CN111228269A - Application of cepharanthine and its salt as iron death inducer in preparing antitumor drugs - Google Patents

Application of cepharanthine and its salt as iron death inducer in preparing antitumor drugs Download PDF

Info

Publication number
CN111228269A
CN111228269A CN202010256288.4A CN202010256288A CN111228269A CN 111228269 A CN111228269 A CN 111228269A CN 202010256288 A CN202010256288 A CN 202010256288A CN 111228269 A CN111228269 A CN 111228269A
Authority
CN
China
Prior art keywords
hydrochloride
cepharanthine
cells
prostate cancer
stephanine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202010256288.4A
Other languages
Chinese (zh)
Inventor
王一飞
王瑶
黄泽秀
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.)
Jinan University
Guangzhou Jinan Biomedicine Research and Development Base Co Ltd
Original Assignee
Jinan 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 Jinan University filed Critical Jinan University
Priority to CN202010256288.4A priority Critical patent/CN111228269A/en
Publication of CN111228269A publication Critical patent/CN111228269A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4741Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having oxygen as a ring hetero atom, e.g. tubocuraran derivatives, noscapine, bicuculline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • 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)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to the technical field of medicines, and provides application of stephanine and salts thereof as an iron death inducer in preparation of antitumor drugs. Therefore, the stephanine and the salt thereof have good anticancer effect and provide a new effective treatment medicine for patients with tumor diseases.

Description

Application of cepharanthine and its salt as iron death inducer in preparing antitumor drugs
Technical Field
The invention relates to the technical field of medicines, in particular to application of stephanine and salt thereof serving as an iron death inducer in preparation of antitumor drugs.
Background
Apoptosis is the most deeply studied type of cell death, however, in recent years, in addition to apoptosis, many other forms of cell death have been discovered, including autophagic cell death, iron death, pyro-death, glutamate apoptosis and exosome-associated cell death. Traditional cancer treatments are primarily directed at apoptosis, however, many cancer cells are chemoresistant and have defects in apoptosis induction, most of the mechanisms controlling apoptotic and non-apoptotic cell death are also independently regulated, and the cells may also experience various stresses leading to apoptotic and non-apoptotic cell death. Most tumors are treated by surgical resection and radiotherapy at an early stage, and are treated in multiple modes in combination with surgery and chemotherapy at a later stage. However, some chemotherapy-resistant tumor cells can escape apoptotic and other death signals through drug efflux, metabolic changes, alterations in DNA damage repair mechanisms, or other unknown mechanisms, and in addition cancer cells also exhibit higher ROS levels than normal cells and develop resistance to pro-inflammatory and pro-apoptotic molecules to maintain self-survival. Iron death (ferrotosis), a novel cell death mode which is independent from apoptosis and other modes in a mechanism, and a novel iron-dependent cell death mode which is found by high-throughput screening of lethal effects of more than 2 ten thousand compounds by DolmaS and the like in 2003 and is used for targeted killing of HRas overexpressed tumor cells induced by Erastin, wherein the novel iron-dependent cell death mode is greatly different from classical cell death modes such as apoptosis, necrosis, autophagy and the like in aspects of morphology, biochemistry and the like. In 2012, Dixon SJ named this cell death as iron death, when cells died iron, mitochondria condensed in cytoplasm, inner membrane ridges decreased or even disappeared, outer membrane density increased, ruptured, intracellular active divalent iron ions increased, and finally lethal lipid peroxide accumulated to cause cell death. Researches show that the iron death inducer Erastin has better cancer inhibition effect in cancers such as liver cancer, breast cancer, ovarian cancer and the like, and more recent researches show that iron death is not only related to tumors, but also plays an important role in diseases such as neurodegenerative diseases, cardiovascular diseases, diabetes, inflammation and the like.
However, few drugs have been found to induce iron death, such as Erastin and RSL-3, an inhibitor of glutathione peroxidase 4. Although there have been more and more recent studies reporting that some natural products, molecules can induce iron death in different cells, such as: artemisinin, salvia miltiorrhiza, arsenic and the like, but the clinical application of the substances through an iron death mechanism is not proved, and more iron death inducers are still to be developed. There is increasing evidence that compounds from natural sources can induce non-apoptotic programmed cell death in cancer cells, and natural compounds have gained wide promise for future anti-cancer therapies.
Cepharanthine (CEP) is a monomeric compound extracted from Cepharanthine belonging to genus Cepharanthaceae, and belongs to the class of alkaloids, and can be used for clinically treating leukopenia, alopecia areata, etc. Cepharanthine Hydrochloride (CH), a semisynthetic compound obtained by salinization of cepharanthine extracted from cepharanthus spicatus of cepharanthus, has a structural formula:
Figure BDA0002437457180000021
at present, the effect of stephanine as an iron death inducer on tumor-related aspects is not reported at home and abroad.
Disclosure of Invention
Aiming at the defects in the prior art, the invention aims to provide the application of stephanine and salt thereof as an iron death inducer in preparing antitumor drugs, and experiments show that: the cepharanthine and salts thereof can induce the cell to generate iron death in the tumor cell to play a role in treating the tumor, and provide a new technical means for treating tumor patients.
Further, the drug is administered by injection.
Further, the tumor is a prostate tumor.
Further, the concentration of stephanine and the salt thereof for inducing the death of the prostate tumor by iron is 1-100 mu M.
Experiments show that the stephanine hydrochloride induces the cells to die by iron in the tumor cells so as to play a role in resisting cancer, and the influence of the single use of the stephanine hydrochloride on the growth of different prostate cancer cells is detected for the first time. The inventor finds that the cells die but not die after the cepharanthine hydrochloride is added, detects the change of the morphology of the prostate cancer cells through an electron microscope, and finds that the mitochondrial structure is changed after the cepharanthine treatment.
The invention selects 1-100 mu M cepharanthine hydrochloride, and determines the drug toxicity of cepharanthine hydrochloride to prostate cancer DU145, LNcap, PC-3 and PC-3M cell strains in the range. After the cepharanthine hydrochloride is added to treat the cells, the active oxygen level in four cell strains is improved, the lipid peroxide accumulation and the expression level of GPX4 (glutathione peroxidase 4) are reduced, and the anti-cancer effect is obvious.
The prostate cancer LNcap cells have the highest sensitivity to the cepharanthine hydrochloride, and the concentration of the cepharanthine hydrochloride is 20 mu M, so that the LNcap cells can be induced to generate iron death.
The invention also provides a tumor cell iron death inducer, which comprises cepharanthine hydrochloride. The cepharanthine hydrochloride has therapeutic potential for neurodegenerative diseases, cardiovascular diseases, etc.
In summary, the invention includes at least one of the following beneficial technical effects:
the stephanine and the salt thereof can induce the cell to generate iron death in the tumor cell and induce the death of apoptosis-tolerant tumor cells, have obvious anti-cancer and anti-drug resistance effects and provide more effective treatment schemes for a large number of tumor patients. Meanwhile, the cepharanthine is used as a clinical medicine, has good safety, wide sources of the cepharanthine hydrochloride, simple and economic acquisition mode and good application prospect in the technical field of anti-tumor.
Drawings
FIG. 1 is a graph of the survival rate of various strains of prostate cancer cells inhibited by stephanine hydrochloride at various concentrations in example 1;
FIG. 2 is a graph of apoptosis of prostate cancer cells following cepharanthine hydrochloride treatment in example 2;
FIG. 3 is a graph showing the change in reactive oxygen species in prostate cancer cells after cepharanthine hydrochloride treatment in example 3;
FIG. 4 is a graph showing the change in intracellular lipid peroxide levels of prostate cancer cells in example 3 after treatment with stephanine hydrochloride and the iron death inhibitor, Fer-1;
FIG. 5 is a graph of the effect of LNcap cells on prostate cancer cell viability following the combination of stephaglabrin hydrochloride with the iron chelator DFO or with the iron death inhibitor Fer-1 of example 3.
Detailed Description
The present invention will be described in further detail with reference to the accompanying drawings and examples.
Example 1 Effect of varying concentrations of stephanine hydrochloride on prostate cancer cell survival
S1, weighing cepharanthine hydrochloride powder by using an electronic balance, placing the cepharanthine hydrochloride powder into a centrifugal tube, adding dimethyl sulfoxide (DMSO) to dissolve the cepharanthine hydrochloride powder to prepare a mother solution with the concentration of 100mM, then diluting the mother solution with DMSO to different concentrations, wherein the concentrations of the cepharanthine hydrochloride are 1 mu M, 20 mu M, 40 mu M, 80 mu M and 100 mu M respectively, adding the cepharanthine hydrochloride powder into a DMEM/F12 culture medium containing 10% fetal calf serum respectively, and performing an experiment for later use;
s2, selecting four strains of prostate cancer cells DU145 (purchased from an ATCC cell bank), LNcap (purchased from an ATCC cell bank), PC-3 (purchased from an ATCC cell bank) and PC-3M (purchased from a national experiment cell resource sharing platform), digesting the prostate cancer cells by using a digestive juice containing 0.075 wt.% of I-type collagenase, 0.5 wt.% of II-type collagenase and 0.1 wt.% of trypsin, digesting at 37 ℃ for 1min, repeatedly blowing the digested suspension by using a straw after digestion, plating the suspension on a corresponding culture dish for subculture or drug experiments, and observing the cell morphology and growth condition under an inverted microscope every day;
s3, inoculating the suspension prepared in the step S2 into a 96-well plate, wherein each well is inoculated with 0.8 multiplied by 10 cells4Culturing prostate cancer cells by using 5 groups of culture media containing drugs with different concentrations prepared in the step S1, setting 20 groups of experimental groups, adding 4 groups of DMSO groups with the same dosage into DMEM/F12 culture media containing 10% fetal calf serum to culture prostate cancer cells as a control group, after overnight culture and wall attachment, respectively adding 10 mu l of 5mg/ml tetramethyl azozolium (MTT) during 24h and 48h of culture, sucking the DMEM/F12 culture media and MTT after 4h of incubation at 37 ℃, adding DMSO, reacting for 30min at room temperature on a shaking table, then measuring an absorbance value at a wavelength of 570nm by using a chemiluminescence instrument, and finally drawing a fitting curve of drug concentration corresponding to cell survival rate by using GraphPad Prism 8.
The invention adopts an MTT method to detect the toxicity of the stephanine hydrochloride to prostate cancer cells DU145, LNcap, PC-3 and PC-3M four strains of cells for 24h and 48h, and calculates to obtain the IC50 value of the stephanine hydrochloride to different prostate cancer cells. The results show that LNcap cells are most sensitive to cepharanthine hydrochloride, see figure 1.
In this example, concentrations of cepharanthine hydrochloride of 1. mu.M, 40. mu.M, 80. mu.M and 100. mu.M, the effect of cepharanthine hydrochloride on prostate cancer cell survival was similar and the fitted curves were similar.
Example 2 testing the Effect of stephanine hydrochloride on the cell death pathway of prostate cancer
S1, adopting the mother liquor with the concentration of 100mM prepared in the embodiment 1, then diluting the mother liquor with a culture medium into different concentrations, wherein the final concentrations of stephanine hydrochloride are respectively 10 mu M and 20 mu M, respectively culturing cells into two groups, setting a group of DMSO as a blank control group (represented by control), wherein the dose of the added DMSO is the same as that of the stephanine hydrochloride, and performing an experiment for later use;
s2, inoculating the suspension of prostate cancer cells LNcap prepared in step S2 of example 1 into a 6-well plate, wherein each well is inoculated with 30X 10 cells4And (2) culturing the prostate cancer cells by using 3 groups of culture media prepared in step S1 of the embodiment 2, collecting the prostate cancer cells in a centrifuge tube when the cells are cultured for 24 hours and 48 hours, centrifuging the cells for 20 minutes at a speed of 1000rpm/min, removing supernatant after centrifugation, re-suspending the cells by using DMEM/F12 culture medium containing 10% fetal calf serum, discarding the culture medium by centrifugation, repeatedly washing the cells for 2 times by using phosphate buffer, then adding binding buffer containing 5 mul of annexin V dye for re-suspending, then adding 10 mul of PI dye, incubating the cells for 5 minutes at room temperature, detecting the apoptosis by using a flow cytometer, and respectively recording the apoptosis of the prostate cancer cells at 24 hours and 48 hours, and referring to FIG. 2.
As can be seen from fig. 2, when the concentration of cepharanthine hydrochloride was 20 μ M, significant cell death was induced in LNcap prostate cancer cells, but the results showed that the proportion of apoptotic cells was not high, suggesting that cepharanthine hydrochloride induces other forms of cell death.
Example 3 determination of the Effect of stephanine hydrochloride on reactive oxygen species in LNcap of prostate cancer cells
S1, miningThe method comprises diluting the suspension of the prostate cancer cells prepared in example 2 to obtain the concentrations of stephanine hydrochloride of 10 μ M and 20 μ M, adding the stephanine hydrochloride into DMEM/F12 culture medium containing 10% fetal bovine serum, dividing into two groups, setting a group of DMSO as a blank control group (indicated by control), adding the DMSO in the same amount as the stephanine hydrochloride, inoculating the suspension of the prostate cancer cells LNcap prepared in example 2 into 6-well plates, inoculating 30 × 10 cells per well4Culturing prostate cancer cells by using 3 groups of culture media prepared in the embodiment, and performing experiments for standby;
s2, diluting the active oxygen detection probe DCFH-DA by using a serum-free culture medium according to the ratio of 1:1000 so that the final concentration is 10 mu M. Removing the culture medium of prostate cancer cells LNcap cultured in a 6-well plate, and adding 2ml of diluted DCFH-DA solution into each well;
s3, putting the 6-hole plate into a cell culture box at 37 ℃ for incubation for 20min, and then washing the cells three times by using a serum-free culture medium to sufficiently remove the DCFH-DA probe which does not enter the cells. Collecting prostate cancer cells LNcap by pancreatin, centrifuging for 25min at the speed of 800rpm/min, re-suspending and cleaning by PBS after centrifuging, centrifuging again for 15min at the speed of 500rpm/min, discarding the supernatant, re-suspending the cells by 200 μ l PBS, and then detecting the fluorescence intensity of DCFH-DA in the cells by a flow analyzer under the conditions of 488nm excitation wavelength and 525nm emission wavelength, referring to FIG. 3.
As can be seen from fig. 3, the concentration of stephanine hydrochloride of 20 μ M has a greater effect on the active oxygen in the prostate cancer cells LNcap, the active oxygen in the prostate cancer cells LNcap is increased more, and the concentration of stephanine hydrochloride of 10 μ M has a slightly lower effect on the active oxygen in the prostate cancer cells LNcap.
Example 4 determination of the Effect of stephanine hydrochloride on the level of lipid peroxides in LNcap of prostate cancer cells
S1, taking the cepharanthine hydrochloride with the concentration of 20 mu M obtained by dilution in the example 3 as one group and taking the solution of the cepharanthine hydrochloride combined inhibitor Fer-1 with the concentration of 20 mu M as one group, in the example, the two groups are respectively added into DMEM/F12 culture medium containing 10% fetal bovine serum, and a group of DMSO (expressed by control) is further set as a blank control group to be addedThe dose of the physiological saline is the same as that of the stephanine hydrochloride, and the suspension of prostate cancer cells LNcap prepared in example 3 is inoculated into a 6-well plate, and each well is inoculated with 30X 10 cells4Culturing the prostate cancer cells in LNcap by using 3 groups of culture media prepared in the step of the embodiment, wherein the experiments are ready for use;
s2, detecting the level of lipid peroxide by using BODIPY C11 as a probe, adding 5 mu M BODIPYC11 into each well of a 6-well plate, incubating in a cell culture box at 37 ℃ for 30min, collecting prostate cancer cells LNcap into a centrifuge tube by trypsinization, centrifuging for 3min at the speed of 1200rpm/min, re-suspending and cleaning with PBS after centrifugation, centrifuging again for 5min at the speed of 700rpm/min, discarding the supernatant, re-suspending the cells with 200 mu l PBS, and further analyzing the fluorescence intensity of the cells under the excitation wavelength of 488nm by using a flow cytometer, wherein the reference is shown in FIG. 4.
As can be seen from fig. 4, stephanine hydrochloride with a concentration of 20 μ M has a greater effect on the LNcap lipid peroxide level of prostate cancer cells than the stephanine hydrochloride combined inhibitor Fer-1, and the lipid peroxide accumulation is higher than that of the LNcap lipid peroxide level of prostate cancer cells in the blank control group compared with that in the other two groups. This result suggests that cepharanthine hydrochloride induces iron death.
Example 5 determination of the Effect of stephanine hydrochloride on inhibiting the viability of prostate cancer LNcap cells by iron death
S1, taking the cepharanthine hydrochloride with the concentration of 20 mu M obtained by dilution in the example 3 as a group, taking the solution of the cepharanthine hydrochloride with the inhibitor Fer-1 with the concentration of 20 mu M as a group, taking the solution of the cepharanthine hydrochloride with the inhibitor iron chelator DFO with the concentration of 20 mu M as a group, respectively adding the three groups into DMEM/F12 culture medium containing 10% fetal bovine serum, inoculating the prostate cancer cell LNcap suspension prepared in the example 3 into a 6-well plate, and inoculating 30 multiplied by 10 cells into each well4Culturing the prostate cancer cells in LNcap by using 3 groups of culture media prepared in the step of the embodiment, wherein the experiments are ready for use;
s2, after overnight culture and adherence, respectively adding 10 mu l of 5mg/ml tetramethyl azodicarbonamide (MTT) during 24h and 48h of culture, after incubation for 4h at 37 ℃, absorbing DMEM/F12 culture medium and MTT, adding DMSO, and reacting for 30min at room temperature on a shaking table;
s3, detecting the level of lipid peroxide by using BODIPY C11 as a probe, adding 5 mu M BODIPYC11 into each well of a 6-well plate, incubating in a cell culture box at 37 ℃ for 30min, collecting prostate cancer cells LNcap into a centrifuge tube by trypsinization, centrifuging for 3min at the speed of 1200rpm/min, re-suspending and cleaning with PBS after centrifugation, centrifuging again for 5min at the speed of 700rpm/min, discarding the supernatant, re-suspending the cells with 200 mu l PBS, and further analyzing the fluorescence intensity of the cells under the excitation wavelength of 488nm by using a flow cytometer, wherein the reference is made to FIG. 5.
As can be seen from fig. 5, the iron death inhibitor Fer-1 or the iron chelator DFO can reverse the inhibition of stephaglabrin hydrochloride on the viability of prostate cancer LNcap cells, and this result demonstrates that stephaglabrin hydrochloride is an iron death inducer.
Example 6 tumor formation test
Selecting male atmoic BALB/c nude mouse (4-6 weeks old), purchasing from Chinese laboratory animal science institute, placing in sterile environment, subcutaneously injecting prostate cancer cell LNcap suspended in 200 μ l matrix gel/PBS (5mg/ml) onto mouse body to construct nude mouse tumor model, and when the tumor volume in mouse body grows to 100mm3Randomly dividing the mice into 4 groups, namely a model group, a stephanine hydrochloride low-dose group of 15mg/kg, a stephanine hydrochloride medium-dose group of 30mg/kg and a stephanine hydrochloride high-dose group of 40mg/kg, measuring the weights of the mice and the sizes of tumors every other day by adopting a mode of intraperitoneal injection every day, killing the mice on day 13 to collect the tumors, wherein the weight of the tumors of each group of mice after the test is shown in a table 1, and the tumor growth rate of each group of mice after the test is shown in a table 2.
TABLE 1
Figure BDA0002437457180000071
TABLE 2
Figure BDA0002437457180000072
According to the data in tables 1 and 2, the tumor weight of the stephanine hydrochloride mice is far lower than that of the model mice, which indicates that the stephanine hydrochloride can inhibit tumor growth, and especially the low-dose stephanine hydrochloride group has the best tumor inhibition effect.
The embodiments of the present invention are preferred embodiments of the present invention, and the scope of the present invention is not limited by these embodiments, so: all equivalent changes made according to the structure, shape and principle of the invention are covered by the protection scope of the invention.

Claims (5)

1. Application of cepharanthine and its salt as iron death inducer in preparing antitumor drugs is provided.
2. Use according to claim 1, characterized in that: the medicament is administered by injection.
3. Use according to claim 1, characterized in that: the tumor is prostate tumor.
4. Use according to claim 3, characterized in that: the concentration of the cepharanthine and salts thereof for inducing the death of the prostate tumor by iron is 1-100 mu M.
5. An agent for inducing iron death in tumor cells, comprising: the iron death inducer comprises cepharanthine hydrochloride.
CN202010256288.4A 2020-04-02 2020-04-02 Application of cepharanthine and its salt as iron death inducer in preparing antitumor drugs Pending CN111228269A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010256288.4A CN111228269A (en) 2020-04-02 2020-04-02 Application of cepharanthine and its salt as iron death inducer in preparing antitumor drugs

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010256288.4A CN111228269A (en) 2020-04-02 2020-04-02 Application of cepharanthine and its salt as iron death inducer in preparing antitumor drugs

Publications (1)

Publication Number Publication Date
CN111228269A true CN111228269A (en) 2020-06-05

Family

ID=70869956

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010256288.4A Pending CN111228269A (en) 2020-04-02 2020-04-02 Application of cepharanthine and its salt as iron death inducer in preparing antitumor drugs

Country Status (1)

Country Link
CN (1) CN111228269A (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111838074A (en) * 2020-07-15 2020-10-30 浙江大学 Method for constructing iron death mouse model by using iron death inducer Erastin and application of method
CN113244252A (en) * 2021-06-09 2021-08-13 广西农业职业技术学院 Application of saponin compound in preparation of iron death inducer
CN113337569A (en) * 2021-05-26 2021-09-03 深圳市人民医院 Method for screening antitumor drugs based on induction of cell inflammatory death
CN113552353A (en) * 2021-07-12 2021-10-26 江南大学 Magnetic particle chemiluminescence kit for diagnosis of PCa and CRPC diseases

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101507725A (en) * 2008-12-31 2009-08-19 河南省医药科学研究院 Use of stephanine and hydrochloride thereof in preparing anti-hepatitis virus medicine
US20150190392A1 (en) * 2014-01-03 2015-07-09 Macau University Of Science And Technology Group of Alkaloids, the Novel Autophagic Enhancers for Treatment of Cancers and Neurodegenerative Conditions Thereof
CN104922677A (en) * 2015-05-27 2015-09-23 广州暨南生物医药研究开发基地有限公司 Application of drug combination containing cepharanthine hydrochloride in preparing drug for treating leukopenia
CN110496225A (en) * 2019-09-28 2019-11-26 广州暨南生物医药研究开发基地有限公司 Stephanine and autophagy inhibitor are combined the application in preparation treatment liver-cancer medicine

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101507725A (en) * 2008-12-31 2009-08-19 河南省医药科学研究院 Use of stephanine and hydrochloride thereof in preparing anti-hepatitis virus medicine
US20150190392A1 (en) * 2014-01-03 2015-07-09 Macau University Of Science And Technology Group of Alkaloids, the Novel Autophagic Enhancers for Treatment of Cancers and Neurodegenerative Conditions Thereof
CN104922677A (en) * 2015-05-27 2015-09-23 广州暨南生物医药研究开发基地有限公司 Application of drug combination containing cepharanthine hydrochloride in preparing drug for treating leukopenia
CN110496225A (en) * 2019-09-28 2019-11-26 广州暨南生物医药研究开发基地有限公司 Stephanine and autophagy inhibitor are combined the application in preparation treatment liver-cancer medicine

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ITA等: "Remarkable enhancement of cytotoxicity of onconase and cepharanthine when used in combination on various tumor cell lines", 《CANCER BIOLOGY & THERAPY》 *
潘柏申等: "《2018检验医学进展》", 31 August 2018, 中华医学电子音像出版社 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111838074A (en) * 2020-07-15 2020-10-30 浙江大学 Method for constructing iron death mouse model by using iron death inducer Erastin and application of method
CN113337569A (en) * 2021-05-26 2021-09-03 深圳市人民医院 Method for screening antitumor drugs based on induction of cell inflammatory death
CN113244252A (en) * 2021-06-09 2021-08-13 广西农业职业技术学院 Application of saponin compound in preparation of iron death inducer
CN113552353A (en) * 2021-07-12 2021-10-26 江南大学 Magnetic particle chemiluminescence kit for diagnosis of PCa and CRPC diseases
CN113552353B (en) * 2021-07-12 2023-08-25 江南大学 Magnetic particle chemiluminescence kit for PCa and CRPC disease diagnosis

Similar Documents

Publication Publication Date Title
CN111228269A (en) Application of cepharanthine and its salt as iron death inducer in preparing antitumor drugs
Sun et al. Discovery and development of tumor glycolysis rate-limiting enzyme inhibitors
CN109320570A (en) A kind of icariside I class compound, derivative, officinal salt and application
CN111658655A (en) Application of cucurbitacin B in preparation of iron death inducer and anti-nasopharyngeal carcinoma drug
CN113181166B (en) Application of curcumenol in preparing anti-lung cancer medicine
CN111166754A (en) Application of cryptotanshinone in preparation of medicine for preventing and treating cachexia skeletal muscle atrophy
CN104817608A (en) Cordycepin salt containing selenium compound, preparation method of cordycepin salt and application
CN112870189B (en) Application of 2-acyl-1-dimethylaminomethyl ferrocene derivative in preparation of drug for targeted therapy of hepatocellular carcinoma
CN112402413B (en) Application of lindley eupatorium herb sesquiterpene lactone B in preparation of anti-liver cancer medicine and anti-liver cancer medicine
CN111393278A (en) Usnea longissima derivative and application thereof in preparation of medicine for treating gallbladder cancer
CN111789830A (en) Application of 2- (2,2, 2-trifluoroethylene) -1, 3-dione compound in preparation of anti-lung cancer drugs
CN112336716A (en) Application of vitamin C and disulfiram in preparation of anti-tumor combined medicine
CN112791081A (en) Application of white ketone in preparation of lung cancer treatment medicine
CN111000850A (en) Application of cryptotanshinone in preparation of drug for overcoming drug resistance of EGFR-TKI used for treating NSCLC
CN114366740B (en) Application of compound A-6 in preparation of broad-spectrum anticancer drugs
Cao et al. Study On Anticancer Effect of Synthetic Biogenic Source of Germacrene A
CN105969725B (en) The purposes of fructus lycii red pigment
CN115645532B (en) Application of isoxazole derivative in preparing brain glioma radiotherapy sensitization drugs
CN104127394A (en) Application of above one of alpha-pinene, beta-pinene, 1,8-eudesmol and camphene in preparation of human blood vessel endothelial cell damage treatment medicines
CN113908148B (en) Application of nobiletin in preparation of anti-cholangiocarcinoma drugs
CN113876774B (en) Application of neoliensinine in preparing medicine for treating leukemia
CN104337807B (en) Nanometer liposome of load paclitaxel and sensitizer and preparation method thereof and application altogether
CN112274514B (en) Application of sinomenine compounds in preparation of medicines for preventing or treating glioma
CN108379282A (en) A kind of drug and application thereof
CN116173008B (en) Application of nardostachyne as iron death inducer

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
RJ01 Rejection of invention patent application after publication
RJ01 Rejection of invention patent application after publication

Application publication date: 20200605