CN116082310A - Bipyridine amide derivative, and preparation and application thereof - Google Patents

Bipyridine amide derivative, and preparation and application thereof Download PDF

Info

Publication number
CN116082310A
CN116082310A CN202310132225.1A CN202310132225A CN116082310A CN 116082310 A CN116082310 A CN 116082310A CN 202310132225 A CN202310132225 A CN 202310132225A CN 116082310 A CN116082310 A CN 116082310A
Authority
CN
China
Prior art keywords
bipyridyl
cells
compound
cell
rxrα
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202310132225.1A
Other languages
Chinese (zh)
Other versions
CN116082310B (en
Inventor
吴振
方美娟
陈俊
赵泰格
何凤明
钟艺晶
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.)
Xiamen University
Original Assignee
Xiamen University
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 Xiamen University filed Critical Xiamen University
Priority to CN202310132225.1A priority Critical patent/CN116082310B/en
Publication of CN116082310A publication Critical patent/CN116082310A/en
Application granted granted Critical
Publication of CN116082310B publication Critical patent/CN116082310B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • 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

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The bipyridyl amide derivative disclosed by the invention can target RXR alpha, block the interaction of p-RXR alpha and PLK1, induce cancer cells to undergo apoptosis and G2/M phase cell cycle arrest, and inhibit tumor cell proliferation, so that the bipyridyl amide derivative can be used for treating and preventing cancers or other related diseases.

Description

Bipyridine amide derivative, and preparation and application thereof
Technical Field
The invention relates to the technical field of chemical medicines, in particular to bipyridyl amide derivatives, and preparation and application thereof.
Background
The retinoid X receptor alpha (Retinoid X receptor alpha, RXR alpha) belongs to the nuclear receptor superfamily of transcription factors, exists in different tissues, and directly or indirectly participates in various biological processes such as apoptosis, metabolism, cell cycle, inflammation and the like. More and more researches show that the abnormal expression and the abnormal function of RXR alpha are closely related to the development of tumor generation, and are important drug targets for treating cancers. Rxrα agonists Bexarotene (Targretin) were approved by the us Food and Drug Administration (FDA) in 1999 for treatment of Cutaneous T Cell Lymphoma (CTCL) patients resistant to prior drug treatment. In recent years, research shows that the novel RXR alpha modulator-acylhydrazone derivative (XS 060, compound B10) can target and block the interaction of p-RXR alpha/PLK 1, induce cancer cells to generate centrosome and chromosome abnormality, DNA damage reaction and G2/M phase retardation, and inhibit tumor growth. Thus, novel rxrα modulators targeting the p-rxrα/PLK1 interaction are an effective strategy for tumor treatment.
Disclosure of Invention
The invention aims to solve the problems in the prior art and provide bipyridyl amide derivatives shown in a general formula (I) or pharmaceutically acceptable salts, hydrates, solvates, metabolites, metabolic precursors or prodrugs thereof.
Figure BDA0004084420290000011
Wherein the bipyridine is 3,4 '-bipyridine or 4,4' -bipyridine; l represents a saturated or unsaturated alkyl chain, preferably-CH 2 -、-C 2 H 4 -or-ch=ch-; r is R 1 4-piperidinemethanol, N-methylpiperazine, 4-methylpiperidine, 4-hydroxypiperidine and 2-methylaminoethanol; r is R 2 Is H, halogen, alkyl, alkoxy, preferably H, para-F, para-CH 3 para-OCH 3 meta-F, meta-CH 3 Or meta-OCH 3
Furthermore, the bipyridyl amide derivative shown in the general formula (I) is any one of the following 29 structural formulas:
Figure BDA0004084420290000021
a pharmaceutical composition comprising said bipyridyl amide derivative, or a pharmaceutically acceptable salt or pharmaceutically acceptable carrier of said derivative.
The pharmaceutically acceptable salt is a pharmaceutically acceptable addition salt of a compound of the general formula (I) and an acid. Among these acids used for salification include inorganic acids, preferably hydrochloric acid, sulfuric acid, phosphoric acid and methanesulfonic acid, and organic acids, preferably acetic acid, trichloroacetic acid, propionic acid, butyric acid, maleic acid, p-toluenesulfonic acid, malic acid, malonic acid, cinnamic acid, citric acid, fumaric acid, camphoric acid, digluconic acid, aspartic acid and tartaric acid.
The pharmaceutically acceptable carrier refers to an excipient or diluent that does not cause significant irritation to the organism and does not interfere with the biological activity and properties of the compound being administered.
The bipyridyl amide derivative or the application of the bipyridyl amide derivative or the pharmaceutically acceptable salt thereof in preparing RXR alpha modulator medicaments.
The bipyridyl amide RXR alpha modulator can be used for treating cancers or tumor-related diseases, including breast cancer, lung cancer, liver cancer and cervical cancer.
The bipyridyl amide derivative or the pharmaceutically acceptable salt thereof has good binding activity with RXR alpha protein and has the effect of inhibiting the growth and development of nude mice xenograft tumors.
Compared with the prior art, the technical scheme of the invention has the beneficial effects that:
the bipyridyl amide derivatives disclosed by the invention can be used for resisting the transcriptional activity of RXR alpha through targeting combination, and destroying the interaction between p-RXR alpha and PLK1 in tumor cells to ensure that the tumor cells generate G2/M phase cell cycle retardation and inhibit the proliferation of the tumor cells, so that the bipyridyl amide derivatives can be used for treating and preventing cancers or tumor-related diseases.
Drawings
FIG. 1 is a graph showing the experimental results of example 3 of the present invention.
FIG. 2 is a graph showing the experimental results of example 5 of the present invention.
FIG. 3 is a graph showing the experimental results of example 6 of the present invention.
FIG. 4 is a graph showing the experimental results of example 7 of the present invention.
FIG. 5 is a graph showing the experimental results of example 8 of the present invention.
FIG. 6 is a graph showing the experimental results of example 9 of the present invention.
FIG. 7 is a graph showing the experimental results of example 10 of the present invention.
FIG. 8 is a graph showing the experimental results of example 11 of the present invention.
FIG. 9 is a graph showing the experimental results of example 12 of the present invention.
FIG. 10 is a graph showing the experimental results of example 13 of the present invention.
FIG. 11 is a graph showing the experimental results of example 14 of the present invention.
Detailed Description
In order to make the technical problems, technical schemes and beneficial effects to be solved more clear and obvious, the invention is further described in detail below with reference to the accompanying drawings and embodiments.
The preparation method of bipyridyl amide derivatives provided by the invention comprises the following steps:
1. substituted or unsubstituted phenylacetic acid or phenylpropionic acid or cinnamic acid is taken as a raw material, thionyl chloride is taken as a solvent, stirring is carried out, heating is carried out until reflux is carried out, substituted or unsubstituted phenylacetyl chloride or phenylpropionyl chloride or cinnamoyl chloride is generated, 2-amino-4-bromopyridine or 2-amino-5-bromopyridine is condensed with the acyl chloride to obtain substituted or unsubstituted N- (4-bromopyridine-2-yl) -3-phenylacetamide, N- (4-bromopyridine-2-yl) -3-phenylpropionamide or N- (4-bromopyridine-2-yl) -3-cinnamamide intermediate (1), N- (5-bromopyridine-2-yl) -3-phenylacetamide, N- (5-bromopyridine-2-yl) -3-phenylpropionamide and N- (5-bromopyridine-2-yl) -3-cinnamide intermediate (2).
2. 4- (4-bromopyridine-2-yl) amine derivatives are obtained by substitution of 4-bromo-2-fluoropyridine, and then reacted with bisboronic acid pinacol ester to obtain the intermediate boric acid ester (3).
3. Suzuki coupling is carried out on the intermediate boric acid ester (3) and the intermediate (1) or (2) respectively to obtain a target compound (I), wherein the specific synthetic route is as follows:
Figure BDA0004084420290000041
in a preferred embodiment of the invention, wherein the bipyridine is 3,4 '-bipyridine or 4,4' -bipyridine; l represents-CH 2 -、-C 2 H 4 -or-ch=ch-; r is R 1 4-piperidinemethanol, N-methylpiperazine, 4-methylpiperidine, 4-hydroxypiperidine and 2-methylaminoethanol; r is R 2 Is H, para-F, para-CH 3 para-OCH 3 meta-F, meta-CH 3 Or meta-OCH 3
TABLE 1 Structure, hydrogen Spectrum of the Compounds of the invention 1 H NMR) and High Resolution Mass Spectrometry (HRMS)
Figure BDA0004084420290000051
Figure BDA0004084420290000061
Figure BDA0004084420290000071
Figure BDA0004084420290000081
Figure BDA0004084420290000091
Figure BDA0004084420290000101
Example 1: preparation of N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [4,4' -bipyridyl ] -2-yl) -3-cinnamamide (A28)
This example illustrates the synthesis of N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [4,4' -bipyridyl ] -2-yl) -3-cinnamide derivatives of the invention using the synthesis of A28 as an example, and the specific procedure is as follows:
(1) Synthesizing an intermediate cinnamoyl chloride: in a dry 100mL reaction flask, cinnamic acid (2.00 g,0.18 mmol) was added at room temperature and thionyl chloride as solvent and heated to reflux for 1h. TLC monitoring the reaction was completed, stopping the reaction, and vacuum spin-drying the solvent under reduced pressure to obtain 2.12g of cinnamoyl chloride as a pale yellow oil, with a yield of 94.6%.
(2) Synthesis of intermediate N- (4-bromopyridin-2-yl) -3-cinnamide: in a dry 50mL reaction flask, 2-amino-4-bromopyridine (1.00 g,5.78 mmol), pyridine (1.4 mL,17.34 mmol) and THF (10 mL) were added sequentially under ice-bath. Stirred under ice bath conditions for 0.5h. A THF mixture of cinnamoyl chloride (1.15 g,6.94 mmol) was then slowly added dropwise while stirring, and the mixture was reacted under ice-bath conditions for 1 hour. TLC monitors that the reaction is finished, the reaction is stopped, the solvent is decompressed, concentrated in vacuum, added with ice water and stirred, solid is separated out, and a filter cake (crude product) is obtained through suction filtration. The crude product obtained was separated by column chromatography on silica gel (petroleum ether: ethyl acetate=5:1, v/v) to give 1.13g of pale yellow N- (4-bromopyridin-2-yl) -3-cinnamamide in 66.5% yield.
(3) Synthesis of intermediate (1- (4-bromopyridin-2-yl) piperidin-4-yl) methanol: dissolving 4-bromo-2-fluoropyridine (2.00 g,11.36 mmol) and 4-piperidinemethanol (1.57 g,13.63 mmol) in 25mL of DMF, adding anhydrous potassium carbonate (4.71 g,34.08 mmol), heating to 90 ℃ for 3h after nitrogen replacement reaction system, stopping heating after TLC detection of complete reaction, pouring the reaction solution into 100mL of ice water under stirring, precipitating solid, suction filtering, taking filter cake, and drying to obtain white solid (1- (4-bromopyridin-2-yl) piperidin-4-yl) methanol 2.89g, yield 94.3%
(4) Synthesis of (1- (4, 5-tetramethyl-1, 3, 2-dioxoborolan-2-yl) pyridin-2-yl) piperidin-4-yl) methanol: after (1- (4-bromopyridin-2-yl) piperidin-4-yl) methanol (2.89 g,10.07 mmol), pinacol ester of bisboronic acid (3.05 g,12 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (0.1 mmol) and dry anhydrous potassium acetate (0.93 g,30 mmol) were added to 25mL of dry 1, 4-dioxane, nitrogen was replaced, the reaction was allowed to proceed to 110℃under nitrogen protection for 3h, TLC was performed to detect (1- (4-bromopyridin-2-yl) piperidin-4-yl) methanol, the solvent was removed by vacuum concentration, ethyl acetate was extracted three times with water (3X 100 mL), the organic phase was combined, the organic phase was washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, the drying agent was removed by filtration, and the residue after concentration of the organic phase under vacuum was separated and purified by column chromatography on silica gel (isocratic elution method, dichloromethane: methanol=20:1, v/v) to obtain white powder (1- (4-bromopyridin-2-yl) piperidin-4-yl) methanol (3X, 5-2-yl) in yield, 5-2-tetramethyl-2-piperidinyl) piperidine (2-yl) methanol) was obtained by heating to a reaction
(5) N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [4,4' -bipyridyl ] -2-yl) -3-cinnamamide (A28): a50 mL round bottom flask was weighed out, intermediate N- (4-bromopyridin-2-yl) -3-cinnamamide (302.0 mg,1 mmol) was dissolved in 5mL of solvent (ethylene glycol dimethyl ether: water=4:1), the reaction was allowed to stand still for 3 times with nitrogen, the reaction was stopped by TLC detection, the solvent was removed by concentration under reduced pressure, and the residue was stirred with silica gel, and purified by column chromatography (isocratic elution method, ethyl acetate=1:1, v/v) to give compound N- (2 '- (4- (hydroxymethyl) piperidin-1, 4' -yl) -2 '-bipyridyl ] -3.150.5 mg as a pale yellow powder, wherein N- (4' - (4-hydroxyphosphinyl) ferrocene ] palladium dichloride (36.6 mg,0.05 mmol) was dissolved in 5mL of solvent (ethylene glycol dimethyl ether: water=4:1), nitrogen was allowed to stand still for 3 times, the reaction was allowed to stand still at 80 ℃, and the solvent was removed by concentration under reduced pressure, and the purification was performed by column chromatography (isocratic elution method: ethyl acetate=1:1, v/v).
Other compounds listed in table 1: n- (2 '- (4-methylpiperidin-1-yl) - [3,4' -bipyridin ] -6-yl) -2- (m-tolyl) acetamide (A1), 2- (3-methoxyphenyl) -N- (2 '- (4-methylpiperidin-1-yl) - [3,4' -bipyridin ] -6-yl) acetamide (A2), 2- (3-fluorophenyl) -N- (2 '- (4-methylpiperidin-1-yl) - [3,4' -bipyridin ] -6-yl) acetamide (A3), N- (2 '- (4-methylpiperidin-1-yl) - [3,4' -bipyridin ] -6-yl) -2- (p-tolyl) acetamide (A4), N- (2 '- (4-methylpiperazin-1-yl) - [3,4' -bipyridin ] -6-yl) -2- (m-tolyl) acetamide (A5), 2- (3-methoxyphenyl) -N- (2 '- (4-methylpiperazin-1-yl) - [3,4' -bipyridin ] -6-yl) acetamide (A6), 2- (3-fluorophenyl) -N- (2 '- (4-methylpiperazin-1-yl) - [3,4' -bipyridin ] -6-yl) acetamide (A7), N- (2 '- (4-methylpiperazin-1-yl) - [3,4' -bipyridin ] -6-yl) -2- (p-tolyl) acetamide (A8), N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [3,4' -bipyridin ] -6-yl) -2- (m-tolyl) acetamide (A9), N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [3,4' -bipyridin ] -6-yl) -2- (3-methoxyphenyl) acetamide (a 10), 2- (3-fluorophenyl) -N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [3,4' -bipyridin ] -6-yl) acetamide (a 11), N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [3,4' -bipyridin ] -6-yl) -2- (p-tolyl) acetamide (a 12), N- (2 '- ((2-hydroxyethyl) (methyl) amino) - [3,4' -bipyridyl ] -6-yl) -2- (m-tolyl) acetamide (A13), N- (2 '- ((2-hydroxyethyl) (methyl) amino) - [3,4' -bipyridyl ] -6-yl) -2- (3-methoxyphenyl) acetamide (A14), 2- (3-fluorophenyl) -N- (2 '- ((2-hydroxyethyl) (methyl) amino) - [3,4' -bipyridyl ] -6-yl) acetamide (A15), N- (2 '- ((2-hydroxyethyl) (methyl) amino) - [3,4' -bipyridyl ] -6-yl) -2- (p-tolyl) acetamide (A16), N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [3,4' -bipyridyl ] -6-yl) cinnamamide (A17), (E) -3- (3-fluorophenyl) -N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [3,4' -bipyridyl ] -6-yl) acrylamide (A18), (E) -N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [3,4' -bipyridyl ] -6-yl) -3- (m-tolyl) acrylamide (a 19), N- (2 '- (4-methylpiperidin-1-yl) - [4,4' -bipyridyl ] -2-yl) -2- (m-tolyl) acetamide (a 20), 2- (3-methoxyphenyl) -N- (2 '- (4-methylpiperidin-1-yl) - [4,4' -bipyridyl ] -2-yl) acetamide (a 21), 2- (3-fluorophenyl) -N- (2 '- (4-methylpiperidin-1-yl) - [4,4' -bipyridyl ] -2-yl) acetamide (a 22), N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [4,4' -bipyridyl ] -2-yl) -2- (m-tolyl) acetamide (a 23), N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [4,4' -bipyridyl ] -2-yl) -2- (3-methoxyphenyl) acetamide (a 24), 2- (3-fluorophenyl) -N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [4,4' -bipyridin ] -2-yl) acetamide (A25), (E) -3- (3-fluorophenyl) -N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [4,4' -bipyridin ] -2-yl) acrylamide (A26), (E) -N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [4,4' -bipyridin ] -2-yl) -3- (m-tolyl) acrylamide (A27), N- (2 '- (4- (hydroxymethyl) piperidin-1-yl) - [4,4' -bipyridin ] -2-yl) -3-phenylpropionamide (A29) are similar to the synthetic procedure for compound A28.
Example 2: evaluation of G2/M phase cell cycle arrest Activity of Compounds of the invention
The effect of the compounds of the invention on the progression of the cell cycle was examined using a flow cytometer. Taking tumor cells MDA-MB-231 in the logarithmic phase, inoculating the tumor cells into a 6-well plate, culturing overnight at 37 ℃, adding a culture medium containing 1 mu M of a compound to be tested, continuously culturing for 12 hours, discarding the culture medium, pre-cooling 1 XPBS for rinsing, collecting the cells into a pre-cooled 1.5mL ep tube after pancreatin digestion, centrifuging at 2500rpm for 5 minutes, removing the supernatant after centrifugation, pre-cooling PBS for two times, removing the supernatant after centrifugation, lightly flicking the tube wall to avoid cell agglomeration, adding 70% ethanol for 4 ℃ for more than 4 hours, 2500rpm, centrifuging for 5 minutes, removing the supernatant after pre-cooling 1 XPBS for two times, lightly flicking the tube wall to avoid cell agglomeration, adding 500 mu L of DAPI solution with a working concentration of 1 mu g/mL (2 mg/mL DAPI storage liquid is prepared by ultrapure water, diluting to a working concentration when in use) suspending the cells, staining the cells at room temperature for 20 minutes, and detecting cell cycle distribution by a flow cytometer. The experimental results are shown in table 2, and the results show that the compounds of the invention can significantly interfere with the cell cycle progression and block the cell cycle in the G2/M phase.
TABLE 2 inhibition of proliferation activity of Compounds A1-A29 and G2/M phase blocking Rate of cancer cells
Figure BDA0004084420290000131
Example 3: evaluation of the Activity of the Compounds of the invention for binding to RXR alpha
The dual-luciferase reporter gene system is an important experimental method for transcriptional level regulation research. The nuclear receptor rxrα acts as a transcription factor, whose transcriptional activity may be affected in the presence of its exogenous ligand. Thus, the effect of the compounds of the invention on rxrα transcriptional activity was first tested using a reporter gene assay.
The logarithmic phase of Hela cells were inoculated in 48-well plates, cultured overnight, each well was simultaneously transfected with 50ng of pBind-RXR alpha-LBD and 50ng of pG5-Luciferase plasmid, transfected for 24 hours, with DMSO as a negative control, 0.1. Mu.M of RXR alpha agonist CD3254 as a positive control, 1. Mu.M of test compound was co-incubated with 0.1. Mu.M of CD3254, 3 duplicate wells were placed in each group, and culture was continued for 24 hours at 37 ℃. And then detecting the fluorescence intensity of the target compound after the action by using a dual-luciferase reporter gene kit (Promega) to obtain the compound which remarkably reduces the RXR alpha transcriptional activity activated by CD 3254. The transcriptional activity of RXRalpha after the compounds were tested by the reporter gene assay is shown in FIG. 1. The results of fig. 1 demonstrate that the compounds of the present invention are capable of significantly inhibiting the transcriptional activity of rxrα.
Example 4: evaluation of the anti-tumor cell proliferation Activity of the Compound (5. Mu.M) of the present invention
Cell lines: human breast cancer cells (MDA-MB-231), human liver cancer cells (HepG) 2 ) Human non-small cell lung cancer cell (A549)
Cell culture: MDA-MB-231, A549, hepG 2 DMEM medium was used.
Preparation of compound test samples: the compounds were dissolved in DMSO to prepare 10mM stock solution for subsequent experiments, which was stored in a-20 ℃ refrigerator.
MTT experiment: taking logarithmic phase cell MDA-MB-231, A549 and HepG 2 Inoculated in 96-well plates and cultured overnight at 37 ℃. Adding 5 mu M compound solution to be tested into each hole, arranging 3 compound holes in each group, taking the DMSO solution diluted in the same proportion as a reference, continuously culturing for 72 hours, adding MTT with the working concentration of 0.5mg/mL, culturing for 3-4 hours at 37 ℃, discarding the supernatant, adding 100 mu L of DMSO into each hole, vibrating the plate for 10 minutes to fully dissolve formazan, and detecting the absorbance at 492nm by an enzyme-labeling instrument. The experimentally measured data is expressed as:
Figure BDA0004084420290000141
and calculating the inhibition rate. The experimental results (Table 2) show that the compounds of the invention have obvious inhibition effect on tumor cell proliferation.
Example 5: selective analysis of Compound A28 binding to RXRalpha-LBD, nur77-LBD and RARgamma-LBD
The experimental procedure is as in example 3. The pBind-RXRalpha-LBD, pBind-Nur77-LBD and pBind-RARgamma-LBD plasmids were transfected into HeLa cells, respectively, with 0.1. Mu.M CD3254, 5. Mu.M tripterine (Celastrol) and 0.1. Mu.M ATRA as positive controls for the three nuclear receptors, respectively. HeLa cells transfected with the above plasmid were treated with 31.25nM A28 for 24h, respectively, and the experimental results are shown in FIG. 2. The results of fig. 2A show that a28 significantly inhibits transcriptional activation of rxrα, while the results of fig. 2B-2C show that a28 has no significant effect on the transcriptional activity of Nur77, rarγ.
Example 6: binding assays of Compound A28 with RXRalpha-LBD
(1) Surface Plasmon Resonance (SPR): determination of dissociation constant (K) of A28 binding to RXRalpha-LBD by surface plasmon technique (SPR) D ). 50. Mu.g of purified RXR alpha-LBD protein was coupled to CM5 sensor chip, and the change in the dissociation process of binding to the protein on the chip surface was captured by SPR detector at various concentrations A28 (0.17. Mu. Mol/L, 0.28. Mu. Mol/L, 0.47. Mu. Mol/L, 0.78. Mu. Mol/L, 1.30. Mu. Mol/L, 2.16. Mu. Mol/L, 3.60. Mu. Mol/L, 6.00. Mu. Mol/L and 10.0. Mu. Mol/L) using a BIAcore T200 instrument, and the binding affinity was calculated by kinetic fitting of the data. As shown in FIG. 3A, the compound A28 can generate good combination with RXRalpha-LBD protein in vitro, K D 39.29.+ -. 1.12nM.
(2) Fluorescence titration experiments: the binding of a28 to rxrα -LBD was analyzed by fluorescence titration experiments. 200. Mu.L of purified RXRalpha-LBD protein solution with the concentration of 1.0. Mu.M is taken into a cuvette, and the A28 solution is added dropwise to ensure that the action concentration is from 9.95nM to 2345.32nM. And (3) sequentially detecting the fluorescence intensity of the small molecule-protein mixed solution with different concentrations by using a fluorescence spectrophotometer at 25 ℃, wherein the width of an excitation slit is 5nm, the width of an emission slit is 10nm, the excitation wavelength is 280nm, and the emission spectrum is recorded as 285-450 nm. The resulting data were plotted using Origin 2021 software and the dissociation constants were calculated according to standard formulas, the experimental results being shown in FIG. 3B. Finally fitting out dissociation constant K of A28 and RXRalpha-LBD combination D 146.93 + -16.17 nM.
(3) Dual luciferase reporter gene assay: the experimental procedure is as in example 3. The binding of compound a28 to rxrα -LBD was analyzed using a dual luciferase reporter assay. FIG. 3C shows the results of an experiment to detect the binding of compound A28 to RXRα -LBD using a dual luciferase reporter system. The results of fig. 3C show that compound a28 inhibits transcriptional activation of rxrα in a concentration dependent manner.
Example 7: evaluation of in vitro anti-tumor cell proliferation Activity of Compound A28
(1) Cell colony formation assay: the ability of compound a28 to inhibit cell proliferation was determined by cell colony formation. Taking 1000-2000 logarithmic MDA-MB-231 cells, inoculating the cells into a 6-well plate, culturing the cells for 48 hours at 37 ℃, adding culture mediums containing different concentrations (0.98, 3.91,15.63,62.50 and 250.00 nM) of A28 for further culturing for one week, discarding the culture mediums, washing the culture mediums for 3 times by 1 XPBS, fixing the methanol at room temperature for 15min, discarding the methanol, washing the culture mediums for 3 times by 1 XPBS, drying the culture mediums, adding a 0.5% crystal violet solution (diluted by PBS) for dyeing, dyeing the culture mediums at room temperature for 10-15min, removing dye liquor, washing the culture mediums with ultra-pure water until the culture mediums have no background color, observing colony count by a microscope, and photographing and recording. The experimental results are shown in fig. 4, and the results show that the compound a28 can significantly inhibit the proliferation of MDA-MB-231 cells in vitro.
(2) MTT experiment:
cell lines: MDA-MB-231, HCC1937, MCF-7 (human breast cancer cell line), A549, H460 (human non-small cell lung cancer cell line), hepG 2 (human liver cancer cell line), hela (human cervical cancer cell line), MCF-10A (human normal mammary epithelial cell), LO 2 (human normal hepatocytes), haCaT (human immortalized epidermal cells), HK2 (human renal cortex proximal tubular epithelial cells).
Cell culture: MDA-MB-231, HCC1937, MCF-7, A549, hepG 2 Hela, haCaT, HK2 used DMEM medium, H460, LO 2 The culture was performed using RPMI-1640 medium, and MCF-10A was performed using MCF-10A-dedicated medium.
The experimental method for each concentration inhibition was the same as in example 4. Calculating half inhibition concentration IC according to inhibition ratio of each concentration 50 The results are shown in Table 3. Experimental results show that the compound A28 has excellent inhibition effect on various cancer cell lines and IC 50 Is between 0.016 and 2.334 mu M; for normal mammary gland cell MCF-10A inhibition was greatly reduced, IC 50 The value was 18.580.+ -. 0.580. Mu.M.
TABLE 3 half inhibition concentration of Compound A28 on each cell line
Figure BDA0004084420290000161
Example 8: compound A28 interferes with cell cycle progression
The experimental procedure is as in example 2. The effect of compound a28 on the progression of the cell cycle was examined by flow cytometry. The logarithmic phase tumor cells MDA-MB-231 and A549 are respectively inoculated into a 6-well plate, cultured overnight at 37 ℃, added with culture media containing different concentrations (15.6, 31.2,62.5,125.0 nM) of A28 for continuous culture for 12 hours, and then the cell cycle distribution is detected by a flow cytometer. The results of the experiment are shown in FIG. 5, and demonstrate that compound A28 inhibited 85.88% and 46.39% of cells in the G2/M phase, respectively, 12h after 125nM action on MDA-MB-231 and A549 cells.
Example 9: compound A28 induces apoptosis
(1) Annexin V-FITC/PI apoptosis detection assay: the effect of compound a28 on apoptosis was examined by flow cytometry. Taking logarithmic phase cells MDA-MB-231 and A549, inoculating the cells into a 6-well plate, culturing overnight at 37 ℃, adding a culture medium containing different concentrations (31.2, 62.5,125.0,250.0 nM) of A28, continuously culturing for 24 hours, discarding the culture medium, pre-cooling for 1×PBS for rinsing, collecting the cells into a pre-cooled 1.5mL ep tube after pancreatin digestion, centrifuging for 5 minutes, removing the supernatant, pre-cooling for two times by PBS, removing the supernatant after centrifugation, lightly flicking the tube wall to avoid cell agglomeration, and using an Annexin V-FITC/PI apoptosis detection kit (Yeasen) for 20 minutes at room temperature and detecting apoptosis by a flow cytometer. The experimental results are shown in FIG. 6A, where compound A28, after 24h at 250nM, resulted in apoptosis in 40.40% and 52.70% of MDA-MB-231 and A549 cells, respectively.
(2) Protein immunoblotting (Western blot) to detect apoptosis: after apoptosis, the protein expression associated with the cells is significantly changed. PARP proteins produce cleavage when cells undergo early apoptosis, and thus PARP cleavage can be used as a marker of early apoptosis. Bcl-2 and Mcl-1 are anti-apoptotic proteins of the Bcl-2 family, which have important regulatory effects on cell growth and differentiation. Bcl-2 and Mcl-1 both inhibit apoptosis by inhibiting cytochrome C release. The effect of compound A28 on the cleavage of the apoptosis marker protein PARP, bcl-2 and Mcl-1 expression was examined by Western blot. Taking logarithmic phase cell MDA-MB-231, inoculating the cell MDA-MB-231 into a 6-well plate, culturing overnight at 37 ℃, adding a culture medium containing A28 for continuous culture for a corresponding time, discarding the culture medium, pre-cooling for 1×PBS for 3 times, adding a cell lysate, performing ice lysis for 30min,12000g, centrifuging at 4 ℃ for 15min, taking a supernatant, adding a loading buffer,100 ℃, boiling for 10min, and detecting a protein sample by Western blot.
The experimental results are shown in fig. 6B and 6C. The results of FIG. 6B show 125nM A28 inducing PARP cleavage in a time-dependent manner and the results of FIG. 6C show A28 acting on MDA-MB-231 cells for 24h to up-regulate PARP cleavage and down-regulate Bcl-2 and Mcl-1 expression in a concentration-dependent manner.
Example 10: compound A28 regulates and relies on RXR alpha to induce cancer cell mitosis disorder and cell proliferation inhibition
Transfecting PX330-sgCTL and PX330-sgRXR alpha plasmids into A549 cells to obtain A549-WT and A549-RXR alpha, respectively -/- And (3) cells. Then using Western blot experiment to detect RXR alpha protein expression in two kinds of cells, using flow cytometry to detect A459-WT and A549-RXR alpha -/- Cell cycle distribution of cells after 12h of Compound A28 (125 nM) detection of A459-WT and A549-RXR alpha Using MTT assay -/- Cell viability of cells after 72h treatment at different concentrations of a 28. Subsequently, the results are shown in fig. 7.
FIG. 7A shows detection of A549-WT and A549-RXR alpha by Western blot -/- Rxrα protein expression in cells. The results of fig. 7A show that PX330-sgrxrα effectively inhibits rxrα expression in cells.
FIGS. 7B and 7C show 125nM A28 treatment A549-WT and A549-RXRalpha -/- After 12h of cells, cell cycle distribution. The results of fig. 7B and 7C show that: in A549-WT cells, A28 was able to block 79.66% of the cell cycle in the G2/M phase; however, when RXR alpha is expressedWhen knocked out, a28 had reduced ability to block the cell cycle, with only 45.99% of cells in G2/M phase.
FIG. 7D shows A549-WT and A549-RXRalpha treated with A28 at different concentrations for 72h -/- Cell proliferation. The results show that A28 inhibits A549-WT and A549-RXRalpha -/- IC for cell proliferation 50 The method comprises the following steps of: 0.1959 + -0.0260 and 3.3619 + -0.1754 μM, suggesting that A28 inhibition of cell proliferation is dependent on RXR alpha expression.
FIGS. 7E and 7F show A28-regulated RXR alpha protein modification. The results of fig. 7E show that a28 induces rxrα protein production of modified bands in a time dependent manner. The results of fig. 7F show that a28 selectively regulates rxrα protein expression without significant impact on Nur77 protein expression, while it has no significant impact on rxrα upstream, cell cycle related CDK1 protein expression.
Fig. 7G shows the source of rxrα modification bands induced by a 28. The results in fig. 7G show that the a 28-induced modified rxrα bands disappeared under the action of phosphatase, indicating that a28 induced rxrα to produce a phosphorylated modification.
Example 11: compound A28 induces mitogenic arrest of cancer cells by disrupting p-RXRalpha interaction with PLK1
The pCMV-Myc-RXRalpha and pCMV-Flag-PLK1 plasmids were transfected into HeLa cells for 24h, and after transfection, heLa cells were treated with 500nM A28 for 3h. The supernatant was discarded, washed twice with pre-chilled 1 XPBS, and pre-chilled cell lysates were added, lysed on ice for 30min, all cells scraped off with a spatula and transferred to pre-chilled 1.5mL ep tubes. The cell lysate was placed in a low temperature centrifuge, 4℃and 12000g, and centrifuged for 15min. Collecting supernatant, collecting 15% protein supernatant, adding loading buffer, and decocting at 100deg.C for 10min to obtain Input. To the remaining protein supernatant, 1. Mu.g of Flag antibody stock was added, followed by incubation overnight at 4℃in a vertical mixer. mu.L of protein A/G agarose beads (protein A/G agarose beads washed three times with 1 XPBS) were added per sample, and then placed in a vertical mixer and incubated at 4℃for 3-4 h to allow for coupling of antibodies to agarose beads. After immunoprecipitation, the supernatant was carefully aspirated off at 4℃at 3000rpm for 5min, the agarose beads left at the bottom of the tube were washed 3-4 times with lysate, the remaining supernatant was aspirated off as much as possible the last time, and 20. Mu.L of a 2X loading buffer was added to the precipitated agarose beads, which were boiled at 100℃for 10min. Protein samples were detected by Western blot experiments.
The experimental results are shown in FIG. 8. FIG. 8 shows the interaction of p-RXRalpha with PLK1 after 500nM A28 treatment of HeLa cells for 3h. The results show that a28 significantly inhibited the interaction of p-rxrα with PLK 1.
Example 12: compound A28 induces spindle disorders and chromosomal misplacement
Thymidine double blocking method: placing a glass slide in a 12-well plate in advance, inoculating MDA-MB-231 cells in logarithmic phase into the 12-well plate, and inoculating 5% CO at 37deg.C 2 Culturing overnight. First blocking: the medium was changed to a medium containing 2mM fresh thymidine (protected from light), 37℃and 5% CO 2 Culturing for 16h; first release: discarding the supernatant, washing with 1×PBS for 2 times, changing into normal culture medium, and culturing for 8 hr; second blocking: the medium was again changed to a medium containing 2mM fresh thymidine (protected from light), 37℃and 5% CO 2 Culturing for 16h; second release: at this point the cells are at the G1/S phase boundary (i.e., 0 h), then enter S phase over time, 8h at G2/M phase, and 10h at M phase.
Immunofluorescent staining: washing the cell climbing slice twice with ice-cold PBS, fixing with 3.7-4.0% paraformaldehyde (in PBS) at room temperature for 15min, washing with PBS three times, adding membrane penetrating fluid (0.1% triton X-100+0.1M Glycine,in PBS), and standing at room temperature for 8min; the slide glass is washed twice by PBS, 30 mu L of diluted primary antibody (the primary antibody is diluted to 1:100 by PBS) is carefully dripped on the slide glass for multiple times, the primary antibody completely covers all cells on the slide glass, and the primary antibody cannot overflow the slide glass due to surface tension, so that the recovery is facilitated, and the slide glass is carefully incubated at 4 ℃ overnight. Recovering the primary antibody, soaking in PBS for 5min, discarding PBS, repeating for 3 times, and washing the primary antibody. Taking 30 mu L of diluted secondary antibody (the secondary antibody is diluted to 1:100 by PBS), carefully dripping the secondary antibody on a glass slide for multiple times, completely covering all cells on the glass slide, and incubating for 2 hours at room temperature without overflowing the glass slide due to surface tension. Discarding the secondary antibody, adding PBS, washing for 5min each time, and washing for 3 times. mu.L of DAPI dye (diluted 1:20000 with PBS) was taken and incubated for 5min at room temperature. Gently washed 5 times with PBS. The slide was cleaned and a slide oil was added to the top. The slide was then transferred to the spot with the oil of the slide using pointed tweezers, with slow movements, taking care not to create bubbles. After capping, the pieces were air-dried in the dark. Observing the prepared slide under a fluorescence microscope, and storing the slide in a refrigerator at-20 ℃ after the experiment is finished.
FIG. 9 shows MDA-MB-231 cells treated with thymidine double block, after addition of 62.5nM A28 for 12h, staining of the corresponding proteins with alpha-tubulin antibodies, staining of chromatin with DAPI, and observation of cell spindle formation and chromosome distribution by confocal microscopy. The results show that the chromosomes of the control group are aligned at the equatorial plate position and the spindle structure is normal. Compared with the control group, the chromosome arrangement of a large number of cells treated by A28 is severely disordered, and the shape of a spindle body is abnormal, so that the cell process is blocked, and the late M phase cannot be entered.
Example 13: pharmacokinetic study of Compound A28
Pharmacokinetic study protocols were approved according to guidelines of the institutional animal care and use committee of the Xiamen university. Preferably compound A28 is evaluated pharmacokinetic in Sprague-Dawley rats of 10-14 weeks of age (200-220 g). The compounds were formulated in 10% DMSO, 2% T-80 and 88% ultrapure water and injected intraperitoneally at a dose of 25mg/kg (i.p.). Female SD rats were randomly divided into 3 groups, and blood samples were stored in tubes containing heparin sodium (10 mg/mL, 30. Mu.L) at 0.083h, 0.25h, 0.5h, 1h, 2h, 4h, 8h, 12h, 24h, 48h after dosing. After centrifugation at 4000rpm for 10min at 4 ℃ (100 μl), plasma was separated from blood. Compound a28 levels in plasma samples were determined by LC/MS using ABI 3200QTRAP triple quadrupole system operating in positive electrospray mode. The pharmacokinetic parameters of compound a28 were fitted by DAS 3.0 software.
Fig. 10 shows the pharmacokinetic versus time profiles of the preferred compound a28, with specific parameters in table 4.
TABLE 4 pharmacokinetic parameters of Compound A28
Figure BDA0004084420290000191
Figure BDA0004084420290000201
Example 14: preferably, the compound A28 can inhibit the growth of breast cancer MDA-MB-231 cell nude mice xenograft tumor
MDA-MB-231 cell suspension was subcutaneously injected into the right anterior axilla of nude mice. A control group (solvent) was set, a low dose group (12.5 mg/kg) and a high dose group (25.0 mg/kg), and administration was continued for 15 days.
FIGS. 11A and 11B show that the preferred compound A28 is capable of significantly inhibiting the growth of MDA-MB-231 cell xenograft tumors;
FIG. 11C shows that the preferred compound A28 is capable of significantly inhibiting the weight of MDA-MB-231 cell xenograft tumors;
fig. 11D shows that the preferred compound a28 did not cause significant weight loss in nude mice during dosing, with no significant toxicity;
FIG. 11E shows representative H & E, ki67 and RXR alpha staining of tumors treated with control and preferred compound A28;
fig. 11F shows H & E staining of the major organs of mice in control and dosing groups.

Claims (10)

1. The bipyridyl amide derivative is characterized by having a structure shown in the following general formula (I):
Figure FDA0004084420280000011
wherein the bipyridine is 3,4 '-bipyridine or 4,4' -bipyridine; l represents a saturated or unsaturated alkyl chain; r is R 1 4-piperidinemethanol, N-methylpiperazine, 4-methylpiperidine, 4-hydroxypiperidine or 2-methylaminoethanol; r is R 2 Is H, halogen, alkyl or alkoxy.
2. Bipyridylamide derivatives according to claim 1, characterized in that: the L is selected from-CH 2 -、-C 2 H 4 -or-ch=ch-.
3. Bipyridylamide derivatives according to claim 1, characterized in that: the R is 2 Selected from H, para-F, para-CH 3 para-OCH 3 meta-F, meta-CH 3 Or meta-OCH 3
4. Bipyridylamide derivatives according to claim 1, characterized in that: the structure is as follows
Figure FDA0004084420280000012
Figure FDA0004084420280000021
5. The method for producing bipyridyl amide derivatives according to any one of claims 1 to 4, comprising the following synthetic routes:
Figure FDA0004084420280000022
6. a medicament, characterized in that: a bipyridyl amide derivative according to any one of claims 1 to 4 or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable carrier thereof.
7. Use of the medicament of claim 6 for the preparation of a cancer therapeutic composition.
8. The use according to claim 7, wherein: the cancer is lung cancer, breast cancer or liver cancer.
9. Use of the medicament of claim 6 as an interaction blocker of p-rxrα with PLK 1.
10. Use of the medicament of claim 6 as a modulator of the nuclear receptor rxrα.
CN202310132225.1A 2023-02-17 2023-02-17 Bipyridine amide derivative, and preparation and application thereof Active CN116082310B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310132225.1A CN116082310B (en) 2023-02-17 2023-02-17 Bipyridine amide derivative, and preparation and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310132225.1A CN116082310B (en) 2023-02-17 2023-02-17 Bipyridine amide derivative, and preparation and application thereof

Publications (2)

Publication Number Publication Date
CN116082310A true CN116082310A (en) 2023-05-09
CN116082310B CN116082310B (en) 2024-08-20

Family

ID=86211970

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310132225.1A Active CN116082310B (en) 2023-02-17 2023-02-17 Bipyridine amide derivative, and preparation and application thereof

Country Status (1)

Country Link
CN (1) CN116082310B (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070293533A1 (en) * 2006-06-06 2007-12-20 Ginn John D SUBSTITUTED 3-AMINO-THIENO[2,3-b]PYRIDINE-2-CARBOXYLIC ACID AMIDE COMPOUNDS AND PROCESSES FOR PREPARING AND THEIR USES
WO2016126085A2 (en) * 2015-02-04 2016-08-11 비욘드바이오주식회사 Heterocyclic compound and pharmaceutical composition comprising same
CN108383779A (en) * 2018-05-03 2018-08-10 华侨大学 A kind of preparation method of 3 '-aryl-bipy 2,2' bipyridyl -6- carboxamides derivatives
CN110078661A (en) * 2019-05-09 2019-08-02 华侨大学 A kind of preparation method of 3- alkenyl-bipy 2,2' bipyridyl -6- carboxamides derivatives
CN110078662A (en) * 2019-05-09 2019-08-02 华侨大学 A kind of preparation method of 3- acyl methyl-bipy 2,2' bipyridyl -6- carboxamides derivatives
CN115611885A (en) * 2022-08-29 2023-01-17 厦门大学 N-substituted-5- ((4-substituted pyrimidin-2-yl) amino) -1H-indole-2-carboxamide derivatives

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070293533A1 (en) * 2006-06-06 2007-12-20 Ginn John D SUBSTITUTED 3-AMINO-THIENO[2,3-b]PYRIDINE-2-CARBOXYLIC ACID AMIDE COMPOUNDS AND PROCESSES FOR PREPARING AND THEIR USES
WO2016126085A2 (en) * 2015-02-04 2016-08-11 비욘드바이오주식회사 Heterocyclic compound and pharmaceutical composition comprising same
CN108383779A (en) * 2018-05-03 2018-08-10 华侨大学 A kind of preparation method of 3 '-aryl-bipy 2,2' bipyridyl -6- carboxamides derivatives
CN110078661A (en) * 2019-05-09 2019-08-02 华侨大学 A kind of preparation method of 3- alkenyl-bipy 2,2' bipyridyl -6- carboxamides derivatives
CN110078662A (en) * 2019-05-09 2019-08-02 华侨大学 A kind of preparation method of 3- acyl methyl-bipy 2,2' bipyridyl -6- carboxamides derivatives
CN115611885A (en) * 2022-08-29 2023-01-17 厦门大学 N-substituted-5- ((4-substituted pyrimidin-2-yl) amino) -1H-indole-2-carboxamide derivatives

Also Published As

Publication number Publication date
CN116082310B (en) 2024-08-20

Similar Documents

Publication Publication Date Title
CN109563060B (en) IDO1 inhibitor and preparation method and application thereof
CN104136437B (en) N-(5S, 6S, 9R)-4-(2-oxo-2,3-dihydro-1H-imidazo [4,5-B] pyridine-1-yl) piperidines-1-carboxylic acid 5-amino-6-(2,3-difluorophenyl)-6,7,8,9-tetrahydrochysene-5H-cycloheptatriene is [B] pyridine-9-base ester Hemisulphate also
CN111902417B (en) Diaryl macrocyclic compound, pharmaceutical composition and application thereof
WO2020228635A1 (en) Egfr kinase inhibitor and application thereof in preparing anti-cancer drug
CN114591293A (en) Fused ring compound as Nav1.8 inhibitor and application thereof
TWI723480B (en) Fused ring derivatives used as fgfr4 inhibitors
CN110577526B (en) Salt of bromodomain structural protein inhibitor and preparation method and application thereof
CN113461668A (en) Novel biphenyl derivative, preparation method and medical application thereof
CN116082310B (en) Bipyridine amide derivative, and preparation and application thereof
JP2022517396A (en) EGFR inhibitor salt, crystalline form and method for producing it
WO2023274280A1 (en) Crystal form of biphenyl derivative inhibitor and preparation method therefor
CN114907387B (en) Pyrimido pyrrole KRAS inhibitor and preparation method and application thereof
CN115536657A (en) Salt of biphenyl derivative inhibitor, crystal form and preparation method thereof
AU2021348707B2 (en) Crystal form of pyridinylphenyl compound and preparation method therefor
CN114507190B (en) Deuterated 1, 4-benzodiazepine-2, 5-diketone compound and application thereof
TW202313604A (en) Pharmaceutically acceptable salt of pyrazoloheteroaryl derivative and crystal form thereof
CN116528859A (en) Crystal form containing free alkali of di-fused ring derivative inhibitor and preparation method and application thereof
CN115715291A (en) Bruton's tyrosine kinase inhibitor and preparation method thereof
CN117645568A (en) N- ([ 4,4' -bipyridine ] -2-yl) cinnamamide derivative, and preparation and application thereof
CN116751186B (en) Preparation and application of estrogen receptor modulator
TWI788444B (en) Crystal form of fgfr4 inhibitor and its preparation method thereof
WO2022095930A1 (en) Deuterated derivative as atx inhibitor, and application thereof
EP4091670A1 (en) Crystal of imidazopyridinone compound or salt thereof
CN116535423A (en) Compound for inhibiting/inducing degradation of EGFR kinase, and pharmaceutical composition and application thereof
CN118812526A (en) CysLT1Receptor antagonist, preparation method, pharmaceutical 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