CN115737619A - Drug target for inhibiting tumor, application and oral drug - Google Patents

Drug target for inhibiting tumor, application and oral drug Download PDF

Info

Publication number
CN115737619A
CN115737619A CN202211457404.4A CN202211457404A CN115737619A CN 115737619 A CN115737619 A CN 115737619A CN 202211457404 A CN202211457404 A CN 202211457404A CN 115737619 A CN115737619 A CN 115737619A
Authority
CN
China
Prior art keywords
cancer
drug target
tumor
drug
translation initiation
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
CN202211457404.4A
Other languages
Chinese (zh)
Inventor
张弓
陈洋
余卓
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.)
Shenzhen Chi Biotech Co ltd
Original Assignee
Shenzhen Chi Biotech Co ltd
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 Shenzhen Chi Biotech Co ltd filed Critical Shenzhen Chi Biotech Co ltd
Priority to CN202211457404.4A priority Critical patent/CN115737619A/en
Priority to PCT/CN2022/138169 priority patent/WO2024108676A1/en
Priority to AU2022472191A priority patent/AU2022472191A1/en
Publication of CN115737619A publication Critical patent/CN115737619A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/194Carboxylic acids, e.g. valproic acid having two or more carboxyl groups, e.g. succinic, maleic or phthalic acid
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The application relates to the field of targeted drug development, in particular to a drug target for inhibiting tumor and application thereof. Specifically, a drug target for inhibiting tumors is provided, and the drug target is eukaryotic cell translation initiation factor EIF2. The invention also provides application of the drug target for inhibiting tumor and an oral drug. By inhibiting translation initiation factor EIF2 to inhibit translation initiation of eukaryotic cells, the malignant phenotype effect of cancer cells can be safely suppressed, so that at least the following safety is realized: 1. unlike other common translation initiation inhibitors (such as CHX and HTT), targeted inhibition of EIF2 does not occupy ribosome resources and is capable of competitively binding ribosomes; 2. the ISR of cells is not caused, and the toxic and side effects on normal cells can be reduced; can be administered orally safely.

Description

Drug target for inhibiting tumor, application and oral drug
Technical Field
The application relates to the field of targeted drug development, in particular to a drug target for inhibiting tumor, application and an oral drug.
Background
The inhibition of the translation of the cancer cells can effectively suppress the growth of the cancer cells, and has a drug development value. However, inhibition of the translation process may lead to severe cytotoxicity, mainly due to: the translation inhibitor occupies ribosome resources, causes cell stress, protein folding disorder and the like, and the factors can influence the clinical application prospect of the translation inhibition related medicine.
In some related technologies, small molecule compounds or mimetics that inhibit a single translation initiation factor have been developed to affect the enzymatic activity, functional structure, modification abundance, etc., thereby achieving the purpose of inhibiting translation initiation. For example, elatol, SAN and 15d-PGJ2, which have been developed to date, inhibit EIF4A1 (PMID: 32014999). However, such inhibitors are likely to cause cell Integrated Stress Response (ISR) (PMID: 32014999). ISR is an adaptive cellular mechanism that nonspecifically inhibits overall protein synthesis in cells, allowing cells to respond to stress and survive (PMID: 27629041).
Central to this pathway is phosphorylation of eIF2 α, which serves to inhibit overall protein synthesis while allowing expression of selected mrnas. Although ISR is primarily a homeostatic process that promotes survival, exposure to severe stress drives signaling to cell death resulting in cytotoxicity. For example, paclitaxel treatment has been reported to induce ISR to provide a survival advantage for cancer in vivo in breast cancer therapy (PMID: 31211507). The reason for this is that ISR can translate into IRES-dependent uarf proteins, such as oncogenes SOX2, MYC, HER2, etc. which can be preferentially translated under ISR induction, making cancer cells resistant to stress, resulting in cancer resistance and recurrence.
Therefore, there is still a great need in the art to find suitable drug targets to achieve accurate targeted therapy of cancer.
Disclosure of Invention
In view of the above, the present application aims to provide a drug target capable of effectively inhibiting tumors, especially malignant phenotypes such as cancer cell proliferation, metastasis and neoplasia, so as to achieve accurate targeted therapy of cancer while avoiding side effects.
In a first aspect, a drug target for inhibiting tumors is provided, which is eukaryotic translation initiation factor EIF2.EIF2 is a protein complex composed of a plurality of constituent proteins, the structure of which is found, for example, in Tomas Adomavicius et al ("The structural basis of translational control by eIF2 phosphorylation", nature Communication, may 13,2019, article number 2136 (2019), https:// www. Natural. Communications/019 41467-10167-3). In some embodiments, the drug target is subunit EIF2S1 of initiation factor EIF2. For example, the protein sequence of EIF2S1 can be found in the NCBI' S library in RefSeq ID: NP-004085. In some embodiments, the drug is aurintricarboxylic acid (ATCA) or an ammonium salt thereof.
By inhibiting the translation initiation factor EIF2, the phosphorylation of the protein can be effectively reduced, thereby inhibiting ISR. Meanwhile, the inhibition of the translation initiation factor EIF2 can prevent ribosome from carrying out translation initiation assembly, does not consume ribosome resources, is not easy to generate various stress reactions in cells, and thus, the effect of safely inhibiting cancer cells is achieved
In some embodiments, the tumor is a malignant tumor selected from the group consisting of: malignant epithelial tumors, sarcomas, myelomas, leukemias, lymphomas, melanomas, head and neck tumors, brain tumors, peritoneal carcinomas, mixed tumors, and childhood malignancies.
In some further embodiments, the malignant epithelial tumors are selected from the group consisting of: lung cancer, breast cancer, liver cancer, pancreatic cancer, colorectal cancer, stomach cancer, gastroesophageal adenocarcinoma, esophageal cancer, small intestine cancer, cardiac cancer, endometrial cancer, ovarian cancer, fallopian tube cancer, vulva cancer, testicular cancer, prostate cancer, penile cancer, kidney cancer, bladder cancer, anal cancer, gallbladder cancer, bile duct cancer, teratoma, and heart tumor.
In some embodiments, the tumor is lung cancer, preferably non-small cell lung cancer.
In some embodiments, the tumor is ovarian cancer, preferably ovarian epithelial cancer.
In some embodiments, the tumor is liver cancer.
In a second aspect, there is provided the use of eukaryotic translation initiation factor EIF2 as a pharmaceutical target for inhibiting tumors.
In some embodiments, the drug is aurintricarboxylic acid (ATCA) or an ammonium salt thereof.
In a third aspect, there is provided the use of a substance that inhibits the eukaryotic translation initiation factor EIF2 in the manufacture of a medicament for the treatment of a tumour.
In some embodiments, the substance is aurintricarboxylic acid (ATCA) or an ammonium salt thereof.
In a fourth aspect, an oral medicament for inhibiting a tumor is provided, comprising an inhibitor capable of inhibiting the translation initiation factor EIF2 of a drug target eukaryotic cell without causing phosphorylation. In some embodiments, the inhibitor is aurintricarboxylic acid (ATCA) or an ammonium salt thereof.
In summary, the present invention can achieve the following advantageous technical effects.
By inhibiting translation initiation factor EIF2 to inhibit translation initiation of eukaryotic cells, the malignant phenotype effect of cancer cells can be safely suppressed, so that at least the following safety is realized:
1. unlike other common translation initiation inhibitors (such as CHX and HTT), targeted inhibition of EIF2 does not occupy ribosome resources and is capable of competitively binding ribosomes; for example, CHX not only inhibits translation initiation but also inhibits translation elongation, thus provoking more stress and greater side effects;
2. the ISR of cells is not caused, and the toxic and side effects on normal cells can be reduced;
3. can be administered orally safely.
Drawings
Figure 1 is a graph of an ATCA-EIF 2S1 binding thermal transfer protein immunoblot.
Figure 2 is a schematic diagram of molecular docking to evaluate binding energy and interaction pattern of ATCA with its target protein EIF2S1.
FIG. 3 is a graph showing the results of the EIF2A phosphoprotein immunoblot assay, wherein the left lane is a control and the right lane is the results after 48 hours of treatment with 1mM ATCA.
FIG. 4 is an electrophoretogram showing the downregulation of SOX2 and HER2 after ATCA treatment.
FIG. 5 is a graph showing the results of a polysome analysis experiment.
FIG. 6 is a Western Blot immunoblot for the detection of nascent peptides.
Figure 7 shows ATCA inhibits cancer malignancy phenotype-associated signaling pathways, specifically showing KEGG pathway enrichment results for downregulated genes after 48 hours of drug treatment.
Detailed Description
The present application will be described in further detail with reference to specific examples.
The inventors of the present application have conducted extensive studies in order to develop a drug capable of effectively inhibiting the translation of cancer cells. As a result, it has unexpectedly been found that by inhibiting the translation initiation factor EIF2, its phosphorylation can be effectively reduced, thereby inhibiting ISR. Meanwhile, the inhibition of the translation initiation factor EIF2 can prevent ribosome from carrying out translation initiation assembly, does not consume ribosome resources, and is not easy to generate various stress reactions in cells, thereby achieving the effect of safely inhibiting cancer cells.
Meanwhile, the inventors of the present application found in the course of their studies that aurintricarboxylic acid (ATCA) can target subunit EIF2S1 that binds to translation initiation factor EIF2, but does not affect its phosphorylation abundance, meaning that ISR is not caused. The expression of SOX2 and HER2 proteins is down-regulated, indicating that the ISR-dependent SOX2 and HER2 small open reading frame translation event (uORF) triggered by EIF2S1 does not occur in the cells. In addition, the ATCA only targets the subunit EIF2S1 combined with the translation initiation factor EIF2, but not combined with the non-target proteins EEF2 and Vincultin.
The present invention has been completed based on the above findings.
Embodiments of the present invention will be described in detail below with reference to specific examples. It will be understood by those of ordinary skill in the art that these examples are provided merely to illustrate some exemplary ways in which the invention may be practiced and are not intended to limit the scope of the invention to these exemplary embodiments.
Example 1: heat transfer immunoblotting CESTA-WB experiment
CESTA-WB experiment: h1299 cells are plated in a 15cm dish for culture, when the density is about 90-100%, the culture medium is sucked away, the dish is washed twice by PBS, 2ml of Buffer (RB Buffer, 10% N-Dodecyl-beta-D-maltoside and 1 Xprotease inhibitor) is added into each dish and is flatly placed on ice for lysis for 30 minutes, cell lysate is collected and is centrifuged at 17000g and 4 ℃ for 10 minutes, and the supernatant is quantified by a BCA kit. 45uL of total protein was taken out of each PCR tube, aurintricarboxylic acid (ATCA) was added to the tubes to a final concentration of 10mM, 1mM, 100uM, 10uM, 1uM, 100nM, 10nM, and 25 ℃ for 30 minutes to allow the ATCA to bind to the protein sufficiently, and the tubes were taken out without ATCA as a control. The experimental group is treated for 3 minutes at 55 ℃, one tube of the control group is treated for 3 minutes at 37 ℃, and the other tube of the control sample is treated for 3 minutes at 50 ℃; centrifuging at the temperature of 4 ℃ for 10 minutes at 17000g, removing denatured proteins, placing the supernatant in a new PCR tube, taking 5ug of the supernatant, adding 5 xSDS Loading Buffer, performing water bath denaturation at the temperature of 100 ℃, and detecting the quantitative trends of EEF2, EIF2S1, beta-actin and Vincultin proteins by using a Western Blot method.
Western Blot experiment: samples and pre-stained protein molecular weight standards were loaded onto SDS-PAGE gels (10cm. Times.10cm) for electrophoresis at 100V,40min.100V,230mA,1h electrotransfer the proteins in SDS-PAGE to nitrocellulose membranes. After membrane transfer, the nitrocellulose membrane was washed with 25ml TBS for 5min at room temperature. Membranes were placed in 25mL of blocking buffer (TBST, 5% skim milk) and incubated for 1 hour at room temperature. Wash three times with 15ml TBST for 5min each. Membranes and EEF2, EIF2S1, β -actin and Vinculin primary antibodies (diluted as 1 1500) were incubated overnight at 4 ℃ in 10mL primary antibody dilution buffer with occasional gentle shaking. Wash three times with 15mL TBST for 5min each. A 5% skim milk was used to formulate 1. Wash three times with 15mL TBST for 5min each. The membrane was mixed with 10mL
Figure BDA0003953795130000043
(0.5ml 20X
Figure BDA0003953795130000042
#7003, 0.5mL 20X peroxide, and 9.0mL purified water) was incubated at room temperature for 1min with occasional gentle agitation. Excess developer solution on the film was drained (not allowed to dry), wrapped in plastic film, and then developed using a BIO-RAD developer, the results of which are shown in FIG. 1.
As can be seen in figure 1, ATCA targets the binding protein EIF2S1 (one of the component proteins of EIF 2) and does not bind the non-target proteins EEF2 and Vinculin.
Example 2 molecular docking analysis: assessment of binding energy and interaction patterns of ATCA with its target protein EIF2S1
To assess the binding energy and interaction pattern of ATCA with its target protein EIF2S1, a protein-ligand docking assay was performed using AutoDock Vina 1.2.2 (PMID: 19499576). The molecular structure of ATCA was first obtained from PubChem compound database (https:// PubChem. Ncbi. Nlm. Nih. Gov /), for example, as shown in the following formula (1).
Figure BDA0003953795130000041
Then, the protein EIF2S1 was downloaded from PDB (https:// www. Rcsb. Org /) (PDB number: 6ybv; resolution:
Figure BDA0003953795130000044
) 3D coordinates of (a). Protein and ligand files were first prepared, and then all protein and molecule files were converted to PDBQI format, with all water molecules removed, and polar amino acid atoms added. The grid box is centered to cover the domains of each protein and accommodate free molecular motion. The butt joint pocket is arranged as one
Figure BDA0003953795130000051
The lattice point distance of the square pocket is as follows: 0.05nm. Molecular docking studies were used by AutoDock Vina 1.2.2 (http:// AutoDock. Script. Edu /) for model visualization. The results of the analysis are shown in FIG. 2.
As can be seen from FIG. 2, the results of molecular docking showed the most stable docking conformation, ATCA can penetrate deep inside the EIF2S1 pocket, forming multiple stable hydrogen bonds with Lys142, pro144, his10, lys96 and Val18 subunits, with a binding energy of-6.7 kcal/mol. This indicates that it forms an effective non-covalent bond and occupies its pocket site, resulting in inhibition of its function and dissociation into competitive bonds. In this way, the inhibition degree of the ATCA on the EIF2S1 can be conveniently adjusted by adjusting the concentration of the ATCA, or the inhibition on the EIF2S1 is released by removing the ATCA, so that flexible control as required is realized.
Example 3EIF2A phosphorylated protein immunoblot assay
Western Blot method: mixing the sample with the prestained protein molecular weightThe standards were loaded on SDS-PAGE gel (10cm. Times.10cm) and electrophoresed at 100V, and reacted for 40min. The treatment was carried out at 100V and 230mA for 1 hour, and then the proteins in the SDS-PAGE were electrically transferred to a nitrocellulose membrane. After membrane transfer, the nitrocellulose membrane was washed with 25ml TBS for 5min at room temperature. Membranes were placed in 25mL of blocking buffer (TBST, 5% skim milk) and incubated for 1 hour at room temperature. Wash three times with 15ml TBST for 5min each time. Membranes and phosphorylated EIF2S1 primary antibody (diluted according to 1 1500) were incubated overnight at 4 ℃ in 10mL primary antibody dilution buffer with periodic gentle shaking. Wash three times with 15mL TBST for 5min each. A 5% skim milk was used to formulate 1. Wash three times with 15mL TBST for 5min each time. The membrane was mixed with 10mL
Figure BDA0003953795130000052
(0.5ml 20X
Figure BDA0003953795130000053
0.5ml of 20X peroxide and 9.0mL of ultrapure water) was incubated at room temperature for 1min with regular gentle agitation. Excess developer solution on the film was drained (no drying), wrapped in a plastic film, and then developed with a BIO-RAD developer.
The results are shown in fig. 3 and 4. As can be seen from fig. 3, ATCA is able to target binding to EIF2S1, but does not affect its phosphorylation abundance, which means that ISR is not caused. As can be seen in fig. 4, SOX2 and HER2 protein expression was down-regulated, indicating that the ISR-dependent SOX2, HER2 small open reading frame translation event (uORF) initiated by EIF2S1 did not occur in the cells.
Example 4 translation initiation efficiency assay and nascent peptide detection in cells
Polysome analysis (Polysome profiling): cells were plated in 75T flasks at 200 million cells per flask and ACTA was added to the experimental groups 24 hours after adherence to the wall to give a working solution concentration of 1mg/ml. The control group was replaced with fresh medium, sampled 24, 48 hours after ATCA treatment, the medium aspirated, PBS washed twice, 2ml lysine Buffer (RB Buffer,1% Tton X-100, 10. Mu.g/. Mu.L cycloheximide) per vial lysed on ice for 30 minutes, and the lysates pipetted into RNase-free EP tubes. Centrifugation was carried out at 17000g for 10 minutes at 4 ℃ and 1.5ml of the supernatant was aspirated into a new EP tube and added to a sucrose density gradient solution (sucrose concentration from top to bottom: 13% -46%, each gradient increasing in concentration by 3%) prepared in advance. The cells were centrifuged at 25400rpm at 4 ℃ for 4 hours and 10 minutes, and then relative quantitative analysis of polysome, monoribosome, and ribosome size subunit was performed by HPLC, and the results are shown in FIG. 5. As can be seen from FIG. 5, the polysome distribution curve did not change much after 24 hours of dosing; after 48 hours of dosing, the polysome fraction decreased significantly, mainly in the form of mononucleosomes. Since ATCA does not affect translation elongation, it is suggested that translation initiation has been slowed effectively, resulting in translation of only one ribosome on most mRNAs.
Detecting nascent peptides: the cells are plated in a 6-well plate, 20 million cells are added into each well, ATCA is added after 24 hours of adherence to ensure that the concentration of the working solution is 1mg/ml, a control group is replaced by new culture solution and is respectively treated for 2,6, 12, 24 and 48 hours, ATCA is not added into the control group, cycloheximide is added into a positive control group for treatment for 10 minutes 25 minutes before sampling, then 10ug/ul puromycin marked nascent peptides are added into all the wells for 15 minutes, the culture medium is sucked away, PBS is washed twice, IP & WB lysate is added for ice lysis for 30 minutes, centrifugation is carried out for 20 minutes at 4 ℃ and 12000g, supernatant is sucked into a new centrifuge tube, 10ug protein is added into a5 xSDS Loading Buffer, water bath denaturation is carried out for 10 minutes at 100 ℃, and quantitative detection is carried out by a Western Blot method.
Western Blot method: samples and pre-stained protein molecular weight standards were loaded onto SDS-PAGE gels (10cm. Times.10cm) for electrophoresis at 100V,40min.100V,230mA,1h electrotransfer of proteins in SDS-PAGE to nitrocellulose membrane. After membrane transfer, the nitrocellulose membrane was washed with 25ml TBS for 5min at room temperature. The membrane was placed in 25mL blocking buffer (TBST, 5% skim milk) and incubated at room temperature for 1 hour. Wash three times with 15ml TBST for 5min each time. Membranes and puromycin primary antibody (diluted as 1 in 1000) were incubated overnight at 4 ℃ in 10mL of primary antibody dilution buffer with occasional gentle shaking. Wash three times with 15mL TBST for 5min each. A 5% skim milk was used to formulate 1. Wash three times with 15mL TBST for 5min each. The membrane was mixed with 10mL
Figure BDA0003953795130000062
(0.5mL 20X
Figure BDA0003953795130000061
0.5ml of 20X peroxide and 9.0mL of ultrapure water) was incubated at room temperature for 1min with occasional gentle agitation. Excess developer on the film was drained (not allowed to dry), wrapped in plastic film, and then developed with a BIO-RAD developer. The results are shown in FIG. 6.
As can be seen in FIG. 6, puromycin covalently binds itself as an "amino acid" to the ends of nascent peptide chains and is detached from the ribosome, and these nascent peptide chains terminated with puromycin are visible with puromycin antibodies. FIG. 6 shows that protein synthesis is not significantly hindered by adding drugs for 2-12 h; after the medicine is added for 24-48h, the newly generated peptide chain is obviously reduced compared with the newly added medicine, and the result proves that the translation initiation inhibition performance of ATCA can effectively reduce the synthesis of protein.
Example 5 transcriptome sequencing
Transcriptome sequencing experiment procedure:
RNA extraction
1mg/mL ATCA was added to the medium of a T75 cell culture flask full of three million H1299 cells as a treatment group, an equal volume of PBS was added as a control group, the culture supernatant was discarded after 48H ATCA treatment, placed on ice, and washed twice with pre-cooled PBS. Discarding PBS, adding 5mL precooled Trizol (total RNA extraction reagent), fully shaking and mixing uniformly, and specifically extracting as follows:
1) Adding 0.2mL of chloroform into every 1000 microliters of Trizol, violently mixing uniformly (vortex) for 15s, standing for 3min at room temperature, and observing that the sample begins to stratify;
2) 12000 Xg, centrifuging for 15min at 4 ℃ to divide the sample into three layers;
3) Carefully pipette the upper aqueous phase, approximately 600. Mu.L, into a new 1.5mL EP tube, taking care not to pipette the middle layer of liquid;
4) Adding 800 μ L isopropanol, inverting, and mixing;
5) Standing at-20 deg.C for 8 hr;
6) After 8h, 12000 Xg, centrifuging for 30min at 4 ℃, and removing supernatant;
7) Adding 1mL of 75% ethanol (precooling for 30min at-20 ℃ in advance);
8) 7500 Xg, centrifuging at 4 deg.C for 5min, and removing supernatant;
9) Repeating steps 7) and 8) once;
10 ) removing residual ethanol, opening the tube cover, and air-drying for 5min;
11 About 30 to 60 microliters of RNase Free pure water was added according to the obtained particle size to dissolve, and this step yielded an RNA standard, which was stored in a-80 ℃ refrigerator.
Enriching Poly-AmRNA:
the sequence was performed according to the standard human poly-AmRNA transcript library and sequencing protocol, and the specific experimental procedures were described in the applicant's published literature (PMID: 23519614, PMID: 30265008).
The procedure used a Novozam poly-A mRNA enrichment kit to enrich for poly-A mRNA. The specific steps are shown in the specification: extracting total poly-A mRNA by adopting a VAHTS mRNA Caputre Beads kit, comprising the following steps:
1) Taking out the mRNA Caputre Beads, and standing to balance the temperature to room temperature;
2) Preparation of RNA samples: diluting 0.01-12.5 μ g of total RNA to 50 μ L with RNase-free water in a nucleic-free PCR tube, and freezing for use;
3) The mRNA Caputre Beads are inverted up and down and are fully and uniformly mixed; sucking 50 mu L of the mixture, adding the mixture into a total RNA sample, and uniformly mixing;
4) Placing the sample in a PCR instrument, and keeping at 65 ℃ for 5min,25 ℃ for 5min and 4 ℃ to enable mRNA to be combined on the magnetic beads;
5) Placing the sample in a magnetic rack for 5min to separate mRNA from total RNA, and removing supernatant;
6) Taking out the sample from the magnetic frame, adding 200 mu L of Beads wash buffer, uniformly blowing and stirring the sample by using a pipette gun, standing the sample on the magnetic frame for 5 minutes, and removing supernatant;
7) Taking out the sample from the magnetic frame, adding 50 mu L of Tris Buffer to resuspend the magnetic beads, and blowing and uniformly mixing the magnetic beads by using a pipette gun;
8) Placing the sample in a PCR instrument, keeping at 80 ℃ for 2min and 25 ℃, and eluting mRNA;
9) Adding 50 mu L of Bead Binding Buffer, and blowing and uniformly mixing by using a liquid transfer gun;
10 ) left at room temperature for 5min to bind mRNA to the magnetic beads;
11 Place the sample on a magnetic rack for 5min to separate mRNA from total RNA; removing the supernatant;
12 Take out the sample from the magnetic rack, put 200 μ L Beads Wash Buffer, mix well, put on the magnetic rack for 5min, remove the supernatant; total mRNA was obtained.
Transcriptome library construction:
library construction was performed using the MGI transcriptome library kit. PE150 sequencing was performed using a MGIseq2000 high throughput sequencing platform. Sequencing Subjects data reads were aligned to the human transcriptome reference sequence using the FANSe3 alignment algorithm with the parameters-L80-E5-I0-S14-B1-U0.
Differential gene screening and enrichment analysis:
two groups of transcriptome sequencing data differential expression genes (the up-down regulation is more than 2 times, and P is less than 0.05) are screened out by using the edgeR, and the KEGG database is used for carrying out gene function and pathway enrichment analysis on the down-regulated genes, so that a representative pathway with a P value less than 0.01 is displayed.
The results are shown in FIG. 7. As can be seen from fig. 7, the most prominent down-regulated pathway is the cell cycle, which is one of the most important pathways for cancer cells to maintain rapid proliferation and growth. In addition, the pathways with significant down-regulation include pathways that contribute significantly to the development of cancer, such as platinum drug resistance, TGF-beta signaling pathway, signaling pathway regulation and differentiation of stem cells, and FoxO signaling pathway. This indicates that ATCA can inhibit these cancer promotion pathways more completely, and effectively suppress the malignant phenotype of cancer cells.
In addition, the cytotoxicity of ATCA was measured using LDH method, and the cytotoxicity of the drug was detected. The results show that at the ATCA concentration of 0.5mg/ml, no significant cytotoxicity is induced to cancer cells and normal cells, and various physiological activities of normal cells HBE are normal, which indicates that the HBE has no significant killing to normal cells, i.e. no cytotoxicity.
In an ATCA (advanced telecom computing architecture) lung cancer cell apoptosis induction experiment, annexin V and propidium iodide are used for staining cancer cells, and then a flow cytometer is used for detecting that lung adenocarcinoma cells A549 and H1299 are obviously apoptotic (P is less than 0.05, n is not less than 4) under the action of 0.5mg/mL ATCA, and normal lung cells HBE are not apoptotic, which indicates that ATCA can initiate cancer cell apoptosis without killing normal cells.
In addition, in other experiments, the ATCT can also induce the apoptosis of ovarian cancer cells and inhibit the proliferation of liver cancer cells.
The above are preferred embodiments of the present application, and the scope of protection of the present application is not limited thereto, so: all equivalent changes made according to the structure, shape and principle of the present application shall be covered by the protection scope of the present application.

Claims (12)

1. A drug target for inhibiting tumor, wherein the drug target is eukaryotic translation initiation factor EIF2.
2. The drug target of claim 1, wherein the drug target is subunit EIF2S1 of initiation factor EIF2.
3. The drug target of claim 1, wherein the drug is an EIF2 inhibitor selected from aurintricarboxylic acid (ATCA) or an ammonium salt thereof.
4. The drug target of claim 1, wherein the tumor is a malignant tumor selected from the group consisting of: malignant epithelial tumors, sarcomas, myelomas, leukemias, lymphomas, melanomas, head and neck tumors, brain tumors, peritoneal carcinomas, mixed tumors, and childhood malignancies.
5. The drug target of claim 4, wherein the malignant epithelial tumors are selected from the group consisting of: lung cancer, breast cancer, liver cancer, pancreatic cancer, colorectal cancer, stomach cancer, gastroesophageal adenocarcinoma, esophageal cancer, small intestine cancer, cardiac cancer, endometrial cancer, ovarian cancer, fallopian tube cancer, vulvar cancer, testicular cancer, prostate cancer, penile cancer, kidney cancer, bladder cancer, anal cancer, gallbladder cancer, bile duct cancer, teratoma, and cardiac tumor.
6. The drug target of claim 4, wherein the tumor is lung cancer, preferably non-small cell lung cancer.
7. The drug target according to claim 4, wherein the tumor is ovarian cancer, preferably ovarian epithelial cancer.
8. The drug target of claim 4, wherein the tumor is liver cancer.
9. Use of a substance that inhibits the eukaryotic translation initiation factor EIF2 in the manufacture of a medicament for the treatment of a tumour.
10. Use according to claim 9, wherein the substance is aurintricarboxylic acid (ATCA) or an ammonium salt thereof.
11. An oral medicament for inhibiting a tumor, comprising an inhibitor capable of inhibiting the translation initiation factor EIF2 targeted to a drug target eukaryotic cell without causing phosphorylation.
12. The oral medicament according to claim 11, characterized in that the inhibitor is aurintricarboxylic acid (ATCA) or an ammonium salt thereof.
CN202211457404.4A 2022-11-21 2022-11-21 Drug target for inhibiting tumor, application and oral drug Pending CN115737619A (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN202211457404.4A CN115737619A (en) 2022-11-21 2022-11-21 Drug target for inhibiting tumor, application and oral drug
PCT/CN2022/138169 WO2024108676A1 (en) 2022-11-21 2022-12-09 Drug target for inhibiting tumor, use thereof, and oral drug
AU2022472191A AU2022472191A1 (en) 2022-11-21 2022-12-09 Drug target for inhibiting tumor, use thereof and oral drug

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202211457404.4A CN115737619A (en) 2022-11-21 2022-11-21 Drug target for inhibiting tumor, application and oral drug

Publications (1)

Publication Number Publication Date
CN115737619A true CN115737619A (en) 2023-03-07

Family

ID=85333742

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211457404.4A Pending CN115737619A (en) 2022-11-21 2022-11-21 Drug target for inhibiting tumor, application and oral drug

Country Status (3)

Country Link
CN (1) CN115737619A (en)
AU (1) AU2022472191A1 (en)
WO (1) WO2024108676A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150290155A1 (en) * 2014-04-15 2015-10-15 Aurin Biotech Inc. Methods and kits for prevention and treatment of cancer
CN107912043A (en) * 2015-07-06 2018-04-13 伊玛提克斯生物技术有限公司 For the cancer of the esophagus and the new type of peptides and peptide combinations of other cancer immunotherapies
CN110638802A (en) * 2019-10-22 2020-01-03 深圳承启生物科技有限公司 Application of ATCA in preparation of medicine for treating tumors
CN114404400A (en) * 2022-01-24 2022-04-29 深圳承启生物科技有限公司 Translation inhibitor without occupying ribosome resource as antitumor drug

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150290155A1 (en) * 2014-04-15 2015-10-15 Aurin Biotech Inc. Methods and kits for prevention and treatment of cancer
CN107912043A (en) * 2015-07-06 2018-04-13 伊玛提克斯生物技术有限公司 For the cancer of the esophagus and the new type of peptides and peptide combinations of other cancer immunotherapies
CN110638802A (en) * 2019-10-22 2020-01-03 深圳承启生物科技有限公司 Application of ATCA in preparation of medicine for treating tumors
CN114404400A (en) * 2022-01-24 2022-04-29 深圳承启生物科技有限公司 Translation inhibitor without occupying ribosome resource as antitumor drug

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DMITRIEV,S.E. ET AL.: "A Quick Guide to Small-Molecule Inhibitors of Eukaryotic Protein Synthesis", 《BIOCHEMISTRY (MOSCOW)》, vol. 85, no. 11, pages 1 - 18 *

Also Published As

Publication number Publication date
AU2022472191A1 (en) 2024-06-06
WO2024108676A1 (en) 2024-05-30

Similar Documents

Publication Publication Date Title
Yan et al. RNA-binding protein KHSRP promotes tumor growth and metastasis in non-small cell lung cancer
Lal et al. Regulation of HuR structure and function by dihydrotanshinone-I
Zhang et al. FoxM1 promotes the migration of ovarian cancer cell through KRT5 and KRT7
Ueda et al. Expression of fatty acid synthase depends on NAC1 and is associated with recurrent ovarian serous carcinomas
Li et al. Lobaplatin arrests cell cycle progression, induces apoptosis and alters the proteome in human cervical cancer cell Line CaSki
Yao et al. Knockdown of ECHS1 protein expression inhibits hepatocellular carcinoma cell proliferation via suppression of Akt activity
Carter et al. Role of human nucleoside transporters in pancreatic cancer and chemoresistance
Ou et al. p53-Induced LINC00893 regulates RBFOX2 stability to suppress gastric cancer progression
Yin et al. Targeting ABCB6 with nitidine chloride inhibits PI3K/AKT signaling pathway to promote ferroptosis in multiple myeloma
Yang et al. Putative N-glycoprotein markers of MCF-7/ADR cancer stem cells from N-glycoproteomics characterization of the whole cell lysate
Xu et al. Emerging roles of hnRNP A2B1 in cancer and inflammation
Yang et al. Hsa_circ_0093884 bound to RNA-binding protein RPS3 ameliorates hepatocyte inflammation in anti-tuberculosis drug-induced liver injury by competitively activating SIRT1
CN114155918A (en) Potential FASN inhibitor test method in drug compound library based on molecular docking and molecular dynamic simulation screening
Daigo et al. A systematic approach to the development of novel therapeutics for lung cancer using genomic analyses
Man et al. TANGO1 interacts with NRTN to promote hepatocellular carcinoma progression by regulating the PI3K/AKT/mTOR signaling pathway
CN115737619A (en) Drug target for inhibiting tumor, application and oral drug
Jia et al. Oncogenic SRSF3 in health and diseases
Tavanez et al. Hepatitis delta virus interacts with splicing factor SF3B155 and alters pre‐mRNA splicing of cell cycle control genes
Pan et al. La protein contributes to cells proliferation and migration and serves as a potential therapeutic target for hepatocellular carcinoma
Sun et al. Proteomic analysis identifies nuclear protein effectors in PKC-δ signaling under high glucose-induced apoptosis in human umbilical vein endothelial cells
Wen et al. YTHDF2 Is a Therapeutic Target for HCC by Suppressing Immune Evasion and Angiogenesis Through ETV5/PD‐L1/VEGFA Axis
Aizaz et al. Interaction analysis of TRIM proteins and innate immunity mediators of common carp (Cyprinus carpio L.) to evaluate its affinity for ubiquitination and innate immune response
CN112870363A (en) Application of human PCID2 protein in preparation or screening of antitumor drugs and compound with antitumor activity
Zhang et al. DBF4 dependent kinase inhibition suppresses hepatocellular carcinoma progression and potentiates anti-programmed cell death-1 therapy
Xu et al. CKAP4-mediated activation of FOXM1 via phosphorylation pathways regulates malignant behavior of glioblastoma cells

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