CN111153899A - Substituted pyridine compound, preparation method and application thereof - Google Patents

Substituted pyridine compound, preparation method and application thereof Download PDF

Info

Publication number
CN111153899A
CN111153899A CN201911043519.7A CN201911043519A CN111153899A CN 111153899 A CN111153899 A CN 111153899A CN 201911043519 A CN201911043519 A CN 201911043519A CN 111153899 A CN111153899 A CN 111153899A
Authority
CN
China
Prior art keywords
alkyl
compound
ring
formula
group
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.)
Granted
Application number
CN201911043519.7A
Other languages
Chinese (zh)
Other versions
CN111153899B (en
Inventor
易磊
宋智泉
田强
张帅
王波
蔡家强
王利春
王晶翼
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.)
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Original Assignee
Sichuan Kelun Biotech Biopharmaceutical 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 Sichuan Kelun Biotech Biopharmaceutical Co Ltd filed Critical Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Publication of CN111153899A publication Critical patent/CN111153899A/en
Application granted granted Critical
Publication of CN111153899B publication Critical patent/CN111153899B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention relates to a substituted pyridine compound, a preparation method and application thereof. In particular, the invention relates to compounds of formula I, processes for their preparationThe use of the compounds of formula I for the prophylaxis and/or treatment of diseases which are associated with the SHP2 enzyme, and pharmaceutical compositions containing the compounds of formula I.

Description

Substituted pyridine compound, preparation method and application thereof
Technical Field
The invention relates to the field of medicinal chemistry, in particular to a substituted pyridine compound serving as an SHP2 inhibitor, a preparation method thereof, a medicinal composition and application thereof in treating SHP2 enzyme-related diseases.
Background
SHP2(src homology 2 domain stabilizing phospho-side phosphatase 2) is a protein tyrosine phosphatase encoded by the gene PTPN11, structurally comprising two N-terminal SH2 (Srchology 2) domains, a Protein Tyrosine Phosphatase (PTP) catalytic domain and a C-terminal tail rich in proline groups and tyrosine phosphorylation sites. In the basal state, the SHP2 adopts a self-inhibiting configuration, with the N-SH2 domain sterically hindering access of the substrate to the active site. When ligand containing phosphorylated tyrosine residue is specifically combined with N-SH2 domain, the configuration of SHP2 is changed, the catalytic active site of PTP domain is exposed, and the inhibition of the catalytic active site is eliminated, so that the signaling cascade reaction initiated by tyrosine phosphorylation is initiated.
SHP2 is present in the cytoplasm and transduces signals from multiple receptor-tyrosine kinases in cells, and is therefore involved in a cascade of many oncogenic signals (e.g., RAS-ERK, PI3K-AKT, JAK-STAT). SHP2 has been reported to bind RAS and undergo a dephosphorylation reaction, enhancing the binding force between RAS-RAF, thereby activating downstream RAS/ERK/MAPK proliferation signaling pathways (Bunda, s., equivalent. nat. Commun.6, 8859.). Additionally, SHP2 has been reported in the literature (Li, J., et al. cancer Res.75,508-518.) to be involved in the programmed T cell death/checkpoint pathway (PD-L1/PD-1). Activated PD-1 recruits SHP2, dephosphorylates co-stimulatory receptor CD28, inhibits T cell function, and promotes immune escape. In view of the recent clinical success of anti-PD-L1/PD-1 therapy, there has been great interest in the development of small molecule SHP2 inhibitors for cancer immunotherapy.
Some SHP2 inhibitors have been reported (see, e.g., WO2015107493a1), but there remains a need in the art for new SHP2 inhibitors, particularly SHP2 inhibitors with high activity and other advantageous properties.
Disclosure of Invention
In one aspect, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof:
Figure BDA0002253496460000021
wherein:
R1selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -OC1-6Alkyl radical, C1-6Alkyl and halo C1-6An alkyl group;
l is selected from the group consisting of a direct bond, -NH-, -CH2-、-CH2CH2-, -CH ═ CH-and-C (═ CH)2)-;
Or L and R1Together with the atoms to which they are attached form a 5-10 membered hydrocarbon ring, a 5-10 membered heterocyclic ring, a 5-10 membered heteroaromatic ring or C6-10Aromatic rings, wherein the hydrocarbon, heterocyclic, heteroaromatic and aromatic rings are each optionally substituted with one or more groups selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -O, -OH, -OC1-6Alkyl, halo C1-6Alkyl and C1-6Alkyl substituent substitution;
x is selected from the group consisting of a direct bond, -S-, -O-, -NR6-、-C(R6)2-and-C (═ CH)2)-;
W1、W2And W3Each independently selected from CH and N;
ring A is selected from a 4-10 membered heterocyclic ring and a 4-10 membered hydrocarbon ring;
R2each occurrence is independently selected from H, halogen, -OH, -OC1-6Alkyl, -CN, C1-6Alkyl radical, C3-6Cycloalkyl radical, C2-6Alkenyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)23-to 10-membered heterocyclic group, C6-10Aryl and 5-10 membered heteroaryl, wherein said alkyl, cycloalkyl, alkenyl, heterocyclyl, aryl and heteroaryl are each optionally substituted with one or more substituents selected from the group consisting of halogen, ═ O, -OH, -OC1-6Alkyl, -CN, C1-6Alkyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl) and-N (C)1-6Alkyl radical)2Substituted with the substituent(s);
or 2R in adjacent positions2Together with the atoms to which they are attached form a 5-10 membered hydrocarbon ring, a 5-10 membered heterocyclic ring, a 5-6 membered heteroaromatic ring or a benzene ring, wherein the hydrocarbon ring, heterocyclic ring, heteroaromatic ring and benzene ring are each optionally substituted with one or more groups selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -O, -OH, -OC1-6Alkyl, halo C1-6Alkyl and C1-6Substitution of alkyl groupsSubstituted by radicals;
R3selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -OH, -COOH, -C (O) OC1-6Alkyl, -C (O) NH2、-OC1-6Alkyl and C1-6Alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, -OH, ═ O, -OC1-6Alkyl, -CN, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl) and-N (C)1-6Alkyl radical)2Substituted with the substituent(s);
R4each occurrence is independently selected from H, halogen, ═ O, -OH, -OC1-6Alkyl, -CN, C1-6Alkyl radical, C3-6Cycloalkyl radical, C2-6Alkenyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl) and-N (C)1-6Alkyl radical)2
R5aAnd R5bAre substituents on the same carbon atom and are each independently selected from H, halogen, -OH, -OC1-6Alkyl, -C1-6alkylene-OH, -CN, halo-C1-6Alkyl radical, C1-6Alkyl radical, C3-6Cycloalkyl radical, C2-6Alkenyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2and-C1-6alkylene-NH2(ii) a Or R5aAnd R5bTogether with the atoms to which they are attached form a 3-8 membered hydrocarbon ring or a 3-8 membered heterocyclic ring, wherein the hydrocarbon ring and heterocyclic ring are each optionally substituted with one or more substituents selected from H, halogen, ═ O, -OH, -OC1-6Alkyl, -C1-6alkylene-OH, -CN, halo-C1-6Alkyl radical, C1-6Alkyl radical, C3-6Cycloalkyl radical, C2-6Alkenyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2and-C1-6alkylene-NH2Substituted with the substituent(s);
R6is H or C1-6An alkyl group;
or, any one of R2And one R6Together with the atoms to which they are attached form a 5-10 membered hydrocarbon ring, a 5-10 membered heterocyclic ring, a 5-6 membered heteroaromatic ring or a benzene ring, wherein the hydrocarbon ring, heterocyclic ring, heteroaromatic ring and benzene ring are each optionally substituted with one or more groups selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -O, -OH, -OC1-6Alkyl, halo C1-6Alkyl and C1-6Alkyl substituent substitution;
g is 0, 1 or 2;
m is 0, 1,2, 3 or 4;
n is 0, 1,2, 3 or 4.
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, and one or more pharmaceutically acceptable carriers.
In a further aspect, the present invention provides the use of a compound of formula (I) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the invention, in the manufacture of a medicament for the prevention or treatment of a SHP2 phosphatase related disease.
In a further aspect, the present invention provides a method for preventing or treating a SHP2 phosphatase-related disease, the method comprising administering a compound of formula (I) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention, to a subject in need thereof.
In another aspect, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention, for use in the prevention or treatment of a SHP2 phosphatase related disease.
In one embodiment, the SHP2 phosphatase-associated disease is a disease that is sensitive to or responsive to SHP2 phosphatase inhibition. In further embodiments, the SHP2 phosphatase-related disease is a neoplastic disorder, including but not limited to solid and hematological malignancies.
In another aspect, the present invention further provides a compound of formula (I) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the invention, for use in a method of preventing or treating a SHP2 phosphatase related disease in combination with additional therapeutic methods, including but not limited to: radiotherapy, chemotherapy, immunotherapy, or a combination thereof.
Detailed Description
Compounds of the invention
In one aspect, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof:
Figure BDA0002253496460000041
wherein:
R1selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -OC1-6Alkyl radical, C1-6Alkyl and halo C1-6An alkyl group;
l is selected from the group consisting of a direct bond, -NH-, -CH2-、-CH2CH2-, -CH ═ CH-and-C (═ CH)2)-;
Or L and R1Together with the atoms to which they are attached form a 5-10 membered hydrocarbon ring, a 5-10 membered heterocyclic ring, a 5-10 membered heteroaromatic ring or C6-10Aromatic rings, wherein the hydrocarbon ring, heterocyclic ring, heteroaromatic ring and aromatic ring are each optionally substituted with oneOr more are selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -O, -OH, -OC1-6Alkyl, halo C1-6Alkyl and C1-6Alkyl substituent substitution;
x is selected from the group consisting of a direct bond, -S-, -O-, -NR6-、-C(R6)2-and-C (═ CH)2)-;
W1、W2And W3Each independently selected from CH and N;
ring A is selected from a 4-10 membered heterocyclic ring and a 4-10 membered hydrocarbon ring;
R2each occurrence is independently selected from H, halogen, -OH, -OC1-6Alkyl, -CN, C1-6Alkyl radical, C3-6Cycloalkyl radical, C2-6Alkenyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)23-to 10-membered heterocyclic group, C6-10Aryl and 5-10 membered heteroaryl, wherein said alkyl, cycloalkyl, alkenyl, heterocyclyl, aryl and heteroaryl are each optionally substituted with one or more substituents selected from the group consisting of halogen, ═ O, -OH, -OC1-6Alkyl, -CN, C1-6Alkyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl) and-N (C)1-6Alkyl radical)2Substituted with the substituent(s);
or 2R in adjacent positions2Together with the atoms to which they are attached form a 5-10 membered hydrocarbon ring, a 5-10 membered heterocyclic ring, a 5-6 membered heteroaromatic ring or a benzene ring, wherein the hydrocarbon ring, heterocyclic ring, heteroaromatic ring and benzene ring are each optionally substituted with one or more groups selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -O, -OH, -OC1-6Alkyl, halo C1-6Alkyl and C1-6Alkyl substituent substitution;
R3selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -OH, -COOH, -C (O) OC1-6Alkyl, -C (O) NH2、-OC1-6Alkyl and C1-6Alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, -OH, ═ O, -OC1-6Alkyl, -CN, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl) and-N (C)1-6Alkyl radical)2Substituted with the substituent(s);
R4each occurrence is independently selected from H, halogen, ═ O, -OH, -OC1-6Alkyl, -CN, C1-6Alkyl radical, C3-6Cycloalkyl radical, C2-6Alkenyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl) and-N (C)1-6Alkyl radical)2
R5aAnd R5bAre substituents on the same carbon atom and are each independently selected from H, halogen, -OH, -OC1-6Alkyl, -C1-6alkylene-OH, -CN, halo-C1-6Alkyl radical, C1-6Alkyl radical, C3-6Cycloalkyl radical, C2-6Alkenyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2and-C1-6alkylene-NH2
Or R5aAnd R5bTogether with the atoms to which they are attached form a 3-8 membered hydrocarbon ring or a 3-8 membered heterocyclic ring, wherein the hydrocarbon ring and heterocyclic ring are each optionally substituted with one or more substituents selected from H, halogen, ═ O, -OH, -OC1-6Alkyl, -C1-6alkylene-OH, -CN, halo-C1-6Alkyl radical, C1-6Alkyl radical, C3-6Cycloalkyl radical, C2-6Alkenyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2and-C1-6alkylene-NH2Substituted with the substituent(s);
R6is H or C1-6An alkyl group;
or, any one of R2And one R6Together with the atoms to which they are attached form a 5-10 membered hydrocarbon ring, a 5-10 membered heterocyclic ring, a 5-6 membered heteroaromatic ring or a benzene ring, wherein the hydrocarbon ring, heterocyclic ring, heteroaromatic ring and benzene ringEach optionally substituted by one or more groups selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -O, -OH, -OC1-6Alkyl, halo C1-6Alkyl and C1-6Alkyl substituent substitution;
g is 0, 1 or 2;
m is 0, 1,2, 3 or 4;
n is 0, 1,2, 3 or 4.
In some embodiments, R1Selected from H, -NH2、-OC1-3Alkyl, halo C1-3Alkyl and C1-3An alkyl group. In a preferred embodiment, R1Selected from H, -NH2and-CH3
In some embodiments, L is selected from the group consisting of a direct bond, -NH-, and-CH2-. In a preferred embodiment, L is selected from the group consisting of a direct bond and-CH2-. In a more preferred embodiment, L is selected from a direct bond.
In some embodiments, L and R1Together with the atoms to which they are attached form a 5-6 membered heterocyclic ring or a 5-6 membered heteroaromatic ring, wherein each of the heterocyclic and heteroaromatic rings is optionally substituted with one or more groups selected from H, -NH2F, Cl, -CN, -O, -OH, halogeno-C1-3Alkyl and C1-3Alkyl substituent substitution; in a preferred embodiment, L and R1Together with the atoms to which they are attached form a 5-6 membered heteroaromatic ring optionally substituted with one or more groups selected from H, -NH2F, Cl, -CN, -O, -OH, halogeno-C1-3Alkyl and C1-3Alkyl substituents. In another preferred embodiment, L and R1Together with the atoms to which they are attached form a pyrazole, pyrrole, pyrroline, imidazole, pyrazine or pyridine ring, wherein each of said rings is optionally substituted by one or more groups selected from H, -NH2F, Cl, -CN, -O, -OH, halogeno-C1-3Alkyl and C1-3Alkyl substituents. In a still further preferred embodiment of the present invention,
Figure BDA0002253496460000051
moieties are selected from the group consisting of:
Figure BDA0002253496460000052
Figure BDA0002253496460000053
in a still further preferred embodiment of the present invention,
Figure BDA0002253496460000061
moieties are selected from the group consisting of:
Figure BDA0002253496460000062
Figure BDA0002253496460000063
in a still further preferred embodiment of the present invention,
Figure BDA0002253496460000064
part is selected from
Figure BDA0002253496460000065
And
Figure BDA0002253496460000066
in some embodiments, X is selected from the group consisting of a direct bond, -S-, -O-, and-C (═ CH)2) -; in some embodiments, X is selected from the group consisting of a direct bond, -S-, and-O-; in a preferred embodiment, X is selected from the group consisting of a direct bond and-S-.
In some embodiments, W1Is CH, W2And W3Each independently selected from CH and N. In a preferred embodiment, W1、W2And W3Are both CH.
In some embodiments, ring a is selected from a 5-6 membered heterocyclic ring and a 5-6 membered hydrocarbon ring; in a preferred embodiment, ring a is selected from a 6-membered nitrogen-containing heterocyclic ring and a 6-membered hydrocarbon ring; in a further preferred embodiment, ring a is selected from the group consisting of a piperidine ring and a cyclohexene ring; in a further preferred embodiment, ring a is selected from the piperidine rings.
In some implementationsIn the scheme, R2Each occurrence is independently selected from H, halogen, -OH, -OC1-6 alkyl, -CN, C1-3 alkyl, halo C1-3 alkyl and-NH2(ii) a In a preferred embodiment, R2Each occurrence is independently selected from H, F, Cl, Br, -OH, -OCH3、-CH3、-CH2CH3、-CF3、-CHF2and-NH2For example, each independently selected from H, F, Cl, -OH, -CH3、-CH2CH3、-CF3、-CHF2and-NH2(ii) a In a further preferred embodiment, R2Each occurrence independently selected from F, Cl, Br, -OCH3、-CH3、-CF3and-NH2(ii) a In a further preferred embodiment, R2Each occurrence independently selected from F, Cl, -CH3、-CF3and-NH2(ii) a In a still further preferred embodiment, R2Each occurrence independently selected from Br, Cl and-NH2
In some embodiments of the present invention, the substrate is,
Figure BDA0002253496460000067
a group selected from: 2, 3-dichlorophenyl, 2-chloro-3-aminophenyl, 2- (trifluoromethyl) pyridin-3-yl, 2-amino-3-chloropyridin-4-yl, 2, 3-dichloropyridin-4-yl, 2-bromo-3-chloropyridin-4-yl, 2-bromo-3-fluoropyridin-4-yl, 2, 6-dichloropyridin-4-yl, 2, 3-difluorophenyl, 3, 4-difluorophenyl, 2-methylpyridin-4-yl, 2-chloropyridin-4-yl, 3-chloropyridin-4-yl, and 2-methoxy-3-chloropyridin-4-yl. In a preferred embodiment of the process according to the invention,
Figure BDA0002253496460000068
a group selected from: 2, 3-dichlorophenyl, 2-chloro-3-aminophenyl and 2, 3-dichloropyridin-4-yl.
In some embodiments, 2R in adjacent positions2Together with the atoms to which they are attached form a 5-7 membered hydrocarbon ring or a 5-7 membered heterocyclic ring, wherein the hydrocarbon ring and heterocyclic ring are each optionally mono-substitutedOne or more selected from H, -NH2、F、Cl、-CN、=O、-OH、-OC1-3Alkyl, halo C1-3Alkyl and C1-3Alkyl substituent substitution; in a preferred embodiment, 2R in adjacent positions2Together with the atoms to which they are attached form a 5-6 membered hydrocarbon ring or a 5-6 membered heterocyclic ring, wherein the hydrocarbon ring and heterocyclic ring are each optionally substituted with one or more groups selected from H, -NH2、F、Cl、=O、-OH、-OCH3and-CH3Substituted with the substituent(s); in a preferred embodiment, 2R in adjacent positions2Together with the atoms to which they are attached form a ring selected from cyclopentene, dioxole and pyrroline, wherein each of said rings is optionally substituted with one or more groups selected from H, -NH2、F、Cl、=O、-OH、-OCH3and-CH3Substituted with the substituent(s); in a more preferred embodiment, the 2R are relative to the point of attachment to X2Respectively in ortho-and meta-positions. In a more preferred embodiment of the process according to the invention,
Figure BDA0002253496460000071
a group selected from:
Figure BDA0002253496460000072
in some embodiments, when X is-NR6-or-C (R)6)2When in the ortho position to the point of attachment of X, an R2And one R6Together with the atoms to which they are attached form a 5-7 membered hydrocarbon ring or a 5-7 membered heterocyclic ring, wherein the hydrocarbon ring and heterocyclic ring are each optionally substituted with one or more groups selected from H, -NH2、F、Cl、-CN、=O、-OH、-OC1-3Alkyl, halo C1-3Alkyl and C1-3Alkyl substituent substitution; in a preferred embodiment, when X is-NR6-or-C (R)6)2When in the ortho position to the point of attachment of X, an R2And one R6Together with the atoms to which they are attached form a 5-6 membered hydrocarbon ring or a 5-6 membered heterocyclic ring, wherein the hydrocarbon ring and heterocyclic ring are each optionally substituted with one or more groups selected from H, -NH2、F、Cl、=O、-OH、-OCH3and-CH3Substituted with the substituent(s); in a preferred embodiment, when X is-NR6-or-C (R)6)2When in the ortho position to the point of attachment of X, an R2And one R6Together with the atoms to which they are attached form a ring selected from cyclohexene and tetrahydropyridine, wherein each of said rings is optionally substituted by one or more groups selected from H, -NH2、F、Cl、-CN、=O、-OH、-OC1-3Alkyl, halo C1-3Alkyl and C1-3Alkyl substituents. In a more preferred embodiment, when X is-NR6-or-C (R)6)2When the pressure in the air is higher than the preset pressure,
Figure BDA0002253496460000073
selected from the group consisting of:
Figure BDA0002253496460000074
in some embodiments, R3Selected from H, -NH2Halogen, -CN, -OH, C1-4Alkyl, -COOH and-C (O) OC1-4Alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ═ O, -OH, -S (═ O)g-(C1-3Alkyl) and-NH2Substituted with the substituent(s); in a preferred embodiment, R3Selected from H, -NH2、F、Cl、-CN、-OH、-CH3、-CF3、-CH2CH3、-CH2F、-CHF2、-CH2OH、-CH2CH2OH、-CH2NH2、-CH2CH2NH2-COOH and-C (O) OEt; in a further preferred embodiment, R3Selected from H, F, -CN, -COOH, -CH2OH、-CH2F、-CHF2and-CH2OH; in a further preferred embodiment, R3Selected from H, F and-CH2OH。
In some embodiments, R4Each occurrence is independently selected from H, F, Cl, ═ O, -OH, -OC1-3Alkyl, -CN, C1-3Alkyl and-NH2(ii) a In thatIn a preferred embodiment, R4Each occurrence is independently selected from H, F, Cl, -OH, -CH3and-NH2(ii) a In a further preferred embodiment, R4Each occurrence independently selected from H and-CH3(ii) a In a further preferred embodiment, R4Is H.
In some embodiments, R5aAnd R5bEach independently selected from H, C1-3Alkyl, -NH2、-C1-3alkylene-OH and-C1-3alkylene-NH2(ii) a Or R5aAnd R5bTogether with the atoms to which they are attached form a 5-6 membered hydrocarbon ring or a 5-6 membered heterocyclic ring, wherein the hydrocarbon ring and heterocyclic ring are each optionally substituted with one or more groups selected from H, F, Cl, C1-3Alkyl, ═ O, -NH2、-OH、-C1-3alkylene-OH and-C1-3alkylene-NH2Substituted with the substituent(s); in a preferred embodiment, R5aAnd R5bEach independently selected from H, -CH3、-CH2CH3、-NH2and-CH2NH2(ii) a Or R5aAnd R5bTogether with the atoms to which they are attached form a cyclopentane, an oxolane or a dihydrooxazole, wherein each of the cyclopentane, oxolane or dihydrooxazole is optionally substituted with one or more groups selected from-CH3and-NH2Substituted with the substituent(s); in a further preferred embodiment, R5aAnd R5bEach independently selected from-CH3and-NH2(ii) a Or R5aAnd R5bTogether with the atoms to which they are attached form a cyclopentane, an oxolane or a dihydrooxazole, wherein each of the cyclopentane, oxolane or dihydrooxazole is simultaneously substituted with a-CH3and-NH2Substituted with the substituent(s); in a further preferred embodiment, R5ais-CH3,R5bis-NH2(ii) a Or R5aAnd R5bTogether with the atoms to which they are attached form a co-substituted-CH3and-NH2Substituted oxolanes, preferably R5aAnd R5bTogether with the atoms to which they are attached form a group
Figure BDA0002253496460000081
In some embodiments, g is 0 or 2.
In some embodiments, m is 0, 1 or 2; in a preferred embodiment, m is 0.
In some embodiments, n is 0, 1 or 2; in a preferred embodiment, n is 1 or 2.
In some embodiments, the present invention provides a compound of formula (II) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof:
Figure BDA0002253496460000082
wherein R is1、R2、R3、R4、R5a、R5b、W1、W2、W3L, X, A, m and n are as defined above.
In some embodiments, the compounds of formula (I) of the present invention have the structure of formula (III) or formula (IV):
Figure BDA0002253496460000083
wherein the content of the first and second substances,
Figure BDA0002253496460000084
represents a single bond or a double bond; u is selected from N and C; when in use
Figure BDA0002253496460000085
Is a single bond, U is N, when
Figure BDA0002253496460000086
Is a double bond, U is C; ring B is selected from a 5-6 membered heterocyclic ring or a 5-6 membered heteroaromatic ring, wherein each of said heterocyclic and heteroaromatic rings is optionally substituted with one or more groups selected from H, -NH2、F、Cl、-CN、=O、-OH, halo C1-3Alkyl and C1-3Alkyl substituent substitution; in a preferred embodiment, ring B is selected from a 5-6 membered heteroaromatic ring. In a preferred embodiment, ring B is selected from a pyrazole ring, a pyrrole ring, a pyrroline ring, an imidazole ring, a pyrazine ring or a pyridine ring; the remaining groups are as defined above. In a preferred embodiment of the process according to the invention,
Figure BDA0002253496460000093
is a single bond, and U is N.
In a preferred embodiment, the compounds of formula (I) of the present invention have the structure of formula (V), formula (VI) or formula (VII):
Figure BDA0002253496460000091
wherein R is2aAnd R2bAre defined as each2Same as R2aAnd R2bMay be the same or different; q1、Q2And Q3Each is independently selected from N, NH, C and CH, and is not simultaneously C or CH; the remaining groups are as defined above.
It will be understood by those skilled in the art that the present invention encompasses compounds resulting from any combination of the various embodiments. Embodiments resulting from the combination of features from one embodiment or preferred features with features from another embodiment or preferred features are also included within the scope of the present invention.
In a preferred embodiment, the present invention provides a compound or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, wherein said compound is selected from the group consisting of:
Figure BDA0002253496460000092
Figure BDA0002253496460000101
Figure BDA0002253496460000111
Figure BDA0002253496460000121
in another aspect, the present invention provides a pharmaceutical composition comprising a compound of formulae (I) - (VII) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, and one or more pharmaceutically acceptable carriers.
In a further aspect, the present invention provides the use of a compound of formulae (I) - (VII) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention, in the manufacture of a medicament for the prevention or treatment of a SHP2 phosphatase related disease.
In a further aspect, the present invention provides a method for preventing or treating a SHP2 phosphatase-related disease, the method comprising administering a compound of formula (I) - (VII) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention, to a subject in need thereof.
In another aspect, the present invention provides a compound of formulae (I) - (VII) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention, for use in the prevention or treatment of a SHP2 phosphatase related disease.
In one embodiment, the SHP2 phosphatase-associated disease is a disease that is sensitive to or responsive to SHP2 phosphatase inhibition. In further embodiments, the SHP2 phosphatase-related disease is a neoplastic disorder, including but not limited to solid and hematological malignancies.
In another aspect, the present invention further provides a compound of formulae (I) - (VII) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a pharmaceutical composition of the invention, for use in a method of preventing or treating a SHP2 phosphatase-associated disease in combination with additional therapeutic methods, including but not limited to: radiotherapy, chemotherapy, immunotherapy, or a combination thereof.
The preparation method of the invention
Yet another aspect of the present invention relates to a process for the preparation of a compound of the present invention, said process comprising:
1. reacting a compound of formula S-1 with a compound of formula S-2 to produce a compound of formula IM-1;
Figure BDA0002253496460000131
2. reacting a compound of formula IM-1 with a compound of formula S-3 to form a compound of formula IM-2;
Figure BDA0002253496460000132
3. deprotecting a compound of formula IM-2 to produce a compound of formula (I);
Figure BDA0002253496460000133
wherein LG is1And LG2Each independently represents a halogen leaving group, or C optionally substituted by halogen1-6An alkylsulfonate leaving group (e.g., a trifluoromethanesulfonate leaving group);
Rcand RdEach independently represents H or a leaving group;
PG1protecting groups representing hydroxy groups (e.g. methyl, tert-butyldimethylSilyl, triisopropylsilyl, benzyl, and methoxymethyl);
the remaining groups are as defined above.
In a preferred embodiment, LG is used1And LG2Each independently represents a halogen such as iodine or chlorine.
In a preferred embodiment, RcAnd RdIndependently selected from H, halogen, boronic acid groups, boronic ester groups, substituted silicon groups, substituted metal groups or C optionally substituted by halogen1-6An alkylsulfonate group. In a more preferred embodiment, RcIs a boronic acid group or a boronic ester group, RdIs H, boric acid group or boric acid ester group.
In step 1, the reaction may be carried out in the presence of a metal catalyst. In a preferred embodiment, the metal catalyst is a metal palladium catalyst, such as tetrakistriphenylphosphine palladium, palladium acetate, tris (dibenzylideneacetone) dipalladium, 1' -bis (diphenylphosphino) dichloroferrocenepalladium, 1, 2-bis-diphenylphosphinoethylpalladium chloride, bis (triphenylphosphine) palladium dichloride, and the like.
In step 2, the reaction may be carried out in the presence of a base and/or a metal catalyst. In a preferred embodiment, the base is an inorganic base, such as potassium phosphate. In another preferred embodiment, the metal catalyst is a metal palladium catalyst, such as tetrakistriphenylphosphine palladium, palladium acetate, tris (dibenzylideneacetone) dipalladium, 1' -bis (diphenylphosphino) dichloroferrocenepalladium, 1, 2-bis-diphenylphosphinoethylpalladium chloride, bis (triphenylphosphine) palladium dichloride, and the like.
In step 3, the reaction may be carried out under acidic or catalytic hydrogenolysis conditions. In a preferred embodiment, the acid is an organic acid, preferably trifluoroacetic acid. In another preferred embodiment, the metal catalyst is palladium on carbon.
The starting materials for the preparation process of the present invention may be obtained from commercial sources or may be prepared according to known methods.
It will be appreciated by those skilled in the art that one or more of the steps in the above routes may be omitted depending on the desired resulting product structure. The order of the reaction steps may also be appropriately adjusted and the protection/deprotection reaction steps may be added or omitted as necessary by those skilled in the art.
The compounds of the formula (II), formula (III), formula (IV), formula (V), formula (VI) and formula (VII) of the present invention and the like can be synthesized by a similar method with reference to the above-mentioned embodiments.
Pharmaceutical compositions, formulations and kits
The present invention also provides a pharmaceutical composition comprising a compound of formulae (I) - (VII) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, and one or more pharmaceutically acceptable carriers, and optionally further comprising one or more second therapeutic agents for treating a SHP2 enzyme-related disorder.
It is a further object of the present invention to provide a process for preparing a pharmaceutical composition of the present invention, said process comprising combining a compound of formula (I) - (VII) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a mixture thereof, with one or more pharmaceutically acceptable carriers. The method may further comprise admixing one or more second therapeutic agents for treating a SHP2 enzyme-related disorder.
Pharmaceutically acceptable carriers that may be employed in the pharmaceutical compositions of the present invention include, but are not limited to, sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is an exemplary carrier when the pharmaceutical composition is administered intravenously. Physiological saline and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Pharmaceutically acceptable carriers include pharmaceutical excipients. Suitable pharmaceutical excipients include, but are not limited to, starch, glucose, lactose, sucrose, gelatin, maltose, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol and the like. The composition may also optionally contain minor amounts of wetting agents, emulsifying agents, lubricating agents, stabilizing agents, or pH buffering agents, and the like. Oral formulations may contain standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. Examples of suitable pharmaceutically acceptable carriers are described in Remington's Pharmaceutical Sciences (2005).
The pharmaceutical composition may be administered in any form as long as it achieves prevention, alleviation, prevention or cure of symptoms in a human or animal patient. For example, various suitable dosage forms can be prepared according to the administration route.
When administered orally, the pharmaceutical composition may be formulated into any orally acceptable dosage form including, but not limited to, tablets, capsules, granules, pills, syrups, oral solutions, oral suspensions, oral emulsions, and the like. Among these, the tablets use carriers such as lactose, corn starch, etc., and can be compressed into tablets optionally in combination with: binding agents, for example acacia or gelatin; disintegrants, such as croscarmellose sodium, crospovidone; lubricants, such as magnesium stearate and the like. Diluents for capsules include lactose, dried corn starch, and the like. Oral suspensions are generally prepared by mixing the active ingredient with suitable emulsifying and suspending agents. Optionally, some sweetener, aromatic or colorant may be added into the above oral preparation.
When administered transdermally or topically, the pharmaceutical compositions may be formulated in the form of suitable ointments, lotions or liniments in which the active ingredient is suspended or dissolved in one or more carriers. Carriers that may be used in ointment formulations include, but are not limited to: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyethylene oxide, polypropylene oxide, emulsifying wax and water; lotions or liniments carriers that may be used include, but are not limited to: mineral oil, sorbitan monostearate, tween 60, 2-octyldodecanol, benzyl alcohol and water.
The pharmaceutical composition can also be used in the form of injection, including injection, sterile powder for injection and concentrated solution for injection. Among the carriers and solvents that may be employed are water, ringer's solution and isotonic sodium chloride solution. In addition, the sterilized fixed oil may also be employed as a solvent or suspending medium, such as a monoglyceride or diglyceride.
In another embodiment, administration of a compound or pharmaceutical composition of the invention may be combined with additional methods of treatment. The additional treatment methods may be selected from, but are not limited to: radiotherapy, chemotherapy, immunotherapy, or a combination thereof. The compounds or pharmaceutical compositions of the present invention may be administered before, during or after the administration of the additional method of treatment. The additional therapeutic methods may be performed simultaneously with, immediately before or after, or at intervals, the administration of the compounds or pharmaceutical compositions of the invention, the mode and sequence of administration being selected and adjusted to the particular therapeutic situation.
Yet another aspect of the present invention relates to a pharmaceutical formulation comprising a compound of formula (I) - (VII), a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, or a mixture thereof, as an active ingredient, or a pharmaceutical composition of the present invention. In some embodiments, the formulation is in the form of a solid formulation, a semi-solid formulation, a liquid formulation, or a gaseous formulation.
It is a further object of the invention to provide an article of manufacture, for example in the form of a kit. Articles of manufacture as used herein are intended to include, but are not limited to, kits and packages. The article of the present invention comprises: (a) a first container; (b) a pharmaceutical composition in a first container, wherein the composition comprises: a first therapeutic agent comprising: a compound of formula (I) - (VII) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, or a mixture thereof; and (c) package insert indicating that the pharmaceutical composition can be used for the treatment of a neoplastic disorder (as defined hereinbefore). In another embodiment, the package insert indicates that the pharmaceutical composition can be used in combination with a second therapeutic agent to treat a neoplastic disorder. The article may further comprise: (d) a second container, wherein components (a) and (b) are located within the second container and component (c) is located within or outside the second container. Being located within the first and second containers means that each container retains the item within its boundaries.
The first container is a container for holding a pharmaceutical composition. The container may be used for preparation, storage, transport and/or individual/bulk sale. The first container is intended to encompass a bottle, jar, vial, flask, syringe, tube (e.g., for a cream preparation), or any other container used to prepare, contain, store, or dispense a pharmaceutical product.
The second container is a container for holding the first container and optionally packaging instructions. Examples of the second container include, but are not limited to, a box (e.g., a carton or a plastic box), a box, a carton, a bag (e.g., a paper or plastic bag), a pouch, and a blister pack. The package insert may be physically adhered to the exterior of the first container via a tie, glue, staple, or other adhesive means, or it may be placed inside the second container without any physical means of adhering to the first container. Alternatively, the package insert is located outside of the second container. When located outside of the second container, it is preferred that the package insert is physically adhered via a tie, glue, staple or other means of adhesion. Alternatively, it may abut or contact the exterior of the second container without physical adhesion.
The package insert is a trademark, label, logo, etc. that lists information related to the pharmaceutical composition located in the first container. The listed information is typically determined by a regulatory agency (e.g., the U.S. food and drug administration) that governs the area in which the article is to be sold. Preferably the package insert specifically lists the indications for which the pharmaceutical composition is approved for use. The package insert may be made of any material from which information contained therein or thereon can be read. Preferably, the package insert is a printable material (e.g., paper, plastic, cardboard, foil, adhesive paper or plastic, etc.) on which the desired information can be formed (e.g., printed or applied).
Methods of treatment and uses
Another object of the present invention is to provide a method for preventing or treating a SHP2 enzyme-related disease, which comprises administering to a subject in need thereof an effective amount of a compound of formulae (I) - (VII) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite or prodrug thereof, or a mixture thereof, or a pharmaceutical composition of the present invention.
According to one embodiment of the invention, the SHP2 enzyme-related diseases that can be prevented or treated using the compounds of the present invention are diseases that are sensitive or responsive to the inhibition of the SHP2 enzyme. In a further embodiment, the SHP2 enzyme-related disease is a neoplastic disorder, including but not limited to solid and hematologic malignancies. In further embodiments, the neoplastic disorder includes, but is not limited to, breast, colorectal, colon, lung (including small cell lung, non-small cell lung, and bronchioloalveolar) and prostate cancer, as well as biliary tract, bone, bladder, head and neck, kidney, liver, gastrointestinal tissue, esophagus, ovary, pancreas, skin, testis, thyroid, uterus, cervix and vulva cancer, and leukemia (including Chronic Lymphocytic Leukemia (CLL), Acute Lymphocytic Leukemia (ALL), and Chronic Myelogenous Leukemia (CML)), multiple myeloma, and lymphoma. In a particular embodiment, the neoplastic disease is colon cancer or lung cancer.
In a preferred embodiment, the disease is cancer. In a further preferred embodiment, the compounds of the invention may be used in combination with chemoradiotherapy or immunotherapy for the prevention or treatment of cancer.
The dosing regimen may be adjusted to provide the best desired response. For example, when administered as an injection, a single bolus, bolus and/or continuous infusion may be administered, and the like. For example, several divided doses may be administered over time, or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is noted that dosage values may vary with the type and severity of the condition being alleviated, and may include single or multiple doses. Generally, the dosage of treatment will vary depending on considerations such as, for example: the age, sex and general health of the patient to be treated; the frequency of treatment and the nature of the desired effect; the degree of tissue damage; duration of symptoms; and other variables that can be adjusted by the individual physician. It is further understood that for any particular individual, the specific dosage regimen will be adjusted over time according to the individual need and the professional judgment of the person administering the composition or supervising the administration of the composition. The amount and regimen of administration of the pharmaceutical composition can be readily determined by one of ordinary skill in the clinical art. For example, the composition or compound of the present invention may be administered in divided doses 4 times per day to 1 time per 3 days, and the amount administered may be, for example, 0.01 to 1000 mg/time. The desired dose may be administered in one or more administrations to achieve the desired result. The pharmaceutical compositions according to the invention may also be provided in unit dosage form.
Advantageous effects
The invention provides a novel high-activity SHP2 inhibitor, which can realize at least one of the following technical effects:
(1) high inhibitory activity against SHP2 enzyme.
(2) Excellent physicochemical properties (e.g. solubility, physical and/or chemical stability).
(3) Excellent pharmacokinetic properties (e.g. good bioavailability, suitable half-life and duration of action).
(4) Excellent safety (lower toxicity and/or fewer side effects, wider therapeutic window), etc.
General terms and definitions
Unless defined otherwise below, all technical and scientific terms used herein are intended to have the same meaning as commonly understood by one of ordinary skill in the art. Reference to the techniques used herein is intended to refer to those techniques commonly understood in the art, including those variations of or alternatives to those techniques that would be apparent to those skilled in the art. While the following terms are believed to be well understood by those skilled in the art, the following definitions are set forth to better explain the present invention.
The terms "comprising," "including," "having," "containing," or "involving," and other variations thereof herein, are inclusive or open-ended and do not exclude additional unrecited elements or method steps. It will be understood by those skilled in the art that terms such as "including" and "comprising" encompass the meaning of "consisting of ….
The term "about" means within. + -. 10%, preferably within. + -. 5%, more preferably within. + -. 2% of the stated value.
Unless otherwise stated, concentrations are by weight and ratios (including percentages) are by mole.
The term "one or more" or similar expressions "at least one" may mean, for example, 1,2, 3,4, 5, 6, 7, 8, 9, 10 or more(s).
When the lower and upper limits of a range of values are disclosed, any value falling within the range and any included range is specifically disclosed. In particular, each range of values (in the form "about a to b", or equivalently, "about a-b") disclosed herein is to be understood as meaning each number and range encompassed within the broader range.
For example, the expression "C1-C6"is to be understood to cover any subrange therein as well as each point value, e.g. C2-C5、C3-C4、C1-C2、C1-C3、C1-C4、C1-C5Etc. and C1、C2、C3、C4、C5、C6And the like. For example, the expression "C3-C10"should also be understood in a similar manner, e.g. to cover any sub-ranges and point values comprised therein, e.g. C3-C9、C6-C9、C6-C8、C6-C7、C7-C10、C7-C9、C7-C8、C8-C9Etc. and C3、C4、C5、C6、C7、C8、C9、C10And the like. Also for example, the expression "3-10 membered" should be understood to encompass any subrange therein as well as each point value, e.g., 3-4, 3-5, 3-6, 3-7, 3-8, 3-9, 4-5, 4-6, 4-7, 4-8, 5-7, 5-8, 6-7, etc., as well as 3,4, 5, 6, 7, 8, 9, 10, etc. Also for example, the expression "5-10 elements" should be understood in a similar manner, e.g., to encompass any subranges and point values subsumed therein, e.g., 5-6 elements, 5-7 elements, 5-8 elements, 5-9 elements, 5-10 elements, 6-7 elements, 6-8 elements, 6-9 elements, 6-10 elements, 7-8 elements, etc., as well as 5, 6, 7, 8, 9, 10 elements, etc.
The term "alkyl", used herein alone or in combination with other groups, refers to a saturated straight or branched chain hydrocarbon group. As used herein, the term "C1-6Alkyl "refers to a saturated straight or branched chain hydrocarbon group having 1 to 6 carbon atoms (e.g., 1,2, 3,4, 5, or 6 carbon atoms). E.g. "C1-6Alkyl "may be methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, or n-hexyl, and the like.
As used herein, the term "alkylene" refers to a saturated, straight or branched chain, divalent hydrocarbon radical. For example, as used herein, the term "C1-6Alkylene "refers to a straight or branched chain divalent hydrocarbon radical saturated with 1 to 6 carbon atoms. Such as methylene, ethylene, propylene or butylene, and the like.
The term "cycloalkyl", as used herein alone or in combination with other groups, refers to a saturated non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (e.g., monocyclic, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl; or bicyclic, including spiro, fused or bridged systems (such as bicyclo [ 1.1.1)]Pentyl, bicyclo [2.2.1]Heptyl, bicyclo [3.2.1]Octyl or bicyclo [5.2.0]Nonyl, decalinyl, etc.). Cycloalkyl radicals including C3-10Cycloalkyl, preferably C3-6Cycloalkyl, more preferably C5-6A cycloalkyl group. For example, the term "C3-10CycloalkanesThe group "refers to cycloalkyl groups having 3 to 10 ring carbon atoms (e.g., 3,4, 5, 6, 7, 8, 9, or 10).
The term "hydrocarbon ring", as used herein alone or in combination with other groups, refers to a saturated or partially unsaturated (i.e., having one or more double and/or triple bonds within the ring, but not forming an aromatic ring system) monocyclic or polycyclic hydrocarbon ring having, for example, from 3 to 10 (e.g., from 4 to 10, from 5 to 10, from 6 to 10, preferably from 5 to 8, more preferably from 5 to 6) ring carbon atoms, including, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclohexenyl, and the like.
The term "halo" or "halogen" group, when used herein alone or in combination with other groups, denotes F, Cl, Br or I.
The term "haloalkyl", as used herein alone or in combination with other groups, refers to an alkyl group as described above wherein one or more hydrogen atoms are replaced by a halogen. For example, the term "halo C1-6Alkyl "refers to C optionally substituted with one or more (e.g., 1-3) halogens1-6An alkyl group. It will be understood by those skilled in the art that when there is more than one halogen substituent, the halogens may be the same or different and may be located on the same or different C atoms. Examples of haloalkyl are, for example, -CH2F、-CHF2、-CF3、-CCl3、-C2F5、-C2Cl5、-CH2CF3、-CH2Cl or-CH2CH2CF3And the like.
The term "alkenyl", as used herein alone or in combination with other groups, refers to a straight or branched chain hydrocarbon group having one or more carbon-carbon double bonds. For example, as used herein, the term "C2-6Alkenyl "means a straight or branched chain hydrocarbon group having 2 to 6 carbon atoms and one, two or three carbon-carbon double bonds, preferably C containing one carbon-carbon double bond2-6An alkenyl group. For example vinyl, 1-propenyl, 2-butenyl, 3-butenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 2-hexenyl-methyl-2-propenyl, 4-methyl-3-pentenyl and the like.
The term "heterocycle", used herein alone or in combination with other groups, refers to a monocyclic or bicyclic non-aromatic ring system (i.e., a 3-10, 4-10, 5-10, 6-10, 3-8, preferably 3-6, more preferably 5-6 membered heterocycle) having, for example, 3-10 (e.g., 4-10, 6-10, 3-8, 3-6 or 5-6) ring atoms, wherein at least one ring atom (e.g., 1,2 or 3) is a heteroatom selected from N, O and S, and the remaining ring atoms are C. The ring system may be saturated (which may also be understood as a corresponding "saturated heterocycloalkyl") or unsaturated (i.e., having one or more double and/or triple bonds within the ring). The term also covers the case where the C atom may be substituted (═ O) and/or the S atom on the ring may be substituted by 1 or 2(═ O). A heterocyclic group is a group derived from a heterocyclic ring by removing one hydrogen atom, and examples thereof include, but are not limited to: oxirane, thienylethyl, aziridine, azetidine, oxetane, thienylbutyl, tetrahydrofuranyl, tetrahydrothienyl, dioxolyl, pyrrolidinyl, pyrrolidinonyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl, tetrahydropyranyl, piperidinyl, morpholinyl, 1, 4-thioxanyl, 1, 4-dioxanyl, dithianyl, tetrahydropyridinyl, thiomorpholinyl, piperazinyl, trithianyl and the like.
The term "aromatic ring", used herein alone or in combination with other groups, refers to an all-carbon monocyclic or fused-ring polycyclic (e.g., bicyclic) aromatic ring having a conjugated pi-electron system. An aryl group is a group derived from an aromatic ring by the removal of one hydrogen atom. As used herein, the term "C6-10Aryl "refers to an aromatic radical derived from an aromatic ring containing 6 to 10 carbon atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, and the like.
The term "heteroaromatic ring", used herein alone or in combination with other groups, refers to an aromatic ring in which one or more (e.g., 1,2 or 3) ring atoms are heteroatoms selected from N, O and S, and the remaining ring atoms are C. Heteroaryl is a group derived from a heteroaromatic ring by removal of one hydrogen atom. Heteroaryl or heteroaromatic rings can be characterized by the number of ring atoms. For example, a 5-to 10-membered heteroaryl group may contain 5-to 10 ring atoms (e.g. 5, 6, 7, 8, 9 or 10), in particular 5, 6, 9, 10 ring atoms, and a 5-to 6-membered heteroaryl group may contain, for example, 5 or 6 ring atoms. And in each case, the heteroaryl or heteroaromatic ring may optionally be further benzo-fused. For example, examples of heteroaryl groups are thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, pyrazinyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl and the like, and benzo derivatives thereof; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, quinolinyl, isoquinolinyl, benzofuranyl, benzothienyl, indolyl, isoindolyl, and the like, and benzo derivatives thereof.
The term "hydroxy" denotes-OH.
The term "cyano" denotes-CN.
The term "nitro" denotes-NO2
The term "amino" denotes-NH2
The terms "substituted" and "substituted" mean that one or more (e.g., one, two, three, or four) hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency in the current situation is not exceeded and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
If a substituent is described as "optionally … substituted," the substituent may be (1) unsubstituted or (2) substituted. If an atom or group is described as optionally substituted with one or more of a list of substituents, one or more hydrogens on the atom or group may be replaced with an independently selected, optional substituent. If a substituent is described as "independently selected from" or "each independently is," each substituent is selected independently of the other. Thus, each substituent may be the same as or different from another (other) substituent. For example, a certain substituent or substitution position or different substituents or substitution positions haveR groups (such as but not limited to R) which may be the same or different symbols2、R3、Rh、Ri、RxAnd/or Ry) In the selection of (3), R's are independently selected from each other, and may be the same or different. The same is true with regard to the choice of values such as d, g, m, n.
Unless indicated, as used herein, the point of attachment of a substituent may be from any suitable position of the substituent.
When a bond of a substituent is shown through a bond connecting two atoms in a ring, then such substituent may be bonded to any ring atom in the substitutable ring.
The invention also includes all pharmaceutically acceptable isotopically-labeled compounds, which are identical to those of the present invention, except that one or more atoms are replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number prevailing in nature. Examples of isotopes suitable for inclusion in compounds of the invention include, but are not limited to, isotopes of hydrogen (e.g., deuterium (g), (b), (c), (d), (2H) Tritium (a)3H) ); isotopes of carbon (e.g. of13C and14C) (ii) a Isotopes of chlorine (e.g. of chlorine)37Cl); isotopes of iodine (e.g. of iodine)125I) (ii) a Isotopes of nitrogen (e.g. of13N and15n); isotopes of oxygen (e.g. of17O and18o); isotopes of phosphorus (e.g. of phosphorus)32P); and isotopes of sulfur (e.g. of34S)。
The term "stereoisomer" denotes an isomer formed as a result of at least one asymmetric center. In compounds having one or more (e.g., one, two, three, or four) asymmetric centers, they can give rise to racemic mixtures, single enantiomers, diastereomeric mixtures, and individual diastereomers. Certain individual molecules may also exist as geometric isomers (cis/trans). Similarly, the compounds of the invention may exist as mixtures of two or more structurally different forms (commonly referred to as tautomers) in rapid equilibrium. Representative examples of tautomers include keto-enol tautomers, phenol-keto tautomers, nitroso-oxime tautomers, imine-enamine tautomers, and the like. It is to be understood that the scope of the present invention encompasses all such isomers or mixtures thereof in any ratio (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%).
Solid (—), solid wedge shapes may be used herein
Figure BDA0002253496460000201
Or virtual wedge shape
Figure BDA0002253496460000202
Carbon-carbon bonds of the compounds of the invention are depicted. The use of a solid line to depict a bond to an asymmetric carbon atom is intended to indicate that all possible stereoisomers (e.g., particular enantiomers, racemic mixtures, etc.) at that carbon atom are included. The use of solid or dashed wedges to depict bonds to asymmetric carbon atoms is intended to indicate that the stereoisomers shown are present. When present in a racemic mixture, solid and dotted wedges are used to define the relative stereochemistry, not the absolute stereochemistry. Unless otherwise indicated, the compounds of the present invention may exist in the form of stereoisomers (which include cis and trans isomers, optical isomers (e.g., R and S enantiomers), diastereomers, geometric isomers, rotamers, conformers, atropisomers, and mixtures thereof). The compounds of the present invention may exhibit more than one type of isomerization and consist of mixtures thereof (e.g., racemic mixtures and diastereomeric pairs).
The invention also encompasses all possible crystalline forms or polymorphs of the compounds of the invention, which may be single polymorphs or mixtures of more than one polymorph in any ratio.
It will also be appreciated that certain compounds of the invention may exist in free form or, where appropriate, in the form of a pharmaceutically acceptable derivative thereof. In the present invention, pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable salts, esters, solvates, N-oxides, metabolites or prodrugs thereof, which upon administration to a patient in need thereof are capable of providing, directly or indirectly, a compound of formula (I) - (VII) or a metabolite thereof. Thus, when reference is made herein to "a compound of the invention," it is also intended to encompass the various derivative forms of the compounds described above.
Pharmaceutically acceptable salts of the compounds of the present invention include acid addition salts and base addition salts thereof. Suitable acid addition salts are formed from acids which form pharmaceutically acceptable salts. Examples include hydrochloride, acetate, aspartate, benzoate, bicarbonate/carbonate, glucoheptonate, gluconate, nitrate, orotate, palmitate and other similar salts. Suitable base addition salts are formed from bases which form pharmaceutically acceptable salts. Examples include aluminum salts, arginine salts, choline salts, magnesium salts, and other similar salts. For reviews of suitable salts see, for example, "Remington's Pharmaceutical Sciences", mack publishing Company, Easton, Pa., (2005); and "handbook of pharmaceutically acceptable salts: properties, Selection and application "(handbook of Pharmaceutical Salts: Properties, Selection, and Use), Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002). Methods for preparing pharmaceutically acceptable salts of the compounds of the present invention are known to those skilled in the art.
As used herein, the term "ester" means an ester derived from a compound described herein, including physiologically hydrolyzable esters (compounds of the invention that can hydrolyze under physiological conditions to release the free acid or alcohol form). The compounds of the invention may themselves also be esters.
The compounds of the invention may be present in the form of solvates, preferably hydrates, wherein the compounds of the invention comprise as structural element of the crystal lattice of the compound a polar solvent, such as in particular water, methanol or ethanol. The amount of polar solvent, particularly water, may be present in stoichiometric or non-stoichiometric proportions.
Those skilled in the art will appreciate that not all nitrogen-containing heterocycles are capable of forming N-oxides, since the available lone pair of electrons is required for oxidation to the oxide. One skilled in the art will recognize nitrogen-containing heterocycles that are capable of forming N-oxides. Those skilled in the art will also recognize that tertiary amines are capable of forming N-oxides. Synthetic methods for preparing N-oxides of heterocycles and tertiary amines are well known to those skilled in the art and include oxidation of heterocycles and tertiary amines with peroxy acids such as peracetic and m-chloroperbenzoic acid (mCPBA), hydrogen peroxide, alkyl hydroperoxides such as t-butyl hydroperoxide, sodium perborate, and dioxiranes (dioxiranes) such as dimethyldioxirane. These methods for preparing N-oxides have been widely described and reviewed in the literature, see for example: l.l.gilchrist, Comprehensive Organic Synthesis, vol.7, pp748-750(a.r.katritzky and a.j.boulton, eds., Academic Press); and G.W.H.Cheeseman and E.S.G.Werstuk, Advances in Heterocyclic Chemistry, vol.22, pp 390-.
Also included within the scope of the present invention are metabolites of the compounds of the present invention, i.e., substances formed in vivo upon administration of the compounds of the present invention. Metabolites of a compound can be identified by techniques well known in the art, and their activity can be characterized by assay methods. Such products may result, for example, from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymatic hydrolysis, etc. of the administered compound. Accordingly, the present invention includes metabolites of the compounds of the present invention, including compounds made by the process of contacting the compounds of the present invention with a mammal for a time sufficient to produce a metabolite thereof.
The present invention further includes within its scope prodrugs of the compounds of the present invention which are certain derivatives of the compounds of the present invention which may themselves have little or no pharmacological activity which, when administered into or onto the body, may be converted to the compounds of the present invention having the desired activity by, for example, hydrolytic cleavage. Typically such prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the desired therapeutically active compound. Additional information on the use of prodrugs can be found in "Pro-drugs as Novel Delivery Systems", volume 14, ACS Symposium Series (t.higuchi and v.stella). Prodrugs of the invention may be prepared, for example, by substituting certain moieties known to those skilled in the art as "pro-moieties" (e.g., "Design of Prodrugs", described in h. bundgaard (Elsevier, 1985)) for appropriate functional groups present in compounds of the invention.
The invention also encompasses compounds of the invention containing a protecting group. In any process for preparing the compounds of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned, thereby forming a chemically protected form of the compounds of the present invention. This may be achieved by conventional protecting Groups, such as those described in t.w.greene & p.g.m.wuts, Protective Groups in Organic Synthesis, John Wiley & Sons,2006, which references are incorporated herein by reference. The protecting group may be removed at a suitable subsequent stage using methods known in the art.
The invention also encompasses methods of making the compounds described herein. It is to be understood that the compounds of the present invention can be synthesized using the methods described herein as well as synthetic methods known in the art of synthetic organic chemistry or variations thereof as would be understood by one of skill in the art. Preferred methods include, but are not limited to, those described below. The reaction may be carried out in a solvent or solvent mixture suitable for the reagents and materials used and for effecting the conversion.
By "pharmaceutically acceptable carrier" in the context of the present invention is meant a diluent, adjuvant, excipient, or vehicle that is administered with the active ingredient and which is, within the scope of sound medical judgment, suitable for contact with the tissues of humans and/or other animals without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio. The terms "active ingredient," "therapeutic agent," "active substance," or "active agent" refer to a chemical entity that is effective in treating one or more symptoms of a target disorder or condition.
The term "effective amount" (e.g., "therapeutically effective amount" or "prophylactically effective amount") as used herein refers to an amount of active ingredient that will achieve the desired effect to some extent upon administration, e.g., to alleviate one or more symptoms of the treated condition or to prevent the occurrence of the condition or symptoms thereof.
As used herein, unless otherwise specified, the term "treating" or "treatment" means reversing, alleviating, inhibiting the progression of, or preventing the disorder or condition to which the term applies, or one or more symptoms of the disorder or condition.
SHP2 phosphatase-related diseases are diseases that are sensitive or responsive to SHP2 phosphatase inhibition, including, but not limited to, solid and hematological malignancies, such as breast, colorectal, colon, lung (including small cell lung, non-small cell lung, and bronchioloalveolar) and prostate cancer, as well as biliary, bone, bladder, head and neck, kidney, liver, gastrointestinal tissue, esophageal, ovarian, pancreatic, skin, testicular, thyroid, uterine, cervical, and vulvar cancers, and leukemias (including Chronic Lymphocytic Leukemia (CLL), Acute Lymphocytic Leukemia (ALL), and Chronic Myelogenous Leukemia (CML)), multiple myeloma, and lymphoma.
As used herein, "individual" includes a human or non-human animal. Exemplary human individuals include human individuals (referred to as patients) having a disease (e.g., a disease described herein) or normal individuals. "non-human animals" in the context of the present invention include all vertebrates, such as non-mammals (e.g., birds, amphibians, reptiles) and mammals, such as non-human primates, livestock and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
Detailed Description
Examples
The present invention will be described in more detail below with reference to examples and test examples, but these examples do not limit the scope of the present invention and can be modified without departing from the scope of the present invention.
Determination of MS using an agilent (esi) mass spectrometer, manufacturer: agilent, model: agilent 6120B.
Preparative high performance liquid chromatography was prepared using Shimadzu LC-8A preparative liquid chromatography (YMC, ODS, 250X 20mm column).
Purifying by thin layer chromatography using silica gel plate of GF 254 (0.4-0.5 nm) produced by cigarette bench.
The reaction is monitored by Thin Layer Chromatography (TLC) or LC-MS using a developer system including, but not limited to: the volume ratio of the solvent is adjusted according to different polarities of the compounds, or triethylamine and the like are added for adjustment.
The column chromatography generally uses Qingdao ocean silica gel with 200-300 meshes as a stationary phase. The eluent system includes but is not limited to dichloromethane and methanol system and n-hexane and ethyl acetate system, the volume ratio of the solvent is adjusted according to different polarities of the compounds, and a small amount of triethylamine and the like can be added for adjustment.
Unless otherwise specified in the examples, the reaction temperature was room temperature (20 ℃ C. to 30 ℃ C.).
Unless otherwise indicated, reagents used in the examples were purchased from Acros Organics, Aldrich chemical company, Nanjing pharmacosome technology, Shanghai Shuyao sub-pharmaceutical technology, and the like.
The abbreviations used herein have the following meanings:
Figure BDA0002253496460000231
preparation examples
Example A: preparation of 6-chloro-3-iodo-4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridine (A-6)
Figure BDA0002253496460000241
The first step is as follows: preparation of Compound 2, 6-dichloro-4-methoxypyridine (A-2)
2, 6-dichloro-4-nitropyridine (A-1) (10.0g,51.8mmol), potassium carbonate (21.5g,155.8mmol) and methanol (42mL) were added to a 100mL microwave tube, followed by heating to 70 ℃ in a microwave and stirring for reaction for 0.5 hour. After the reaction, the temperature is reduced to 25 ℃, then 30mL of ethyl acetate is added for dilution, the mixture is filtered, a filter cake is rinsed by ethyl acetate (30mL), and the filtrate is concentrated to dryness to obtain the compound A-2(9.0g, the yield is 98%).
The second step is that: preparation of Compound 2, 6-dichloro-4-methoxy-pyridine-3-carbaldehyde (A-3)
2, 6-dichloro-4-methoxy-pyridine (A-2) (4.5g,25.42mmol) and anhydrous THF (100mL) were added to a 250mL three-necked flask, dissolved with stirring under nitrogen, and cooled to-78 ℃. Subsequently, nBuLi (27.81mmol,12.85mL,2.5N in THF) was added dropwise to the reaction over 20 minutes, and the reaction was incubated for 0.5 hour. DMF (3.70g,50.56mmol) was added dropwise to the reaction mixture over 15 minutes, and the reaction was incubated for 2 hours after the addition. Saturated ammonium chloride (100mL) was slowly added to the reaction solution to quench the reaction, ethyl acetate (60mL) was added to extract twice, the organic phases were combined, washed with saturated brine (150mL) 1 time, the organic phase was dried over anhydrous sodium sulfate, and the crude product was purified by silica gel column chromatography to give Compound A-3(4.5g, 86% yield).
The third step: preparation of 6-chloro-4-methoxy-1H-pyrazolo [3,4-b ] pyridine (A-4)
A250 mL three-necked flask was charged with 2, 6-dichloro-4-methoxy-pyridine-3-carbaldehyde (A-3) (4.5g,21.84mmol) and THF (50mL), and 80% hydrazine hydrate (2.5g,32.76mmol) was added with stirring. After the addition, the temperature was raised to 50 ℃ to react for 1 hour, and the solvent was removed by concentration under reduced pressure. Dioxane (40mL) was added to the residue, and the mixture was transferred to a microwave tube and heated to 150 ℃ by microwave for 8 hours. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography to give Compound A-4(1.0g, yield 25%).
The fourth step: preparation of 6-chloro-3-iodo-4-methoxy-1H-pyrazolo [3,4-b ] pyridine (A-5)
6-chloro-4-methoxy-1H-pyrazolo [3,4-b ] pyridine (A-4) (1.0g,5.4mmol), NIS (2.5g,11mmol) and 1, 2-dichloroethane (30mL) were charged into a 100mL reaction flask, heated to 80 ℃ for reaction for 16 hours, concentrated under reduced pressure to remove the solvent, and the crude product was purified by silica gel column chromatography to give Compound A-5(1.5g, 89% yield).
The fifth step: preparation of 6-chloro-3-iodo-4-methoxy-1- (2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridine (A-6)
6-chloro-3-iodo-4-methoxy-1H-pyrazolo [3,4-b ] pyridine (A-5) (1.50g,4.84mmol) and anhydrous THF (20mL) were added to a 100mL three-necked flask and cooled to 0 ℃ under nitrogen. 60% NaH (0.23g,5.81mmol) was added to the reaction and stirred at constant temperature for 0.5 h, followed by the addition of SEMCl (0.97g, 5.81mmol), stirring was continued for 10 min and then allowed to warm to room temperature for 20 min. The reaction was quenched by slowly adding saturated ammonium chloride (30mL), extracted twice with ethyl acetate (20mL), the organic phases were combined, washed 1 time with saturated brine (40mL), dried over anhydrous sodium sulfate, and the crude product was purified by silica gel column chromatography to give Compound A-6(1.90g, 89% yield).
EXAMPLE 1 preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (2, 3-dichlorophenyl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM1)
Figure BDA0002253496460000251
The first step is as follows: preparation of 6-chloro-3- (2, 3-dichlorophenyl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridine (1-1)
6-chloro-3-iodo-4-methoxy-1- (2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridine (A-6) (0.190g,0.43mmol), 2, 3-dichlorophenylboronic acid (0.082g,0.43mmol), tetrakis triphenylphosphine palladium (50mg, 43. mu. mol), sodium carbonate (0.091g,0.86mmol), dioxane (8mL) and water (1mL) were charged into a 20mL microwave tube, and the mixture was heated to 100 ℃ for 1 hour under nitrogen substitution in a microwave oven. After completion of the reaction, the temperature was lowered to 25 ℃ and the reaction mixture was filtered, and the filtrate was concentrated and purified by silica gel column chromatography to obtain Compound 1-1(0.170g, yield 86%).
The second step is that: preparation of tert-butyl (1- (3- (2, 3-dichlorophenyl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -4-methylpiperidin-4-yl) carbamate (1-2)
A10 mL reaction flask was charged with 6-chloro-3- (2, 3-dichlorophenyl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridine (1-1) (0.17g,0.37mmol), (4-methylpiperidin-4-yl) carbamic acid tert-butyl ester (0.16g,0.74mmol), potassium phosphate (0.25g,1.18mmol) and NMP (8mL), heated to 120 ℃ and reacted for 6 hours. After the reaction was complete, the temperature was reduced to 25 ℃ and water (15mL) was added. Ethyl acetate (20mL) was extracted 1 time, washed 1 time with saturated brine (20mL), dried over anhydrous sodium sulfate, and the crude product was concentrated under reduced pressure and purified by silica gel column chromatography to give compound 1-2(94mg, yield 40%).
The third step: preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (2, 3-dichlorophenyl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM1)
A10 mL reaction flask was charged with tert-butyl (1- (3- (2, 3-dichlorophenyl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -4-methylpiperidin-4-yl) carbamate (1-2) (90mg,0.14mmol) and trifluoroacetic acid (5mL), and heated to 80 ℃ for 6 hours. After the reaction is finished, the temperature is reduced to 25 ℃, and the mixture is concentrated under reduced pressure and dried. The residue was added with 40% aqueous hydrobromic acid (15mL) and heated to 100 ℃ for 4 hours. After the reaction, the temperature was decreased to 25 ℃ and the pH was adjusted to 9 with 4N NaOH, ethyl acetate (10mL) was extracted 2 times, the organic phases were combined, washed with saturated brine (20mL) 1 time, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude product, which was subjected to reverse phase HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% aqueous trifluoroacetic acid) to prepare the trifluoroacetate salt of compound TM 1(9 mg, yield 13%).
MS m/z(ESI):392.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.95(s,1H),10.80(br,1H),7.92(s,3H),7.69(dd,J=7.4Hz,2.2Hz,1H),7.46-7.39(m,2H),6.04(s,1H),4.00-3.97(m,2H),3.30-3.25(m,2H),1.76-1.66(m,4H),1.38(s.3H).
Example 2: preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- ((2, 3-dichlorophenyl) thio) -1H pyrazolo [3,4-b ] pyridin-4-ol (TM2)
Figure BDA0002253496460000261
The first step is as follows: preparation of 6-chloro-3- ((2, 3-dichlorophenyl) thio) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridine (2-1)
A50 mL reaction flask was charged with 6-chloro-3-iodo-4-methoxy-1- (2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridine (A-6) (0.300g,0.682mmol), 2, 3-dichlorothiophenol (0.122g,0.682mmol), N-diisopropylethylamine (0.176g,1.364mmol), 4, 5-bis diphenylphosphine-9, 9-dimethylxanthene (39.5mg, 68.2. mu. mol), palladium acetate (15.3mg, 68.2. mu. mol), and dioxane (10 mL). The reaction was carried out at 80 ℃ for 3 hours under nitrogen. After the reaction, the temperature was reduced to 25 ℃ and the reaction mixture was extracted twice with water (30mL) and ethyl acetate (20 mL). The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and the crude product was concentrated under reduced pressure and purified by silica gel column chromatography to give compound 2-1(125mg, yield 37%).
The second step is that: preparation of tert-butyl (1- (3- ((2, 3-dichlorophenyl) thio) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -4-methylpiperidin-4-yl) carbamate (2-2)
To a 15mL microwave tube were added 6-chloro-3- ((2, 3-dichlorophenyl) thio) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridine (2-1) (125mg,254.6 μmol), (4-methylpiperidin-4-yl) carbamic acid tert-butyl ester (218mg,1.02mmol), potassium phosphate (162mg,763.2 μmol), and NMP (5 mL). The reaction mixture was reacted at 120 ℃ for 4 hours under microwave, then cooled to 25 ℃, and water (15mL) and ethyl acetate (10mL) were added to the reaction mixture to extract 2 times, and the organic phase was washed with water, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The crude product was purified by column chromatography to give compound 2-2(98mg, yield 58%).
The third step: preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- ((2, 3-dichlorophenyl) thio) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM2)
A trifluoroacetate salt of TM2 (20mg, yield 25%) was synthesized in a similar manner to that described for the third step in example 1, except that 2-2 was used in this step instead of 1-2 in the third step in example 1.
MS m/z(ESI):424.0[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.33(s,1H),10.90(br,1H),7.89(s,3H),7.44-7.41(dd,J=8.0Hz,1.3Hz,1H),7.20-7.16(t,J=8.0Hz,1H),.6.71-6.68(dd,J=8.0Hz,1.3Hz,1H),5.99(s,1H),4.00-3.95(m,2H),3.30-3.24(m,2H),1.73-1.67(m,4H),1.37(s.3H).
Example 3: preparation of 6- ((3S,4S) -4-amino-3-methyl-2-oxa-8-azaspiro [4.5] decan-8-yl) -3- (2, 3-dichlorophenyl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM3)
Figure BDA0002253496460000271
The first step is as follows: preparation of (3S,4S) -8- (3-iodo-4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine (3-1)
A-6(50mg,0.11mmol), (3S,4S) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine (39mg,0.23mmol), potassium phosphate anhydrous (72mg,0.34mmol) and N-methylpyrrolidone (1mL) were charged into a 5mL reaction flask at 25 ℃ and the temperature was raised to 130 ℃ for reaction for 2 hours. After the reaction was completed, the reaction mixture was diluted with water (10mL), extracted with ethyl acetate (10mL) three times, the combined organic phases were washed with saturated brine (30mL), dried over anhydrous sodium sulfate, filtered, and the solvent was evaporated under reduced pressure, and the crude product was purified by preparative thin layer chromatography to give compound 3-1(40mg, yield 61%).
The second step is that: preparation of (3S,4S) -8- (3- (2, 3-dichlorophenyl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine (3-2)
3-1(10mg,0.020mmol) and 2, 3-dichlorophenylboronic acid (4mg,0.020mmol) were dissolved in 1, 4-dioxane (1mL) and water (0.250mL) at 25 ℃, tetrakistriphenylphosphine-palladium (5mg,0.004mmol) and cesium carbonate (10mg,0.030mmol) were added, replaced with nitrogen 3 times, and the temperature was raised to 100 ℃ for 1 hour. After the reaction was completed, the temperature was reduced to 25 ℃ and diluted with water (20mL), extracted with ethyl acetate (20mL) for 3 times, the combined organic phases were washed with saturated brine (30mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give crude 3-2(10mg, yield 100%) which was used directly in the next reaction.
The third step: preparation of 6- ((3S,4S) -4-amino-3-methyl-2-oxa-8-azaspiro [4.5] decan-8-yl) -3- (2, 3-dichlorophenyl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM3)
3-2(10mg,0.06mmol) was dissolved in 40% HBr (1mL) at 25 deg.C for 48 h and concentrated to dryness after completion of the reaction. The crude product was prepared by reverse phase HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% aqueous trifluoroacetic acid) to give the trifluoroacetate salt of compound TM 3(4 mg, yield 40%).
MS m/z(ESI):448.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.90(s,1H),10.69(s,1H),7.87(s,3H),7.69(d,J=7.3Hz,1H),7.47-7.38(m,2H),6.02(s,1H),4.22-4.17(m,1H),4.14-4.07(m,1H),4.05-3.98(m,1H),3.89(d,J=9.2Hz,1H),3.71(d,J=9.0Hz,1H),3.41-3.34(m,1H),3.15-3.00(m,2H),1.79-1.65(m,3H),1.56-1.48(m,1H),1.21(d,J=6.5Hz,3H).
Example 4: preparation of 6- ((3S,4S) -4-amino-3-methyl-2-oxo-8-azaspiro [4.5] decan-8-yl) -3- (2, 3-dichloropyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM8)
Figure BDA0002253496460000281
The first step is as follows: preparation of (3S,4S) -8- (3- (2, 3-dichloropyridin-4-yl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -3-methyl-2-oxo-8-pyrazolo [4.5] decan-4-amine (8-1)
8-1(150mg, yield 48%) was synthesized in a similar manner to that described for the second step in example 3, except that 2, 3-dichloro-pyridine-4-boronic acid pinacol ester was used in this step instead of 2, 3-dichlorophenylboronic acid in the second step in example 3.
The second step is that: preparation of 6- ((3S,4S) -4-amino-3-methyl-2-oxo-8-azaspiro [4.5] decan-8-yl) -3- (2, 3-dichloropyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM8)
A10 mL reaction flask was charged with 8-1(90mg,0.14mmol) and trifluoroacetic acid (2mL), and heated to 80 ℃ for 6 hours. After the reaction is finished, the temperature is reduced to 25 ℃, and the mixture is concentrated under reduced pressure and dried. The residue was added concentrated hydrochloric acid (15mL) and heated to 100 ℃ for 4 hours. After the reaction, the temperature was decreased to 25 ℃ and the pH was adjusted to 9 with 4N NaOH, ethyl acetate (10mL) was extracted 2 times, the organic phases were combined, washed with saturated brine (20mL) 1 time, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude product, which was subjected to reverse phase HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% aqueous formic acid) to give the formate salt of TM8 (26mg, yield 34%).
MS m/z(ESI):449.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.19(s,1H),10.96(s,1H),8.45(d,J=4.8Hz,1H),7.97(s,3H),7.58(d,J=4.8Hz,1H),6.09(s,1H),4.21-4.16(m,1H),4.16-4.07(m,1H),4.07-3.98(m,1H),3.93-3.86(m,1H),3.73-3.68(m,1H),3.40-3.34(m,1H),3.09-3.03(m,2H),1.86-1.63(m,3H),1.60-1.50(m,1H),1.22(d,J=6.5Hz,3H).
Example 5: preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (2, 3-dichloropyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM61)
Figure BDA0002253496460000291
The first step is as follows: preparation of tert-butyl (1- (3-iodo-4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -4-methylpiperidin-4-yl) carbamate (61-1)
61-1(500mg, yield 71%) was synthesized in a similar manner to the procedure described for the first step in example 3, except that tert-butyl (4-methylpiperidin-4-yl) carbamate was used in this step in place of (3S,4S) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine in the first step in example 3.
The second step is that: preparation of tert-butyl (1- (3- (2, 3-dichloropyridin-4-yl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -4-methylpiperidin-4-yl) carbamate (61-2)
61-2(180mg, yield 35%) was synthesized in a similar manner to that described for the second step in example 3, except that 2, 3-dichloro-pyridine-4-boronic acid pinacol ester was used instead of 2, 3-dichlorophenylboronic acid in the second step in example 3 and 61-1 was used instead of 3-1 in example 3 in this step.
The third step: preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (2, 3-dichloropyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM61)
Crude TM61 was synthesized in a similar manner as described for the second step in example 4, except that 61-2 was used in this step instead of 8-1 in the second step in example 4. The crude product was prepared by reverse phase HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% aqueous formic acid) to give the formate salt of compound TM61 (17mg, yield 14%).
MS m/z(ESI):393.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.21(s,1H),10.97(s,1H),8.44(d,J=4.9Hz,1H),8.02(s,3H),7.58(d,J=4.9Hz,1H),6.08(s,1H),3.99(m,2H),3.38-3.20(m,2H),1.79-1.66(m,4H),1.38(s,3H).
Example 6: preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (2-bromo-3-chloropyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM62)
Figure BDA0002253496460000301
The first step is as follows: preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (2-bromo-3-chloropyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM62)
A trifluoroacetate salt of TM62 (25mg, yield 15%) was synthesized in a similar manner to that described for the third step in example 1 except that 61-2 was used in this step instead of 1-2 in example 1.
MS m/z(ESI):437.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.20(s,1H),10.95(s,1H),8.41(d,J=4.8Hz,1H),7.88(s,3H),7.58(d,J=4.8Hz,1H),6.06(s,1H),4.04-3.95(m,2H),3.32-3.23(m,2H),1.76-1.65(m,4H),1.38(s,3H).
Example 7: preparation of 6- ((3S,4S) -4-amino-3-methyl-2-oxa-8-azaspiro [4.5] decan-8-yl) -3- (2-bromo-3-chloropyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM63)
Figure BDA0002253496460000302
The first step is as follows: preparation of (3S,4S) -8- (3- (2-bromo-3-chloropyridin-4-yl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine (63-1)
63-1(10mg, yield 16%) was synthesized in a similar manner to that described for the second step in example 3, except that 2-bromo-3-chloro-pyridine-4-boronic acid was used in this step instead of 2, 3-dichlorophenylboronic acid in the second step in example 3.
The second step is that: preparation of 6- ((3S,4S) -4-amino-3-methyl-2-oxa-8-azaspiro [4.5] decan-8-yl) -3- (2-bromo-3-chloropyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM63)
A crude product of TM63 was synthesized in a similar manner to that described in the third step of example 1, except that 63-1 was used in this step instead of 1-2 in example 1. The crude product was prepared by reverse phase HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% aqueous formic acid) to give the formate salt of compound TM63 (6mg, yield 67%).
MS m/z(ESI):493.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.18(s,1H),10.90(s,1H),8.42(d,J=4.8Hz,1H),7.86(s,3H),7.58(d,J=4.8Hz,1H),6.05(s,1H),4.23-4.17(m,1H),4.15-4.10(m,1H),4.06-4.00(m,1H),3.91-3.88(m,1H),3.72-3.70(m,1H),3.41-3.35(m,1H),3.12-3.02(m,2H),1.75-1.66(m,3H),1.54-1.49(m,1H),1.21(d,J=6.5Hz,3H).
Example 8: preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (2-bromo-3-fluoropyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM64)
Figure BDA0002253496460000311
The first step is as follows: preparation of tert-butyl (1- (3- (2-chloro-3-fluoropyridin-4-yl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -4-methylpiperidin-4-yl) carbamate (64-1)
Compound 64-1(90mg, yield 20%) was synthesized in a similar manner to the procedure described for the second step in example 3, except that 2-chloro-3-fluoro-pyridine-4-boronic acid was used instead of 2, 3-dichlorophenylboronic acid in the second step in example 3 and 61-1 was used instead of 3-1 in example 3 in this step.
The second step is that: preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (2-bromo-3-fluoropyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM64)
TM64 trifluoroacetate salt (17mg, yield 48%) was synthesized in a similar manner to that described for the third step in example 1, except that 64-1 was used in this step instead of 1-2 in example 1.
MS m/z(ESI):422.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.34(s,1H),11.19(brs,1H),8.33(d,J=4.9Hz,1H),7.93(s,3H),7.76(m,1H),6.12(s,1H),4.01-3.98(m,2H),3.52-3.14(m,2H),1.72(s,4H),1.38(s,3H).
EXAMPLE 9 preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (2, 6-dichloro-4-pyridinyl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM65)
Figure BDA0002253496460000321
The first step is as follows: preparation of tert-butyl (1- (3- (2, 6-dichloro-pyridin-4-yl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -4-methylpiperidin-4-yl) carbamate (65-1)
Compound 65-1(90mg, yield 87%) was synthesized in a similar manner to that described for the second step in example 3, except that 2, 6-dichloro-4-pyridineboronic acid was used instead of 2, 3-dichlorophenylboronic acid in the second step in example 3 and 61-1 was used instead of 3-1 in example 3 in this step.
The second step is that: preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (2, 6-dichloro-4-pyridinyl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM65)
Crude TM65 was synthesized in a similar manner as described for the second step in example 4, except that 65-1 was used in this step instead of 8-1 in the second step in example 4. The crude product was prepared by reverse phase HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% aqueous formic acid) to give compound TM65(12mg, yield 32%).
MS m/z(ESI):393.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.26(s,1H),8.60(s,2H),8.36(s,1H),6.02(s,1H),3.95-3.88(m,2H),3.27-3.23(m,2H),1.73-1.52(m,4H),1.33(s,3H).
EXAMPLE 10 preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (3, 4-difluorophenyl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM66)
Figure BDA0002253496460000322
The first step is as follows: preparation of tert-butyl (1- (3- (3, 4-difluorophenyl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -4-methylpiperidin-4-yl) carbamate (66-1)
Compound 66-1(63mg, yield 62%) was synthesized in a similar manner to that described for the second step in example 3, except that 3, 4-difluorophenylboronic acid was used in this step instead of 2, 3-dichlorophenylboronic acid in the second step in example 3 and 61-1 was used instead of 3-1 in example 3.
The second step is that: preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (3, 4-difluorophenyl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM66)
TM66 trifluoroacetate salt (34mg, yield 51%) was synthesized in a similar manner to that described for the third step in example 1, except that 66-1 was used in this step instead of 1-2 in example 1.
MS m/z(ESI):360.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.00(s,1H),11.32(s,1H),8.14-8.05(m,1H),7.99-7.82(m,4H),7.55-7.44(m,1H),6.16-6.09(m,1H),4.03-3.94(m,2H),3.33-3.23(m,2H),1.78-1.66(m,4H),1.38(s,3H).
EXAMPLE 11 preparation of 6- (4-amino-4-methylpiperidin-1-yl) -5-chloro-3- (2, 3-dichloropyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM67)
Figure BDA0002253496460000331
The first step is as follows: preparation of tert-butyl (1- (5-chloro-3- (2, 3-dichloropyridin-4-yl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -4-methylpiperidin-4-yl) carbamate (67-1)
A10 mL reaction flask was charged with 61-2(120mg, 188.2. mu. mol) and acetonitrile (10mL), and NCS (101mg, 752.7. mu. mol) was further added to conduct a reaction at 40 ℃ for 20 hours. The reaction mixture was concentrated, and the crude product was purified by preparative thin layer chromatography to give compound 67-1(72mg, yield 59%).
The second step is that: preparation of 6- (4-amino-4-methylpiperidin-1-yl) -5-chloro-3- (2, 3-dichloropyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM67)
Crude TM67 was synthesized in a similar manner to that described for the third step in example 1, except that 67-1 was used in this step instead of 1-2 in example 1. The crude product was prepared by reverse phase HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% aqueous formic acid) to give compound TM67(25mg, yield 55%).
MS m/z(ESI):427.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.66(s,1H),8.33(d,J=4.8Hz,1H),8.10-7.85(br,2H),7.75-7.69(m,1H),3.45-3.35(m,2H),3.02-2.88(m,2H),1.95–1.65(m,4H),1.30(s.3H).
EXAMPLE 12 preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (2-methylpyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM68)
Figure BDA0002253496460000341
The first step is as follows: preparation of tert-butyl (1- (3- (2-methylpyridin-4-yl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazolo [3,4-b ] pyridin-6-yl) -4-methylpiperidin-4-yl) carbamate (66-1)
Compound 68-1(90mg, yield 95%) was synthesized in a similar manner to that described in the second step of example 3, except that 2-methylpyridin-4-ylboronic acid was used in this step in place of 2, 3-dichlorophenylboronic acid in the second step of example 3 and 61-1 was used in place of 3-1 in example 3.
The second step is that: preparation of 6- (4-amino-4-methylpiperidin-1-yl) -3- (2-methylpyridin-4-yl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM68)
Crude TM65 was synthesized in a similar manner as described for the second step in example 4, except that 68-1 was used in this step instead of 8-1 in the second step in example 4. The crude product was prepared by reverse phase HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% aqueous trifluoroacetic acid) to give the trifluoroacetate salt of compound TM68 (20mg, yield 38%).
MS m/z(ESI):339.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.79(s,1H),12.03(brs,1H),8.78(d,J=6.2Hz,1H),8.64-8.48(m,2H),8.05(s,3H),6.32(s,1H),4.05-3.95(m,2H),3.39-3.24(m,2H),2.75(s,3H),1.75-1.54(m,4H),1.39(s,3H).
Example 13: preparation of 3- (2-amino-3-chloro-pyridin-4-yl) -6- (4-amino-4-methyl-1-piperidinyl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM69)
Figure BDA0002253496460000342
The first step is as follows: preparation of (1- (3- (3-chloro-2- ((2, 4-dimethoxybenzyl) amino) pyridin-4-yl) -4-methoxy-1- ((2- (trimethylsilyl) ethoxy) methyl-1H-pyrazolo [3,4-b ] pyridin-6-yl) -4-methylpiperidin-4-yl) carbamic acid tert-butyl ester (69-1) 61-2(80mg,125.46umol), 2, 4-dimethoxybenzylamine (105mg,627.28umol) and NMP (2mL) were added to a 10mL microwave tube, heated to 150 ℃ in a microwave reactor and reacted for 3 hours, 10mL of water was added to the reaction solution, ethyl acetate (10mL) was extracted twice, the organic phases were combined, and washed 1 time with saturated brine (20mL), drying with anhydrous sodium sulfate, and concentrating under reduced pressure to obtain crude product. The crude product was purified by preparative thin layer chromatography to give compound 69-1(21mg, yield 22%).
The second step is that: preparation of 3- (2-amino-3-chloro-pyridin-4-yl) -6- (4-amino-4-methyl-1-piperidinyl) -1H-pyrazolo [3,4-b ] pyridin-4-ol (TM69)
Crude TM2 was synthesized in a similar manner as described for the third step in example 1, except that 69-1 was used in this step instead of 1-2 in the third step in example 1. The crude product was prepared by reverse phase HPLC (mobile phase A: acetonitrile, mobile phase B: 0.05% aqueous formic acid) to give compound TM69(4mg, yield 40%).
MS m/z(ESI):374.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ:12.79(s,1H),7.89(d,J=4.8Hz,1H),6.65(d,J=4.8Hz,1H),6.25(s,2H),5.93(s,1H),3.56-3.54(m,4H),1.49-1.39(m,4H),1.10(s,3H).
Biological examples
Test example 1: SHP2 (protein tyrosine phosphatase) in vitro enzyme activity inhibition assay
The test system comprises:
kinase enzymes: recombinant full-length human PTPN11(SHP2), Active (Signal Chemcatalog: P38-20G)
Substrate: 6,8-Difluoro-4-methylumbelliferyl phosphate (DiFMUP) (Invitrogen catalog: D6567) activating peptide: IRS1_ pY1172(dPEG8) pY1222(BPS Bioscience Catalog:79319-2)
Termination reagent: bpv (phen) (Abcam Catalog: ab141436)
Test parameters are as follows:
SHP2 concentration: 0.5 nM; concentration of difmuup: 200 mu M; IRS-1 concentration: 0.5 mu M; bpv: 160 μ M buffer system: 60mM Hepes pH 7.2; 75mM NaCl; 75mM KCl; 0.05% P-20; 1mM EDTA; 5mM DTT;
compound incubation and enzyme activation time: room temperature for 60 minutes
Enzyme and substrate reaction time: room temperature for 30 minutes
Parameters of the microplate reader: BMG PHERAStar Fluorescence with excitation wavelength of 340nm and emission wavelength of 450nm
The test steps are as follows:
and (3) incubating a mixture of a compound to be detected and phosphatase SHP2 with activation peptide IRS-1 at room temperature for 60 minutes in a buffer solution system, adding a substrate DiFMUP to start reaction, incubating at room temperature for 30 minutes, adding bpv to stop the reaction, putting the reaction plate into an enzyme labeling instrument, and reading the fluorescence value of each hole in the plate by adopting an end-point method.
Data processing:
relative inhibitory activity was calculated for each concentration group using a vehicle group (containing 0.5nM SHP2,200 μ M difup, 0.5 μ M IRS-1,160 μ M bpv, 0.05% DMSO) as a negative control and a reaction buffer group (200 μ M difup, 0.5 μ M IRS-1,160 μ M bpv, 0.05% DMSO) as a blank control, and the inhibitory rate was 100% - (test group-blank group)/(vehicle group-blank group) × 100%. Half maximal Inhibitory Concentration (IC) of the compound was calculated according to a four parameter model fitting curve50)。
And (3) test results:
the inhibition of SHP2 activity by compounds was determined as described above and the results are shown in table 1.
TABLE 1 SHP2 test results of enzyme activity inhibition
Compound numbering IC50(nM)
Example 1 36.0
Example 2 90.0
Example 3 51.0
Example 4 5.3
Example 5 4.8
Example 6 1.3
Example 7 8.8
Example 8 2.0
Example 9 6.3
Example 10 57.5
Example 11 9.5
Example 12 28.0
Example 13 59.9
And (4) conclusion:
in an SHP2 enzyme activity inhibition test, the compound of the invention shows stronger inhibition activity.
Test example 2: inhibition assay of cell proliferation Activity of KYSE-520 with Compounds
The test system comprises:
cell name/manufacturer: KYSE-520/JCRB Cell Bank
Kit name/manufacturer: CellTiter-
Figure BDA0002253496460000361
Luminescent Cell visual Assay, Promega, experimental parameters:
cell number: 1500 cells/well
Plating a culture medium: KYSE-520:1640+ 10% FBS
Adding a culture medium: KYSE-520:1640+ 10% FBS
Compound incubation conditions: 37 ℃ and 5% CO2
Incubation time: 5d
Detecting the temperature: RT (reverse transcription)
Parameters of the microplate reader: BMG PHERAStar FS Luminescent
The test steps are as follows:
culturing the cellsIn a medium containing 10% fetal bovine serum, the medium was left at 37 ℃ with 5% CO2Culturing is carried out under culture conditions. And (3) paving a proper amount of cells into a 96-well plate, and culturing overnight in an incubator to ensure that the cells are attached to the wall. The next day, the medium was removed, complete medium containing the prediluted compound was added and incubated at 37 ℃ for 5 d. On the fifth day, the detection reagent CellTiter-GLo was added to each well, and Relative Luminescence Units (RLU) of each well were detected by chemiluminescence.
Data processing:
background values were obtained using CellTiter-Glo in cell-free medium.
Cell viability ═ 100% of (sample RLU-background RLU)/(vehicle RLU-background RLU),
maximum inhibition rate of 100% -cell viabilityMaximum concentration
Half maximal Inhibitory Concentration (IC) of the compound was calculated according to a four parameter model fitting curve50)。
And (3) test results:
the inhibitory activity of the compound on the proliferation of KYSE-520 (human esophageal squamous carcinoma cells) was measured according to the above-mentioned method, and the results are shown in Table 2.
TABLE 2 inhibition of KYSE-520 cell proliferation Activity by Compounds
Figure BDA0002253496460000371
And (4) conclusion:
the compound has stronger cell proliferation inhibition activity on KYSE-520.

Claims (24)

1. A compound of formula (I) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof:
Figure FDA0002253496450000011
wherein:
R1selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -OC1-6Alkyl radical, C1-6Alkyl and halo C1-6An alkyl group;
l is selected from the group consisting of a direct bond, -NH-, -CH2-、-CH2CH2-, -CH ═ CH-and-C (═ CH)2)-;
Or L and R1Together with the atoms to which they are attached form a 5-10 membered hydrocarbon ring, a 5-10 membered heterocyclic ring, a 5-10 membered heteroaromatic ring or C6-10Aromatic rings, wherein the hydrocarbon, heterocyclic, heteroaromatic and aromatic rings are each optionally substituted with one or more groups selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -O, -OH, -OC1-6Alkyl, halo C1-6Alkyl and C1-6Alkyl substituent substitution;
x is selected from the group consisting of a direct bond, -S-, -O-, -NR6-、-C(R6)2-and-C (═ CH)2)-;
W1、W2And W3Each independently selected from CH and N;
ring A is selected from a 4-10 membered heterocyclic ring and a 4-10 membered hydrocarbon ring;
R2each occurrence is independently selected from H, halogen, -OH, -OC1-6Alkyl, -CN, C1-6Alkyl radical, C3-6Cycloalkyl radical, C2-6Alkenyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)23-to 10-membered heterocyclic group, C6-10Aryl and 5-10 membered heteroaryl, wherein said alkyl, cycloalkyl, alkenyl, heterocyclyl, aryl and heteroaryl are each optionally substituted with one or more substituents selected from the group consisting of halogen, ═ O, -OH, -OC1-6Alkyl, -CN, C1-6Alkyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl) and-N (C)1-6Alkyl radical)2Substituted with the substituent(s);
or 2R in adjacent positions2Together with the atoms to which they are attached form a 5-10 membered hydrocarbon ring, a 5-10 membered heterocyclic ring, a 5-6-membered heteroaromatic ring or phenyl ring, wherein the hydrocarbon ring, heterocyclic ring, heteroaromatic ring and phenyl ring are each optionally substituted with one or more groups selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -O, -OH, -OC1-6Alkyl, halo C1-6Alkyl and C1-6Alkyl substituent substitution;
R3selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -OH, -COOH, -C (O) OC1-6Alkyl, -C (O) NH2、-OC1-6Alkyl and C1-6Alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, -OH, ═ O, -OC1-6Alkyl, -CN, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl) and-N (C)1-6Alkyl radical)2Substituted with the substituent(s);
R4each occurrence is independently selected from H, halogen, ═ O, -OH, -OC1-6Alkyl, -CN, C1-6Alkyl radical, C3-6Cycloalkyl radical, C2-6Alkenyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl) and-N (C)1-6Alkyl radical)2
R5aAnd R5bAre substituents on the same carbon atom and are each independently selected from H, halogen, -OH, -OC1-6Alkyl, -C1-6alkylene-OH, -CN, halo-C1-6Alkyl radical, C1-6Alkyl radical, C3-6Cycloalkyl radical, C2-6Alkenyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2and-C1-6alkylene-NH2
Or R5aAnd R5bTogether with the atoms to which they are attached form a 3-8 membered hydrocarbon ring or a 3-8 membered heterocyclic ring, wherein the hydrocarbon ring and heterocyclic ring are each optionally substituted with one or more substituents selected from H, halogen, ═ O, -OH, -OC1-6Alkyl, -C1-6alkylene-OH, -CN, halo-C1-6Alkyl radical, C1-6Alkyl radical, C3-6Cycloalkyl radical, C2-6Alkenyl, -S (═ O)g-(C1-6Alkyl), -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2and-C1-6alkylene-NH2Substituted with the substituent(s);
R6is H or C1-6An alkyl group;
or, any one of R2And one R6Together with the atoms to which they are attached form a 5-10 membered hydrocarbon ring, a 5-10 membered heterocyclic ring, a 5-6 membered heteroaromatic ring or a benzene ring, wherein the hydrocarbon ring, heterocyclic ring, heteroaromatic ring and benzene ring are each optionally substituted with one or more groups selected from H, -NH2、-NH(C1-6Alkyl), -N (C)1-6Alkyl radical)2Halogen, -CN, -O, -OH, -OC1-6Alkyl, halo C1-6Alkyl and C1-6Alkyl substituent substitution;
g is 0, 1 or 2;
m is 0, 1,2, 3 or 4;
n is 0, 1,2, 3 or 4.
2. A compound of formula (I) according to claim 1, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically-labeled compound, metabolite or prodrug thereof, wherein R is1Selected from H, -NH2、-OC1-3Alkyl, halo C1-3Alkyl and C1-3Alkyl, preferably R1Selected from H, -NH2and-CH3
3. A compound of formula (I) according to claim 1 or 2, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically-labeled compound, metabolite or prodrug thereof, wherein L is selected from the group consisting of a direct bond, -NH-and-CH2-, preferably L is selected from the group consisting of a direct bond and-CH2-。
4. A compound of formula (I) according to any one of claims 1 to 3 or a salt thereofA pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug, wherein L and R1Together with the atoms to which they are attached form a 5-6 membered heterocyclic ring or a 5-6 membered heteroaromatic ring, wherein each of the heterocyclic and heteroaromatic rings is optionally substituted with one or more groups selected from H, -NH2F, Cl, -CN, -O, -OH, halogeno-C1-3Alkyl and C1-3Alkyl substituent substitution;
preferably, L and R1Together with the atoms to which they are attached form a 5-6 membered heteroaromatic ring optionally substituted with one or more groups selected from H, -NH2F, Cl, -CN, -O, -OH, halogeno-C1-3Alkyl and C1-3Alkyl substituent substitution;
also preferably, L and R1Together with the atoms to which they are attached form a pyrazole, pyrrole, pyrroline, imidazole, pyrazine or pyridine ring, wherein each of said rings is optionally substituted by one or more groups selected from H, -NH2F, Cl, -CN, -O, -OH, halogeno-C1-3Alkyl and C1-3Alkyl substituent substitution;
it is further preferred that the first and second liquid crystal compositions,
Figure FDA0002253496450000031
moieties are selected from the group consisting of:
Figure FDA0002253496450000032
Figure FDA0002253496450000033
it is further preferred that the first and second liquid crystal compositions,
Figure FDA0002253496450000034
part is selected from
Figure FDA0002253496450000035
5. According to the rightA compound of formula (I) as claimed in any one of claims 1-4 wherein X is selected from the group consisting of a direct bond, -S-, -O-and-C (═ CH), or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof2) -; preferably, X is selected from the group consisting of a direct bond, -S-, and-O-; more preferably, X is selected from a direct bond and-S-.
6. A compound of formula (I) according to any one of claims 1 to 5 or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, wherein W is1Is CH, W2And W3Each independently selected from CH and N; preferably, W1、W2And W3Are both CH.
7. A compound of formula (I) according to any one of claims 1-6, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, wherein ring a is selected from a 5-6 membered heterocyclic ring and a 5-6 membered hydrocarbon ring; preferably, ring a is selected from a 6-membered nitrogen-containing heterocyclic ring and a 6-membered hydrocarbon ring; more preferably, ring a is selected from the group consisting of piperidine ring and cyclohexene ring.
8. A compound of formula (I) according to any one of claims 1 to 7, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically-labeled compound, metabolite or prodrug thereof, wherein R is2Each occurrence is independently selected from H, halogen, -OH, -OC1-6Alkyl, -CN, C1-3Alkyl, halo C1-3Alkyl and-NH2(ii) a Preferably, R2Each occurrence is independently selected from H, F, Cl, Br, -OCH3、-OH、-CH3、-CH2CH3、-CF3、-CHF2and-NH2(ii) a More preferably, R2Each occurrence independently selected from F, Cl, Br, -OCH3、-CH3、-CF3and-NH2(ii) a Further preferably, R2Each occurrence independently selected from F, Cl, -CH3、-CF3and-NH2(ii) a In a still further preferred embodiment, R2Each occurrence independently selected from Br, Cl and-NH2
9. A compound of formula (I) according to any one of claims 1 to 8, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, wherein 2R at adjacent positions2Together with the atoms to which they are attached form a 5-7 membered hydrocarbon ring or a 5-7 membered heterocyclic ring, wherein the hydrocarbon ring and heterocyclic ring are each optionally substituted with one or more groups selected from H, -NH2、F、Cl、-CN、=O、-OH、-OC1-3Alkyl, halo C1-3Alkyl and C1-3Alkyl substituent substitution;
preferably 2R in adjacent positions2Together with the atoms to which they are attached form a 5-6 membered hydrocarbon ring or a 5-6 membered heterocyclic ring, wherein the hydrocarbon ring and heterocyclic ring are each optionally substituted with one or more groups selected from H, -NH2、F、Cl、=O、-OH、-OCH3and-CH3Substituted with the substituent(s);
also preferably, 2R's in adjacent positions2Together with the atoms to which they are attached form a ring selected from cyclopentene, dioxole and pyrroline, wherein each of said rings is optionally substituted with one or more groups selected from H, -NH2、F、Cl、=O、-OH、-OCH3and-CH3Substituted with the substituent(s);
more preferably, said 2R are relative to the point of attachment to X2Respectively at ortho-position and meta-position;
it is still further preferred that the first and second substrates are,
Figure FDA0002253496450000041
is selected from the followingThe group of (a):
Figure FDA0002253496450000042
Figure FDA0002253496450000043
10. a compound of formula (I) according to any one of claims 1 to 9, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically-labelled compound, metabolite or prodrug thereof, wherein when X is-NR6-or-C (R)6)2When in the ortho position to the point of attachment of X, an R2And one R6Together with the atoms to which they are attached form a 5-7 membered hydrocarbon ring or a 5-7 membered heterocyclic ring, wherein the hydrocarbon ring and heterocyclic ring are each optionally substituted with one or more groups selected from H, -NH2、F、Cl、-CN、=O、-OH、-OC1-3Alkyl, halo C1-3Alkyl and C1-3Alkyl substituent substitution;
preferably, when X is-NR6-or-C (R)6)2When in the ortho position to the point of attachment of X, an R2And one R6Together with the atoms to which they are attached form a 5-6 membered hydrocarbon ring or a 5-6 membered heterocyclic ring, wherein the hydrocarbon ring and heterocyclic ring are each optionally substituted with one or more groups selected from H, -NH2、F、Cl、=O、-OH、-OCH3and-CH3Substituted with the substituent(s);
also preferably, when X is-NR6-or-C (R)6)2When in the ortho position to the point of attachment of X, an R2And one R6Together with the atoms to which they are attached form a ring selected from cyclohexene and tetrahydropyridine, wherein each of said rings is optionally substituted by one or more groups selected from H, -NH2、F、Cl、-CN、=O、-OH、-OC1-3Alkyl, halo C1-3Alkyl and C1-3Alkyl substituent substitution;
even more preferably, when X is-NR6-or-C (R)6)2When the pressure in the air is higher than the preset pressure,
Figure FDA0002253496450000044
selected from the group consisting of:
Figure FDA0002253496450000045
Figure FDA0002253496450000046
11. a compound of formula (I) according to any one of claims 1 to 10, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically-labelled compound, metabolite or prodrug thereof, wherein R is3Selected from H, -NH2Halogen, -CN, -OH, C1-4Alkyl, -COOH and-C (O) OC1-4Alkyl, wherein said alkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, ═ O, -OH, -S (═ O)g-(C1-3Alkyl) and-NH2Substituted with the substituent(s); preferably, R3Selected from H, -NH2、F、Cl、-CN、-OH、-CH3、-CF3、-CH2CH3、-CH2F、-CHF2、-CH2OH、-CH2CH2OH、-CH2NH2、-CH2CH2NH2-COOH and-C (O) OEt; more preferably, R3Selected from H, F, -CN, -COOH, -CH2OH、-CH2F、-CHF2and-CH2OH; more preferably, R3Selected from H, F and-CH2OH。
12. A compound of formula (I) according to any one of claims 1 to 11, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically-labeled compound, metabolite or prodrug thereof, wherein R is4Each occurrence is independently selected from H, F, Cl, ═ O, -OH, -OC1-3Alkyl, -CN, C1-3Alkyl and-NH2(ii) a In a preferred embodiment, R4Each occurrence is independently selected from H, F, Cl, -OH, -CH3and-NH2(ii) a In a further preferred embodiment, R4Each occurrence independently selected from H and-CH3
13. A compound of formula (I) according to any one of claims 1 to 12, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically-labelled compound, metabolite or prodrug thereof, wherein R is5aAnd R5bEach independently selected from H, C1-3Alkyl, -NH2、-C1-3alkylene-OH and-C1-3alkylene-NH2(ii) a Or R5aAnd R5bTogether with the atoms to which they are attached form a 5-6 membered hydrocarbon ring or a 5-6 membered heterocyclic ring, wherein the hydrocarbon ring and heterocyclic ring are each optionally substituted with one or more groups selected from H, F, Cl, C1-3Alkyl, ═ O, -NH2、-OH、-C1-3alkylene-OH and-C1-3alkylene-NH2Substituted with the substituent(s);
preferably, R5aAnd R5bEach independently selected from H, -CH3、-CH2CH3、-NH2and-CH2NH2(ii) a Or R5aAnd R5bTogether with the atoms to which they are attached form a cyclopentane, an oxolane or a dihydrooxazole, wherein each of the cyclopentane, oxolane or dihydrooxazole is optionally substituted with one or more groups selected from-CH3and-NH2Substituted with the substituent(s);
more preferably, R5aAnd R5bEach independently selected from-CH3and-NH2(ii) a Or R5aAnd R5bTogether with the atoms to which they are attached form a cyclopentane, an oxolane or a dihydrooxazole, wherein each of the cyclopentane, oxolane or dihydrooxazole is simultaneously substituted with a-CH3and-NH2Is substituted with the substituent(s).
14. A compound of formula (I) according to any one of claims 1-13, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, wherein g is 0 or 2.
15. A compound of formula (I) according to any one of claims 1-14, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, wherein m is 0, 1 or 2; preferably, m is 0.
16. A compound of formula (I) according to any one of claims 1-15, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, wherein N is 0, 1 or 2; preferably, n is 1 or 2.
17. A compound of formula (II) or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof:
Figure FDA0002253496450000051
wherein R is1、R2、R3、R4、R5a、R5b、W1、W2、W3L, X, A, m and n are as defined in any one of claims 1 to 16.
18. A compound of formula (I) according to any one of claims 1-16, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, wherein the compound of formula (I) has the structure of formula (III) or formula (IV):
Figure FDA0002253496450000061
wherein the content of the first and second substances,
Figure FDA0002253496450000063
represents a single bond or a double bond; u is selected from N and C; when in use
Figure FDA0002253496450000065
Is a single bond, U is N, when
Figure FDA0002253496450000064
Is a double bond, U is C; ring B is selected from a 5-6 membered heterocyclic ring or a 5-6 membered heteroaromatic ring, wherein each of said heterocyclic and heteroaromatic rings is optionally substituted with one or more groups selected from H, -NH2F, Cl, -CN, -O, -OH, halogeno-C1-3Alkyl and C1-3Alkyl substituent substitution; preferably, ring B is selected from a 5-6 membered heteroaromatic ring; more preferably, ring B is selected from a pyrazole ring, a pyrrole ring, a pyrroline ring, an imidazole ring, a pyrazine ring or a pyridine ring; the remaining groups are as defined in any one of claims 1 to 16; preferably, the first and second electrodes are formed of a metal,
Figure FDA0002253496450000066
is a single bond, and U is N.
19. A compound of formula (I) according to any one of claims 1-16, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, wherein the compound of formula (I) has the structure of formula (V), formula (VI) or formula (VII):
Figure FDA0002253496450000062
wherein R is2aAnd R2bAre defined as each2Same as R2aAnd R2bMay be the same or different; q1、Q2And Q3Each is independently selected from N, NH, C and CH, and is not simultaneously C or CH; the remaining groups are as defined in any one of claims 1 to 16.
20. A compound or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite or prodrug thereof, wherein said compound is selected from the group consisting of:
Figure FDA0002253496450000071
Figure FDA0002253496450000081
Figure FDA0002253496450000091
21. a pharmaceutical composition comprising a compound of any one of claims 1-20, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, and one or more pharmaceutically acceptable carriers.
22. Use of a compound of any one of claims 1-20, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, N-oxide, isotopically labeled compound, metabolite, or prodrug thereof, or a pharmaceutical composition of claim 21, in the manufacture of a medicament for the prevention or treatment of a SHP2 phosphatase-related disease.
23. The use of claim 22, wherein the SHP2 phosphatase-associated disease is a disease sensitive to or responsive to SHP2 phosphatase inhibition; preferably, the SHP2 phosphatase-related disease is a neoplastic disease including, but not limited to, solid and hematological malignancies, such as breast cancer, colorectal cancer, colon cancer, lung cancer (including small cell lung cancer, non-small cell lung cancer and bronchioloalveolar cancer) and prostate cancer, as well as biliary tract cancer, bone cancer, bladder cancer, head and neck cancer, kidney cancer, liver cancer, gastrointestinal tissue cancer, esophageal cancer, ovarian cancer, pancreatic cancer, skin cancer, testicular cancer, thyroid cancer, uterine cancer, cervical cancer and vulvar cancer, and leukemias (including Chronic Lymphocytic Leukemia (CLL), Acute Lymphocytic Leukemia (ALL) and Chronic Myelogenous Leukemia (CML)), multiple myeloma and lymphoma.
24. A process for the preparation of a compound according to any one of claims 1 to 20, which process comprises:
(1) reacting a compound of formula S-1 with a compound of formula S-2 to produce a compound of formula IM-1;
Figure FDA0002253496450000101
(2) reacting a compound of formula IM-1 with a compound of formula S-3 to form a compound of IM-2; and
Figure FDA0002253496450000102
(3) deprotecting a compound of formula IM-2 to produce a compound of formula (I);
Figure FDA0002253496450000103
wherein LG is1And LG2Each independently represents a halogen leaving group, or C optionally substituted by halogen1-6An alkylsulfonate leaving group (e.g., a trifluoromethanesulfonate leaving group);
Rcand RdEach independently represents H or a leaving group;
PG1a protective group representing a hydroxyl group (e.g., methyl group, t-butyldimethylsilyl group, triisopropylsilyl group, benzyl group, and methoxymethyl group);
the remaining groups are as defined in any one of claims 1 to 20.
CN201911043519.7A 2018-11-08 2019-10-30 Substituted pyridine compound, preparation method and application thereof Active CN111153899B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201811324879 2018-11-08
CN201811324879X 2018-11-08

Publications (2)

Publication Number Publication Date
CN111153899A true CN111153899A (en) 2020-05-15
CN111153899B CN111153899B (en) 2023-12-01

Family

ID=70555899

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201911043519.7A Active CN111153899B (en) 2018-11-08 2019-10-30 Substituted pyridine compound, preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN111153899B (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112194652A (en) * 2020-11-30 2021-01-08 北京华氏开元医药科技有限公司 4-hydroxymethyl-1H-indole compound, preparation method and application
CN112402385A (en) * 2020-11-30 2021-02-26 北京华氏开元医药科技有限公司 4-hydroxymethyl-1H-indole compound pharmaceutical preparation and preparation method thereof
US10934302B1 (en) 2018-03-21 2021-03-02 Relay Therapeutics, Inc. SHP2 phosphatase inhibitors and methods of use thereof
WO2021218752A1 (en) * 2020-04-26 2021-11-04 Betta Pharmaceuticals Co., Ltd Shp2 inhibitors, compositions and uses thereof
WO2022042331A1 (en) * 2020-08-25 2022-03-03 四川科伦博泰生物医药股份有限公司 Heterocyclic compound, and preparation method therefor and use thereof
WO2022237676A1 (en) * 2021-05-12 2022-11-17 药雅科技(上海)有限公司 Preparation and application of shp2 phosphatase inhibitor
US11529347B2 (en) 2016-09-22 2022-12-20 Relay Therapeutics, Inc. SHP2 phosphatase inhibitors and methods of use thereof
US11591336B2 (en) 2017-05-26 2023-02-28 D. E. Shaw Research, Llc Substituted pyrazolo[3,4-b]pyrazines as SHP2 phosphatase inhibitors
CN115960109A (en) * 2021-05-31 2023-04-14 药雅科技(上海)有限公司 Preparation and application of fused-ring SHP2 phosphatase inhibitor
US11629145B2 (en) 2016-10-24 2023-04-18 D. E. Shaw Research, Llc SHP2 phosphatase inhibitors and methods of use thereof
US11701354B2 (en) 2017-09-29 2023-07-18 D. E. Shaw Research, Llc Pyrazolo[3,4-b]pyrazine derivatives as SHP2 phosphatase inhibitors
US11890281B2 (en) 2019-09-24 2024-02-06 Relay Therapeutics, Inc. SHP2 phosphatase inhibitors and methods of making and using the same

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105899493A (en) * 2014-01-17 2016-08-24 诺华股份有限公司 1 -(triazin-3-yi_/pyridazin-3-yl)-piper(-azine)idine derivatives and compositions thereof for inhibiting the activity of SHP2
CN105899491A (en) * 2014-01-17 2016-08-24 诺华股份有限公司 1 -pyridazin-/triazin-3-yl-piper(-azine)/idine/pyrolidine derivatives and and compositions thereof for inhibiting the activity of SHP2
CN105916845A (en) * 2014-01-17 2016-08-31 诺华股份有限公司 N-azaspirocycloalkane substituted n-heteroaryl compounds and compositions for inhibiting the activity of shp2
WO2016203404A1 (en) * 2015-06-19 2016-12-22 Novartis Ag Compounds and compositions for inhibiting the activity of shp2
WO2018057884A1 (en) * 2016-09-22 2018-03-29 Relay Therapeutics, Inc. Shp2 phosphatase inhibitors and methods of use thereof
WO2018130928A1 (en) * 2017-01-10 2018-07-19 Novartis Ag Pharmaceutical combination comprising an alk inhibitor and a shp2 inhibitor
WO2018136265A1 (en) * 2017-01-23 2018-07-26 Revolution Medicines, Inc. Bicyclic compounds as allosteric shp2 inhibitors

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105899493A (en) * 2014-01-17 2016-08-24 诺华股份有限公司 1 -(triazin-3-yi_/pyridazin-3-yl)-piper(-azine)idine derivatives and compositions thereof for inhibiting the activity of SHP2
CN105899491A (en) * 2014-01-17 2016-08-24 诺华股份有限公司 1 -pyridazin-/triazin-3-yl-piper(-azine)/idine/pyrolidine derivatives and and compositions thereof for inhibiting the activity of SHP2
CN105916845A (en) * 2014-01-17 2016-08-31 诺华股份有限公司 N-azaspirocycloalkane substituted n-heteroaryl compounds and compositions for inhibiting the activity of shp2
WO2016203404A1 (en) * 2015-06-19 2016-12-22 Novartis Ag Compounds and compositions for inhibiting the activity of shp2
WO2018057884A1 (en) * 2016-09-22 2018-03-29 Relay Therapeutics, Inc. Shp2 phosphatase inhibitors and methods of use thereof
WO2018130928A1 (en) * 2017-01-10 2018-07-19 Novartis Ag Pharmaceutical combination comprising an alk inhibitor and a shp2 inhibitor
WO2018136265A1 (en) * 2017-01-23 2018-07-26 Revolution Medicines, Inc. Bicyclic compounds as allosteric shp2 inhibitors

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11529347B2 (en) 2016-09-22 2022-12-20 Relay Therapeutics, Inc. SHP2 phosphatase inhibitors and methods of use thereof
US11629145B2 (en) 2016-10-24 2023-04-18 D. E. Shaw Research, Llc SHP2 phosphatase inhibitors and methods of use thereof
US11591336B2 (en) 2017-05-26 2023-02-28 D. E. Shaw Research, Llc Substituted pyrazolo[3,4-b]pyrazines as SHP2 phosphatase inhibitors
US11701354B2 (en) 2017-09-29 2023-07-18 D. E. Shaw Research, Llc Pyrazolo[3,4-b]pyrazine derivatives as SHP2 phosphatase inhibitors
US10934302B1 (en) 2018-03-21 2021-03-02 Relay Therapeutics, Inc. SHP2 phosphatase inhibitors and methods of use thereof
US11890281B2 (en) 2019-09-24 2024-02-06 Relay Therapeutics, Inc. SHP2 phosphatase inhibitors and methods of making and using the same
WO2021218752A1 (en) * 2020-04-26 2021-11-04 Betta Pharmaceuticals Co., Ltd Shp2 inhibitors, compositions and uses thereof
WO2022042331A1 (en) * 2020-08-25 2022-03-03 四川科伦博泰生物医药股份有限公司 Heterocyclic compound, and preparation method therefor and use thereof
CN112402385A (en) * 2020-11-30 2021-02-26 北京华氏开元医药科技有限公司 4-hydroxymethyl-1H-indole compound pharmaceutical preparation and preparation method thereof
CN112402385B (en) * 2020-11-30 2022-04-01 北京华氏开元医药科技有限公司 4-hydroxymethyl-1H-indole compound pharmaceutical preparation and preparation method thereof
CN112194652A (en) * 2020-11-30 2021-01-08 北京华氏开元医药科技有限公司 4-hydroxymethyl-1H-indole compound, preparation method and application
WO2022237676A1 (en) * 2021-05-12 2022-11-17 药雅科技(上海)有限公司 Preparation and application of shp2 phosphatase inhibitor
CN115960109A (en) * 2021-05-31 2023-04-14 药雅科技(上海)有限公司 Preparation and application of fused-ring SHP2 phosphatase inhibitor

Also Published As

Publication number Publication date
CN111153899B (en) 2023-12-01

Similar Documents

Publication Publication Date Title
CN111153899B (en) Substituted pyridine compound, preparation method and application thereof
JP7089061B2 (en) 2-Amino-pyridine or 2-amino-pyrimidine derivative as a cyclin-dependent kinase inhibitor
AU2017311645B2 (en) Pyridopyrimdinone CDK2/4/6 inhibitors
CN114127053B (en) Substituted pyrazine compound, preparation method and application thereof
EP2857404B1 (en) IMIDAZO[1,2-b]PYRIDAZINE DERIVATIVES AS KINASE INHIBITORS
CA3139769A1 (en) Nitrogen-containing heterocyclic derivative regulator, preparation method therefor and application thereof
KR20130129244A (en) Substituted 6,6-fused nitrogenous heterocyclic compounds and uses thereof
RU2742485C2 (en) Heterocyclic compound used as a fgfr inhibitor
CN111484491B (en) Substituted pyrido-cyclic compounds, process for their preparation and their use
CN114981268A (en) Pyrimidine-4 (3H) -ketone heterocyclic compound, preparation method and application thereof in medicine and pharmacology
AU2017389818B2 (en) Quinazoline compound and preparation method, application, and pharmaceutical compostion thereof
KR20220028072A (en) Heterocyclic compounds as BET inhibitors
CN113330009B (en) Azacyclic compounds, preparation method and application thereof
JP2020530020A (en) A novel heterocyclic compound as a CDK8 / 19 inhibitor
CN116113418A (en) Heterocyclic compounds, their preparation and use
WO2022161222A1 (en) Heterocyclic shp2 inhibitor, preparation method therefor, and use thereof
CN113387962A (en) Pyrazolo [3,4-d ] pyrimidine-3-one derivative, pharmaceutical composition and application thereof
KR20200135827A (en) Imidazolidin-2-one derivatives substituted as PRMT5 inhibitors
CN112752758B (en) Imidazo [4,5-c ] cinnolin-2-one compounds as selective modulators of ATM kinase and uses thereof
EP3012259A1 (en) Five-membered heterocyclic pyridine compounds and preparation method and use thereof
CN111848605B (en) Substituted pyrido [3,4-b ] pyrazin-2 (1H) -one compound, preparation method and application thereof
WO2021057867A1 (en) Class of cdk inhibitor based on organic arsine, preparation method and application thereof
CA3213823A1 (en) Selective modulators of ataxia telangiectasia mutated (atm) kinase and uses thereof
US20230399332A1 (en) IMIDAZO[1,2-a]PYRAZINE OR PYRAZOLO[1,5-a]PYRIMIDINE DERIVATIVE AND USE THEREOF
WO2023250083A1 (en) Small molecule inhibitors of dyrk/clk and uses 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