US20230265526A1 - Cd133 related to anticancer agent resistance in colon cancer and use thereof - Google Patents

Cd133 related to anticancer agent resistance in colon cancer and use thereof Download PDF

Info

Publication number
US20230265526A1
US20230265526A1 US18/186,129 US202318186129A US2023265526A1 US 20230265526 A1 US20230265526 A1 US 20230265526A1 US 202318186129 A US202318186129 A US 202318186129A US 2023265526 A1 US2023265526 A1 US 2023265526A1
Authority
US
United States
Prior art keywords
protein
gene
cells
egfr
resistance
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US18/186,129
Inventor
Je Sang Ko
Min-soo Kang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Korea University Research and Business Foundation
Original Assignee
Korea University Research and Business Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Korea University Research and Business Foundation filed Critical Korea University Research and Business Foundation
Priority to US18/186,129 priority Critical patent/US20230265526A1/en
Publication of US20230265526A1 publication Critical patent/US20230265526A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57419Specifically defined cancers of colon
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • 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
    • C12N15/1138Non-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 against receptors or cell surface proteins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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.
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0006Modification of the membrane of cells, e.g. cell decoration
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70596Molecules with a "CD"-designation not provided for elsewhere in G01N2333/705
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present invention relates to a use of CD133 involved in anticancer agent resistance in colon cancer.
  • anticancer agents against various types of cancer have been developed to date, only a few types of cancer can be completely cured with an anticancer agent alone, this is because cancer cells do not respond to an anticancer agent in cancer treatment with the anticancer agent, or tumors are effectively reduced in an early stage, but develop anticancer agent resistance during or after treatment. Therefore, for effective anticancer treatment, resistance to an anticancer agent, for example, anticancer agent resistance of cancer cells, has to be overcome.
  • a mutation in the KRAS, NRAS or BRAF gene results in production of a protein having a modified signaling characteristic in tumor cells, and such a mutation has been known to be associated with unsuccessful results in cancer treatment using a therapeutic antibody targeting an epithelial growth factor receptor (EGFR), for example, gefitinib, cetuximab or panitumumab (Amado, Wolf et al. 2008; Karapetis, Khambata-Ford et al. 2008; Di Nicolantonio, Martini et al. 2008; Loupakis, Ruzzo et al. 2009; Lievre, Bachet et al. 2006).
  • EGFR epithelial growth factor receptor
  • KRAS G13D known as a colon cancer mutant cell line was reported to have resistance (tolerance) to an anticancer agent targeting EGFR.
  • CD133 is related to resistance of colon cancer cells against an anticancer agent targeting EGFR, and thus the present invention was completed.
  • the present invention is directed to providing a biomarker composition for diagnosing resistance to an EGFR-targeting agent in colon cancer, which includes CD133 protein or a gene encoding the same.
  • the present invention is also directed to providing a kit for diagnosing resistance to an EGFR-targeting agent in colon cancer.
  • the present invention is also directed to providing a method of providing information required for diagnosis of resistance to an EGFR-targeting agent in colon cancer.
  • the inventors had confirmed, through a study on a KRAS G13D mutant colon cancer cell line having resistance to an EGFR-targeting agent, that the cell migration to and invasiveness of adjacent normal cells are highly improved by transferring the mutant to adjacent cells via microvesicles, the migration of KRAS G13D via microvesicles imparts anticancer agent resistance to adjacent colon cancer cells, and the CD133 protein controlling the release of microvesicles is related to resistance to an EGFR-targeting agent, and thus the present invention was completed.
  • the present invention is characterized by confirming whether colon cancer cells have resistance to an EGFR-targeting agent using the activity of the CD133 gene and an active protein, which is a product thereof, as a biomarker.
  • the present invention provides a biomarker composition for diagnosing resistance to an EGFR-targeting agent in colon cancer, which includes the CD133 protein or a gene encoding the same.
  • the biomarker of the present invention may be a marker with anticancer agent resistance, which is an EGFR-targeting agent, and since it has excellent accuracy and reliability as a marker for diagnosing anticancer agent resistance, the biomarker may be used to diagnose the occurrence, development and/or metastasis of resistant cancer, and treat resistant cancer.
  • tolerance means that an organism does not sensitively respond to a drug, and thus withstands the drug's effects.
  • CD133 used as a biomarker for resistance diagnosis in the present invention is one of the representative CD-type proteins expressed on the surface of cancer stem cells, which are reported to be mainly present in cancer stem cells of colon cancer, liver cancer, pancreatic cancer and lung cancer.
  • colon cancer used herein is a collective term referring to rectal cancer, colorectal cancer and anal cancer.
  • the EGFR-targeting agent of the present invention refers to an anticancer agent, which may be any EGFR-targeting agent exhibiting an anticancer effect, and is used interchangeably with an EGFR-targeting drug.
  • the EGFR-targeting agent is one or more selected from the group consisting of cetuximab, gefitinib, erlotinib, panitumumab, PKI-166, EKB-569, HKI-272 (WAY-177820), icotinib, brigatinib, afatinib, lapatinib, canertinib, AEE788, XL647, and Zactima.
  • the EGFR-targeting agent is cetuximab, gefitinib, erlotinib or panitumumab, and most preferably, gefitinib.
  • diagnosis refers to the detection of a pathological condition, which means, in terms of the purpose of the present invention, the determination of the presence or absence of resistance, and the development or alleviation of symptoms of a disease by examining the presence or absence of the expression of a biomarker for diagnosing resistance to an EGFR-targeting agent.
  • the “diagnosis biomarker” used herein refers to a material that can be used in diagnosis by distinguishing the presence or absence of resistance to an EGFR-targeting agent, and includes organic biomolecules such as polypeptides or nucleic acids (e.g., mRNA, etc.), lipids, glycolipids, glycoproteins, saccharides (a monosaccharide, a disaccharide, an oligosaccharide, etc.), which increase or decrease in cancer cells resistant to an anticancer agent, compared to cancer cells.
  • the biomarker for resistance diagnosis according to the present invention may be a protein expressed from the CD133 gene whose expression level is increased in resistant cancer cells with respect to an EGFR-targeting agent, compared to general cancer cells.
  • composition for diagnosing resistance to an EGFR-targeting agent may include an agent that measures an mRNA expression level of the CD133 gene or an amount of a protein expressed from the gene, and such an agent may be an oligonucleotide having a complementary sequence to CD133 mRNA, for example, a primer or nucleic acid probe specifically binding to CD133 mRNA, or an antibody specific for CD133 protein.
  • the primer refers to a single-stranded oligonucleotide that is able to serve as a starting point of template-directed DNA synthesis under suitable conditions (that is, four different types of nucleoside triphosphates and a polymerase) in a suitable buffer at a suitable temperature.
  • suitable conditions that is, four different types of nucleoside triphosphates and a polymerase
  • a suitable length of the primer may vary according to various factors, for example, a temperature and the usage of the primer.
  • the primer sequence is not required to be perfectly complementary to a partial sequence of the template, and it is sufficient that the primer sequence has sufficient complementarity within a range in which the primer can do an intrinsic action when hybridized with the template.
  • the primer of the present invention does not need to have a perfectly complementary sequence to the nucleotide sequence of a gene, which is the template, and it is reasonable that the primer has a sufficient complementarity within a range in which the primer can do an intrinsic action when hybridized with the gene sequence.
  • the primer according to the present invention is preferably used in a gene amplification reaction.
  • the amplification reaction refers to a reaction of amplifying a nucleic acid molecule, and amplification reactions of such a gene are well known in the art, and may include, for example, polymerase chain reaction (PCR), reverse transcriptase chain reaction (RT-PCR), ligase chain reaction (LCR), transcription-mediated amplification (TMA), and nucleic acid sequence-based amplification (NASBA).
  • PCR polymerase chain reaction
  • RT-PCR reverse transcriptase chain reaction
  • LCR ligase chain reaction
  • TMA transcription-mediated amplification
  • NASBA nucleic acid sequence-based amplification
  • the nucleic acid probe refers to a natural or modified monomer or linear oligomer consisting of linkages of such monomers, and includes a deoxyribonucleotide and a ribonucleotide, may be specifically hybridized with a target nucleotide sequence, and naturally occurs or is artificially synthesized.
  • the probe according to the present invention may be a single chain, and preferably, an oligodeoxyribonucleotide.
  • the probe of the present invention may include natural dNMPs (i.e., dAMP, dGMP, dCMP and dTMP), or nucleotide analogs or derivatives.
  • the probe of the present invention may include a ribonucleotide.
  • the probe of the present invention may include backbone-modified nucleotides, such as a peptide nucleic acid (PNA), phosphorothioate DNA, phosphodithioate DNA, phosphoroamidate DNA, amide-linked DNA, MMI-linked DNA, 2′-O-methyl RNA, a-DNA and methyl phosphonate DNA; sugar-modified nucleotides, such as 2′-O-methyl RNA, 2′-fluoro RNA, 2′-amino RNA, 2′-O-alkyl DNA, 2′-O-allyl DNA, 2′-O-alkynyl DNA, hexose DNA, pyranosyl RNA and anhydrohexitol DNA; base-modified nucleotides, such as pyrimidines with a C-5 substituent (the substituent may be fluoro-, bromo-, chloro-, iodo-, methyl-, ethyl-
  • the CD133-specific antibody may be a polyclonal antibody, a monoclonal antibody, a human antibody or a humanized antibody.
  • antibody fragments examples include Fab, Fab′, F(ab′) 2 and Fv fragments; a diabody; a linear antibody (Zapata et al., Protein Eng.8(10):1057-1062(1995)); a single-chain antibody molecule; and a multi-specific antibody formed from an antibody fragment.
  • Fv is a minimal antibody fragment including a complete antigen-recognizing and binding region. This region consists of a dimer of one heavy chain variable region and one light chain variable region, which are firmly bound to each other by a non-covalent bond.
  • the polyclonal antibody may be prepared by one or more injections of an immunizing agent in combination with an immune adjuvant, if necessary, into a mammal.
  • an immunizing agent and/or an immune adjuvant is/are subcutaneously or intraperitoneally injected into a mammal several times.
  • the immunizing agent may be a protein of the present invention or a fusion protein thereof It may be effective that an immunizing agent, as well as a protein known to be immunogenic, is injected into an immunized mammal.
  • the monoclonal antibody according to the present invention may be prepared by the hybridoma method disclosed in the literature (Kohler et al., Nature, 256:495 (1975)), or a recombinant DNA method (refer to U.S. Pat. No. 4,816,576).
  • the monoclonal antibody may also be isolated from a phage antibody library using the technology disclosed in the literature (Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991)).
  • a part of a heavy chain and/or a light chain is identical to or has homology with a corresponding sequence of an antibody derived from a specific species or an antibody belonging to a specific antibody class or subclass, whereas the remainder of the chain(s) includes an antibody derived from a different species, an antibody belonging to a different antibody class or subclass or a “chimeric” antibody which is identical or homologous to a fragment of such an antibody (Morrison et al., Proc. Natl. Acad. Sci. USA, 81:6851-6855 (1984)).
  • a “humanized” form of a non-human (e.g., rodents) antibody is a chimeric immunoglobulin including a minimal sequence derived from a non-human immunoglobulin, an immunoglobulin chain or a fragment thereof (e.g., Fv, Fab, Fab′, F(ab′) 2 or a different antigen-binding sequence of an antibody).
  • the humanized antibody includes a human immunoglobulin (recipient antibody) in which a complementarity-determining region (CDR) residue of a recipient is substituted with a CDR residue of a non-human species (donor antibody) such as a mouse, rat or rabbit, having desired specificity, affinity and ability.
  • CDR complementarity-determining region
  • aFv framework residue of the human immunoglobulin is substituted with a corresponding non-human residue.
  • the humanized antibody may include a recipient antibody, or a residue which is not found in an introduced CDR or framework sequence.
  • the humanized antibody substantially includes one or more, generally two or more variable domains, and here, all or substantially all CDR regions correspond to a region of a non-human immunoglobulin, and all or substantially all FR regions correspond to a region of a human immunoglobulin sequence.
  • the humanized antibody includes at least a part of a variable region (Fc) of an immunoglobulin, generally, a part of a human immunoglobulin region (Presta, Curr. Op. Struct. Biol. 2:593-596 (1992)).
  • composition for diagnosing resistance to an EGFR-targeting agent according to the present invention may be included in the form of a kit.
  • the kit may include a primer, probe or antibody which is able to measure the expression level of the CD133 gene or an amount of the CD133 protein, and the definition thereof is as described above.
  • reagents required for PCR amplification for example, a buffer, a DNA polymerase (e.g., a heat-stable DNA polymerase obtained from Thermusaquaticus (Taq), Thermusthermophilus (Tth), Thermusfiliformis, Thermisflavus, Thermococcusliteralis or Pyrococcusfuriosus (Pfu)), a DNA polymerase cofactor and dNTPs may be included, and when the kit is used in an immunoassay, the kit of the present invention may optionally include a secondary antibody and a marker substrate. Further, the kit according to the present invention may be manufactured in multiple individual packages or compartments containing the above-mentioned reagent components.
  • a DNA polymerase e.g., a heat-stable DNA polymerase obtained from Thermusaquaticus (Taq), Thermusthermophilus (Tth), Thermusfiliformis, Thermisflav
  • composition for diagnosing resistance to an EGFR-targeting agent according to the present invention may be included in the form of a microarray.
  • a primer, probe or antibody that is able to measure the expression level of the CD133 protein or gene encoding the same is used as a hybridizable array element, and fixed on a substrate.
  • a preferable substrate may be a suitable rigid or semi-rigid support, for example, a membrane, a filter, a chip, a slide, a wafer, a fiber, a magnetic or non-magnetic bead, a gel, a tubing, a plate, a polymer, a microparticle and a capillary.
  • the hybridizable array element may be arranged and immobilized on the substrate, and such immobilization may be performed by a chemical binding method or a covalent binding method such as UV.
  • the hybridizable array element may be bound to a glass surface which is modified to include an epoxy compound or an aldehyde group, or bound to a polylysine-coated surface using UV.
  • the hybridizable array element may bind to a substrate by a linker (e.g., an ethylene glycol oligomer or a diamine).
  • a sample applied to the microarray of the present invention when a sample applied to the microarray of the present invention is a nucleic acid, it may be labeled and hybridized with an array element fixed on the microarray.
  • Hybridization conditions may vary, and detection and analysis of a hybridization degree may be performed in various ways according to a labeling material.
  • the present invention provides a method of providing information for diagnosing resistance to an EGFR-targeting agent, which includes measuring the expression level of the CD133 gene or the expressed protein in a biological sample isolated from a patient, and more specifically, the method may include (a) measuring the expression level of the CD133 gene or an amount of the expressed protein in a biological sample of a patient; and (b) measuring the expression level of the gene and an amount of the expressed protein from a sample of a normal control and comparing it with the measurement result obtained in Step (a).
  • the method of measuring the expression level of a gene or an amount of a protein encoded by the gene may be performed by a known process of isolating mRNA or a protein from a biological sample using a known technique.
  • the biological sample refers to a sample obtained from the living body, which is different from a normal control in terms of the expression level of the gene or a protein level according to the occurrence or progression of resistance to an EGFR-targeting agent, and the sample may be, but is not limited to, tissue, a cell, blood, serum, plasma, saliva or urine.
  • the measurement of the expression level of a gene is preferably to measure an mRNA level, and methods of measuring an mRNA level include, but are not limited to, RT-PCR, real time reverse transcription polymerase chain reaction, RNase protection assay, Northern blotting and DNA chip assay.
  • the measurement of a protein level may use an antibody, and in this case, the CD133 protein in a biological sample and an antibody specific for the protein forms a binding product, that is, an antigen-antibody complex, and an amount of antigen-antibody complex formation may be quantitatively measured from the size of a signal of a detection label.
  • the detection level may be selected from the group consisting of an enzyme, a fluorescent material, a ligand, a light-emitting material, a microparticle, a redox molecule and a radioisotope, but the present invention is not limited thereto.
  • Analysis methods for measuring a protein level may include, but not limited to, Western blotting, ELISA, radioimmunoassay, radioimmunodiffusion, Ouchterlonyimmunodiffusion, rocket immunoelectrophoresis, immunohistochemical staining, immunoprecipitation assay, complement fixation assay, FACS, and protein chip assay.
  • the present invention may confirm mRNA expression levels or protein amounts in the control, a patient resistant to an EGFR-targeting agent, or a patient suspected of having resistance to an EGFR-targeting agent through the detection methods described above, and the expression levels are compared to be able to diagnose the occurrence and progression of the resistance to an EGFR-targeting agent.
  • the expression level of the CD133 gene according to the present invention or an amount of the expressed protein is higher than that of the normal control sample, it can be determined that a patient has resistance to an EGFR-targeting agent.
  • the present invention also relates to a composition for preventing or treating resistance to an EGFR-targeting agent, which includes a CD133 inhibitor.
  • the present invention also provides a use of a CD133 inhibitor for preparing a pharmaceutical composition for preventing or treating resistance to an EGFR-targeting agent.
  • CD133 is highly expressed in cancer cells with resistance to an EGFR-targeting agent, as described above, and the expression of CD133 promotes budding of microvesicles, increases cell migration and invasiveness by delivering the oncogenic protein KRAS G13D to adjacent cells using microvesicles, and thus is involved in the occurrence of anticancer agent resistance.
  • the CD133 inhibitor may be used in treatment of resistant cancer.
  • composition for preventing or treating resistance to an EGFR-targeting agent of the present invention may include an agent for reducing mRNA expression of the CD133 gene or protein expression thereof, or degrading function or activity.
  • the CD133 protein inhibitor may be a peptide or compound which is bound with the CD133 protein to regulate a signal of a nerve differentiation pathway.
  • the inhibitor may be selected by a screening method exemplified below, such as protein structure analysis, and may be designed using a method known in the art.
  • the CD133 protein inhibitor may be a material which is bound with the CD133 protein consisting of an amino acid sequence represented by SEQ ID NO: 2 to inhibit activity.
  • a polyclonal antibody, monoclonal antibody, human antibody or humanized antibody against the CD133 protein may be used, and the definition of the antibody is as described above.
  • Resistant cancer may be prevented or treated by inhibiting the function of CD133 in cells using the antibody.
  • the functional or activity inhibitor of the CD133 protein according to the present invention may be delivered using a liposome, a virus, a gene gun, a polymer, ultrasonication or an electric shock, but the present invention is not particularly limited thereto.
  • the CD133 gene may be DNA encoding the same or mRNA transcribed therefrom. Accordingly, the inhibitor of the gene may be an inhibitor which is bound to the gene itself to interfere with transcription or bound to mRNA transcribed from the gene to interfere with the translation of mRNA.
  • the inhibitor of the CD133 gene includes all types of inhibitors that inhibit the expression of the CD133 gene.
  • an inhibitor may be a peptide, nucleic acid or compound binding to the gene.
  • the inhibitor may be selected by a screening method to be described below, such as cell-based screening, and may be designed by a method known in the art.
  • the inhibitor may be an antisense-oligonucleotide, siRNA, shRNA or miRNA against the CD133 gene or a vector including the same.
  • an antisense-oligonucleotide, siRNA, shRNA, miRNA or a vector including the same may be manufactured using a method known in the art.
  • the inhibitor may be a nucleic acid molecule prepared to inhibit the expression of the CD133 gene consisting of SEQ ID NO: 1.
  • RNA refers to double-stranded RNA inducing RNA interference through the cleavage of mRNA of a target gene, and consists of an RNA strand of a sense sequence having the sequence like mRNA of the target gene and an RNA strand of an antisense sequence having a sequence complementary thereto.
  • the siRNA may include siRNA synthesized in vitro, or a form expressed by inserting a base sequence encoding siRNA into an expression vector.
  • vector refers to a gene construct including foreign DNA inserted into a genome encoding a polypeptide.
  • the vector related to the present invention is a vector in which a nucleic acid sequence inhibiting the gene is inserted into a genome, and such a vector may be a DNA vector, a plasmid vector, a cosmid vector, a bacteriophage vector, an enzyme vector, or a viral vector.
  • the antisense may have a sequence complementary to an entire or partial mRNA sequence transcribed from the CD133 gene or a fragment thereof, and bind to the mRNA to inhibit the expression of the CD133 gene or a fragment thereof.
  • shRNAi short hairpin RNAi
  • shRNAi short hairpin RNAi
  • composition of the present invention may further include a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier includes a carrier and a vehicle, which are generally used in the pharmaceutical field, and specifically, includes an ion exchange resin, alumina, aluminum stearate, lecithin, a serum protein (e.g., human serum albumin), a buffer material (e.g., various types of phosphates, glycine, sorbic acid, potassium sorbate, or a partial glyceride compound of a saturated vegetable fatty acid), water, a salt or electrolyte (e.g., protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride or a zinc salt), colloidal silica, magnesium trisilicate, polyvinylpyrrolidone, a cellulose-based substrate, polyethylene glycol, sodium carboxymethylcellulose, polyarylate, wax, polyethylene glycol or lanolin, but the present invention is not limited thereto.
  • an ion exchange resin e.g., Amberlite IR-120, Amberlite,
  • composition of the present invention may further include a lubricant, a wetting agent, an emulsifier, a suspending agent, or a preservative, other than the above-described components.
  • the composition according to the present invention may be prepared as an aqueous solution for parenteral administration, and preferably, a Hank's solution, a Ringer's solution or a buffer solution such as physically buffered saline is used.
  • An aqueous injectable suspension may include a substrate that can increase the viscosity of the suspension such as sodium carboxymethylcellulose, sorbitol or dextran.
  • composition of the present invention may be systemically or locally administered, and formulated in a suitable form by known technology for such administration.
  • the composition may be administered by mixing the active compound with an inactive diluent or edible carrier, encapsulating all components with a hard or soft gelatin capsule or compressing all components in the form of a tablet.
  • the active compound may be mixed with an excipient, and then formed as an edible tablet, a buccal tablet, a troche, a capsule, an elixir, a suspension, a syrup or a wafer.
  • CD133 is well dissolved in saline or a buffer solution, after storage in a freeze-dried state, an effective amount of CD133 may be mixed in saline or a buffer solution to prepare a solution suitable for intravenous injection, subcutaneous injection, muscle injection, intraperitoneal injection or transdermal injection, right before the administration.
  • the effective amount of the active ingredient of the pharmaceutical composition of the present invention refers to an amount required for prevention, inhibition or alleviation of a disease.
  • the effective amount may be adjusted by various factors such as the type and severity of a disease, the type and content of the active ingredient and other components, contained in the composition, the type of a dosage form, and the age, body weight, general health condition, gender and diet of a patient, administration time, administration route, a release rate of the composition, a treatment period, and a co-used drug.
  • the inhibitor of the present invention may be administered once or several times a day, when the type of the inhibitor is a compound, it may be administered at a dose of 0.1 ng/kg to 10 g/kg, when the type of the inhibitor is a polypeptide, protein or antibody, it may be administered at a dose of 0.1 ng/kg to 10 g/kg, or when the type of the inhibitor is an antisense-oligonucleotide, siRNA, shRNAi or miRNA, it may be administered at a dose of 0.01 ng/kg to 10 g/kg.
  • the present invention also provides a method of treating an animal with resistance to an EGFR-targeting agent, which includes administering a composition including a pharmaceutically effective amount of a CD133 inhibitor for preventing or treating resistance to an EGFR-targeting agent to a subject.
  • a pharmaceutical composition and an administration method, which are used in the method of treating resistance to an EGFR-targeting agent, are as described above, and the common description between them will be omitted to avoid excessive complexity of the specification.
  • subjects to which the pharmaceutical composition for preventing or treating resistance to an EGFR-targeting agent may be administered include all kinds of animals.
  • the subjects may be non-human animals such as dogs, cats, and mice.
  • the present invention also provides a method of screening a drug for preventing or treating resistance to an EGFR-targeting agent, which includes bringing the CD133 gene into contact with a candidate ex vivo, and determining whether the candidate promotes or inhibits the expression of the gene.
  • the present invention provides a method of screening a drug for preventing or treating resistance to an EGFR-targeting agent, which includes bringing the CD133 protein into contact with a candidate ex vivo, and determining whether the candidate improves or inhibits the function or activity of the protein.
  • a candidate to be analyzed may be brought into contact with resistant cancer cells having the gene or protein.
  • the candidate may be an individual nucleic acid, protein or peptide, another extract or natural substance, or a compound, which is expected or randomly selected to have a potential as a material promoting or inhibiting the mRNA transcription or mRNA translation into a protein in the base sequence of the CD133 gene or a drug improving or inhibiting the function or activity of the CD133 protein according to a common selection method.
  • the expression level of the gene or the amount or activity of the protein may be measured, and as a result of the measurement, when the expression level of the gene or the amount or activity of the protein is measured as increased or decreased, the candidate may be determined as a material for treating or preventing resistant cancer.
  • the method of measuring the expression level of the gene, or the amount or activity of the protein may be performed by various methods known in the art, and may be, for example, RT-PCR, real time-polymerase chain reaction, Western blotting, Northern blotting, ELISA, radioimmunoassay (RIA), radioimmunodiffusion and immunoprecipitation assay, but the present invention is not limited thereto.
  • the candidate exhibiting an activity of inhibiting gene expression or a protein function, obtained by the screening method of the present invention may be a candidate for a resistant cancer therapeutic agent.
  • the candidate for a therapeutic agent against cancer resistant to an EGFR-targeting agent serves as a leading compound in the development of a therapeutic agent against resistance to an EGFR-targeting agent, and modifies and optimizes the structure of the leading compound to exhibit an effect of inhibiting the CD133 gene or the function of a protein expressed therefrom, thereby developing a novel therapeutic agent against cancer resistant to an EGFR-targeting agent.
  • the present invention identifies that the CD133 protein expressed in colon cancer increases resistance to an EGFR-targeting agent, and thus the CD133 protein can be used as a novel target protein for diagnosing and treating resistant cancer as well as general cancer.
  • FIGS. 1 A- 1 E show the result of identifying a CD133 expression regulatory mechanism in liver cancer cells: (A) the change in CD133 expression according to EGF expression; (B-C) the change in CD133 expression according to an EGF downstream signaling molecule, NF- ⁇ B; and (D-E) the relationship between NF- ⁇ B and CD133, confirmed by promoter activity.
  • FIGS. 2 A- 2 E show the result of confirming that CD133 is involved in promotion of microvesicle budding in liver cancer cells: (A) confirmation of the relationship between an EGF signaling system and microvesicle and TNT formation; (B) confirmation of microvesicle and TNT formation in a CD133—overexpressing cell line; and (C-E) confirmation of the change in activation of Rac1 and RhoA associated with microvesicle budding and a downstream signaling factor, Erk1/2, of RhoA according to CD133 expression
  • FIGS. 3 A- 3 B show the result of analyzing microvesicles budded from cancer cells: (A) the result of confirming whether CD133 is included in microvesicles budded from various cancer cells; and (B) the result of observing the migration of microvesicles containing CD133 to adjacent cells.
  • FIGS. 4 A- 4 F show the result of confirming that CD133 is involved in oncogenic protein transport and microvesicle budding in colon cancer: (A) confirmation whether an oncogenic protein is contained in microvesicles according to CD133 expression; (B-C) confirmation that CD133 expression contributes to microvesicle budding and a size change; (D) confirmation that CD133 expression contributes to the transport of an oncogenic protein via microvesicles and cell proliferation; and (E-F) confirmation that the transport of an oncogenic protein via microvesicles contributes to the change in cell migration to and invasiveness of adjacent normal cells.
  • FIGS. 5 A- 5 F show the result of confirming that CD133—containing microvesicles induce anticancer agent resistance (gefitinib) in colon cancer: (A-B) confirmation of the capability of CD133—containing microvesicles to induce anticancer agent resistance (gefitinib) by confirmation of the proliferation of cancer cells; (C-D) the result of confirming that cancer cell proliferation is caused by an oncogenic protein transported by microvesicles through the change in KRAS downstream signaling molecule and target gene expression; and (E-F) confirmation that cell migration to and invasiveness of adjacent normal cells are changed by the oncogenic protein transported by CD133—containing microvesicles in the presence of gefitinib.
  • CD133 NCBI Gene ID: 8842
  • EGF epithelial growth factor
  • CD133 expression was investigated. Specifically, for transfection and Western blotting, a specific gene (plasmid vector or siRNA) was expressed in cells using a transfection reagent. For general plasmid vector transfection, 24 hours after transfection, cells were harvested, and for siRNA transfection, 48 hours after transfection, cells were harvested. For Western blotting, the harvested cells were lysed in RIPA buffer, and centrifuged at 12,000 g for 20 minutes at 4° C., followed by collecting a supernatant. The collected supernatant was subjected to SDS-PAGE gel electrophoresis, and transferred onto a nitrocellulose membrane, followed by detecting the expression of a desired protein using suitable antibodies.
  • plasmid vector or siRNA plasmid vector or siRNA
  • FIG. 1 B it was observed that NF- ⁇ B is involved in EGF-induced CD133 expression ( FIG. 1 B ). It was confirmed that, when the expression of NF- ⁇ B subunits, such as p50 (NCBI Gene ID: 4790) and p65 (NCBI Gene ID: 5970), was inhibited, CD133 expression decreases ( FIG. 1 C ).
  • NF- ⁇ B subunits such as p50 (NCBI Gene ID: 4790) and p65 (NCBI Gene ID: 5970
  • FIG. 1 C Promoter activity was observed in a promoter near CD133 ORF. Specifically, to measure the promoter activity, a CD133 ORF promoter region was cloned in a special vector expressing luciferase. Cells were simultaneously transfected with the luciferase vector and a Renilla plasmid vector for quantification.
  • EGF EGF-like growth factor
  • TNT TNT formation
  • RhoA and Rac1 are known to be important, and thus an expression pattern of the gene according to the CD133 expression pattern was observed by small GTPase pull-down assay.
  • small GTPases such as ARF6, RhoA and Rac1
  • Rac1 activity after transfected with CD133, cells were harvested, and activity was measured using a Rac1 activation Assay kit.
  • RhoA activity after CD133 transfection, cells were harvested, and activity was measured using a RhoA activation Assay kit.
  • Microvesicles were observed by microvesicle microscopic analysis. Specifically, microvesicles were isolated from the culture solution of CD133-transfected cells and stained with WGA-488, general cells were treated with the stained microvesicles, and after 12 hours, fluorescence intensity was measured using a LSM700 Meta confocal microscope.
  • the microvesicles were released from various types of cancer cells, and contained CD133 ( FIG. 3 A ).
  • CD133 (red)-containing microvesicles were isolated by centrifugation, and treated with WGA to stain the cell membrane. It was observed that, when the cell line was treated with the microvesicles, the microvesicles were transported to adjacent cells along with CD133 ( FIG. 3 B ).
  • the used vector is a vector prepared by substituting a neomycin antibiotic region with Zeomycin in a pSilencer 2.1-U6 neo vector (www.thermo fisher.com/kr/ko/home/life-science/dna-rnapurification-analysis/napamisc/vector-maps/psilencer-2-1-u6-hygro-vectormap.html).
  • microvesicles To isolate microvesicles, the culture solutions of a CD133 normal expression cell line and a CD133 expression-inhibited cell line were collected, and then each cell culture solution was centrifuged at 20,000 g for 1 hour, thereby obtaining a supernatant. After discarding the supernatant, microvesicles were isolated.
  • microvesicle budding was measured by a Malvern Nanosight Nanoparticle Tracking Analysis system.
  • KRAS (NCBI Gene ID: 3845) G13D is a well-known oncogenic protein, and usually found as a KRAS mutant in colon cancer. It was observed that, when CD133 expression was inhibited in a colon cancer cell line, KRAS G13D was not contained in the microvesicles ( FIG. 4 A ). In addition, it was confirmed that CD133 expression is involved in determination of the amount and sizes (100 to 200 nm) of microvesicle budding ( FIGS. 4 B and 4 C ).
  • microvesicles were isolated from the CD133 expression-inhibited cell line and the general cell line and then normal cells were treated with the microvesicles.
  • microvesicle-recipient cells were cultured in an 8- ⁇ m pore insert, treated with microvesicles, and then after 48 hours, migrating cells counted.
  • an 8- ⁇ m pore insert was coated with Matrigel, and then recipient cells were incubated. Forty-eight hours after microvesicle treatment, cells migrating through the Matrigel were counted.
  • gefitinib is an anticancer material that inhibits EGFR activity and thus controls cancer cells. It was reported that, in certain types of cancer in which EGFR downstream signaling molecules including KRAS are activated, gefitinib efficacy was insignificant. It was inferred that the transport of KRAS G13D to adjacent cells by CD133-containing microvesicles is involved in resistance to such an EGFR-targeting agent.
  • Cells being cultured to confirm the above inference were incubated with gefitinib and the microvesicles for a suggested time, cell viability was analyzed using an EZ-cytox kit at specific time, and the absorbance was measured at 570 nm using a microplate reader, followed by calculating a cell growth rate. Twenty-four hours after treatment with the microvesicles and gefitinib, the cells were harvested, and then fixed with 95% cold ethanol. The fixed cells were treated with RNase and propidium iodide to stain DNA, and then the stained DNA was analyzed by FACS.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Plant Pathology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to a use of CD133 involved in resistance to an EGFR-targeting agent in colon cancer. The CD133 protein may be used as a novel target protein for diagnosing and treating resistant cancer as well as general cancer.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a Rule 53(b) Divisional of U.S. application Ser. No. 16/848,273 filed Apr. 14, 2020, which claims priority based on Korean Patent Application No. 10-2019-0043765 filed Apr. 15, 2019. The contents of all of the above-identified applications are incorporated herein by reference in their entireties.
  • INCORPORATION BY REFERENCE OF SEQUENCE LISTING
  • The content of the electronically submitted sequence listing, file name: Q285877_sequence listing as filed.XML; size: 7,331 bytes; and date of creation: Mar. 17, 2023, is hereby incorporated by reference in their entirety.
  • TECHNICAL FIELD
  • The present invention relates to a use of CD133 involved in anticancer agent resistance in colon cancer.
  • BACKGROUND ART
  • Although many anticancer agents against various types of cancer have been developed to date, only a few types of cancer can be completely cured with an anticancer agent alone, this is because cancer cells do not respond to an anticancer agent in cancer treatment with the anticancer agent, or tumors are effectively reduced in an early stage, but develop anticancer agent resistance during or after treatment. Therefore, for effective anticancer treatment, resistance to an anticancer agent, for example, anticancer agent resistance of cancer cells, has to be overcome.
  • Generally, a mutation in the KRAS, NRAS or BRAF gene results in production of a protein having a modified signaling characteristic in tumor cells, and such a mutation has been known to be associated with unsuccessful results in cancer treatment using a therapeutic antibody targeting an epithelial growth factor receptor (EGFR), for example, gefitinib, cetuximab or panitumumab (Amado, Wolf et al. 2008; Karapetis, Khambata-Ford et al. 2008; Di Nicolantonio, Martini et al. 2008; Loupakis, Ruzzo et al. 2009; Lievre, Bachet et al. 2006). For example, KRAS G13D known as a colon cancer mutant cell line was reported to have resistance (tolerance) to an anticancer agent targeting EGFR.
  • For this reason, to improve a therapeutic effect of an anticancer agent targeting EGFR for cancer treatment, it is urgent to solve such a problem of anticancer agent resistance. Research on the identification of a specific cause or mechanism of the anticancer agent resistance has been persistently conducted, but it is a reality that little is known about it.
  • Nevertheless, the inventors had identified that, through a resistance study for colon cancer mutant cell lines resistant to an anticancer agent targeting EGFR, CD133 is related to resistance of colon cancer cells against an anticancer agent targeting EGFR, and thus the present invention was completed.
  • TECHNICAL PROBLEM
  • The present invention is directed to providing a biomarker composition for diagnosing resistance to an EGFR-targeting agent in colon cancer, which includes CD133 protein or a gene encoding the same.
  • The present invention is also directed to providing a kit for diagnosing resistance to an EGFR-targeting agent in colon cancer.
  • The present invention is also directed to providing a method of providing information required for diagnosis of resistance to an EGFR-targeting agent in colon cancer.
  • TECHNICAL SOLUTION
  • The inventors had confirmed, through a study on a KRAS G13D mutant colon cancer cell line having resistance to an EGFR-targeting agent, that the cell migration to and invasiveness of adjacent normal cells are highly improved by transferring the mutant to adjacent cells via microvesicles, the migration of KRAS G13D via microvesicles imparts anticancer agent resistance to adjacent colon cancer cells, and the CD133 protein controlling the release of microvesicles is related to resistance to an EGFR-targeting agent, and thus the present invention was completed.
  • The present invention is characterized by confirming whether colon cancer cells have resistance to an EGFR-targeting agent using the activity of the CD133 gene and an active protein, which is a product thereof, as a biomarker.
  • To this end, the present invention provides a biomarker composition for diagnosing resistance to an EGFR-targeting agent in colon cancer, which includes the CD133 protein or a gene encoding the same.
  • The biomarker of the present invention may be a marker with anticancer agent resistance, which is an EGFR-targeting agent, and since it has excellent accuracy and reliability as a marker for diagnosing anticancer agent resistance, the biomarker may be used to diagnose the occurrence, development and/or metastasis of resistant cancer, and treat resistant cancer.
  • The term “resistance” or “tolerance” used herein means that an organism does not sensitively respond to a drug, and thus withstands the drug's effects.
  • CD133 used as a biomarker for resistance diagnosis in the present invention is one of the representative CD-type proteins expressed on the surface of cancer stem cells, which are reported to be mainly present in cancer stem cells of colon cancer, liver cancer, pancreatic cancer and lung cancer.
  • The term “colon cancer” used herein is a collective term referring to rectal cancer, colorectal cancer and anal cancer.
  • In addition, the EGFR-targeting agent of the present invention refers to an anticancer agent, which may be any EGFR-targeting agent exhibiting an anticancer effect, and is used interchangeably with an EGFR-targeting drug. Preferably, the EGFR-targeting agent is one or more selected from the group consisting of cetuximab, gefitinib, erlotinib, panitumumab, PKI-166, EKB-569, HKI-272 (WAY-177820), icotinib, brigatinib, afatinib, lapatinib, canertinib, AEE788, XL647, and Zactima. More preferably, the EGFR-targeting agent is cetuximab, gefitinib, erlotinib or panitumumab, and most preferably, gefitinib.
  • The term “diagnosis” used herein refers to the detection of a pathological condition, which means, in terms of the purpose of the present invention, the determination of the presence or absence of resistance, and the development or alleviation of symptoms of a disease by examining the presence or absence of the expression of a biomarker for diagnosing resistance to an EGFR-targeting agent.
  • The “diagnosis biomarker” used herein refers to a material that can be used in diagnosis by distinguishing the presence or absence of resistance to an EGFR-targeting agent, and includes organic biomolecules such as polypeptides or nucleic acids (e.g., mRNA, etc.), lipids, glycolipids, glycoproteins, saccharides (a monosaccharide, a disaccharide, an oligosaccharide, etc.), which increase or decrease in cancer cells resistant to an anticancer agent, compared to cancer cells. The biomarker for resistance diagnosis according to the present invention may be a protein expressed from the CD133 gene whose expression level is increased in resistant cancer cells with respect to an EGFR-targeting agent, compared to general cancer cells.
  • The composition for diagnosing resistance to an EGFR-targeting agent may include an agent that measures an mRNA expression level of the CD133 gene or an amount of a protein expressed from the gene, and such an agent may be an oligonucleotide having a complementary sequence to CD133 mRNA, for example, a primer or nucleic acid probe specifically binding to CD133 mRNA, or an antibody specific for CD133 protein.
  • The primer refers to a single-stranded oligonucleotide that is able to serve as a starting point of template-directed DNA synthesis under suitable conditions (that is, four different types of nucleoside triphosphates and a polymerase) in a suitable buffer at a suitable temperature. A suitable length of the primer may vary according to various factors, for example, a temperature and the usage of the primer. In addition, the primer sequence is not required to be perfectly complementary to a partial sequence of the template, and it is sufficient that the primer sequence has sufficient complementarity within a range in which the primer can do an intrinsic action when hybridized with the template. Therefore, the primer of the present invention does not need to have a perfectly complementary sequence to the nucleotide sequence of a gene, which is the template, and it is reasonable that the primer has a sufficient complementarity within a range in which the primer can do an intrinsic action when hybridized with the gene sequence. In addition, the primer according to the present invention is preferably used in a gene amplification reaction. The amplification reaction refers to a reaction of amplifying a nucleic acid molecule, and amplification reactions of such a gene are well known in the art, and may include, for example, polymerase chain reaction (PCR), reverse transcriptase chain reaction (RT-PCR), ligase chain reaction (LCR), transcription-mediated amplification (TMA), and nucleic acid sequence-based amplification (NASBA).
  • The nucleic acid probe refers to a natural or modified monomer or linear oligomer consisting of linkages of such monomers, and includes a deoxyribonucleotide and a ribonucleotide, may be specifically hybridized with a target nucleotide sequence, and naturally occurs or is artificially synthesized. The probe according to the present invention may be a single chain, and preferably, an oligodeoxyribonucleotide. The probe of the present invention may include natural dNMPs (i.e., dAMP, dGMP, dCMP and dTMP), or nucleotide analogs or derivatives. In addition, the probe of the present invention may include a ribonucleotide. For example, the probe of the present invention may include backbone-modified nucleotides, such as a peptide nucleic acid (PNA), phosphorothioate DNA, phosphodithioate DNA, phosphoroamidate DNA, amide-linked DNA, MMI-linked DNA, 2′-O-methyl RNA, a-DNA and methyl phosphonate DNA; sugar-modified nucleotides, such as 2′-O-methyl RNA, 2′-fluoro RNA, 2′-amino RNA, 2′-O-alkyl DNA, 2′-O-allyl DNA, 2′-O-alkynyl DNA, hexose DNA, pyranosyl RNA and anhydrohexitol DNA; base-modified nucleotides, such as pyrimidines with a C-5 substituent (the substituent may be fluoro-, bromo-, chloro-, iodo-, methyl-, ethyl-, vinyl-, formyl-, ethynyl-, propynyl-, alkynyl-, thiazolyl-, imidazolyl-, or pyridyl-), 7-deazapurines with a C-7 substituent (the substituent may be fluoro-, bromo-, chloro-, iodo-, methyl-, ethyl-, vinyl-, formyl-, alkynyl-, alkenyl-, thiazolyl-, imidazolyl-, or pyridyl-); inosine; and diaminopurine.
  • The CD133-specific antibody may be a polyclonal antibody, a monoclonal antibody, a human antibody or a humanized antibody.
  • Examples of the antibody fragments include Fab, Fab′, F(ab′)2 and Fv fragments; a diabody; a linear antibody (Zapata et al., Protein Eng.8(10):1057-1062(1995)); a single-chain antibody molecule; and a multi-specific antibody formed from an antibody fragment.
  • When an antibody is digested with papain, two identical antigen-binding fragments, that is, “Fab” fragments having a single antigen-binding region, and the remainder, “Fc” fragment, are obtained. When treated with pepsin, a F(ab′)2 fragment which has two antigen-binding regions and is still able to be crosslinked to an antigen is produced. Fv is a minimal antibody fragment including a complete antigen-recognizing and binding region. This region consists of a dimer of one heavy chain variable region and one light chain variable region, which are firmly bound to each other by a non-covalent bond.
  • A method of preparing a polyclonal antibody is known to those of ordinary skill in the art. The polyclonal antibody may be prepared by one or more injections of an immunizing agent in combination with an immune adjuvant, if necessary, into a mammal. Generally, an immunizing agent and/or an immune adjuvant is/are subcutaneously or intraperitoneally injected into a mammal several times. The immunizing agent may be a protein of the present invention or a fusion protein thereof It may be effective that an immunizing agent, as well as a protein known to be immunogenic, is injected into an immunized mammal.
  • The monoclonal antibody according to the present invention may be prepared by the hybridoma method disclosed in the literature (Kohler et al., Nature, 256:495 (1975)), or a recombinant DNA method (refer to U.S. Pat. No. 4,816,576). The monoclonal antibody may also be isolated from a phage antibody library using the technology disclosed in the literature (Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991)).
  • In the monoclonal antibody of the present invention, specifically, when exhibiting desired activity, a part of a heavy chain and/or a light chain is identical to or has homology with a corresponding sequence of an antibody derived from a specific species or an antibody belonging to a specific antibody class or subclass, whereas the remainder of the chain(s) includes an antibody derived from a different species, an antibody belonging to a different antibody class or subclass or a “chimeric” antibody which is identical or homologous to a fragment of such an antibody (Morrison et al., Proc. Natl. Acad. Sci. USA, 81:6851-6855 (1984)).
  • A “humanized” form of a non-human (e.g., rodents) antibody is a chimeric immunoglobulin including a minimal sequence derived from a non-human immunoglobulin, an immunoglobulin chain or a fragment thereof (e.g., Fv, Fab, Fab′, F(ab′)2 or a different antigen-binding sequence of an antibody). In most cases, the humanized antibody includes a human immunoglobulin (recipient antibody) in which a complementarity-determining region (CDR) residue of a recipient is substituted with a CDR residue of a non-human species (donor antibody) such as a mouse, rat or rabbit, having desired specificity, affinity and ability. In some cases, aFv framework residue of the human immunoglobulin is substituted with a corresponding non-human residue. In addition, the humanized antibody may include a recipient antibody, or a residue which is not found in an introduced CDR or framework sequence. Generally, the humanized antibody substantially includes one or more, generally two or more variable domains, and here, all or substantially all CDR regions correspond to a region of a non-human immunoglobulin, and all or substantially all FR regions correspond to a region of a human immunoglobulin sequence. In addition, the humanized antibody includes at least a part of a variable region (Fc) of an immunoglobulin, generally, a part of a human immunoglobulin region (Presta, Curr. Op. Struct. Biol. 2:593-596 (1992)).
  • The composition for diagnosing resistance to an EGFR-targeting agent according to the present invention may be included in the form of a kit.
  • The kit may include a primer, probe or antibody which is able to measure the expression level of the CD133 gene or an amount of the CD133 protein, and the definition thereof is as described above.
  • When the kit is employed in PCR amplification, optionally, reagents required for PCR amplification, for example, a buffer, a DNA polymerase (e.g., a heat-stable DNA polymerase obtained from Thermusaquaticus (Taq), Thermusthermophilus (Tth), Thermusfiliformis, Thermisflavus, Thermococcusliteralis or Pyrococcusfuriosus (Pfu)), a DNA polymerase cofactor and dNTPs may be included, and when the kit is used in an immunoassay, the kit of the present invention may optionally include a secondary antibody and a marker substrate. Further, the kit according to the present invention may be manufactured in multiple individual packages or compartments containing the above-mentioned reagent components.
  • In addition, the composition for diagnosing resistance to an EGFR-targeting agent according to the present invention may be included in the form of a microarray.
  • In the microarray of the present invention, a primer, probe or antibody that is able to measure the expression level of the CD133 protein or gene encoding the same is used as a hybridizable array element, and fixed on a substrate. A preferable substrate may be a suitable rigid or semi-rigid support, for example, a membrane, a filter, a chip, a slide, a wafer, a fiber, a magnetic or non-magnetic bead, a gel, a tubing, a plate, a polymer, a microparticle and a capillary. The hybridizable array element may be arranged and immobilized on the substrate, and such immobilization may be performed by a chemical binding method or a covalent binding method such as UV. For example, the hybridizable array element may be bound to a glass surface which is modified to include an epoxy compound or an aldehyde group, or bound to a polylysine-coated surface using UV. In addition, the hybridizable array element may bind to a substrate by a linker (e.g., an ethylene glycol oligomer or a diamine).
  • Meanwhile, when a sample applied to the microarray of the present invention is a nucleic acid, it may be labeled and hybridized with an array element fixed on the microarray. Hybridization conditions may vary, and detection and analysis of a hybridization degree may be performed in various ways according to a labeling material.
  • In addition, the present invention provides a method of providing information for diagnosing resistance to an EGFR-targeting agent, which includes measuring the expression level of the CD133 gene or the expressed protein in a biological sample isolated from a patient, and more specifically, the method may include (a) measuring the expression level of the CD133 gene or an amount of the expressed protein in a biological sample of a patient; and (b) measuring the expression level of the gene and an amount of the expressed protein from a sample of a normal control and comparing it with the measurement result obtained in Step (a).
  • The method of measuring the expression level of a gene or an amount of a protein encoded by the gene may be performed by a known process of isolating mRNA or a protein from a biological sample using a known technique.
  • The biological sample refers to a sample obtained from the living body, which is different from a normal control in terms of the expression level of the gene or a protein level according to the occurrence or progression of resistance to an EGFR-targeting agent, and the sample may be, but is not limited to, tissue, a cell, blood, serum, plasma, saliva or urine.
  • The measurement of the expression level of a gene is preferably to measure an mRNA level, and methods of measuring an mRNA level include, but are not limited to, RT-PCR, real time reverse transcription polymerase chain reaction, RNase protection assay, Northern blotting and DNA chip assay.
  • The measurement of a protein level may use an antibody, and in this case, the CD133 protein in a biological sample and an antibody specific for the protein forms a binding product, that is, an antigen-antibody complex, and an amount of antigen-antibody complex formation may be quantitatively measured from the size of a signal of a detection label. The detection level may be selected from the group consisting of an enzyme, a fluorescent material, a ligand, a light-emitting material, a microparticle, a redox molecule and a radioisotope, but the present invention is not limited thereto. Analysis methods for measuring a protein level may include, but not limited to, Western blotting, ELISA, radioimmunoassay, radioimmunodiffusion, Ouchterlonyimmunodiffusion, rocket immunoelectrophoresis, immunohistochemical staining, immunoprecipitation assay, complement fixation assay, FACS, and protein chip assay.
  • Accordingly, the present invention may confirm mRNA expression levels or protein amounts in the control, a patient resistant to an EGFR-targeting agent, or a patient suspected of having resistance to an EGFR-targeting agent through the detection methods described above, and the expression levels are compared to be able to diagnose the occurrence and progression of the resistance to an EGFR-targeting agent.
  • In addition, according to the method of providing information for the diagnosis of the resistance to an EGFR-targeting agent according to the present invention, when the expression level of the CD133 gene according to the present invention or an amount of the expressed protein is higher than that of the normal control sample, it can be determined that a patient has resistance to an EGFR-targeting agent.
  • The present invention also relates to a composition for preventing or treating resistance to an EGFR-targeting agent, which includes a CD133 inhibitor.
  • The present invention also provides a use of a CD133 inhibitor for preparing a pharmaceutical composition for preventing or treating resistance to an EGFR-targeting agent.
  • According to the present invention, CD133 is highly expressed in cancer cells with resistance to an EGFR-targeting agent, as described above, and the expression of CD133 promotes budding of microvesicles, increases cell migration and invasiveness by delivering the oncogenic protein KRAS G13D to adjacent cells using microvesicles, and thus is involved in the occurrence of anticancer agent resistance.
  • Therefore, the CD133 inhibitor may be used in treatment of resistant cancer.
  • To this end, the composition for preventing or treating resistance to an EGFR-targeting agent of the present invention may include an agent for reducing mRNA expression of the CD133 gene or protein expression thereof, or degrading function or activity.
  • The CD133 protein inhibitor may be a peptide or compound which is bound with the CD133 protein to regulate a signal of a nerve differentiation pathway. The inhibitor may be selected by a screening method exemplified below, such as protein structure analysis, and may be designed using a method known in the art.
  • Specifically, the CD133 protein inhibitor may be a material which is bound with the CD133 protein consisting of an amino acid sequence represented by SEQ ID NO: 2 to inhibit activity.
  • In addition, as the protein inhibitor, a polyclonal antibody, monoclonal antibody, human antibody or humanized antibody against the CD133 protein may be used, and the definition of the antibody is as described above.
  • Resistant cancer may be prevented or treated by inhibiting the function of CD133 in cells using the antibody.
  • The functional or activity inhibitor of the CD133 protein according to the present invention may be delivered using a liposome, a virus, a gene gun, a polymer, ultrasonication or an electric shock, but the present invention is not particularly limited thereto.
  • The CD133 gene may be DNA encoding the same or mRNA transcribed therefrom. Accordingly, the inhibitor of the gene may be an inhibitor which is bound to the gene itself to interfere with transcription or bound to mRNA transcribed from the gene to interfere with the translation of mRNA.
  • Therefore, the inhibitor of the CD133 gene includes all types of inhibitors that inhibit the expression of the CD133 gene. For example, such an inhibitor may be a peptide, nucleic acid or compound binding to the gene. The inhibitor may be selected by a screening method to be described below, such as cell-based screening, and may be designed by a method known in the art.
  • In one embodiment, the inhibitor may be an antisense-oligonucleotide, siRNA, shRNA or miRNA against the CD133 gene or a vector including the same. Such an antisense-oligonucleotide, siRNA, shRNA, miRNA or a vector including the same may be manufactured using a method known in the art. Specifically, the inhibitor may be a nucleic acid molecule prepared to inhibit the expression of the CD133 gene consisting of SEQ ID NO: 1.
  • The term “siRNA” used herein refers to double-stranded RNA inducing RNA interference through the cleavage of mRNA of a target gene, and consists of an RNA strand of a sense sequence having the sequence like mRNA of the target gene and an RNA strand of an antisense sequence having a sequence complementary thereto.
  • The siRNA may include siRNA synthesized in vitro, or a form expressed by inserting a base sequence encoding siRNA into an expression vector.
  • The “vector” used herein refers to a gene construct including foreign DNA inserted into a genome encoding a polypeptide.
  • The vector related to the present invention is a vector in which a nucleic acid sequence inhibiting the gene is inserted into a genome, and such a vector may be a DNA vector, a plasmid vector, a cosmid vector, a bacteriophage vector, an enzyme vector, or a viral vector.
  • In addition, the antisense may have a sequence complementary to an entire or partial mRNA sequence transcribed from the CD133 gene or a fragment thereof, and bind to the mRNA to inhibit the expression of the CD133 gene or a fragment thereof.
  • In addition, the short hairpin RNAi (shRNAi) may be manufactured by a conventional method using a human or mouse shRNAi common base sequence region as a target.
  • In addition, the pharmaceutical composition of the present invention may further include a pharmaceutically acceptable carrier.
  • The pharmaceutically acceptable carrier includes a carrier and a vehicle, which are generally used in the pharmaceutical field, and specifically, includes an ion exchange resin, alumina, aluminum stearate, lecithin, a serum protein (e.g., human serum albumin), a buffer material (e.g., various types of phosphates, glycine, sorbic acid, potassium sorbate, or a partial glyceride compound of a saturated vegetable fatty acid), water, a salt or electrolyte (e.g., protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride or a zinc salt), colloidal silica, magnesium trisilicate, polyvinylpyrrolidone, a cellulose-based substrate, polyethylene glycol, sodium carboxymethylcellulose, polyarylate, wax, polyethylene glycol or lanolin, but the present invention is not limited thereto.
  • In addition, the composition of the present invention may further include a lubricant, a wetting agent, an emulsifier, a suspending agent, or a preservative, other than the above-described components.
  • In one aspect, the composition according to the present invention may be prepared as an aqueous solution for parenteral administration, and preferably, a Hank's solution, a Ringer's solution or a buffer solution such as physically buffered saline is used. An aqueous injectable suspension may include a substrate that can increase the viscosity of the suspension such as sodium carboxymethylcellulose, sorbitol or dextran.
  • The composition of the present invention may be systemically or locally administered, and formulated in a suitable form by known technology for such administration. For example, for oral administration, the composition may be administered by mixing the active compound with an inactive diluent or edible carrier, encapsulating all components with a hard or soft gelatin capsule or compressing all components in the form of a tablet. For oral administration, the active compound may be mixed with an excipient, and then formed as an edible tablet, a buccal tablet, a troche, a capsule, an elixir, a suspension, a syrup or a wafer.
  • Various forms for injection or parenteral administration may be prepared according to a technique known or commonly used in the art. Since CD133 is well dissolved in saline or a buffer solution, after storage in a freeze-dried state, an effective amount of CD133 may be mixed in saline or a buffer solution to prepare a solution suitable for intravenous injection, subcutaneous injection, muscle injection, intraperitoneal injection or transdermal injection, right before the administration.
  • The effective amount of the active ingredient of the pharmaceutical composition of the present invention refers to an amount required for prevention, inhibition or alleviation of a disease.
  • Accordingly, the effective amount may be adjusted by various factors such as the type and severity of a disease, the type and content of the active ingredient and other components, contained in the composition, the type of a dosage form, and the age, body weight, general health condition, gender and diet of a patient, administration time, administration route, a release rate of the composition, a treatment period, and a co-used drug. While not limited thereto, for example, in the case of an adult, the inhibitor of the present invention may be administered once or several times a day, when the type of the inhibitor is a compound, it may be administered at a dose of 0.1 ng/kg to 10 g/kg, when the type of the inhibitor is a polypeptide, protein or antibody, it may be administered at a dose of 0.1 ng/kg to 10 g/kg, or when the type of the inhibitor is an antisense-oligonucleotide, siRNA, shRNAi or miRNA, it may be administered at a dose of 0.01 ng/kg to 10 g/kg.
  • The present invention also provides a method of treating an animal with resistance to an EGFR-targeting agent, which includes administering a composition including a pharmaceutically effective amount of a CD133 inhibitor for preventing or treating resistance to an EGFR-targeting agent to a subject.
  • A pharmaceutical composition and an administration method, which are used in the method of treating resistance to an EGFR-targeting agent, are as described above, and the common description between them will be omitted to avoid excessive complexity of the specification.
  • Meanwhile, subjects to which the pharmaceutical composition for preventing or treating resistance to an EGFR-targeting agent may be administered include all kinds of animals. For example, the subjects may be non-human animals such as dogs, cats, and mice.
  • The present invention also provides a method of screening a drug for preventing or treating resistance to an EGFR-targeting agent, which includes bringing the CD133 gene into contact with a candidate ex vivo, and determining whether the candidate promotes or inhibits the expression of the gene.
  • In addition, the present invention provides a method of screening a drug for preventing or treating resistance to an EGFR-targeting agent, which includes bringing the CD133 protein into contact with a candidate ex vivo, and determining whether the candidate improves or inhibits the function or activity of the protein.
  • According to the screening method of the present invention, first, a candidate to be analyzed may be brought into contact with resistant cancer cells having the gene or protein.
  • The candidate may be an individual nucleic acid, protein or peptide, another extract or natural substance, or a compound, which is expected or randomly selected to have a potential as a material promoting or inhibiting the mRNA transcription or mRNA translation into a protein in the base sequence of the CD133 gene or a drug improving or inhibiting the function or activity of the CD133 protein according to a common selection method.
  • Afterward, in cells treated with a candidate, the expression level of the gene or the amount or activity of the protein may be measured, and as a result of the measurement, when the expression level of the gene or the amount or activity of the protein is measured as increased or decreased, the candidate may be determined as a material for treating or preventing resistant cancer.
  • The method of measuring the expression level of the gene, or the amount or activity of the protein may be performed by various methods known in the art, and may be, for example, RT-PCR, real time-polymerase chain reaction, Western blotting, Northern blotting, ELISA, radioimmunoassay (RIA), radioimmunodiffusion and immunoprecipitation assay, but the present invention is not limited thereto.
  • The candidate exhibiting an activity of inhibiting gene expression or a protein function, obtained by the screening method of the present invention, may be a candidate for a resistant cancer therapeutic agent.
  • The candidate for a therapeutic agent against cancer resistant to an EGFR-targeting agent serves as a leading compound in the development of a therapeutic agent against resistance to an EGFR-targeting agent, and modifies and optimizes the structure of the leading compound to exhibit an effect of inhibiting the CD133 gene or the function of a protein expressed therefrom, thereby developing a novel therapeutic agent against cancer resistant to an EGFR-targeting agent.
  • Details related to genetic engineering technology in the present invention will become more apparent from the content disclosed in Sambrook, et al. Molecular Cloning, A Laboratory Manual, Cold Spring Harbor laboratory Press, Cold Spring Harbor, N.Y. (2001); and Frederick M. Ausubel et al., Current protocols in molecular biology volume 1, 2, 3, John Wiley & Sons, Inc. (1994).
  • ADVANTAGEOUS EFFECTS
  • The present invention identifies that the CD133 protein expressed in colon cancer increases resistance to an EGFR-targeting agent, and thus the CD133 protein can be used as a novel target protein for diagnosing and treating resistant cancer as well as general cancer.
  • DESCRIPTION OF DRAWINGS
  • The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.
  • FIGS. 1A-1E show the result of identifying a CD133 expression regulatory mechanism in liver cancer cells: (A) the change in CD133 expression according to EGF expression; (B-C) the change in CD133 expression according to an EGF downstream signaling molecule, NF-κB; and (D-E) the relationship between NF-κB and CD133, confirmed by promoter activity.
  • FIGS. 2A-2E show the result of confirming that CD133 is involved in promotion of microvesicle budding in liver cancer cells: (A) confirmation of the relationship between an EGF signaling system and microvesicle and TNT formation; (B) confirmation of microvesicle and TNT formation in a CD133—overexpressing cell line; and (C-E) confirmation of the change in activation of Rac1 and RhoA associated with microvesicle budding and a downstream signaling factor, Erk1/2, of RhoA according to CD133 expression
  • FIGS. 3A-3B show the result of analyzing microvesicles budded from cancer cells: (A) the result of confirming whether CD133 is included in microvesicles budded from various cancer cells; and (B) the result of observing the migration of microvesicles containing CD133 to adjacent cells.
  • FIGS. 4A-4F show the result of confirming that CD133 is involved in oncogenic protein transport and microvesicle budding in colon cancer: (A) confirmation whether an oncogenic protein is contained in microvesicles according to CD133 expression; (B-C) confirmation that CD133 expression contributes to microvesicle budding and a size change; (D) confirmation that CD133 expression contributes to the transport of an oncogenic protein via microvesicles and cell proliferation; and (E-F) confirmation that the transport of an oncogenic protein via microvesicles contributes to the change in cell migration to and invasiveness of adjacent normal cells.
  • FIGS. 5A-5F show the result of confirming that CD133—containing microvesicles induce anticancer agent resistance (gefitinib) in colon cancer: (A-B) confirmation of the capability of CD133—containing microvesicles to induce anticancer agent resistance (gefitinib) by confirmation of the proliferation of cancer cells; (C-D) the result of confirming that cancer cell proliferation is caused by an oncogenic protein transported by microvesicles through the change in KRAS downstream signaling molecule and target gene expression; and (E-F) confirmation that cell migration to and invasiveness of adjacent normal cells are changed by the oncogenic protein transported by CD133—containing microvesicles in the presence of gefitinib.
  • MODES OF THE INVENTION
  • Hereinafter, the advantages and features of the present invention and the methods of accomplishing the same may be clearly understood with reference to the detailed description of exemplary embodiments and the accompanying drawings. However, the present invention is not limited to the exemplary embodiments disclosed below, and may be embodied in many different forms. These exemplary embodiments are merely provided to complete the disclosure of the present invention and fully convey the scope of the present invention to those of ordinary skill in the art, and the present invention should be defined by only the accompanying claims.
  • <Example 1>Confirmation of CD133 Expression Regulatory Mechanism in Cancer Cells
  • An epithelial growth factor (EGF) signaling system has been reported to be significant in the occurrence and development of cancer. CD133 (NCBI Gene ID: 8842) is known as a marker of a cancer stem cell, and particularly, to play a crucial role in formation of liver cancer stem cells. Therefore, the correlation between the EGF signaling system and CD133 expression in a liver cancer cell line was investigated.
  • After treatment with an inhibitor against an EGF downstream signaling molecule, CD133 expression was investigated. Specifically, for transfection and Western blotting, a specific gene (plasmid vector or siRNA) was expressed in cells using a transfection reagent. For general plasmid vector transfection, 24 hours after transfection, cells were harvested, and for siRNA transfection, 48 hours after transfection, cells were harvested. For Western blotting, the harvested cells were lysed in RIPA buffer, and centrifuged at 12,000 g for 20 minutes at 4° C., followed by collecting a supernatant. The collected supernatant was subjected to SDS-PAGE gel electrophoresis, and transferred onto a nitrocellulose membrane, followed by detecting the expression of a desired protein using suitable antibodies. As a result, it was observed that NF-κB is involved in EGF-induced CD133 expression (FIG. 1B). It was confirmed that, when the expression of NF-κB subunits, such as p50 (NCBI Gene ID: 4790) and p65 (NCBI Gene ID: 5970), was inhibited, CD133 expression decreases (FIG. 1C). Promoter activity was observed in a promoter near CD133 ORF. Specifically, to measure the promoter activity, a CD133 ORF promoter region was cloned in a special vector expressing luciferase. Cells were simultaneously transfected with the luciferase vector and a Renilla plasmid vector for quantification. Twenty-four hours after transfection, the cells were treated with EGF for 14 hours, lysed with lysis buffer and centrifuged to obtain a supernatant, and then luciferase activity was measured using the Luminometer 20/20 (FIG. 1D). In a promoter which had been subjected to treatment with an NF-κB inhibitor and an NF-κB subunit-binding site, promoter activity was not observed (FIG. 1D). Taken together, it was confirmed that EGF activates NF-κB and regulates CD133 expression at the transcription level (FIG. 1A).
  • <Example 2>Identification of CD133 Role in Cancer Cells
  • The relationship between EGF and microvesicle and TNT formation was investigated by microscopic analysis. Specifically, after 14 hours of EGF treatment, cells were fixed with 4% paraformaldeyde and treated with WGA-488 and DAPI for 10 minutes to stain the cell membrane and nucleus. Fluorescence intensity was measured using an LSM700 Meta confocal microscope.
  • The results showed that, when a liver cancer cell line was treated with EGF, compared to a control, microvesicle and TNT formation increases between cells, and this phenomenon is decreased by treatment with the NF-κB inhibitor (WGA, cell membrane staining, FIG. 2A). In addition, in a CD133 expression-stable cell line, it was confirmed that microvesicles and TNT formation rates highly increase (FIG. 2B). Therefore, it was expected that microvesicle budding is closely related to the change in CD133 expression.
  • In addition, to bud microvesicles from the cell membrane, the regulation of activities of small GTPases such as ARF6, RhoA and Rac1 is known to be important, and thus an expression pattern of the gene according to the CD133 expression pattern was observed by small GTPase pull-down assay. Specifically, to measure Rac1 activity, after transfected with CD133, cells were harvested, and activity was measured using a Rac1 activation Assay kit. To measure RhoA activity, after CD133 transfection, cells were harvested, and activity was measured using a RhoA activation Assay kit. As a result, it was seen that, in the CD133 expression-stable cell line, Rac1 (NCBI Gene ID: 5879) activity decreases, RhoA (NCBI Gene ID: 387) activity increases, and thus the activity of the downstream signaling molecule Erk1/2 (NCBI Gene ID: 5595, 5594) increases (FIG. 2E). This result showed that a certain level of CD133 expression is essential for microvesicle formation, and particularly, small GTPase activity is regulated to promote microvesicle budding.
  • <Example 3>Analysis of Physiological Properties of Microvesicles Released from Cancer Cells
  • Microvesicles were observed by microvesicle microscopic analysis. Specifically, microvesicles were isolated from the culture solution of CD133-transfected cells and stained with WGA-488, general cells were treated with the stained microvesicles, and after 12 hours, fluorescence intensity was measured using a LSM700 Meta confocal microscope.
  • It was observed that the microvesicles were released from various types of cancer cells, and contained CD133 (FIG. 3A). In addition, the CD133 (red)-containing microvesicles were isolated by centrifugation, and treated with WGA to stain the cell membrane. It was observed that, when the cell line was treated with the microvesicles, the microvesicles were transported to adjacent cells along with CD133 (FIG. 3B).
  • <Example 4>Identification of CD133 Role in Colon Cancer
  • Forty-eight hours after a HCT116 cell line was transfected with a shCD133 vector into which the shRNA sequence (5′: GAGUCGGAAACUGGCAGAUAGCAAU-3′: SEQ ID NO: 3) knocking down the CD133 expression was inserted to prepare a CD133 expression-inhibited cell line, selection was performed with a selection marker Zeocin. Afterward, single colonies were selected and subcultured, and subjected to Western blotting with wild-type HCT116 to verify CD133 knockdown. The used vector is a vector prepared by substituting a neomycin antibiotic region with Zeomycin in a pSilencer 2.1-U6 neo vector (www.thermo fisher.com/kr/ko/home/life-science/dna-rnapurification-analysis/napamisc/vector-maps/psilencer-2-1-u6-hygro-vectormap.html).
  • To isolate microvesicles, the culture solutions of a CD133 normal expression cell line and a CD133 expression-inhibited cell line were collected, and then each cell culture solution was centrifuged at 20,000 g for 1 hour, thereby obtaining a supernatant. After discarding the supernatant, microvesicles were isolated.
  • From the isolated microvesicles, the amount and sizes of microvesicle budding were measured by a Malvern Nanosight Nanoparticle Tracking Analysis system.
  • KRAS (NCBI Gene ID: 3845) G13D is a well-known oncogenic protein, and usually found as a KRAS mutant in colon cancer. It was observed that, when CD133 expression was inhibited in a colon cancer cell line, KRAS G13D was not contained in the microvesicles (FIG. 4A). In addition, it was confirmed that CD133 expression is involved in determination of the amount and sizes (100 to 200 nm) of microvesicle budding (FIGS. 4B and 4C).
  • To observe the transport of KRAS G13D to adjacent cells, microvesicles were isolated from the CD133 expression-inhibited cell line and the general cell line and then normal cells were treated with the microvesicles. Specifically, for cell migration assay, microvesicle-recipient cells were cultured in an 8-μm pore insert, treated with microvesicles, and then after 48 hours, migrating cells counted. For invasion assay, an 8-μm pore insert was coated with Matrigel, and then recipient cells were incubated. Forty-eight hours after microvesicle treatment, cells migrating through the Matrigel were counted.
  • As a result, it was confirmed that, in normal cells treated with the microvesicles isolated from the general cell line, KRAS G13D is transported to adjacent cells via the microvesicles along with CD133, and the activation of KRAS downstream signaling molecules Akt (NCBI Gene ID: 207) and Erk1/2 increases (FIG. 4D). The microvesicle-mediated KRAS G13D transport induced increases in cell migration to (FIG. 4E) and invasiveness (FIG. 4F) of normal cells. Based on this result, it was able to be confirmed that CD133 regulates a microvesicle transport material, and plays a crucial role in microvesicle size and budding.
  • <Example 5>Confirmation of Induction of Anticancer Agent Resistance to Adjacent Cells by CD133—Containing Microvesicles in Colon Cancer
  • While gefitinib is an anticancer material that inhibits EGFR activity and thus controls cancer cells, it was reported that, in certain types of cancer in which EGFR downstream signaling molecules including KRAS are activated, gefitinib efficacy was insignificant. It was inferred that the transport of KRAS G13D to adjacent cells by CD133-containing microvesicles is involved in resistance to such an EGFR-targeting agent.
  • Cells being cultured to confirm the above inference were incubated with gefitinib and the microvesicles for a suggested time, cell viability was analyzed using an EZ-cytox kit at specific time, and the absorbance was measured at 570 nm using a microplate reader, followed by calculating a cell growth rate. Twenty-four hours after treatment with the microvesicles and gefitinib, the cells were harvested, and then fixed with 95% cold ethanol. The fixed cells were treated with RNase and propidium iodide to stain DNA, and then the stained DNA was analyzed by FACS.
  • As a result, it was confirmed that, when treated along with gefitinib, microvesicles extracted from CD133 expression-inhibited colon cancer cells do not contribute to cell proliferation of adjacent recipient cells, but microvesicles extracted from CD133-expressing cells restore cell proliferation even when gefitinib is treated (FIGS. 5A and 5B). To demonstrate that such a change is caused by KRAS G13D transport by microvesicles, the change in KRAS downstream signaling molecules and target genes in microvesicle-recipient cells was observed. It was confirmed that the CD133-mediated KRAS G13D transport increases the activities of KRAS downstream signaling molecules FAK (NCBI Gene ID: 5747), Akt and Erk1/2 in the recipient cells (FIG. 5C). In addition, it was observed that mRNA expressions of KRAS target genes SERVIVIN (NCBI Gene ID: 332) and CCNB1 (NCBI Gene ID: 891) increase, anti-apoptotic mRNA (BCLXL, BCL2L2 (NCBI Gene ID: 598, 599)) increases, and pro-apoptotic mRNA (BIM (NCBI Gene ID: 10018)) decreases (FIG. 5D). This shows that the cell migration to and invasiveness of recipient cells receiving the oncogenic protein (KRAS G13D)—containing microvesicles increase, and thus the development of cancer is maintained after treatment with an anticancer agent. In conclusion, in colon cancer, CD133—containing microvesicles are involved in transport of the KRAS oncogenic protein to induce anticancer agent resistance to adjacent cells, thereby accelerating cancer development.

Claims (1)

1. A method of treating resistance to Gefitinib in colon cancer, the method comprising administering an inhibitor against a CD133 gene to a subject resistant to Gefitinib, wherein the inhibitor is a peptide, a compound, or an antibody against the CD133 protein.
US18/186,129 2019-04-15 2023-03-17 Cd133 related to anticancer agent resistance in colon cancer and use thereof Pending US20230265526A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/186,129 US20230265526A1 (en) 2019-04-15 2023-03-17 Cd133 related to anticancer agent resistance in colon cancer and use thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR1020190043765A KR102198404B1 (en) 2019-04-15 2019-04-15 CD133 related to anticancer drug resistance in colon cancer and use thereof
KR10-2019-0043765 2019-04-15
US16/848,273 US11639530B2 (en) 2019-04-15 2020-04-14 CD133 related to anticancer agent resistance in colon cancer and use thereof
US18/186,129 US20230265526A1 (en) 2019-04-15 2023-03-17 Cd133 related to anticancer agent resistance in colon cancer and use thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/848,273 Division US11639530B2 (en) 2019-04-15 2020-04-14 CD133 related to anticancer agent resistance in colon cancer and use thereof

Publications (1)

Publication Number Publication Date
US20230265526A1 true US20230265526A1 (en) 2023-08-24

Family

ID=72748882

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/848,273 Active 2040-12-14 US11639530B2 (en) 2019-04-15 2020-04-14 CD133 related to anticancer agent resistance in colon cancer and use thereof
US18/186,129 Pending US20230265526A1 (en) 2019-04-15 2023-03-17 Cd133 related to anticancer agent resistance in colon cancer and use thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/848,273 Active 2040-12-14 US11639530B2 (en) 2019-04-15 2020-04-14 CD133 related to anticancer agent resistance in colon cancer and use thereof

Country Status (2)

Country Link
US (2) US11639530B2 (en)
KR (1) KR102198404B1 (en)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1777523A1 (en) * 2005-10-19 2007-04-25 INSERM (Institut National de la Santé et de la Recherche Médicale) An in vitro method for the prognosis of progression of a cancer and of the outcome in a patient and means for performing said method
US20100040637A1 (en) * 2005-11-23 2010-02-18 Karen Van Orden Methods and Composition for Treating Diseases Targeting Prominin-1 (CD133)
KR102055350B1 (en) * 2017-09-28 2019-12-13 고려대학교 산학협력단 Biomarker for Diagnosis of Anticancer drug Resistance of Colon Cancer and Uses thereof

Also Published As

Publication number Publication date
US11639530B2 (en) 2023-05-02
KR20200121122A (en) 2020-10-23
KR102198404B1 (en) 2021-01-05
US20200325545A1 (en) 2020-10-15

Similar Documents

Publication Publication Date Title
JP2019150027A (en) Methods and compositions for predicting response to eribulin
WO2016152352A1 (en) Melanoma-specific biomarker and use thereof
KR102216590B1 (en) Composition for treatment and diagnosis of pancreatic neuroendocrine tumors
KR101315570B1 (en) NLK as a marker for the diagnosis of hepatocellular carcinomas and as a therapeutic agent thereof
KR101875935B1 (en) A Biomarker of the resistance about HER2 inhibitor
US10688197B2 (en) Non-alcoholic fatty liver regulator 14-3-3 protein
US11639530B2 (en) CD133 related to anticancer agent resistance in colon cancer and use thereof
EP3516071B1 (en) Methods and pharmaceutical compositions for the treatment of lung cancer
CN112462072B (en) Application of TMUB1 protein in preparation of tumor immunosuppressive molecule detection agent
US20180207294A1 (en) Non-alcoholic fatty liver regulating factor 14-3-3 protein
KR102055350B1 (en) Biomarker for Diagnosis of Anticancer drug Resistance of Colon Cancer and Uses thereof
US20210164982A1 (en) Pharmaceutical use of actinin-4 involved in induction of cervical cancer
KR101150410B1 (en) Use of S100P as a hepatocellular carcinomar diagnostic marker
KR101385783B1 (en) Deleted in breast cancer-1 as a marker for the diagnosis of hepatocellular carcinomas and as a therapeutic agent thereof
KR102238258B1 (en) A novel biomarker for diagnosing liver cancer and a treating method using thereof
KR101535346B1 (en) Pharmaceutical use of c-Yes for differentiation therapy of cancer stem cell
US20240102105A1 (en) Biomarker for predicting responsiveness to anticancer agent and use thereof
KR20170002364A (en) Marker for diagnosing HER2 inhibitor resistance cancer, diagnostic kit comprising the same and method for diagnosing HER2 inhibitor resistance cancer
WO2017054759A1 (en) Prevention, diagnosis and treatment of cancer overexpressing gpr160
KR101637543B1 (en) Marker for diagnosing HER2 inhibitor resistance cancer, diagnostic kit comprising the same and method for diagnosing HER2 inhibitor resistance cancer
KR101640042B1 (en) Marker for diagnosing HER2 inhibitor resistance cancer, diagnostic kit comprising the same and method for diagnosing HER2 inhibitor resistance cancer
KR101637545B1 (en) Marker for diagnosing HER2 inhibitor resistance cancer, diagnostic kit comprising the same and method for diagnosing HER2 inhibitor resistance cancer
KR101640049B1 (en) Marker for diagnosing HER2 inhibitor resistance cancer, diagnostic kit comprising the same and method for diagnosing HER2 inhibitor resistance cancer
KR20230029140A (en) Composition for detecting EGFR targeted therapy resistance and screening method for inhibiting EGFR targeted therapy resistance
KR20230010414A (en) Biomarker composition for predicting metastasis of triple negative breast cancer comprising CSDE1

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION