CN113999233A - BTK inhibitor ring derivative, preparation method and pharmaceutical application thereof - Google Patents

BTK inhibitor ring derivative, preparation method and pharmaceutical application thereof Download PDF

Info

Publication number
CN113999233A
CN113999233A CN202110840971.7A CN202110840971A CN113999233A CN 113999233 A CN113999233 A CN 113999233A CN 202110840971 A CN202110840971 A CN 202110840971A CN 113999233 A CN113999233 A CN 113999233A
Authority
CN
China
Prior art keywords
substituted
alkyl
methyl
membered
amino
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202110840971.7A
Other languages
Chinese (zh)
Inventor
张晨
廖雨亭
何平
陈孝刚
宣兆利
叶飞
唐平明
万松林
李瑶
倪佳
严庞科
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.)
Tibet Haisike Pharmaceutical Co ltd
Original Assignee
Sichuan Haisco Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sichuan Haisco Pharmaceutical Co Ltd filed Critical Sichuan Haisco Pharmaceutical Co Ltd
Publication of CN113999233A publication Critical patent/CN113999233A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Abstract

The invention provides a BTK inhibitor ring derivative, a preparation method and a pharmaceutical application thereof. The BTK inhibitor ring derivative is a compound shown in a general formula (I) or a stereoisomer, a solvate, a prodrug, a metabolite, a pharmaceutically acceptable salt or a co-crystal thereof. The BTK inhibitor ring derivatives are useful for the treatment of BTK related diseases such as tumors or autoimmune diseases. B-L-K (I).

Description

BTK inhibitor ring derivative, preparation method and pharmaceutical application thereof
Technical Field
The invention relates to a compound shown in a general formula (I) or a stereoisomer, a solvate, a prodrug, a metabolite, a pharmaceutically acceptable salt or a eutectic crystal thereof, an intermediate and a preparation method thereof, and application in BTK related diseases such as tumors or autoimmune system diseases.
Background
Bruton's Tyrosine Kinase (BTK) is a member of the non-receptor protein tyrosine kinase Tec family, is a key regulator in the B cell antigen receptor (BCR) signaling pathway, and is distributed in the lymphatic, hematopoietic, and blood systems. BTK mutations cause activation of signal pathways such as proliferation, differentiation and angiogenesis of downstream tumor cells, leading to X-linked agammaglobulinemia, non-hodgkin's lymphoma (NHL) and many B-cell malignancies, including Chronic Lymphocytic Leukemia (CLL), mantle cell lymphoma and diffuse large B-cell lymphoma. Because the BTK is mainly expressed in B cells and marrow cells, the BTK is a target with better targeting and safety.
PROTAC (protease targeting chimera) molecules are bifunctional compounds capable of simultaneously combining targeting proteins and E3 ubiquitin ligase, and the compounds can induce the recognition of the targeting proteins by proteasomes of cells, cause the degradation of the targeting proteins and effectively reduce the content of the targeting proteins in the cells. By introducing ligands capable of binding different target proteins into the ProTAC molecule, the application of the PROTAC technology to the treatment of various diseases becomes possible, and the technology has attracted much attention in recent years.
Disclosure of Invention
The invention develops a BTK inhibitor which has novel structure, good drug effect, high bioavailability and higher safety and is used for treating BTK related diseases such as tumors or autoimmune system diseases.
The invention develops a PROTAC compound of a BTK inhibitor and E3 ubiquitin ligase, which has novel structure, good drug effect, high bioavailability and higher safety and can inhibit or degrade BTK, and is used for treating BTK related diseases such as tumors or autoimmune system diseases.
The invention relates to a compound as shown in a general formula (I) or a stereoisomer, a solvate, a prodrug, a metabolite, a pharmaceutically acceptable salt or a eutectic crystal thereof, wherein
B-L-K (I)。
According to some embodiments of the invention, wherein L is selected from the group consisting of-Ak 1-Cy1-Ak2-Cy2-Ak3-Cy3-Ak4-Cy4-Ak 5-;
according to some embodiments of the invention, Ak1, Ak2, Ak3, Ak4 and Ak5 are each independently selected from CH2、O、-NRb0-C ═ O, C ≡ C or a bond;
according to some embodiments of the invention, wherein Ak1 and Ak5 are bonds, Ak2, Ak3 and Ak4 are each independently selected from CH2、O、-NRb0-or a bond;
according to some embodiments of the invention, B is selected from B1-W1-B2-B3-B4-, Ak1, Ak2, Ak3, Ak4 or Ak5, which are not bonds, are not directly linked to each other;
according to some embodiments of the invention, Ak1, Ak2, Ak3, Ak4 and Ak5 are each independently selected from CH2O, C ═ O, C ≡ C or a bond, and at least 4 of Ak1, Ak2, Ak3, Ak4, and Ak5 are selected from the bonds;
according to some embodiments of the invention, wherein each of Cy1, Cy2, Cy3, and Cy4 is independently selected from 3-to 12-membered heterocycle, 3-to 12-membered cycloalkyl, 6-to 10-membered aryl, or bond, said heterocycle, cycloalkyl, or aryl optionally further substituted with 0, 1,2,3, or 4 substituents selected from H, F, Cl, Br, I, OH, NH2、oxo、CF3、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl, CN, COOH or C1-4Alkoxy, said heterocycle containing 1,2,3, or 4 heteroatoms selected from O, S, N;
according to some embodiments of the invention, wherein each of Cy1, Cy2, Cy3, and Cy4 is independently selected from a bond, a 4-7 membered heteromonocyclic ring, a 5-10 membered heterobicyclic ring, a 6-12 membered heterospirocyclic ring, a 7-10 membered heterobridged ring, a 4-7 membered monocycloalkyl, a 5-10 membered benzocycloalkyl, a 6-12 membered spirocycloalkyl, a 7-10 membered bridged cycloalkyl, or a 6-10 membered aryl, said aryl, cycloalkyl, heteromonocyclic, heterobicyclic, heterospirocyclic, or heterobridged ring optionally further substituted with 0, 1,2,3, or 4 substituents selected from H, F, Cl, Br, I, OH, NH, F, Cl, Br, I, OH, and ci2、oxo、CF3、COOH、CN、C1-4Alkyl, hydroxy substituted C1-4Alkyl, halogen substituted C1-4Alkyl or C1-4Substituted by alkoxy substituents, said hetero-monocyclic, hetero-monocyclicAnd the bicyclic, heterospirocyclic or heterobridged ring contains 1,2,3 or 4 heteroatoms selected from O, S, N;
according to some embodiments of the invention, wherein each of Cy1, Cy2, Cy3 and Cy4 is independently selected from one of the following substituted or unsubstituted groups: a bond, phenyl, naphthyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclobutylcyclobutyl, cyclobutylcyclopentyl, cyclobutylcyclohexyl, cyclopentylcyclopentyl, cyclopentylcyclohexyl, cyclohexylcyclohexylcyclohexyl, cyclopropylcyclobutylcyclopentyl, cyclopropylcyclopentylyl, cyclopropylcyclohexylcyclohexyl, cyclobutylspirocyclobutyl, cyclobutylspirocyclopentyl, cyclobutylspirocyclohexyl, cyclopentylpirocyclopentyl, cyclopentylpirocyclohexyl, cyclohexylspirocyclohexyl, cyclopropylspirocyclobutyl, cyclopropylspirocyclopentyl, cyclopropylspirocyclohexyl, azetidinyl, azepinyl, piperidinyl, morpholinyl, piperazinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazolyl, tetrazolyl, cyclopropylazacyclobutyl, cyclopropylazacyclopentyl, cyclopropylazacylclocyclohexyl, Cyclopropylazapiperidinyl, cyclobutylazacyclobutyl, cyclobutylazacyclopentyl, cyclobutylazacyclohexyl, cyclobutylazopiperidinyl, cyclopentazacyclobutyl, cyclopentazacyclopentyl, cyclopentazacyclohexyl, cyclopentazacyclohexadinyl, cyclopentazacyclobutyl, cyclopentazacyclopentyl, cyclohexylazacyclohexyl, cyclohexylazacyclobutylazetidinyl, azetidinylcyclopentyl, azetidinylcyclohexyl, azetidinylboroazetidinyl, azetidinylcyclopentyl, azetidinylcyclohexyl, azetidinylcyclopentylgroup, azetidinopentylazacyclopentyl, azetidinohexylazacyclohexadecylohexylyl, azacyclopentadipiperidinyl, azetidinylcyclobutyl, azetidinohexylazacyclopentyl, azacyclohexazacyclopentadinyl, azacyclohexadipinyl, azetidinyl, azetidinylcohexylazacyclohexadinyl, azacyclohexadinyl, azacyclohexylpiperidinyl, azacyclohexadipinyl, azacyclohexadinyl, and the like, Cyclobutyl spiroazetidinyl, cyclopentyl spiroazetidinyl, cyclohexyl spiroazetidinylA group, a cyclohexylspiroazacyclohexylgroup, an azetidinylspiroazetidinyl group, an azetidinylspiroazacyclopentylpentyl group, an azetidinylspiroazacyclohexylgroup, an azetidinylspiroazetidinyl group, an azetidinylspiroazacyclohexylgroup, a cyclobutyl spiropiperidinyl group, a cyclopentylspiropiperidinyl group, a cyclohexylspiropiperidinyl group, an azetidinylspiropiperidinyl group, an azetidinylspiroazacylpiperidinyl group, an acid derivative thereof, a salt thereof, and a pharmaceutically acceptable salt thereof,
Figure BDA0003178773240000031
Figure BDA0003178773240000032
When substituted, is optionally further substituted with 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, NH2、oxo、CF3、CN、C1-4Alkyl, CF3、CH2OH, COOH or C1-4Substituted by a substituent of alkoxy;
according to some embodiments of the invention, when B is selected from B1-W1-B2-B3-B4-, Cy1, Cy2, Cy3 and Cy4 cannot be bonds at the same time;
according to some embodiments of the invention, each of Cy1, Cy2, Cy3 and Cy4 is independently selected from one of the following substituted or unsubstituted groups: a bond, azetidinyl, piperidine, morpholine, piperazine, cyclopropyloazetidinyl, cyclobutyloazetidinyl, cyclopentoazetidinyl, cyclohexyloazetidinyl, azetidinyl, azetidinoazetidinyl, azetidinyl, and azetidinylAn azacyclopentyl, an azacyclopenta-azacyclohexyl, an azacyclopenta-piperidine, an azacyclohexa-azetidinyl, an azacyclohexa-azacyclopentyl, an azacyclohexa-azacyclohexyl, a cyclobutyl-spiroazetidinyl, a cyclopentyl-spiroazetidinyl, a cyclohexyl-spiroazetidinyl, an azetidinyl-spiroazetidinyl, an, Azacyclohexylspirocyclohexylpiperidine, cyclopentylspiropiperidine, cyclohexylspiropiperidine, azetidinylspiropiperidine, azacyclohexylspiropiperidine, azacyclobutylspiropiperidine, azacyclobutylpiperidine, or,
Figure BDA0003178773240000041
Figure BDA0003178773240000042
Figure BDA0003178773240000043
Or
Figure BDA0003178773240000044
When substituted, is optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、COOH、CN、oxo、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
according to some embodiments of the invention, each Cy1, Cy2, Cy3, Cy4 is independently selected from one of the following substituted or unsubstituted groups: a bond,
Figure BDA0003178773240000045
Figure BDA0003178773240000046
Figure BDA0003178773240000051
When substituted, is optionally further substituted with 0 to 4 substituents selected from H, F, CF3Methyl, oxo, hydroxymethyl, COOH, CN or NH2Substituted with the substituent(s);
according to some embodiments of the invention, wherein B is selected from B1-W1-B2-B3-B4-or
Figure BDA0003178773240000052
According to some embodiments of the invention wherein B1 is selected from a 6 membered heteroaromatic ring or phenyl further optionally substituted with 0, 1,2,3 or 4Rb1(iii) substituted, said heteroaromatic ring contains 1,2,3, or 4 heteroatoms selected from O, S, N;
according to some embodiments of the invention, wherein B1 is selected from phenyl or pyridyl, said phenyl or pyridyl further optionally substituted with 0, 1,2,3 or 4Rb1Substituted;
according to some embodiments of the invention, wherein W1 is selected from-O-, -S-, -NH-, -NHCO-, or-CONH-;
according to some embodiments of the invention, wherein W1 is selected from-O-, -NHCO-, or-CONH-;
according to some embodiments of the invention wherein B2 is selected from a 6 membered heteroaromatic ring or phenyl optionally further optionally substituted with 0, 1,2,3 or 4Rb2(iii) substituted, said heteroaromatic ring contains 1,2,3, or 4 heteroatoms selected from O, S, N;
according to some embodiments of the invention, wherein B2 is selected from phenyl or pyridyl, said phenyl or pyridyl further optionally substituted with 0, 1,2,3 or 4Rb2Substituted;
according to some embodiments of the invention, wherein B3 is selected from 8-10 membered heterorings, said heterorings optionally further optionally substituted with 0, 1,2,3 or4Rb3(iii) substituted, said heteroring contains 1,2,3, or 4 heteroatoms selected from O, S, N;
according to some embodiments of the invention, wherein B3 is selected from substituted or unsubstituted imidazopyrimidines, pyrazolopyrimidines, imidazopyrazines, pyrazolopyrazines, imidazotetrahydropyrimidines, pyrazolotetrahydropyrimidines, when substituted, optionally further substituted with 0, 1,2,3, or 4Rb3Substituted;
according to some embodiments of the invention, wherein B4 is selected from the group consisting of 4-7 membered monocycloalkyl, 6-12 membered spirocycloalkyl, 5-10 membered benzocycloalkyl, and 7-10 membered bridged cycloalkyl, said monocycloalkyl, spirocycloalkyl, benzocycloalkyl, and bridged cycloalkyl groups optionally further optionally substituted with 0, 1,2,3, or 4Rb4Substituted;
according to some embodiments of the invention, wherein B4 is selected from cyclohexyl, cyclopentyl, cyclobutyl, said cyclohexyl, cyclopentyl, cyclobutyl being further optionally substituted by 0, 1,2,3 or 4Rb4Substituted;
according to some embodiments of the invention, B5Or B6Each independently selected from one of the following substituted or unsubstituted groups: phenyl or 5-6 membered heteroaryl, when substituted, optionally further substituted with 0 to 4Rb5(iii) substituted, said heteroaryl contains 1 to 4 heteroatoms selected from O, S, N;
according to some embodiments of the invention, B5Or B6Each independently selected from substituted or unsubstituted phenyl or 6 membered heteroaryl, when substituted, optionally further substituted with 0 to 4Rb1(iii) substituted, said heteroaryl contains 1 to 3N atoms;
according to some embodiments of the invention, B5Or B6Each independently selected from one of the following substituted or unsubstituted groups: phenyl or pyridyl, when substituted, optionally further substituted with 0 to 2Rb1Substituted;
according to some embodiments of the invention, wherein Rb0Selected from H, C1-4Alkyl, 3 to 12 membered cycloalkyl or 3 to 12 membered heterocycle containing 1,2,3 or 4 substituents selected from O,S, N;
according to some embodiments of the invention, wherein Rb0Is selected from methyl;
according to some embodiments of the invention, wherein Rb1、Rb2、Rb3Or Rb4Each independently selected from H, F, Cl, Br, I, OH, NH2、CN、CF3、COOH、CONH2、C1-4Alkyl, hydroxy substituted C1-4Alkyl, halogen substituted C1-4Alkyl or C1-4Alkoxy, said alkyl and alkoxy being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, NH2、CN、CONH2、C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
according to some embodiments of the invention, Rb5Each independently selected from H, F, Cl, Br, I, OH, NH2、CN、CF3、-C(=O)NH2、-C(=O)NH-C1-4Alkyl, -C (═ O) N (C)1-4Alkyl radical)2、C1-4Alkyl or C1-4Alkoxy, said alkyl or alkoxy being optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I or OH;
according to some embodiments of the invention, Rb5Each independently selected from H, F, Cl, Br, I, OH, CF3、C1-4Alkyl or C1-4Alkoxy, said alkyl or alkoxy being optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I;
according to some embodiments of the invention, Rb5Each independently selected from H, F, Cl, Br, I, methoxy or ethoxy;
according to some embodiments of the invention, Rb6Selected from H, C1-4Alkyl, halogen substituted C1-4Alkyl or hydroxy substituted C1-4An alkyl group;
according to some embodiments of the invention, Rb6Selected from H, C1-4An alkyl group;
according to some embodiments of the invention, Rb6Is selected from H;
according to some embodiments of the invention, Rb7、Rb8Each independently selected from H, F, Cl, Br, I, OH, NH2、CN、CF3、-C(=O)NH2、-C(=O)NH-C1-4Alkyl, -C (═ O) N (C)1-4Alkyl radical)2、C1-4Alkyl or C1-4Alkoxy, said alkyl or alkoxy being optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I or OH;
according to some embodiments of the invention, Rb7、Rb8Each independently selected from H, F, Cl, Br, I, NH2、CF3、-C(=O)NH2、C1-4Alkyl or C1-4Alkoxy, said alkyl or alkoxy being optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I;
according to some embodiments of the invention, Rb7、Rb8Each independently selected from H, F, NH2、CF3、-C(=O)NH2
According to some embodiments of the invention, Rb7、Rb8Taken together with the carbon atoms to which they are attached form a 5-6 membered heteroaryl or phenyl group optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、CN、CF3、-C(=O)NH2、-C(=O)NH-C1-4Alkyl, -C (═ O) N (C)1-4Alkyl radical)2、C1-4Alkyl or C1-4Alkoxy, said heteroaryl having 1 to 4 heteroatoms selected from O, S, N;
according to some embodiments of the invention, m is selected from 0, 1 or 2;
according to some embodiments of the invention, m is selected from 0 or 1;
according to some embodiments of the invention, m is selected from 1;
according to some embodiments of the invention, wherein B is selected from
Figure BDA0003178773240000071
Figure BDA0003178773240000072
According to some embodiments of the invention, wherein B is selected from
Figure BDA0003178773240000073
Figure BDA0003178773240000081
According to some embodiments of the invention, wherein B is selected from
Figure BDA0003178773240000082
Figure BDA0003178773240000083
According to some embodiments of the invention, wherein Rb1、Rb2、Rb3、Rb4Each independently selected from H, F, Cl, Br, I, OH, NH2、CN、CONH2、CF3COOH, hydroxymethyl, methyl or methoxy, said methyl or methoxy being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I;
according to some embodiments of the invention, wherein Rb1、Rb2Each independently selected from H, F, Cl, Br, OH, NH2、CN、CF3Hydroxymethyl, methyl or methoxy;
according to some embodiments of the invention, wherein each of n1, n2, n3, n4 is independently selected from 0, 1,2,3 or 4;
according to some embodiments of the invention, wherein B is selected from
Figure BDA0003178773240000084
Figure BDA0003178773240000091
According to the bookSome embodiments of the invention wherein L is selected from
Figure BDA0003178773240000092
Figure BDA0003178773240000093
Figure BDA0003178773240000094
Wherein the left side of the connecting rod is connected with the B,
according to some embodiments of the invention, wherein L may be selected from
Figure BDA0003178773240000095
Figure BDA0003178773240000096
Figure BDA0003178773240000101
Wherein the left side is connected with B;
according to some embodiments of the invention, wherein L may be selected from
Figure BDA0003178773240000102
Figure BDA0003178773240000103
Figure BDA0003178773240000104
Wherein the left side is connected with B;
according to some embodiments of the invention, wherein L is selected from
Figure BDA0003178773240000105
Figure BDA0003178773240000106
Figure BDA0003178773240000107
Wherein the left side is connected with B;
according to some embodiments of the invention, wherein L is selected from the group consisting of a bond,
Figure BDA0003178773240000108
Figure BDA0003178773240000109
Figure BDA0003178773240000111
Wherein the left side is connected with B;
according to some embodiments of the invention, wherein L is selected from
Figure BDA0003178773240000112
Figure BDA0003178773240000113
Figure BDA0003178773240000121
Figure BDA0003178773240000131
Figure BDA0003178773240000132
Wherein the left side is connected with B;
according to some embodiments of the invention, wherein L is selected from
Figure BDA0003178773240000141
Figure BDA0003178773240000142
Figure BDA0003178773240000151
Figure BDA0003178773240000161
Figure BDA0003178773240000171
Figure BDA0003178773240000181
Figure BDA0003178773240000191
Figure BDA0003178773240000192
Wherein the left side is connected with B;
according to some embodiments of the invention, wherein L is selected from
Figure BDA0003178773240000193
Figure BDA0003178773240000194
Figure BDA0003178773240000195
Wherein the left side is connected with B;
according to some embodiments of the invention, wherein K is selected from
Figure BDA0003178773240000196
Figure BDA0003178773240000201
According to some embodiments of the invention, wherein K is selected from
Figure BDA0003178773240000202
Figure BDA0003178773240000203
Figure BDA0003178773240000211
According to some embodiments of the invention, wherein K is selected from
Figure BDA0003178773240000212
Figure BDA0003178773240000213
According to some embodiments of the invention, wherein K is selected from
Figure BDA0003178773240000214
Figure BDA0003178773240000215
Figure BDA0003178773240000221
According to some embodiments of the invention, wherein ring E or F is selected from a benzene ring or a 5-6 membered heteroaromatic ring containing 1,2 heteroatoms selected from O, S, N;
according to some embodiments of the invention, wherein Rk2Is selected from CH2、C=O、S=O、SO2
According to some embodiments of the invention, wherein Rk2Is selected from CH2Or C ═ O;
according to some embodiments of the invention, wherein Rk1、Rk3Or Rk4Each independently selected from H, F, Cl, Br, I, OH, NH2、CF3、CN、COOH、C1-4Alkyl or C1-4An alkoxy group;
according to some embodiments of the invention, wherein Rk1、Rk3Or Rk4Each independently selected from H, CH3F, Cl, Br, I, OH or NH2
According to some embodiments of the invention, wherein Rk1、Rk3Or Rk4Each independently selected from H, F, Cl, Br, I, OH or NH2
According to some embodiments of the invention, wherein Rk5Is selected from C ═ O or
Figure BDA0003178773240000222
According to some implementations of the inventionScheme wherein, M1Is selected from the group consisting of a bond, -CH2-C (═ O) NH-or-C (═ O) CH2NH-;
According to some embodiments of the invention, wherein M is2Is selected from-NHC (═ O) -C1-6Alkyl, 8-10 membered hetero-cyclic or-NHC (═ O) -C3-6Cycloalkyl, said alkyl, hetero-fused ring or cycloalkyl being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, NH2、C1-4Alkyl, oxo or C1-4Substituted by a substituent of alkoxy;
according to some embodiments of the invention, wherein M is2Is selected from-NHC (═ O) -C1-6Alkyl or-NHC (═ O) -C3-6Cycloalkyl, said alkyl or cycloalkyl being optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
according to some embodiments of the invention, wherein M is3Is selected from-NH-or-O-;
according to some embodiments of the invention, wherein Rk6Is selected from C1-6Alkyl, said alkyl being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, C1-6Alkyl or C3-6Cycloalkyl, substituted with a substituent;
according to some embodiments of the invention, wherein Rk6Selected from methyl, ethyl, propyl, isopropyl, tert-butyl, isobutyl or sec-butyl;
according to some embodiments of the invention, wherein Rk7Each independently selected from H, F, Cl, Br, I, OH, SH, C1-6Alkyl radical, C1-6Alkoxy or C1-6Alkylthio radical, C1-6Alkyl formyloxy, said alkyl, alkoxy or alkylthio being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
according to some embodiments of the invention, wherein Rk7Each independently selected from H, F, OH, SH, methyl, methoxy or-SCH3
According to some embodiments of the invention, wherein Rk8、Rk9Each independently selected from H, C1-6Alkyl or C3-6Cycloalkyl, said alkyl or cycloalkyl being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, NH2、C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
according to some embodiments of the invention, wherein Rk8、Rk9Each independently selected from H, methyl, ethyl, cyclopropyl or cyclobutyl;
according to some embodiments of the invention, wherein Rk10Selected from 5-6 membered heteroaryl, said heteroaryl optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, CF3、CN、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl radical, C1-4Alkoxy or C3-6Cycloalkyl, substituted with a substituent;
according to some embodiments of the invention, wherein G is selected from 6-10 membered aryl or 5-10 membered heteroaryl, said aryl or heteroaryl is optionally further substituted with 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, CF3、CN、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl radical, C1-4Alkoxy or C3-6Cycloalkyl, said heteroaryl containing 1,2,3, or 4 heteroatoms selected from N, O, S;
according to some embodiments of the invention, wherein each of p1 or p2 is independently selected from 0, 1,2,3 or 4;
according to some embodiments of the invention, wherein each of p1 or p2 is independently selected from 0, 1 or 2;
according to some embodiments of the invention, wherein K is selected from
Figure BDA0003178773240000231
Figure BDA0003178773240000232
Figure BDA0003178773240000241
According to some embodiments of the invention, wherein K is selected from
Figure BDA0003178773240000242
Figure BDA0003178773240000243
Figure BDA0003178773240000251
According to some embodiments of the invention, wherein K is selected from
Figure BDA0003178773240000252
Figure BDA0003178773240000253
Figure BDA0003178773240000261
According to some embodiments of the invention, wherein K is selected from
Figure BDA0003178773240000262
Figure BDA0003178773240000263
Figure BDA0003178773240000271
The present invention provides a first embodiment relating to a compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein
B-L-K (I);
L is selected from-Ak 1-Cy1-Ak2-Cy2-Ak3-Cy3-Ak4-Cy4-Ak 5-;
Ak1、Ak2、ak3, Ak4 and Ak5 are each independently selected from CH2、O、-NRb0-C ═ O, C ≡ C or a bond;
cy1, Cy2, Cy3 and Cy4 are each independently selected from 3 to 12 membered heterocyclic ring, 3 to 12 membered cycloalkyl, 6 to 10 membered aryl or bond, said heterocyclic, cycloalkyl or aryl being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, NH2Oxo ═ O), CF3、CN、COOH、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl or C1-4Alkoxy, said heterocycle containing 1,2,3, or 4 heteroatoms selected from O, S, N;
cy1, Cy2, Cy3, and Cy4 cannot be bonds at the same time;
when Ak1, Ak2, Ak3, Ak4 or Ak5 are not bonds, the bonds can not be directly connected with each other;
b is selected from B1-W1-B2-B3-B4-or
Figure BDA0003178773240000281
B1 is selected from 6-membered heteroaromatic ring or phenyl further optionally substituted with 0, 1,2,3 or 4Rb1(iii) substituted, said heteroaromatic ring contains 1,2,3, or 4 heteroatoms selected from O, S, N;
w1 is selected from-O-, -S-, -NH-, -NHCO-or-CONH-;
b2 is selected from 6-membered heteroaromatic ring or phenyl optionally further optionally substituted with 0, 1,2,3 or 4Rb2(iii) substituted, said heteroaromatic ring contains 1,2,3, or 4 heteroatoms selected from O, S, N;
b3 is selected from 8-10 membered heteroring, optionally further optionally substituted with 0, 1,2,3 or 4Rb3(iii) substituted, said heteroring contains 1,2,3, or 4 heteroatoms selected from O, S, N;
b4 is selected from the group consisting of 4-7 membered monocycloalkyl, 6-12 membered spirocycloalkyl, 5-10 membered benzocycloalkyl, and 7-10 membered bridged cycloalkyl, said monocycloalkyl, spirocycloalkyl, benzocycloalkyl, and bridged cycloalkyl groups optionally further substituted with 0, 1,2,3, or 4Rb4Substituted;
B5or B6Each independently selected from one of the following substituted or unsubstituted groups: phenyl or 5-6 membered heteroaryl, when substituted, optionally further substituted with 0 to 4Rb5(iii) substituted, said heteroaryl contains 1 to 4 heteroatoms selected from O, S, N;
Rb0selected from H, C1-4Alkyl, 3 to 12 membered cycloalkyl or 3 to 12 membered heterocycle containing 1,2,3 or 4 heteroatoms selected from O, S, N;
Rb1、Rb2、Rb3or Rb4Each independently selected from H, F, Cl, Br, I, OH, NH2、CN、CF3、COOH、CONH2、C1-4Alkyl, hydroxy substituted C1-4Alkyl, halogen substituted C1-4Alkyl or C1-4Alkoxy, said alkyl and alkoxy being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, NH2、CN、CONH2、C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
Rb5each independently selected from H, F, Cl, Br, I, OH, NH2、CN、CF3、-C(=O)NH2、-C(=O)NH-C1-4Alkyl, -C (═ O) N (C)1-4Alkyl radical)2、C1-4Alkyl or C1-4Alkoxy, said alkyl or alkoxy being optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I or OH;
Rb6selected from H, C1-4Alkyl, halogen substituted C1-4Alkyl or hydroxy substituted C1-4An alkyl group;
Rb7、Rb8each independently selected from H, F, Cl, Br, I, OH, NH2、CN、CF3、-C(=O)NH2、-C(=O)NH-C1-4Alkyl, -C (═ O) N (C)1-4Alkyl radical)2、C1-4Alkyl or C1-4Alkoxy, said alkyl or alkoxy being optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I or OH;
or Rb7、Rb8Taken together with the carbon atoms to which they are attached form a 5-6 membered heteroaryl or phenyl group optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、CN、CF3、-C(=O)NH2、-C(=O)NH-C1-4Alkyl, -C (═ O) N (C)1-4Alkyl radical)2、C1-4Alkyl or C1-4Alkoxy, said heteroaryl having 1 to 4 heteroatoms selected from O, S, N;
m is selected from 0, 1 or 2;
k is selected from
Figure BDA0003178773240000291
Figure BDA0003178773240000292
Ring E or F is selected from a benzene ring or a 5-6 membered heteroaromatic ring containing 1,2 heteroatoms selected from O, S, N;
Rk2is selected from CH2、C=O、S=O、SO2
Rk1、Rk3Or Rk4Each independently selected from H, F, Cl, Br, I, OH, NH2、CF3、CN、COOH、C1-4Alkyl or C1-4An alkoxy group;
Rk5is selected from C ═ O or
Figure BDA0003178773240000301
M1Is selected from the group consisting of a bond, -CH2-C (═ O) NH-or-C (═ O) CH2NH-;
M2Is selected from-NHC (═ O) -C1-6Alkyl, 8-10 membered hetero-cyclic or-NHC (═ O) -C3-6Cycloalkyl, said alkyl, hetero-fused ring or cycloalkyl being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, NH2、C1-4Alkyl, oxo or C1-4Substituted by a substituent of alkoxy;
M3is selected from-NH-or-O-;
Rk6is selected from C1-6Alkyl, said alkyl being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, C1-6Alkyl or C3-6Cycloalkyl, substituted with a substituent;
Rk7each independently selected from H, F, Cl, Br, I, OH, SH, C1-6Alkyl radical, C1-6Alkoxy or C1-6Alkylthio radical, C1-6Alkyl formyloxy, said alkyl, alkoxy or alkylthio being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
Rk8、Rk9each independently selected from H, C1-6Alkyl or C3-6Cycloalkyl, said alkyl or cycloalkyl being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, NH2、C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
Rk10selected from 5-6 membered heteroaryl, said heteroaryl optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, CF3、CN、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl radical, C1-4Alkoxy or C3-6Cycloalkyl, substituted with a substituent;
g is selected from 6-10 membered aryl or 5-10 membered heteroaryl, said aryl or heteroaryl being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, CF3、CN、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl radical, C1-4Alkoxy or C3-6Cycloalkyl, said heteroaryl containing 1 to 4 heteroatoms selected from N, O, S;
p1 or p2 are each independently selected from 0, 1,2,3 or 4.
The second embodiment of the present invention relates to a compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein
Cy1, Cy2, Cy3 and Cy4 are each independently selected from a bond, 4-7 membered heteromonocyclic ring, 5-10 membered heterobicyclic ring, 6-12 membered heterospirocyclic ring, 7-10 membered heterobridged ring, 4-7 membered monocycloalkyl, 5-10 membered benzocycloalkyl, 6-12 membered spirocycloalkyl, 7-10 membered bridged cycloalkyl or 6-10 membered aryl, said aryl, cycloalkyl, heteromonocyclic, heterobicyclic, heterospirocyclic or heterobridged ring being optionally further substituted with 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, NH, Cl, Br, I, O2、oxo、CF3、COOH、CN、C1-4Alkyl, hydroxy substituted C1-4Alkyl, halogen substituted C1-4Alkyl or C1-4(ii) alkoxy, said heteromonocyclic, heterobicyclic, heterospirocyclic, or heterobridged ring containing 1,2,3, or 4 heteroatoms selected from O, S, N;
b is selected from B1-W1-B2-B3-B4-;
b1 is selected from phenyl or pyridyl further optionally substituted by 0, 1,2,3 or 4Rb1Substituted;
w1 is selected from-O-, -NHCO-, or-CONH-;
b2 is selected from phenyl or pyridyl further optionally substituted by 0, 1,2,3 or 4Rb2Substituted;
b3 is selected from substituted or unsubstituted imidazopyrimidines, pyrazolopyrimidines, imidazopyrazines, pyrazolopyrazines, imidazotetrahydropyrimidines, pyrazolotetrahydropyrimidines, when substituted, optionally further substituted with 0, 1,2,3 or 4Rb3Substituted;
b4 is selected from cyclohexyl, cyclopentyl and cyclobutyl, which are further optionally substituted by 0, 1,2,3 or 4Rb4Substituted;
k is selected from
Figure BDA0003178773240000311
Figure BDA0003178773240000312
Rk2Is selected from CH2、C=O;
Rk1、Rk3Or Rk4Each independently selected from H, F, Cl, Br, I, OH or NH2
M1Is selected from the group consisting of a bond, -CH2-C (═ O) NH-or-C (═ O) CH2NH-;
M2Is selected from-NHC (═ O) -C1-6Alkyl, 8-10 membered hetero-cyclic or-NHC (═ O) -C3-6Cycloalkyl, said alkyl, hetero-fused ring or cycloalkyl being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, NH2、C1-4Alkyl, oxo or C1-4Substituted by a substituent of alkoxy;
Rk6is selected from C1-6Alkyl, said alkyl being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, C1-6Alkyl or C3-6Cycloalkyl, substituted with a substituent;
Rk7each independently selected from H, F, Cl, Br, I, OH, SH, C1-6Alkyl radical, C1-6Alkoxy or C1-6Alkylthio radical, C1-6Alkyl formyloxy, said alkyl, alkoxy or alkylthio being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
Rk8、Rk9each independently selected from H, C1-6Alkyl or C3-6Cycloalkyl, said alkyl or cycloalkyl being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, NH2、C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
Rk10selected from 5-6 membered heteroaryl, said heteroaryl optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, CF3、CN、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl radical, C1-4Alkoxy or C3-6Cycloalkyl, substituted with a substituent;
the other groups are as defined in the first embodiment.
The third embodiment of the present invention relates to a compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein
Cy1, Cy2, Cy3 and Cy4 are each independently selected from the group consisting of a bond, phenyl, naphthyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclobutylcyclobutyl, cyclobutylcyclopentyl, cyclobutylocyclocyclohexyl, cyclopentylcyclohexyl, cyclohexylocyclocyclohexyl, cyclopropylcyclobutylcyclobutyl, cyclopropylocyclocyclopentyl, cyclopropylcyclohexylcyclohexyl, cyclobutylspirocyclopentyl, cyclobutylspirocyclohexyl, cyclopentylprofospiranyl, cyclopentylprofecolhexyl, cyclohexylspirocyclohexyl, cyclopropylspirocyclobutyl, cyclopropylspirocyclopentyl, cyclopropylspirocyclohexyl, azetidinyl, piperidinyl, morpholinyl, piperazinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazolyl, tetrazolyl, cyclopropyloazetidinyl, Cyclopropylazacyclohexylyl, cyclopropylazapiperidinyl, cyclobutylazacyclobutyl, cyclobutylazacyclopentyl, cyclobutylazacyclohexyl, cyclobutylpiperidinyl, cyclopentazacyclobutyl, cyclopentazacyclopentyl, cyclopentazacyclohexyl, cyclopentazacyclopentyl, cyclohexylazacyclobutyl, cyclohexylazacyclopentyl, cyclohexylazacyclohexyl, azetidinyl, azetidinoazetidinyl, azetidinyl, cyclopentyl, azetidinyl, cyclopentyl, cyclohexyl-and azetidinyl, Azacyclohexylpiperidinyl, cyclobutyl spiroazetidinyl, cyclopentyl spiroazetidinyl, cyclohexyl spiroazetidinylA group, a cyclohexylspiroazacyclohexylgroup, an azetidinylspiroazetidinyl group, an azetidinylspiroazacyclopentylpentyl group, an azetidinylspiroazacyclohexylgroup, an azetidinylspiroazetidinyl group, an azetidinylspiroazacyclohexylgroup, a cyclobutyl spiropiperidinyl group, a cyclopentylspiropiperidinyl group, a cyclohexylspiropiperidinyl group, an azetidinylspiropiperidinyl group, an azetidinylspiroazacylpiperidinyl group, an acid derivative thereof, a salt thereof, and a pharmaceutically acceptable salt thereof,
Figure BDA0003178773240000331
Figure BDA0003178773240000332
When substituted, is optionally further substituted with 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I, OH, NH2、oxo、CF3、CN、C1-4Alkyl, CF3、CH2OH, COOH or C1-4Substituted by a substituent of alkoxy;
b is selected from
Figure BDA0003178773240000333
Figure BDA0003178773240000334
Rb1、Rb2、Rb3Or Rb4Each independently selected from H, F, Cl, Br, I, OH, NH2、CN、CONH2、CF3COOH, hydroxymethyl, methyl or methoxy, said methyl or methoxy being optionally further substituted by 0, 1,2,3 or 4 substituents selected from H, F, Cl, Br, I;
n1, n2, n3, n4 are each independently selected from 0, 1,2,3 or 4;
the other groups are defined as in the first and second embodiments.
The fourth embodiment of the present invention relates to a compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein
L is selected from
Figure BDA0003178773240000335
Figure BDA0003178773240000336
Figure BDA0003178773240000341
Or L may be selected from
Figure BDA0003178773240000342
Figure BDA0003178773240000343
Figure BDA0003178773240000351
Or L may be selected from
Figure BDA0003178773240000352
Figure BDA0003178773240000353
The left side of the L is connected with the B, and the right side of the L is connected with the K;
b is selected from
Figure BDA0003178773240000354
Figure BDA0003178773240000355
Rb1、Rb2Each independently selected from H, F, Cl, Br, OH, NH2、CN、CF3Hydroxymethyl, methyl or methoxy;
k is selected from
Figure BDA0003178773240000356
Figure BDA0003178773240000357
Figure BDA0003178773240000361
The other groups are as defined in the first, second or third schemes.
The fifth embodiment of the present invention relates to a compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal thereof, wherein
B is selected from
Figure BDA0003178773240000362
At least 4 of Ak1, Ak2, Ak3, Ak4 and Ak5 are selected from bonds;
cy1, Cy2, Cy3 and Cy4 are each independently selected from a bond, a 4-7 membered azamonocyclic ring, a 5-10 membered azabicyclic ring, a 7-10 membered heterobridged ring or a 6-12 membered nitrogen-containing heterospirocyclic ring, said heteromonocyclic, heterofused, heterobridged or heterospirocyclic ring optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I, OH, COOH, CN, NH, and mixtures thereof2、oxo、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl or C1-4(ii) alkoxy, said heteromonocyclic, heterobicyclic, heterobridged, or heterospirocyclic ring containing 1 to 4 heteroatoms selected from O, S, N;
k is selected from
Figure BDA0003178773240000371
Figure BDA0003178773240000372
The other groups are as defined in the first embodiment.
The sixth embodiment of the present invention relates to a compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal thereof, wherein
Ak1、Ak2. Ak3, Ak4 and Ak5 are each independently selected from CH2O, or a bond, and at least 4 of Ak1, Ak2, Ak3, Ak4, and Ak5 are selected from the group consisting of bonds;
cy1, Cy2, Cy3 and Cy4 are each independently selected from one of the following substituted or unsubstituted groups: a bond, azetidinyl, azepinyl, piperidine, morpholine, piperazine, cyclopropyloazetidinyl, cyclobutyloazetidinyl, cyclopentoazetidinyl, cyclohexyloazetidinyl, azetidinyl, azetidinoheteroazetidinyl, cyclohexyloazetidinyl, azetidinoazetidinyl, azetidinyl, azetidinoheteroazetidinyl, azetidinoazetidinyl, cyclopentoazetidinyl, azetidinyl, cyclopentaizacetyl, cyclopentaizazetidinyl, cyclopentaizacyloheteropiperidyl, cyclopentaizacylo-azacyclobutylazacycloteyl, cyclopentaizacylo-azacycloteyl, cyclopentaizacylo-azacyclotebucin, cyclopentaizacylo-azacyclo-pentyl, cyclopentaidyl, cyclopentaizacylo-azacyclo-hexyl, and piperidine, Azetidinyl, azacyclohexylazapiperidino, cyclobutyl spiroazetidinyl, cyclopentyl spiroazetidinyl, cyclohexyl spiroazetidinyl, cyclohexylspiroazetidinyl, azetidinyl spiroazetidinyl, azetidinyl, and the like, Cyclohexyl spiropiperidine, azetidinyl spiropiperidine, azepinyl spiropiperidine, azetidinyl spiropiperidine, and azetidinyl spiropiperidine,
Figure BDA0003178773240000381
Figure BDA0003178773240000382
Figure BDA0003178773240000383
When substituted, is optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、COOH、CN、oxo、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
B5or B6Each independently selected from substituted or unsubstituted phenyl or 6 membered heteroaryl, when substituted, optionally further substituted with 0 to 4Rb1(iii) substituted, said heteroaryl contains 1 to 3N atoms;
Rb5independently selected from H, F, Cl, Br, I, OH, CF3、C1-4Alkyl or C1-4Alkoxy, said alkyl or alkoxy being optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I;
Rb6selected from H, C1-4An alkyl group;
Rb7、Rb8each independently selected from H, F, Cl, Br, I, NH2、CF3、-C(=O)NH2、C1-4Alkyl or C1-4Alkoxy, said alkyl or alkoxy being optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I;
m is selected from 0 or 1;
k is selected from
Figure BDA0003178773240000384
Figure BDA0003178773240000385
Figure BDA0003178773240000391
Rk2Each independently selected from CH2Or C ═ O;
Rk1、Rk3or Rk4Each independently selected from H, CH3F, Cl, Br, I, OH or NH2
M1Is selected from the group consisting of a bond, -CH2-C (═ O) NH-or-C (═ O) CH2NH-;
M2Is selected from-NHC (═ O) -methyl, -NHC (═ O) -ethyl, -NHC (═ O) -cyclopropyl, -NHC (═ O) -cyclobutyl, -NHC (═ O) -cyclopentyl or-NHC (═ O) -cyclohexyl, said methyl, ethyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl being optionally further substituted by 0 to 4 groups selected from H, F, Cl, Br, I, OH, NH2、C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
Rk6selected from methyl, ethyl, propyl, isopropyl, tert-butyl, isobutyl or sec-butyl;
Rk7each independently selected from H, F, OH, SH, methyl, methoxy or-SCH3
Rk8、Rk9Each independently selected from H, methyl, ethyl, cyclopropyl or cyclobutyl;
p1 or p2 are each independently selected from 0, 1 or 2;
the other groups are defined as in the first and fifth schemes.
The seventh embodiment of the present invention relates to a compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal thereof, wherein
Cy1, Cy2, Cy3 and Cy4 are each independently selected from one of the following substituted or unsubstituted groups: a bond,
Figure BDA0003178773240000392
Figure BDA0003178773240000401
Figure BDA0003178773240000402
When substituted, is optionally further substituted with 0 to 4 substituents selected from H, F, CF3Methyl, oxo, hydroxymethyl, COOH, CN or NH2Substituted with the substituent(s);
b is selected from
Figure BDA0003178773240000403
K is selected from
Figure BDA0003178773240000404
Figure BDA0003178773240000405
Figure BDA0003178773240000411
The other groups are as defined in the first, fifth or sixth schemes.
The eighth embodiment of the present invention relates to a compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein
L is selected from a bond,
Figure BDA0003178773240000412
Figure BDA0003178773240000413
Figure BDA0003178773240000414
Wherein the left side is connected with B;
or L is selected from
Figure BDA0003178773240000415
Figure BDA0003178773240000416
Figure BDA0003178773240000421
Figure BDA0003178773240000431
Figure BDA0003178773240000441
Figure BDA0003178773240000442
Wherein the left side is connected with B;
or L is selected from
Figure BDA0003178773240000443
Figure BDA0003178773240000444
Figure BDA0003178773240000451
Figure BDA0003178773240000461
Figure BDA0003178773240000471
Figure BDA0003178773240000481
Figure BDA0003178773240000491
Figure BDA0003178773240000492
Wherein the left side is connected with B;
or L is selected from
Figure BDA0003178773240000493
Figure BDA0003178773240000501
Figure BDA0003178773240000502
Wherein the left side is connected with B;
the other groups are as defined in the first, fifth, sixth or seventh embodiment.
The ninth embodiment of the present invention relates to a compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal thereof, wherein
B is selected from
Figure BDA0003178773240000503
Figure BDA0003178773240000504
Or B is selected from
Figure BDA0003178773240000505
Figure BDA0003178773240000511
The other groups are as defined in the first, fifth, sixth, seventh or eighth schemes.
The tenth embodiment of the present invention relates to a compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal thereof, wherein
K is selected from
Figure BDA0003178773240000512
Figure BDA0003178773240000513
Figure BDA0003178773240000521
The other groups are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth or ninth embodiments.
The eleventh embodiment of the present invention relates to a compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein
K is selected from
Figure BDA0003178773240000522
Figure BDA0003178773240000523
Figure BDA0003178773240000531
The other groups are as defined in the first, second, third, fourth, fifth, sixth, seventh, eighth or ninth embodiments.
Some embodiments of the present invention relate to a compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein the compound is selected from one of the following structures:
Figure BDA0003178773240000532
Figure BDA0003178773240000541
Figure BDA0003178773240000551
Figure BDA0003178773240000561
Figure BDA0003178773240000571
Figure BDA0003178773240000581
Figure BDA0003178773240000591
some embodiments of the present invention relate to a compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein the salt is selected from the group consisting of trifluoroacetate.
The invention relates to a pharmaceutical composition, which comprises the compound or stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or eutectic crystal thereof, and a pharmaceutically acceptable carrier.
The invention relates to application of the compound or stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or eutectic crystal thereof in preparing a medicament for treating diseases related to BTK activity or expression.
The invention relates to application of the compound or stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or eutectic crystal thereof in preparing a medicament for treating diseases related to inhibiting or degrading BTK.
The compound or the stereoisomer, the solvate, the prodrug, the metabolite, the pharmaceutically acceptable salt or the eutectic crystal thereof can be used for treating tumors or autoimmune diseases.
The compound or the stereoisomer, the solvate, the prodrug, the metabolite, the pharmaceutically acceptable salt or the eutectic crystal thereof are applied to tumors, wherein the tumors are selected from non-Hodgkin's lymphoma, chronic lymphocytic leukemia, mantle cell lymphoma and B cell lymphoma, and the autoimmune diseases are selected from rheumatoid arthritis or psoriasis.
Unless stated to the contrary, the terms used in the specification and claims have the following meanings.
Where carbon, hydrogen, oxygen, sulfur, nitrogen or F, Cl, Br, I are involved in the radicals and compounds of the invention, including their isotopes, and where carbon, hydrogen, oxygen, sulfur or nitrogen are involved in the radicals and compounds of the invention, optionally further replaced by one or more of their corresponding isotopesWherein the isotope of carbon comprises12C、13C and14c, isotopes of hydrogen including protium (H), deuterium (D, also called deuterium), tritium (T, also called deuterium), isotopes of oxygen including16O、17O and18isotopes of O, sulfur including32S、33S、34S and36isotopes of S, nitrogen include14N and15isotopes of N, F include17F and19isotopes of F, chlorine including35Cl and37cl, isotopes of bromine including79Br and81Br。
"halogen" means F, Cl, Br or I.
"halo-substituted" refers to F, Cl, Br, or I substitution, including but not limited to 1 to 10 substituents selected from F, Cl, Br, or I, 1 to 6 substituents selected from F, Cl, Br, or I, and 1 to 4 substituents selected from F, Cl, Br, or I. "halogen-substituted" is simply referred to as "halo".
"alkyl" refers to a substituted or unsubstituted straight or branched chain saturated aliphatic hydrocarbon group, including, but not limited to, alkyl of 1 to 20 carbon atoms, alkyl of 1 to 8 carbon atoms, alkyl of 1 to 6 carbon atoms, alkyl of 1 to 4 carbon atoms. Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, neo-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, and various branched isomers thereof; alkyl groups, as used herein, are defined in accordance with the present definition. The alkyl group may be monovalent, divalent, trivalent or tetravalent.
"hydrocarbyl" refers to a substituted or unsubstituted, straight or branched chain, saturated or unsaturated group consisting of carbon and hydrogen atoms. The hydrocarbyl group may be monovalent, divalent, trivalent, or tetravalent.
"heteroalkyl" means that 1 or more (including but not limited to 2,3, 4,5, or 6) carbon atoms in a substituted or unsubstituted alkyl group are replaced with a heteroatom (including but not limited to N, O or S). Non-limiting examples include-X (CH)2)v-X(CH2)v-X(CH2) v-H (v is an integer from 1 to 5, X is independently selected from a bond or a heteroatomHeteroatoms include, but are not limited to N, O or S, and at least 1X is selected from the heteroatom, and either N or S in the heteroatom can be oxidized to various oxidation states). The heteroalkyl group may be monovalent, divalent, trivalent, or tetravalent.
"alkylene" refers to a substituted or unsubstituted, straight and branched chain, divalent saturated hydrocarbon radical, including- (CH)2)v- (v is an integer of 1 to 10), examples of the alkylene group include, but are not limited to, methylene, ethylene, propylene, butylene, and the like.
"Heteroalkylidene" means a substituted or unsubstituted alkylene group in which 1 or more (including but not limited to 2,3, 4,5, or 6) carbon atoms are replaced with a heteroatom (including but not limited to N, O or S). Non-limiting examples include-X (CH)2)v-X(CH2)v-X(CH2) v-, v is an integer from 1 to 5, each X is independently selected from the group consisting of a bond, N, O or S, and at least 1X is selected from the group consisting of N, O or S.
"cycloalkyl" refers to a substituted or unsubstituted saturated carbocyclic hydrocarbon group, typically having from 3 to 10 carbon atoms, non-limiting examples including cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl, and the like. Cycloalkyl as found herein, is as defined above. The cycloalkyl group may be monovalent, divalent, trivalent or tetravalent.
"heterocycloalkyl" refers to a substituted or unsubstituted saturated heteroatom-containing cyclic hydrocarbon group, including but not limited to 3 to 10 atoms, 3 to 8 atoms, containing 1 to 3 heteroatoms selected from N, O or S, optionally substituted N, S in the ring of the heterocycloalkyl can be oxidized to various oxidation states. Heterocycloalkyl groups may be attached to a heteroatom or carbon atom, heterocycloalkyl groups may be attached to an aromatic ring or to a non-aromatic ring, heterocycloalkyl groups may be attached to a bridged or spiro ring, non-limiting examples include oxiranyl, aziridinyl, oxetanyl, azetidinyl, tetrahydrofuryl, tetrahydro-2H-pyranyl, dioxolanyl, dioxanyl, pyrrolidinyl, piperidinyl, imidazolidinyl, oxazolidinyl, oxazinanyl, morpholinyl, hexahydropyrimidyl, piperazinyl. The heterocycloalkyl group can be monovalent, divalent, trivalent, or tetravalent.
"alkenyl" means a substituted or unsubstituted straight and branched chain unsaturated hydrocarbon group having at least 1, and typically 1,2 or 3 carbon double bonds, the backbone including, but not limited to, 2 to 10, 2 to 6 or 2 to 4 carbon atoms, examples of alkenyl include, but are not limited to, vinyl, allyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1-methyl-1-butenyl, 2-methyl-3-butenyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 1-methyl-1-pentenyl, 2-methyl-1-pentenyl, 1-heptenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 1-octenyl, 3-octenyl, 1-nonenyl, 3-nonenyl, 1-decenyl, 4-decenyl, 1, 3-butadiene, 1, 3-pentadiene, 1, 4-hexadiene, and the like; alkenyl groups are present herein, the definition of which is consistent with the present definition. The alkenyl group may be monovalent, divalent, trivalent or tetravalent.
"alkynyl" refers to substituted or unsubstituted, straight and branched chain, monovalent unsaturated hydrocarbon radicals having at least 1, and typically 1,2 or 3 carbon-carbon triple bonds, including, but not limited to, 2 to 10 carbon atoms, 2 to 6 carbon atoms, 2 to 4 carbon atoms in the backbone, and examples of alkynyl include, but are not limited to, ethynyl, propargyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1-pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1-methyl-1-butynyl, 2-methyl-3-butynyl, 1-hexynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, 2-hexynyl, 5-hexynyl, 1-methyl-1-pentynyl, 2-methyl-1-pentynyl, 1-heptynyl, 2-heptynyl, 3-heptynyl, 4-heptynyl, 1-octynyl, 3-octynyl, 1-nonynyl, 3-nonynyl, 1-decynyl, 4-decynyl and the like; alkynyl groups can be monovalent, divalent, trivalent or tetravalent.
"alkoxy" means a substituted or unsubstituted-O-alkyl group. Non-limiting examples include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, n-hexoxy, cyclopropoxy, and cyclobutoxy.
"Carbocyclyl group"Or "carbocyclic ring" refers to a substituted or unsubstituted saturated or unsaturated aromatic or non-aromatic ring, which may be a 3-to 8-membered monocyclic, 4-to 12-membered bicyclic, or 10-to 15-membered tricyclic ring system, to which the carbocyclic group may be attached, optionally monocyclic, bridged, or spiro. Non-limiting examples include cyclopropane, cyclobutane, cyclopentane, cyclohexane, cycloheptane, 1-cyclopentyl-1-alkenyl, 1-cyclopentyl-2-alkenyl, 1-cyclopentyl-3-alkenyl, cyclohexyl, 1-cyclohexyl-2-alkenyl, 1-cyclohexyl-3-alkenyl, cyclohexenyl, benzene ring, naphthalene ring, and mixtures thereof,
Figure BDA0003178773240000621
"carbocyclyl" or "carbocycle" may be monovalent, divalent, trivalent or tetravalent.
"heterocyclyl" or "heterocycle" refers to a substituted or unsubstituted saturated or unsaturated aromatic or non-aromatic ring which may be a 3-to 8-membered monocyclic, 4-to 12-membered bicyclic, or 10-to 15-membered tricyclic ring system and contain 1 or more (including but not limited to 2,3, 4, or 5) heteroatoms selected from N, O or S, optionally substituted N, S of the heterocyclyl ring being oxidizable to various oxidation states. The heterocyclic group may be attached to a heteroatom or carbon atom, the heterocyclic group may be attached to an aromatic ring or a non-aromatic ring, the heterocyclic group may be attached to a bridged or spiro ring, non-limiting examples of which include oxiranyl, aziridinyl, oxetanyl, azetidinyl, 1, 3-dioxolanyl, 1, 4-dioxolanyl, 1, 3-dioxanyl, azepinyl, pyridyl, furyl, thienyl, pyranyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, piperidinyl, morpholinyl, thiomorpholinyl, 1, 3-dithiayl, dihydrofuranyl, dihydropyranyl, dithiapentyl, tetrahydrofuranyl, tetrahydropyrrolyl, tetrahydroimidazolyl, tetrahydrothiazolyl, tetrahydropyranyl, benzimidazolyl, benzopyridyl, pyrrolopyridyl, dihydropyranyl, spirocyclo-pyridyl, spiro-pyridyl, spirocyclo-pyridyl, oxacycloheptyl, azanyl, pyridyl, oxathianyl, thianyl, thienyl, pyridyl, etc, Benzodihydrofuranyl, pyrrolyl, pyrazolyl, thiazolyl, oxazolyl, pyrazinyl, indazolyl, benzothienyl, benzofuranyl, benzopyrinylPyrrolyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, benzopyridyl, benzopyrimidinyl, benzopyrazinyl, piperazinyl, azabicyclo [3.2.1]Octyl, azabicyclo [5.2.0 ] groups]Nonoalkyl oxatricyclo [5.3.1.1 ]]Dodecyl, azaadamantyl, oxaspiro [3.3 ]]A heptylalkyl group,
Figure BDA0003178773240000622
Figure BDA0003178773240000623
"Heterocyclyl" or "heterocycle" may be monovalent, divalent, trivalent or tetravalent.
"spiro" or "spirocyclic" refers to a polycyclic group that shares a single atom (referred to as a spiro atom) between substituted or unsubstituted monocyclic rings, the number of ring atoms in the spiro system including, but not limited to, 5 to 20, 6 to 14, 6 to 12, 6 to 10, wherein one or more of the rings may contain 0 or more (including but not limited to 1,2,3 or 4) double bonds, and optionally may contain 0 to 5 rings selected from N, O or S (═ O)nA heteroatom of (a).
Figure BDA0003178773240000631
The "spiro" or "spirocyclic group" may be monovalent, divalent, trivalent, or tetravalent.
"fused ring" or "fused ring group" refers to a polycyclic group in which each ring in the system shares an adjacent pair of atoms with other rings in the system, wherein one or more rings may contain 0 or more (including but not limited to 1,2,3, or 4) double bonds, and may be substituted or unsubstituted, and each ring in the fused ring system may contain 0 to 5 heteroatoms or heteroatom-containing groups (including but not limited to those selected from N, S (═ O)nOr O, n is 0, 1 or 2). The number of ring atoms in the fused ring system includes, but is not limited to, 5 to 20, 5 to 14, 5 to 12, 5 to 10. Non-limiting examples include:
Figure BDA0003178773240000632
Figure BDA0003178773240000633
the "fused ring" or "fused ring group" may be monovalent, divalent, trivalent or tetravalent.
"bridged ring" or "bridged ring group" refers to a substituted or unsubstituted polycyclic group containing any two atoms not directly linked, which may contain 0 or more double bonds, and any ring in the ring system may contain 0 to 5 heteroatoms or heteroatom-containing groups (including but not limited to N, S (═ O)nOr O, wherein n is 0, 1, 2). The number of ring atoms includes, but is not limited to, 5 to 20, 5 to 14, 5 to 12, or 5 to 10. Non-limiting examples include
Figure BDA0003178773240000641
Figure BDA0003178773240000642
Cubane and adamantane. The "bridge ring" or "bridge ring group" may be monovalent, divalent, trivalent or tetravalent.
"carbocyclyl", "spirocarbocyclyl" or "carbospirocyclic" refers to a "spiro" ring whose ring system consists of only carbon atoms. The "carbospiro", "spirocyclic carbocyclyl", "spirocarbocyclyl" or "carbospiro" group, as referred to herein, is defined in accordance with the definition of spiro.
"carbocyclic", "fused carbocyclic", or "carbocyclic" refers to "fused rings" in which the ring system consists of only carbon atoms. "carbocyclic ring", "fused carbocyclic ring group", "fused carbocyclic group", or "fused carbocyclic group" as used herein, is defined in accordance with fused rings.
"Carbobridged ring", "bridged carbocyclyl" or "carbocyclyl" refers to a "bridged ring" in which the ring system consists of only carbon atoms. "Carbobridged ring", "bridged carbocyclyl", or "carbobridyclyl" as used herein, is defined in accordance with the definition of bridged ring.
"Heteromonocyclic", "monocyclic heterocyclyl" or "heteromonocyclic" refers to "heterocyclyl" or "heterocycle" of a monocyclic ring system, and heterocyclyl, "monocyclic heterocyclyl" or "heteromonocyclic" as found herein, is defined consistent with the definition of heterocycle.
"Heterocyclo", "heterocyclocyclyl" or "heterocyclocyclyl" means a "fused ring" containing heteroatoms. The term "fused ring" as used herein refers to a "fused ring," fused ring group, "fused heterocyclic group," or "fused ring group," which is defined as being fused.
"Heterospirocyclic", "heterospirocyclic", "spiroheterocyclic" or "heterospirocyclic" refers to "spirocyclic" containing heteroatoms. The definition of heterospiro, "heterospiro ring", "spiro heterocyclic" or "heterospiro ring" as used herein is consistent with spiro rings.
"heterobridged ring," "heterobridged ring group," "bridged heterocyclic group," or "heterobridged ring group" refers to a "bridged ring" containing a heteroatom. The term "heterobridged ring", "heterobridged ring group", "bridged heterocyclic group" or "heterobridged ring group", as used herein, is defined in accordance with the bridged ring.
"aryl" or "aromatic ring" refers to a substituted or unsubstituted aromatic hydrocarbon group having a single or fused ring, the number of ring atoms in the aromatic ring including, but not limited to, 6 to 18, 6 to 12, or 6 to 10 carbon atoms. The aryl ring may be fused to a saturated or unsaturated carbocyclic or heterocyclic ring in which the ring to which the parent structure is attached is an aryl ring, non-limiting examples of which include a benzene ring, a naphthalene ring, a substituted naphthalene group, a substituted or substituted naphthalene group, a substituted benzene or substituted benzene ring, a substituted benzene or substituted benzene ring, a substituted benzene ring, or substituted benzene ring, a substituted benzene ring, or substituted benzene ring, a substituted benzene or substituted benzene ring, a substituted benzene ring, or substituted benzene ring, a substituted benzene ring, or substituted benzene ring, a substituted benzene,
Figure BDA0003178773240000651
The "aryl" or "aromatic ring" may be monovalent, divalent, trivalent, or tetravalent. When divalent, trivalent, or tetravalent, the attachment site is located on the aryl ring.
"heteroaryl" or "heteroaromatic ring" refers to a substituted or unsubstituted aromatic hydrocarbon group and contains from 1 to 5 selected heteroatoms or heteroatom-containing groups (including but not limited to N, O or S (═ O) n, n is 0, 1, 2), and the number of ring atoms in the heteroaromatic ring includes but is not limited to 5 to 15, 5 to 10, or 5 to 6. Non-limiting examples of heteroaryl groups include, but are not limited to, pyridyl, furyl, thienyl, pyridyl, pyranyl, N-alkylpyrrolyl, pyrimidinyl, pyrryl, pyridyl, substituted pyridyl, substituted pyridyl, and the like,Pyrazinyl, pyridazinyl, imidazolyl, benzopyrazolyl, benzimidazole, benzopyridine, pyrrolopyridine, and the like. The heteroaryl ring may be fused to a saturated or unsaturated carbocyclic or heterocyclic ring in which the ring to which the parent structure is attached is a heteroaryl ring, non-limiting examples of which include
Figure BDA0003178773240000652
Heteroaryl, as used herein, is defined in accordance with the present definition. Heteroaryl groups can be monovalent, divalent, trivalent, or tetravalent. When divalent, trivalent, or tetravalent, the attachment site is on the heteroaryl ring.
The "5-membered and 5-membered heteroaromatic ring" means a 5-and 5-membered fused heteroaromatic ring in which at least 1 ring of 2 fused rings contains 1 or more heteroatoms (including but not limited to O, S or N), and the whole group has aromaticity, and non-limiting examples include pyrrolopyrrole rings, pyrazolopyrrole rings, pyrazolopyrazole rings, pyrrolofuran rings, pyrazolofuran rings, pyrrolothiophene rings, pyrazolothiophene rings.
"5 and 6 membered heteroaromatic ring" means a 5 and 6 membered fused heteroaromatic ring wherein at least 1 ring of the 2 fused rings contains more than 1 heteroatom (including but not limited to O, S or N) and the entire group is aromatic, non-limiting examples include benzo 5 membered heteroaryl, 6 membered heteroaromatic ring and 5 membered heteroaromatic ring.
"substituted" or "substituted" means substituted with 1 or more (including but not limited to 2,3, 4, or 5) substituents including but not limited to H, F, Cl, Br, I, alkyl, cycloalkyl, alkoxy, haloalkyl, thiol, hydroxy, nitro, mercapto, amino, cyano, isocyano, aryl, heteroaryl, heterocyclyl, bridged ring, spiro ring, fused ring, hydroxyalkyl, ═ O, carbonyl, aldehyde, carboxylic acid, formate, - (CH)2)m-C(=O)-Ra、-O-(CH2)m-C(=O)-Ra、-(CH2)m-C(=O)-NRbRc、-(CH2)mS(=O)nRa、-(CH2)m-alkenyl-Ra、ORdOr- (CH)2)m-alkynesradical-Ra(wherein m, n are 0, 1 or 2), arylthio, thiocarbonyl, silyl or-NRbRcEtc. wherein R isbAnd RcIndependently selected from the group consisting of H, hydroxy, amino, carbonyl, alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, sulfonyl, trifluoromethanesulfonyl, and optionally, RbAnd RcFive or six membered cycloalkyl or heterocyclyl groups may be formed.
"contains 1 to 5 heteroatoms selected from O, S, N" means containing 1,2,3, 4 or 5 heteroatoms selected from O, S, N.
"0 to X substituents selected from …" means substituted with 0, 1,2,3 … X substituents selected from …, X being selected from any integer between 1 and 10. If "substituted with 0 to 4 substituents selected from …" is intended to mean substituted with 0, 1,2,3, or 4 substituents selected from …. If "substituted with 0 to 5 substituents selected from …" is meant substituted with 0, 1,2,3, 4, or 5 substituents selected from …. By "heterobridged ring is optionally further substituted with 0 to 4 substituents selected from H or F" is meant that the heterobridged ring is optionally further substituted with 0, 1,2,3 or 4 substituents selected from H or F.
The ring of X-Y element (X is selected from the integer less than or equal to Y and greater than or equal to 3, and Y is selected from any integer between 4 and 12) includes the ring of X +1, X +2, X +3, X +4 … Y element. Rings include heterocyclic, carbocyclic, aromatic, aryl, heteroaryl, cycloalkyl, heteromonocyclic, heterospirocyclic, or heterobridged rings. For example, "4-7 membered heteromonocyclic" refers to a 4-, 5-, 6-, or 7-membered heteromonocyclic ring, and "5-10 membered heterobicyclic ring" refers to a 5-, 6-, 7-, 8-, 9-, or 10-membered heterobicyclic ring.
"optional" or "optionally" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs or does not. Such as: "alkyl optionally substituted with F" means that the alkyl group may, but need not, be substituted with F, and the description includes the case where the alkyl group is substituted with F and the case where the alkyl group is not substituted with F.
By "pharmaceutically acceptable salt" or "pharmaceutically acceptable salt thereof" is meant a salt of a compound of the invention that retains the biological effectiveness and properties of the free acid or free base obtained by reaction with a non-toxic inorganic or organic base, and the free base obtained by reaction with a non-toxic inorganic or organic acid.
"pharmaceutical composition" refers to a mixture of one or more compounds of the present invention, or stereoisomers, tautomers, deuteroides, solvates, prodrugs, metabolites, pharmaceutically acceptable salts or co-crystals thereof, and other chemical components, wherein "other chemical components" refers to pharmaceutically acceptable carriers, excipients, and/or one or more other therapeutic agents.
By "carrier" is meant a material that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
"excipient" refers to an inert substance added to a pharmaceutical composition to facilitate administration of a compound. Non-limiting examples include calcium carbonate, calcium phosphate, sugars, starches, cellulose derivatives (including microcrystalline cellulose), gelatin, vegetable oils, polyethylene glycols, diluents, granulating agents, lubricants, binders, and disintegrating agents.
By "prodrug" is meant a compound of the invention that is metabolically convertible in vivo to a biologically active compound. Prodrugs of the invention are prepared by modifying an amino or carboxyl group in a compound of the invention, which modification may be removed by routine manipulation or in vivo, to yield the parent compound. When a prodrug of the present invention is administered to a mammalian subject, the prodrug is cleaved to form a free amino or carboxyl group.
"cocrystal" refers to a crystal of an Active Pharmaceutical Ingredient (API) and a cocrystal former (CCF) bound by hydrogen bonding or other non-covalent bonds, wherein the API and CCF are both solid in their pure state at room temperature and a fixed stoichiometric ratio exists between the components. A co-crystal is a multi-component crystal that contains both a binary co-crystal formed between two neutral solids and a multicomponent co-crystal formed between a neutral solid and a salt or solvate.
"animal" is meant to include mammals, such as humans, companion animals, zoo animals, and livestock, preferably humans, horses, or dogs.
"stereoisomers" refers to isomers resulting from the different arrangement of atoms in a molecule, including cis, trans isomers, enantiomers and conformational isomers.
"tautomer" refers to functional group isomers resulting from rapid movement of an atom in two positions in a molecule, such as keto-enol isomers and amide-imino-enol isomers, and the like.
"optional" or "optionally" or "selective" or "selectively" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "a heterocyclic group optionally substituted with an alkyl group" means that the alkyl group may, but need not, be present, and the description includes the case where the heterocyclic group is substituted with an alkyl group, and the case where the heterocyclic group is not substituted with an alkyl group.
“IC50"is the concentration of drug or inhibitor required to inhibit half of a given biological process (or some component of the process such as an enzyme, receptor, cell, etc.).
Detailed Description
To accomplish the objects of the present invention, the compounds "commercially available chemicals" used in the reactions described herein are prepared from standard commercial sources including Shanghai Aladdin Biotechnology GmbH, Shanghai Merlin Biotechnology GmbH, Sigma-Aldrich, Afahesa (China) Chemicals GmbH, echiei (Shanghai) chemical developments GmbH, Annagi chemistry, Shanghai Tantaceae technology GmbH, Corlon Chemicals, Bailinguo science GmbH, and the like, starting from commercially available chemicals and/or compounds described in the chemical literature according to organic synthesis techniques known to those skilled in the art.
References and monographs in this field detail the synthesis of reactants useful in the preparation of the compounds described herein, or provide articles describing the preparation for reference. These references and monographs include: "Synthetic Organic Chemistry", John Wiley & Sons, Inc., New York; sandler et al, "Organic Functional Group precursors," 2nd ed, Academic Press, New York, 1983; h.o.house, "Modern Synthetic Reactions", 2nd ed., w.a.benjamin, inc.menlo Park, calif.1972; gilchrist, "Heterocyclic Chemistry", 2nd Ed., John Wiley & Sons, New York, 1992; march, "Advanced Organic Chemistry: Reactions, mechanics and Structure", 4th Ed., Wiley Interscience, New York, 1992; fuhrhop, J.and Penzlin G. "Organic Synthesis: hubs, Methods, Starting Materials", Second, Revised and Enlarged Edition (1994) John Wiley & Sons ISBN: 3527-29074-5; hoffman, R.V. "Organic Chemistry, An Intermediate Text" (1996) Oxford University Press, ISBN 0-19-509618-5; larock, R.C. "Comprehensive Organic Transformations: A Guide to Functional Group Preparations" 2nd Edition (1999) Wiley-VCH, ISBN: 0-471-; march, J. "Advanced Organic Chemistry: Reactions, mechanics, and Structure" 4th Edition (1992) John Wiley & Sons, ISBN: 0-471-; otera, J. (editor) "Modern carbon Chemistry" (2000) Wiley-VCH, ISBN: 3-527-; patai, S. "Patai's 1992Guide to the Chemistry of Functional Groups" (1992) Interscience ISBN: 0-471-; solomons, T.W.G. "Organic Chemistry" 7th Edition (2000) John Wiley & Sons, ISBN: 0-471-; stowell, J.C., "Intermediate Organic Chemistry" 2nd Edition (1993) Wiley-Interscience, ISBN: 0-471-; "Industrial Organic Chemicals: Starting Materials and Intermediates: An Ullmann's Encyclopedia" (1999) John Wiley & Sons, ISBN: 3-527-; "Organic Reactions" (1942-2000) John Wiley & Sons, in over 55 volumes; and "Chemistry of Functional Groups" John Wiley & Sons, in 73volumes.
Specific and similar reactants can be selectively identified by an index of known chemicals prepared by the chemical abstracts society of america, which is available in most public and university libraries and online. Chemicals that are known but not commercially available in catalogs are optionally prepared by custom chemical synthesis plants, many of which standard chemical supply plants (e.g., those listed above) provide custom synthesis services. References to the preparation and selection of pharmaceutically acceptable Salts of the compounds described herein are P.H.Stahl & C.G.Wermuth "Handbook of Pharmaceutical Salts", Verlag Helvetica Chimica Acta, Zurich,2002.
The compounds of the present invention can also be synthesized by any of the following methods:
the first synthesis method comprises the following steps:
Figure BDA0003178773240000681
obtaining a corresponding general formula (A-2) by the nucleophilic substitution reaction or the coupling reaction of the general formula (A-1) and the general formula (Z-2), and obtaining a corresponding general formula (A-4) by the coupling reaction or the substitution reaction of the general formula (A-2) and the general formula (A-3);
and a second synthesis method comprises the following steps:
Figure BDA0003178773240000691
the general formula (Z-1) and the general formula (Z-2) are subjected to reductive amination, nucleophilic substitution reaction or coupling reaction to obtain a corresponding general formula (Z-3), and the general formula (Z-3) is subjected to protecting group removal to obtain a compound of a general formula (Z-3'); the general formula (Z-3') and the general formula (Z-4) are subjected to nucleophilic substitution reaction or coupling reaction to obtain a corresponding general formula (Ia);
the general formula (Z-3 ') and the general formula (Z-5) are subjected to reductive amination, nucleophilic substitution reaction or coupling reaction to obtain a corresponding general formula (Z-6), and the general formula (Z-6) is subjected to protecting group removal to obtain a compound of the general formula (Z-6'); the general formula (Z-6') and the general formula (Z-4) are subjected to nucleophilic substitution reaction or coupling reaction to obtain a corresponding general formula (Ib);
the general formula (Z-6 ') and the general formula (Z-7) are subjected to reductive amination, nucleophilic substitution reaction or coupling reaction to obtain a corresponding general formula (Z-8), and the general formula (Z-8) is subjected to protecting group removal to obtain a compound of the general formula (Z-8'); the general formula (Z-8') and the general formula (Z-4) are subjected to nucleophilic substitution reaction or coupling reaction to obtain the corresponding general formula (Ic);
the general formula (Z-8 ') and the general formula (Z-9) are subjected to reductive amination, nucleophilic substitution reaction or coupling reaction to obtain a corresponding general formula (Z-10), the general formula (Z-10) is subjected to protecting group removal to obtain a compound of the general formula (Z-10 '), and the general formula (Z-10 ') is subjected to nucleophilic substitution reaction or coupling reaction with the general formula (Z-4) to obtain a corresponding general formula (Id);
R1、R4、R6、R8each independently selected from (═ O), -CHO, F, Cl, Br, I, OTf, CH2Cl、CH2Br、CH2I、CH2OTf、CH2OMs、CH2OTs;
R2、R5、R7、R9Each independently selected from H, (═ O), -CHO, F, Cl, Br, I or an amino protecting group, preferably Boc;
R3、R10selected from the group consisting of independently H, NH2、F、Cl、Br、I、OTf、OH;
X and R11Each independently selected from F, Cl, Br, I, OTf, halogenated magnesium, B (OH)2Borate, alkyl-substituted tin, zinc halide, copper halide, and the like.
The following examples illustrate the technical solutions of the present invention in detail, but the scope of the present invention includes but is not limited thereto.
The compounds used in the reactions described herein are prepared according to organic synthesis techniques known to those skilled in the art, starting from compounds described in the commercial chemicals and/or chemical literature. "commercial chemicals" are obtained from regular commercial sources, and suppliers include: tatan science and technology, Annaiji chemistry, Shanghai Demer, Chengdong chemical engineering, Shaoshao chemical technology, Nanjing Yashi, Yaogongkang and Bailingwei science and technology.
The structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) or (and) Mass Spectrometry (MS). NMR shift (. delta.) of 10-6The units in (ppm) are given. NMR was measured using (Bruker Avance III 400 and Bruker Avance 300) nuclear magnetic spectrometers in deuterated dimethyl sulfoxide (DMSO-d)6) Deuterated chloroform (CDCl)3) Deuterated methanol (CD)3OD), internal standard Tetramethylsilane (TMS);
MS measurement (Agilent 6120B (ESI)) and Agilent 6120B (APCI));
HPLC was carried out using an Agilent 1260DAD high pressure liquid chromatograph (Zorbax SB-C18100X 4.6mm, 3.5. mu.M);
the thin layer chromatography silica gel plate adopts HSGF254 of tobacco yellow sea or GF254 of Qingdao, the specification of silica gel plate used by Thin Layer Chromatography (TLC) is 0.15mm-0.20mm, and the specification of thin layer chromatography separation and purification product is 0.4mm-0.5 mm;
the column chromatography generally uses 200-mesh and 300-mesh silica gel of the Tibet yellow sea silica gel as a carrier.
SEM:
Figure BDA0003178773240000701
THP:
Figure BDA0003178773240000702
Boc: a tert-butoxycarbonyl group; ms:
Figure BDA0003178773240000703
TBS:
Figure BDA0003178773240000704
MTBE methyl tert-butyl ether; bn:
Figure BDA0003178773240000705
DIPEA: n, N-diisopropylethylamine; DMAc: n, N-dimethylacetamide; DMSO, DMSO: dimethyl sulfoxide; DCM: dichloromethane; cbz:
Figure BDA0003178773240000706
NMP: n-methyl pyrrolidone; tf: a trifluoromethanesulfonyl group; ts: a p-toluenesulfonyl group; TMS: trimethylsilyl; DME: ethylene glycol dimethyl ether; DCM: dichloromethane; DCE: 1, 2-dichloroethane; pd2dba3: tris (dibenzylidene) acetone dipalladium; john Phos: (2-biphenyl) di-tert-butylphosphine; THF: tetrahydrofuran; the DIAD: diisopropyl azodicarboxylate; CDI: n, N' -carbonyldiimidazole; MsCl: methanesulfonyl chloride; TFA: trifluoroacetic acid.
Examples
Example 1
5-amino-1- (1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxo-2, 3-dihydro-1H-isoindol-5-yl) piperidin-4-yl) -3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamide ] methyl } phenyl) -1H-pyrazole-4-carboxamide (Compound 1)
5-amino-1-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl)piperidin-4-yl)-3-(4-{[(5-fluoro-2-methoxyphenyl)formamido]methyl}phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000711
The first step is as follows: 4- (Methanesulfonyloxy) piperidine-1-carboxylic acid tert-butyl ester (1b)
tert-butyl 4-(methanesulfonyloxy)piperidine-1-carboxylate
Figure BDA0003178773240000712
Tert-butyl 4-hydroxypiperidine-1-carboxylate 1a (5.00g, 24.84mmol) and triethylamine (3.77g, 37.26mmol) were dissolved in 30mL of dichloromethane, and methanesulfonyl chloride (3.41g, 29.81mmol) was added dropwise in an ice-water bath, and after completion of the addition, the mixture was naturally returned to room temperature and stirred for 1 hour. After the reaction was completed, 50mL of water was added for extraction. The organic phase was dried over anhydrous sodium sulfate and the crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether (v/v) ═ 1:10) and concentrated to give the title compound as a yellow oil (1b) (6.5g, 93.66% yield).
The second step is that: (4- { [ (5-fluoro-2-methoxyphenyl) carboxamido ] methyl } phenyl) boronic acid (1d)
(4-{[(5-fluoro-2-methoxyphenyl)formamido]methyl}phenyl)boronic acid
Figure BDA0003178773240000721
The compound 5-fluoro-2-methoxybenzoic acid 1c (0.71g,4.10mmol) and 4-aminomethylphenylboronic acid hydrochloride (0.92g,4.92mmol), N-methylimidazole (0.84g,10.25mmol) were added to 15mL of dry DMF, TCFH (2.87g,10.25mmol) was added in portions under ice-water bath, and the mixture was allowed to naturally warm to room temperature and stirred for 1 hour. The reaction solution was quenched with water (50mL), extracted with EA ((20mL × 3), the organic phases were combined, washed with saturated brine (20mL × 3), dried over anhydrous sodium sulfate, filtered, the solvent was removed from the filtrate under reduced pressure, and the residue was separated by column chromatography (PET: EA ═ 1:1) to give the objective compound 1d (0.94g, yield: 74.89%).
MS m/z=304.1[M+H]+
The third step: 5-amino-3-bromo-1H-pyrazole-4-carbonitrile (1f)
5-amino-3-bromo-1H-pyrazole-4-carbonitrile
Figure BDA0003178773240000722
The compound 5-amino-1H-pyrazole-4-carbonitrile (1e) (1.0g,9.25mmol) was dissolved in 15mL of dry DMF, NBS (2.47g,13.88mmol) was added portionwise at room temperature, and after the addition was completed, the mixture was stirred at room temperature for 3 hours. The reaction solution was quenched with water (50mL), extracted with EA ((20mL × 3), the organic phases were combined, washed with saturated brine (20mL × 3), dried over anhydrous sodium sulfate, filtered, the solvent was removed from the filtrate under reduced pressure, and the residue was separated by column chromatography (PET: EA ═ 2:1) to give the objective compound (1f) (1.30g, yield: 75.15%).
MS m/z=187.0,189.1[M+H]+
The fourth step: 4- (5-amino-3-bromo-4-cyano-1H-pyrazol-1-yl) piperidine-1-carboxylic acid tert-butyl ester (1g)
tert-butyl 4-(5-amino-3-bromo-4-cyano-1H-pyrazol-1-yl)piperidine-1-carboxylate
Figure BDA0003178773240000723
The compound 5-amino-3-bromo-1H-pyrazole-4-carbonitrile (1f) (1.0g,5.35mmol), tert-butyl 4- (methanesulfonyloxy) piperidine-1-carboxylate (1b) (1.79g,6.42mmol) and cesium carbonate (2.61g,8.02mmol) were dissolved in 15mL of dry DMF and stirred at room temperature overnight. The reaction solution was quenched with water (50mL), extracted with EA ((20mL × 3), the organic phases were combined, washed with saturated brine (20mL × 3), dried over anhydrous sodium sulfate, filtered, the solvent was removed from the filtrate under reduced pressure, and the residue was separated by column chromatography (PET: EA ═ 2:1) to obtain 1g (0.90g, yield: 45.46%) of the objective compound.
MS m/z=370.1,372.2[M+H]+
The fifth step: 4- (5-amino-4-cyano-3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamido ] methyl } phenyl) -1H-pyrazol-1-yl) piperidine-1-carboxylic acid tert-butyl ester (1H)
tert-butyl 4-(5-amino-4-cyano-3-(4-{[(5-fluoro-2-methoxyphenyl)formamido]methyl}phenyl)-1H-pyrazol-1-yl)piperidine-1-carboxylate
Figure BDA0003178773240000731
The compound tert-butyl 4- (5-amino-3-bromo-4-cyano-1H-pyrazol-1-yl) piperidine-1-carboxylate (1g) (0.50g,1.35mmol), (4- (5-fluoro-2-methoxybenzamide) methyl) phenyl) boronic acid (1d) (0.53g,1.76mmol), cesium carbonate (0.92g,2.82mmol), Pd (dppf) Cl2(103.4mg,0.14mmol) was added to a mixed solvent of 10mL dioxane and water (v/v. sub.5: 1), and the mixture was replaced with nitrogen 3 times, heated to 110 ℃ and reacted for 3 hours. The reaction mixture was quenched with water (50mL), EA extracted ((20 mL. times.3), the combined organic phases were washed with saturated brine (20 mL. times.3), dried over anhydrous sodium sulfate, filtered, the solvent was removed from the filtrate under reduced pressure, and the residue was separated by column chromatography (DCM: CH)3OH ═ 10:1) gave the title compound (1h) (0.42g, yield: 56.69%).
MS m/z=549.1[M+1]+
And a sixth step: 5-amino-3- (4- [ (5-fluoro-2-methoxyphenyl) carboxamido ] methyl } phenyl) -1- (piperidin-4-yl) -1H-pyrazole-4-carboxamide (1i)
5-amino-3-(4-{[(5-fluoro-2-methoxyphenyl)formamido]methyl}phenyl)-1-(piperidin-4-yl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000732
The compound 4- (5-amino-4-cyano-3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamido)]Methyl } phenyl) -1H-pyrazol-1-yl) piperidine-1-carboxylic acid tert-butyl ester (1H)) (100mg,0.18mmoL) was dissolved in 5mL of sulfuric acid (90%), heated to 100 ℃ and reacted for 1 hour, the reaction solution was cooled to room temperature, PH was adjusted to 10 in an ice-water bath, EA extraction ((20mL × 3), organic phases were combined, saturated brine washing (20mL × 3), drying over anhydrous sodium sulfate, filtration, solvent removal from the filtrate under reduced pressure, and column chromatography of the residue was performed (DCM: CH)3OH ═ 10:1) to obtain the objective compound (1i) (0.06g, yield: 70.5%).
MS m/z=467.2[M+H]+
The seventh step: 5-amino-1- (1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxo-2, 3-dihydro-1H-isoindol-5-yl) piperidin-4-yl) -3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamide ] methyl } phenyl) -1H-pyrazole-4-carboxamide (Compound 1)
5-amino-1-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl)piperidin-4-yl)-3-(4-{[(5-fluoro-2-methoxyphenyl)formamido]methyl}phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000741
Reacting the compound 5-amino-3- (4- [ (5-fluoro-2-methoxyphenyl) formamido]Methyl } phenyl) -1- (piperidin-4-yl) -1H-pyrazole-4-carboxamide (1i) (60.0mg,0.13mmol), 2- (2, 6-dioxo-3-piperidyl) -5-fluoroisoindoline-1, 3-dione (69.0mg, 0.25mmol) were dissolved in 2mL of DMSO, N-diisopropylethylamine (49.0mg, 0.38mmol) was added, and after completion of the addition, the reaction was carried out at 90 ℃ for 2 hours. After the reaction, the reaction mixture was cooled to room temperature, 10mL of water was added, the mixture was filtered, the filter cake was dissolved in 20mL of dichloromethane, the solution was washed with 5mL of saturated sodium chloride, dried over anhydrous sodium sulfate, and the crude product was concentrated under reduced pressure and subjected to column chromatography (DCM: CH)3OH ═ 10:1) gave the title compound 1 as a yellow solid (20.0mg, 23% yield).
MS m/z=723.2[M+1]+
1H NMR(400MHz,CD3OD)δ7.67(d,1H),7.60(dd,1H),7.51–7.44(m,4H),7.39(d,1H),7.29–7.20(m,2H),7.17–7.11(m,1H),5.05(dd,1H),4.65(s,2H),4.45–4.34(m,1H),4.24–4.15(m,2H),3.93(s,3H),3.23–3.11(m,2H),2.92–2.19(m,1H),2.78–2.67(m,2H),2.28–2.01(m,5H).
Example 2
5-amino-1- (1- (1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxo-2, 3-dihydro-1H-isoindol-5-yl) azetidin-3-yl) piperidin-4-yl) -3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamide ] methyl } phenyl) -1H-pyrazole-4-carboxamide (Compound 2)
5-amino-1-(1-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl)azetidin-3-yl)piperidin-4-yl)-3-(4-{[(5-fluoro-2-methoxyphenyl)formamido]methyl}phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000751
The first step is as follows: n- [ (4- (5-amino-4-cyano-1- (piperidin-4-yl) -1H-pyrazol-3-yl) phenyl) methyl ] -5-fluoro-2-methoxybenzamide (2a)
N-[(4-(5-amino-4-cyano-1-(piperidin-4-yl)-1H-pyrazol-3-yl)phenyl)methyl]-5-fluoro-2-methoxybenzamide
Figure BDA0003178773240000752
Tert-butyl 4- (5-amino-4-cyano-3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamido ] methyl } phenyl) -1H-pyrazol-1-yl) piperidine-1-carboxylate (1H) (0.55g,1.01mmoL) was dissolved in 20mL of HCl/dioxane, reacted at room temperature for 3 hours, and the reaction solution was directly concentrated to dryness to give a crude product of the objective compound (2a) (0.41g) which was used directly in the next reaction.
MS m/z=449.2[M+H]+
The second step is that: 3- (4- (5-amino-4-cyano-3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamido ] methyl } phenyl) -1H-pyrazol-1-yl) piperidin-1-yl) azetidine-1-carboxylic acid tert-butyl ester (2b)
tert-butyl 3-(4-(5-amino-4-cyano-3-(4-{[(5-fluoro-2-methoxyphenyl)formamido]methyl}phenyl)-1H-pyrazol-1-yl)piperidin-1-yl)azetidine-1-carboxylate
Figure BDA0003178773240000761
Intermediate N- [ (4- (5-amino-4-cyano-1- (piperidin-4-yl) -1H-pyrazol-3-yl) phenyl) methyl ] -5-fluoro-2-methoxybenzamide hydrochloride (2a) (0.40g, 0.824mmol) was dissolved in 20mL of N, N-dimethylacetamide, 3-oxoazetidine-1-carboxylic acid tert-butyl ester (0.565g, 3.30mmol), anhydrous magnesium sulfate 0.5g, acetic acid 0.5mL, stirred at 70 ℃ for 1H, sodium cyanoborohydride (335mg, 5.5mmol) was added, and the reaction was continued overnight. After the reaction was completed, the reaction mixture was cooled to room temperature, and extracted with 50mL of ethyl acetate and 20mL of water. The organic phase was dried over anhydrous sodium sulfate and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1-5:1) and concentrated to give the title compound as a yellow solid (2b) (0.38g, 76.31% yield)
MS m/z=604.2[M+1]+
The third step: 5-amino-1- (1- (azetidin-3-yl) piperidin-4-yl) -3- (4- [ (5-fluoro-2-methoxyphenyl) carboxamide ] methyl } phenyl) -1H-pyrazole-4-carboxamide (2c)
5-amino-1-(1-(azetidin-3-yl)piperidin-4-yl)-3-(4-{[(5-fluoro-2-methoxyphenyl)formamido]methyl}phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000762
Tert-butyl 3- (4- (5-amino-4-cyano-3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamido ] methyl } phenyl) -1H-pyrazol-1-yl) piperidin-1-yl) azetidine-1-carboxylate (2b) (0.08g, 0.13mmol) was dissolved in 5mL sulfuric acid (90%), heated to 100 ℃ for 1 hour, the reaction was cooled to room temperature, PH was adjusted to 10 in an ice water bath, EA extraction ((20mL × 3), organic phases were combined, washed with saturated brine (20mL × 3), dried over anhydrous sodium sulfate, filtered, the solvent was removed from the filtrate under reduced pressure, and the residue was separated by column chromatography (DCM: CH3OH ═ 10:1) to give the target compound (2c) (0.05g, yield: 71.5%).
MS m/z=522.2[M+H]+
The fourth step: 5-amino-1- (1- (1- (2- (2, 6-oxopiperidin-3-yl) -1, 3-dioxo-2, 3-dihydro-1H-isoindol-5-yl) azetidin-3-yl) piperidin-4-yl) -3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamide ] methyl } phenyl) -1H-pyrazole-4-carboxamide (Compound 2)
5-amino-1-(1-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl)azetidin-3-yl)piperidin-4-yl)-3-(4-{[(5-fluoro-2-methoxyphenyl)formamido]methyl}phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000771
The compound 5-amino-1- (1- (azetidin-3-yl) piperidin-4-yl) -3- (4- [ (5-fluoro-2-methoxyphenyl) carboxamide ] methyl } phenyl) -1H-pyrazole-4-carboxamide (2c) (50.0mg,0.10mmol), 2- (2, 6-dioxo-3-piperidinyl) -5-fluoroisoindoline-1, 3-dione (69.0mg, 0.25mmol) was dissolved in 5mL of DMSO, N-diisopropylethylamine (49.0mg, 0.38mmol) was added, and after completion of the addition, reaction was carried out at 90 ℃ for 2 hours. After the reaction was completed, it was cooled to room temperature, 10mL of water was added, it was filtered, the filter cake was dissolved in 20mL of dichloromethane, it was then washed with 5mL of saturated sodium chloride, dried over anhydrous sodium sulfate, and the crude product obtained was concentrated under reduced pressure and sent to the liquid phase for preparation to obtain the title compound 2 as a yellow solid (10.0mg, yield 14%). preparation conditions: instrument and preparative column: the liquid phase was prepared using WATERS 2767, column format Xbridge C18,5 μm, 19mm x 250mm in internal diameter x length.
The preparation method comprises the following steps: the crude product was dissolved in DMF and filtered through a 0.45 μm filter to prepare a sample solution.
Mobile phase system: acetonitrile/(containing 0.05% ammonia) water. Gradient elution: the acetonitrile content is 30-75%, and the elution time is 15 min.
MS m/z=778.3[M+1]+
Example 3
5-amino-1- (1- (1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxo-2, 3-dihydro-1H-isoindol-5-yl) azetidin-3-yl) piperidin-4-yl) -3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamido ] methyl } phenyl) -1H-pyrazole-4-carboxamide (Compound 3)
5-amino-1-(1-(1-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl)azetidin-3-yl)azetidin-3-yl)piperidin-4-yl)-3-(4-{[(5-fluoro-2-methoxyphenyl)formamido]methyl}phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000781
The first step is as follows: n- [ (4- (5-amino-1- (1- (azetidin-3-yl) piperidin-4-yl) -4-cyano-1H-pyrazol-3-yl) phenyl) methyl ] -5-fluoro-2-methoxybenzamide (3a)
N-[(4-(5-amino-1-(1-(azetidin-3-yl)piperidin-4-yl)-4-cyano-1H-pyrazol-3-yl)phenyl)methyl]-5-fluoro-2-methoxybenzamide
Figure BDA0003178773240000782
The compound tert-butyl 3- (4- (5-amino-4-cyano-3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamido ] methyl } phenyl) -1H-pyrazol-1-yl) piperidin-1-yl) azetidine-1-carboxylate (2b) (0.20g,0.33mmoL) was dissolved in 20mL HCl/dioxane, reacted at room temperature for 3 hours, and the reaction solution was directly concentrated to dryness to give a crude product of the objective compound (3a) (0.18g) which was used directly in the next reaction.
MS m/z=504.2[M+H]+
The second step is that: tert-butyl 3- (3- (4- (5-amino-4-cyano-3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamido ] methyl } phenyl) -1H-pyrazol-1-yl) piperidin-1-yl) azetidine-1-carboxylate (3b)
tert-butyl 3-(3-(4-(5-amino-4-cyano-3-(4-{[(5-fluoro-2-methoxyphenyl)formamido]methyl}phenyl)-1H-pyrazol-1-yl)piperidin-1-yl)azetidin-1-yl)azetidine-1-carboxylate
Figure BDA0003178773240000791
Intermediate N- [ (4- (5-amino-1- (1- (azetidin-3-yl) piperidin-4-yl) -4-cyano-1H-pyrazol-3-yl) phenyl) methyl ] -5-fluoro-2-methoxybenzamide (3a) (0.16g, 0.33mmol) was dissolved in 20mL of N, N-dimethylacetamide, tert-butyl 3-oxoazetidine-1-carboxylate (0.285g, 1.65mmol), anhydrous magnesium sulfate 0.5g, acetic acid 0.5mL was added, stirred at 70 ℃ for 1H, sodium cyanoborohydride (335mg, 5.5mmol) was added, and the reaction was continued overnight. After the reaction was completed, the reaction mixture was cooled to room temperature, and extracted with 50mL of ethyl acetate and 20mL of water. The organic phase was dried over anhydrous sodium sulfate and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1-5:1) and concentrated to give the title compound as a yellow solid (3b) (0.08g, 36.86% yield)
MS m/z=659.3[M+1]+
The third step: 5-amino-1- (1- (1- (azetidin-3-yl) piperidin-4-yl) -3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamide ] methyl } phenyl) -1H-pyrazole-4-carboxamide (3c)
5-amino-1-(1-(1-(azetidin-3-yl)azetidin-3-yl)piperidin-4-yl)-3-(4-{[(5-fluoro-2-methoxyphenyl)formamido]methyl}phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000792
The compound tert-butyl 3- (3- (4- (5-amino-4-cyano-3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamide)]Methyl } phenyl) -1H-pyrazol-1-yl) piperidin-1-yl) azetidine-1-carboxylate (3b) (0.08g, 0.12mmol) was dissolved in 5mL sulfuric acid (90%), heated to 100 ℃ and reacted for 1 hour, the reaction solution was cooled to room temperature, PH was adjusted to 10 in an ice water bath, EA extraction ((20mL × 3), organic phases were combined, washed with saturated brine (20mL × 3), dried over anhydrous sodium sulfate, filtered, the filtrate was freed of the solvent under reduced pressure, and the residue was separated by column chromatography (DCM: CH: DCM)3OH ═ 10:1) to obtain the objective compound (3c) (0.03g, yield: 42.8%).
MS m/z=577.4[M+H]+
The fourth step: 5-amino-1- (1- (1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxo-2, 3-dihydro-1H-isoindol-5-yl) azetidin-3-yl) piperidin-4-yl) -3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamido ] methyl } phenyl) -1H-pyrazole-4-carboxamide (Compound 3)
5-amino-1-(1-(1-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-5-yl)azetidin-3-yl)azetidin-3-yl)piperidin-4-yl)-3-(4-{[(5-fluoro-2-methoxyphenyl)formamido]methyl}phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000801
The compound 5-amino-1- (1- (1- (azetidin-3-yl) piperidin-4-yl) -3- (4- { [ (5-fluoro-2-methoxyphenyl) carboxamide ] methyl } phenyl) -1H-pyrazole-4-carboxamide (3c) (30.0mg,0.05mmol), 2- (2, 6-dioxo-3-piperidinyl) -5-fluoroisoindoline-1, 3-dione (69.0mg, 0.25mmol) was dissolved in 5mL of DMSO, N-diisopropylethylamine (49.0mg, 0.38mmol) was added, and after completion of the addition, the reaction was carried out at 90 ℃ for 2 hours. After the reaction was completed, it was cooled to room temperature, 10mL of water was added, the mixture was filtered, the filter cake was dissolved in 20mL of dichloromethane, the mixture was washed with 5mL of saturated sodium chloride, dried over anhydrous sodium sulfate, and the crude product obtained was concentrated under reduced pressure and sent to the liquid phase for preparation to give the title compound 3 as a yellow solid (5.0mg, yield 11.53%)
The preparation conditions are as follows: instrument and preparative column: the liquid phase was prepared using WATERS 2767, column format Xbridge C18,5 μm, 19mm x 250mm in internal diameter x length.
The preparation method comprises the following steps: the crude product was dissolved in DMF and filtered through a 0.45 μm filter to prepare a sample solution.
Mobile phase system: acetonitrile/(containing 0.05% ammonia) water. Gradient elution: the acetonitrile content is 30-75%, and the elution time is 15 min.
MS m/z=417.2[M/2+1]+
Example 4:
5-amino-1- (1- (1- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindolin-5-yl) azacyclobutane-3-yl) methyl) azetidin-3-yl) piperidin-4-yl) -3- (4- ((5-fluoro-2-methoxybenzoyl) methyl) phenyl) -1H-pyrazole-4-carboxamide (Compound 4)
5-amino-1-(1-(1-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)azetidin-3-yl)piperidin-4-yl)-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000811
The first step is as follows: tert-butyl 3- [ [3- [4- [ 5-amino-4-cyano-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazol-1-yl ] -1-piperidinyl ] azetidin-1-yl ] methyl ] azetidine-1-carbonate (4a)
tert-butyl3-[[3-[4-[5-amino-4-cyano-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazol-1-yl]-1-piperidyl]azetidin-1-yl]methyl]azetidine-1-carboxylate
Figure BDA0003178773240000812
Compound 3a (0.5g, 1mmol) was dissolved in 5mL of N, N-dimethylacetamide, 3-formylazetidine-1-carboxylic acid tert-butyl ester (370mg, 2.0mmol), anhydrous magnesium sulfate 0.5g, 1 drop of acetic acid, stirred at 70 ℃ for 1h, sodium triacetoxyborohydride (422mg, 2.0mmol) was added, and the reaction was continued for 1 h. After the reaction was completed, the reaction mixture was cooled to room temperature, and extracted with 10mL of ethyl acetate and 10mL of water. The organic phase was dried over anhydrous sodium sulfate and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1-5:1) and concentrated to give the title compound as a yellow solid 4a (250mg, 37.4% yield)
MS m/z=673.3[M+1]+
The second step is that: 5-amino-1- [1- [1- (azetidin-3-ylmethyl) azetidin-3-yl ] -4-piperidinyl ] -3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (4b)
5-amino-1-[1-[1-(azetidin-3-ylmethyl)azetidin-3-yl]-4-piperidyl]-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240000821
After completion of the reaction, the compound (4a) (250mg, 0.37mmol) was dissolved in 2.5mL of 90% sulfuric acid, heated to 90 ℃, stirred for 1 hour, adjusted to PH 8 to 9 with an aqueous ammonia solution at 0 ℃, extracted with dichloromethane (20mL × 4), washed once with saturated brine (20mL × 1), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound (4b) as a yellow solid (200mg, 92%) which was used in the next reaction.
MS m/z=591.3[M+1]+
The third step: 5-amino-1- (1- (1- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindolin-5-yl) azacyclobutane-3-yl) methyl) azetidin-3-yl) piperidin-4-yl) -3- (4- ((5-fluoro-2-methoxybenzoyl) methyl) phenyl) -1H-pyrazole-4-carboxamide (Compound 4)
5-amino-1-(1-(1-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)azetidin-3-yl)piperidin-4-yl)-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000822
Compound (4b) (100mg,0.15mmol), 2- (2, 6-dioxo-3-piperidyl) -5-fluoroisoindoline-1, 3-dione (124mg, 0.42mmol) were dissolved in 5mL of DMSO, N-diisopropylethylamine (120mg, 0.93mmol) was added, and the reaction was carried out at 90 ℃ for 2 hours after the addition. After completion of the reaction, it was cooled to room temperature, 10mL of water was added, and the filtrate was filtered, and the filter cake was dissolved in 20mL of dichloromethane, washed with 5mL of saturated sodium chloride, dried over anhydrous sodium sulfate, and the crude product was concentrated under reduced pressure and subjected to column chromatography (DCM: MeOH ═ 20:1) to give the title compound 4 as a yellow solid (10mg, yield 7.0%).
MS m/z=847.3[M+1]+
1H NMR(400MHz,DMSO-d6)δ11.04(s,1H),8.81(t,1H),7.62(d,1H),7.51(dd,1H),7.48–7.38(m,4H),7.37–7.29(m,1H),7.18(dd,1H),6.76(d,1H),6.62(dd,1H),6.32(br.s,2H),5.04(dd,1H),4.54(d,2H),4.15–4.02(m,3H),3.89(s,3H),3.72–3.64(m,2H),3.54–3.38(m,2H),2.96–2.69(m 8H),2.62–2.53(m,2H),2.07–1.72(m,8H).
Example 5:
5-amino-1- [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] -4-piperidinyl ] methyl ] azetidin-3-yl ] -4-piperidinyl ] -3- [4- [ [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (Compound 5)
5-amino-1-[1-[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]azetidin-3-yl]-4-piperidyl]-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240000831
The first step is as follows: tert-butyl 4- [ [3- [4- [ 5-amino-4-cyano-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazol-1-yl ] -1-piperidinyl ] azetidin-1-yl ] methyl ] piperidine-1-carbonate (compound 5a)
tert-butyl4-[[3-[4-[5-amino-4-cyano-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazol-1-yl]-1-piperidyl]azetidin-1-yl]methyl]piperidine-1-carboxylate
Figure BDA0003178773240000832
Compound 4a (0.5g, 1.0mmol) was dissolved in 10mL of N, N-dimethylacetamide, and added1-Boc-4-piperazine Pyridinecarboxaldehyde(0.84g, 4.0mmol), anhydrous magnesium sulfate 0.5g, acetic acid 3 drops, stirring at 70 ℃ for 1h, adding sodium triacetoxyborohydride (0.84g, 4.0mmol), and continuing the reaction for 1 h. After the reaction was completed, the reaction mixture was cooled to room temperature, and extracted with 50mL of ethyl acetate and 20mL of water. Drying the organic phase with anhydrous sodium sulfate to obtain crude productPurification by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1-5:1), and concentration gave the title compound as a yellow solid (5a) (0.25g, yield 35.9%)
MS m/z=701.3[M+H]+
The second step is that: 5-amino-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] -1- [1- [1- (4-piperidinylmethyl) azepin-3-yl ] -4-piperidinyl ] pyrazole-4-carboxamide (5b)
5-amino-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]-1-[1-[1-(4-piperidylmethyl)azetidin-3-yl]-4-piperidyl]pyrazole-4-carboxamide
Figure BDA0003178773240000841
Tert-butyl 4- [ [3- [4- [ 5-amino-4-cyano-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazol-1-yl ] -1-piperidinyl ] azetidin-1-yl ] methyl ] piperidine-1-carboxylate (5a) (250mg, 0.35mmol) was dissolved in 2.5mL of 90% sulfuric acid, warmed to 90 ℃, stirred for 1 hour, after completion of the reaction, after adjusting the PH to 8 to 9 with an aqueous ammonia solution at 0 ℃, the mixture was extracted with dichloromethane (20mL × 4), washed once with saturated brine (20mL × 1), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain the title compound (5b) as a yellow solid (200mg, 90%) which was used in the next reaction.
MS m/z=619.3[M+1]+
The third step: 5-amino-1- [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] -4-piperidinyl ] methyl ] azetidin-3-yl ] -4-piperidinyl ] -3- [4- [ [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (Compound 5)
5-amino-1-[1-[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]methyl]azetidin-3-yl]-4-piperidyl]-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240000851
5-amino-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] -1- [1- [1- (4-piperidinylmethyl) azepin-3-yl ] -4-piperidinyl ] pyrazole-4-carboxamide (5b) (100mg,0.18mmol), 2- (2, 6-dioxo-3-piperidinyl) -5-fluoroisoindoline-1, 3-dione (124mg, 0.42mmol) were dissolved in 5mL of DMSO, N-diisopropylethylamine (120mg, 0.93mmol) was added, and after completion of the addition, the reaction was carried out at 90 ℃ for 2 hours. After completion of the reaction, it was cooled to room temperature, 10mL of water was added, and the filtrate was filtered, and the filter cake was dissolved in 20mL of dichloromethane, washed with 5mL of saturated sodium chloride, dried over anhydrous sodium sulfate, and the crude product was concentrated under reduced pressure and subjected to column chromatography (DCM: MeOH ═ 20:1) to give the title compound 5 as a yellow solid (5mg, yield 3.4%).
MS m/z=875.3[M+1]+
1H NMR(400MHz,DMSO-d6)δ11.05(s,1H),8.81(t,1H),7.64(d,1H),7.51(dd,1H),7.48–7.38(m,4H),7.37–7.27(m,2H),7.25–7.15(m,2H),6.32(br.s,2H),5.05(dd,1H),4.55(d,2H),4.17–3.96(m,3H),3.89(s,3H),3.60–3.37(m,2H),3.01–2.71(m,8H),2.63–2.53(m,2H),2.44–2.33(m,1H),2.05–1.53(m,11H),1.23–1.09(m,2H).
Example 6:
5-amino-1- [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] -3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (Compound 6)
5-amino-1-[1-[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]-4-piperidyl]-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240000852
Figure BDA0003178773240000861
The first step is as follows: 4- [4- [ 5-amino-4-cyano-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazol-1-yl ] -1-piperidinyl ] piperidine-1-carboxylic acid tert-butyl ester (compound 6a)
tert-butyl4-[4-[5-amino-4-cyano-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazol-1-yl]-1-piperidyl]piperidine-1-carboxylate
Figure BDA0003178773240000862
Intermediate 2a (1.45g, 3.0mmol) was dissolved in 10mL of N, N-dimethylacetamide and added1-Boc-4-piperazine Pyridinones as inhibitors of HIV replication(1.2g, 6.0mmol), anhydrous magnesium sulfate 0.5g, acetic acid 3 drops, stirring at 70 ℃ for 1h, adding sodium triacetoxyborohydride (1.26g, 6.0mmol), and continuing the reaction for 1 h. After the reaction was completed, the reaction mixture was cooled to room temperature, and extracted with 50mL of ethyl acetate and 20mL of water. The organic phase was dried over anhydrous sodium sulfate and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1-5:1) and concentrated to give the title compound as a yellow solid (6a) (1.45g, 76.7% yield)
MS m/z=632.3[M+H]+
The second step is that: 5-amino-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] -1- [1- (4-piperidinyl) -4-piperidinyl ] pyrazole-4-carboxamide (6b)
5-amino-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]-1-[1-(4-piperidyl)-4-piperidyl]pyrazole-4-carboxamide
Figure BDA0003178773240000871
After completion of the reaction, the compound 6a (145mg, 0.23mmol) was dissolved in 2.5mL of 90% sulfuric acid, heated to 90 ℃, stirred for 1 hour, adjusted to PH 8 to 9 with an aqueous ammonia solution at 0 ℃, extracted with dichloromethane (20mL × 4), washed once with saturated brine (20mL × 1), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound (6b) as a yellow solid (145mg, 93%), which was used directly in the next reaction.
MS m/z=550.3[M+1]+
The third step: 5-amino-1- [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] -3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (Compound 6)
5-amino-1-[1-[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-4-piperidyl]-4-piperidyl]-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240000872
Compound (6b) (100mg,0.18mmol), 2- (2, 6-dioxo-3-piperidyl) -5-fluoroisoindoline-1, 3-dione (124mg, 0.42mmol) were dissolved in 5mL of DMSO, N-diisopropylethylamine (120mg, 0.93mmol) was added, and the reaction was carried out at 90 ℃ for 2 hours after the addition. After completion of the reaction, it was cooled to room temperature, 10mL of water was added, and the filtrate was filtered, and the filter cake was dissolved in 20mL of dichloromethane, washed with 5mL of saturated sodium chloride, dried over anhydrous sodium sulfate, and the crude product was concentrated under reduced pressure and subjected to column chromatography (DCM: MeOH ═ 20:1) to obtain the title compound 6as a yellow solid (5mg, yield 3.4%).
MS m/z=806.3[M+1]+
Example 7:
5-amino-1- [1- [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] azetidin-3-yl ] -4-piperidinyl ] -3- [4- [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (Compound 7)
5-amino-1-[1-[1-[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]azetidin-3-yl]-4-piperidyl]-4-piperidyl]-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240000881
The first step is as follows: n- [ [4- [ 5-amino-4-cyano-1- [1- (4-piperidinyl) -4-piperidinyl ] pyrazol-3-yl ] phenyl ] methyl ] -5-fluoro-2-methoxy-benzamide (compound 7a)
N-[[4-[5-amino-4-cyano-1-[1-(4-piperidyl)-4-piperidyl]pyrazol-3-yl]phenyl]methyl]-5-fluoro-2-methoxy-benzamide
Figure BDA0003178773240000882
After the reaction was completed, the intermediate 6a (1.0g, 1.58mmol) was dissolved in 10mL of DCM, and 2mL of trifluoroacetic acid was added thereto, and the mixture was stirred at room temperature for 1 hour, and then concentrated under reduced pressure, dichloromethane 5m was added again, and l was adjusted to PH 8 to 9 with an aqueous ammonia solution at 0 ℃, extracted with dichloromethane (20mL × 4), washed once with saturated brine (20mL × 1), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain the title compound as a crude yellow solid 7a (1.0g), which was used in the next reaction.
MS m/z=532.3[M+H]+
The second step is that: tert-butyl 3- [4- [4- [ 5-amino-4-cyano-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazol-1-yl ] -1-piperidinyl ] azetidine-1-carboxylate (7b)
tert-butyl 3-[4-[4-[5-amino-4-cyano-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazol-1-yl]-1-piperidyl]-1-piperidyl]azetidine-1-carboxylate
Figure BDA0003178773240000891
N- [ [4- [ 5-amino-4-cyano-1- [1- (4-piperidinyl) -4-piperidinyl ] pyrazol-3-yl ] phenyl ] methyl ] -5-fluoro-2-methoxy-benzamide compound 7a (1.0g, 1.87mmol) was dissolved in 10mL of N, N-dimethylacetamide, tert-butyl 3-azetidine-1-carboxylate (0.85g, 5mmol), anhydrous magnesium sulfate 0.5g, 1 drop of acetic acid, stirred at 70 ℃ for 1h, sodium triacetoxyborohydride (1.05mg, 5mmol) was added, and the reaction was continued for 1 h. After the reaction was completed, the reaction mixture was cooled to room temperature, and extracted with 10mL of ethyl acetate and 10mL of water. The organic phase was dried over anhydrous sodium sulfate and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1-5:1) and concentrated to give the title compound as a yellow solid 7b (0.5g, 38.7% yield)
MS m/z=687.3[M+1]+
The third step: 5-amino-1- [1- [1- (azetidin-3-yl) -4-piperidinyl ] -3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (7c)
5-amino-1-[1-[1-(azetidin-3-yl)-4-piperidyl]-4-piperidyl]-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240000892
After completion of the reaction, the compound (7b) (0.5g, 0.72mmol) was dissolved in 2.5mL of 90% sulfuric acid, the temperature was raised to 90 ℃, and the mixture was stirred for 1 hour, and then the PH was adjusted to 8 to 9 with an aqueous ammonia solution at 0 ℃, and the mixture was extracted with dichloromethane (20mL × 4), washed once with saturated brine (20mL × 1), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain the title compound (7c) as a yellow solid (300mg, 70%) which was used in the next reaction.
MS m/z=605.3[M+1]+
The fourth step: 5-amino-1- [1- [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] azetidin-3-yl ] -4-piperidinyl ] -3- [4- [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (Compound 7)
5-amino-1-[1-[1-[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]azetidin-3-yl]-4-piperidyl]-4-piperidyl]-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240000901
Compound 7c (100mg,0.15mmol), 2- (2, 6-dioxo-3-piperidyl) -5-fluoroisoindoline-1, 3-dione (124mg, 0.42mmol) were dissolved in 5mL of DMSO, N-diisopropylethylamine (120mg, 0.93mmol) was added, and the reaction was carried out at 90 ℃ for 2 hours after the addition. After completion of the reaction, it was cooled to room temperature, 10mL of water was added, and the filtrate was filtered, and the filter cake was dissolved in 20mL of dichloromethane, washed with 5mL of saturated sodium chloride, dried over anhydrous sodium sulfate, and the crude product was concentrated under reduced pressure and subjected to column chromatography (DCM: MeOH ═ 20:1) to obtain the title compound 7 as a yellow solid (10mg, yield 7.0%).
MS m/z=861.3[M+1]+
Example 8:
5-amino-1- (1'- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindol-5-yl) azetidin-3-yl) methyl) - [1,4' -bipiperidin ] -4-yl) -3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide
5-amino-1-(1'-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)-[1,4'-bipiperidin]-4-yl)-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000902
Figure BDA0003178773240000911
The first step is as follows: 3- ((4- (5-amino-4-cyano-3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazol-1-yl) - [1,4 '-bipiperidine ] -1' -yl) methyl) azetidine-1-carboxylic acid tert-butyl ester (8a)
tert-butyl 3-((4-(5-amino-4-cyano-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazol-1-yl)-[1,4'-bipiperidin]-1'-yl)methyl)azetidine-1-carboxylate
Figure BDA0003178773240000912
Compound (7a) (170mg,0.20mmol) was dissolved in 15mL of DCE, sodium carbonate (67mg, 0.80mmol) was added, and the mixture was stirred at room temperature for 30min, tert-butyl 3-formylazetidine-1-carboxylate (74mg,0.40mmol) was added, and after stirring at room temperature for 25 min, sodium triacetoxyborohydride (127mg, 0.60mmol) was added, and the mixture was stirred at room temperature overnight. To the reaction system was slowly added 20mL of an aqueous sodium bicarbonate solution, extracted with DCM (50mL × 3), the organic phase was washed with 50mL of water, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1-4:1) to give tert-butyl 3- ((4- (5-amino-4-cyano-3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazol-1-yl) - [1,4 '-bipiperidin ] -1' -yl) methyl) azetidine-1-carboxylate (8a) (120mg, yield: 81.5%).
MS m/z=701.4[M+1]+
The fourth step: 5-amino-1- (1'- (azetidin-3-ylmethyl) - [1,4' -bispiperidine ] -4-yl) -3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (8b)
5-amino-1-(1'-(azetidin-3-ylmethyl)-[1,4'-bipiperidin]-4-yl)-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000921
Mixing 3- ((4- (5-amino-4-cyano-3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazol-1-yl) - [1,4' -bipiperidine]-1' -yl) methyl) azetidine-1-carboxylic acid tert-butyl ester (8a) (120mg,0.17mmol) was dissolved in 2mL 90% H2SO4And the mixture is stirred for 1.5 hours at 90 ℃ in an external bath. Cooling, slowly pouring the system into sodium carbonate aqueous solution under ice water external bath, extracting with dichloromethane, drying with anhydrous sodium sulfate, and spin-drying to obtain 5-amino-1- (1'- (azetidine-3-ylmethyl) - [1,4' -bipiperidine]-4-yl) -3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (8b) (85mg, yield: 74.5%).
MS m/z=619.4[M+1]+
The fifth step: 5-amino-1- (1'- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindol-5-yl) azetidin-3-yl) methyl) - [1,4' -bipiperidine ] -4-yl) -3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (Compound 8)
5-amino-1-(1'-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)-[1,4'-bipiperidin]-4-yl)-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000922
5-amino-1- (1'- (azetidin-3-ylmethyl) - [1,4' -bispiperidine ] -4-yl) -3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (8b) (80mg,0.13mmol) was dissolved in 4 mL of DMSO, 1.0mL of DIPEA and 2- (2, 6-dioxopiperidin-3-yl) -5-fluoroisoindoline-1, 3-dione (54mg, 0.19mmol) were added, the reaction was stirred at 80 ℃ in an external bath for 5 hours, the reaction was cooled, 50mL of water was added, filtration was performed, the solid was collected, washed with water, dissolved in dichloromethane, dried over anhydrous sodium sulfate, after concentration under reduced pressure, the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1) to give 5-amino-1- (1'- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindol-5-yl) azetidin-3-yl) methyl) - [1,4' -bipiperidin ] -4-yl) -3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (compound 8) (40mg, yield: 35.2%).
MS m/z=875.4[M+1]+
1H NMR(400MHz,CD3OD)δ7.66–7.58(m,2H),7.55–7.43(m,4H),7.29–7.20(m,1H),7.15(dd,1H),6.78(d,1H),6.62(dd,1H),5.04(dd,1H),4.67(s,2H),4.23–4.05(m,3H),3.95(s,3H),3.78–3.67(m,2H),3.22–2.95(m,5H),2.91–2.60(m,5H),2.54–2.37(m,3H),2.27–2.03(m,5H),2.02–1.84(m,4H),1.71–1.54(m,2H).
Example 9:
5-amino-1- (1'- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindol-5-yl) pyrrolidin-3-yl) methyl) - [1,4' -bipiperidine ] -4-yl) -3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (Compound 9)
5-amino-1-(1'-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)pyrrolidine-3-yl)methyl)-[1,4'-bipiperidin]-4-yl)-3-(4-((5-fluoro-2-methoxy benzamido)methyl)phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000931
The first step is as follows: 3- ((4- (5-amino-4-cyano-3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazol-1-yl) - [1,4 '-bipiperidine ] -1' -yl) methyl) pyrrolidine-1-carboxylic acid tert-butyl ester (9a)
tert-butyl 3-((4-(5-amino-4-cyano-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazol-1-yl)-[1,4'-bipiperidin]-1'-yl)methyl)pyrrolidine-1-carboxylate
Figure BDA0003178773240000941
Compound (7a) (170mg crude, 0.20mmol) was dissolved in 15mL DCE, sodium carbonate (67mg, 0.80mmol) was added, and the mixture was stirred at room temperature for 30min, tert-butyl 3-formylpyrrolidine-1-carboxylate (80mg,0.40mmol) was added, and after stirring at room temperature for 25 min, sodium triacetoxyborohydride (127mg, 0.60mmol) was added, and the mixture was stirred at room temperature overnight. To the reaction system was slowly added 20mL of an aqueous sodium hydrogencarbonate solution, followed by extraction with DCM (50mL × 3), washing of the organic phase with 50mL of water, drying over anhydrous sodium sulfate, concentration under reduced pressure, and purification of the crude product by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1-4:1) to give tert-butyl 3- ((4- (5-amino-4-cyano-3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazol-1-yl) - [1,4 '-bipiperidin ] -1' -yl) methyl) pyrrolidine-1-carboxylate (9a) (95mg, yield: 61.5%).
MS m/z=715.4[M+1]+
The second step is that: 5-amino-3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1- (1'- (pyrrolidin-3-ylmethyl) - [1,4' -bipiperidine ] -4-yl) -1H-pyrazole-4-carboxamide (9b)
5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1'-(pyrrolidin-3-ylmethyl)-[1,4'-bipiperidin]-4-yl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000942
Mixing 3- ((4- (5-amino-4-cyano-3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazol-1-yl) - [1,4' -bipiperidine]-1' -Yl) methyl) pyrrolidine-1-carboxylic acid tert-butyl ester (9a) (90mg,0.13mmol) was dissolved in 2mL of 90% H2SO4And the mixture is stirred for 1.5 hours at 90 ℃ in an external bath. Cooling, slowly pouring the system into sodium carbonate aqueous solution under ice water external bath, extracting with dichloromethane, drying with anhydrous sodium sulfate, and spin-drying to obtain 5-amino-3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1- (1'- (pyrrolidine-3-ylmethyl) - [1,4' -bipiperidine]-4-yl) -1H-pyrazole-4-carboxamide (9b) (60mg, yield: 75.2%).
MS m/z=633.4[M+1]+
The third step: 5-amino-1- (1'- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindol-5-yl) pyrrolidin-3-yl) methyl) - [1,4' -bipiperidine ] -4-yl) -3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (Compound 9)
5-amino-1-(1'-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)pyrrolidine-3-yl)methyl)-[1,4'-bipiperidin]-4-yl)-3-(4-((5-fluoro-2-methoxy benzamido)methyl)phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000951
5-amino-3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1- (1'- (pyrrolidin-3-ylmethyl) - [1,4' -bipiperidine ] -4-yl) -1H-pyrazole-4-carboxamide (9b) (60mg,0.10mmol) was dissolved in 4 mL DMSO, 0.5mL DIPEA and 2- (2, 6-dioxopiperidin-3-yl) -5-fluoroisoindoline-1, 3-dione (40mg, 0.15mmol) were added, the reaction was stirred at 80 ℃ for 5 hours in an external bath, the reaction was cooled, 15mL water was added, filtration was carried out, the solid was collected, washed with water, the solid was dissolved with dichloromethane, dried over anhydrous sodium sulfate, after concentration under reduced pressure, the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1) to give 5-amino-1- (1'- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindol-5-yl) pyrrolidin-3-yl) methyl) - [1,4' -bipiperidin ] -4-yl) -3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (compound 9) (25mg, yield: 29.0%).
MS m/z=889.3[M+1]+
1H NMR(400MHz,CD3OD)δ7.66–7.58(m,2H),7.54–7.45(m,4H),7.29–7.21(m,1H),7.19–7.13(m,1H),6.99–6.94(m,1H),6.80(dd,1H),5.04(dd,1H),4.67(s,2H),4.58–4.47(m,1H),4.18–4.06(m,1H),3.95(s,3H),3.69–3.38(m,4H),3.23–3.01(m,5H),2.92–2.60(m,4H),2.54–2.42(m,4H),2.28–2.04(m,5H),2.04–1.77(m,5H),1.72–1.58(m,2H).
Example 10:
5-amino-1- (1'- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindolin-5-yl) piperidin-4-yl) methyl) - [1,4' -bipiperidine ] -4-yl) -3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (Compound 10)
5-amino-1-(1'-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)-[1,4'-bipiperidin]-4-yl)-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000961
The first step is as follows: 4- ((4- (5-amino-4-cyano-3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazol-1-yl) - [1,4 '-bipiperidine ] -1' -yl) methyl) piperidine-1-carboxylic acid tert-butyl ester (10a)
tert-butyl 4-((4-(5-amino-4-cyano-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazol-1-yl)-[1,4'-bipiperidin]-1'-yl)methyl)piperidine-1-carboxylate
Figure BDA0003178773240000962
Compound (7a) (340mg of crude product, 0.40mmol) was dissolved in 25mL of DCE, and sodium hydrogencarbonate (135mg, 1.60mmol) was added thereto, followed by stirring at room temperature for 30min, tert-butyl 4-formylpiperidine-1-carboxylate (170mg,0.80mmol) was added thereto, followed by stirring at room temperature for 25 min, followed by addition of sodium triacetoxyborohydride (254mg, 1.20mmol) and stirring at room temperature overnight. To the reaction system was slowly added 20mL of an aqueous sodium hydrogencarbonate solution, followed by extraction with DCM (50mL × 3), washing of the organic phase with 50mL of water, drying over anhydrous sodium sulfate, concentration under reduced pressure, and purification of the crude product by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1-4:1) to give tert-butyl 4- ((4- (5-amino-4-cyano-3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazol-1-yl) - [1,4 '-bipiperidin ] -1' -yl) methyl) piperidine-1-carboxylate (10a) (190mg, yield: 62%).
MS m/z=729.4[M+1]+
The second step is that: 5-amino-3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1- (1'- (piperidin-4-ylmethyl) - [1,4' -bipiperidine ] -4-yl) -1H-pyrazole-4-carboxamide (10b)
5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1'-(piperidin-4-ylmethyl)-[1,4'-bipiperidin]-4-yl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000971
Mixing 4- ((4- (5-amino-4-cyano-3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazol-1-yl) - [1,4' -bipiperidine]-1' -Yl) methyl) piperidine-1-carboxylic acid tert-butyl ester (10a) (190mg,0.26mmol) was dissolved in 3mL 90% H2SO4And the mixture is stirred for 1.5 hours at 90 ℃ in an external bath. Cooling, slowly pouring the system into sodium carbonate aqueous solution under ice water bath, extracting with dichloromethane, drying with anhydrous sodium sulfate, and spin-drying to obtain 5-amino-3- (4- (((5-fluoro-2-methoxybenzyl)Amido) methyl) phenyl) -1- (1'- (piperidin-4-ylmethyl) - [1,4' -bipiperidine]-4-yl) -1H-pyrazole-4-carboxamide (10b) (140mg, yield: 83.2%).
MS m/z=647.4[M+1]+
The third step: 5-amino-1- (1'- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindolin-5-yl) piperidin-4-yl) methyl) - [1,4' -bipiperidine ] -4-yl) -3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (Compound 10)
5-amino-1-(1'-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)-[1,4'-bipiperidin]-4-yl)-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000972
5-amino-3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1- (1'- (piperidin-4-ylmethyl) - [1,4' -bipiperidine ] -4-yl) -1H-pyrazole-4-carboxamide (10b) (70mg,0.11mmol) was dissolved in 4 mL of DMSO, 0.5mL of DIPEA and 2- (2, 6-dioxopiperidin-3-yl) -5-fluoroisoindoline-1, 3-dione (50mg, 0.15mmol) were added, the reaction was stirred at 80 ℃ in an external bath for 5 hours, the reaction was cooled, 15mL of water was added, filtration was performed, the solid was collected, washed with water, the solid was dissolved with dichloromethane, dried over anhydrous sodium sulfate, after concentration under reduced pressure, the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1) to give 5-amino-1- (1'- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindolin-5-yl) piperidin-4-yl) methyl) - [1,4' -bipiperidine ] -4-yl) -3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (compound 10) (20mg, yield: 20.2%).
MS m/z=903.4[M+1]+
Example 11:
5-amino-1- (1'- ((1- (2- (2, 6-dioxopiperidin-3-yl) -6-fluoro-1, 3-dioxoisoindol-5-yl) piperidin-4-yl) methyl) - [1,4' -bipiperidin ] -4-yl) -3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (Compound 11)
5-amino-1-(1'-((1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)-[1,4'-bipiperidin]-4-yl)-3-(4-((5-fluoro-2-methoxybenz-amido)methyl)phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240000981
5-amino-3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1- (1'- (piperidin-4-ylmethyl) - [1,4' -bipiperidine ] -4-yl) -1H-pyrazole-4-carboxamide (10b) (70mg,0.11mmol) was dissolved in 4 mL of DMSO, 0.5mL of DIPEA and 2- (2, 6-dioxopiperidin-3-yl) -5-fluoroisoindoline-1, 3-dione (50mg, 0.15mmol) were added, the reaction was stirred at 80 ℃ in an external bath for 5 hours, the reaction was cooled, 15mL of water was added, filtration was performed, the solid was collected, washed with water, the solid was dissolved with dichloromethane, dried over anhydrous sodium sulfate, after concentration under reduced pressure, the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1) to give 5-amino-1- (1'- ((1- (2- (2, 6-dioxopiperidin-3-yl) -6-fluoro-1, 3-dioxoisoindol-5-yl) piperidin-4-yl) methyl) - [1,4' -bipiperidine ] -4-yl) -3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (compound 11) (25mg, yield: 24.6%).
MS m/z=921.3[M+1]+
Example 12:
5-amino-1- [1- [2- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] -2-azaspiro [3.5] non-7-yl ] -4-piperidinyl ] -3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (Compound 12)
5-amino-1-[1-[2-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-2-azaspiro[3.5]nonan-7-yl]-4-piperidyl]-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240000991
The first step is as follows: tert-butyl 7- [4- [ 5-amino-4-cyano-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazol-1-yl ] -1-piperidinyl ] -2-azaspiro [3.5] nonane-2-carbonate (12a)
tert-butyl 7-[4-[5-amino-4-cyano-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazol-1-yl]-1-piperidyl]-2-azaspiro[3.5]nonane-2-carboxylate
Figure BDA0003178773240000992
Intermediate 2a (145mg, 0.3mmol) was dissolved in 5mL of N, N-dimethylacetamide, and tert-butyl 7-oxo-2-azaspiro [3.5] nonane-2-carboxylate (239mg, 1.0mmol), anhydrous magnesium sulfate 0.5g, 1 drop of acetic acid was added, stirred at 70 ℃ for 1h, and sodium triacetoxyborohydride (211mg, 1.0mmol) was added, and the reaction was continued for 1 h. After the reaction was completed, the reaction mixture was cooled to room temperature, and extracted with 10mL of ethyl acetate and 10mL of water. The organic phase was dried over anhydrous sodium sulfate, and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1-5:1), and concentrated to give the title compound as a yellow solid (12a) (100mg, yield 46.7%)
MS m/z=672.3[M+H]+
The second step is that: 5-amino-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] -1- [1- (4-piperidinyl) -4-piperidinyl ] pyrazole-4-carboxamide (12b)
5-amino-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]-1-[1-(4-piperidyl)-4-piperidyl]pyrazole-4-carboxamide
Figure BDA0003178773240001001
Tert-butyl 7- [4- [ 5-amino-4-cyano-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazol-1-yl ] -1-piperidinyl ] -2-azaspiro [3.5] nonane-2-carbonate (12a) (100mg,0.15mmol) was dissolved in 2.5mL of 90% sulfuric acid, the temperature was raised to 90 ℃, stirring was performed for 1 hour, after completion of the reaction, the PH was adjusted to 8 to 9 with an aqueous ammonia solution at 0 ℃, dichloromethane was used for extraction (20mL × 4), the mixture was washed once with saturated brine (20mL × 1), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound (12b) as a yellow solid (100mg, 89%) which was used directly in the next reaction.
MS m/z=590.3[M+1]+
The third step: 5-amino-1- [1- [2- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] -2-azaspiro [3.5] non-7-yl ] -4-piperidinyl ] -3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (Compound 12)
5-amino-1-[1-[2-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-2-azaspiro[3.5]nonan-7-yl]-4-piperidyl]-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240001011
5-amino-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] -1- [1- (4-piperidinyl) -4-piperidinyl ] pyrazole-4-carboxamide (12b) (100mg,0.15mmol), 2- (2, 6-dioxo-3-piperidinyl) -5-fluoroisoindoline-1, 3-dione (124mg, 0.42mmol) was dissolved in 5mL of DMSO, N-diisopropylethylamine (120mg, 0.93mmol) was added, and after the addition, the reaction was carried out at 90 ℃ for 2 hours. After completion of the reaction, it was cooled to room temperature, 10mL of water was added, and the filtrate was filtered, and the filter cake was dissolved in 20mL of dichloromethane, washed with 5mL of saturated sodium chloride, dried over anhydrous sodium sulfate, and the crude product was concentrated under reduced pressure and subjected to column chromatography (DCM: MeOH ═ 20:1) to give the title compound 12 as a yellow solid (10mg, yield 7.0%).
MS m/z=846.3[M+1]+
1H NMR(400MHz,CD3OD)δ7.67–7.46(m,6H),7.29–7.10(m,2H),6.84–6.77(m,1H),6.69–6.60(m,1H),5.05(dd,1H),4.71–4.62(m,2H),4.51–4.37(m,1H),3.96(s,3H),3.88–3.66(m,6H),3.44–3.16(m,3H),2.93–2.63(m,3H),2.47–2.04(m,9H),1.82–1.57(m,4H).
Example 13:
1- [1- [ (3aR,6aS) -2- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindol-5-yl ] -3,3a,4,5,6,6 a-hexahydro-1H-cyclopenta [ c ] pyrrol-5-yl ] -4-piperidyl ] -5-amino-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (Compound 13)
1-[1-[(3aR,6aS)-2-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-3,3a,4,5,6,6a-hexahydro-1H-cyclopenta[c]pyrrol-5-yl]-4-piperidyl]-5-amino-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240001012
Figure BDA0003178773240001021
The first step is as follows: tert-butyl (3aR,6aS) -5- [4- [ 5-amino-4-cyano-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazol-1-yl ] -1-piperidinyl ] -3,3a,4,5,6,6 a-hexahydro-1H-cyclopenty [ c ] pyrrole-2-carbonate (Compound 13a)
tert-butyl(3aR,6aS)-5-[4-[5-amino-4-cyano-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazol-1-yl]-1-piperidyl]-3,3a,4,5,6,6a-hexahydro-1H-cyclopenta[c]pyrrole-2-carboxylate
Figure BDA0003178773240001022
Intermediate 2a (145mg, 0.3mmol) was dissolved in 5mL of N, N-dimethylacetamide, and (3aR,6aS) -5-octahydrocyclopenta [ C ] pyrrole-2 (1H) -carboxylic acid tert-butyl ester (225mg, 1.0mmol), anhydrous magnesium sulfate 0.5g, 1 drop of acetic acid, stirred at 70 ℃ for 1H, sodium triacetoxyborohydride (211mg, 1.0mmol) was added, and the reaction was continued for 1H. After the reaction was completed, the reaction mixture was cooled to room temperature, and extracted with 10mL of ethyl acetate and 10mL of water. The organic phase was dried over anhydrous sodium sulfate and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1-5:1) and concentrated to give the title compound as a yellow solid 13a (100mg, 50.8% yield)
MS m/z=658.3[M+H]+
The second step is that: 1- [1- [ (3aR,6aS) -1,2,3,3a, 4,5,6,6 a-octahydrocyclopenta [ c ] pyrrol-5-yl ] -4-piperidinyl ] -5-amino-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (13b)
1-[1-[(3aR,6aS)-1,2,3,3a,4,5,6,6a-octahydrocyclopenta[c]pyrrol-5-yl]-4-piperidyl]-5-amino-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240001031
Tert-butyl (3aR,6aS) -5- [4- [ 5-amino-4-cyano-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazol-1-yl ] -1-piperidinyl ] -3,3a,4,5,6,6 a-hexahydro-1H-cyclopenta [ c ] pyrrole-2-carbonate (compound 13a) (100mg,0.15mmol) was dissolved in 2.5mL of 90% sulfuric acid, the temperature was raised to 90 ℃, stirring was performed for 1 hour, after completion of the reaction, the pH was adjusted to 8 to 9 with an aqueous ammonia solution at 0 ℃, dichloromethane was extracted (20 mL. times.4), washed once with saturated brine (20 mL. times.1), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound (13b) aS a yellow crude solid (87mg), directly used for the next reaction.
MS m/z=576.3[M+1]+
The third step: 1- [1- [ (3aR,6aS) -2- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindol-5-yl ] -3,3a,4,5,6,6 a-hexahydro-1H-cyclopenta [ c ] pyrrol-5-yl ] -4-piperidyl ] -5-amino-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (Compound 13)
1-[1-[(3aR,6aS)-2-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]-3,3a,4,5,6,6a-hexahydro-1H-cyclopenta[c]pyrrol-5-yl]-4-piperidyl]-5-amino-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240001032
1- [1- [ (3aR,6aS) -1,2,3,3a, 4,5,6,6 a-octahydrocyclopenta [ c ] pyrrol-5-yl ] -4-piperidinyl ] -5-amino-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (13b) (100mg,0.15mmol), 2- (2, 6-dioxo-3-piperidinyl) -5-fluoroisoindoline-1, 3-dione (124mg, 0.42mmol) were dissolved in 5mL of DMSO, N-diisopropylethylamine (120mg, 0.93mmol) was added, and after the addition, the reaction was carried out at 90 ℃ for 2 hours. After completion of the reaction, it was cooled to room temperature, 10mL of water was added, and the filtrate was filtered, and the filter cake was dissolved in 20mL of dichloromethane, washed with 5mL of saturated sodium chloride, dried over anhydrous sodium sulfate, and the crude product was concentrated under reduced pressure and subjected to column chromatography (DCM: MeOH ═ 20:1) to give the title compound 13 as a yellow solid (10mg, yield 7.0%).
MS m/z=832.3[M+1]+
1H NMR(400MHz,CD3OD)δ7.70–7.41(m,6H),7.29–7.20(m,1H),7.19–7.11(m,1H),7.09–7.01(m,1H),6.95–6.86(m,1H),5.05(dd,1H),4.66(s,2H),4.49–4.37(m,1H),3.95(m,3H),3.85–3.64(m,3H),3.61–3.45(m,4H),3.24–3.10(m,2H),3.04–2.91(m,2H),2.91–2.54(m,5H),2.46–2.17(m,4H),2.15–2.05(m,1H),1.79–1.62(m,2H).
Example 14:
5-amino-1- [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] azetidin-3-yl ] methyl ] -4-piperidinyl ] -3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (Compound 14)
5-amino-1-[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]azetidin-3-yl]methyl]-4-piperidyl]-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240001041
The first step is as follows: tert-butyl 3- [ [4- [ 5-amino-4-cyano-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazol-1-yl ] -1-piperidinyl ] methyl ] azetidine-1-carbonate (compound 14a)
tert-butyl3-[[4-[5-amino-4-cyano-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazol-1-yl]-1-piperidyl]methyl]azetidine-1-carboxylate
Figure BDA0003178773240001051
Intermediate 2a (145mg, 0.3mmol) was dissolved in 5mL of N, N-dimethylacetamide, 3-formylazetidine-1-carboxylic acid tert-butyl ester (185mg, 1.0mmol), anhydrous magnesium sulfate 0.5g, 1 drop of acetic acid, stirred at 70 ℃ for 1h, sodium triacetoxyborohydride (211mg, 1.0mmol) was added, and the reaction was continued for 1 h. After the reaction was completed, the reaction mixture was cooled to room temperature, and extracted with 10mL of ethyl acetate and 10mL of water. The organic phase was dried over anhydrous sodium sulfate and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 10:1-5:1) and concentrated to give the title compound as a yellow solid 14a (100mg, 54.1% yield)
MS m/z=618.3[M+H]+
The second step is that: 5-amino-1- [1- (azetidin-3-ylmethyl) -4-piperidinyl ] -3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (14b)
5-amino-1-[1-(azetidin-3-ylmethyl)-4-piperidyl]-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240001052
Tert-butyl 3- [ [4- [ 5-amino-4-cyano-3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazol-1-yl ] -1-piperidinyl ] methyl ] azetidine-1-carbonate (compound 14a) (100mg, 0.19mmol) was dissolved in 2.5mL 90% sulfuric acid, the temperature was raised to 90 ℃, stirred for 1 hour, after completion of the reaction, the PH was adjusted to 8 to 9 with an aqueous ammonia solution at 0 ℃, extracted with dichloromethane (20mL × 4), washed once with a saturated saline solution (20mL × 1), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the title compound (14b) as a crude yellow solid (100mg) which was used directly in the next reaction.
MS m/z=536.2[M+1]+
The third step: 5-amino-1- [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] azetidin-3-yl ] methyl ] -4-piperidinyl ] -3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (Compound 14)
5-amino-1-[1-[[1-[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-5-yl]azetidin-3-yl]methyl]-4-piperidyl]-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]pyrazole-4-carboxamide
Figure BDA0003178773240001061
5-amino-1- [1- (azetidin-3-ylmethyl) -4-piperidinyl ] -3- [4- [ [ (5-fluoro-2-methoxy-benzoyl) amino ] methyl ] phenyl ] pyrazole-4-carboxamide (14b) (100mg,0.15mmol), 2- (2, 6-dioxo-3-piperidinyl) -5-fluoroisoindoline-1, 3-dione (124mg, 0.42mmol) was dissolved in 5mL of DMSO, N-diisopropylethylamine (120mg, 0.93mmol) was added, and after completion of the addition, reaction was carried out at 90 ℃ for 2 hours. After completion of the reaction, it was cooled to room temperature, 10mL of water was added, and the filtrate was filtered, and the filter cake was dissolved in 20mL of dichloromethane, washed with 5mL of saturated sodium chloride, dried over anhydrous sodium sulfate, and the crude product was concentrated under reduced pressure and subjected to column chromatography (DCM: MeOH ═ 20:1) to give the title compound 14 as a yellow solid (10mg, yield 7.0%).
MS m/z=792.3[M+1]+
1H NMR(400MHz,CD3OD)δ7.69–7.45(m,6H),7.29–7.20(m,1H),7.19–7.11(m,1H),6.86–6.77(m,1H),6.71–6.62(m,1H),5.05(dd,1H),4.66(s,2H),4.53–4.40(m,1H),4.34–4.21(m,2H),3.95(s,3H),3.91–3.83(m,2H),3.79–3.69(m,1H),3.66–3.57(m,2H),3.39–3.33(m,1H),3.28–3.15(m,1H),2.91–2.63(m,4H),2.50–2.33(m,2H),2.31–2.19(m,2H),2.16–2.05(m,1H).
Example 15:
5-amino-1- (1- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindolin-5-yl) piperidin-4-yl) methyl) piperidin-4-yl) -3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (Compound 15)
5-amino-1-(1-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperidin-4-yl)-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240001071
The first step is as follows: tert-butyl 4- ((4- (5-amino-4-cyano-3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazol-1-yl) piperidin-1-yl) methyl) piperidine-1-carboxylate (15a)
tert-butyl 4-((4-(5-amino-4-cyano-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazol-1-yl)piperidin-1-yl)methyl)piperidine-1-carboxylate
Figure BDA0003178773240001072
Compound (2a) (0.67g,1.5mmol) was dissolved in 20mL of DCE, and tert-butyl 4-formylpiperidine-1-carboxylate (0.64g,3.0mmol) was added. After stirring at room temperature for 10min, sodium triacetoxyborohydride (0.7g, 3.3mmol) was added thereto, and the reaction was stirred at room temperature for 12 h. To the reaction solution, 50mL of a saturated sodium bicarbonate solution was slowly dropped, 30mL of dichloromethane was extracted, dried over anhydrous sodium sulfate, and after concentration under reduced pressure, the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give tert-butyl 4- ((4- (5-amino-4-cyano-3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazol-1-yl) piperidin-1-yl) methyl) piperidine-1-carboxylate (15a) (0.5g, yield: 52%).
LC-MS m/z=646.3[M+1]+
The second step is that: 5-amino-3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1- (1- (piperidin-4-ylmethyl) piperidin-4-yl) -1H-pyrazole-4-carboxamide (15b)
5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1-(piperidin-4-ylmethyl)piperidin-4-yl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240001081
Tert-butyl 4- ((4- (5-amino-4-cyano-3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazol-1-yl) piperidin-1-yl) methyl) piperidine-1-carboxylate (15a) (0.5g,0.78mmol) was dissolved in 10mL sulfuric acid (W% ═ 90%), and stirred at 90 ℃ for 1H. After cooling to room temperature, 30mL of purified water was added to dilute the solution, the pH of the system was adjusted to about 10.0 with sodium carbonate solid, the solution was extracted with 60mL of dichloromethane, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude 5-amino-3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1- (1- (piperidin-4-ylmethyl) piperidin-4-yl) -1H-pyrazole-4-carboxamide (15b) (0.34g, yield: 77%) which was directly subjected to the next reaction.
MS m/z=564.3[M+1]+
The third step: 5-amino-1- (1- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindolin-5-yl) piperidin-4-yl) methyl) piperidin-4-yl) -3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (Compound 15)
5-amino-1-(1-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperidin-4-yl)-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240001082
5-amino-3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1- (1- (piperidin-4-ylmethyl) piperidin-4-yl) -1H-pyrazole-4-carboxamide (15b) (0.2g,0.35mmol) was dissolved in 10mL of DMSO, 1.0mL of DIPEA and 2- (2, 6-dioxopiperidin-3-yl) -5-fluoroisoindoline-1, 3-dione (0.2g, 0.7mmol) were added, the reaction mixture was stirred at 90 ℃ for 5 hours while being externally bathed, cooled, diluted with 50mL of ethyl acetate, washed with 50mL of water, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and then the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v): 20:1), to give 5-amino-1- (1- ((1- (2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindolin-5-yl) piperidin-4-yl) methyl) piperidin-4-yl) -3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (15) (0.15g, yield: 52%)
MS m/z=820.3[M+1]+
1H NMR(400MHz,DMSO-d6)δ11.06(br.s,1H),8.82(t,1H),7.64(d,1H),7.55–7.38(m,5H),7.37–7.27(m,2H),7.26–7.15(m,2H),6.33(br.s,2H),5.06(dd,1H),4.55(d,2H),4.15–4.06(m,1H),4.06–3.97(m,2H),3.90(s,3H),3.01–2.89(m,5H),2.63–2.53(m,2H),2.21–2.12(m,2H),2.09–1.92(m,5H),1.85–1.73(m,5H),1.21–1.08(m,2H).
Example 16:
5-amino-1- (1- ((1- (2- (2, 6-dioxopiperidin-3-yl) -6-fluoro-1, 3-dioxoisoindol-5-yl) piperidin-4-yl) methyl) piperidin-4-yl) -3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide
5-amino-1-(1-((1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperidin-4-yl)-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1H-pyrazole-4-carboxamide
Figure BDA0003178773240001091
5-amino-3- (4- (((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1- (1- (piperidin-4-ylmethyl) piperidin-4-yl) -1H-pyrazole-4-carboxamide (15b) (0.2g,0.35mmol) was dissolved in 10mL of DMSO, 1.0mL of DIPEA and 2- (2, 6-dioxopiperidin-3-yl) -5, 6-difluoroisoindoline-1, 3-dione (0.2g, 0.7mmol) were added, the reaction mixture was stirred at 90 ℃ for 5 hours while being externally bathed, and the reaction mixture was cooled, diluted with 50mL of ethyl acetate, washed with 50mL of water, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and then the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v): 20:1), to give 5-amino-1- (1- ((1- (2- (2, 6-dioxopiperidin-3-yl) -6-fluoro-1, 3-dioxoisoindol-5-yl) piperidin-4-yl) methyl) piperidin-4-yl) -3- (4- ((5-fluoro-2-methoxybenzamido) methyl) phenyl) -1H-pyrazole-4-carboxamide (Compound 16) (0.14g, yield: 47%)
MS m/z=838.3[M+1]+
1H NMR(400MHz,DMSO-d6δ11.09(s,1H),8.82(t,1H),7.68(d,1H),7.55–7.29(m,7H),7.18(dd,1H),6.33(s,2H),5.09(dd,1H),4.55(d,2H),4.18–4.06(m,1H),3.89(s,3H),3.65–3.54(m,2H),3.00–2.81(m,5H),2.65–2.53(m,2H),2.25–2.17(m,2H),2.09–1.93(m,5H),1.88–1.75(m,5H),1.34–1.20(m,2H).
Example 17:
5- (4- ((4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) methyl) piperidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Compound 17)
5-(4-((4-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)methyl)piperidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione
Figure BDA0003178773240001101
The first step is as follows: 4-Methylbenzenesulfonic acid 4-oxocyclohexyl ester (17b)
4-oxocyclohexyl 4-methylbenzenesulfonate
Figure BDA0003178773240001102
4-Hydroxycyclohex-1-one (17a) (10g,87.61mmol) was dissolved in 120mL of dichloromethane, pyridine (10.4g, 131.5mmol) was added, p-toluenesulfonyl chloride (18.4g,96.52mmol) was added slowly in portions, and after the addition was complete, stirring was carried out at room temperature for 16 h. To the reaction mixture was added 300mL of methylene chloride, and the organic phase was washed successively with 0.5mol/L hydrochloric acid solution (100mL), 100mL of saturated sodium bicarbonate solution and 100mL of water, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give 4-oxocyclohexyl 4-methylbenzenesulfonate (17b) (15g, yield: 64%).
The second step is that: 4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohex-1-one (17c)
4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexan-1-one
Figure BDA0003178773240001111
3- (4-Phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (9.1g, 30.00mmol) was dissolved in 120mL of DMF, potassium carbonate (12.43g, 90.00mmol) was added, the temperature of the reaction was raised to 70 ℃, 4-oxocyclohexyl 4-methylbenzenesulfonate (17b) (15.0g,55.9mmol) was added slowly in portions, and after the addition, the reaction was stirred at 70 ℃ for 4H. The reaction mixture was cooled to room temperature, 200mL of water was added, and the mixture was extracted with 200mL of ethyl acetate, the organic phase was washed with 100mL of water, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude product, which was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 25:1-15:1), to give 4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohex-1-one (17c) (3.0g, yield: 25%).
LCMS m/z=400.2[M+1]+
The third step: 4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazine-1-carboxylic acid tert-butyl ester (17d)
tert-butyl 4-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazine-1-carboxylate
Figure BDA0003178773240001112
4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohex-1-one (17c) (200mg,0.50mmol) was dissolved in 15ml of DCE, tert-butyl piperazine-1-carboxylate (186mg,1.00mmol) was added, stirring was carried out at room temperature for 30min, sodium triacetoxyborohydride (212mg,1.00mmol) was added, and stirring was carried out at room temperature for 16H. To the reaction solution, 30mL of an aqueous sodium bicarbonate solution was slowly added, extraction was performed with 100mL of dichloromethane, the organic phase was washed with 100mL of water, dried over anhydrous sodium sulfate, and concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give tert-butyl 4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazine-1-carboxylate (17d) (210mg, yield: 74%).
LCMS m/z=570.3[M+1]+
The fourth step: trifluoroacetic acid salt of 3- (4-phenoxyphenyl) -1- (4- (piperazin-1-yl) cyclohexyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (17e)
3-(4-phenoxyphenyl)-1-(4-(piperazin-1-yl)cyclohexyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine trifluoroacetate
Figure BDA0003178773240001121
Tert-butyl 4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazine-1-carboxylate (17d) (200mg,0.35mmol) was dissolved in 15mL of dichloromethane, 8mL of trifluoroacetic acid was added, and the mixture was stirred at room temperature for 2H. The reaction system was concentrated under reduced pressure to give a crude product of 3- (4-phenoxyphenyl) -1- (4- (piperazin-1-yl) cyclohexyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (17e) as the trifluoroacetate salt (0.25 g).
LCMS m/z=470.3[M+1]+
The fifth step: 4- ((4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) methyl) piperidine-1-carboxylic acid tert-butyl ester (17f)
tert-butyl 4-((4-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)methyl)piperidine-1-carboxylate
Figure BDA0003178773240001122
The trifluoroacetate salt (0.12g) of the above crude 3- (4-phenoxyphenyl) -1- (4- (piperazin-1-yl) cyclohexyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (17e) was dissolved in 15mL of DCE, solid sodium bicarbonate (60mg, 0.71mmol) was added, and the mixture was stirred at room temperature for 20min, tert-butyl 4-formylpiperidine-1-carboxylate (73mg,0.34mmol) was added, and the mixture was stirred at room temperature for 30min, sodium triacetoxyborohydride (72mg, 0.34mmol) was added, and the mixture was stirred at room temperature for 16H. To the reaction solution, 30mL of a saturated sodium bicarbonate solution was slowly added, extraction was performed with dichloromethane (30mL × 3), the organic phase was washed with 50mL of water, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give tert-butyl 4- ((4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) methyl) piperidine-1-carboxylate (17f) (100mg, two-step yield from compound 17 d: 89%).
LCMS m/z=667.4[M+1]+
And a sixth step: trifluoroacetic acid salt of 3- (4-phenoxyphenyl) -1- (4- (4- (piperidin-4-ylmethyl) piperazin-1-yl) cyclohexyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (17g)
3-(4-phenoxyphenyl)-1-(4-(4-(piperidin-4-ylmethyl)piperazin-1-yl)cyclohexyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine trifluoroacetate
Figure BDA0003178773240001131
Tert-butyl 4- ((4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) methyl) piperidine-1-carboxylate (17f) (100mg,0.15mmol) was dissolved in 10mL DCM and 5mL trifluoroacetic acid was added and stirred at room temperature for 2H. The reaction system was concentrated under reduced pressure to give a crude product of 3- (4-phenoxyphenyl) -1- (4- (4- (piperidin-4-ylmethyl) piperazin-1-yl) cyclohexyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (17g) as the trifluoroacetate salt (0.15 g).
LCMS m/z=567.3[M+1]+
The seventh step: 5- (4- ((4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) methyl) piperidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Compound 17)
5-(4-((4-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)methyl)piperidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione
Figure BDA0003178773240001132
The above crude 3- (4-phenoxyphenyl) -1- (4- (4- (piperidin-4-ylmethyl) piperazin-1-yl) cyclohexyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (17g) as trifluoroacetate (0.15g) was dissolved in 5mL of DMSO, solid sodium bicarbonate (61mg, 0.73mmol) was added and stirred at room temperature for 10min, 0.5mL of DIPEA and 2- (2, 6-dioxopiperidin-3-yl) -5-fluoroisoindoline-1, 3-dione (see WO2017197056 for synthesis) (60mg, 0.22mmol) were added and the reaction was stirred at 80 ℃ for 5H. The reaction mixture was cooled to room temperature, 50mL of water was added, and the filtrate was filtered, and the filter cake was collected, washed with 10mL of water, the solid was dissolved in 100mL of dichloromethane, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude product, which was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 15:1) to give 5- (4- ((4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) methyl) piperidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (compound 17) (70mg, two-step yield from compound 17 f: 57%).
1H NMR(400MHz,CDCl3)δ8.63(br.s,1H),8.38(s,1H),7.70–7.61(m,3H),7.43–7.35(m,2H),7.29–7.26(m,1H),7.21–7.12(m,3H),7.12–7.00(m,3H),5.58(br.s,2H),5.02–4.87(m,2H),4.00–3.88(m,2H),3.02–2.92(m,2H),2.92–2.63(m,7H),2.63–2.51(m,2H),2.51–2.39(m,2H),2.31–2.08(m,6H),1.95–1.77(m,6H),1.67–1.52(m,2H),1.36–1.22(m,3H).
LCMS m/z=823.4[M+1]+
Example 18:
trifluoroacetate salt of 5- (3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) azetidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Compound 18)
5-(3-(4-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione trifluoroacetate
Figure BDA0003178773240001141
The first step is as follows: 3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) azetidine-1-carboxylic acid tert-butyl ester (18a)
tert-butyl 3-(4-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)azetidine-1-carboxylate
Figure BDA0003178773240001142
The trifluoroacetate salt (460mg) of the above crude 3- (4-phenoxyphenyl) -1- (4- (piperazin-1-yl) cyclohexyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (17e) is dissolved in 25mL of DCE, solid sodium bicarbonate (240mg, 2.86mmol) is added, stirring is carried out at room temperature for 20min, tert-butyl 3-oxaazetidine-1-carboxylate (240mg,1.40mmol) is added, stirring is carried out at room temperature for 30min, sodium triacetoxyborohydride (297mg, 1.40mmol) is added, and stirring is carried out at room temperature for 16H. To the reaction solution, 30mL of a saturated sodium bicarbonate solution was slowly added, extraction was performed with dichloromethane (30mL × 3), the organic phase was washed with 50mL of water, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give tert-butyl 3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) azetidine-1-carboxylate (18a) (400mg, two-step yield from compound 17 d: > 99%).
LCMS m/z=625.4[M+1]+
The second step is that: trifluoroacetic acid salt of 1- (4- (4- (azetidin-3-yl) piperazin-1-yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (18b)
1-(4-(4-(azetidin-3-yl)piperazin-1-yl)cyclohexyl)-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine trifluoroacetate
Figure BDA0003178773240001151
Tert-butyl 3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) azetidine-1-carboxylate (18a) (200mg,0.32mmol) was dissolved in 10mL DCM and 5mL trifluoroacetic acid was added and stirred at rt for 2H. The reaction system was concentrated under reduced pressure to give a crude 1- (4- (4- (azetidin-3-yl) piperazin-1-yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (18b) as the trifluoroacetate salt (260 mg).
LCMS m/z=525.3[M+1]+
The third step: trifluoroacetate salt of 5- (3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) azetidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Compound 18)
5-(3-(4-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione trifluoroacetate
Figure BDA0003178773240001161
The above crude trifluoroacetate salt (260mg) of 1- (4- (4- (azetidin-3-yl) piperazin-1-yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (18b) was dissolved in 10mL of DMSO, solid sodium bicarbonate (121mg, 1.44mmol) was added and stirred at room temperature for 10min, 0.5mL of DIPEA and 2- (2, 6-dioxopiperidin-3-yl) -5-fluoroisoindoline-1, 3-dione (synthesis see WO2017197056) (120mg, 0.43mmol) were added and the reaction stirred at 80 ℃ for 5H. The reaction mixture was cooled to room temperature, 20mL of water was added, the mixture was filtered, the filter cake was collected, the filter cake was washed with 10mL of water, the solid was dissolved in 100mL of dichloromethane, dried over anhydrous sodium sulfate, and the crude product was concentrated under reduced pressure and purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 15:1) to give a trifluoroacetate salt (75mg) of 5- (3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) azetidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (compound 18).
1H NMR(400MHz,CDCl3)δ9.27(br.s,1H),8.42–8.36(m,1H),7.69–7.60(m,3H),7.43–7.34(m,2H),7.21–7.03(m,5H),6.81–6.75(m,1H),6.56–6.48(m,1H),5.70(br.s,2H),4.99–4.70(m,2H),4.16–4.04(m,2H),3.95–3.84(m,2H),3.49–3.35(m,1H),2.95–2.78(m,2H),2.78–2.65(m,4H),2.65–2.50(m,3H),2.50–2.35(m,2H),2.26–2.08(m,4H),2.02–1.66(m,5H),1.66–1.50(m,1H).
LCMS m/z=781.3[M+1]+
Example 18-1:
5- (3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) azetidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (compound 18)
5-(3-(4-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)azetidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione
Figure BDA0003178773240001162
The trifluoroacetate salt (85mg) of 5- (3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) azetidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (compound 18) was dissolved in 50mL of dichloromethane and 5mL of methanol, washed with saturated aqueous sodium bicarbonate solution (30 mL. times.3), the organic layer was dried over anhydrous sodium sulfate, concentrated under reduced pressure, 10mL of petroleum ether and 2mL of ethyl acetate were added to the residue, stirred for 0.5H, filtered, and the filter cake was dried under vacuum to give 5- (3- (4- (4-amino-3-) (5- (4-amino-3-) (compound 18) 4-Phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) azetidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (compound 18) (60 mg).
1H NMR(400MHz,CDCl3)δ9.12–8.90(m,1H),8.42–8.34(m,1H),7.69–7.60(m,3H),7.43–7.34(m,2H),7.22–7.02(m,5H),6.81–6.75(m,1H),6.56–6.48(m,1H),5.64(br.s,2H),4.99–4.70(m,2H),4.16–4.04(m,2H),3.95–3.85(m,2H),3.50–3.35(m,1H),2.95–2.78(m,2H),2.78–2.63(m,4H),2.63–2.50(m,3H),2.50–2.35(m,2H),2.28–2.05(m,5H),1.95–1.45(m,5H).
LCMS m/z=781.3[M+1]+
Example 19:
trifluoroacetic acid salt of 5- (3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) - [1,3 '-diazacyclobutane ] -1' -yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Compound 19)
5-(3-(4-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)-[1,3'-biazetidin]-1'-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione trifluoroacetate
Figure BDA0003178773240001171
The first step is as follows: 3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) - [1,3 '-diazacyclobutane ] -1' -carboxylic acid tert-butyl ester (19a)
tert-butyl 3-(4-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)-[1,3'-biazetidine]-1'-carboxylate
Figure BDA0003178773240001181
The trifluoroacetate salt (260mg) of the above crude 1- (4- (4- (azetidin-3-yl) piperazin-1-yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (18b) was dissolved in 25mL of DCE, solid sodium bicarbonate (135mg, 1.61mmol) was added and stirred at room temperature for 20min, tert-butyl 3-oxoazetidine-1-carboxylate (110mg,0.64mmol) was added and stirred at room temperature for 30min, sodium triacetoxyborohydride (136mg, 0.64mmol) was added and stirred at room temperature for 16H. To the reaction solution, 30mL of a saturated aqueous sodium bicarbonate solution was slowly added, extraction was performed with dichloromethane (30mL × 3), the organic phase was washed with 50mL of water, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1-8:1) to give tert-butyl 3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) - [1,3 '-diazacyclobutane ] -1' -carboxylate (19a) (110mg, two-step yield from compound 18 a: 51%).
LCMS m/z=680.4[M+1]+
The second step is that: trifluoroacetate salt of 1- (4- (4- ([1,3' -diaza-but ] -3-yl) piperazin-1-yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (19b)
1-(4-(4-([1,3'-biazetidin]-3-yl)piperazin-1-yl)cyclohexyl)-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine trifluoroacetate
Figure BDA0003178773240001182
Tert-butyl 3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) - [1,3 '-diazacyclobutane ] -1' -carboxylate (19a) (100mg,0.15mmol) was dissolved in 10mL of dichloromethane, 5mL of trifluoroacetic acid was added, and the mixture was stirred at room temperature for 2H. The reaction system was concentrated under reduced pressure to give a crude 1- (4- (4- ([1,3' -bisazetidin ] -3-yl) piperazin-1-yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (19b) as the trifluoroacetate salt (140 mg).
LCMS m/z=580.3[M+1]+
The third step: trifluoroacetic acid salt of 5- (3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) - [1,3 '-diazacyclobutane ] -1' -yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Compound 19)
5-(3-(4-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperazin-1-yl)-[1,3'-biazetidin]-1'-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione trifluoroacetate
Figure BDA0003178773240001191
The trifluoroacetate salt (140mg) of the crude 1- (4- (4- ([1,3' -diazepin ] -3-yl) piperazin-1-yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (19b) described above is dissolved in 10mL of DMSO, solid sodium bicarbonate (74mg, 0.88mmol) is added, stirring is carried out at room temperature for 10min, 0.5mL of DIPEA and 2- (2, 6-dioxopiperidin-3-yl) -5-fluoroisoindoline-1, 3-dione (synthesis is described in WO2017197056) (60mg, 0.22mmol) are added, and the reaction is stirred at 80 ℃ for 5H. The reaction mixture was cooled to room temperature, 20mL of water was added, the mixture was filtered, the filter cake was collected, the filter cake was washed with 10mL of water, the solid was dissolved in 100mL of dichloromethane, dried over anhydrous sodium sulfate, and the crude product was concentrated under reduced pressure and purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 15:1) to give a trifluoroacetate salt (40mg) of 5- (3- (4- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperazin-1-yl) - [1,3 '-diazacyclobutane ] -1' -yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (compound 19).
1H NMR(400MHz,CDCl3)δ8.94(br.s,1H),8.38(s,1H),7.70–7.60(m,3H),7.43–7.35(m,2H),7.21–7.05(m,5H),6.77(d,1H),6.50(dd,1H),5.60(br.s,2H),5.01–4.87(m,2H),4.07–3.98(m,2H),3.88–3.81(m,2H),3.71–3.62(m,1H),3.55–3.47(m,2H),3.16–3.01(m,3H),2.92–2.65(m,5H),2.56–2.36(m,6H),2.26–2.08(m,3H),1.95–1.77(m,5H),1.77–1.62(m,2H).
LCMS m/z=836.4[M+1]+
Example 20:
trifluoroacetate salt of 5- (4- ((1- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperidin-4-yl) methyl) piperazin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Compound 20)
5-(4-((1-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperidin-4-yl)methyl)piperazin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione trifluoroacetate
Figure BDA0003178773240001201
The first step is as follows: 4- ((1- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperidin-4-yl) methyl) piperazine-1-carboxylic acid tert-butyl ester (20a)
tert-butyl 4-((1-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperidin-4-yl)methyl)piperazine-1-carboxylate
Figure BDA0003178773240001202
4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohex-1-one (17c) (200mg,0.50mmol) was dissolved in 15mL of DCE, acetic acid (0.25mL,4.375mmol) and tert-butyl 4- (piperidin-4-ylmethyl) piperazine-1-carboxylate (284mg,1.00mmol) were added (see WO2020051564 for synthesis), stirred at room temperature for 30min, sodium triacetoxyborohydride (212mg,1.00mmol) was added, and stirred at room temperature for 16H. To the reaction solution, 30mL of a saturated sodium bicarbonate solution was slowly added, extraction was performed with 100mL of dichloromethane, the organic phase was washed with 100mL of water, dried over anhydrous sodium sulfate, and concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 15:1) to give tert-butyl 4- ((1- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperidin-4-yl) methyl) piperazine-1-carboxylate (20a) (170mg, yield: 51%).
LCMS m/z=667.4[M+1]+
The second step is that: trifluoroacetic acid salt of 3- (4-phenoxyphenyl) -1- (4- (4- (piperazin-1-ylmethyl) piperidin-1-yl) cyclohexyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (20b)
3-(4-phenoxyphenyl)-1-(4-(4-(piperazin-1-ylmethyl)piperidin-1-yl)cyclohexyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine trifluoroacetate
Figure BDA0003178773240001211
Tert-butyl 4- ((1- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperidin-4-yl) methyl) piperazine-1-carboxylate (20a) (150mg,0.22mmol) was dissolved in 8mL of dichloromethane, 3mL of trifluoroacetic acid was added, and the mixture was stirred at room temperature for 2H. The reaction system was concentrated under reduced pressure to give a crude product of 3- (4-phenoxyphenyl) -1- (4- (4- (piperazin-1-ylmethyl) piperidin-1-yl) cyclohexyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (20b) as the trifluoroacetate salt (0.22 g).
LCMS m/z=567.4[M+1]+
The third step: trifluoroacetate salt of 5- (4- ((1- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperidin-4-yl) methyl) piperazin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Compound 20)
5-(4-((1-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperidin-4-yl)methyl)piperazin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione trifluoroacetate
Figure BDA0003178773240001212
The above crude trifluoroacetate salt of 3- (4-phenoxyphenyl) -1- (4- (4- (piperazin-1-ylmethyl) piperidin-1-yl) cyclohexyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (20b) (0.22g) was dissolved in 5mL of DMSO, solid sodium bicarbonate (95mg, 1.13mmol) was added and stirred at room temperature for 10min, 0.5mL of DIPEA and 2- (2, 6-dioxopiperidin-3-yl) -5-fluoroisoindoline-1, 3-dione (see WO2017197056 for synthesis) (93mg, 0.34mmol) were added and the reaction was stirred at 80 ℃ for 5H. The reaction mixture was cooled to room temperature, 20mL of water was added, the filtrate was collected, the filter cake was washed with 10mL of water, the solid was dissolved in 100mL of dichloromethane, dried over anhydrous sodium sulfate, and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 15:1) after concentration under reduced pressure to give a trifluoroacetate salt (100mg) of 5- (4- ((1- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperidin-4-yl) methyl) piperazin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (compound 20).
1H NMR(400MHz,CDCl3)δ8.39(s,1H),7.72–7.60(m,3H),7.43–7.34(m,2H),7.30–7.26(m,1H),7.20–7.11(m,3H),7.11–7.01(m,3H),5.70(br.s,2H),5.03–4.68(m,2H),3.47–3.37(m,4H),3.23–2.94(m,2H),2.93–2.66(m,3H),2.60–2.50(m,4H),2.50–2.38(m,2H),2.38–2.08(m,9H),2.05–1.93(m,1H),1.93–1.68(m,5H),1.68–1.48(m,2H).
LCMS m/z=823.4[M+1]+
Example 20-1:
5- (4- ((1- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperidin-4-yl) methyl) piperazin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Compound 20)
5-(4-((1-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)piperidin-4-yl)methyl)piperazin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione
Figure BDA0003178773240001221
The trifluoroacetate salt (60mg) of 5- (4- ((1- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperidin-4-yl) methyl) piperazin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (compound 20) was dissolved in 50mL of dichloromethane and 5mL of methanol, washed with a saturated aqueous sodium bicarbonate solution (30 mL. times.3), the organic layer was dried over anhydrous sodium sulfate, concentrated under reduced pressure, the residue was added with 10mL of petroleum ether and 2mL of ethyl acetate, stirred for 0.5H, filtered, and the filter cake was dried under vacuum to give 5- (4- ((1- (4- (4-amino-3- (4-Phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) piperidin-4-yl) methyl) piperazin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (compound 20) (40 mg).
1H NMR(400MHz,CDCl3)δ8.39(s,1H),7.72–7.60(m,3H),7.43–7.34(m,2H),7.30–7.26(m,1H),7.20–7.11(m,3H),7.11–7.01(m,3H),5.71(br.s,2H),5.03–4.68(m,2H),3.47–3.35(m,4H),3.17–2.92(m,2H),2.92–2.64(m,3H),2.60–2.50(m,4H),2.50–2.35(m,2H),2.38–1.96(m,10H),1.92–1.65(m,5H),1.65–1.45(m,2H).
LCMS m/z=823.4[M+1]+
Example 21:
5- (3- (7- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) -2, 7-diazaspiro [3.5] non-2-yl) azetidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Compound 21)
5-(3-(7-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)-2,7-diazaspiro[3.5]nonan-2-yl)azetidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione
Figure BDA0003178773240001231
The first step is as follows: 7- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) -2, 7-diazaspiro [3.5] nonane-2-carboxylic acid tert-butyl ester (21a)
tert-butyl 7-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)-2,7-diazaspiro[3.5]nonane-2-carboxylate
Figure BDA0003178773240001232
4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohex-1-one (17c) (150mg,0.38mmol) was dissolved in 15mL of DCE, acetic acid (0.25mL,4.375mmol) and tert-butyl 2, 7-diazaspiro [3.5] non-2-carboxylate (171mg,0.76mmol) were added, stirred at room temperature for 30min, sodium triacetoxyborohydride (160mg,0.75mmol) was added, and stirred at room temperature for 16H. To the reaction solution, 30mL of a saturated sodium bicarbonate solution was slowly added, extraction was performed with 100mL of dichloromethane, the organic phase was washed with 100mL of water, dried over anhydrous sodium sulfate, and concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give tert-butyl 7- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) -2, 7-diazaspiro [3.5] nonane-2-carboxylate (21a) (70mg, yield: 30%).
LCMS m/z=610.3[M+1]+
The second step is that: trifluoroacetate salt of 1- (4- (2, 7-diazaspiro [3.5] non-7-yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (21b)
1-(4-(2,7-diazaspiro[3.5]nonan-7-yl)cyclohexyl)-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine trifluoroacetate
Figure BDA0003178773240001241
Tert-butyl 7- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) -2, 7-diazaspiro [3.5] nonane-2-carboxylate (21a) (70mg,0.11mmol) was dissolved in 8mL of dichloromethane, 3mL of trifluoroacetic acid was added, and the mixture was stirred at room temperature for 2H. The reaction system was concentrated under reduced pressure to give a crude 1- (4- (2, 7-diazaspiro [3.5] non-7-yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (21b) as the trifluoroacetate salt (0.12 g).
LCMS m/z=510.3[M+1]+
The third step: 3- (7- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) -2, 7-diazaspiro [3.5] non-2-yl) azetidine-1-carboxylic acid tert-butyl ester (21c)
tert-butyl 3-(7-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)-2,7-diazaspiro[3.5]nonan-2-yl)azetidine-1-carboxylate
Figure BDA0003178773240001242
The trifluoroacetate salt (0.12g) of the above crude 1- (4- (2, 7-diazaspiro [3.5] non-7-yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (21b) was dissolved in 15mL of DCE, solid sodium bicarbonate (48mg, 0.57mmol) was added and stirred at room temperature for 20min, tert-butyl 3-oxoazetidine-1-carboxylate (40mg,0.23mmol) was added and stirred at room temperature for 30min, sodium triacetoxyborohydride (50mg, 0.24mmol) was added and stirred at room temperature for 16H. To the reaction solution, 30mL of a saturated sodium bicarbonate solution was slowly added, extraction was performed with dichloromethane (30mL × 3), the organic phase was washed with 50mL of water, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give tert-butyl 3- (7- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) -2, 7-diazaspiro [3.5] non-2-yl) azetidine-1-carboxylate (21c) (35mg, two-step yield from compound 21 a: 48%).
LCMS m/z=665.4[M+1]+
The fourth step: 1- (4- (2- (azetidin-3-yl) -2, 7-diazaspiro [3.5] non-7-yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (21d) trifluoroacetate
1-(4-(2-(azetidin-3-yl)-2,7-diazaspiro[3.5]nonan-7-yl)cyclohexyl)-3-(4-phenoxy-phenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine trifluoroacetate
Figure BDA0003178773240001251
Tert-butyl 3- (7- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) -2, 7-diazaspiro [3.5] non-2-yl) azetidine-1-carboxylate (21c) (35mg,0.05mmol) was dissolved in 10mL DCM, and 5mL trifluoroacetic acid was added and stirred at room temperature for 2H. The reaction system was concentrated under reduced pressure to give crude 1- (4- (2- (azetidin-3-yl) -2, 7-diazaspiro [3.5] non-7-yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (21d) trifluoroacetate (60 mg).
LCMS m/z=565.3[M+1]+
The fifth step: 5- (3- (7- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) -2, 7-diazaspiro [3.5] non-2-yl) azetidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Compound 21)
5-(3-(7-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)-2,7-diazaspiro[3.5]nonan-2-yl)azetidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione
Figure BDA0003178773240001252
The crude 1- (4- (2- (azetidin-3-yl) -2, 7-diazaspiro [3.5] non-7-yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (21d) trifluoroacetate (60mg) was dissolved in 5mL of DMSO, solid sodium bicarbonate (21mg, 0.25mmol) was added and stirred at room temperature for 10min, 0.5mL of DIPEA and 2- (2, 6-dioxopiperidin-3-yl) -5-fluoroisoindoline-1, 3-dione (synthesis see WO2017197056) (21mg, 0.08mmol) were added and the reaction stirred at 80 ℃ for 5H. The reaction mixture was cooled to room temperature, 20mL of water was added, the mixture was filtered, the filter cake was collected, the filter cake was washed with 10mL of water, the solid was dissolved in 100mL of dichloromethane, dried over anhydrous sodium sulfate, and the crude product was concentrated under reduced pressure to be purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 15:1) to give 5- (3- (7- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) -2, 7-diazaspiro [3.5] non-2-yl) azetidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindolin-1, 3-dione (compound 21) (15mg, two-step yield from compound 21 c: 37%).
1H NMR(400MHz,CDCl3)δ8.49(br.s,1H),8.38(s,1H),7.68–7.60(m,3H),7.43–7.34(m,2H),7.21–7.04(m,5H),6.79–6.74(m,1H),6.54–6.47(m,1H),5.52(br.s,2H),5.13–4.68(m,2H),4.07–3.98(m,2H),3.86–3.77(m,2H),3.72–3.61(m,1H),3.13–3.04(m,4H),2.95–2.65(m,4H),2.65–2.37(m,5H),2.26–2.08(m,4H),1.97–1.54(m,7H).
LCMS m/z=821.3[M+1]+
Example 22:
5- (3- (5- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) azetidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Compound 22)
5-(3-(5-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)azetidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione
Figure BDA0003178773240001261
The first step is as follows: 5- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) hexahydropyrrolo [3,4-c ] pyrrole-2 (1H) -carboxylic acid tert-butyl ester (22a)
tert-butyl 5-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate
Figure BDA0003178773240001271
4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohex-1-one (17c) (150mg,0.38mmol) was dissolved in 15mL of DCE, acetic acid (0.25mL,4.375mmol) and tert-butyl hexahydropyrrolo [3,4-c ] pyrrole-2 (1H) -carboxylate (160mg,0.75mmol) were added, stirring at room temperature for 30min, sodium triacetoxyborohydride (160mg,0.75mmol) was added, and stirring at room temperature for 16H. To the reaction solution, 30mL of a saturated sodium bicarbonate solution was slowly added, extraction was performed with 100mL of dichloromethane, the organic phase was washed with 100mL of water, dried over anhydrous sodium sulfate, and concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give tert-butyl 5- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) hexahydropyrrolo [3,4-c ] pyrrole-2 (1H) -carboxylate (22a) (120mg, yield: 53%).
LCMS m/z=596.3[M+1]+
The second step is that: trifluoroacetate salt of 1- (4- (hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) cyclohexyl) -3- (4-phenoxy-phenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (22b)
1-(4-(hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)cyclohexyl)-3-(4-phenoxy-phenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine trifluoroacetate
Figure BDA0003178773240001272
Tert-butyl 5- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) hexahydropyrrolo [3,4-c ] pyrrole-2 (1H) -carboxylate (22a) (120mg,0.20mmol) was dissolved in 8mL DCM and 3mL trifluoroacetic acid was added. Stirred at room temperature for 2 h. The reaction system was concentrated under reduced pressure to give a crude 1- (4- (hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) cyclohexyl) -3- (4-phenoxy-phenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (22b) as the trifluoroacetate salt (145 mg).
LCMS m/z=496.3[M+1]+
The third step: 3- (5- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) azetidine-1-carboxylic acid tert-butyl ester (22c)
tert-butyl 3-(5-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)azetidine-1-carboxylate
Figure BDA0003178773240001281
The trifluoroacetate salt (145mg) of the above crude 1- (4- (hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) cyclohexyl) -3- (4-phenoxy-phenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (22b) was dissolved in 15mL DCE, solid sodium bicarbonate (85mg, 1.00mmol) was added and stirred at room temperature for 20min, tert-butyl 3-oxoazetidine-1-carboxylate (68mg,0.40mmol) was added and stirred at room temperature for 30min, sodium triacetoxyborohydride (85mg, 0.40mmol) was added and stirred at room temperature for 16H. To the reaction solution, 30mL of a saturated sodium bicarbonate solution was slowly added, extraction was performed with dichloromethane (30mL × 3), the organic phase was washed with 50mL of water, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20:1) to give tert-butyl 3- (5- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) azetidine-1-carboxylate (22c) (110mg, two-step yield from compound 22 a: 85%).
LCMS m/z=651.4[M+1]+
The fourth step: trifluoroacetic acid salt of 1- (4- (5- (azetidin-3-yl) hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (22d)
1-(4-(5-(azetidin-3-yl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)cyclohexyl)-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine trifluoroacetate
Figure BDA0003178773240001282
Tert-butyl 3- (5- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) azetidine-1-carboxylate (22c) (100mg,0.15mmol) was dissolved in 10mL DCM and 5mL trifluoroacetic acid was added and stirred at room temperature for 2H. The reaction system was concentrated under reduced pressure to give a crude 1- (4- (5- (azetidin-3-yl) hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (22d) as the trifluoroacetate salt (130 mg).
LCMS m/z=551.3[M+1]+
The fifth step: 5- (3- (5- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) azetidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Compound 22)
5-(3-(5-(4-(4-amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)cyclohexyl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)azetidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione
Figure BDA0003178773240001291
The trifluoroacetate salt (130mg) of the crude 1- (4- (5- (azetidin-3-yl) hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) cyclohexyl) -3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-4-amine (22d) was dissolved in 5mL DMSO, solid sodium bicarbonate (63mg, 0.75mmol) was added, stirring was carried out at room temperature for 10min, 0.5mL DIPEA and 2- (2, 6-dioxopiperidin-3-yl) -5-fluoroisoindoline-1, 3-dione (synthesis see WO2017197056) (64mg, 0.23mmol) were added and the reaction was stirred at 80 ℃ for 5H. The reaction mixture was cooled to room temperature, 20mL of water was added, and the mixture was filtered, and the filter cake was collected, washed with 10mL of water, the solid was dissolved in 100mL of dichloromethane, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude product which was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 15:1) to give 5- (3- (5- (4- (4-amino-3- (4-phenoxyphenyl) -1H-pyrazolo [3,4-d ] pyrimidin-1-yl) cyclohexyl) hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) azetidin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (compound 22) (70mg, two-step yield from compound 22 c: 58%).
1H NMR(400MHz,CDCl3)δ9.24(br.s,1H),8.39(s,1H),7.69(d,2H),7.60(d,1H),7.31(t,2H),7.17–7.06(m,3H),7.04–6.92(m,2H),6.72(d,1H),6.47–6.39(m,1H),5.70(br.s,2H),4.97–4.78(m,2H),4.07–3.97(m,2H),3.94–3.84(m,2H),3.56–3.46(m,1H),3.00–2.64(m,6H),2.62–2.41(m,5H),2.40–2.20(m,2H),2.16–2.05(m,2H),2.04–1.85(m,4H),1.85–1.74(m,2H),1.74–1.55(m,2H).
LCMS m/z=807.3[M+1]+
Test example
1. Cell proliferation inhibition assay
The Mino culture medium is RPMI1640+ 10% FBS, and is cultured at 37 deg.C and 5% CO2An incubator. Cells were plated in 96-well plates. Wherein, the Mino cells are 5000 cells/well, and each well is 90 mu L. Each well was dosed with 10 μ L of compound at different concentrations. Each concentration was set to 3 more wells, the last column was DMSO vehicle control, at 37 ℃, 5% CO2The culture was continued under the conditions for 72 hours. After 72 hours, 100. mu.L of detection reagent (Ce) was added to each wellll visual Assay, Promega, G7573), mixed for 2 minutes, incubated at room temperature for 10 minutes, and the fluorescence signal value was measured with a microplate reader (PHERAstar FSX). Using origin9.2 software, the IC of compounds to inhibit cell proliferation was calculated50The value, and the inhibition Max inhi% at the highest concentration of compound was calculated according to formula (1).
Max inhi.%=(1-T72 administration of drugs/T72 vehicle) X 100% of formula (1).
IC for inhibiting Mino cell proliferation50The results are shown in Table 1.
TABLE 1 IC for inhibition of Mino cell proliferation50Value of
Serial number Compound numbering IC50(nM)
1 Compound 8 73
2 Compound 12 412
3 Compound 17 186
4 Trifluoroacetic acid salt of Compound 18 21
5 Trifluoroacetic acid salt of Compound 19 80
6 Trifluoroacetic acid salt of Compound 20 111
7 Compound 21 147
8 Compound 22 286
And (4) conclusion: the compound synthesized by the technology has a certain inhibition effect on the proliferation of Mino cells (mantle cell lymphoma cells). Specific values of activity are shown in Table 1.
2. In vitro BTK C481S kinase assay
Kinase BTK C481S (Carna, Cat.No. 08-547) was prepared into 2.5X kinase solution, substrates FAM-P2(GL Biochem, Cat.No.112394) and ATP ((Sigma, Cat.No. A7699-1G) were prepared into 2.5X substrate solution, 5. mu.L of compounds with different concentrations were added to 384-well plates, 10. mu.L of 2.5X kinase solution was added, incubated at room temperature for 10 minutes, 10. mu.L of 2.5X substrate solution was added, after incubation at 28 ℃ for an appropriate time, 30. mu.L of stop solution was added to terminate the reaction, and the detection was performed using a Caliper EZ reader2 instrument, the inhibition calculation formula is shown in formula 2, where max is the DMSO control reading, min is the negative control reading, and conversion is the compound reading, and IC was calculated using XLIPT extended-in version5.4.0.8 software according to the inhibition value50The value is obtained.
Inhibition ratio [% ], [ ([ max-conversion ]/[ max-min ] ]) [% ], and [ 100% ]
The results of the activity test are shown in table 2:
TABLE 2 IC of the Compounds of the invention against BTK C481S kinase in vitro50(nM)
Compound (I) BTK C481S IC50(nM)
Compound 1 17nM
Compound 6 21nM
Compound 7 24nM
Compound 8 19nM
Compound 13 23nM
Compound 14 28nM
Compound 15 47nM
Compound 16 45nM
And (4) conclusion: the compound disclosed by the invention has good inhibitory activity on in vitro BTK C481S kinase.
3. Pharmacokinetic testing of rats
Purpose of the experiment: in this test, the test substance is administered to SD rats via single-dose vein and gavage, and the concentration of the test substance in the plasma of the rats is measured to evaluate the pharmacokinetic characteristics and bioavailability of the test substance in the rats.
Test animals: male SD rats, about 200g, 6-8 weeks old, 6 per compound. Purchased from Woods laboratories Inc.
The test method comprises the following steps: on the day of the experiment, 6 SD rats were randomly grouped by body weight. The food is fasted for 12-14 h before administration for 1 day, and is fed for 4h after administration.
Figure BDA0003178773240001311
Figure BDA0003178773240001321
Doses are in free base.
Sampling
Before and after administration isoflurane anesthesia 0.1mL of blood was drawn from the orbit and placed in an EDTAK2 centrifuge tube. Centrifuging at 5000rpm and 4 deg.C for 10min, and collecting plasma.
Plasma time points were collected in groups G1& G2:
G1:0,5min,15min,30min,1,2,4,6,8,24h;
G2:0,15min,30min,1,2,4,6,8,24h.
all samples were stored at-80 ℃ before analytical testing. The samples were tested by HPLC-MS/MS.
Table 3 pharmacokinetic parameters of the compounds in rat plasma
Test compounds Mode of administration AUC0-t(ng/ml·h) F(%)
Compound 18 i.g.(20mg/kg) 5698±750 19.6±2.6
Compound 20 i.g.(20mg/kg) 560±147 6.78±1.8
Injecting: compound was administered i.g. (gavage).
And (4) conclusion: the compound synthesized by the technology of the invention has certain oral bioavailability in rat bodies.

Claims (16)

1. A compound of formula (I) or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein
B-L-K (I);
L is selected from-Ak 1-Cy1-Ak2-Cy2-Ak3-Cy3-Ak4-Cy4-Ak 5-;
ak1, Ak2, Ak3, Ak4 and Ak5 are each independently selected from CH2、O、-NRb0-C ═ O, C ≡ C or a bond;
cy1, Cy2, Cy3 and Cy4 are each independently selected from 3 to 12 membered heterocyclic ring, 3 to 12 membered cycloalkyl, 6 to 10 membered aryl or bond, said heterocyclic, cycloalkyl or aryl being optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、oxo、CF3、C1-4Alkyl, hydroxy substituted C1-4Alkyl, halogen substituted C1-4Alkyl, CN, COOH orC1-4Alkoxy, said heterocycle containing 1 to 4 heteroatoms selected from O, S, N;
b is selected from B1-W1-B2-B3-B4-or
Figure FDA0003178773230000011
When B is selected from B1-W1-B2-B3-B4-, Cy1, Cy2, Cy3 and Cy4 cannot be bonds at the same time;
when B is selected from B1-W1-B2-B3-B4-, Ak1, Ak2, Ak3, Ak4 or Ak5 is not a bond, the two can not be directly connected with each other;
b1 is selected from 6-membered heteroaromatic ring or phenyl further optionally substituted with 0 to 4Rb1(iii) substituted, said heteroaromatic ring contains 1 to 4 heteroatoms selected from O, S, N;
w1 is selected from-O-, -S-, -NH-, -NHCO-or-CONH-;
b2 is selected from 6-membered heteroaromatic ring or phenyl optionally further optionally substituted with 0 to 4Rb2(iii) substituted, said heteroaromatic ring contains 1 to 4 heteroatoms selected from O, S, N;
b3 is selected from 8-10 membered heterorings, optionally further optionally substituted with 0 to 4Rb3(iii) substituted, said heteroring contains 1 to 4 heteroatoms selected from O, S, N;
b4 is selected from the group consisting of 4-7 membered monocycloalkyl, 6-12 membered spirocycloalkyl, 5-10 membered benzocycloalkyl, and 7-10 membered bridged cycloalkyl, said monocycloalkyl, spirocycloalkyl, benzocycloalkyl, and bridged cycloalkyl groups optionally further optionally substituted with 0 to 4Rb4Substituted;
B5or B6Each independently selected from one of the following substituted or unsubstituted groups: phenyl or 5-6 membered heteroaryl, when substituted, optionally further substituted with 0 to 4Rb5(iii) substituted, said heteroaryl contains 1 to 4 heteroatoms selected from O, S, N;
Rb0selected from H, C1-4Alkyl, 3 to 12 membered cycloalkyl or 3 to 12 membered heterocycle containing 1 to 4 heteroatoms selected from O, S, N;
Rb1、Rb2、Rb3or Rb4Each independently selected from H, F, Cl, Br, I, OH, NH2、CN、CF3、COOH、CONH2、C1-4Alkyl, hydroxy substituted C1-4Alkyl, halogen substituted C1-4Alkyl or C1-4Alkoxy, said alkyl and alkoxy being optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、CN、CONH2、C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
Rb5each independently selected from H, F, Cl, Br, I, OH, NH2、CN、CF3、-C(=O)NH2、-C(=O)NH-C1-4Alkyl, -C (═ O) N (C)1-4Alkyl radical)2、C1-4Alkyl or C1-4Alkoxy, said alkyl or alkoxy being optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I or OH;
Rb6selected from H, C1-4Alkyl, halogen substituted C1-4Alkyl or hydroxy substituted C1-4An alkyl group;
Rb7、Rb8each independently selected from H, F, Cl, Br, I, OH, NH2、CN、CF3、-C(=O)NH2、-C(=O)NH-C1-4Alkyl, -C (═ O) N (C)1-4Alkyl radical)2、C1-4Alkyl or C1-4Alkoxy, said alkyl or alkoxy being optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I or OH;
or Rb7、Rb8Taken together with the carbon atoms to which they are attached form a 5-6 membered heteroaryl or phenyl group optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、CN、CF3、-C(=O)NH2、-C(=O)NH-C1-4Alkyl, -C (═ O) N (C)1-4Alkyl radical)2、C1-4Alkyl or C1-4Alkoxy, said heteroaryl having 1 to 4 heteroatoms selected from O, S, N;
m is selected from 0, 1 or 2;
k is selected from
Figure FDA0003178773230000021
Figure FDA0003178773230000022
Figure FDA0003178773230000031
Ring E or F is selected from a benzene ring or a 5-6 membered heteroaromatic ring containing 1 to 2 heteroatoms selected from O, S, N;
Rk2is selected from CH2、C=O、S=O、SO2
Rk1、Rk3Or Rk4Each independently selected from H, F, Cl, Br, I, OH, NH2、CF3、CN、COOH、C1-4Alkyl or C1-4An alkoxy group;
Rk5is selected from C ═ O or
Figure FDA0003178773230000032
M1Is selected from the group consisting of a bond, -CH2-C (═ O) NH-or-C (═ O) CH2NH-;
M2Is selected from-NHC (═ O) -C1-6Alkyl, 8-10 membered hetero-cyclic or-NHC (═ O) -C3-6Cycloalkyl, said alkyl, hetero-cyclo or cycloalkyl optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、C1-4Alkyl, oxo or C1-4Substituted by a substituent of alkoxy;
M3is selected from-NH-or-O-;
Rk6is selected from C1-6Alkyl, said alkyl being optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, C1-6Alkyl or C3-6Cycloalkyl, substituted with a substituent;
Rk7each independently selected from H, F, Cl, Br, I, OH, SH, C1-6Alkyl radical, C1-6Alkoxy or C1-6Alkylthio radical, C1-6Alkyl formyloxy, said alkyl, alkoxy or alkylthio being optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
Rk8、Rk9each independently selected from H, C1-6Alkyl or C3-6Cycloalkyl, said alkyl or cycloalkyl being optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
Rk10selected from 5-6 membered heteroaryl, said heteroaryl optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, CF3、CN、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl radical, C1-4Alkoxy or C3-6Cycloalkyl, substituted with a substituent;
g is selected from 6-10 membered aryl or 5-10 membered heteroaryl, said aryl or heteroaryl being optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, CF3、CN、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl radical, C1-4Alkoxy or C3-6Cycloalkyl, said heteroaryl containing 1 to 4 heteroatoms selected from N, O, S;
p1 or p2 are each independently selected from 0, 1,2,3 or 4.
2. A compound according to claim 1 or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein
Cy1, Cy2, Cy3 and Cy4 are each independently selected from a bond, a 4-7 membered heteromonocyclic ring, a 5-10 membered heterobicyclic ring, a 6-12 membered heterospirocyclic ring, a 7-10 membered heterobridged ring, a 4-7 membered monocycloalkyl, a 5-10 membered heterocycloalkyl, a 6-12 membered spirocycloalkyl, a 7-10 membered bridged cycloalkyl or a 6-10 membered aryl, all of whichThe aryl, cycloalkyl, heteromonocyclic, heterobicyclic, heterospirocyclic or heterobridged ring is optionally further substituted by 0 to 4 groups selected from H, F, Cl, Br, I, OH, NH2、oxo、CF3、COOH、CN、C1-4Alkyl, hydroxy substituted C1-4Alkyl, halogen substituted C1-4Alkyl or C1-4Alkoxy, said hetero-monocyclic, hetero-fused ring, hetero-spiro ring or hetero-bridged ring containing 1 to 4 heteroatoms selected from O, S, N;
b is selected from B1-W1-B2-B3-B4-;
b1 is selected from phenyl or pyridyl further optionally substituted with 0 to 4Rb1Substituted;
w1 is selected from-O-, -NHCO-, or-CONH-;
b2 is selected from phenyl or pyridyl further optionally substituted with 0 to 4Rb2Substituted;
b3 is selected from substituted or unsubstituted imidazopyrimidines, pyrazolopyrimidines, imidazopyrazines, pyrazolopyrazines, imidazotetrahydropyrimidines, pyrazolotetrahydropyrimidines, when substituted, optionally further substituted with 0 to 4Rb3Substituted;
b4 is selected from cyclohexyl, cyclopentyl and cyclobutyl, said cyclohexyl, cyclopentyl and cyclobutyl being further optionally substituted by 0 to 4Rb4Substituted;
k is selected from
Figure FDA0003178773230000041
Figure FDA0003178773230000042
Figure FDA0003178773230000051
Rk2Is selected from CH2Or C ═ O;
Rk1、Rk3or Rk4Each independently selected from H, F, Cl, Br, I, OH or NH2
M1Is selected from the group consisting of a bond, -CH2-C(=O) NH-or-C (═ O) CH2NH-;
M2Is selected from-NHC (═ O) -C1-6Alkyl, 8-10 membered hetero-cyclic or-NHC (═ O) -C3-6Cycloalkyl, said alkyl, hetero-cyclo or cycloalkyl optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、C1-4Alkyl, oxo or C1-4Substituted by a substituent of alkoxy;
Rk6is selected from C1-6Alkyl, said alkyl being optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, C1-6Alkyl or C3-6Cycloalkyl, substituted with a substituent;
Rk7each independently selected from H, F, Cl, Br, I, OH, SH, C1-6Alkyl radical, C1-6Alkoxy or C1-6Alkylthio radical, C1-6Alkyl formyloxy, said alkyl, alkoxy or alkylthio being optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
Rk8、Rk9each independently selected from H, C1-6Alkyl or C3-6Cycloalkyl, said alkyl or cycloalkyl being optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
Rk10selected from 5-6 membered heteroaryl, said heteroaryl optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, CF3、CN、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl radical, C1-4Alkoxy or C3-6Cycloalkyl, substituted with a substituent;
p1 or p2 are each independently selected from 0, 1,2,3 or 4.
3. A compound according to claim 2 or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein
Cy1, Cy2, Cy3 and Cy4 are each independently selected from substitutedOr one of the following unsubstituted groups: a bond, phenyl, naphthyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclobutylcyclobutyl, cyclobutylcyclopentyl, cyclobutylcyclohexyl, cyclopentylcyclopentyl, cyclopentylcyclohexyl, cyclohexylcyclohexylcyclohexyl, cyclopropylcyclobutylcyclopentyl, cyclopropylcyclopentylyl, cyclopropylcyclohexylcyclohexyl, cyclobutylspirocyclobutyl, cyclobutylspirocyclopentyl, cyclobutylspirocyclohexyl, cyclopentylpirocyclopentyl, cyclopentylpirocyclohexyl, cyclohexylspirocyclohexyl, cyclopropylspirocyclobutyl, cyclopropylspirocyclopentyl, cyclopropylspirocyclohexyl, azetidinyl, azepinyl, piperidinyl, morpholinyl, piperazinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazolyl, tetrazolyl, cyclopropylazacyclobutyl, cyclopropylazacyclopentyl, cyclopropylazacylclocyclohexyl, Cyclopropylazapiperidinyl, cyclobutylazacyclobutyl, cyclobutylazacyclopentyl, cyclobutylazacyclohexyl, cyclobutylazopiperidinyl, cyclopentazacyclobutyl, cyclopentazacyclopentyl, cyclopentazacyclohexyl, cyclopentazacyclohexadinyl, cyclopentazacyclobutyl, cyclopentazacyclopentyl, cyclohexylazacyclohexyl, cyclohexylazacyclobutylazetidinyl, azetidinylcyclopentyl, azetidinylcyclohexyl, azetidinylboroazetidinyl, azetidinylcyclopentyl, azetidinylcyclohexyl, azetidinylcyclopentylgroup, azetidinopentylazacyclopentyl, azetidinohexylazacyclohexadecylohexylyl, azacyclopentadipiperidinyl, azetidinylcyclobutyl, azetidinohexylazacyclopentyl, azacyclohexazacyclopentadinyl, azacyclohexadipinyl, azetidinyl, azetidinylcohexylazacyclohexadinyl, azacyclohexadinyl, azacyclohexylpiperidinyl, azacyclohexadipinyl, azacyclohexadinyl, and the like, Cyclobutyl spiroazetidinyl, cyclopentyl spiroazetidinyl, cyclohexyl spiroazetidinyl, cyclobutyl spiroazetidinyl, azetidinyl spiroazetidinyl, cyclopentyl spiroazetidinyl, cyclohexyl, cyclopentyl spiroazetidinyl, cyclohexyl, cyclopentyl, cyclohexyl, cyclopentyl spiroazetidinyl, cyclohexyl, cyclopentyl, cyclohexyl, cyclopentyl spiroazetidinyl, cyclohexyl, or cyclohexyl,An azacyclopentyl spiroazacyclohexyl group, an azacyclohexyl spiroazetidinyl group, a cyclobutyl spiropiperidinyl group, a cyclopentyl spiropiperidinyl group, a cyclohexyl spiropiperidinyl group, an azetidinyl spiropiperidinyl group, an azacyclopentyl spiropiperidinyl group, an azacyclohexyl spiropiperidinyl group, an azacyclopentyl spiropiperidinyl group, an azacyclohexyl spiropiperidinyl group, a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable carrier, a pharmaceutically,
Figure FDA0003178773230000061
Figure FDA0003178773230000062
When substituted, is optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、oxo、CF3、CN、C1-4Alkyl, CF3、CH2OH, COOH or C1-4Substituted by a substituent of alkoxy;
b is selected from
Figure FDA0003178773230000063
Figure FDA0003178773230000064
Rb1、Rb2、Rb3、Rb4Each independently selected from H, F, Cl, Br, I, OH, NH2、CN、CONH2、CF3COOH, hydroxymethyl, methyl or methoxy, said methyl or methoxy being optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I;
n1, n2, n3 and n4 are respectively and independently selected from 0, 1,2,3 or 4.
4. A compound according to claim 3 or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein
L is selected from
Figure FDA0003178773230000071
Figure FDA0003178773230000072
Or L may be selected from
Figure FDA0003178773230000073
Figure FDA0003178773230000074
Or L may be selected from
Figure FDA0003178773230000081
Figure FDA0003178773230000082
Or L may be selected from
Figure FDA0003178773230000083
Figure FDA0003178773230000084
The left side of the L is connected with the B, and the right side of the L is connected with the K;
b is selected from
Figure FDA0003178773230000085
Figure FDA0003178773230000086
Rb1、Rb2Each independently selected from H, F, Cl, Br, OH, NH2、CN、CF3Hydroxymethyl, methyl or methoxy;
k is selected from
Figure FDA0003178773230000091
Figure FDA0003178773230000092
5. The compound of claim 1, or a stereoisomer, deutero-compound, solvate, prodrug, metabolite, pharmaceutically acceptable salt, or co-crystal thereof, wherein,
b is selected from
Figure FDA0003178773230000101
At least 4 of Ak1, Ak2, Ak3, Ak4 and Ak5 are selected from bonds;
cy1, Cy2, Cy3 and Cy4 are each independently selected from a bond, a 4-7 membered azamonocyclic ring, a 5-10 membered azabicyclic ring, a 7-10 membered heterobridged ring or a 6-12 membered nitrogen-containing heterospirocyclic ring, said heteromonocyclic, heterofused, heterobridged or heterospirocyclic ring optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I, OH, COOH, CN, NH, and mixtures thereof2、oxo、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl or C1-4(ii) alkoxy, said heteromonocyclic, heterobicyclic, heterobridged, or heterospirocyclic ring containing 1 to 4 heteroatoms selected from O, S, N;
k is selected from
Figure FDA0003178773230000102
Figure FDA0003178773230000103
6. The compound of claim 5, or a stereoisomer, deutero-compound, solvate, prodrug, metabolite, pharmaceutically acceptable salt, or co-crystal thereof, wherein,
ak1, Ak2, Ak3, Ak4 and Ak5 are each independently selected from CH2O, or a bond, and at least 4 of Ak1, Ak2, Ak3, Ak4, and Ak5 are selected from the group consisting of bonds;
cy1, Cy2, Cy3 and Cy4 are each independently selected from one of the following substituted or unsubstituted groups: a bond, azetidinyl, azacyclopentyl, piperazinePyridine, morpholine, piperazine, cyclopropyloazetidinyl, cyclobutylazetidinyl, cyclobutylpipetidinyl, cyclopentaizetidinyl, cyclopentaietidactyl, cyclopentaietioazetidinyl, cyclopentaizaclidinyl, cyclopentaiclidinyl, cyclohexylazepinyl, cyclohexylazaclidinyl, azetidinyl, azetidinoazetidinyl, azetidinobutylazinyl, azetidinobutylazocycloazetidinyl, azetidinoazetidinyl, cyclopentaizacyclopentyl, azetidinohexyl, azetidinyl, azetidinoazetidinyl, cyclopentaizacyclobutylyl, cyclopentaizacylo-azacyclobutylyl, cyclopentaizacylohexyl, cyclopentaizacylpiperidine, azetidinyl, cyclopentaizacylobutyryl, cyclopentaizacylo-azacyclobutylyl, cyclopentaizacylohexyl, cyclopentaizacylohexylyl, cyclopentaizacylbutyl, and azetidinyl, Azetidinyl, azacyclohexylazapiperidyl, cyclobutyl spiroazetidinyl, cyclopentyl spiroazetidinyl, cyclohexyl spiroazetidinyl, azetidinyl spiroazetidinyl, azetidinyl spiroazetidinyl, cyclohexylspiropiperidinyl, azetidinyl spiroazetidinyl, cyclobutyl spiropiperidinyl, azetidinyl spiroazetidinyl, azetidinyl spiropiperidinyl, azetidinyl, and the like, An azacyclopentyl spiropiperidine, an azacyclohexyl spiropiperidine,
Figure FDA0003178773230000111
Figure FDA0003178773230000112
Figure FDA0003178773230000113
When substituted, is optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、COOH、CN、oxo、C1-4Alkyl, halogen substituted C1-4Alkyl, hydroxy substituted C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
B5or B6Each independently selected from substituted or unsubstituted phenyl or 6 membered heteroaryl, when substituted, optionally further substituted with 0 to 4Rb1(iii) substituted, said heteroaryl contains 1 to 3N atoms;
Rb5independently selected from H, F, Cl, Br, I, OH, CF3、C1-4Alkyl or C1-4Alkoxy, said alkyl or alkoxy being optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I;
Rb6selected from H, C1-4An alkyl group;
Rb7、Rb8each independently selected from H, F, Cl, Br, I, NH2、CF3、-C(=O)NH2、C1-4Alkyl or C1-4Alkoxy, said alkyl or alkoxy being optionally further substituted with 0 to 4 substituents selected from H, F, Cl, Br, I;
m is selected from 0 or 1;
k is selected from
Figure FDA0003178773230000121
Figure FDA0003178773230000122
Rk2Each independently selected from CH2Or C ═ O;
Rk1、Rk3or Rk4Each independently selected from H, CH3F, Cl, Br, I, OH or NH2
M1Is selected from the group consisting of a bond, -CH2-C (═ O) NH-or-C (═ O) CH2NH-;
M2Selected from-NHC (═ O) -methyl, -NHC (═ O) -ethyl, -NHC (═ O) -cyclopropyl, -NHC (═ O)O) -cyclobutyl, -NHC (═ O) -cyclopentyl or-NHC (═ O) -cyclohexyl, said methyl, ethyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl being optionally further substituted by 0 to 4 substituents selected from H, F, Cl, Br, I, OH, NH2、C1-4Alkyl or C1-4Substituted by a substituent of alkoxy;
Rk6selected from methyl, ethyl, propyl, isopropyl, tert-butyl, isobutyl or sec-butyl;
Rk7each independently selected from H, F, OH, SH, methyl, methoxy or-SCH3
Rk8、Rk9Each independently selected from H, methyl, ethyl, cyclopropyl or cyclobutyl;
p1 or p2 are each independently selected from 0, 1 or 2.
7. The compound of claim 6, or a stereoisomer, deutero-compound, solvate, prodrug, metabolite, pharmaceutically acceptable salt, or co-crystal thereof, wherein,
cy1, Cy2, Cy3 and Cy4 are each independently selected from one of the following substituted or unsubstituted groups: a bond,
Figure FDA0003178773230000131
Figure FDA0003178773230000132
When substituted, is optionally further substituted with 0 to 4 substituents selected from H, F, CF3Methyl, oxo, hydroxymethyl, COOH, CN or NH2Substituted with the substituent(s);
b is selected from
Figure FDA0003178773230000133
K is selected from
Figure FDA0003178773230000134
Figure FDA0003178773230000135
Figure FDA0003178773230000141
8. The compound of claim 7, or a stereoisomer, deutero-compound, solvate, prodrug, metabolite, pharmaceutically acceptable salt, or co-crystal thereof, wherein,
l is selected from a bond,
Figure FDA0003178773230000142
Figure FDA0003178773230000143
Figure FDA0003178773230000144
Wherein the left side is connected with B; or L is selected from
Figure FDA0003178773230000151
Figure FDA0003178773230000152
Figure FDA0003178773230000161
Figure FDA0003178773230000171
Figure FDA0003178773230000172
Wherein the left side is connected with B;
orL is selected from
Figure FDA0003178773230000173
Figure FDA0003178773230000174
Figure FDA0003178773230000181
Figure FDA0003178773230000191
Figure FDA0003178773230000201
Figure FDA0003178773230000211
Figure FDA0003178773230000221
Figure FDA0003178773230000231
Wherein the left side is connected with B;
or L is selected from
Figure FDA0003178773230000232
Figure FDA0003178773230000233
Figure FDA0003178773230000234
Wherein the left side is connected to B.
9. A compound according to claim 1 or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein
B is selected from
Figure FDA0003178773230000235
Figure FDA0003178773230000236
Or B is selected from
Figure FDA0003178773230000241
Figure FDA0003178773230000242
10. The compound of claim 4 or 7, or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein
K is selected from
Figure FDA0003178773230000243
Figure FDA0003178773230000244
Figure FDA0003178773230000251
11. A compound according to claim 10 or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein
K is selected from
Figure FDA0003178773230000252
Figure FDA0003178773230000253
Figure FDA0003178773230000261
12. A compound or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein the compound is selected from one of the following structures:
Figure FDA0003178773230000262
Figure FDA0003178773230000271
Figure FDA0003178773230000281
Figure FDA0003178773230000291
Figure FDA0003178773230000301
Figure FDA0003178773230000311
Figure FDA0003178773230000321
13. a compound according to any one of claims 1 to 12, or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, wherein the salt is selected from trifluoroacetate salt.
14. A pharmaceutical composition comprising a compound of any one of claims 1-13, or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, and a pharmaceutically acceptable carrier.
15. Use of a compound of any one of claims 1-13, or a stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, and the pharmaceutical composition of claim 14, for the manufacture of a medicament for the treatment of a disease associated with inhibition or degradation of BTK.
16. The use according to any one of claims 15, wherein the disease is selected from a tumor or an autoimmune disease.
CN202110840971.7A 2020-07-28 2021-07-26 BTK inhibitor ring derivative, preparation method and pharmaceutical application thereof Pending CN113999233A (en)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
CN2020107289535 2020-07-28
CN202010728953 2020-07-28
CN2020107966051 2020-08-13
CN202010796605 2020-08-13
CN202010918423 2020-09-04
CN2020109184237 2020-09-04
CN2020112265954 2020-11-10
CN202011226595 2020-11-10

Publications (1)

Publication Number Publication Date
CN113999233A true CN113999233A (en) 2022-02-01

Family

ID=79920987

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110840971.7A Pending CN113999233A (en) 2020-07-28 2021-07-26 BTK inhibitor ring derivative, preparation method and pharmaceutical application thereof

Country Status (1)

Country Link
CN (1) CN113999233A (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115557933A (en) * 2021-07-01 2023-01-03 杭州和正医药有限公司 Bruton tyrosine kinase and mutant degradation agent, composition and application thereof
WO2023072270A1 (en) * 2021-10-29 2023-05-04 百极弘烨(南通)医药科技有限公司 Protac compound, pharmaceutical composition comprising same, preparation method therefor, and use thereof
WO2023227080A1 (en) * 2022-05-25 2023-11-30 百极弘烨(南通)医药科技有限公司 Protac compound, pharmaceutical composition containing same, and preparation method therefor and use thereof

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115557933A (en) * 2021-07-01 2023-01-03 杭州和正医药有限公司 Bruton tyrosine kinase and mutant degradation agent, composition and application thereof
WO2023274390A1 (en) * 2021-07-01 2023-01-05 杭州和正医药有限公司 Bruton's tyrosine kinase and mutant degrader, composition and application thereof
WO2023072270A1 (en) * 2021-10-29 2023-05-04 百极弘烨(南通)医药科技有限公司 Protac compound, pharmaceutical composition comprising same, preparation method therefor, and use thereof
WO2023227080A1 (en) * 2022-05-25 2023-11-30 百极弘烨(南通)医药科技有限公司 Protac compound, pharmaceutical composition containing same, and preparation method therefor and use thereof

Similar Documents

Publication Publication Date Title
CA3089936C (en) Substituted quinazoline and pyridopyrimidine derivatives useful as anticancer agents
TWI762939B (en) BTK inhibitor cyclic derivatives, preparation method and pharmaceutical application thereof
TWI471326B (en) Substituted n-(1h-indazol-4-yl)imidazo[1,2-a]pyridine-3-carboxamide compounds as cfms inhibitors
RU2677884C2 (en) Substituted nicotinimide inhibitors of btk, their preparation and use in treatment of cancer, inflammation and autoimmune diseases
CN107629059B (en) Compounds useful as inhibitors of ATR kinase
EP2534151B1 (en) 8-methyl-1-phenyl-imidazol[1,5-a]pyrazine compounds
CN113999233A (en) BTK inhibitor ring derivative, preparation method and pharmaceutical application thereof
CN112812100B (en) Compound capable of degrading BTK kinase, preparation method and pharmaceutical application thereof
CN110799190A (en) Heterocyclic compounds
KR20100098714A (en) Benzofuropyrimidinones as protein kinase inhibitors
CN111050765B (en) Spiro compounds and methods of making and using the same
CN116134028A (en) Compound capable of degrading BTK kinase, preparation method and pharmaceutical application thereof
CN113292536A (en) Compound capable of degrading Bcr-Abl or PARP and preparation method and pharmaceutical application thereof
CN114437035A (en) Compound for inhibiting and degrading IRAK4, pharmaceutical composition and pharmaceutical application thereof
WO2006046135A2 (en) Pyrazolo[4,3-d] pyrimidine derivatives useful as pde-5 inhibitors
CN114507235A (en) Compound capable of degrading BTK kinase, preparation method and pharmaceutical application thereof
WO2022111526A1 (en) Benzene ring derivative, and composition and pharmaceutical use thereof
CN117425646A (en) Phosphono derivative, composition thereof and pharmaceutical application thereof
CN112279848A (en) Pan-JAKs inhibitor and application thereof
CN116888108B (en) Novel EGFR degradation agent
CN107849047A (en) A kind of biphenyl derivatives and preparation method thereof and purposes in medicine
CN114181196A (en) Compound for inhibiting and degrading PARP enzyme, preparation method and pharmaceutical application thereof
AU2019379213B2 (en) Six-membered fused with six-membered heterocyclic compound and uses thereof serving as protein receptor kinase inhibitor
CN112390785A (en) BET family bromodomain inhibitor ring derivative, preparation method and pharmaceutical application thereof
US11584737B2 (en) Heterocyclic compound

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
TA01 Transfer of patent application right

Effective date of registration: 20231218

Address after: 856099 Xingfu Jiayuan Economic Development Zone, Gyerba, Nedong District, Shannan City, Tibet Autonomous Region

Applicant after: Tibet Haisike Pharmaceutical Co.,Ltd.

Address before: 611130 No.136 Baili Road, Wenjiang cross strait science and Technology Park, Chengdu, Sichuan

Applicant before: SICHUAN HAISCO PHARMACEUTICAL Co.,Ltd.

TA01 Transfer of patent application right