CN111518065A - Parthenolide derivative and preparation method and application thereof - Google Patents

Parthenolide derivative and preparation method and application thereof Download PDF

Info

Publication number
CN111518065A
CN111518065A CN202010525365.1A CN202010525365A CN111518065A CN 111518065 A CN111518065 A CN 111518065A CN 202010525365 A CN202010525365 A CN 202010525365A CN 111518065 A CN111518065 A CN 111518065A
Authority
CN
China
Prior art keywords
compound
dmapt
glioblastoma
formula
parthenolide
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.)
Withdrawn
Application number
CN202010525365.1A
Other languages
Chinese (zh)
Inventor
杨东林
张亚军
李勇
秦红霞
何刘军
黄玖红
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.)
Chongqing University of Arts and Sciences
Original Assignee
Chongqing University of Arts and Sciences
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 Chongqing University of Arts and Sciences filed Critical Chongqing University of Arts and Sciences
Priority to CN202010525365.1A priority Critical patent/CN111518065A/en
Publication of CN111518065A publication Critical patent/CN111518065A/en
Withdrawn legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/77Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D307/93Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems condensed with a ring other than six-membered
    • 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
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/06Peri-condensed systems

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (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 invention relates to the technical field of organic synthesis, in particular to a parthenolide derivative and a preparation method and application thereof. The invention takes the feverfew as the initial main raw material, and synthesizes 11 feverfew lactone derivatives through a series of chemical reactions, and the derivatives have certain antiproliferative activity on glioblastoma cells. The invention also proves that the derivative DMAPT-D6 containing deuterium can remarkably induce the accumulation of active oxygen of glioblastoma cells, thereby causing DNA damage in the glioblastoma cells. In addition, DMAPT-D6 promoted caspase-dependent death receptor-mediated exogenous apoptosis, suggesting that DNA damage induced by DMAPT-D6 could induce glioblastoma cell apoptosis. The two are combined, and the ROS accumulation caused by the treatment of DMAPT-D6 causes DNA damage and then causes death receptor mediated apoptosis, which shows that the DMAPT-D6 with the novel component has the treatment potential for treating glioblastoma and can be applied to the preparation of drugs for treating glioblastoma.

Description

Parthenolide derivative and preparation method and application thereof
Technical Field
The invention relates to the technical field of organic synthesis, in particular to a parthenolide derivative and a preparation method and application thereof.
Background
Reactive Oxygen Species (ROS) are a class of short half-life, highly Reactive, oxygen-containing, aerobic metabolic byproducts, including superoxide, hydroxyl radicals, and hydroxide. Cellular reactive oxygen species are produced intracellularly mainly by mitochondria, NADPH oxidase, peroxisomes and endoplasmic reticulum. New data indicate that reactive oxygen species act as double-edged swords in cells. Low levels of ROS are necessary for cell survival and proliferation; in contrast, excess ROS can cause oxidative stress, resulting in DNA damage, apoptosis, and necrosis. DNA damage is caused by chemical and physical factors that can lead to a complex series of processes including cell cycle arrest, DNA repair, modulation of cellular checkpoints, initiation of apoptosis, etc. For this reason, increasing cellular ROS production to induce DNA damage and cell death is a well-known and effective anti-cancer strategy.
Parthenolide (PTL), a Sesquiterpene Lactone (SL) isolated from the buds of feverfew, has received particular attention for its anti-tumor activity in a variety of human cancer cell lines. PTL induces cytotoxicity in a variety of solid tumors, including colorectal, melanoma, pancreatic, breast, prostate, and glioblastoma, but is ineffective in normal tissues. The lack of stability and poor solubility of PTL under acidic and alkaline conditions and in media containing 0.5% serum is a major factor limiting its pharmacological applications.
Gliomas are the most aggressive primary intracranial malignancies of the central nervous system, posing a fatal threat to human health. Currently, the World Health Organization (WHO) classifies gliomas into grades four (I-IV) according to their prognostic histopathological features, with grade IV Glioblastoma (GBM) being the most fatal. Despite tremendous advances in glioma research, including surgery, postoperative adjuvant radiation therapy, and chemotherapy, glioma patients consistently have less than 5% survival 5 years after diagnosis. Therefore, new therapeutic regimens are needed, such as obtaining new drugs that target proliferative tumor cells, to prolong the survival of brain tumor patients, especially GBM patients. Despite the great progress currently made in the treatment of glioblastoma, the prognosis is poor due to resistance to radiotherapy and chemotherapy, with an excessive incidence of morbidity and mortality. There is an urgent need to develop new drugs for inhibiting the proliferation and growth of glioblastoma cells and reversing the multidrug resistance of glioblastoma cells.
Disclosure of Invention
In view of the above, the present invention aims to provide parthenolide derivatives, and a preparation method and applications thereof, wherein the PTL derivatives are synthesized by increasing the solubility of PTL, have a certain inhibitory effect on cell activity of glioblastoma, and in particular, DMAPT-D6 containing deuterium can inhibit the growth of glioblastoma cells by inducing DNA damage due to excessive ROS accumulation, so that the growth of the glioblastoma cells can be inhibited, and the PTL derivatives can be used for preparing a drug for treating glioblastoma.
The invention solves the technical problems by the following technical means:
the invention provides a parthenolide derivative, wherein the structural general formula of the derivative is any one of formula (I), formula (II), formula (III) and formula (IV), and the structural formulas of the formula (I), the formula (II), the formula (III) and the formula (IV) are as follows:
Figure BDA0002533618230000021
wherein, the structural formula of R is ═ CH2
Figure BDA0002533618230000022
Preferably, the derivative is one of the following compounds:
Figure BDA0002533618230000023
Figure BDA0002533618230000031
another aspect of the present invention provides a method for preparing the above parthenolide derivative, wherein the method comprises the following steps:
Figure BDA0002533618230000032
preferably, the preparation method specifically comprises the following steps:
compound PTL and p-toluenesulfonic acid were stirred for reaction in dichloromethane and saturated NaHCO was used3The reaction was quenched, and the resulting organic layer was washed with saturated brine, anhydrous Na2SO4Drying, concentrating under reduced pressure, and recrystallizing with acetone to obtain compound MCL;
stirring compound MCL and m-chloroperoxybenzoic acid in dichloromethane for epoxidation reaction, and sequentially adding Na to the obtained reactant2SO4、NaHCO3And saturated brine, anhydrous Na2SO4Drying, concentrating under reduced pressure, and recrystallizing with acetone to obtain compound 1;
stirring compound 1 and phosphorus oxychloride in pyridine for reaction, adding diethyl ether, and sequentially using NaHCO to obtain reaction mixture3And saturated brine, anhydrous Na2SO4Drying, concentrating under reduced pressure, and separating and purifying on silica gel column to obtain Arglabin;
separating compound PTL, MCL, compound 1 and Arglabin with dimethylamine in tetrahydrofuran under alkaline condition, adding dichloromethane into the obtained reaction mixture, washing with saturated saline, and adding anhydrous Na2SO4Drying, and concentrating under reduced pressure to respectively obtain a compound DMAPT, a compound 2, a compound 3 and a compound 4;
separating compound PTL, MCL, compound 1 and Arglabin in tetrahydrofuran with dimethyl-d 6-amine hydrochloride under alkaline condition, adding dichloromethane into the obtained reaction mixture, washing with saturated saline, and adding anhydrous Na2SO4Drying, concentrating under reduced pressure to obtain compounds DMAPT-D6, 5, 6 and 6And (7) a compound.
Preferably, the compound DMAPT-D6 is prepared by the following specific method:
20mgPTL was dissolved in 2mL tetrahydrofuran, followed by addition of 50mgK2CO3And 20mg of dimethyl-d 6-amine hydrochloride were stirred overnight, 20mL of dichloromethane was added, the mixture was washed with saturated brine, and the resulting organic layer was purified over anhydrous Na2SO4Drying and concentrating under reduced pressure to obtain the compound DMAPT-D6.
In addition, the invention also provides application of the parthenolide derivative, and application of the derivative in preparation of a medicine for treating glioblastoma.
Preferably, the compound DMAPT-D6 is used for preparing a medicine for treating glioblastoma.
Preferably, the compound DMAPT-D6 is used for preparing a medicine for treating glioblastoma, and the compound DMAPT-D6 is used for accumulating intracellular active oxygen to cause DNA damage in glioblastoma cells so as to induce the apoptosis of the glioblastoma cells.
Preferably, the drug for treating glioblastoma comprises a parthenolide derivative or a pharmaceutically acceptable salt, a hydrate or a combination thereof and an auxiliary material.
The invention takes PTL as the initial main raw material, and synthesizes 11 parthenolide derivatives through a series of chemical reactions, and the derivatives have certain antiproliferative activity on glioblastoma cells, in particular, deuterium-containing compound DMAPT-D6 has stronger activity on U87 and LN229 cells. The present invention also demonstrates that DMAPT-D6 significantly induces the accumulation of Reactive Oxygen Species (ROS) in glioblastoma cells, leading to DNA damage in glioblastoma cells. In addition, DMAPT-D6 promoted caspase-dependent death receptor-mediated exogenous apoptosis, suggesting that DNA damage induced by DMAPT-D6 could induce glioblastoma cell apoptosis. Taken together, the data of the present invention indicate that ROS accumulation resulting from DMAPT-D6 treatment leads to DNA damage and then to death receptor-mediated apoptosis, suggesting that DMAPT-D6 with a novel composition has therapeutic potential for the treatment of glioblastoma.
Drawings
FIG. 1 is a graph of cell growth curves of U87 cells and LN229 cells treated with compound DMAPT-D6;
FIG. 2 is a graph of cell growth curves for 24h, 48h, 72h of U87 cells and LN229 cells treated with various concentrations of compound DMAPT-D6;
FIG. 3 is a graph showing the results of a colony formation test;
FIG. 4 is a graph showing the results of the EdU staining test;
FIG. 5 is a graph of flow cytometry analysis cell cycle results;
FIG. 6 is a graph showing the effect of different concentrations of DMAPT-D6 treatment on cell cycle-related proteins P27, CDK1, CDK2, CylinB and Cylin E;
FIG. 7 is a graph showing the effect of treatment with DMAPT-D6 compound at various concentrations on intracellular reactive oxygen species;
FIG. 8 is a graph showing the effect of treatment with the compound DMAPT-D6 at various concentrations on DNA damage;
FIG. 9 is a graph showing the effect of different concentrations of the compound DMAPT-D6 on oxidative damage and DNA repair signal pathway related proteins NRF2, gamma H2AX, 53BP1, DNA LIG IV;
FIG. 10 is a graph showing the effect of treatment with the compound DMAPT-D6 at various concentrations on cell death;
FIG. 11 is a graph showing the results of the analysis of glioma cell apoptosis by Annexin V-FITC/PI in combination with flow cytometry;
FIG. 12 is a graph showing the effect of treatment with DMAPT-D6 on the expression of proteins involved in the cell death receptor signaling pathway at various concentrations;
FIG. 13 is a graph showing the results of recovery of cell death using the caspase inhibitor Z-VAD-FMK;
FIG. 14 is a graph showing the results of the effect of using the inhibitor Z-VAD-FMK on expression of relevant pathway proteins.
Detailed Description
The technical solutions in the embodiments of the present invention will be clearly and completely described below with reference to the drawings in the embodiments of the present invention, and it is obvious that the described embodiments are only a part of the embodiments of the present invention, and not all of the embodiments. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
The parthenolide derivatives are prepared from the main raw materials of Parthenolide (PTL) through chemical reaction, and the parthenolide used in the invention is synthesized by adopting the prior art and purchased. The structural general formula of the parthenolide derivative is any one of formula (I), formula (II), formula (III) and formula (IV), and the structural formulas of formula (I), formula (II), formula (III) and formula (IV) are as follows:
Figure BDA0002533618230000061
wherein, the structural formula of R is ═ CH2
Figure BDA0002533618230000062
The synthetic route of the parthenolide derivative is as follows:
Figure BDA0002533618230000063
specifically, the specific preparation method of each derivative is as follows:
the product testing conditions in the following examples are as follows: on a 400MHz solid nuclear magnetic resonance spectrometer (Bruker AVANCEIII400MHz), recording by taking Tetramethylsilicon (TMS) as an internal standard1H and13C NMR。1h NMR data are reported below: chemical shift, in ppm (), multiplicity (s ═ singlet, d ═ doublet, t ═ triplet, m ═ multiplet), coupling constant (Hz), relative intensity;13the C NMR data are reported below: chemical shift (ppm).
Example 1
The structural formula of compound MCL is as follows:
Figure BDA0002533618230000071
the preparation method of compound MCL is as follows: to 100mLCH2Cl2To this solution was added 86mg of p-toluenesulfonic acid (0.5mmol) to prepare a p-toluenesulfonic acid solution, and the solution was stirred at 20mL CH2Cl2Adding parthenolide (14mmol) 3.5g to obtain parthenolide solution, adding parthenolide solution dropwise into p-toluenesulfonic acid solution at room temperature, stirring at room temperature overnight, and adding 20mL saturated NaHCO3The reaction was quenched, and the resulting organic layer was washed with saturated brine (2 × 20mL), anhydrous Na2SO4Drying and concentration under reduced pressure gave a crude residue which was recrystallized from acetone to give 3.2g of a pale yellow crystalline solid, Compound MCL, in a yield of 91% by calculation.
1H NMR(400MHz,CDCl3)6.22(d,J=3.3Hz,1H),5.51(d,J=3.0Hz,1H),3.82(t,J=10.3Hz,1H),2.80–2.59(m,3H),2.40(dd,J=16.3,8.4Hz,1H),2.30–2.14(m,3H),2.14–2.04(m,
1H),1.84–1.74(m,2H),1.69(s,3H),1.40–1.28(m,4H).13C NMR(101MHz,CDCl3)169.71,
138.88,131.87,130.88,119.39,84.45,80.25,58.69,49.62,38.38,34.97,30.08,25.81,23.88,22.73.
Example 2
The structural formula of compound 1 is as follows:
Figure BDA0002533618230000072
the preparation method of the compound 1 comprises the following steps:
1.75g of the compound MCL (mmol) prepared in example 1 and 1.8g of m-chloroperoxybenzoic acid (10.5mmol) are added to 50mL of CH at room temperature2Cl2Stirring overnight, the resulting reaction mixture was then washed with Na2SO4(2×30mL)、NaHCO3(2 × 50mL) and saturated brine (2 × 30mL), anhydrous Na2SO4Drying and concentration under reduced pressure gave a crude residue which was recrystallized from acetone to give 1.3g of a crystalline solid, Compound 1, in a calculated yield of 70%.
1H NMR(400MHz,CDCl3)6.20(d,J=3.3Hz,1H),5.48(d,J=3.0Hz,1H),4.05(t,J=10.4Hz,1H),2.81(s,1H),2.38–2.19(m,4H),2.04–1.82(m,4H),1.70–1.61(m,1H),1.48(s,3H),
1.46–1.37(m,1H),1.30(s,3H).13C NMR(101MHz,CDCl3)169.62,138.16,119.55,81.83,79.71,69.89,62.19,55.62,49.45,37.37,33.41,29.53,23.26,21.97.
Example 3
The structural formula of the compound Arglabin is as follows:
Figure BDA0002533618230000081
the preparation method of the compound Arglabin comprises the following steps:
264mg of the compound 1(1.0mmol) prepared in example 2 are added to 5mL of pyridine at 0 ℃ and stirred, 300. mu.L of phosphorus oxychloride is further added to the obtained solution under stirring, the mixture is stirred for 2 hours, 30mL of diethyl ether is added, and the obtained organic layer is sequentially treated with NaHCO3And saturated brine, anhydrous Na2SO4Drying and concentration under reduced pressure gave a crude residue which was isolated and purified on a silica gel column to give 112mg of the compound Arglabin in a calculated yield of 45%.
1H NMR(400MHz,CDCl3)6.15(d,J=3.3Hz,1H),5.58(s,1H),5.42(d,J=3.1Hz,1H),4.01(t,J=10.2Hz,1H),2.94(d,J=10.7Hz,1H),2.83–2.74(m,1H),2.29–2.11(m,3H),2.07–2.01(m,1H),1.99(d,J=7.9Hz,3H),1.88–1.82(m,1H),1.55–1.45(m,1H),1.35(d,J=6.4Hz,3H).13C NMR(101MHz,CDCl3)170.43,140.57,139.14,124.91,118.27,82.89,72.52,62.68,52.85,51.05,39.71,33.48,22.79,21.45,18.25.
Example 4
The compound DMAPT has the following structural formula:
Figure BDA0002533618230000082
the compound DMAPT was prepared as follows: 20mg of Parthenolide (PTL) was dissolved in 2mL of tetrahydroFuran (THF), then adding 10mgK respectively2CO3And 0.5mL dimethylamine (40 wt% in water), the resulting mixture was stirred at room temperature overnight, and 20mL CH was added2Cl2Washed with saturated brine (2 × 20mL), and the resulting organic layer was purified over anhydrous Na2SO4Drying and concentration under reduced pressure gave 22mg of a yellow solid, compound DMAPT, in a yield of 93%.
1H NMR(400MHz,CDCl3)5.25–5.16(m,1H),3.86(t,J=9.1Hz,1H),2.89–2.82(m,1H),2.78–2.70(m,2H),2.57(d,J=11.2Hz,1H),2.39(s,6H),2.33–2.02(m,7H),1.74–1.62(m,4H),1.30(s,3H),1.26–1.20(m,1H).13C NMR(101MHz,CDCl3)176.21,134.68,125.12,82.29,66.38,61.56,57.31,48.21,46.15,45.77,41.09,36.68,29.81,24.12,17.23,16.93.HRMS(ESI)m/z calcd for C17H28NO3 +(M+H)+294.20637,found 294.20624.
Example 5
The structural formula of compound 2 is as follows:
Figure BDA0002533618230000083
the preparation method of the compound 2 comprises the following steps: 20mg of the compound MCL prepared in example 1 was dissolved in 2mL of Tetrahydrofuran (THF), and 10mgK was added thereto2CO3And 0.5mL dimethylamine (40 wt% in water), the resulting mixture was stirred at room temperature overnight, and 20mL CH was added2Cl2Washed with saturated brine (2 × 20mL), and the resulting organic layer was purified over anhydrous Na2SO4Drying and concentration under reduced pressure gave 23mg of a yellow solid, Compound 2, in a 97% yield.
1H NMR(400MHz,CDCl3)3.76(t,J=10.3Hz,1H),2.67(dd,J=12.9,5.0Hz,1H),2.60–2.49(m,2H),2.38–2.28(m,2H),2.20(s,6H),2.14–2.03(m,4H),1.96(d,J=11.4Hz,1H),1.72(dd,J=15.1,8.3Hz,2H),1.60(s,3H),1.21(d,J=17.0Hz,4H).13C NMR(101MHz,CDCl3)176.00,130.84,130.34,83.13,79.34,57.34,57.11,49.97,44.93,43.68,37.43,34.37,29.00,26.32,22.75,21.81.HRMS(ESI)m/z calcd for C17H28NO3 +(M+H)+294.20637,found 294.20630.
Example 6
The structural formula of compound 3 is as follows:
Figure BDA0002533618230000091
the preparation method of the compound 3 comprises the following steps: 20mg of Compound 1 prepared in example 2 was dissolved in 2mL of Tetrahydrofuran (THF), and 10mgK was added thereto2CO3And 0.5mL dimethylamine (40 wt% in water), the resulting mixture was stirred at room temperature overnight, and 20mL CH was added2Cl2Washed with saturated brine (2 × 20mL), and the resulting organic layer was purified over anhydrous Na2SO4Drying and concentration under reduced pressure gave 22mg of a yellow solid, Compound 3, in a calculated yield of 96%.
1H NMR(400MHz,CDCl3)4.00(t,J=10.4Hz,1H),2.63(dd,J=13.0,5.0Hz,1H),2.51–2.44(m,1H),2.31–2.24(m,1H),2.17(s,6H),2.09–2.03(m,1H),1.88–1.81(m,3H),1.80–1.70(m,2H),1.66–1.53(m,3H),1.39(s,3H),1.34–1.27(m,1H),1.21(s,3H).13C NMR(101MHz,CDCl3)175.94,80.60,78.67,68.84,61.23,56.89,54.27,49.39,44.93,43.22,36.42,32.57,28.37,22.20.HRMS(ESI)m/z calcd for C17H28NO4 +(M+H)+310.20128,found310.20114.
Example 7
The structural formula of compound 4 is as follows:
Figure BDA0002533618230000092
the preparation method of the compound 4 comprises the following steps: 20mg of Arglabin, a compound prepared in example 3, was dissolved in 2mL of Tetrahydrofuran (THF), and 10mgK was added thereto2CO3And 0.5mL dimethylamine (40 wt% in water), the resulting mixture was stirred at room temperatureAt night, 20mL of CH was added2Cl2Washed with saturated brine (2 × 20mL), and the resulting organic layer was purified over anhydrous Na2SO4Drying and concentration under reduced pressure gave 23mg of a yellow solid, Compound 4, in a calculated yield of 92%.
1H NMR(400MHz,CDCl3)5.49(s,1H),3.94(t,J=10.2Hz,1H),2.68(ddd,J=21.9,17.7,7.6Hz,3H),2.49(dd,J=13.0,6.1Hz,1H),2.23(dd,J=11.8,5.5Hz,1H),2.17(s,6H),2.10–2.00(m,2H),1.86(s,4H),1.56(dd,J=22.8,10.8Hz,2H),1.39(d,J=12.5Hz,1H),1.26(s,3H).13C NMR(101MHz,CDCl3)176.72,139.74,123.71,81.53,71.49,61.65,56.99,51.43,50.87,45.04,43.55,38.56,32.66,21.83,21.70,17.27.HRMS(ESI)m/zcalcd for C17H26NO3 +(M+H)+292.19072,found 292.19067.
Example 8
The compound DMAPT-D6 has the following structural formula:
Figure BDA0002533618230000101
the compound DMAPT-D6 was prepared as follows: 20mg of Parthenolide (PTL) was dissolved in 2mL of Tetrahydrofuran (THF), and 50mg K was added2CO3And 20mg of dimethylamine-d 6-amine hydrochloride, the resulting mixture was stirred at room temperature overnight and 20mL of CH was added2Cl2Washed with saturated brine (2 × 20mL), and the resulting organic layer was purified over anhydrous Na2SO4Drying and concentration under reduced pressure gave 20mg of a yellow solid, compound DMAPT-D6, in 83% yield.
1H NMR(400MHz,CDCl3)5.14(d,J=9.9Hz,1H),3.76(t,J=9.0Hz,1H),2.68(dt,J=6.7,3.9Hz,2H),2.56(dd,J=13.2,4.7Hz,1H),2.35–2.27(m,2H),2.25–2.16(m,2H),2.12–1.96(m,4H),1.62(d,J=6.1Hz,3H),1.60–1.50(m,1H),1.23(s,3H),1.17(dd,J=13.1,5.6Hz,1H).13C NMR(101MHz,CDCl3)175.44,133.64,124.10,81.10,65.50,60.43,56.57,46.92,45.57,40.12,35.67,28.98,23.11,16.23,15.92.HRMS(ESI)m/z calcd forC17H22D6NO3 +(M+H)+300.24403,found 300.24438.
Example 9
The structural formula of compound 5 is as follows:
Figure BDA0002533618230000102
the preparation method of the compound 5 comprises the following steps: 20mg of MCL prepared in example 1 was dissolved in 2mL of Tetrahydrofuran (THF), and 50mgK was added thereto2CO3And 20mg of dimethylamine-d 6-amine hydrochloride, the resulting mixture was stirred at room temperature overnight and 20mL of CH was added2Cl2Washed with saturated brine (2 × 20mL), and the resulting organic layer was purified over anhydrous Na2SO4Drying and concentration under reduced pressure gave 23mg of a yellow solid, compound 5, in a calculated yield of 95%.
1H NMR(400MHz,CDCl3)3.75(t,J=10.3Hz,1H),2.70–2.45(m,4H),2.31(dt,J=11.6,5.3Hz,2H),2.21–2.04(m,4H),1.97(dd,J=22.6,11.1Hz,1H),1.78–1.67(m,2H),1.61(s,3H),1.23(s,3H),1.21–1.15(m,1H).13C NMR(101MHz,CDCl3)176.06,130.86,130.32,83.11,79.33,57.36,57.10,49.94,43.76,37.41,34.39,29.00,26.36,22.75,21.81.HRMS(ESI)m/z calcd for C17H22D6NO3 +(M+H)+300.24403,found 300.24490.
Example 10
The structural formula of compound 6 is as follows:
Figure BDA0002533618230000111
the preparation method of the compound 6 comprises the following steps: 20mg of Compound 1 prepared in example 2 was dissolved in 2mL of Tetrahydrofuran (THF), and 50mgK was added thereto2CO3And 20mg of dimethylamine-d 6-amine hydrochloride, the resulting mixture was stirred at room temperature overnight and 20mL of CH was added2Cl2Washed with saturated brine (2 × 20mL), and the resulting organic layer was purified over anhydrous Na2SO4Drying and concentration under reduced pressure gave 21mg of a yellow solid, compound 6, in a calculated yield of 90%.
1H NMR(400MHz,CDCl3)4.00(t,J=10.4Hz,1H),2.61(dd,J=13.0,5.0Hz,1H),2.47(dd,J=13.0,6.2Hz,1H),2.28–2.12(m,3H),2.09–2.02(m,1H),1.84(dd,J=11.6,6.2Hz,3H),1.79–1.73(m,1H),1.65–1.55(m,2H),1.39(s,3H),1.30(d,J=11.8Hz,1H),1.21(s,3H).13C NMR(101MHz,CDCl3)176.00,80.56,78.66,68.85,61.23,56.84,54.27,49.38,43.27,36.42,32.58,28.37,22.20.HRMS(ESI)m/z calcd for C17H22D6NO4 +(M+H)+316.23895,found316.23895.
Example 11
The structural formula of compound 7 is as follows:
Figure BDA0002533618230000112
the preparation method of the compound 7 comprises the following steps: 20mg of Arglabin, a compound prepared in example 3, was dissolved in 2mL of Tetrahydrofuran (THF), and 50mgK was added thereto2CO3And 20mg of dimethylamine-d 6-amine hydrochloride, the resulting mixture was stirred at room temperature overnight and 20mL of CH was added2Cl2Washed with saturated brine (2 × 20mL), and the resulting organic layer was purified over anhydrous Na2SO4Drying and concentration under reduced pressure gave 23mg of a yellow solid, Compound 7, in a 91% yield.
1H NMR(400MHz,CDCl3)5.56(s,1H),4.01(t,J=10.2Hz,1H),2.86–2.67(m,3H),2.57(dd,J=13.1,6.0Hz,1H),2.33–2.00(m,5H),1.93(s,3H),1.46(dd,J=18.6,6.3Hz,2H),1.33(s,3H).13C NMR(101MHz,CDCl3)177.74,140.75,124.75,82.58,72.51,62.68,57.78,52.46,51.84,44.57,39.58,33.67,29.71,29.33,27.23,22.86,22.73,18.29,14.11.HRMS(ESI)m/z calcd for C17H20D6NO3 +(M+H)+298.22838,found 298.22818.
Example 12
In this example, the effect of the parthenolide derivatives prepared in examples 1 to 11 on glioblastoma was examined, specifically as follows.
The human glioblastoma cell lines U87 and LN229 used in this example were purchased from Cobioer, both of which were mycoplasma free and have been identified by STR testing. Both the U87 and LN229 cell lines were cultured in DMEM medium, purchased from HyClone, containing 10% Fetal Bovine Serum (FBS) and 1% penicillin-streptomycin solution (100U/ml Penicillium and 100. mu.g/ml streptomycin) at 37 ℃ in 5% CO2Culturing in a constant temperature incubator. The cell culture steps are as follows:
(1) after counting U87 cells with a fully automatic cell counter, U87 cells and LN229 were diluted to 5 × 10 with DMEM medium3Obtaining cell suspension per mL;
(2) adding 100 mu L of cell suspension into each hole of a 96-hole plate, blowing, uniformly mixing, and culturing in an incubator at 37 ℃ for 24 hours;
(3) PTL and the compounds prepared in examples 1 to 11 were diluted with dimethyl sulfoxide to concentrations of 0.5. mu.M, 1.0. mu.M, 1.5. mu.M, and 2.0. mu.M, respectively, and added to the respective cultured cell lines, followed by incubation at 37 ℃ in an incubator for 48 hours;
(4) MTT solution (20. mu.L/well) was added and incubated at 37 ℃ for 4h in an incubator;
(5) the supernatant was removed, the crystals formed were dissolved in DMSO (200. mu.L/well) and then assayed for OD570 nm (OD570) using a microplate reader (Bio-Tek, VT, USA) and analyzed by GraphPad Prism 7.0, the effect of each compound on U87 cell viability being shown in Table 1, the data in Table 1 showing that the parthenolide derivatives prepared according to the invention all have different degrees of effect on U87 cell viability, with the IC of compound DMAPT-D6 in U87 cells50The value was 15.5. mu.M.
Figure BDA0002533618230000121
TABLE 1
Compound DMAPT-D6 vs U87 cellsAnd LN229 cell viability as shown in FIG. 1, the data of FIG. 1 indicate the IC of DMAPT-D6 in U87 cells and LN229 cells50The values were 15.5. mu.M and 11.15. mu.M, respectively. To further observe the inhibitory effect of compound DMAPT-D6 on glioblastoma, glioblastoma cells were treated at concentrations of 2.5. mu.M, 5. mu.M, 10. mu.M, 20. mu.M, and 40. mu.M for 24h, 48h, and 72h, respectively, and the relationship between cell growth and exposure time and dose was observed, the growth profile of which is shown in FIG. 2, and the growth profile of FIG. 2 shows that compound DMAPT-D6 decreased the proliferative capacity of U87 and LN229 cells, indicating that the compound has dose-and time-dependent cytotoxicity on glioblastoma cells.
To further confirm the growth inhibitory effect of DMAPT-D6 on glioblastoma cells, colony formation assays were performed in this example, using human glioblastoma cells U87 and LN229 at a cell concentration of 1 × 103The cells/cell/well were inoculated on a six-well plate, cultured overnight, and after treating the cells with the compound DMAPT-D6 at concentrations of 0. mu.M, 5. mu.M, 10. mu.M, and 20. mu.M for 48 hours, the treated cells were cultured with a fresh medium for 15 days, fixed with 4% paraformaldehyde solution for 30 minutes, the cells were stained with 1% crystal violet for 30 minutes to identify colonies, and the number of colonies of at least 50 cells was counted under a 4-fold microscope, the results are shown in FIG. 3. FIG. 3 shows that DMAPT-D6 has significant inhibitory effect on the growth of glioblastoma cells, and is dose-dependent, and DMAPT-D6 has significant inhibitory effect on the growth of glioblastoma cells compared to the control group without DMAPT-D6. The EdU staining test was performed, and the test results are shown in fig. 4, and fig. 4 shows that the cell number was significantly reduced in a dose-dependent manner after treatment with compound DMAPT-D6 in the EdU staining test, indicating that compound DMAPT-D6 has a significant inhibitory effect on the proliferation of U87 and LN229 cells.
To further understand the mechanism of cytotoxic effect of the compound DMAPT-D6 on glioblastoma cells, this example also performed cell cycle analysis on U87 and LN229 cells treated with the compound DMAPT-D6 or without DMAPT-D6. Specifically, glioblastoma cells in a logarithmic growth phase were collected and transferred to a six-well plate, the cell density in each well was 30%, the cells were treated with the compound DMAPT-D6 at concentrations of 0 μ M, 5 μ M, 10 μ M, and 20 μ M for 48 hours, collected and used for flow cytometry analysis, for cell cycle determination, the collected cells were fixed with 70% ethanol at 4 ℃ for 24 hours, and then washed 3 times with PBS, and then, the cells were incubated with PBS containing 50mg/ml propidium iodide and 100mg/ml RNase at 37 ℃ for 0.5 hour, and finally, the stained cells were analyzed by BD-accuri-C6 flow cytometry, and the results were statistically compared and visualized automatically using FlowJo software; for apoptosis detection, cells were harvested and processed using Annexin V-FITC/PI apoptosis assay kit according to the manufacturer's instructions, fluorescence activated cells were analyzed within 1 hour using a BD-AccuriTM-C6 flow cytometer, and the apoptosis rate was analyzed by FlowJo software, with the results shown in FIG. 5. FIG. 5 shows that compound DMAPT-D6 has significant dose-dependent induced cell cycle arrest on both U87 and LN229 cells, arresting the cell cycle in S phase.
Based on the above experiments, immunoblot analysis was also performed in this example, and specifically, collected glioblastoma cells were analyzed in the presence of HaltTMThe protein samples (30 μ g/lane) were separated by electrophoresis after 30min of lysis in RIPA buffer of protease and phosphatase inhibitors (Beyotime, shanghai, china), and after quantification of total protein concentration using BCA protein assay kit (Beyotime, shanghai, china), transferred to PVDF membrane (Millipore, Billerica, MA, usa), followed by blocking the membrane with 5% BSA for 2h at room temperature, incubation of one antibody overnight at 4 ℃, followed by incubation with the corresponding IRDye 800 CW-goat anti-mouse IgG (h + L) or IRDye 800-LT donkey anti-rabbit IgG (h + L) secondary antibody, followed by imaging using Odyssey infrared fluorescence imaging system (Li-cor, NE, usa), and using β -actin supernatant as a control, as shown in fig. 6, the immunoblot results show that the compound DMAPT-D6 significantly reduced the protein levels of cyclin B, cyclin E, 1 and CDK 34, while the protein levels of CDK 56 and pt 39229 in cell line increased with increasing the dose of the compound dmaln-46 6.
The above data indicate that compound DMAPT-D6 may inhibit cell proliferation by inducing cell cycle arrest in the S phase of glioblastoma cells.
To further understand the mechanism by which DMAPT-D6 induces inhibition of cell proliferation, the production of Reactive Oxygen Species (ROS) in U87 and LN229 cells was observed using a fluorescence microscope. Specifically, the formation of ROS was detected using an ROS detection kit (Beyotime, shanghai, china), glioblastoma cells treated with various concentrations of the compound DMAPT-D6 were collected, centrifuged at 800rpm for 5 minutes, the resulting precipitate after centrifugation was resuspended in serum-free DMEM containing DCFH-DA (10 μ M) and incubated for 20 minutes, and finally, the cell suspension was centrifuged and washed 3 times with serum-free DMEM, and then observed by a fluorescence microscope (Olympus IX53/DP80, Japan), and as a result, the reactive oxygen species level generated by the treatment with various concentrations of DMAPT-D6 was significantly higher than that of the control group which had not been treated with DMAPT-D6, as shown in fig. 7.
Several studies have shown that intracellular ROS accumulation can induce DNA damage and lead to DNA damage responses, and in order to evaluate whether DMAPT-D6 can initiate the DNA damage process caused by excessive intracellular ROS, phosphohistone γ H2AX was detected in U87 and LN229 cell lines by immunofluorescence and immunoblotting, the procedure for immunofluorescence was as follows: glioblastoma cells were grown in 24-well plates and then treated with varying concentrations of DMAPT-D6 for 48 hours, washed 3 times with PBS, the cells were fixed in 4% paraformaldehyde for 30 minutes, permeabilized in 0.1% Triton X-100 for 10 minutes, and blocked in immunostaining blocking buffer (Beyotime, shanghai) at 37 ℃ for 30 minutes, the cells were incubated with anti- γ H2AX (1: 250) antibody, respectively, at 4 ℃ overnight, then the stained cells were washed 3 times with PBS and incubated with Alexa Fluor-coupled secondary antibody (1: 2000) for 1 hour at room temperature, the nuclei were labeled with 1mg/mL DAPI for 30 minutes, images were captured with a fluorescence microscope (Olympus IX53/DP80, japan), and the immunofluorescence results are shown in figure 8. The results shown in fig. 8 indicate that in U87 and LN229 cells, the green signal representing γ H2AX was significantly accumulated in the nucleus in a dose-dependent manner and γ H2AX was significantly upregulated compared to the control, indicating DNA double strand breaks in TNBC cells. In order to detect whether the DMAPT-D6 destroys the DNA Repair (DR) process, the expression level of DR-related proteins such as p53 binding protein 1(53BP1) and DNA ligase IV (LIG IV) in glioblastoma cells was detected by immunoblotting, and the results of immunoblotting are shown in FIG. 9. FIG. 9 shows that expression of NRF2 is dose-dependently upregulated with increasing DMAPT-D6 concentration after DMAPT-D6 treatment, suggesting that too much ROS accumulate in the cells. Furthermore, as shown in fig. 9, DMAPT-D6 significantly caused a reduction in the expression of both 53BP1 and DNA LIG IV proteins in a dose-dependent manner, showing an inhibitory effect on DR.
Disproportionate increases in ROS and severe DNA damage can induce endogenous and exogenous apoptotic pathways, mediated by mitochondrial and cell death receptor signaling, respectively. Based on the effect of DMAPT-D6 on ROS induction and subsequent DNA damage, the present invention also discusses whether DMAPT-D6 can promote the initiation of apoptosis. Notably, PI staining analysis showed a significant increase in the percentage of PI positive cells in U87 and LN229 cell lines with increasing concentrations of DMAPT-D6, as shown in fig. 10, indicating that DMAPT-D6 induced cell death. To further confirm the effect of DMAPT-D6 on apoptosis, Annexin V-FITC/PI analysis was performed by flow cytometry 48 hours after U87 and LN229 cells were treated with DMAPT-D6, as shown in fig. 11, DMAPT-D6 induced 7.28% and 10.7% late apoptosis (Annexin V-FITC and PI positive cells) in U87 and LN229 cell lines, respectively, at low doses of 5 μ M; at a dose of 20 μ M, the percentage of apoptosis rose to 46.9% and 34.7%, respectively. Next, the expression of external apoptosis signaling pathway-related proteins was analyzed to determine whether cell death receptors participate in the apoptotic response to DMAPT-D6, and consistent with the above results, cell death receptor signaling pathway-related proteins such as DR3, DR5, FADD, TRADD were significantly up-regulated with increasing concentrations of DMAPT-D6, as shown in fig. 12. Subsequently, Caspase precursor (Procaspase)8 is cleaved to produce the active enzyme form of Caspase 8(Caspase 8) due to the increase of FADD and TRADD. In addition, downstream caspase 3 and PARP are splice-activated in U87 and LN229 cells, meaning that it mediates exogenous apoptosis through death receptors, as shown in fig. 12. To further demonstrate DMAPT-D6-induced apoptosis mediated by cell death receptors, a Caspase inhibitor, Z-VAD-FMK, was used to test the recovery effect of DMAPT-D6-induced apoptosis, as shown in FIG. 13, which can significantly rescue DMAPT-D6-induced apoptosis and partially restore the protein levels of DNA Lig IV, DR3, Caspase8, 3 and PARP when treated with Z-VAD-FMK, as shown in FIG. 14, indicating that DMAPT-D6-induced apoptosis mediated by death receptors is dependent on caspases.
Taken together, the compound DMAPT-D6 of the present invention significantly up-regulated the activity of proteins associated with death receptor signaling pathways, such as DR3, DR5, FADD, TRADD, and cysteine proteases 3, 8, and PARP, indicating that death receptor-mediated exogenous apoptosis is induced following DMAPT-D6 treatment; the results show that U87 and LN229 cells after being treated with compound DMAPT-D6 induce excessive ROS accumulation to cause DNA damage, and further induce cell cycle S phase block and cell death receptor mediated cell external apoptosis signal pathway, thereby inhibiting growth of glioblastoma cells and playing an anti-cancer role on the glioblastoma cells.
Therefore, the compound DMAPT-D6 is a potential anticancer drug with active oxygen regulation capacity, can be used as a lead compound to be applied to the preparation of drugs for treating glioblastoma, and particularly to the preparation of drugs for treating glioblastoma, wherein the drugs for treating glioblastoma comprise the 11 parthenolide derivatives or pharmaceutically acceptable salts, hydrates or combinations and auxiliary materials thereof.
Although the present invention has been described in detail with reference to the preferred embodiments, it will be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the spirit and scope of the invention as defined in the appended claims. The techniques, shapes, and configurations not described in detail in the present invention are all known techniques.

Claims (9)

1. The parthenolide derivative is characterized in that the structural general formula of the derivative is any one of a formula (I), a formula (II), a formula (III) and a formula (IV), and the structural formulas of the formula (I), the formula (II), the formula (III) and the formula (IV) are as follows:
Figure FDA0002533618220000011
wherein, the structural formula of R is ═ CH2Or
Figure FDA0002533618220000012
2. The parthenolide derivative according to claim 1, wherein the derivative is one of the following compounds:
Figure FDA0002533618220000013
Figure FDA0002533618220000021
3. the preparation method of the parthenolide derivative is characterized by comprising the following steps:
Figure FDA0002533618220000022
4. the method for preparing a parthenolide derivative according to claim 3, wherein the method specifically comprises the following steps:
compound PTL and p-toluenesulfonic acid were stirred for reaction in dichloromethane and saturated NaHCO was used3The reaction was quenched, and the resulting organic layer was washed with saturated brine, anhydrous Na2SO4Drying, concentrating under reduced pressure, and recrystallizing with acetone to obtain compound MCL;
stirring the compound MCL and m-chloroperoxybenzoic acid in dichloromethane for epoxidation reaction to obtainTo the reactant is sequentially added with Na2SO4、NaHCO3And saturated brine, anhydrous Na2SO4Drying, concentrating under reduced pressure, and recrystallizing with acetone to obtain compound 1;
stirring compound 1 and phosphorus oxychloride in pyridine for reaction, adding diethyl ether, and sequentially using NaHCO to obtain reaction mixture3And saturated brine, anhydrous Na2SO4Drying, concentrating under reduced pressure, and purifying and separating on silica gel column to obtain Arglabin;
separating compound PTL, MCL, compound 1 and Arglabin with dimethylamine in tetrahydrofuran under alkaline condition, adding dichloromethane into the obtained reaction mixture, washing with saturated saline, and adding anhydrous Na2SO4Drying, and concentrating under reduced pressure to respectively obtain a compound DMAPT, a compound 2, a compound 3 and a compound 4;
separating compound PTL, MCL, compound 1 and Arglabin in tetrahydrofuran with dimethyl-d 6-amine hydrochloride under alkaline condition, adding dichloromethane into the obtained reaction mixture, washing with saturated saline, and adding anhydrous Na2SO4Drying and concentrating under reduced pressure to obtain the compound DMAPT-D6, the compound 5, the compound 6 and the compound 7 respectively.
5. The parthenolide derivative according to claim 4, wherein the compound DMAPT-D6 is prepared by the following method:
20mgPTL was dissolved in 2mL tetrahydrofuran, followed by addition of 50mgK2CO3And 20mg of dimethyl-d 6-amine hydrochloride were stirred overnight, 20mL of dichloromethane was added, the mixture was washed with saturated brine, and the resulting organic layer was purified over anhydrous Na2SO4Drying and concentrating under reduced pressure to obtain the compound DMAPT-D6.
6. Use of a parthenolide derivative according to claim 1 or 2 in the manufacture of a medicament for the treatment of glioblastoma.
7. The use of a parthenolide derivative according to claim 6, wherein compound DMAPT-D6 is used in the preparation of a medicament for the treatment of glioblastoma.
8. The use of the parthenolide derivative according to claim 7, wherein the compound DMAPT-D6 is used for preparing a drug for treating glioblastoma through accumulation of intracellular reactive oxygen species, resulting in DNA damage in glioblastoma cells, thereby inducing glioblastoma cell apoptosis.
9. The use of a parthenolide derivative according to claim 8, wherein the medicament for treating glioblastoma comprises a parthenolide derivative or a pharmaceutically acceptable salt, hydrate or combination thereof and an adjuvant.
CN202010525365.1A 2020-06-10 2020-06-10 Parthenolide derivative and preparation method and application thereof Withdrawn CN111518065A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010525365.1A CN111518065A (en) 2020-06-10 2020-06-10 Parthenolide derivative and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010525365.1A CN111518065A (en) 2020-06-10 2020-06-10 Parthenolide derivative and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN111518065A true CN111518065A (en) 2020-08-11

Family

ID=71912928

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010525365.1A Withdrawn CN111518065A (en) 2020-06-10 2020-06-10 Parthenolide derivative and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN111518065A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115403545A (en) * 2021-05-28 2022-11-29 南京中医药大学 Guaiane sesquiterpene prodrug and application thereof

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115403545A (en) * 2021-05-28 2022-11-29 南京中医药大学 Guaiane sesquiterpene prodrug and application thereof
CN115403545B (en) * 2021-05-28 2024-03-26 南京中医药大学 Guaiane sesquiterpene prodrug and application thereof

Similar Documents

Publication Publication Date Title
Bisi et al. Coumarin derivatives as potential antitumor agents: Growth inhibition, apoptosis induction and multidrug resistance reverting activity
Chou et al. Synthesis of furopyrazole analogs of 1-benzyl-3-(5-hydroxymethyl-2-furyl) indazole (YC-1) as novel anti-leukemia agents
CA2736097C (en) Carbazole compounds for inhibition of nf-kb activity
Vianna et al. Selective cytotoxicity and apoptosis induction in glioma cell lines by 5-oxygenated-6, 7-methylenedioxycoumarins from Pterocaulon species
Gao et al. Synthesis and anticancer activity of some novel 2-phenazinamine derivatives
Li et al. Synthesis, structure–activity relationship and biological evaluation of anticancer activity for novel N-substituted sophoridinic acid derivatives
Chipoline et al. Molecular mechanism of action of new 1, 4-naphthoquinones tethered to 1, 2, 3-1H-triazoles with cytotoxic and selective effect against oral squamous cell carcinoma
Han et al. Design, synthesis and biological evaluation of thieno [3, 2-d] pyrimidine derivatives containing aroyl hydrazone or aryl hydrazide moieties for PI3K and mTOR dual inhibition
Ma et al. Melatonin derivatives combat with inflammation-related cancer by targeting the Main Culprit STAT3
Kumari et al. Stereoselective synthesis of carbohydrate fused pyrano [3, 2-c] pyranones as anticancer agents
Yao et al. Stereoisomeric guaiacylglycerol-β-coniferyl aldehyde ether induces distinctive apoptosis by downregulation of MEK/ERK pathway in hepatocellular carcinoma cells
Zeng et al. Selective stabilization of multiple promoter G-quadruplex DNA by using 2-phenyl-1H-imidazole-based tanshinone IIA derivatives and their potential suppressing function in the metastatic breast cancer
Li et al. S3I-201 derivative incorporating naphthoquinone unit as effective STAT3 inhibitors: Design, synthesis and anti-gastric cancer evaluation
Zhou et al. Synthesis and evaluation of novel 12-aryl berberine analogues with hypoxia-inducible factor-1 inhibitory activity
CN111518065A (en) Parthenolide derivative and preparation method and application thereof
JP2021523934A (en) Aminopyrazine and related compounds useful as mitochondrial deconjugation agents
WO2012100723A1 (en) Use of artemisinin b in preparation of antitumor drug
Ostrowska et al. Anticancer effects of O-aminoalkyl derivatives of alloxanthoxyletin and seselin
Awasthi et al. Antitumor activity of a pexidartinib bioisostere inhibiting CSF1 production and CSF1R kinase activity in human hepatocellular carcinoma
Nyein et al. Synthesis and anti-glioblastoma effects of artemisinin-isothiocyanate derivatives
Rhee et al. Synthesis, cytotoxicity and topoisomerase II inhibitory activity of benzonaphthofurandiones
Yang et al. Toward chelerythrine optimization: Analogues designed by molecular simplification exhibit selective growth inhibition in non-small-cell lung cancer cells
Xu et al. Design, Synthesis, and Biological Evaluation of Novel Amyl Ester Tethered Dihydroartemisinin‐Isatin Hybrids as Potent Anti‐Breast Cancer Agents
CN107011312A (en) Jungermanniaceae D nitrogen containing derivative and preparation method thereof and the purposes in treatment tumor disease
JP2016522223A (en) Substance having tyrosine kinase inhibitory activity and preparation method and use 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
WW01 Invention patent application withdrawn after publication

Application publication date: 20200811

WW01 Invention patent application withdrawn after publication