WO2011088605A1 - Use of leptin in enhancing therapeutic efficacy of medroxyprogesterone acetate on liver cancer - Google Patents

Use of leptin in enhancing therapeutic efficacy of medroxyprogesterone acetate on liver cancer Download PDF

Info

Publication number
WO2011088605A1
WO2011088605A1 PCT/CN2010/001662 CN2010001662W WO2011088605A1 WO 2011088605 A1 WO2011088605 A1 WO 2011088605A1 CN 2010001662 W CN2010001662 W CN 2010001662W WO 2011088605 A1 WO2011088605 A1 WO 2011088605A1
Authority
WO
WIPO (PCT)
Prior art keywords
liver cancer
mpa
leptin
cells
estrogen
Prior art date
Application number
PCT/CN2010/001662
Other languages
French (fr)
Chinese (zh)
Inventor
王森稔
叶耀宗
李金德
杨翔荏
杨晓芳
Original Assignee
高雄医学大学
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
Priority claimed from US12/689,718 external-priority patent/US20110098219A1/en
Application filed by 高雄医学大学 filed Critical 高雄医学大学
Publication of WO2011088605A1 publication Critical patent/WO2011088605A1/en

Links

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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/405Indole-alkanecarboxylic acids; Derivatives thereof, e.g. tryptophan, indomethacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/2264Obesity-gene products, e.g. leptin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis

Definitions

  • the invention relates to a method for treating liver cancer by using a thin hormone, in particular to a therapeutic drug, a course of treatment or a screening platform for administering a therapeutically effective amount to a liver cancer patient, in particular to increase or decrease the leptin (Leptin).
  • a thin hormone in particular to a therapeutic drug, a course of treatment or a screening platform for administering a therapeutically effective amount to a liver cancer patient, in particular to increase or decrease the leptin (Leptin).
  • the physiological concentration of leptin in patients with liver cancer is combined with medroxyprogesterone acetate (MPA) and then contacted with liver cancer cells to kill liver cancer cells.
  • MPA medroxyprogesterone acetate
  • liver cancer is a common malignant tumor in the eastern countries, and it is found by clinical observation that it contains 5 ⁇ ⁇ 3 ⁇ 4)% of liver cancer is associated with hepatitis and cirrhosis.
  • the statistics of the top ten causes of death in Taiwan, China, show that since 1982, cancer has leapt to the top, with liver cancer being the first cause of all cancer deaths for men and for women. It is also the second highest, and it kills 4,000 people every year, which is enough to show that it poses a great threat to the health of Chinese people. Therefore, early diagnosis and treatment of liver cancer will be one of the research priorities in the future.
  • VEGF Vascular Endothelial Growth Factor
  • liver cancer has gradually tended to treat liver cancer patients with hormone therapy, and clinical and laboratory studies have also found that liver cancer is also found.
  • Glucocorticoid Antagonists for example: Progesterone and RU486 can inhibit the expression of a-fetoprotein in human liver cancer. It suggests that progesterone has a potential for hormone therapy in liver cancer.
  • progesterone acetate, a progesterone analog can effectively inhibit the growth of liver cancer cells.
  • the MPA is derived from the luteinizing hormone (17-OH-progesterone) and is administered orally as a synthetic steroid. Its structure is similar to that of the natural luteinizing hormone, except that it has a methyl group at the ⁇ - 6 position (Methyl Group). There is an acetate functional group (Acetoxy Group) at position 17.
  • the MPA is a luteinizing hormone with antiestrogenic activity. In postmenopausal breast cancer, it completely inhibits the concentration of plasma estrogen and also inhibits the release of Luteining Hormone (LH). Stimulates the growth of the endometrium and produces typical body hormone changes in breast acinar cells.
  • the MPA action machine is still unclear, but is believed to involve interaction with intracellular hormone receptors.
  • MPA binds to receptors of estrogen, progesterone and androgen, and in particular, binding to androgen receptors is particularly important for the cytotoxic effects of this drug.
  • MPA can inhibit the size of liver cancer tumors and prolong the survival rate of liver cancer patients.
  • MPA may affect the process of liver cancer evolution and improve the prognosis of patients.
  • the use of MPA to treat clinical liver cancer patients may be helpful for patient survival, but because MPA has multiple pharmacological biological activities, its mechanism of action needs to be further explored.
  • estrogen is a hormone of a single-chain protein component and is generally considered to play an important role in weight maintenance and energy regulation
  • MPA is a progesterone-derived synthetic steroid.
  • MPA can cause apoptosis of liver cancer cell lines in a system in vivo and in vitro, that is, the MPA system can effectively inhibit the growth of liver cancer cell lines in a cell line system; however, relevant research abroad
  • MPA was found that treatment of liver cancer patients with MPA had no significant effect on prolonging survival.
  • MPA is currently on the market for cancer, there is no exhilarating effect on liver cancer patients.
  • the main object of the present invention is to overcome the above problems encountered in the prior art and to provide an application of attenuating MPA with a leptin to inhibit liver cancer cells.
  • a secondary object of the present invention is to provide a method for directly treating MPC patients with liver cancer patients in the serum of liver cancer patients or in liver cancer tissues, so as to achieve the pharmacological action of enhancing MPA.
  • Another object of the present invention is to provide a method for treating liver cancer patients by combining leptin and MPA into a drug when the expression of lept hormone in the serum of liver cancer patients or liver cancer tissues is high, so as to achieve the pharmacological action of strengthening MPA. .
  • a further object of the present invention is to provide a method for increasing leptin by 5-hydroxy-trypton (5-HTP) when the expression of leptin in serum or liver cancer tissues of liver cancer patients is low. After the amount, MPA treatment is given to achieve the pharmacological action of strengthening MPA.
  • 5-HTP 5-hydroxy-trypton
  • the present invention relates to a method for treating liver cancer by using a lepid hormone, comprising a therapeutic drug, a course of treatment or a screening platform for administering a therapeutically effective amount to a liver cancer patient, characterized in that the hormone is increased or the liver cancer patient is raised.
  • the physiological concentration of lean hormone combined with a MPA is contacted with liver cancer cells, and the interaction of the estrogen and its progesterone receptor through the receptor enhances and accelerates the MPA to kill liver cancer cells, thereby normalizing the liver epithelial cell line and cancer cells.
  • the strain has a specific pharmacological effect, so that it can have no adverse side effects on normal liver cells, and can prolong the overall survival rate of liver cancer patients.
  • the level of expression of the estrogen can further serve as a predictor of prophylactic factors for the treatment of MPA in patients with liver cancer and a prognostic factor for survival.
  • Figure 1 is a schematic diagram showing the analysis of the prognosis survival time of patients with liver cancer using MPA after surgery.
  • Figure 2 is a schematic diagram showing the analysis of cell growth by estrogen at different concentrations in the present invention.
  • Figure 3 is a schematic diagram showing the analysis of cell growth by MPA at various concentrations in the present invention.
  • Figure 4 is a graphical representation of the analysis of cell growth after treatment with leptin and MPA in the present invention.
  • Fig. 5 is a schematic view showing the cell type of the present invention after treating cells with leptin and MPA.
  • Fig. 6 A is a schematic diagram showing the effect of the present invention on cell cycle after treating cells with leptin and MPA.
  • Fig. 6B is a schematic diagram showing the analysis of cell cycle after treating cells with estrogen and MPA.
  • Figure 7 Schematic diagram of the effect of the present invention on apoptosis-related proteins after treatment with leptin and MPA.
  • Fig. 8 A is a schematic diagram showing the interaction of the leptin receptor with the progesterone receptor receptor by immunoprecipitation.
  • Figure 8B is a schematic representation of the interaction of the leptin receptor with the progesterone receptor receptor by immunofluorescence staining.
  • Figure 9 Schematic diagram of the analysis of the present invention for inhibiting the reduction of the estrogen receptor and enhancing the action of MPA.
  • Figure 10 A is a schematic representation of the increase in expression of the long form of the estrogen receptor in the present invention to allow apoptosis of the cells after dosing.
  • Figure 10B is a schematic diagram showing the analysis of apoptosis of cells after dosing by increasing the amount of progesterone receptor expression.
  • Fig. 1 is a schematic diagram showing changes in the amount of JAK/STAT pathway after drug treatment in the present invention.
  • Fig. 1 2 is a schematic diagram showing changes in the amount of expression of the MAPK pathway of the present invention after drug treatment.
  • Fig. 1 3 A is a schematic diagram showing changes in the amount of expression of the JAK/STAT path of the present invention in a short period of time after drug treatment.
  • Fig. 1 3 B is a schematic diagram showing changes in the amount of expression of the MAPK pathway of the present invention in a short time by drug treatment.
  • Fig. 14 is a schematic diagram showing the degradation of p-STAT3 by inhibiting the activation of ERK1/2 and the expression of PIAS3 in the present invention.
  • Fig. 15 is a schematic diagram showing the activity analysis of a large amount of leptin in the cells to enhance MPA inhibition of cell growth in the cells.
  • Fig. 1 6 A is a schematic diagram of the analysis of the STAT3 related message pathway by MPA treatment in the present invention.
  • Fig. 1 6 B is a schematic diagram of the analysis of the MAPK-related message pathway by MPA treatment in the present invention.
  • Fig. 17 is a schematic diagram showing the analysis of the growth of normal liver epithelial cell lines by the combination of leptin and MPA in the present invention.
  • Fig. 1 8 A is a schematic diagram showing the analysis of STAT3 related message pathways after treatment of lean liver cells with MPA by normal liver cells of the present invention.
  • Fig. 1 8 B is a schematic diagram of the MAPK-related message pathway of normal liver cells of the present invention treated with estrogen and MPA.
  • the invention relates to a method for treating liver cancer by using a lepid hormone, and can be used as statistical data on liver cancer cell lines (HepG2 Cells) and clinical liver cancer patients.
  • the invention comprises a therapeutic drug, a course of treatment or a screening platform for administering a medicinal effective amount to a liver cancer patient, which is characterized by a combination of a leptin (Leptin) or a physiological concentrating agent for increasing the physiological concentration of leptin in a liver cancer patient.
  • a leptin leptin
  • a physiological concentrating agent for increasing the physiological concentration of leptin in a liver cancer patient.
  • MPA melanocytogenes
  • the interaction of the estrogen and its progesterone receptor through the receptor enhances and accelerates the MPA inhibiting the killing effect of the liver cancer cells, thereby normalizing the liver epithelial cell line.
  • the drug or course of treatment with leptin combined with MPA can cause the cancer cells of liver cancer patients to decrease in size, that is, the drug can respectively reduce the size or quality of liver cancer cells, and is normal.
  • the adverse side effects of non-toxic killing of liver cells can effectively prolong the overall survival rate of liver cancer patients.
  • the invention has found that the effects of lean hormone and MPA on liver cancer cells and prolonging the survival rate of patients have been found in the study of in vitro and in some clinical patients. Accordingly, the combination of leptin and MAP was designed as a therapeutic drug for the treatment of liver cancer patients, and it was also found that the physiological concentration of lean hormone in normal humans can enhance the efficacy of MPA.
  • MPA is treated at a dose of 500 mg (mg) per day in a clinical patient in a preferred embodiment of the present invention, and therefore, after the present invention utilizes a combination of MPA to increase the physiological concentration of estrogen or estrogen , can achieve better therapeutic effects.
  • FIG. 1 for the analysis of the prognosis survival time of the liver cancer patients after treatment with MPA.
  • the present invention compares the results of clinical analysis of clinical liver cancer patients in the above preferred embodiments, wherein the performance of leukemia in liver cancer tissues and the survival time of MPA and patients after surgery can be found. There is no meaningful relationship.
  • the P values shown in the upper left and upper right of Figure 1 are 0. 383 and 0. 171; respectively, there is MPA after surgery.
  • the analysis showed that the high-performance of the estrogen was longer than the low-performance one, as shown in the lower left of Figure 1, the P value was 0.001.
  • the prognosis and survival rate of patients with liver cancer who have high expression of estrogen after surgery are significantly better than those with low expression of estrogen. Therefore, it is sufficient to confirm that the leptin can enhance the effect of MPA on the treatment of liver cancer, and the expression level of the estrogen can further serve as a predictor of the efficacy of the treatment of liver cancer patients with MPA (Predictive Factors) and its survival time.
  • Prognostic Factors in which the higher the expression of the estrogen, the more obvious the inhibitory effect of the intensive sputum on hepatocellular carcinoma cells, which can be used as a liver cancer patient to receive hormone therapy (ie, sputum) after surgery. Indicators, in turn, can help maintain or prolong the survival of liver cancer patients.
  • the prognosis of patients with liver cancer who have higher expression of estrogen is better than that of patients with low expression of estrogen, and the survival rate can be extended for nearly five years. This is among other related technologies. Undiscovered.
  • the present invention also demonstrates in an in vitro experiment that leptin enhances the action of sputum to inhibit liver cancer cells through certain mechanisms, and in addition to combining leptin and MAP into a therapeutic drug or treatment, the present invention may also provide A platform for screening for lean hormones to determine whether to perform MPA treatment.
  • the combination of leptin and MPA is a new treatment that can be used in patients with liver cancer. There are no reports or studies at home and abroad. The difference from other known literatures is that the present invention combines MPA with estrogen as a new treatment method, so that it can effectively improve the shortcomings of the known technique that only MPA is used to treat liver cancer patients without prolonging the survival rate.
  • the present invention further explores the anti-cancer role of leptin combined with MPA in liver cancer cells and the molecular mechanism of its action.
  • Hepatocellular carcinoma cell lines were treated with leptin and MPA, and their cell viability was observed.
  • the cell cycle, apoptosis and mechanism of action were analyzed.
  • the related message transmission pathways involved in estrogen and MPA were also explored.
  • the effects of the pharmacological effects of leptin combined with MPA on normal liver epithelial cell lines were examined by observing their drug specificity, in order to further understand the use of hormone therapy in liver cancer.
  • Example 1 Effect of leptin on the growth of hepatoma cells
  • FIG. 2 is a schematic diagram of the analysis of cell growth by different hormones in the present invention.
  • LH lean hormone
  • LL lean hormone
  • the above-mentioned determination of the main concentration depends on the range of physiological concentration of 10 ng as a normal person, and the effect of the estrogen on the cells can be observed in the physiological range, and the higher concentration determines whether the higher the concentration of the estrogen is for the cells. The impact will be greater.
  • the experimental results showed that after treatment of cells with high or low concentrations of estrogen for 24 hours, it was found in the XTT assay that leptin had no significant effect on the growth of cancer cells.
  • Example 2 Effect of MPA on the growth of hepatoma cells
  • FIG. 3 is a schematic diagram of the analysis of cell growth of MPA at different concentrations in the present invention.
  • two different concentrations of MPA were used to treat the cells, with a high concentration of MPA (MH) of 10 and a low concentration of MPA (ML) of 10 ⁇ M.
  • the experimental results showed that after treatment with high concentration of MPA for 24 hours, the cells were significantly inhibited by XTT assay. After 48 hours of treatment, the cell inhibition phenomenon was significantly increased.
  • the low concentration of MPA did not significantly inhibit the effect at 24 or 48 hours. Therefore, it indicates that the inhibitory effect of MPA on cells with increasing concentration and time is better.
  • FIG. 4 is a schematic diagram of the analysis of cell growth after treatment with lean hormone combined with MPA.
  • cells were treated with two different concentrations of estrogen in combination with two different concentrations of MPA.
  • the results of the experiment showed that the high concentration of the estrogen combined with the high concentration of MPA to treat the cells for 24 hours, the XTT assay showed that the inhibition of cell growth was indeed better than the MPA alone, and its P value was 0.001, and The P value of 0.01% is better than that of the MPA alone.
  • the experimental results show that leptin can indeed enhance the effect of MPA on cell growth, and the higher the concentration of leptin, the more obvious the inhibitory effect.
  • Example 4 Leptin combined with MPA causes increased apoptosis
  • FIG. 5 is a schematic diagram of the cell type of the present invention treated with leukemia combined with MPA.
  • the estrogen does enhance the cytostatic effect of MPA
  • the results of the cell type are observed under the microscope, as in the experiments of the above examples, the combination of leptin After MPA treatment, the degree of apoptosis of Apoptosis was significantly greater than that of MPA alone.
  • the present invention further observes the nuclear form by immunofluorescence. After the cells were treated with estrogen and MPA for 24 hours, the nucleus was stained with fluorescent dye (DAPI) and observed under a fluorescence microscope at 40X. The results were also consistent, and the degree of nuclear atrophy was more obvious than that of MPA alone. Therefore, it can be seen from the above results that leptin does enhance and accelerate the apoptosis caused by MPA.
  • DAPI fluorescent dye
  • FIG. 6A and FIG. 6B are respectively a schematic diagram showing the effect of cell cycle on cell treatment after treatment with leukemia combined with MPA, and the cell cycle analysis of the present invention after treating cells with leptin and MPA.
  • this example is to collect cells treated with estrogen or MPA and estrogen combined with MPA for 24 hours, by flow cytometry (Flow Cytometry) Analyzes the effect of the drug on the Cell Cycle and counts the portion of Sub-Gl/Apoptosis in the cell cycle as Student's - test.
  • flow cytometry Flow Cytometry
  • the experimental results showed that the cells treated with MPA had an increasing trend in sub-G1, indicating a marked increase in apoptotic cells, but decreased in the G1 and G2/M phases, but increased in the S phase;
  • the analysis showed that the increase of sub-G1 was higher than that of MPA alone, and its P value was 0.002; while the low concentration of lean hormone combined with high concentration of MPA Sub-
  • the increase trend of Gl is also higher than that of MPA alone, and its P value is 0.04, which is lower in G1 and G2/M than in MPA alone, and higher in S phase.
  • the ratio of leukemia combined with MPA leads to apoptosis compared with MPA alone.
  • the effect may be to increase MPA inhibition of G1 and G2/M phases, or to arrest the cell cycle in S phase.
  • FIG. 7 the effect of the present invention on apoptosis-related proteins after treatment with leptin and MPA is shown.
  • the present invention uses Western blotting to analyze whether the apoptosis-related proteins are changed after treatment with estrogen and MPA for 24 hours.
  • the cell proteins were anti-c leaved caspase 3, anti-cleaved caspase 7 and anti-cleaved PARP (Po 1 y-ADP-r i bose - Polymerase)
  • the antibody was analyzed by Western blotting.
  • Example 7 The interaction between the leptin receptor (ob-R) and the progesterone receptor (PgR) after dosing is shown in Fig. 8A and Fig. 8B, respectively, which are analyzed by immunoprecipitation according to the present invention.
  • FIG. 8B Further explore whether there is some interaction between the leptin receptor and the progesterone receptor.
  • the cells are treated with leptin or MPA alone, and lean hormone combined with MPA for 30 minutes.
  • the cell protein was immunoprecipitated with anti-pgR antibody, and the anti-ob-R antibody was used to analyze whether the leptin receptor and the progesterone receptor were combined by the Western blotting method. After 30 minutes of MPA addition, the leptin receptor does bind to the progesterone receptor. After 30 minutes of treatment with estrogen and MPA, the leptin receptor binding to the progesterone receptor was more pronounced, as shown in Figure 8A. The same result was obtained by immunofluorescence staining.
  • Example 8 Decreasing the expression of the estrogen receptor to reduce the effect of leptin combined with MPA
  • FIG. 9 a schematic diagram of the analysis of the present invention for inhibiting the reduction of the estrogen receptor to enhance the action of MPA.
  • the leptin receptor and progesterone receptors will interact with each other after dosing.
  • the hormone will lose the effect of strengthening the MPA inhibiting cells, and will not increase the therapeutic effect of MPA.
  • the leptin receptor siRNA was transferred to the cell culture day at 20 nM, and the next day was changed.
  • the serum-free (Serum-Free) medium was treated with leptin and MPA for 24 hours, and the effect of cell growth was analyzed by XTT assay.
  • the effect of leptin on strengthening MPA is mainly through thin Hormones and their receptors regulate this function.
  • Example 9 Increasing the expression levels of the leptin receptor and the progesterone receptor, and increasing the apoptosis of the cells after dosing, as shown in Fig. 10A and Fig. 10B, respectively, adding long-type lean hormones to the present invention.
  • the expression of the receptor allows the cells to be analyzed for apoptosis after dosing, and the present invention increases the expression of the progesterone receptor to visualize the apoptosis of the cells after dosing.
  • the therapeutic effect of leptin-enhancing MPA did indeed act through the activation of the estrogen receptor.
  • ob-Rb plasmid was transferred to cell culture for one day, the next day was replaced with serura-free medium, and then combined with estrogen and MPA for 24 hours, XTT
  • the assay analyzes the effects of cell growth by observing the effects of the cells after dosing by expressing the cells in large amounts of the long-term leptin receptor (ob-Rb).
  • ob-Rb long-term leptin receptor
  • Fig. 10A when the cells express a large amount of long-term leptin receptors, the effect of improving MPA treatment is markedly increased.
  • long-type leptin receptors appear to increase the performance of progesterone receptors.
  • PgR plasmid was further transferred to cell culture for one day, and the next day was replaced with serum-free medium, and then treated with leptin and MPA for 24 hours, and the effect of cell growth was analyzed by XTT assay.
  • the effect of enhancing the action of MPA is observed by expressing the cells in large amounts of progesterone receptors.
  • the results are shown in Fig. 10B, which is also in line with the expectation of the present invention, and indeed, when the cells express a large amount of progesterone receptors, the therapeutic effect of MPA is also enhanced.
  • it was found that the action of leptin combined with MPA is due to the indirect increase of progesterone receptor expression by the leptin receptor, thereby improving the therapeutic effect of MPA.
  • Example 10 Lean Hormone Enhances MPA Inhibition JAK/STAT Message Delivery Path
  • FIG. 1 1 is a schematic diagram showing changes in the JAK/STAT path of the present invention after drug treatment.
  • this example uses western blot method to analyze whether the downstream related proteins are treated by the combination of leptin and MPA for 24 hours. change.
  • p-ob-R, ob-R and PgR were inhibited after 24 hours of treatment with the drug.
  • the effect of inhibition by lean hormone combined with MPA was more obvious than that of MPA alone.
  • the results were also suppressed. Therefore, it was found that MPA inhibits the progesterone receptor and related proteins downstream, so that it loses its original function, and the inhibitory effect of the cells after treatment with leptin and MPA is more obvious.
  • Example 11 Leptin Enhances MPA Inhibition MAPK Message Delivery Path
  • FIG. 12 a schematic diagram showing the change of the MAPK pathway of the present invention after the drug treatment.
  • this example attempts to observe whether the MAPK-related message pathway and the upstream and downstream proteins also change, and the protein expression is analyzed by Western blotting under the same conditions. Real The results showed that the expression levels of the APK-related proteins p-JNK, JNK, p-ERKl/2, ERKl/2, p-p38 and p38 were all inhibited, demonstrating that MPA not only inhibits the downstream proteins associated with the leptin receptor. It also inhibits other message pathways to inhibit the growth of liver cancer cells, and its estrogen also enhances MPA inhibition.
  • Example 12 Observation of changes in estrogen molecules by MPA on a short time point
  • FIG. 1 3 A and FIG. 1 3 B are schematic diagrams showing changes in the JAK/STAT path in the short-term performance of the JAK/STAT path in the present invention, and changes in the performance of the APK path in the short-term period of the present invention.
  • the results of the above experiments showed that the proteins in the JAK2/STAT3 and MAPK-related signal pathways of the drug-treated cells were inhibited in 24 hours, possibly because the cells were in apoptotic state at 24 hours.
  • Related protein degradation does not reveal which of the relevant message pathways changes, so this example uses short time points to observe changes in related proteins.
  • the cells were treated with high concentrations of estrogen and high concentrations of MPA and high concentrations of estrogen combined with high concentrations of MPA for 30 minutes, 3 hours and 6 hours, respectively. Collecting cells according to time points JAK/STAT-related proteins are analyzed by Western blotting methods for protein expression. The results showed that in the 30th minute, the combination of lean hormone and MPA began to degrade p-ob-R, and the degree of degradation became more obvious with time. However, P-STAT3 (Tyr705) was also inhibited at the third hour, and the effect of inhibition was also more obvious than that of leptin combined with MPA, as shown in Fig. 13 A.
  • the present invention is intended to inhibit the activation of ERK1/2 and the expression of PIAS3 to reduce the degradation of p-STAT3.
  • the above findings showed that p-ERK1/2 alone was overexpressed with MPA and lean hormone combined with MPA at the 30th minute and the third hour.
  • This example attempts to find a relevant path to clarify p-ERKl/2 activation.
  • p-ERKl/2 activation induces the activation of P-STAT3 inhibitor-activated transcriptional activator 3 (PIAS3) and inhibits the expression of P-STAT3 (Tyr705).
  • Apoptosis (as shown in Figure 1 3 B). Therefore, in this example, the cells were added with 10 p-ERKl/2 inhibitor (U0126) 4 hours before the dosing, and then the cells were treated with estrogen and MPA for 3 hours, and the cellular proteins were collected and analyzed by Western blotting. The amount of performance. It was found by experiments that the phosphorylation of p-ERK1/2 was inhibited, and PIAS3 was not induced to activate, and the expression of p-STAT3 (Tyr705) was not inhibited.
  • MPA may indirectly activate PIAS3 by activating p-ERK1/2 to inhibit apoptosis of p-STAT3 (Tyr705), which enhances the action of MPA to activate p-ERK1/2. Increased apoptosis.
  • Example 14 Cell mass expression of leptin enhances MPA activity in inhibiting cell growth
  • FIG. 15 to FIG. 1 6B are respectively a schematic diagram showing the activity analysis of a large amount of estrogen-enhanced MPA inhibiting cell growth in the cells of the present invention, and the present invention is related to STAT3 by MPA treatment after a large amount of expression of leukemia in the cells.
  • a schematic diagram of the analysis of the message path, and a schematic diagram of the analysis of the MAPK-related message pathway by MPA treatment after the present invention expresses a large amount of lean hormone in the cell.
  • the combination of lean hormone and MPA inhibits the hematopoietic cells of the liver cancer cells
  • the present invention in this embodiment, in order to test whether the liver cancer cells themselves can secrete a large amount of lean hormones, Will enhance the role of MPA in inhibiting liver cancer cells. So after the laser quality thin body (pcLeptin) colonization transferred to the cell culture one day, the next day medium was replaced with serum-free, and then processed to 10- 4 M MPA for 24 hours, XTT assay Analysis of the cell growth. As shown in the left half of Figure 15, the cell's transient translocation of the estrogen gene allows the cells to secrete a large amount of estrogen.
  • pcLeptin laser quality thin body
  • the secretion of the estrogen by the cells itself also enhances the inhibition of liver cancer cells by MPA.
  • the medium was collected and subjected to immunoprecipitation using a leptin antibody, followed by Western blot analysis, as shown in the right half of Fig. 15, when the leptin gene was expressed in a large amount in the cell. , will cause the cells to secrete leptin from the body and can be measured in the medium.
  • the protein expression of the STAT and MAPK-related message pathways is also analyzed by Western blotting. As shown in FIG.
  • the pro-liposome is transferred to the cell culture for one day, the next day.
  • the JAK/STAT-related protein was analyzed by Western blotting. It was found that when the cells expressed a large amount of lean hormone, MPA was added for 24 hours. The degree of STAT3 activation was also more pronounced than that of cells treated with MPA alone, and the cellular MAPK message pathway-related proteins were analyzed by Western blotting under the same conditions, as shown in Figure 16 B, in the MAPK-related protein ERK, JNK. , p38, c-fos and c-jun are also the same result.
  • Example 15 Effect of leptin combined with MPA on normal liver epithelial cell lines
  • FIG. 1 7 to FIG. 1 8B are respectively a schematic diagram of the analysis of the growth of normal liver epithelial cell lines by the combination of lean hormone and MPA, and the STAT3 related message path of the normal liver cells of the present invention treated with lean hormone combined with MPA.
  • the cell growth ie, survival rate
  • the treatment of liver cancer cells with the combination of lean hormone and MPA can indeed enhance the effect of MPA inhibiting liver cancer cells, and is better than using MPA alone; in addition, the present invention displays liver cancer cell lines in large amounts to express hormone proteins. The same effect was also found. Accordingly, it is expected that the present invention will be applied to the treatment of clinical liver cancer patients in the future.
  • the present invention can detect the expression amount of leukemia in the serum of liver cancer patients or liver cancer tissues; in a preferred embodiment, for patients with higher performance, direct treatment with MPA can be considered, thereby achieving enhanced MPA inhibition.
  • the present invention can also combine leptin and MPA to design a drug for treating liver cancer patients, thereby expanding the scope of use of the drug and enhancing the pharmacological action of MPA;
  • the present invention may further be administered to MPA after increasing the amount of estrogen in a patient with a low expression of estrogen, such as 5-hydroxy-trypton (5-HTP). Therefore, the present invention can help liver cancer patients maintain a longer survival time.
  • the present invention is a method for treating liver cancer by using a thin hormone, which can effectively improve various disadvantages, and includes a therapeutic drug, a course of treatment or a screening platform for administering a therapeutically effective amount to a liver cancer patient.
  • the cell has a specific pharmacological effect on the normal liver epithelial cell line and the cancer cell line, and the donor can have no adverse side effects on the normal liver cell, and can prolong the overall survival rate of the liver cancer patient, thereby further making the present invention
  • the production can be more progressive, more practical, and more in line with the needs of the user. It has indeed met the requirements for the invention patent application, and loves to file a patent application according to law.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Endocrinology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Genetics & Genomics (AREA)
  • Obesity (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Use of leptin in enhancing the effect of medroxyprogesterone acetate on inhibition of hepatocarcinoma cells. When the level of leptin in hepatoma patients is higher, medroxyprogesterone acetate is administrated directly or combined with leptin. When the level of leptin in patients is lower, 5-hydroxy tryptophan is administrated for increasing the level of leptin, and then medroxyprogesterone acetate is administrated.

Description

说 明 书  Description
痩激素在强化醋酸甲羟孕酮治疗肝癌中的应用  Application of sputum hormone in strengthening medroxyprogesterone acetate for treatment of liver cancer
技术领域 Technical field
本发明系有关于一种以瘦激素治疗肝癌的应用, 尤指涉及一种对肝癌病患投予医药有 效量的一治疗药物、 疗程或筛选平台, 特别系指将瘦激素 (Leptin ) 或提高肝癌病患瘦激 素生理浓度之合并一醋酸甲羟孕酮 (Medroxyprogesterone acetate, MPA ) 后与肝癌细胞 接触以毒杀肝癌细胞者。  The invention relates to a method for treating liver cancer by using a thin hormone, in particular to a therapeutic drug, a course of treatment or a screening platform for administering a therapeutically effective amount to a liver cancer patient, in particular to increase or decrease the leptin (Leptin). The physiological concentration of leptin in patients with liver cancer is combined with medroxyprogesterone acetate (MPA) and then contacted with liver cancer cells to kill liver cancer cells.
背景技术 Background technique
根据美国方面的统计, 过去二十年间, 美国肝癌发生率有显著增加的情形, 且男性系 女性的三倍, 而黑人系白人的两倍。 另外, 对于中国台湾、 日本、 韩国及大陆等东方国家 方面而言, 由于属于 B 型肝炎病毒及肝硬化的高盛行区, 故肝癌在东方国家为常见恶性肿 瘤, 且由临床观察发现, 含有 5§〜¾)%的肝癌与肝炎及肝硬化症有关。 于其中, 以中国台 湾地区十大死亡原因统计数据显示, 自公元一九八二年以来, 癌症即跃居首位, 其中以肝 癌对男性而言系所有癌症死因的第一位, 而对于女性而言也高居第二位, 且每年更造成四 千人死亡, 足以显见其对国人健康造成极大的威胁。 因此, 肝癌早期诊断及治疗将是未来 研究重点之一。  According to US statistics, there has been a significant increase in the incidence of liver cancer in the United States over the past two decades, with three times as many as men and two times as many as blacks. In addition, for the eastern countries such as Taiwan, Japan, South Korea and the mainland, because of the high-risk area of hepatitis B virus and cirrhosis, liver cancer is a common malignant tumor in the eastern countries, and it is found by clinical observation that it contains 5 § ~3⁄4)% of liver cancer is associated with hepatitis and cirrhosis. Among them, the statistics of the top ten causes of death in Taiwan, China, show that since 1982, cancer has leapt to the top, with liver cancer being the first cause of all cancer deaths for men and for women. It is also the second highest, and it kills 4,000 people every year, which is enough to show that it poses a great threat to the health of Chinese people. Therefore, early diagnosis and treatment of liver cancer will be one of the research priorities in the future.
在目前临床上肝癌治疗方式大都以外科手术来治疗并改善病人的存活率, 但却只有 10〜15 %的肝癌病人可用于手术治疗, 其余不能以手术治疗的病人则试图以放射疗法、 化 学疗法或免疫疗法来延长肝癌病人的存活率, 但其结果都不尽理想, 且化学治疗及放射治 疗亦会对于正常人体细胞带来损害, 进而引发病人种种不适的副作用发生。  In the current clinical treatment of liver cancer, most of them are treated with surgery to improve the survival rate of patients, but only 10 to 15% of patients with liver cancer can be used for surgery. The rest of patients who cannot be treated with surgery are trying to use radiation therapy or chemotherapy. Or immunotherapy to prolong the survival rate of patients with liver cancer, but the results are not satisfactory, and chemotherapy and radiation therapy will also cause damage to normal human cells, which may lead to side effects of various discomforts.
有鉴于此, 在其它相关研究及报导中, 另发现由脂肪细胞所分泌的细胞激素——瘦激 素与肝癌的发生有相当密切的关系性。 截至目前为止, 已被证实与瘦激素有关的癌症, 包 括有子宫内膜癌及乳癌等。 研究指出, 瘦激素透过与其相对应的膜表面接受器结合后, 可 藉由转录讯息传递活化子 3 (STAT3 ) 的讯息传递系统来调控基因表现, 而这些下游基因有 许多都与癌症发生有关, 如: 血管内皮生长因子 (Vascular Endothelial Growth Factor, VEGF) 。 然而, 近年来荷尔蒙治疗在肝癌治疗上似乎开启一页新的契机, 故在肝癌治疗方 式上己慢慢地趋向于以荷尔蒙疗法来治疗肝癌病人, 且在临床上及实验室研究亦发现肝癌 系一种性荷尔蒙依赖性的肿瘤型态, 并以雄性激素居多, 与类固醇荷尔蒙及性荷尔接受器 的改变有明显相关。 最近有一种对于肝癌病人新的治疗方式, 糖皮质类固醇的拮抗剂 (Glucocorticoid Antagonists ) , 例如: 黄体酮及 RU486 在人类肝癌可以抑制甲型胎儿 蛋白 ( a -fetoprotein) 的表现。 暗示着黄体酮在肝癌荷尔蒙治疗上具有潜在的可能性。 目前已有研究发现醋酸甲羟孕酮这一类黄体酮类似物能有效地抑制肝癌细胞的生长。 该 MPA系由黄体激素 (17- OH- progesterone) 衍生而来, 为一种合成类固醇的口服给药, 其结构与天然的黄体激素相似, 不同处在于 α - 6 位置有甲基(Methyl Group) 与 17 位 置有乙酸酯官能基 (Acetoxy Group) 。 该 MPA 系一种有抗雌激素活性 (Antiestrogenic Activity) 的黄体激素, 在停经后的乳癌, 其会完全地抑制血浆雌激素的浓度, 同时亦会 抑制黄体激素 (Luteining Hormone, LH) 的释放、 剌激子宫内膜的生长以及在乳房腺泡 细胞 (Acinar Cells ) 产生典型体激素变化。 该 MPA作用机转至今仍不明确, 但被认为涉 及与细胞内荷尔蒙接受器的交互作用。 在其它研究中也指出 MPA会与雌激素、 黄体激素及 雄性素的受体结合, 其中尤以与雄性素受体的结合对此药的细胞毒性作用特别重要。 近来 亦有研究指出 MPA 会抑制肝癌肿瘤的大小及延长肝癌病人的存活率, 相关地研究也证实 MPA可能影响肝癌演化的过程进而改善病人治疗的预后。 如此, 利用 MPA治疗临床上的肝 癌病人或许对于病人之存活率系有所帮助的, 但由于 MPA 具多重药理生物活性, 故对于其 作用机制仍需进一步地探讨。 In view of this, in other related research and reports, it was found that the cytokine secreted by fat cells, leptin, has a close relationship with the occurrence of liver cancer. To date, cancers related to lean hormones have been identified, including endometrial cancer and breast cancer. Studies have shown that by binding to their corresponding membrane surface receptors, estrogen can regulate gene expression through transcriptional signaling of the activator 3 (STAT3) signaling system, many of which are associated with cancer. , such as: Vascular Endothelial Growth Factor (VEGF). However, in recent years, hormone therapy seems to open a new opportunity in the treatment of liver cancer. Therefore, the treatment of liver cancer has gradually tended to treat liver cancer patients with hormone therapy, and clinical and laboratory studies have also found that liver cancer is also found. A sex-dependent hormone type with predominantly androgen, which is significantly associated with changes in steroid hormones and sex receptors. Recently, there is a new treatment for liver cancer patients, Glucocorticoid Antagonists, for example: Progesterone and RU486 can inhibit the expression of a-fetoprotein in human liver cancer. It suggests that progesterone has a potential for hormone therapy in liver cancer. At present, it has been found that progesterone acetate, a progesterone analog, can effectively inhibit the growth of liver cancer cells. The MPA is derived from the luteinizing hormone (17-OH-progesterone) and is administered orally as a synthetic steroid. Its structure is similar to that of the natural luteinizing hormone, except that it has a methyl group at the α - 6 position (Methyl Group). There is an acetate functional group (Acetoxy Group) at position 17. The MPA is a luteinizing hormone with antiestrogenic activity. In postmenopausal breast cancer, it completely inhibits the concentration of plasma estrogen and also inhibits the release of Luteining Hormone (LH). Stimulates the growth of the endometrium and produces typical body hormone changes in breast acinar cells. The MPA action machine is still unclear, but is believed to involve interaction with intracellular hormone receptors. In other studies, it has also been pointed out that MPA binds to receptors of estrogen, progesterone and androgen, and in particular, binding to androgen receptors is particularly important for the cytotoxic effects of this drug. Recently, studies have also indicated that MPA can inhibit the size of liver cancer tumors and prolong the survival rate of liver cancer patients. Related studies have also confirmed that MPA may affect the process of liver cancer evolution and improve the prognosis of patients. Thus, the use of MPA to treat clinical liver cancer patients may be helpful for patient survival, but because MPA has multiple pharmacological biological activities, its mechanism of action needs to be further explored.
对此, 已有一些临床试验证明 MPA 对末期肝癌病患具有抑制效果, 相关研究也证明它 能抑制肝癌细胞株 (HepG2 Cells ) 的生长。 同时, 在本案申请人之前研究分析结果当中, 并没有发现肝癌组织中瘦激素的表现情形以及有无手术后使用 MPA与病患存活时间呈现有 意义的关系 (如图 1所示, P值分别为 0. 383与 0. 171 ) 。 另外, 肝癌组织中微血管密度值 以及 Ki- 67的表现, 也证明与病患存活时间没有呈现有意义的关系。  In this regard, some clinical trials have shown that MPA has an inhibitory effect on end-stage liver cancer patients, and related studies have also shown that it can inhibit the growth of HepG2 cells. At the same time, in the research results of the applicant's previous study, there was no significant relationship between the expression of leptin in liver cancer tissues and the use of MPA after surgery and the survival time of patients (as shown in Figure 1, P values were respectively 0. 383 and 0. 171). In addition, the microvessel density values in liver cancer tissues and the performance of Ki-67 also showed no significant relationship with the survival time of patients.
综上所述, 瘦激素属于单链蛋白质成分的荷尔蒙, 一般被认为在体重维持及能量调节 上扮演重要角色, 而 MPA则为黄体激素衍生的合成类固醇。 目前相关研究已证实 MPA确实 可在活体内暨试管内的系统中造成肝癌细胞株的细胞凋亡, 即该 MPA系可在细胞株系统中 有效地抑制肝癌细胞株生长; 然而, 在国外相关研究中却发现, 以 MPA 去治疗肝癌病患对 于延长存活率并没有显著影响。 虽然目前 MPA系己上市之癌症用药, 然而对于肝癌病患并 没有令人振奋之疗效。 因此, 以 MPA在延长肝癌病人存活率上尚未达到统计上的差异, 且 除了缺乏在临床有效证据外, 亦未出现有考虑根据肝癌病患血清及肝癌组织中如瘦激素等 生物标志的表现量来配合 MPA 的合并给药。 故, 一般已用者无法符合使用者于实际使用时 所需。  In summary, estrogen is a hormone of a single-chain protein component and is generally considered to play an important role in weight maintenance and energy regulation, while MPA is a progesterone-derived synthetic steroid. At present, relevant studies have confirmed that MPA can cause apoptosis of liver cancer cell lines in a system in vivo and in vitro, that is, the MPA system can effectively inhibit the growth of liver cancer cell lines in a cell line system; however, relevant research abroad However, it was found that treatment of liver cancer patients with MPA had no significant effect on prolonging survival. Although MPA is currently on the market for cancer, there is no exhilarating effect on liver cancer patients. Therefore, there is no statistical difference in the survival rate of patients with liver cancer by MPA, and in addition to the lack of clinically valid evidence, there is no consideration of the biomarkers such as estrogen and other biomarkers in liver cancer tissues and liver cancer tissues. To cooperate with the combined administration of MPA. Therefore, the average user has not been able to meet the needs of the user in actual use.
发明内容 Summary of the invention
本发明主要目的在于, 克服已知技艺所遭遇的上述问题并提供一种以瘦激素强化 MPA 对于抑制肝癌细胞的应用。  The main object of the present invention is to overcome the above problems encountered in the prior art and to provide an application of attenuating MPA with a leptin to inhibit liver cancer cells.
本发明次要目的在于, 提供一种当肝癌病患血清中或肝癌组织中瘦激素表现量较高 时, 直接给予 MPA治疗肝癌病患, 以利达到强化 MPA的药理作用。 本发明另一目的在于, 提供一种当肝癌病患血清中或肝癌组织中瘦激素的表现量较高 时, 将瘦激素及 MPA合并成一药物治疗肝癌病患, 以利达到强化 MPA的药理作用。 A secondary object of the present invention is to provide a method for directly treating MPC patients with liver cancer patients in the serum of liver cancer patients or in liver cancer tissues, so as to achieve the pharmacological action of enhancing MPA. Another object of the present invention is to provide a method for treating liver cancer patients by combining leptin and MPA into a drug when the expression of lept hormone in the serum of liver cancer patients or liver cancer tissues is high, so as to achieve the pharmacological action of strengthening MPA. .
本发明再一目的在于, 提供一种当肝癌病患血清中或肝癌组织中瘦激素的表现量较低 时, 以 5-羟色氨酸 (5- hydroxy- trypton, 5- HTP) 提高瘦激素量后, 再给予 MPA治疗, 以 利达到强化 MPA的药理作用。  A further object of the present invention is to provide a method for increasing leptin by 5-hydroxy-trypton (5-HTP) when the expression of leptin in serum or liver cancer tissues of liver cancer patients is low. After the amount, MPA treatment is given to achieve the pharmacological action of strengthening MPA.
为达以上目的, 本发明系一种以瘦激素治疗肝癌的应用, 系包含对肝癌病患投予医药 有效量的一治疗药物、 疗程或筛选平台, 其特征系将瘦激素或提高肝癌病患瘦激素生理浓 度合并一 MPA后与肝癌细胞接触, 以该瘦激素透过其受体与黄体酮受体的交互作用加强及 加速该 MPA毒杀肝癌细胞, 藉以对于正常肝脏上皮细胞株与癌细胞株具有专一性的药理作 用, 以供可对正常肝脏细胞无不良副作用同时, 并能延长肝癌病患整体存活率。 其中, 该 瘦激素的表现量高低更进一步可作为肝癌病患对 MPA 治疗有无疗效的预测因子 (Predictive Factors) 及其存活时间的预后因子 (Prognostic Factors) 。  In order to achieve the above object, the present invention relates to a method for treating liver cancer by using a lepid hormone, comprising a therapeutic drug, a course of treatment or a screening platform for administering a therapeutically effective amount to a liver cancer patient, characterized in that the hormone is increased or the liver cancer patient is raised. The physiological concentration of lean hormone combined with a MPA is contacted with liver cancer cells, and the interaction of the estrogen and its progesterone receptor through the receptor enhances and accelerates the MPA to kill liver cancer cells, thereby normalizing the liver epithelial cell line and cancer cells. The strain has a specific pharmacological effect, so that it can have no adverse side effects on normal liver cells, and can prolong the overall survival rate of liver cancer patients. Among them, the level of expression of the estrogen can further serve as a predictor of prophylactic factors for the treatment of MPA in patients with liver cancer and a prognostic factor for survival.
附图说明 DRAWINGS
图 1, 系本发明以肝癌病患术后使用 MPA治疗的预后存活时间的分析示意图。  Figure 1 is a schematic diagram showing the analysis of the prognosis survival time of patients with liver cancer using MPA after surgery.
图 2 , 系本发明以瘦激素在不同浓度下对细胞生长的分析示意图。  Figure 2 is a schematic diagram showing the analysis of cell growth by estrogen at different concentrations in the present invention.
图 3, 系本发明以 MPA在不同浓度下对细胞生长的分析示意图。  Figure 3 is a schematic diagram showing the analysis of cell growth by MPA at various concentrations in the present invention.
图 4, 系本发明以瘦激素合并 MPA处理后对细胞生长的分析示意图。  Figure 4 is a graphical representation of the analysis of cell growth after treatment with leptin and MPA in the present invention.
图 5, 系本发明以瘦激素合并 MPA处理细胞后之细胞型态示意图。  Fig. 5 is a schematic view showing the cell type of the present invention after treating cells with leptin and MPA.
图 6 A, 系本发明以瘦激素合并 MPA处理细胞后对细胞周期的影响示意图。  Fig. 6 A is a schematic diagram showing the effect of the present invention on cell cycle after treating cells with leptin and MPA.
图 6 B, 本发明以瘦激素合并 MPA处理细胞后对细胞周期的分析示意图。  Fig. 6B is a schematic diagram showing the analysis of cell cycle after treating cells with estrogen and MPA.
图 7, 系本发明以瘦激素合并 MPA处理后对细胞凋亡相关蛋白质的影响示意图。  Figure 7. Schematic diagram of the effect of the present invention on apoptosis-related proteins after treatment with leptin and MPA.
图 8 A , 系本发明以免疫沉淀法分析瘦激素受体与黄体酮受体接受器交互结合作用的 示意图。  Fig. 8 A is a schematic diagram showing the interaction of the leptin receptor with the progesterone receptor receptor by immunoprecipitation.
图 8 B, 系本发明以免疫荧光染色分析瘦激素受体与黄体酮受体接受器交互结合作用 的示意图。  Figure 8B is a schematic representation of the interaction of the leptin receptor with the progesterone receptor receptor by immunofluorescence staining.
图 9, 系本发明抑制瘦激素受体减少加强 MPA作用的分析示意图。  Figure 9. Schematic diagram of the analysis of the present invention for inhibiting the reduction of the estrogen receptor and enhancing the action of MPA.
图 1 0 A, 系本发明增加长型瘦激素受体的表现量使细胞在加药后的凋亡分析示意 图。  Figure 10 A is a schematic representation of the increase in expression of the long form of the estrogen receptor in the present invention to allow apoptosis of the cells after dosing.
图 1 0 B, 系本发明增加黄体酮受体之表现量使细胞在加药后的凋亡分析示意图。 图 1 1, 系本发明 JAK/STAT路径以药物处理后表现量的变化示意图。  Figure 10B is a schematic diagram showing the analysis of apoptosis of cells after dosing by increasing the amount of progesterone receptor expression. Fig. 1 is a schematic diagram showing changes in the amount of JAK/STAT pathway after drug treatment in the present invention.
图 1 2, 系本发明 MAPK路径以药物处理后表现量的变化示意图。  Fig. 1 2 is a schematic diagram showing changes in the amount of expression of the MAPK pathway of the present invention after drug treatment.
图 1 3 A, 系本发明 JAK/STAT路径以药物处理短时间内表现量的变化示意图。 图 1 3 B, 系本发明 MAPK路径以药物处理短时间内表现量的变化示意图。 Fig. 1 3 A is a schematic diagram showing changes in the amount of expression of the JAK/STAT path of the present invention in a short period of time after drug treatment. Fig. 1 3 B is a schematic diagram showing changes in the amount of expression of the MAPK pathway of the present invention in a short time by drug treatment.
图 1 4, 系本发明以抑制 ERK1/2 的活化及 PIAS3 的表现量减少 p-STAT3 的降解示意 图。  Fig. 14 is a schematic diagram showing the degradation of p-STAT3 by inhibiting the activation of ERK1/2 and the expression of PIAS3 in the present invention.
图 1 5, 系本发明于细胞内大量表现瘦激素加强 MPA 抑制细胞生长的活性分析示意 图。  Fig. 15 is a schematic diagram showing the activity analysis of a large amount of leptin in the cells to enhance MPA inhibition of cell growth in the cells.
图 1 6 A, 系本发明于细胞内大量表现瘦激素后以 MPA 处理对于 STAT3相关讯息路径 的分析示意图。  Fig. 1 6 A is a schematic diagram of the analysis of the STAT3 related message pathway by MPA treatment in the present invention.
图 1 6 B, 系本发明于细胞内大量表现瘦激素后以 MPA 处理对于 MAPK相关讯息路径 的分析示意图。  Fig. 1 6 B is a schematic diagram of the analysis of the MAPK-related message pathway by MPA treatment in the present invention.
图 1 7, 系本发明以瘦激素合并 MPA对正常肝脏上皮细胞株生长的分析示意图。  Fig. 17 is a schematic diagram showing the analysis of the growth of normal liver epithelial cell lines by the combination of leptin and MPA in the present invention.
图 1 8 A, 系本发明正常肝脏细胞以瘦激素合并 MPA处理后对 STAT3相关讯息路径的 分析示意图。  Fig. 1 8 A is a schematic diagram showing the analysis of STAT3 related message pathways after treatment of lean liver cells with MPA by normal liver cells of the present invention.
图 1 8 B, 系本发明正常肝脏细胞以瘦激素合并 MPA处理后对 MAPK相关讯息路径的分 析示意图。  Fig. 1 8 B is a schematic diagram of the MAPK-related message pathway of normal liver cells of the present invention treated with estrogen and MPA.
具体实施方式 detailed description
本发明系一种以瘦激素治疗肝癌的应用, 可作为在肝癌细胞株 (HepG2 Cells ) 上及临 床肝癌病患的统计数据。 其包含对肝癌病患投予医药有效量的一治疗药物、 疗程或筛选平 台, 其特征系将瘦激素 (Leptin) 或提高肝癌病患瘦激素生理浓度之合并一细胞毒性剂-醋 酸甲羟孕酮 (Medroxyprogesterone acetate, MPA) 后与肝癌细胞接触, 以该瘦激素透过 其受体与黄体酮受体的交互作用加强及加速该 MPA抑制肝癌细胞的毒杀效果, 藉以对于正 常肝脏上皮细胞株与癌细胞株具有专一性的药理作用, 俾供以瘦激素合并 MPA之药物或疗 程可造成肝癌病患癌细胞大小降低, 即该药物可分别地造成肝癌细胞尺寸或质量减少, 并 且对正常肝脏细胞无毒杀之不良副作用产生, 进而能有效延长肝癌病患之整体存活率。  The invention relates to a method for treating liver cancer by using a lepid hormone, and can be used as statistical data on liver cancer cell lines (HepG2 Cells) and clinical liver cancer patients. The invention comprises a therapeutic drug, a course of treatment or a screening platform for administering a medicinal effective amount to a liver cancer patient, which is characterized by a combination of a leptin (Leptin) or a physiological concentrating agent for increasing the physiological concentration of leptin in a liver cancer patient. After exposure to melanocytogenes (MPA), the interaction of the estrogen and its progesterone receptor through the receptor enhances and accelerates the MPA inhibiting the killing effect of the liver cancer cells, thereby normalizing the liver epithelial cell line. It has a specific pharmacological effect with cancer cell lines. The drug or course of treatment with leptin combined with MPA can cause the cancer cells of liver cancer patients to decrease in size, that is, the drug can respectively reduce the size or quality of liver cancer cells, and is normal. The adverse side effects of non-toxic killing of liver cells can effectively prolong the overall survival rate of liver cancer patients.
本发明在活体外的研究及部分临床病患实证上己发现瘦激素与 MPA对于肝癌细胞有显 著抑制效果及延长病患存活率的现象。 据此, 将瘦激素及 MAP 合并设计成一治疗药物, 用 以治疗肝癌病患, 并且亦发现在正常人瘦激素生理浓度就几可以加强 MPA疗效。 于其中, MPA在本发明一较佳实施例中的临床病人以每日 500 毫克 (mg) 的剂量来治疗, 因此, 在 本发明利用将瘦激素或提高病患瘦激素生理浓度之合并 MPA之后, 可达到更好治疗作用。 请参阅图 1所示, 系本发明以肝癌病患术后使用 MPA 治疗预后存活时间的分析示意图。 如 图所示: 系本发明以上述较佳实施例中将临床肝癌病患统计分析结果, 其中, 可发现肝癌 组织中瘦激素之表现情形以及有无手术后使用 MPA 与病患的存活时间并没有呈现有意义的 关系, 如图 1中左上及右上所示 P值分别为 0. 383与 0. 171; 惟, 在手术后有使用 MPA的 肝癌病患中, 分析结果发现瘦激素高表现者比低表现者有较长存活时间, 如图 1中左下所 示 P值为 0. 008。 The invention has found that the effects of lean hormone and MPA on liver cancer cells and prolonging the survival rate of patients have been found in the study of in vitro and in some clinical patients. Accordingly, the combination of leptin and MAP was designed as a therapeutic drug for the treatment of liver cancer patients, and it was also found that the physiological concentration of lean hormone in normal humans can enhance the efficacy of MPA. Among them, MPA is treated at a dose of 500 mg (mg) per day in a clinical patient in a preferred embodiment of the present invention, and therefore, after the present invention utilizes a combination of MPA to increase the physiological concentration of estrogen or estrogen , can achieve better therapeutic effects. Please refer to FIG. 1 for the analysis of the prognosis survival time of the liver cancer patients after treatment with MPA. As shown in the figure: The present invention compares the results of clinical analysis of clinical liver cancer patients in the above preferred embodiments, wherein the performance of leukemia in liver cancer tissues and the survival time of MPA and patients after surgery can be found. There is no meaningful relationship. The P values shown in the upper left and upper right of Figure 1 are 0. 383 and 0. 171; respectively, there is MPA after surgery. In the case of liver cancer, the analysis showed that the high-performance of the estrogen was longer than the low-performance one, as shown in the lower left of Figure 1, the P value was 0.001.
经此临床实验可知, 瘦激素表现量高的肝癌病患在外科手术后接受荷尔蒙 (即 MPA) 治疗的预后及存活率明显地优于瘦激素表现量低的肝癌病患。 因此, 足以确认该瘦激素可 加强 MPA在肝癌治疗上的效果, 且该瘦激素的表现量高低更进一步可作为肝癌病患对 MPA 治疗有无疗效的预测因子 (Predictive Factors ) 及其存活时间的预后因子 (Prognostic Factors) , 其中尤以该瘦激素的表现量越高, 其强化 ΜΡΑ对肝癌细胞的抑制效果越明显, 藉此可作为肝癌病患在外科手术后接受荷尔蒙疗法 (即 ΜΡΑ) 的指标, 进而能有助于维持 或延长肝癌病患的存活时间。  According to this clinical experiment, the prognosis and survival rate of patients with liver cancer who have high expression of estrogen after surgery are significantly better than those with low expression of estrogen. Therefore, it is sufficient to confirm that the leptin can enhance the effect of MPA on the treatment of liver cancer, and the expression level of the estrogen can further serve as a predictor of the efficacy of the treatment of liver cancer patients with MPA (Predictive Factors) and its survival time. Prognostic Factors, in which the higher the expression of the estrogen, the more obvious the inhibitory effect of the intensive sputum on hepatocellular carcinoma cells, which can be used as a liver cancer patient to receive hormone therapy (ie, sputum) after surgery. Indicators, in turn, can help maintain or prolong the survival of liver cancer patients.
基于上述结果, 瘦激素表现量较高的肝癌病患使用 ΜΡΑ 的预后会比瘦激素表现量低的 病患还要好, 并且存活率可延长将近五年之久, 此为其它相关己知技术中未被发现之。 而 本发明在活体外的实验中也证实瘦激素会透过某些机制增强 ΜΡΑ抑制肝癌细胞的作用, 且 除了可将瘦激素及 MAP 合并设计成一治疗药物或疗程之外, 本发明亦可提供瘦激素的筛选 平台以决定是否执行 MPA治疗。  Based on the above results, the prognosis of patients with liver cancer who have higher expression of estrogen is better than that of patients with low expression of estrogen, and the survival rate can be extended for nearly five years. This is among other related technologies. Undiscovered. However, the present invention also demonstrates in an in vitro experiment that leptin enhances the action of sputum to inhibit liver cancer cells through certain mechanisms, and in addition to combining leptin and MAP into a therapeutic drug or treatment, the present invention may also provide A platform for screening for lean hormones to determine whether to perform MPA treatment.
瘦激素合并 MPA 系一个新的能运用在肝癌病患的治疗方式, 在目前国内外并没有相关 报导及研究。 与其它己知文献不同点在于本发明系将 MPA 合并瘦激素为一个新的治疗方 式, 故可有效改善已知技术单单仅以 MPA 来治疗肝癌病患而达不到延长存活率效果的缺 点。  The combination of leptin and MPA is a new treatment that can be used in patients with liver cancer. There are no reports or studies at home and abroad. The difference from other known literatures is that the present invention combines MPA with estrogen as a new treatment method, so that it can effectively improve the shortcomings of the known technique that only MPA is used to treat liver cancer patients without prolonging the survival rate.
本发明后续探讨瘦激素合并 MPA在肝癌细胞中的抑癌角色及其作用的分子机转。 利用 肝癌细胞株以瘦激素合并 MPA处理之后观察其细胞存活率, 并针对其细胞周期、 细胞凋亡 及作用机制进行分析, 同时亦探讨瘦激素及 MPA所参与相关讯息传递路径。 另外, 更检测 瘦激素合并 MPA 的药理作用对于正常肝脏上皮细胞株的影响, 透过观察其药物专一性, 以 期更进一步了解荷尔蒙治疗在肝癌上的运用。  The present invention further explores the anti-cancer role of leptin combined with MPA in liver cancer cells and the molecular mechanism of its action. Hepatocellular carcinoma cell lines were treated with leptin and MPA, and their cell viability was observed. The cell cycle, apoptosis and mechanism of action were analyzed. The related message transmission pathways involved in estrogen and MPA were also explored. In addition, the effects of the pharmacological effects of leptin combined with MPA on normal liver epithelial cell lines were examined by observing their drug specificity, in order to further understand the use of hormone therapy in liver cancer.
实施例 1: 瘦激素对肝癌细胞生长的影响  Example 1: Effect of leptin on the growth of hepatoma cells
请参阅图 2所示, 系本发明以瘦激素在不同浓度下对细胞生长的分析示意图。 如图所 示: 为了解瘦激素对于肝癌细胞的影响, 本实施例分别以两种不同浓度的瘦激素来处理细 胞, 其中高浓度的瘦激素 (LH) 为 100 奈克 (ng) , 低浓度的瘦激素 (LL) 为 10 ng。 于 其中, 上述主要浓度的决定取决于 10 ng 为正常人生理浓度的范围, 可在生理范围时观察 瘦激素对于细胞的影响, 并且就高浓度之决定可了解瘦激素的浓度越高是否对于细胞的影 响就会越大。 经实验结果显示, 以高浓度或低浓度的瘦激素来处理细胞 24小时后, 在 XTT assay分析中发现, 瘦激素对于癌细胞的生长并没有显著影响。  Please refer to FIG. 2, which is a schematic diagram of the analysis of cell growth by different hormones in the present invention. As shown in the figure: In order to understand the effect of leptin on liver cancer cells, in this example, cells were treated with two different concentrations of estrogen, wherein the high concentration of lean hormone (LH) was 100 ng (ng), low concentration The lean hormone (LL) is 10 ng. Among them, the above-mentioned determination of the main concentration depends on the range of physiological concentration of 10 ng as a normal person, and the effect of the estrogen on the cells can be observed in the physiological range, and the higher concentration determines whether the higher the concentration of the estrogen is for the cells. The impact will be greater. The experimental results showed that after treatment of cells with high or low concentrations of estrogen for 24 hours, it was found in the XTT assay that leptin had no significant effect on the growth of cancer cells.
实施例 2: MPA对肝癌细胞生长的影响 请参阅图 3所示, 系本发明以 MPA 在不同浓度下对细胞生长之分析示意图。 如图所 示: 为证明 MPA在治疗肝癌上的效果, 本实施例中选用两种不同浓度的 MPA来处理细胞, 高浓度的 MPA ( MH) 为 10、, 低浓度的 MPA ( ML) 为 10M。 经实验结果显示, 以 XTT assay 分析在高浓度 MPA 处理细胞 24 小时后, 对细胞有明显抑制现象, 在处理 48 小时 后, 对细胞抑制现象有明显增加。 而低浓度 MPA不论在 24 或 48 小时并没有明显抑制效 果。 故表示 MPA随着浓度及时间增加对于细胞的抑制效果就越好。 Example 2: Effect of MPA on the growth of hepatoma cells Please refer to FIG. 3, which is a schematic diagram of the analysis of cell growth of MPA at different concentrations in the present invention. As shown in the figure: To demonstrate the effect of MPA on the treatment of liver cancer, in this example, two different concentrations of MPA were used to treat the cells, with a high concentration of MPA (MH) of 10 and a low concentration of MPA (ML) of 10 M. The experimental results showed that after treatment with high concentration of MPA for 24 hours, the cells were significantly inhibited by XTT assay. After 48 hours of treatment, the cell inhibition phenomenon was significantly increased. The low concentration of MPA did not significantly inhibit the effect at 24 or 48 hours. Therefore, it indicates that the inhibitory effect of MPA on cells with increasing concentration and time is better.
实施例 3: 瘦激素加强 MPA毒杀肝癌细胞的作用  Example 3: Effect of leptin on MPA poisoning of liver cancer cells
请参阅图 4所示, 系本发明以瘦激素合并 MPA 处理后对细胞生长的分析示意图。 如图 所示: 在本实施例中以两种不同浓度的瘦激素搭配两种不同浓度的 MPA来处理细胞。 经实 验结果显示, 高浓度的瘦激素合并高浓度的 MPA来处理细胞 24小时后, 以 XTT assay分析 发现, 其抑制细胞生长效果确实较单独使用 MPA还要好, 其 P值为 0. 001, 而在低浓度瘦 激素合并高浓度 MPA抑制效果也较单独使用 MPA还要好, 其 P值为 0. 01。 惟在高浓度或低 浓度瘦激素合并低浓度 MPA 时, 则并没有较明显的抑制效果。 故经实验结果可确认瘦激素 确实能加强 MPA抑制细胞生长的作用, 且瘦激素浓度越高抑制效果就越明显。  Please refer to FIG. 4, which is a schematic diagram of the analysis of cell growth after treatment with lean hormone combined with MPA. As shown in the figure: In this example, cells were treated with two different concentrations of estrogen in combination with two different concentrations of MPA. The results of the experiment showed that the high concentration of the estrogen combined with the high concentration of MPA to treat the cells for 24 hours, the XTT assay showed that the inhibition of cell growth was indeed better than the MPA alone, and its P value was 0.001, and The P value of 0.01% is better than that of the MPA alone. However, when high concentration or low concentration of lean hormone combined with low concentration of MPA, there is no obvious inhibitory effect. Therefore, the experimental results show that leptin can indeed enhance the effect of MPA on cell growth, and the higher the concentration of leptin, the more obvious the inhibitory effect.
实施例 4: 瘦激素合并 MPA造成细胞凋亡增加  Example 4: Leptin combined with MPA causes increased apoptosis
请参阅图 5所示, 系本发明以瘦激素合并 MPA 处理细胞后的细胞型态示意图。 如图所 示- 从上述结果得知, 瘦激素确实会加强 MPA对细胞抑制效果, 而在本实施例中经显微镜 下观察到细胞型态的结果, 亦如同上述各实施例实验, 瘦激素合并 MPA 处理之后, 细胞凋 亡 (Apoptosis ) 萎缩的程度比单独加入 MPA为之明显。 再者, 本发明更进一步以免疫荧光 法观察细胞核型态。 将细胞以瘦激素合并 MPA处理 24小时过后, 再以荧光染剂 (DAPI ) 染 细胞核, 以荧光显微镜 40X 下观察, 得到结果也是一致的, 其细胞核萎缩程度比单独加入 MPA为之明显。 故经以上结果得知瘦激素确实会加强及加速 MPA所造成细胞凋亡的作用。  Please refer to FIG. 5, which is a schematic diagram of the cell type of the present invention treated with leukemia combined with MPA. As shown in the figure - from the above results, it is known that the estrogen does enhance the cytostatic effect of MPA, and in the present example, the results of the cell type are observed under the microscope, as in the experiments of the above examples, the combination of leptin After MPA treatment, the degree of apoptosis of Apoptosis was significantly greater than that of MPA alone. Furthermore, the present invention further observes the nuclear form by immunofluorescence. After the cells were treated with estrogen and MPA for 24 hours, the nucleus was stained with fluorescent dye (DAPI) and observed under a fluorescence microscope at 40X. The results were also consistent, and the degree of nuclear atrophy was more obvious than that of MPA alone. Therefore, it can be seen from the above results that leptin does enhance and accelerate the apoptosis caused by MPA.
实施例 5: 瘦激素合并 MPA对细胞周期的影响  Example 5: Effect of leptin combined with MPA on cell cycle
请参阅图 6 A及图 6 B所示, 系分别为本发明以瘦激素合并 MPA处理细胞后对细胞周 期的影响示意图、 及本发明以瘦激素合并 MPA处理细胞后对细胞周期的分析示意图。 如图 所示: 为更进一步了解瘦激素合并 MPA在肝癌细胞中的角色, 本实施例系收取单独以瘦激 素或 MPA以及瘦激素合并 MPA处理 24 小时后的细胞, 以流式细胞仪 (Flow Cytometry) 分析药物对细胞周期 (Cell Cycle) 的影响, 并将细胞周期中 Sub- Gl/Apoptosis的部分以 T检定 (Student ' s - test ) 统计数值。 经实验结果显示, 以 MPA处理过的细胞在 sub - G1 有上升趋势, 表示凋亡细胞有明显地增加, 而在 G1及 G2/M期则系下降的, 但在 S期系有 上升现象; 在高浓度瘦激素合并高浓度 MPA来处理细胞 24小时后, 分析发现, sub- G1 增 加趋势较单独使用 MPA为高, 其 P值为 0. 002; 而在低浓度瘦激素合并高浓度 MPA时 sub- Gl增加趋势也较单独使用 MPA为高, 其 P值为 0. 04, 在 G1及 G2/M期比单独加 MPA为低, S 期则为高。 由以上结果得知, 瘦激素合并 MPA 导致细胞凋亡比例比单独加 MPA为之明 显, 另外其作用可能系增加 MPA抑制 G1及 G2/M期, 或是使细胞周期停滞在 S期。 Please refer to FIG. 6A and FIG. 6B, which are respectively a schematic diagram showing the effect of cell cycle on cell treatment after treatment with leukemia combined with MPA, and the cell cycle analysis of the present invention after treating cells with leptin and MPA. As shown in the figure: To further understand the role of leptin and MPA in liver cancer cells, this example is to collect cells treated with estrogen or MPA and estrogen combined with MPA for 24 hours, by flow cytometry (Flow Cytometry) Analyzes the effect of the drug on the Cell Cycle and counts the portion of Sub-Gl/Apoptosis in the cell cycle as Student's - test. The experimental results showed that the cells treated with MPA had an increasing trend in sub-G1, indicating a marked increase in apoptotic cells, but decreased in the G1 and G2/M phases, but increased in the S phase; After treatment with high concentration of lean hormone and high concentration of MPA for 24 hours, the analysis showed that the increase of sub-G1 was higher than that of MPA alone, and its P value was 0.002; while the low concentration of lean hormone combined with high concentration of MPA Sub- The increase trend of Gl is also higher than that of MPA alone, and its P value is 0.04, which is lower in G1 and G2/M than in MPA alone, and higher in S phase. From the above results, it is known that the ratio of leukemia combined with MPA leads to apoptosis compared with MPA alone. In addition, the effect may be to increase MPA inhibition of G1 and G2/M phases, or to arrest the cell cycle in S phase.
实施例 6: 细胞凋亡相关蛋白质的分析  Example 6: Analysis of apoptosis-related proteins
请参阅图 7所示, 系本发明以瘦激素合并 MPA处理后对细胞凋亡相关蛋白质的影响示 意图。 如图所示: 既已证明瘦激素合并 MPA会使细胞凋亡增加, 本发明再利用西方墨点法 分析细胞以瘦激素合并 MPA处理 24小时之后, 其细胞凋亡相关蛋白质是否有所变化。 如图 所示, 以不同浓度瘦激素合并不同浓度 MPA 处理细胞 24 小时之后, 细胞蛋白质以 anti- c leaved caspase 3、 anti-cleaved caspase 7及 anti - cleaved PARP ( Po 1 y-ADP-r i bose- polymerase) 抗体进行西方墨点法分析, 由结果发现单独以 MPA 处理后的细胞在 cleaved caspase 3/7 与 cleaved PARP 都有明显地被活化, 而瘦激素合并 MPA造成之 cleaved caspase 3/7与 cleaved PARP活化效果都比单独使用 MPA更加明显。  Referring to Fig. 7, the effect of the present invention on apoptosis-related proteins after treatment with leptin and MPA is shown. As shown in the figure: It has been shown that the combination of leptin and MPA increases the apoptosis. The present invention uses Western blotting to analyze whether the apoptosis-related proteins are changed after treatment with estrogen and MPA for 24 hours. As shown in the figure, after treatment with different concentrations of estrogen and different concentrations of MPA for 24 hours, the cell proteins were anti-c leaved caspase 3, anti-cleaved caspase 7 and anti-cleaved PARP (Po 1 y-ADP-r i bose - Polymerase) The antibody was analyzed by Western blotting. It was found that cells treated with MPA alone were significantly activated in cleaved caspase 3/7 and cleaved PARP, and cleaved caspase 3/7 caused by leptin combined with MPA The cleaved PARP activation effect is more pronounced than with MPA alone.
实施例 7: 瘦激素受体 (ob- R) 与黄体酮受体 (PgR) 在加药后的相互结合作用 请参阅图 8 A及图 8 B所示, 分别为本发明以免疫沉淀法分析瘦激素受体与黄体酮受 体接受器交互结合作用的示意图、 及本发明以免疫荧光染色分析瘦激素受体与黄体酮受体 接受器交互结合作用的示意图。 如图所示: 进一步探讨瘦激素受体及黄体酮受体之间是否 存在某些交互作用, 本实施例中将细胞先单独以瘦激素或 MPA, 以及瘦激素合并 MPA处理 30分钟后, 收取细胞蛋白质以 anti-pgR抗体进行免疫沉淀法, 再用 anti-ob-R抗体以西 方墨点法分析瘦激素受体及黄体酮受体是否会因加药后而结合来共同作用, 结果发现在加 入 MPA 30分钟后, 瘦激素受体确实跟黄体酮受体有结合现象。 而瘦激素合并 MPA处理细胞 30分钟后, 瘦激素受体跟黄体酮受体结合现象更加地明显, 如图 8 A所示。 在免疫荧光染 色也同样得到相同之结果, 以药物处理细胞 10分钟后, 经 FITC染黄体酮受体、 Texas- Red 染瘦激素受体、 以及 phalloidin 染细胞膜, 在荧光显微镜 40X 下观察, 当单独加入 MPA 时, 会有少许之瘦激素受体会跟黄体酮受体结合。 在加入瘦激素和 MPA后, 此时结合会更 加地明显, 如图 8 B所示。 故经实验得知, 瘦激素受体与黄体酮受体确实会互相作用进而 对细胞产生影响。  Example 7: The interaction between the leptin receptor (ob-R) and the progesterone receptor (PgR) after dosing is shown in Fig. 8A and Fig. 8B, respectively, which are analyzed by immunoprecipitation according to the present invention. Schematic diagram of the interaction of the leptin receptor with the progesterone receptor receptor, and the present invention by immunofluorescence staining for the interaction of the leptin receptor with the progesterone receptor receptor. As shown in the figure: Further explore whether there is some interaction between the leptin receptor and the progesterone receptor. In this example, the cells are treated with leptin or MPA alone, and lean hormone combined with MPA for 30 minutes. The cell protein was immunoprecipitated with anti-pgR antibody, and the anti-ob-R antibody was used to analyze whether the leptin receptor and the progesterone receptor were combined by the Western blotting method. After 30 minutes of MPA addition, the leptin receptor does bind to the progesterone receptor. After 30 minutes of treatment with estrogen and MPA, the leptin receptor binding to the progesterone receptor was more pronounced, as shown in Figure 8A. The same result was obtained by immunofluorescence staining. After treating the cells with the drug for 10 minutes, FITC stained progesterone receptor, Texas-Red stained leptin receptor, and phalloidin stained cell membrane, observed under fluorescence microscope 40X, when alone When MPA is added, a small amount of the estrogen receptor will bind to the progesterone receptor. After the addition of lean hormone and MPA, the binding is more pronounced at this point, as shown in Figure 8B. Therefore, it has been experimentally found that the leptin receptor and the progesterone receptor do interact to affect cells.
实施例 8: 降低瘦激素受体的表现减少瘦激素合并 MPA后的效果  Example 8: Decreasing the expression of the estrogen receptor to reduce the effect of leptin combined with MPA
请参阅图 9所示, 系本发明抑制瘦激素受体减少加强 MPA作用的分析示意图。 如图所 示- 瘦激素受体及黄体酮受体会因为加药后而互相结合来共同作用, 在本实施例中继续探 讨既然瘦激素会增加 MPA抑制细胞效果, 而瘦激素结合至相对接受器来启动作用, 如果抑 制瘦激素受体的表现是否瘦激素就会因而失去加强 MPA抑制细胞之作用, 而不会去增加 MPA之治疗效果。 如图所示, 将瘦激素受体 siRNA以 20nM转殖至细胞培养一天, 第二天换 上无血清 (Serum- Free ) 之培养基, 再合并瘦激素及 MPA处理细胞 24 小时之后, 以 XTT assay 分析细胞生长之影响。 经实验结果中得知, 当瘦激素受体之 siRNA 降低瘦激素受体 之表现量时, 瘦激素确实无法去加强 MPA抑制细胞之作用, 由此可知瘦激素会加强 MPA之 效果主要系经由瘦激素及其接受器来调控此功能。 Please refer to FIG. 9 for a schematic diagram of the analysis of the present invention for inhibiting the reduction of the estrogen receptor to enhance the action of MPA. As shown in the figure - the leptin receptor and progesterone receptors will interact with each other after dosing. In this example, we continue to explore that since leptin increases MPA inhibition of cell effects, and estrogen binding to relative acceptance In order to initiate the action, if the inhibition of the expression of the estrogen receptor is negative, the hormone will lose the effect of strengthening the MPA inhibiting cells, and will not increase the therapeutic effect of MPA. As shown in the figure, the leptin receptor siRNA was transferred to the cell culture day at 20 nM, and the next day was changed. The serum-free (Serum-Free) medium was treated with leptin and MPA for 24 hours, and the effect of cell growth was analyzed by XTT assay. According to the experimental results, when the TNF of the leptin receptor reduces the expression of the estrogen receptor, the leptin can not strengthen the effect of MPA inhibiting cells. It can be seen that the effect of leptin on strengthening MPA is mainly through thin Hormones and their receptors regulate this function.
实施例 9: 增加瘦激素受体及黄体酮受体的表现量使细胞在加药后的凋亡增加 请参阅图 1 0 A及图 1 0 B所示, 分别为本发明增加长型瘦激素受体的表现量使细胞 在加药后的凋亡分析示意图、 及本发明增加黄体酮受体的表现量使细胞在加药后的凋亡分 析示意图。 如图所示: 由以上结果发现瘦激素加强 MPA 的治疗效果确实经由瘦激素受体活 化来作用。 因此, 为了再证实这项研究, 本实施例将 ob- Rb plasmid 转殖至细胞培养一 天, 第二天换上 serura- free 的培养基, 再合并瘦激素及 MPA处理细胞 24 小时之后, 以 XTT assay 分析细胞生长影响, 藉由将细胞大量表现长型瘦激素受体 (ob- Rb) 以观察细胞 在加药后的影响。 结果如图 1 0 A所示, 当细胞大量表现长型瘦激素受体时, 确实对于提 高 MPA 治疗效果有明显增加现象。 另外, 长型瘦激素受体似乎也会增加黄体酮受体的表 现。 有鉴于此, 本实施例另外再 PgR plasmid 转殖至细胞培养一天, 第二天换上 serum - free培养基, 再合并瘦激素及 MPA处理细胞 24小时之后, 以 XTT assay分析细胞生长的 影响, 藉由将细胞大量表现黄体酮受体以观察是否会加强 MPA作用的效果。 结果如图 1 0 B所示, 同样符合本发明预期, 当细胞大量表现黄体酮受体时, 确实也增强 MPA 的治疗效 果。 在本实施例中发现瘦激素合并 MPA作用, 系由于瘦激素受体间接地增加黄体酮受体的 表现, 进而提高 MPA治疗效果。  Example 9: Increasing the expression levels of the leptin receptor and the progesterone receptor, and increasing the apoptosis of the cells after dosing, as shown in Fig. 10A and Fig. 10B, respectively, adding long-type lean hormones to the present invention. The expression of the receptor allows the cells to be analyzed for apoptosis after dosing, and the present invention increases the expression of the progesterone receptor to visualize the apoptosis of the cells after dosing. As shown in the figure: From the above results, it was found that the therapeutic effect of leptin-enhancing MPA did indeed act through the activation of the estrogen receptor. Therefore, in order to reconfirm this study, in this example, ob-Rb plasmid was transferred to cell culture for one day, the next day was replaced with serura-free medium, and then combined with estrogen and MPA for 24 hours, XTT The assay analyzes the effects of cell growth by observing the effects of the cells after dosing by expressing the cells in large amounts of the long-term leptin receptor (ob-Rb). As a result, as shown in Fig. 10A, when the cells express a large amount of long-term leptin receptors, the effect of improving MPA treatment is markedly increased. In addition, long-type leptin receptors appear to increase the performance of progesterone receptors. In view of this, in this example, PgR plasmid was further transferred to cell culture for one day, and the next day was replaced with serum-free medium, and then treated with leptin and MPA for 24 hours, and the effect of cell growth was analyzed by XTT assay. The effect of enhancing the action of MPA is observed by expressing the cells in large amounts of progesterone receptors. The results are shown in Fig. 10B, which is also in line with the expectation of the present invention, and indeed, when the cells express a large amount of progesterone receptors, the therapeutic effect of MPA is also enhanced. In this example, it was found that the action of leptin combined with MPA is due to the indirect increase of progesterone receptor expression by the leptin receptor, thereby improving the therapeutic effect of MPA.
实施例 10: 瘦激素加强 MPA抑制 JAK/STAT讯息传递路径  Example 10: Lean Hormone Enhances MPA Inhibition JAK/STAT Message Delivery Path
请参阅图 1 1所示, 系本发明 JAK/STAT路径以药物处理后表现量的变化示意图。 如图 所示: 为找出瘦激素与 MPA合并作用后启动下游哪些相关蛋白质的变化, 本实施例利用西 方墨点法分析肝癌细胞以瘦激素合并 MPA处理 24 小时之后其下游相关蛋白质是否有所改 变。 经实验结果得知细胞以药物处理 24小时之后, 皆抑制了 p- ob- R、 ob-R与 PgR, 同时 也发现瘦激素合并 MPA去处理细胞后, 抑制之效果也比单独处理 MPA较为明显, 此外, 在 JAK2/STAT3相关路径之蛋白质方面, 结果也系抑制的。 因此, 从结果得知 MPA会去抑制黄 体酮受体及下游之相关蛋白质, 使其失去原有功能, 且瘦激素合并 MPA处理后的细胞, 其 抑制效果更加明显。  Please refer to FIG. 1 1 , which is a schematic diagram showing changes in the JAK/STAT path of the present invention after drug treatment. As shown in the figure: In order to find out which related proteins are changed after the combination of leptin and MPA, this example uses western blot method to analyze whether the downstream related proteins are treated by the combination of leptin and MPA for 24 hours. change. According to the experimental results, it was found that p-ob-R, ob-R and PgR were inhibited after 24 hours of treatment with the drug. It was also found that the effect of inhibition by lean hormone combined with MPA was more obvious than that of MPA alone. In addition, in terms of proteins in the JAK2/STAT3 related pathway, the results were also suppressed. Therefore, it was found that MPA inhibits the progesterone receptor and related proteins downstream, so that it loses its original function, and the inhibitory effect of the cells after treatment with leptin and MPA is more obvious.
实施例 11: 瘦激素加强 MPA抑制 MAPK讯息传递路径  Example 11: Leptin Enhances MPA Inhibition MAPK Message Delivery Path
请参阅图 1 2所示, 系本发明 MAPK路径以药物处理后表现量的变化示意图。 如图所 示: 在瘦激素下游蛋白质 JAK2/STAT3 皆被抑制之后, 本实施例试图观察 MAPK相关讯息路 径及上下游蛋白质是否也有所变化, 在相同条件下, 以西方墨点法分析蛋白质表现。 经实 验结果得知 APK相关蛋白质 p- JNK、 JNK、 p-ERKl/2, ERKl/2、 p- p38及 p38之表现量都系 被抑制的, 证明 MPA 不仅会去抑制瘦激素受体下游相关蛋白, 也同样会去抑制其它讯息路 径来抑制肝癌细胞生长, 其瘦激素也更加提升 MPA抑制作用。 Please refer to FIG. 12 for a schematic diagram showing the change of the MAPK pathway of the present invention after the drug treatment. As shown in the figure: After the downstream hormone protein JAK2/STAT3 was inhibited, this example attempts to observe whether the MAPK-related message pathway and the upstream and downstream proteins also change, and the protein expression is analyzed by Western blotting under the same conditions. Real The results showed that the expression levels of the APK-related proteins p-JNK, JNK, p-ERKl/2, ERKl/2, p-p38 and p38 were all inhibited, demonstrating that MPA not only inhibits the downstream proteins associated with the leptin receptor. It also inhibits other message pathways to inhibit the growth of liver cancer cells, and its estrogen also enhances MPA inhibition.
实施例 12: 以短时间点观察瘦激素合并 MPA对讯息蛋白分子的变化  Example 12: Observation of changes in estrogen molecules by MPA on a short time point
请参阅图 1 3 A及图 1 3 B所示, 分别为本发明 JAK/STAT路径以药物处理短时间内表 现量的变化示意图、 及本发明 APK路径以药物处理短时间内表现量的变化示意图。 如图所 示- 由以上实验结果显示在 24小时以药物处理过的细胞 JAK2/STAT3及 MAPK相关讯号路径 的蛋白质都被抑制, 可能原因系因为在 24小时之时间点细胞皆处于凋亡状态造成相关蛋白 质降解, 并看不出系哪一条相关讯息路径起变化, 于是本实施例利用短时间点来观察相关 蛋白质变化。 将细胞处理高浓度之瘦激素与高浓度之 MPA及高浓度之瘦激素合并高浓度之 MPA, 分别处理 30分钟、 3小时及 6小时。 依照时间点来收集细胞 JAK/STAT相关蛋白质以 西方墨点法来分析蛋白质之表现量。 结果显示, 在第 30分钟瘦激素合并 MPA就己经开始降 解 p-ob-R, 且随着时间之增加降解之程度就越明显。 而 P-STAT3 (Tyr705)在第 3小时也 幵始有被抑制之现象, 抑制之效果也系瘦激素合并 MPA 比单独使用还要明显, 如图 1 3 A 所示。 另外, 在相同条件下以西方墨点法观察 MAPK上、 下游相关蛋白质表现量的变化, p - JN 在第 3小时有明显地被抑制下来之趋势, 而 p- ERK1/2及 PIAS3在第 30分钟及第 3小 时的时候, MPA与瘦激素合并 MPA有过度表现的现象, 如图 1 3 B所示。  Please refer to FIG. 1 3 A and FIG. 1 3 B , which are schematic diagrams showing changes in the JAK/STAT path in the short-term performance of the JAK/STAT path in the present invention, and changes in the performance of the APK path in the short-term period of the present invention. . As shown in the figure - the results of the above experiments showed that the proteins in the JAK2/STAT3 and MAPK-related signal pathways of the drug-treated cells were inhibited in 24 hours, possibly because the cells were in apoptotic state at 24 hours. Related protein degradation does not reveal which of the relevant message pathways changes, so this example uses short time points to observe changes in related proteins. The cells were treated with high concentrations of estrogen and high concentrations of MPA and high concentrations of estrogen combined with high concentrations of MPA for 30 minutes, 3 hours and 6 hours, respectively. Collecting cells according to time points JAK/STAT-related proteins are analyzed by Western blotting methods for protein expression. The results showed that in the 30th minute, the combination of lean hormone and MPA began to degrade p-ob-R, and the degree of degradation became more obvious with time. However, P-STAT3 (Tyr705) was also inhibited at the third hour, and the effect of inhibition was also more obvious than that of leptin combined with MPA, as shown in Fig. 13 A. In addition, under the same conditions, Western blotting method was used to observe the changes in the expression of MAPK and downstream related proteins. p-JN was significantly inhibited at the 3rd hour, while p-ERK1/2 and PIAS3 were at 30th. At the minute and the third hour, MPA and the estrogen combined with MPA showed excessive performance, as shown in Figure 13B.
实施例 13: p-ERKl/2诱导 PIAS3活化抑制 P-STAT3  Example 13: p-ERKl/2 induction PIAS3 activation inhibition P-STAT3
请参阅图 1 4所示, 系本发明以抑制 ERK1/2的活化及 PIAS3的表现量减少 p- STAT3的 降解示意图。 如图所示: 上述发现 p- ERK1/2在第 30分钟及第 3小时单独使用 MPA与瘦激 素合并 MPA有过度表现之现象, 本实施例试图找出相关路径以厘清 p-ERKl/2 活化与 p - STAT3 (Tyr705) 抑制之相关性。 结果得知 p-ERKl/2活化诱使 P-STAT3 之抑制者-活化态 转录讯息传递活化子的抑制蛋白 3 (Protein inhibitor of activated STAT3, PIAS3) 活 化而抑制 P-STAT3 (Tyr705 ) 之表现使细胞凋亡 (如图 1 3 B所示) 。 于是本实施例将细 胞在加药前 4小时先加入 10 p-ERKl/2 inhibitor (U0126) 后, 再加入瘦激素及 MPA处 理细胞 3 个小时, 收取细胞蛋白质以西方墨点法分析其相关蛋白质的表现量。 经实验结果 发现抑制了 p- ERK1/2之磷酸化, 且 PIAS3也没有被诱使活化, 其 p- STAT3 (Tyr705) 之表 现量亦没有被抑制下来。 由以上结果得知, MPA 也许系透过活化 p- ERK1/2 而间接活化 PIAS3使抑制 p- STAT3 (Tyr705) 促使细胞凋亡, 其中瘦激素加强了 MPA活化 p- ERK1/2之 作用使细胞凋亡增加。  Referring to Figure 14, the present invention is intended to inhibit the activation of ERK1/2 and the expression of PIAS3 to reduce the degradation of p-STAT3. As shown in the figure: The above findings showed that p-ERK1/2 alone was overexpressed with MPA and lean hormone combined with MPA at the 30th minute and the third hour. This example attempts to find a relevant path to clarify p-ERKl/2 activation. Correlation with p-STAT3 (Tyr705) inhibition. As a result, it was found that p-ERKl/2 activation induces the activation of P-STAT3 inhibitor-activated transcriptional activator 3 (PIAS3) and inhibits the expression of P-STAT3 (Tyr705). Apoptosis (as shown in Figure 1 3 B). Therefore, in this example, the cells were added with 10 p-ERKl/2 inhibitor (U0126) 4 hours before the dosing, and then the cells were treated with estrogen and MPA for 3 hours, and the cellular proteins were collected and analyzed by Western blotting. The amount of performance. It was found by experiments that the phosphorylation of p-ERK1/2 was inhibited, and PIAS3 was not induced to activate, and the expression of p-STAT3 (Tyr705) was not inhibited. From the above results, MPA may indirectly activate PIAS3 by activating p-ERK1/2 to inhibit apoptosis of p-STAT3 (Tyr705), which enhances the action of MPA to activate p-ERK1/2. Increased apoptosis.
实施例 14: 细胞大量表现瘦激素加强 MPA抑制细胞生长的活性 请参阅图 1 5〜图 1 6 B所示, 分别为本发明于细胞内大量表现瘦激素加强 MPA抑制 细胞生长的活性分析示意图、 本发明于细胞内大量表现瘦激素后以 MPA 处理对于 STAT3相 关讯息路径的分析示意图、 及本发明于细胞内大量表现瘦激素后以 MPA 处理对于 MAPK相 关讯息路径的分析示意图。 如图所示: 以上述实施例, 瘦激素合并 MPA抑制肝癌细胞其瘦 激素属于外加的实验方式, 然而本发明于此实施例中, 为测试当肝癌细胞本身可分泌大量 瘦激素时, 是否也会提升 MPA抑制肝癌细胞作用。 于是将瘦激素质体 (pcLeptin ) 转殖至 细胞培养一天, 第二天换上 serum-free的培养基, 再以 10—4M MPA处理细胞 24小时之后, 以 XTT assay 分析细胞生长之影响。 如图 1 5左半边所示, 藉细胞短暂性转殖瘦激素基因 使细胞自体能分泌大量瘦激素, 再加入 MPA 24小时之后发现, 细胞本身分泌瘦激素后同样 会加强 MPA 抑制肝癌细胞作用。 此外, 将瘦激素基因大量表现后, 收集其培养基利用瘦激 素抗体进行免疫沉淀法, 接着进行西方墨点法分析结果, 如图 1 5右半边所示, 瘦激素基 因大量表现在细胞中时, 会使细胞自体分泌出瘦激素, 并可在培养基中测得。 再者, 本实 施例亦收取细胞蛋白质以西方墨点法分析 STAT及 MAPK相关讯息路径的蛋白质表现量, 如 图 1 6 A所示, 将瘦激素质体转殖至细胞培养一天, 第二天换上 serum- free的培养基, 再 以 Ιθ ΐ MPA处理细胞 24小时之后, JAK/STAT相关蛋白质以西方墨点法分析结果, 结果发 现当细胞大量表现瘦激素时再加入 MPA 24小时之后, 抑制 STAT3活化之程度也系比单独处 理 MPA之细胞还要明显, 且在相同条件下以西方墨点法分析细胞 MAPK讯息路径相关蛋白 质, 如图 1 6 B所示, 在 MAPK相关讯息蛋白质 ERK、 JNK、 p38、 c-fos及 c- jun也系相同 的结果。 Example 14: Cell mass expression of leptin enhances MPA activity in inhibiting cell growth Please refer to FIG. 15 to FIG. 1 6B, which are respectively a schematic diagram showing the activity analysis of a large amount of estrogen-enhanced MPA inhibiting cell growth in the cells of the present invention, and the present invention is related to STAT3 by MPA treatment after a large amount of expression of leukemia in the cells. A schematic diagram of the analysis of the message path, and a schematic diagram of the analysis of the MAPK-related message pathway by MPA treatment after the present invention expresses a large amount of lean hormone in the cell. As shown in the figure: In the above examples, the combination of lean hormone and MPA inhibits the hematopoietic cells of the liver cancer cells, and the present invention is in this embodiment. However, in the present embodiment, in order to test whether the liver cancer cells themselves can secrete a large amount of lean hormones, Will enhance the role of MPA in inhibiting liver cancer cells. So after the laser quality thin body (pcLeptin) colonization transferred to the cell culture one day, the next day medium was replaced with serum-free, and then processed to 10- 4 M MPA for 24 hours, XTT assay Analysis of the cell growth. As shown in the left half of Figure 15, the cell's transient translocation of the estrogen gene allows the cells to secrete a large amount of estrogen. After adding the MPA for 24 hours, it is found that the secretion of the estrogen by the cells itself also enhances the inhibition of liver cancer cells by MPA. In addition, after a large amount of expression of the leptin gene, the medium was collected and subjected to immunoprecipitation using a leptin antibody, followed by Western blot analysis, as shown in the right half of Fig. 15, when the leptin gene was expressed in a large amount in the cell. , will cause the cells to secrete leptin from the body and can be measured in the medium. Furthermore, in this example, the protein expression of the STAT and MAPK-related message pathways is also analyzed by Western blotting. As shown in FIG. 16A, the pro-liposome is transferred to the cell culture for one day, the next day. After replacing the serum-free medium and treating the cells with Ιθ ΐ MPA for 24 hours, the JAK/STAT-related protein was analyzed by Western blotting. It was found that when the cells expressed a large amount of lean hormone, MPA was added for 24 hours. The degree of STAT3 activation was also more pronounced than that of cells treated with MPA alone, and the cellular MAPK message pathway-related proteins were analyzed by Western blotting under the same conditions, as shown in Figure 16 B, in the MAPK-related protein ERK, JNK. , p38, c-fos and c-jun are also the same result.
实施例 15: 瘦激素合并 MPA对正常肝脏上皮细胞株的影响  Example 15: Effect of leptin combined with MPA on normal liver epithelial cell lines
请参阅图 1 7〜图 1 8 B所示, 分别为本发明以瘦激素合并 MPA对正常肝脏上皮细胞 株生长的分析示意图、 本发明正常肝脏细胞以瘦激素合并 MPA处理后对 STAT3相关讯息路 径的分析示意图、 及本发明正常肝脏细胞以瘦激素合并 MPA处理后对 MAPK相关讯息路径的 分析示意图。 如图所示: 本实施例以相同条件下将瘦激素合并 MPA处理正常肝脏上皮细胞 株 THLE- 3经 24及 48小时之后, 藉由 XTT assay分析瘦激素合并 MPA后对细胞生长 (即存 活率) 的影响。 如图 1 7所示, 由实验结果显示, 单独处理瘦激素及 MPA或瘦激素合并 MPA对于正常肝脏上皮细胞株的存活率及毒杀性皆没有显著影响。 再者, 如图 1 8 A及图 1 8 B所示, 本实施例亦于相同条件下分析 STAT及 MAPK相关讯息路径的蛋白质表现量, 结果显示瘦激素及 MPA对于相关讯息蛋白质也都没有显著影响。  Please refer to FIG. 1 7 to FIG. 1 8B, which are respectively a schematic diagram of the analysis of the growth of normal liver epithelial cell lines by the combination of lean hormone and MPA, and the STAT3 related message path of the normal liver cells of the present invention treated with lean hormone combined with MPA. The analysis diagram of the analysis, and the analysis of the MAPK-related message pathway after the treatment of the normal liver cells of the present invention with the combination of lean hormone and MPA. As shown in the figure: In this example, after the same conditions were used to treat the normal liver epithelial cell line THLE-3 with MPA for 24 and 48 hours, the cell growth (ie, survival rate) was analyzed by XTT assay after estrogen combined with MPA. ) Impact. As shown in Figure 17, the results of the experiment showed that treatment of leptin and MPA or leptin alone with MPA had no significant effect on the survival rate and toxicity of normal liver epithelial cell lines. Furthermore, as shown in Fig. 18A and Fig. 18B, this example also analyzes the protein expression of STAT and MAPK-related message pathways under the same conditions, and the results show that both leptin and MPA are not significant for related protein. influences.
藉此, 经上述各实验证实, 以瘦激素合并 MPA处理肝癌细胞之后, 确实可强化 MPA抑 制肝癌细胞的效果, 并且比单独使用 MPA更佳; 此外, 本发明将肝癌细胞株大量表现瘦激 素蛋白质时, 亦发现相同效果。 据此, 未来可望将本发明构想运用在临床肝癌病人的治疗 上。 例如: 本发明可以检测肝癌病患血清中或肝癌组织中瘦激素的表现量; 于一较佳实施 例中, 对于表现量较高的病患, 可考虑直接给予 MPA治疗, 进而达到强化 MPA抑制肝肿瘤 的作用; 于另一较佳实施例中, 本发明亦可将瘦激素及 MPA合并设计成一药物治疗肝癌病 患, 藉此可扩大药物的使用范围并强化 MPA 的药理作用; 于再一较佳实施例中, 本发明另 可以如 5-羟色氨酸 (5- hydroxy- trypton, 5- HTP ) 提高瘦激素表现量较低之病患之瘦激素 量后, 再给予 MPA 治疗。 因此, 本发明可帮助肝癌病患维持更长久的存活时间。 另外, 在 本发明以相同条件处理正常肝细胞株时, 亦发现瘦激素及 MPA并不会对正常肝细胞株产生 影响, 显示此药物或疗程并不会对病患产生不良副作用, 可相较于已知的化学治疗及放射 治疗对于正常人体的伤害, 提供给肝癌病患一个新的契机。 Therefore, it has been confirmed by the above experiments that the treatment of liver cancer cells with the combination of lean hormone and MPA can indeed enhance the effect of MPA inhibiting liver cancer cells, and is better than using MPA alone; in addition, the present invention displays liver cancer cell lines in large amounts to express hormone proteins. The same effect was also found. Accordingly, it is expected that the present invention will be applied to the treatment of clinical liver cancer patients in the future. On. For example, the present invention can detect the expression amount of leukemia in the serum of liver cancer patients or liver cancer tissues; in a preferred embodiment, for patients with higher performance, direct treatment with MPA can be considered, thereby achieving enhanced MPA inhibition. The effect of liver tumors; in another preferred embodiment, the present invention can also combine leptin and MPA to design a drug for treating liver cancer patients, thereby expanding the scope of use of the drug and enhancing the pharmacological action of MPA; In a preferred embodiment, the present invention may further be administered to MPA after increasing the amount of estrogen in a patient with a low expression of estrogen, such as 5-hydroxy-trypton (5-HTP). Therefore, the present invention can help liver cancer patients maintain a longer survival time. In addition, when the normal liver cell strain is treated under the same conditions in the present invention, it is also found that the estrogen and MPA do not affect the normal liver cell line, indicating that the drug or the course of treatment does not cause adverse side effects to the patient, The known chemotherapeutic and radiotherapy treatments for normal human injuries provide a new opportunity for liver cancer patients.
综上所述, 本发明系一种以瘦激素治疗肝癌的应用, 可有效改善己用的种种缺点, 系 包含对肝癌病患投予医药有效量的一治疗药物、 疗程或筛选平台, 可将瘦激素或提高肝癌 病患瘦激素生理浓度之合并一醋酸甲羟孕酮后与肝癌细胞接触, 以该瘦激素透过其受体与 黄体酮受体之交互作用加强及加速该 MPA毒杀肝癌细胞, 藉以对于正常肝脏上皮细胞株与 癌细胞株具有专一性之药理作用, 俾供可对正常肝脏细胞无不良副作用同时, 并能延长肝 癌病患之整体存活率者, 进而使本发明之产生能更进步、 更实用、 更符合使用者所须, 确 已符合发明专利申请要件, 爱依法提出专利申请。  In summary, the present invention is a method for treating liver cancer by using a thin hormone, which can effectively improve various disadvantages, and includes a therapeutic drug, a course of treatment or a screening platform for administering a therapeutically effective amount to a liver cancer patient. Lean hormone or increase the physiological concentration of leptin in patients with liver cancer combined with medroxyprogesterone acetate and contact with liver cancer cells, and the interaction of the leptin with its progesterone receptor enhances and accelerates the MPA poisoning of liver cancer The cell has a specific pharmacological effect on the normal liver epithelial cell line and the cancer cell line, and the donor can have no adverse side effects on the normal liver cell, and can prolong the overall survival rate of the liver cancer patient, thereby further making the present invention The production can be more progressive, more practical, and more in line with the needs of the user. It has indeed met the requirements for the invention patent application, and loves to file a patent application according to law.

Claims

权 利 要 求 书 Claim
1 . 一种以瘦激素治疗肝癌的应用, 系包含对肝癌病患投予医药有效量的一治疗药 物、 疗程或筛选平台, 其特征系将瘦激素或提高肝癌病患瘦激素生理浓度之合并一醋酸甲 羟孕酮 (MPA) 后与肝癌细胞接触, 以该瘦激素透过其受体与黄体酮受体的交互作用加强及 加速该 MPA毒杀肝癌细胞, 藉以对于正常肝脏上皮细胞株与癌细胞株具有专一性的药理作 用, 以供对正常肝脏细胞无毒杀的不良副作用产生同时, 并能延长肝癌病患的整体存活 率; 以及  1 . A method for treating liver cancer by using a thin hormone, comprising a therapeutic drug, a course of treatment or a screening platform for administering a medicinal effective amount to a liver cancer patient, characterized in that the combination of the lean hormone or the physiological concentration of the lean hormone of the liver cancer patient is combined. After medroxyprogesterone acetate (MPA) is contacted with liver cancer cells, the interaction of the leptin with its progesterone receptor through the receptor enhances and accelerates the MPA to kill liver cancer cells, thereby allowing for normal liver epithelial cell lines. The cancer cell strain has a specific pharmacological effect for the simultaneous side effects of non-toxic killing of normal liver cells, and can prolong the overall survival rate of liver cancer patients;
其中, 该瘦激素的表现量高低更进一步作为肝癌病患对 MPA治疗有无疗效的预测因子 (Predictive Factors ) 及其存活时间的预后因子 (Prognostic Factors) 。  Among them, the expression level of the estrogen further as a predictor of the efficacy of the treatment of MPA in patients with liver cancer (Predictive Factors) and its survival factor (Prognostic Factors).
2 . 根据权利要求 1所述的以瘦激素治疗肝癌的应用, 其特征在于, 该 MPA为一细胞 毒性剂。  The use of leukemia for treating liver cancer according to claim 1, wherein the MPA is a cytotoxic agent.
3 . 根据权利要求 1所述的以瘦激素治疗肝癌的应用, 其特征在于, 该药物造成肝癌 细胞尺寸或肝癌细胞质量的缩小。  The use of the leukemia for treating liver cancer according to claim 1, characterized in that the drug causes a reduction in the size of the liver cancer cells or the quality of the liver cancer cells.
4 . 根据权利要求 1所述的以瘦激素治疗肝癌的应用, 其特征在于, 可作为在肝癌细 胞株上及临床肝癌病患的统计数据。  The use of leukemia for treating liver cancer according to claim 1, which is useful as statistical data on liver cancer cell lines and clinical liver cancer patients.
5 . 根据权利要求 1所述的以瘦激素治疗肝癌的应用, 其特征在于, 该肝癌病患血清 中或肝癌组织中瘦激素的表现量较高者, 系直接给予 MPA 治疗肝癌病患, 以利达到强化 MPA的药理作用。  The use of the leukemia for treating liver cancer according to claim 1, wherein the liver cancer patient has a higher amount of expression of leukemia in the serum or liver cancer tissue, and the patient is directly treated with MPA to treat the liver cancer patient, To achieve the pharmacological effects of strengthening MPA.
6 . 根据权利要求 1所述的以瘦激素治疗肝癌的应用, 其特征在于, 该肝癌病患血清 中或肝癌组织中瘦激素的表现量较高者, 系将瘦激素及 MPA合并成一药物治疗肝癌病患, 以利达到强化 MPA的药理作用。  The use of the method for treating hepatocellular carcinoma with a leptin according to claim 1, wherein the liver cancer patient has a higher expression of estrogen in the serum or the liver cancer tissue, and the leukemia and the MPA are combined into one drug treatment. For patients with liver cancer, in order to achieve the pharmacological effects of strengthening MPA.
7 . 根据权利要求 1所述的以瘦激素治疗肝癌的应用, 其特征在于, 该肝癌病患血清 中或肝癌组织中瘦激素的表现量较低者, 系以 5-羟色氨酸 (5-hydroxy-trypton, 5-HTP) 提高瘦激素量后, 再给予 MPA治疗, 以利达到强化 MPA的药理作用。  The use of the method for treating liver cancer with a leptin according to claim 1, wherein the liver cancer patient has a lower expression level of lean hormone in the serum or liver cancer tissue, and is 5-hydroxytryptophan (5). -hydroxy-trypton, 5-HTP) After increasing the amount of estrogen, it is given MPA treatment to enhance the pharmacological effects of MPA.
8 . 根据权利要求 1所述的以瘦激素治疗肝癌的应用, 其特征在于, 该瘦激素的表现 量越高, 其强化 MPA对肝癌细胞的抑制效果越明显。  The use of the leukemia for treating liver cancer according to claim 1, characterized in that the higher the expression of the estrogen, the more effective the effect of enhancing MPA on hepatoma cells is.
PCT/CN2010/001662 2010-01-19 2010-10-22 Use of leptin in enhancing therapeutic efficacy of medroxyprogesterone acetate on liver cancer WO2011088605A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US12/689,718 US20110098219A1 (en) 2009-10-22 2010-01-19 System for Curing Hepatic Cancer with Leptin
US12/689,718 2010-01-19

Publications (1)

Publication Number Publication Date
WO2011088605A1 true WO2011088605A1 (en) 2011-07-28

Family

ID=44307687

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2010/001662 WO2011088605A1 (en) 2010-01-19 2010-10-22 Use of leptin in enhancing therapeutic efficacy of medroxyprogesterone acetate on liver cancer

Country Status (1)

Country Link
WO (1) WO2011088605A1 (en)

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE CA 2007, Database accession no. 147:163443 *
HOU ZHENJIANG ET AL.: "Correlation of leptin level in blood serum of hepatocellular carcinoma patients with nutritional state and prognosis.", ZHONGGUO LAONIANXUE ZAZHI (CHINESE)., vol. 28, no. 16, August 2008 (2008-08-01), pages 1602 - 1604 *
XU XIA ET AL.: "Interventional Therapy Combined with Medroxyprogesterone Acetate for Advanced Hepatic Carcinoma.", ZHONGLIUXUE ZAZHI (CHINESE), vol. 8, no. 6, 2002, pages 367 *
YAN GUANGTAO.: "Leptin may be a key target in treatment of tumors.", J CHINESE PLA POSTGRAD MED SCH (CHINESE), vol. 31, no. 1, January 2010 (2010-01-01), pages 1 - 3 *

Similar Documents

Publication Publication Date Title
Yu et al. Dexrazoxane ameliorates doxorubicin-induced cardiotoxicity by inhibiting both apoptosis and necroptosis in cardiomyocytes
Nooshinfar et al. Melatonin, an inhibitory agent in breast cancer
Li et al. Epigenetic reactivation of estrogen receptor-α (ERα) by genistein enhances hormonal therapy sensitivity in ERα-negative breast cancer
Liu et al. FoxO3 reverses 5-fluorouracil resistance in human colorectal cancer cells by inhibiting the Nrf2/TR1 signaling pathway
Chen et al. Shikonin inhibits triple-negative breast cancer-cell metastasis by reversing the epithelial-to-mesenchymal transition via glycogen synthase kinase 3β-regulated suppression of β-catenin signaling
Chai et al. Coptisine from Rhizoma coptidis exerts an anti-cancer effect on hepatocellular carcinoma by up-regulating miR-122
Han et al. Piperine (PP) enhanced mitomycin-C (MMC) therapy of human cervical cancer through suppressing Bcl-2 signaling pathway via inactivating STAT3/NF-κB
Chen et al. Baicalein resensitizes tamoxifen‐resistant breast cancer cells by reducing aerobic glycolysis and reversing mitochondrial dysfunction via inhibition of hypoxia‐inducible factor‐1α
Chi et al. Chemotherapy resistance and metastasis-promoting effects of thyroid hormone in hepatocarcinoma cells are mediated by suppression of FoxO1 and Bim pathway
Huang et al. Astragalus polysaccharide induces the apoptosis of human hepatocellular carcinoma cells by decreasing the expression of Notch1
Zhuang et al. Pien Tze Huang inhibits tumor cell proliferation and promotes apoptosis via suppressing the STAT3 pathway in a colorectal cancer mouse model
Forte et al. Obesity, diabetes, the cardiorenal syndrome, and risk for cancer
Wang et al. Anti-inflammatory effect of miltirone on inflammatory bowel disease via TLR4/NF-κB/IQGAP2 signaling pathway
Marzioni et al. An oestrogen receptor β-selective agonist exerts anti-neoplastic effects in experimental intrahepatic cholangiocarcinoma
Lewis-Wambi et al. Buthionine sulfoximine sensitizes antihormone-resistant human breast cancer cells to estrogen-induced apoptosis
EP2391358A1 (en) Therapeutic treatment of cancer and dysplasia of the cervix or vagina using estrogen antagonists
Zhao et al. Oral administration of Prunella vulgaris L improves the effect of taxane on preventing the progression of breast cancer and reduces its side effects
Shi et al. Synuclein γ Stimulates Membrane-Initiated Estrogen Signaling by Chaperoning Estrogen Receptor (ER)-α36, a Variant of ER-α
Kim et al. Rosiglitazone promotes tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis by reactive oxygen species-mediated up-regulation of death receptor 5 and down-regulation of c-FLIP
Xu et al. The effect of JAK2 knockout on inhibition of liver tumor growth by inducing apoptosis, autophagy and anti-proliferation via STATs and PI3K/AKT signaling pathways
Zhang et al. Britannin stabilizes T cell activity and inhibits proliferation and angiogenesis by targeting PD-L1 via abrogation of the crosstalk between Myc and HIF-1α in cancer
Hu et al. The disulfiram/copper complex induces autophagic cell death in colorectal cancer by targeting ULK1
Li et al. Impact of Taurine on the proliferation and apoptosis of human cervical carcinoma cells and its mechanism
Li et al. Liriodenine induces the apoptosis of human laryngocarcinoma cells via the upregulation of p53 expression
Serini et al. Role of β-catenin signaling in the anti-invasive effect of the omega-3 fatty acid DHA in human melanoma cells

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10843648

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10843648

Country of ref document: EP

Kind code of ref document: A1