WO2016013909A1 - Method for screening therapeutic agent for cancer using interaction between ikba and aurkc - Google Patents

Method for screening therapeutic agent for cancer using interaction between ikba and aurkc Download PDF

Info

Publication number
WO2016013909A1
WO2016013909A1 PCT/KR2015/007751 KR2015007751W WO2016013909A1 WO 2016013909 A1 WO2016013909 A1 WO 2016013909A1 KR 2015007751 W KR2015007751 W KR 2015007751W WO 2016013909 A1 WO2016013909 A1 WO 2016013909A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
protein
ikba
aurkc
fluorescent protein
Prior art date
Application number
PCT/KR2015/007751
Other languages
French (fr)
Korean (ko)
Inventor
정영호
한은희
김남두
김현경
Original Assignee
한국기초과학지원연구원
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 한국기초과학지원연구원 filed Critical 한국기초과학지원연구원
Publication of WO2016013909A1 publication Critical patent/WO2016013909A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor 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
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances

Definitions

  • the present invention relates to a method for screening cancer therapeutic agents through the interaction of IkBa and AURKC. More particularly, the present invention relates to a binding inhibitor compound of ikBa and AURKC and a composition for preventing and treating cancer comprising the same as an active ingredient. .
  • IkBa (nuc lear factor of kappa light polypeptide gene enhancer in B eel I s inhibitor, alpha) protein is a type of control subunit that inhibits NF- ⁇ ⁇ activity.
  • IkBa binds to the nuclear position signal (NLS) site of NF-kB to mask the nuclear position signal so that NF-kB remains inactivated in the cytoplasm.
  • IkBa also prevents the NF—kB translation factor from binding to the DNA needed for NF-kB to function.
  • IkBa protein has been found to have IkBa protein mutations in some Hawkins lymphoma cells. This mutation inactivates IkBa, resulting in chronic activation of NF-kB. Lymphoma tumors have been reported to be directly associated with the development of malignant tumors (Cabannes E et al, Oncogene 18 (20): 3063770).
  • SerURe / threonine-protein kinase 13 (AURKC) protein is an enzyme encoded by the AUPKC gene (Bernard M et al., Genomi cs 53 (3): 40679). This gene encodes the aurora subfami ly of serine and threonine protein kinase.
  • the encoded protein is a chromosomal transport protein that complexes with Aurora B and the internal centrosome protein and can play a role in tissue microtubules with respect to centrosome and spindle function during mitosis. This gene is overexpressed in some cancer cell lines, indicating that it is involved in tumor signal transduction.
  • the growth, differentiation, and death of cells can be attributed to protein-protein, protein-nucleic acid. It is made by the interaction between high molecular materials. Out-of-cell signals pass through receptors on the cell membrane and pass through the cell's nucleus through several biochemical reactions in the cytoplasm, where they express specific genes. This intracellular delivery of these external signals is achieved by interactions between proteins at different levels. For example, growth factors or cytokines bind to the corresponding receptors on the cell surface, which induces clustering of the receptors. Ligand aggregation by these receptors causes the intracellular domains of the receptors to aggregate together, leading to interactions with various proteins involved in intracellular signaling.
  • This signaling system creates intermediate proteins that can deliver multiple levels of signal through protein kinase phosphorylation, protein phosphatase dephosphorylation, and so on, and eventually the signal is transferred to transcriptional activators.
  • Activated transcriptional promoters bind to DNA and interact with basic transcriptional apparatus such as RNA polymerase that synthesizes mRNA to activate specific genes. Therefore, these interactions control specific transcription, specifically in response to specific tissues, developmental processes and external stimuli.
  • the cause of the disorder is that abnormal interactions such as alteration, inhibition, and promotion between specific proteins are caused by invasion of foreign substances or genetic variation of internally active proteins. Therefore, the research that has been continued because the substance that can control this interaction can provide a method of treatment for the disease caused by it.
  • yeast two hybrids Y2H
  • fluorescence resonance energy transfer FRET
  • fluorescence resonance energy transfer FRET
  • recombination of split proteins biniolecular fluorescence com
  • Bi-FC recombination of split proteins
  • the inventors of the present invention have completed a cancer drug screening method using IkBa and AURKC based on the system for confirming the protein-protein interaction.
  • an object of the present invention is to provide a method for screening a cancer drug through IkBa and AURKC.
  • Another object of the present invention is to provide a compound screened through the cancer drug screening method.
  • Still another object of the present invention is to provide a composition for preventing and treating cancer, which comprises the screened compound as an active ingredient.
  • Another object of the present invention is to provide a use of the screened compound for the manufacture of a prophylactic and therapeutic agent for cancer.
  • Still another object of the present invention is to provide a method for preventing and treating cancer, wherein the screened compound is administered to an individual in need thereof in an effective amount.
  • the present invention provides a method for screening a cancer drug through IkBa and AURKC.
  • the present invention provides a compound screened through the cancer drug screening method.
  • the present invention provides a composition for preventing and treating cancer comprising the screened compound as an active ingredient.
  • (A) (i) expressing a fusion protein in which sequentially bound fusion proteins, a first marker, and IkBa, and a fusion protein in which a second marker, AURKC are sequentially coupled, Preparing a cell expressing a fusion protein to which a label, IkBa is sequentially coupled, mobile mothers, second markers, and AURKC are sequentially coupled to each other;
  • the present invention provides a method for screening a cancer drug, wherein the cancer drug candidate is identified as a cancer drug when it inhibits binding of IkBa protein and AURKC protein.
  • the present invention used intracellular localization of proteins generated by external stimulation or intrinsic signaling mechanisms.
  • a mobile parent whose position is moved by an external stimulus or an intrinsic signal transduction operation, a constituent in which the labeling material and the IkBa or AURKC which can be tracked are sequentially designed, and the labeling material and the AURKC or IkBa are A second construct was sequentially designed.
  • the first label and the second label should each contain one different IkBa and one AURKC.
  • the second component should be composed of the second marker, AURKC, and if the first component is the carrier, the first marker, and the AURKC, the second component is: It should consist of two markers, IkBa.
  • the present invention determines whether the first component and the second component bind to each other, and whether the first component and the second component interfere with the binding of the first component and the second component to the cancer therapeutic agent. If inhibition of the binding of the first component and the second component is determined as a cancer treatment.
  • Mobile hair in the present invention is a site that functions to move the crab 1 to a specific area in the cell.
  • movement to a specific region within a cell is caused by an external signal. It is possible to migrate haphazardly or intrinsically, and certain regions within the cell represent separate, discrete and identifiable elements present in the cell as intracellular structures.
  • the intracellular specific region may preferably be a membrane structure such as a cell membrane or a nuclear membrane or intracellular organelles such as endoplasmic reticulum, Golgi apparatus, mitochondria, lysosomes, and other intracellular specific regions.
  • the "mobile mothers" of the present invention is a protein kinase C (protein kinase C,
  • cPKC classic PKC; PKC-alpha, P C-beta, PKC— ga ⁇ a), nPKC (novel PKC; PKC-delta, PKC-epsilon, PKC-eta, PKOtheta) It is done.
  • the carriers of the present invention may use a variant of PKC, and the variant is more preferably a variant in which the intrinsic phosphorylation activity of PKC is eliminated in order to minimize disturbance caused by the intrinsic signaling mechanism.
  • the "first label” of the present invention is GFP (Green Fluorescent Protein)
  • Enhanced Green Fluorescent Protein EGFP
  • Red Fluorescent Protein RFP
  • Monomeric Red Fluorescent Protein mRFP
  • DsRed Red fluorescent protein
  • Cyan Fluorescent Protein CFP
  • Cyan Green Fluorescent Protein CGFP
  • YFP Yel low Fluorescent Protein
  • AzG AzG
  • HcR HcRed, Heteractis cr ispa red fluorescent protein
  • BFP Blue Fluorescent Protein
  • the "second label material” of the present invention is different from the one label material of the crab and GFP (Green
  • EGFP Fluorescent Protein
  • EGFP Enhanced Green Fluorescent Protein
  • RFP Red Fluorescent Protein
  • mRFP Monomeric Red Fluorescent Protein
  • DsRed Red fluorescent protein
  • CFP Cyan Fluorescent Protein
  • CFP Cyan Green Fluorescent Protein
  • YFP Yel low Fluorescent Protein
  • AzGCAzami Green HFP
  • HcRCHcRed Heteractis cr ispa red fluorescent protein
  • BFP Blue Fluorescent Protein
  • fusion protein refers to a protein or polypeptide having an amino acid sequence derived from two or more proteins. It may also comprise a linkage region of amino acids between amino acid moieties derived from.
  • the cell may be a cell of an animal, a plant, a yeast and a bacterium. Preferably, the cell may easily receive a first construct and a second construct, which are introduced from outside of the bacterium, and may be used for intracellular organelles such as cytoplasm and nucleus. Cells with distinct boundaries are preferred.
  • CHO-kl ATCC CCL-61, Cricetulus griseus, hamster, Chinese
  • HEK293 ATCCCRL-1573, Homo sapiens, human
  • HeLa ATCC CCL-2, Homo sapiens, human
  • SH-SY5Y ATCC CRL-2266, Homo sapiens, human
  • Swiss 3T3 ATCC CCL-92, Mus musculus, mouse
  • NIH / 3T3CATCC CRL-1658 Mus musculus, mouse
  • L-929 ATCC CCL-1, Mus musculus, mouse
  • Rat2 ATCCCRL-1764, Rat t us norvegicus, rat
  • RBL-2H3 ATCC CRL-2256, Rat t us norvegicus, rat
  • MDCK ATCC CCL-34, Can is familiar is).
  • the cells comprising the crab first construct and the second construct may be prepared by molecular biological methods known in the art.
  • the cells are transformed into individual expression vectors capable of expressing the first component and the second component or expression vectors capable of expressing both the first component and the second component, and then the system is expressed in the expression vector. Preferred is a method by which the first and second constructs are expressed.
  • each expression vector into cells can be carried out using transformation methods known in the art, such as calcium phosphate, calcium chloride, rubidium chloride, microprojectile bombardment, electroporation, Particle gun bombardment, silicon carbide whiskers, sonication, PEG-mediated fusion, microinjection, liposome mediation (1 iposome-mediated method), magnetic nanoparticle mediated method, and the like.
  • transformation methods known in the art such as calcium phosphate, calcium chloride, rubidium chloride, microprojectile bombardment, electroporation, Particle gun bombardment, silicon carbide whiskers, sonication, PEG-mediated fusion, microinjection, liposome mediation (1 iposome-mediated method), magnetic nanoparticle mediated method, and the like.
  • standard recombinant DNA and molecular cloning techniques used in the present invention are well known in the art and described in the following literature (Sambrook, J., Fritsch, EF and Maniatis, T., Molecular Cloning: A Laboratory).
  • reaction refers to the proximal state between a particular ligand or compound, or a portion or fragment thereof, and a portion of the second molecule of interest. It may be non-covalent or covalent as a result of hydrogen-bonding, van der Waals interactions, electrostatic or hydrophobic interactions. 12-myri state 13-acetate,
  • Phorbol ester TPA (12-ot et r adecanoy lphor bo 1 -13-acetate), PDBu (phorbol 12, 13-dibutyrate), Adenosine triphosphate (ATP), tr idecanoic ac id, arachidonic acid, linoleic acid, DiC8 It is characterized in that selected from the group consisting of 130C937.
  • the "treatment of signal substance" of the present invention is characterized by treating PM Phorbol 12-myri state 13-acetate, Phorbol ester) at a concentration of 50nM to 5 ⁇ . More preferably luM.
  • concentration of the PMA is less than 50 nM, movement of the mobile mothers using the PKC is not difficult, and when the concentration of the PMA is greater than 5 uM, abnormal phenomena (eg, cell death signal disturbances, etc.) occur due to overtreatment of the chemical.
  • the "confirming the interaction” is to detect the distribution of the first label and the second label, for example, coverslips containing the cells to be identified.
  • It may be a method of fixing the coverslip to a perfusion chamber and mounting it on a material stage of a confocal laser fluorescence microscope (Cal Zeiss LSM710) and obtaining images of the component vectors before and after external stimulation.
  • the present invention provides a compound screened through the cancer drug screening method.
  • the compound of the present invention may be represented by the following formula (1). [Formula 1]
  • the compound of the present invention is characterized by inhibiting IkBa protein and AURKC protein binding.
  • the compound of the present invention is characterized by inhibiting the binding of the IkBa protein and AURKC protein to show cancer prevention and treatment effects.
  • the "cancer” of the present invention is gastric cancer, lung cancer, liver cancer, colon cancer, small intestine cancer, pancreatic cancer, brain cancer, bone cancer, melanoma, breast cancer, sclerosis, uterine cancer, cervical cancer head and neck cancer, esophageal cancer, thyroid cancer, parathyroid cancer, kidney Cancer, sarcoma, prostate cancer, urethral cancer, bladder cancer, hematologic cancer, lymphoma, psoriasis or fibroadenoma.
  • the present invention provides a composition for preventing and treating cancer, comprising an inhibitor of interaction between IkBa and AURKC as an active ingredient.
  • the inhibitor may be represented by the following formula (1).
  • composition of the present invention may further comprise suitable carriers, excipients and diluents commonly used in the manufacture of pharmaceutical compositions. It may also be used in the form of powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols and the like, oral formulations, external preparations, suppositories, and sterile injectable solutions according to conventional methods. Suitable formulations known in the art are preferably those disclosed in Remington's Pharmaceut i Cal Science, recently, Mack Publ i Shing Company, Easton PA.
  • Carrier excipients and diluents that may be included include lactose, dextrose, sucrose, sorbitol, manny, xylly, erythritol, maltitol, starch, acacia rubber, alginine. , Gelatin, Calcium Phosphate Shampoo, Cellulose, Methyl Cellulose, Microcrystalline Salose Polyvinyl Pyridone, Water, Methylhydroxy Benzoate, Propylhydroxyxbenzoate, Talc, Magnesium Stearate and Mineral oil and the like.
  • composition When formulating the composition, it is prepared using diluents or excipients such as layering agents, extenders, binders, wetting agents, disintegrating agents, and surfactants which are commonly used.
  • Solid preparations for oral administration include tablets, pills, powders, granules, capsules, and the like. Such solid preparations may include at least one excipient such as starch, calcium carbonate, sucrose, lactose, It is prepared by mixing gelatin.
  • lubricants such as magnesium stearate and talc are also used.
  • Oral liquid preparations include suspensions, solvents, emulsions, and syrups.In addition to the commonly used simple diluents, water and liquid paraffin, various excipients such as wetting agents, sweeteners, fragrances, and preservatives are included. Can be.
  • Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilized preparations, suppositories.
  • non-aqueous solvent and suspending agent vegetable oils such as propylene glycol, polyethylene glycol olive oil, injectable esters such as ethyl oleate, and the like can be used.
  • witemsol macrogol, twenty-one, cacao butter, laurin butter, glycerogelatin and the like can be used.
  • treatment refers to amelioration of a particular disease or disorder symptom, which may include treating such disorder, substantially preventing the development of the disorder, or improving the condition of the subject.
  • treatment refers to the full spectrum of treatment for a given disorder that afflicts a patient, which alleviates one or most of the symptoms resulting from that disorder, Treating certain disorders or preventing the development of a disorder.
  • the pharmaceutical composition of the present invention can be administered in parallel with a known compound having an effect of preventing and treating cancer or a known compound having an effect of inhibiting cancer metastasis.
  • the present invention provides a use of the inhibitor of the interaction between IkBa and AURKC for the manufacture of a prophylactic and therapeutic agent for cancer.
  • the present invention provides a method for preventing and treating cancer, comprising administering an effective inhibitor of the interaction between IkBa and AURKC to a subject in need thereof.
  • the inhibitor may be represented by the following formula (1).
  • H Inhibitors of interaction between IkBa and AURKC of the present invention can be administered in an effective amount via several routes including oral, transdermal, subcutaneous, intravenous or intramuscular.
  • the term 'effective amount' refers to an amount that exhibits a therapeutic and prophylactic effect or a cancer metastasis inhibiting effect when administered to a patient
  • the term 'subject' includes an animal, preferably a mammal, particularly a human. It may be an animal, cells, tissues, organs and the like derived from the animal. The subject may be a patient (pat i ent) in need of treatment.
  • the interaction inhibitor between IkBa and AURKC of the present invention may be administered by itself or prepared and prepared in various formulations as described above, and is preferably administered until a desired effect, i.e., cancer prevention and treatment effect, is obtained.
  • the compounds of the present invention and their pharmaceutically acceptable salts can be administered by various routes according to methods known in the art. That is, orally or parenterally, such as oral, intramuscular, intravenous, intradermal, intraarterial, intramedullary, intradural, intramural, intranasal, intravaginal, intra rectal sublingual or subcutaneous, gastrointestinal tract, mucosa or respiratory tract. May be administered. Preferably it can be applied directly to the skin.
  • the compounds of the present invention and pharmaceutically acceptable salts thereof may be administered in a form that is bound to or encapsulated within a molecule that induces high affinity binding to the target cell or tissue.
  • Inhibitors of interactions between IkBa and AURKC of the present invention may be prepared using techniques known in the art, such as sterols (e.g. cholesterol), lipids (e.g. cationic lipids, bivalent or liposomes) or target cell specific binding agents (e.g. Ligands recognized by target cell specific receptors).
  • Suitable coupling or crosslinking agents can include, for example, Protein A, carbodiimide, N-succinimidyl-3- (2-pyridyldithio) propiotate (SPDP) and the like. These formulations are described in Remington's Pharmaceut i cal Science, 15th Edition, 1975, Mack Publ i Shing Company, East on, Pennsyl vani a).
  • the present invention provides a method for screening a cancer drug through the interaction of IkBa and AURKC. More specifically, the present invention provides a binding inhibitor compound of IkBa and AURKC and a composition for preventing and treating cancer comprising the same as an active ingredient.
  • the screening method of the present invention can be usefully used in that the cancer therapeutic agent can be easily screened depending on the inhibition of binding of IkBa and AURKC.
  • FIG. 1 is an overexpression of TMD-mRFP-IkBa (first construct) and EGFP-AURKC (second construct) in HEK293T cell line and analyzed by a method using cell imaging (Registration No. 10-0948767) (left) Panel; first component, middle panel; second component, right panel; first component + second component).
  • Figure 2 shows pTMD-mRFP-C3 p i asm i d (Crab 1 construct) and EGFP-AURKC (near 12 constructs)
  • TMD-mRFP-IkBa first construct
  • EGFP-AURKC second construct
  • 5A to 5D show gastric cancer cell lines AGS (A) and SNU638 b) and colorectal cancer cell lines.
  • FIG. 6 shows that TMD-mRFP-IkBa (first construct) and EGFP-AURKC (second construct) are overexpressed in HEK293T cells (Panel A) and CH0-K1 cell lines (Panel B) in the same manner and Compound 50 ⁇ M was treated for 3 hours. After that, 1 ⁇ ⁇ (external stimulus) was treated for 3 minutes to analyze the inhibition of binding of the two proteins in real time (left panel: 1st component, middle panel: 2nd component ⁇ right panel: 1st component + 1st) 2 components).
  • FIG. 7 shows a wound healing assay (A), a Transwell migration assay (B) and a soft agar assay by culturing breast cancer cell line MDA-MB-231 cells.
  • A wound healing assay
  • B Transwell migration assay
  • B soft agar assay by culturing breast cancer cell line MDA-MB-231 cells.
  • c soft agar assay by culturing breast cancer cell line MDA-MB-231 cells.
  • HEK293 ATCC CRL-1573, Homo sapiens, human
  • MDA-MB-231 ATCC HTB-26, Homo sapiens, human
  • A549 ATCC CC ⁇ 185 Homo sapiens, human
  • HepG2 ATCC HB -8065, HepG2, Homo sapiens, human
  • Culture conditions of animal cell lines used in the present invention was used for cell line culture method of ATCC (American Type Culture Collection), which is the organ of each cell line.
  • CH0-K1 was used for F-12 culture
  • HEK293 and MDA-MB-231 cell lines were used for DMEM culture
  • A549 and HepG2 cells were used for RPMI 1640, and other culture conditions were used in the same manner.
  • the common culture method of each cell is as follows (detailed culture conditions may vary depending on the purpose of the skilled person). Incubation at pH 7.4 with 25 niM HEPES, 10% FBS (fetal bovine serum, v / v), 100 units / ml penicillin, 100 ug / ml straptomycin In cell (F-12 and DMEM), each cell line was incubated in an incubator maintained at a temperature of 37 ° C., CO 2 partial pressure of 5%.
  • Intracellular gene introduction method used in the embodiment of the present invention used Fugene HD (Promega), one of the liposome-based methods commonly used, all conditions for gene introduction, such as the concentration of the gene is a manufacturer
  • Fugene HD Promega
  • the coverslips were placed in 6-plates, and the cells were cultured for one day, and then exchanged with 2 ml of fresh culture without penicillin, streptomycin, or FBS.
  • About one transgenic white sample was added to the culture medium without 0.1 ml of penicillin and streptomycin FBS, followed by complete mixing and addition of 3 Fugene HD reagents.
  • the solution was allowed to stand at room temperature for 15 minutes, then added to each well of a 6-plate containing the coverslips in which the cells were grown, incubated for 4 hours, and 200 ⁇ l of FBS was added for transformation for 18 hours. It was.
  • the first constituent includes a module (mobile mothers) that can move to the cell membrane when the protein evenly expressed in the cytoplasm has been treated with the signal amorphous substance, and can be analyzed using these microscopes.
  • the protein is labeled and bound, and finally consists of a fusion sequence to which the target substance can be bound.
  • the first construct was constructed using restriction enzymes EcoRI / Kpnl (IkBa) and EcoRI / Xinal (AURKC) with IkBa and AURKC in a TMD-mRFP-empty vector.
  • TMD—mRFP-empty vector is a vector comprising a Protein Kinase C mutant (TMD) represented by SEQ ID NO: 5 and mRFP represented by SEQ ID NO: 7.
  • IkBa is the template for pcDNA3.1-IkBa vector (NCBI Reference Sequence:
  • NM_020529.2 purchased from Mediscov Inc
  • PCR was performed using primers of SEQ ID NO: 1 and SEQ ID NO: 2.
  • AURKC performed PCR using a PPTB7-AURKC vector (GenBank: BC002363) as a template and primers of SEQ ID NO: 3 and SEQ ID NO: 4. Obtained after PCR amplification using primers Water was prepared by inserting into the EcoRI / Kpnl (IkBa) and EcoRI / Xmal (AURKC) positions of the pTMD-mRFP-C3 vector.
  • the second construct includes a label for analyzing the movement of a target substance having intrinsic properties of binding to a target substance bound to the first component.
  • Crab bicomponent uses green fluorescent protein (EGFP), a fluorescence that is different from the label contained in the first construct.
  • the second construct was constructed using restriction enzymes EcoRI / Kpnl (IkBa) and EcoRI / Xmal (AURKC) from IkBa and AURKC in an EFRP-C3 vector (clontech) of a previously registered patent (e.g. 10-1217718). .
  • IkBa is the template for the pcDNA3.1—IkBa vector (NCBI Reference Sequence:
  • AURKC was performed using a PPTB7-AUR C vector (GenBank: BC002363) as a template and using primers of SEQ ID NO: 3 and SEQ ID NO: 4. After PCR amplification using the primers, the obtained product was prepared by inserting into the EcoRI / Kpnl (IkBa) and EcoRI / Xnial (AURKC) positions of the pTMD-mRFP-C3 vector.
  • the confocal laser microscope uses a 488 nni Argon laser (EGFP) and a 543 nm HeNe laser (niRFP) to excite the fluorescent label, and the fluorescent signal generated from each fluorescent label is a band path filter BP505.
  • EGFP 488 nni Argon laser
  • niRFP 543 nm HeNe laser
  • the fluorescent signal generated from each fluorescent label is a band path filter BP505.
  • -530 EGFP
  • Long path filter LP560 or BP560-630 niRFP
  • the red fluorescence caused by the first component vector including the moving mothers (TMD) after the external stimulation using the confocal laser fluorescence microscope is transferred to the cell membrane.
  • the green fluorescence by the second material vector for the target material without the mobile hairs is distributed evenly in the cytoplasm as in stimulation.
  • a 12-element vector is an external stimulus
  • the migration of the second constituent to the cell membrane is necessarily based on the binding of the target substance to the target substance.
  • IkBa proteins regulated by TNF-a pha are known to be involved in various cellular signaling mechanisms in cells. Of particular interest among the various mechanisms is that they have a close relationship with provoking inflammation in response to various risk signals occurring in vivo.
  • the binding of AURKC protein interacting with IkBa was performed in real time using living cells.
  • the first and second constructs prepared above were transformed into HEK293T and CH0-K1 cell lines, and their fluorescence was confirmed at 0 and 3 minutes.
  • the first construct (TMD-mRFP-IkBa) was evenly distributed in the cell at 0 minutes, and then moved to the cell membrane after 3 minutes (left panel in Fig. 1).
  • EGFP-AURKC was also distributed evenly in the cell at 0 min and then moved to the cell membrane with the first construct after 3 min (panel in FIG. 1). This means that the first component and the second component are combined.
  • the first component is replaced by TMD-mFRP-AU KC and the second component is replaced by EGRP—IkBa.
  • HEK2953T and CH0-K1 cell lines were transformed and fluorescence was detected at 0 and 3 minutes.
  • the first construct (TMD-mRFP-AURKC) was evenly distributed in the cells at 0 minutes, and then moved to the cell membrane after 3 minutes (Fig. 3 is the left panel).
  • the two constructs (EGFP-IkBa) were also distributed evenly within the cell at 0 min and then migrated to the cell membrane with the first construct after 3 min ( Figure 3-middle panel). This means that the first component and the second component are combined.
  • the first and second compositions prepared above are prepared.
  • the cells were transformed into coverslips in which HEK293T and CH0-K1 cells grew. Afterwards, IkBa protein and AURKC protein binding inhibitor candidates were Treated during.
  • the coverslip is secured to the perfusion chamber, mounted on the stage of the confocal laser fluorescence microscope (Cal Zeiss LSM710), for component vectors before and after external stimulation ( ⁇ Phorbol ester). Image was acquired.
  • Figure 6 is an analysis of the binding inhibitory effect of the IkBa protein and AURKC protein of the compound TMD-mRFP-IkBa (first component) and EGFP-AURKC (component 2) in the same manner in HEK293T cells and CH0-K1 cell line Overexpressed and treated 50 ⁇ M of the compound for 3 hours. Thereafter, 1 ⁇ of ⁇ (external stimulus) was treated for 3 minutes to analyze the inhibition of binding of both proteins in real time. It was confirmed that the above compound did not bind the IkBa protein and the AURKC protein, and the red fluorescence reflecting the moving mothers and IkBa moved to the cell membrane, but the green fluorescence reflecting the AURKC protein was located in the cytoplasm. Therefore, it can be seen that the compound represented by Chemical Formula 1 can be used as an inhibitor that inhibits the binding of IkBa and AURKC.
  • Cytotoxicity of the compounds used in the examples of the invention was used a commonly used CCK8 kit (Dojindo Molecular Technologies) and all conditions were performed according to the manufacturer's instructions. More specifically, cells were cultured in 96-plate for one day, 10, 50, and 100 ⁇ were treated with the compound and incubated for 24 and 48 hours, followed by 10 ⁇ l of CCK8 solution, and the absorbance was measured at 450 nm for one hour.
  • gastric cancer cell lines AGS and SNU638 (FIG. 5, B) and colon cancer cell lines HCT116 and SW620 (C), uterine cancer cell line Hela (MA) ⁇ breast cancer cell line MDA—MB-231 (F), as shown in FIG. Cytotoxicity was measured in lung cancer cell line A549 and HepG2 cell line, and these cancer cells were inhibited by compounds that inhibit the interaction of IkBa and AURKC. Death increased.
  • the cancer cell migration assay was performed using a breast cancer cell line MDA—MB-231 cultured in DMEM containing 10% FBS at 37 ° C. 5% CO 2 conditions (FIG. 7A). Cells are incubated for 24 hours in 24 well tissue culture plates (2 ⁇ 10 5 cells). Thereafter, the wound is slowly cut across the center of the well using a lnil pipette tip without changing medium. At this time, use the tip upright with the bottom of the well, cut the wound in one direction, and march twice with medium to remove the fallen cells, and then supplement the new medium. In addition, 25 ⁇ M of compounds inhibiting the interaction of the plate group IkBa with AURKC were treated and incubated for 48 hours. This is followed by two marches with IX PBS followed by fixation with 3.7% paraformaldehyde for 30 minutes. The fixed cells were stained for 30 minutes by dissolving 1% crystal vial in 2% ethane and observed using a microscope.
  • the control group ( ⁇ ) without treatment of the compound that inhibits the interaction of IkBa and AURKC in the breast cancer cell line was narrower than that of the treatment with the compound (25 ⁇ ). Confirmed.
  • the cells grew on the wounded portion with the tip to fill the space, but when the compound was treated, the cells did not grow on the wounded portion and the space remained. That is, it was confirmed that the compound which inhibits the interaction of IkBa and AURKC inhibits the proliferation of cells.
  • the transwell migration assay used a breast cancer cell line MDA-MB-231 incubated with 1 FBS of DMEM (FIG. 7B).
  • FBS 1 FBS of DMEM
  • To prepare the transwell 0.5% FBS and 2.6 ⁇ 1 DMEM were placed in the lower part of the 24 well, and 8 well sized transwell was added. Then, cells (1 ⁇ 10 5 cells) were added to the prepared transwells. Then, ⁇ and 25 ⁇ were added to the compounds that inhibit the interaction between IkBa and AURKC, and the cells were incubated for 2 hours and 30 minutes at 37 ° C, 5% C0 2 , to move down the filter fitted with the cells. It was.
  • cancer cell proliferation was inhibited by a compound that inhibits the interaction between IkB a and AURKC as in ⁇ 7-1>.
  • the colony morphology was analyzed by culturing the breast cancer cell line MDA-MB—231 in agar medium prepared using agarose by a Sot agar assay (FIG. 7). First, 4% agar was dissolved and kept warm in a 56 ° C constant temperature bath, and 10% FBS DMEM was kept at 37 ° C. In order to make a lower layer of agar medium to culture the cells, DMEM 5 [ii l containing 0.75% agar and 10% FBS was placed in a 60 mm culture dish and waited until it solidified. 10% including 0.36% agar to make the next upstairs
  • the present invention relates to a method for screening a cancer therapeutic agent through the interaction of IkBa and AURKC. More specifically, IkBa and AURKC binding inhibitor compound and the active ingredient It relates to a composition for preventing and treating cancer.
  • the present invention is effective in that cancer therapeutic agents can be screened at the molecular level and the action of the therapeutic agents can be known through intermolecular interactions.

Abstract

The present invention relates to a method for screening a therapeutic agent for cancer using interaction between IkBa and AURKC and, more specifically, to an IkBa and AURKC binding inhibitor compound, and a composition for the prevention and treatment of cancer comprising the same as an active ingredient. The present invention is effective in that a therapeutic agent for cancer can be screened at a molecular level and the action of the therapeutic agent can be identified through molecular interaction.

Description

【명세서】  【Specification】
【발명의 명칭】  [Name of invention]
IkBa 및 AURKC의 상호작용을 통한 암 치료제 스크리닝 방법 【기술분야】  Cancer drug screening method through interaction of IkBa and AURKC
<ι> 본 출원은 2014년 7월 24일에 출원된 대한민국 특허출원 제 10-2014—0094260 호를 우선권으로 주장하고, 상기 명세서 전체는 본 출원의 참고문헌이다,  <ι> This application claims the priority of Korean Patent Application No. 10-2014—0094260, filed July 24, 2014, the entirety of which is a reference of the present application,
<2>  <2>
<3> 본 발명은 IkBa 및 AURKC의 상호작용을 통한 암 치료제 스크리닝 방법에 관 한 것이다ᅳ 더욱 상세하게는 ikBa와 AURKC의 결합 억제제 화합물 및 이를 유효성분 으로 포함하는 암 예방 및 치료용 조성물에 관한 것이다.  The present invention relates to a method for screening cancer therapeutic agents through the interaction of IkBa and AURKC. More particularly, the present invention relates to a binding inhibitor compound of ikBa and AURKC and a composition for preventing and treating cancer comprising the same as an active ingredient. .
<4>  <4>
【배경기술】  Background Art
<5> IkBa(nuc lear factor of kappa l ight polypept ide gene enhancer in Bᅳ eel I s inhibi tor , alpha) 단백질은 NF— κ Β의 활성을 저해하는 제어소단위의 한 종류이다. IkBa는 NF-kB의 핵위치신호 (NLS) 부위에 결합하여 핵위치신호를 가림으로써 NF-kB 가 세포질 내에서 불활성화 상태로 유지되도록 한다. 또한 IkBa는 NF— kB의 번역 인 자가 NF-kB이 기능하기 위해 필요한 DNA에 결합하는 것을 막는다. IkBa 단백질은 몇몇 호킨스 림프종 세포에서 IkBa 단백질 돌연변이가 발견되었다. 이 돌연변이는 IkBa를 불활성화 하여 NF-kB가 만성적으로 활성화되도록 한다. 하여 림프종 종양이 악성 종양으로의 발병과 직접적 연관이 있는 것으로 보고되었다 (Cabannes E et al , Oncogene 18 (20): 3063770) .  <5> IkBa (nuc lear factor of kappa light polypeptide gene enhancer in B eel I s inhibitor, alpha) protein is a type of control subunit that inhibits NF- κ Β activity. IkBa binds to the nuclear position signal (NLS) site of NF-kB to mask the nuclear position signal so that NF-kB remains inactivated in the cytoplasm. IkBa also prevents the NF—kB translation factor from binding to the DNA needed for NF-kB to function. IkBa protein has been found to have IkBa protein mutations in some Hawkins lymphoma cells. This mutation inactivates IkBa, resulting in chronic activation of NF-kB. Lymphoma tumors have been reported to be directly associated with the development of malignant tumors (Cabannes E et al, Oncogene 18 (20): 3063770).
<6>  <6>
<7> AURKC(Ser ine/threonine-protein kinase 13) 단백질은 AUPKC 유전자에 의해 코딩되는 효소이다 (Bernard M et al . , Genomi cs 53 (3): 40679) . 이 유전자는 세린 및 트레오닌 단백질 카이네이즈의 오로라 하위군 (aurora subfami ly)을 인코딩한다. 인코딩 된 단백질은 오로라 B 및 내부 중심 소체 단백질과 복합체를 형성하고 유사 분열동안 중심체 및 스핀들 (spindle) 기능과 관련하여 조직 미세 소관에 역할을 할 수 있는 염색체 운송 단백질이다. 이 유전자는 몇몇 암 세포주에서 과발현되는데, 이는 종양의 신호 전달에 관여한다는 것을 나타낸다.  SerURe / threonine-protein kinase 13 (AURKC) protein is an enzyme encoded by the AUPKC gene (Bernard M et al., Genomi cs 53 (3): 40679). This gene encodes the aurora subfami ly of serine and threonine protein kinase. The encoded protein is a chromosomal transport protein that complexes with Aurora B and the internal centrosome protein and can play a role in tissue microtubules with respect to centrosome and spindle function during mitosis. This gene is overexpressed in some cancer cell lines, indicating that it is involved in tumor signal transduction.
<8>  <8>
<9> 한편, 세포들의 성장, 분화, 이동 죽음 등은 단백질-단백질, 단백질 -핵산 등의 고분자 물질들간의 상호작용 (interaction)에 의해 이루어진다. 세포 외부의 신호는 세포막의 수용체를 통과하여 세포질내에서 여러 생화학적 반웅을 통해 세포 의 핵 내로 전달되며, 거기서 특정 유전자들을 발현시킨다. 이러한 외부 신호의 세 포내 전달과정은 여러 단계의 단백질간 상호작용에 의해 이루어진다. 예로써 성장 조절인자 (growth factor)나 사이토카인 (cytokine) 등은 세포 표면의 해당 수용체 (receptor)에 결합하고 이러한 결합은 수용체가 서로 뭉치도록 (cluster ing) 유도한 다. 이러한 수용체의 리간드에 의한 뭉침은 그 수용체의 세포질내 도메인끼리도 서 로 뭉치도록 유도함으로써 세포내 신호전달과정에 관계된 여러 단백질들과의 상호 작용을 유발한다. 이 신호전달체계는 단백질 키나제에 의한 단백질의 인산화, 단백 질 포스파타제에 의한 탈인산화등을 통해 여러 단계의 신호를 전달 할 수 있는 중 간체 단백질을 만들면서 결국 그의 신호를 전사촉진 단백질 (transcriptional activator)에 전달한다 (Helden, C. H. , Cell 80, 213-223(1995)). 활성화된 전사촉 진 단백질은 DNA에 결합하고, mRNA 를 합성하는 RNA 중합효소 등 기본적인 전사 조 절 단백질들 (basal transcriptional apparatus)과 상호작용하여 특정 유전자들을 활성화시킨다. 따라서 이러한 상호작용은 특정 조직, 발생학적 과정 및 외부자극 등에 반웅하여 특이적으로 전사가 일어나도록 조절하는 것이다. 이때 외부물질의 침입 또는 내부 활성 단백질의 유전적 변이 등에 의해 특정 단백질간의 상호작용이 변경 , 억제 , 촉진 등 비정상적으로 일어나게 된 것이 병 (disorder)의 원인이라고 할 수 있다. 따라서 이러한 상호작용을 조절할 수 있는 물질은 곧 그에 기인된 병 에 대한 치료의 방법을 제공할 수 있으므로 이에 관한 연구가 계속되어 왔다. On the other hand, the growth, differentiation, and death of cells can be attributed to protein-protein, protein-nucleic acid. It is made by the interaction between high molecular materials. Out-of-cell signals pass through receptors on the cell membrane and pass through the cell's nucleus through several biochemical reactions in the cytoplasm, where they express specific genes. This intracellular delivery of these external signals is achieved by interactions between proteins at different levels. For example, growth factors or cytokines bind to the corresponding receptors on the cell surface, which induces clustering of the receptors. Ligand aggregation by these receptors causes the intracellular domains of the receptors to aggregate together, leading to interactions with various proteins involved in intracellular signaling. This signaling system creates intermediate proteins that can deliver multiple levels of signal through protein kinase phosphorylation, protein phosphatase dephosphorylation, and so on, and eventually the signal is transferred to transcriptional activators. (Helden, CH, Cell 80, 213-223 (1995)). Activated transcriptional promoters bind to DNA and interact with basic transcriptional apparatus such as RNA polymerase that synthesizes mRNA to activate specific genes. Therefore, these interactions control specific transcription, specifically in response to specific tissues, developmental processes and external stimuli. At this time, it can be said that the cause of the disorder is that abnormal interactions such as alteration, inhibition, and promotion between specific proteins are caused by invasion of foreign substances or genetic variation of internally active proteins. Therefore, the research that has been continued because the substance that can control this interaction can provide a method of treatment for the disease caused by it.
<ιο> 현재까지 알려진 생체고분자 물질들 간의 상호작용, 특히 결합특성을 분석하 기 위한 방법으로 교차결합 (crossl inking), 친화크로마토그래피 (affinity chromatography) , 면역침강법 ( i mmunoprec i p i t at i on , IP) 등의 전통적인 in vitro 방법들이 사용되고 있으나 이들 방법들은 단백질의 생산과 분리 /정제 과정이 요구 되며 시험관내의 완층액의 조건과 추출된 단백질의 2차적인 변형 등의 문제점이 있 어 생체내에서 일어나는 실제 상호작용과 상이한 정보를 제공할 수 있다는 단점을 가지고 있다. <ιο> Cross-linking, affinity chromatography, immunoprecipitation (i mmunoprec ipit at i on, Traditional in vitro methods such as IP) are used, but these methods require the production and separation / purification of proteins, and in vivo, due to problems such as in vitro lye solution and secondary modification of extracted proteins. The disadvantage is that it can provide different information than the actual interaction that takes place.
<ιι> 이러한 in vitro 방법들의 단점을 보완하기 위해 세포내에서 직접 수행하는 이스트 투 하이브리드 (yeast twohybrid, Y2H), 형광공명에너지전이 (fluorescence resonance energy transfer , FRET) , 분할 단백질의 재결합 (biniolecular fluorescence com lementation, Bi-FC) 방법들이 개발되어 사용되고 있다.  <ιι> To complement the shortcomings of these in vitro methods, yeast two hybrids (Y2H), fluorescence resonance energy transfer (FRET), and recombination of split proteins (biniolecular fluorescence com) lementation (Bi-FC) methods have been developed and used.
<12> <13> 본 연구진은 단백질 인산화효소 C(prote i n k i nase C) 이동모들, 제 1표지물질 및 목적물질아 포함된 제 1구성물과 제 2표지물질 및 표지물질이 포함된 제 2구성물을 세포내에서 발현시켜 그 발현된 목적물질과 표적물질의 상호작용 여부를 확인할 수 있는 시스템을 개발하여 특허 등톡을 받은 바 있다 (등톡번호 : 10-0948767) . 이 시 스템은 단백질 간의 상호작용을 확인할 수 있다는 점에서 유용한 실험 방법을 제시 하였다. 하지만 본 시스템을 통해 치료제 후보물질을 스크리닝한 선행 연구는 아직 알려진 바 없다. <12> <13> The researchers performed intracellular intracellular preparations of protein kinase C carriers, first and second markers and markers. The company developed a system that can express whether the expressed target substance and the target substance interact with each other and received patent registration (equivalent number: 10-0948767). This system presents a useful experimental method in that it can confirm the interaction between proteins. However, no previous studies have screened for drug candidates through this system.
<14>  <14>
【발명의 상세한 설명】  [Detailed Description of the Invention]
【기술적 과제】  [Technical problem]
<15> 이에 본 발명자들 상기 단백질과 단백질의 상호작용을 확인하는 시스템을 토 대로 IkBa와 AURKC를 이용한 암 치료제 스크리닝 방법을 완성하였다.  Thus, the inventors of the present invention have completed a cancer drug screening method using IkBa and AURKC based on the system for confirming the protein-protein interaction.
< 16> 따라서, 본 발명의 목적은 IkBa와 AURKC를 통한 암치료제 스크리닝 방법을 제공하는 것이다. Accordingly, an object of the present invention is to provide a method for screening a cancer drug through IkBa and AURKC.
<17> 본 발명의 또 다른 목적은, 상기 암 치료제 스크리닝 방법을 통해 스크리닝 된 화합물을 제공하는 것이다.  Another object of the present invention is to provide a compound screened through the cancer drug screening method.
<18> 본 발명의 또 다른 목적은, 상기 스크리닝된 화합물을 유효성분으로 포함하 는 암 예방 및 치료용 조성물을 제공하는 것이다. Still another object of the present invention is to provide a composition for preventing and treating cancer, which comprises the screened compound as an active ingredient.
< 19> 본 발명의 또 다른 목적은, 상기 스크리닝된 화합물의 암 예방 및 치료제 제 조를 위한 용도를 제공한다.  Another object of the present invention is to provide a use of the screened compound for the manufacture of a prophylactic and therapeutic agent for cancer.
<20> 본 발명의 또 다른 목적은, 상기 스크링된 화합물을 이를 필요로 하는 개체 에 유효량으로 투여하는 것을 특징으로 하는 암 예방 및 치료 방법을 제공한다.Still another object of the present invention is to provide a method for preventing and treating cancer, wherein the screened compound is administered to an individual in need thereof in an effective amount.
<21> <21>
【기술적 해결방법】  Technical Solution
<22> 상기의 목적을 달성하기 위해, 본 발명은 IkBa와 AURKC를 통한 암치료제 스 크리닝 방법을 제공한다.  In order to achieve the above object, the present invention provides a method for screening a cancer drug through IkBa and AURKC.
<23> 본 발명의 또 다른 목적을 달성하기 위해, 본 발명은 상기 암 치료제 스크리 닝 방법을 통해 스크리닝된 화합물을 제공한다.  In order to achieve another object of the present invention, the present invention provides a compound screened through the cancer drug screening method.
<24> 본 발명의 또 다른 목적을 달성하기 위해, 본 발명은 상기 스크리닝된 화합 물을 유효성분으로 포함하는 암 예방 및 치료용 조성물을 제공한다.  In order to achieve another object of the present invention, the present invention provides a composition for preventing and treating cancer comprising the screened compound as an active ingredient.
<25>  <25>
<26> 이하 본 발명을 상세히 설명한다 . <27> Hereinafter, the present invention will be described in detail. <27>
<28> 본 발명은  <28> The present invention
<29> ( a ) ( i ) 이동모들, 제 1표지물질, IkBa가 순차적으로 결합된 융합 단백질 및 제 2표지물질, AURKC가 순차적으로 결합된 융합 단백질을 발현하거나 ( Π ) 이동모 들, 제 1표지물질, IkBa가 순차적으로 결합된 융합 단백질 및 이동모들, 제 2표지물 질, AURKC가 순차적으로 결합된 융합 단백질을 발현하는 세포를 제조하는 단계; (A) (i) expressing a fusion protein in which sequentially bound fusion proteins, a first marker, and IkBa, and a fusion protein in which a second marker, AURKC are sequentially coupled, Preparing a cell expressing a fusion protein to which a label, IkBa is sequentially coupled, mobile mothers, second markers, and AURKC are sequentially coupled to each other;
<30> (b) 암 치료제 후보 물질을 첨가하는 단계 ; (B) adding a cancer drug candidate;
<3i> (c) 암 치료제 후보 물질과 (a)단계의 융합 단백질 간 상호작용이 이루어지 도록 하는 단계 ;  (C) allowing the cancer drug candidate to interact with the fusion protein of step (a);
<32> (d ) 신호물질을 처리하는 단계 ; 및  (D) processing the signal material; And
<33> ( e ) 암 치료제 후보 물질과 세포 내 융합 단백질의 분포를 통해 상호작용을 확인하는 단계를 포함하며  (E) confirming the interaction through the distribution of the candidate drug for cancer therapy and the intracellular fusion protein,
<34> 암 치료제 후보물질이 IkBa 단백질과 AURKC 단백질의 결합을 억제할 경우 암 치료제로 판별하는 것을 특징으로 하는 암 치료제 스크리닝 방법을 제공한다.  The present invention provides a method for screening a cancer drug, wherein the cancer drug candidate is identified as a cancer drug when it inhibits binding of IkBa protein and AURKC protein.
<35>  <35>
<36> 본 발명은 IkBa와 AURKC간의 상호작용을 직접적으로, 그리고 실시간으로 분 석할 수 있도록 하기 위하여, 외부자극 또는 내재적인 신호전달기작에 의해 발생되 는 단백질의 세포내 위치이동을 이용하였다. 즉, 외부자극 또는 내재적인 신호전달 가작에 의해 위치가 이동되는 이동모들, 이를 추적할 수 있는 표지물질과 IkBa 또 는 AURKC가 순차적으로 결합된 게 1구성물을 디자인하고, 표지물질과 AURKC 또는 IkBa가 순차적으로 결합된 제 2구성물을 디자인하였다. 제 1표지물질과 제 2표지물질 은 각각 IkBa와 AURKC 서로 다른 하나씩을 포함해야 한다. 예를 들어, 제 1구성물이 이동모들, 제 1표지물질, IkBa라면 제 2구성물은 제 2표지물질, AURKC로 구성되어야 하며 제 1구성물이 이동모들, 게 1표지물질, AURKC라면 제 2구성물은 게 2표지물질, IkBa로 구성되어야 한다.  In order to analyze the interaction between IkBa and AURKC directly and in real time, the present invention used intracellular localization of proteins generated by external stimulation or intrinsic signaling mechanisms. In other words, a mobile parent whose position is moved by an external stimulus or an intrinsic signal transduction operation, a constituent in which the labeling material and the IkBa or AURKC which can be tracked are sequentially designed, and the labeling material and the AURKC or IkBa are A second construct was sequentially designed. The first label and the second label should each contain one different IkBa and one AURKC. For example, if the first component is a carrier, the first marker, and IkBa, the second component should be composed of the second marker, AURKC, and if the first component is the carrier, the first marker, and the AURKC, the second component is: It should consist of two markers, IkBa.
<37>  <37>
<38> 본 발명은 제 1구성물과 제 2구성물이 결합하며, 암 치료제 후보물질의 게 1구 성물과 제 2구성물의 결합을 방해하는지의 여부에 따라서 암 치료제인지 아닌지 구 분된다. 만약 제 1구성물과 제 2구성물의 결합을 억제할 경우 암 치료제로 판별한다. According to the present invention, whether the first component and the second component bind to each other, and whether the first component and the second component interfere with the binding of the first component and the second component to the cancer therapeutic agent. If inhibition of the binding of the first component and the second component is determined as a cancer treatment.
<39> <39>
<40> 본 발명에서 이동모들은 게 1구성물을 세포내의 특정 영역으로 이동시키는 기 능을 하는 부위이다. 상기에서 세포 내의 특정 영역으로의 이동은 외부 신호에 의 해 또는 내재적으로 이동할 수 있으며, 세포내 특정 영역은 세포내 구조물로서 세 포내에 존재하는 구분되고 (separate), 구별되며 (discreet), 증빙 가능한 (identifiable) 요소를 나타낸다. 상기 세포내 특정 영역은 바람직하게는 세포막, 핵막 등의 막 구조물 또는 소포체, 골지체, 미토콘드리아, 리소좀 등의 세포내 소 기관 및 그 외 세포내 특정 영역일 수 있다. Mobile hair in the present invention is a site that functions to move the crab 1 to a specific area in the cell. In this case, movement to a specific region within a cell is caused by an external signal. It is possible to migrate haphazardly or intrinsically, and certain regions within the cell represent separate, discrete and identifiable elements present in the cell as intracellular structures. The intracellular specific region may preferably be a membrane structure such as a cell membrane or a nuclear membrane or intracellular organelles such as endoplasmic reticulum, Golgi apparatus, mitochondria, lysosomes, and other intracellular specific regions.
<4i> 본 발명의 상기 "이동모들" 은 단백질 인산화 효소 C(protein kinase C,<4i> The "mobile mothers" of the present invention is a protein kinase C (protein kinase C,
P C), cPKC( classical PKC; PKC-alpha, P C-beta, PKC— ga隱 a), nPKC( novel PKC; PKC-delta, PKC-epsilon, PKC-eta, PKOtheta)로 이루어진 군에서 선택된 것임을 특징으로 한다. PC), cPKC (classic PKC; PKC-alpha, P C-beta, PKC— ga 隱 a), nPKC (novel PKC; PKC-delta, PKC-epsilon, PKC-eta, PKOtheta) It is done.
<42> 이들은 모두 C1 도메인이라는 부분을 공통적으로 가지고 있으며, C1 도메인 에 DAG(diacyl glycerol) 또는 TPA(phorbol ester, PMA)와 결합함으로써 세포막으 로의 위치 이동이 유도된다. 본 발명의 이동모들로 바람직하게는 PKC의 변이체를 사용할 수 있으며, 상기 변이체는 내재적인 신호전달기작으로 인한 교란현상을 최 소화하기 위하여 PKC의 내재적인 인산화 활성을 제거한 변이체인 것이 더욱 바람직 하다.  They all have a common part called the C1 domain, and their binding to the cell membrane is induced by binding to DAG (diacyl glycerol) or TPA (phorbol ester (PMA)) in the C1 domain. Preferably, the carriers of the present invention may use a variant of PKC, and the variant is more preferably a variant in which the intrinsic phosphorylation activity of PKC is eliminated in order to minimize disturbance caused by the intrinsic signaling mechanism.
<43> 본 발명의 상기 "제 1표지물질" 은 GFP(Green Fluorescent Protein), The "first label" of the present invention is GFP (Green Fluorescent Protein),
EGFP( Enhanced Green Fluorescent Protein) , RFP(Red Fluorescent Protein) , mRFP(Monomer ic Red Fluorescent Protein) , DsRed(Discosoma sp. red fluorescent protein) , CFP(Cyan Fluorescent Protein) , CGFP(Cyan Green Fluorescent Protein) , YFP(Yel low Fluorescent Protein) , AzG(Azami Green), HcR(HcRed, Heteractis cr ispa red fluorescent protein) 및 BFP(Blue Fluorescent Protein)로 이루어진 군에서 선택된 것임을 특징으로 한다. Enhanced Green Fluorescent Protein (EGFP), Red Fluorescent Protein (RFP), Monomeric Red Fluorescent Protein (mRFP), Discosoma sp. Red fluorescent protein (DsRed), Cyan Fluorescent Protein (CFP), Cyan Green Fluorescent Protein (CGFP), YFP (Yel low Fluorescent Protein), AzG (Azami Green), HcR (HcRed, Heteractis cr ispa red fluorescent protein) and BFP (Blue Fluorescent Protein).
<44> 본 발명의 상기 "제 2표지물질" 은 게 1표지물질과 서로 다르며 GFP(Green The "second label material" of the present invention is different from the one label material of the crab and GFP (Green
Fluorescent Protein) , EGFP( Enhanced Green Fluorescent Protein) , RFP(Red Fluorescent Protein) , mRFP(Monomer ic Red Fluorescent Protein) , DsRed(Discosoma sp. red fluorescent protein) , CFP(Cyan Fluorescent Protein) , CGFP(Cyan Green Fluorescent Protein) , YFP(Yel low Fluorescent Protein) , AzGCAzami Green) , HcRCHcRed, Heteractis cr ispa red fluorescent protein) 및 BFP(Blue Fluorescent Protein)로 이루어진 군에서 선택된 것임을 특징으로 한다.Fluorescent Protein (EGFP), Enhanced Green Fluorescent Protein (EGFP), Red Fluorescent Protein (RFP), Monomeric Red Fluorescent Protein (mRFP), Discosoma sp. Red fluorescent protein (DsRed), Cyan Fluorescent Protein (CFP), Cyan Green Fluorescent (CGFP) Protein), YFP (Yel low Fluorescent Protein), AzGCAzami Green (HFP), HcRCHcRed, Heteractis cr ispa red fluorescent protein) and BFP (Blue Fluorescent Protein).
<45> <45>
<46> 본 발명의 용어 "융합 단백질' '은 2가지 이상의 단백질로부터 유래된 아미노 산 서열을 갖는 단백질 또는 폴리펩티드를 지칭한다. 융합 단백질은 별개의 단백질 로부터 유래된 아미노산 부분들 간의 아미노산의 연결성 영역을 포함할 수도 있다. 상기 세포는 동물, 식물, 효모 및 박테리아의 세포일 수 있으며, 바람직하게 는 박테리아 외의 경우 외부로부터 도입되는 제 1구성물 및 제 2구성물을 잘 받아들 일 수 있고, 세포질과 핵 등의 세포내 소기관의 경계가 뚜렷하게 구별되는 세포가 바람직하다. 더 바람직하게는 CHO-kl(ATCC CCL-61, Cricetulus griseus, hamster, Chinese), HEK293(ATCCCRL-1573, Homo sapiens, human), HeLa(ATCC CCL-2 , Homo sapiens, human) , SH-SY5Y(ATCC CRL-2266 , Homo sapiens, human) , Swiss 3T3(ATCC CCL-92 , Mus musculus, mouse) , 3T3-L1CATCC CL-173, Mus musculus, mouse) , NIH/3T3CATCC CRL-1658, Mus musculus, mouse), L-929(ATCC CCL-1, Mus musculus, mouse), Rat2(ATCCCRL-1764, Rat t us norvegicus, rat), RBL-2H3(ATCC CRL-2256, Rat t us norvegicus, rat), MDCK(ATCC CCL-34 , Can is familiar is)일 수 있다. 또한 그 밖에 각종 줄기세포, 각종 조직에서 추출된 세포 및 인위적으로 만들어진 유사 세포막 구조물일 수 있다. 본 발명에서 게 1구성물 및 제 2구성물을 포함하는 세포는 당업계에서 공지된 분자생물학적 방법을 통해 제조될 수 있다. 이에 제한되지는 않으나, 제 1구성물 및 제 2구성물을 발현할 수 있는 각각의 발현백터 또는 제 1구성물 및 제 2구성물을 모두 발현할 수 있는 발현백터로 세포를 형질전환한 뒤, 발현백터에서 계 1구성물 및 제 2 구성물이 발현되도록 하는 방법이 바람직하다. 세포로의 각각의 발현백터의 형질전환은 당업계에 공지된 형질전환방법, 예 를 들면, 인산칼습법, 염화칼슘법, 염화루비듐법, 미세사출법 (microprojectile bombardment ) , 일렉트로포레이션 (electroporation), 입자 총 충격 (particle gun bombardment ) , 실리콘 탄화물 위스커 (Silicon carbide whiskers), 초음파 처리 (sonication), PEG-매개 융합법 (PEG-mediated fusion), 미세주입법 (microinjection), 리포좀 매개법 ( 1 iposome-mediated method), 자성나노입자 매개 법 (magnetic nanopart icle一 mediated method) 등에 의해 수행될 수 있다. 한편, 본 발명에서 사용된 표준 재조합 DNA 및 분자 클로닝 기술은 당해 분 야에 널리 공지되어 있고, 다음 문헌에 기재되어 있다 (Sambrook, J., Fritsch, E. F. and Maniatis, T., Molecular Cloning: A Laboratory Manual, 2nd ed. , Cold Spring Harbor Laboratory: Cold Spring Harbor , NY (1989); by Si lhavy, T. J., Bennan , M. L. and Enquist , L. W., Experiments with Gene Fusions, Cold Spring Harbor Laboratory: Cold Spring Harbor , NY (1984); and by Ausubel , F. M. et al . , Current Protocols in Molecular Biology, published by Greene Publishing Assoc. and Wi ley-lnterscience (1987)) . As used herein, the term “fusion protein” refers to a protein or polypeptide having an amino acid sequence derived from two or more proteins. It may also comprise a linkage region of amino acids between amino acid moieties derived from. The cell may be a cell of an animal, a plant, a yeast and a bacterium. Preferably, the cell may easily receive a first construct and a second construct, which are introduced from outside of the bacterium, and may be used for intracellular organelles such as cytoplasm and nucleus. Cells with distinct boundaries are preferred. More preferably CHO-kl (ATCC CCL-61, Cricetulus griseus, hamster, Chinese), HEK293 (ATCCCRL-1573, Homo sapiens, human), HeLa (ATCC CCL-2, Homo sapiens, human), SH-SY5Y ( ATCC CRL-2266, Homo sapiens, human), Swiss 3T3 (ATCC CCL-92, Mus musculus, mouse), 3T3-L1CATCC CL-173, Mus musculus, mouse), NIH / 3T3CATCC CRL-1658, Mus musculus, mouse) , L-929 (ATCC CCL-1, Mus musculus, mouse), Rat2 (ATCCCRL-1764, Rat t us norvegicus, rat), RBL-2H3 (ATCC CRL-2256, Rat t us norvegicus, rat), MDCK (ATCC CCL-34, Can is familiar is). In addition, it may be other stem cells, cells extracted from various tissues, and artificially made similar membrane structures. In the present invention, the cells comprising the crab first construct and the second construct may be prepared by molecular biological methods known in the art. Although not limited thereto, the cells are transformed into individual expression vectors capable of expressing the first component and the second component or expression vectors capable of expressing both the first component and the second component, and then the system is expressed in the expression vector. Preferred is a method by which the first and second constructs are expressed. The transformation of each expression vector into cells can be carried out using transformation methods known in the art, such as calcium phosphate, calcium chloride, rubidium chloride, microprojectile bombardment, electroporation, Particle gun bombardment, silicon carbide whiskers, sonication, PEG-mediated fusion, microinjection, liposome mediation (1 iposome-mediated method), magnetic nanoparticle mediated method, and the like. Meanwhile, standard recombinant DNA and molecular cloning techniques used in the present invention are well known in the art and described in the following literature (Sambrook, J., Fritsch, EF and Maniatis, T., Molecular Cloning: A Laboratory). Manual, 2nd ed., Cold Spring Harbor Laboratory: Cold Spring Harbor, NY (1989); by Si lhavy, TJ, Bennan, ML and Enquist, LW, Experiments with Gene Fusions, Cold Spring Harbor Laboratory: Cold Spring Harbor, NY (1984); and by Ausubel, FM et al. , Current Protocols in Molecular Biology, published by Greene Publishing Assoc. and Wi ley-lnterscience (1987)).
<55>  <55>
<56> 본 발명에 사용된 용어 "상호 작용' '이란 특정 리간드 또는 화합물, 또는 그 의 일부 또는 단편과, 관심있는 제 2 분자의 일부 간의 근접한 상태를 지칭한다. 상 호 작용은, 예를 들어 수소-결합, 반 데르 발스 (van der Waals) 상호 작용, 정전 기 또는 소수성 상호 작용의 결과로서 비―공유적이거나, 또는 공유적일 수 있다. <57> 본 발명의 상기 "신호물질" 은 PM Phorbol 12-myri state 13-acetate, As used herein, the term “interaction” refers to the proximal state between a particular ligand or compound, or a portion or fragment thereof, and a portion of the second molecule of interest. It may be non-covalent or covalent as a result of hydrogen-bonding, van der Waals interactions, electrostatic or hydrophobic interactions. 12-myri state 13-acetate,
Phorbol ester) , TPA( 12-ot et r adecanoy lphor bo 1 -13-acet a t e ) , PDBu(phorbol 12, 13-dibutyrate) , ATP(Adenosine triphosphate) , tr idecanoic ac i d , arachidonic acid, linoleic acid, DiC8, 130C937로 이루어진 군에서 선택된 것임을 특징으로 한다. Phorbol ester), TPA (12-ot et r adecanoy lphor bo 1 -13-acetate), PDBu (phorbol 12, 13-dibutyrate), Adenosine triphosphate (ATP), tr idecanoic ac id, arachidonic acid, linoleic acid, DiC8 It is characterized in that selected from the group consisting of 130C937.
<58>  <58>
<59> 본 발명의 상기 "신호물질의 처리" 는 PM Phorbol 12-myri state 13- acetate, Phorbol ester)를 50nM 내지 5υΜ의 농도로 처리하는 것임을 특징으로 한 다. 더 바람직하게는 luM로 처리할 수 있다. 상기 PMA의 처리농도가 50nM 미만의 경우 PKC를 사용한 이동모들의 이동이 층분하지 않고, 5uM 초과의 경우 화학물질의 과다처리에 의한 비정상적인 현상들 (세포사멸 신호교란 등)이 발생하여 바람직하 지 못하다.  The "treatment of signal substance" of the present invention is characterized by treating PM Phorbol 12-myri state 13-acetate, Phorbol ester) at a concentration of 50nM to 5υΜ. More preferably luM. When the concentration of the PMA is less than 50 nM, movement of the mobile mothers using the PKC is not difficult, and when the concentration of the PMA is greater than 5 uM, abnormal phenomena (eg, cell death signal disturbances, etc.) occur due to overtreatment of the chemical.
<60>  <60>
<61> 상기 "상호작용을 확인하는 단계" 는 제 1표지물질과 제 2표지물질의 분포를 검출하는 것으로, 예를 들어 확인하고자 하는 세포를 포함하는 커버슬립 The "confirming the interaction" is to detect the distribution of the first label and the second label, for example, coverslips containing the cells to be identified.
(coverslip)을 유체기 (perfusion chamber)에 고정시켜 공초점 레이저형광 현미경 ( 칼자이스 LSM710)의 제물대에 장착하고 외부 자극 전과 외부자극 후의 구성물 백터 들에 대한 영상을 획득하는 방법일 수 있다. It may be a method of fixing the coverslip to a perfusion chamber and mounting it on a material stage of a confocal laser fluorescence microscope (Cal Zeiss LSM710) and obtaining images of the component vectors before and after external stimulation.
<62>  <62>
<63> 본 발명은 상기 암 치료제 스크리닝 방법을 통해 스크리닝된 화합물을 제공 한다.  The present invention provides a compound screened through the cancer drug screening method.
<64> 본 발명의 상기 화합물은 하기 화학식 1로 표시될 수 있다. 【화학식 1] The compound of the present invention may be represented by the following formula (1). [Formula 1]
Figure imgf000010_0001
Figure imgf000010_0001
본 발명의 상기 화합물은 IkBa 단백질과 AURKC 단백질 결합을 억제하는 것을 특징으로 한다.  The compound of the present invention is characterized by inhibiting IkBa protein and AURKC protein binding.
본 발명의 상기 화함물이 IkBa 단백질과 AURKC 단백질의 결합을 억제하여 암 예방 및 치료 효과를 보이는 것을 특징으로 한다.  The compound of the present invention is characterized by inhibiting the binding of the IkBa protein and AURKC protein to show cancer prevention and treatment effects.
본 발명의 상기 "암" 은 위암, 폐암, 간암, 대장암, 소장암, 췌장암, 뇌암, 뼈암, 흑색종, 유방암, 경화성선증, 자궁암, 자궁경부암 두경부암, 식도암, 갑상 선암, 부갑상선암, 신장암, 육종, 전립선암, 요도암, 방광암, 혈액암, 림프종, 건 선 또는 섬유선종으로 이루어진 군으로부터 선택되는 것을 특징으로 한다. 본 발명은 IkBa와 AURKC간의 상호작용 저해제를 유효성분으로 포함하는 암 예방 및 치료용 조성물을 제공한다. 상기 저해제는 하기 화학식 1로 표시될 수 있다.  The "cancer" of the present invention is gastric cancer, lung cancer, liver cancer, colon cancer, small intestine cancer, pancreatic cancer, brain cancer, bone cancer, melanoma, breast cancer, sclerosis, uterine cancer, cervical cancer head and neck cancer, esophageal cancer, thyroid cancer, parathyroid cancer, kidney Cancer, sarcoma, prostate cancer, urethral cancer, bladder cancer, hematologic cancer, lymphoma, psoriasis or fibroadenoma. The present invention provides a composition for preventing and treating cancer, comprising an inhibitor of interaction between IkBa and AURKC as an active ingredient. The inhibitor may be represented by the following formula (1).
[화학식 1]  [Formula 1]
Figure imgf000010_0002
Figure imgf000010_0002
본 발명의 조성물은 약학적 조성물의 제조에 통상적으로 사용하는 적절한 담 체, 부형제 및 회석제를 더 포함할 수 있다. 또한 통상의 방법에 따라 산제, 과립 제, 정제, 캡술제, 현탁액, 에멀견, 시럽, 에어로졸 등의 경구형 제형, 외용제, 좌 제 및 멸균 주사용액의 형태로 제형화하여 사용될 수 있다. 당해 기술 분야에 알려 진 적합한 제제는 문헌 (Remington' s Pharmaceut i cal Science , 최근, Mack Publ i shing Company, Easton PA)에 개시되어 있는 것을 사용하는 것이 바람직하다. 포함될 수 있는 담체 부형제 및 희석제로는 락토오스, 덱스트로오스, 수크로오스, 소르비를, 만니를, 자일리를, 에리스리틀, 말티틀, 전분, 아카시아 고무, 알지네이 트, 젤라틴 , 칼슘포스페이트 칼슴 실리케이트 , 셀를로오스, 메틸 셀를로오스, 미 정질 샐를로오스 폴리비닐 피를리돈, 물, 메틸히드록시 벤조에이트, 프로필히드톡 시 벤조에이트, 탈크, 마그네슘 스테아레이트 및 광물유 등이 있다. 상기 조성물을 제제화할 경우에는 보통 사용하는 층진제, 증량제, 결합제, 습윤제, 붕해제, 계면 활성제 등의 희석제 또는 부형제를 사용하여 조제된다. 경구투여를 위한 고형제제 에는 정제, 환제, 산제, 과립제, 캡슐제 등이 포함되며, 이라한 고형제제는 상기 조성물에 적어도 하나 이상의 부형제 예를 들면 , 전분 , 칼슘 카보네이트 , 수크로오 스, 락토오스, 젤라틴 등을 섞어 조제된다. 또한 단순한 부형제 이외에 마그네슘 스테아레이트, 탈크 같은 윤활제들도 사용된다. 경구를 위한 액상 제제로는 현탁 제, 내용액제, 유제, 시럽게 등이 해당되는데 흔히 사용되는 단순 회석제인 물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존 제 등이 포함될 수 있다. 비경구 투여를 위한 제제에는 멸균된 수용액, 비수성용 제, 현탁제, 유제, 동결건조 제제, 좌제가 포함된다. 비수성용제, 현탁제로는 프로 필렌글리콜, 폴리에틸렌 글리콜 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기제로는 위템솔, 마크로 골, 트원 61, 카카오지, 라우린지, 글리세로제라틴 등이 사용될 수 있다. The composition of the present invention may further comprise suitable carriers, excipients and diluents commonly used in the manufacture of pharmaceutical compositions. It may also be used in the form of powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols and the like, oral formulations, external preparations, suppositories, and sterile injectable solutions according to conventional methods. Suitable formulations known in the art are preferably those disclosed in Remington's Pharmaceut i Cal Science, recently, Mack Publ i Shing Company, Easton PA. Carrier excipients and diluents that may be included include lactose, dextrose, sucrose, sorbitol, manny, xylly, erythritol, maltitol, starch, acacia rubber, alginine. , Gelatin, Calcium Phosphate Shampoo, Cellulose, Methyl Cellulose, Microcrystalline Salose Polyvinyl Pyridone, Water, Methylhydroxy Benzoate, Propylhydroxyxbenzoate, Talc, Magnesium Stearate and Mineral oil and the like. When formulating the composition, it is prepared using diluents or excipients such as layering agents, extenders, binders, wetting agents, disintegrating agents, and surfactants which are commonly used. Solid preparations for oral administration include tablets, pills, powders, granules, capsules, and the like. Such solid preparations may include at least one excipient such as starch, calcium carbonate, sucrose, lactose, It is prepared by mixing gelatin. In addition to simple excipients, lubricants such as magnesium stearate and talc are also used. Oral liquid preparations include suspensions, solvents, emulsions, and syrups.In addition to the commonly used simple diluents, water and liquid paraffin, various excipients such as wetting agents, sweeteners, fragrances, and preservatives are included. Can be. Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilized preparations, suppositories. As the non-aqueous solvent and suspending agent, vegetable oils such as propylene glycol, polyethylene glycol olive oil, injectable esters such as ethyl oleate, and the like can be used. As the base of the suppository, witemsol, macrogol, twenty-one, cacao butter, laurin butter, glycerogelatin and the like can be used.
<77> 본 발명의 상기 "치료" 란 특정 질병 또는 장애 증상을 개선시키는 것을 지 칭하고, 이는 이러한 장애를 치유하거나 장애 발병을 실질적으로 예방하거나, 또 는 대상체의 상태를 개선시키는 것을 포함할 수 있다. 본원에 사용된 바와 같은 용 어 "치료"는 환자를 고통스럽게 하는 소정의 장애에 대한 전 스펙트럼의 치료를 지 칭하는데, 이에는 해당 장애로부터 비롯된 한 가지 증상 또는 대부분의 증상을 완 화시키거나, 특정 장애를 치유하거나, 또는 장애 발병을 예방하는 것이 포함된다. The term “treatment” of the present invention refers to amelioration of a particular disease or disorder symptom, which may include treating such disorder, substantially preventing the development of the disorder, or improving the condition of the subject. . As used herein, the term “treatment” refers to the full spectrum of treatment for a given disorder that afflicts a patient, which alleviates one or most of the symptoms resulting from that disorder, Treating certain disorders or preventing the development of a disorder.
<78> <78>
<79> 나아가 , 본 발명의 약학적 조성물은 암을 예방 및 치료하는 효과를 가지는 공지의 화합물 또는 암 전이를 억제하는 효과를 가지는 공지의 화합물과 병행하여 투여할 수 있다.  Furthermore, the pharmaceutical composition of the present invention can be administered in parallel with a known compound having an effect of preventing and treating cancer or a known compound having an effect of inhibiting cancer metastasis.
<80>  <80>
<8i> 본 발명은 IkBa와 AURKC간의 상호작용 저해제의 암 예방 및 치료제 제조를 위한 용도를 제공한다.  <8i> The present invention provides a use of the inhibitor of the interaction between IkBa and AURKC for the manufacture of a prophylactic and therapeutic agent for cancer.
<82> 본 발명은 IkBa와 AURKC간의 상호작용 저해제를 이를 필요로 하는 개체에 유 효량으로 투여하는 것을 특징으로 하는 암 예방 및 치료방법을 제공한다.  The present invention provides a method for preventing and treating cancer, comprising administering an effective inhibitor of the interaction between IkBa and AURKC to a subject in need thereof.
<83> 상기 저해제는 하기 화학식 1로 표시될 수 있다. <83> The inhibitor may be represented by the following formula (1).
[화학식 1]  [Formula 1]
H
Figure imgf000012_0001
본 발명의 IkBa와 AURKC간의 상호작용 저해제는 유효량으로 경구, 경피, 피 하, 정맥 또는 근육을 포함한 여러 경로를 통해 투여될 수 있다. 상기에서 '유효 량' 이란 환자에게 투여하였을 때, 암의 치료 및 예방 효과 또는 암 전이 억제효과 를 나타내는 양을 말하며, 상기 개체 (subject ) ' 란 동물, 바람직하게는 포유동 물, 특히 인간을 포함하는 동물일 수 있으며, 동물에서 유래한 세포, 조직, 기관 등일 수도 있다. 상기 개체는 치료가 필요한 환자 (pat i ent )일 수 있다. 상기 본 발명의 IkBa와 AURKC간의 상호작용 저해제는 그 자체를 투여하거나 상술한 바와 같은 여러 제형으로 제조되어 투여될 수 있으며, 바람직하게는 원하는 효과 즉, 암 예방 및 치료 효과가 도출될 때까지 투여될 수 있다. 본 발명의 화합 물 및 이의 약학적으로 허용가능한 염은 당업계에 공지된 방법에 따라 다양한 경로 로 투여될 수 있다. 즉, 경구 또는 비경구, 예컨대 구강, 근육내, 정맥내, 피내, 동맥내, 골수내, 경막내, 목강내, 비강내, 질내, 직장내 설하 또는 피하 투여되거 나, 위장관, 점막 또는 호흡기로 투여될 수 있다. 바람직하게는 피부에 직접 도포 될 수 있다. 또한 본 발명의 화합물 및 이의 약학적으로 허용가능한 염은 표적 세 포나 조직에 고친화성 결합을 유발하는 분자와 결합되거나 상기 분자 내에 캡슐화 된 형태로 투여될 수 있다. 본 발명의 IkBa와 AURKC간의 상호작용 저해제는 당업계 에 공지된 기술을 이용하여 스테를 (예: 콜레스테롤) , 지질 (예: 양이온 지질, 비로 좀 또는 리포좀) 또는 표적 세포 특이적 결합게 (예: 표적세포 특이적 수용체에 의 해 인지되는 리간드)와 결합될 수 있다. 적합한 커플링제 또는 가교제로는 예컨대 단백질 A, 카보디이미드, N-숙신이미딜 -3-(2-피리딜디티오)프로피오테이트 (SPDP) 등을 포함할 수 있다. 이들 제형은 제약 화학에 일반적으로 공지된 처방서인 문헌 (Remington' s Pharmaceut i cal Science , 15th Edi t ion , 1975, Mack Publ i shing Company , East on , Pennsyl vani a)에 기술되어 있다.
H
Figure imgf000012_0001
Inhibitors of interaction between IkBa and AURKC of the present invention can be administered in an effective amount via several routes including oral, transdermal, subcutaneous, intravenous or intramuscular. As used herein, the term 'effective amount' refers to an amount that exhibits a therapeutic and prophylactic effect or a cancer metastasis inhibiting effect when administered to a patient, and the term 'subject' includes an animal, preferably a mammal, particularly a human. It may be an animal, cells, tissues, organs and the like derived from the animal. The subject may be a patient (pat i ent) in need of treatment. The interaction inhibitor between IkBa and AURKC of the present invention may be administered by itself or prepared and prepared in various formulations as described above, and is preferably administered until a desired effect, i.e., cancer prevention and treatment effect, is obtained. Can be. The compounds of the present invention and their pharmaceutically acceptable salts can be administered by various routes according to methods known in the art. That is, orally or parenterally, such as oral, intramuscular, intravenous, intradermal, intraarterial, intramedullary, intradural, intramural, intranasal, intravaginal, intra rectal sublingual or subcutaneous, gastrointestinal tract, mucosa or respiratory tract. May be administered. Preferably it can be applied directly to the skin. In addition, the compounds of the present invention and pharmaceutically acceptable salts thereof may be administered in a form that is bound to or encapsulated within a molecule that induces high affinity binding to the target cell or tissue. Inhibitors of interactions between IkBa and AURKC of the present invention may be prepared using techniques known in the art, such as sterols (e.g. cholesterol), lipids (e.g. cationic lipids, bivalent or liposomes) or target cell specific binding agents (e.g. Ligands recognized by target cell specific receptors). Suitable coupling or crosslinking agents can include, for example, Protein A, carbodiimide, N-succinimidyl-3- (2-pyridyldithio) propiotate (SPDP) and the like. These formulations are described in Remington's Pharmaceut i cal Science, 15th Edition, 1975, Mack Publ i Shing Company, East on, Pennsyl vani a).
<93>  <93>
【유리한 효과】  Advantageous Effects
<94> 따라서 본 발명은 IkBa 및 AURKC의 상호작용을 통한 암 치료제 스크리닝 방 법을 제공한다. 더욱 상세하게는 IkBa와 AURKC의 결합 억제제 화합물 및 이를 유효 성분으로 포함하는 암 예방 및 치료용 조성물을 제공한다. 본 발명의 스크리닝 방 법은 IkBa와 AURKC의 결합 억제 유무에 따라서 암 치료제를 쉽게 스크리닝할 수 있 다는 점에서 유용하게 이용될 수 있다.  Accordingly, the present invention provides a method for screening a cancer drug through the interaction of IkBa and AURKC. More specifically, the present invention provides a binding inhibitor compound of IkBa and AURKC and a composition for preventing and treating cancer comprising the same as an active ingredient. The screening method of the present invention can be usefully used in that the cancer therapeutic agent can be easily screened depending on the inhibition of binding of IkBa and AURKC.
<95>  <95>
【도면의 간단한 설명】  [Brief Description of Drawings]
<96> 도 1은 TMD-mRFP-IkBa (제 1구성물)과 EGFP-AURKC (제 2구성물)을 HEK293T 세포 주에 과발현 시키고 세포 이미징을 이용한 방법 (등록번호 10— 0948767)으로 분석한 것이다 (왼쪽 패널; 제 1 구성물, 가운데 패널; 제 2 구성물, 오른쪽 패널; 제 1구 성물 + 제 2구성물) .  FIG. 1 is an overexpression of TMD-mRFP-IkBa (first construct) and EGFP-AURKC (second construct) in HEK293T cell line and analyzed by a method using cell imaging (Registration No. 10-0948767) (left) Panel; first component, middle panel; second component, right panel; first component + second component).
<97> 도 2는 pTMD-mRFP-C3 p i asm i d (게 1구성물)와 EGFP-AURKC (거 12구성물)를 Figure 2 shows pTMD-mRFP-C3 p i asm i d (Crab 1 construct) and EGFP-AURKC (near 12 constructs)
HEK293T 세포주에 과발현시킨 후 1 M의 PMA (외부자극)를 3분간 처리한 결과를 나타낸 것이다 (왼쪽 패널; 제 1 구성물, 가운데 패널; 제 2 구성물 오른쪽 패널; 제 1구성물 + 제 2구성물) . Results of overexpression in HEK293T cell line followed by treatment with 1 M PMA (external stimulus) for 3 minutes (left panel; first construct, middle panel; second construct right panel; first construct + second construct).
<98> 도 3-가는 TMD-niRFP-AURKC (게 1구성물)와 EGFP- IkBa (제 2구성물)를 HEK293T 세 포 내 과발현시킨 후 1 μ Μ의 Ρ Α (외부자극)를 3분간 처리하여 두 단백질의 결합 을 실시간으로 분석한 결과이다 (왼쪽패널; 제 1 구성물, 가운데 패널; 제 2 구성 물, 오른쪽 패널; 제 1구성물 + 제 2구성물) . 도 3-나는 pTMD-mRFP— C3 p l asm i d ( 제 1구성물)와 EGFP- IkBa (게 2구성물)를 HEK293T 세포에 과발현시킨 후 1 μ Μ의 PMA (외부자극)를 3분간 처리하여 두 단백질의 결합을 실시간으로 분석한 결과이다 (왼 쪽패널; 제 1 구성물, 가운데 패널; 제 2 구성물, 오른쪽 패널; 제 1구성물 + 제 2구성물) .  3-98. TMD-niRFP-AURKC (Crab 1 constituent) and EGFP-IkBa (Second constituent) were overexpressed in HEK293T cells and treated with 1 μΜ Ρ Α (external stimulus) for 3 minutes. Results of real-time analysis of protein binding (left panel; first component, middle panel; second component, right panel; first component + second component). 3-I overexpresses pTMD-mRFP—C3 pl asm id (first construct) and EGFP-IkBa (crab construct) to HEK293T cells, followed by treatment of 1 μM PMA (external stimulus) for 3 minutes. Results of real-time analysis of binding (left panel; first component, middle panel; second component, right panel; first component + second component).
<99> 도 4는 TMD-mRFP-IkBa (제 1구성물)과 EGFP-AURKC (제 2구성물)을 CH0-K1 세포 주에 과발현시킨 후 1 μ Μ의 ΡΜΑ (외부자극)를 3분간 처리하여 두 단백질의 결합을 실시간으로 나타낸 사진이다 (왼쪽패널 : 제 1 구성물, 가운데 패널 : 제 2 구성물, 오른쪽 패널 : 제 1구성물 + 제 2구성물) .  4 shows that TMD-mRFP-IkBa (first construct) and EGFP-AURKC (second construct) were overexpressed in the CH0-K1 cell line, followed by treatment of 1 μΜ ΡΜΑ (external stimulus) for 3 minutes. This is a photo showing the binding of proteins in real time (left panel: first component, middle panel: second component, right panel: first component + second component).
<ι οο> 도 5a 내지 도 5d는 위암 세포주 AGS (가)와 SNU638 나) 및 대장암 세포주 5A to 5D show gastric cancer cell lines AGS (A) and SNU638 b) and colorectal cancer cell lines.
HCT116(다)과 SW620(라), 자궁암 세포주 He l a (마) , 유방암 세포주 MDA-MB-23U바) , 폐암 세포주 A549(사) 그리고 간암 세포주 HepG2(아)에서 세포독성을 측정한 결과 를 나타낸 그래프이다. HCT116 (C) and SW620 (D), uterine cancer cell line He la (E), breast cancer cell line MDA-MB-23U), Cytotoxicity was measured in lung cancer cell line A549 (A) and liver cancer cell line HepG2 (H).
<ιοι> 도 6은 TMD-mRFP-IkBa (제 1구성물)과 EGFP-AURKC (제 2구성물)을 HEK293T 세 포 (패널 가)와 CH0-K1 세포주 (패널 나)에 동일한 방법으로 과발현 시키고 화합물 50 μΜ을 3시간 처리하였다. 그 후 1 μΜ의 ΡΜΑ (외부자극)를 3분간 처리하여 두 단백질의 결합 억제를 실시간으로 분석한 사진이다 (왼쪽패널: 제 1 구성물, 가운데 패널: 제 2 구성물ᅳ 오른쪽 패널 : 제 1구성물 + 제 2구성물).  FIG. 6 shows that TMD-mRFP-IkBa (first construct) and EGFP-AURKC (second construct) are overexpressed in HEK293T cells (Panel A) and CH0-K1 cell lines (Panel B) in the same manner and Compound 50 μM was treated for 3 hours. After that, 1 μΜ ΡΜΑ (external stimulus) was treated for 3 minutes to analyze the inhibition of binding of the two proteins in real time (left panel: 1st component, middle panel: 2nd component ᅳ right panel: 1st component + 1st) 2 components).
<102> 도 7은 유방암 세포주 MDA-MB-231 세포를 배양하여 상처 치유 분석 (Wound healing assay) (가) , 트렌스웰 이동 분석 (Transwell migration assay) (나) 그리고 연 한천 분석 (soft agar assay) (다)을 한 결과로 IkBa와 AURKC의 상호작용을 저해 하는 화합물이 암세포 증식에 미치는 영향을 확인한 결과이다.  FIG. 7 shows a wound healing assay (A), a Transwell migration assay (B) and a soft agar assay by culturing breast cancer cell line MDA-MB-231 cells. As a result of (c), the effect of compounds inhibiting the interaction of IkBa and AURKC on cancer cell proliferation was confirmed.
<103>  <103>
【발명의 실시를 위한 형태】  [Form for implementation of invention]
<104> 이하 본 발명을 상세히 설명한다 . Hereinafter, the present invention will be described in detail.
<105> 단, 하기 실시예는 본 발명을 예시하는 것일 뿐, 본 발명의 내용이 하기 실 시예에 한정되는 것은 아니다.  However, the following examples are merely to illustrate the invention, but the content of the present invention is not limited to the following examples.
<106>  <106>
<107> <실시예 1>  <Example 1>
<108> 동물세포주 및 형질전환  <108> Animal Cell Lines and Transformation
<109>  <109>
<ιιο> <1-1> 동물세포주 및 이의 배양  <ιιο> <1-1> Animal cell lines and their culture
<ιιι> 본 발명에서의 실험은 CHOKKATCC CCL-61, Cricetulus griseus, hamster,  <ιιι> Experiments in the present invention are CHOKKATCC CCL-61, Cricetulus griseus, hamster,
Chinese), HEK293(ATCC CRL-1573, Homo sapiens, human), MDA-MB-231 (ATCC HTB-26 , Homo sapiens, human) , A549 (ATCC CCᄂ一 185 Homo sapiens , human) , HepG2 (ATCC HB-8065, HepG2, Homo sapiens, human) 세포주를 사용하였다. 본 발명에 사용된 동 물세포주의 배양조건은 각 세포주의 분양기관인 ATCC(American Type Culture Col lection)사의 세포주 배양방법을 이용하였다. 단, CH0-K1은 F-12 배양액을 사용 하였고, HEK293, MDA-MB-231 세포주는 DMEM 배양액을 사용하였고, A549, HepG2 세 포주는 RPMI 1640 배양액을 사용하였으며 기타 배양조건은 동일하게 사용하였다. 한편, 각 세포들의 공통적인 배양방법은 다음과 같다 (당업자의 목적에 의해 세부적 인 배양 조건은 달라질 수 있다. ) . 25 niM HEPES, 10% FBS( fetal bovine serum, v/v), 100 units/ml 페니실린, 100 ug/ml 스트랩토마이신이 들어있는 pH7.4의 배양 액 (F-12 및 DMEM) 속에서 각 세포주를 온도 37°C, C02분압 5%가 유지되는 배양기에 서 배양하였다. Chinese), HEK293 (ATCC CRL-1573, Homo sapiens, human), MDA-MB-231 (ATCC HTB-26, Homo sapiens, human), A549 (ATCC CC 一 185 Homo sapiens, human), HepG2 (ATCC HB -8065, HepG2, Homo sapiens, human) cell line was used. Culture conditions of animal cell lines used in the present invention was used for cell line culture method of ATCC (American Type Culture Collection), which is the organ of each cell line. However, CH0-K1 was used for F-12 culture, HEK293 and MDA-MB-231 cell lines were used for DMEM culture, A549 and HepG2 cells were used for RPMI 1640, and other culture conditions were used in the same manner. On the other hand, the common culture method of each cell is as follows (detailed culture conditions may vary depending on the purpose of the skilled person). Incubation at pH 7.4 with 25 niM HEPES, 10% FBS (fetal bovine serum, v / v), 100 units / ml penicillin, 100 ug / ml straptomycin In cell (F-12 and DMEM), each cell line was incubated in an incubator maintained at a temperature of 37 ° C., CO 2 partial pressure of 5%.
<112>  <112>
<!13> <1-2> 세포주의 형질전환  <! 13> <1-2> Transformation of Cell Line
<114> 본 발명의 실시예에서 사용된 세포내 유전자 도입방법은 일반적으로 사용되 는 리포좀 -기반 방법 중의 하나인 Fugene HD (Promega)을 사용하였으며, 유전자의 농도 등 유전자 도입을 위한 모든 조건은 제조사의 지침을 따라 수행하였다. 보다 구체적으로는 6-플레이트에 커버슬립을 넣고 세포를 하루 동안 배양한 후, 페니실 린과 스트렙토마이신, FBS가 없는 2 ml의 신선한 배양액으로 교환하였다. 약 1 정도의 형질전흰ᅳ 시료들을 0.1 ml의 페니실린과 스트렙토마이신ᅳ FBS가 없는 배양 액에 첨가한 후 완전히 섞고 3 의 Fugene HD 시약을 첨가하여 섞어주었다. 이 용액을 상온에서 15분간 놓아둔 후 세포가 자라고 있는 커버슬립이 들어있는 6-플 레이트의 각 웰 (well)에 첨가하여 4시간 동안 배양 한 뒤 FBS를 200 μ 1 넣고 18시 간 형질전환되도록 하였다.  Intracellular gene introduction method used in the embodiment of the present invention used Fugene HD (Promega), one of the liposome-based methods commonly used, all conditions for gene introduction, such as the concentration of the gene is a manufacturer Follow the instructions. More specifically, the coverslips were placed in 6-plates, and the cells were cultured for one day, and then exchanged with 2 ml of fresh culture without penicillin, streptomycin, or FBS. About one transgenic white sample was added to the culture medium without 0.1 ml of penicillin and streptomycin FBS, followed by complete mixing and addition of 3 Fugene HD reagents. The solution was allowed to stand at room temperature for 15 minutes, then added to each well of a 6-plate containing the coverslips in which the cells were grown, incubated for 4 hours, and 200 μl of FBS was added for transformation for 18 hours. It was.
<] 15>  <] 15>
<116> <실시예 2>  <Example 2>
<ιΐ7> 제 1구성물 및 제 2구성물의 디자인 및 제작  <ιΐ7> Design and manufacture of the first and second components
<118>  <118>
<1!9> <2-1> 제 1구성물의 설계 및 제작  <1! 9> <2-1> Design and Fabrication of First Component
<120> 본 발명에서 제 1구성물이란 세포질에서 고르게 발현되어 있는 단백질이 신호 무질을 처리하였을 때 세포막으로 이동할 수 있는 모듈 (이동모들)을 포함하며, 이 들 현미경을 이용하여 분석할 수 있는 적색형광단백질이 표지되어 결합되어 있고 마지막으로 목적물질이 결합될 수 있는 융합서열로 구성된다. 제 1구성물은 TMD- mRFP-empty 백터에 IkBa 및 AURKC를 제한효소 EcoRI/Kpnl (IkBa) 및 EcoRI/Xinal (AURKC)를 이용하여 제작하였다. TMD— mRFP-empty 백터는 서열번호 5로 표시되는 Protein Kinase C mutant, (TMD)와 서열번호 7으로 표시되는 mRFP를 포함하는 백터 이다.  In the present invention, the first constituent includes a module (mobile mothers) that can move to the cell membrane when the protein evenly expressed in the cytoplasm has been treated with the signal amorphous substance, and can be analyzed using these microscopes. The protein is labeled and bound, and finally consists of a fusion sequence to which the target substance can be bound. The first construct was constructed using restriction enzymes EcoRI / Kpnl (IkBa) and EcoRI / Xinal (AURKC) with IkBa and AURKC in a TMD-mRFP-empty vector. TMD—mRFP-empty vector is a vector comprising a Protein Kinase C mutant (TMD) represented by SEQ ID NO: 5 and mRFP represented by SEQ ID NO: 7.
<i2i> IkBa는 주형으로 pcDNA3.1-IkBa 백터 (NCBI Reference Sequence:  <i2i> IkBa is the template for pcDNA3.1-IkBa vector (NCBI Reference Sequence:
NM_020529.2; purchased from Mediscov Inc)를 사용하고, 서열번호 1 및 서열번호 2의 프라이머를 사용하여 PCR을 수행하여 제조하였다. 또한, AURKC는 주형으로 PPTB7-AURKC 백터 (GenBank : BC002363)를 사용하고, 서열번호 3 및 서열번호 4의 프라이머를 사용하여 PCR올 수행하였다. 프라이머를 사용하여 PCR 증폭한 후 수득 물을 pTMD-mRFP-C3 백터의 EcoRI/Kpnl (IkBa) 및 EcoRI/Xmal (AURKC)위치에 삽입하 여 제작하였다. NM_020529.2; purchased from Mediscov Inc), and PCR was performed using primers of SEQ ID NO: 1 and SEQ ID NO: 2. In addition, AURKC performed PCR using a PPTB7-AURKC vector (GenBank: BC002363) as a template and primers of SEQ ID NO: 3 and SEQ ID NO: 4. Obtained after PCR amplification using primers Water was prepared by inserting into the EcoRI / Kpnl (IkBa) and EcoRI / Xmal (AURKC) positions of the pTMD-mRFP-C3 vector.
<122>  <122>
<123> <2-2> 제 2구성물의 설계 및 제작  <123> <2-2> Design and Fabrication of Second Component
<124> 제 2구성물이란 계 1구성물에 결합된 목적물질과 결합하는 내재적인 특성을 가 진 표적물질의 이동을 분석하기 위한 표지물을 포함한다. 게 2구성물은 제 1구성물에 포함된 표지와 다른 형광인 녹색형광단백질 (EGFP)을 이용한다. 제 2구성물은 기존에 등록된 특허 (등톡번호: 10-1217718)의 EFRP-C3 백터 (clontech)에 IkBa 및 AURKC를 제한효소 EcoRI/Kpnl (IkBa) 및 EcoRI/Xmal (AURKC)를 이용하여 제작하였다. <125> IkBa는 주형으로 pcDNA3.1— IkBa 백터 (NCBI Reference Sequence: The second construct includes a label for analyzing the movement of a target substance having intrinsic properties of binding to a target substance bound to the first component. Crab bicomponent uses green fluorescent protein (EGFP), a fluorescence that is different from the label contained in the first construct. The second construct was constructed using restriction enzymes EcoRI / Kpnl (IkBa) and EcoRI / Xmal (AURKC) from IkBa and AURKC in an EFRP-C3 vector (clontech) of a previously registered patent (e.g. 10-1217718). . <K> IkBa is the template for the pcDNA3.1—IkBa vector (NCBI Reference Sequence:
M_020529.2; purchased from Mediscov Inc)를 사용하고, 서열번호 1및 서열번호 2 의 프라이머를 사용하여 PCR을 수행하여 제조하였다. 또한, AURKC는 주형으로 PPTB7-AUR C 백터 (GenBank : BC002363)를 사용하고, 서열번호 3 및 서열번호 4의 프라이머를 사용하여 PCR을 수행하였다. 프라이머를 사용하여 PCR 증폭한 후 수득 물을 pTMD-mRFP-C3 백터의 EcoRI/Kpnl (IkBa) 및 EcoRI/Xnial (AURKC)위치에 삽입하 여 제작하였다.  M_020529.2; purchased from Mediscov Inc), and PCR was performed using primers of SEQ ID NO: 1 and SEQ ID NO: 2. In addition, AURKC was performed using a PPTB7-AUR C vector (GenBank: BC002363) as a template and using primers of SEQ ID NO: 3 and SEQ ID NO: 4. After PCR amplification using the primers, the obtained product was prepared by inserting into the EcoRI / Kpnl (IkBa) and EcoRI / Xnial (AURKC) positions of the pTMD-mRFP-C3 vector.
<126>  <126>
<127> <실시예 3>  <127> <Example 3>
<128> 제 1구성물 및 제 2구성물의 세포 내 이동 확인  Intracellular migration of the first and second constructs
<129> 제 1구성물과 제 2구성물 백터가 도입된 세포가 자라고 있는 커버슬립  <129> Coverslips in which cells in which the first and second construct vectors are introduced grow
(coverslip)을 유체기 (perfusion chamber)에 고정시켜 공초점 레이저형광현미경 (칼 자이스 LSM710)의 제물대에 장착하고 외부자극 전과 외부자극 (ΙμΜ PMA(Phorbol ester) 처리) 후의 구성물 백터들에 대한 영상을 획득하였다.  (coverslip) mounted in a perfusion chamber, mounted on the product of the confocal laser fluorescence microscope (Cal Zeiss LSM710) and image of the composition vectors before and after external stimulation (ΙμΜ Phorbol ester treatment) Obtained.
<130> 공초점 레이저현미경의 레이저는 488 nni Argon 레이저 (EGFP), 543 nm HeNe 레이저 (niRFP)를 사용하여 형광표지를 여기 (excitation)시키고, 각 형광표지에서 발생하는 형광신호는 band path 필터 BP505-530 (EGFP), Long path 필터 LP560 또 는 BP560-630 (niRFP)를 사용하였으며, 각 형광들 간의 간섭을 완전히 제거한 후 영 상을 획득하였다. The confocal laser microscope uses a 488 nni Argon laser (EGFP) and a 543 nm HeNe laser (niRFP) to excite the fluorescent label, and the fluorescent signal generated from each fluorescent label is a band path filter BP505. -530 (EGFP), Long path filter LP560 or BP560-630 (niRFP) were used, and images were obtained after the interference between each fluorescence was completely removed.
<131> 그 결과 도 1, 도 3-가 및 도 4와 같이 공초점 레이저형광현미경을 이용하여 외 부 자극 후 이동모들 (TMD)이 포함된 제 1구성물 백터에 의한 적색 형광은 세포막으 로 이동되지만 이동모들이 없는 표적물질용 제 2구성물 백터에 의한 녹색 형광은 자 극전과 동일하게 세포질에 고루 분포하고 있다. 따라서 거 12구성물 백터는 외부자극 에 반웅하지 않으며, 제 2구성물의 세포막으로의 이동은 반드시 목적물질과 표적물 질의 결합을 전제로 한다. As a result, as shown in FIGS. 1, 3, and 4, the red fluorescence caused by the first component vector including the moving mothers (TMD) after the external stimulation using the confocal laser fluorescence microscope is transferred to the cell membrane. However, the green fluorescence by the second material vector for the target material without the mobile hairs is distributed evenly in the cytoplasm as in stimulation. Thus, a 12-element vector is an external stimulus Inevitably, the migration of the second constituent to the cell membrane is necessarily based on the binding of the target substance to the target substance.
<132>  <132>
<133> <실시여 Γ 4>  <133> <embodiment Γ 4>
<134> 제 1구성물 및 제 2구성물을 이용하여 IkBa과 AURKC의 세포내 결합 분석  Intracellular Binding Analysis of IkBa and AURKC Using First and Second Constructs
<135> TNF-a l pha에 의해 조절되는 IkBa 단백질의 상호작용은 세포 내에서 다양한 세포신호전달기작에 관여하는 것으로 알려져 있다. 다양한 기작 중 특히 주목되고 있는 것은 생체내에서 발생하는 각종 위해성 신호에 대웅하는 염증유발과 밀접한 관련성을 가지고 있다는 것이다. IkBa와 상호작용하는 AURKC 단백질의 결합여부를 살아있는 세포를 이용하여 실시간으로 실험을 실시하였다.  The interaction of IkBa proteins regulated by TNF-a pha is known to be involved in various cellular signaling mechanisms in cells. Of particular interest among the various mechanisms is that they have a close relationship with provoking inflammation in response to various risk signals occurring in vivo. The binding of AURKC protein interacting with IkBa was performed in real time using living cells.
<136> 상기에서 제조한 제 1구성물 및 제 2구성물을 HEK293T와 CH0-K1 세포주에 형질 전환하였고, 이를 0분과 3분째에 형광을 확인하였다.  The first and second constructs prepared above were transformed into HEK293T and CH0-K1 cell lines, and their fluorescence was confirmed at 0 and 3 minutes.
<137> 그 결과, 도 1에서와 같이, 제 1구성물 (TMD-mRFP-IkBa)은 0분째에는 세포내에 고르게 분포하다가, 3분 후에 세포막으로 이동하였으며 (도 1 왼쪽 패널) , 제 2구성물 (EGFP-AURKC)도 역시 0분째에는 세포내에 고르게 분포하다가 3분 후 제 1구성물과 더불어 세포막으로 이동함을 알 수 있었다 (도 1 가운데 패널) . 이는 제 1구성물과 제 2구성물이 결합하였음을 의미한다.  As a result, as shown in Fig. 1, the first construct (TMD-mRFP-IkBa) was evenly distributed in the cell at 0 minutes, and then moved to the cell membrane after 3 minutes (left panel in Fig. 1). EGFP-AURKC) was also distributed evenly in the cell at 0 min and then moved to the cell membrane with the first construct after 3 min (panel in FIG. 1). This means that the first component and the second component are combined.
<138>  <138>
<139> 반대로 제 1구성물을 TMD-mFRP-AU KC로 게 2구성물을 EGRP— IkBa로 바꾸어  In contrast, the first component is replaced by TMD-mFRP-AU KC and the second component is replaced by EGRP—IkBa.
HEK2953T와 CH0-K1 세포주에 형질전환 하였고, 이를 0분과 3분째에 형광을 확인하 였다.  HEK2953T and CH0-K1 cell lines were transformed and fluorescence was detected at 0 and 3 minutes.
<140> 그 결과 도 3-가에서와 같이, 제 1구성물 (TMD-mRFP-AURKC)은 0분째에는 세포 내에 고르게 분포하다가, 3분 후에 세포막으로 이동하였으며 (도 3-가 왼쪽 패널), 제 2구성물 (EGFP-IkBa)도 역시 0분째에는 세포내에 고르게 분포하다가 3분 후 제 1구 성물과 더불어 세포막으로 이동함을 알 수 있었다 (도 3-가 가운데 패널) . 이는 제 1 구성물과 제 2구성물이 결합하였음을 의미한다.  As a result, as shown in Fig. 3A, the first construct (TMD-mRFP-AURKC) was evenly distributed in the cells at 0 minutes, and then moved to the cell membrane after 3 minutes (Fig. 3 is the left panel). The two constructs (EGFP-IkBa) were also distributed evenly within the cell at 0 min and then migrated to the cell membrane with the first construct after 3 min (Figure 3-middle panel). This means that the first component and the second component are combined.
< 141 >  <141>
<142> <실시여】 5>  <142> <Release> 5>
<143> IkBa과 AURKC의 결합을 억제하는 화합물 스크리닝  <143> Screening Compounds That Inhibit the Binding of IkBa and AURKC
<144> 상기 스크리닝 방법을 이용하여 상기에서 제조한 제 1구성물 및 제 2구성물을  Using the screening method, the first and second compositions prepared above are prepared.
6-플레이트 내에 HEK293T와 CH0-K1 세포가 자라고 있는 커버슬립 (covers l i p)에 형 질전환 하였다. 그 후 IkBa 단백질과 AURKC 단백질 결합 억제제 후보물질을 3시간 동안 처리하였다. 커버슬립 (coverslip)을 유체기 (perfusion chamber)에 고정시켜 공초점 레이저형광 현미경 (칼자이스 LSM710)의 제물대에 장착하고 외부 자극 전과 외부자극 (ΙμΜ PMA(Phorbol ester)) 후의 구성물 백터들에 대한 영상을 획득하였 다. The cells were transformed into coverslips in which HEK293T and CH0-K1 cells grew. Afterwards, IkBa protein and AURKC protein binding inhibitor candidates were Treated during. The coverslip is secured to the perfusion chamber, mounted on the stage of the confocal laser fluorescence microscope (Cal Zeiss LSM710), for component vectors before and after external stimulation (ΙμΜ Phorbol ester). Image was acquired.
그 결과, IkBa 단백질과 AURKC 단백질의 상호작용을 억제하는 하기 화학식 1 로 표시되는 화합물을 스크리닝 하였다.  As a result, the compounds represented by the following formula (1) inhibiting the interaction of IkBa protein and AURKC protein were screened.
[화학식 1]  [Formula 1]
Figure imgf000018_0001
Figure imgf000018_0001
도 6은 상기 화합물의 IkBa 단백질과 AURKC 단백질의 결합 저해 효과를 분석 한 것으로 TMD-mRFP-IkBa (제 1구성물)과 EGFP-AURKC (게 2구성물)을 HEK293T 세포와 CH0-K1 세포주에 동일한 방법으로 과발현 시키고 상기 화합물 50 μΜ을 3시간동안 처리하였다. 그 후 1 μΜ의 ΡΜΑ (외부자극)를 3분간 처리하여 두 단백질의 결합 억 제를 실시간으로 분석하였다. 위 화합물에 의해 IkBa 단백질과 AURKC 단백질이 결 합하지 못하고 이동모들 및 IkBa를 반영하는 적색형광은 세포막으로 이동하였으나 AURKC 단백질을 반영하는 녹색형광은 그대로 세포질에 위치하였음을 확인하였다. 따라서 상기 화학식 1로 표시되는 화합물이 IkBa와 AURKC의 결합을 저해하는 저해제 로 사용될 수 있다는 것을 보여준다.  Figure 6 is an analysis of the binding inhibitory effect of the IkBa protein and AURKC protein of the compound TMD-mRFP-IkBa (first component) and EGFP-AURKC (component 2) in the same manner in HEK293T cells and CH0-K1 cell line Overexpressed and treated 50 μM of the compound for 3 hours. Thereafter, 1 μΜ of ΡΜΑ (external stimulus) was treated for 3 minutes to analyze the inhibition of binding of both proteins in real time. It was confirmed that the above compound did not bind the IkBa protein and the AURKC protein, and the red fluorescence reflecting the moving mothers and IkBa moved to the cell membrane, but the green fluorescence reflecting the AURKC protein was located in the cytoplasm. Therefore, it can be seen that the compound represented by Chemical Formula 1 can be used as an inhibitor that inhibits the binding of IkBa and AURKC.
<실시예 6> <Example 6>
스크리닝된 화합물의 세포독성 측정  Cytotoxicity Measurements of Screened Compounds
발명의 실시예에서 사용된 화합물의 세포독성은 일반적으로 사용되는 CCK8 키트 (Dojindo Molecular Technologies)를 사용하였으며 모든 조건은 제조사의 지 침을 따라 수행하였다. 보다 구체적으로 96-플레이트에 세포를 하루 동안 배양한 후 10, 50, 100 μΜ의 화합물을 처리하고 24, 48 시간 배양한 뒤 CCK8 용액을 10 μ ΐ 넣고 한 시간 뒤 450 nm에서 흡광도를 측정하였다.  Cytotoxicity of the compounds used in the examples of the invention was used a commonly used CCK8 kit (Dojindo Molecular Technologies) and all conditions were performed according to the manufacturer's instructions. More specifically, cells were cultured in 96-plate for one day, 10, 50, and 100 μΜ were treated with the compound and incubated for 24 and 48 hours, followed by 10 μl of CCK8 solution, and the absorbance was measured at 450 nm for one hour.
그 결과 도 5 와 같이 위암 세포주 AGS와 SNU638 (도 5 가, 나) 및 대장암 세 포주 HCT116과 SW620 (다, 라) , 자궁암 세포주 Hela (마)ᅳ 유방암 세포주 MDA— MB- 231(바), 폐암 세포주 A549(사) 그리고 간암 세포주 HepG2(아)에서 세포독성을 측 정한 결과 IkBa와 AURKC의 상호작용을 저해하는 화합물에 의해 이들 암세포의 세포 사멸이 증가하였다. As a result, gastric cancer cell lines AGS and SNU638 (FIG. 5, B) and colon cancer cell lines HCT116 and SW620 (C), uterine cancer cell line Hela (MA) ᅳ breast cancer cell line MDA—MB-231 (F), as shown in FIG. Cytotoxicity was measured in lung cancer cell line A549 and HepG2 cell line, and these cancer cells were inhibited by compounds that inhibit the interaction of IkBa and AURKC. Death increased.
<154>  <154>
<155> <실시예 7>  <155> <Example 7>
<156> 스크리닝 된 화합물의 암세포 증식 억제  Inhibition of Cancer Cell Proliferation of Screened Compounds
<157>  <157>
<158> <7-1> 암세포 이동 능력 (migration) 분석 방법  <158> <7-1> Analysis method of cancer cell migration ability
<159> 암세포 이동 능력 (migration) 분석 방법은 37°C 5% C02 조건에서 10% FBS가 들어간 DMEM에서 배양한 유방암 세포주 MDA— MB-231를 이용했다 (도 7가). 세포를 24 웰 조직 배양 플레이트에 (2 X 105 cells) 로 24시간 배양한다. 그 후 배지를 바꾸 지 않고, 천천히 lnil 피펫 팁을 이용하여 웰의 중앙을 가로질러 상처를 낸다. 이 때ᅳ 팁을 웰 바닥과 수직으로 세워서 사용하고, 상처를 한 방향으로 낸다, 그리고 떨어진 세포들을 제거하기 위해 배지로 두 번 행군 뒤 새 배지를 보층해 준다. 그 리고 실험 군 플레이트쎄 IkBa와 AURKC의 상호작용을 저해하는 화합물 25μΜ을 처 리한 뒤 48시간 동안 배양한다. 그 뒤에 IX PBS로 2번 행군 뒤 30분 동안 3.7% 파 라포름알데하이드를 이용해 고정시킨다. 고정된 세포를 2% 에탄을에 1% 크리스탈 바이을렛을 녹여서 30분간 염색한 뒤 현미경을 이용해서 관찰했다. The cancer cell migration assay was performed using a breast cancer cell line MDA—MB-231 cultured in DMEM containing 10% FBS at 37 ° C. 5% CO 2 conditions (FIG. 7A). Cells are incubated for 24 hours in 24 well tissue culture plates (2 × 10 5 cells). Thereafter, the wound is slowly cut across the center of the well using a lnil pipette tip without changing medium. At this time, use the tip upright with the bottom of the well, cut the wound in one direction, and march twice with medium to remove the fallen cells, and then supplement the new medium. In addition, 25 μM of compounds inhibiting the interaction of the plate group IkBa with AURKC were treated and incubated for 48 hours. This is followed by two marches with IX PBS followed by fixation with 3.7% paraformaldehyde for 30 minutes. The fixed cells were stained for 30 minutes by dissolving 1% crystal vial in 2% ethane and observed using a microscope.
<160> 그 결과 도 7가와 같이 유방암 세포주에 IkBa와 AURKC의 상호작용을 저해하 는 화합물을 처리하지 않은 대조군 (ΟμΜ)의 경우 상처의 폭이 상기 화합물을 처리 한 경우 (25μΜ)에 비해 좁은 것을 확인했다. 대조군의 경우에는 팁으로 상처 낸 부 위에 세포가 자라서 공간을 채웠지만, 화합물을 처리한 경우에는 상처 낸 부위에 세포가 자라지 않아서 공간이 그대로 남아 있었다. 즉, IkBa와 AURKC의 상호작용을 저해하는 화합물이 세포의 증식을 억제하는 것을 확인하였다. As a result, as shown in FIG. 7, the control group (ΟμΜ) without treatment of the compound that inhibits the interaction of IkBa and AURKC in the breast cancer cell line was narrower than that of the treatment with the compound (25μΜ). Confirmed. In the case of the control group, the cells grew on the wounded portion with the tip to fill the space, but when the compound was treated, the cells did not grow on the wounded portion and the space remained. That is, it was confirmed that the compound which inhibits the interaction of IkBa and AURKC inhibits the proliferation of cells.
<161>  <161>
<162> <7-2> 트랜스 웰 이동 측정  <162> <7-2> Transwell Transfer Measurement
<163> 트랜스 웰 이동 측정법 (Transwell migration assay)는 1 FBS인 DMEM으로 배양한 유방암 세포주 MDA-MB— 231를 이용했다 (도 7나). 트랜스 웰을 준비하는 것 은 24 웰 아래 부분에 0.5% FBS와 2.6ηᅵ 1 DMEM을 넣고, 8 구멍 크기인 트랜스 웰 을 넣었다. 그 다음 준비된 트랜스 웰에 세포를 (1 X 105 cells)를 넣어주었다. 그 리고 IkBa와 AURKC의 상호작용을 저해하는 화합물을 ΙΟμΜ, 25μΜ을 넣어준 다음 세포를 37°C, 5% C02에서 2시간 30분 동안 배양하며, 세포가 끼운 필터의 아래 쪽을 향해 이동하도록 하였다. 2시간 30분 후에 필터를 빼서 필터 위의 남아 있는 세포 잔해를 면봉을 이용해서 제거하였다. 그리고 필터 아래쪽의 세포들은 5% 글루타알 데하이드로 10분간 고정한 뒤 1% 크리스탈 바이올렛이 녹은 2% 에탄올로 20분간 염 색하였다. 크리스탈 바이올렛을 제거하고, 현미경으로 관찰하며 필터의 아래쪽 부 분의 세포 수를 측정했다. 이 때, 필터의 다른 부분들을 선택하여 관찰한 뒤 평균 을 냈다. The transwell migration assay used a breast cancer cell line MDA-MB-231 incubated with 1 FBS of DMEM (FIG. 7B). To prepare the transwell, 0.5% FBS and 2.6η 1 DMEM were placed in the lower part of the 24 well, and 8 well sized transwell was added. Then, cells (1 × 10 5 cells) were added to the prepared transwells. Then, ΙΟμΜ and 25μΜ were added to the compounds that inhibit the interaction between IkBa and AURKC, and the cells were incubated for 2 hours and 30 minutes at 37 ° C, 5% C0 2 , to move down the filter fitted with the cells. It was. After 2 hours and 30 minutes, remove the filter and the remaining cells on the filter The debris was removed using a cotton swab. The cells under the filter were fixed with 5% glutaraldehyde for 10 minutes and stained with 2% ethanol dissolved in 1% crystal violet for 20 minutes. The crystal violet was removed, observed under a microscope and the cell number in the lower part of the filter was measured. At this time, the other parts of the filter were selected and observed and averaged.
< 164> 그 결과를 도 7나와 같이 그래프를 통해 나타냈다. 유방암 세포주에 IkBa와  The results are shown through a graph as shown in FIG. 7. IkBa and Breast Cancer Cell Lines
AURKC의 상호작용을 저해하는 화합물을 처리한 경우에 대조군에 비해 이동하는 세 포의 수가 감소하는 것을 확인했다. 그리고 ΙΟ μ Μ을 넣어준 것보다 25 μ Μ의 화합물 을 넣어 주었을 때 이동하는 세포의 수가 더 감소하는 것을 확인했다.  When treated with a compound that inhibits the interaction of AURKC it was confirmed that the number of moving cells compared to the control. In addition, when 25 μM of compound was added, the number of cells to be moved decreased more than that of ΙΟ μΜ.
<165> 따라서 상기 <7-1>과 같이 IkB a와 AURKC의 상호작용을 저해하는 화합물에 의해 암세포 증식이 억제되는 것을 확인하였다.  Thus, it was confirmed that cancer cell proliferation was inhibited by a compound that inhibits the interaction between IkB a and AURKC as in <7-1>.
<166>  <166>
<167> <7~3> 연 한천 분석  <167> <7 ~ 3> Annual Agar Analysis
<168> 연 한천 분석 방법 (Sof t agar assay)으로 아가로스를 이용하여 만든 한천 배 지에 유방암 세포주 MDA-MB— 231를 배양하여 콜로니 형태를 분석하였다 (도 7다) . 먼저, 4% 한천 (agar )을 녹여 56°C 항온 수조에서 따뜻하게 유지시키고, 10% FBS DMEM을 37°C로 온도를 유지시켰다. 세포를 배양할 한천 배지의 아래층을 만들기 위 해 , 0.75% 한천과 10% FBS를 포함한 DMEM 5[ii l을 60mm 배양접시 (cul ture di sh)에 넣 고 굳을 때까지 기다렸다. 그 다음 위층을 만들기 위해, 0.36% 한천을 포함한 10% The colony morphology was analyzed by culturing the breast cancer cell line MDA-MB—231 in agar medium prepared using agarose by a Sot agar assay (FIG. 7). First, 4% agar was dissolved and kept warm in a 56 ° C constant temperature bath, and 10% FBS DMEM was kept at 37 ° C. In order to make a lower layer of agar medium to culture the cells, DMEM 5 [ii l containing 0.75% agar and 10% FBS was placed in a 60 mm culture dish and waited until it solidified. 10% including 0.36% agar to make the next upstairs
FBS DMEM 3ml에 세포를 3 X Ιθ' ce l l s 만큼 넣고, IkB a와 AURKC의 상호작용을 억제 하는 화합물 25 μ Μ을 넣어서 굳은 한천 배지 위에 넣고 굳을 때까지 기다렸다. 배 양 접시에 표기 한 뒤 37°C 배양기에서 3주간 배양한다. 그 다음에 콜로니의 수와 모양을 관찰했다ᅳ Cells were placed in 3 ml of FBS DMEM by 3 X Ιθ 'ce lls, and 25 μM of compounds inhibiting the interaction between IkB a and AURKC were placed on the solid agar medium and allowed to solidify. Mark on the culture plate and incubate for 3 weeks in a 37 ° C incubator. Then we observed the number and shape of the colonies.
<169> 그 결과 IkB a와 AURKC의 상호작용을 억제하는 화합물을 넣은 배지에서의 콜로니의 모양이 화합물을 처리하지 않은 대조군의 콜로니와 비교했을 때 더 동그 랗고 작은 것을 확인했다. 또한 화합물을 처리하지 않은 대조군보다 화합물을 처리 한 경우에 콜로니 수가 더 적은 것을 확인하였다. IkB a와 AURKC의 상호작용을 저 해하는 화합물에 의해 세포의 암화가 억제되는 것을 확인하였다.  As a result, it was confirmed that the shape of colonies in the medium containing the compound that inhibits the interaction between IkB a and AURKC was rounder and smaller than the colonies of the control group not treated with the compound. In addition, it was confirmed that the number of colonies when the compound was treated than the control group was not treated. It was confirmed that cancer cell inhibition was inhibited by a compound that inhibited the interaction between IkB a and AURKC.
<170>  <170>
【산업상 이용가능성】  Industrial Applicability
<i 7i> 본 발명은 IkBa 및 AURKC의 상호작용을 통한 암 치료제 스크리닝 방법에 관 한 것이다. 더욱 상세하게는 IkBa와 AURKC의 결합 억제제 화합물 및 이를 유효성분 으로 포함하는 암 예방 및 치료용 조성물에 관한 것이다. 본 발명은 암 치료제를 분자적 수준에서 스크리닝 할 수 있고 분자간의 상호작용을 통해 치료제의 작용을 알 수 있다는 점에서 효과적이다. <i 7i> The present invention relates to a method for screening a cancer therapeutic agent through the interaction of IkBa and AURKC. More specifically, IkBa and AURKC binding inhibitor compound and the active ingredient It relates to a composition for preventing and treating cancer. The present invention is effective in that cancer therapeutic agents can be screened at the molecular level and the action of the therapeutic agents can be known through intermolecular interactions.

Claims

【청구의 범위】 【Scope of Claim】
【청구항 1】 【Claim 1】
(a) ( i ) 이동모들, 제 1표지물질, IkBa가 순차적으로 결합된 융합 단백질 및 제 2표지물질, AURKC가 순차적으로 결합된 융합 단백질을 발현하거나 (Π) 이동모 들, 제 1표지물질, IkBa가 순차적으로 결합된 융합 단백질 및 이동모들, 제 2표지물 질, AURKC가 순차적으로 결합된 융합 단백질을 발현하는 세포를 제조하는 단계; (a) (i) mobile hairs, a first labeling material, a fusion protein to which IkBa is sequentially bound, a second labeling substance, and AURKC are sequentially bound to express a fusion protein or (Π) mobile hairs, a first labeling material, Preparing cells expressing a fusion protein to which IkBa is sequentially bound, mobile hairs, a second marker, and AURKC are sequentially bound to each other;
(b) 암 치료제 후보 물질을 첨가하는 단계; (b) adding a cancer treatment candidate;
(c) 암 치료제 후보 물질과 (a)단계의 융합 단백질 간 상호작용이 이루어지 도록 하는 단계 ; (c) allowing interaction between the cancer treatment candidate and the fusion protein of step (a);
(d) 신호물질을 처리하는 단계 ; 및 (d) processing the signal material; and
(e) 암 치료제 후보 물질과 세포 내 융합 단백질의 분포를 통해 상호작용을 확인하는 단계를 포함하며 (e) Contains the step of confirming the interaction between the cancer treatment candidate and the fusion protein within the cell;
암 치료제 후보물질이 IkBa 단백질과 AURKC 단백질의 결합을 억제할 경우 암 치료제로 판별하는 것을 특징으로 하는 암 치료제 스크리닝 방법. A cancer treatment screening method characterized in that a cancer treatment candidate is identified as a cancer treatment when it inhibits the binding of the IkBa protein and the AURKC protein.
【청구항 2】 【Claim 2】
제 1항에 있어서, 상기 이동모들은 단백질 인산화 효소 C(protein kinase C, PKC), cPKC(classical PKC; PKC— alpha, PKC-beta, PKC-ga画 a), nPKC( novel PKC; PKC-delta, P C-epsilon, P C-eta, PKC-theta)로 이루어진 군에서 선택된 것임을 특징으로 하는 방법 . According to claim 1, the mobile hairs include protein kinase C (PKC), cPKC (classical PKC; PKC— alpha, PKC-beta, PKC-ga画 a), nPKC (novel PKC; PKC-delta, A method characterized in that it is selected from the group consisting of P C-epsilon, P C-eta, PKC-theta).
【청구항 3】 【Claim 3】
게 1항에 있어서, 상기 게 1표지물질은 GFP(Green Fluorescent Protein), EGFP( Enhanced Green Fluorescent Protein) , RFPCRed Fluorescent Protein) , mRFPCMonomer ic Red Fluorescent Protein) , DsRed(Discosoma sp. red fluorescent protein) , CFP(Cyan Fluorescent Protein) , CGFP(Cyan Green Fluorescent Protein) , YFP(Yel low Fluorescent Protein) , AzG(Azami Green) , HcR(HcRed, Heteract is crispa red fluorescent protein) 및 BFP(Blue Fluorescent Protein)로 이루어진 군에서 선택된 것임을 특징으로 하는 방법. According to item 1, the crab 1 labeling substances are GFP (Green Fluorescent Protein), EGFP (Enhanced Green Fluorescent Protein), RFPCRed Fluorescent Protein), mRFPCMonomer ic Red Fluorescent Protein), DsRed (Discosoma sp. red fluorescent protein), and CFP. (Cyan Fluorescent Protein), CGFP (Cyan Green Fluorescent Protein), YFP (Yel low Fluorescent Protein), AzG (Azami Green), HcR (HcRed, Heteract is crispa red fluorescent protein), and BFP (Blue Fluorescent Protein). A method characterized as selected.
【청구항 4] [Claim 4]
제 1항에 있어서, 상기 제 2표지물질은 제 1표지물질과 서로 다르며 GFP(Green Fluorescent Protein) , EGFP( Enhanced Green Fluorescent Protein) , RFP(Red Fluorescent Protein) , mRFP(Monomer ic Red Fluorescent Protein) , DsRed(Discosoma sp. red fluorescent protein) , CFP(Cyan Fluorescent Protein) , CGFP(Cyan Green Fluorescent Protein) , YFP(Yel low Fluorescent Protein) , AzG(Azami Green) , HcR(HcRed, Heteract is cr ispa red fluorescent protein) 및 The method of claim 1, wherein the second labeling material is different from the first labeling material and is GFP (Green Fluorescent Protein), EGFP (Enhanced Green Fluorescent Protein), RFP (Red Fluorescent Protein), mRFP (Monomer ic Red Fluorescent Protein), DsRed (Discosoma sp. red fluorescent protein), CFP (Cyan Fluorescent Protein), CGFP (Cyan Green Fluorescent) Protein), YFP (Yel low Fluorescent Protein), AzG (Azami Green), HcR (HcRed, Heteract ispa red fluorescent protein) and
BFP(Blue Fluorescent Protein)로 이루어진 군에서 선택된 것임을 특징으로 하는 방법 . A method characterized in that it is selected from the group consisting of BFP (Blue Fluorescent Protein).
【청구항 5】 【Claim 5】
제 1항에 있어서, 상기 신호물질은 PMMPhorbol 12-myri state 13-acetate, Phorbol ester) , TPA(12-otetradecanoylphorbol-13-acetate) , PDBu(phorbol 12, 13-dibutyrate) , ATP(Adenosine triphosphate) , tr idecanoic acid, arachidonic acid, linoleic acid, DiC8, 130C937로 이루어진 군에서 선택된 것임 을 특징으로 하는 방법 . The method of claim 1, wherein the signaling substance is PMMPhorbol 12-myri state 13-acetate, Phorbol ester), TPA (12-otetradecanoylphorbol-13-acetate), PDBu (phorbol 12, 13-dibutyrate), ATP (Adenosine triphosphate), A method characterized in that it is selected from the group consisting of tr idecanoic acid, arachidonic acid, linoleic acid, DiC8, and 130C937.
【청구항 6】 【Claim 6】
게 1항에 있어서, 상기 신호물질의 처리는 PMMPhorbol 12-myri state 13- acetate, Phorbol ester)를 50nM 내지 5uM의 농도로 처리하는 것임을 특징으로 하 는 방법 . The method according to item 1, wherein the treatment of the signal material is treated with PMMPhorbol 12-myri state 13-acetate, Phorbol ester) at a concentration of 50nM to 5uM.
【청구항 7】 【Claim 7】
제 1항의 암 치료제 스크리닝 방법을 통해 스크리닝된 화합물. A compound screened through the cancer treatment drug screening method of claim 1.
【청구항 8] [Claim 8]
제 7항에 있어서, 상기 화합물은 하기 화학식 1로 표시되는 화합물. The compound according to claim 7, wherein the compound is represented by the following formula (1).
[화학식 1] [Formula 1]
Figure imgf000023_0001
Figure imgf000023_0001
【청구항 9】 제 7항에 있어서, 상기 화합물은 IkBa 단백질과 AURKC 단백질 결합을 억제하 는 것을 특징으로 하는 화합물. 【Claim 9】 The compound according to claim 7, wherein the compound inhibits the binding of IkBa protein and AURKC protein.
【청구항 10】 【Claim 10】
게 7항에 있어서, 상기 화합물이 IkBa 단백질과 AURKC 단백질의 결합을 억제 하여 암 예방 및 효과를 보이는 것을 특징으로 하는 화합물. The compound according to item 7, wherein the compound inhibits the binding of IkBa protein and AURKC protein, thereby preventing cancer and showing effects.
【청구항 11】 【Claim 11】
IKBa와 AURKC간의 상호작용 저해제를 유효성분으로 포함하는 암 예방 및 치 료용 조성물. A composition for preventing and treating cancer comprising an inhibitor of the interaction between IKBa and AURKC as an active ingredient.
【청구항 12】 【Claim 12】
제 11항에 있어서, 상기 저해제는 하기 화학식 1로 표시되는 화합물인 것을 특징으로 하는 암 예방 및 치료용 조성물. The composition for preventing and treating cancer according to claim 11, wherein the inhibitor is a compound represented by the following formula (1).
[화학식 [Chemical formula
Figure imgf000024_0001
Figure imgf000024_0001
【청구항 13] [Claim 13]
제 11항에 있어서, 상기 암은 위암, 폐암, 간암, 대장암, 소장암, 췌장암, 뇌 암 뼈암, 혹색종, 유방암, 경화성선증, 자궁암, 자궁경부암, 두경부암, 식도암, 갑상선암, 부갑상선암, 신장암, 육종, 전립선암, 요도암, 방광암, 혈액암, 림프종, 건선 또는 섬유선종으로 이루어진 군으로부터 선택되는 것을 특징으로 하는 조성 물. The method of claim 11, wherein the cancer is stomach cancer, lung cancer, liver cancer, colon cancer, small intestine cancer, pancreatic cancer, brain cancer, bone cancer, xenograft, breast cancer, sclerosing gonadosis, uterine cancer, cervical cancer, head and neck cancer, esophageal cancer, thyroid cancer, parathyroid cancer, A composition selected from the group consisting of kidney cancer, sarcoma, prostate cancer, urethral cancer, bladder cancer, blood cancer, lymphoma, psoriasis, or fibroadenoma.
【청구항 14】 【Claim 14】
IKBa와 AURKC간의 상호작용 저해제의 암 예방 및 치료제 제조를 위한 용도. Use of inhibitors of the interaction between IKBa and AURKC for the manufacture of cancer prevention and treatment.
【청구항 15】 【Claim 15】
제 14항에 있어서, 상기 저해제는 하기 화학식 1로 표시되는 화합물인 것을 특징으로 하는 암 예방 및 치료제 제조를 위한 용도. The method of claim 14, wherein the inhibitor is a compound represented by the following formula (1): Use for manufacturing characterized cancer prevention and treatment.
[화학식 1] [Formula 1]
Figure imgf000025_0001
Figure imgf000025_0001
【청구항 16] [Claim 16]
제 15항에 있어서, 상기 암은 위암 폐암, 간암, 대장암, 소장암, 췌장암, 뇌 암 뻐암 혹색종, 유방암, 경화성선증, 자궁암, 자궁경부암, 두경부암, 식도암, 갑상선암, 부갑상선암 신장암, 육종, 전립선암, 요도암, 방광암, 혈액암, 림프종, 건선 또는 섬유선종으로 이루어진 군으로부터 선택되는 것을 특징으로 하는 암 예 방 및 치료제 제조를 위한 용도. The method of claim 15, wherein the cancers include stomach cancer, lung cancer, liver cancer, colon cancer, small intestine cancer, pancreatic cancer, brain cancer, leukoma, breast cancer, sclerosing gonadosis, uterine cancer, cervical cancer, head and neck cancer, esophageal cancer, thyroid cancer, parathyroid cancer, and kidney cancer. Use for the manufacture of cancer prevention and treatment, characterized in that it is selected from the group consisting of sarcoma, prostate cancer, urethral cancer, bladder cancer, hematological cancer, lymphoma, psoriasis or fibroadenoma.
【청구항 17】 【Claim 17】
IKBa와 AURKC간의 상호작용 저해제를 이를 필요로 하는 개체에 유효량으로 투여하는 것을 특징으로 하는 암 예방 및 치료 방법. A cancer prevention and treatment method comprising administering an effective amount of an inhibitor of the interaction between IKBa and AURKC to an individual in need thereof.
【청구항 18】 【Claim 18】
제 17항에 있어서, 상기 저해제는 하기 화학식 1로 표시되는 화합물인 것을 특징으로 하는 암 예방 및 치료 방법. The method of claim 17, wherein the inhibitor is a compound represented by the following formula (1).
[화학식 1] [Formula 1]
Figure imgf000025_0002
Figure imgf000025_0002
【청구항 19】 【Claim 19】
제 18항에 있어서, 상기 암은 위암, 폐암, 간암, 대장암, 소장암, 췌장암, 뇌 암, 뼈암, 흑색종, 유방암, 경화성선증, 자궁암, 자궁경부암, 두경부암, 식도암, 갑상선암, 부갑상선암, 신장암, 육종, 전립선암, 요도암, 방광암, 혈액암, 림프종, 건선 또는 섬유선종으로 이루어진 군으로부터 선택되는 것을 특징으로 하는 암 예 방 및 치료 방법. The method of claim 18, wherein the cancer is stomach cancer, lung cancer, liver cancer, colon cancer, small intestine cancer, pancreatic cancer, brain cancer, bone cancer, melanoma, breast cancer, sclerosing gonadosis, uterine cancer, cervical cancer, head and neck cancer, esophageal cancer, thyroid cancer, and parathyroid cancer. , Kidney cancer, sarcoma, prostate cancer, urethral cancer, bladder cancer, blood cancer, lymphoma, A method for preventing and treating cancer, characterized in that the cancer is selected from the group consisting of psoriasis or fibroadenoma.
PCT/KR2015/007751 2014-07-24 2015-07-24 Method for screening therapeutic agent for cancer using interaction between ikba and aurkc WO2016013909A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20140094260 2014-07-24
KR10-2014-0094260 2014-07-24

Publications (1)

Publication Number Publication Date
WO2016013909A1 true WO2016013909A1 (en) 2016-01-28

Family

ID=55163364

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2015/007751 WO2016013909A1 (en) 2014-07-24 2015-07-24 Method for screening therapeutic agent for cancer using interaction between ikba and aurkc

Country Status (2)

Country Link
KR (1) KR101738680B1 (en)
WO (1) WO2016013909A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20070048660A (en) * 2004-07-20 2007-05-09 씨지케이 주식회사 System for detecting molecular interactions
KR100948767B1 (en) * 2008-11-12 2010-03-23 한국기초과학지원연구원 Method for detecting interactions between two and more biological macromolecules

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20070048660A (en) * 2004-07-20 2007-05-09 씨지케이 주식회사 System for detecting molecular interactions
KR100948767B1 (en) * 2008-11-12 2010-03-23 한국기초과학지원연구원 Method for detecting interactions between two and more biological macromolecules

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BERGQVIST, S. ET AL.: "The IkBa/NF-kB complex has two hot spots, one at either end of the interface", PROTEIN SCIENCE, vol. 17, 2008, pages 2051 - 2058 *
DATABASE PubChem 4 June 2005 (2005-06-04), Database accession no. 654580 *
KATSHA, A. ET AL.: "Aurora kinase A promotes inflammation and tumorigenesis in mice and human gastric neoplasia", GASTROENTEROLOGY, vol. 145, no. 6, 2013, pages 1312 - 1322 and 1322.e1-1322.e8 *

Also Published As

Publication number Publication date
KR101738680B1 (en) 2017-05-22
KR20160012957A (en) 2016-02-03

Similar Documents

Publication Publication Date Title
US8349798B2 (en) Caveolin peptides and their use as therapeutics
Pardo et al. Role of voltage-gated potassium channels in cancer
US20200038471A1 (en) Small molecule cancer treatments that cause necrosis in cancer cells but do not affect normal cells
Dráberová et al. Differential expression of human γ‐tubulin isotypes during neuronal development and oxidative stress points to a γ‐tubulin‐2 prosurvival function
US8173360B2 (en) Cell death inhibitor
Lu et al. Discovery of a subtype-selective, covalent inhibitor against palmitoylation pocket of TEAD3
CN111658644B (en) Small-molecule STAT3 inhibitor WZ-2-033 and application thereof in preparation of medicines for treating breast cancer and gastric cancer
EP3605102B1 (en) Use of a composition comprising an aimp2-dx2 protein for diagnosing lung cancer
US20140248254A1 (en) The use of ache as nuclease
CN111484492B (en) Substituted pyridino-imidazole compound and application thereof in preparation of medicine for treating malignant tumor diseases
WO2008046107A2 (en) Novel inhibitors of the egfr kinase targeting the asymmetric activating dimer interface
Pang et al. Sodium cantharidate induces Apoptosis in breast cancer cells by regulating energy metabolism via the protein phosphatase 5-p53 axis
CN113730409B (en) Application of quinacrine in preparation of medicine for treating pulmonary fibrosis
WO2016013909A1 (en) Method for screening therapeutic agent for cancer using interaction between ikba and aurkc
Ren et al. SIPAR negatively regulates STAT3 signaling and inhibits progression of melanoma
CN110960677B (en) Use of SAGE1 inhibitor in preparation of medicine or kit
KR101749670B1 (en) Method for screening anti-cancer agents comprising detecting interactions of IkBα and RPS6KA2
CN109453384A (en) Application of the NOD1 in the product that preparation inhibits tumour SRC signal path
US20090111733A1 (en) Par-4 related methods and compositions
KR101704772B1 (en) IkB RPS6KA2 Pharmaceutical composition comprising small-molecule inhibitors of IkBand RPS6KA2 interaction for preventing and treating cancer
US20150174191A1 (en) Pharmaceutical composition for treating hyperproliferative diseases
CN116135873A (en) Kit, P53-targeting polypeptide and application thereof in preparation of medicines for treating cancers
CN115925881A (en) Polypeptide for inhibiting SET protein nucleoplasm shuttle and application thereof
US20100035825A1 (en) Treatment Of Disease By Inducing Cell Apoptosis
CN116655807A (en) Specific targeting protein antagonistic polypeptide, preparation method and application thereof

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: 15824295

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: 15824295

Country of ref document: EP

Kind code of ref document: A1