CN116265470A - Heterocyclic compound as HPK1 inhibitor and application thereof - Google Patents

Heterocyclic compound as HPK1 inhibitor and application thereof Download PDF

Info

Publication number
CN116265470A
CN116265470A CN202211624824.7A CN202211624824A CN116265470A CN 116265470 A CN116265470 A CN 116265470A CN 202211624824 A CN202211624824 A CN 202211624824A CN 116265470 A CN116265470 A CN 116265470A
Authority
CN
China
Prior art keywords
compound
membered heterocyclyl
alkyl
cycloalkyl
optionally
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
CN202211624824.7A
Other languages
Chinese (zh)
Inventor
胡治隆
刘力锋
唐锋
周峰
唐任宏
任晋生
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.)
Nanjing Zaiming Pharmaceutical Co ltd
Original Assignee
Jiangsu Simcere Pharmaceutical 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 Jiangsu Simcere Pharmaceutical Co Ltd filed Critical Jiangsu Simcere Pharmaceutical Co Ltd
Publication of CN116265470A publication Critical patent/CN116265470A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/056Ortho-condensed systems with two or more oxygen atoms as ring hetero atoms in the oxygen-containing ring
    • 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
    • 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
    • C07D471/12Heterocyclic 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 three hetero rings
    • C07D471/14Ortho-condensed systems
    • 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

Abstract

The invention provides a compound shown as a formula (I) or pharmaceutically acceptable salt thereof, a pharmaceutical composition, a preparation method thereof and application of the compound serving as an HPK1 inhibitor.

Description

Heterocyclic compound as HPK1 inhibitor and application thereof
The invention claims the priority and the rights of Chinese patent application with patent application number 202111552237.7 and the name of heterocyclic compound used as HPK1 inhibitor and application thereof, which are submitted to the China national intellectual property office on 12 and 17 days of 2021. The entire disclosure of the above-identified prior application is incorporated herein by reference in its entirety.
Technical Field
The invention relates to a novel heterocyclic compound or pharmaceutically acceptable salt thereof, a pharmaceutical composition containing the same and application of the heterocyclic compound or pharmaceutically acceptable salt thereof as an HPK1 inhibitor in preventing or treating related diseases.
Background
One of the main features of cancer is immune evasion capability. Tumor cells inhibit their recognition and attack by the body's immune system through a variety of complex mechanisms. Several strategies for tumor immunotherapy have been devised to counteract this immunosuppression, including mechanisms that interfere with negative regulatory effector T cell function, such as PD1/PDL1 immune checkpoint inhibitors, by blocking the interaction of PD1 and PDL1, to counteract T lymphocyte immunosuppression in PDL 1-highly expressing cancer cells, and antibody development against PD1 or PDL1 inhibitors has also been demonstrated for clinical benefit in a variety of cancer types. In addition, therapeutic antibodies that block the interaction between CD80/CD86 and the T cell co-inhibitory receptor (CTLA-4) can promote T cell expansion in lymphoid tissues at various levels. In addition to these cell surface related proteins, intracellular signaling was found to be involved in immune down regulation, where HPK1 (hematopoietic progenitor kinase 1, also known as MAP4K 1) specifically expressed in hematopoietic cells is a serine/threonine kinase, primarily involved in immune down regulation in cells.
Studies have found that inactivation of HPK1 in human and mouse cells is often accompanied by the development of autoimmune diseases, suggesting that HPK1 regulates immune tolerance in the body. For example, peripheral mononuclear cells (PBMC) from psoriatic arthritis patients and T cells from systemic lupus erythematosus patients all found down-regulation of HPK1 expression (J Autoimmun 2011,37 (3), 180-9); mouse model experiments found that HPK 1-deleted mice were more prone to autoimmune meningitis (Nat Immunol 2007,8 (1), 84-91). In vitro studies demonstrated that antigen stimulation of T and B lymphocytes derived from HPK1 deletions had a stronger activating effect (Cancer immunol. Immunother.2010,59 (3), 419-429), indicating negative regulation of T and B lymphocyte function by HPK 1. In addition, HPK 1-deleted dendritic cells (DC cells) exhibited more potent antigen presentation and T cell activation properties, suggesting that HPK1 is also involved in immune regulation of DC cells.
Upon activation of the T Cell Receptor (TCR) and B Cell Receptor (BCR), cytoplasmic HPK1 is recruited to the vicinity of the cell membrane to be activated, which activates the HPK1 phosphorylates the adaptor protein SLP76 or LAT, thus activating SLP76 as a docking site for the negative regulator protein 14-3-3 pi, mediating the ubiquitination of SLP76, ultimately leading to instability of the TCR signal complex, thus down-regulating TCR signal (J.cell biol.2011,195 (5), 839-853). It has also been found that HPK1 can be activated by PGE2 (prostaglandin E2) in a PKA-dependent manner, and possibly even by immunosuppressive factors expressed by tumor cells (Blood 2003,101 (9), 3687-3689).
HPK1 compared with wild type -/- The mice show stronger growth inhibition effect on the growth of inoculated isogenic lung cancer tumors. Demonstration of HPK by anti-tumor immune response studies on T cell transplantation mouse models -/- The strong anti-tumor effect of knockout is at least partially T cell dependent. The contribution of dendritic cells to the antitumor activity is also achieved by the fact that dendritic cells are derived from HPK1 -/- DC cell transplantation experiments of deficient mouse bone marrow were confirmed (J.Immunol.2009, 182 (10), 6187-61). Recently, it was found that the HPK1 transgenic mice, which catalyze the inactivation of enzymes, are also effective in inhibiting glioblastoma GL261 growth and enhancing the efficacy of anti-PD 1 treatment of MC38 tumors, as compared to wild-type HPK transgenic mice. Therefore, HPK1 is a potential anti-tumor therapeutic target, and the development of small molecule inhibitors against HPK1 kinase, whether as single drugs or in combination with other immunomodulatory therapeutic strategies, is expected to be effective in anti-tumor therapy.
Disclosure of Invention
The invention provides a compound shown in a formula (I) or pharmaceutically acceptable salt thereof:
Figure BDA0004003727240000021
wherein, the liquid crystal display device comprises a liquid crystal display device,
X 1 selected from N or CR 1 ,R 1 Selected from H, halogen, CN, SH, NH 2 、C 1 -C 10 Alkyl, C 1 -C 10 Alkoxy, C 3 -C 10 Cycloalkyl, C 3 -C 10 Cycloalkyloxy, 4-7 membered heterocyclyl, 4-7 membered heterocyclyloxy or
Figure BDA0004003727240000022
Each R is 1a Identical or different, R is the same or different 1a Independently selected from C 1 -C 10 Alkyl, C 3 -C 10 Cycloalkyl or 4-7 membered heterocyclyl, said SH, NH 2 、C 1 -C 10 Alkyl, C 1 -C 10 Alkoxy, C 3 -C 10 Cycloalkyl, C 3 -C 10 Cycloalkyloxy, 4-7 membered heterocyclyl or 4-7 membered heterocyclyloxy optionally substituted with R 1b Substitution;
X 2 selected from N or CR 2 ,R 2 Selected from H, halogen, CN, SH, NH 2 、C 1 -C 10 Alkyl, C 1 -C 10 Alkoxy, C 3 -C 10 Cycloalkyl, C 3 -C 10 Cycloalkyloxy, 4-7 membered heterocyclyl or 4-7 membered heterocyclyloxy, said SH, NH 2 、C 1 -C 10 Alkyl, C 1 -C 10 Alkoxy, C 3 -C 10 Cycloalkyl, C 3 -C 10 Cycloalkyloxy, 4-7 membered heterocyclyl or 4-7 membered heterocyclyloxy optionally substituted with R 2b Substitution;
alternatively, X 1 、X 2 Taken together form a 4-14 membered heterocyclyl or a 5-6 membered heteroaryl, said 4-14 membered heterocyclyl or 5-6 membered heteroaryl optionally being substituted with R 2b Substitution; when X is 1 、X 2 When the ring is not formed,X 1 、X 2 with only double bonds, when X 1 、X 2 X is the same as X in the ring formation 1 、X 2 Connected by single bond or double bond;
R 3 、R 4 each independently selected from H, halogen, CN, OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl, said OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl optionally being substituted by R 3b Substitution;
R 5 selected from OH or NH 2 Said OH or NH 2 Optionally by R 5b Substitution; and when R is 5 Selected from optionally R 5b Substituted NH 2 When X is 1 、X 2 Taken together form a 4-14 membered heterocyclyl or a 5-6 membered heteroaryl, said 4-14 membered heterocyclyl or 5-6 membered heteroaryl optionally being substituted with R 2b Substituted, or X 1 Selected from CR 1 ,R 1 Selected from SH or
Figure BDA0004003727240000023
Each R is 1a Identical or different, R is the same or different 1a Independently selected from C 1 -C 10 Alkyl, C 3 -C 10 Cycloalkyl or 4-7 membered heterocyclyl, said SH optionally being substituted by R 1b Substituted, X 2 Selected from N;
X 3 、X 4 selected from any one of the following:
1)X 3 selected from N or CR 6 ,X 4 Selected from N or CR 7 ,X 3 、X 4 Connected by double bonds, R 6 、R 7 Each independently selected from H, halogen, CN, OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl, said OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl optionally being substituted by R 6b Substitution; or alternatively, the process may be performed,
2)X 3 、X 4 one of them is selected from C (=O) and the other is selected from NR 8 ,X 3 、X 4 Connected by a single bond, R is 8 Selected from H, C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl, said C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl optionally being substituted by R 8b Substitution; or alternatively, the process may be performed,
3)X 3 、X 4 are joined together to form a 4-14 membered heterocyclyl or 5-6 membered heteroaryl, X 3 、X 4 Linked by a double bond, said 4-14 membered heterocyclyl or 5-6 membered heteroaryl optionally being bound by R 9b Substitution;
each R is 1b Or R is 3b Independently selected from deuterium, F, cl, br, I, CN, =o, OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl, C 2 -C 3 Alkenyl, C 2 -C 3 Alkynyl or 4-7 membered heterocyclyl, said OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl, C 2 -C 3 Alkenyl, C 2 -C 3 Alkynyl or 4-7 membered heterocyclyl are optionally substituted by R c Substitution;
each R is 2b 、R 5b 、R 6b 、R 8b Or R is 9b Independently selected from F, cl, br, I, CN, =o, OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl, said OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl optionally being substituted by R c Substitution;
each R is c Independently selected from F, cl, br, I, CN, =o, OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl;
with the proviso that the compound of formula (I) does not comprise
Figure BDA0004003727240000031
Figure BDA0004003727240000032
In some embodiments, R 1 Selected from SH, NH 2 、C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 3 -C 6 Cycloalkyl, C 3 -C 6 Cycloalkyloxy, 4-7 membered heterocyclyl, 4-7 membered heterocyclyloxy or
Figure BDA0004003727240000033
The R is 1a Independently selected from C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl, said SH, NH 2 、C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 3 -C 6 Cycloalkyl, C 3 -C 6 Cycloalkyloxy, 4-7 membered heterocyclyl or 4-7 membered heterocyclyloxy optionally substituted with R 1b And (3) substitution.
In some embodiments, X 1 Selected from N or CR 1 ,R 1 Selected from SH, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 3 -C 6 Cycloalkyl or C 3 -C 6 Cycloalkyloxy, said SH, C 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, C 3 -C 6 Cycloalkyl or C 3 -C 6 Cycloalkyl oxy is optionally substituted with R 1b And (3) substitution.
In some embodiments, X 1 Selected from N or CR 1 ,R 1 Selected from SH, C 1 -C 6 Alkoxy, C 3 -C 6 Cycloalkyloxy or
Figure BDA0004003727240000034
The SH, C 1 -C 6 Alkoxy or C 3 -C 6 Cycloalkyl oxy is optionally substituted with R 1b And (3) substitution.
In some embodiments, X 1 Selected from N or CR 1 ,R 1 Selected from SH, C 1 -C 6 Alkoxy or C 3 -C 6 Cycloalkyloxy, said SH, C 1 -C 6 Alkoxy or C 3 -C 6 Cycloalkyl oxy is optionally substituted with R 1b And (3) substitution.
In some embodiments, X 1 Selected from N or CR 1 ,R 1 Selected from SH, methoxy, cyclopropyloxy or
Figure BDA0004003727240000035
Said SH, methoxy or cyclopropyloxy group being optionally substituted by R 1b And (3) substitution.
In some embodiments, X 1 Selected from N or CR 1 ,R 1 Selected from SH, methoxy or cyclopropyloxy, said SH, methoxy or cyclopropyloxy being optionally substituted by R 1b And (3) substitution.
In some embodiments, R 1a Selected from C 1 -C 3 An alkyl group. In some embodiments, R 1b Selected from deuterium, F, cl, br, I, CN, = O, C 1 -C 6 Alkyl, C 2 -C 3 Alkenyl or C 2 -C 3 Alkynyl groups.
In some embodiments, R 1b Selected from deuterium or C 1 -C 3 An alkyl group.
In some embodiments, X 1 Selected from CR 1 ,R 1 Selected from SCH 3 、OCH 3 、OCD 3 Cyclopropyloxy or
Figure BDA0004003727240000041
In some embodiments, X 1 Selected from N or CR 1 ,R 1 Selected from SCH 3 、OCH 3 、OCD 3 Or cyclopropyloxy.
In some embodiments, X 2 Selected from N.
In some embodiments, X 1 、X 2 Taken together form a 5-10 membered heterocyclyl or a 5-6 membered heteroaryl, said 5-10 membered heterocyclyl or 5-6 membered heteroaryl optionally being substituted with R 2b And (3) substitution.
In some embodiments, X 1 、X 2 Are joined together to form
Figure BDA0004003727240000042
Figure BDA0004003727240000043
Wherein Y is selected from CH 2 NH, O or S, said ≡>
Figure BDA0004003727240000044
Figure BDA0004003727240000045
Optionally by R 2b And (3) substitution.
In some embodiments, X 1 、X 2 Are joined together to form
Figure BDA0004003727240000046
Said->
Figure BDA0004003727240000047
Optionally by R 2b And (3) substitution.
In some embodiments, X 1 、X 2 Are joined together to form
Figure BDA0004003727240000048
Said->
Figure BDA0004003727240000049
Optionally by R 2b And (3) substitution.
In some embodiments, X 1 、X 2 Are joined together to form
Figure BDA00040037272400000410
In some embodiments, R 3 、R 4 Each independently selected from H, halogen, CN or C 1 -C 3 Alkyl, said C 1 -C 3 Alkyl is optionally substituted with R 3b And (3) substitution.
In some embodiments, R 3 、R 4 Each independently selected from H or optionally R 3b Substitution C 1 -C 3 An alkyl group.
In some embodiments, R 3 、R 4 Each independently selected from optionally R 3b Substitution C 1 -C 3 An alkyl group.
In some embodiments, R 3 、R 4 One of which is selected from H or methyl and the other of which is selected from methyl. In some embodiments, R 3 、R 4 Are all selected from methyl groups. In some embodiments, R 5 Selected from optionally R 5b Substituted OH.
In some embodiments, R 5 Selected from OH or NH 2 Said OH or NH 2 Optionally by R 5b Substitution; and when R is 5 Selected from optionally R 5b Substituted NH 2 When X is 1 、X 2 Taken together form a 4-14 membered heterocyclyl or a 5-6 membered heteroaryl, said 4-14 membered heterocyclyl or 5-6 membered heteroaryl optionally being substituted with R 2b And (3) substitution.
In some embodiments, R 5 Selected from OH or NH 2 Said OH or NH 2 Optionally by R 5b Substitution; and when R is 5 Selected from optionally R 5b Substituted NH 2 When X is 1 、X 2 Are joined together to form
Figure BDA0004003727240000051
Figure BDA0004003727240000052
Said->
Figure BDA0004003727240000053
Optionally by R 2b Substituted, or X 1 Selected from CR 1 ,R 1 Selected from SH or->
Figure BDA0004003727240000054
Each R is 1a Identical or different, R is the same or different 1a Independently selected from C 1 -C 3 Alkyl, said SH is C 1 -C 3 Alkyl substitution, X 2 Selected from the group consisting ofN。
In some embodiments, when R 5 Selected from optionally R 5b Substituted NH 2 When X is 1 、X 2 Taken together form a 5-10 membered heterocyclyl or a 5-6 membered heteroaryl, said 5-10 membered heterocyclyl or 5-6 membered heteroaryl optionally being substituted with R 2b And (3) substitution.
In some embodiments, R 5 Selected from OH or NH 2 Said OH or NH 2 Optionally by R 5b Substituted, X 1 、X 2 Are joined together to form
Figure BDA0004003727240000055
Figure BDA0004003727240000056
Wherein Y is selected from CH 2 NH, O or S, said ≡>
Figure BDA0004003727240000057
Figure BDA0004003727240000058
Optionally by R 2b And (3) substitution.
In some embodiments, R 5 Selected from OH or NH 2 Said OH or NH 2 Optionally by R 5b Substituted, X 1 、X 2 Are joined together to form
Figure BDA0004003727240000059
Said->
Figure BDA00040037272400000510
Figure BDA00040037272400000511
Optionally by R 2b And (3) substitution.
In some embodiments, each R 2b 、R 5b Each independently selected from F, cl, br, I, CN, C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl.
In some embodiments, R 5 Selected from OH or NH 2 ,X 1 、X 2 Are joined together to form
Figure BDA0004003727240000061
Figure BDA0004003727240000062
Wherein Y is selected from CH 2 NH, O or S.
In some embodiments, R 5 Selected from OH or NH 2 ,X 1 、X 2 Are joined together to form
Figure BDA0004003727240000063
Figure BDA0004003727240000064
In some embodiments, R 5 Selected from OH or NH 2 . In some embodiments, X 3 Selected from N or CR 6 ,X 4 Selected from N, X 3 、X 4 Connected by double bonds, R 6 Selected from C 1 -C 6 Alkyl or C 3 -C 6 Cycloalkyl group, the C 1 -C 6 Alkyl or C 3 -C 6 Cycloalkyl is optionally substituted with R 6b And (3) substitution.
In some embodiments, X 3 Selected from CR 6 ,X 4 Selected from N, X 3 、X 4 Connected by double bonds, R 6 Selected from C optionally substituted by halogen 1 -C 3 An alkyl group.
In some embodiments, X 3 Selected from CR 6 ,X 4 Selected from N, X 3 、X 4 Connected by double bonds, R 6 Selected from C optionally substituted by F 1 -C 3 An alkyl group.
In some embodiments, X 3 Selected from CR 6 ,X 4 Selected from N, X 3 、X 4 Connected by double bonds, R 6 Selected from C (CH) 3 ) 2 F。
In some casesIn embodiments, X 3 、X 4 Are joined together to form a 4-7 membered heterocyclic group, X 3 、X 4 Linked by a double bond, said 4-7 membered heterocyclic group optionally being R 9b And (3) substitution.
In some embodiments, X 3 、X 4 Are joined together to form a 5-6 membered heterocyclic group, X 3 、X 4 Linked by a double bond, said 5-6 membered heterocyclic group optionally being R 9b And (3) substitution.
In some embodiments, R 9b Selected from F, cl, br, I, CN, =o or C 1 -C 3 An alkyl group.
In some embodiments, X 3 、X 4 Are joined together to form
Figure BDA0004003727240000065
In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt thereof is selected from the group consisting of a compound of formula (I-1), formula (I-2), formula (I-3), formula (I-4), or formula (I-5), or a pharmaceutically acceptable salt thereof:
Figure BDA0004003727240000066
Figure BDA0004003727240000071
wherein, the liquid crystal display device comprises a liquid crystal display device,
n is selected from 0, 1, 2, 3, 4 or 5; r is R 5 Selected from OH or NH 2 Said OH or NH 2 Optionally by R 5b Substitution; the R is 2b 、R 5b As defined above.
In some embodiments, n in the compounds of formula (I-1), formula (I-2), formula (I-3), formula (I-4), or formula (I-5) is selected from 0, 1, or 2.
In some embodiments, n in the compounds of formula (I-1), formula (I-2), formula (I-3), formula (I-4), or formula (I-5) is selected from 0.
In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt thereof is selected from the compounds of formula (I-6) below:
Figure BDA0004003727240000072
the X is 1 、X 2 、X 3 、X 4 、R 3 、R 4 As defined above.
In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt thereof is selected from the compounds of formula (I-7) below:
Figure BDA0004003727240000073
the X is 1 、X 2 、X 3 、X 4 、R 3 、R 4 As defined above.
In some embodiments, X in the compound of formula (I-7) 1 Selected from N or CR 1 ,R 1 Selected from SH or
Figure BDA0004003727240000074
The SH is optionally substituted by R 1b And (3) substitution.
In some embodiments, X in the compound of formula (I-7) 1 Selected from CR 1 ,R 1 Selected from SCH 3 Or (b)
Figure BDA0004003727240000075
In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt thereof is selected from the following compounds or pharmaceutically acceptable salts thereof:
Figure BDA0004003727240000081
Figure BDA0004003727240000091
the invention also provides a pharmaceutical composition which comprises the compound shown in the formula (I) or pharmaceutically acceptable salt thereof and pharmaceutically acceptable auxiliary materials.
Further, the invention relates to application of a compound shown in the formula (I) or pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof in preparing a medicament for preventing or treating HPK1 related diseases.
Further, the invention relates to application of a compound shown in the formula (I) or pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof in preparing a medicament for preventing or treating tumors.
Further, the invention relates to the use of a compound represented by formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for preventing or treating HPK 1-related diseases.
Further, the invention relates to the use of a compound represented by formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the prevention or treatment of tumors.
Further, the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for preventing or treating HPK 1-related diseases.
Further, the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, for preventing or treating a tumor.
The invention also relates to a method of treating a disease associated with HPK1 comprising administering to a patient a therapeutically effective dose of a pharmaceutical formulation comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof as described herein.
The invention also relates to a method of treating a tumour, which comprises administering to a patient a therapeutically effective dose of a pharmaceutical formulation comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof as described herein.
In some embodiments, the HPK 1-associated disease is selected from a tumor.
Definition and description of terms
Unless otherwise indicated, the terms used in the present invention have the following meanings, and the groups and term definitions recited in the present invention, including as examples, exemplary definitions, preferred definitions, definitions recited in tables, definitions of specific compounds in the examples, and the like, may be arbitrarily combined and combined with each other. A particular term, unless otherwise defined, shall not be construed as being ambiguous or otherwise unclear, but shall be construed in accordance with the ordinary meaning in the art. When trade names are presented herein, it is intended to refer to their corresponding commercial products or active ingredients thereof.
Herein, a method of manufacturing a semiconductor device
Figure BDA0004003727240000101
Representing the ligation site.
The graphic representation of racemates or enantiomerically pure compounds herein is from Maehr, J.chem. Ed.1985, 62:114-120. Unless otherwise indicated, wedge keys and virtual wedge keys are used
Figure BDA0004003727240000102
Representing the absolute configuration of a stereogenic center, using the black real and virtual keys +. >
Figure BDA0004003727240000103
Representing the relative configuration of a stereocenter (e.g., the cis-trans configuration of a alicyclic compound).
The term "tautomer" refers to a functional group isomer that results from the rapid movement of an atom in a molecule at two positions. The compounds of the present invention may exhibit tautomerism. Tautomeric compounds may exist in two or more interconvertible species. Tautomers generally exist in equilibrium and attempts to isolate individual tautomers often result in a mixture whose physicochemical properties are consistent with the mixture of compounds. The location of the equilibrium depends on the chemical nature of the molecule. For example, among many aliphatic aldehydes and ketones such as acetaldehyde, the ketone type predominates; whereas, among phenols, the enol form is dominant. The present invention encompasses all tautomeric forms of the compounds.
The term "stereoisomers" refers to isomers arising from the spatial arrangement of atoms in a molecule, and includes cis-trans isomers, enantiomers and diastereomers.
The compounds of the present invention may have asymmetric atoms such as carbon atoms, sulfur atoms, nitrogen atoms, phosphorus atoms or asymmetric double bonds, and thus the compounds of the present invention may exist in specific geometric or stereoisomeric forms. Particular geometric or stereoisomeric forms may be cis and trans isomers, E and Z geometric isomers, (-) -and (+) -enantiomers, (R) -and (S) -enantiomers, diastereomers, (D) -isomers, (L) -isomers, and racemic or other mixtures thereof, such as enantiomerically or diastereomerically enriched mixtures, all of which fall within the definition of compounds of the invention. Additional asymmetric carbon atoms, asymmetric sulfur atoms, asymmetric nitrogen atoms, or asymmetric phosphorus atoms may be present in the substituents such as alkyl groups, and all such isomers and mixtures thereof are included within the definition of compounds of the invention. The asymmetric atom-containing compounds of the present invention may be isolated in optically pure form or in racemic form, which may be resolved from racemic mixtures or synthesized by using chiral starting materials or chiral reagents.
The term "substituted" means that any one or more hydrogen atoms on a particular atom is substituted with a substituent, provided that the valence of the particular atom is normal and the substituted compound is stable. When the substituent is oxo (i.e., =o), meaning that two hydrogen atoms are substituted, oxo does not occur on the aromatic group.
The term "optionally" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. For example, ethyl "optionally" substituted with halogen means that ethyl may be unsubstituted (CH 2 CH 3 ) Monosubstituted (CH) 2 CH 2 F、CH 2 CH 2 Cl, etc.), polysubstituted (CHFCH 2 F、CH 2 CHF 2 、CHFCH 2 Cl、CH 2 CHCl 2 Etc.) or fully substituted (CF) 2 CF 3 、CF 2 CCl 3 、CCl 2 CCl 3 Etc.). It will be appreciated by those skilled in the art that for any group comprising one or more substituents, no substitution or pattern of substitution is introduced that is sterically impossible and/or synthetic.
When any variable (e.g. R a 、R b ) Where the composition or structure of a compound occurs more than once, its definition is independent in each case. For example, if a group is substituted with 2R b Substituted, each R b There are independent options.
When the number of one linking group is 0, such as- (CH) 2 ) 0 -, indicating that the linking group is a bond.
When one of the variables is selected from the group consisting of a chemical bond or is absent, the two groups representing its attachment are directly linked, e.g., when L in A-L-Z represents a bond, it is meant that the structure is actually A-Z.
The linking group referred to herein is arbitrary in its linking direction unless the linking direction is indicated. For example when building blocks
Figure BDA0004003727240000111
L of (3) 1 Selected from "C 1 -C 3 alkylene-O ", in which case L 1 Either the rings Q and R can be connected in a direction from left to right 1 Form a "ring Q-C 1 -C 3 alkylene-O-R 1 ", rings Q and R may be connected in a right-to-left direction 1 Form a "ring Q-O-C 1 -C 3 Alkylene group-R 1 ”。
When the bond of a substituent is cross-linked to two atoms on a ring, the substituent may be bonded to any atom on the ring. For example, structural units
Figure BDA0004003727240000112
R represents 5 Substitution may occur at any position on the phenyl ring.
Herein, keys depicted by solid lines and dashed lines
Figure BDA0004003727240000113
Represents a single bond or a double bond.
C herein m -C n Refers to having an integer number of carbon atoms in the m-n range. For example "C 1 -C 10 By "is meant that the group may have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, 6 carbon atoms, 7 carbon atoms, 8 carbon atoms, 9 carbon atoms, or 10 carbon atoms.
The term "alkyl" refers to a compound of the formula C n H 2n+1 The alkyl group may be linear or branched. The term "C 1 -C 10 Alkyl "is understood to mean a straight-chain or branched saturated hydrocarbon radical having 1,2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms. Specific examples of the alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, 2-methylbutyl, 1-ethylpropyl, 1, 2-dimethylpropyl, neopentyl, 1-dimethylpropyl, and 4-methylpentyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 2-ethylbutyl, 1-ethylbutyl, 3-dimethylbutyl, 2-dimethylbutyl, 1-dimethylbutyl, 2, 3-dimethylbutyl, 1, 3-dimethylbutyl, or 1, 2-dimethylbutyl, and the like; the term "C 1 -C 6 Alkyl "is understood to mean an alkyl group having 1 to 6 carbon atoms, specific examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, neopentyl, hexyl, 2-methylpentyl, and the like. The term "C 1 -C 3 Alkyl "is understood to mean a straight-chain or branched saturated alkyl group having 1 to 3 carbon atoms. The "C 1 -C 10 Alkyl "may contain" C 1 -C 6 Alkyl "or" C 1 -C 3 Alkyl "and the like, said" C 1 -C 6 The alkyl group may further comprise "C 1 -C 3 An alkyl group.
The term "alkoxy" refers to a group generated by the loss of a hydrogen atom on a hydroxyl group of a straight or branched chain alcohol, and is understood to be "alkyloxy" or "alkyl-O-". The term "C 1 -C 10 Alkoxy "is understood to mean" C 1 -C 10 Alkyloxy "or" C 1 -C 10 alkyl-O- "; the term "C 1 -C 6 Alkoxy "is understood to mean" C 1 -C 6 Alkyloxy "or" C 1 -C 6 alkyl-O- ". The "C 1 -C 10 Alkoxy "may contain" C 1 -C 6 Alkoxy "and" C 1 -C 3 Alkoxy "and the like, said" C 1 -C 6 Alkoxy groups may further comprise "C 1 -C 3 An alkoxy group.
The term "alkenyl" refers to an unsaturated aliphatic hydrocarbon group consisting of carbon and hydrogen atoms, straight or branched chain, and having at least one double bond. The term "C 2 -C 3 Examples of alkenyl "include vinyl, (E) -2-methylvinyl, (Z) -2-methylvinyl, and the like.
The term "alkynyl" refers to a straight or branched chain unsaturated aliphatic hydrocarbon group consisting of carbon and hydrogen atoms having at least one triple bond. The term "C 2 -C 3 Examples of alkynyl groups include ethynyl (-C.ident.CH), prop-1-ynyl (-C.ident.CCH) 3 ) Etc.
The term "cycloalkyl" refers to a carbocycle that is fully saturated and exists as a single ring, fused ring, bridged ring, or spiro ring, etc. Unless otherwise indicated, the carbocycle is typically a 3 to 12 membered ring. The term "C 3 -C 10 Cycloalkyl "is understood to mean a saturated monocyclic, fused, spiro or bridged ring having 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms. Specific examples of the cycloalkyl group include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, norbornyl (bicyclo [ 2.2.1)]Heptyl), bicyclo [2.2.2]Octyl, adamantyl, spiro [4.5 ]]Decyl, and the like. The term "C 3 -C 10 Cycloalkyl "may contain" C 3 -C 6 Cycloalkyl ", the term" C 3 -C 6 Cycloalkyl "is understood to mean a saturated monocyclic or bicyclic hydrocarbon ring having 3, 4, 5, or 6 carbon atoms, specific examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
The term "cycloalkyloxy" is understood as "cycloalkyl-O-".
The term "heterocyclyl" refers to a fully saturated or partially saturated monocyclic, fused, spiro, or bridged ring radical having 1, 2, 3, 4, or 5 heteroatoms or groups of heteroatoms (i.e., groups of heteroatoms) in the ring atoms, including but not limited to nitrogen (N), oxygen (O), sulfur (S), phosphorus (P), boron (B), and-S (=o) 2 -、-S(=O)-、-P(=O) 2 -P (=o) -, -NH-, -S (=o) (=nh) -, -C (=o) NH-, or-NHC (=o) NH-, etc. The term "4-14 membered heterocyclyl" refers to a heterocyclyl having a number of ring atoms of 4,5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 and having from 1 to 5 heteroatoms or groups of heteroatoms independently selected from those described above. "4-14 membered heterocyclic group" may include the ranges of "4-7 membered heterocyclic group", "5-6 membered heterocyclic group", and the like. The term "4-7 membered heterocyclic group" means a heterocyclic group having 4,5, 6 or 7 ring atoms and containing 1, 2, 3,4 or 5 heteroatoms or groups independently selected from the group consisting of the above. The term "5-6 membered heterocyclic group" means a heterocyclic group having 5 or 6 ring atoms and containing 1, 2, 3 or 4 heteroatoms or groups of heteroatoms independently selected from those described above in the ring atoms. Specific examples of 4-membered heterocyclyl groups include, but are not limited to, azetidinyl or oxetanyl; specific examples of 5-membered heterocyclyl groups include, but are not limited to, tetrahydrofuranyl, dioxolyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl, 4, 5-dihydro-oxazolyl, or 2, 5-dihydro-1H-pyrrolyl; specific examples of 6 membered heterocyclyl groups include, but are not limited to, tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, trithianyl, tetrahydropyridinyl or 4H- [1,3,4 ] ]Thiadiazinyl; specific examples of 7-membered heterocyclyl groups include, but are not limited to, diazepinyl. The heterocyclic group may also be a bicyclic hetero ringCyclic and tricyclic heterocyclic groups, wherein specific examples of 5,5 membered bicyclic groups include, but are not limited to, hexahydrocyclopenta [ c ]]Pyrrol-2 (1H) -yl; specific examples of 5,6 membered bicyclo groups include, but are not limited to, hexahydropyrrolo [1,2-a ]]Pyrazin-2 (1H) -yl, 5,6,7, 8-tetrahydro- [1,2,4]Triazolo [4,3-a ]]Pyrazinyl or 5,6,7, 8-tetrahydroimidazo [1,5-a ]]And pyrazinyl. "heterocyclyl" in the present invention may include "heterocycloalkyl", for example "4-7 membered heterocyclyl" may include "4-7 membered heterocycloalkyl", and the like. In the present invention, although some bicyclic heterocyclic groups contain a benzene ring or a heteroaryl ring in part, the heterocyclic groups as a whole are not aromatic.
The term "heterocyclyloxy" is understood to mean "heterocyclyl-O-".
The term "heteroaryl" refers to a monocyclic or fused polycyclic aromatic ring system containing at least one ring atom selected from N, O, S, the remaining ring atoms being aromatic ring groups of C. The term "5-10 membered heteroaryl" is understood to include such mono-or bicyclic aromatic ring systems: it has 5,6,7,8, 9 or 10 ring atoms, in particular 5 or 6 or 9 or 10 ring atoms, and it contains 1 to 5, preferably 1 to 3 heteroatoms independently selected from N, O and S. In particular, the heteroaryl group is selected from thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl and the like, and their benzo derivatives, such as benzofuranyl, benzothienyl, benzothiazolyl, benzoxazolyl, benzisoxazolyl, benzimidazolyl, benzotriazole, indazolyl, indolyl, isoindolyl and the like; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, or the like, and their benzo derivatives, such as quinolinyl, quinazolinyl, or isoquinolinyl, or the like; or an axcinyl group, an indolizinyl group, a purinyl group, etc., and their benzo derivatives; or cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthyridinyl, pteridinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, or phenoxazinyl, and the like. The term "5-6 membered heteroaryl" refers to an aromatic ring system having 5 or 6 ring atoms and which contains 1 to 3, preferably 1 to 2 heteroatoms independently selected from N, O and S.
The term "halo" or "halogen" refers to fluorine, chlorine, bromine or iodine.
The term "therapeutically effective amount" means an amount of a compound of the invention that (i) treats or prevents a particular disease, condition, or disorder, (ii) alleviates, ameliorates, or eliminates one or more symptoms of a particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of a particular disease, condition, or disorder described herein. The amount of the compound of the present invention that constitutes a "therapeutically effective amount" will vary depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but can be routinely determined by one of ordinary skill in the art based on his own knowledge and disclosure.
The term "pharmaceutically acceptable" is intended to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The term "pharmaceutically acceptable salt" refers to salts of pharmaceutically acceptable acids or bases, including salts of compounds with inorganic or organic acids, and salts of compounds with inorganic or organic bases.
The term "pharmaceutical composition" refers to a mixture of one or more compounds of the invention or salts thereof and pharmaceutically acceptable excipients. The purpose of the pharmaceutical composition is to facilitate the administration of the compounds of the invention to an organism.
The term "pharmaceutically acceptable excipients" refers to those excipients which do not significantly stimulate the organism and which do not impair the biological activity and properties of the active compound. Suitable excipients are well known to the person skilled in the art, such as carbohydrates, waxes, water soluble and/or water swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like.
The words "comprise" or "include" and variations thereof such as "comprises" or "comprising" are to be interpreted in an open, non-exclusive sense, i.e. "including but not limited to.
The invention also includes isotopically-labeled compounds of the invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic weight or mass number different from the atomic weight or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine, and chlorine, such as, respectively 2 H、 3 H、 11 C、 13 C、 14 C、 13 N、 15 N、 15 O、 17 O、 18 O、 31 P、 32 P、 35 S、 18 F、 123 I、 125 I and 36 cl, and the like.
Certain isotopically-labeled compounds of the invention (e.g., with 3 H is H 14 C-tag) can be used in compound and/or substrate tissue distribution analysis. Tritiation (i.e 3 H) And carbon-14 (i.e 14 C) Isotopes are particularly preferred for their ease of preparation and detectability. Positron emitting isotopes, such as 15 O、 13 N、 11 C and C 18 F can be used in Positron Emission Tomography (PET) studies to determine substrate occupancy. Isotopically-labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the schemes and/or examples below by substituting an isotopically-labeled reagent for an non-isotopically-labeled reagent.
The pharmaceutical compositions of the present invention may be prepared by combining the compounds of the present invention with suitable pharmaceutically acceptable excipients, for example, in solid, semi-solid, liquid or gaseous formulations such as tablets, pills, capsules, powders, granules, ointments, emulsions, suspensions, suppositories, injections, inhalants, gels, microspheres, aerosols and the like.
Typical routes of administration of the compounds of the invention or pharmaceutically acceptable salts thereof or pharmaceutical compositions thereof include, but are not limited to, oral, rectal, topical, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, intramuscular, subcutaneous, intravenous administration.
The pharmaceutical compositions of the present invention may be manufactured by methods well known in the art, such as conventional mixing, dissolving, granulating, emulsifying, lyophilizing, and the like.
In some embodiments, the pharmaceutical composition is in oral form. For oral administration, the pharmaceutical compositions may be formulated by mixing the active compound with pharmaceutically acceptable excipients well known in the art. These excipients enable the compounds of the present invention to be formulated into tablets, pills, troches, dragees, capsules, liquids, gels, slurries, suspensions and the like for oral administration to a patient.
The solid oral compositions may be prepared by conventional mixing, filling or tabletting methods. For example, it can be obtained by the following method: the active compound is mixed with solid auxiliary materials, the resulting mixture is optionally milled, if desired with other suitable auxiliary materials, and the mixture is then processed to granules, giving a tablet or dragee core. Suitable excipients include, but are not limited to: binders, diluents, disintegrants, lubricants, glidants or flavoring agents, and the like.
The pharmaceutical compositions may also be suitable for parenteral administration, such as sterile solutions, suspensions or lyophilized products in suitable unit dosage forms.
In all methods of administration of the compounds of formula I described herein, the dosage administered is from 0.01mg/kg to 200mg/kg body weight, preferably from 0.05mg/kg to 50mg/kg body weight, more preferably from 0.1mg/kg to 30mg/kg body weight, either alone or in divided doses.
The compounds of the present invention may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combining them with other chemical synthetic methods, and equivalent alternatives well known to those skilled in the art, preferred embodiments including but not limited to the examples of the present invention.
The chemical reactions of the embodiments of the present invention are accomplished in a suitable solvent that is compatible with the chemical changes of the present invention and the reagents and materials required therefor. In order to obtain the compounds of the present invention, it is sometimes necessary for a person skilled in the art to modify or select the synthesis steps or reaction schemes on the basis of the embodiments already present.
The invention adopts the following abbreviations:
Figure BDA0004003727240000141
Detailed Description
The invention is described in detail below by way of examples, which are not meant to limit any disadvantages of the invention. The present invention has been described in detail herein, and specific embodiments thereof are also disclosed, it will be apparent to those skilled in the art that various changes and modifications can be made to the specific embodiments of the invention without departing from the spirit and scope of the invention. All reagents used in the present invention are commercially available and can be used without further purification.
Unless otherwise indicated, the ratio of the mixed solvent is a volume mixing ratio.
Unless otherwise indicated,% refers to weight percent wt%.
The compounds being obtained by hand or by hand
Figure BDA0004003727240000142
Software naming, commercial compounds are referred to by vendor catalog names.
The structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) and/or Mass Spectrometry (MS). The unit of NMR shift is 10 -6 (ppm). The solvent for NMR measurement is deuterated dimethyl sulfoxide, deuterated chloroform, deuterated methanol, etc., and the internal standard is Tetramethylsilane (TMS); IC (integrated circuit) 50 "means half inhibition concentration" means concentration at which half of the maximum inhibition effect is achieved.
The eluent or mobile phase can be a mixed eluent or mobile phase consisting of two or more solvents, the ratio of which is the volume ratio of the solvents, for example, 0-10% methanol/dichloromethane represents that the volume ratio of methanol to dichloromethane in the mixed eluent or mobile phase is 0:100-10:100.
Synthesis of intermediate M-1:
Figure BDA0004003727240000151
step 1:
the starting material M-1-1 (10 g) was dissolved in DMF (100 mL) and N-bromosuccinimide (NBS, 11.8 g) was added in portions. After the reaction solution was reacted at 25℃for 2 hours, the reaction solution was poured into 50mL of water, and the solid was precipitated and filtered, and after washing the cake three times with water (5 mL), the cake was dissolved in toluene (100 mL) and concentrated to give a crude compound M-1-2 (12 g).
Step 2:
compound M-1-2 (12 g) was dissolved in POCl 3 (100g) The reaction was continued for 3 hours after the reaction temperature was gradually increased from 25℃to 110 ℃. The reaction mixture was cooled to 25 ℃ and concentrated under reduced pressure, and after dissolving the residue in dichloromethane (50 mL), the residue was slowly added to a saturated aqueous sodium bicarbonate solution (200 mL), the organic phase was dried over anhydrous sodium sulfate by extraction with dichloromethane (100 ml×3), filtered, and the filtrate was concentrated to give crude intermediate M-1 (10 g).
Synthesis of intermediate M-2:
Figure BDA0004003727240000152
step 1:
bis (cyclopentadienyl) zirconium hydroxide (CP 2 ZrHCl,1.2 g) was dispersed in a solution of dichloromethane (40 mL) and starting material M-2-1 (6 g) and pinacol biborate (9.8 g) were slowly added at 0deg.C. The reaction mixture was warmed to 45℃and cooled to room temperature after 48 hours of reaction. The reaction solution was concentrated under reduced pressure, and purified and separated by silica gel column chromatography (petroleum ether: ethyl acetate=100:1) to give compound M-2-2 (10 g).
Step 2:
under the protection of nitrogen, the compound M-2-2 (5 g) is dispersed in THF (50 mL) and water (12 mL), and the compound M-2-3 (5.9 g) and phosphorus are added in sequencePotassium acid (15.8 g) and Pd (dppf) Cl 2 DCM (1 g). The reaction mixture was reacted at 70℃for 2 hours, then cooled to room temperature, dried over anhydrous sodium sulfate, concentrated, and purified by silica gel column chromatography (petroleum ether: ethyl acetate=20:1) to give compound M-2-4 (4 g). LC-MS (ESI) m/z=222.2 [ M+H ] ] +
Step 3:
compound M-2-4 (4 g) was dissolved in trifluoromethanesulfonic acid (TfOH, 30 mL) at room temperature. After the reaction solution was heated to 80 ℃ and reacted for 3 hours, the reaction solution was cooled to room temperature, the pH of the reaction solution was adjusted to 7-8 with saturated sodium carbonate, extracted with dichloromethane (100 ml×3), the organic phase was dried over anhydrous sodium sulfate and filtered, and concentrated under reduced pressure to give compound M-2-5 (3.8 g). LC-MS (ESI) m/z=208.1 [ M+H ]] +
Step 4:
compound M-2-5 (3.8 g) and pyridine hydrochloride (10.5 g) were mixed, warmed to 150 ℃ and reacted for 30 minutes, cooled to room temperature, the reaction solution was poured into water (150 mL), extracted with dichloromethane (100 mL x 3), and the organic phase was dried over anhydrous sodium sulfate and filtered, and concentrated under reduced pressure to give a crude product. This crude product was dispersed in toluene (20 mL) and 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU, 8 g) was added. After heating to 100 ℃ for 12 hours, cooling to room temperature, adjusting the pH of the reaction solution to 7-8 by using an aqueous hydrochloric acid solution (1N), extracting with dichloromethane (100 ml x 3), drying and filtering an organic phase anhydrous sodium sulfate, and concentrating under reduced pressure to obtain a compound M-2-6 (3.2 g).
Step 5:
compound M-2-6 (3.2 g) was dispersed in phosphorus oxychloride (60 mL) at room temperature. After 1 hour of reaction at 100 ℃, the reaction solution was cooled to room temperature, the solvent was removed by concentration, the pH of the residue was adjusted to 8-9 with saturated sodium carbonate solution, extracted with dichloromethane (100 mL. Times.3), the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure and purified by liquid chromatography (column: flash Spherical C; mobile phase: A: water; B: acetonitrile, B%:0% -40%,50 mL/min) to give Compound M-2-7 (650 mg). LC-MS (ESI) m/z=212.0 [ M+H ] ] +
Step 6:
compounds M-2-7 (650 mg) was dissolved in N-methylpyrrolidone (NMP, 20 mL), and 2, 4-dimethoxybenzyl amine (DMBNH) 2 770 mg) and N, N-diisopropylethylamine (DIEA, 780 mg). The reaction mixture was heated to 80℃and reacted for 2 hours, followed by concentrating the reaction mixture under reduced pressure to give Compound M-2-8 (1.1 g). LC-MS (ESI) m/z=311.2 [ M+H ]] +
Step 7:
compound M-2-8 (1.1 g) was dissolved in dioxane hydrochloride solution (1 mol/L,20 mL), heated to 80℃for 2 hours, cooled to room temperature, concentrated under reduced pressure to remove the solvent, the residue was pH adjusted to 8-9 with saturated sodium carbonate solution, extracted with ethyl acetate (50 mL. Times.3), the organic phase was dried over anhydrous sodium sulfate and filtered, and concentrated under reduced pressure to give intermediate M-2 (400 mg). LC-MS (ESI) m/z=193.1 [ M+H ]] +
Synthesis of intermediate M-3:
Figure BDA0004003727240000161
step 1:
compound M-3-1 (2 g) and compound M-3-2 (1.15 g) were dissolved in EtOH (15 mL) and TEA (3.23 g) was added dropwise at 0deg.C. After 30 min, the reaction was allowed to warm to room temperature for 12 h, the reaction was concentrated, the residue was taken up in water (100 ml), extracted with ethyl acetate (50 ml x 3), dried over anhydrous sodium sulfate and filtered, and concentrated under reduced pressure to give compound M-3-3 (1.2 g).
Step 2:
compound M-3-3 (1.2 g) was dissolved in 2-methyltetrahydrofuran (20 mL) under nitrogen. MeMgBr (7.18 mL,3N diethyl ether solution) was added dropwise at-20deg.C, and the reaction mixture was heated to 25deg.C for further 2 hours. Quench with saturated aqueous ammonium chloride (5 mL), add water (100 mL), extract with ethyl acetate (50 mL x 3), dry over anhydrous sodium sulfate and filter, concentrate under reduced pressure to give compound M-3-4 (0.7 g). LC-MS (ESI) m/z=154.1 [ M+H ] ] +
Step 3:
compound M-3-4 (0.7 g) was dissolved in methylene chloride (10 mL) at 0deg.C, and diethylaminosulfur trifluoride (DAST, 3.68 g) was added dropwise. After 0.5 hour of reactionThe reaction mixture was warmed to room temperature and reacted for 12 hours. To the reaction solution was added water (50 mL) and saturated sodium bicarbonate solution to adjust ph=7-8, extracted with ethyl acetate (50 ml×3), dried over anhydrous sodium sulfate and filtered, concentrated under reduced pressure, and purified by silica gel column chromatography (dichloromethane: methanol=20:1) to give intermediate M-3 (0.40 g). LC-MS (ESI) m/z=156.1 [ M+H ]] +
Example 1: synthesis of Compound 001:
Figure BDA0004003727240000171
step 1:
under argon, cyclopropane (495 mg) was added to THF (25 mL). NaH (310 mg,60% mass fraction) was added under an ice-water bath, and after 20 minutes of reaction, intermediate M-1 (2.16 g) was added. The reaction mixture was warmed to room temperature, quenched by addition of saturated ammonium chloride solution (10 mL) after 45 minutes, extracted with ethyl acetate (3 x 50 mL), and the organic phase was dried over anhydrous sodium sulfate, concentrated under reduced pressure to give a crude product, which was purified by column chromatography (petroleum ether: ethyl acetate=10:1) to give compound 1-2 (2.03 g). LC-MS (ESI) m/z=298.9 [ M+H ]] +
Step 2:
tributyl (1-ethoxyethylene) tin (2.81 g), tetrakis (triphenylphosphine) palladium (783.1 mg) and compounds 1-2 (2.03 g) were dissolved in toluene (20 mL) under argon. After reaction at 80 ℃ for 6 hours, saturated KF aqueous solution (30 mL) was added and stirred for 1 hour, extracted with ethyl acetate (3 x 50 mL), and the organic phase was dried over anhydrous sodium sulfate, and separated and purified by column chromatography under reduced pressure (petroleum ether: ethyl acetate=10:1) to give compounds 1 to 3 (1.89 g). LC-MS (ESI) m/z=291.1 [ M+H ] ] +
Step 3:
compounds 1-3 (1.2 g) were dissolved in THF (12 mL) and H at room temperature 2 To O (2 mL) was added aqueous HCl (1.5N, 2751. Mu.L). After 1 hour of reaction, quench with saturated sodium bicarbonate solution (10 mL), extract with ethyl acetate (3 x 30 mL), dry the organic phase over anhydrous sodium sulfate, and isolate and purify the compound 1-4 (850 mg) by column chromatography under reduced pressure. LC-MS (ESI) m/z=263.1 [ M+H ]] +
Step 4:
compounds 1-4 (850 mg) were dissolved in THF (19 mL) under argon and MeMgBr (3M, 3.24 mL) was added dropwise at 0deg.C. The reaction mixture was slowly warmed to room temperature, after 2 hours of reaction, quenched by addition of saturated ammonium chloride solution (10 mL), extracted with ethyl acetate (3×30 mL), and the organic phase was dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified by column chromatography (petroleum ether: ethyl acetate=3:1) to give compounds 1 to 5 (750 mg). LC-MS (ESI) m/z=279.1 [ M+H ]] +
Step 5:
compounds 1-5 (100 mg), intermediate M-2 (76 mg), potassium acetate (176 mg), and (2-dicyclohexylphosphine-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) (2 '-methylamino-1, 1' -biphenyl-2-yl) palladium methanesulfonate (Brettphos Pd G4, 33 mg) and 2-dicyclohexylphosphine-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl (Brettphos, 39 mg) were dissolved in dioxane (3 mL) under argon. After reacting at 100℃for 1 hour, the reaction solution was concentrated under reduced pressure, and purified by using a liquid chromatography column (column: flash Spherical C; mobile phase: A: water; B: acetonitrile, B%:0% -30%,40 mL/min) to give compound 001 (95 mg).
LC-MS(ESI):m/z=435.2[M+H] +
1 H NMR(400MHz,DMSO-d6)δ10.66(s,1H),9.28(s,1H),9.21(d,J=0.9Hz,1H),8.17(s,1H),8.08(d,J=8.7Hz,1H),7.36(d,J=8.7Hz,1H),5.20(s,1H),4.62–4.54(m,1H),4.53–4.70(m,1H),3.04–2.92(m,1H),1.71(s,6H),1.46(d,J=7.1Hz,3H),1.39(d,J=6.5Hz,3H),0.86–0.82(m,4H).
Example 2: synthesis of compound 002:
Figure BDA0004003727240000181
step 1:
intermediate M-1 (900 mg) was dissolved in THF (20 mL) at room temperature, and sodium methyl mercaptide (250 mg) was added. After 4 hours of reaction, the reaction mixture was poured into water (60 mL), extracted with ethyl acetate (30 mL x 3), the organic phase was dried over anhydrous sodium sulfate and filtered, and concentrated under reduced pressure to give compound 2-2 (1.1 g).
Step 2:
compound 2-2 (1.1 g) was dispersed in toluene (20 mL) under nitrogen, and tributyl (1-ethoxyethylene) tin (4.86 g) and tetraphenylphosphine palladium (1.12 g) were added. After reaction at 80 ℃ for 4 hours, the reaction solution was directly concentrated and purified by silica gel column chromatography (petroleum ether: ethyl acetate=10:1) to give compound 2-3 (720 mg). LC-MS (ESI) m/z=281.0 [ M+H ]] +
Step 3:
compound 2-3 (720 mg) was dissolved in tetrahydrofuran (10 mL) and water (2 mL), and dioxane solution (1N, 0.3 mL) of hydrochloric acid was added. After 1h at rt, the reaction mixture was adjusted to pH 8-9 with saturated sodium carbonate solution, extracted with dichloromethane (30 ml x 3), dried over anhydrous sodium sulfate and filtered, concentrated under reduced pressure to give compound 2-4 (660 mg). LC-MS (ESI) m/z=253.0 [ M+H ]] +
Step 4:
compounds 2-4 (660 mg) were dissolved in THF (20 mL) under nitrogen. MeMgBr (1 Nin THF,5 mL) was added at 0deg.C, the reaction was continued at room temperature for 4 hours with slow warming, the pH of the reaction solution was adjusted to 7-8 with saturated ammonium chloride solution, extracted with dichloromethane (50 mL. Times.3), dried over anhydrous sodium sulfate and filtered, and concentrated under reduced pressure to give compound 2-5 (560 mg). LC-MS (ESI) m/z=269.0 [ M+H ] ] +
Step 5:
compounds 2-5 (170 mg) and intermediate M-3 (110 mg) were dissolved in 1, 4-dioxane (20 mL) under nitrogen, and Brettphos (40 mg), brettphos Pd G4 (69 mg) and potassium acetate (292 mg) were added sequentially. After 2 hours of reaction at 100 ℃, the reaction solution was concentrated and purified by high performance liquid chromatography (chromatographic column: gemini NX-C18; mobile phase: A is water; B is acetonitrile, B%:0% -40%,20 mL/min) to obtain compound 002 (15.4 mg).
LC-MS(ESI):m/z=388.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ10.68(s,1H),9.35(s,1H),9.14(s,1H),8.49(s,1H),8.47(d,J=5.9Hz,1H),7.36(d,J=5.9Hz,1H),5.30(s,1H),2.66(s,3H),1.79(s,3H),1.74(s,3H),1.71(s,6H).
Example 3: synthesis of compound 003:
Figure BDA0004003727240000191
step 1:
intermediate M-1 (2 g) was dissolved in methanol (20 mL), potassium carbonate (2.96 g) was added, and the reaction was allowed to proceed at 25℃for 5 hours, followed by LCMS monitoring the reaction. The reaction was concentrated, the residue was taken up in water (50 mL), extracted with dichloromethane (50 mL x 3) and the organic phase dried over anhydrous sodium sulphate and concentrated to give crude compound 3-2 (1.8 g). LC-MS (ESI) m/z=272.9 [ M+H ]] +
Step 2:
compound 3-2 (1.8 g) was dissolved in toluene (20 mL), tributyl (1-ethoxyethylene) tin (2.86 g) and tetrakis (triphenylphosphine) palladium (0.76 g), N were added 2 The reaction was allowed to react at 80℃for 12 hours under protection, and LCMS was monitored for completion of the reaction. The reaction mixture was cooled to 25℃and saturated aqueous potassium fluoride (20 mL) was added thereto, stirred for 1 hour, extracted with ethyl acetate (50 mL. Times.3), and the organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give a crude compound 3-3 (1.6 g). LC-MS (ESI) m/z=265.1 [ M+H ] ] +
Step 3:
compound 3-3 (1.6 g) was dissolved in tetrahydrofuran (15 mL) and water (3 mL), and aqueous hydrochloric acid (0.4 mL, 2M) was added dropwise and reacted at 25℃for 1 hour, followed by LCMS monitoring the reaction. Saturated aqueous sodium bicarbonate (20 mL) was added to the reaction, extracted with ethyl acetate (50 mL x 3), and the organic phase dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated to give crude compound 3-4 (1.2 g). LC-MS (ESI) m/z=237.0 [ M+H ]] +
Step 4:
compound 3-4 (1.2 g) was dissolved in tetrahydrofuran (15 mL), cooled to-20deg.C, and N was replaced 2 After three times of protection, meMgBr (5.1 mL, 3M) was added dropwise and the reaction was resumed at 25℃for 2 hours, and LCMS monitored to be complete. The reaction mixture was quenched by dropping saturated aqueous ammonium chloride (5 mL), then added with water (50 mL), extracted with ethyl acetate (50 mL. 3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give the compound3-5 (0.7 g). LC-MS (ESI) m/z=253.1 [ M+H ]] +
Step 5:
compound 4 (0.10 g) and intermediate M-3 (61.41 mg) were dissolved in dioxane (10 mL), followed by addition of potassium acetate (116.5 mg), 2-dicyclohexylphosphine-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl (21.2 mg) and (2-dicyclohexylphosphine-3, 6-dimethoxy-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) (2 '-methylamino-1, 1' -biphenyl-2-yl) methanesulfonic acid palladium (36.4 mg), N 2 The reaction was allowed to react at 100℃for 2 hours under protection, and LCMS was monitored to complete the reaction. The reaction was cooled to 25 ℃, water (50 mL) was added, extracted with ethyl acetate (50 mL x 3), the organic phase dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated and purified by silica gel column chromatography to give compound 003 (16.7 mg).
LC-MS(ESI):m/z=372.1[M+H] +
1 H NMR(400MHz,Methanol-d 4 )δ9.35(s,1H),9.04(s,1H),8.37(d,J=6.0Hz,1H),8.16(s,1H),7.34(d,J=6.0Hz,1H),4.12(s,3H),1.84(s,3H),1.81-1.77(m,9H).
Example 4: synthesis of compound 004:
Figure BDA0004003727240000201
step 1:
intermediate M-1 (500 mg) was dissolved in dry tetrahydrofuran (10 mL), to which sodium methyl mercaptide (151.3 mg) was added and reacted overnight at room temperature. After completion of the reaction, LCMS monitored, water (30 mL) was added to the reaction solution and ethyl acetate (30 mL) was extracted twice. The organic phase was washed once with water (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated to give 520mg of intermediate 4-2.LC-MS (ESI) m/z=288.91 [ M+H ]] +
Step 2:
intermediate 4-2 (500 mg) was dissolved in toluene (10 mL), tributyl (1-ethoxyvinyl) tin (748 mg) and tetraphenylpalladium phosphate (200 mg) were added in this order, and the mixture was reacted overnight at 80℃under nitrogen. LCMS monitored reaction was complete. The reaction solution was cooled to room temperature, and a potassium fluoride solution (20 mL) was added thereto, followed by stirring at room temperature for 1h.Ethyl acetate (20 mL) was extracted twice, the combined organic phases were washed with water (40 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give intermediate 4-3, which was taken to the next step in theoretical amount. LC-MS (ESI) m/z=281.04 [ M+H ] ] +
Step 3:
the crude intermediate 4-3 (500 mg) was dispersed in tetrahydrofuran (5 mL) and water (1 mL), to which diluted hydrochloric acid (0.5 mL, 1.5M) was added and reacted at 25℃for 4 hours. LCMS monitored reaction was complete. Ethyl acetate (20 mL) and aqueous sodium bicarbonate (20 mL) were added to the reaction, the mixture was separated, the aqueous phase was extracted with ethyl acetate (20 mL), the combined organic phases were washed with water (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give crude product, which was purified by column chromatography (petroleum ether: ethyl acetate=10:1-3:1) to give 230mg of intermediate 4-4.LC-MS (ESI) m/z=253.01 [ M+H ]] +
Step 4:
intermediate 4-4 (150 mg) was dissolved in 1, 4-dioxane (5 mL), intermediate M-2 (114 mg), brettphos (64 mg), brettphos Pd G4 (109 mg) and potassium acetate (291 mg) were added in this order, and the mixture was reacted at 100℃for 4 hours under nitrogen. LCMS monitored reaction was complete. The reaction solution was cooled to room temperature, filtered through celite, and the filtrate was concentrated to give a crude product which was purified by column chromatography (petroleum ether: ethyl acetate=3:1-1:1) to give 150mg of intermediate 4-5.LC-MS (ESI) m/z=409.13 [ M+H ]] +
Step 5:
intermediate 4-5 (150 mg) was dissolved in isopropanol (5 mL), to which was added sodium cyanoborohydride (115 mg) and ammonium acetate (566 mg), and reacted under closed conditions at 90℃for 2 hours. LCMS monitored reaction was complete. The reaction solution was cooled to room temperature, water (20 mL) and methylene chloride (20 mL) were added, the solution was separated, the aqueous phase was extracted with methylene chloride (20 mL), the combined organic phases were dried over anhydrous sodium sulfate, filtered, and concentrated to give a crude product, which was subjected to high performance liquid chromatography (column: YMC 18; mobile phase: A was water (containing 0.1% aqueous ammonia); B%:45% -65%, B was acetonitrile, 40 mL/min) to give compound 004 (26 mg). LC-MS (ESI) m/z=410.10 [ M+H ] ] +
1 H NMR(400MHz,DMSO-d 6 )δ10.83(d,J=8.0Hz,1H),9.33(s,1H),8.89(d,J=4.7Hz,1H),8.58(d,J=8.9Hz,1H),8.11(d,J=8.7Hz,1H),7.32(dd,J=8.7,2.8Hz,1H),4.65(dd,J=14.3,6.9Hz,1H),4.58–4.49(m,1H),3.09(q,J=8.0Hz,1H),2.66(s,3H),1.54–1.38(m,9H).
Example 5: synthesis of Compound 005:
Figure BDA0004003727240000211
step 1:
dissolving intermediate 3-5 (110 mg) in ultra-dry dichloromethane (5 mL), protecting with nitrogen, cooling to 0deg.C, and dropwise adding boron trifluoride diethyl etherate (180 mg) and TMSN 3 (150 mg), slowly warmed to room temperature, and reacted for 3 hours, followed by TLC monitoring the completion of the reaction. The reaction was poured into cold saturated sodium bicarbonate solution (30 mL), extracted with ethyl acetate (10 mL x 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated to give intermediate 5-2 (115 mg).
Step 2:
the starting material 5-2 (200 mg) was dispersed in dichloromethane (10 mL), 1, 4-dioxane solution of hydrogen chloride (0.5 mL,4 m) was added, reacted overnight at 25 ℃, and TLC detection was complete (petroleum ether: ethyl acetate=3:1). The reaction was adjusted to neutral pH with saturated sodium carbonate solution, extracted with dichloromethane (15 ml x 3), the organic phase dried over anhydrous sodium sulfate, filtered and the filtrate concentrated to give intermediate 5-3 (180 mg).
Step 3:
intermediate 5-3 (180 mg) was dissolved in acetonitrile (10 mL), and triethylamine (210 mg) and phosphorus oxybromide (40 mg) were added to react at 80℃for 6 hours. LCMS detected reaction completion. The reaction was cooled to room temperature, poured into an ice saturated sodium bicarbonate solution (30 mL), extracted with ethyl acetate (15 mL x 3), the organic phase dried over anhydrous sodium sulfate, filtered and the filtrate concentrated to give 170mg of intermediate 5-4.LC-MS (ESI) m/z=325.97, 327.97[ M+H ] ] +
Step 4:
intermediate 5-4 (150 mg) and dimethyl sulfenamide (43 mg) were dissolved in 1, 4-dioxane (10 mL), and Pd was added sequentially 2 (dba) 3 (42 mg), xantphos (27 mg) and cesium carbonate (300 mg)mg), under nitrogen protection at 70℃for 12h. LCMS detected reaction completion. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated and purified by column chromatography (petroleum ether: ethyl acetate=3:1) to give 110mg of crude intermediate 5-5.LC-MS (ESI) m/z=339.07 [ M+H ]] +
Step 5:
intermediate 5-5 (100 mg) and intermediate M-3 (55 mg) were dissolved in 1, 4-dioxane (10 mL), and Brettphos (38 mg), brettphos Pd G4 (65 mg) and potassium acetate (174 mg) were added in this order and reacted at 100℃under nitrogen for 4 hours. LCMS detected reaction completion. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated and purified by column chromatography (petroleum ether: ethyl acetate=1:1) to give 130mg of intermediate 5-6.LC-MS (ESI) m/z=458.20 [ M+H ]] +
Step 6:
intermediate 5-6 (130 mg) was dissolved in dry ethyl acetate (10 mL), and platinum dioxide (20 mg) was added thereto and reacted at 40℃for 2 hours under a hydrogen atmosphere. LCMS detected reaction completion. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated to give a crude product, which was subjected to high performance liquid chromatography (column: YMC 18; mobile phase: A was water (0.1% formic acid), B%:30% -70%, B was acetonitrile, 40 mL/min) to give compound 005 (30 mg).
LC-MS(ESI):m/z=432.20[M+H] +
1 H NMR(400MHz,DMSO-d 6 ) δ10.57 (s, 1H), 9.44 (s, 1H), 9.04 (s, 1H), 8.44 (d, j=5.8 hz, 1H), 8.25 (s, 1H), 8.11 (s, 1H), 7.33 (d, j=5.9 hz, 1H), 3.53 (s, 6H), 1.78 (s, 3H), 1.74-1.68 (m, 9H). Example 6: synthesis of compound 006:
Figure BDA0004003727240000221
/>
step 1:
intermediate 5-3 (150 mg) was dissolved in tetrahydrofuran (5 mL), sodium methyl mercaptide (40 mg) was added, the reaction was carried out at 25℃for 12h, and the completion of the reaction was detected by TLC. The reaction solution was poured into water, extracted with ethyl acetate (15 ml×3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give 110mg of crude intermediate 6-2.
Step 2:
intermediate 6-2 (100 mg) and intermediate M-3 (58 mg) were dissolved in 1, 4-dioxane (10 mL), and Brettphos (40 mg), brettphos Pd G4 (69 mg) and potassium acetate (190 mg) were added in this order and reacted at 100℃under nitrogen for 4 hours. TLC detection was complete. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated and purified by column chromatography (petroleum ether: ethyl acetate=1:1) to give 110mg of intermediate 6-3.
Step 3:
intermediate 6-3 (100 mg) was dissolved in 80% ethanol aqueous solution (10 mL), and ammonium chloride (25 mg) and zinc powder (32 mg) were added to react at 80℃for 15min, and the completion of the reaction was detected by LCMS. The reaction was cooled to room temperature, filtered, the filtrate was collected and poured into water, extracted with ethyl acetate (15 mL x 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate concentrated to give crude product, which was purified by high performance liquid chromatography (column: YMC 18; mobile phase: a water (0.1% aqueous ammonia); B%:45% -65%, B acetonitrile, 40 mL/min) to give compound 006 (18 mg).
LC-MS(ESI):m/z=387.20[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ10.70(s,1H),9.35(s,1H),9.26(s,1H),8.52(s,1H),8.50–8.44(m,2H),7.42–7.36(m,1H),2.66(s,3H),1.79(s,3H),1.73(s,3H),1.68(s,6H).
Example 7: synthesis of Compounds 007, 007-P1, 007-P2:
Figure BDA0004003727240000231
step 1:
intermediate M-1 (1.8 g) was dissolved in a mixed solution of deuterated methanol (15 mL) and tetrahydrofuran (15 mL), potassium carbonate (2.7 g) was added, and the reaction was stirred at 25℃overnight, and LCMS monitored for completion. The reaction was concentrated, water (50 mL) was added to the residue, extracted with dichloromethane (15 mL x 3), the combined organic phases were dried over anhydrous sodium sulfate, filtered and the filtrate concentrated to give 1.4g of intermediate 7-2.LC-MS (ESI) m/z=275.9 [ M+H ]] +
Step 2:
intermediate 7-2 (1.5 g) was dissolved in toluene (30 mL), tributyl (1-ethoxyvinyl) tin (2.4 g) and tetra-triphenylphosphine palladium (626 mg) were added in this order, and reacted at 80℃for 12h under nitrogen. LCMS monitored the reaction was complete. The reaction solution was cooled to 25℃and a saturated aqueous KF solution (50 mL) was added thereto and stirred for 1h. Extraction with ethyl acetate (20 ml x 3) and drying of the organic phase over anhydrous sodium sulphate, filtration and concentration of the filtrate gives 1.3g of crude intermediate 7-3. The crude product is put into the next step according to the theoretical amount. LC-MS (ESI) m/z=268.07 [ M+H ]] +
Step 3:
the crude intermediate 7-3 (1.5 g) was dispersed in a mixed solution of tetrahydrofuran (15 mL) and water (1 mL), diluted hydrochloric acid (0.4 mL, 1.5M) was added, and the mixture was stirred at 25℃for 2 hours. LCMS monitored the reaction was complete. Ethyl acetate (50 mL) and aqueous sodium hydrogencarbonate (30 mL) were added sequentially to the reaction solution, the solution was separated, the aqueous phase was extracted with ethyl acetate (20 mL), the combined organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated and purified by crude column chromatography (petroleum ether: ethyl acetate=3:1) to give 1.2g of intermediate 7-4.LC-MS (ESI) m/z=240.04 [ M+H ] ] +
Step 4:
intermediate 7-4 (270 mg) was dissolved in tetrahydrofuran (10 mL), cooled to 0deg.C under nitrogen protection, and a tetrahydrofuran solution of methylmagnesium bromide (1.1 mL, 3M) was added dropwise, the reaction was stirred at 0deg.C for 2h, and LCMS monitored the reaction. To the reaction mixture were added ethyl acetate (20 mL) and a saturated ammonium chloride (20 mL), and the mixture was separated, and an aqueous phase was extracted with ethyl acetate (20 mL). The combined organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give 230mg of intermediate 7-5.LC-MS (ESI) m/z=256.07 [ M+H ]] +
Step 5:
intermediate 7-5 (220 mg) was dissolved in 1, 4-dioxane (5 mL), intermediate M-2 (160 mg), brettphos (46 mg), brettphos Pd G4 (79 mg) and potassium acetate (505 mg) were added in this order, and reacted at 80℃for 4 hours under nitrogen. LCMS monitored the reaction was complete. The reaction solution was cooled to 25 ℃, filtered through celite, and the filtrate was concentrated to give 007 crude product, which was prepared by supercritical liquid chromatography (SFC; column DAICEL CHIRALPAK IC (250 mm. Times.30 mm,10 um)) and mobile phase using ethanol (0.1% aqueous ammonia) as eluent as a mixture of polarity (30% -30%) to give 007-P1 (22.9 mg) and 007-P2 (46.3 mg).
007-P1
Rt=1.658min
LC-MS(ESI):m/z=412.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ10.66(s,1H),9.31(s,1H),9.27(s,1H),8.15(s,1H),8.12-8.06(m,1H),7.41-7.33(m,1H),5.21(s,1H),4.64-4.54(m,1H),3.04-2.94(m,1H),1.70(s,6H),1.48-1.44(m,3H),1.42-1.38(m,3H).
007-P2
Rt=2.961min
LC-MS(ESI):m/z=412.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ10.66(s,1H),9.31(s,1H),9.28(s,1H),8.15(s,1H),8.12-8.06(m,1H),7.41-7.36(m,1H),5.21(s,1H),4.64-4.54(m,1H),3.04-2.94(m,1H),1.71(s,6H),1.48-1.44(m,3H),1.41-1.38(m,3H).
Example 8: synthesis of Compounds 008, 008-P1, 008-P2:
Figure BDA0004003727240000241
Intermediate 3-4 (150 mg) was dissolved in 1, 4-dioxane (8 mL), intermediate M-2 (114 mg), brettphos (64 mg), brettphos Pd G4 (109 mg) and potassium acetate (291 mg) were added in this order, and the mixture was reacted at 100℃for 4 hours under nitrogen. LCMS monitored the reaction was complete. The reaction solution was cooled to 25 ℃, filtered through celite, and the filtrate was concentrated to give 008 crude product, which was prepared by supercritical liquid chromatography (SFC; column DAICEL CHIRALPAK IC (250 mm. Times.30 mm,10 um), mobile phase: a mixture of ethanol (containing 0.1% ammonia water) with polarity (50% -50%) as eluent to give 008-P1 (11.1 mg) and 008-P2 (28.8 mg).
008-P1
Rt=1.657min
LC-MS(ESI):m/z=409.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ10.56(s,1H),9.56(s,1H),9.55(s,1H),8.16(s,1H),8.16-8.13(m,1H),7.38-7.33(m,1H),5.21(s,1H),4.60-4.56(m,1H),4.12(s,3H),3.03-2.98(m,1H),1.74(s,6H),1.70-1.58(m,3H),1.47-1.37(m,3H).
008-P2
Rt=1.661min
LC-MS(ESI):m/z=409.1[M+H] +
1 H NMR(400MHz,MeOD)δ9.27(s,1H),9.36(s,1H),8.17(s,1H),8.17-8.14(m,1H),7.35-7.31(m,1H),4.68-4.60(m,1H),4.15(s,3H),3.08-3.04(m,1H),1.86-1.80(m,6H),1.80-1.92(m,3H),1.51-1.28(m,3H).
Example 9: synthesis of Compound 009:
Figure BDA0004003727240000251
intermediate 5-4 (180 mg) was dissolved in 1, 4-dioxane (10 mL), intermediate M-2 (102 mg), brettphos (86 mg), brettphos Pd G4 (98 mg) and potassium acetate (261 mg) were added in this order, and reacted at 100℃under nitrogen for 4 hours. LCMS monitored the reaction was complete. The reaction solution was cooled to room temperature, filtered through celite, and the filtrate was concentrated to give a crude product, which was subjected to high performance liquid chromatography (column: YMC 18; A was water (containing 0.1% ammonia water); B%:45% -65%, B was acetonitrile, 40 mL/min) to give Compound 009 (50 mg).
LC-MS(ESI):m/z=469.20[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ10.58(s,1H),9.40(s,1H),9.33(s,1H),8.16(s,1H),8.06(d,J=8.7Hz,1H),7.34(d,J=8.7Hz,1H),4.63–4.57(m,1H),3.52(s,6H),2.99(dt,J=13.5,7.2Hz,1H),1.96(s,2H),1.64(d,J=2.4Hz,6H),1.44(d,J=7.0Hz,3H),1.38(d,J=6.5Hz,3H).
Example 10: synthesis of Compound 010:
Figure BDA0004003727240000252
Intermediate 5-3 (260 mg) was dissolved in tetrahydrofuran (10 mL), and methylthio was addedSodium alkoxide (56 mg) was reacted overnight at 25 ℃. LCMS monitored the reaction was complete. To the reaction solution were added ethyl acetate (20 mL) and water (20 mL), the mixture was separated, the aqueous phase was extracted with ethyl acetate (20 mL), and the combined organic phases were washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give 220mg of intermediate 10-2.LC-MS (ESI) m/z=294.05 [ M+H ]] +
Intermediate 10-2 (180 mg) was dissolved in 1, 4-dioxane (5 mL), and intermediate M-2 (118 mg), brettphos (99 mg), brettphos Pd G4 (113 mg) and potassium acetate (301 mg) were added in this order and reacted at 100℃under nitrogen for 6 hours. LCMS monitored the reaction was complete. The reaction solution was cooled to room temperature, filtered through celite, and the filtrate was concentrated to give a crude product which was purified by column chromatography (petroleum ether: ethyl acetate=10:1-3:1) to give 240mg of intermediate 10-3.LC-MS (ESI) m/z=450.16 [ M+H ]] +
Intermediate 10-3 (180 mg) was dispersed in a mixed solution of ethanol (10 mL) and water (2 mL), followed by addition of zinc powder (70 mg) and ammonium chloride (57 mg), and reacted at 80℃for 1 hour. LCMS monitored the reaction was complete. The reaction solution was cooled to room temperature, ethyl acetate (20 mL) and water (30 mL) were added, the solution was separated, the aqueous phase was extracted with ethyl acetate (20 ml×3), the combined organic phases were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give a crude product which was subjected to high performance liquid chromatography (column: YMC 18; mobile phase: a: water (containing 0.1% ammonia); B%:45% -65%, B was acetonitrile, 40 mL/min) to give compound 010 (16 mg).
LC-MS(ESI):m/z=424.20[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ10.75(s,1H),9.41(s,1H),9.33(s,1H),8.48(s,1H),8.10(d,J=8.7Hz,1H),7.42(d,J=8.7Hz,1H),4.62(dd,J=6.6,5.3Hz,1H),3.05–2.96(m,1H),2.66(s,3H),1.71(d,J=3.9Hz,6H),1.44(d,J=7.1Hz,3H),1.38(d,J=6.5Hz,3H).
Example 11: synthesis of Compound 011:
Figure BDA0004003727240000261
step 1:
the starting material 11-1 (10 g) was dissolved in acetic acid (100 mL), bromine (10.37 g) was added dropwise and reacted overnight at 25 ℃. LCMS monitored the reaction was complete. To the reaction solution were added ethyl acetate (200 mL) and water (200 mL), the solution was separated, and the aqueous phase was extracted with ethyl acetate (100 ml×2). The organic phases were combined, washed with saturated brine (300 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give 15g of intermediate 11-2.LC-MS (ESI) m/z= 232.94, 234.94[ M+H ]] +
Step 2:
intermediate 11-2 (15 g) was dissolved in methanol (150 mL), concentrated sulfuric acid (17 g) was added to the solution in an ice bath, and the reaction was carried out at 60℃for 10 hours. LCMS monitored the reaction was complete. The reaction mixture was concentrated, aqueous sodium bicarbonate was added to the residue to adjust the pH to 7, extracted with ethyl acetate (70 mL x 3), the organic phases were combined, washed with saturated brine (200 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated to give 10g of intermediate 11-3.LC-MS (ESI) m/z= 246.95, 248.95[ M+H ]] +
Step 3:
intermediate 11-3 (10 g) was dispersed in aqueous ammonia (60 mL) and reacted at 50℃for 12 hours under closed conditions. LCMS monitored the reaction was complete. The reaction mixture was cooled to room temperature, and ethyl acetate (100 mL) was added thereto to separate the solution. The aqueous phase was extracted with ethyl acetate (20 x 5 ml) and the combined organic phases were dried over anhydrous sodium sulfate, filtered and the filtrate concentrated to give 10g of intermediate 11-4. m/z= 231.95, 233.95[ m+h ] ] +
Step 4:
intermediate 11-4 (10 g) was dissolved in N, N-dimethylformamide (150 mL), and 1, 2-dibromoethane (15 g) and cesium carbonate (26 g) were added in this order to react at 70℃for 10 hours. LCMS monitored the reaction was complete. The reaction mixture was cooled to room temperature, and ethyl acetate (200 mL) and water (150 mL) were added thereto to separate the solutions. The aqueous phase was extracted with ethyl acetate (70 x 3 ml), the combined organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give crude product which was purified by column chromatography (dichloromethane: methanol=10:1) to give 5g of intermediate 11-5.LC-MS (ESI) m/z= 257.97, 259.97[ M+H ]] +
Step 5:
intermediate 11-5 (2 g) was dissolved in dry tetrahydrofuran (50 mL), cooled to 0deg.C, and added with borane dimethyl sulfide under nitrogen blanketThe reaction was continued for 16h at 65℃after stirring the tetrahydrofuran solution (13 mL, 3M) slowly warmed to 25 ℃. LCMS monitored the reaction was complete. To the reaction mixture was added methanol (20 mL), the mixture was quenched, concentrated, diluted hydrochloric acid (20 mL) was added to the residue to precipitate a solid, and the solid was filtered and the cake was dried to obtain 2g of intermediate 11-6.LC-MS (ESI) m/z= 243.99, 245.99[ M+H ]] +
Step 6:
intermediate 11-6 (1.3 g) and methyl 2, 2-dimethoxyacetate (857 mg) were added to the tube, and reacted at 100℃for 1 hour. LCMS monitored the reaction was complete. Toluene (5 mL) was added to the reaction solution, and the mixture was concentrated to obtain intermediate 11-7, and the crude product was directly fed to the next step in accordance with the theoretical amount. LC-MS (ESI) m/z=346.02 [ M+H ] ] +
Step 7:
intermediate 11-7 (1.85 g) was dissolved in concentrated sulfuric acid (7 mL) and reacted overnight at 25 ℃. LCMS monitored the reaction was complete. The reaction solution was added to ice water (20 mL), and aqueous ammonia was added dropwise thereto to adjust the pH to 5, to precipitate a solid, which was filtered and the cake was dried to obtain 1.5g of intermediate 11-8.LC-MS (ESI) m/z= 281.97, 283.97[ M+H ]] +
Step 8:
intermediate 11-8 (1 g) was dissolved in dry dichloromethane (15 mL), triethylamine (717 mg), ethylsulfonyl chloride (547 mg) and 4-dimethylaminopyridine (22 mg) were added in this order, and reacted overnight at 25 ℃. LCMS monitored the reaction was complete. The reaction mixture was concentrated and the residue was purified by column chromatography (petroleum ether: ethyl acetate=10:1-1:1) to give 80mg of intermediate 11-9.LC-MS (ESI) m/z= 373.96, 375.96[ M+H ]] +
Step 9:
intermediate 11-9 (80 mg) was dissolved in toluene (1 mL), tributyl (1-ethoxyethylene) tin (93 mg) and tetrakis triphenylphosphine palladium (25 mg) were added in this order, and reacted at 80℃under nitrogen for 16h. LCMS monitored the reaction was complete. The reaction solution was cooled to room temperature, and an aqueous potassium fluoride solution was then added thereto, followed by stirring at room temperature for 1 hour. Ethyl acetate (20 mL) and water (20 mL) were added and the mixture was partitioned. The aqueous phase was extracted with ethyl acetate (20 mL). The combined organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give intermediate 11-10. The crude product is directly put into the next step according to the theoretical amount. LC-MS (ESI) m/z=366.09 [ M ] +H] +
Step 10:
the crude intermediate 11-10 (270 mg) was dispersed in a mixed solution of tetrahydrofuran (5 mL) and water (5 mL), diluted hydrochloric acid (0.1 mL, 3M) was added, and the mixture was reacted at 25℃for 4 hours. LCMS monitored the reaction was complete. To the reaction solution was added saturated aqueous sodium bicarbonate (20 mL), extracted with ethyl acetate (10 mL x 2), the combined organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated to give crude product which was purified by pre-TLC (petroleum ether: ethyl acetate=2:1) to give 100mg of intermediate 11-11.LC-MS (ESI) m/z=338.06 [ M+H ]] +
Step 11:
intermediate 11-11 (100 mg) was dissolved in 1, 4-dioxane (3 mL), and intermediate M-2 (46 mg), brettphos (32 mg), brettphos Pd G4 (55 mg) and potassium acetate (145 mg) were added in sequence and reacted at 100℃for 8 hours under nitrogen. LCMS monitored the reaction was complete. The reaction mixture was concentrated to give crude product which was purified by pre-TLC (petroleum ether: ethyl acetate=2:1) to give 80mg of intermediate 11-12.LC-MS (ESI) m/z=420.15 [ M+H ]] +
Step 12:
intermediate 11-12 (50 mg) was dissolved in isopropanol (2 mL), and sodium cyanoborohydride (37 mg) and ammonium acetate (184 mg) were added in this order, followed by reaction at 90℃for 2 hours under closed conditions. LCMS monitored the reaction was complete. The reaction mixture was cooled to room temperature, and water (20 mL) and methylene chloride (20 mL) were added in this order to separate the solutions. The aqueous phase was extracted with dichloromethane (20 mL), the combined organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated to give crude product, which was subjected to supercritical liquid chromatography (SFC; column DAICEL CHIRALPAK IC (150 mm. Times.30 mm. Times.50 um), mobile phase [ water (0.1% formic acid) -ACN ]; B%:15% -35%,11 min) to give compounds 011-P1 (0.8 mg) and 011-P2 (1.2 mg).
011-P1
Rt=1.213min
LC-MS(ESI):m/z=412.1[M+H] +
1 H NMR(400MHz,MeOD)δ9.25(s,1H),8.76(s,1H),8.55(br,1H),8.14-8.10(m,1H),7.49(s,1H),7.22-7.18(m,1H),5.10-5.05(m,1H),4.56-4.52(m,1H),4.46-4.42(m,3H),3.10-3.02(m,1H),1.72-1.70(s,3H),1.62-1.58(m,3H),1.57-1.54(m,3H).
011-P2
Rt=1.234min
LC-MS(ESI):m/z=412.1[M+H] +
1 H NMR(400MHz,MeOD)δ9.25(s,1H),8.81(s,1H),8.56(br,1H),8.14-8.11(m,1H),7.49(s,1H),7.20-7.17(m,1H),5.10-5.02(m,1H),4.56-4.22(m,5H),3.12-3.02(m,1H),1.68-1.63(s,3H),1.61-1.58(m,3H),1.57-1.54(m,3H).
Biological Activity and related Property test cases
The compounds in the following test examples were all prepared according to the methods of the above examples of the present invention.
Test example 1: HPK1 kinase Activity assay
Experimental materials:
HPK1 (MAP 4K 1) 35948 was purchased from Signalchem, #M23-11G
MBP 35951 was purchased from Signalchem, #M42-51N
ADP-Glo was purchased from Promega, # V9102
DMSO was purchased from Sigma
384-well assay plate was purchased from Perkin Elmer #6007290
384-well assay plate was purchased from LABCYTE
MgCl 2 、MnCl 2 DTT, tween-20, HEPES, BSA purchased from Sigma
Experimental instrument:
nano-scale acoustic pipetting system:
Figure BDA0004003727240000281
650LIQUID HANDLERS(LABCYTE,USA)
multi-label detection analyzer: envision Multilabel Reader (PerkinElmer, USA)
The experimental method comprises the following steps:
the inhibition activity of the synthesized compound on HPK1 kinase was detected by using a luminescence kinase assay (ADP-GloTM) developed by Promega corporation. The specific method comprises the following steps: for compounds
Figure BDA0004003727240000282
Gradient is carried outDiluted and transferred into reaction plates at 50 nL/well (384 Kong Baiban, perkin Elmer # 6007290), starting compound concentration 100nm, 3-fold gradient dilution, 10 concentration points; HPK1 was buffered with kinase reaction buffer (50 mM HEPES (pH 7.5), 0.01% Tween-20,5mM MgCl) 2 The reaction wells were diluted to the appropriate concentration with 0.01% BSA and 0.05mM DTT, 3. Mu.l enzyme (final concentration 50 nM) or kinase reaction buffer was added to each well, the reaction plate was placed in a centrifuge, centrifuged at 1000 rpm for 30 seconds and incubated on ice for 30 minutes. Mu.l/well of 2.5 XATP (62.5. Mu.M)/MBP substrate mixture (250. Mu.g/mL) was added, centrifuged at 1000 rpm for 30 seconds and incubated at room temperature for 60 minutes. Add 5. Mu.L/well ADP-Glo and mix well and react for 40 minutes at room temperature. ADP-Glo detection substrate was added, 10. Mu.L/well, and incubated for 30 minutes at room temperature. The chemiluminescent signal was read in an enzyme-labeled instrument (Envision, perkin Elmer). Test compound inhibition (n=2) was calculated as: inhibition% = (maximum signal value-signal value for each well)/(maximum signal value-minimum signal value) ×100%. The maximum signal value is the reading value of the strongest enzyme reaction activity only containing DMSO; the minimum signal value is the read of the wells without enzyme. Data were imported into MS Excel and curve fitted using XLFit Excel add-in version 5.4.0.8: y=bottom+ (Top-Bottom)/(1+ (IC) 50 X ≡HillSlope), IC is calculated from the fitted curve 50
The test results are shown in Table 1.
TABLE 1 HPK1 enzymatic inhibitory Activity of the Compounds of the invention
Figure BDA0004003727240000283
Figure BDA0004003727240000291
Test example 2: t cell activation assay
Experimental materials:
jurkat T cells were purchased from ATCC
RPMI1640 is purchased from Gibco (ThermoFisher, USA)
PBS was purchased from Gibco (ThermoFisher, USA)
anti-CD 3 monoclonal antibody (OKT 3) was purchased from BD Biosciences #566685
anti-CD 28 monoclonal antibody (CD 28.2) was purchased from BD Biosciences #555725
Human IL-2ELISA detection kit was purchased from BD Biosciences #555190
96 well cell culture plates were purchased from Corning
Experimental instrument:
CO 2 cell incubator: thermoFisher (USA)
Multi-label detection analyzer: envision Multilabel Reader (PerkinElmer, USA)
Cell counter: vi-CELL (Beckman, USA)
The experimental method comprises the following steps:
the 96-well cell culture plate is pre-treated with anti-CD 3 antibody, the anti-CD 3 antibody is diluted to 2 mug/mL by PBS, 100 mug/well is added, the cell culture plate is incubated for 4 hours at 37 ℃, and then washed for 1-2 times by PBS, and the cell culture plate is spin-dried for standby; jurkat T cells were collected, counted using a cytometer, and cell density was adjusted to 1X10 per well 5 Cell numbers were seeded into a new cell culture plate. Dissolving the compound with DMSO and performing gradient dilution, adding corresponding holes of the culture plate inoculated with cells, controlling the final concentration of the DMSO below 0.1%, and performing gradient dilution with initial concentration of the compound of 10 mu M and 3 times, wherein the concentration points are 8; the cell culture plates were placed in a 37℃incubator and pre-incubated for 1 hour. Transfer of preincubated Jurkat T cells to CD3 antibody coated cell culture plates, 100. Mu.L/well, cell count 1X10 5 The method comprises the steps of carrying out a first treatment on the surface of the Adding an anti-CD 28 antibody to obtain a final concentration of 1 mug/mL; the cell culture plates were placed in a carbon dioxide cell incubator for 48 hours. Taking cell culture supernatant, diluting with a proper amount, and detecting the content of human IL-2 by adopting an ELISA method. Quantitative conversion was performed based on the amount of standard. Fold count of IL-2 production: fold = IL-2 production/minimum IL-2 production, minimum IL-2 production i.e. IL-2 production in DMSO blank wells. Maximum effect refers to the highest fold production of IL-2 under drug treatment.
The test results are shown in Table 2.
TABLE 2T cell activation assay results for the compounds of the invention
Figure BDA0004003727240000301
Test example 3: pharmacokinetic studies in mice
6 BALB/c mice (purchased from Peking Vitre Liwa laboratory animal technologies Co., ltd.) were randomly divided into 2 groups (3 each). The test compounds were formulated into dosing solutions which were administered to animals in IV and PO at doses of 2 and 10mg/kg. After administration, blood samples were taken at a series of preset time points (time points see table below), each time point being sampled from a group of animals. The blood sample is converted to a plasma sample using a general procedure and the concentration of the test compound in the sample is analyzed by LC-MS/MS. The data obtained are shown in Table 3.
Figure BDA0004003727240000302
Figure BDA0004003727240000311
TABLE 3 PK parameters of Compound 001 in BALB/c mice
Modes of administration and PK parameters Compound 001
IV dose(mpk) 2
IV Cl(mL/min/kg) 12.3
IV AUC(h*ng/mL) 2792
IV Vss(L/kg) 2.8
PO dose(mpk) 10
PO T 1/2 (h) 2.4
PO C max (ng/mL) 2251
PO AUC(h*ng/mL) 14577
F(%) 101

Claims (13)

1. A compound of formula (I) or a pharmaceutically acceptable salt thereof:
Figure FDA0004003727230000011
wherein, the liquid crystal display device comprises a liquid crystal display device,
X 1 selected from N or CR 1 ,R 1 Selected from H, halogen, CN, SH, NH 2 、C 1 -C 10 Alkyl, C 1 -C 10 Alkoxy, C 3 -C 10 Cycloalkyl, C 3 -C 10 Cycloalkyloxy, 4-7 membered heterocyclyl, 4-7 membered heterocyclyloxy or
Figure FDA0004003727230000012
Each R is 1a Identical or different, R is the same or different 1a Independently selected from C 1 -C 10 Alkyl, C 3 -C 10 Cycloalkyl or 4-7 membered heterocyclyl, said SH, NH 2 、C 1 -C 10 Alkyl, C 1 -C 10 Alkoxy, C 3 -C 10 Cycloalkyl, C 3 -C 10 Cycloalkyloxy, 4-7 membered heterocyclyl or 4-7 membered heterocyclyloxy optionally substituted with R 1b Substituted, X 2 Selected from N or CR 2 ,R 2 Selected from H, halogen, CN, SH, NH 2 、C 1 -C 10 Alkyl, C 1 -C 10 Alkoxy, C 3 -C 10 Cycloalkyl, C 3 -C 10 Cycloalkyloxy, 4-7 membered heterocyclyl or 4-7 membered heterocyclyloxy, said SH, NH 2 、C 1 -C 10 Alkyl, C 1 -C 10 Alkoxy, C 3 -C 10 Cycloalkyl, C 3 -C 10 Cycloalkyloxy, 4-7 membered heterocyclyl or 4-7 membered heterocyclyloxy optionally substituted with R 2b Substitution; alternatively, X 1 、X 2 Taken together form a 4-14 membered heterocyclyl or a 5-6 membered heteroaryl, said 4-14 membered heterocyclyl or 5-6 membered heteroaryl optionally being substituted with R 2b Substitution; when X is 1 、X 2 X when not forming a ring 1 、X 2 With only double bonds, when X 1 、X 2 X is the same as X in the ring formation 1 、X 2 Connected by single bond or double bond;
R 3 、R 4 each independently selected from H, halogen, CN, OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl, said OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl optionally being substituted by R 3b Substitution;
R 5 selected from OH or NH 2 Said OH or NH 2 Optionally by R 5b Substitution of said OH or NH 2 Optionally by R 5b Substitution; and when R is 5 Selected from optionally R 5b Substituted NH 2 When X is 1 、X 2 Taken together form a 4-14 membered heterocyclyl or a 5-6 membered heteroaryl, said 4-14 membered heterocyclyl or 5-6 membered heteroaryl optionally being substituted with R 2b Substituted, or X 1 Selected from CR 1 ,R 1 Selected from SH or
Figure FDA0004003727230000013
Each R is 1a Identical or different, R is the same or different 1a Independently selected from C 1 -C 10 Alkyl, C 3 -C 10 Cycloalkyl or 4-7 membered heterocyclyl, said SH optionally being substituted by R 1b Substituted, X 2 Selected from N;
X 3 、X 4 selected from any one of the following:
1)X 3 selected from N or CR 6 ,X 4 Selected from N or CR 7 ,X 3 、X 4 Connected by double bonds, R 6 、R 7 Each independently selected from H, halogen, CN, OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl, said OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl optionally being substituted by R 6b Substitution; or alternatively, the process may be performed,
2)X 3 、X 4 one of them is selected from C (=O) and the other is selected from NR 8 ,X 3 、X 4 Connected by a single bond, R is 8 Selected from H, C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl, said C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl optionally being substituted by R 8b Substitution; or alternatively, the process may be performed,
3)X 3 、X 4 are joined together to form a 4-14 membered heterocyclyl or 5-6 membered heteroaryl, X 3 、X 4 Linked by a double bond, said 4-14 membered heterocyclyl or 5-6 membered heteroaryl optionally being bound by R 9b Substitution;
each R is 1b Or R is 3b Independently selected from deuterium, F, cl, br, I, CN, =o, OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl, C 2 -C 3 Alkenyl, C 2 -C 3 Alkynyl or 4-7 membered heterocyclyl, said OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl, C 2 -C 3 Alkenyl, C 2 -C 3 Alkynyl or 4-7 membered heterocyclyl are optionally substituted by R c Substitution;
each R is 2b 、R 5b 、R 6b 、R 8b Or R is 9b Independently selected from F, cl, br, I, CN, =o, OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl, said OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl optionally being substituted by R c Substitution;
each R is c Independently selected from F, cl, br, I, CN, =o, OH, NH 2 、C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl or 4-7 membered heterocyclyl;
with the proviso that the compound of formula (I) does not comprise
Figure FDA0004003727230000021
Figure FDA0004003727230000022
2. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein X 1 Selected from N or CR 1 ,R 1 Selected from SH, C 1 -C 6 Alkoxy, C 3 -C 6 Cycloalkyloxy or
Figure FDA0004003727230000023
The SH, C 1 -C 6 Alkoxy or C 3 -C 6 Cycloalkyl oxy is optionally substituted with R 1b And (3) substitution.
3. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 2, wherein X 1 Selected from N or CR 1 ,R 1 Selected from SH, methoxy, cyclopropyloxy or
Figure FDA0004003727230000031
Said SH, methoxy or cyclopropyloxy group being optionally substituted by R 1b And (3) substitution.
4. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein X 1 、X 2 Taken together form a 5-10 membered heterocyclyl or a 5-6 membered heteroaryl, said 5-10 membered heterocyclyl or 5-6 membered heteroaryl optionally being substituted with R 2b And (3) substitution.
5. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 4, wherein X 1 、X 2 Are joined together to form
Figure FDA0004003727230000032
Figure FDA0004003727230000033
Wherein Y is selected from CH 2 NH, O or S, said ≡>
Figure FDA0004003727230000034
Figure FDA0004003727230000035
Optionally by R 2b And (3) substitution.
6. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein X 3 Selected from CR 6 ,X 4 Selected from N, X 3 、X 4 Connected by double bonds, R 6 Selected from C optionally substituted by halogen 1 -C 3 An alkyl group.
7. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein X 3 、X 4 Are joined together to form a 4-7 membered heterocyclic group, X 3 、X 4 Linked by a double bond, said 4-7 membered heterocyclic group optionally being R 9b Substitution, said R 9b Selected from F, cl, br, I, CN, =o or C 1 -C 3 An alkyl group.
8. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 7, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is selected from the group consisting of the compounds of formula (I-6)
Figure FDA0004003727230000036
9. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 7, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is selected from the group consisting of the compounds of formula (I-7)
Figure FDA0004003727230000041
10. The compound of formula (I-7) or a pharmaceutically acceptable salt thereof according to claim 9, wherein X 1 Selected from N or CR 1 ,R 1 Selected from SH or
Figure FDA0004003727230000042
The SH is optionally substituted by R 1b And (3) substitution.
11. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is selected from the following compounds or pharmaceutically acceptable salts thereof:
Figure FDA0004003727230000043
Figure FDA0004003727230000051
Figure FDA0004003727230000061
12. a pharmaceutical composition comprising a compound according to any one of claims 1 to 11, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable adjuvant.
13. Use of a compound according to any one of claims 1 to 11, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 12, for the manufacture of a medicament for the prevention or treatment of HPK 1-related diseases.
CN202211624824.7A 2021-12-17 2022-12-16 Heterocyclic compound as HPK1 inhibitor and application thereof Pending CN116265470A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2021115522377 2021-12-17
CN202111552237 2021-12-17

Publications (1)

Publication Number Publication Date
CN116265470A true CN116265470A (en) 2023-06-20

Family

ID=86742844

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211624824.7A Pending CN116265470A (en) 2021-12-17 2022-12-16 Heterocyclic compound as HPK1 inhibitor and application thereof

Country Status (1)

Country Link
CN (1) CN116265470A (en)

Similar Documents

Publication Publication Date Title
EP3052476B1 (en) Substituted nicotinimide inhibitors of btk and their preparation and use in the treatment of cancer, inflammation and autoimmune disease
TW202110843A (en) Nitrogen-containing heterocyclic derivative regulator, preparation method therefor and application thereof
CN101678014B (en) Heterocyclic kinase modulators
CA2792508C (en) Piperidin-4-yl azetidine derivatives as jak1 inhibitors
CN106922146B (en) Pyrazole carboxamide compounds for the treatment of diseases mediated by Bruton's Tyrosine Kinase (BTK)
TW201127385A (en) N-containing heteroaryl derivatives as JAK3 kinase inhibitors
CA2883426A1 (en) Pyrrolotriazinone derivatives as pi3k inhibitors
JP2021506793A (en) Substituted bicyclic heterocyclic compounds as PRMT5 inhibitors
TW201546062A (en) Compositions useful for treating disorders related to KIT
CN102762565A (en) Pyrido[3,2-d]pyrimidine PI3[delta] inhibitor compounds and methods of use
KR20130116358A (en) Heterocyclic compounds as pi3 kinase inhibitors
CN110461841B (en) Nitrogen heteroaryl derivative with CSF1R inhibitory activity, and preparation method and application thereof
CN115279760A (en) Novel HPK1 inhibitor and preparation method and application thereof
JP7077323B2 (en) Quinazoline compound and its preparation method, use and pharmaceutical composition
WO2022184116A1 (en) New sos1 inhibitor, preparation method therefor and use thereof
KR20130029756A (en) N-7 substituted purine and pyrazolopyrimidine compounds, compositions and methods of use
CA3114259A1 (en) Aminonorbornane derivative and manufacture method therefor and use thereof
EP3546462B1 (en) Novel oxoisoquinoline derivative
WO2022171034A1 (en) Bicyclic compound as hpk1 inhibitor and application thereof
CN114685490A (en) Pyrrolopyridine compound and application thereof
CN114685489A (en) Polysubstituted nitrogen-containing heterocyclic compound and application thereof
CN116023378A (en) Lactam compounds as HPK1 inhibitors and uses thereof
WO2023041049A1 (en) Heterocyclic compound as sos1 inhibitor and uses thereof
JP7406674B2 (en) Spirocyclic compounds
WO2023036252A1 (en) Pyrrolopyrimidine or pyrrolopyridine derivative and medical use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
TA01 Transfer of patent application right

Effective date of registration: 20230816

Address after: 210032 No.99, Huakang Road, Jiangbei new district, Nanjing City, Jiangsu Province

Applicant after: Nanjing Zaiming Pharmaceutical Co.,Ltd.

Address before: 210042 699 Xuanwu Road, Xuanwu District, Nanjing, Jiangsu -18

Applicant before: JIANGSU SIMCERE PHARMACEUTICAL Co.,Ltd.

TA01 Transfer of patent application right