WO2021194276A1 - Pharmaceutical composition, for prevention or treatment of cancer, comprising 13-hydroxyoctadecadienoic acid as active ingredient - Google Patents

Pharmaceutical composition, for prevention or treatment of cancer, comprising 13-hydroxyoctadecadienoic acid as active ingredient Download PDF

Info

Publication number
WO2021194276A1
WO2021194276A1 PCT/KR2021/003708 KR2021003708W WO2021194276A1 WO 2021194276 A1 WO2021194276 A1 WO 2021194276A1 KR 2021003708 W KR2021003708 W KR 2021003708W WO 2021194276 A1 WO2021194276 A1 WO 2021194276A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
hode
cells
tumor
pharmaceutical composition
Prior art date
Application number
PCT/KR2021/003708
Other languages
French (fr)
Korean (ko)
Inventor
김세윤
변영주
박승주
Original Assignee
한국과학기술원
고려대학교 세종산학협력단
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 한국과학기술원, 고려대학교 세종산학협력단 filed Critical 한국과학기술원
Publication of WO2021194276A1 publication Critical patent/WO2021194276A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/201Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids having one or two double bonds, e.g. oleic, linoleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Definitions

  • the present invention was made by the project number 2015061639 under the support of the Ministry of Science, ICT and Future Planning. Development of source technology and establishment of hepatocyte-based toxicity and drug efficacy evaluation system”, hosted by the Korea Advanced Institute of Science and Technology, and the research period is from October 1, 2015 to September 30, 2016.
  • the present invention relates to a pharmaceutical composition for preventing or treating cancer comprising 13-HODE (13-Hydroxyoctadecadienoic acid) as an active ingredient.
  • 13-HODE 13-Hydroxyoctadecadienoic acid
  • Cancer is the most common and serious disease that threatens human health, and research and development of anticancer drugs is important for prolonging the lifespan of cancer patients.
  • cancer treatment methods have made a lot of progress due to the rapid development of oncogenomics and molecular pharmacology and the development of new anticancer drugs, but there is still a need for the development of new therapeutic agents.
  • Endometrial cancer is one of the most common cancers in women. Although the incidence rate of endometrial cancer varies by region, it ranks fourth in the United States after breast cancer, lung cancer, and skin cancer. In 2015, US statistics showed 54,870 new cases and 10,170 deaths. In Korea, 20,859 patients occurred in 2015, and endometrial cancer is the 7th most common cancer among all female cancer patients, and the incidence rate is increasing.
  • Endometrial cancer mainly occurs during menopause in women and is known to occur more in developed countries than in developing countries.
  • endometrial cancer is highly correlated with obesity, and 81% of patients with a BMI of 30 or higher are diagnosed with endometrial cancer.
  • the incidence rate among younger women has been increasing over the past 10 years.
  • the treatment method for endometrial cancer may vary depending on the disease stage, tumor size, age and general condition of the patient, and whether or not a child is desired, but surgery is mostly performed as the primary treatment. Additional treatment methods may include radiation therapy or chemotherapy. Endometrial cancer is treated with hormone therapy with progesterone or chemotherapy without surgery. Chemotherapy uses doxorubicin and cisplatin, which leads to hematologic toxicity and side effects such as hair loss, dizziness, and vomiting. For recurrent or advanced endometrial cancer, taxanes and platinum-containing drugs (TC therapy) are administered adjuvantly.
  • TC therapy platinum-containing drugs
  • mTOR (mechanistic target of rapamycin) protein is a serine/threonine kinase and is a key signaling factor that regulates cell growth, cell cycle, protein synthesis and glucose metabolism.
  • PI3K, Akt emtors and higher-level factors
  • Activation of emtor signals in cancer is caused by mutations in the emtor gene itself, high-level oncogenes (PI3K, Akt) that enhance the activity of emtor, and mutations in tumor suppressor factors (eg PTEN, TSC1/2).
  • the present inventors made intensive research efforts to develop compounds for inhibiting cancer cell growth and killing cancer cells.
  • the present invention was completed by identifying that the composition containing 13-HODE (13-Hydroxyoctadecadienoic acid) as an active ingredient is effective in treating cancer.
  • an object of the present invention is to provide a pharmaceutical composition for preventing or treating cancer comprising 13-HODE (13-Hydroxyoctadecadienoic acid) as an active ingredient.
  • the present invention provides a pharmaceutical composition for preventing or treating cancer comprising 13-HODE (13-Hydroxyoctadecadienoic acid) as an active ingredient.
  • 13-HODE 13-Hydroxyoctadecadienoic acid
  • the 13-HODE corresponds to an mTOR signaling inhibitor.
  • the emtor signal transduction inhibitor refers to a substance that inhibits the activity of emtor itself or inhibits the activity of an upper-level factor that increases the activity of emtor.
  • Targets whose activity is inhibited by the emtor signaling inhibitor may include, but are not limited to, emtor, PI3K, Akt, S6K, and S6.
  • the 13-HODE is 13-HODE, a pharmaceutically acceptable salt thereof, or an optical isomer thereof.
  • the cancer is a solid cancer.
  • solid cancer has characteristics distinguishing it from blood cancer, and includes bladder, breast, intestine, kidney, lung, liver, brain, esophagus, gallbladder, ovary, pancreas, stomach, cervix, thyroid, prostate and skin. It is a cancer consisting of a mass caused by abnormal cell growth in various solid organs such as
  • the solid cancer is brain tumor, benign astrocytoma, malignant astrocytoma, pituitary adenoma, meningioma, brain lymphoma, oligodendroglioma, ependymoma, brainstem tumor, head and neck tumor, laryngeal cancer, oropharyngeal cancer, nasal/sinus cancer , nasopharyngeal cancer, salivary gland cancer, hypopharyngeal cancer, thyroid cancer, oral cancer, chest tumor, small cell lung cancer, non-small cell lung cancer, thymus cancer, mediastinal tumor, esophageal cancer, breast cancer, male breast cancer, abdominal tumor, stomach cancer, liver cancer, gallbladder cancer, biliary tract cancer, Pancreatic cancer, small intestine cancer, colorectal cancer, rectal cancer, anal cancer, bladder cancer, kidney cancer, male genital tumor, penile cancer, prostate cancer, female genital tumor, cervical cancer,
  • the brain tumor is selected from the group consisting of meningioma, pituitary tumor, hemangioblastoma, epidermoid tumor, glioma cyst, glioma, oligodendrocyte glioma, glioblastoma, and metastatic brain tumor.
  • the cancer is a hematologic cancer.
  • hematologic cancer refers to cancer occurring in components constituting blood, and refers to malignant tumors occurring in blood, hematopoietic organs, lymph nodes, lymphatic organs, and the like.
  • the hematologic cancer is acute myeloid leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, acute monocytic leukemia, multiple myeloma, Hodgkin's lymphoma, and non-Hodgkin's lymphoma. is selected from, but not limited thereto.
  • the pharmaceutical composition of the present invention may include a pharmaceutically acceptable carrier in addition to the composition as an active ingredient.
  • Pharmaceutically acceptable carriers included in the pharmaceutical composition of the present invention are commonly used in formulation, and include lactose, dextrose, sucrose, sorbitol, mannitol, starch, acacia gum, calcium phosphate, alginate, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, methyl cellulose, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil, and the like. it's not going to be
  • the pharmaceutical composition of the present invention may further include a lubricant, a wetting agent, a sweetening agent, a flavoring agent, an emulsifying agent, a suspending agent, a preservative, and the like, in addition to the above components.
  • a lubricant e.g., a talc, a kaolin, a kaolin, a kaolin, a kaolin, kaolin, kaolin, kaolin, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, a talct, a talct, a talct, a stevia, glycerin, glycerin, glycerin,
  • the pharmaceutical composition of the present invention may be administered orally or parenterally, for example, intrathecal administration, intravenous administration, subcutaneous administration, intradermal administration, intramuscular administration, intraperitoneal administration, intrasternal administration, intratumoral administration, intranasal administration , intracerebral administration, intracranial administration, intrapulmonary administration, rectal administration, etc., but is not limited thereto.
  • a suitable dosage of the pharmaceutical composition of the present invention varies depending on factors such as formulation method, administration mode, age, weight, sex, pathological condition, food, administration time, administration route, excretion rate and response sensitivity of the patient, An ordinarily skilled physician can readily determine and prescribe an effective dosage (a pharmaceutically effective amount) for the desired treatment or prophylaxis.
  • the daily dose of the pharmaceutical composition of the present invention is 0.0001-100 mg/kg.
  • the term “pharmaceutically effective amount” refers to an amount sufficient to prevent or treat the above-mentioned diseases.
  • prevention refers to the prevention or protective treatment of a disease or disease state.
  • treatment refers to reduction, suppression, sedation or eradication of a disease state.
  • the pharmaceutical composition of the present invention is prepared in unit dosage form by formulating using a pharmaceutically acceptable carrier and/or excipient according to a method that can be easily carried out by a person of ordinary skill in the art to which the present invention pertains. or may be prepared by incorporation into a multi-dose container.
  • the dosage form may be prepared in various ways such as oral medicine, injection, etc., in the form of a solution, suspension or emulsion in oil or aqueous medium, or in the form of an extract, powder, suppository, powder, granule, tablet or capsule, dispersant or stable Additional topics may be included.
  • the pharmaceutical composition comprising 13-HODE of the present invention has an effect of inhibiting the emtor signaling process.
  • the pharmaceutical composition of the present invention has the effect of inhibiting the growth of cancer cells and the effect of inhibiting the growth of tumors in vivo. Therefore, the pharmaceutical composition of the present invention has an anticancer effect on cancer, more specifically endometrial cancer.
  • the present invention provides a method for preventing or treating cancer comprising administering to a subject a pharmaceutical composition comprising the above-described 13-HODE of the present invention as an active ingredient. .
  • administer refers to directly administering a therapeutically or prophylactically effective amount of a composition of the present invention to a subject (individual) suffering from, or likely to suffer from, the subject disease. It means that the same amount is formed in the body of
  • the "therapeutically effective amount” of the composition means an amount of the composition sufficient to provide a therapeutic or prophylactic effect to a subject to which the composition is administered, and includes a “prophylactically effective amount”.
  • the term "subject (subject)” is a mammal, including humans, mice, rats, guinea pigs, dogs, cats, horses, cattle, pigs, monkeys, chimpanzees, baboons and rhesus monkeys. . Most specifically, the subject of the present invention is a human.
  • the method for preventing or treating cancer of the present invention includes administering the pharmaceutical composition according to an aspect of the present invention, the description thereof is omitted to avoid excessive redundancy of the present specification for overlapping content. .
  • the present invention provides a pharmaceutical composition for preventing or treating cancer comprising 13-HODE (13-Hydroxyoctadecadienoic acid) as an active ingredient.
  • 13-HODE 13-Hydroxyoctadecadienoic acid
  • composition of the present invention can be used as an anticancer agent by inhibiting the growth of cancer cells and exhibiting the efficacy of killing cancer cells.
  • Figure 3a shows the results of analyzing the molecular position of the emtor protein to which 13-HODE binds through hydrogen-deuterium substitution mass spectrometry.
  • Figure 3b shows the catalytic cleft of the emtor protein, which is the molecular site at which 13-HODE binds.
  • Figure 4a shows the results of analyzing proteins related to emtor signaling in cells after 13-HODE was treated with respect to JHUEM7 cells through immunoprecipitation.
  • Figure 4b shows the results of analyzing proteins related to emtor signaling in cells after 13-HODE was treated with respect to JHUEM7 cells through immunoprecipitation.
  • Figure 4c shows the results of analysis of proteins related to emtor signaling in cells after 13-HODE was treated with respect to JHUEM7 cells through immunoprecipitation.
  • Figure 5a shows the results of analyzing proteins related to emtor signaling in cells after 13-HODE was treated with respect to JHUEM7 cells through western blotting.
  • Figure 5b shows the results of analyzing proteins related to emtor signaling in HCT116, SW480, HT29, MDA-MB-468, and U-373 MG cells after treatment with 13-HODE through Western blotting.
  • 6a shows the results of analyzing the viability of JHUEM7 cells after treatment with 13-HODE.
  • Figure 6b shows the results of analyzing the viability of MDA-MB-468 cells after treatment with 13-HODE.
  • 7a shows the results of analyzing the degree of cancer cell colony proliferation after treatment with 13-HODE for JHUEM7 cells.
  • Figure 7b shows the results of analyzing the degree of cancer cell colony proliferation after 13-HODE treatment with respect to MDA-MB-468 cells.
  • Figure 8a shows the results of analyzing the size change of the tumor after treatment with 13-HODE JHUEM7 cells xenografted into mice.
  • Figure 8b shows the results of observing the size change of the tumor after treatment with 13-HODE JHUEM7 cells xenografted into mice.
  • Figure 10a shows the results of analyzing the size change of the tumor after treatment with 13-HODE of MDA-MB-468 cells xenografted into mice.
  • Figure 10b shows the results of observing the size change of the tumor after treatment with 13-HODE of MDA-MB-468 cells xenografted into mice.
  • 11a shows the results of analyzing significant genes using RNA-seq analysis after treating JHUEM7 cells with DMSO control drug and 150 ⁇ M 13-HODE for 48 hours, respectively.
  • 11b shows the results of performing pathway enrichment analysis using RNA-seq analysis after treating JHUEM7 cells with DMSO control drug and 150 ⁇ M 13-HODE for 48 hours, respectively.
  • 11c shows the results of performing pathway enrichment analysis using RNA-seq analysis after treating JHUEM7 cells with DMSO control drug and 150 ⁇ M 13-HODE for 48 hours, respectively.
  • FIG. 13 shows the results of analyzing proteins related to emtor signaling in JHUEM7 cells overexpressing ALOX15 through western blotting.
  • 15A shows the results of analyzing proteins related to emtor signaling in MCF7 cells overexpressing ALOX15 through Western blotting.
  • 15B shows the results of analyzing the degree of colony proliferation of MCF7 cells overexpressing ALOX15.
  • Figure 16a shows the results of analyzing proteins related to emtor signaling in cells after 13-HODE was treated with respect to HEC59 cells through western blotting.
  • Figure 16b shows the results of analyzing the viability of HEC59 cells after treatment with 13-HODE.
  • Figure 16c shows the results of analyzing the degree of cancer cell colony proliferation after 13-HODE treatment on HEC59 cells.
  • % used to indicate the concentration of a specific substance is (weight/weight) % for solid/solid, (weight/volume) % for solid/liquid, and Liquid/liquid is (volume/volume) %.
  • HEK293, HEK293T, MDA-MB-468, MCF7, HEC59 and U373MG cells were cultured in high-glucose DMEM medium supplemented with 2 mM glutamine, 100 ⁇ g/ml penicillin-streptomycin and 10% fetal bovine serum (FBS, Atlas Biological). cultured. JHUEM7 cells were cultured in DMEM/F12 (Thermo Fisher Scientific) supplemented with 1X non-essential amino acids (Sigma Aldrich, Missouri, USA) and 10% FBS.
  • SW480 and HT29 cells were cultured in RPMI1640 medium supplemented with 2 mM glutamine, 100 ⁇ g/ml penicillin-streptomycin and 10% fetal bovine serum (FBS, Atlas Biological). All cell lines were cultured at 37 °C, 5% CO 2 conditions.
  • HEK293 and HEK293T cells were purchased from ATCC (American Type Culture Collection, Virginia, USA), and JHUEM7 cells were purchased from RIKEN (Saitama, Japan).
  • JHUEM7 cells were lysed with 1x CHAPS lysis buffer.
  • the composition of the CHAPS buffer is 50 mM HEPES, 150 mM NaCl, 0.4% CHAPS, 50 mM NaF, 10 mM Na-pyrophosphate, 100 mM ⁇ -glycerophosphate at pH 7.4, and a protease inhibitor.
  • Antibodies used in the Western blotting were anti-mTOR antibody (Cell Signaling Technology #2972, Massachusetts, USA) and anti-raptor antibody (Bethyl Laboratories A300-553A, Alabama, USA).
  • Biotin-labeled 13-HODE was bound to emtor and raptor (regulatory protein that forms a protein complex with emtor). However, biotin-labeled 13-HODE showed a low binding affinity to emtor and raptor in the presence of unlabeled 13-HODE ( FIG. 2 ).
  • Protein sequence coverage maps were obtained using undeuterated samples as follows: 3.5 ⁇ L of ⁇ 50 ⁇ M Emtor C-terminal fragment, 25 mM HEPES, pH 7.4, 50 mM KCl, 10 mM MgCl 2 , ice-cold 10% glycerol diluted with 96.5 ⁇ L quench solution (100 mM glycine, 25 mM TCEP, pH 2.5).
  • the sample was subjected to Waters HDX nanoAcquity UPLC (Waters, Milford, Mass.) and subjected to in-line pepsin treatment (Waters Enzymate BEH pepsin 40 column). Peptide fragments were separated through an Acquitty UPLC BEH C18 peptide trap and column. After separation, the peptides were eluted directly with a Waters Synapt G2 mass spectrometer (Waters, Milford, MA) using a gradient of 5% to 35% acetonitrile (0.1% formic acid). MS E data were obtained via 20 to 30 V lamp trap CE. Peptides were analyzed using Waters' ProteinLynx Global Server 2.5.1 (PLGS).
  • PLGS Waters' ProteinLynx Global Server 2.5.1
  • D 2 0 was added to the sample to initiate a deuterium substitution reaction.
  • the deuterium substitution reaction was performed under various conditions (10 seconds, 1 minute, 10 minutes and 2 hours).
  • % D was used to calculate the percent deuterated plot ( ⁇ % D) . Confidence intervals for the ⁇ % D plot were determined and adjusted for percent deuterated using a fully deuterated control.
  • Example 4 Treatment and analysis of 13-HODE on Emtor kinase isolated through immunoprecipitation
  • HEK293T cells were washed once with ice-cold PBS and lysed with ice-cold lysis buffer.
  • the composition of the lysis buffer is HEPES 40 mM, 120 mM NaCl, 2 mM EDTA, 10 mM pyrophosphate, 10 mM sodium fluoride, 0.3% CHAPS, IX protease inhibitor cocktail at pH 7.4.
  • the cell lysate was incubated at 4° C. for 10 minutes and centrifuged at 13,000 rpm for 10 minutes to collect the supernatant.
  • 2 ⁇ g of anti-mTOR antibody (Cell Signaling Technology #2972, Massachusetts, USA) was added to 2 mg of the supernatant and spin-incubated at 4° C. for 1.5 hours.
  • 20 ⁇ L of agarose beads (Pierce) was added and incubated for an additional hour.
  • Emtor immunoprecipitates were washed twice with the lysis buffer and twice with kinase wash buffer.
  • the composition of the kinase wash buffer is 25 mM HEPES, 20 mM potassium chloride, pH 7.4.
  • the kinase assay was performed at 37° C. for 15 minutes using 15 ⁇ L of mTOR Complex 1 kinase buffer and 150 ng of S6K1 substrate. The reaction was stopped by adding 10 ⁇ L sample buffer, heated for 5 minutes, and then analyzed by Western blotting.
  • Antibodies used in the Western blotting were anti-T389 p-S6K antibody (Cell Signaling Technology #9205, Massachusetts, USA), anti-S473 p-Akt antibody (Cell Signaling Technology #9271), and anti-S6K antibody (Cell Signaling Technology).
  • Example 5 Emtor kinase analysis of 13-HODE-treated cells by Western blotting
  • Example 5-1 Emtor kinase analysis of 13-HODE-treated JHUEM7 cells by Western blotting
  • 35 mm cell culture dishes were seeded with JHUEM7 3 ⁇ 10 5 cells.
  • the next day 68 or 135 uM of 13-HODE or 10 nM Torin1 was mixed with the cell culture medium and treated, and then 48 hours later, emtor signaling was analyzed by western blotting.
  • the 13-HODE or Torin1 mixed culture medium was freshly replaced every 24 hours.
  • the cells were lysed with NP-40 lysis buffer.
  • the composition of NP-40 is 50 mM Tris-HCl at pH 7.5, 150 mM NaCl, 1% NP-40 substitute, 1 mM EDTA at pH 8.0, 50 mM NaF, 10 mM Na-Pi lophosphate (Na-pyrophosphate), 15 mM Na 3 VO 4 , 100 mM ⁇ -glycerophosphate, and a protease inhibitor.
  • the whole cell lysate was mixed with 5X SDS sample buffer to adjust the final concentration to 1-2 ⁇ g/ ⁇ L and then heated for 5 minutes. Then, Western blotting was performed with the heated cell lysate.
  • Antibodies used in the Western blotting were anti-T389 p-S6K antibody (Cell Signaling Technology #9205, Massachusetts, USA), anti-S240/244 p-S6 antibody (Cell Signaling Technology #5364), anti-T37/46 p-4EBP1 antibody (Cell Signaling Technology #9459), anti-S473 p-Akt antibody (Cell Signaling Technology #9271), anti-T308 p-Akt antibody (Cell Signaling Technology #4056), anti-T638/641 p-PKCa Antibody (Cell Signaling Technology #9375), Anti-S6K Antibody (Cell Signaling Technology #9202), Anti-S6 Antibody (Cell Signaling Technology #2217), Anti-4EBP1 Antibody (Cell Signaling Technology #9452), Anti-Akt Antibody ( Cell Signaling Technology #9272), anti-PKCa antibody (Cell Signaling Technology #2056).
  • Example 5-2 Emtor kinase analysis of 13-HODE-treated HCT003, SW480, HT29, MDA-MB-468 and U-373MG cells by Western blotting
  • the experimental method was the same as the method described in Example 5-1, except that HCT003, SW480, HT29, MDA-MB-468, and U-373MG cells were used instead of JHUEM7 cells.
  • JHUEM7 cells or MDA-MB-468 cells (1 ⁇ 10 4 cells/mL) were seeded in 96 well plates. After overnight incubation, cells were treated with DMSO or 135 ⁇ M of 13-HODE for various times. Then, 20 ⁇ L of MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) solution (2.5 mg/mL in PBS) was added to each well, Then, incubated at 37 °C for an additional 1 hour. The OD (optical density) value of each well was measured at 492 nm with a SpectraMax Paradigm Reader (Molecular Devices).
  • MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
  • cells (4 x 10 4 cells/well) were seeded in 24 well plates and cultured overnight, and cells were cultured with DMSO or 135 ⁇ M of 13-HODE for 4 days. The resulting colonies were then fixed with methanol at -20 °C for 30 min and stained with 1% crystal violet for 15 min.
  • Example 7 Colony proliferation analysis of 13-HODE-treated cells
  • JHUEM7 cells or MDA-MB-468 cells (4 x 10 4 cells/well) were seeded in a 24-well plate and cultured overnight, and the cells were cultured with DMSO or 135 ⁇ M of 13-HODE for 4 days. The resulting colonies were then fixed with methanol at -20 °C for 30 min and stained with 1% crystal violet for 15 min.
  • Example 8-1 The anticancer effect of 13-HODE confirmed through the experiment of nude mice implanted with JHUEM7 cells
  • the size of the tumor was examined after treatment with 13-HODE in the tumor-transplanted mice.
  • mice were implanted by subcutaneous injection into 8-12 week old female BALB/c nude mice. After the mean tumor volume reached 50 mm 3 , mice were randomly assigned to 3 different groups (6 mice/group). The mouse weight and tumor diameter were measured once every two days. Tumor volume using a caliper was evaluated according to the following formula 0.5x (width) 2 x (Length ), a Student's T test was used to determine the P-values.
  • 13-HODE treatment mice received intratumoral or intravenous injection of 13-HODE at 10 mg/kg every 2 days after initiation of the experiment.
  • Example 8-2 The anticancer effect of 13-HODE confirmed through an experiment in nude mice implanted with MDA-MB-468 cells
  • the size of the tumor was examined after treatment with 13-HODE in the tumor-transplanted mice.
  • the experimental method is the same as that described in Example 8-1, except that MDA-MB-468 cells were used instead of JHUEM7 cells.
  • DEG analysis is an analysis that selects a candidate group for a significant gene (Differentially Expressed Genes) with a difference in expression between the control group and the control group by measuring and statistically processing the expression value of the gene.
  • 13-HODE treatment group 3729 genes were decreased and 3842 genes were increased ( FIG. 11a ).
  • cancer-related regulatory pathways such as liver cancer, pancreatic cancer, and breast cancer were detected ( FIG. 11b ), and cell cycle, aging, and fatty acid metabolism-related mechanistic pathways were also detected ( FIG. 11c ).
  • Example 10 Viability of cells treated with 13-HODE and 13-HODE measured inside cells
  • the amount of 13-HODE inside the 13-HODE-treated JHUEM7 cells was quantitatively analyzed.
  • the amount of 13-HODE inside the cell was measured as follows.
  • 13-HODE was extracted from ⁇ 1.4 mL cell supernatant using solid phase extraction (SPE).
  • SPE solid phase extraction
  • a 60 mg Oasis HLB (Waters) SPE cartridge was washed and pre-conditioned sequentially with ethyl acetate, methanol and (0.1 % acetic acid + 5 % methanol) aqueous solution.
  • 10 ⁇ L of 0.2 mg/mL EDTA and BHT with MeOH:H 2 O (50:50) as solvents were added to the adsorbent bed of the SPE column.
  • An equal volume (0.1% acetic acid + 5% MeOH) aqueous solution was added to the cell supernatant.
  • 13-HODE was determined using an LC-MS/MS system equipped with an Agilent 1290 HPLC (Agilent) and QTRAP 5500 mass spectrometry (AB Sciex).
  • a reversed-phase column (Pursuit5 C18, 150 ⁇ 2.1 mm) was used with mobile phase A (0.1 % aqueous acetic acid solution) and mobile phase B (0.1 % acetic acid in acetonitrile/methanol (84/16) solvent).
  • LC was run at 250 ⁇ L/min and 35°C. The separation gradient is as follows: 35% B at 0 min, 35% B hold for 0.25 min, 35% to 45% of B for 0.75 min, 45% hold of B for 2 min, 45% of B for 5.5 min.
  • MRM Multiple reaction monitoring
  • Example 11 Emtor kinase analysis of JHUEM7 cells overexpressing ALOX15 (human lipoxygenase15-1)
  • the experimental method is the same as the method described in Example 5, except that the cells used and 13-HODE were not treated.
  • As a control group cells not overexpressing ALOX15 were used.
  • Example 12 Anticancer effect of 13-HODE confirmed through an experiment in nude mice implanted with JHUEM7 cells overexpressing ALOX15 (human lipoxygenase15-1)
  • mice transplanted with ALOX15 overexpressing cells were transplanted with ALOX15 overexpressing cells.
  • the experimental method was the same as that described in Example 8, except that ALOX15 overexpressing JHEUM7 cells were used instead of JHUEM7 cells and 13-HODE was not treated.
  • ALOX15 overexpressing JHEUM7 cells were used instead of JHUEM7 cells and 13-HODE was not treated.
  • nude mice transplanted with cells not overexpressing ALOX15 were used.
  • Example 13 Emtor kinase of MCF7 cells overexpressing ALOX15 (human lipoxygenase15-1) and colony proliferation analysis of cells
  • Emtor kinase and cell colony proliferation assays were performed using MCF7 cells overexpressing ALOX15.
  • the experimental method is the same as that described in Examples 5 and 7, except that the cells used and 13-HODE were not treated.
  • MCF7 cells overexpressing wild-type ALOX15 were used, and as a control group and a comparison group, a control overexpressing cell incapable of enzyme expression and a mutant ALOX15 T560M cell in which enzyme activity was lost were used as the control group.
  • Example 14 Cellular Emtor kinase, Cell Viability, Cell Colony Proliferation Analysis Using HEC59 Cells
  • Emtor kinase, cell viability, and cell colony proliferation assays performed in Examples 5, 6, and 7 were performed using HEC59 cells.
  • HEC59 cells are cancer cells (endometrial cancer cells) and are cells with the emtor mutation (mTOR E1799K ).
  • the experimental method is the same as that described in Examples 5, 6, and 7 except for the cells used.
  • 13-HODE exerts an anticancer effect on various cancers such as colorectal cancer, breast cancer, glioblastoma, endometrial cancer, liver cancer, and pancreatic cancer.

Landscapes

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

Abstract

The present invention relates to a pharmaceutical composition, for the prevention or treatment of cancer, comprising 13-hydroxyoctadecadienoic acid (13-HODE) as an active ingredient. The composition of the present invention exhibits efficacy of inhibiting the growth of cancer cells and killing cancer cells, and thus can be used as an anticancer agent.

Description

13-하이드록시옥타데카디에노산을 유효성분으로 포함하는 암의 예방 또는 치료용 약제학적 조성물Pharmaceutical composition for preventing or treating cancer comprising 13-hydroxyoctadecadenoic acid as an active ingredient
본 발명은 미래창조과학부 지원 하에서 과제고유번호 2015061639에 의해 이루어진 것으로서, 상기 과제의 연구관리전문기관은 한국생명공학연구원, 연구사업명은 “원천기술개발사업”, 연구과제명은 “인간 줄기세포 유래 간세포분화 원천기술 개발과 간세포 기반 독성·약효 평가 시스템 확립”, 주관기관은 한국과학기술원, 연구기간은 2015.10.01 ~ 2016.09.30 이다. The present invention was made by the project number 2015061639 under the support of the Ministry of Science, ICT and Future Planning. Development of source technology and establishment of hepatocyte-based toxicity and drug efficacy evaluation system”, hosted by the Korea Advanced Institute of Science and Technology, and the research period is from October 1, 2015 to September 30, 2016.
본 특허출원은 2020년 3월 25일에 대한민국 특허청에 제출된 대한민국 특허출원 제10-2020-0036500호에 대하여 우선권을 주장하며, 상기 특허출원의 개시사항은 본 명세서에 참조로서 삽입된다. This patent application claims priority to Korean Patent Application No. 10-2020-0036500 filed with the Korean Intellectual Property Office on March 25, 2020, the disclosure of which is incorporated herein by reference.
본 발명은 13-HODE(13-Hydroxyoctadecadienoic acid)를 유효성분으로 포함하는 암의 예방 또는 치료용 약제학적 조성물에 관한 것이다.The present invention relates to a pharmaceutical composition for preventing or treating cancer comprising 13-HODE (13-Hydroxyoctadecadienoic acid) as an active ingredient.
암은 인간의 건강을 위협하는 가장 흔하고 심각한 질병이며, 항암제의 연구와 개발은 암환자 수명의 연장을 위해 중요하다. 최근 몇 년 동안 암 치료방법은 암유전체학과 분자약학의 급속한 발전과 새로운 항암제 개발로 많은 발전을 이루었지만, 여전히 새로운 치료제의 개발이 필요하다.Cancer is the most common and serious disease that threatens human health, and research and development of anticancer drugs is important for prolonging the lifespan of cancer patients. In recent years, cancer treatment methods have made a lot of progress due to the rapid development of oncogenomics and molecular pharmacology and the development of new anticancer drugs, but there is still a need for the development of new therapeutic agents.
자궁내막암은 여성에서 발생율이 높은 암 중 하나이다. 자궁내막암의 발생율은 지역에 따라서 다르나 미국에서는 유방암 폐암 피부암에 이어서 4위를 차지하고 있다. 2015년 미국 통계에서 54,870명의 새로운 환자가 발생하였으며, 10,170명의 환자가 사망하였다. 우리나라에서는 2015년 20,859명의 환자가 발생하여, 자궁내막암은 전체 여성 암환자 중 7번째로 많이 발생하는 암이며, 그 발생율은 점점 증가하고 있는 추세이다. Endometrial cancer is one of the most common cancers in women. Although the incidence rate of endometrial cancer varies by region, it ranks fourth in the United States after breast cancer, lung cancer, and skin cancer. In 2015, US statistics showed 54,870 new cases and 10,170 deaths. In Korea, 20,859 patients occurred in 2015, and endometrial cancer is the 7th most common cancer among all female cancer patients, and the incidence rate is increasing.
자궁내막암의 사망률은 20년전에 비하여 2 배로 증가하였으며, 10년 전에 비하여 약 8 % 증가하였다. 자궁내막암은 주로 여성의 폐경기에 발생하고 개발도상국보다는 선진국에서 더 많이 발생하는 것으로 알려져 있다. 특히 자궁내막암은 비만과의 상관성이 높아 BMI 30 이상인 환자 중 81 %가 자궁내막암으로 진단된다. 또한, 최근 10년동안 보다 젊은 여성들에서의 발병율이 높아지고 있는 추세이다.The mortality rate of endometrial cancer has doubled compared to 20 years ago, and it has increased by about 8% compared to 10 years ago. Endometrial cancer mainly occurs during menopause in women and is known to occur more in developed countries than in developing countries. In particular, endometrial cancer is highly correlated with obesity, and 81% of patients with a BMI of 30 or higher are diagnosed with endometrial cancer. In addition, the incidence rate among younger women has been increasing over the past 10 years.
자궁내막암의 치료방법은 질병의 진행 단계, 종양의 크기, 환자의 나이 및 전신상태, 아이를 원하는지 여부 등에 따라 달라질 수 있지만 대부분 일차적인 치료로서 수술을 시행한다. 추가적인 치료방법으로는 방사선 치료나 항암화학요법 등이 진행될 수 있다. 자궁내막암의 치료는 프로게스테론 제재로 호르몬 요법을 시행하거나 수술을 하지 않고 항암화학요법을 병행하기도 한다. 화학치료는 독소루비신과 시스플라틴이 사용되며, 그로인해 혈액학적 독성반응과 탈모, 어지러움증, 구토와 같은 부작용이 따른다. 재발성 또는 진행된 자궁내막암은 탁산(taxanes) 및 백금 함유 약물(TC therapy)가 보조적으로 투여된다.The treatment method for endometrial cancer may vary depending on the disease stage, tumor size, age and general condition of the patient, and whether or not a child is desired, but surgery is mostly performed as the primary treatment. Additional treatment methods may include radiation therapy or chemotherapy. Endometrial cancer is treated with hormone therapy with progesterone or chemotherapy without surgery. Chemotherapy uses doxorubicin and cisplatin, which leads to hematologic toxicity and side effects such as hair loss, dizziness, and vomiting. For recurrent or advanced endometrial cancer, taxanes and platinum-containing drugs (TC therapy) are administered adjuvantly.
엠토르 (mTOR, mechanistic target of rapamycin) 단백질은 세린/트레오닌 카이네이즈(kinase)로서 세포의 성장, 세포주기, 단백질 합성 및 포도당 대사등을 조절하는 핵심 신호전달인자이다. 특히 세포성장 신호조절의 핵심 단백질로서 30 %의 고형암세포에서 비정상적으로 활성이 증가되어 있으며, 암세포에서 이러한 엠토르 및 상위단계 인자들 (PI3K, Akt)이 가장 많이 변화되어 있는 것으로 알려져 있다. 암에서의 엠토르 신호의 활성화는 엠토르 유전자 자체의 돌연변이, 엠토르의 활성을 높이는 상위단계의 발암유전자 (PI3K, Akt) 및 종양억제인자 (예 PTEN, TSC1/2)의 돌연변이에 의해 발생한다. 따라서 엠토르 연관 신호전달의 억제는 단백질 합성저해, 지질 합성저해, 세포 성장 억제를 유발하여 세포사멸까지 초래할 수 있다. 따라서 암세포 성장의 억제와 암세포 사멸을 위한 엠토르 연관 신호전달 억제제의 개발의 필요성이 있다. mTOR (mechanistic target of rapamycin) protein is a serine/threonine kinase and is a key signaling factor that regulates cell growth, cell cycle, protein synthesis and glucose metabolism. In particular, as a key protein for regulating cell growth signaling, its activity is abnormally increased in 30% of solid cancer cells, and it is known that these emtors and higher-level factors (PI3K, Akt) are the most changed in cancer cells. Activation of emtor signals in cancer is caused by mutations in the emtor gene itself, high-level oncogenes (PI3K, Akt) that enhance the activity of emtor, and mutations in tumor suppressor factors (eg PTEN, TSC1/2). . Therefore, inhibition of emtor-associated signaling can lead to cell death by inhibiting protein synthesis, inhibiting lipid synthesis, and inhibiting cell growth. Therefore, there is a need for the development of an emtor-related signaling inhibitor for the inhibition of cancer cell growth and cancer cell death.
본 발명자들은 암세포 성장의 억제와 암세포 사멸을 위한 화합물을 개발하고자 예의 연구 노력하였다. 그 결과 13-HODE(13-Hydroxyoctadecadienoic acid)를 유효성분으로 포함하는 조성물의 경우 암 치료에 효과가 있다는 것을 규명함으로써, 본 발명을 완성하게 되었다. The present inventors made intensive research efforts to develop compounds for inhibiting cancer cell growth and killing cancer cells. As a result, the present invention was completed by identifying that the composition containing 13-HODE (13-Hydroxyoctadecadienoic acid) as an active ingredient is effective in treating cancer.
따라서, 본 발명의 목적은 13-HODE(13-Hydroxyoctadecadienoic acid)를 유효성분으로 포함하는 암의 예방 또는 치료용 약제학적 조성물을 제공하는 것이다. Accordingly, an object of the present invention is to provide a pharmaceutical composition for preventing or treating cancer comprising 13-HODE (13-Hydroxyoctadecadienoic acid) as an active ingredient.
본 발명의 일 양태에 따르면, 본 발명은 13-HODE(13-Hydroxyoctadecadienoic acid)를 유효성분으로 포함하는 암의 예방 또는 치료용 약제학적 조성물을 제공한다.According to one aspect of the present invention, the present invention provides a pharmaceutical composition for preventing or treating cancer comprising 13-HODE (13-Hydroxyoctadecadienoic acid) as an active ingredient.
상기 13-HODE는 엠토르(mTOR) 신호전달 억제제에 해당한다. The 13-HODE corresponds to an mTOR signaling inhibitor.
엠토르 신호전달 억제제는 엠토르 자체의 활성을 억제하거나 엠토르의 활성을 높이는 상위단계의 인자의 활성을 억제하는 물질을 의미한다. 상기 엠토르 신호전달 억제제에 의해 활성이 억제되는 표적은 엠토르, PI3K, Akt, S6K, S6 등이 있을 수 있으나 이에 한정되는 것은 아니다. The emtor signal transduction inhibitor refers to a substance that inhibits the activity of emtor itself or inhibits the activity of an upper-level factor that increases the activity of emtor. Targets whose activity is inhibited by the emtor signaling inhibitor may include, but are not limited to, emtor, PI3K, Akt, S6K, and S6.
본 발명의 일 구체예에 있어서, 상기 13-HODE는 13-HODE, 이의 약제학적으로 허용되는 염 또는 이의 광학이성질체이다. In one embodiment of the present invention, the 13-HODE is 13-HODE, a pharmaceutically acceptable salt thereof, or an optical isomer thereof.
본 발명의 일 구현예에 있어서, 상기 암은 고형암이다.In one embodiment of the present invention, the cancer is a solid cancer.
본 명세서 상의 용어 "고형암"이란 혈액암과는 구별되는 특징을 지니고, 방광, 유방, 장, 신장, 폐, 간, 뇌, 식도, 쓸개, 난소, 췌장, 위, 자궁경부, 갑상선, 전립선 및 피부 등의 여러 고형 장기(solid organ)에서 비정상적으로 세포가 성장하여 발생한 덩어리로 이루어진 암이다. As used herein, the term "solid cancer" has characteristics distinguishing it from blood cancer, and includes bladder, breast, intestine, kidney, lung, liver, brain, esophagus, gallbladder, ovary, pancreas, stomach, cervix, thyroid, prostate and skin. It is a cancer consisting of a mass caused by abnormal cell growth in various solid organs such as
본 발명의 일 구체예에 있어서, 상기 고형암은 뇌종양, 양성성상세포종, 악성성상세포종, 뇌하수체 선종, 뇌수막종, 뇌림프종, 핍지교종, 상의세포종, 뇌간종양, 두경부 종양, 후두암, 구인두암, 비강/부비동암, 비인두암, 침샘암, 하인두암, 갑상선암, 구강암, 흉부종양, 소세포성 폐암, 비소세포성 폐암, 흉선암, 종격동 종양, 식도암, 유방암, 남성유방암, 복부종양, 위암, 간암, 담낭암, 담도암, 췌장암, 소장암, 대장암, 직장암, 항문암, 방광암, 신장암, 남성 생식기종양, 음경암, 전립선암, 여성생식기종양, 자궁경부암, 자궁내막암, 난소암, 자궁육종, 질암, 여성외부생 식기암, 여성요도암 및 피부암으로 이루어지는 군으로부터 선택되는 것이나 이에 한정되는 것은 아니다. 보다 구체적으로 상기 고형암은 자궁내막암, 대장암, 교모세포종, 유방암, 간암, 췌장암이다. In one embodiment of the present invention, the solid cancer is brain tumor, benign astrocytoma, malignant astrocytoma, pituitary adenoma, meningioma, brain lymphoma, oligodendroglioma, ependymoma, brainstem tumor, head and neck tumor, laryngeal cancer, oropharyngeal cancer, nasal/sinus cancer , nasopharyngeal cancer, salivary gland cancer, hypopharyngeal cancer, thyroid cancer, oral cancer, chest tumor, small cell lung cancer, non-small cell lung cancer, thymus cancer, mediastinal tumor, esophageal cancer, breast cancer, male breast cancer, abdominal tumor, stomach cancer, liver cancer, gallbladder cancer, biliary tract cancer, Pancreatic cancer, small intestine cancer, colorectal cancer, rectal cancer, anal cancer, bladder cancer, kidney cancer, male genital tumor, penile cancer, prostate cancer, female genital tumor, cervical cancer, endometrial cancer, ovarian cancer, uterine sarcoma, vaginal cancer, exogenous female It is selected from the group consisting of gastric cancer, female urethral cancer, and skin cancer, but is not limited thereto. More specifically, the solid cancer is endometrial cancer, colorectal cancer, glioblastoma, breast cancer, liver cancer, and pancreatic cancer.
상기 뇌종양은 뇌수막종, 뇌하수체종양, 혈관모세포종, 유표피종, 교질낭종, 신경교종, 희돌기세포교종, 교모세포종, 전이성 뇌종양으로 이루어지는 군으로부터 선택되는 것이다. The brain tumor is selected from the group consisting of meningioma, pituitary tumor, hemangioblastoma, epidermoid tumor, glioma cyst, glioma, oligodendrocyte glioma, glioblastoma, and metastatic brain tumor.
본 발명의 일 구현예에 있어서, 상기 암은 혈액암이다. In one embodiment of the present invention, the cancer is a hematologic cancer.
본 명세서 상의 용어 "혈액암"이란 혈액을 구성하는 성분에 생긴 암을 지칭하는 것으로, 혈액, 조혈기관, 림프절, 림프기관 등에 발생한 악성 종양을 의미한다. As used herein, the term “hematologic cancer” refers to cancer occurring in components constituting blood, and refers to malignant tumors occurring in blood, hematopoietic organs, lymph nodes, lymphatic organs, and the like.
본 발명의 일 구체예에 있어서, 상기 혈액암은 급성골수구성백혈병, 급성림프구성 백혈병, 만성골수성백혈병, 만성림프구성백혈병, 급성단구성백혈병, 다발성 골수종, 호지킨림프종 및 비호지킨 림프종으로 이루어진 군으로부터 선택되는 것이나 이에 한정되는 것은 아니다. In one embodiment of the present invention, the hematologic cancer is acute myeloid leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, acute monocytic leukemia, multiple myeloma, Hodgkin's lymphoma, and non-Hodgkin's lymphoma. is selected from, but not limited thereto.
본 발명의 약제학적 조성물은 유효성분인 상기 조성물 외에 약제학적으로 허용되는 담체를 포함할 수 있다.The pharmaceutical composition of the present invention may include a pharmaceutically acceptable carrier in addition to the composition as an active ingredient.
본 발명의 약제학적 조성물에 포함되는 약제학적으로 허용되는 담체는 제제시에 통상적으로 이용되는 것으로서, 락토스, 덱스트로스, 수크로스, 솔비톨, 만니톨, 전분, 아카시아 고무, 인산 칼슘, 알기네이트, 젤라틴, 규산 칼슘, 미세결정성 셀룰로스, 폴리비닐피롤리돈, 셀룰로스, 물, 시럽, 메틸 셀룰로스, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 활석, 스테아르산 마그네슘 및 미네랄 오일 등을 포함하나, 이에 한정되는 것은 아니다. Pharmaceutically acceptable carriers included in the pharmaceutical composition of the present invention are commonly used in formulation, and include lactose, dextrose, sucrose, sorbitol, mannitol, starch, acacia gum, calcium phosphate, alginate, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, methyl cellulose, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil, and the like. it's not going to be
본 발명의 약제학적 조성물은 상기 성분들 이외에 윤활제, 습윤제, 감미제, 향미제, 유화제, 현탁제, 보존제 등을 추가로 포함할 수 있다. 적합한 약제학적으로 허용되는 담체 및 제제는 Remington's Pharmaceutical Sciences (19th ed., 1995)에 상세히 기재되어 있다.The pharmaceutical composition of the present invention may further include a lubricant, a wetting agent, a sweetening agent, a flavoring agent, an emulsifying agent, a suspending agent, a preservative, and the like, in addition to the above components. Suitable pharmaceutically acceptable carriers and agents are described in detail in Remington's Pharmaceutical Sciences (19th ed., 1995).
본 발명의 약제학적 조성물은 경구 또는 비경구로 투여할 수 있고, 예컨대 척추강 내 투여, 정맥내 투여, 피하 투여, 피내 투여, 근육내 투여, 복강내 투여, 흉골 내 투여, 종양 내 투여, 비내 투여, 뇌내 투여, 두개골 내 투여, 폐내 투여 및 직장내 투여 등으로 투여할 수 있으나 이에 한정되는 것은 아니다.The pharmaceutical composition of the present invention may be administered orally or parenterally, for example, intrathecal administration, intravenous administration, subcutaneous administration, intradermal administration, intramuscular administration, intraperitoneal administration, intrasternal administration, intratumoral administration, intranasal administration , intracerebral administration, intracranial administration, intrapulmonary administration, rectal administration, etc., but is not limited thereto.
본 발명의 약제학적 조성물의 적합한 투여량은 제제화 방법, 투여방식, 환자의 연령, 체중, 성, 병적 상태, 음식, 투여 시간, 투여 경로, 배설 속도 및 반응 감응성과 같은 요인들에 의해 다양하며, 보통으로 숙련된 의사는 소망하는 치료 또는 예방에 효과적인 투여량(약제학적 유효량)을 용이하게 결정 및 처방할 수 있다. 본 발명의 바람직한 구현예에 따르면, 본 발명의 약제학적 조성물의 1일 투여량은 0.0001-100 ㎎/㎏이다. A suitable dosage of the pharmaceutical composition of the present invention varies depending on factors such as formulation method, administration mode, age, weight, sex, pathological condition, food, administration time, administration route, excretion rate and response sensitivity of the patient, An ordinarily skilled physician can readily determine and prescribe an effective dosage (a pharmaceutically effective amount) for the desired treatment or prophylaxis. According to a preferred embodiment of the present invention, the daily dose of the pharmaceutical composition of the present invention is 0.0001-100 mg/kg.
본 명세서에서 용어 "약제학적 유효량"은 상술한 질환을 예방 또는 치료하는 데 충분한 양을 의미한다.As used herein, the term “pharmaceutically effective amount” refers to an amount sufficient to prevent or treat the above-mentioned diseases.
본 명세서에서 용어 “예방”은 질환 또는 질환 상태의 방지 또는 보호적인 치료를 의미한다. 본 명세서에서 용어 “치료”는 질환 상태의 감소, 억제, 진정 또는 근절을 의미한다.As used herein, the term “prevention” refers to the prevention or protective treatment of a disease or disease state. As used herein, the term “treatment” refers to reduction, suppression, sedation or eradication of a disease state.
본 발명의 약제학적 조성물은 당해 발명이 속하는 기술분야에서 통상의 지식을 가진 자가 용이하게 실시할 수 있는 방법에 따라, 약제학적으로 허용되는 담체 및/또는 부형제를 이용하여 제제화 함으로써 단위 용량 형태로 제조되거나 또는 다용량 용기 내에 내입시켜 제조될 수 있다. 이때 제형은 내복약, 주사제 등 다양하게 제조될 수 있고, 오일 또는 수성 매질중의 용액, 현탁액 또는 유화액 형태이거나 엑스제, 산제, 좌제, 분말제, 과립제, 정제 또는 캅셀제 형태일 수도 있으며, 분산제 또는 안정화제를 추가적으로 포함할 수 있다.The pharmaceutical composition of the present invention is prepared in unit dosage form by formulating using a pharmaceutically acceptable carrier and/or excipient according to a method that can be easily carried out by a person of ordinary skill in the art to which the present invention pertains. or may be prepared by incorporation into a multi-dose container. At this time, the dosage form may be prepared in various ways such as oral medicine, injection, etc., in the form of a solution, suspension or emulsion in oil or aqueous medium, or in the form of an extract, powder, suppository, powder, granule, tablet or capsule, dispersant or stable Additional topics may be included.
본 발명의 실시예에 따르면, 본 발명의 13-HODE를 포함하는 약제학적 조성물은 엠토르 신호전달과정을 억제하는 효과가 있다. According to an embodiment of the present invention, the pharmaceutical composition comprising 13-HODE of the present invention has an effect of inhibiting the emtor signaling process.
또한, 본 발명의 실시예에 따르면, 본 발명의 상기 약제학적 조성물은 암세포의 성장을 억제하는 효과 및 생체내에서 종양의 성장을 억제하는 효과가 있다. 따라서, 본 발명의 상기 약제학적 조성물은 암, 보다 구체적으로는 자궁내막암에 대해 항암 효과가 있다.In addition, according to an embodiment of the present invention, the pharmaceutical composition of the present invention has the effect of inhibiting the growth of cancer cells and the effect of inhibiting the growth of tumors in vivo. Therefore, the pharmaceutical composition of the present invention has an anticancer effect on cancer, more specifically endometrial cancer.
본 발명의 다른 일 양태에 따르면, 본 발명은 상술한 본 발명의 13-HODE를 유효성분으로 포함하는 약제학적 조성물을 대상체(subject)에 투여하는 단계를 포함하는 암의 예방 또는 치료방법을 제공한다. According to another aspect of the present invention, the present invention provides a method for preventing or treating cancer comprising administering to a subject a pharmaceutical composition comprising the above-described 13-HODE of the present invention as an active ingredient. .
본 명세서에서 사용된 용어, "투여" 또는 "투여하다"는 본 발명의 조성물의 치료적, 또는 예방적 유효량을 상기 대상 질환을 겪거나, 겪을 가능성이 있는 대상체(개체)에 직접적으로 투여함으로써 대상체의 체내에서 동일한 양이 형성되도록 하는 것을 말한다. As used herein, the term "administration" or "administer" refers to directly administering a therapeutically or prophylactically effective amount of a composition of the present invention to a subject (individual) suffering from, or likely to suffer from, the subject disease. It means that the same amount is formed in the body of
상기 조성물의 "치료적 유효량"은 조성물을 투여하고자 하는 대상체에게 치료적 또는 예방적 효과를 제공하기에 충분한 조성물의 함량을 의미하며, 이에 "예방적 유효량"을 포함하는 의미이다.The "therapeutically effective amount" of the composition means an amount of the composition sufficient to provide a therapeutic or prophylactic effect to a subject to which the composition is administered, and includes a "prophylactically effective amount".
또한, 본 명세서에서 사용된 용어, "대상체(개체)"는 인간, 마우스, 랫트, 기니아 피그, 개, 고양이, 말, 소, 돼지, 원숭이, 침팬지, 비비 및 붉은털 원숭이 등을 포함하는 포유류이다. 가장 구체적으로는, 본 발명의 대상체는 인간이다.In addition, as used herein, the term "subject (subject)" is a mammal, including humans, mice, rats, guinea pigs, dogs, cats, horses, cattle, pigs, monkeys, chimpanzees, baboons and rhesus monkeys. . Most specifically, the subject of the present invention is a human.
본 발명의 상기 암의 예방, 또는 치료방법은, 본 발명의 일 양태인 약제학적 조성물을 투여하는 단계를 포함하는 방법이므로, 중복되는 내용에 대해서는 본 명세서의 과도한 중복성을 피하기 위하여 그 기재를 생략한다.Since the method for preventing or treating cancer of the present invention includes administering the pharmaceutical composition according to an aspect of the present invention, the description thereof is omitted to avoid excessive redundancy of the present specification for overlapping content. .
본 발명의 특징 및 이점을 요약하면 다음과 같다:The features and advantages of the present invention are summarized as follows:
본 발명은 13-HODE(13-Hydroxyoctadecadienoic acid)를 유효성분으로 포함하는 암의 예방 또는 치료용 약제학적 조성물을 제공한다. The present invention provides a pharmaceutical composition for preventing or treating cancer comprising 13-HODE (13-Hydroxyoctadecadienoic acid) as an active ingredient.
본 발명의 조성물은 암 세포의 성장을 억제하고 암 세포를 사멸시키는 효능을 나타내어 항암제로 사용이 가능하다. The composition of the present invention can be used as an anticancer agent by inhibiting the growth of cancer cells and exhibiting the efficacy of killing cancer cells.
도 1은 13-HODE의 화학식을 나타낸다.1 shows the chemical formula of 13-HODE.
도 2는 풀 다운 분석(pull-down assay)을 통해 엠토르 단백질과 13-HODE 사이의 결합을 분석한 결과를 나타낸다.2 shows the results of analyzing the binding between the emtor protein and 13-HODE through a pull-down assay.
도 3a는 수소-중수소 치환 질량분석법을 통해 13-HODE가 결합하는 엠토르 단백질의 분자적 위치를 분석한 결과를 나타낸다. Figure 3a shows the results of analyzing the molecular position of the emtor protein to which 13-HODE binds through hydrogen-deuterium substitution mass spectrometry.
도 3b는 13-HODE가 결합하는 분자적 위치인 엠토르 단백질의 촉매 중열(catalytic cleft)을 나타낸다. Figure 3b shows the catalytic cleft of the emtor protein, which is the molecular site at which 13-HODE binds.
도 4a는 면역침강을 통해 JHUEM7 세포에 대하여 13-HODE를 처리한 후 세포의 엠토르 신호전달과 관련된 단백질을 분석한 결과를 나타낸다. Figure 4a shows the results of analyzing proteins related to emtor signaling in cells after 13-HODE was treated with respect to JHUEM7 cells through immunoprecipitation.
도 4b는 면역침강을 통해 JHUEM7 세포에 대하여 13-HODE를 처리한 후 세포의 엠토르 신호전달과 관련된 단백질을 분석한 결과를 나타낸다. Figure 4b shows the results of analyzing proteins related to emtor signaling in cells after 13-HODE was treated with respect to JHUEM7 cells through immunoprecipitation.
도 4c는 면역침강을 통해 JHUEM7 세포에 대하여 13-HODE를 처리한 후 세포의 엠토르 신호전달과 관련된 단백질을 분석한 결과를 나타낸다. Figure 4c shows the results of analysis of proteins related to emtor signaling in cells after 13-HODE was treated with respect to JHUEM7 cells through immunoprecipitation.
도 5a는 웨스턴 블롯팅을 통해 JHUEM7 세포에 대하여 13-HODE를 처리한 후 세포의 엠토르 신호전달과 관련된 단백질을 분석한 결과를 나타낸다. Figure 5a shows the results of analyzing proteins related to emtor signaling in cells after 13-HODE was treated with respect to JHUEM7 cells through western blotting.
도 5b는 웨스턴 블롯팅을 통해 HCT116, SW480, HT29, MDA-MB-468, U-373 MG 세포에 대하여 13-HODE를 처리한 후 세포의 엠토르 신호전달과 관련된 단백질을 분석한 결과를 나타낸다. Figure 5b shows the results of analyzing proteins related to emtor signaling in HCT116, SW480, HT29, MDA-MB-468, and U-373 MG cells after treatment with 13-HODE through Western blotting.
도 6a는 JHUEM7 세포에 대하여 13-HODE를 처리한 후 세포의 생존율을 분석한 결과를 나타낸다.6a shows the results of analyzing the viability of JHUEM7 cells after treatment with 13-HODE.
도 6b는 MDA-MB-468 세포에 대하여 13-HODE를 처리한 후 세포의 생존율을 분석한 결과를 나타낸다. Figure 6b shows the results of analyzing the viability of MDA-MB-468 cells after treatment with 13-HODE.
도 7a는 JHUEM7 세포에 대하여 13-HODE를 처리한 후 암세포 콜로니 증식 정도를 분석한 결과를 나타낸다. 7a shows the results of analyzing the degree of cancer cell colony proliferation after treatment with 13-HODE for JHUEM7 cells.
도 7b는 MDA-MB-468 세포에 대하여 13-HODE를 처리한 후 암세포 콜로니 증식 정도를 분석한 결과를 나타낸다. Figure 7b shows the results of analyzing the degree of cancer cell colony proliferation after 13-HODE treatment with respect to MDA-MB-468 cells.
도 8a는 생쥐에 이종이식한 JHUEM7 세포를 13-HODE로 처리한 후 종양의 크기 변화를 분석한 결과를 나타낸다.Figure 8a shows the results of analyzing the size change of the tumor after treatment with 13-HODE JHUEM7 cells xenografted into mice.
도 8b는 생쥐에 이종이식한 JHUEM7 세포를 13-HODE로 처리한 후 종양의 크기 변화를 관찰한 결과를 나타낸다.Figure 8b shows the results of observing the size change of the tumor after treatment with 13-HODE JHUEM7 cells xenografted into mice.
도 9는 웨스턴 블롯팅을 통해 생쥐에서 적출한 종양 시료를 분석한 결과를 나타낸다.9 shows the results of analysis of tumor samples extracted from mice through Western blotting.
도 10a는 생쥐에 이종이식한 MDA-MB-468 세포를 13-HODE로 처리한 후 종양의 크기 변화를 분석한 결과를 나타낸다.Figure 10a shows the results of analyzing the size change of the tumor after treatment with 13-HODE of MDA-MB-468 cells xenografted into mice.
도 10b는 생쥐에 이종이식한 MDA-MB-468 세포를 13-HODE로 처리한 후 종양의 크기 변화를 관찰한 결과를 나타낸다.Figure 10b shows the results of observing the size change of the tumor after treatment with 13-HODE of MDA-MB-468 cells xenografted into mice.
도 11a는 JHUEM7 세포에 각각 DMSO 대조군 약물과 150 μM 13-HODE를 48 시간 처리한 후 RNA-seq 분석을 이용하여 유의한 유전자를 분석한 결과를 나타낸다.11a shows the results of analyzing significant genes using RNA-seq analysis after treating JHUEM7 cells with DMSO control drug and 150 μM 13-HODE for 48 hours, respectively.
도 11b는 JHUEM7 세포에 각각 DMSO 대조군 약물과 150 μM 13-HODE를 48 시간 처리한 후 RNA-seq 분석을 이용하여 경로 농축 분석을 수행한 결과를 나타낸다.11b shows the results of performing pathway enrichment analysis using RNA-seq analysis after treating JHUEM7 cells with DMSO control drug and 150 μM 13-HODE for 48 hours, respectively.
도 11c는 JHUEM7 세포에 각각 DMSO 대조군 약물과 150 μM 13-HODE를 48 시간 처리한 후 RNA-seq 분석을 이용하여 경로 농축 분석을 수행한 결과를 나타낸다. 11c shows the results of performing pathway enrichment analysis using RNA-seq analysis after treating JHUEM7 cells with DMSO control drug and 150 μM 13-HODE for 48 hours, respectively.
도 12는 13-HODE가 처리된 JHUEM7 세포의 생존율 및 세포 내부의 13-HODE 양을 정량적으로 분석한 결과를 나타낸다. 12 shows the results of quantitative analysis of the viability of 13-HODE-treated JHUEM7 cells and the amount of 13-HODE inside the cells.
도 13은 웨스턴 블롯팅을 통해 ALOX15를 과발현하는 JHUEM7 세포의 엠토르 신호전달과 관련된 단백질을 분석한 결과를 나타낸다.13 shows the results of analyzing proteins related to emtor signaling in JHUEM7 cells overexpressing ALOX15 through western blotting.
도 14는 생쥐에 ALOX15를 과발현하는 JHUEM7 세포를 이식한 후 종양의 크기 변화를 분석한 결과를 나타낸다.14 shows the results of analyzing the change in tumor size after transplanting JHUEM7 cells overexpressing ALOX15 into mice.
도 15a는 웨스턴 블롯팅을 통해 ALOX15를 과발현하는 MCF7 세포의 엠토르 신호전달과 관련된 단백질을 분석한 결과를 나타낸다.15A shows the results of analyzing proteins related to emtor signaling in MCF7 cells overexpressing ALOX15 through Western blotting.
도 15b는 ALOX15를 과발현하는 MCF7 세포 콜로니 증식 정도를 분석한 결과를 나타낸다. 15B shows the results of analyzing the degree of colony proliferation of MCF7 cells overexpressing ALOX15.
도 16a는 웨스턴 블롯팅을 통해 HEC59 세포에 대하여 13-HODE를 처리한 후 세포의 엠토르 신호전달과 관련된 단백질을 분석한 결과를 나타낸다. Figure 16a shows the results of analyzing proteins related to emtor signaling in cells after 13-HODE was treated with respect to HEC59 cells through western blotting.
도 16b는 HEC59 세포에 대하여 13-HODE를 처리한 후 세포의 생존율을 분석한 결과를 나타낸다.Figure 16b shows the results of analyzing the viability of HEC59 cells after treatment with 13-HODE.
도 16c는 HEC59 세포에 대하여 13-HODE를 처리한 후 암세포 콜로니 증식 정도를 분석한 결과를 나타낸다.Figure 16c shows the results of analyzing the degree of cancer cell colony proliferation after 13-HODE treatment on HEC59 cells.
이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 보다 구체적으로 설명하기 위한 것으로, 본 발명의 요지에 따라 본 발명의 범위가 이들 실시예에 의해 제한되지 않는다는 것은 당업계에서 통상의 지식을 가진 자에 있어서 자명할 것이다.Hereinafter, the present invention will be described in more detail through examples. These examples are only for illustrating the present invention in more detail, and it will be apparent to those skilled in the art that the scope of the present invention is not limited by these examples according to the gist of the present invention. .
실시예Example
본 명세서 전체에 걸쳐, 특정 물질의 농도를 나타내기 위하여 사용되는 "%"는 별도의 언급이 없는 경우, 고체/고체는 (중량/중량) %, 고체/액체는 (중량/부피) %, 그리고 액체/액체는 (부피/부피) %이다.Throughout this specification, "%" used to indicate the concentration of a specific substance is (weight/weight) % for solid/solid, (weight/volume) % for solid/liquid, and Liquid/liquid is (volume/volume) %.
실시예 1: 세포의 배양 방법Example 1: Cell culture method
HEK293, HEK293T, MDA-MB-468, MCF7, HEC59 및 U373MG 세포는 2 mM 글루타민, 100 μg/ml 페니실린-스트렙토마이신 및 10 % 소태아 혈청(FBS, Atlas Biological)이 보충된 high-glucose DMEM 배지에서 배양하였다. JHUEM7 세포는 1X 비필수 아미노산(Sigma Aldrich, Missouri, USA) 및 10 % FBS가 보충된 DMEM/F12(Thermo Fisher Scientific)에서 배양하였다. SW480, HT29 세포는 2 mM 글루타민, 100 μg/ml 페니실린-스트렙토마이신 및 10 % 소태아 혈청(FBS, Atlas Biological)이 보충된 RPMI1640 배지에서 배양하였다. 모든 세포주는 37 ℃, 5 % CO2 조건에서 배양하였다. HEK293, HEK293T, MDA-MB-468, MCF7, HEC59 and U373MG cells were cultured in high-glucose DMEM medium supplemented with 2 mM glutamine, 100 µg/ml penicillin-streptomycin and 10% fetal bovine serum (FBS, Atlas Biological). cultured. JHUEM7 cells were cultured in DMEM/F12 (Thermo Fisher Scientific) supplemented with 1X non-essential amino acids (Sigma Aldrich, Missouri, USA) and 10% FBS. SW480 and HT29 cells were cultured in RPMI1640 medium supplemented with 2 mM glutamine, 100 μg/ml penicillin-streptomycin and 10% fetal bovine serum (FBS, Atlas Biological). All cell lines were cultured at 37 °C, 5% CO 2 conditions.
HEK293 및 HEK293T 세포는 ATCC(American Type Culture Collection, Virginia, USA)로부터 구입하였고, JHUEM7 세포는 RIKEN(Saitama, Japan)으로부터 구입하였다. HEK293 and HEK293T cells were purchased from ATCC (American Type Culture Collection, Virginia, USA), and JHUEM7 cells were purchased from RIKEN (Saitama, Japan).
실시예 2: 풀다운 분석(pull-down assay)를 통한 엠토르와 13-HODE 사이의 관계 분석Example 2: Analysis of the relationship between Emtor and 13-HODE through pull-down assay
JHUEM7 세포를 1x CHAPS 용해 버퍼로 용해하였다. CHAPS 버퍼의 조성은 pH 7.4의 50 mM HEPES, 150 mM NaCl, 0.4% CHAPS, 50 mM NaF, 10 mM Na-파이로포스페이트, 100 mM β-글리세로포스페이트, 및 프로테아제 억제제이다.JHUEM7 cells were lysed with 1x CHAPS lysis buffer. The composition of the CHAPS buffer is 50 mM HEPES, 150 mM NaCl, 0.4% CHAPS, 50 mM NaF, 10 mM Na-pyrophosphate, 100 mM β-glycerophosphate at pH 7.4, and a protease inhibitor.
2 mg의 총 세포 용해물을 1) 10 μM의 비오틴, 2) 10 μM의 비오틴 표지된 13-HODE(US, Michigan, Cayman Chemical), 3) 10 μM의 비오틴 및 10 μM의 비오틴 표지된 13-HODE, 또는 4) 10 μM의 비오틴 표지된 13-HODE 및 20 μM의 라벨되지 않은 13-HODE와 함께 혼합하였다. 스트렙타비딘 비드(Streptavidin bead)를 상기 반응물에 첨가하고 추가적으로 한시간동안 더 혼합한 뒤, 10 μL 샘플 버퍼를 첨가하여 반응을 멈추게 하였고, 5분 동안 가열한 후, 웨스턴 블로팅으로 분석하였다. 2 mg of total cell lysate were prepared with 1) 10 μM biotin, 2) 10 μM biotin-labeled 13-HODE (US, Michigan, Cayman Chemical), 3) 10 μM biotin and 10 μM biotin-labeled 13- HODE, or 4) 10 μM biotin labeled 13-HODE and 20 μM unlabeled 13-HODE. Streptavidin beads (Streptavidin bead) were added to the reaction mixture and mixed for an additional hour. Then, 10 μL sample buffer was added to stop the reaction, and after heating for 5 minutes, analysis was performed by Western blotting.
상기 웨스턴 블로팅에서 사용된 항체는 항-mTOR 항체(Cell Signaling Technology #2972, Massachusetts, USA), 항-raptor 항체(Bethyl Laboratories A300-553A, Alabama, USA)이다. Antibodies used in the Western blotting were anti-mTOR antibody (Cell Signaling Technology #2972, Massachusetts, USA) and anti-raptor antibody (Bethyl Laboratories A300-553A, Alabama, USA).
비오틴 표지된 13-HODE는 엠토르 및 랩터(raptor, 엠토르와 함께 단백질 복합체를 이루는 조절 단백질)와 결합하였다. 다만, 비오틴 표지된 13-HODE는 비표지된 13-HODE의 존재하에서 엠토르 및 랩터와 낮은 결합력을 보였다 (도 2). Biotin-labeled 13-HODE was bound to emtor and raptor (regulatory protein that forms a protein complex with emtor). However, biotin-labeled 13-HODE showed a low binding affinity to emtor and raptor in the presence of unlabeled 13-HODE ( FIG. 2 ).
상기 결과를 통해, 13-HODE와 엠토르가 상호작용을 한다는 사실을 알 수 있다. From the above results, it can be seen that 13-HODE and emtor interact.
실시예 3: 수소-중수소 치환 질량분석법을 통한 엠토르와 13-HODE 사이의 관계 분석Example 3: Analysis of the Relationship Between Emtor and 13-HODE by Hydrogen-Deuterium Substitution Mass Spectrometry
실시예 2에서 확인한 13-HODE와 엠토르 사이의 상호작용을 보다 구체적으로 확인하기 위해, 수소-중수소 치환 질량분석법을 수행하였다.In order to more specifically confirm the interaction between 13-HODE and emtor confirmed in Example 2, hydrogen-deuterium substitution mass spectrometry was performed.
수소-중수소 치환 질량분석법을 통해 13-HODE가 결합하는 엠토르 C-말단의 구조적, 형태적 변화를 관찰하였고, 13-HODE의 작용 메커니즘 및 엠토르에 결합하는 분자적 위치를 확인하였다. Through hydrogen-deuterium substitution mass spectrometry, structural and conformational changes at the C-terminus of Emtor to which 13-HODE binds were observed, and the mechanism of action of 13-HODE and the molecular position of binding to Emtor were confirmed.
단백질 서열 커버리지 맵은 다음과 같은 중수소로 치환되지 않은 시료를 사용하여 확보하였다: 3.5 μL의 ~50 μM 엠토르 C-말단 단편, pH 7.4의 25mM HEPES, 50mM KCl, 10 mM MgCl2, 얼음으로 냉각된 96.5 μL 퀜치(quench) 용액(100 mM 글리신, 25 mM TCEP, pH 2.5)에 의해 희석된 10% 글리세롤.Protein sequence coverage maps were obtained using undeuterated samples as follows: 3.5 μL of ~50 μM Emtor C-terminal fragment, 25 mM HEPES, pH 7.4, 50 mM KCl, 10 mM MgCl 2 , ice-cold 10% glycerol diluted with 96.5 µL quench solution (100 mM glycine, 25 mM TCEP, pH 2.5).
상기 샘플을 Waters HDX nanoAcquity UPLC(Waters, Milford, MA)를 거쳐 in-line 펩신 처리(Waters Enzymate BEH pepsin 40 column)하였다. 펩티드 단편들을 Acquitty UPLC BEH C18 펩티드 트랩 및 컬럼을 통해 분리하였다. 분리 후에, 5 % 내지 35 % 아세토니트릴 (0.1 % 포름산) 구배를 사용하여 펩티드를 Waters Synapt G2 질량 분석기 (Waters, Milford, MA)로 직접 용리시켰다. MSE 데이터는 20 내지 30 V 램프 트랩 CE를 통해 얻었다. 펩타이드는 Waters의 ProteinLynx Global Server 2.5.1 (PLGS)을 사용하여 분석하였다. The sample was subjected to Waters HDX nanoAcquity UPLC (Waters, Milford, Mass.) and subjected to in-line pepsin treatment (Waters Enzymate BEH pepsin 40 column). Peptide fragments were separated through an Acquitty UPLC BEH C18 peptide trap and column. After separation, the peptides were eluted directly with a Waters Synapt G2 mass spectrometer (Waters, Milford, MA) using a gradient of 5% to 35% acetonitrile (0.1% formic acid). MS E data were obtained via 20 to 30 V lamp trap CE. Peptides were analyzed using Waters' ProteinLynx Global Server 2.5.1 (PLGS).
13-HODE가 결합하는 엠토르 C-말단을 분석하기 위해 다음과 같은 시료를 사용하였다: 3.5 μL의 ~50 μM 엠토르 C-말단 단편, pH 7.4의 25 mM HEPES, 50 mM KCl, 10 mM MgCl2, 10 % 글리세롤, 1% DMSO, 130 μM 13-HODE. 대조군으로는 13-HODE를 포함하지 않는 시료를 사용하였다. The following samples were used to analyze the Emtor C-terminus to which 13-HODE binds: 3.5 µL of -50 µM Emtor C-terminal fragment, 25 mM HEPES, pH 7.4, 50 mM KCl, 10 mM MgCl 2 , 10% glycerol, 1% DMSO, 130 μM 13-HODE. As a control, a sample not containing 13-HODE was used.
중수소 치환 반응을 일으키기 위해 상기 시료에 D20를 첨가하였다. 중수소 치환 반응은 다양한 조건으로(10 초, 1 분, 10 분 및 2 시간) 수행하였다. D 2 0 was added to the sample to initiate a deuterium substitution reaction. The deuterium substitution reaction was performed under various conditions (10 seconds, 1 minute, 10 minutes and 2 hours).
역 교환 수정(Back exchange correction)을 퀜치 전에 6 M 중수소화구아니딘 DCI (deuterated Guanidine DCl)를 첨가하여 수행하였다. 퀜치는 냉각된 100 mM 글리신 버퍼, 5 mM TCEP를 포함하는 65 μL 용액을 사용하여 수행하였다. 마지막으로, Waters DynamX 3.0 software를 통해 수소-중수소 치환에서의 중수소 흡수(% D)를 반응 시간별로 분석하였다.Back exchange correction was performed by adding 6 M deuterated guanidine DCI (DCI) before quenching. Quenching was performed using a 65 μL solution containing chilled 100 mM glycine buffer, 5 mM TCEP. Finally, deuterium uptake (% D) in hydrogen-deuterium substitution was analyzed by reaction time through Waters DynamX 3.0 software.
펩티드에 대한 배양 시간 t에서의 중수소 흡수율 (% D)의 표준화된 백분율을 다음과 같이 계산하였다: % D = (100 (mt-m0)) / (mf-m0). (mt: 인큐베이션 시간 t에서 중심 질량, m0: 중수소화되지 않은 대조군의 중심 질량, mf: 완전 중수소화 된 대조군의 중심 질량) 상기 % D를 사용하여 중수소화 백분율 플롯(△% D)을 계산하였다. △% D 플롯에 대한 신뢰 구간을 결정하고 완전 중수소화된 대조군을 사용하여 중수소화 백분율로 조정하였다.The normalized percentage of deuterium uptake (% D) at incubation time t for the peptide was calculated as follows: % D = (100 (mt-m0)) / (mf-m0). (mt: central mass at incubation time t, m0: central mass of non-deuterated control, mf: central mass of fully deuterated control) The % D was used to calculate the percent deuterated plot (Δ% D) . Confidence intervals for the Δ% D plot were determined and adjusted for percent deuterated using a fully deuterated control.
13-HODE의 존재하에, 일부 영역은 변경된 중수소 흡수 동역학을 나타내었고, 이는 주로 키나아제 도메인이였다 (도 3a). In the presence of 13-HODE, some regions exhibited altered deuterium uptake kinetics, mainly the kinase domain (Fig. 3a).
따라서, 상기 결과와 기존에 알려진 엠토르의 크리스탈 구조를 종합하여, 13-HODE의 결합부위가 키나아제 단백질의 촉매 중열(catalytic cleft)에 가로질러 걸쳐있는 것을 확인하였다 (도 3b). Therefore, by combining the above results with the previously known crystal structure of Emtor, it was confirmed that the binding site of 13-HODE spanned across the catalytic cleft of the kinase protein ( FIG. 3b ).
실시예 4: 면역침강을 통하여 분리한 엠토르 키나아제에 13-HODE를 처리 및 분석Example 4: Treatment and analysis of 13-HODE on Emtor kinase isolated through immunoprecipitation
엠토르 키나아제 분석을 수행하기 위해, HEK293T 세포를 얼음으로 냉각된 PBS로 한번 세척하고, 얼음으로 냉각된 용해 버퍼로 용해하였다. 용해 버퍼의 조성은 pH 7.4의 HEPES 40 mM, 120mM NaCl, 2 mM EDTA, 10 mM 파이로포스페이트, 10 mM 플루오린화나트륨, 0.3% CHAPS, 1X 프로테아제 억제제 칵테일이다. To perform the Emtor kinase assay, HEK293T cells were washed once with ice-cold PBS and lysed with ice-cold lysis buffer. The composition of the lysis buffer is HEPES 40 mM, 120 mM NaCl, 2 mM EDTA, 10 mM pyrophosphate, 10 mM sodium fluoride, 0.3% CHAPS, IX protease inhibitor cocktail at pH 7.4.
세포 용해물을 4 ℃에서 10분동안 배양하고, 13,000 rpm에서 10분 동안 원심 분리하여 상층액을 수집하였다. 2 μg 항-mTOR 항체(Cell Signaling Technology #2972, Massachusetts, USA)를 2 mg 상층액에 첨가하고 4 ℃에서 1.5시간 동안 회전 배양하였다. 20 μL의 아가로즈 비드(Pierce)를 첨가하고 추가적으로 한시간동안 더 배양하였다.The cell lysate was incubated at 4° C. for 10 minutes and centrifuged at 13,000 rpm for 10 minutes to collect the supernatant. 2 μg of anti-mTOR antibody (Cell Signaling Technology #2972, Massachusetts, USA) was added to 2 mg of the supernatant and spin-incubated at 4° C. for 1.5 hours. 20 μL of agarose beads (Pierce) was added and incubated for an additional hour.
엠토르 면역침전물을 상기 용해 버퍼로 2회 및 키나아제 세척 버퍼로 2회 세척하였다. 키나아제 세척 버퍼의 조성은 pH 7.4의 25 mM HEPES, 20 mM 염화칼륨이다. Emtor immunoprecipitates were washed twice with the lysis buffer and twice with kinase wash buffer. The composition of the kinase wash buffer is 25 mM HEPES, 20 mM potassium chloride, pH 7.4.
그 후, 15 μL의 엠토르 C1(mTOR Complex 1) 키나아제 버퍼 및 150 ng의 S6K1 기질을 이용하여 37 ℃에서 15 분동안 키나아제 분석을 수행하였다. 10 μL 샘플 버퍼를 첨가하여 반응을 멈추게 하였고, 5분 동안 가열한 후, 웨스턴 블로팅으로 분석하였다.Then, the kinase assay was performed at 37° C. for 15 minutes using 15 μL of mTOR Complex 1 kinase buffer and 150 ng of S6K1 substrate. The reaction was stopped by adding 10 μL sample buffer, heated for 5 minutes, and then analyzed by Western blotting.
상기 웨스턴 블로팅에서 사용된 항체는 항-T389 p-S6K 항체(Cell Signaling Technology #9205, Massachusetts, USA), 항-S473 p-Akt 항체(Cell Signaling Technology #9271), 항-S6K 항체(Cell Signaling Technology #9202), 항-Akt 항체(Cell Signaling Technology #9272), 항-mTOR 항체(Cell Signaling Technology #2972,), 항-mLST 항체(Cell Signaling Technology #3274,), 항-raptor 항체(Bethyl Laboratories A300-553A, Alabama, USA), 항-rictor 항체(Bethyl Laboratories A400-458A)이다. Antibodies used in the Western blotting were anti-T389 p-S6K antibody (Cell Signaling Technology #9205, Massachusetts, USA), anti-S473 p-Akt antibody (Cell Signaling Technology #9271), and anti-S6K antibody (Cell Signaling Technology). Technology #9202), Anti-Akt Antibody (Cell Signaling Technology #9272), Anti-mTOR Antibody (Cell Signaling Technology #2972,), Anti-mLST Antibody (Cell Signaling Technology #3274,), Anti-raptor Antibody (Bethyl Laboratories) A300-553A, Alabama, USA), an anti-rictor antibody (Bethyl Laboratories A400-458A).
13-HODE의 존재 하에, 엠토르 C1 기질인 S6K1은 용량 의존적으로 억제되었다 (도 4a). 13-HODE의 존재 하에, S473에서 엠토르 C2(mTOR Complex 2) 기질인 Akt의 인산화는 추가로 차단되었다(도 4b). 상기 결과는 13-HODE가 나타내는 엠토르 C1 및 엠토르 C2에 대한 대사 산물의 억제활성을 나타낸다.In the presence of 13-HODE, the Emtor Cl substrate, S6K1, was dose-dependently inhibited (Fig. 4a). In the presence of 13-HODE, phosphorylation of Akt, a substrate of mTOR C2 (mTOR Complex 2) at S473, was further blocked ( FIG. 4b ). The above results show the inhibitory activity of metabolites for Emtor C1 and Emtor C2, which 13-HODE exhibits.
또한, 엠토르 C1 억제에 필요한 13-HODE의 농도와 관련하여, 엠토르 C2와 동일한 억제 수준을 달성하기 위해서는 더 적은 양이 필요하였고, 이는 13-HODE가 엠토르 C1에 대한 보다 강력한 활성을 나타낸다는 것을 의미한다. In addition, with respect to the concentration of 13-HODE required for Emtor C1 inhibition, a smaller amount was required to achieve the same level of inhibition as Emtor C2, indicating that 13-HODE has a stronger activity on Emtor C1. Means that.
13-HODE가 엠토르 키나아제 도메인을 직접 표적으로 하는지에 대한 추가 확인을 위해, 엠토르의 키나아제 도메인을 형성하는 아미노산 1362 내지 2549를 포함하는 재조합 엠토르 C-말단 단편을 사용하여 In-vitro 키나아제 분석을 수행하였다. 그 결과, 13-HODE는 S6K1의 인산화를 억제하였다 (도 4c). For further confirmation that 13-HODE directly targets the emtor kinase domain, an in-vitro kinase assay using a recombinant emtor C-terminal fragment comprising amino acids 1362 to 2549 forming the kinase domain of emtor was performed. As a result, 13-HODE inhibited phosphorylation of S6K1 ( FIG. 4c ).
상기 결과는 13-HODE가 엠토르의 키나아제 활성을 직접적으로 억제하고, 엠토르 키나아제 도메인 부분을 직접 표적한다는 사실을 나타낸다.These results indicate that 13-HODE directly inhibits the kinase activity of emtor and directly targets the emtor kinase domain portion.
실시예 5: 웨스턴 블로팅을 통한 13-HODE가 처리된 세포의 엠토르 키나아제 분석Example 5: Emtor kinase analysis of 13-HODE-treated cells by Western blotting
실시예 5-1. 웨스턴 블로팅을 통한 13-HODE가 처리된 JHUEM7 세포의 엠토르 키나아제 분석Example 5-1. Emtor kinase analysis of 13-HODE-treated JHUEM7 cells by Western blotting
JHUEM7 세포에서 엠토르 신호전달에 대한 활성을 분석하기 위해 엠토르 키나아제를 분석하였다. In order to analyze the activity of emtor signaling in JHUEM7 cells, emtor kinase was analyzed.
35mm 세포 배양 디쉬에 JHUEM7 3 x 105 개의 세포를 시딩하였다. 다음날 68 혹은 135 uM의 13-HODE 또는 10nM Torin1을 세포 배양액에 혼합하여 처리한 뒤 48시간 후에 웨스턴블로팅을 통해 엠토르 신호전달 분석을 하였다. 13-HODE 또는 Torin1 혼합 배양액은 매 24시간마다 새롭게 교체해주었다.35 mm cell culture dishes were seeded with JHUEM7 3×10 5 cells. The next day, 68 or 135 uM of 13-HODE or 10 nM Torin1 was mixed with the cell culture medium and treated, and then 48 hours later, emtor signaling was analyzed by western blotting. The 13-HODE or Torin1 mixed culture medium was freshly replaced every 24 hours.
상기 세포를 NP-40 용해 버퍼로 용해하였다. NP-40의 조성은 pH 7.5의 50 mM 트리스-HCl, 150 mM NaCl, 1% NP-40 서브스티튜트(NP-40 substitute), pH 8.0의 1 mM EDTA, 50 mM NaF, 10 mM Na-파이로포스페이트(Na-pyrophosphate), 15 mM Na3VO4, 100 mM β-글리세로포스페이트(β-glycerophosphate), 및 프로테아제 억제제이다.The cells were lysed with NP-40 lysis buffer. The composition of NP-40 is 50 mM Tris-HCl at pH 7.5, 150 mM NaCl, 1% NP-40 substitute, 1 mM EDTA at pH 8.0, 50 mM NaF, 10 mM Na-Pi lophosphate (Na-pyrophosphate), 15 mM Na 3 VO 4 , 100 mM β-glycerophosphate, and a protease inhibitor.
전체 세포 용해물을 5X SDS 샘플 버퍼와 혼합하여 최종 농도를 1-2 μg/μL 로 조정한 후 5 분간 가열하였다. 그 후 가열된 세포 용해물으로 웨스턴 블로팅을 수행하였다. The whole cell lysate was mixed with 5X SDS sample buffer to adjust the final concentration to 1-2 μg/μL and then heated for 5 minutes. Then, Western blotting was performed with the heated cell lysate.
상기 웨스턴 블로팅에서 사용된 항체는 항-T389 p-S6K 항체(Cell Signaling Technology #9205, Massachusetts, USA), 항-S240/244 p-S6 항체(Cell Signaling Technology #5364), 항-T37/46 p-4EBP1 항체(Cell Signaling Technology #9459), 항-S473 p-Akt 항체(Cell Signaling Technology #9271), 항-T308 p-Akt 항체(Cell Signaling Technology #4056), 항-T638/641 p-PKCa 항체(Cell Signaling Technology #9375), 항-S6K 항체(Cell Signaling Technology #9202), 항-S6 항체(Cell Signaling Technology #2217), 항-4EBP1 항체(Cell Signaling Technology #9452), 항-Akt 항체(Cell Signaling Technology #9272), 항-PKCa 항체(Cell Signaling Technology #2056)이다. Antibodies used in the Western blotting were anti-T389 p-S6K antibody (Cell Signaling Technology #9205, Massachusetts, USA), anti-S240/244 p-S6 antibody (Cell Signaling Technology #5364), anti-T37/46 p-4EBP1 antibody (Cell Signaling Technology #9459), anti-S473 p-Akt antibody (Cell Signaling Technology #9271), anti-T308 p-Akt antibody (Cell Signaling Technology #4056), anti-T638/641 p-PKCa Antibody (Cell Signaling Technology #9375), Anti-S6K Antibody (Cell Signaling Technology #9202), Anti-S6 Antibody (Cell Signaling Technology #2217), Anti-4EBP1 Antibody (Cell Signaling Technology #9452), Anti-Akt Antibody ( Cell Signaling Technology #9272), anti-PKCa antibody (Cell Signaling Technology #2056).
상기 실험을 통해, 13-HODE를 JHUEM7 세포에 처리하는 경우, T389 S6K1의 인산화는 감소, 즉 엠토르 신호전달 과정이 억제된다는 사실을 확인하였다(도 5a).Through the above experiment, it was confirmed that when 13-HODE was treated in JHUEM7 cells, phosphorylation of T389 S6K1 was reduced, that is, the emtor signaling process was inhibited ( FIG. 5a ).
실시예 5-2. 웨스턴 블로팅을 통한 13-HODE가 처리된 HCT003, SW480, HT29, MDA-MB-468, U-373MG 세포의 엠토르 키나아제 분석Example 5-2. Emtor kinase analysis of 13-HODE-treated HCT003, SW480, HT29, MDA-MB-468 and U-373MG cells by Western blotting
HCT003, SW480, HT29, MDA-MB-468, U-373MG 세포에서 엠토르 신호전달에 대한 활성을 분석하기 위해 엠토르 키나아제를 분석하였다. In order to analyze the activity of emtor signaling in HCT003, SW480, HT29, MDA-MB-468, and U-373MG cells, emtor kinase was analyzed.
실험 방법은 JHUEM7 세포 대신에 HCT003, SW480, HT29, MDA-MB-468, U-373MG 세포를 사용한 것을 제외하고는 실시예 5-1에 기재된 방법과 동일한 방법을 사용하였다. The experimental method was the same as the method described in Example 5-1, except that HCT003, SW480, HT29, MDA-MB-468, and U-373MG cells were used instead of JHUEM7 cells.
실험을 통해, 13-HODE를 HCT116 세포, SW480세포, HT29 세포, MDA-MB-468 세포, U-373 MG 세포에 처리하는 경우 T389 S6K1, S240/244, T37/46의 인산화가 감소되는 것을 확인, 즉 엠토르 신호전달 과정이 억제된다는 사실을 확인하였다(도 5b).Through the experiment, it was confirmed that phosphorylation of T389 S6K1, S240/244, and T37/46 was reduced when 13-HODE was treated with HCT116 cells, SW480 cells, HT29 cells, MDA-MB-468 cells, and U-373 MG cells. , that is, it was confirmed that the emtor signaling process was inhibited (FIG. 5b).
실시예 6: 13-HODE가 처리된 세포의 생존력 분석Example 6: Viability analysis of cells treated with 13-HODE
13-HODE의 항암효과를 확인하기 위해, 13-HODE를 처리한 JHUEM7 세포 및 MDA-MB-468 세포의 생존력을 분석하였다. To confirm the anticancer effect of 13-HODE, the viability of 13-HODE-treated JHUEM7 cells and MDA-MB-468 cells was analyzed.
JHUEM7 세포 또는 MDA-MB-468 세포(1 x 104 세포/mL)를 96 웰 웰플레이트에 시딩하였다. 오버나잇 배양하고, 세포를 DMSO 또는 135 μM의 13-HODE로 다양한 시간동안 처리하였다. 그 후, 20 μL의 MTT(3-(4,5-디메틸티아졸-2-일)-2,5-디페닐테트라졸륨 브로마이드) 용액(PBS에 2.5mg/mL)을 각 웰에 첨가하였고, 그 후 추가적으로 1시간동안 37 ℃로 배양하였다. 각 웰의 OD(optical density, 광학밀도) 값은 SpectraMax Paradigm Reader (Molecular Devices)로 492 nm에서 측정하였다. 콜로니 형성 시험을 위해, 세포(4 x 104 세포/웰)를 24 웰 플레이트에 시딩하여 오버나잇 배양하고, 세포를 DMSO 또는 135 μM의 13-HODE로 4일동안 배양하였다. 그 후 생성된 콜로니를 -20 ℃에서 30 분 동안 메탄올로 고정시키고 15 분 동안 1 % 크리스탈 바이올렛으로 염색하였다. JHUEM7 cells or MDA-MB-468 cells (1×10 4 cells/mL) were seeded in 96 well plates. After overnight incubation, cells were treated with DMSO or 135 μM of 13-HODE for various times. Then, 20 μL of MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) solution (2.5 mg/mL in PBS) was added to each well, Then, incubated at 37 °C for an additional 1 hour. The OD (optical density) value of each well was measured at 492 nm with a SpectraMax Paradigm Reader (Molecular Devices). For colony formation test, cells (4 x 10 4 cells/well) were seeded in 24 well plates and cultured overnight, and cells were cultured with DMSO or 135 μM of 13-HODE for 4 days. The resulting colonies were then fixed with methanol at -20 °C for 30 min and stained with 1% crystal violet for 15 min.
실험을 통해 13-HODE로 처리한 웰에서의 세포 증식율이 13-HODE를 처리하지 않는 웰에서의 증식율보다 현저히 낮다는 것을 확인하였다 (도 6a, 도 6b). Through the experiment, it was confirmed that the cell proliferation rate in the wells treated with 13-HODE was significantly lower than that in the wells not treated with 13-HODE ( FIGS. 6A and 6B ).
이러한 결과는 13-HODE가 매우 뛰어난 항암 효과를 갖고 있음을 나타낸다. These results indicate that 13-HODE has a very good anticancer effect.
실시예 7: 13-HODE가 처리된 세포의 콜로니 증식 분석Example 7: Colony proliferation analysis of 13-HODE-treated cells
JHUEM7 세포주 및 MDA-MB-468 세포주에서 13-HODE와 암세포 콜로니 증식율의 연관성을 확인하기 위해, JHUEM7 세포 또는 MDA-MB-468 세포주를 배양하는 웰에 13-HODE 첨가 시 암세포 콜로니 증식율을 살펴보았다.In order to confirm the association between 13-HODE and cancer cell colony growth rate in JHUEM7 cell line and MDA-MB-468 cell line, the cancer cell colony growth rate was examined when 13-HODE was added to wells culturing JHUEM7 cells or MDA-MB-468 cell line.
JHUEM7 세포 또는 MDA-MB-468 세포(4 x 104 세포/웰)를 24 웰 플레이트에 시딩하여 오버나잇 배양하고, 세포를 DMSO 또는 135 μM의 13-HODE로 4일동안 배양하였다. 그 후 생성된 콜로니를 -20 ℃에서 30 분 동안 메탄올로 고정시키고 15 분 동안 1 % 크리스탈 바이올렛으로 염색하였다. JHUEM7 cells or MDA-MB-468 cells (4 x 10 4 cells/well) were seeded in a 24-well plate and cultured overnight, and the cells were cultured with DMSO or 135 μM of 13-HODE for 4 days. The resulting colonies were then fixed with methanol at -20 °C for 30 min and stained with 1% crystal violet for 15 min.
실험을 통해 13-HODE로 처리한 웰에서의 세포 증식율이 13-HODE를 처리하지 않는 웰에서의 증식율보다 현저히 낮다는 것을 확인하였다. 이러한 결과는 13-HODE가 매우 뛰어난 항암 효과를 갖고 있음을 나타낸다 (도 7a, 도 7b). Through the experiment, it was confirmed that the cell proliferation rate in the wells treated with 13-HODE was significantly lower than that in the wells not treated with 13-HODE. These results indicate that 13-HODE has a very excellent anticancer effect ( FIGS. 7a and 7b ).
실시예 8: 동물 실험을 통해 확인한 13-HODE의 항암 효과 분석Example 8: Analysis of the anticancer effect of 13-HODE confirmed through animal experiments
실시예 8-1. JHUEM7 세포를 이식한 누드 마우스 실험을 통해 확인한 13-HODE의 항암 효과Example 8-1. The anticancer effect of 13-HODE confirmed through the experiment of nude mice implanted with JHUEM7 cells
생쥐 이종이식 모델에서의 13-HODE의 항암 효과를 확인하기 위해, 종양이 이식된 생쥐에 13-HODE을 처리한 후 종양의 크기 변화를 살펴보았다. In order to confirm the anticancer effect of 13-HODE in the mouse xenograft model, the size of the tumor was examined after treatment with 13-HODE in the tumor-transplanted mice.
동물실험은 한국과학기술원 동물실험윤리위원회가 승인한 가이드라인을 통해 수행하였다. JHUEM7 세포(4 x 106)를 8-12 주령의 암컷 BALB/c 누드 마우스에 피하주사하여 이식하였다. 평균 종양 부피가 50 mm3에 도달한 후, 마우스를 무작위로 3 개의 다른 그룹에(6 마리/그룹) 할당하였다. 마우스의 체중과 종양의 직경을 이틀에 한번 측정하였다. 종양 부피는 캘리퍼스를 이용하여 일반식 0.5x(width)2x(Length)에 따라 평가하였고, P-values를 결정하기 위해 스튜던트 T 검정을 이용하였다. 13-HODE 처리를 위해, 마우스는 실험 개시 후 2일마다 10 mg/kg의 13-HODE의 종양 내 주사 또는 정맥 주사를 받았다.Animal experiments were performed according to guidelines approved by the Animal Experimentation Ethics Committee of the Korea Advanced Institute of Science and Technology. JHUEM7 cells (4 x 10 6 ) were implanted by subcutaneous injection into 8-12 week old female BALB/c nude mice. After the mean tumor volume reached 50 mm 3 , mice were randomly assigned to 3 different groups (6 mice/group). The mouse weight and tumor diameter were measured once every two days. Tumor volume using a caliper was evaluated according to the following formula 0.5x (width) 2 x (Length ), a Student's T test was used to determine the P-values. For 13-HODE treatment, mice received intratumoral or intravenous injection of 13-HODE at 10 mg/kg every 2 days after initiation of the experiment.
실험을 통해 13-HODE를 처리한 생쥐 이종이식 모델의 종양의 성장은 대조군과 비교하였을 때 성장이 억제되는 것을 확인하였다 (도 8a, 도 8b).Through the experiment, it was confirmed that the growth of the tumor in the mouse xenograft model treated with 13-HODE was inhibited when compared to the control group ( FIGS. 8a and 8b ).
또한, 웨스턴 블롯팅을 통해 13-HODE를 처리한 종양에서 T389 S6K1의 인산화 및 S473 AKT의 인산화가 감소한 것을 확인하였다 (도 9). In addition, it was confirmed that phosphorylation of T389 S6K1 and phosphorylation of S473 AKT were decreased in the 13-HODE-treated tumor through Western blotting ( FIG. 9 ).
실시예 8-2. MDA-MB-468 세포를 이식한 누드 마우스 실험을 통해 확인한 13-HODE의 항암 효과Example 8-2. The anticancer effect of 13-HODE confirmed through an experiment in nude mice implanted with MDA-MB-468 cells
생쥐 이종이식 모델에서의 13-HODE의 항암 효과를 확인하기 위해, 종양이 이식된 생쥐에 13-HODE을 처리한 후 종양의 크기 변화를 살펴보았다. In order to confirm the anticancer effect of 13-HODE in the mouse xenograft model, the size of the tumor was examined after treatment with 13-HODE in the tumor-transplanted mice.
실험 방법은 JHUEM7 세포 대신에 MDA-MB-468 세포를 사용한 것을 제외하고는 실시예 8-1에 기재된 방법과 동일하다. The experimental method is the same as that described in Example 8-1, except that MDA-MB-468 cells were used instead of JHUEM7 cells.
실험을 통해 13-HODE를 처리한 생쥐 이종이식 모델의 종양의 성장은 대조군과 비교하였을 때 성장이 억제되는 것을 확인하였다 (도 10a, 도 10b).Through the experiment, it was confirmed that the growth of the tumor in the mouse xenograft model treated with 13-HODE was inhibited when compared to the control group ( FIGS. 10a and 10b ).
이러한 결과는 13-HODE가 엠토르 신호전달 과정을 억제하고 매우 뛰어난 항암 효과를 갖고 있음을 나타낸다.These results indicate that 13-HODE inhibits the emtor signaling process and has a very excellent anticancer effect.
실시예 9: 13-HODE가 처리된 세포의 유의 유전자 발현 및 기전 분석(RNA-seq assay)Example 9: Analysis of significant gene expression and mechanism of 13-HODE-treated cells (RNA-seq assay)
13-HODE의 약물 기전과 유전자간의 연관성을 확인하기 위해 RNA-seq 실험인 유의 유전자 분석(Differentially Expressed Genes analysis, DEG analysis) 및 경로 농축 분석(Pathway enrichment analysis)을 통해 유의하게 발현하는 유전자 및 관련 기전에 대한 분석을 수행하였다.Genes and related mechanisms that are significantly expressed through RNA-seq experiments, Differentially Expressed Genes analysis (DEG analysis) and Pathway enrichment analysis, to confirm the association between the drug mechanism of 13-HODE and genes analysis was performed.
DEG 분석은 유전자의 발현값을 측정하고 통계적으로 처리하여 대조군과 비교군 간에 발현이 차이가 나는 유의한 유전자(Differentially Expressed Genes) 후보군을 선발하는 분석이다. JHUEM7 세포에 각각 DMSO 대조군 약물과 150 μM 13-HODE를 48 시간 처리한 후 RNA-seq 분석을 이용하여 유의한 유전자를 분석한 결과. 13-HODE 처리군에서 3729 개의 유전자가 감소하였고 3842 개의 유전자가 증가하였다(도 11a).DEG analysis is an analysis that selects a candidate group for a significant gene (Differentially Expressed Genes) with a difference in expression between the control group and the control group by measuring and statistically processing the expression value of the gene. The result of analyzing significant genes using RNA-seq analysis after treating JHUEM7 cells with DMSO control drug and 150 μM 13-HODE for 48 hours, respectively. In the 13-HODE treatment group, 3729 genes were decreased and 3842 genes were increased ( FIG. 11a ).
상기 결과는 13-HODE 처리에 의하여 엠토르 신호 전달 경로 관련 유전자군의 변화가 있다는 것을 나타낸다. The above results indicate that there is a change in the gene group related to the emtor signal transduction pathway by 13-HODE treatment.
또한, 경로 농축 분석을 통해 간암, 췌장암, 유방암 등의 암 관련 조절 경로들이 검출되었고 (도 11b), 그 외에 세포 주기, 노화, 지방산 대사 관련 기전 경로 등이 검출되었다 (도 11c).In addition, through the pathway enrichment analysis, cancer-related regulatory pathways such as liver cancer, pancreatic cancer, and breast cancer were detected ( FIG. 11b ), and cell cycle, aging, and fatty acid metabolism-related mechanistic pathways were also detected ( FIG. 11c ).
실시예 10: 13-HODE가 처리된 세포의 생존율 및 세포 내부에서 측정된 13-HODEExample 10: Viability of cells treated with 13-HODE and 13-HODE measured inside cells
13-HODE가 세포 내부로 유입이 잘되는지 확인하기 위해 13-HODE가 처리된 JHUEM7 세포 내부의 13-HODE 양을 정량적으로 분석하였다. In order to check whether 13-HODE flows well into the cell, the amount of 13-HODE inside the 13-HODE-treated JHUEM7 cells was quantitatively analyzed.
실험결과 DMSO를 처리한 대조군에 비해 13-HODE 처리한 세포는 대조군에 비해 세포 내에서 13-HODE 양이 높게 관찰되었다 (도 12).As a result of the experiment, 13-HODE-treated cells compared to the DMSO-treated control group showed a higher amount of 13-HODE in the cells than the control group ( FIG. 12 ).
세포 내부의 13-HODE 양은 다음과 같은 방법으로 측정하였다. The amount of 13-HODE inside the cell was measured as follows.
13-HODE는 고체상 추출(solid phase extraction, SPE)을 사용하여 ~1.4 mL 세포 상층액에서 추출하였다. 60mg Oasis HLB (Waters) SPE 카트리지를 세척하고 에틸 아세테이트, 메탄올 및 (0.1 % 아세트산 + 5 % 메탄올) 수용액으로 순차적으로 프리 컨디셔닝하였다. MeOH:H2O (50:50)를 용매로 하는 10 μL의 0.2 mg/mL의 EDTA 및 BHT를 SPE 컬럼의 흡착제 베드에 첨가하였다. 동일한 부피 (0.1 % 아세트산 + 5 % MeOH) 수용액을 세포 상층액에 첨가하였다. 50 μL의 500 nM 13-HODE-d4도 내부 표준 물질로 샘플 용액에 첨가하였다. 상기 용액을 SPE 컬럼에 추가하였다. 컬럼을 2 컬럼 부피 (0.1 % 아세트산 + 5 % MeOH) 수용액으로 세척한 후, 컬럼을 진공을 사용하여 건조하였다. 마지막으로 13-HODE는 0.5 mL 메탄올에 이어 1.5 mL 에틸 아세테이트로 용출하였다. 샘플 용액을 진공 원심 분리기를 사용하여 건조시킨 다음 분석할 때까지 -20℃에서 보관하였다. 건조된 물질은 LC-MS/MS 분석 전에 40 μL의 메탄올로 환원하였다. 13-HODE는 Agilent 1290 HPLC (Agilent) 및 QTRAP 5500 질량 분석법 (AB Sciex)이 장착된 LC-MS/MS 시스템을 사용하여 측정하였다. 역상 컬럼 (Pursuit5 C18, 150 × 2.1 mm)을 이동상 A (0.1 % 아세트산 수용액) 및 이동상 B (아세토 니트릴/메탄올(84/16)을 용매로 하는 0.1 % 아세트산)와 함께 사용하였다. LC는 250 μL/min 및 35 ℃에서 실행하였다. 분리 구배는 다음과 같다: 0 분에 35 % B, 0.25 분 동안 35 % B 유지, 0.75 분 동안 B의 35 %에서 45 %로, 2 분 동안 B의 45 % 유지, 5.5 분 동안 B의 45 %에서 56 %로, 5.5 분 동안 B의 56 %에서 65 %로, 6 분 동안 B의 65 % 유지, 1.5 분 동안 B의 65 %에서 ~ 95 %로 유지, 5.5 분 동안 B의 95 % 유지, 0.1 분 동안 B의 95 %에서 35 %로, B의 35 %에서 2.9 분 동안 유지. 다중 반응 모니터링 (MRM) 모드는 13-HODE의 음이온 모드에서 수행되었으며, 13-HODE의 특정 전이에 해당하는 추출된 이온 크로마토 그램을 정량화에 사용하였다 (Q1/Q3 = 295/195, RT = 13-HODE의 경우 18.09 분; Q1/Q3 = 299/198, 13-HODE-d4의 경우 RT = 17.93 분). 13-HODE의 보정 범위는 1-10000 nM (r2 ≥ 0.99)이다. 데이터 분석은 Analyst 1.5.2를 사용하여 수행하였다.13-HODE was extracted from ~1.4 mL cell supernatant using solid phase extraction (SPE). A 60 mg Oasis HLB (Waters) SPE cartridge was washed and pre-conditioned sequentially with ethyl acetate, methanol and (0.1 % acetic acid + 5 % methanol) aqueous solution. 10 μL of 0.2 mg/mL EDTA and BHT with MeOH:H 2 O (50:50) as solvents were added to the adsorbent bed of the SPE column. An equal volume (0.1% acetic acid + 5% MeOH) aqueous solution was added to the cell supernatant. 50 μL of 500 nM 13-HODE-d4 was also added to the sample solution as an internal standard. The solution was added to the SPE column. After washing the column with 2 column volumes (0.1 % acetic acid + 5 % MeOH) aqueous solution, the column was dried using vacuum. Finally, 13-HODE was eluted with 0.5 mL methanol followed by 1.5 mL ethyl acetate. The sample solution was dried using a vacuum centrifuge and then stored at -20°C until analysis. The dried material was reduced with 40 μL of methanol before LC-MS/MS analysis. 13-HODE was determined using an LC-MS/MS system equipped with an Agilent 1290 HPLC (Agilent) and QTRAP 5500 mass spectrometry (AB Sciex). A reversed-phase column (Pursuit5 C18, 150×2.1 mm) was used with mobile phase A (0.1 % aqueous acetic acid solution) and mobile phase B (0.1 % acetic acid in acetonitrile/methanol (84/16) solvent). LC was run at 250 μL/min and 35°C. The separation gradient is as follows: 35% B at 0 min, 35% B hold for 0.25 min, 35% to 45% of B for 0.75 min, 45% hold of B for 2 min, 45% of B for 5.5 min. to 56% of B, from 56% to 65% of B for 5.5 min, hold 65% of B for 6 min, hold 65% to ~95% of B for 1.5 min, hold 95% of B for 5.5 min, 0.1 From 95% to 35% of B for min and hold for 2.9 min at 35% of B. Multiple reaction monitoring (MRM) mode was performed in anion mode of 13-HODE, and extracted ion chromatograms corresponding to specific transitions of 13-HODE were used for quantification (Q1/Q3 = 295/195, RT = 13- 18.09 min for HODE; Q1/Q3 = 299/198, RT = 17.93 min for 13-HODE-d4). The calibration range for 13-HODE is 1-10000 nM (r2 ≥ 0.99). Data analysis was performed using Analyst 1.5.2.
실시예 11: ALOX15 (human lipoxygenase15-1)을 과발현하는 JHUEM7 세포의 엠토르 키나아제 분석Example 11: Emtor kinase analysis of JHUEM7 cells overexpressing ALOX15 (human lipoxygenase15-1)
13-HODE의 합성을 촉매하는 것으로 알려진 ALOX15 (human lipoxygenase15-1)을 과발현하는 JHUEM7 세포를 이용하여 13-HODE가 엠토르 신호전달에 미치는 영향을 분석하였다. The effect of 13-HODE on emtor signaling was analyzed using JHUEM7 cells overexpressing ALOX15 (human lipoxygenase15-1), which is known to catalyze the synthesis of 13-HODE.
실험 방법은 사용한 세포 및 13-HODE를 처리하지 않는 것을 제외하고는 실시예 5에 기재된 방법과 동일하다. 대조군은 ALOX15을 과발현하지 않은 세포를 사용하였다. The experimental method is the same as the method described in Example 5, except that the cells used and 13-HODE were not treated. As a control group, cells not overexpressing ALOX15 were used.
실험결과 ALOX15를 과발현하는 세포는 T389 S6K1, S473, T308의 인산화가 감소하는 것을 확인, 즉 엠토르 신호전달 과정이 억제된다는 사실을 확인하였다 (도 13).As a result of the experiment, it was confirmed that the phosphorylation of T389 S6K1, S473, and T308 was decreased in the cells overexpressing ALOX15, that is, the emtor signaling process was inhibited (FIG. 13).
실시예 12: ALOX15 (human lipoxygenase15-1)을 과발현하는 JHUEM7 세포를 이식한 누드 마우스 실험을 통해 확인한 13-HODE의 항암 효과Example 12: Anticancer effect of 13-HODE confirmed through an experiment in nude mice implanted with JHUEM7 cells overexpressing ALOX15 (human lipoxygenase15-1)
생쥐 이종이식 모델에서의 13-HODE의 항암 효과를 확인하기 위해, ALOX15를 과발현하는 세포를 이식한 생쥐의 종양 크기 변화를 살펴보았다. In order to confirm the anticancer effect of 13-HODE in a mouse xenograft model, the tumor size change in mice transplanted with ALOX15 overexpressing cells was examined.
실험 방법은 JHUEM7 세포 대신에 ALOX15 과발현 JHEUM7 세포를 사용한 것 및 13-HODE를 처리하지 않는 것을 제외하고는 실시예 8에 기재된 방법과 동일하다. 대조군은 ALOX15을 과발현하지 않은 세포를 이식한 누드 마우스를 이용하였다. The experimental method was the same as that described in Example 8, except that ALOX15 overexpressing JHEUM7 cells were used instead of JHUEM7 cells and 13-HODE was not treated. As a control group, nude mice transplanted with cells not overexpressing ALOX15 were used.
실험을 통해 ALOX15 과발현 JHEUM7 세포를 이식한 생쥐 이종이식 모델의 종양의 성장은 대조군과 비교하였을 때 성장이 억제되는 것을 확인하였다 (도 14).Through the experiment, it was confirmed that the growth of the tumor in the mouse xenograft model transplanted with ALOX15 overexpressing JHEUM7 cells was inhibited when compared to the control group ( FIG. 14 ).
이러한 결과는 13-HODE가 엠토르 신호전달 과정을 억제하고 항암 효과를 갖고 있음을 나타낸다.These results indicate that 13-HODE inhibits the emtor signaling process and has an anticancer effect.
실시예 13: ALOX15 (human lipoxygenase15-1)을 과발현하는 MCF7 세포의 엠토르 키나아제 및 세포의 콜로니 증식 분석Example 13: Emtor kinase of MCF7 cells overexpressing ALOX15 (human lipoxygenase15-1) and colony proliferation analysis of cells
ALOX15를 과발현하는 MCF7 세포를 이용하여 엠토르 키나아제 및 세포 콜로니 증식 분석을 수행하였다. Emtor kinase and cell colony proliferation assays were performed using MCF7 cells overexpressing ALOX15.
실험 방법은 사용한 세포 및 13-HODE를 처리하지 않는 것을 제외하고는 실시예 5 및 실시예 7에 기재된 방식과 동일하다. 실험군은 야생형 ALOX15를 과발현하는 MCF7 세포를 사용하였으며, 대조군 및 비교군으로 각각 효소발현이 안되는 대조군 과발현 세포 및 효소활성이 상실된 돌연변이 ALOX15 T560M 세포를 사용하였다. The experimental method is the same as that described in Examples 5 and 7, except that the cells used and 13-HODE were not treated. For the experimental group, MCF7 cells overexpressing wild-type ALOX15 were used, and as a control group and a comparison group, a control overexpressing cell incapable of enzyme expression and a mutant ALOX15 T560M cell in which enzyme activity was lost were used as the control group.
실험 결과 야생형 ALOX15 과발현 JHEUM7 세포에서 S240/244, S473, T308의 인산화가 감소하는 것을 확인하였다. 즉, 엠토르 신호 전달 과정이 억제되는 것을 확인하였다. 이러한 효과는 ALO15 T560M 돌연변이 세포주에서는 관찰되지 않았다 (도 15a). 콜로니 성장분석을 통하여 야생형 ALOX15 발현 MCF7 세포주에서의 콜로니 성장 억제가 관찰되었으나 T560M 돌연변이 세포주에서는 콜로니 성장억제 효과가 없었다 (도 15b). As a result of the experiment, it was confirmed that phosphorylation of S240/244, S473, and T308 was decreased in wild-type ALOX15 overexpressing JHEUM7 cells. That is, it was confirmed that the emtor signal transduction process was inhibited. This effect was not observed in the ALO15 T560M mutant cell line ( FIG. 15A ). Colony growth inhibition was observed in the wild-type ALOX15-expressing MCF7 cell line through colony growth analysis, but there was no colony growth inhibitory effect in the T560M mutant cell line ( FIG. 15b ).
상기 결과는 13-HODE에 의해 엠토르 신호전달 과정 및 암 세포의 증식이 억제된다는 사실을 나타낸다. These results indicate that the emtor signaling process and the proliferation of cancer cells are inhibited by 13-HODE.
실시예 14: HEC59 세포를 이용한 세포의 엠토르 키나아제, 세포 생존력, 세포의 콜로니 증식 분석Example 14: Cellular Emtor kinase, Cell Viability, Cell Colony Proliferation Analysis Using HEC59 Cells
HEC59 세포를 이용하여 실시예 5, 실시예 6, 실시예 7에서 수행한 엠토르 키나아제, 세포 생존력, 세포 콜로니 증식 분석을 수행하였다. HEC59 세포는 암세포(자궁내막암 세포)이며, 엠토르 돌연변이 (mTORE1799K)를 갖는 세포이다. Emtor kinase, cell viability, and cell colony proliferation assays performed in Examples 5, 6, and 7 were performed using HEC59 cells. HEC59 cells are cancer cells (endometrial cancer cells) and are cells with the emtor mutation (mTOR E1799K ).
실험 방법은 사용한 세포를 제외하고는 실시예 5, 실시예 6, 실시예 7에 기재된 방식과 동일하다. The experimental method is the same as that described in Examples 5, 6, and 7 except for the cells used.
실험 결과, 13-HODE를 HEC59 세포에 처리하는 경우 T389 S6K1, S240/244, S473의 인산화를 감소되는 것을 확인, 즉 엠토르 신호전달 과정이 억제된다는 사실을 확인하였다(도 16a).As a result of the experiment, it was confirmed that when 13-HODE was treated in HEC59 cells, phosphorylation of T389 S6K1, S240/244, and S473 was reduced, that is, the emtor signaling process was inhibited ( FIG. 16a ).
또한, 13-HODE로 처리한 웰에서의 세포 증식율이 13-HODE를 처리하지 않는 웰에서의 증식율보다 현저히 낮다는 것을 확인하였다 (도 16b, 도 16c). In addition, it was confirmed that the cell proliferation rate in the wells treated with 13-HODE was significantly lower than that in the wells not treated with 13-HODE ( FIGS. 16B and 16C ).
상기 결과들은 13-HODE가 대장암, 유방암, 교모세포종, 자궁내막암, 간암, 췌장암 등 다양한 암에 항암 효과를 발휘한다는 점을 나타낸다. The above results indicate that 13-HODE exerts an anticancer effect on various cancers such as colorectal cancer, breast cancer, glioblastoma, endometrial cancer, liver cancer, and pancreatic cancer.

Claims (6)

13-HODE(13-Hydroxyoctadecadienoic acid)를 유효성분으로 포함하는 암의 예방 또는 치료용 약제학적 조성물. A pharmaceutical composition for preventing or treating cancer comprising 13-HODE (13-Hydroxyoctadecadienoic acid) as an active ingredient.
제 1 항에 있어서, 상기 암은 고형암인 것인, 암의 예방 또는 치료용 약제학적 조성물.The pharmaceutical composition for preventing or treating cancer according to claim 1, wherein the cancer is a solid cancer.
제 2 항에 있어서, 상기 고형암은 뇌종양, 양성성상세포종, 악성성상세포종, 뇌하수체 선종, 뇌수막종, 뇌림프종, 핍지교종, 상의세포종, 뇌간종양, 두경부 종양, 후두암, 구인두암, 비강/부비동암, 비인두암, 침샘암, 하인두암, 갑상선암, 구강암, 흉부종양, 소세포성 폐암, 비소세포성 폐암, 흉선암, 종격동 종양, 식도암, 유방암, 남성유방암, 복부종양, 위암, 간암, 담낭암, 담도암, 췌장암, 소장암, 대장암, 직장암, 항문암, 방광암, 신장암, 남성 생식기종양, 음경암, 전립선암, 여성생식기종양, 자궁경부암, 자궁내막암, 난소암, 자궁육종, 질암, 여성외부생 식기암, 여성요도암 및 피부암으로 이루어지는 군으로부터 선택되는 것인, 암의 예방 또는 치료용 약제학적 조성물.According to claim 2, wherein the solid cancer is brain tumor, benign astrocytoma, malignant astrocytoma, pituitary adenoma, meningioma, cerebral lymphoma, oligodendroglioma, ependymoma, brainstem tumor, head and neck tumor, laryngeal cancer, oropharyngeal cancer, nasal/sinus cancer, nasopharyngeal cancer , salivary gland cancer, hypopharyngeal cancer, thyroid cancer, oral cancer, chest tumor, small cell lung cancer, non-small cell lung cancer, thymus cancer, mediastinal tumor, esophageal cancer, breast cancer, male breast cancer, abdominal tumor, stomach cancer, liver cancer, gallbladder cancer, biliary tract cancer, pancreatic cancer, small intestine cancer Cancer, colorectal cancer, rectal cancer, anal cancer, bladder cancer, kidney cancer, male genital tumor, penile cancer, prostate cancer, female genital tumor, cervical cancer, endometrial cancer, ovarian cancer, uterine sarcoma, vaginal cancer, external genital cancer, A pharmaceutical composition for preventing or treating cancer, which will be selected from the group consisting of female urethral cancer and skin cancer.
제 1 항에 있어서, 상기 암은 혈액암인 것인, 암의 예방 또는 치료용 약제학적 조성물.The pharmaceutical composition for preventing or treating cancer according to claim 1, wherein the cancer is a blood cancer.
제 4 항에 있어서, 상기 혈액암은 급성골수구성백혈병, 급성림프구성 백혈병, 만성골수성백혈병, 만성림프구성백혈병, 급성단구성백혈병, 다발성 골수종, 호지킨림프종 및 비호지킨 림프종으로 이루어진 군으로부터 선택되는 것인, 암의 예방 또는 치료용 약제학적 조성물. 5. The method of claim 4, wherein the hematologic cancer is selected from the group consisting of acute myelocytic leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, acute monocytic leukemia, multiple myeloma, Hodgkin's lymphoma and non-Hodgkin's lymphoma. The pharmaceutical composition for the prevention or treatment of cancer.
13-HODE를 유효성분으로 포함하는 약제학적 조성물을 대상체(subject)에 투여하는 단계를 포함하는 암의 예방 또는 치료방법.A method for preventing or treating cancer, comprising administering to a subject a pharmaceutical composition comprising 13-HODE as an active ingredient.
PCT/KR2021/003708 2020-03-25 2021-03-25 Pharmaceutical composition, for prevention or treatment of cancer, comprising 13-hydroxyoctadecadienoic acid as active ingredient WO2021194276A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20200036500 2020-03-25
KR10-2020-0036500 2020-03-25

Publications (1)

Publication Number Publication Date
WO2021194276A1 true WO2021194276A1 (en) 2021-09-30

Family

ID=77892388

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2021/003708 WO2021194276A1 (en) 2020-03-25 2021-03-25 Pharmaceutical composition, for prevention or treatment of cancer, comprising 13-hydroxyoctadecadienoic acid as active ingredient

Country Status (2)

Country Link
KR (1) KR20210120890A (en)
WO (1) WO2021194276A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20020091399A (en) * 2001-05-30 2002-12-06 조윤희 Metabolites of essential unsaturated fatty acids exerting anti-proliferative effect on human breast cancer cells
JP2013116862A (en) * 2011-12-02 2013-06-13 National Institute Of Advanced Industrial Science & Technology Antibody specific to hode, diagnostic method of disease originated from oxidation stress, kit, hybridoma, and immunological detection method
WO2019199673A1 (en) * 2018-04-09 2019-10-17 President And Fellows Of Harvard College Modulating nuclear receptors and methods of using same

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20020091399A (en) * 2001-05-30 2002-12-06 조윤희 Metabolites of essential unsaturated fatty acids exerting anti-proliferative effect on human breast cancer cells
JP2013116862A (en) * 2011-12-02 2013-06-13 National Institute Of Advanced Industrial Science & Technology Antibody specific to hode, diagnostic method of disease originated from oxidation stress, kit, hybridoma, and immunological detection method
WO2019199673A1 (en) * 2018-04-09 2019-10-17 President And Fellows Of Harvard College Modulating nuclear receptors and methods of using same

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KODAKA ATSUSHI, HAYAKAWA YUKI, ALSAYEGH RAWAA JAFFAR, YASUHARA TADASHI, TOMODA HIROSHI, OKU TERUAKI, DAN SHINGO, TSUIJI MAKOTO, TS: "Stereoisomer-Specific Induction of G2/M Phase Arrest and Apoptosis by 9-(<i>E</i>,<i>Z</i>)-Hydroxyoctadecadienoic Acid in Mouse Lymphoma Cells", BIOLOGICAL & PHARMACEUTICAL BULLETIN, vol. 42, no. 6, 1 June 2019 (2019-06-01), JP, pages 937 - 943, XP055853490, ISSN: 0918-6158, DOI: 10.1248/bpb.b18-00935 *
TAVAKOLI-TARAKI MASOUMEH, FATEMEH, KARAMI-TEHRANI: "Apoptosis Induced by 13-S-hydroxyoctadecadienoic acid in the Breast Cancer Cell Lines, MCF-7 and MDA-MB-231", IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES, vol. 16, no. 4, 1 January 2013 (2013-01-01), pages 661 - 667, XP055853461 *

Also Published As

Publication number Publication date
KR20210120890A (en) 2021-10-07

Similar Documents

Publication Publication Date Title
Cohen et al. Acetylation of the C terminus of Ku70 by CBP and PCAF controls Bax-mediated apoptosis
AU2021107577A4 (en) Flavagline derivatives for inhibition of kras oncogene activation
WO2021107644A1 (en) USE OF COMPOSITION FOR ENHANCING ANTICANCER EFFECT, COMPRISING ERRγ INHIBITOR AS ACTIVE INGREDIENT
US9249153B2 (en) Pharmaceutical composition for treating aging-associated diseases, containing progerin expression inhibitor as active ingredient, and screening method of said progerin expression inhibitor
WO2016018088A1 (en) Novel biomarker for predicting sensitivity to met inhibitor, and use thereof
WO2016003158A2 (en) New compound for inhibiting binding between dx2 protein and p14/arf protein, and pharmaceutical composition for treating or preventing cancer disease containing same as effective ingredient
WO2021194276A1 (en) Pharmaceutical composition, for prevention or treatment of cancer, comprising 13-hydroxyoctadecadienoic acid as active ingredient
Lu et al. The protective effect of isoflurane pretreatment on liver IRI by suppressing noncanonical pyroptosis of liver macrophages
WO2014157965A1 (en) Composition for treatment or metastasis suppression of cancers which includes p34 expression inhibitor or activity inhibitor as active ingredient
WO2017043779A1 (en) Stk32c gene relevant to breast cancer and use thereof
WO2023140664A1 (en) Pharmaceutical composition for preventing or treating liver fibrosis induced by non-alcoholic steatohepatitis, containing, as active ingredient, expression inhibitor of lipocalin 2 or receptor thereof
Jiang et al. Multifunctions of CRIF1 in cancers and mitochondrial dysfunction
WO2011090283A2 (en) Composition containing inhibitors of the expression or activity of sh3rf2 for preventing or treating cancer
WO2017048069A1 (en) Novel pyrazoline derivative and use thereof
WO2020076124A1 (en) Pharmaceutical composition for preventing or treating cancer, containing malate-aspartate shuttle inhibitor and carnitine acylcarnitine carrier shuttle inhibitor
WO2024085603A1 (en) Composition for preventing or treating c-myc-associated diseases through inhibition of expression of ubiquitin-specific protease 47 (usp47), and use thereof
WO2017164486A1 (en) Composition for inhibiting resistance to anticancer agents, containing tesk1 inhibitor, and method for screening for tesk1 inhibitor
WO2023163301A1 (en) Peptide that inhibits formation of kitenin dimer, and use for same
WO2023018168A1 (en) Composition for preventing or treating cancer comprising msh2 or msh6 inhibitor as active ingredient
WO2020242225A1 (en) COMPOSITION FOR PREVENTING OR TREATING PROTEIN CONFORMATIONAL DISORDERS, COMPRISING IRE1α KINASE ACTIVATOR AS ACTIVE INGREDIENT
WO2023068835A1 (en) Pak4 inhibitor and uses thereof
WO2021107730A1 (en) Synthetic-baicalein-derivative as chemotherapeutic agent
WO2022177121A1 (en) Prohibitin companion diagnostic composition for leukemia targeted treatment
WO2022169342A2 (en) Pharmaceutical composition for inhibiting cancer metastasis
WO2023167544A1 (en) Biomarker comprising fam167a for diagnosis of resistance against bcr-abl-independent tyrosine kinase inhibitor, and composition targeting fam167a for preventing or treating chronic myeloid leukemia

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: 21774342

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: 21774342

Country of ref document: EP

Kind code of ref document: A1