CN114349657B - Azo microtubulin inhibitor and preparation method and application thereof - Google Patents

Azo microtubulin inhibitor and preparation method and application thereof Download PDF

Info

Publication number
CN114349657B
CN114349657B CN202111529543.9A CN202111529543A CN114349657B CN 114349657 B CN114349657 B CN 114349657B CN 202111529543 A CN202111529543 A CN 202111529543A CN 114349657 B CN114349657 B CN 114349657B
Authority
CN
China
Prior art keywords
azo
tubulin
preparation
cancer
disease
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
CN202111529543.9A
Other languages
Chinese (zh)
Other versions
CN114349657A (en
Inventor
王尊元
马臻
黄文海
沈正荣
曾申昕
梁美好
章迟啸
潘有禄
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.)
Hangzhou Medical College
Original Assignee
Hangzhou Medical College
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 Hangzhou Medical College filed Critical Hangzhou Medical College
Priority to CN202111529543.9A priority Critical patent/CN114349657B/en
Publication of CN114349657A publication Critical patent/CN114349657A/en
Application granted granted Critical
Publication of CN114349657B publication Critical patent/CN114349657B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses an azo compound for inhibiting tubulin, a preparation method and application thereof. The invention takes substituted aniline as raw material, and after diazotization reaction of sodium nitrite, the azo micromolecule is obtained by coupling reaction of the substituted aniline and substituted phenol in an organic solvent. The azo compound for inhibiting the tubulin does not show pharmacological activity in dark environment, but shows affinity to the tubulin under the illumination of specific wavelength due to the special photoisomerization property, and further damages the balance of the tubulin, which can be applied to the preparation of medicines for preventing or treating diseases caused by excessive growth of the tubulin.

Description

Azo microtubulin inhibitor and preparation method and application thereof
Technical Field
The invention relates to an azo compound, in particular to an azo compound capable of inhibiting tubulin and pharmaceutically usable salt thereof, a preparation method thereof and application thereof in medicaments for inhibiting tubulin.
Background
Microtubules (microtubules) are dimers of two types of alpha and beta tubulin, are distributed in a network or bundle shape in cells, and participate in the formation of cytoskeleton, maintenance of cell morphology, cell contraction, intracellular material transport, cell division, and the like. Therefore, by inhibiting the polymerization of tubulin into microtubules in the cell division process or inhibiting the depolymerization of microtubules into tubulin, the mitosis can not be carried out or stagnated, and the interruption of the cell mitosis process can have larger influence on cells, so that the growth of the cells is inhibited, and finally the generation of apoptosis is induced, thereby achieving the purpose of inhibiting the proliferation and the growth of the cells. Given that cancer cells differ most from normal cells in that mitosis of cancer cells is abnormally frequent and uncontrolled, blocking tubulin synthesis is particularly important for the treatment of malignant tumors.
It has now been found that a large number of natural, synthetic and semisynthetic compounds have the effect of affecting the microtubule system, interfering with its normal function, and are collectively known as tubulin inhibitors because most of them bind to tubulin. These tubulin inhibitors have found wide application in medicine, such as anti-tumor, antifungal, insect repellent, etc. Tubulin has become an important target for research and development of novel antitumor drugs, and tubulin inhibitors have also become important antitumor drugs that are effective in clinical use. The action mechanism of the inhibitor is that in tumor cells which are rapidly divided, the inhibition of tubulin polymerization leads the spindle body not to be formed or promotes the tubulin polymerization, leads the spindle body not to be restored to a microtubule network again so as to interfere the normal mitosis process of the cells, leads the mitosis process of the cells to be interrupted and to be stopped in a period, thereby leading the tumor cells to be apoptotic and playing an anti-tumor role. The microtubule plays a very key role in the growth and development of tumor cells, and particularly, the effectiveness of the targeted drugs of taxol, colchicine, vinblastine and the like in clinical treatment of tumors makes the microtubule an ideal target for anti-tumor drug research. Table 1 lists some of the marketed tubulin inhibitors, and many of them are currently under development at various stages.
TABLE 1 microtubulin inhibitors which are partly marketed
Combretastatin A4, CA4 was obtained from african pinus (Combretum caffrum) in south africa. CA4 has cis-stilbene structure which can simulate Colchicine, thus it can target microtubulin in vivo, inhibit its polymerization, and further destroy tumor blood vessel, thereby playing an anti-tumor role. CA4 is effective on various tumor models, and is a very promising antitumor drug, however, the water solubility of CA4 is very poor, and only 11.8 mug/ml, so that the oral bioavailability is lower; meanwhile, the characteristics that the stilbene parent nucleus is easy to become trans, so that the activity disappears limit the clinical application value. In order to overcome the defect of CA4, scientific researchers have conducted extensive structure-activity relationship research on CA4, and developed a water-soluble phosphate prodrug CA4P of CA4, which is currently in clinical trials in stage III.
However, currently, tubulin inhibitors, including CA4, are nonspecific and their biological activity cannot be limited to specific tumor sites, and are toxic to normal cells and healthy cells. This non-specificity can cause serious systemic side effects in the treatment of cancer, such as cardiotoxicity and neurotoxicity, which greatly limit the dosage and route of administration of such inhibitors; at the same time, the development of acquired drug resistance also greatly weakens their clinical therapeutic value.
Disclosure of Invention
The invention aims at overcoming the defects of the prior art and provides an azo compound with a tubulin inhibiting effect, pharmaceutically usable salts thereof, a synthesis method thereof and application thereof in medicines for degrading tubulin.
The aim of the invention is realized by the following technical scheme: an azo compound with tubulin inhibiting effect, having the following structure:
wherein R1 and R2 are independently selected from hydrogen, carbonyl, acetyl, propionyl, butyryl, benzoyl, substituted benzoyl, benzyl, substituted benzyl, phenyl, substituted phenyl, methyl, ethyl, propyl, butyl, trifluoromethyl, alkenyl, allyl, alkynyl, propargyl, and propargyl;
r3, R4, R5, R6, R7, R8, R9, R10 are independently selected from hydrogen, hydroxy, amino, halogen, methyl, ethyl, propyl, butyl, trifluoromethyl, methoxy, ethoxy, propoxy, aryl, substituted aryl;
x, Y is independently hydrogen, oxygen, sulfur or nitrogen.
Further, the azo compound with tubulin inhibition effect is selected from the following structures:
r1 and R2 are independently selected from hydrogen, carbonyl, acetyl, methyl, ethyl, propyl, butyl, trifluoromethyl, alkenyl, allyl, alkynyl, propargyl and propargyl;
r3, R4, R5, R6, R7, R8, R9, R10 are independently selected from hydrogen, hydroxy, amino, halogen, methoxy, ethoxy, propoxy;
x, Y is independently hydrogen, oxygen or nitrogen.
More particularly, the azo compound with tubulin inhibition effect is selected from the following structures:
the azo compound for degrading the tubulin can be used singly or can be prepared into pharmaceutically acceptable salts by a conventional method, wherein the pharmaceutically acceptable salts are hydrochloride, hydrobromide, hydroiodide, sulfate, bisulfate, phosphate, acetate, propionate, butyrate, oxalate, tartrate, methanesulfonate, p-toluenesulfonate, fumarate, taurine, citrate, succinate or mixed salts thereof.
The preparation method of the azo microtubulin inhibitor comprises the following steps:
diazotizing substituted aniline of formula 1, and coupling with substituted benzene compound containing active XH group of formula 2 to generate azo compound of formula 3; then with Z-R 1 Performing condensation reaction to obtain a target compound of the formula I;
wherein R is 1 -R 10 X, Y are the same as in formula I; z is halogen atom Br or I.
The invention relates to a preparation method of azo compounds with tubulin inhibition effect, which specifically comprises the following steps:
diazotizing substituted aniline, and coupling with substituted benzene compound containing active XH group to generate azo compound; depending on the structural requirements, further condensation reactions may be required to obtain the target compounds. Wherein R1 to R10, X, Y are the same as before; z is halogen atom Br or I.
The invention also provides application of the azo compound and pharmaceutically acceptable salt thereof in pharmacy, which comprises the following specific steps: for the preparation of a pharmaceutical preparation for the prevention or treatment of diseases caused by overexpression of tubulin. The diseases are breast cancer, leukemia, skin cancer, cervical cancer, esophageal cancer, lung cancer, glioma and the like, and also comprise neurodegenerative diseases such as Alzheimer's disease, amyotrophic lateral sclerosis, cataract, parkinson's disease, creutzfeldt-Jakob disease and Huntington's disease, and further comprise other diseases such as acute gout, arthralgia, familial mediterranean fever, liver cirrhosis and the like.
Compared with the prior art, the invention has the following beneficial effects:
the azo compound for inhibiting the tubulin exists in a trans isomer with high stability in a dark environment, and does not show pharmacological activity; but, due to its characteristic photoisomerization properties, it turns into cis-isomer under light of a specific wavelength, shows affinity to tubulin, destroys the balance of tubulin, inhibits its polymerization, and further destroys the growth of cells. Therefore, the azo compound for inhibiting the microtubulin can be applied to the preparation of medicaments for preventing or treating diseases caused by excessive growth of the microtubulin, including tumors such as breast cancer, leukemia, skin cancer, cervical cancer, esophageal cancer, lung cancer, glioma and the like, neurodegenerative diseases such as Alzheimer disease, amyotrophic lateral sclerosis, cataract, parkinson's disease, creutzfeldt-Jakob disease and Huntington's disease, and other diseases such as acute gout, arthralgia, familial mediterranean fever, liver cirrhosis and the like.
Description of the drawings:
FIG. 1. UV-visible absorption spectrum of Azo-F under 368nm (blue light) irradiation;
FIG. 2. UV-visible absorption spectrum of Azo-F at 625nm (red light);
FIG. 3 HPLC spectra of Azo-F compounds without irradiation;
FIG. 4. Spectra of Azo-F compounds after 20min exposure to blue light;
FIG. 5. Spectra of Azo-F compounds after 100min exposure to blue light;
FIG. 6 shows a molecular docking diagram (panel a) and a partial enlargement (panel b) of a compound and a protein.
Detailed Description
The structure, the preparation method and the use of the pharmaceutical preparation for preventing or treating diseases caused by the overexpression of tubulin according to the present invention are further described below with reference to examples, but the present invention is not limited thereto.
Analytical data for the samples were determined by the following instrument:
the thermometer is uncorrected; bruker DRX400 nuclear magnetic resonance apparatus; agilent 5975 mass spectrometer; bruker Vector 22 IR spectrometer.
Example 1 Synthesis of Azo-H
A100 ml three-necked flask was selected, added with a magnetic rotor, fitted with a thermometer and a constant pressure dropping funnel, and placed on a magnetic stirrer. 1.83g (10 mmol) of 3,4, 5-trimethoxyaniline was weighed, 5ml of 95% ethanol solution was added, the mixture was cooled to-10℃with glycerol at-20℃and the magnetic stirrer was turned on to stir and dissolve the mixture, and the reaction solution was bright yellow.
2ml of 37% concentrated hydrochloric acid is measured and added into the reaction liquid dropwise, and the cooling liquid is replaced periodically during the dripping period, so that the temperature of the reaction liquid is ensured to be lower than 0 ℃, and the reaction liquid is converted into beige turbid liquid.
1.39g (20 mmol) of sodium nitrite was weighed out and prepared as a 5ml aqueous solution, which was likewise cooled in a cooling liquid. After the hydrochloric acid is added dropwise, the sodium nitrite solution is added dropwise, and the temperature is also noted during the dropwise, so that the reaction temperature is always lower than 0 ℃. After the addition, the solution appeared orange-yellow. The reaction was continued with stirring for 1h while maintaining the reaction solution at a temperature below 0 ℃.
8g of sodium hydroxide was weighed and dissolved in 50ml of water to prepare a sodium hydroxide solution.
0.85g (9 mmol) of phenol was weighed, 5ml of sodium hydroxide solution prepared before addition was added, and after dissolution, it was cooled to-10℃with a cooling liquid.
And (3) dropwise adding the solution into diazotization reaction liquid, and regulating the pH value to be 7 finally after the dripping is finished. The cooling liquid was removed, and the reaction was stirred at room temperature for 2h. The mixture was separated, extracted with ethyl acetate and concentrated, and the product was an orange flaky solid. 1 H-NMR(CDCl 3 ,ppm):7.85(d,2H,Ph),7.22(s,2H,Ph),6.95(d,2H,Ph),3.95(s,6H,OCH3),3.93(s,3H,OCH3). 13 C-NMR(CDCl 3 ,ppm):158.6,153.4,148.7,146.8,140,124.9,115.9,100.1,61.1,56.2.MS(ESI)m/z:[M+H] + =288.
Example 2 Synthesis of Azo-F
The same procedure as in example 1 was followed using difluorophenol as starting material and the product was a tan solid. 1 H-NMR(CDCl 3 ,ppm):7.20(s,2H,Ph),6.55(d,2H,Ph),3.95(s,6H,OCH 3 ),3.94(s,3H,OCH 3 ). 13 C-NMR(CDCl 3 ,ppm):158.7,158.6,156.1,153.4,148.7,146.8,140,124.9,115.9,100.1,61.1,56.2.MS(ESI)m/z:[M+H] + =324.
EXAMPLE 3 Synthesis of Azo-Cl
The same procedure as in example 1 was followed using dichlorophenol as a starting material and the product was a black solid. 1 H-NMR(CDCl 3 ,ppm):7.12(2H,s,Ph),6.85(2H,s,Ph),3.88(9H,s,OCH 3 ). 13 C-NMR(CDCl 3 ,ppm):155.8,153.5,148.3,141.2,141.1,128.4,116.6,100.6,61.2,56.3.MS(ESI)m/z:[M+H] + =356.
EXAMPLE 4 Synthesis of Azo-H
The same procedure as in example 1 was followed using o-propargyloxyphenol as starting material, the product being a tan solid. 1 H-NMR(CDCl 3 ,ppm):7.20(s,2H,Ph),6.55(d,2H,Ph),3.95(s,6H,OCH3),3.94(s,3H,OCH3). 13 C-NMR(CDCl 3 ,ppm):158.7,158.6,156.1,153.4,149.2,140.4,100.5,61.1,56.2.
EXAMPLE 5 Synthesis of Azo-Me
The Azo-H prepared in example 4 was 0.55g, potassium carbonate 0.3g and an excess of methyl iodide were added to 2mL of DMF and the reaction stirred at room temperature overnight. After the reaction, water was added and extracted with ethyl acetate to give a brown yellow solid. 1 H-NMR(CDCl 3 ,ppm):7.66(1H,dd,Ph),7.63(1H,d,Ph),7.23(2H,s,Ph),7.02(1H,d,CH),4.87(2H,f,CH2),3.97(9H,s,3×OCH3),3.93(3H,s,OCH3).
EXAMPLE 6 Synthesis of Azo-Pr
Azo-Pr was obtained by substituting bromopropane for methyl iodide as described in example 5. MS (ESI) M/z: [ M+H ]] + =384.
EXAMPLE 7 Synthesis of Azo-Bu
The procedure as described in example 5 was followed using bromobutane instead of iodomethane to give a tan solid. MS (ESI) m/z:[M+H] + =398.
Example 8 preparation of Azo-Bu hydrochloride
46mg of Azo-Bu synthesized in example 7 was dissolved in acetone, and dry HCl gas was slowly introduced to supersaturation with stirring. White crystals are precipitated in an ice-water bath to obtain hydrochloride with a yield of 80 percent.
EXAMPLE 9 photoisomerization property study (taking the product of example 2 as an example)
1) Ultraviolet-visible absorption spectrum
Precisely weighing the product (Azo-F) of the example 2, adding the product into a 100mL volumetric flask, and adding ethanol to fix the volume; absorbing 2mL into another volumetric flask to prepare 0.002mol/L solution, testing under U3010 UV-Vis spectrophotometer (Hitachi, cell optical path is lcm, japan), and measuring ultraviolet-visible absorption spectrum at room temperature under 200-700nm with solvent as reference. Maximum absorption was found at 368 nm. The cuvette was placed under 368nm illumination and scanned every 15min to see its spectral appearance under illumination.
The results are shown in FIG. 1. It can be seen that the absorption intensity gradually becomes weaker with the increase of the irradiation time, whereas the case of the sub-absorption peak around 260nm is contrary, and the absorption intensity gradually becomes stronger with the increase of the irradiation time.
In the same way, azo-F was scanned once every 15min under the irradiation of 625nm (red light).
The results are shown in FIG. 2. It can be seen that as the irradiation time is prolonged, the absorption intensity thereof at 368nm becomes gradually strong, and the sub-absorption peak around 260nm becomes gradually weak.
It can be seen that the effect of light of different wavelengths on such azo compounds is different, which also illustrates to some extent that such azo compounds counteract photoisomerization.
2) Research on photoisomerization phenomenon before and after blue light irradiation
After the prepared Azo-F compound was irradiated with blue light (wavelength range 450 to 470 nm) for a predetermined period of time, changes in the peak pattern and the content before and after irradiation with light were measured by HPLC (Shimadzu A20 type).
HPLC spectra of Azo-F compounds without irradiation are shown in FIG. 3. The compound is mainly shown as two peaks in the solution, the peaks are 3.894min and 11.159min respectively, and the content is 41.48% and 50.86% respectively, which indicates that the cis-trans conformations exist in the solution and a certain equilibrium state is achieved.
The spectrum of the Azo-F compound after exposure to blue light for 20min is shown in FIG. 4. It can be seen that the peak at 3.816min was significantly reduced, the content was reduced to 15.75%; the other main peak around 11min has a content greatly increased to 78.62%. It is shown that after 20min of irradiation, the cis-trans conformations of the azobenzene compound are mutually converted, wherein the trans-conformations are more dominant.
The spectrum after continuing to irradiate for 100min is shown in fig. 5. It can be seen that the peak at 3.816min is significantly reduced and substantially vanished by 100min blue light irradiation; the other major peak around 11min had a significant increase in content, reaching 93.9%, indicating substantially complete conformation conversion of azobenzene.
EXAMPLE 10 photoisomerization performance study (taking the product of example 7 as an example)
7.96mg of the product of example 7 (Azo-Bu) was placed in a 10mL volumetric flask A, and the chromatographic methanol was dissolved and the volume was fixed. Another 100. Mu.L of the solution in flask A was placed in flask B of 10mL and the chromatographic methanol was used to determine the volume. After the preparation, 1mL of the solution in volumetric flask B was placed in 6 different EP tubes, respectively, and numbered. The prepared EP tube was placed in a thin layer chromatography scanner and irradiated with red light (620-630 nm) and one of the tubes was taken out at 15min,30min,1h,2h,4h,8h and placed in LC-MS to see the ratio change of cis-to trans-structure (cis-polar is large, early peak).
The wavelength was changed as above, and the sample was irradiated with yellow light (580-585 nm), green light (520-530 nm), blue light (450-470 nm), violet light (405 nm), and violet light 395nm for 15min,30min,1h,2h,4h,8h, respectively, and data were recorded.
Table 2: content of cis-trans isomer under different wavelength illumination (%), area normalization method
It can be seen that under the irradiation of four wavelengths of red light (620-630 nm), yellow light (580-585 nm), green light (520-530 nm) and blue light (450-470 nm), the interconversion of cis-and trans-structures of the two azobenzene compounds can not be obviously caused. While the purple light at 395nm and 405nm can obviously cause the structural interconversion of the two azobenzene compounds.
EXAMPLE 11 molecular docking
Molecular docking is a common and effective method for current computer-aided drug design, which guides us to screen compounds with good adaptability on the active site of proteins and reveals ideal binding patterns of small molecules. The CDOCKER program provides a refined docking approach for a multitude of ligands with a single target protein, and this grid-based docking approach utilizes the CHARMM force field, where the protein remains immobilized while allowing ligand to change conformation, with results obtained in terms of-CDOCKER energy and-CDOCKER interaction, with higher values for greater favorable binding between the protein and ligand. Before pharmacological activity detection, the synthesized small molecular compound is subjected to molecular docking with tubulin by using computer-aided drug design software Discovery studio 4.0 of Chuangteng company.
The docking results are shown in fig. 6. It can be seen that, in order of-CDocker_interaction_energy, azo-Bu and Azo-Pr each have a plurality of conformations scored higher than that of the small molecule CA-4 of the original protein (the name 5LYJ of its binding protein is shown), and the top-ranked conformations are consistent with the conformations of the small molecules in the original protein, and are cis structures, indicating that the designed compounds may have similar or stronger tubulin inhibition as CA-4.
Example 12 antiproliferative activity of small molecule compounds:
HeLa cells (as a model of human cervical adenocarcinoma) and H157 cells (a model of lung cancer) were maintained in DMEM/F12 medium supplemented with 10% FBS under standard cell culture conditions. 3,000 cells per well were seeded into two sets of 96-well plates and the cells were incubated three times with a series of dilutions of the individual compounds for four days. During four days of compound treatment, one group of cells grew in the dark and the other group of cells grew under pulsed ultraviolet light (390-400 nm, 10 seconds every 0.5 h). At the end of compound treatment, 3- (4, 5-dimethyltetrazol-2-yl) -2, 5-diphenyltetrazolium bromide (MTT) reagent was added to each well and incubated with cells for 1 hour at 37 ℃. Crystals formed in each 96 well were then dissolved using DMSO. Optical density values were measured at wavelengths 570nm and 630nm using BioTek Synergy H4 (BioTek Instruments, winooski, VT), and the difference between these two optical density values was used to analyze the relative cell viability in each 96 well. IC50 values were calculated by Graphpad Prism software using a sigmoidal dose response plot.
The results are shown in Table 3 below:
IC under dark/ultraviolet (395 nm) 50 Value (MTT method measurement)
a No detection was observed within the experimental test concentrations
It can be seen that the azo derivative of the CA4 shows weak cell proliferation inhibition effect on Hela\MCF-7 cells under dark conditions, and shows strong cell proliferation inhibition effect under illumination of specific wavelength, and the inhibition activity of the compound 8 with the strongest activity on the Hela cells is 7.5 times stronger than that of positive control CA 4.
In conclusion, the azo derivative of CA4 provided by the invention has unique photoisomerization property, and can exist in an inactive or less active trans-isomer (trans-) state in dark; under the irradiation of light with a certain wavelength, the inactive trans isomer can be quickly converted into the cis isomer (cis-) with biological activity, the polymerization of the targeted tubulin is inhibited, and the targeted single cell death can be realized. The azo derivative of CA4 can be transported in vivo in a nontoxic manner and activated at a target site, so that the targeting problem of a tubulin inhibitor is solved to a certain extent.

Claims (5)

1. An azo small molecular compound is characterized by having the following structure:
2. the pharmaceutically acceptable salt of the azo small molecule compound of claim 1, wherein the pharmaceutically acceptable salt is a hydrochloride, a hydrobromide, or a mixed salt thereof.
3. The use of an azo small molecule compound according to claim 1 for the preparation of a pharmaceutical formulation for the prevention or treatment of diseases caused by tubulin overexpression.
4. The use of an azo small molecule compound according to claim 2 in the preparation of a pharmaceutical formulation for the prevention or treatment of diseases caused by tubulin overexpression.
5. The use according to claim 3 or 4, wherein the condition caused by tubulin overexpression is breast cancer, leukemia, skin cancer, cervical cancer, esophageal cancer, lung cancer, glioma, alzheimer's disease, amyotrophic lateral sclerosis, cataract, parkinson's disease, creutzfeldt-jacob disease, huntington's disease, acute gout, joint pain, familial mediterranean fever or cirrhosis.
CN202111529543.9A 2021-12-14 2021-12-14 Azo microtubulin inhibitor and preparation method and application thereof Active CN114349657B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111529543.9A CN114349657B (en) 2021-12-14 2021-12-14 Azo microtubulin inhibitor and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111529543.9A CN114349657B (en) 2021-12-14 2021-12-14 Azo microtubulin inhibitor and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN114349657A CN114349657A (en) 2022-04-15
CN114349657B true CN114349657B (en) 2023-12-22

Family

ID=81099640

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111529543.9A Active CN114349657B (en) 2021-12-14 2021-12-14 Azo microtubulin inhibitor and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN114349657B (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109574871A (en) * 2018-11-23 2019-04-05 上海交通大学 A kind of acetylamino azobenzene derivative and its preparation and application

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170051149A1 (en) * 2014-04-29 2017-02-23 Ecole Normale Superieure De Lyon Azoaryls as reversibly modulatable tubulin inhibitors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109574871A (en) * 2018-11-23 2019-04-05 上海交通大学 A kind of acetylamino azobenzene derivative and its preparation and application

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Borowiak M 等.Photoswitchable Inhibitors of Microtubule Dynamics Optically Control Mitosis and Cell Death.《CELL》.2015,第162卷(第2期),第404页图1,第S6-S7页,第406页左栏倒数第1段,第404页右栏第3段. *
Photoswitchable Inhibitors of Microtubule Dynamics Optically Control Mitosis and Cell Death;Borowiak M 等;《CELL》;第162卷(第2期);第404页图1,第S6-S7页,第406页左栏倒数第1段,第404页右栏第3段 *

Also Published As

Publication number Publication date
CN114349657A (en) 2022-04-15

Similar Documents

Publication Publication Date Title
Zhu et al. Design, synthesis, and evaluation of chalcone analogues incorporate α, β-Unsaturated ketone functionality as anti-lung cancer agents via evoking ROS to induce pyroptosis
Ferte et al. Flavonoid-related modulators of multidrug resistance: synthesis, pharmacological activity, and structure− activity relationships
Kumbhare et al. Synthesis and biological evaluation of novel Mannich bases of 2-arylimidazo [2, 1-b] benzothiazoles as potential anti-cancer agents
Mahboobi et al. Synthetic 2-aroylindole derivatives as a new class of potent tubulin-inhibitory, antimitotic agents
AU2012250497C1 (en) Prenylated hydroxystilbenes
Choi et al. Design and synthesis of 3, 4-dihydro-2H-benzo [h] chromene derivatives as potential NF-κB inhibitors
Attalah et al. Ethyl benzoate bearing pyrrolizine/indolizine moieties: Design, synthesis and biological evaluation of anti-inflammatory and cytotoxic activities
US20170051149A1 (en) Azoaryls as reversibly modulatable tubulin inhibitors
Hayun et al. Synthesis, anti-inflammatory and antioxidant activity of mannich bases of dehydrozingerone derivatives
Iqbal et al. Design, synthesis, and biological evaluation of imidazopyridine‐linked thiazolidinone as potential anticancer agents
Ramalingam et al. Synthesis and biological evaluation of calothrixins B and their deoxygenated analogs
Prinsloo et al. Synthesis and in vitro antileishmanial efficacy of novel quinazolinone derivatives
CN114349657B (en) Azo microtubulin inhibitor and preparation method and application thereof
Elgaafary et al. Synthesis of β-enaminonitrile-linked 8-methoxy-1H-benzo [f] chromene moieties and analysis of their antitumor mechanisms
Wani et al. Anticancer activity of a novel quinazolinone-chalcone derivative through cell cycle arrest in pancreatic cancer cell line
Venkatesh et al. Design and synthesis of novel sulphamide tethered quinazolinone hybrids as potential antitumor agents
Dai et al. Sophoridine Derivatives Induce Apoptosis and Autophagy to Suppress the Growth of Triple‐Negative Breast Cancer through Inhibition of mTOR Signaling
KR20050096954A (en) Breast cancer resistance protein(bcrp) inhibitor
Özdemir et al. Synthesis and evaluation of a new series of arylidene indanones as potential anticancer agents
Tozkoparan et al. Novel 3, 6‐Disubstituted 7H‐1, 2, 4‐Triazolo [3, 4‐b][1, 3, 4] thiadiazines: Synthesis, Characterization, and Evaluation of Analgesic/Anti‐inflammatory, Antioxidant Activities
CN111094262B (en) 1, 3-dioxane-4, 6-diketone compound, preparation method thereof, pharmaceutical composition and application thereof
US20150051255A1 (en) Glucose transporter inhibitors
Patel et al. Sulfonylpiperazines based on a flavone as antioxidant and cytotoxic agents
CN114349817A (en) VHL ligand-based tubulin degrading agent and application thereof
CN110526854B (en) Alpha, beta-unsaturated ketone derivative, preparation method and application thereof as medicine

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