CN114728943A - Compounds as BCR-ABL inhibitors - Google Patents

Compounds as BCR-ABL inhibitors Download PDF

Info

Publication number
CN114728943A
CN114728943A CN202180006423.2A CN202180006423A CN114728943A CN 114728943 A CN114728943 A CN 114728943A CN 202180006423 A CN202180006423 A CN 202180006423A CN 114728943 A CN114728943 A CN 114728943A
Authority
CN
China
Prior art keywords
compound
amino
alkyl
preparation
hydroxy
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN202180006423.2A
Other languages
Chinese (zh)
Inventor
张寅生
陆鹏
李久香
杨加举
秦慧
叶嘉炜
诸丽娟
汪纪楠
施伟
王晓金
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chia Tai Tianqing Pharmaceutical Group Co Ltd
Original Assignee
Chia Tai Tianqing Pharmaceutical Group Co Ltd
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 Chia Tai Tianqing Pharmaceutical Group Co Ltd filed Critical Chia Tai Tianqing Pharmaceutical Group Co Ltd
Publication of CN114728943A publication Critical patent/CN114728943A/en
Pending legal-status Critical Current

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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Landscapes

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

Abstract

The field of pharmaceutical chemistry, compounds serving as BCR-ABL inhibitors, namely compounds shown as a formula (I) or pharmaceutically acceptable salts thereof, a preparation method thereof, a pharmaceutical composition containing the compounds, and application of the compounds in preparing medicines for treating BCR-ABL related diseases.

Description

Compounds as BCR-ABL inhibitors
Reference to related applications
This application claims the benefit and priority of the chinese patent application having application number 202010062317.3 filed on 19.01.2020 to the intellectual property office of the people's republic of china and the benefit and priority of the chinese patent application having application number 202011246125.4 filed on 10.11.2020 to the intellectual property office of the people's republic of china, the entire contents of which are hereby incorporated by reference in their entirety.
Technical Field
The application belongs to the field of pharmaceutical chemistry, and provides a compound serving as a BCR-ABL inhibitor, a preparation method thereof, a pharmaceutical composition containing the compound, and application of the compound in preparation of medicines for treating BCR-ABL related diseases.
Background
Chronic Myelocytic Leukemia (CML) is the main disease species of chronic leukemia in China, and accounts for about 70% of chronic leukemia. Chromosome abnormalities are present in more than 90% of the cases. Mainly translocate the long arm of chromosome 9 and 22 to form Bcr-Abl fusion gene and express protein p-210. The tyrosine kinase activity of p-210 is much stronger than that of p-150 (normal c-Abl gene expression product), which causes abnormal proliferation and differentiation of hematopoietic stem cells, and finally triggers CML.
Imatinib is the first Bcr-Abl targeted therapeutic drug on the market and is currently used as a first-line therapeutic drug for the treatment of various stages of CML. Nilotinib, dasatinib and bosutinib (second generation Bcr-Abl inhibitors) are approved for the treatment of imatinib-resistant or intolerant CML. The second generation Bcr-Abl inhibitors all have superior potency to imatinib and are effective against almost all imatinib resistant mutant types, but still do not address the problem of mutant resistance of T315I (gatekeeper). The T315I mutation accounts for about 20% of patients with drug resistance of the first and second generation Bcr-Abl inhibitors, and the marketing of ponatinib (third generation Bcr-Abl inhibitor) relieves the predicament that patients with T315I drug resistance mutation can not be treated with drugs, and is the only choice for patients with failure of the first and second generation Bcr-Abl inhibitor treatment.
ABL001 is a Bcr-ABL allosteric inhibitor developed by noval, which is disclosed in WO2013171639, currently in phase III clinical research. ABL001 is a potent, selective inhibitor of BCR-ABL, active against most mutants, such as T315I. (Andrew A. Wylie et al (2017) Nature 543, 733-737).
Figure PCTCN2021072699-APPB-000001
At present, no BCR-ABL allosteric inhibitor is on the market, so that a high-efficiency and low-toxicity Bcr-ABL inhibitor for T315I is needed in clinic, and a new BCR-ABL allosteric inhibitor needs to be further developed.
Disclosure of Invention
In one aspect, the present application provides a compound of formula (I) or a pharmaceutically acceptable salt thereof,
Figure PCTCN2021072699-APPB-000002
wherein,
R 1is selected from
Figure PCTCN2021072699-APPB-000003
Or alternatively
Figure PCTCN2021072699-APPB-000004
R 2Selected from hydrogen, 3-10 membered heterocyclyl or amino, wherein said 3-10 membered heterocyclyl or amino is optionally substituted with one or more RaSubstitution;
R aselected from hydroxy, cyano, halogen,
Figure PCTCN2021072699-APPB-000005
C 1-6Alkyl radical, C1-6Alkoxy, di (C)1-6Alkyl) amino, or substituted by one or more hydroxy or C1-6Alkoxy-substituted C1-6An alkyl group;
R 3is selected from-OCF2H, wherein said-OCF2H is optionally substituted with halogen;
R 4selected from hydrogen, C1-6Alkyl radical, C3-6Cycloalkyl or 3-10 membered heterocyclyl, wherein C1-6Alkyl radical, C3-6Cycloalkyl or 3-10 membered heterocyclyl is optionally substituted with one or more RbSubstitution;
R bselected from deuterium, hydroxy, C1-6Alkoxy radical, C1-6Alkyl, amino, mono (C)1-6Alkyl) amino or di (C)1-6Alkyl) amino;
R 5is selected from C1-6An alkyl group;
ring a is selected from 5-6 membered heteroaryl or phenyl.
In some embodiments, RaSelected from hydroxy, halogen, C1-6Alkyl, di (C) 1-6Alkyl) amino, or C substituted by one or more hydroxy groups1-6An alkyl group; rbSelected from hydroxy, C1-6Alkoxy radical, C1-6Alkyl, amino, mono (C)1-6Alkyl) amino or di (C)1-6Alkyl) amino.
In some embodiments, R1Is selected from
Figure PCTCN2021072699-APPB-000006
Other variables are as defined herein.
In some embodiments, R1Is selected from
Figure PCTCN2021072699-APPB-000007
Other variables are as defined herein.
In some embodiments, R2Selected from hydrogen, 3-10 membered heterocycloalkyl, 3-10 membered heterocycloalkenyl or amino, wherein said 3-10 membered heterocycloalkyl, 3-10 membered heterocycloalkenyl or amino is optionally substituted with one or more RaSubstitution; other variables are as defined herein.
In some embodiments, R2Selected from hydrogen, 4-6 membered heterocyclyl, 7-9 membered spiroheterocyclyl or amino, wherein said 4-6 membered heterocyclyl, 7-9 membered spiroheterocyclyl or amino is optionally substituted with one or more RaSubstitution; other variables are as defined herein.
In some embodiments, R2Selected from hydrogen, 4-6 membered heterocyclyl or amino, wherein said 4-6 membered heterocyclyl or amino is optionally substituted with one or more RaSubstitution; other variables are as defined herein.
In some embodiments, R2Selected from hydrogen, 4-6 membered heterocycloalkyl, 6 membered heterocycloalkenyl, 7 or 9 membered spiroheterocycloalkyl or amino, wherein said 4-6 membered heterocycloalkyl, 6 membered heterocycloalkenyl, 7 or 9 membered spiroheterocycloalkyl or amino is optionally substituted with one or more R aSubstitution; other variables are as defined herein.
In some embodiments, R2Selected from the group consisting of hydrogen, pyrrolidinyl, morpholinyl, piperazinyl, azetidinyl, piperidinyl, 1,2,3, 6-tetrahydropyridinyl, tetrahydropyranyl, 3, 4-dihydropyranyl, 3, 6-dihydropyranyl, 2-oxa-7-azaspiro [3.5]]Nonyl, 2-oxa-6-azaspiro [3.3]]Heptalkyl or amino, wherein the pyrrolidinyl, morpholinyl, piperazinyl, azetidinyl, piperidinyl, 1,2,3, 6-tetrahydropyridinyl, tetrahydropyranyl, 3, 4-dihydropyranyl, 3, 6-dihydropyranyl, 2-oxa-7-azaspiro [3.5]]Nonanyl, 2-oxa-6-azaspiro [3.3]Heptylalkyl or amino optionally substituted with one or more RaSubstitution; other variables are as defined herein.
In some embodiments, R2Selected from hydrogen, pyrrolidinyl, morpholinyl, piperazinyl, azetidinyl, piperidinyl, 3, 4-dihydropyranyl, or amino, wherein said pyrrolidinyl, morpholinyl, piperazinyl, azetidinyl, piperidinyl, 3, 4-dihydropyranyl, or amino is optionally substituted with one or more RaSubstitution; other variables are as defined herein.
In some embodiments, R2Selected from hydrogen,
Figure PCTCN2021072699-APPB-000008
Figure PCTCN2021072699-APPB-000009
Or an amino group, wherein
Figure PCTCN2021072699-APPB-000010
Figure PCTCN2021072699-APPB-000011
Or amino optionally substituted with one or more RaSubstitution; othersThe variables are as defined herein.
In some embodiments, RaSelected from hydroxy, cyano, halogen,
Figure PCTCN2021072699-APPB-000012
C 1-4Alkyl radical, C1-3Alkoxy, di (C)1-3Alkyl) amino groups or substituted by one or more hydroxy groups or C1-3Alkoxy-substituted C1-4An alkyl group; other variables are as defined herein.
In some embodiments, RaSelected from hydroxy, halogen, C1-3Alkyl, di (C)1-3Alkyl) amino, or C substituted by one or more hydroxy groups1-3An alkyl group; other variables are as defined herein.
In some embodiments, RaSelected from hydroxy, cyano, fluoro,
Figure PCTCN2021072699-APPB-000013
Methyl, methoxy, 2-hydroxyethyl, 2-methoxyethyl, 2-methyl-2-hydroxypropyl, di (methyl) amino or hydroxymethyl; other variables are as defined herein.
In some embodiments, RaSelected from hydroxy, fluoro, methyl, 2-hydroxyethyl, di (methyl) amino or hydroxymethyl; other variables are as defined herein.
In some embodiments, R2Selected from the group consisting of hydrogen, pyrrolidinyl, morpholinyl, piperazinyl, azetidinyl, piperidinyl, 1,2,3, 6-tetrahydropyridinyl, tetrahydropyranyl, 3, 4-dihydropyranyl, 3, 6-dihydropyranyl, 2-oxa-7-azaspiro [3.5]]Nonanyl, 2-oxa-6-azaspiro [3.3]Heptaalkyl or amino, wherein said pyrrolidinyl is optionally substituted with one hydroxy, cyano, fluoro, cyano, nitro, or a pharmaceutically acceptable salt thereof,
Figure PCTCN2021072699-APPB-000014
Methoxy or di (methyl) amino, wherein the piperazinyl, piperidinyl or morpholinyl is optionally substituted with one or two hydroxy or methyl groups, wherein the azetidinyl is optionally substituted with one or two hydroxy, fluoro, cyano, methyl, methoxy or hydroxymethyl groups, wherein the amino is optionally substituted with one or more 2-hydroxyethyl, methyl, 2-methoxyethyl or 2-methyl-2-hydroxypropyl groups; other variables are as defined herein.
In some embodiments, R2Selected from hydrogen, pyrrolidinyl, morpholinyl, piperazinyl, azetidinyl, piperidinyl, 3, 4-dihydropyranyl, or amino, wherein said pyrrolidinyl is optionally substituted with one hydroxy, fluoro, or di (methyl) amino, wherein said piperazinyl or piperidinyl is optionally substituted with one methyl, wherein said azetidinyl is optionally substituted with one hydroxy or hydroxymethyl, wherein said amino is optionally substituted with one or more 2-hydroxyethyl or methyl; other variables are as defined herein.
In some embodiments, R2Selected from hydrogen,
Figure PCTCN2021072699-APPB-000015
Figure PCTCN2021072699-APPB-000016
Figure PCTCN2021072699-APPB-000017
Other variables are as defined herein.
In some embodiments, R2Selected from hydrogen,
Figure PCTCN2021072699-APPB-000018
Figure PCTCN2021072699-APPB-000019
Figure PCTCN2021072699-APPB-000020
Other variables are as defined herein.
In some embodiments, R3Is selected from-OCF3or-OCF2Cl; other variables are as defined herein.
In some embodiments, R4Selected from hydrogen, C1-6Alkyl radical, C3-6Cycloalkyl or 3-to 10-membered heterocycloalkyl, wherein C1-6Alkyl radical, C3-6Cycloalkyl or 3-10 membered heterocycloalkyl optionally substituted with one or more RbSubstitution; other variables are as defined herein.
In some embodiments, R4Selected from hydrogen, C1-5Alkyl radical, C3-6Cycloalkyl or 4-6 membered heterocycloalkyl, wherein C1-5Alkyl radical, C3-6Cycloalkyl or 4-6 membered heterocycloalkyl optionally substituted with one or more RbSubstitution; other variables are as defined herein.
In some embodiments, R4Selected from hydrogen, C1-5Alkyl, cyclopropyl or 6-membered heterocycloalkyl, wherein C1-5Alkyl, cyclopropyl or 6-membered heterocycloalkyl optionally substituted with one or more RbSubstitution; other variables are as defined herein.
In some embodiments, R4Selected from hydrogen, methyl, ethyl, 2-methylpropyl, 3-methylbutyl, cyclopropyl, tetrahydropyranyl or piperidinyl, wherein methyl, ethyl, 2-methylpropyl, 3-methylbutyl, cyclopropyl, tetrahydropyranyl or piperidinyl is optionally substituted by one or more RbSubstitution; other variables are as defined herein.
In some embodiments, R4Selected from hydrogen, methyl, ethyl, 2-methylpropyl, 3-methylbutyl, cyclopropylA base,
Figure PCTCN2021072699-APPB-000021
Figure PCTCN2021072699-APPB-000022
Wherein methyl, ethyl, 2-methylpropyl, 3-methylbutyl, cyclopropyl, methyl, ethyl, propyl, isopropyl, and the like,
Figure PCTCN2021072699-APPB-000023
Optionally substituted by one or more RbSubstitution; other variables are as defined herein.
In some embodiments, RbSelected from deuterium, hydroxy, C1-3Alkoxy radical, C1-3Alkyl, amino, mono (C)1-3Alkyl) amino or di (C)1-3Alkyl) amino; the other variables are as defined herein.
In some embodiments, RbSelected from hydroxy, C1-3Alkoxy radical, C1-3Alkyl, amino, mono (C)1-3Alkyl) amino or di (C)1-3Alkyl) amino; other variables are as defined herein.
In some embodiments, RbSelected from deuterium, hydroxy, methoxy, methyl or di (ethyl) amino; other variables are as defined herein.
In some embodiments, RbSelected from hydroxy, methoxy, methyl or di (ethyl) amino; other variables are as defined herein.
In some embodiments, R4Selected from hydrogen, methyl, ethyl, 2-methylpropyl, 3-methylbutyl, cyclopropyl, tetrahydropyranyl or piperidinyl, wherein methyl is optionally substituted with three deuterium groups, wherein ethyl is optionally substituted with one hydroxy, methoxy or di (ethyl) amino group, wherein 2-methylpropyl or 3-methylbutyl is optionally substituted with one hydroxy group, wherein piperidineOptionally substituted with one methyl group; other variables are as defined herein.
In some embodiments, R4Selected from hydrogen, methyl, d3-methyl, cyclopropyl,
Figure PCTCN2021072699-APPB-000024
Figure PCTCN2021072699-APPB-000025
Other variables are as defined herein.
In some embodiments, R4Selected from hydrogen; other variables are as defined herein.
In some embodiments, R4Selected from methyl or cyclopropyl; the other variables are as defined herein.
In some embodiments, R5Is selected from C1-3An alkyl group; other variables are as defined herein.
In some embodiments, R5Is selected from methyl; other variables are as defined herein.
In some embodiments, ring a is selected from a 5-6 membered nitrogen containing heteroaryl or phenyl; other variables are as defined herein.
In some embodiments, ring a is selected from pyrrolyl, pyridinyl, or phenyl; other variables are as defined herein.
In some embodiments, ring a is selected from pyrrolyl; other variables are as defined herein.
In some embodiments, a building block
Figure PCTCN2021072699-APPB-000026
Is selected from
Figure PCTCN2021072699-APPB-000027
Figure PCTCN2021072699-APPB-000028
Other variables are as defined herein.
In some embodiments, a building block
Figure PCTCN2021072699-APPB-000029
Is selected from
Figure PCTCN2021072699-APPB-000030
Is further selected from
Figure PCTCN2021072699-APPB-000031
Figure PCTCN2021072699-APPB-000032
Other variables are as defined herein.
In some embodiments, R1Is selected from
Figure PCTCN2021072699-APPB-000033
Or
Figure PCTCN2021072699-APPB-000034
R 2Selected from hydrogen, 3-10 membered heterocyclyl or amino, wherein said 3-10 membered heterocyclyl or amino is optionally substituted with one or more RaSubstitution;
R aselected from hydroxy, halogen, C1-6Alkyl, di (C)1-6Alkyl) amino or C substituted by one or more hydroxy groups1-6An alkyl group;
R 3is selected from-OCF2H, wherein said-OCF 2H is optionally substituted with halo;
R 4selected from hydrogen, C1-6Alkyl radical, C3-6Cycloalkyl or 3-10 membered heterocyclyl, wherein C1-6Alkyl radical, C3-6Cycloalkyl or 3-10 membered heterocyclyl is optionally substituted with one or more RbSubstitution;
R bselected from hydroxy, C1-6Alkoxy radical, C1-6Alkyl, amino, mono (C)1-6Alkyl) amino or di (C)1-6Alkyl) amino;
R 5is selected from C1-6An alkyl group;
ring a is selected from 5-6 membered heteroaryl or phenyl.
In another aspect, the present application provides a compound of formula (II) or a pharmaceutically acceptable salt thereof,
Figure PCTCN2021072699-APPB-000035
wherein,
R 3is selected from-OCF3or-OCF2Cl;
R 2、R 4And RaAnd RbAs defined above.
In another aspect, the present application provides a compound of formula (III), or a pharmaceutically acceptable salt thereof,
Figure PCTCN2021072699-APPB-000036
wherein,
structural unit
Figure PCTCN2021072699-APPB-000037
Is selected from
Figure PCTCN2021072699-APPB-000038
R 3Is selected from-OCF3or-OCF2Cl;
R 2And RaAs defined above.
In some embodiments, the present application provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein,
R 1is selected from
Figure PCTCN2021072699-APPB-000039
Or
Figure PCTCN2021072699-APPB-000040
R 2Selected from hydrogen, 4-10 membered heterocyclyl or amino containing 1-3 heteroatoms selected from N or O or S, wherein said 4-10 membered heterocyclyl or amino is optionally substituted with one or more RaSubstitution;
R aselected from hydroxy, cyano, halogen,
Figure PCTCN2021072699-APPB-000041
C 1-6Alkyl radical, C1-6Alkoxy, di (C)1-6Alkyl) amino, or substituted by one or more hydroxy or C1-6Alkoxy-substituted C1-6An alkyl group;
R 3is selected from-OCF3、-OCF 2Cl or-OCF2Br;
R 4Selected from hydrogen, C1-6Alkyl radical, C3-6Cycloalkyl, or a 5-to 7-membered heterocyclic group containing 1 to 3 heteroatoms selected from N or O or S, wherein C1-6Alkyl radical, C3-6Cycloalkyl or 5-7 membered heterocyclyl is optionally substituted with one or more RbSubstitution;
R bselected from deuterium, hydroxy, C1-6Alkoxy radical, C1-6An alkyl group;
R 5is selected from C1-6An alkyl group;
ring a is selected from 5-6 membered heteroaryl or phenyl containing 1-3 heteroatoms selected from N or O or S.
In some embodiments, the present application provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein,
R 1is selected from
Figure PCTCN2021072699-APPB-000042
Or
Figure PCTCN2021072699-APPB-000043
R 2Selected from hydrogen, a 4-9 membered heterocyclyl or amino group containing 1-3 (e.g. 1,2 or 3) heteroatoms selected from N or O or S (preferably N or O), wherein said 4-9 membered heterocyclyl or amino group is optionally substituted by 1 or 2RaSubstitution;
R aselected from hydroxy, cyano, halogen (preferably F),
Figure PCTCN2021072699-APPB-000044
C 1-3Alkyl (preferably methyl, ethyl), C1-3Alkoxy (preferably methoxy, ethoxy), di (C)1-6Alkyl) amino, or by a hydroxy or C group1-3Alkoxy (preferably methoxy, ethoxy) substituted C1-4An alkyl group;
R 3is selected from-OCF3or-OCF2Cl;
R 4Selected from hydrogen, C1-4Alkyl, aryl, heteroaryl, and heteroaryl,C 3Cycloalkyl, or a 6-membered heterocyclic group containing 1-2 (preferably 1) heteroatoms selected from N or O or S, wherein C is1-4Alkyl radical, C3Cycloalkyl or 6-membered heterocyclyl is optionally substituted with one or more RbSubstitution;
R bselected from deuterium, hydroxy and C1-3Alkoxy (e.g., methoxy, ethoxy, propoxy);
R 5is selected from C1-3Alkyl (e.g., methyl, ethyl, propyl);
ring a is selected from 5-6 membered heteroaryl or phenyl containing 1-2 (preferably 1) N heteroatoms.
In some embodiments, the present application provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein,
R 1is selected from
Figure PCTCN2021072699-APPB-000045
Or
Figure PCTCN2021072699-APPB-000046
R 2Selected from hydrogen, a 4-9 membered heterocyclyl or amino group containing 1-3 (e.g. 1,2 or 3) heteroatoms selected from N or O or S (preferably N or O), wherein said 4-9 membered heterocyclyl or amino group is optionally substituted by 1 or 2RaSubstitution;
R aselected from hydroxy, cyano, halogen (preferably F),
Figure PCTCN2021072699-APPB-000047
C 1-3Alkyl (preferably methyl, ethyl), C1-3Alkoxy (preferably methoxy, ethoxy), di (C)1-6Alkyl) amino, or by a hydroxy or C group1-3Alkoxy (preferably methoxy, ethoxy) substituted C1-4An alkyl group;
R 3is-OCF2Cl;
R 4Selected from hydrogen, C1-3Alkyl (preferably methyl, ethyl), C3Cycloalkyl, or a 6-membered heterocyclic group containing 1 heteroatom selected from N or O (preferably O heteroatom), wherein C1-3Alkyl is optionally substituted by one or more RbSubstitution;
R bis selected from deuterium;
R 5is selected from C1-3Alkyl (preferably methyl, ethyl);
ring a is a 5-membered heteroaryl or phenyl group containing 1-2 (preferably 1) N heteroatoms.
In some embodiments, the present application provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein,
R 1is selected from
Figure PCTCN2021072699-APPB-000048
Or
Figure PCTCN2021072699-APPB-000049
R 2Is selected from
Figure PCTCN2021072699-APPB-000050
Figure PCTCN2021072699-APPB-000051
Figure PCTCN2021072699-APPB-000052
R 3Is selected from-OCF3or-OCF2Cl;
R 4Selected from hydrogen, methyl, ethyl, propylCyclopropyl, hydroxymethyl, hydroxyethyl, hydroxypropyl, hydroxy 2-methylpropyl, methoxymethyl, methoxyethyl, methoxypropyl, ethoxymethyl, ethoxyethyl, ethoxypropyl, propoxymethyl, propoxyethyl, propoxypropyl,
Figure PCTCN2021072699-APPB-000053
Wherein methyl, ethyl and propyl are optionally substituted with one or more deuterium;
R 5selected from methyl, ethyl and propyl;
ring a is selected from pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl or phenyl.
In some embodiments, the present application provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein,
R 1is selected from
Figure PCTCN2021072699-APPB-000054
Or
Figure PCTCN2021072699-APPB-000055
R 2Is selected from
Figure PCTCN2021072699-APPB-000056
Figure PCTCN2021072699-APPB-000057
R 3is-OCF2Cl;
R 4Selected from hydrogen, methyl, d3-methyl, cyclopropyl or
Figure PCTCN2021072699-APPB-000058
R 5Is selected from methyl;
ring A is selected from pyrrolyl or phenyl.
In the present application, there are also embodiments derived from any combination of the above variables.
In another aspect, the present application provides a compound, or a pharmaceutically acceptable salt thereof,
Figure PCTCN2021072699-APPB-000059
Figure PCTCN2021072699-APPB-000060
Figure PCTCN2021072699-APPB-000061
Figure PCTCN2021072699-APPB-000062
in another aspect, the present application provides a compound, or a pharmaceutically acceptable salt thereof,
Figure PCTCN2021072699-APPB-000063
Figure PCTCN2021072699-APPB-000064
in another aspect, the present application also provides a pharmaceutical composition comprising a compound of the present application, or a pharmaceutically acceptable salt thereof, as described above. In some embodiments, the pharmaceutical compositions of the present application further comprise a pharmaceutically acceptable excipient.
In another aspect, the present application also provides a method for treating and/or preventing BCR-ABL related diseases, comprising administering to a mammal, preferably a human, in need thereof a therapeutically effective amount of the above-described compound of the present application or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof.
In another aspect, the present application also provides a use of the above compound or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof of the present application in the preparation of a medicament for treating and/or preventing BCR-ABL related diseases.
In another aspect, the present application also provides the use of the above-mentioned compound of the present application or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the treatment and/or prevention of BCR-ABL related diseases.
In another aspect, the present application also provides a compound of the present application, a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof for treating and/or preventing BCR-ABL related diseases.
In some embodiments, the BCR-ABL associated disease is selected from cancer, for example leukemia (such as chronic myeloid leukemia).
Technical effects
The compound or the pharmaceutically acceptable salt thereof can show a proliferation inhibition effect on K562 cells and Ba/F3 cells transfected by BCR-ABL T315I, has good cell activity, and can show good in vivo pharmacokinetic properties.
Definition of
The following terms used in the present application have the following meanings, unless otherwise specified. A particular term should not be considered as ambiguous or unclear without special definition, but rather construed according to ordinary meaning in the art. When a trade name appears herein, it is intended to refer to its corresponding commodity or its active ingredient.
When a covalent bond in some structural unit or group is not attached to a particular atom in this application, it is meant that the covalent bond can be attached to any atom in the structural unit or group, as long as the valence bond attachment rules are not violated.
The term "substituted" means that any one or more hydrogen atoms on a particular atom is replaced with a substituent, so long as the valence of the particular atom is normal and the substituted compound is stable. When the substituent is oxo (i.e., ═ O), meaning that two hydrogen atoms are substituted, oxo does not occur on the aryl.
The terms "optionally" or "optionally" mean that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. When a group is "optionally" substituted, it means that the group may be unsubstituted or substituted, e.g., ethyl "optionally" is substituted with halo, meaning that ethyl may be unsubstituted (CH)2CH 3) Monosubstituted (e.g. CH)2CH 2F) Polysubstituted (e.g. CHFCH)2F、CH 2CHF 2Etc.) or completely substituted (CF)2CF 3). It will be appreciated by those skilled in the art that any group containing one or more substituents will not incorporate any substitution or substitution pattern which is sterically impossible and/or cannot be synthesized.
Herein Cm-nIt is the moiety that has an integer number of carbon atoms in the given range. E.g. "C1-6By "is meant that the group can have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, or 6 carbon atoms. E.g. C1-3Meaning that the group may have 1 carbon atom, 2 carbon atoms, 3 carbon atoms.
When any variable (e.g., R) occurs more than one time in the composition or structure of a compound, its definition in each case is independent. Thus, for example, if a group is substituted with 2R, then there are separate options for each R.
When the number of one linking group is 0, e.g. - (CH)2) 0-, denotes that the linking group is a covalent bond。
When one of the variables is selected from a covalent bond, it means that the two groups to which it is attached are directly linked, for example, in A-L '-Z where L' represents a covalent bond, it means that the structure is actually A-Z.
When a substituent's bond is cross-linked to two atoms on a ring, such substituent may be bonded to any atom on the ring. For example, a structural unit
Figure PCTCN2021072699-APPB-000065
Meaning that it may be substituted at any position on the cyclohexyl or cyclohexadiene.
The term "halo" or "halogen" refers to fluorine, chlorine, bromine and iodine.
The term "hydroxy" refers to an-OH group.
The term "amino" refers to-NH2A group.
The term "cyano" refers to the group — CN.
The term "alkyl" refers to a group of formula CnH 2n+1A hydrocarbon group of (2). The alkyl group may be linear or branched. For example, the term "C1-6Alkyl "means an alkyl group having 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, neopentyl, hexyl, 2-methylpentyl, and the like). Similarly, the alkyl portion (i.e., alkyl) of alkoxy, alkylamino, dialkylamino, alkylsulfonyl and alkylthio groups have the same definitions as above. Also for example, the term "C1-3Alkyl "refers to alkyl groups containing 1 to 3 carbon atoms (e.g., methyl, ethyl, propyl, and isopropyl).
The term "alkoxy" refers to-O-alkyl.
The term "alkylamino" refers to-NH-alkyl.
The term "dialkylamino" refers to-N (alkyl)2
The term "cycloalkyl" means fully saturated and may be in the form ofMonocyclic, bridged or spiro carbon rings. Unless otherwise indicated, the carbocycle is typically a 3 to 10 membered ring, such as a 3,4, 5, 6, 7, 8, 9 or 10 membered ring. Non-limiting examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornyl (bicyclo [2.2.1 ] n]Heptyl), bicyclo [2.2.2]Octyl, adamantyl, bicyclo [1.1.1]Pent-1-yl and the like. E.g. C3-4Cycloalkyl groups include cyclopropyl and cyclobutyl.
The term "heterocyclyl" refers to a non-aromatic ring that is fully saturated or partially unsaturated (but not fully unsaturated heteroaromatic) and may exist as a single ring, a bridged ring, or a spiro ring. Unless otherwise indicated, the heterocyclic ring is typically a 3-to 10-membered ring containing 1 to 3 heteroatoms (preferably 1 or 2 heteroatoms) independently selected from sulfur, oxygen and/or nitrogen, or a 4-to 6-membered ring, for example, a 4-membered ring, a 5-membered ring, a 6-membered ring, a 7-membered ring, an 8-membered ring or a 9-membered ring. Non-limiting examples of heterocyclyl groups include, but are not limited to, oxiranyl, tetrahydrofuryl, dihydrofuranyl, 3, 4-dihydropyranyl, 3, 6-dihydropyranyl, pyrrolidinyl, N-methylpyrrolidinyl, dihydropyrrolyl, piperidinyl, piperazinyl, pyrazolidinyl, 4H-pyranyl, morpholinyl, thiomorpholinyl, tetrahydrothienyl, 2-oxa-7-azaspiro [3.5] nonanyl, 2-oxa-6-azaspiro [3.3] heptanyl, and the like.
The term "heterocycloalkyl" refers to a cyclic group that is fully saturated and may exist as a single ring, a bridged ring, or a spiro ring. Unless otherwise indicated, the heterocyclic ring is typically a 3 to 10 membered ring, or a 4 to 6 membered ring containing 1 to 3 heteroatoms (preferably 1 or 2 heteroatoms) independently selected from sulfur, oxygen and/or nitrogen. Examples of 3-membered heterocycloalkyl include, but are not limited to, oxiranyl, thietanyl, cycloazenyl, non-limiting examples of 4-membered heterocycloalkyl include, but are not limited to, azetidinyl, oxetanyl, thiabutinyl, examples of 5-membered heterocycloalkyl include, but are not limited to, tetrahydrofuranyl, tetrahydrothienyl, pyrrolidinyl, isoxazolidinyl, oxazolidinyl, isothiazolidinyl, thiazolidinyl, imidazolidinyl, examples of tetrahydropyrazolyl, 6-membered heterocycloalkyl include, but are not limited to, piperidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, morpholinyl, piperazinyl, 1, 4-thialkyl, 1, 4-dioxanyl, thiomorpholinyl, 1, 3-dithianyl, 1, 4-dithianyl, and examples of 7-membered heterocycloalkyl include, but are not limited to, azepanyl, oxepanyl, thiepanyl. Monocyclic heterocycloalkyl groups having 5 or 6 ring atoms are preferred.
The term "heterocycloalkenyl" refers to a non-aromatic ring that is partially unsaturated (but not fully unsaturated heteroaromatic) and may exist as a single ring, a bridged ring, or a spiro ring. Unless otherwise indicated, the heterocyclic ring is typically a 3 to 10 membered ring containing 1 to 3 heteroatoms (preferably 1 or 2 heteroatoms) independently selected from sulfur, oxygen and/or nitrogen, or a 4 to 6 membered ring. Non-limiting examples of heterocycloalkenyl include, but are not limited to, dihydrofuranyl, 3, 4-dihydropyranyl, 3, 6-dihydropyranyl, dihydropyrrolyl, 4H-pyranyl, and the like.
The term "spiroheterocyclyl" refers to a fully saturated or partially unsaturated (but not fully saturated) spirocyclic ring in which one or more ring atoms are heteroatoms (preferably 1 or 2 heteroatoms) selected from sulfur, oxygen and/or nitrogen, the remaining ring atoms being carbon. Preferably 6 to 14, more preferably 6 to 10. Spiroheterocycles are classified as mono-, di-or polyspiroheterocycles, preferably mono-or di-spiroheterocycles, more preferably 4-or 5-or 6-membered mono-spiroheterocycles, depending on the number of spiro atoms shared between the rings. Non-limiting examples of spiroheterocycles include
Figure PCTCN2021072699-APPB-000066
The term "spiroheterocycloalkyl" refers to a completely saturated spiroheterocyclyl group.
The term "heteroaryl" refers to a monocyclic or fused polycyclic ring system containing as ring atoms at least one (e.g., 1-4, 1-3, or 1-2, e.g., 1,2, or 3) heteroatom selected from N, O, S, the remaining ring atoms being C, and having at least one aromatic ring. Preferred heteroaryl groups have a single 5 to 8 membered ring, or multiple fused rings containing 6 to 14, especially 6 to 10 ring atoms. Non-limiting examples of heteroaryl groups include, but are not limited to, pyrrolyl, furanyl, thienyl, imidazolyl, oxazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, quinolinyl, isoquinolinyl, tetrazolyl, triazolyl, triazinyl, benzofuranyl, benzothienyl, indolyl, isoindolyl, and the like.
The term "treating" means administering a compound or formulation described herein to ameliorate or eliminate a disease or one or more symptoms associated with the disease, and includes:
(i) inhibiting the disease or disease state, i.e., arresting its development;
(ii) alleviating the disease or condition, i.e., causing regression of the disease or condition.
The term "preventing" means administering a compound or formulation described herein to prevent a disease or one or more symptoms associated with the disease, and includes: prevention of a disease or condition occurs in a mammal, particularly when such mammal is susceptible to the disease condition, but has not yet been diagnosed as having the disease condition.
The term "therapeutically effective amount" means an amount of a compound of the present application that (i) treats or prevents a particular disease, condition, or disorder, (ii) alleviates, ameliorates, or eliminates one or more symptoms of a particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of a particular disease, condition, or disorder described herein. The amount of a compound of the present application that constitutes a "therapeutically effective amount" varies depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but can be routinely determined by those skilled in the art with their own knowledge and this disclosure.
The term "pharmaceutically acceptable" is intended to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
As the pharmaceutically acceptable salt, for example, a metal salt, an ammonium salt, a salt with an organic base, a salt with an inorganic acid, a salt with an organic acid, a salt with a basic or acidic amino acid, and the like can be mentioned.
The term "pharmaceutical composition" refers to a mixture of one or more compounds of the present application or salts thereof and pharmaceutically acceptable excipients. The purpose of the pharmaceutical composition is to facilitate administration of the compounds of the present application to an organism.
The term "pharmaceutically acceptable adjuvants" refers to those adjuvants which do not have a significant irritating effect on the organism and do not impair the biological activity and properties of the active compound. Suitable adjuvants are well known to those skilled in the art, such as carbohydrates, waxes, water-soluble and/or water-swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water, and the like.
The word "comprise" or "comprises", and variations such as "comprises" or "comprising", will be understood in an open, non-exclusive sense, i.e., "including but not limited to", meaning that elements, components, and steps other than those listed may be included.
The compounds of the present invention may exist in specific geometric or stereoisomeric forms. The present invention contemplates all such compounds, including cis and trans isomers, (-) -and (+) -enantiomers, (R) -and (S) -enantiomers, diastereomers, (D) -isomers, (L) -isomers, as well as racemic and other mixtures thereof, such as enantiomerically or diastereomerically enriched mixtures, all of which are within the scope of the present invention. Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All such isomers, as well as mixtures thereof, are included within the scope of the present invention.
Unless otherwise indicated, "(D)" or "(+)" means dextrorotation, "(L)" or "(-) -means levorotation," (DL) "or" (±) "means racemization.
Using solid wedge keys, unless otherwise indicated
Figure PCTCN2021072699-APPB-000067
And wedge dotted bond
Figure PCTCN2021072699-APPB-000068
Showing the absolute configuration of a solid centre, by means of straight solid keys
Figure PCTCN2021072699-APPB-000069
And straight dotted line bond
Figure PCTCN2021072699-APPB-000070
Showing the relative configuration of the centres of solids, by wavy lines
Figure PCTCN2021072699-APPB-000071
Representing solid-line keys of wedge shape
Figure PCTCN2021072699-APPB-000072
Or wedge dotted bond
Figure PCTCN2021072699-APPB-000073
Or by wavy lines
Figure PCTCN2021072699-APPB-000074
Indicating straight solid-line keys
Figure PCTCN2021072699-APPB-000075
And straight dotted bond
Figure PCTCN2021072699-APPB-000076
Optically active (R) -and (S) -isomers as well as D and L isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If one of the enantiomers of a compound of the invention is desired, it can be prepared by asymmetric synthesis or derivatization with a chiral auxiliary, wherein the resulting diastereomeric mixture is separated and the auxiliary group is cleaved to provide the pure desired enantiomer. Alternatively, when the molecule contains a basic functional group (e.g., amino) or an acidic functional group (e.g., carboxyl), diastereomeric salts are formed with an appropriate optically active acid or base, followed by diastereomeric resolution by conventional methods known in the art, and the pure enantiomers are recovered. Furthermore, separation of enantiomers and diastereomers is typically accomplished by using chromatography employing a chiral stationary phase, optionally in combination with chemical derivatization (e.g., carbamate formation from amines).
The compounds of the present invention may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. For example, the compounds may be labelled with radioactive isotopes, such as tritium (A), (B), (C) and C)3H) Iodine-125 (125I) Or C-14(14C) .1. the As another example, deuterium can be substituted for hydrogen with deuterium to form a deuterated drug, such as d3The-methyl represents that three hydrogen atoms on the methyl are all replaced by deuterium atoms, the bond formed by deuterium and carbon is firmer than the bond formed by common hydrogen and carbon, and compared with an un-deuterated drug, the deuterated drug has the advantages of reducing toxic and side effects, increasing the stability of the drug, enhancing the curative effect, prolonging the biological half-life period of the drug and the like. All isotopic variations of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present invention. "optional" or "optionally" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
The present application also includes isotopically-labeled compounds of the present application, which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Can be incorporated into this applicationExamples of isotopes of compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine and chlorine, such as respectively2H、 3H、 11C、 13C、 14C、 13N、 15N、 15O、 17O、 18O、 31P、 32P、 35S、 18F、 123I、 125I and36cl, and the like.
Certain isotopically-labelled compounds of the present application (e.g. with3H and14c-labeled ones) can be used in compound and/or substrate tissue distribution assays. Tritiated (i.e. by tritiation)3H) And carbon-14 (i.e.14C) Isotopes are particularly preferred for their ease of preparation and detectability. Positron emitting isotopes, such as15O、 13N、 11C and18f can be used in Positron Emission Tomography (PET) studies to determine substrate occupancy. Isotopically labeled compounds of the present application can generally be prepared by following procedures analogous to those disclosed in the schemes and/or in the examples below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
In addition, heavier isotopes are used (such as deuterium (i.e., deuterium)2H) Substitution may provide certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements), and thus may be preferred in certain circumstances, wherein deuterium substitution may be partial or complete, partial deuterium substitution meaning that at least one hydrogen is substituted with at least one deuterium, all such forms of the compounds being encompassed within the scope of the present application.
The compounds of the present application may be asymmetric, e.g., having one or more stereoisomers. Unless otherwise indicated, all stereoisomers include, for example, enantiomers and diastereomers. The compounds of the present application containing asymmetric carbon atoms can be isolated in optically active pure form or in racemic form. The optically active pure form can be resolved from a racemic mixture or synthesized by using chiral starting materials or chiral reagents.
The pharmaceutical compositions of the present application can be prepared by combining the compounds of the present application with suitable pharmaceutically acceptable excipients, for example, can be formulated into solid, semi-solid, liquid or gaseous formulations, such as tablets, pills, capsules, powders, granules, ointments, emulsions, suspensions, suppositories, injections, inhalants, gels, microspheres, aerosols, and the like.
Typical routes of administration of a compound of the present application or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof include, but are not limited to, oral, rectal, topical, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, intramuscular, subcutaneous, intravenous administration.
The pharmaceutical compositions of the present application can be manufactured by methods well known in the art, such as conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, lyophilizing, and the like.
In some embodiments, the pharmaceutical composition is in an oral form. For oral administration, the pharmaceutical compositions may be formulated by mixing the active compounds with pharmaceutically acceptable excipients well known in the art. These adjuvants enable the compounds of the present application to be formulated as tablets, pills, lozenges, dragees, capsules, liquids, gels, slurries, suspensions and the like, for oral administration to a patient.
Solid oral compositions may be prepared by conventional mixing, filling or tableting methods. For example, it can be obtained by the following method: the active compounds are mixed with solid adjuvants, optionally the mixture obtained is milled, if desired with further suitable adjuvants, and the mixture is then processed to granules, to give tablets or dragee cores. Suitable excipients include, but are not limited to: binders, diluents, disintegrants, lubricants, glidants, sweeteners or flavoring agents, and the like.
The pharmaceutical compositions may also be adapted for parenteral administration, as sterile solutions, suspensions or lyophilized products in suitable unit dosage forms.
Therapeutic dosages of the compounds of the present application may be determined, for example, by: the particular use of the treatment, the mode of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of the compound of the present application in the pharmaceutical composition may not be fixed, depending on a variety of factors including dosage, chemical properties (e.g., hydrophobicity), and the route of administration. For example, the compounds of the present application can be provided for parenteral administration by a physiological buffered aqueous solution containing about 0.1-10% w/v of the compound. Some typical dosage ranges are from about 1. mu.g/kg to about 1g/kg body weight/day. In certain embodiments, the dosage range is from about 0.01mg/kg to about 100mg/kg of body weight per day. The dosage will likely depend on such variables as the type and extent of progression of the disease or disorder, the general health status of the particular patient, the relative biological efficacy of the selected compound, the excipient formulation and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
In this document, unless the context clearly dictates otherwise, singular terms encompass plural referents and vice versa.
All patents, patent applications, and other established publications are herein expressly incorporated by reference for the purpose of description and disclosure. These publications are provided solely for their disclosure prior to the filing date of the present application. All statements as to the date of these documents or representation as to the contents of these documents is based on the information available to the applicant and does not constitute any admission as to the correctness of the dates of these documents or the contents of these documents. Moreover, any reference herein to such publications in any country does not constitute an admission that the publications form part of the common general knowledge in the art.
The compounds of the present application may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combinations thereof with other chemical synthetic methods, and equivalents thereof known to those skilled in the art, with preferred embodiments including, but not limited to, the examples of the present application.
The chemical reactions of the embodiments herein are carried out in a suitable solvent that is compatible with the chemical changes of the present application and the reagents and materials required therefor. In order to obtain the compounds of the present application, it is sometimes necessary for a person skilled in the art to modify or select the synthesis steps or reaction schemes based on the existing embodiments.
An important consideration in the art of synthetic route planning is the selection of suitable protecting Groups for reactive functional Groups (e.g., amino Groups as used herein), for example, see Greene's Protective Groups in Organic Synthesis (4th Ed.) Hoboken, New Jersey: John Wiley & Sons, Inc.
The compounds of general formula (I) of the present application can be prepared by a person skilled in the art of organic synthesis by the following route:
synthesis of intermediate (i):
Figure PCTCN2021072699-APPB-000077
wherein,
R 2、R 3is as defined above, and R2Is not hydrogen.
Synthesis of intermediate (ii):
Figure PCTCN2021072699-APPB-000078
wherein,
R 3as defined above, R2Is hydrogen.
Synthesis of intermediate (iii):
the first scheme is as follows:
Figure PCTCN2021072699-APPB-000079
scheme II:
Figure PCTCN2021072699-APPB-000080
wherein,
R 4、R 5and ring a is as defined above.
Preparation of the target compound:
Figure PCTCN2021072699-APPB-000081
wherein,
R 2、R 3、R 4、R 5and ring a is as defined above.
The following abbreviations are used in this application:
SOCl 2represents thionyl chloride; TEA stands for triethylamine; oxone represents Oxone complex salt; NBS represents N-bromosuccinimide; pd (PPh)3) 4Represents tetrakis (triphenylphosphine) palladium; PdCl2(dppf) represents 1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride; DCM represents dichloromethane; DMSO represents dimethyl sulfoxide; DMF stands for N, N-dimethylformamide.
Detailed Description
For clarity, the invention is further illustrated by examples, which are not intended to limit the scope of the application. It will be apparent to those skilled in the art that various changes and modifications can be made in the specific embodiments of the invention without departing from the spirit and scope of the invention. All reagents used herein were commercially available and were used without further purification.
Example 1: preparation of Compound 1
Figure PCTCN2021072699-APPB-000082
Figure PCTCN2021072699-APPB-000083
Step A: preparation of Compound 1-1
Adding 300mL of toluene and 5-bromo-6-chloronicotinic acid (15.0g) into a 500mL three-necked bottle in sequence, dropwise adding thionyl chloride (14.79g) into the system at room temperature, heating to 70-80 ℃ after the addition, reacting for 4 hours, stopping the reaction, and concentrating under reduced pressure to obtain brown oily matter; dichloromethane (300mL) was added thereto, stirring was turned on and 4- (chlorodifluoromethoxy) aniline (12.28g) was added dropwise to the system; after the dropwise addition, triethylamine (12.58g) was added dropwise; after dropping, the reaction is carried out for 4h at room temperature. Adding 100mL of saturated sodium bicarbonate water solution into the reaction solution, stirring for 10min, filtering and collecting a filter cake; and separating the mother liquor to obtain an organic phase, adding 100mL of saturated sodium chloride aqueous solution, stirring, separating to obtain the organic phase, adding the filter cake into the organic phase, concentrating under reduced pressure to obtain a residue, and purifying by column chromatography to obtain 20.44g of the compound 1-1.
And B, step B: preparation of Compounds 1-2
Adding isopropanol (10mL), the compound 1-1(2.0g) obtained in the step A, (R) -3-pyrrolidinol (0.498g), N, N-diisopropylethylamine (1.229g) and magnetons into a 50mL sealed tube in sequence, sealing the tube, and placing the sealed tube into a microwave reactor to react for 30min at 130 ℃. Concentrating the reaction solution under reduced pressure to obtain brown residue, adding anhydrous ethanol (4mL) into the brown residue, pulping for 10min, filtering, leaching the filter cake with a small amount of anhydrous ethanol, and collecting the filter cake; the filter cake was placed in a vacuum oven and dried under vacuum at 50 ℃ to constant weight to obtain 1.744g of compound 1-2.
MS(ESI,[M+H] +)m/z:462.0/464.0.
And C: preparation of Compounds 1-3
To a 25mL single-neck flask were added in sequence dimethyl sulfoxide (5mL), 2, 5-dibromopyridine (100mg), 2-methyl-2-mercapto urea sulfate (120mg), and cesium carbonate (550mg), and after completion of the addition, the temperature was raised to 75-85 ℃ for reaction for 4 hours. Cooling to room temperature, filtering to obtain light yellow mother liquor, adding 20mL of water, extracting with 10mL of dichloromethane for 2 times, mixing organic phases, adding 10mL of saturated sodium chloride aqueous solution, stirring, washing, separating liquid to obtain an organic phase, drying with anhydrous sodium sulfate, performing suction filtration, and concentrating under reduced pressure to obtain 153mg of compound 1-3; the product was used in the next reaction without purification.
MS(ESI,[M+H] +)m/z:203.9.
Step D: preparation of Compounds 1-4
To a 250mL three-necked flask were added isopropanol (90mL) and water (30mL) in this order, and 5-bromo-2- (methylthio) pyridine (3.2g) prepared according to the method in step C was added to the system with stirring, followed by addition of Oxone (Oxone, complex salt of potassium hydrogen persulfate, 22.17g) in portions. After the addition, the reaction was kept at room temperature. Cooling to room temperature, filtering to obtain white mother liquor, and concentrating under reduced pressure; adding 50mL of ethyl acetate and 30mL of water for washing, extracting the water phase for 1 time by using 50mL of ethyl acetate, combining organic phases, adding 30mL of saturated sodium chloride aqueous solution for washing, separating liquid to obtain an organic phase, and concentrating under reduced pressure to obtain 3.70g of a compound 1-4.
MS(ESI,[M+H] +)m/z:224.1.
Step E: preparation of Compounds 1-5
And (3) sequentially adding 1, 4-dioxane (10mL), the 5-bromo-2-methylsulfonylpyridine (100mg) obtained in the step D, the pinacol diboron diboride (164mg), potassium acetate (85mg) and the [1,1' -bis (diphenylphosphine) ferrocene ] palladium dichloride dichloromethane complex (10mg) into a 25mL three-necked flask, reacting for 4 hours after the addition and nitrogen replacement, heating to 80-90 ℃, stopping the reaction, cooling to room temperature, and directly using for the next reaction without separation and purification.
Step F: preparation of Compound 1
To the reaction solution obtained in the above step E, the compound 1-2(163mg) obtained in the above step B, potassium carbonate (146mg), deionized water (1.5mL), tetrakis (triphenylphosphine) palladium (10mg) were added in this order; after the addition, nitrogen is replaced, and the temperature is raised to 80-90 ℃ for reaction for 4 h. Cooling to room temperature, filtering the reaction, collecting mother liquor, adding 15mL ethyl acetate, stirring, separating liquid, adding 15mL saturated sodium chloride aqueous solution, stirring, separating liquid, and purifying by column chromatography to obtain 60mg of compound 1.
1H NMR(500MHz,DMSO-d 6)δ10.22(s,1H),8.88(m,1H),8.81(d,J=2.3Hz,1H),8.17-8.11(m,3H),7.93–7.82(m,2H),7.35(d,J=8.8Hz,2H),4.91(d,J=3.5Hz,1H),4.27–4.18(m,1H),3.46–3.37(m,1H),3.36(s,3H),3.28–3.19(m,2H),2.86(d,J=11.4Hz,1H),1.91–1.81(m,1H),1.81–1.71(m,1H).
MS(ESI,[M+H] +)m/z:539.2/541.1.
Example 2: preparation of Compound 2
Figure PCTCN2021072699-APPB-000084
Step A: preparation of Compound 2-1
Trifluoromethyl benzene (30mL), 5-bromo-2-methylnicotinic acid ethyl ester (3.0g), N-bromosuccinimide (2.62g) and azobisisobutyronitrile (0.395g) are sequentially added into a 50mL single-neck bottle, and after the addition is finished, the temperature is raised to 70-80 ℃ for reaction for 6 hours after nitrogen replacement. After cooling to room temperature, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue, which was purified by column chromatography to give 1.731g of Compound 2-1.
MS(ESI,[M+H] +)m/z:321.9/323.9.
And B: preparation of Compound 2-2
Adding 30mL of ammonia isopropanol solution (2mol/L) into a 100mL single-neck bottle in sequence to obtain a compound 2-1(1.484g) obtained in the step A, and reacting at room temperature for 6h after the addition is finished; concentrating the reaction solution under reduced pressure; the obtained residue was subjected to column chromatography to give 768mg of the compound 2-2.
1H NMR(500MHz,DMSO-d 6):δ8.93(s,1H),8.87(s,1H),8.28(s,1H),4.39(s,2H).
And C: preparation of Compounds 2-3
Referring to the procedure of step E of example 1, a reaction solution containing compound 2-3 was prepared using compound 2-2 obtained in step B, cooled to room temperature, and used in the next reaction without separation and purification.
Step D: preparation of Compound 2
Compound 2 was prepared by the method of step F of example 1 using the reaction mixture obtained in step C above and compound 1-2 obtained in step B of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.19(s,1H),8.92(s,1H),8.81-8.76(m,2H),8.11–8.05(m,2H),7.86(d,J=8.8Hz,2H),7.33(d,J=8.6Hz,2H),4.87(d,J=3.4Hz,1H),4.51(s,2H),4.21–4.16(m,1H),3.41–3.39(m,1H),3.25–3.15(m,2H),2.88(d,J=11.4Hz,1H),1.87–1.79(m,1H),1.76–1.69(m,1H).
MS(ESI,[M+H] +)m/z:516.2/518.2.
Example 3: preparation of Compound 3
Figure PCTCN2021072699-APPB-000085
Compound 3 was prepared by the method referenced to step F of example 1, using compound 1-2 and 3-methanesulfonylphenylboronic acid, obtained in step B of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.23(s,1H),8.78(d,J=2.3Hz,1H),8.06(d,J=2.4Hz,1H),7.96–7.91(m,2H),7.90–7.84(m,2H),7.79–7.73(m,2H),7.34(d,J=8.7Hz,2H),4.86(d,J=3.3Hz,1H),4.19(q,J=3.6Hz,1H),3.45–3.36(m,1H),3.29(s,3H),3.27–3.22(m,1H),3.21–3.14(m,1H),2.85(d,J=11.4Hz,1H),1.88–1.79(m,1H),1.77–1.70(m,1H).
MS(ESI,[M+H] +)m/z:538.1/540.1.
Example 4: preparation of Compound 4
Figure PCTCN2021072699-APPB-000086
Step A: preparation of Compound 4-1
Compound 4-1 was prepared by reacting compound 1-1 with N-methylpiperazine according to the preparation method of step B of example 1.
MS(ESI,[M+H] +)m/z:475.1/477.1.
And B: preparation of Compound 4
Compound 4 was prepared by reacting the reaction mixture containing Compound 2-3 obtained in step C of example 2 with Compound 4-1 of step A described above, according to the preparation method of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.37(s,1H),9.05(s,1H),8.94(s,1H),8.83(d,J=1.8Hz,1H),8.35(s,1H),8.23(s,1H),7.90(d,J=8.9Hz,2H),7.39(d,J=8.7Hz,2H),4.54(s,2H),3.17(t,J=4.7Hz,4H),2.28(t,J=4.7Hz,4H),2.17(s,3H).
MS(ESI,[M+H] +)m/z:529.3.
Example 5: preparation of Compound 5
Figure PCTCN2021072699-APPB-000087
Step A: preparation of Compound 5-1
Compound 5-1 was prepared by reacting compound 1-1 with morpholine according to the procedure for the preparation of step B of example 1.
MS(ESI,[M+H] +)m/z:462.0/464.0.
And B: preparation of Compound 5
Compound 5 was prepared by reacting the reaction mixture containing Compound 2-3 obtained in step C of example 2 with Compound 5-1 of step A described above, according to the preparation method of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.29(s,1H),8.98(s,1H),8.85(s,1H),8.75(s,1H),8.28(s,1H),8.15(s,1H),7.80(d,J=8.5Hz,2H),7.29(d,J=8.6Hz,2H),4.44(s,2H),3.63–3.36(m,4H),3.18–2.89(m,4H).
MS(ESI,[M+H] +)m/z:516.2.
Example 6: preparation of Compound 6
Figure PCTCN2021072699-APPB-000088
Step A: preparation of Compound 6-1
Referring to the preparation method of step B of example 1, compound 6-1 was prepared by reacting compound 1-1 with (R) -3-dimethylaminopyrrolidine.
MS(ESI,[M+H] +)m/z:489.3/491.3.
And B: preparation of Compound 6
Referring to the preparation method of step F of example 1, compound 6 was prepared by reacting compound 6-1 of step A with the reaction solution containing compound 2-3 obtained in step C of example 2.
1H NMR(500MHz,DMSO-d 6)δ10.23(s,1H),8.92(s,1H),8.86–8.74(m,2H),8.18–8.06(m,2H),7.87(d,J=9.0Hz,2H),7.34(d,J=8.7Hz,2H),4.51(s,2H),3.26–3.09(m,2H),3.09–2.93(m,1H),2.77–2.60(m,1H),2.12(s,6H),2.03–1.94(m,1H),1.72–1.59(m,2H).
MS(ESI,[M+H] +)m/z:543.3.
Example 7: preparation of Compound 7
Figure PCTCN2021072699-APPB-000089
Step A: preparation of Compound 7-1
Referring to the preparation method of step B of example 1, compound 7-1 is prepared by reacting compound 1-1 with azetidine.
MS(ESI,[M+H] +)m/z:448.1/450.1.
And B: preparation of Compound 7
Referring to the preparation method of step F of example 1, compound 7 was prepared by reacting the reaction solution containing compound 2-3 obtained in step C of example 2 with compound 7-1 of step A described above.
1H NMR(500MHz,DMSO-d 6)δ10.24(s,1H),8.93(s,1H),8.83(s,1H),8.79(s,1H),8.17–8.07(m,2H),7.87(d,J=8.4Hz,2H),7.35(d,J=8.2Hz,2H),5.57(d,J=5.8Hz,1H),4.52(s,2H),4.45–4.31(m,1H),3.94–3.81(m,2H),3.53–3.43(m,2H).
MS(ESI,[M+H] +)m/z:502.1.
Example 8: preparation of Compound 8
Figure PCTCN2021072699-APPB-000090
Step A: preparation of Compound 8-1
Referring to the preparation method of step B of example 1, compound 8-1 is prepared by reacting compound 1-1 with tetrahydropyrrole.
MS(ESI,[M+H] +)m/z:446.0/448.0.
And B: preparation of Compound 8
Referring to the preparation method of step F of example 1, compound 8 was prepared by reacting the reaction solution containing compound 2-3 obtained in step C of example 2 with compound 8-1 of step A described above.
1H NMR(500MHz,DMSO-d 6)δ10.17(s,1H),8.90(s,1H),8.79(d,J=2.5Hz,1H),8.78(d,J=2.4Hz,1H),8.12–8.05(m,2H),7.92–7.77(m,2H),7.33(d,J=8.6Hz,2H),4.50(s,2H),3.19–3.04(m,4H),1.85–1.65(m,4H).
MS(ESI,[M-H] -)m/z:498.1/500.1.
Example 9: preparation of Compound 9
Figure PCTCN2021072699-APPB-000091
Step A: preparation of Compound 9-1
Compound 9-1 was prepared according to the method of example 1, step a, using 5-bromo-6-chloronicotinic acid and 4-trifluoromethoxyaniline.
MS(ESI,[M-H] -)m/z:393.0.
And B: preparation of Compound 9-2
Compound 9-2 is prepared using compound 9-1 obtained in the above step A and (R) -3-pyrrolidinol, by a method similar to step B of example 1.
MS(ESI,[M+H] +)m/z:446.1.
And C: preparation of Compound 9
Compound 9 was prepared by the method of step F of example 1 using the reaction solution containing Compound 2-3 obtained in step C of example 2 and Compound 9-2 obtained in step B above.
1H NMR(500MHz,DMSO-d 6)δ10.18(s,1H),8.92(s,1H),8.86–8.72(m,2H),8.16–8.01(m,2H),7.86(d,J=8.7Hz,2H),7.35(d,J=8.4Hz,2H),4.94–4.82(m,1H),4.52(s,2H),4.19(s,1H),3.44–3.37(m,1H),3.28–3.15(m,2H),2.95–2.83(m,1H),1.90–1.79(m,1H),1.79–1.67(m,1H).
MS(ESI,[M+H] +)m/z:500.2.
Example 10: preparation of Compound 10
Figure PCTCN2021072699-APPB-000092
Step A: preparation of Compound 10-2
Referring to the preparation method of step B of example 2, compound 10-2 was prepared by substituting an ethanol solution of methylamine (30% -35%, m/m) for an isopropanol solution of ammonia (2 mol/L).
MS(ESI,[M+H] +)m/z:227.1/229.1.
And B: preparation of Compound 10-3
Referring to the procedure of step E of example 1, a reaction solution containing compound 10-3 was prepared using compound 10-2 obtained in step A, cooled to room temperature, and used in the next reaction without separation and purification.
Step C: preparation of Compound 10
Compound 10 was prepared by the method of step F of example 1 using the reaction solution containing compound 10-3 obtained in step B above and compound 1-2 obtained in step B of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.18(s,1H),8.78(d,J=2.4Hz,1H),8.76(d,J=2.1Hz,1H),8.07(d,J=2.4Hz,1H),8.05(d,J=2.1Hz,1H),7.97–7.80(m,2H),7.34(d,J=8.7Hz,2H),4.86(d,J=3.5Hz,1H),4.53(s,2H),4.21–4.15(m,1H),3.34(s,3H),3.25–3.13(m,2H),3.07–2.95(m,1H),2.87(d,J=11.3Hz,1H),1.88–1.78(m,1H),1.76–1.68(m,1H).
MS(ESI,[M+H] +)m/z:530.2/532.2.
Example 11: preparation of Compound 11
Figure PCTCN2021072699-APPB-000093
Step A: preparation of Compound 11-1
To a 100mL single-necked flask were sequentially added 20mL of absolute ethanol, cyclopropylamine (532mg), and compound 2-1(600mg), prepared by the method of step A in example 2. After 6 hours of reaction at room temperature, the reaction mixture was concentrated under reduced pressure, and the residue obtained was subjected to column chromatography to give 405mg of compound 11-1.
1H NMR(500MHz,DMSO-d 6)δ8.86(d,J=2.3Hz,1H),8.27(d,J=2.1Hz,1H),4.43(s,2H),3.01–2.93(m,1H),0.91–0.85(m,2H),0.85–0.77(m,2H).
MS(ESI,[M+H] +)m/z:253.0/255.0.
And B: preparation of Compound 11-2
Referring to the procedure of step E of example 1, a reaction solution containing compound 11-2 was prepared using compound 11-1 obtained in the above step A, cooled to room temperature, and used in the next step without isolation and purification.
And C: preparation of Compound 11
Compound 11 was prepared by the method of step F of example 1 using the reaction solution containing Compound 11-2 obtained in step B above and Compound 1-2 obtained in step B of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.19(s,1H),8.78(d,J=2.4Hz,1H),8.77(d,J=2.1Hz,1H),8.08(d,J=2.4Hz,1H),8.06(d,J=2.1Hz,1H),7.93–7.80(m,2H),7.34(d,J=8.6Hz,2H),4.86(d,J=3.5Hz,1H),4.60(s,2H),4.21–4.15(m,1H),3.41–3.35(m,1H),3.36-3.30(m,2H),3.24–3.16(m,2H),3.16-3.11(m,3H),2.87(d,J=11.3Hz,1H),1.88–1.77(m,1H),1.76–1.68(m,1H).
MS(ESI,[M+H] +)m/z:556.3/558.2.
Example 12: preparation of Compound 12
Figure PCTCN2021072699-APPB-000094
Step A: preparation of Compound 12-1
Referring to the procedure of step A of example 1, 5-bromo-6-chloronicotinic acid was replaced with 5-bromonicotinic acid to give compound 12-1
1H NMR(500MHz,DMSO-d 6)δ10.68(s,1H),9.07(d,J=1.9Hz,1H),8.92(d,J=2.2Hz,1H),8.55(t,J=2.1Hz,1H),7.96–7.83(m,2H),7.44–7.33(m,2H).
MS(ESI,[M+H] +)m/z:377.0/379.0/381.0.
Step B preparation of Compound 12
Compound 12 was prepared by the method of step F of example 1 using the reaction solution containing compound 2-3 obtained in step C of example 2 and compound 12-1 obtained in step A above.
1H NMR(500MHz,DMSO-d 6)δ10.68(s,1H),9.28–9.21(m,2H),9.13(s,1H),8.93(s,1H),8.74(s,1H),8.56(s,1H),7.91(d,J=8.6Hz,2H),7.40(d,J=8.6Hz,2H),4.52(s,2H).
MS(ESI,[M-H] -)m/z:429.0/431.0.
Example 13: preparation of Compound 13
Figure PCTCN2021072699-APPB-000095
Step A: preparation of Compound 13-1
A25 mL single-necked flask was charged with 5mL of absolute ethanol, compound 2-1 prepared in step A of example 2 (0.15g), and 2-methoxyethylamine (0.18g) in this order, and reacted at room temperature for 6 hours. The reaction solution was concentrated under reduced pressure, and the obtained residue was subjected to column chromatography to give 0.12g of compound 13-1.
MS(ESI,[M+H] +)m/z:271.0.
And B: preparation of Compound 13-2
Referring to the procedure of step E of example 1, a reaction solution containing compound 13-2 was prepared using compound 13-1 obtained in the above step A, cooled to room temperature, and used in the next step without isolation and purification.
And C: preparation of Compound 13
Compound 13 was prepared by the method of step F of example 1 using the reaction solution containing compound 13-2 obtained in the above step B and compound 12-1 obtained in step A of example 12.
1H NMR(500MHz,DMSO-d 6)δ10.69(s,1H),9.25(s,2H),9.14(s,1H),8.75(s,1H),8.58(s,1H),7.92(d,J=9.0Hz,2H),7.41(d,J=8.8Hz,2H),4.67(s,2H),3.76-3.78(m,2H),3.61-3.64(m,2H),3.30(s,3H).
MS(ESI,[M+H] +)m/z:489.2.
Example 14: preparation of Compound 14
Figure PCTCN2021072699-APPB-000096
Step A: preparation of Compound 14-1
Compound 14-1 was prepared according to the procedure of example 13, step a, substituting starting ethanolamine for 2-methoxyethylamine.
MS(ESI,[M+H] +)m/z:257.2/259.2.
And B: preparation of Compound 14-2
Referring to the procedure of step E of example 1, using compound 14-1 obtained in the above step A, a reaction solution containing compound 14-2 was prepared, cooled to room temperature, and used in the next reaction without separation and purification.
And C: preparation of Compound 14
Compound 14 was prepared by the method of step F of example 1 using the reaction solution containing compound 14-2 obtained in step B above and compound 12-1 obtained in step A of example 12.
1H NMR(500MHz,DMSO-d 6)δ10.69(s,1H),9.25(s,2H),9.14(s,1H),8.75(s,1H),8.58(s,1H),7.92(d,J=9.1Hz,2H),7.41(d,J=9.0Hz,2H),4.87-4.89(m,1H),4.69(s,2H),3.66-3.69(m,4H).
MS(ESI,[M+H] +)m/z:475.1.
Example 15: preparation of Compound 15
Figure PCTCN2021072699-APPB-000097
Figure PCTCN2021072699-APPB-000098
Step A: preparation of Compound 15-1
Compound 15-1 was prepared according to the procedure of example 13, step a, substituting the starting material, 1-amino-2-methyl-2-propanol, for 2-methoxyethylamine.
MS(ESI,[M+H] +)m/z:285.0/287.0.
And B, step B: preparation of Compound 15-2
Referring to the procedure of step E of example 1, using compound 15-1 obtained in the above step A, a reaction solution containing compound 15-2 was prepared, cooled to room temperature, and used in the next reaction without separation and purification.
And C: preparation of Compound 15
Compound 15 was obtained by the method of step F of example 1 using the reaction solution containing compound 15-2 obtained in the above step B and compound 12-1 obtained in step A of example 12.
1H NMR(500MHz,DMSO-d 6):δ10.69(s,1H),9.25(s,2H),9.14(s,1H),8.76(s,1H),8.59(s,1H),7.93(d,J=8.5Hz,2H),7.41(d,J=8.5Hz,2H),4.80(s,2H),4.74(s,1H),3.53(s,2H),1.16(s,6H).
MS(ESI,[M-H] -)m/z:501.1.
Example 16: preparation of Compound 16
Figure PCTCN2021072699-APPB-000099
Step A: preparation of Compound 16-1
Compound 16-1 was prepared according to the procedure of example 13, step a, substituting the starting material, 4-aminotetrahydropyran, for 2-methoxyethylamine.
MS(ESI,[M+H] +)m/z:297.0/299.0.
And B: preparation of Compound 16-2
Referring to the procedure of step E of example 1, a reaction solution containing compound 16-2 was prepared using compound 16-1 obtained in the above step A, cooled to room temperature, and used in the next step without isolation and purification.
And C: preparation of Compound 16
Compound 16 was prepared by the method of step F of example 1 using the reaction solution containing compound 16-2 obtained in step B above and compound 12-1 obtained in step A of example 12.
1H NMR(500MHz,DMSO-d 6):δ11.21(s,1H),9.44(m,2H),9.33(s,1H),9.17(m,1H),8.73(d,J=2.0Hz,1H),8.05(d,J=9.0Hz,2H),7.47(d,J=8.5Hz,2H),4.72(s,2H),4.42(m,1H),4.03(m,2H),3.53(m,2H),1.95(m,2H),1.79(m,2H).
MS(ESI,[M-H] -)m/z:513.1.
Example 17: preparation of Compound 17
Figure PCTCN2021072699-APPB-000100
Step A: preparation of Compound 17-1
Compound 17-1 was prepared by the method of example 1, step B, substituting the starting material, 3-hydroxyazetidine hydrochloride, for (R) -3-pyrrolidinol.
And B, step B: preparation of Compound 17
Compound 17 was obtained by the method of step F of example 1 using the reaction solution containing compound 11-2 obtained in step B of example 11 and the reaction solution containing compound 17-1 obtained in step A.
1H NMR(500MHz,DMSO-d 6)δ10.23(s,1H),8.79(d,J=4.8Hz,2H),8.07(s,2H),7.86(d,J=8.8Hz,2H),7.35(d,J=8.5Hz,2H),5.54(d,J=6.1Hz,1H),4.53(s,2H),4.36-4.37(m,1H),3.84-3.88(m,2H),3.45-3.48(m,2H),3.02(s,1H),0.82-0.84(m,4H).
MS(ESI,[M+H] +)m/z:542.1.
Example 18: preparation of Compound 18
Figure PCTCN2021072699-APPB-000101
Step A: preparation of Compound 18-1
Compound 18-1 was prepared by the method of example 1, step B, substituting starting morpholine for (R) -3-pyrrolidinol.
And B: preparation of Compound 18
Compound 18 was prepared by the method of step F of example 1 using the reaction solution containing compound 10-3 obtained in step B of example 10 and the reaction solution containing compound 18-1 obtained in step A described above.
1H NMR(500MHz,DMSO-d 6)δ10.36(s,1H),9.02(d,J=2.0Hz,1H),8.82(s,1H),8.34(s,1H),8.21(s,1H),7.87(d,J=9.1Hz,2H),7.36(d,J=9.0Hz,2H),4.60(s,2H),3.52-3.53(m,4H),3.14(s,3H),3.13-3.14(m,4H).
MS(ESI,[M+H] +)m/z:530.1.
Example 19: preparation of Compound 19
Figure PCTCN2021072699-APPB-000102
Step A: preparation of Compound 19
Compound 19 was obtained by the method referred to step F of example 1, using the reaction solution containing compound 10-3 obtained in step B of example 10 and compound 17-1 obtained in step A of example 17.
1H NMR(500MHz,DMSO-d 6)δ10.23(s,1H),8.78-8.80(m,2H),8.09(s,2H),7.85-7.87(m,2H),7.34-7.36(m,2H),5.54(d,J=6.2Hz,1H),4.60(s,2H),4.33-4.39(m,1H),3.85-3.88(m,2H),3.44-3.48(m,2H), 3.15(s,3H).
MS(ESI,[M+H] +)m/z:516.1.
Example 20: preparation of Compound 20
Figure PCTCN2021072699-APPB-000103
Step A: preparation of Compound 20-1
Compound 20-1 was prepared by the method of step B of example 1, substituting the starting material, 2-oxa-7-azaspiro [3.5] nonane, for (R) -3-pyrrolidinol.
And B: preparation of Compound 20
Compound 20 was prepared by the method of step F of example 1 using the reaction solution containing compound 10-3 obtained in step B of example 10 and the reaction solution containing compound 20-1 obtained in step A described above.
1H NMR(500MHz,DMSO-d 6)δ10.33(s,1H),8.99(s,1H),8.79(s,1H),8.30(s,1H),8.18(s,1H),7.86(d,J=8.9Hz,2H),7.36(d,J=8.7Hz,2H),4.61(s,2H),4.27(s,4H),3.14(s,3H),3.07(s,4H),1.70(s,4H).
MS(ESI,[M+H] +)m/z:570.2.
Example 21: preparation of Compound 21
Figure PCTCN2021072699-APPB-000104
Step A: preparation of Compound 21-1
Referring to the procedure for the preparation of step B of example 1, compound 1-1 and 2-oxa-6-aza-spiro [3.3] heptane were reacted to give compound 21-1.
MS(ESI,[M+H] +)m/z:474.1.
And B, step B: preparation of Compound 21
Compound 21 was prepared by reacting the reaction mixture containing compound 10-3 obtained in step B of example 10 with compound 21-1 obtained in step A described above, according to the procedure of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.23(s,1H),8.79(s,2H),8.08(s,2H),7.86(d,J=8.0Hz,2H),7.35(d,J=7.7Hz,2H),4.62(s,2H),4.57(s,4H),3.88(s,4H),3.16(s,3H).
MS(ESI,[M+H] +)m/z:542.3/544.2.
Example 22: preparation of Compound 22
Figure PCTCN2021072699-APPB-000105
Step A: preparation of Compound 22-1
Compound 22-1 was prepared by reacting compound 1-1 with 3-cyanoazetidine hydrochloride according to the procedure for the preparation of step B of example 1.
MS(ESI,[M-H] -)m/z:455.0/457.0.
And B: preparation of Compound 22
Compound 22 can be prepared by reacting compound 22-1 obtained in step A with the reaction mixture containing compound 10-3 obtained in step B of example 10 according to the procedure of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.30(s,1H),8.90–8.74(m,2H),8.19–8.06(m,2H),7.86(d,J=8.7 Hz,2H),7.35(d,J=8.6Hz,2H),4.60(s,2H),3.96(t,J=8.8Hz,2H),3.88–3.81(m,2H),3.74–3.66(m,1H),3.15(s,3H).
MS(ESI,[M-H] -)m/z:523.2.0/525.2.
Example 23: preparation of Compound 23
Figure PCTCN2021072699-APPB-000106
Step A: preparation of Compound 23
Compound 23 was prepared by the method of step F of example 1, using the reaction mixture containing Compound 11-2 obtained in step B of example 11 and Compound 22-1 obtained in step A of example 22.
1H NMR(500MHz,DMSO-d 6)δ10.30(s,1H),9.01–8.70(m,2H),8.29–8.00(m,2H),7.86(d,J=8.7Hz,2H),7.35(d,J=8.6Hz,2H),4.53(s,2H),3.96(t,J=8.8Hz,2H),3.84(t,J=7.1Hz,2H),3.76–3.64(m,1H),3.09–2.93(m,1H),0.99–0.89(m,2H),0.88–0.77(m,2H).
MS(ESI,[M-H] -)m/z:549.2/551.1.
Example 24: preparation of Compound 24
Figure PCTCN2021072699-APPB-000107
Step A: preparation of Compound 24-1
Referring to the preparation method of step B of example 1, compound 24-1 is prepared by reacting compound 1-1 with 3-methoxyazetidine.
MS(ESI,[M-H] -)m/z:460.1/462.1.
And B, step B: preparation of Compound 24
Compound 24 was prepared by reacting the reaction mixture containing compound 10-3 obtained in step B of example 10 with compound 24-1 obtained in step A described above, according to the procedure of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.25(s,1H),8.90–8.71(m,2H),8.19–8.05(m,2H),7.86(d,J=8.6Hz,2H),7.35(d,J=8.6Hz,2H),4.60(s,2H),4.16–4.07(m,1H),3.92–3.79(m,2H),3.58–3.46(m,2H),3.14(s,3H),3.12(s,3H).
MS(ESI,[M-H] -)m/z:527.91/529.90.
Example 25: preparation of Compound 25
Figure PCTCN2021072699-APPB-000108
Step A: preparation of Compound 25-1
Referring to the preparation method of step B of example 1, compound 25-1 was prepared by reacting compound 1-1 with 2-methoxyethylamine.
MS(ESI,[M-H] -)m/z:448.08/450.05.
And B, step B: preparation of Compound 25
Compound 25 was prepared by reacting compound 25-1 obtained in step A with the reaction solution containing compound 10-3 obtained in step B of example 10 according to the procedure in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.15(s,1H),8.78(d,J=2.0Hz,1H),8.74(d,J=2.3Hz,1H),8.09(d,J=2.1Hz,1H),7.93(d,J=2.4Hz,1H),7.90–7.80(m,2H),7.33(d,J=8.7Hz,2H),6.70(t,J=5.6Hz,1H),4.60(s,2H),3.55(q,J=5.9Hz,2H),3.47(t,J=6.0Hz,2H),3.25(s,3H),3.15(s,3H).
MS(ESI,[M-H] -)m/z:516.2/518.2.
Example 26: preparation of Compound 26
Figure PCTCN2021072699-APPB-000109
Step A: preparation of Compound 26-1
Referring to the preparation method of step B of example 1, compound 26-1 was prepared by reacting compound 1-1 with 1-amino-2-methyl-2-propanol.
MS(ESI,[M-H] -)m/z:462.0.
And B: preparation of Compound 26
Compound 26 can be prepared by reacting compound 26-1 obtained in step A with the reaction solution containing compound 10-3 obtained in step B of example 10 according to the procedure in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.16(s,1H),8.85(d,J=2.1Hz,1H),8.71(d,J=2.4Hz,1H),8.17(d,J=2.1Hz,1H),7.96(d,J=2.4Hz,1H),7.89–7.78(m,2H),7.33(d,J=8.7Hz,2H),6.22(t,J=5.8Hz,1H),4.69(s,1H),4.61(s,2H),3.42(d,J=5.8Hz,2H),3.15(s,3H),1.10(s,6H).
MS(ESI,[M-H] -)m/z:530.3/532.3.
Example 27: preparation of Compound 27
Figure PCTCN2021072699-APPB-000110
Step A: preparation of Compound 27-1
Referring to the preparation method of step B of example 1, compound 27-1 was prepared by reacting compound 1-1 with N-methyl-2-hydroxyethylamine.
1H NMR(500MHz,DMSO-d 6)δ10.68(s,1H),9.00(d,J=2.3Hz,1H),8.98(d,J=2.0Hz,1H),8.95(s,1H),8.53(d,J=2.3Hz,1H),8.34(d,J=2.0Hz,1H),7.88(d,J=9.1Hz,2H),7.40(d,J=8.9Hz,2H),4.55(s,2H).
And B: preparation of Compound 27
Compound 27 was prepared by reacting compound 27-1 obtained in the above step A with the reaction solution containing compound 10-3 obtained in step B of example 10, according to the procedure of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.25(s,1H),8.85(s,1H),8.76(s,1H),8.17(s,1H),8.12(s,1H),7.86(d,J=8.7Hz,2H),7.35(d,J=8.4Hz,2H),4.66(t,J=4.7Hz,1H),4.59(s,2H),3.53(t,J=5.7Hz,2H),3.43(t,J=6.0Hz,2H),3.14(s,3H),2.71(s,3H).
MS(ESI,[M-H] -)m/z:516.2/518.1.
Example 28: preparation of Compound 28
Figure PCTCN2021072699-APPB-000111
Step A: preparation of Compound 28-1
Referring to the preparation method of step B of example 1, compound 28-1 is prepared by reacting compound 1-1 with azetidine hydrochloride.
1H NMR(500MHz,DMSO-d 6)δ10.24(s,1H),8.68(d,J=2.0Hz,1H),8.29(d,J=2.0Hz,1H),7.85(d,J=9.1Hz,2H),7.34(d,J=9.0Hz,2H),4.31(t,J=7.6Hz,4H),2.27(p,J=7.6Hz,2H).
And B: preparation of Compound 28
Compound 28 was prepared by reacting the reaction mixture containing Compound 2-3 obtained in step C of example 2 with Compound 28-1 obtained in step A described above, according to the procedure of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ8.96(d,J=1.9Hz,1H),8.76(d,J=2.1Hz,1H),8.50(d,J=2.1Hz,1H), 8.31(d,J=1.9Hz,1H),7.96(d,J=9.1Hz,2H),7.37(d,J=8.9Hz,2H),5.59(s,4H),4.56(s,2H),2.56(p,J=1.9Hz,1H).
MS(ESI,[M-H] -)m/z:484.16/486.14.
Example 29: preparation of Compound 29
Figure PCTCN2021072699-APPB-000112
Step A: preparation of Compound 29-1
Compound 29-1 was prepared by reacting compound 1-1 with 3-fluoroazetidine hydrochloride according to the procedure for the preparation of step B of example 1.
And B, step B: compound 29 preparation
Compound 29 can be prepared by reacting compound 29-1 obtained in step A above with the reaction mixture containing compound 2-3 obtained in step C of example 2 by the method described in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.28(s,1H),8.94(s,1H),8.86(d,J=2.0Hz,1H),8.82(d,J=2.2Hz,1H),8.15(d,J=2.2Hz,1H),8.14(d,J=2.0Hz,1H),7.88(d,J=9.1Hz,2H),7.36(d,J=8.9Hz,2H),5.47–5.16(m,1H),4.52(s,2H),4.10–3.96(m,2H),3.83–3.68(m,2H).
MS(ESI,[M-H] -)m/z:502.20/504.25.
Example 30: preparation of Compound 30
Figure PCTCN2021072699-APPB-000113
Step A: preparation of Compound 30
Compound 30 was prepared by the method of step F of example 1, using the reaction mixture containing Compound 10-3 obtained in step B of example 10 and Compound 28-1 obtained in step A of example 28.
1H NMR(500MHz,DMSO-d 6)δ10.69(s,1H),9.25(d,J=2.1Hz,1H),9.14(d,J=2.0Hz,1H),8.75(t,J=2.1Hz,1H),8.57(d,J=2.1Hz,1H),7.92(d,J=9.1Hz,2H),7.41(d,J=9.0Hz,2H),4.64(s,2H),3.70–3.60(m,2H),3.43–3.37(m,2H),3.25(s,3H),1.95–1.84(m,2H).
MS(ESI,[M-H] -)m/z:498.14/500.10.
Example 31: preparation of Compound 31
Figure PCTCN2021072699-APPB-000114
Step A: preparation of Compound 31
Compound 31 can be prepared by reacting the reaction mixture containing compound 10-3 obtained in step B of example 10 with compound 29-1 obtained in step A of example 29, according to the procedure of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.28(s,1H),8.83(d,J=2.0Hz,1H),8.82(d,J=2.2Hz,1H),8.14(d,J=2.2Hz,1H),8.12(d,J=2.0Hz,1H),7.87(d,J=9.1Hz,2H),7.36(d,J=8.9Hz,2H),5.41–5.22(m,1H),4.61(s,2H),4.11–3.96(m,2H),3.81–3.67(m,2H),3.15(s,3H).
MS(ESI,[M-H] -)m/z:516.13/518.11.
Example 32: preparation of Compound 32
Figure PCTCN2021072699-APPB-000115
Step A: preparation of Compound 32
Compound 32 was prepared by reacting the reaction mixture containing compound 10-3 obtained in step B of example 10 with compound 8-1 obtained in step A of example 8, according to the procedure of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.19(s,1H),8.78(d,J=2.3Hz,1H),8.77(d,J=2.0Hz,1H),8.08(d,J=2.2Hz,1H),8.07(d,J=1.9Hz,1H),7.87(d,J=9.1Hz,2H),7.34(d,J=8.8Hz,2H),4.59(s,2H),3.14(s,3H),3.13–3.04(m,4H),1.82–1.67(m,4H).
MS(ESI,[M-H] -)m/z:512.20/514.16.
Example 33: preparation of Compound 33
Figure PCTCN2021072699-APPB-000116
Step A: preparation of Compound 33-1
Referring to the preparation method of step B of example 1, compound 33-1 was prepared by reacting compound 1-1 with (S) -3-hydroxymethyltetrahydropyrrole hydrochloride.
1H NMR(500MHz,DMSO-d 6)δ10.23(s,1H),8.69(d,J=2.1Hz,1H),8.35(d,J=2.1Hz,1H),7.86(d,J=9.1Hz,2H),7.34(d,J=9.0Hz,2H),3.78–3.66(m,4H),1.97–1.83(m,4H).
And B: preparation of Compound 33
Compound 33 can be produced by reacting the reaction mixture containing compound 10-3 obtained in step B of example 10 with compound 33-1 obtained in the above-mentioned step by the method described in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.20(s,1H),8.79(d,J=1.9Hz,1H),8.78–8.74(m,1H),8.10(d,J=1.9Hz,1H),8.08(d,J=2.1Hz,1H),7.87(d,J=9.0Hz,2H),7.34(d,J=8.7Hz,2H),4.60(s,2H),3.94–3.80(m,1H),3.31–3.25(m,1H),3.25–3.17(m,2H),3.16(s,3H),3.15(s,3H),3.12–3.06(m,1H),1.95–1.88(m,1H),1.88–1.79(m,1H).
MS(ESI,[M-H] -)m/z:542.19/544.20.
Example 34: preparation of Compound 34
Figure PCTCN2021072699-APPB-000117
Step A: preparation of Compound 34
Compound 34 was obtained by reacting the reaction mixture containing compound 2-3 obtained in step C of example 2 with compound 33-1 obtained in step A of example 33 in accordance with the procedure in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.20(s,1H),8.92(s,1H),8.82–8.76(m,2H),8.11(d,J=2.3Hz,1H),8.10–8.07(m,1H),7.87(d,J=9.1Hz,2H),7.35(d,J=8.9Hz,2H),4.51(s,2H),3.90(p,J=4.8,2.3Hz,1H),3.31–3.26(m,1H),3.26–3.17(m,2H),3.16(s,3H),3.13–3.05(m,1H),1.96–1.89(m,1H),1.89–1.77(m,1H).
MS(ESI,[M-H] -)m/z:528.18/530.22.
Example 35: preparation of Compound 35
Figure PCTCN2021072699-APPB-000118
Step A: preparation of Compound 35-1
Referring to the preparation method of step B of example 1, compound 1-1 and (S) -3-cyanopyrrolidine hydrochloride should be prepared to give compound 35-1.
1H NMR(500MHz,DMSO-d 6)δ10.30(s,1H),8.72(d,J=1.9Hz,1H),8.41(d,J=1.9Hz,1H),7.87(d,J=9.0Hz,2H),7.35(d,J=8.8Hz,2H),4.05–3.97(m,1H),3.95–3.86(m,2H),3.85–3.77(m,1H),3.53(p,J=6.4Hz,1H),2.38–2.28(m,1H),2.27–2.16(m,1H).
And B: preparation of Compound 35
Compound 35 was prepared by reacting the reaction mixture containing Compound 10-3 obtained in step B of example 10 with Compound 35-1 obtained in the above-mentioned step, according to the procedure of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.26(s,1H),8.81(d,J=2.3Hz,1H),8.79(d,J=2.0Hz,1H),8.14(d,J=2.3Hz,1H),8.12(d,J=2.0Hz,1H),7.87(d,J=9.1Hz,2H),7.35(d,J=8.9Hz,2H),4.61(s,2H),3.57–3.48(m,1H),3.46–3.36(m,2H),3.26–3.17(m,2H),3.15(s,3H),2.23–2.13(m,1H),2.11–2.02(m,1H).
MS(ESI,[M-H] -)m/z:537.18/539.19.
Example 36: preparation of Compound 36
Figure PCTCN2021072699-APPB-000119
Step A: preparation of Compound 36
Compound 36 can be prepared by reacting the reaction mixture containing Compound 2-3 obtained in step C of example 2 with Compound 35-1 obtained in step A of example 35, according to the procedure of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.25(s,1H),8.93(s,1H),8.81(d,J=2.1Hz,2H),8.17–8.12(m,2H),7.87(d,J=9.1Hz,2H),7.35(d,J=8.9Hz,2H),4.52(s,2H),3.58–3.50(m,1H),3.47–3.37(m,2H),3.28–3.12(m,2H),2.25–2.13(m,1H),2.13–2.03(m,1H).
MS(ESI,[M-H] -)m/z:523.18/525.18.
Example 37: preparation of Compound 37
Figure PCTCN2021072699-APPB-000120
Step A: preparation of Compound 37-1
Referring to the preparation method of step B of example 1, compound 37-1 was prepared by reacting compound 1-1 with ethyl 4-aminobutyrate hydrochloride.
1H NMR(500MHz,DMSO-d 6)δ10.19(s,1H),8.70–8.62(m,1H),8.29(d,J=2.0Hz,1H),7.86(d,J=9.1Hz,2H),7.34(d,J=8.9Hz,2H),7.08(t,J=5.8Hz,1H),4.05(q,J=7.1Hz,2H),3.47(q,J=6.6Hz,2H), 2.34(t,J=7.4Hz,2H),1.84(q,J=7.1Hz,2H),1.17(t,J=7.1Hz,3H).
And B, step B: preparation of Compound 37-2
A50 mL single-neck flask was charged with compound 37-1(660mg) obtained in step A and 1M aqueous sodium hydroxide solution (10mL), and heated to 80 ℃ for 2 hours. The pH was adjusted to 5 with 6M dilute hydrochloric acid, diluted with 10mL of water and extracted 3 times with ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and concentrated to give 600mg of compound 37-2.
1H NMR(500MHz,DMSO-d 6)δ12.06(s,1H),10.20(s,1H),8.66(d,J=2.0Hz,1H),8.29(d,J=2.0Hz,1H),7.86(d,J=9.1Hz,2H),7.34(d,J=8.9Hz,2H),7.08(t,J=5.7Hz,1H),3.46(q,J=6.6Hz,2H),2.27(t,J=7.4Hz,2H),1.81(p,J=7.2Hz,2H).
And C: preparation of Compound 37-3
A50 mL single-neck flask was charged with the compound 37-2(600mg) obtained in the above step B, DMF (20mL) and triethylamine (162mg), and a DMF (5mL) solution of pentafluorophenyl diphenyl phosphate (482mg) was added dropwise at room temperature, and the mixture was heated to 50 ℃ to react, after completion of the reaction. 150mL of water was added, extracted three times with ethyl acetate, the organic phases were combined and washed three times with saturated aqueous sodium chloride solution. The organic phase was dried over anhydrous sodium sulfate and concentrated, and the crude product was purified by column chromatography to give 489mg of compound 37-3.
1H NMR(500MHz,DMSO-d 6)δ10.68(s,1H),8.97(d,J=1.8Hz,1H),8.66(d,J=1.8Hz,1H),7.88(d,J=9.0Hz,2H),7.39(d,J=8.8Hz,2H),3.90(t,J=6.9Hz,2H),2.49(d,J=8.1Hz,2H),2.18(p,J=7.4Hz,2H).
Step D: preparation of Compound 37
Compound 37 can be obtained by reacting the reaction mixture containing Compound 10-3 obtained in step B of example 10 with Compound 37-3 obtained in the above-mentioned step by the method described in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.63(s,1H),9.01(d,J=2.2Hz,1H),8.84(d,J=1.9Hz,1H),8.49–8.42(m,1H),8.20(d,J=2.0Hz,1H),7.89(d,J=9.1Hz,2H),7.40(d,J=8.9Hz,2H),4.59(s,2H),4.14(t,J=6.8Hz,2H),3.14(s,3H),2.18(t,J=7.7Hz,2H),2.07(p,J=8.0,7.4Hz,2H).
MS(ESI,[M+H] +)m/z:528.3/530.2.
Example 38: preparation of Compound 38
Figure PCTCN2021072699-APPB-000121
Step A: preparation of Compound 38-1
Referring to the preparation method of step B of example 1, compound 38-1 was prepared by reacting compound 1-1 with (R) -3-methoxypyrrolidine.
And B: preparation of Compound 38
Compound 38 can be prepared by reacting the reaction mixture containing compound 2-3 obtained in step C of example 2 with compound 38-1 obtained in step A described above, by the method described in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.20(s,1H),8.92(s,1H),8.79(d,J=2.5Hz,2H),8.10(m,2H),7.86(d,J=8.0Hz,2H),7.53(d,J=7.5Hz,2H),4.51(s,2H),3.89(s,1H),3.25(m,3H),3.17(s,3H),3.10(m,1H),1.85(m,2H).
MS(ESI,[M+H] +)m/z:530.1.
Example 39: preparation of Compound 39
Figure PCTCN2021072699-APPB-000122
Step A: preparation of Compound 39
Compound 39 can be prepared by reacting the reaction mixture containing Compound 10-3 obtained in step B of example 10 with Compound 38-1 obtained in step 38 of example 1 by the method described in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.20(s,1H),8.78(m,2H),8.08(m,2H),7.86(d,J=9.0Hz,2H),7.34(d, J=8.5Hz,2H),4.60(s,2H),3.89(s,1H),3.25(m,3H),3.20(s,3H),3.18(s,3H),3.12(m,1H),1.85(m,2H).
MS(ESI,[M+H] +)m/z:544.2.
Example 40: preparation of Compound 40
Figure PCTCN2021072699-APPB-000123
Step A: preparation of Compound 40-1
Referring to the preparation method of step B of example 1, compound 40-1 is prepared by reacting compound 1-1 with (R) -3-cyanopyrrolidine.
And B: preparation of Compound 40
Compound 40 was prepared by reacting the reaction mixture containing Compound 2-3 obtained in step C of example 2 with Compound 40-1 obtained in step A described above, according to the procedure of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.25(s,1H),8.92(s,1H),8.81(s,2H),8.14(d,J=1.5Hz,2H),7.86(d,J=8.5Hz,2H),7.35(d,J=8.5Hz,2H),4.52(s,2H),3.52(m,1H),3.39(m,2H),3.18(m,2H),2.12(m,2H).
HRMS(ESI,[M+H] +)m/z:525.1.
EXAMPLE 41 preparation of Compound 41
Figure PCTCN2021072699-APPB-000124
Step A: preparation of Compound 41
Compound 41 was prepared by reacting the reaction mixture containing Compound 10-3 obtained in step B of example 10 with Compound 40-1 obtained in step A of example 40 according to the procedure in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.26(s,1H),8.79(d,J=5.0Hz,2H),8.12(d,J=8.0Hz,2H),7.86(d,J=8.5Hz,2H),7.34(d,J=8.5Hz,2H),4.61(s,2H),3.52(s,1H),3.40(d,J=7.0Hz,2H),3.18(m,2H),3.15(s,3H),2.12(m,2H).
HRMS(ESI,[M+H] +)m/z:539.1418.
Example 42: preparation of Compound 42
Figure PCTCN2021072699-APPB-000125
Step A: preparation of Compound 42-1
To a 25mL three-necked flask, 2-2(0.931g) of the compound obtained in step B of example 2 was charged,1, 4-dioxane (20mL), pinacol diboron ester (1.664g), PdCl2(dppf) (0.266g), potassium acetate (1.072g), after addition, N2After protection, the temperature is raised to 90 ℃ for reaction for 2 h. After the reaction was completed, the reaction system was cooled to room temperature, and the compound 1-1(1.5g) obtained in step A of example 1, potassium carbonate (1.509g), Pd (Ph) were sequentially added to the system3P) 4(0.421g) and water (5mL) were added, the mixture was replaced with nitrogen, and the mixture was heated to 110 ℃ to react for 2 hours. After completion of the reaction, the reaction mixture was filtered, and the filtrate was collected, washed with ethyl acetate (50mL) and saturated brine (25mL), separated, dried over anhydrous sodium sulfate, and concentrated. The crude product was purified by column chromatography to give 0.5g of compound 42-1.
1H NMR(500MHz,DMSO-d 6)δ10.68(s,1H),9.00(d,J=2.3Hz,1H),8.98(d,J=2.0Hz,1H),8.95(s,1H),8.53(d,J=2.3Hz,1H),8.34(d,J=2.0Hz,1H),7.88(d,J=9.1Hz,2H),7.40(d,J=8.9Hz,2H),4.55(s,2H).
MS(ESI,[M+H] -)m/z:463.1.
And B: preparation of Compound 42-2
To a 35mL microwave tube, compound 42-2(0.3g), 1, 4-dioxane (20mL), 4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester (0.199g), bis (tri-tert-butylphosphine) palladium (0) (0.033g), cesium carbonate (0.420g) and water (4.00mL) were added in this order, the reaction was placed in a microwave reactor under microwave conditions: at 150 deg.C for 30 min. After the reaction was completed, 50mL of ethyl acetate and 20mL of water were added to the reaction solution, the organic phase was collected by liquid separation, and washed three times with 20mL of a saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, and concentrated. The crude product was purified by column chromatography to give 210mg of compound 42-2.
1H NMR(500MHz,DMSO-d 6)δ10.62(s,1H),9.12(d,J=2.5Hz,1H),8.90(s,1H),8.83(d,J=2.0Hz,1H),8.40(d,J=2.5Hz,1H),8.24(d,J=2.0Hz,1H),7.89(m,2H),7.39(d,J=8.5Hz,2H),5.63(s,1H),4.50(s,2H),4.28(s,2H),3.35(s,2H),1.98(s,2H),1.41(s,9H).
MS(ESI,[M+Na] +)m/z:634.3.
And C: preparation of Compound 42
To a 25mL single-necked flask, compound 42-2(50mg) obtained in the above step and a solution of 4M hydrogen chloride in 1, 4-dioxane (5mL) were added in this order, and the mixture was stirred at room temperature for 1 hour. After the reaction was completed, the reaction solution was concentrated to remove the solvent, and the obtained crude product was purified by column chromatography to obtain 40mg of compound 42.
1H NMR(500MHz,DMSO-d 6)δ10.87(s,1H),9.48(s,2H),9.15(d,J=2.0Hz,1H),8.94(s,2H),8.47(d,J=2.0Hz,1H),8.33(d,J=2.0Hz,1H),7.94(d,J=8.5Hz,2H),7.39(d,J=8.5Hz,2H),5.66(s,1H),5.51(s,2H),4.15(s,2H),3.11(d,J=4.0Hz,2H),2.17(s,2H).
MS(ESI,[M+H] +)m/z:512.1.
Example 43: preparation of Compound 43
Figure PCTCN2021072699-APPB-000126
Compound 43 was prepared by reacting compound 42-1 obtained in step a of example 42 with 3, 6-dihydro-2H-pyran-4-boronic acid pinacol ester according to the procedure of example 42, step B.
1H NMR(500MHz,DMSO-d 6)δ10.62(s,1H),9.12(d,J=2.0Hz,1H),8.91(s,1H),8.83(d,J=1.5Hz,1H),8.39(d,J=1.5Hz,1H),8.21(d,J=1.5Hz,1H),7.89(d,J=8.0Hz,2H),7.39(d,J=7.5Hz,2H),5.64(s,1H),4.52(s,2H),3.95(d,J=2.0Hz,2H),3.72(t,J=5.5Hz,2H),2.45(s,2H).
HRMS(ESI,[M+H] +)m/z:513.1174.
Example 44: preparation of Compound 44
Figure PCTCN2021072699-APPB-000127
Step A: preparation of Compound 44-1
Compound 44-1 was prepared by reacting compound 1-1 obtained in step a of example 1 with compound 10-2 obtained in step a of example 10, according to the procedure of step a of example 42.
1H NMR(500MHz,DMSO-d 6)δ10.69(s,1H),8.97(m,2H),8.56(d,J=2.0Hz,1H),8.32(d,J=2.0Hz,1H),7.86(d,J=9.5Hz,2H),7.05(d,J=9.0Hz,2H),4.64(s,2H),3.15(s,3H).
MS(ESI,[M+H] +)m/z:479.2.
And B: preparation of Compound 44-2
Referring to the procedure of step B of example 42, compound 44-2 was prepared using the reaction of compound 44-1 obtained in the above step A with 3, 6-dihydro-2H-pyran-4-boronic acid pinacol ester.
MS(ESI,[M+H] +)m/z:527.3.
And C: preparation of Compound 44
To a 100mL single-necked flask, 44-2(150mg) obtained in the above procedure and methanol (20mL) were sequentially added, Pd/C (1.671mg) was added, and hydrogen gas was substituted three times, followed by stirring at room temperature overnight. Filtering after the reaction is completed, concentrating the filtrate to obtain a crude product, and purifying by column chromatography to obtain 88mg of the compound 44.
1H NMR(500MHz,DMSO-d 6)δ10.59(s,1H),9.15(d,J=2.0Hz,1H),8.83(d,J=1.5Hz,1H),8.19(m,2H),7.88(d,J=9.0Hz,2H),7.38(d,J=8.5Hz,2H),4.64(s,2H),3.86(dd,J=3.5Hz,11Hz,2H),3.22(t,J=11.5Hz,2H),3.16(s,3H),3.00(m,1H),1.95(m,2H),1.60(d,J=12.5Hz,2H).
MS(ESI,[M+H] +)m/z:529.1.
Example 45: preparation of Compound 45
Figure PCTCN2021072699-APPB-000128
Step A: preparation of Compound 45-1
Referring to the preparation method of step B of example 1, compound 45-1 is prepared by reacting compound 1-1 with 4-hydroxypiperidine.
MS(ESI,[M+H] +)m/z:476.1/478.1.
And B: preparation of Compound 45
Compound 45 was prepared by reacting the reaction mixture containing Compound 10-3 obtained in step B of example 10 with Compound 45-1 obtained in step A described above, according to the preparation method of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.31(s,1H),8.98(d,J=1.6Hz,1H),8.78(d,J=2.1Hz,1H),8.32–8.24(m,1H),8.16(d,J=2.0Hz,1H),7.86(d,J=9.0Hz,2H),7.35(d,J=8.7Hz,2H),4.67(d,J=4.0Hz,1H),4.60(s,2H),3.58(dq,J=8.3,4.1Hz,1H),3.44(d,J=13.1Hz,2H),3.14(s,3H),2.86(t,J=10.5Hz,2H),1.63(d,J=9.8Hz,2H),1.28(d,J=9.5Hz,2H).
MS(ESI,[M+H] +)m/z:544.1.
Example 46: preparation of Compound 46
Figure PCTCN2021072699-APPB-000129
Step A: preparation of Compound 46
Referring to the preparation process of step F of example 1, compound 46 was prepared by reacting compound 45-1 obtained in step A of example 45 with the reaction solution containing compound 11-2 obtained in step B of example 11.
1H NMR(500MHz,DMSO-d 6)δ10.31(s,1H),8.99(d,J=1.8Hz,1H),8.78(d,J=2.2Hz,1H),8.27(d,J=1.7Hz,1H),8.15(d,J=2.1Hz,1H),7.86(d,J=9.0Hz,2H),7.35(d,J=8.8Hz,2H),4.67(d,J=4.0Hz,1H),4.53(s,2H),3.58(dq,J=8.4,4.2Hz,1H),3.45(d,J=13.1Hz,2H),3.01(tt,J=7.4,4.0Hz,1H),2.86(t,J=10.5Hz,2H),1.64(d,J=9.9Hz,2H),1.29(d,J=9.5Hz,2H),0.92(d,J=3.3Hz,2H),0.83(d,J=5.3Hz,2H).
MS(ESI,[M+H] +)m/z:570.2.
Example 47: preparation of Compound 47
Figure PCTCN2021072699-APPB-000130
Step A: preparation of Compound 47-1
Referring to the procedure for the preparation of step B of example 1, compound 47-1 was prepared by reacting compound 1-1 with 3-methylhydroxyazetidine hydrochloride.
MS(ESI,[M-H] -)m/z:460.0/462.0.
And B: preparation of Compound 47
Referring to the preparation process of step F of example 1, compound 47 was prepared by reacting compound 47-1 obtained in the above step A with the reaction solution containing compound 10-3 obtained in step B of example 10.
1H NMR(500MHz,Methanol-d 4)δ8.82(d,J=1.9Hz,1H),8.78(d,J=2.2Hz,1H),8.17(d,J=1.9Hz,1H),8.05(d,J=2.2Hz,1H),7.79(d,J=9.0Hz,2H),7.27(d,J=8.9Hz,2H),4.62(s,2H),3.84(t,J=8.6Hz,2H),3.64–3.53(m,4H),3.26(s,3H),2.70(p,J=8.0Hz,1H).
MS(ESI,[M+H] +)m/z:530.1.
Example 48: preparation of Compound 48
Figure PCTCN2021072699-APPB-000131
Step A: preparation of Compound 48
Compound 48 can be obtained by reacting the reaction mixture containing Compound 11-2 obtained in step B of example 11 with Compound 47-1 obtained in step A of example 47 according to the preparation process in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.21(s,1H),8.78(d,J=8.4Hz,2H),8.06(d,J=15.2Hz,2H),7.86(d,J=8.6Hz,2H),7.34(d,J=8.3Hz,2H),4.68(s,1H),4.53(s,2H),3.69(t,J=7.9Hz,2H),3.43(dd,J=11.6,6.0Hz,4H),3.01(s,1H),2.61(s,1H),0.92(s,2H),0.83(d,J=5.5Hz,2H).
MS(ESI,[M+H] +)m/z:556.2.
Example 49: preparation of Compound 49
Figure PCTCN2021072699-APPB-000132
Step A: preparation of Compound 49-1
Compound 49-1 was prepared by reacting compound 1-1 with cis-2, 6-dimethylmorpholine according to the procedure for preparation of step B of example 1.
MS(ESI,[M+H] +)m/z:490.0/492.0.
And B: preparation of Compound 49
Compound 49 can be prepared by reacting compound 49-1 obtained in step A with the reaction mixture containing compound 10-3 obtained in step B of example 10 according to the preparation process of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.35(s,1H),8.99(d,J=1.8Hz,1H),8.80(d,J=2.2Hz,1H),8.30(d,J=1.8Hz,1H),8.20(d,J=2.2Hz,1H),7.87(d,J=9.1Hz,2H),7.36(d,J=8.8Hz,2H),4.61(s,2H),3.50(d,J=16.4Hz,2H),3.44(d,J=12.6Hz,2H),3.15(s,3H),2.48–2.40(m,2H),0.96(d,J=6.2Hz,6H).
MS(ESI,[M+H] +)m/z:558.2.
Example 50: preparation of Compound 50
Figure PCTCN2021072699-APPB-000133
Step A: preparation of Compound 50-1
Referring to the preparation method of step B of example 1, Compound 50-1 was prepared by reacting compound 1-1 with 3-methyl-3-acridine alkoxide.
MS(ESI,[M+H] -)m/z:460.0/462.0.
And B: preparation of Compound 50
Compound 50 was prepared by reacting the reaction mixture containing Compound 11-2 obtained in step B of example 11 with Compound 50-1 obtained in step A described above, according to the preparation method of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.23(s,1H),8.79(dd,J=7.3,1.9Hz,2H),8.07(dd,J=4.7,2.0Hz,2H),7.86(d,J=9.0Hz,2H),7.34(d,J=8.8Hz,2H),5.48(s,1H),4.53(s,2H),3.62–3.48(m,4H),3.01(tt,J=7.4,3.9Hz,1H),1.27(s,3H),0.92(d,J=3.3Hz,2H),0.84(d,J=5.3Hz,2H).
MS(ESI,[M+H] +)m/z:556.2.
Example 51: preparation of Compound 51
Figure PCTCN2021072699-APPB-000134
Step A: preparation of Compound 51-1
Under the protection of nitrogen, (S) - (-) -2-carboxycyclobutylamine (200mg), tetrahydrofuran (10mL) and borane tetrahydrofuran complex (425mg) are sequentially added into a 50mL sealed tube, and after sealing, the sealed tube is placed into an oil bath pot and heated to 72 ℃ for stirring and reaction overnight. Methanol (5mL) was added to the reaction mixture and stirred for 10min, and the reaction mixture was concentrated under reduced pressure to give a pale yellow residue 51-1, which was used in the next reaction without separation and purification.
And B: preparation of Compound 51-2
Referring to the preparation method of step B of example 1, compound 51-2 was prepared by reacting compound 51-1 with compound 1-1 obtained in the above step A.
MS(ESI,[M-H] -)m/z:460.0/462.0.
And C: preparation of Compound 51
Compound 51 was prepared by reacting compound 51-2 of step B described above with the reaction solution containing compound 10-3 of step B of example 10 according to the preparation process of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.22(s,1H),8.85(d,J=2.0Hz,1H),8.76(d,J=2.3Hz,1H),8.14(d,J=2.1Hz,1H),8.08(d,J=2.3Hz,1H),7.86(d,J=9.1Hz,2H),7.34(d,J=9.0Hz,2H),4.88(s,1H),4.59(s,2H),4.44(tt,J=7.5,4.1Hz,1H),3.69(dt,J=10.6,5.1Hz,1H),3.55(dt,J=11.2,5.7Hz,1H),3.19(q,J=7.9Hz,1H),3.14(s,3H),2.12(q,J=7.6Hz,2H).
MS(ESI,[M-H] -)m/z:528.2.
Example 52: preparation of Compound 52
Figure PCTCN2021072699-APPB-000135
Step A: preparation of Compound 52-1
Referring to the preparation method of example 51, step A, (R) - (-) -2-carboxycyclobutylamine and borane tetrahydrofuran complex were reacted to prepare compound 52-1.
And B: preparation of Compound 52-2
Referring to the preparation method of step B of example 1, compound 52-2 was prepared by reacting compound 52-1 obtained in the above step A with compound 1-1.
MS(ESI,[M+H] +)m/z:462.1/464.1.
Step C: preparation of Compound 52
Compound 52 was obtained by reacting the reaction mixture containing Compound 10-3 obtained in step B of example 10 with Compound 52-2 obtained in step B described above, according to the preparation process in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.22(s,1H),8.85(d,J=2.0Hz,1H),8.76(d,J=2.3Hz,1H),8.14(d,J=2.1Hz,1H),8.08(d,J=2.4Hz,1H),7.89–7.83(m,2H),7.34(d,J=9.0Hz,2H),4.90(t,J=5.8Hz,1H),4.59(s,2H),4.44(tt,J=7.5,4.2Hz,1H),3.69(dt,J=10.6,5.1Hz,1H),3.60–3.50(m,1H),3.19(q,J=8.0Hz,1H),3.14(s,3H),2.12(q,J=7.6Hz,2H).
MS(ESI,[M+H] +)m/z:530.3.
Example 53: preparation of Compound 53
Figure PCTCN2021072699-APPB-000136
Step A: preparation of Compound 53-1
Referring to the preparation method of step B of example 1, compound 53-1 was prepared by reacting compound 1-1 with (R) -3-fluoropyrrolidine.
MS(ESI,[M+H] +)m/z:464.0/466.0.
And B: preparation of Compound 53
Referring to the preparation process of step F of example 1, compound 53 was prepared by reacting compound 53-1 obtained in step A above with the reaction solution containing compound 2-3 obtained in step C of example 2.
1H NMR(500MHz,DMSO-d 6)δ10.23(s,1H),8.93(s,1H),8.82(d,J=1.7Hz,1H),8.81(d,J=2.3Hz,1H),8.13(d,J=2.3Hz,1H),8.13–8.11(m,1H),7.88(s,1H),7.86(s,1H),7.36(s,1H),7.34(s,1H),5.41–5.16(m,1H),4.52(s,2H),3.54–3.42(m,1H),3.34–3.22(m,3H),2.14–1.91(m,2H).
MS(ESI,[M-H] -)m/z:516.16/518.20.
Example 54: preparation of Compound 54
Figure PCTCN2021072699-APPB-000137
Step A: preparation of Compound 54
Compound 54 can be obtained by reacting the reaction mixture containing Compound 10-3 obtained in step B of example 10 with Compound 53-1 obtained in step A of example 53 by the preparation method described in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.23(s,1H),8.80(s,2H),8.11(d,J=11.2Hz,2H),7.87(d,J=8.8Hz,2H),7.35(d,J=8.5Hz,2H),5.39–5.18(m,1H),4.60(s,2H),3.54–3.40(m,1H),3.33–3.19(m,3H),3.15(s,3H),2.14–1.89(m,2H).
MS(ESI,[M-H] -)m/z:530.16/532.14.
Example 55: preparation of Compound 55
Figure PCTCN2021072699-APPB-000138
Step A: preparation of Compound 55-1
Referring to the preparation method of step B of example 1, compound 55-1 was prepared by reacting compound 1-1 with (S) -3-fluoropyrrolidine.
MS(ESI,[M+H] +)m/z:464.3/466.3.
And B: preparation of Compound 55
Compound 55 was prepared by reacting compound 55-1 of step A described above with the reaction solution containing compound 2-3 obtained in step C of example 2, according to the preparation method of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.23(s,1H),8.93(s,1H),8.81(s,2H),8.24–8.03(m,2H),7.87(d,J=7.8Hz,2H),7.35(d,J=7.6Hz,2H),5.40–5.16(m,1H),4.52(s,2H),3.57–3.41(m,1H),3.33–3.15(m,3H),2.20–1.87(m,2H).
MS(ESI,[M+H] +)m/z:518.3/520.3.
Example 56: preparation of Compound 56
Figure PCTCN2021072699-APPB-000139
Step A: preparation of Compound 56
Compound 56 can be obtained by reacting the reaction mixture containing Compound 10-3 obtained in step B of example 10 with Compound 55-1 obtained in step A of example 55 according to the preparation process in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.23(s,1H),8.86–8.76(m,2H),8.13(d,J=2.3Hz,1H),8.12–8.06(m,1H),7.87(d,J=9.1Hz,2H),7.35(d,J=8.9Hz,2H),5.37–5.18(m,1H),4.60(s,2H),3.55–3.40(m,1H),3.34–3.19(m,3H),3.15(s,3H),2.17–1.88(m,2H).
MS(ESI,[M+H] +)m/z:532.3/534.2.
Example 57: preparation of Compound 57
Figure PCTCN2021072699-APPB-000140
Step A: preparation of Compound 57-1
Adding 10mL of absolute ethyl alcohol, the compound 2-1(400mg) obtained in the step A of the example 2, diisopropylethylamine (428mg) and deuterated methylamine hydrochloride (97mg) into a 50mL single-neck flask in sequence, and reacting for 6h at room temperature after the addition is finished; concentrating the reaction solution under reduced pressure; the residue was subjected to column chromatography to give compound 57-1.
MS(ESI,[M+H] +)m/z:230.1/231.9.
And B: preparation of Compound 57-2
Referring to the procedure of step E of example 1, a reaction solution containing compound 57-2 was prepared using compound 57-1 obtained in the above step A, cooled to room temperature, and used for the next reaction without separation and purification.
And C: preparation of Compound 57
Compound 57 was prepared by the method referenced as step F in example 1 using the reaction solution containing compound 57-2 obtained in the above step B and compound 33-1 obtained in step A in example 33.
1H NMR(500MHz,DMSO-d 6)δ10.20(s,1H),8.78(d,J=2.4Hz,1H),8.77(d,J=2.2Hz,1H),8.09(d,J=2.3Hz,1H),8.07(d,J=2.1Hz,1H),7.90–7.82(m,2H),7.34(d,J=8.6Hz,2H),4.59(s,2H),3.93–3.85(m,1H),3.30–3.16(m,2H),3.15(s,3H),3.09(d,J=11.9Hz,1H),1.95–1.78(m,2H).
MS(ESI,[M-H] -)m/z:545.2/547.2.
Example 58: preparation of Compound 58
Figure PCTCN2021072699-APPB-000141
Step A: preparation of Compound 58
Compound 58 was prepared by reacting compound 5-1, obtained in step a of example 5, with (3- (methylsulfonyl) phenyl) boronic acid, according to the procedure described in step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.42(s,1H),8.82(d,J=2.3Hz,1H),8.27(s,1H),8.18(d,J=2.3Hz,1H),8.04(d,J=7.7Hz,1H),7.95(d,J=7.8Hz,1H),7.88(d,J=9.1Hz,2H),7.81(t,J=7.8Hz,1H),7.36(d,J=8.8Hz,2H),3.58–3.54(m,4H),3.30(s,3H),3.19–3.08(m,4H).
MS(ESI,[M+H] +)m/z:538.3/540.2.
Example 59: preparation of Compound 59
Figure PCTCN2021072699-APPB-000142
Step A: preparation of Compound 59-1
1, 4-dioxane (15mL), 1-tert-butoxycarbonyl-pyrrole-2-boronic acid (423mg), the compound 1-2(700mg) obtained in step B of example 1, potassium carbonate (626mg), tetrakis (triphenylphosphine) palladium (87mg), purified water (3mL) and magnetite were sequentially added to a 20mL microwave tube, and after purging with nitrogen, the mixture was placed in a microwave reactor and reacted at 130 ℃ for 1.5 hours; vacuum concentrating, and purifying the crude product by column chromatography to obtain 500mg of compound 59-1.
MS(ESI,[M-H] -)m/z:447.16/449.14.
And B: preparation of Compound 59-2
Adding ultra-dry tetrahydrofuran (15mL), the compound 59-1(400mg) obtained in the step A and azobisisobutyronitrile (21mg) into a 100mL single-neck bottle, and cooling to-78 ℃ under the nitrogen protection condition after adding; dibromohydantoin (94mg) was added to the system in portions, and the reaction was incubated for 2h after the addition. Vacuum concentrating, and purifying the crude product by column chromatography to obtain 370mg of compound 59-2.
MS(ESI,[M-H] -)m/z:525.1/527.1.
And C: preparation of Compound 59-3
To a 20mL microwave tube were added 1, 4-dioxane (15mL), 59-2(370mg) of the compound obtained in step B, N-diisopropylethylamine (94mg), sodium thiomethoxide (51.0mg), tris (dibenzylidene-BASE acetone) dipalladium (50mg), 4, 5-bis diphenylphosphine-9, 9-dimethylxanthene (63mg), and magnetite, and the mixture was placed in a microwave reactor after purging with nitrogen and reacted at 130 ℃ for 2 hours. Vacuum concentrating, and purifying by column chromatography to obtain 250mg of compound 59-3.
MS(ESI,[M-H] -)m/z:493.2/495.2.
Step D: preparation of Compound 59
To a 25mL single-neck flask were added isopropanol (5mL), purified water (1.5mL), compound 59-3 from step C (230mg), and potassium peroxymonosulfonate (195mg) in that order, and the reaction was carried out at room temperature for 0.5 h. Ethyl acetate was added to the reaction system to extract 3 times, and the organic phases were combined and washed with a saturated aqueous sodium sulfite solution and a saturated brine in this order 2 times each. The organic phase was collected, dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the crude product was purified by column chromatography to give 20mg of compound 59.
1H NMR(500MHz,DMSO-d 6)δ12.50(s,1H),10.18(s,1H),8.76(d,J=2.4Hz,1H),8.08(d,J=2.4Hz,1H),7.93–7.81(m,2H),7.40–7.28(m,2H),6.80(dd,J=3.6,2.4Hz,1H),6.28(dd,J=3.7,2.3Hz,1H),4.88(d,J=3.4Hz,1H),4.23(s,1H),3.52–3.42(m,1H),3.21(s,3H),2.97(d,J=11.6Hz,1H),1.90–1.81(m,1H),1.80–1.72(m,1H).
MS(ESI,[M-H] -)m/z:525.2/527.2.
Example 60: preparation of Compound 60
Figure PCTCN2021072699-APPB-000143
Step A: preparation of Compound 60-1
The title compound 60-1 was prepared using compound 1-1 and 2- (methylamino) ethan-1-ol obtained in step A of example 1, according to the procedure of step B of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.68(s,1H),9.00(d,J=2.3Hz,1H),8.98(d,J=2.0Hz,1H),8.95(s,1H),8.53(d,J=2.3Hz,1H),8.34(d,J=2.0Hz,1H),7.88(d,J=9.1Hz,2H),7.40(d,J=8.9Hz,2H),4.55(s,2H).
And B, step B: preparation of Compound 60
Compound 60 was prepared by reacting compound 60-1 obtained in step A above with the reaction solution containing compound 2-3 obtained in step C of example 2, according to the procedure of step F of example 1.
1H NMR(500MHz,DMSO-d 6)δ10.25(s,1H),8.91(s,1H),8.88(s,1H),8.76(s,1H),8.19(s,1H),8.14(s,1H),7.87(d,J=8.8Hz,2H),7.35(d,J=8.5Hz,2H),4.66(t,J=4.9Hz,1H),4.51(s,2H),3.54(q,J=5.7Hz,2H),3.44(t,J=6.0Hz,2H),2.71(s,3H).
MS(ESI,[M+H] +)m/z:504.2/506.2.
Example 61: preparation of Compound 61
Figure PCTCN2021072699-APPB-000144
Step A: preparation of Compound 61-1
Compound 61-1 was prepared by reacting compound 2-1 with 1-tert-butoxycarbonyl-4-aminopiperidine instead of 2-methoxyethylamine, according to the procedure as in step a of example 13.
And B: preparation of Compound 61-2
Referring to the procedure of step E of example 1, a reaction solution containing compound 61-2 was prepared using compound 61-1 obtained in the above step A, cooled to room temperature, and used in the next reaction without separation and purification.
Step C: preparation of Compound 61-3
Compound 61-3 was prepared by the method of step F of example 1 using the reaction solution containing compound 61-2 obtained in step B above and compound 12-1 obtained in step A of example 12.
MS(ESI,[M-H] -)m/z:612.29/614.30.
Step D: preparation of Compound 61
To a 50mL round-bottomed flask, compound 61-3(430mg) obtained in the above step A, methylene chloride (10mL) and a 6M hydrogen chloride-dioxane solution (2.5mL) were added, and the mixture was stirred at room temperature overnight, after completion of the reaction, the reaction mixture was concentrated, followed by addition of ethyl acetate (10mL) and stirring at room temperature for 4 hours. Filter and wash the filter cake with ethyl acetate. The filter cake was collected and dried to yield 60mg of compound 61.
1H NMR(500MHz,DMSO-d 6)δ9.50–9.45(m,1H),9.41(s,1H),9.38(d,J=6.7Hz,1H),9.34(s,1H),8.74(s,1H),8.02(d,J=9.0Hz,2H),7.40(d,J=8.5Hz,2H),4.64(s,2H),4.41(d,J=11.7Hz,1H),3.37(d,J=12.1Hz,2H),3.07(t,J=11.7Hz,2H),2.14(d,J=13.3Hz,2H),1.95(d,J=12.3Hz,2H).
MS(ESI,[M+H] +)m/z:514.3/516.4.
Test example 1 proliferation inhibitory Effect of Compounds on K562 cells
Collecting K562 cells in exponential growth phase, collecting the cells to a centrifuge tube, centrifuging for 5min at a low-speed desk centrifuge at 1000 rpm, discarding the supernatant, and adding 5mL of complete culture medium (RPMI1640 basic medium + 10% FBS) to the pipette for cell resuspension. Counting with a cell counter, diluting the complete medium, adjusting the cell density to 6X 104Adding equal amount of RPMI1640 basic culture medium to adjust serum concentration to 5% and cell density to 3 × 104one/mL. Inoculating to 96-well plate with multi-channel pipette at 100 μ L/well, placing at 37 deg.C and 5% CO2Culturing in a cell culture box with saturated humidity. After 24h incubation, compounds were loaded using a nanoliter loader, with 8 concentrations (1000nM,250nM,63nM,16nM,3.9nM,0.98nM,0.24nM,0.061nM) per compound, 2 duplicate wells per concentration, with cells without compound as negative controls and medium without cells as blank controls. After 72 hours, adding CCK-8 with 10 mu L/hole,detecting the light absorption value of the Envision microplate reader at 450nm after 2 hours, calculating the inhibition rate, wherein the inhibition rate (%) (negative control group average value-experimental group average value)/(negative control group average value-blank group average value) is multiplied by 100%, taking the logarithm of the concentration of the compound as the abscissa, the inhibition rate as the ordinate, analyzing four parameters, fitting a dose-effect curve, and calculating the IC50The results are shown in Table 1.
Experimental example 2 proliferation inhibitory Effect of Compounds on Ba/F3 cells transfected with BCR-ABL T315I
The Ba/F3-BCR-ABL1-T315I cells (purchased from Bai Biotech Co., Ltd., Nanjing) in the exponential growth phase are taken, the cells are collected into a centrifuge tube, a low-speed desk type centrifuge is used for 1000 r/min, the centrifuge tube is centrifuged for 5min, the supernatant is discarded, and a pipettor is used for adding 5mL of complete culture medium (RPMI1640 basic medium + 10% FBS) for cell resuspension. Counting with a cell counter, diluting the complete medium, and adjusting the cell density to 6X 104Adding the same amount of RPMI1640 basic medium to adjust the serum concentration to 5% and the cell density to 3 × 104one/mL. Inoculating to 96-well plate with multi-channel pipette at 100 μ L/well, placing at 37 deg.C and 5% CO2Culturing in a cell culture box with saturated humidity. After 24h incubation, compounds were loaded using a nanoliter loader at 8 concentrations (1000nM,250nM,63nM,16nM,3.9nM,0.98nM,0.24nM,0.061nM) per compound, 2 replicate wells per concentration, with cells without compound as negative controls and medium without cells as blank controls. Adding CCK-8 and 10 mu L/well after 72 hours, adding CCK8 into Ba/F3-BCR-ABL1-T315I cells, placing the cells at 37 ℃ after adding CCK8, detecting the light absorption value at 450nm by using an enzyme labeling instrument after 1 hour, calculating the inhibition rate, wherein the inhibition rate (%) (negative control group mean value-experimental group mean value)/(negative control group mean value-blank group mean value) x 100%, taking the logarithm of the compound concentration as the abscissa and the inhibition rate as the ordinate, performing four-parameter analysis, fitting a dose-effect curve, and calculating IC50The results are shown in Table 1.
TABLE 1
Figure PCTCN2021072699-APPB-000145
Figure PCTCN2021072699-APPB-000146
Test example 3 evaluation of pharmacokinetics in mice
ICR mice (purchased from Shanghai Seipaibikai laboratory animals Co., Ltd.) weighed 18-22 g, were adapted for 3-5 days, and were randomly grouped, 9 mice per group, and each compound was administered by gavage at a dose of 10 mg/kg. The test animals (ICR mice) were fasted overnight before and food 4h after administration, and water was freely available both before and after and during the experiment. After the intragastric administration, 0.1mL of blood is collected from eye sockets for 0.25h (15min), 0.5h (30min), 1h, 2h, 3h, 4h, 6h, 8h, 10h and 24h (3-4 time points are collected from each mouse, 3 mice are collected at each time point), EDTA-K2 is anticoagulated, and the blood plasma is transferred to the conditions of 4 ℃, 4000rpm and 10min within 30min for centrifugal separation. All plasma was collected and immediately stored at-20 ℃ for testing.
Sucking 30 mu L of plasma sample to be detected and standard yeast sample, adding 300 mu L of acetonitrile solution containing an internal standard (diazepam 20ng/mL), shaking and uniformly mixing for 5min, centrifuging at 13000rpm for 10min, taking 70 mu L of supernatant, adding 70 mu L of ultrapure water for dilution, uniformly mixing, sucking 1 mu L of ultrapure water for LC/MS/MS determination, and recording a chromatogram.
Oral exposure of the compounds was assessed by in vivo pharmacokinetic experiments in mice. The relevant pharmacokinetic parameters were fitted using DAS3.2.5 software and the results are shown in Table 2.
TABLE 2
Figure PCTCN2021072699-APPB-000147

Claims (15)

  1. A compound of formula (I) or a pharmaceutically acceptable salt thereof,
    Figure PCTCN2021072699-APPB-100001
    wherein,
    R 1is selected from
    Figure PCTCN2021072699-APPB-100002
    Or
    Figure PCTCN2021072699-APPB-100003
    R 2Selected from hydrogen, 3-10 membered heterocyclyl or amino, wherein said 3-10 membered heterocyclyl or amino is optionally substituted with one or more RaSubstitution;
    R aselected from hydroxy, cyano, halogen,
    Figure PCTCN2021072699-APPB-100004
    C 1-6Alkyl radical, C1-6Alkoxy, di (C)1-6Alkyl) amino, or substituted by one or more hydroxy or C1-6Alkoxy-substituted C1-6An alkyl group;
    R 3is selected from-OCF2H, wherein said-OCF2H is optionally substituted with halogen;
    R 4selected from hydrogen, C1-6Alkyl radical, C3-6Cycloalkyl or 3-10 membered heterocyclyl, wherein C1-6Alkyl radical, C3-6Cycloalkyl or 3-10 membered heterocyclyl is optionally substituted with one or more RbSubstitution;
    R bselected from deuterium, hydroxy, C1-6Alkoxy radical, C1-6Alkyl, amino, mono (C)1-6Alkyl) amino or di (C)1-6Alkyl) amino;
    R 5is selected from C1-6An alkyl group;
    ring a is selected from 5-6 membered heteroaryl or phenyl.
  2. A compound of formula (I) or a pharmaceutically acceptable salt thereof as claimed in claim 1 wherein R2Selected from hydrogen, 4-6 membered heterocyclyl, 7-9 membered spiroheterocyclyl or amino, wherein said 4-6 membered heterocyclyl, 7-9 membered spiroheterocyclyl or amino is optionally substituted with one or more RaSubstitution; or, R2Selected from hydrogen, 4-6 membered heterocyclyl or amino, wherein said 4-6 membered heterocyclyl or amino is optionally substituted with one or more RaSubstitution; or, R2Selected from hydrogen, 4-6 membered heterocycloalkyl, 6 membered heterocycloalkenyl, 7 or 9 membered spiroheterocycloalkyl or amino, wherein said 4-6 membered heterocycloalkyl, 6 membered heterocycloalkenyl, 7 or 9 membered spiroheterocycloalkyl or amino is optionally substituted with one or more RaSubstitution; or, R2Selected from the group consisting of hydrogen, pyrrolidinyl, morpholinyl, piperazinyl, azetidinyl, piperidinyl, 1,2,3, 6-tetrahydropyridinyl, tetrahydropyranyl, 3, 4-dihydropyranyl, 3, 6-dihydropyranyl, 2-oxa-7-azaspiro [3.5]]Nonanyl, 2-oxa-6-azaspiro [3.3]Heptylalkyl group or amino group, wherein the pyrrolidinyl, morpholinyl, piperazinyl, azetidinyl, piperidinyl, 1,2,3, 6-tetrahydropyridinyl, tetrahydropyranyl, 3, 4-dihydropyranyl, 3, 6-dihydropyranyl, 2-oxa-7-azaspiro [3.5]]Nonanyl, 2-oxa-6-azaspiro [3.3]Heptylalkyl or amino optionally substituted with one or more RaSubstitution; or, R2Selected from hydrogen,
    Figure PCTCN2021072699-APPB-100005
    Figure PCTCN2021072699-APPB-100006
    Figure PCTCN2021072699-APPB-100007
  3. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 or 2 wherein RaSelected from hydroxy, halogen, C1-6Alkyl, di (C)1-6Alkyl) amino or C substituted by one or more hydroxy groups1-6An alkyl group; or, RaSelected from hydroxy, cyano, halogen,
    Figure PCTCN2021072699-APPB-100008
    C 1-4Alkyl radical, C1-3Alkoxy, di (C)1-3Alkyl) amino groups or substituted by one or more hydroxy groups or C1-3Alkoxy-substituted C1-4An alkyl group; or, RaSelected from hydroxy, halogen, C1-3Alkyl, di (C)1-3Alkyl) amino or C substituted by one or more hydroxy groups1-3An alkyl group; or, RaSelected from hydroxy, cyano, fluoro,
    Figure PCTCN2021072699-APPB-100009
    Methyl, methoxy, 2-hydroxyethyl, 2-methoxyethyl, 2-methyl-2-hydroxypropyl, di (methyl) amino or hydroxymethyl.
  4. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-3, wherein R3Is selected from-OCF3or-OCF2Cl。
  5. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-4 wherein R4Selected from hydrogen, C1-5Alkyl radical, C3-6Cycloalkyl or 4-6 membered heterocycloalkyl, wherein C1-5Alkyl radical, C3-6Cycloalkyl or 4-6 membered heterocycloalkyl optionally substituted with one or moreR isbSubstitution; or, R4Selected from hydrogen, C1-5Alkyl, cyclopropyl or 6-membered heterocycloalkyl, wherein C1-5Alkyl, cyclopropyl or 6-membered heterocycloalkyl optionally substituted with one or more RbSubstitution; or, R4Selected from hydrogen, methyl, ethyl, 2-methylpropyl, 3-methylbutyl, cyclopropyl, tetrahydropyranyl or piperidinyl, wherein methyl, ethyl, 2-methylpropyl, 3-methylbutyl, cyclopropyl, tetrahydropyranyl or piperidinyl is optionally substituted by one or more RbSubstitution; or, R4Selected from hydrogen, methyl, d3-methyl, cyclopropyl,
    Figure PCTCN2021072699-APPB-100010
    Figure PCTCN2021072699-APPB-100011
  6. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-5 wherein RbSelected from hydroxy, C1-6Alkoxy radical, C1-6Alkyl, amino, mono (C)1-6Alkyl) amino or di (C)1-6Alkyl) amino; or, RbSelected from deuterium, hydroxy, C1-3Alkoxy radical, C1-3Alkyl, amino, mono (C)1-3Alkyl) amino or di (C)1-3Alkyl) amino; or, RbSelected from hydroxy, C1-3Alkoxy radical, C1-3Alkyl, amino, mono (C)1-3Alkyl) amino or di (C)1-3Alkyl) amino; or, RbSelected from deuterium, hydroxy, methoxy, methyl or di (ethyl) amino.
  7. A compound of formula (I) as claimed in any one of claims 1 to 6 or a pharmaceutical thereofThe above acceptable salt, wherein R5Is selected from C1-3An alkyl group; or, R5Selected from methyl.
  8. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-7 wherein ring a is selected from 5-6 membered nitrogen containing heteroaryl or phenyl; alternatively, ring a is selected from pyrrolyl, pyridyl or phenyl.
  9. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-8 wherein the structural unit
    Figure PCTCN2021072699-APPB-100012
    Is selected from
    Figure PCTCN2021072699-APPB-100013
    Or, a structural unit
    Figure PCTCN2021072699-APPB-100014
    Is selected from
    Figure PCTCN2021072699-APPB-100015
    Is further selected from
    Figure PCTCN2021072699-APPB-100016
  10. The compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-9, which is a compound of formula (II) or a compound of formula (III),
    Figure PCTCN2021072699-APPB-100017
    wherein,
    R 3is selected from-OCF3or-OCF2Cl; or
    Figure PCTCN2021072699-APPB-100018
    Wherein,
    structural unit
    Figure PCTCN2021072699-APPB-100019
    Is selected from
    Figure PCTCN2021072699-APPB-100020
    R 3Is selected from-OCF3or-OCF2Cl。
  11. A compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein,
    R 1is selected from
    Figure PCTCN2021072699-APPB-100021
    Or
    Figure PCTCN2021072699-APPB-100022
    R 2Selected from hydrogen, 4-10 membered heterocyclyl or amino containing 1-3 heteroatoms selected from N or O or S, wherein said 4-10 membered heterocyclyl or amino is optionally substituted with one or more RaSubstitution;
    R aselected from hydroxy, cyano, halogen,
    Figure PCTCN2021072699-APPB-100023
    C 1-6Alkyl radical, C1-6Alkoxy, di (C)1-6Alkyl) amino, or substituted by one or more hydroxy or C1-6Alkoxy-substituted C1-6An alkyl group;
    R 3is selected from-OCF3、-OCF 2Cl or-OCF2Br;
    R 4Selected from hydrogen, C1-6Alkyl radical, C3-6Cycloalkyl, or 5-7 membered heterocyclyl containing 1-3 heteroatoms selected from N or O or S, wherein C1-6Alkyl radical, C3-6Cycloalkyl or 5-7 membered heterocyclyl is optionally substituted with one or more RbSubstitution;
    R bselected from deuterium, hydroxy, C1-6Alkoxy radical, C1-6An alkyl group;
    R 5is selected from C1-6An alkyl group;
    ring A is selected from 5-6 membered heteroaryl or phenyl containing 1-3 heteroatoms selected from N or O or S.
  12. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-11, wherein the compound of formula (I) is selected from:
    Figure PCTCN2021072699-APPB-100024
    Figure PCTCN2021072699-APPB-100025
    Figure PCTCN2021072699-APPB-100026
    Figure PCTCN2021072699-APPB-100027
  13. a compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-12, wherein the compound of formula (I) is selected from:
    Figure PCTCN2021072699-APPB-100028
    Figure PCTCN2021072699-APPB-100029
  14. a pharmaceutical composition comprising a compound of any one of claims 1-13, or a pharmaceutically acceptable salt thereof.
  15. Use of a compound according to any one of claims 1 to 13, or a pharmaceutically acceptable salt thereof, a pharmaceutical composition according to claim 14, for the manufacture of a medicament for the treatment and/or prevention of a BCR-ABL associated disease; optionally, the disease is selected from cancer; alternatively, the disease is selected from leukemia; alternatively, the disease is selected from chronic myeloid leukemia.
CN202180006423.2A 2020-01-19 2021-01-19 Compounds as BCR-ABL inhibitors Pending CN114728943A (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
CN2020100623173 2020-01-19
CN202010062317 2020-01-19
CN202011246125 2020-11-10
CN2020112461254 2020-11-10
PCT/CN2021/072699 WO2021143927A1 (en) 2020-01-19 2021-01-19 Compound acting as bcr-abl inhibitor

Publications (1)

Publication Number Publication Date
CN114728943A true CN114728943A (en) 2022-07-08

Family

ID=76864757

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180006423.2A Pending CN114728943A (en) 2020-01-19 2021-01-19 Compounds as BCR-ABL inhibitors

Country Status (2)

Country Link
CN (1) CN114728943A (en)
WO (1) WO2021143927A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL308614A (en) * 2021-05-28 2024-01-01 Chia Tai Tianqing Pharmaceutical Group Co Ltd Compound used as bcr-abl inhibitor
WO2024156938A1 (en) 2023-01-28 2024-08-02 Orion Corporation Cbl-b inhibitors

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104302638A (en) * 2012-05-15 2015-01-21 诺华股份有限公司 Benzamide derivatives for inhibiting the activity of abl1, abl2 and bcr-abl1
CN104334529A (en) * 2012-05-15 2015-02-04 诺华股份有限公司 Compounds and compositions for inhibiting the activity of ABL1, ABL2 AND BCR-ABL1
CN104379574A (en) * 2012-05-15 2015-02-25 诺华股份有限公司 Benzamide derivatives for inhibiting the activity of ABL1, ABL 2 and BCR-ABL 1
WO2017186148A1 (en) * 2016-04-29 2017-11-02 Astar Biotech Llc Novel heterocyclic compounds as tyrosine kinase bcr-abl inhibitors
WO2018133826A1 (en) * 2017-01-20 2018-07-26 深圳市塔吉瑞生物医药有限公司 (hetero)arylamide compound for inhibiting protein kinase activity

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104302638A (en) * 2012-05-15 2015-01-21 诺华股份有限公司 Benzamide derivatives for inhibiting the activity of abl1, abl2 and bcr-abl1
CN104334529A (en) * 2012-05-15 2015-02-04 诺华股份有限公司 Compounds and compositions for inhibiting the activity of ABL1, ABL2 AND BCR-ABL1
CN104379574A (en) * 2012-05-15 2015-02-25 诺华股份有限公司 Benzamide derivatives for inhibiting the activity of ABL1, ABL 2 and BCR-ABL 1
WO2017186148A1 (en) * 2016-04-29 2017-11-02 Astar Biotech Llc Novel heterocyclic compounds as tyrosine kinase bcr-abl inhibitors
WO2018133826A1 (en) * 2017-01-20 2018-07-26 深圳市塔吉瑞生物医药有限公司 (hetero)arylamide compound for inhibiting protein kinase activity

Also Published As

Publication number Publication date
WO2021143927A1 (en) 2021-07-22

Similar Documents

Publication Publication Date Title
CN112778276A (en) Compound as SHP2 inhibitor and application thereof
EP4148056A1 (en) Sos1 inhibitor containing phosphorus
CN109641897B (en) Bcl-2 selective inhibitors, their preparation and use
CN113527335A (en) Macrocyclic compound as EGFR inhibitor and application thereof
JP2022506442A (en) Trifluoromethyl Substituted Sulfamide-based Selective BCL-2 Inhibitor
CN113825754B (en) Disubstituted sulfonamide selective BCL-2 inhibitors including methyl and trifluoromethyl
CN114728943A (en) Compounds as BCR-ABL inhibitors
TW202325300A (en) Fused imide derivative
CN112867717A (en) Compounds useful as kinase inhibitors and uses thereof
CN114907326A (en) Amido and bicyclic inhibitors of TYK2
CN116547276A (en) TYK2 inhibitor compounds containing an amide group and a heterocycloalkyl group
CN108329274B (en) Bruton's tyrosine kinase inhibitors
CN114685532A (en) Macrocyclic compound and medical application thereof
CN115433179A (en) Benzopyrimidine compounds and medical application thereof
EP4371981A1 (en) Sulfur/phosphorus-containing aryl compound and application thereof
EP4177258A1 (en) Arylphosphine oxide compounds and use thereof
EP4155304A1 (en) Compound used as ret kinase inhibitor and application thereof
CN113666877A (en) Alkoxy and amido containing TYK2 inhibitor compound
CN114805438A (en) Phosphoacyl-containing TYK2 inhibitor compounds
CN110776511A (en) Quinoline ring-containing BTK inhibitors
IL308614A (en) Compound used as bcr-abl inhibitor
WO2023072248A1 (en) Pyridyl-containing compound
CN116891468A (en) Thiazole compound and application thereof
CN115925689A (en) Cyclopropyl-containing nuclear export protein inhibitors
CN116768870A (en) Compound with benzyloxy aryl ether structure, preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination