CN113845531A - Pyrazolo ring compound, pharmaceutical composition containing same, preparation method and application thereof - Google Patents

Pyrazolo ring compound, pharmaceutical composition containing same, preparation method and application thereof Download PDF

Info

Publication number
CN113845531A
CN113845531A CN202010597780.8A CN202010597780A CN113845531A CN 113845531 A CN113845531 A CN 113845531A CN 202010597780 A CN202010597780 A CN 202010597780A CN 113845531 A CN113845531 A CN 113845531A
Authority
CN
China
Prior art keywords
radical
cycloalkyl
alkyl
aryl
optionally substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202010597780.8A
Other languages
Chinese (zh)
Inventor
贾兰齐
游泽金
孙晓阳
冉茂盛
陈泉龙
朱军
李伟
王太津
田强
宋宏梅
薛彤彤
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
Priority to CN202010597780.8A priority Critical patent/CN113845531A/en
Publication of CN113845531A publication Critical patent/CN113845531A/en
Pending legal-status Critical Current

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • 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

Description

Pyrazolo ring compound, pharmaceutical composition containing same, preparation method and application thereof
Technical Field
The invention belongs to the technical field of medicines, and relates to a pyrazolo ring compound, a pharmaceutical composition containing the pyrazolo ring compound, a preparation method of the pyrazolo ring compound, and application of the pyrazolo ring compound in preparation of medicines for treating HPK 1-related diseases.
Background
Hematopoietic progenitor protein kinase 1 (HPK 1), also known as mitogen-activated protein kinase kinase kinase kinase kinase kinase 1 (MAP 4K1), is a member of the Ste-20 silk/threonine family, which family members also include MAP4K2(GCK), MAP4K3(GLK), MAP4K4(HGK), MAP4K5(KHS) and MAP4K6 (MINK). HPK1 is primarily expressed restrictively in hematopoietic cells, including early progenitor cells, as well as hematopoietic cell lines, such as human acute lymphoblastic leukemia cells (MOLT4), human lymphoma cells (Daudi), and the like. Under physiological conditions, HPK1 functions as peripheral immune tolerance via the JNK/SAPK signaling pathway. However, in certain tumor progression, HPK1 down-regulates T cell, B cell responses, and activation of dendritic cells. When a T Cell Receptor (TCR) binds to a ligand (CD3/28), HPK1 is activated by a TCR signal path, and the activated HPK1 can phosphorylate serine 376 of a joint protein SLP-76, so that 14-3-3 protein is recruited to cause instability of a TCR complex, and finally, T cell response is weakened and T cell proliferation is inhibited. Therefore, it has been increasingly appreciated that the inhibition of the immune down-regulation of HPK1 in tumor therapy exerts an anti-tumor effect on the immune system of the patient himself. It has been shown that the antigen presentation of mouse dendritic cells, which had been specifically depleted in HPK1 enzyme activity, was enhanced (Alzabin, Bhardwaj et al 2009). Similarly, lewis lung carcinoma transplanted tumor mice had significantly increased tumor suppression rates after receiving HPK1 knockout T cell transplantation therapy compared to the control group (wild type T cells) (Alzabin, pyrajan et al 2010). Therefore, a small-molecule inhibitor of HPK1 is searched as a new direction for tumor immunotherapy.
Disclosure of Invention
The present invention provides novel, highly active HPK1 inhibitor compounds that can be used alone or in combination with other drugs for the treatment of neoplastic diseases.
A first aspect of the present invention relates to a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof:
Figure BDA0002557649670000011
wherein the content of the first and second substances,
R1selected from hydrogen, C (O) Ra、CO2Ra、C(O)NRbRc、S(O)Ra、SO2Ra、SO2NRbRc、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 3-8 membered heterocyclyl and 5-14 membered heteroaryl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 3-8 membered heterocyclyl and 5-14 membered heteroaryl are each optionally substituted with one or more RdSubstitution;
R2selected from hydrogen, halogen, CN, NO2、C(O)Ra、CO2Ra、C(O)NRbRc、S(O)Ra、SO2Ra、SO2NRbRc、NRbRc、NHC(O)Ra、NHCO2Ra、OC(O)Ra、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C14Aryl and 5-14 membered heteroaryl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with one or more RdSubstitution;
R3selected from hydrogen, C (O) Ra、CO2Ra、C(O)NRbRc、S(O)Ra、SO2Ra、SO2NRbRc、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl, said C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-10 membered heterocyclyl fused ringsAnd C is1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 5-10 membered heterocyclyl are each optionally substituted with one or more RdSubstitution;
Raindependently selected from C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl, said C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 5-10 membered heterocyclyl are each optionally substituted with one or more RdSubstitution;
Rband RcEach independently selected from hydrogen and C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6alkoxy-C1-C6Alkyl and 4-7 membered heterocyclyl; or, RbAnd RcTogether with the nitrogen atom to which they are attached form a 4-7 membered heterocyclyl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6alkoxy-C1-C6Each of alkyl and 4-7 membered heterocyclyl is optionally substituted with one or more RdSubstitution;
x is selected from CH and N;
Rdindependently selected from: hydrogen, hydroxy, halogen, oxo, CN, NO2、C(O)Re、CO2Re、NRfSO2Rg、S(O)Re、SO2Re、C(O)NRfRg、SO2NRfRg、NRfRg、NRfC(O)Rg、C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C1-C6alkoxy-C1-C6Alkoxy radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl, said C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C1-C6alkoxy-C1-C6Alkoxy radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl are each optionally substituted with one or more RjSubstitution;
Reindependently selected from hydrogen, C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl;
Rfand RgEach independently selected from hydrogen and C1-C6Alkyl radical, C3-C7Cycloalkyl and 4-7 membered heterocyclyl, wherein said C is1-C6Alkyl is optionally substituted by C1-C6Alkoxy or NRhRiSubstitution; or, RbAnd RcTogether with the nitrogen atom to which they are attached form a 4-7 membered heterocyclyl;
Rhand RiEach independently selected from hydrogen and C1-C6An alkyl group; and is
RjIndependently selected from halogen, hydroxy, CN, C (O) Re、NRfRg、C(O)NRbRe、NRbC(O)ReAnd 3-8 membered heterocyclic groups.
Another aspect of the present invention provides a pharmaceutical composition comprising a compound of the present invention, or a stereoisomer, tautomer, pharmaceutically acceptable salt, polymorph, co-crystal, solvate, metabolite, prodrug or any mixture of two or more thereof, together with one or more pharmaceutically acceptable carriers.
Another aspect of the invention provides a kit comprising:
1) a compound of the invention or a stereoisomer, tautomer, pharmaceutically acceptable salt, polymorph, co-crystal, solvate, metabolite, prodrug or any mixture of two or more thereof; and
2) optionally packaging and/or instructions.
Another aspect of the invention provides the use of a compound of the invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof, or a pharmaceutical composition of the invention, for the manufacture of a medicament for the treatment of a disease associated with HPK 1.
Another aspect of the invention provides a compound of the invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof, or a pharmaceutical composition of the invention or a kit of the invention, for use in the treatment of a disease associated with HPK 1.
Another aspect of the present invention provides a method of treating a disease associated with HPK1, comprising administering to a subject in need thereof an effective amount of a compound of the present invention, or a stereoisomer, tautomer, pharmaceutically acceptable salt, polymorph, co-crystal, solvate, metabolite or prodrug thereof, or any mixture of two or more thereof, or a pharmaceutical composition of the present invention, and optionally comprising co-administering other agents for treating a disease or disorder associated with HPK 1.
Another aspect of the present invention provides a method for preparing the compound of the present invention, which is carried out according to the following reaction scheme 1 or 2:
scheme 1:
Figure BDA0002557649670000031
wherein R is1、R2、R3X is as defined above;
the first step is as follows: the compound I-1 is subjected to coupling, condensation or alkylation reaction to form a compound I-2, wherein LG1Is a leaving group such as halogen (e.g., Cl, Br or I), OTs, OTf, etc.;
the second step is that: carrying out coupling or nucleophilic substitution reaction on the compound I-2 to generate a compound I-3, wherein PG is an amino protecting group, such as Boc, Cbz, Bn, PMB and the like;
the third step: carrying out deprotection reaction on the compound I-3 to generate a compound I-4;
the fourth step: the compound I-4 is subjected to coupling, condensation or alkylation reaction to generate the target compound I.
Scheme 2:
Figure BDA0002557649670000032
wherein R is1、R2、R3X is as defined above;
the first step is as follows: the compound I-1 is subjected to coupling, condensation or alkylation reaction to form a compound I-2, wherein LG1Is a leaving group such as halogen (e.g., Cl, Br or I), OTs, OTf, etc.;
the second step is that: the compound I-2 is subjected to coupling or nucleophilic substitution reaction to generate the target compound I.
Compounds and methods of preparation
It is an object of the present invention to provide a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof:
Figure BDA0002557649670000033
wherein the content of the first and second substances,
R1selected from hydrogen, C (O) Ra、CO2Ra、C(O)NRbRc、S(O)Ra、SO2Ra、SO2NRbRc、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 3-8 membered heterocyclyl and 5-14 membered heteroaryl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 3-8 membered heterocyclyl and 5-14 membered heteroaryl are each optionally substituted with one or more RdSubstitution;
R2selected from hydrogen, halogen, CN, NO2、C(O)Ra、CO2Ra、C(O)NRbRc、S(O)Ra、SO2Ra、SO2NRbRc、NRbRc、NHC(O)Ra、NHCO2Ra、OC(O)Ra、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C14Aryl and 5-14 membered heteroaryl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with one or more RdSubstitution;
R3selected from hydrogen, C (O) Ra、CO2Ra、C(O)NRbRc、S(O)Ra、SO2Ra、SO2NRbRc、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl, said C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 5-10 membered heterocyclyl are each optionally substituted with one or more RdSubstitution;
Raindependently selected from C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl, said C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 5-10 membered heterocyclyl are each optionally substituted with one or more RdSubstitution;
Rband RcEach independently selected from hydrogen and C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical C1-C6Alkyl and 4-7 membered heterocyclyl; or, RbAnd RcTogether with the nitrogen atom to which they are attached form a 4-7 membered heterocyclyl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical C1-C6Each of alkyl and 4-7 membered heterocyclyl is optionally substituted with one or more RdSubstitution;
x is selected from CH and N;
Rdindependently selected from: hydrogen, hydroxy, halogen, oxo, CN, NO2、C(O)Re、CO2Re、NRfSO2Rg、S(O)Re、SO2Re、C(O)NRfRg、SO2NRfRg、NRfRg、NRfC(O)Rg、C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical C1-C6Alkoxy radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl, said C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C1-C6alkoxy-C1-C6Alkoxy radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl are each optionally substituted with one or more RjSubstitution;
Reindependently selected from hydrogen, C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl;
Rfand RgEach independently selected from hydrogen and C1-C6Alkyl radical, C3-C7Cycloalkyl and 4-7 membered heterocyclyl, wherein said C is1-C6Alkyl is optionally substituted by C1-C6Alkoxy or NRhRiSubstitution; or, RbAnd RcTogether with the nitrogen atom to which they are attached form a 4-7 membered heterocyclyl;
Rhand RiEach independently selected from hydrogen and C1-C6An alkyl group; and is
RjIndependently selected from halogen, hydroxy, CN, C (O) Re、NRfRg、C(O)NRbRe、NRbC(O)ReAnd 3-8 membered heterocyclic groups.
According to some embodiments of the invention, R1Selected from hydrogen, C (O) Ra、CO2Ra、C(O)NRbRc、S(O)Ra、SO2Ra、SO2NRbRc、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl, 3-8 membered heterocyclyl and 5-10 membered heteroaryl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl, 3-8 membered heterocyclyl and 5-10 membered heteroaryl are each optionally substituted with one or more RdAnd (4) substitution.
In some embodiments of the invention, R1Selected from hydrogen, C (O) Ra、C1-C6Alkyl radical, C3-C7Cycloalkyl and 3-8 membered heterocyclyl, said C1-C6Alkyl radical, C3-C7Cycloalkyl and 3-8 membered heterocyclyl are each optionally substituted with one or more RdAnd (4) substitution.
In some embodiments of the invention, R1Selected from hydrogen, C1-C6Alkyl radical, C3-C7Cycloalkyl radicalsAnd 3-8 membered heterocyclic group, said C1-C6Alkyl radical, C3-C7Cycloalkyl and 3-8 membered heterocyclyl are each optionally substituted with RdAnd (4) substitution.
In some embodiments of the invention, R1Is selected from C1-C6Alkyl and 3-8 membered heterocyclic group, said C1-C6Alkyl and 3-8 membered heterocyclyl are each optionally substituted with RdAnd (4) substitution.
In some embodiments of the invention, R1Is selected from C1-C6Alkyl and 5-6 membered azacycloalkyl, said C1-C6Alkyl is optionally substituted with a group selected from: hydroxy, C1-C6Alkoxy, halogen, and 5-6 membered heterocyclyl.
In some embodiments of the invention, R1Is selected from C1-C6Alkyl and pyrrolidinyl, said C1-C6Alkyl is optionally substituted with a group selected from: hydroxy, C1-C6Alkoxy, halogen and morpholinyl.
In some embodiments of the invention, R1Is selected from C1-C6Alkyl and pyrrolidinyl, said C1-C6Alkyl is optionally substituted with a group selected from: hydroxy, methoxy, fluoro and morpholinyl.
In some embodiments of the invention, R1Selected from the group consisting of methyl, ethyl, isopropyl, isobutyl, sec-butyl, morpholin-2-ylmethyl, 2-hydroxyethyl, 2-methoxyethyl, 2-fluoroethyl and pyrrolidinyl.
In some embodiments of the invention, R1Selected from the group consisting of isopropyl, sec-butyl, morpholin-2-ylmethyl, 2-hydroxyethyl, 2-methoxyethyl, 2-fluoroethyl and pyrrolidinyl.
According to some embodiments of the invention, R2Selected from hydrogen, halogen, CN, NO2、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C10Aryl and 5-to 10-membered heteroaryl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted with one or more RdAnd (4) substitution.
In some embodiments of the invention, R2Selected from hydrogen, halogen, C1-C6Alkyl and C3-C7Cycloalkyl radical, said C1-C6Alkyl and C3-C7Cycloalkyl is each optionally substituted by one or more RdAnd (4) substitution.
In some embodiments of the invention, R2Selected from hydrogen, halogen and C1-C6Alkyl radical, said C1-C6Alkyl is optionally substituted by one or more RdAnd (4) substitution.
In some embodiments of the invention, R2Is hydrogen.
According to some embodiments of the invention, R3Selected from hydrogen, C (O) Ra、CO2Ra、C(O)NRbRc、S(O)Ra、SO2Ra、SO2NRbRc、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl and 5-to 10-membered heteroaryl, said C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl, 5-10 membered heteroaryl and 5-10 membered heterocyclyl are each optionally substituted with one or more RdAnd (4) substitution.
In some embodiments of the invention, R3Selected from C (O) Ra、CO2Ra、C(O)NRbRc、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl and 5-to 10-membered heteroaryl, said C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-10 membered heterocyclyl fused ringsAnd C is1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl, 5-10 membered heteroaryl and 5-10 membered heterocyclyl are each optionally substituted with one or more RdAnd (4) substitution.
In some embodiments of the invention, R3Selected from C (O) Ra、CO2Ra、C6-C10Aryl and 5-to 10-membered heteroaryl, said C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C6-C10Aryl, 5-10 membered heteroaryl, C3-C7Cycloalkyl and 5-10 membered heterocyclyl are each optionally substituted with 1,2,3 or 4RdAnd (4) substitution.
In some embodiments of the invention, R3Selected from C (O) Ra、C6-C10Aryl and 5-to 10-membered heteroaryl, said C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C6-C10Aryl, 5-10 membered heteroaryl, C3-C7Cycloalkyl and 5-10 membered heterocyclyl are each optionally substituted with 1,2,3 or 4RdAnd (4) substitution.
In some embodiments of the invention, R3Selected from C (O) RaPhenyl and 5-9 membered heteroaryl, each of which is optionally substituted with C5-C6Cycloalkyl or 5-9 membered heterocyclyl fused and said phenyl, 5-9 membered heteroaryl, C5-C6Cycloalkyl and 5-9 membered heterocyclyl are each optionally substituted with 1,2,3 or 4RdAnd (4) substitution.
In some embodiments of the invention, R3Selected from C (O) RaPhenyl and 5-9 membered heteroaryl, each of which is optionally substituted with C5-C6Cycloalkyl or 5-9 membered heterocyclyl fused and said phenyl, 5-9 membered heteroaryl, C5-C6Cycloalkyl and 5-9 membered heterocyclyl are each optionally substituted with 1,2,3 or 4RdSubstitution;
Raselected from the group consisting of cyclopropyl, cyclobutyl, isopropyl, pyrazolyl, indazolyl, indolyl and
Figure BDA0002557649670000051
the cyclopropyl, pyrazolyl, indazolyl, indolyl and
Figure BDA0002557649670000052
optionally substituted with 1,2,3 or 4 fluoro or methyl groups;
Rdindependently selected from: fluoro, vinyl, pyrrolidinyl, ethyl, N-propyl, isopropyl, oxo, methyl, methoxy, -N (CH)3)2、-NH(CH2)2-NH(CH3) Isobutyl, morpholinyl, piperidinyl, - (CH)2)2N(CH3)2And amino, said vinyl, methyl, ethyl, n-propyl, pyrrolidinyl being optionally substituted with one or more hydroxy, fluoro, CN, acetyl or morpholinyl.
In some embodiments of the invention, R3Selected from pyrazolyl, pyridyl, pyrimidinyl, indolyl, indazolyl or pyrimidopyrazolyl, optionally substituted by 1 or 2RdSubstitution; and R isdIndependently selected from oxo, amino, C1-C6Alkyl and pyrrolidinyl, said C1-C6Alkyl and pyrrolidinyl are each optionally substituted with hydroxy, CN or acetyl.
In some embodiments of the present invention, the first and second substrates are,
R1is selected from C1-C6Alkyl and pyrrolidinyl, said C1-C6Alkyl is optionally substituted with a group selected from: hydroxy, C1-C6Alkoxy, halogen and morpholinyl;
R2is hydrogen;
R3is selected from
Figure BDA0002557649670000061
Figure BDA0002557649670000062
Figure BDA0002557649670000063
The above-mentioned
Figure BDA0002557649670000064
Figure BDA0002557649670000065
Figure BDA0002557649670000066
Optionally substituted by 1 or 2 or 3 or 4RdSubstitution;
Rdindependently selected from fluoro, oxo, amino, methyl, ethyl, N-propyl, isopropyl, isobutyl, methoxy, -N (CH)3)2、-NH(CH2)2NH2、C2-C6Alkenyl, morpholinyl, piperidinyl and pyrrolidinyl, said methyl, ethyl, n-propyl, isopropyl, -NH (CH)2)2NH2、C2-C6Alkenyl and pyrrolidinyl are each optionally substituted with one or more groups selected from hydroxy, methyl, halo, morpholinyl, -N (CH)3)2CN or acetyl; and is
X is selected from CH and N.
In some embodiments of the present invention, the first and second substrates are,
R1selected from isopropyl, sec-butyl, morpholin-2-ylmethyl, 2-hydroxyethyl, 2-methoxyethyl, 2-fluoroethyl and pyrrolidinyl;
R2is hydrogen;
R3is selected from
Figure BDA0002557649670000067
Figure BDA0002557649670000068
Figure BDA0002557649670000071
And the number of the first and second electrodes,
x is selected from CH and N.
The present invention encompasses compounds of formula I obtained by any combination of the above preferred groups.
According to some embodiments of the invention, the compounds of the invention have the structure of formula II:
Figure BDA0002557649670000072
wherein the content of the first and second substances,
R1、R2x and RaAs defined above.
In some embodiments of the invention, RaIs selected from C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl, said C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 5-10 membered heterocyclyl are each optionally substituted with one or more RdIs substituted in which RdAs defined above.
In some embodiments of the invention, RaIs selected from C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl and 5-to 10-membered heteroaryl, said C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted by 1 or 2RdIs substituted in which RdAs defined above.
In the inventionIn some embodiments, RaIs selected from C1-C6Alkyl radical, C3-C7Cycloalkyl and 5-10 membered heteroaryl, said 5-10 membered heteroaryl being optionally fused with a 5-6 membered heterocyclyl, and said C1-C6Alkyl radical, C3-C7Cycloalkyl, 5-10 membered heteroaryl and 5-6 membered heterocyclyl are each optionally substituted with 1 or 2 substituents independently selected from halogen and C1-C6Alkyl groups.
In some embodiments of the invention, RaIs selected from C1-C6Alkyl radical, C3-C7Cycloalkyl, pyrrolyl, pyrazolyl and imidazolyl each optionally fused to a benzene or tetrahydropyrimidine ring, and C1-C6Alkyl radical, C3-C7Cycloalkyl, pyrrolyl, pyrazolyl, imidazolyl, phenyl and tetrahydropyrimidine ring each optionally substituted by 1 or 2 substituents independently selected from halogen and C1-C6Alkyl groups.
In some embodiments of the invention, RaSelected from cyclopropyl, fluorocyclopropyl, difluorocyclopropyl, cyclobutyl, isopropyl, pyrazolyl, methyl pyrazolyl, indazolyl, indolyl and tetrahydropyrimidizolyl.
In some embodiments of the present invention, the first and second substrates are,
R1is selected from C1-C6Alkyl and pyrrolidinyl, said C1-C6Alkyl is optionally substituted with a group selected from: hydroxy, C1-C6Alkoxy, halogen and morpholinyl;
R2is hydrogen;
Rais selected from C1-C6Alkyl radical, C3-C7Cycloalkyl, pyrrolyl, pyrazolyl and imidazolyl each optionally fused to a benzene or tetrahydropyrimidine ring, and C1-C6Alkyl radical, C3-C7Cycloalkyl, pyrrolyl, pyrazolyl, imidazolyl, phenyl and tetrahydropyrimidine rings are each optionally substituted by 1 or 2 substituents independently selected from halogenAnd C1-C6Radical substitution of alkyl; and is
X is CH.
In some embodiments of the present invention, the first and second substrates are,
R1selected from isopropyl, sec-butyl, morpholin-2-ylmethyl, 2-hydroxyethyl, 2-methoxyethyl, 2-fluoroethyl and pyrrolidinyl;
R2is hydrogen;
Raselected from cyclopropyl, fluorocyclopropyl, difluorocyclopropyl, cyclobutyl, isopropyl, pyrazolyl, methyl pyrazolyl, indazolyl, indolyl and tetrahydropyrimidizolyl pyrazolyl; and is
X is CH.
According to some embodiments of the invention, the compounds of the invention have the structure of formula III:
Figure BDA0002557649670000081
wherein the content of the first and second substances,
R4selected from hydrogen, C (O) Re、CO2Re、C(O)NRfRg、S(O)Re、SO2Re、SO2NRfRg、C1-C6Alkyl radical, C2-C6Alkenyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl, said C1-C6Alkyl radical, C2-C6Alkenyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with 1 or 2RjSubstitution;
R5selected from hydrogen, NRfRg、C1-C6Alkyl radical, C3-C7Cycloalkyl, 3-8 membered heterocyclyl, C6-C14Aryl and 5-14 membered heteroaryl, said C1-C6Alkyl radical, C3-C7Cycloalkyl, 3-8 membered heterocyclyl, C6-C14Aryl and 5-14 membered heteroaryl are each optionallyGround is 1 or 2RjSubstitution; and is
R1、R2、X、Rf、Rg、ReAnd RjAs defined above.
In some embodiments of the invention, R4Is selected from C1-C6Alkyl and C2-C6Alkenyl radical, said C1-C6Alkyl and C2-C6Each alkenyl group is optionally substituted by 1 or 2 substituents independently selected from halogen, NRfRgAnd 5-6 membered heterocyclyl.
In some embodiments of the invention, R4Is selected from C1-C6Alkyl and C2-C6Alkenyl radical, said C1-C6Alkyl and C2-C6Each alkenyl group is optionally substituted with 1 or 2 substituents independently selected from halogen, N (C)1-C6Alkyl radical)2And morpholinyl.
In some embodiments of the invention, R4Selected from 2-fluorovinyl, 2-difluoroethyl, isopropyl, isobutyl, and mixtures thereof,
Figure BDA0002557649670000091
Figure BDA0002557649670000092
And 2- (N, N-dimethylamino) ethyl.
In some embodiments of the present invention, the first and second substrates are,
R5selected from hydrogen, NRfRgAnd 5-6 membered heterocyclyl;
Rfand RgEach independently selected from hydrogen and C1-C6Alkyl radical, wherein said C1-C6Alkyl is optionally substituted by NRhRiSubstitution; and is
RhAnd RiEach independently selected from hydrogen and C1-C6An alkyl group.
In some embodiments of the present invention, the first and second substrates are,
R5selected from hydrogen,NRfRgPyrrolidinyl, morpholinyl, and piperidinyl;
Rfand RgEach independently selected from hydrogen and C1-C6Alkyl radical, wherein said C1-C6Alkyl is optionally substituted by NRhRiSubstitution; and;
Rhand RiEach independently selected from hydrogen and C1-C6An alkyl group.
In some embodiments of the invention, R5Selected from the group consisting of hydrogen, pyrrolidinyl, morpholinyl, piperidinyl, 2- (N-methylamino) ethylamino, and 2- (N, N-dimethylamino) ethylamino.
In some embodiments of the present invention, the first and second substrates are,
R1is C1-C6An alkyl group;
R2is hydrogen;
R4is selected from C1-C6Alkyl and C2-C6Alkenyl radical, said C1-C6Alkyl and C2-C6Each alkenyl group is optionally substituted with 1 or 2 substituents independently selected from halogen, N (C)1-C6Alkyl radical)2And morpholinyl;
R5selected from hydrogen, NRfRgPyrrolidinyl, morpholinyl, and piperidinyl;
Rfand RgEach independently selected from hydrogen and C1-C6Alkyl radical, wherein said C1-C6Alkyl is optionally substituted by NRhRiSubstitution;
Rhand RiEach independently selected from hydrogen and C1-C6An alkyl group; and is
X is CH.
In some embodiments of the invention, R5Selected from the group consisting of hydrogen, pyrrolidinyl, morpholinyl, piperidinyl, 2- (N-methylamino) ethylamino, and 2- (N, N-dimethylamino) ethylamino.
In some embodiments of the present invention, the first and second substrates are,
R1is C1-C6An alkyl group;
R2is hydrogen;
R4selected from the group consisting of 2-fluorovinyl, 2-difluoroethyl, isopropyl, isobutyl, and mixtures thereof,
Figure BDA0002557649670000093
And 2- (N, N-dimethylamino) ethyl;
R5selected from hydrogen, pyrrolidinyl, morpholinyl, piperidinyl, 2- (N-methylamino) ethylamino, and 2- (N, N-dimethylamino) ethylamino; and the number of the first and second electrodes,
x is CH.
According to some embodiments of the invention, the compounds of the invention have the structure of formula IV:
Figure BDA0002557649670000101
wherein the content of the first and second substances,
R6and R7Each independently selected from hydrogen, halogen, CN, NO2、C(O)Re、CO2Re、C(O)NRfRg、S(O)Re、SO2Re、SO2NRfRg、NRfRg、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C14Aryl and 5-14 membered heteroaryl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with one or more RjSubstitution;
R8and R9Each independently selected from hydrogen, NRfRgAnd C1-C6An alkyl group; or, R8And R9Together with the carbon atom to which they are attached form C3-C7Cycloalkyl or carbonyl, said C1-C6Alkyl and C3-C7Cycloalkyl is each optionally substituted by one or more RjSubstitution;
y is selected from CH and N;
u and V are each independently selected from CRfRg、NReO, S (O) and SO2
m and n are each independently selected from 0, 1 and 2, and the sum of m and n is 0, 1 or 2; and is
R1、R2、X、Re、Rf、RgAnd RjAs defined above.
In some embodiments of the invention, R6And R7Each independently selected from hydrogen, halogen and C1-C6An alkoxy group.
In some embodiments of the invention, R6And R7Each independently selected from hydrogen, fluoro and methoxy.
In some embodiments of the invention, R8And R9Each independently selected from hydrogen and NRfRgWherein R isfAnd RgAs defined above; or, R8And R9Together with the carbon atom to which it is attached form a carbonyl group.
In some embodiments of the present invention, the first and second substrates are,
R8is hydrogen;
R9selected from hydrogen and NRfRg(ii) a And is
RfAnd RgEach independently is C1-C6An alkyl group;
or, R8And R9Together with the carbon atom to which it is attached form a carbonyl group.
In some embodiments of the present invention, the first and second substrates are,
R8is hydrogen; and is
R9Selected from hydrogen and N, N-dimethylamino;
or, R8And R9Together with the carbon atom to which it is attached form a carbonyl group.
In some embodiments of the invention, Y is CH.
In some embodiments of the present invention, the first and second substrates are,
u and V are each independently selected from CRfRgAnd NRe
ReIndependently selected from hydrogen and C1-C6An alkyl group; and is
RfAnd RgEach independently selected from hydrogen and C1-C6An alkyl group.
In some embodiments of the invention, U and V are each independently selected from CH2And N (C)1-C6Alkyl groups).
In some embodiments of the present invention, the first and second substrates are,
u is CH2(ii) a And V is selected from CH2And N (C)1-C6Alkyl groups).
In some embodiments of the present invention, the first and second substrates are,
u is CH2(ii) a And is
V is selected from CH2、NCH(CH3)2And N (CH)3)。
In some embodiments of the present invention, the first and second substrates are,
R1is C1-C6An alkyl group;
R2is hydrogen;
R6and R7Each independently selected from hydrogen, halogen and C1-C6An alkoxy group;
R8is hydrogen;
R9selected from hydrogen and NRfRg
RfAnd RgEach independently is C1-C6An alkyl group;
or, R8And R9Together with the carbon atom to which it is attached form a carbonyl group;
y is CH;
u is CH2
V is selected from CH2And N (C)1-C6Alkyl groups);
m and n are each independently selected from 0, 1 and 2, and the sum of m and n is 0, 1 or 2; and is
X is selected from CH and N.
In some embodiments of the present invention, the first and second substrates are,
R1is isopropyl;
R2is hydrogen;
R6and R7Each independently selected from hydrogen, fluoro and methoxy;
R8is hydrogen;
R9selected from hydrogen and NRfRg
RfAnd RgEach independently is methyl;
or, R8And R9Together with the carbon atom to which it is attached form a carbonyl group;
y is CH;
u is CH2
V is selected from CH2、NCH(CH3)2And N (CH)3);
m and n are each independently selected from 0, 1 and 2, and the sum of m and n is 0, 1 or 2; and is
X is selected from CH and N.
According to some embodiments of the invention, the compounds of the invention have the structure of formula V:
Figure BDA0002557649670000111
wherein the content of the first and second substances,
R10selected from hydrogen, C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl, said C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl are each optionally substituted with one or moreRjSubstitution;
R11、R12、R13、R14each independently selected from hydrogen, halogen, hydroxy, C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl, said C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl are each optionally substituted with one or more RjSubstitution; or, R11、R12Together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl group; or, R13、R14Together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl group; and is
R1、R2X and RjAs defined above.
In some embodiments of the invention, R10Is C1-C6An alkyl group.
In some embodiments of the invention, R10Selected from methyl and isopropyl.
In some embodiments of the invention, R11、R12、R13、R14Each independently selected from hydrogen and C1-C6An alkyl group; or, R11、R12Together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl group; or, R13、R14Together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl group.
In some embodiments of the invention, R11、R12、R13、R14Each independently selected from hydrogen and methyl; or, R11、R12Together with the carbon atom to which it is attached form a cyclopropyl group; or, R13、R14Together with the carbon atom to which it is attached, form a cyclopropyl group.
In some embodiments of the inventionIn, R11、R12Each independently selected from hydrogen and C1-C6Alkyl, and R13、R14Together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl group.
In some embodiments of the invention, R11、R12Each independently selected from hydrogen and methyl, and R13、R14Together with the carbon atom to which it is attached, form a cyclopropyl group.
In some embodiments of the present invention, the first and second substrates are,
R1is C1-C6An alkyl group;
R2is hydrogen;
R10is C1-C6An alkyl group;
R11、R12、R13、R14each independently selected from hydrogen and C1-C6An alkyl group; or, R11、R12Together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl group; or, R13、R14Together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl group; and is
X is CH.
In some embodiments of the present invention, the first and second substrates are,
R1is C1-C6An alkyl group;
R2is hydrogen;
R10is C1-C6An alkyl group;
R11、R12、R13、R14each independently selected from hydrogen and C1-C6An alkyl group; or, R11、R12Each independently selected from hydrogen and C1-C6Alkyl radical, R13、R14Together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl group; and is
X is CH.
In some embodiments of the present invention, the first and second substrates are,
R1is isopropyl;
R2is hydrogen;
R10selected from methyl and isopropyl;
R11、R12each independently is hydrogen, R13、R14Together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl group; and is
X is CH.
According to some embodiments of the invention, the compound of the invention is selected from:
Figure BDA0002557649670000121
Figure BDA0002557649670000131
Figure BDA0002557649670000141
another object of the present invention is to provide a method for preparing the compound of the present invention, which is carried out according to the following reaction scheme 1 or 2:
scheme 1:
Figure BDA0002557649670000142
wherein R is1、R2、R3X is as defined above;
the first step is as follows: the compound I-1 is subjected to coupling, condensation or alkylation reaction to form a compound I-2, wherein LG1Is a leaving group such as halogen (e.g., Cl, Br or I), OTs, OTf, etc.;
the second step is that: carrying out coupling or nucleophilic substitution reaction on the compound I-2 to generate a compound I-3, wherein PG is an N atom protecting group, such as Boc, Cbz, Bn, PMB and the like;
the third step: carrying out deprotection reaction on the compound I-3 to generate a compound I-4;
the fourth step: the compound I-4 is subjected to coupling, condensation or alkylation reaction to generate the target compound I.
Scheme 2:
Figure BDA0002557649670000143
wherein R is1、R2、R3X is as defined above;
the first step is as follows: the compound I-1 is subjected to coupling, condensation or alkylation reaction to form a compound I-2, wherein LG1Is a leaving group such as halogen (e.g., Cl, Br or I), OTs, OTf, etc.;
the second step is that: the compound I-2 is subjected to coupling or nucleophilic substitution reaction to generate the target compound I.
Pharmaceutical compositions and methods of treatment
It is another object of the present invention to provide a pharmaceutical composition comprising a compound of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof, together with one or more pharmaceutically acceptable carriers.
It is another 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 the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof, with one or more pharmaceutically acceptable carriers.
It is another object of the present invention to provide a pharmaceutical formulation comprising a compound of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof, or a pharmaceutical composition of the present invention.
It is another object of the present invention to provide a kit comprising:
1) a compound of the invention or a stereoisomer, tautomer, pharmaceutically acceptable salt, polymorph, co-crystal, solvate, metabolite, prodrug or any mixture of two or more thereof, or a pharmaceutical composition of the invention; and
2) optionally packaging and/or instructions.
Another object of the present invention is to provide the use of a compound of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof, or a pharmaceutical composition of the present invention, for the manufacture of a medicament for the treatment of a disease related to HPK 1. In particular, the disease is a tumor.
It is another object of the present invention to provide a compound of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof or a pharmaceutical composition of the present invention or a kit of the present invention for use in the treatment of a disease associated with HPK 1. In particular, the disease is a tumor.
It is another object of the present invention to provide a method of treating a disease associated with HPK1, comprising administering to a subject in need thereof an effective amount of a compound of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof, or a pharmaceutical composition of the present invention, and optionally comprising co-administering other agents for treating a disease or disorder associated with HPK 1. In particular, the disease is a tumor.
By "pharmaceutically acceptable carrier" in the context of the present invention is meant a diluent, adjuvant, excipient, or vehicle that is administered together with a therapeutic agent 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.
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. Examples of suitable pharmaceutically acceptable carriers are described in Remington's Pharmaceutical Sciences (1990).
The pharmaceutical compositions of the present invention may act systemically and/or locally. For this purpose, they may be administered by a suitable route, for example by injection, intravenous, intraarterial, subcutaneous, intraperitoneal, intramuscular or transdermal administration; or by oral, buccal, nasal, transmucosal, topical, in the form of ophthalmic preparations or by inhalation.
For these routes of administration, the pharmaceutical compositions of the present invention may be administered in suitable dosage forms.
The pharmaceutical compositions of the present invention may optionally be administered in combination with other agents that have at least some effect in the treatment of various diseases. In some embodiments, the present invention provides a combination preparation of a compound of the invention and an additional therapeutic agent for simultaneous, separate or sequential use in therapy.
The term "effective amount" as used herein refers to an amount of a compound that, when administered, will alleviate one or more symptoms of the condition being treated to some extent.
The dosing regimen may be adjusted to provide the best desired response. For example, a single bolus may be administered, 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. 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 of a compound of the invention administered will depend on the subject being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound, and the judgment of the prescribing physician. Generally, an effective dose is from about 0.0001 to about 50mg per kg body weight per day, e.g., from about 0.01 to about 10 mg/kg/day (single or divided administration). For a 70kg human, this may amount to about 0.007 mg/day to about 3500 mg/day, e.g., about 0.7 mg/day to about 700 mg/day. In some cases, dosage levels not higher than the lower limit of the aforesaid range may be sufficient, while in other cases still larger doses may be employed without causing any harmful side effects, provided that the larger dose is first divided into several smaller doses to be administered throughout the day.
The compound of the invention may be present in the pharmaceutical composition in an amount or amount of about 0.01mg to about 1000 mg.
As used herein, unless otherwise specified, the term "treating" means reversing, alleviating, inhibiting the progression of, or preventing such a disorder or condition, or one or more symptoms of such a disorder or condition, to which such term applies.
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.).
Definition of
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.
As used herein, the terms "comprises," "comprising," "has," "containing," or "involving," and other variations thereof herein, are inclusive or open-ended and do not exclude additional unrecited elements or method steps.
The term "alkyl" as used herein is defined as a straight or branched chain saturated aliphatic hydrocarbon group. In some embodiments, the alkyl group has 1 to 10 carbon atoms, e.g., 1 to 8 carbon atoms (C)1-C8Alkyl), 1 to 6 carbon atoms (C)1-C6Alkyl), 1 to 4 carbon atoms (C)1-C4Alkyl), 1 to 3 carbon atoms (C)1-C3Alkyl), 2 to 6 carbon atoms (C)2-C6Alkyl), 2 to 4 carbon atoms (C)2-C4Alkyl) or 3 to 4 carbon atoms (C)3-C4Alkyl groups). For example, as used herein, the term "C1-C6Alkyl "refers to a straight or branched chain group having 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, or n-hexyl). In some embodiments, the alkyl group is optionally substituted with one or more (such as 1 to 3) suitable substituents such as halogen (when this group is referred to as "haloalkyl", e.g., -CF)3、-C2F5、-CHF2、-CH2F、-CH2CF3、-CH2Cl or-CH2CH2CF3Etc.).
As used herein, the term "alkenyl" refers to a hydrocarbon group containing at least one C ═ C double bond. The alkenyl group may be a straight or branched chain alkenyl group and contains 2 to 15 carbon atoms. E.g. "C" herein2-6Alkenyl "is alkenyl containing 2 to 6 carbon atoms. Non-limiting examples of alkenyl groups include ethenyl, propenyl, n-butenyl, 3-methylbut-2-enyl, n-pentenyl, octenyl and decenyl. An alkenyl radical may be unsubstituted or substituted by one or more identical or different substituentsAnd (4) substitution.
As used herein, the term "cycloalkyl" refers to a saturated 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, etc.), optionally substituted with one or more (such as 1 to 3) suitable substituents. The cycloalkyl group has 3 to 15, such as 3 to 10 carbon atoms, 3 to 7 carbon atoms, 3 to 6 carbon atoms, 3 to 5 carbon atoms, 5 to 7 carbon atoms, 4 to 6 carbon atoms, or 5 to 6 carbon atoms, and the like. For example, as used herein, the term "C3-C7Cycloalkyl "refers to a saturated monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl) having 3 to 7 ring carbon atoms, optionally substituted with one or more (such as 1 to 3) suitable substituents, for example, methyl-substituted cyclopropyl.
As used herein, the term "aryl" refers to an all-carbon monocyclic or fused ring polycyclic aromatic group having a conjugated pi-electron system. For example, as used herein, the term "C6-C14Aryl "means an aromatic radical containing from 6 to 14 carbon atoms, the term" C6-C10Aryl "means an aromatic group containing 6 to 10 carbon atoms, such as phenyl or naphthyl. Aryl is optionally substituted with one or more (such as 1 to 3) suitable substituents (e.g. halogen, -OH, -CN, -NO)2、C1-C6Alkyl, etc.).
As used herein, the term "heteroaryl" refers to a monocyclic, bicyclic or tricyclic aromatic ring system having 5 to 14 ring atoms, in particular having 5,6, 7, 8, 9, 10, 11, 12, 13 or 14 ring atoms, in particular 1,2,3,4, 5,6, 7, 8, 9 or 10 carbon atoms, and which contains at least one heteroatom (e.g. oxygen, nitrogen or sulfur) which may be the same or different. And, the heteroaryl ring may be fused to an aryl, heterocyclyl, or cycloalkyl ring, wherein the ring linked together with the parent structure is a heteroaryl ring. For example, as used herein, the term "5-14 membered heteroaryl" means a heteroaryl group containing 5 to 14 ring atoms. Specific examples of heteroaryl groups include, but are not limited to, thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl and the like, or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl and the like, or azaindolyl, pyrimidopyrazolyl, pyrrolopyridyl, pyrazolopyrimidine and the like, and their benzo derivatives such as indazolyl, indolyl, isoindolyl, quinolyl, isoquinolyl and the like.
As used herein, the term "halo" or "halogen" group is defined to include F, Cl, Br, or I.
The term "alkoxy," as used herein, means an alkyl group, as defined above, appended to the parent molecular moiety through an oxygen atom. C1-C6Representative examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, pentyloxy, hexyloxy, and the like.
As used herein, the term "heterocyclyl" refers to a monocyclic or polycyclic group having, for example, 2,3,4, 5,6, 7, 8, 9 carbon atoms in the ring and one or more (e.g., 1,2,3, or 4) selected from C (═ O), O, S, S (═ O), S (═ O)2And NR (R represents a hydrogen atom or a substituent such as, but not limited to, an alkyl group or a cycloalkyl group). The heterocyclic group may be saturated or unsaturated. Saturated heterocyclyl groups can be referred to as heterocycloalkyl groups, such as 3-8 membered heterocycloalkyl groups, 5-6 membered heterocycloalkyl groups, and the like. Unless otherwise specifically indicated in the specification, a heterocyclyl group may be a monocyclic, bicyclic, tricyclic or higher ring system, which may include fused, bridged or spiro ring systems. In particular, a 3-10 membered heterocyclyl is a group having 3-10 carbon atoms and heteroatoms in the ring, e.g., having 4 to 10, 4 to 7, 4 to 6, 5 to 10, 5 to 7, or 5 to 6 carbon atoms and heteroatoms (referred to as 4 to 10, 4 to 7, 4 to 6, 5 to 10, 5 to 7, and 5 to 6 membered heterocyclyl, respectively), such as, but not limited to, oxiranyl, aziridinyl, azetidinylAzepinyl, oxetanyl, tetrahydrofuryl, pyrrolidinyl, pyrrolidinonyl, imidazolidinyl, pyrazolidinyl, tetrahydropyranyl, azacycloheptyl
Figure BDA0002557649670000172
Mesityl, piperidyl, morpholinyl, dithianyl (dithianyl), thiomorpholinyl, piperazinyl, trithianyl (trithianyl), and the like; and bicyclic derivatives thereof, such as, but not limited to, pyrrolidinyl-cyclopropyl, cyclopent-aziridinyl, pyrrolidinyl-cyclobutyl, pyrrolidinyl-pyrrolidinyl, pyrrolidinyl-piperidinyl, pyrrolidinyl-piperazinyl, pyrrolidinyl-morpholinyl, piperidinyl-morpholinyl; or a benzo derivative or a heteroaryl and derivative; or spiro derivatives, e.g. but not limited to
Figure BDA0002557649670000171
And the like.
The term "substituted" means that one or more (e.g., 1,2,3, or 4) hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency at the present time is not exceeded and 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 with … …," the substituent may be (1) unsubstituted or (2) substituted. If a carbon of a substituent is described as being optionally substituted with one or more of the list of substituents, one or more hydrogens on the carbon (to the extent of any hydrogens present) may be replaced individually and/or together with an independently selected optional substituent. If the nitrogen of a substituent is described as being optionally substituted with one or more of the list of substituents, then one or more hydrogens on the nitrogen (to the extent any hydrogen is present) may each be replaced with an independently selected optional substituent.
If a substituent is described as being "independently selected from" a group, each substituent is selected independently of the other. Thus, each substituent may be the same as or different from another (other) substituent.
If a variable or substituent can be selected from different variations and the variable or substituent occurs more than once, then the variations can be the same or different.
As used herein, the term "one or more" means 1 or more than 1, such as 2,3,4, 5,6, 7, 8, 9, 10, etc., under reasonable conditions.
Unless indicated, as used herein, the point of attachment of a substituent may be from any suitable position of the substituent.
The invention also includes all pharmaceutically acceptable isotopic 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 which predominates in nature. Examples of isotopes suitable for inclusion in compounds of the invention include, but are not limited to, isotopes of hydrogen (e.g. hydrogen)2H、3H) (ii) a Isotopes of carbon (e.g. of11C、13C and14C) (ii) a Isotopes of chlorine (e.g. of chlorine)36Cl); isotopes of fluorine (e.g. of fluorine)18F) (ii) a Isotopes of iodine (e.g. of iodine)123I and125I) (ii) a Isotopes of nitrogen (e.g. of13N and15n); isotopes of oxygen (e.g. of15O、17O and18o); isotopes of phosphorus (e.g. of phosphorus)32P); and isotopes of sulfur (e.g. of35S)。
The term "stereoisomer" denotes an isomer formed as a result of at least one asymmetric center. In compounds having one or more (e.g., 1,2,3, or 4) asymmetric centers, they can result in 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. For example, dihydropyrimidine groups, 2(1H) -pyridonyl groups, and the like may exist in solution in equilibrium in the following tautomeric forms. It is understood that the scope of this application encompasses all such isomers or mixtures thereof in any ratio (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%).
Unless otherwise indicated, the compounds of the present invention are intended to exist as stereoisomers, including 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 present invention encompasses all possible crystalline forms or polymorphs of the compounds of the present 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 be present in free form for use in therapy 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, solvates, metabolites or prodrugs thereof, which upon administration to a patient in need thereof are capable of providing, directly or indirectly, a compound of the present invention or a metabolite or residue 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. For a review of suitable Salts, see Stahl and Wermuth, "Handbook of Pharmaceutical Salts: Properties, Selection, and Use" (Wiley-VCH, 2002). Methods for preparing pharmaceutically acceptable salts of the compounds of the present invention are known to those skilled in the art.
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.
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. Such products may result, for example, from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, 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. Further 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) and "Bioreversible Carriers in Drug Design," Pergamon Press,1987(E.B.Roche editions, American Pharmaceutical Association). 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 can be achieved by conventional protecting Groups, for example, as described in Protective Groups in Organic Chemistry, ed.j.f.w.mcomie, Plenum Press, 1973; and T.W.Greene & P.G.M.Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons,1991, which are incorporated herein by reference. The protecting group may be removed at a suitable subsequent stage using methods known in the art. As used herein, the term "about" when used in reference to a value or range of values means that the value or range of values and the range of errors acceptable to one skilled in the art for that value or range of values, for example, the range of errors is ± 10%, 5%, 4%, 3%, 2%, 1%, 0.5%, etc.
Advantageous effects of the invention
The compound of the invention has strong inhibitory activity on HPK1, good physicochemical properties (such as solubility, physical and/or chemical stability), good pharmacokinetic properties (such as good bioavailability, proper blood drug concentration, half-life and action duration), good safety (lower toxicity (such as lower cardiac and hepatic toxicity) and/or fewer side effects, wider therapeutic window) and other excellent properties.
Detailed Description
Examples
The invention is further described below in connection with examples, which are not intended to limit the scope of the invention.
The abbreviations in the present invention have the following meanings:
Figure BDA0002557649670000191
Figure BDA0002557649670000201
the structure of the compound of the invention is determined by nuclear magnetic resonance spectrum (1H NMR) or Mass Spectrometry (MS).
The reaction was monitored by Thin Layer Chromatography (TLC) or LCMS.
The microwave reaction was performed using a BiotageInitiator + microwave reactor.
The column chromatography generally uses 200-300 mesh silica gel (Qingdao ocean) as a stationary phase. The system of eluents comprises: a: petroleum ether/ethyl acetate; b: dichloromethane/methanol, the volume ratio of the solvent is adjusted according to the polarity of the compound.
In the following examples, the reaction temperature was room temperature (20 ℃ C. to 30 ℃ C.), unless otherwise specified.
Reagents used in this application were purchased from Acros Organics, Aldrich Chemical Company, or Tereber Chemical, among others.
Intermediate int.1: 1-isopropyl-1H-pyrazolo [4,3-c ] pyridin-6-amine
Figure BDA0002557649670000202
The first step is as follows: 6-chloro-1-isopropyl-1H-pyrazolo [4,3-c ] pyridine (int.1b)
6-chloro-1H-pyrazolo [4,3-c ] pyridine (int.1a,6g,39.07mmol) is dissolved in 50mL DMF, NaH (3.13g,78.14mmol, 60% purity) is added under ice-bath, and then stirred for 0.5H. Then 2-iodopropane (9.96g,58.61mmol) was added and reacted at room temperature for 2 h. After the reaction was completed, the reaction solution was quenched by adding saturated ammonium chloride solution, extracted with EA (50mL × 3), and the organic phases were combined and dried over anhydrous sodium sulfate. The organic phase was spin-dried and purified by flash column chromatography (eluent system a) to yield compound int.1b (4.58 g).
MS(ESI,m/z):196.0[M+H]+.
The second step is that: (1-isopropyl-1H-pyrazolo [4,3-c ] pyridin-6-yl) carbamic acid tert-butyl ester (int.1c)
Int.1b (4.45g,22.74mmol), tert-butyl carbamate (13.32g,113.72mmol), Pd2(dba)3(2.08g,2.27mmol), Brettphos (2.44g,4.55mmol) and Cs2CO3(18.53g,56.86mmol) was dissolved in 50mL of 1, 4-dioxane, N2Heating to 110 ℃ under protection, and reacting for 5 h. After the reaction was completed, the reaction mixture was filtered through celite, and the filtrate was dried by spin-drying and purified by flash column chromatography (eluent system a) to obtain compound int.1c (4.8 g).
MS(ESI,m/z):277.1[M+H]+.
The third step: 1-isopropyl-1H-pyrazolo [4,3-c ] pyridin-6-amine (int.1)
Int.1c (800mg,2.90mmol) was dissolved in 10mL DCM and 10mL TFA and stirred at RT overnight. After the reaction is finished, adding saturated sodium bicarbonate solution to quench the reaction, and adjusting the pH value to be alkaline. Extracted with EA (50mL × 3), the organic phases were combined and dried over anhydrous sodium sulfate. The organic phase was spin-dried and purified by flash column chromatography (eluent system a) to yield compound int.1(200 mg).
MS(ESI,m/z):177.1[M+H]+.
Intermediate int.2: 4, 6-dichloro-1- (2, 2-difluoroethyl) -1H-pyrrolo [2,3-b ] pyridine
Figure BDA0002557649670000211
4, 6-dichloro-1H-pyrrolo [2,3-b ]]Pyridine (int.2a,1g,5.35mmol), Cs2CO3(3.49g,10.69mmol) and 1, 1-difluoro-2-iodoethane (1.54g,8.02mmol) were dissolved in 10mL of DMF and heated to 70 ℃ for 1 h. After the reaction was completed, the reaction was quenched by adding water, extracted with EA (50mL × 3), and the organic phases were combined and dried over anhydrous sodium sulfate. The organic phase was spin-dried and purified by flash column chromatography (eluent system a) to yield compound int.2(1.1 g).
MS(ESI,m/z):251.0[M+H]+.
Intermediate int.3: 2-bromo-5, 5, 6-trimethyl-5, 6-dihydro-4H-pyrazoline [1,5-d ]][1,4]Diaza derivatives
Figure BDA0002557649670000213
-7(8H) -one
Figure BDA0002557649670000212
The first step is as follows: 2- (3, 5-dibromo-1H-pyrazol-1-yl) acetonitrile (Int.3b)
3, 5-dibromo-1H-pyrazole (int.3a,1.5g,6.64mmol) was dissolved in 20mL of DMF, and chloroacetonitrile (750mg,9.96mmol) and potassium carbonate (1.84g,13.28mmol) were added, followed by heating to 40 ℃ for 2H. After the reaction was completed, the reaction was quenched by addition of water, extracted with EA (50mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, the organic phases were dried and purified by flash column chromatography (eluent system a) to yield compound int.3b (1.5 g).
MS(ESI,m/z):265.0[M+H]+.
The second step is that: (3-bromo-5- (2-methylprop-1-enyl) -1H-pyrazol-1-yl) acetonitrile (int.3c)
Compound 1b (1.5g, 5.56mmol), 2-methyl-1-propenylboronic acid pinacol ester (1.03g, 5.56mmol), Pd (dppf) Cl2(413mg, 0.56mmol) and K2CO3(3.13g, 21.65mmol) was added to a mixed solvent of 10mL of 1, 4-dioxane and 10mL of water, and N was added2Heating to 100 ℃ under protection, and reacting for 2 h. After the reaction was completed, the reaction mixture was filtered through celite, and the filtrate was dried by spin-drying and purified by flash column chromatography (eluent system a) to obtain compound int.3c (1 g).
MS(ESI,m/z):240.0[M+H]+.
The third step: 2-bromo-5, 5-dimethyl-5, 6-dihydro-4H-pyrazoline [1,5-d ]][1,4]Diaza derivatives
Figure BDA0002557649670000222
-7(8H) -one (int.3d)
Int.3c (1g,4.16mmol) was added to 20mL of methanesulfonic acid, heated to 65 ℃ and reacted for 72 h. After the reaction is finished, adjusting the pH to 8-9 by using a 1N sodium hydroxide solution. Extracting with EA (50 mL. multidot.3), mixing the organic phases, drying with anhydrous sodium sulfate, spin-drying the organic phase, and subjecting to preparative high performance liquid chromatography (preparation method: Prep-HPLC (instrument model: Agilent 1260, chromatography column: Waters SunAire Prep C)18OBD (19 mm. times.150 mm. times.5.0. mu.m); temperature of the chromatographic column: 25 ℃; flow rate: 20.0 mL/min; detection wavelength: 214 nm; elution gradient: (0 min: 10% A, 90% B; 16.0 min: 90% A, 10% B); mobile phase A: 100% acetonitrile; mobile phase B: 0.05% aqueous formic acid. Compound retention time Rt5.6min) to yield compound int.3d (200 mg).
MS(ESI,m/z):258.0[M+H]+.
The fourth step: 2-bromo-5, 5, 6-trimethyl-5, 6-dihydro-4H-pyrazoline [1,5-d ]][1,4]Diaza derivatives
Figure BDA0002557649670000223
-7(8H) -one (int.3)
Int.3d (100mg, 387.42. mu. mol) was dissolved in 10mL THF, and potassium tert-butoxide (52mg, 464.91. mu. mol) was added. After stirring at room temperature for 0.5h, iodomethane (110mg, 774.85. mu. mol) was added and stirring was continued for 1 h. After the reaction, 2mL of methanol was added to the reaction solution, the organic phase was spin-dried, and subjected to preparative high performance liquid chromatography (preparation method: Prep-HPLC (Instrument model: Agilent 1260, chromatography column: Waters SunAire Prep C)18OBD (19 mm. times.150 mm. times.5.0. mu.m); temperature of the chromatographic column: 25 ℃; flow rate: 20.0 mL/min; detection wavelength: 214 nm; elution gradient: (0 min: 10% A, 90% B; 16.0 min: 90% A, 10% B); mobile phase A: 100% acetonitrile; mobile phase B: 0.05% aqueous formic acid. Compound retention time Rt6.1min) to yield compound int.3(45 mg).
MS(ESI,m/z):272.0[M+H]+.
Example 1: (Z) -N- (1- (2-fluorovinyl) -1H-pyrrolo [2,3-b ] pyridin-6-yl) -1-isopropyl-1H-pyrazolo [4,3-c ] pyridin-6-amine (Compound 1)
Figure BDA0002557649670000221
Int.1(42.74mg, 170.24. mu. mol), Int.2(30mg, 170.24. mu. mol), tBuONa (49.08mg, 510.73. mu. mol), xantphos (9.85mg, 17.02. mu. mol) and Pd2(dba)3(15.59mg, 17.02. mu. mol) was added to a 10mL microwave tube. 3mL of DMF was added, and the mixture was heated to 100 ℃ by microwave for 2 hours. After the reaction, the reaction mixture was filtered through celite, and the filtrate was dried by preparative high performance liquid chromatography (preparation method: Prep-HPLC (Instrument model: Agilent 1260, chromatography column: Waters SunAire Prep C)18OBD (19 mm. times.150 mm. times.5.0. mu.m); temperature of the chromatographic column: 25 ℃; flow rate: 20.0 mL/min; detection wavelength: 214 nm; elution gradient: (0 min: 10% A, 90% B; 16.0 min: 90% A, 10% B); mobile phase A: 100% acetonitrile; flow ofPhase B: 0.05% aqueous formic acid. Compound retention time Rt9.3min) to give compound 1(2.11 mg).
MS(ESI,m/z):371.1[M+H]+.
1H NMR(400MHz,MeOD)δ8.74(s,1H),8.52(s,1H),8.08(s,1H),7.84(d,J=8.4Hz,1H),7.59(dd,J=3.7,1.8Hz,1H),7.07–6.98(dd,J=35.4,4.2Hz,1H),6.95–6.93(d,J=8.4Hz,1H),6.95–6.74(dd,J=74.0,4.3Hz,1H),6.56–6.55(d,J=3.7Hz,1H),4.88–4.85(m,1H)1.63–1.62(d,J=6.7Hz,6H).
Example 2: n- (1- (2, 2-difluoroethyl) -4- (pyrrolidin-3-yl) -1H-pyrrolo [2,3-b ] pyridin-6-yl) -1-isopropyl-1H-pyrazolo [4,3-c ] pyridin-6-amine (Compound 2)
Figure BDA0002557649670000231
The first step is as follows: n- (4-chloro-1- (2, 2-difluoroethyl) -1H-pyrrolo [2,3-b ] pyridin-6-yl) -1-isopropyl-1H-pyrazolo [4,3-c ] pyridin-6-amine (Compound 2a)
Compounds int.1(400mg,2.27mmol), int.2(570mg,2.27mmol), Cs2CO3(1.48g,4.54mmol), Xantphos (131.34mg, 226.99. mu. mol) and Pd2(dba)3(207.86mg, 226.99. mu. mol) was dissolved in 10mL1, 4-dioxane, heated to 90 ℃ and reacted overnight. After the reaction was completed, the mixture was filtered through celite, and the filtrate was spin-dried and purified by flash column chromatography (eluent system a) to give compound 2a (220 mg).
MS(ESI,m/z):391.0[M+H]+.
The second step is that: tert-butyl 3- (1- (2, 2-difluoroethyl) -6- (((1-isopropyl-1H-pyrazolo [4,3-c ] pyridin-6-yl) amino ] -1H-pyrrolo [2,3-b ] pyridin-4-yl) -2, 5-dihydro-1H-pyrrole-1-carboxylate (Compound 2b)
Compound 2a (100mg, 266.10. mu. mol), 3- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2, 5-dihydropyrrole-1-carboxylic acid tert-butyl ester (117.82mg, 399.15. mu. mol), Pd (dppf) Cl2(19.47mg, 26.61. mu. mol) and Cs2CO3(173.50mg, 532.19. mu. mol) was dissolved in 10mL of 1, 4-dioxoSix rings and 1mL water. N is a radical of2Heating to 90 ℃ under protection, and reacting for 2 h. After the reaction was completed, the mixture was filtered through celite. The filtrate was spin-dried and purified by flash column chromatography (eluent system a) to give compound 2b (38 mg).
MS(ESI,m/z):524.2[M+H]+.
The third step: tert-butyl 3- (1- (2, 2-difluoroethyl) -6- (((1-isopropyl-1H-pyrazolo [4,3-c ] pyridin-6-yl) amino ] -1H-pyrrolo [2,3-b ] pyridin-4-yl) pyrrolidine-1-carboxylic acid ester (Compound 2c)
Compound 2b (10.00mg, 19.10. mu. mol), 10% Pd/C (2.32mg, 19.10. mu. mol) was dissolved in 10mL MeOH, H2The reaction was carried out overnight under protection. After the reaction was completed, the reaction solution was filtered. The filtrate was spun dry to give compound 2c (8mg) which was used directly in the next reaction.
MS(ESI,m/z):526.2[M+H]+.
The fourth step: n- (1- (2, 2-difluoroethyl) -4- (pyrrolidin-3-yl) -1H-pyrrolo [2,3-b ] pyridin-6-yl) -1-isopropyl-1H-pyrazolo [4,3-c ] pyridin-6-amine (Compound 2)
Compound 2c (8mg, 15.22. mu. mol) was dissolved in 10mL DCM, 10mL TFA was added, and the reaction was carried out at room temperature for 2 h. After the reaction, the reaction mixture was concentrated and subjected to preparative high performance liquid chromatography (preparation method: Prep-HPLC (Instrument model: Agilent 1260, chromatography column: Waters SunAire Prep C)18OBD (19 mm. times.150 mm. times.5.0. mu.m); temperature of the chromatographic column: 25 ℃; flow rate: 20.0 mL/min; detection wavelength: 214 nm; elution gradient: (0 min: 10% A, 90% B; 16.0 min: 90% A, 10% B); mobile phase A: 100% acetonitrile; mobile phase B: 0.05% aqueous formic acid. Compound retention time Rt3.8min) to give compound 2(3.57 mg).
MS(ESI,m/z):426.2[M+H]+.
1H NMR(400MHz,MeOD)δ8.76(s,1H),8.64(s,1H),8.10(s,1H),7.26–7.25(d,J=3.5Hz,1H),6.84(s,1H),6.59–6.58(d,J=3.6Hz,1H),6.47–6.17(tt,J=55.6,3.9Hz,1H),4.92–4.90(m,1H),4.78–4.70(td,J=14.6,4.0Hz,2H),3.93–3.82(m,2H),3.70–3.58(m,1H),3.51–3.41(m,2H),2.66–2.49(m,1H),2.42–2.28(m,1H),1.60–1.58(t,J=9.3Hz,6H).
Example 3: n- (1- (morpholin-2-ylmethyl) -1H-pyrazolo [4,3-c ] pyridin-6-yl) cyclopropanecarboxamide (Compound 3)
Figure BDA0002557649670000241
The first step is as follows: 6-chloro-1-trityl-1H-pyrazolo [4,3-c ] pyridine (Compound 3a)
Int.1a (4g,26.05mmol) was dissolved in 60mL THF, NaH (1.80g,46.88mmol, 60% purity) was added to the reaction at 0 deg.C and stirred for 0.5 h. Triphenylchloromethane (18.88g,67.72mmol) was added to the reaction system, stirred at 0 ℃ for 0.5h, warmed to room temperature, and reacted for 2 h. After the reaction was completed, extraction was performed with EA (50mL × 3), and the organic phases were combined and dried over anhydrous sodium sulfate. The organic phase was spin dried and purified by flash column chromatography (eluent system a) to give compound 3b (8.4 g).
MS(ESI,m/z):396.1[M+H]+.
The second step is that: (1-trityl-1H-pyrazolo [4,3-c ] pyridin-6-yl) carbamic acid tert-butyl ester (Compound 3b)
Compound 3b (8.4g, 21.22mmol), tert-butylcarbamate (12.43g, 106.09mmol), Pd2(dba)3(1.94mg, 2.12mmol), Brettphos (2.28g, 4.24mmol) and Cs2CO3(17.28g, 53.05mmol) was dissolved in 100mL of 1, 4-dioxane, N2Heating to 110 ℃ under protection, and reacting for 6 h. After the reaction was completed, the mixture was filtered through celite. The filtrate was spin dried and purified by flash column chromatography (eluent system a) to give compound 3b (7.2 g).
MS(ESI,m/z):477.0[M+H]+.
The third step: (Cyclopropanecarbonyl) (1-trityl-1H-pyrazolo [4,3-c ] pyridin-6-yl) carbamic acid tert-butyl ester (Compound 3c)
Compound 3b (7.1g,14.90mmol) and DIPEA (7.7g,59.59mmol) were dissolved in 100mL DCM, cyclopropanecarbonyl chloride (3.11g,29.80mmol) was slowly added dropwise to the reaction mixture at 0 deg.C, and the reaction was carried out at room temperature for 2 h. After completion of the reaction, the reaction mixture was directly concentrated to obtain compound 3c (8 g).
MS(ESI,m/z):545.0[M+H]+.
The fourth step: n- (1H-pyrazolo [4,3-c ] pyridin-6-yl) cyclopropanecarboxamide (Compound 3d)
Compound 3c (8.0g,14.69mmol) was dissolved in 80mL DCM and 20mL TFA and reacted at room temperature for 5 h. After the reaction is finished, the pH is adjusted to about 8-9 by using saturated sodium bicarbonate. Extracted with EA (50mL × 3), the organic phases were combined and dried over anhydrous sodium sulfate. The organic phase was spin dried and purified by flash column chromatography (eluent system a) to give compound 3d (2.3 g).
MS(ESI,m/z):203.1[M+H]+.
The fifth step: tert-butyl 2- ((6- (cyclopropanecarboxamido) -1H-pyrazolo [4,3-c ] pyridin-1-yl) methyl) morpholine-4-carboxylate (compound 3e)
Compound 3d (200mg, 989.06. mu. mol) and tert-butyl 2- (bromomethyl) morpholine-4-carboxylate (415.64mg,1.48mmol) were dissolved in 8mL DMF and K was added2CO3(272.98mg,1.98mmol), the temperature was raised to 60 ℃ and the reaction was stirred for 15 h. After the reaction was completed, extraction was performed with EA (50mL × 3), and the organic phases were combined and dried over anhydrous sodium sulfate. The organic phase was spin dried and purified by flash column chromatography (eluent system a) to give compound 3e (300 mg).
MS(ESI,m/z):402.0[M+H]+.
And a sixth step: n- (1- (morpholin-2-ylmethyl) -1H-pyrazolo [4,3-c ] pyridin-6-yl) cyclopropanecarboxamide (Compound 3)
Compound 3e (300mg, 747.28. mu. mol) was added to 12mL DCM and 3mL TFA and reacted at room temperature with stirring for 2 h. After the reaction is finished, the pH is adjusted to about 8-9 by saturated sodium bicarbonate, and the mixture is subjected to preparative high performance liquid chromatography (preparation method: Prep-HPLC (instrument model: Agilent 1260, chromatographic column: Waters SunAire Prep C)18OBD (19 mm. times.150 mm. times.5.0. mu.m); temperature of the chromatographic column: 25 ℃; flow rate: 28.0 mL/min; detection wavelength: 214 nm; elution gradient: (2 min: 8% A, 92% B; 15.0 min: 40% A, 60% B); mobile phase A: 100% acetonitrile; mobile phase B: 0.05% aqueous ammonium bicarbonate solution. Compound retention time Rt7.12min) compound 3(130mg) was isolated.
MS(ESI,m/z):302.0[M+H]+.
1H NMR(400MHz,DMSO-d6)δ10.91(s,1H),8.84(d,J=0.7Hz,1H),8.21(d,J=3.4Hz,2H),4.37–4.22(m,2H),3.82–3.73(m,1H),3.64(d,J=11.0Hz,1H),3.31(td,J=11.0,4.2Hz,1H),2.76(d,J=11.8Hz,1H),2.66–2.53(m,2H),2.45(t,J=11.0Hz,1H),2.11–2.00(m,1H),0.89–0.77(m,4H).
Example 4: 2- ((1-isopropyl-1H-pyrazolo [4, 3-c)]Pyridin-6-yl) amino) -5,5, 6-trimethyl-5, 6-dihydro-4H-pyrazoline [1,5-d][1,4]Diaza derivatives
Figure BDA0002557649670000253
-7(8H) -one (Compound 4)
Figure BDA0002557649670000251
Int.3(15mg, 56.75. mu. mol), Int.1(15mg, 56.75. mu. mol), cesium carbonate (37mg, 113.50. mu. mol), Xantphos (3.3mg, 5.67. mu. mol) and Pd2(dba)3(5.2mg, 5.67. mu. mol) in 10mL1, 4-dioxane, N2Heating to 100 ℃ under protection, and reacting for 2 h. After the reaction was completed, the mixture was filtered through celite. The filtrate was spin-dried and subjected to preparative high performance liquid chromatography (preparation method: Prep-HPLC (instrument model: Agilent 1260, column: Waters SunAire Prep C)18OBD (19 mm. times.150 mm. times.5.0. mu.m); temperature of the chromatographic column: 25 ℃; flow rate: 20.0 mL/min; detection wavelength: 214 nm; elution gradient: (0 min: 10% A, 90% B; 16.0 min: 90% A, 10% B); mobile phase A: 100% acetonitrile; mobile phase B: 0.05% aqueous formic acid. Compound retention time Rt4.2min) to give compound 4(1.37 mg).
MS(ESI,m/z):368.2[M+H]+.
1H NMR(400MHz,MeOD)δ8.65(s,1H),8.03(s,1H),7.57(s,1H),6.09(s,1H),5.05(s,2H),4.82–4.79(m,1H),3.24(s,2H),2.92(s,3H),1.55–1.53(d,J=6.7Hz,6H).
Example 5: n- (1-isopropyl-1H-pyrazolo [3,4-d ] pyrimidin-6-yl) -6-methoxy-2-methyl-1, 2,3, 4-tetrahydroisoquinolin-7-amine (compound 5)
Figure BDA0002557649670000252
The first step is as follows: 2, 4-dichloropyrimidine-5-carbaldehyde (Compound 5b)
Compound 5a (3g,13.17mmol) was dissolved in 30mL of THF, and isopropyl magnesium chloride-lithium chloride (11.14mL,14.48mmol) was added to the reaction system at-78 deg.C, followed by reaction for 40 minutes with stirring. Then a solution of morpholine-4-aldehyde (4.55g,39.50mmol) in 30mL of THF was added to the reaction, stirred at-78 deg.C for 10min, then raised to-40 deg.C and reacted for 1 h. After the reaction was completed, the reaction was quenched with 1N hydrochloric acid. Extracted with EA (50mL × 3), the organic phases were combined and dried over anhydrous sodium sulfate. The organic phase was spin dried and purified by flash column chromatography (eluent system a) to give compound 5b (830 mg).
MS(ESI,m/z):177.0[M+H]+.
The second step is that: 6-chloro-1H-pyrazolo [3,4-d ] pyrimidine (compound 5c)
Compound 5b (800mg,4.52mmol) was dissolved in 10mL THF, and a solution of hydrazine hydrate (452.55mg,9.04mmol) in 10mL THF was added to the reaction at 0 deg.C, followed by reaction for 1h with stirring at room temperature. After the reaction was completed, extraction was performed with EA (50mL × 3), and the organic phases were combined and dried over anhydrous sodium sulfate. The organic phase was spin dried and purified by flash column chromatography (eluent system a) to give compound 5c (190 mg).
MS(ESI,m/z):155.1[M+H]+.
The third step: 6-chloro-1-isopropyl-1H-pyrazolo [3,4-d ] pyrimidine (compound 5d)
Compound 5c (180mg,1.16mmol) was dissolved in 5mL of DMF, the system was left at 0 ℃ and NaH (55.9mg,46.88mmol, 60% purity) was added to the reaction system and reacted for 0.5h with stirring. Finally, 2-iodopropane (296.96mg,1.75mmol) was added to the reaction system, and the reaction was allowed to warm to room temperature for 1 hour. After the reaction was completed, extraction was performed with EA (50mL × 3), and the organic phases were combined and dried over anhydrous sodium sulfate. The organic phase was spin dried and purified by flash column chromatography (eluent system a) to give compound 5d (120 mg).
MS(ESI,m/z):197.1[M+H]+.
The fourth step: n- (1-isopropyl-1H-pyrazolo [3,4-d ] pyrimidin-6-yl) -6-methoxy-2-methyl-1, 2,3, 4-tetrahydroisoquinolin-7-amine (compound 5)
Compound 5d (49.09mg, 249.67. mu. mol) and 6-methoxy-2-methyl-1, 2,3, 4-tetrahydroisoquinolin-7-amine (40mg, 208.05. mu. mol) were added to 3mL of isopropanol and 0.02mL of concentrated hydrochloric acid, and the mixture was heated to 80 ℃ and reacted with stirring for 15 hours. After the reaction, the reaction mixture was cooled, concentrated, and subjected to preparative high performance liquid chromatography (preparation method: Prep-HPLC (instrument model: Agilent 1260, column: Waters SunAire Prep C)18OBD (19 mm. times.150 mm. times.5.0. mu.m); temperature of the chromatographic column: 25 ℃; flow rate: 28.0 mL/min; detection wavelength: 214 nm; elution gradient: (3 min: 10% A, 90% B; 16.0 min: 70% A, 30% B); mobile phase A: 100% acetonitrile; mobile phase B: 0.05% aqueous ammonium bicarbonate solution. Compound retention time Rt8.1min) compound 5(10mg) was isolated.
MS(ESI,m/z):353.1[M+H]+.
1H NMR(400MHz,CDCl3)δ8.80(s,1H),8.35(s,1H),7.90(d,J=3.4Hz,2H),6.64(s,1H),5.10–4.98(m,1H),3.89(s,3H),3.62(s,2H),2.91(t,J=5.8Hz,2H),2.71(t,J=5.9Hz,2H),2.51(s,3H),1.62(d,J=6.7Hz,6H).
The compounds in the following table were synthesized according to the corresponding methods in the above examples:
Figure BDA0002557649670000261
Figure BDA0002557649670000271
Figure BDA0002557649670000281
biological evaluation
The following experimental examples further describe and explain the present invention, but these examples are not intended to limit the scope of the present invention.
Experimental example 1: inhibition assay of HPK1 enzymatic activity by Compounds
Compounds were tested for HPK1 enzymatic activity using an ATP-glossay assay, reagents (HPK1,5x kinase buffer, substrate (mbp), ATP) were purchased from Promega corporation, and protocol and optimization were recommended based on the instructions for the reagents.
Mu.l 2.5 XHPK 1(2 ng/. mu.l) was added to the 384 well plates followed by 1. mu.l of the 5 Xrange of concentrations of the test compound, and after incubation for 10min at room temperature 2. mu.l 2.5 XMBP (0.25. mu.g/. mu.l)/ATP (25. mu.M) was added and incubated for 1h at room temperature. After completion of the incubation, 5. mu.l of ATP-Glo reagent (ATP-Glo reagent) was added to each well of the 384-well plate, incubated at room temperature, the reaction was terminated, and the remaining ATP was removed. After 40min, 10. mu.l of ATP-Glo detection buffer (ATP-Glo detection buffer) was added to each well and reacted at room temperature for 1 hour. After the reaction was completed, the 384 well plates were transferred to BMG LABTECH (PHERAstar) for reading, and the mode was selected: luminescence, data presentation: relative chemiluminescence values RLU (relative Luminescence units).
Data processing: the inhibition of HPK1 enzymatic activity by the compound was calculated according to the following formula:
compound inhibition ratio (%) ═ RLUMaximum of-RLUBlank space)-(RLUTreatment of-RLUBlank space))/(RLUMaximum of-RLUBlank space)×100%
Wherein, RLUMaximum ofThe values are relative chemiluminescence values of the group treated with no compound; RLUBlank spaceRelative chemiluminescence values for the group without HPK1 and without compound treatment; RLUTreatment ofIs the relative chemiluminescence value of the compound treatment group. IC was calculated by Graphpad Prism software log (inhibitor) vs. response-variable slope (four parameter) fitting50
Compound (I) HPK1 IC50(nM)
1 60.79
2 4.22
3 175.07
4 60.87
5 15.91
Therefore, the compounds (such as the compounds 1-5) have better inhibition effect on the enzymatic activity of HPK 1.

Claims (15)

1. A compound of formula I or a stereoisomer, tautomer, pharmaceutically acceptable salt, polymorph, co-crystal, solvate, metabolite, prodrug or any mixture of two or more thereof:
Figure FDA0002557649660000011
wherein the content of the first and second substances,
R1selected from hydrogen, C (O) Ra、CO2Ra、C(O)NRbRc、S(O)Ra、SO2Ra、SO2NRbRc、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 3-8 membered heterocyclyl and 5-14 membered heteroaryl, said C1-C6Alkyl radical, C3-C7CycloalkanesBase, C6-C14Aryl, 3-8 membered heterocyclyl and 5-14 membered heteroaryl are each optionally substituted with one or more RdSubstitution;
R2selected from hydrogen, halogen, CN, NO2、C(O)Ra、CO2Ra、C(O)NRbRc、S(O)Ra、SO2Ra、SO2NRbRc、NRbRc、NHC(O)Ra、NHCO2Ra、OC(O)Ra、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C14Aryl and 5-14 membered heteroaryl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with one or more RdSubstitution;
R3selected from hydrogen, C (O) Ra、CO2Ra、C(O)NRbRc、S(O)Ra、SO2Ra、SO2NRbRc、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl, said C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 5-10 membered heterocyclyl are each optionally substituted with one or more RdSubstitution;
Raindependently selected from C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl, said C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 5-10 membered heterocyclyl are each optionally substituted with one or more RdSubstitution;
Rband RcEach independently selected from hydrogen and C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical C1-C6Alkyl and 4-7 membered heterocyclyl; or, RbAnd RcTogether with the nitrogen atom to which they are attached form a 4-7 membered heterocyclyl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical C1-C6Each of alkyl and 4-7 membered heterocyclyl is optionally substituted with one or more RdSubstitution;
x is selected from CH and N;
Rdindependently selected from: hydrogen, hydroxy, halogen, oxo, CN, NO2、C(O)Re、CO2Re、NRfSO2Rg、S(O)Re、SO2Re、C(O)NRfRg、SO2NRfRg、NRfRg、NRfC(O)Rg、C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical C1-C6Alkoxy radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl, said C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical C1-C6Alkoxy radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl are each optionally substituted with one or more RjSubstitution;
Reindependently selected from hydrogen, C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl;
Rfand RgEach independently selected from hydrogen and C1-C6Alkyl radical, C3-C7Cycloalkyl and 4-7 membered heterocyclyl, wherein said C is1-C6Alkyl is optionally substituted by C1-C6Alkoxy or NRhRiSubstitution; or, RbAnd RcTogether with the nitrogen atom to which they are attached form a 4-7 membered heterocyclyl;
Rhand RiEach independently selected from hydrogen and C1-C6An alkyl group; and is
RjIndependently selected from halogen, hydroxy, CN, C (O) Re、NRfRg、C(O)NRbRe、NRbC(O)ReAnd 3-8 membered heterocyclic groups.
2. A compound of claim 1, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug, or any mixture of two or more thereof,
R1selected from hydrogen, C (O) Ra、CO2Ra、C(O)NRbRc、S(O)Ra、SO2Ra、SO2NRbRc、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl, 3-8 membered heterocyclyl and 5-10 membered heteroaryl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl, 3-8 membered heterocyclyl and 5-10 membered heteroaryl are each optionally substituted with one or more RdSubstitution;
preferably, R1Selected from hydrogen, C (O) Ra、C1-C6Alkyl radical, C3-C7Cycloalkyl and 3-8 membered heterocyclyl, said C1-C6Alkyl radical, C3-C7Cycloalkyl and 3-8 membered heterocyclyl are each optionally substituted with one or more RdSubstitution;
more preferably, R1Selected from hydrogen, C1-C6Alkyl radical, C3-C7Cycloalkyl and 3-8 membered heterocyclyl, said C1-C6Alkyl radical, C3-C7Cycloalkyl and 3-8 membered heterocyclyl are each optionally substituted with RdSubstitution;
more preferably, R1Is selected from C1-C6Alkyl and 5-6 membered azacycloalkyl, said C1-C6Alkyl is optionally substituted with a group selected from: hydroxy, C1-C6Alkoxy, halogen, and 5-6 membered heterocyclyl;
more preferably, R1Is selected from C1-C6Alkyl and pyrrolidinyl, said C1-C6Alkyl is optionally substituted with a group selected from: hydroxy, C1-C6Alkoxy, halogen and morpholinyl;
more preferably, R1Is selected from C1-C6Alkyl and pyrrolidinyl, said C1-C6Alkyl is optionally substituted with a group selected from: hydroxy, methoxy, fluoro and morpholinyl;
particularly preferably, R1Selected from the group consisting of isopropyl, sec-butyl, morpholin-2-ylmethyl, 2-hydroxyethyl, 2-methoxyethyl, 2-fluoroethyl and pyrrolidinyl.
3. A compound of claim 1 or 2, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof,
R2selected from hydrogen, halogen, CN, NO2、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C10Aryl and 5-to 10-membered heteroaryl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted with one or more RdSubstitution;
preferably, R2Selected from hydrogenHalogen, C1-C6Alkyl and C3-C7Cycloalkyl radical, said C1-C6Alkyl and C3-C7Cycloalkyl is each optionally substituted by one or more RdSubstitution;
more preferably, R2Selected from hydrogen, halogen and C1-C6Alkyl radical, said C1-C6Alkyl is optionally substituted by one or more RdSubstitution;
particularly preferably, R2Is hydrogen.
4. A compound according to any one of claims 1 to 3, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof,
R3selected from hydrogen, C (O) Ra、CO2Ra、C(O)NRbRc、S(O)Ra、SO2Ra、SO2NRbRc、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl and 5-to 10-membered heteroaryl, said C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl, 5-10 membered heteroaryl and 5-10 membered heterocyclyl are each optionally substituted with one or more RdSubstitution;
preferably, R3Selected from C (O) Ra、CO2Ra、C(O)NRbRc、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl and 5-to 10-membered heteroaryl, said C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C1-C6Alkyl radical, C3-C7A cycloalkyl group, a,C6-C10Aryl, 5-10 membered heteroaryl and 5-10 membered heterocyclyl are each optionally substituted with one or more RdSubstitution;
more preferably, R3Selected from C (O) Ra、CO2Ra、C6-C10Aryl and 5-to 10-membered heteroaryl, said C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C6-C10Aryl, 5-10 membered heteroaryl, C3-C7Cycloalkyl and 5-10 membered heterocyclyl are each optionally substituted with 1,2,3 or 4RdSubstitution;
more preferably, R3Selected from C (O) Ra、C6-C10Aryl and 5-to 10-membered heteroaryl, said C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C6-C10Aryl, 5-10 membered heteroaryl, C3-C7Cycloalkyl and 5-10 membered heterocyclyl are each optionally substituted with 1,2,3 or 4RdSubstitution;
more preferably, R3Selected from C (O) RaPhenyl and 5-9 membered heteroaryl, each of which is optionally substituted with C5-C6Cycloalkyl or 5-9 membered heterocyclyl fused and said phenyl, 5-9 membered heteroaryl, C5-C6Cycloalkyl and 5-9 membered heterocyclyl are each optionally substituted with 1,2,3 or 4RdAnd (4) substitution.
5. A compound of claim 4, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug, or any mixture of two or more thereof,
R3selected from pyrazolyl, pyridyl, pyrimidinyl, indolyl, indazolyl or pyrimidopyrazolyl, optionally substituted by 1 or 2RdSubstitution; and is
RdIndependently selected from oxo, amino, C1-C6Alkyl and pyrrolidinyl, said C1-C6Alkyl and pyrrolidinyl are each optionally substituted with hydroxy, CN or acetyl.
6. A compound according to any one of claims 1 to 3, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof, wherein the compound is a compound of formula II:
Figure FDA0002557649660000031
wherein the content of the first and second substances,
Rais selected from C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl, said C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 5-10 membered heterocyclyl are each optionally substituted with one or more RdSubstitution;
preferably, RaIs selected from C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl and 5-to 10-membered heteroaryl, said C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted with C3-C7Cycloalkyl or 5-to 10-membered heterocyclyl fused, and said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C6-C10Aryl and 5-10 membered heteroaryl are each optionally substituted by 1 or 2RdSubstitution;
more preferably, RaIs selected from C1-C6Alkyl radical, C3-C7Cycloalkyl and 5-to 10-membered heteroaryl, said5-10 membered heteroaryl optionally fused with 5-6 membered heterocyclyl, and said C1-C6Alkyl radical, C3-C7Cycloalkyl, 5-10 membered heteroaryl and 5-6 membered heterocyclyl are each optionally substituted with 1 or 2 substituents independently selected from halogen and C1-C6Radical substitution of alkyl;
more preferably, RaIs selected from C1-C6Alkyl radical, C3-C7Cycloalkyl, pyrrolyl, pyrazolyl and imidazolyl each optionally fused to a benzene or tetrahydropyrimidine ring, and C1-C6Alkyl radical, C3-C7Cycloalkyl, pyrrolyl, pyrazolyl, imidazolyl, phenyl or tetrahydropyrimidine ring each optionally substituted by 1 or 2 substituents independently selected from halogen and C1-C6Radical substitution of alkyl;
particularly preferably, RaSelected from cyclopropyl, fluorocyclopropyl, difluorocyclopropyl, cyclobutyl, isopropyl, pyrazolyl, methyl pyrazolyl, indazolyl, indolyl and tetrahydropyrimidizolyl.
7. A compound according to any one of claims 1 to 3, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof, wherein the compound is of formula III:
Figure FDA0002557649660000041
wherein the content of the first and second substances,
R4selected from hydrogen, C (O) Re、CO2Re、C(O)NRfRg、S(O)Re、SO2Re、SO2NRfRg、C1-C6Alkyl radical, C2-C6Alkenyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl, said C1-C6Alkyl radical, C2-C6Alkenyl radical, C3-C7Cycloalkyl radical, C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with 1 or 2RjSubstitution; and is
R5Selected from hydrogen, NRfRg、C1-C6Alkyl radical, C3-C7Cycloalkyl, 3-8 membered heterocyclyl, C6-C14Aryl and 5-14 membered heteroaryl, said C1-C6Alkyl radical, C3-C7Cycloalkyl, 3-8 membered heterocyclyl, C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with 1 or 2RjSubstitution;
preferably, the first and second electrodes are formed of a metal,
R4is selected from C1-C6Alkyl and C2-C6Alkenyl radical, said C1-C6Alkyl and C2-C6Each alkenyl group is optionally substituted by 1 or 2 substituents independently selected from halogen, NRfRgAnd 5-6 membered heterocyclyl;
R5selected from hydrogen, NRfRgAnd 5-6 membered heterocyclyl;
Rfand RgEach independently selected from hydrogen and C1-C6Alkyl radical, wherein said C1-C6Alkyl is optionally substituted by NRhRiSubstitution; and;
Rhand RiEach independently selected from hydrogen and C1-C6An alkyl group;
it is particularly preferred that,
R4selected from 2-fluorovinyl, 2-difluoroethyl, isopropyl, isobutyl, and mixtures thereof,
Figure FDA0002557649660000042
And 2- (N, N-dimethylamino) ethyl; and is
R5Selected from the group consisting of hydrogen, pyrrolidinyl, morpholinyl, piperidinyl, 2- (N-methylamino) ethylamino, and 2- (N, N-dimethylamino) ethylamino.
8. A compound according to any one of claims 1 to 3, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof, wherein the compound is of formula IV:
Figure FDA0002557649660000043
wherein the content of the first and second substances,
R6and R7Each independently selected from hydrogen, halogen, CN, NO2、C(O)Re、CO2Re、C(O)NRfRg、S(O)Re、SO2Re、SO2NRfRg、NRfRg、C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C14Aryl and 5-14 membered heteroaryl, said C1-C6Alkyl radical, C3-C7Cycloalkyl radical, C1-C6Alkoxy radical, C6-C14Aryl and 5-14 membered heteroaryl are each optionally substituted with one or more RjSubstitution;
R8and R9Each independently selected from hydrogen, NRfRgAnd C1-C6An alkyl group; or, R8And R9Together with the carbon atom to which they are attached form C3-C7Cycloalkyl or carbonyl, said C1-C6Alkyl and C3-C7Cycloalkyl is each optionally substituted by one or more RjSubstitution;
y is selected from CH and N;
u and V are each independently selected from CRfRg、NReO, S (O) and SO2(ii) a And is
m and n are each independently selected from 0, 1 and 2, and the sum of m and n is 0, 1 or 2;
preferably, the first and second electrodes are formed of a metal,
R6and R7Each independently selected from hydrogen, halogen and C1-C6An alkoxy group;
R8and R9Each independently selected from hydrogen and NRfRg(ii) a Or, R8And R9Together with the carbon atom to which it is attached form a carbonyl group;
y is CH;
u and V are each independently selected from CRfRgAnd NRe
ReIndependently selected from hydrogen and C1-C6An alkyl group;
Rfand RgEach independently selected from hydrogen and C1-C6An alkyl group; and is
m and n are each independently selected from 0, 1 and 2, and the sum of m and n is 0, 1 or 2;
it is particularly preferred that,
R6and R7Each independently selected from hydrogen, fluoro and methoxy;
R8is hydrogen;
R9selected from hydrogen and N, N-dimethylamino;
or, R8And R9Together with the carbon atom to which it is attached form a carbonyl group;
y is CH;
u is CH2
V is selected from CH2、NCH(CH3)2And N (CH)3) (ii) a And is
m and n are each independently selected from 0, 1 and 2, and the sum of m and n is 0, 1 or 2.
9. A compound according to any one of claims 1 to 3, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof, wherein the compound is of formula V:
Figure FDA0002557649660000051
wherein the content of the first and second substances,
R10selected from hydrogen, C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl, said C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl are each optionally substituted with one or more RjSubstitution; and is
R11、R12、R13、R14Each independently selected from hydrogen, halogen, hydroxy, C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl, said C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Alkoxy radical, C3-C7Cycloalkyl radical, C6-C14Aryl, 5-14 membered heteroaryl and 3-8 membered heterocyclyl are each optionally substituted with one or more RjSubstitution; or, R11、R12Together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl group; or, R13、R14Together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl group;
preferably, the first and second electrodes are formed of a metal,
R10is C1-C6An alkyl group; and is
R11、R12、R13、R14Each independently selected from hydrogen and C1-C6An alkyl group; or, R11、R12Together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl group; or, R13、R14Together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl group;
it is particularly preferred that,
R10selected from methyl and isopropyl; and is
R11、R12、R13、R14Each independently selected from hydrogen and methyl; or, R11、R12Together with the carbon atom to which it is attached form a cyclopropyl group; or, R13、R14Together with the carbon atom to which it is attached, form a cyclopropyl group.
10. The compound of claim 1, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug, or any mixture of two or more thereof, wherein the compound is selected from the group consisting of:
Figure FDA0002557649660000061
Figure FDA0002557649660000071
11. a pharmaceutical composition comprising a compound of any one of claims 1 to 10, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug, or any mixture of two or more thereof, and one or more pharmaceutically acceptable carriers.
12. A kit, comprising:
1) a compound according to any one of claims 1 to 10 or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof, or a pharmaceutical composition according to claim 11; and
2) optionally packaging and/or instructions.
13. Use of a compound according to any one of claims 1 to 10, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a polymorph, a co-crystal, a solvate, a metabolite, a prodrug or any mixture of two or more thereof, for the manufacture of a medicament for the treatment of a disease associated with HPK1, in particular a tumor.
14. The use of claim 13, wherein the medicament further comprises an additional agent for treating a disease or disorder associated with HPK 1.
15. A process for the preparation of a compound according to any one of claims 1 to 10, according to the following scheme 1 or 2:
scheme 1:
Figure FDA0002557649660000081
wherein R is1、R2、R3X is as defined in any one of claims 1 to 10;
the first step is as follows: the compound I-1 is subjected to coupling, condensation or alkylation reaction to form a compound I-2, wherein LG1Is a leaving group such as halogen (e.g., Cl, Br or I), OTs, OTf, etc.;
the second step is that: carrying out coupling or nucleophilic substitution reaction on the compound I-2 to generate a compound I-3, wherein PG is an amino protecting group, such as Boc, Cbz, Bn, PMB and the like;
the third step: carrying out deprotection reaction on the compound I-3 to generate a compound I-4;
the fourth step: carrying out coupling, condensation or alkylation reaction on the compound I-4 to generate a target compound I;
scheme 2:
Figure FDA0002557649660000082
wherein R is1、R2、R3X is as defined in any one of claims 1 to 10;
the first step is as follows: the compound I-1 is subjected to coupling, condensation or alkylation reaction to form a compound I-2, wherein LG1Is a leaving group such as halogen (e.g., Cl, Br or I), OTs, OTf, etc.;
the second step is that: the compound I-2 is subjected to coupling or nucleophilic substitution reaction to generate the target compound I.
CN202010597780.8A 2020-06-28 2020-06-28 Pyrazolo ring compound, pharmaceutical composition containing same, preparation method and application thereof Pending CN113845531A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010597780.8A CN113845531A (en) 2020-06-28 2020-06-28 Pyrazolo ring compound, pharmaceutical composition containing same, preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010597780.8A CN113845531A (en) 2020-06-28 2020-06-28 Pyrazolo ring compound, pharmaceutical composition containing same, preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN113845531A true CN113845531A (en) 2021-12-28

Family

ID=78972095

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010597780.8A Pending CN113845531A (en) 2020-06-28 2020-06-28 Pyrazolo ring compound, pharmaceutical composition containing same, preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN113845531A (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022197641A1 (en) 2021-03-15 2022-09-22 Rapt Therapeutics, Inc. 1h-pyrazolo[3,4-d]pyrimidin-6-yl-amine derivatives as hematopoietic progenitor kinase 1 (hpk1) modulators and/or inhibitors for the treatment of cancer and other diseases
US11453681B2 (en) 2019-05-23 2022-09-27 Gilead Sciences, Inc. Substituted eneoxindoles and uses thereof
WO2023208127A1 (en) * 2022-04-27 2023-11-02 南京明德新药研发有限公司 Heteroaryl-substituted bicyclic compound and use thereof
US11897878B2 (en) 2018-10-31 2024-02-13 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds
US11925631B2 (en) 2018-10-31 2024-03-12 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11897878B2 (en) 2018-10-31 2024-02-13 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds
US11925631B2 (en) 2018-10-31 2024-03-12 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds
US11453681B2 (en) 2019-05-23 2022-09-27 Gilead Sciences, Inc. Substituted eneoxindoles and uses thereof
WO2022197641A1 (en) 2021-03-15 2022-09-22 Rapt Therapeutics, Inc. 1h-pyrazolo[3,4-d]pyrimidin-6-yl-amine derivatives as hematopoietic progenitor kinase 1 (hpk1) modulators and/or inhibitors for the treatment of cancer and other diseases
US11918582B2 (en) 2021-03-15 2024-03-05 Rapt Therapeutics, Inc. Pyrazole pyrimidine compounds and uses thereof
WO2023208127A1 (en) * 2022-04-27 2023-11-02 南京明德新药研发有限公司 Heteroaryl-substituted bicyclic compound and use thereof

Similar Documents

Publication Publication Date Title
EP3712151B1 (en) (s)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-5-amine as shp2 inhibitor for the treatment of cancer
CN111153901B (en) Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
CN111484480B (en) Polycyclic derivative inhibitor, preparation method and application thereof
CN113845531A (en) Pyrazolo ring compound, pharmaceutical composition containing same, preparation method and application thereof
WO2021169990A1 (en) Kras inhibitors for treating cancers
CN113795483A (en) Carboxamide-pyrimidine derivatives as SHP2 antagonists
EP2769980A1 (en) Pyrazoloquinoline derivative
EP2945623A1 (en) Hedgehog pathway signaling inhibitors and therapeutic applications thereof
KR20210099611A (en) Heteroaromatic derivative control agent, preparation method and use thereof
CN111315747A (en) Dihydropyrazolone pyrimidine compound and preparation method and application thereof
WO2019228341A1 (en) Inhibitor containing tricyclic derivative, preparation method therefor, and application thereof
KR20220061958A (en) Heterobicyclic amides as inhibitors of CD38
CN115605479A (en) Pyridoneopyrimidine derivative, preparation method thereof and application thereof in medicine
CN115710266A (en) Pyridopyrimidine derivative, preparation method thereof and application thereof in medicines
CN113754682B (en) Compound having macrocyclic structure and use thereof
WO2022068929A1 (en) Pyrimidinedione compound and use thereof
KR20220119702A (en) Pyridazinyl thiazolecarboxamide compound
AU2022280845A1 (en) Nitrogen-containing heterocyclic compound, preparation method therefor, and application thereof in medicines
CA3114147A1 (en) Fgfr4 inhibitor and use thereof
EP4223759A1 (en) Pyrazolopyridazinone compound, and pharmaceutical composition and use thereof
CN109790160B (en) Pyrido five-membered aromatic ring compound, preparation method and application thereof
CN115916768A (en) Sulfur-containing isoindoline derivative, preparation method and medical application thereof
US20220259210A1 (en) Pyrazolone-Fused Pyrimidine Compound, Preparation Method for Same and Applications Thereof
CN113993870A (en) Imidazo pyrimidine compound containing polycyclic group, preparation method and application thereof
CN114573584A (en) DNA-PK selective inhibitor and preparation method and application thereof

Legal Events

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