WO2012115437A2 - Pharmaceutical composition for cancer treatment or metastasis inhibition containing expression or activation inhibitors of map7d2 protein, novel cancer therapeutic target - Google Patents

Pharmaceutical composition for cancer treatment or metastasis inhibition containing expression or activation inhibitors of map7d2 protein, novel cancer therapeutic target Download PDF

Info

Publication number
WO2012115437A2
WO2012115437A2 PCT/KR2012/001316 KR2012001316W WO2012115437A2 WO 2012115437 A2 WO2012115437 A2 WO 2012115437A2 KR 2012001316 W KR2012001316 W KR 2012001316W WO 2012115437 A2 WO2012115437 A2 WO 2012115437A2
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
map7d2
protein
expression
metastasis
Prior art date
Application number
PCT/KR2012/001316
Other languages
French (fr)
Korean (ko)
Other versions
WO2012115437A3 (en
Inventor
권기선
김선영
최소영
도소희
박성섭
이광표
Original Assignee
한국생명공학연구원
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from KR1020120016917A external-priority patent/KR101376599B1/en
Application filed by 한국생명공학연구원 filed Critical 한국생명공학연구원
Publication of WO2012115437A2 publication Critical patent/WO2012115437A2/en
Publication of WO2012115437A3 publication Critical patent/WO2012115437A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.

Definitions

  • compositions for treating cancer or inhibiting cancer metastasis comprising inhibitors of the expression or activity of the MAP 7 D 2 protein as a novel cancer treatment target
  • the present invention relates to a pharmaceutical composition for treating cancer or inhibiting cancer metastasis, comprising an inhibitor of expression or activity of MAP7D2 protein, which is a novel cancer growth or cancer metastasis therapeutic target.
  • Cancer Progression is Activation of Cancer-Induced Genes (oncogene) and Tumor Suppressor Genes
  • Cancer is a disease that is very heterogeneous in its cause and progression. Morphological and pathological Although similar in nature, they progress in significantly different forms at the molecular level. Recent clinical experience supports this: ERBB2 and EGFR, the targets of successful clinical drugs such as Herceptin and Zephytinib, are mutated or overexpressed in only about 20-30% of all cancer patients. have.
  • MAP7D2 (MAP7 domain containing 2) is another protein named FLJ 14503, MGC104944, RP11-393H10.2, which is a type of microtubule-associated protein 7 domain.
  • the present inventors have conducted research to discover targeted cancer treatment targets, and have established a large-scale gene expression database of cancer tissues through original data mining techniques.
  • the present invention was completed by identifying MAP7D2 that is overexpressed in lung cancer, colorectal cancer, ovarian cancer, gastric cancer, and liver cancer and remarkably inhibiting the growth, infiltration and metastasis of cancer cells by inhibition of MAP7D2 expression.
  • An object of the present invention is the MAP7D2 protein, which is a novel cancer growth or cancer metastasis therapeutic target. It is to provide a pharmaceutical composition for treating cancer or inhibiting cancer metastasis, containing an inhibitor of expression or activity as an active ingredient.
  • Another object of the present invention is to provide a method for screening cancer growth, invasion or metastasis inhibitors using MAP7D2.
  • Another object of the present invention is to provide a method for monitoring or diagnosing cancer in a subject using MAP7D2.
  • the present invention provides a pharmaceutical composition for treating cancer or inhibiting cancer metastasis, containing an inhibitor of the expression or activity of MAP7D2 (MAP7 domain containing 2) protein as an active ingredient.
  • MAP7D2 MAP7 domain containing 2
  • the present invention provides a method for screening a candidate substance for treating cancer or inhibiting cancer metastasis, comprising the following steps:
  • ⁇ 2i> 2 measuring the expression level of MAP7D2 protein in the cell line
  • the present invention provides a method for screening a candidate substance for cancer treatment or cancer metastasis suppression comprising the following steps:
  • the present invention provides a method for monitoring or diagnosing cancer using MAP7D2 protein:
  • the present invention also provides a method for preventing cancer, comprising administering to a subject a pharmaceutically effective amount of an MAP7D2 protein expression or activity inhibitor.
  • the present invention also provides a method for treating cancer or inhibiting cancer metastasis, comprising administering a pharmaceutically effective amount of an MAP7D2 protein expression or active inhibitor to a subject with cancer. Provide the law.
  • the present invention comprises the step of measuring the expression level of MAP7D2 in cancer cells using any one or more of the nucleic acid complementary to the antibody or gene specifically binding to MAP7D2, the diagnosis of cancer, confirming the treatment result Or provide a method of assessing prognosis.
  • the present invention also provides a kit for diagnosing cancer, comprising any one or more of nucleic acids complementary to antibodies or / genes that specifically bind to MAP7D2.
  • the present invention also provides an inhibitor of MAP7D2 protein expression or activity for use in the prophylaxis or treatment of cancer or the pharmaceutical composition for inhibiting cancer metastasis.
  • the present invention provides nucleic acids complementary to antibodies or genes that specifically bind to MAP7D2 for use in cancer diagnostic kits.
  • a large-scale gene expression database of human cancer tissue was constructed to discover a novel targeting anticancer candidate target MAP7D2 through data mining techniques, and the MAP7D2 is a kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and the like. It is particularly remarkably overexpressed in tissue or cells of liver cancer, and inhibition of MAP7D2 significantly inhibits the growth, migration, invasion and metastasis of cancer cells and induces apoptosis, thereby inhibiting the expression or activity inhibitor of MAP7D2 protein.
  • a pharmaceutical composition containing as an ingredient may be usefully used as an active ingredient of a pharmaceutical composition for treating cancer or inhibiting cancer metastasis.
  • FIG. 1 is a diagram showing the results of analyzing the expression pattern of the MAP7D2 gene in clinical tissues with the microarray platform U133plus2.
  • Figure 2 is a diagram showing the results of analyzing the expression of the MAP7D2 gene in the cancer cell line microarray platform U133plus2.
  • FIG. 3 illustrates the use of GAPDH as an internal control for standardization of RNA amounts.
  • Figure shows the expression of MAP7D2 in colorectal cancer cell line.
  • Figure shows the expression of MAP7D2 in liver cancer cell line.
  • FIG. 5 uses GAPDH as an internal control for standardization of RNA amounts.
  • FIG. 6 shows the results of RT-PCR to compare the expression of MAP7D2 in normal and lung cancer tissues of lung cancer patients:
  • -Act in beta actin (control); N: normal tissue; And T: cancer tissue.
  • FIG. 7 shows the results of RT-PCR to compare the expression patterns of MAP7D2 in normal and liver cancer tissues of liver cancer patients:
  • ⁇ -act in beta actin (control); N: normal tissue; And T: cancer tissue.
  • FIG. 8 shows the renal cancer cell line A498 knocked down by the expression of MAP7D2 by siRNA.
  • MAP7D2 gene was confirmed by RT-PCR.
  • Figure 9 shows the results of confirming the inhibitory effect of cell growth in the renal cancer cell line A498 knocked down expression of MAP7D2 by siRNA.
  • FIG. 10 is a diagram confirming the expression of the MAP7D2 gene by RT-PCR in lung cancer cell line NCI-H1703 in which expression of MAP7D2 is knocked down by shRNA.
  • FIG. 11 is a view of morphology of lung cancer cell line NCI-H1703 observed under a microscope (X100) 5 days after knocking down MAP7D2 expression (shCtrl: Nontarget shControl RNA).
  • FIG. 12 is a graph showing changes in cell growth due to knockdown of the MAP7D2 gene in lung cancer cell line NCI-H1703 (bar: standard deviation).
  • FIG. 13 is a diagram showing the change in cell migration by knockdown of MAP7D2 gene in lung cancer cell line NCI-H1703:
  • A A picture of a microscopic observation of a cancer cell migration experiment using a transwell plate of NCI-H1703 cells transduced with shRNA (X100 magnification);
  • FIG. 14 is a diagram showing the change of cell invasion by knockdown of MAP7D2 gene in lung cancer cell line NCI-H1703:
  • the present invention provides a pharmaceutical composition for treating cancer or inhibiting cancer metastasis, comprising an MAP7D2 (MAP7 domain containing 2) protein expression or activity inhibitor as an active ingredient.
  • MAP7D2 MAP7 domain containing 2
  • the MAP7D2 protein preferably has an amino acid sequence as set forth in SEQ ID NO: 1, but is not limited thereto.
  • the cancer is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer, and more preferably lung cancer or kidney cancer, but is not limited thereto.
  • the inhibitor of expression of the MAP7D2 protein is from the group consisting of antisense nucleotides, small hairpin RNAs, small interfering RNAs, and ribozymes that complementarily bind to the mRNA of the MAP7D2 gene.
  • the MAP7D2 protein inhibitor is any one selected from the group consisting of compounds, peptides, peptide mimetics, substrate analogs, aptamers, and antibodies that complementarily bind to MAP7D2 proteins. Is preferably, but is not limited thereto.
  • the siRNA is composed of a 15 to 30 mer sense sequence selected from the base sequence of the mRNA of the gene encoding a human MAP7D2 protein and an antisense sequence complementary to the sense sequence, wherein
  • the sense sequence is not particularly limited thereto, but is preferably composed of 25 bases, but is not limited thereto, and more preferably, SEQ ID NO: 4 or 5, but is not limited thereto.
  • the antisense nucleotide binds to (generalizes) the complementary sequencing of DNA, immature -mRNA, or mature mRNA, as defined by the Watson-click base pair, thereby disrupting the flow of genetic information as a protein in DNA. will be.
  • the nature of antisense nucleotides that are specific for the target sequence makes them exceptionally versatile. Since antisense nucleotides are long chains of monomeric units they can be easily synthesized for the target RNA sequence. Many recent studies have demonstrated the utility of antisense nucleotides as biochemical means for studying target proteins (Rothenberg et al., J. Natl. Cancer Inst., 81: 1539-1544, 1999).
  • antisense nucleotides can be considered as a new type of inhibitor because of recent advances in oligonucleotide chemistry and in the synthesis of nucleotides that exhibit improved cell adsorption, target binding affinity and nuclease resistance.
  • Peptide Minetics binds to the binding domain of the MAP7D2 protein. It is to inhibit the activity of the inhibitory MAP7D2 protein.
  • Peptide or non-systematic seuneun peptide may be a non-peptide, S p i coupling (Benkirane, N., et al J. Biol Chem, 271:... 33218-33224, 1996) and engaged by the same, a non-peptide bond Can be composed of amino acids.
  • a "conformational ly constrained" peptide, between cyclic mimetics, at least one exocyclic domain, binding moiety (binding amino acid) and active site It may be a click mimetics.
  • Peptide mimetics are structured similar to the secondary structural properties of the MAP7D2 protein and are either antibodies (Park, BW et al. Nat Biotechnol 18, 194-198, 2000) or water soluble receptors (Takasaki, W. et al. Nat Biotechnol 15, 1266). -1270, 1997), which can mimic the inhibitory properties of large molecules, and may be novel small molecules that work with the same effect as natural antagonists (Wrighton, NC et al. Nat Biotechnol 15, 1261-1265, 1997).
  • the aptamer is a single chain DNA or RNA molecule
  • Oligomers that bind with specific affinity and selectivity to specific chemical or biological molecules by evolutionary methods using oligonucleotide libraries called systemat ic evo 1 on i 1 and g of s by exponential enrichment (SELEX) Can be obtained separately (C. Tuerand L. Gold, Science 249, 505-510, 2005; AD Ellington and JW Szostak, Nature 346, 818-822, 1990; M. Famulok, et. Al., Acc. Chem. Res. 33, 591-599, 2000; DS Wilson and Szostak, Annu. Rev. Biochem. 68, 611-647, 1999).
  • Aptamers can specifically bind to targets and modulate the activity of the targets, such as by blocking the ability of the targets to function through binding.
  • the antibody may specifically and directly bind to MAP7D2 to effectively inhibit MAP7D2 activity.
  • MAP7D2 it is preferable to use a polyclonal antibody or a monoclonal antibody.
  • the antibody that specifically binds to MAP7D2 may be prepared by known methods known to those skilled in the art, and commercially known MAP7D2 antibodies may be purchased and used.
  • the antibody can be prepared by injecting the immunogen MAP7D2 protein into an external host according to conventional methods known to those skilled in the art. External hosts include mammals such as mice, rats, sheep and rabbits. Immunogens are injected intramuscularly, intraperitoneally or subcutaneously, and can generally be administered in combination with an adjuvant to increase antigenicity. Blood is drawn periodically from an external host to determine the titer and antigen Antibodies can be isolated by collecting specific serum.
  • the composition may have inhibitory activity of cancer proliferation, migration, invasion or metastasis.
  • the present inventors describe the gene expression of NCBI.
  • the present inventors selected genes that are specifically overexpressed in specific cancer tissues, and selected cell lines in which the genes were expressed higher than a certain level. At this time, the gene was selected as a novel target gene with little reported on cancer. In this process, MAP7D2 was selected as a candidate target gene for targeted chemotherapy, and the expression of MAP7D2 was analyzed by microarray analysis. As a result, it was found that the renal cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer were expressed higher than normal tissues or other cancer tissues (see FIG. 1).
  • the present inventors transduced short hairpin RNA (shRNA) or small interfering RNA (LNA) genes into lung or kidney cancer cell lines, which show high expression levels of MAP7D2. Expression was inhibited and growth, migration, infiltration and metastasis of cancer cells of the cell line were observed. As a result, it was confirmed that MAP7D2 gene expression was inhibited by introduction of shRNA or siRNA into cancer cells (see FIGS. 8 and 10), and proliferation of cancer cells was markedly reduced and cell death was increased. 9 and 11 to 12, it was observed that the migration and infiltration of cancer cells are inhibited (see FIGS. 13 and 14). Therefore, it was found that inhibition of expression or activity of MAP7D2 inhibits the proliferation of cancer cells such as lung cancer or kidney cancer and plays a decisive role in invasion and metastasis.
  • shRNA short hairpin RNA
  • LNA small interfering RNA
  • MAP7D2 is overexpressed in the tissues of renal cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer and the cell lines of the cancer, and the inhibition of the expression of the gene inhibits the growth, migration and invasion of cancer cells.
  • Inhibitors of the expression or activity of MAP7D2 protein may be usefully used as an active ingredient of a pharmaceutical composition for treating cancer or inhibiting cancer metastasis.
  • the pharmaceutical composition of the present invention may be prepared by including one or more pharmaceutically acceptable carriers in addition to the above-described active ingredients for administration.
  • Pharmaceutically acceptable carriers include saline, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol, liposomes and one or more of these components in combination.
  • Other conventional additives such as antioxidants, buffers, bacteriostatics, can be added.
  • diluents, dispersants, surfactants, binders and lubricants can be added in addition to formulate into injectable formulations, such as aqueous solutions, suspensions, emulsions, pills, capsules, granules or tablets, which will act specifically on target organs.
  • injectable formulations such as aqueous solutions, suspensions, emulsions, pills, capsules, granules or tablets, which will act specifically on target organs.
  • Target organ specific antibodies or other ligands can be used in combination with the carrier so that they can be used.
  • Remington's Pharmaceutical Science Remington's Pharmaceutical Science (Recent), Mack Publishing Company, Easton PA, it is preferred according to each disease or component. It may be formulated.
  • the nucleotides or nucleic acids used in the present invention may be prepared for the purpose of oral, topical, parenteral, intranasal, intravenous, intramuscular, subcutaneous, intraocular, transdermal and the like.
  • the nucleic acid or vector is used in injectable form.
  • the area to be treated may be mixed with any pharmaceutically acceptable media for injectable compositions for direct infusion.
  • the pharmaceutical compositions of the present invention may in particular comprise isotonic sterile solutions or lyophilized compositions which allow the composition of injectable solutions upon the addition of dry, in particular sterile water or appropriate physiological saline.
  • nucleic acid used in tumors of the tumor is advantageous because it allows the treatment efficiency to be focused on the infected tissue.
  • the dosage of nucleic acid used can be adjusted by various parameters, in particular by gene, vector, mode of administration used, disease in question or alternatively required duration of treatment. In addition, the range varies depending on the patient's weight, age, sex, health status, diet, administration time, administration method, excretion rate and the severity of the disease.
  • the daily dose is about 0.0001 to 100 /, preferably 0.001 to 10 /, preferably administered once to several times a day.
  • the present invention provides a method for screening a candidate substance for cancer treatment or cancer metastasis inhibition using MAP7D2 protein.
  • the cancer is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer, and more preferably lung cancer or kidney cancer, but is not limited thereto.
  • 3) preferably, but not limited to, selecting a test substance whose expression level of the MAP7D2 protein is reduced compared to a control group not treated with the test substance.
  • the cell line of step 1) is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer cell lines, and the lung cancer or kidney cancer cell lines. More preferably, it is not limited to this.
  • the expression level of the protein of step 2) is immunoprecipitation method.
  • the activity level of the MAP7D2 protein is compared to the control group not treated with the test substance. It is preferable to include the step of screening a reduced test substance compared to but is not limited thereto.
  • the activity level of the protein of step 2) is SDS-PAGE ,.
  • Method, enzyme immunoassay (ELISA), mass spectrometry and protein chip is preferably measured by any one selected from the group consisting of, but not limited to.
  • a novel targeting anticancer candidate target MAP7D2 was selected through a data mining technique based on a large gene expression database of human cancer tissue constructed in the present invention. Cancer cells that are overexpressed in tissues and cells of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer, and inhibited MAP7D2 inhibit growth, migration, invasion and metastasis, and induce apoptosis. Since it has been determined to play a crucial role in the method, the method of measuring the expression or activity of MAP7D2 protein can be usefully used for screening a substance for inhibiting cancer treatment or cancer metastasis.
  • the present invention provides a method for monitoring or diagnosing cancer using MAP7D2 protein.
  • MAP7D2 in step 1) preferably has an amino acid sequence set forth in SEQ ID NO: 1, but is not limited thereto.
  • the cancer of step 2) is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, stomach cancer and liver cancer, and more preferably lung cancer or kidney cancer. Or not limited thereto.
  • the present invention provides a method for preventing cancer, comprising administering to a subject a pharmaceutically effective amount of an MAP7D2 protein expression or activity inhibitor.
  • the present invention provides a method for treating cancer or inhibiting cancer metastasis, comprising administering a pharmaceutically effective amount of an MAP7D2 protein expression or active inhibitor to a subject with cancer. Provide the law.
  • the pharmaceutically effective amount is 0.0001 to 100 mg / kg, preferably
  • the dosage can be varied according to the body of the specific patient, age, sex, health status, 'diet, administration time, administration method, clearance, such as the severity of the disease.
  • the MAP7D2 protein expression or activity inhibitor may be administered orally or orally during clinical administration and intraperitoneal, rectal, subcutaneous, intravenous, intramuscular, intrauterine It can be administered by injection, cerebrovascular injection, or intrathoracic injection, and can be used in the form of general pharmaceutical preparations.
  • the cancer is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer, and more preferably lung cancer or kidney cancer, but is not limited thereto.
  • the subject is a vertebrate and preferably a mammal, more preferably an experimental animal such as a rat, rabbit, guinea pig, hamster, dog, or cat, and most preferably anthropoid animals such as chimpanzees or gorillas. to be.
  • an experimental animal such as a rat, rabbit, guinea pig, hamster, dog, or cat
  • anthropoid animals such as chimpanzees or gorillas. to be.
  • the present inventors constructed a large-scale gene expression database of human cancer tissue to find a novel targeted anticancer candidate target through data mining techniques.
  • MAP7D2 is overexpressed in tissues and cells of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer, and inhibition of MAP7D2 inhibits the growth, migration, invasion and metastasis of lung cancer or kidney cancer cells and apoptosis.
  • This induction confirmed the possibility of MAP7D2 as a target of a novel anticancer agent, and therefore inhibitors of the expression or activity of MAP7D2 protein can be usefully used for the treatment of cancer or inhibition of cancer metastasis.
  • the present invention includes the steps of measuring the expression level of MAP7D2 in cancer cells using at least one of an antibody specifically binding to MAP7D2 or a nucleic acid complementary to the gene, the result of the diagnosis and treatment of cancer It provides a method of evaluating or evaluating prognosis.
  • the cancer is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer, and more preferably lung cancer or kidney cancer, but is not limited thereto.
  • detection of expression of MAP7D2 elevated above normal It means that the patient has cancer.
  • detection of normal expression of MAP7D2 in diagnostic samples of individuals treated or receiving cancer means that the cancer treatment is successful, and detection of elevated MAP7D2 above normal in the diagnostic samples indicates that treatment should be continued. do.
  • detection of normal expression of MAP7D2 in a diagnostic sample of a subject with cancer means a good prognosis, and detection of MAP7D2 elevated above normal in the diagnostic sample means a poor prognosis.
  • the present invention provides a kit for diagnosing cancer comprising any one or more of nucleic acids complementary to an antibody or gene that specifically binds to MAP7D2.
  • the cancer diagnostic kit may further include one or more substances and reaction product detection reagents and instructions for reacting with MAP7D2.
  • the one or more substances that react with MAP7D2 may be RNA or DNA complementary to the RNA or DNA of MAP7D2, and an antibody that binds to the MAP7D2 protein and the reagent for detecting the reaction product may be a nucleic acid or protein label and a coloring reagent. have.
  • a novel targeting anticancer candidate target MAP7D2 was selected through a data mining technique based on a large gene expression database of human cancer tissue constructed in the present invention. Cancer cells that are overexpressed in tissues and cells of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer, and inhibited MAP7D2 inhibit growth growth, invasion and metastasis, and induce apoptosis, thereby inducing MAP7D2 to cancer cells. It was confirmed to play a decisive role. Therefore, the expression level of MAP7D2 can be monitored and cancer can be diagnosed.
  • the present invention is for use in the prevention or treatment of cancer or to inhibit cancer metastasis
  • Inhibitors of MAP7D2 protein expression or activity are provided.
  • the MAP7D2 protein preferably has an amino acid sequence as set forth in SEQ ID NO: 1, but is not limited thereto.
  • the inhibitor of expression of the MAP7D2 protein may be any one selected from the group consisting of antisense nucleotides, short interfering RNAs, and short hairpin RNAs, which complementarily bind to mRNA of a gene. Preferred but not limited to.
  • the inhibitor of activity of the MAP7D2 protein binds complementarily to the MAP7D2 protein. It is preferably one selected from the group consisting of a compound, a peptide, a peptide mimetics, an aptamer and an antibody, but is not limited thereto.
  • the cancer is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer, and more preferably lung cancer or kidney cancer, but is not limited thereto.
  • the inventors constructed a large-scale gene expression database of human cancer tissue to discover novel targeted anticancer candidate targets through data mining techniques.
  • MAP7D2 is overexpressed in tissues and cells of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer, and inhibition of MAP7D2 inhibits the growth, migration, infiltration and metastasis of lung cancer or kidney cancer cells, and apoptosis. Because of this induction, inhibitors of the expression or activity of the MAP7D2 protein can be usefully used for preventing or treating cancer or inhibiting cancer metastasis.
  • the present invention provides a nucleic acid complementary to an antibody or gene that specifically binds to MAP7D2 for use in a kit for cancer diagnosis.
  • the cancer diagnostic kit may further include one or more substances and reaction products for detecting the reaction with MAP7D2 and instructions thereof.
  • the one or more substances that react with MAP7D2 may be RNA or DNA complementary to the RNA or DNA of MAP7D2, and an antibody that binds to MAP7D2 protein
  • the reagent for detecting the reaction product may be a nucleic acid or protein label and a coloring reagent.
  • a novel targeting anticancer candidate target MAP7D2 was selected through a data mining technique based on a large gene expression database of human cancer tissue constructed in the present invention. Cancer cells that are overexpressed in tissues and cells of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer, and inhibit MAP7D2 growth, migration, invasion and metastasis, and induce apoptosis, thereby inhibiting MAP7D2 from cancer cells. It was confirmed that it plays a decisive role in. Thus, nucleic acids complementary to antibodies or / genes that specifically bind to MAP7D2 can be usefully used in cancer diagnostic kits.
  • the database contains gene expression data for more than 22,000 normal and cancerous tissues from almost all tissues, as shown in Table 2 below.
  • COPA Cancer Outlier Profile Analysis
  • Example ⁇ 1-1> C0PA analysis was applied in a method different from the conventional approach to discover therapeutic targets from a cancer gene expression database constructed independently (Tomlins et al. 2005). .
  • the targeted anticancer target genes of therapeutic agents effectively used in the clinic are overexpressed by gene mutation or gene amplification in only a part of the patients.
  • ERBB2 a target of Herceptin
  • EGFR a target of Iressa or Celuximab
  • Overexpression by gene mutation or gene amplification is observed only in about 10-20% of the population.
  • the outlier analysis method compares the average of the top 10-20% of the patient group with the average of the normal person, rather than comparing the average of the patient group and the normal person to find genes with different expressions as in the conventional t-test. Since it is effective, this cancer outlier profile analysis (COPA) was performed.
  • COPA cancer outlier profile analysis
  • the C0PA assay was applied to select genes that could be targeted for effective targeting therapy in each cancer from among drug-targeting target receptors, kinases, transporters, and channel proteins.
  • Therapeutic drug targets currently in clinical trials or mainly on the market are mostly cell surface molecules such as receptors, channel transporters, kinases and cell adhesion molecules (CAMs).
  • Candidate target genes were identified.
  • Candidate target genes were identified based on the following criteria. i) Gene expression database mining for clinical tissues formed a pool of candidate target genes that are expressed more specifically in specific cancer tissues than in normal tissues. ii) Select genes that are specifically overexpressed in several cancers to become target genes in the gene pool formed.
  • MAP7D2 As a result, as shown in FIG. 1, as a result of confirming the expression pattern of MAP7D2 in the clinical tissues of cancer patients, kidney cancer, lung cancer, colon cancer, liver cancer, and ovary It was observed that the expression of MAP7D2 was increased in cancer tissues such as cancer and stomach cancer compared to normal tissues, and thus, MAP7D27 ⁇ could be an effective therapeutic target for the treatment of cancer (FIG. 1). ).
  • the expression pattern in the clinical tissue of MAP7D2 is derived from kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer in accordance with the results confirmed in the cancer tissue of Figure 1 It can be seen that they are relatively high in one cancer cell line (FIG. 2).
  • MAP7D2 is best expressed for each target cancer selected based on the gene expression database for each cell line, in order to select cancer cell lines to be used in the experiment. The cell lines were confirmed, which are shown in Table 3 below.
  • RT-PCR was performed to confirm the expression patterns of MAP7D2, respectively.
  • 11 types of cancer cell lines (4 types of colorectal cancer, 4 types of lung cancer, and 3 types of liver cancer) were used in the easy-BLUE TM Total RNA Extraction kit (Sol Gent, Cat #: 17061).
  • cDNA was synthesized using a DiaStar RT kit (SolGent, Cat #: DR13-R10K) with each total RNA 2 / g. cDNA was distilled with sterile distilled water to prepare 2% cDNA, followed by 10 pmole Taq DNA polymerase (Solgent Korea) and primer [Forward primer: 5 '-CTCGAGAGAACAGATTATG-3', sequence No. 2; Reverse primer: RT-PCR was performed using 5'-CTCACTTGTGGAGACACATC-3 ', SEQ ID NO: 3]. At this time, as a gel loading control RT-PCR of GAPDH was performed. The PCR programs used were as shown in Table 4 below.
  • MAP7D2 gene is highly expressed in cell lines such as colorectal cancer, lung cancer and liver cancer as shown in Table 3 above ( 3 to 5).
  • RNA 2 and the DiaStar RT kit were supplied from the Korea Human Resource Base Bank of Pusan National University Hospital. Each tissue was crushed finely using a mortar and pestle while frozen with liquid nitrogen. Using the RNeasyMini kit (Qiagene, Cat #: 74104), the pulverized tissue was isolated from the total RNA, and then the total amount of RNA 2 and the DiaStar RT kit (SolGent, Cat #: DR13-R10K). CDNA was synthesized.
  • cDNA was distilled with sterile distilled water to prepare 2% cDNA, followed by 10 pmole / Taq DNA polymerase (Solgent, Korea) and primer primer: 5 '-CTCGAGAGAACAGAGATTATG-3', SEQ ID NO: 2; Reverse primer: RT-PCR was performed using 5'-CTTCACTTGTGGAGACACATC-3 ', SEQ ID NO: 3]. At this time, RT-PCR of ⁇ -act in was performed as a gel loading control, and the PCR program used is shown in Table 4 above.
  • MAP7D2 was highly expressed in lung cancer tissues and more than 33% in liver cancer tissues when compared with normal tissues (FIGS. 6 and 7).
  • the knockdown method was used to inhibit MAP7D2 gene expression and to analyze the effect on cancer cell growth by MAP7D2 gene expression inhibition.
  • MAP7D2 siRNA was used to determine how MAP7D2 gene expression inhibition affects the growth of kidney cancer cells.
  • each plate has a Cell Counting Kit-
  • the degree of cell proliferation was observed by measuring absorbance at 450 nm in Victor microplate reader.
  • MAP7D2 shRNA was used to cut.
  • Virus particles were prepared by transfecting the shRNA into 293T using Transfection Enhancing Reagent (WelGene, Cat #: TR001-02). After 48 hours, the virus culture supernatant was recovered and filtered with syringe filter (mi 11 ipore, Cat #: SLHP033RS), and then mixed 1: 1 with fresh culture medium, in order to improve the effect of infection. 4 yg / niL polybrene (SIGMA, Cat #: L107689) was added and transfected into lung cancer cell lines NCI-H1703 and kidney cancer cell line A498, respectively.
  • Transfection Enhancing Reagent WelGene, Cat #: TR001-02
  • syringe filter mi 11 ipore, Cat #: SLHP033RS
  • 4 yg / niL polybrene SIGMA, Cat #: L107689 was added and transfected into lung cancer cell lines NCI-H1703 and kidney cancer cell line A498, respectively.
  • MAP7D2 shRNA # 2 or # 3 was transfected.
  • cell growth was markedly decreased and cell death was increased compared to the control group (Nontarget shControlRNA) (FIG. 11).
  • MAP7D2 shRNA # 2 was confirmed to exhibit about 70% growth inhibition and apoptosis effect compared to the control group (FIG. 12).
  • MAP7D2 has a clear oncogene addiction in renal and lung cancers, and thus may be a good target for chemotherapy in renal and lung cancers.
  • Transwell (transwell Kcoastar, 3422) system was used to confirm whether the inhibition of MAP7D2 expression by cancer cells, the expression of MAP7D2 by the shRNA described in Example ⁇ 3-2> Lung cancer cell line NCI-H1703 was inhibited.
  • Nontarget shRNA Control Transduction particles were used as negative control shRNA to exclude nonspecific reaction by lentiviral vector shRNA in cells, and the lentiviral vector to which fluorescent material was attached was used.
  • the introduction efficiency was predicted. The efficiency of introduction was about 80% or more. After 48 hours of phenotypic infection, 2 ug / ml of puromycin was used for 4 days of selection, and then the cancer cells were recovered and examined for their ability to migrate.
  • the cells were suspended in transfer medium at 4 ⁇ 10 cells / ml.
  • the 24-well transwell plate is then coated with 0.05% gelatin (Sigma G1393) on the underside of the insert (i nser t) for one hour at room temperature and after one hour, the remaining gelatin in the insert.
  • 600 ul of RPMI transfer medium with 53 ⁇ 4 FBS was added to the chamber.
  • the cells were prepared in advance, the cells were placed in the insert into 4xl0 4 year old catcher / 100 ul and incubated for 24 hours at 37 ° C / 5% CO 2 conditions.
  • the cells whose MAP7D2 expression was suppressed by shRNA # 2 and # 4 were about 80% compared with the cells into which the nontarget shRNA (shCtr 1) introduced as a control was introduced. It showed a clear movement inhibitory effect (Fig. 13), it can be seen that MAP7D2 plays a critical role in the migration of cancer cells.
  • the cells were washed twice with RPMI invasion media (RPMI, lOmM HEPES, 0.5% BSA), and then the cells were suspended in the infiltration medium at 4 ⁇ 10 cells / ml.
  • 24-well transwell plates (8 urn pore size, costar 3422) were diluted with Matrigel (BD 354234) in serum-free media (RPMI, lOmM HEPES) at 1 mg / ml at room temperature on the top of the insert. Coating for one hour at. After one hour, the Matrigel remaining in the insert was removed and washed once with serum-free medium. Then, 600 ul RPMI infiltration medium with 5% FBS was added.
  • RPMI invasion media RPMI, lOmM HEPES, 0.5% BSA
  • the cells whose MAP7D2 expression was suppressed by shRNA # 2 and # 4 were about 70% compared with the cells into which the nontarget shRNA (shCtr 1) introduced as a control was introduced. It showed a significant degree of invasion inhibitory effect (FIG. 14), and it can be seen that MAP7D2 plays a critical role in cancer cell invasion.
  • the above ingredients were mixed and layered in an airtight cloth to prepare a powder.
  • tablets were prepared by tableting according to a conventional method for preparing tablets.
  • the capsule was prepared by filling in gelatin capsules according to a conventional method for preparing capsules.
  • the solution was layered in a 5 mi type I ampoule made of clear glass, encapsulated under the upper grid of air by dissolving the glass, and sterilized by autoclaving at 12 C C for at least 15 minutes to prepare an injection solution.
  • the present invention enables the development of cancer therapeutic agents using MAP7D2 expression or activity inhibitors, and the development of a method for monitoring or diagnosing cancer using MAP7D2 as a biomarker. It may be usefully used for screening cancer growth or metastasis inhibitors.

Abstract

The present invention relates to the pharmaceutical composition for cancer treatment or metastasis inhibition containing expression or activation inhibitors of MAP7D2 protein as an active ingredient. More specifically a novel anti-cancer therapeutic target has been discovered using data-mining techniques by building a large-scale gene expression database of human cancer tissues, and, based on the gene expression profiles of cancer cell lines, appropriate cancer cell lines were selected and experimentally verified through the inhibition of candidate target genes. Through these, MAP7D2 has been verified as a novel potential anti-cancer target which is over-expressed in a variety of cancer tissues or cells, such as cancer tissues or cells of the kidney, lung, large-intestine, ovary, stomach and liver, and inhibition of which impedes the growth, movement, infiltration and metastasis of cancer cells, thereby leading to cell destruction. As such, expression levels of MAP7D2 can be useful as a diagnostic marker for cancer and an MAP7D2 protein expression or activation inhibitor can be used as an active ingredient in a pharmaceutical composition for cancer treatment or metastasis inhibition.

Description

【명세서】  【Specification】
【발명의명칭】  [Name of invention]
신규한 암 치료 표적인 MAP 7 D 2 단백질의 발현 또는 활성 억제제를 포 함하는 암 치료 또는 암 전이 억제용 약학적 조성물  Pharmaceutical compositions for treating cancer or inhibiting cancer metastasis comprising inhibitors of the expression or activity of the MAP 7 D 2 protein as a novel cancer treatment target
【기술분야】  Technical Field
<ι> 본 발명은 신규한 암 성장 또는 암 전이 치료 표적인 MAP7D2 단백질의 발현 또는 활성 억제제를 포함하는 암 치료 또는 암 전이 억제용 약학적 조성물에 관한 것이다.  The present invention relates to a pharmaceutical composition for treating cancer or inhibiting cancer metastasis, comprising an inhibitor of expression or activity of MAP7D2 protein, which is a novel cancer growth or cancer metastasis therapeutic target.
<2>  <2>
【배경기술】  Background Art
<3> 암 진행 과정은 암 유발 유전자 (oncogene)의 활성화와 종양 억제 유전자 <3> Cancer Progression is Activation of Cancer-Induced Genes (oncogene) and Tumor Suppressor Genes
(tumor suppressor gene)의 비활성화를 필요로 하는 복합적인 과정이다. 최근에, 인간 암을 연구 또는 치료하기 위해 마우스 모델을 이용한 연구에서는, 종양 유지 를 위해 특정 암 유발 유전자 (예를 들어, H— ras, K-ras, Myc 등)들의 지속적인 발 현이 필요하며, 이러한 단일 암 유발 유전자의 비활성화에 의해 암이 효과적으로 치료되는 현상이 확인되고 있다. 이는 암 유발 유전자 중독 (oncogene addiction) 으로 정의되고 있으며, 현재 항암 연구의 중요한 목표는, 암에서 이러한 중독된 암 유발 유전자를 찾기 위해 융합 유전체학 (Integrative Genomics)와 시스템 생물학 (systems biology)과 같은 새로운 방법들을 통합하여 효과적인 표적화 표적을 찾는 것이다. This is a complex process that requires the deactivation of the tumor suppressor gene. Recently, studies using mouse models to study or treat human cancers require the continuous expression of certain cancer-causing genes (eg, H- ras, K-ras, Myc, etc.) for tumor maintenance. It is confirmed that cancer is effectively treated by inactivation of a single cancer causing gene. This is defined as oncogene addiction, and an important goal of current anticancer research is to find new methods such as Integrative Genomics and Systems Biology to find these addicted cancer-causing genes in cancer. To find an effective targeting target.
<4> 허셉틴 (Herceptin), 리툭산 (Rituxan), 제피티닙 (Gef it inib) 및 글리백 Herceptin, Rituxan, Gefitinib, and Glybag
(Gleevec) 등 성공적인 표적화 항암 치료제가 임상에서 쓰이면서 표적화 항암제 시 장은 큰 성장을 보이고 있으며, 다국적 제약회사들은 새로운 표적화 항암 치료제를 개발하기 위해 많은 연구 개발을 진행하고 있다. 현재 표적화 항암 표적에 대한 치료제가 개발되어 전임상 또는 임상 시험 단계에 있는 것이 다수 있으나, 여전히 각종 암에서 새로운 표적을 확보하려는 노력이 전세계적으로 진행되고 있다. 대표 적인 연구로는 미국 NCI와 NHGRI가 공동으로 진행하고 있는 TCGA The Cancer Genome Atlas) 연구로 각종 암환자의 엑손 시퀀싱 (exon sequencing)을 통해 암에서 빈번하게 발생하는 중요한 돌연변이를 찾는 방식으로 새로운 표적을 확보하려고 하 고 있다. As successful targeted anticancer drugs such as Gleevec are being used in the clinic, the targeted anticancer drug market is growing rapidly, and multinational pharmaceutical companies are conducting a lot of research and development to develop new targeted anticancer drugs. There are a number of therapeutic agents for targeted anticancer targets that are currently in preclinical or clinical trials, but efforts are still underway worldwide to acquire new targets for various cancers. A representative study is the TCGA The Cancer Genome Atlas, which is jointly conducted by NCI and NHGRI in the United States. Exxon sequencing of various cancer patients finds new targets by finding important mutations that occur frequently in cancer. I'm trying to secure it.
<5>  <5>
<6> 암은 그 원인 및 진행 과정이 대단히 이질적인 질병으로서 형태학적, 병리학 적으로는 비슷하지만 분자수준에서는 서로 현저히 다른 형태로 진행한다 . 최근의 임상 경험도 이를 뒷받침 하는데, 허셉틴이나 제피티닙과 같이 임상에서 성공적으 로 쓰이고 있는 약물들의 표적인 ERBB2 및 EGFR들은 전체 암환자 중 약 20~30% 정 도의 일부 환자들에서만 돌연변이되거나 과발현되고 있다. <6> Cancer is a disease that is very heterogeneous in its cause and progression. Morphological and pathological Although similar in nature, they progress in significantly different forms at the molecular level. Recent clinical experience supports this: ERBB2 and EGFR, the targets of successful clinical drugs such as Herceptin and Zephytinib, are mutated or overexpressed in only about 20-30% of all cancer patients. have.
<7> 이와 같이, 암 진행 과정이 서로 이질적이기 때문에, 중요 표적들을 찾기 위 해서는 가능한 한 많은 임상 시료를 확보, 분석하는 것이 필요하다. 현재 전 세계 적으로도 몇몇 연구 그룹만이 대규모 유전자 발현 데이터베이스를 확보하고 이를 이용해 표적화 암 치료 표적 발굴 연구를 진행하고 있으며, 국내의 많은 연구들은 아직 대규모 데이터베이스를 확보하지 못한 한계를 가지고 있다.  As the cancer progression is heterogeneous, it is necessary to acquire and analyze as many clinical samples as possible to find important targets. Currently, only a few research groups worldwide have a large gene expression database and are using it to discover targeted cancer treatment targets, and many studies in Korea have yet to secure large databases.
<8>  <8>
<9> MAP7D2(MAP7 domain containing 2)는 FLJ 14503, MGC104944, RP11-393H10.2의 다른 이름으로 불리는 단백질로, 미세소관 연관 단백질 (microtubule-associated protein) 7 도메인의 한 종류이다.  MAP7D2 (MAP7 domain containing 2) is another protein named FLJ 14503, MGC104944, RP11-393H10.2, which is a type of microtubule-associated protein 7 domain.
<ιο> 현재까지 MAP7D2에 관한 연구는 자폐 스펙트럼 장애 (Autism spectrum disorder, ASD)의 진단을 위한 연구에서 X-염색체 (chromosome) 상에서 발현이 감소 하는 유전자 (미국공개특허 2010/0029009 A1), 또는 X-염색체 연관 정신지체 (chromosome X- linked mental retardat ion)를 지닌 환자의 유전자 복제수 변이의 검출을 위한 X-chromosome의 발현 분석에서 Case 4 환자의 XP22.12에서 검출되는 약 1Mb의 복제에 영향을 주는 유전자로 기재되어 있을 뿐 (BMCGenomics, 2007, 8:443, 2007) , 구체적으로 MAP7D2 유전자에 대한 정보, 이의 기능 및 암과의 연관 성은 전혀 알려진 바 없다. To date, studies on MAP7D2 have been directed to genes with reduced expression on the X-chromosome in studies for the diagnosis of Autism spectrum disorder (ASD) (US Patent Publication No. 2010/0029009 A1), or X Expression analysis of X-chromosome for detection of gene copy number variation in patients with chromosome X-linked mental retardat ions affected the replication of approximately 1 Mb detected in X P 22.12 of Case 4 patients. The state is only listed as a gene (BMCGenomics, 2007, 8: 443, 2007), specifically information about the MAP7D2 gene, its function and its association with cancer are not known at all.
<11>  <11>
<12> 이에, 본 발명자들은 표적화 암 치료 표적을 발굴하기 위하여 연구를 진행한 결과, 암 조직의 대규모 유전자 발현 데이터베이스를 구축하여 독창적인 데이터마 이닝 기법을 통해, 새로운 표적화 항암 후보 표적으로서 신장암, 폐암, 대장암, 난 소암, 위암 및 간암에서 과발현되는 MAP7D2를 발굴하였고, MAP7D2의 발현 저해에 의해 암세포의 성장, 침윤 및 전이가 현저하게 억제됨을 확인함으로써 본 발명을 완성하였다.  Accordingly, the present inventors have conducted research to discover targeted cancer treatment targets, and have established a large-scale gene expression database of cancer tissues through original data mining techniques. The present invention was completed by identifying MAP7D2 that is overexpressed in lung cancer, colorectal cancer, ovarian cancer, gastric cancer, and liver cancer and remarkably inhibiting the growth, infiltration and metastasis of cancer cells by inhibition of MAP7D2 expression.
<13>  <13>
【발명의 내용】  [Content of invention]
【기술적 과제】  [Technical problem]
<14> 본 발명의 목적은 신규한 암 성장 또는 암 전이 치료 표적인 MAP7D2 단백질 의 발현 또는 활성 억제제를 유효성분으로 함유하는 암 치료 또는 암 전이 억제용 약학적 조성물을 제공하는 것이다. <14> An object of the present invention is the MAP7D2 protein, which is a novel cancer growth or cancer metastasis therapeutic target. It is to provide a pharmaceutical composition for treating cancer or inhibiting cancer metastasis, containing an inhibitor of expression or activity as an active ingredient.
<15> 또한, 본 발명의 또 다른 목적은 MAP7D2를 이용하여, 암의 성장, 침윤 또는 전이 억제제를 스크리닝하는 방법을 제공하는 것이다.  Another object of the present invention is to provide a method for screening cancer growth, invasion or metastasis inhibitors using MAP7D2.
<16> 아울러, 본 발명의 또 다른 목적은 MAP7D2를 이용하여 개체 내 암을 모니터 링 또는 진단하는 방법을 제공하는 것이다. In addition, another object of the present invention is to provide a method for monitoring or diagnosing cancer in a subject using MAP7D2.
<17>  <17>
【기술적 해결방법】  Technical Solution
<18> 상기 목적을 달성하기 위하여, 본 발명은 MAP7D2(MAP7 domain containing 2) 단백질의 발현 또는 활성 억제제를 유효성분으로 함유하는 암 치료 또는 암 전이 억제용 약학적 조성물을 제공한다.  In order to achieve the above object, the present invention provides a pharmaceutical composition for treating cancer or inhibiting cancer metastasis, containing an inhibitor of the expression or activity of MAP7D2 (MAP7 domain containing 2) protein as an active ingredient.
<19> 또한, 본 발명은 하기의 단계를 포함하는 암 치료 또는 암 전이 억제용 후보 물질의 스크리닝 방법을 제공한다:  In addition, the present invention provides a method for screening a candidate substance for treating cancer or inhibiting cancer metastasis, comprising the following steps:
<20> 1) MAP7D2 단백질의 발현 세포주에 피검물질을 처리하는 단계;  1) treating the test substance to the cell line expressing MAP7D2 protein;
<2i> 2) 상기 세포주에서 MAP7D2 단백질의 발현 정도를 측정하는 단계; 및  <2i> 2) measuring the expression level of MAP7D2 protein in the cell line; And
<22> 3) 상기 MAP7D2 단백질의 발현 정도가 피검물질을 처리하지 않은 대조군에 비해 감소한 피검물질을 선별하는 단계 .  3) selecting a test substance whose expression level of the MAP7D2 protein is decreased compared to a control group not treated with the test substance.
<23> 또한, 본 발명은 하기의 단계를 포함하는 암 치료 또는 암 전이 억제용 후보 물질의 스크리닝 방법을 제공한다:  In addition, the present invention provides a method for screening a candidate substance for cancer treatment or cancer metastasis suppression comprising the following steps:
<24> 1) MAP7D2 단백질에 피검물질을 처리하는 단계;  1) treating the test substance to the MAP7D2 protein;
<25> 2) 상기 MAP7D2 단백질의 활성 정도를 측정하는 단계 ; 및  2) measuring the activity of the MAP7D2 protein; And
<26> 3) 상기 MAP7D2 단백질의 활성 정도가 피검물질을 처리하지 않은 대조군에 비해 감소한 피검물질을 선별하는 단계 .  3) selecting the test substance whose activity level of the MAP7D2 protein is reduced compared to the control group not treated with the test substance.
<27> 또한, 본 발명은 MAP7D2 단백질을 이용한 암의 모니터링 또는 진단 방법을 제공한다: In addition, the present invention provides a method for monitoring or diagnosing cancer using MAP7D2 protein:
<28> 1) 피검체 유래 시료에서 MAP7D2 단백질의 발현 수준을 측정하는 단계; 및 1) measuring the expression level of MAP7D2 protein in a subject-derived sample; And
<29> 2) MAP7D2 단백질의 발현 수준이 대조군보다 증가된 피검체를 암에 걸릴 위 험이 있는 개체로 판정하는 단계. 2) Determining that the subject with increased expression level of MAP7D2 protein than the control group as a subject at risk of cancer.
<30> 또한, 본 발명은 약학적으로 유효한 양의 MAP7D2 단백질 발현 또는 활성 억 제제를 개체에 투여하는 단계를 포함하는 암 예방 방법올 제공한다. The present invention also provides a method for preventing cancer, comprising administering to a subject a pharmaceutically effective amount of an MAP7D2 protein expression or activity inhibitor.
<3i> 또한, 본 발명은 약학적으로 유효한 양의 MAP7D2 단백질 발현 또는 활성 억 제제를 암에 걸린 개체에 투여하는 단계를 포함하는 암 치료 또는 암 전이 억제 방 법을 제공한다. <3i> The present invention also provides a method for treating cancer or inhibiting cancer metastasis, comprising administering a pharmaceutically effective amount of an MAP7D2 protein expression or active inhibitor to a subject with cancer. Provide the law.
<32> 또한, 본 발명은 MAP7D2에 특이적으로 결합하는 항체 또는 유전자에 상보적인 핵산 중 어느 하나 이상을 이용하여 암세포에서의 MAP7D2 발현 수준을 측 정하는 단계를 포함하는, 암의 진단, 치료 결과 확인 또는 예후를 평가하는 방법을 제공한다.  In addition, the present invention comprises the step of measuring the expression level of MAP7D2 in cancer cells using any one or more of the nucleic acid complementary to the antibody or gene specifically binding to MAP7D2, the diagnosis of cancer, confirming the treatment result Or provide a method of assessing prognosis.
<33> 또한, 본 발명은 MAP7D2에 특이적으로 결합하는 항체 또는 / 유전자에 상보적인 핵산 중 어느 하나 이상을 포함하는 암 진단용 키트를 제공한다.  The present invention also provides a kit for diagnosing cancer, comprising any one or more of nucleic acids complementary to antibodies or / genes that specifically bind to MAP7D2.
<34> 또한, 본 발명은 암의 예방 또는 치료, 또는 암 전이 억제용 약학적 조성물 에 사용하기 위한, MAP7D2 단백질 발현 또는 활성 억제제를 제공한다.  The present invention also provides an inhibitor of MAP7D2 protein expression or activity for use in the prophylaxis or treatment of cancer or the pharmaceutical composition for inhibiting cancer metastasis.
<35> 아을러, 본 발명은 암 진단용 키트에 사용하기 위한, MAP7D2에 특이적으로 결합하는 항체 또는 유전자에 상보적인 핵산을 제공한다.  In addition, the present invention provides nucleic acids complementary to antibodies or genes that specifically bind to MAP7D2 for use in cancer diagnostic kits.
<36>  <36>
【유리한 효과】  Advantageous Effects
<37> 본 발명에서는 인체 암 조직의 대규모 유전자 발현 데이터베이스를 구축하여 데이터마이닝 기법을 통해 신규한 표적화 항암 후보 표적인 MAP7D2를 발굴하였고, 상기 MAP7D2는 신장암, 폐암, 대장암, 난소암, 위암 및 간암의 조직 또는 세포에서 특이적으로 현저하게 과발현되며, MAP7D2의 억제에 의해 암 세포의 성장, 이동, 침 윤 및 전이가 현저히 저해되고 세포사멸이 유도됨으로써, MAP7D2 단백질의 발현 또 는 활성 억제제를 유효성분으로 함유하는 약학적 조성물은 암의 치료 또는 암전이 억제용 약학적 조성물의 유효성분으로서 유용하게 이용될 수 있다.  In the present invention, a large-scale gene expression database of human cancer tissue was constructed to discover a novel targeting anticancer candidate target MAP7D2 through data mining techniques, and the MAP7D2 is a kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and the like. It is particularly remarkably overexpressed in tissue or cells of liver cancer, and inhibition of MAP7D2 significantly inhibits the growth, migration, invasion and metastasis of cancer cells and induces apoptosis, thereby inhibiting the expression or activity inhibitor of MAP7D2 protein. A pharmaceutical composition containing as an ingredient may be usefully used as an active ingredient of a pharmaceutical composition for treating cancer or inhibiting cancer metastasis.
<38>  <38>
【도면의 간단한 설명】  [Brief Description of Drawings]
<39> 도 1은 MAP7D2 유전자의 임상 조직에서의 발현 양상을 마이크로어레이 플랫 폼 U133plus2로 분석한 결과를 나타낸 그림이다.  FIG. 1 is a diagram showing the results of analyzing the expression pattern of the MAP7D2 gene in clinical tissues with the microarray platform U133plus2.
<40> 도 2는 MAP7D2 유전자의 암세포주에서의 발현 양상을 마이크로어레이 플랫 폼 U133plus2로 분석한 결과를 나타낸 그림이다. Figure 2 is a diagram showing the results of analyzing the expression of the MAP7D2 gene in the cancer cell line microarray platform U133plus2.
<4i> 도 3은 RNA 양의 표준화를 위해 GAPDH를 내부 대조군으로서 사용하여, 3 illustrates the use of GAPDH as an internal control for standardization of RNA amounts.
MAP7D2의 대장암 세포주에서의 발현 양상을 나타낸 그림이다.  Figure shows the expression of MAP7D2 in colorectal cancer cell line.
<42> 도 4는 RNA 양의 표준화를 위해 GAPDH를 내부 대조군으로서 사용하여, 4 uses GAPDH as an internal control for standardization of RNA amounts.
MAP7D2의 간암 세포주에서의 발현 양상을 나타낸 그림이다.  Figure shows the expression of MAP7D2 in liver cancer cell line.
<43> 도 5는 RNA 양의 표준화를 위해 GAPDH를 내부 대조군으로서 사용하여, FIG. 5 uses GAPDH as an internal control for standardization of RNA amounts.
MAP7D2의 폐암 세포주에서의 발현 양상을 나타낸 그림이다. <44> 도 6은 폐암 환자들의 정상 조직과 폐암 조직에서 MAP7D2의 발현 양상을 비 교하기 위해 RT-PCR을 수행한 결과이다: Figure shows the expression of MAP7D2 in lung cancer cell line. FIG. 6 shows the results of RT-PCR to compare the expression of MAP7D2 in normal and lung cancer tissues of lung cancer patients:
<45> -act in: 베타 액틴 (대조군); N: 정상 조직; 및 T: 암조직. -Act in: beta actin (control); N: normal tissue; And T: cancer tissue.
<46> 도 7은 간암 환자들의정상 조직과 간암 조직에서 MAP7D2의 발현 양상을 비교 하기 위해 RT-PCR을 수행한 결과이다:  FIG. 7 shows the results of RT-PCR to compare the expression patterns of MAP7D2 in normal and liver cancer tissues of liver cancer patients:
<47> β -act in: 베타 액틴 (대조군); N: 정상 조직; 및 T: 암조직. Β-act in: beta actin (control); N: normal tissue; And T: cancer tissue.
<48> 도 8은 siRNA에 의해, MAP7D2의 발현이 녹다운된 신장암 세포주 A498에서  FIG. 8 shows the renal cancer cell line A498 knocked down by the expression of MAP7D2 by siRNA.
MAP7D2 유전자의 발현을 RT-PCR을 통해 확인한 그림이다.  The expression of MAP7D2 gene was confirmed by RT-PCR.
<4 > 도 9은 siRNA에 의해, MAP7D2의 발현이 녹다운된 신장암 세포주 A498에서 세 포 성장의 저해 효과를 확인한 결과이다. Figure 9 shows the results of confirming the inhibitory effect of cell growth in the renal cancer cell line A498 knocked down expression of MAP7D2 by siRNA.
<50> 도 10은 shRNA에 의해, MAP7D2의 발현이 녹다운된 폐암 세포주 NCI-H1703에 서 MAP7D2 유전자의 발현을 RT-PCR을 통해 확인한 그림이다. 10 is a diagram confirming the expression of the MAP7D2 gene by RT-PCR in lung cancer cell line NCI-H1703 in which expression of MAP7D2 is knocked down by shRNA.
<5i> 도 11는 MAP7D2의 발현을 녹다운한 5일 후, 폐암 세포주 NCI-H1703의 형태를 현미경 (X100)을 통해 관찰한 그림이다 (shCtrl: Nontarget shControl RNA). <5i> FIG. 11 is a view of morphology of lung cancer cell line NCI-H1703 observed under a microscope (X100) 5 days after knocking down MAP7D2 expression (shCtrl: Nontarget shControl RNA).
<52> 도 12은 폐암 세포주 NCI-H1703에서 MAP7D2 유전자의 녹다운에 의한 세포 성 장의 변화를 나타낸 그래프이다 (bar: 표준편차). 12 is a graph showing changes in cell growth due to knockdown of the MAP7D2 gene in lung cancer cell line NCI-H1703 (bar: standard deviation).
<53> 도 13은 폐암 세포주 NCI-H1703에서 MAP7D2 유전자의 녹다운에 의한 세포 이 동의 변화를 나타낸 그림이다: FIG. 13 is a diagram showing the change in cell migration by knockdown of MAP7D2 gene in lung cancer cell line NCI-H1703:
<54> A: shRNA로 형질도입한 NCI-H1703 세포의 트랜스웰 플레이트를 이용한 암세 포 이동 실험을 현미경으로 관찰한 그림 (X100 배율); 및 A: A picture of a microscopic observation of a cancer cell migration experiment using a transwell plate of NCI-H1703 cells transduced with shRNA (X100 magnification); And
<55> B: MAP7D2 발현 감소에 의한 NCI-H1703 이동 변화를 수치화한 그래프 (X200 B: A graph quantifying changes in NCI-H1703 migration due to decreased MAP7D2 expression (X200)
배율에서 임의적으로 고른 8 면의 세포수를 계수함, bar: 표준편차).  Count the number of randomly selected 8-sided cells in magnification, bar: standard deviation).
<56> 도 14는 폐암 세포주 NCI-H1703에서 MAP7D2 유전자의 녹다운에 의한 세포 침 윤의 변화를 나타낸 그림이다: FIG. 14 is a diagram showing the change of cell invasion by knockdown of MAP7D2 gene in lung cancer cell line NCI-H1703:
<57> A: shRNA로 형질도입한 NCI-H1703 세포의 트랜스웰 플레이트를 이용한 암세 포 침윤 실험을 현미경으로 관찰한 그림 (X100 배율); 및 A: microscopic observation of cancer cell infiltration experiments using transwell plates of NCI-H1703 cells transduced with shRNA (X100 magnification); And
<58> B: MAP7D2 발현 감소에 의한 NCI-H1703 침윤 변화를 수치화한 그래프 (X200 B: A graph quantifying changes in NCI-H1703 invasion due to decreased MAP7D2 expression (X200)
배율에서 임의적으로 고른 8개 필드 (field)의 세포 수를 계수함, bar: 표준편차). Count the number of cells in 8 fields randomly chosen at magnification, bar: standard deviation).
<59> <59>
【발명의 실시를 위한 최선의 형태】  [Best form for implementation of the invention]
<60> 이하, 본 발명을 상세히 설명한다. <62> 본 발명은 MAP7D2(MAP7 domain containing 2) 단백질의 발현 또는 활성 억제 제를 유효성분으로 함유하는 암 치료 또는 암 전이 억제용 약학적 조성물을 제공한 다. Hereinafter, the present invention will be described in detail. The present invention provides a pharmaceutical composition for treating cancer or inhibiting cancer metastasis, comprising an MAP7D2 (MAP7 domain containing 2) protein expression or activity inhibitor as an active ingredient.
<63> 상기 MAP7D2 단백질은 서열번호 1로 기재되는 아미노산 서열을 가지는 것이 바람직하나 이에 한정되지 않는다.  The MAP7D2 protein preferably has an amino acid sequence as set forth in SEQ ID NO: 1, but is not limited thereto.
<64> 상기 암은 신장암, 폐암, 대장암, 난소암, 위암 및 간암으로 구성된 군으로 부터 선택되는 어느 하나인 것이 바람직하고, 폐암 또는 신장암인 것이 더욱 바람 직하나, 이에 한정되지 않는다 .  The cancer is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer, and more preferably lung cancer or kidney cancer, but is not limited thereto.
<65> 상기 MAP7D2 단백질의 발현 억제제는 MAP7D2 유전자의 mRNA에 상보적으로 결 합하는 안티센스 뉴클레오티드, 짧은 헤어핀 RNA(small hairpin RNA), 작은 간섭 RNA( small interfering RNA) 및 리보자임 (ribozyme)으로 구성된 군으로부터 선택되 는 어느 하나인 것이 바람직하고, 상기 MAP7D2 단백질의 활성 억제제는 MAP7D2 단 백질에 상보적으로 결합하는 화합물, 펩티드, 펩티드 미메틱스, 기질유사체, 앱타 머 및 항체로 구성된 군으로부터 선택되는 어느 하나인 것이 바람직하나 이에 한정 되지 않는다.  The inhibitor of expression of the MAP7D2 protein is from the group consisting of antisense nucleotides, small hairpin RNAs, small interfering RNAs, and ribozymes that complementarily bind to the mRNA of the MAP7D2 gene. The MAP7D2 protein inhibitor is any one selected from the group consisting of compounds, peptides, peptide mimetics, substrate analogs, aptamers, and antibodies that complementarily bind to MAP7D2 proteins. Is preferably, but is not limited thereto.
<66> 상기 siRNA는 인간 MAP7D2 단백질을 암호화하는 유전자의 mRNA의 염기서열 내에서 선택되는 15 내지 30머 (mer)의 센스 서열 및 상기 센스 서열에 상보적으로 결합하는 안티센스 서열로 구성되며, 이때, 상기 센스 서열은 특별히 이에 제한되 는 것은 아니나, 25개의 염기로 구성되는 것이 바람직하나 이에 한정되지 않으며, 서열번호 4 또는 5인 것이 더욱 바람직하나, 이에 한정되지 않는다.  The siRNA is composed of a 15 to 30 mer sense sequence selected from the base sequence of the mRNA of the gene encoding a human MAP7D2 protein and an antisense sequence complementary to the sense sequence, wherein The sense sequence is not particularly limited thereto, but is preferably composed of 25 bases, but is not limited thereto, and more preferably, SEQ ID NO: 4 or 5, but is not limited thereto.
<67> 상기 안티센스 뉴클레오티드는 왓슨 -클릭 염기쌍에 정의된 바에 따라, DNA, 미성숙 -mRNA 또는 성숙된 mRNA의 상보적 염기서열에 결합 (흔성화)하여 DNA에서 단 백질로서 유전정보의 흐름을 방해하는 것이다. 표적 서열에 특이성이 있는 안티센 스 뉴클레오티드의 성질은 그것들을 예외적으로 다기능이 되도톡 한다. 안티센스 뉴클레오티드는 모노머 단위의 긴 사슬이기 때문에 이들은 표적 RNA 서열에 대해 쉽게 합성될 수 있다. 최근 많은 연구들은 표적 단백질을 연구하기 위한 생화학적 수단으로 안티센스 뉴클레오티드의 유용성을 증명하였다 (Rothenberg et al . , J. Natl. Cancer Inst., 81:1539-1544, 1999). 올리고뉴클레오티드 화학 및 향상된 세포흡착, 표적결합 친화도 및 뉴클레아제 내성을 나타내는 뉴클레오티드 합성 분 야에서 최근 많은 진보가 있었으므로 안티센스 뉴클레오티드의 사용은 새로운 형태 의 억제제로 고려될 수 있다.  The antisense nucleotide binds to (generalizes) the complementary sequencing of DNA, immature -mRNA, or mature mRNA, as defined by the Watson-click base pair, thereby disrupting the flow of genetic information as a protein in DNA. will be. The nature of antisense nucleotides that are specific for the target sequence makes them exceptionally versatile. Since antisense nucleotides are long chains of monomeric units they can be easily synthesized for the target RNA sequence. Many recent studies have demonstrated the utility of antisense nucleotides as biochemical means for studying target proteins (Rothenberg et al., J. Natl. Cancer Inst., 81: 1539-1544, 1999). The use of antisense nucleotides can be considered as a new type of inhibitor because of recent advances in oligonucleotide chemistry and in the synthesis of nucleotides that exhibit improved cell adsorption, target binding affinity and nuclease resistance.
<68> 상기 펩티드 미메틱스 (Peptide Minetics)는 MAP7D2 단백질의 결합 도메인을 억제하는 MAP7D2 단백질의 활성을 억제하는 것이다. 펩티드 미메틱스는 펩티드 또 는 비펩티드일 수 있고, pSi 결합 (Benkirane, N. , et al. J. Biol. Chem., 271:33218-33224, 1996)과 같은, 비펩티드 결합에 의해 결합된 아미노산으로 구성 될 수 있다. 또한, "구조적으로 강제된 (conformational ly constrained)" 펩티드, 사이클릭 미메틱스 (cyclic mimetics), 적어도 하나의 액소사이클릭 도메인 (exocyclic domain), 결합 부분 (결합 아미노산) 및 활성 부위를 포함하는 사이클릭 미메틱스일 수 있다. 펩티드 미메틱스는 MAP7D2 단백질의 이차구조 특성과 유사하 게 구조화되고 항체 (Park, B. W. et al . Nat Biotechnol 18, 194-198, 2000) 또는 수용성 수용체 (Takasaki, W. et al . Nat Biotechnol 15, 1266-1270, 1997)와 같은 거대한 분자의 억제 특성을 모방할 수 있으며, 천연의 길항제와 동등한 효과로 작 용할 수 있는 신규한 소분자일 수 있다 (Wrighton, N. C. et al. Nat Biotechnol 15, 1261-1265, 1997). Peptide Minetics binds to the binding domain of the MAP7D2 protein. It is to inhibit the activity of the inhibitory MAP7D2 protein. Peptide or non-systematic seuneun peptide may be a non-peptide, S p i coupling (Benkirane, N., et al J. Biol Chem, 271:... 33218-33224, 1996) and engaged by the same, a non-peptide bond Can be composed of amino acids. Also, a "conformational ly constrained" peptide, between cyclic mimetics, at least one exocyclic domain, binding moiety (binding amino acid) and active site It may be a click mimetics. Peptide mimetics are structured similar to the secondary structural properties of the MAP7D2 protein and are either antibodies (Park, BW et al. Nat Biotechnol 18, 194-198, 2000) or water soluble receptors (Takasaki, W. et al. Nat Biotechnol 15, 1266). -1270, 1997), which can mimic the inhibitory properties of large molecules, and may be novel small molecules that work with the same effect as natural antagonists (Wrighton, NC et al. Nat Biotechnol 15, 1261-1265, 1997).
<69> 상기 앱타머 (aptamer)는 단일 사슬 DNA 또는 RNA 분자로서 ,  The aptamer is a single chain DNA or RNA molecule,
SELEX(systemat ic evo 1 ut i on of 1 i gands by exponential enrichment)라 불리는 올 리고뉴클레오타이드 (oligonucleotide) 라이브러리를 이용한 진화적인 방법에 의해 특정 화학 분자나 생물학적 분자에 높은 친화력과 선별력을 갖고 결합하는 올리고 머를 분리하여 수득할 수 있다 (C. Tuerand L. Gold, Science 249, 505 - 510, 2005; A. D. Ellington and J. W. Szostak, Nature 346, 818 - 822, 1990; M. Famulok, et . al. , Acc. Chem. Res. 33, 591 - 599, 2000; D. S. Wilson and Szostak, Annu. Rev. Biochem. 68, 611 - 647, 1999) . 앱타머는 표적에 특이적으 로 결합하고 표적의 활성을 조정할 수 있는데, 예컨대, 결합을 통하여 표적이 기능 하는 능력을 차단할 수 있다.  Oligomers that bind with specific affinity and selectivity to specific chemical or biological molecules by evolutionary methods using oligonucleotide libraries called systemat ic evo 1 on i 1 and g of s by exponential enrichment (SELEX) Can be obtained separately (C. Tuerand L. Gold, Science 249, 505-510, 2005; AD Ellington and JW Szostak, Nature 346, 818-822, 1990; M. Famulok, et. Al., Acc. Chem. Res. 33, 591-599, 2000; DS Wilson and Szostak, Annu. Rev. Biochem. 68, 611-647, 1999). Aptamers can specifically bind to targets and modulate the activity of the targets, such as by blocking the ability of the targets to function through binding.
<70> 상기 항체는 MAP7D2에 특이적이고 직접적으로 결합하여 MAP7D2의 활성을 효 과적으로 억제할 수 있다. 상기 MAP7D2에 특이적으로 결합하는 항체는 폴리클로날 (polyclonal) 항체 또는 모노클로날 (monoclonal ) 항체를 사용하는 것이 바람직하 다. 상기 MAP7D2에 특이적으로 결합하는 항체는 당업자에게 알려진 공지의 방법으 로 제작하여도 무방하며 , 상업적으로 알려진 MAP7D2 항체를 구입하여 사용할 수 있 다. 상기 항체는 당업자에게 알려진 종래 방법에 따라 면역원인 MAP7D2 단백질을 외부 숙주에 주사함으로써 제조될 수 있다. 외부 숙주는 마우스, 래트, 양, 토끼 와 같은 포유동물을 포함한다. 면역원은 근내, 복강내 또는 피하 주사방법으로 주 사되며, 일반적으로 항원성을 증가시키기 위한 보조제 (adjuvant)와 함께 투여할 수 있다. 외부 숙주로부터 정기적으로 혈액을 채취하여 형상된 역가 및 항원에 대한 특이성을 보이는 혈청을 수거하여 항체를 분리할 수 있다. The antibody may specifically and directly bind to MAP7D2 to effectively inhibit MAP7D2 activity. As the antibody specifically binding to MAP7D2, it is preferable to use a polyclonal antibody or a monoclonal antibody. The antibody that specifically binds to MAP7D2 may be prepared by known methods known to those skilled in the art, and commercially known MAP7D2 antibodies may be purchased and used. The antibody can be prepared by injecting the immunogen MAP7D2 protein into an external host according to conventional methods known to those skilled in the art. External hosts include mammals such as mice, rats, sheep and rabbits. Immunogens are injected intramuscularly, intraperitoneally or subcutaneously, and can generally be administered in combination with an adjuvant to increase antigenicity. Blood is drawn periodically from an external host to determine the titer and antigen Antibodies can be isolated by collecting specific serum.
<71> 상기 조성물은 암의 증식, 이동, 침윤 또는 전이의 저해 활성을 갖을 수 있 다. The composition may have inhibitory activity of cancer proliferation, migration, invasion or metastasis.
<72>  <72>
<73> 본 발명의 구체적인 실시예에서, 본 발명자들은 NCBI의 Gene Expression  In a specific embodiment of the present invention, the present inventors describe the gene expression of NCBI.
Omnibus와 EBI의 ArrayExpress의 유전자 발현 데이터로부터, 마이크로어레이 분석 을 통해 인체 시료의 유전자 발현 데이터베이스를 구축하였고 (표 1 참조), 상기 데 이터베이스에는 정상 및 암 조직에 대한 유전자 발현 데이터가 포함되어있다 (표 2 참조). 독자적으로 구축한 상기 암 유전자 발현 데이터베이스로부터 암 치료 표적 을 발굴하기 위하여, 암 이상값 프로파일 분석 (Cancer Outlier Profile Analysis, COPA)을 적용하였다 (Tomlins et al . 2005).  From the gene expression data of OmniExpress and EBI's ArrayExpress, a microarray analysis was used to build a gene expression database of human samples (see Table 1), which contains gene expression data for normal and cancerous tissues (see Table 1). See Table 2). Cancer Outlier Profile Analysis (COPA) was applied to identify cancer treatment targets from the cancer gene expression database built independently (Tomlins et al. 2005).
<74> 본 발명자들은 구축된 유전자 발현 데이터베이스로부터 암 치료를 위한 후보 표적 유전자를 선별하기 위해 특정 암 조직에서 특이적으로 과발현되는 유전자를 선정하여 그 유전자가 일정 수준이상 높게 발현되는 세포주를 선별하였다. 이때, 상기 유전자는 암 관련하여 보고된 것이 거의 없는 신규한 표적 유전자인 것으로 선정된 것이며, 이와 같은 과정으로, MAP7D2를 표적화 항암 치료의 후보 표적 유전 자로서 선별하여, MAP7D2의 발현을 마이크로어레이 분석을 통해 확인해 본 결과, 신장암, 폐암, 대장암, 난소암, 위암 및 간암 등의 조직에서 정상조직 또는 다른 암 조직들에 비해 높게 발현되는 것으로 나타났고 (도 1 참조), 이와 일치하게, 신 장암, 폐암, 대장암, 난소암, 위암 및 간암 세포주에서 비교적 높게 발현됨을 확인 하였다 (도 2 참조). 또한, 마이크로어레이 분석 결과에서 정상조직 또는 다른 암 들에서 보다 MAP7D2 발현이 현저히 높게 나타난, 대장암, 폐암 및 간암 세포주에서 의 MAP7D2 유전자 발현을 확인한 결과, 상기 세포주에서 MAP7D2 유전자가 높게 발 현됨을 확인하였다 (도 3 내지 도 5 참조). 또한, 폐암 및 간암 조직에서도 정상 조직에 비해 MAP7D2가 높게 발현됨을 확인하였으며 (도 6 및 도 7 참조), 이를 통해 MAP7D2가 대장암, 폐암 또는 간암의 표적 단백질로서 이용 가능함을 알 수 있었다. <75> 또한, 본 발명자들은, 특히 높은 MAP7D2의 발현량을 나타낸, 폐암 또는 신장 암 세포주에 짧은 헤어핀 RNA( short hairpin RNA, shRNA) 또는 작은 간섭 RNA( small interfering RNA)를 형질도입하여 MAP7D2 유전자의 발현을 억제시키고, 상기 세포주의 암 세포의 성장, 이동, 침윤 및 전이를 관찰하였다. 그 결과, shRNA또는 siRNA의 암 세포내 도입에 의해 MAP7D2 유전자 발현이 억제됨을 확인하 였고 (도 8 및 도 10 참조), 암세포의 증식이 현저하게 감소하였고 세포사멸이 증가 하였으며 (도 9 및, 도 11 내지 도 12 참조), 암세포의 이동 및 침윤이 저해됨을 관 찰하였다 (도 13 및 도 14 참조). 따라서, MAP7D2의 발현 또는 활성 억제는 폐암 또는 신장암과 같은 암세포의 증식을 저해하고, 침윤 및 전이에 결정적인 역할을 하는 것을 알 수 있었다. In order to select candidate target genes for cancer treatment from the constructed gene expression database, the present inventors selected genes that are specifically overexpressed in specific cancer tissues, and selected cell lines in which the genes were expressed higher than a certain level. At this time, the gene was selected as a novel target gene with little reported on cancer. In this process, MAP7D2 was selected as a candidate target gene for targeted chemotherapy, and the expression of MAP7D2 was analyzed by microarray analysis. As a result, it was found that the renal cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer were expressed higher than normal tissues or other cancer tissues (see FIG. 1). , Lung cancer, colorectal cancer, ovarian cancer, gastric cancer and liver cancer cell lines were confirmed to be relatively high expression (see Figure 2). In addition, the results of MAP7D2 gene expression in colorectal cancer, lung cancer, and liver cancer cell lines, which showed significantly higher MAP7D2 expression than normal tissues or other cancers, showed that the MAP7D2 gene was highly expressed in the cell lines. (See FIGS. 3-5). In addition, it was confirmed that MAP7D2 was expressed higher in lung cancer and liver cancer tissues than normal tissues (see FIGS. 6 and 7), and it could be seen that MAP7D2 can be used as a target protein of colorectal cancer, lung cancer, or liver cancer. In addition, the present inventors transduced short hairpin RNA (shRNA) or small interfering RNA (LNA) genes into lung or kidney cancer cell lines, which show high expression levels of MAP7D2. Expression was inhibited and growth, migration, infiltration and metastasis of cancer cells of the cell line were observed. As a result, it was confirmed that MAP7D2 gene expression was inhibited by introduction of shRNA or siRNA into cancer cells (see FIGS. 8 and 10), and proliferation of cancer cells was markedly reduced and cell death was increased. 9 and 11 to 12, it was observed that the migration and infiltration of cancer cells are inhibited (see FIGS. 13 and 14). Therefore, it was found that inhibition of expression or activity of MAP7D2 inhibits the proliferation of cancer cells such as lung cancer or kidney cancer and plays a decisive role in invasion and metastasis.
<76> 따라서, MAP7D2는 신장암, 폐암, 대장암, 난소암, 위암 및 간암의 조직 및 상기 암의 세포주에서 과발현하고 상기 유전자의 발현이 억제되면 암 세포의 성장, 이동 및 침윤이 저해됨으로, MAP7D2 단백질의 발현 또는 활성 억제제는 암 치료 또 는 암 전이 억제용 약학적 조성물의 유효성분으로서 유용하게 이용될 수 있다. Therefore, MAP7D2 is overexpressed in the tissues of renal cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer and the cell lines of the cancer, and the inhibition of the expression of the gene inhibits the growth, migration and invasion of cancer cells. Inhibitors of the expression or activity of MAP7D2 protein may be usefully used as an active ingredient of a pharmaceutical composition for treating cancer or inhibiting cancer metastasis.
<77>  <77>
<78> 본 발명의 MAP7D2 단백질의 발현 또는 활성 억제제를 유효성분으로 함유하는 약학적 조성물은, 조성물 총 중량에 대하여 상기 유효성분을 0.0001 내지 50 중량 % 로 포함하는 것이 바람직하나 이에 한정되지 않는다. <78> A pharmaceutical composition containing an inhibitor of the expression or activity of MAP7D2 protein of the present invention as an active ingredient, wherein the active ingredient is 0 . It is preferable to include in 0001 to 50% by weight, but is not limited thereto.
<7 > 본 발명의 약학적 조성물은, 투여를 위해서 상기 기재한 유효성분 이외에 추 가로 약제학적으로 허용 가능한 담체를 1종 이상 포함하여 제조할 수 있다. 약제 학적으로 허용 가능한 담체는 식염수, 멸균수, 링거액, 완충 식염수, 덱스트로즈 용액, 말토 덱스트린 용액, 글리세를, 에탄올, 리포좀 및 이들 성분 중 1 성분 이 상을 흔합하여 사용할 수 있으며, 필요에 따라 항산화제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다. 또한 희석제, 분산제, 계면활성제, 결합제 및 윤활제를 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주사용 제형, 환 약, 캡슐, 과립 또는 정제로 제제화할 수 있으며, 표적 기관에 특이적으로 작용할 수 있도록 표적 기관 특이적 항체 또는 기타 리간드를 상기 담체와 결합시켜 사용 할 수 있다. 더 나아가 당해 기술분야의 적정한 방법으로 또는 레밍턴의 문헌 (Remington' s Pharmaceutical Science (최근판), Mack Publishing Company, Easton PA)에 개시되어 있는 방법을 이용하여 각 질환에 따라 또는 성분에 따라 바람직하 게 제제화할 수 있다.  <7> The pharmaceutical composition of the present invention may be prepared by including one or more pharmaceutically acceptable carriers in addition to the above-described active ingredients for administration. Pharmaceutically acceptable carriers include saline, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol, liposomes and one or more of these components in combination. Other conventional additives, such as antioxidants, buffers, bacteriostatics, can be added. In addition, diluents, dispersants, surfactants, binders and lubricants can be added in addition to formulate into injectable formulations, such as aqueous solutions, suspensions, emulsions, pills, capsules, granules or tablets, which will act specifically on target organs. Target organ specific antibodies or other ligands can be used in combination with the carrier so that they can be used. Furthermore, according to the appropriate method in the art or using the method disclosed in Remington's Pharmaceutical Science (Recent), Mack Publishing Company, Easton PA, it is preferred according to each disease or component. It may be formulated.
<80> 본 발명에서 사용되는 뉴클레오티드 또는 핵산은, 경구, 국소, 비경구, 비 내, 정맥 내, 근육 내, 피하, 안 내, 경피 등의 투여를 목적으로 제조될 수 있다. <81> 바람직하게는, 핵산 또는 백터가 주사가능한 형태로 사용된다. 이에 따라서 특히 처리될 영역으로는 직접적인 주입을 위하여 주사 가능한 조성물을 위한 임의 의 약학적으로 허용되는 매개체와 혼합될 수 있다. 본 발명의 약학적 조성물은 특 히 등장 멸균 용액 또는 건조 특히 멸균수 또는 적절한 생리 식염수의 첨가에 따라 주사 가능한 용액의 조성을 가능케 하는 동결건조 조성물을 포함할 수 있다. 환자 의 종양으로의 핵산의 직접적인 주입은 치료 효율을 감염된 조직에 집중시키도록 하므로 유리하다. 사용되는 핵산의 투여량은 다양한 파라미터, 특히 유전자, 백 터, 사용되는 투여 방식, 문제시되는 질병 또는 대안적으로 요구되는 치료기간에 의해 조절될 수 있다. 또한, 환자의 체중, 연령, 성별, 건강상태, 식이, 투여시 간, 투여방법, 배설율 및 질환의 중증도 등에 따라 그 범위가 다양하다. 일일 투 여량은 약 0.0001 내지 100 /이고, 바람직하게는 0.001 내지 10 /이며, 하루 1회 내지 수회 나누어 투여하는 것이 바람직하다. The nucleotides or nucleic acids used in the present invention may be prepared for the purpose of oral, topical, parenteral, intranasal, intravenous, intramuscular, subcutaneous, intraocular, transdermal and the like. Preferably, the nucleic acid or vector is used in injectable form. Thus, in particular, the area to be treated may be mixed with any pharmaceutically acceptable media for injectable compositions for direct infusion. The pharmaceutical compositions of the present invention may in particular comprise isotonic sterile solutions or lyophilized compositions which allow the composition of injectable solutions upon the addition of dry, in particular sterile water or appropriate physiological saline. patient Direct injection of nucleic acid into tumors of the tumor is advantageous because it allows the treatment efficiency to be focused on the infected tissue. The dosage of nucleic acid used can be adjusted by various parameters, in particular by gene, vector, mode of administration used, disease in question or alternatively required duration of treatment. In addition, the range varies depending on the patient's weight, age, sex, health status, diet, administration time, administration method, excretion rate and the severity of the disease. The daily dose is about 0.0001 to 100 /, preferably 0.001 to 10 /, preferably administered once to several times a day.
<82>  <82>
<83> 또한, 본 발명은 MAP7D2 단백질을 이용한 암 치료 또는 암 전이 억제용 후보 물질의 스크리닝 방법을 제공한다.  In addition, the present invention provides a method for screening a candidate substance for cancer treatment or cancer metastasis inhibition using MAP7D2 protein.
<84> 상기 암은 신장암, 폐암, 대장암, 난소암, 위암 및 간암으로 구성된 군으로 부터 선택되는 어느 하나인 것이 바람직하고, 폐암 또는 신장암인 것이 더욱 바람 직하나, 이에 한정되지 않는다.  The cancer is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer, and more preferably lung cancer or kidney cancer, but is not limited thereto.
<85> 구체적으로, 상기 방법은  Specifically, the method
<86> 1) MAP7D2 단백질의 발현 세포주에 피검물질을 처리하는 단계;  1) treating the test substance to the cell line expressing MAP7D2 protein;
<87> 2) 상기 세포주에서 MAP7D2 단백질의 발현 정도를 측정하는 단계 ; 및  2) measuring the expression level of MAP7D2 protein in the cell line; And
<88> 3) 상기 MAP7D2 단백질의 발현 정도가 피검물질을 처리하지 않은 대조군에 비해 감소한 피검물질을 선별하는 단계를 포함하는 것이 바람직하나 이에 한정되지 않는다.  3) preferably, but not limited to, selecting a test substance whose expression level of the MAP7D2 protein is reduced compared to a control group not treated with the test substance.
<89> 상기 방법에 있어서, 단계 1)의 세포주는 신장암, 폐암, 대장암, 난소암, 위 암 및 간암 세포주로 구성된 군으로부터 선택되는 어느 하나인 것이 바람직하고, 폐암 또는 신장암 세포주인 것이 더욱 바람직하나, 이에 한정되지 않는다 .  In the above method, the cell line of step 1) is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer cell lines, and the lung cancer or kidney cancer cell lines. More preferably, it is not limited to this.
<90> 상기 방법에 있어서, 단계 2)의 단백질의 발현 정도는 면역침강법  In the above method, the expression level of the protein of step 2) is immunoprecipitation method.
(immunoprecipitation), 방사능면역분석법 (RIA) , 효소면역분석법 (ELISA), 면역조직 화학, RT-PCR, 웨스턴 블롯 (Western Blotting) 및 유세포 분석법 (FACS)으로 이루어 진 군으로부터 선택된 어느 하나로 측정하는 것이 바람직하나 당업자에게 알려진 전사체 또는 그로부터 코딩된 단백질의 양을 측정하는 모든 방법을 사용할 수 있 다.  (immunoprecipitation), radioimmunoassay (RIA), enzyme immunoassay (ELISA), immunohistochemistry, RT-PCR, Western blotting and flow cytometry (FACS). Any method of measuring the amount of transcript or protein encoded therefrom known to one skilled in the art can be used.
<91> 또 다른 방법은,  <91> Another way is
<92> 1) MAP7D2 단백질에 피검물질을 처리하는 단계;  1) treating the test substance to the MAP7D2 protein;
<93> 2) 상기 MAP7D2단백질의 활성 정도를 측정하는 단계 ; 및  2) measuring the activity of the MAP7D2 protein; And
<94> 3) 상기 MAP7D2 단백질의 활성 정도가 피검물질을 처리하지 않은 대조군에 비해 감소한 피검물질을 선별하는 단계를 포함하는 것이 바람직하나 이에 한정되지 않는다. 3) The activity level of the MAP7D2 protein is compared to the control group not treated with the test substance. It is preferable to include the step of screening a reduced test substance compared to but is not limited thereto.
<95> 상기 방법에 있어서, 단계 2)의 단백질의 활성 정도는 SDS-PAGE,.면역형광  In the above method, the activity level of the protein of step 2) is SDS-PAGE ,.
법, 효소면역분석법 (ELISA), 질량분석 및 단백질 칩으로 구성된 군으로부터 선택되 는 어느 하나로 측정하는 것이 바람직하나 이에 한정되지 않는다.  Method, enzyme immunoassay (ELISA), mass spectrometry and protein chip is preferably measured by any one selected from the group consisting of, but not limited to.
<96>  <96>
<97> 본 발명의 구체적인 실시예에서, 본 발명에서 구축한, 인체 암 조직의 대규 모 유전자 발현 데이터베이스를 바탕으로 하는 데이터마이닝 기법을 통해 신규한 표적화 항암 후보 표적인 MAP7D2를 선별하였고, 상기 MAP7D2는 신장암, 폐암, 대장 암, 난소암, 위암 및 간암의 조직 및 세포에서 과발현되고, MAP7D2가 억제된 암세 포는 성장, 이동, 침윤 및 전이가 저해되고 세포사멸이 유도됨으로써, 이를 통해 MAP7D2가 암세포에 결정적인 역할을 하는 것을 확인하였으므로, MAP7D2 단백질의 발현 또는 활성 정도를 측정하는 방법은 암의 치료 또는 암전이 억제용 물질을 스 크리닝하는 데 유용하게 이용될 수 있다.  In a specific embodiment of the present invention, a novel targeting anticancer candidate target MAP7D2 was selected through a data mining technique based on a large gene expression database of human cancer tissue constructed in the present invention. Cancer cells that are overexpressed in tissues and cells of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer, and inhibited MAP7D2 inhibit growth, migration, invasion and metastasis, and induce apoptosis. Since it has been determined to play a crucial role in the method, the method of measuring the expression or activity of MAP7D2 protein can be usefully used for screening a substance for inhibiting cancer treatment or cancer metastasis.
<98>  <98>
<99> 또한, 본 발명은 MAP7D2 단백질을 이용한 암의 모니터링 또는 진단 방법을 제공한다.  In addition, the present invention provides a method for monitoring or diagnosing cancer using MAP7D2 protein.
<100> 구체적으로, 상기 방법은  Specifically, the method
<ιοι> 1) 피검체 유래 시료에서 MAP7D2 단백질의 발현 수준을 측정하는 단계; 및 <ιοι> 1) measuring the expression level of MAP7D2 protein in a subject-derived sample; And
<102> 2) MAP7D2 단백질의 발현 수준이 대조군보다 증가된 피검체를 암에 걸릴 위 험이 있는 개체로 판정하는 단계를 포함하는 것이 바람직하나 이에 한정되지 않는 다. 2) preferably, but not limited to, a step of determining a subject having an increased risk of expression of MAP7D2 protein as a subject at risk of cancer.
<103> 상기 방법에 있어서, 단계 1)의 MAP7D2는 서열번호 1로 기재되는 아미노산 서열을 가지는 것이 바람직하나, 이에 한정되지 않는다.  In the above method, MAP7D2 in step 1) preferably has an amino acid sequence set forth in SEQ ID NO: 1, but is not limited thereto.
<104> 상기 방법에 있어서, 단계 2)의 암은 신장암, 폐암, 대장암, 난소암, 위암 및 간암으로 구성된 군으로부터 선택되는 어느 하나인 것이 바람직하고, 폐암 또는 신장암인 것이 더욱 바람직하나, 이에 한정되지 않는다. In the above method, the cancer of step 2) is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, stomach cancer and liver cancer, and more preferably lung cancer or kidney cancer. Or not limited thereto.
<105>  <105>
<106> 또한, 본 발명은 약학적으로 유효한 양의 MAP7D2 단백질 발현 또는 활성 억 제제를 개체에 투여하는 단계를 포함하는 암 예방 방법을 제공한다.  In addition, the present invention provides a method for preventing cancer, comprising administering to a subject a pharmaceutically effective amount of an MAP7D2 protein expression or activity inhibitor.
<!07> 또한, 본 발명은 약학적으로 유효한 양의 MAP7D2 단백질 발현 또는 활성 억 제제를 암에 걸린 개체에 투여하는 단계를 포함하는 암 치료 또는 암 전이 억제 방 법을 제공한다. In another aspect, the present invention provides a method for treating cancer or inhibiting cancer metastasis, comprising administering a pharmaceutically effective amount of an MAP7D2 protein expression or active inhibitor to a subject with cancer. Provide the law.
<i08> 상기 약학적으로 유효한 양이란 0.0001 내지 100 mg/kg이고, 바람직하게는  <i08> The pharmaceutically effective amount is 0.0001 to 100 mg / kg, preferably
0.001 내지 10 mg/kg이며, 이에 한정되는 것은 아니다. 투여량은 특정 환자의 체 중, 연령, 성별, 건강상태, '식이, 투여기간, 투여방법, 제거율, 질환의 중증도 등 에 따라 변화될 수 있다. 0.001 to 10 mg / kg, but is not limited thereto. The dosage can be varied according to the body of the specific patient, age, sex, health status, 'diet, administration time, administration method, clearance, such as the severity of the disease.
<109> 상기 MAP7D2 단백질 발현 또는 활성 억제제는 임상 투여 시에 경구 또는 비 경구로 투여가 가능하며 비경구 투여시 복강내주사, 직장내주사, 피하주사, 정맥주 사, 근육내주사, 자궁내 경막주사, 뇌혈관내 주사 또는 흉부내 주사에 의해 투여될 수 있고, 일반적인 의약품 제제의 형태로 사용될 수 있다.  The MAP7D2 protein expression or activity inhibitor may be administered orally or orally during clinical administration and intraperitoneal, rectal, subcutaneous, intravenous, intramuscular, intrauterine It can be administered by injection, cerebrovascular injection, or intrathoracic injection, and can be used in the form of general pharmaceutical preparations.
<110> 상기 암은 신장암, 폐암, 대장암, 난소암, 위암 및 간암으로 구성된 군으로 부터 선택되는 어느 하나인 것이 바람직하고, 폐암 또는 신장암인 것이 더욱 바람 직하나, 이에 한정되지 않는다.  The cancer is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer, and more preferably lung cancer or kidney cancer, but is not limited thereto.
<111> 상기 개체는 척추동물이고 바람직하게는 포유동물이며, 그보다 바람직하게는 쥐, 토끼, 기니아피그, 햄스터, 개, 고양이와 같은 실험동물이고, 가장 바람직하게 는 침팬지, 고릴라와 같은 유인원류 동물이다.  The subject is a vertebrate and preferably a mammal, more preferably an experimental animal such as a rat, rabbit, guinea pig, hamster, dog, or cat, and most preferably anthropoid animals such as chimpanzees or gorillas. to be.
<112>  <112>
<Π3> 본 발명의 구체적인 실시예에서, 본 발명자들은 인체 암 조직의 대규모 유전 자 발현 데이터베이스를 구축하여 데이터마이닝 기법을 통해 신규한 표적화 항암 후보 표적을 발굴하였다. 특히, MAP7D2가 신장암, 폐암, 대장암, 난소암, 위암 및 간암의 조직 및 세포에서 과발현되고, MAP7D2의 억제에 의해 폐암 또는 신장암 세 포의 성장, 이동, 침윤 및 전이가 저해되고 세포사멸이 유도됨으로써, 신규한 항암 제의 표적으로서 MAP7D2의 가능성을 확인하였으므로, MAP7D2 단백질의 발현 또는 활성 억제제는 암의 치료 또는 암전이 억제에 유용하게 이용될 수 있다. In a specific embodiment of the present invention, the present inventors constructed a large-scale gene expression database of human cancer tissue to find a novel targeted anticancer candidate target through data mining techniques. In particular, MAP7D2 is overexpressed in tissues and cells of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer, and inhibition of MAP7D2 inhibits the growth, migration, invasion and metastasis of lung cancer or kidney cancer cells and apoptosis. This induction confirmed the possibility of MAP7D2 as a target of a novel anticancer agent, and therefore inhibitors of the expression or activity of MAP7D2 protein can be usefully used for the treatment of cancer or inhibition of cancer metastasis.
<114> <114>
<Π5> 또한, 본 발명은 MAP7D2에 특이적으로 결합하는 항체 또는 ?유전자에 상보적인 핵산 중 어느 하나 이상을 이용하여 암세포에서의 MAP7D2 발현 수준을 측 정하는 단계를 포함하는, 암의 진단, 치료 결과 확인 또는 예후를 평가하는 방법을 제공한다.  <Π5> In addition, the present invention includes the steps of measuring the expression level of MAP7D2 in cancer cells using at least one of an antibody specifically binding to MAP7D2 or a nucleic acid complementary to the gene, the result of the diagnosis and treatment of cancer It provides a method of evaluating or evaluating prognosis.
<116> 상기 암은 신장암, 폐암, 대장암, 난소암, 위암 및 간암으로 구성된 군으로 부터 선택되는 어느 하나인 것이 바람직하고, 폐암 또는 신장암인 것이 더욱 바람 직하나, 이에 한정되지 않는다.  The cancer is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer, and more preferably lung cancer or kidney cancer, but is not limited thereto.
<Π7> 상기 암 진단 방법에 있어서, 정상 이상으로 상승된 MAP7D2의 발현 검출은 환자가 암에 걸렸음을 의미한다. 또한, 암으로 치료를 받았거나 받고 있는 개체의 진단시료에서 MAP7D2의 정상적인 발현 검출은 암 치료가 성공적임을 의미하고, 상 기 진단시료에서 정상 이상으로 상승된 MAP7D2의 검출은 치료를 계속해야 한다는 것을 의미한다. 아을러, 암에 걸린 개체의 진단시료에서 MAP7D2의 정상적인 발현 검출은 예후가 좋다는 것을 의미하고, 상기 진단시료에서 정상 이상으로 상승된 MAP7D2의 검출은 예후가 좋지 않다는 것을 의미한다. <Π7> In the cancer diagnosis method, detection of expression of MAP7D2 elevated above normal It means that the patient has cancer. In addition, detection of normal expression of MAP7D2 in diagnostic samples of individuals treated or receiving cancer means that the cancer treatment is successful, and detection of elevated MAP7D2 above normal in the diagnostic samples indicates that treatment should be continued. do. In addition, detection of normal expression of MAP7D2 in a diagnostic sample of a subject with cancer means a good prognosis, and detection of MAP7D2 elevated above normal in the diagnostic sample means a poor prognosis.
<118> .  <118>.
<ιΐ9> 또한, 본 발명은 MAP7D2에 특이적으로 결합하는 항체 또는 유전자에 상보적인 핵산 중 어느 하나 이상을 포함하는 암 진단용 키트를 제공한다. In addition, the present invention provides a kit for diagnosing cancer comprising any one or more of nucleic acids complementary to an antibody or gene that specifically binds to MAP7D2.
<120> 상기 암 진단용 키트는 MAP7D2와 반웅하는 하나 이상의 물질 및 반응 생성물 검출용 시약과 이에 대한 지시 사항을 추가적으로 포함할 수 있다. 예를 들어, MAP7D2와 반웅하는 하나 이상의 물질은 MAP7D2의 RNA 또는 DNA에 상보적인 RNA 또 는 DNA, 및 MAP7D2 단백질에 결합하는 항체일 수 있고 반웅 생성물 검출용 시약은 핵산 또는 단백질 표지 및 발색시약일 수 있다. The cancer diagnostic kit may further include one or more substances and reaction product detection reagents and instructions for reacting with MAP7D2. For example, the one or more substances that react with MAP7D2 may be RNA or DNA complementary to the RNA or DNA of MAP7D2, and an antibody that binds to the MAP7D2 protein and the reagent for detecting the reaction product may be a nucleic acid or protein label and a coloring reagent. have.
<121>  <121>
<122> 본 발명의 구체적인 실시예에서, 본 발명에서 구축한, 인체 암 조직의 대규 모 유전자 발현 데이터베이스를 바탕으로 하는 데이터마이닝 기법을 통해 신규한 표적화 항암 후보 표적인 MAP7D2를 선별하였고, 상기 MAP7D2는 신장암, 폐암, 대장 암, 난소암, 위암 및 간암의 조직 및 세포에서 과발현되고, MAP7D2가 억제된 암세 포는 성장 이동, 침윤 및 전이가 저해되고 세포사멸이 유도됨으로써, 이를 통해 MAP7D2가 암세포에 결정적인 역할을 하는 것을 확인하였다. 따라서, MAP7D2의 발 현 수준 확인을 통해 암을 모니터링하거나, 암을 진단할 수 있다.  In a specific embodiment of the present invention, a novel targeting anticancer candidate target MAP7D2 was selected through a data mining technique based on a large gene expression database of human cancer tissue constructed in the present invention. Cancer cells that are overexpressed in tissues and cells of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer, and inhibited MAP7D2 inhibit growth growth, invasion and metastasis, and induce apoptosis, thereby inducing MAP7D2 to cancer cells. It was confirmed to play a decisive role. Therefore, the expression level of MAP7D2 can be monitored and cancer can be diagnosed.
<123>  <123>
<124> 또한, 본 발명은 암의 예방 또는 치료, 또는 암 전이 억제에 사용하기 위한,  In addition, the present invention is for use in the prevention or treatment of cancer or to inhibit cancer metastasis,
MAP7D2 단백질 발현 또는 활성 억제제를 제공한다.  Inhibitors of MAP7D2 protein expression or activity are provided.
<125> 상기 MAP7D2 단백질은 서열번호 1로 기재되는 아미노산 서열을 가지는 것이 바람직하나 이에 한정되지 않는다. The MAP7D2 protein preferably has an amino acid sequence as set forth in SEQ ID NO: 1, but is not limited thereto.
<126> 상기 MAP7D2 단백질의 발현 억제제는 유전자의 mRNA에 상보적으로 결 합하는 안티센스 뉴클레오티드, 작은 간섭 RNA(short interfering RNA) 및 짧은 헤 어핀 RNA(short hairpin RNA)로 이루어진 군으로부터 선택된 어느 하나인 것이 바 람직하나 이에 한정되지 않는다. The inhibitor of expression of the MAP7D2 protein may be any one selected from the group consisting of antisense nucleotides, short interfering RNAs, and short hairpin RNAs, which complementarily bind to mRNA of a gene. Preferred but not limited to.
<127> 상기 MAP7D2 단백질의 활성 억제제는 MAP7D2 단백질에 상보적으로 결합하는 화합물, 펩티드, 펩티드 미메틱스, 앱타머 및 항체로 이루어진 군으로부터 선택된 어느 하나인 것이 바람직하나 이에 한정되지 않는다. The inhibitor of activity of the MAP7D2 protein binds complementarily to the MAP7D2 protein. It is preferably one selected from the group consisting of a compound, a peptide, a peptide mimetics, an aptamer and an antibody, but is not limited thereto.
<128> 상기 암은 신장암, 폐암, 대장암, 난소암, 위암 및 간암으로 구성된 군으로 부터 선택되는 어느 하나인 것이 바람직하고, 폐암 또는 신장암인 것이 더욱 바람 직하나, 이에 한정되지 않는다.  The cancer is preferably any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer, and more preferably lung cancer or kidney cancer, but is not limited thereto.
<129>  <129>
<130> 본 발명의 구체적인 실시예에서, 본 발명자들은 인체 암 조직의 대규모 유전 자 발현 데이터베이스를 구축하여 데이터마이닝 기법을 통해 신규한 표적화 항암 후보 표적을 발굴하였다. 특히, MAP7D2가 신장암, 폐암, 대장암, 난소암, 위암 및 간암의 조직 및 세포에서 과발현되고, MAP7D2의 억제에 의해 폐암 또는 신장암 세 포의 성장, 이동, 침윤 및 전이가 저해되고 세포사멸이 유도되므로, MAP7D2 단백질 의 발현 또는 활성 억제제는 암의 예방 또는 치료, 또는 암 전이 억제에 유용하게 이용될 수 있다.  In a specific embodiment of the present invention, the inventors constructed a large-scale gene expression database of human cancer tissue to discover novel targeted anticancer candidate targets through data mining techniques. In particular, MAP7D2 is overexpressed in tissues and cells of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer, and inhibition of MAP7D2 inhibits the growth, migration, infiltration and metastasis of lung cancer or kidney cancer cells, and apoptosis. Because of this induction, inhibitors of the expression or activity of the MAP7D2 protein can be usefully used for preventing or treating cancer or inhibiting cancer metastasis.
<131>  <131>
<132> 아울러, 본 발명은 암 진단용 키트에 사용하기 위한, MAP7D2에 특이적으로 결합하는 항체 또는 유전자에 상보적인 핵산을 제공한다.  In addition, the present invention provides a nucleic acid complementary to an antibody or gene that specifically binds to MAP7D2 for use in a kit for cancer diagnosis.
<133> 상기 암 진단용 키트는 MAP7D2와 반웅하는 하나 이상의 물질 및 반웅 생성물 검출용 시약과 이에 대한 지시 사항을 추가적으로 포함할 수 있다. 예를 들어, MAP7D2와 반웅하는 하나 이상의 물질은 MAP7D2의 RNA 또는 DNA에 상보적인 RNA 또 는 DNA, 및 MAP7D2 단백질에 결합하는 항체일 수 있고 반웅 생성물 검출용 시약은 핵산 또는 단백질 표지 및 발색시약일 수 있다.  The cancer diagnostic kit may further include one or more substances and reaction products for detecting the reaction with MAP7D2 and instructions thereof. For example, the one or more substances that react with MAP7D2 may be RNA or DNA complementary to the RNA or DNA of MAP7D2, and an antibody that binds to MAP7D2 protein, and the reagent for detecting the reaction product may be a nucleic acid or protein label and a coloring reagent. have.
<134>  <134>
<135> 본 발명의 구체적인 실시예에서, 본 발명에서 구축한, 인체 암 조직의 대규 모 유전자 발현 데이터베이스를 바탕으로 하는 데이터마이닝 기법을 통해 신규한 표적화 항암 후보 표적인 MAP7D2를 선별하였고, 상기 MAP7D2는 신장암, 폐암, 대장 암, 난소암, 위암 및 간암의 조직 및 세포에서 과발현되고, MAP7D2가 억제된 암세 포는 성장, 이동, 침윤 및 전이가 저해되고 세포사멸이 유도됨으로써, 이를 통해 MAP7D2가 암세포에 결정적인 역할을 하는 것을 확인하였다. 따라서, MAP7D2에 특 이적으로 결합하는 항체 또는 / 유전자에 상보적인 핵산은 암 진단용 키트에 유용하게 사용될 수 있다.  In a specific embodiment of the present invention, a novel targeting anticancer candidate target MAP7D2 was selected through a data mining technique based on a large gene expression database of human cancer tissue constructed in the present invention. Cancer cells that are overexpressed in tissues and cells of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer, and inhibit MAP7D2 growth, migration, invasion and metastasis, and induce apoptosis, thereby inhibiting MAP7D2 from cancer cells. It was confirmed that it plays a decisive role in. Thus, nucleic acids complementary to antibodies or / genes that specifically bind to MAP7D2 can be usefully used in cancer diagnostic kits.
<136>  <136>
【발명의 실시를 위한 형태】 이하, 본 발명을 실시예 및 제조예에 의하여 상세히 설명한다. [Form for implementation of invention] Hereinafter, the present invention will be described in detail by way of examples and preparation examples.
단, 하기 실시예 및 제조예는 본 발명을 구체적으로 예시하는 것이며, 본 발 명의 내용이 상기 실시예 및 제조예에 의해 한정되는 것은 아니다.  However, the following Examples and Preparation Examples specifically illustrate the present invention, and the contents of the present invention are not limited to the Examples and Preparation Examples.
<실시예 1> 데이터베이스 구축 및 암유발유전자중독 (oncogene addiction) 을 보이는 신규한 항암제 표적 유전자의 선별 Example 1 Screening of a Novel Anticancer Target Gene Showing Database Construction and Oncogene Addiction
<1-1> 대규모 인체 암 조직의 유전자 발현 데이터베이스  <1-1> Gene Expression Database of Large-scale Human Cancer Tissues
NCBI의 Gene Expression Omnibus 및 EBI의 ArrayExpress의 공개된 유전자 발 현 데이터베이스로부터 450가지 이상의 데이터 세트를 수집 및 분석하여 하기 표 1 에 나타낸 바와 같이, 33,000개 이상의 시료를 포함하는 차별화된 유전자 발현 데 이터베이스를 구축하였다. 구체적으로는, 서로 다른 데이터 세트들을 통합하여 분 석할 수 있도록 가능한 한 마이크로어레이 원시 데이터 파일로부터 일관된 방법으 로 데이터들을 통합 및 정리하였다. 첫 번째로, Affymetrix human chip platform 을 사용한 데이터들만 수집하였고, 두 번째로, 원시 데이터인 CEL 파일로부터 Affymetrix 고유 알고리즘인 MAS5(microarray suite 5) 방법을 적용하여, 각 유전 자 수준에서의 발현값을 구하였고, 전반적인 평균 표준화 (global mean normalization)를 동일하게 적용하여, 각 마이크로어레이 플랫폼 (microarray platform, U133Plus2)으로 10,000개 이상의 시료를 하나로 통합하여 메타 분석하는 것이 가능하도록 하였다.  Collected and analyzed more than 450 data sets from NCBI's Gene Expression Omnibus and EBI's ArrayExpress published database to generate differentiated gene expression databases comprising more than 33,000 samples, as shown in Table 1 below. Built. Specifically, we have consolidated and organized the data in a consistent way from the microarray raw data file as much as possible to integrate and analyze different data sets. First, only data using the Affymetrix human chip platform was collected. Second, the expression value at each gene level was determined by applying Affymetrix's unique algorithm, the microarray suite 5 (MAS5) method, from the original CEL file. By applying the global mean normalization in the same way, each microarray platform (U133Plus2) was able to integrate more than 10,000 samples into one and perform meta-analysis.
【표 1】 Table 1
인체 시료 유전자 발현 데이터베이스 현황  Human sample gene expression database
Figure imgf000017_0001
Figure imgf000017_0001
<145>  <145>
<146> 상기 데이터베이스에는 하기 표 2에 나타낸 바와 같이, 거의 모든 조직으로 부터 유래한, 22 ,000개 이상의 정상 및 암 조직에 대한 유전자 발현 데이터를 포함 하고 있다.  The database contains gene expression data for more than 22,000 normal and cancerous tissues from almost all tissues, as shown in Table 2 below.
<147>  <147>
<148> 【표 2] 조직 π 정상 합계 <148> [Table 2] Tissue π normal sum
방광 (Bladder) 126 23 149  Bladder 126 23 149
험액 (Blood) 1416 1127 2543  Blood 1416 1127 2543
골수 (Bone Marrow) 1736 5 1741  Bone Marrow 1736 5 1741
뇌 (Brain) 1285 2239 3524  Brain 1285 2239 3524
유방 (Breast) 3738 158 3896  Breast 3738 158 3896
자궁경부 (Cervix 138 46 184  Cervical (Cervix 138 46 184
대장 (Colon) 1116 231 1347  Colon 1116 231 1347
자궁내막 (Endomet r i urn ) 84 81 165  Endomet i urn 84 81 165
식도 (Esoohagus) 32 32 64  Esoohagus 32 32 64
두경부 (Head Neck) 253 42 295  Head Neck 253 42 295
신장 (Kidnev) 928 166 1094  Kidnev 928 166 1094
간 (Liver ) 354 83 437  Liver 354 83 437
폐 (L匿) 1290 426 1716  Lung (L 匿) 1290 426 1716
림프점 (Lvnrnh node) 25 12 37  Lvnrnh node 25 12 37
림프구 ( LvmDhocvt e ) 180 175 355  Lymphocyte (LvmDhocvt e) 180 175 355
근육 (Muscle) 0 622 622  Muscle 0 622 622
난소 (Ovarv) 977 17 994  Ovarv 977 17 994
췌장 (Pancreas) 94 35 129  Pancreas 94 35 129
저립선 (Prostate) 473 188 661  Prostate 473 188 661
피부 (Skin) 437 80 517  Skin 437 80 517
소장 (Small intestine) 14 7 21  Small intestine 14 7 21
비장 (SDleen) 4 10 14  Spleen 4 10 14
위장 (Stomach) 295 61 356  Stomach 295 61 356
정소 (Test is) 206 26 232  Test is 206 26 232
갑상선 (Thvroid) 130 52 182  Thyroid (Thvroid) 130 52 182
자궁 (Uterus) 130 25 155  Uterus 130 25 155
기타 410 268 678  Others 410 268 678
합계 15454 6661 22115  Total 15454 6661 22115
<149>  <149>
<150> <1-2> 암 이상값 프로파일 분석 (Cancer Outlier Profile Analysis, COPA)을 사용한 암 환자에서의 유전자 발현변화 확인  <150> <1-2> Identification of Gene Expression Changes in Cancer Patients Using Cancer Outlier Profile Analysis (COPA)
<151> 상기 실시예 <1-1>과 같이, 독자적으로 구축한 암 유전자 발현 데이터베이스 로부터 치료 표적을 발굴하기 위하여 기존의 접근법과는 차별화된 방법으로 C0PA 분석을 적용하였다 (Tomlins et al . 2005) . 현재 임상에서 효과적으로 쓰이는 치료 제의 표적화 항암 표적 유전자들은 전체 환자 중 일부에서만 유전자 돌연변이 또는 유전자 증폭에 의한 과발현이 관찰되고 있다. 예컨대, 허셉틴 (Herceptin)의 표적 인 ERBB2는 전체 유방암 환자 증 약 25~30¾ 정도에서만 과발현되는 것으로 나타났 고, 이레사 (Iressa) 또는 세룩시맵 (Cetuximab)의 표적인 EGFR도 폐암 환자나 뇌종 양 환자의 약 10-20% 정도에서만 유전자 돌연변이 또는 유전자 증폭에 의한 과발현 이 관찰된다. 이와 같이, 효과적인 표적 유전자들은 전체 환자 중 일부 환자에서 만 변이가 관찰되므로, 기존의 t-test와 같이 환자군과 정상인들의 평균을 비교하 여 발현에 차이가 나는 유전자를 찾는 방법보다는 환자군 중 상위 10~20%와 정상인 들의 평균을 비교하는 outlier 분석 방법이 더욱 효과적이므로, 이러한 암 이상값 프로파일 분석 (Cancer Outlier Profile Analysis, COPA)를 실시하였다. As described in Example <1-1>, C0PA analysis was applied in a method different from the conventional approach to discover therapeutic targets from a cancer gene expression database constructed independently (Tomlins et al. 2005). . Currently, the targeted anticancer target genes of therapeutic agents effectively used in the clinic are overexpressed by gene mutation or gene amplification in only a part of the patients. For example, ERBB2, a target of Herceptin, was found to be overexpressed in only about 25-30¾ of breast cancer patients, and EGFR, a target of Iressa or Celuximab, was also used in patients with lung cancer or brain tumors. Overexpression by gene mutation or gene amplification is observed only in about 10-20% of the population. As such, effective target genes may be present in some patients. Since only the variation is observed, the outlier analysis method compares the average of the top 10-20% of the patient group with the average of the normal person, rather than comparing the average of the patient group and the normal person to find genes with different expressions as in the conventional t-test. Since it is effective, this cancer outlier profile analysis (COPA) was performed.
<152>  <152>
<153> <1-3>신규한 표적화 치료 표적 유전자의 선별  <1-3> Screening of New Targeted Therapeutic Target Genes
<i54> C0PA 분석법을 적용하여 의약품이 될 만한 (druggable) 표적인 수용체, 키나 제, 수송체, 및 채널 단백질들 중에서 각 암에서 효과적인 표적화 치료의 표적이 될 수 있는 유전자들을 선별하였다. 현재까지 임상 시험에 들어가 있거나 주로 시 판되고 있는 치료용 약물 표적들은 수용체, 채널 수송체, 키나제 및 세포부착분자 (Cell adhesion molecules, CAMs) 등과 같은 세포표면분자들이 대부분이므로, 주로 이러한 것들올 위주로 분석하여 후보 표적 유전자를 발굴하였다. 후보 표적 유전 자는 하기와 같은 기준으로 발굴하였다. i ) 임상 조직에 대한 유전자 발현 데이 터베이스 마이닝을 통해 정상조직에 비해 특정 암 조직에서 특이적으로 많이 발현 되고 있는 후보 표적 유전자의 풀 (pool)을 형성하였다. ii) 형성된 유전자 풀 내 에서 표적 유전자가 되도록 여러 암에서 특이적으로 과발현되어 있는 유전자를 선 별하는데, 이는 여러 암에서 특정 표적 유전자가 많이 발현되어 있을수록 암 형성 에 직접적이고도 중요한 유전자일 가능성이 높기 때문이다. iii) 표적 유전자가 많 이 발현되고 있는 임상 조직과 동일한 조직에서 유래한 암 세포주들 중에서 그 표 적 유전자가 일정 수준이상 많이 발현되고 있는 세포주를 선별하였다. iv) 선별된 후보 표적 유전자들에 대해 문헌 조사를 수행하여 되도록 암 관련하여 연구가 많이 진행되어 있지 않은 유전자를 선별하여 신규한 중독되는 암 유발 유전자 (addicted oncogene)를 찾고자 하였다. 이와 같은 기준을 바탕으로 후보 표적 유전자들 중에 서 MAP7D2를 표적화 항암 치료를 위한 일차 후보 표적 유전자로서 선택하여 검증하 였다.  <i54> The C0PA assay was applied to select genes that could be targeted for effective targeting therapy in each cancer from among drug-targeting target receptors, kinases, transporters, and channel proteins. Therapeutic drug targets currently in clinical trials or mainly on the market are mostly cell surface molecules such as receptors, channel transporters, kinases and cell adhesion molecules (CAMs). Candidate target genes were identified. Candidate target genes were identified based on the following criteria. i) Gene expression database mining for clinical tissues formed a pool of candidate target genes that are expressed more specifically in specific cancer tissues than in normal tissues. ii) Select genes that are specifically overexpressed in several cancers to become target genes in the gene pool formed. The more expression of a specific target gene in various cancers, the more likely it is to be a direct and important gene for cancer formation. Because. iii) Among cancer cell lines derived from the same tissues as the clinical tissues in which the target genes are expressed a lot, the cell lines in which the target genes were expressed at a certain level or more were selected. iv) A literature search was conducted on selected candidate target genes to find new addicted oncogenes by selecting genes that have not been studied as much as possible. Based on these criteria, MAP7D2 was selected and validated as the primary candidate target gene for targeted chemotherapy among candidate target genes.
<155> 그 결과, 도 1에 나타낸 바와 같이, 암 환자의 임상조직에서 MAP7D2의 발현 양상을 확인한 결과, 신장암 (kidney), 폐암 (lung), 대장암 (colon), 간암 (liver), 난소암 (ovary) 및 위암 (stomach) 등의 암 조직에서 정상조직에 비해 MAP7D2의 발현 이 증가되어 있음을 관찰하였으며, 이를 통해 MAP7D27} 암 치료에 효과적인 치료 표적이 될 수 있음을 알 수 있었다 (도 1).  As a result, as shown in FIG. 1, as a result of confirming the expression pattern of MAP7D2 in the clinical tissues of cancer patients, kidney cancer, lung cancer, colon cancer, liver cancer, and ovary It was observed that the expression of MAP7D2 was increased in cancer tissues such as cancer and stomach cancer compared to normal tissues, and thus, MAP7D27} could be an effective therapeutic target for the treatment of cancer (FIG. 1). ).
<156>  <156>
<157> <실시예 2> MAP7D2의 다양한 암에서의 발현량 확인 <i58> <2-l> 암 세포주에서 마이크로어레이 플랫폼을 통한 MAP7D2 발현량의 확인Example 2 Confirmation of Expression of MAP7D2 in Various Cancers <i58><2-l> Identification of MAP7D2 Expression Levels through Microarray Platform in Cancer Cell Lines
<159> 다양한 암 세포주에서 MAP7D2의 발현 양상을 마이크로어레이 플랫품 Microarray Platforms for Expression of MAP7D2 in Various Cancer Cell Lines
U133plus2A 유전자 칩을 이용하여 분석하였다.  Analysis was performed using the U133plus2A gene chip.
<160> 그 결과, 도 2에 나타낸 바와 같이, MAP7D2의 임상조직에서의 발현양상은, 도 1의 암 조직에서 확인한 결과와 일치하게 신장암, 폐암, 대장암, 난소암, 위암 및 간암에서 유래한 암세포주들에서 비교적 높게 발현됨을 알 수 있었다 (도 2). As a result, as shown in Figure 2, the expression pattern in the clinical tissue of MAP7D2 is derived from kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer and liver cancer in accordance with the results confirmed in the cancer tissue of Figure 1 It can be seen that they are relatively high in one cancer cell line (FIG. 2).
<161> 그리고, 이러한 도 1 및 도 2의 결과를 토대로 하여, 실험에 사용할 암 세포 주를 선별하기 위해, 각각의 세포주에 대한 유전자 발현 데이터베이스를 바탕으로 선별한 각 표적 암 별로 MAP7D2를 가장 잘 발현하는 세포주들을 확인하였으며, 이 를 하기 [표 3]에 나타내었다. 1 and 2, MAP7D2 is best expressed for each target cancer selected based on the gene expression database for each cell line, in order to select cancer cell lines to be used in the experiment. The cell lines were confirmed, which are shown in Table 3 below.
<162>  <162>
<163> 【표 3】  <163> [Table 3]
Figure imgf000020_0001
Figure imgf000020_0001
<164>  <164>
<165> <2-2> 암세포주에서 역전사 중합효소연쇄반웅 (Reverse Tanscription- <165> <2-2> Reverse Transcription Polymerase Chain Reaction in Cancer Cell Lines
Polymerase Chain Reaction, RT-PCR)을 통한 MAP7D2 발현량의 확인 Confirmation of MAP7D2 Expression by Polymerase Chain Reaction (RT-PCR)
<i66> MAP7D2가 다른 암에 비해 과발현되어 있던 대장암, 폐암 및 간암 세포주에서  <i66> In colorectal, lung and liver cancer cell lines where MAP7D2 was overexpressed compared to other cancers
MAP7D2의 발현 양상을 각각 확인하기 위하여 RT-PCR을 수행하였다.  RT-PCR was performed to confirm the expression patterns of MAP7D2, respectively.
<167> 구체적으로, 11종류의 다양한 암 세포주 (대장암 4종류, 폐암 4종류,간암 3종 류)에서 easy-BLUE™ Total RNA Extraction kit (Sol Gent, Cat #: 17061)으로 총Specifically, 11 types of cancer cell lines (4 types of colorectal cancer, 4 types of lung cancer, and 3 types of liver cancer) were used in the easy-BLUE ™ Total RNA Extraction kit (Sol Gent, Cat #: 17061).
RNA를 분리한 다음, 각각의 총 RNA 2//g으로, DiaStar RT kit(SolGent, Cat#:DR13- R10K)를 사용하여 cDNA를 합성하였다. cDNA를 멸균 증류수로 회석하여 2% cDNA를 제조한 후 10 pmole Taq DNA 폴리머라제 (polymerase) (한국 솔젠트사) 및 프라이 머 [정방향 프라이머 (Forward pr imer ): 5 ' -CTCGAGAGAACAGATTATG-3 ', 서열번호 2; 역 방향 프라이머 (Reverse primer): 5'- CTCACTTGTGGAGACACATC-3 ' , 서열번호 3]를 사 용하여 RT-PCR을 수행하였다. 이 때, 겔 로딩 대조군 (Gel loading control)으로써 GAPDH의 RT-PCR을 수행하였다 이용된 PCR프로그램은 하기 [표 4]와 같았다: 【표 4】 After RNA was isolated, cDNA was synthesized using a DiaStar RT kit (SolGent, Cat #: DR13-R10K) with each total RNA 2 / g. cDNA was distilled with sterile distilled water to prepare 2% cDNA, followed by 10 pmole Taq DNA polymerase (Solgent Korea) and primer [Forward primer: 5 '-CTCGAGAGAACAGATTATG-3', sequence No. 2; Reverse primer: RT-PCR was performed using 5'-CTCACTTGTGGAGACACATC-3 ', SEQ ID NO: 3]. At this time, as a gel loading control RT-PCR of GAPDH was performed. The PCR programs used were as shown in Table 4 below.
Figure imgf000021_0001
Figure imgf000021_0001
<170>  <170>
<i7i> 그 결과, 도 3 내지 도 5에 나타낸 바와 같이, 상기 [표 3]의 결과와 유사하 게, MAP7D2 유전자가 대장암, 폐암 및 간암 등의 세포주들에서 높게 발현되어 있음 을 확인하였다 (도 3 내지 도 5).  <i7i> As a result, as shown in Figures 3 to 5, it was confirmed that the MAP7D2 gene is highly expressed in cell lines such as colorectal cancer, lung cancer and liver cancer as shown in Table 3 above ( 3 to 5).
<172>  <172>
<173> <2-3> 암 조직에서 역전사 중합효소연쇄반웅을 통한 MAP7D2 발현량 확인 <173> <2-3> Confirmation of MAP7D2 Expression by Reverse Transcriptase Polymerase Chain Reaction in Cancer Tissues
<174> 폐암 및 간암 조직에서 MAP7D2의 발현 양상을 확인하기 위하여, 폐암 또는 간암 환자의 정상 조직과 암 조직을 각각 분리하여 , RT-PCR을 수행하였다.In order to confirm the expression pattern of MAP7D2 in lung cancer and liver cancer tissues, normal tissues and cancer tissues of lung cancer or liver cancer patients were separated from each other, and RT-PCR was performed.
<175> 구체적으로 , 폐암, 간암의 인체자원은 부산 대학교 병원 한국 인체 자원 거 점은행으로부터 공급받아 사용하였다. 각 조직을 액체질소를 사용하여 얼린 상태 에서 막자사발을 이용하여 잘게 분쇄하였다. 분쇄한 조직을 RNeasyMini kit (Qiagene, Cat #: 74104)을 사용하여 명시된 방법대로, 총 RNA를 분리한 다음, 각 각의 총 RNA 2으로, DiaStar RT kit(SolGent, Cat#:DR13-R10K)를 사용하여 cDNA를 합성하였다. cDNA를 멸균 증류수로 회석하여 2% cDNA를 제조한 후 10 pmole/ Taq DNA 폴리머라제 (polymerase) (한국 솔젠트사) 및 프라이메정방향 프라이머 (Forward primer): 5 ' -CTCGAGAGAACAGATTATG-3 ' , 서열번호 2; 역방향 프라이머 (Reverse primer): 5'- CTTCACTTGTGGAGACACATC-3 ', 서열번호 3]를 사용하여 RT-PCR을 수행하 였다. 이 때, 겔 로딩 대조군 (Gel loading control)으로써 β -act in의 RT-PCR을 수행하였으며, 이용된 PCR프로그램은 상기 [표 4]와 같다. Specifically, the human resources of lung cancer and liver cancer were supplied from the Korea Human Resource Base Bank of Pusan National University Hospital. Each tissue was crushed finely using a mortar and pestle while frozen with liquid nitrogen. Using the RNeasyMini kit (Qiagene, Cat #: 74104), the pulverized tissue was isolated from the total RNA, and then the total amount of RNA 2 and the DiaStar RT kit (SolGent, Cat #: DR13-R10K). CDNA was synthesized. cDNA was distilled with sterile distilled water to prepare 2% cDNA, followed by 10 pmole / Taq DNA polymerase (Solgent, Korea) and primer primer: 5 '-CTCGAGAGAACAGAGATTATG-3', SEQ ID NO: 2; Reverse primer: RT-PCR was performed using 5'-CTTCACTTGTGGAGACACATC-3 ', SEQ ID NO: 3]. At this time, RT-PCR of β-act in was performed as a gel loading control, and the PCR program used is shown in Table 4 above.
<176> 그 결과, 정상 조직과 비교하였을 때, 폐암 조직에서는 ΈΆ 이상, 간암 조직 에서는 33% 이상 MAP7D2가높게 발현되어 있음을 확인하였다 (도 6 및 도 7).As a result, it was confirmed that MAP7D2 was highly expressed in lung cancer tissues and more than 33% in liver cancer tissues when compared with normal tissues (FIGS. 6 and 7).
<177> <177>
<178> <실시예 3> MAP7D2 유전자 발현 억제의 암세포 성장에 대한 영향 분석 Example 3 Analysis of the Effect of MAP7D2 Gene Expression Inhibition on Cancer Cell Growth
<i79> MAP7D2를 가장 잘 발현하고 있는 세포주 중에서 비교적 빨리 자라고 전이력 이 높은, 폐암 세포주인 NCI-H1703 및 신장암인 A498 세포주를 사용하여 MAP7D2 녹 다운 (knockdown) 방법을 이용하여 MAP7D2 유전자의 발현을 억제하고, MAP7D2 유전 자 발현 억제에 의한 암 세포 성장에 대한 영향을 분석하였다. <i79> MAP7D2 rust using the lung cancer cell line NCI-H1703 and kidney cancer A498 cell line, which grow relatively quickly among the cell lines that express MAP7D2 best, and have high metastasis. The knockdown method was used to inhibit MAP7D2 gene expression and to analyze the effect on cancer cell growth by MAP7D2 gene expression inhibition.
<180>  <180>
<i8i> <3-1> MAP7D2 유전자 발현 억제의 신장암 세포 성장에 대한 영향  <i8i> <3-1> Effects of MAP7D2 Gene Expression Inhibition on Kidney Cancer Cell Growth
<i82> MAP7D2 유전자 발현 억제가 신장암 세포의 성장에 어떠한 영향을 미치는지 확인하기 위하여, MAP7D2 siRNA를 이용하였다.  <i82> MAP7D2 siRNA was used to determine how MAP7D2 gene expression inhibition affects the growth of kidney cancer cells.
<183> 구체적으로, 2개의 MAP7D2 siRNA(#l Duplex; 5'- Specifically, two MAP7D2 siRNAs (#l Duplex; 5′-)
GCAGGAGAUGUUGGGAAAGAA/UUCUUUCCCAACAUCUCCUGC-3 ' , 서열번호 4 및 #2: Duplex; 5 ' -CCCAAAUUAUUAGCUCGGAAU/AUUCCGAGCUAAUAAUUUGGG-3 ' , 서열번호 5)를 바이오니아에 서 구입하여 이용하였다. 상기 언급한 siRNA는 대조군인 control siRNA(Bioneer CAT#: SN_1002)와 Amaxa를 함께 사용한 전기천공법을 통해 신장암 세포주 A498에 도입되었으며, 전기천공법을 수행한 지 24시간 후, MAP7D2의 발현 저해 정도에 따 른 세포의 성장을 대조군과 비교하여 조사하였다. 세포 증식을 조사하기 위해, 우 선 1~4 X 104의 세포를 24-웰 플레이트 (plate)에 분주하여 37°C/5% C02의 조건에서 배양한 후, 각각 48 시간 동안 배양하였으며, 배양이 끝난 세포를 회수하여 상기 < 실시예 2-2>에 기재된 RT-PCR 방 을 이용하여 신장암 세포에서 MAP7D2 siRNA에 의 한 MAP7D2 단백질의 발현 억제를 확인하였다. GCAGGAGAUGUUGGGAAAGAA / UUCUUUCCCAACAUCUCCUGC-3 ', SEQ ID NO: 4 and # 2: Duplex; 5'-CCCAAAUUAUUAGCUCGGAAU / AUUCCGAGCUAAUAAUUUGGG-3 ', SEQ ID NO: 5) was purchased from Bioneer and used. Said siRNA was introduced into renal cancer cell line A498 by electroporation using control siRNA (Bioneer CAT #: SN_1002) and Amaxa as a control group, and the inhibition of MAP7D2 expression was observed 24 hours after electroporation. Cell growth according to the comparison was investigated compared to the control. In order to examine cell proliferation, firstly, cells of 1-4 × 10 4 were dispensed into 24-well plates, incubated at 37 ° C / 5% C0 2 , and incubated for 48 hours each. The cultured cells were recovered, and the inhibition of expression of MAP7D2 protein by MAP7D2 siRNA was confirmed in kidney cancer cells using the RT-PCR method described in Example 2-2.
<184> 또한, 정해진 시간이 경과한 후, 각각의 플레이트에 Cell Counting Kit-In addition, after a predetermined time elapses, each plate has a Cell Counting Kit-
8(CCK-8,D0JIND0, Cat#: CK04-11) 용액 50 ul를 첨가하여 1시간 정도 어두운 상태 에서 37 C02 인큐베이터에서 반웅시킨 다음, 100 ul씩 96 웰 플레이트로 옮기고, Add 50 ul of 8 (CCK-8, D0JIND0, Cat #: CK04-11) solution and react in a 37 C0 2 incubator in the dark for 1 hour, then transfer 100 ul into a 96 well plate.
Victor microplate reader에서 450nm로 흡광도를 측정하여 세포 증식 정도를 관찰 하였다. The degree of cell proliferation was observed by measuring absorbance at 450 nm in Victor microplate reader.
<185> 그 결과, 도 6에 나타낸 바와 같이, 대조군에 비해 siRNA #2를 500nM 처리한 경우, 내부 대조군 (internal control)으로서 사용한 GAPDH에 대비하여 뚜렷한 MAP7D2 발현 억제효과를 나타냈다 (도 8).  As a result, as shown in FIG. 6, when siRNA # 2 was treated with 500 nM compared to the control group, the inhibitory effect of MAP7D2 expression was clearly observed in comparison with GAPDH used as an internal control (FIG. 8).
<186> 또한, 도 7에서 나타낸 바와 같이, 대조군으로 사용된 control siRNA와 비교 하여 MAP7D2 siRNA로 그 발현이 저해된 경우, 신장암 세포주 A498에서는 48시간 후 , 약 60% 정도의 뚜렷한 성장의 저해 및 세포가사멸됨을 확인하였다 (도 9). In addition, as shown in FIG. 7, when the expression of the MAP7D2 siRNA was inhibited compared to the control siRNA used as a control, in the renal cancer cell line A498 after 48 hours, the inhibition of marked growth of about 60% and It was confirmed that the cells were killed (FIG. 9).
<187> <187>
<i88> <3-2> MAP7D2 유전자 발현 억제의 폐암 세포 성장에 대한 영향  <i88> <3-2> Effect of MAP7D2 Gene Expression Inhibition on Lung Cancer Cell Growth
<i89> MAP7D2 유전자 발현 억제가 폐암 세포의 성장에 어떠한 영향을 미치는지 확 인하기 위하예 MAP7D2 shRNA를 이용하였다. <i89> How MAP7D2 Gene Expression Inhibition Affects Lung Cancer Cell Growth Example MAP7D2 shRNA was used to cut.
<190> 구체적으로, TRC 렌티바이러스 플라스미드 백터 (lenticiral plasmid vecotr) pLKO.l-puro(Sigma)에 있는 MAP7D2 작은 헤어핀 (small-hairpin) RNA 클론들은Specifically, the MAP7D2 small-hairpin RNA clones in the TRC lenticiral plasmid vecotr pLKO.l-puro (Sigma)
Sigma사의 MISSION™ shRNA clones 1 ibraries (SIGMA, Cat#: SHCLNG-應 _ 152780) 중에 서 구입하여 사용하였다. 이것은 4종류의 shRNA 컨스트럭트 (construct) 세트로 구 성되어 있으며, 각각의 TRC 번호 TRCN0000108390, TRCN0000108391 , TRCN0000108392 및 TRCN0000108394로, 각각 MAP7D2 shRNA#l부터 #4로 명명하여 사용하였다. 렌티 바이러스 (Lentiviral) MAP7D2 shRNA와 더불어, 음성 대조군으로 Nontarget shRNA Control Transduction part icles(SIGMA, cat#: SHC002V)을 사용하였고, 형질도입 효율의 확인과 shRNA 전달을 최적화하기 위한 양성 대조군으로는 MISS0N TurboGFP Control Transduction part icles(SIGMA, Cat#: SHC003V)을 함께 사용하였다. 이It was purchased from Sigma's MISSION ™ shRNA clones 1 ibraries (SIGMA, Cat #: SHCLNG- 應 152780). It consists of four sets of shRNA constructs, named TRCN0000108390, TRCN0000108391, TRCN0000108392, and TRCN0000108394, respectively, and used MAP7D2 shRNA # 1 to # 4. In addition to the Lentiviral MAP7D2 shRNA, a nontarget shRNA Control Transduction part icles (SIGMA, cat #: SHC002V) was used as a negative control, and the MISS0N TurboGFP Control was used as a positive control to confirm transduction efficiency and optimize shRNA delivery. Transduction part icles (SIGMA, Cat #: SHC003V) were used together. this
- TM 、 、 TO 푸, Lipofectamine 2000(invitrogen, Cat#: 11668— 027) 및 Enhancer Q-TM 、 、 TO Fu , Lipofectamine 2000 (invitrogen, Cat #: 11668—027) and Enhancer Q
Transfection Enhancing Reagent (We lGene, Cat#: TR001-02)을 사용하여 293T에 상 기 shRNA를 형질감염하여 바이러스 입자를 제조하였다. 48 시간 뒤 , 바이러스 배 양 상층액을 회수하여 syringe f i lter(mi 11 ipore,Cat#: SLHP033RS)로 여과한 다음, 신선한 배양 배지와 1:1로 흔합하였고, 여기에 감염의 효과를 향상시키기 위하여, 4 yg/niL의 polybrene(SIGMA, Cat#:L107689)을 넣어 폐암 세포주 NCI-H1703과 신장 암 세포주 A498에 각각 형질감염하였다. 16시간 후, 신선한 배지로 교체한 다음, 48시간 동안 배양하고, MAP7D2 shRNA가 형질감염된 NCI-H1703 또는 A498 세포를 선 별하기 위하여 2 yg/ml의 퓨로마이신 (puromycin)으로 3일 동안 처리하여 선별하였 다. 이때, 선별 대조군으로 shRNA가 형질도입되지 않은 각각의 암 세포주에 동량 의 퓨로마이신 (puromycin)을 처리하여 관찰하였다. 퓨로마이신 (puromycin) 선별 후, 배양이 끝난 세포를 회수하여 상기 <실시예 2-2〉에 기재된 RT-PCR 방법을 이용 하여 발현 저해 여부를 확인하고, MAP7D2 발현 저해 정도에 따른 세포 성장률을 대 조군과 비교하여 조사하였다. Virus particles were prepared by transfecting the shRNA into 293T using Transfection Enhancing Reagent (WelGene, Cat #: TR001-02). After 48 hours, the virus culture supernatant was recovered and filtered with syringe filter (mi 11 ipore, Cat #: SLHP033RS), and then mixed 1: 1 with fresh culture medium, in order to improve the effect of infection. 4 yg / niL polybrene (SIGMA, Cat #: L107689) was added and transfected into lung cancer cell lines NCI-H1703 and kidney cancer cell line A498, respectively. After 16 hours, replaced with fresh medium, incubated for 48 hours, and screened by treatment with 2 yg / ml puromycin for 3 days to select NCI-H1703 or A498 cells transfected with MAP7D2 shRNA. It was. At this time, as a selection control, each cancer cell line that was not transduced with shRNA was observed by treating the same amount of puromycin (puromycin). After puromycin screening, the cultured cells were recovered and the expression was inhibited using the RT-PCR method described in Example 2-2, and the cell growth rate according to the inhibition of MAP7D2 expression was controlled. It was investigated in comparison with.
<i i> 그 결과, 도 10에 나타낸 바와 같이, 대조군에 비해 shMAP7D2 #1 내지 #4모 두, 내부 대조군 (internal control)으로서 사용한 β-액틴 (actin)에 대비하여 뚜렷 한 MAP7D2 발현 억제효과를 나타냈다 (도 10).  As a result, as shown in FIG. 10, all of shMAP7D2 # 1 to # 4 compared to the control group showed clear MAP7D2 expression inhibition effect compared to β-actin used as an internal control. (FIG. 10).
<192> 또한, 도 11 및 도 12에 나타낸 바와 같이, 폐암 세포주 I I-H1703에 4종류 의 MAP7D2 shRNA(#l~#4) 각각을 형질감염한 경우, shMAP7D2 #2 또는 #3을 형질감염 한 폐암 세포주에서 대조군 (Nontarget shControlRNA)에 비해 세포 성장이 현저하 게 감소하였고, 세포 사멸이 증가함을 확인하였다 (도 11). 특히, MAP7D2 shRNA #2 는 5일 동안 대조군에 비해 약 70%정도의 성장 저해 및 세포사멸 효과를 나타냄을 확인하였다 (도 12). In addition, as shown in FIGS. 11 and 12, when each of four types of MAP7D2 shRNAs (#l to # 4) were transfected into lung cancer cell line I I-H1703, shMAP7D2 # 2 or # 3 was transfected. In one lung cancer cell line, cell growth was markedly decreased and cell death was increased compared to the control group (Nontarget shControlRNA) (FIG. 11). In particular, MAP7D2 shRNA # 2 Was confirmed to exhibit about 70% growth inhibition and apoptosis effect compared to the control group (FIG. 12).
<193>  <193>
<194> 이를 통해, 10¾> 혈청이 들어있는 정상 배지에서도, MAP7D2 발현 억제가 성장 인자의 의존성을 낮추고 세포 사멸을 유도하여, 최종적으로 세포 성장의 정도를 낮 추었음을 확인할 수 있었으며, 따라서, MAP7D2는 신장암 및 폐암에서 유전자 중독 (oncogene addiction)을 뚜렷하게 나타내므로, 신장암과 폐암에서 항암치료를 위한 좋은 타켓이 될 수 있다.  Through this, it was confirmed that suppression of MAP7D2 expression in the normal medium containing 10¾ serum lowered the dependence of growth factors and induced cell death, thereby finally lowering the extent of cell growth. Thus, MAP7D2 Has a clear oncogene addiction in renal and lung cancers, and thus may be a good target for chemotherapy in renal and lung cancers.
<195>  <195>
<196> <실시예 4> MAP7D2 발현 억제의 암세포 이동에 대한 영향 분석  <Example 4> Analysis of the effect on cancer cell migration of inhibition of MAP7D2 expression
<i 7> MAP7D2의 발현 억제에 의해 암세포의 이동 억제 효과를 나타내는지를 확인하 기 위하여, 트랜스웰 (transwellKcoastar, 3422) 시스템을 이용하였고, 상기 실시 예 <3-2>에 기재된 shRNA로 MAP7D2의 발현을 저해시킨 폐암 세포주 NCI-H1703를 사 용하였다. 이때 CCK-8 실험과 동일하게, 세포에서 렌티바이러스 백터 shRNA에 의 한 비특이적인 반웅을 배제하기 위하여 Nontarget shRNA Control Transduction particles을 음성 대조군 shRNA로 사용하였고, 형광물질이 부착된 렌티바이러스 백 터를 사용하여 도입 효율을 예측하였다. 도입 효율은 약 80% 이상 수준이었고, 형 질감염 48 시간 후, 2 ug/ml의 퓨로마이신 (puromycin)을 처리하여 4일간의 선별을 거친 다음, 암세포를 회수하여 암세포의 이동 능력을 조사하였다.  <i 7> Transwell (transwell Kcoastar, 3422) system was used to confirm whether the inhibition of MAP7D2 expression by cancer cells, the expression of MAP7D2 by the shRNA described in Example <3-2> Lung cancer cell line NCI-H1703 was inhibited. At this time, as in CCK-8 experiment, Nontarget shRNA Control Transduction particles were used as negative control shRNA to exclude nonspecific reaction by lentiviral vector shRNA in cells, and the lentiviral vector to which fluorescent material was attached was used. The introduction efficiency was predicted. The efficiency of introduction was about 80% or more. After 48 hours of phenotypic infection, 2 ug / ml of puromycin was used for 4 days of selection, and then the cancer cells were recovered and examined for their ability to migrate.
<1 8> 구체적으로, 암세포의 이동 능력을 확인하기 위해, 트립신 (trypsinXGibco  <1 8> Specifically, to determine the ability of cancer cells to migrate, trypsin (trypsinXGibco
25300)으로 MAP7D2의 발현을 억제하고 퓨로마이신 (puromycin)으로 선별한 NCI- H1703 세포를 수득한 다음, RPMI migration media(RPMI, 10mM HEPES, 0.5% BSA)로 25300) to inhibit the expression of MAP7D2 and obtain NCI-H1703 cells screened with puromycin, followed by RPMI migration media (RPMI, 10 mM HEPES, 0.5% BSA).
2회 세척한 후에 4χ10ΰ세포수 /ml 로 이동 배지에 세포를 현탁하였다. 이후, 24-웰 트랜스웰 플레이트는 인서트 (insert) 아래쪽 면에 0.05% 젤라틴 (Sigma G1393)을 사 용하여 실온에서 한 시간 동안 코팅하고 한 시간이 지난 후, 인서트 (insert)에 남 아있는 젤라틴을 제거하고 PBS로 1회 세척하였다. 상기 과정이 끝난 후, 5¾ FBS가 첨가된 600 ul의 RPMI 이동 배지를 챔버 (chamber)에 첨가하였다. 소독된 핀셋을 사용하여 인서트를 챔버에 넣은 다음, 미리 준비해둔, 세포를 인서트 안에 4xl04세 포수 /100 ul로 넣고, 37°C/5% CO2의 조건에서 24시간 동안 배양하였다. 세포의 이 동을 측정하기 위해, 먼저 인서트의 위쪽 면을 PBS에 적신 면봉으로 닦아내고 3.7% 파라포름알데히드 (paraformaldehyde) 500 ul가 포함된 챔버에 인서트를 넣어서 실 온에서 30분간 고정하였다. 그런 다음, 1¾ 크리스탈 바이올렛 (crystal violet)/100 ltiM 붕산나트륨 (Sodium Borate, NaBorate) 500 ul에 30분간 염색하여 물로 세척한 후에 건조시켜서 현미경으로 xlOO 배율에서 세포수를 측정하였다. After washing twice, the cells were suspended in transfer medium at 4χ10 cells / ml. The 24-well transwell plate is then coated with 0.05% gelatin (Sigma G1393) on the underside of the insert (i nser t) for one hour at room temperature and after one hour, the remaining gelatin in the insert. Was removed and washed once with PBS. After the procedure, 600 ul of RPMI transfer medium with 5¾ FBS was added to the chamber. After inserting the insert into the chamber using sterile tweezers, the cells were prepared in advance, the cells were placed in the insert into 4xl0 4 year old catcher / 100 ul and incubated for 24 hours at 37 ° C / 5% CO 2 conditions. To measure cell migration, first wipe the top of the insert with a cotton swab moistened in PBS and insert the insert into a chamber containing 500 ul of 3.7% paraformaldehyde. It fixed for 30 minutes at temperature. Then, 1¾ crystal violet / 100 ltiM sodium borate (Sodium Borate, NaBorate) was stained with 500 ul for 30 minutes, washed with water and dried to measure the number of cells at xlOO magnification under a microscope.
<199> 그 결과, 도 11에서 나타낸 바와 같이, shRNA#2 및 #4에 의해 MAP7D2의 발현 이 억제된 세포는 대조군으로 사용된 nontarget shRNA(shCtr 1)가 도입된 세포와 비 교하여 약 80% 정도의 뚜렷한 이동 저해 효과를 나타내었으며 (도 13), 이를 통해, MAP7D2가 암세포의 이동에 있어 결정적인 역할을 한다는 것을 알 수 있었다.As a result, as shown in FIG. 11, the cells whose MAP7D2 expression was suppressed by shRNA # 2 and # 4 were about 80% compared with the cells into which the nontarget shRNA (shCtr 1) introduced as a control was introduced. It showed a clear movement inhibitory effect (Fig. 13), it can be seen that MAP7D2 plays a critical role in the migration of cancer cells.
<200> <200>
<20i> <실시예 5> MAP7D2 발현 억제의 암세포 침윤에 대한 영향 분석  <Example 5> Analysis of the effect on cancer cell invasion of MAP7D2 expression inhibition
<202> MAP7D2의 발현 억제의 암세포 침윤에 대한 영향을 분석하였다.  The effect of inhibition of expression of MAP7D2 on cancer cell invasion was analyzed.
<203> 구체적으로, RPMI invasion media(RPMI, lOmM HEPES, 0.5% BSA)로 2회 세척 한 후에 4x10세포수 /ml 로 침윤 배지에 세포를 현탁하였다. 24-웰 트랜스웰 플레 이트 (8 urn 공극 크기, costar 3422)는 마트리겔 (matrigel )(BD 354234)을 1 mg/ml로 serum-free media(RPMI, lOmM HEPES)에 희석하여 인서트 위쪽 면에 실온에서 한 시 간 동안 코팅하였다. 한 시간 후, 인서트에 남아있는 마트리겔을 제거하고 무혈청 배지로 1회 세척하였다. 이후, 5% FBS가 첨가된 600 ul의 RPMI 침윤 배지를 첨가 하였다. 소독된 핀셋을 사용하여 인서트를 배지가 들어있는 챔버에 넣은 다음/ 미 리 준비하여 둔 NCI-H1703 세포를 인서트 안에 100 ul 첨가하여 37°C/5% CO2의 조건 에서 24시간 배양하였다. 마트리겔을 통과한 침윤된 세포를 측정하기 위해 인서트 의 위쪽 면을 PBS에 적신 면봉으로 닦아내고 3.7% 파라포름알데히드 (paraformaldehyde) 500 uKSigma HT50)가 들어있는 챔버에 인서트를 넣고 실온에 서 30분간 고정하였다. 그런 다음, 1% 크리스탈 바이을렛 (crystal violet )(Sigma C3886)/100 mM NaBorate (Sigma S9640) 500 ul에 30분간 염색하고 물로 세척한 후 건조시켜서 현미경으로 xlOO 과 x200 배율에서 사진을 찍고 세포 수를 세었다. Specifically, the cells were washed twice with RPMI invasion media (RPMI, lOmM HEPES, 0.5% BSA), and then the cells were suspended in the infiltration medium at 4 × 10 cells / ml. 24-well transwell plates (8 urn pore size, costar 3422) were diluted with Matrigel (BD 354234) in serum-free media (RPMI, lOmM HEPES) at 1 mg / ml at room temperature on the top of the insert. Coating for one hour at. After one hour, the Matrigel remaining in the insert was removed and washed once with serum-free medium. Then, 600 ul RPMI infiltration medium with 5% FBS was added. Using sterilized tweezers, insert the insert into the chamber containing the medium / pre-prepared NCI-H1703 cells were incubated for 24 hours at 37 ° C / 5% CO 2 by adding 100 ul into the insert. To measure the infiltrated cells that passed through the Matrigel, wipe the top of the insert with a cotton swab moistened with PBS, insert the insert into a chamber containing 3.7% paraformaldehyde 500 uKSigma HT50) and fix for 30 minutes at room temperature. . Then, 1% crystal violet (Sigma C3886) / 100 mM NaBorate (Sigma S9640) was stained with 500 ul for 30 minutes, washed with water, dried and photographed at xlOO and x200 magnification under a microscope. Counted.
<204> 그 결과, 도 12에서 나타낸 바와 같이, shRNA#2 및 #4에 의해 MAP7D2의 발현 이 억제된 세포는 대조군으로 사용된 nontarget shRNA(shCtr 1 )가 도입된 세포와 비 교하여 약 70% 정도의 뚜렷한 침윤 저해 효과를 나타내었으며 (도 14), 이를 통해, MAP7D2가 암세포의 침윤에 있어 결정적인 역할을 한다는 것을 알 수 있었다.As a result, as shown in FIG. 12, the cells whose MAP7D2 expression was suppressed by shRNA # 2 and # 4 were about 70% compared with the cells into which the nontarget shRNA (shCtr 1) introduced as a control was introduced. It showed a significant degree of invasion inhibitory effect (FIG. 14), and it can be seen that MAP7D2 plays a critical role in cancer cell invasion.
<205> <205>
<206> <제조예 1> 약학적 제제의 제조  Preparation Example 1 Preparation of Pharmaceutical Formulation
<207> 1. 산제의 제조  1. Preparation of powder
<208> MAP7D2의 발현 또는 활성 억제제 2 g <209> 유당 1 g <208> 2 g of inhibitor of expression or activity of MAP7D2 <209> lactose 1 g
<210> 상기의 성분을 흔합하고 기밀포에 층진하여 산제를 제조하였다. The above ingredients were mixed and layered in an airtight cloth to prepare a powder.
<211> <211>
<212> 2. 정제의 제조 2. Preparation of Tablets
<213> MAP7D2의 발현 또는 활성 억제제 100 mg <213> Inhibitor of MAP7D2 expression or activity 100 mg
<214> 옥수수전분 100 nig <214> corn starch 100 nig
<215> 유 당 100 mg <215> Lactose 100 mg
<216> 스테아린산 마그네슘 2 nig 2 mg of magnesium stearate
<217> 상기의 성분을 흔합한 후, 통상의 정제의 제조방법에 따라서 타정하여 정제 제조하였다. After the above components were mixed, tablets were prepared by tableting according to a conventional method for preparing tablets.
<218>  <218>
<21 > 3. 캡슐제의 제조 3. Preparation of Capsules
<220> MAP7D2의 발현 또는 활성 억제제 100 nig <220> 100 nig of inhibitor of expression or activity of MAP7D2
<221> 극 Oᄀ ·- -J ᄇ <221> Oa --- J J
~ r nr 100 rag  ~ r nr 100 rag
<222> 유 당 100 mg <222> Lactose 100 mg
<223> 스테아린산 마그네슘 2 mg <223> magnesium stearate 2 mg
<224> 상기의 성분을 흔합한 후, 통상의 캡슐제의 제조방법에 따라서 젤라틴 캡슐 에 충전하여 캡슐제를 제조하였다. After mixing the above components, the capsule was prepared by filling in gelatin capsules according to a conventional method for preparing capsules.
<225>  <225>
<226> 4. 주사제의 제조 4. Preparation of Injectables
<227> MAP7D2의 발현 또는 활성 억제제 10 //g/m Inhibitor of Expression or Activity of MAP7D2 10 // g / m
<228> 묽은 염산 BP pH 7.6로 될 때까지 Dilute hydrochloric acid BP until pH 7.6
<229> 주사용 염화나트륨 BP 최대 1 m« <229> Injectable sodium chloride BP up to 1 m «
<230> 적당한 용적의 주이용 염화나트륨 BP 중에 MAP7D2의 발현 또는 활성을 억제 제를 용해시키고, 생성된 용액의 pH를 묽은 염산 BP를 이용하여 pH 7.6로 조절하 고, 주이용 염화나트륨 BP를 이용하여 용적을 조절하고 충분히 흔합하였다. 용액 을 투명 유리로 된 5 mi 타입 I 앰플 중에 층전시키고, 유리를 용해시킴으로써 공 기의 상부 격자하에 봉입시키고, 12C C에서 15 분 이상 오토클래이브시켜 살균하여 주사액제를 제조하였다. Dissolve the inhibitor in the expression or activity of MAP7D2 in a suitable volume of sodium chloride BP in a suitable volume, adjust the pH of the resulting solution to pH 7.6 using dilute hydrochloric acid BP, and volume using a sodium chloride BP for main volume. Was adjusted and mixed well. The solution was layered in a 5 mi type I ampoule made of clear glass, encapsulated under the upper grid of air by dissolving the glass, and sterilized by autoclaving at 12 C C for at least 15 minutes to prepare an injection solution.
<231>  <231>
<232> 5. 환의 제조 5. Preparation of Rings
<233> MAP7D2의 발현 또는 활성 억제제 1 <234> 유당 1.5 g Inhibitors of expression or activity of MAP7D2 1 <234> Lactose 1.5 g
<235> 글리세린 1 g <235> 1 g of glycerin
<236> 자일리를 0.5 g <236> 0.5 g of Xili
<237> 상기의 성분을 흔합한 후, 통상의 방법에 따라 1 환 당 4 g이 I도록 제조하 였다 After mixing the above components, it was prepared to I 4 g per ring according to a conventional method.
<238>  <238>
<239> 6. 과립의 제조 6. Preparation of Granules
<240> MAP7D2의 발현 또는 활성 억제제 150 rag <240> Inhibitor of Expression or Activity of MAP7D2 150 rag
<241> 대두 추출물 50 nig <241> soybean extract 50 nig
<242> 포도당 200 rag <242> glucose 200 rag
<243> 전분 600 rag <243> starch 600 rag
<244> 상기의 성분을 흔합한 후, 30% 에탄을 100 mg을 첨가하여 60°C에서 건조하여 과립을 형성한 후 포에 충진하였다. After mixing the above components, 100 mg of 30% ethane was added, dried at 60 ° C. to form granules, and then filled into fabric.
<245>  <245>
【산업상 이용 가능성】  [Industrial availability]
<246> 상기에서 살펴본 바와 같이 , 본 발명을 통해 MAP7D2의 발현 또는 활성 억제 제를 이용한 암 치료제 개발이 가능하고, MAP7D2를 바이오마커로 이용하여 암을 모 니터링 또는 진단할 수 있는 방법을 개발할 수 있으며, 암 성장 또는 전이 억제제 를 스크리닝하는데 유용하게 이용될 수 있다.  As described above, the present invention enables the development of cancer therapeutic agents using MAP7D2 expression or activity inhibitors, and the development of a method for monitoring or diagnosing cancer using MAP7D2 as a biomarker. It may be usefully used for screening cancer growth or metastasis inhibitors.

Claims

【청구의 범위】 [Range of request]
【청구항 1】  [Claim 1]
MAP7D2(MAP7 domain containing 2) 단백질의 발현 또는 활성 억제제를 유효 성분으로 함유하는 암 치료 또는 암 전이 억제용 약학적 조성물.  A pharmaceutical composition for treating cancer or inhibiting cancer metastasis, comprising as an active ingredient an inhibitor of MAP7D2 (MAP7 domain containing 2) protein expression or activity.
【청구항 2】 [Claim 2]
제 1항에 있어서, 상기 MAP7D2 단백질은 서열번호 1로 기재되는 아미노산 서 열을 갖는 것을 특징으로 하는 암 치료 또는 암 전이 억제용 약학적 조성물.  The pharmaceutical composition for treating cancer or inhibiting cancer metastasis according to claim 1, wherein the MAP7D2 protein has an amino acid sequence as set forth in SEQ ID NO: 1.
【청구항 3】 [Claim 3]
제 1항에 있어서, 상기 MAP7D2 단백질의 발현 억제제는 MAP7D2 유전자의 mRNA에 상보적으로 결합하는 안티센스 뉴클레오티드, 짧은 헤어핀 RNA, 작은 간섭 RNA( small interfering RNA) 및 리보자임 (r ibozyme)으로 구성된 군으로부터 선택되 는 어느 하나인 것을 특징으로 하는 암 치료 또는 암 전이 억제용 약학적 조성물.  The method of claim 1, wherein the inhibitor of expression of the MAP7D2 protein is selected from the group consisting of antisense nucleotides, short hairpin RNAs, small interfering RNAs, and ribozymes that complementarily bind to mRNAs of the MAP7D2 genes. A pharmaceutical composition for treating cancer or inhibiting cancer metastasis, characterized in that any one.
【청구항 4] [Claim 4]
제 1항에 있어서, 상기 MAP7D2 단백질의 활성 억제제는 MAP7D2 단백질에 상 보적으로 결합하는 화합물, 펩티드, 펩티드 미메틱스, 기질유사체, 앱타머 및 항체 로 구성된 군으로부터 선택되는 어느 하나인 것을 특징으로 하는 암 치료 또는 암 전이 억제용 약학적 조성물.  The method according to claim 1, wherein the activity inhibitor of the MAP7D2 protein is any one selected from the group consisting of compounds, peptides, peptide mimetics, substrate analogs, aptamers and antibodies that binds to the MAP7D2 protein complementarily. Pharmaceutical compositions for treating cancer or inhibiting cancer metastasis.
【청구항 5】 [Claim 5]
제 1항에 있어서, 상기 암은 신장암, 폐암, 대장암, 난소암, 위암 및 간암으 로 구성된 군으로부터 선택되는 어느 하나인 것을 특징으로 하는 암 치료 또는 암 전이 억제용 약학적 조성물.  The pharmaceutical composition for treating cancer or inhibiting cancer metastasis according to claim 1, wherein the cancer is any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer.
【청구항 6] [Claim 6]
1) MAP7D2 단백질의 발현 세포주에 피검물질을 처리하는 단계;  1) treating the test substance to the cell line expressing MAP7D2 protein;
2) 상기 세포주에서 MAP7D2 단백질의 발현 정도를 측정하는 단계; 및  2) measuring the expression level of MAP7D2 protein in the cell line; And
3) 상기 MAP7D2 단백질의 발현 정도가 피검물질을 처리하지 않은 대조군에 비해 감소한 피검물질을 선별하는 단계를 포함하는, 암 치료 또는 암 전이 억제용 후보 물질의 스크리닝 방법 . 3) screening a test substance for treating cancer or inhibiting cancer metastasis, comprising selecting a test substance in which the expression level of the MAP7D2 protein is reduced compared to a control group not treated with the test substance.
【청구항 7】 [Claim 7]
제 6항에 있어서, 단계 2)의 단백질의 발현 정도는 면역침강법 (i圍 unoprecipitation), 방사능면역분석법 (RIA) , 효소면역분석법 (ELISA), 면역조직 화학, RT-PCR, 웨스턴 블롯 (Western Blotting) 및 유세포 분석법 (FACS)으로 이루어 진 군으로부터 선택된 어느 하나로 측정하는 것을 특징으로 하는 방법 .  The method of claim 6, wherein the expression level of the protein of step 2) is determined by immunoprecipitation, radioimmunoassay (RIA), enzyme immunoassay (ELISA), immunohistochemistry, RT-PCR, Western blot (Western). Blotting) and flow cytometry (FACS).
【청구항 8] [Claim 8]
1) MAP7D2 단백질에 피검물질을 처리하는 단계;  1) treating the test substance to the MAP7D2 protein;
2) 상기 MAP7D2 단백질의 활성 정도를 측정하는 단계 ; 및  2) measuring the activity of the MAP7D2 protein; And
3) 상기 MAP7D2 단백질의 활성 정도가 피검물질을 처리하지 않은 대조군에 비해 감소한 피검물질을 선별하는 단계를 포함하는, 암 치료 또는 암 전이 억제용 후보 물질의 스크리닝 방법 .  3) screening a test substance for treating cancer or inhibiting cancer metastasis, comprising selecting a test substance whose activity level of the MAP7D2 protein is reduced compared to a control group not treated with the test substance.
【청구항 9】 [Claim 9]
제 8항에 있어서, 단계 2)의 단백질의 활성 정도는 SDS-PAGE, 면역형광법, 효소면역분석법 (ELISA), 질량분석 및 단백질 칩으로 이루어진 군으로부터 선택되는 어느 하나로 측정하는 것을 특징으로 하는 방법 .  The method of claim 8, wherein the activity of the protein of step 2) is measured by any one selected from the group consisting of SDS-PAGE, immunofluorescence, enzyme immunoassay (ELISA), mass spectrometry and protein chip.
【청구항 10] [Claim 10]
1) 피검체 유래 시료에서 MAP7D2 단백질의 발현 수준을 측정하는 단계; 및 1) measuring the expression level of MAP7D2 protein in a subject-derived sample; And
2) MAP7D2 단백질의 발현 수준이 대조군보다 증가된 피검체를 암에 걸릴 위 험이 있는 개체로 판정하는 단계를 포함하는, 암 진단 방법. 2) A method for diagnosing cancer, comprising: determining a subject having an increased expression level of MAP7D2 protein than a control group as a subject at risk of cancer.
【청구항 11】 [Claim 11]
제 6항, 제 8항 또는 제 10항에 있어서, 상기 암은 신장암, 폐암, 대장암, 난소암, 위암 및 간암으로 구성된 군으로부터 선택되는 어느 하나인 것을 특징으로 하는 방법 .  The method of claim 6, 8, or 10, wherein the cancer is any one selected from the group consisting of kidney cancer, lung cancer, colon cancer, ovarian cancer, gastric cancer, and liver cancer.
【청구항 12] [Claim 12]
약학적으로 유효한 양의 MAP7D2 단백질 발현 또는 활성 억제제를 개체에 투 여하는 단계를 포함하는 암 예방 방법. A method of preventing cancer, comprising administering to a subject a pharmaceutically effective amount of an MAP7D2 protein expression or activity inhibitor.
【청구항 13] [Claim 13]
약학적으로 유효한 양의 MAP7D2 단백질 발현 또는 활성 억제제를 암에 걸린 개체에 투여하는 단계를 포함하는 암 치료 또는 암 전이 억제 방법.  A method of treating cancer or inhibiting cancer metastasis comprising administering a pharmaceutically effective amount of a MAP7D2 protein expression or activity inhibitor to a subject with cancer.
【청구항 14] [Claim 14]
MAP7D2에 특이적으로 결합하는 항체 또는 ; ?유전자에 상보적인 핵산 중 어느 하나 이상을 이용하여 암세포에서의 MAP7D2 발현 수준을 측정하는 단계를 포 함하는, 암의 진단, 치료 결과 확인 또는 예후를 평가하는 방법 .  An antibody that specifically binds to MAP7D2 or; ? A method of diagnosing cancer, confirming the outcome of a cancer, or assessing the prognosis, comprising measuring MAP7D2 expression levels in cancer cells using one or more of the genes complementary to the gene.
【청구항 15] [Claim 15]
MAP7D2에 특이적으로 결합하는 항체 또는 유전자에 상보적인 핵산 중 어느 하나 이상을 포함하는 암 진단용 키트.  Cancer diagnostic kit comprising any one or more of nucleic acids complementary to the antibody or gene specifically binding to MAP7D2.
【청구항 16】 [Claim 16]
암의 예방 또는 치료, 또는 암 전이 억제용 약학적 조성물에 사용하기 위한, MAP7D2 단백질 발현 또는 활성 억제제.  Inhibitor of MAP7D2 protein expression or activity for use in the prophylaxis or treatment of cancer, or the pharmaceutical composition for inhibiting cancer metastasis.
【청구항 17】 [Claim 17]
암 진단용 키트에 사용하기 위한, MAP7D2에 특이적으로 결합하는 항체 또는 7^; ^유전자에 상보적인 핵산.  An antibody or 7 ^; ^ nucleic acid complementarily binding to MAP7D2 for use in a cancer diagnostic kit.
PCT/KR2012/001316 2011-02-25 2012-02-21 Pharmaceutical composition for cancer treatment or metastasis inhibition containing expression or activation inhibitors of map7d2 protein, novel cancer therapeutic target WO2012115437A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR10-2011-0017247 2011-02-25
KR20110017247 2011-02-25
KR10-2012-0016917 2012-02-20
KR1020120016917A KR101376599B1 (en) 2011-02-25 2012-02-20 Pharmaceutical composition for the treatment of cancers or inhibition of metastasis containing the expression or activity inhibitors of MAP7D2, a novel cancer therapeutic target

Publications (2)

Publication Number Publication Date
WO2012115437A2 true WO2012115437A2 (en) 2012-08-30
WO2012115437A3 WO2012115437A3 (en) 2012-12-20

Family

ID=46721331

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2012/001316 WO2012115437A2 (en) 2011-02-25 2012-02-21 Pharmaceutical composition for cancer treatment or metastasis inhibition containing expression or activation inhibitors of map7d2 protein, novel cancer therapeutic target

Country Status (1)

Country Link
WO (1) WO2012115437A2 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080292546A1 (en) * 2003-06-09 2008-11-27 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
EP2149613A1 (en) * 2008-07-28 2010-02-03 Greenwood Genetic Center, Inc. Methods for determining dysregulation of methylation of brain expressed genes on the X chromosome to diagnose autism spectrum disorders

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080292546A1 (en) * 2003-06-09 2008-11-27 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
EP2149613A1 (en) * 2008-07-28 2010-02-03 Greenwood Genetic Center, Inc. Methods for determining dysregulation of methylation of brain expressed genes on the X chromosome to diagnose autism spectrum disorders

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BERND HEUBECK ET AL.: 'Tumor-associated antigenic pattern in squamous cell carcinomas of the head and neck - Analysed by SEREX' EUROPEAN JOURNAL OF CANCER 2006, ISSN 0959-8049 *
MADRIGAL ET AL.: 'X-chromosome tiling path array detection of copy number variants in patients with chromosome X-linked mental retardation' BMC GENOMICS vol. 8, no. 443, 2007, ISSN 1471-2164 *

Also Published As

Publication number Publication date
WO2012115437A3 (en) 2012-12-20

Similar Documents

Publication Publication Date Title
Pan et al. MicroRNA-149 inhibits proliferation and invasion of glioma cells via blockade of AKT1 signaling
Liu et al. CCL5 promotes VEGF-dependent angiogenesis by down-regulating miR-200b through PI3K/Akt signaling pathway in human chondrosarcoma cells
US9365851B2 (en) Spalt-like transcription factor 4 (SALL4) and uses thereof
Xu et al. HOXD13 suppresses prostate cancer metastasis and BMP4‐induced epithelial‐mesenchymal transition by inhibiting SMAD1
US9650638B2 (en) Aptamer for periostin and anti-cancer composition including same
KR20210056959A (en) Composition for preventing or treating of liver cancer
US20200326343A1 (en) Gene and its expression product promoting the occurrence and development of cancer and application
KR20170019635A (en) Method for providing the information for chronic myeloid leukemia
CN109321656B (en) Use of protein DEPDC1 as marker for diagnosing triple-negative breast cancer
US11510911B2 (en) Method for prediction of susceptibility to sorafenib treatment by using SULF2 gene, and composition for treatment of cancer comprising SULF2 inhibitor
CN108245680B (en) Application of RIPK4 as target site in preparation of medicines for treating bladder cancer
CN105189786A (en) FALZ for use as a target for therapies to treat cancer
WO2011129427A9 (en) Diagnostic agent and therapeutic agent for cancer
Hu et al. Down regulation of human positive coactivator 4 suppress tumorigenesis and lung metastasis of osteosarcoma
Duijkers et al. Downregulation of Axl in non-MYCN amplified neuroblastoma cell lines reduces migration
Liao et al. Non‑SMC condensin I complex subunit H promotes the malignant progression and cisplatin resistance of breast cancer MCF‑7 cells
KR101376599B1 (en) Pharmaceutical composition for the treatment of cancers or inhibition of metastasis containing the expression or activity inhibitors of MAP7D2, a novel cancer therapeutic target
KR101525229B1 (en) Pharmaceutical composition for the treatment of cancers or inhibition of cancer metastasis containing the inhibitors of Gpr171 expression or activity
WO2012115437A2 (en) Pharmaceutical composition for cancer treatment or metastasis inhibition containing expression or activation inhibitors of map7d2 protein, novel cancer therapeutic target
KR101817386B1 (en) Method for screening agent for treatment of cancers or inhibition of cancer metastasis using ELK3, and pharmaceutical composition for the treatment of cancers or inhibition of cancer metastasis containing the inhibitors of ELK3 expression or activity
KR101171863B1 (en) Pharmaceutical composition for the treatment of cancers or inhibition of metastasis containing the expression or activity inhibitors of PRPF4, a novel cancer therapeutic target
US20150258173A1 (en) Compositions for modulating invasion ability of a tumor and methods thereof
CN111150726B (en) Application of 4-hydroxy-2-methylene butyric acid (5-formoxyl furan-2-methyl) ester in preparation of liver cancer treatment drug
US20220364184A1 (en) Compositions and methods for treatment of a poor prognosis subtype of colorectal cancer
JP5098013B2 (en) Method for establishing cell line of human malignant ovarian germ cell tumor, human malignant ovarian embryo tumor cell line, and use 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: 12748989

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12748989

Country of ref document: EP

Kind code of ref document: A2