CN112574179B - DNA-PK inhibitors - Google Patents

DNA-PK inhibitors Download PDF

Info

Publication number
CN112574179B
CN112574179B CN202011038984.4A CN202011038984A CN112574179B CN 112574179 B CN112574179 B CN 112574179B CN 202011038984 A CN202011038984 A CN 202011038984A CN 112574179 B CN112574179 B CN 112574179B
Authority
CN
China
Prior art keywords
inhibitors
compound
pharmaceutically acceptable
alkyl
butyl
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
CN202011038984.4A
Other languages
Chinese (zh)
Other versions
CN112574179A (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 CN112574179A publication Critical patent/CN112574179A/en
Application granted granted Critical
Publication of CN112574179B publication Critical patent/CN112574179B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention belongs to the technical field of medicines. In particular, the invention relates toA compound useful as an inhibitor of DNA-dependent protein kinase (DNA-PK), a pharmaceutically acceptable salt thereof, a deuterogen thereof and a stereoisomer thereof, a pharmaceutical composition and a preparation containing the compound, the pharmaceutically acceptable salt thereof, the deuterogen thereof and the stereoisomer thereof, and uses of the compound, the pharmaceutically acceptable salt thereof, the deuterogen thereof and the stereoisomer thereof.

Description

DNA-PK inhibitors
Technical Field
The invention belongs to the technical field of medicines. In particular, the invention relates to a compound which can be used as an inhibitor of DNA-dependent protein kinase (DNA-PK), pharmaceutically acceptable salts, deuterons and stereoisomers thereof, a pharmaceutical composition and a preparation containing the compound, the pharmaceutically acceptable salts, the deuterons and the stereoisomers thereof, and applications of the compound, the pharmaceutically acceptable salts, the deuterons and the stereoisomers 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 on DNA metabolic processes, resulting in DNA damage. After DNA damage, a series of cellular responses such as damaged DNA repair can be triggered, and the repair results in the improvement of cell survival, which is one of the mechanisms of tumor cells resisting to radiotherapy and chemotherapy. DNA double strand breaks, if not repaired in time and integrity, can lead to cell death through apoptosis or/and mitotic disturbances. Therefore, by inhibiting the repair of these DNA damages, the sensitivity of cells to radiotherapy and chemotherapy can be improved, and the proliferation of cells can be inhibited.
In human and other higher eukaryotes, repair of DNA Double Strand Breaks (DSBs) 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. Non-homologous end-joining 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 phosphatidylinositol 3 kinase (PI3K) superfamily member, is a serine/threonine protein kinase; the PI3K superfamily also includes ATM, ATR, mTOR and 4 PI3K isomers. When DNA-PK binds to a DNA fragment, its kinase activity is activated. The important function of Ku is to combine with the end of DNA, recruit DNA-PKcs, and the two compose DNA-PK holoenzyme and activate DNA-PKcs; the activated DNA-PKcs directs Artemis to bind to the damaged site, relies on its ribozyme activity for DNA end-cutting treatment to facilitate ligation repair, then the XRCC 4/DNA-Ligase IV complex is recruited by the activated DNA-PKcs, and finally the DNA-Ligase IV localizes and ligates the broken DNA double-stranded end to complete repair. 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 non-ligatable ends or other forms of damage at the endpoints; finally, the DNA ends are ligated.
Because tumor cells have a higher basal level of endogenous replication stress and DNA damage (oncogene-induced replication stress) and the efficiency of DNA repair mechanisms in tumor cells is low, the sensitivity of tumor cells to DNA-PK increases and the effectiveness of chemo-radiotherapy increases. Normal cells are less sensitive to DNA-PK inhibition than tumor cells, and the safety window is increased.
Therefore, it is desirable to develop compounds that are inhibitors of DNA-PK that synergistically enhance the effects of chemotherapy and radiation therapy while reducing damage to normal cells and reducing side effects.
Disclosure of Invention
The invention aims to provide a compound which has a novel structure and a good inhibition effect on DNA-PK. 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:
the invention provides a compound shown in the following general formula (I), pharmaceutically acceptable salt thereof, a deuteride thereof or a stereoisomer thereof:
Figure BDA0002705312530000021
wherein, X1、X2、X3、X4、X5、X6Are each independently selected from-C (R)3) -or-N-;
y is selected from-NR4-、-CH2-, -S-, -O-, -C (O), -S (O) -or-S (O)2-;
Each R3Each independently selected from hydrogen, halogen, hydroxyl, cyano, amino, nitro, C1-6Alkyl radical, C1-6Alkylamino radical, C1-6Alkoxyamino, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy, amino C1-6Alkoxy radical, C1-6Alkylthio, halo C1-6Alkylthio, hydroxy C1-6Alkylthio or amino radicals C1-6An alkylthio group;
R2is selected from C1-6Alkyl, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl, -NH-R4、-O-R4or-S-R4
R1Each R4Each independently selected from H, C1-6Alkyl radical, C3-6Cycloalkyl, halo C1-6Alkyl radical, C1-6alkyl-C3-6Cycloalkyl, halo C3-6A cycloalkyl group;
ring a and ring B are each independently selected from 6-10 membered aryl or 5-10 membered heteroaryl optionally substituted with 1 to 4 substituents selected from the group consisting of: deuterium atom, halogen, hydroxy, cyano, nitro, amino, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl radical, C1-6Alkoxy, hydroxy C1-6Alkoxy, amino C1-6Alkoxy, halo C1-6Alkoxy radical, C3-6Cycloalkyl radical, C1-6alkyl-C3-6Cycloalkyl or halo C3-6A cycloalkyl group;
m is selected from 0, 1,2,3 or 4.
In certain embodiments, X4、X5、X6Is selected from-C (R)3)-。
In certain embodiments, ring a is selected from phenyl or 5-6 membered nitrogen containing heteroaryl optionally substituted with 1-4 substituents selected from the group consisting of: halogen, hydroxy, cyano, nitro, amino, C1-6Alkyl, halo C1-6Alkyl radical, C1-6Alkoxy, hydroxy C1-6Alkyl, amino C1-6Alkyl, hydroxy C1-6Alkoxy, amino C1-6Alkoxy, halo C1-6Alkoxy radical, C3-6Cycloalkyl radical, C1-6alkyl-C3-6Cycloalkyl or halo C3-6A cycloalkyl group.
In certain embodiments, ring B is selected from phenyl or 5-6 membered heteroaryl.
In certain embodiments, ring a is selected from the following ring systems:
Figure BDA0002705312530000031
Figure BDA0002705312530000032
independently represent the position at which ring A is connected to ring B;
ring B is selected from the following ring systems:
Figure BDA0002705312530000033
Figure BDA0002705312530000041
independently represent the positions at which ring B and Y, or ring B and ring A, are joined;
each Q1、Q2Are the same or different and are each independently selected from the group consisting of absent, hydrogen atom, deuterium atom, halogen, hydroxy, cyano, nitro, amino, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl radical, C1-6Alkoxy, hydroxy C1-6Alkoxy, amino C1-6Alkoxy radical, C3-6Cycloalkyl radical, C1-6alkyl-C3-6Cycloalkyl or halo C3-6A cycloalkyl group;
n is selected from 0, 1,2 or 3.
In certain embodiments, each Q2Each independently selected from the group consisting of absent, hydrogen atom, fluorine, chlorine, bromine, iodine, hydroxyl, amino, nitro, cyano, methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, trifluoromethyl, methoxy, ethoxy, propoxy, and isopropoxy.
In certain embodiments, each Q2Each independently selected from the group consisting of absent, hydrogen atom, fluorine, chlorine, bromine, iodine, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, hydroxy, cyano, amino or nitro.
In certain embodiments, X4、X5、X6Are each independently selected from-C (R)3)-。
In certain embodiments, Y is selected from-NR4-、-CH2-, -S-or-O-.
In certain embodiments, each R is4Each independently selected from H, C1-6Alkyl radical, C3-6Cycloalkyl, halo C1-6Alkyl radical, C1-6alkyl-C3-6Cycloalkyl, halo C3-6A cycloalkyl group.
In certain embodiments, each R is4Each independently selected from H, C1-4Alkyl radical, C3-6Cycloalkyl, halo C1-4Alkyl radical, C1-4alkyl-C3-6Cycloalkyl, halo C3-6A cycloalkyl group.
In certain embodiments, each R is4Each independently selected from H, C1-4Alkyl or C3-6A cycloalkyl group.
In certain embodiments, ring a is selected from the following ring systems:
Figure BDA0002705312530000042
Figure BDA0002705312530000043
independently represent the position at which ring A is connected to ring B;
each Q1Each independently selected from the group consisting of absent, deuterium atom, halogen, hydroxy, cyano, nitro, amino, C1-4Alkyl, halo C1-4Alkyl radical, C1-4Alkoxy, hydroxy C1-4Alkyl or amino C1-4An alkyl group;
n is selected from 0, 1,2 or 3.
In certain embodiments, ring B is selected from the following ring systems:
Figure BDA0002705312530000051
Figure BDA0002705312530000052
are respectively and independentlyIndicates the position at which ring B is connected to ring Y or ring B is connected to ring A.
In certain embodiments, the compound, a pharmaceutically acceptable salt thereof, a deuteride thereof, or an isomer thereof, has a structure represented by the following general formula (II):
Figure BDA0002705312530000053
in certain embodiments, X1、X3、X4Are each independently selected from-C (R)3) or-N-.
In certain embodiments, R1Selected from hydrogen, C1-4Alkyl or C3-6A cycloalkyl group.
In certain embodiments, R2Is selected from di (C)1-6Alkyl) amino, -NH-R4、-O-R4or-S-R4
Each R4Each independently selected from H, C1-6Alkyl radical, C3-6Cycloalkyl, halo C1-6Alkyl radical, C1-6alkyl-C3-6Cycloalkyl or halo C3-6A cycloalkyl group.
In certain embodiments, each R is4Each independently selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
In certain embodiments, each R is3Each independently selected from hydrogen, halogen, hydroxy, cyano, amino, nitro or C1-4An alkyl group.
In certain embodiments, m is selected from 1,2, or 3.
In certain embodiments, X1Is selected from-N-;
X3、X4are each independently selected from-C (R)3)-;
R1Selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl;
R2selected from methylamino, dimethylamino, ethylamino, diethylamino, n-propylamino, isopropylamino, n-butylamino, isobutylamino, sec-butylamino, tert-butylamino, cyclopropylamino, cyclobutylamino, cyclopentylamino or cyclohexylamino;
each R3Each independently selected from hydrogen, fluoro, chloro, bromo, iodo, hydroxy, amino, cyano, nitro, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, or tert-butyl;
ring a is selected from the following ring systems:
Figure BDA0002705312530000061
each Q1Each independently selected from deuterium atom, fluorine, chlorine, bromine, iodine, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, hydroxy, cyano, amino or nitro;
n is selected from 0, 1,2 or 3;
m is selected from 1,2 or 3.
In certain embodiments, ring a is selected from the following ring systems:
Figure BDA0002705312530000062
in certain embodiments, X1Is selected from-N-.
In certain embodiments, X3、X4Are each independently selected from-C (R)3)。
In certain embodiments, R1Selected from hydrogen, methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, trifluoromethyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
In certain embodiments, R1Selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, cyclopropylA group selected from cyclobutyl, cyclopentyl and cyclohexyl.
In certain embodiments, R2Selected from methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, methylamino, dimethylamino, ethylamino, diethylamino, n-propylamino, isopropylamino, n-butylamino, isobutylamino, sec-butylamino, tert-butylamino, cyclopropylamino, cyclobutylamino, cyclopentylamino or cyclohexylamino.
In certain embodiments, R2Selected from methylamino, dimethylamino, ethylamino, diethylamino, n-propylamino, isopropylamino, n-butylamino, isobutylamino, sec-butylamino, tert-butylamino, cyclopropylamino, cyclobutylamino, cyclopentylamino or cyclohexylamino.
In certain embodiments, each R is3Each independently selected from hydrogen, fluoro, chloro, bromo, iodo, hydroxy, amino, cyano, nitro, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl or tert-butyl.
In certain embodiments, each R is3Each independently selected from hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, amino, cyano or nitro.
In certain embodiments, each Q1Each independently selected from the group consisting of absent, deuterium atom, fluorine, chlorine, bromine, iodine, hydroxyl, amino, cyano, nitro, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, trifluoromethyl, hydroxymethyl, aminomethyl, methoxy, ethoxy, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
In certain embodiments, each Q1Each independently selected from the group consisting of absent, deuterium atom, fluorine, chlorine, bromine, iodine, hydroxyl, amino, cyano, nitro, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, or tert-butyl.
In certain embodiments, each Q1Each independently selected from the group consisting of absent, deuterium atom, fluorine, chlorine, bromine, iodine, methyl, and ethylAlkyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, hydroxy, cyano, amino or nitro. In certain embodiments, each Q1Each independently selected from deuterium atom, fluorine, chlorine, bromine, iodine, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, hydroxyl, cyano, amino or nitro.
In certain embodiments, each Q1Each independently selected from the group consisting of absent, deuterium atom, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, and tert-butyl.
In certain embodiments, n is selected from 0, 1,2, or 3.
In certain embodiments, n is selected from 0 or 1.
In certain embodiments, the compound, a pharmaceutically acceptable salt thereof, a deuteride thereof, or an isomer thereof, has a structure represented by the following general formula (III):
Figure BDA0002705312530000071
wherein, X3Is selected from-C (R)3) -or-N-;
R1selected from hydrogen or C1-4An alkyl group;
R2is selected from di (C)1-6Alkyl) amino, -NH-R4、-O-R4or-S-R4
R3Selected from hydrogen, halogen, hydroxy, cyano, amino, nitro or C1-4An alkyl group;
each R4Each independently selected from H, C1-6Alkyl radical, C3-6Cycloalkyl, halo C1-6Alkyl radical, C1-6alkyl-C3-6Cycloalkyl or halo C3-6A cycloalkyl group;
each Q1Each independently selected from deuterium atom, fluorine, chlorine, bromine, iodine, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, hydroxy, cyano, amino or nitro;
n is selected from 0, 1,2 or 3;
m is selected from 1,2 or 3.
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 of the present invention, a specific compound of formula (I), formula (II), or formula (III), a pharmaceutically acceptable salt thereof, a deutero-isomer thereof, and a stereoisomer thereof, is selected from the compounds shown in table 1 below:
TABLE 1
Figure BDA0002705312530000081
Figure BDA0002705312530000091
Figure BDA0002705312530000101
The invention also provides a pharmaceutical preparation, which contains the compound shown in the general formula (I), the general formula (II) or the general formula (III), the pharmaceutically acceptable salt, the deuteron or the stereoisomer thereof, and one or more medicinal carriers and/or diluents; the pharmaceutical preparation can be prepared into any clinically or pharmaceutically acceptable dosage form.
The invention also provides a pharmaceutical composition which contains the compound shown in the general formula (I), the general formula (II) or the general formula (III), the pharmaceutically acceptable salt, the deuteron or the stereoisomer thereof and one or more second therapeutic active agents. In certain embodiments, the second therapeutically active agent is 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 agents, angiogenesis inhibitors, cell growth inhibitors, targeting antibodies, HMG-CoA reductase inhibitors, and prenyl protein transferase inhibitors.
In certain embodiments, the ingredients to be combined (e.g., the compound of the present invention, a pharmaceutically acceptable salt thereof, a deutero-derivative thereof, a stereoisomer thereof, and the second therapeutically active agent) may be administered simultaneously or separately in sequential order. For example, the second therapeutic agent can be administered before, concurrently with, or after the administration of the compound of the present invention, a pharmaceutically acceptable salt thereof, a deuteron thereof, or a stereoisomer thereof. Furthermore, the components to be combined may also be administered in combination in the same formulation or in separate and distinct formulations.
The pharmaceutically acceptable carrier and/or diluent useful in the pharmaceutical composition or pharmaceutical formulation of the present invention may be any conventional carrier and/or diluent in the art of pharmaceutical formulation, and the selection of a particular carrier and/or diluent will depend on the mode of administration or the type and state of the disease used to treat a particular patient. The preparation of suitable pharmaceutical compositions for a particular mode of administration is well within the knowledge of those skilled in the pharmaceutical art.
In certain embodiments of the present invention, the above-described pharmaceutical compositions may be administered to a patient or subject in need of such treatment by oral, parenteral, rectal, or pulmonary administration, and the like. 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 pharmaceutical composition can also be prepared into injections, including injection solutions, 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 inhalant or a spray.
In another aspect, the present invention also relates to the use of the compound of the aforementioned general formula (I) or formula (II), a pharmaceutically acceptable salt thereof, a deutero-derivative thereof, or an isomer thereof in the preparation of 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 preparation containing the compound shown in the general formula (I) or the formula (II), the pharmaceutically acceptable salt, the deuteron 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.
Further, the invention also relates to application of a pharmaceutical composition containing the compound shown in the general formula (I) or the formula (II), the pharmaceutically acceptable salt, the deuteron 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 the compound of the aforementioned general formula (I), general formula (II) or general formula (III), a pharmaceutically acceptable salt thereof, a deuteron thereof, or an isomer thereof in the preparation of a medicament for sensitizing cancer cells to an anticancer agent and/or radiotherapy.
Furthermore, the invention also relates to application of a pharmaceutical preparation containing the compound shown in the general formula (I) or the formula (II), pharmaceutically acceptable salt, deuteron or isomer thereof in preparing a medicament for sensitizing cancer cells to anticancer agents and/or radiotherapy.
Furthermore, the invention also relates to application of a pharmaceutical composition containing the compound shown in the general formula (I), the general formula (II) or the general formula (III), pharmaceutically acceptable salt thereof, deuteron or isomer thereof in preparing a medicament for sensitizing cancer cells to anticancer agents and/or radiotherapy. The anti-cancer agents are as described above and include 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 agents, angiogenesis inhibitors, cytostatic agents, targeting antibodies, HMG-CoA reductase inhibitors, and prenyl protein transferase inhibitors.
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 formula (I), formula (II) or formula (III), a pharmaceutically acceptable salt thereof, a deuteroid thereof or a stereoisomer thereof, a pharmaceutical preparation or a pharmaceutical composition as described above; the disease associated with DNAPK overactivation 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 radiotherapy, the method comprising administering to a patient in need thereof an effective amount of the compound of the aforementioned general formula (I), general formula (II), or general formula (III), a pharmaceutically acceptable salt thereof, a deutero-isomer thereof, the aforementioned pharmaceutical preparation or pharmaceutical composition; the anti-cancer agent is as described above.
In another aspect, the invention also provides a process for the preparation of a compound of the invention:
a process for preparing a compound of formula (I):
Figure BDA0002705312530000121
wherein Y' is selected from fluorine, chlorine, bromine or iodine. Corresponding to X1-X6、R1、R2、R3、R4Ring A, ring B, Q1、Q2Y, m, n are as defined aboveThe method is as follows.
The preparation method comprises the following steps:
removing a protecting group from the intermediate 1 under an acidic condition to generate an intermediate 2;
reacting the intermediate 2 with the intermediate 3 under an alkaline condition to generate an intermediate 4;
the intermediate 4 generates a compound shown in a general formula (I) under the action of a Lawson reagent.
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, 1, 4-dioxane, trifluoroacetic acid, water, etc., 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 refers to the condition containing organic base or inorganic base, the organic base includes but is not limited to pyridine, triethylamine, N-dimethylaniline, sodium methoxide, potassium ethoxide, potassium tert-butoxide, sodium tert-butoxide, potassium acetate, N-diisopropylethylamine and the like; preferred inorganic bases include, but are not limited to, 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 includes but is not limited to formic acid, acetic acid, trifluoroacetic acid, propionic acid, benzoic acid, methanesulfonic acid, p-toluenesulfonic acid, tartaric acid and the like; inorganic acids include, but are not limited to, hydrochloric acid, concentrated sulfuric acid, hydrobromic acid, hydrofluoric acid, nitric acid, nitrous acid, boric acid, and the like.
The protecting group includes, but is not limited to, (1) alkoxycarbonyl protecting groups such as benzyloxycarbonyl (Cbz), tert-butoxycarbonyl (Boc), fluorenyl methoxycarbonyl (Fmoc), allyloxycarbonyl (Alloc), trimethylsilyloxycarbonyl (Teoc), and the like; (2) acyl-based protecting groups such as phthaloyl (Pht), p-toluenesulfonyl (Tos), trifluoroacetyl (Tfa), o- (p) nitrobenzenesulfonyl (Ns), pivaloyl and the like; (3) alkyl-based protecting groups, trityl (Trt), 2, 4-dimethoxybenzyl (Dmb), p-methoxybenzyl (PMB), benzyl (Bn), and the like.
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.
[ DEFINITIONS AND GENERAL TERMS ]
In the specification and claims of this application, compounds are named according to chemical structural formula, and if the name and chemical structural formula of a compound do not match when the same compound is represented, the chemical structure is taken as the standard.
In the present application, unless otherwise specified, scientific and technical terms used herein have the 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. Where 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.
"halogen" as referred to herein means fluorine, chlorine, bromine and iodine, preferably fluorine and chlorine.
"C" according to the invention1-6Alkyl "denotes straight or branched alkyl having 1 to 6 carbon atoms, including for example" C1-4Alkyl group "," C1-3Alkyl group "," C1-2Alkyl group "," C2-4Alkyl group "," C2-3Alkyl group "," C3-4Alkyl "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 invention1-4Alkyl "means C1-6Having 1-4 carbon atoms in the alkyl radicalA specific example.
"C" according to the invention1-6Alkoxy "means" C1-6alkyl-O- ", said" C1-6Alkyl "is as defined above.
"C" according to the invention1-4Alkoxy "means" C1-4alkyl-O- ", said" C1-4Alkyl "is as defined above.
The "hydroxy group C" of the present invention1-6Alkyl, amino C1-6Alkyl, halo C1-6Alkyl "means C1-6One or more hydrogens of the alkyl group are each replaced by one or more hydroxyl groups, amino groups or halogens. C1-6Alkyl is as defined above.
The "hydroxy group C" of the present invention1-6Alkoxy, amino C1-6Alkoxy, halo C1-6Alkoxy "means" C1-6One or more hydrogens of "alkoxy" are replaced with one or more hydroxy, amino, or halogen.
"C" according to the invention1-6Alkylamino radical, C1-6Alkoxyamino, di (C)1-6Alkyl) amino "means independently C1-6alkyl-NH-, C1-6alkyl-O-NH-),
Figure 1
"C" according to the invention1-6Alkylthio "means" C1-6alkyl-S- ", said" C1-6Alkyl "is as defined above.
The "hydroxy group C" of the present invention1-6Alkylthio, amino C1-6Alkylthio, halo C1-6Alkylthio "means" C1-6Alkylthio "is one in which one or more hydrogens are replaced with one or more hydroxy, amino, or halogen.
"C" according to the invention3-6Cycloalkyl "refers to a monocyclic saturated and nonaromatic alkyl group containing 3 to 6 carbon atoms, specific examples of which include, but are not limited to: a cyclopropane group (cyclopropyl), a cyclobutane group (cyclobutyl), a cyclopentyl group (cyclopentyl), a cyclohexane group (cyclohexyl), and the like.
The "halo C" of the present invention3-6Cycloalkyl "means" C3-6One or more hydrogens of "cycloalkyl" are replaced with one or more halogens.
"C" according to the invention1-6alkyl-C3-6Cycloalkyl "means" C3-6Cycloalkyl "wherein one or more hydrogens are replaced by one or more C1-6Alkyl groups are substituted.
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 referred to herein includes "5-to 8-membered heteroaryl" and "8-to 10-membered fused heteroaryl".
The "5-to 8-membered 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. "5-8 membered heteroaryl" includes, for example, "5-7 membered heteroaryl", "5-6 membered nitrogen-containing heteroaryl", "6 membered nitrogen-containing heteroaryl", and the like, wherein the heteroatoms in the "nitrogen-containing heteroaryl" contain at least one nitrogen atom, e.g., 1 or 2 nitrogen atoms, or, alternatively, one nitrogen atom and 1 or 2 other heteroatoms (e.g., oxygen atoms and/or sulfur atoms), or, alternatively, 2 nitrogen atoms and 1 or 2 other heteroatoms (e.g., oxygen atoms and/or sulfur atoms).
Specific examples of the "5-to 8-membered 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 heteroaryl group" according to the present invention means a specific example containing 5 to 6 ring atoms, at least one of which is a heteroatom such as nitrogen atom, oxygen atom or sulfur atom.
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.
Optionally, a ring atom (e.g., a carbon atom, a nitrogen atom, or a sulfur atom) in the cyclic structure may be oxo. Specific examples thereof include, but are not limited to: 2-furyl, 3-furyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-pyrimidyl, pyridazinyl (e.g. 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g. 5-tetrazolyl), triazolyl (e.g. 2-triazolyl and 5-triazolyl), 2-thienyl, 3-thienyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-pyrrolyl, 3-pyridyl, 4-pyrimidinyl, 5-pyridazinyl, pyridazinyl (e.g. 3-triazolyl), 2-thiazolyl, 5-thiazolyl, tetrazolyl, triazolyl (e.g. 5-triazolyl), 2-thienyl, etc, Pyrazolyl (e.g., 2-pyrazolyl), isothiazolyl, 1,2, 3-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 3-triazolyl, 1,2, 3-thiadiazolyl, 1,3, 4-thiadiazolyl, 1,2, 5-thiadiazolyl, pyrazinyl, 1,3, 5-triazinyl, and the like.
The term "optionally substituted with …" as used herein means that one or more hydrogen atoms on the substituent may be "substituted" or "unsubstituted" by one or more substituents.
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 compound3H, 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 basic functional groups present in the compounds(e.g., -NH)2Etc.) with a suitable inorganic or organic anion (acid), 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 term "deuteron" as used herein refers to one or more of the structures of the compounds1H quilt2H (also denoted as "D") replaces the structure formed.
An "effective amount" as used herein refers to 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.
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.
Unless otherwise depicted or described, structures recited herein include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers of each asymmetric center. Thus, individual stereochemical isomers as well as enantiomeric, diastereomeric and geometric (or conformational) mixtures of the compounds of the present invention are within the scope of the invention. Compounds that have been drawn with stereochemical centers (usually defined by using shaded or bold bonds) are stereochemically pure, but the absolute stereochemistry is not yet defined. Such compounds may have the R or S configuration. In those cases where absolute configuration has been determined, the chiral center is labeled with (R) or (S) in the plot.
Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention. In addition, unless otherwise specified, the structures depicted herein are also intended to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, having the structure of the invention except that hydrogen is replaced by deuterium or tritium or enriched with13C or14Carbon substitution of C12Compounds of C are within the scope of the invention. Such compounds are useful as analytical tools, probes in bioassays or as DNA-PK inhibitors with improved therapeutic properties.
Advantageous effects of the invention
1. The compound, the pharmaceutically acceptable salt, the deuteron or the stereoisomer thereof has excellent DNA-PK inhibition effect, good pharmacokinetic property in organisms, lasting effect 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, the deuteron or the stereoisomer thereof have better treatment 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.
Detailed Description
The present invention will be described in further detail below with reference to specific examples. It should be understood that the scope of the present invention is not limited to the following examples. All the technologies that can be realized based on the above contents of the present invention belong to the scope of the present invention.
1 preparation example of the Compound of the present invention
In the preparation examples, the abbreviations have the following meanings:
KOH: potassium hydroxide EA: ethyl acetate DCM: methylene dichloride
DMAP: 4-dimethylaminopyridine Na2SO4: sodium sulfate TFA: trifluoroacetic acid
DMF: n, N-dimethylformamide THF: tetrahydrofuran Pd/C: palladium/carbon catalyst
MeOH: methanol EtOH: ethanol Tf2O: trifluoromethanesulfonic anhydride
EDCI: 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride DIEA: n, N-diisopropylethylamine
Lawson's reagent: 2, 4-bis (p-methoxyphenyl) -1, 3-dithio-diphosphetane-2, 4 sulfide
HATU: 2- (7-azabenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate
Preparation example 1: preparation of N-methyl-8- (1- ((2 '-methyl- [4,5' -bipyrimidin ] -6-yl) amino) propan-2-yl) quinoline-4-carbothioic acid amide (Compound 1)
1. Preparation of 8- (3- ((tert-butoxycarbonyl) amino) prop-1-en-2-yl) quinoline-4-carboxylic acid
Figure BDA0002705312530000181
Tert-butyl (2- (4-cyanoquinolin-8-yl) allyl) carbamate (1.2g,3.9mmol), KOH (0).87g,15.5mmol) dissolved in ethanol (30mL), water (10mL) and reacted at 110 ℃ for 10 h. After the reaction, adjusting pH to 5-6 with dilute hydrochloric acid, adding water (20mL), EA extracting for three times (3X 30mL), combining organic phases, and passing through anhydrous Na2SO4Drying, filtering and spin-drying to obtain 770mg of product with a yield of 60.3%.
2. Preparation of tert-butyl (2- (4- (methylcarbamoyl) quinolin-8-yl) allyl) carbamate
Figure BDA0002705312530000182
8- (3- ((tert-butoxycarbonyl) amino) prop-1-en-2-yl) quinoline-4-carboxylic acid (770mg), methylamine hydrochloride (317mg, 4.70mmol), EDCI (893mg,4.70mmol), DMAP (862mg,7.05mmol) were dissolved in DCM (20mL) and reacted at 25 ℃ for 2 h. After the reaction, water (20mL) is added, extraction is carried out for three times, organic phases are combined and are treated by anhydrous Na2SO4Drying, filtering, spin-drying, and reverse C18 separation (acetonitrile/water, acetonitrile 30% -50%) to obtain 800mg crude product.
3. Preparation of tert-butyl (2-4- (methylcarbamoyl) quinolin-8-yl) propyl) carbamate
Figure BDA0002705312530000183
(2- (4- (methylcarbamoyl) quinolin-8-yl) allyl) carbamic acid tert-butyl ester (800mg crude from above) was dissolved in methanol (20mL), Pd/C (200mg), H was added2Reacting for 1h at 25 ℃ under the environment. After the reaction was completed, suction filtration was performed, the filtrate was spin-dried, and forward separation (DCM: MeOH ═ 20:1) gave 646mg of the product in 80.5% yield over two steps.
4. Preparation of 8- (1-aminopropyl-2-yl) -N-methylquinoline-4-carboxamide
Figure BDA0002705312530000184
Tert-butyl (2- (4- (methylcarbamoyl) quinolin-8-yl) propyl) carbamate (646mg) was dissolved in DCM (10mL), and TFA (6mL) was added dropwise and reacted at 25 ℃ for 1 h. And (5) after the reaction is finished, spin-drying to obtain a crude product.
5. Preparation of N-methyl-8- (1- ((2 '-methyl- [4,5' -bipyrimidin ] -6-yl) amino) propan-2-yl) quinoline-4-carboxamide
Figure BDA0002705312530000191
8- (1-Aminoprop-2-yl) -N-methylquinoline-4-carboxamide (300mg of crude from above), 6-chloro-2 '-methyl-4, 5' -bipyrimidine (0.29g,1.4mmol), Na2CO3(0.59g,5.6mmol), dissolved in THF (20mL), water (10mL), reacted at 80 ℃ for 10 h. Adding water (10mL), EA (3X 10mL) for extraction three times, combining the organic phases, and passing through anhydrous Na2SO4Drying, filtration, spin-drying and forward separation (DCM: MeOH ═ 20:1) afforded 130mg of product.
6. Preparation of N-methyl-8- (1- ((2 '-methyl- [4,5' -bipyrimidin ] -6-yl) amino) propan-2-yl) quinoline-4-thiocarboxamide
Figure BDA0002705312530000192
N-methyl-8- (1- ((2' -methyl- [4,5' -bipyrimidin ] -6-yl) amino) propan-2-yl) quinoline-4-carboxamide (130mg,0.31 mmol), Lawson's reagent (254mg,0.62mmol) was dissolved in THF (20mL) and reacted at 75 ℃ for 12 h. After the reaction was complete, spin-dried, NaOH solution (20mL) was added, EA was extracted three times (3 × 10mL), the organic phases were combined, filtered, spin-dried, and separated by spatula (DCM: MeOH ═ 8:1) to afford the title compound 55 mg.
Molecular formula C23H23N7Molecular weight of S429.5 LC-MS (M/e) 430.2(M + H)+)
1H-NMR(400MHz,MeOD)δ:9.09(s,2H),8.90(s,1H),8.40(s,1H),7.84-7.86(d,1H, J=8Hz),7.69-7.71(d,1H,J=8Hz),7.50-7.54(t,1H),7.32(s,1H),6.75(s,1H),455(s,4H), 3.80(s,2H),2.73(s,3H),1.45(s,3H)。
Preparation example 2: preparation of (R) -N-methyl-8- (1- (((2 '-methyl- [4,5' -pyrimidine ] ] -6-yl) amino) propan-2-yl) quinoline-4-carbothioamide (Compound 1-1)
1. Preparation of tert-butyl (2- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) allyl) carbamate
Figure BDA0002705312530000201
To N-tert-butoxycarbonylaminopropyne (150.0g,967.7mmol), B at 0 deg.C2(Pin)2(300.0g, 1181.1 mmol), CuCl (10.0g,100.8mmol), t-BuONa (15.0g,156.1mmol), and P (t-Bu)3MeOH (75.0mL,1875.0mmol) was slowly added dropwise to a suspension of (25.0g,123.6 mmol) 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) gave the crude compound (330.0 g) which was used directly in the next step.
2. Preparation of tert-butyl (2- (4-hydroxyquinolin-8-yl) allyl) carbamate
Figure BDA0002705312530000202
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) Cl2(15.0g,20.5mmol), and Na2CO3A 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 added2(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 BDA0002705312530000203
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 BDA0002705312530000211
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 dropwise2O (120.4g,427.0mmol), and after the addition was complete, the mixture was stirred at-20 ℃ for 20 min. 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 BDA0002705312530000212
8- (1- (((tert-butoxycarbonyl) amino) propan-2-yl) quinolin-4-yl trifluoromethanesulfonate (120.0g,276.5mmol), Zn (CN)2(40.8g,347.5mmol) and Pd (PPh)3)4A 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 BDA0002705312530000213
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 BDA0002705312530000221
8- (1- ((tert-butoxycarbonyl) amino) propan-2-yl) quinoline-4-carboxylic acid (37.7g,114.0mmol), HATU (86.9g, 228.0mmol), MeNH2A 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, beating 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-aminopropan-2-yl) -N-methylquinoline-4-carboxamide hydrochloride
Figure BDA0002705312530000222
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.
9. Preparation of (R) -N-methyl-8- (1- ((2 '-methyl- [4,5' -bipyrimidin ] -6-yl) amino) propan-2-yl) quinoline-4-carboxamide
Figure BDA0002705312530000223
Prepared by the method of reference preparation 1(5) starting material 8- (1-aminopropyl-2-yl) -N-methylquinoline-4-carboxamide was replaced by (R) -8- (1-aminopropyl-2-yl) -N-methylquinoline-4-carboxamide hydrochloride.
10. Preparation of (R) -N-methyl-8- (1- ((2 '-methyl- [4,5' -bipyrimidin ] -6-yl) amino) propan-2-yl) quinoline-4-carbothioamide
Figure BDA0002705312530000231
(R) -N-methyl-8- (1- ((2' -methyl- [4,5' -bipyrimidin ] -6-yl) amino) propan-2-yl) quinoline-4-carboxamide (25mg, 0.06mmol) was dissolved in THF (10mL), Lawson's reagent (49mg,0.12mmol) was added, and the reaction was allowed to proceed for 16 hours at 72 ℃. EA (30mL) and water (10mL) were added to the reaction mixture to extract a liquid, and the organic phase was dried and purified by C18 reverse phase column chromatography (water/methanol 3/7) to obtain a crude product (20mg), which was then purified by silica gel plate (DCM: MeOH 20:1) to obtain the objective compound (10 mg).
The molecular formula is as follows: c23H23N7S molecular weight: 429.5LC-MS (M/e):430.2(M + H)
1HNMR(400MHz,MeOD):9.21-9.08(m,2H),9.03-8.86(m,1H),8.42(s,1H),7.92-7.85 (m,1H),7.76-7.68(m,1H),7.61-7.54(m,1H),7.41-6.75(m,2H),4.62-4.52(q,J=7.2Hz,1H), 3.92-3.73(m,2H),3.30(s,3H),2.74(s,3H),1.55-1.45(m,3H).
2 pharmacological Activity test of Compounds of the present invention
The advantageous effects of the compounds of the present invention are further illustrated below by pharmacological test examples, but this should not be construed as the compounds of the present invention having only the following advantageous effects.
In the examples, abbreviations have the following meanings:
EDTA: ethylenediaminetetraacetic acid
DMSO, DMSO: dimethyl sulfoxide
Tris (Tris): tris (hydroxymethyl) aminomethane
Brij-35: polyoxyethylene lauryl ether
DTT: dithiothreitol
Experimental example 1: in vitro enzymatic Activity of Compounds of the invention
And (3) testing the sample: the structural formula and the preparation method of the compound shown in the table 1 are shown in preparation examples.
Experimental reagent:
name (R) Brand
ADP-Glo Kinase Assay Promege
DNA-PK Promege
The experimental method comprises the following steps:
1. preparing 1 time of kinase buffer solution and stop solution
1) 1-fold kinase buffer
40mM Tris,pH 7.5
0.0055%Brij-35
20mM MgCl2
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.
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 for 2 hours at 28 ℃.
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 of the DMSO control, min refers to the conversion of the no enzyme active control, and conversion refers to the conversion 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.
9. Results
The compounds shown in the table 1 of the invention have the inhibitory activity of 1nM-100nM on DNA-PK kinase, and can effectively inhibit the activity of DNA-PK kinase, for example, the inhibitory activity of 5.3nM (IC) on DNA-PK kinase of the compound 150) The DNA-PK kinase inhibitory activity of Compound 1-1 was 1.6nM (IC)50) Are effective DNA-PK kinase inhibitors.
Figure BDA0002705312530000251

Claims (7)

1. A compound shown in a general formula (II), pharmaceutically acceptable salt thereof, a deuteron compound thereof or a stereoisomer thereof,
Figure FDA0003540050990000011
wherein, the first and the second end of the pipe are connected with each other,
X1is N;
X3is selected from-C (R)3) -or-N-;
X4is selected from-C (R)3)-;
R1Selected from hydrogen or C1-4An alkyl group;
R2is selected from-NH-R4
Each R3Each independently selected from hydrogen, halogen or C1-4An alkyl group;
each R4Each independently selected from H, C1-6Alkyl or halo C1-6An alkyl group;
m is 1;
ring a is selected from the following ring systems:
Figure FDA0003540050990000012
Figure FDA0003540050990000013
independently represent the position at which ring A is connected to ring B;
each Q1Each independently selected from the group consisting of absent, deuterium atom, halogen, C1-4Alkyl or halo C1-4An alkyl group;
n is selected from 0, 1,2 or 3.
2. The compound of claim 1, a pharmaceutically acceptable salt thereof, a deutero-compound thereof, or a stereoisomer thereof, having a structure represented by formula (III),
Figure FDA0003540050990000014
wherein, X3Is selected from-C (R)3) -or-N-;
R1selected from hydrogen or C1-4An alkyl group;
R2is selected from-NH-R4
R3Selected from hydrogen, halogen or C1-4An alkyl group;
each R4Each independently selected from H, C1-6Alkyl or halo C1-6An alkyl group;
each Q1Each independently selected from deuterium atom, fluorine, chlorine, bromine, iodine, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl or tert-butyl;
n is selected from 0, 1,2 or 3;
m is 1.
3. The compound of claim 1, a pharmaceutically acceptable salt thereof, a deuteride thereof, or a stereoisomer thereof, selected from:
Figure FDA0003540050990000021
Figure FDA0003540050990000031
4. a pharmaceutical formulation comprising a compound of any one of claims 1 to 3, a pharmaceutically acceptable salt thereof, a deuteride thereof, or a stereoisomer thereof, comprising one or more pharmaceutically acceptable carriers and/or diluents; the pharmaceutical preparation can be prepared into any clinically or pharmaceutically acceptable dosage form.
5. A pharmaceutical composition comprising a compound of any one of claims 1-3, a pharmaceutically acceptable salt thereof, a deuterogen thereof, or a stereoisomer thereof, comprising one or more second therapeutically active agents selected from the group consisting of anticancer agents selected from the group consisting of 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.
6. Use of a compound according to any one of claims 1 to 3, a pharmaceutically acceptable salt thereof, a deuterode thereof or a stereoisomer thereof, a pharmaceutical formulation according to claim 4 or a pharmaceutical composition according to claim 5, for the preparation of a medicament for the prevention and/or treatment of benign tumors or cancers selected from carcinoma in situ and metastatic cancer, in combination with radiation therapy and/or one or more anti-cancer agents.
7. Use of a compound of any one of claims 1-3, a pharmaceutically acceptable salt thereof, a deutero-derivative thereof, or an isomer thereof, a pharmaceutical formulation of claim 4, or a pharmaceutical composition of claim 5, in the manufacture of a medicament for sensitizing cancer cells to an anti-cancer agent and/or radiation therapy.
CN202011038984.4A 2019-09-29 2020-09-28 DNA-PK inhibitors Active CN112574179B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910936274 2019-09-29
CN2019109362744 2019-09-29

Publications (2)

Publication Number Publication Date
CN112574179A CN112574179A (en) 2021-03-30
CN112574179B true CN112574179B (en) 2022-05-10

Family

ID=75119688

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011038984.4A Active CN112574179B (en) 2019-09-29 2020-09-28 DNA-PK inhibitors

Country Status (1)

Country Link
CN (1) CN112574179B (en)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK3459942T3 (en) * 2012-04-24 2021-03-08 Vertex Pharma DNA-PK inhibitors
LT3424920T (en) * 2013-10-17 2020-08-10 Vertex Pharmaceuticals Incorporated Co-crystals of (s)-n-methyl-8-(1-((2`-methyl-[4,5`-bipyrimidin]-6-yl)amino)propan-2-yl)quinoline-4-carboxamide and deuterated derivatives thereof as dna-pk inhibitors
LT3057959T (en) * 2013-10-17 2018-06-11 Vertex Pharmaceuticals Incorporated Dna-pk inhibitors
MX2019003317A (en) * 2016-09-27 2019-08-05 Vertex Pharma Method for treating cancer using a combination of dna-damaging agents and dna-pk inhibitors.

Also Published As

Publication number Publication date
CN112574179A (en) 2021-03-30

Similar Documents

Publication Publication Date Title
TWI828008B (en) Glp-1r modulating compounds
US10696659B2 (en) Compounds having estrogen receptor alpha degradation activity and uses thereof
TWI819518B (en) Glp-1r modulating compounds
JP6522646B2 (en) Substituted 4,5,6,7-tetrahydro-pyrazolo [1,5-α] pyrazine derivatives as ROS1 inhibitors and 5,6,7,8-tetrahydro-4H-pyrazolo [1,5-α] [1,1 4] Diazepine derivatives
CN108329311B (en) Tricyclic compound as selective estrogen receptor down-regulator and application thereof
TW202246237A (en) Glp-1r modulating compounds
CN104080455A (en) Certain chemical entities, compositions, and methods
EP3214079B1 (en) Six-membered ring benzo derivatives as dpp-4 inhibitor and use thereof
CN112047948A (en) Kras mutant inhibitors
TWI469975B (en) 5-hydroxypyrimidine-4-carboxamide derivatives
JP2017508779A5 (en)
US11312702B2 (en) Substituted quinoline-8-carbonitrile derivatives having androgen receptor degradation activity and uses thereof
WO2016088813A1 (en) Novel diazabicyclo[2.2.2]octane derivative
CN103159755B (en) Alpha-glycosidase inhibitor preparation method and purpose
US20150306070A1 (en) Use of maleimide derivatives for preventing and treating leukemia
CN108299420B (en) Pentacyclic compounds as selective estrogen receptor down-regulators and uses thereof
CN111909144A (en) Quinazoline DNA-PK inhibitor
JP6927548B2 (en) Certain protein kinase inhibitors
CN111499591A (en) ROR gamma modulators
CN105175309A (en) N-benzyl-5/6-formyl amino indol-2-carboxylic acid derivative and application thereof
CN112574179B (en) DNA-PK inhibitors
CN112574211B (en) Heterocyclic kinase inhibitors
CN113372345B (en) Deuterated heterocyclic kinase inhibitors
CN111909147B (en) DNA-PK inhibitors
CN111253403B (en) Amino pyran deuterated derivative, composition and application thereof

Legal Events

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