CN117838865A - Application of USP7 inhibitor in preparation of medicines for treating or preventing cholangiocarcinoma - Google Patents

Application of USP7 inhibitor in preparation of medicines for treating or preventing cholangiocarcinoma Download PDF

Info

Publication number
CN117838865A
CN117838865A CN202410052981.8A CN202410052981A CN117838865A CN 117838865 A CN117838865 A CN 117838865A CN 202410052981 A CN202410052981 A CN 202410052981A CN 117838865 A CN117838865 A CN 117838865A
Authority
CN
China
Prior art keywords
usp7
cholangiocarcinoma
bile duct
duct cancer
treating
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
CN202410052981.8A
Other languages
Chinese (zh)
Inventor
张呈生
张鹏
万亿
马锦荣
李晨曦
马俊鹏
笪艳艳
万懿琪
李晓庄
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 Affiliated Hospital of Nanchang University
Original Assignee
First Affiliated Hospital of Nanchang 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 Affiliated Hospital of Nanchang University filed Critical First Affiliated Hospital of Nanchang University
Priority to CN202410052981.8A priority Critical patent/CN117838865A/en
Publication of CN117838865A publication Critical patent/CN117838865A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/70Mechanisms involved in disease identification
    • G01N2800/7023(Hyper)proliferation
    • G01N2800/7028Cancer

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Public Health (AREA)
  • Urology & Nephrology (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biomedical Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Cell Biology (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Food Science & Technology (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Inorganic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention belongs to the field of biological medicine, and particularly relates to application of a USP7 inhibitor in preparation of a medicine for treating or preventing cholangiocarcinoma. In the invention, USP7 is used as a molecular marker, and USP7 antibody is prepared into a kit for detecting cholangiocarcinoma; and combining the USP7 inhibitor with gemcitabine and platinum chemotherapeutics to prepare the medicine for treating or preventing bile duct cancer. The invention provides a new treatment strategy for patients with bile duct cancer incapable of being radically treated by operation, and also establishes a new marker for predicting prognosis of the patients with bile duct cancer.

Description

Application of USP7 inhibitor in preparation of medicines for treating or preventing cholangiocarcinoma
Technical Field
The invention belongs to the field of biological medicine, and particularly relates to application of a USP7 inhibitor in preparation of a medicine for treating or preventing cholangiocarcinoma.
Background
Bile duct cancer (CCA) lacks ideal therapeutic strategies due to its low early diagnosis rate, resulting in poor prognosis, and places a heavy mental and economic burden on countless households. Current treatments for cholangiocarcinoma include surgery, radiation therapy, chemotherapy, and limited targeted therapies, where surgical resection is the sole curative treatment for cholangiocarcinoma. However, most patients have a diagnosis after the medical treatment of jaundice due to bile duct blockage, so that the patients are difficult to radically treat by operation, and the main treatment mode is systematic treatment mainly based on chemotherapy. Under normal conditions, cholangiocarcinoma is insensitive to radiotherapy and chemotherapy, the median total survival time of the advanced first-line chemotherapy (gemcitabine plus cisplatin) and the second-line chemotherapy (FOLFOX) of cholangiocarcinoma is short, the prognosis of patients with cholangiocarcinoma is limited, and the untargeted cytotoxicity of chemotherapeutics brings serious side effects. Although a small number of molecular targeted drugs have been approved for clinical treatment of cholangiocarcinoma, the existing targeted drugs have stricter requirements on indications, and most of them can only be used for adjuvant treatment of advanced cholangiocarcinoma, and the proportion and degree of benefited patients are low. Therefore, there is an urgent need to elucidate the molecular mechanism of biliary tract carcinogenesis and development of new therapeutic targets and development of targeted drugs.
Disclosure of Invention
Based on this, the present invention provides the use of USP7 inhibitors in the manufacture of a medicament for the treatment or prevention of cholangiocarcinoma, at least solving one of the problems of the prior art.
In a first aspect, the invention provides the use of a USP7 inhibitor in the manufacture of a medicament for the treatment or prophylaxis of cholangiocarcinoma.
In some preferred embodiments, the USP7 inhibitor is one or more of P5091, P22077, BAY 11-708, NSC 632839, XL177A, HBX 19818, GEN-6776.
In some more preferred embodiments, the USP7 inhibitor is P5091 or P22077.
In some preferred embodiments, the USP7 inhibitor is used at a concentration of 15mg/kg to 40mg/kg, e.g., 20mg/kg, 25mg/kg, 30mg/kg, 35mg/kg, 40mg/kg, etc.
In some preferred embodiments, the drug for treating or preventing cholangiocarcinoma contains Gemcitabine (Gemcitabine) and a platinum substance, which is one or more of cisplatin (cispratin), carboplatin (Carboplatin), oxaliplatin (Oxaliplatin).
In some preferred embodiments, the gemcitabine is used at a concentration of 20mg/kg to 50mg/kg, such as 20mg/kg, 25mg/kg, 30mg/kg, 35mg/kg, 40mg/kg, etc.; the platinum-based material is used at a concentration of 1 to 10mg/mg, for example, 1mg/kg, 2.5mg/kg, 5mg/kg, 7.5mg/kg, 10mg/kg, etc.
In some preferred embodiments, the cholangiocarcinoma is one or more of intrahepatic cholangiocarcinoma, portafas cholangiocarcinoma, and extrahepatic cholangiocarcinoma.
In a second aspect, the present invention provides a medicament for treating or preventing cholangiocarcinoma comprising a USP7 inhibitor, gemcitabine and a platin, the platin being one or more of cisplatin, carboplatin, oxaliplatin. The medicine can be an intravenous injection preparation or a combination of an intravenous injection preparation and an oral preparation. When the medicament is a combination of an intravenous formulation and an oral formulation, the intravenous formulation contains a USP7 inhibitor and the oral formulation contains gemcitabine and a platinum group substance.
In some preferred embodiments, the medicament for treating or preventing cholangiocarcinoma further comprises a pharmaceutically acceptable carrier.
In some preferred embodiments, the pharmaceutically acceptable carrier is physiological saline.
Due to the adoption of the technical scheme, the embodiment of the invention has at least the following beneficial effects:
(1) USP7 is used as a molecular marker, and clinical sample verification proves that the prognosis situation of a bile duct cancer patient can be well predicted, and the USP7 can be used as a molecular marker for evaluating bile duct cancer, can be applied to development of a bile duct cancer prognosis prediction detection kit, can be used for guiding clinical individuation treatment, and can improve the treatment accuracy;
(2) The USP7 inhibitor can remarkably inhibit the growth of bile duct cancer cells and bile duct cancer human xenograft model (PDX) tumors, and can be used for guiding clinical bile duct cancer treatment tests and later clinical application;
(3) The USP7 inhibitor combined with gemcitabine and cisplatin can remarkably inhibit the growth of bile duct cancer, provides a new treatment scheme for bile duct cancer patients difficult to radically treat by operation, and can be used for guiding clinical bile duct cancer treatment tests and later clinical application.
Drawings
The following drawings are only for purposes of illustration and explanation of the present invention and are not intended to limit the scope of the invention.
FIG. 1 shows the analysis of USP7 mRNA levels in biliary tract cancer tumor tissue and paracancerous/normal tissue by TCGA and GEO databases in an example of the present invention.
FIG. 2 shows protein levels of USP7 on bile duct cancerous tumor tissue and paracancerous tissue by WB assay 12 in an embodiment of the invention.
FIG. 3 shows the results of immunohistochemical staining of cholangiocarcinoma tissue chip (left) and quantification of USP7 expression levels (right) in the examples of the present invention.
Figure 4 shows a patient survival curve in an embodiment of the present invention.
FIG. 5 is a graph showing the results of cell proliferation experiments in examples of the present invention.
FIG. 6 is a graph showing the results of clone formation in the examples of the present invention.
FIG. 7 shows the results of cell cycle flow cytometry in an embodiment of the present invention.
Figure 8 shows the statistics of cell cycle flow cytometry in an embodiment of the present invention.
FIG. 9 shows the quantitative proteomics results of knockdown USP7 in the examples of the invention.
Fig. 10 is a diagram of endpoint mice tumor-bearing in PDX model experiments in accordance with embodiments of the present invention.
FIG. 11 shows the change in tumor volume of mice during the experiment in the examples of the present invention.
FIG. 12 is a graph showing the tumor size at the final sacrifice of mice in the examples of the present invention.
FIG. 13 is a statistical plot of tumor weights at the final sacrifice of mice in the examples of the present invention.
In the figures, if p <0.05, then there is considered a significant difference.
Detailed Description
The invention will be more readily understood by the following examples in conjunction with the general description thereof, which are given solely for the purpose of illustrating certain aspects of the invention and are not intended to be limiting thereof. The experimental methods or conditions used in the examples were carried out according to the conventional methods or manufacturer's instructions unless otherwise specified, and the materials and reagents used in the examples were obtained from commercial sources unless otherwise specified.
In the cell, 70% -80% of the protein is degraded through ubiquitin-proteasome pathway, deubiquitinase (DUBs) is a protease capable of specifically shearing covalent links between substrate protein and ubiquitin molecules, and the inverse regulation of ubiquitin modification of the substrate protein can be realized, so that the steady state or function of the substrate protein is regulated and controlled. More and more studies indicate that abnormal regulation of deubiquitinase plays an important role in the development and progression of tumors. Ubiquitin-specific protease 7 (ubiquitin-specific protease, usp 7) is a star molecule in the deubiquitinase protein family. USP7, also known as herpes virus associated ubiquitin specific protease (HAUSP), is one of the most widely studied DUBs, is abnormally high expressed in various tumors such as breast cancer, colorectal cancer, etc., and is closely related to the poor prognosis of patients. USP7 has a broad range of substrates, such as P53, PTEN, FOXO4, N-MYC, etc., and is involved in regulating a variety of signaling pathways associated with tumorigenic development. Thus, targeting USP7 is considered a promising cancer treatment strategy, several USP7 inhibitors have been developed and show good efficacy in preclinical tumor models in vitro and in vivo. However, no report has been made at present on the abundance of USP7 expression, functional effects in cholangiocarcinoma, and whether it can be used as a potential therapeutic target.
The inventors found through studies that USP7 is highly expressed in the tissues of cholangiocarcinoma patients and is associated with poor prognosis of cholangiocarcinoma patients, knocking out USP7 or using an inhibitor of USP7 can block tumor cells in S phase of the cell cycle and significantly inhibit malignant proliferation of cholangiocarcinoma cells. Further using quantitative proteomics techniques, the inventors have found that USP7 affects proliferation of tumor cells by regulating expression of a range of cyclin-related proteins. USP7 is thus a potentially effective target for the treatment of cholangiocarcinoma, but monotherapy with tumors tends to quickly develop resistance, and combination therapy strategies are considered as effective approaches to avoid resistance. At present, no combination treatment strategy and effect for treating bile duct cancer by utilizing a targeted therapy-USP 7 inhibitor combined with a first-line chemotherapy method of bile duct cancer-gemcitabine plus cisplatin therapy are reported. The inventor confirms that the USP7 inhibitor or the combined gemcitabine and cisplatin treatment strategy can effectively inhibit the growth of bile duct cancer through the evaluation of a bile duct cancer patient-derived xenograft tumor model (PDX model). These findings reveal the molecular mechanisms of USP7 to promote tumorigenesis by modulating the cell cycle and suggest that therapeutic approaches targeting USP7 or/and combined gemcitabine plus cisplatin are novel strategies for cholangiocarcinoma treatment.
In order to improve the treatment effect of patients with bile duct cancer which cannot be radically treated by surgical operation and prolong the survival rate of the patients, the invention provides the application of the USP7 inhibitor in preparing medicaments for treating or preventing the bile duct cancer and the medicaments for treating or preventing the bile duct cancer. Meanwhile, the invention also provides a method for using USP7 as a molecular marker of bile duct cancer to develop a bile duct cancer detection kit, which can be used for predicting the survival condition of a bile duct cancer patient. The technical scheme and the effect of the invention are verified in the following experiment.
1. Analysis of expression level of USP7 in cholangiocarcinoma by database
Analysis of cholangiocarcinoma transcriptome data in TCGA and GEO databases, as shown in fig. 1, showed that USP7 mRNA levels were higher in tumor tissue than in normal/paracancerous tissue. This result suggests that USP7 is specifically highly expressed in bile duct cancer tumor tissue, while it is relatively low in normal tissue.
2. Western immunoblotting experiment (Western blot)
Clinical tumor samples were obtained from the first affiliated hospital at the university of western traffic, all samples were obtained with informed consent from the patient, and the study was approved by the ethical committee at the university of western traffic (approval No. XJTUAE 2023-1604). By searching the medical records, 12 pairs of normal and paracancerous tissues of the cholangiocarcinoma patient are obtained, then cell lysate is added, the tissues are ground by a homogenizer, and after centrifugation at 12000rpm for 5 minutes, the supernatant is collected. Protein fluid was quantified by operating according to the Bradford method protein concentration assay kit (bio, SK 3031) instructions. SDS-PAGE electrophoresis and membrane transfer are then carried out, then PVDF membrane is blocked and antibody incubation is carried out, and finally exposure is carried out.
Wherein, the cell lysate is composed of 50mM Tris-HCl, 250mM NaCl, 1mM EDTA, 50mM NaF, 0.5% Triton X-100, pH 7.4, 1mM DTT, 1mM PMSF and Cocktail (Sigma, 1:100) added before use.
As shown in fig. 2, USP7 protein levels in tumor tissue (T) of cholangiocarcinoma patients were higher than in paired paracancerous tissue (N). This result suggests that USP7 is specifically highly expressed in bile duct cancer tumor tissue, while it is relatively low in normal tissue.
3. Immunohistochemistry of cholangiocarcinoma tissue chip
Bile duct cancer tissue chips were from Shanghai core Biotechnology Inc., all samples were taken with informed consent from the patient. Immunohistochemical staining with USP7 (CST) antibody, scoring the samples according to the degree of staining and the proportion of stained cells of USP7 to obtain an immunohistochemical index (H-Score ), H-score= (weak positive percentage x 1) + (medium positive percentage x 2) + (strong positive percentage x 3), pi refers to the proportion of cells of a certain intensity in positive cells, i refers to staining intensity, typically a value of 1, 2 or 3; a final immunohistochemical index was obtained.
The results are shown in FIG. 3, where the H-score of normal tissue is about 80 points, and the H-score of cholangiocarcinoma tissue is about 200 points, the protein level of USP7 is significantly higher in cholangiocarcinoma tissue than in normal tissue.
4. Survival curve drawing
The expression level of USP7 in cholangiocarcinoma and the total survival time of the patient are obtained by searching the TCGA database, then the patient is divided into two groups of high expression and low expression of USP7 according to the expression condition of the USP7, and the survival curve of the patient is drawn by using the R language surviviner and survivinal package.
The results are shown in FIG. 4, where the higher the mRNA expression level of USP7, the shorter the patient survival time.
5. Cell viability assay and clone formation
Human normal bile duct epithelial cell line (HIBEpic) and bile duct cancer cell line (HuCCT 1, RBE, HCCC-9810) were purchased from American Type Culture Collection (ATCC), and the cells were inoculated in 60mm dishes at 37℃with 5% CO 2 In culture, cells in the logarithmic growth phase were tested using DMEM medium (Gibco, usa) supplemented with 10% fetal bovine serum (Gibco, usa), 100 units/ml penicillin (bi-cloudy days, china) and 100mg/ml streptomycin (bi-cloudy days, china).
5000 cells HIBEpic, huCCT, RBE and HCCC-9810 were seeded into 96-well cell culture plates, treated one day later with DMSO, 5. Mu. M P5091, 7.5. Mu. M P22077 (Selleck), viability of the cells was detected on days 1, 3, 5, 7 using CCK8 detection kit (Tao Shu organisms), respectively, and proliferation curves were plotted using Graphpad.
20000 HIBEpic, huCCT, RBE and HCCC-9810 cells were seeded into 12-well cell culture plates, treated with DMSO, 10. Mu. M P5091, 12.5. Mu. M P22077, respectively, after one day, fixed with 4% PFA after 72 hours, and stained with 0.1% crystal violet.
As shown in fig. 5 and 6, USP7 inhibitors P5091, P22077 can significantly inhibit the proliferation and clonogenic capacity of tumor cells without significantly affecting normal human bile duct epithelial cells.
7. Cell cycle detection
Digestive bile duct cancer cells with pancreatin without EDTA and collecting in a 1.5 mL centrifuge tube, centrifuging at 1000xg for 5min, discarding the supernatant; 1mL of precooled PBS is added to resuspend the cells, 1000xg is centrifuged for 5min, and the supernatant is discarded; re-suspending cells by using 250 mu L of precooled PBS, slowly dripping 750 mu L of precooled absolute ethyl alcohol, and uniformly mixing; the sample is placed at the temperature of minus 20 ℃ and fixed for 2 hours to overnight, 1000xg is centrifuged for 5 minutes, and the supernatant is discarded; 1mL pre-chilled PBS resuspended cells, centrifuged at 1000Xg for 5min, and the supernatant discarded. 100. Mu.L RNase A (10 mg/mL) was added to the cell pellet, mixed well and incubated at 37℃for 30min. Dyeing: 200 mu l of propidium iodide dye solution is added into the sample, evenly mixed and incubated at 4 ℃ for 30min in a dark place, and the sample is detected by a flow cytometer.
As shown in fig. 7 and 8, USP7 inhibitors P5091, P22077 can significantly inhibit the progression of the cell cycle of cholangiocarcinoma cells and primarily block the tissue in S phase.
8. Quantitative proteomics experiments
Pancreatin digested USP7 shRNA knockdown and USP7 non-knockdown cholangiocarcinoma RBE cell lines, and cell pellet was collected by centrifugation. 4 volumes of lysis buffer (8M urea, 1% protease inhibitor) was added to each cell pellet and sonicated. Centrifugation was performed at 12000 and g for 10 min at 4℃to remove cell debris, and the supernatant was transferred to a new centrifuge tube, and protein concentration was measured using the BCA kit. And (3) carrying out enzymolysis on the protein of each sample in an equivalent amount, and regulating the volume to be consistent by using a lysate. Adding 1 time of pre-cooled acetone, mixing by vortex, adding 4 times of pre-cooled acetone, and precipitating at-20deg.C for 2 h.4500 g, centrifuging for 5min, discarding the supernatant, and washing the precipitate with pre-cooled acetone for 2 times. After air drying the precipitate, TEAB was added to a final concentration of 200. 200 mM, the precipitate was broken up by sonication, trypsin was added at a ratio of 1:50 (protease: protein, m/m), and enzymatic hydrolysis was carried out overnight. Dithiothreitol (DTT) was added to a final concentration of 5 mM and reduced at 56℃for 30min. Then Iodoacetamide (IAM) was added to give a final concentration of 11 mM and incubated at room temperature for 15 min in the dark.
Constructing a spectra Library: the peptide fragments were dissolved in liquid chromatography mobile phase a and then separated using a NanoElute ultra high performance liquid system. Mobile phase a is an aqueous solution containing 0.1% formic acid, 2% acetonitrile; mobile phase B was an acetonitrile solution containing 0.1% formic acid.
Setting a liquid phase gradient: 0-10 min,9% -24% mobile phase B;10-14 min,24% -35% of mobile phase B;14-17 min,35% -80% of mobile phase B;17-20 min,80% mobile phase B, flow rate maintained at 1000 nL/min. Peptide fragments were isolated by ultra-high performance liquid chromatography and injected into a Capilliry ion source for ionization and then subjected to timsTOF Pro (Bruker) mass spectrometry for data acquisition.
DIA data acquisition: the liquid phase parameters are consistent with those of the warehouse building. Peptide fragments were separated by ultra high performance liquid chromatography and analyzed by timsTOF Pro (Bruker) mass spectrometry. The ion source voltage was set to 1.75 kV and both the peptide fragment parent ion and its secondary fragments were detected and analyzed using TOF. The data acquisition mode uses a data independent parallel accumulation serial fragmentation (dia-PASEF) mode, the scanning range of the primary mass spectrum is set to 400-1500 m/z, 10 PASEF mode acquisitions are carried out after one primary mass spectrum is acquired, the secondary mass spectrum scanning takes 100-1700 m/z as an interval, and every 25 m/z as a window.
As shown in FIG. 9, quantitative proteomic experiments were performed to knock down the expression of USP7 by shRNA, and found that the down-regulation of the expression of USP7 can affect the expression levels of a plurality of the cyclin-involved regulatory proteins including cyclin D1 (CCND 1), D3 (CCND 3) and the like.
9. Evaluation of patient-derived tumor xenograft (PDX) model targeting USP7 in combination with gemcitabine plus cisplatin for cholangiocarcinoma
Animal experiments were approved by the ethical committee of the university of traffic of western America (approval number: XJTUAE 2023)-1604). NCG mice (severe immunodeficiency, 6-8 weeks old, jiangsu Jiyaokang) were subcutaneously transplanted with tumor tissues of cholangiocarcinoma patients. When the tumor volume is 100-150mm 3 Mice were randomly divided into the following 4 groups:
control group: treating with physiological Saline (Saline);
gem+cis group: treatment with gemcitabine (25 mg/kg) plus cisplatin (5 mg/kg);
p5091 group: treatment with USP7 inhibitor P5091 (25 mg/kg);
Gem+cis+P5091 group: treatment was performed with gemcitabine (25 mg/kg) plus cisplatin (5 mg/kg) plus USP7 inhibitor P5091 (25 mg/kg).
Gemcitabine plus cisplatin is administered by gavage once a week; p5091 is administered by intraperitoneal injection, once a day; the length and width of the tumor were measured every two days using vernier calipers, according to the formula 4pi/3× (width/2) 2 Tumor volume size was calculated by x (length/2).
As shown in fig. 10-13, the bile duct cancer responds to treatment with gemcitabine plus cisplatin in combination with USP7 inhibitor P5091, USP7 inhibitor P5091 shows a significant inhibition of bile duct cancer tumor growth and is far superior in effect to the clinical bile duct cancer first line treatment regimen (gemcitabine plus cisplatin); the treatment strategy of the USP7 inhibitor P5091 combined with gemcitabine and cisplatin can further inhibit the growth of bile duct cancer tumor (tumor size, growth speed and final tumor weight) on a single drug basis. As can be seen from FIG. 11, the tumor volume of gemcitabine plus cisplatin plus USP7 inhibitor group P5091 (Gem+cis+P5091) after 20 days of treatment was 100mm 3 About, the tumor volume of the gemcitabine plus cisplatin group (Gem+cis) is 400mm 3 The above. As can be seen from FIG. 13, after 20 days of treatment, the tumor weight of the gemcitabine plus cisplatin plus USP7 inhibitor group P5091 (Gem+cis+P 5091) was around 0.05g, while the tumor volume of the gemcitabine plus cisplatin group (Gem+cis) was around 0.65 g.
10. Preparation of bile duct cancer detection kit
The bile duct cancer detection kit comprises reagents for detecting the expression level of USP7, wherein each reagent comprises a first antibody and a second antibody, the first antibody is a USP7 specific antibody, and the second antibody is an antibody which is homologous with the first antibody and is provided with a horseradish peroxidase label. The kit may also include conventional containers, instructions for use, positive controls, negative controls, buffers, adjuvants, and solvents. The instructions of the application describe how to detect the tumor by using the kit, and how to judge the tumor development and select the treatment scheme by using the detection result. The components of the kit may be packaged in the form of an aqueous medium.
11. Bile duct cancer detection kit for detecting bile duct cancer
The detection method of the bile duct cancer detection kit is Western Blot (WB, western blotting experiment) and IHC (IHC, immunohistochemical experiment), is a technology commonly used for separating and identifying proteins in research, and is widely applied to the research fields of molecular biology, biochemistry, cytobiology, immunogenetics and the like.
The WB experiment is to separate the mixed protein sample by polyacrylamide gel electrophoresis (PAGE), transfer the separated protein sample onto nitrocellulose or PVDF membrane by electric field device imprinting, then combine the membrane with the corresponding first antibody specifically, then combine the membrane with the second antibody with horseradish peroxidase, and finally detect the combined antibody by chemiluminescence. Since antibodies bind only to the target protein, only a clear band is generally visible, the gray scale of which corresponds to the mass of the target protein.
IHC experiments are based on the specificity of antigen-antibody reactions, i.e. antibodies can only be detected when they bind to the corresponding antigen. In the experiment, tissues or cells were first fixed on a slide and appropriately treated to expose the antigen. The specific antibody is then bound to an antigen in a tissue or cell to form an antigen-antibody complex. Finally, the complex is presented on the glass slide through a chromogenic reaction, so that the detection of the antigen is realized, and the color of the staining corresponds to the quality of the target protein.
In conclusion, USP7 can be used as a molecular marker for predicting survival of cholangiocarcinoma patients; the combination therapy regimen of USP7 inhibitors in combination with gemcitabine plus cisplatin chemotherapy may significantly inhibit the malignant growth of cholangiocarcinoma.
The above description is merely illustrative of the present invention, and the scope of the present invention is not limited thereto, and any changes or substitutions that may be easily conceived by those skilled in the art within the technical scope of the present invention should be covered in the scope of the present invention.

Claims (10)

  1. Use of an usp7 inhibitor in the manufacture of a medicament for the treatment or prophylaxis of cholangiocarcinoma.
  2. 2. The use according to claim 1, wherein the USP7 inhibitor is one or more of P5091, P22077, BAY 11-708, NSC 632839, XL177A, HBX 19818, GEN-6776.
  3. 3. The use according to claim 2, wherein the USP7 inhibitor is P5091 or P22077.
  4. 4. The use according to claim 2, wherein the medicament for treating or preventing cholangiocarcinoma comprises gemcitabine and a platinum substance, wherein the platinum substance is one or more of cisplatin, carboplatin, oxaliplatin.
  5. 5. The use of claim 4, wherein the bile duct cancer is one or more of intrahepatic bile duct cancer, portal biliary duct cancer, and extrahepatic bile duct cancer.
  6. 6. A medicament for treating or preventing cholangiocarcinoma comprising a USP7 inhibitor, gemcitabine and a platinum species, the platinum species being one or more of cisplatin, carboplatin, oxaliplatin.
  7. 7. The medicament for treating or preventing cholangiocarcinoma according to claim 6, wherein the USP7 inhibitor is one or more of P5091, P22077, BAY 11-708, NSC 632839, XL177A, HBX 19818, GEN-6776.
  8. 8. The medicament for treating or preventing cholangiocarcinoma according to claim 7, wherein the USP7 inhibitor is P5091 or P22077.
  9. 9. The medicament for treating or preventing cholangiocarcinoma according to claim 7, further comprising a pharmaceutically acceptable carrier.
  10. 10. The medicament for treating or preventing cholangiocarcinoma according to claim 9, wherein the pharmaceutically acceptable carrier is physiological saline.
CN202410052981.8A 2024-01-15 2024-01-15 Application of USP7 inhibitor in preparation of medicines for treating or preventing cholangiocarcinoma Pending CN117838865A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202410052981.8A CN117838865A (en) 2024-01-15 2024-01-15 Application of USP7 inhibitor in preparation of medicines for treating or preventing cholangiocarcinoma

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202410052981.8A CN117838865A (en) 2024-01-15 2024-01-15 Application of USP7 inhibitor in preparation of medicines for treating or preventing cholangiocarcinoma

Publications (1)

Publication Number Publication Date
CN117838865A true CN117838865A (en) 2024-04-09

Family

ID=90538162

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202410052981.8A Pending CN117838865A (en) 2024-01-15 2024-01-15 Application of USP7 inhibitor in preparation of medicines for treating or preventing cholangiocarcinoma

Country Status (1)

Country Link
CN (1) CN117838865A (en)

Similar Documents

Publication Publication Date Title
Gao et al. Inhibition of AIM2 inflammasome-mediated pyroptosis by Andrographolide contributes to amelioration of radiation-induced lung inflammation and fibrosis
Li et al. ITGB1 enhances the radioresistance of human non-small cell lung cancer cells by modulating the DNA damage response and YAP1-induced epithelial-mesenchymal transition
Hua et al. Suppression of glioblastoma by targeting the overactivated protein neddylation pathway
Liu et al. NLRP3 inflammasome activation by estrogen promotes the progression of human endometrial cancer
Tirado et al. Roscovitine is an effective inducer of apoptosis of Ewing's sarcoma family tumor cells in vitro and in vivo
US11452775B2 (en) Detection of arginine methylation of EGFR for prediction of resistance to therapy
US20180104232A1 (en) Methods for determining prognosis for breast cancer patients
Feng et al. Proteome profiling of Epstein–Barr virus infected nasopharyngeal carcinoma cell line: Identification of potential biomarkers by comparative iTRAQ-coupled 2D LC/MS-MS analysis
Farrell et al. Targeting the fatty acid binding proteins disrupts multiple myeloma cell cycle progression and MYC signaling
Li et al. Kinesin family member 20B regulates tongue cancer progression by promoting cell proliferation
Yang et al. Blockage of glycolysis by targeting PFKFB3 suppresses the development of infantile hemangioma
Ding et al. SCP2-mediated cholesterol membrane trafficking promotes the growth of pituitary adenomas via Hedgehog signaling activation
Yu et al. B4GALNT2 Gene Promotes Proliferation, and Invasiveness and Migration Abilities of Model Triple Negative Breast Cancer (TNBC) Cells by Interacting With HLA-B Protein
Xiao et al. USP39-mediated deubiquitination of Cyclin B1 promotes tumor cell proliferation and glioma progression
CN117487915A (en) Application of H2AFV in diagnosis, prognosis, prevention or treatment of liver cancer
Liu et al. Suppression of CCT3 inhibits melanoma cell proliferation by downregulating CDK1 expression
CN117838865A (en) Application of USP7 inhibitor in preparation of medicines for treating or preventing cholangiocarcinoma
Li et al. Silencing of E3 ubiquitin ligase RNF8 enhances ionizing radiation sensitivity of medulloblastoma cells by promoting the deubiquitination of PCNA
Pluskalová et al. Photodynamic treatment (ALA-PDT) suppresses the expression of the oncogenic Bcr-Abl kinase and affects the cytoskeleton organization in K562 cells
Shi et al. Inhibition of ClC-5 suppresses proliferation and induces apoptosis in cholangiocarcinoma cells through the Wnt/β-catenin signaling pathway
Wei et al. PSME3 induces radioresistance and enhances aerobic glycolysis in cervical cancer by regulating PARP1
Yang et al. Clock gene NR1D1 might be a novel target for the treatment of bladder cancer
Cheng et al. NEDD4 E3 ubiquitin protein ligase serves an important role in cutaneous melanoma occurrence and development
Gregory et al. Pancreatic Tumor-Derived Extracellular Vesicles Stimulate Schwann Cell Phenotype Indicative of Perineural Invasion via IL-8 Signaling
CN114010791B (en) Application of MyD88-IFN gamma R1 dimer as target in preparation of medicine for preventing colon cancer

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