CN108341835B - Boron-containing compounds as tyrosine kinase inhibitors - Google Patents

Boron-containing compounds as tyrosine kinase inhibitors Download PDF

Info

Publication number
CN108341835B
CN108341835B CN201710047432.1A CN201710047432A CN108341835B CN 108341835 B CN108341835 B CN 108341835B CN 201710047432 A CN201710047432 A CN 201710047432A CN 108341835 B CN108341835 B CN 108341835B
Authority
CN
China
Prior art keywords
compound
formula
pharmaceutically acceptable
acceptable salt
hydrogen
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.)
Active
Application number
CN201710047432.1A
Other languages
Chinese (zh)
Other versions
CN108341835A (en
Inventor
张寅生
高勇
任景
王庆璘
赵大敏
周宇
吴喆阳
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
Priority to CN201710047432.1A priority Critical patent/CN108341835B/en
Publication of CN108341835A publication Critical patent/CN108341835A/en
Application granted granted Critical
Publication of CN108341835B publication Critical patent/CN108341835B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic Table
    • C07F5/02Boron compounds
    • C07F5/025Boronic and borinic acid compounds

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present application is in the field of medicine and specifically relates to boron containing compounds useful as tyrosine kinase inhibitors, processes for their preparation, pharmaceutical compositions containing them and their use in the treatment of tyrosine kinase mediated cell proliferative disorders. The compound has good inhibition effect on tyrosine kinase.

Description

Boron-containing compounds as tyrosine kinase inhibitors
Technical Field
The application belongs to the field of medicines, and particularly relates to a compound shown in a formula (I), a preparation method thereof, a pharmaceutical composition containing the compound, and application of the compound in treating cell proliferative diseases mediated by tyrosine kinase.
Background
Cancer, one of the leading causes of morbidity and mortality worldwide, is a cell proliferative disease. Protein kinases play a very important role in regulating cell growth, proliferation, invasion, differentiation and the like. One of the major mechanisms by which protein kinases achieve cellular regulation is through transmembrane extracellular signal transduction, which in turn regulates biochemical pathways within the cell. Protein phosphorylation represents a process by which intracellular signals propagate from molecule to molecule, ultimately leading to a cellular response. Protein phosphorylation occurs primarily at serine, threonine or tyrosine residues, and protein kinases have therefore been classified by the specificity of their phosphorylation sites, i.e., serine/threonine kinases and tyrosine kinases. Since phosphorylation is a ubiquitous process in cells and since cellular phenotype is largely influenced by the activity of these pathways, protein kinase inhibitors have important uses in the treatment of human and animal diseases. Among them, tyrosine kinases include, but are not limited to, Epidermal Growth Factor Receptor (EGFR) tyrosine kinase, Anaplastic Lymphoma Kinase (ALK), and Janus kinase (JAK).
Among them, EGFR is a member of the erbB receptor family of transmembrane protein tyrosine kinases, and forms homodimers on cell membranes by binding to its ligand (e.g., the endogenous epidermal growth factor EGF). The formation of these dimers can lead to phosphorylation of key tyrosine residues within the cell, thereby activating multiple downstream signaling pathways within the cell. These intracellular signaling pathways play important regulatory roles in cell proliferation, survival, and anti-apoptotic processes. Dysregulation of EGFR signal transduction pathways, including increased expression of ligands and receptors, EGFR gene amplification and mutation, can promote transformation of normal cells into tumor cells, and play an important role in proliferation, invasion, metastasis and angiogenesis of tumor cells. Therefore, EGFR is an important target for the development of anti-tumor drugs.
Among them, ALK is a cell membrane-spanning Receptor Tyrosine Kinase (RTK), and belongs to the insulin receptor subfamily. Intact ALK has the typical RTK three-part structure, an extracellular region, a lipophilic transmembrane region, and an intracellular tyrosine kinase. ALK fusion proteins, mutations and overexpression are associated with a variety of diseases, such as non-small cell lung cancer, intestinal cancer or renal cancer.
Among them, Janus kinases (JAKs) are a family of tyrosine kinases consisting of JAK1, JAK2, JAK3 and TYK 2. JAKs play a crucial role in cytokine signaling. Downstream substrates of JAK family kinases include transcriptional signal sensing and activation (STAT) proteins. JAK kinases are associated with a variety of diseases, for example JAK3 is associated with many aberrant immune responses, solid tumors or hematologic malignancies; JAK2, for example, is associated with myeloproliferative diseases such as polycythemia vera.
In view of the important role of tyrosine kinase in vivo, the development of tyrosine kinase inhibitors suitable for being used as therapeutic drugs is particularly important, and the invention unexpectedly discovers that a series of boron-containing compounds have good inhibition effect on tyrosine kinase.
Disclosure of Invention
In one aspect, the present application relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof,
Figure BDA0001216697810000021
wherein the content of the first and second substances,
R1selected from hydrogen, fluorine, chlorine, bromine, iodine, alkyl or alkoxyWherein alkyl is optionally substituted with fluoro, chloro, bromo or iodo;
x is selected from NH or O;
R4and R6Each independently selected from hydrogen, alkyl, alkoxy or-NHC (═ O) Ra
R3And R7Each independently selected from hydrogen or alkoxy;
R5selected from hydrogen, alkylamino, alkyl, cycloalkyl, alkoxy or heterocycloalkyl, wherein said alkylamino, alkyl, cycloalkyl, alkoxy or heterocycloalkyl is optionally substituted with: o, -S (O)2RbAlkyl, -NRcRdOr a heterocycloalkyl group;
Raselected from alkyl, alkenyl, alkynyl, cyano or cycloalkyl;
Rbselected from alkyl groups;
Rcand RdEach independently selected from hydrogen, alkyl, alkenyl or cycloalkyl;
cy is selected from
Figure BDA0001216697810000022
R2Each independently selected from fluoro, chloro, bromo, iodo or alkoxy;
n is 0 to 4;
Z1and Z2Each independently selected from hydrogen or C1-C6An alkyl group;
the A ring is a 5-6 membered ring, which is optionally substituted with alkyl.
In some embodiments, R1Selected from hydrogen, fluorine, chlorine, bromine, iodine, C1-C6Alkyl or C1-C6Alkoxy radical, wherein C1-C6Alkyl is optionally substituted with fluorine, chlorine, bromine or iodine.
In some preferred embodiments, R1Selected from hydrogen, fluorine, chlorine, bromine, C1-C3Alkyl or C1-C3Alkoxy radical, wherein C1-C3Alkyl is optionally substituted with fluorine.
In some more preferred embodiments, R1Selected from hydrogen, fluoro, chloro, bromo, methyl or methoxy, wherein methyl is optionally substituted with 1-3 fluoro.
In some most preferred embodiments, R1Selected from hydrogen, fluorine, chlorine, bromine, methyl, methoxy or trifluoromethyl.
In some most preferred embodiments, R1Selected from hydrogen, fluorine, chlorine, bromine or methyl.
In some embodiments, R4And R6Each independently selected from hydrogen and C1-C6Alkyl radical, C1-C6Alkoxy or-NHC (═ O) Ra
In some preferred embodiments, R4And R6Each independently selected from hydrogen and C1-C3Alkyl radical, C1-C3Alkoxy or-NHC (═ O) Ra
In some more preferred embodiments, R4And R6Each independently selected from hydrogen, methyl or-NHC (═ O) Ra
In some embodiments, RaIs selected from C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, cyano or C3-C6Cycloalkyl, preferably C1-C3Alkyl radical, C2-C3Alkenyl radical, C2-C3Alkynyl, cyano or C3-C6Cycloalkyl, more preferably C2-C3Alkenyl, most preferably vinyl.
In some embodiments, R4And R6At least one is selected from hydrogen.
In some most preferred embodiments, R4And R6Each independently selected from hydrogen, methyl or
Figure BDA0001216697810000031
And R is4And R6At least one is selected from hydrogen.
In some most preferred embodiments, R4And R6Each independently selected from hydrogen or
Figure BDA0001216697810000032
And R is4And R6At least one is selected from hydrogen.
In some most preferred embodiments, R4And R6Each independently selected from hydrogen or methyl, and R4And R6At least one is selected from hydrogen.
In some embodiments, R3And R7Each independently selected from hydrogen or C1-C6An alkoxy group.
In some preferred embodiments, R3And R7Each independently selected from hydrogen or C1-C3An alkoxy group.
In some more preferred embodiments, R3And R7Each independently selected from hydrogen, methoxy or isopropyloxy.
In some embodiments, R3And R7At least one is selected from hydrogen.
In some most preferred embodiments, R3And R7Each independently selected from hydrogen, methoxy or isopropyloxy, and R3And R7At least one is selected from hydrogen. In some most preferred embodiments, R3And R7Each independently selected from hydrogen or methoxy, and R3And R7At least one is selected from hydrogen.
In some embodiments, R5Selected from hydrogen, C1-C6Alkylamino radical, C1-C6Alkyl radical, C3-C6Cycloalkyl radical, C1-C6Alkoxy or 3-6 membered heterocycloalkyl, wherein said C1-C6Alkylamino radical, C1-C6Alkyl radical, C3-C6Cycloalkyl radical, C1-C6Alkoxy or 3-6 membered heterocycloalkyl optionally substituted with: o, -S (O)2Rb、C1-C6Alkyl, -NRcRdOr optionally substituted 3-6 membered heterocycloalkyl.
In some preferred embodiments, R5Selected from hydrogen, C1-C3Alkylamino radical, C1-C3Alkyl radical, C3-C6Cycloalkyl radical, C1-C3Alkoxy or 5-6 membered heterocycloalkyl, wherein said C1-C3Alkylamino radical, C1-C3Alkyl radical, C3-C6Cycloalkyl radical, C1-C3Alkoxy or 5-6 membered heterocycloalkyl optionally substituted with: o, -S (O)2Rb、C1-C6Alkyl, -NRcRdOr optionally substituted 5-6 membered heterocycloalkyl.
In some more preferred embodiments, R5Selected from the group consisting of hydrogen, ethylamino, ethoxy, tetrahydrofuranyl, tetrahydrothienyl, pyrrolidinyl, tetrahydropyrazolyl, piperidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, morpholinyl, piperazinyl, 1, 4-thiaxanyl, 1, 4-dioxanyl, thiomorpholinyl, or 1, 4-dithianyl, wherein said ethylamino, ethoxy, tetrahydrofuranyl, tetrahydrothienyl, pyrrolidinyl, tetrahydropyrazolyl, piperidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, morpholinyl, piperazinyl, 1, 4-thiaxanyl, 1, 4-dioxanyl, thiomorpholinyl, or 1, 4-dithianyl is optionally substituted with: o, -S (O)2Rb、C1-C3Alkyl, -NRcRdOr optionally is covered with C1-C6Alkyl-substituted 5-6 membered heterocycloalkyl.
In some more preferred embodiments, R5Selected from hydrogen, ethylamino, ethoxy, morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl; wherein ethoxy is optionally substituted by-NRcRdPyrrolidinyl, tetrahydropyrazolyl, piperidinyl, or piperazinyl; wherein morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl is optionally substituted with: o, -S (O)2Rb、C1-C3Alkyl, -NRcRdOr optionally is covered with C1-C3An alkyl-substituted 5-6 membered nitrogen-containing heterocycloalkyl group; wherein ethylamino is optionally substituted by C1-C3Alkyl or-NRcRdAnd (4) substitution.
In some embodiments, RbIs selected from C1-C6An alkyl group.
In some more preferred embodiments, RbIs selected from C1-C3An alkyl group.
In some embodiments, RcAnd RdEach independently selected from hydrogen and C1-C6Alkyl radical, C2-C6Alkenyl or C3-C6A cycloalkyl group.
In some preferred embodiments, RcAnd RdEach independently selected from hydrogen or C1-C3An alkyl group.
In some more preferred embodiments, R5Selected from hydrogen, ethylamino, ethoxy, morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl; wherein ethoxy is optionally substituted by-N (CH)3)2Or
Figure BDA0001216697810000041
Substitution; wherein morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl is optionally substituted by ═ O,
Figure BDA0001216697810000042
Methyl, -N (CH)3)2Optionally C1-C3Pyrrolidinyl substituted by alkyl, optionally C1-C3Alkyl-substituted tetrahydropyrazolyl, optionally substituted by C1-C3Alkyl-substituted piperidinyl or optionally substituted by C1-C3Alkyl substituted piperazinyl substitution; wherein ethylamino is optionally substituted by methyl or-N (CH)3)2And (4) substitution.
In some more preferred embodiments, R5Selected from hydrogen, ethylamino, ethoxy,
Figure BDA0001216697810000043
Figure BDA0001216697810000044
Wherein ethoxy is optionally substituted by-N (CH)3)2Or
Figure BDA0001216697810000045
Substitution; wherein
Figure BDA0001216697810000046
Figure BDA0001216697810000047
Optionally substituted by ═ O,
Figure BDA0001216697810000048
Methyl, -N (CH)3)2Pyrrolidinyl, tetrahydropyrazolyl, piperidinyl, piperazinyl, 4-methylpiperazinyl; wherein ethylamino is optionally substituted by methyl or-N (CH)3)2And (4) substitution.
In some more preferred embodiments, R5Selected from hydrogen, ethylamino, ethoxy,
Figure BDA0001216697810000049
Figure BDA00012166978100000410
Wherein ethoxy is optionally substituted by-N (CH)3)2Or
Figure BDA00012166978100000411
Substitution; wherein
Figure BDA00012166978100000412
Optionally substituted by methyl or
Figure BDA00012166978100000413
Substitution; wherein
Figure BDA00012166978100000414
Optionally piperazinyl, 4-methylpiperazinyl or-N (CH)3)2Substitution; wherein
Figure BDA00012166978100000415
Optionally substituted with ═ O; in which the ethylamino group is simultaneously substituted by one methyl group and one-N (CH)3)2And (4) substitution.
In some most preferred embodiments, R5Selected from hydrogen, ethylamino or ethoxy; wherein ethoxy is optionally substituted by-N (CH)3)2Substitution; wherein ethylamino is optionally substituted by methyl or-N (CH)3)2And (4) substitution.
In some most preferred embodiments, R5Selected from hydrogen, ethoxy,
Figure BDA00012166978100000416
Figure BDA00012166978100000417
Wherein ethoxy is optionally substituted
Figure BDA00012166978100000418
Substitution; wherein
Figure BDA00012166978100000419
Optionally substituted by methyl or
Figure BDA0001216697810000051
Substitution; wherein
Figure BDA0001216697810000052
Optionally is covered with
Figure BDA0001216697810000053
or-N (CH)3)2Substitution; wherein
Figure BDA0001216697810000054
Optionally substituted by ═ O.
In some most preferred embodiments, R5Selected from hydrogen,
Figure BDA0001216697810000055
Figure BDA0001216697810000056
In some embodiments, R2Each independently selected from fluorine, chlorine, bromine, iodine or C1-C6An alkoxy group.
In some preferred embodiments, R2Each independently selected from fluorine, chlorine, bromine, iodine or C1-C3An alkoxy group.
In some more preferred embodiments, R2Each independently selected from fluorine, chlorine, bromine, iodine.
In some more preferred embodiments, R2Each independently selected from methoxy, ethoxy or isopropoxy.
In some embodiments, n is 0or 1.
In some embodiments, Cy is
Figure BDA0001216697810000057
Comprises that
Figure BDA0001216697810000058
Preferably, it is
Figure BDA0001216697810000059
In some embodiments, Cy is
Figure BDA00012166978100000510
Comprises that
Figure BDA00012166978100000511
Preferably, it is
Figure BDA00012166978100000512
In some embodiments, Cy is
Figure BDA00012166978100000513
Comprises that
Figure BDA00012166978100000514
Figure BDA0001216697810000061
Preferably, it is
Figure BDA0001216697810000062
Figure BDA0001216697810000063
Wherein m is 1 or 2, R8And R9Each independently selected from hydrogen or C1-C6Alkyl, preferably hydrogen or C1-C3Alkyl, most preferably hydrogen or methyl.
In some embodiments, Cy is
Figure BDA0001216697810000064
Comprises that
Figure BDA0001216697810000065
Figure BDA0001216697810000066
Preferably, it is
Figure BDA0001216697810000067
Figure BDA0001216697810000068
R8And R9Each independently selected from hydrogen or C1-C3Alkyl, preferably hydrogen or methyl, most preferably hydrogen.
In some embodiments, Cy is
Figure BDA0001216697810000069
Figure BDA00012166978100000610
In some embodiments, the present application relates to a compound of formula (II) or a compound of formula (III), or a pharmaceutically acceptable salt thereof,
Figure BDA00012166978100000611
wherein the content of the first and second substances,
R1、R3、R4、R5、R6、R7and X is as defined for compounds of formula (I);
structural unit
Figure BDA0001216697810000071
The definition of (A) is the same as that of the compound of formula (I).
In some embodiments, the present application relates to compounds of formula (IV) or a pharmaceutically acceptable salt thereof,
Figure BDA0001216697810000072
wherein the content of the first and second substances,
R1、R3、R5、R7and Cy is as defined for compounds of formula (I).
In some embodiments, the present application relates to a compound of formula (V) or formula (VI), or a pharmaceutically acceptable salt thereof,
Figure BDA0001216697810000073
wherein the content of the first and second substances,
R1、R3、R5、R7and X is as defined for compounds of formula (I);
structural unit
Figure BDA0001216697810000074
The definition of (A) is the same as that of the compound of formula (I).
In some embodiments, the present application relates to compounds of formula (VII), or a pharmaceutically acceptable salt thereof,
Figure BDA0001216697810000075
wherein the content of the first and second substances,
R1、R3、R4、R6、R7x and Cy are as defined for compounds of formula (I);
Y1is selected from N or CH;
Y2selected from NH, CH2S or O;
q is 0 to 11;
R12selected from ═ O, -S (═ O)2RbAlkyl, -NRcRdOr optionally substituted heterocycloalkyl;
Rbselected from alkyl groups;
Rcand RdEach independently selected from hydrogen, alkyl or cycloalkyl.
In some embodiments, R12Selected from ═ O, -S (═ O)2Rb、C1-C6Alkyl, -NRcRdOr optionally substituted 3-6 membered heterocycloalkyl.
In some preferred embodiments, R12Selected from ═ O, -S (═ O)2Rb、C1-C3Alkyl, -NRcRdOr optionally substituted 5-6 membered heterocycloalkyl.
In some more preferred embodiments, R12Selected from ═ O, -S (═ O)2Rb、C1-C3Alkyl, -NRcRdOr optionally is covered with C1-C6Alkyl-substituted 5-6 membered heterocycloalkyl.
In some more preferred embodiments, R12Selected from ═ O, -S (═ O)2Rb、C1-C3Alkyl, -NRcRdOr optionally is covered with C1-C3Alkyl-substituted 5-6 membered heterocycloalkyl.
In some more preferred embodiments, R12Selected from ═ O, -S (═ O)2Rb、C1-C3Alkyl, -NRcRdOr optionally is covered with C1-C3An alkyl-substituted 5-6 membered nitrogen-containing heterocycloalkyl group.
In some embodiments, RbIs selected from C1-C6An alkyl group.
In some more preferred embodiments, RbIs selected from C1-C3An alkyl group.
In some embodiments, RcAnd RdEach independently selected from hydrogen and C1-C6Alkyl or C3-C6A cycloalkyl group.
In some preferred embodiments, RcAnd RdEach independently selected from hydrogen or C1-C3An alkyl group.
In some more preferred embodiments, R12Is selected from ═ O,
Figure BDA0001216697810000081
Methyl, -N (CH)3)2Optionally C1-C3Piperazinyl substituted by alkyl, optionally C1-C3Pyrrolidinyl substituted by alkyl, optionally C1-C3Alkyl-substituted tetrahydropyrazolyl or optionally substituted by C1-C3Alkyl-substituted piperidinyl.
In some more preferred embodiments, R12Is selected from ═ O,
Figure BDA0001216697810000082
Methyl or-N (CH)3)2
In some preferred embodiments, q is 0 to 6.
In some more preferred embodiments, q is 0or 1.
In some more preferred embodiments, q is 0.
In some more preferred embodiments, q is 1, and R is12And Y2And (4) connecting.
In some most preferred embodiments, the building blocks
Figure BDA0001216697810000083
Is selected from
Figure BDA0001216697810000084
Figure BDA0001216697810000085
It will be appreciated that the choice of q does not violate the valence bond joining rule, e.g. R when q is 1112Cannot be selected from ═ O, Y2Nor can it be selected from O.
In some embodiments, the application relates to a compound of formula (VIII) or formula (IX), or a pharmaceutically acceptable salt thereof,
Figure BDA0001216697810000091
wherein the content of the first and second substances,
R1、R3、R4、R6、R7and X is as defined for compounds of formula (I);
R12、Y1、Y2and q is as defined for compound of formula (VII);
structural unit
Figure BDA0001216697810000092
The definition of (A) is the same as that of the compound of formula (I).
In some embodiments, the present application relates to the following compounds, or pharmaceutically acceptable salts thereof:
Figure BDA0001216697810000093
Figure BDA0001216697810000101
Figure BDA0001216697810000111
in some embodiments, the present application relates to the following compounds:
Figure BDA0001216697810000121
in another aspect, the present application relates to a pharmaceutical composition comprising a compound of formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI), formula (VII), formula (VIII) or formula (IX) or a pharmaceutically acceptable salt thereof, as described herein. In some embodiments, the pharmaceutical compositions of the present application further comprise a pharmaceutically acceptable excipient.
In another aspect, the present application relates to a method of treating a tyrosine kinase mediated disease in a mammal, comprising administering to a mammal, preferably a human, in need of such treatment a therapeutically effective amount of a compound of formula (i), formula (ii), formula (iii), formula (iv), formula (v), formula (vi), formula (vii), formula (viii) or formula (ix), including but not limited to EGFR tyrosine kinase, ALK or JAK; such diseases include, but are not limited to, cell proliferative diseases (e.g., cancer).
In another aspect, the present application relates to a method of treating a disease mediated by EGFR tyrosine kinase in a mammal, comprising administering to a mammal, preferably a human, in need of such treatment a therapeutically effective amount of a compound of formula (iv), (v) or (vi) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. Such diseases include, but are not limited to, cell proliferative diseases (e.g., cancer).
In another aspect, the application relates to a method of treating a disease mediated by ALK or JAK in a mammal, including but not limited to a cell proliferative disease (e.g., cancer), comprising administering to a mammal, preferably a human, in need of such treatment a therapeutically effective amount of a compound of formula (vii), formula (viii), or formula (ix), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
In yet another aspect, the present application relates to the use of a compound of formula (i), formula (ii), formula (iii), formula (iv), formula (v), formula (vi), formula (vii), formula (viii) or formula (ix), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for the treatment of a disease mediated by a tyrosine kinase, including but not limited to EGFR tyrosine kinase, ALK or JAK, including but not limited to a cell proliferative disease such as cancer.
In yet another aspect, the present application relates to the use of a compound of formula (iv), (v) or (vi), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for the treatment of a disease mediated by EGFR tyrosine kinase, including but not limited to cell proliferative disorders (e.g., cancer).
In a further aspect, the application relates to the use of a compound of formula (vii), formula (viii) or formula (ix), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for the treatment of a disease mediated by ALK or JAK, including but not limited to cell proliferative disorders (e.g., cancer).
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.
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.
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. For example, ethyl is "optionally" substituted with fluoro or chloro, meaning that ethyl may be unsubstituted (e.g., -CH2CH3) Monosubstituted (e.g. -CH)2CH2F、CHClCH2F) Polysubstituted (e.g. -CHFCH)2F、-CHClCH2F、-CH2CHCl2、-CH2CHF2Etc.) or fully substituted (-CFClCF)3、-CF2CF3). 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; "C3-6By "is meant that the group may have 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, or 6 carbon atoms.
The dotted line (- - - - -) in the structural unit or the substituent group herein indicates that the atom in the structural unit or the substituent group and the substituted group may form a covalent bond when bonded to a specific atom. For example,
Figure BDA0001216697810000131
quilt
Figure BDA0001216697810000132
Substituted, meaning a substituent group
Figure BDA0001216697810000133
Wherein the C atom to which is attached may be substituted with a substituent
Figure BDA0001216697810000134
Any atom on the above forms a covalent bond, and the formed group includes, but is not limited to, those not violating the valence bond connection rule
Figure BDA0001216697810000135
Figure BDA0001216697810000136
When- - -is not attached to a particular atom (e.g.,
Figure BDA0001216697810000137
) Denotes- -any atom which may be attached to the structural unitWithout violating the valence bond connection rules. Thus, for example, structural units
Figure BDA0001216697810000141
Comprises that
Figure BDA0001216697810000142
Also include, for example
Figure BDA0001216697810000143
And the like.
When any variable (e.g. R)2、R8、R9Or R12) When a compound occurs more than one time in its composition or structure, its definition in each case is independent. Thus, for example, (R)2)nRepresents a group represented by n R2Substituted, then each R2All have independent options; specifically, for example, when n ═ 2, it represents that one group is substituted with 2R2And each R is substituted2There is a separate option.
The term "alkylamino" refers to the group-NH-alkyl.
The term "trifluoromethyl" refers to-CF3A group.
The term "alkyl" refers to a group of formula CnH2n+1A hydrocarbon group of (1). 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 of an alkoxy group (i.e., alkyl) has the same definition as above; the term "C1-3Alkyl "refers to an alkyl group containing 1 to 3 carbon atoms (e.g., methyl, ethyl, n-propyl, isopropyl).
The term "alkoxy" refers to-O-alkyl.
The term "alkylamino" refers to-NH-alkyl.
The term "alkenyl" refers to a straight or branched chain of at least one carbon and hydrogen atomUnsaturated aliphatic hydrocarbon group having double bonds. Non-limiting examples of alkenyl groups include, but are not limited to, ethenyl, 1-propenyl, 2-propenyl, 1-butenyl, isobutenyl, 1, 3-butadienyl, and the like. For example, the term "C2-6Alkyl "refers to an alkenyl group containing 2 to 6 carbon atoms; the term "C2-C3Alkenyl "means an alkenyl group having 2 to 3 carbon atoms (e.g., ethenyl, 1-propenyl, 2-propenyl).
The term "cycloalkyl" refers to a carbon ring that is fully saturated and may exist as a single ring, a bridged ring, or a spiro ring. Unless otherwise indicated, the carbocycle is typically a 3 to 10 membered ring. Non-limiting examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornyl (bicyclo [2.2.1] heptyl), bicyclo [2.2.2] octyl, adamantyl, and the like.
The term "heterocycloalkyl" refers to a cyclic group that is fully saturated and may exist as a monocyclic, bicyclic, or spiro ring. Unless otherwise indicated, the heterocyclic ring is typically a 3 to 7 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, thietanyl, examples of 5-membered heterocycloalkyl include, but are not limited to, tetrahydrofuranyl, tetrahydrothienyl, pyrrolidinyl, isoxazolidinyl, oxazolidinyl, isothiazolidinyl, thiazolidinyl, imidazolidinyl, tetrahydropyrazolyl, pyrrolinyl, examples of 6-membered heterocycloalkyl include, but are not limited to, piperidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, morpholinyl, piperazinyl, 1, 4-thiaxanyl, 1, 4-dioxanyl, thiomorpholinyl, 1, 2-dithianyl, 1, 4-dithianyl, tetrahydropyranyl, 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 "treating" means administering a compound or formulation described herein to prevent, ameliorate or eliminate a disease or one or more symptoms associated with the disease, and includes:
(i) preventing the occurrence of a disease or condition in a mammal, particularly when such mammal is susceptible to the disease condition, but has not yet been diagnosed as having the disease condition;
(ii) inhibiting the disease or disease state, i.e., arresting its development;
(iii) alleviating the disease or condition, i.e., causing regression of the disease or 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 words "comprise" or "comprise" and variations thereof such as "comprises" or "comprising," are to be understood in an open, non-exclusive sense, i.e., "including but not limited to.
The intermediates and compounds of the present application may also exist in different tautomeric forms, and all such forms are included within the scope of the present application. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies that can interconvert via a low energy barrier. For example, proton tautomers (also referred to as proton transfer tautomers) include interconversion via proton migration, such as keto-enol and imine-enamine isomerizations. A specific example of a proton tautomer is an imidazole moiety, wherein the proton can migrate between two ring nitrogens. Valence tautomers include interconversion by recombination of some of the bonding electrons.
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. Examples of isotopes that can be incorporated into compounds of the present application 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 where deuterium substitution may be partial or complete, partial deuterium substitution meaning that at least one hydrogen is substituted with at least one deuterium.
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.
In all methods of administration of the compounds of formula (I), formula (II), formula (III), formula (IV), formula (V), formula (VI), formula (VII), formula (VIII) or formula (IX) described herein, the compounds are administered in a daily dose of 0.01 to 200mg/kg body weight, in single or divided doses.
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 (such as amino Groups in the present application), for example, reference may be made to Greene's Protective Groups in Organic Synthesis (4th Ed.) Hoboken, New Jersey: John Wiley & Sons, Inc. all references cited herein are incorporated herein in their entirety.
In some embodiments, the compounds of formula (i) herein may be prepared by those skilled in the art of organic synthesis via scheme 1 using standard methods in the art:
route 1:
Figure BDA0001216697810000171
wherein the content of the first and second substances,
R1、R3、R4、R5、R6、R7x and Cy are as defined for compounds of formula (I).
In some embodiments, the compounds of formula (i) herein may be prepared by those skilled in the art of organic synthesis via scheme 2 using standard methods in the art:
route 2:
Figure BDA0001216697810000181
wherein the content of the first and second substances,
R1、R3、R4、R5、R6、R7x and Cy are as defined for compounds of formula (I).
In some embodiments, the compounds of formula (II), formula (IV), formula (V), formula (VII), or formula (VIII) may be prepared by methods described above with reference to schemes 1-2.
In some embodiments, the compounds of formula (iii) herein may be prepared by one skilled in the art of organic synthesis via scheme 3 using standard methods in the art:
route 3:
Figure BDA0001216697810000182
wherein the content of the first and second substances,
R1、R2、R3、R4、R5、R6、R7x and n are as defined for compounds of formula (III).
In some embodiments, the compounds of formula (iv) herein may be prepared by those skilled in the art of organic synthesis via scheme 4 using standard methods in the art:
route 4:
Figure BDA0001216697810000183
wherein the content of the first and second substances,
R1、R3、R5、R6、R7x and Cy are as defined for compounds of formula (IV).
As shown in scheme 4, a compound of formula 12a is reacted with acryloyl chloride to provide a compound of formula 13a, and the compound of formula 13a is reduced to the nitro group to provide a compound of formula 14a (the compound of formula 14a is also commercially available or synthesized with reference to TW 201512176A). Reacting a compound of formula 3 (commercially available as such) with a compound of formula 4a (commercially available or as referred to WO2011022337A1, WO2012109164A1) under basic conditions to obtain a compound of formula 5 a. Reacting the compound shown in the formula 14a with the compound shown in the formula 5a under an acidic condition to obtain the compound shown in the formula (IV).
In some embodiments, the compounds of formula (iv) herein may be prepared by those skilled in the art of organic synthesis via scheme 5 using standard methods in the art:
route 5:
Figure BDA0001216697810000191
wherein the content of the first and second substances,
R1、R3、R5、R7x and Cy are as defined for formula (IV)A compound (I) is provided.
As shown in scheme 5a, a compound of formula 15a (which can be synthesized with reference to TW 201512176A) and a compound of formula 4a are reacted under heating to obtain a compound of formula (iv).
In some embodiments, compounds of formula (vi) herein can be prepared by those skilled in the art of organic synthesis via scheme 6 using standard methods in the art:
route 6:
Figure BDA0001216697810000192
wherein the content of the first and second substances,
R1、R2、R3、R5、R7x and n are as defined for compounds of formula (VI).
As shown in scheme 6, compounds of formula 3a and compounds of formula 9a (which are directly commercially available) are reacted under basic conditions to provide compounds of formula 10. The compound of formula 10a and the compound of formula 14a are reacted under acidic conditions to provide the compound of formula 16 a. Removing the pinacol protection from the compound of the formula 16a to obtain the compound of the formula (VI).
In some embodiments, the compounds of formula (vii) herein may be prepared by those skilled in the art of organic synthesis via scheme 7 using standard methods in the art:
route 7:
Figure BDA0001216697810000201
wherein the content of the first and second substances,
R1、R3、R4、R6、R7、R12、X、Y1、Y2q, and Cy are as defined for the compound of formula (VII).
In some embodiments, the compounds of formula (IX) herein may be prepared by those skilled in the art of organic synthesis via scheme 8 using standard procedures in the art:
route 8:
Figure BDA0001216697810000202
wherein the content of the first and second substances,
R1、R2、R3、R4、R6、R7、R12、X、Y1、Y2q and n are as defined for compounds of formula (IX);
the following abbreviations are used in this application:
the invention employs the following abbreviations: eq represents equivalent, equivalent; TLC for thin layer chromatography; EtOH stands for ethanol; MeOH represents methanol; HCl represents hydrogen chloride; DCM represents dichloromethane; n-BuOH represents n-butanol; 2-BuOH represents sec-butyl alcohol; PTSA represents p-toluenesulfonic acid; DIAD represents diisopropyl azodicarboxylate; DMF represents N, N-dimethylformamide; DMSO represents dimethyl sulfoxide; BOC represents tert-butylcarbonyl as an amine protecting group; O/N stands for overnight; THF represents tetrahydrofuran; boc2O represents di-tert-butyl dicarbonate; TFA represents trifluoroacetic acid; DIPEA stands for diisopropylethylamine.
Detailed Description
The present invention is described in detail below by way of examples, but is not meant to be limited to any of the disadvantages of the present invention. Having described the present application in detail and disclosed specific embodiments thereof, it will be apparent to those skilled in the art that various changes and modifications can be made therein without departing from the spirit and scope thereof. All solvents used herein are commercially available and can be used without further purification. The starting compounds for the syntheses described herein are commercially available or can be prepared by methods known in the art.
Example 1 preparation of N- (3- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound I-1)
Figure BDA0001216697810000211
Step (1) preparation of 6- ((2, 5-dichloropyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound 3)
To a reaction flask were added compound 1(4.92g, 26.85mmol), compound 2(4.0g, 26.85mmol), DIPEA (6.94g, 53.70mmol) and n-butanol (40mL), and the mixture was refluxed under nitrogen for 1 hour. After the reaction was completed, the reaction solution was concentrated to dryness under reduced pressure, 50mL of water was added to the concentrate, and ultrasonic treatment was carried out to precipitate a solid, which was washed with suction filtration and water, and finally dried under reduced pressure at 50 ℃ to obtain Compound 3(7.94 g).
1H-NMR(500M,DMSO-d6)δ9.63(s,1H),9.28(s,1H),8.36~8.37(d,J=0.5Hz,1H),7.79(s,1H),7.58~7.60(dd,J=2Hz,3.4Hz,1H),7.43~7.45(d,J=8Hz,1H),5.01(s,2H).
HRMS(ESI,[M+H]+)m/z:295.9991.
Step (2) preparation of N- (3- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound I-1)
To a reaction flask was added compound 3(0.30g, 1.01mmol), compound 4(0.24g, 1.11mmol), trifluoroacetic acid (0.23g, 2.02mmol), sec-butanol (10 mL); the microwave is heated to 140 ℃ and the power is 150W, and the reaction is carried out for 1 hour. After completion of the reaction, saturated aqueous sodium bicarbonate was added to adjust the pH to basic, dichloromethane (20mL, three times) was extracted, and the organic phases were combined and dried over anhydrous magnesium sulfate. The drying agent was removed by suction filtration and the filtrate was concentrated under reduced pressure to remove the solvent. Column chromatography (100% dichloromethane to 5% methanol/dichloromethane gradient elution) followed by concentration under reduced pressure and drying afforded compound I-1(0.16 g).
1H-NMR(500M,DMSO-d6)δ9.93(s,1H),9.18(s,1H),8.91(s,1H),8.13(s,1H),8.09(s,1H),7.86(s,1H),7.76~7.80(t,J=10.5Hz,1H),7.40~7.42(t,J=7.0Hz,1H),7.28~7.30(d,J=8.5Hz,1H),6.97~6.98(d,J=8.5Hz,1H),6.39~6.45(dd,J=10.0Hz,17.0Hz,1H),6.20~6.23(d,J=17.0Hz,1H),5.71~5.73(d,J=10.5Hz,1H),4.95(s,2H),4.10(s,3H).
HRMS(ESI,[M+H]+)m/z:452.1148.
Example 2 preparation of N- (3- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) phenyl) acrylamide (Compound I-2)
Figure BDA0001216697810000221
Compound I-2 was prepared by the method of step (2) of reference example 1, starting from compounds 3 and 5.
1H-NMR(500M,DMSO-d6)δ10.01(s,1H),9.28(s,1H),9.18(s,1H),8.95(s,1H),8.14(s,1H),7.74(s,1H),7.72~7.74(m,2H),7.43~7.45(d,J=8.0Hz,1H),7.37~7.39(d,J=8.0Hz,1H),7.23~7.24(d,J=7.5Hz,1H),7.02~7.06(t,J=8.0Hz,1H),6.42~6.48(m,1H),6.22~6.26(m,1H),5.72~5.75(dd,J=1.5Hz,10.0Hz,1H),5.01(s,2H).
HRMS(ESI,[M+H]+)m/z:422.1066.
Example 3 preparation of N- (3- ((4- ((1-hydroxy-1, 3-dihydrobenzo [ C ] [1,2] oxaborzol-6-yl) amino) -5-methylpyrimidin-2-yl) amino) phenyl) acrylamide (Compound I-3)
Figure BDA0001216697810000222
Step (1) preparation of 6- ((2-chloro-5-methylpyridin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound 3-5)
Compounds 3 to 5 were prepared by the method of step (1) of reference example 1 using compounds 1 to 5 and compound 2 as starting materials.
1H-NMR(500M,DMSO-d6)δ9.35(s,1H),9.24(s,1H),7.93~7.95(m,2H),7.70~7.72(dd,J=2.0Hz,8.5Hz,1H),7.38~7.40(d,J=8.0Hz,1H),4.98(s,2H),3.95(s,3H).
HRMS(ESI,[M+H]+)m/z:292.0652.
Step (2) preparation of N- (3- ((4- ((1-hydroxy-1, 3-dihydrobenzo [ C ] [1,2] oxazol-6-yl) amino) -5-methylpyrimidin-2-yl) amino) phenyl) acrylamide (Compound I-3)
Compound I-3 was prepared by the method of step (2) of reference example 1, starting from compounds 3-5 and 5.
MS(ESI,[M+H]+)m/z:417.2.
Example 4 preparation of (3- ((2- ((5-Acrylamido-4- (2- (dimethylamino) ethoxy) -2-methoxyphenyl) amino) -5-chloropyrimidin-4-yl) amino) phenyl) boronic acid (Compound I-4)
Preparation of Compound 5-1:
Figure BDA0001216697810000231
step (1) preparation of 2-fluoro-4-methoxy-5-nitroaniline (Compound 8-1)
To reaction flask 1 were added compound 8(20.0g, 141.70mmol), methyl tert-butyl ether (160mL) and tetrahydrofuran (40mL), and the mixture was stirred at 0 ℃ for 30 minutes; concentrated nitric acid (65%, 15.11g, 155.87mmol) was slowly added dropwise to the reaction mixture, reacted at 0 ℃ for 1 hour, washed with methyl tert-butyl ether by suction filtration to give the nitrate of compound 8, and dichloromethane (100mL) was added to the nitrate to give a dichloromethane suspension of the nitrate. Concentrated sulfuric acid (98%, 139.20g, 142mmol) was added to reaction flask 2, and the resulting dichloromethane suspension of nitrate salt of compound 8 was added dropwise to reaction flask 2 at 0 ℃. After reacting for 3 hours, completely reacting, and adding ammonia water to adjust the pH value to be alkaline; the organic phase was washed with brine, and dried over anhydrous magnesium sulfate. The drying agent was removed by suction filtration, the solvent was removed by concentration under reduced pressure, and finally dried in an oven (45 ℃ C.) under reduced pressure to give compound 8-1(22.768 g).
1H-NMR(500M,DMSO-d6)δ7.37~7.39(d,J=9.0Hz,1H),7.18~7.20(d,J=12.5Hz,1H),5.24(s,2H),3.82(s,3H).
HRMS(ESI,[M+H]+)m/z:187.0511.
Step (2) preparation of 2- (2- (dimethylamino) ethoxy) -4-methoxy-5-nitroaniline (Compound 8-2)
N, N-dimethylethanolamine (2.87g, 32.2mmol) was placed in a 250mL three-necked flask and dissolved by adding 40mL of DMF. Sodium hydride (59%, 1.31g, 32.2mmol) was added slowly with stirring at-5 ℃ for additional 50 minutes, and a solution of compound 8-1(3g, 16.12mmol) in DMF (9mL) was added dropwise over 30 minutes. After stirring was continued for 30 minutes, the temperature was returned to room temperature and the mixture was stirred overnight. The reaction was quenched by addition of 15mL of water, concentrated under reduced pressure to remove the solvent by evaporation, and purified by column chromatography to give 8-2(4g) as a brown oily compound.
1H NMR(500MHz,DMSO-d6)δ7.30(s,1H),6.82(s,1H),4.81(s,2H),4.23(t,J=5.8Hz,2H),3.88(s,3H),2.72(t,J=5.8Hz,2H),2.27(s,6H).
HRMS(ESI,[M+H]+)m/z:256.1384.
Step (3) preparation of N- (2- (2- (dimethylamino) ethoxy) -4-methoxy-5-nitrophenyl) acrylamide (Compound 8-3)
Compound 8-2(3.56g, 13.95mmol) was placed in a 100mL three-necked flask, 70mL of tetrahydrofuran and triethylamine (5.8mL,41.8mmol) were added, and the mixture was placed in a low-temperature reaction tank at-10 ℃. A solution of acryloyl chloride (1.28g, 16.74mmol) in tetrahydrofuran (20mL) was added dropwise over 30 minutes. The reaction was continued at-10 ℃ for 1 hour. After returning to room temperature, the reaction was completed after 3 hours. The reaction was quenched by addition of 20mL of saturated sodium carbonate solution and the reaction solvent tetrahydrofuran was mostly evaporated to dryness. 100mL of saturated sodium carbonate solution was added, sonicated for 10 minutes, filtered, the filter cake was washed with water, and dried under reduced pressure to give compound 8-3(4.1 g).
1H NMR(500MHz,DMSO-d6)δ9.52(s,1H),8.70(s,1H),7.04(s,1H),6.62(dd,J=17.0,10.2Hz,1H),6.28(dd,J=17.0,2.0Hz,1H),5.79(dd,J=10.2,2.0Hz,1H),4.35(t,J=5.8Hz,2H),4.00(s,3H),2.73(t,J=5.8Hz,2H),2.27(s,6H).
HRMS(ESI,[M+H]+)m/z:310.1421.
Step (4) preparation of N- (5-amino-2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound 5-1)
Placing the compound 8-3(3.5g,11.32mmol) in a 100mL eggplant-shaped bottle, adding ethanol (20mL) and water (5mL), adding iron powder (3.79g, 67.9mmol) and ammonium chloride (424mg, 7.92mmol), heating to 80 ℃, reacting for 2 hours, removing most of the solvent by reduced pressure evaporation, extracting with dichloromethane (50mL, three times), washing with a small amount of water, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering, evaporating to dryness, and purifying by column chromatography to obtain the compound 5-1(1.5 g).
1H NMR(300MHz,DMSO-d6)δ9.79(s,1H),7.44(s,1H),6.67(s,1H),6.33(dd,J=16.9,9.9Hz,1H),6.18(dd,J=17.0,2.3Hz,1H),5.70(dd,J=9.8,2.3Hz,1H),4.45(brs,2H),4.01(t,J=5.4Hz,2H),3.73(s,3H),2.44(t,J=5.4Hz,2H),2.23(s,6H).
HRMS(ESI,[M+H]+)m/z:280.1646.
Preparation of Compound I-4:
Figure BDA0001216697810000241
step (1) preparation of 2, 5-dichloro-N- (3- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) pyrimidin-4-amine (compound 3-10)
Compounds 3 to 10 were prepared by the method of step (1) of reference example 1 using Compounds 1 and 2 to 1 as starting materials.
1H-NMR(500M,DMSO-d6)δ9.53(s,1H),8.37(s,1H),7.84(s,1H),7.79~7.81(d,J=8.0Hz,1H),7.49~7.50(d,J=7.5Hz,1H),7.41~7.44(t,J=7.5Hz,1H),1.31(s,12H).
HRMS(ESI,[M+H]+)m/z:366.0941.
Step (2) preparation of N- (5- ((5-chloro-4- ((3- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-26)
Compound I-26 was prepared by substituting trifluoroacetic acid for p-toluenesulfonic acid using compound 3-10 and compound 5-1 as starting materials in the same manner as in step (2) of example 1.
Step (3) preparation of (3- ((2- ((5-acrylamido-4- (2- (dimethylamino) ethoxy) -2-methoxyphenyl) amino) -5-chloropyrimidin-4-yl) amino) phenyl) boronic acid (Compound I-4)
Compound I-26(200mg, 0.328mmol) was placed in a 100mL single-neck flask, acetone (2mL) and water (2mL) were added, sodium periodate (141mg, 0.657mmol) and ammonium acetate (51mg, 0.657mmol) were added, and the reaction was completed at room temperature for 2 hours. Extracting with ethyl acetate (10mL for three times), washing with water, drying with anhydrous sodium sulfate, filtering, evaporating to dryness, and purifying by column chromatography to obtain compound I-4(50 mg).
1H NMR(500MHz,DMSO-d6)δ9.61(s,1H),8.59(s,1H),8.23(s,1H),8.04(s,1H),7.97(s,1H),7.91(s,1H),7.83~7.76(m,2H),7.45(d,J=7.2Hz,1H),7.21~7.13(m,1H),6.86(s,1H),6.46(dd,J=16.9,10.1Hz,1H),6.19(d,J=16.8Hz,1H),5.72(d,J=10.5Hz,1H),5.36-5.31(m,1H),4.19~4.13(m,2H),3.78(s,3H),2.64~2.57(m,2H),2.28(s,6H).
MS(ESI,[M+H]+)m/z:527.0.
Example 5 preparation of (4- ((2- ((5-Acrylamido-4- (2- (dimethylamino) ethoxy) -2-methoxyphenyl) amino) -5-chloropyrimidin-4-yl) amino) phenyl) boronic acid (Compound I-5)
Figure BDA0001216697810000251
Step (1) preparation of 2, 5-dichloro-N- (4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) pyrimidin-4-amine (compound 3-11)
Compounds 3 to 11 were prepared by the method of step (1) of reference example 1 using Compounds 1 and 2 to 2 as starting materials.
1H-NMR(500M,DMSO-d6)δ9.58(s,1H),8.42(s,1H),7.68~7.70(d,J=8.0Hz,2H),7.63~7,65(d,J=8.0Hz,2H),1.31(s,12H).
HRMS(ESI,[M+H]+)m/z:366.0937.
Step (2) preparation of (4- ((2- ((5-Acrylamido-4- (2- (dimethylamino) ethoxy) -2-methoxyphenyl) amino) -5-chloropyrimidin-4-yl) amino) phenyl) boronic acid (Compound I-5)
Compound I-5 was prepared by the method of step (2) in the synthesis of compound I-26 in reference example 4, starting from compounds 3-11 and 5-1.
1H-NMR(500M,DMSO-d6)δ9.66(br,1H),8.57(s,1H),8.14~8.18(d,2H),8.06(s,1H),7.82(m,2H),7.63(m,4H),6.89~6.91(d,1H),6.47(m,1H),6.15~6.18(d,1H),5.67~5.69(d,1H),4.21(br,2H),3.77(s,3H),2.67(s,2H),2.32(s,6H).
MS(ESI,[M+H]+)m/z:527.3.
Example 6 preparation of N- (5- ((5-chloro-4- ((2- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) amino) pyrimidin-2-yl) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-6)
Figure BDA0001216697810000261
Step (1) preparation of 2, 5-dichloro-N- (2- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) pyrimidin-4-amine (compound 3-12)
Compounds 3 to 12 were prepared by the method of step (1) of reference example 1 using Compound 1 and Compounds 2 to 3 as starting materials.
1H-NMR(500M,DMSO-d6)δ9.68(s,1H),8.44(s,1H),8.12~8.13(d,J=8.5Hz,1H),7.72~7.73(d,J=7.0Hz,1H),7.57~7.61(t,J=7.5Hz,1H),7.20~7.23(t,J=7.5Hz,1H),1.29(s,12H).
HRMS(ESI,[M+H]+)m/z:366.0945.
Step (2) preparation of N- (5- ((5-chloro-4- ((2- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) amino) pyrimidin-2-yl) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-6)
Compound I-6 was prepared by the method of step (2) in the synthesis of compound I-26 in reference example 4, starting from compounds 3-12 and 5-1.
MS(ESI,[M+H]+)m/z:527.3.
Example 7 preparation of N- (5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-5-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-7)
Figure BDA0001216697810000262
Step (1) preparation of 5- ((2, 5-dichloropyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound 3-13)
Compounds 3 to 13 were prepared by the method of step (1) of reference example 1 using compounds 1 and 2 to 4 as starting materials.
1H-NMR(500M,MeOD)δ7.07~7.10(t,J=7.5Hz,1H),6.75(s,1H),6.69~6.71(d,J=7.0Hz,1H),6.64~6.65(d,J=7.5Hz,1H),4.51(s,2H).
MS(ESI,[M+H]+)m/z:295.9.
Step (2) preparation of N- (5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-5-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-7)
Compound I-7 was prepared by the method of step (2) in the synthesis of compound I-26 in reference example 4, starting from compounds 3-13 and 5-1.
1H-NMR(500M,DMSO-d6)δ9.63(s,1H),8.96(br,1H),8.65(s,1H),8.24(s,1H),8.14(s,1H),8.08(s,1H),7.81(s,1H),7.53(s,2H),6.90(s,1H),6.44~6.49(dd,1H),6.14~6.18(d,1H),5.68~5.70(d,J=10.5Hz,1H),4.78(s,2H),4.19~4.21(t,J=5.5Hz,2H),3.75(s,3H),2.62~2.64(t,J=5.5Hz,2H),2.29(s,6H).
MS(ESI,[M+H]+)m/z:538.9.
Example 7a preparation of 2- (2-Acrylamido-4- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborolan-5-yl) amino) pyrimidin-2-yl) amino) -5-methoxyphenoxy) -N, N-dimethylethane-1-ammonium methanesulfonate (Compound I-7-21)
Figure BDA0001216697810000271
Adding the compound I-7(341mg, 633mmol, 1eq) into methanol (50mL), adding accurately weighed methanesulfonic acid (60mg, 632mmol, 1eq), stirring for 20 minutes, clarifying the solution, concentrating the reaction solution to obtain a crude product 360mg, adding diethyl ether (40mL), pulping, filtering, washing the filter cake with diethyl ether (10mL), and drying to obtain the compound I-7-21(329mg) with the purity of 95.93% (UPLC).
1H NMR(500MHz,DMSO-d6)δ9.55(s,1H),9.28(s,1H),9.00(brs,1H),8.87(s,1H),8.46(brs,1H),8.12(s,1H),8.09(s,1H),7.79(s,1H),7.57(dd,J=8and 12Hz,2H),6.90(s,1H),6.61(dd,J=10and 16.5Hz,1H),6.19(dd,J=1.5and 15.5Hz,1H),5.72(d,J=10.5Hz,1H),4.80(s,1H),4.47(t,J=5,2H),3.79(s,3H),3.61(d,J=8.5Hz,2H),2.94(d,J=3.5Hz,6H),2.38(s,3H).
MS(ESI,[M+H]+):m/z 539.1.
Example 8 preparation of N-5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-7-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-8)
Figure BDA0001216697810000281
Step (1) preparation of 7- ((2, 5-dichloropyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound 3-14)
Compounds 3 to 14 were prepared by the method of step (1) of reference example 1 using compounds 1 and 2 to 5 as starting materials.
1H NMR(500MHz,DMSO-d6)δ9.52(s,1H),8.95(s,1H),8.46(s,1H),8.03(d,J=8.0Hz,1H),7.56(t,J=7.8Hz,1H),7.21(d,J=7.8Hz,1H),5.04(s,2H).
MS(ESI,[M+H]+)m/z:295.9.
Step (2) preparation of N-5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-7-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-8)
Compound I-8 was prepared by the method of step (2) in the synthesis of compound I-26 in reference example 4, starting from compounds 3-14 and 5-1.
1H NMR(500MHz,DMSO-d6)δ9.63(d,J=17.1Hz,2H),8.42-8.32(m,2H),8.28-8.18(m,2H),8.13(s,1H),7.30-7.22(m,1H),7.00(d,J=7.5Hz,1H),6.92(s,1H),6.47(dd,J=17.0,10.2Hz,1H),6.17(d,J=16.9Hz,1H),5.70(d,J=10.3Hz,1H),4.99(s,2H),4.21(t,J=5.7Hz,2H),3.77(s,3H),2.63(t,J=5.7Hz,2H),2.29(s,6H).
HRMS(ESI,[M+H]+)m/z:539.1932.
Example 8 preparation of 8a N-5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-7-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide mesylate (Compound I-8-21)
Figure BDA0001216697810000282
Compound I-8(215mg, 0.38mmol) was placed in a 50mL single-necked eggplant-type bottle, and 3mL of methanol was added. Methanesulfonic acid (36mg, 0.38mmol) in 1mL methanol (1mL) was added dropwise and stirred at room temperature for 30 min. The reaction solution was heated to 50 ℃ and stirred for 1 hour. Evaporating the reaction solution to dryness, adding 3mL of acetone and 5 drops of methanol, pulping, performing ultrasonic treatment for 1 minute, filtering, washing a filter cake with a small amount of acetone, and drying to obtain a compound I-8-21(180 mg).
1H NMR(500MHz,DMSO-d6)δ10.19(s,1H),9.69~9.50(m,2H),9.02(brs,1H),8.78(brs,1H),8.20(s,1H),8.15~8.04(m,2H),7.39~7.29(m,1H),7.08(d,J=7.6Hz,1H),6.91(s,1H),6.80(dd,J=16.9,10.2Hz,1H),6.19(d,J=17.0Hz,1H),5.71(d,J=10.3Hz,1H),5.00(s,2H),4.50-4.40(m,2H),3.82(s,3H),3.62-3.57(m,2H),2.90(s,6H),2.37(s,3H).
MS(ESI,[M+H]+)m/z:539.1.
Example 9 preparation of N-5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-4-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-9)
Figure BDA0001216697810000291
Step (1) preparation of 4- ((2, 5-dichloropyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound 3-15)
Compounds 3 to 15 were prepared by the method of step (1) of reference example 1 using compounds 1 and 2 to 6 as starting materials.
1H-NMR(500M,DMSO-d6)δ9.52(s,1H),9.03(s,1H),8.37(s,1H),7.69~7.70(m,1H),7.44~7.45(d,J=6.5Hz,2H),4.93(s,2H).
MS(ESI,[M+H]+)m/z:295.9.
Step (2) preparation of N-5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-4-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-9)
Compound I-9 was prepared by the method of step (2) in the synthesis of compound I-26 in reference example 4, starting from compounds 3-15 and 5-1.
1H-NMR(500M,DMSO-d6)δ9.57(s,1H),9.15(s,1H),8.49(s,1H),8.15(s,1H),8.04(s,1H),7.82(s,1H),7.51~7.55(m,2H),7.25~7.28(t,J=7.5Hz,1H),6.79(s,1H),6.41~6.46(dd,1H),6.18~6.21(d,1H),5.71~5.74(d,J=10.5Hz,1H),4.95(s,2H),4.12~4.14(t,J=5.5Hz,2H),3.73(s,3H),2.54~2.57(t,J=5.5Hz,3H),2.26(s,6H).
MS(ESI,[M+H]+)m/z:539.3.
Example 9a preparation of 2- (2-acrylamido-4- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-4-yl) amino) pyrimidin-2-yl) amino) -5-methoxyphenoxy) -N, N-dimethylethane-1-ammonium methanesulfonate (Compound I-9-21)
Figure BDA0001216697810000301
Compound I-9-21 was prepared by the method of step (3) in the synthesis of Compound I-8-21 in reference example 8, starting with Compound I-9.
1H-NMR(500M,DMSO-d6)δ9.57(br,1H),9.26(br,2H),8.62(br,1H),8.13(s,1H),8.01(s,1H),7.59~7.60(d,J=7.0Hz,1H),7.50~7.51(t,J=6.0Hz,2H),7.31~7.34(t,J=14.5Hz,1H),7.12~7.14(d,J=8.0Hz,1H),6.82(s,1H),6.61~6.67(dd,J=17.0,10.5Hz,1H),6.22~6.25(d,J=17.0Hz,1H),5.74~5.76(d,J=10.0Hz,1H),4.95(s,2H),4.40(br,2H),3.77(s,3H),3.58(br,2H),2.92(s,6H),2.39(s,3H).
MS(ESI,[M+H]+)m/z:539.3.
Example 10 preparation of N- (5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) oxy) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-10)
Figure BDA0001216697810000302
Step (1) preparation of 6- ((2, 5-dichloropyridin-4-yl) oxy) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound 3-16)
Compounds 3 to 16 were prepared by the method of step (1) of reference example 1 using compounds 1 and 2 to 7 as starting materials.
1H-NMR(500M,DMSO-d6)δ9.30(s,1H),8.83(s,1H),7.53~7.56(m,2H),7.40~7.42(d,J=8.5Hz,1H),5.05(s,2H).
MS(ESI,[M+H]+)m/z:296.9.
Step (2) preparation of N- (5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) oxy) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-10)
Compound I-10 was prepared by the method of step (2) in the synthesis of compound I-26 in reference example 4, starting from compounds 3-16 and 5-1.
1H-NMR(500M,DMSO-d6)δ9.53(s,1H),9.24(br,1H),8.34(s,2H),7.97(s,1H),7.46(s,1H),7.40~7.41(d,J=8.0Hz,1H),7.33~7.35(d,J=8.5Hz,1H),6.78(s,1H),6.42~6.48(dd,1H),6.18~6.21(d,1H),5.71~5.73(d,J=10.5Hz,1H),5.00(s,2H),4.11~4.13(t,J=5.0Hz,2H),3.73(s,3H),2.58(t,2H),2.26(s,6H).
MS(ESI,[M+H]+)m/z:540.3.
EXAMPLE 11 preparation of N- (5- ((5-chloro-4- ((5-fluoro-1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-11)
Figure BDA0001216697810000311
Step (1) preparation of 6- ((2, 5-dichloropyridin-4-yl) amino) -5-fluorobenzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound 3-17)
Compounds 3 to 17 were prepared by the method of step (1) of reference example 1 using compounds 1 and 2 to 8 as starting materials.
1H-NMR(500M,DMSO-d6)δ9.66(s,1H),9.34(br,1H),8.39(s,1H),7.71~7.72(d,J=8.0Hz,1H),7.40~7.42(d,J=10.0Hz,1H),5.02(s,2H).
MS(ESI,[M+H]+)m/z:313.9.
Step (2) preparation of N- (5- ((5-chloro-4- ((5-fluoro-1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-11)
Compound I-11 was prepared by the method of step (2) in the synthesis of compound I-26 in reference example 4, starting from compounds 3-17 and 5-1.
1H-NMR(500M,DMSO-d6)δ9.49(s,1H),9.23(br,1H),8.75(s,1H),8.07~8.08(d,J=8.0Hz,1H),8.05(s,1H),7.73~7.75(d,J=7.5Hz,1H),7.61(s,1H),7.22~7.24(d,J=10.0Hz,1H),6.78(s,1H),6.46~6.50(m,1H),6.17~6.21(dd,1H),5.70~5.72(d,J=10.5Hz,1H),4.98(s,2H),4.13~4.14(t,2H),3.78(s,3H),2.69(t,2H),2.33(s,6H).
MS(ESI,[M+H]+)m/z:557.1.
EXAMPLE 12 preparation of N- (5- ((5-chloro-4- ((1-hydroxy-3, 4-dihydro-1H-benzo [ c ] [1,2] oxaborolin-7-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) propenylamide (Compound I-12)
Figure BDA0001216697810000321
Step (1) preparation of 7- ((2, 5-dichloropyrimidin-4-yl) amino) -3, 4-dihydro-1H-benzo [ c ] [1,2] oxaborolnin-1-ol (Compound 3-18)
Compounds 3 to 18 were prepared by the method of step (1) of reference example 1 using compounds 1 and 2 to 9 as starting materials.
1H-NMR(500M,DMSO-d6)δ9.53(s,1H),846(br,1H),8.35(s,1H),7.76(s,1H),7.55~7.56(d,J=8Hz,1H),7.23~7.25(d,J=8.5Hz,1H),4.09~4.11(t,J=5.5Hz,2H),2.86~2.89(t,J=5.5Hz,2H).
MS(ESI,[M+H]+)m/z:310.0
Step (2) preparation of N- (5- ((5-chloro-4- ((1-hydroxy-3, 4-dihydro-1H-benzo [ c ] [1,2] oxaborozin-7-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) propenylamide (Compound I-12)
Compound I-12 was prepared by the method of step (2) in the synthesis of compound I-26 in reference example 4, starting from compounds 3-18 and 5-1.
1H-NMR(500M,DMSO-d6)δ9.68(s,1H),8.64(s,1H),8.37(s,1H),8.24(s,1H),8.04(s,1H),7.87(s,1H),7.78~7.80(d,J=8.0Hz,1H),7.70(s,1H),7.04~7.05(d,J=8.0Hz,1H),6.85(s,1H),6.58(m,1H),6.17~6.20(dd,1H),5.70~5.72(d,J=10Hz,1H),4.02(br,2H),4.04~4.07(t,J=5.5Hz,2H),3.79(s,3H),2.79~2.81(t,J=5.5Hz,4H),2.38(s,6H).
MS(ESI,[M+H]+)m/z:551.4.
Example 13 preparation of N- (3- ((4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) -5- (trifluoromethyl) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound I-13)
Figure BDA0001216697810000322
Adding compounds 3-19(260mg, 0.698mmol), compound 2(131mg, 0.767mmol) and ethanol as solvent into a reaction flask; heated to reflux for 4 hours under nitrogen. The reaction solution was evaporated to dryness by rotary evaporation and purified by column chromatography to give compound I-13(390mg, 83%).
1H NMR(500MHz,DMSO-d6)δ9.90(s,1H),9.14(s,1H),8.65(s,1H),8.39-8.25(m,2H),7.88(brs,1H),7.70~7.57(m,2H),7.40(dd,J=8.8,2.2Hz,1H),7.24(d,J=7.4Hz,1H),6.97(d,J=8.9Hz,1H),6.40(dd,J=16.9,10.1Hz,1H),6.21(dd,J=17.0,2.1Hz,1H),5.72(dd,J=10.1,2.1Hz,1H),4.93(s,2H),3.75(s,3H).
HRMS(ESI,[M+H]+)m/z:486.1252.
Example 14 preparation of N- (3- ((4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) -5- (trifluoromethyl) pyrimidin-2-yl) amino) phenyl) acrylamide (Compound I-14)
Figure BDA0001216697810000331
Starting from compounds 3 to 20 and 2, compound I-14 was prepared by the method of step (1) of reference example 13.
1H NMR(500MHz,DMSO-d6)δ10.21~10.01(m,2H),9.33(brs,1H),8.44(s,1H),7.72(s,1H),7.64(brs,1H),7.54(d,J=7.9Hz,1H),7.42(d,J=7.9Hz,1H),7.32~7.21(m,2H),6.93(brs,1H),6.45(dd,J=17.0,10.1Hz,1H),6.23(dd,J=17.0,2.1Hz,1H),5.74(dd,J=10.1,2.1Hz,1H),5.04(s,2H).
HRMS(ESI,[M+H]+)m/z:456.1514.
Example 15 preparation of N- (5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-15)
Figure BDA0001216697810000332
Compound I-15 was prepared by the method of step (2) in the synthesis of compound I-26 in reference example 4, starting from compounds 3 and 5-1.
1H NMR(500MHz,DMSO-d6)δ9.61(s,1H),9.16(s,1H),8.78(s,1H),8.21(s,1H),8.05(s,1H),7.85(s,1H),7.83~7.70(m,2H),7.22(d,J=8.1Hz,1H),6.85(s,1H),6.43(dd,J=17.0,10.2Hz,1H),6.16(d,J=17.0Hz,1H),5.70(d,J=10.2Hz,1H),4.93(s,2H),4.14(t,J=5.7Hz,2H),3.79(s,3H),2.58(t,J=5.7Hz,2H),2.27(s,6H).
HRMS(ESI,[M+H]+)m/z:539.2084.
Example 15 preparation of 15a N- (5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide mesylate (Compound I-15-21)
Figure BDA0001216697810000341
Compound I-15-21 was prepared by the method of step (3) in the synthesis of Compound I-8-21 in reference example 8, starting with Compound I-15.
1H NMR(500MHz,DMSO-d6)δ9.34(s,1H),9.13(s,1H),8.77(s,1H),8.14(s,1H),8.06(s,1H),7.82-7.73(m,3H),7.25(d,J=8.1Hz,1H),6.85(s,1H),6.51(s,1H),6.21(dd,J=16.9,2.0Hz,1H),5.74(dd,J=10.0,2.1Hz,1H),4.94(s,2H),4.37(t,J=5.1Hz,2H),3.83(s,3H),3.49~3.31(m,2H),2.84(s,6H),2.36(s,3H).
MS(ESI,[M+H]+)m/z:539.1.
Example preparation of 166- ((5-chloro-2- ((2-methoxyphenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-16)
Figure BDA0001216697810000342
Compound I-16 was prepared by the method of step (2) of reference example 1 using compound 3 and compound 6-4 as starting materials.
1H NMR(500MHz,DMSO-d6)δ9.21(s,1H),9.03(s,1H),8.13(s,1H),7.95(d,J=7.8Hz,1H),7.88(s,1H),7.71(s,1H),7.64(d,J=8.1Hz,1H),7.40(d,J=8.0Hz,1H),6.98(d,J=8.0Hz,1H),6.92(t,J=7.6Hz,1H),6.71(t,J=7.6Hz,1H),5.03(s,2H),3.82(s,3H).
HRMS(ESI,[M+H]+)m/z:383.1143.
Example 17 preparation of N- (5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl (meth) amino) -4-methoxyphenyl) acrylamide (Compound I-17)
Figure BDA0001216697810000351
Step (1) N1- (2- (dimethylamino) ethyl) -5-methoxy-N1Preparation of (E) -methyl-4-nitrophenyl-1, 2-diamine (Compound 8-4)
To a reaction flask was added Compound 8-1(11.00g, 59.14), N1,N1,N2Trimethyl-1, 2-diamine (9.06g, 88.71mmol), potassium carbonate (12.26g, 88.71mmol) and the solvent DMF (60 mL). Under the protection of nitrogen, the mixture was heated to 90 ℃. After completion of the reaction, water was added to the reaction, extracted with ethyl acetate (three times 100mL), the organic phases were combined and washed with brine, the organic phase was dried over anhydrous magnesium sulfate, then the drying agent was removed by suction filtration, the solvent was removed by concentration under reduced pressure, and finally dried under reduced pressure in an oven at 50 ℃ to obtain compound 8-4(14.858 g).
1H-NMR(500M,MeOD)δ7.33(s,1H),6.81(s,1H),3.88(s,3H),3.16~3.19(t,J=6.5Hz,2H),2.77(s,3H),2.60~2.62(t,J=6.5Hz,2H),2.33(s,6H).
MS(ESI,[M+H]+)m/z:269.1.
Step (2) preparation of N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5-nitrophenyl) acrylamide (Compound 8-5)
To a reaction flask, add compound 8-4(1.0g, 3.73mmol), tetrahydrofuran (10mL) and water (1mL) and stir at 0 ℃; 3-Chloropropylchloride (0.57g, 4.47mmol) was added dropwise to the reaction flask. After the completion of the dropwise addition, the reaction was warmed to room temperature, and after the completion of the reaction, solid sodium hydroxide (0.597g, 14.92mmol) was further added to the reaction mixture. After stirring, the mixture was concentrated under reduced pressure to remove tetrahydrofuran, a small amount of water was added, and the mixture was extracted with ethyl acetate (10mL, three times). The organic phases were combined, dried over anhydrous magnesium sulphate, freed from the drying agent by suction filtration, concentrated under reduced pressure to remove the solvent and finally dried in an oven at 50 ℃ under reduced pressure to give compound 8-5(1.09 g).
1H-NMR(500M,DMSO-d6)δ8.58(s,1H),6.89~6.894(s,1H),6.50~6.56(dd,1H),6.39~6.43(dd,1H),5.82~5.84(d,J=10.5Hz,1H),3.97(s,3H),3.20~3.22(t,J=5.5Hz,2H),2.81(s,3H),2.59~2.62(t,J=5.5Hz,2H),2.33(s,6H).
MS(ESI,[M+H]+)m/z:323.1.
Step (3) preparation of N- (5-amino-2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (Compound 5-2)
To a reaction flask were added compound 8-5(1.0g, 3.10mmol), iron powder (0.869g, 15.52mmol), ammonium chloride (0.830g, 15.52mmol), solvent ethanol (14mL) and water (4mL), and the reaction was heated to 85 ℃. After completion of the reaction, water (20mL) was added, followed by extraction with ethyl acetate (40mL, three times), and the organic phases were combined and washed with saturated brine. The resulting organic phase was dried over anhydrous magnesium sulfate, then the drying agent was removed by suction filtration, and the filtrate was concentrated under reduced pressure to remove the solvent. Finally, the mixture was dried in an oven at 50 ℃ under reduced pressure to obtain Compound 5-2(0.628g)
1H-NMR(500M,DMSO-d6)δ10.06(br,1H),7.66(s,1H),6.80(s,1H),6.28~6.33(dd,J=9.5Hz,16.5Hz,1H),6.18~6.22(dd,J=2.0Hz,17.0Hz,1H),5.69~5.72(dd,J=1.5Hz,10.0Hz,1H),4.62(br,2H),2.18~2.80(t,J=5.5Hz,2H),2.61(s,3H),2.17(s,6H).
HRMS(ESI,[M+H]+)m/z:293.1976.
Step (4) preparation of N- (5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl (meth) amino) -4-methoxyphenyl) acrylamide (Compound I-17)
Compound I-17 was prepared by the method of step (2) in the synthesis of compound I-26 in reference example 4, starting from compound 3 and compound 5-2.
1H NMR(500MHz,DMSO-d6)δ9.97(s,1H),9.14(s,1H),8.78(s,1H),8.48(s,1H),8.08(s,1H),7.86(s,1H),7.80(s,1H),7.77(d,J=9.6Hz,1H),7.23(d,J=8.2Hz,1H),6.95(s,1H),6.46~6.36(m,1H),6.18(d,J=16.8Hz,1H),5.72(d,J=11.4Hz,1H),4.93(s,2H),3.79(s,3H),2.91~2.84(m,2H),2.67(s,3H),2.41~2.31(m,2H),2.24(s,6H).
MS(ESI,[M+H]+)m/z:552.2.
Example 18 preparation of N- (2- (2- (dimethylamino) ethoxy) -5- ((5-fluoro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborzol-6-yl) amino) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound I-18)
Figure BDA0001216697810000361
Step (1) preparation of 6- ((2-chloro-5-fluoropyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound 3-22)
Compounds 3 to 22 were prepared by the method of step (1) of reference example 1 using compounds 1 to 8 and compound 2 as starting materials.
1H NMR(500MHz,DMSO-d6)δ10.04(s,1H),9.29(s,1H),8.31(s,1H),7.91(s,1H),7.71(d,J=8.1Hz,1H),7.42(d,J=8.1Hz,1H),4.99(s,2H).
HRMS(ESI,[M+H]+)m/z:280.0472.
Step (2) preparation of N- (2- (2- (dimethylamino) ethoxy) -5- ((5-fluoro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborzol-6-yl) amino) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound I-18)
Compound I-18 was prepared by the method of step (2) in the synthesis of compound I-26 in reference example 4, starting from compound 3-22 and compound 5-1.
1H NMR(500MHz,DMSO-d6)δ9.65(s,1H),9.32(s,1H),8.38(s,1H),8.03(d,J=3.6Hz,1H),7.91~7.86(m,2H),7.64(s,1H),7.49(d,J=7.9Hz,1H),7.25(d,J=8.2Hz,1H),7.12(d,J=7.9Hz,1H),6.86(s,1H),6.44(dd,J=17.0,10.2Hz,1H),6.17(d,J=16.9Hz,1H),5.71(d,J=11.3Hz,1H),4.93(s,2H),4.14(t,J=5.4Hz,2H),3.82(s,3H),2.57(t,J=5.4Hz,2H),2.26(s,6H).
MS(ESI,[M+H]+)m/z:523.1.
Example 19 preparation of N- (2- (2- (dimethylamino) ethoxy) -5- ((4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound I-19)
Figure BDA0001216697810000371
Step (1) preparation of 6- ((2-chloropyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound 3-23)
Compounds 3 to 23 were prepared by the method of step (1) of reference example 1 using compounds 1 to 9 and compound 2 as starting materials.
1H NMR(500MHz,DMSO-d6)δ10.10(s,1H),9.28(s,1H),8.14(d,J=5.7Hz,1H),7.90-7.79(m,2H),7.41(d,J=8.1Hz,1H),6.76(d,J=5.7Hz,1H),4.97(s,2H).
HRMS(ESI,[M+H]+)m/z:262.0543.
Step (2) preparation of N- (2- (2- (dimethylamino) ethoxy) -5- ((4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound I-19)
Compound I-19 was prepared by the method of step (2) in the synthesis of compound I-26 in reference example 4, starting with compound 3-23 and compound 5-1.
1H NMR(500MHz,DMSO-d6)δ9.69(s,1H),9.32(s,1H),8.46(s,1H),7.95(d,J=5.7Hz,1H),7.89(d,J=8.3Hz,1H),7.76(s,1H),7.62(s,1H),7.24(d,J=8.3Hz,1H),6.88(s,1H),6.44(dd,J=16.9,10.2Hz,1H),6.21-6.13(m,2H),5.71(d,J=10.6Hz,1H),4.92(s,2H),4.15(t,J=5.6Hz,2H),3.83(s,3H),2.58(t,J=5.6Hz,2H),2.26(s,6H).
MS(ESI,[M+H]+)m/z:505.1.
Example 19a preparation of 2- (2-acrylamido-4- ((4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) -5-methoxyphenoxy) -N, N-dimethylethane-1-ammonium methanesulfonate (Compound I-19-21)
Figure BDA0001216697810000381
To a reaction flask was added compound I-19(0.53g, 1.05mmol), methanol (3mL) and stirred at room temperature; methanesulfonic acid (100.98mg, 1.12mmol) in methanol (2mL) was slowly added to the reaction. After the reaction was gradually cleared, the reaction mixture was heated to 50 ℃ and reacted for 1 hour. Then concentrating under reduced pressure to remove methanol, adding methyl tert-butyl ether (5mL) and pulping; a solid was obtained by suction filtration, and finally dried under reduced pressure to obtain Compound I-19-21(0.546 g).
1H NMR(500MHz,DMSO-d6)δ9.41(s,1H),9.39(s,1H),9.16(s,1H),8.37(s,1H),7.95~7.96(d,J=6.0Hz,1H),7.88~7.90(d,J=8.0Hz,1H),7.77(s,1H),7.69(br,1H),7.25~7.27(d,J=8.5Hz,1H),6.89(s,1H),6.53~6.59(dd,J=17.0,10.5Hz,1H),6.21-6.24(m,2H),5.74~5.76(d,J=10.5Hz,1H),4.93(s,2H),4.38~4.40(t,J=4.0Hz,2H),3.88(s,3H),3.44(m,2H),2.84(s,6H),2.37(s,3H).
MS(ESI,[M+H]+)m/z:505.1.
EXAMPLE 20 preparation of N- (5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-1-6-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (Compound I-20)
Figure BDA0001216697810000382
Step (1) preparation of 6- ((2, 5-dichloropyridin-4-yl) amino) -3, 3-dimethylbenzo [ C ] [1,2] oxazol-1 (3H) -ol (Compound 3-24) (preparation of Compound 3-24)
Compounds 3 to 24 were prepared by the method of step (1) of reference example 1 using Compound 1 and Compounds 2 to 10 as starting materials.
1H NMR(500MHz,DMSO-d6)δ9.55(s,1H),9.08(s,1H),8.36(s,1H),7.72(d,1H),7.57(dd,1H),7.44(d,1H),1.47(s,6H).
HRMS(ESI,[M+H]+)m/z:324.0759.
Step (2) preparation of N- (5- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-1-6-yl) amino) pyrimidin-2-yl) amino) -2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (compound I-20) compound I-20 was prepared by the method of step (2) in the synthesis of compound I-26 in reference example 4, starting with compounds 3-24 and 5-1.
1H NMR(500MHz,DMSO-d6)δ9.64(s,1H),8.69(s,1H),8.25(s,1H),8.05(s,1H),7.92(s,1H),7.82~7.71(m,2H),7.23(d,J=8.2Hz,1H),6.87(s,1H),6.47(dd,J=17.0,10.2Hz,1H),6.20(d,J=16.9Hz,1H),5.72(d,J=10.2Hz,1H),4.15(t,J=5.7Hz,2H),3.79(s,3H),2.58(t,J=5.7Hz,2H),2.26(s,6H),1.43(s,6H).
MS(ESI,[M+H]+)m/z:567.1.
Example 21 preparation of N- (3- ((4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborzol-6-yl) amino) -5-methylpyrimidin-2-yl) amino) phenyl) acrylamide (Compound I-21)
Figure BDA0001216697810000391
Step (1) preparation of 6- ((2-chloro-5-methylpyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound 3-25)
Compounds 3 to 25 were prepared by the method of step (1) of reference example 1 using compounds 1 to 10 and compound 2 as starting materials.
1H NMR(500MHz,DMSO-d6)δ9.24(s,1H),8.94(s,1H),8.02(s,1H),7.84(s,1H),7.64(d,J=8.1Hz,1H),7.41(d,J=8.1Hz,1H),4.99(s,2H),2.17(s,3H).
HRMS(ESI,[M+H]+)m/z:276.0692.
Step (2) preparation of N- (3- ((4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) -5-methylpyrimidin-2-yl) amino) phenyl) acrylamide (Compound I-21)
Compound I-21 was prepared by the method of step (2) of reference example 1, starting from compound 3-25 and compound 5.
1H NMR(500MHz,DMSO-d6)δ9.97(s,1H),9.11(s,1H),8.87(s,1H),8.36(s,1H),7.92-7.87(m,2H),7.83(dd,J=8.2,1.9Hz,1H),7.76(s,1H),7.49(d,J=8.1Hz,1H),7.34(d,J=8.2Hz,1H),7.21(d,J=7.9Hz,1H),7.04(t,J=8.1Hz,1H),6.45(dd,J=17.0,10.2Hz,1H),6.22(dd,J=17.0,2.1Hz,1H),5.72(dd,J=10.1,2.1Hz,1H),4.98(s,2H),2.12(s,3H).
HRMS(ESI,[M+H]+)m/z:402.1720.
Example 22 preparation of N- (3- ((4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborzol-6-yl) amino) pyrimidin-2-yl) amino) phenyl) acrylamide (Compound I-22)
Figure BDA0001216697810000401
Compound I-22 was prepared by the method of step (2) of reference example 1, starting from compound 3-25 and compound 5.
1H NMR(500MHz,DMSO-d6)δ9.16(s,1H),8.34(s,1H),7.94(s,1H),7.86(d,J=8.8Hz,1H),7.82(s,1H),7.69(dd,J=8.2,2.1Hz,1H),7.36(d,J=8.2Hz,1H),7.18(s,1H),6.59(d,J=2.6Hz,1H),6.33(dd,J=8.8,2.6Hz,1H),5.00(s,2H),3.80(s,3H),3.12~3.00(m,4H),2.48~2.43(m,4H),2.23(s,3H),2.10(s,3H).
HRMS(ESI,[M+H]+)m/z:461.2462.
Example 23 preparation of N- (3- ((5-fluoro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) phenyl) acrylamide (Compound I-23)
Figure BDA0001216697810000402
Compound I-23 was prepared by the method of step (2) of reference example 1, starting from compound 3-22 and compound 5.
1H NMR(500MHz,DMSO-d6)δ10.01(s,1H),9.40(s,1H),9.15(s,1H),9.11(s,1H),8.10(d,J=3.6Hz,1H),7.95~7.90(m,2H),7.82(s,1H),7.46(d,J=8.0Hz,1H),7.35(d,J=8.1Hz,1H),7.25(d,J=7.8Hz,1H),7.12(t,J=8.0Hz,1H),6.46(dd,J=17.0,10.1Hz,1H),6.24(d,J=15.7Hz,1H),5.73(d,J=10.7Hz,1H),4.98(s,2H).
HRMS(ESI,[M+H]+)m/z:406.1700.
Example 24 preparation of N- (3- ((5-bromo-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) phenyl) acrylamide (Compound I-24)
Figure BDA0001216697810000411
Step (1) preparation of 6- ((5-bromo-2-chloro-pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (compound 3-26)
Compounds 3 to 26 were prepared by the method of step (1) of reference example 1 using compounds 1 to 11 and compound 2 as starting materials.
1H NMR(500MHz,DMSO-d6)δ9.41(s,1H),9.27(s,1H),8.45(s,1H),7.76(s,1H),7.55(d,J=8.1Hz,1H),7.43(d,J=8.1Hz,1H),5.01(s,2H).
HRMS(ESI,[M+H]+)m/z:339.9653.
Step (2) preparation of N- (3- ((5-bromo-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) phenyl) acrylamide (Compound I-24)
Compound I-24 was prepared by the method of step (2) of reference example 1, starting from compound 3-26 and compound 5.
1H NMR(500MHz,DMSO-d6)δ9.99(s,1H),9.27(s,1H),9.17(s,1H),8.69(s,1H),8.21(s,1H),7.80(s,1H),7.72~7.66(m,2H),7.44~7.35(m,2H),7.25~7.19(m,1H),7.01(t,J=8.1Hz,1H),6.44(dd,J=17.0,10.1Hz,1H),6.23(d,J=16.9Hz,1H),5.77~5.69(m,1H),5.01(s,2H).
HRMS(ESI,[M+H]+)m/z:466.0649.
EXAMPLE 25 preparation of N- (2- (2- (dimethylamino) ethoxy) -5- ((4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborolan-6-yl) amino) (trifluoromethyl) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound I-25)
Figure BDA0001216697810000412
Step (1) preparation of (2-chloro-5- (trifluoromethyl) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborole-1- (3H) -ol (Compound 3-27)
6-aminobenzo [ c ] [1,2] oxaborole-1 (3H) -ol (4.000g, 26.854mmol, 1eq) and 2, 4-dichloro-5- (trifluoromethyl) pyrimidine (5.826g, 26.854mmol, 1eq) were dissolved in isopropanol (100mL), N-Diisopropylethylamine (DIPEA) (6.941g, 53.707mmol, 2eq) was added dropwise under ice-water bath (0 ℃), reacted overnight at room temperature, the reaction solution was concentrated to dryness, and the residue was dissolved in 250mL of ethyl acetate, washed with water and saturated brine once, dried, filtered, concentrated under reduced pressure, and purified by preparative chromatography to give compound 3-27(2.2g) with a purity of 99% (UPLC).
1H NMR(500MHz,DMSO-d6)δ9.60(s,1H),9.23(brs,1H),8.57(s,1H),7.69(d,J=1.5Hz,1H),7.50(dd,J=2.0and8.5Hz,1H),7.45(d,J=8.0Hz,1H),5.02(s,2H).
MS(ESI):m/z 330.0[M+H]+,228.1[M-H]-.
Step (2) preparation of N- (2- (2- (dimethylamino) ethoxy) -5- ((4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborolan-6-yl) amino) (trifluoromethyl) pyrimidin-2-yl) amino) -4-methoxyphenyl) acrylamide (Compound I-25)
Adding the compounds 3-27(500mg, 1.518mmol, 1.0eq), N- (5-amino-2- (2- (dimethylamino) ethoxy) -4-methoxyphenyl) acrylamide (509mg, 1.821mmol, 1.2eq) to 2-butanol (20mL), adding p-toluenesulfonic acid (433mg, 2.276mmol, 1.5eq), stirring at room temperature for 1 minute, heating and refluxing for 1 hour, completing the reaction, cooling to room temperature, adding a methanol solution of ammonia (7mol/L) to adjust the pH to 8, concentrating to prepare sand, and performing column chromatography to obtain the compound I-25(664mg) with purity of 97% (UPLC).
1H NMR(500MHz,DMSO-d6)δ10.12(brs,1H),9.48(s,1H),9.06(s,1H),8.45(s,1H),8.25(s,1H),8.06(s,1H),7.61(m,1H),7.51(d,J=8.0Hz,1H),7.11(d,J=8.0Hz,1H),6.81(s,1H),6.70(m,1H),6.20(dd,J=1.5and17Hz,1H),5.68(d,J=10.5Hz,2H),4.95(s,2H),4.35(s,2H),3.80(s,3H),3.39(s,2H),2.78(d,J=3.0Hz,6H).
MS(ESI):m/z 573.0[M+H]+.
Example 25a preparation of (2-acrylamido-4- ((4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborolan-6-yl) amino) -5- (trifluoromethyl) amino) -5-methoxyphenoxy) -N, N-dimethylethane-1-ammonium methanesulfonate (Compound I-25-21)
Figure BDA0001216697810000421
Compound I-25(567mg, 0.991mmol, 1.0eq) was dissolved in methanol (25mL), methanesulfonic acid (95.2mg, 0.991mmol, 1.0eq) was added with stirring, the reaction solution was clarified after 10 minutes of stirring, and the reaction solution was concentrated to dryness to give compound I-25-21(640mg) with a purity of 97% (UPLC).
1H NMR(500MHz,DMSO-d6)δ9.95(brs,1H),9.60(brs,1H),9.41(s,1H),8.39(brs,1H),7.99(s,1H),7.62(m,1H),7.51(m,1H),7.29(s,1H),7.12(d,J=8.0Hz,1H),6.86(s,1H),6.73(m,1H),6.20(m,1H),5.71(m,1H),4.98(s,2H),4.42(s,2H),3.82(s,3H),3.59(d,J=4.5Hz,2H),2.90(d,J=4.5Hz,6H).2.40(s,3H).
MS(ESI):m/z 573.1[M+H]+.
Example preparation of 266- ((5-chloro-2- ((2-methoxy-4- (4-methylpiperazin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-26)
Figure BDA0001216697810000431
Compound I-26 was prepared by the method of step (2) of reference example 1, starting from compound 3 and compound 6.
1H-NMR(500M,DMSO-d6)δ9.21(s,1H),8.67(s,1H),8.06(s,1H),7.88(s,1H),7.62~7.66(m,3H),7.35~7.37(d,J=8.5Hz,1H),6.63~6.634(d,J=2.0Hz,1H),6.34~6.36(d,J=9.0Hz,1H),5.01(s,2H),3.79(s,3H),3.25(m,4H)2.99(m,4H),2.61(s,3H).
HRMS(ESI,[M+H]+)m/z:481.1911.
Example 276 preparation of- ((5-chloro-2- ((2-methoxy-4- (4- (4-methylpiperazin-1-yl) piperidin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-17)
Figure BDA0001216697810000432
Compound I-17 was prepared by the method of step (2) of reference example 1 using compound 3 and compound 6-2 as starting materials.
1H NMR(500MHz,DMSO-d6)δ9.20(s,1H),8.85(s,1H),8.04(s,1H),7.87(brs,1H),7.64(d,J=8.1Hz,1H),7.60(s,1H),7.56(d,J=8.6Hz,1H),7.34(d,J=8.1Hz,1H),6.56(d,J=2.5Hz,1H),6.33~6.26(m,1H),4.99(s,2H),3.77(s,3H),3.62(d,J=12.0Hz,2H),2.62~2.53(m,3H),2.49~2.20(m,8H),2.15(s,3H),1.85-1.77(m,2H),1.53~1.42(m,2H).
HRMS(ESI,[M+H]+)m/z:564.2754.
Example 286 preparation of- ((5-chloro-2- (phenylamino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-28)
Figure BDA0001216697810000441
Compound I-28 was prepared by the method of step (2) of reference example 1, starting from compound 3 and compound 6-3.
1H NMR(500MHz,DMSO-d6)δ10.40(s,1H),10.07(s,1H),8.35(s,1H),7.86(s,1H),7.58(d,J=8.1Hz,1H),7.51-7.37(m,3H),7.15(t,J=7.7Hz,2H),6.99(t,J=7.3Hz,1H),5.04(s,2H).
HRMS(ESI,[M+H]+)m/z:353.0968.
Example preparation of 296- ((5-chloro-2- ((4- (4-methylpiperazin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-29)
Figure BDA0001216697810000442
Compound I-29 was prepared by the method of step (2) of reference example 1, starting from compounds 3 and 6-1.
1H NMR(500MHz,DMSO-d6)δ9.21(s,1H),9.02(brs,1H),8.87(s,1H),8.07(s,1H),7.87(s,1H),7.64(d,J=7.8Hz,1H),7.45~7.35(m,3H),6.74(d,J=8.6Hz,2H),5.02(s,2H),3.10~2.99(m,4H),2.65~2.54(m,4H),2.32(s,3H).
HRMS(ESI,[M+H]+)m/z:451.1826.
Example preparation of 306- ((5-chloro-2- ((4- (4-dimethylamino) piperidin-1-yl) -2-methoxyphenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-30)
Figure BDA0001216697810000443
Compound I-30 was prepared by the method of step (2) of reference example 1 using compound 3 and compound 6-5 as starting materials.
1H NMR(500MHz,DMSO-d6)δ9.20(s,1H),8.85(s,1H),8.05(s,1H),7.88(s,1H),7.65(d,J=8.0Hz,1H),7.62~7.54(m,2H),7.35(d,J=8.1Hz,1H),6.58(s,1H),6.32(d,J=8.3Hz,1H),5.00(s,2H),3.78(s,3H),3.62(d,J=11.7Hz,2H),2.61(t,J=12.1Hz,2H),2.25~2.12(m,7H),1.88~1.79(m,2H),1.53~1.41(m,2H).
HRMS(ESI,[M+H]+)m/z:509.2259.
Example preparation of 316- ((5-chloro-2- ((2-isopropyl-5-methyl-4- (piperidin-4-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-31)
Figure BDA0001216697810000451
Compound I-31 was prepared by the method of step (2) of reference example 1 using compound 3 and compound 6-6 as starting materials.
1H NMR(500MHz,DMSO-d6)δ9.04(s,1H),8.10(s,1H),7.80(s,1H),7.63(s,1H),7.57(dd,J=8.1,2.0Hz,1H),7.53(s,1H),7.41(d,J=8.1Hz,1H),6.73(s,1H),4.99(s,2H),4.58~4.51(m,1H),3.14~3.07(m,2H),2.74~2.64(m,2H),2.05~1.94(m,1H),1.84(s,3H),1.83(s,3H),1.62~1.53(m,4H),1.27(s,3H),1.26(s,3H).
HRMS(ESI,[M+H]+)m/z:508.2320.
Example preparation of 327- ((5-chloro-2- ((2-isopropoxy-5-methyl-4- (piperidin-4-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-32)
Figure BDA0001216697810000452
Compound I-32 was prepared by the method of step (2) of reference example 1, starting from compound 3-14 and compound 6-6.
1H NMR(500MHz,DMSO-d6)δ8.43(s,1H),8.24-8.14(m,2H),7.95(s,1H),7.60(s,1H),7.32(t,J=7.7Hz,1H),7.07(d,J=7.4Hz,1H),6.83(s,1H),5.00(s,2H),4.58-4.48(m,1H),3.10-3.05(m,2H),2.80-2.72(m,1H),2.68-2.61(m,2H),2.20(s,3H),1.67-1.51(m,4H),1.25(s,3H),1.24(s,3H).
MS(ESI,[M+H]+)m/z:508.1.
Example 337 preparation of- ((5-chloro-2- ((2-methoxy-4- (4- (4-methylpiperazin-1-yl) piperidin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-33)
Figure BDA0001216697810000461
Compound I-33 was prepared by the method of step (2) in the synthesis of reference example 1, starting from compound 3-14 and compound 6-2.
1H NMR(500MHz,DMSO-d6)δ9.61(s,1H),8.34(s,1H),8.24(s,1H),8.18(s,1H),8.10(s,1H),7.38(d,J=8.5Hz,1H),7.28(t,J=7.5Hz,1H),7.03(d,J=7.5Hz,1H),6.64(s,1H),6.49(dd,J=1.5,7Hz,1H),5.01(s,2H),3.76(s,3H),3.73(m,2H),2.69(t,J=11.5Hz,2H),2.57~2.37(m,5H),2.34(s,3H),1.86(d,J=11.5Hz,2H),1.51~1.58(m,2H),1.23~1.28(m,4H).
MS(ESI,[M+H]+)m/s:564.1.
Example preparation of 347- ((5-chloro-2- ((4- (4- (dimethylamino) piperidin-1-yl) -2-methoxyphenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-34)
Figure BDA0001216697810000462
Compound I-34 was prepared by the method of step (2) of reference example 1, starting from compound 3-14 and compound 6-5.
1H NMR(500MHz,DMSO-d6)δ9.61(s,1H),8.34(s,1H),8.24(br,1H),8.18(s,1H),8.10(s,1H),7.37~7.38(d,J=8.5Hz,1H),7.27~7.30(t,J=7.5Hz,1H),7.03~7.04(d,J=7.0Hz,1H),6.64~6.65(d,J=2.0Hz,1H),6.49~6.51(dd,J=2.5Hz,9.0Hz,1H),5.00(s,2H),3.76(s,3H),3.72~3.75(t,2H),2.67~2.72(t,J=11.0Hz,2H),2.25(s,6H),1.86~1.88(m,2H),1.49~1.57(m,2H).
MS(ESI,[M+H]+)m/z:509.2.
Example 356 preparation of- ((2- ((2-methoxy-4- (4-methylpiperazin-1-yl) phenyl) amino) -5-methylpyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-35)
Figure BDA0001216697810000463
Compound I-35 was prepared by the method of step (2) of reference example 1, starting from compound 3-25 and compound 6.
1H NMR(500MHz,DMSO-d6)δ9.16(s,1H),8.34(s,1H),7.94(s,1H),7.86(d,J=8.8Hz,1H),7.82(s,1H),7.69(dd,J=8.2,2.1Hz,1H),7.36(d,J=8.2Hz,1H),7.18(s,1H),6.59(d,J=2.6Hz,1H),6.33(dd,J=8.8,2.6Hz,1H),5.00(s,2H),3.80(s,3H),3.12~3.00(m,4H),2.48~2.43(m,4H),2.23(s,3H),2.10(s,3H).
HRMS(ESI,[M+H]+)m/z:461.2462.
Example 366- ((2- ((2-methoxy-4- (4-methylpiperazin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-36) preparation
Figure BDA0001216697810000471
Compound I-36 was prepared by the method of step (2) of reference example 1, starting from compound 3-23 and compound 6.
1H NMR(500MHz,DMSO-d6)δ9.36(s,1H),9.18(s,1H),7.96-7.90(m,2H),7.88(d,J=8.7Hz,1H),7.75(d,J=8.0Hz,1H),7.46(s,1H),7.32(d,J=8.1Hz,1H),6.64(s,1H),6.47(d,J=8.5Hz,1H),6.18(d,J=5.4Hz,1H),4.97(s,2H),3.83(s,3H),3.17~3.12(m,4H),2.63~2.55(m,4H),2.32(s,3H).
HRMS(ESI,[M+H]+)m/z:447.2316.
Example 376 preparation of- ((5-fluoro-2- ((2-methoxy-4- (4-methylpiperazin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-37)
Figure BDA0001216697810000472
Compound I-37 was prepared by the method of step (2) of reference example 1, starting from compound 3-22 and compound 6.
1H NMR(500MHz,DMSO-d6)δ9.35(s,1H),9.20(s,1H),8.08~7.97(m,2H),7.81~7.69(m,2H),7.48(s,1H),7.34(d,J=8.0Hz,1H),6.61(s,1H),6.41(dd,J=8.9,2.8Hz,1H),4.99(s,2H),3.80(s,3H),3.15~3.02(m,4H),2.50~2.43(m,4H),2.25(s,3H).
HRMS(ESI,[M+H]+)m/z:465.2192.
Example preparation of 386- ((5-bromo-2- ((2-methoxy-4- (4-methylpiperazin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-38)
Figure BDA0001216697810000481
Compound I-38 was prepared by the method of step (2) of reference example 1, starting from compound 3-26 and compound 6.
1H NMR(500MHz,DMSO-d6)δ9.18(s,1H),8.60(s,1H),8.12(s,1H),7.83(s,1H),7.65-7.58(m,2H),7.55(d,J=8.7Hz,1H),7.35(d,J=8.2Hz,1H),6.57(d,J=2.5Hz,1H),6.28(d,J=8.5Hz,1H),5.00(s,2H),3.78(s,3H),3.09~3.04(m,4H),2.47~2.41(m,4H),2.23(s,3H).
HRMS(ESI,[M+H]+)m/z:525.1382.
Example 39 preparation of (3- ((5-chloro-2- ((2-isopropoxy-5-methyl-4- (piperidin-4-yl) phenyl) amino) pyrimidin-4-yl) amino) phenyl) boronic acid (Compound I-39)
Figure BDA0001216697810000482
Dissolve compound 3-10(1g, 2.73mmol) and compound 6-6(0.96g, 3.0mmol) in 2-butanol (10mL) with stirring, and finally add trifluoroacetic acid (0.47g, 4.10 mmol). Microwave reaction at 120 deg.c for 2 hr. And after the reaction is finished, decompressing and concentrating to remove the solvent, adding 50mL of ethyl acetate to dilute the reaction solution, adjusting the pH of a water layer to 7-8 by using a saturated sodium bicarbonate solution, standing for layering, extracting the water layer for three times by using 50mL of ethyl acetate, combining organic layers, decompressing and concentrating to remove the solvent, and obtaining a crude product of the compound I-39-1. And separating by column chromatography to obtain compound I-39-1(1.02 g).
Compound I-39-1(700mg, 1.21mmol) was added to a 100mL single-neck flask, dissolved in 50mL tetrahydrofuran and 10mL purified water, followed by addition of sodium periodate (259mg, 1.21mmol), and the reaction was stirred at room temperature with dropwise addition of 2mol/L aqueous hydrochloric acid (0.605mL, 1.21mmol) on an ice bath. After the reaction, the solution was concentrated under reduced pressure to give a brown oil. A mixed solution (50mL) of dichloromethane/methanol (dichloromethane to methanol volume ratio of 10:1) is added to dissolve, a saturated sodium bicarbonate solution is used for adjusting the pH value to 6-7, and an aqueous layer is extracted three times by using a mixed solution (10mL) of dichloromethane/methanol (dichloromethane to methanol volume ratio of 10:1) to combine organic layers. The solvent was removed by concentration under reduced pressure to give a brown oil, which was isolated by column chromatography to give Compound I-39(110 mg).
1H NMR(500MHz,DMSO-d6)δ8.108(s,1H),7.995(s,2H),7.893(s,1H),7.737(s,1H)7.644~7.613(m,2H),7.566(s,1H),7.346~7.316(m,1H),6.740(s,1H),4.534~4.486(m,1H),3.314~3.273(m,2H),3.031~2.872(m,3H),1.998(s,3H),1.935~1.1.831(m,2H),1.784~1.710(m,2H),1.287~1.266(d,6H).
13C NMR(125MHz,DMSO-d6)δ157.88,156.83,155.02,144.84,138.15,136.54,130.74,130.28,130.11,128.30,128.06,127.41,126.28,121.38,111.51,104.39,71.71,44.13,35.25,29.26,22.40,18.86.
MS(ESI,[M+H]+)m/z:496.
Example 40 preparation of (3- ((5-chloro-2- ((2-methoxy-4- (4- (4-methylpiperazin-1-yl) piperidin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) phenyl) boronic acid (Compound I-40)
Figure BDA0001216697810000491
Compound 3-10(960mg, 2.63mmol) and compound 6-1(880mg, 2.89mmol) were dissolved in 2-butanol (10mL) with stirring and finally trifluoroacetic acid (0.45g, 3.95mmol) was added. Microwave reaction at 110 deg.c for 2 hr. After the reaction, the solvent was removed by concentration under reduced pressure, the reaction mixture was diluted with 50mL of methylene chloride and 10mL of purified water, the pH of the aqueous layer was adjusted to 7 with a saturated sodium bicarbonate solution, the aqueous layer was extracted three times with methylene chloride (10mL) by standing for layer separation, and the organic layers were combined and concentrated under reduced pressure to remove the solvent, to give a crude product of the compound I-40-1. Separating by column chromatography to obtain compound I-40-1(900 mg).
To a 100mL single-neck flask was added compound I-40-1(1.2g, 1.89mmol), followed by addition of 4.8mL each of methanol and n-pentane, stirring to dissolve, and finally addition of isobutylboronic acid (0.77g, 7.55mmol) and 1mol/L aqueous hydrochloric acid (3.6mL, 3.6 mmol). Stir vigorously at room temperature for 5 hours. The mixture was allowed to stand for separation, the pentane layer was discarded, the methanol layer was extracted three times with 10mL of n-pentane, the solvent was removed by concentration under reduced pressure, the mixture was dissolved in a mixed solution (50mL) of methylene chloride/methanol (the volume ratio of methylene chloride to methanol was 10:1), and the pH was adjusted to neutrality with a saturated sodium bicarbonate solution. The aqueous layer was extracted with a mixed solution (10mL) of methylene chloride/methanol (volume ratio of methylene chloride to methanol: 10:1) three times and the organic layers were combined and separated by column chromatography to give compound I-40(200 mg).
1H NMR(500MHz,DMSO-d6)δ8.705(s,1H),8.035~8.012(m,3H),7.890(s,1H),7.659~7.643(m,1H),7.618~7.590(m,2H),7.577~7.542(m,1H),6.574~6.569(m,1H),6.343~6.322(m,1H),3.776(s,3H),3.659~3.633(m,2H),3.180(s,1H),2.636~2.591(m,4H),2.475~2.368(m,4H),2.234(s,3H),1.860~1.837(m,2H),1.550~1.485(m,2H),1.307~1.188(m,2H).
13C NMR(125MHz,DMSO-d6)δ158.67,156.53,154.99,150.91,148.25,138.23,130.20,129.74,127.80,125.59,122.91,120.90,107.76,103.65,100.90,61.35,56.02,55.23,49.38,49.07,48.66,45.75,28.20.
MS(ESI,[M+H]+)m/z:552.1.
EXAMPLE 41 preparation of (3- ((5-chloro-2- ((4- (4- (dimethylamino) piperidin-1-yl) -2-methoxyphenyl) amino) pyrimidin-4-yl) amino) phenyl) boronic acid (Compound I-41)
Figure BDA0001216697810000501
Dissolve compound 3-10(2g, 5.46mmol) and compound 6-5(1.5g, 6.02mmol) in 2-butanol (10mL) with stirring, and finally add trifluoroacetic acid (0.94g, 8.24 mmol). The reaction was heated with a microwave at 120 ℃ for 2 hours. After the reaction, the solvent was removed under reduced pressure, a mixed solution (50mL) of dichloromethane/methanol (dichloromethane to methanol volume ratio 10:1) was added, the pH of the aqueous layer was adjusted to neutrality with a saturated aqueous solution of sodium bicarbonate, the mixture was allowed to stand for separation, the aqueous layer was extracted three times with a mixed solution (50mL) of dichloromethane/methanol (dichloromethane to methanol volume ratio 10:1), the organic layers were combined, and the solvent was removed by concentration under reduced pressure to give a crude product of compound I-41-1. Separating by column chromatography to obtain compound I-41-1(2 g).
To a 100mL single-neck flask was added compound I-41-1(870mg, 1.5mmol), 3.8mL each of methanol and n-pentane was added and dissolved with stirring, and finally isobutylboronic acid (611mg, 6mmol) and 1mol/L aqueous hydrochloric acid (2.85mL, 0.285mmol) were added. Stir vigorously at room temperature for 12 hours. Standing for layering, removing a pentane layer, extracting a methanol layer twice with 10mL of n-pentane, adjusting the pH value to 6-7 with a saturated sodium bicarbonate aqueous solution, extracting twice with 50mL of a dichloromethane/methanol (the volume ratio of dichloromethane to methanol is 10:1) mixed solution, retaining an organic layer, concentrating under reduced pressure to remove a solvent to obtain a yellow oily substance, and performing column chromatography to obtain a compound I-41(120 mg).
1H NMR(500MHz,DMSO-d6)δ8.753(s,1H),8.052~8.8.042(m,3H),7.893(s,1H),7.655~7.615(m,3H),7.562~7.547(m,1H),7.297~7.266(m,1H),6.613~6.610(m,1H),6.364~6.348(m,1H),3.787(s,3H),3.768~3.743(m,2H),3.270~3.223(m,1H),2.721(s,6H),2.657~2.633(m,2H),2.125~2.102(m,2H),1.787~1.722(m,2H).
13C NMR(125MHz,DMSO-d6)δ158.89,156.54,155.00,150.88,147.51,138.19,130.25,129.86,127.83,125.66,122.85,121.43,108.04,103.71,101.31,62.80,56.09,48.67,26.03.
MS(ESI,[M+H]+)m/z:497.1.
Example 426 preparation of- ((5-chloro-2- ((4- (piperidin-4-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-42)
Figure BDA0001216697810000502
Compound 3(1g,3.3mmol), compound 6-7(1.2g,3.96mmol), p-toluenesulfonic acid (682mg,3.96mmol) were placed in a 250mL three-necked flask, 2-butanol (50mL) was added, nitrogen substitution was performed, then heating and refluxing were performed for 6 hours, after the reaction was completed, the solvent was removed by concentration under reduced pressure, 20mL of ethyl acetate, 20mL of dichloromethane, and 100mL of water were added, the pH of the system was adjusted to 10 with saturated aqueous sodium carbonate solution, and the organic phase was separated and collected. The aqueous phase was extracted twice with 20mL of dichloromethane. The organic phases were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to remove the solvent, giving the crude compound I-42-1, which was used in the next step without purification.
Suspending the compound I-42-1 in 20mL of methanol, adding 4mL of concentrated hydrochloric acid under magnetic stirring, heating and stirring at 50 ℃ in an oil bath for 2.5h, cooling the system to room temperature after the reaction is finished, separating out white solid, filtering to remove the solid, and concentrating the filtrate under reduced pressure to dryness. The residue was taken up in 20mL of water and adjusted to pH 8 with saturated aqueous sodium carbonate solution, whereupon a grey solid precipitated. The gray solid was slurried with ethanol (5mL) overnight and suction filtered to give compound I-42(100 mg).
1H NMR(500MHz,DMSO-d6)δ9.13(s,1H),8.89(s,1H),8.10(s,1H),7.84(s,1H),7.63(d,J=5.0Hz,1H),7.46(d,J=6.0Hz,2H),7.41(d,J=7.0Hz,1H),6.95(d,J=5.5Hz,2H),5.02(s,2H),3.02(d,J=9.0Hz,2H),2.57(d,J=11.5Hz,2H),2.44(s,1H),1.63~1.06(m,6H).
MS(ESI,[M+H]+)m/z:436.1.
Example 436- ((5-chloro-2- ((4-morpholinylphenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-43) preparation
Figure BDA0001216697810000511
Placing compound 3(0.5g,1.69mmol), compound 6-8(361mg,2.02mmol) and trifluoroacetic acid (0.38mL,5.07mmol) in a 100mL three-necked flask, adding 10mL of 2-butanol, replacing with nitrogen, heating in oil bath under reflux for 5h, after the reaction is finished, evaporating the solvent under reduced pressure. The residue was taken up in 30mL of dichloromethane and 20mL of water and the pH was adjusted to 8 with saturated aqueous sodium carbonate solution. The layers were separated and the aqueous phase was extracted twice more with 20mL of dichloromethane. The water phase is emulsion, and is filtered to obtain off-white powdery solid. The solid was dried under vacuum at 50 ℃ for 4 hours to give Compound I-43(400 mg).
1H NMR(500MHz,DMSO-d6)δ8.99(s,1H),8.84(s,1H),8.07(s,1H),7.86(s,1H),7.64(d,J=8.0Hz,1H),7.43~7.40(m,3H),6.78~6.73(m,2H),5.02(s,2H),3.73(t,J=4.5,5.0Hz,4H),2.98(t,J=4.5,5.0Hz,4H).
MS(ESI,[M+H]+)m/z:438.1.
Example preparation of 446- ((5-chloro-2- ((4- (2- (pyrrolidin-1-yl) ethoxy) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-44)
Figure BDA0001216697810000512
Compound 3(0.5g,1.69mmol), compound 6-9(418mg,2.02mmol) were placed in a 100mL three-necked flask, 5mL of 2-butanol was added, trifluoroacetic acid (0.38mL,5.07mmol) was further added thereto, nitrogen gas was replaced, the mixture was heated in an oil bath under reflux for 4 hours, and the solvent was removed by concentration under reduced pressure after the completion of the reaction. Ammonia gas in methanol was added to adjust pH 8, and methanol was added to the solution. Column chromatography gave the crude product, which was slurried with ethyl acetate (5mL) and filtered under suction to give compound I-44(340 mg).
1H NMR(500MHz,DMSO-d6)δ9.18(s,1H),9.05(s,1H),8.86(s,1H),8.08(s,1H),7.85(s,1H),7.64(d,J=8.0Hz,1H),7.46~7.40(m,3H),6.71(d,J=9.0Hz,2H),5.02(s,2H),3.98(t,J=6.0,5.5Hz,2H),2.77(t,J=5.5,6.0Hz,2H),2.69~2.33(m,4H),1.69(t,J=3.5,3.0Hz,4H).
MS(ESI,[M+H]+)m/z:466.1.
Example 456 preparation of- ((2- ((4- (4-methylpiperazin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1- (3H) -ol (Compound I-45)
Figure BDA0001216697810000521
Compound 2(2.0g,13.4mmol), compounds 1-9(2.0g,13.4mmol), n-butanol (30mL), DIPEA (4.4mL,26.8mmol) were placed in a 100mL three-necked flask, replaced with nitrogen, heated in an oil bath under reflux for 9h, and the solvent was dried by spinning after the reaction was complete to give a brown oil. 60mL of water was added to the oil, sonicated until a solid appeared, and filtered with suction to give a yellow solid. The yellow solid was slurried with 10mL of ethanol and suction filtered to give off-white powder compound 3-23(1.84 g).
The compound 3-23(0.5g,2.3mmol) and the compound 6-1(484mg,2.5mmol) were placed in a 100mL three-necked flask, 5mL of 2-butanol was added, trifluoroacetic acid (0.5mL,6.9mmol) was further added thereto, nitrogen gas was replaced, the mixture was heated in an oil bath under reflux for 3 hours, and the solvent was spin-dried after the reaction. Ammonia gas in methanol was added to adjust pH 8, and methanol was added to the solution. And (4) performing column chromatography to obtain a crude product. The crude product was purified by TLC to give Compound I-45(110 mg).
1H NMR(500MHz,DMSO-d6)δ9.40(s,1H),9.28(s,1H),8.93(s,1H),8.15(s,1H),7.95(d,J=5.5Hz,1H),7.78(d,J=8.0Hz,1H),7.61(d,J=7.5Hz,2H),7.32(d,J=8.0Hz,1H),6.85(d,J=9.0Hz,2H),6.21(d,J=6.0Hz,1H),4.97(s,2H),3.08(t,J=4.5,4.5Hz,4H),2.51(t,J=5.0,5.0Hz,4H),2.29(s,3H).
MS(ESI,[M+H]+)m/z:417.1.
Example preparation of 467- ((5-chloro-2- ((4- (4-methylpiperazin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-46)
Figure BDA0001216697810000531
Compound 3-14(500mg,1.69mmol), compound 6-1(387mg,2.02mmol), 2-butanol (5mL) and trifluoroacetic acid (0.4mL,5.07mmol) were placed in a 25mL pressure resistant tube and reacted with microwave heating at 140 ℃ for 2 hours. The reaction mixture was filtered with suction, and the filter cake was washed with a mixed solvent of ethanol and methylene chloride (10mL:10 mL). The filtrate was evaporated to dryness under reduced pressure to give an oil, which was dissolved in 30mL of dichloromethane, the pH was adjusted to 8 with saturated aqueous sodium carbonate solution, and the organic phase was separated and taken out. The aqueous phase was extracted with 15mL dichloromethane, the organic phases were combined, dried over anhydrous sodium sulfate and chromatographed to give the crude product. The crude product was slurried with ethanol (10mL) and suction filtered to give a white solid, compound I-46(140 mg).
1H NMR(500MHz,DMSO-d6)δ9.60(s,1H),9.22(s,1H),8.45(s,1H),8.40(s,1H),8.17(s,1H),7.48~7.41(m,3H),7.10(d,J=7.5Hz,1H),6.90(d,J=9.0Hz,2H),5.03(s,2H),3.09(t,J=4.5,5.0Hz,4H),2.47(t,J=4.5,5.0Hz,4H),2.23(s,3H).
MS(ESI,[M+H]+)m/z:451.2.
Example preparation of 471- (4- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-5-yl) amino) pyrimidin-2-yl) amino) phenyl) -4-methylpiperazine-1, 4-dimesylate (Compound I-47)
Figure BDA0001216697810000532
The compound 3-31(1.5g,5.2mmol) and the compound 6-1(1.19g,6.2mmol) were placed in a 250mL three-necked flask, 15mL of 2-butanol was added thereto, trifluoroacetic acid (1.0mL,15.0mmol) was further added thereto, nitrogen gas was replaced, the mixture was heated under reflux for 6 hours in an oil bath, and after the reaction was completed, the solvent was removed by concentration under reduced pressure. Chloroform was added to the residue until the solution became clear, a saturated aqueous solution of sodium hydrogencarbonate was adjusted to pH 7, and the organic phase was separated and washed once with saturated brine. Drying with anhydrous sodium sulfate, concentrating under reduced pressure to dryness, purifying by column chromatography to obtain brown crude product, pulping the crude product with ethyl acetate (10mL), vacuum filtering to obtain off-white solid (1g), suspending in acetone, adding 0.22g methanesulfonic acid, heating in 50 deg.C oil bath, stirring for 40 min, and concentrating under reduced pressure to remove solvent. Adding ethanol, concentrating under reduced pressure to dry, and repeating for 2 times. Acetone (6mL) was slurried and suction filtered to give a white solid, Compound I-47(517 mg).
1H NMR(500MHz,DMSO-d6)δ9.88(s,1H),9.70(s,2H),8.29(s,1H),7.73(d,J=7.5Hz,2H),7.56(d,J=7.5Hz,1H),7.34(d,J=8.5Hz,2H),6.92(d,J=8.5Hz,2H),4.93(s,2H),3.78(d,J=13.0Hz,2H),3.54(d,J=11.5Hz,2H),3.20~2.88(m,7H),2.41(s,6H).
MS(ESI,[M+H]+)m/z:451.0.
Example 487 preparation of- ((5-chloro-2- ((4- (4-methylpiperazin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) -3, 4-dihydro-1H-benzo [ c ] [1,2] boran-1-ol (Compound I-48)
Figure BDA0001216697810000541
Compound 3-18(0.5g,1.613mmol) and compound 6-1(0.37g,1.694mmol) were dissolved in 2-butanol (8mL) with stirring, trifluoroacetic acid (0.55g,4.839mmol) was added, and the reaction was heated with microwave at 160 ℃ for 1.5 hours. And after the reaction is finished, adding 10mL of purified water and 10mL of dichloromethane to dilute the reaction solution, adjusting the pH value of a water layer to 7-8 by using a saturated sodium bicarbonate solution to generate a large amount of gray solid, and performing suction filtration on the suspension to obtain a white-like solid. Dissolving the solid in 10ml of methanol, heating until the solid refluxes, closing the heating, naturally cooling to room temperature, and carrying out suction filtration to obtain the off-white solid. Drying in vacuo afforded Compound I-48(247 mg).
1H NMR(500MHz,DMSO-d6)δ8.976(s,1H),8.755(s,1H),8.412(s,1H),8.050(s,1H),7.798~7.794(m,1H),7.596~7.581(m,1H),7.397~7.379(m,2H),7.208~7.191(m,1H),6.732~6.714(m,2H),4.122~4.099(t,2H),3.025~3.005(m,4H),2.558~2.873(m,2H),2.454~2.434(m,4H),2.221(s,3H).
13C NMR(125MHz,DMSO-d6)δ158.40,156.73,146.45,133.00,127.20,120.85,116.13,63.83,55.18,49.37,46.25,31.97.
MS(ESI,[M+H]+)m/z:465.1.
Example preparation of 496- ((5-chloro-2- ((4- (4-methylpiperazin-1-yl) phenyl) amino) pyrimidin-4-yl) -5-fluorobenzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-49)
Figure BDA0001216697810000542
Compounds 3-32(0.5g,1.59mmol) and 6-1(0.367g,1.67mmol) were dissolved in 2-butanol (8mL) and stirred to dissolve, trifluoroacetic acid (0.54g,4.78mmol) was added, and the reaction was heated to reflux for 6 hours. After the reaction is finished, the 2-butanol is removed by rotary evaporation under reduced pressure. 10mL of purified water and 10mL of methylene chloride were added thereto, and the pH of the aqueous layer was adjusted to 7 to 8 with a saturated aqueous solution of sodium bicarbonate to obtain a suspension. The suspension was suction filtered to give an off-white solid. Drying in vacuo afforded Compound I-49(310 mg).
1H NMR(500MHz,DMSO-d6)δ9.269(s,1H),8.980(s,1H),8.824(s,1H),8.066(s,1H),7.765~7.750(m,2H),7.437~7.417(m,1H),7.261~7.244(m,2H),6.627~6.609(m,2H),5.042(s,2H),3.184~3.173(m,4H),2.999(br,4H),2.280(s,3H).
13C NMR(125MHz,DMSO-d6)δ158.32,157.53,131.76,120.26,116.06,109.78,109.60,70.15,54.93,49.07.
MS(ESI,[M+H]+)m/z:469.2.
Example preparation of- ((5-chloro-2- ((4- (4- (dimethylamino) piperidin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-50)
Figure BDA0001216697810000551
Compound 3(2g,6.76mmol) and compound 6-10(1.55g,7.10mmol) were dissolved in 2-butanol (20mL) with stirring, trifluoroacetic acid (2.31g,20.28mmol) was added, and the reaction was heated to reflux for 6 hours. And (3) after the reaction is finished, evaporating 2-butanol under reduced pressure, adding 10mL of purified water and 10mL of dichloromethane into the residue, adjusting the pH value of a water layer to 7-8 by using a saturated sodium bicarbonate solution to obtain a suspension, carrying out suction filtration on the suspension to obtain an off-white solid, and carrying out vacuum drying to obtain a compound I-50(1 g).
1H NMR(500MHz,CDCl3-d1)δ8.767~8.744(m,2H),8.481~8.465(m,1H),8.205~8.157(m,2H),8.127~8.107(m,1H),7.685~7.653(m,2H),5.912(s,2H),4.430(br,2H),3.483~3.444(m,2H),3.134-3.119(m,7H),2.746(br,2H),2.484~2.462(m,2H).
13C NMR(125MHz,CDCl3-d1)δ161.96,160.01,157.60,153.37,150.99,141.03,136.36,129.22,127.55,125.24,125.16,121.56,108.20,75.07,65.99,54.19,44.97,31.69.
MS(ESI,[M+H]+)m/z:479.1.
Example preparation of 514- ((5-chloro-2- ((4- (4-methylpiperazin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-51)
Figure BDA0001216697810000552
The compounds 3-33(0.5g,1.69mmol) and 6-1(0.34g,1.77mmol) were dissolved in 2-butanol (8mL) with stirring, and trifluoroacetic acid (0.578g,5.07mmol) was added thereto, followed by reaction with microwave heating at 140 ℃ for 2 hours. After the reaction, the solvent was distilled off under reduced pressure to give a brown oil. The reaction solution was diluted with 10mL of purified water and 10mL of dichloromethane, and the pH of the aqueous layer was adjusted to 7-8 with a saturated aqueous solution of sodium bicarbonate to give a large amount of gray solid. The suspension was filtered with suction to give a grey solid. 50mL of ethanol is added into the mixture to be heated and dissolved, then the mixture is naturally cooled to room temperature, and a sample is filtered by suction to obtain a gray solid. Drying in vacuo afforded Compound I-51(180 mg).
1H NMR(500MHz,CDCl3-d1)δ8.956-8.889(m,1H),8.644~8.526(m,2H),8.439~8.267(m,2H),8.248~8.094(m,2H),7.759~7.607(m,2H),6.023~5.997(m,2H),4.153(br,4H),3.751(br,4H),3.441(s,3H).
13C NMR(125MHz,CDCl3-d1)δ161.84,160.34,157.84,152.23,150.05,136.83,135.65,132.19,131.80,131.11,124.72,121.12,108.10,74.20,58.46,53.03,48.95.
MS(ESI,[M+H]+)m/z:451.1.
Example 526 preparation of- ((5-fluoro-2- ((4- (4-methylpiperazin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-52)
Figure BDA0001216697810000561
Compound 3-22(1.0g,3.57mmol) and compound 6-1(0.72g,3.75mmol) were dissolved in 2-butanol (8mL) with stirring, trifluoroacetic acid (1.22g,10.71mmol) was added, and the reaction was heated with microwave at 160 ℃ for 2 hours. After the reaction, the solvent was distilled off under reduced pressure to give a brown oil. The reaction solution was diluted with 10mL of purified water and 10mL of dichloromethane, and the pH of the aqueous layer was adjusted to 7-8 with a saturated sodium bicarbonate solution. The dichloromethane layer was evaporated under reduced pressure to remove the solvent to give an oil. The oil was dissolved by heating with 10mL of methanol, then cooled naturally to room temperature, and the sample was filtered with suction to give a gray solid. Drying in vacuo afforded Compound I-52(200 mg).
1H NMR(500MHz,CDCl3-d1)δ8.843(s,1H),8.689~8.682(m,1H),8.586~8.570(m,1H),8.278~8.242(m,3H),8.154~8.137(m,1H),7.749~7.731(m,2H),5.944(s,3H),4.043~4.035(m,4H),3.586(br,4H),3.282(s,3H).
13C NMR(125MHz,CDCl3-d1)δ159.79,154.50,154.42,152.95,150.15,145.64,143.55,143.38,141.28,137.36,128.51,126.65,125.23,124.88,121.37,75.09,58.59,53.42,49.14.
MS(ESI,[M+H]+)m/z:435.1
Example 534 preparation of (4- ((5-chloro-4- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) pyrimidin-2-yl) amino) phenyl) thiomorpholine 1, 1-dioxide (Compound I-53)
Figure BDA0001216697810000571
Compound 3(0.498g,1.68mmol) and compound 6-11(0.40g,1.76mmol) were dissolved in 2-butanol (8mL) with stirring, and trifluoroacetic acid (0.574g,5.04mmol) was added and the reaction was heated with a microwave at 140 ℃ for 2 hours. And after the reaction is finished, adding 5mL of purified water and 10mL of chloroform to dilute the reaction solution, and adjusting the pH value of a water layer to 7-8 by using a saturated sodium bicarbonate solution. The aqueous layer was discarded, the organic layer was evaporated under reduced pressure to remove the solvent, 10mL of ethanol was added to the residue and heated to reflux to dissolve it, then it was naturally cooled to room temperature, the sample was filtered under suction to obtain a gray solid, which was dried under vacuum to obtain Compound I-53(400 mg).
1H NMR(500MHz,DMSO-d6)δ9.172(s,1H),9.044(s,1H),8.856(s,1H),8.080(s,1H),7.855(s,1H),7.652~7.636(m,1H),7.460~7.404(m,3H),6.833~6.816(m,2H),5.026(s,2H),3.640(br,4H),3.105(br,4H).
13C NMR(125MHz,DMSO-d6)δ158.31,156.83,155.03,142.97,137.80,133.71,127.62,126.60,121.75,120.91,116.93,103.61,70.36,50.34,48.14.
MS(ESI,[M+H]+)m/z:486.0.
Example preparation of 546- ((5-chloro-2- ((4- (4- (ethylsulfonyl) piperazin-1-yl) phenyl) amino) pyrimidin-4-yl) amino) benzo [ c ] [1,2] oxaborzol-1 (3H) -ol (Compound I-54)
Figure BDA0001216697810000572
Compound 3(0.537g,1.27mmol) and compound 6-12(0.36g,1.33mmol) were dissolved in 2-butanol (8mL), and trifluoroacetic acid (0.45g,3.99mmol) was added thereto with stirring and dissolved, followed by reaction with microwave heating at 140 ℃ for 2 hours. After the reaction, the solvent was distilled off under reduced pressure. The reaction solution was diluted with 10mL of purified water and 50mL of ethyl acetate, and the pH of the aqueous layer was adjusted to 7-8 with a saturated aqueous solution of sodium bicarbonate. Extraction was performed twice with ethyl acetate (50mL), the combined organic layers were evaporated under reduced pressure to remove the solvent, and the residue was purified by column chromatography and dried in vacuo to give compound I-54(120 mg).
1H NMR(500MHz,DMSO-d6)δ9.169(s,1H),9.026(s,1H),8.848(s,1H),8.070(s,1H),7.862~7.859(m,1H),7.647~7.633(m,1H),7.445~7.403(m,3H),6.783~6.765(m,2H),5.032(s,2H),3.305~3.282(m,2H),3.125~3.066(m,8H),1.261~1.231(t,3H).
13C NMR(125MHz,DMSO-d6)δ158.31,156.84,155.01,145.83,137.82,133.84,127.59,126.60,121.75,120.57,117.12,103.54,70.36,56.50,49.87,45.69,42.89,19.02,7.95.
MS(ESI,[M+H]+)m/z:528.
Experimental example 1 Activity experiments on inhibition of EGFR (WT), EGFR (T790M/L858R) and EGFR (T790M) in vitro
1.1 EGFR (WT) inhibitory Activity Screen
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20) 100 ng/. mu.L of EGFR (WT) stock solution was diluted, 6. mu.L of diluted EGFR (WT) stock solution (final concentration of 0.5 ng/. mu.L) was added to each well, and different compounds of each concentration dissolved in DMSO were added to each well, 7 different concentrations were set for each compound, and 2 duplicate wells were set for a blank control well (containing no enzyme) and a negative control well (containing enzyme, adding vehicle DMSO). After the enzyme had reacted with the compound or vehicle for 10 minutes, 5X 25. mu.M ATP (final concentration of 5. mu.M) prepared with kinase buffer and 5X 0.5. mu.M substrate (final concentration of 0.1. mu.M, ULight-poly GT) were mixed in a 1:1 ratio and then 4. mu.L of the mixture per well was added to the wells; after the plate was sealed and the plate was coated with the membrane, after 1 hour at room temperature, 5. mu.L of 4X 40mM EDTA (final concentration: 10mM) was added to each well for 5 minutes at room temperature, 5. mu.L of 4X 8nM detection reagent (final concentration: 2nM, Eu-anti-phosphorus-tyrosine antibody) was added to each well, the mixture was incubated at room temperature for 1 hour, and then detected by the Htrf method using a plate reader (excitation 320nM, emission 665 nM). Calculating the inhibition ratio, and calculating IC50Values (table 1).
1.2 EGFR (T790M/L858R) inhibition Activity screening
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20) 70 ng/. mu.L of EGFR (T790M L858R) stock solution was diluted, 6. mu.L of diluted EGFR (T790M L858R) was added per well (final concentration 0.02 ng/. mu.L), and DMSO-solubilized different compounds were added to the wells at respective concentrations, 7 different concentrations being set for each compound, and blank control wells (no enzyme) and negative control wells (enzyme-containing, vehicle DMSO-containing) were set, and 2 replicate wells were set. After the enzyme reacts with the compound or the solvent for 30min, 5 × 5 μ M ATP (final concentration of 1 μ M) prepared with a kinase buffer solution and 5 × 0.5 μ M substrate (final concentration of 0.1 μ M, ULight-poly GT) are mixed in a ratio of 1:1, and then 4 μ L of the mixture per well is added to the well; after the plate is sealed and the plate is covered with a membrane, after 1 hour at room temperature, 5. mu.L of 4X 40mM EDTA (final concentration 10mM) is added to each well for 5min at room temperature, and 5. mu.L of 4X 8nM detection reagent (final concentration 2nM, Eu-anti-phosphorus-tyrosine antibody) is added to each well and incubated for 1 hour at room temperature; and (3) detecting by a plate reader Htrf method (excitation is 320nm, and emission is 665 nm). Calculating the inhibition ratio, and calculating IC50Values (table 1).
1.3 EGFR (T790M) inhibitory Activity screening
With kinase buffer (50mM HEPES, 50mM MgCl)2、10mM MnCl21mM DTT, 25mM SEB) 114.8 ng/. mu.L of EGFR (T790M) stock was diluted, 6. mu.L of diluted EGFR (T790M) (final concentration of 1.5 ng/. mu.L) was added to each well, DMSO-solubilized various concentrations of different compounds were added to each well, 7 concentrations of each compound were set, and 2 replicate wells were set for a blank control well (no enzyme) and a negative control well (enzyme-containing, vehicle DMSO). After the enzyme reacts with the compound or vehicle for 10min, 5 × 20 μ M ATP (final concentration of 4 μ M) prepared with kinase buffer and 5 × 5 μ M Substrate (final concentration of 1 μ M TK Substrate-Substrate) were mixed at a ratio of 1:1, and then 4 μ L of the mixture per well was added to the wells; after the plate is sealed and the membrane is sealed, after the reaction is carried out for 1h at room temperature, 5 mu L of 1 multiplied TK Antibody and 62.5nM XL-665 are added into each hole, and after the mixture is mixed according to the ratio of 1:1, 10 mu L of each hole is added into each hole; after the plate is sealed by a sealing film, incubating for 1 hour at room temperature; and (3) detecting by a plate reader Htrf method (excitation is 320nm, and emission is 665 nm).
In the present invention, EGFR (wt) refers to wild EGFR; EGFR (T790M/L858R) refers to EGFR with double mutations of T790M and L858R; EGFR (T790M) refers to EGFR with the T790M mutation.
Table 1 shows the activity data of the compounds on wild-type EGFR, two mutant EGFR (T790M/L858R, T790M). Activity IC provided by a Compound designated "A50Less than or equal to 10 nM; IC provided by a compound whose activity is designated as "B50Is 10-100 nM; IC provided by a compound whose activity is designated as "C50100-; activity assignment IC provided by "D" Compounds501000-; IC provided by a compound whose activity is designated as "E50Is more than or equal to 10000 nM.
Table 1: inhibitory Activity of Compounds against EGFR (WT), EGFR (T790M/L858R) and EGFR (T790M)
Figure BDA0001216697810000591
Figure BDA0001216697810000601
Figure BDA0001216697810000611
Experimental example 2 test for inhibitory Activity of in vitro kinases ALK (WT), ALK (C1156Y) and ALK (L1196M)
2.1 ALK (WT) inhibitory Activity Screen
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20) 330 ng/. mu.L of ALK (WT) stock solution was diluted, 6. mu.L of diluted ALK (WT) (final concentration of 0.01 ng/. mu.L) was added to each well, DMSO-dissolved different compounds at respective concentrations were added to each well, 7 concentrations of each compound were set, and 2 duplicate wells were set with a blank control well (containing no enzyme) and a negative control well (containing enzyme, adding vehicle DMSO). After the enzyme reacts with the compound or the solvent for 10min, 50uM ATP (terminal ATP) prepared by using a kinase buffer solution is addedConcentration 10uM) and 0.5. mu.M substrate (final concentration 0.1. mu.M, ULight-poly GT) at a ratio of 1:1, and then adding 4. mu.L of the mixture per well to the wells; after the plate is sealed and the plate is covered with a membrane, after reacting for 2h at room temperature, 5 μ L of 40mM EDTA (final concentration is 10mM) is added into each hole, the room temperature is 5min, 5 μ L of 8nM detection reagent (final concentration is 2nM, Eu-anti-phosphorus-tyrosine antibody) is added into each hole, and the incubation is carried out for 1h at room temperature; BMG instruments read plates (excitation 620nm, emission 665 nm). Calculating the inhibition ratio, and calculating IC50Values (table 2).
2.2 ALK (C1156Y) inhibition Activity screening
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20) 325 ng/. mu.L of ALK (C1156Y) stock solution was diluted, 6. mu.L of diluted ALK (C1156Y) (final concentration of 0.03ng/ul) was added to each well, DMSO-dissolved different compounds were added to each well at respective concentrations, 7 concentrations were set for each compound, and blank control wells (no enzyme) and negative control wells (enzyme-containing, vehicle DMSO-added) were set at 2 duplicate wells. After the enzyme has reacted with the compound or vehicle for 10min, 5 × 50 μ M ATP (final concentration 10 μ M) prepared in kinase buffer is mixed with 5 × 500nM substrate (final concentration 100nM, ULight-poly-GT) in a 1:1 ratio, and 4 μ L of the mixture per well is added to the wells; after the plate was sealed and the plate was coated with a membrane, after reacting at room temperature for 2 hours, 5. mu.L of 4X 40mM EDTA (final concentration: 10mM) was added to each well for 5 minutes at room temperature, and 5. mu.L of 4X 8nM detection reagent (final concentration: 2nM, Ab detection antibody, PE) was added to each well and incubated at room temperature for 1 hour; BMG instruments read plates (excitation 320or 340nm, emission 665 nm). Calculating the inhibition ratio, and calculating IC50Values (table 2).
2.3 ALK (L1196M) inhibitory Activity screening
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20) 271 ng/. mu.L of ALK (L1196M) stock solution was diluted, 6ul of diluted ALK (L1196M) (final concentration of 0.03 ng/. mu.L) was added to each well, DMSO-dissolved different compounds at various concentrations were added to the wells, 7 concentrations were set for each compound, and blank control wells (no enzyme) and negative control wells (enzyme-containing, vehicle DMSO-added) were set for 2 replicate wells. After the enzyme reacts with the compound or the solvent for 10min, the mixture is used5 X50. mu.M ATP (final concentration 10. mu.M) in kinase buffer was mixed with 5 X500 nM substrate (final concentration 100nM, ULight-poly-GT) at a ratio of 1:1 and then added to wells as 4. mu.L of the mixture per well; after the plate was sealed and the plate was coated with a membrane, after reacting at room temperature for 2 hours, 5. mu.L of 4X 40mM EDTA (final concentration: 10mM) was added to each well for 5 minutes at room temperature, and 5. mu.L of 4X 8nM detection reagent (final concentration: 2nM, Ab detection antibody, PE) was added to each well and incubated at room temperature for 1 hour; BMG instruments read plates (excitation 320or 340nm, emission 665 nm). Calculating the inhibition ratio, and calculating IC50Values (table 2).
In the present invention, ALK (wt) means wild ALK, unless otherwise specified; ALK (C1156Y) refers to ALK with a C1156Y mutation; ALK (L1196M) refers to ALK with the L1196M mutation.
Table 2 shows the activity data of compounds on ALK (wt), ALK (C1156Y), ALK (L1196M). Activity IC provided by a Compound designated "A50Less than or equal to 30 nM; IC provided by a compound whose activity is designated as "B50Is 30-100 nM; IC provided by a compound whose activity is designated as "C50100-; activity assignment IC provided by "D" Compounds50Is more than or equal to 1000 nM.
Table 2: inhibitory Activity of Compounds on ALK (WT), ALK (C1156Y) and ALK (L1196M)
Figure BDA0001216697810000621
Figure BDA0001216697810000631
Experimental example 3 inhibition of in vitro kinase JAK1, JAK2, and JAK3
3.1 JAK1 inhibitory Activity screening
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20) 50 ng/. mu.L of JAK1 stock solution was diluted, 6. mu.L of diluted JAK1 (final concentration of 0.5 ng/. mu.L) was added to each well, 7 compounds each dissolved in DMSO were added to each wellThe concentration was determined by mixing a blank control well (containing no enzyme) and a negative control well (containing enzyme and DMSO in vehicle) with 2 duplicate wells. After 30min of reaction of the enzyme with the compound or vehicle, 5 × 250 μ M ATP (final concentration 50 μ M) and 5 × 0.5 μ M substrate (final concentration 0.1 μ M, μ M Light-poly GT) in kinase buffer were mixed as 1:1 mixing, adding 4 mu L of mixture into each hole; after the plate was sealed with a membrane, after reacting at room temperature for 2 hours, 5. mu.L of 4X 40mM EDTA (final concentration: 10mM) was added to each well for 5 minutes at room temperature, and 5. mu.L of 4X 8nM detection reagent (final concentration: 2nM, Ab detection antibody, PE) was added to each well and incubated at room temperature for 1 hour; htrf method (excitation 620nm, emission 665 nm). Calculating the inhibition ratio, and calculating IC50Values (table 1).
3.2 JAK2 inhibitory Activity screening
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20) 50 ng/. mu.L of JAK2 stock solution was diluted, 6. mu.L of diluted JAK2 (final concentration of 0.005 ng/. mu.L) was added to each well, DMSO-dissolved different compounds at respective concentrations were added to each well, 7 concentrations of each compound were set, and 2 duplicate wells were set with a blank control well (containing no enzyme) and a negative control well (containing enzyme, adding vehicle DMSO). After 30min of reaction of the enzyme with the compound or vehicle, 5 × 100 μ M ATP (final concentration 20 μ M) and 5 × 0.5 μ M substrate (final concentration 0.1 μ M, μ M Light-poly GT) in kinase buffer were mixed as 1:1 mixing, adding 4 mu L of mixture into each hole; after the plate was sealed and the plate was coated with a membrane, after reacting at room temperature for 2 hours, 5. mu.L of 4X 40mM EDTA (final concentration: 10mM) was added to each well for 5 minutes at room temperature, and 5. mu.L of 4X 8nM detection reagent (final concentration: 2nM, Ab detection antibody, PE) was added to each well and incubated at room temperature for 1 hour; htrf method (excitation 620nm, emission 665 nm). Calculating the inhibition ratio, and calculating IC50Values (table 1).
3.3 JAK3 inhibitory Activity screening
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20) 50 ng/. mu.L of JAK3 stock solution was diluted, 6. mu.L of diluted JAK3 (final concentration of 0.01 ng/. mu.L) was added to each well, and DMSO-dissolved different compounds were added to each well at different concentrations7 concentrations, blank control wells (no enzyme) and negative control wells (enzyme, vehicle DMSO), 2 replicate wells. After 30min of reaction of the enzyme with the compound or vehicle, 5 × 100 μ M ATP (final concentration 20 μ M) and 5 × 0.5 μ M substrate (final concentration 0.1 μ M, μ M Light-poly GT) in kinase buffer were mixed as 1:1 mixing, adding 4 mu L of mixture into each hole; after the plate was sealed and the plate was coated with a membrane, after reacting at room temperature for 2 hours, 5. mu.L of 4X 40mM EDTA (final concentration: 10mM) was added to each well for 5 minutes at room temperature, and 5. mu.L of 4X 8nM detection reagent (final concentration: 2nM, Ab detection antibody, PE) was added to each well and incubated at room temperature for 1 hour; htrf method (excitation 620nm, emission 665 nm). Calculating the inhibition ratio, and calculating IC50Values (table 1).
3.4 TYK2 inhibition Activity screening
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20) 50ng/μ L of TYK2 stock solution was diluted, 6 μ L of diluted TYK2 (final concentration of 0.5ng/μ L) was added per well, DMSO-dissolved different compounds at various concentrations were added to the wells, 7 concentrations per compound, blank control empty (no enzyme) and negative control wells (enzyme containing, vehicle DMSO), 2 replicate wells. After 30min of reaction of the enzyme with the compound or vehicle, 5 × 100 μ M ATP (final concentration of 20 μ M) and 5 × 0.5 μ M substrate (final concentration of 0.1 μ M, μ Light-poly GT) in kinase buffer were mixed as described in 1:1 mixing, adding 4 mu L of mixture into each hole; after the plate was sealed and the plate was coated with a membrane, after reacting at room temperature for 2 hours, 5. mu.L of 4X 40mM EDTA (final concentration: 10mM) was added to each well for 5 minutes at room temperature, and 5. mu.L of 4X 8nM detection reagent (final concentration: 2nM, Ab detection antibody, PE) was added to each well and incubated at room temperature for 1 hour; plate reading (excitation 620nm, emission 665nm) was done with MD instruments. Calculating the inhibition ratio, and calculating IC50Values (table 3).
Table 3 shows the activity data of compounds against JAK1, JAK2, JAK 3. Activity IC provided by a Compound designated "A50Less than or equal to 10 nM; IC provided by a compound whose activity is designated as "B50Is 10-100 nM; IC provided by a compound whose activity is designated as "C50100-; activity assignment IC provided by "D" Compounds50Is more than or equal to 1000 nM.
Table 3: inhibitory Activity of Compounds against JAK1, JAK2, JAK3, TYK2
Figure BDA0001216697810000651
Figure BDA0001216697810000661
In the activity test experiments described above, the following terms, if not otherwise specified, have the following meanings:
EDTA refers to ethylene diamine tetraacetic acid; EGTA refers to ethylene glycol bis (2-aminoethyl ether) tetraacetic acid; DTT refers to dithiothreitol; 0.01% Tween 20 refers to Tween 20 with a mass fraction of 0.01%; "4-fold gradient dilution" means that 3 volumes of the diluted solution are added to 1 volume of stock solution 1 to give stock solution 2; then 1 volume of stock solution 2 is taken and added with 3 volumes of diluted solution to obtain stock solution 3; by analogy, solutions with different concentrations are obtained; "Final concentration" means the concentration in the whole reaction system at the start of the reaction, and is based on the total volume of the reaction; "X" indicates a multiple, i.e., a multiple of the current concentration based on the final concentration.

Claims (72)

1. A compound of formula (I) or a pharmaceutically acceptable salt thereof,
Figure FDA0002775244510000011
wherein the content of the first and second substances,
R1selected from hydrogen, fluorine, chlorine, bromine, iodine, C1-C6Alkyl or C1-C6Alkoxy radical, wherein C1-C6Alkyl is optionally substituted with fluoro, chloro, bromo or iodo;
x is selected from NH or O;
R3and R7Each independently selected from hydrogen or C1-C6An alkoxy group;
R4is selected from-NHC (═ O) Ra;R6Selected from hydrogen; r5Selected from hydrogen, C1-C6Alkylamino or C1-C6Alkoxy, wherein said C1-C6Alkylamino or C1-C6The alkoxy group is substituted with: c1-C6Alkyl or-NRcRd
Or, R4And R6Each independently selected from hydrogen or C1-C6An alkyl group; r5Selected from ethoxy, morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl, wherein said morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl is optionally substituted with: o, -S (O)2Rb、C1-C6Alkyl, -NRcRdOr optionally is covered with C1-C6An alkyl-substituted 5-6 membered heterocycloalkyl wherein the ethoxy group is substituted with pyrrolidinyl;
Raselected from vinyl groups;
Rbis selected from C1-C6An alkyl group;
Rcand RdEach independently selected from hydrogen or C1-C6An alkyl group;
cy is selected from
Figure FDA0002775244510000012
Or
Figure FDA0002775244510000013
R2Each independently selected from fluorine, chlorine, bromine, iodine or C1-C6An alkoxy group;
n is 0 to 4;
Z1and Z2Each independently selected from hydrogen or C1-C6An alkyl group;
the A ring is a 5-6 membered ring, optionally substituted with C1-C6Alkyl substitution;
wherein the compound of formula (I) is not:
Figure FDA0002775244510000014
Figure FDA0002775244510000021
2. a compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is1Selected from hydrogen, fluorine, chlorine, bromine, C1-C3Alkyl or C1-C3Alkoxy radical, wherein C1-C3Alkyl is optionally substituted with fluorine.
3. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is1Selected from hydrogen, fluorine, chlorine, bromine, methyl, methoxy or trifluoromethyl.
4. A compound of formula (I) or a pharmaceutically acceptable salt thereof, as claimed in claim 1, R4And R6Each independently selected from hydrogen or C1-C3An alkyl group; r5Selected from ethoxy, morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl, wherein said morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl is optionally substituted with: o, -S (O)2Rb、C1-C3Alkyl, -NRcRdOr optionally is covered with C1-C6An alkyl-substituted 5-6 membered heterocycloalkyl, wherein the ethoxy group is substituted with pyrrolidinyl.
5. A compound of formula (I) or a pharmaceutically acceptable salt thereof, as claimed in claim 1, R4And R6Each independently selected from hydrogen or methyl; r5Selected from ethoxy, morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl, wherein said morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl is optionally substitutedThe following groups are substituted: o, -S (O)2Rb、C1-C3Alkyl, -NRcRdOr optionally is covered with C1-C6An alkyl-substituted 5-6 membered heterocycloalkyl, wherein the ethoxy group is substituted with pyrrolidinyl.
6. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4And R6At least one is selected from hydrogen.
7. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is3And R7Each independently selected from hydrogen or C1-C3An alkoxy group.
8. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is3And R7Each independently selected from hydrogen, methoxy or isopropyloxy.
9. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is3And R7At least one is selected from hydrogen.
10. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is3And R7Each independently selected from hydrogen, methoxy or isopropyloxy, and R3And R7At least one is selected from hydrogen.
11. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is3And R7Each independently selected from hydrogen or methoxy, and R3And R7At least one is selected from hydrogen.
12. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4Is selected from-NHC (═ O) Ra;R6Selected from hydrogen; r5Selected from hydrogen, C1-C3Alkylamino or C1-C3Alkoxy, wherein said C1-C3Alkylamino or C1-C3The alkoxy group is substituted with: c1-C6Alkyl or-NRcRd
13. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4Is selected from-NHC (═ O) Ra;R6Selected from hydrogen; r5Selected from hydrogen, ethylamino or ethoxy, wherein said ethylamino or ethoxy is substituted with: c1-C3Alkyl or-NRcRd
14. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4Is selected from-NHC (═ O) Ra;R6Selected from hydrogen; r5Selected from hydrogen, ethylamino or ethoxy, wherein ethoxy is-NRcRdIn which the ethylamino group is substituted by C1-C3Alkyl or-NRcRdAnd (4) substitution.
15. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4And R6Each independently selected from hydrogen or C1-C6An alkyl group; r5Selected from ethoxy, piperidinyl, morpholinyl, piperazinyl or thiomorpholinyl, wherein said piperidinyl, morpholinyl, piperazinyl or thiomorpholinyl is optionally substituted with: o, -S (O)2Rb、C1-C3Alkyl, -NRcRdOr optionally is covered with C1-C6An alkyl-substituted 5-6 membered heterocycloalkyl, wherein the ethoxy group is substituted with pyrrolidinyl.
16. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4And R6Are each independently selected from hydrogenOr C1-C6An alkyl group; r5Selected from ethoxy, morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl, wherein morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl is optionally substituted with: o, -S (O)2Rb、C1-C3Alkyl, -NRcRdOr optionally is covered with C1-C3An alkyl-substituted 5-6 membered nitrogen-containing heterocycloalkyl wherein the ethoxy group is substituted with pyrrolidinyl.
17. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R isbIs selected from C1-C3An alkyl group.
18. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R iscAnd RdEach independently selected from hydrogen or C1-C3An alkyl group.
19. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4Is selected from-NHC (═ O) Ra;R6Selected from hydrogen; r5Selected from hydrogen, ethylamino or ethoxy, wherein ethoxy is replaced by-N (CH)3)2In which the ethylamino group is substituted by methyl or-N (CH)3)2And (4) substitution.
20. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4Is selected from-NHC (═ O) Ra;R6Selected from hydrogen; r5Selected from hydrogen, ethylamino or ethoxy, wherein ethoxy is replaced by-N (CH)3)2In which the ethylamino group is simultaneously substituted by one methyl group and one-N (CH)3)2And (4) substitution.
21. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4Is selected from-NHC (═ O) Ra;R6Selected from hydrogen; r5Selected from hydrogen、
Figure FDA0002775244510000031
22. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4And R6Each independently selected from hydrogen or C1-C6An alkyl group; r5Selected from ethoxy, morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl, wherein ethoxy is substituted
Figure FDA0002775244510000041
Wherein morpholinyl, piperidinyl, piperazinyl or thiomorpholinyl is optionally substituted by ═ O,
Figure FDA0002775244510000042
Methyl, -N (CH)3)2Optionally C1-C3Alkyl-substituted piperidinyl or optionally substituted by C1-C3Alkyl substituted piperazinyl.
23. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4And R6Each independently selected from hydrogen or C1-C6An alkyl group; r5Selected from ethoxy,
Figure FDA0002775244510000043
Wherein the ethoxy group is
Figure FDA0002775244510000044
Is substituted in which
Figure FDA0002775244510000045
Optionally substituted by ═ O,
Figure FDA0002775244510000046
Methyl, -N (CH)3)2Piperidinyl, piperazinyl4-methylpiperazinyl substitution.
24. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4And R6Each independently selected from hydrogen or C1-C6An alkyl group; r5Selected from ethoxy,
Figure FDA0002775244510000047
Wherein the ethoxy group is
Figure FDA0002775244510000048
Is substituted in which
Figure FDA0002775244510000049
Optionally substituted by methyl or
Figure FDA00027752445100000410
Is substituted in which
Figure FDA00027752445100000411
Optionally piperazinyl, 4-methylpiperazinyl or-N (CH)3)2Is substituted in which
Figure FDA00027752445100000412
Optionally substituted by ═ O.
25. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4And R6Each independently selected from hydrogen or C1-C6An alkyl group; r5Selected from ethoxy,
Figure FDA00027752445100000413
Wherein the ethoxy group is
Figure FDA00027752445100000414
Is substituted in which
Figure FDA00027752445100000415
Optionally substituted by methyl or
Figure FDA00027752445100000416
Is substituted in which
Figure FDA00027752445100000417
Optionally is covered with
Figure FDA00027752445100000418
or-N (CH)3)2Is substituted in which
Figure FDA00027752445100000419
Optionally substituted by ═ O.
26. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is4And R6Each independently selected from hydrogen or C1-C6An alkyl group; r5Is selected from
Figure FDA00027752445100000420
Figure FDA00027752445100000421
27. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 26 wherein R is4And R6Each independently selected from hydrogen or C1-C6An alkyl group; r5Is selected from
Figure FDA00027752445100000422
Figure FDA0002775244510000051
28. As claimed in claim 1A compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein R2Each independently selected from fluorine, chlorine, bromine, iodine or C1-C3An alkoxy group.
29. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is2Each independently selected from fluorine, chlorine, bromine, iodine.
30. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein R is2Each independently selected from methoxy, ethoxy or isopropoxy.
31. A compound of formula (i), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein n is 0or 1.
32. A compound of formula (i), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein Cy is selected from
Figure FDA0002775244510000052
Figure FDA0002775244510000053
33. A compound of formula (i), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein Cy is selected from
Figure FDA0002775244510000054
Figure FDA0002775244510000055
34. A compound of formula (i), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein Cy is selected from
Figure FDA0002775244510000056
Figure FDA0002775244510000057
35. A compound of formula (i), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein Cy is selected from
Figure FDA0002775244510000058
Wherein m is 1 or 2, R8And R9Each independently selected from hydrogen or C1-C6An alkyl group.
36. A compound of formula (i), or a pharmaceutically acceptable salt thereof, as claimed in claim 35 wherein Cy is selected from
Figure FDA0002775244510000059
Figure FDA0002775244510000061
Wherein m is 1 or 2, R8And R9Each independently selected from hydrogen or C1-C3An alkyl group.
37. A compound of formula (i), or a pharmaceutically acceptable salt thereof, as claimed in claim 35 wherein m is 1 or 2, R8And R9Each independently selected from hydrogen or methyl.
38. A compound of formula (i), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein Cy is selected from
Figure FDA0002775244510000062
Figure FDA0002775244510000063
Wherein R is8And R9Each independently selected from hydrogen or C1-C3An alkyl group.
39. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 38 wherein R is8And R9Each independently selected from hydrogen or methyl.
40. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 38 wherein R is8And R9Each independently selected from hydrogen.
41. A compound of formula (i), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein Cy is selected from
Figure FDA0002775244510000064
Figure FDA0002775244510000065
42. A compound of formula (II) or a pharmaceutically acceptable salt thereof,
Figure FDA0002775244510000071
wherein the content of the first and second substances,
R1、R2、R3、R4、R5、R6、R7x, n and Z2As defined in any one of claims 1 to 31;
m is 1 or 2;
R8and R9Each independently selected from hydrogen or C1-C6An alkyl group;
wherein the compound of formula (II) is not:
Figure FDA0002775244510000072
43. the method of claim 42The compound of formula (II) or a pharmaceutically acceptable salt thereof, wherein the structural unit
Figure FDA0002775244510000073
As defined for Cy in any one of claims 35 to 40.
44. A compound of formula (II) or a pharmaceutically acceptable salt thereof, as claimed in claim 42 wherein the structural unit
Figure FDA0002775244510000081
Is selected from
Figure FDA0002775244510000082
Figure FDA0002775244510000083
45. A compound of formula (V) or a pharmaceutically acceptable salt thereof,
Figure FDA0002775244510000084
wherein the content of the first and second substances,
R1、R2、R3、R5、R7、R8、R9、m、n、Z2and X is as defined in claim 42.
46. A compound of formula (III) or a pharmaceutically acceptable salt thereof,
Figure FDA0002775244510000085
wherein the content of the first and second substances,
R1、R2、R3、R4、R5、R6、R7x, n are as defined inAny one of claims 1-31;
wherein the compound of formula (III) is not:
Figure FDA0002775244510000086
47. a compound of formula (III) or a pharmaceutically acceptable salt thereof as claimed in claim 46 wherein the structural unit
Figure FDA0002775244510000087
Is as defined for Cy in any one of claims 33 to 34.
48. A compound of formula (III) or a pharmaceutically acceptable salt thereof as claimed in claim 46 wherein the structural unit
Figure FDA0002775244510000088
Is selected from
Figure FDA0002775244510000091
Figure FDA0002775244510000092
49. A compound of formula (VI) or a pharmaceutically acceptable salt thereof,
Figure FDA0002775244510000093
wherein the content of the first and second substances,
R1、R2、R3、R5、R7n and X are as defined in claim 46.
50. A compound of formula (VII) or a pharmaceutically acceptable salt thereof,
Figure FDA0002775244510000094
wherein the content of the first and second substances,
R1、R3、R7x and Cy are as defined in any one of claims 1-3, 7-11 or 32-41;
R4and R6Each independently selected from hydrogen and C1-C6Alkyl or C1-C6An alkoxy group;
Y1is selected from N or CH;
Y2selected from NH, CH2S or O;
q is 0 to 11;
R12selected from ═ O, -S (═ O)2Rb、C1-C6Alkyl, -NRcRdOr optionally is covered with C1-C6Alkyl-substituted 5-6 membered heterocycloalkyl;
Rbis selected from C1-C6An alkyl group;
Rcand RdEach independently selected from hydrogen or C1-C6An alkyl group;
wherein the compound of formula (VII) is not:
Figure FDA0002775244510000101
51. a compound of formula (VII) or a pharmaceutically acceptable salt thereof, as claimed in claim 50 wherein R is4And R6Each independently selected from hydrogen and C1-C3Alkyl or C1-C3An alkoxy group.
52. A compound of formula (VII) or a pharmaceutically acceptable salt thereof, as claimed in claim 50 wherein R is4And R6Each independently selected from hydrogen or methyl.
53. A compound of formula (VII) or a pharmaceutically acceptable salt thereof, as claimed in claim 50 wherein R is4And R6At least one is selected from hydrogen.
54. A compound of formula (VII) or a pharmaceutically acceptable salt thereof, as claimed in claim 50 wherein R is12Selected from ═ O, -S (═ O)2Rb、C1-C6Alkyl, -NRcRdOr optionally is covered with C1-C3Alkyl-substituted 5-6 membered heterocycloalkyl.
55. A compound of formula (VII) or a pharmaceutically acceptable salt thereof, as claimed in claim 50 wherein R is12Selected from ═ O, -S (═ O)2Rb、C1-C3Alkyl, -NRcRdOr optionally is covered with C1-C3An alkyl-substituted 5-6 membered nitrogen-containing heterocycloalkyl group.
56. A compound of formula (VII) or a pharmaceutically acceptable salt thereof, as claimed in claim 50 wherein R iscAnd RdEach independently selected from hydrogen or C1-C3An alkyl group.
57. A compound of formula (VII) or a pharmaceutically acceptable salt thereof, as claimed in claim 50 wherein R is12Is selected from ═ O,
Figure FDA0002775244510000102
Methyl, -N (CH)3)2Optionally C1-C3Piperazinyl substituted by alkyl or optionally substituted by C1-C3Alkyl-substituted piperidinyl.
58. A compound of formula (VII) or a pharmaceutically acceptable salt thereof, as claimed in claim 50 wherein R is12Is selected from ═ O,
Figure FDA0002775244510000111
Figure FDA0002775244510000112
Methyl or-N (CH)3)2
59. A compound of formula (VII) according to claim 50 wherein q is 0or 1, or a pharmaceutically acceptable salt thereof.
60. A compound of formula (VII) or a pharmaceutically acceptable salt thereof, as claimed in claim 50 wherein the structural unit
Figure FDA0002775244510000113
Is selected from
Figure FDA0002775244510000114
61. A compound of formula (VII) or a pharmaceutically acceptable salt thereof, as claimed in claim 60 wherein the structural unit
Figure FDA0002775244510000115
Is selected from
Figure FDA0002775244510000116
62. A compound of formula (VIII) or a pharmaceutically acceptable salt thereof,
Figure FDA0002775244510000117
wherein the content of the first and second substances,
R1、R2、R3、R7x, n and Z2As defined in any one of claims 1 to 3, 7 to 11 or 35 to 40;
R4、R6、R12、Y1、Y2and q is as defined in any one of claims 50 to 61;
m is 1 or 2;
R8and R9Each independently selected from hydrogen or C1-C6An alkyl group;
wherein the compound of formula (VIII) is not:
Figure FDA0002775244510000118
Figure FDA0002775244510000121
63. a compound of formula (VIII) according to claim 62, or a pharmaceutically acceptable salt thereof, wherein the building blocks
Figure FDA0002775244510000122
Is as defined for Cy in any one of claims 41 to 45.
64. A compound of formula (VIII) according to claim 62, or a pharmaceutically acceptable salt thereof, wherein the building blocks
Figure FDA0002775244510000123
Is selected from
Figure FDA0002775244510000124
Figure FDA0002775244510000125
65. A compound of formula (IX), or a pharmaceutically acceptable salt thereof,
Figure FDA0002775244510000126
wherein the content of the first and second substances,
R1、R2、R3、R7x, n are as defined in any one of claims 1-3, 7-11 or 32-34;
R4、R6、R12、Y1、Y2and q is as defined in any one of claims 50 to 61;
wherein the compound of formula (IX) is not:
Figure FDA0002775244510000131
66. the compound of formula (IX) or a pharmaceutically acceptable salt thereof of claim 65, wherein the structural unit
Figure FDA0002775244510000132
As defined for Cy in any one of claims 32 to 34.
67. The compound of formula (IX) or a pharmaceutically acceptable salt thereof of claim 65, wherein the structural unit
Figure FDA0002775244510000133
Is selected from
Figure FDA0002775244510000134
Figure FDA0002775244510000135
68. The following compounds, or a pharmaceutically acceptable salt thereof,
Figure FDA0002775244510000136
Figure FDA0002775244510000141
Figure FDA0002775244510000151
69. the following compounds are described in detail,
Figure FDA0002775244510000161
70. a pharmaceutical composition comprising a compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, or a compound of claim 69.
71. Use of a compound of any one of claims 1-68, or a pharmaceutically acceptable salt thereof, a compound of claim 69, or a pharmaceutical composition of claim 70, in the manufacture of a medicament for treating a tyrosine kinase mediated disease.
72. The use of claim 71, wherein the tyrosine kinase is selected from EGFR tyrosine kinase, ALK, or JAK.
CN201710047432.1A 2017-01-22 2017-01-22 Boron-containing compounds as tyrosine kinase inhibitors Active CN108341835B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201710047432.1A CN108341835B (en) 2017-01-22 2017-01-22 Boron-containing compounds as tyrosine kinase inhibitors

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201710047432.1A CN108341835B (en) 2017-01-22 2017-01-22 Boron-containing compounds as tyrosine kinase inhibitors

Publications (2)

Publication Number Publication Date
CN108341835A CN108341835A (en) 2018-07-31
CN108341835B true CN108341835B (en) 2021-01-15

Family

ID=62974365

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201710047432.1A Active CN108341835B (en) 2017-01-22 2017-01-22 Boron-containing compounds as tyrosine kinase inhibitors

Country Status (1)

Country Link
CN (1) CN108341835B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3969458A4 (en) 2019-05-13 2023-05-17 Borah, Inc. Chemical compounds
EP3994142A4 (en) * 2019-07-03 2023-09-06 Borah, Inc. Chemical compounds
WO2021061823A1 (en) * 2019-09-23 2021-04-01 Boragen, Inc. Chemical compounds

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1625400A (en) * 2002-02-01 2005-06-08 里格尔药品股份有限公司 2,4-pyrimidinediamine compounds and their uses
CN106478700A (en) * 2015-08-26 2017-03-08 杭州雷索药业有限公司 The phenyl aminess kinases inhibitor that boryl replaces

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1625400A (en) * 2002-02-01 2005-06-08 里格尔药品股份有限公司 2,4-pyrimidinediamine compounds and their uses
CN106478700A (en) * 2015-08-26 2017-03-08 杭州雷索药业有限公司 The phenyl aminess kinases inhibitor that boryl replaces

Also Published As

Publication number Publication date
CN108341835A (en) 2018-07-31

Similar Documents

Publication Publication Date Title
AU2021245168B2 (en) Amine-substituted aryl or heteroaryl compounds as ehmt1 and ehmt2 inhibitors
CN112955459A (en) Bicyclic peptide ligands and uses thereof
CA2988896A1 (en) Fused-ring or tricyclic aryl pyrimidine compound used as kinase inhibitor
AU2014356069A1 (en) Novel amino pyrimidine derivatives
EP3640247B1 (en) Syk inhibitor and use method therefor
WO2012061337A1 (en) Fgfr2 modulators
JP2024505732A (en) Pyridopyrimidinone derivatives and their production methods and uses
CN108341835B (en) Boron-containing compounds as tyrosine kinase inhibitors
JP7077323B2 (en) Quinazoline compound and its preparation method, use and pharmaceutical composition
CN111377907A (en) Polysubstituted phenylamino pyrimidine derivative and preparation method and application thereof
CN112812128A (en) Macrocyclic compounds as ALK and ROS modulators
TW202204351A (en) Compounds having a macrocyclic structure and uses thereof
CN108329274B (en) Bruton's tyrosine kinase inhibitors
CN109942556A (en) Pyrimidinone compound and its application
JP2022515309A (en) Substituted aryl compounds, their production methods and uses
CN113045569B (en) Compounds useful as RET kinase inhibitors and uses thereof
CN114105976A (en) Selective ROCK2 kinase inhibitors
CN103965161A (en) Substituted 2-aminopyridine inhibitor for protein kinase
WO2021104441A1 (en) Polyaromatic compound as egfr kinase inhibitor
CN115894381A (en) 2,4,5-trisubstituted pyrimidine compound and preparation method and application thereof
TW202313574A (en) 7-nitro-8-hydroxyquinoline derivative, its preparation method and its medical use
CN105541792B (en) Polycyclic class PI3K inhibitor
CN110776511B (en) Quinoline ring-containing BTK inhibitors
CN113966331A (en) Triaromatic ring compound containing urea structure and application thereof
CN113004252B (en) Aryl imidazole derivative 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
GR01 Patent grant
GR01 Patent grant