CN113372345B - Deuterated heterocyclic kinase inhibitors - Google Patents

Deuterated heterocyclic kinase inhibitors Download PDF

Info

Publication number
CN113372345B
CN113372345B CN202110215181.XA CN202110215181A CN113372345B CN 113372345 B CN113372345 B CN 113372345B CN 202110215181 A CN202110215181 A CN 202110215181A CN 113372345 B CN113372345 B CN 113372345B
Authority
CN
China
Prior art keywords
deuterium
hydrogen
compound
pharmaceutically acceptable
independently selected
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.)
Active
Application number
CN202110215181.XA
Other languages
Chinese (zh)
Other versions
CN113372345A (en
Inventor
刘斌
陈博
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.)
Shandong Xuanzhu Pharma Co Ltd
Original Assignee
Xuanzhu Pharma 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 Xuanzhu Pharma Co Ltd filed Critical Xuanzhu Pharma Co Ltd
Publication of CN113372345A publication Critical patent/CN113372345A/en
Application granted granted Critical
Publication of CN113372345B publication Critical patent/CN113372345B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Abstract

The invention belongs to the technical field of medicines, and particularly relates to a deuterated heterocyclic DNA-PK inhibitor compound shown in a general formula (I), pharmaceutically acceptable salts and isomers thereof, a pharmaceutical composition and a preparation containing the compound, the pharmaceutically acceptable salts and the isomers thereof, a method for preparing the compound, the pharmaceutically acceptable salts and the isomers thereof, and applications of the compound, the pharmaceutically acceptable salts and the isomers thereof.

Description

Deuterated heterocyclic kinase inhibitors
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a heterocyclic DNA-PK inhibitor compound, pharmaceutically acceptable salts and isomers thereof, a pharmaceutical composition and a preparation containing the compound, the pharmaceutically acceptable salts and the isomers thereof, a method for preparing the compound, the pharmaceutically acceptable salts and the isomers thereof, and applications of the compound, the pharmaceutically acceptable salts and the isomers thereof.
Background
Cancer is a malignant disease which is difficult to treat all over the world, has high treatment difficulty and high mortality rate, brings heavy burden to patients and families, and is a main disease affecting the health of residents in China. In recent years, the incidence of cancer in our country has increased significantly, the mortality rate has also gradually increased, and cancer prevention and treatment face a severe situation.
Currently, radiotherapy and chemotherapy are the most effective means of treating cancer, while radiotherapy is the most effective non-surgical treatment of malignancies. Radiation and a considerable number of anticancer drugs can act directly or indirectly on DNA or DNA metabolic processes, resulting in DNA damage, of which DNA Double Strand Break (DSB) is the most lethal for cancer cells. After DNA damage, a series of cellular responses such as damaged DNA repair can be initiated, and the repair results in the improvement of cancer cell survival, which is one of the mechanisms of tumor cells resisting to radiotherapy and chemotherapy. If a DNA double strand break is not repaired in time and integrity, cancer cells die as a result of apoptosis or/and mitotic disturbances. Therefore, by inhibiting the repair of such DNA damage, the sensitivity of cancer cells to radiotherapy and chemotherapy can be improved, and the proliferation of cells can be inhibited.
In human and other higher eukaryotes, DSB repair is mainly performed by DNA-Dependent Protein Kinase (DNA-PK) dominated DNA non-homologous end joining (NHEJ), thereby repairing damaged DNA and maintaining cellular activity and genomic stability. NHEJ repair is primarily involved in G1/S phase DNA damage repair and does not require DNA end-joining templates. NHEJ repair requires an economic coordination of many proteins and signaling pathways. The heterodimer of the Ku70/80 subunit and the catalytic subunit DNA-dependent protein kinase (DNA-PKcs), together, constitute an active DNA-PK enzyme complex.
DNA-PKcs belongs to the member of the phosphatidylinositol 3 kinase (PI3K) superfamily, a serine/threonine protein kinase; the PI3K superfamily also includes ATM, ATR, mTOR and 4 PI3K isomers. When DNA-PK binds to a DNA break, its kinase activity is activated. The important function of Ku is to bind to the DNA termini, recruit DNA-PKcs, which make up DNA-PK holoenzymes and activate DNA-PKcs; activated DNA-PKcs directs the Artemis protein (an endonuclease) to bind to the damaged site, DNA end-breaking is performed by virtue of its ribozyme activity to facilitate ligation repair, then the XRCC 4/DNA-ligase IV complex is recruited by the activated DNA-PKcs, and finally the broken DNA double-stranded end is targeted and ligated by DNA-ligase IV to complete repair. XRCC4 is a protein that forms a complex with DNA-ligase IV and increases the activity of DNA-ligase IV. DNA-PKcs present 40 amino acid residues that can be autophosphorylated, with the most typical autophosphorylation sites occurring at Ser2056(POR cluster) and Thr2609(ABCDE cluster). NHEJ is thought to develop through three key steps: recognition of DSB-binding of Ku70/80 to incomplete DNA ends recruits two molecules of DNA-PKcs to the adjacent side of the DSB; performing DNA processing to remove the end-pointed non-ligatable ends or other forms of damage; finally, the DNA ends are ligated.
Tumor cells are more sensitive to DNA-PK because they have a higher basal level of endogenous replication stress (oncogene-induced replication stress) and DNA damage, and the DNA repair mechanisms are less efficient in tumor cells.
At present, the development of a high-efficiency and good-selectivity DNA-PK inhibitor has important clinical significance, can synergistically enhance the effects of radiotherapy and chemotherapy, effectively inhibit the growth of tumors, and simultaneously can effectively reduce the damage to normal cells and reduce side effects.
Disclosure of Invention
The invention aims to solve the technical problem of providing a heterocyclic compound which has a novel structure and a good inhibition effect on DNA-PK, in particular to a deuterated heterocyclic compound. Further, such compounds may be used to increase the sensitivity of a subject to radiation therapy and/or one or more anti-cancer agents. Furthermore, the compounds can be used for preventing and/or treating benign tumors or cancers by combining with radiotherapy and/or one or more anticancer agents.
The technical scheme of the invention is as follows:
in one aspect, the present invention provides a compound represented by the following general formula (I), a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0002952928540000021
wherein the content of the first and second substances,
X 1 、X 2 、X 3 、X 4 、X 5 、X 6 、X 7 each independently selected from C, C (R) or N;
x is selected from-CR a R b -、-C(O)-、-O-、-NR a -, -S-, -S (O) -or-S (O) 2 -;
Y is selected from O or S;
ring A is selected from 6-10 membered aryl or 5-10 membered heteroaryl optionally substituted with a substituent; the substituents are selected from deuterium, halogen, nitro, cyano, or the following groups optionally deuterated: hydroxy, amino, C 1-6 Alkyl radical, C 1-6 Alkylamino radical, di (C) 1-6 Alkyl) amino, halo C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Alkylthio, halo C 1-6 Alkoxy, halo C 1-6 Alkylthio, hydroxy C 1-6 Alkoxy, hydroxy C 1-6 Alkylthio, amino C 1-6 Alkoxy, amino C 1-6 Alkylthio radical, C 3-6 Cycloalkyl radical, C 3-6 A heterocyclic group;
each R, R 1 Each independently selected from hydrogen, deuterium, halogen, nitro, cyano, or the following optionally deuterated: hydroxy, amino, C 1-6 Alkyl radical, C 1-6 Alkylamino radical, di (C) 1-6 Alkyl) amino, halo C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Alkylthio, halo C 1-6 Alkoxy, halo C 1-6 Alkylthio, hydroxy C 1-6 Alkoxy, hydroxy C 1-6 Alkylthio, amino C 1-6 Alkoxy, amino C 1-6 Alkylthio radical, C 3-6 Cycloalkyl radical, C 3-6 A heterocyclic group;
R 2 is selected from-NR c R d 、-O-R c 、-S-R c Or the following group optionally deuterated: c 1-6 Alkyl, halo C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 An alkyl group;
R a 、R b 、R c 、R d each independently selected from hydrogen, deuterium, C 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 3-6 Heterocyclyl, wherein said C is 1-6 Alkyl radical, C 3-6 Cycloalkyl radical, C 3-6 The heterocyclic radical being optionally deuterated, C 1-6 Alkyl, deuterated C 1-6 Alkyl, halogen, hydroxyl, deuterated hydroxyl, amino and deuterated amino;
R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、R 9 each independently selected from hydrogen, deuterium, halogen, or the following optionally deuterated: hydroxy, amino, C 1-6 Alkyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Alkylthio, halo C 1-6 Alkoxy, haloGeneration C 1-6 An alkylthio group;
m is selected from 0, 1,2 or 3;
and X 1 、X 2 、X 3 、X 4 、X 5 、X 6 、X 7 X, ring A, R 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、R 9 At least one of which is deuterium or deuterated.
In certain embodiments, Y is S.
In certain embodiments, Y is O.
In certain embodiments, ring a is selected from phenyl, 5-8 membered monocyclic heteroaryl optionally substituted with substituents; the substituents are selected from deuterium, halogen, nitro, cyano, or the following groups optionally deuterated: amino group, C 1-6 Alkyl radical, C 1-6 Alkylamino radical, di (C) 1-6 Alkyl) amino, halo C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Alkylthio, halo C 1-6 Alkoxy or halo C 1-6 An alkylthio group.
In certain embodiments, ring a is selected from phenyl, 5-6 membered monocyclic heteroaryl optionally substituted with substituents; the substituents are selected from deuterium, halogen, nitro, cyano, or the following groups optionally deuterated: amino group, C 1-6 Alkyl radical, C 1-6 Alkylamino radical, di (C) 1-6 Alkyl) amino, halo C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Alkylthio, halo C 1-6 Alkoxy or halo C 1-6 An alkylthio group.
In certain embodiments, ring a is selected from phenyl or 5-6 membered monocyclic heteroaryl.
In certain embodiments, ring a is selected from phenyl or 5-6 membered nitrogen containing monocyclic heteroaryl optionally substituted with a substituent; the substituents are selected from deuterium, halogen, nitro, cyano, or the following groups optionally deuterated: amino group, C 1-6 Alkyl radical, C 1-6 Alkylamino radical, di (C) 1-6 Alkyl) amino, halo C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Alkylthio, halo C 1-6 Alkoxy or halo C 1-6 An alkylthio group.
In certain embodiments, ring a is selected from phenyl or 5-6 membered nitrogen containing monocyclic heteroaryl.
In certain embodiments, ring a is selected from phenyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, or triazinyl optionally substituted with a substituent; the substituents are selected from deuterium, halogen, nitro, cyano, trifluoromethyl, trifluoromethoxy, or the following group optionally deuterated: amino, methyl, ethyl, propyl, isopropyl, methylamino, dimethylamino, monofluoromethyl, difluoromethyl, hydroxymethyl, aminomethyl, methoxy, ethoxy, propoxy, isopropoxy, methylthio, monofluoromethoxy, difluoromethoxy.
In certain embodiments, ring a is selected from phenyl, pyrrolyl, imidazolyl, pyrazolyl, thiazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, or triazinyl.
In certain embodiments, ring a is selected from phenyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, or triazinyl, optionally substituted with a substituent; the substituents are selected from deuterium, halogen, nitro, cyano, trifluoromethyl, trifluoromethoxy, or the following group optionally deuterated: amino, methyl, ethyl, propyl, isopropyl, methylamino, dimethylamino, monofluoromethyl, difluoromethyl, hydroxymethyl, aminomethyl, methoxy, ethoxy, propoxy, isopropoxy, methylthio, monofluoromethoxy, difluoromethoxy.
In certain embodiments, ring a is selected from phenyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, or triazinyl.
In certain embodiments, each R, R 1 Each independently selected from hydrogen, deuterium, halogen, nitro, cyano, or optionally deuteratedThe following groups: amino group, C 1-6 Alkyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Alkylthio, halo C 1-6 Alkoxy, halo C 1-6 An alkylthio group.
In certain embodiments, each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, nitro, cyano, trifluoromethyl, trifluoromethoxy, trifluoromethylthio, or the following optionally deuterated: amino, methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, methoxy, ethoxy, methylthio, ethylthio, monofluoromethoxy, difluoromethoxy;
R 1 selected from hydrogen, deuterium, fluorine, chlorine, bromine, iodine, trifluoromethyl, trifluoromethoxy, or the following group optionally deuterated: amino, methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy.
In certain embodiments, each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, nitro, cyano, trifluoromethyl, trifluoromethoxy, or the following optionally deuterated: amino, methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy.
In certain embodiments, R 1 Selected from hydrogen, deuterium, fluorine, chlorine, bromine, iodine, trifluoromethyl, trifluoromethoxy, or the following group optionally deuterated: amino, methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy.
In certain embodiments, R 2 Is selected from-NR c R d 、-O-R c 、-S-R c Or the following group optionally deuterated: c 1-6 Alkyl, halo C 1-6 An alkyl group;
R c selected from hydrogen, deuterium, C 1-6 Alkyl radical, said C 1-6 Alkyl being optionally deuterium, C 1-6 Alkyl, deuterated C 1-6 Alkyl, halogen, hydroxy, deuterated hydroxy, amino, deuterated aminoSubstitution; r d Selected from hydrogen or deuterium.
In certain embodiments, R 2 Is selected from-NR c R d 、-O-R c 、-S-R c Or the following group optionally deuterated: c 1-6 Alkyl, halo C 1-6 An alkyl group; r c Selected from hydrogen, deuterium, trifluoromethyl, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl; r d Selected from hydrogen or deuterium.
In certain embodiments, R 2 Is selected from-NR c R d 、-O-R c 、-S-R c Trifluoromethyl, or the following group optionally deuterated: methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl;
R c selected from hydrogen, deuterium, trifluoromethyl, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl; r d Selected from hydrogen or deuterium.
In certain embodiments, X is selected from-CR a R b -、-C(O)-、-O-、-NR a -, -S-, -S (O) -or-S (O) 2 -;
R a 、R b Each independently selected from hydrogen or deuterium.
In certain embodiments, R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、R 9 Each independently selected from hydrogen, deuterium, halogen, trifluoromethyl, trifluoromethoxy, or the following group optionally deuterated: hydroxyl, amino, methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy.
In certain preferred embodiments, R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、R 9 Each independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or the following group optionally deuterated: methyl, ethyl, propyl, isopropyl, methoxy, ethoxy.
In certain embodiments, R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、R 9 Each independently selected from hydrogen or deuterium.
In certain embodiments, X 1 、X 2 、X 3 、X 4 、X 5 、X 6 、X 7 Each independently selected from C, C (R) or N;
y is selected from O or S;
x is selected from-CR a R b -、-O-、-NR a -or-S-;
ring a is selected from phenyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl or triazinyl optionally substituted with a substituent; the substituents are selected from deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or the following group optionally deuterated: amino, methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, methoxy, ethoxy, propoxy, isopropoxy, monofluoromethoxy, difluoromethoxy;
each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, trifluoromethylthio, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, methoxy, ethoxy, methylthio, ethylthio, monofluoromethoxy, difluoromethoxy;
R 1 selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or optionally substituted group of: amino, methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy;
R 2 is selected from-NR c R d 、-O-R c or-S-R c
R a 、R b Each independently selected from hydrogen or deuterium;
R c selected from hydrogen, deuterium, trifluoromethyl, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl; r d Selected from hydrogen or deuterium;
R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、R 9 each independently selected from hydrogen, deuterium, halogen, trifluoromethyl, trifluoromethoxy, or the following group optionally deuterated: methyl, ethyl, propyl, isopropyl, methoxy, ethoxy;
m is selected from 0, 1 or 2.
In certain embodiments, X 1 、X 2 、X 3 、X 4 、X 5 、X 6 、X 7 Each independently selected from C, C (R) or N;
y is selected from O or S;
x is selected from-CR a R b -、-O-、-NR a -or-S-;
ring a is selected from phenyl, pyridinyl or pyrimidinyl optionally substituted with a substituent; the substituents are selected from deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or the following group optionally deuterated: amino, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy;
each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, trifluoromethylthio, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy;
R 1 selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or optionally substituted group of: amino, methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy;
R 2 is selected from-NR c R d 、-O-R c or-S-R c
R a 、R b Each independently selected from hydrogen or deuterium;
R c selected from hydrogen, deuterium, trifluoromethyl, or the following optionally deuterated: methyl, ethyl, propyl, isopropylPropyl, monofluoromethyl, difluoromethyl; r d Selected from hydrogen or deuterium;
R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、R 9 each independently selected from hydrogen or deuterium;
m is selected from 0, 1 or 2.
In certain embodiments, X 3 Is N; x 1 、X 2 、X 4 、X 5 、X 6 、X 7 Each independently selected from C, C (R) or N;
each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, methoxy, ethoxy. In certain embodiments, X 2 Is N, X 5 Is N, X 1 、X 3 、X 4 、X 6 、X 7 Each independently selected from C, C (R) or N;
each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, methoxy, ethoxy.
In certain embodiments, X 3 Is N; x 1 、X 2 、X 4 、X 5 、X 6 、X 7 Each independently selected from C or C (R);
each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, methoxy, ethoxy.
In certain embodiments, X 2 Is N, X 5 Is N, X 1 、X 3 、X 4 、X 6 、X 7 Each independently selected from C or C (R);
each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, methoxy, ethoxy.
In certain embodiments, X 4 Is N; x 1 、X 2 、X 3 、X 5 、X 6 、X 7 Each independently selected from C, C (R) or N;
each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, methoxy, ethoxy.
In certain embodiments, X 3 Is N; x 4 Is N; x 1 、X 2 、X 5 、X 6 、X 7 Each independently selected from C, C (R) or N;
each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, methoxy, ethoxy.
In certain embodiments, X 2 Is N; x 4 Is N; x 1 、X 3 、X 5 、X 6 、X 7 Each independently selected from C, C (R) or N;
each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, methoxy, ethoxy.
The technical solutions of the present invention can be combined with each other to form a new technical solution, and the formed new technical solution is also included in the scope of the present invention.
In certain embodiments, the compound of formula (I), a pharmaceutically acceptable salt thereof, or an isomer thereof, is selected from the group consisting of:
Figure BDA0002952928540000081
Figure BDA0002952928540000091
Figure BDA0002952928540000101
in another aspect, the present invention also provides a pharmaceutical preparation, which contains the compound described in the aforementioned general formula (I), its pharmaceutically acceptable salt or its isomer, and one or more pharmaceutically acceptable excipients, and the pharmaceutical preparation can be any pharmaceutically acceptable dosage form. Pharmaceutically acceptable excipients are substances which are non-toxic, compatible with the active ingredient and otherwise biologically suitable for use in the organism. The choice of a particular excipient will depend on the mode of administration or disease type and state used to treat a particular patient.
In certain embodiments, the pharmaceutical formulations described above may be administered to a patient or subject in need of such treatment by oral, parenteral, rectal, or pulmonary administration, among others. For oral administration, the pharmaceutical composition can be prepared into oral preparations, for example, conventional oral solid preparations such as tablets, capsules, pills, granules and the like; it can also be made into oral liquid, such as oral solution, oral suspension, syrup, etc. When the composition is formulated into oral preparations, appropriate filler, binder, disintegrating agent, lubricant, etc. can be added. For parenteral administration, the above pharmaceutical preparations may also be prepared into injections, including injections, sterile powders for injection and concentrated solutions for injection. The injection can be prepared by conventional method in the existing pharmaceutical field, and can be prepared without adding additives or adding suitable additives according to the properties of the medicine. For rectal administration, the pharmaceutical composition may be formulated as a suppository or the like. For pulmonary administration, the pharmaceutical composition may be formulated as an inhalation formulation, aerosol, powder spray, or the like.
In another aspect, the invention also relates to the use of the compound of the general formula (I), its pharmaceutically acceptable salt or its isomer in the preparation of a medicament for preventing and/or treating diseases such as benign tumors or cancers, wherein the medicament is combined with radiotherapy and/or one or more anticancer agents, and the cancers comprise carcinoma in situ and metastatic cancers.
Furthermore, the invention also relates to application of a pharmaceutical preparation containing the compound shown in the general formula (I), the pharmaceutically acceptable salt thereof or the isomer thereof in preparing a medicament for preventing and/or treating diseases such as benign tumors or cancers, wherein the medicament can be combined with radiotherapy and/or one or more anticancer agents, and the cancers comprise carcinoma in situ and metastatic cancers.
In another aspect, the present invention also relates to the use of a compound of the aforementioned general formula (I), a pharmaceutically acceptable salt thereof, or an isomer thereof for the manufacture of a medicament for sensitizing cancer cells to anticancer agents and/or ionizing radiation.
Furthermore, the invention also relates to application of a pharmaceutical preparation containing the compound shown in the general formula (I), the pharmaceutically acceptable salt thereof or the isomer thereof in preparing a medicament for sensitizing cancer cells to anticancer agents and/or ionizing radiation.
The ionizing radiation refers to the radiation of various energy rays received by a patient during the radiotherapy process.
In another aspect, the present invention also provides a pharmaceutical composition comprising a compound of the foregoing general formula (I), a pharmaceutically acceptable salt or isomer thereof, and one or more second therapeutically active agents selected from the group consisting of anticancer agents including mitotic inhibitors, alkylating agents, antimetabolites, DNA chimerics, antitumor antibiotics, growth factor inhibitors, signaling inhibitors, cell cycle inhibitors, enzyme inhibitors, retinoid receptor modulators, proteasome inhibitors, topoisomerase inhibitors, biological response modifiers, hormonal agents, angiogenesis inhibitors, cell growth inhibitors, targeting antibodies, HMG-CoA reductase inhibitors, and prenyl protein transferase inhibitors.
In certain embodiments, the second therapeutically active agent can be a drug that reduces or reduces one or more side effects of a compound of the invention when used to treat a disease in a subject, or can enhance the efficacy of a compound of the invention.
In certain embodiments, the pharmaceutical composition further comprises one or more pharmaceutically acceptable excipients, as described above.
In another aspect, the present invention also relates to the use of a pharmaceutical composition containing the compound of the general formula (I), its pharmaceutically acceptable salt or its isomer for preparing a medicament for preventing and/or treating diseases such as benign tumor or cancer, which can be combined with radiotherapy and/or one or more anticancer agents, wherein the cancer includes carcinoma in situ and metastatic cancer.
Further, the invention also relates to application of a pharmaceutical composition containing the compound shown in the general formula (I), the pharmaceutically acceptable salt thereof or the isomer thereof in preparing a medicament for sensitizing cancer cells to anticancer agents and/or ionizing radiation.
In another aspect, the present invention also provides a method for treating a disease associated with DNAPK overactivation, the method comprising administering to a patient in need thereof an effective amount of a compound of the aforementioned general formula (I), a pharmaceutically acceptable salt thereof or an isomer thereof, the aforementioned pharmaceutical preparation or pharmaceutical composition; the disease associated with DNAPK over-activation is selected from benign tumors or cancers, including carcinoma in situ and metastatic carcinoma.
In another aspect, the present invention also provides a method for enhancing the sensitivity of a patient to an anticancer agent or radiation therapy, which comprises administering to a patient in need thereof an effective amount of a compound represented by the aforementioned general formula (I), a pharmaceutically acceptable salt thereof or an isomer thereof, the aforementioned pharmaceutical preparation or pharmaceutical composition; the anti-cancer agent is as described above.
In another aspect, the present invention also provides a kit comprising:
(a) an effective amount of one or more compounds of the general formula (I), pharmaceutically acceptable salts thereof or isomers thereof,
and (b) an effective amount of one or more anti-cancer agents.
The anti-cancer agent is as described above.
By "effective amount" is meant a dosage of a drug that prevents, alleviates, retards, inhibits or cures a condition in a subject. The size of the administered dose is related to the administration mode of the drug, the pharmacokinetics of the medicament, the severity of the disease, the individual signs (sex, weight, height, age) of the subject, and the like.
In the present invention, unless otherwise defined, scientific and technical terms used herein have meanings commonly understood by those skilled in the art, however, in order to better understand the present invention, definitions of some terms are provided below. To the extent that the definitions and explanations of terms provided herein do not conform to the meanings commonly understood by those skilled in the art, the definitions and explanations of terms provided herein shall control.
The "halogen" as referred to herein means a fluorine atom, a chlorine atom, a bromine atom or an iodine atom.
"C" according to the invention 1-6 Alkyl "denotes a straight or branched chain alkyl group containing 1 to 6 carbon atoms, including for example" C 1-4 Alkyl group "," C 1-3 Alkyl group "," C 1-2 Alkyl group "," C 2-6 Alkyl group "," C 2-5 Alkyl group "," C 2-4 Alkyl group and C 2-3 Alkyl group "," C 3-6 Alkyl group "," C 3-5 Alkyl group "," C 3-4 Alkyl "and the like, specific examples include, but are not limited to: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, 2-methylbutyl, neopentyl, 1-ethylpropyl, n-hexyl, isohexyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 3-dimethylbutyl, 2-dimethylbutyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 1, 2-dimethylpropyl, and the like. "C" according to the invention 1-4 Alkyl "means C 1-6 Specific examples of the alkyl group having 1 to 4 carbon atoms.
"C" according to the invention 1-6 Alkoxy "means" C 1-6 alkyl-O- ", said" C 1-6 Alkyl "is as defined above. "C" according to the invention 1-4 Alkoxy "means" C 1-4 alkyl-O- ", said" C 1-4 Alkyl radicals "such asAs defined above.
"C" according to the invention 1-6 Alkylthio "means" C 1-6 alkyl-S- ", said" C 1-6 Alkyl "is as defined above. "C" according to the invention 1-4 Alkylthio "means" C 1-4 alkyl-S- ", said" C 1-4 Alkyl "is as defined above.
The "hydroxy group C" of the present invention 1-6 Alkyl, amino C 1-6 Alkyl, halo C 1-6 Alkyl "means C 1-6 One or more hydrogens of the alkyl group are each replaced by one or more hydroxyl groups, amino groups or halogens. C 1-6 Alkyl is as previously defined
The "hydroxy group C" of the present invention 1-6 Alkoxy, amino C 1-6 Alkoxy, halo C 1-6 Alkoxy "means" C 1-6 One or more hydrogens of "alkoxy" are replaced with one or more hydroxy, amino, or halogen.
The "hydroxy group C" of the present invention 1-6 Alkylthio, amino C 1-6 Alkylthio, halo C 1-6 Alkylthio "means" C 1-6 Alkylthio "is one in which one or more hydrogens are replaced with one or more hydroxy, amino, or halogen.
"C" according to the invention 1-6 Alkylamino radical, di (C) 1-6 Alkyl) amino "means independently C 1-6 alkyl-NH-),
Figure BDA0002952928540000121
The "6-to 10-membered aryl" as referred to in the present invention includes "6-to 8-membered monocyclic aryl" and "8-to 10-membered fused ring aryl".
The "6-to 8-membered monocyclic aryl group" as referred to herein means a monocyclic aryl group having 6 to 8 ring carbon atoms, and examples thereof include phenyl and the like.
The "8-to 10-membered fused ring aryl" as referred to herein means an unsaturated aromatic cyclic group having 8 to 10 ring carbon atoms, formed by two or more cyclic structures sharing two adjacent atoms with each other, and is preferably a "9-to 10-membered fused ring aryl", and specific examples thereof are naphthyl and the like.
The "5-to 10-membered heteroaryl" as used herein includes "5-to 8-membered monocyclic heteroaryl" and "8-to 10-membered fused heteroaryl".
The "5-to 8-membered monocyclic heteroaryl group" according to the present invention means a monocyclic cyclic group having aromaticity, which contains 5 to 8 ring atoms, at least one of which is a heteroatom such as nitrogen atom, oxygen atom or sulfur atom. Optionally, a ring atom (e.g., a carbon atom, a nitrogen atom, or a sulfur atom) in the cyclic structure may be oxo. "5-to 8-membered monocyclic heteroaryl" includes, for example, "5-to 7-membered monocyclic heteroaryl", "5-to 6-membered nitrogen-containing monocyclic heteroaryl", "6-membered nitrogen-containing monocyclic heteroaryl", and the like, in which the heteroatom contains at least one nitrogen atom, for example, contains 1 or 2 nitrogen atoms, or, contains one nitrogen atom and 1 or 2 other heteroatoms (e.g., oxygen atom and/or sulfur atom), or, contains 2 nitrogen atoms and 1 or 2 other heteroatoms (e.g., oxygen atom and/or sulfur atom).
Specific examples of the "5-to 8-membered monocyclic heteroaryl group" include, but are not limited to, furyl, thienyl, pyrrolyl, thiazolyl, isothiazolyl, thiadiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, imidazolyl, pyrazolyl, 1,2, 3-triazolyl, 1,2, 4-triazolyl, 1,2, 3-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,3, 4-oxadiazolyl, pyridyl, 2-pyridonyl, 4-pyridonyl, pyrimidinyl, pyridazinyl, pyrazinyl, 1,2, 3-triazinyl, 1,3, 5-triazinyl, 1,2,4, 5-tetrazinyl, and the like. The "5-6 membered monocyclic heteroaryl" refers to a specific example containing 5 to 6 ring atoms in the 5-8 membered monocyclic heteroaryl.
The "8-to 10-membered fused heteroaryl group" as used herein refers to an unsaturated aromatic cyclic structure having 8 to 10 ring atoms (at least one of which is a heteroatom such as nitrogen atom, oxygen atom or sulfur atom) formed by two or more cyclic structures sharing two adjacent atoms with each other. Optionally, a ring atom (e.g., a carbon atom, a nitrogen atom, or a sulfur atom) in the cyclic structure may be oxo. Including "9-to 10-membered fused heteroaryl", "8-to 9-membered fused heteroaryl", and the like, which may be fused in a benzo-5-6-membered heteroaryl, 5-6-membered heteroaryl and 5-6-membered heteroaryl, and the like; specific examples include, but are not limited to: pyrrolopyrrole, pyrrolofuran, pyrazolopyrrole, pyrazolothiophene, furothiophene, pyrazoloxazole, benzofuranyl, benzisofuranyl, benzothiophenyl, indolyl, isoindolyl, benzoxazolyl, benzimidazolyl, indazolyl, benzotriazolyl, quinolinyl, 2-quinolinonyl, 4-quinolinonyl, 1-isoquinolinyl, acridinyl, phenanthridinyl, pyridazinyl, phthalazinyl, quinazolinyl, quinoxalinyl, purinyl, naphthyridinyl, and the like.
"C" according to the invention 3-6 Cycloalkyl "refers to a saturated or partially saturated monocyclic ring group containing 3 to 6 ring atoms and having no aromaticity, as defined in the present invention by" C 3-6 Cycloalkyl "includes" C 3-6 Saturated cycloalkyl radicals "and" C 3-6 Partially saturated cycloalkyl radical ", preferably" C 3-4 Cycloalkyl group "," C 5-6 Cycloalkyl group "," C 3-5 Cycloalkyl groups "and the like. Examples thereof include, but are not limited to: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cyclohexene, etc.
"C" according to the invention 3-6 The heterocyclic group "means a saturated or partially saturated and non-aromatic monocyclic cyclic group containing at least one hetero atom (e.g., containing 1,2,3, 4 or 5) which is a nitrogen atom, an oxygen atom and/or a sulfur atom and has 3 to 6 ring atoms, and optionally, a ring atom (e.g., a carbon atom, a nitrogen atom or a sulfur atom) in the cyclic structure may be oxo. "C" according to the invention 3-6 Heterocyclyl "includes" C 3-6 Saturated heterocyclic radicals "and" C 3-6 Partially saturated heterocyclyl ". Said "C 3-6 Heterocyclyl "preferably" C 3-4 Heterocyclic group "," C 3-5 Heterocyclic group "," C 5-6 Heterocyclyl "and the like. Specific examples thereof include, but are not limited to: aziridinyl, 2H-aziridinyl, diazacyclopropyl, 3H-diazacyclopropenyl, azetidinyl, 1, 4-dioxanyl, 1, 3-diazacycloAn oxacyclohexyl group, a 1, 3-dioxolanyl group, a 1, 4-dioxadienyl group, a tetrahydrofuryl group, a dihydropyrrolyl group, a pyrrolidinyl group, an imidazolidinyl group, a 4, 5-dihydroimidazolyl group, a pyrazolidinyl group, a 4, 5-dihydropyrazolyl group, a 2, 5-dihydrothienyl group, a tetrahydrothienyl group, a 4, 5-dihydrothiazolyl group, a thiazolidinyl group, a piperidyl group, a tetrahydropyridinyl group, a piperidonyl group, a tetrahydropyridinyl group, a dihydropiperidonyl group, a piperazinyl group, a morpholinyl group and the like.
The term "optionally substituted" as used herein refers to both the case where one or more hydrogen atoms on a substituted group are "substituted" or "unsubstituted" with one or more substituents, and when a group is "substituted" with a substituent, it is preferably substituted with 1 to 3 substituents, more preferably 1 to 2 substituents.
The term "deuterated" as used herein means that one or more hydrogen atoms of a deuterated group are "substituted" with one or more deuterium atoms, either partially or fully deuterated.
As used herein, "optionally deuterated" refers to both deuterated and non-deuterated groups, wherein "deuterated" is as defined above.
The "anticancer agent" of the present invention refers to an agent having a certain therapeutic effect on tumors, including, but not limited to, mitotic inhibitors, alkylating agents, antimetabolites, DNA chimerics, antitumor antibiotics, growth factor inhibitors, signal transduction inhibitors, cell cycle inhibitors, enzyme inhibitors, retinoid receptor modulators, proteasome inhibitors, topoisomerase inhibitors, biological response modifiers, hormonal drugs, angiogenesis inhibitors, cell growth inhibitors, targeting antibodies, HMG-CoA reductase inhibitors, prenyl protein transferase inhibitors, and the like; the tumor includes benign tumor and cancer.
The chemotherapy is the abbreviation of chemical drug therapy, and achieves the purpose of treatment mainly by using chemical therapeutic drugs to kill cancer cells.
The "radiotherapy" in the invention refers to a tumor treatment method, i.e. tumor radiotherapy, which mainly uses radioactive rays to perform local tumor treatment, wherein the "radioactive rays" include alpha, beta and gamma rays generated by radioactive isotopes, and x rays, electron beams, proton beams and other particle beams generated by various x-ray treatment machines or accelerators.
"pharmaceutically acceptable salt" as used herein refers to an acidic functional group (e.g., -COOH, -OH, -SO) present in a compound 3 H, etc.) with a suitable inorganic or organic cation (base), including salts with alkali or alkaline earth metals, ammonium salts, salts with nitrogen-containing organic bases; and salts of basic functional groups present in the compounds (e.g., -NH2, etc.) with suitable inorganic or organic anions (acids), including salts with inorganic or organic acids (e.g., carboxylic acids, etc.).
"isomers" as used herein means that the compounds of the present invention contain one or more asymmetric centers and thus are available as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. The compounds of the present invention may have asymmetric centers that each independently produce two optical isomers. The scope of the present invention includes all possible optical isomers and mixtures thereof. The compounds of the present invention, if they contain an olefinic double bond, include cis-isomers and trans-isomers, unless otherwise specified. The compounds described herein may exist in tautomeric (one of the functional group isomers) forms having different points of attachment of hydrogen through one or more double bond shifts, e.g., a ketone and its enol form are keto-enol tautomers. The compounds of the present invention contain a spiro ring structure, and substituents on the ring may be present on both sides of the ring to form the opposite cis (cis) and trans (trans) isomers, depending on the steric structure of the ring. Each tautomer and mixtures thereof are included within the scope of the present invention. All enantiomers, diastereomers, racemates, meso, cis-trans isomers, tautomers, geometric isomers, epimers, mixtures thereof and the like of the compounds are included within the scope of the present invention.
The compounds of the invention may be prepared by enantiospecific synthesis or by resolution from a mixture of enantiomers in such a way as to give the individual enantiomers. Conventional resolution techniques include resolving mixtures of enantiomers of the starting material or the final product using various well-known chromatographic methods.
When the stereochemistry of the disclosed compounds is named or depicted by structure, the named or depicted stereoisomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% pure by weight relative to the other stereoisomers. When a single isomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% pure by weight. The optical purity wt% is the ratio of the weight of an enantiomer to the weight of the enantiomer plus the weight of its optical isomer.
In another aspect, the present invention also provides a process for the preparation of a compound of formula (I) according to the invention:
Figure BDA0002952928540000161
wherein E is boronic acid or boronic ester; hal 'and Hal' are respectively and independently selected from F, Cl, Br and I; corresponding to X 1 -X 7 、R 1 -R 9 X, Y, ring A, m are as previously defined.
The method comprises the following steps:
(a) the intermediate 1 and the intermediate 2 are subjected to a Suzuki coupling reaction under the action of a catalyst to generate an intermediate 3;
(b) and reacting the intermediate 3 with the intermediate 4 under alkaline conditions to generate the compound of the general formula (I).
In another aspect, the present invention also provides a method for preparing intermediate 1 in the above preparation method:
Figure BDA0002952928540000162
wherein E is boronic acid or boronic ester; hal is selected from F, Cl, Br and I; corresponding to X 1 -X 7 As defined aboveThe above-mentioned; d' is a hydrogen or deuterium atom. The method comprises the following steps:
(a) reacting the intermediate 6 with tert-butylamine under acidic conditions to generate an intermediate 7;
(b) the intermediate 7 reacts under an acidic condition to generate an intermediate 8;
(c) reacting the intermediate 8 with chloroacetaldehyde to generate an intermediate 9;
(d) the intermediate 9 reacts with B under the action of catalyst 2 (PIN) 2 The reaction produced intermediate 1.
In the above preparation method, all reactions can be carried out in a conventional solvent, including but not limited to DMSO, DMF, acetonitrile, methanol, ethanol, tetrahydrofuran, toluene, dimethyl ether, dichloromethane, chloroform, dichloroethane, 1, 4-dioxane, trifluoroacetic acid, and water, and a single solvent or a mixed solvent of two or more solvents can be used in the reaction process. Alternatively, if a certain reactant is a liquid, the reaction may be carried out in the absence of another solvent.
The alkaline condition is a condition containing organic base or inorganic base, and the organic base is independently selected from pyridine, triethylamine, N-dimethylaniline, sodium methoxide, potassium ethoxide, potassium tert-butoxide, sodium tert-butoxide, potassium acetate, N-diisopropylethylamine and the like; preferably an inorganic base independently selected from potassium carbonate, sodium carbonate, cesium carbonate, sodium hydride, potassium hydroxide, sodium hydroxide, lithium hydroxide, potassium acetate, sodium acetate, potassium phosphate, sodium phosphate and the like.
The acidic condition refers to a condition containing organic acid or inorganic acid, and the organic acid is independently selected from formic acid, acetic acid, trifluoroacetic acid, propionic acid, benzoic acid, methanesulfonic acid, p-toluenesulfonic anhydride, tartaric acid and the like; the inorganic acid is independently selected from hydrochloric acid, concentrated sulfuric acid, hydrobromic acid, hydrofluoric acid, nitric acid, nitrous acid, boric acid, and the like.
The catalyst is independently selected from Pd (PPh) 3 ) 4 、PdCl 2 (PPh 3 ) 2 、PdCl 2 (MeCN) 2 、Pd(dppf)Cl 2 、Ph 2 P(CH 2 ) 2 PPh 2 (dppe)、Ph 2 P(CH 2 ) 3 PPh 2 (dppp)、Pd 2 (dba) 3 One or more of palladium chloride, palladium acetate, palladium triphenylphosphine and tricyclohexylphosphine.
In the present invention, the compounds and intermediates of the present invention can be isolated and purified using methods well known to those skilled in the art of organic synthesis. Examples of conventional methods for isolating and purifying compounds may include, but are not limited to: chromatography on a solid support (e.g. silica gel, alumina or silica derivatized with alkylsilanes), thin-layer chromatography, distillation at various pressures, vacuum sublimation, trituration, for example, by the methods described below: "Vogel's Textbook of Practical Organic Chemistry",5th edition (1989), Furniss et al, pub. Longman Scientific & Technical, Essex CM 202 JE, England.
It is understood that the chemical reaction, if involving reactive groups such as-NH-which need not participate in the reaction 2 The reactive groups-OH, -COOH, etc. can be protected prior to the next reaction by methods known to those skilled in the art, including but not limited to, the formation of esters, amides, alkylamines, ethers, etc. of the reactive groups. Common methods of carboxyl protection include, but are not limited to, ester formation with aliphatic or aromatic alcohols, amide or hydrazide formation with amines or hydrazines. Common amino protecting groups include, but are not limited to: (1) an alkoxycarbonyl amino-protecting group such as benzyloxycarbonyl (Cbz), tert-butoxycarbonyl (Boc), fluorenyl methoxycarbonyl (Fmoc), allyloxycarbonyl (Alloc), trimethylsilethoxycarbonyl (Teoc), etc.; (2) acyl amino groups such as phthaloyl (Pht), p-toluenesulfonyl (Tos), trifluoroacetyl (Tfa), o- (p) nitrobenzenesulfonyl (Ns), pivaloyl and the like; (3) alkyl amino protecting groups, trityl (Trt), 2, 4-dimethoxybenzyl (Dmb), p-methoxybenzyl (PMB), benzyl (Bn), and the like. Common hydroxyl protecting groups include, but are not limited to, silyl ether protecting groups, benzyl ether protecting groups, alkoxymethyl ethers or alkoxy-substituted methyl ethers, acetyl, benzoyl, pivaloyl and the like. After the reaction of the protecting group is completed, the reaction of removing protecting group can be performed by methods known to those skilled in the art, and the conditions for removing protecting group include, but are not limited to, deprotection under acidic conditionsDeprotection under alkaline condition, catalytic hydrogenation deprotection, etc.; the acidic and basic conditions are as defined above.
The raw materials and/or intermediates directly used in the preparation method of the present invention can be commercially or self-prepared, and the intermediate can be obtained by a person skilled in the art according to a known conventional chemical reaction preparation method, and the preparation method thereof is also within the protection scope of the present invention.
Advantageous effects of the invention
1. The compound, the pharmaceutically acceptable salt or the isomer thereof has excellent DNA-PK inhibitory action, good pharmacokinetic properties in organisms, lasting action and high bioavailability, and can enhance the sensitivity of cancer cells to radiotherapy and/or one or more anticancer agents.
2. The compound, the pharmaceutically acceptable salt thereof or the isomer thereof has better therapeutic effect on benign tumors and cancers.
3. The compound of the invention has simple preparation process, high medicine purity, stable quality and easy large-scale industrial production.
Drawings
The accompanying drawings, which are included to provide a further understanding of the disclosure and are incorporated in and constitute a part of this application, illustrate embodiment(s) of the disclosure and together with the description serve to explain the disclosure and not to limit the disclosure. In the drawings:
fig. 1 is the tumor weight results of the combination of the compound of the present invention and doxorubicin hydrochloride liposome injection after drug effect experiment administration on nude mice loaded with NCI-H1048 small cell lung cancer transplantation tumor model, wherein the abscissa is each experimental group and the ordinate is tumor weight (g), the sum is a statistical result, the sum represents the comparison between the combination group and the vehicle control group, the sum represents the comparison between the combination group and the doxorubicin single group, and the sum represents the P value < 0.05; and denotes a P value < 0.05; denotes P value < 0.001.
FIG. 2 shows the results of T/C (%) after administration of the compound of the present invention in combination with doxorubicin hydrochloride liposome injection to nude mice loaded with NCI-H1048 small cell lung cancer transplanted tumor model, wherein the abscissa is the time (days) after tumor inoculation and the ordinate is the relative tumor proliferation rate T/C (%).
Detailed description of the preferred embodiments
The technical solutions of the present invention will be described below in conjunction with the specific embodiments, and the above-mentioned contents of the present invention will be further described in detail, but it should not be understood that the scope of the above-mentioned subject matter of the present invention is limited to the following examples. All the technologies realized based on the above contents of the present invention belong to the scope of the present invention.
Abbreviations:
DCM is dichloromethane; MeOH: methanol; b is 2 (PIN) 2 Pinacol ester diboron; pd (dppf) 2 Cl 2 1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride; pd 2 (dba) 3 : tris (dibenzylideneacetone) dipalladium; EA: ethyl acetate; PE: petroleum ether; pd (PPh) 3 ) 2 Cl 2 : bis (triphenylphosphine) palladium dichloride; selectfluor: 1-chloromethyl-4-fluoro-1, 4-diazobicyclo 2.2.2 octane bis (tetrafluoroborate) salt; tf (f) 2 O: trifluoromethanesulfonic anhydride; HATU: 2- (7-azabenzotriazole) -N, N' -tetramethyluronium hexafluorophosphate; DIEA: n, N-diisopropylethylamine; SFC: supercritical fluid chromatography.
Preparation example one: preparation of (R) -8- (1-aminopropyl-2-yl) -N-methylquinoline-4-carboxamide hydrochloride
1. Preparation of tert-butyl (2- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) allyl) carbamate
Figure BDA0002952928540000191
To N-tert-butoxycarbonylaminopropyne (150.0g,967.7mmol), B at 0 deg.C 2 (Pin) 2 (300.0g, 1181.1mmol), CuCl (10.0g,100.8mmol), t-BuONa (15.0g,156.1mmol), and P (t-Bu) 3 MeOH (75.0mL,1875.0mmol) was slowly added dropwise to a suspension of (25.0g,123.6mmol) in 3.0L of toluene, and after the addition was complete, the system was warmed to 20 ℃ and stirred for 16 h. Silica gel column chromatography (petroleum ether: ethyl acetate 5:1) was performed to give the crude compound (330.0g) as a crude productAnd then used in the next step.
2. Preparation of tert-butyl (2- (4-hydroxyquinolin-8-yl) allyl) carbamate
Figure BDA0002952928540000192
A mixture containing 4-hydroxy-8-bromoquinoline (150.0g,669.6mmol), tert-butyl (2- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) allyl) carbamate (288.0g,1014.1mmol), Pd (dppf) Cl 2 (15.0g,20.5mmol), and Na 2 CO 3 A suspension of (144.0g,1358.5mmol) 1, 4-dioxane (1.9L) and water (0.2L) was stirred at 100 ℃ for 16h and tert-butyl (2- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) allyl) carbamate (90.0g) and Pd (dppf) Cl were added 2 (12.0g), stirring was continued at 100 ℃ for 16 hours. Direct silica gel column chromatography (dichloromethane: methanol ═ 20:1) afforded the crude compound (120.0g) which was used directly in the next step.
3. Preparation of tert-butyl (2- (4-hydroxyquinolin-8-yl) propyl) carbamate
Figure BDA0002952928540000201
A suspension (1.5L) of tert-butyl (2- (4-hydroxyquinolin-8-yl) allyl) carbamate in methanol containing (100.0g,333.3mmol) and 10% Pd/C (50.0g) was stirred under hydrogen at 40 ℃ for 4 hours, filtered through Celite, and the solvent was spin-dried to obtain the product (86.0g, yield: 85.4%).
4. Preparation of 8- (1- (((tert-butoxycarbonyl) amino) prop-2-yl) quinolin-4-yl trifluoromethanesulfonate
Figure BDA0002952928540000202
To a solution of tert-butyl (2- (4-hydroxyquinolin-8-yl) propyl) carbamate (86.0g,284.8mmol) and pyridine (111.8g,1415.2mmol) in 1.0L of dichloromethane at-20 deg.C was slowly added Tf dropwise 2 O(120.4g,427.0mmol) and stirring at-20 ℃ for 20min after the addition. The reaction was quenched by addition of saturated aqueous citric acid (500mL), and the organic phase was separated, dried over anhydrous sodium sulfate, and spin-dried to give the product (120.0g, yield: 97.0%).
5. Preparation of tert-butyl (2- (4-cyanoquinolin-8-yl) propyl) carbamate
Figure BDA0002952928540000203
8- (1- (((tert-butoxycarbonyl) amino) prop-2-yl) quinolin-4-yl trifluoromethanesulfonate (120.0g,276.5mmol), Zn (CN) 2 (40.8g,347.5mmol) and Pd (PPh) 3 ) 4 A suspension (1.4L) of (16.8g,14.5mmol) in methanol was stirred at 100 ℃ for 12 hours under nitrogen. The solvent was dried, water (500.0mL) was added, extraction was performed with ethyl acetate (500.0mL × 3), and the organic phases were combined and dried over anhydrous sodium sulfate. Column chromatography over silica gel (petroleum ether: ethyl acetate: 10:1) gave the product (76.0g, yield: 88.4%).
6. Preparation of 8- (1- ((tert-butoxycarbonyl) amino) propan-2-yl) quinoline-4-carboxylic acid
Figure BDA0002952928540000211
A mixture of tert-butyl (2- (4-cyanoquinolin-8-yl) propyl) carbamate (76.0g,243.6mmol) and KOH (136.8g,2442.9mmol) in ethanol (800.0mL) and water (280.0mL) was stirred at 120 ℃ for 12 hours. The pH was adjusted to 5 to 6 with dilute hydrochloric acid to precipitate a solid, which was then filtered to obtain the product (41.0g, yield: 50.9%).
7. Preparation of (R) - (2- (2- (4- (methylcarbamoyl) quinolin-8-yl) propyl) carbamic acid tert-butyl ester
Figure BDA0002952928540000212
8- (1- ((tert-butoxycarbonyl) amino) propan-2-yl) quinoline-4-carboxylic acid (37.7g,114.0mmol), HATU (86.9g,228.0mmol), MeNH 2. A suspension (500.0mL) of HCl (15.4g,228.0mmol) and DIEA (29.4g,228.0mmol) in dichloromethane was stirred at 10 ℃ for 16 h. Water (500.0mL) was added, extracted with dichloromethane (500.0mL x 3), and the organic phases were combined and dried over anhydrous sodium sulfate. Silica gel column chromatography (dichloromethane: methanol 60: 1-20: 1) to obtain crude product, pulping the crude product with ethyl acetate to obtain racemate (23.9g), and SFC separation (column model: CHIRALPAX AY-H (AYH0 CE-vc0010.46cm i.d. 25cm L), mobile phase: n-hexane/ethanol 90:10(V: V), peak time: 16.928min) to obtain target compound (12g, yield: 30.6%).
8. Preparation of (R) -8- (1-aminopropyl-2-yl) -N-methylquinoline-4-carboxamide hydrochloride
Figure BDA0002952928540000213
Tert-butyl (2- (4- (methylcarbamoyl) quinolin-8-yl) propyl) carbamate (3.0g, 8.7mmol) was dissolved in a solution of hydrogen chloride in 1, 4-dioxane (30.0mL) and stirred at 20 ℃ for 1.5 hours. Spin dry to give the crude product (2.6g) which was used directly in the next step.
The first embodiment is as follows: preparation of (R) -8- (1- ((6- (imidazo [1,2-a ] pyridin-7-yl-5, 6,8-d3) pyrimidin-4-yl) amino) propan-2-yl) -N-methylquinoline-4-carboxamide (Compound 1-1)
1. 1-pyridine-d-oxides 5 Preparation of
Figure BDA0002952928540000221
Pyridine-d 5 (12.0g,142.7mmol) was added to DCM (120mL), m-chloroperoxybenzoic acid (85%, 34.8g,171.4mmol) was added in portions at 0 ℃ and reacted at 20 ℃ for 16 hours. The system was filtered under reduced pressure, the filter cake was washed with DCM (10ml × 2), and the filtrate was concentrated and purified by silica gel column chromatography (DCM: MeOH ═ 10:1) to give the product (10.5g, 73.5% yield).
2. 4-nitropyridine-1-oxide-2, 3,5,6-d 4 Preparation of
Figure BDA0002952928540000222
1-pyridine-d oxide 5 (9.5g,94.9mmol) was added to a reaction flask containing concentrated sulfuric acid (45mL) at 0 deg.C, followed by addition of fuming nitric acid (30mL) at 0 deg.C, stirring for 10 minutes, and then reaction at 130 deg.C for 4 hours. Pouring ice water (300mL) into the system, slowly adding sodium carbonate solid (80g), carrying out vacuum filtration on the system, taking a filter cake as a target product (5g), extracting the filtrate by using DCM (200mL x 3), drying the organic phase by using anhydrous sodium sulfate, carrying out spin drying to obtain a crude product 10.0g, adding DCM (50mL) for ultrasonic treatment, carrying out vacuum filtration on insoluble substances, drying the filter cake to obtain a target compound (3g), and combining the two filter cakes to obtain the target compound (8g, yield 58.5%).
3. 4-chloropyridine-1-oxide-2, 3,5,6-d 4 Preparation of
Figure BDA0002952928540000223
4-nitropyridine-1-oxide-2, 3,5,6-d 4 (7.0g,48.6mmol) was added to acetyl chloride (50mL) and reacted at 50 ℃ for 1 hour. Silica gel column chromatography (DCM: MeOH ═ 15:1) was performed on the system to give the product (6.0g, 92.8% yield).
4. N- (tert-butyl) -4-chloropyridine-3, 5,6-d 3 Preparation of (E) -2-amines
Figure BDA0002952928540000224
4-chloropyridine-1-oxide-2, 3,5,6-d 4 (5.5g,41.3mmol) was added to a mixed solution of DCM (90mL) and chloroform (30mL), tert-butylamine (15.1g,206.6mmol) was added thereto, p-toluenesulfonic anhydride (27.0g,82.7mmol) was added in portions at 0 ℃ and the mixture was reacted at 0 ℃ for 1 hour. The reaction system was suction filtered under reduced pressure, and the filtrate was concentrated and subjected to silica gel column chromatography (PE: EA ═ 20:1) to give the product (3.4g, yield 44.0%).
5. 4-chloropyridine-3, 5,6-d 3 Preparation of (E) -2-amines
Figure BDA0002952928540000231
Mixing N- (tert-butyl) -4-chloropyridine-3, 5,6-d 3 -2-amine (3.2g,17.1mmol) was added to trifluoroacetic acid (15mL) and reacted at 80 ℃ for 16 hours. The system was concentrated, EA (80mL) and saturated sodium carbonate solution (30mL) were added, the solution was separated by extraction, and the organic phase was dried by silica gel column chromatography (PE: EA ═ 1:1) to give the product (2.0g, 89.2% yield).
6. 7-chloroimidazo [1,2-a ]]Pyridine-5, 6,8-d 3 Preparation of
Figure BDA0002952928540000232
4-chloropyridine-3, 5,6-d 3 -2-amine (1.9g,14.5mmol) was added to water (20mL) followed by chloroacetaldehyde (1.7g,21.7mmol) and reacted at 100 ℃ for 4 hours. The pH of the system was adjusted to alkalinity with sodium carbonate solution (10mL), EA (40mL × 2) was added for extraction and separation, the organic phase was concentrated, and the product was obtained by silica gel column chromatography (PE: EA ═ 1:1) (1.8g, yield 80.1%).
7. (imidazo [1, 2-a)]Pyridin-7-yl-5, 6,8-d 3 ) Preparation of boric acid
Figure BDA0002952928540000233
Reacting 7-chloroimidazo [1,2-a ]]Pyridine-5, 6,8-d 3 (620mg,4.0mmol) was dissolved in 1, 4-dioxane (12mL) and B was added 2 (PIN) 2 (1.5g,5.9mmol),Pd 2 (dba) 3 (366mg,0.4mmol), tricyclohexylphosphine (224mg,0.8mmol), potassium acetate (784mg,8.0mmol), N 2 Under the condition, the reaction is carried out for 2 hours at the temperature of 105 ℃ by microwave, and the system is directly used for the next step.
8. 7- (6-Chloropyrimidin-4-yl) imidazo [1,2-a]Pyridine-5, 6,8-d 3 Preparation of
Figure BDA0002952928540000241
Will (imidazo [1, 2-a)]Pyridin-7-yl-5, 6,8-d 3 ) Boric acid (reaction system of the previous step) with additional water (1mL), 4, 6-dichloropyrimidine (1.2g,8.1mmol), Pd (dppf) 2 Cl 2 (295mg,0.40mmol), cesium carbonate (3.9g,12.0mmol), N 2 And reacting for 2 hours at 120 ℃ by using microwaves. EA (100mL) and aqueous sodium chloride (80mL) were added to the reaction mixture to extract a liquid, and the organic phase was concentrated and subjected to silica gel column chromatography (DCM: MeOH ═ 50:1) to give a crude product (2.0g), followed by C18 reverse phase column chromatography (water/methanol ═ 10/90) to give the objective compound (1.0g, crude product).
9. (R) -8- (1- ((6- (imidazo [1, 2-a))]Pyridin-7-yl-5, 6,8-d 3 ) Preparation of pyrimidin-4-yl) amino) propan-2-yl) -N-methylquinoline-4-carboxamide
Figure BDA0002952928540000242
Mixing 7- (6-chloropyrimidin-4-yl) imidazo [1,2-a]Pyridine-5, 6,8-d 3 (270mg crude) was dissolved in THF (12mL) and water (3mL), and (R) -8- (1-aminopropan-2-yl) -N-methylquinoline-4-carboxamide hydrochloride (70mg crude) was added, sodium carbonate (150mg,1.4mmol) was added, and the reaction was carried out at 85 ℃ for 18 hours. The resulting mixture was concentrated and purified by silica gel column chromatography (DCM: MeOH ═ 20:1) to give a crude product (100mg), and then by silica gel plate chromatography (DCM: MeOH ═ 15:1) to give the desired compound (60 mg).
The molecular formula is as follows: c 25 H 20 D 3 N 7 O molecular weight: 440.5LC-MS (M/e): 441.2(M + H)
1 HNMR(400MHz,MeOD):8.97(s,1H),8.42(s,1H),8.03(d,J=8.0Hz,1H),7.97(s,1H),7.79(d,J=2.4Hz,1H),7.72(m,1H),7.63(t,J=8.0Hz,1H),7.52(m,1H),6.91-6.75(m,1H),4.62(q,J=7.2Hz,1H),3.95-3.79(m,2H),3.02(s,3H),1.55-1.45(m,3H).
Example two: preparation of (R) -8- (1- ((6- (3-fluoroimidazo [1,2-a ] pyridin-7-yl-5, 6,8-d3) pyrimidin-4-yl) amino) propan-2-yl) -N-methylquinoline-4-carboxamide (Compound 2-1)
1. 7-chloro-3-fluoroimidazo [1,2-a ]]Pyridine-5, 6,8-d 3 Preparation of (2)
Figure BDA0002952928540000251
Reacting 7-chloroimidazo [1,2-a ]]Pyridine-5, 6,8-d 3 (900mg,5.8mmol) was dissolved in THF (30mL), 60% NaH (348.0mg,8.7mmol) was added at 5 deg.C, after 15min of reaction Selectfluor (4.1g,11.6mmol) was added, the temperature was raised to 65 deg.C and the reaction was allowed to proceed for 18 h. The resulting mixture was concentrated and subjected to silica gel column chromatography (EA: PE ═ 1:3) to obtain the product (270.0mg, yield 26.9%).
2. (3-Fluoroimidazo [1, 2-a)]Pyridin-7-yl-5, 6,8-d 3 ) Preparation of boric acid
Figure BDA0002952928540000252
Reacting 7-chloroimidazo [1,2-a ]]Pyridine-5, 6,8-d 3 (240mg,1.4mmol) was dissolved in 1, 4-dioxane (10mL) and B was added 2 (PIN) 2 (529mg,2.1mmol),Pd 2 (dba) 3 (128.2mg,0.14mmol), tricyclohexylphosphine (78.5mg,0.28mmol), potassium acetate (274.4mg,2.8mmol), N 2 And reacting for 1 hour at 120 ℃ by using microwaves, and directly using the system in the next step.
3. 7- (6-Chloropyrimidin-4-yl) -3-fluoroimidazo [1,2-a]Pyridine-5, 6,8-d 3 Preparation of
Figure BDA0002952928540000253
Reacting (3-fluoroimidazo [1,2-a ]]Pyridin-7-yl-5, 6,8-d 3 ) Boric acid (reaction system above) with additional water (1mL), 4, 6-dichloropyrimidine (311mg,2.1mmol), Pd (dppf) Cl was added 2 (76.8mg,0.105mmol), cesium carbonate (1.37g,4.2mmol), N 2 The reaction was carried out by microwave at 120 ℃ for 1 hour, and the resulting mixture was concentrated and subjected to silica gel column chromatography (DCM: MeOH: 50:1) to obtain the objective compound (190mg, yield 54.1%).
4. (R) -8- (1- ((6- (3-fluoroimidazo [1, 2-a))]Pyridin-7-yl-5, 6,8-d 3 ) Preparation of pyrimidin-4-yl) amino) propan-2-yl) -N-methylquinoline-4-carboxamide
Figure BDA0002952928540000261
Reacting 7- (6-chloropyrimidin-4-yl) -3-fluoroimidazo [1,2-a]Pyridine-5, 6,8-d 3 (162mg,0.65mmol) was dissolved in THF (12mL) and water (3mL), and (R) -8- (1-aminopropyl-2-yl) -N-methylquinoline-4-carboxamide hydrochloride (120mg,0.43mmol) was added, sodium carbonate (228mg,2.1mmol) was added, followed by reaction at 85 ℃ for 16 hours. The system was concentrated and chromatographed on silica gel column (DCM: MeOH ═ 20:1) to give the product (70mg, 35.5% yield).
The molecular formula is as follows: c 25 H 19 D 3 FN 7 O molecular weight: 458.5LC-MS (M/e): 459.3(M + H)
1 HNMR(400MHz,MeOD):8.97(s,1H),8.44(s,1H),8.04(d,J=8.0Hz,1H),7.80(d,J=3.4Hz,1H),7.64(t,J=8.0Hz,1H),7.62-7.58(m,1H),7.52-7.41(m,1H),6.91-6.75(m,1H),4.68-4.58(m,1H),3.95-3.79(m,2H),3.02(s,3H),1.55-1.45(m,3H).
The compounds shown in the following table were prepared using the same or similar methods as the examples above:
Figure BDA0002952928540000271
experimental protocol
An exemplary experimental scheme of a portion of the compounds of the invention is provided below to show the advantageous activity and advantageous technical effects of the compounds of the invention. It should be understood, however, that the following experimental protocols are only illustrative of the present disclosure and are not intended to limit the scope of the present disclosure.
Experimental example 1 in vitro cytological Activity of Compounds of the invention
Abbreviations
EDTA: ethylenediaminetetraacetic acid
DMSO (dimethylsulfoxide): dimethyl sulfoxide
Tris (Tris): tris (hydroxymethyl) aminomethane
Brij-35: polyoxyethylene lauryl ether
DTT: dithiothreitol
And (3) testing the sample: the structural formula and the preparation method of the compound are shown in the examples.
Experimental reagent:
name (R) Brand
ADP-Glo Kinase Assay Promege
DNA-PK Promege
The experimental method comprises the following steps:
1. 1-fold kinase buffer solution is prepared
1) 1-fold kinase buffer
40mM Tris,pH 7.5
0.0055%Brij-35
20mM MgCl 2
0.05mM DTT
2. Compound preparation
1) The initial concentration of the compound to be detected was 1. mu.M, and the concentration was set to 100-fold, that is, 100. mu.M. Mu.l of 10mM compound was taken and 198. mu.l of 100% DMSO was added to prepare a 100. mu.M solution of the compound. 100 μ l of 100-fold compound was added to the second well of the 96-well plate, and 60 μ l of 100% DMSO was added to the other wells. Mu.l of compound from the second well was added to the third well and diluted sequentially 3-fold further down for a total of 10 concentrations.
2) Transfer the highest concentration (400nM) of 100. mu.l of 100% DMSO and the positive control Wortmannin to two empty wells as Max and Min wells, respectively.
3) Echo was used to transfer 50nl of compound to 384-well plates.
3. Preparation of 2 Xkinase solution
1) A2-fold DNA-PK kinase solution was prepared using a 1-fold kinase buffer.
2) Transfer 2.5. mu.l of 2-fold enzyme solution to 384-well reaction wells.
3) Shaking, mixing, and standing at room temperature.
4. Preparation of 2 Xsubstrate solution
1) A2-fold substrate solution was prepared using 1-fold kinase buffer.
2) Transfer 2.5. mu.l of 2-fold substrate solution to 384-well reaction wells to initiate the reaction.
3) Oscillating and mixing evenly.
5. Kinase reaction and termination
1) The 384 well plates were capped and incubated at 28 ℃ for 3 hours.
2) Transfer 5. mu.l ADP-Glo reagent and incubate at 28 ℃ for 2 hours.
6. Detection of reaction results
1) The reaction was stopped by transferring 10. mu.l of the kinase detection reagent to reaction wells of a 384-well plate.
2) Rest for 30 minutes at room temperature.
7. Data reading
Sample values were read at Envision.
8. Inhibition rate calculation
1) Data is copied from Envision.
2) This was converted to inhibition data.
Percent inhibition is (max-conversion)/(max-min) 100. where max refers to the conversion rate of the DMSO control, min refers to the conversion rate of the no enzyme control, and conversion refers to the conversion rate at each concentration of test compound.
3) Data were imported into MS Excel and curve-fitted using XLFit Excel add-in version 5.4.0.8.
The experimental results are as follows:
TABLE 1 in vitro enzymatic Activity data for Compounds of the invention
Figure BDA0002952928540000291
And (4) experimental conclusion:
the result shows that the compound has better inhibition effect on the activity of DNA-PK kinase.
Experimental example 2 CD1 mouse pharmacokinetics experiment of the Compound of the present invention
And (3) testing the sample: the compound 2-1 of the invention is prepared by self, and the chemical name and the preparation method are shown in the preparation examples of each compound.
The test animals were: CD1 mice, female, purchased from Beijing Wintolidian laboratory animal technology, Inc., weighing 22-25g, for a total of 12 mice.
Preparing a test article:
the preparation method of the blank solvent (1) comprises the following steps: weighing 28g of HP-beta-CD, adding a proper amount of water for injection to dissolve the HP-beta-CD, then fixing the volume of the water for injection to 100mL, and uniformly mixing in a vortex manner to obtain 28% HP-beta-CD.
The preparation method of the blank solvent (2) comprises the following steps: weighing 20g of HPC, slowly adding 500mL of stirred purified water, then adding 1mL of Tween 80, stirring until the mixture is clear and transparent, diluting to 1000mL, and uniformly stirring to obtain 2% of HPC + 0.1% of Tween 80.
IV (bolus IV) administration:
taking the compound 2-1(2.45mg), adding DMSO (116 mu L), shaking for dissolving, then adding PEG400(116 mu L), vortex and mixing uniformly, finally adding a blank solvent (1) (2.10mL), vortex and mixing uniformly, and keeping the temperature at 50 ℃ for 20min to obtain a clear solution of 1mg/mL, which is used as an IV administration solution of the compound 2-1.
PO (intragastric) administration:
weighing the compound 2-1(3.98mg), placing the compound in a tissue grinder, adding 3.78mL of blank solvent (2), and grinding uniformly at 1200 r/min to obtain suspension liquid medicine with the concentration of 1mg/mL, wherein the suspension liquid medicine is used as PO administration liquid medicine of the compound 2-1.
Experimental method
Figure BDA0002952928540000301
Approximately 50. mu.L of whole blood was collected at each time point by the canthus and placed in the eye containing EDTA-K 2 Centrifuging at 8000 rpm at 4 deg.C for 6min in an anticoagulant tube to obtain plasma sample, and freezing at-80 deg.C in a refrigerator for analysis.
Plasma sample analysis
Adopting a protein precipitation method: taking 20 mu L of a plasma sample, adding 200 mu L of an internal standard (acetonitrile solution containing 50ng/mL of tolbutamide), vortexing for 10min, then centrifuging for 20min at 4000 rpm, taking 100 mu L of supernatant, adding 100 mu L of water, vortexing and uniformly mixing for 3min, and then analyzing by LC-MS/MS.
Results of the experiment
TABLE 2 evaluation results of CD1 mouse PK
Figure BDA0002952928540000311
AUC 0-t Area under curve 0 → t when representing drug; AUC inf Area under curve 0 → ∞ when drug is represented; CL represents clearance; v ss Representing the steady state apparent distribution volume; t is a unit of 1/2 Represents a terminal elimination half-life; t is max Represents the time to peak; c max Expression peak concentration; f% represents the absolute bioavailability.
Conclusion of the experiment
As can be seen from the data in table 2, the compound of the present invention has excellent pharmacokinetic properties, and has high exposure and bioavailability.
Experimental example 3 combination of the Compound of the present invention and Doxil injection of Doxil hydrochloride for nude mice having NCI-H1048 mouse Drug effect experiment of cell lung cancer transplantable tumor model
1. Experimental drugs and materials
1.1 test article
Control compound: prepared according to the prior art method, has the following structure,
Figure BDA0002952928540000312
and (5) a reference substance.
Test compounds: compound 2-1 of the present invention was prepared according to the methods of the examples.
1.2 solvent
Control vehicle: 0.5% MC; test compound vehicle: 2% HPC + 0.1% tween 80.
1.3 reagents
RPMI-1640 culture medium, fetal bovine serum, penicillin, streptomycin, trypan blue, pancreatin, DMSO, 75% ethanol, etc., and doxorubicin hydrochloride liposome injection.
2. Laboratory animal
The species are as follows: rodent nude mice; quantity: 36 are provided; sex: a female; specification: 6 weeks old.
3. Experimental methods
3.1 preparation of Single cell suspensions
Taking NCI-H1048 small cell lung cancer cells in logarithmic growth phase, conventionally digesting with 0.25% pancreatin, suspending in RPMI-1640 culture medium without fetal bovine serum, adding matrix glue at a ratio of 1:1, mixing uniformly, and adjusting to cell concentration of 1 x 10 8 one/mL.
3.2 preparation of tumor-bearing mice
36 nude mice were subcutaneously placed on the right back of the mice, wiped with an alcohol cotton swab and then injected subcutaneously with 0.1ml of single cell suspension using a 1ml syringe.
3.3 Experimental groups
Measuring the diameter of the transplanted tumor by a vernier caliper until the tumor grows to 200mm 3 On the left and right sides, animals are randomly grouped according to the tumor volume and the weight of the mice, before grouping, undesirable tumors such as scattered tumors, long tumors, superficial tumors, small tumors or large tumors are removed, then, tumor-bearing mice are randomly divided into a Vehicle control (Vehicle) group, a doxorubicin 6mg/kg single-use group, a control 100mg/kg + doxorubicin 6mg/kg combined group and a compound 2-1100 mg/kg + doxorubicin 6mg/kg combined group according to the weight and the tumor volume, the number of animals in each group and the administration scheme are shown in the following table:
TABLE 3 administration route, dose and period of drug effect experiment of nude mouse loaded NCI-H1048 small cell lung cancer model
Figure BDA0002952928540000321
Figure BDA0002952928540000331
3.4 administration of drugs
On the day of administration, doxorubicin 6mg/kg single-use group and combined group tail vein (i.v.) were injected with doxorubicin hydrochloride liposome injection once a week (q.w.), the vehicle control group was injected with an equal volume of 5% glucose injection, after 16h, the combined group was orally administered (p.o.) with a dose of compound, the vehicle control group and doxorubicin 6mg/kg single-use group were administered with an equal volume of control vehicle for 4 consecutive days (qd 4), and both doxorubicin hydrochloride liposome injection and compound were treated for 2 cycles (7 days is one cycle), and detailed administration method, administration dose and administration route are shown in table 3.
3.5 tumor diameter measurement
Each group of mice was dynamically observed for the antitumor effect of the test substance using a method for measuring tumor size, starting on the day of administration. Tumor diameter was measured 2 times per week, and mice were weighed simultaneously for each measurement.
3.6 tumor tissue denudation
After the administration, nude mice were sacrificed by cervical dislocation, subcutaneous tumor tissues of the mice were taken, photographed, and weighed.
4. Index of experimental evaluation
4.1 evaluation index of antitumor Effect as relative tumor proliferation Rate T/C (%)
The formula for Tumor Volume (TV) is represented by V-1/2 × a × b 2 (a major diameter; b minor diameter);
relative Tumor Volume (RTV) ═ Vt/V0(Vt is tumor volume at each measurement, V0 is tumor volume at the time of group administration);
T/C% (TRTV/CRTV) × 100% (TRTV: mean RTV in treatment group; CRTV: mean RTV in vehicle control group).
The evaluation standard of curative effect is as follows: T/C > 40% is ineffective; T/C% is less than or equal to 40%, and meanwhile, statistical differences exist, namely P is less than 0.05, and the effect is achieved.
4.2 tumor weight: at the end of the experiment, nude mice were sacrificed by cervical dislocation, subcutaneous tumor tissues were taken, photographed and weighed.
5. Statistical treatment
All experimental results are expressed by mean value + -SEM (standard error), on-Way ANOVA (One-Way Analysis of variance ) is adopted to evaluate the overall difference on the premise of judging the normality and the homogeneity of the variance of the data, when the overall difference is significant, LSD t-test Analysis method is used for comparing the groups, the Dunnett's T3 Analysis method is used for comparing the groups when the variance is not uniform, and P <0.05 is the significant difference. All data were analyzed using SPSS 18.0.
6. The experimental results are as follows:
the NCI-H1048 model results show that the control product and doxorubicin combined group has the tumor inhibiting effect (P <0.05) compared with the Vehicle group, and has no sensitization effect compared with the doxorubicin single group. The combination of compound 2-1+ doxorubicin group had very significant tumor-inhibiting effect compared to the Vehicle group (x, P < 0.001); compared with the doxorubicin single group, the compound has sensitization effect (&, P <0.05), and specific results are shown in figure 1 (& represents comparison between the combined group and a solvent control group, & represents comparison between the combined group and the doxorubicin single group). Moreover, the relative tumor proliferation rate (T/C%) of the compound 2-1+ doxorubicin combination group is obviously lower than that of the control substance + doxorubicin combination group, and the specific result is shown in figure 2.
7. And (4) experimental conclusion:
the compound 2-1 of the invention has better tumor inhibiting effect and sensitization effect than the reference compound.

Claims (11)

1. A compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof,
Figure FDA0003668138860000011
wherein the content of the first and second substances,
X 1 is C;
X 2 、X 4 、X 5 、X 6 、X 7 each independently selected from C (R);
X 3 is N;
x is selected from-NR a -;R a Is hydrogen or deuterium;
y is selected from O or S;
ring a is selected from pyrimidinyl optionally substituted with a substituent that is deuterium;
each R is independently selected from hydrogen, deuterium, halogen, or the following optionally deuterated: hydroxy, amino, C 1-6 Alkyl, halo C 1-6 Alkyl, hydroxy C 1-6 Alkyl, amino C 1-6 Alkyl radical, C 1-6 Alkoxy or halo C 1-6 An alkoxy group;
R 1 selected from optionally deuterated methyl;
R 2 is selected from-NR c R d ;R d Is hydrogen or deuterium;
R c is methyl, wherein said methyl is optionally substituted with deuterium;
R 3 、R 4 、R 5 、R 6 、R 7 each independently selected from hydrogen, deuterium, halogen, or the following optionally deuterated: hydroxy, amino, C 1-6 Alkyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy, halo C 1-6 An alkoxy group;
R 8 、R 9 each independently selected from hydrogen or deuterium;
m is 1;
and X 1 、X 2 、X 3 、X 4 、X 5 、X 6 、X 7 X, ring A, R 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、R 9 At least one of which is deuterium or deuterated.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof,
each R is independently selected from hydrogen, deuterium, halogen, or the following optionally deuterated: amino group, C 1-6 Alkyl, halo C 1-6 Alkyl radical, C 1-6 Alkoxy or halo C 1-6 An alkoxy group.
3. The compound of claim 1, or a pharmaceutically acceptable salt thereof,
each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or the following optionally deuterated: amino, methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, methoxy, ethoxy, monofluoromethoxy or difluoromethoxy.
4. The compound of claim 1, or a pharmaceutically acceptable salt thereof,
R 3 、R 4 、R 5 、R 6 、R 7 each independently selected from hydrogen, deuterium, halogen, trifluoromethyl, trifluoromethoxy, or the following group optionally deuterated: hydroxyl, amino, methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy.
5. The compound of claim 1, or a pharmaceutically acceptable salt thereof,
X 1 is C;
X 2 、X 4 、X 5 、X 6 、X 7 each independently selected from C (R);
X 3 is N; y is O;
x is selected from-NR a -;
Ring a is pyrimidinyl;
each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, monofluoromethyl, difluoromethyl, methoxy, ethoxy, monofluoromethoxy, difluoromethoxy;
R 1 selected from methyl optionally deuterated;
R 2 is selected from-NR c R d
R a Selected from hydrogen or deuterium;
R c selected from optionally deuterated methyl;
R d selected from hydrogen or deuterium;
R 3 、R 4 、R 5 、R 6 、R 7 each independently selected from hydrogen or deuterium;
R 8 、R 9 each independently selected from hydrogen or deuterium;
m is 1;
and X 1 、X 2 、X 3 、X 4 、X 5 、X 6 、X 7 X, ring A, R 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 、R 8 、R 9 At least one of which is deuterium or deuterated.
6. The compound of any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof,
X 3 is N; x 1 Is C; x 2 、X 4 、X 5 、X 6 、X 7 Each independently selected from C (R);
each R is independently selected from hydrogen, deuterium, fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, or the following optionally deuterated: methyl, ethyl, propyl, isopropyl, methoxy, ethoxy.
7. A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure FDA0003668138860000021
Figure FDA0003668138860000031
8. a pharmaceutical formulation comprising a compound according to any one of claims 1 to 7, or a pharmaceutically acceptable salt thereof, in a pharmaceutically acceptable dosage form, comprising one or more pharmaceutically acceptable excipients.
9. A pharmaceutical composition comprising a compound according to any one of claims 1 to 7, or a pharmaceutically acceptable salt thereof, comprising one or more second therapeutically active agents selected from the group consisting of anti-cancer agents including mitotic inhibitors, alkylating agents, anti-metabolites, DNA chimerics, anti-tumor antibiotics, growth factor inhibitors, signaling inhibitors, cell cycle inhibitors, enzyme inhibitors, retinoid receptor modulators, proteasome inhibitors, topoisomerase inhibitors, biological response modifiers, hormonal drugs, angiogenesis inhibitors, cell growth inhibitors, targeting antibodies, HMG-CoA reductase inhibitors and prenyl protein transferase inhibitors.
10. Use of a compound according to any one of claims 1 to 7 or a pharmaceutically acceptable salt thereof, a pharmaceutical formulation according to claim 8, or a pharmaceutical composition according to claim 9, in the manufacture of a medicament for the prevention and/or treatment of benign tumours or cancers, including carcinoma in situ and metastatic carcinoma, in combination with radiotherapy and/or one or more anti-cancer agents.
11. Use of a compound of any one of claims 1-7, or a pharmaceutically acceptable salt thereof, a pharmaceutical formulation of claim 8, or a pharmaceutical composition of claim 9, in the manufacture of a medicament for sensitizing cancer cells to an anti-cancer agent and/or radiation therapy.
CN202110215181.XA 2020-02-25 2021-02-25 Deuterated heterocyclic kinase inhibitors Active CN113372345B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010115530 2020-02-25
CN2020101155306 2020-02-25

Publications (2)

Publication Number Publication Date
CN113372345A CN113372345A (en) 2021-09-10
CN113372345B true CN113372345B (en) 2022-08-16

Family

ID=77570624

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110215181.XA Active CN113372345B (en) 2020-02-25 2021-02-25 Deuterated heterocyclic kinase inhibitors

Country Status (1)

Country Link
CN (1) CN113372345B (en)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG10201704095UA (en) * 2012-04-24 2017-06-29 Vertex Pharma Dna-pk inhibitors

Also Published As

Publication number Publication date
CN113372345A (en) 2021-09-10

Similar Documents

Publication Publication Date Title
CN106986863B (en) DNA-PK inhibitors
TWI772291B (en) Substituted pyrrolopyrimidine CDK inhibitors, pharmaceutical compositions containing the same and uses thereof
TW202128691A (en) Kras mutein inhibitors
DE69828408T2 (en) NAPHTHYRIDINONE FOR INHIBITING THE CELL REPRODUCTION THROUGH PROTEIN TYROSINE KINASE AND CELL CYCLE KINASE
CN112047948A (en) Kras mutant inhibitors
CN108329311B (en) Tricyclic compound as selective estrogen receptor down-regulator and application thereof
EP3312180B1 (en) Use of pteridinone derivative serving as egfr inhibitor
EA021463B1 (en) Purine derivatives and their use as modulators of toll-like receptor 7
KR20060132850A (en) Imidazo[4,5-c]pyridine compounds and methods of antiviral treatment
CN113896710A (en) SHP2 inhibitor and application thereof
CN111732575B (en) N- (3- (pyrimidine-2-yl) phenyl) benzene sulfonamide derivative, pharmaceutical composition, preparation method and application
US9724331B2 (en) Use of maleimide derivatives for preventing and treating leukemia
CN111592541B (en) Macrocyclic kinase inhibitors and uses thereof
CN111362925A (en) 4-pyrimidine formamide compound, pharmaceutical composition, preparation method and application
CN113549092B (en) Tricyclic kinase inhibitors
CN108299420B (en) Pentacyclic compounds as selective estrogen receptor down-regulators and uses thereof
CN111909144A (en) Quinazoline DNA-PK inhibitor
CN112574211B (en) Heterocyclic kinase inhibitors
CN113372345B (en) Deuterated heterocyclic kinase inhibitors
CN113416181B (en) Quinazoline derivative and application thereof
CN112574179B (en) DNA-PK inhibitors
WO2017133542A1 (en) Pharmaceutical composition comprising pyridopyrimidine derivative or pharmaceutically acceptable salt thereof
TW458977B (en) 6,7-disubstituted-4-aminopyrido[2,3-D] pyrimidine compounds
CN111303163B (en) Compound with JAK kinase inhibitory activity, preparation method, composition and application
CN111909147B (en) DNA-PK inhibitors

Legal Events

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