WO2008075713A1 - Marker for diagnosis of cancer, and target molecule for therapy - Google Patents

Marker for diagnosis of cancer, and target molecule for therapy Download PDF

Info

Publication number
WO2008075713A1
WO2008075713A1 PCT/JP2007/074408 JP2007074408W WO2008075713A1 WO 2008075713 A1 WO2008075713 A1 WO 2008075713A1 JP 2007074408 W JP2007074408 W JP 2007074408W WO 2008075713 A1 WO2008075713 A1 WO 2008075713A1
Authority
WO
WIPO (PCT)
Prior art keywords
gene
small cell
cancer
trim33
polynucleotide
Prior art date
Application number
PCT/JP2007/074408
Other languages
French (fr)
Japanese (ja)
Inventor
Johji Inazawa
Sana Yokoi
Issei Imoto
Hitoshi Tsuda
Original Assignee
Tokyo Medical And Dental University
Daiichi Sankyo Co., Ltd.
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 Tokyo Medical And Dental University, Daiichi Sankyo Co., Ltd. filed Critical Tokyo Medical And Dental University
Publication of WO2008075713A1 publication Critical patent/WO2008075713A1/en

Links

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
    • G01N33/57415Specifically defined cancers of breast
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds

Definitions

  • the present invention relates to a means for diagnosing small cell cancer such as small cell lung cancer or breast cancer, and preferably means for utilizing amplification or expression enhancement of a specific gene as a cancer diagnostic marker.
  • the present invention also relates to a means for treating small cell cancer such as small cell lung cancer or breast cancer by suppressing amplification or expression enhancement of the specific gene.
  • TRIM33 also called tripartite motif 33, sometimes called “Ectodermin” has been identified as a factor involved in ectoderm formation in Xenopus embryogenesis, and by ubiquitinating Smad4, a mesoderm-inducing factor It is known to inhibit (Non-Patent Document 1: Cell, 121, pp.87-99, 2005). Regarding the relationship between TRIM33 gene and cancer, TRIM3 3 gene has been shown to increase expression in colorectal cancer! /, And its ability to be demonstrated S /! (Non-patent document 1, Fig. 7D And 7E).
  • RNA interference suppression of TRIM33 gene expression in the melanoma C32 cell line or breast cancer MB231 cell line by RNA interference has been shown to suppress the growth of these cells (Figs. 7F and 7G). Proliferation inhibition has been shown to be Smad4-dependent (Fig. 7G).
  • TRM33 gene is amplified or increased in breast cancer cells. Relationship between TRM33 gene and other cancers (eg small cell lung cancer) There is no report about. Further, TRM33 gene is a gene present in the genetic child seat 1Ro13.1, the force is also reported that the human genome 1 ⁇ 13 area you are amplified in breast cancer s (Non-Patent Document 2: Cancer Research, 60, pp.4519- 4525, 2000; Non-Patent Document 3: Cancer Genet Cytogenet, 117, pp.153-158, 2000), these publications show that specific TRIM33 genes are amplified or upregulated in breast cancer! / No suggestion! /, And no teaching.
  • the gene expression pattern in cancer is the gene expression pattern in the tissue of the origin.
  • the gene expression pattern differs for each tissue that often inherits the turn. Therefore, while there are genes that change in common across cancer types, in general, gene expression patterns in cancer differ depending on the tissue of origin (for example, classification such as colon cancer and lung cancer). (Molecular biology of the cell (3rd edition) .1256
  • Non-Patent Document 1 suggests the relationship between TRIM33 gene and other cancers (for example, small cell lung cancer)! / I think.
  • Non-Patent Document 1 Cell, 121, pp.87-99, 2005
  • Non-patent document 2 Cancer Research, 60, pp.4519-4525, 2000
  • Non-patent document 3 Cancer Genet Cytogenet, 117, pp.153-158, 2000
  • An object of the present invention is to provide means for using amplification or increased expression of a specific gene as a marker for cancer diagnosis, preferably means for diagnosing small cell cancer such as small cell lung cancer or breast cancer. is there.
  • Another object of the present invention is to provide a means for treating small cell cancer such as small cell lung cancer or breast cancer by suppressing the amplification or increased expression of the specific gene. Means to do
  • TRIM33 gene is amplified in small cell carcinoma such as small cell lung cancer and breast cancer cells, and its expression is enhanced. I found out. Furthermore, we found that the expression level of TRIM33 gene is a prognostic factor for breast cancer. Further, the gene can be used as a marker for small cell carcinoma such as small cell lung cancer and breast cancer, and by detecting the amplification or expression enhancement using, for example, a probe that can specifically bind to the gene, It was found that small cell carcinoma such as small cell lung cancer and breast cancer can be diagnosed easily and reliably.
  • RNA interference by inhibiting the expression of the TRIM33 gene by means such as RNA interference, it is possible to remarkably suppress the growth of small cell carcinomas such as small cell lung cancer and breast cancer cells, thereby producing small cell lung cancer. It has been found that it can treat small cell cancer and breast cancer.
  • the present invention is based on the above findings. Based on this, it was completed.
  • the present inventors have doubled a specific region on 1 ⁇ 13.2 (hereinafter also referred to as an amplicon according to the present invention) in a cell line derived from human small cell lung cancer (LU-140). It was found that amplification was performed on a minute chromosome (dmin) (Fig. 1 (C), (D)). Furthermore, the present inventors found that the TRIM33 gene was amplified in a homogeneous chromosome region (HSR) in a human breast cancer-derived cell line (MCF7) (Fig. 2, (A)). .
  • HSR homogeneous chromosome region
  • MCF7 human breast cancer-derived cell line
  • the amplicon according to the present invention has, as its original locus, 1 ⁇ 13 ⁇ 2 substantially represented by four types of BAC clones (RP1 1-1115211, RP11-1129E18, RP11-625J15, RP11-254K1). Present in the chromosomal region.
  • These BAC clones are clones of the BAC library (RPCI human BAC library 11) of human chromosome fragments created from human blood-derived chromosomes.
  • Each clone in the BAC library represents a certain fragment (+ strand, strand) of the chromosome.
  • the terminal portion of each clone (+ strand, strand) was sequenced, and its base sequence became clear.
  • the full-length nucleotide sequence of each clone (+ strand, single strand) of the BAC library has been revealed!
  • the partial base sequence of the end of RP11-1152I1 (+ strand) is the base sequence described in SEQ ID NO: 1.
  • the partial base sequence at the end of the RP11— 1129E18 (+ strand) region is Shiki Iki (GenBank accession number: AQ697738) described in SEQ ID NO: 3.
  • the partial base sequence at the end of the RP11-1129E18 (-strand) region is the base sequence described in SEQ ID NO: 4 (GenBank accession number: AQ720255).
  • the partial base sequence at the end of the RP11-625J15 (+ strand) region is the base sequence described in SEQ ID NO: 5 (GenBank accession number: AQ405632).
  • the partial base sequence at the end of the RP11-625J15 (strand) region is the base sequence described in SEQ ID NO: 6 (GenBank Accession No: AQ455093).
  • the partial base sequence at the end of the RP11-254K1 (+ strand) region is the base set forth in SEQ ID NO: 7. Sequence (GenBank accession number: AQ478938). The partial base sequence at the end of the RP11—254K1 (—strand) region is the base sequence described in SEQ ID NO: 8 (GenBank Accession No: AQ478940). Since these 4 clones partially overlap, it is possible to create a contig sequence formed from them.
  • nucleotide sequence described in GenBank accession number NT-0192713.18 is Contig's IJ of the genomic fragment of the first human chromosome.
  • RP11—115211, RP11—1129E18, RP1 1—625J15 and RP11—254K1 are the 10597582th power and 10752202, respectively, in the base banking IJ described in GenBank accession number NT—019273 ⁇ 18.
  • the Contig sequence consisting of these 4 clones is the base sequence from the 1097582nd position of IJ No. 10597582, described in GenBank accession number NT _019273-18 (about 0 ⁇ It is considered to correspond to an area of 5 Mbps.
  • the amplicon according to the present invention is a base sequence consisting of a base sequence from GenBank accession number NT-0119273.18, IJ No. 10597582, No. 11 to No. 121,868.
  • the force S is considered to be a chromosomal region substantially represented by the polynucleotide represented by.
  • the chromosomal region substantially represented by the polynucleotide represented by the nucleotide sequence consisting of nucleotide sequence 1059758 from the 2nd to 11121868 of the nucleotide sequence described in GenBank accession number NT-0192773.18 is TRIM33
  • BCAS2 breast carcino ma amplified sequence—2
  • DENND2C DENN / MADD dom ain containing 2C
  • the transcription region of the TRIM33 gene is GenBank accession number NT-019273.
  • the transcription region of the BCAS2 gene is substantially indicated by a polynucleotide represented by the nucleotide sequence up to the 11031950th position of the 11017863th nucleotide of the nucleotide sequence described in GenBank accession number NT-019273.18. It is thought to be conformed to the chromosomal region. Therefore, the transcription region of the BCAS2 gene is considered to be conformed to the chromosomal region substantially indicated by the polynucleotide represented by RP11-254K1.
  • the transcription region of the DENND2C gene is a polynucleotide represented by the nucleotide sequence from the 11033155th force of IJ's 11033155th to 11120417th described in GenBank accession number NT-0192 73.18. It is thought that it is actually conformed to the chromosomal region shown. Therefore, the transcription region of the DENND2C gene is considered to be coordinated with a chromosomal region substantially corresponding to the polynucleotide represented by RP11-254K1.
  • the present invention provides a diagnostic marker for small cell cancer or breast cancer, preferably lung small cell cancer or breast cancer, comprising the amplicon according to the present invention.
  • the disease site of a patient who develops small cell cancer such as small cell lung cancer or breast cancer is amplified by the amplicon according to the present invention as compared to the site of a healthy subject as a control. It is thought that there is. Therefore, when the amplicon according to the present invention is amplified in a biological sample derived from lung or milk tissue isolated from a certain person, compared to that isolated from a healthy person, the person is Diagnosed as having or having a high probability of suffering from small cell cancer or breast cancer, and / or having a poor or high prognosis.
  • Amplification of the amplicon according to the present invention means that the portion included in the amplicon is also amplified. Therefore, it is clear that the portion contained in the amplicon according to the present invention also serves as a diagnostic marker for small cell cancer such as small cell lung cancer or breast cancer.
  • a diagnostic marker for small cell cancer or breast cancer preferably small cell lung cancer or breast cancer comprising a portion contained in the amplicon according to the present invention.
  • the chromosome region where the TRIM33 gene is conformed Region chromosomal region where BCAS2 gene is conformed, chromosomal region where DENND2C gene is conformed, chromosomal region where TRIM33 gene transcription region is conformed, chromosomal region where BCAS2 gene transcription region is conformed, transcription region of DENND2C gene
  • examples include the chromosomal region to be conformed, TRIM33 gene, BCAS2 gene, and DENND2C gene.
  • the present inventors As a result of the expression analysis of the TRIM33 gene, the BCAS2 gene, and the DENND2C gene in 20 cell lines derived from human small cell lung cancer, the present inventors have found that the expression levels of the respective genes ( The amount of each gene transcript) was found to be increased compared to the expression level of each gene in healthy lungs (Fig. 1 (E)). Furthermore, the present inventor measured the amount of TRIM33 protein in cells using a cell line derived from small cell lung cancer (4 types) and a cell line derived from breast cancer (MCF7). It was found to be increased compared to the TRIM33 protein mass (Fig. 2 (B)).
  • the present inventors use an immunochemical technique to increase the expression of the TRIM33 gene only in the small cell carcinoma part in a mixed small cell lung cancer (mixed small cell cancer and linear cancer) tissue.
  • Figure 3 Furthermore, the present inventors have found that expression of the TRIM3 3 gene is increased in breast cancer tissue using an immunochemical technique (FIG. 3).
  • the prognosis of patients whose TRI M33 gene expression is strongly detected in breast cancer tissue is poor compared to the prognosis of patients whose expression of TRIM33 gene is not detected or weakly detected in breast cancer tissue.
  • FIG. 6 (A) (B) the prognosis of patients whose TRI M33 gene expression is strongly detected in breast cancer tissue.
  • the transcription product of the gene conforming to the amplicon according to the present invention and the protein encoded by the gene also serve as a diagnostic marker for small cell cancer such as small cell lung cancer or breast cancer. It is done. That is, when the expression of the gene is increased in a biological sample derived from lung or milk tissue isolated from a certain person, the person is suffering from small cell cancer such as small cell lung cancer or breast cancer. Diagnosed as having a strong or likely likelihood and / or having a poor or likely prognosis.
  • diagnosis of small cell cancer or breast cancer preferably small cell lung cancer or breast cancer comprising a transcription product of a gene conforming to the amplicon according to the present invention or a protein encoded by the gene, is preferable.
  • Markers are provided.
  • genes conforming to the amplicon according to the present invention include TRIM33 gene, BCAS2 gene, and DENND2C gene.
  • amplification of the amplicon according to the present invention in mammals including humans which is a diagnostic agent for small cell cancer or breast cancer, preferably small cell lung cancer or breast cancer, and A diagnostic agent capable of detecting an increased expression of a gene conforming to the amplicon is provided.
  • the presence of the above-mentioned diagnostic agent for diagnosis of human small cell cancer or breast cancer preferably human small cell cancer or breast cancer, small cell cancer or breast cancer, preferably small cell lung cancer or breast cancer is suspected.
  • the above diagnostic agent for prognosis of patients with small cell cancer or breast cancer preferably small cell lung cancer or breast cancer.
  • amplification of the amplicon according to the present invention and / or increased expression of a gene conforming to the amplicon is detected in a biological sample isolated from a human.
  • the above diagnostic agent comprising a nucleic acid probe that hybridizes with a region under stringent conditions; the base sequence described in GenBank Accession Number NT-0192713.18 and the base sequence consisting of up to 11 121868, or its complementary base
  • a polynucleotide comprising at least 15 consecutive nucleotides contained in the sequence or a condition that is stringent with the polynucleotide
  • the above diagnostic agent comprising a polynucleotide consisting of at least 15 bases that hybridizes underneath; a chromosomal region to
  • a method for diagnosing small cell cancer or breast cancer, preferably small cell lung cancer or breast cancer wherein the amplicon according to the present invention is used in a biological sample isolated from a human. And / or an increase in the expression of a gene conforming to the amplicon, and preferably a step of detecting an increase in the TRIM33 gene and / or an increase in the expression of the TRIM33 gene is provided.
  • a method for prognosing small cell cancer or breast cancer, preferably small cell lung cancer or breast cancer which comprises a step of detecting an increase in expression of the gene to be detected, preferably amplification of TRIM33 gene and / or detection of increased expression of TRIM33 gene .
  • the above diagnostic method characterized by using any one of the above diagnostic agents.
  • the present invention provides a method for screening a substance having a growth inhibitory action on small cell cancer cells or breast cancer cells, preferably lung small cell cancer cells or breast cancer cells, which is conformed to the amplicon according to the present invention.
  • a screening method comprising a step of selecting a substance having an action of suppressing amplification and / or expression of a gene, preferably TRIM33 gene, is provided.
  • a substance having an action of suppressing amplification and / or expression of a gene conforming to the amplicon according to the present invention preferably an action of suppressing amplification and / or expression of a TRIM33 gene.
  • a medicament for treating small cell cancer or breast cancer preferably small cell lung cancer or breast cancer, which contains, as an active ingredient, a substance having the above, for example, a substance screened by the screening method described above.
  • a double-stranded polynucleotide comprising a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 17 of the Sequence Listing and a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 18 of the Sequence Listing;
  • Examples include a double-stranded polynucleotide comprising a polynucleotide represented by the base sequence set forth in SEQ ID NO: 19 in the sequence listing and a polynucleotide represented by the base sequence set forth in SEQ ID NO: 20 in the sequence listing.
  • a method for treating small cell cancer or breast cancer preferably small cell cancer such as small cell lung cancer, more preferably small cell lung cancer, wherein a therapeutically effective amount of the above substance is applied to small cell lung cancer or the like.
  • a therapeutic method comprising the steps of administering to a small cell cancer patient or a breast cancer patient, preferably a small cell cancer patient such as a small cell lung cancer, more preferably a small cell lung cancer patient, and TRIM33 for the manufacture of the medicament
  • Use of a substance having an action of suppressing gene amplification and / or expression is provided by the present invention.
  • the present invention also provides a growth inhibitor for small cell carcinoma cells such as small cell lung cancer or breast cancer cells, comprising a double-stranded polynucleotide selected from the following group. .
  • a double-stranded polynucleotide comprising a polynucleotide represented by the base sequence set forth in SEQ ID NO: 17 of the sequence listing and a polynucleotide represented by the base sequence set forth in SEQ ID NO: 18 of the sequence listing
  • (2 ) A double-stranded polynucleotide comprising a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 19 in the Sequence Listing and a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 20 in the Sequence Listing.
  • FIG. 1 shows the TRM33 amplification ((A) to (D)) and expression level ((E)) in SCLC cell lines.
  • A A representative CGH array image of LU-140 cells is shown.
  • B shows the replication number profile of chromosome 1 in LU-140 cells.
  • C Representative FISH images of RP11-625J15 (TRIM33, green signal) and control RP11-315D10 (RET, red signal) hybridized to the metaphase chromosome of SCLC cell line LU-140.
  • D Amplicon map of 1 ⁇ 13.2 site in LU-140 cell line.
  • E shows TRIM33 expression in SCLC cell lines and normal lung as measured by RT-PCR analysis.
  • FIG. 2 shows TRM33 amplification and overexpression in breast cancer cell lines.
  • A BAC RP11-625J15 (TRIM33, green signal) and RP 11 _315D 10 (RET, red signal) were used. The results of a two-color FISH test on metaphase chromosomes derived from the breast cancer cell line MCF7 are shown.
  • B Immunoblotting of protein extracts in 4 SCLC, 2 NSCLC, and 1 breast cancer cell lines
  • FIG. 3 is a photograph showing the immunohistochemical staining of TRIM33 protein in lung primary small cell carcinoma and breast cancer. Shows strong nuclear staining (A-D) in small cell carcinoma and low expression in adenocarcinoma (D, tip of arrow) and low expression in normal lung (stars in A and C). Intensive nuclear staining in invasive ductal cell carcinoma (F), invasive lobular carcinoma (H, arrow tip), and intraepithelial ductal carcinoma (G), and low expression in normal mammary gland (H, arrow) Show.
  • A-D strong nuclear staining
  • F invasive ductal cell carcinoma
  • H, arrow tip invasive lobular carcinoma
  • G intraepithelial ductal carcinoma
  • H, arrow normal mammary gland
  • FIG. 4 shows the effect of enhanced TRIM33 expression on cancer cell proliferation.
  • A Western blotting results using protein extract and anti-Myc antibody are shown.
  • B Number of colonies formed by TRIM33 transform HeLa and SBC-3 cells.
  • C Growth-promoting action of TRIM33 on SBC-3 cells established as a clone that stably expresses TRI M33, and SBC-3 cells transfected with an empty vector (empty) as a control The results are shown.
  • FIG. 5 is a graph showing the growth inhibitory effect of siRNA targeting TRM33 mRNA.
  • A Shows the protein level of TRIM33 protein determined by Western blotting.
  • B shows the effect of siRNA oligonucleotides on the survival of SBC_5 and MCF7 cells.
  • C Control and TRM33_siRNA-treated The results of measuring the growth inhibitory action of TGF- / 3 on MCF7 cells by BrdU incorporation are shown.
  • FIG. 6 shows the correlation between the expression level of TRIM33 and the survival rate in 134 cases of breast cancer.
  • A The overall survival of the TRIM33 high group is significantly shorter than that of the TRIM33 low group.
  • B The progression-free survival of the TRIM33 high group is also significantly shorter than that of the TRIM33 low group.
  • the diagnostic marker provided by the present invention is a marker for cancer diagnosis detected in patients with small cell cancer such as small cell lung cancer or breast cancer, and the amplification of the amplicon according to the present invention and / or the present invention. Increased expression of genes conforming to such amplicons It is a marker for cancer diagnosis.
  • diagnosis marker refers to a biologically-derived substance used to determine the presence or absence of a certain disease, the degree thereof, or the prognosis after surgery, or the substance. Means the substance. Therefore, the marker for cancer diagnosis means a biological substance used for determining the presence or absence of cancer, its degree, or the prognosis after the operation, or a substance originating from the substance.
  • a marker for diagnosing cancer according to the present invention is preferably a marker for diagnosing small cell cancer such as small cell lung cancer or breast cancer.
  • the amplicon according to the present invention or a portion thereof, or a gene conforming to the amplicon, a transcription product of the gene, or the gene It is characterized by consisting of gene products.
  • the amplicon according to the present invention or a portion thereof is amplified and / or the expression of the gene is increased, the individual to be diagnosed has small cell cancer such as small cell lung cancer or breast cancer. Diagnosed as having or likely to be affected.
  • the prognosis of a patient with small cell cancer or breast cancer, such as small cell lung cancer, in which the amplicon according to the present invention or a portion thereof is amplified and / or the expression of the gene is increased is poor. It is diagnosed that the possibility is high.
  • genes that are conformed to the amplicon according to the present invention include TRIM33 gene, BC AS2 gene, and DENND2C gene.
  • the TRIM33 gene (sometimes simply referred to as “TRIM33”) is a gene involved in ectoderm formation in Xenopus embryogenesis and is inhibited by ubiquitination of the mesoderm-inducing factor Smad4 Responsible for the process. Information on this gene is available to those skilled in the art (Non-Patent Document l: Cell, 121, pp. 87-99, 2005). Since the TRIM33 gene is present in the chromosome of any mammalian animal and the TRI M33 protein encoded thereby is expressed in any mammalian animal, the cancer marker of the present invention is Forces available in mammals The most preferred subject is a human. For example, the full length of human-derived TRIM33 gene is exemplified in SEQ ID NO: 22 in the sequence listing.
  • the BCAS2 gene is a gene that conforms to the region of chromosome 1 ⁇ 13.3 to 21 and was found to be amplified in breast cancer (Cancer Lett., 185, pp.219-223, 2002). This Genetic information is available to those skilled in the art.
  • GenBank accession number NM-005872 discloses the full-length ORF base sequence of the BCAS2 gene.
  • DENND2C gene Information on the DENND2C gene is available to those skilled in the art.
  • the full-length ORF base sequence of the DENND2C gene is disclosed in GenBank accession number NM-198459.
  • the amplicon according to the present invention can be obtained from mammals including humans by a method known per se. Specifically, for example, the cell nucleus is separated and purified from mammals including humans, the genomic DNA contained in the cell nucleus is extracted, the genomic DNA is fragmented with a restriction enzyme, etc.
  • a genomic library is created by incorporating DNA into a vector such as BAC. From this genomic library, the probe for detecting the amplicon according to the present invention (for example, RP11-115211, RP11-1129E18, RP11-625J15, RP11-254K1, etc.) is used to simply amplify the amplicon according to the present invention. Can be separated.
  • the amplicon according to the present invention comprises a polynucleotide represented by a nucleotide sequence consisting of a base sequence of GenBank accession number NT-0192713.18, IJ No. 10597582, No. 11121868, and the like. Since it is considered to be a substantially chromosomal region, for example, the probe can be designed based on this base sequence.
  • a part or all of the amplicon according to the present invention can be used.
  • a part of the amplicon according to the present invention is used, two or more of the amplicons according to the present invention are used. May be used.
  • a chromosomal region in which the TRIM33 gene is conformed As part of the amplicon according to the present invention, a chromosomal region in which the TRIM33 gene is conformed, a chromosomal region in which the BCAS 2 gene is conformed, a chromosomal region in which the DENND2C gene is conformed, and a transcription region of the TRIM33 gene.
  • a chromosomal region to which the transcription region of the DENND2C gene conforms examples include a chromosomal region to which the transcription region of the DENND2C gene conforms, and a chromosomal region to which the transcription region of the BCAS2 gene conforms.
  • the chromosomal region in which a certain gene is conformed means a part of the genome in which the gene is located, and the genome including the sense strand of the gene and the corresponding antisense strand! / Means part.
  • a “gene” can be defined as a unit of genetic function defined by a base sequence of a certain region in high molecular DNA or RNA.
  • Eukaryotic genes can include exons and introns.
  • a gene can include a region on a nucleic acid having a specific control function such as a promoter or an operator.
  • a sense strand of a chromosomal region in which a gene is coordinated is also included in the gene by reverse transcription of the transcription product of the gene.
  • a single-stranded polynucleotide or a partial polynucleotide thereof forming the amplicon according to the present invention can be exemplified.
  • a cDNA library is prepared from an appropriate source in which the expression of the gene has been confirmed according to a conventional method, and the gene is obtained from the library. It can be obtained by selecting a desired clone using an appropriate probe or primer peculiar to the gene. Examples of gene origin include various cells and tissues in which the gene expression has been confirmed, or cultured cells derived from these cells (such as cell lines derived from small cell carcinoma such as breast cancer or small cell lung cancer). it can.
  • Isolation of total RNA from these sources, isolation and purification of mRNA, acquisition of cDNA and its cloning, etc. can all be performed according to conventional methods.
  • Selection of a desired clone from the cDNA library can be performed, for example, by confirming the expressed protein for each clone using a known protein expression system and further using the function of the protein as an index.
  • An example of the function of TRI M33 is the ubiquitination activity of Smad4.
  • the full-length ORF of the TRIM33 gene is disclosed in, for example, GenBank accession number NM-015906.3 or SEQ ID NO: 22 in the sequence listing. Based on these sequences, an appropriate probe or a specific probe specific to the TRIM33 gene Primers can be designed.
  • An example of such a primer is a primer consisting of a base sequence shown in SEQ ID NOs: 9 and 10 in the sequence listing.
  • the full-length ORF of the BCAS2 gene is disclosed in, for example, NM-005872.2, and it is possible to design appropriate probes and primers specific to the BCAS2 gene based on this sequence.
  • the ORF full length of the DENND2C gene is disclosed in, for example, NM 198459.2, and based on this sequence, DENND2C c It is possible to design appropriate probes and primers specific to the gene.
  • the desired gene can be obtained from various cells and tissues in which the expression of the gene has been confirmed by general means such as PCR using the above primers.
  • Reaction conditions for PCR are not particularly limited and can be selected by those skilled in the art. For example, 94 ° C for 30 seconds (denaturation), 55 ° C for 30 seconds, 1 minute (annealing), 72 ° C
  • a reaction step consisting of 2 minutes (elongation) is defined as one cycle. For example, after 30 cycles, the reaction can be performed at 72 ° C for 7 minutes.
  • the amplified DNA fragment can be cloned into an appropriate vector that can be amplified in a host such as Escherichia coli.
  • Techniques such as primer preparation and cloning of target genes are well known and commonly used by those skilled in the art. For example, Molecular Cloning 2nd Edition, Current Protocols in Molecular Biogy. Supplementl-38, John Wiley & Sons (1987—1997). ) Etc. can be performed according to the method described in S).
  • DNA consisting of a base sequence having a deletion, substitution and / or addition of one to several bases in the base sequence of the human TRIM33 gene is suitable for chemical synthesis, genetic engineering, or mutagenesis.
  • a genetic engineering technique can be used in addition to a method of contacting a drug that becomes a mutagen with the TRIM33 gene or a method of irradiating ultraviolet rays.
  • Site-specific mutagenesis which is one of the genetic engineering methods, is useful because it can introduce a specific mutation at a specific position, and follows the method described in Molecular Cloning, 2nd edition, above. It can be carried out.
  • the BCAS2 gene and DENND2C gene can also be obtained in the same manner as the TRIM33 gene.
  • a gene conforming to the amplicon according to the present invention a transcription product of the gene, or a gene product of the gene may be used.
  • the transcript of the gene conforming to the amplicon according to the present invention can be obtained in the same manner as the acquisition of the gene conforming to the amplicon according to the present invention.
  • a method for obtaining a gene product (eg, TRIM33 protein, BCAS2 protein, DENND2C protein, etc.) of a gene conforming to the amplicon according to the present invention is not particularly limited. Obtain the gene according to the method and apply this DNA
  • the gene product may be expressed by introducing it into an appropriate expression system, and the protein expressed by an appropriate method may be separated and purified.
  • the type of vector is not particularly limited.
  • the vector should replicate autonomously (for example, a plasmid), or be integrated into the genome of the host cell when introduced into the host cell and replicated together with the integrated chromosome. You can use a cluster.
  • an expression vector can be used, and it is desirable that elements necessary for transcription (eg, promoter, terminator, selection marker, etc.) are functionally linked.
  • a promoter is a DNA sequence that exhibits transcriptional activity in a host cell, and can be appropriately selected by those skilled in the art depending on the type of host.
  • a transformant can be prepared by introducing a recombinant vector containing the gene into an appropriate host (eg, bacteria, yeast, fungi, and higher eukaryotic cells). The gene product is cultured in an appropriate nutrient medium under conditions that allow gene expression, and the expressed gene product can be isolated and purified by an appropriate means available to those skilled in the art.
  • the diagnostic agent for small cell cancer such as small cell lung cancer or breast cancer provided by the present invention is amplifying and / or conforming to the amplicon according to the present invention in mammals including humans.
  • a diagnostic agent capable of detecting increased expression of a gene preferably a diagnostic agent for diagnosis of small cell cancer or breast cancer such as human small cell lung cancer, small cell cancer such as small cell lung cancer or Diagnostic agents for diagnosis of patients suspected of having breast cancer, amplification of amplicons according to the present invention and / or increased expression of genes conforming to the amplicons in biological samples isolated from humans It is a diagnostic agent that can detect.
  • Amplification of the amplicon according to the present invention means that the portion included in the amplicon is also amplified. Therefore, a diagnostic agent that can detect a portion contained in the amplicon according to the present invention is also included in the diagnostic agent according to the present invention.
  • amplification of the amplicon according to the present invention means that the number of copies of the amplicon according to the present invention increases.
  • the amplification of the amplicon according to the present invention is not limited to the amplification at its original locus (1 ⁇ 13.2), but also includes amplification in a homogeneously stained region or a double microchromosome.
  • the degree of amplification of the amplicon according to the present invention is increased as long as the amount of the amplicon contained in the test sample is increased compared with the amount of the amplicon in the healthy subject as a control! / , And les, not particularly limited.
  • the gene conformed to the amplicon according to the present invention means a gene included in the amplicon according to the present invention, for example, TRIM33 gene, BCAS2 gene, DENND2C gene Is exemplified.
  • TRIM33 gene a gene included in the amplicon according to the present invention
  • BCAS2 gene a gene included in the amplicon according to the present invention
  • DENND2C gene a gene included in the amplicon according to the present invention.
  • the diagnostic agent for small cell cancer such as small cell lung cancer or breast cancer provided by the present invention is suitably used for diagnosis of humans suspected of having small cell cancer such as small cell lung cancer or breast cancer.
  • the biological sample include collected blood, body fluid, sputum, milk, biopsy tissue, or tissue separated by surgery.
  • Examples of the diagnostic agent capable of detecting amplification of the amplicon according to the present invention include diagnostic agents capable of detecting amplification of a gene conforming to the amplicon according to the present invention.
  • diagnostic agents examples include diagnostic agents that can detect TRIM33 gene amplification, diagnostic agents that can detect BCAS2 gene amplification, and diagnostic agents that can detect DENND2C gene amplification. it can.
  • a diagnostic agent capable of detecting the amplification of the TRIM33 gene for example, a diagnostic agent containing a nucleic acid probe that hybridizes under stringent conditions with a chromosomal region in which the TRIM33 gene is conformed; Diagnostic agent containing a nucleic acid probe that hybridizes under stringent conditions with the chromosomal region to which the transcription region conforms; Diagnostic agent containing a nucleic acid probe that hybridizes under stringent conditions with the TRIM33 gene; GenBan kcession number NT— A nucleotide sequence consisting of at least 15 bases contained in the nucleotide sequence consisting of the 10843084th to 10961466th nucleotides of the IJ base described in 019273.18 or its complementary nucleotide sequence, or a stringent condition with the polynucleotide Polynucleoside consisting of at least 15 bases that hybridize below It can be exemplified diagnostic agent comprising a de
  • a diagnostic agent comprising a nucleic acid probe represented by any one of the base sequences described in SEQ ID NOs: 5 to 8 in the sequence listing, SEQ ID NO: 9 to 10 in the sequence listing
  • a diagnostic agent containing a primer consisting of one nucleotide sequence can be exemplified.
  • BCAS2 residue Diagnostic agent containing a nucleic acid probe that hybridizes under stringent conditions with the chromosomal region to which the gene is conformed; Nucleic acid probe that hybridizes under stringent conditions with the chromosomal region to which the transcription region of the BCAS2 gene is conformed Diagnostic agent; Diagnostic agent comprising a nucleic acid probe that hybridizes with the BCAS2 gene under stringent conditions; GenBank accession number NT-0119273.18 Examples thereof include a diagnostic agent comprising a polynucleotide comprising at least 15 consecutive bases contained in the complementary base sequence or a polynucleotide comprising at least 15 bases that hybridizes with the polynucleotide under stringent conditions.
  • a diagnostic agent containing a nucleic acid probe represented by any one of the nucleotide sequences set forth in SEQ ID NOs: 7 to 8 in the sequence listing, SEQ ID NOS: 11 to 12 in the sequence listing A diagnostic agent containing a primer consisting of one base sequence can be exemplified.
  • a diagnostic agent capable of detecting the amplification of the DENND2C gene for example, a diagnostic agent containing a nucleic acid probe that hybridizes under stringent conditions with a chromosomal region in which the DENN D2C gene conforms; a transcription region of the DENND2C gene; Diagnostic agent containing nucleic acid probe that hybridizes under stringent conditions with the chromosomal region to which it conforms; Diagnostic agent containing nucleic acid probe that hybridizes with DEN ND2C gene under stringent conditions; GenBank Accession Number NT— 019273.18 Or a polynucleotide comprising at least 15 contiguous bases contained in the base sequence consisting of the 11033155th to the 11120417th base sequence or the complementary base sequence thereof, or hybridizing with the polynucleotide under stringent conditions Polynuclear consisting of at least 15 bases It can be exemplified diagnostic agent comprising a plastid.
  • a diagnostic agent comprising a nucleic acid probe represented by any one of the nucleotide sequences set forth in SEQ ID NOs: 7 to 8 in the sequence listing, SEQ ID NOS: 13 to 14 in the sequence listing Examples thereof include a diagnostic agent containing a primer consisting of any one of the described base sequences.
  • examples of the diagnostic agent capable of detecting amplification of the amplicon according to the present invention include diagnostic agents including a nucleic acid probe that hybridizes with the amplicon according to the present invention under stringent conditions.
  • diagnostic agents include GenBank succession A base sequence consisting of at least 15 bases included in the base sequence consisting of up to the 10597582th power of IJ, No. 111 to 21868, or its complementary base sequence described in the number ⁇ _019273 ⁇ 18
  • a diagnostic agent containing a polynucleotide comprising at least 15 bases that hybridizes with a nucleotide under stringent conditions can be preferably exemplified. More specifically, a diagnostic agent containing a nucleic acid probe represented by any one of the base sequences described in SEQ ID NOs:! To 4 in the sequence listing can be preferably exemplified.
  • the length of the polynucleotide (including a nucleic acid probe or primer) contained in the diagnostic agent according to the present invention is preferably at least 15 bases (Molecular Cloning: A laboratory Manual, 2nd). Ed., Old Spring Harbor Laboratory, old Spring Harbor. N., 1989; sometimes referred to as Molecular Cloning 2nd edition).
  • the diagnostic marker according to the present invention is specifically detected by Southern hybridization or Northern hybridization, the length of the polynucleotide may be about 19 to 40 bp (Molecular Cloning No. 1). 2 edition).
  • the length of the polynucleotide can be preferably exemplified by about 300 to 3000 bp (the first clinical FISH protocol). Chromosome ⁇ Gene diagnosis method 1st edition, Joji Inazawa et al., 1997; sometimes referred to as clinical FISH protocol).
  • the detection of the amplification of the amplicon according to the present invention using the diagnostic agent capable of detecting the amplification of the amplicon according to the present invention includes, for example, conventional methods such as FISH method, Southern blot method, CGH array method, etc. Can be implemented.
  • CGH array method first, DNA from the test sample and normal tissue DNA as a control are labeled with FITC (green) and Texas red (red), respectively, by nick translation, and equal amounts of both labeled DNAs.
  • This solution is prepared by adding the solution to a slide glass labeled with the diagnostic agent according to the present invention after the preparation of the mixed solution and performing competitive hybridization and detecting fluorescence. The power to do S.
  • stringent conditions refers to, for example, 6 X SSC, 0.5% SDS and 50% honremamide in the night. This refers to the condition of washing at 68 ° C in a solution of 0.1 X SSC, 0.5% S DS after warming at C.
  • the polynucleotide to be hybridized may not be a polynucleotide having a base sequence complementary to the base sequence of the other polynucleotide to be hybridized. Hybridization can be performed, for example, according to the method described in the Molecular Cloning Second Edition or the Clinical FISH Protocol.
  • the "chromosomal region substantially represented by a certain polynucleotide” means a portion of a chromosome corresponding to a double-stranded polynucleotide formed from the polynucleotide and its complementary polynucleotide.
  • corresponding includes complete matching, and high homology (for example, 80% or more, preferably 90% or more, more preferably 93% or more, particularly preferably 95%). More preferably 98% or more).
  • a hybridized under stringent conditions with a chromosomal region in which the gene conformed to the amplicon according to the present invention is conformed is as follows. Examples of such genes include TRIM33 gene, BCAS2 gene, and DENND2C gene.
  • any one of SEQ ID NOS: 5 to 8 in the sequence listing, a nucleic acid probe represented by the base sequence set forth in 1, and a primer represented by the base sequence set forth in SEQ ID NO: 9 or 10 in the sequence listing Can be preferably exemplified.
  • a polynucleotide that hybridizes under stringent conditions with the chromosomal region to which the BCAS2 gene is conformed a polynucleotide that hybridizes under stringent conditions with the chromosomal region to which the transcription region of the BCAS 2 gene is conformed, and the BCAS2 gene and string.
  • the BCAS2 gene sequence the base sequence IJ described in SEQ ID NO: 7 in the sequence listing, the base sequence IJ described in SEQ ID NO: 8 in the sequence listing, the base sequence described in SEQ ID NO: 11 in the sequence listing,
  • the nucleotide sequence according to SEQ ID NO: 12 any force selected from the group consisting of: a polynucleotide having at least 15 bases contiguous in the nucleotide sequence of 1 or a complementary nucleotide sequence thereof, or under stringent conditions with the polynucleotide Examples of polynucleotides that hybridize with each other.
  • nucleic acid probe represented by the base sequence described in SEQ ID NO: 7 or 8 in the sequence listing, and a primer represented by the base sequence described in SEQ ID NO: 11 or 12 in the sequence listing may be preferably exemplified. it can.
  • a polynucleotide that hybridizes under stringent conditions with a chromosomal region in which the DENND2C gene is conformed and a polynucleotide that hybridizes under stringent conditions with a chromosomal region in which the transcription region of the DENND2C gene is conformed, DENND2C
  • a polynucleotide that hybridizes under stringent conditions with the gene included in the nucleotide sequence consisting of the 11033155th to 11120417th of the nucleotide sequence described in GenBank Session Number NT-019273 or its complementary nucleotide sequence Examples thereof include a polynucleotide comprising at least 15 consecutive nucleotides, or a polynucleotide that hybridizes with the polynucleotide under stringent conditions.
  • nucleic acid probe represented by the base sequence described in SEQ ID NO: 7 or 8 in the sequence listing, and a primer represented by the base sequence described in SEQ ID NO: 13 or 14 in the sequence listing are preferably exemplified. Can do.
  • a diagnostic agent for small cell cancer such as small cell lung cancer or breast cancer provided by the present invention
  • Drugs can be exemplified.
  • Such a diagnostic agent can detect increased expression of the gene conforming to the amplicon according to the present invention.
  • a diagnostic agent comprising a polynucleotide comprising at least 15 bases that hybridizes under stringent conditions with a transcription product of a gene conforming to the amplicon according to the present invention
  • a polynucleotide comprising at least 15 bases that hybridizes with a transcription product of TRIM33 gene under stringent conditions and a polynucleotide comprising at least 15 bases that hybridizes with a transcription product of BCAS2 gene under stringent conditions.
  • DENND2C gene transcript and string Or at least one of these forces can be exemplified.
  • the polynucleotide (including the nucleic acid probe or primer) contained in the diagnostic agent according to the present invention includes the base sequence of the amplicon according to the present invention, the base sequence of the gene conformed to the amplicon according to the present invention, etc. And can be produced by a conventional method, for example, a chemical synthesis method.
  • a diagnostic agent capable of detecting the gene product of the gene conforming to the amplicon according to the present invention can be exemplified.
  • the gene product of the gene conforming to the amplicon according to the present invention is recognized, preferably specifically recognized. Diagnostic agents including antibodies can be exemplified. Examples of the gene product of the gene conforming to the amplicon according to the present invention include TRIM33 protein, BCAS2 protein or DENND2C protein.
  • a diagnostic agent containing an antibody that recognizes the TRIM33 protein preferably a polypeptide represented by the amino acid sequence set forth in SEQ ID NO: 21 in the Sequence Listing can be exemplified.
  • the antibody herein may be either a polyclonal antibody or a monoclonal antibody, or may be a chimeric antibody in which antibodies derived from two or more species are combined.
  • An antibody can be produced using the gene product or a fragment thereof as an antigen.
  • the antigen is composed of at least 8, preferably at least 10, more preferably at least 12, even more preferably 15 or more amino acids.
  • a region represented by an amino acid sequence unique to the gene product is preferable to use as an antigen.
  • per se known antibody production methods can be used.
  • the antigen is administered to the animal by a method known per se to induce immunity, and the antibody is recovered from the serum of the immunized animal by a known antibody recovery method, or the antibody-producing cells are recovered and known per se Can be produced by introducing means for transformation into permanent proliferating cells.
  • the polynucleotide (including the case of being a nucleic acid probe or primer) or antibody contained in the diagnostic agent according to the present invention is the amplification of the amplicon according to the present invention and / or the expression of the gene conforming to the amplicon.
  • the renucleotide, the nucleic acid probe, the primer or the antibody can be labeled with an appropriate label such as a fluorescent label or a radioisotope.
  • an appropriate label such as a fluorescent label or a radioisotope.
  • labeling means are well known and commonly used by those skilled in the art.
  • the method for diagnosing small cell cancer such as small cell lung cancer or breast cancer comprises amplifying the amplicon according to the present invention and / or the amplicon in a biological sample isolated from human force. It includes a step of detecting increased expression of a gene conforming to.
  • the method includes a step of detecting amplification of the amplicon according to the present invention using the polynucleotide, nucleic acid probe, or primer described above, or conforms to the amplicon using the antibody described above.
  • a step of detecting increased expression of the gene It is also preferable to detect the amplification of the amplicon according to the present invention and the increased expression of the gene conforming to the amplicon by combining the above two steps. More specifically, the above diagnostic method including the step of detecting amplification of TRIM33 gene and / or enhanced expression of TRIM33 gene can be exemplified.
  • the amplification of the amplicon according to the present invention and / or amplification of the amplicon according to the present invention is performed on a biological sample isolated from a human. It is possible to detect an increase in the expression of a gene conforming to the.
  • the diagnostic method including the step of detecting the amplification of TRIM33 gene and / or the increased expression of TRIM33 gene, typically, (1) the amount of TRIM33 gene and / or TRIM33 contained in a biological sample isolated from human A step of measuring the expression level of the gene product of the gene, and (2) the amount of the TRIM33 gene and / or the expression level of the gene product of the TRIM33 gene measured in (1) above and included in a healthy human-derived biological sample It is preferable to include a step of comparing the amount of the TRIM33 gene and / or the expression level of the gene product of the TRIM33 gene. Prepare in advance the amount of TRIM33 gene and / or the expression level of the gene product of TRIM33 gene contained in a healthy human-derived biological sample as a standard value, and perform the above step (2) using this standard value. You can also.
  • the screening method provided by the present invention is a screening method for a substance having a growth inhibitory action on small cell cancer cells such as small cell lung cancer or breast cancer cells, and is conformed to the amplicon according to the present invention. Suppresses the amplification and / or expression of genes It includes the step of selecting a substance to have!
  • the substance selected by the above method can be used as an active ingredient of a medicament for the treatment of small cell cancer such as small cell lung cancer or breast cancer, and the substance selected by the above method can be used as a pulmonary substance. It can be used as a growth inhibitor for small cell cancer cells such as small cell cancer or breast cancer cells.
  • the types of substances subject to screening are not particularly limited, and include, for example, organic low molecular weight compounds, high molecular weight compounds, inorganic compounds, nucleic acids (including small molecule RNA and non-natural bases used for RNAi, etc. Any substance is included, including nucleic acids and the like, sugars, lipids, or peptides (including oligopeptides or polypeptides).
  • a screening method including the step of detecting an inhibitory effect on the amplification and / or expression of the TRIM33 gene by a test substance, typically, (1) a small cell, such as small cell lung cancer, TRIM33 in a cancer cell or a breast cancer cell It is preferable to include a step of measuring the number of gene copies and / or the expression level of the gene product of the TRIM33 gene in the presence or absence of the test substance.
  • Small cell carcinoma cells such as small cell lung cancer or breast cancer cells are isolated from humans including patients.
  • Small cell cancer cells or breast cancer cells may be used, but for example, small cell cancer cells or breast cancer cells such as small cell lung cancer that stably express the TRIM33 protein may be prepared and used as established cell lines. Good.
  • a small cell carcinoma cell line for example, LU-140
  • MCF7 breast cancer cell line
  • the number of copies of the TRIM33 gene can be measured by a conventional method using the above-described nucleic acid probe or primer, for example, the Northern blot method or the CGH method.
  • the expression level of the gene product of TRIM33 gene can be detected, for example, by Western plotting.
  • the medicament provided by the present invention is a medicament for the treatment of small cell cancer such as small cell lung cancer or breast cancer, and amplification and / or amplification of a gene conforming to the amplicon according to the present invention. It contains a substance having an action of suppressing the expression of the gene as an active ingredient.
  • the medicament include a substance having an action of amplifying the TRIM33 gene and / or suppressing the expression of the gene as an active ingredient. Or, for example, on It may be a medicine containing a substance screened by the screening method described above as an active ingredient.
  • the medicament of the present invention can be administered to mammals including humans.
  • the administration form of the medicament of the present invention is not particularly limited and can be administered orally or parenterally.
  • the medicament of the present invention may be a substance that is an active ingredient, but a pharmaceutical composition containing a substance that is an active ingredient and a pharmacologically and pharmaceutically acceptable additive for pharmaceutical preparation is prepared and administered. I hope to do it.
  • the medicament of the present invention containing a protein as an active ingredient can be formulated according to a conventional method for preparing a protein preparation, and the medicament of the present invention containing a polynucleotide as an active ingredient can also be used in the art. It can be formulated and used by various means.
  • the medicament of the present invention containing a nucleic acid can also be used, for example, in the form of a recombinant vector containing a nucleic acid which is an active ingredient.
  • the above recombinant vector contains various sequences necessary for gene expression or promoting gene expression so that the gene product is efficiently expressed in vivo from the nucleic acid which is an active ingredient. Incorporate them in the proper order!
  • the total length is not particularly limited.
  • the antisense polynucleotide may be, for example, an oligonucleotide having 10 bases or more, preferably 15 bases or more, so that complementary binding is possible.
  • an antibody preferably a monoclonal antibody
  • the antibody can be produced by a usual method, and is preferably a gene product of a gene conforming to the amplicon according to the present invention, preferably Monoclonal antibodies capable of specifically binding to the TRIM33 protein can also be produced by a general method of those skilled in the art.
  • a cell growth inhibitor for small cell carcinoma cells such as small cell lung cancer or breast cancer cells
  • a preferred polynucleotide as an active ingredient of the medicament of the present invention for example, the following (1) or (2) duplex Polynucleotide: (1) a double-stranded polynucleotide comprising a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 17 of the Sequence Listing and a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 18 of the Sequence Listing; And (2) a double-stranded polynucleotide comprising a polynucleotide represented by the base sequence set forth in SEQ ID NO: 19 of the sequence listing and a polynucleotide represented by the base sequence set forth in SEQ ID NO: 20 of the sequence listing
  • the force S that can be, is not limited to these.
  • Examples of pharmaceutical additives that are pharmacologically and pharmaceutically acceptable include excipients, disintegrating agents or disintegrating aids, binders, lubricants, coating agents, dyes, diluents, The ability to use bases, solubilizers or solubilizers, isotonic agents, pH adjusters, stabilizers, propellants, adhesives, etc.
  • Examples of pharmaceutical compositions suitable for oral administration include, for example, tablets, capsules, powders, fine granules, granules, liquids, syrups and the like.
  • compositions suitable for parenteral administration include injections, drops, suppositories, inhalants, transdermal absorption agents, eye drops, ear drops, ointments, creams, or patches. be able to.
  • the dosage of the medicament of the present invention is not particularly limited, and an appropriate dosage may be selected according to various conditions such as the type of substance as an active ingredient, the purpose of treatment or prevention, the age and symptoms of the patient, the route of administration and the like.
  • the power that can be selected In general, an adult can be selected from a dose range of about 0.001 mg per day; about 100 Omg.
  • an appropriate medium RPMI-1640 or Dulbecco with 10% urine fetal serum and 100 units / ml penicillin / 100 g / ml streptomycin. Maintained in modified Eagle medium).
  • a CGH array (MCG Cancer Array -800) was prepared using 800 BAC / PAC clones with genes and STS (sequence_tagged site) markers that may be important for cancer development and progression (Cancer Sci., 95 , Pp.559-563, 2004), and hybridization was performed according to the method described in the literature (Cancer Sci., 96, pp.100-110, 2005; Cancer Sci., 96, pp. 676-683, 2005). Specifically, the test DNA from each GBM cell line and the control DNA obtained from healthy male volunteers were labeled with Cy3_dCTP or Cy5_dCTP, respectively, precipitated with ethanol in the presence of Cot-1 DNA, and then the hybridization mixture.
  • Metaphase chromosomes were prepared from each cell line. FISH analysis was performed by using the BAC located near the region to be analyzed as a probe by the method reported previously (Clin. Cancer Res., 15, pp.4705-4713, 2003).
  • Single-stranded cDNA was generated using total RNA strength, Superscript First-Strand Synthesis System (Invitrogen), and amplified using specific primers for each gene (TRM33 gene, BCAS2 gene, or FLJ37099 gene).
  • TRM33 gene Superscript First-Strand Synthesis System
  • BCAS2 gene BCAS2 gene
  • FLJ37099 gene specific primers for each gene
  • GPDH glyceraldehyde-3-phosphate dehydrogenase gene
  • pCMV-Tag3_TRM33 The plasmid that expresses Myc-tagged TRIM33 (pCMV-Tag3_TRM33) is cloned in frame into the eukaryotic expression vector pCMV-Tag3 (Stratagene) together with the above RT-PCR product, which is the entire coding region of TRIM33.
  • pCMV-Tag3_TRIM33 or empty vector pCMV-Tag3_mock
  • FuGENE6 Roche
  • the expression of TRIM33 protein in transiently transfected cells was detected 48 hours after transfection. This was confirmed by Western blotting analysis using Signaling Technology). Cells were incubated for 2 weeks in 6-well plates with appropriate concentrations of G418, fixed with 70% ethanol and stained with crystal violet.
  • Stable TRIM33 transfectants and controls were obtained by transfecting pCMV-Tag3_TRM33 or pCMV-Tag3_mock, respectively, into cells with low expression of TRIM33. 2 ⁇ 10 3 cells were seeded in 96-well plates for measurement of cell proliferation. The number of viable cells was measured by colorimetric quantitative analysis (cell counting kit_8, Dojindo Laboratories) using a water-soluble tetrazolium salt.
  • tissue microarray (TMA) blocks of tissue core samples taken from the center of each tumor were prepared.
  • TMA tissue microarray
  • tissue cores were collected from each representative tissue piece, and these cores were placed on the receiving block using a Tissue Microarrayer (Beecher Instruments).
  • TMAs containing 138 core samples were constructed using a 2.0 mm diameter core, placed 7 to 0.8 mm apart in the receiving block. The paraffin in the sample was removed with various concentrations of ethanol and rehydrated.
  • a tumor was TRM33 negative when all malignant cells were not nuclear-stained, and a tumor was TRM33-positive when nuclear staining was observed in more than 50% of the tumor cell cells.
  • Small lymphocytes (not showing pl6 nuclear staining) were used as a negative internal control and the following criteria were applied.
  • SiRNA treatment was performed according to the previously reported method (Cell, 121, pp. 87-99, 2005). Using Lipofectamine 2000 protocol (Invitrogen), double-stranded RNA oligomers (Sigma genosys, 75 ng m 2 ) were transfected into each cell. As a control-siRNA, an unrelated sequence of RNA was used.
  • the dmin pattern was not shown in the strain, indicating that it was located away from the 1 pl3 amplicon (FIG. 1D).
  • the affected sites that were the least common among the 13 amplicons in BAC RP 11-115211 and 1 ⁇ 11-25411 were identified, that is, the sites having the TRIM33, BCAS2, and FLJ37099 genes.
  • the amplicon size was only 500kb.
  • Figure 1 shows the amplification ((A) to (D)) and expression level ((E) of TRIM33 in SCLC cell lines.
  • A Representative CGH array of LU-140 cells Image shows a marked increase in TRIM33 replication as a clear green signal (arrow)
  • B shows the replication profile of chromosome 1 in LU-140 cells.
  • C RP11-625J15 (TRIM33, green signal) hybridized to metaphase chromosome of SCLC cell line LU-140 and control RP11-3
  • a typical FISH image of 15D10 shows that TRIM33 (green) is amplified on a double microchromosome BAC RP11-315D10 shows two signals in the same cell line
  • D Amplicon map of 1 ⁇ 13 ⁇ 2 site in LU-140 cell line 7 types used as probes in FIS sputum test Shows the position of BAC (vertical bar) and three genes (TRIM33, BCAS2, and FLJ37099), the number of replicates per cell and LU -Range of dmin determined by FISH test of metaphase chromosomes derived from -140 cells.
  • TRIM33, BCAS2, and FLJ37099 are individual targets for amplification. These genes were detected in all SCLC cell lines except Lu-141 and S-1 by semiquantitative RT-PCR. The expression level of was examined. As shown in Figure 1E, TRIM33, BCAS2, and FL J37099 expression levels are normal in 17 (85%), 4 (20%), and 4 (20%) of 20 cell lines, respectively. Of these three genes, TR IM33 was most frequently overexpressed compared to lung cDNA.
  • Figure 1 (E) shows TRIM33 expression in SCLC cell lines and normal lung as measured by RT-PCR analysis.
  • ACC-LC_80 1: ACC-LC_80, 2: ACC_LC_173, 3 ⁇ u_130, 4: Lul34A, 5: Lul34B, 6: Lu_143, 7: 87-5, 8: S_2, 9: ACC_LC_5, 10: ACC_LC_172, 11: ACC_LC_170, 12 : PC_6, 13: SBC_5, 14: Lu_24, 15: Lu_135, 16: Lu_13 8, 17: Lu-139, 18: Lu-165, 19: SBC-3, 20: Lu-140, 21: Normal lung. Overexpression of TRIM33 was observed in 17 out of 20 cell lines (85.0%).
  • MCF7 breast cancer cell line (Cancer Genet. Cytogenet., 117, pp.153-158, 2000) that gave high level expression in this region by CGH reported previously was subjected to FIS H test, and TRIM33 replication in MCF7 I checked the number.
  • TRIM33 protein Expression levels of TRIM33 protein in four SCLC cell lines (LU-24, LU_139, LU_165, and LU_140), two NSCLC cell lines (PC-10 and A549), and one breast cancer cell line (MCF7) Compared. As shown in FIG. 2B, the maximum amount of TRIM33 was observed in LU-140 and MCF7, and the minimum amount in the NSCLC cell line. The expression level of TRIM33 protein was consistent with the amplification level of this gene.
  • Fig. 2 shows the amplification and overexpression of TRIM33 in breast cancer cell lines.
  • A shows the results of a two-color FISH test for metaphase chromosomes derived from breast cancer cell line MCF7 using BA C RP11-625J15 (TRIM33, green signal) and RP11_315D10 (RET, red signal).
  • B AC RP11-625J15 gave a strong signal as HSR in MCF7.
  • BAC RP11-31 5D10 gave two signals in the same cell line.
  • B shows immunoblotting of protein extracts in 4 types of SCLC, 2 types of NSCLC, and 1 type of breast cancer cell line.
  • TRIM33 protein coincided with the amplification level of the gene in LU-140 and MCF7.
  • FIG. 3 is a photograph showing immunohistochemical staining of TRM33 protein in primary small cell lung cancer and breast cancer.
  • FIG. 4 shows the effect of enhanced TRIM33 expression on cancer cell proliferation.
  • a construct containing the full-length sequence of TRIM33 and a Myc tag or an empty vector (pCMV-Tag3_mock, controller) was transfected into HeLa and SCLC cell line SBC-3.
  • A The results of Western blotting using 50 g of protein extract and anti-Myc antibody are shown. This indicates that Myc-tagged TRIM33 protein is expressed in the cells transiently tranfected with pCMV-Tag3_TRIM33!
  • the upper row shows the results of Western blot analysis of 2 clones (clone 1 and 2) transfected with pCMV_Tag3B-TRIM33 using 50 ⁇ g of protein extract and anti-Myc-tag antibody. Both clones expressed Myc-tagged TRM33 protein!
  • the lower graph shows the effect of stable TRIM3 3 expression on the proliferation of SBC-3 cells. Cell viability was measured on a given day with water-soluble tetrazolium salt assembly (each point on the graph is the average of three independent test results and the bar represents SE).
  • TRM33 suppresses the anti-proliferative effect of TGF- ⁇ by reducing the Smad response
  • control and TRIM33_siRNA-treated MCF7 cell DNA synthesis / cell cycle progression were assessed by BrdU incorporation.
  • Inhibition of TRIM33 inhibited the basal rate of BrdU incorporation and markedly enhanced the growth-suppressing action induced by exogenous TGF-3 (Fig. 5C). This result indicates that TRM33 exerts a growth promoting effect on SCLC and breast cancer cells by attenuating the cell growth inhibitory effect of TGF- / 3 by TRM33.
  • Fig. 5 shows the growth inhibitory action of siRNA targeting TRIM33 mRNA.
  • MCF7 cells showing amplification and overexpression of TRIM33 and SBC-5 cells showing overexpression of TRIM33 were treated with siRNA targeting TRIM33 or control siRNA using Lipofectamine 2000.
  • A Protein level of TRIM33 protein determined by Western blotting. Cells were treated with 75 ng m 2 oligonucleotide (TRIM33 or controller) or Lipofectamine 2000 alone, and cells were collected 48 and 120 hours after transfer. Untreated cells were maintained under the same conditions. 50 g of whole cell lysate was separated by polyacrylamide gel electrophoresis and analyzed by immunoblotting.
  • TRM33 is a prognostic factor for breast cancer
  • a prognostic factor analysis was performed on 134 cases with a clear prognosis among the 138 breast cancer cases stained in (g) above.
  • the TRIM33 high group is significant (p value: 0.0285 or 0 ⁇ 011) for both overall survival and progression-free survival. ) Had a poor prognosis. From these results, TRIM33 was considered useful as a prognostic marker.
  • nucleotide sequence set forth in SEQ ID NO: 2 RP11-115211 probe, -strand
  • nucleotide sequence set forth in SEQ ID NO: 4 RP11-1129E18 probe, -strand Base sequence described in SEQ ID NO: 5: RP11-625J15 probe, + strand, NT— 019273 (10389 3839, 120498679): 10785181-10949822 (Length: 164642) Start: 114679019-114 843660 (Length: 164642)
  • nucleotide sequence set forth in SEQ ID NO: 8 RP11-254KI probe, -strand
  • SEQ ID NO: 9 nucleotide sequence: F primer used for TRM33 gene expression analysis
  • SEQ ID NO: 10 nucleotide sequence: TRM33 gene expression analysis
  • R primer SEQ ID NO: 11 nucleotide sequence: BCAS2 gene
  • F primer used for expression analysis Base sequence described in SEQ ID NO: 12: used for expression analysis of BCAS2 gene
  • Base sequence described in SEQ ID NO: 13 F primer used for expression analysis of DENND2C gene
  • Base sequence described in 14 R primer used for expression analysis of DENND2C gene
  • Base sequence described in SEQ ID NO: 17 TRM33 siRNA (l) sense strand
  • Amino acid sequence set forth in SEQ ID NO: 21 antigen of TRIM33 antibody
  • Nucleotide sequence described in SEQ ID NO: 25 Nucleotide sequence consisting of Nos. 10597582 to 11121868 of the nucleotide sequence described in GenBank accession number NT-0192713.18 Industrial applicability
  • the present invention provides a means for easily and reliably diagnosing small cell cancer such as small cell lung cancer and breast cancer. According to the present invention, a medicament for the treatment of small cell cancer such as small cell lung cancer and breast cancer is provided.

Abstract

Disclosed is a marker for use in the diagnosis of small cell carcinoma or breast cancer, which comprises a chromosomal region, a gene or a protein selected from the group consisting of the following members: (1) a chromosomal region substantially comprising a polynucleotide having a nucleotide sequence lying between position-10597582 and position-11121868 in the nucleotide sequence depicted in GenBank Accession Number NT_019273.18; (2) TRIM33 (tripartite motif 33) gene; (3) BCAS2 (breast carcinoma amplified sequence-2) gene; (4) DENND2C (DENN/MADD domain containing 2C) gene; (5) TRIM33 protein; (6) BCAS2 protein; and (7) DENND2C protein.

Description

明 細 書  Specification
癌診断用マーカーならびに治療の標的分子  Markers for cancer diagnosis and target molecules for treatment
技術分野  Technical field
[0001] 本発明は、肺小細胞癌などの小細胞癌又は乳癌の診断を行うための手段、好まし くは特定の遺伝子の増幅又は発現亢進を癌診断用マーカーとして利用する手段に 関する。  The present invention relates to a means for diagnosing small cell cancer such as small cell lung cancer or breast cancer, and preferably means for utilizing amplification or expression enhancement of a specific gene as a cancer diagnostic marker.
また、本発明は上記の特定の遺伝子の増幅又は発現亢進を抑制することにより肺 小細胞癌などの小細胞癌又は乳癌を治療する手段に関する。  The present invention also relates to a means for treating small cell cancer such as small cell lung cancer or breast cancer by suppressing amplification or expression enhancement of the specific gene.
背景技術  Background art
[0002] TRIM33 (tripartite motif 33、「Ectodermin」と呼ばれる場合もある)はアフリカッメガ エルの胚形成において外胚葉形成に関与する因子として同定され、中胚葉誘導因 子である Smad4をュビキチン化することにより阻害することが知られている (非特許文 献 1 : Cell, 121, pp.87- 99, 2005)。 TRIM33遺伝子とがんとの関係については、 TRIM3 3遺伝子が大腸癌にお!/、て発現亢進して!/、ること力 S示されて!/、る(非特許文献 1、 Fig. 7D及び 7E)。また、メラノーマ C32細胞株又は乳癌 MB231細胞株内の TRIM33遺伝子 の発現を RNA干渉法で抑制するとこれらの細胞の増殖が抑制されることが示されて おり (同 Fig.7F及び 7G)、その細胞増殖抑制は Smad4依存的であることが示されている (同 Fig.7G)。  [0002] TRIM33 (also called tripartite motif 33, sometimes called “Ectodermin”) has been identified as a factor involved in ectoderm formation in Xenopus embryogenesis, and by ubiquitinating Smad4, a mesoderm-inducing factor It is known to inhibit (Non-Patent Document 1: Cell, 121, pp.87-99, 2005). Regarding the relationship between TRIM33 gene and cancer, TRIM3 3 gene has been shown to increase expression in colorectal cancer! /, And its ability to be demonstrated S /! (Non-patent document 1, Fig. 7D And 7E). In addition, suppression of TRIM33 gene expression in the melanoma C32 cell line or breast cancer MB231 cell line by RNA interference has been shown to suppress the growth of these cells (Figs. 7F and 7G). Proliferation inhibition has been shown to be Smad4-dependent (Fig. 7G).
[0003] しかしながら、上記刊行物には、乳癌細胞内において TRIM33遺伝子の増幅又は 発現亢進が生じていることについての報告はなぐ TRM33遺伝子とその他の癌(例 えば肺小細胞癌など)との関係についても報告がない。また、 TRM33遺伝子は遺伝 子座 1ρ13.1に存在する遺伝子であり、乳癌においてヒトゲノム 1ρ13領域が増幅してい るとの報告もある力 s (非特許文献 2 : Cancer Research, 60, pp.4519-4525, 2000;非特 許文献 3 : Cancer Genet Cytogenet, 117, pp.153-158, 2000)、これらの刊行物には乳 癌にぉレ、て特定の TRIM33遺伝子が増幅又は発現亢進して!/、ることは示唆な!/、し教 示されていない。 [0003] However, the above publications do not report that the TRIM33 gene is amplified or increased in breast cancer cells. Relationship between TRM33 gene and other cancers (eg small cell lung cancer) There is no report about. Further, TRM33 gene is a gene present in the genetic child seat 1Ro13.1, the force is also reported that the human genome 1ρ13 area you are amplified in breast cancer s (Non-Patent Document 2: Cancer Research, 60, pp.4519- 4525, 2000; Non-Patent Document 3: Cancer Genet Cytogenet, 117, pp.153-158, 2000), these publications show that specific TRIM33 genes are amplified or upregulated in breast cancer! / No suggestion! /, And no teaching.
[0004] また、癌における遺伝子発現パターンは、発生母地の組織における遺伝子発現パ ターンを受け継いでいることが多ぐ組織ごとに遺伝子の発現パターンは相違する。 従って、癌種を越えて共通して変化が認められる遺伝子が存在する一方で、一般的 には、癌における遺伝子発現パターンは、発生母地の組織 (例えば、大腸癌や肺癌 といった分類)により異なると考えられる(Molecular biology of the cell (第 3版) .1256[0004] In addition, the gene expression pattern in cancer is the gene expression pattern in the tissue of the origin. The gene expression pattern differs for each tissue that often inherits the turn. Therefore, while there are genes that change in common across cancer types, in general, gene expression patterns in cancer differ depending on the tissue of origin (for example, classification such as colon cancer and lung cancer). (Molecular biology of the cell (3rd edition) .1256
-1257等)。かかる観点からも、非特許文献 1は、 TRIM33遺伝子とその他の癌(例え ば、肺小細胞癌など)の関係につ!/、て示唆なレ、し教示されて!/、な!/、と考える。 -1257). From this point of view, Non-Patent Document 1 suggests the relationship between TRIM33 gene and other cancers (for example, small cell lung cancer)! / I think.
非特許文献 1 : Cell, 121, pp.87- 99, 2005  Non-Patent Document 1: Cell, 121, pp.87-99, 2005
非特許文献 2: Cancer Research, 60, pp.4519-4525, 2000  Non-patent document 2: Cancer Research, 60, pp.4519-4525, 2000
非特許文献 3: Cancer Genet Cytogenet, 117, pp.153-158, 2000  Non-patent document 3: Cancer Genet Cytogenet, 117, pp.153-158, 2000
発明の開示  Disclosure of the invention
発明が解決しょうとする課題  Problems to be solved by the invention
[0005] 本発明の課題は、特定の遺伝子の増幅又は発現亢進を癌診断用マーカーとして 利用する手段、好ましくは肺小細胞癌などの小細胞癌又は乳癌の診断を行う手段を 提供することにある。 [0005] An object of the present invention is to provide means for using amplification or increased expression of a specific gene as a marker for cancer diagnosis, preferably means for diagnosing small cell cancer such as small cell lung cancer or breast cancer. is there.
また、本発明の別の課題は、上記の特定の遺伝子の増幅又は発現亢進を抑制す ることにより肺小細胞癌などの小細胞癌又は乳癌を治療する手段を提供することにあ 課題を解決するための手段  Another object of the present invention is to provide a means for treating small cell cancer such as small cell lung cancer or breast cancer by suppressing the amplification or increased expression of the specific gene. Means to do
[0006] 本発明者らは上記の課題を解決すべく鋭意研究を行った結果、肺小細胞癌などの 小細胞癌及び乳癌細胞において TRIM33遺伝子が増幅しており、かつその発現が 亢進していることを見出した。さらに、 TRIM33遺伝子の発現レベルが乳癌の予後因 子であることを見出した。また、該遺伝子を肺小細胞癌などの小細胞癌及び乳癌の マーカーとして用いることができ、その増幅又は発現亢進を例えば該遺伝子に特異 的に結合可能なプローブなどを用いて検出することにより、肺小細胞癌などの小細 胞癌及び乳癌を簡便かつ確実に診断できることを見出した。さらに、 TRIM33遺伝 子の発現を例えば RNA干渉法などの手段により阻害することによって肺小細胞癌な どの小細胞癌及び乳癌細胞の増殖を顕著に抑制することができ、それによつて肺小 細胞癌などの小細胞癌及び乳癌を治療できることを見出した。本発明は上記知見に 基づレ、て完成されたものである。 [0006] As a result of intensive studies to solve the above problems, the present inventors have found that TRIM33 gene is amplified in small cell carcinoma such as small cell lung cancer and breast cancer cells, and its expression is enhanced. I found out. Furthermore, we found that the expression level of TRIM33 gene is a prognostic factor for breast cancer. Further, the gene can be used as a marker for small cell carcinoma such as small cell lung cancer and breast cancer, and by detecting the amplification or expression enhancement using, for example, a probe that can specifically bind to the gene, It was found that small cell carcinoma such as small cell lung cancer and breast cancer can be diagnosed easily and reliably. Furthermore, by inhibiting the expression of the TRIM33 gene by means such as RNA interference, it is possible to remarkably suppress the growth of small cell carcinomas such as small cell lung cancer and breast cancer cells, thereby producing small cell lung cancer. It has been found that it can treat small cell cancer and breast cancer. The present invention is based on the above findings. Based on this, it was completed.
[0007] 本発明者らは、ヒト肺小細胞癌由来の細胞株(LU— 140)において 1ρ13. 2上の 特定の領域 (以下、本発明に係るアンプリコンと称することもある)が二重微染色体(d ouble minute chromosome ; dmin)で増幅していることを見出した(図 1 (C) , (D ) )。さらに、本発明者らは、ヒト乳癌由来の細胞株(MCF7)において TRIM33遺伝 子が均一染色体領域(homogenously staining region ; HSR)で増幅しているこ とを見出した(図 2、(A) )。  [0007] The present inventors have doubled a specific region on 1ρ13.2 (hereinafter also referred to as an amplicon according to the present invention) in a cell line derived from human small cell lung cancer (LU-140). It was found that amplification was performed on a minute chromosome (dmin) (Fig. 1 (C), (D)). Furthermore, the present inventors found that the TRIM33 gene was amplified in a homogeneous chromosome region (HSR) in a human breast cancer-derived cell line (MCF7) (Fig. 2, (A)). .
[0008] 本発明に係るアンプリコンは、その本来の座位として、 4種類の BACクローン(RP1 1— 115211、 RP11 - 1129E18, RP11— 625J15、 RP11— 254K1)で実質的に 示される 1ρ13· 2の染色体領域に存在する。これらの BACクローンは、ヒト血液由来 の染色体から作成されたヒト染色体断片の BACライブラリー(RPCI human BAC li brary 11)のクローンである。 BACライブラリーの各クローンは、該染色体のある断片 (+鎖、 鎖)を示す。各クローン(+鎖、 鎖)の末端部分が配列解析され、その塩 基配列が明ら力、となっている。なお、 BACライブラリーの各クローン(+鎖、一鎖)の 全長塩基配列は明らかにされて!/、な!/、。  [0008] The amplicon according to the present invention has, as its original locus, 1ρ13 · 2 substantially represented by four types of BAC clones (RP1 1-1115211, RP11-1129E18, RP11-625J15, RP11-254K1). Present in the chromosomal region. These BAC clones are clones of the BAC library (RPCI human BAC library 11) of human chromosome fragments created from human blood-derived chromosomes. Each clone in the BAC library represents a certain fragment (+ strand, strand) of the chromosome. The terminal portion of each clone (+ strand, strand) was sequenced, and its base sequence became clear. In addition, the full-length nucleotide sequence of each clone (+ strand, single strand) of the BAC library has been revealed!
[0009] RP11 - 1152I1 (+鎖)の末端の部分塩基配列は、配列番号 1に記載の塩基配列  [0009] The partial base sequence of the end of RP11-1152I1 (+ strand) is the base sequence described in SEQ ID NO: 1.
(GenBankァクセッション番号: AQ749680)である。 RP11 - 115211 (—鎖)の末 端の部分塩基配列は、配列番号 2に記載の塩基配列(GenBankァクセッション番号 : AQ749954)である。  (GenBank accession number: AQ749680). The partial base sequence at the end of RP11-115211 (—strand) is the base sequence described in SEQ ID NO: 2 (GenBank accession number: AQ749954).
[0010] RP11— 1129E18 ( +鎖)領域の末端の部分塩基配列は、配列番号 3に記載の塩 基酉己歹 IJ (GenBankァクセッション番号: AQ697738)である。 RP11— 1129E18 (— 鎖)領域の末端の部分塩基配列は、配列番号 4に記載の塩基配列(GenBankァク セッション番号: AQ720255)である。  [0010] The partial base sequence at the end of the RP11— 1129E18 (+ strand) region is Shiki Iki (GenBank accession number: AQ697738) described in SEQ ID NO: 3. The partial base sequence at the end of the RP11-1129E18 (-strand) region is the base sequence described in SEQ ID NO: 4 (GenBank accession number: AQ720255).
[0011] RP11— 625J15 ( +鎖)領域の末端の部分塩基配列は、配列番号 5に記載の塩基 配列(GenBankァクセッション番号: AQ405632)である。 RP11— 625J15 ( 鎖) 領域の末端の部分塩基配列は、配列番号 6に記載の塩基配列(GenBankァクセッ シヨン番号: AQ455093)である。  [0011] The partial base sequence at the end of the RP11-625J15 (+ strand) region is the base sequence described in SEQ ID NO: 5 (GenBank accession number: AQ405632). The partial base sequence at the end of the RP11-625J15 (strand) region is the base sequence described in SEQ ID NO: 6 (GenBank Accession No: AQ455093).
[0012] RP11 - 254K1 (+鎖)領域の末端の部分塩基配列は、配列番号 7に記載の塩基 配列(GenBankァクセッション番号: AQ478938)である。 RP11— 254K1 (—鎖) 領域の末端の部分塩基配列は、配列番号 8に記載の塩基配列(GenBankァクセッ シヨン番号: AQ478940)である。これら 4クローンは部分的に重複しているためそれ らから形成される Contig配列を作成することが可能である。 [0012] The partial base sequence at the end of the RP11-254K1 (+ strand) region is the base set forth in SEQ ID NO: 7. Sequence (GenBank accession number: AQ478938). The partial base sequence at the end of the RP11—254K1 (—strand) region is the base sequence described in SEQ ID NO: 8 (GenBank Accession No: AQ478940). Since these 4 clones partially overlap, it is possible to create a contig sequence formed from them.
[0013] 一方、 International Human Genome sequencing Consortiumiま、ヒトクノ ムの塩基配列を決定し、その塩基配列を公表している(Nature Vol. 431、 pp. 93 1 945、 2004)。 NCBIは、この結果を受けて、ヒトゲノム断片の Contig配列を公表 している。 [0013] On the other hand, the human human genome sequencing consortium has been determined and its nucleotide sequence has been published (Nature Vol. 431, pp. 93 1 945, 2004). In response to this result, NCBI has published the Contig sequence of the human genome fragment.
[0014] GenBankァクセッション番号 NT— 019273.18に記載の塩基配列は、ヒト第 1染色 体のゲノム断片の Contig酉己歹 IJである。 RP11— 115211、 RP11 - 1129E18, RP1 1— 625J15及び RP11— 254K1は、 GenBankァクセッション番号 NT— 019273· 1 8に記載の塩基酉己歹 IJにおいて、それぞれ、第 10597582番目力、ら第 10752202番 目、第 10746859番目力、ら第 10891175番目、第 10785181番目力、ら第 109498 22番目、第 10934043番目力、ら第 11121868番目に申目当すると考えられる。  [0014] The nucleotide sequence described in GenBank accession number NT-0192713.18 is Contig's IJ of the genomic fragment of the first human chromosome. RP11—115211, RP11—1129E18, RP1 1—625J15 and RP11—254K1 are the 10597582th power and 10752202, respectively, in the base banking IJ described in GenBank accession number NT—019273 · 18. No. 10746859, No. 10891175, No. 10785181, No. 109498, No. 22, 10934043, No. 11, etc.
[0015] 従って、これら 4クローンからなる Contig配列は、 GenBankァクセッション番号 NT _019273· 18に記載の塩基酉己歹 IJの第 10597582番目力、ら第 11 121868番目まで の塩基配列(およそ、 0· 5Mbpの領域)に相当すると考えられる。  [0015] Therefore, the Contig sequence consisting of these 4 clones is the base sequence from the 1097582nd position of IJ No. 10597582, described in GenBank accession number NT _019273-18 (about 0 · It is considered to correspond to an area of 5 Mbps.
[0016] 以上のことから、本発明に係るアンプリコンは、 GenBankァクセッション番号 NT— 019273.18に記載の塩基酉己歹 IJの第 10597582番目力、ら第 11 121868番目まで力、 らなる塩基配列で表されるポリヌクレオチドにより実質的に示される染色体領域と考え ること力 Sでさる。  [0016] Based on the above, the amplicon according to the present invention is a base sequence consisting of a base sequence from GenBank accession number NT-0119273.18, IJ No. 10597582, No. 11 to No. 121,868. The force S is considered to be a chromosomal region substantially represented by the polynucleotide represented by.
[0017] GenBankァクセッション番号 NT— 019273.18に記載の塩基配列の第 1059758 2番目から第 11121868番目までからなる塩基配列で表されるポリヌクレオチドによ り実質的に示される染色体領域には、 TRIM33遺伝子、 BCAS2 (breast carcino ma amplified sequence— 2)遺伝子および DENND2C (DENN/MADD dom ain containing 2C)遺伝子が配座していると考えられている。  [0017] The chromosomal region substantially represented by the polynucleotide represented by the nucleotide sequence consisting of nucleotide sequence 1059758 from the 2nd to 11121868 of the nucleotide sequence described in GenBank accession number NT-0192773.18 is TRIM33 The gene, BCAS2 (breast carcino ma amplified sequence—2) gene and DENND2C (DENN / MADD dom ain containing 2C) gene are considered to be coordinated.
[0018] TRIM33遺伝子の転写領域は、 GenBankァクセッションナンバー NT— 019273.  [0018] The transcription region of the TRIM33 gene is GenBank accession number NT-019273.
18に記載された塩基酉己歹 IJの第 10843084番目力、ら 10961466番目までの塩基酉己 列で表されるポリヌクレオチドにより実質的に示される染色体領域に配座していると考 えられている。従って、 TRIM33遺伝子の転写領域は、 RP11— 625J15及び RP11 — 254K1で形成される Contig配列で表されるポリヌクレオチドにより実質的に示さ れる染色体領域に配座して!/、ると考えられる。 Base Iki listed in 18 IJ's 10843084th power, et al. It is considered to be conformed to the chromosomal region substantially indicated by the polynucleotide represented by the column. Therefore, it is considered that the transcription region of the TRIM33 gene is conformed to the chromosomal region substantially represented by the polynucleotide represented by the contig sequence formed by RP11-625J15 and RP11-254K1! /.
[0019] BCAS2遺伝子の転写領域は、 GenBankァクセッションナンバー NT— 019273.1 8に記載された塩基配列の第 11017863番目力も第 11031950番目までの塩基配 列で表されるポリヌクレオチドにより実質的に示される染色体領域に配座していると考 えられている。従って、 BCAS2遺伝子の転写領域は、 RP11— 254K1で表されるポ リヌクレオチドにより実質的に示される染色体領域に配座していると考えられる。  [0019] The transcription region of the BCAS2 gene is substantially indicated by a polynucleotide represented by the nucleotide sequence up to the 11031950th position of the 11017863th nucleotide of the nucleotide sequence described in GenBank accession number NT-019273.18. It is thought to be conformed to the chromosomal region. Therefore, the transcription region of the BCAS2 gene is considered to be conformed to the chromosomal region substantially indicated by the polynucleotide represented by RP11-254K1.
[0020] DENND2C遺伝子の転写領域は、 GenBankァクセッションナンバー NT— 0192 73.18に記載された塩基酉己歹 IJの第 11033155番目力、ら第 11120417番目までの塩 基配列で表されるポリヌクレオチドで実質的に示される染色体領域に配座していると 考えられる。従って、 DENND2C遺伝子の転写領域は、 RP11— 254K1で表される ポリヌクレオチドに実質的に相当する染色体領域に配座していると考えられる。  [0020] The transcription region of the DENND2C gene is a polynucleotide represented by the nucleotide sequence from the 11033155th force of IJ's 11033155th to 11120417th described in GenBank accession number NT-0192 73.18. It is thought that it is actually conformed to the chromosomal region shown. Therefore, the transcription region of the DENND2C gene is considered to be coordinated with a chromosomal region substantially corresponding to the polynucleotide represented by RP11-254K1.
[0021] 本発明により、本発明に係るアンプリコンからなる小細胞癌又は乳癌、好ましくは肺 小細胞癌又は乳癌の診断用マーカーが提供される。肺小細胞癌などの小細胞癌又 は乳癌を発症して!/、る患者のその疾患部位は、対照となる健常者の該部位と比較し て、本発明に係るアンプリコンが増幅していると考えられる。従って、ある者から分離 した肺又は乳組織由来の生体試料において、健常者から分離したそれと比較して、 本発明に係るアンプリコンが増幅している場合には、その者は、肺小細胞癌などの小 細胞癌又は乳癌に罹患している力、またはその可能性が高い、及び/又は、予後が 不良であるまたはその可能性が高いと診断される。  The present invention provides a diagnostic marker for small cell cancer or breast cancer, preferably lung small cell cancer or breast cancer, comprising the amplicon according to the present invention. The disease site of a patient who develops small cell cancer such as small cell lung cancer or breast cancer is amplified by the amplicon according to the present invention as compared to the site of a healthy subject as a control. It is thought that there is. Therefore, when the amplicon according to the present invention is amplified in a biological sample derived from lung or milk tissue isolated from a certain person, compared to that isolated from a healthy person, the person is Diagnosed as having or having a high probability of suffering from small cell cancer or breast cancer, and / or having a poor or high prognosis.
[0022] 本発明に係るアンプリコンが増幅することは、該アンプリコンに含まれる部分も増幅 することを意味する。従って、本発明に係るアンプリコンに含まれる部分も、肺小細胞 癌などの小細胞癌又は乳癌の診断用マーカーとなることは明らかである。  [0022] Amplification of the amplicon according to the present invention means that the portion included in the amplicon is also amplified. Therefore, it is clear that the portion contained in the amplicon according to the present invention also serves as a diagnostic marker for small cell cancer such as small cell lung cancer or breast cancer.
[0023] すなわち、本発明により、本発明に係るアンプリコンに含まれる部分からなる小細胞 癌又は乳癌、好ましくは肺小細胞癌又は乳癌の診断用マーカーが提供される。本発 明に係るアンプリコンに含まれる部分として、 TRIM33遺伝子が配座する染色体領 域、 BCAS2遺伝子が配座する染色体領域、 DENND2C遺伝子が配座する染色体 領域、 TRIM33遺伝子の転写領域が配座する染色体領域、 BCAS2遺伝子の転写 領域が配座する染色体領域、 DENND2C遺伝子の転写領域が配座する染色体領 域、 TRIM33遺伝子、 BCAS2遺伝子、 DENND2C遺伝子が例示される。 That is, according to the present invention, there is provided a diagnostic marker for small cell cancer or breast cancer, preferably small cell lung cancer or breast cancer comprising a portion contained in the amplicon according to the present invention. As part of the amplicon according to the present invention, the chromosome region where the TRIM33 gene is conformed Region, chromosomal region where BCAS2 gene is conformed, chromosomal region where DENND2C gene is conformed, chromosomal region where TRIM33 gene transcription region is conformed, chromosomal region where BCAS2 gene transcription region is conformed, transcription region of DENND2C gene Examples include the chromosomal region to be conformed, TRIM33 gene, BCAS2 gene, and DENND2C gene.
[0024] 本発明者らは、ヒト肺小細胞癌由来の 20種類の細胞株を対象として、 TRIM33遺 伝子、 BCAS2遺伝子、 DENND2C遺伝子の発現解析を行った結果、それぞれの 遺伝子の発現量 (各遺伝子の転写産物の量)は、健常の肺における各遺伝子の発 現量と比較して、亢進してレ、ることを見出した(図 1 (E) )。さらに、本発明者は、肺小 細胞癌由来の細胞株(4種)および乳癌由来の細胞株(MCF7)を用いて、細胞内の TRIM33タンパク質量を測定した結果、ヒト肺上皮細胞株 A549内の TRIM33タン パク質量と比べて、亢進していることを見出した(図 2 (B) )。また、本発明者らは、免 疫化学的手法を用いて、混合型小細胞肺癌(小細胞癌と線癌の混合)組織において は小細胞癌の部分のみに、 TRIM33遺伝子の発現が亢進していることを見出した( 図 3)。さらに、本発明者らは、免疫化学的手法を用いて、乳癌組織において TRIM3 3遺伝子の発現が亢進していることを見出した(図 3)。また、乳癌組織において TRI M33遺伝子の発現が強く検出される患者の予後は、乳癌組織において TRIM33遺 伝子の発現が検出されないまたは弱く検出される患者の予後と比較して、不良であ ることを見出した(図 6 (A) (B) )。  [0024] As a result of the expression analysis of the TRIM33 gene, the BCAS2 gene, and the DENND2C gene in 20 cell lines derived from human small cell lung cancer, the present inventors have found that the expression levels of the respective genes ( The amount of each gene transcript) was found to be increased compared to the expression level of each gene in healthy lungs (Fig. 1 (E)). Furthermore, the present inventor measured the amount of TRIM33 protein in cells using a cell line derived from small cell lung cancer (4 types) and a cell line derived from breast cancer (MCF7). It was found to be increased compared to the TRIM33 protein mass (Fig. 2 (B)). In addition, the present inventors use an immunochemical technique to increase the expression of the TRIM33 gene only in the small cell carcinoma part in a mixed small cell lung cancer (mixed small cell cancer and linear cancer) tissue. (Figure 3). Furthermore, the present inventors have found that expression of the TRIM3 3 gene is increased in breast cancer tissue using an immunochemical technique (FIG. 3). In addition, the prognosis of patients whose TRI M33 gene expression is strongly detected in breast cancer tissue is poor compared to the prognosis of patients whose expression of TRIM33 gene is not detected or weakly detected in breast cancer tissue. (Fig. 6 (A) (B)).
[0025] 以上の知見から、本発明に係るアンプリコンに配座する遺伝子の転写産物及び該 遺伝子がコードする蛋白質も、肺小細胞癌などの小細胞癌又は乳癌の診断用マー カーとなると考えられる。すなわち、ある者から分離した肺又は乳組織由来の生体試 料において該遺伝子の発現が亢進している場合には、その者は、肺小細胞癌などの 小細胞癌又は乳癌に罹患している力、またはその可能性が高い、及び/又は、予後 が不良であるまたはその可能性が高いと診断される。  [0025] From the above findings, it is considered that the transcription product of the gene conforming to the amplicon according to the present invention and the protein encoded by the gene also serve as a diagnostic marker for small cell cancer such as small cell lung cancer or breast cancer. It is done. That is, when the expression of the gene is increased in a biological sample derived from lung or milk tissue isolated from a certain person, the person is suffering from small cell cancer such as small cell lung cancer or breast cancer. Diagnosed as having a strong or likely likelihood and / or having a poor or likely prognosis.
[0026] 従って、本発明により、本発明に係るアンプリコンに配座する遺伝子の転写産物又 は該遺伝子がコードする蛋白質からなる小細胞癌又は乳癌、好ましくは肺小細胞癌 又は乳癌のの診断用マーカーが提供される。本発明に係るアンプリコンに配座する 遺伝子として、 TRIM33遺伝子、 BCAS2遺伝子、 DENND2C遺伝子が例示される [0027] 別の観点からは、本発明により、小細胞癌又は乳癌、好ましくは肺小細胞癌又は乳 癌の診断薬であってヒトを含む哺乳動物において、本発明に係るアンプリコンの増幅 及び/又は該アンプリコンに配座する遺伝子の発現亢進を検出することができる診 断薬が提供される。好ましい態様によれば、ヒトの小細胞癌又は乳癌、好ましくはヒト の肺小細胞癌又は乳癌の診断用の上記診断薬、小細胞癌又は乳癌、好ましくは肺 小細胞癌又は乳癌の存在が疑われる患者の診断用の上記診断薬、及び小細胞癌 又は乳癌患者、好ましくは肺小細胞癌又は乳癌患者の予後診断用の上記診断薬が 提供される。 Therefore, according to the present invention, diagnosis of small cell cancer or breast cancer, preferably small cell lung cancer or breast cancer comprising a transcription product of a gene conforming to the amplicon according to the present invention or a protein encoded by the gene, is preferable. Markers are provided. Examples of genes conforming to the amplicon according to the present invention include TRIM33 gene, BCAS2 gene, and DENND2C gene. [0027] From another viewpoint, according to the present invention, amplification of the amplicon according to the present invention in mammals including humans, which is a diagnostic agent for small cell cancer or breast cancer, preferably small cell lung cancer or breast cancer, and A diagnostic agent capable of detecting an increased expression of a gene conforming to the amplicon is provided. According to a preferred embodiment, the presence of the above-mentioned diagnostic agent for diagnosis of human small cell cancer or breast cancer, preferably human small cell cancer or breast cancer, small cell cancer or breast cancer, preferably small cell lung cancer or breast cancer is suspected. And the above diagnostic agent for prognosis of patients with small cell cancer or breast cancer, preferably small cell lung cancer or breast cancer.
[0028] より好まし!/、態様によれば、ヒトから分離された生体試料にぉレ、て本発明に係るアン プリコンの増幅及び/又は該アンプリコンに配座する遺伝子の発現亢進を検出する ことができる上記診断薬; GenBankァクセッションナンバー NT— 019273.18に記 載された塩基配列の第 10597582番目から第 11121868番目までからなる塩基配 列で表されるポリヌクレオチドにより実質的に示される染色体領域とストリンジェントな 条件下でハイブリダィズする核酸プローブを含む上記診断薬; GenBankァクセッショ ンナンバー NT— 019273.18に記載された塩基配列の第 10597582番目力も第 11 121868番目までからなる塩基配列若しくはその相補的塩基配列に含まれる連続す る少なくとも 15塩基からなるポリヌクレオチド又は該ポリヌクレオチドとストリンジェント な条件下でハイブリダィズする少なくとも 15塩基からなるポリヌクレオチドを含む上記 診断薬; TRIM33遺伝子が配座する染色体領域とストリンジェントな条件下でハイブ リダィズする核酸プローブを含む上記診断薬; TRIM33遺伝子の転写領域が配座 する染色体領域とストリンジェントな条件下でハイブリダィズする核酸プローブを含む 上記診断薬; TRIM33遺伝子とストリンジェントな条件下でハイブリダィズする核酸プ ローブを含む上記診断薬; GenBankァクセッションナンバー NT— 019273.18に記 載された塩基配列の第 10843084番目力、ら第 10961466番目までからなる塩基配 列若しくはその相補的塩基配列に含まれる連続する少なくとも 15塩基からなるポリヌ クレオチド又は該ポリヌクレオチドとストリンジェントな条件下でハイブリダィズする少な くとも 15塩基からなるポリヌクレオチドを含む上記診断薬; BCAS2遺伝子が配座する 染色体領域とストリンジェントな条件下でハイブリダィズする核酸プローブを含む上記 診断薬; BCAS2遺伝子の転写領域が配座する染色体領域とストリンジェントな条件 下でハイブリダィズする核酸プローブを含む上記診断薬; BCAS2遺伝子とストリンジ ェントな条件下でハイブリダィズする核酸プローブを含む上記診断薬; GenBankァク セッションナンバー NT— 019273.18に記載された塩基配列の第 11017863番目 から第 11031950番目までからなる塩基配列若しくはその相補的塩基配列に含まれ る連続する少なくとも 15塩基からなるポリヌクレオチド又は該ポリヌクレオチドとストリン ジェントな条件下でハイブリダィズする少なくとも 15塩基からなるポリヌクレオチドを含 む上記診断薬; DENND2C遺伝子が配座する染色体領域とストリンジェントな条件 下でハイブリダィズする核酸プローブを含む上記診断薬; DENND2C遺伝子の転 写領域が配座する染色体領域とストリンジェントな条件下でハイブリダィズする核酸プ ローブを含む上記診断薬; DENND2C遺伝子とストリンジェントな条件下でハイプリ ダイズする核酸プローブを含む上記診断薬; GenBankァクセッションナンバー NT— 019273.18に記載された塩基酉己歹 IJの第 11033155番目力、ら第 11120417番目ま でからなる塩基配列若しくはその相補的塩基配列に含まれる連続する少なくとも 15 塩基からなるポリヌクレオチド又は該ポリヌクレオチドとストリンジェントな条件下でハイ ブリダィズする少なくとも 15塩基からなるポリヌクレオチドを含む上記診断薬;本発明 に係るアンプリコンに配座する遺伝子の転写産物とストリンジェントな条件下でハイブ リダィズする少なくとも 15塩基からなるポリヌクレオチドを含む上記診断薬; TRIM33 遺伝子の転写産物とストリンジェントな条件下でハイブリダィズする少なくとも 15塩基 からなるポリヌクレオチドを含む上記診断薬; BCAS2遺伝子の転写産物とストリンジ ェントな条件下でハイブリダィズする少なくとも 15塩基からなるポリヌクレオチドを含む 上記診断薬; DENND2C遺伝子の転写産物とストリンジェントな条件下でハイブリダ ィズする少なくとも 15塩基からなるポリヌクレオチドを含む上記診断薬;配列表の配列 番号 1〜8のうちいずれ力、 1の塩基配列からなる核酸プローブを含む上記診断薬;配 列表の配列番号 9〜; 14のうちいずれ力、 1の塩基配列からなるプライマーを含む上記 診断薬;検出する手段が放射性同位元素標識された検出する手段である上記診断 薬;本発明に係るアンプリコンに配座する遺伝子の遺伝子産物を特異的に認識する 抗体を含む上記の診断薬; TRIM33タンパク質、 BCAS2タンパク質又は DENND2 Cタンパク質を特異的に認識する抗体を含む上記の診断薬;モノクローナル抗体又 はポリクローナル抗体である上記診断薬;配列表の配列番号 21に記載のアミノ酸配 列で表されるポリペプチドを特異的に認識する抗体を含む上記の診断薬;及び検出 する手段がェンザィムィムノアッセィ (ELISA)である上記の診断薬が提供される。 [0028] More preferred! / According to the embodiment, amplification of the amplicon according to the present invention and / or increased expression of a gene conforming to the amplicon is detected in a biological sample isolated from a human. Chromosome substantially represented by the polynucleotide represented by the nucleotide sequence consisting of the 10597582th to 11121868th nucleotide sequences described in GenBank Accession Number NT-0192773.18 The above diagnostic agent comprising a nucleic acid probe that hybridizes with a region under stringent conditions; the base sequence described in GenBank Accession Number NT-0192713.18 and the base sequence consisting of up to 11 121868, or its complementary base A polynucleotide comprising at least 15 consecutive nucleotides contained in the sequence or a condition that is stringent with the polynucleotide The above diagnostic agent comprising a polynucleotide consisting of at least 15 bases that hybridizes underneath; a chromosomal region to which the TRIM33 gene conforms and a nucleic acid probe that hybridizes under stringent conditions; a transcription region of the TRIM33 gene Diagnostic agent containing a nucleic acid probe that hybridizes under stringent conditions with a chromosomal region that sits; Diagnostic agent containing a nucleic acid probe that hybridizes under stringent conditions with the TRIM33 gene; GenBank accession number NT-019273.18 A nucleotide sequence consisting of at least 15 bases contained in the 10843084th power of the base sequence described above, the 10961466th base sequence or the complementary base sequence thereof or a stringent condition with the polynucleotide Hybridize with at least 15 BCAS2 gene is conformationally; the diagnostic agent comprising the polynucleotide consisting of group The above diagnostic agent comprising a nucleic acid probe that hybridizes under stringent conditions with a chromosomal region; the above diagnostic agent comprising a nucleic acid probe that hybridizes under stringent conditions with a chromosomal region where the transcription region of the BCAS2 gene conforms; and the BCAS2 gene The above diagnostic agent comprising a nucleic acid probe that hybridizes under stringent conditions; the nucleotide sequence of 11017863 to 11031950 of the nucleotide sequence described in GenBank session number NT-0192713.18 or its complementary nucleotide sequence The diagnostic agent comprising a polynucleotide comprising at least 15 consecutive nucleotides or a polynucleotide comprising at least 15 nucleotides that hybridizes with the polynucleotide under stringent conditions; a chromosomal region to which the DENND2C gene is coordinated and a stringent Under the conditions The above-mentioned diagnostic agent containing a nucleic acid probe that hybridizes; the above-mentioned diagnostic agent containing a nucleic acid probe that hybridizes under stringent conditions with the chromosomal region where the transcriptional region of the DENND2C gene conforms; and highly amplified under stringent conditions with the DENND2C gene; The above-mentioned diagnostic agent containing a nucleic acid probe for soybean; The diagnostic agent comprising a polynucleotide comprising at least 15 consecutive nucleotides or a polynucleotide comprising at least 15 nucleotides hybridized under stringent conditions with the polynucleotide; a gene conforming to the amplicon according to the present invention; Hybridize with transcripts under stringent conditions The above diagnostic agent comprising a polynucleotide comprising at least 15 bases; a diagnostic agent comprising a polynucleotide comprising at least 15 bases that hybridizes with a TRIM33 gene transcription product under stringent conditions; the BCAS2 gene transcription product and stringage A diagnostic agent comprising a polynucleotide comprising at least 15 bases that hybridizes under the present conditions; a diagnostic agent comprising a polynucleotide comprising at least 15 bases that hybridizes with the DENND2C gene transcript under stringent conditions; The diagnostic agent comprising a nucleic acid probe consisting of one nucleotide sequence of any one of the sequence numbers 1 to 8 in the sequence table; the above comprising a primer consisting of the nucleotide sequence of the first one of the sequence numbers 9 to 14 of the sequence listing Diagnostic agent; means to detect is radioisotope-labeled means to detect The above diagnostic agent; specifically recognizes the gene product of the gene conforming to the amplicon according to the present invention The above diagnostic agent comprising an antibody; the above diagnostic agent comprising an antibody that specifically recognizes TRIM33 protein, BCAS2 protein or DENND2 C protein; the above diagnostic agent which is a monoclonal antibody or a polyclonal antibody; Provided is the above-mentioned diagnostic agent comprising an antibody that specifically recognizes the polypeptide represented by the amino acid sequence described above; and the above-mentioned diagnostic agent whose means for detection is Enzymno assay (ELISA) .
[0029] 別の観点からは、本発明により、小細胞癌又は乳癌、好ましくは肺小細胞癌又は乳 癌の診断方法であって、ヒトから分離された生体試料において、本発明に係るアンプ リコンの増幅及び/又は該アンプリコンに配座する遺伝子の発現亢進、好ましくは、 TRIM33遺伝子の増幅及び/又は TRIM33遺伝子の発現亢進を検出する工程を 含む診断方法が提供される。例えば、このような診断方法として、小細胞癌又は乳癌 、好ましくは肺小細胞癌又は乳癌の患者から分離された生体試料において、本発明 に係るアンプリコンの増幅及び/又は該アンプリコンに配座する遺伝子の発現亢進、 好ましくは、 TRIM33遺伝子の増幅及び/又は TRIM33遺伝子の発現亢進を検出 する工程を含む、小細胞癌又は乳癌、好ましくは肺小細胞癌又は乳癌の予後診断 方法が例示される。上記発明の好ましい態様によれば、上記のいずれかの診断薬を 用いることを特徴とする上記の診断方法が提供される。  From another viewpoint, according to the present invention, there is provided a method for diagnosing small cell cancer or breast cancer, preferably small cell lung cancer or breast cancer, wherein the amplicon according to the present invention is used in a biological sample isolated from a human. And / or an increase in the expression of a gene conforming to the amplicon, and preferably a step of detecting an increase in the TRIM33 gene and / or an increase in the expression of the TRIM33 gene is provided. For example, as such a diagnostic method, amplification of the amplicon according to the present invention and / or conformation to the amplicon in a biological sample isolated from a patient with small cell cancer or breast cancer, preferably lung small cell cancer or breast cancer. A method for prognosing small cell cancer or breast cancer, preferably small cell lung cancer or breast cancer, which comprises a step of detecting an increase in expression of the gene to be detected, preferably amplification of TRIM33 gene and / or detection of increased expression of TRIM33 gene . According to a preferred aspect of the present invention, there is provided the above diagnostic method characterized by using any one of the above diagnostic agents.
[0030] 本発明者らは、 TRIM33遺伝子の発現を抑制する siRNAにより、肺小細胞癌など の小細胞癌又は乳癌由来の細胞の増殖が阻害されることを見出した(図 5)。従って 、さらに別の観点からは、小細胞癌細胞又は乳癌細胞、好ましくは肺小細胞癌細胞 又は乳癌細胞に対する増殖阻害作用を有する物質のスクリーニング方法であって、 本発明に係るアンプリコンに配座する遺伝子好ましくは TRIM33遺伝子の増幅及び /又は発現を抑制する作用を有する物質を選択する工程を含むスクリーニング方法 が提供される。  [0030] The present inventors have found that siRNA that suppresses the expression of the TRIM33 gene inhibits the growth of cells derived from small cell carcinoma such as small cell lung cancer or breast cancer (FIG. 5). Accordingly, from yet another aspect, the present invention provides a method for screening a substance having a growth inhibitory action on small cell cancer cells or breast cancer cells, preferably lung small cell cancer cells or breast cancer cells, which is conformed to the amplicon according to the present invention. A screening method comprising a step of selecting a substance having an action of suppressing amplification and / or expression of a gene, preferably TRIM33 gene, is provided.
[0031] また、本発明により、本発明に係るアンプリコンに配座する遺伝子の増幅及び/又 は発現を抑制する作用を有する物質、好ましくは TRIM33遺伝子の増幅及び/又 は発現を抑制する作用を有する物質、例えば上記のスクリーニング方法によりスクリ 一ユングされた物質を有効成分として含む小細胞癌又は乳癌、好ましくは肺小細胞 癌又は乳癌の治療のための医薬が提供される。 TRIM33遺伝子の発現を抑制する 作用を有する物質として、配列表の配列番号 17に記載の塩基配列で表されるポリヌ クレオチドと配列表の配列番号 18に記載の塩基配列で表されるポリヌクレオチドとか らなる 2重鎖ポリヌクレオチド、配列表の配列番号 19に記載の塩基配列で表されるポ リヌクレオチドと配列表の配列番号 20に記載の塩基配列で表されるポリヌクレオチド とからなる 2重鎖ポリヌクレオチドを例示することができる。 [0031] Further, according to the present invention, a substance having an action of suppressing amplification and / or expression of a gene conforming to the amplicon according to the present invention, preferably an action of suppressing amplification and / or expression of a TRIM33 gene. There is provided a medicament for treating small cell cancer or breast cancer, preferably small cell lung cancer or breast cancer, which contains, as an active ingredient, a substance having the above, for example, a substance screened by the screening method described above. Suppresses expression of TRIM33 gene As a substance having an action, a double-stranded polynucleotide comprising a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 17 of the Sequence Listing and a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 18 of the Sequence Listing; Examples include a double-stranded polynucleotide comprising a polynucleotide represented by the base sequence set forth in SEQ ID NO: 19 in the sequence listing and a polynucleotide represented by the base sequence set forth in SEQ ID NO: 20 in the sequence listing. .
[0032] さらに、小細胞癌又は乳癌、好ましくは肺小細胞癌などの小細胞癌、さらに好ましく は肺小細胞癌の治療方法であって、上記の物質の治療有効量を肺小細胞癌などの 小細胞癌患者又は乳癌患者、好ましくは肺小細胞癌などの小細胞癌患者、さらに好 ましくは肺小細胞癌患者に投与する工程を含む治療方法、及び上記医薬の製造の ための TRIM33遺伝子の増幅及び/又は発現を抑制する作用を有する物質の使 用が本発明により提供される。  [0032] Further, a method for treating small cell cancer or breast cancer, preferably small cell cancer such as small cell lung cancer, more preferably small cell lung cancer, wherein a therapeutically effective amount of the above substance is applied to small cell lung cancer or the like. A therapeutic method comprising the steps of administering to a small cell cancer patient or a breast cancer patient, preferably a small cell cancer patient such as a small cell lung cancer, more preferably a small cell lung cancer patient, and TRIM33 for the manufacture of the medicament Use of a substance having an action of suppressing gene amplification and / or expression is provided by the present invention.
[0033] また、本発明により、肺小細胞癌などの小細胞癌細胞又は乳癌細胞に対する増殖 阻害剤であって、以下の群より選ばれる 2重鎖ポリヌクレオチドを含む増殖阻害剤が 提供される。 (1)配列表の配列番号 17に記載の塩基配列で表されるポリヌクレオチド と配列表の配列番号 18に記載の塩基配列で表されるポリヌクレオチドとからなる 2重 鎖ポリヌクレオチド、及び(2)配列表の配列番号 19に記載の塩基配列で表されるポリ ヌクレオチドと配列表の配列番号 20に記載の塩基配列で表されるポリヌクレオチドと 力、らなる 2重鎖ポリヌクレオチド。  [0033] The present invention also provides a growth inhibitor for small cell carcinoma cells such as small cell lung cancer or breast cancer cells, comprising a double-stranded polynucleotide selected from the following group. . (1) a double-stranded polynucleotide comprising a polynucleotide represented by the base sequence set forth in SEQ ID NO: 17 of the sequence listing and a polynucleotide represented by the base sequence set forth in SEQ ID NO: 18 of the sequence listing; and (2 ) A double-stranded polynucleotide comprising a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 19 in the Sequence Listing and a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 20 in the Sequence Listing.
図面の簡単な説明  Brief Description of Drawings
[0034] [図 1]SCLC細胞株中の TRM33の増幅((A)〜(D))及び発現レベル((E))を示した図で ある。 (A) LU-140細胞の代表的な CGHアレイイメージを示す。(B) LU-140細胞中の 第 1染色体の複製数プロファイルを示す。(C) SCLC細胞株 LU-140の分裂中期染色 体にハイブリダィズさせた RP11-625J15(TRIM33、緑シグナル)及びコントロール RP11 -315D10(RET、赤シグナル)による代表的な FISHイメージを示す。(D) LU-140細胞株 中の 1ρ13·2部位のアンプリコンマップを示す。(E) RT-PCR分析で測定した SCLC細 胞株及び正常肺における TRIM33の発現を示す。  [0034] FIG. 1 shows the TRM33 amplification ((A) to (D)) and expression level ((E)) in SCLC cell lines. (A) A representative CGH array image of LU-140 cells is shown. (B) shows the replication number profile of chromosome 1 in LU-140 cells. (C) Representative FISH images of RP11-625J15 (TRIM33, green signal) and control RP11-315D10 (RET, red signal) hybridized to the metaphase chromosome of SCLC cell line LU-140. (D) Amplicon map of 1ρ13.2 site in LU-140 cell line. (E) shows TRIM33 expression in SCLC cell lines and normal lung as measured by RT-PCR analysis.
[図 2]乳癌細胞株における TRM33の増幅及び過剰発現を示した図である。 (A) BAC RP11-625J15(TRIM33、緑シグナノレ)及び RP 11 _315D 10(RET、赤シグナノレ)を用レ、た 乳癌細胞株 MCF7由来の分裂中期染色体に対する 2色 FISH試験の結果を示す。(B) 4種の SCLC、 2種の NSCLC、及び 1種の乳癌細胞株における蛋白抽出物の免疫ブロ FIG. 2 shows TRM33 amplification and overexpression in breast cancer cell lines. (A) BAC RP11-625J15 (TRIM33, green signal) and RP 11 _315D 10 (RET, red signal) were used. The results of a two-color FISH test on metaphase chromosomes derived from the breast cancer cell line MCF7 are shown. (B) Immunoblotting of protein extracts in 4 SCLC, 2 NSCLC, and 1 breast cancer cell lines
[図 3]肺の原発性小細胞癌及び乳癌における TRIM33タンパク質の免疫組織化学染 色を示した写真である。小細胞癌における強い核染色 (A-D)及び腺癌 (D、矢の先端) における低発現、並びに正常肺 (A及び C中の星印)における低発現を示す。侵襲性 腺管細胞癌 、 F)、侵襲性小葉癌 (H、矢の先端)、及び上皮内腺管癌 (G)における強 度の核染色、及び正常乳腺 (H、矢印)における低発現を示す。 FIG. 3 is a photograph showing the immunohistochemical staining of TRIM33 protein in lung primary small cell carcinoma and breast cancer. Shows strong nuclear staining (A-D) in small cell carcinoma and low expression in adenocarcinoma (D, tip of arrow) and low expression in normal lung (stars in A and C). Intensive nuclear staining in invasive ductal cell carcinoma (F), invasive lobular carcinoma (H, arrow tip), and intraepithelial ductal carcinoma (G), and low expression in normal mammary gland (H, arrow) Show.
[図 4]癌細胞増殖に対する TRIM33発現亢進の効果を示した図である。(A)蛋白抽出 物及び抗 Myc抗体を用いたウェスタンブロッツティングの結果を示す。(B)TRIM33トラ ンスフエタト HeLa及び SBC-3細胞により形成されたコロニー数を示す。(C)安定に TRI M33を発現するクローンとして樹立された SBC-3細胞における TRIM33の該 SBC-3細 胞に対する増殖促進作用、及びコントロールとして空ベクター (empty)でトランスフエク トした SBC-3細胞の結果を示す。 FIG. 4 shows the effect of enhanced TRIM33 expression on cancer cell proliferation. (A) Western blotting results using protein extract and anti-Myc antibody are shown. (B) Number of colonies formed by TRIM33 transform HeLa and SBC-3 cells. (C) Growth-promoting action of TRIM33 on SBC-3 cells established as a clone that stably expresses TRI M33, and SBC-3 cells transfected with an empty vector (empty) as a control The results are shown.
[図 5]TRM33 mRNAを標的とする siRNAによる増殖阻害作用を示した図である。 (A) ウェスタンブロッテイングによって決定された TRIM33タンパク質の蛋白のレベルを示 す。(B)SBC_5及び MCF7細胞の生存に対する siRNAオリゴヌクレオチドの影響を示す 。(C)コントロール及び TRM33_siRNA-処理 MCF7細胞に対する TGF- /3の増殖阻害 作用を BrdU取り込みで測定した結果を示す。  FIG. 5 is a graph showing the growth inhibitory effect of siRNA targeting TRM33 mRNA. (A) Shows the protein level of TRIM33 protein determined by Western blotting. (B) shows the effect of siRNA oligonucleotides on the survival of SBC_5 and MCF7 cells. (C) Control and TRM33_siRNA-treated The results of measuring the growth inhibitory action of TGF- / 3 on MCF7 cells by BrdU incorporation are shown.
[図 6]乳癌の 134症例を対象とした TRIM33の発現レベルと生存率の相関を示した図 である。 (A) TRIM33 high群の全体生存期間(overall survival)は TRIM33 low群のそ れと比較して有意に短いことを示す。(B) TRIM33 high群の無増悪期間(progression- free survival)も同様に TRIM33 low群のそれと比較して有意に短いことを示す。  FIG. 6 shows the correlation between the expression level of TRIM33 and the survival rate in 134 cases of breast cancer. (A) The overall survival of the TRIM33 high group is significantly shorter than that of the TRIM33 low group. (B) The progression-free survival of the TRIM33 high group is also significantly shorter than that of the TRIM33 low group.
園 7]肺以外の小細胞癌臨床検体 9例について TRM33染色を行った結果を示す。 発明を実施するための最良の形態 7] Shows the results of TRM33 staining of 9 clinical cases of small cell carcinoma other than lung. BEST MODE FOR CARRYING OUT THE INVENTION
本発明により提供される診断用マーカーは、肺小細胞癌などの小細胞癌又は乳癌 の患者において検出される癌診断用マーカーであり、本発明に係るアンプリコンの増 幅及び/又は本発明に係るアンプリコンに配座する遺伝子の発現亢進を指標とする 癌診断用マーカーである。 The diagnostic marker provided by the present invention is a marker for cancer diagnosis detected in patients with small cell cancer such as small cell lung cancer or breast cancer, and the amplification of the amplicon according to the present invention and / or the present invention. Increased expression of genes conforming to such amplicons It is a marker for cancer diagnosis.
[0036] 本明細書において、「診断用マーカー」とは、ある者のある疾患の罹患の有無、そ の程度又はその術後の予後を判定するために用いられる生体由来物質又は該物質 を起源とする物質を意味する。従って、癌診断用マーカーとは、癌の罹患の有無、そ の程度又はその術後の予後を判定するために用いられる生体由来物質又は該物質 を起源とする物質を意味する。本発明に係る癌診断用マーカーとして好ましく肺小細 胞癌などの小細胞癌又は乳癌診断用マーカーを例示できる。  [0036] In this specification, "diagnostic marker" refers to a biologically-derived substance used to determine the presence or absence of a certain disease, the degree thereof, or the prognosis after surgery, or the substance. Means the substance. Therefore, the marker for cancer diagnosis means a biological substance used for determining the presence or absence of cancer, its degree, or the prognosis after the operation, or a substance originating from the substance. A marker for diagnosing cancer according to the present invention is preferably a marker for diagnosing small cell cancer such as small cell lung cancer or breast cancer.
[0037] 本発明により提供される診断用マーカーの具体的な態様として、本発明に係るアン プリコン若しくはその部分、又は、該アンプリコンに配座する遺伝子、該遺伝子の転 写産物若しくは該遺伝子の遺伝子産物からなることを特徴としている。本発明に係る アンプリコン又はその部分が増幅しており、及び/又は該遺伝子の発現が亢進して いる場合には、診断対象となった個体が肺小細胞癌などの小細胞癌又は乳癌に罹 患している力、、または、その可能性が高いと診断される。あるいは、本発明に係るアン プリコン又はその部分が増幅しており及び/又は該遺伝子の発現が亢進している肺 小細胞癌などの小細胞癌又は乳癌の患者について、その予後は不良であるあるい はその可能性が高レ、と診断される。  [0037] As a specific embodiment of the diagnostic marker provided by the present invention, the amplicon according to the present invention or a portion thereof, or a gene conforming to the amplicon, a transcription product of the gene, or the gene It is characterized by consisting of gene products. When the amplicon according to the present invention or a portion thereof is amplified and / or the expression of the gene is increased, the individual to be diagnosed has small cell cancer such as small cell lung cancer or breast cancer. Diagnosed as having or likely to be affected. Alternatively, the prognosis of a patient with small cell cancer or breast cancer, such as small cell lung cancer, in which the amplicon according to the present invention or a portion thereof is amplified and / or the expression of the gene is increased is poor. It is diagnosed that the possibility is high.
[0038] 本発明に係るアンプリコンに配座する遺伝子として、例えば、 TRIM33遺伝子、 BC AS2遺伝子、 DENND2C遺伝子が例示される。  [0038] Examples of genes that are conformed to the amplicon according to the present invention include TRIM33 gene, BC AS2 gene, and DENND2C gene.
[0039] TRIM33遺伝子(単に「TRIM33」と標記することもある)はアフリカッメガエルの胚 形成において外胚葉形成に関与する遺伝子であり、中胚葉誘導因子である Smad4 をュビキチン化することにより阻害する過程を担っている。この遺伝子の情報は当業 者に利用可能である(非特許文献 l : Cell、 121、 pp. 87— 99、 2005)。 TRIM33遺 伝子は任意の哺乳類動物の染色体に存在しており、かつそれによりコードされる TRI M33タンパク質は任意の哺乳類動物において発現していることから、本発明の癌マ 一力一は任意の哺乳類動物において利用できる力 最も好ましい対象はヒトである。 例えば、ヒト由来の TRIM33遺伝子の全長を配列表の配列番号 22に例示した。  [0039] The TRIM33 gene (sometimes simply referred to as “TRIM33”) is a gene involved in ectoderm formation in Xenopus embryogenesis and is inhibited by ubiquitination of the mesoderm-inducing factor Smad4 Responsible for the process. Information on this gene is available to those skilled in the art (Non-Patent Document l: Cell, 121, pp. 87-99, 2005). Since the TRIM33 gene is present in the chromosome of any mammalian animal and the TRI M33 protein encoded thereby is expressed in any mammalian animal, the cancer marker of the present invention is Forces available in mammals The most preferred subject is a human. For example, the full length of human-derived TRIM33 gene is exemplified in SEQ ID NO: 22 in the sequence listing.
[0040] BCAS2遺伝子は染色体 1ρ13. 3〜21の領域に配座する遺伝子であり、乳癌にお いて増幅する遺伝子として見出された(Cancer Lett., 185, pp.219-223, 2002)。この 遺伝子の情報は当業者にとって利用可能である。例えば、 GenBankァクセッション 番号 NM— 005872に BCAS2遺伝子の ORF全長の塩基配列が開示されている。 [0040] The BCAS2 gene is a gene that conforms to the region of chromosome 1ρ13.3 to 21 and was found to be amplified in breast cancer (Cancer Lett., 185, pp.219-223, 2002). this Genetic information is available to those skilled in the art. For example, GenBank accession number NM-005872 discloses the full-length ORF base sequence of the BCAS2 gene.
[0041] DENND2C遺伝子の情報は当業者にとって利用可能である。例えば、 GenBank ァクセッション番号 NM— 198459に DENND2C遺伝子の ORF全長の塩基配列が開 示されている。 [0041] Information on the DENND2C gene is available to those skilled in the art. For example, the full-length ORF base sequence of the DENND2C gene is disclosed in GenBank accession number NM-198459.
[0042] 本発明に係るアンプリコンは、ヒトを含む哺乳動物から自体公知の方法により入手 可能である。具体的には、例えば、ヒトを含む哺乳動物からその細胞核を分離 ·精製 し、その細胞核に含まれるゲノム DNAを抽出した上、該ゲノム DNAを制限酵素など により断片化し、断片化された該ゲノム DNAを BACなどのベクターへ組み込みこと によりゲノムライブラリーを作成する。このゲノムライブラリーから、本発明に係るアンプ リコンを検出するためのプローブ(例えば、 RP11— 115211、 RP11 - 1129E18, R P11 - 625J15, RP11— 254K1等)用いて、本発明に係るアンプリコンを単離可能 である。あるいは、本発明に係るアンプリコンは、 GenBankァクセッション番号 NT— 019273.18に記載の塩基酉己歹 IJの第 10597582番目力、ら第 11121868番目まで力、 らなる塩基配列で表されるポリヌクレオチドにより実質的に示される染色体領域と考え られるため、例えば、この塩基配列に基づき前記のプローブを設計することも可能で ある。  [0042] The amplicon according to the present invention can be obtained from mammals including humans by a method known per se. Specifically, for example, the cell nucleus is separated and purified from mammals including humans, the genomic DNA contained in the cell nucleus is extracted, the genomic DNA is fragmented with a restriction enzyme, etc. A genomic library is created by incorporating DNA into a vector such as BAC. From this genomic library, the probe for detecting the amplicon according to the present invention (for example, RP11-115211, RP11-1129E18, RP11-625J15, RP11-254K1, etc.) is used to simply amplify the amplicon according to the present invention. Can be separated. Alternatively, the amplicon according to the present invention comprises a polynucleotide represented by a nucleotide sequence consisting of a base sequence of GenBank accession number NT-0192713.18, IJ No. 10597582, No. 11121868, and the like. Since it is considered to be a substantially chromosomal region, for example, the probe can be designed based on this base sequence.
[0043] 本発明により提供される診断用マーカーとして、本発明に係るアンプリコンの一部 又は全部を用いることができ、一部を用いる場合には、 2以上の本発明に係るアンプ リコンの一部を用いてもよい。  [0043] As a diagnostic marker provided by the present invention, a part or all of the amplicon according to the present invention can be used. When a part of the amplicon according to the present invention is used, two or more of the amplicons according to the present invention are used. May be used.
[0044] 本発明に係るアンプリコンの一部として、 TRIM33遺伝子が配座する染色体領域、 BCAS 2遺伝子が配座する染色体領域、 DENND2C遺伝子が配座する染色体領 域、 TRIM33遺伝子の転写領域が配座する染色体領域、 BCAS2遺伝子の転写領 域が配座する染色体領域、 DENND2C遺伝子の転写領域が配座する染色体領域 を例示できる。  [0044] As part of the amplicon according to the present invention, a chromosomal region in which the TRIM33 gene is conformed, a chromosomal region in which the BCAS 2 gene is conformed, a chromosomal region in which the DENND2C gene is conformed, and a transcription region of the TRIM33 gene. Examples include a chromosomal region to which the transcription region of the DENND2C gene conforms, and a chromosomal region to which the transcription region of the BCAS2 gene conforms.
[0045] 本明細書において、ある遺伝子が配座する染色体領域とは、該遺伝子が位置する ゲノムの一部を意味し、該遺伝子のセンス鎖及び対応するアンチセンス鎖!/、ずれを 含むゲノム部分を意味する。本発明に係るアンプリコンの一部の入手は、本発明に 係るアンプリコンの入手方法と同様に、当業者であれば実施可能である。 [0045] In this specification, the chromosomal region in which a certain gene is conformed means a part of the genome in which the gene is located, and the genome including the sense strand of the gene and the corresponding antisense strand! / Means part. To obtain a part of the amplicon according to the present invention, Similar to the method of obtaining such amplicons, those skilled in the art can implement it.
[0046] 本明細書において、「遺伝子」とは高分子 DNAまたは RNAでの一定の領域の塩 基配列により規定される遺伝の作用単位として定義され得る。真核生物の遺伝子は ェキソンやイントロンを含むことができる。さらに遺伝子はプロモーターやオペレータ 一などの特定の制御機能をもつ核酸上の領域も含むことができる。ある遺伝子が配 座する染色体領域のセンス鎖ゃ該遺伝子の転写産物を逆転写してなる cDNAも該 遺伝子に含まれる。 In the present specification, a “gene” can be defined as a unit of genetic function defined by a base sequence of a certain region in high molecular DNA or RNA. Eukaryotic genes can include exons and introns. In addition, a gene can include a region on a nucleic acid having a specific control function such as a promoter or an operator. A sense strand of a chromosomal region in which a gene is coordinated is also included in the gene by reverse transcription of the transcription product of the gene.
[0047] また、本発明に係るアンプリコンの一部として、本発明に係るアンプリコンを形成す る 1本鎖ポリヌクレオチド又はその部分ポリヌクレオチドを例示できる。  [0047] Further, as a part of the amplicon according to the present invention, a single-stranded polynucleotide or a partial polynucleotide thereof forming the amplicon according to the present invention can be exemplified.
[0048] 本発明に係るアンプリコンに配座する遺伝子の取得は、具体的には、該遺伝子の 発現が確認されている適当な起源から常法に従って cDNAライブラリーを調製し、該 ライブラリーから、該遺伝子に特有の適当なプローブやプライマーを用いて所望のク ローンを選択することにより取得可能である。遺伝子の起源としては、該遺伝子の発 現が確認されている各種の細胞や組織、またはこれらに由来する培養細胞(乳癌又 は肺小細胞癌などの小細胞癌由来の細胞株など)を例示できる。  [0048] To obtain a gene conforming to the amplicon according to the present invention, specifically, a cDNA library is prepared from an appropriate source in which the expression of the gene has been confirmed according to a conventional method, and the gene is obtained from the library. It can be obtained by selecting a desired clone using an appropriate probe or primer peculiar to the gene. Examples of gene origin include various cells and tissues in which the gene expression has been confirmed, or cultured cells derived from these cells (such as cell lines derived from small cell carcinoma such as breast cancer or small cell lung cancer). it can.
これら起源からの全 RNAの分離、 mRNAの分離や精製、 cDNAの取得とそのクロ 一ユング等はいずれも常法に従って実施することができる。 cDNAライブラリーからの 所望のクローンの選択は、例えば公知の蛋白質発現系を利用して各クローンについ て発現蛋白質の確認を行い、さらに該蛋白質の機能を指標にして実施できる。 TRI M33の機能として、 Smad4のュビキチン化活性が例示される。  Isolation of total RNA from these sources, isolation and purification of mRNA, acquisition of cDNA and its cloning, etc. can all be performed according to conventional methods. Selection of a desired clone from the cDNA library can be performed, for example, by confirming the expressed protein for each clone using a known protein expression system and further using the function of the protein as an index. An example of the function of TRI M33 is the ubiquitination activity of Smad4.
[0049] TRIM33遺伝子の ORF全長は、例えば、 GenBankァクセッションナンバー NM— 015906. 3又は配列表の配列番号 22に開示されており、これらの配列に基づいて TRIM33遺伝子に特有の適当なプローブやプライマーを設計することが可能である 。このようなプライマーとして、例えば、配列表の配列番号 9および 10に記載の塩基 酉己列からなるプライマーを例示できる。 BCAS2遺伝子の ORF全長は、例えば、 NM —005872. 2に開示されており、本配列に基づいて BCAS2遺伝子に特有の適当 なプローブやプライマーを設計することが可能である。 DENND2C遺伝子の ORF全 長は、例えば、 NM 198459. 2に開示されており、本配列に基づいて DENND2 c遺伝子に特有の適当なプローブやプライマーを設計することが可能である。 [0049] The full-length ORF of the TRIM33 gene is disclosed in, for example, GenBank accession number NM-015906.3 or SEQ ID NO: 22 in the sequence listing. Based on these sequences, an appropriate probe or a specific probe specific to the TRIM33 gene Primers can be designed. An example of such a primer is a primer consisting of a base sequence shown in SEQ ID NOs: 9 and 10 in the sequence listing. The full-length ORF of the BCAS2 gene is disclosed in, for example, NM-005872.2, and it is possible to design appropriate probes and primers specific to the BCAS2 gene based on this sequence. The ORF full length of the DENND2C gene is disclosed in, for example, NM 198459.2, and based on this sequence, DENND2C c It is possible to design appropriate probes and primers specific to the gene.
[0050] 例えば、所望の遺伝子を該遺伝子の発現が確認されている各種の細胞や組織か ら、上記のプライマーを用いて PCR法などの汎用の手段により該遺伝子を取得する こともできる。 PCR法の反応条件は特に限定されず当業者が適宜選択することがで きる力 例えば、 94°Cで 30秒間(変性)、 55°Cで 30秒、 1分間(アニーリング)、 72°Cで 2 分間(伸長)からなる反応工程を 1サイクルとして、例えば 30サイクル行った後、 72°C で 7分間反応させる条件などを挙げることができる。  [0050] For example, the desired gene can be obtained from various cells and tissues in which the expression of the gene has been confirmed by general means such as PCR using the above primers. Reaction conditions for PCR are not particularly limited and can be selected by those skilled in the art. For example, 94 ° C for 30 seconds (denaturation), 55 ° C for 30 seconds, 1 minute (annealing), 72 ° C A reaction step consisting of 2 minutes (elongation) is defined as one cycle. For example, after 30 cycles, the reaction can be performed at 72 ° C for 7 minutes.
[0051] 例えば、 TRIM33遺伝子自体を取得する場合には、増幅された DNA断片を大腸 菌等の宿主で増幅可能な適切なベクター中にクローニングすることができる。プライ マー調製及び目的遺伝子のクローニングなどの手法は当業者に周知かつ慣用であ り、 列えば、モレキュラークロ一ユング第 2版、 Current Protocols in Molecular Biol ogy. Supplementl〜38, John Wiley&Sons (1987— 1997)等に記載の方法に準じて 行うこと力 Sできる。また、ヒト TRIM33遺伝子の塩基配列において 1から数個の塩基の 欠失、置換及び/又は付加を有する塩基配列からなる DNAは、例えば、化学合成 、遺伝子工学的手法、又は突然変異誘発などの当業者に既知の任意の方法で取得 すること力 Sできる。例えば、 TRIM33遺伝子に対して変異原となる薬剤を接触させる 方法又は紫外線を照射する方法のほか、遺伝子工学的手法等を用いることができる 。遺伝子工学的手法の一つである部位特異的変異所発法は特定の位置に特定の 変異を導入できる手法であることから有用であり、上掲モレキュラークローニング第 2 版に記載の方法に準じて行うことができる。 BCAS2遺伝子や DENND2C遺伝子に ついても TRIM33遺伝子と同様に取得され得る。  [0051] For example, when the TRIM33 gene itself is obtained, the amplified DNA fragment can be cloned into an appropriate vector that can be amplified in a host such as Escherichia coli. Techniques such as primer preparation and cloning of target genes are well known and commonly used by those skilled in the art. For example, Molecular Cloning 2nd Edition, Current Protocols in Molecular Biogy. Supplementl-38, John Wiley & Sons (1987—1997). ) Etc. can be performed according to the method described in S). In addition, DNA consisting of a base sequence having a deletion, substitution and / or addition of one to several bases in the base sequence of the human TRIM33 gene is suitable for chemical synthesis, genetic engineering, or mutagenesis. It can be acquired in any way known to the supplier. For example, a genetic engineering technique can be used in addition to a method of contacting a drug that becomes a mutagen with the TRIM33 gene or a method of irradiating ultraviolet rays. Site-specific mutagenesis, which is one of the genetic engineering methods, is useful because it can introduce a specific mutation at a specific position, and follows the method described in Molecular Cloning, 2nd edition, above. It can be carried out. The BCAS2 gene and DENND2C gene can also be obtained in the same manner as the TRIM33 gene.
[0052] 本発明により提供される診断用マーカーとして、本発明に係るアンプリコンに配座 する遺伝子、該遺伝子の転写産物あるいは該遺伝子の遺伝子産物を用いてもよ!/、。 本発明に係るアンプリコンに配座する遺伝子の転写産物は、本発明に係るアンプリコ ンに配座する遺伝子の取得と同様に取得可能である。  [0052] As a diagnostic marker provided by the present invention, a gene conforming to the amplicon according to the present invention, a transcription product of the gene, or a gene product of the gene may be used. The transcript of the gene conforming to the amplicon according to the present invention can be obtained in the same manner as the acquisition of the gene conforming to the amplicon according to the present invention.
[0053] 本発明に係るアンプリコンに配座する遺伝子の遺伝子産物(例えば、 TRIM33タン パク質、 BCAS2タンパク質、 DENND2Cタンパク質など)を取得する方法は特に限 定されないが、典型的には、上記の方法に従って該遺伝子を取得し、この DNAを適 当な発現系に導入することにより該遺伝子産物を発現させ、適宜の方法で発現され たタンパク質を分離及び精製すればよい。ベクターの種類は特に限定されず、例え ば、 自立的に複製するベクター(例えばプラスミド等)、あるいは宿主細胞に導入され た際に宿主細胞のゲノムに組み込まれ、組み込まれた染色体と共に複製されるべク ターなどを利用できる。好ましくは発現ベクターを用いることができ、転写に必要な要 素等(例えばプロモーター、ターミネータ一、選択マーカー等)が機能的に連結され ていることが望ましい。プロモーターは宿主細胞において転写活性を示す DNA配列 であり、宿主の種類に応じて当業者が適宜選択することができる。該遺伝子を含む組 み換えベクターを適当な宿主 (例えば細菌、酵母、真菌、及び高等真核細胞等)に 導入することによって形質転換体を作製することができ、得られた形質転換体を該遺 伝子の発現を可能にする条件下で適切な栄養培地中で培養し、発現した該遺伝子 産物を当業者に利用可能な適宜の手段により分離'精製することができる。 [0053] A method for obtaining a gene product (eg, TRIM33 protein, BCAS2 protein, DENND2C protein, etc.) of a gene conforming to the amplicon according to the present invention is not particularly limited. Obtain the gene according to the method and apply this DNA The gene product may be expressed by introducing it into an appropriate expression system, and the protein expressed by an appropriate method may be separated and purified. The type of vector is not particularly limited. For example, the vector should replicate autonomously (for example, a plasmid), or be integrated into the genome of the host cell when introduced into the host cell and replicated together with the integrated chromosome. You can use a cluster. Preferably, an expression vector can be used, and it is desirable that elements necessary for transcription (eg, promoter, terminator, selection marker, etc.) are functionally linked. A promoter is a DNA sequence that exhibits transcriptional activity in a host cell, and can be appropriately selected by those skilled in the art depending on the type of host. A transformant can be prepared by introducing a recombinant vector containing the gene into an appropriate host (eg, bacteria, yeast, fungi, and higher eukaryotic cells). The gene product is cultured in an appropriate nutrient medium under conditions that allow gene expression, and the expressed gene product can be isolated and purified by an appropriate means available to those skilled in the art.
[0054] 本発明により提供される肺小細胞癌などの小細胞癌又は乳癌の診断薬は、ヒトを含 む哺乳動物において、本発明に係るアンプリコンの増幅及び/又は該アンプリコン に配座する遺伝子の発現亢進を検出することができる診断薬であり、好ましくは、ヒト の肺小細胞癌などの小細胞癌又は乳癌の診断のための診断薬、肺小細胞癌などの 小細胞癌又は乳癌の存在が疑われる患者の診断用の診断薬、ヒトから分離された生 体試料にお!/、て本発明に係るアンプリコンの増幅及び/又は該アンプリコンに配座 する遺伝子の発現亢進を検出することができる診断薬である。  [0054] The diagnostic agent for small cell cancer such as small cell lung cancer or breast cancer provided by the present invention is amplifying and / or conforming to the amplicon according to the present invention in mammals including humans. A diagnostic agent capable of detecting increased expression of a gene, preferably a diagnostic agent for diagnosis of small cell cancer or breast cancer such as human small cell lung cancer, small cell cancer such as small cell lung cancer or Diagnostic agents for diagnosis of patients suspected of having breast cancer, amplification of amplicons according to the present invention and / or increased expression of genes conforming to the amplicons in biological samples isolated from humans It is a diagnostic agent that can detect.
[0055] 本発明に係るアンプリコンが増幅することは、該アンプリコンに含まれる部分も増幅 することを意味する。従って、本発明に係るアンプリコンに含まれる部分を検出するこ とができる診断薬も本発明に係る診断薬に含まれる。  [0055] Amplification of the amplicon according to the present invention means that the portion included in the amplicon is also amplified. Therefore, a diagnostic agent that can detect a portion contained in the amplicon according to the present invention is also included in the diagnostic agent according to the present invention.
[0056] 本明細書において、本発明に係るアンプリコンの増幅とは、本発明に係るアンプリコ ンのコピー数が増加することを意味する。本発明に係るアンプリコンの増幅はその本 来の座位(1ρ13. 2)における増幅に限らず、均一染色領域や二重微染色体におけ る増幅も含まれる。本発明に係るアンプリコンの増幅の程度は、被験試料に含まれる 該アンプリコンの量が対照となる健常体における該アンプリコンの量と比較して増加 して!/、る限りにお!/、て、特に制限されなレ、。 [0057] 本明細書において、本発明に係るアンプリコンに配座する遺伝子とは、本発明に係 るアンプリコンに内包される遺伝子を意味し、例えば、 TRIM33遺伝子、 BCAS2遺 伝子、 DENND2C遺伝子が例示される。本発明に係るアンプリコンに配座する遺伝 子の発現亢進の程度は、被験試料に含まれる該遺伝子の発現が対照となる健常体 における該遺伝子の発現と比べて亢進している限りにおいて、特に制限されない。 In the present specification, amplification of the amplicon according to the present invention means that the number of copies of the amplicon according to the present invention increases. The amplification of the amplicon according to the present invention is not limited to the amplification at its original locus (1ρ13.2), but also includes amplification in a homogeneously stained region or a double microchromosome. The degree of amplification of the amplicon according to the present invention is increased as long as the amount of the amplicon contained in the test sample is increased compared with the amount of the amplicon in the healthy subject as a control! / , And les, not particularly limited. [0057] In the present specification, the gene conformed to the amplicon according to the present invention means a gene included in the amplicon according to the present invention, for example, TRIM33 gene, BCAS2 gene, DENND2C gene Is exemplified. As long as the expression of the gene conforming to the amplicon according to the present invention is enhanced compared to the expression of the gene in a healthy subject as a control, the expression of the gene contained in the test sample is particularly increased. Not limited.
[0058] 本発明により提供される肺小細胞癌などの小細胞癌又は乳癌の診断薬は、肺小細 胞癌などの小細胞癌又は乳癌の存在が疑われるヒトの診断に好適に用いられる。生 体試料として、例えば、採取された血液、体液、喀痰、乳汁、生検組織、又は手術に より分離された組織を例示可能である。  [0058] The diagnostic agent for small cell cancer such as small cell lung cancer or breast cancer provided by the present invention is suitably used for diagnosis of humans suspected of having small cell cancer such as small cell lung cancer or breast cancer. . Examples of the biological sample include collected blood, body fluid, sputum, milk, biopsy tissue, or tissue separated by surgery.
[0059] 本発明に係るアンプリコンの増幅を検出することができる診断薬として、本発明に係 るアンプリコンに配座する遺伝子の増幅を検出することができる診断薬を例示できる [0059] Examples of the diagnostic agent capable of detecting amplification of the amplicon according to the present invention include diagnostic agents capable of detecting amplification of a gene conforming to the amplicon according to the present invention.
。このような診断薬として、例えば、 TRIM33遺伝子の増幅を検出することができる診 断薬、 BCAS2遺伝子の増幅を検出することができる診断薬、 DENND2C遺伝子の 増幅を検出することができる診断薬を例示できる。 . Examples of such diagnostic agents include diagnostic agents that can detect TRIM33 gene amplification, diagnostic agents that can detect BCAS2 gene amplification, and diagnostic agents that can detect DENND2C gene amplification. it can.
[0060] TRIM33遺伝子の増幅を検出することができる診断薬として、例えば、 TRIM33遺 伝子が配座する染色体領域とストリンジェントな条件下でハイブリダィズする核酸プロ ーブを含む診断薬; TRIM33遺伝子の転写領域が配座する染色体領域とストリンジ ェントな条件下でハイブリダィズする核酸プローブを含む診断薬; TRIM33遺伝子と ストリンジェントな条件下でハイブリダィズする核酸プローブを含む診断薬; GenBan kァクセッションナンバー NT— 019273.18に記載された塩基酉己歹 IJの第 10843084 番目から第 10961466番目までからなる塩基配列若しくはその相補的塩基配列に 含まれる連続する少なくとも 15塩基からなるポリヌクレオチド又は該ポリヌクレオチドと ストリンジェントな条件下でハイブリダィズする少なくとも 15塩基からなるポリヌクレオ チドを含む診断薬を例示できる。より具体的には、配列表の配列番号 5〜8に記載の 塩基配列のうちいずれか 1の塩基配列で表される核酸プローブを含む診断薬、配列 表の配列番号 9〜; 10に記載の塩基配列のうちいずれ力、 1の塩基配列からなるプライ マーを含む診断薬を例示できる。  [0060] As a diagnostic agent capable of detecting the amplification of the TRIM33 gene, for example, a diagnostic agent containing a nucleic acid probe that hybridizes under stringent conditions with a chromosomal region in which the TRIM33 gene is conformed; Diagnostic agent containing a nucleic acid probe that hybridizes under stringent conditions with the chromosomal region to which the transcription region conforms; Diagnostic agent containing a nucleic acid probe that hybridizes under stringent conditions with the TRIM33 gene; GenBan kcession number NT— A nucleotide sequence consisting of at least 15 bases contained in the nucleotide sequence consisting of the 10843084th to 10961466th nucleotides of the IJ base described in 019273.18 or its complementary nucleotide sequence, or a stringent condition with the polynucleotide Polynucleoside consisting of at least 15 bases that hybridize below It can be exemplified diagnostic agent comprising a de. More specifically, a diagnostic agent comprising a nucleic acid probe represented by any one of the base sequences described in SEQ ID NOs: 5 to 8 in the sequence listing, SEQ ID NO: 9 to 10 in the sequence listing A diagnostic agent containing a primer consisting of one nucleotide sequence can be exemplified.
[0061] BCAS2遺伝子の増幅を検出することができる診断薬として、例えば、 BCAS2遺 伝子が配座する染色体領域とストリンジェントな条件下でハイブリダィズする核酸プロ ーブを含む診断薬; BCAS2遺伝子の転写領域が配座する染色体領域とストリンジェ ントな条件下でハイブリダィズする核酸プローブを含む診断薬; BCAS2遺伝子とスト リンジェントな条件下でハイブリダィズする核酸プローブを含む診断薬; GenBankァ クセッションナンバー NT— 019273.18に記載された塩基配列の第 11017863番目 力も第 11031950番目までからなる塩基配列若しくはその相補的塩基配列に含まれ る連続する少なくとも 15塩基からなるポリヌクレオチド又は該ポリヌクレオチドとストリン ジェントな条件下でハイブリダィズする少なくとも 15塩基からなるポリヌクレオチドを含 む診断薬を例示できる。より具体的には、配列表の配列番号 7〜8に記載の塩基配 列のうちいずれか 1の塩基配列で表される核酸プローブを含む診断薬、配列表の配 列番号 11〜; 12に記載の塩基配列のうちいずれ力、 1の塩基配列からなるプライマー を含む診断薬を例示できる。 [0061] As a diagnostic agent capable of detecting amplification of the BCAS2 gene, for example, BCAS2 residue Diagnostic agent containing a nucleic acid probe that hybridizes under stringent conditions with the chromosomal region to which the gene is conformed; Nucleic acid probe that hybridizes under stringent conditions with the chromosomal region to which the transcription region of the BCAS2 gene is conformed Diagnostic agent; Diagnostic agent comprising a nucleic acid probe that hybridizes with the BCAS2 gene under stringent conditions; GenBank accession number NT-0119273.18 Examples thereof include a diagnostic agent comprising a polynucleotide comprising at least 15 consecutive bases contained in the complementary base sequence or a polynucleotide comprising at least 15 bases that hybridizes with the polynucleotide under stringent conditions. More specifically, a diagnostic agent containing a nucleic acid probe represented by any one of the nucleotide sequences set forth in SEQ ID NOs: 7 to 8 in the sequence listing, SEQ ID NOS: 11 to 12 in the sequence listing A diagnostic agent containing a primer consisting of one base sequence can be exemplified.
[0062] DENND2C遺伝子の増幅を検出することができる診断薬として、例えば、 DENN D2C遺伝子が配座する染色体領域とストリンジェントな条件下でハイブリダィズする 核酸プローブを含む診断薬; DENND2C遺伝子の転写領域が配座する染色体領 域とストリンジェントな条件下でハイブリダィズする核酸プローブを含む診断薬; DEN ND2C遺伝子とストリンジェントな条件下でハイブリダィズする核酸プローブを含む診 断薬; GenBankァクセッションナンバー NT— 019273.18に記載された塩基配列の 第 11033155番目から第 11120417番目までからなる塩基配列若しくはその相補 的塩基配列に含まれる連続する少なくとも 15塩基からなるポリヌクレオチド又は該ポ リヌクレオチドとストリンジヱントな条件下でハイブリダィズする少なくとも 15塩基からな るポリヌクレオチドを含む診断薬を例示することができる。より具体的には、配列表の 配列番号 7〜8に記載の塩基配列のうちいずれか 1の塩基配列で表される核酸プロ ーブを含む診断薬、配列表の配列番号 13〜; 14に記載の塩基配列のうちいずれか 1 の塩基配列からなるプライマーを含む診断薬を例示できる。  [0062] As a diagnostic agent capable of detecting the amplification of the DENND2C gene, for example, a diagnostic agent containing a nucleic acid probe that hybridizes under stringent conditions with a chromosomal region in which the DENN D2C gene conforms; a transcription region of the DENND2C gene; Diagnostic agent containing nucleic acid probe that hybridizes under stringent conditions with the chromosomal region to which it conforms; Diagnostic agent containing nucleic acid probe that hybridizes with DEN ND2C gene under stringent conditions; GenBank Accession Number NT— 019273.18 Or a polynucleotide comprising at least 15 contiguous bases contained in the base sequence consisting of the 11033155th to the 11120417th base sequence or the complementary base sequence thereof, or hybridizing with the polynucleotide under stringent conditions Polynuclear consisting of at least 15 bases It can be exemplified diagnostic agent comprising a plastid. More specifically, a diagnostic agent comprising a nucleic acid probe represented by any one of the nucleotide sequences set forth in SEQ ID NOs: 7 to 8 in the sequence listing, SEQ ID NOS: 13 to 14 in the sequence listing Examples thereof include a diagnostic agent containing a primer consisting of any one of the described base sequences.
[0063] また、本発明に係るアンプリコンの増幅を検出することができる診断薬として、本発 明に係るアンプリコンとストリンジェントな条件下でハイブリダィズする核酸プローブを 含む診断薬を例示できる。このような診断薬として、例えば、 GenBankァクセッション ナンバー ΝΤ_019273· 18に記載された塩基酉己歹 IJの第 10597582番目力、ら第 111 21868番目までからなる塩基配列若しくはその相補的塩基配列に含まれる連続する 少なくとも 15塩基からなるポリヌクレオチド又は該ポリヌクレオチドとストリンジェントな 条件下でハイブリダィズする少なくとも 15塩基からなるポリヌクレオチドを含む診断薬 を好ましく例示できる。より具体的には、配列表の配列番号;!〜 4に記載の塩基配列 のうちいずれか 1の塩基配列で表される核酸プローブを含む診断薬を好ましく例示 できる。 [0063] Further, examples of the diagnostic agent capable of detecting amplification of the amplicon according to the present invention include diagnostic agents including a nucleic acid probe that hybridizes with the amplicon according to the present invention under stringent conditions. Examples of such diagnostic agents include GenBank succession A base sequence consisting of at least 15 bases included in the base sequence consisting of up to the 10597582th power of IJ, No. 111 to 21868, or its complementary base sequence described in the number ΝΤ_019273 · 18 A diagnostic agent containing a polynucleotide comprising at least 15 bases that hybridizes with a nucleotide under stringent conditions can be preferably exemplified. More specifically, a diagnostic agent containing a nucleic acid probe represented by any one of the base sequences described in SEQ ID NOs:! To 4 in the sequence listing can be preferably exemplified.
[0064] 本発明に係る診断薬に含まれるポリヌクレオチド (核酸プローブ又はプライマーであ る場合も含む)の長さは、少なくとも 15塩基以上であることが好ましい(Molecular Clo nlng: A laboratory Manual, 2nd Ed., し old Spring Harbor Laboratory, し old Spn ng Harbor. N.,1989 ;モレキュラークローニング第 2版と称する場合がある)。例えば、 サザンハイブリダィゼーシヨンやノーザンハイブリダィゼーシヨンにより本発明に係る 診断用マーカーを特異的に検出する場合には、前記ポリヌクレオチドの長さは 19〜 40bp程度でもよい(モレキュラークローニング第 2版)。また、例えば、 CGH (Compa rative genomic hybridization)法により本発明に係る診断用マーカーを検出する 場合には、前記ポリヌクレオチドの長さとして 300〜3000bp程度を好ましく例示でき る(臨床 FISHプロトコ一ルー目で見る染色体 ·遺伝子診断法一第 1版、稲澤譲治ら、 1997年;臨床 FISHプロトコールと称する場合がある)。  [0064] The length of the polynucleotide (including a nucleic acid probe or primer) contained in the diagnostic agent according to the present invention is preferably at least 15 bases (Molecular Cloning: A laboratory Manual, 2nd). Ed., Old Spring Harbor Laboratory, old Spring Harbor. N., 1989; sometimes referred to as Molecular Cloning 2nd edition). For example, when the diagnostic marker according to the present invention is specifically detected by Southern hybridization or Northern hybridization, the length of the polynucleotide may be about 19 to 40 bp (Molecular Cloning No. 1). 2 edition). For example, when the diagnostic marker according to the present invention is detected by the CGH (Comparative genomic hybridization) method, the length of the polynucleotide can be preferably exemplified by about 300 to 3000 bp (the first clinical FISH protocol). Chromosome · Gene diagnosis method 1st edition, Joji Inazawa et al., 1997; sometimes referred to as clinical FISH protocol).
[0065] 本発明に係るアンプリコンの増幅を検出することができる診断薬を用いた本発明に 係るアンプリコンの増幅の検出は、例えば、 FISH法、サザンブロット法、 CGHアレイ 法など慣用の方法により実施可能である。例えば、 CGHアレイ法を用いる場合、まず 被験試料由来の DNAと対照となる正常組織由来 DNAをニックトランスレーションで それぞれ FITC (緑色)とテキサスレッド(赤色)でラベリングし、双方の標識 DNAの等 量からなる混合溶液を調製した上、本発明に係る診断薬がラベルされて!/、るスライド グラスに本溶液を加えて競合的なハイブリダィゼーシヨンを実施し、蛍光を検出する ことより実施すること力 Sできる。本発明に係るアンプリコンが増幅している場合には、該 当箇所において、被験試料由来の DNAを標識した FITC由来の緑色が観察される [0066] 本明細書において「ストリンジェントな条件下」とは、例えば、 6 X SSC、 0. 5% SD Sおよび 50%ホノレムアミドの溶 ί夜中で 42。Cにてカロ温した後、 0. 1 X SSC、 0. 5% S DSの溶液中で 68°Cにて洗浄する条件をいう。ハイブリダィゼーシヨンするポリヌクレ ォチドは、ハイブリダィゼーシヨンする他方のポリヌクレオチドの塩基配列の相補的塩 基配列を有するポリヌクレオチドでなくてもよい。ハイブリダィゼーシヨンは、例えば、 前記のモレキュラークローニング第 2版や前記の臨床 FISHプロトコールに記載され てレ、る方法に準じて行うことができる。 [0065] The detection of the amplification of the amplicon according to the present invention using the diagnostic agent capable of detecting the amplification of the amplicon according to the present invention includes, for example, conventional methods such as FISH method, Southern blot method, CGH array method, etc. Can be implemented. For example, when using the CGH array method, first, DNA from the test sample and normal tissue DNA as a control are labeled with FITC (green) and Texas red (red), respectively, by nick translation, and equal amounts of both labeled DNAs. This solution is prepared by adding the solution to a slide glass labeled with the diagnostic agent according to the present invention after the preparation of the mixed solution and performing competitive hybridization and detecting fluorescence. The power to do S. When the amplicon according to the present invention is amplified, the green color derived from FITC labeled with the DNA derived from the test sample is observed at the relevant location. [0066] As used herein, "stringent conditions" refers to, for example, 6 X SSC, 0.5% SDS and 50% honremamide in the night. This refers to the condition of washing at 68 ° C in a solution of 0.1 X SSC, 0.5% S DS after warming at C. The polynucleotide to be hybridized may not be a polynucleotide having a base sequence complementary to the base sequence of the other polynucleotide to be hybridized. Hybridization can be performed, for example, according to the method described in the Molecular Cloning Second Edition or the Clinical FISH Protocol.
[0067] 本明細書において、「あるポリヌクレオチドにより実質的に示される染色体領域」とは 、該ポリヌクレオチドとその相補的ポリヌクレオチドから形成される 2本鎖ポリヌクレオ チドに相当する染色体の部分を意味する。ここで「相当する」とは、完全に一致するこ とを含み、また、高い相同性 (例えば、 80%以上、好ましくは、 90%以上、さらに好ま しくは 93%以上、特に好ましくは 95%以上、最も好ましくは 98%以上)を有することも 含む。  [0067] In the present specification, the "chromosomal region substantially represented by a certain polynucleotide" means a portion of a chromosome corresponding to a double-stranded polynucleotide formed from the polynucleotide and its complementary polynucleotide. To do. As used herein, “corresponding” includes complete matching, and high homology (for example, 80% or more, preferably 90% or more, more preferably 93% or more, particularly preferably 95%). More preferably 98% or more).
[0068] 本発明により提供される肺小細胞癌などの小細胞癌又は乳癌の診断薬として、本 発明に係るアンプリコンに配座する遺伝子が配座する染色体領域とストリンジェントな 条件下でハイブリダィズするポリヌクレオチドを例示できる。このような遺伝子として、 例えば、 TRIM33遺伝子、 BCAS2遺伝子、 DENND2C遺伝子を例示できる。  [0068] As a diagnostic agent for small cell cancer such as small cell lung cancer or breast cancer provided by the present invention, a hybridized under stringent conditions with a chromosomal region in which the gene conformed to the amplicon according to the present invention is conformed. An example of such a polynucleotide is as follows. Examples of such genes include TRIM33 gene, BCAS2 gene, and DENND2C gene.
[0069] TRIM33遺伝子が配座する染色体領域とストリンジェントな条件下でハイブリダィズ するポリヌクレオチドとして、 TRIM33遺伝子の転写領域が配座する染色体領域とス トリンジェントな条件下でハイブリダィズするポリヌクレオチド、 TRIM33遺伝子とストリ ンノ一 NT_019273に記載された塩基酉己歹 IJの第 10847955番目力、ら第 1096138 2番目までからなる塩基配列若しくはその相補的塩基配列に含まれる連続する少なく とも 15塩基からなるポリヌクレオチド又は該ポリヌクレオチドとストリンジェントな条件下 好ましくは、 TRIM33遺伝子配列、配列表の配列番号 5に記載の塩基配歹 IJ、配列 表の配列番号 6に記載の塩基配列、配列表の配列番号 7に記載の塩基配歹 1]、配列 表の配列番号 8に記載の塩基配歹 1]、配列表の配列番号 9に記載の塩基配列及び配 列表の配列番号 10に記載の塩基配列からなる群より選ばれるいずれ力、 1の塩基配 列若しくはその相補的塩基配列において連続する少なくとも 15塩基を有するポリヌク レオチド又は該ポリヌクレオチドとストリンジェントな条件下でハイブリダィズするポリヌ クレオチド、さらに好ましくは、配列表の配列番号 5に記載の塩基配歹 IJ、配列表の配 列番号 6に記載の塩基配歹 IJ、配列表の配列番号 7に記載の塩基配歹 IJ、配列表の配 列番号 8に記載の塩基配歹 1]、配列表の配列番号 9に記載の塩基配列及び配列表の 配列番号 10に記載の塩基配列からなる群より選ばれるいずれ力、 1の塩基配列若しく はその相補的塩基配列において連続する少なくとも 15塩基を有するポリヌクレオチド ドを例示できる。より具体的には、配列表の配列番号 5〜8のうちいずれ力、 1に記載の 塩基配列で表される核酸プローブ、配列表の配列番号 9又 10に記載の塩基配列で 表されるプライマーを好ましく例示することができる。 [0069] A polynucleotide that hybridizes under stringent conditions with the chromosomal region to which the TRIM33 gene is construed as a polynucleotide that hybridizes under stringent conditions with the chromosomal region to which the TRIM33 gene is conformed, TRIM33 gene And Strintnoichi NT_019273 IJ's 10847955th power, 1096138 polynucleotide consisting of at least 15 consecutive bases contained in the base sequence consisting of up to the 2nd base sequence or its complementary base sequence, or Preferably under stringent conditions with the polynucleotide, TRIM33 gene sequence, nucleotide sequence IJ described in SEQ ID NO: 5 in the sequence listing, nucleotide sequence described in SEQ ID NO: 6 in the sequence listing, SEQ ID NO: 7 in the sequence listing Base sequence 1], the base sequence described in SEQ ID NO: 8 in the sequence listing, and the nucleotide sequence described in SEQ ID NO: 9 in the sequence listing Distribution Any force selected from the group consisting of the base sequence set forth in SEQ ID NO: 10 in the sequence table, a polynucleotide having at least 15 bases continuous in one base sequence or its complementary base sequence, or under stringent conditions with the polynucleotide More preferably, the nucleotide sequence IJ described in SEQ ID NO: 5 in the sequence listing, the base sequence IJ described in SEQ ID NO: 6 in the sequence listing, and the nucleotide sequence described in SEQ ID NO: 7 in the sequence listing.歹 IJ, the base sequence described in SEQ ID NO: 8 in the sequence listing 1], the base sequence described in SEQ ID NO: 9 in the sequence listing, and the force selected from the group consisting of the base sequence set forth in SEQ ID NO: 10 in the sequence listing And a polynucleotide having at least 15 bases that are continuous in the base sequence of 1 or its complementary base sequence. More specifically, any one of SEQ ID NOS: 5 to 8 in the sequence listing, a nucleic acid probe represented by the base sequence set forth in 1, and a primer represented by the base sequence set forth in SEQ ID NO: 9 or 10 in the sequence listing Can be preferably exemplified.
BCAS2遺伝子が配座する染色体領域とストリンジェントな条件下でハイブリダィズ するポリヌクレオチドとして、 BCAS 2遺伝子の転写領域が配座する染色体領域とスト リンジェントな条件下でハイブリダィズするポリヌクレオチド、 BCAS2遺伝子とストリン ノ ー NT_019273に記載された塩基酉己歹 IJの第 11017863番目力、ら第 11031950 番目までからなる塩基配列若しくはその相補的塩基配列に含まれる連続する少なく とも 15塩基からなるポリヌクレオチド又は該ポリヌクレオチドとストリンジェントな条件下 でハイブリダィズするポリヌクレオチドを例示できる。好ましくは、 BCAS2遺伝子配列 、配列表の配列番号 7に記載の塩基配歹 IJ、配列表の配列番号 8に記載の塩基配歹 IJ、 配列表の配列番号 11に記載の塩基配列、配列表の配列番号 12に記載の塩基配列 力、らなる群より選ばれるいずれ力、 1の塩基配列若しくはその相補的塩基配列におい て連続する少なくとも 15塩基を有するポリヌクレオチド又は該ポリヌクレオチドとストリ ンジェントな条件下でハイブリダィズするポリヌクレオチドを例示できる。より具体的に は、配列表の配列番号 7又は 8に記載の塩基配列で表される核酸プローブ、配列表 の配列番号 11又 12に記載の塩基配列で表されるプライマーを好ましく例示すること ができる。 [0071] DENND2C遺伝子が配座する染色体領域とストリンジェントな条件下でハイブリダ ィズするポリヌクレオチドとして、 DENND2C遺伝子の転写領域が配座する染色体 領域とストリンジェントな条件下でハイブリダィズするポリヌクレオチド、 DENND2C遺 伝子とストリンジェントな条件下でハイブリダィズするポリヌクレオチド、 GenBankァク セッションナンバー NT— 019273に記載された塩基配列の第 11033155番目から 第 11120417番目までからなる塩基配列若しくはその相補的塩基配列に含まれる連 続する少なくとも 15塩基からなるポリヌクレオチド又は該ポリヌクレオチドとストリンジェ ントな条件下でハイブリダィズするポリヌクレオチドを例示できる。好ましくは、 DENN D2C遺伝子配列、配列表の配列番号 7に記載の塩基配歹 IJ、配列表の配列番号 8に 記載の塩基配列、配列表の配列番号 13に記載の塩基配歹 1]、配列表の配列番号 14 に記載の塩基配列からなる群より選ばれるいずれか 1の塩基配列若しくはその相補 的塩基配列において連続する少なくとも 15塩基を有するポリヌクレオチド又は該ポリ きる。より具体的には、配列表の配列番号 7又は 8に記載の塩基配列で表される核酸 プローブ、配列表の配列番号 11又 12に記載の塩基配列で表されるプライマーを好 ましく例示すること力 Sできる。より具体的には、配列表の配列番号 7又は 8に記載の塩 基配列で表される核酸プローブ、配列表の配列番号 13又 14に記載の塩基配列で 表されるプライマーを好ましく例示することができる。 As a polynucleotide that hybridizes under stringent conditions with the chromosomal region to which the BCAS2 gene is conformed, a polynucleotide that hybridizes under stringent conditions with the chromosomal region to which the transcription region of the BCAS 2 gene is conformed, and the BCAS2 gene and string. A polynucleotide consisting of at least 15 consecutive bases contained in the base sequence consisting of the 11017863th force of IJ No. 11017863, the 1103950th, or its complementary base sequence described in NT NT019273, or the polynucleotide And polynucleotides that hybridize under stringent conditions. Preferably, the BCAS2 gene sequence, the base sequence IJ described in SEQ ID NO: 7 in the sequence listing, the base sequence IJ described in SEQ ID NO: 8 in the sequence listing, the base sequence described in SEQ ID NO: 11 in the sequence listing, The nucleotide sequence according to SEQ ID NO: 12, any force selected from the group consisting of: a polynucleotide having at least 15 bases contiguous in the nucleotide sequence of 1 or a complementary nucleotide sequence thereof, or under stringent conditions with the polynucleotide Examples of polynucleotides that hybridize with each other. More specifically, a nucleic acid probe represented by the base sequence described in SEQ ID NO: 7 or 8 in the sequence listing, and a primer represented by the base sequence described in SEQ ID NO: 11 or 12 in the sequence listing may be preferably exemplified. it can. [0071] A polynucleotide that hybridizes under stringent conditions with a chromosomal region in which the DENND2C gene is conformed, and a polynucleotide that hybridizes under stringent conditions with a chromosomal region in which the transcription region of the DENND2C gene is conformed, DENND2C A polynucleotide that hybridizes under stringent conditions with the gene, included in the nucleotide sequence consisting of the 11033155th to 11120417th of the nucleotide sequence described in GenBank Session Number NT-019273 or its complementary nucleotide sequence Examples thereof include a polynucleotide comprising at least 15 consecutive nucleotides, or a polynucleotide that hybridizes with the polynucleotide under stringent conditions. Preferably, the DENN D2C gene sequence, the base sequence IJ described in SEQ ID NO: 7 of the sequence listing, the base sequence described in SEQ ID NO: 8 of the sequence listing, the base sequence 1] described in SEQ ID NO: 13 of the sequence listing, A polynucleotide having at least 15 bases contiguous in any one base sequence selected from the group consisting of the base sequences set forth in SEQ ID NO: 14 in the sequence table or a complementary base sequence thereof, or the polynucleotide. More specifically, a nucleic acid probe represented by the base sequence described in SEQ ID NO: 7 or 8 in the sequence listing, and a primer represented by the base sequence described in SEQ ID NO: 11 or 12 in the sequence listing are preferably exemplified. That power S. More specifically, a nucleic acid probe represented by the base sequence described in SEQ ID NO: 7 or 8 in the sequence listing, and a primer represented by the base sequence described in SEQ ID NO: 13 or 14 in the sequence listing are preferably exemplified. Can do.
[0072] 本発明により提供される肺小細胞癌などの小細胞癌又は乳癌の診断薬として、例 えば、本発明に係るアンプリコンに配座する遺伝子の転写産物を検出することができ る診断薬を例示できる。このような診断薬は、本発明に係るアンプリコンに配座する遺 伝子の発現亢進を検出可能である。  [0072] As a diagnostic agent for small cell cancer such as small cell lung cancer or breast cancer provided by the present invention, for example, a diagnosis capable of detecting a transcription product of a gene conforming to the amplicon according to the present invention. Drugs can be exemplified. Such a diagnostic agent can detect increased expression of the gene conforming to the amplicon according to the present invention.
[0073] このような診断薬として、例えば、本発明に係るアンプリコンに配座する遺伝子の転 写産物とストリンジェントな条件下でハイブリダィズする少なくとも 15塩基からなるポリ ヌクレオチドを含む診断薬を例示できる。より具体的には、 TRIM33遺伝子の転写産 物とストリンジェントな条件下でハイブリダィズする少なくとも 15塩基からなるポリヌクレ ォチド、 BCAS2遺伝子の転写産物とストリンジェントな条件下でハイブリダィズする 少なくとも 15塩基からなるポリヌクレオチド、 DENND2C遺伝子の転写産物とストリン ち少なくともいずれ力、 1を含む診断薬を例示できる。 [0073] As such a diagnostic agent, for example, a diagnostic agent comprising a polynucleotide comprising at least 15 bases that hybridizes under stringent conditions with a transcription product of a gene conforming to the amplicon according to the present invention can be exemplified. . More specifically, a polynucleotide comprising at least 15 bases that hybridizes with a transcription product of TRIM33 gene under stringent conditions, and a polynucleotide comprising at least 15 bases that hybridizes with a transcription product of BCAS2 gene under stringent conditions. DENND2C gene transcript and string Or at least one of these forces can be exemplified.
[0074] 本発明に係る診断薬に含まれるポリヌクレオチド (核酸プローブ又はプライマーを含 む)は、本発明に係るアンプリコンの塩基配列、本発明に係るアンプリコンに配座する 遺伝子の塩基配列等に基づいて設計し、慣用の方法、例えば、化学合成法により製 造することが可能である。  [0074] The polynucleotide (including the nucleic acid probe or primer) contained in the diagnostic agent according to the present invention includes the base sequence of the amplicon according to the present invention, the base sequence of the gene conformed to the amplicon according to the present invention, etc. And can be produced by a conventional method, for example, a chemical synthesis method.
[0075] 本発明に係る診断薬として、例えば、本発明に係るアンプリコンに配座する遺伝子 の遺伝子産物を検出することができる診断薬を例示できる。本発明に係るアンプリコ ンに配座する遺伝子の遺伝子産物を検出することができる診断薬として、本発明に 係るアンプリコンに配座する遺伝子の遺伝子産物を認識する、好ましくは特異的に認 識する抗体を含む診断薬を例示できる。本発明に係るアンプリコンに配座する遺伝 子の遺伝子産物として、 TRIM33タンパク質、 BCAS2タンパク質又は DENND2C タンパク質を例示できる。具体的には、 TRIM33タンパク質、好ましくは、配列表の配 列番号 21に記載のアミノ酸配列で表されるポリペプチドを認識する抗体を含む診断 薬を好ましく例示できる。ここでの抗体は、ポリクローナル抗体又はモノクローナル抗 体のいずれでもよぐまた、 2種類以上の生物種由来の抗体を組み合わせたキメラ抗 体でもよい。  [0075] As the diagnostic agent according to the present invention, for example, a diagnostic agent capable of detecting the gene product of the gene conforming to the amplicon according to the present invention can be exemplified. As a diagnostic agent capable of detecting the gene product of the gene conforming to the amplicon according to the present invention, the gene product of the gene conforming to the amplicon according to the present invention is recognized, preferably specifically recognized. Diagnostic agents including antibodies can be exemplified. Examples of the gene product of the gene conforming to the amplicon according to the present invention include TRIM33 protein, BCAS2 protein or DENND2C protein. Specifically, a diagnostic agent containing an antibody that recognizes the TRIM33 protein, preferably a polypeptide represented by the amino acid sequence set forth in SEQ ID NO: 21 in the Sequence Listing can be exemplified. The antibody herein may be either a polyclonal antibody or a monoclonal antibody, or may be a chimeric antibody in which antibodies derived from two or more species are combined.
[0076] 抗体は、本遺伝子産物またはその断片を抗原として用いて作製できる。抗原は、少 なくとも 8個、好ましくは少なくとも 10個、より好ましくは少なくとも 12個、さらに好ましく は 15個以上のアミノ酸で構成される。本遺伝子産物に特異的な抗体を作成するため には、本遺伝子産物に固有なアミノ酸配列で表される領域を抗原として用いることが 好ましい。抗体の産生には、自体公知の抗体作製法を利用できる。上記抗原を自体 公知の方法により動物に投与して免疫誘導を行い、免疫誘導された動物の血清から 自体公知の抗体回収法により抗体を回収するか、あるいは、抗体産生細胞を回収し て自体公知の永久増殖性細胞への形質転換手段を導入することにより生産できる。  [0076] An antibody can be produced using the gene product or a fragment thereof as an antigen. The antigen is composed of at least 8, preferably at least 10, more preferably at least 12, even more preferably 15 or more amino acids. In order to produce an antibody specific to the gene product, it is preferable to use a region represented by an amino acid sequence unique to the gene product as an antigen. For the production of antibodies, per se known antibody production methods can be used. The antigen is administered to the animal by a method known per se to induce immunity, and the antibody is recovered from the serum of the immunized animal by a known antibody recovery method, or the antibody-producing cells are recovered and known per se Can be produced by introducing means for transformation into permanent proliferating cells.
[0077] 本発明に係る診断薬に含まれるポリヌクレオチド (核酸プローブ、プライマーである 場合を含む)又は抗体は、本発明に係るアンプリコンの増幅及び/又は該アンプリコ ンに配座する遺伝子の発現亢進を検出する機能が阻害されない限りにおいて、該ポ リヌクレオチド、該核酸プローブ、該プライマー又は該抗体等を、蛍光標識又は放射 性同位元素などの適宜の標識により標識化することが可能である。このような標識手 段は当業者に周知かつ慣用されている。 [0077] The polynucleotide (including the case of being a nucleic acid probe or primer) or antibody contained in the diagnostic agent according to the present invention is the amplification of the amplicon according to the present invention and / or the expression of the gene conforming to the amplicon. As long as the function of detecting enhancement is not inhibited, The renucleotide, the nucleic acid probe, the primer or the antibody can be labeled with an appropriate label such as a fluorescent label or a radioisotope. Such labeling means are well known and commonly used by those skilled in the art.
[0078] 本発明により提供される肺小細胞癌などの小細胞癌又は乳癌の診断方法は、ヒト 力、ら分離された生体試料において、本発明に係るアンプリコンの増幅及び/又は該 アンプリコンに配座する遺伝子の発現亢進を検出する工程を含むことを特徴としてい る。好ましくは、上記に説明したポリヌクレオチド、核酸プローブ、プライマーを用いて 本発明に係るアンプリコンの増幅を検出する工程を含み、又は、上記に説明した抗 体を用いて該アンプリコンに配座する遺伝子の発現亢進を検出する工程を含む。上 記の 2つの工程を組合わせて、本発明に係るアンプリコンの増幅及び該アンプリコン に配座する遺伝子の発現亢進を検出することも好ましい。より具体的には、 TRIM33 遺伝子の増幅及び/又は TRIM33遺伝子の発現亢進を検出する工程を含む上記 診断方法を例示できる。  [0078] The method for diagnosing small cell cancer such as small cell lung cancer or breast cancer provided by the present invention comprises amplifying the amplicon according to the present invention and / or the amplicon in a biological sample isolated from human force. It includes a step of detecting increased expression of a gene conforming to. Preferably, the method includes a step of detecting amplification of the amplicon according to the present invention using the polynucleotide, nucleic acid probe, or primer described above, or conforms to the amplicon using the antibody described above. A step of detecting increased expression of the gene. It is also preferable to detect the amplification of the amplicon according to the present invention and the increased expression of the gene conforming to the amplicon by combining the above two steps. More specifically, the above diagnostic method including the step of detecting amplification of TRIM33 gene and / or enhanced expression of TRIM33 gene can be exemplified.
[0079] 一般的には、健常のヒト由来の生体試料を対照として用いることにより、ヒトから分離 された生体試料にお!/、て、本発明に係るアンプリコンの増幅及び/又は該アンプリコ ンに配座する遺伝子の発現亢進を検出することができる。 TRIM33遺伝子の増幅及 び/又は TRIM33遺伝子の発現亢進を検出する工程を含む診断方法では、典型 的には、(1)ヒトから分離された生体試料に含まれる TRIM33遺伝子の量及び/又 は TRIM33遺伝子の遺伝子産物の発現量を測定する工程、及び(2)上記(1)で測 定した TRIM33遺伝子の量及び/又は TRIM33遺伝子の遺伝子産物の発現量と 健常のヒト由来の生体試料に含まれる TRIM33遺伝子の量及び/又は TRIM33遺 伝子の遺伝子産物の発現量を比較する工程を含むことが好ましい。健常のヒト由来 の生体試料に含まれる TRIM33遺伝子の量及び/又は TRIM33遺伝子の遺伝子 産物の発現量を標準値としてあらかじめ用意しておき、この標準値を用いて上記の 工程(2)を行うこともできる。  [0079] In general, by using a biological sample derived from a normal human as a control, the amplification of the amplicon according to the present invention and / or amplification of the amplicon according to the present invention is performed on a biological sample isolated from a human. It is possible to detect an increase in the expression of a gene conforming to the. In the diagnostic method including the step of detecting the amplification of TRIM33 gene and / or the increased expression of TRIM33 gene, typically, (1) the amount of TRIM33 gene and / or TRIM33 contained in a biological sample isolated from human A step of measuring the expression level of the gene product of the gene, and (2) the amount of the TRIM33 gene and / or the expression level of the gene product of the TRIM33 gene measured in (1) above and included in a healthy human-derived biological sample It is preferable to include a step of comparing the amount of the TRIM33 gene and / or the expression level of the gene product of the TRIM33 gene. Prepare in advance the amount of TRIM33 gene and / or the expression level of the gene product of TRIM33 gene contained in a healthy human-derived biological sample as a standard value, and perform the above step (2) using this standard value. You can also.
[0080] 本発明により提供されるスクリーニング方法は、肺小細胞癌などの小細胞癌細胞又 は乳癌細胞に対する増殖阻害作用を有する物質のスクリーニング方法であって、本 発明に係るアンプリコンに配座する遺伝子の増幅及び/又は発現を抑制する作用を 有する物質を選択する工程を含むことを特徴として!/、る。上記の方法により選択され た物質は、肺小細胞癌などの小細胞癌又は乳癌の治療のための医薬の有効成分と して用いることができ、また、上記の方法により選択された物質を肺小細胞癌などの 小細胞癌細胞又は乳癌細胞に対する増殖阻害剤として用いることができる。スクリー ユングの対象となる物質の種類は特に制限されず、例えば、有機低分子化合物のほ カゝ、高分子化合物、無機化合物、核酸類 (RNAiなどに用いる小分子 RNAや非天然 型塩基を含む核酸などを含む)、糖類、脂質類、又はペプチド類 (オリゴペプチド又 はポリペプチド類を含む)などの任意の物質が包含される。 [0080] The screening method provided by the present invention is a screening method for a substance having a growth inhibitory action on small cell cancer cells such as small cell lung cancer or breast cancer cells, and is conformed to the amplicon according to the present invention. Suppresses the amplification and / or expression of genes It includes the step of selecting a substance to have! The substance selected by the above method can be used as an active ingredient of a medicament for the treatment of small cell cancer such as small cell lung cancer or breast cancer, and the substance selected by the above method can be used as a pulmonary substance. It can be used as a growth inhibitor for small cell cancer cells such as small cell cancer or breast cancer cells. The types of substances subject to screening are not particularly limited, and include, for example, organic low molecular weight compounds, high molecular weight compounds, inorganic compounds, nucleic acids (including small molecule RNA and non-natural bases used for RNAi, etc. Any substance is included, including nucleic acids and the like, sugars, lipids, or peptides (including oligopeptides or polypeptides).
[0081] 被験物質による TRIM33遺伝子の増幅及び/又は発現に対する抑制作用を検出 する工程を含むスクリーニング方法では、典型的には、(1)肺小細胞癌などの小細胞 癌細胞又は乳癌細胞における TRIM33遺伝子の複製数及び/又は TRIM33遺伝 子の遺伝子産物の発現量を被験物質の存在下又は非存在下で測定する工程を含 むことが好ましい。肺小細胞癌などの小細胞癌細胞又は乳癌細胞としては、患者を 含むヒトから分離 '採取した生体試料 (例えば手術により分離した癌細胞を含む組織 片など)から分離された肺小細胞癌などの小細胞癌細胞又は乳癌細胞を用いてもよ いが、例えば、安定に TRIM33タンパク質を発現する肺小細胞癌などの小細胞癌細 胞又は乳癌細胞を樹立細胞株として調製して用いてもよい。あるいは、すでに樹立さ れた当業者に入手可能な肺小細胞癌などの小細胞癌細胞株 (例えば、 LU- 140) 又は乳癌細胞株(MCF7)を用いてもよ!/、。 TRIM33遺伝子の遺伝子産物を測定す るかわりに TRIM33遺伝子の転写産物を測定することも可能である。  [0081] In a screening method including the step of detecting an inhibitory effect on the amplification and / or expression of the TRIM33 gene by a test substance, typically, (1) a small cell, such as small cell lung cancer, TRIM33 in a cancer cell or a breast cancer cell It is preferable to include a step of measuring the number of gene copies and / or the expression level of the gene product of the TRIM33 gene in the presence or absence of the test substance. Small cell carcinoma cells such as small cell lung cancer or breast cancer cells are isolated from humans including patients. 'Small cell lung cancer isolated from a collected biological sample (for example, a tissue fragment containing cancer cells separated by surgery) Small cell cancer cells or breast cancer cells may be used, but for example, small cell cancer cells or breast cancer cells such as small cell lung cancer that stably express the TRIM33 protein may be prepared and used as established cell lines. Good. Alternatively, a small cell carcinoma cell line (for example, LU-140) or a breast cancer cell line (MCF7) that has already been established and is available to those skilled in the art may be used! Instead of measuring the TRIM33 gene product, it is also possible to measure the TRIM33 gene transcript.
[0082] TRIM33遺伝子の複製数は、上記の核酸プローブやプライマーを用いた慣用の 方法、例えば、ノーザンブロット法や CGH法により測定可能である。 TRIM33遺伝子 の遺伝子産物の発現量は、例えば、ウェスタンプロット法などにより検出可能である。  [0082] The number of copies of the TRIM33 gene can be measured by a conventional method using the above-described nucleic acid probe or primer, for example, the Northern blot method or the CGH method. The expression level of the gene product of TRIM33 gene can be detected, for example, by Western plotting.
[0083] 本発明により提供される医薬は、肺小細胞癌などの小細胞癌又は乳癌の治療のた めの医薬であって、本発明に係るアンプリコンに配座する遺伝子の増幅及び/又は 該遺伝子の発現を抑制する作用を有する物質を有効成分として含むことを特徴とし て!/、る。 TRIM33遺伝子の増幅及び/又は該遺伝子の発現を抑制する作用を有す る物質を有効成分として含む上記医薬を好ましく例示できる。あるいは、例えば、上 記のスクリーニング方法によりスクリーニングされた物質を有効成分として含む医薬で あってもよい。本発明の医薬はヒトを含む哺乳類動物に投与することができる。また、 本発明の医薬の投与形態は特に限定されず、経口的又は非経口的に投与すること ができる。本発明の医薬は、有効成分である物質であってもよいが、有効成分である 物質とともに薬理学的及び製剤学的に許容される製剤用添加物とを含む医薬組成 物を調製して投与することが望ましレ、。 [0083] The medicament provided by the present invention is a medicament for the treatment of small cell cancer such as small cell lung cancer or breast cancer, and amplification and / or amplification of a gene conforming to the amplicon according to the present invention. It contains a substance having an action of suppressing the expression of the gene as an active ingredient. Preferable examples of the medicament include a substance having an action of amplifying the TRIM33 gene and / or suppressing the expression of the gene as an active ingredient. Or, for example, on It may be a medicine containing a substance screened by the screening method described above as an active ingredient. The medicament of the present invention can be administered to mammals including humans. The administration form of the medicament of the present invention is not particularly limited and can be administered orally or parenterally. The medicament of the present invention may be a substance that is an active ingredient, but a pharmaceutical composition containing a substance that is an active ingredient and a pharmacologically and pharmaceutically acceptable additive for pharmaceutical preparation is prepared and administered. I hope to do it.
[0084] 例えば、タンパク質を有効成分として含む本発明の医薬は、通常のタンパク質製剤 の調製方法に従って製剤化することができ、ポリヌクレオチドを有効成分として含む 本発明の医薬も、当業界で利用可能な手段で製剤化して用いることができる。核酸 を含む本発明の医薬は、例えば、有効成分である核酸を含む組換えベクターの形態 で用いることもできる。上記の組換えベクターには、有効成分である該核酸から生体 内で効率的に遺伝子産物が発現されるように、遺伝子発現のために必要な、あるい は遺伝子発現を促進する各種の配列を適宜の順番で組み込んでお!/、てもよ!/、。本 発明の医薬がアンチセンスポリヌクレオチドを含む場合、全長も特に制限されることは ない。アンチセンスポリヌクレオチドは、例えば、相補的結合が可能になるように 10塩 基以上、好ましくは 15塩基以上のオリゴヌクレオチドであってもよい。また、本発明の 医薬の有効成分として抗体、好ましくはモノクローナル抗体を用いる場合には、抗体 は通常の方法で製造することができ、本発明に係るアンプリコンに配座する遺伝子の 遺伝子産物、好ましくは、 TRIM33タンパク質に特異的に結合可能なモノクローナル 抗体も当業者の汎用の方法で製造することができる。  [0084] For example, the medicament of the present invention containing a protein as an active ingredient can be formulated according to a conventional method for preparing a protein preparation, and the medicament of the present invention containing a polynucleotide as an active ingredient can also be used in the art. It can be formulated and used by various means. The medicament of the present invention containing a nucleic acid can also be used, for example, in the form of a recombinant vector containing a nucleic acid which is an active ingredient. The above recombinant vector contains various sequences necessary for gene expression or promoting gene expression so that the gene product is efficiently expressed in vivo from the nucleic acid which is an active ingredient. Incorporate them in the proper order! When the medicament of the present invention contains an antisense polynucleotide, the total length is not particularly limited. The antisense polynucleotide may be, for example, an oligonucleotide having 10 bases or more, preferably 15 bases or more, so that complementary binding is possible. In addition, when an antibody, preferably a monoclonal antibody, is used as the active ingredient of the medicament of the present invention, the antibody can be produced by a usual method, and is preferably a gene product of a gene conforming to the amplicon according to the present invention, preferably Monoclonal antibodies capable of specifically binding to the TRIM33 protein can also be produced by a general method of those skilled in the art.
[0085] 肺小細胞癌などの小細胞癌細胞又は乳癌細胞に対する細胞増殖阻害剤あるいは 本発明の医薬の有効成分として好ましいポリヌクレオチドとして、例えば、以下の(1) 又は(2)の 2重鎖ポリヌクレオチド:(1)配列表の配列番号 17に記載の塩基配列で表 されるポリヌクレオチドと配列表の配列番号 18に記載の塩基配列で表されるポリヌク レオチドとからなる 2重鎖ポリヌクレオチド、及び(2)配列表の配列番号 19に記載の 塩基配列で表されるポリヌクレオチドと配列表の配列番号 20に記載の塩基配列で表 されるポリヌクレオチドとからなる 2重鎖ポリヌクレオチドを例示することができる力 S、こ れらに限定されることはない。 [0086] 薬理学的及び製剤学的に許容しうる製剤用添加物としては、例えば、賦形剤、崩 壊剤ないし崩壊補助剤、結合剤、滑沢剤、コーティング剤、色素、希釈剤、基剤、溶 解剤ないし溶解補助剤、等張化剤、 pH調節剤、安定化剤、噴射剤、及び粘着剤等 を用いること力 Sできる。経口投与に適する医薬組成物の例としては、例えば、錠剤、 カプセル剤、散剤、細粒剤、顆粒剤、液剤、シロップ剤等を挙げることができる。非経 口投与に適する医薬組成物としては、例えば、注射剤、点滴剤、座剤、吸入剤、経 皮吸収剤、点眼剤、点耳剤、軟膏剤、クリーム剤、又は貼付剤等を挙げることができ る。本発明の医薬の投与量は特に限定されず、有効成分である物質の種類、治療又 は予防の目的、患者の年齢や症状、投与経路などの種々の条件に応じて適宜の投 与量を選択することが可能である力 一般的には、成人 1日あたり 0. OOlmg〜; 100 Omg程度の投与量範囲から選択することができる。 [0085] As a cell growth inhibitor for small cell carcinoma cells such as small cell lung cancer or breast cancer cells, or a preferred polynucleotide as an active ingredient of the medicament of the present invention, for example, the following (1) or (2) duplex Polynucleotide: (1) a double-stranded polynucleotide comprising a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 17 of the Sequence Listing and a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 18 of the Sequence Listing; And (2) a double-stranded polynucleotide comprising a polynucleotide represented by the base sequence set forth in SEQ ID NO: 19 of the sequence listing and a polynucleotide represented by the base sequence set forth in SEQ ID NO: 20 of the sequence listing The force S that can be, is not limited to these. [0086] Examples of pharmaceutical additives that are pharmacologically and pharmaceutically acceptable include excipients, disintegrating agents or disintegrating aids, binders, lubricants, coating agents, dyes, diluents, The ability to use bases, solubilizers or solubilizers, isotonic agents, pH adjusters, stabilizers, propellants, adhesives, etc. Examples of pharmaceutical compositions suitable for oral administration include, for example, tablets, capsules, powders, fine granules, granules, liquids, syrups and the like. Examples of pharmaceutical compositions suitable for parenteral administration include injections, drops, suppositories, inhalants, transdermal absorption agents, eye drops, ear drops, ointments, creams, or patches. be able to. The dosage of the medicament of the present invention is not particularly limited, and an appropriate dosage may be selected according to various conditions such as the type of substance as an active ingredient, the purpose of treatment or prevention, the age and symptoms of the patient, the route of administration and the like. The power that can be selected In general, an adult can be selected from a dose range of about 0.001 mg per day; about 100 Omg.
実施例  Example
[0087] 以下、実施例により本発明をさらに具体的に説明する力 本発明の範囲は下記の 実施例に限定されることはない。  [0087] Hereinafter, the present invention will be described more specifically by way of examples. The scope of the present invention is not limited to the following examples.
例 1  Example 1
(A)材料と方法  (A) Materials and methods
(a)細胞株及び原発癌  (a) Cell lines and primary cancer
合計 22種の SCLC細胞株及び 1種の MCF7を使用し、細胞を 10%のゥシ胎児血清及 び 100単位/ mlペニシリン/ 100 g/mlストレプトマイシンを添加した適切な培地 (RPMI -1640又はダルベッコ改変イーグル培地)中で維持した。 10人の SCLC患者の手術中 に分離された原発癌試料、及び 141人の乳癌患者から手術中に分離された原発癌 試料を入手した。これらの患者はいずれも術前放射線照射、化学療法、又は免疫療 法を受けていなかった。  A total of 22 SCLC cell lines and 1 MCF7 were used, and the cells were cultured in an appropriate medium (RPMI-1640 or Dulbecco with 10% urine fetal serum and 100 units / ml penicillin / 100 g / ml streptomycin. Maintained in modified Eagle medium). Primary cancer samples isolated during surgery of 10 SCLC patients and primary cancer samples isolated during surgery from 141 breast cancer patients were obtained. None of these patients received preoperative radiation, chemotherapy, or immunotherapy.
[0088] (b)CGHアレイ分析 [0088] (b) CGH array analysis
癌の発生や進行に重要であろうと考えられる遺伝子や STS(sequence_tagged site) マーカーを持つ 800個の BAC/PACクローンを用いて CGHアレイ(MCG Cancer Array -800)を調製し (Cancer Sci., 95, pp.559-563, 2004)、文献記載の方法に従ってハイ ブリダィゼーシヨンを fiつた (Cancer Sci., 96, pp.100-110, 2005; Cancer Sci., 96, pp. 676-683, 2005)。すなわち、各 GBM細胞株からの被検 DNA及び健常男性ボランティ ァから得た対照 DNAをそれぞれ Cy3_dCTP又は Cy5_dCTPでラベルし、 Cot-1 DNA の存在下でエタノールにより沈殿させ、ハイブリダィゼーシヨン'ミックス (50%ホルムァ ミド、 10%硫酸デキストラン、 2 X生理食塩-クェン酸緩衝液 [SSC]、 4%硫酸ドデシルナ トリウム [SDS]、 pH 7)で再溶解して 75°Cで 10分間変性させた。 37°Cで 10分間プレイン キュベーシヨンした後、アレイスライドに各反応液をそれぞれ塗布し、 42°Cで 48〜72 時間インキュベートした。 A CGH array (MCG Cancer Array -800) was prepared using 800 BAC / PAC clones with genes and STS (sequence_tagged site) markers that may be important for cancer development and progression (Cancer Sci., 95 , Pp.559-563, 2004), and hybridization was performed according to the method described in the literature (Cancer Sci., 96, pp.100-110, 2005; Cancer Sci., 96, pp. 676-683, 2005). Specifically, the test DNA from each GBM cell line and the control DNA obtained from healthy male volunteers were labeled with Cy3_dCTP or Cy5_dCTP, respectively, precipitated with ethanol in the presence of Cot-1 DNA, and then the hybridization mixture. (50% formamide, 10% dextran sulfate, 2X physiological saline-citrate buffer [SSC], 4% dodecyl sodium sulfate [SDS], pH 7) and redissolved at 75 ° C for 10 minutes . After pre-incubation at 37 ° C for 10 minutes, each reaction solution was applied to the array slide and incubated at 42 ° C for 48 to 72 hours.
[0089] ハイブリダィゼーシヨンの後、スライドを 50%ホルムアミド溶液で 1回、 50°Cの 2 X SSC( pH 7.0)で 15分間、 2 X SSCで 1回、 50°Cの 0.1% SDSで 15分間、及び 0.1% Nonidet P-4 0(pH 8.0)を含む 0.1 mol/Lのリン酸ナトリウムバッファ一中で室温下に 1回 (15分間)洗 浄し、 GenePix 4000B (Axon Instruments)でスキャンした。得られたイメージを GenePi X Pro 4.1イメージングソフトウェアで分析した。アレイ全体の Log2比の 2/3の平均値が 0になるように蛍光比を標準化した。ゼロから有意 (〉2 SD)に逸脱した平均比は異常 ィ直とみなした。  [0089] After hybridization, slides were once in 50% formamide solution, 15 minutes at 50 ° C 2X SSC (pH 7.0), once in 2X SSC, 0.1% SDS at 50 ° C. Wash for 15 minutes at room temperature and once in a 0.1 mol / L sodium phosphate buffer containing 0.1% Nonidet P-40 (pH 8.0) at room temperature (15 minutes), then with GenePix 4000B (Axon Instruments). Scanned. The resulting images were analyzed with GenePi X Pro 4.1 imaging software. The fluorescence ratio was standardized so that the average value of 2/3 of the Log2 ratio of the entire array was 0. An average ratio that deviated from zero to significant (> 2 SD) was considered abnormally straight.
[0090] (c)蛍光 in situハイブリダィゼーシヨン  [0090] (c) Fluorescence in situ hybridization
各細胞株から分裂中期染色体を調製した。先に報告した方法により (Clin. Cancer Res., 15, pp.4705-4713, 2003)、分析すべき領域付近に位置する BACをプローブとし て用いて FISH分析を行った。  Metaphase chromosomes were prepared from each cell line. FISH analysis was performed by using the BAC located near the region to be analyzed as a probe by the method reported previously (Clin. Cancer Res., 15, pp.4705-4713, 2003).
(d)RT-PCR  (d) RT-PCR
全 RNA力、ら Superscript First-Strand Synthesis System(Invitrogen)を使用して単 鎖 cDNAを生成させ、各遺伝子 (TRM33遺伝子、 BCAS2遺伝子、又は FLJ37099遺伝 子)の特異的プライマーを用いて増幅させた。 cDNA合成の効率を評価するためにグ リセルアルデヒド- 3-リン酸デヒドロゲナーゼ遺伝子 (GAPDH)を同時に増幅した。  Single-stranded cDNA was generated using total RNA strength, Superscript First-Strand Synthesis System (Invitrogen), and amplified using specific primers for each gene (TRM33 gene, BCAS2 gene, or FLJ37099 gene). To evaluate the efficiency of cDNA synthesis, the glyceraldehyde-3-phosphate dehydrogenase gene (GAPDH) was amplified simultaneously.
[0091] (e)抗 TRIM33抗体の製造 [0091] (e) Production of anti-TRIM33 antibody
キーホールリンペットへモシ二アン (KLH)への結合の便宜のためにァミノ末端システ インを用いて下記ペプチド(GenBankァクセッション番号: NP— 056990に記載のヒト T RIM33アミノ酸配列第 1114-1127番目力、らなるペプチド RKRLKSDERPVHIK)を合成し た。このペプチドをゥサギに注射により 2回投与し、採血して抗血清を調製した (Opero n Biotechnologies Group)D For the convenience of binding to keyhole limpet to mossianan (KLH), the amino acid sequence 1114-1127 of human TR RIM33 described in the following peptide (GenBank accession number: NP-056990) was used using an amino terminal system. The second peptide, RKRLKSDERPVHIK) was synthesized. This peptide was administered twice to a rabbit by injection, and blood was collected to prepare an antiserum (Opero n Biotechnologies Group) D
(e)—過性トランスフエクシヨン、ウェスタンブロット、及びコロニー形成  (e) —Transient transformation, Western blot, and colony formation
Mycタグを付した TRIM33(pCMV-Tag3_TRM33)を発現するプラスミドは、 TRIM33の 全コード領域である上記 RT-PCR産物を Mycェピトープと共に真核生物発現ベクター pCMV- Tag3(Stratagene)中にインフレームでクローニングすることにより得た。コロニ 一形成アツセィのため pCMV- Tag3_TRIM33又は空ベクター (pCMV- Tag3_mock)を F uGENE6(ロッシュ)を用いて細胞にトランスフエタトした。文献記載の方法に従って(Ca ncer Sci., 94, pp.344-349, 2003)、一過性トランスフエタトした細胞における TRIM33タ ンパク質の発現をトランスフエタト後 48時間に抗 Myc抗体(Cell Signaling Technology) を用いたウェスタンブロッテイング分析により確認した。適当な濃度の G418を用いて 6 ゥエルプレート中で細胞を 2週間インキュベートし、 70%エタノールで固定してクリスタ ルバイオレットで染色した。  The plasmid that expresses Myc-tagged TRIM33 (pCMV-Tag3_TRM33) is cloned in frame into the eukaryotic expression vector pCMV-Tag3 (Stratagene) together with the above RT-PCR product, which is the entire coding region of TRIM33. Was obtained. For colony formation, pCMV-Tag3_TRIM33 or empty vector (pCMV-Tag3_mock) was transfected into cells using FuGENE6 (Roche). According to the method described in the literature (Cancer Sci., 94, pp.344-349, 2003), the expression of TRIM33 protein in transiently transfected cells was detected 48 hours after transfection. This was confirmed by Western blotting analysis using Signaling Technology). Cells were incubated for 2 weeks in 6-well plates with appropriate concentrations of G418, fixed with 70% ethanol and stained with crystal violet.
(ί)細胞増殖アツセィ (ί) Cell Growth Atsey
TRIM33を低発現する細胞中にそれぞれ pCMV-Tag3_TRM33又は pCMV-Tag3_m ockをトランスフエタトして安定な TRIM33トランスフエクタント及びコントロールを得た。 細胞増殖の測定用に 96ゥヱルプレート中に 2 X 103個の細胞を播種した。生細胞数は 水溶性テトラゾリゥム塩を用いた比色定量分析 (cell counting kit_8、同仁化学研究所 )により測定した。 Stable TRIM33 transfectants and controls were obtained by transfecting pCMV-Tag3_TRM33 or pCMV-Tag3_mock, respectively, into cells with low expression of TRIM33. 2 × 10 3 cells were seeded in 96-well plates for measurement of cell proliferation. The number of viable cells was measured by colorimetric quantitative analysis (cell counting kit_8, Dojindo Laboratories) using a water-soluble tetrazolium salt.
(g)免疫組織化学 (g) Immunohistochemistry
文献記載の方法に従って (Lab. Invest., 85, pp.257-266, 2005)、ホルマリン固定化 パラフィン包埋組織切片を用いて間接 IHC法を行った。 138人の患者から切除された 原発性乳癌試料力 調製された組織片を用い、各腫瘍の中央部から採取した組織コ ァ試料の組織マイクロアレイ (TMA)ブロックを調製した。 TMAブロックの構築のために 、それぞれ代表的な組織片から 1又は 2個の組織コアを採取し、 Tissue Microarrayer( Beecher Instruments)を用いて受容側ブロックにこれらのコアを乗せた。直径 2.0 mm のコアを使用し、受容側ブロック中でそれら力 .7〜0.8 mm離れるように配置し 138コ ァ試料を含む 3個の TMAを構築した。各種濃度のエタノールで試料のパラフィンを除 き再水和させた。 10 mMクェン酸バッファー (pH 6.0)中で 10分間マイクロ波照射する 前処理により抗原を再生させた。冷却後、内在ペルォキシダーゼを封鎖するために 3 %過酸化水素で切片を処理し、ついで抗ヒト TRM33ポリクローナル抗体又は健常ゥ サギ血清と 4°Cで終夜反応させた。切片をすすぎ、ゥサギ EnViSi0n+ペルォキシダー ゼ (Dako)とインキュベートし、 0.05%過酸化水素及び 3,3'-ジァミノべンジジンで染色し 、さらにへマトキシリンで対比染色した。 LU-140細胞株を陽性対照として用いた。免 疫染色における陰性対照として組織切片を一次抗体及び免疫ペプチドの混合物とィ ンキュペートした。 LU-140細胞では全ての癌細胞核で強く染色されて!/、た (2+)。 According to the method described in the literature (Lab. Invest., 85, pp.257-266, 2005), an indirect IHC method was performed using formalin-fixed paraffin-embedded tissue sections. Primary breast cancer sample force excised from 138 patients Using prepared tissue pieces, tissue microarray (TMA) blocks of tissue core samples taken from the center of each tumor were prepared. For the construction of the TMA block, one or two tissue cores were collected from each representative tissue piece, and these cores were placed on the receiving block using a Tissue Microarrayer (Beecher Instruments). Three TMAs containing 138 core samples were constructed using a 2.0 mm diameter core, placed 7 to 0.8 mm apart in the receiving block. The paraffin in the sample was removed with various concentrations of ethanol and rehydrated. Microwave irradiation in 10 mM citrate buffer (pH 6.0) for 10 minutes The antigen was regenerated by pretreatment. After cooling, the sections were treated with 3% hydrogen peroxide to sequester endogenous peroxidase, and then reacted with anti-human TRM33 polyclonal antibody or healthy rabbit serum at 4 ° C overnight. Sections were rinsed, incubated with Usagi EnVi S i 0 n + Peruokishida peptidase (Dako), and stained with 0.05% hydrogen peroxide and 3,3' Jiamino base Njijin were counterstained with hematoxylin to further. The LU-140 cell line was used as a positive control. Tissue sections were incubated with a mixture of primary antibody and immunopeptide as a negative control for immunostaining. LU-140 cells were strongly stained in all cancer cell nuclei! /, (2+).
[0093] (h)IHC染色分析 [0093] (h) IHC staining analysis
各患者の臨床結果を知らされて!/、な!/、2名の病理学者によりスライドの判定を行つ た。すべての悪性細胞が核染色されていない場合に腫瘍を TRM33陰性とし、 50%を 超える腫瘍細胞細胞において核染色が認めれた場合にその腫瘍を TRM33陽性とし た。陰性内部コントロールとして小リンパ球 (pl6の核染色を示さない)を使用し、下記 の基準を適用した。  After being informed of the clinical outcome of each patient! /, Na! /, Two pathologists judged the slides. A tumor was TRM33 negative when all malignant cells were not nuclear-stained, and a tumor was TRM33-positive when nuclear staining was observed in more than 50% of the tumor cell cells. Small lymphocytes (not showing pl6 nuclear staining) were used as a negative internal control and the following criteria were applied.
:免疫反応性の腫瘍細胞は検出されない  : Immunoreactive tumor cells are not detected
1 +:腫瘍細胞の 50〜 100%が弱い陽性を示す  1 +: 50-100% of tumor cells are weakly positive
2+:腫瘍細胞の 50〜 100%が強い陽性を示す  2+: 50-100% of tumor cells are strongly positive
1+又は 2+: TRIM33の発現異常  1+ or 2+: Abnormal expression of TRIM33
[0094] (i)siRNA [0094] (i) siRNA
先に報告された方法 (Cell, 121, pp.87-99, 2005)に従って siRNA処理を行なった。 Li pofectamine 2000 protocol(Invitrogen)を用ぃ飞 2本鎖 RNAオリゴマー (Sigma genosys 、 75ngん m2)を各細胞にトランスフエタトした。コントロール- siRNAとしては無関係なシ ークエンスの RNAを用いた。 SiRNA treatment was performed according to the previously reported method (Cell, 121, pp. 87-99, 2005). Using Lipofectamine 2000 protocol (Invitrogen), double-stranded RNA oligomers (Sigma genosys, 75 ng m 2 ) were transfected into each cell. As a control-siRNA, an unrelated sequence of RNA was used.
(j)BrdU分析  (j) BrdU analysis
siRNAを媒体として 24時間サイレンシングした後に 96ゥエル'プレート中に 2 X 103個 の細胞を播種した。細胞が付着した後、細胞を無血清で 48時間維持した。細胞は無 処理又は 24時間サイト力イン (TGF- /3 1、 R&D)処理のいずれかとした。 BrdU取り込 みのために細胞を 10 M BrdUと 3時間インキュベートし、 BrdU labeling and Detectio n Kit II (Roche)の指示書に従って処理した。実験は 3回繰り返した。 [0095] (B)結果 After silencing for 24 hours using siRNA as a vehicle, 2 × 10 3 cells were seeded in 96-well plates. After the cells attached, the cells were maintained in serum free for 48 hours. Cells were either untreated or treated with 24-hour cytoforce-in (TGF- / 31, R & D). Cells were incubated with 10 M BrdU for 3 hours for BrdU incorporation and processed according to the instructions of BrdU labeling and Detection Kit II (Roche). The experiment was repeated three times. [0095] (B) Results
(a)SCLC細胞株の CGHアレイ分析  (a) CGH array analysis of SCLC cell lines
22種の SCLC細胞株の全てに同一バッチの MCG Cancer Array-800スライドを用い てそれらの細胞における遺伝子複製数の変化をアツセィした。表 1は 22種全ての細 胞株の全ゲノムの複製数の増減を示す。表 2にはそれらのうちで高レベル増幅を与 えたクローンを示した。全ての細胞株においてある程度の増減が認められた。 CGHァ レイ分析により lp、 lq、 3q、 5p、 12p、 17q、 20q、 Xp、及び Xqの複製数の増加傾向、及 び 3p、 4q、 5q、 13q、 17p、及び 22qの複製数の低下傾向が示唆された。 22種の SCLC 細胞株のうち 7種ににおいて高レベルの増幅が認められた Gog2比〉 2、表 2)。それら 7種の遺伝子のうち 2種の遺伝子 MYCLl(lp34.2)及び MYC(8q24.21)の高レベルの増 幅が 2種の細胞株中でそれぞれ認められた。  All 22 SCLC cell lines were subjected to the same batch of MCG Cancer Array-800 slides to assess changes in gene replication in those cells. Table 1 shows the increase or decrease in the number of whole genome replicas of all 22 cell lines. Table 2 shows those clones that gave high level amplification. Some increase or decrease was observed in all cell lines. CGH array analysis shows that lp, lq, 3q, 5p, 12p, 17q, 20q, Xp, and Xq are increasing in number of copies, and 3p, 4q, 5q, 13q, 17p, and 22q are decreasing in number Was suggested. High levels of amplification were observed in 7 out of 22 SCLC cell lines Gog2 ratio> 2, Table 2). Of these seven genes, two genes, MYCLl (lp34.2) and MYC (8q24.21), showed high levels of amplification in the two cell lines, respectively.
[0096] [表 1]  [0096] [Table 1]
Most frequer 丄 y gained and/ or lost clones Most frequer 丄 y gained and / or lost clones
Alteration Gene Locus Number Frequency (%)  Alteration Gene Locus Number Frequency (%)
EPS 15 lp32. 3 9 41% 顧 CI lq22 11 50%  EPS 15 lp32. 3 9 41% Customer CI lq22 11 50%
ARHGEF2 lq22 9 41%  ARHGEF2 lq22 9 41%
PRCC lq23. 1 10 46%  PRCC lq23. 1 10 46%
ABCC5 3q27. 1 11 50%  ABCC5 3q27. 1 11 50%
EIF4G1 3q27. 1 10 46%  EIF4G1 3q27. 1 10 46%
CDH12 5pl4. 3 11 50%  CDH12 5pl4. 3 11 50%
PC4 5pl 3. 3 11 50%  PC4 5pl 3.3 3 50%
SKP2 5pl3. 2 11 50%  SKP2 5pl3. 2 11 50%
DAB2 5pl3. 1 9 41%  DAB2 5pl3. 1 9 41%
ZNF131 5pl2 9 41%  ZNF131 5pl2 9 41%
RAD52 12pl3. 33 10 46% 丽 T3, CDC27 17q21. 32 11 50%  RAD52 12pl3. 33 10 46% 丽 T3, CDC27 17q21. 32 11 50%
C0L1A1, ABCC3 17q21. 33 9 41%  C0L1A1, ABCC3 17q21. 33 9 41%
PPM ID 17q23. 2 9 41%  PPM ID 17q23. 2 9 41%
ITGB4 17q25. 1 9 41%  ITGB4 17q25. 1 9 41%
BIRC5 17q25. 3 10 46%  BIRC5 17q25. 3 10 46%
SHGC- 103396 17q25. 3 9 41% 賺 iiz -ミvu fcld πζ-ϋ OζAV. SHGC- 103396 17q25. 3 9 41% 賺 iiz -mi vu fcld πζ-ϋ OζAV.
επ¾επ¾
n。 n.
Figure imgf000033_0001
Figure imgf000033_0001
a画  a
CvJ  CvJ
隱 1  隱 1
Figure imgf000033_0002
Figure imgf000033_0002
譲 H  Concession H
SO 22 種の肺小細胞癌細胞中で高レベル増幅を示 SO High level amplification in 22 small cell lung cancer cells
した遺伝子 (log2 > 2. 0)  Genes (log2> 2. 0)
Locus Gene Number  Locus Gene Number
1ρ34. 2 MYCL1 , RLF 2  1ρ34.2 2 MYCL1, RLF 2
8q24. 21 MYC 2  8q24. 21 MYC 2
lpl3. 2 TRIM33 1  lpl3. 2 TRIM33 1
19ql2 CC E1 1  19ql2 CC E1 1
Xpl l. 3 PCTK1 1  Xpl l. 3 PCTK1 1
[0098] (b)FISH法による lpl3アンプリコンの決定 [0098] (b) Determination of lpl3 amplicon by FISH method
1ρ13が SCLCにおいて顕著な DNA複製増加を与える新規な領域であることから (図 1A及び Β)、 1ρ13アンプリコンのマップを作成するため、 CGHアレイ分析においてこの 領域での複製数の増加を与えた SCLC細胞株につ!/、て FISH試験を行った。この増幅 領域をカバーする 7個の FISH用 BACを選択した (図 1C)。 4種の BAC(RP11_1152I1、 R P11-1129E18、 RP_11625J15、及び RP11-254K1)は LU-140中の二重微小染色体 (dm in)上で増幅されたが、他の 3種の BACは同一の細胞株中で dminパターンを示さず、 1 pl3アンプリコンから外れた位置であることが示された (図 1D)。このようにして BAC RP 11-115211と1^11-2541 1にぉぃて 13ァンプリコン中で最小かっ共通の被影響部位 、すなわち TRIM33、 BCAS2、及び FLJ37099遺伝子を有する部位を特定した。アンプ リコンのサイズは 500kbにすぎなかった。  Since 1ρ13 is a novel region that gives a marked increase in DNA replication in SCLC (Figure 1A and Β), the CGH array analysis gave an increase in the number of copies in this region to create a map of 1ρ13 amplicons. The SCLC cell line was! / And the FISH test was performed. Seven FISH BACs covering this amplification region were selected (Fig. 1C). Four BACs (RP11_1152I1, RP11-1129E18, RP_11625J15, and RP11-254K1) were amplified on the double microchromosome (dm in) in LU-140, while the other three BACs were the same cell. The dmin pattern was not shown in the strain, indicating that it was located away from the 1 pl3 amplicon (FIG. 1D). In this way, the affected sites that were the least common among the 13 amplicons in BAC RP 11-115211 and 1 ^ 11-25411 were identified, that is, the sites having the TRIM33, BCAS2, and FLJ37099 genes. The amplicon size was only 500kb.
[0099] 図 1は SCLC細胞株中の TRIM33の増幅((A)〜(D))及び発現レベル ((E》を示した 図である。 (A) LU-140細胞の代表的な CGHアレイイメージを示す。 TRIM33の複製数 の著しい増加が明瞭な緑シグナル (矢印)として検知された。 (B) LU-140細胞中の第 1 染色体の複製数プロファイルを示す。 1ρ13·2位における TRIM33の複製数比率 (log2 比)が著しく増加した (比率 =2.4、矢印)。 (C) SCLC細胞株 LU-140の分裂中期染色体 にハイブリダィズさせた RP11-625J15(TRIM33、緑シグナル)及びコントロール RP11-3 15D10(RET、赤シグナル)による代表的な FISHイメージを示す。 TRIM33(緑)が二重微 小染色体上で増幅されること示す。 BAC RP11-315D10は同一細胞株中で 2種のシグ ナルを与えた。(D) LU-140細胞株中の 1ρ13·2部位のアンプリコンマップを示す。 FIS Η試験にプローブとして使用した 7種の BAC (縦のバー)及び 3つの遺伝子 (TRIM33、 BCAS2、及び FLJ37099)の位置を示す。グラフは 1個の細胞あたりの複製数、及び LU -140細胞由来の中期染色体の FISH試験によって決定された dminの範囲を示す。 [0099] Figure 1 shows the amplification ((A) to (D)) and expression level ((E) of TRIM33 in SCLC cell lines. (A) Representative CGH array of LU-140 cells Image shows a marked increase in TRIM33 replication as a clear green signal (arrow) (B) shows the replication profile of chromosome 1 in LU-140 cells. Replication ratio (log2 ratio) increased significantly (ratio = 2.4, arrow) (C) RP11-625J15 (TRIM33, green signal) hybridized to metaphase chromosome of SCLC cell line LU-140 and control RP11-3 A typical FISH image of 15D10 (RET, red signal) shows that TRIM33 (green) is amplified on a double microchromosome BAC RP11-315D10 shows two signals in the same cell line (D) Amplicon map of 1ρ13 · 2 site in LU-140 cell line 7 types used as probes in FIS sputum test Shows the position of BAC (vertical bar) and three genes (TRIM33, BCAS2, and FLJ37099), the number of replicates per cell and LU -Range of dmin determined by FISH test of metaphase chromosomes derived from -140 cells.
[0100] (c)SCLC細胞株中の TRIM33の過剰発現  [0100] (c) Overexpression of TRIM33 in SCLC cell line
TRIM33, BCAS2、及び FLJ37099が増幅の個々のターゲットであるか否かを判断す るために、半定量的 RT-PCRにより Lu-141及び S-1以外の全 SCLC細胞株においてこ れらの遺伝子の発現レベルを調べた。図 1Eに示すように、 TRIM33, BCAS2、及び FL J37099の発現レベルは 20種の細胞株のうちそれぞれ 17種 (85%)、 4種 (20%)、及び 4種( 20%)において正常な肺 cDNAと比較して上昇しており、これらの 3種の遺伝子のうち TR IM33が最も高頻度に過剰発現していた。図 1(E)は RT-PCR分析で測定した SCLC細 胞株及び正常肺における TRIM33の発現を示す。 1:ACC-LC_80、 2:ACC_LC_173、 3丄 u_130、 4:Lul34A、 5:Lul34B、 6:Lu_143、 7:87-5、 8:S_2、 9:ACC_LC_5、 10:ACC _LC_172、 11:ACC_LC_170、 12:PC_6、 13:SBC_5、 14:Lu_24、 15:Lu_135、 16:Lu_13 8、 17:Lu- 139、 18:Lu- 165、 19:SBC- 3、 20:Lu- 140、 21:正常肺。 20種の細胞株のうち 17種 (85.0%)において TRIM33の過剰発現が認められた。  To determine whether TRIM33, BCAS2, and FLJ37099 are individual targets for amplification, these genes were detected in all SCLC cell lines except Lu-141 and S-1 by semiquantitative RT-PCR. The expression level of was examined. As shown in Figure 1E, TRIM33, BCAS2, and FL J37099 expression levels are normal in 17 (85%), 4 (20%), and 4 (20%) of 20 cell lines, respectively. Of these three genes, TR IM33 was most frequently overexpressed compared to lung cDNA. Figure 1 (E) shows TRIM33 expression in SCLC cell lines and normal lung as measured by RT-PCR analysis. 1: ACC-LC_80, 2: ACC_LC_173, 3 丄 u_130, 4: Lul34A, 5: Lul34B, 6: Lu_143, 7: 87-5, 8: S_2, 9: ACC_LC_5, 10: ACC_LC_172, 11: ACC_LC_170, 12 : PC_6, 13: SBC_5, 14: Lu_24, 15: Lu_135, 16: Lu_13 8, 17: Lu-139, 18: Lu-165, 19: SBC-3, 20: Lu-140, 21: Normal lung. Overexpression of TRIM33 was observed in 17 out of 20 cell lines (85.0%).
[0101] (d)乳癌細胞株における TRIM33の増幅  [0101] (d) Amplification of TRIM33 in breast cancer cell lines
乳癌細胞株での 1ρ13の高頻度増幅が報告されている (Cancer Res., 60, pp.4519-4 525, 2000)。先に報告されている CGHによりこの領域において高レベル発現を与えた MCF7乳癌細胞株 (Cancer Genet. Cytogenet., 117, pp.153-158, 2000)について FIS H試験を行い、 MCF7における TRIM33の複製数を調べた。 1種の BAC(625J15)は MC F7においても相同染色領域 (HSRs)として強いシグナルを与えた (図 2A)  High frequency amplification of 1ρ13 in breast cancer cell lines has been reported (Cancer Res., 60, pp.4519-4 525, 2000). MCF7 breast cancer cell line (Cancer Genet. Cytogenet., 117, pp.153-158, 2000) that gave high level expression in this region by CGH reported previously was subjected to FIS H test, and TRIM33 replication in MCF7 I checked the number. One BAC (625J15) gave a strong signal as a homologous staining region (HSRs) in MC F7 (Fig.2A)
(e)SCLC及び乳癌細胞株における TRM33タンパク質の過剰発現  (e) Overexpression of TRM33 protein in SCLC and breast cancer cell lines
4種の SCLC細胞株 (LU-24、 LU_139、 LU_165、及び LU_140)、 2種の NSCLC細胞 株 (PC-10及び A549)、及び 1種の乳癌細胞株(MCF7)における TRIM33タンパク質の 発現レベルを比較した。図 2Bに示すように、 LU-140及び MCF7において最大量の T RIM33が認められ、 NSCLC細胞株においては最小量であった。 TRIM33タンパク質の 発現レベルはこの遺伝子の増幅レベルと一致した。  Expression levels of TRIM33 protein in four SCLC cell lines (LU-24, LU_139, LU_165, and LU_140), two NSCLC cell lines (PC-10 and A549), and one breast cancer cell line (MCF7) Compared. As shown in FIG. 2B, the maximum amount of TRIM33 was observed in LU-140 and MCF7, and the minimum amount in the NSCLC cell line. The expression level of TRIM33 protein was consistent with the amplification level of this gene.
[0102] 図 2は乳癌細胞株における TRIM33の増幅及び過剰発現を示した図である。 (A) BA C RP11-625J15(TRIM33、緑シグナル)及び RP11_315D10(RET、赤シグナル)を用い た乳癌細胞株 MCF7由来の分裂中期染色体に対する 2色 FISH試験の結果を示す。 B AC RP11-625J15は MCF7において HSRとして強いシグナルを与えた。 BAC RP11-31 5D10は同一細胞株中で 2種の信号を与えた。(B) 4種の SCLC、 2種の NSCLC、及び 1 種の乳癌細胞株における蛋白抽出物の免疫ブロッテイングを示す。 l:Lu_24、 2:Lu-l 39、 3:Lu- 165、 4:Lu- 140、 5:A549、 6:PC- 10、及び 7:MCF7。 TRIM33タンパク質の発 現レベルは、 LU-140及び MCF7における該遺伝子の増幅レベルと一致した。 [0102] Fig. 2 shows the amplification and overexpression of TRIM33 in breast cancer cell lines. (A) shows the results of a two-color FISH test for metaphase chromosomes derived from breast cancer cell line MCF7 using BA C RP11-625J15 (TRIM33, green signal) and RP11_315D10 (RET, red signal). B AC RP11-625J15 gave a strong signal as HSR in MCF7. BAC RP11-31 5D10 gave two signals in the same cell line. (B) shows immunoblotting of protein extracts in 4 types of SCLC, 2 types of NSCLC, and 1 type of breast cancer cell line. l: Lu_24, 2: Lu-l 39, 3: Lu-165, 4: Lu-140, 5: A549, 6: PC-10, and 7: MCF7. The expression level of TRIM33 protein coincided with the amplification level of the gene in LU-140 and MCF7.
[0103] (ί)原発性 SCLC及び乳癌腫瘍の中の TRM33タンパク質の過剰発現  [0103] (ί) Overexpression of TRM33 protein in primary SCLC and breast cancer tumors
原発腫瘍にお!/、て TRIM33が過剰発現して!/、るか否かを確認するために免疫組織 化学的に 10種の原発性 SCLC及び 141種の原発性乳癌を調べた。図 3A-Dに示すよ うに、非腫瘍対照と比較して 10種の SCLC腫瘍の全て (100%)において核中での TRIM 33の高発現が認められた。混合小細胞癌 (腺癌コンポーネントと複合した肺小細胞癌 )の場合には小細胞癌 (図 3C-D)の部分でのみ TRIM33が過剰発現していた。また、 原発性乳癌腫瘍においては 141種のうちの 126種 (89%)において TRIM33の核染色が 認められ、 TRM33の発現が増加していた (図 3E-H)。腺管上皮内癌においても TRIM 33の核染色が認められた (図 3G)。対照として正常な乳腺においては非常に弱い染 色のみが認められた (図 3H)。  To confirm whether TRIM33 was overexpressed in primary tumors! /, Immunohistochemically, 10 primary SCLCs and 141 primary breast cancers were examined. As shown in FIGS. 3A-D, high expression of TRIM 33 in the nucleus was observed in all 10 SCLC tumors (100%) compared to non-tumor controls. In the case of mixed small cell carcinoma (small cell lung cancer combined with adenocarcinoma component), TRIM33 was overexpressed only in the small cell carcinoma (Figure 3C-D) part. In primary breast cancer tumors, nuclear staining of TRIM33 was observed in 126 (89%) of 141 types, and the expression of TRM33 was increased (Fig. 3E-H). Nuclear staining of TRIM 33 was also observed in ductal carcinoma in situ (Fig. 3G). Only very weak staining was observed in the normal mammary gland as a control (Fig. 3H).
[0104] 図 3は肺の原発性小細胞癌及び乳癌における TRM33タンパク質の免疫組織化学 染色を示した写真である。小細胞癌における強い核染色 (A-D)及び腺癌 (D、矢の先 端)における低発現、並びに正常肺 (A及び C中の星印)における低発現を示す。侵襲 性腺管細胞癌 、 F)、侵襲性小葉癌 (H、矢の先端)、及び上皮内腺管癌 (G)における 強度の核染色、及び正常乳腺 (H、矢印)における低発現を示す。  [0104] Fig. 3 is a photograph showing immunohistochemical staining of TRM33 protein in primary small cell lung cancer and breast cancer. Intense nuclear staining (A-D) in small cell carcinoma and low expression in adenocarcinoma (D, tip of arrow) and low expression in normal lung (stars in A and C). Intense nuclear staining in invasive ductal cell carcinoma (F), invasive lobular carcinoma (H, tip of arrow), and intraepithelial ductal carcinoma (G), and low expression in normal mammary gland (H, arrow).
[0105] (g)SCLCにおける TRIM33の腫瘍形成性  [0105] (g) Tumorigenicity of TRIM33 in SCLC
SCLC細胞の細胞増殖に対する TRM33の影響を調べる目的で TRIM33の全長配列 を用いてコロニー形成アツセィを行った。図 4Bに示すように、トランスフエタト及びそれ に続く薬剤耐性コロニーの選択の 2週間後において、空ベクターを導入した細胞に 比べて TRIM33でトランスフエタトした細胞により形成された大コロニーの数が増加して V、た。この増殖促進作用を確認するため外因性 TRIM33を安定発現する SBC-3細胞 株を樹立した (SBC-3-TRM33細胞、図 4C上段)。予想のとおり、空プラスミドでトラン スフエタトした細胞に比べて SBC-3-TRM33細胞の増殖速度は非常に大き力、つた。 [0106] 図 4は癌細胞増殖に対する TRIM33発現亢進の効果を示した図である。 TRIM33の 全長配列を含み Mycタグを付した構築物 (pCMV-Tag3_TRM33)又は空ベクター (pC MV-Tag3_mock、コントローノレ)を HeLa及び SCLC細胞株 SBC-3にトランスフエタトした 。(A) 50 gの蛋白抽出物及び抗 Myc抗体を用いたウェスタンブロッツティングの結果 を示す。 pCMV-Tag3_TRIM33で一過性トランスフエタトした細胞にお!/、て Mycタグ付 き TRIM33タンパク質が発現して!/、ることを示す。(B)トランスフエタト及びそれに続く薬 剤耐性コロニー選択の 2週間後に 6ゥエルプレート中で TRIM33トランスフエタト HeLa及 び SBC-3細胞により形成されたコロニーは空ベクターでトランスフエタトした細胞により 形成されるコロニーよりも多数であった。下のグラフはコロニー形成を定量分析した結 果を示す。 2 mmを超えるコロニーの数をカウントして 3回の独立した実験の平均土 SE M (図中のバーは SEを示す)として示した。 (C) pCMV_Tag3B_TRIM33でトランスフエク トし G418で選択することにより安定に TRIM33を発現するクローンとして樹立された SB C-3細胞における TRIM33の該 SBC-3細胞に対する増殖促進作用、及びコントロール として空ベクター (empty)でトランスフエタトした SBC-3細胞の結果を示す。上段は 50 μ gの蛋白抽出物及び抗 Myc-タグ抗体を用レ、て pCMV_Tag3B-TRIM33でトランスフエ タトした 2クローン (クローン 1及び 2)をウェスタンブロット分析した結果を示す。両クロー ンとも Myc-タグ付き TRM33タンパク質を発現して!/、た。下段のグラフは安定な TRIM3 3発現が SBC-3細胞の増殖に及ぼす影響を示す。所定の日に細胞の生存を水溶性 テトラゾリゥム塩アツセィで測定した(グラフの各点は 3回の独立した試験結果の平均 値として示し、バーは SEを示す)。 To investigate the effect of TRM33 on cell proliferation of SCLC cells, a colony formation assay was performed using the full-length sequence of TRIM33. As shown in Figure 4B, the number of large colonies formed by cells tranfected with TRIM33 compared to cells transfected with the empty vector at 2 weeks after selection of the transfectant and subsequent drug-resistant colonies. Increased by V. In order to confirm this growth promoting action, an SBC-3 cell line stably expressing exogenous TRIM33 was established (SBC-3-TRM33 cell, upper part of FIG. 4C). As expected, the growth rate of SBC-3-TRM33 cells was much greater than that of cells tranfected with the empty plasmid. [0106] Fig. 4 shows the effect of enhanced TRIM33 expression on cancer cell proliferation. A construct (pCMV-Tag3_TRM33) containing the full-length sequence of TRIM33 and a Myc tag or an empty vector (pCMV-Tag3_mock, controller) was transfected into HeLa and SCLC cell line SBC-3. (A) The results of Western blotting using 50 g of protein extract and anti-Myc antibody are shown. This indicates that Myc-tagged TRIM33 protein is expressed in the cells transiently tranfected with pCMV-Tag3_TRIM33! (B) Two weeks after selection of the transpetate and subsequent drug-resistant colonies, the colonies formed by TRIM33 transfect HeLa and SBC-3 cells in the 6-well plate were transferred to cells transfected with the empty vector. There were more than colonies formed. The lower graph shows the results of quantitative analysis of colony formation. The number of colonies exceeding 2 mm was counted and expressed as the average soil SEM (bars in the figure indicate SE) of three independent experiments. (C) Growth-promoting effect of TRIM33 on SBC-3 cells in SBC-3 cells established as clones that stably express TRIM33 by transfection with pCMV_Tag3B_TRIM33 and selection with G418, and an empty vector as a control ( The results for SBC-3 cells transfected with empty) are shown. The upper row shows the results of Western blot analysis of 2 clones (clone 1 and 2) transfected with pCMV_Tag3B-TRIM33 using 50 μg of protein extract and anti-Myc-tag antibody. Both clones expressed Myc-tagged TRM33 protein! The lower graph shows the effect of stable TRIM3 3 expression on the proliferation of SBC-3 cells. Cell viability was measured on a given day with water-soluble tetrazolium salt assembly (each point on the graph is the average of three independent test results and the bar represents SE).
[0107] (h)TRIM33を標的とする siRNAによる増殖阻害  [0107] (h) Growth inhibition by siRNA targeting TRIM33
TRM33の抑制が SCLC又は乳癌細胞の増殖を抑制するか否かを試験する目的で TRIM33を高発現する SBC-5及び MCF7細胞にお!/、て TRIM33 siRNAの活性を調べ た。 TRIM33を標的とする siRNAは内因性 TRIM33タンパク質をトランスフエタトの 48〜1 20時間において顕著に減少させた力 コントロール siRNA又はトランスフエタト試薬で は減少は認められなかった(図 5A)。また、 TRIM33の発現抑制により SBC-5及び MC F7細胞の増殖が阻害された (図 5B)。これらの結果は TRIM33の過剰発現が SCLC及 び乳癌細胞の増殖促進に関与していることを示しており、 TRM33の発現抑制作用を 有する物質により SCLC及び乳癌細胞の増殖を抑制できることを示している。 To test whether suppression of TRM33 suppresses the growth of SCLC or breast cancer cells, the activity of TRIM33 siRNA was examined in SBC-5 and MCF7 cells that highly express TRIM33! The siRNA targeting TRIM33 significantly reduced the endogenous TRIM33 protein from 48 to 120 hours after transfectation. No decrease was observed with the force control siRNA or transfate reagent (FIG. 5A). In addition, suppression of TRIM33 expression inhibited the growth of SBC-5 and MC F7 cells (FIG. 5B). These results indicate that overexpression of TRIM33 is involved in promoting the growth of SCLC and breast cancer cells, It shows that the growth of SCLC and breast cancer cells can be suppressed by the substances possessed.
[0108] (i)TGF- βが惹起する細胞分裂停止に対する TRM33の抑制作用 [0108] (i) Inhibitory effect of TRM33 on TGF-β-induced cell division arrest
TRM33が Smadレスポンスを減じることにより TGF- βの抗増殖作用を抑制する可能 性を検討するため、 BrdU取り込みによりコントロール及び TRIM33_siRNA-処理 MCF7 細胞の DNA合成/細胞周期進行をアツセィした。 TRIM33の抑制により BrdU取り込み の基礎速度が阻害され、外因性の TGF- /3により惹起される増殖抑制作用が顕著に 増強された (図 5C)。この結果は、 TGF- /3が担う細胞増殖抑制作用を TRM33が減弱 することにより TRM33が SCLC及び乳癌細胞に対して増殖促進作用を発揮することを 示している。  In order to investigate the possibility that TRM33 suppresses the anti-proliferative effect of TGF-β by reducing the Smad response, control and TRIM33_siRNA-treated MCF7 cell DNA synthesis / cell cycle progression were assessed by BrdU incorporation. Inhibition of TRIM33 inhibited the basal rate of BrdU incorporation and markedly enhanced the growth-suppressing action induced by exogenous TGF-3 (Fig. 5C). This result indicates that TRM33 exerts a growth promoting effect on SCLC and breast cancer cells by attenuating the cell growth inhibitory effect of TGF- / 3 by TRM33.
[0109] 図 5は TRIM33 mRNAを標的とする siRNAによる増殖阻害作用を示した図である。 T RIM33の増幅及び過剰発現を示す MCF7細胞及び TRIM33の過剰発現を示す SBC- 5細胞をリポフエクタミン 2000を用いて TRIM33を標的とする siRNA又はコントロール siR NAで処理した。(A)ウェスタンブロッテイングによって決定された TRIM33タンパク質の 蛋白のレべノレ。 75 ngん m2のオリゴヌクレオチド (TRIM33又はコントローノレ)あるいはリ ポフエクタミン 2000のみで細胞を処理し、トランスフエタトの後 48及び 120時間に細胞 を集めた。未処理の細胞を同一条件で維持した。全細胞溶解物 50 gをポリアクリル アミドゲル電気泳動により分離し、ィムノブロッテイングで分析した。(B)SBC_5及び MC F7細胞の生存に対する siRNAオリゴヌクレオチドの影響を示した。 MTTアツセィにより トランスフエタト後の所定の時間に細胞の生存数を決定した。結果はリボフヱクタミン 単独処理(コントロール)細胞の吸光度に対するパーセントとして示し、各点は 3回の 独立した実験の平均値土 SEで示した。(C)コントロール及び TRM33_siRNA-処理 MC F7細胞に対する TGF- /3の増殖阻害作用を BrdU取り込みで測定した結果を示す。グ ラフは DNA合成が活発に行われている細胞の割合を示し、コントロール siRNAで処理 した細胞の吸光度に対するパーセントとして示した。結果は 3回の独立した実験の平 均値士 SEとして示した。 [0109] Fig. 5 shows the growth inhibitory action of siRNA targeting TRIM33 mRNA. MCF7 cells showing amplification and overexpression of TRIM33 and SBC-5 cells showing overexpression of TRIM33 were treated with siRNA targeting TRIM33 or control siRNA using Lipofectamine 2000. (A) Protein level of TRIM33 protein determined by Western blotting. Cells were treated with 75 ng m 2 oligonucleotide (TRIM33 or controller) or Lipofectamine 2000 alone, and cells were collected 48 and 120 hours after transfer. Untreated cells were maintained under the same conditions. 50 g of whole cell lysate was separated by polyacrylamide gel electrophoresis and analyzed by immunoblotting. (B) The effect of siRNA oligonucleotides on the survival of SBC_5 and MCF7 cells was shown. The number of viable cells was determined at a predetermined time after the transfer by MTT assay. The results are expressed as a percentage of the absorbance of the ribofactoramine alone treated (control) cells, and each point is expressed as the mean value of three independent experiments. (C) Control and TRM33_siRNA-treated The results of measuring the growth inhibitory action of TGF- / 3 on MC F7 cells by BrdU incorporation are shown. Graphs show the percentage of cells with active DNA synthesis and are expressed as a percentage of the absorbance of cells treated with control siRNA. The results are shown as mean valuer SE of three independent experiments.
[0110] 例 2  [0110] Example 2
(A)材料と方法  (A) Materials and methods
予後因子解析 各症例を TRIM33の染色性を指標として TRIM33 low群(-および 1+)と TRIM33 high 群(2+)に分類し、各群の全体生存期間(overall survival)および無増悪期間(progres sion-free survival)を検討した。解析は STAT VIEW softwareを用い、 aplan-Meier c urveを作製し、 Logrank testにて p値が 0.05未満を有意とした。 Prognostic factor analysis Each case was classified into TRIM33 low group (-and 1+) and TRIM33 high group (2+) using TRIM33 staining as an index, and overall survival and progression-free period (progression-free) of each group survival) was examined. For the analysis, STAT VIEW software was used to prepare an aplan-Meier curve, and a p value of less than 0.05 was determined to be significant by the Logrank test.
(B)結果  (B) Result
予後における TRIM33の発現解析  Expression analysis of TRIM33 in prognosis
TRM33の発現レベルが乳癌の予後因子であるか否かに関して、前記 (g)で染色し た 138の乳癌の症例のうち予後の明らかな 134例を対象として、予後因子解析を実施 した。図 6(A)(B)で示されるように、全体生存期間(overall survival)および無増悪期 間(progression-free survival)のいずれも、 TRIM33 high群が有意(p値: 0.0285又は 0· 011)に予後不良であった。この結果から、 TRIM33は予後予測のマーカーとして有用 であると考えられた。  Regarding whether or not the expression level of TRM33 is a prognostic factor for breast cancer, a prognostic factor analysis was performed on 134 cases with a clear prognosis among the 138 breast cancer cases stained in (g) above. As shown in Figures 6 (A) and 6 (B), the TRIM33 high group is significant (p value: 0.0285 or 0 · 011) for both overall survival and progression-free survival. ) Had a poor prognosis. From these results, TRIM33 was considered useful as a prognostic marker.
[0111] 例 3 [0111] Example 3
肺以外の小細胞癌臨床検体 9例 (食道癌 5例、副鼻腔腫瘍 2例、尿管癌 1例、大腸 癌 1例)について TRM33染色を行なった。染色及び判定は例 1に記載した方法と同 様に行なった。その結果、 9例中 7例 (食道癌 5例中 4例、副鼻腔腫瘍 2例中 2例、尿管 癌 1例中 1例、大腸癌 1例中 0例)において腫瘍部の染色性が亢進していた。図 7に染 色の結果を示す。この結果により、 TRIM33は肺癌を含む様々な臓器における小細胞 癌の診断用マーカーとして有用であることが示された。  Nine clinical specimens of small cell carcinoma other than lung (5 cases of esophageal cancer, 2 cases of sinus tumor, 1 case of ureteral cancer, 1 case of colon cancer) were subjected to TRM33 staining. Staining and determination were performed in the same manner as described in Example 1. As a result, 7 out of 9 cases (4 out of 5 cases of esophageal cancer, 2 out of 2 cases of sinus tumor, 1 out of 1 ureteral cancer, 0 out of 1 colon cancer) It was increased. Figure 7 shows the dyeing results. This result shows that TRIM33 is useful as a diagnostic marker for small cell carcinoma in various organs including lung cancer.
[0112] 配列表には以下の配列を示した。 [0112] The following sequences are shown in the Sequence Listing.
配列番号 1に記載の塩基配列: RP11-115211プローブ, +strand, NT— 019273(10389 3839, 120498679) : 10597582 - 10752202 (Length: 154621) Start: 114491420 - 114 646040 (Length: 154621)  Base sequence described in SEQ ID NO: 1: RP11-115211 probe, + strand, NT— 019273 (10389 3839, 120498679): 10597582-10752202 (Length: 154621) Start: 114491420-114 646040 (Length: 154621)
配列番号 2に記載の塩基配列: RP11-115211プローブ, -strand,  The nucleotide sequence set forth in SEQ ID NO: 2: RP11-115211 probe, -strand,
配列番号 3に記載の塩基配列: RP11-1129E18プローブ, +strand, NT— 019273(1038 93839, 120498679) : 10746859 - 10891175 (Length: 144317) Start: 114640697 - 11 4785013 (Length: 144317)  Base sequence described in SEQ ID NO: 3: RP11-1129E18 probe, + strand, NT— 019273 (1038 93839, 120498679): 10746859-10891175 (Length: 144317) Start: 114640697-11 4785013 (Length: 144317)
配列番号 4に記載の塩基配列: RP11-1129E18プローブ, -strand 配列番号 5に記載の塩基配列: RP11-625J15プローブ, +strand, NT— 019273(10389 3839, 120498679) : 10785181 - 10949822 (Length: 164642) Start: 114679019 - 114 843660 (Length: 164642) The nucleotide sequence set forth in SEQ ID NO: 4: RP11-1129E18 probe, -strand Base sequence described in SEQ ID NO: 5: RP11-625J15 probe, + strand, NT— 019273 (10389 3839, 120498679): 10785181-10949822 (Length: 164642) Start: 114679019-114 843660 (Length: 164642)
配列番号 6に記載の塩基配列: RP11-625J15プローブ, -strand Base sequence described in SEQ ID NO: 6: RP11-625J15 probe, -strand
配列番号 7に記載の塩基配列: RP11-254KIプローブ, +strand, NT— 019273(103893 839, 120498679) : 10934043 - 11121868 (Length: 187826) Start: 114827881 - 1150 15706 (Length: 187826) Base sequence described in SEQ ID NO: 7: RP11-254KI probe, + strand, NT— 019273 (103893 839, 120498679): 10934043-11121868 (Length: 187826) Start: 114827881-1150 15706 (Length: 187826)
配列番号 8に記載の塩基配列: RP11-254KIプローブ, -strand The nucleotide sequence set forth in SEQ ID NO: 8: RP11-254KI probe, -strand
配列番号 9に記載の塩基配列: TRM33遺伝子の発現解析に用いた Fプライマー 配列番号 10に記載の塩基配列: TRM33遺伝子の発現解析に用いた Rプライマー 配列番号 11に記載の塩基配列: BCAS2遺伝子の発現解析に用いた Fプライマー 配列番号 12に記載の塩基配列: BCAS2遺伝子の発現解析に用 1/、た Rプライマー 配列番号 13に記載の塩基配列: DENND2C遺伝子の発現解析に用いた Fプライマー 配列番号 14に記載の塩基配列: DENND2C遺伝子の発現解析に用いた Rプライマー 配列番号 17に記載の塩基配列: TRM33 siRNA(l)センス鎖 SEQ ID NO: 9 nucleotide sequence: F primer used for TRM33 gene expression analysis SEQ ID NO: 10 nucleotide sequence: TRM33 gene expression analysis R primer SEQ ID NO: 11 nucleotide sequence: BCAS2 gene F primer used for expression analysis Base sequence described in SEQ ID NO: 12: used for expression analysis of BCAS2 gene R primer Base sequence described in SEQ ID NO: 13 F primer used for expression analysis of DENND2C gene SEQ ID NO: Base sequence described in 14: R primer used for expression analysis of DENND2C gene Base sequence described in SEQ ID NO: 17: TRM33 siRNA (l) sense strand
配列番号 18に記載の塩基配列: TRM33 siRNA(l)アンチセンス鎖 The nucleotide sequence set forth in SEQ ID NO: 18: TRM33 siRNA (l) antisense strand
配列番号 19に記載の塩基配列: TRM33 siRNA(2)センス鎖 The nucleotide sequence set forth in SEQ ID NO: 19: TRM33 siRNA (2) sense strand
配列番号 20に記載の塩基配列: TRM33 siRNA(3)アンチセンス鎖 The nucleotide sequence set forth in SEQ ID NO: 20: TRM33 siRNA (3) antisense strand
配列番号 21に記載のアミノ酸配列: TRIM33抗体の抗原 Amino acid sequence set forth in SEQ ID NO: 21: antigen of TRIM33 antibody
配列番号 25に記載の塩基配列: GenBankァクセッション番号 NT— 019273.18に記載 の塩基配列の第 10597582番目から第 11121868番目までからなる塩基配列 産業上の利用可能性 Nucleotide sequence described in SEQ ID NO: 25: Nucleotide sequence consisting of Nos. 10597582 to 11121868 of the nucleotide sequence described in GenBank accession number NT-0192713.18 Industrial applicability
本発明により、肺小細胞癌などの小細胞癌及び乳癌を簡便かつ確実に診断できる 手段が提供される。本発明により、肺小細胞癌などの小細胞癌及び乳癌の治療のた めの医薬が提供される。  The present invention provides a means for easily and reliably diagnosing small cell cancer such as small cell lung cancer and breast cancer. According to the present invention, a medicament for the treatment of small cell cancer such as small cell lung cancer and breast cancer is provided.

Claims

請求の範囲 下記の群より選ばれる染色体領域、遺伝子又はタンパク質からなる、小細胞癌又は 乳癌の診断用マーカー; A diagnostic marker for small cell cancer or breast cancer, comprising a chromosomal region, gene or protein selected from the following group;
(1) GenBankァクセッションナンバー NT— 019273.18に記載された塩基配列の第 1059 7582番目力、ら第 11121868番目までからなる塩基配列で表されるポリヌクレオチドによ り実質的に示される染色体領域、  (1) Chromosomal region substantially represented by a polynucleotide represented by the nucleotide sequence consisting of the 1059th 7582th force of the nucleotide sequence described in GenBank Accession Number NT-0192713.18, and the 11121868th nucleotide sequence,
(2) TRIM33 (tripartite motif 33)遺伝子、  (2) TRIM33 (tripartite motif 33) gene,
(3) BCAS2 (breast carcinoma ampliiied sequence_2)退 子、  (3) BCAS2 (breast carcinoma ampliiied sequence_2)
(4) DENND2C (DENN/MADD domain containing 2C)遺伝子、  (4) DENND2C (DENN / MADD domain containing 2C) gene,
(5) TRIM33タンパク質、  (5) TRIM33 protein,
(6) BCAS2タンパク質、及び  (6) BCAS2 protein, and
(7) DENND2Cタンパク質。  (7) DENND2C protein.
TRM33遺伝子又は TRM33タンパク質からなる、小細胞癌又は乳癌の診断用マーカ 小細胞癌が肺小細胞癌である請求項 1又は 2に記載の診断用マーカー。  The diagnostic marker for small cell cancer or breast cancer comprising the TRM33 gene or TRM33 protein. The diagnostic marker according to claim 1 or 2, wherein the small cell cancer is small cell lung cancer.
小細胞癌又は乳癌の診断薬であって、ヒトを含む哺乳類動物において下記 (1)の染 色体領域の増幅及び/又は下記 (2)の遺伝子の発現亢進を検出することができる診 断薬; A diagnostic agent for small cell cancer or breast cancer that can detect the amplification of the chromosomal region of (1) below and / or the increased expression of the gene of (2) below in mammals including humans. ;
(1) GenBankァクセッションナンバー NT— 019273.18に記載された塩基配列の第 1059 7582番目力、ら第 11121868番目までからなる塩基配列で表されるポリヌクレオチドによ り実質的に示される染色体領域、  (1) Chromosomal region substantially represented by a polynucleotide represented by the nucleotide sequence consisting of the 1059th 7582th force of the nucleotide sequence described in GenBank Accession Number NT-0192713.18, and the 11121868th nucleotide sequence,
(2) TRIM33 (tripartite motif 33)退 子、 BCAS2 (breast carcinoma amplified sequence -2)遺伝子、又は DENND2C (DENN/MADD domain containing 2C)遺伝子。  (2) TRIM33 (tripartite motif 33) gene, BCAS2 (breast carcinoma amplified sequence -2) gene, or DENND2C (DENN / MADD domain containing 2C) gene.
ヒトの小細胞癌又は乳癌の診断のための請求項 4に記載の診断薬。 The diagnostic agent according to claim 4 for diagnosis of human small cell cancer or breast cancer.
小細胞癌が肺小細胞癌である請求項 4又は 5に記載の診断薬。 6. The diagnostic agent according to claim 4 or 5, wherein the small cell cancer is small cell lung cancer.
ヒトから分離された生体試料にお!/、て、下記 (1)の染色体領域の増幅及び/又は下 記 (2)の遺伝子の発現亢進を検出することができる請求項 5又は 6に記載の診断薬;The biological sample isolated from a human can detect the amplification of the chromosomal region of (1) below and / or the enhanced expression of the gene of (2) below. Diagnostic agent;
(l)GenBankァクセッションナンバー NT 019273.18に記載された塩基配列の第 1059 7582番目力、ら第 11121868番目までからなる塩基配列で表されるポリヌクレオチドによ り実質的に示される染色体領域、 (l) GenBank accession number NT 019273.18 A chromosomal region substantially represented by a polynucleotide represented by the nucleotide sequence consisting of the 7582th power, and the 11121868th,
(2)TRIM33 (tripartite motif 33ノ退 子、 BCAS2 (breast carcinoma amplified sequence -2)遺伝子、又は DENND2C (DENN/MADD domain containing 2C)遺伝子。  (2) TRIM33 (tripartite motif 33 gene), BCAS2 (breast carcinoma amplified sequence-2) gene, or DENND2C (DENN / MADD domain containing 2C) gene.
[8] GenBankァクセッションナンバー NT— 019273.18に記載された塩基配列の第 1059758 2番目から第 11121868番目までからなる塩基配列で表されるポリヌクレオチドにより実 質的に示される染色体領域とストリンジェントな条件下でハイブリダィズする核酸プロ ーブを含む、請求項 4な!/、し 7の!/、ずれか 1項に記載の診断薬。  [8] GenBank accession number NT-0109273.18 of the nucleotide sequence described in the 1059758 2nd to 11121868 nucleotide sequence and the chromosomal region substantially indicated by the polynucleotide represented by the nucleotide sequence The diagnostic agent according to claim 4, which comprises a nucleic acid probe that hybridizes under conditions.
[9] GenBankァクセッションナンバー NT— 019273.18に記載された塩基配列の第 1059758 2番目から第 11121868番目までからなる塩基配列若しくはその相補的塩基配列に含 まれる連続する少なくとも 15塩基からなるポリヌクレオチド又は該ポリヌクレオチドとス トリンジヱントな条件下でハイブリダィズする少なくとも 15塩基からなるポリヌクレオチド を含む、請求項 4な!/、し 7の!/、ずれか 1項に記載の診断薬。  [9] Polynucleotide consisting of at least 15 contiguous bases contained in the base sequence consisting of the 2nd to 11121868th base sequence of the nucleotide sequence described in GenBank accession number NT-0119273.18 or its complementary base sequence The diagnostic agent according to claim 4, wherein the diagnostic agent comprises a polynucleotide comprising at least 15 bases that hybridizes with the polynucleotide under stringent conditions.
[10] TRM33遺伝子が配座する染色体領域とストリンジェントな条件下でハイブリダィズす る核酸プローブを含む、請求項 4ないし 7のいずれか 1項に記載の診断薬。  [10] The diagnostic agent according to any one of claims 4 to 7, comprising a nucleic acid probe that hybridizes under stringent conditions with a chromosomal region to which the TRM33 gene is conformed.
[11] GenBankァクセッションナンバー NT— 019273.18に記載された塩基配列の第 1084308 4番目から第 10961466番目までからなる塩基配列若しくはその相補的塩基配列に含 まれる連続する少なくとも 15塩基からなるポリヌクレオチド又は該ポリヌクレオチドとス トリンジヱントな条件下でハイブリダィズする少なくとも 15塩基からなるポリヌクレオチド を含む、請求項 4な!/、し 7の!/、ずれか 1項に記載の診断薬。  [11] Polynucleotide consisting of at least 15 contiguous bases contained in the base sequence consisting of the 4th to 10961466th base sequences of the nucleotide sequence described in GenBank accession number NT-0109273.18 or its complementary base sequence The diagnostic agent according to claim 4, wherein the diagnostic agent comprises a polynucleotide comprising at least 15 bases that hybridizes with the polynucleotide under stringent conditions.
[12] 配列表の配列番号 1〜8のうちいずれ力、 1の塩基配列からなる核酸プローブを含む、 請求項 4な!/、し 7の!/、ずれか 1項に記載の診断薬。  [12] The diagnostic agent according to [4], comprising a nucleic acid probe consisting of one nucleotide sequence of SEQ ID NOs: 1 to 8 in the sequence listing.
[13] 配列表の配列番号 9〜; 14のうちいずれ力、 1の塩基配列からなるプライマーを含む、 請求項 4な!/、し 7の!/、ずれか 1項に記載の診断薬。  [13] The diagnostic agent according to [4], which comprises a primer consisting of one nucleotide sequence of SEQ ID NOs: 9 to 14 in the sequence listing.
[14] 下記の群より選ばれるタンパク質を特異的に認識する抗体を含む、請求項 4ないし 7 の!/、ずれか 1項に記載の診断薬;  [14] The diagnostic agent according to claim 4 or 7, wherein the diagnostic agent comprises an antibody that specifically recognizes a protein selected from the following group;
(DTRIM33 (tripartite motif 33)タンパク質、  (DTRIM33 (tripartite motif 33) protein,
(2)BCAS2 (breast carcinoma amplified sequence-2)タンパク質又は (3)DENND2C (DENN/MADD domain containing 2C)タンパク質。 (2) BCAS2 (breast carcinoma amplified sequence-2) protein or (3) DENND2C (DENN / MADD domain containing 2C) protein.
[15] 抗体がモノクローナル抗体又はポリクローナル抗体である請求項 14に記載の診断薬 15. The diagnostic agent according to claim 14, wherein the antibody is a monoclonal antibody or a polyclonal antibody.
[16] 小細胞癌又は乳癌の診断方法であって、ヒトから分離された生体試料にお!/、て下記 ( [16] A method for diagnosing small cell cancer or breast cancer in a biological sample isolated from humans!
1)の染色体領域の増幅及び/又は下記 (2)の遺伝子の発現亢進を検出する工程を 含む診断方法;  A diagnostic method comprising the step of detecting the amplification of the chromosomal region of 1) and / or the enhanced expression of the gene of (2) below;
(1) GenBankァクセッションナンバー NT— 019273.18に記載された塩基配列の第 1059 7582番目力、ら第 11121868番目までからなる塩基配列で表されるポリヌクレオチドによ り実質的に示される染色体領域、  (1) Chromosomal region substantially represented by a polynucleotide represented by the nucleotide sequence consisting of the 1059th 7582th force of the nucleotide sequence described in GenBank Accession Number NT-0192713.18, and the 11121868th nucleotide sequence,
(2) TRIlVw3 (tripartite motif «53ノ退 ナ、 BCAS2 (breast carcinoma amplined sequence -2)遺伝子、又は DENND2C (DENN/MADD domain containing 2C)遺伝子。  (2) TRIlVw3 (tripartite motif «53 knockout gene, BCAS2 (breast carcinoma amplined sequence -2) gene, or DENND2C (DENN / MADD domain containing 2C) gene).
[17] TRIM33 (tripartite motif 33)遺伝子の増幅及び/又は TRIM33遺伝子の発現亢進を 検出する工程を含む小細胞癌又は乳癌の診断方法。  [17] A method for diagnosing small cell cancer or breast cancer, comprising a step of detecting amplification of TRIM33 (tripartite motif 33) gene and / or increased expression of TRIM33 gene.
[18] 小細胞癌が肺小細胞癌である請求項 16又は 17に記載の診断方法。 18. The diagnostic method according to claim 16 or 17, wherein the small cell cancer is small cell lung cancer.
[19] 請求項 4な!/、し 15の!/、ずれか 1項に記載の診断薬を用いる請求項 16な!/、し 18の!/、 ずれか 1項に記載の診断方法。 [19] The diagnostic method according to claim 16, wherein the diagnostic agent according to claim 1 is used, and the diagnostic agent according to claim 1 is used.
[20] 小細胞癌細胞又は乳癌細胞に対する増殖阻害作用を有する物質のスクリーニング 方法であって、下記の群より選ばれるいずれ力、 1の遺伝子の増幅及び/又は発現を 抑制する作用を有する物質を選択する工程を含む方法; [20] A screening method for a substance having a growth inhibitory action on small cell cancer cells or breast cancer cells, wherein a substance having an action of suppressing amplification and / or expression of a gene of any one selected from the following group: A method comprising a step of selecting;
(DTRIM33 (tripartite motif 33)遺伝子、  (DTRIM33 (tripartite motif 33) gene,
(2) Bし AS2 (Dreast carcinoma amplined sequence- 2)退 子、及び  (2) B2 AS2 (Dreast carcinoma amplined sequence-2)
(3) DENND2C (DENN/MADD domain containing 2C)遺伝子。  (3) DENND2C (DENN / MADD domain containing 2C) gene.
[21] 小細胞癌細胞又は乳癌細胞に対する増殖阻害作用を有する物質のスクリーニング 方法であって、 TRIM33 (tripartite motif 33)遺伝子の増幅及び/又は発現を抑制す る作用を有する物質を選択する工程を含む方法。  [21] A method for screening a substance having an inhibitory action on growth of small cell cancer cells or breast cancer cells, comprising a step of selecting a substance having an action of suppressing amplification and / or expression of a TRIM33 (tripartite motif 33) gene. Including methods.
[22] 小細胞癌細胞が肺小細胞癌細胞である請求項 20又は 21に記載のスクリーニング方 法。  [22] The screening method according to [20] or [21], wherein the small cell carcinoma cells are lung small cell carcinoma cells.
[23] 小細胞癌又は乳癌の治療のための医薬であって、下記の群より選ばれるいずれか 1 の遺伝子の増幅及び/又は発現を抑制する作用を有する物質を有効成分として含 む医薬; [23] A medicament for the treatment of small cell cancer or breast cancer, any one selected from the following group 1 A medicament comprising as an active ingredient a substance having an action of suppressing amplification and / or expression of the gene of
(DTRIM33 (tripartite motif 33)遺伝子、  (DTRIM33 (tripartite motif 33) gene,
(2) Bし AS2 (Dreast carcinoma amplined sequence- 2)退 子、及び  (2) B2 AS2 (Dreast carcinoma amplined sequence-2)
(3) DENND2C (DENN/MADD domain containing 2C)遺伝子。  (3) DENND2C (DENN / MADD domain containing 2C) gene.
[24] 小細胞癌又は乳癌の治療のための医薬であって、 TRIM33 (tripartite motif 33)遺伝 子の増幅及び/又は発現を抑制する作用を有する物質を有効成分として含む医薬  [24] A medicament for treating small cell cancer or breast cancer, comprising a substance having an action of suppressing amplification and / or expression of a TRIM33 (tripartite motif 33) gene as an active ingredient
[25] 小細胞癌の治療のための医薬であって、 TRIM33 (tripartite motif 33)遺伝子の増幅 及び/又は発現を抑制する作用を有する物質を有効成分として含む医薬。 [25] A medicament for treating small cell cancer, comprising a substance having an action of suppressing amplification and / or expression of TRIM33 (tripartite motif 33) gene as an active ingredient.
[26] 小細胞癌が肺小細胞癌である請求項 23ないし 25のいずれか 1項に記載の医薬。 26. The medicament according to any one of claims 23 to 25, wherein the small cell cancer is small cell lung cancer.
[27] 小細胞癌細胞又は乳癌細胞に対する増殖阻害剤であって、以下の群より選ばれる 2 重鎖ポリヌクレオチドを有効成分として含む増殖阻害剤; [27] A growth inhibitor for small cell cancer cells or breast cancer cells, comprising a double-stranded polynucleotide selected from the following group as an active ingredient;
(1)配列表の配列番号 17に記載の塩基配列で表されるポリヌクレオチドと配列表の配 列番号 18に記載の塩基配列で表されるポリヌクレオチドとからなる 2重鎖ポリヌクレオ チド、及び  (1) a double-stranded polynucleotide comprising a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 17 in the sequence listing and a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 18 in the sequence listing; and
(2)配列表の配列番号 19に記載の塩基配列で表されるポリヌクレオチドと配列表の配 列番号 20に記載の塩基配列で表されるポリヌクレオチドとからなる 2重鎖ポリヌクレオ チドを含む増殖阻害剤。  (2) Proliferation comprising a double-stranded polynucleotide consisting of a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 19 in the sequence listing and a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 20 in the sequence listing Inhibitor.
[28] 小細胞癌細胞が肺小細胞癌細胞である請求項 27に記載の増殖阻害剤。  [28] The growth inhibitor according to claim 27, wherein the small cell carcinoma cell is a lung small cell carcinoma cell.
[29] 小細胞癌又は乳癌の治療のための医薬であって、以下の群より選ばれる 2重鎖ポリ ヌクレオチドを有効成分として含む医薬; [29] A medicament for the treatment of small cell cancer or breast cancer, comprising a double-stranded polynucleotide selected from the following group as an active ingredient;
(1)配列表の配列番号 17に記載の塩基配列で表されるポリヌクレオチドと配列表の配 列番号 18に記載の塩基配列で表されるポリヌクレオチドとからなる 2重鎖ポリヌクレオ チド、及び  (1) a double-stranded polynucleotide comprising a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 17 in the sequence listing and a polynucleotide represented by the nucleotide sequence set forth in SEQ ID NO: 18 in the sequence listing; and
(2)配列表の配列番号 19に記載の塩基配列で表されるポリヌクレオチドと配列表の配 列番号 20に記載の塩基配列で表されるポリヌクレオチドとからなる 2重鎖ポリヌクレオ チドを含む医薬。 小細胞癌が肺小細胞癌である請求項 29に記載の医薬。 (2) A pharmaceutical comprising a double-stranded polynucleotide comprising a polynucleotide represented by the base sequence set forth in SEQ ID NO: 19 in the sequence listing and a polynucleotide represented by the base sequence set forth in SEQ ID NO: 20 in the sequence listing. . 30. The medicament according to claim 29, wherein the small cell cancer is small cell lung cancer.
PCT/JP2007/074408 2006-12-20 2007-12-19 Marker for diagnosis of cancer, and target molecule for therapy WO2008075713A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2006342462A JP2010051172A (en) 2006-12-20 2006-12-20 Marker for diagnosis of cancer and target molecule oslc1 for therapy
JP2006-342462 2006-12-20

Publications (1)

Publication Number Publication Date
WO2008075713A1 true WO2008075713A1 (en) 2008-06-26

Family

ID=39536340

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2007/074408 WO2008075713A1 (en) 2006-12-20 2007-12-19 Marker for diagnosis of cancer, and target molecule for therapy

Country Status (2)

Country Link
JP (1) JP2010051172A (en)
WO (1) WO2008075713A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110387424A (en) * 2019-07-12 2019-10-29 安徽农业大学 BCAS2 gene is used to judge the application of starting time country fair boar puberty
WO2020069661A1 (en) * 2018-10-04 2020-04-09 Cdi Laboratories, Inc. Serological biomarkers for early diagnosis of lung cancer

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2005304497A (en) * 2004-03-25 2005-11-04 Joji Inasawa Method for detecting cancer using specified cancer-related gene and method for inhibiting the cancer

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2005304497A (en) * 2004-03-25 2005-11-04 Joji Inasawa Method for detecting cancer using specified cancer-related gene and method for inhibiting the cancer

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
DUPONT S. ET AL.: "Germ-layer Specification and Control of Cell Growth by Ectodermin, a Smad4 Ubiquitin Ligase", CELL, vol. 121, 2005, pages 87 - 99 *
FOROZAN F. ET AL.: "Comparative Genomic Hybridization Analysis of 38 Breast Cancer Cell Lines: A Basis for Interpreting Complementary DNA Microarray Data", CANCER RES., vol. 60, 2000, pages 4519 - 4525, XP002304931 *
MAASS N. ET AL.: "Amplification of the BCAS2 gene at chromosome 1p13.3-21 in human primary breast cancer", CANCER LETT., vol. 185, 2002, pages 219 - 223, XP002277815, DOI: doi:10.1016/S0304-3835(02)00286-0 *
NAGASAKI K. ET AL.: "Identification of a novel gene, DAM1, amplified at chromosome 1p13.3-21 region in human breast cancer cell lines", CANCER LETT., vol. 140, 1999, pages 219 - 226, XP009029869, DOI: doi:10.1016/S0304-3835(99)00091-9 *
YOKOI S. ET AL.: "A Novel Target Gene, SKP2, within the 5p13 Amplicon That Is Frequently Detected in Small Cell Lung Cancers", AM. J. PATHOL., vol. 161, no. 1, 2002, pages 207 - 216, XP002463141 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020069661A1 (en) * 2018-10-04 2020-04-09 Cdi Laboratories, Inc. Serological biomarkers for early diagnosis of lung cancer
WO2020069637A1 (en) * 2018-10-04 2020-04-09 Cdi Laboratories, Inc. Serological biomarkers for early diagnosis of lung cancer
US20220003768A1 (en) * 2018-10-04 2022-01-06 Cdi Laboratories, Inc. Serological biomarkers for early diagnosis of lung cancer
CN110387424A (en) * 2019-07-12 2019-10-29 安徽农业大学 BCAS2 gene is used to judge the application of starting time country fair boar puberty

Also Published As

Publication number Publication date
JP2010051172A (en) 2010-03-11

Similar Documents

Publication Publication Date Title
EP2740742B1 (en) Fusion gene of kif5b gene and ret gene, and method for determining effectiveness of cancer treatment targeting fusion gene
US7867709B2 (en) Method for detecting cancer and method for suppressing cancer
KR101944555B1 (en) Method for the diagnosis, prognosis and treatment of breast cancer metastasis
KR100863405B1 (en) Markers for Diagnosis of Cancer and Its Use
KR20150122731A (en) Method for the prognosis and treatment of cancer metastasis
JP2012504426A (en) Compositions, kits and methods for cancer diagnosis, prognosis and monitoring using GOLPH3
KR20080005193A (en) Breast cancer related gene znfn3a1
US20080182250A1 (en) Method for detecting multiple myeloma and method for inhibiting the same
JP2009165473A (en) Cancer
TW201204393A (en) Diagnostic agent and therapeutic agent of cancer
WO2008075713A1 (en) Marker for diagnosis of cancer, and target molecule for therapy
US9090942B2 (en) Method for detecting esophageal carcinoma and agent for suppressing esophageal carcinoma
US9229003B2 (en) Method for detecting thyroid carcinoma
JP4617257B2 (en) Human cancer-related genes, products encoded by them and applications
EP1045926B1 (en) Gene family with transformation modulating activity
EP1183392A1 (en) Diagnosis, prognosis and treatment of cancer related to the barx2 gene
KR100891992B1 (en) Markers for Diagnosis of Cancer and Its Use
US20100255503A1 (en) Markers for diagnosis of cancer and its use
KR20110036560A (en) Composition for diagnosing colorectal cancer and use thereof
KR20110036559A (en) Composition for diagnosing colorectal cancer and use thereof
KR20070032074A (en) Method of predicting the responsiveness of a tumour to erbb receptor drugs
KR20110036557A (en) Composition for diagnosing colorectal cancer and use thereof
KR20110036556A (en) Composition for diagnosing colorectal cancer and use thereof
KR20110036558A (en) Composition for diagnosing colorectal cancer and use thereof
KR20110036561A (en) Composition for diagnosing colorectal cancer and use thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07850878

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07850878

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP