[go: up one dir, main page]

WO2023172032A1 - Composition for preventing or treating cancer comprising nmur2 inhibitor as active ingredient - Google Patents

Composition for preventing or treating cancer comprising nmur2 inhibitor as active ingredient Download PDF

Info

Publication number
WO2023172032A1
WO2023172032A1 PCT/KR2023/003111 KR2023003111W WO2023172032A1 WO 2023172032 A1 WO2023172032 A1 WO 2023172032A1 KR 2023003111 W KR2023003111 W KR 2023003111W WO 2023172032 A1 WO2023172032 A1 WO 2023172032A1
Authority
WO
WIPO (PCT)
Prior art keywords
nmur2
cancer
preventing
pharmaceutical composition
inhibitor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
PCT/KR2023/003111
Other languages
French (fr)
Korean (ko)
Inventor
한태수
정은선
김장성
반현승
노유나
황진성
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Korea Research Institute of Bioscience and Biotechnology KRIBB
Original Assignee
Korea Research Institute of Bioscience and Biotechnology KRIBB
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 Korea Research Institute of Bioscience and Biotechnology KRIBB filed Critical Korea Research Institute of Bioscience and Biotechnology KRIBB
Publication of WO2023172032A1 publication Critical patent/WO2023172032A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the present invention relates to a composition for preventing or treating cancer.
  • Cancer has a high mortality rate worldwide and is the most common cause of death in Western societies after cardiovascular disease.
  • colon cancer, breast cancer, and prostate cancer are continuously increasing due to the aging of the population, the generalization of high-fat diet due to westernization of diet, rapid increase in environmental pollutants, and increase in alcohol consumption.
  • anticancer substances that can prevent and treat cancer and contribute to the improvement of human health, healthy quality of life, and human health.
  • brain and central nervous system cancers are ranked 3rd in men and 2nd in women, and 3rd among men and women among the crude incidence rates for the 0 to 14-year-old group, according to national cancer registration statistics.
  • Brain tumors occur in approximately 3,000 people per year in Korea. Benign brain tumors can be cured through curative surgery, but malignant brain tumors are treated with a combination of surgery, radiation therapy, and chemotherapy.
  • Commonly occurring brain tumors include glioma, meningoencephalopathy, pituitary adenoma, metastatic brain tumor, and acoustic schwannoma.
  • glioma glioblastoma
  • glioblastoma is known to be the most common malignant tumor, grade 4 in the WHO brain tumor classification, and has an incidence rate of 1 It is said to be about 3 to 4 per 100,000 people per year.
  • Treatment methods for brain tumors include surgery, radiation therapy, and chemotherapy. Generally, chemotherapy is used after removing the cancer through surgery and radiation therapy.
  • the first-line chemotherapy treatment for brain tumors includes Temozolomide, which alkylates or methylates intracellular DNA, and the second-line drug is Avastin-irinotecan. Irinotecan).
  • One object of the present invention is to provide a pharmaceutical composition for preventing or treating cancer.
  • Another object of the present invention is to provide a method for screening agents for treating cancer.
  • the present invention provides a pharmaceutical composition for preventing or treating cancer containing an NMUR2 inhibitor as an active ingredient.
  • the NMUR2 inhibitor may be an NMUR2 expression inhibitor or an NMUR2 activity inhibitor.
  • the NMUR2 inhibitor may be a compound having the following formula (I) or a pharmaceutically acceptable salt thereof.
  • the cancer may be one in which NMUR2 is overexpressed.
  • the cancer may be a brain tumor.
  • the present invention includes the steps of treating cells overexpressing NMUR2 with an anticancer drug candidate; and measuring the expression or activity level of NMUR2 in cells treated with the candidate substance.
  • NMUR2 which is targeted by the pharmaceutical composition of the present invention, is a novel target that has not been previously known as a target for the prevention or treatment of cancer.
  • the pharmaceutical composition of the present invention which contains an NMUR2 expression or activity inhibitor as an active ingredient, is a conventional anticancer agent. It has the advantage of being able to be used as an alternative anticancer agent in cases of refractoriness or resistance.
  • NMUR2 activity inhibitors have a significantly lower IC 50 value compared to temozolomide, a conventional anticancer drug, and can be used alone as an effective ingredient for the prevention or treatment of cancer.
  • the pharmaceutical composition of the present invention when used in combination with conventional anticancer drugs, exhibits a synergistic effect compared to when each is used alone, so it has the advantage of being able to be used as an anticancer adjuvant.
  • NMUR2 expression inhibitors can inhibit cancer cell proliferation and cell migration, promote cancer cell death, and inhibit metastasis, so they can be used as effective anticancer agents.
  • Figures 1A, 1B, and 1C show the structure of the pcDNA3.1 plasmid vector expressed by the CMV promoter and inserted with the base sequence encoding NMUR2 and the base sequence encoding IRES-GFP, the bright field and fluorescence of GFP expression, and the expression of the vector in the HTLA cell line.
  • This is a graph showing the intensity of luciferase when introduced into and treated with NMU-25, the ligand of NMUR2.
  • Figure 2a is a graph showing the intensity of luciferase when the HTLA cell line was treated with NMU-25 and the compound of Formula I.
  • Figure 2b is a graph showing the intensity of luciferase when the HTLA cell line was treated with NMU-25 and the compound of Formula I at various concentrations.
  • Figures 3A and 3B are graphs measuring the IC 50 of temozolomide and the compound of Formula I against brain tumor cell lines U-87MG, T-98G, and U-373MG, and graphs showing the activity of caspase 3/7 according to concentration. .
  • Figures 4A and 4B are graphs confirming the OD450, caspase 3/7 activity, and CI values when temozolomide and the compound of Formula I are co-treated.
  • Figure 5 is a graph confirming IC 50 when gastric cancer, colon cancer, and skin cancer cell lines SNU-638, HCT116, and SK-MEL-2 were treated with the compound of Formula I.
  • Figure 6 is a diagram analyzing the correlation between genetic mutations and NMUR2 expression in glioblastoma and glioma samples.
  • Figure 7 is a diagram confirming that, unlike the glioblastoma cell line into which the pcDNA empty vector was introduced, the glioblastoma cell line into which the pcDNA3.1 NMUR2-overexpression vector was introduced expresses NMUR2 at a high level.
  • Figure 8 is a diagram confirming that the proliferation rate of glioblastoma cells in the NMUR2 overexpressing cell line is increased compared to the control group.
  • Figure 9 is a diagram confirming that the migration of glioblastoma cells in the NMUR2 overexpressing cell line is increased compared to the control group.
  • Figure 10 is a diagram confirming that the expression of NMUR2 was significantly reduced in glioblastoma cell lines treated with siRNA against NMUR2.
  • Figure 11 is a diagram confirming that the proliferation rate of cells in glioblastoma cell lines treated with siRNA against NMUR2 was reduced compared to the control group.
  • Figure 12 is a diagram confirming the decrease in cell migration in glioblastoma cell lines treated with siRNA against NMUR2 compared to the control group.
  • Figure 13 is a diagram confirming that the compound of Formula 1, a compound that inhibits NMUR2, inhibited cell migration of a glioblastoma cell line.
  • composition for the treatment or prevention of cancer 1.
  • One aspect of the present invention provides a pharmaceutical composition for treating or preventing cancer.
  • the pharmaceutical composition for treating or preventing cancer of the present invention contains an inhibitor of NMUR2 (Neuromedin U Receptor 2; NCBI GenBank ID: 56923) as an active ingredient.
  • NMUR2 Neuropeptide U Receptor 2
  • GPCR G-protein coupled receptor
  • the NMUR2 inhibitor may be an NMUR2 expression inhibitor or an NMUR2 activity inhibitor.
  • NMUR2 expression inhibitor or NMUR2 activity inhibitor is used to collectively refer to all agents that reduce the expression or activity of NMUR2, and specifically, NMUR2 at the transcription, mRNA, or translation level. It may include any agent that inhibits the activity of NMUR2 by suppressing its expression by reducing the expression level of the gene or by interfering with the activity of the NMUR2 protein.
  • NMUR2 expression inhibitor or NMUR2 activity inhibitor can be used without limitation in its form, such as a compound, nucleic acid, peptide, virus, or vector containing the nucleic acid that can inhibit the expression or activity of NMUR2 by targeting NMUR2.
  • the NMUR2 expression inhibitor may be a miRNA, siRNA or shRNA, or an antisense oligonucleotide that binds complementary to the mRNA of the NMUR2 gene.
  • it may be siRNA
  • the NMUR2 activity inhibitor is specific for the NMUR2 protein. It may be an aptamer, antibody, or low-molecular-weight compound that binds to.
  • miRNA nucleic acid molecules that can mediate RNA interference or gene silencing, and can inhibit the expression of target genes, thereby producing efficient gene knockdown. down) method or gene therapy method.
  • miRNA, siRNA, and shRNA can inhibit NMUR2 by acting specifically on NMUR2 to cleave the NMUR2 mRNA molecule and induce RNA interference (RNAi).
  • miRNAs, siRNAs and shRNAs can be synthesized chemically or enzymatically.
  • the siRNA may include the nucleotide sequences represented by SEQ ID NO: 5 and SEQ ID NO: 6, or the nucleotide sequences represented by SEQ ID NO: 7 and SEQ ID NO: 8.
  • the term “aptamer” refers to a single-stranded oligonucleotide, about 20 to 60 nucleotides in size, and a nucleic acid molecule that has binding activity to a given target molecule. It has a variety of three-dimensional structures depending on its sequence, and can have high affinity for specific substances, like an antigen-antibody reaction.
  • An aptamer can inhibit the activity of a certain target molecule by binding to it.
  • the aptamer of the present invention may be RNA, DNA, modified nucleic acid, or a mixture thereof, and may also be in a linear or circular form.
  • the aptamer may bind to NMUR2 and serve to inhibit the activity of NMUR2.
  • Such an aptamer can be prepared from the sequence of NMUR2 by a method known to those skilled in the art.
  • an antibody refers to a polypeptide that includes at least one immunoglobulin variable region sequence and specifically binds to a given antigen.
  • an antibody may comprise a heavy (H) chain variable region and a light (L) chain variable region.
  • the term "low molecular weight compound” is a natural or synthetic compound with a molecular weight of 1000 Da or less, such as 800 Da or less, especially 500 Da or less, and may bind to the NMUR2 protein and interfere with the activity of NMUR2.
  • the low molecular weight compound may be a compound having the formula of ⁇ Formula I> below or a pharmaceutically acceptable salt thereof.
  • salt refers to inorganic and organic acid addition salts of the compound.
  • the “pharmaceutically acceptable salt” may be a salt that does not cause serious irritation to the organism to which the compound is administered and does not impair the biological activity and physical properties of the compound, and may be an inorganic acid salt, an organic acid salt, or a metal salt.
  • the inorganic acid salt may be hydrochloride, bromate, phosphate, sulfate, or disulfate.
  • the organic acid salts include formate, acetate, propionate, lactate, oxalate, tartrate, malate, maleate, citrate, fumarate, besylate, camsylate, edicyl salt, trichloroacetic acid, and trifluoroacetate. , benzoate, gluconate, methanesulfonate, glycolate, succinate, 4-toluenesulfonate, galacturonate, embonate, glutamate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, or aspart. It may be an acid salt.
  • the metal salt may be a calcium salt, sodium salt, magnesium salt, strontium salt, or potassium salt.
  • cancer refers to colorectal cancer, including colon cancer and rectal cancer, breast cancer, uterine cancer, cervical cancer, ovarian cancer, prostate cancer, head and neck carcinoma, melanoma, myeloma, leukemia, lymphoma, stomach cancer, lung cancer, pancreatic cancer, liver cancer, Esophageal cancer, small intestine cancer, perianal cancer, fallopian tube carcinoma, endometrial carcinoma, vaginal carcinoma, vulvar carcinoma, Hodgkin's disease, bladder cancer, kidney cancer, ureteral cancer, renal cell carcinoma, renal pelvic carcinoma, bone cancer, skin cancer, head cancer, and neck cancer.
  • cutaneous melanoma intraocular melanoma, endocrine cancer, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, or central nervous system (CNS) cancer, such as colon cancer, stomach cancer, and lung cancer.
  • CNS central nervous system
  • it may be pancreatic cancer, liver cancer, small intestine cancer, skin cancer, or central nervous system cancer, especially central nervous system cancer.
  • the central nervous system cancer may be a brain tumor.
  • the “brain tumor” refers to any tumor that occurs within the skull and includes all tumors that occur in the brain and structures surrounding the brain.
  • the brain tumor may be glioma, meningoencephalopathy, pituitary adenoma, metastatic brain tumor, acoustic schwannoma, glioblastoma, etc., but is not limited thereto.
  • the cancer may be one in which NMUR2 is overexpressed.
  • the pharmaceutical composition of the present invention contains an NMUR2 inhibitor as an active ingredient, it can exhibit a particularly excellent prevention or treatment effect against cancer in which NMUR2 is overexpressed.
  • the term “treatment” refers to clinical intervention to alter the natural processes of the individual or cell being treated, which may be performed during the course of the clinical pathology or to prevent it.
  • the desired therapeutic effects include preventing the occurrence or recurrence of the disease, alleviating symptoms, reducing all direct or indirect pathological consequences of the disease, preventing metastasis, reducing the rate of disease progression, and alleviating the disease state. or temporarily alleviating, remission, or improving prognosis.
  • the present invention includes all actions to improve the course of cancer by administering a composition containing a substance that inhibits NMUR2.
  • prevention refers to all actions that inhibit or delay the onset of cancer by administering a composition containing a substance that inhibits NMUR2 according to the present invention.
  • the pharmaceutical composition of the present invention may further include appropriate carriers, excipients, or diluents commonly used in the preparation of pharmaceutical compositions.
  • composition containing a pharmaceutically acceptable carrier may be in various oral or parenteral dosage forms. When formulated, it can be prepared using diluents or excipients such as commonly used fillers, extenders, binders, wetting agents, disintegrants, and surfactants.
  • Solid preparations for oral administration may include tablets, powders, granules, capsules, etc., and such solid preparations may contain one or more compounds and at least one excipient, such as starch, calcium carbonate, sucrose, or lactose. It can be prepared by mixing (lactose), gelatin, etc. Additionally, in addition to simple excipients, lubricants such as magnesium stearate, talc, etc. may also be used.
  • Liquid preparations for oral administration include suspensions, oral solutions, emulsions, and syrups. In addition to the commonly used simple diluents such as water and liquid paraffin, various excipients such as wetting agents, sweeteners, fragrances, and preservatives may be included. there is.
  • Preparations for parenteral administration may include sterilized aqueous solutions, non-aqueous solutions, suspensions, emulsions, freeze-dried preparations, and suppositories.
  • Non-aqueous solvents and suspensions may include propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable ester such as ethyl oleate.
  • injectable ester such as ethyl oleate.
  • As a base for suppositories witepsol, macrogol, tween 61, cacao, laurel, glycerogelatin, etc. can be used.
  • the pharmaceutical composition of the present invention is not limited thereto, but includes tablets, pills, powders, granules, capsules, suspensions, oral solutions, emulsions, syrups, sterilized aqueous solutions, non-aqueous solutions, suspensions, emulsions, and freezing. It may have any one formulation selected from the group consisting of dry formulations.
  • the pharmaceutical composition of the present invention may be administered in a daily dose of about 0.0001 mg/kg to about 10 g/kg, and may be administered in a daily dosage of about 0.001 mg/kg to about 1 g/kg.
  • the dosage may vary depending on the degree of purification of the mixture, the patient's condition (age, gender, weight, etc.), and the severity of the condition being treated.
  • the total daily dose may be divided and administered several times during the day, if necessary.
  • the compound of formula (I) is an NMUR2 inhibitor that inhibits the activity of NMUR2 (see Figure 2), and it was confirmed that the conventional brain tumor cell lines U-87MG, T-98G, and U-373MG were used as brain tumor cell lines. It was confirmed that it exhibited a much better therapeutic effect than the therapeutic agent temozolomide (see Figures 3A and 4A).
  • the compound of Formula I in order to confirm the effect of the compound of Formula I on other cancer types, the compound of Formula I was administered alone to SNU-638, HCT116, and SK-MEL-2, which are gastric cancer, colon cancer, and skin cancer cell lines, respectively. and was confirmed to have an IC 50 similar to that of brain tumor cell lines.
  • Another aspect of the present invention provides a kit for preventing or treating cancer.
  • the kit for preventing or treating cancer of the present invention may include the pharmaceutical composition and an existing anticancer agent.
  • the kit for preventing or treating cancer of the present invention may additionally include a carrier, diluent, excipient, or a combination of two or more commonly used in pharmaceutical compositions.
  • the kit may further include a notice in a form established by a government agency for regulating the manufacture, use, or sale of pharmaceuticals or biological products.
  • composition for treating or preventing cancer ' Since the pharmaceutical composition is the same as described in ' 1. Pharmaceutical composition for treating or preventing cancer ', further description is omitted.
  • the “existing anticancer agent” refers to a chemotherapy treatment agent that has been previously used to inhibit the proliferation of cancer cells.
  • the existing anticancer agent as described above may be an anticancer agent targeting a target other than NMUR2, for example, temozolomide or Avastin-irinotecan, and especially temozolomide.
  • the pharmaceutical composition contains an NMUR2 inhibitor as an active ingredient, it can further increase the anticancer effect by acting on a different target than the existing anticancer agent.
  • the pharmaceutical composition and the existing anticancer agent may be administered simultaneously or sequentially.
  • the pharmaceutical composition and the existing anticancer drug are administered sequentially, the pharmaceutical composition may be administered first and then the existing anticancer drug may be administered later, or the existing anticancer drug may be administered first and then the pharmaceutical composition. It may also be administered later. Therefore, the kit may further include an instruction manual containing instructions on the above administration method.
  • the cancer is the same as described in ‘ 1.
  • Pharmaceutical composition for treating or preventing cancer ’ may overexpress NMUR2, and may further be refractory or resistant to the existing anticancer drugs.
  • Another aspect of the present invention provides a method for screening anticancer drugs.
  • the anticancer drug screening method of the present invention includes treating cells overexpressing NMUR2 with an anticancer drug candidate; And measuring the expression or activity level of NMUR2 in cells treated with the candidate substance.
  • the “anticancer drug candidate” refers to a substance expected to exhibit an effect of inhibiting the proliferation of cancer cells.
  • the anticancer drug candidate may be treated at a concentration of 1 ⁇ M to 40 ⁇ M.
  • the anticancer drug candidate may be treated at a concentration of 5 ⁇ M, 10 ⁇ M, 15 ⁇ M, 20 ⁇ M, 25 ⁇ M, 30 ⁇ M, or 35 ⁇ M.
  • expression level measurement may refer to the process of confirming the presence and expression level of mRNA of a gene, or the presence and expression level of protein expressed from the gene.
  • the presence or expression level of mRNA of the gene can be confirmed by measuring the amount of mRNA of the gene. Analysis methods for this include RT-PCR and competitive RT-PCR using primer pairs, probes, or anti-sense nucleotides for the mRNA of NMUR2. PCR), real-time RT-PCR, RNase protection assay (RPA), Northern blotting, DNA chip, etc., but are not limited to these.
  • RPA RNase protection assay
  • Northern blotting DNA chip, etc.
  • Analysis methods for this include Western blot, ELISA (enzyme linked immunosorbent assay), RIA (Radioimmunoassay), radioimmunodiffusion, and Ouktero using an antibody that specifically binds to the NMUR2 protein.
  • Ouchterlony immunodiffusion method, rocket immunoelectrophoresis, tissue immunostaining, immunoprecipitation assay, complement fixation assay, FACS, protein chip, etc. are limited to these. It doesn't work.
  • the “activity level measurement” is a process of confirming whether a protein expressed from a gene is active, and is accomplished by confirming whether and to what extent the protein is functional.
  • the anticancer drug screening method may further include determining the candidate substance as an anticancer drug when the measured expression or activity level of NMUR2 is significantly lower than that of cells not treated with the candidate substance.
  • the "significantly low case” means that the expression or activity level of NMUR2 is lower by 10% or more compared to cells not treated with the candidate substance, such as 20% or more, 30% or more, 40% or more, 50% or more. It may be above or below 60%, and especially may be below 70%.
  • the screening method can be performed in vivo or in vitro, and is not particularly limited. Candidate substances may be known substances or new substances, and large-scale screening can be performed through, for example, plant extracts or chemical libraries. Through this, it is possible to discover agents that can suppress cancer, especially brain tumors, by inhibiting the expression or activity of NMUR2.
  • a cell line into which an NMUR2 expression vector was introduced was constructed.
  • an NMUR2-IRES-GFP expression vector was constructed by inserting a base sequence encoding NMUR2 and a base sequence encoding IRES-GFP into the pcDNA3.1 plasmid vector expressed by the CMV promoter, and expressing the NMUR2-IRES-GFP.
  • HTLA_NMUR2-IRES-GFP cells were created by transducing the vector into the PRESTO-TANGO HTLA cell line, which expresses luciferase in a manner dependent on beta-arrestin signaling activity.
  • a cell line into which the expression vector was successfully transduced was secured.
  • the obtained cell line was treated with NMU-25, a NMUR2 ligand, and 6 hours later, the activity of luciferase was observed to increase, confirming that the activity of NMUR2 was increased, and a drug screening cell line selective for NMUR2 was created.
  • the cell lines into which the expression vector was successfully transduced were cultured at a density of 20,000 cells/well in a 5% CO 2 incubator at 37°C for 24 hours. Afterwards, 10 ⁇ l of DMSO (100 ⁇ M) and the compound of Formula I (100 ⁇ M) diluted in DPBS (Dulbecco's Phosphate Buffered Saline) were treated so that the final concentration was 10 ⁇ M, or the compound of Formula I was treated at 0.1 and 1, respectively. After diluting to 10, 50, 100, and 200 ⁇ M, 10ul was applied per well so that the final concentrations were 0, 0.01, 1, 5, 10, and 20 ⁇ M, respectively. 15 minutes after treatment with DMSO and the compound of Formula I, 10 ⁇ l of NMU-25, a ligand for NMUR2, was treated to a final concentration of 20 nM.
  • DPBS Dens Phosphate Buffered Saline
  • luciferase activity solution (One-glo, Promega) was added at a density of 50 ⁇ l/well, reacted at room temperature for 5 minutes, and then luciferase Activity was measured.
  • the luciferase activity significantly increased in the group treated with DMSO and NMU-25, a ligand of NMUR2, while the group treated simultaneously with NMU-25, a ligand of NMUR2, and the compound of Formula I showed luciferase activity. No change in rase activity was observed, confirming that the compound of Formula I inhibits NMUR2 (see Figure 2a).
  • the degree of inhibition of NMUR2 by the compound of Formula I in the experimental group repeatedly showed a high inhibition effect of more than 70%.
  • the IC 50 of the compound of Formula I and temozolomide was confirmed in brain tumor cell lines U-87MG, T-98G, and U-373MG (Korea Cell Line Bank).
  • the IC 50 of temozolomide was confirmed to be 1000 ⁇ M or more, showing a minimal inhibitory effect on brain tumor cells even at high concentrations, while the IC 50 of the compound of Formula I was confirmed to be 6 to 7 ⁇ M.
  • the compound of Formula I can inhibit brain tumor cell growth even when used alone at a lower concentration than temozolomide, which is currently used as a brain tumor treatment (see Figure 3A).
  • Temozolomide IC 50 ( ⁇ M) Compound IC of Formula I 50 ( ⁇ M) U-87MG 14621 6.412 T-98G 1666 6.160 U-373MG 2630 7.994
  • U-87MG and U-373MG were placed in a 96-well white plate at a density of 8,000 cells/well each, and T-98G was placed at a density of 5,000 cells/well, and then cultured in an incubator at 37°C and 5% CO 2 for 24 hours, and the chemical formula Compound I was treated at concentrations of 0, 5, and 10 ⁇ M, respectively. After 48 hours, the cells were treated with 30 ⁇ l of Caspase 3/7-Glo (Promega) solution, waited for 30 minutes at room temperature and in the dark, and changes in cell death rate were measured. Significant induction of cell death was observed when treated with 10 ⁇ M compound of formula I in all three brain tumor cell lines (see Figure 3B).
  • T-98G a brain tumor cell line
  • temozolomide and the compound of formula (I) were treated alone or in combination.
  • the final concentration of the compound of Formula I was When treated alone at a concentration of 10 ⁇ M, the cell growth rate and cell death rate increased. In addition, higher cell growth and cell death rates were observed when treated in combination with temozolomide at the same dose as above, compared to when the compound of Formula I was treated alone (see Figure 4A).
  • the brain tumor cell line T-98G was cultured in a 96-well plate at 3000 cells/well for 24 hours, and then temozolomide was added at a concentration of 0. to 1000 mM and simultaneously treated with the compound of formula I at a concentration of 1 to 20 ⁇ M. The degree to which they influence each other when diluted to 1/2 of the highest concentration was calculated as the value of the combination index.
  • SNU-638, HCT116, and SK-MEL-2 which are cell lines for stomach cancer, colon cancer, and skin cancer, were cultured in a 96-well plate at a density of 3000 cells/well for 24 hours in a 5% CO2 incubator at 37°C, and then HCT116 and SK-MEL-2 was treated with a compound of formula I (final concentration 0 ⁇ 20 ⁇ M) diluted to 1/2 of the highest concentration, and for SNU-638, a compound of formula I (final concentration 0 ⁇ 100 ⁇ M) was treated. It was diluted to 1/2 of the highest concentration and treated with 10 ⁇ l each. After culturing for 48 hours after drug treatment, cell growth rate was measured using the CCK8 kit according to the manufacturer's instructions.
  • IC 50 values were similar to those in brain tumor cell lines (see Figure 5).
  • TCGA Cancer Genome Atlas
  • a cell line into which an NMUR2 expression vector was introduced was created.
  • nucleotide sequence encoding NMUR2 was inserted into the pcDNA3.1 plasmid vector to construct a pcDNA3.1 NMUR2-overexpression vector, and the pcDNA empty vector and pcDNA3.1 NMUR2-overexpression vector were added to the glioblastoma cell line T-98G, respectively. was transduced using Lipofectamine 2000 (Thermo Scientific), and after RNA extraction and cDNA synthesis 72 hours later, overexpression of NMUR2 was confirmed by qPCR.
  • the nucleotide sequences of the primers used in qPCR are shown in Table 4 below.
  • the glioblastoma cell line into which the pcDNA empty vector was introduced and the glioblastoma cell line into which the pcDNA3.1 NMUR2-overexpression vector was introduced were distributed in a 96-well plate at a density of 3000 cells/well and incubated at 37°C with 5% CO 2 . After 24, 48, 72, and 96 hours of culture, cell proliferation was analyzed using the CCK8 kit (Dojindo Molecular Technologies, Inc.).
  • the glioblastoma cell line introduced with the pcDNA empty vector and the glioblastoma cell line introduced with the pcDNA3.1 NMUR2-overexpression vector were placed in the upper chamber of an 8 ⁇ m 24-well 'insert' (FALCON) at 5x10 cells each in 500 ⁇ l of serum-free medium. It was put in with. The lower chamber was filled with 500 ⁇ l of medium containing 10% FBS. Afterwards, the transwell was cultured at 37°C and 5% CO 2 for 18 hours, fixed with formaldehyde diluted to 4% using distilled water and 100% methanol, and then diluted to 0.1% using 20X PBS (LPS solution). Stained with crystal violet (SIGMA).
  • siNMUR2-1 and siNMUR2-2 were transduced into T-98G, a glioblastoma cell line, respectively, using lipofectamineRNAiMAX (Thermo Scientific) (Table 5 below). ), 72 hours later, qPCR was performed using the method described in Example 7 above to measure the expression level of NMUR2.
  • siRNA against NMUR2 inhibits the growth of glioblastoma and can be used as a preventive or therapeutic agent for glioblastoma.
  • the cell lines transduced with siNMUR2-1 and siNMUR2-2, respectively, into T-98G, a glioblastoma cell line were placed in the upper chamber of an 8 ⁇ m 24-well 'insert' (FALCON), 5x104 each, along with 500 ⁇ l of serum-free medium. I put it in.
  • the lower chamber was filled with 500 ⁇ l of medium containing 10% FBS.
  • the transwell was cultured at 37°C and 5% CO 2 for 18 hours, fixed with formaldehyde diluted to 4% using distilled water and 100% methanol, and then diluted to 0.1% using 20X PBS (LPS solution). Stained with crystal violet (SIGMA).
  • siRNA against NMUR2 can inhibit the migration of glioblastoma and metastasis of cancer, and can be used as a preventive or therapeutic agent for glioblastoma.
  • glioblastoma cell lines U-87MG, T-98G, and U-373MG were placed in an upper chamber at 1x10 cells each with 500 ⁇ l of serum-free medium, and the concentration of the compound of Formula 1 in the medium was 0 (DMSO). , the compound of Formula 1 was treated to concentrations of 5, 10, and 20 ⁇ M.
  • the lower chamber was filled with 500 ⁇ l of medium containing 10% FBS.
  • the transwell was cultured at 37°C and 5% CO 2 for 20 hours, fixed with formaldehyde diluted to 4% using distilled water and 100% methanol, and then diluted to 0.1% using 20X PBS (LPS solution). Stained with crystal violet (SIGMA).
  • the compound of Formula 1 which is an inhibitor for NMUR2, can inhibit the migration of glioblastoma and inhibit cancer metastasis, and can be used as a preventive or therapeutic agent for glioblastoma.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Urology & Nephrology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Hematology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Cell Biology (AREA)
  • Biophysics (AREA)
  • Food Science & Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Physics & Mathematics (AREA)
  • General Engineering & Computer Science (AREA)
  • Toxicology (AREA)
  • Tropical Medicine & Parasitology (AREA)

Abstract

The present application relates to a pharmaceutical composition for preventing or treating cancer comprising an NMUR2 expression inhibitor or an NMUR2 activity inhibitor as an active ingredient. A pharmaceutical composition of the present invention comprising an NMUR2 inhibitor according to the present invention as an active ingredient is advantageous in that the composition can be used as an alternative anticancer agent in cases where unresponsiveness or resistance to conventional anticancer agents is exhibited.

Description

NMUR2 억제제를 유효성분으로 포함하는 암의 예방 또는 치료용 조성물Composition for preventing or treating cancer containing an NMUR2 inhibitor as an active ingredient

[관련 출원과의 상호 인용][Cross-citation with related applications]

본 출원은 2022년 03월 08일에 출원된 한국 특허출원 제2022-0029570호에 기초한 우선권의 이익을 주장하며, 해당 문헌에 개시된 모든 내용은 본 명세서의 일부로서 포함된다.This application claims the benefit of priority based on Korean Patent Application No. 2022-0029570 filed on March 8, 2022, and all contents disclosed in the document are included as part of this specification.

[기술분야][Technology field]

본 발명은 암의 예방 또는 치료를 위한 조성물에 관한 것이다.The present invention relates to a composition for preventing or treating cancer.

암은 세계적으로 높은 사망률을 보이고 있으며, 서구 사회에서는 심혈관 질환 다음으로 가장 일반적인 사망 원인이다. 특히, 인구의 고령화, 식생활의 서구화로 인한 고지방식의 섭취의 일반화, 환경 오염 물질의 급격한 증가, 음주량의 증가 등으로 대장암, 유방암, 전립선암 등이 지속적으로 증가하는 추세에 있다. 이러한 실정에서 암의 예방 및 치료를 가능하게 하여 인간 건강의 증진, 건강한 삶의 질 향상 및 인류 보건 증진에 기여할 수 있는 항암 물질의 창출이 절실히 요구되고 있다.Cancer has a high mortality rate worldwide and is the most common cause of death in Western societies after cardiovascular disease. In particular, colon cancer, breast cancer, and prostate cancer are continuously increasing due to the aging of the population, the generalization of high-fat diet due to westernization of diet, rapid increase in environmental pollutants, and increase in alcohol consumption. In this situation, there is an urgent need for the creation of anticancer substances that can prevent and treat cancer and contribute to the improvement of human health, healthy quality of life, and human health.

그 중에서도 뇌 및 중추신경계 암은 국가암등록통계 발표 자료에 따르면 0세에서 14세군 조발생률 순위 중, 남자 3위 여자 2위에 위치하며 남녀 전체에서 3위를 차지하고 있다. 뇌종양은 국내에서 1년에 3000여명의 빈도로 발생하며, 양성 뇌종양은 근치적 수술을 통해 완치가 가능하지만 악성 뇌종양은 수술, 방사선요법, 항암화학요법을 병합하여 치료를 하고 있다. 일반적으로 발생하는 뇌종양은 신경교종, 뇌수막증, 뇌하수체선종, 전이성 뇌종양, 청신경초종 등이 있으며, 이 중 신경교종(교모세포종)은 WHO 뇌종양 분류 4등급의 가장 흔한 악성 종양으로 알려져 있고, 발병률은 1년에 10만명당 약 3~4명으로 알려져 있다.Among them, brain and central nervous system cancers are ranked 3rd in men and 2nd in women, and 3rd among men and women among the crude incidence rates for the 0 to 14-year-old group, according to national cancer registration statistics. Brain tumors occur in approximately 3,000 people per year in Korea. Benign brain tumors can be cured through curative surgery, but malignant brain tumors are treated with a combination of surgery, radiation therapy, and chemotherapy. Commonly occurring brain tumors include glioma, meningoencephalopathy, pituitary adenoma, metastatic brain tumor, and acoustic schwannoma. Among these, glioma (glioblastoma) is known to be the most common malignant tumor, grade 4 in the WHO brain tumor classification, and has an incidence rate of 1 It is said to be about 3 to 4 per 100,000 people per year.

뇌종양의 치료방법으로는 수술, 방사선요법, 항암화학요법 등이 있으며, 일반적으로 수술 및 방사선요법으로 암을 제거한 후 항암화학요법을 사용한다. 뇌종양 치료를 위한 일차적인(first-line) 항암화학요법 치료제로는 세포 내 DNA를 알킬화하거나 메틸화하는 테모졸로마이드(Temozolomide)가 있으며, 이차적인(second-line) 약제로는 아바스틴-이리노테칸(Avastin-Irinotecan)가 있다.Treatment methods for brain tumors include surgery, radiation therapy, and chemotherapy. Generally, chemotherapy is used after removing the cancer through surgery and radiation therapy. The first-line chemotherapy treatment for brain tumors includes Temozolomide, which alkylates or methylates intracellular DNA, and the second-line drug is Avastin-irinotecan. Irinotecan).

그러나, 뇌종양 치료에 사용되는 테모졸로마이드에 대한 약제의 불응성 및 내성으로 인해 환자의 치료율이 낮고 재발 및 전이 발생이 빈번하여 환자의 예후가 불량한 문제가 있고, 이로 인해 악성 뇌종양의 5년 생존율은 20~30% 정도로 종양 치료 과정의 초기에 많은 환자가 사망하고 있어, 이를 치료하기 위한 신규 표적의 발굴이 필요한 실정이다.However, due to drug refractoriness and resistance to temozolomide, which is used to treat brain tumors, the cure rate of patients is low and recurrence and metastasis occur frequently, resulting in poor patient prognosis. As a result, the 5-year survival rate of malignant brain tumors is low. As many patients die early in the tumor treatment process (about 20-30%), there is a need to discover new targets to treat this.

본 발명의 일 목적은 암의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.One object of the present invention is to provide a pharmaceutical composition for preventing or treating cancer.

본 발명의 다른 목적은 암의 치료용 제제의 스크리닝 방법을 제공하는 것이다.Another object of the present invention is to provide a method for screening agents for treating cancer.

상기 목적을 달성하기 위하여, 본 발명은 NMUR2 억제제를 유효성분으로 포함하는 암의 예방 또는 치료용 약학적 조성물을 제공한다.In order to achieve the above object, the present invention provides a pharmaceutical composition for preventing or treating cancer containing an NMUR2 inhibitor as an active ingredient.

상기 NMUR2 억제제는 NMUR2 발현 억제제 또는 NMUR2 활성 억제제일 수 있다.The NMUR2 inhibitor may be an NMUR2 expression inhibitor or an NMUR2 activity inhibitor.

상기 NMUR2 억제제는 하기 <화학식 I>의 화학식을 갖는 화합물 또는 그의 제약상 허용되는 염일 수 있다.The NMUR2 inhibitor may be a compound having the following formula (I) or a pharmaceutically acceptable salt thereof.

<화학식 I><Formula I>

Figure PCTKR2023003111-appb-img-000001
Figure PCTKR2023003111-appb-img-000001

상기 암은 NMUR2가 과발현된 것일 수 있다.The cancer may be one in which NMUR2 is overexpressed.

또한, 상기 암은 뇌종양일 수 있다. Additionally, the cancer may be a brain tumor.

또한, 상기 목적을 달성하기 위하여, 본 발명은 NMUR2가 과발현된 세포에 항암제 후보 물질을 처리하는 단계; 및 상기 후보 물질이 처리된 세포에서 NMUR2의 발현 또는 활성 수준을 측정하는 단계;를 포함하는, 항암제 스크리닝 방법을 제공한다.In addition, in order to achieve the above object, the present invention includes the steps of treating cells overexpressing NMUR2 with an anticancer drug candidate; and measuring the expression or activity level of NMUR2 in cells treated with the candidate substance.

본 발명의 약학적 조성물이 표적화하는 NMUR2는 종래 암의 예방 또는 치료를 위한 표적으로는 전혀 알려져 있지 않던 신규 표적인바, NMUR2 발현 또는 활성 억제제를 유효성분으로 하는 본 발명의 약학적 조성물은 종래의 항암제들에 불응성을 나타내거나 내성을 나타내는 경우의 대체 항암제로 이용될 수 있는 장점이 있다.NMUR2, which is targeted by the pharmaceutical composition of the present invention, is a novel target that has not been previously known as a target for the prevention or treatment of cancer. The pharmaceutical composition of the present invention, which contains an NMUR2 expression or activity inhibitor as an active ingredient, is a conventional anticancer agent. It has the advantage of being able to be used as an alternative anticancer agent in cases of refractoriness or resistance.

특히, NMUR2 활성 억제제의 경우, 종래의 항암제인 테모졸로마이드에 비하여 현저하게 낮은 IC50 값을 나타내는바, 그 단독으로도 암의 예방 또는 치료를 위한 유효성분이 될 수 있다. 또한, 본 발명의 약학적 조성물의 경우, 종래의 항암제들과 병용됨으로써 각각 단독으로 이용되는 경우에 비해 상승효과를 나타내는바, 항암 보조제로서도 이용될 수 있는 장점이 있다. 또한, NMUR2의 발현 억제제의 경우, 암의 세포 증식 및 세포 이동을 억제하여, 암 세포의 사멸을 촉진하고, 전이를 억제할 수 있어, 효과적인 항암제로 사용될 수 있다.In particular, NMUR2 activity inhibitors have a significantly lower IC 50 value compared to temozolomide, a conventional anticancer drug, and can be used alone as an effective ingredient for the prevention or treatment of cancer. In addition, the pharmaceutical composition of the present invention, when used in combination with conventional anticancer drugs, exhibits a synergistic effect compared to when each is used alone, so it has the advantage of being able to be used as an anticancer adjuvant. In addition, NMUR2 expression inhibitors can inhibit cancer cell proliferation and cell migration, promote cancer cell death, and inhibit metastasis, so they can be used as effective anticancer agents.

도 1A, 1B 및 1C는 CMV 프로모터로 발현되고 NMUR2를 암호화하는 염기서열 및 IRES-GFP를 암호화하는 염기서열이 삽입된 pcDNA3.1 플라스미드 벡터의 구조, bright field와 GFP 발현 형광 및 상기 벡터를 HTLA 세포주에 도입하고 NMUR2의 리간드인 NMU-25를 처리하였을 때의 루시퍼라제(luciferase)의 강도를 나타낸 그래프이다. Figures 1A, 1B, and 1C show the structure of the pcDNA3.1 plasmid vector expressed by the CMV promoter and inserted with the base sequence encoding NMUR2 and the base sequence encoding IRES-GFP, the bright field and fluorescence of GFP expression, and the expression of the vector in the HTLA cell line. This is a graph showing the intensity of luciferase when introduced into and treated with NMU-25, the ligand of NMUR2.

도 2a는 상기 HTLA 세포주에 NMU-25와 화학식 I의 화합물을 처리했을 때 루시퍼라제의 강도를 나타낸 그래프이다. Figure 2a is a graph showing the intensity of luciferase when the HTLA cell line was treated with NMU-25 and the compound of Formula I.

도 2b는 상기 HTLA 세포주에 NMU-25와 화학식 I의 화합물을 농도별로 처리했을 때 루시퍼라제의 강도를 나타낸 그래프이다.Figure 2b is a graph showing the intensity of luciferase when the HTLA cell line was treated with NMU-25 and the compound of Formula I at various concentrations.

도 3A 및 3B는 뇌종양 세포주인 U-87MG, T-98G 및 U-373MG에 대한 테모졸로마이드와 화학식 I의 화합물의 IC50을 측정한 그래프와 농도에 따른 caspase 3/7의 활성을 나타낸 그래프이다. Figures 3A and 3B are graphs measuring the IC 50 of temozolomide and the compound of Formula I against brain tumor cell lines U-87MG, T-98G, and U-373MG, and graphs showing the activity of caspase 3/7 according to concentration. .

도 4A 및 도 4B는 테모졸로마이드와 화학식 I의 화합물을 병용처리할 때의 OD450, caspase 3/7의 활성 및 CI 값을 확인한 그래프이다. Figures 4A and 4B are graphs confirming the OD450, caspase 3/7 activity, and CI values when temozolomide and the compound of Formula I are co-treated.

도 5는 위암, 대장암 및 피부암 세포주인 SNU-638, HCT116 및 SK-MEL-2에 화학식 I의 화합물을 처리하였을 때의 IC50을 확인한 그래프이다. Figure 5 is a graph confirming IC 50 when gastric cancer, colon cancer, and skin cancer cell lines SNU-638, HCT116, and SK-MEL-2 were treated with the compound of Formula I.

도 6은 교모세포종(glioblastoma) 및 신경교종(glioma) 샘플들에서 유전적 돌연변이와 NMUR2 발현 간의 상관 관계를 분석한 도이다.Figure 6 is a diagram analyzing the correlation between genetic mutations and NMUR2 expression in glioblastoma and glioma samples.

도 7은 pcDNA 공벡터가 도입된 교모세포종 세포주와 달리, pcDNA3.1 NMUR2-과발현 벡터가 도입된 교모세포종 세포주는 NMUR2를 높은 수준으로 발현하는 것을 확인한 도이다.Figure 7 is a diagram confirming that, unlike the glioblastoma cell line into which the pcDNA empty vector was introduced, the glioblastoma cell line into which the pcDNA3.1 NMUR2-overexpression vector was introduced expresses NMUR2 at a high level.

도 8은 NMUR2 과발현 세포주에서 교모세포종 세포의 증식도가 대조군에 비하여 증가하는 것을 확인한 도이다.Figure 8 is a diagram confirming that the proliferation rate of glioblastoma cells in the NMUR2 overexpressing cell line is increased compared to the control group.

도 9는 NMUR2 과발현 세포주에서 교모세포종 세포의 이동이 대조군에 비하여 증가하는 것을 확인한 도이다.Figure 9 is a diagram confirming that the migration of glioblastoma cells in the NMUR2 overexpressing cell line is increased compared to the control group.

도 10은 NMUR2에 대한 siRNA를 처리한 교모세포종 세포주에서 NMUR2의 발현이 유의미하게 감소한 것을 확인한 도이다.Figure 10 is a diagram confirming that the expression of NMUR2 was significantly reduced in glioblastoma cell lines treated with siRNA against NMUR2.

도 11은 NMUR2에 대한 siRNA를 처리한 교모세포종 세포주에서 세포의 증식도가 대조군에 비하여 감소한 것을 확인한 도이다.Figure 11 is a diagram confirming that the proliferation rate of cells in glioblastoma cell lines treated with siRNA against NMUR2 was reduced compared to the control group.

도 12는 NMUR2에 대한 siRNA를 처리한 교모세포종 세포주에서 세포의 이동 대조군에 비하여 감소한 것을 확인한 도이다.Figure 12 is a diagram confirming the decrease in cell migration in glioblastoma cell lines treated with siRNA against NMUR2 compared to the control group.

도 13은 NMUR2을 억제하는 화합물인 화학식 1의 화합물이 교모세포종 세포주의 세포 이동을 억제한 것을 확인한 도이다.Figure 13 is a diagram confirming that the compound of Formula 1, a compound that inhibits NMUR2, inhibited cell migration of a glioblastoma cell line.

이하, 본 발명을 상세히 설명한다. Hereinafter, the present invention will be described in detail.

1. 암의 치료 또는 예방용 약학적 조성물1. Pharmaceutical composition for the treatment or prevention of cancer

본 발명은 일 측면은 암의 치료 또는 예방용 약학적 조성물을 제공한다.One aspect of the present invention provides a pharmaceutical composition for treating or preventing cancer.

상기 본 발명의 암의 치료 또는 예방용 약학적 조성물은 NMUR2(Neuromedin U Receptor 2; NCBI GenBank ID: 56923)의 억제제를 유효성분으로 포함한다.The pharmaceutical composition for treating or preventing cancer of the present invention contains an inhibitor of NMUR2 (Neuromedin U Receptor 2; NCBI GenBank ID: 56923) as an active ingredient.

상기 "NMUR2(Neuromedin U Receptor 2)"는 유전자의 명칭 또는 상기 유전자에 의해 암호화되는 단백질의 명칭으로서 사용된다. NMUR2 유전자에 의해 암호화되는 NMUR2 단백질은 G-단백질 연결 수용체(G-protein coupled receptor; GPCR)의 일종으로, 뉴로펩티드 호르몬인 NMU(뉴로메딘 U; neuromedin U) 및 NMS(뉴로메딘 S; neuromedin S)와 결합하여 세포의 성장, 증식, 사멸 등 각종 세포 반응을 활성화시키는 역할을 한다.The “NMUR2 (Neuromedin U Receptor 2)” is used as the name of the gene or the name of the protein encoded by the gene. The NMUR2 protein, encoded by the NMUR2 gene, is a type of G-protein coupled receptor (GPCR) and is responsible for the neuropeptide hormones NMU (neuromedin U) and NMS (neuromedin S). It plays a role in activating various cellular reactions such as cell growth, proliferation, and death by combining with.

상기 NMUR2 억제제는 NMUR2 발현 억제제 또는 NMUR2 활성 억제제일 수 있다.The NMUR2 inhibitor may be an NMUR2 expression inhibitor or an NMUR2 activity inhibitor.

본 발명에서 용어 "NMUR2 발현 억제제 또는 NMUR2 활성 억제제"는 NMUR2의 발현 또는 활성을 감소시키는 제제를 모두 통칭하는 의미로 사용되며, 구체적으로는 전사(transcription), mRNA, 또는 번역(translation) 수준에서 NMUR2 유전자의 발현양을 감소시킴으로써 발현을 억제하거나, NMUR2 단백질의 활성을 방해함으로써 NMUR2의 활성을 억제하는 모든 제제를 포함할 수 있다.In the present invention, the term "NMUR2 expression inhibitor or NMUR2 activity inhibitor" is used to collectively refer to all agents that reduce the expression or activity of NMUR2, and specifically, NMUR2 at the transcription, mRNA, or translation level. It may include any agent that inhibits the activity of NMUR2 by suppressing its expression by reducing the expression level of the gene or by interfering with the activity of the NMUR2 protein.

상기 "NMUR2 발현 억제제 또는 NMUR2 활성 억제제"는 NMUR2의 발현 또는 NMUR2을 표적으로 하여 활성을 억제할 수 있는 화합물, 핵산, 펩타이드, 바이러스 또는 상기 핵산을 포함하는 벡터 등 그 형태에 제한 없이 사용 가능하다. 예를 들어서, 상기 NMUR2 발현 억제제는 NMUR2 유전자의 mRNA에 상보적으로 결합하는 miRNA, siRNA 또는 shRNA나, 안티센스 올리고뉴클레오티드 등일 수 있고, 특히, siRNA일 수 있으며, 상기 NMUR2 활성 억제제는 NMUR2 단백질에 특이적으로 결합하는 앱타머(aptamer), 항체 또는 저분자 화합물 등일 수 있다. The “NMUR2 expression inhibitor or NMUR2 activity inhibitor” can be used without limitation in its form, such as a compound, nucleic acid, peptide, virus, or vector containing the nucleic acid that can inhibit the expression or activity of NMUR2 by targeting NMUR2. For example, the NMUR2 expression inhibitor may be a miRNA, siRNA or shRNA, or an antisense oligonucleotide that binds complementary to the mRNA of the NMUR2 gene. In particular, it may be siRNA, and the NMUR2 activity inhibitor is specific for the NMUR2 protein. It may be an aptamer, antibody, or low-molecular-weight compound that binds to.

본 발명에서 용어, "miRNA", "siRNA" 및 "shRNA"는 RNA 방해 또는 유전자 사일런싱(silencing)을 매개할 수 있는 핵산 분자로서, 표적 유전자의 발현을 억제할 수 있기 때문에 효율적인 유전자 녹다운(knock down) 방법 또는 유전자 치료 방법으로 사용된다. miRNA, siRNA 및 shRNA는 본 발명의 목적상 NMUR2에 특이적으로 작용하여 NMUR2 mRNA 분자를 절단하여 RNA 간섭(RNAi; RNA interference) 현상을 유도함으로써, 상기 NMUR2를 억제할 수 있다. miRNA, siRNA 및 shRNA는 화학적으로 또는 효소학적으로 합성될 수 있다. 특히, 상기 siRNA는 서열번호 5 및 서열번호 6으로 표시되는 염기 서열, 또는 서열번호 7 및 서열번호 8로 표시되는 염기 서열을 포함할 수 있다.In the present invention, the terms "miRNA", "siRNA", and "shRNA" are nucleic acid molecules that can mediate RNA interference or gene silencing, and can inhibit the expression of target genes, thereby producing efficient gene knockdown. down) method or gene therapy method. For the purpose of the present invention, miRNA, siRNA, and shRNA can inhibit NMUR2 by acting specifically on NMUR2 to cleave the NMUR2 mRNA molecule and induce RNA interference (RNAi). miRNAs, siRNAs and shRNAs can be synthesized chemically or enzymatically. In particular, the siRNA may include the nucleotide sequences represented by SEQ ID NO: 5 and SEQ ID NO: 6, or the nucleotide sequences represented by SEQ ID NO: 7 and SEQ ID NO: 8.

본 발명에서 용어, "앱타머(aptamer)"는 단일가닥 올리고뉴클레오 티드로서, 20 내지 60 뉴클레오티드 정도의 크기이며, 소정의 표적 분자에 대한 결합 활성을 갖는 핵산 분자를 말한다. 서열에 따라 다양한 3 차원 구조를 가지며, 항원-항체 반응처럼 특정 물질과 높은 친화력을 가질 수 있다. 앱타머는 소정의 표적 분자에 결합함으로써, 소정의 표적 분자의 활성을 저해할 수 있다. 본 발명의 앱타머는 RNA, DNA, 변형된(modified) 핵산 또는 이들의 혼합물일 수 있으며, 또한 직쇄상 또는 환상의 형태일 수 있다. 바람직하게 상기 앱타머는 NMUR2에 결합하여 NMUR2의 활성을 저해하는 역할을 할 수 있다. 이와 같은 앱타머는 NMUR2의 서열로부터 당업자가 공지의 방법에 의해 제조할 수 있다.In the present invention, the term “aptamer” refers to a single-stranded oligonucleotide, about 20 to 60 nucleotides in size, and a nucleic acid molecule that has binding activity to a given target molecule. It has a variety of three-dimensional structures depending on its sequence, and can have high affinity for specific substances, like an antigen-antibody reaction. An aptamer can inhibit the activity of a certain target molecule by binding to it. The aptamer of the present invention may be RNA, DNA, modified nucleic acid, or a mixture thereof, and may also be in a linear or circular form. Preferably, the aptamer may bind to NMUR2 and serve to inhibit the activity of NMUR2. Such an aptamer can be prepared from the sequence of NMUR2 by a method known to those skilled in the art.

본 발명에서 용어 "항체"는 적어도 1 개의 이뮤노글로불린 가변 영역 서열을 포함하고 주어진 항원에 특이적으로 결합하는 폴리펩티드를 지칭한다. 예를 들어, 항체는 중쇄(H) 가변 영역 및 경쇄(L) 가변 영역을 포함할 수 있다.As used herein, the term “antibody” refers to a polypeptide that includes at least one immunoglobulin variable region sequence and specifically binds to a given antigen. For example, an antibody may comprise a heavy (H) chain variable region and a light (L) chain variable region.

본 발명에서 용어 "저분자 화합물"은 분자량이 1000 Da 이하, 예컨대 800Da 이하, 특히 500Da 이하인 천연 또는 합성의 화합물로서, 상기 NMUR2 단백질에 결합되어 NMUR2의 활성을 방해하는 것일 수 있다. 특히, 상기 저분자 화합물은 하기 <화학식 I>의 화학식을 갖는 화합물 또는 그의 약학적으로 허용가능한 염일 수 있다. In the present invention, the term "low molecular weight compound" is a natural or synthetic compound with a molecular weight of 1000 Da or less, such as 800 Da or less, especially 500 Da or less, and may bind to the NMUR2 protein and interfere with the activity of NMUR2. In particular, the low molecular weight compound may be a compound having the formula of <Formula I> below or a pharmaceutically acceptable salt thereof.

<화학식 I><Formula I>

Figure PCTKR2023003111-appb-img-000002
Figure PCTKR2023003111-appb-img-000002

상기 "약학적으로 허용가능한"의 의미는 유효성분의 활성을 억제하지 않으면서 적용(처방) 대상이 적응 가능한 수준 이상의 독성을 지니지 않는다는 의미이다. 상기 "염"은 화합물의 무기 및 유기산 부가염을 의미한다. 상기 "약학적으로 허용가능한 염"은 화합물이 투여되는 유기체에 심각한 자극을 유발하지 않고 화합물의 생물학적 활성과 물성들은 손상시키지 않는 염일 수 있고, 무기산 염, 유기산 염 또는 금속 염일 수 있다. 상기 무기산염은 염산염, 브롬산염, 인산염, 황산염, 또는 이황산염일 수 있다. 상기 유기산염은 포름산염, 아세트산염, 프로피온산염, 젖산염, 옥살산염, 주석산염, 말산염, 말레인산염, 구연산염, 푸마르산염, 베실산염, 캠실산염, 에디실염, 트리클로로아세트산, 트리플루오로아세트산염, 벤조산염, 글루콘산염, 메탄술폰산염, 글리콜산염, 숙신산염, 4-톨루엔술폰산염, 갈룩투론산염, 엠본산염, 글루탐산염, 에탄술폰산염, 벤젠술폰산염, p-톨루엔술폰산염, 또는 아스파르트산염일 수 있다. 상기 금속염은 칼슘염, 나트륨염, 마그네슘염, 스트론튬염, 또는 칼륨염일 수 있다.The meaning of “pharmaceutically acceptable” means that it does not inhibit the activity of the active ingredient and does not have toxicity beyond a level acceptable to the subject of application (prescription). The term “salt” refers to inorganic and organic acid addition salts of the compound. The “pharmaceutically acceptable salt” may be a salt that does not cause serious irritation to the organism to which the compound is administered and does not impair the biological activity and physical properties of the compound, and may be an inorganic acid salt, an organic acid salt, or a metal salt. The inorganic acid salt may be hydrochloride, bromate, phosphate, sulfate, or disulfate. The organic acid salts include formate, acetate, propionate, lactate, oxalate, tartrate, malate, maleate, citrate, fumarate, besylate, camsylate, edicyl salt, trichloroacetic acid, and trifluoroacetate. , benzoate, gluconate, methanesulfonate, glycolate, succinate, 4-toluenesulfonate, galacturonate, embonate, glutamate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, or aspart. It may be an acid salt. The metal salt may be a calcium salt, sodium salt, magnesium salt, strontium salt, or potassium salt.

본 발명에서 용어 "암"은 결장암 및 직장암을 포함하는 대장암, 유방암, 자궁암, 자궁경부암, 난소암, 전립선암, 두경부암종, 흑색종, 골수종, 백혈병, 림프종, 위암, 폐암, 췌장암, 간암, 식도암, 소장암, 항문부근암, 나팔관암종, 자궁내막암종, 질암종, 음문암종, 호지킨병, 방광암, 신장암, 수뇨관암, 신장세포암종, 신장골반암종, 골암, 피부암, 두부암, 경부암, 피부흑색종, 안구내흑색종, 내분비선암, 갑상선암, 부갑상선암, 부신암, 연조직육종, 요도암, 음경암 또는 중추신경계(central nervous system;CNS) 암일 수 있고, 예컨대 대장암, 위암, 폐암, 췌장암, 간암, 소장암, 피부암 또는 중추신경계 암일 수 있고, 특히 중추신경계 암일 수 있다.In the present invention, the term "cancer" refers to colorectal cancer, including colon cancer and rectal cancer, breast cancer, uterine cancer, cervical cancer, ovarian cancer, prostate cancer, head and neck carcinoma, melanoma, myeloma, leukemia, lymphoma, stomach cancer, lung cancer, pancreatic cancer, liver cancer, Esophageal cancer, small intestine cancer, perianal cancer, fallopian tube carcinoma, endometrial carcinoma, vaginal carcinoma, vulvar carcinoma, Hodgkin's disease, bladder cancer, kidney cancer, ureteral cancer, renal cell carcinoma, renal pelvic carcinoma, bone cancer, skin cancer, head cancer, and neck cancer. , cutaneous melanoma, intraocular melanoma, endocrine cancer, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, or central nervous system (CNS) cancer, such as colon cancer, stomach cancer, and lung cancer. , it may be pancreatic cancer, liver cancer, small intestine cancer, skin cancer, or central nervous system cancer, especially central nervous system cancer.

상기 중추신경계 암은 뇌종양일 수 있다. 상기 "뇌종양"은 두개골 내에 생기는 모든 종양을 말하며 뇌 및 뇌 주변 구조물에서 발생하는 모든 종양을 포함한다. 특히, 상기 뇌종양은 신경교종, 뇌수막증, 뇌하수체선종, 전이성 뇌종양, 청신경초종, 교모세포종 등일 수 있으나, 이에 한정되는 것은 아니다.The central nervous system cancer may be a brain tumor. The “brain tumor” refers to any tumor that occurs within the skull and includes all tumors that occur in the brain and structures surrounding the brain. In particular, the brain tumor may be glioma, meningoencephalopathy, pituitary adenoma, metastatic brain tumor, acoustic schwannoma, glioblastoma, etc., but is not limited thereto.

한편, 상기 암은 NMUR2가 과발현된 것일 수 있다. 본 발명의 약학적 조성물은 NMUR2 억제제를 유효성분으로 포함하고 있으므로, NMUR2가 과발현된 암에 대하여 특히 우수한 예방 또는 치료의 효과를 나타낼 수 있다.Meanwhile, the cancer may be one in which NMUR2 is overexpressed. Since the pharmaceutical composition of the present invention contains an NMUR2 inhibitor as an active ingredient, it can exhibit a particularly excellent prevention or treatment effect against cancer in which NMUR2 is overexpressed.

본 발명에서 용어, "치료"는 치료하고자 하는 개개인 또는 세포의 천연 과정을 변경시키기 위해 임상적으로 개입하는 것을 지칭하고, 이는 임상 병리 상태가 진행되는 동안 또는 이를 예방하기 위해 수행할 수 있다. 목적하는 치료 효과에는 질병의 발생 또는 재발을 예방하고, 증상을 완화시키며, 질병에 따른 모든 직접 또는 간접적인 병리학적 결과를 저하시키며, 전이를 예방하고, 질병 진행 속도를 감소시키며, 질병 상태를 경감 또는 일시적으로 완화시키며, 차도시키거나 예후를 개선시키는 것이 포함된다. 본 발명에서는 NMUR2를 억제하는 물질을 포함하는 조성물의 투여로 암의 경과를 호전시키는 모든 행위를 포함한다. 또한, "예방"은 본 발명에 따른 NMUR2를 억제하는 물질을 포함하는 조성물의 투여로 암의 발병을 억제 또는 지연시키는 모든 행위를 말한다.As used herein, the term “treatment” refers to clinical intervention to alter the natural processes of the individual or cell being treated, which may be performed during the course of the clinical pathology or to prevent it. The desired therapeutic effects include preventing the occurrence or recurrence of the disease, alleviating symptoms, reducing all direct or indirect pathological consequences of the disease, preventing metastasis, reducing the rate of disease progression, and alleviating the disease state. or temporarily alleviating, remission, or improving prognosis. The present invention includes all actions to improve the course of cancer by administering a composition containing a substance that inhibits NMUR2. In addition, “prevention” refers to all actions that inhibit or delay the onset of cancer by administering a composition containing a substance that inhibits NMUR2 according to the present invention.

본 발명의 약학적 조성물은 약학적 조성물의 제조에 통상적으로 사용하는 적절한 담체, 부형제 또는 희석제를 추가로 포함할 수 있다.The pharmaceutical composition of the present invention may further include appropriate carriers, excipients, or diluents commonly used in the preparation of pharmaceutical compositions.

약학적으로 허용 가능한 담체를 포함하는 조성물은 경구 또는 비경구의 여러 가지 제형일 수 있다. 제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면 활성제 등의 희석제 또는 부형제를 사용하여 조제될 수 있다.The composition containing a pharmaceutically acceptable carrier may be in various oral or parenteral dosage forms. When formulated, it can be prepared using diluents or excipients such as commonly used fillers, extenders, binders, wetting agents, disintegrants, and surfactants.

경구 투여를 위한 고형제제에는 정제환제, 산제, 과립제, 캡슐제 등이 포함될 수 있으며, 이러한 고형 제제는 하나 이상의 화합물에 적어도 하나 이상의 부형제, 예를 들면, 전분, 탄산 칼슘, 수크로오스(sucrose) 또는 락토오스(lactose), 젤라틴 등을 섞어 조제될 수 있다. 또한, 단순한 부형제 이외에 스테아린산 마그네슘, 탈크 등과 같은 윤활제들도 사용될 수 있다. 경구 투여를 위한 액상제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데 흔히 사용되는 단순 희석제인 물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등이 포함될 수 있다.Solid preparations for oral administration may include tablets, powders, granules, capsules, etc., and such solid preparations may contain one or more compounds and at least one excipient, such as starch, calcium carbonate, sucrose, or lactose. It can be prepared by mixing (lactose), gelatin, etc. Additionally, in addition to simple excipients, lubricants such as magnesium stearate, talc, etc. may also be used. Liquid preparations for oral administration include suspensions, oral solutions, emulsions, and syrups. In addition to the commonly used simple diluents such as water and liquid paraffin, various excipients such as wetting agents, sweeteners, fragrances, and preservatives may be included. there is.

비경구투여를 위한 제제에는 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결건조제제, 좌제가 포함될 수 있다. 비수성용제, 현탁용제로는 프로필렌글리콜(propylene glycol), 폴리에틸렌 글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기제로는 위텝솔(witepsol), 마크로골, 트윈(tween) 61, 카카오지, 라우린지, 글리세로젤라틴 등이 사용될 수 있다.Preparations for parenteral administration may include sterilized aqueous solutions, non-aqueous solutions, suspensions, emulsions, freeze-dried preparations, and suppositories. Non-aqueous solvents and suspensions may include propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable ester such as ethyl oleate. As a base for suppositories, witepsol, macrogol, tween 61, cacao, laurel, glycerogelatin, etc. can be used.

또한, 본 발명의 약학적 조성물은 이에 제한되지는 않으나, 정제, 환제, 산제, 과립제, 캡슐제, 현탁제, 내용액제, 유제, 시럽제, 멸균된 수용액, 비수성용제, 현탁제, 유제, 및 동결건조제제로 이루어진 군으로부터 선택되는 어느 하나의 제형을 가질 수 있다.In addition, the pharmaceutical composition of the present invention is not limited thereto, but includes tablets, pills, powders, granules, capsules, suspensions, oral solutions, emulsions, syrups, sterilized aqueous solutions, non-aqueous solutions, suspensions, emulsions, and freezing. It may have any one formulation selected from the group consisting of dry formulations.

본 발명의 약학적 조성물은 매일 약 0.0001 mg/kg 내지 약 10 g/kg이 투여될 수 있으며, 약 0.001 mg/kg 내지 약 1 g/kg의 1일 투여 용량으로 투여될 수 있다. 그러나 상기 투여량은 상기 혼합물의 정제 정도, 환자의 상태(연령, 성별, 체중 등), 치료하고 있는 상태의 심각성 등에 따라 다양할 수 있다. 필요에 따라 편리성을 위하여 1일 총 투여량은 하루 동안 여러 번 나누어 투여될 수 있다.The pharmaceutical composition of the present invention may be administered in a daily dose of about 0.0001 mg/kg to about 10 g/kg, and may be administered in a daily dosage of about 0.001 mg/kg to about 1 g/kg. However, the dosage may vary depending on the degree of purification of the mixture, the patient's condition (age, gender, weight, etc.), and the severity of the condition being treated. For convenience, the total daily dose may be divided and administered several times during the day, if necessary.

본 발명의 구체적인 실시예에서는 상기 화학식 I의 화합물이 NMUR2 억제제로서 NMUR2의 활성을 억제하는 것임을 확인하였고(도 2 참조), 뇌종양 세포주인 U-87MG, T-98G 및 U-373MG에 대하여 종래의 뇌종양 치료제인 테모졸로마이드에 비하여 훨씬 우수한 치료 효과를 나타냄을 확인하였다(도 3A 및 4A 참조).In a specific example of the present invention, it was confirmed that the compound of formula (I) is an NMUR2 inhibitor that inhibits the activity of NMUR2 (see Figure 2), and it was confirmed that the conventional brain tumor cell lines U-87MG, T-98G, and U-373MG were used as brain tumor cell lines. It was confirmed that it exhibited a much better therapeutic effect than the therapeutic agent temozolomide (see Figures 3A and 4A).

본 발명의 또 다른 일실시예에서는 다른 암종에 대한 화학식 I의 화합물의 효과를 확인하기 위해 각각 위암, 대장암 및 피부암 세포주인 SNU-638, HCT116 및 SK-MEL-2에 화학식 I의 화합물을 단독으로 처리하였고 뇌종양 세포주들에서와 유사한 수준의 IC50을 나타냄을 확인하였다.In another embodiment of the present invention, in order to confirm the effect of the compound of Formula I on other cancer types, the compound of Formula I was administered alone to SNU-638, HCT116, and SK-MEL-2, which are gastric cancer, colon cancer, and skin cancer cell lines, respectively. and was confirmed to have an IC 50 similar to that of brain tumor cell lines.

2. 암의 예방 또는 치료용 키트2. Kit for preventing or treating cancer

본 발명의 다른 측면은, 암의 예방 또는 치료용 키트를 제공한다.Another aspect of the present invention provides a kit for preventing or treating cancer.

상기 본 발명의 암의 예방 또는 치료용 키트는 상기 약학적 조성물과 기존의 항암제를 포함할 수 있다. 본 발명의 암의 예방 또는 치료용 키트는 약학적 조성물에 통상적으로 사용되는 담체, 희석제, 부형제 또는 이들 둘 이상의 조합을 추가적으로 포함할 수 있다. 또한 상기 키트는 의약품이나 생물학적 제제의 제조, 사용 또는 판매를 규제를 위한 정부 기관이 정한 형태의 안내문을 더 포함할 수 있다.The kit for preventing or treating cancer of the present invention may include the pharmaceutical composition and an existing anticancer agent. The kit for preventing or treating cancer of the present invention may additionally include a carrier, diluent, excipient, or a combination of two or more commonly used in pharmaceutical compositions. In addition, the kit may further include a notice in a form established by a government agency for regulating the manufacture, use, or sale of pharmaceuticals or biological products.

상기 약학적 조성물은 상기 '1. 암의 치료 또는 예방용 약학적 조성물 '에서 설명한 바와 동일하므로, 추가적인 설명은 생략한다.Since the pharmaceutical composition is the same as described in ' 1. Pharmaceutical composition for treating or preventing cancer ', further description is omitted.

상기 "기존의 항암제"는 암세포의 증식을 억제하기 위하여 기사용되어 온 화학요법 치료제를 의미한다. 상기와 같은 기존의 항암제는 NMUR2이 아닌 다른 표적에 대한 항암제일 수 있고, 예를 들어 테모졸로마이드 또는 아바스틴-이리노테칸일 수 있고, 특히 테모졸로마이드일 수 있다.The “existing anticancer agent” refers to a chemotherapy treatment agent that has been previously used to inhibit the proliferation of cancer cells. The existing anticancer agent as described above may be an anticancer agent targeting a target other than NMUR2, for example, temozolomide or Avastin-irinotecan, and especially temozolomide.

상기 약학적 조성물은 NMUR2 억제제를 유효성분으로 포함하고 있으므로, 상기 기존의 항암제와는 다른 표적에 작용하여 항암 효과를 더욱 상가적으로 상승시킬 수 있다.Since the pharmaceutical composition contains an NMUR2 inhibitor as an active ingredient, it can further increase the anticancer effect by acting on a different target than the existing anticancer agent.

상기 약학적 조성물과 상기 기존의 항암제는 동시에 투여될 수도 있고, 순차적으로 투여될 수도 있다. 상기 약학적 조성물과 상기 기존의 항암제가 순차적으로 투여되는 경우, 상기 약학적 조성물이 먼저 투여된 후 상기 기존의 항암제가 나중에 투여될 수도 있고, 상기 기존의 항암제가 먼저 투여된 후 상기 약학적 조성물이 나중에 투여될 수도 있다. 따라서 상기 키트에는 상기와 같은 투여 방법에 관한 설명이 기재된 설명서가 더 포함될 수 있다. The pharmaceutical composition and the existing anticancer agent may be administered simultaneously or sequentially. When the pharmaceutical composition and the existing anticancer drug are administered sequentially, the pharmaceutical composition may be administered first and then the existing anticancer drug may be administered later, or the existing anticancer drug may be administered first and then the pharmaceutical composition. It may also be administered later. Therefore, the kit may further include an instruction manual containing instructions on the above administration method.

상기 암은 상기 '1. 암의 치료 또는 예방용 약학적 조성물 '에서 설명한 바와 동일하다. 특히, 상기 암은 NMUR2가 과발현된 것일 수 있고, 나아가 상기 기존의 항암제에 불응성이거나 내성을 가지는 것일 수 있다.The cancer is the same as described in ‘ 1. Pharmaceutical composition for treating or preventing cancer ’. In particular, the cancer may overexpress NMUR2, and may further be refractory or resistant to the existing anticancer drugs.

본 발명의 구체적인 실시예에서는 테모졸로마이드와 상기 화학식 I의 화합물을 병용 처리한 경우에, 이들 각각의 약물을 단일 처리할 때보다 뇌종양 세포의 성장이 유의미하게 억제되고 세포의 사멸은 증가됨을 확인하였다. 또한, 상기 테모졸로마이드 및 화학식 I의 화합물의 병용처리 효과를 확인하기 위하여 상기 두 약물을 병용처리하여 CI(병용지수; Combination Index)를 확인하였을 때, 병용처리시 상승효과(synergism)가 있음을 확인하였다.In a specific example of the present invention, it was confirmed that when temozolomide and the compound of formula (I) were treated in combination, the growth of brain tumor cells was significantly inhibited and cell death was increased compared to when each drug was treated alone. . In addition, in order to confirm the effect of the combination treatment of the temozolomide and the compound of Formula I, the two drugs were treated in combination and the CI (Combination Index) was checked, and it was found that there was a synergism during the combination treatment. Confirmed.

3. 항암제 스크리닝 방법3. Anticancer drug screening method

본 발명의 또 다른 측면은 항암제 스크리닝 방법을 제공한다.Another aspect of the present invention provides a method for screening anticancer drugs.

상기 본 발명의 항암제 스크리닝 방법은 NMUR2가 과발현된 세포에, 항암제 후보 물질을 처리하는 단계; 및 상기 후보 물질이 처리된 세포에서 NMUR2의 발현 또는 활성 수준을 측정하는 단계;를 포함한다. The anticancer drug screening method of the present invention includes treating cells overexpressing NMUR2 with an anticancer drug candidate; And measuring the expression or activity level of NMUR2 in cells treated with the candidate substance.

상기 "항암제 후보 물질"은 암세포의 증식을 억제하는 효과를 나타낼 것으로 기대되는 물질을 말한다. The “anticancer drug candidate” refers to a substance expected to exhibit an effect of inhibiting the proliferation of cancer cells.

상기 항암제 후보 물질은 1 μM 내지 40 μM의 농도로 처리될 수 있다. 예를 들어서, 상기 항암제 후보 물질은 5 μM, 10 μM, 15 μM, 20 μM, 25 μM, 30 μM 또는 35 μM의 농도로 처리될 수 있다. The anticancer drug candidate may be treated at a concentration of 1 μM to 40 μM. For example, the anticancer drug candidate may be treated at a concentration of 5 μM, 10 μM, 15 μM, 20 μM, 25 μM, 30 μM, or 35 μM.

상기 "발현 수준 측정"이란 유전자의 mRNA의 존재 여부와 발현 정도를 확인하거나, 유전자에서 발현된 단백질의 존재 여부와 발현 정도를 확인하는 과정을 의미할 수 있다. 상기 유전자의 mRNA의 존재 여부나 발현 정도의 확인은 상기 유전자의 mRNA의 양을 측정함으로써 알 수 있다. 이를 위한 분석 방법으로는 NMUR2의 mRNA에 대한 mRNA에 대한 프라이머(primer) 쌍, 프로브(probe), 또는 안티센스 뉴클레오티드(anti-sense nucleotide) 등을 이용한, RT-PCR, 경쟁적 RT-PCR(competitive RT-PCR), 실시간 RT-PCR (real-time RTPCR), RNase 보호 분석법(RPA; RNase protection assay), 노던 블랏팅 (Northern blotting), DNA 칩 등이 있으나 이로 제한되는 것은 아니다. 또한, 상기 유전자에서 발현된 단백질의 존재 여부나 발현 정도의 확인은 상기 유전자의 단백질의 양을 확인할 수 있다. 이를 위한 분석 방법으로는, NMUR2 단백질에 대하여 특이적으로 결합하는 항체를 이용한, 웨스턴블랏, ELISA (enzyme linked immunosorbent assay), 방사선면역분석(RIA: Radioimmunoassay), 방사면역확산법(radioimmunodiffusion), 오우크테로니(Ouchterlony) 면역 확산법, 로케트(rocket) 면역전기영동, 조직면역염색, 면역침전 분석법(Immunoprecipitation assay), 보체고정분석법(complement fixation assay), FACS, 단백질 칩(protein chip) 등이 있으나, 이에 제한되는 것은 아니다. 또한, 상기 "활성 수준 측정"은 유전자에서 발현된 단백질이 활성을 갖는지 확인하는 과정으로, 상기 단백질의 기능성 여부와 정도를 확인함으로써 이루어진다.The term “expression level measurement” may refer to the process of confirming the presence and expression level of mRNA of a gene, or the presence and expression level of protein expressed from the gene. The presence or expression level of mRNA of the gene can be confirmed by measuring the amount of mRNA of the gene. Analysis methods for this include RT-PCR and competitive RT-PCR using primer pairs, probes, or anti-sense nucleotides for the mRNA of NMUR2. PCR), real-time RT-PCR, RNase protection assay (RPA), Northern blotting, DNA chip, etc., but are not limited to these. Additionally, the presence or expression level of the protein expressed from the gene can be confirmed by confirming the amount of the protein of the gene. Analysis methods for this include Western blot, ELISA (enzyme linked immunosorbent assay), RIA (Radioimmunoassay), radioimmunodiffusion, and Ouktero using an antibody that specifically binds to the NMUR2 protein. Ouchterlony immunodiffusion method, rocket immunoelectrophoresis, tissue immunostaining, immunoprecipitation assay, complement fixation assay, FACS, protein chip, etc. are limited to these. It doesn't work. In addition, the “activity level measurement” is a process of confirming whether a protein expressed from a gene is active, and is accomplished by confirming whether and to what extent the protein is functional.

또한, 상기 항암제 스크리닝 방법은, 상기 측정된 NMUR2의 발현 또는 활성 수준이, 후보 물질이 처리되지 않은 세포에 비해 유의미하게 낮은 경우, 상기 후보 물질을 항암제로 판정하는 단계를 더 포함할 수 있다. In addition, the anticancer drug screening method may further include determining the candidate substance as an anticancer drug when the measured expression or activity level of NMUR2 is significantly lower than that of cells not treated with the candidate substance.

상기 "유의미하게 낮은 경우"는 상기 후보 물질이 처리되지 않은 세포에 비해 상기 NMUR2의 발현 또는 활성 수준이 10% 이상 낮은 경우를 의미하고, 예컨대 20% 이상, 30% 이상, 40% 이상, 50% 이상 또는 60% 이상 낮은 경우일 수 있고, 특히 70% 이상 낮은 경우일 수 있다. 상기 스크리닝 방법은 생체 내(in vivo) 또는 시험관 내(in vitro)에서 수행될 수 있으며, 특별히 제한되지 않는다. 후보 물질은 공지된 물질 또는 신규 물질일 수 있으며, 예를 들어 식물 추출물 또는 케미컬 라이브러리(chemical library)를 통하여 대규모로 스크리닝을 수행할 수 있다. 이를 통해 NMUR2의 발현 또는 활성을 억제하여 암, 특히 뇌종양을 억제할 수 있는 제제를 발굴할 수 있다.The "significantly low case" means that the expression or activity level of NMUR2 is lower by 10% or more compared to cells not treated with the candidate substance, such as 20% or more, 30% or more, 40% or more, 50% or more. It may be above or below 60%, and especially may be below 70%. The screening method can be performed in vivo or in vitro, and is not particularly limited. Candidate substances may be known substances or new substances, and large-scale screening can be performed through, for example, plant extracts or chemical libraries. Through this, it is possible to discover agents that can suppress cancer, especially brain tumors, by inhibiting the expression or activity of NMUR2.

이하, 본 발명의 이해를 돕기 위하여 바람직한 제조예 및 실험예를 제시한다. 그러나 하기의 제조예 및 실험예는 본 발명을 보다 쉽게 이해하기 위하여 제공되는 것일 뿐, 제조예 및 실험예에 의해 본 발명의 내용이 한정되는 것은 아니다.Below, preferred preparation examples and experimental examples are presented to aid understanding of the present invention. However, the following manufacturing examples and experimental examples are provided only to make the present invention easier to understand, and the content of the present invention is not limited by the manufacturing examples and experimental examples.

[실시예 1][Example 1]

화학식 I의 화합물의 NMUR2 억제 활성 확인 Confirmation of NMUR2 inhibitory activity of compounds of formula I

화학식 I의 화합물의 NMUR2 억제 활성을 확인하기 위하여 NMUR2 발현 벡터를 도입한 세포주를 제작하였다. 구체적으로, CMV 프로모터로 발현되는 pcDNA3.1 플라스미드 벡터에 NMUR2를 암호화하는 염기서열 및 IRES-GFP를 암호화하는 염기서열을 삽입하여 NMUR2-IRES-GFP 발현 벡터를 구축하고, 상기 NMUR2-IRES-GFP 발현 벡터를 beta-arrestin 신호 활성에 의존적으로 루시퍼라제를 발현하는 PRESTO-TANGO HTLA 세포주에 형질 도입하여 HTLA_NMUR2-IRES-GFP 세포를 제작하였다.To confirm the NMUR2 inhibitory activity of the compound of Formula I, a cell line into which an NMUR2 expression vector was introduced was constructed. Specifically, an NMUR2-IRES-GFP expression vector was constructed by inserting a base sequence encoding NMUR2 and a base sequence encoding IRES-GFP into the pcDNA3.1 plasmid vector expressed by the CMV promoter, and expressing the NMUR2-IRES-GFP. HTLA_NMUR2-IRES-GFP cells were created by transducing the vector into the PRESTO-TANGO HTLA cell line, which expresses luciferase in a manner dependent on beta-arrestin signaling activity.

유세포 분석기(FACS)를 활용하여 GFP 형광 단백질을 발현하는 단일 클론을 선별함으로써, 상기 발현 벡터가 성공적으로 형질 도입된 세포주를 확보하였다. 상기 확보된 세포주에 NMUR2의 리간드인 NMU-25를 처리하고 6시간 뒤에 루시퍼라제의 활성이 증가되는 것을 관찰함으로써 NMUR2의 활성이 증가하는 것을 확인하였고, NMUR2에 선택적인 약물 스크리닝 세포주를 제작하였다.By selecting a single clone expressing the GFP fluorescent protein using flow cytometry (FACS), a cell line into which the expression vector was successfully transduced was secured. The obtained cell line was treated with NMU-25, a NMUR2 ligand, and 6 hours later, the activity of luciferase was observed to increase, confirming that the activity of NMUR2 was increased, and a drug screening cell line selective for NMUR2 was created.

96-well 화이트 플레이트 상에서, 발현 벡터가 성공적으로 형질 도입된 상기 세포주 20000개/well의 밀도로 37℃, 5% CO2 인큐베이터로 24시간동안 배양하였다. 이후, DPBS(Dulbecco's Phosphate Buffered Saline)에 희석한 DMSO(100 μM)와 화학식 I의 화합물(100 μM)을 well당 10μl 처리하여 최종 농도가 각각 10μM이 되도록, 또는 화학식 I의 화합물을 각각 0.1, 1, 10, 50, 100, 200μM로 희석한 후 well당 10ul 처리하여 최종 농도가 각각 0, 0.01, 1, 5, 10, 20μM이 되도록 처리하였다. DMSO와 화학식 I의 화합물을 처리하고 15분 후에, 최종 농도가 20nM이 되도록 NMUR2의 리간드인 NMU-25를 10μl 처리하였다.On a 96-well white plate, the cell lines into which the expression vector was successfully transduced were cultured at a density of 20,000 cells/well in a 5% CO 2 incubator at 37°C for 24 hours. Afterwards, 10 μl of DMSO (100 μM) and the compound of Formula I (100 μM) diluted in DPBS (Dulbecco's Phosphate Buffered Saline) were treated so that the final concentration was 10 μM, or the compound of Formula I was treated at 0.1 and 1, respectively. After diluting to 10, 50, 100, and 200μM, 10ul was applied per well so that the final concentrations were 0, 0.01, 1, 5, 10, and 20μM, respectively. 15 minutes after treatment with DMSO and the compound of Formula I, 10 μl of NMU-25, a ligand for NMUR2, was treated to a final concentration of 20 nM.

이후, 세포를 37℃, 5% CO2 인큐베이터에서 6시간동안 배양한 후, 루시퍼라제 활성 용액(One-glo, Promega)을 50μl/well의 밀도로 추가하여 상온에서 5분간 반응시킨 다음, 루시퍼라제 활성을 측정하였다. DMSO만을 처리한 군에 비하여 DMSO와 NMUR2의 리간드인 NMU-25를 함께 처리한 군에서 루시퍼라제 활성이 유의미하게 증가한 반면, NMUR2의 리간드인 NMU-25 및 화학식 I의 화합물을 동시에 처리한 군에서는 루시퍼라제 활성의 변화가 관찰되지 않아, 화학식 I의 화합물이 NMUR2를 억제하는 것을 확인할 수 있었다(도 2a 참조). 상기 조건 및 방법으로 실험을 세 번 반복하였을 때, 실험군에서 화학식 I의 화합물에 의한 NMUR2의 억제 정도는 반복적으로 70 % 이상의 높은 억제 효과를 보였다. Afterwards, the cells were cultured in a 37°C, 5% CO 2 incubator for 6 hours, then luciferase activity solution (One-glo, Promega) was added at a density of 50 μl/well, reacted at room temperature for 5 minutes, and then luciferase Activity was measured. Compared to the group treated with DMSO alone, the luciferase activity significantly increased in the group treated with DMSO and NMU-25, a ligand of NMUR2, while the group treated simultaneously with NMU-25, a ligand of NMUR2, and the compound of Formula I showed luciferase activity. No change in rase activity was observed, confirming that the compound of Formula I inhibits NMUR2 (see Figure 2a). When the experiment was repeated three times under the above conditions and methods, the degree of inhibition of NMUR2 by the compound of Formula I in the experimental group repeatedly showed a high inhibition effect of more than 70%.

NNC 05-2090에 의한 억제효과 (% inhibition)Inhibition effect by NNC 05-2090 (% inhibition) 1차 Primary 78.06 78.06 2차 Secondary 80.79 80.79 3차 3rd 74.26 74.26

또한, 화학식 I의 화합물을 농도별로 처리하였을 때, 농도에 따라 루시퍼라제 활성이 감소하는 것을 확인할 수 있었다(도 2b 참조).위와 같은 결과는, 화학식 I의 화합물은 NMUR2를 효과적으로 억제할 수 있음을 보여준다.In addition, when the compound of Formula I was treated at different concentrations, it was confirmed that luciferase activity decreased depending on the concentration (see Figure 2b). The above results show that the compound of Formula I can effectively inhibit NMUR2. It shows.

[실시예 2][Example 2]

뇌종양 세포주에서 화학식 I의 화합물의 ICIC of compounds of formula I in brain tumor cell lines 5050 확인 check

뇌종양 세포주인 U-87MG, T-98G 및 U-373MG(한국세포주은행)에서 상기 화학식 I의 화합물과 테모졸로마이드의 IC50을 확인하였다. The IC 50 of the compound of Formula I and temozolomide was confirmed in brain tumor cell lines U-87MG, T-98G, and U-373MG (Korea Cell Line Bank).

96-well 배양 플레이트에 U-87MG, U-373MG는 8000개/well, T-98G는 5000개/well의 밀도로 37℃, 5% CO2 인큐베이터에서 24시간동안 배양하였고, 화학식 I의 화합물을 DPBS에 희석하여 최종 농도가 0, 1, 2, 5, 10 및 20 μM 이 되도록 처리하였다. 테모졸로마이드는 0, 106, 213, 425, 850 및 1700 μM의 농도로 배지에 직접 녹인 후 배지를 바꾸는 방식으로 처리하였다. 약물 처리 후 48시간 경과 시, CCK8 키트(Dojindo Molecular Technologies, Inc.)를 이용하여 제조사의 지시에 따라 세포 생존율을 측정하였고, 이에 기반하여 IC50 값을 도출하였다. In a 96-well culture plate, U-87MG, U-373MG were cultured at a density of 8,000/well, and T-98G was 5,000/well in a 37°C, 5% CO 2 incubator for 24 hours, and the compound of Formula I was cultured in a 96-well culture plate. It was diluted in DPBS so that the final concentrations were 0, 1, 2, 5, 10, and 20 μM. Temozolomide was dissolved directly in the medium at concentrations of 0, 106, 213, 425, 850, and 1700 μM and then treated by changing the medium. 48 hours after drug treatment, cell viability was measured using the CCK8 kit (Dojindo Molecular Technologies, Inc.) according to the manufacturer's instructions, and the IC 50 value was derived based on this.

이때, 테모졸로마이드의 IC50은 1000 μM 이상으로 확인되어, 고농도에서도 뇌종양 세포의 억제 효과가 미미한 반면, 화학식 I의 화합물의 IC50는 6 내지 7 μM으로 확인되었다. 즉, 화학식 I의 화합물은 기존 뇌종양 치료제로 사용되고 있는 테모졸로마이드보다 낮은 농도에서 단독으로 사용되어도 뇌종양 세포 성장을 억제할 수 있다(도 3A 참조). At this time, the IC 50 of temozolomide was confirmed to be 1000 μM or more, showing a minimal inhibitory effect on brain tumor cells even at high concentrations, while the IC 50 of the compound of Formula I was confirmed to be 6 to 7 μM. In other words, the compound of Formula I can inhibit brain tumor cell growth even when used alone at a lower concentration than temozolomide, which is currently used as a brain tumor treatment (see Figure 3A).

Temozolomide ICTemozolomide IC 5050 (μM)(μM) 화학식 I의 화합물 ICCompound IC of Formula I 5050 (μM)(μM) U-87MGU-87MG 1462114621 6.4126.412 T-98GT-98G 16661666 6.1606.160 U-373MGU-373MG 26302630 7.9947.994

96-well 화이트 플레이트에 U-87MG, U-373MG는 각 8,000개/well, T-98G는 5000개/well의 밀도로 넣어준 후 37℃, 5% CO2 인큐베이터에서 24시간동안 배양하였고, 화학식 I의 화합물을 각각 0, 5, 10 μM의 농도로 처리하였다. 이후, 48시간이 경과한 뒤에 Caspase 3/7-Glo(Promega) 용액을 30 μl 처리하고 실온, 암조건에서 30분간 대기한 후 세포 사멸율의 변화를 측정하였다. 세 종의 뇌종양 세포주 모두에서 화학식 I의 화합물을 10 μM로 처리하였을 때 유의미하게 세포 사멸 유도가 관찰되었다(도 3B 참조). U-87MG and U-373MG were placed in a 96-well white plate at a density of 8,000 cells/well each, and T-98G was placed at a density of 5,000 cells/well, and then cultured in an incubator at 37°C and 5% CO 2 for 24 hours, and the chemical formula Compound I was treated at concentrations of 0, 5, and 10 μM, respectively. After 48 hours, the cells were treated with 30 μl of Caspase 3/7-Glo (Promega) solution, waited for 30 minutes at room temperature and in the dark, and changes in cell death rate were measured. Significant induction of cell death was observed when treated with 10 μM compound of formula I in all three brain tumor cell lines (see Figure 3B).

[실시예 3][Example 3]

뇌종양 세포주에서 테모졸로마이드 및 화학식 I의 화합물의 병용처리에 의한 상승작용적 효과 확인 Confirmation of synergistic effect by combined treatment of temozolomide and compound of formula I in brain tumor cell lines

뇌종양 세포주인 T-98G에서 테모졸로마이드 및 화학식 I의 화합물을 각각 단독으로 처리하거나 병용으로 처리하였을 때의 세포 성장 및 세포 사멸을 관찰하였다. Cell growth and cell death were observed in T-98G, a brain tumor cell line, when temozolomide and the compound of formula (I) were treated alone or in combination.

뇌종양 세포주 T98G를 96-well 배양 플레이트와 96-well 화이트 플레이트에 3000개/well의 밀도로 24시간동안 배양한 뒤에 테모졸로마이드를 최종 농도 1mM으로 단독으로 처리하였을 때에 비하여, 화학식 I의 화합물을 최종 농도 10μM으로 단독으로 처리하였을 때 세포 성장율 및 세포 사멸율이 증가하였다. 또, 화학식 I의 화합물을 단독으로 처리하였을 때보다 테모졸로마이드와 위와 동일한 용량으로 병용 처리하였을 때 더 높은 세포 성장율 및 세포 사멸율을 나타내었다(도 4A 참조). Compared to when the brain tumor cell line T98G was cultured in a 96-well culture plate and a 96-well white plate at a density of 3000 cells/well for 24 hours and then treated alone with temozolomide at a final concentration of 1mM, the final concentration of the compound of Formula I was When treated alone at a concentration of 10 μM, the cell growth rate and cell death rate increased. In addition, higher cell growth and cell death rates were observed when treated in combination with temozolomide at the same dose as above, compared to when the compound of Formula I was treated alone (see Figure 4A).

이를 통하여 테모졸로마이드 및 화학식 I의 화합물의 병용 처리에 의한 뇌종양 환자의 치료 효과 증대를 기대할 수 있다.Through this, it can be expected that the treatment effect of brain tumor patients will be increased by the combined treatment of temozolomide and the compound of formula (I).

[실시예 4][Example 4]

뇌종양 세포주에서 테모졸로마이드 및 화학식 I의 화합물의 병용처리에 의한 병용지수 확인 Confirmation of combination index by combined treatment of temozolomide and compound of formula I in brain tumor cell lines

약물간 상호작용의 정도를 나타내는 병용지수(CI; Combination Index)를 확인하기 위하여, 뇌종양 세포주 T-98G를 96-well 플레이트에 3000개/well의 세포로 24시간 배양한 뒤에 테모졸로마이드를 농도 0 내지 1000 mM로 처리하고 동시에 화학식 I의 화합물을 농도 1 내지 20 μM로 처리하였다. 최고 농도 대비 1/2로 희석하여 처리하였을 경우에 서로에게 영향을 주는 정도를 병용지수의 값으로 계산하였다. 병용지수의 값이 1.0 미만인 것이 상승작용이 있음을 나타내는 것으로서, 테모졸로마이드와 화학식 I의 화합물을 병용으로 처리하였을 때 CI=0.686로 계산되어 상승효과가 있음을 확인하였다(도 4B 참조).To check the combination index (CI), which indicates the degree of drug-drug interaction, the brain tumor cell line T-98G was cultured in a 96-well plate at 3000 cells/well for 24 hours, and then temozolomide was added at a concentration of 0. to 1000 mM and simultaneously treated with the compound of formula I at a concentration of 1 to 20 μM. The degree to which they influence each other when diluted to 1/2 of the highest concentration was calculated as the value of the combination index. A combination index value of less than 1.0 indicates a synergistic effect, and when temozolomide and the compound of Formula I were treated in combination, CI = 0.686 was calculated, confirming the existence of a synergistic effect (see Figure 4B).

[실시예 5][Example 5]

위암, 대장암 및 피부암 세포주에서 화학식 I의 화합물의 ICIC of compounds of formula I in gastric, colon and skin cancer cell lines 5050 확인 check

위암, 대장암 및 피부암의 세포주인 SNU-638, HCT116 및 SK-MEL-2를 96-well 플레이트에 3000개/well의 밀도로 37℃, 5% CO2 인큐베이터에서 24시간동안 배양한 뒤에, HCT116과 SK-MEL-2는 화학식 I의 화합물(최종 농도 0~20 μM)을 최고 농도 대비 1/2로 희석하여 처리하였고, SNU-638의 경우에는 화학식 I의 화합물(최종 농도 0~100 μM)을 최고 농도 대비 1/2로 희석하여, 10 μl씩 처리하였다. 약물 처리 후에 48시간동안 배양한 후에, CCK8 키트를 이용하여 제조사의 지시에 따라 세포 성장률을 측정하였다.SNU-638, HCT116, and SK-MEL-2, which are cell lines for stomach cancer, colon cancer, and skin cancer, were cultured in a 96-well plate at a density of 3000 cells/well for 24 hours in a 5% CO2 incubator at 37°C, and then HCT116 and SK-MEL-2 was treated with a compound of formula I (final concentration 0~20 μM) diluted to 1/2 of the highest concentration, and for SNU-638, a compound of formula I (final concentration 0~100 μM) was treated. It was diluted to 1/2 of the highest concentration and treated with 10 μl each. After culturing for 48 hours after drug treatment, cell growth rate was measured using the CCK8 kit according to the manufacturer's instructions.

아래 표 3과 같이, 뇌종양 세포주에서와 유사한 IC50 값을 나타내었다(도 5 참조).As shown in Table 3 below, IC 50 values were similar to those in brain tumor cell lines (see Figure 5).

세포주cell line ICIC 5050 (μM)(μM) SNU-638SNU-638 9.6579.657 HCT116HCT116 8.3268.326 SK-MEL-2SK-MEL-2 5.1565.156

이러한 수치는 화학식 I의 화합물이 뇌종양 이외의 다른 암종에서도 세포의 성장을 억제하는 효과를 나타내는 결과이다.These values indicate that the compound of Formula I has an effect of inhibiting cell growth in cancer types other than brain tumors.

[실시예 6][Example 6]

NUMR2 발현과 신경교종 및 교모세포종 환자의 유전자 변이 간 상관관계 분석Correlation analysis between NUMR2 expression and genetic mutations in glioma and glioblastoma patients

게놈 시퀀싱 및 생물정보학을 사용하여 암의 원인이 되는 유전적 돌연변이를 목록화한 The Cancer Genome Atlas(TCGA)을 이용하여, 교모세포종(glioblastoma) 및 신경교종(glioma) 샘플들에서 유전적 돌연변이와 NMUR2 발현 간의 상관 관계를 분석하였다.Genetic mutations and NMUR2 were identified in glioblastoma and glioma samples using The Cancer Genome Atlas (TCGA), which catalogs cancer-causing genetic mutations using genome sequencing and bioinformatics. The correlation between expressions was analyzed.

그 결과, 도 6에 나타난 바와 같이, 시토신-포스페이트-구아닌(CpG) 섬 메틸화 표현형(CpG island methylator phenotype, G-CIMP)이 높게 발생하는 경우의 샘플은 236개인데 반해, 낮게 발생하는 경우의 샘플은 17개로 나타나, G-CIMP가 높게 발생하는 경우가 낮게 발생하는 경우보다 NMUR2 발현이 유의미하게 증가한 것을 확인하였다.As a result, as shown in Figure 6, there are 236 samples in cases where the cytosine-phosphate-guanine (CpG) island methylator phenotype (G-CIMP) occurs at a high level, whereas there are 236 samples in cases where the cytosine-phosphate-guanine (CpG) island methylator phenotype (G-CIMP) occurs at a low level. was found to be 17, and it was confirmed that NMUR2 expression was significantly increased in cases where G-CIMP occurred at high levels compared to cases where G-CIMP occurred at low levels.

위와 같은 결과는 G-CIMP가 높게 발생하는 교모세포종 또는 신경교종의 경우, NUMR2의 발현도 높아져 있음을 보여준다.The above results show that in the case of glioblastoma or glioma with a high level of G-CIMP, the expression of NUMR2 is also high.

[실시예 7][Example 7]

NMUR2 과발현 세포주 제작Construction of NMUR2 overexpressing cell lines

NMUR2 과발현 세포주를 제작하기 위하여, NMUR2 발현 벡터를 도입한 세포주를 제작하였다.In order to create a cell line overexpressing NMUR2, a cell line into which an NMUR2 expression vector was introduced was created.

구체적으로, pcDNA3.1 플라스미드 벡터에 NMUR2를 암호화하는 염기서열을 삽입하여, pcDNA3.1 NMUR2-과발현 벡터를 구축하고, 교모세포종 세포주인 T-98G에 각각 pcDNA 공벡터와 pcDNA3.1 NMUR2-과발현 벡터를 리포펙타민 2000 (Thermo Scientific)을 이용하여 형질 도입하고, 72시간 뒤 RNA 추출 및 cDNA 합성 후, NMUR2의 과발현 여부를 qPCR로 확인하였다. qPCR에 사용된 프라이머의 염기 서열은 아래 표 4와 같다.Specifically, the nucleotide sequence encoding NMUR2 was inserted into the pcDNA3.1 plasmid vector to construct a pcDNA3.1 NMUR2-overexpression vector, and the pcDNA empty vector and pcDNA3.1 NMUR2-overexpression vector were added to the glioblastoma cell line T-98G, respectively. was transduced using Lipofectamine 2000 (Thermo Scientific), and after RNA extraction and cDNA synthesis 72 hours later, overexpression of NMUR2 was confirmed by qPCR. The nucleotide sequences of the primers used in qPCR are shown in Table 4 below.

명칭designation 서열(5'→3')Sequence (5'→3') 서열번호sequence number NMUR2 정방향NMUR2 forward AGA TGT GGC GCA ACT ACC CAGA TGT GGC GCA ACT ACC C 1One NMUR2 역방향NMUR2 reverse CGA AGC ACA CGG TCT CAA AGACGA AGC ACA CGG TCT CAA AGA 22 GAPDH 정방향GAPDH forward CTC TGC TCC TCCTGT TCG ACCTC TGC TCC TCCTGT TCG AC 33 GAPDH 역방향GAPDH reverse TTA AAA GCA GCC CTG GTG ACTTA AAA GCA GCC CTG GTG AC 44

그 결과, 도 7에 나타난 바와 같이, pcDNA 공벡터가 도입된 교모세포종 세포주와 달리, pcDNA3.1 NMUR2-과발현 벡터가 도입된 교모세포종 세포주는 NMUR2를 높은 수준으로 발현하는 것을 확인하였다.As a result, as shown in Figure 7, unlike the glioblastoma cell line into which the pcDNA empty vector was introduced, the glioblastoma cell line into which the pcDNA3.1 NMUR2-overexpression vector was introduced was confirmed to express NMUR2 at a high level.

[실시예 8][Example 8]

NMUR2 과발현 세포주의 세포 증식도 분석Cell proliferation analysis of NMUR2 overexpressing cell lines

상기 실시예 7에서 제작한 pcDNA 공벡터가 도입된 교모세포종 세포주와 pcDNA3.1 NMUR2-과발현 벡터가 도입된 교모세포종 세포주에서 세포 증식도를 분석하였다.Cell proliferation was analyzed in the glioblastoma cell line introduced with the pcDNA empty vector prepared in Example 7 and the glioblastoma cell line introduced with the pcDNA3.1 NMUR2-overexpression vector.

구체적으로, pcDNA 공벡터가 도입된 교모세포종 세포주와 pcDNA3.1 NMUR2-과발현 벡터가 도입된 교모세포종 세포주를 96-웰 플레이트에 3000개/웰의 밀도로 분주하고, 37℃, 5% CO2 배양 후, 24, 48, 72, 96 시간 후에 CCK8 키트(Dojindo Molecular Technologies, Inc.)를 이용하여 세포의 증식도를 분석하였다.Specifically, the glioblastoma cell line into which the pcDNA empty vector was introduced and the glioblastoma cell line into which the pcDNA3.1 NMUR2-overexpression vector was introduced were distributed in a 96-well plate at a density of 3000 cells/well and incubated at 37°C with 5% CO 2 . After 24, 48, 72, and 96 hours of culture, cell proliferation was analyzed using the CCK8 kit (Dojindo Molecular Technologies, Inc.).

그 결과, 도 8에 나타난 바와 같이, NMUR2 과발현 세포주에서 교모세포종 세포의 증식도가 대조군에 비하여 증가하는 것을 확인하였다.As a result, as shown in Figure 8, it was confirmed that the proliferation rate of glioblastoma cells in the NMUR2 overexpressing cell line increased compared to the control group.

위와 같은 결과는 NMUR2가 과발현되는 교모세포종에서 암 세포가 빠른 속도로 증식할 수 있음을 보여준다.The above results show that cancer cells can proliferate at a rapid rate in glioblastoma where NMUR2 is overexpressed.

[실시예 9][Example 9]

NMUR2 과발현 세포주의 세포 이동 분석Cell migration analysis of NMUR2 overexpressing cell lines

상기 실시예 7에서 제작한 pcDNA 공벡터가 도입된 교모세포종 세포주와 pcDNA3.1 NMUR2-과발현 벡터가 도입된 교모세포종 세포주에서 세포 이동을 분석하였다.Cell migration was analyzed in the glioblastoma cell line introduced with the pcDNA empty vector prepared in Example 7 and the glioblastoma cell line introduced with the pcDNA3.1 NMUR2-overexpression vector.

구체적으로, pcDNA 공벡터가 도입된 교모세포종 세포주와 pcDNA3.1 NMUR2-과발현 벡터가 도입된 교모세포종 세포주를 8μm 24-웰 'insert'(FALCON)의 upper chamber에 각각 5x104 개씩 500μl의 무혈청 배지와 함께 넣어주었다. Lower chamber에는 10%의 FBS가 포함된 배지 500㎕로 채웠다. 이후에 transwell을 37℃, 5% CO2로 18시간 동안 배양하였으며, 증류수를 이용하여 4%로 희석한 포름알데히드와 100% 메탄올로 고정한 뒤 20X PBS(LPS solution)를 이용하여 0.1%로 희석한 crystal violet (SIGMA)으로 염색하였다.Specifically, the glioblastoma cell line introduced with the pcDNA empty vector and the glioblastoma cell line introduced with the pcDNA3.1 NMUR2-overexpression vector were placed in the upper chamber of an 8μm 24-well 'insert' (FALCON) at 5x10 cells each in 500μl of serum-free medium. It was put in with. The lower chamber was filled with 500 μl of medium containing 10% FBS. Afterwards, the transwell was cultured at 37°C and 5% CO 2 for 18 hours, fixed with formaldehyde diluted to 4% using distilled water and 100% methanol, and then diluted to 0.1% using 20X PBS (LPS solution). Stained with crystal violet (SIGMA).

그 결과, 도 9에 나타난 바와 같이, NMUR2 과발현 세포주에서 교모세포종 세포의 이동이 대조군에 비하여 증가하는 것을 확인하였다.As a result, as shown in Figure 9, it was confirmed that the migration of glioblastoma cells in the NMUR2 overexpressing cell line increased compared to the control group.

위와 같은 결과는 NMUR2가 과발현되는 교모세포종에서 암 세포의 이동이 증가되어 주변 조직으로 전이될 수 있음을 보여준다.The above results show that the migration of cancer cells in glioblastoma overexpressing NMUR2 is increased and can metastasize to surrounding tissues.

[실시예 10][Example 10]

NMUR2에 대한 siRNA가 도입된 교모세포종에서 NMUR2 발현 변화 확인Confirmation of NMUR2 expression changes in glioblastoma introduced with siRNA against NMUR2

NMUR2에 대한 siRNA가 도입된 교모세포종에서 NMUR2 발현 변화를 확인하기 위하여, 교모세포종 세포주인 T-98G에 각각 siNMUR2-1, siNMUR2-2를 lipofectamineRNAiMAX (Thermo Scientific)를 이용하여 형질 도입하고(아래 표 5), 72시간 뒤에 상기 실시예 7에 기재된 방법으로 qPCR을 진행하여, NMUR2의 발현 수준을 측정하였다.To confirm changes in NMUR2 expression in glioblastoma into which siRNA against NMUR2 was introduced, siNMUR2-1 and siNMUR2-2 were transduced into T-98G, a glioblastoma cell line, respectively, using lipofectamineRNAiMAX (Thermo Scientific) (Table 5 below). ), 72 hours later, qPCR was performed using the method described in Example 7 above to measure the expression level of NMUR2.

명칭designation 서열(5'→3')Sequence (5'→3') 서열번호sequence number siNMUR2-1 sensesiNMUR2-1 sense AGA AUG CUU CCU GGA UCU AAGA AUG CUU CCU GGA UCU A 55 siNMUR2-1 anti-sensesiNMUR2-1 anti-sense UAG AUG CAG GAA GCA UUC UUAG AUG CAG GAA GCA UUC U 66 siNMUR2-2 sensesiNMUR2-2 sense GCU GAC CGA AGA UAU AGGGCU GAC CGA AGA UAU AGG 77 siNMUR2-2 anti-sensesiNMUR2-2 anti-sense ACC UAU AUC UUC GGU CAG CACC UAU AUC UUC GGU CAG C 88

그 결과, 도 10에 나타난 바와 같이, NMUR2에 대한 siRNA를 처리한 교모세포종 세포주에서 NMUR2의 발현이 유의미하게 감소한 것을 확인하였다.As a result, as shown in Figure 10, it was confirmed that the expression of NMUR2 was significantly decreased in glioblastoma cell lines treated with siRNA against NMUR2.

[실시예 11][Example 11]

NMUR2에 대한 siRNA가 도입된 교모세포종의 세포 증식도 분석Analysis of cell proliferation of glioblastoma introduced with siRNA against NMUR2

상기 실시예 10에서 제작한 교모세포종 세포주인 T-98G에 각각 siNMUR2-1, siNMUR2-2를 형질 도입한 세포주에서 세포 증식도를 분석하였다.Cell proliferation was analyzed in the cell lines transduced with siNMUR2-1 and siNMUR2-2, respectively, into T-98G, a glioblastoma cell line prepared in Example 10.

구체적으로, 교모세포종 세포주인 T-98G에 각각 siNMUR2-1, siNMUR2-2를 형질 도입한 세포주를 96-웰 플레이트에 3000개/웰의 밀도로 분주하고, 37℃, 5% CO2 배양 후, 24, 48, 72, 96 시간 후에 CCK8 키트(Dojindo Molecular Technologies, Inc.)를 이용하여 세포의 증식도를 분석하였다.Specifically, the cell lines transduced with siNMUR2-1 and siNMUR2-2, respectively, into T-98G, a glioblastoma cell line, were distributed at a density of 3000 cells/well in a 96-well plate, and incubated at 37°C with 5% CO 2 . After 24, 48, 72, and 96 hours of culture, cell proliferation was analyzed using the CCK8 kit (Dojindo Molecular Technologies, Inc.).

그 결과, 도 11에 나타난 바와 같이, NMUR2에 대한 siRNA를 처리한 교모세포종 세포주에서 세포의 증식도가 대조군에 비하여 감소하는 것을 확인하였다.As a result, as shown in Figure 11, it was confirmed that the proliferation rate of cells in glioblastoma cell lines treated with siRNA against NMUR2 was decreased compared to the control group.

위와 같은 결과는 NMUR2에 대한 siRNA가 교모세포종의 성장을 억제하여, 교모세포종의 예방 또는 치료제로 사용될 수 있음을 보여준다.The above results show that siRNA against NMUR2 inhibits the growth of glioblastoma and can be used as a preventive or therapeutic agent for glioblastoma.

[실시예 12][Example 12]

NMUR2에 대한 siRNA가 도입된 교모세포종의 세포 이동 분석Cell migration analysis of glioblastoma introduced with siRNA against NMUR2

상기 실시예 10에서 제작한 교모세포종 세포주인 T-98G에 각각 siNMUR2-1, siNMUR2-2를 형질 도입한 세포주에서 세포 이동을 분석하였다.Cell migration was analyzed in T-98G, a glioblastoma cell line prepared in Example 10, in which siNMUR2-1 and siNMUR2-2 were transduced, respectively.

구체적으로, 교모세포종 세포주인 T-98G에 각각 siNMUR2-1, siNMUR2-2를 형질 도입한 세포주를 8μm 24-웰 'insert'(FALCON)의 upper chamber에 각각 5x104 개씩 500μl의 무혈청 배지와 함께 넣어주었다. Lower chamber에는 10%의 FBS가 포함된 배지 500㎕로 채웠다. 이후에 transwell을 37℃, 5% CO2로 18시간 동안 배양하였으며, 증류수를 이용하여 4%로 희석한 포름알데히드와 100% 메탄올로 고정한 뒤 20X PBS(LPS solution)를 이용하여 0.1%로 희석한 crystal violet (SIGMA)으로 염색하였다.Specifically, the cell lines transduced with siNMUR2-1 and siNMUR2-2, respectively, into T-98G, a glioblastoma cell line, were placed in the upper chamber of an 8μm 24-well 'insert' (FALCON), 5x104 each, along with 500μl of serum-free medium. I put it in. The lower chamber was filled with 500 μl of medium containing 10% FBS. Afterwards, the transwell was cultured at 37°C and 5% CO 2 for 18 hours, fixed with formaldehyde diluted to 4% using distilled water and 100% methanol, and then diluted to 0.1% using 20X PBS (LPS solution). Stained with crystal violet (SIGMA).

그 결과, 도 12에 나타난 바와 같이, NMUR2에 대한 siRNA를 처리한 교모세포종 세포주에서 세포의 이동 대조군에 비하여 감소하는 것을 확인하였다.As a result, as shown in Figure 12, it was confirmed that cell migration was decreased in glioblastoma cell lines treated with siRNA against NMUR2 compared to the control group.

위와 같은 결과는 NMUR2에 대한 siRNA가 교모세포종의 이동을 억제하여, 암의 전이를 억제할 수 있어, 교모세포종의 예방 또는 치료제로 사용될 수 있음을 보여준다.The above results show that siRNA against NMUR2 can inhibit the migration of glioblastoma and metastasis of cancer, and can be used as a preventive or therapeutic agent for glioblastoma.

[실시예 13][Example 13]

화학식 1의 화합물이 교모세포종의 세포 이동에 미치는 영향 분석Analysis of the effect of compounds of formula 1 on cell migration of glioblastoma

NMUR2을 억제하는 화합물인 화학식 1의 화합물이 교모세포종 세포주의 세포 이동에 미치는 영향을 분석하였다.The effect of the compound of Formula 1, a compound that inhibits NMUR2, on cell migration of glioblastoma cell lines was analyzed.

구체적으로, 교모세포종 세포주인 U-87MG, T-98G, U-373MG에 upper chamber에 각각 1x104 개씩 500㎕의 무혈청 배지와 함께 넣어주었으며, 배지에서의 화학식 1 화합물 농도가 각각 0(DMSO), 5, 10, 20μM이 되도록 화학식 1 화합물을 처리하였다. Lower chamber에는 10%의 FBS가 포함된 배지 500㎕로 채웠다. 이후에 transwell을 37℃, 5% CO2로 20시간 동안 배양하였으며, 증류수를 이용하여 4%로 희석한 포름알데히드와 100% 메탄올로 고정한 뒤 20X PBS(LPS solution)를 이용하여 0.1%로 희석한 crystal violet (SIGMA)으로 염색하였다.Specifically, glioblastoma cell lines U-87MG, T-98G, and U-373MG were placed in an upper chamber at 1x10 cells each with 500 ㎕ of serum-free medium, and the concentration of the compound of Formula 1 in the medium was 0 (DMSO). , the compound of Formula 1 was treated to concentrations of 5, 10, and 20 μM. The lower chamber was filled with 500 μl of medium containing 10% FBS. Afterwards, the transwell was cultured at 37°C and 5% CO 2 for 20 hours, fixed with formaldehyde diluted to 4% using distilled water and 100% methanol, and then diluted to 0.1% using 20X PBS (LPS solution). Stained with crystal violet (SIGMA).

그 결과, 도 13에 나타난 바와 같이, IC50값 농도 이상인 10μM과 20μM 사이에서 이동한 세포의 수가 급격하게 감소한 것을 확인하였다.As a result, as shown in Figure 13, it was confirmed that the number of cells migrating between 10 μM and 20 μM, which is above the IC 50 value concentration, decreased rapidly.

위와 같은 결과는 NMUR2에 대한 억제제인 화학식 1의 화합물이 교모세포종의 이동을 억제하여, 암의 전이를 억제할 수 있어, 교모세포종의 예방 또는 치료제로 사용될 수 있음을 보여준다.The above results show that the compound of Formula 1, which is an inhibitor for NMUR2, can inhibit the migration of glioblastoma and inhibit cancer metastasis, and can be used as a preventive or therapeutic agent for glioblastoma.

Claims (13)

NMUR2 발현 억제제 또는 NMUR2 활성 억제제를 유효성분으로 포함하는 암의 예방 또는 치료용 약학적 조성물. A pharmaceutical composition for preventing or treating cancer comprising an NMUR2 expression inhibitor or an NMUR2 activity inhibitor as an active ingredient. 청구항 1에 있어서,In claim 1, 상기 NMUR2 발현 억제제는 miRNA, siRNA 및 shRNA 로 이루어진 군으로부터 선택되는 것이고, 상기 NMUR2 활성 억제제는 항체, 앱타머 및 저분자 화합물로 이루어진 군으로부터 선택되는 것인, 암의 예방 또는 치료용 약학적 조성물. The NMUR2 expression inhibitor is selected from the group consisting of miRNA, siRNA, and shRNA, and the NMUR2 activity inhibitor is selected from the group consisting of antibodies, aptamers, and small molecule compounds. A pharmaceutical composition for preventing or treating cancer. 청구항 1에 있어서,In claim 1, 상기 NMUR2 발현 억제제는 서열번호 5 및 서열번호 6으로 표시되는 염기 서열을 포함하는 것인, 암의 예방 또는 치료용 약학적 조성물.The NMUR2 expression inhibitor is a pharmaceutical composition for preventing or treating cancer, comprising the base sequences represented by SEQ ID NO: 5 and SEQ ID NO: 6. 청구항 1에 있어서,In claim 1, 상기 NMUR2 발현 억제제는 서열번호 7 및 서열번호 8로 표시되는 염기 서열을 포함하는 것인, 암의 예방 또는 치료용 약학적 조성물.The NMUR2 expression inhibitor is a pharmaceutical composition for preventing or treating cancer, comprising the base sequences represented by SEQ ID NO: 7 and SEQ ID NO: 8. 청구항 1에 있어서,In claim 1, 상기 NMUR2 활성 억제제는 하기 화학식을 갖는 화합물 또는 그의 제약상 허용되는 염을 포함하는 것인, 암의 예방 또는 치료용 약학적 조성물. A pharmaceutical composition for preventing or treating cancer, wherein the NMUR2 activity inhibitor includes a compound having the following formula or a pharmaceutically acceptable salt thereof. <화학식 I><Formula I>
Figure PCTKR2023003111-appb-img-000003
Figure PCTKR2023003111-appb-img-000003
청구항 1 내지 청구항 5 중 어느 한 항에 있어서, The method according to any one of claims 1 to 5, 상기 암은 뇌종양, 중추신경계 암, 신경교종, 교모세포종, 뇌수막증, 뇌하수체선종, 전이성 뇌종양, 청신경초종, 위암, 대장암 및 피부암으로 이루어진 군으로부터 선택되는 것인, 암의 예방 또는 치료용 약학적 조성물. The cancer is selected from the group consisting of brain tumor, central nervous system cancer, glioma, glioblastoma, meningitis, pituitary adenoma, metastatic brain tumor, acoustic neuroma, stomach cancer, colon cancer, and skin cancer, and pharmaceuticals for the prevention or treatment of cancer. Composition. 청구항 1 내지 청구항 5 중 어느 한 항에 있어서, The method according to any one of claims 1 to 5, 상기 암은 신경교종, 교모세포종, 뇌수막증, 뇌하수체선종, 전이성 뇌종양 및 청신경초종으로 이루어진 군으로부터 선택되는 것인, 암의 예방 또는 치료용 약학적 조성물.A pharmaceutical composition for preventing or treating cancer, wherein the cancer is selected from the group consisting of glioma, glioblastoma, meningitis, pituitary adenoma, metastatic brain tumor, and acoustic neuroma. 청구항 1 내지 청구항 5 중 어느 한 항에 있어서, The method according to any one of claims 1 to 5, 상기 암은 위암, 대장암 및 피부암으로 이루어진 군으로부터 선택되는 것인, 암의 예방 또는 치료용 약학적 조성물.A pharmaceutical composition for preventing or treating cancer, wherein the cancer is selected from the group consisting of stomach cancer, colon cancer, and skin cancer. 청구항 1 내지 청구항 5 중 어느 한 항에 있어서, The method according to any one of claims 1 to 5, 상기 암은 NMUR2(Neuromedin U Receptor 2)가 과발현된 것인, 암의 예방 또는 치료용 약학적 조성물.A pharmaceutical composition for preventing or treating cancer, wherein the cancer is one in which NMUR2 (Neuromedin U Receptor 2) is overexpressed. 청구항 1 내지 청구항 5 중 어느 한 항의 약학적 조성물; 및 The pharmaceutical composition of any one of claims 1 to 5; and 항암제;anticancer drugs; 를 포함하는, 암의 예방 또는 치료용 약학적 키트.A pharmaceutical kit for preventing or treating cancer, comprising: 청구항 10에 있어서,In claim 10, 상기 항암제는 테모졸로마이드(Temozolomide)인 것인, 키트.The kit, wherein the anticancer agent is temozolomide. NMUR2가 과발현된 세포에, 항암제 후보 물질을 처리하는 단계; 및Treating cells overexpressing NMUR2 with an anticancer drug candidate; and 상기 후보 물질이 처리된 세포에서 NMUR2의 발현 또는 활성 수준을 측정하는 단계;Measuring the expression or activity level of NMUR2 in cells treated with the candidate substance; 를 포함하는 항암제 스크리닝 방법.Anticancer drug screening method comprising. 청구항 12에 있어서,In claim 12, 상기 측정된 NMUR2의 발현 또는 활성 수준이, 후보 물질이 처리되지 않은 세포에 비해 낮은 경우, 상기 후보 물질을 항암제로 판정하는 단계;를 더 포함하는, 항암제 스크리닝 방법.If the measured expression or activity level of NMUR2 is lower than that of cells not treated with the candidate substance, determining the candidate substance as an anticancer drug.
PCT/KR2023/003111 2022-03-08 2023-03-07 Composition for preventing or treating cancer comprising nmur2 inhibitor as active ingredient Ceased WO2023172032A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2022-0029570 2022-03-08
KR20220029570 2022-03-08

Publications (1)

Publication Number Publication Date
WO2023172032A1 true WO2023172032A1 (en) 2023-09-14

Family

ID=87935495

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/003111 Ceased WO2023172032A1 (en) 2022-03-08 2023-03-07 Composition for preventing or treating cancer comprising nmur2 inhibitor as active ingredient

Country Status (2)

Country Link
KR (1) KR20230132715A (en)
WO (1) WO2023172032A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6071932A (en) * 1995-05-05 2000-06-06 British Technology Group Intercorporate Licensing Limited Carbazolypiperines as GABA uptake inhibitors
WO2004067725A2 (en) * 2003-01-30 2004-08-12 Regeneron Pharmaceuticals, Inc. Methods of identifying modulators of nmur2-mediated activity

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6071932A (en) * 1995-05-05 2000-06-06 British Technology Group Intercorporate Licensing Limited Carbazolypiperines as GABA uptake inhibitors
WO2004067725A2 (en) * 2003-01-30 2004-08-12 Regeneron Pharmaceuticals, Inc. Methods of identifying modulators of nmur2-mediated activity

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
PENG SHENG, LU YINGJUN, LI PENGYI, LIU PEIRONG, SHI XIAOWEI, LIU CHUNLIANG, ZHANG YU, LIU SHASHA, WANG JING: "The short interference RNA (siRNA) targeting NMUR2 relieves nociception in a bone cancer pain model of rat through PKC-ERK and PI3K-AKT pathways", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, ELSEVIER, AMSTERDAM NL, vol. 512, no. 3, 1 May 2019 (2019-05-01), Amsterdam NL , pages 616 - 622, XP093089575, ISSN: 0006-291X, DOI: 10.1016/j.bbrc.2019.03.067 *
PENG YUAN, HUANG JINSHENG, XIAO HONG, WU TENG, SHUAI XINTAO: "Codelivery of temozolomide and siRNA with polymeric nanocarrier for effective glioma treatment", INTERNATIONAL JOURNAL OF NANOMEDICINE, vol. Volume 13, 1 June 2018 (2018-06-01), pages 3467 - 3480, XP093089576, DOI: 10.2147/IJN.S164611 *
PRZYGODZKA PATRYCJA, SOCHACKA EWELINA, SOBOSKA KAMILA, PACHOLCZYK MARCIN, PAPIEWSKA-PAJĄK IZABELA, PRZYGODZKI TOMASZ, PŁOCIŃSKI PR: "Neuromedin U induces an invasive phenotype in CRC cells expressing the NMUR2 receptor", JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH, vol. 40, no. 1, 1 December 2021 (2021-12-01), pages 283, XP093089627, DOI: 10.1186/s13046-021-02073-8 *
ROH ROHYUNA: "Target validation and inhibitor exploration screening of NMUR2 for glioblastoma treatment", MASTER'S THESIS, BIOSCIENCE (FUNCTIONAL GENOMICS), UNIVERSITY OF SCIENCE AND TECHNOLOGY, 1 February 2023 (2023-02-01), University of Science And Technology, XP093089633, Retrieved from the Internet <URL:http://www.riss.or.kr/search/detail/DetailView.do?p_mat_type=be54d9b8bc7cdb09&control_no=0d3b8359ab672048ffe0bdc3ef48d419&keyword=> [retrieved on 20231009] *

Also Published As

Publication number Publication date
KR20230132715A (en) 2023-09-18

Similar Documents

Publication Publication Date Title
Erik et al. Antiapoptotic action of hypoxia-inducible factor-1α in human endothelial cells
WO2020080861A1 (en) Gastric cancer treatment composition comprising syt11 inhibitor as active ingredient
WO2012064146A2 (en) Anticancer composition containing gkn 1
WO2021241981A1 (en) Composition for preventing or treating cancer
WO2018004240A1 (en) Use of nupr1 in diagnosis and treatment of brain tumor
WO2016018089A1 (en) Novel biomarker for predicting sensitivity to parp inhibitor, and use thereof
WO2010062143A9 (en) Anticancer composition comprising antitumor agent and substance having inhibitory effects on l1cam activity and expression
WO2023172032A1 (en) Composition for preventing or treating cancer comprising nmur2 inhibitor as active ingredient
WO2023096126A1 (en) Method for screening mdsc inhibitor
WO2023163497A1 (en) Pharmaceutical composition for preventing or treating colorectal cancer comprising ednra inhibitor as active ingredient
WO2018105921A2 (en) Use of leucine-zipper protein for diagnosis or treatment of fatty liver
WO2017164568A1 (en) Method for predicting survival rate and prognosis of esophageal cancer patient by measuring protein expression level of v-atpase subunit v1e1
WO2017007241A1 (en) Method for determining sensitivity to simultaneous inhibitor against parp and tankyrase
WO2024177416A1 (en) Pharmaceutical composition for treating egfr targeted therapy-resistant cancer
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
WO2017061828A1 (en) Pharmaceutical composition for preventing or treating cancer comprising inhibitor of plrg1 (pleiotropic regulator 1) as active ingredient
WO2023068820A1 (en) Composition for treating or preventing cancer
WO2011087343A2 (en) Composition for treating cancer related to an hpv infection
KR102080223B1 (en) Pharmaceutical composition for treating xCT inhibitor- resistant cancer
WO2016200246A1 (en) Novel biomarker for diagnosing resistance to anticancer agent for biliary tract cancer and use thereof
WO2021137506A1 (en) Composition for inhibiting growth of cancer stem cells, containing wdr34 inhibitor, and use thereof
WO2021075853A1 (en) Composition comprising emp3 inhibitor for inhibiting growth of cancer stem cell and use thereof
WO2025178391A1 (en) Pharmaceutical composition containing odc1 inhibitor as active ingredient for inhibiting anticancer drug tolerance, recovering anticancer drug responsiveness, or enhancing anticancer drug sensitivity
WO2023075123A1 (en) Pharmaceutical composition for treatment and metastasis inhibition of colorectal cancer, comprising mirnas, and use thereof
WO2015056970A1 (en) Composition for preventing or treating osteoporosis, containing progranulin inhibitor as active ingredient

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: 23767133

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: 23767133

Country of ref document: EP

Kind code of ref document: A1