CN113330009A - Nitrogen heterocyclic compound, preparation method and application thereof - Google Patents

Nitrogen heterocyclic compound, preparation method and application thereof Download PDF

Info

Publication number
CN113330009A
CN113330009A CN202080008849.7A CN202080008849A CN113330009A CN 113330009 A CN113330009 A CN 113330009A CN 202080008849 A CN202080008849 A CN 202080008849A CN 113330009 A CN113330009 A CN 113330009A
Authority
CN
China
Prior art keywords
compound
radical
cancer
membered
aryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202080008849.7A
Other languages
Chinese (zh)
Other versions
CN113330009B (en
Inventor
李桂英
陈忠辉
冉茂盛
梅小波
韩晓军
蔡家强
田强
宋宏梅
薛彤彤
王晶翼
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Original Assignee
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sichuan Kelun Biotech Biopharmaceutical Co Ltd filed Critical Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Publication of CN113330009A publication Critical patent/CN113330009A/en
Application granted granted Critical
Publication of CN113330009B publication Critical patent/CN113330009B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings

Abstract

The invention relates to a nitrogen heterocyclic compound, a preparation method and application thereof, in particular to a compound shown in a formula I, a stereoisomer, a tautomer or a mixture thereof of the compound, a pharmaceutically acceptable salt, a eutectic crystal, a polymorphic substance or a solvate of the compound, or a stable isotope derivative, a metabolite or a prodrug of the compound, a preparation method and application thereof in treating tumor-related diseases.

Description

Nitrogen heterocyclic compound, preparation method and application thereof Technical Field
The invention belongs to the field of medicines, and particularly relates to a compound shown in a formula I, a stereoisomer, a tautomer or a mixture of the compound, a pharmaceutically acceptable salt, a eutectic crystal, a polymorphic substance or a solvate of the compound, or a stable isotope derivative, a metabolite or a prodrug of the compound. The compounds of the invention are useful as modulators (e.g., inhibitors) of PRMT5, for the treatment of diseases associated with PRMT5 activity (e.g., neoplastic diseases).
Figure PCTCN2020077581-APPB-000001
Background
PRMT5(Protein Arginine methyl transferase 5) belongs to an epigenetic enzyme, is one of the members of the PRMT family (human body has PRMT1-11), and can catalyze the methylation modification of Arginine residues of histone and some non-histone substrates. PRMT5 is widely present in the nucleus and cytoplasm of human cells, including tissues such as heart, muscle and testis. The arginine methylation is classified into I, II and III according to different catalytic arginine methylation modes, the PRMT5 belongs to type II symmetric double methylation (sDMA) PRMT, and the methyl donor is S-adenosylmethionine (SAM).
The PRMT5 regulates the expression degree of various target proteins by catalyzing the arginine methylation of a substrate, participates in various physiological functions, and has important effects on the proliferation, the metastasis and the malignant transformation of tumor cells. Methylation modification of histone by PRMT5 leads to silencing of cancer suppressor genes such as p53, ST7, NM23 and Rb, and further promotes the occurrence and development of tumors. The regulation of non-histones by PRMT5 is mainly characterized by affecting the localization and expression of transcription factors (NF-. kappa.B/P65, E2F1, HoxA \ GATA4), programmed cell death protein 4(PDCD4), cell cycle and survival-related regulatory protein E2F1, hypoxia inducible factor 1(HIF-1), cyclin-dependent kinases (CDKs), PI3K/Akt, etc. (Koh CM, Bezzi M, Guccione E. curr Mol Bio Rep,2015,1(1): 19-28). In lung cancer cells, PRMT5 can inhibit miR-99 family transcription, increase FGFR3 expression, activate Erk1/2 and Ak pathways, leading to tumor cell growth and metastasis (Pengyu stringing, Nan Zhao, et al cancer Letters,2018,427, 38-48). PRMT5 methylates Eif4e and FGFR3 in colon cancer, promoting tumor cell growth (ZHANG B, DONG S, ZHU R, et al. Oncostatt, 2015,6(26): 22799-.
In conclusion, the PRMT5 target point has a clear mechanism for regulating tumor proliferation and metastasis, and is a promising tumor treatment target point. No inhibitors of the PRMT5 target are currently on the market. Therefore, there is a need to develop new PRMT5 inhibitors with high efficacy and low toxicity to meet clinical needs.
Disclosure of Invention
The invention aims to provide a compound shown in a formula I, a preparation method thereof and application thereof in preventing and/or treating diseases related to PRMT5 activity, such as tumor diseases.
In one aspect, the present invention relates to a compound of formula I, a stereoisomer, a tautomer, or a mixture thereof of said compound, a pharmaceutically acceptable salt, a co-crystal, a polymorph, or a solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound:
Figure PCTCN2020077581-APPB-000002
wherein:
ring A is selected from C6-12Aryl and 5-to 10-membered heteroaryl, said C6-12Aryl, 5-10 membered heteroaryl may be optionally substituted with one or more of halogen, OH, CN, C1-4Alkyl radical, C1-4An alkoxy group;
ring B is selected from C6-12Aryl, 5-10 membered heteroaryl, C3-8Cycloalkyl, 4-10 membered heterocyclyl, 9-12 membered arylheterocyclyi, 9-12 membered aryloateroaryl and 9-12 membered aryloaycloalkyl;
R aand RbIndependently selected from H, halogen, OH, CN, NH2、NO 2、C 1-4Alkyl radical, C1-4Haloalkyl, C1-4Hydroxyalkyl radical, C1-4Alkoxy radical, C1-4Haloalkoxy and C3-8Cycloalkyl, or two RaAre linked to form a 3-8 membered cycloalkyl or 3-8 membered heterocyclyl; when a plurality of RaWhen occurring simultaneously, each RaMay be attached to the same or different carbon atoms and may be the same or differentThe same is carried out; when a plurality of RbWhen occurring simultaneously, each RbMay be attached to the same or different carbon atoms and may be the same or different;
R cselected from halogen, OH, CN, NO2、C 1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, 9-12 membered aryloateroaryl, 9-12 membered aryloateroheterocyclyl, 9-12 membered aryloaycloalkyl, CO2R 20、 C(O)SR 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、OS(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、C(O)NR 21SO 2R 25、OR 26、SR 26、OC(O)R 20、OCO 2R 20、OC(O)NR 21R 22、NR 23C(O)NR 21R 22、NR 23C(O)OR 20、C(=NR 21)R 22、C(=NR 21)OR 20、OC(=NR 23)R 21、OC(=NR 23)OR 20、C(=NR 23)NR 21R 22、OC(=NR 23)NR 21R 22、C(=NOR 20)R 24And NNR21R 22(ii) a Said C is1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, 9-12 membered aryloateroaryl, 9-12 membered aryloateroheterocyclyl, 9-12 membered aryloaycloalkyl may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Haloalkyl, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, 9-12 membered arylheterocyclo, 9-12 membered arylcycloalkylo, CO2R 20、C(O)SR 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、OS(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、C(O)NR 21SO 2R 25、OR 26、SR 26、OC(O)R 20、OCO 2R 20、OC(O)NR 21R 22、NR 23C(O)NR 21R 22、NR 23C(O)OR 20、C(=NR 21)R 22、C(=NR 21)OR 20、OC(=NR 23)R 21、OC(=NR 23)OR 20、C(=NR 23)NR 21R 22、OC(=NR 23)NR 21R 22、C(=NOR 20)R 24、=NNR 21R 22(ii) a When it is muchR iscWhen occurring simultaneously, each RcMay be the same or different;
R 20、R 21、R 22、R 23、R 24、R 25、R 26each independently selected from: H. c1-6Alkyl radical, C1-6Hydroxyalkyl radical, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl, 4-10 membered heterocyclyl, C6-12Aryl and 5-10 membered heteroaryl; said C is1-6Alkyl radical, C1-6Hydroxyalkyl radical, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl, 4-10 membered heterocyclyl, C6-12Aryl, 5-10 membered heteroaryl may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Haloalkoxy, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl-C1-4Alkyl-, CO2R 30、C(O)R 30、C(O)NR 31R 32、NR 33C(O)R 34、NR 31R 32、S(O) 2R 35、NR 31SO 2R 35、S(O)NR 31R 32、S(O) 2NR 31R 32(ii) a Or R21And R22Taken together to form a 3-8 membered heterocyclyl;
R 30、R 31、R 32、R 33、R 34and R35Each independently selected from: H. c1-6Alkyl radical, C1-6Hydroxyalkyl radical, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl, 4-10 membered heterocyclyl, C6-12Aryl and 5-10 membered heteroaryl; said C is1-6Alkyl radical, C1-6Hydroxyalkyl radical, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl, 4-10 membered heterocyclyl, C6-12Aryl, 5-10 membered heteroaryl may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Haloalkoxy, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl; or R31And R32Taken together to form a 3-8 membered heterocyclyl;
m is selected from 0, 1 and 2;
n is selected from 0, 1 and 2, and m + n is less than or equal to 3;
p is selected from 0 and 1;
q is selected from 0, 1,2,3 and 4;
t is selected from 0, 1,2,3 and 4;
s is selected from 0, 1,2,3,4 and 5.
In a preferred embodiment, ring A is C6-12Aryl radical, said C6-12Aryl groups may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-4Alkyl radical, C1-4An alkoxy group; preferably, a is phenyl.
In a preferred embodiment, ring B is selected from C6-12Aryl and 5-10 membered heteroaryl; preferably, ring B is selected from phenyl, pyridyl, pyridazinyl, pyrimidinyl and pyrazinyl.
In a preferred embodiment, RaSelected from H, halogen, C1-4Alkyl and C1-4An alkoxy group; when a plurality of RaWhen occurring simultaneously, each RaMay be attached to the same or different carbon atoms and may be the same or different.
In a more preferred embodiment, RaIs H.
In a preferred embodiment, RbSelected from H, halogen, OH, CN, C1-4Alkyl radical, C1-4Haloalkyl, C1-4Hydroxyalkyl radical, C1-4Alkoxy and C1-4A haloalkoxy group; when a plurality of RbWhen occurring simultaneously, each RbMay be attached to the same or different carbon atoms and may be the same or different.
In a more preferred embodiment, RbIs H.
In a preferred embodiment, RcSelected from halogen, OH, CN, NO2、C 1-6Alkyl radical, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, CO2R 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、OR 26、SR 26、OC(O)R 20And NR23C(O)NR 21R 22(ii) a Said C is1-6Alkyl radical, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl may be optionally substituted with one or more of halogen, OH, CN, C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Haloalkyl, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, 9-12 membered arylheterocyclo, 9-12 membered arylcycloalkylo, CO2R 20、C(O)SR 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、OS(O) 2R 25、NR 21SO 2R 25、 S(O)NR 21R 22、S(O) 2NR 21R 22、C(O)NR 21SO 2R 25、OR 26、SR 26、OC(O)R 20、OCO 2R 20、OC(O)NR 21R 22、NR 23C(O)NR 21R 22、NR 23C(O)OR 20、C(=NR 21)R 22、C(=NR 21)OR 20、OC(=NR 23)R 21、OC(=NR 23)OR 20、C(=NR 23)NR 21R 22、OC(=NR 23)NR 21R 22、C(=NOR 20)R 24、=NNR 21R 22(ii) a When a plurality of RcWhen occurring simultaneously, each RcMay be the same or different.
In a preferred embodiment, RcSelected from halogen, OH, CN, NO2、C 1-4Alkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl radical, C1-4Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, CO2R 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、OR 26、SR 26、OC(O)R 20And NR23C(O)NR 21R 22(ii) a Said C is1-4Alkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl radical, C1-4Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-4Alkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl radical, C1-4Haloalkyl, C1-4Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, CO2R 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、OR 26、SR 26、OC(O)R 20、NR 23C(O)NR 21R 22(ii) a When a plurality of RcWhen occurring simultaneously, each RcMay be the same or different.
In a preferred embodiment, R20、R 21、R 22、R 23、R 24、R 25、R 26Each independently selected from H, C1-4Alkyl radical、C 1-4Hydroxyalkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl, 4-10 membered heterocyclyl, C6-12Aryl and 5-10 membered heteroaryl; said C is1-4Alkyl radical, C1-4Hydroxyalkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl, 4-10 membered heterocyclyl, C6-12Aryl, 5-10 membered heteroaryl may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-4Alkyl radical, C1-4Haloalkyl, C1-4Haloalkoxy, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl radical, C1-4Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl-C1-4Alkyl-, CO2R 30、C(O)R 30、C(O)NR 31R 32、NR 33C(O)R 34、NR 31R 32、S(O) 2R 35、NR 31SO 2R 35、S(O)NR 31R 32、S(O) 2NR 31R 32(ii) a Or R21And R22Joined together to form a 3-8 membered heterocyclic group.
In a preferred embodiment, R20、R 21、R 22、R 23、R 24、R 25、R 26Each independently selected from H, C1-3Alkyl radical, C1-3Hydroxyalkyl radical, C1-3Heteroalkyl group, C1-3Alkoxy radical, C3-6Cycloalkyl, 4-6 membered heterocyclyl, C6-10Aryl and 5-6 membered heteroaryl; said C is1-3Alkyl radical, C1-3Hydroxyalkyl radical, C1-3Heteroalkyl group, C1-3Alkoxy radical, C3-6Cycloalkyl, 4-to 6-memberedHeterocyclic group, C6-10Aryl, 5-6 membered heteroaryl may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-3Alkyl radical, C1-3Haloalkyl, C1-3Haloalkoxy, C1-3Heteroalkyl group, C1-3Alkoxy radical, C3-6Cycloalkyl radical, C1-3Hydroxyalkyl, 4-6 membered heterocyclyl, C6-10Aryl, 5-6 membered heteroaryl-C1-4Alkyl-, CO2R 30、C(O)R 30、C(O)NR 31R 32、NR 33C(O)R 34、NR 31R 32、S(O) 2R 35、NR 31SO 2R 35、S(O)NR 31R 32、S(O) 2NR 31R 32(ii) a Or R21And R22Joined together to form a 3-8 membered heterocyclic group.
In a preferred embodiment, R30、R 31、R 32、R 33、R 34、R 35Each independently selected from H, C1-4Alkyl radical, C1-4Hydroxyalkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-6Cycloalkyl, 4-6 membered heterocyclyl, C6-10Aryl and 5-6 membered heteroaryl; said C is1-4Alkyl radical, C1-4Hydroxyalkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-6Cycloalkyl, 4-6 membered heterocyclyl, C6-10Aryl, 5-6 membered heteroaryl may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-4Alkyl radical, C1-4Haloalkyl, C1-4Haloalkoxy, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-6Cycloalkyl radical, C1-4Hydroxyalkyl, 4-6 membered heterocyclyl, C6-10Aryl, 5-6 membered heteroaryl; or R31And R32Joined together to form a 3-8 membered heterocyclic group.
In a preferred embodiment, R30、R 31、R 32、R 33、R 34、R 35Each independently selected from H, C1-4Alkyl radical, C3-6Cycloalkyl and C1-4An alkoxy group.
In a preferred embodiment, R30、R 31、R 32、R 33、R 34、R 35Each independently selected from the group consisting of H, methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, cyclopropyl, and propoxy.
In a preferred embodiment, RcIndependently selected from halogen, C1-6Alkyl radical, C3-8Cycloalkyl radical, C1-6Alkoxy, 6-membered heterocyclic group, NR21R 22、OR 26And SR26Wherein R is21、R 22、R 26Each independently selected from: H. c1-6Alkyl, 4-6 membered heterocyclyl, 5-6 membered heteroaryl and C3-8Cycloalkyl radical, said C1-6Alkyl, 4-6 membered heterocyclyl, 5-6 membered heteroaryl and C3-8Cycloalkyl is optionally substituted with one or more of the following substituents: c1-6Alkyl, 4-6 membered heterocyclyl, 5-6 membered heteroaryl, C3-8Cycloalkyl radical, C1-6alkyl-C (O) -, C3-6cycloalkyl-C (O) -, C1-4alkyl-S (O)2And 6-membered heteroaryl-C1-4An alkyl group-.
In a preferred embodiment, RcIndependently selected from halogen, C1-4Alkyl radical, C3-6Cycloalkyl radical, C1-4Alkoxy, morpholinyl, NR21R 22、OR 26And SR26Wherein R is21、R 22、R 26Each independently selected from: H. optionally substituted piperidinyl, optionally substituted pyrrolidinyl, optionally substituted azetidinyl, optionally substituted oxetanyl, optionally substituted tetrahydrofuranyl, optionally substituted tetrahydropyranyl, optionally substituted pyridylmethyl, optionally substituted cyclohexyl, optionally substituted cyclobutyl, and optionally substituted cyclopropylmethyl, wherein the optionally substituted group is optionally substituted with one or more of the following substituents: c1-4alkyl-C (O) -, C3-6cycloalkyl-C (O) -, C1-4alkyl-S (O)2-and 6-membered heteroarylmethyl (preferably pyrazinylmethyl); preferably, the substituent is located on the nitrogen atom of the piperidinyl, pyrrolidinyl, azetidinyl.
In a more preferred embodiment, RcIndependently selected from halogen (e.g. chlorine), methyl, ethyl, isopropyl, cyclopropyl, methoxy, morpholino, methyl, ethyl, methyl, propyl, butyl, or a salt, butyl, or a salt thereof,
Figure PCTCN2020077581-APPB-000003
Figure PCTCN2020077581-APPB-000004
In a preferred embodiment, RcIn the meta and/or para position relative to the carbonyl group attached to ring B.
In a preferred embodiment, m is 1.
In a preferred embodiment, n is selected from 1 and 2.
In a preferred embodiment, p is 1.
In a preferred embodiment, q is 0.
In a preferred embodiment, t is 0.
In a preferred embodiment, s is selected from 1 and 2.
In some embodiments of the invention, the compound has the structure of formula II-A:
Figure PCTCN2020077581-APPB-000005
wherein, ring A, Ra、R b、R cM, n, p, q, s, t are as defined above for formula I.
In some embodiments of the invention, the compound has the structure of formula II-B:
Figure PCTCN2020077581-APPB-000006
wherein, ring A, Ra、R b、R cM, n, p, q, s, t are as defined above for formula I.
In some embodiments of the invention, the compound has the structure of formula II-C:
Figure PCTCN2020077581-APPB-000007
wherein, ring A, Ra、R b、R cM, n, p, q, s, t are as defined above for formula I.
In some embodiments of the invention, the compound of formula II-A, II-B or II-C:
ring A is C6-12Aryl radical, said C6-12Aryl groups may be optionally substituted with one or more of halogen, OH, CN, C1-4Alkyl radical, C1-4An alkoxy group;
R aselected from H, halogen, C1-4Alkyl and C1-4An alkoxy group; when a plurality of RaWhen occurring simultaneously, each RaMay be attached to the same or different carbon atoms and may be the same or different;
R bSelected from H, halogen, OH, CN, C1-4Alkyl radical, C1-4Haloalkyl, C1-4Hydroxyalkyl radical, C1-4Alkoxy and C1-4A haloalkoxy group; when a plurality of RbWhen occurring simultaneously, each RbMay be attached to the same or different carbon atoms and may be the same or different;
R cselected from halogen, OH, CN, NO2、C 1-4Alkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl radical, C1-4Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, CO2R 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、OR 26、SR 26、OC(O)R 20And NR23C(O)NR 21R 22(ii) a Said C is1-4Alkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl radical, C1-4Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-4Alkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl radical, C1-4Haloalkyl, C1-4Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, CO2R 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、OR 26、SR 26、OC(O)R 20、NR 23C(O)NR 21R 22(ii) a When a plurality of RcWhen occurring simultaneously, each RcMay be the same or different;
R 20、R 21、R 22、R 23、R 24、R 25、R 26each independently selected from H, C1-3Alkyl radical, C1-3Hydroxyalkyl radical, C1-3Heteroalkyl group, C1-3Alkoxy radical, C3-6Cycloalkyl, 4-6 membered heterocyclyl, C6-10Aryl and 5-6 membered heteroaryl; said C is1-3Alkyl radical, C1-3Hydroxyalkyl radical, C1-3Heteroalkyl group, C1-3Alkoxy radical, C3-6Cycloalkyl, 4-6 membered heterocyclyl, C6-10Aryl, 5-6 membered heteroaryl may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-3Alkyl radical, C1-3Haloalkyl, C1-3Haloalkoxy, C1-3Heteroalkyl group, C1-3Alkoxy radical, C3-6Cycloalkyl radical, C1-3Hydroxyalkyl, 4-6 membered heterocyclyl, C6-10Aryl, 5-6 membered heteroaryl-C1-4Alkyl-, CO2R 30、C(O)R 30、C(O)NR 31R 32、NR 33C(O)R 34、NR 31R 32、S(O) 2R 35、NR 31SO 2R 35、S(O)NR 31R 32、S(O) 2NR 31R 32(ii) a Or R21And R22Taken together to form a 3-8 membered heterocyclyl;
R 30、R 31、R 32、R 33、R 34、R 35each independently selected from H, methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, cyclopropyl, and propoxy;
m is selected from 0, 1 and 2;
n is selected from 0, 1 and 2, and m + n is less than or equal to 3;
p is selected from 0 and 1;
q is selected from 0, 1,2,3 and 4;
s is selected from 0, 1,2,3,4 and 5; and is
t is selected from 0, 1,2,3 and 4.
In some embodiments of the invention, the compound of formula II-a:
ring A is phenyl; raIs H; rbIs H;
R cselected from halogen (e.g. chlorine), methyl, ethyl, isopropyl, cyclopropyl, methoxy, morpholino, methyl, ethyl, propyl, methyl, propyl, butyl, or a salt thereof,
Figure PCTCN2020077581-APPB-000008
m is selected from 1 and 2;
n is selected from 1 and 2, and m + n is less than or equal to 3;
p is 1;
q is 1;
s is selected from 1 and 2; and t is 1.
In some embodiments of the invention, the compound of formula II-B:
ring A is phenyl; raIs H; rbIs H;
R cis selected from
Figure PCTCN2020077581-APPB-000009
m is 1;
n is 1;
p is 1;
q is 1;
s is 1; and t is 1.
In some embodiments of the invention, the compound of formula II-C:
ring A is phenyl; raIs H; rbIs H;
R cis selected from
Figure PCTCN2020077581-APPB-000010
m is 1;
n is 1;
p is 1;
q is 1;
s is 1; and t is 1.
In some embodiments of the invention, the compound of formula II-C:
ring A is phenyl; raIs H; rbIs H;
R cis selected from
Figure PCTCN2020077581-APPB-000011
m is selected from 1 and 2;
n is selected from 1 and 2, and m + n is less than or equal to 3;
p is 1;
q is 1;
s is 1; and t is 1.
In some embodiments of the invention, the compounds of the invention are selected from, but not limited to:
Figure PCTCN2020077581-APPB-000012
Figure PCTCN2020077581-APPB-000013
in another aspect, the present invention provides a pharmaceutical composition comprising a compound as described above, a stereoisomer, a tautomer, or a mixture thereof of said compound, a pharmaceutically acceptable salt, a co-crystal, a polymorph, or a solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound. Optionally, the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers.
In some embodiments, the pharmaceutical composition is for use in the prevention and/or treatment of a disease associated with PRMT5 activity (e.g., a neoplastic disease).
In another aspect, the present invention provides a pharmaceutical formulation comprising a compound as described above, a stereoisomer, a tautomer, or a mixture thereof of said compound, a pharmaceutically acceptable salt, a co-crystal, a polymorph, or a solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound, or a pharmaceutical composition as described above.
In another aspect, the present invention provides a use of a compound as described above, a stereoisomer, a tautomer, or a mixture thereof, a pharmaceutically acceptable salt, a co-crystal, a polymorph, or a solvate of the compound, or a stable isotope derivative, metabolite, or prodrug of the compound, or a pharmaceutical composition as described above, or a pharmaceutical formulation as described above, for the manufacture of a medicament for the prevention and/or treatment of a disease associated with PRMT5 activity (e.g. a neoplastic disease).
In another aspect, the present invention provides the use of a compound as described above, a stereoisomer, a tautomer, or a mixture thereof, a pharmaceutically acceptable salt, a co-crystal, a polymorph, or a solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound, or a pharmaceutical composition as described above, for the manufacture of a formulation for modulating (e.g. reducing) the activity of PRMT 5.
In another aspect, the present invention provides a method of preparing a pharmaceutical composition of the present invention, comprising combining a compound of the present invention, or a stereoisomer, a tautomer, or a mixture thereof, or a pharmaceutically acceptable salt, polymorph, co-crystal, solvate, metabolite, or prodrug thereof, with one or more pharmaceutically acceptable carriers.
In another aspect, the present invention provides a method of treating a disease associated with PRMT5 activity (e.g. a neoplastic disease), comprising administering to a subject in need thereof a therapeutically and/or prophylactically effective amount of a compound of the present invention, a stereoisomer, a tautomer or a mixture thereof of said compound, a pharmaceutically acceptable salt, a co-crystal, a polymorph or a solvate of said compound, or a stable isotope derivative, metabolite or prodrug of said compound, or a pharmaceutical composition as described above, or a pharmaceutical formulation as described above.
In the present invention, the disease associated with PRMT5 activity is a tumor disease, including but not limited to: brain tumor, lung cancer, squamous cell carcinoma, bladder cancer, stomach cancer, ovarian cancer, peritoneal cancer, pancreatic cancer, breast cancer, head and neck cancer, cervical cancer, endometrial cancer, rectal cancer, liver cancer, kidney cancer, esophageal adenocarcinoma, esophageal squamous cell carcinoma, prostate cancer, cancer of the female genital tract, carcinoma in situ, leukemia, lymphoma, neurofibroma, thyroid cancer, bone cancer, skin cancer, brain cancer, colon cancer, testicular cancer, gastrointestinal stromal tumor, prostate tumor, mast cell tumor, multiple myeloma, melanoma, glioma, or sarcoma.
Definition of terms
Unless defined otherwise below, all technical and scientific terms used herein are intended to have the same meaning as commonly understood by one of ordinary skill in the art. Reference to the techniques used herein is intended to refer to those techniques commonly understood in the art, including those variations of or alternatives to those techniques that would be apparent to those skilled in the art. While the following terms are believed to be well understood by those skilled in the art, the following definitions are set forth to better explain the present invention.
The terms "comprising," "including," "having," "containing," or "involving," and other variations thereof herein, are inclusive or open-ended and do not exclude additional unrecited elements or method steps.
The term "halo" refers to a halogen substituent, and includes F, Cl, Br, or I. It is to be understood that when referred to as a group, the term "halogen" refers to a halogen substituent, including F, Cl, Br, or I.
The term "alkyl" is a straight or branched chain saturated aliphatic hydrocarbon group. The term "C1-6Alkyl "and" C1-4Alkyl "refers to a straight or branched chain alkyl group having 1 to 6 carbon atoms and 1-4 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl or n-hexyl, respectively.
The term "heteroalkyl" refers to an alkyl group having one or more backbone chain atoms selected from atoms other than carbon (e.g., oxygen, nitrogen, sulfur, phosphorus, or combinations thereof), wherein the heteroatom is not terminal to the heteroalkyl group. The numerical ranges refer to the number of carbons in the chain, C1-6Heteroalkyl is meant to include 1 to 6 carbon atoms, for example containing 2,3,4, 5 or 6 carbon atoms. For example, -CH2OCH 2CH 3The radical being designated C3A heteroalkyl group. For example, heteroalkyl groups contain 1-3, e.g., 1-2, preferably 1, heteroatoms selected from oxygen, nitrogen and sulfur.
The term "alkoxy" refers to a group having the structure "alkyl-O-", wherein alkyl is as defined above, preferably C1- 6Alkoxy radical, C1-4Alkoxy or C1- 3An alkoxy group. C1- 6Representative examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, isopropoxy, n-propoxy, n-butoxy, isobutoxy, t-butoxy, pentyloxy, hexyloxy, and the like.
The term "haloalkyl" refers to a group consisting ofOne or more (such as 1 to 3) identically or differently halogen-substituted alkyl groups, the term "C1-6Haloalkyl "and" C1-4Haloalkyl "refers to haloalkyl groups having 1 to 6 carbon atoms and 1-4 carbon atoms, respectively, e.g., -CF3、-C 2F 5、-CHF 2、-CH 2F、-CH 2CF 3、-CH 2Cl or-CH2CH 2CF 3And the like.
The term "hydroxyalkyl" refers to a group formed by replacement of a hydrogen atom of an alkyl group by one or more, e.g. one, hydroxyl group, e.g. C1-6Hydroxyalkyl radical, C1-4Hydroxyalkyl or C1-3Hydroxyalkyl groups, examples of which include, but are not limited to, hydroxymethyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, -CH (OH) CH3And the like.
The term "alkenyl" refers to a monovalent straight or branched chain hydrocarbon group containing one or more carbon-carbon double bonds, including but not limited to C2-6Alkenyl radical, C2-4Alkenyl, e.g. -CH ═ CH2、-CH 2CH=CH 2、-C(CH 3)=CH 2、-CH 2-CH=CH-CH 3And the like.
The term "alkynyl" refers to a monovalent straight or branched chain hydrocarbon radical having one or more carbon-carbon triple bonds, including but not limited to C2-6Alkynyl, C2-4Alkynyl groups such as ethynyl, 2-propynyl, 2-butynyl, and 1, 3-butadiynyl and the like.
The term "cycloalkyl" refers to a saturated or unsaturated non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring group, including, but not limited to, monocyclic cycloalkyl (such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl), and bicycloalkyl, including spiro, fused (fused) ring, or bridged ring systems (i.e., spirocycloalkyl, fused (fused) ring alkyl, and bridged cycloalkyl, such as bicyclo [1.1.1] pentyl, bicyclo [2.2.1] heptyl, and the like).
The term "3-8 membered cycloalkyl" or "C3-8Cycloalkyl "is used interchangeably and refers to a cycloalkyl group having 3 to 8 ring-forming carbon atoms, which may be a monocycloalkyl group, e.g., C3-6Cycloalkyl radical, C3-5Cycloalkyl radical, C3-4Cycloalkyl, e.g. cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and may also be bicycloalkyl, e.g. C3-8Spiro cycloalkyl, C3-8Bridged cycloalkyl radical, C3-8Condensed ring alkyl, C5-7Spiro cycloalkyl, C5-7Bridged cycloalkyl radical, C4-7A fused ring alkyl group.
The term "fused ring" or "fused ring" refers to a ring system formed by two or more ring structures sharing two adjacent atoms with each other.
The term "spiro" refers to a ring system formed by two or more cyclic structures sharing a ring atom with each other.
The term "bridged ring" refers to a ring system formed by two or more cyclic structures sharing two atoms not directly attached to each other.
The term "aryl" refers to an all-carbon monocyclic or fused polycyclic aromatic group having a conjugated pi-electron system. As used herein, the term "C6-12Aryl "means an aryl group containing from 6 to 12 carbon atoms, e.g. C6-10Aryl, for example, is phenyl or naphthyl.
The term "arylocycloalkyl" refers to a fused ring group formed by an aryl and a cycloalkyl (e.g., monocyclic cycloalkyl) group sharing two adjacent atoms with each other, wherein the point of attachment to other groups can be either on the aryl or on the cycloalkyl. The term "9-12 membered arylcycloalkylalkyl" refers to arylcycloalkylalkyl groups containing a total of 9-12 ring atoms, such as, for example, phenylcyclopentyl, phenylcyclohexyl,
Figure PCTCN2020077581-APPB-000014
the term "heterocyclyl" refers to a group having 2 or more (e.g., 3,4, 5,6, 7, 8, 9, 10, 11, 12, 13, or 14) carbon atoms and oneA saturated or partially unsaturated monocyclic or polycyclic (e.g. bicyclic, spirocyclic or bridged) group of one or more (e.g. 1,2,3 or 4) heteroatoms including, but not limited to, oxygen, nitrogen, sulfur atoms, carbon and sulfur heteroatoms of said heterocyclic group being optionally substituted by oxo (oxo) (e.g. to form C-O, S (═ O) or S (═ O)2)。
The terms "4-10 membered heterocyclyl", "3-8 membered heterocyclyl" mean heterocyclyl containing 4-10 or 3-8 ring atoms, respectively, including but not limited to 4-8 membered heterocyclyl, 4-7 membered heterocyclyl, 4-6 membered heterocyclyl, 5-6 membered heterocyclyl, 3-7 membered heterocyclyl, 4 membered heterocyclyl, 5 membered heterocyclyl, 6 membered heterocyclyl and the like, which heterocyclyl contains one or more (e.g., 1,2 or 3, more e.g., 1 or 2) heteroatoms selected from oxygen, nitrogen and sulfur, e.g., 4-7 membered nitrogen containing heterocyclyl, 4-7 membered oxygen containing heterocyclyl, 4-7 membered sulfur containing heterocyclyl, 5-6 membered nitrogen containing heterocyclyl, 5-6 membered oxygen containing heterocyclyl, 5-6 membered sulfur containing heterocyclyl. The "nitrogen-containing heterocyclic group", "oxygen-containing heterocyclic group", "sulfur-containing heterocyclic group" optionally further contains one or more other hetero atoms selected from oxygen, nitrogen, sulfur. Examples of 4-10 membered heterocyclyl or 3-8 membered heterocyclyl include, but are not limited to, oxiranyl, aziridinyl, azetidinyl, oxetanyl, tetrahydrofuranyl, pyrrolidinyl, pyrrolidinonyl, imidazolidinyl, pyrazolidinyl, tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, trithianyl,
Figure PCTCN2020077581-APPB-000015
And the like.
In the present invention, a heterocyclic group may form a ring-merged structure with a heterocyclic group or a cycloalkyl group, and the point of attachment of the ring-merged structure to other groups may be on any of the heterocyclic groups or the cycloalkyl group, and therefore, the heterocyclic group of the present invention also includes, but is not limited to, heterocyclo-heterocyclic groups, heterocyclo-cycloalkyl groups, monocyclic heterocyclo-monocyclic heterocyclic groups, monocyclic heterocyclo-monocyclic cycloalkyl groups, such as 3-7-membered (mono) heterocyclo-3-7-membered (mono) heterocyclic groups, 3-7-membered (mono) heterocyclo- (mono) cycloalkane-cyclic groupsRadical, 3-7 membered (mono) heterocyclo C4-6(mono) cycloalkyl, examples of which include, but are not limited to, pyrrolidinyl-cyclopropyl, cyclopent-aziridinyl, pyrrolidinyl-cyclobutyl, pyrrolidinyl-pyrrolidinyl, pyrrolidinylpiperidinyl, pyrrolidinylpiperazinyl, piperidino-morpholinyl, pyrrolidinyl-piperazinyl, piperidinyl-piperidinyl, pyrrolidinyl-piperidinyl, piperidinyl-and/or piperidinyl-cyclopropyl, pyrrolidinyl-and-pyrrolidinyl, piperidinyl-and-pyrrolidinyl, and the like,
Figure PCTCN2020077581-APPB-000016
Figure PCTCN2020077581-APPB-000017
In the present invention, the heterocyclic group also includes bridged heterocyclic groups and spiro heterocyclic groups.
The term "bridged heterocyclic ring" refers to a cyclic structure containing one or more (e.g., 1,2,3, or 4) heteroatoms (e.g., oxygen, nitrogen, sulfur atoms) formed by two saturated rings that share two ring atoms that are not directly connected, including, but not limited to, 7-10 membered bridged heterocyclic rings, 8-10 membered bridged heterocyclic rings, 7-10 membered nitrogen-containing bridged heterocyclic rings, 7-10 membered oxygen-containing bridged heterocyclic rings, 7-10 membered sulfur-containing bridged heterocyclic rings, and the like, e.g.
Figure PCTCN2020077581-APPB-000018
Figure PCTCN2020077581-APPB-000019
Figure PCTCN2020077581-APPB-000020
And the like. The "nitrogen-containing bridged heterocyclic ring", "oxygen-containing bridged heterocyclic ring", "sulfur-containing bridged heterocyclic ring" optionally further contains one or more other heteroatoms selected from oxygen, nitrogen, sulfur.
The term "spiroheterocycle" refers to a ring formed by two or more saturated rings sharing a ring atom and containing one or more (e.g., 1,2,3, or 4) heteroatoms (e.g., oxygen, nitrogen, sulfur atoms)Son) including, but not limited to, 5-10 membered spiroheterocycles, 6-10 membered nitrogen-containing spiroheterocycles, 6-10 membered oxygen-containing spiroheterocycles, 6-10 membered sulfur-containing spiroheterocycles, and the like, e.g.
Figure PCTCN2020077581-APPB-000021
Figure PCTCN2020077581-APPB-000022
Figure PCTCN2020077581-APPB-000023
Said "nitrogen-containing spiroheterocycle", "oxygen-containing spiroheterocycle", "sulfur-containing spiroheterocycle" optionally further contains one or more other heteroatoms selected from oxygen, nitrogen, sulfur. The term "6-to 10-membered nitrogen-containing spiroheterocyclic group" means a spiroheterocyclic group containing a total of 6 to 10 ring atoms and in which at least one ring atom is a nitrogen atom.
The term "arylheterocyclo" refers to a cyclic group formed by an aryl group and a heterocyclyl group sharing two adjacent carbon atoms with each other, and the point of attachment to the other group may be on the aryl or heterocyclyl group. Wherein aryl and heterocyclyl are as defined above. For example, as used herein, the term "9-12 membered arylheterocyclo" means a group containing a total of 9-12 ring atoms of the arylheterocyclo, including but not limited to 9-10 membered benzoheterocyclo, e.g., phenyl and 5-8 membered heterocyclo, e.g., phenyl and 5-6 membered heterocyclo, e.g., benzo 5-6 membered monocyclic heterocyclo, benzo 5-6 membered nitrogen containing monocyclic heterocyclo, benzo 5-6 membered oxygen containing monocyclic heterocyclo, benzo 5-6 membered sulfur containing heterocyclo, said "nitrogen containing heterocyclyl", "oxygen containing heterocyclyl", "sulfur containing heterocyclyl" optionally also containing one or more other heteroatoms selected from oxygen, nitrogen, sulfur. The carbon and sulfur heteroatoms of the heterocyclyl are optionally oxo (oxo) (e.g., to form C ═ O, S (═ O) or S (═ O)2)。
Examples include, but are not limited to: indazolyl group,
Figure PCTCN2020077581-APPB-000024
Figure PCTCN2020077581-APPB-000025
The term "heteroaryl" refers to a monocyclic or polycyclic aromatic group containing one or more of the same or different heteroatoms, including monocyclic heteroaryl and bicyclic or polycyclic ring systems containing at least one heteroaromatic ring (an aromatic ring system containing at least one heteroatom), which may have 5,6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 ring atoms, for example 5,6, 7, 8, 9, or 10 ring atoms. The heteroatom may be oxygen, nitrogen or sulfur.
The term "5-10 membered heteroaryl" means a heteroaryl group containing 5 to 10 ring atoms, including 5-6 membered heteroaryl, 5-6 membered monocyclic heteroaryl, 5-membered monocyclic heteroaryl, 6 membered monocyclic heteroaryl, and the like, said heteroaryl group containing one or more (e.g., 1,2, or 3, more e.g., 1 or 2) heteroatoms selected from oxygen, nitrogen, and sulfur, e.g., 5-10 membered nitrogen-containing heteroaryl, 5-10 membered oxygen-containing heteroaryl, 5-10 membered sulfur-containing heteroaryl, 5-6 membered nitrogen-containing heteroaryl, 5-6 membered oxygen-containing heteroaryl, 5-6 membered sulfur-containing heteroaryl, 5-6 membered nitrogen-containing monocyclic heteroaryl, 5-6 membered oxygen-containing monocyclic heteroaryl, 5-6 membered sulfur-containing monocyclic heteroaryl. The "nitrogen-containing heteroaryl", "oxygen-containing heteroaryl", "sulfur-containing heteroaryl", "nitrogen-containing monocyclic heteroaryl", "oxygen-containing monocyclic heteroaryl", "sulfur-containing monocyclic heteroaryl" optionally further contains one or more other heteroatoms selected from oxygen, nitrogen, sulfur. Examples thereof include, but are not limited to, thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl and the like, or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl and the like, and 5-to 10-membered heterocyclic groups containing these groups.
In the present invention, a heteroaryl group (e.g., a monocyclic heteroaryl group) may be substituted with an aryl group (e.g., a monocyclic aryl group such as phenyl), a heterocyclic group (e.g., a monocyclic heterocyclic group), a cycloalkyl group (e.g., a monocyclic cycloalkyl group),Heteroaryl (e.g., another monocyclic heteroaryl) shares two adjacent atoms with each other to form a fused ring structure, the point of attachment of which may be on either the heteroaryl ring or on other rings, including but not limited to (mono) heteroarylo (mono) heteroaryl, (mono) heteroarylo (monocyclic) aryl, (mono) heteroarylo (mono) heterocyclyl, and (mono) heteroarylo (mono) cycloalkyl, such as 5-6 membered (mono) heteroarylo 5-6 membered (mono) heteroaryl, 5-6 membered (mono) heteroarylo-phenyl, 5-6 membered (mono) heteroarylo 5-6 membered (mono) heterocyclyl, or 5-6 membered (mono) heteroarylo C4-6(Mono) cycloalkyl (e.g., 5-6 membered heteroarylbenzocyclobutyl, 5-6 membered heteroarylbenzocyclopentyl, 5-6 membered heteroarylbenzocyclohexyl), examples of which include, but are not limited to, indolyl, isoindolyl, indazolyl, benzimidazole, quinolinyl, isoquinolinyl, cyclohexyl, and the like,
Figure PCTCN2020077581-APPB-000026
Figure PCTCN2020077581-APPB-000027
And the like.
The term "aryloateroaryl" refers to a fused ring radical of an aryl (e.g., monocyclic aryl, such as phenyl) and a heteroaryl (e.g., monocyclic heteroaryl, such as 5-6 membered monocyclic heteroaryl), the point of attachment of which to other groups may be either on the aromatic ring or the heteroaromatic ring. The "aryloateroaryl" includes, but is not limited to, monocyclic aryl and monocyclic heteroaryl. The term "9-12 membered aryloateroaryl" refers to aryloateroaryl groups containing a total of 9-12 ring atoms, such as benzo 5-6 membered nitrogen containing monocyclic heteroaryl.
The term "heteroarylcycloalkylalkyl" refers to heteroaryl (e.g., monocyclic heteroaryl, such as 5-6 membered monocyclic heteroaryl) and cycloalkyl (e.g., C)4-6Cycloalkyl) groups, the point of attachment to other groups may be on the heteroaromatic ring or on the cycloalkyl group. The "heteroarylcycloalkyl" includes, but is not limited to, monocyclic heteroarylmonocyclic cycloalkyl. The term "9-10 membered heteroarylcycloalkylalkyl" refers to heteroarylcycloalkylalkyl groups containing a total of 9-10 ring atomsFor example, 4-6 membered nitrogen-containing monocyclic heteroaryl group C4-6A monocyclic cycloalkyl group.
The term "substituted" means that one or more (e.g., 1,2,3, or 4) hydrogens on the designated group is replaced with a selection from the indicated group, provided that the designated atom's normal valency at the present instance is not exceeded and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
If a substituent is described as "optionally substituted with … …," the substituent may be (1) unsubstituted or (2) substituted. If a carbon of a substituent is described as optionally substituted with one or more of the list of substituents, one or more hydrogens on the carbon (to the extent any hydrogen present) may each be replaced with an independently selected optional substituent. If the nitrogen of a substituent is described as being optionally substituted with one or more of the list of substituents, then one or more hydrogens on the nitrogen (to the extent any hydrogen is present) may each be replaced with an independently selected optional substituent.
If a substituent is described as "independently selected from … …," each substituent is selected independently of the other. Thus, each substituent may be the same as or different from another (other) substituent.
As used herein, the term "one or more" means 1 or more than 1, such as 2,3,4, 5,6, 7, 8, 9 or 10, under reasonable conditions.
Unless indicated, as used herein, the point of attachment of a substituent may be from any suitable position of the substituent.
The invention also includes all pharmaceutically acceptable isotopic compounds of the invention, which are identical to those of the compounds of the invention, except that one or more atoms are replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number which predominates in nature. Examples of isotopes suitable for inclusion in compounds of the invention include, but are not limited to, isotopes of hydrogen (e.g. hydrogen)2H、 3H);Isotopes of carbon (e.g. of11C、 13C and14C) (ii) a Isotopes of chlorine (e.g. of chlorine)36Cl); isotopes of fluorine (e.g. of fluorine)18F) (ii) a Isotopes of iodine (e.g. of iodine)123I and125I) (ii) a Isotopes of nitrogen (e.g. of13N and15n); isotopes of oxygen (e.g. of15O、 17O and18o); isotopes of phosphorus (e.g. of phosphorus)32P); and isotopes of sulfur (e.g. of35S). The term "stable isotopic derivative" refers to a stable compound formed by the replacement of one or more atoms in a compound of the present invention by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominant in nature.
The term "stereoisomer" refers to an isomer of a compound that is formed by the inclusion of at least one asymmetric center. In compounds having one or more (e.g., 1,2,3, or 4) asymmetric centers, they can result in racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Certain individual molecules may also exist as geometric isomers (cis/trans). The compounds of the invention may exist as a mixture of two or more different structural forms in rapid equilibrium (commonly referred to as tautomers). Representative examples of tautomers include keto-enol tautomers, phenol-keto tautomers, nitroso-oxime tautomers, imine-enamine tautomers, and the like. For example, a nitroso-oxime may exist in solution in equilibrium with the following tautomeric forms:
Figure PCTCN2020077581-APPB-000028
it is to be understood that the scope of this application encompasses all such isomers or mixtures thereof in any proportion (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%).
Unless otherwise indicated, the compounds of the present invention may exist as stereoisomers (which includes cis and trans isomers), optical isomers (e.g., R and S enantiomers), diastereomers, geometric isomers, rotamers, conformers, atropisomers, or mixtures thereof. The compounds of the present invention may exhibit more than one type of isomerization and consist of mixtures thereof (e.g., racemic mixtures and diastereomeric pairs).
The present invention encompasses all possible crystalline forms or polymorphs of the compounds of the present invention, which may be single polymorphs or mixtures of more than one polymorph in any ratio. It will also be appreciated that certain compounds of the invention may be present in free form for use in therapy or, where appropriate, in the form of a pharmaceutically acceptable derivative thereof. In the present invention, pharmaceutically acceptable derivatives include, but are not limited to: pharmaceutically acceptable salts, solvates, metabolites or prodrugs thereof, which upon administration to a patient in need thereof are capable of providing, directly or indirectly, a compound of the present invention or a metabolite or residue thereof. Thus, when reference is made herein to "a compound of the invention," it is also intended to encompass the various derivative forms of the compounds described above.
Pharmaceutically acceptable salts of the compounds of the present invention include pharmaceutically acceptable acid addition salts and base addition salts thereof. Such as hexafluorophosphate, meglumine salts and the like. For a review of suitable Salts, see Stahl and Wermuth, "Handbook of Pharmaceutical Salts: Properties, Selection, and Use" (Wiley-VCH, 2002).
By "pharmaceutically acceptable carrier" in the context of the present invention is meant a diluent, adjuvant, excipient, or vehicle that is administered together with a therapeutic agent and which is, within the scope of sound medical judgment, suitable for contact with the tissues of humans and/or other animals without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio.
Pharmaceutically acceptable carriers that may be employed in the pharmaceutical compositions of the present invention include, but are not limited to, sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is an exemplary carrier when the pharmaceutical composition is administered intravenously. Physiological saline and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, maltose, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol and the like. The composition may also optionally contain minor amounts of wetting agents, emulsifying agents, or pH buffering agents. Oral formulations may contain standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. Examples of suitable pharmaceutically acceptable carriers are described in Remington's Pharmaceutical Sciences (1990).
The pharmaceutical compositions of the present invention may act systemically and/or locally. For this purpose, they may be administered by a suitable route, for example by injection, intravenous, intraarterial, subcutaneous, intraperitoneal, intramuscular or transdermal administration; or by oral, buccal, nasal, transmucosal, topical, in the form of ophthalmic preparations or by inhalation.
For these routes of administration, the pharmaceutical compositions of the present invention may be administered in suitable dosage forms.
Such dosage forms include, but are not limited to, tablets, capsules, lozenges, hard candies, powders, sprays, creams, ointments, suppositories, gels, pastes, lotions, ointments, aqueous suspensions, injectable solutions, elixirs, syrups.
The term "effective amount" as used herein refers to an amount of a compound that, when administered, will alleviate one or more symptoms of the condition being treated to some extent.
The dosing regimen may be adjusted to provide the best desired response. For example, a single bolus may be administered, several divided doses may be administered over time, or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is noted that dosage values may vary with the type and severity of the condition being alleviated, and may include single or multiple doses. It is further understood that for any particular individual, the specific dosage regimen will be adjusted over time according to the individual need and the professional judgment of the person administering the composition or supervising the administration of the composition.
The amount of a compound of the invention administered will depend on the subject being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound, and the judgment of the prescribing physician. Generally, an effective dose is from about 0.0001 to about 50mg per kg body weight per day, e.g., from about 0.01 to about 10 mg/kg/day (single or divided administration). For a 70kg human, this may amount to about 0.007 mg/day to about 3500 mg/day, e.g., about 0.7 mg/day to about 700 mg/day. In some cases, dosage levels not higher than the lower limit of the aforesaid range may be sufficient, while in other cases still larger doses may be employed without causing any harmful side effects, provided that the larger dose is first divided into several smaller doses to be administered throughout the day.
The compound of the invention may be present in the pharmaceutical composition in an amount or amount of about 0.01mg to about 1000 mg.
As used herein, unless otherwise specified, the term "treating" means reversing, alleviating, inhibiting the progression of, or preventing the progression of, the disorder or condition to which the term applies, or one or more symptoms of such disorder or condition.
As used herein, "individual" includes a human or non-human animal. Exemplary human individuals include human individuals (referred to as patients) having a disease (e.g., a disease described herein) or normal individuals. "non-human animals" in the context of the present invention include all vertebrates, such as non-mammals (e.g., birds, amphibians, reptiles) and mammals, such as non-human primates, livestock and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
The compounds of the invention may be present in the form of solvates, preferably hydrates, wherein the compounds of the invention comprise as structural element of the crystal lattice of the compound a polar solvent, such as in particular water, methanol or ethanol. The amount of polar solvent, particularly water, may be present in stoichiometric or non-stoichiometric proportions.
Also included within the scope of the present invention are metabolites of the compounds of the present invention, i.e., substances formed in vivo upon administration of the compounds of the present invention. Such products may result, for example, from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic hydrolysis, etc. of the administered compound. Accordingly, the present invention includes metabolites of the compounds of the present invention, including compounds obtained by the process of contacting the compounds of the present invention with a mammal for a time sufficient to produce a metabolite thereof.
The present invention further includes within its scope prodrugs of the compounds of the present invention which are certain derivatives of the compounds of the present invention which may themselves have little or no pharmacological activity which, when administered into or onto the body, may be converted to the compounds of the present invention having the desired activity by, for example, hydrolytic cleavage. Typically such prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the desired therapeutically active compound. Further information on the use of prodrugs can be found in "Pro-drugs as Novel Delivery Systems", volume 14, ACS Symposium Series (T.Higuchi and V.Stella) and "Bioreversible Carriers in Drug Design," Pergamon Press,1987(E.B.Roche editions, American Pharmaceutical Association). Prodrugs of the invention may be prepared, for example, by substituting certain moieties known to those skilled in the art as "pro-moieties" (e.g., "Design of Prodrugs", described in h. bundgaard (Elsevier, 1985)) for appropriate functional groups present in compounds of the invention.
The invention also encompasses compounds of the invention containing a protecting group. In any process for preparing the compounds of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned, thereby forming a chemically protected form of the compounds of the present invention. This can be achieved by conventional protecting Groups, for example, as described in Protective Groups in Organic Chemistry, ed.j.f.w.mcomie, Plenum Press, 1973; and T.W.Greene & P.G.M.Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons,1991, which are incorporated herein by reference. The protecting group may be removed at a suitable subsequent stage using methods known in the art.
Preparation method
Synthesis of Compounds of formula I-a
Figure PCTCN2020077581-APPB-000029
Ring A, ring B, Ra、R b、R cM, n, p, q, s, t are as defined above for formula I;
the first step is as follows: the compound I-1 and the compound I-2 are subjected to reductive amination reaction to generate an intermediate I-3.
The solvent is methanol, ethanol, acetic acid, THF, 1, 2-dichloroethane or DCM, etc., and the reducing agent is sodium borohydride, sodium cyanoborohydride, sodium triacetoxyborohydride, etc., at 0-80 deg.C.
The second step is that: deprotection of compound I-3 under acidic conditions produces intermediate I-4.
The solvent is THF, CH3CN, DCM or dioxane, etc., the acid used is HCl or TFA, etc., and the temperature is 0 ℃ to 110 ℃.
The third step: the compound I-4 and the compound I-5 or the acyl chloride of the compound I-5 are subjected to condensation reaction to generate an intermediate I-6.
The solvent is DMF, DCM, 1, 2-dichloroethane or THF, the condensing agent is HATU, HBTU, TBTU, DCC/HOBt, EDCI/HOBt, etc., and the base is TEA, DIPEA, Cs2CO 3、Na 2CO 3、AcOK、NaHCO 3Or K2CO 3Etc. at a temperature of 0 ℃ to 60 ℃.
The fourth step: the compound I-6 reacts with R under the action of alkalic-H is substituted or subjected to a coupling reaction (e.g., Buchwald-Hartwig reaction) to produce compound I-a.
Under non-catalytic conditions, the bases used are t-Buona, t-BuOK, t-BuOLi, Cs2CO 3、LiHMDS、LDA、NaHMDS、KHMDS、K 3PO 4、Na 2CO 3、AcOK、NaHCO 3Or K2CO 3And the like, wherein the solvent is methanol, ethanol, iPrOH, toluene, xylene, THF, DME, dioxane, DMF, DMSO or NMP and the like, and the temperature is 20-140 ℃;
the catalyst used in the coupling reaction under the catalytic condition is Pd (OAc)2、Pd 2(dba) 3、Pd(dba) 2、PdCl 2、Pd(PPh 3) 4、Pd(dppf)Cl 2、Pd(acac) 2Or Pd (allyl)2Etc., the ligand used is PPh3XPhos, SPhos, RuPhos, XantPhos, Dppf, BINOL, BINAP or Pcy3Etc., the alkali is t-Buona, t-BuOK, t-Buoli, Cs2CO 3、LiHMDS、LDA、NaHMDS、KHMDS、K 3PO 4、Na 2CO 3、 AcOK、NaHCO 3Or K2CO 3And the like, and the solvent is toluene, xylene, THF, DME, dioxane, DMF, DMSO, NMP or the like, and the temperature is 60 ℃ to 140 ℃.
Synthesis of Compounds of formula I-b
Figure PCTCN2020077581-APPB-000030
Ring A, Ra、R b、R cM, n, p, q, t are as defined above for formula I;
the first step is as follows: the compound II-1 and the compound II-2 are subjected to ring closure under the action of alkali to generate an intermediate II-3.
The solvent is water, the alkali is sodium hydroxide, potassium hydroxide, etc. and the temperature is 0-100 deg.c. The second step is that: the compound II-3 generates an intermediate II-4 under the action of a chlorinating agent.
The solvent is DCM, chloroform or 1, 2-dichloroethane, the chlorinating agent is phosphorus oxychloride, thionyl chloride and the like, and the temperature is 25-110 ℃.
The third step: the compound II-4 and the compound I-4 are condensed under the action of alkali to generate an intermediate I-b.
The solvent is DCM, chloroform, 1, 2-dichloroethane, DMF, etc., and the base is TEA, DIPEA, Cs2CO 3、Na 2CO 3、AcOK、NaHCO 3Or K2CO 3Etc. at a temperature of 0 ℃ to 60 ℃.
Advantageous effects
The compound of the invention has obvious inhibitory activity to PRMT5 and a signal channel thereof, has no obvious toxic or side effect, and can be used for treating diseases (such as tumors) related to the activity of PRMT 5.
Detailed Description
The invention is further described below in connection with examples, which are not intended to limit the scope of the invention.
In this application, when chemical names and structural formulae are inconsistent, the structural formulae should be taken as a control unless the context suggests that the chemical name and not the structural formula is correct.
The abbreviations in the present invention have the following meanings:
Figure PCTCN2020077581-APPB-000031
Figure PCTCN2020077581-APPB-000032
the structure of the compound of the invention is determined by nuclear magnetic resonance1HNMR) and/or Mass Spectrometry (MS) identification.
1H NMR chemical shifts (. delta.) in parts per million (ppm)ppm) was recorded.1H NMR was measured by JEOL Eclipse400 Nuclear magnetic Analyzer using deuterated methanol (CD)3OD), deuterated chloroform (CDCl)3) Hexadeuterio dimethyl sulfoxide (DMSO-d)6) Internal standard is Tetramethylsilane (TMS). s: singlet, d: doublet, t: triplet, q: quartet, dd: doublet, qd: quartet, ddd: double doublet, ddt: double triplet, dddd: double doublet, m: multiplet, br: broad (broad), J: coupling constant, Hz: hertz's scale
MS was determined using an Agilent 6120B mass spectrometer.
The reaction was monitored by Thin Layer Chromatography (TLC) or LC-MS.
LC-MS instrument: agilent 6125B.
The compound can be separated and purified by a silica gel thick preparation plate for chromatography, silica gel column chromatography, preparative high performance liquid chromatography (Prep-HPLC) and Flash column chromatography (Flash column chromatography).
The chromatography silica gel thick preparation plate adopts a cigarette platform yellow sea HSGF254 type preparation plate.
Column chromatography generally uses Qingdao ocean silica gel of 200-300 meshes as a carrier. The system of eluents comprises: a: dichloromethane and methanol; b: petroleum ether and ethyl acetate, the volume ratio of the solvent is adjusted according to the polarity of the compound.
Preparative high performance liquid chromatography (Prep-HPLC) using Agilent 1260 chromatography.
Flash column chromatography used an Agela medium pressure rapid purification preparation system (MP-200).
The microwave reaction used a BiotageInitiator + (400W, RT-300 ℃ C.) microwave reactor.
In the examples, the reaction temperature is, unless otherwise specified, room temperature (20 ℃ C. to 30 ℃ C.).
Reagents used in this application were purchased from Acros Organics, Aldrich Chemical Company, or Tereber Chemical, among others.
Example 1: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyrimidin-4-yl) amino) piperidin-1-yl) ethanone (Compound 1)
Figure PCTCN2020077581-APPB-000033
The first step is as follows: 4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carboxylic acid tert-butyl ester (1c)
Compound 1a (1.80g,9.01mmol) and compound 1b (1.00g, 7.51mmol) were added sequentially to a mixed solution of dichloromethane (20mL) and glacial acetic acid (20mL), stirred at room temperature for 30min, sodium triacetoxyborocyanide (3.18g,15.02mmol) was added portionwise to the reaction system, and stirring at room temperature was continued for 3 h. The reaction was quenched with saturated aqueous ammonium chloride, extracted with dichloromethane, the organic phase was washed with saturated aqueous NaCl, extracted, the organic phase was concentrated and the residue was purified by flash column chromatography (eluent system B) to give compound 1c (600 mg). MS (ESI, M/z) 317.3[ M + H ]] +
The second step is that: 2- (piperidin-4-yl) -1,2,3, 4-tetrahydroisoquinoline (1d)
Compound 1c (360mg, 1.14mmol) was added to a mixed solution of concentrated hydrochloric acid (5mL) and tetrahydrofuran (5mL) and stirred at room temperature for 2 h. The solvent was concentrated under reduced pressure and the residue was taken without further purification to give the hydrochloride salt of compound 1d (286 mg). MS (ESI, M/z):217.2[ M + H] +
The third step: (6-Chloropyrimidin-4-yl) (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (1f)
The hydrochloride salt of compound 1d (286mg,1.11mmol), 1e (160mg, 1.01mmol), DIPEA (522mg, 4.04mmol) and HATU (383mg, 1.01mmol) were added to DMF (4mL) and stirred at room temperature for 2 h. The reaction mixture was poured into water, extracted with ethyl acetate, the organic phase was concentrated, and the residue was subjected to flash column chromatography (eluent system a) to give compound 1f (220 mg). MS (ESI, M/z):357.2[ M + H] +
The fourth step: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyrimidin-4-yl) amino) piperidin-1-yl) ethanone (1)
Compound 1f (30mg, 84.07. mu. mol),1g (30mg, 168.14. mu. mol) and TEA (60mg, 588.49. mu. mol) were added to iPrOH (1.5mL) and heated to 90 ℃ and stirred for 2 h. The solvent was concentrated and the residue was subjected to preparative high performance liquid chromatography to give compound 1(21 mg).
MS(ESI,m/z):463.3[M+H] +.
1H NMR(CDCl 3,400MHz)δ8.54(s,1H),7.13-7.11(m,3H),7.04-7.02(m,1H),6.61(s,1H),5.32(t,J=6.8Hz,1H),4.75(d,J=12.8Hz,1H),4.55(d,J=14Hz,1H),4.08(d,J=13.2Hz,1H),3.95-3.79(m,3H),3.22-3.18(m,1H),3.16-3.07(m,1H),2.86-2.79(m,7H),2.12(s,3H),2.11-1.95(m,4H),1.81-1.70(m,2H),1.47-1.36(m,2H).
Example 2: (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) (6- (oxetan-3-ylamino) pyrimidin-4-yl) methanone (Compound 2)
Figure PCTCN2020077581-APPB-000034
Compound 1f (50mg, 140.12. mu. mol), 2a (20mg, 280.23. mu. mol) and TEA (43mg, 420.35. mu. mol) were added to iPrOH (2mL), heated to 90 ℃ and stirred for 2 h. The solvent was concentrated, and the residue was separated by preparative high performance liquid chromatography to give compound 2(35 mg).
MS(ESI,m/z):394.2[M+H] +.
1H NMR(CDCl 3,400MHz)δ8.60(s,1H),7.20-7.12(m,3H),7.08-7.04(m,1H),6.67(s,1H),5.92(d,J=6.4Hz,1H),5.17-5.07(m,3H),4.82-4.75(m,1H),4.62(t,J=6.0Hz,2H),4.12-4.05(m,1H),3.94-3.84(m,2H),3.20-3.10(m,1H),3.01-2.84(m,6H),2.15-2.08(m,1H),2.03-1.96(m,1H),1.83-1.70(m,2H).
Example 3: (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) (6- ((tetrahydrofuran-3-yl) amino) pyrimidin-4-yl) methanone (Compound 3)
Figure PCTCN2020077581-APPB-000035
Compound 1f (30mg, 84.07. mu. mol), 3a (21mg, 168.14. mu. mol) and TEA (60mg, 588.49. mu. mol) were added to iPrOH (1.5mL) and heated to 90 ℃ with stirring for 2 h. The reaction solution was concentrated, and the residue was separated by preparative high performance liquid chromatography to give compound 3(21 mg).
MS(ESI,m/z):408.3[M+H] +.
1H NMR(CDCl 3,400MHz)δ8.57(s,1H),7.16-7.09(m,3H),7.06-7.02(m,1H),6.61(s,1H),5.46(d,J=5.2Hz,1H),4.75(d,J=12.8Hz,1H),4.55(s,1H),4.07(d,J=13.6Hz,1H),4.02-3.83(m,5H),3.77-3.72(m,1H),3.15-3.05(m,1H),3.01-2.77(m,6H),2.39-2.28(m,1H),2.13-1.85(m,3H),1.80-1.67(m,2H).
Example 4: 1- (3- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyrimidin-4-yl) amino) azetidin-1-yl) ethanone (compound 4)
Figure PCTCN2020077581-APPB-000036
Compound 1f (100mg, 280.23. mu. mol), 4a (64mg, 424.48. mu. mol) and TEA (142mg,1.4mmol) were added to iPrOH (5mL) and heated to 90 ℃ and stirred for 2 h. The solvent was concentrated, and the residue was separated by preparative high performance liquid chromatography to give compound 4(45 mg).
MS(ESI,m/z):435.2[M+H] +.
1H NMR(CDCl 3,400MHz)δ8.55(s,1H),7.17-7.08(m,3H),7.06-6.99(m,1H),6.69(s,1H),4.72-4.70(m,2H),4.48(t,J=8.0Hz,1H),4.41-4.33(m,1H),4.05-4.02(m,1H),3.98-3.88(m,2H),3.84(s,2H),3.17-3.05(m,1H),2.96-2.78(m,6H),2.08-2.05(m,1H),1.98-1.85(m,4H),1.77-1.68(m,2H).
Example 5: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyrimidin-4-yl) amino) piperidin-1-yl) -2-methylpropan-1-one (Compound 5)
Figure PCTCN2020077581-APPB-000037
Compound 1f (300mg, 840.69. mu. mol), 5a (286mg, 1.68mmol) and TEA (425mg,4.2mmol) were added to iPrOH (5mL) and heated to 90 ℃ and stirred for 2 h. The solvent was concentrated, and the residue was separated by preparative high performance liquid chromatography to give compound 5(100 mg).
MS(ESI,m/z):491.2[M+H] +.
1H NMR(CDCl 3,400MHz)δ8.54(s,1H),7.13-7.11(m,3H),7.06-7.00(m,1H),6.60(s,1H),5.36(d,J=7.0Hz,1H),4.73(d,J=13.5Hz,1H),4.58(d,J=12.9Hz,1H),4.07(d,J=13.6Hz,1H),3.94(d,J=13.4Hz,1H),3.85(s,2H),3.21-3.09(m,2H),2.92-2.75(m,8H),2.21-2.07(m,3H),1.96-1.93(m,1H),1.73-1.70(m,2H),1.48-1.35(m,2H),1.14(d,J=6.1Hz,6H).
Example 6: (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) (6- ((pyridin-2-ylmethyl) amino) pyrimidin-4-yl) methanone (Compound 6)
Figure PCTCN2020077581-APPB-000038
Compound 1f (100mg, 280.23. mu. mol), 6a (61mg, 560.46. mu. mol) and TEA (142mg,1.4mmol) were added to iPrOH (5mL) and heated to 90 ℃ and stirred for 2 h. The solvent was concentrated, and the residue was separated by preparative high performance liquid chromatography to give compound 6(75 mg).
MS(ESI,m/z):429.2[M+H] +.
1H NMR(CDCl 3,400MHz)δ8.61(s,1H),8.57(d,J=4.8Hz,1H),7.69(td,J=8.0,2.0Hz,1H),7.29(d,J=8.0Hz,1H),7.24-7.21(m,1H),7.13-7.08(m,3H),7.05-7.01(m,1H),6.69(s,1H),6.49(s,1H),4.77-4.71(m,3H),4.03-4.00(m,1H),3.81(s,2H),3.13-3.05(m,1H),2.92-2.75(m,6H),2.05-2.00(m,1H),1.92-1.88(m,1H),1.75-1.65(m,2H).
Example 7: (S) -1- (3- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyrimidin-4-yl) amino) pyrrolidin-1-yl) ethanone (Compound 7)
Figure PCTCN2020077581-APPB-000039
Compound 1f (0.1g, 280.23. mu. mol), 7a (33.56mg, 261.80. mu. mol) and DIPEA (90.54mg, 700.58. mu. mol) were added to iPrOH (2mL) and stirred with microwave heating to 90 ℃ for 2 h. The solvent was concentrated and the residue was subjected to preparative high performance liquid chromatography to give compound 7(20 mg).
MS(ESI,m/z):449.3[M+H] +.
1H NMR(CDCl 3,400MHz)δ8.57(s,0.5H),8.56(s,0.5H)7.15-7.09(m,3H),7.04-7.02(m,1H),6.66(s,0.5H),6.64(s,0.5H),5.98-5.88(br,1H),4.72(d,J=13.6Hz,1H),4.60-4.50(br,1H),4.04(d,J=10.0Hz 1H),3.88-3.77(m,3H),3.65-3.50(m,2.5H),3.38-3.33(m,0.5H),3.14-3.08(m,1H),2.95-2.81(m,6H),2.33-2.21(m,1H),2.08-2.04(m,4H),1.97-1.91(m,2H),1.77-1.70(m,2H).
Example 8: (R) -1- (3- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyrimidin-4-yl) amino) pyrrolidin-1-yl) ethanone (Compound 8)
Figure PCTCN2020077581-APPB-000040
Compound 1f (0.1g, 280.23. mu. mol), 8a (55.36mg, 336.28. mu. mol) and DIPEA (90.54mg, 700.58. mu. mol) were added to iPrOH (2mL) and reacted for 2h with microwave heating to 90 ℃. The solvent was concentrated and the residue was subjected to preparative high performance liquid chromatography to give compound 8(20 mg).
MS(ESI,m/z):449.3[M+H] +
1H NMR(CDCl 3,400MHz)δ8.57(s,0.5H),8.56(s,0.5H)7.15-7.09(m,3H),7.04-7.02(m,1H),6.67(s,0.5H),6.65(s,0.5H),5.98-5.88(br,1H),4.73(d,J=13.2Hz,1H),4.60-4.50(br,1H),4.04(d,J=13.6Hz 1H),3.88-3.77(m,3H),3.65-3.50(m,2.5H),3.38-3.34(m,0.5H),3.14-3.07(m,1H),2.95-2.81(m,6H),2.37-2.20(m,1H),2.08-2.04(m,4H),1.97-1.91(m,2H),1.77-1.70(m,2H).
Example 9: (6- ((cyclopropylmethyl) amino) pyrimidin-4-yl) (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (Compound 9)
Figure PCTCN2020077581-APPB-000041
Compound 1f (0.1g, 280.23. mu. mol), 9a (25.91mg, 364.30. mu. mol) and DIPEA (90.54mg, 700.58. mu. mol) were added to iPrOH (2mL) and reacted for 2h with microwave heating to 90 ℃. The solvent was concentrated and the residue was subjected to preparative high performance liquid chromatography to give compound 9(70 mg).
MS(ESI,m/z):392.2[M+H] +
1H NMR(CDCl 3,400MHz)δ8.53(s,1H)7.14-7.08(m,3H),7.03-7.00(m,1H),6.57(s,1H),5.33(s,1H),4.76-4.72(m,1H),4.06-4.02(m,1H),3.85-3.77(m,2H),3.19-3.06(m,3H),2.89-2.76(m,6H),2.05-2.00(m,1H),1.93-1.90(m,1H),1.76-1.63(m,2H),1.11-1.04(m,1H),0.66-0.56(m,2H),0.28-0.25(m,2H).
Example 10: (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) (6- ((pyridin-3-methyl) amino) pyrimidin-4-yl) methanone (Compound 10)
Figure PCTCN2020077581-APPB-000042
Compound 1f (100mg, 280.23. mu. mol), 10a (45.46mg, 420.35. mu. mol) and TEA (198.50mg,1.96mmol) were added to iPrOH (5mL) and heated to 90 ℃ and stirred for 2 h. The solvent was concentrated, and the residue was separated by preparative high performance liquid chromatography to give compound 10(80 mg).
MS(ESI,m/z):429.2[M+H] +.
1H NMR(400MHz,CDCl 3)δ8.59(d,J=1.8Hz,1H),8.57(s,1H),8.53(d,J=4.7Hz,1H),7.65(d,J=7.9Hz,1H),7.25(s,1H),7.15-7.08(m,3H),7.04-7.00(m,1H),6.67(s,1H),6.12(s,1H),4.70-4.62(m,3H),4.03(d,J=13.5Hz,1H),3.84(s,2H),3.13-3.04(m,1H),2.96-2.89(m,4H),2.84-2.76(m,2H),2.04(d,J=12.8Hz,1H),1.93(d,J=12.8Hz,1H),1.75-1.65(m,2H).
Example 11: (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) (6- ((1- (methylsulfonyl) piperidin-4-yl) amino) pyrimidin-4-yl) methanone (Compound 11)
Figure PCTCN2020077581-APPB-000043
Compound 1f (100mg, 280.23. mu. mol), 11a (74.93mg, 420.35. mu. mol) and TEA (198.50mg,1.96mmol) were added to iPrOH (5mL) and heated to 90 ℃ and stirred for 2 h. The solvent was concentrated, and the residue was separated by preparative high performance liquid chromatography to give compound 11(30 mg).
MS(ESI,m/z):499.2[M+H] +.
1H NMR(400MHz,CDCl 3)δ8.52(s,1H),7.15-7.08(m,3H),7.05-7.00(m,1H),6.62(s,1H),5.91(s,1H),4.71(d,J=13.1Hz,1H),4.02(d,J=13.0Hz,1H),3.82-3.73(m,4H),3.11(t,J=11.7Hz,1H),2.94-2.75(m,12H),2.07-2.04(m,3H),1.93-1.90(m,1H),1.74-1.58(m,4H).
Example 12: (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) (6- ((pyridin-4-ylmethyl) amino) pyrimidin-4-yl) methanone (Compound 12)
Figure PCTCN2020077581-APPB-000044
Compound 1f (100mg, 280.23. mu. mol), 12a (45.46mg, 420.35. mu. mol) and TEA (198.50mg,1.96mmol) were added to iPrOH (5mL) and heated to 90 ℃ and stirred for 2 h. The solvent was concentrated, and the residue was separated by preparative high performance liquid chromatography to give compound 12(60 mg).
MS(ESI,m/z):429.2[M+H] +.
1H NMR(400MHz,CDCl 3)δ8.55(s,1H),8.53(d,J=4.8Hz,2H),7.19(d,J=4.9Hz,2H),7.15-7.07(m,3H),7.05-6.99(m,1H),6.70(s,1H),6.41(s,1H),4.64(t,J=11.1Hz,3H),4.02(d,J=13.1Hz,1H),3.85-3.75(m,2H),3.09(t,J=11.7Hz,1H),2.89-2.77(m,6H),2.01(d,J=12.0Hz,1H),1.90(d,J=12.0Hz,1H),1.74-1.59(m,2H).
Example 13: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) -2-methylpyrimidin-4-yl) amino) piperidin-1-yl) ethanone (Compound 13)
Figure PCTCN2020077581-APPB-000045
The first step is as follows: 6-hydroxy-2-methylpyrimidine-4-carboxylic acid (13c)
13a (5g,23.79mmol) was dissolved in water (16mL), aqueous NaOH (3.8mL,6.25mol/L) was added dropwise at room temperature, stirring was carried out at 25 ℃ for 1 hour, 13b (4.95g,52.34mmol) was dissolved in water (5mL) and then added to the reaction system, pH was adjusted to 11 with aqueous NaOH (6.25mol/L) under ice bath, stirring was continued at that temperature for 40min, pH was adjusted to 1 with aqueous hydrochloric acid (12mol/L), suction filtration was carried out under reduced pressure, the cake was washed with aqueous hydrochloric acid (2 × 10mL,0.1mol/L), and drying was carried out to obtain compound 13c (1.8g) MS (ESI, M/z):155.2[ M + H/z): 155.2] +.
The second step is that: 6-chloro-2-methylpyrimidine-4-carbonyl chloride (13d)
13c (500mg,3.24mmol) was added to POCl3(3mL), warm to 110 ℃ and stir for 30min until all the solids are dissolved, cool to room temperature, concentrate the solvent under reduced pressure, and use the residue directly in the next reaction.
The third step: (6-chloro-2-methylpyrimidin-4-yl) - (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (13e)
Adding 1d (794mg,2.75mmol) and DIPEA (3.25g,25.13mmol) into dichloromethane (10mL), dissolving 13d (600mg,3.14mmol) in dichloromethane (5mL), dropping into the reaction system, stirring at 25 deg.C for 1H, adding water (20mL) into the reaction system, extracting, separating, concentrating the organic phase, purifying the residue by flash column chromatography (eluent system A) to obtain compound 13e (300mg) MS (ESI, M/z):371.2[ M + H ] M] +.
The fourth step: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) -2-methylpyrimidin-4-yl) amino) piperidin-1-yl) ethanone (13)
Compound 13e (100mg, 269.63. mu. mol),1g (96mg, 539.27. mu. mol) and TEA (191mg,1.89mmol) were added to iPrOH (1.5mL) and heated to 90 ℃ and stirred for 2 h. The solvent was concentrated and the residue was subjected to preparative high performance liquid chromatography to give compound 13(40 mg).
MS(ESI,m/z):477.2[M+H] +.
1H NMR(CDCl 3,400MHz)δ7.16-7.07(m,3H),7.05-6.99(m,1H),6.37(s, 1H),5.34(s,1H),4.72(d,J=13.2Hz,1H),4.51(d,J=13.7Hz,1H),4.03-3.96(m,2H),3.83-3.77(m,3H),3.29-3.17(m,1H),3.14-3.09(m,1H),2.97-2.72(m,7H),2.50(s,3H),2.11(s,3H),2.09-1.96(m,3H),1.95-1.84(m,1H),1.76-1.59(m,2H),1.48-1.33(m,2H).
Example 14: (6- (Cyclobutylamino) -2-methylpyrimidin-4-yl) (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (Compound 14)
Figure PCTCN2020077581-APPB-000046
Compound 13e (60mg, 161.78. mu. mol) and 14a (57mg, 808.90. mu. mol) were added to iPrOH (2mL) and heated to 90 ℃ with stirring for 3 h. The solvent was concentrated and the residue was isolated by preparative high performance liquid chromatography to give compound 14(15 mg).
MS(ESI,m/z):406.3[M+H] +.
1H NMR(CDCl 3,400MHz)δ7.24-7.14(m,3H),7.09-7.07(m,1H),6.33(s,1H),5.38(s,1H),4.85-4.81(d,J=13.6Hz,1H),5.17-5.07(m,3H),4.11(m,1H),3.20-3.06(m,6H),2.82-2.76(m,1H),2.51-2.40(m,5H),2.19(m,2H),1.94-1.78(m,6H).
Example 15: 1- (4- ((3- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) phenyl) amino) piperidin-1-yl) ethanone (Compound 15)
Figure PCTCN2020077581-APPB-000047
The first step is as follows: 3-bromobenzoyl chloride (15b)
Compound 15a (0.5g,2.49mmol), oxalyl chloride (379.07mg,2.98mmol) were added to dichloromethane (10mL), DMF (0.1mL) was added dropwise to the reaction mixture, stirred at room temperature for 2h, the solvent was concentrated, and the residue was directly taken to the next reaction.
The second step is that: (3-bromophenyl) (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (15c)
Hydrochloride (630mg, 2.49mmol) of compound 1d and DIPEA (963mg, 7.47mmol) were added to dichloromethane (5mL), dichloromethane (5mL) containing compound 15b (2.49mmol) was added dropwise to the reaction solution, stirred at room temperature for 2h, water (10mL) was added to the reaction system, extraction was performed, the organic layer was concentrated under reduced pressure, and the residue was purified by flash column chromatography (eluent system a) to obtain compound 15c (500 mg). MS (ESI, M/z) 399.1[ M + H ]] +
The third step: 1- (4- ((3- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) phenyl) amino) piperidin-1-yl) ethanone (15)
Compound 15c (150.00mg, 375.64. mu. mol), Ruphos (35.01mg, 75.13. mu. mol),1g (100.67mg, 563.45. mu. mol), Pd2(dba) 3(34.37mg, 37.56. mu. mol) and sodium tert-butoxide (108.30mg,1.13mmol) were added successively to toluene (10mL) and stirred at 90 ℃ for 6 hours. The reaction solution was cooled to room temperature, filtered, washed with water, extracted with ethyl acetate, the organic layer was concentrated, and the residue was purified using a preparative TLC plate (developing solvent dichloromethane: methanol 10:1) to give compound 15(5 mg).
MS(ESI,m/z):461.3[M+H] +.
1H NMR(CDCl 3,400MHz)δ7.18-7.10(m,4H),7.06-7.03(m,1H),6.69-6.61(m,3H),4.79(s,1H),4.52-4.48(d,J=13.6Hz,1H),3.91-3.79(m,4H),3.55-3.50(m,1H),3.22-3.17(m,1H),2.98-2.81(m,8H),2.14-2.05(m,6H),1.36-1.25(m,6H).
Example 16: 1- (4- ((2- (4- (3, 4-dihydroisoquinolin-2- (1H) -yl) piperidine-1-carbonyl) pyridin-4-yl) amino) piperidin-1-yl) ethanone (Compound 16)
Figure PCTCN2020077581-APPB-000048
The first step is as follows: 4-Bromopyridinecarboxylic acid chloride (16b)
Compound 16a (0.65g,3.22mmol), oxalyl chloride (490.38mg,3.86mmol) and DMF (0.1mL) were added to dichloromethane (10mL), stirred at room temperature for 2h, the solvent was concentrated and the residue was directly taken to the next reaction.
The second step is that: (4-Bromopyridin-2-yl) (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (16c)
The hydrochloride of compound 1d (811mg, 3.22mmol) and DIPEA (1.25g, 9.66mmol) were added to dichloromethane (10mL), a dichloromethane solution (5mL) containing 16b (3.22mmol) was added dropwise at room temperature, stirred at room temperature for 2h, washed with water, extracted, the organic layer was concentrated, and the residue was purified by flash column chromatography (eluent system a) to give compound 16c (600 mg). MS (ESI, M/z) 400.1[ M + H] +
The third step: 1- (4- ((2- (4- (3, 4-dihydroisoquinolin-2- (1H) -yl) piperidine-1-carbonyl) pyridin-4-yl) amino) piperidin-1-yl) ethanone (16)
Compound 16c (300mg, 749.42. mu. mol), Ruphos (69.85mg, 149.88. mu. mol),1g (200.84mg,1.12mmol), Pd2(dba) 3(68.57mg, 74.94. mu. mol) and sodium tert-butoxide (216.06mg,2.25mmol) were added successively to toluene (5mL) and reacted at 90 ℃ for 6 hours. The reaction solution was cooled to room temperature, filtered, washed with water, extracted, and the organic layer was concentrated, and the residue was purified using a preparative TLC plate (dichloromethane: methanol 10:1 as a developing solvent) to give compound 16(20 mg).
MS(ESI,m/z):462.3[M+H] +.
1H NMR(CDCl 3,400MHz)δ8.16-8.14(m,1H),7.13-7.09(m,3H),7.04-7.00(m,1H),6.78-6.77(m,1H),6.46-6.44(m,1H),4.81-4.78(m,1H),4.54-4.51(m,1H),4.40-4.39(m,1H),4.14-4.11(m,1H),3.89-3.81(m,3H),3.60-3.58(m,1H),3.24-3.17(m,1H),3.13-3.07(m,1H),2.96-2.79(m,6H),2.19-1.93(m,7H),1.76-1.74(m,2H),1.44-1.37(m,2H).
Example 17: 1- (4- ((4- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyridin-2-yl) amino) piperidin-1-yl) ethanone (Compound 17)
Figure PCTCN2020077581-APPB-000049
The first step is as follows: 2-Bromoiisonicotinoyl chloride (17b)
Compound 17a (0.5g,2.48mmol), oxalyl chloride (377.22mg,2.97mmol) were added to dichloromethane (10mL), DMF (0.1mL) was added dropwise to the reaction mixture, stirred at room temperature for 2h, and the residue was directly taken to the next reaction.
The second step is that: (2-Bromopyridin-4-yl) (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (17c)
The hydrochloride salt of compound 1d (625mg, 2.48mmol), DIPEA (1.1g, 8.68mmol) were added dropwise to dichloromethane (10mL), a solution of compound 17b (2.48mmol) in dichloromethane (5mL) was added dropwise at room temperature, stirred at room temperature for 2h, washed with water, extracted, the organic layer concentrated and the residue purified by flash column chromatography (eluent system a) to give compound 17c (300 mg). MS (ESI, M/z) 400.1[ M + H] +
The third step: 1- (4- ((4- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyridin-2-yl) amino) piperidin-1-yl) ethanone (17)
Compound 17c (0.2g, 499.61. mu. mol), Ruphos (46.56mg, 99.92. mu. mol),1g (133.89mg, 749.42. mu. mol), Pd2(dba) 3(45.71mg, 49.96. mu. mol) and sodium tert-butoxide (144.04mg,1.50mmol) were added successively to toluene (5mL) and stirred at 90 ℃ for 6 h. The reaction solution was cooled to room temperature, filtered, washed with water, extracted, and the organic layer was concentrated, and the residue was purified using a preparative TLC plate (dichloromethane: methanol 10:1 as a developing solvent) to give compound 17(10 mg). MS (ESI, M/z) 462.3[ M + H ]] +.
1H NMR(CDCl 3,400MHz)δ8.05(s,1H),7.08-7.03(m,3H),6.97-6.95(m,1H),6.46-6.44(m,1H),6.31(s,1H),4.69-4.57(m,2H),4.45-4.41(m,1H),3.87-3.72(m,5H),3.19-3.12(m,1H),3.01-2.94(m,1H),2.88-2.76(m,6H),2.08-1.88(m,7H),1.66-1.54(m,2H),1.36-1.27(m,2H).
Example 18: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyrimidin-4-yl) oxy) piperidin-1-yl) ethanone (compound 18)
Figure PCTCN2020077581-APPB-000050
18a (642mg,4.48mmol), sodium tert-butoxide (539mg,5.6mmol) was added to THF (15mL) and stirred at 25 ℃ for 1h, 1f (800mg, 2.24mmol) was added and stirred at 25 ℃ for 5h, the solvent was concentrated and the residue was purified by flash column chromatography (eluent system A) to give compound 18(180 mg).
MS(ESI,m/z):464.3[M+H] +.
1H NMR(CDCl 3,400MHz)δ8.75(s,1H),7.15-7.09(m,3H),7.04-7.02(m,1H),6.95(s,1H),5.44-5.40(m,1H),4.77(d,J=13.4Hz,1H),3.99-3.86(m,4H),3.70-3.68(m,1H),3.56-3.48(m,1H),3.46-3.37(m,1H),3.13-3.01(m,1H), 2.94-2.84(m,6H),2.13(s,3H),2.11-1.92(m,4H),1.87-1.69(m,4H).
Example 19: (6- ((1- (cyclopropylcarbonyl) piperidin-4-yl) amino) pyrimidin-4-yl) (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (Compound 19)
Figure PCTCN2020077581-APPB-000051
The first step is as follows: tert-butyl 4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyrimidin-4-yl) amino) piperidine-1-carboxylate (19b)
Compound 1f (300mg, 840.69. mu. mol), 19a (337mg, 1.68mmol) and TEA (595.48mg,5.88mmol) were added to iPrOH (10mL) and heated to 90 ℃ and stirred for 2 h. The solvent was concentrated and the residue was purified by flash column chromatography (eluent system a) to give compound 19b (430 mg). MS (ESI, M/z):521.3[ M + H] +
The second step is that: (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) (6- (piperidin-4-ylamino) pyrimidin-4-yl) methanone (19c)
Compound 19b (430mg, 825.87. mu. mol) was added to a solution of dioxane hydrochloride (4mol/L,10mL) and stirred at room temperature for 2 h. The solvent was concentrated and the residue was taken without further purification to give the hydrochloride salt of compound 19c (360 mg). MS (ESI, M/z) 421.3[ M + H ]] +
The third step: (6- ((1- (cyclopropylcarbonyl) piperidin-4-yl) amino) pyrimidin-4-yl) (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (19)
The hydrochloride salt of compound 19c (360mg, 679.33. mu. mol), 19d (58.48mg, 679.33. mu. mol), DIPEA (526.78mg, 4.08mmol) and HATU (310mg, 815.2. mu. mol) were added to dichloromethane (10mL) and stirred at room temperature for 2 h. Water washing, dichloromethane extraction, concentration of the organic phase and purification of the residue by flash column chromatography (eluent system a) gave compound 19(150 mg).
MS(ESI,m/z):489.3[M+H] +
1H NMR(CDCl 3,400MHz)δ8.53(s,1H),7.20-7.09(m,3H),7.08-7.02(m,1H),6.66(d,J=0.9Hz,1H),5.69(s,1H),4.76(d,J=13.5Hz,1H),4.54-4.52(m, 1H),4.27-3.92(m,5H),3.36-3.30(m,1H),3.16-2.97(m,6H),2.91-2.76(m,2H),2.13-2.03(m,4H),1.90-1.71(m,3H),1.57-1.36(m,2H),1.02-0.94(m,2H),0.80-0.73(m,2H).
Example 20: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) -2-isopropylpyrimidin-4-yl) amino) piperidin-1-yl) ethanone (compound 20)
Figure PCTCN2020077581-APPB-000052
The first step is as follows: 6-hydroxy-2-isopropylpyrimidine-4-carboxylic acid (20b)
Dissolving 13a (3g,14.27mmol) in water (10mL), dropping NaOH aqueous solution (3.8mL,6.25mol/L), stirring at 25 deg.C for 1h, adding aqueous solution (5mL) containing 20a (2.71g,31.4mmol), adjusting pH to 11-12 with NaOH aqueous solution (6.25mol/L) under ice bath, and maintaining under ice bathStirring for 40min, adjusting to pH 1 with aqueous hydrochloric acid (12mol/L), filtering, washing the filter cake with aqueous solution (2X 10mL,0.1mol/L HCl), and drying to give compound 20b (1.2g) MS (ESI, M/z):183.2[ M + H: (M + H): 183.2: (M + H): compound] +
The second step is that: 6-chloro-2-isopropylpyrimidine-4-carbonyl chloride (20c)
20b (500mg,2.74mmol) was added to POCl3(5mL) was heated to 110 ℃ under nitrogen and stirred for 30min, and the solvent was concentrated to give compound 20c (0.5 g). The residue was used in the next reaction without purification.
The third step: (6-chloro-2-isopropylpyrimidin-4-yl) - (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (20d)
Adding 1d hydrochloride (792mg,2.74mmol) and DIPEA (2.48g,19.17mmol) into dichloromethane (15mL), adding dichloromethane (5mL) containing 20d (600mg,2.74mmol) dropwise into the reaction system, stirring at 25 deg.C for 1H, adding water (20mL) into the reaction system, washing with water, extracting, concentrating the organic phase, and purifying the residue by flash column chromatography (eluent system A) to obtain compound 20d (700mg). MS (ESI, M/z):399.2[ M + H] +.
The fourth step: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) -2-isopropylpyrimidin-4-yl) amino) piperidin-1-yl) ethanone (20)
Compound 20d (100mg, 250.67. mu. mol),1g (89.57mg, 501.34. mu. mol) and TEA (177.56mg,1.75mmol) were added to iPrOH (5mL) and heated to 90 ℃ and stirred for 16 h. The solvent was concentrated and the residue was subjected to preparative high performance liquid chromatography to give compound 20(50 mg).
MS(ESI,m/z):505.2[M+H] +
1H NMR(CDCl 3,400MHz)δ7.23-7.17(m,2H),7.16-7.10(m,1H),7.08-7.04(m,1H),6.46(s,1H),5.20-5.18(m,1H),4.81(d,J=12.2Hz,1H),4.54-4.49(m,1H),4.29-3.87(m,4H),3.84-3.80(m,1H),3.33-2.98(m,7H),2.97-2.92(m,1H),2.86-2.82(m,2H),2.19-2.14(m,6H),2.07-2.04(m,1H),1.92-1.88(m,2H),1.47-1.41(m,2H),1.26(d,J=6.9Hz,6H).
Example 21: 1- (4- ((2-cyclopropyl-6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyrimidin-4-yl) amino) piperidin-1-yl) ethanone (compound 21)
Figure PCTCN2020077581-APPB-000053
The first step is as follows: 2-cyclopropyl-6-hydroxypyrimidine-4-carboxylic acid (21b)
Dissolving 13e (5g,23.79mmol) in water (16mL), dropping an aqueous solution of NaOH (3.8mL,6.25mol/L), stirring at 25 ℃ for 1h, adding an aqueous solution (5mL) containing 21a (6.6g,54.72mmol), adjusting the pH to 11-12 with an aqueous solution of NaOH (6.25mol/L) under ice bath, stirring for 40min while maintaining ice bath, adjusting the pH to 1 with an aqueous solution of hydrochloric acid (8mol/L), filtering, washing the cake with an aqueous solution of hydrochloric acid (2 × 10mL,0.1mol/L), concentrating under reduced pressure, and drying to obtain compound 21b (2 g). MS (ESI, M/z) 181.1[ M + H ]] +.
The second step is that: 6-chloro-2-cyclopropylpyrimidine-4-carbonylchloro (21c)
21b (200mg,1.1mmol) was added to POCl3(2mL), stirred at 110 ℃ for 30min, the solvent was concentrated and the residue was used directly in the next reaction.
The third step: (6-chloro-2-cyclopropylpyrimidin-4-yl) - (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (21d)
Adding hydrochloride of 1d (308mg,1.2mmol) and DIPEA (358mg,2.8mmol) into dichloromethane (10mL), adding dichloromethane (5mL) solution containing 21c (1.1mmol) dropwise, stirring at room temperature for 1H, adding water (20mL) into the reaction system, washing with water, extracting, concentrating the organic phase, and purifying the residue by flash column chromatography (eluent system A) to obtain compound 21d (100mg) MS (ESI, M/z):397.2[ M + H: (M + H)] +.
The fourth step: 1- (4- ((2-cyclopropyl-6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyrimidin-4-yl) amino) piperidin-1-yl) ethanone (21)
Compound 21d (100mg, 252.63. mu. mol),1g (90mg, 503.4. mu. mol) and DIPEA (98mg, 755.8. mu. mol) were added to NMP (2mL), heated to 90 ℃ with a microwave, stirred for 3h, the solvent was concentrated, and the residue was isolated by preparative high performance liquid chromatography to give compound 21(15 mg).
MS(ESI,m/z):503.3[M+H] +
1H NMR(CDCl 3,400MHz)δ7.10-7.04(m,3H),6.99-6.97(m,1H),6.29(s,1H),4.99(s,1H),4.69(d,J=13.6Hz,1H),4.44(d,J=14.0Hz,1H),4.06-4.02(m,1H),3.89(s,2H),3.74(d,J=14.0Hz,1H),3.17-3.10(m,1H),3.03-2.96(m,5H),2.75(q,J=12.4Hz,2H),2.04-1.93(m,8H),1.72-1.69(m,2H),1.37-1.17(m,4H),0.97-0.87(m,4H).
Example 22: 1- (4- ((2-chloro-6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyrimidin-4-yl) amino) piperidin-1-yl) ethanone (compound 22)
Figure PCTCN2020077581-APPB-000054
The first step is as follows: 6- ((1-acetylpiperidin-4-yl) amino) -2-chloropyrimidine-4-carboxylic acid (22b)
Compound 22a (300mg,1.55mmol), 1g (555mg, 3.11mmol) and TEA (1.10g,10.88mmol) were added to iPrOH (10mL) and heated to 90 ℃ and stirred for 2 h. The solvent was concentrated and the residue was purified by flash column chromatography (eluent system a) to give compound 22b (250 mg). MS (ESI, M/z) 299.2[ M + H ]] +
The second step is that: 1- (4- ((2-chloro-6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) pyrimidin-4-yl) amino) piperidin-1-yl) ethanone (22)
Compound 22b (127mg, 425.14. mu. mol), hydrochloride salt of 1d (129mg, 445.86. mu. mol), DIPEA (220mg, 1.70mmol) and HATU (162mg, 425.14. mu. mol) were added to DMF (3mL) and stirred at room temperature for 2 h. The reaction solution was poured into water, extracted with ethyl acetate, and the organic phase was concentrated, and the residue was subjected to preparative high performance liquid chromatography to give compound 22(170 mg).
MS(ESI,m/z):497.1[M+H] +.
1H NMR(CDCl 3,400MHz)7.17-7.09(m,3H),7.06-7.02(m,1H),6.53(s,1H),6.04(s,1H),4.68(d,J=13.2Hz,1H),4.52(d,J=14Hz,1H),4.20(s,1H),4.02(d,J=13.2Hz,1H),3.89(s,2H),3.82(d,J=14Hz,1H),3.27-3.10(m,2H),2.99-2.78(m,7H),2.12(s,3H),2.11-1.95(m,4H),1.80-1.66(m,2H),1.48-1.36(m,2H).
Example 23: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) -2-methoxypyrimidin-4-yl) amino) piperidin-1-yl) ethanone (Compound 23)
Figure PCTCN2020077581-APPB-000055
Compound 22(50mg, 100.60. mu. mol), sodium methoxide (14mg, 251.49. mu. mol) were added to MeOH (3mL), heated to 60 ℃ and stirred for 3 h. After cooling to room temperature, the reaction was quenched by addition of saturated ammonium chloride solution (1mL), the solvent was concentrated, and the residue was separated by preparative high performance liquid chromatography to give compound 23(15 mg).
MS(ESI,m/z):493.2[M+H] +.
1H NMR(CDCl 3,400MHz)δ7.23-7.13(m,3H),7.10-7.05(m,1H),6.31(s,1H),5.38(m,1H),4.79(d,J=13.5Hz,1H),4.52(d,J=13.7Hz,1H),4.19(d,J=14.0Hz,1H),4.09(s,2H),3.92(s,3H),3.81(d,J=14.0Hz,1H),3.29-2.98(m,7H),2.81(q,J=11.6Hz,2H),2.20(d,J=13.6Hz,2H),2.11(s,3H),2.03(d,J=12.8Hz,2H),1.95-1.80(m,2H),1.46-1.33(m,2H).
Example 24: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) -2-morpholinopyrimidin-4-yl) amino) piperidin-1-yl) ethanone (compound 24)
Figure PCTCN2020077581-APPB-000056
Compound 22(50mg, 100.60. mu. mol), morpholine (35mg, 402.39. mu. mol) and DIPEA (52mg, 402.39. mu. mol) were added to iPrOH (3mL), and the reaction was heated to 120 ℃ with microwave stirring for 3 h. The reaction solution was concentrated, and the residue was separated by preparative high performance liquid chromatography to give compound 24(23 mg).
MS(ESI,m/z):548.2[M+H] +.
1H NMR(CDCl 3,400MHz)7.21-7.11(m,3H),7.07-7.03(m,1H),5.93(s,1H),4.86(t,J=6.8Hz,1H),4.76(d,J=13.5Hz,1H),4.49(d,J=13.6Hz,1H),4.18(d,J=13.6Hz,1H),3.97(s,2H),3.81(d,J=14.0Hz,1H),3.73(s,8H),3.20(td,J=13.9,2.9Hz,1H),3.11-2.91(m,6H),2.81-2.79(m,2H),2.12(s,3H),2.10-1.98(m,4H),1.84-1.71(m,2H),1.47-1.34(m,2H).
Example 25: 1- (4- (4- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) phenoxy) piperidin-1-yl) ethanone (Compound 25)
Figure PCTCN2020077581-APPB-000057
The first step is as follows: phenyl 4- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) acetate (25b)
Compound 1d (1g,3.96mmol), 25a (855.24mg, 4.75mmol), DIPEA (1.28g, 9.89mmol) and HATU (2.26g, 5.93mmol) were added to DMF (10mL) and stirred at room temperature for 2 h. The reaction was poured into water, extracted with ethyl acetate, the organic phase was concentrated and the residue was subjected to flash column chromatography (eluent system a) to give compound 25b (1.2 g). MS (ESI, M/z):379.2[ M + H] +
The second step is that: (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) (4-hydroxyphenyl) methanone (25c)
Compound 25b (1g,2.64mmol) and NaOH (2mL,2mol/L) were added to methanol (30mL), stirred at room temperature for 1h, pH adjusted to 6-7 with hydrochloric acid, extracted (DCM: isopropanol ═ 3:1), the organic phase concentrated and the residue chromatographed on flash column (eluent system a) to give compound 25c (0.56 g). MS (ESI, M/z) 337.2[ M + H ]] +
The third step: 4-hydroxy-N-tert-butoxycarbonyl-piperidine (25e)
25d (1.00g, 5.02mmol) was added to ethanol (20mL), and sodium borohydride (284.82mg,7.53mmol) was added to the reaction in portions under ice-bath, and stirred at room temperature for 4 h. Quenching the reaction with saturated aqueous ammonium chloride solution, extracting with dichloromethane, concentrating the organic phase under reduced pressure, and purifying the residue with flash columnPurification by chromatography (eluent system B) gave compound 25e (1.01 g). MS (ESI, M/z):302.3[ M + H] +
The fourth step: 4- (4- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) phenoxy) piperidine-1-carboxylic acid tert-butyl ester (25f)
25c (300mg, 891.73. mu. mol), 25e (358.94mg,1.78mmol) and PPh were placed under nitrogen3(467.77mg,1.78mmol) was added to dry THF (10mL), DIAD (360.63mg,1.78mmol) was added under ice-bath, and stirred at room temperature for 5 h. The reaction was poured into water, extracted with ethyl acetate, the organic phase was concentrated and the residue was subjected to flash column chromatography (eluent system a) to give compound 25f (0.46 g). MS (ESI, M/z) 520.2[ M + H ]] +
The fifth step: (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) (4-piperidin-4-yloxy) phenyl) methanone (25g)
Compound 25f (0.4g, 769.71. mu. mol) was added to a solution of dioxane hydrochloride (5mL,4mol/L) and stirred at room temperature for 2 h. To the reaction mixture was added an aqueous solution (10mL), extracted with ethyl acetate, and the organic phase was separated, concentrated, and the residue was purified without further purification to give 25g of a compound (280 mg). MS (ESI, M/z) 420.2[ M + H ]] +
And a sixth step: 1- (4- (4- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) phenoxy) piperidin-1-yl) ethanone (25)
25g (30mg, 60.92. mu. mol) was added to DCM (3mL), and triethylamine (30.82mg, 304.58. mu. mol) and acetyl chloride (9.56mg, 121.83. mu. mol) were added under ice-bath and stirred at room temperature for 2 h. The reaction solution was poured into water, extracted with ethyl acetate, and the organic phase was concentrated, and the residue was subjected to preparative high performance liquid chromatography to give compound 25(8 mg).
MS(ESI,m/z):462.3[M+H] +.
1H NMR(400MHz,CDCl 3)δ7.42-7.37(m,2H),7.14(m,3H),7.06-7.03(m,1H),6.95-6.90(m,2H),4.63-4.57(m,1H),3.92(s,1H),3.80-3.63(m,4H),3.43(m,1H),2.95(m,5H),2.13(s,3H),2.05(s,2H),2.00-1.77(m,6H),1.71(d,J=9.2Hz, 2H),1.26(t,J=6.5Hz,2H).
Example 26: (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) (4- ((1-pyrazin-2-ylmethyl) piperidin-4-yl) oxy) phenyl) methanone (Compound 26)
Figure PCTCN2020077581-APPB-000058
25g(100mg,238.35μmol)、K 2CO 3(98.82mg, 715.04. mu. mol) was added to DMF (3mL), 26a (66.57mg, 262.18. mu. mol) was added under ice-bath, and stirred at room temperature for 15 h. The reaction solution was poured into water, extracted with ethyl acetate, and the organic phase was concentrated, and the residue was subjected to preparative high performance liquid chromatography to give compound 26(35 mg).
MS(ESI,m/z):512.3[M+H]+.
1H NMR(400MHz,CDCl 3)δ8.70(d,J=1.2Hz,1H),8.54(dd,J=2.4,1.6Hz,1H),8.48(d,J=2.5Hz,1H),7.37(d,J=8.7Hz,2H),7.15-7.08(m,3H),7.04-7.01(m,1H),6.91(d,J=8.7Hz,2H),4.70(s,1H),4.45-4.37(m,1H),3.82(s,2H),3.73(s,2H),2.90(m,5H),2.82-2.73(m,3H),2.43(m,2H),2.21(s,1H),2.09-1.83(m,7H),1.64(d,J=7.8Hz,2H).
Example 27: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) azepan-1-carbonyl) pyrimidin-4-yl) amino) piperidin-1-yl) ethanone (compound 27)
Figure PCTCN2020077581-APPB-000059
The first step is as follows: 4- (3, 4-dihydroisoquinolin-2 (1H) -yl) azepane-1-carboxylic acid tert-butyl ester (27b)
Compound 27a (1.00g,4.69mmol) and 1b (749.41mg, 5.63mmol) were added sequentially to a mixed solution of tetrahydrofuran (20mL) and glacial acetic acid (1mL), stirred at room temperature for 30min, sodium cyanoborohydride (1.18g,18.76mmol) was added portionwise to the reaction system, and stirring at room temperature was continued for 3 h. The reaction was quenched with saturated aqueous ammonium chloride, extracted with dichloromethane, the solvent was concentrated under reduced pressure, and the residue was purified by flash column chromatography (eluent system B) to give compound 27B (500 mg). MS (ESI, m/z):331.3[M+H] +
The second step is that: 2- (azepan-4-yl) -1,2,3, 4-tetrahydroisoquinoline dihydrochloride (27c)
Compound 27b (450.00mg,1.36mmol) was added to a 4mol/L solution of dioxane hydrochloride (15mL) and stirred at room temperature for 2 h. Concentration under reduced pressure gave the hydrochloride salt of compound 27c (363.32 mg). MS (ESI, M/z) 231.2[ M + H ] +.
The third step: (6-Chloropyrimidin-4-yl) (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) azepan-1-yl) methanone (27d)
The hydrochloride salt of compound 27c (400mg,1.50mmol), 1e (261.46mg, 1.65mmol), DIPEA (775.03mg, 6.00mmol) and HATU (684.07mg, 1.80mmol) were added to DMF (4mL) and stirred at room temperature for 2 h. The reaction mixture was poured into water, extracted with ethyl acetate, the organic phase was concentrated, and the residue was subjected to flash column chromatography (eluent system a) to give compound 27d (130 mg). MS (ESI, M/z) 371.2[ M + H ]] +
The fourth step: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) azepan-1-carbonyl) pyrimidin-4-yl) amino) piperidin-1-yl) ethanone (27)
Compound 27d (40mg, 107.85. mu. mol),1g (38.54mg, 215.71. mu. mol) and TEA (76.4mg, 754.97. mu. mol) were added to iPrOH (2mL), heated to 90 ℃ and stirred for 5 h. The solvent was concentrated and the residue was subjected to preparative high performance liquid chromatography to give compound 27(15 mg).
MS(ESI,m/z):477.3[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ8.43(s,1H),7.60(d,J=7.5Hz,1H),7.08-7.03(m,4H),6.49(s,1H),4.25(d,J=13.0Hz,1H),4.08(s,1H),3.79(d,J=13.5Hz,1H),3.72-3.61(m,3H),3.46-3.43(m,2H),3.29-3.13(m,2H),2.79-2.63(m,6H),2.01(s,3H),1.94-1.77(m,6H),1.58-1.55(m,2H),1.43-1.22(m,2H).
Example 28: (2- ((1- (cyclopropylcarbonyl) piperidin-4-yl) amino) pyridin-4-yl) (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (Compound 28)
Figure PCTCN2020077581-APPB-000060
Compound 17c (0.1g, 249.81. mu. mol), Ruphos (23.33mg, 49.96. mu. mol), 28a (84.05 mg, 499.61. mu. mol), Pd2(dba) 3(22.88mg, 24.98. mu. mol) and sodium tert-butoxide (72.02mg,749.42mmol) were added successively to toluene (5mL) and stirred at 95 ℃ for 20 hours. The reaction solution was cooled to room temperature, filtered, washed with water, extracted, and the organic layer concentrated, and the residue was subjected to preparative high performance liquid chromatography to give compound 28(13 mg). MS (ESI, M/z) 488.3[ M + H ]] +.
1H NMR(400MHz,DMSO- d6)δ8.01(d,J=5.2Hz,1H),7.10-7.02(m,4H),6.70(d,J=7.6Hz,1H),6.43-6.42(dd,J=5.2Hz,1.2Hz,1H),6.40(s,1H),4.46(d,J=13.2Hz,1H),4.25-4.17(m,2H),3.99-3.98(m,1H),3.69(s,2H),3.61(d,J=13.6Hz,1H),3.29-3.23(m,3H),3.03(t,J=12.4Hz,1H),2.81-2.69(m,6H),2.02-1.78(m,4H),1.46-1.23(m,4H),0.72-0.67(m,4H).
Example 29: (2- (cyclohexylamino) pyridin-4-yl) (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (Compound 29)
Figure PCTCN2020077581-APPB-000061
Compound 17c (0.1g, 249.81. mu. mol), Ruphos (23.33mg, 49.96. mu. mol), 29a (49.55mg, 499.61. mu. mol), Pd2(dba) 3(22.88mg, 24.98. mu. mol) and sodium tert-butoxide (72.02mg,749.42mmol) were added successively to toluene (5mL) and stirred at 95 ℃ for 20 hours. The reaction solution was cooled to room temperature, filtered, washed with water, extracted, and the organic layer was concentrated, and the residue was subjected to preparative high performance liquid chromatography to give compound 29(15 mg). MS (ESI, M/z):419.3[ M + H] +.
1H NMR(400MHz,DMSO- d6)δ7.98(d,J=5.6Hz,1H),7.10-7.02(m,4H),6.55(d,J=7.6Hz,1H),6.38-6.36(m,2H),4.45(d,J=13.2Hz,1H),3.69-3.58(m,4H),3.02(t,J=12.8Hz,1H),2.76-2.65(m,5H),1.91-1.68(m,6H),1.48-1.42(m,3H),1.32-1.12(m,6H).
Example 30: (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) (2-methyl-6- ((tetrahydro-2H-pyran-4-yl) oxy) pyrimidin-4-yl) methanone (compound 30)
Figure PCTCN2020077581-APPB-000062
30a (55mg, 539.27. mu. mol), sodium tert-butoxide (65mg, 674.08. mu. mol) were added to tetrahydrofuran (3mL), stirred at room temperature for 1 hour, and a solution of compound 13e (100mg, 269.63. mu. mol) in tetrahydrofuran (1mL) was added to the reaction and stirred at room temperature for 3 hours. The reaction was quenched by addition of saturated ammonium chloride (1mL), the solvent was concentrated, and the residue was subjected to preparative high performance liquid chromatography to give compound 30(20 mg).
MS(ESI,m/z):437.2[M+H] +.
1H NMR(CDCl 3,400MHz)δ7.26-7.15(m,3H),7.12-7.06(m,1H),6.74(s,1H),5.38(tt,J=8.3,4.1Hz,1H),4.87(d,J=13.6Hz,1H),4.18(s,2H),4.06(d,J=14.0Hz,1H),3.98(dt,J=11.7,4.6Hz,2H),3.67-3.59(m,2H),3.42-3.07(m,6H),2.82(t,J=12.0Hz,1H),2.61(s,3H),2.36-2.19(m,2H),2.11-2.02(m,2H),1.98-1.76(m,4H).
Example 31: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) -2-ethylpyrimidin-4-yl) amino) piperidin-1-yl) ethanone (Compound 31)
Figure PCTCN2020077581-APPB-000063
The first step is as follows: 6-hydroxy-2-ethylpyrimidine-4-carboxylic acid (31b)
Dissolving 13a (2g,9.52mmol) in water (15mL), dropping NaOH aqueous solution (1.6mL,6.25mol/L), stirring at 25 ℃ for 1h, adding 31a (2.27g,20.94mmol) dissolved in water (5mL) to the reaction system, adjusting pH to 11-12 with NaOH aqueous solution (6.25mol/L) under ice bath, keeping stirring under ice bath for 40min, adjusting pH to 1 with hydrochloric acid aqueous solution (12mol/L), filtering, and adding hydrochloric acid aqueous solution (2) to the filter cake10mL,0.1mol/L HCl), concentrating the solvent under reduced pressure, and drying to give compound 31b (0.75g) MS (ESI, M/z):169.2[ M + H] +.
The second step is that: 6-chloro-2-ethylpyrimidine-4-carbonyl chloride (31c)
31b (700mg,4.16mmol) was added to SOCl2(10mL), DMF (304.27mg, 4.16mmol) was added, stirred at 85 ℃ for 4h, the reaction solution was cooled to room temperature, and the solvent was concentrated under reduced pressure to give the crude 31c, which was used directly in the next reaction.
The third step: (6-chloro-2-ethylpyrimidin-4-yl) - (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) methanone (31d)
Adding 1d hydrochloride (1.12g,3.87mmol) and DIPEA (3.50g,27.11mmol) into dichloromethane (10mL), dissolving 31c (793.95mg,3.87mmol) in dichloromethane (5mL), dropping into the reaction system, stirring at 25 deg.C for 1H, adding water (20mL) into the reaction solution, extracting with dichloromethane, washing the organic phase with saturated sodium chloride solution, filtering, concentrating the solvent under reduced pressure, and purifying the residue by flash column chromatography (eluent system A) to obtain compound 31d (800mg) MS (ESI, M/z):385.2[ M + H ] 385.] +.
The fourth step: 1- (4- ((6- (4- (3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carbonyl) -2-ethylpyrimidin-4-yl) amino) piperidin-1-yl) ethanone (31)
Compound 31d (70mg, 181.86. mu. mol),1g (65mg, 363.73. mu. mol) and TEA (74mg, 727.46. mu. mol) were added to iPrOH (2mL), heated to 90 ℃ and stirred for 2 h. The solvent was concentrated and the residue was subjected to preparative high performance liquid chromatography to give compound 31(15 mg).
MS(ESI,m/z):491.3[M+H] +.
1H NMR(400MHz,CDCl 3)δ7.22-7.16(m,3H),7.08(d,J=6.6Hz,1H),6.48(s,1H),5.35(s,1H),4.81(d,J=13.0Hz,1H),4.51(d,J=13.5Hz,1H),4.17-4.12(m,3H),3.82(d,J=13.5Hz,1H),3.30-3.04(m,7H),2.89-2.71(m,4H),2.25-1.80(m,10H),1.49-1.37(m,2H),1.28(t,J=7.6Hz,3H).
In the invention, the compounds 1-31 (except the compounds 15-19) are separated and purified by using an active 1260 type HPLC, the column temperature is 25 ℃, and the separation conditions are shown in the following table:
Figure PCTCN2020077581-APPB-000064
Figure PCTCN2020077581-APPB-000065
Figure PCTCN2020077581-APPB-000066
drug efficacy screening method and data
Experimental example 1 detection of inhibitory Effect of the Compound of the present invention on the enzymatic Activity of PRMT5/MEP50
The influence of the compound on the activity of PRMT5/MEP50 enzyme is tested by adopting a TR-FRET (time-resolved fluorescence) detection method in the experiment, so that the inhibition effect of the compound on signal path paths such as methylation of arginine residues of histone and non-histone on the enzyme molecule level is evaluated.
The kit used was: PRMT5TR-FRET Assay Kit (BPSbyscience)
The experimental steps are as follows:
1) taking a proper amount of DMSO solution of a 10mM compound to be detected, firstly diluting the compound to be detected by using DMSO in proportion, and then diluting the compound to be detected into 5 multiplied compound to be detected solution containing 1% DMSO concentration by using an assoy buffer; detecting by adopting a white shallow-hole 384-microtiter plate, adding 5 multiplied by the concentration of a compound solution to be detected into the plate, and detecting by 2 mu L/hole;
2) adding 4 XPRMT 5/MEP50(4 times of the final reaction system concentration of PRMT5/MEP50, namely 30 ng/muL) and 2.5 muL/hole (the final reaction system concentration is 10 muL, and the final reaction system concentration of PRMT5/MEP50 is 7.5 ng/muL), and incubating with the compound to be tested for 30 min;
3) adding 1.82 × substrate mixture (S-adenosylmethionine, biotinylated histone H4 peptide substrate, final reaction system concentration of 3 μ M, 0.5rxns/μ L and 5.5 μ L/hole respectively), mixing, reacting at room temperature for 2 hr;
4) adding 4 xEu into the reaction system according to the instructions in the kit3+Labeled antibody (4 × Eu)3+The concentration of the labeled antibody is 4 times of the concentration of the final detection system, provided by TR-FRET Assay Kit, BPSbeiioscience Cat #52120) solution, 5 mu L/hole, slowly oscillating, keeping out of the sun, and reacting for 30min at room temperature;
5) according to the instructions in the Kit, 4 Xfluorescent dye labeled receptor (4 Xfluorescent dye labeled receptor concentration is 4 times of the concentration of the final detection system, provided by TR-FRET Assay Kit, BPSbeiioscience Cat #52120) solution is added into the reaction system, 5 mu L/hole is slowly oscillated, and the reaction is carried out for 30min at room temperature in a dark place;
6) data for detection of TR-FRET mode, IC50Value estimation IC using simplified formula50Residual activity%/(1-residual activity%) x (x ═ compound concentration).
The test compounds of the invention all adopt four concentration gradients (1000, 100, 10, 0nM) to test the inhibition effect on PRMT5/MEP50 enzyme, and the results of the inhibition effect are shown in Table 1.
TABLE 1 inhibition of the activity of the PRMT5/MEP50 enzymes by the compounds of the invention
Compound (I) PRMT5/MEP50IC 50(μM)
1 0.07±0.01
2 0.54±0.01
3 0.43±0.05
4 0.31±0.03
5 0.19±0.01
6 0.86±0.09
7 0.58±0.01
8 1.00±0.15
9 0.78±0.10
10 0.90±0.09
11 0.21±0.03
12 0.34±0.11
13 0.05±0.02
14 0.60±0.02
15 0.56±0.04
16 0.12±0.04
17 0.26±0.01
18 0.95±0.17
19 0.19±0.12
20 0.40±0.08
21 0.23±0.04
22 0.11±0.02
23 0.19±0.02
24 0.40±0.05
25 1.10±0.06
26 0.90±0.04
27 0.09±0.03
28 0.38±0.07
29 0.70±0.26
30 0.15±0.04
31 0.40±0.03
The results show that the compound of the invention has obvious inhibition effect on the enzymatic activity of PRMT5/MEP 50.

Claims (24)

  1. A compound of formula I, a stereoisomer, a tautomer, or a mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound:
    Figure PCTCN2020077581-APPB-100001
    wherein:
    ring A is selected from C6-12Aryl and 5-to 10-membered heteroaryl, said C6-12Aryl, 5-10 membered heteroaryl may be optionally substituted with one or more of halogen, OH, CN, C1-4Alkyl radical, C1-4An alkoxy group;
    ring B is selected from C6-12Aryl, 5-10 membered heteroaryl, C3-8Cycloalkyl, 4-10 membered heterocyclyl, 9-12 membered arylheterocyclyi, 9-12 membered aryloateroaryl and 9-12 membered aryloaycloalkyl;
    R aand RbIndependently selected from H, halogenElement, OH, CN, NH2、NO 2、C 1-4Alkyl radical, C1-4Haloalkyl, C1-4Hydroxyalkyl radical, C1-4Alkoxy radical, C1-4Haloalkoxy and C3-8Cycloalkyl, or two RaAre linked to form a 3-8 membered cycloalkyl or 3-8 membered heterocyclyl; when a plurality of RaWhen occurring simultaneously, each RaMay be attached to the same or different carbon atoms and may be the same or different; when a plurality of RbWhen occurring simultaneously, each RbMay be attached to the same or different carbon atoms and may be the same or different;
    R cselected from halogen, OH, CN, NO2、C 1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, 9-12 membered aryloateroaryl, 9-12 membered aryloateroheterocyclyl, 9-12 membered aryloaycloalkyl, CO2R 20、C(O)SR 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、OS(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、C(O)NR 21SO 2R 25、OR 26、SR 26、OC(O)R 20、OCO 2R 20、OC(O)NR 21R 22、NR 23C(O)NR 21R 22、NR 23C(O)OR 20、C(=NR 21)R 22、C(=NR 21)OR 20、OC(=NR 23)R 21、OC(=NR 23)OR 20、C(=NR 23)NR 21R 22、OC(=NR 23)NR 21R 22、C(=NOR 20)R 24And NNR21R 22(ii) a Said C is1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, 9-12 membered aryloateroaryl, 9-12 membered aryloateroheterocyclyl, 9-12 membered aryloaycloalkyl may be optionally substituted with one or more of halogen, OH, CN, C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Haloalkyl, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, 9-12 membered arylheterocyclo, 9-12 membered arylcycloalkylo, CO2R 20、C(O)SR 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、OS(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、C(O)NR 21SO 2R 25、OR 26、SR 26、OC(O)R 20、OCO 2R 20、OC(O)NR 21R 22、NR 23C(O)NR 21R 22、NR 23C(O)OR 20、C(=NR 21)R 22、C(=NR 21)OR 20、OC(=NR 23)R 21、OC(=NR 23)OR 20、C(=NR 23)NR 21R 22、OC(=NR 23)NR 21R 22、C(=NOR 20)R 24、=NNR 21R 22(ii) a When a plurality of RcWhen occurring simultaneously, each RcMay be the same or different;
    R 20、R 21、R 22、R 23、R 24、R 25、R 26each independently selected from: H. c1-6Alkyl radical, C1-6Hydroxyalkyl radical, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl, 4-10 membered heterocyclyl, C6-12Aryl and 5-10 membered heteroaryl; said C is1-6Alkyl radical, C1-6Hydroxyalkyl radical, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl, 4-10 membered heterocyclyl, C6-12Aryl, 5-10 membered heteroaryl may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Haloalkoxy, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl-C1-4Alkyl-, CO2R 30、C(O)R 30、C(O)NR 31R 32、NR 33C(O)R 34、NR 31R 32、S(O) 2R 35、NR 31SO 2R 35、S(O)NR 31R 32、S(O) 2NR 31R 32(ii) a Or R21And R22Taken together to form a 3-8 membered heterocyclyl;
    R 30、R 31、R 32、R 33、R 34and R35Each independently selected from: H. c1-6Alkyl radical, C1-6Hydroxyalkyl radical, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl, 4-10 membered heterocyclyl, C6-12Aryl and 5-10 membered heteroaryl; said C is1-6Alkyl radical, C1-6Hydroxyalkyl radical, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl, 4-10 membered heterocyclyl, C6-12Aryl, 5-10 membered heteroaryl may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Haloalkoxy, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl; or R31And R32Taken together to form a 3-8 membered heterocyclyl;
    m is selected from 0, 1 and 2;
    n is selected from 0, 1 and 2, and m + n is less than or equal to 3;
    p is selected from 0 and 1;
    q is selected from 0, 1,2,3 and 4;
    t is selected from 0, 1,2,3 and 4;
    s is selected from 0, 1,2,3,4 and 5.
  2. The compound of claim 1, a stereoisomer, tautomer, or mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph or solvate of said compound, or a stable isotopic derivative, metabolite or prodrug of said compound, wherein ring a is C6-12Aryl radical, said C6-12Aryl groups may be optionally substituted with one or more of halogen, OH, CN, C1-4Alkyl radical, C1-4An alkoxy group; preferably, a is phenyl.
  3. The compound of claim 1 or 2, a stereoisomer, tautomer, or mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph or solvate of said compound, or a stable isotopic derivative, metabolite or prodrug of said compound, wherein ring B is selected from C6-12Aryl and 5-10 membered heteroaryl; preferably, ring B is selected from phenyl, pyridyl, pyridazinyl, pyrimidinyl and pyrazinyl.
  4. The compound of any one of claims 1-3, a stereoisomer, tautomer, or mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound, wherein R isaSelected from H, halogen, C1-4Alkyl and C1-4An alkoxy group; when a plurality of RaWhen occurring simultaneously, each RaMay be attached to the same or different carbon atoms and may be the same or different;
    more preferably, RaIs H.
  5. Compounds according to any one of claims 1-4A stereoisomer, a tautomer, or a mixture thereof of said compound, a pharmaceutically acceptable salt, a co-crystal, a polymorph or a solvate of said compound, or a stable isotopic derivative, metabolite or prodrug of said compound, wherein R isbSelected from H, halogen, OH, CN, C1-4Alkyl radical, C1-4Haloalkyl, C1-4Hydroxyalkyl radical, C1-4Alkoxy and C1-4A haloalkoxy group; when a plurality of RbWhen occurring simultaneously, each RbMay be attached to the same or different carbon atoms and may be the same or different;
    more preferably, RbIs H.
  6. The compound of any one of claims 1-5, a stereoisomer, tautomer, or mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound, wherein R iscSelected from halogen, OH, CN, NO2、C 1-6Alkyl radical, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, CO2R 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、OR 26、SR 26、OC(O)R 20And NR23C(O)NR 21R 22(ii) a Said C is1-6Alkyl radical, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl may be optionally substituted with one or more of halogen, OH, CN, C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Heteroalkyl group, C1-6Alkoxy radical, C3-8Cycloalkyl radical, C1-6Haloalkyl, C1-6Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, 9-12 membered arylheterocyclo, 9-12 membered arylcycloalkylo, CO2R 20、C(O)SR 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、OS(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、C(O)NR 21SO 2R 25、OR 26、SR 26、OC(O)R 20、OCO 2R 20、OC(O)NR 21R 22、NR 23C(O)NR 21R 22、NR 23C(O)OR 20、C(=NR 21)R 22、C(=NR 21)OR 20、OC(=NR 23)R 21、OC(=NR 23)OR 20、C(=NR 23)NR 21R 22、OC(=NR 23)NR 21R 22、C(=NOR 20)R 24、=NNR 21R 22(ii) a When a plurality of RcWhen occurring simultaneously, each RcMay be the same or different;
    preferably, RcSelected from halogen, OH, CN, NO2、C 1-4Alkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl radical, C1-4Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, CO2R 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、OR 26、SR 26、OC(O)R 20And NR23C(O)NR 21R 22(ii) a Said C is1-4Alkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl radical, C1-4Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl may be optionally substituted with one or more of halogen, OH, CN, C1-4Alkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl radical, C1-4Haloalkyl, C1-4Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, CO2R 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、OR 26、SR 26、OC(O)R 20、NR 23C(O)NR 21R 22(ii) a When a plurality of RcWhen occurring simultaneously, each RcMay be the same or different.
  7. The compound of any one of claims 1-6, a stereoisomer, tautomer, or mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound, wherein R iscIndependently selected from: halogen, C1-6Alkyl radical, C3-8Cycloalkyl radical, C1-6Alkoxy, 6-membered heterocyclic group, NR21R 22、OR 26And SR26Wherein R is21、R 22、R 26Each independently selected from: H. c1-6Alkyl, 4-6 membered heterocyclyl, 5-6 membered heteroaryl and C3-8Cycloalkyl radical, said C1-6Alkyl, 4-6 membered heterocyclyl, 5-6 membered heteroaryl and C3-8Cycloalkyl is optionally substituted with one or more of the following substituents: c1-6Alkyl, 4-6 membered heterocyclyl, 5-6 membered heteroaryl, C3-8Cycloalkyl radical, C1-6alkyl-C (O) -, C3-6cycloalkyl-C (O) -, C1-4alkyl-S (O)2And 6-membered heteroaryl-C1-4Alkyl-;
    preferably, RcIndependently selected from: halogen, C1-4Alkyl radical, C3-6Cycloalkyl radical, C1-4Alkoxy, morpholinyl, NR21R 22、OR 26And SR 26Wherein R is21、R 22、R 26Each independently selected from: H. optionally substituted piperidinyl, optionally substituted pyrrolidinyl, optionally substituted azetidinyl, optionally substituted oxetanyl, optionally substituted tetrahydrofuranyl, optionally substituted tetrahydropyranyl, optionally substituted pyridylmethyl, optionally substituted cyclohexyl, optionally substituted cyclobutyl, and optionally substituted cyclopropylmethyl, wherein the optionally substituted group is optionally substituted with one or more of the following substituents: c1-4alkyl-C (O) -, C3-6cycloalkyl-C (O) -, C1-4alkyl-S (O)2-and 6-membered heteroarylmethyl (preferably pyrazinylmethyl); preferably, said substituent is located on the nitrogen atom of said piperidinyl, pyrrolidinyl, azetidinyl;
    more preferably, RcIndependently selected from: halogen (e.g. chloro), methyl, ethyl, isopropyl, cyclopropyl, methoxy, morpholino, trifluoromethyl,
    Figure PCTCN2020077581-APPB-100002
    Figure PCTCN2020077581-APPB-100003
  8. The compound of any one of claims 1-7, a stereoisomer, tautomer, or mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound, wherein R iscIn the meta and/or para position relative to the carbonyl group attached to ring B.
  9. The compound of any one of claims 1-8, a stereoisomer, tautomer, or mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph or solvate of said compound, or a stable isotopic derivative, metabolite or prodrug of said compound, wherein m is 1; and/or
    n is selected from 1 and 2; and/or
    p is 1; and/or
    q is 0; and/or
    t is 0; and/or
    s is selected from 1 and 2.
  10. The compound according to any one of claims 1-9, a stereoisomer, a tautomer, or a mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound, wherein said compound has the structure of formula II-a:
    Figure PCTCN2020077581-APPB-100004
    wherein, ring A, Ra、R b、R cM, n, p, q, s, t are as defined in one of claims 1 to 9.
  11. The compound of any one of claims 1-9, a stereoisomer, a tautomer, or a mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound, wherein said compound has the structure of formula II-B:
    Figure PCTCN2020077581-APPB-100005
    wherein, ring A, Ra、R b、R cM, n, p, q, s, t are as defined in one of claims 1 to 9.
  12. The compound of any one of claims 1-9, a stereoisomer, a tautomer, or a mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound, wherein said compound has the structure of formula II-C:
    Figure PCTCN2020077581-APPB-100006
    wherein, ring A, Ra、R b、R cM, n, p, q, s, t are as defined in one of claims 1 to 9.
  13. The compound according to any one of claims 10-12, a stereoisomer, a tautomer, or a mixture thereof of the compound, a pharmaceutically acceptable salt, a co-crystal, a polymorph, or a solvate of the compound, or a stable isotopic derivative, metabolite, or prodrug of the compound, wherein:
    ring A is C6-12Aryl radical, said C6-12Aryl groups may be optionally substituted with one or more of halogen, OH, CN, C1-4Alkyl radical, C1-4An alkoxy group;
    R aselected from H, halogen, C1-4Alkyl and C1-4An alkoxy group; when a plurality of RaWhen occurring simultaneously, each RaMay be attached to the same or different carbon atoms and may be the same or different;
    R bselected from H, halogen, OH, CN, C1-4Alkyl radical, C1-4Haloalkyl, C1-4Hydroxyalkyl radical, C1-4Alkoxy and C1-4A haloalkoxy group; when a plurality of RbWhen occurring simultaneously, each RbMay be attached to the same or different carbon atoms and may be the same or different;
    R cselected from halogen, OH, CN, NO2、C 1-4Alkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl radical, C1-4Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, CO2R 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、OR 26、SR 26、OC(O)R 20And NR23C(O)NR 21R 22(ii) a Said C is1-4Alkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl radical, C1-4Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-4Alkyl radical, C1-4Heteroalkyl group, C1-4Alkoxy radical, C3-8Cycloalkyl radical, C1-4Haloalkyl, C1-4Hydroxyalkyl, 4-to 10-membered heterocyclic group, C6-12Aryl, 5-10 membered heteroaryl, CO2R 20、C(O)R 20、C(O)NR 21R 22、NR 23C(O)R 24、NR 21R 22、S(O) 2R 25、NR 21SO 2R 25、S(O)NR 21R 22、S(O) 2NR 21R 22、OR 26、SR 26、OC(O)R 20、NR 23C(O)NR 21R 22(ii) a When a plurality of RcWhen occurring simultaneously, each RcMay be the same or different;
    R 20、R 21、R 22、R 23、R 24、R 25、R 26each independently selected from H, C1-3Alkyl radical, C1-3Hydroxyalkyl radical, C1-3Heteroalkyl group, C1-3Alkoxy radical, C3-6Cycloalkyl, 4-6 membered heterocyclyl, C6-10Aryl and 5-6 membered heteroaryl; said C is1-3Alkyl radical, C1-3Hydroxyalkyl radical, C1-3Heteroalkyl group, C1-3Alkoxy radical, C3-6Cycloalkyl, 4-6 membered heterocyclyl, C6-10Aryl, 5-6 membered heteroaryl may be optionally substituted with one or more of the following substituents: halogen, OH, CN, C1-3Alkyl radical, C1-3Haloalkyl, C1-3Haloalkoxy, C1-3Heteroalkyl group, C1-3Alkoxy radical, C3-6Cycloalkyl radical, C1-3Hydroxyalkyl, 4-6 membered heterocyclyl, C6-10Aryl, 5-6 membered heteroaryl-C1-4Alkyl-, CO2R 30、C(O)R 30、C(O)NR 31R 32、NR 33C(O)R 34、NR 31R 32、S(O) 2R 35、NR 31SO 2R 35、S(O)NR 31R 32、S(O) 2NR 31R 32(ii) a Or R21And R22Taken together to form a 3-8 membered heterocyclyl;
    R 30、R 31、R 32、R 33、R 34、R 35each independently selected from H, methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, cyclopropyl, and propoxy;
    m is selected from 0, 1 and 2;
    n is selected from 0, 1 and 2, and m + n is less than or equal to 3;
    p is selected from 0 and 1;
    q is selected from 0, 1,2,3 and 4;
    s is selected from 0, 1,2,3,4 and 5; and is
    t is selected from 0, 1,2,3 and 4.
  14. The compound of claim 13, a stereoisomer, tautomer, or mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph or solvate of said compound, or a stable isotopic derivative, metabolite or prodrug of said compound, wherein:
    ring A is phenyl; raIs H; rbIs H;
    R cselected from halogen (e.g. chlorine), methyl, ethyl, isopropyl, cyclopropyl, methoxy, morpholino, methyl, ethyl, propyl, methyl, propyl, butyl, or a salt thereof,
    Figure PCTCN2020077581-APPB-100007
    m is selected from 1 and 2;
    n is selected from 1 and 2, and m + n is less than or equal to 3;
    p is 1;
    q is 1;
    s is selected from 1 and 2; and t is 1.
  15. The compound of claim 13, a stereoisomer, tautomer, or mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph or solvate of said compound, or a stable isotopic derivative, metabolite or prodrug of said compound, wherein:
    ring A is phenyl; raIs H; rbIs H;
    R cis selected from
    Figure PCTCN2020077581-APPB-100008
    m is 1;
    n is 1;
    p is 1;
    q is 1;
    s is 1; and t is 1.
  16. The compound of claim 13, a stereoisomer, tautomer, or mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph or solvate of said compound, or a stable isotopic derivative, metabolite or prodrug of said compound, wherein:
    ring A is phenyl; raIs H; rbIs H;
    R cis selected from
    Figure PCTCN2020077581-APPB-100009
    m is 1;
    n is 1;
    p is 1;
    q is 1;
    s is 1; and t is 1.
  17. The compound of claim 1, a stereoisomer, tautomer, or mixture thereof of said compound, a pharmaceutically acceptable salt, co-crystal, polymorph or solvate of said compound, or a stable isotopic derivative, metabolite or prodrug of said compound, wherein said compound is selected from the group consisting of:
    Figure PCTCN2020077581-APPB-100010
    Figure PCTCN2020077581-APPB-100011
  18. a pharmaceutical composition comprising a compound according to any one of claims 1-17, a stereoisomer, a tautomer, or a mixture thereof of said compound, a pharmaceutically acceptable salt, a co-crystal, a polymorph, or a solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound; optionally, the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers.
  19. A pharmaceutical formulation comprising a compound according to any one of claims 1-17, a stereoisomer, a tautomer, or a mixture thereof, a pharmaceutically acceptable salt, a co-crystal, a polymorph, or a solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound, or a pharmaceutical composition according to claim 18.
  20. Use of a compound according to any one of claims 1-17, a stereoisomer, a tautomer, or a mixture thereof, a pharmaceutically acceptable salt, a co-crystal, a polymorph, or a solvate of said compound, or a stable isotope derivative, metabolite, or prodrug of said compound, or a pharmaceutical composition according to claim 18, or a pharmaceutical formulation according to claim 19, for the preparation of a medicament for the prophylaxis and/or treatment of a disease associated with PRMT5 activity, such as a neoplastic disease;
    preferably, the neoplastic disease is selected from: brain tumor, lung cancer, squamous cell carcinoma, bladder cancer, stomach cancer, ovarian cancer, peritoneal cancer, pancreatic cancer, breast cancer, head and neck cancer, cervical cancer, endometrial cancer, rectal cancer, liver cancer, kidney cancer, esophageal adenocarcinoma, esophageal squamous cell carcinoma, prostate cancer, cancer of the female genital tract, carcinoma in situ, leukemia, lymphoma, neurofibroma, thyroid cancer, bone cancer, skin cancer, brain cancer, colon cancer, testicular cancer, gastrointestinal stromal tumor, prostate tumor, mast cell tumor, multiple myeloma, melanoma, glioma, or sarcoma.
  21. A compound according to any one of claims 1-17, a stereoisomer, a tautomer, or a mixture thereof, a pharmaceutically acceptable salt, a co-crystal, a polymorph, or a solvate of said compound, or a stable isotopic derivative, metabolite, or prodrug of said compound, or a pharmaceutical composition according to claim 18, or a pharmaceutical formulation according to claim 19, for use in the prevention and/or treatment of a disease associated with PRMT5 activity, such as a neoplastic disease;
    preferably, the neoplastic disease is selected from: brain tumor, lung cancer, squamous cell carcinoma, bladder cancer, stomach cancer, ovarian cancer, peritoneal cancer, pancreatic cancer, breast cancer, head and neck cancer, cervical cancer, endometrial cancer, rectal cancer, liver cancer, kidney cancer, esophageal adenocarcinoma, esophageal squamous cell carcinoma, prostate cancer, cancer of the female genital tract, carcinoma in situ, leukemia, lymphoma, neurofibroma, thyroid cancer, bone cancer, skin cancer, brain cancer, colon cancer, testicular cancer, gastrointestinal stromal tumor, prostate tumor, mast cell tumor, multiple myeloma, melanoma, glioma, or sarcoma.
  22. A method of treating a disease associated with PRMT5 activity, comprising administering to a subject in need thereof a therapeutically and/or prophylactically effective amount of a compound of any one of claims 1-17, a stereoisomer, tautomer, or mixture thereof, a pharmaceutically acceptable salt, co-crystal, polymorph, or solvate of said compound, or a stable isotope derivative, metabolite, or prodrug of said compound, or a pharmaceutical composition of any one of claims 18, or a pharmaceutical formulation of claim 19;
    preferably, the disease associated with PRMT5 activity is a neoplastic disease; more preferably, the neoplastic disease is selected from: brain tumor, lung cancer, squamous cell carcinoma, bladder cancer, stomach cancer, ovarian cancer, peritoneal cancer, pancreatic cancer, breast cancer, head and neck cancer, cervical cancer, endometrial cancer, rectal cancer, liver cancer, kidney cancer, esophageal adenocarcinoma, esophageal squamous cell carcinoma, prostate cancer, cancer of the female genital tract, carcinoma in situ, leukemia, lymphoma, neurofibroma, thyroid cancer, bone cancer, skin cancer, brain cancer, colon cancer, testicular cancer, gastrointestinal stromal tumor, prostate tumor, mast cell tumor, multiple myeloma, melanoma, glioma, or sarcoma.
  23. A method of synthesizing a compound of formula I-a, said method comprising the steps of:
    Figure PCTCN2020077581-APPB-100012
    ring A, ring B, Ra、R b、R cM, n, p, q, s, t are as defined in one of claims 1 to 9;
    the first step is as follows: carrying out reductive amination reaction on the compound I-1 and the compound I-2 to generate an intermediate I-3;
    the second step is that: deprotecting the compound I-3 under acidic conditions to produce an intermediate I-4;
    the third step: carrying out condensation reaction on the compound I-4 and the compound I-5 or acyl chloride of the compound I-5 to generate an intermediate I-6;
    the fourth step: the compound I-6 reacts with R under the action of alkalic-H is substituted or subjected to a coupling reaction, such as Buchwald-Hartwig reaction, to yield compound I-a.
  24. A method of synthesizing a compound of formula I-b, said method comprising the steps of:
    Figure PCTCN2020077581-APPB-100013
    ring A, Ra、R b、R cM, n, p, q, t are as defined in one of claims 1 to 9;
    the first step is as follows: the compound II-1 and the compound II-2 are subjected to ring closure under the action of alkali to generate an intermediate II-3;
    the second step is that: the compound II-3 generates an intermediate II-4 under the action of a chlorinating reagent;
    the third step: the compound II-4 and the compound I-4 are condensed under the action of alkali to generate an intermediate I-b.
CN202080008849.7A 2019-03-12 2020-03-03 Azacyclic compounds, preparation method and application thereof Active CN113330009B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN201910184914 2019-03-12
CN2019101849140 2019-03-12
PCT/CN2020/077581 WO2020182018A1 (en) 2019-03-12 2020-03-03 Nitrogen heterocyclic compound, preparation method therefor and use thereof

Publications (2)

Publication Number Publication Date
CN113330009A true CN113330009A (en) 2021-08-31
CN113330009B CN113330009B (en) 2024-02-09

Family

ID=72426918

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202080008849.7A Active CN113330009B (en) 2019-03-12 2020-03-03 Azacyclic compounds, preparation method and application thereof

Country Status (2)

Country Link
CN (1) CN113330009B (en)
WO (1) WO2020182018A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11077101B1 (en) 2018-07-18 2021-08-03 Tango Therapeutics, Inc. Compounds and methods of use
KR20220123229A (en) * 2019-12-17 2022-09-06 머크 샤프 앤드 돔 엘엘씨 PRMT5 inhibitors
MX2023001379A (en) 2020-07-31 2023-06-15 Tango Therapeutics Inc Piperidin-1- yl-n-pyrydi ne-3-yl-2-oxoacet am ide derivatives useful for the treatment of mtap-deficient and/or mt a-accumulating cancers.
CN115340528A (en) 2021-05-13 2022-11-15 上海翊石医药科技有限公司 Compound with anti-tumor activity and application thereof

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000000472A1 (en) * 1998-06-30 2000-01-06 Du Pont Pharmaceuticals Company 5-ht7 receptor antagonists
WO2014128465A1 (en) * 2013-02-20 2014-08-28 Cancer Therapeutics Crc Pty Ltd 2-(hetero)aryl-benzimidazole and imidazopyridine derivatives as inhibitors of asparagime emethyl transferase
CN105339351A (en) * 2013-03-14 2016-02-17 Epizyme股份有限公司 Arginine methyltransferase inhibitors and uses thereof
CN105452226A (en) * 2012-12-21 2016-03-30 Epizyme股份有限公司 Teatrahydro- and dihydro-isoquinoline PRMT5 inhibitors and uses thereof
CN105980374A (en) * 2013-12-19 2016-09-28 拜耳制药股份公司 Substituted piperidinyl-tetrahydroquinolines
CN106458978A (en) * 2013-12-19 2017-02-22 拜耳制药股份公司 Substituted piperidinyl-tetrahydroquinolines and their use as [alpha]-2c adrenoreceptor antagonists
US20170283407A1 (en) * 2014-09-03 2017-10-05 Ctxt Pty Ltd Tetrahydroisoquinoline derived prmt5-inhibitors
CN108570059A (en) * 2017-03-09 2018-09-25 中国科学院上海药物研究所 A kind of compound and its preparation and application with PRMT5 inhibitory activity
WO2019002074A1 (en) * 2017-06-29 2019-01-03 Bayer Aktiengesellschaft Thiazole compounds useful as prmt5 inhibitors

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000000472A1 (en) * 1998-06-30 2000-01-06 Du Pont Pharmaceuticals Company 5-ht7 receptor antagonists
CN105452226A (en) * 2012-12-21 2016-03-30 Epizyme股份有限公司 Teatrahydro- and dihydro-isoquinoline PRMT5 inhibitors and uses thereof
WO2014128465A1 (en) * 2013-02-20 2014-08-28 Cancer Therapeutics Crc Pty Ltd 2-(hetero)aryl-benzimidazole and imidazopyridine derivatives as inhibitors of asparagime emethyl transferase
CN105339351A (en) * 2013-03-14 2016-02-17 Epizyme股份有限公司 Arginine methyltransferase inhibitors and uses thereof
CN105980374A (en) * 2013-12-19 2016-09-28 拜耳制药股份公司 Substituted piperidinyl-tetrahydroquinolines
CN106458978A (en) * 2013-12-19 2017-02-22 拜耳制药股份公司 Substituted piperidinyl-tetrahydroquinolines and their use as [alpha]-2c adrenoreceptor antagonists
US20170283407A1 (en) * 2014-09-03 2017-10-05 Ctxt Pty Ltd Tetrahydroisoquinoline derived prmt5-inhibitors
CN108570059A (en) * 2017-03-09 2018-09-25 中国科学院上海药物研究所 A kind of compound and its preparation and application with PRMT5 inhibitory activity
WO2019002074A1 (en) * 2017-06-29 2019-01-03 Bayer Aktiengesellschaft Thiazole compounds useful as prmt5 inhibitors

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KONGKAI ZHU,ET AL.: ""Discovery of new potent protein arginine methyltransferase 5 (PRMT5) inhibitors by assembly of key pharmacophores from known inhibitors"", 《BIOORGANIC & MEDICINAL CHEMISTRY LETTERS》, vol. 28, pages 3693 - 3699, XP085555731, DOI: 10.1016/j.bmcl.2018.10.026 *
来源于SCIENTIFIC EXCHANGE,INC等提供的产品目录: ""STN检索报告"", 《数据库REGSTRY(在线)》, pages 1 - 26 *

Also Published As

Publication number Publication date
WO2020182018A1 (en) 2020-09-17
CN113330009B (en) 2024-02-09

Similar Documents

Publication Publication Date Title
CN113365988B (en) SHP2 inhibitor and application thereof
CN113767103B (en) Novel spirocyclic K-Ras G12C inhibitors
CN112142735B (en) Condensed cyanopyridine compound, preparation method and application
WO2021043077A1 (en) Substituted pyrazine compound and preparation method therefor and use thereof
CN113330009B (en) Azacyclic compounds, preparation method and application thereof
TWI585088B (en) Imidazo[1,2-b]pyridazine analogues as kinase inhibitors
CN113544128A (en) KRAS-G12C inhibitors
CN112778276A (en) Compound as SHP2 inhibitor and application thereof
KR20200021051A (en) Use of Rheto-associated protein kinase inhibitors, pharmaceutical compositions containing Rheto-associated protein kinase inhibitors, methods of manufacture and uses of pharmaceutical compositions
CN110467615A (en) Azolopyrimidines include its pharmaceutical composition and its preparation method and application
CN115315427B (en) HPK1 inhibitor and preparation method and application thereof
CN113748114A (en) Quinazoline compound and application thereof in medicine
JP2023530838A (en) Substituted Pyrazine Compounds, Pharmaceutical Compositions Containing Such Compounds, and Uses Thereof
JP2022511213A (en) Cell necrosis inhibitor, its preparation method and its use
CN115279749A (en) SHP2 inhibitor and composition and application thereof
CA2993096A1 (en) Fused ring pyrimidine compound, intermediate, and preparation method, composition and use thereof
CN113527299B (en) Nitrogen-containing condensed ring compound, preparation method and application
US20220144847A1 (en) Heterocyclic compound, pharmaceutical composition comprising same, preparation method therefor, and use thereof
WO2022017434A1 (en) Compound having kinase inhibitory activity
CN114728932A (en) Polyarylates as EGFR kinase inhibitors
CN112513041B (en) Tricyclic compounds
WO2023081637A1 (en) Fused tetracyclic quinazoline derivatives as inhibitors of erbb2
CN114075141A (en) Opioid receptor 'bias' ligand, preparation method and application thereof in medicine
CN116640094A (en) Compounds as PRMT1 inhibitors and uses thereof
CN113880804A (en) Novel benzimidazole compounds

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant