CN114426470A - Application of human PCID2 protein in preparation or screening of antitumor drugs and compound with antitumor activity - Google Patents

Application of human PCID2 protein in preparation or screening of antitumor drugs and compound with antitumor activity Download PDF

Info

Publication number
CN114426470A
CN114426470A CN202210062797.2A CN202210062797A CN114426470A CN 114426470 A CN114426470 A CN 114426470A CN 202210062797 A CN202210062797 A CN 202210062797A CN 114426470 A CN114426470 A CN 114426470A
Authority
CN
China
Prior art keywords
pcid2
compound
protein
tumor
liver cancer
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
CN202210062797.2A
Other languages
Chinese (zh)
Inventor
白仲添
张保新
席莉莉
周建业
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.)
First Hospital of Lanzhou University
Original Assignee
First Hospital of Lanzhou University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by First Hospital of Lanzhou University filed Critical First Hospital of Lanzhou University
Priority to CN202210062797.2A priority Critical patent/CN114426470A/en
Publication of CN114426470A publication Critical patent/CN114426470A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/76Ketones containing a keto group bound to a six-membered aromatic ring
    • C07C49/84Ketones containing a keto group bound to a six-membered aromatic ring containing ether groups, groups, groups, or groups
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C205/00Compounds containing nitro groups bound to a carbon skeleton
    • C07C205/45Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by at least one doubly—bound oxygen atom, not being part of a —CHO group

Abstract

The invention belongs to the technical field of biotechnology and gene therapy, and particularly relates to application of human PCID2 protein in preparation or screening of antitumor drugs and an antitumor compound. The PCID2 gene is found to promote the proliferation of tumor cells and regulate the cell cycle, is a key molecule for regulating the occurrence and development of tumors, and can be used as a target protein for preparing or screening antitumor drugs; meanwhile, the human PCID2 protein is used as a target protein, and a class of anti-tumor compounds targeting the PCID2 protein are screened out by a molecular docking technology, and the compounds can obviously inhibit the proliferation of tumor cells and promote the apoptosis of the tumor cells.

Description

Application of human PCID2 protein in preparation or screening of antitumor drugs and compound with antitumor activity
The present case is the divisional application of former application, former patent number: 2021103641290, respectively; the original patent name is as follows: the application of human PCID2 protein in preparing or screening antitumor drugs and compounds with antitumor activity; the original application date: 2021, 4 months and 3 days.
Technical Field
The invention belongs to the technical field of new accurate cancer medical targets and medicines, and particularly relates to application of human PCID2 protein in preparation or screening of antitumor medicines and a compound with antitumor activity.
Background
The tumor is a disease of cell damage or malignant and unlimited proliferation, wherein liver cancer is one of common malignant tumors, and accounts for the sixth place and the fourth place of the global tumor morbidity. The new liver cancer of our country accounts for more than 50% of the whole world every year. Liver cancer is occult, early symptoms are not obvious, the intrahepatic and extrahepatic metastasis rate is high, and the prognosis is very poor. Tumor chemotherapy is currently the primary means of treating malignant tumors. However, drug resistance, especially acquired multidrug resistance, has become a major obstacle to current tumor chemotherapy. Most of tumor chemotherapy drugs applied to clinical application generate therapeutic effects by inducing apoptosis of tumor cells, and thus, an anti-apoptosis pathway is a main mechanism for drug resistance of tumor cells.
Active replication of cellular DNA and rapid cycle of mitosis are the basis for maintaining abnormal proliferation of tumors, and cell cycle-specific targeted therapy is therefore also considered to be the most promising anti-cancer strategy. However, the currently clinically approved drugs have strong toxic and side effects and poor target specificity, so that the application effect of the cell cycle-directed inhibitor in the clinical treatment of tumors is unsatisfactory. In normal cells, DNA synthesis replication occurs at G0/G1 and S phase, but the intense DNA replication stress of tumor cells can abnormally activate Chromosome Fragile Sites (CFSs), causing DNA replication to occur at various nodes of the cell cycle, especially M phase, to maintain genome stability and quality, which is a unique feature and defect of tumor cell DNA replication. If a key target point for coordinating the DNA replication pressure can be found, the stability of the tumor cell DNA can be thoroughly destroyed, and then the cell proliferation is fundamentally inhibited. Dynamic new therapies based on specific targeting of tumor cell DNA synthesis hold promise to improve the therapeutic outcome of tumors, and the development of corresponding cell cycle specific multi-target natural small molecule inhibitors has created promise for clinical treatment of tumors.
The existing antitumor chemotherapeutic drugs still have defects in treatment due to strong toxic and side effects and poor specificity and targeting. In the field of liver cancer treatment, although sorafenib, sovitinib and regorafenib have certain curative effects on the treatment of advanced liver cancer, each has defects. In the future, the development of the antagonist of the multifunctional molecule/pathway targeting the growth specificity of cells such as HIF-1 alpha, RAAS and the like is of great significance for improving the curative effect of liver cancer and prolonging the prognosis.
The natural medicine and the derivatives thereof have broad-spectrum biological characteristics in the treatment of serious diseases such as tumor and the like, have low toxic and side effects, and have obvious advantages in the research and development of tumor-specific target antagonists. Therefore, the development of multi-target natural small molecule inhibitors is a promising direction for the targeting treatment of tumor molecules, but the structural optimization of the high-efficiency antagonist is not started due to the lack of specific functional targets.
PCID2 is a key transporter that regulates cell cycle checkpoints. The PCID2 gene is found to promote the proliferation of tumor cells and regulate the cell cycle, is a key molecule for regulating the occurrence and development of tumors, can be used as a target protein for screening antitumor drugs, and is used for screening the antitumor drugs; meanwhile, the invention screens out a kind of anti-tumor compounds targeting PCID2 molecules by a molecular docking technology, and the anti-tumor compounds can obviously inhibit tumor cell proliferation and promote tumor cell apoptosis and have obvious anti-tumor activity.
Disclosure of Invention
The invention provides a compound shown as a formula (I) or a pharmaceutically acceptable salt thereof,
Figure BDA0003478981180000021
wherein, R is1Hydrogen and hydroxyl; r2Hydrogen, methyl, methoxy, halogen and nitro.
Preferably, the halogen is chlorine.
In a fifth aspect, the invention provides an application of a compound shown as a formula (I) or a pharmaceutically acceptable salt thereof in preparing an anti-tumor medicament.
Preferably, the tumor is a tumor of the digestive system.
Preferably, the tumor is liver cancer.
Preferably, the compound or the pharmaceutically acceptable salt thereof is added with a pharmaceutically acceptable carrier and/or auxiliary materials, and can be prepared into any one dosage form of tablets, capsules, granules, powder, pills, tinctures, vinum, soft extracts and mixtures.
The invention has the beneficial effects that: the invention screens out a kind of antitumor drugs targeting human PCID2 protein by molecular docking technology, and the drugs can obviously inhibit liver cancer cell proliferation, promote liver cancer cell apoptosis and have the activity of obviously inhibiting liver cancer.
Drawings
FIG. 1 is a schematic representation of the PCID2 domain and PCID2-DSS1 crystal structure; wherein, A is a schematic diagram of a PCID2 structural domain, and B is a schematic diagram of a crystal structure of PCID2-DSS 1;
FIG. 2 shows the binding between chalcone compound 11 and PCID2 protein obtained by molecular docking;
FIG. 3 shows the inhibition of hepatoma cells by Compound 11 and Compound 1;
FIG. 4 shows the results of detecting the PCID2 protein expression levels in the liver cancer cells SMCC7721, MHCC97H, Huh7 and normal liver cell L02 after the drug intervention of the compound 11; wherein the DMCC is compound 11;
FIG. 5 shows the real-time unmarked results of the pharmacodynamic analysis anaplerosis experiment RTCA before and after 24h of the compound 11 in the liver cancer cell Huh7 cell; wherein OE represents PCID2 overexpression.
Detailed Description
The invention provides an application of a PCID2 gene and/or a PCID2 protein in preparing a medicament for treating tumors. In order to make the technical means, the creation characteristics, the achievement purposes and the effects of the invention easy to understand, the invention is further described with the specific embodiments. The scope of the invention is not limited to the examples described below.
The sources of reagents, vectors, cells described in the following examples are as follows:
liver cancer cells SMCC7721, Huh7, MHCC97H, HCCLM3, Hep3B, PLC, Bel-7404, Bel-7402 and HEK-293T cells were from Shanghai institute of bioscience for Biological Sciences Cell Resource Center; the vectors Ubi-MCS-3FLAG-SV40-EGFP-IRES-puromycin, hU6-MCS-CMV-EGFP, plas-043-pBiFC-YC-hGdown1, plas-044-pBiFC-VN-hGdown1 were from Shanghai Genechem Co., Ltd. Other reagents not described are of conventional origin.
Example 1 antitumor Compounds and tumor inhibition experiments
1. Screening for anti-tumor Compounds
(1) Determination of the Crystal Structure of the PCID2 Domain and the PCID2-DSS1 Complex
Human PCID2 binds to disordered protein DSS1(26S proteosome complex subunit, 26S proteasome complex subunit) (PCID 2-DSS1 complex for short) to maintain the PCID2 protein structure stable. The crystal structures of the PCID2 domain and the PCID2-DSS1 complex were determined in this example, and the results are shown in FIG. 1, in which the WH (wired helix) domain is the main interaction site and is one of the important functional regions of the PCID2 protein.
(2) Preparation of virtual Compound libraries
The chalcone derivative is designed by a biological electronic isostere, a similarity structure transition technology and other methods at three substitution positions of a chalcone matrix by using a shape similarity method (such as ROCS) and a fingerprint similarity method and screening from an existing small molecule database (such as SPECS, Chemdiv, TCMD and the like). In order to ensure the druggability of the target compound, the drug-like property evaluation is carried out on the analog molecule library.
(3) Virtual screening based on molecular docking:
first adopt
Figure BDA0003478981180000031
Glide molds in softwareThe block was subjected to a molecular docking method to analyze the interaction of compound 11 with PCID2, including hydrogen bonding, van der waals interactions, electrostatic interactions, etc., to obtain a binding mode of the two interactions. Then, compounds in the analogue molecule library are butted to an active site of PCID2, and screening is carried out by sequentially carrying out butting methods with different accuracies. And finally, performing cluster analysis on the result of the virtual screening, and selecting target molecules (compounds 1-12) which have stronger binding effect with the PCID2 theoretically according to the joint score value and the experience.
2. Chemical synthesis of target molecules
(1) Synthesis method
The target compound was synthesized in the references (J Med Chem,2015,58, 1795-: firstly, 2, 4-dihydroxyacetophenone (a) or 2,4, 6-trihydroxyacetophenone is taken as a raw material, an intermediate 2, 4-dimethoxyacetophenone (b) or 2-hydroxy-4, 6-dimethoxyacetophenone is obtained through methylation, and secondly, the target molecules and the analogues thereof are obtained through condensation reaction of the 2, 4-dimethoxyacetophenone or 2-hydroxy-4, 6-dimethoxyacetophenone and different substituted benzaldehydes.
Figure BDA0003478981180000041
(2) Structural characterization of drug molecules
After obtaining the target molecule, characterizing and identifying the structure and purity of all drug molecules by using Nuclear Magnetism (NMR), Mass Spectrum (MS), High Performance Liquid Chromatography (HPLC), and the like, wherein the nuclear magnetism data are shown as follows:
compound 1
(E)-1-(2-hydroxy-4,6-dimethoxyphenyl)-3-(2-methoxyphenyl)prop-2-en-1-one:1H NMR(400MHz,CDCl3)δ14.43(s,1H),8.19(d,J=16Hz,1H),8.01(d,J=15.6Hz,1H),7.65(dd,J=8,1.6Hz,1H),7.40(m,1H),7.03(t,1H),6.97(d,J=8.4Hz,1H),6.14(d,J=2.4Hz,1H),5.99(d,J=2.4Hz,1H),3.94(s,3H),3.94(s,3H),3.86(s,3H);13C NMR(100MHz,CDCl3)δ193.00,168.30,166.00,162.47,158.59,37.77,131.29,128.71,127.82,124.50,120.63,111.14,106.38,93.74,91.11,55.68,55.47,55.42。
Compound 2
(E)-1-(2-hydroxy-4,6-dimethoxyphenyl)-3-(3-methoxyphenyl)prop-2-en-1-one:1H NMR(400MHz,CDCl3)δ14.27(s,1H),7.90(d,J=15.6Hz,1H),7.76(d,J=15.6Hz,1H),7.35(t,1H),7.22(d,J=7.6Hz,1H),7.13(d,J=2Hz,1H),6.95(dd,J=5.6,2Hz,1H),6.12(d,J=2.4Hz,1H),5.97(d,J=2Hz,1H),3.92(s,3H),3.86(s,3H),3.84(s,3H);13C NMR(100MHz,CDCl3)δ192.49,168.33,,166.20,162.44,159.80,142.04,136.90,129.76,127.80,120.81,115.51,113.56,106.24,93.75,91.16,55.74,55.47,55.18。
Compound 3
(E)-1-(2-hydroxy-4,6-dimethoxyphenyl)-3-(4-methoxyphenyl)prop-2-en-1-one:1H NMR(400MHz,CDCl3)δ14.41(s,1H),7.84(d,J=16Hz,1H),7.80(d,J=16Hz,1H),7.58(dd,J=6.8,1.6Hz,2H),6.94(d,J=8.8Hz,2H),6.13(d,J=1.2Hz,1H),5.97(d,J=2Hz,1H),,3.92(s,3H),3.85(s,3H),3.84(s,3H);13C NMR(100MHz,CDCl3)δ192.51,168.31,165.96,162.40,161.30,142.38,130.04,128.23,125.05,114.29,106.27,93.76,91.13,55.75,55.48,55.31。
Compound 4
(E)-3-(2-chlorophenyl)-1-(2-hydroxy-4,6-dimethoxyphenyl)prop-2-en-1-one:1H NMR(400MHz,CDCl3)δ14.20(s,1H),8.17(d,J=15.6Hz,1H),7.90(d,J=15.6Hz,1H),7.71(t,1H),7.45(m,1H),7.32(dd,J=4.4,2.4Hz,2H),6.12(d,J=2.4Hz,1H),5.97(d,J=2.4Hz,1H),3.91(s,3H),3.85(s,3H);13C NMR(100MHz,CDCl3)δ192.34,168.48,166.42,162.49,137.89,135.39,133.84,130.71,130.27,130.03,127.81,127.01,126.92,93.80,91.31,55.90,55.65。
Compound 5
(E)-3-(3-chlorophenyl)-1-(2-hydroxy-4,6-dimethoxyphenyl)prop-2-en-1-one:1H NMR(400MHz,CDCl3)δ14.18(s,1H),7.89(d,J=15.6Hz,1H),7.70(d,J=15.6Hz,1H),7.57(s,1H),7.31(m,1H),7.20(dd,J=4,0.8Hz,2H),6.12(d,J=2.4Hz,1H),5.97(d,J=2.4Hz,1H),3.93(s,3H),3.85(s,3H);13C NMR(100MHz,CDCl3)δ192.21,168.42,166.42,162.47,140.37,137.44,134.80,130.06,129.77,128.85,127.83,126.55,106.25,93.79,91.29,55.89,55.58。
Compound 6
(E)-3-(4-chlorophenyl)-1-(2-hydroxy-4,6-dimethoxyphenyl)prop-2-en-1-one:1H NMR(400MHz,CDCl3)δ14.22(s,1H),7.88(d,J=15.6Hz,1H),7.36(d,J=15.6Hz,1H),7.54(d,J=8.4Hz,2H),7.39(d,J=8Hz,2H),6.12(d,J=2.4Hz,1H),5.97(d,J=2.4Hz,1H),3.92(s,3H),3.84(s,3H);13C NMR(100MHz,CDCl3)δ192.28,168.42,166.34,162.44,140.71,135.81,134.06,129.42,129.10,128.01,106.25,93.80,91.28,55.84,55.57。
Compound 7
(E)-1-(2,4-dimethoxyphenyl)-3-(2-methoxyphenyl)prop-2-en-1-one:1H NMR(400MHz,CDCl3)δ8.03(d,J=16Hz,1H),7.76(d,J=8.8Hz,1H),7.61(dd,J=7.6,1.2Hz,1H),7.57(d,J=16Hz,1H),7.37(m,1H),6.99(d,J=7.2Hz,1H),6.95(t,1H),6.57(dd,J=8.8,2.4Hz,1H),6.50(d,J=2.4Hz,1H),3.90(s,3H),3.89(s,3H),3.87(s,3H);13C NMR(100MHz,CDCl3)δ190.93,163.88,160.23,158.5,137.41,132.68,131.09,128.55,127.63,124.38,122.42,120.55,111.07,105.06,98.55,55.60,55.41,55.37。
Compound 8
(E)-1-(2,4-dimethoxyphenyl)-3-(3-methoxyphenyl)prop-2-en-1-one:1H NMR(400MHz,CDCl3)δ7.70(d,J=8.8Hz,1H),7.66(d,J=15.6Hz,1H),7.51(d,J=15.6,1H),7.33(t,1H),7.21(d,J=7.6Hz,1H),7.12(t,1H),6.94(m,1H),6.58(dd,J=8.8,2.4Hz,1H),6.50(d,J=2.4Hz,1H),3.90(s,3H),3.87(s,3H),3.84(s,3H);13C NMR(100MHz,CDCl3)δ190.12,164.07,160.26,159.65,141.57,136.66,132.63,129.60,127.32,121.86,120.66,115.35,113.26,105.13,98.37,55.50,55.30,55.03。
Compound 9
(E)-1-(2,4-dimethoxyphenyl)-3-(4-methoxyphenyl)prop-2-en-1-one:1H NMR(400MHz,CDCl3)δ7.76(d,J=8.4Hz,1H),7.68(d,J=15.6Hz,1H),7.57(d,J=8.4,2H),7.41(d,J=15.6Hz,1H),6.39(d,J=8.8Hz,2H),6.59(dd,J=8.8,2.4Hz,1H),6.51(d,J=2Hz,1H),3.91(s,3H),3.88(s,3H),3.85(s,3H);13C NMR(100MHz,CDCl3)δ190.56,163.90,161.17,160.19,141.97,132.63,129.90,128.09,124.95,122.39,114.22,105.06,98.6,55.67,55.45,55.29。
Compound 10
(E)-3-(2-chlorophenyl)-1-(2,4-dimethoxyphenyl)prop-2-en-1-one:1H NMR(400MHz,CDCl3)δ8.06(d,J=15.6Hz,1H),7.79(d,J=8.4Hz,1H),7.71(m,1H),7.51(d,J=16Hz,1H),7.43(m,1H),7.30(m,2H),6.59(dd,J=8.8,2.4Hz,1H),6.50(d,J=2.4Hz,1H),3.91(s,3H),3.88(s,3H);13C NMR(100MHz,CDCl3)δ190.09,164.31,160.43,137.60,135.21,133.74,132.97,130.53,130.09,129.61,127.67,126.90,121.85,105.27,98.54,55.67,55.49。
Compound 11
(E)-3-(3-chlorophenyl)-1-(2,4-dimethoxyphenyl)prop-2-en-1-one:1H NMR(400MHz,CDCl3)δ7.79(d,J=8.8Hz,1H),7.62(t,2H),7.53(d,J=16Hz,1H),7.47(dd,J=8,1.6Hz,1H),7.34(t,2H),6.59(dd,J=8.8,2.4Hz,1H),6.51(d,J=2.4Hz,1H),3.92(s,3H),3.88(s,3H);13CNMR(100MHz,CDCl3)δ189.77,164.35,160.45,139.89,137.28,134.63,132.87,129.94,129.58,128.30,127.69,126.41,121.72,105.29,98.45,55.65,55.43。
Compound 12
(E)-3-(4-chlorophenyl)-1-(2,4-dimethoxyphenyl)prop-2-en-1-one:1H NMR(400MHz,CDCl3)δ7.78(d,J=8.8Hz,1H),7.64(d,J=15.6Hz,1H),7.54(t,3H),7.37(d,J=8.8Hz,2H),6.59(dd,J=8.4,2Hz,1H),6.50(d,J=2.4Hz,1H),3.91(s,3H),3.88(s,3H);13C NMR(100MHz,CDCl3)δ189.92,164.29,160.41,140.22,135.6,133.95,132.88,129.33,128.98,127.58,121.90,105.26,98.53,55.67,55.46。
The structural formula of the obtained compound is shown as the following formula 1-12:
Figure BDA0003478981180000071
3. cell viability assay
Cell viability was measured using a Cell Counting Kit (CCK-8) (Dojindo). Cells in the log phase were trypsinized and counted according to the instructions provided with the kit, and cells were seeded in 96-well plates (150 μ L/well) at the appropriate concentration, with 6 duplicate wells per group. After conventional culture is carried out for 24h, 48h, 72h and 96h respectively, the original culture medium is sucked dry, and 110 mu L/hole CCK-8 solution detection mixed solution is added into the culture plate (10 mu L CCK-8 solution is added into each 100 mu L complete culture medium); and (3) after the culture plate is incubated in a cell temperature incubator for 2h, detecting the OD value at 450nm by using an enzyme-labeling instrument, and drawing a cell proliferation curve according to the detection result. The compound is used for intervening liver cancer cells, and the cell activity is detected through a CCK8 cell activity experiment; and detecting the expression level of PCID2 in the liver cancer cells after the intervention of the compounds by Western blot.
Cell activity experiments show that the compounds 1-12 have half of fatality rate IC to liver cancer cells50As shown in table 1 below; wherein the compound 11(3- (3-chlorphenyl) -1- (2, 4-dimethoxyphenyl) -2-propylene-1-ketone, ((E) -3- (3-chlorophenyl) -1- (2, 4-dimethoxyphenyl) prop-2-en-1-one)) has the strongest effect on liver cancer cells and half of fatality rate IC (integrated circuit) on the liver cancer cells50It was 9.286. mu. mol/L. And its interaction with PCID2 is strongest as shown in fig. 2.
TABLE 1 half-lethality of Compounds 1-12 to hepatoma cells
Compound numbering 1 2 3 4 5 6
IC50(μmol/L) ND 39.34 450.9 ND 30.49 39.42
Compound numbering 7 8 9 10 11 12
IC50(μmol/L) ND 10.73 ND ND 9.286 ND
Note: ND means not detected
The screened compounds 1-12 can inhibit the proliferation of liver cancer cells, wherein the inhibition activity of the compound 1 on the proliferation of the liver cancer cells is weaker, and the compound 11 has the strongest inhibition activity on the proliferation of the liver cancer cells. The results of the cell proliferation experiments are shown in fig. 3, which provides a result graph of the inhibitory activity of only compounds 1 and 11 on the proliferation of liver cancer cells.
4. Effect of Compound 11 drug intervention on hepatoma cell proliferation
(1) Influence of compound 11 drug intervention on expression level of PCID2 in hepatoma cells
Performing compound 11 drug-stem prognosis on liver cancer cells SMCC7721, MHCC97H, Huh7 cell lines and normal liver cells L02, and detecting the expression level of the PCID2 protein of the liver cancer cells by Western blot.
The experimental result is shown in fig. 4, compared with the solvent control group (DMSO), the expression level of the PCID2 protein of the liver cancer cell is significantly reduced after the compound 11 drug is dried, and the expression of the PCID2 of the normal liver cell line L02 cell is not significantly affected. The result shows that the compound 11 can obviously inhibit the expression of the gene PCID2 in the liver cancer cell and has an influence on the gene PCID2 of a normal liver cell.
(2) After gene PCID2 is over-expressed, compound 11 medicine intervenes the influence on liver cancer cell proliferation
The liver cancer Huh7 cell line was divided into a blank group (Huh7-NC), a compound 11 drug intervention group (Huh7-11#), a solvent control group (Huh7-DMSO) and a PCID2 overexpression compound 11 drug intervention group (Huh7-OE-11 #). RTCA real-time label-free detection of the growth of the liver cancer Huh7 cell line.
The results of the RTCA real-time unmarked experiment are shown in FIG. 5, and before drug intervention (before 24 h), the PCID2 overexpression can remarkably promote the growth of liver cancer cells; after drug intervention (after 24 h), the proliferation capacity of liver cancer cells in the compound 11 drug intervention group (Huh7-11#) and the PCID2 overexpression compound 11 drug intervention group (Huh7-OE-11#) was significantly inhibited. The result shows that the compound 11 can obviously inhibit the proliferation of normal liver cancer cells and also can obviously inhibit the proliferation of the liver cancer cells over-expressed by PCID2, which indicates that the compound 11 inhibits the proliferation of the liver cancer cells by inhibiting PCID 2.
The above description is only for details of a specific exemplary embodiment of the present invention, and it is obvious to those skilled in the art that various modifications and changes may be made in the present invention in the practical application process according to specific preparation conditions, and the present invention is not limited thereto. All that comes within the spirit and principle of the invention is to be understood as being within the scope of the invention.

Claims (5)

1. A compound of formula (I) or a pharmaceutically acceptable salt thereof,
Figure FDA0003478981170000011
wherein, R is1Hydrogen and hydroxyl; r2Hydrogen, methyl, methoxy, halogen and nitro.
2. Use of a compound according to claim 1 or a pharmaceutically acceptable salt thereof in the preparation of an anti-tumor medicament.
3. The use of claim 2, wherein the tumor is a tumor of the digestive system.
4. The use of claim 3, wherein the tumor is liver cancer.
5. The compound or the pharmaceutically acceptable salt thereof according to claim 1, wherein the compound or the pharmaceutically acceptable salt thereof is added with a pharmaceutically acceptable carrier and/or an auxiliary material, and can be prepared into any dosage form of tablets, capsules, granules, powder, pills, tinctures, vinum, soft extracts and mixtures.
CN202210062797.2A 2021-04-03 2021-04-03 Application of human PCID2 protein in preparation or screening of antitumor drugs and compound with antitumor activity Pending CN114426470A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210062797.2A CN114426470A (en) 2021-04-03 2021-04-03 Application of human PCID2 protein in preparation or screening of antitumor drugs and compound with antitumor activity

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210062797.2A CN114426470A (en) 2021-04-03 2021-04-03 Application of human PCID2 protein in preparation or screening of antitumor drugs and compound with antitumor activity
CN202110364129.0A CN112870363B (en) 2021-04-03 2021-04-03 Application of human PCID2 protein in preparation or screening of antitumor drugs and compound with antitumor activity

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN202110364129.0A Division CN112870363B (en) 2021-04-03 2021-04-03 Application of human PCID2 protein in preparation or screening of antitumor drugs and compound with antitumor activity

Publications (1)

Publication Number Publication Date
CN114426470A true CN114426470A (en) 2022-05-03

Family

ID=76040443

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202110364129.0A Active CN112870363B (en) 2021-04-03 2021-04-03 Application of human PCID2 protein in preparation or screening of antitumor drugs and compound with antitumor activity
CN202210062797.2A Pending CN114426470A (en) 2021-04-03 2021-04-03 Application of human PCID2 protein in preparation or screening of antitumor drugs and compound with antitumor activity

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN202110364129.0A Active CN112870363B (en) 2021-04-03 2021-04-03 Application of human PCID2 protein in preparation or screening of antitumor drugs and compound with antitumor activity

Country Status (1)

Country Link
CN (2) CN112870363B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115019881A (en) * 2022-07-15 2022-09-06 普瑞基准科技(北京)有限公司 Small molecule anti-tumor effect identification method and system based on gene protein activity

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6323136A (en) * 1986-03-14 1988-01-30 Seizo Miyata Nonlinear optical element
CN106102758A (en) * 2013-11-11 2016-11-09 夸利蒂赫布丘蒂克斯公司 Therapeutic compound that slips is derivative and using method thereof
US20180289682A1 (en) * 2015-10-08 2018-10-11 The Regents Of The University Of California Compounds and methods for promoting stress resistance
CN110028475A (en) * 2019-05-13 2019-07-19 中国药科大学 Novel C DK9 inhibitor, preparation method and application based on benzofuran structure
CN111269104A (en) * 2020-01-23 2020-06-12 温州医科大学 Chalcone analogue and application thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008111464A1 (en) * 2007-03-05 2008-09-18 Eisai R & D Management Co., Ltd. Method for examination of action of anti-cancer agent utilizing splicing defect as measure
CN103215292B (en) * 2012-01-18 2015-10-07 中国科学院生物物理研究所 The hybridoma cell line of the solubility expression of people Pcid2 albumen and the monoclonal antibody 2D7-F11 of anti-human Pcid2 albumen and this antibody of secretion
US20190033306A1 (en) * 2014-05-13 2019-01-31 The University Of Chicago Recurrent fusion genes in human cancers

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6323136A (en) * 1986-03-14 1988-01-30 Seizo Miyata Nonlinear optical element
CN106102758A (en) * 2013-11-11 2016-11-09 夸利蒂赫布丘蒂克斯公司 Therapeutic compound that slips is derivative and using method thereof
US20180289682A1 (en) * 2015-10-08 2018-10-11 The Regents Of The University Of California Compounds and methods for promoting stress resistance
CN110028475A (en) * 2019-05-13 2019-07-19 中国药科大学 Novel C DK9 inhibitor, preparation method and application based on benzofuran structure
CN111269104A (en) * 2020-01-23 2020-06-12 温州医科大学 Chalcone analogue and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CA: "RN:1663469-39-3等", REGISTRY(STN), 17 March 2015 (2015-03-17), pages 1 - 46 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115019881A (en) * 2022-07-15 2022-09-06 普瑞基准科技(北京)有限公司 Small molecule anti-tumor effect identification method and system based on gene protein activity
CN115019881B (en) * 2022-07-15 2022-10-21 普瑞基准科技(北京)有限公司 Small molecule anti-tumor effect identification method and system based on gene protein activity

Also Published As

Publication number Publication date
CN112870363A (en) 2021-06-01
CN112870363B (en) 2022-02-18

Similar Documents

Publication Publication Date Title
Chen et al. Computational discovery of niclosamide ethanolamine, a repurposed drug candidate that reduces growth of hepatocellular carcinoma cells in vitro and in mice by inhibiting cell division cycle 37 signaling
Liu et al. Identifying the antiproliferative effect of astragalus polysaccharides on breast cancer: coupling network pharmacology with targetable screening from the cancer genome atlas
Liu et al. The modulatory properties of Astragalus membranaceus treatment on triple-negative breast cancer: an integrated pharmacological method
Fan et al. Integrating network pharmacology deciphers the action mechanism of Zuojin capsule in suppressing colorectal cancer
Govindarasu et al. In silico modeling and molecular docking insights of kaempferitrin for colon cancer-related molecular targets
Xing et al. Identification of key candidate genes and pathways in hepatocellular carcinoma by integrated bioinformatical analysis
Yang et al. Network pharmacology reveals polyphyllin II as one hit of nano Chinese medicine monomers against nasopharyngeal carcinoma
Baek et al. Identification of a novel anticancer mechanism of Paeoniae Radix extracts based on systematic transcriptome analysis
CN114426470A (en) Application of human PCID2 protein in preparation or screening of antitumor drugs and compound with antitumor activity
Yue et al. Focus on the molecular mechanisms of cisplatin resistance based on multi-omics approaches
Qi et al. Investigating the mechanism of scutellariae barbata herba in the treatment of colorectal cancer by network pharmacology and molecular docking
Liu et al. Gene profiling of HepG2 cells following nitidine chloride treatment: An investigation with microarray and Connectivity Mapping
Zhang et al. Molecular docking and in vitro experiments verified that kaempferol induced apoptosis and inhibited human HepG2 cell proliferation by targeting BAX, CDK1, and JUN
Dai et al. Study on the molecular mechanism of the herbal couple Sparganii Rhizoma-Curcumae Rhizoma in the treatment of lung cancer based on network pharmacology
Pang et al. Competing endogenous RNA and coexpression network analysis for identification of potential biomarkers and therapeutics in association with metastasis risk and progression of prostate cancer
Solanki et al. Combined transcriptomics and in-silico approach uncovers the role of prognostic biomarkers in hepatocellular carcinoma
Xie et al. Design, synthesis, and biological evaluation of novel EF24 and EF31 analogs as potential IκB kinase β inhibitors for the treatment of pancreatic cancer
Lu et al. Identification of key genes in hepatocellular carcinoma and validation of the candidate gene, cdc25a, using gene set enrichment analysis, meta‑analysis and cross‑species comparison
Su et al. Biological evaluation and molecular docking of Rhein as a multi-targeted radiotherapy sensitization agent of nasopharyngeal carcinoma
Joshi et al. In silico ADME/pharmacokinetic and target prediction studies of ethambutol as drug molecule
Guo et al. Network pharmacological study of compound kushen injection in esophageal cancer
Poleboyina et al. Homology Modeling, Screening, and Identification of Potential FOXO6 Inhibitors Curtail Gastric Cancer Progression: an In Silico Drug Repurposing Approach
Nair et al. Application of comprehensive bioinformatics approaches to reconnoiter crucial genes and pathways underpinning hepatocellular carcinoma: a drug repurposing endeavor
Ahmed et al. Structure-guided design and development of vanillin-triazole conjugates as potential MARK4 inhibitors targeting hepatocellular carcinoma
Yang et al. Small-molecule drugs of colorectal cancer: Current status and future directions

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