CN114349739A - Azo compound for degrading tubulin and synthesis method and application thereof - Google Patents

Azo compound for degrading tubulin and synthesis method and application thereof Download PDF

Info

Publication number
CN114349739A
CN114349739A CN202111528045.2A CN202111528045A CN114349739A CN 114349739 A CN114349739 A CN 114349739A CN 202111528045 A CN202111528045 A CN 202111528045A CN 114349739 A CN114349739 A CN 114349739A
Authority
CN
China
Prior art keywords
tubulin
azo
azo compound
degrading
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.)
Pending
Application number
CN202111528045.2A
Other languages
Chinese (zh)
Inventor
王尊元
马臻
黄文海
沈正荣
梁美好
章迟啸
曾申昕
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 CN202111528045.2A priority Critical patent/CN114349739A/en
Publication of CN114349739A publication Critical patent/CN114349739A/en
Pending legal-status Critical Current

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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/12Ophthalmic agents for cataracts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Abstract

The invention discloses an azo compound for degrading tubulin, a synthetic method and application thereof. The method takes azo micromolecules with tubulin inhibition as raw materials, and azo compounds for degrading the tubulin are obtained through further reaction. The preparation method of the azo compound for degrading the tubulin has the characteristics of low cost, simple method operation, mild conditions and the like. The azo compound for degrading 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 specific photoisomerization performance, and further promotes the degradation of the tubulin, so that the azo compound can be applied to the preparation of medicaments for preventing or treating diseases caused by excessive increase of the tubulin.

Description

Azo compound for degrading tubulin and synthesis method and application thereof
Technical Field
The invention relates to an azo compound, in particular to an azo compound (based on a CRBN ligand) capable of degrading tubulin, a pharmaceutically usable salt thereof, a synthetic method thereof and application thereof in a medicament for selectively degrading the tubulin.
Background
Microtubules (microtubule) are dimers formed by two types of tubulin, alpha and beta, and are distributed in the form of networks or bundles in cells, and are involved in the formation of cytoskeleton, maintenance of cell morphology, cell contraction, transport of intracellular substances, 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, mitosis can not be performed or stopped, the interruption of the cell mitosis process can have greater influence on cells, so that the growth of the cells is inhibited, and the occurrence of apoptosis is finally induced, thereby achieving the purpose of inhibiting the proliferation and the growth of the cells. Since cancer cells differ most from normal cells in that their mitosis is frequent and uncontrolled, blocking tubulin synthesis is of particular importance in the treatment of malignant tumors.
Combretastatin (Combretastatin a4, CA4) was obtained from the south african shrub african windmill (combretam cafrum). The cis-stilbene structure of CA4 can target tubulin in body, inhibit its polymerization and further destroy tumor blood vessel, so that it has the function of resisting tumor. CA4 is effective in various tumor models and is a promising antitumor drug, however, CA4 has extremely poor water solubility, only 11.8 μ g/ml, so that the oral bioavailability is low; meanwhile, the clinical application value of the stilbene derivative is limited due to the characteristic that the stilbene derivative parent nucleus is easy to be destructed into trans form to cause activity disappearance. In order to overcome the defects of CA4, researchers have conducted extensive structure-activity relationship research on CA4 and developed a water-soluble phosphate prodrug CA4P, which is currently in phase III clinical trials.
However, the tubulin inhibitors including CA4 are currently non-specific, their biological activity cannot be localized to specific tumor sites, and are toxic to normal and healthy cells. This non-specificity can cause severe systemic side effects in the treatment of cancer, such as cardiotoxicity and neurotoxicity, which greatly limits the dosage and route of administration of such inhibitors; at the same time, the development of acquired resistance greatly diminishes their clinical therapeutic value.
In order to enhance the targeting property, the azobenzene optical switch is a good modification approach in view of the stilbene core structure of CA4 and the activity change of cis-trans isomers thereof. The azobenzene compound contains a special conjugated pi chemical structure, so that the azobenzene compound has photoisomerization performance, namely: the trans-isomer and the cis-isomer can be mutually converted after the light irradiation of a special wave band. In 2015, the international journal of apex "Cell" reports: borowjak et al synthesized several azotized derivatives of CA4, PSTs, using azobenzene in place of the stilbene fragment in CA4 (i.e., C-N-bond) (Borowjak et al 2015.Cell 162, 403-. The research finds that: because azo compounds have unique photoisomerization performance, PSTs exist in an inactive trans isomer (trans-) state in the dark or under normal daily irradiation; under the irradiation of light with certain wavelength, the inactive trans isomer can be quickly converted into cis isomer (cis-), which has biological activity, targets tubulin, inhibits the polymerization of the tubulin, and can realize targeted single cell death.
The azo derivative of CA4 can be transported in vivo without toxicity and activated at a target site, thereby solving the targeting problem of the tubulin inhibitor to a certain extent. However, the drawback of acquired resistance inherent in tubulin inhibitors has not been addressed because they still inhibit tubulin to exert a pharmacological effect, require a certain dose to bind and occupy the active site of tubulin for a long period of time to inhibit its activity.
Disclosure of Invention
The invention aims to overcome the defects of the prior art and provides an azo compound capable of degrading tubulin, a pharmaceutically usable salt thereof, a synthesis method thereof and application thereof in a medicament for selectively degrading tubulin.
The purpose of the invention is realized by the following technical scheme: an azo compound for degrading tubulin, which has the following structure:
Figure BDA0003409694490000021
wherein R1 is selected from hydrogen, acetyl, propionyl, butyryl, benzoyl, substituted benzoyl, benzyl, substituted benzyl, phenyl, substituted phenyl, methyl, ethyl, propyl, butyl, trifluoromethyl;
x, Y, W is independently oxygen, or nitrogen;
n is selected from an integer between 0 and 16.
More specifically, the azo compound is selected from the following structures:
r1 is selected from hydrogen, methyl, ethyl, propyl, butyl, pentyl, hexyl, trifluoromethyl;
x, Y, W is independently oxygen, sulfur or nitrogen;
n is selected from an integer between 1 and 12.
More specifically, the azo compound is selected from the following structures:
r1 is selected from hydrogen, methyl, butyl, pentyl, hexyl;
x, Y, W is independently oxygen, or nitrogen;
n is selected from an integer between 1 and 12.
The azo compound for degrading tubulin can be used alone or 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, taurate, citrate, succinate or mixed salts thereof.
The synthesis method of the azo compound is characterized by comprising the following steps:
1) reacting bromo acid with lenalidomide in a solvent in the presence of anhydrous potassium carbonate to obtain a white solid bromide;
2) reacting white solid bromide with sodium azide in a solvent to obtain azide, carrying out Click reaction on the azide and an azo compound in the presence of catalytic amount of Cu salt, and separating to obtain the target product with the structure shown in the formula I.
The invention also provides a preparation method of the azo compound for degrading the tubulin, which comprises the following steps: the LigandA a and LigandB compounds were first prepared or purchased according to literature methods and attached to linker using different chemistries depending on the substituents R1, R2. Examples are as follows:
Figure BDA0003409694490000031
the reaction steps are as follows:
the commercial bromoacid reacts with the commercial lenalidomide in a solvent (DMF) in the presence of anhydrous potassium carbonate to obtain white solid bromide. The bromide reacts with sodium azide in a solvent (DMF) to obtain the azide. Carrying out Click reaction on the azide and a micromolecular azo compound in the presence of catalytic amount of Cu salt, and separating to obtain a target product.
The invention also provides an application of the azo compound and the pharmaceutically acceptable salt thereof in pharmacy, which comprises the following specific steps: the pharmaceutical preparation is used for preparing a pharmaceutical preparation for preventing or treating diseases caused by over-expression of tubulin, wherein the diseases comprise breast cancer, leukemia, skin cancer, cervical cancer, esophageal cancer, lung cancer, glioma and the like, and also comprise acute gout, arthralgia, familial mediterranean fever and liver cirrhosis.
Compared with the prior art, the invention has the following beneficial effects:
based on the rapid development of ProTAC (Proteolysis-targeting chimaaras, a proteolytic targeting chimera), the invention solves the problem of acquired drug resistance inherent in small molecule inhibitors in a protein degradation mode: the azo compound and the pharmaceutically available salt thereof provided by the invention do not need to occupy active sites for a long time like an inhibitor, but only need to be combined with tubulin for a short time to form a ternary complex to induce the protein degradation effect, and the function can be restored only by re-synthesis after the protein is degraded, so that the generation of drug resistance is greatly delayed; it is also effective for those proteins that have developed resistance to drugs. Therefore, the method can be applied to the preparation of medicaments for preventing or treating diseases caused by over-expression of tubulin.
Description of the drawings:
FIG. 1 is a HRMS diagram of the target product HAzo-C11-DA synthesized in example 5;
FIG. 2 is a graph showing the degradation effect of the target product HAzo-C11-DA protein.
Detailed Description
The structure, preparation method and application in preparing pharmaceutical preparations for preventing or treating diseases caused by over-expression of tubulin will be further described in the following without limiting the invention.
Analytical data for the samples were determined by the following instruments:
the thermometer is not corrected; bruker DRX400 nmr; agilent model 5975 mass spectrometer; bruker Vector22 infrared spectrometer.
EXAMPLE 1 Synthesis of Azo-H
Figure BDA0003409694490000041
A100 ml three-necked flask is selected, a magnetic rotor is added, a thermometer and a constant pressure dropping funnel are arranged on the magnetic stirrer. Weighing 1.83g (10mmol) of 3,4, 5-trimethoxyaniline, adding 5ml of 95% ethanol solution, cooling to-10 ℃ with glycerol at-20 ℃, opening a magnetic stirrer, and stirring for dissolving, wherein the reaction solution is bright yellow.
Measuring 2ml of 37% concentrated hydrochloric acid, dropwise adding into the reaction solution, and periodically replacing a cooling liquid during the dropwise adding period to ensure that the temperature of the reaction solution is lower than 0 ℃ and the reaction solution is changed into beige turbid liquid.
Sodium nitrite 1.39g (20mmol) was weighed out to prepare a 5ml aqueous solution, which was similarly cooled in a cooling liquid. After the hydrochloric acid is dropwise added, dropwise adding a sodium nitrite solution, and paying attention to the temperature during the dropwise adding process to ensure that the reaction temperature is always lower than 0 ℃. After the addition, the solution appeared orange-yellow. The reaction was stirred for 1h while maintaining the temperature below 0 ℃.
8g of sodium hydroxide is weighed and dissolved in 50ml of water to prepare a sodium hydroxide solution.
O-propargyloxyphenol (9mmol) was weighed, 5ml of the previously prepared sodium hydroxide solution was added, and after dissolution, the solution was cooled to-10 ℃.
Dropwise adding the solution into the diazotization reaction solution, and adjusting the pH value to 7 after the dropwise adding is finished. The cooling liquid was removed and the reaction was stirred at room temperature for 2 h. Separating, extracting with ethyl acetate, and concentrating to obtain a product as a brown yellow solid.1H-NMR(CDCl3,ppm):7.20(s,2H,Ph),6.55(d,2H,Ph),3.95(s,6H,OCH3),3.94(s,3H,OCH3).13C-NMR(CDCl3,ppm):158.7,158.6,156.1,153.4,149.2,140.4,100.5,61.1,56.2.
EXAMPLE 2 Synthesis of Azo-Me
Figure BDA0003409694490000051
0.55g of Azo-H prepared in example 1, 0.3g of potassium carbonate and excess methyl iodide were added to 2mL of DMF, and the reaction was stirred at room temperature of 25 ℃ overnight. After the reaction, water was added and extracted with ethyl acetate to obtain a tan solid.1H-NMR(CDCl3,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 3 Synthesis of Azo-Pr
Figure BDA0003409694490000052
Azo-Pr was obtained as described in example 2, substituting methyl iodide with propyl bromide. MS (ESI) M/z [ M + H ]]+=384.
Example 4 Synthesis of Azo-Bu
Figure BDA0003409694490000053
The procedure as described in example 2 was followed, substituting methyl iodide with butyl bromide to give a tan solid.1HNMR(400MHz,CDCl3)δ7.56(td,J=4.6,2.2Hz,1H),7.15(s,1H),6.94(d,J=9.2Hz,1H),4.78(d,J=2.4Hz,1H),4.05(t,J=6.6Hz,1H),3.90(s,3H),3.86(s,1H),3.50(t,J=6.7Hz,1H),2.47(t,J=2.4Hz,1H),1.89–1.73(m,2H),1.50(s,2H).MS(ESI)m/z:[M+H]+=398.
Example 5 Synthesis of HAzo-C11-DA
Figure BDA0003409694490000061
Commercially available bromoundecanoic acid (1.3 g) was dissolved in DMF, and thionyl chloride (2 mL) was added thereto, followed by reaction at 90 ℃ for 2 hours. After removing excess SOCl2 under reduced pressure, 1g of commercially available lenalidomide and 2g of anhydrous potassium carbonate were added at room temperature, and the mixture was reacted at 60 ℃ overnight. After the reaction is finished, white solid bromide is obtained after extraction and concentration and is directly used for the next reaction.
105mg of bromide was weighed out and dissolved in 2mL of DMF, and 40mg of sodium azide was added thereto to react at 60 ℃ overnight. After the reaction, a large amount of water is added, and white solid azide is obtained after centrifugation.1H NMR(400MHz,CDCl 3)δ(ppm)8.59(1H,s,NH),7.88(1H,s,NH),7.69(2H,t,Ph),7.46(1H,t,Ph),5.10(1H,d,CH),4.36(2H,s,CH2),3.26(2H,t,CH2),2.78(2H,m,CH2),2.41(2H,t,CH2),2.20(2H,dd,CH2),1.71(2H,t,CH2),1.59(2H,t,CH2),1.28-1.35(12H,m,CH2).
50mg of an azide and 50mg of an Azo-H compound obtained in example 1 and a catalytic amount of CuBr (about 2mg) were weighed out and placed in a reaction flask, and after 2mL of DMSO was added and dissolved, a small amount of water was added and the reaction was carried out at room temperature and 25 ℃ for 24 hours with stirring. After the reaction is finished, water is added, EA is used for extraction, column chromatography separation is carried out, and white solid HAzo-C11-DA is obtained after concentration. HRMS: found [ M + H]+811.3749, Calc C42H51N8O9 811.3779. The HRMS spectrum of the product is shown in figure 1.
Example 6 Synthesis of HAzo-C6-DA
Figure BDA0003409694490000062
By the same procedure as in example 5, commercially available bromohexanoic acid was used in place of bromoundecanoic acid, and separation was carried out by column chromatography to obtain HAzo-C6-DA as a white solid. MS: [ M + H ]]+=740。
Example 7 Synthesis of HAzo-C8-DA
Figure BDA0003409694490000071
In the same manner as in example 5, commercially availableAnd (3) replacing bromoundecanoic acid with bromooctanoic acid, and performing column separation to obtain white solid HAzo-C8-DA. MS: [ M + H ]]+=768。
Example 8 preparation of HAzo-C11-DA hydrochloride
46mg of HAzo-C11-DA synthesized according to claim 5 was dissolved in acetone, and dried HCl gas was slowly introduced into the mixture while stirring until supersaturated. White crystals were precipitated in an ice-water bath as the hydrochloride salt in a yield of 80%.
EXAMPLE 9 study of the Photoisomerizing Properties of Small molecule Compounds (example 4)
7.96mg of the target product is placed in a 10mL volumetric flask A, and the chromatographic methanol is dissolved to fix the volume. Another 100. mu.L of the solution in the volumetric flask A is placed in a 10mL volumetric flask B, and the volume is determined by the chromatographic methanol. After the preparation, 1mL of the solution in the volumetric flask B is taken and placed in 6 different EP tubes respectively, and the solutions are numbered. The prepared EP tube is placed in a thin layer chromatography scanner and irradiated by red light (620-630nm), one of the tubes is taken out at 15min, 30min, 1h, 2h, 4h and 8h and is placed in an LC-MS to check the change of the ratio of cis-structure to trans-structure (the cis-polarity is large, the peak emergence is early).
The operation is the same as above, the wavelength is changed, yellow light (580-. The results are shown in Table 1.
TABLE 1 cis-trans isomer content (%, area normalization) under different wavelength illumination
Figure BDA0003409694490000081
As can be seen, under the irradiation of four wavelengths of red light (620-630nm), yellow light (580-585nm), green light (520-530nm) and blue light (450-470nm), the interconversion between the cis-structure and the trans-structure of the two azobenzene compounds can not be obviously caused. While the purple light of 395nm and 405nm can obviously cause the structural interconversion of the two azobenzene compounds.
Example 10 antiproliferative activity of small molecule compounds:
HeLa cells (as a model of human cervical adenocarcinoma) and MCF-7 cells (a model of breast 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 for four days in triplicate with a series of individual compound dilutions. During four days of compound treatment, one group of cells was grown in the dark, while the other group was grown under pulsed UV light (390-400nm, 10 seconds per 0.5 h). At the end of compound treatment, 3- (4, 5-dimethylthiazol-2-yl) -2, 5-diphenyltetrazolium bromide (MTT) reagent was added to each well and incubated with cells for 1 hour at 37 ℃. The crystals formed in each 96 well were then dissolved using DMSO. Optical density values were measured at wavelengths of 570nm and 630nm using a BioTek Synergy H4(BioTek Instruments, Winooski, VT) and the difference between the two optical density values was used to analyze relative cell viability in each 96 well. IC (integrated circuit)50Values were calculated by Graphpad Prism software using a sigmoidal dose response plot.
The results are shown in table 2 below:
TABLE 2 IC in darkness/UV (395nm)50Value (MTT method determination)
Figure BDA0003409694490000091
aNot detected within the experimental test concentration
As can be seen, the azo derivative of CA4 shows weaker cell proliferation inhibition effect on Hela \ MCF-7 cells under dark conditions, but shows stronger cell proliferation inhibition effect under the illumination of specific wavelength, and the inhibition activity of the compound 8 with the strongest activity on Hela cells is 7.5 times stronger than that of the positive control CA 4.
Example 11 degradation of tubulin (exemplified by the product of example 5):
MCF-7 cell lines were given different concentrations of HAzo-C11-DA and exposed to light at a wavelength of 390-400nm for 10 seconds every 0.5h for a total of 4 h. The cells were collected, lysed on ice for 30min after adding a lysate (50mM NaCl,5mM EDTA, 0.5% SDS, 0.1mM sodium orthovanadate, 50. mu.g/ml aprotinin, 1mM phenylthiononyl fluoride, and 10mM Tris-HCl; pH 7.4), centrifuged at 12000rpm for 15 minutes at 4 ℃, and the supernatant protein was quantitated and subjected to SDS-PAGE; after electrophoresis, transferring the sample to a nitrocellulose membrane, then carrying out immunoreaction, sequentially carrying out room temperature sealing for 1 hour, incubating overnight at 4 ℃ primary antibody, incubating for 2 hours at room temperature secondary antibody, washing the membrane for 2 hours by TBST, adding ECL solution, and carrying out accurate warm bath for 1 minute; draining the film, exposing in a dark box for several minutes, developing and fixing; the X-ray film was scanned by a scanner and then the grayscale values of the bands were read by MetaMorph software, and statistical analysis was performed with GAPDH as an internal reference.
The results are shown in FIG. 2.
It can be seen that the product HAzo-C11-DA of example 5 of the present invention showed a slight degradation effect on tubulin at various concentrations, and the lack of activity may be related to the failure to obtain a suitable cleavage site and also to the length of the linker chain. Since the compounds are designed based on the ProTAC principle, other compounds with different chain lengths can also show the degradation activity on tubulin.
The azo-based compound degrading tubulin according to the present invention shows no pharmacological activity in dark environment, but shows affinity to tubulin under light of a specific wavelength due to its characteristic photoisomerization property, and further promotes tubulin degradation, which may be applied in the preparation of a drug for preventing or treating diseases caused by excessive increase of tubulin, including tumors such as breast cancer, leukemia, skin cancer, cervical cancer, esophageal cancer, lung cancer, glioma, etc., and also including neurodegenerative diseases such as alzheimer's disease, amyotrophic lateral sclerosis, cataract, parkinson's disease, creutzfeldt-jakob disease, huntington's disease, and also including other diseases such as acute gout, joint pain, familial mediterranean fever, liver cirrhosis, etc.

Claims (8)

1. An azo compound for degrading tubulin, which is characterized by having a structure shown in formula I:
Figure FDA0003409694480000011
wherein R is1Selected from the group consisting of hydrogen, acetyl, propionyl, butyryl, benzoyl, substituted benzoyl, benzyl, substituted benzyl, phenyl, substituted phenyl, methyl, ethyl, propyl, butyl, pentyl, hexyl, trifluoromethyl;
x, Y, W is independently oxygen, sulfur or nitrogen;
n is selected from an integer between 0 and 16.
2. The azo-based compound of claim 1, wherein R is1Selected from hydrogen, methyl, ethyl, propyl, butyl, pentyl, hexyl, trifluoromethyl;
x, Y, W is independently oxygen, sulfur or nitrogen;
n is selected from an integer between 1 and 12.
3. The azo-based compound of claim 1, wherein R is1Selected from hydrogen, methyl, butyl, pentyl, hexyl;
x, Y, W is independently oxygen, or nitrogen;
n is selected from an integer between 1 and 12.
4. The pharmaceutically acceptable salt of the azo-based compound of any one of claims 1 to 3, wherein the pharmaceutically acceptable salt is a hydrochloride, hydrobromide, hydroiodide, sulfate, bisulfate, phosphate, acetate, propionate, butyrate, oxalate, tartrate, methanesulfonate, p-toluenesulfonate, fumarate, taurate, citrate, succinate, or a mixed salt thereof.
5. A method for synthesizing an azo compound according to any one of claims 1 to 3, comprising the steps of:
1) reacting bromo acid with lenalidomide in a solvent in the presence of anhydrous potassium carbonate to obtain a white solid bromide;
2) reacting white solid bromide with sodium azide in a solvent to obtain azide, carrying out Click reaction on the azide and an azo compound in the presence of catalytic amount of Cu salt, and separating to obtain the target product with the structure shown in the formula I.
6. Use of an azo compound according to any one of claims 1 to 3 for the preparation of a pharmaceutical preparation for the prevention or treatment of a disease caused by overexpression of tubulin.
7. Use of the azo compound of claim 4 in the preparation of a pharmaceutical formulation for the prevention or treatment of a disease caused by overexpression of tubulin.
8. The use according to claim 6 or 7, wherein the disease caused by overexpression of tubulin is breast cancer, leukemia, skin cancer, cervical cancer, esophageal cancer, lung cancer, glioma, neurodegenerative diseases such as Alzheimer's disease, amyotrophic lateral sclerosis, cataract, Parkinson's disease, Creutzfeldt-Jakob disease, Huntington's disease, acute gout, joint pain, familial Mediterranean fever or liver cirrhosis.
CN202111528045.2A 2021-12-14 2021-12-14 Azo compound for degrading tubulin and synthesis method and application thereof Pending CN114349739A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111528045.2A CN114349739A (en) 2021-12-14 2021-12-14 Azo compound for degrading tubulin and synthesis method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111528045.2A CN114349739A (en) 2021-12-14 2021-12-14 Azo compound for degrading tubulin and synthesis method and application thereof

Publications (1)

Publication Number Publication Date
CN114349739A true CN114349739A (en) 2022-04-15

Family

ID=81098915

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111528045.2A Pending CN114349739A (en) 2021-12-14 2021-12-14 Azo compound for degrading tubulin and synthesis method and application thereof

Country Status (1)

Country Link
CN (1) CN114349739A (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015166295A1 (en) * 2014-04-29 2015-11-05 Ecole Normale Superieure De Lyon Azoaryls as reversibly modulatable tubulin inhibitors
CN109761970A (en) * 2019-01-26 2019-05-17 中国药科大学 A kind of targeting proteins degradation chimeric molecule, preparation method and application
CN110753693A (en) * 2016-12-23 2020-02-04 阿尔维纳斯运营股份有限公司 EGFR proteolytic targeting chimeric molecules and related methods of use
CN112480081A (en) * 2020-12-07 2021-03-12 沈阳药科大学 Difunctional molecular compound for inducing SHP2 protein degradation based on Cereblan ligand
CN112552293A (en) * 2019-09-25 2021-03-26 珠海宇繁生物科技有限责任公司 PROTAC small molecular compound and application thereof
CN113307793A (en) * 2020-08-27 2021-08-27 杭州医学院 Compound for inducing Tau protein degradation based on CRBN ligand, preparation method, pharmaceutical composition and application thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015166295A1 (en) * 2014-04-29 2015-11-05 Ecole Normale Superieure De Lyon Azoaryls as reversibly modulatable tubulin inhibitors
CN110753693A (en) * 2016-12-23 2020-02-04 阿尔维纳斯运营股份有限公司 EGFR proteolytic targeting chimeric molecules and related methods of use
CN109761970A (en) * 2019-01-26 2019-05-17 中国药科大学 A kind of targeting proteins degradation chimeric molecule, preparation method and application
CN112552293A (en) * 2019-09-25 2021-03-26 珠海宇繁生物科技有限责任公司 PROTAC small molecular compound and application thereof
CN113307793A (en) * 2020-08-27 2021-08-27 杭州医学院 Compound for inducing Tau protein degradation based on CRBN ligand, preparation method, pharmaceutical composition and application thereof
CN112480081A (en) * 2020-12-07 2021-03-12 沈阳药科大学 Difunctional molecular compound for inducing SHP2 protein degradation based on Cereblan ligand

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BIAN, JINLEI; REN等: "Discovery of Wogonin-based PROTACs against CDK9 and capable of achieving antitumor activity", BIOORGANIC CHEMISTRY *

Similar Documents

Publication Publication Date Title
Ghate et al. Synthesis of vanillin ethers from 4-(bromomethyl) coumarins as anti-inflammatory agents
Bhandari et al. Design, synthesis and evaluation of antiinflammatory, analgesic and ulcerogenicity studies of novel S-substituted phenacyl-1, 3, 4-oxadiazole-2-thiol and Schiff bases of diclofenac acid as nonulcerogenic derivatives
Jadhav et al. Synthesis and anti-inflammatory evaluation of methylene bridged benzofuranyl imidazo [2, 1-b][1, 3, 4] thiadiazoles
Bano et al. Synthesis and biological evaluation of some new 2-pyrazolines bearing benzene sulfonamide moiety as potential anti-inflammatory and anti-cancer agents
US4719047A (en) Anthracene derivatives
JP2007509842A (en) Compounds containing nitric oxide moieties for inducing the expression of ApoA1 for the treatment of cardiovascular disorders
Aytaç et al. Synthesis of 3, 6-disubstituted 7H-1, 2, 4-triazolo [3, 4-b]-1, 3, 4-thiadiazines as novel analgesic/anti-inflammatory compounds
US4511582A (en) Phenanthrene derivatives
Yang et al. Structure-activity relationship studies of phenothiazine derivatives as a new class of ferroptosis inhibitors together with the therapeutic effect in an ischemic stroke model
ES2909403T3 (en) Prenylated hydroxystilbenes
US4551282A (en) Triphenylene derivatives
Bhole et al. Synthesis and antitumor activity of (4-hydroxyphenyl)[5-substituted alkyl/aryl)-2-thioxo-1, 3, 4-thiadiazol-3-yl] methanone and [(3, 4-disubstituted)-1, 3-thiazol-2ylidene]-4-hydroxybenzohydrazide
Choi et al. Design and synthesis of 3, 4-dihydro-2H-benzo [h] chromene derivatives as potential NF-κB inhibitors
Liu et al. Synthesis and biological evaluation of novel 5, 6, 7-trimethoxy flavonoid salicylate derivatives as potential anti-tumor agents
Li et al. O2-(2, 4-dinitrophenyl) diazeniumdiolates derivatives: design, synthesis, cytotoxic evaluation and reversing MDR in MCF-7/ADR cells
EP3252039B1 (en) Compound containing indoleacetic acid core structure and use thereof
CN114349739A (en) Azo compound for degrading tubulin and synthesis method and application thereof
Vergara et al. Synthesis, antiprotozoal activity and cytotoxicity in U-937 macrophages of triclosan–hydrazone hybrids
HUT54617A (en) Process for producing new substituted acyl-phenols and pharmaceutical compositions containing them
Tomita et al. Determination of endogenous melatonin in the individual pineal glands of inbred mice using precolumn oxidation reversed-phase micro-high-performance liquid chromatography
CN114349817A (en) VHL ligand-based tubulin degrading agent and application thereof
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
KR20050096954A (en) Breast cancer resistance protein(bcrp) inhibitor
CN114349657B (en) Azo microtubulin inhibitor and preparation method and application thereof
Salman et al. Studies in antifertility agents. 50. Stereoselective binding of d-and l-centchromans to estrogen receptors and their antifertility activity

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