CN107213153B - Application of zoledronic acid compound in preparation of medicine for treating cancer - Google Patents

Application of zoledronic acid compound in preparation of medicine for treating cancer Download PDF

Info

Publication number
CN107213153B
CN107213153B CN201710448499.6A CN201710448499A CN107213153B CN 107213153 B CN107213153 B CN 107213153B CN 201710448499 A CN201710448499 A CN 201710448499A CN 107213153 B CN107213153 B CN 107213153B
Authority
CN
China
Prior art keywords
idp
cancer
cells
zoledronic acid
hct116
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.)
Active
Application number
CN201710448499.6A
Other languages
Chinese (zh)
Other versions
CN107213153A (en
Inventor
彭莹
林建国
邱玲
李珂
吕高超
徐栋
刘清竹
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.)
Jiangsu Institute of Nuclear Medicine
Original Assignee
Jiangsu Institute of Nuclear Medicine
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 Jiangsu Institute of Nuclear Medicine filed Critical Jiangsu Institute of Nuclear Medicine
Priority to CN201710448499.6A priority Critical patent/CN107213153B/en
Publication of CN107213153A publication Critical patent/CN107213153A/en
Application granted granted Critical
Publication of CN107213153B publication Critical patent/CN107213153B/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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides an application of a zoledronic acid compound in preparing a medicine for treating cancer, wherein the zoledronic acid compound can be used for treating cancer, can be widely used for treating breast cancer, neuroblastoma, osteosarcoma, prostatic cancer, liver cancer, gastric cancer and colon cancer, and particularly has a remarkable effect in treating colon cancer, and the IC50 of 72h of the existing zoledronic acid (ZOL) acting on HCT116 cells is 25.8 mu M measured, and the zoledronic acid compound (M) disclosed by the invention4IDP) had an IC50 of 0.98. mu.M on HCT116 cells for 72h, compared to an IC50 of ZOL which is M4IC50 of IDP 26.3 times, M4IDP has a more potent cytotoxic effect on HCT116 cells than ZOL and has a lower IC50 value, M4IDP has great potential to be exploited for the treatment of colon cancer.

Description

Application of zoledronic acid compound in preparation of medicine for treating cancer
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and particularly relates to an application of a zoledronic acid compound in preparation of a cancer treatment drug.
Background
Bisphosphonates (BPs) are organic analogs of inorganic pyrophosphate (PPi) which are organic compounds characterized by two P-C bonds to the same carbon atom. By varying the substituents attached to the carbon atoms, various BPs have been developed. Among them, zoledronic acid (ZOL) is a third generation bisphosphonate, and to carbon atoms are attached a hydroxyl group and an imidazole ring containing 2 nitrogen atoms (FIG. 22).
The zoledronic acid and other bisphosphonates can be specifically combined with hydroxyapatite in bone, have strong bone resorption resisting effect, inhibit actin formation by directly changing the shape of osteoclast, interfere the absorption and reabsorption of osteoclast to bone, also inhibit the release of peptide in bone matrix, break the vicious circle called 'seed and soil mechanism' between tumor and bone formation, and inhibit the growth of tumor cells. Therefore, the zoledronic acid can be widely used for treating osteoporosis, osteitis deformans, hypercalcemia, tumor-related bone diseases and the like in clinic. Recent research shows that zoledronic acid has direct antiproliferative and proapoptotic activity on various cancer cells such as brain glioma, liver cancer, prostate cancer, breast cancer and the like, so that zoledronic acid is expected to be a new weapon for treating the cancers.
Colorectal cancer (CRC) is a cancer originating in the colon or rectum, and is also the third most common cancer in men and the second most common cancer in women worldwide. Colorectal cancer is endemic, with almost 55% occurring in more developed regions. With the development of social economy and the change of life style of people, the incidence rate of colorectal cancer in China is on a rapid rising trend. Colorectal cancer ranks fourth in cancer mortality worldwide, and current therapies often fail to eliminate potential metastatic foci, and new therapies remain to be developed. Some studies have shown that zoledronic acid inhibits colorectal cancer cell proliferation and causes apoptosis, an effect that has also been demonstrated in mouse transplanted tumor models. However, because the research on the aspect is very few, the inhibition effect and the potential mechanism of the bisphosphonate such as zoledronic acid on the growth of colorectal cancer cells are not well researched, and because the molecular weight (Mr 272) of zoledronic acid is small, the influence of the structure is large, and small differences in the structure can generate large influence on the drug effect of the bisphosphonate such as zoledronic acid, the drug effect result of the bisphosphonate such as zoledronic acid on cancer cells is difficult to predict, the optimization of the structure of the bisphosphonate such as zoledronic acid and the further discovery of high-efficiency and low-toxicity drugs have great challenge and practical significance, and the systematic screening of pharmacodynamics has important value. However, the treatment of cancer with the zoledronic acid compound 1-hydroxy-2- (4-methyl-1H-imidazolyl) -ethane-1, 1-bisphosphonic acid has not been disclosed in the prior artThe related reports of the symptoms, such as synthesis of 4-methylimidazole bisphosphate and the like99TcmThe zoledronic acid compound 1-hydroxy-2- (4-methyl-1H-imidazolyl) -ethane-1, 1-diphosphonic acid (M) disclosed in marker4IDP) is used only for99TcmMarking, not proposing M4Whether IDP has an anticancer effect or not, let alone the intensive studies on its inhibitory effect on cancer cells and its potential mechanism, and therefore on bisphosphonates such as M mentioned above4The use of IDP in the treatment of cancer, such as colon cancer, still requires further development and investigation.
Disclosure of Invention
Therefore, the technical problem to be solved by the invention is to provide the application of the zoledronic acid compound in preparing the medicine for treating the cancer.
Therefore, the invention provides the application of the zoledronic acid compound in preparing the medicine for treating the cancer; the zoledronic acid compound has a structure shown in a formula (I):
Figure BDA0001321845030000021
the use of the compound, wherein the cancer comprises breast cancer, neuroblastoma, osteosarcoma, prostate cancer, liver cancer, stomach cancer and colon cancer.
The medicine for treating cancer comprises medicines for resisting human breast cancer cells MDA-MB-231, neuroblastoma cells SH-SY5Y, osteosarcoma cells U2OS, prostate cancer cells PC-3, liver cancer cells SMMC-7721, stomach cancer cells SGC-7901 and colon cancer cells HCT 116.
The invention also provides an anticancer drug which takes the compound shown in the formula (I) as an effective component.
The anticancer drug is a clinically acceptable preparation prepared by taking the compound shown in the formula (I) as an active ingredient and selectively adding conventional auxiliary materials according to a conventional process.
The preparation of the anticancer drug comprises tablets, capsules, granules, syrups, powders, pills, tinctures, vinum, soft extracts, freeze-dried powders, pastilles or mixtures.
The technical scheme of the invention has the following advantages:
1. the invention provides an application of a zoledronic acid compound in preparing a medicine for treating cancer, wherein the zoledronic acid compound can be used for treating cancer, and has an inhibiting effect on the growth of human breast cancer cells MDA-MB-231, neuroblastoma cells SH-SY5Y, osteosarcoma cells U2OS, prostate cancer cells PC-3, liver cancer cells SMMC-7721, stomach cancer cells SGC-7901 and colon cancer cells HCT116, so that the zoledronic acid compound can be widely used for treating breast cancer, neuroblastoma, osteosarcoma, prostate cancer, liver cancer, stomach cancer and colon cancer, particularly has a remarkable effect on the aspect of treating colon cancer, and the IC50 of the existing zoledronic acid (ZOL) which acts on HCT116 cells for 72h is measured to be 25.8 mu M, and the zoledronic acid compound (M39M) disclosed by the invention can be used for treating cancer4IDP) had an IC50 of 0.98. mu.M on HCT116 cells for 72h, compared to an IC50 of ZOL which is M4IC50 of IDP 26.3 times, M4IDP has a more potent cytotoxic effect on HCT116 cells than ZOL and has a lower IC50 value, M4IDP has great potential to be developed and applied to the treatment of colon cancer, and is verified to be M4When the concentration of IDP is up to 5 mu M, the cell activity of human normal liver cells LO2 is not obviously influenced, and when M is used, the cell activity is not influenced4When the concentration of IDP is as high as 2.5 mu M, the cell activity of human normal gastric mucosa cell GES-1 is not obviously influenced, which shows that M4IDP has good selective inhibition effect on human colon cancer cell HCT116, M4IDP is hopefully developed into a high-efficiency and low-toxicity medicament for treating human colon cancer.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the description of the embodiments or the prior art will be briefly described below, and it is obvious that the drawings in the following description are some embodiments of the present invention, and other drawings can be obtained by those skilled in the art without creative efforts.
FIG. 1 is a scheme showing the preparation of 2- (4-methyl-1H-imidazolyl) acetic acid, a compound prepared in example 1 of the present invention1An H-NMR spectrum;
FIG. 2 is a diagram of the preparation of 1-hydroxy-2- (4-methyl-1H-imidazolyl) -ethane-1, 1-bisphosphonic acid, compound prepared in example 1 of the present invention1An H-NMR spectrum;
FIG. 3 shows M in Experimental example 1 of the present invention4FIG. 2 shows the results of experiments in which IDP inhibited MDA-MB-231, SH-SY5Y, U2OS, PC-3, SMMC-7721, SGC-7901 and HCT116 cell proliferation;
FIG. 4 shows M in Experimental example 2 of the present invention4CCK-8 analysis result chart of HCT116 cell proliferation inhibited by IDP with different action time; results are expressed as mean ± SD,. p<0.05,**p<0.01, compared to respective controls;
FIG. 5 shows different concentrations M in Experimental example 2 of the present invention4CCK-8 assay of IDP or ZOL on HCT116 cells for 72 h; results are expressed as mean ± SD,. star.p<0.01;
FIG. 6 shows different concentrations M in Experimental example 2 of the present invention4The result of CCK-8 analysis of the effect of IDP on HCT116, LO2 and GES-1 cells for 72 h; results are expressed as mean ± SD,. star.p<0.01, with the same concentration M4Comparing the cell viability of HCT116 cells under IDP;##p<0.01, compared to respective controls;
FIG. 7 is a graph showing the results of a colony formation experiment in Experimental example 2 of the present invention;
FIG. 8 is a histogram of the colony formation experiment in Experimental example 2 of the present invention; p <0.01, compared to control;
FIG. 9A shows M in Experimental example 3 of the present invention4A flow cytometry analysis result graph of the cell cycle when IDP acts for 0 h;
FIG. 9B shows M in Experimental example 3 of the present invention4A flow cytometry analysis result graph of the cell cycle when IDP acts for 24 h;
FIG. 9C shows M in Experimental example 3 of the present invention4A flow cytometry analysis result graph of the cell cycle when IDP acts for 48 hours;
FIG. 9D shows M in Experimental example 3 of the present invention4A flow cytometry analysis result graph of the cell cycle when IDP acts for 72 h;
FIG. 10 shows M in Experimental example 3 of the present invention4The distribution ratio of the cell population in G1 and S, G2/M phases of different action time of IDP results; results are expressed as mean ± SD,. p<0.05,**p<0.01, compared to respective controls;
FIG. 11 shows M in Experimental example 3 of the present invention4The effect of IDP on HCT116 cell cycle-related protein expression;
FIG. 12 shows the results of the Annexin V/PI double staining assay in Experimental example 3 of the present invention, wherein bar is 50 μm;
FIG. 13 shows the results of Hoechst33258 staining analysis in Experimental example 3 of the present invention;
FIG. 14 shows M in Experimental example 3 of the present invention4The effect of IDP on the expression of apoptosis-related proteins cleared PARP and active-caspase 3;
FIG. 15 shows M in Experimental example 3 of the present invention4Experimental results for IDP promotion of ROS accumulation in HCT116 cells; results are expressed as mean ± SD,. p<0.05,**p<0.01, compared to a control;
FIG. 16 shows M in Experimental example 3 of the present invention4The result of the influence of IDP on the expression of the endoplasmic reticulum stress related marker protein;
FIG. 17 shows the results of MDC staining test in Experimental example 3 of the present invention;
FIG. 18 shows M in Experimental example 3 of the present invention4The effect of IDP on the expression level of autophagy-related proteins;
FIG. 19 shows M in Experimental example 3 of the present invention4The effect of IDP on HCT116 autophagy flow resulted in bar being 20 μm;
FIG. 20 shows M in Experimental example 3 of the present invention4The result of the effect of IDP on the expression of PTEN protein of HCT116 cells and key effector molecules of PI3K/Akt/mTOR pathway;
FIG. 21 shows M in Experimental example 3 of the present invention4The possible mechanistic patterns of IDP-induced G1 phase arrest, apoptosis and autophagy of HCT116 cells;
FIG. 22 is the chemical structure of ZOL.
Detailed Description
The technical solutions of the present invention will be described clearly and completely with reference to the accompanying drawings, and it should be understood that the described embodiments are some, but not all embodiments of the present invention. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention. In addition, the technical features involved in the different embodiments of the present invention described below may be combined with each other as long as they do not conflict with each other.
The compound shown as the formula (I) can be synthesized according to 4-methylimidazole diphosphonic acid and the synthesis method thereof99TcmThe synthesis method of 4-methylimidazole bisphosphate disclosed in marker "can also be synthesized according to the preparation method described in example 1 below.
Example 1
This example provides a process for preparing a compound of formula (I), comprising the steps of:
Figure BDA0001321845030000051
the synthetic route of the compound (I) is as follows:
Figure BDA0001321845030000061
synthesis of Compound Ethyl 2- (4-methyl-1H-imidazolyl) acetate represented by Q1 and formula (A-2)
A150 mL three-necked flask was charged with 8.2g (0.1mol) of 4-methylimidazole represented by the formula (A-1), KOH8.4g (0.15mol), and K2CO313.8g (0.1mol), tetrabutylammonium bromide 0.7g (2mmol) and CH2Cl275mL of the mixture was stirred at room temperature for 0.5h, 11.2mL (0.1mol) of ethyl bromoacetate was slowly added dropwise thereto, the mixture was refluxed for 8h and then filtered, and the filter cake was made of CH2Cl2Washing with 40mL for 2 times, washing the filtrate with 150mL of saturated NaCl solution, separating the layers, and purifying the organic phase with anhydrous Na2SO4After drying, the solvent was distilled off to obtain 10.6g of a tan viscous liquid, i.e., ethyl 2- (4-methyl-1H-imidazolyl) acetate, which is a compound represented by the formula (A-2).
Synthesis of Compound 2- (4-methyl-1H-imidazolyl) acetic acid represented by Q2 and formula (A-3)
Adding 8.4g (0.05mol) of ethyl 2- (4-methyl-1H-imidazolyl) acetate of the compound (A-2) obtained in the step Q1, 100mL of distilled water and 1mL of concentrated hydrochloric acid into a 250mL three-neck flask, heating and refluxing for reaction for 8H, adding 2g of activated carbon, refluxing and decoloring for 0.5H, then carrying out hot filtration to obtain a light brown aqueous solution, distilling to remove the solvent to obtain a light brown viscous liquid, adding acetone into the viscous liquid, stirring, precipitating a solid, carrying out suction filtration, drying, and then recrystallizing by using isopropanol to obtain 6.1g of a white solid, namely the compound 2- (4-methyl-1H-imidazolyl) acetic acid of the compound (A-3), wherein the yield is as follows: 54.8 percent; a white solid; mp: 153-156 ℃; IR (KBr) (. sigma./cm)-1:3328,1644,1603,1450;ESI-MS(m/z):139(M);1H NMR(500MHz,DMSO):9.06(s,1H,CHring),7.45(s,1H,CHring),5.14(s,2H,CH 2COOH),2.22(s,3H,CH 3) ), [ isomers: 9.00(s,1H, CH)ring),7.42(s,1H,CHring),5.10(s,2H,CH 2COOH),2.22(s,3H,CH 3)](see FIG. 1 for1H-NMR spectrum); c6H8N2O2Elemental analysis (calculated)/%: c: 51.38%, H: 5.67%, O: 22.95%, N: 19.99 percent.
Q3, Compound 1-hydroxy-2- (4-methyl-1H-imidazolyl) -ethane-1, 1-bisphosphonic acid (M) of formula (I)4IDP) Synthesis
A250 mL three-necked flask was charged with 5.6g (0.04mol) of 2- (4-methyl-1H-imidazolyl) acetic acid, which is the compound represented by (A-3) obtained in step Q2, 40mL of chlorobenzene, and 85% by mass of H3PO48mL, uniformly mixing, heating the mixture to 100 ℃, stirring and reacting for 0.5H, then slowly dropwise adding 10.4mL of phosphorus trichloride (after dropwise adding is finished for about 0.5H) when the temperature is reduced to 65 ℃, continuously heating to 100 ℃, reacting for 3H, cooling, adding 50mL of 9mol/L hydrochloric acid for reflux reaction for 6H, cooling, pouring the reaction liquid into cold ethanol, separating out a solid, performing suction filtration, drying, and recrystallizing with water to obtain 3.4g of a white solid, namely the target final product, namely the compound 1-hydroxy-2- (4-methyl-1H-imidazolyl) -ethane-1, 1-diphosphonic acid (M) shown in the formula (I)4IDP). The yield is 29.7%; white solid;mp:227-231℃;IR(KBr),σ/cm-1:3398,3200,2930,2875,1665,1620,1448,1245;ESI-MS(m/z):285(M-);1H NMR(500MHz,DMSO):7.54(3,1H,CHring),6.86(s,1H,CHring),4.30(s,2H,NCH 2),2.01(s,3H,CH 3) And [ isomer: 7.78(s,1H, CH)ring),6.57(s,1H,CHring),4.30(s,2H,CH 2COOH),2.14(s,3H,CH 3)](see FIG. 2 for1H-NMR spectrum); c6H12N2O7P2Elemental analysis (calculated)/%: c: 25.19%, H: 4.18%, O: 39.11%, N: 9.83%, P: 21.69 percent.
Example 2
This example provides an anticancer agent containing the compound represented by formula (I) prepared in example 1 as an active ingredient.
Furthermore, the anticancer drug is a clinically acceptable preparation prepared by taking the compound shown in the formula (I) as an active ingredient and selectively adding conventional auxiliary materials according to a conventional process, wherein the preparation comprises tablets, capsules, granules, syrups, powders, pills, tinctures, vinum, soft extracts, freeze-dried powders, lozenges or mixtures.
Examples of the experiments
The compound 1-hydroxy-2- (4-methyl-1H-imidazolyl) -ethane-1, 1-diphosphonic acid (M) shown as the formula (I) in the invention4IDP) inhibitory effect on cancer cells
The following experimental examples relate to the following experimental materials:
fetal Bovine Serum (FBS), antibiotics, DMEM, purchased under Gibco brand in the united states;
hoechst33258, dansyl cadaverine (MDC), MTT, DMSO from Sigma, usa;
annexin V/PI staining kit purchased from Roche;
CCK-8 kit, crystal violet, cell cycle analysis kit, Reactive Oxygen Species (ROS) analysis kit, RIPA lysate, mChery-GFP-LC 3B expressing adenovirus (Ad-mChery-GFP-LC 3B), and antibodies such as p27, Cyclin D1, PARP, active-caspase3, CHOP, Bip, LC3B, PTEN, phospho-GSK-3 beta (Ser9), GSK-3 beta, Bad and the like, were purchased from Biyunnan corporation;
antibodies such as phospho-Rb (Ser780), ERO1L, p62, ATG5, phospho-PDK1(Ser241) were purchased from Abcam;
antibodies such as phospho-Akt (Ser473), Akt, phospho-mTOR (Ser2448), mTOR, phospho-p70S6K (Thr389) and the like were purchased from CST corporation, USA;
PERK, beta-actin, HRP labeled secondary antibody and Western Blotting Luminol reagent are products of Santa Cruz company in the United states;
human breast cancer cell line MDA-MB-231, human neuroblastoma cell line SH-SY5Y, human osteosarcoma cell line U2OS, human prostate cancer cell line PC-3, human liver cancer cell line SMMC-7721, human gastric cancer cell line SGC-7901, human colon cancer cell line HCT116, human liver cell line LO2 and human gastric mucosal cell line GES-1 were purchased from American ATCC company in culture medium of 90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin, 5% CO at 37 deg.C2Culturing under the condition;
medicine preparation: m4IDP is prepared in example 1 and zoledronic acid (ZOL) is a commercially available product, M4Both IDP and ZOL were more than 98% pure, dissolved in sterile water to a concentration of 10mM, and stored at-20 ℃ for use diluted with medium (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin) to the desired concentration.
Experimental example 1M4Inhibitory Effect of IDP on various human cancer cells
This example used CCK-8 to assay for M4Growth inhibitory effect of IDP on human-derived breast cancer cell MDA-MB-231, neuroblastoma cell SH-SY5Y, osteosarcoma cell U2OS, prostate cancer cell PC-3, liver cancer cell SMMC-7721, stomach cancer cell SGC-7901 and colon cancer cell HCT 116. The specific method comprises the following steps:
experimental groups: the above cancer cells were plated in 96-well plates at a density of 4000 cells/well, and 100. mu.L of the medium (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin) was plated in each well at 37 ℃ with 5% CO2After culturing for 24h under the condition, adding the extract into each well respectivelyInto 100. mu.L of a mixture containing M at various concentrations4The medium of IDP, said M4The concentration of IDP was 0.625. mu.M, 1.25. mu.M, 2.5. mu.M, 5. mu.M, 10. mu.M, 20. mu.M, 40. mu.M and 60. mu.M in this order, at 37 ℃ with 5% CO2Culturing under the above conditions for 72h, adding 20 μ L CCK-8 solution into each well, and continuing to culture at 37 deg.C and 5% CO2Culturing for 4h under the condition, and detecting the absorbance value of the culture solution under the condition of 450nm by using a SpetraMax M5 enzyme-labeling instrument to obtain an OD value of an experimental group; control group: only 200. mu.L of medium, M, was added per well4Healthy HCT116 cells with an IDP concentration of 0. mu.M were used as a control group, and OD values of the control group were obtained according to the experiment. Cell viability was expressed as the percentage of the OD of the experimental group to the OD of the control group. The results are shown in FIG. 3, M4IDP has inhibitory effect on all of the above-mentioned cancer cells, and the effect is enhanced with increasing drug concentration. Wherein M is4The IDP has the strongest inhibitory effect on human colon cancer HCT116 cells, which indicates that M4IDP has higher inhibition potential on human colon cancer HCT116 cells.
Experimental example 2M4Inhibitory Effect of IDP on human Colon cancer HCT116 cells
2.1 cytotoxicity assays
2.1.1 detection of M Using CCK-8 assay4Growth inhibitory effect of IDP on HCT116 cells. The specific method comprises the following steps:
experimental groups: HCT116 cells were plated at 4000/well density into 96-well plates with 100. mu.L of the medium (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin) per well at 37 ℃ with 5% CO2After culturing for 24h under the condition, 100 μ L of different M concentrations are added into each well4IDP medium, said medium containing different concentrations of M4M in IDP Medium4The concentrations of IDP were 0.3125. mu.M, 0.625. mu.M, 1.25. mu.M, 2.5. mu.M, 5. mu.M, 10. mu.M, 20. mu.M, 5% CO at 37 ℃ respectively2Culturing under the conditions for 24h, 48h or 72h, adding 20 μ L CCK-8 solution into each well, and continuing to culture at 37 deg.C and 5% CO2Culturing for 4h under the condition, and detecting the absorbance value under the condition of 450nm by using a SpetraMax M5 enzyme-labeling instrument; control group: only 200. mu.L of medium, M, was added per well4Health with 0 μ M concentration of IDPHCT116 cells were used as a control group, and OD values of the control group were obtained according to the experiment. Cell viability was expressed as the percentage of the OD of the experimental group to the OD of the control group, see FIG. 4, M4IDP has inhibitory effect on HCT116 cells, and the effect is enhanced with concentration or time, and after 24 hr, the concentration is as low as 2.5 μ M4IDP can inhibit HCT116 cell activity, and after 48 hr, the concentration of M is as low as 0.3125 μ M4IDP can inhibit HCT116 cell activity, and after 72h, M4The greater ability of IDP to inhibit HCT116 cell activity indicates that M is present4IDP has a high inhibitory potential against HCT116 cells.
2.1.2 comparison of M Using CCK-8 analysis4Growth inhibitory effects of IDP and ZOL on HCT116 cells. The specific method comprises the following steps:
experimental groups: HCT116 cells were plated at 4000/well density into 96-well plates with 100. mu.L of the medium (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin) per well at 37 ℃ with 5% CO2After culturing for 24h under the condition, 100 μ L of different M concentrations are added into each well4IDP or ZOL medium containing M at different concentrations4M in IDP or ZOL Medium4The concentration of IDP or ZOL was 0.625. mu.M, 1.25. mu.M, 2.5. mu.M, 5. mu.M, 10. mu.M, 20. mu.M, respectively, at 37 ℃ with 5% CO2Culturing for 72h, adding 20 μ L CCK-8 solution into each well, culturing for 4h, and detecting absorbance value at 450nm with Speramax M5 enzyme-labeling instrument to obtain M4OD values of IDP or ZOL experimental groups; control group: only 200. mu.L of medium, M, was added per well4Healthy HCT116 cells with 0. mu.M concentration of IDP or ZOL were used as a control group, and OD values of the control group were obtained according to the experiment. Cell viability was expressed as the percentage of the OD of the experimental group to the OD of the control group. The results are shown in FIG. 5, from which M is known4IDP was more cytotoxic than ZOL on HCT116 cells and M was measured4The IC50 of the IDP on HCT116 cells for 72h was 0.98. mu.M, and the IC50 of ZOL on HCT116 cells for 72h was determined to be 25.8. mu.M, ZOL being the same as M4IDP vs. HCT116 at 72h IC50, ZOL at IC50 was M426.3 times IDP, show M4IDP has a stronger cytotoxic effect on HCT116 cells than ZOLAnd has a lower IC50 value, M4IDP is promising for applications in the treatment of colon cancer.
2.1.3 comparison of M Using CCK-8 analysis4Growth inhibitory effects of IDP on human colon cancer cells HCT116 and human liver cells LO2, human gastric mucosal cells GES-1. The specific method comprises the following steps:
spreading human colon cancer cell HCT116, human liver cell LO2, and human gastric mucosa cell GES-1 at 4000/well density to 96-well plate, wherein each well contains 100 μ L of the culture medium (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin), and the culture medium is cultured at 37 deg.C and 5% CO2After culturing for 24h under the condition, 100 μ L of different M concentrations are added into each well4The medium of IDP, said M4The concentration of IDP was 0.625. mu.M, 1.25. mu.M, 2.5. mu.M, 5. mu.M, and 10. mu.M in this order, 5% CO at 37 ℃2Culturing under the above conditions for 72h, adding 20 μ L CCK-8 solution into each well, and continuing to culture at 37 deg.C and 5% CO2Culturing for 4h under the condition, and detecting the absorbance value of the culture solution under the condition of 450nm by using an American SpetraMaxM5 enzyme-labeling instrument to obtain an OD value of an experimental group; control group: only 200. mu.L of medium, M, was added per well4Healthy cells with an IDP concentration of 0. mu.M were used as a control group, and the OD value of the control group was obtained according to the experiment. Cell viability was expressed as the percentage of the OD of the experimental group to the OD of the control group. The results are shown in FIG. 6, M4The cytotoxic effect of IDP on human colon cancer HCT116 cells is stronger than that of human normal liver cells LO2 and human normal gastric mucosa cells GES-1, and the concentration of M is as high as 5 mu M4The inhibition rate of IDP on HCT116 reaches 65%, but the IDP has no obvious influence on the cell activity of LO 2; m at a concentration of up to 2.5. mu.M4The inhibition rate of IDP on HCT116 reaches 55%, but the IDP has no obvious influence on the cell activity of GES-1. This indicates that M is compared with the normal human cells LO2 and GES-14IDP has good selective inhibition effect on human colon cancer cell HCT116, M4IDP is hopefully developed into a high-efficiency and low-toxicity medicament for treating human colon cancer.
2.2 cloning experiments
HCT116 cells were plated at a density of 300/well in 12-well plates with 1mL of the medium per well (90% DMEM, 10)% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin), 5% CO at 37 deg.C2After 24h of incubation under conditions, the medium was replaced with various concentrations of M4IDP medium containing various concentrations of M4M in IDP Medium4The IDP concentrations were 0. mu.M, 0.625. mu.M, 1.25. mu.M, 2.5. mu.M, respectively, and continued at 37 ℃ with 5% CO2After 14 days of culture under the conditions, the HCT116 cells were fixed with methanol and stained with 1% crystal violet for 10min, rinsed well with water and dried in air, and then observed under a microscope and clones containing more than 50 cells were counted. The results are shown in FIGS. 7 and 8, using M4The number of clones of IDP-treated cells was significantly less than that of the cells without M addition4Number of IDP-treated cell clones, this inhibitory effect being dependent on M4The increase in IDP concentration is enhanced.
Experimental example 3M4IDP causes human colon cancer HCT116 cell G1 phase block, apoptosis and autophagy by blocking PI3K/Akt/mTOR
3.1 cell cycle Studies
HCT116 cells were plated at 2 × 105Individual/well density was plated in 6-well plates, 2mL of the medium per well (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin), 5% CO at 37 deg.C2After 24h incubation under conditions, 2.5. mu.M M was used4IDP was treated for 0h (control), 24h, 48h, 72h, respectively. Then, HCT116 cells were collected, fixed with 75% ethanol by volume, and then, HCT116 cells were resuspended in a 50. mu.g/mL PI solution, the treated cells were analyzed by a Flow cytometer of BD company, Germany, and the data obtained were analyzed by Flow Jo software. The results are shown in FIGS. 9A-D and FIG. 10, where it can be seen that M increases with time4IDP causes accumulation of cells in the G1 phase, and cells in the S and G2/M phases gradually decrease.
It is well known that the cell cycle is negatively regulated by CDK, p27, etc., and positively regulated by the cyclin-CDK complex. To study M4The possible mechanism of IDP causing G1 phase block is that the expression of some proteins related to G1/S checkpoint, such as p27, cyclin D1 and phosphine-Rb, is detected by Western Blotting method, and HCT116 cell is treated with 2 × 105Of one/holeDensity plated in 6-well plates, 2mL of the medium per well (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin), 5% CO at 37 ℃2After 24h incubation under conditions, 2.5. mu.M M was used4IDP (dichloro-diphenyl-trichloroethane) treatment of HCT116 cells for 0h, 24h, 48h and 72h, then cracking of HCT116 cells by using a proper amount of RIPA lysate, 12,000 × g high-speed centrifugation for 5min, taking supernatant, measuring the protein concentration of the supernatant by using a BCA method, adding a proper volume of 6 × SDS loading buffer solution into the solution containing 30 mu g of protein, boiling water bath for 5min, then carrying out short-time centrifugation, then loading all the samples into 10% SDS polyacrylamide gel for electrophoresis, setting the voltage for electrophoresis as 100V, transferring the protein on the 10% SDS polyacrylamide gel onto a PVDF membrane by using a Bio-Rad wet electric transfer membrane instrument after the electrophoresis is finished, transferring the protein onto the PVDF membrane by using 100V voltage for 1h, transferring the protein onto the PVDF membrane by using p27, CyclinD1 or pho-Rb antibody with the dilution concentration of 1:1000 after the membrane transfer, and obtaining the detection result shown in figure 114IDP increased the expression of CDK inhibitory molecule p27 and inhibited the expression of cyclin D1 and phospho-Rb (p-Rb), indicating that M4The G1 phase block caused by IDP may be attributed to its effect on p27, CyclinD1 and phospho-Rb.
3.2 apoptosis assay
Experimental group (M)4IDP) HCT116 cells were treated with 3 × 104One/well density was plated in 24-well plates, 500. mu.L of the medium (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin) per well, 5% CO at 37 ℃2After 24h incubation under conditions, 2.5. mu.M M was used4Treating with IDP for 72h, then absorbing the culture medium, adding Annexin V/PI staining solution prepared according to the requirements of the kit, incubating at room temperature for 10min, observing under an Olympus IX51 fluorescence microscope, and taking a picture; the Control group (Control) was performed according to the above experimental group procedure, but only 500. mu.L of the medium was added to each well, i.e., without M4IDP-treated healthy HCT116 cells. The results are shown in FIG. 12, where the arrows indicate apoptotic cells whose cell membranes were stained green by Annexin V-FITC, the circles indicate apoptotic middle and late cells whose nuclei were stained red by PI and dead cells, and M is shown4IDP induced significantly more apoptotic cells than the control group.
Then makeMorphological characteristics of apoptotic cells were visualized by Hoechst33258 staining analysis by staining HCT116 cells with 3 × 104One/well density was plated in 24-well plates, 500. mu.L of the medium (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin) per well, 5% CO at 37 ℃2After 24h incubation under conditions, 2.5. mu.M M was used4IDP was treated for 24h, 48h, 72h, then cells were fixed with 4% paraformaldehyde, then cells were stained with 2. mu.g/mL Hoechst33258 for 5min, observed under an Olympus IX51 fluorescence microscope and photographed, and the Control (Control) was performed as described above, but only 500. mu.L of medium was added per well, i.e., no M added4IDP-treated healthy HCT116 cells. The results are shown in FIG. 13, from which M is known4IDP causes chromatin contraction and fragmentation of HCT116 cells (indicated by arrows).
The indicator proteins of apoptosis, namely clean PARP and active-caspase3, are detected by using a Western Blotting method, namely, the method comprises the step of dissolving HCT116 cells in 3 × 104One/well density was plated in 24-well plates, 500. mu.L of the medium (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin) per well, 5% CO at 37 ℃2After 24h incubation under conditions, 2.5. mu.M M was used4IDP (dichloro-diphenyl-trichloroethane) treatment of HCT116 cells for 0h, 24h, 48h and 72h, cell lysis by using a proper amount of RIPA lysate, high-speed centrifugation for 5min at 12,000 × g, supernatant taking, BCA (burst amplification factor) method for measuring the protein concentration of the supernatant, adding a proper volume of 6 × SDS (sodium dodecyl sulfate) sample buffer solution into the obtained solution containing 30 mu g of protein, boiling water bath for 5min, short-time centrifugation, then full sample loading into 10% SDS polyacrylamide gel for electrophoresis, setting the voltage at 100V, after the electrophoresis is finished, using a Bio-Rad wet electric transfer membrane instrument to transfer the protein on the 10% SDS polyacrylamide gel to a PVDF membrane, using 100V voltage to transfer the membrane for 1h, using PARP, active-caspase3 and β -active antibodies with the dilution concentration of 1:1000 to detect the protein on the 10% SDS-polyacrylamide gel, and the result is shown in figure 144Increased duration of IDP action, which further illustrates M4IDP causes HCT116 cells to undergo apoptosis.
3.3 intracellular ROS accumulation and endoplasmic reticulum stress Effect assay
3.3.1 intracellular ROS accumulation assay
ROS accumulation may play an important role in apoptosis by altering the internal environment of the cell, and intracellular ROS accumulation is detected by contacting HCT116 cells with 2 × 105Individual/well density was plated in 6-well plates, 2mL of the medium per well (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin), 5% CO at 37 deg.C2After 24h incubation under conditions, M was added at various concentrations4The culture medium of IDP is treated for 24h, and the culture medium contains M with different concentrations4M in IDP Medium4Concentrations of IDP were 0. mu.M, 0.625. mu.M, 1.25. mu.M, 2.5. mu.M, respectively, and cells were collected as required in the Reactive Oxygen Species (ROS) assay kit, washed once with PBS, and incubated in 10. mu.M DCFH-DA protected from light at 37 ℃ for 30 min. The fluorescence signal was detected by flow cytometry. The results are shown in FIG. 15, M4Elevated ROS levels in HCT116 cells 24h after IDP treatment, suggesting M4IDP causes the accumulation of ROS.
3.3.2 endoplasmic reticulum stress Effect detection
ROS may cause endoplasmic reticulum stress-mediated unfolded protein reaction to influence cell growth and apoptosis, and the method for detecting endoplasmic reticulum stress effect comprises the following steps of detecting the expression condition of endoplasmic reticulum stress-related protein by using Western Blotting method, and detecting HCT116 cells with 2 × 105Individual/well density was plated in 6-well plates, 2mL of the medium per well (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin), 5% CO at 37 deg.C2After 24h incubation under conditions, 2.5. mu.M M was used4IDP treatment of HCT116 cells for 0h, 24h, 48h and 72h, collecting proteins, electrophoresis, membrane transfer, antibody incubation and exposure according to a conventional method, namely using a proper amount of RIPA lysate to lyse the cells, centrifuging at a high speed of 12,000 × g for 5min, taking supernatant, measuring the protein concentration of the supernatant by using a BCA method, adding a proper volume of 6 × SDS loading buffer solution into the obtained solution containing 30 mu g of protein, carrying out boiling water bath for 5min, centrifuging for a short time, loading all the supernatant into 10% SDS polyacrylamide gel for electrophoresis, setting the voltage at 100V during electrophoresis, and after the electrophoresis is finished, using a Bio-Rad wet electric membrane converter to polymerize 10% SDS by using a Bio-Rad wet electric membrane converterThe protein on the acrylamide gel is transferred to a PVDF membrane, the membrane is transferred for 1h by adopting 100V voltage, CHOP, Bip, ERO1L and PERK and β -actin antibodies with the dilution concentration of 1:1000 are used for detection after the membrane is transferred, the result is shown in figure 16, the expressions of endoplasmic reticulum stress-related proteins CHOP, Bip, ERO1L and PERK are all increased, and the result shows that M is M4IDP causes endoplasmic reticulum stress effects in HCT116 cells.
3.4 autophagy study
M detection by MDC staining4Whether IDP causes autophagy of HCT116 or not, MDC is commonly used as a specific marker stain for autophagic vesicles by contacting HCT116 cells with 2 × 105Individual/well density was plated in 6-well plates, 2mL of the medium per well (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin), 5% CO at 37 deg.C2After 24h incubation under conditions, 2.5. mu.M M was used4IDP was treated for 24h, 48h, 72h, respectively, and the cells were washed once with ice-cold PBS, and then incubated in 50 μ M MDC for 30min at 37 ℃. Cells were washed once with ice-cold PBS, fixed with 4% paraformaldehyde, observed under a fluorescence microscope and photographed, and the Control (Control) was performed as described above, but only 2mL of medium was added per well, i.e., without M4IDP-treated healthy HCT116 cells. The results are shown in FIG. 17, M4IDP caused accumulation of MDC-stained vacuoles, suggesting M4IDP may cause autophagy in HCT116 cells.
Then the expression of the autophagy-related marker protein is detected by using a Western Blotting method, and the HCT116 cells are treated with 2 × 105Individual/well density was plated in 6-well plates, 2mL of the medium per well (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin), 5% CO at 37 deg.C2After 24h incubation under conditions, 2.5. mu.M M was used4IDP treating HCT116 cells for 0h, 24h, 48h and 72h, collecting protein, electrophoresis, transferring membrane, incubating antibody and exposing according to conventional method, i.e. using appropriate amount of RIPA lysate to lyse cells, centrifuging at high speed of 12,000 × g for 5min, collecting supernatant, measuring protein concentration of the supernatant by BCA method, adding appropriate volume of 6 × SDS buffer solution into 30 μ g protein-containing solution, and boiling waterBathing for 5min, then centrifuging for a short time, then loading all samples into 10% SDS polyacrylamide gel for electrophoresis, setting the voltage at 100V during electrophoresis, transferring the proteins on the 10% SDS polyacrylamide gel onto a PVDF membrane by using a Bio-Rad wet electric transfer membrane instrument after the electrophoresis is finished, transferring the membrane for 1h by using the 100V voltage, and then detecting by using LC3B, Atg5, p62 and β -actin antibodies with the dilution concentration of 1:10004IDP causes autophagy in HCT116 cells, but a longer duration of action (72h) may cause diminished autophagy flow.
And finally performing autophagic flow analysis. Ad-mCherry-GFP-LC3B is an adenovirus capable of expressing mCherry-GFP-LC3B fusion protein, can effectively express fusion protein of red fluorescent protein mCherry, green fluorescent protein GFP and LC3B in target cells after infection, presents bright red and green fluorescence, and can be used for detecting autophagy of cells. After infecting the cells, mCherry-GFP-LC3B was present in the cytoplasm as diffuse yellow fluorescence (combined effect of mCherry and GFP) under a fluorescent microscope in the case of non-autophagy; in the case of autophagy, mCherry-GFP-LC3B was concentrated on the autophagosome membrane under a fluorescence microscope and appeared as yellow spots; when autophagosomes fused to lysosomes, they appeared as red spots due to partial quenching of GFP fluorescence. The specific analysis method is as follows: HCT116 cells were plated onto glass-bottomed cell culture dishes, and cells were confluent to 20% -30% for infection: the cells were first washed 2 times with medium and then infected under 20MOI Ad-mCherry-GFP-LC3B for 24h, then 2.5. mu.M M4The infected cells were treated with IDP and observed and photographed using Olympus confocal microscope at treatment times of 0h, 3h, 6h, 16h, 24h, 48h, 72 h. The results are shown in fig. 19, where the mCherry and GFP proteins were uniformly distributed in the cytoplasm at 0h treatment time, indicating that autophagy did not occur; at 3h treatment, the mCherry and GFP proteins were visible under the fluorescent microscope to aggregate and may overlap into yellow spots (bright spots in the figure), indicating autophagosome formation, which may last for up to 24h and follow thereThe increase of the physiological time and the increase of yellow spots (bright spots in the figure) show that autophagosomes are gradually increased and mature, and the autophagy degree is gradually increased; after 48 hours of treatment, red spots are increased under a fluorescence microscope, green signals are reduced, and the red spots (arrows indicate bright spots) can be seen after overlapping, which indicates that autophagosome is fused with lysosome and autophagy flow is increased; at 72h treatment, yellow spots (bright spots within circles) and red spots (arrows indicate bright spots) in the overlaid images were both less than at 48h, indicating a diminished degree of autophagy.
3.5 Signal Path Studies
The PI3K/Akt/mTOR pathway plays an important role in regulating cell cycle, apoptosis and autophagy. To determine M4Whether the inhibitory effect of IDP on HCT116 cells is associated with this pathway, we investigated M4Effect of IDP on representative Signal protein molecules on this Signal pathway HCT116 cells were treated with 2 × 105Individual/well density was plated in 6-well plates, 2mL of the medium per well (90% DMEM, 10% FBS, 100U/mL penicillin, 0.1mg/mL streptomycin), 5% CO at 37 deg.C2After 24h incubation under conditions, 2.5. mu.M M was used4Treating HCT116 cells for 0h, 24h, 48h and 72h with IDP, collecting protein, performing electrophoresis, transferring membrane, incubating antibody and exposing, namely lysing the cells with an appropriate amount of RIPA lysate, centrifuging at 12,000 × g for 5min at high speed, collecting supernatant, measuring the protein concentration of the supernatant by BCA method, adding an appropriate volume of 6 × SDS loading buffer solution to the solution containing 30. mu.g of protein, boiling water bath for 5min, centrifuging for a short time, loading the whole solution to 10% SDS polyacrylamide gel for electrophoresis at a voltage of 100V, transferring the protein on the 10% SDS polyacrylamide gel to PVDF membrane by a Bio-Rad wet electric membrane transfer apparatus after electrophoresis, transferring the protein to membrane at a voltage of 100V for 1h, transferring the protein to membrane by PTEN, phospho-PDK1(p-PDK1), phospho-t (p-phot), Akt, mTOR (mTOR), mTOR-3570, GSP-3526-3670, GSP 19-11-P-3670, and detecting the protein, the protein concentration, the antibody and the antibody are detected by BCA-36454IDP causes downregulation of phospho-Akt, phospho-mTOR, phospho-p70S6K and upregulation of phospho-GSK-3 β on Akt, mTThe total protein content of OR and GSK-3 β has no significant influence, the expression of pro-apoptotic protein Bad which is a downstream molecule of Akt is increased, and the expression of protein of phosphorylated form PDK1 which is an upstream kinase of Akt is reduced, which indicates that M is a protein with a high activity4IDP may act directly on the upstream molecule of PDK 1; increased expression of PTEN, a negative regulatory molecule of PI3K/Akt pathway. These results show that M4IDP could inhibit the PI3K/Akt/mTOR signaling pathway by up-regulating PTEN expression.
In summary, the above studies of this example show that Akt can inhibit p27 expression and increase cyclin D1 expression by inactivating GSK-3 β, thereby positively regulating G1/S cell cycle progression, and therefore, M4IDP-induced G1 phase arrest may be associated with Akt and GSK-3 β, Bad protein is associated with the initiation of apoptosis, non-phosphorylated Bad protein can promote apoptosis by combining with and inactivating anti-apoptotic protein Bcl-2 or Bcl-xL, activated Akt can inhibit apoptosis and promote cell survival by inducing Bad phosphorylation, therefore, M4IDP-induced apoptosis of HCT116 cells may be associated with inhibition of Akt phosphorylation and increased Bad protein expression. mTOR is a downstream effector molecule of Akt, and together with its downstream target p70S6K, exerts a negative regulatory effect on autophagy, M4Autophagy by IDP is likely to be associated with M4IDP is involved in the inhibition of the Akt/mTOR/p70S6K pathway. PDK1, which is an upstream molecule of Akt and a downstream molecule of PIP3, can be phosphorylated by PIP3 and causes phosphorylation of Akt. The tumor suppressor PTEN can negatively regulate the PI3K pathway, dephosphorylate PIP3 into PIP2, and thereby negatively regulate Akt activity. In summary, M4IDP-induced arrest of G1 phase, apoptosis and autophagy in HCT116 cells may be attributed to the production of PTEN, which further affects the downstream molecules PDK1, Akt, mTOR, GSK-3 β and Bad, as shown in fig. 21.
It should be understood that the above examples are only for clarity of illustration and are not intended to limit the embodiments. Other variations and modifications will be apparent to persons skilled in the art in light of the above description. And are neither required nor exhaustive of all embodiments. And obvious variations or modifications therefrom are within the scope of the invention.

Claims (2)

1. The use of a zoledronic acid compound in the preparation of a medicament for treating cancer; the zoledronic acid compound has a structure shown in a formula (I):
Figure 806011DEST_PATH_IMAGE001
(I);
the cancer is selected from breast cancer, prostate cancer and colon cancer.
2. The use according to claim 1, wherein the cancer treatment drug is selected from the group consisting of drugs against human-derived breast cancer cells MDA-MB-231, prostate cancer cells PC-3 and colon cancer cells HCT 116.
CN201710448499.6A 2017-06-14 2017-06-14 Application of zoledronic acid compound in preparation of medicine for treating cancer Active CN107213153B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201710448499.6A CN107213153B (en) 2017-06-14 2017-06-14 Application of zoledronic acid compound in preparation of medicine for treating cancer

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201710448499.6A CN107213153B (en) 2017-06-14 2017-06-14 Application of zoledronic acid compound in preparation of medicine for treating cancer

Publications (2)

Publication Number Publication Date
CN107213153A CN107213153A (en) 2017-09-29
CN107213153B true CN107213153B (en) 2020-09-08

Family

ID=59948565

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201710448499.6A Active CN107213153B (en) 2017-06-14 2017-06-14 Application of zoledronic acid compound in preparation of medicine for treating cancer

Country Status (1)

Country Link
CN (1) CN107213153B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116509847A (en) * 2023-05-10 2023-08-01 重庆医科大学 Application of 1,4-methylimidazole acetic acid in preparation of obesity prevention and treatment medicines or foods

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
4-甲基咪唑双膦酸的合成及其99Tcm标记;翟好真;《同位素》;20101120;第23卷(第4期);225-229 *
Anti-proliferation effect of zoledronic acid on human colon cancer line;Fu-Shi Han et al.;《Asian Pacific Journal of Tropical Medicine》;20160220;第9卷(第2期);164-168 *

Also Published As

Publication number Publication date
CN107213153A (en) 2017-09-29

Similar Documents

Publication Publication Date Title
US20210024482A1 (en) Novel Group of STAT3 Pathway Inhibitors and Cancer Stem Cell Pathway Inhibitors
CN107271675B (en) Anti-human ADRB3 monoclonal antibody and application thereof in disease diagnosis and treatment
Zhang et al. Evaluation of anticancer effect in vitro and in vivo of iridium (III) complexes on gastric carcinoma SGC-7901 cells
Ji et al. Antineoplastic effects and mechanisms of micheliolide in acute myelogenous leukemia stem cells
Liu et al. Effects of the tyrosine protein kinase inhibitor genistein on the proliferation, activation of cultured rat hepatic stellate cells
Yu et al. Discovery of an orally selective inhibitor of signal transducer and activator of transcription 3 using advanced multiple ligand simultaneous docking
Zhang et al. Inhibition of PDGF-BB-induced proliferation and migration in VSMCs by proanthocyanidin A2: Involvement of KDR and Jak-2/STAT-3/cPLA2 signaling pathways
Chipoline et al. Molecular mechanism of action of new 1, 4-naphthoquinones tethered to 1, 2, 3-1H-triazoles with cytotoxic and selective effect against oral squamous cell carcinoma
Sarkar et al. Epigallocatechin-3-gallate inhibits osteoclastic differentiation by modulating mitophagy and mitochondrial functions
CN112457281B (en) Small molecule inhibitor for blocking combination of COVID-19 spinous protein and human angiotensin converting enzyme 2 and application thereof
Pei et al. Selective STAT3 inhibitor alantolactone ameliorates osteoarthritis via regulating chondrocyte autophagy and cartilage homeostasis
Kwak et al. Blood-brain barrier-permeable fluorone-labeled dieckols acting as neuronal ER stress signaling inhibitors
CA2387591A1 (en) Use of indirubine derivatives for making medicines
Prateep et al. Avicequinone B sensitizes anoikis in human lung cancer cells
CN107213153B (en) Application of zoledronic acid compound in preparation of medicine for treating cancer
CN102438612A (en) Nitrile derivatives and their pharmaceutical use and compositions
Wang et al. Paeonol protects mitochondrial injury and prevents pulmonary vascular remodeling in hypoxia
Feng et al. Scaffold hopping of celastrol provides derivatives containing pepper ring, pyrazine and oxazole substructures as potent autophagy inducers against breast cancer cell line MCF-7
JP6442615B2 (en) Compounds containing indoleacetic acid core structure and their applications
Wang et al. MCU Inhibitor Ruthenium Red Alleviates the Osteoclastogenesis and Ovariectomized Osteoporosis via Suppressing RANKL‐Induced ROS Production and NFATc1 Activation through P38 MAPK Signaling Pathway
Wang et al. Anthraquinone-bridged diruthenium (II) complexes inhibit migration and invasion of human hepatocarcinoma MHCC97-H cells
Wang et al. Garcinol and its analogues: Synthesis, cytotoxic activity and mechanistic investigation
Chan et al. Enantiomerically pure β-dipeptide derivative induces anticancer activity against human hormone-refractory prostate cancer through both PI3K/Akt-dependent and-independent pathways
Bai et al. A novel mitochondria‐targeting compound exerts therapeutic effects against melanoma by inducing mitochondria‐mediated apoptosis and autophagy in vitro and in vivo
Jin et al. GSK 650394 inhibits osteoclasts differentiation and prevents bone loss via promoting the activities of antioxidant enzymes in vitro and in vivo

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