、R3独立选自H、甲基、甲氧基、-CN;优选地,R1选自H、甲基、甲氧基、-CN;优选地,R1选自H、甲基、甲氧基;优选地,R3为H。
  12. 根据权利要求1~8任意一项所述的化合物,其特征在于,R4选自H、C1~C6烷基、C2~C6烯基、3~10元环烷基、3~10元环烯基、C1~C6烷氧基、C1~C6烷硫基、-OH、-CN、-C(=O)H、-C(=O)NR19R20、-C(=O)OR21,所述烷基可选地被卤素所取代,其中R19、R20、R21独立选自H、C1~C6烷基;
    优选地,R4选自C1~C6烷基、C2~C6烯基、3~5元环烷基、5元环烯基、C1~C6烷氧基、C1~C6烷硫基、-OH、-CN、-C(=O)H、-C(=O)NR19R20、-C(=O)OR21,所述烷基可选地被卤素所取代,其中R19、R20、R21独立选自H、C1~C6烷基;
    优选地,R4选自C1~C6烷基、-OH;
    优选地,R4选自甲基、乙基、二氟甲基、乙烯基、环丙烷基、环戊烯基、甲氧基、甲硫基、-OH、-CN、-C(=O)H、-C(=O)NH2、-C(=O)N(CH3)2、-C(=O)OH、-C(=O)OCH3
  13. 根据权利要求1~8任意一项所述的化合物,其特征在于,R4为卤素;优选地,R4为氯。
  14. 根据权利要求1~13任意一项所述的化合物,其特征在于,当R13为烷基时,X为一单键,所述烷基可选地被选自-OH、烷氧基、卤素、-C(=O)NR9R10、-C(=O)OR11的取代基所取代,R9、R10、R11独立选自H、烷基。
  15. 根据权利要求1~13任意一项所述的化合物,其特征在于,R2为-X-R13,X为一单键,R13为C1~C6烷基,所述C1~C6烷基可选地被选自-OH、C1~C6烷氧基、卤素、-C(=O)NR9R10、-C(=O)OR11、-NR22R23的取代基所取代,R9、R10、R11、R22、R23独立选自H、C1~C6烷基;
    优选地,R2为-X-R13,X为一单键,R13为C1~C6烷基,所述C1~C6烷基可选地被选自-OH、卤素、-C(=O)NR9R10、-NR22R23的取代基所取代,R9、R10、R22、R23独立选自C1~C6烷基;
    优选地,R2为-X-R13,X为一单键,R13为C1~C6烷基,所述C1~C6烷基可选地被选自-OH、-F、-C(=O)N(CH3)2、-N(CH3)2的取代基所取代;
    优选地,R2选自-CH3
  16. 根据权利要求1~13任意一项所述的化合物,其特征在于,R2为-X-R13,X为一单键,R13为C1~C6烷基,所述C1~C6烷基可选地被选自-OH、C1~C6烷氧基、卤素、-C(=O)NR9R10、-C(=O)OR11的取代基所取代,R9、R10、R11独立选自H、C1~C6烷基;
    优选地,R2为-X-R13,X为一单键,R13为C1~C6烷基,所述C1~C6烷基可选地被选自-OH、卤素、-C(=O)NR9R10的取代基所取代,R9、R10独立选自C1~C6烷基;
    优选地,R2为-X-R13,X为一单键,R13为C1~C6烷基,所述C1~C6烷基可选地被选自-OH、-F、-C(=O)N(CH3)2的取代基所取代;
    优选地,R2选自-CH3
  17. 根据权利要求1~13任意一项所述的化合物,其特征在于,R2为-X-R13,X选自C1~C3亚烷基、一单键,R13选自6~12元芳基、5~12元杂芳基,所述杂芳基含有选自N、O、S的杂原子,所述芳基、杂芳基可选地被选自卤素、C1~C6烷基、C1~C6烷氧基、-OH、-CN、-NR14R15的取代基所取代,R14、R15独立选自H、C1~C6烷基;
    优选地,R2为-X-R13,X为亚甲基、一单键,R13为苯基、5~6元杂芳基,所述杂芳基含有1~2个选自N、O、S的杂原子,所述苯基、杂芳基可选地被选自卤素、C1~C6烷基、C1~C6烷氧基、-OH、-CN、-NR14R15的取代基所取代,R14、R15独立选自H、C1~C6烷基;
    优选地,R2为-X-R13,X为一单键,R13为吡啶基、嘧啶基,所述吡啶基、嘧啶基可选地被C1~C6烷氧基所取代;
    优选地,R2选自
  18. 根据权利要求1~13任意一项所述的化合物,其特征在于,R2为-X-R13,X为亚甲基、一单键,R13为苯基、5~6元杂芳基,所述杂芳基含有1个选自N、O、S的杂原子,所述苯基、杂芳基可选地被选自卤素、C1~C6烷基、C1~C6烷氧基、-OH、-CN、-NR14R15的取代基所取代,R14、R15独立选自H、C1~C6烷基;
    优选地,R2为-X-R13,X为一单键,R13为吡啶基,所述吡啶基可选地被C1~C6烷氧基所取代;
    优选地,R2
  19. 根据权利要求1~13任意一项所述的化合物,其特征在于,R2为-X-R13,X选自C1~C3亚烷基、一单键,R13选自3~10元环烷基、3~10元杂环烷基、3~10元环烯基、3~10元杂环烯基,所述杂环烷基、杂环烯基含有选自 N、O、S的杂原子,所述环烷基、杂环烷基、环烯基、杂环烯基可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被选自卤素、-OH、C1~C6烷氧基、-NR17R18的取代基所取代;
    优选地,R2为-X-R13,X选自C1~C3亚烷基、一单键,R13选自3~10元环烷基、3~10元杂环烷基,所述杂环烷基含有选自N、O、S的杂原子,所述环烷基、杂环烷基可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被选自卤素、-OH的取代基所取代;
    优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自3~10元环烷基、4~8元杂环烷基,所述杂环烷基含有1~2个选自N、O、S的杂原子,所述环烷基、杂环烷基可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被选自卤素、-OH的取代基所取代;
    优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自环丙烷基、环丁烷基、环戊烷基、二环[1.1.1]戊烷基、环己烷基、金刚烷基、氮杂环丁烷基、四氢呋喃基、四氢吡喃基、四氢噻喃基、四氢吡咯基、哌啶基、哌嗪基、1,4-亚乙基哌啶基、吗啉基,上述基团可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被选自卤素、-OH的取代基所取代;
    优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自3~6元环烷基、5~8元杂环烷基,所述杂环烷基含有1~2个选自N、O、S的杂原子,所述环烷基、杂环烷基可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被选自卤素、-OH的取代基所取代;
    优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自环丙烷基、环丁烷基、环戊烷基、二环[1.1.1]戊烷基、环己烷基、四氢呋喃基、四氢吡喃基、四氢噻喃基、四氢吡咯基、哌啶基、1,4-亚乙基哌啶基、吗啉基,上述基团可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被选自卤素、-OH的取代基所取代;
    优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自3~6元环烷基,所述环烷基可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被卤素所取代;
    优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自环丙烷基、环丁烷基、环戊烷基、环己烷基,上述基团可选地被选自卤素、C1~C6烷基、-Y-R16的取代基所取代,所述C1~C6烷基可选地进一步被卤素所取代。
  20. 根据权利要求1~13、19任意一项所述的化合物,其特征在于,R17、R18独立选自H、C1~C6烷基;优选地,R17、R18独立选自H、甲基;优选地,R17、R18均为甲基。
  21. 根据权利要求19所述的化合物,其特征在于,R2为-X-R13,X选自亚甲基、一单键,R13选自环丙烷基、环丁烷基、环戊烷基、二环[1.1.1]戊烷基、环己烷基、金刚烷基、氮杂环丁烷基、四氢呋喃基、四氢吡喃基、四氢噻喃基、四氢吡咯基、哌啶基、哌嗪基、1,4-亚乙基哌啶基、吗啉基,上述基团可选地被选自-F、甲基、的取代基所取代,其中甲基可选地进一步被选自-F、-OH的取代基所取代;
    优选地,R2选自:
    优选地,R2为-X-R13,X选自亚甲基、一单键,R13选自环丙烷基、环丁烷基、环戊烷基、二环[1.1.1]戊烷基、环己烷基、四氢呋喃基、四氢吡喃基、四氢噻喃基、四氢吡咯基、哌啶基、1,4-亚乙基哌啶基、吗啉基,上述基团可选地被选自-F、甲基、的取代基所取代,其中甲基可选地进一步被选自-F、-OH的取代基所取代;
    优选地,R2选自:
  22. 根据权利要求19或21所述的化合物,其特征在于,R2为-X-R13,X为一单键,R13选自环丙烷基、环丁烷基、环戊烷基、环己烷基,上述基团可选地被选自-F、甲基的取代基所取代,其中甲基可选地进一步被-F所取代;
    优选地,R2选自:
  23. 根据权利要求1~13、19任意一项所述的化合物,其特征在于,Y为C1~C3亚烷基,R16选自3~10元环烷基、3~10元杂环烷基、3~10元环烯基、3~10元杂环烯基、6~12元芳基、5~12元杂芳基,所述杂环烷基、杂环烯基、杂芳基含有选自N、O、S的杂原子;
    优选地,Y为C1~C3亚烷基,R16为3~10元杂环烷基,所述杂环烷基含有1个选自N、O、S的杂原子;
    优选地,Y为亚甲基,R16为5元杂环烷基,所述杂环烷基含有1个选自N、O、S的杂原子;
    优选地,Y为亚甲基,R16为四氢吡咯基。
  24. 根据权利要求23所述的化合物,其特征在于,R2
  25. 根据权利要求1~13、19、20任意一项所述的化合物,其特征在于,Y选自R16选自C1~C6烷基、C1~C6烷氧基、-NR17R18、3~10元环烷基、3~10元杂环烷基、3~10元环烯基、3~10元杂环烯基、6~12元芳基、5~12元杂芳基,所述杂环烷基、杂环烯基、杂芳基含有选自N、O、S的杂原子;
    优选地,Y选自R16选自C1~C6烷基、C1~C6烷氧基、-NR17R18、3~10元杂环烷基、6~12元芳基、5~12元杂芳基,所述杂环烷基、杂芳基含有1~2个选自N、O、S的杂原子;
    优选地,Y选自R16选自C1~C6烷基、C1~C6烷氧基、-NR17R18、5~6元杂环烷基、6元芳基、5~6元杂芳基,所述杂环烷基、杂芳基含有1~2个选自N、O的杂原子;
    优选地,Y选自R16选自甲基、甲氧基、-N(CH3)2、吗啉基、苯基、异噁唑基。
  26. 根据权利要求25所述的化合物,其特征在于,R2选自
    优选地,R2选自
  27. 根据权利要求1~13任意一项所述的化合物,其特征在于,R2选自环烷基、杂环烷基、环烯基、杂环烯基,所述环烷基、杂环烷基、环烯基、杂环烯基可选地被选自卤素、烷基、卤素取代的烷基、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;或者,R2选自烷基,所述烷基可选地被选自卤素、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;其中,R9、R10、R11独立选自H、烷基;
    优选地,R2选自3~10元环烷基、3~10元杂环烷基、3~10元环烯基、3~10元杂环烯基,所述杂环烷基、杂环烯基含有选自N、O、S的杂原子,所述环烷基、杂环烷基、环烯基、杂环烯基可选地被选自卤素、烷基、卤素取代的烷基、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;或者,R2选自C1~C6烷基,所述C1~C6烷基可选地被选自卤素、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;其中,R9、R10、R11独立选自H、烷基;
    优选地,R2选自3~6元环烷基、3~6元杂环烷基、5~6元环烯基、5~6元杂环烯基,所述杂环烷基、杂环烯基含有选自N、O、S的杂原子,所述环烷基、杂环烷基、环烯基、杂环烯基可选地被选自卤素、烷基、卤素取代的烷基、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;或者,R2选自C1~C6烷基,所述C1~C6烷基可选地被选自卤素、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;其中,R9、R10、R11独立选自H、烷基;
    优选地,R2选自3~6元环烷基、3~6元杂环烷基,所述杂环烷基含有1~2个选自N、O、S的杂原子,所述环烷基、杂环烷基可选地被选自卤素、C1~C6烷基、卤素取代的C1~C6烷基、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;或者,R2选自C1~C6烷基,所述C1~C6烷基可选地被选自卤素、-C(=O)NR9R10的取代基所取代;其中,R9、R10、R11独立选自C1~C6烷基;
    优选地,R2选自3~6元环烷基、5~6元杂环烷基,所述杂环烷基含有1个选自O、S的杂原子,所述环烷基、杂环烷基可选地被选自卤素、C1~C3烷基、卤素取代的C1~C3烷基、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;或者,R2选自C1~C3烷基,所述C1~C3烷基可选地被选自卤素、-C(=O)NR9R10的取代基所取代;其中,R9、R10、R11独立选自C1~C3烷基;
    优选地,R2选自环丙烷基、环丁烷基、环戊烷基、二环[1.1.1]戊烷基、环己烷基、四氢呋喃基、四氢吡喃基、四氢噻喃基,上述基团可选地被选自卤素、C1~C3烷基、卤素取代的C1~C3烷基、-C(=O)NR9R10、-C(=O)OR11的取代基所取代;或者,R2选自甲基、乙基、丙基,上述基团可选地被选自卤素、-C(=O)NR9R10的取代基所取代;其中,R9、R10、R11独立选自C1~C3烷基;
    优选地,R2选自环丙烷基、环丁烷基、环戊烷基、二环[1.1.1]戊烷基、环己烷基、四氢呋喃基、四氢吡喃基、四氢噻喃基,上述基团可选地被选自氟、甲基、三氟甲基、-C(=O)N(CH3)2、-C(=O)OCH3的取代基所取代;或者,R2选自甲基、乙基、丙基,上述基团可选地被选自氟、-C(=O)N(CH3)2的取代基所取代;
    优选地,R2选自: -CH3 优选地,为如下构型:
  28. 根据权利要求1~27任意一项所述的化合物,其特征在于,选自如下之一:



  29. 药物组合物,其特征在于,含有权利要求1~28任意一项所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,以及药学上可接受的辅料或者辅助性成分。
  30. 权利要求1~28任意一项所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,或权利要求29所述的药物组合物在制备SOS1抑制剂中的用途。
  31. 权利要求1~28任意一项所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,或权利要求29所述的药物组合物在制备用于治疗由SOS1介导的疾病的药物中的用途。
  32. 根据权利要求31所述用途,其特征在于,所述疾病选自:癌症、病原性皮疹病;
    优选地,所述癌症选自:非小细胞肺癌、肺癌、胰腺癌、卵巢癌、膀胱癌、前列腺癌、慢性粒细胞白血病、结直肠癌、脑癌、肝癌、肾癌、胃癌、及乳腺癌;
    所述病原性皮疹病选自:努南综合症、心面皮肤综合症、I型遗传性牙龈纤维瘤病。
  33. 权利要求1~28任意一项所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,或权利要求29所述的药物组合物在制备治疗致使SOS1蛋白过度表达的疾病的药物中的用途。
  34. 权利要求1~28任意一项所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,或权利要求29所述的药物组合物在制备治疗SOS1蛋白过度表达所致疾病的药物中的用途。
  35. 药物组合物,其特征在于,含有同时或者分别给药的权利要求1~28任意一项所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,以及RAS抑制剂。
  36. 根据权利要求35所述的药物组合物,其特征在于,所述RAS抑制剂为KRAS抑制剂;优选地,所述RAS抑制剂为KRAS G12C抑制剂;优选地,所述RAS抑制剂为Adagrasib。
  37. 权利要求35或36所述的药物组合物在制备治疗癌症的药物中的用途。
  38. 治疗由SOS1介导的疾病的方法,其特征在于,包括向受试者施用权利要求1~28任意一项所述的化合物,或其互变异构体、立体异构体、溶剂化物、代谢产物、同位素标记物、药学上可接受的盐、共晶,或权利要求29、35或36任意一项所述的药物组合物的步骤。
PCT/CN2023/104412 2022-07-01 2023-06-30 哒嗪并吡啶酮衍生物及其用途 WO2024002318A1 (zh)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN202210765419.0 2022-07-01
CN202210765419 2022-07-01
CN202211318236 2022-10-26
CN202211318236.0 2022-10-26
CN202310164106.4 2023-02-24
CN202310164106 2023-02-24

Publications (1)

Publication Number Publication Date
WO2024002318A1 true WO2024002318A1 (zh) 2024-01-04

Family

ID=89383334

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/104412 WO2024002318A1 (zh) 2022-07-01 2023-06-30 哒嗪并吡啶酮衍生物及其用途

Country Status (1)

Country Link
WO (1) WO2024002318A1 (zh)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111372932A (zh) * 2017-12-21 2020-07-03 勃林格殷格翰国际有限公司 作为sos1抑制剂的新颖苄氨基取代吡啶并嘧啶酮及衍生物
WO2021127429A1 (en) * 2019-12-20 2021-06-24 Mirati Therapeutics, Inc. Sos1 inhibitors
CN113200981A (zh) * 2021-02-10 2021-08-03 杭州英创医药科技有限公司 作为sos1抑制剂的杂环化合物
WO2023125737A1 (en) * 2021-12-29 2023-07-06 Silexon Ai Technology Co., Ltd. Heterocyclic compounds and use thereof
WO2023135260A1 (en) * 2022-01-14 2023-07-20 Jazz Pharmaceuticals Ireland Limited Novel amine-substituted phthalazines and derivatives as sos1 inhibitors
WO2023143147A1 (zh) * 2022-01-28 2023-08-03 上海优理惠生医药有限公司 一种哒嗪并吡啶酮类化合物、其药物组合物及应用

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111372932A (zh) * 2017-12-21 2020-07-03 勃林格殷格翰国际有限公司 作为sos1抑制剂的新颖苄氨基取代吡啶并嘧啶酮及衍生物
WO2021127429A1 (en) * 2019-12-20 2021-06-24 Mirati Therapeutics, Inc. Sos1 inhibitors
CN113200981A (zh) * 2021-02-10 2021-08-03 杭州英创医药科技有限公司 作为sos1抑制剂的杂环化合物
WO2023125737A1 (en) * 2021-12-29 2023-07-06 Silexon Ai Technology Co., Ltd. Heterocyclic compounds and use thereof
WO2023135260A1 (en) * 2022-01-14 2023-07-20 Jazz Pharmaceuticals Ireland Limited Novel amine-substituted phthalazines and derivatives as sos1 inhibitors
WO2023143147A1 (zh) * 2022-01-28 2023-08-03 上海优理惠生医药有限公司 一种哒嗪并吡啶酮类化合物、其药物组合物及应用

Similar Documents

Publication Publication Date Title
JP7394074B2 (ja) 治療用化合物
CN111484477B (zh) 一种苯并吡啶酮杂环化合物及其用途
ES2857251T3 (es) Compuestos polifluorados que actúan como inhibidores de la tirosina cinasa de bruton
JP7046940B2 (ja) 神経栄養因子チロシンキナーゼ受容体阻害剤として用いられるアミノピラゾロピリミジン化合物
US20200123172A1 (en) Cot modulators and methods of use thereof
TW201716415A (zh) 作為蛋白質激酶之調節劑的掌性二芳基巨環
JP7200120B2 (ja) Mk2阻害剤として有用なヘテロアリール化合物
WO2019134539A1 (zh) 二氢吡唑酮并嘧啶类化合物及其制备方法和用途
CN114026104B (zh) Cot调节剂及其使用方法
TWI828712B (zh) 作為trk抑制劑的雜環化合物
BR112014017749B1 (pt) Composto alquinilbenzeno 3,5-dissubstituído e sal do mesmo
CN113950481A (zh) 大环化合物
JP2024505732A (ja) ピリドピリミジノン系誘導体及びその製造方法と使用
CN112745335A (zh) 一种三并杂环化合物及其用途
CN112851663A (zh) 一种并杂环化合物及其用途
WO2021228248A1 (zh) 氮杂稠环酰胺类化合物及其用途
CN113024544A (zh) 一种含氰基并杂环化合物及其用途
CN112707905A (zh) 一种三并杂环化合物及其制备方法和用途
WO2022028506A1 (zh) Sos1抑制剂、包含其的药物组合物及其用途
JP2023522863A (ja) Egfr阻害剤としての三環式化合物
EP4065230A1 (en) Therapeutic compounds
CN113527299A (zh) 一类含氮稠环类化合物、制备方法和用途
CN112300173B (zh) 一类含氮多环类化合物、制备方法和用途
WO2018228275A1 (zh) 作为mnk抑制剂的杂环化合物
WO2023125928A1 (zh) Menin抑制剂及其用途

Legal Events

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

Ref document number: 23830482

Country of ref document: EP

Kind code of ref document: A1