CN112439067A - Application of SGLT2 inhibitor in preparation of product for improving sensitivity of antitumor drugs - Google Patents

Application of SGLT2 inhibitor in preparation of product for improving sensitivity of antitumor drugs Download PDF

Info

Publication number
CN112439067A
CN112439067A CN201910826149.8A CN201910826149A CN112439067A CN 112439067 A CN112439067 A CN 112439067A CN 201910826149 A CN201910826149 A CN 201910826149A CN 112439067 A CN112439067 A CN 112439067A
Authority
CN
China
Prior art keywords
tumor
drug
product
chemotherapeutic
cells
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.)
Granted
Application number
CN201910826149.8A
Other languages
Chinese (zh)
Other versions
CN112439067B (en
Inventor
谢伟东
钟今
孙鹏博
丁艺佩
张雅鸥
许乃寒
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 Graduate School Tsinghua University
Original Assignee
Shenzhen Graduate School Tsinghua 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 Shenzhen Graduate School Tsinghua University filed Critical Shenzhen Graduate School Tsinghua University
Priority to CN201910826149.8A priority Critical patent/CN112439067B/en
Publication of CN112439067A publication Critical patent/CN112439067A/en
Application granted granted Critical
Publication of CN112439067B publication Critical patent/CN112439067B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

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
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/381Heterocyclic compounds having sulfur as a ring hetero atom having five-membered rings
    • 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
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses application of an SGLT2 inhibitor in preparation of a product for improving the sensitivity of an anti-tumor medicament. The invention discovers that the SGLT2 inhibitor Canagliflozin can inhibit the expression and function of P-glycoprotein in tumor cells and inhibit the autophagy phenomenon of stress protection of the tumor cells in the face of chemotherapy drug treatment, thereby improving the sensitivity of the tumor cells to the chemotherapy drugs and enhancing the treatment effect of the drugs. The inhibitor can exist in the form of liquid, dry powder or extract, can be prepared into various dosage forms with health care or medicinal efficacy such as capsules, tablets, injection, pills, granules, powder and the like with various conventional auxiliary materials, and can be combined with other chemotherapeutic drugs for treating malignant tumors.

Description

Application of SGLT2 inhibitor in preparation of product for improving sensitivity of antitumor drugs
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to application of an SGLT2 inhibitor in preparation of products for improving the sensitivity of antitumor drugs, enhancing the treatment effect of antitumor drugs and treating tumors.
Background
Malignant tumor is cancer cell developed due to genetic and environmental carcinogenic factors, which results in the loss of normal regulation of local tissue cells in human body at gene level and has the characteristics of unlimited proliferation, metastasis and transformation. Malignant tumors not only seriously harm the health of human beings, but also greatly affect the lives of patients, and also form great medical burden on families and society. The number of cases of malignant tumor and death cases in China are the first in the world, are the most serious public health problem at present, and are not beneficial to social stability and economic development.
Currently, malignant tumors are mainly treated by surgical resection, chemotherapy, radiotherapy and the like, wherein chemotherapy is one of the most effective methods for controlling the spread of cancerous cells in vivo. However, chemotherapy often has the disadvantages of great side effects and easy generation of drug resistance by tumor cells. Drug resistance is the most important cellular defense mechanism when tumor cells are attacked by chemotherapeutic drugs, relates to various antitumor drugs commonly used in clinical practice, and is one of the most serious obstacles to successful chemotherapy of tumors.
Drug resistance of tumors is associated with multiple factors, of which the mechanism of multidrug resistance mediated by P-glycoprotein (P-glycoprotein) is one of the key breakthrough. The P-glycoprotein is glycoprotein encoded by a multidrug resistance gene (MDR1) and has the molecular weight of 170KD, has the function of a drug pump, releases energy after combining ATP to discharge chemotherapy drug molecules in cells to the outside of the cells, changes the drug concentration in the cells, and reduces the sensitivity of tumor cells to drugs.
Autophagy is a ubiquitous phenomenon in eukaryotic cells and is capable of updating the metabolism and energy of cells, thereby facilitating the maintenance of cell homeostasis. However, autophagy is bilateral to tumors and plays an important role in the development and treatment of tumors. Inducing autophagy in some cases can inhibit tumor growth and cause autophagy-related cell death. However, most often, in the face of conditions that are not conducive to survival, such as hypoxia, nutrient deficiency, DNA damage, etc., autophagy can be regulated by scavenging oxygen free radicals, damaging mitochondria, etc., to provide stress protection for tumor cells.
Canagliflozin (CAN for short) is an SGLT2 inhibitor, and is a type of diabetes drug on the market. The CAN has the action mechanism of selectively inhibiting sodium-glucose cotransporter SGLT2 on the proximal convoluted tubule and inhibiting the absorption of glucose by the proximal convoluted tubule, thereby reducing the blood sugar. Both safety and efficacy have been subject to clinical evaluation.
Disclosure of Invention
The invention aims to provide application of an SGLT2 inhibitor in preparation of products for improving the sensitivity of antitumor drugs, enhancing the treatment effect of antitumor drugs and treating tumors.
The invention firstly protects the new application of the SGLT2 inhibitor or the pharmaceutically acceptable salt and ester thereof.
The invention protects the application of SGLT2 inhibitor or pharmaceutically acceptable salt and ester thereof in any one of the following A1) -A8):
A1) preparing a product for preventing and/or treating tumors;
A2) preparing a product for enhancing the treatment effect of the antitumor drug or the chemotherapeutic drug;
A3) preparing a product for inhibiting tumor drug resistance;
A4) preparing a product for improving the sensitivity of tumor cells to anti-tumor drugs or chemotherapeutic drugs;
A5) preparing a product for promoting the accumulation of the antitumor drug or the chemotherapeutic drug in the tumor cells;
A6) preparing a product for reducing the expression level of the P-glycoprotein of the tumor cells;
A7) preparing a product for inhibiting autophagy induced by an anti-tumor drug, a chemotherapeutic drug or an autophagy inducer;
A8) preparing the product for inhibiting the growth of the tumor.
The invention also protects a product, the active ingredient of which is SGLT2 inhibitor or pharmaceutically acceptable salt and ester thereof;
the function of the product is any one of the following B1) -B8):
B1) preventing and/or treating tumors;
B2) enhancing the therapeutic effect of the anti-tumor drug or the chemotherapeutic drug;
B3) inhibiting tumor drug resistance;
B4) improving the sensitivity of tumor cells to anti-tumor drugs or chemotherapeutic drugs;
B5) promoting the accumulation of the antitumor drug or the chemotherapeutic drug in the tumor cells;
B6) reducing the expression level of tumor cell P-glycoprotein;
B7) inhibiting autophagy induced by antineoplastic agent or chemotherapeutic agent or autophagy inducer;
B8) inhibiting tumor growth.
The invention also provides an anti-tumor composition, which consists of the SGLT2 inhibitor or pharmaceutically acceptable salt and ester thereof and anti-tumor drugs or chemotherapeutic drugs;
the anti-tumor composition has the functions of any one of the following B1) -B8):
B1) preventing and/or treating tumors;
B2) enhancing the therapeutic effect of the anti-tumor drug or the chemotherapeutic drug;
B3) inhibiting tumor drug resistance;
B4) improving the sensitivity of tumor cells to anti-tumor drugs or chemotherapeutic drugs;
B5) promoting the accumulation of the antitumor drug or the chemotherapeutic drug in the tumor cells;
B6) reducing the expression level of tumor cell P-glycoprotein;
B7) inhibiting autophagy induced by antineoplastic agent or chemotherapeutic agent or autophagy inducer;
B8) inhibiting tumor growth.
In any of the above applications or products or anti-tumor compositions, the reduction of the expression level of the P-glycoprotein in the tumor cells is the reduction of the protein expression level of the P-glycoprotein in the tumor cells; further, the reduction of the protein expression level of the tumor cell P-glycoprotein is realized by enhancing the degradation of the tumor cell P-glycoprotein; furthermore, the enhancement of the degradation of the tumor cell P-glycoprotein is realized by a proteasome degradation pathway.
In any of the above applications or products or anti-tumor compositions, the inhibition of autophagy induced by the anti-tumor drug or chemotherapeutic drug or autophagy inducer is characterized by reducing the LC3 II/LC 3I ratio and/or increasing the expression level of p 62; the autophagy induced by the anti-tumor drug or the chemotherapeutic drug or the autophagy inducer blocks autophagy by activating p-ULK1(Ser 757).
In any of the above uses or products or anti-tumor compositions, the inhibition of tumor growth is manifested by a reduction in tumor cell volume and/or a reduction in tumor cell weight.
In any of the above applications or products or anti-tumor compositions, the SGLT2 inhibitor is Canagliflozin (CAN), the CAS accession number of the inhibitor is 842133-18-0, and the molecular formula of the inhibitor is C24H25FO5S, molecular weight 444.52. The structural formula is shown as formula I.
Figure BDA0002189148290000031
In any of the above applications or products or anti-tumor compositions, the anti-tumor drug or chemotherapeutic drug may be an antibiotic anti-tumor drug or chemotherapeutic drug. In a specific embodiment of the present invention, the antibiotic anti-tumor drug or chemotherapeutic drug is Doxorubicin (Doxorubicin, Doxo for short), CAS number is 23214-92-8, and molecular formula is C27H29NO11And the molecular weight is 543.52. The structural formula is shown as a formula II.
Figure BDA0002189148290000032
In any of the above applications or products or anti-tumor compositions, the tumor may be a common tumor in the prior art, such as liver cancer, colon cancer or breast cancer; the tumor cell can be common tumor cells in the prior art, such as liver cancer cells (such as human liver cancer cell HepG2, adriamycin-resistant human liver cancer cell HepG2-ADR), colon cancer cells (such as human colon cancer cell HCT116) or breast cancer cells (such as human breast cancer cell MCF 7).
In any of the above applications or products or anti-tumor compositions, the product may be a medicament and the anti-tumor composition may be an anti-tumor pharmaceutical composition. The product (medicine) or the antitumor composition (antitumor medicine composition) can be used together with other antitumor medicines or chemotherapeutic medicines for treating malignant tumors. When needed, one or more pharmaceutically acceptable carriers can be added into the product (medicament) or the anti-tumor composition (anti-tumor medicament composition); the carrier includes diluent, excipient, filler, binder, wetting agent, disintegrating agent, absorption enhancer, surfactant, adsorption carrier, lubricant, etc. which are conventional in the pharmaceutical field.
The product (medicine) or antitumor composition (antitumor medicine composition) can be made into injection, tablet, powder, granule, capsule, oral liquid, paste, cream, etc.; the medicaments in various dosage forms can be prepared according to the conventional method in the pharmaceutical field.
The above product (drug) or antitumor composition (antitumor drug composition) can be introduced into body such as muscle, intradermal, subcutaneous, vein, mucosal tissue by injection, spray, nasal drop, eye drop, penetration, absorption, physical or chemical mediated method; or mixed or coated with other materials and introduced into body.
The invention discovers that the SGLT2 inhibitor Canagliflozin has new application in research: can inhibit the expression and function of P-glycoprotein in tumor cells, and inhibit autophagy phenomenon of stress protection of tumor cells in the face of chemotherapy drug treatment, thereby improving the sensitivity of tumor cells to chemotherapy drugs and enhancing the treatment effect of drugs. The inhibitor can exist in the form of liquid, dry powder or extract, can be prepared into various dosage forms with health care or medicinal efficacy such as capsules, tablets, injection, pills, granules, powder and the like with various conventional auxiliary materials, and can also be combined with other chemotherapeutic drugs for treating malignant tumors. And the SGLT2 inhibitor Canagliflozin has been subjected to clinical safety tests and is therefore relatively safe.
Drawings
FIG. 1 is a graph showing that CAN enhances the anti-tumor effect of Doxorubicin in different tumor cell lines. The final concentration of CAN is 40 mu M, and the final concentration of Doxorubicin is 0.175-4.2 mu M; data are expressed as mean ± standard deviation (n ═ 3), a P <0.05, aa P <0.01vs negative control (Nor); b P <0.05, bb P <0.01vs CAN alone control (CAN 40); c P <0.05, cc P <0.01vs correspond to the same concentration Doxo group.
FIG. 2 shows the accumulation of Doxorubicin in different tumor cell lines. The final concentration of Doxorubicin is 0.35 mu M; data are expressed as mean ± standard deviation (n ═ 3).
FIG. 3 is a graph showing the effect of CAN on the accumulation of Doxorubicin in different tumor cell lines. The final concentration of CAN is 40 mu M, and the final concentration of Doxorubicin is 0.35 mu M; data are presented as mean ± standard deviation (n ═ 3) —, # P <0.001vs Doxorubicin alone control (Doxo).
FIG. 4 is a graph showing the effect of CAN and Doxorubicin on P-glycoprotein expression at protein level and mRNA level. Nor, normal control; CAN, the administration concentration of the CAN is 40 MuM; doxo, Doxorubicin was administered at a concentration of 0.35. mu.M. Data are expressed as mean ± standard deviation (n ═ 3) —, P <0.05vs Nor.
FIG. 5 shows the mechanism of the effect of CAN and Doxorubicin on P-glycoprotein. The concentration of CAN was 40. mu.M, that of Doxorubicin was 0.35. mu.M, that of MG132 was 5. mu.M, and that of Bafilomycin A1 was 10 nM.
FIG. 6 shows that CAN inhibits autophagy in HepG2 cells stimulated by Doxorubicin. The final concentration of CAN was 40. mu.M, and the final concentration of Doxorubicin was 0.35. mu.M.
FIG. 7 shows the early stage of CAN inhibition of autophagy in HepG2 cells. The final concentration of CAN was 40. mu.M, the final concentration of Rapamycin was 0.5. mu.M, and the final concentration of Bafilomycin A1 was 10 nM.
FIG. 8 shows CAN activation of ULK-Ser757 phosphorylation sites. The final concentration of CAN was 40. mu.M, and the final concentration of Doxorubicin was 0.35. mu.M.
FIG. 9 is a graph showing the effect of CAN and Doxorubicin on tumor volume in nude mice. N6-8, P <0.05vs Nor.
FIG. 10 shows the effect of CAN and Doxorubicin on pathological changes of tumor tissues in nude mice. The magnification of the picture is 40 x, and the area indicated by the arrow in the lower right corner of the picture is magnified to 200 x.
Detailed Description
The following examples are given to facilitate a better understanding of the invention, but do not limit the invention. The experimental procedures in the following examples are conventional unless otherwise specified. The test materials used in the following examples were purchased from a conventional biochemical reagent store unless otherwise specified. The quantitative tests in the following examples, all set up three replicates and the results averaged.
Half maximal Inhibitory Concentrations (IC) in the following examples50) Refers to the concentration of the drug Doxorubicin in inducing apoptosis of 50% of tumor cells.
Example 1 detection of synergistic Effect of CAN and antitumor drug Doxorubicin
First, experimental material
1. A compound: canagliflozin (CAN) and Doxorubicin (Doxorubicin, Doxo) are products of medchemexpress (mce). The Kancogl stock number is HY-10451, and the Adriamycin stock number is HY-15142A.
2. Cell source: human liver cancer cells (HepG2), human colon cancer cells (HCT116) and human breast cancer cells (MCF7) are all products of Shanghai cell bank of Chinese academy of sciences, and adriamycin-resistant human liver cancer cells (HepG2-ADR) are products of Shanghai Egyptian Biotech Co.
3. Cell biological reagents: DMEM high-sugar powder medium (CP2205) and pancreatin (Trypsin, R001100) are all products of ThermoFisher Scientific company; fetal bovine serum (FBS, P30-3302) is a product of Pan Biotech. Thiazole blue reagent (MTT, 97062) 378) is a product of the Amreso company.
4. Experiment consumables: cell culture plates of 10cm, 6-well and 12-well plates for cell culture, 24-well plates, pipettes, and centrifuge tubes are all products of Biofil corporation.
5. An experimental instrument: the microplate reader (Epoch) is a product of Biotek corporation, usa, and the flow cytometer (BD Accuri C6) is a product of BD corporation, usa.
Second, Experimental methods
1. The cytotoxicity test of the combined effect of CAN and Doxorubicin was carried out as follows:
(1) MTT was formulated into a working solution at a concentration of 5mg/mL in sterile PBS and filter sterilized through a 0.22 μm filter.
(2) Cells with good growth state (human liver cancer cell (HepG2), human breast cancer cell (MCF7) or adriamycin-resistant human liver cancer cell (HepG2-ADR)) are cultured at 4 × 103The density of individual cells/well was seeded into 96-well plates in DMEM cell culture medium containing 10% fetal bovine serum. The cells are treated by CAN and/or Doxorubicin, and different experimental groups are set according to different administration doses, the final concentration of the Doxorubicin ranges from 0.0 mu M to 2.8 mu M, and the final concentration of the CAN is 40 mu M. Three replicate wells were set for each group, and 200 μ Ι _ of PBS solution was added to the wells around the 96-well plate to prevent evaporation of the medium. The control group and each experimental group were administered at the following doses:
nor: the final concentration of CAN is 0.0 mu M; the final concentration of Doxorubicin is 0.0 μ M;
CAN 40: the final concentration of CAN is 40 MuM; the final concentration of Doxorubicin is 0.0 μ M;
doxo0.025: the final concentration of CAN is 0.0 mu M; the final concentration of Doxorubicin is 0.025 mu M;
doxo0.05: the final concentration of CAN is 0.0 mu M; the final concentration of Doxorubicin is 0.05. mu.M;
doxo0.175: the final concentration of CAN is 0.0 mu M; the final concentration of Doxorubicin is 0.175. mu.M;
doxo0.35: the final concentration of CAN is 0.0 mu M; the final concentration of Doxorubicin is 0.35. mu.M;
doxo0.7: the final concentration of CAN is 0.0 mu M; the final concentration of Doxorubicin is 0.7 mu M;
doxo1.4: the final concentration of CAN is 0.0 mu M; the final concentration of Doxorubicin is 1.4. mu.M;
doxo2.8: the final concentration of CAN is 0.0 mu M; the final concentration of Doxorubicin is 2.8 μ M;
CAN40+ doxo 0.025: the final concentration of CAN is 40 MuM; the final concentration of Doxorubicin is 0.025 mu M;
CAN40+ doxo 0.05: the final concentration of CAN is 40 MuM; the final concentration of Doxorubicin is 0.05. mu.M;
CAN40+ doxo 0.175: the final concentration of CAN is 40 MuM; the final concentration of Doxorubicin is 0.175. mu.M;
CAN40+ doxo 0.35: the final concentration of CAN is 40 MuM; the final concentration of Doxorubicin is 0.35. mu.M;
CAN40+ doxo 0.7: the final concentration of CAN is 40 MuM; the final concentration of Doxorubicin is 0.7 mu M;
CAN40+ doxo 1.4: the final concentration of CAN is 40 MuM; the final concentration of Doxorubicin is 1.4. mu.M;
CAN40+ doxo 2.8: the final concentration of CAN is 40 MuM; the final concentration of Doxorubicin was 2.8. mu.M.
(3) After 48 hours of incubation, 10 μ L of MTT solution was added to each well, and after 4 hours of incubation, the medium was aspirated and 100 μ L DMSO was added to each well to dissolve the blue-violet formazan crystals in each well. The measurement was performed by microplate spectrophotometry at a wavelength of 490nm, and the half-maximal Inhibitory Concentration (IC) was calculated50)。
2. In order to search for a mechanism of synergistic action between CAN and Doxorubicin, the characteristic that Doxorubicin has fluorescence per se is utilized, and the content of adriamycin in cells is detected by a flow cytometry method so as to observe whether CAN improves drug accumulation in tumor cells to increase cytotoxicity of the tumor cells. The method comprises the following specific steps:
1) sensitivity of different tumor cells to Doxorubicin: MCF-7 cells, HepG2 cells and HepG2-ADR cells were divided into two groups: nor group and Doxo group. Doxo was cultured at a final concentration of 0.35. mu.M for 12 hours.
2) Accumulation of Doxorubicin drug: HepG2 and HepG2-ADR cells were divided into three groups: nor group, Doxo group and CAN + Doxo group. The final concentration of CAN and Doxo were 40. mu.M and 0.7. mu.M, respectively, and the culture was performed for 12 hours.
Each group of cells was trypsinized into single cells, harvested by centrifugation and washed twice with PBS to exclude residual Doxorubicin in the medium from interfering with subsequent measurements. And (4) resuspending the cells with PBS and detecting on a machine. Since Doxorubicin can emit red fluorescence under excitation of green laser (around 535 nm) with a peak around 620nm, the mean fluorescence intensity in cells can be measured by setting FL2 channel to assess accumulation of Doxorubicin.
Third, experimental results
1. The combination of CAN and Doxorubicin CAN enhance the effect of chemotherapy
To determine the effect of CAN on tumor cells, cytotoxicity assays for CAN and Doxorubicin were performed on three tumor cells, human liver cancer cell (HepG2), human breast cancer cell (MCF7), or Doxorubicin-resistant human liver cancer cell (HepG 2-ADR).
The results are shown in FIG. 1. The results show that: the combination showed a stronger cytotoxic effect for all three types of tumor cells than Doxorubicin, which was used alone at the same dose in each combination. IC of all three cells after combination compared to Doxorubicin alone at the same dose50All significantly reduced, wherein the IC of HepG2 cells50IC of HepG2-ADR cells decreased from 0.871. mu.M to 0.265. mu.M50From 1.743. mu.M to 0.894. mu.M.
2. CAN CAN increase drug accumulation of Doxorubicin in tumor cells
The accumulation of Doxorubicin by different tumor cells under normal conditions is shown in fig. 2. The results show that: the maximum concentration of the Doxorubicin drug in MCF-7 cells, the second highest in HepG2 cells and the lowest in HepG2-ADR cells indicate that the degree of absorption of Doxorubicin by different tumor cell lines is different, which may be related to the difference in expression of P-glycoprotein in different cell lines.
The accumulation of Doxorubicin by different tumor cells with the addition of CAN is shown in fig. 3. The results show that: for HepG2 cells and HepG2-ADR cells, the intracellular fluorescence intensity of the CAN + Doxo combined treatment group was significantly higher than that of the Doxo group alone, increased by 23.5% and 53.1% respectively, indicating that CAN promote accumulation of Doxorubicin in tumor cells.
Example 2 Effect of CAN on P-glycoprotein expression level and mechanism study
First, experimental material
1. Biochemical reagents: p-gp (ab170904) is a product of Abcam corporation; p62(#88588) is a product of Cell Signal Technology corporation; LC3(L7543) is a product of Sigma; the β -actin antibody (A1978) is a product of Sigma Co; the proteasome inhibitor MG132(HY-13259) is a product of MedChemexpress; the autophagy inhibitor BafA1(HY-100558) is a product of MedChemexpress.
2. Molecular biological reagents: siRNA is a product of acute bio-technology ltd, guangzhou; trizol is a product of Invitrogen corporation (15596-026); oligo dT, RNase inhibitor (RRI), dNTP (10mM) M-MLV reverse transcriptase, M-MLV 5 XBuffer, SYBR Green are all products of Takara corporation; primers were synthesized by Invitrogen, and the primer sequences are shown in Table 1.
TABLE 1 primer sequences
Primer and method for producing the same Forward sequence (Forward) Reverse sequence (Reverse) Sequence number of NCBI Length of
Human P-gp TTGCTGCTTACATTCAGGTTTCA AGCCTATCTCCTGTCGCATTA NM_000927 105
Human P62 GCACCCCAATGTGATCTGC CGCTACACAAGTCGTAGTCTGG NM_001142299 93
Human Actin CATGTACGTTGCTATCCAGGC CTCCTTAATGTCACGCACGAT NM_001101 250
3. An experimental instrument: vertical electrophoresis tank/vertical spin tank (Tanon) is a product of Shanghai Tianneng technologies, Inc. The real-time fluorescent quantitative PCR instrument (Qtowering 2.2) is a product of analytical jena, Germany.
The other reagents were the same as in example 1.
Second, Experimental methods
1. Western Blotting detection of protein expression level of P-glycoprotein in different tumor cell lines
The expression level of P-glycoprotein in different tumor cell lines is detected, and the specific steps are as follows:
1) collecting a protein sample: cells with good growth state are divided into 4X 10 cells3The density of each cell/well was inoculated into a 96-well plate of DMEM cell culture medium containing 10% fetal bovine serum, and the cells were subjected to drug administration treatment using CAN and/or Doxorubicin, and cultured after the drug administration treatment. The well plates after plating, dosing, incubation for a specified time were removed from the cells and placed on ice. The culture medium is aspirated, and each well is washed twice by gently adding a proper amount of PBS along the wall of the well, and the residual culture medium is removed. And adding a proper amount of cell lysate according to the number of the cells, and standing for 5-10 minutes on ice. And fully blowing and beating the cells in the hole, collecting cell lysate into an EP tube, placing the EP tube into a refrigerated centrifuge for 10 minutes at 4 ℃, and centrifuging at 12000rpm, wherein the supernatant is the collected protein sample.
2) Protein concentration determination: and (3) adding 2 mu L of protein sample into each hole of a 96-well plate, adding and mixing with Coomassie brilliant blue G250200 mu L, and detecting absorbance at 595nm by using an enzyme-linked immunosorbent assay. The absorbance of a series of BSA standards of known concentration was measured in the same manner, and a standard curve of concentration versus absorbance was plotted to calculate the concentration of the unknown protein sample.
3) Electrophoretic sample preparation and gel electrophoresis: according to the results of the concentration measurement in the previous step, each sample was leveled into an electrophoresis sample having the same protein content and the same volume, and 5 Xloading buffer was added to denature the protein by a metal bath at 100 ℃ for 10 minutes. And (3) placing the prepared rubber plate in an electrophoresis tank, adding electrophoresis liquid, spotting, adding protein marker, and then starting rubber running. Firstly running at constant voltage of 80V for 30 minutes, and after running the upper concentrated gel, performing electrophoresis at constant voltage of 120V until the electrophoresis is finished.
4) Film transfer: and taking out the gel after electrophoresis, sequentially assembling membrane transferring clamps from the negative electrode to the positive electrode according to the sandwich structure of the sponge, the filter paper, the gel, the NC membrane, the filter paper and the sponge, placing the membrane transferring clamps in a membrane transferring groove, pouring membrane transferring liquid, and transferring the membrane for 1.5 hours at constant pressure of 80V.
5) And (3) sealing: preparing 5% skimmed milk (TBST), taking out the NC membrane after membrane transfer, washing with TBST for 3 min, placing in the sealing solution, and sealing at 40rpm for 1-2 hr.
6) Incubating the primary antibody: primary antibodies to the desired target protein were prepared in advance and diluted with 3% BSA (TBST). The blocking solution was removed, the desired band of interest was cut off according to the size of the protein of interest, primary antibody was added, and the mixture was incubated overnight at 4 ℃.
7) Washing the membrane: primary antibody was recovered and TBST was added to wash the membrane for 10 min at 120rpm, repeated 4 times.
8) Incubation of secondary antibody: according to the source of the primary antibody, the secondary antibodies of the mouse, the rabbit and the goat are respectively incubated, and the secondary antibodies are diluted by 2.5% skimmed milk (TBST formula) and shaken at 40rpm for 1-2 hours at room temperature.
9) Washing the membrane: the secondary antibody was recovered and TBST was added to wash the membrane at 120rpm for 10 minutes, which was repeated 5 times.
10) Incubation substrate: the film was carefully removed from the TBST, gently washed in ultrapure water, and drained slightly on absorbent paper. And uniformly dripping the prepared ECL substrate onto the protein film, reacting for 2 minutes, and then placing the film into an exposure clamp to enter a dark room for development.
11) And (3) developing: and (3) placing the film into the exposure clamp, exposing for a plurality of minutes, and adjusting the tabletting time according to the depth of the film strip. And taking out the film, sequentially placing the film in a developing solution and a fixing solution, washing the film by using clear water, and drying the film.
12) And (3) analysis results: and marking the developed film, scanning the film into a gray mode picture, and analyzing the result.
2. RT-qPCR detection of P-glycoprotein mRNA expression level
(1) Extraction of Total RNA
1) Cells with good growth state are divided into 1.5X 10 cells5The density of each cell/well was inoculated into a 6-well plate of DMEM cell culture medium containing 10% fetal bovine serum, and the cells were subjected to drug administration treatment using CAN and/or Doxorubicin, and cultured after the drug administration treatment. The well plates after plating, dosing, incubation for a specified time were removed from the cells and placed on ice. The medium was discarded and 1mL of PBS solution was added to each well for 2 washes and 1mL of RNAioso Plus was added. The cells were lysed on ice and placed on a shaker at 120rpm for 5 minutes before being collected in a 1.5mL centrifuge tube.
2) 200 mu L of chloroform is added into each tube of sample, and the mixture is placed on a shaking instrument to be vigorously shaken and emulsified, and then is kept stand for 10 minutes on ice until delamination.
3) The tube was transferred to a refrigerated centrifuge and centrifuged at 12000rpm for 10 minutes at 4 ℃ to see that the liquid in the tube was divided into three layers, and the uppermost clear liquid was aspirated into a new tube, taking care not to aspirate the lower white liquid. Adding isopropanol with the same volume, slightly reversing the upper part and the lower part, mixing evenly, and standing for 10 minutes on ice.
4) The tube was transferred to a refrigerated centrifuge and centrifuged at 12000rpm for 10 minutes at 4 ℃ to allow a small amount of white precipitate to be observed at the bottom of the tube, and the supernatant was discarded and 1mL of 75% ethanol (DEPC in water) was added to each tube. The tube wall was washed by gently inverting the top and bottom, centrifuged at 12000rpm at 4 ℃ for 5 minutes, and the supernatant was discarded.
5) After repeated washing 2 times, the supernatant was discarded, the tube was centrifuged for 5 minutes, and the residual liquid was carefully aspirated off with a micropipette.
6) The centrifuge tube cap was opened and air dried for 8 minutes, and 20. mu.L of DEPC water was added to each tube to dissolve the precipitate. RNA concentration was determined using Nanodrop and leveled by DEPC water according to concentration. If the reverse transcription is not to be performed next, the sample is stored at-80 ℃.
(2) Reverse transcription of mRNA
1) Preparing an RNA sample according to the previous step, placing the RNA sample on ice, and preparing a reverse transcription system according to the following formula: 1 μ L of RNA template, 1 μ L of Oligo dT 1 μ L, M-MLV Reverse Transcriptase 1 μ L, 4 μ L, RRI 1 μ L of 5 XM-MLV buffer, and 11 μ L of 10Mm dNTP 1 μ L, DEPC water.
2) And (2) fully and uniformly mixing the system prepared in the step 1) to avoid bubbles, and carrying out reverse transcription on a PCR instrument. The setting procedure is as follows: 40 ℃, 1 hour → 70 ℃, 15 minutes → 4 ℃ for a period of time.
(3) mRNA real-time fluorescent quantitative PCR
1) Preparing a qPCR reaction system from a cDNA sample obtained by reverse transcription according to the following formula: cDNA 0.5. mu.L, Forward primer 0.5. mu.L, Reverse primer 0.5. mu.L, SYBR Green 5. mu. L, DEPC water 3.5. mu.L.
2) And (2) subpackaging the prepared sample in the step 1) into 8 connecting pipes, and fully and uniformly mixing to avoid bubbles. The qPCR reaction was performed according to the following procedure: the signal was collected at 90 ℃, 1 min, 95 ℃, 15 sec, 60 ℃, 45 sec, 40 cycles.
3) And (3) making three multiple holes for each sample, counting the experimental results, and comparing the ratio of the target gene and the reference gene of each sample. The sequences of the target gene and the reference gene are shown in Table 1.
Third, experimental results
1. Effect of CAN and Doxorubicin on P-glycoprotein expression levels
The expression level of P-glycoprotein in different tumor cell lines was measured and the results are shown in FIG. 4. The results show that: the expression of P-glycoprotein was differentiated in different tumor cell lines, with the highest P-glycoprotein content in HepG2-ADR cells, and the next in HepG2 cells, with little P-glycoprotein expression in MCF-7 and HCT116 cells (FIG. 4A). The results are consistent with the accumulation of Doxorubicin in each cell line in example 1, indicating that cells with high P-glycoprotein expression have strong efflux ability for Doxorubicin, and can effectively reduce the concentration of Doxorubicin in the cells.
In order to deeply search the action mechanism of CAN, HepG2 cells are selected to carry out subsequent experiments. HepG2 cells were treated with CAN and/or Doxorubicin at the doses shown in FIG. 4B and FIG. 4D. The results show that: when CAN or Doxorubicin was used alone, CAN reduced the P-glycoprotein expression level in HepG2 cells, whereas Doxorubicin increased the P-glycoprotein expression level in HepG2 cells in a dose-dependent relationship (FIG. 4B). When both CAN and Doxorubicin were used in combination, the protein expression level of P-glycoprotein was significantly reduced compared to Doxorubicin alone (FIG. 4D).
To investigate whether reduction of the protein expression level of P-glycoprotein by CAN was involved in affecting the gene expression of P-glycoprotein, the P-glycoprotein mRNA expression levels in each group (Nor group, CAN group, Doxo group and CAN + Doxo group) after 12h and 24h of the administration treatment were measured by RT-qPCR, respectively, and the results are shown in FIG. 4C. The results show that: there was no significant difference in mRNA expression levels of the P-glycoproteins of each group (fig. 4C).
2. Mechanism of influence of CAN and Doxorubicin on P-glycoprotein
From the above experiments, it was found that the effect of CAN on P-glycoprotein expression in HepG2 cells was not due to increased gene expression, and thus it was verified whether CAN decreased protein content by enhancing P-glycoprotein degradation.
Tumor cells HepG2 cells were treated with CAN or Doxorubicin in combination with a proteasome inhibitor MG132 or an autophagy inhibitor BafA1, respectively, at the administration concentrations and the culture time shown in FIG. 5, and the protein expression level of P-glycoprotein was measured by Western Blotting after the treatment.
The results show that: the degradation enhancement of the P-glycoprotein by the CAN is mainly realized by a proteasome degradation way; the reason why Doxorubicin increased the P-glycoprotein content was that gene expression was stimulated by administration (FIG. 5).
Example 3 study of the Effect of CAN and Doxorubicin on HepG2 autophagy
First, experimental material
1. Biochemical reagents: p62(#88588) is a product of Cell Signal Technology corporation; LC3(L7543) is a product of Sigma, ULK1(#8054) is a product of Cell Signal Technology, P-ULK1(#6888P) is a product of Cell Signal Technology, mTOR (#2983) is a product of Cell Signal Technology, P-mTOR (#5536) is a product of Cell Signal Technology, and β -actin antibody (A1978) is a product of Sigma; EBSS (14155-; rapamycin (HY-10219) is a product of MedChemexpress; the autophagy inhibitor Bafilomycin A1(HY-100558) is a product of MedChemexpress.
2. An experimental instrument: the same apparatus as in example 2.
3. Other reagents: the same reagents as in example 1.
Second, Experimental methods
The procedure is as in example 2.
Third, experimental results
1. CAN CAN inhibit autophagy of HepG2 cell caused by Doxorubicin stimulation
HepG2 cells were treated with CAN and/or Doxorubicin at a final CAN concentration of 40. mu.M and a final Doxorubicin concentration of 0.35. mu.M, and the level of autophagy was evaluated at various times (3h, 6h, 12h, 24h, 48 h).
The results are shown in FIG. 6. The results show that: after Doxorubicin acts on HepG2 cells, the expression level of p62 is reduced, the ratio of LC3 II to LC 3I is increased, and autophagy is promoted. The tumor cell is a stress protection mechanism generated by the response of the tumor cell to the stress of the drug, and the induction of autophagy can help to remove damaged organelles and the like, thereby being beneficial to the survival of the cell. And after the CAN is added, the ratio of LC3 II/LC 3I CAN be obviously reduced, and meanwhile, the expression level of p62 is improved, which shows that the CAN CAN effectively inhibit autophagy induced by Doxorubicin.
2. Early stage of CAN inhibition HepG2 cell autophagy
To further determine the effect of CAN on HepG2 autophagy levels, HepG2 autophagy levels were tested using different doses of CAN (0 μ M, 10 μ M, 20 μ M, 40 μ M) and 40 μ M CAN at different time points in culture (0h, 6h, 12h, 24 h).
The results are shown in FIGS. 7A and 7B. The results show that: CAN effectively inhibit autophagy levels, and has a dose-dependent and time-dependent relationship.
The level of autophagy was assessed using CAN in combination with the autophagy inducer EBSS, Rapamycin and the autophagy inhibitor Bafilomycin a 1.
The results are shown in fig. 7C, 7D and 7E. The results show that: CAN CAN inhibit autophagy induced by autophagy inducer EBSS or Rapamycin, and LC3 II/LC 3I ratio is all reduced.
3. CAN activates ULK-Ser757 phosphorylation site, and inhibits autophagy activation
Related studies have shown that, under nutritionally sufficient conditions, high activity mTOR phosphorylates the serine/threonine protein kinase Unc-51-like kinase 1(Unc-51-like kinase, ULK1) Ser757, thereby blocking ULK1 activation, disrupting the interaction between ULK1 and the adenosine 5' -phosphate-activated protein kinase (AMPK), thereby blocking the autophagy activation of the complex of ULK1, the molecular mass 200kD focal adhesion kinase family interacting protein of 200kD (FIP 200), and ATG 13.
HepG2 cells were treated with CAN and/or Doxorubicin at a final CAN concentration of 40. mu.M and a final Doxorubicin concentration of 0.35. mu.M, and p-ULK1(S757) of HepG2 cells was examined 24 hours after treatment.
The results are shown in FIG. 8. The results show that: ULK1 under CAN treatment showed increased phosphorylation at the 757 site, whereas Doxorubicin treatment showed no significant change in the level of phosphorylation of mTOR, indicating that CAN blocks autophagy by activating p-ULK1(Ser 757).
Example 4 model establishment of subcutaneous transplanted tumor in nude mice and drug efficacy evaluation of CAN and Doxorubicin
First, experimental material
1. Experimental animals and food: the male nude mouse (BALB/cSlc-nu/nu) of four-week old is a product of the Guangdong province medical experimental animal center (SPF grade, permit number: Guangdong invigilation word, SCXK (Guangdong) 2013-. SPF grade food is a product of the medical laboratory animal center in guangdong province.
2. Chemical reagents: 10% formalin: 50mL of 40% formalin, NaH2PO4·2H2O 1.75g,Na2HPO4·12H2O3.25 g and deionized water to 500 mL. Anesthetic agent: 10% of urethane injection.
Second, Experimental methods
1. Establishment of tumor-bearing animal model
HepG2 cells were cultured conventionally, digested with trypsin and counted up to the logarithmic growth phase, and after centrifugation, the cell number was adjusted to 10 with serum-free DMEM medium7And adding 10% matrigel, and injecting liver cancer cells at the right thigh part or the armpit of the nude mouse by a subcutaneous injection method. The injection amount is 1 × 10 per nude mouse6Individual cells, 1 week after injection, were observed for tumor growth, excluding non-tumorigenic mice.
2. Group administration
After tumor inoculation, the long diameter and the short diameter of the tumor of the mice are measured by a vernier caliper every day, and after 10 days, the tumor-bearing nude mice are randomly divided into 4 groups when the maximum diameter of the tumor is 5-7 mm, and 5 mice in each group. The administration is carried out in groups. Each group was dosed as follows:
nor: gavage 0.5% sodium carboxymethylcellulose once a day; sterilized PBS solution was injected intraperitoneally every two days.
CAN: dissolving CAN with 0.5% sodium carboxymethylcellulose, and administering in a dosage of 50mg/kg by intragastric administration once a day; injecting sterilized PBS solution into abdominal cavity once every two days; the treatment was given for a total of 25 days.
Doxo (dox): gavage 0.5% sodium carboxymethylcellulose once a day; the Doxorubicin is prepared by a sterilized PBS solution, the administration dosage is 2.5mg/kg, and the administration mode is intraperitoneal injection once every two days; the treatment was given for a total of 25 days.
CAN + Doxo (CAN + DOX): dissolving CAN with 0.5% sodium carboxymethylcellulose, and administering in a dosage of 50mg/kg by intragastric administration once a day; the Doxorubicin is prepared by a sterilized PBS solution, the administration dosage is 2.5mg/kg, and the administration mode is intraperitoneal injection; the treatment was given for a total of 25 days.
3. Observation index
1) Body weight change in nude mice: and recording the weight change of the animals every day in the test process, drawing a weight change curve, and observing the influence of the medicament on the weight of the animals.
2) Survival time and number of surviving nude mice: and recording the death condition of the animals at any time in the test process, calculating the survival time and survival number of the nude mice after the test is finished, and drawing a survival time curve graph.
3) Volume change and tumor growth inhibition rate of tumor: observing the growth condition of the tumor every day in the test process, and calculating the tumor volume according to a formula; after the experiment was completed, the animals were sacrificed, the tumor tissue was removed, and the tumor growth inhibition rate was calculated by weighing according to the formula. Tumor volume is L × W2(L, W represent the longest and shortest tumor diameters, respectively); the tumor growth inhibition rate is [ (A-B)/A ]]X 100% (a and B are tumor weight of control and treatment groups, respectively).
4) Tumor tissues are taken out, fixed by a PB solution containing 40g/L paraformaldehyde overnight, dehydrated by a sucrose solution of 300g/L for 24h, frozen sliced, the thickness of the slice is 25 mu m, HE staining is carried out, and the tumorigenesis condition of HepG2 cells in nude mice is observed.
Third, experimental results
1. Effect of CAN and Doxorubixin on tumor volume in nude mice
The results of the tumor volume and tumor weight changes after the treatment of each group of mice are shown in FIG. 9. The results show that: compared with the Nor group, the growth rate of the tumors in the CAN group is slightly reduced, while the tumors in the Doxo (DOX) group are obviously reduced, but the effect of the CAN + Doxo (CAN + DOX) group is most remarkable. On day 25 post-dose, mean tumor volume in the Nor group reached 545.4mm3The CAN + Doxo (CAN + DOX) group is only 171.3mm3. After the dosing treatment, nude mice were sacrificed and tumor tissue was removed and weighed. The mean tumor weight was 0.34g in the Nor group and 0.15g in the CAN + Doxo (CAN + DOX) group, indicating significant differences (FIG. 9).
2. Effect of CAN and Doxorubixin on pathological changes of tumor tissues of nude mice
After the administration, each group of tumor tissues was sectioned and HE stained.
The results are shown in FIG. 10. The results show that: in the Nor group, the tumor tissue without any drug treatment is composed of tumor parenchyma and a small amount of interstitium, and pathological nuclear division is much, which indicates that the tumor malignancy degree is high. The cells treated by CAN show small area necrosis, and the cells treated by Doxo (DOX) have large tumor cell necrosis, while the tumor tissues of CAN + Doxo (CAN + DOX) group are basically completely necrotic, and the tissues in the tumor are in amorphous red stain without formed cell nucleus. In vivo experiments show that the combination of CAN and Doxorubicin CAN effectively kill HepG2 cells and inhibit tumor growth (figure 10).
The foregoing is only a preferred embodiment of the present invention, and it should be noted that, for those skilled in the art, various modifications and decorations can be made without departing from the technical principle of the present invention, and these modifications and decorations should also be regarded as the protection scope of the present invention.

Claims (10)

  1. Use of an SGLT2 inhibitor or a pharmaceutically acceptable salt, ester thereof in any one of the following a1) -a 8):
    A1) preparing a product for preventing and/or treating tumors;
    A2) preparing a product for enhancing the treatment effect of the antitumor drug or the chemotherapeutic drug;
    A3) preparing a product for inhibiting tumor drug resistance;
    A4) preparing a product for improving the sensitivity of tumor cells to anti-tumor drugs or chemotherapeutic drugs;
    A5) preparing a product for promoting the accumulation of the antitumor drug or the chemotherapeutic drug in the tumor cells;
    A6) preparing a product for reducing the expression level of the P-glycoprotein of the tumor cells;
    A7) preparing a product for inhibiting autophagy induced by an anti-tumor drug, a chemotherapeutic drug or an autophagy inducer;
    A8) preparing the product for inhibiting the growth of the tumor.
  2. 2. A product contains SGLT2 inhibitor or its pharmaceutically acceptable salt and ester as active ingredient;
    the function of the product is any one of the following B1) -B8):
    B1) preventing and/or treating tumors;
    B2) enhancing the therapeutic effect of the anti-tumor drug or the chemotherapeutic drug;
    B3) inhibiting tumor drug resistance;
    B4) improving the sensitivity of tumor cells to anti-tumor drugs or chemotherapeutic drugs;
    B5) promoting the accumulation of the antitumor drug or the chemotherapeutic drug in the tumor cells;
    B6) reducing the expression level of tumor cell P-glycoprotein;
    B7) inhibiting autophagy induced by antineoplastic agent or chemotherapeutic agent or autophagy inducer;
    B8) inhibiting tumor growth.
  3. 3. An anti-tumor composition, which consists of SGLT2 inhibitor or pharmaceutically acceptable salt, ester thereof and anti-tumor drugs or chemotherapeutic drugs.
  4. 4. The anti-tumor composition according to claim 3, wherein: the anti-tumor composition has the functions of any one of the following B1) -B8):
    B1) preventing and/or treating tumors;
    B2) enhancing the therapeutic effect of the anti-tumor drug or the chemotherapeutic drug;
    B3) inhibiting tumor drug resistance;
    B4) improving the sensitivity of tumor cells to anti-tumor drugs or chemotherapeutic drugs;
    B5) promoting the accumulation of the antitumor drug or the chemotherapeutic drug in the tumor cells;
    B6) reducing the expression level of tumor cell P-glycoprotein;
    B7) inhibiting autophagy induced by antineoplastic agent or chemotherapeutic agent or autophagy inducer;
    B8) inhibiting tumor growth.
  5. 5. The use according to claim 1 or the product according to claim 2 or the anti-tumor composition according to claim 3 or 4, characterized in that: the expression level of the tumor cell P-glycoprotein is reduced, namely, the protein expression level of the tumor cell P-glycoprotein is reduced;
    and/or, said reducing the protein expression level of tumor cell P-glycoprotein is achieved by enhancing the degradation of tumor cell P-glycoprotein;
    and/or, said enhancing the degradation of tumor cell P-glycoprotein is effected by a proteasome degradation pathway.
  6. 6. The use according to claim 1 or the product according to claim 2 or the anti-tumor composition according to claim 3 or 4, characterized in that: the autophagy induced by the anti-tumor drug, the chemotherapeutic drug or the autophagy inducer is embodied in that the LC3 II/LC 3I ratio is reduced and/or the protein expression level of p62 is improved;
    and/or inhibiting autophagy induced by the antitumor drug or the chemotherapeutic drug or the autophagy inducer blocks autophagy by activating p-ULK1(Ser 757).
  7. 7. The use according to claim 1 or the product according to claim 2 or the anti-tumor composition according to claim 3 or 4, characterized in that: the inhibition of tumor growth is manifested in a reduction in tumor cell volume and/or a reduction in tumor cell weight.
  8. 8. The use according to claim 1 or the product according to claim 2 or the anti-tumor composition according to claim 3 or 4, characterized in that: the SGLT2 inhibitor is Canagliflozin.
  9. 9. The use according to claim 1 or the product according to claim 2 or the anti-tumor composition according to claim 3 or 4, characterized in that: the anti-tumor drug or the chemotherapeutic drug is an antibiotic anti-tumor drug or a chemotherapeutic drug;
    and/or the antibiotic anti-tumor drug or the chemotherapeutic drug is adriamycin Doxorubicin.
  10. 10. The use according to claim 1 or the product according to claim 2 or the anti-tumor composition according to claim 3 or 4, characterized in that: the tumor is liver cancer, colon cancer or breast cancer;
    and/or, the tumor cell is liver cancer cell, colon cancer cell or breast cancer cell;
    and/or, the product is a medicament.
CN201910826149.8A 2019-09-03 2019-09-03 Application of SGLT2 inhibitor in preparation of product for improving sensitivity of antitumor drugs Active CN112439067B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910826149.8A CN112439067B (en) 2019-09-03 2019-09-03 Application of SGLT2 inhibitor in preparation of product for improving sensitivity of antitumor drugs

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910826149.8A CN112439067B (en) 2019-09-03 2019-09-03 Application of SGLT2 inhibitor in preparation of product for improving sensitivity of antitumor drugs

Publications (2)

Publication Number Publication Date
CN112439067A true CN112439067A (en) 2021-03-05
CN112439067B CN112439067B (en) 2023-03-17

Family

ID=74734381

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910826149.8A Active CN112439067B (en) 2019-09-03 2019-09-03 Application of SGLT2 inhibitor in preparation of product for improving sensitivity of antitumor drugs

Country Status (1)

Country Link
CN (1) CN112439067B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022161510A1 (en) * 2021-01-29 2022-08-04 南京医科大学附属逸夫医院 Use of canagliflozin in preparation of drug for treating nodular sclerosis-mediated disease

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016041530A1 (en) * 2014-09-16 2016-03-24 Zentiva, K.S. Complexes of canagliflozin and cyclodextrins
WO2016134486A1 (en) * 2015-02-27 2016-09-01 Steinberg Gregory Use of canagliflozin and derivatives thereof in the treatment of cancer
CN110038009A (en) * 2019-05-13 2019-07-23 浙江大学 Canagliflozin application in preparation of anti-tumor drugs

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016041530A1 (en) * 2014-09-16 2016-03-24 Zentiva, K.S. Complexes of canagliflozin and cyclodextrins
WO2016134486A1 (en) * 2015-02-27 2016-09-01 Steinberg Gregory Use of canagliflozin and derivatives thereof in the treatment of cancer
CN110038009A (en) * 2019-05-13 2019-07-23 浙江大学 Canagliflozin application in preparation of anti-tumor drugs

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
HAMID R. SAMARI ET AL.: "Inhibition of Hepatocytic Autophagy by Adenosine, Aminoimidazole-4-carboxamide Riboside, and N6-Mercaptopurine Riboside", 《THE JOURNAL OF BIOLOGICAL CHEMISTRY》 *
XU, CHENKE ET AL.: "Canagliflozin exerts anti-inflammatory effects by inhibiting intracellular glucose metabolism and promoting autophagy in immune cells", 《BIOCHEMICAL PHARMACOLOGY》 *
张丽波: "坎格列净的临床安全性研究进展", 《临床合理用药杂志》 *
郑克立: "《临床肾移植学》", 30 November 2006, 科学技术文献出版社 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022161510A1 (en) * 2021-01-29 2022-08-04 南京医科大学附属逸夫医院 Use of canagliflozin in preparation of drug for treating nodular sclerosis-mediated disease

Also Published As

Publication number Publication date
CN112439067B (en) 2023-03-17

Similar Documents

Publication Publication Date Title
CN107106580A (en) The composition for the treatment of cancer stem cell
CN106659735A (en) Pharmaceutical solution having anti-tumor effect-enhancing and toxicity-reducing effect, and pharmaceutical composition comprising same
Chen et al. 15, 16-dihydrotanshinone I induces apoptosis and inhibits the proliferation, migration of human osteosarcoma cell line 143B in vitro
Yang et al. Oridonin suppresses human gastric cancer growth in vitro and in vivo via inhibition of VEGF, integrin β3, and PCNA
CN111358952A (en) Anti-tumor pharmaceutical composition, preparation and application thereof
KR20120000579A (en) Method of treating hepatocellular carcinoma
KR20120020639A (en) Composition comprising salvia miltiorrhiza for treatment of prostate cancer
CN112439067B (en) Application of SGLT2 inhibitor in preparation of product for improving sensitivity of antitumor drugs
WO2019237688A1 (en) Application of niemann-pick c1 protein in diagnosis and treatment of cancer
Peng et al. Dehydrocostus lactone inhibits the proliferation of esophageal cancer cells in vivo and in vitro through ROS-mediated apoptosis and autophagy
CN113940936A (en) Application of dihydrotanshinone I in preparation of Nrf2 inhibitor
CN102048727A (en) Application of formononetin in preparing of medicament for restricting angiogenesis
CN114917219B (en) Application of isoorientin combined with cisplatin in preparation of medicine for reversing lung cancer resistance
TW202038960A (en) Combination of a mcl-1 inhibitor and midostaurin, uses and pharmaceutical compositions thereof
CN108714149A (en) Purposes of the garden burnet active constituent in preparing anti-tumor drug
AU2020255063A1 (en) Combined use of A-nor-5α androstane compound drug and anticancer drug
CN112972455B (en) Application of compound in preparation of antitumor drugs
CN103933569B (en) Anti-lung cancer pharmaceutical composition, its application, pill case and package
CN109381470B (en) C21 steroid saponification compound in marsdenia tenacissima and application thereof
EA007004B1 (en) Non-nucleosidic inhibitors of reverse transcriptase as antagonists of cell prolifiration and inducers of cell differentiation
CN115006538B (en) Application of SDCBP inhibitor in preparation of anti-esophageal cancer drugs
RU2784809C2 (en) Combined product containing dicycloplatin and method for its production and use
CN100584352C (en) Fructus Schisandrae Chinensis and extract thereof the purposes in preparation treatment tumor multi-medicine drug-resistant medicine
TWI607752B (en) Use of a composition containing 4-acetyl-antroquinonol b for preparing pharmaceutical compositions for inhibiting growth of ovarian cancer cells
Liu et al. Antiproliferative and apoptotic effects of strychnine in human hepatoma HepG2 cells and its mechanism

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
GR01 Patent grant
GR01 Patent grant