WO2007026960A1 - Utilisation du gène mocs3 à des fins thérapeutiques ou diagnostiques - Google Patents

Utilisation du gène mocs3 à des fins thérapeutiques ou diagnostiques Download PDF

Info

Publication number
WO2007026960A1
WO2007026960A1 PCT/JP2006/317705 JP2006317705W WO2007026960A1 WO 2007026960 A1 WO2007026960 A1 WO 2007026960A1 JP 2006317705 W JP2006317705 W JP 2006317705W WO 2007026960 A1 WO2007026960 A1 WO 2007026960A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
gene
protein
antibody
moc
Prior art date
Application number
PCT/JP2006/317705
Other languages
English (en)
Japanese (ja)
Inventor
Shinichirou Niwa
Yasutaka Makino
Tomoki Ikuta
Kazuya Arai
Takayuki Shindou
Hiromichi Ogura
Original Assignee
Link Genomics, Inc.
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 Link Genomics, Inc. filed Critical Link Genomics, Inc.
Priority to JP2007533382A priority Critical patent/JPWO2007026960A1/ja
Publication of WO2007026960A1 publication Critical patent/WO2007026960A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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

Definitions

  • the present invention relates to the MCS3 gene, which is a gene specifically amplified in cancer, and its therapeutic or diagnostic use.
  • Cancers Malignant tumors (cancers) are characterized by lethality due to generalization through proliferation, invasion, and metastasis.
  • Local therapies such as surgical resection or radiotherapy cannot adequately address metastatic recurrent cancer, and the development of systemic pharmacotherapy is expected to improve the outcome of cancer treatment in the future.
  • Chemotherapy which is the current center of cancer drug therapy, often uses cell killing agents that act directly on the DNA and / or RNA of cancer cells and cause cell death. For example, bone marrow cells, germ cells, hair matrix cells, and gastrointestinal epithelial cells also acted on normal cells with many divisions, resulting in strong side effects.
  • EGFR epidermal growth factor receptor
  • tyrosine kinase inhibitor ylessa gene: gefitinib
  • Herceptin gene: trastuzumab
  • HE R-2 human epidermal growth factor receptor 2
  • the present inventors have found that the gene that is frequently amplified in cancer (eg, colorectal cancer) is the MOC S 3 gene. I found it.
  • the present inventors have further found that cancer cell proliferation can be suppressed by inhibiting the expression of MOCS 3 protein in colorectal cancer cell lines and cervical cancer cell lines, thereby completing the present invention. It came to. That is, the present invention provides the following cancer therapeutic agents, screening methods for candidate substances having a cancer-suppressing activity, cancer diagnostic agents, cancer diagnostic kits, cancer diagnostic methods, and the like.
  • a therapeutic agent for cancer comprising an OC S 3 gene expression inhibitor as an active ingredient.
  • the cancer therapeutic agent according to (1) above comprising a substance selected from the group consisting of
  • a cancer therapeutic agent comprising a MOC S 3 protein activity inhibitor as an active ingredient.
  • the cancer therapeutic agent according to (3) above comprising a substance selected from the group consisting of
  • a screening method comprising a step of selecting a compound to be reduced.
  • a screening method comprising a step of selecting a compound that binds to the MOCS 3 protein.
  • a cancer therapeutic agent comprising the antibody according to (9) above.
  • a cancer diagnostic agent comprising the antibody according to (9) above.
  • a cancer diagnostic agent comprising a nucleotide sequence that can be hyper-predated under stringent hyper-precipitation conditions in the base sequence of the MOC S 3 gene or a part thereof.
  • a kit for cancer diagnosis containing a polynucleotide comprising a base sequence that can be hyperpredified under high-precipitation conditions that are stringent to the base sequence of the MO C S 3 gene or a part thereof.
  • a method for treating cancer comprising a step of administering a MOC S 3 gene expression inhibitor to a patient.
  • a method for treating cancer comprising the step of administering a MO CS 3 protein activity inhibitor to a patient.
  • a MOC S 3 gene expression inhibitor for the manufacture of a medicament for treating cancer.
  • the present invention provides novel drugs, kits and methods useful for the treatment and Z or diagnosis of cancer, particularly colorectal cancer, and a screening method for candidate compounds having cancer suppressive action.
  • FIG. 1 is a histogram showing the frequency of the MO C S 3 gene relative to the degree of gene amplification in 20 samples from colon cancer patients.
  • Fig. 2 is an optical micrograph (phase contrast image) showing the results of RNA i analysis when siRNA of the MCS3 gene was transfected into the colon cancer cell line RKOE6.
  • Fig. 3 is a graph showing the results of quantitative evaluation of RNA i effect by RT-PCR when sciRNA of MOC S3 gene was transfected into colorectal cancer cell line RKOE6.
  • FIG. 4 is a graph showing the results of evaluating the RNA i effect by measuring the number of viable cells when the siC of the MOC S 3 gene was transfected into the colon cancer cell line RKOE 6.
  • Fig. 5 is a photograph showing the results of verifying the RNA i effect with an optical microscope when siRNA of the MO CS 3 gene was transfected into a cell line CCD 18 Co derived from normal colon tissue. Phase difference image).
  • Figure 6 is a graph showing the results of quantitative RT-PCR evaluation of RNA i effects when MO CS 3 gene siRNA was transfected into cell line CCD 18 Co derived from normal colon tissue. is there.
  • Fig. 7 is a graph showing the results of verifying the RNA i effect by measuring the number of viable cells when transfecting siRNA of the MO CS 3 gene into the cell line C CD 1 8 Co derived from normal colon tissue .
  • Fig. 8 is a photograph showing the results of Northern hybridization performed using various normal organ tissues.
  • Fig. 9 is an optical micrograph (fluorescence image) of a portion (6 cells) of cancer cells in a sample tissue derived from a colon cancer patient analyzed by the FISH method.
  • FIG. 10 is an optical micrograph (bright field image) showing the results of evaluation by immunohistostaining that the M0CS3 gene protein is expressed in cancer cells of the specimen tissue.
  • Fig. 11 is a graph showing the results of verifying the RNAi effect by measuring the number of viable cells when transfection of the MO C S3 gene siRNA into the cervical cancer cell line HeLa cell line.
  • Fig. 12 is an optical micrograph (differential interference image) showing the results of taking a detailed observation of the behavior of the experiment in Fig. 11 taken in a time-series under a microscope.
  • Figures 13A and B are graphs showing the results of (A) serum derived from a colon cancer patient and (B) serum derived from a healthy subject analyzed by mass spectrometry, respectively.
  • FIGS. 14A to C show the correspondence between the peak and amino acid (or amino acid sequence) shown in FIG. 13 determined by MS / MS analysis.
  • Fig. 1 5 _ 1 and Fig. 1 5-2 are the organs analyzed by immunohistochemical staining, where expression was observed in the stomach (A: stomach), colon (B: colon), rectum (C: rectum), Retina (D: epiploon), lungs (E, F, G ⁇ lung), nest (I, J: ovary), prostate (L: prostate), lymph node (M: Lymp node), bone tissue (0: bone), mediastinum (R: mediastinum), and weakly expressed milk (H: breast), spleen (K: spleen), throat (P: throat), and adipose tissue (Q: fatty tissue) ) An optical micrograph (bright field image) of the sample from origin.
  • the present inventors verified a gene amplified by the array CGH method using a sample derived from a colorectal cancer patient, and identified a gene amplification region specific for colorectal cancer. It was. We found that human MOC S 3 (Molybdenum cofactorsynthesis 3) gene is frequently found in colorectal cancer patients among the regions where amplification occurs frequently in the samples.
  • human MOC S 3 Molybdenum cofactorsynthesis 3
  • the human MCS3 gene found by the present inventors is involved in the synthesis system of oxidoreductases containing molybdenum.
  • Molybdenum is a trace element contained in many foods such as legumes, dairy products, and meat, and is known to play an important role in vivo. Molybdenum combines with Molybdopterin (MTP) to form M o 1 ynden um cofactor (M oco. Mo co consists of xanthine oxidase (XO), aldehyde oxidase (AO), and sulfite oxidase ( It is essential for the activity of the three enzymes SO) XO is involved in the metabolism of purine derivatives, AO catalyzes the oxidation of aldehydes to strong sulfonic acids, and SO is required for the metabolism of sulfur-containing amino acids. An enzyme that oxidizes sulfite to sulfuric acid.
  • MTP Molybdopterin
  • Mo co consists of xanthine oxidase (XO), aldehyde oxidase (AO), and sulfite
  • Molybdenum deficiency has been reported in patients who have clinical signs such as increased heartbeat, headache, psychiatric disorder, and coma and are receiving long-term parenteral nutrition. Symptoms of this molybdenum deficiency include very low uric acid levels in serum and urine due to low XO activity, and low urinary sulfuric acid levels in urine due to low soo activity. Molybdenum cofactor deficiency due to congenital anomalies that show symptoms such as severe brain damage and ocular vitreous dislocation is known (Sardesai, VM (1 9 9 3) Nu tr C lin P ract. 8 (6 ), 2 7 7-8 1).
  • Molybdenum cofactor deficiency has been reported to involve two mutations in Mo 1 ybden um cofactor synthesisl (MOC S l) and 2 (MOC S 2) (Reiss, J. eta 1. (1 9 9 8) Am. J. Hum. G enet. 1 0 3, 6 3 9-644; Reiss, J. Eta 1. (1 9 9 9) Am. J. Hum. G enet. 64, 7 0 6-7 1 1 ) c
  • MO CS 3 is homologous to Moe B on the N-terminal side, but homologous to Rhodanese (thiosu 1 fate: cyanidesulfurtransferase, EC 2. 8. 1. 1) on the C-terminal side ( M endel, R. R. & S chwa rz, G. (2 0 0 2) M et. Ions Biol. Syst. 3 9, 3 1 7— 3 6 8).
  • MOC S 3 transfers sulfur to MO C S 2 and activates M OC S 2 in the MTP synthesis system.
  • Activated MOC S 2 converts Precursor Z synthesized from GTP by MOC S 1 into MTP (Matthies, A. eta 1. (2 0 04) PNA S 1 0 1 (1 6), 5 946-5 9 5 1; Mathesies, A. eta 1. (2 0 0 5) B ioch em. 44, 7 9 1 2— 7 9 2 0).
  • MTP synthesized in this way combines with molybdenum and becomes Mo co.
  • M o co is essential for the activity of three enzymes: xanthine oxidase (X O), aldehyde oxidase (AO), and sulfite oxidase (S O).
  • the present inventors have also confirmed that the growth of cancer cells can be suppressed by suppressing the expression of the MO C S 3 gene by RNA i (RNA interference). Therefore, it is possible to treat cancer by suppressing the expression of the MCS3 gene. It is also possible to diagnose cancer by measuring the expression level of the MOC S 3 gene.
  • RNA i RNA interference
  • the present invention provides (1) a substance that inhibits expression of MO CS 3 gene as an active ingredient And (2) a cancer therapeutic agent containing, as an active ingredient, a MCS3 protein activity inhibitor.
  • MOC S 3 gene refers to a human MO CS 3 gene consisting of 24 5 8 bases registered in the Accession No .: NM_ 0 1 4 48 4 in the NC BI nucleotide database ( SEQ ID NO: 1) means, but not limited thereto, for example, a variant that has been altered by having one or more base substitutions, deletions, additions, or insertions in the base sequence of the gene.
  • a gene consisting of a polynucleotide comprising a base sequence that can be hybridized under high-precipitation conditions that are stringent to the base sequence of the gene or its complementary sequence is also referred to as “MOC S 3 gene”. It shall be included.
  • Hypridization is a known method or a method similar thereto, for example, molecular cloning (Molecu 1 r Cloning Third E dition, J. S amb rooketa 1., C old S pr ing Harbor L ab. Press . 2 0 0 1) and the like.
  • the “stringent condition” may be a low stringent condition, a moderate stringent condition, or a high stringent condition.
  • “Low stringent conditions” are, for example, 5 XSSC, 5 X Denhardt's solution, 0.5% SDS, 50% formamide, 32 ° C.
  • “Medium stringent conditions” are, for example, conditions of 5 XSSC, 5 X Denhardt's solution, 0.5% SDS, 50% formamide, and 42 ° C. “High stringent conditions” are, for example, conditions of 5 XSSC, 5 X Denhardt's solution, 0.5% SDS, 50% formamide, 50. Under these conditions, it can be expected that DNA having high homology can be efficiently obtained as the temperature is increased. However, it affects the stringency of the hybridization. Multiple factors such as temperature, probe concentration, probe length, ionic strength, time, and salt concentration can be considered, and those skilled in the art can select similar factors to achieve the same stringency. It can be realized.
  • polynucleotide that can be hybridized the base sequence of SEQ ID NO: 1, for example, 70% or more, when calculated using the default parameter—by homology search software such as FAS TA, BLAST, etc. 7 5% or more, 80% or more, 85% or more, 90% or more, 9 1% or more, 9 2% or more, 93% or more, 94% or more, 95% or more, 96% or more, Examples include polynucleotides having 9 7% or more, 98% or more, and 99% or more identity.
  • inhibition of gene expression means any event in a series of events from gene to protein production (for example, transcription (production of mRNA), translation (production of protein)) By inhibiting the production of the protein encoded by the gene.
  • MOC S 3 protein consists of 460 amino acid residues registered under Accession No .: NP— 0-5 5 2 9 9 on the basis of NC BI protein data. Retains the human MO CS 3 protein (SEQ ID NO: 2) and substantially the same activity as this protein (for example, one or more activities selected from enzyme activities such as Rhodanese activity and Tiocarboxylation activity). A mutant protein consisting of an amino acid sequence in which deletion, substitution, insertion, and addition or deletion of one to a plurality of amino acid residues has occurred in the amino acid sequence.
  • the amino acid mutation site and number in the above mutant protein are not particularly limited as long as the mutant protein retains substantially the same activity as the original protein, but the number of mutations is, for example, 1 to 50. 1 to 40 pieces, 1 to 30 pieces, 1 to 25 pieces, 1 to 20 pieces, 1 to 15 pieces, 1 to 10 pieces, 1 to 9 pieces, 1-8 pieces, 1-7 pieces! ⁇ 6 pieces (1 ⁇ several pieces), 1 ⁇ 5 pieces, 1 ⁇ 4 pieces, 1 ⁇ 3 pieces, 1-2 pieces, 1 piece. In general, the smaller the number of mutations, the better.
  • such a mutant protein has an amino acid sequence of SEQ ID NO: 2 and about 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 9 1% or more, 9 2% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, 99% or more amino acid sequences having 9% or more identity
  • a protein having substantially the same activity as the original protein In general, the larger the homology value, the better.
  • the MO CS 3 protein includes “partial peptides” of the MO CS 3 protein.
  • the partial peptide of the MO CS 3 protein is a partial peptide consisting of a partial amino acid sequence of the amino acid sequence (SEQ ID NO: 2) of the M 0 CS 3 protein. Any substance may be used as long as it has an activity similar to that of protein.
  • SEQ ID NO: 2 at least 20, preferably at least 50, more preferably at least 70, more preferably at least 100, most preferably at least Examples thereof include a polypeptide having an amino acid sequence consisting of at least 200 amino acid residues.
  • these polypeptides contain an amino acid sequence corresponding to the portion involved in the activity of MOCS 3 protein.
  • the partial peptide used in the present invention is one or more of the above-mentioned polypeptides in the amino acid sequence (for example, about 1 to 20 pieces, more preferably about 1 to 10 pieces, still more preferably 1 to 5 amino acid residues) may be changed by deletion, addition, substitution, or insertion.
  • the MO CS 3 protein used in the present invention can be prepared from cells or tissues expressing the protein. Further, these proteins can be synthesized by a known peptide synthesizer, or can be prepared by a recombinant method using an appropriate host cell selected from prokaryotic organisms or eukaryotic organisms.
  • the MO CS 3 protein used in the present invention is although it may be derived from these species, it is preferably derived from a human.
  • Substantially the same activity indicates that these activities are qualitatively equivalent. Accordingly, enzyme activities (such as Rhodanese activity and Tiocarboxylation activity) are equivalent (for example, about 0.01 to 100 times, preferably about 0.5 to 20 times, more preferably about 0.5 to 2). However, quantitative factors such as the degree of activity and the molecular weight of the protein may be different. Enzyme activity is measured according to known methods described in literature such as Maties, A. eta 1. (2 0 0 4) P NAS 1 0 1 (1 6), 5 9 4 6-5 9 5 1, etc. For example, it can be measured according to the screening method described later.
  • B LAS TN and B LAS TX Programs called B LAS TN and B LAS TX based on the BL AS T algorithm have been developed (A 1 tschu 1 SF, eta 1: J Mol B iol 2 1 5: 4 0 3, 1 9 9 0 )
  • amino acid sequences using B LAS TX no.
  • B LAS T and Gap p e d B LAS T programs use the default parameters of each program.
  • cancer therapeutic agent includes anticancer agents, cancer metastasis inhibitors, cancer cell apoptosis inducers, cancer cell proliferation inhibitors, cancer cell infiltration inhibitors, cancer preventive agents, and the like. Used in meaning.
  • cancer (or cancer) and “tumor” are used as terms having the same meaning.
  • the present invention provides a cancer treatment agent containing, as an active ingredient, an expression inhibitor of MCS3 gene. To do.
  • MOC S 3 gene expression inhibitor is not limited as long as it inhibits the expression of the MOC S 3 gene.
  • MOCS 3 gene to MO CS 3 mRNA and a substance that inhibits translation from MOC S 3 mRNA to MOC S 3 protein.
  • Examples of substances that inhibit transcription from the MO C S 3 gene to MO C S 3 mR N A include
  • Examples of substances that block translation from MOC S 3 mRN A to MO C S 3 protein include
  • RNAI action on M0 C S 3 m RN A or a part thereof e.g, si RNA
  • nucleic acid means RNA or DNA.
  • nucleic acid here contains not only purine and pyrimidine bases, but also modifications. It may include those with other heterocyclic bases decorated. Such modifications may include methylated purines and pyrimidines, acylated purines and pyrimidines, acylated purines and pyrimidines, or other heterocycles. Modified nucleosides and modified nucleotides may also be modified at the sugar moiety, for example, one or more hydroxyl groups are replaced by halogens, aliphatic groups, etc., or ethers, amines, etc. It may be converted to a functional group.
  • RNA i is a phenomenon in which the expression of a foreign gene and a target endogenous gene are both inhibited when a double-stranded RNA having the same or similar sequence as the target gene sequence is introduced into the cell.
  • the RNA used here is, for example, 19 to 30 bases in length; double-stranded RNA that causes RNA interference, such as ds RNA (doublestrand RNA), siRNA (smallinterfering RNA) or sh RNA (shorthairpin RNA).
  • RNA can be locally delivered to a desired site by a delivery system such as ribosome, and it can be expressed locally using a vector that produces the above-mentioned double-stranded RNA.
  • ds RNA, si RNA or sh RNA double-stranded RNA
  • the preparation method and use method of such double-stranded RNA are known from many literatures (Special Table 2 0 0 2-5 1 6 0 6 2; US) Publication No. 2 0 0 2Z0 8 6 3 5 6 A; Nature Genetics, 2 4 (2), F e b., 1 80-1 8 3; Genesis, 2 6 (4), Ap ril , 24 0-2 44; Nature, S pe.
  • the length of the double-stranded RNA exhibiting the RNA i effect used in the present invention is usually 19 to 30 bases, preferably 20 to 27 bases, more preferably 21 to 25 bases, most preferably Is 2 1 to 2 3 bases.
  • siRNA used in Example 3 can be used.
  • the term “antisense nucleic acid” or “antisense polynucleotide” has a polynucleotide that is complementary to at least a part of a DNA region of interest, and the polynucleotide is a small part of the region.
  • the term refers to a nucleic acid that can be hybridized with a part.
  • the antisense nucleic acid of the present invention is RNA, DNA, or a modified nucleic acid (RNA, DNA).
  • the antisense nucleic acid of the present invention is RNA, DNA or a modified nucleic acid (RNA, DNA).
  • modified nucleic acids include, but are not limited to, nucleic acid sulfur derivatives, thiophosphate derivatives, and those that are resistant to the degradation of oligonucleotides. is not.
  • the antisense nucleic acid to be used is linked downstream of a suitable promoter, and preferably a sequence containing a transcription termination signal is linked on the 3 ′ side.
  • the nucleic acid thus prepared can be transformed into a desired animal using a known method.
  • the sequence of the antisense nucleic acid is preferably a sequence complementary to the endogenous gene of the animal to be transformed or a part thereof, but is completely complementary as long as the gene expression can be effectively suppressed. It doesn't have to be.
  • a complementary antisense sequence in the untranslated region near the 5 'end of the mRNA of the MO C S 3 gene is effective in inhibiting gene translation.
  • a sequence complementary to the coding region or 3 ′ untranslated region can also be used.
  • Antisense nucleic acid effective for inhibiting gene translation is about 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 9% or more of the transcript of the target gene. 5% or more complementarity.
  • the length of the antisense nucleic acid is at least about 10 bases (for example, about 10 to 40), preferably about 15 bases or more More preferably about 100 bases or more, and still more preferably about 500 bases or more.
  • Antisense nucleic acids can be designed with reference to known literature (for example, Hirashima and Inoue, New Chemistry Experiment Course 2 Nucleic acid IV gene replication and expression, Japan Biochemical Society, Tokyo Chemical Dojin, 1 9 9 3, p. 3 1 9-34 7), J. Kawakamieta 1., P harmtech J apa n. V ol. 8, p. 24 7, 1 9 9 2; V o 1. 8, p. 3 9 5, 1 9 9 2; S. T. C rookeeta 1., ed., An tisense R esearchand Ap plications , CR CP ress, 1 9 9 3 etc.).
  • a nucleic acid having a lipozyme activity that specifically cleaves a transcript of the MCS3 gene can be used as an active ingredient.
  • liposome activity refers to a nucleic acid that specifically cleaves mRNA, which is a transcription product of a target gene. Some liposomes are larger than 400 nucleotides, such as group I introns and MRNA contained in RNase P, but about 40 nucleotides called hammerheads and hepins. Some have the active domain of (protein nucleic acid enzyme, 1990, 35, p. 2 191). For example, FEBSL ett, 1 9 8 8, 2 2 8, p.
  • a compound other than a nucleic acid that inhibits the transcriptional activity of the MOC S3 gene can be used as an active ingredient.
  • a compound is, for example, a compound that binds to a factor involved in the expression / transcription of the MOCS3 gene.
  • a compound may be a natural product or a synthetic compound.
  • Such a compound can be obtained by the screening method described below.
  • Cancer therapeutic agent containing an activity inhibitor of MOC S 3 protein in another embodiment, the present invention also provides activity of MO CS 3 protein.
  • a cancer therapeutic agent containing an inhibitor is provided.
  • MO C S 3 protein activity inhibitor includes, for example,
  • antibody means an antibody that reacts with the full length or fragment of a protein. There is no particular limitation on the form of the antibody of the present invention.
  • anti-MOC S 3 antibodies can be prepared by methods known to those skilled in the art. Details of the anti-MO C S 3 antibody will be described later. '
  • MO CS 3 protein mutant having a dominant negative property relative to MO CS 3 protein refers to an endogenous wild type MO CS 3 protein expressed by expressing a gene encoding the same. This refers to a protein having a function of eliminating or reducing the quality activity (see Kunihiro Tsuchida, Gene Activity Inhibition Experiment, edited by Yoshikazu Tahira, Yodosha (2 0 1) 2 6-3 2).
  • a compound other than the above antibody or mutant that binds to MOCS3 protein can be used as an active ingredient.
  • a compound is, for example, a compound that binds to MO CS 3 protein and inhibits its activity.
  • Such a compound may be a natural product or a synthetic compound. Such compounds should be obtained by screening methods described below. Is possible.
  • the substance capable of inhibiting the activity of the above-described MCS3 protein of the present invention can be used as a cancer therapeutic agent. 2. Screening method for substances that inhibit the activity or expression of MO C S 3 protein
  • the present invention also provides a method for screening a candidate compound having a cancer suppressing action.
  • One preferred embodiment is a method using as an index the binding between the MOCS 3 protein and the test compound.
  • a compound that binds to MCS3 protein is expected to have an effect of inhibiting the activity of MOCS3 protein.
  • the compound preferably binds to the active site of the MOCS3 protein.
  • the MOC S 3 protein is contacted with a test compound.
  • the MOCS 3 protein can be used, for example, in a purified form of the MOCS3 protein, in a form expressed intracellularly or extracellularly, or affinity, depending on the indicator for detecting binding to the test compound. It can be in the form bound to a column.
  • the test compound used in this method can be appropriately labeled as necessary. Examples of the label include a radiolabel and a fluorescent label.
  • test compound used for this method.
  • natural compounds, organic compounds, inorganic compounds, proteins, peptides, etc. as well as compound libraries, gene library expression products, cell extracts, cell culture supernatants, fermented microorganism products, oceans
  • compound libraries include, but are not limited to, biological extracts and plant extracts.
  • the binding between the M CS3 protein and the test compound can be detected by, for example, a label attached to the test compound bound to the MOCS3 protein.
  • the binding activity between a protein and a test compound can be measured by a known method (for example, measurement of Nitratereductase activity or Sulfurtransferase activity (Matthies, A. eta 1. (2 0 04) PNAS 1 0 1 (Refer to (16), 594 6-5 9 5 1)). In this method, a test compound that binds to the MOCS3 protein and inhibits its activity is then selected.
  • the compound isolated by this method is expected to have a cancer suppressing action and is useful as a cancer therapeutic agent.
  • Another embodiment of the screening method of the present invention is a method using the expression of the MOCS3 gene as an index.
  • a test compound is brought into contact with a cell expressing the MCS3 gene.
  • a cell expressing the MCS3 gene examples include cells derived from pets, livestock, etc., such as humans, mice, cats, cats, dogs, eagle, birds, but not limited to these.
  • Cells that express the MCS3 gene include cells that express the endogenous MOCS3 gene, or cells that have had the exogenous MOCS3 gene introduced and expressed. can do.
  • a cell in which an exogenous MOC S 3 gene is expressed can usually be produced by introducing an expression vector into which a MOC S 3 gene has been inserted into a host cell.
  • the expression vector can be prepared by general genetic engineering techniques.
  • test compound used in this method is not particularly limited.
  • a natural compound, an organic compound, an inorganic compound, a single compound such as a protein, a peptide, etc., and a compound library, an expression product of a gene library, Cell extracts, cell culture supernatants, fermented microorganism products, marine organism extracts, plant extracts, etc. are used.
  • the “contact” of the test compound to the cell expressing the MOC S 3 gene usually involves adding the test compound to the culture medium of the cell expressing the MOC S 3 gene. However, it is not limited to this method.
  • “contact” can be performed by introducing a DNA vector expressing the protein into the cell.
  • the expression level of the MOCS 3 gene is then measured.
  • expression of a gene includes both transcription and translation.
  • the expression level of a gene can be measured by methods known to those skilled in the art. For example, mRNA is extracted from cells expressing the MO CS 3 gene according to a conventional method, and the transcription level of the gene is obtained by carrying out the Northern hypridase-chasion method or RT-PCR method using this mRNA as a cocoon. Can be measured.
  • the promoter region of the MOC S 3 gene can be isolated according to a conventional method and detected downstream of the labeled gene (eg, luminescence, fluorescence, coloration, etc.
  • the transcription level of the gene can also be measured by linking the gene (including but not limited to) and observing the activity of the marker gene.
  • protein fractions are collected from cells that express the MO CS 3 gene, and the expression level of the MO CS 3 protein is detected by electrophoresis such as SDS-PAGE to measure the translation level of the gene. Can also be done.
  • the antibody used for detecting the MOCS3 protein is not particularly limited as long as it is a detectable antibody. For example, both a monoclonal antibody and a polyclonal antibody can be used.
  • the present invention also provides an anti-MOCS 3 antibody, a cancer therapeutic agent containing this antibody, and the like.
  • the cancer therapeutic agent is used for cancer targeted therapy or targeted drug delivery.
  • anti-MOC S 3 antibody includes an antibody that specifically binds to MCS3 protein (including fragments (partial peptides) or salts thereof).
  • the anti-MOCS3 antibody used in the present invention may be a polyclonal antibody or a monoclonal antibody.
  • the class of the antibody is not particularly limited, and includes antibodies having any isotype such as IgG, IgM, IgA, IgD, or IgE. IgG or IgM is preferable, and IgG is more preferable in view of ease of purification.
  • antibody is used to mean any antibody fragment or derivative.
  • the antibody of the present invention can be produced by a known method. Methods for producing such antibodies are well known in the art (eg, Har 1 ow E. & Lane D., Antibody, Old Spr «ing Harbor Laboratory Pres (1 9 8 8)). See).
  • the protein used as the sensitizing antigen is usually MOCS 3 protein or a salt thereof.
  • the MO CS 3 protein also includes a partial peptide thereof, which is not limited to, for example, a fragment of the amino acid sequence of SEQ ID NO: 2, for example, 20 or more, A partial peptide having 40 or more, 60 or more, 80 or more, 100 or more consecutive amino acid sequence portions.
  • a partial peptide having 40 or more, 60 or more, 80 or more, 100 or more consecutive amino acid sequence portions For example, an amino (N) terminal fragment or a carboxy (C) terminal fragment is used.
  • the partial peptide used in the present invention one or more (preferably about 1 to 10, more preferably several (1 to 6)) amino acid residues in the above amino acid sequence are deleted. , Substitutions, insertions and Z or additions.
  • salts of MO CS 3 protein or partial peptides used herein include salts with inorganic acids (eg, hydrochloric acid, sulfuric acid), and salts with organic acids (eg, acetic acid, formic acid, propionic acid). Is used.
  • the M0 CS3 protein of the present invention used as a sensitizing antigen for antibody acquisition is not limited to the animal species from which it is derived, but is preferably a protein derived from a mammal such as a mouse or human, and particularly a human-derived protein. Is preferred.
  • M0 CS 3 protein The above-described MOC S 3 protein, a partial peptide thereof or a salt thereof (in the present specification, these are collectively referred to as “M0 CS 3 protein”) as an antigen, and a mammal such as a rat , Mouse, and rabbit.
  • the dose of antigen per animal is 0.1 to 100 mg when adjuvant is not used, and 1 to 100 zg when adjuvant is used.
  • adjuvants include Freund's complete adjuvant (FCA), Freund's incomplete adjuvant (FIA), and aluminum hydroxide adjuvant. Immunization is performed mainly by injecting intravenously, subcutaneously or intraperitoneally.
  • the interval between immunizations is not particularly limited, and immunization is carried out 1 to 10 times, preferably 2 to 5 times at intervals of several days to several weeks, preferably at intervals of 2 to 5 weeks. Then, antibody-producing cells are collected 1 to 60 days after the last immunization day, preferably 1 to 14 days later.
  • antibody-producing cells include spleen cells, lymph node cells, peripheral blood cells and the like, and spleen cells or local lymph node cells are preferred.
  • (ii) Cell fusion Cell fusion between antibody-producing cells and myeloma cells is performed in order to obtain a hybridoma.
  • a myeloma cell to be fused with an antibody-producing cell a generally available cell line of an animal such as a mouse can be used.
  • the cell line to be used has drug selectivity and cannot survive in a HAT selection medium (including hypoxanthine, aminopterin, and thymidine) in an unfused state, but can survive only in a state fused with antibody-producing cells. It is preferable to have Examples of myeoma cells include mouse myeoma cells such as X 6 3 A g. 8. 65 3, NS I / 1—Ag 4-1, NS 0 Z 1, and rat myeloma such as YB 2Z0. Cell lines.
  • Cell fusion consists of 1 X 10 s to 1 X 10 7 Zm 1 antibody-producing cells and 2 X 1 0 5 in animal cell culture media such as serum-free DMEM, RPMI — 1 6 40 medium, etc.
  • animal cell culture media such as serum-free DMEM, RPMI — 1 6 40 medium, etc.
  • ⁇ 2 X 1 0 6 cells / m 1 of the Mie port Ichima cells were mixed (antibody producing cells and myeloma port Ichima cells and cell ratio of 2: 1 to 3: 1 is preferred)
  • cells fusion promoter Perform fusion reaction in the presence.
  • the cell fusion promoter polyethylene glycol having an average molecular weight of 100 to 60,000 daltons can be used.
  • antibody-producing cells and myeloma cells can be fused using a commercially available cell fusion device using an electric stimulus (for example, electoral position).
  • the cell suspension is appropriately diluted with, for example, RPM 1-1640 medium containing fetal bovine serum, and then plated on a microtiter plate at about 3 ⁇ 10 5 cells / wel1. Add the selective medium to each well, and then change the selected medium appropriately. As a result, cells that grow 14 days before or after the start of culture in the selective medium can be obtained as a hybridoma.
  • screening is performed to determine whether antibodies that react with the MOC S protein are present in the culture supernatant of the growing hybridoma. The screening of hypridoromas can be carried out in accordance with usual methods, and is particularly limited. It is not a thing.
  • a portion of the culture supernatant contained in a well grown as a hybrid can be collected and screened by enzyme immunoassay, radioimmunoassay, or the like. Cloning of fused cells is performed by limiting dilution. Finally, a hybridoma that is a cell producing a monoclonal antibody that reacts with the MOCS protein is established.
  • a normal cell culture method As a method for collecting a monoclonal antibody from the hybridoma obtained as described above, a normal cell culture method, ascites formation method, or the like can be employed.
  • hypridoma containing 10% urchin fetal serum in RPMI-1 640 medium, MEM medium, or serum-free medium or other animal cell culture medium (for example, 37 ° C, and incubated 5% C_ ⁇ 7-1 4 days at 2 concentrations), to obtain the culture supernatant antibody.
  • the hive re dormer about 1 X 1 0 7 cells were administered intraperitoneally to the mammal of the same species as the animal from which the myeloma cells are derived, is a large amount proliferating High Priestess de one Ma. Ascites is collected after 1-2 weeks.
  • a known method such as ammonium sulfate salting-out method, ion exchange chromatography, gel filtration, affinity chromatography, etc. is appropriately selected, or It can be purified by combining these.
  • the above-mentioned antigen is administered to mammals such as rabbits, mice, and rabbits.
  • the dose of antigen per animal is 0.1 to LOO mg when no adjuvant is used, and 10 to 100 g when adjuvant is used.
  • adjuvants include Freund's complete adjuvant (FCA), Freund's incomplete adjuvant (FIA), and aluminum hydroxide adjuvant.
  • FCA Freund's complete adjuvant
  • FIA Freund's incomplete adjuvant
  • Immunization is performed mainly by injecting intravenously, subcutaneously or intraperitoneally.
  • the immunity interval is There is no particular limitation, and immunization is performed 1 to 10 times, preferably 2 to 5 times at intervals of several days to several weeks, preferably 2 to 5 weeks.
  • enzyme immunoassay enzyme immunoassay (ELISA (enzyme -1 unosorbentassy) or EIA (enzyme unoassay)
  • radioimmunoassay RIA ( radioi mmu noassay)
  • Etc. blood is collected on the day when the maximum antibody titer is shown to obtain antiserum.
  • an antibody (column adsorbed fraction) that reacts with the MO C S 3 protein is collected by applying the polyclonal antibody in the antiserum to an affinity column fixed with the MO C S 3 protein.
  • the reactivity of the polyclonal antibody in the antiserum against the MOC S 3 protein can be measured by the ELISA method or the like.
  • 2 fragment For example, pepsin or papain
  • Humanized antibodies are described, for example, by Riec hma nn et al. (Riec hma nn J o 1 Biol. Oct 5; 2 0 3 (3): 8 2 5—8, 1 9 8 8), and J ones et al. J ones et al. Nature 3 2 1: 5 2 2-5 2 5, 1 986)).
  • Chimeric antibodies include, for example, “Experimental Medicine (Special issue), V o 1. 1.6, No. 1 0, 1 9 8 8”, Japanese Patent Publication No. 3-7 3 2 8 , Humanized antibodies include, for example, “Nature Genetics, Vol. 15, p. 1 4 6-1 5 6, 1 997”, “Nature Genetics, Vol. 7, p. 1 ⁇ 3-2 1, 1 9 94 '', JP 4-5 0 4 3 6 5 gazette, International application publication WO 9 4-2 5 5 8 5 gazette, etc., ⁇ Nikkei Science, June issue, Pp. 40 to 50, 1 995 "," Nature, Vol. 3 6 8, p. 8 5 6-8 5 9, 1 9 94 " Each can be produced with reference to No.
  • the antibody that binds to the MOCS 3 protein of the present invention can be used for the purpose of, for example, suppressing the growth or metastasis of cancer cells.
  • a human antibody or a human-type antibody is preferred in order to reduce immunogenicity.
  • the antibody When used as a diagnostic agent, the antibody may be labeled with a labeling substance for monitoring or the like (for example, a radioisotope, a fluorescent substance, etc.). If necessary, it can be labeled with radioactive substances, fluorescent compounds, etc. Among the most commonly used fluorescent labeling compounds are fluorescein sothiocyanate, rhodamine, phycoerythrin and fluorescein. Similarly, the antibody MOCS3 antibody can be labeled using a bioluminescent compound. The presence of a bioluminescent protein is measured by detecting the presence of fluorescence. Important bioluminescent compounds for this purpose are luciferin, luciferase and aequorin.
  • a labeling substance for monitoring or the like for example, a radioisotope, a fluorescent substance, etc.
  • fluorescent labeling compounds are fluorescein sothiocyanate, rhodamine, phycoerythrin and fluorescein.
  • the antibody of the present invention can be used for specifically detecting MCS3 protein or the like present in a sample such as a body fluid or tissue.
  • production of antibody columns used to purify MOCS 3 protein, etc., detection of MOCS 3 protein, etc. in each fraction during purification, analysis of the behavior of MOCS 3 protein in the test cells Can be used for such as.
  • the anti-MOCS 3 antibody used in the present invention may be an agent having neutral activity that attenuates the activity of the antigen in the therapeutic agent or diagnostic agent of the present invention. Depending on the condition, it can be used in combination with other drugs for producing a therapeutic effect. Therefore, in another aspect, the present invention provides a combination of an anti-MOCS 3 antibody and another drug for use in targeted therapy or targeted imaging of cancer (eg, colorectal cancer). Body, composition containing such a complex Etc. are also provided. According to such an embodiment, the anti-MOC S 3 antibody used in the present invention is used to target other drugs having a therapeutic effect or a labeling agent for diagnosis, etc., to highly express the MO CS 3 protein. Can be delivered to.
  • Examples of the “other drug” used in the present invention include a viral vector or a non-viral vector for introducing a gene into a target, such as a radioisotope, a therapeutic protein, or a small molecule drug. .
  • examples of the “radioisotope” include radioactive halogen elements such as fluorine-18, iodine-1225 ( 125I ), and iodine-1313. These radioactive halogen elements can also be widely used as radiotherapeutic agents or radiodiagnostic agents by labeling them with antibodies or peptides in the same manner as the above-mentioned radioactive metal elements. For example, odorization with 125 I or 131 I can be bound to an antibody or antibody fragment by a known method such as the chloramine T method.
  • technetium one 9 9 m for the diagnosis indium one 1 1 1 and gallium one 6 7 (6 7 G a) such,
  • rhenium one 1 8 6 1 S 6 R e
  • rhenium 1 8 8 1 88 Re
  • a metal chelator is usually used.
  • metal chelating agents include EDTA, DTPA, diaminodithio compound, cyclam, and DOTA. These chelating agents may be pre-bonded to the antibody and then labeled with a radioactive metal, or may be labeled with the antibody after forming a radioactive metal chelate.
  • a site force-in that activates cells responsible for immunity is suitable.
  • human interleukin 2 human granulocyte-macrophage colony stimulating factor , Macrophage colony stimulating factor, human interleukin 12 and the like.
  • ricin, diphtheria toxin, etc. are used to directly kill colon cancer cells.
  • the toxins can be used.
  • a fusion antibody with a therapeutic protein a cDNA that encodes the therapeutic protein is linked to a cDNA that encodes the antibody or antibody fragment, and a DNA that encodes the fusion antibody is constructed.
  • a fusion antibody can be produced by inserting the expression vector into an expression vector for organisms or eukaryotes and introducing the expression vector into a prokaryote or eukaryote.
  • “Small molecule drug” is used herein to mean a diagnostic or therapeutic compound other than “radioisotope” or “therapeutic protein”.
  • small molecule drugs include alkylating agents such as nitrogen mustard and cyclophosphamide, antimetabolites such as 5-fluorouracil and methotrexate, daunomycin, bleomycin, mitomycin C, daunorubicin, Anticancer agents such as antibiotics such as doxorubicin, plant algaide like vincristine, vinblastine, vindesine, hormonal agents such as evening moxifen, dexamethasone (Clinical Oncology (Japan Clinical Oncology Society 1 9 9 6 Cancer and Chemotherapy company))), or steroids such as cortisone hyde, prednisone, non-sterols such as aspirin and indomethacin, immunomodulators such as gold zomarate and penicillamine, cyclophosphamide , Immunosuppression such
  • daunomycin and antibody can be bound by binding between daunomycin and the amino group of the antibody via dartal aldehyde, or binding of the amino acid group of daunomycin and the antibody's loxyl group via water-soluble calpositimide. For example, the method of combining them.
  • a viral vector modified so as to be able to bind to the anti-MOCS 3 antibody of the present invention can be used (for example, an adenovirus vector (Wang, P., eta 1. (1 9 9 5) Soma tic Celland Mo 1 ec. 2 1, 4 2 9— 44 1), retroviral vectors (N aviaux RK, eta 1. (1 9 9 6) J. Virol 7 0, 5 7 0 1-5 7 0 5), lentiviral vectors ( N aldini, L. (1 9 9 8) Curr. Opin. B iotechnol. 9, 45 7- 46 3)).
  • an adenovirus vector Wang, P., eta 1. (1 9 9 5) Soma tic Celland Mo 1 ec. 2 1, 4 2 9— 44 1
  • retroviral vectors N aviaux RK, eta 1. (1 9 9 6) J. Virol 7 0, 5 7 0 1-5 7 0 5
  • lentiviral vectors
  • Such viral vectors have therapeutic effects such as inducing apoptosis of cancer cells at target sites (eg, colon cancer) such as cell proliferation-related genes, apoptosis-related genes, immune control genes, etc.
  • target sites eg, colon cancer
  • the gene to be played (therapeutic gene) is incorporated.
  • Anti-MOCS3 antibody-binding viral vector when administered to patients in need of gene therapy together with anti-MOCS3 antibody, antigen recognized by anti-MOCS3 antibody (ie MOCS3) Can be targeted to the site where is present.
  • the anti-MOC S 3 antibody and the other drug can be combined chemically or genetically.
  • “chemical bond” includes ionic bond, hydrogen bond, covalent bond, bond by intermolecular force, bond by hydrophobic interaction, etc.
  • “gene engineering bond” For example, it includes the binding mode between an antibody and a therapeutic protein when a fusion protein comprising an antibody and a therapeutic protein is produced using a technique such as genetic recombination.
  • a cancer therapeutic agent containing the MOC S 3 gene expression inhibitor of the present invention a cancer therapeutic agent containing a M OC S 3 protein activity inhibitor, a therapeutic agent containing the anti-MOC S 3 antibody of the present invention
  • the anti-M OC S3 antibody used in the present invention is one of a viral vector or a non-viral vector carrying a radioisotope, a therapeutic protein, a small molecule drug, and a therapeutic gene, or any of these
  • a therapeutic agent that is chemically or genetically engineered with the combination can be formulated based on a known method.
  • a pharmaceutically acceptable carrier can be added as necessary according to a conventional method.
  • surfactants for example, surfactants, excipients, coloring agents, flavoring agents, preservatives, stabilizers, buffering agents, suspending agents, tonicity agents, binders, disintegrating agents, lubricants, fluidity promoters,
  • examples include corrigents, but are not limited to these, and other conventional carriers can be used as appropriate.
  • Specific examples include light anhydrous carboxylic acid, lactose, crystalline cellulose, mannitol, starch, carmellose calcium, carmellose sodium, hydroxypropylcellulose, hydroxypropylmethylcellulose, polyvinyl alcohol, decylaminoacetate, polyvinyl Examples include pyrrolidone, gelatin, medium-chain fatty acid tridallylide, polyoxyethylene hydrogenated castor oil 60, sucrose, carboxymethylcellulose, corn starch, and inorganic salts.
  • Examples of the dosage form of the therapeutic agent of the present invention include tablets, powders, pills, powders, granules, fine granules, soft and hard capsules, film coating agents, and pellets as oral agents. , Sublinguals, pastes, etc. As parenterals, injections, suppositories, transdermal agents, ointments, plasters, liquids for external use, etc. can be mentioned.
  • the dosage form can be selected.
  • the active substance M0 C S 3 protein activity (or MO C S 3 gene expression) inhibitor may be contained in the preparation in an amount of 0.1 to 99.9% by weight.
  • the dose of the active ingredient of the drug of the present invention varies depending on the administration subject, target organ, symptom, administration method, etc.
  • oral administration for example, generally for patients (as 6 O k) About 0.1 mg to l, OO Omg, preferably about 1.0 to; I 0 0 mg, more preferably about 1.0 to 50 mg per day.
  • the single dose varies depending on the subject of administration, target organs, symptoms, administration method, etc.
  • patients (for 60 kg) About 0.1 to 3 O mg per day, preferably about 0.1 to 2 Omg, more preferably about 0.1 to 1 Omg is conveniently administered by intravenous injection.
  • the final decision can be made as appropriate based on the judgment of a doctor or veterinarian in consideration of the type of dosage form, administration method, patient age and weight, patient symptoms, and the like.
  • the preparation thus obtained can be administered to, for example, humans and other mammals (eg, rat, rabbit, hidge, buyu, ushi, cat, inu, monkey, etc.). In the case of animals other than humans, the amount converted per 60 kg can be administered.
  • mammals eg, rat, rabbit, hidge, buyu, ushi, cat, inu, monkey, etc.
  • the amount converted per 60 kg can be administered.
  • the therapeutic agent of the present invention is cancer (for example, colorectal cancer, stomach cancer, lung cancer, breast cancer, prostate cancer, esophageal cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder).
  • cancer for example, colorectal cancer, stomach cancer, lung cancer, breast cancer, prostate cancer, esophageal cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder.
  • Prevention / treatment of uterine cancer eg cervical cancer, endometrial cancer
  • testicular cancer eg cervical cancer, endometrial cancer
  • thyroid cancer spleen cancer
  • ovarian cancer brain tumor, blood tumor, etc.
  • the drug of the present invention contains a MCS3 protein activity inhibitor or MCS3 gene expression inhibitor as an active ingredient, it can be used as an anticancer agent, a cancer metastasis inhibitor, a cancer cell apoptosis inducer, or the like. Can be used.
  • the target cell, tissue, organ, or cancer type is not limited to a specific type.
  • the agent of the present invention is both a MOCS 3 protein activity inhibitor and a MOCS 3 gene expression inhibitor. May be included.
  • an antisense nucleic acid When an antisense nucleic acid is used in the therapeutic agent of the present invention, the antisense nucleic acid is inserted alone or into an appropriate vector such as a retroviral vector, an adenoviral vector, an adenoviral association virus vector, Administration can be according to known means.
  • Antisense nucleic acids can be formulated alone or with a physiologically acceptable carrier and administered via a catheter such as a gene gun or a hydrogel catheter.
  • a virus vector such as a recombinant adenovirus particle and an anti-MOCS 3 antibody
  • these may be used alone, but in general, pharmaceuticals are used.
  • an acceptable carrier include those already described above.
  • Body and aqueous isotonic solutions such as water, saline, glucose, human albumin are preferred.
  • additives, preservatives, preservatives, balance, etc. that are usually used in pharmaceutics can also be added.
  • the pharmaceutical composition thus prepared can be administered by an appropriate administration form and administration route depending on the disease to be treated.
  • administration forms include emulsions, syrups, force capsules, tablets, granules, injections, ointments and the like.
  • administering usually from 10 3 to 10 15 virus particles are administered once per adult. However, it may be changed depending on the disease state and the nature of the target cell / tissue.
  • the administration frequency may be once to several times a day, the administration period may be from 1 day to several months or more, and one set is set to 1 to several times, and many sets are administered intermittently over a long period of time. May be.
  • viral vector particles or viral vector nucleic acid molecules used in the present invention can be used for detection of specific cells and / or tissues, or diagnosis of disease states.
  • viral vector particles obtained by incorporating a detectable marker gene into a viral vector nucleic acid molecule and transfecting it into an appropriate host cell can be combined with an anti-MOCS 3 antibody to detect and diagnose tumor cells.
  • an anti-MOCS 3 antibody can be combined with an anti-MOCS 3 antibody to detect and diagnose tumor cells.
  • it can be used for Alternatively, it can be used to detect and diagnose tumor cells by binding a detectable label to the anti-MCS3 antibody.
  • the present invention also provides a diagnostic agent for cancer.
  • the diagnostic agent for cancer of the present invention comprises (a) an antibody against the MOCS 3 protein, or (b) a condition under high hydration conditions stringent to the base sequence of the MOCS 3 gene or a part thereof. It contains a polynucleotide having a base sequence that can be hyperprimed in. Diagnostic agent and diagnostic method using antibody MOCS 3 antibody Since antibodies against MCS CS protein can specifically recognize MOCS 3 protein, etc., MOCS 3 protein in the test solution can be quantified.
  • the diagnostic method using the anti-MOCS 3 antibody of the present invention includes, for example, (a) a step of bringing a subject-derived biological sample into contact with an antibody against the MOCS 3 protein, and (b) the sample.
  • the labeled anti-MOCS 3 antibody is used to detect and Z or quantitate the binding of MCS3 protein or a fragment thereof to the anti-MOCS 3 antibody. .
  • subject-derived biological sample refers to a subject-derived tissue, cell, or body fluid (eg, blood (including whole blood, plasma, serum, etc.), urine, lymph fluid, saliva, sweat, semen Etc.)
  • a “subject” is usually a human subject who has or is expected to undergo a cancer screening, such as a human subject who has or is suspected of having cancer. included .
  • cancers are colon cancer, stomach cancer, lung cancer, breast cancer, prostate cancer, esophageal cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterus.
  • cervical cancer endometrial cancer
  • testicular cancer thyroid cancer, knee cancer, ovarian cancer, brain tumor, blood tumor, etc. preferable.
  • Immunoassays for detecting the expression of MOCS 3 in a biological sample derived from a subject as described above are based on the ability to suspect cancer (eg, colorectal cancer), and from a subject who is at risk for cancer.
  • the method includes contacting the collected biological sample with an anti-MOCS 3 antibody under conditions that cause binding of a specific antigen and an antibody, and then measuring the amount of immunospecific binding by the antibody. Using such antibody binding, the presence and Z or increased expression of the MOCS 3 protein is detected. In this case, detection of increased MOCS 3 protein expression is an indicator of disease state. If necessary, compare the level of MOCS 3 protein in the biological sample with that of healthy individuals without cancer. Also good.
  • a biological sample such as a serum sample is contacted with a solid support or carrier such as nitrocellulose for the purpose of immobilizing all proteins present in the sample.
  • the support is then washed with buffer and subsequently treated with detectably labeled anti-MOCS 3 antibody.
  • the solid support is then washed twice with buffer to remove unbound antibody.
  • the amount of bound antibody on the solid support is measured according to well-known methods.
  • the detection conditions suitable for each measurement can be appropriately determined by those skilled in the art using conventional test methods.
  • the antibody is conjugated to an enzyme, such as that used in the enzyme immunoassay (EIA) [Voi 1 er, “The Enzyme-Linked Immunosorbent Assay” (ELISA), 1 9 7 8, Diagnostic Holyzons, 2: 1 ⁇ 7, M icrobiological A ssociates Quarterly Pub 1 ication, Walkersvi 1 1 e.MD; V. oi 1 er, A. by J. Clin. Pathol., 3 1: 5 0 7-5 2 0, 1 9 7 8: B Utier, J. E. by Me t.
  • EIA enzyme immunoassay
  • Enzymes that bind to antibodies can be visualized by, for example, spectrophotometric measurements.
  • an appropriate substrate, preferably chromogenic Enzymes that can be used to attach detectable labels to antibodies include, but are not limited to, peroxidase and alkaline phosphatase. This can be achieved by a colorimetric method using a chromogenic substrate.
  • radioimmunoassay RIA
  • sandwich immunoassay immunometric method
  • FIA fluorescence immunoassay
  • T RFIA time-resolved fluorescence immunoassay
  • EIA enzyme immunoassay
  • LIA luminescence immunoassay
  • ELIA electrochemiluminescence immunoassay
  • latex agglutination method immunoprecipitation Atssey, sedimentation reaction method, gel diffusion sedimentation reaction method, immunology Examples include a diffusion assay, an agglutination assay, a complement binding assay, an immunoradiometric assay, a fluorescence immunoassay, and an immunoassay selected from the group consisting of a protein A immunoassay (WO 0 0/1 42 2 7 gazette page 3 9 line 25 to page 4 2 line 8 line EP 1 1 1 1 1 04 7 A 2 paragraph [0 1 1 5] page 1 9 page 3 Line 5 to page 20, line 47
  • various diseases associated with MO CS 3 protein dysfunction can be diagnosed by using the in vivo MOCS 3 protein quantification method using the antibody of the present invention.
  • an increase in the concentration of MOC S 3 protein is detected, it may be caused by, for example, a disease caused by overexpression of MO CS 3 protein (eg, cancer (eg, colorectal cancer)).
  • a disease caused by overexpression of MO CS 3 protein eg, cancer (eg, colorectal cancer)
  • the anti-MCS3 antibody of the present invention can also be used for diagnosis by i n v i v o.
  • the preparation and use of antibody preparations that can be used herein are well known in the art.
  • antibody-chelating agents are described in Nuc 1. Med. Biol. 1 9 9 0 1 7: 2 47-2 54.
  • An antibody having a paramagnetic ion as a label used in magnetic resonance imaging is described in, for example, MagnneticReso nance Medi medine 1 9 9 1 2 2: 3 39 9-342. 5.2 Diagnostic agents and diagnostic methods using polynucleotide (eg, DNA) probes
  • a probe or primer designed based on the nucleotide sequence of the MCS3 gene can be used.
  • a diagnostic method includes, for example, (a) a biological sample derived from a subject, Contacting the nucleotide sequence of the MOCS 3 gene or a fragment thereof with a polynucleotide (probe) having a base sequence capable of being pre-primed under stringent high-prediction conditions; and (b) in the sample And a step of detecting and / or quantifying the hybridization of the polynucleotide with MOCS 3 gene or a fragment thereof.
  • the DNA of MO C S 3 gene (or a gene fragment thereof) in a biological sample derived from a subject is detected and Z or quantified using the probe.
  • the length of the base sequence used as a probe is, for example, 12 bases or more, 15 bases or more, 18 bases or more, 21 bases or more, 24 bases or more, 27 bases or more, 30 bases or more, or even longer It may be a polynucleotide fragment of length.
  • the low, medium or high stringency conditions described above may be used.
  • the “base sequence capable of hybridizing under hybridization conditions stringent to the base sequence of the MOCS 3 gene or a fragment thereof” includes the base sequence of the MCS3 gene or a fragment thereof.
  • a target sequence can be detected or quantified using a known method using a specific polynucleotide probe or primer for the MCS3 gene.
  • known methods include Southern hybridization, Northern hybridization, RT-PCR method, PCR-SSCP method (Genomics, Vol. 5, 8 7 4-8 7 9 (1 9 8 9)), Proceedingsofthe National A cad emy of Sciencesofthe Un ited Statesof Am erica, Vol. 86, 2 7 6 6-2 7 70 (1 9 8 9)), FI SH method, DNA chip or array C GH (Comparative Genetic Hybridization) method, etc. Can be used. Quantitative detection can be performed by quantitative RT-PCR.
  • the array C GH method is an application of the chromosomal C GH method (Ka 1 1 ioniemi, A. eta 1. (1 9 9 2) Science 2 5 8, 8 1 8— 8 2 1).
  • a DNA chip on which genomic DNA fragments (BAC, PAC, YAC, etc.) covering the chromosomal region are spotted at high density slide the cancer-derived DNA labeled with different dyes and normal DNA. It is a method to detect the DNA copy number abnormality in cancer with high resolution by simultaneously performing hybridization on the above genomic DNA fragment and detecting its binding state (Pinke 1, D. eta 1. (1 9 9 8) N at. G enet. 2 0, 2 0 7-2 1 1).
  • the mRNA level of MC S3 in the cell is determined based on the standard gene (housekeeping gene (for example, Shaper, NL et al., J. Mammary, G land B iol. N eop 1 asia 3 (1 9 9 8) 3 1 5-3 24; Wu, YY and Rees, J. L., A cta D e rm. V enereol. 8 0 (2 0 0 0) 2-3) mRNA levels can be compared, preferably by RT-PCR.
  • the standard gene housekeeping gene (for example, Shaper, NL et al., J. Mammary, G land B iol. N eop 1 asia 3 (1 9 9 8) 3 1 5-3 24; Wu, YY and Rees, J. L., A cta D e rm. V enereol. 8 0 (2 0 0 0) 2-3) mRNA levels can be compared,
  • the target sequence (DNA, mRNA, etc.) is detected and quantified by the above-mentioned method and overexpression of the MOC S 3 gene is confirmed, for example, a disease caused by overexpression of MOC S 3 (for example, (Eg, colorectal cancer, cervical cancer)) or is likely to be affected in the future.
  • a disease caused by overexpression of MOC S 3 for example, (Eg, colorectal cancer, cervical cancer)
  • the present invention also provides a kit for detecting and / or quantifying a MOCS 3 protein or a fragment thereof in a body fluid sample of a subject containing an anti-MOCS 3 antibody as a cancer marker. Furthermore, the MOCS 3 gene or a fragment thereof in a biological sample derived from a subject, which contains a base sequence that can be hybridized under stringent high prehybridization conditions to the MOCS 3 gene or a part of the base sequence thereof, is cancerated. It also provides kits for detection and Z or quantification as markers. These kits are used to detect cancer cancer by the above-described immunological technique or hybridization method.
  • cancers examples include colon cancer, stomach cancer, lung cancer, breast cancer, prostate cancer, esophageal cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterus.
  • Cancer eg cervical cancer, endometrial cancer
  • testicular cancer thyroid cancer, knee cancer, ovarian cancer, brain tumor, blood tumor, etc. preferable.
  • cancer marker refers to a subject's body fluid (eg, blood, urine, lymph fluid, saliva, sweat, semen, etc.) or cells or tissues that are not derived from normal tissue, or A molecule that is selectively upregulated in cancer cells or tissues, and that the presence of the molecule in the body fluid or cells or tissues of a subject indicates or indicates the presence of cancer.
  • body fluid eg, blood, urine, lymph fluid, saliva, sweat, semen, etc.
  • a molecule that is selectively upregulated in cancer cells or tissues indicates or indicates the presence of cancer.
  • the kit according to the first aspect contains a component for detecting and / or quantifying the M O C S 3 antigen (including M O C S 3 protein and its partial peptide) in a body fluid sample from a subject.
  • a component for detecting and / or quantifying the M O C S 3 antigen including M O C S 3 protein and its partial peptide
  • MOCS 3 protein is detected and Z or quantified by ELISA
  • Such components detect and Z or quantitate the level of MOCS 3 in, for example, tissue sections or body fluid samples such as blood and urine Can be used for.
  • Such antibodies may be labeled with radioactivity, fluorescence, colorimetry, or enzyme labeling.
  • the kit of the present invention may contain a labeled secondary antibody.
  • the kit according to the second aspect is a hybridizer under high-precipitation conditions stringent to the MOCS 3 gene or a part of its nucleotide sequence.
  • a polynucleotide comprising a base sequence that can be synthesized is contained.
  • the kit of the present invention may contain the above polynucleotide immobilized on a DNA chip.
  • the kit of the present invention is typically a container in addition to an anti-MOC S 3 antibody, MCS3 gene or a part of the base sequence that can be hybridized under stringent high-precipitation conditions. And label.
  • the label on or with the container may indicate that the drug is used to detect a colorectal cancer marker.
  • other items such as instructions for use, labeled secondary antibodies, and the like may be further included.
  • Example 1 Identification of colon cancer specific amplification gene by array C GH method
  • sample preparation of 200 colorectal cancer specimens and verification based on the array CGH method were performed.
  • Fig. 1 is a histogram showing the frequency of the MCS3 gene relative to the degree of amplification in 200 colorectal cancer patients.
  • Table 2 shows the degree of amplification (GZR value) and frequency of 200 samples of colon cancer patients with MOC S 3 gene. The average value is the average value for samples with a GZR value of 1.2 or higher. is doing.
  • Example 2 Verification of the degree of gene amplification in a colon-derived cultured cell line
  • the degree of amplification in a colon cancer-derived cultured cell line was verified for a high-frequency gene region in colon cancer patients.
  • the cultured cell line used was RKOE 6, Caco 2, which is a colorectal cancer-derived cell line.
  • Genomic DNA was extracted from the cultured cells according to the protocol attached to the kit using Blood & Cel lCu lure DNA Kit (QIAGEN).
  • Table 3 shows the degree of amplification (GZ R value) of the MCS3 gene in a cell line derived from colorectal cancer. As shown, in the colon cancer-derived cell line, it was found that amplification occurred in the MCS3 gene located in BAC Clone RP 1 1-3 79 D 4. (Table 3)
  • Quantitative PCR was performed to confirm the amplification of the MOCS3 gene region. Quantitative PCR was performed using S YB RG reen RT—PCRR eaents Applied B iosys terns) according to the attached protocol. ied Biosyst ern s). Primers were synthesized using the following sequences (commissioned to OPERON).
  • Table 4 shows the degree of amplification of the MO CS 3 gene in a colon cancer-derived cell line. Values are relative to control DNA (normal). As shown, in the colon cancer-derived cell line, it was found that amplification occurred in the MOCS 3 gene region. (Table 4)
  • the MO C S 3 gene which was frequently amplified in 200 colorectal cancer patients, is a colorectal cancer-derived cell line, and
  • RNAi analysis was performed and the phenotype was observed.
  • the cell line was purchased from ATCC and cultured according to the attached protocol.
  • siRNA we selected specific 21mer in the gene and synthesized siRNA targeting the sequence (commissioned to QIAGEN).
  • siRNA introduction into RKOE 6 cells Oligofectamine (Invitrogen) was used, and ⁇ ⁇ ⁇ ⁇ si RNA was introduced into the cells according to the attached protocol. N egative control RNA (QI AGEN) was used as a control. The cells were observed under an inverted microscope for 4 days after introduction into the cells.
  • cDNA was synthesized according to the attached protocol by using Su pe r s c r i p t I I I F i r s t—S t r a n d S y n t h e s i S S y s t em f o r RT—P C R (I n v i t ro g e n).
  • Quantitative RT-PCRR was performed using this cDNA as a saddle. Quantitative PCR was performed using S YB RG reen RT— PCRR eaents (A p ⁇ 1 ied B iosys t ern s), according to the attached protocol. 7 5 0 0 R eal — T i me PCRS yst ern ( (Applied Biosys terns)). Primer 1 synthesized the following sequence (consigned to O P ERON) and used it.
  • GAPDH G lyceraldehyde— 3—phosphatedehydrogenas e
  • Control Reagents Ap plied B ios yst em s was used to determine the expression level of GAPDH and the relative ratio was calculated
  • si RN A The number of viable cells after introduction of A 1 Amar Blue (Biosource) and Wa 1 1 ac 1 4 2 0 Mu ltilabel / L um inescence Coutor ARVO (P erkin E l me r). Result
  • RNAi analysis of the MOC S 3 gene was performed using RKO E 6 which is a cell line derived from colorectal cancer.
  • Fig. 3 shows the results of quantitative RT-PCRR after 24 hours of siRNA of the MOCS3 gene in RK o E6 cells and harvesting the cells 24 hours later.
  • the relative amount of N C was shown using the relative ratio of GAP DH.
  • the effect of RNA level was confirmed by quantitative RT-PCR.
  • the expression level of all three types of siRNA was significantly reduced (Fig. 3).
  • Fig. 4 shows the results of measuring the number of viable cells with the measurement reagent using the cells on the 4th day after Transfection of MO CS 3 gene siRNA to RKO E6 cells.
  • the graph shows the relative amount to NC.
  • the number of viable cells was measured, and as with the phenotype, the number of cells was significantly reduced compared to N egative Control (NC) ( ( Figure 4)
  • All three siRNAs (a, b, c) of the MOC S 3 gene were significant (p ⁇ 0. 0 1) in the t-test.
  • Example 4 Functional analysis by RNAI analysis using a colon-derived normal cell line
  • the RNAI analysis was performed using a cell line derived from a normal tissue of the large intestine. Has been verified to be cancer specific.
  • the cell line used was C CD 1 8 CO 2 purchased from AT C. The culture conditions were in accordance with the attached protocol. The a sequence of Example 3 was used as the siRNA used.
  • L i p o f e c t a min e 20 00 (Inv i t r o g e n) was used, and 25 n M of siRNA was introduced into the cells according to the attached protocol.
  • N at i v e C o n t r o ls i RNA Q I AG E N
  • the cells were observed under an inverted microscope for 5 days after introduction.
  • RNA i The effects of RNA i on the MO C S 3 gene in the cell line C CD 18 C o derived from normal colon tissue are as follows.
  • FIG. 5 shows an observation image obtained on the 5th day after Transfection of MOC S3 gene siRNA on CD 18 Co cells (upper: X 40; lower: X 2 200).
  • a, b, and c are the si RNs of the MOC S 3 gene A 3 types, NC indicates negative control. As shown, phenotypic observations were almost the same as NC, and no effect was observed (Figure 5).
  • Figure 6 shows the results of quantitative RT-PCR after cells were collected 24 hours after transcription of siCNA3 of McCS3 gene to CCD18CO cells.
  • the relative amount to NC was shown using the contrast of GAPDH.
  • Figure 7 shows that after the transfection of the siRNA of the MOC S 3 gene in CCD18 CO cells.
  • Fig. 5 shows the results of measuring the number of viable cells with the measurement reagent using the cells on day 5.
  • the graph shows the relative amount with respect to NC, and as in the phenotype, it is almost the same as that of Negative Control (NC). No effect was observed (Fig. 7).
  • the organ-specific expression of the M0CS3 gene in normal tissues was performed by the Northern hyperprecipitation method known in the art.
  • Fig. 8 is a photograph showing the results of Northern high pri- dialysis performed using various normal organ tissues. As shown in the figure, a band was observed around 2.5 kb in the testis, and the expression level of other organs was confirmed to be small. The expression level is small in normal tissues and testis-specific, so it can be estimated that the effects of drugs targeting this gene are small on normal tissues.
  • Example 6 Evaluation of gene amplification
  • the FISH method was used to evaluate the occurrence of gene amplification in the M0CS3 gene region in cancer cells of the specimen tissue. result
  • Figure 9 is an optical micrograph (fluorescence image) of a portion of the observed cancer cells (for 6 cells). As shown, more than 3 spots of signal were found in cancer cells. It was confirmed that the M0CS 3 gene region was amplified in 10 specimens whose gene amplification degree (G / R) was 1.2 or more by the array CGH method. It was also shown that gene amplification occurred from the early stage to the late stage of the disease state. This indicates that the M0CS3 gene region can be applied not only as a molecular target for cancer therapeutics, but also for cancer diagnosis by the FISH method.
  • Example 7 Evaluation of cancer specificity
  • the M0CS 3 gene was expressed in cancer specimen tissues by immunostaining methods known in the art.
  • FIG. 10 shows the results of immunohistochemical evaluation of the expression of M0C S 3 gene protein in cancer cells of the specimen tissue. As shown in the figure, it was confirmed that it was expressed specifically in the cancerous part. Staining was observed in epithelial cells with irregularly-structured overlapping nuclei in poorly differentiated glandular cancer. On the other hand, there was no staining in the lamina intestinal gland where the cells were well-ordered.
  • the M0CS3 gene knockdown showed a growth-inhibiting effect.
  • the effect of the cervical cancer cell line was evaluated by the RNAi analysis method described above.
  • RNAi analysis was carried out in the same manner as described in Example 3 using the above three siRNAs (Example 3: Table 5).
  • siRNA Oligofeminte (Invitrogen) was used, and siRNA of ⁇ was introduced into the cells according to the attached protocol.
  • siRNA negative RNA Co nt r o 1 si RNA (Q I AGEN) was used. Result
  • Fig. 11 shows the results of measuring the number of viable cells with the measuring reagent using the cells on the 4th day after Transfectioion of MCS3 gene siRNA on HeLa cells.
  • the graph shows the relative amount to NC.
  • siRNAs (a, b, c) of MCS3 gene 15% of siRNA b and 25% of siRNA c were inhibited. Significant in 0.01 t test).
  • Serum specimens of colorectal cancer patients 10 L and healthy subject serum specimens 10 / xL were diluted with 500 L of diluted buffer solution (lOmM Tris HC1 ph 7.4 + 150 mM NaCl), then ProteomeLab IgY-12 SC Proteome partitioning kit (BECKMAN COULTER: A24618) was used to remove serum abundant proteins such as albumin and globulin. Dithiothreitol (Wako: 049_08972) was added to the obtained fraction to a final concentration of 10 mM, and the reduction reaction was performed at 60 ° C for 30 minutes.
  • odoacetamide SIGMA: 144-48-9
  • SIGMA 144-48-9
  • the alkylation reaction was carried out at room temperature for 30 minutes in the light-shielded state.
  • add 4 times the volume of cold acetone Wako: 014-08681, -20 ° C
  • the recovered protein was dissolved in 80 zL of 2M urea + lOOmM ammonium bicarbonate solution. After dissolution, a portion was subjected to protein concentration measurement by the BCA method. Trypsin (Promega: V511C) was added to the sample protein at 1/50 (w / w), and digestion was performed at 37 ° C for 16 hours.
  • the sample separated by Nano-HPLC is PicoTip (New Object: FS360-20- 10-D-20) and introduced continuously into an ion trap mass spectrometer directly connected.
  • HCT Plus Bruker Dal tonics
  • the sample was ionized under the conditions of a capillary voltage of 1500 V, an end plate offset value of 500 V, a dry gas flow rate of 12 L / min, and a dry gas temperature of 250 ° C.
  • the ion trap was set to capture 2 Da before and after the target m / z, and MS / MS analysis was performed in MRM mode.
  • Figures 13 A and B show the results of analysis of the above methods for (A) serum from colon cancer patients and (B) serum from healthy subjects, respectively.
  • Figures 14A to C show the correspondence between the peaks shown in Figure 13 and amino acids (or amino acid sequences) determined by MS / MS analysis.
  • an ion peak corresponding to the fragment ion of the target molecule was clearly observed in the serum from colon cancer patients. That is, as shown in FIG. 14B, a partial sequence called EPSL LQ is determined at least from the C-terminal side. Since it corresponds to the amino acid sequence at position 3, the target peptide fragment (SEQ ID NO: 1 3) at amino acid sequences 3 2 6 to 3 3 5 was identified. Such an ion peak corresponding to the partial sequence was not found in the analysis using the serum of healthy subjects (see Fig. 14C). Therefore, it is strongly suggested that this target molecule (ie, MO CS 3 protein) may have increased cancer abundance specifically in cancer.
  • Example 10 Evaluation of specificity in various cancer types
  • Whether the M0CS3 gene was expressed in organ cancers other than colorectal cancer was determined by an immunostaining method known in the art.
  • Immunohistochemical staining was performed with anti-M0CS3 gene polyclonal antibody using Multiple organ cancer tissue microarray for testing tumor tissue specificity of therapeutic antibodies (US Bioma), in which 96 samples of cancer tissues from 35 organs were placed. .
  • the immunohistochemical staining method was revised to YamasMta S & Okada Y. (2005) J Histochem Cytochem. Nov; 53 (11): 1421-32 for the activation method, and Watanabe / Nakano enzyme antibody method was revised for the staining method. According to the 4th edition (interdisciplinary planning). Usagi IgG was used as a negative control. MM.
  • Fig. 15-1 and Fig. 15-2 show the expression of the stomach (A: stomach), colon (B: colon), rectum (C: rectum), retina (D: epiploon). ), Lung (E, F, G: lung), ovary (I, J: ovary), prostate (L: prostate), lymph node (M: Lymp node), bone tissue (O: bone), and mediastinum ( R: mediastinum).
  • H milk
  • spleen K: spleen
  • throat P
  • Q adipose tissue
  • M0CS3 is upregulated in a wide range of cancer types. This suggests that M0CS3 has shown cancer specificity as a molecular target for cancer therapeutics, and can be applied to a wide range of cancer types, not just colorectal cancer. It was. This also applies to cancer diagnosis, suggesting the significant effectiveness of M0CS3.
  • the present invention provides a therapeutic agent for cancer, a diagnostic agent, a diagnostic method, a therapeutic method, and a kit used therefor. Therefore, the present invention is useful in fields such as cancer diagnosis or targeted therapy.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Cell Biology (AREA)
  • Oncology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Microbiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Food Science & Technology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

L’invention concerne un agent thérapeutique pour le cancer qui comprend un inhibiteur de l'expression ou de l’activation de la protéine MOCS3 ; une méthode pour dépister un composé pouvant être utilisé comme principe actif de l'agent thérapeutique ; un anticorps dirigé contre la protéine MOCS3 ; un agent ou une méthode de diagnostic du cancer utilisant l'anticorps ou quelque chose de similaire ; et autres.
PCT/JP2006/317705 2005-08-31 2006-08-31 Utilisation du gène mocs3 à des fins thérapeutiques ou diagnostiques WO2007026960A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2007533382A JPWO2007026960A1 (ja) 2005-08-31 2006-08-31 Mocs3遺伝子の治療的又は診断的用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2005252304 2005-08-31
JP2005-252304 2005-08-31

Publications (1)

Publication Number Publication Date
WO2007026960A1 true WO2007026960A1 (fr) 2007-03-08

Family

ID=37809034

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2006/317705 WO2007026960A1 (fr) 2005-08-31 2006-08-31 Utilisation du gène mocs3 à des fins thérapeutiques ou diagnostiques

Country Status (2)

Country Link
JP (1) JPWO2007026960A1 (fr)
WO (1) WO2007026960A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2955235A3 (fr) * 2009-01-30 2016-03-02 Koninklijke Philips N.V. Procédés de sous-classification de tumeurs du sein

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005054461A2 (fr) * 2003-11-26 2005-06-16 Whitehead Institute For Biomedical Research Regulateurs de transcription et procedes associes

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005054461A2 (fr) * 2003-11-26 2005-06-16 Whitehead Institute For Biomedical Research Regulateurs de transcription et procedes associes

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CANNELLA C. ET AL.: "Polyclonal antibodies to bovine rhodanese", ITALIAN JOURNAL OF BIOCHEMISTRY, vol. 37, no. 4, 1988, pages 235A - 236A, XP003003875 *
MATTHIES A. ET AL.: "Molybdenum Cofactor Biosynthesis in Humans: Identification of a Persulfide Group in the Rhodanese-like Domain of MOCS3 by Mass Spectrometry", BIOCHEMISTRY, vol. 44, no. 21, 2005, pages 7912 - 7920, XP003003876 *
MERRILL G.A. ET AL.: "Detection of timer-dependent and oxidatively induced antigens of bovine liver rhodanese with monoclonal antibodies", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 263, no. 36, 1988, pages 19324 - 19330, XP003003874 *
PALLINI R. ET AL.: "A role for the mitochondrial tiosulfate sulfurtransferase (rhodanese) in tumor cells", ACTA MEDICA ROMANA, vol. 30, no. 3-4, 1992, pages 225 - 228, XP003003877 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2955235A3 (fr) * 2009-01-30 2016-03-02 Koninklijke Philips N.V. Procédés de sous-classification de tumeurs du sein

Also Published As

Publication number Publication date
JPWO2007026960A1 (ja) 2009-03-12

Similar Documents

Publication Publication Date Title
US20060110397A1 (en) AIMP2-DX2 and its uses
CA3020650C (fr) Kinase ros dans le cancer du poumon
JP2008517590A (ja) 癌の拡張及び/又は転移の発生の識別、診断、防止又は治療におけるスラグ遺伝子又はその複製、転写又は発現生成物を使用する使用方法
EP2452948B1 (fr) Nouveau marqueur tumoral
KR101374667B1 (ko) 혈소판 유래 성장 인자 수용체-알파를 발현하는 비-소 세포폐암 종양들의 동정
KR101928618B1 (ko) Tk1 단백질의 발현이 항진된 결장직장암 환자에 대한 치료효과 예측 방법
WO2011129427A1 (fr) Agent de diagnostic et agent thérapeutique pour les cancers
SG187674A1 (en) Bard1 isoforms in lung and colorectal cancer and use thereof
US9789159B2 (en) Diagnosis and treatment of prostate cancer
US8389684B2 (en) Tumor biomarker
JP5354634B2 (ja) ヒトabh8タンパク質、それをコードする遺伝子、およびこれらの治療的又は診断的用途
WO2007026960A1 (fr) Utilisation du gène mocs3 à des fins thérapeutiques ou diagnostiques
JP6341859B2 (ja) がんマーカーおよびその用途
WO2007037538A9 (fr) Application therapeutique ou diagnostique du gene spo11
JP6312861B2 (ja) がん発症の予測、早期診断、及び治療剤としてのnkx6.3の用途
JPWO2007037560A1 (ja) Sgk2遺伝子の治療的又は診断的用途
WO2007037532A1 (fr) Application therapeutique ou diagnostique du gene srms
WO2007037533A9 (fr) Application therapeutique ou diagnostique du gene ppp1r3d
AU2014203702B2 (en) New tumor marker
WO2007037555A9 (fr) Application therapeutique ou diagnostique du gene dusp15
WO2007037550A9 (fr) Application therapeutique ou diagnostique du gene tsta3
JPWO2005021739A1 (ja) Nox1ポリペプチドに対する抗体、Nox1遺伝子を利用したガン診断方法、及びガン増殖抑制物質のスクリーニング方法
JP2010166873A (ja) Pomp遺伝子およびpsma7遺伝子の治療的又は診断的用途
Chiriboga et al. Stromal Cell–Derived Factor-1A and CXCR4 Expression in Hemangioblastoma and Clear Cell-Renal Cell Carcinoma: von Hippel-Lindau Loss-of-Function Induces Expression of a Ligand and Its Receptor

Legal Events

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

Ref document number: 2007533382

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 06797581

Country of ref document: EP

Kind code of ref document: A1