WO2004000346A1 - Preventives/remedies for cancer - Google Patents

Preventives/remedies for cancer Download PDF

Info

Publication number
WO2004000346A1
WO2004000346A1 PCT/JP2003/007926 JP0307926W WO2004000346A1 WO 2004000346 A1 WO2004000346 A1 WO 2004000346A1 JP 0307926 W JP0307926 W JP 0307926W WO 2004000346 A1 WO2004000346 A1 WO 2004000346A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
cancer
amino acid
acid sequence
present
Prior art date
Application number
PCT/JP2003/007926
Other languages
French (fr)
Japanese (ja)
Inventor
Yuichi Hikichi
Ryosuke Katsuyama
Yuichi Kakoi
Original Assignee
Takeda Chemical Industries, 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 Takeda Chemical Industries, Ltd. filed Critical Takeda Chemical Industries, Ltd.
Priority to AU2003243951A priority Critical patent/AU2003243951A1/en
Publication of WO2004000346A1 publication Critical patent/WO2004000346A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • the present invention relates to an agent for preventing and treating cancer, a diagnostic agent, and the like.
  • a microarray method in which cDNA or oligonucleotides are immobilized has been developed. Techniques for finding disease-specific changes in gene expression have become widespread, and their usefulness has been confirmed. For example, Af fymetrix's GeneChip system is being widely used for diagnosing diseases such as cancer and finding drug discovery target genes.
  • antisense oligonucleotides hybridize to RNAs with complementary sequences and induce RNase H degradation of RMs to inhibit protein translation, or to inhibit direct protein synthesis by hybridization. Bring. Because it is possible to specifically suppress the function of the target gene, it is frequently used as a means of analyzing the function of genes, and some antisense oligonucleotides are being developed for clinical application.
  • RIPK1 Interacting Serine / Threonine Kinase 1 (hereinafter referred to as RIPK1) was discovered, but a molecule showing homology to this RIPK1, RIPK2, has recently been cloned (J. Biol. Chem. 273). , 12296-12300, 1998). Subsequently, RIPK2 binds to apoptosis-related molecules such as Caspase and c IAP, and also binds RIPK2 to MCF7 and 293T. It has been reported that apoptosis is induced by forced expression in cells (J. Biol. Chem. 273, 12296-12300, 1998 and J. Biol. Chem. 273, 16968-16975, 1998).
  • RIPK2 is a molecule that acts downstream of Toll-like Receptor (TLR) -2, 3, 4 and is involved in innate immunity ( Nature 416, 190-194, 2002; Nature 416, 194-199, 2002).
  • TLR Toll-like Receptor
  • the present inventors have conducted intensive studies to solve the above problems, and as a result, have found the RIM2 gene as a gene whose expression is significantly increased in cancer tissues, particularly malignant cancer tissues. Furthermore, they found that apoptosis was induced by suppressing the function of the RIPK2 gene, which has been considered to be a factor that induces apoptosis.Based on this finding, the inventors conducted further studies and completed the present invention. Reached.
  • a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a compound that inhibits the activity of a salt thereof, or a salt thereof Prevention and treatment of cancer
  • a prophylactic or therapeutic agent for cancer comprising the antisense polynucleotide according to (3) above,
  • Cancer is colorectal, breast, lung, prostate, esophagus, stomach, liver, biliary, spleen, kidney, bladder, uterus, ovary (1), (2), cancer, testis, thyroid, thyroid, brain or hematological
  • the cancer is breast cancer, (1), (2), (5) or
  • a diagnostic agent for cancer comprising an antibody against a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof;
  • a diagnostic agent for cancer comprising a polynucleotide encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof,
  • Cancer is colorectal, breast, lung, prostate, esophagus, stomach, liver, biliary, spleen, kidney, bladder, uterus, ovary (8) or cancer, testicular cancer, thyroid cancer, kidney cancer, brain tumor or blood tumor
  • a prophylactic / therapeutic agent for cancer comprising a compound having a Receptor-Interacting Serine / Threonine Kinase 2 activity inhibitory activity or a salt thereof,
  • a prophylactic / therapeutic agent for cancer comprising a compound having an inhibitory effect on the expression of Receptor-Interacting Serine / Threonine Kinase 2 or a salt thereof,
  • a prophylactic or therapeutic agent for cancer characterized by using a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof; Screening method,
  • a method for screening a pharmaceutical compound which comprises using a polynucleotide encoding a protein or a partial peptide thereof, (15b) a compound for preventing or treating cancer, The method according to (15a) above, which is a compound used for the prevention and treatment of Z and a compound having Z or cancer prevention and treatment effects.
  • An agent for preventing or treating cancer which can be obtained by using the screening method according to (13) or (15) or the screening kit according to (14) or (16).
  • an agent for preventing or treating cancer comprising the compound or salt thereof according to (17b) above, (18) a protein or a partial peptide which has the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a compound which inhibits the activity of the salt or a salt thereof Apoptosis action promoter,
  • a method for screening an apoptosis-activating agent which comprises using a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof.
  • a compound that inhibits the activity of a protein, or a partial peptide or a salt thereof, having the same or substantially the same amino acid sequence as that represented by SEQ ID NO: 1 in a mammal; Or a salt thereof, or a compound that inhibits the expression of a gene of the protein or an effective amount of a salt thereof, comprising the steps of: • (25) inhibits the activity of a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof, or expresses a gene of the protein Cancer prevention and treatment methods characterized by inhibiting
  • a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 used in the present invention (hereinafter sometimes referred to as the protein of the present invention or the protein used in the present invention) are cells of human warm-blooded animals (eg, guinea pig, rat, mouse, chicken, egret, bush, hidge, horse, monkey, etc.) (eg, hepatocytes, spleen cells, nerve cells, glial cells, Glands i3 cells, bone marrow cells, mesangial cells, Langerhans cells, epidermal cells, epithelial cells, goblet cells, endothelial cells, smooth muscle cells, fibroblasts, fibrocytes, muscle cells, fat cells, immune cells (eg, Macrophages, T cells, B cells, natural killer cells, obesity cells, neutrophils, basophils, eosinophils, monocytes), megakaryocytes, synovial cells, cartilage Cells,
  • progenitor cells stem cells or cancer cells, etc.
  • any tissue where these cells are present such as the brain, various parts of the brain (eg, olfactory bulb, amygdala, basal sphere, hippocampus, thalamus, hypothalamus, Cerebral cortex, medulla, cerebellum), spinal cord, pituitary, stomach, kidney, kidney, liver, gonad, thyroid, gall bladder, bone marrow, adrenal gland, skin, muscle, lung, digestive tract (eg, large intestine, small intestine), blood vessels , Heart, thymus, spleen, submandibular gland, peripheral blood, prostate, testis, ovary, placenta, uterus, bone, joints, skeletal muscle, etc., or a synthetic protein .
  • olfactory bulb amygdala, basal sphere, hippocampus, thalamus, hypothalamus, Cerebral cortex, medulla, cerebellum
  • amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 is about 50% or more, preferably about 60% or more, and more preferably about 50% or more of the amino acid sequence represented by SEQ ID NO: 1.
  • Amino acid sequence homology can be calculated using a homology calculation algorithm NCBI BLAST (National Center for Biotechnology Information Basic Local Search Tool).
  • Examples of the protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 include, for example, a protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 described above. However, a protein having substantially the same activity as the protein containing the amino acid sequence represented by SEQ ID NO: 1 is preferable. Examples of substantially equivalent activities include, for example, signal transduction activity (eg, RIPK2 intracellular signal transduction activity), phosphorylation activity, and the like. Substantially identical indicates that the properties are qualitatively (eg, physiologically or pharmacologically) identical.
  • the signal transduction activity and the phosphorylation activity are equivalent (eg, about 0.01 to 100 times, preferably about 0.1 to 10 times, more preferably 0.5 to 2 times).
  • the quantitative factors such as the degree of these activities and the molecular weight of the protein may be different.
  • the signal transduction activity and phosphorylation activity can be measured by a method known per se, for example, the method described in J. Biol. Chem. 273, 16968-16975, 1998 or a method analogous thereto. . RIPK 2 is autophosphorylated in signaling Activates the NF KB signal.
  • phosphorylation activity can be determined by introducing an expression vector for the protein of the present invention (eg, RI PK2) into cells, solubilizing the cells with an appropriate solubilization buffer, and immunoprecipitation. After obtaining a protein (eg, RI PK2), it is reacted with radiolabeled ATP in an appropriate buffer, and the reaction mixture is subjected to SDS gel electrophoresis, and the protein of the present invention is obtained by autoradiography or the like. (Eg, RI PK 2) can be measured for the degree of autophosphorylation.
  • the signal transduction activity can be determined by introducing an expression vector for the protein of the present invention (eg, RI PK2) into a cell together with a repo overnight gene containing an NF ⁇ B binding sequence in a promoter or an enhancer.
  • an expression vector for the protein of the present invention eg, RI PK2
  • a repo overnight gene containing an NF ⁇ B binding sequence in a promoter or an enhancer.
  • a liquid containing a signal transduction activity or a suppressive substance such as a microorganism cell disruption solution, a microorganism culture supernatant, a eukaryotic cell disruption solution, or a eukaryotic cell culture supernatant is added, and the reporter expression level is known per se.
  • the repo overnight expression level can be measured using, for example, a reporter protein activity using luciferase activity, alkaline phosphatase activity, or the like as an index.
  • Examples of the protein used in the present invention include: (1) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 1 (for example, about 1 to 100, preferably about 1 to 30, An amino acid sequence in which about 1 to 10 amino acids have been deleted, more preferably a number (1 to 5) of amino acids, and 2 or more amino acids in the amino acid sequence represented by SEQ ID NO: 1 (for example, 1 to 1) An amino acid sequence to which about 00, preferably about 1 to 30, preferably about 1 to 10, and more preferably a number (1 to 5) of amino acids are added; 3 represented by SEQ ID NO: 1
  • the amino acid sequence contains one or more (for example, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, and more preferably about 1 to 5) amino acids.
  • Inserted amino acid sequence, 4 1 in the amino acid sequence represented by SEQ ID NO: 1 Or two or more (for example, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, and more preferably the number (1 to 5)) of other amino acids
  • So-called muteins such as proteins containing an amino acid sequence substituted with or an amino acid sequence obtained by combining them are also included.
  • the amino acid sequence is inserted, deleted or substituted as described above, the position of the insertion, deletion or substitution is not particularly limited.
  • the protein has a N-terminus at the left end (amino terminus) and a C-terminus at the right end (capillary end) according to the convention of peptide labeling.
  • the protein used in the present invention including the protein containing the amino acid sequence represented by SEQ ID NO: 1, has a C-terminal lipoxyl group (-C00H), carboxylate (-C00-), amide (- It may be either C0NH 2 ) or ester (-C00R).
  • R in the ester e.g., methyl, Echiru, n- propyl, isopropyl, C, such as n- butyl, _ 6 alkyl groups, such as cyclopentyl, C 3 _ 8 cycloalkyl group such as cyclohexyl cyclo, for example, phenyl, alpha-naphthyl of which C 6 _ l2 7 aryl group, e.g., benzyl, one Nafuchiru C, such as phenylene Lou C Bok 2 ⁇ alkyl group or alpha-naphthylmethyl, such as phenethyl, such as _ 2 alkyl groups C 7 _ 14 Ararukiru group, such as pin bar opening Iruokishimechiru group is used.
  • C such as n- butyl
  • _ 6 alkyl groups such as cyclopentyl
  • a protein in which the carboxyl group is amidated or esterified is also included in the protein used in the present invention.
  • the ester in this case, for example, the above-mentioned C-terminal ester and the like are used.
  • the protein used in the present invention the amino acid residue (e.g., main Chionin residues) of N-terminal Amino group protecting groups (e.g., formyl group, _ 6 Ashiru groups such as C Myu6 Arukanoiru such Asechiru group ), N-terminal dalluminin residue generated by cleavage in vivo, oxidized with lipamine, substituents on the side chains of amino acids in the molecule (eg-0H , -SH, amino group, imidazole group, I Ndoru group, in Guanijino group, etc.) a suitable protecting group (e.g., formyl group, C such Asechi group, such as C WINCH 6 Ashiru groups such as _ 6 Arukanoiru group)
  • a suitable protecting group e.g., formyl group, C such Asechi group, such as C WINCH 6 Ashiru groups such as _ 6 Arukanoiru group
  • protein used in the present invention include, for example, protein containing an amino acid sequence represented by SEQ ID NO: 1.
  • the partial peptide of the protein used in the present invention examples include: It is a partial peptide of the protein used, and preferably may be any peptide having the same properties as the protein used in the present invention described above. Specifically, for the purpose of preparing the antibody of the present invention described below, a peptide having the 27th to 578th amino acid sequence in the amino acid sequence represented by SEQ ID NO: 1 and the like can be mentioned. For example, at least 20 or more, preferably 50 or more, more preferably 70 or more, more preferably 100 or more, and most preferably 200 or more of the constituent amino acid sequences of the protein used in the present invention. Peptides having more than one amino acid sequence are used.
  • one or more (preferably about 1 to 10, more preferably a number (1 to 5)) amino acids in the amino acid sequence are deleted.
  • 1 or 2 or more (preferably, about 1 to 20, more preferably, about 1 to 10, and more preferably, about 1 to 5) amino acids are added to the amino acid sequence.
  • 1 or 2 or more (preferably, about 1 to 20; more preferably, about 1 to 10; more preferably, 1 to 5) amino acids are inserted into the amino acid sequence.
  • one or more (preferably about 1 to 10, more preferably several, and more preferably about 1 to 5) amino acids in the amino acid sequence are replaced with other amino acids. It may be.
  • the partial peptide used in the present invention may be any of the ester (-C00R).
  • the partial peptide used in the present invention includes those having a carboxyl group (or carboxylate) in addition to the C-terminal, N-terminal amino acid residues ( (E.g., methionine residue) whose amino group is protected by a protecting group, glutamine residue generated by cleavage of N-terminal side in vivo and pyrodartamine oxidation, substitution on the side chain of amino acid in the molecule is Also included are those protected with an appropriate protecting group, and complex peptides such as so-called glycopeptides to which sugar chains are bonded.
  • a carboxyl group or carboxylate
  • N-terminal amino acid residues (E.g., methionine residue) whose amino group is protected by a protecting group, glutamine residue generated by cleavage of N-terminal side in vivo and pyrodartamine oxidation, substitution on the side chain of amino acid in the molecule is Also included are those protected with an appropriate protecting group, and complex peptides such as so-
  • the partial peptide used in the present invention can also be used as an antigen for producing an antibody.
  • salts with physiologically acceptable acids eg, inorganic acids, organic acids
  • bases eg, alkali metal salts
  • Such salts include, for example, salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid) or organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid) Succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid).
  • inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid
  • organic acids eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid
  • Succinic acid tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid.
  • the protein, its partial peptide, or a salt thereof used in the present invention can be produced from the above-mentioned human or warm-blooded animal cell or tissue by a known method for purifying a protein, or a DNA encoding the protein. Can also be produced by culturing a transformant containing It can also be produced according to the peptide synthesis method described below.
  • the human or mammalian tissues or cells are homogenized, then extracted with an acid or the like, and the extract is subjected to reverse phase chromatography, ion exchange chromatography. Purification and isolation can be performed by combining chromatography such as chromatography.
  • a commercially available resin for protein synthesis can be usually used.
  • a resin include chloromethyl resin, hydroxymethyl resin, benzhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, and PAM.
  • Resin 4-hydroxymethylmethylphenylacetamidomethyl resin, polyacrylamide resin, 4- (2,, 4, dimethoxyphenylhydroxymethyl) phenoxy resin, 4- (2 ′, 4,1-dimethoxyphenylmethyl resin F moc aminoethyl) phenoxy resin.
  • an amino acid having an ⁇ -amino group and a side chain functional group appropriately protected is condensed on the resin in accordance with the sequence of the target protein according to various known condensation methods.
  • a protein or partial peptide is cleaved from the resin, and at the same time, various protecting groups are removed.
  • an intramolecular disulfide bond formation reaction is performed in a highly diluted solution to obtain the target protein or partial Obtain peptides or their amides.
  • the protected amino acid is added directly to the resin along with a racemization inhibitor (eg, HOB t, HO OB t), or as a symmetrical anhydride or H ⁇ B t ester or H ⁇ OB t ester After the protected amino acid is activated in advance, it can be added to the resin.
  • a racemization inhibitor eg, HOB t, HO OB t
  • the solvent used for activating the protected amino acid or condensing with the resin can be appropriately selected from solvents known to be usable for the protein condensation reaction.
  • solvents known to be usable for the protein condensation reaction for example,
  • Acid amides such as N, N-dimethylformamide, N, N-dimethylacetamide and N-methylvinylidone; halogenated hydrocarbons such as methylene chloride and chloroform; alcohols such as trifluoroethanol; Sulfoxides such as dimethylsulfoxide, ethers such as pyridine, dioxane and tetrahydrofuran, nitriles such as acetonitrile and propionitrile, esters such as methyl acetate and ethyl acetate, or an appropriate mixture thereof are used.
  • the reaction temperature is appropriately selected from a range that is known to be used for a protein bond formation reaction, and is usually appropriately selected from a range of about ⁇ 20 ° C.
  • the activated amino acid derivative is usually used in a 1.5 to 4-fold excess.
  • the condensation is insufficient, sufficient condensation can be performed by repeating the condensation reaction without removing the protecting group. If sufficient condensation cannot be obtained even after repeating the reaction, unreacted amino acids can be acetylated using acetic anhydride or acetylimidazole so that subsequent reactions are not affected. .
  • Examples of the protecting group for the amino group of the starting material include Z, Boc, t-pentyloxycarbonyl, isopolnyoxycarbonyl, 4-methoxybenzyloxycarbonyl, C11Z, Br-Z, a Damantyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 212-trophenylsulfenyl, diphenyl Phosphinothio oil, Fmoc, and the like are used. '
  • the lipoxyl group can be, for example, alkyl esterified (eg, methyl, ethyl, propyl, butyl, t-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.).
  • alkyl esterified eg, methyl, ethyl, propyl, butyl, t-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.
  • Alkyl esterification eg, benzyl ester, 4-nitrobenzyl ester, 4-methoxybenzyl ester, 4-chlorobenzyl ester, benzhydryl esterification), phenacyl esterification, benzyloxycarponyl It can be protected by hydrazide, t-butoxycarbonyl hydrazide, trityl hydrazide and the like.
  • the hydroxyl group of serine can be protected, for example, by esterification or etherification.
  • groups appropriately used for the esterification for example, low-grade (C ,. 6) Arukanoiru groups such Asechiru group, Aroiru group such Benzoiru group, Benjiruokishika Ruponiru group, and a group derived from carbonic acid such as ethoxycarbonyl group Used.
  • a group suitable for etherification include a benzyl group, a tetrahydropyranyl group, and a t-butyl group.
  • the protecting group of the phenolic hydroxyl group of tyrosine for example, Bz l, C 1 2 - Bz l, 2- two Torobenjiru, B r- Z, such as t one-butyl is used.
  • protecting group for histidine imidazole for example, Tos, 4-methoxy-2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, Fmoc and the like are used.
  • Tos 4-methoxy-2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, Fmoc and the like are used.
  • Examples of the activated carbonyl group of the raw material include, for example, a corresponding acid anhydride, azide, active ester [alcohol (for example, pentachlorophenol, 2,4,5-trichloromouth phenol, 2,4 Dinitrophenol, cyanomethyl alcohol, paranitrophenol, H ⁇ NB, N-hydroxysuccinimide, N-hydroxyphthalimide, and ester with H'OB t).
  • active ester for example, pentachlorophenol, 2,4,5-trichloromouth phenol, 2,4 Dinitrophenol, cyanomethyl alcohol, paranitrophenol, H ⁇ NB, N-hydroxysuccinimide, N-hydroxyphthalimide, and ester with H'OB t.
  • Methods for removing (eliminating) the protecting group include, for example, catalytic reduction in a hydrogen stream in the presence of a catalyst such as Pd-black or Pd-carbon, or anhydrous hydrogen fluoride, Acid treatment with sulfonic acid, trifluoromethanesulfonic acid, trifluoroacetic acid or a mixture thereof, base treatment with diisopropylethylamine, triethylamine, piperidine, piperazine, etc. Reduction by a system is also used.
  • the elimination reaction by the above-mentioned acid treatment is generally carried out at a temperature of about 120 ° C. to 40 ° C.
  • anisol for example, anisol, phenol, thioanisole, methacresol, paracresol
  • a force thione scavenger such as dimethyl sulfide, 1,4-butanedithiol, 1,2-ethanedithiol and the like.
  • the 2,4-dinitrophenyl group used as an imidazole protecting group of histidine is removed by thiophenol treatment, and the formyl group used as an indole protecting group of tributofan is 1,2-ethanedithiol, 1,4- In addition to deprotection by acid treatment in the presence of butanedithiol, etc., it is also removed by alkali treatment with dilute sodium hydroxide solution, dilute ammonia and the like.
  • the protection of the functional group which should not be involved in the reaction of the raw materials, the protecting group, the elimination of the protective group, the activation of the functional group involved in the reaction, and the like can be appropriately selected from known groups or known means.
  • an amide of a protein or partial peptide for example, first, after amidating and protecting the ⁇ -hydroxyl group of the amino acid at the carboxy terminal, a peptide (protein) chain is added to the amino group side with a desired chain length.
  • the protein or partial peptide from which only the K-amino group protecting group at the ⁇ -terminal of the peptide chain has been removed, and the protein or partial peptide from which only the C-terminal carboxyl group-protecting group has been removed And condensing these proteins or peptides in a mixed solvent as described above. Details of the condensation reaction are the same as described above.
  • the crude protein or peptide can be purified using various known purification means, and the main fraction can be freeze-dried to obtain the desired amide of the protein or peptide.
  • an ester of a protein or peptide for example, After condensing the ⁇ -hydroxyl group of an amino acid with a desired alcohol to form an amino acid ester, a desired protein or peptide ester can be obtained in the same manner as in the case of a protein or peptide amide.
  • the partial peptide used in the present invention or a salt thereof can be produced according to a peptide synthesis method known per se, or by cleaving the protein used in the present invention with an appropriate peptidase.
  • a method for synthesizing a peptide for example, any of a solid phase synthesis method and a liquid phase synthesis method may be used. That is, a partial peptide or an amino acid capable of constituting the partial peptide used in the present invention is condensed with the remaining portion, and when the product has a protecting group, the protecting peptide is removed to produce the desired peptide. can do.
  • Known condensation methods and elimination of protecting groups include, for example, the methods described in the following 1 to 5.
  • the partial peptide used in the present invention can be purified and isolated by a combination of ordinary purification methods such as solvent extraction, distillation, column chromatography, 'liquid chromatography', and recrystallization.
  • the partial peptide obtained by the above method is a free form, it can be converted to an appropriate salt by a known method or a method analogous thereto, and conversely, when the partial peptide is obtained by a salt, the known method or Can be converted into a free form or another salt by a method according to the above.
  • the polynucleotide encoding the protein used in the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the protein used in the present invention.
  • it is DNA.
  • the DNA include genomic DNA, genomic DNA library, the above-described cell-tissue-derived cDNA, Any of the above-described cell / tissue-derived cDNA library and synthetic DNA may be used.
  • the vector used for the library may be any of pacteriophage, plasmid, cosmid, phagemid and the like.
  • reverse RNA was directly prepared using a preparation of total RNA or mRNA fraction from the above-mentioned cell 'tissue.
  • RT-PCR method transcriptase polymerase chain reaction
  • Examples of the DNA encoding the protein used in the present invention include a DNA containing the base sequence represented by SEQ ID NO: 2 or the base sequence represented by SEQ ID NO: 2 and high stringency. Any DNA that contains a nucleotide sequence that hybridizes under the following conditions and encodes a protein having substantially the same properties as the protein containing the amino acid sequence represented by SEQ ID NO: 1 described above. It may be something. Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 2 under conditions of eight istrines include, for example, about 50% or more, preferably about 6% with the nucleotide sequence represented by SEQ ID NO: 2. 0% or more, more preferably about 70% or more, more preferably about 80% or more, particularly preferably about 90% or more, most preferably about 95% or more. DNA or the like is used.
  • Hybridization can be performed according to a method known per se or a method analogous thereto, for example, the method described in Molecular Cloning 2nd (J. Saibrook et al., Cold Spring Harbor Lab. Press, 1989). When a commercially available library is used, it can be performed according to the method described in the attached instruction manual. More preferably, the reaction can be performed under high stringency conditions.
  • High stringent end conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 20 mM, and a temperature of about 50 to 70: preferably about 60 to 6
  • the conditions at 5 ° C are shown.
  • the case where the sodium concentration is about 19 mM and the temperature is about 65 ° C. is most preferable.
  • DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 1 a DNA containing the base sequence represented by SEQ ID NO: 2 or the like is used.
  • the polynucleotide (eg, DNA) encoding the partial peptide used in the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the partial peptide used in the present invention.
  • any of a genomic DNA, a genomic DNA library, the above-described cell / tissue-derived cDNA, the above-described cell / tissue-derived cDNA library, and synthetic DNA may be used.
  • Examples of the DNA encoding the partial peptide used in the present invention include, for example, a DNA containing a part of a DNA containing the base sequence represented by SEQ ID NO: 2, or a base represented by SEQ ID NO: 2.
  • a DNA containing a base sequence that hybridizes with the sequence under high stringent conditions, and a DNA containing a part of a DNA encoding a protein having substantially the same activity as the protein of the present invention is used.
  • the DNA that can be hybridized with the base sequence represented by SEQ ID NO: 2 has the same meaning as described above.
  • D which completely encodes the protein and partial peptide used in the present invention (hereinafter, these may be simply referred to as the protein of the present invention in the description of cloning and expression of the DNA encoding them).
  • DNA amplified by a PCR method using a synthetic DNA primer containing a part of the nucleotide sequence encoding the protein of the present invention, or a DNA incorporated into an appropriate vector is used as the DNA of the present invention.
  • the method of hybridization is, for example,
  • the DNA base sequence can be converted using the ODA-LA PCR method using PCR, a known kit, for example, Mutan TM -super Express Km (Takara Shuzo), Mutan TM- K (Takara Shuzo), or the like. , Gapped duplex method, Kunkel method and the like, or a method analogous thereto.
  • the DNA encoding the cloned protein can be used as it is depending on the purpose, or can be used after digesting with a restriction enzyme or adding a linker, if desired.
  • the DNA may have ATG as a translation initiation codon at the 5 'end and TAA, TGA or TAG as a translation termination codon at the 3' end. These translation initiation codon and translation termination codon can also be added using an appropriate synthetic DNA adapter.
  • the expression vector for the protein of the present invention includes, for example, (a) cutting out a DNA fragment of interest from DNA encoding the protein of the present invention, and (mouth) ligating the DNA fragment downstream of a promoter in an appropriate expression vector. It can be manufactured by
  • the vector examples include a plasmid derived from Escherichia coli (eg, pBR322, pBR325, pUC12, pUC13), a plasmid derived from Bacillus subtilis (eg, pUB110, pTP5, pC194), a plasmid derived from yeast (eg, p SH19, pSH15), pacteriophage such as phage ⁇ , animal viruses such as retrovirus, vaccinia virus, baculovirus, etc., pAl-11, pXTl, pRc / CMV, pRcZRSV, pcDNA I / N eo or the like is used.
  • Escherichia coli eg, pBR322, pBR325, pUC12, pUC13
  • Bacillus subtilis eg, pUB110, pTP5, pC194
  • yeast eg, p SH19, pSH
  • the promoter used in the present invention may be any promoter as long as it is appropriate for the host used for gene expression.
  • SRo when an animal cell is used as a host, SRo; Promoter, SV40 promoter, LTR promoter, CMV promoter, HSV-TK promoter and the like can be mentioned.
  • CMV cytomegalovirus
  • trp promoter one lac promoter, re cA promoter, AP L promoter - ter, 1
  • rho P promoter one such as T7 promoter
  • the host is a strain of the genus Bacillus, SP01 promoter
  • yeast such as overnight, SPO2 promoter, penP promoter, and the like
  • PHO5 promoter, PGK promoter, GAP promoter, ADH promoter, and the like are preferable.
  • the expression vector may contain, in addition to the above, an enhancer, a splicing signal, a poly-A addition signal, a selection promoter, and an SV40 replication origin (hereinafter sometimes abbreviated as SV40 ori), if desired. Can be used.
  • Selection methods include, for example, a dihydrofolate reductase (hereinafter sometimes abbreviated as dh fr) gene [methotrexate (MTX) resistance], an ampicillin resistance gene (hereinafter abbreviated as Amp 1 ) If there is), the neomycin resistance gene (hereinafter, Ne o sometimes abbreviated as r, G418 resistance), etc. in the like.
  • a signal sequence suitable for the host is added to the N-terminal side of the protein of the present invention.
  • the PhoA 'signal sequence and the OmpA * signal sequence are used.
  • the host is a Bacillus genus
  • the ⁇ -amylase signal sequence and subtilisin signal sequence are used.
  • Signal sequence, SUC2 signal sequence, etc. If the host is an animal cell, use insulin 'signal sequence, Hi-I interferon signal sequence, antibody molecule, signal sequence, etc. it can.
  • a transformant can be produced.
  • Escherichia bacteria for example, Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells, and the like are used.
  • Escherichia include, for example, Escherichia coli.
  • Bacillus bacteria examples include, for example, Bacillus' subtilis (Bacillus).
  • yeast examples include Saccharomyces cerevisiae AH22, AH22R—, NA87-11A, DKD—5D, 20B—12, Schizosaccharomyces pombe NC YC 1913, NCYC2036, and Pichia pastoris (Pichia pastor is) K M71 or the like is used.
  • insect cells for example, when the virus is Ac NPV, the cell line derived from the larvae of the night moth (Spodoptera frugiperda cell; S ; cell), the MG1 cell derived from the midgut of Trichoplusia ni, High Five TM cells, cells derived from Mamestra brassicae or cells derived from Estigmena acrea are used.
  • S sleep moth
  • MG1 cell derived from the midgut of Trichoplusia ni
  • High Five TM cells cells derived from Mamestra brassicae or cells derived from Estigmena acrea are used.
  • BmNPV a cell line derived from silkworm (Bombyx mori N cell; BmN cell) is used.
  • Sf9 cells for example, Sf9 cells (ATCC CRL1711), Sf21 cells (Vaughn, J.L., et al., In Vivo, 13, 213-217, (1977)) and the like are used.
  • insects for example, silkworm larvae are used [Maeda et al., Nature, Vol. 315, 592 (1985)].
  • animal cells for example, monkey cell COS-7, Vero, Chinese cell, Muster cell CHO (hereinafter abbreviated as CHO cell), dh fr gene-deficient chinini-zhamster cell CHO (hereinafter, CHO (dh fr)) Cells), mouse L cells, mouse AtT-20, mouse myeloma cells, rat GH3, human FL cells, and the like.
  • the yeast can be transformed according to the method described in, for example, Methods in Enzymology, Vol. 194, 182-187 (1991), Proc. Natl. Acad. Sci. USA, Vol. 75, 1929 (1978). Can be. Insect cells or insects can be transformed, for example, according to the method described in Bio / Technology, 6, 47-55 (1988).
  • a liquid medium is suitable as a medium used for the cultivation, and a carbon source necessary for the growth of the transformant is contained therein.
  • Nitrogen sources inorganic substances and others.
  • carbon sources include glucose, dextrin, soluble starch, and sucrose.
  • nitrogen sources include ammonium salts, nitrates, corn chip liquor, peptone, potato zein, meat extract, soybean meal, and potato extract.
  • the inorganic or organic substance such as a liquid and the inorganic substance include calcium chloride, sodium dihydrogen phosphate, and magnesium chloride.
  • yeast extract, vitamins, growth promoting factors and the like may be added.
  • the pH of the medium is preferably about 5-8.
  • an M9 medium containing glucose and casamino acids is preferable.
  • a drug such as 3 / 3-indolylacrylic acid can be added to make the promoter work efficiently if necessary.
  • the cultivation is usually performed at about 15 to 43 ° C for about 3 to 24 hours, and if necessary, aeration or stirring may be added.
  • the cultivation is usually performed at about 30 to 40 for about 6 to 24 hours, and if necessary, aeration and stirring may be added.
  • the culture medium When culturing an insect cell or a transformant in which the host is an insect, the culture medium is supplemented with Grace's Insect Medium (Grace, TCC, Nature, 195,788 (1962)). Those appropriately added are used.
  • the pH of the medium is preferably adjusted to about 6.2 to 6.4. Culture is usually performed at about 27 ° C for about 3 to 5 days, and aeration and agitation are added as necessary.
  • a MEM medium containing about 5 to 20% fetal bovine serum [Science, 122, 501 (1952)]
  • a DMEM medium [Virology , Vol. 8, 396 (1959)]
  • RPMI 1640 medium [The] journal of the American Medical Association 199, 519 (1967)]
  • 199 medium for example, a MEM medium containing about 5 to 20% fetal bovine serum [Science, 122, 501 (1952)], a DMEM medium [Virology , Vol. 8, 396 (1959)], RPMI 1640 medium [The] journal of the American Medical Association 199, 519 (1967)], 199 medium
  • the pH is about 6-8.
  • the cultivation is usually performed at about 30 to 40 ° C for about 15 to 60 hours, and aeration and stirring are added as necessary.
  • the protein of the present invention can be produced in the cells, in the cell membrane, or outside the cells of the transformant.
  • the protein of the present invention can be separated and purified from the culture by, for example, the following method.
  • the cells or cells When extracting the protein of the present invention from cultured cells or cells, after culturing, the cells or cells are collected by a known method, suspended in an appropriate buffer, and subjected to sonication, lysozyme and Z or freeze-thawing. After the cells or cells are destroyed by the method, a method of obtaining a crude extract of the protein by centrifugation or filtration is appropriately used.
  • the buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant such as Triton X-1100 TM.
  • Purification of the protein contained in the culture supernatant or extract obtained in this manner is a separation known per se. Purification methods can be combined appropriately. As these known separation and purification methods, solubility such as salting-out and solvent precipitation is used. PT / JP2003 / 007926
  • the protein thus obtained when the protein thus obtained is obtained as a free form, it can be converted to a salt by a method known per se or a method analogous thereto, and conversely, when the protein is obtained as a salt, a method known per se or It can be converted to a free form or other salts by a method analogous thereto.
  • the protein produced by the recombinant can be arbitrarily modified or the polypeptide can be partially removed by the action of an appropriate protein modifying enzyme before or after purification.
  • an appropriate protein modifying enzyme for example, trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, glycosidase and the like are used.
  • the presence of the protein of the present invention thus produced can be measured by, for example, enzymatic immunoassay western blotting using a specific antibody.
  • the antibody against the protein or partial peptide or a salt thereof used in the present invention is any of a polyclonal antibody and a monoclonal antibody as long as it can recognize the protein or partial peptide or a salt thereof used in the present invention. You may.
  • An antibody against the protein or partial peptide used in the present invention or a salt thereof (hereinafter sometimes simply referred to as the protein of the present invention in the description of the antibody) uses the protein of the present invention as an antigen, It can be produced according to a known antibody or antiserum production method.
  • the protein of the present invention is administered to a warm-blooded animal itself or together with a carrier or diluent at a site capable of producing an antibody upon administration.
  • Antibody production ability upon administration TJP2003 / 007926
  • Complete Freund's adjuvant or incomplete Freund's adjuvant may be given to increase the level.
  • Administration is usually performed once every 2 to 6 weeks, for a total of 2 to 10 times.
  • warm-blooded animal to be used include monkeys, egrets, dogs, guinea pigs, mice, rats, sheep, goats, and chickens, and mice and rats are preferably used.
  • a warm-blooded animal immunized with an antigen for example, an individual with an antibody titer is selected from a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization and contained in them.
  • the hybridoma producing the monoclonal antibody can be prepared by fusing the animal-producing cells obtained with myeloma cells of the same or different species.
  • the antibody titer in the antiserum can be measured, for example, by reacting the labeled protein described below with the antiserum, and then measuring the activity of the labeling agent bound to the antibody.
  • the fusion operation can be performed according to a known method, for example, the method of Kohler and Milstein [Nature, 256, 495 (1975)].
  • the fusion promoter include polyethylene glycol (PEG) and Sendai virus, but PEG is preferably used.
  • myeloma cells examples include myeloma cells of warm-blooded animals such as NS-1, P3U1, SP 2/0, and AP-1, but P3U1 is preferably used.
  • the preferred ratio between the number of antibody-producing cells (spleen cells) used and the number of myeloma cells used is about 1: 1 to 20: 1, and PEG (preferably PEG 1000 to PEG 6000) is used at a concentration of about 10 to 80%.
  • the cell fusion can be carried out efficiently by adding the mixture and incubating at 20 to 40 ° C., preferably 30 to 37, for 1 to 10 minutes.
  • a hybridoma culture supernatant is added to a solid phase (eg, a microplate) on which a protein antigen is adsorbed directly or together with a carrier.
  • a solid phase eg, a microplate
  • an anti-immunoglobulin antibody labeled with a radioactive substance or enzyme (anti-mouse immunoglobulin antibody is used if the cells used for cell fusion are mouse) or protein A
  • a monoclonal antibody bound to the solid phase A monoclonal antibody bound to a solid phase is prepared by adding a hybridoma supernatant to a solid phase to which an anti-immunoglobulin antibody or protein A has been adsorbed, adding a protein labeled with a radioactive substance, an enzyme, or the like. And a method for detecting antibody.
  • the selection of the monoclonal antibody can be performed according to a method known per se or a method analogous thereto. Usually, it can be performed in a medium for animal cells supplemented with HAT (hypoxanthine, aminopterin, thymidine).
  • HAT hyperxanthine, aminopterin, thymidine
  • any medium can be used as long as the hybridoma can grow.
  • RPMI 164 medium containing 1 to 20%, preferably 10 to 20% fetal bovine serum, GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd. ))
  • serum-free medium for hybridoma culture SFM-101, Nissui Pharmaceutical Co., Ltd.
  • the culture temperature is usually from 20 to 40 ° C, preferably about 37.
  • the culture time is generally 5 days to 3 weeks, preferably 1 week to 2 weeks. Cultivation can usually be performed under 5% CO2.
  • the antibody titer of the hybridoma culture supernatant can be measured in the same manner as the measurement of the antibody titer in the antiserum described above.
  • Monoclonal antibodies can be separated and purified by methods known per se, for example, immunoglobulin separation and purification methods (eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE) Absorption and desorption method, ultracentrifugation method, gel filtration method, antigen binding Solid phase or specific purification method of collecting antibody only with an active adsorbent such as protein A or protein G and dissociating the bond to obtain the antibody) Can do it.
  • immunoglobulin separation and purification methods eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE) Absorption and desorption method, ultracentrifugation method, gel filtration method, antigen binding Solid phase or specific purification method of collecting antibody only with an active adsorbent such as protein A or protein G and dissociating the bond to obtain the antibody) Can do it.
  • the polyclonal antibody of the present invention can be produced by a method known per se or a method analogous thereto. For example, an immunizing antigen (protein antigen) itself or a complex thereof with a carrier protein is formed, and immunization is performed on a warm-blooded animal in the same manner as in the above-described method for producing a monoclonal antibody.
  • the antibody can be produced by collecting the antibody-containing substance of the antibody and separating and purifying the antibody.
  • the type of carrier-protein and the mixing ratio of carrier and hapten depend on the efficiency of antibody against hapten immunized by cross-linking with carrier.
  • any substance may be cross-linked at any ratio.
  • serum serum albumin, psiloculopurine, hemocyanin, etc. may be used in a weight ratio of about 0.1 to 2 with respect to 1 hapten.
  • a method of coupling at a rate of 0, preferably about 1 to 5 is used.
  • various condensing agents can be used for force coupling between the hapten and the carrier.
  • an active ester reagent containing a daltaraldehyde, a carbodiimide, a maleimide active ester, a thiol group or a dithioviridyl group is used.
  • the condensation product is administered to a warm-blooded animal itself or together with a carrier or diluent at a site where antibody production is possible.
  • Complete Freund's adjuvant ⁇ Incomplete Freund's adjuvant may be administered to enhance body production during administration. The administration is usually made once every about 2 to 6 weeks, for a total of about 3 to 10 times.
  • the polyclonal antibody can be collected from the blood, ascites, etc., preferably from the blood of a warm-blooded animal immunized by the above method.
  • the measurement of the polyclonal antibody titer in the antiserum can be performed in the same manner as the measurement of the antibody titer in the antiserum described above. Separation and purification of the polyclonal antibody can be performed according to the same method for separation and purification of immunoglobulin as in the above-described separation and purification of the monoclonal antibody.
  • Polynucleotide encoding the protein or partial peptide used in the present invention eg, DNA (hereinafter, in the description of antisense polynucleotides, these DNAs may be abbreviated as the DNA of the present invention).
  • the nucleotide sequence of the polynucleotide of the present invention eg, DNA
  • Any antisense polynucleotide may be used as long as it contains a basic or substantially complementary base sequence or a part thereof and has an action capable of suppressing the expression of the DNA. Antisense DNA is preferred.
  • the nucleotide sequence substantially complementary to the DNA of the present invention is, for example, the entire nucleotide sequence or a partial nucleotide sequence of the nucleotide sequence complementary to the DNA of the present invention (that is, the complementary strand of the DNA of the present invention). And about 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more.
  • the antisense polynucleotide in the case of the antisense polynucleotide thus obtained, it is at least about 70%, preferably about 80%, complementary to the complementary sequence of the nucleotide sequence (for example, the nucleotide sequence near the start codon) of the N-terminal site of the protein of the present invention. % Or more, more preferably about 90% or more, and most preferably about 95% or more of the antisense polynucleotide which is directed to RNA degradation by RNase H.
  • an antisense polynucleotide containing a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of DNA containing the nucleotide sequence represented by SEQ ID NO: 2, or a part thereof Preferably, for example, an antisense polynucleotide containing a nucleotide sequence complementary to the nucleotide sequence of DNA containing the nucleotide sequence represented by SEQ ID NO: 2, or a part thereof (more preferably, a nucleotide sequence represented by SEQ ID NO: 2 A base sequence complementary to the base sequence of DNA containing the base sequence to be prepared, or an antisense polynucleotide containing a part thereof.
  • An antisense polynucleotide is usually composed of about 10 to 40 bases, preferably about 15 to 30 bases.
  • the phosphate residues (phosphates) of each nucleotide constituting the antisense DNA are, for example, chemically modified phosphorus such as phospho-thiolate, methylphosphonate, and phosphorodithionate. It may be substituted by an acid residue.
  • the sugar (deoxyliposome) of each nucleotide may be substituted with a chemically modified sugar structure such as 2,1-methylation, and the base portion (pyrimidine, purine) may also be chemically modified. Any one may be used as long as it hybridizes to DNA containing the nucleotide sequence represented by SEQ ID NO: 2.
  • These antisense polynucleotides can be produced using a known DNA synthesizer or the like.
  • an antisense polynucleotide capable of inhibiting the replication or expression of the protein gene of the present invention has been cloned or T JP2003 / 007926
  • nucleotide 29 It can be designed and synthesized based on the nucleotide sequence information of the DNA encoding the determined protein.
  • a polynucleotide nucleic acid
  • Polynucleotides complementary to the selected sequence of the protein-related RNA of the present invention, and polynucleotides capable of specifically hybridizing with the protein-related RNA of the present invention can be used in vivo and in vitro.
  • corresponding means having homology or being complementary to a nucleotide, base sequence or a specific sequence of a nucleic acid including a gene.
  • corresponding means having homology or being complementary to a nucleotide, base sequence or a specific sequence of a nucleic acid including a gene.
  • correspondence between a nucleotide, base sequence or nucleic acid and a peptide (protein) is a nucleotide
  • the palindrome region at the 'end and the hairpin loop at the 3' end can be selected as preferred regions of interest, but any region within the protein gene can be selected as the region of interest.
  • the relationship between the target nucleic acid and the polynucleotide complementary to at least a part of the target region is as follows: The relationship between the target nucleic acid and the polynucleotide capable of hybridizing with the target is
  • Antisense polynucleotides are polynucleotides containing 2-dexoxy D-ribose, polynucleotides containing D-reports, other types of polynucleotides that are N-glycosides of purine or pyrimidine bases, or non-polynucleotides.
  • Other polymers having a nucleotide backbone eg, commercially available protein nucleic acids and synthetic sequence-specific nucleic acid polymers
  • other polymers containing special bonds provided that such polymers are found in DNA and RNA
  • Base pairing including nucleotides having a configuration that allows base attachment
  • Bridges and may also be unmodified polynucleotides (or unmodified oligonucleotides), or even those with known modifications, such as those with a label known in the art, capped, Methylated, one or more natural nucleotides replaced by analogs, modified intramolecular nucleotides, e.g., uncharged bonds (e.g., methylphosphonates, phosphotriesters, phosphoramidates, Those with a charged bond or a sulfur-containing bond (eg, phosphorothioate, phosphorodithioate, etc.), eg, proteins (nucleases, nuclease inhibitors, toxins, antibodies, signals) Peptides, poly-L-lysine, etc.) and sugars (eg, Compounds with side groups such as monosaccharide, etc.), capped, Methylated, one or more natural nucleotides replaced by analogs, modified intramolecular nucleotides,
  • nucleoside may include not only those containing purine and pyrimidine bases but also those having other modified heterocyclic bases. Such modifications may include methylated purines and pyrimidines, acylated purines and pyrimidines, or other heterocycles. Modified nucleotides and modified nucleotides may also be modified at the sugar moiety, e.g., where one or more hydroxyl groups have been replaced with halogens, aliphatic groups, etc., or ethers, It may be converted to a functional group such as amine.
  • the antisense polynucleotide (nucleic acid) of the present invention is an RNA, a DNA, or a modified nucleic acid (RNA, DNA).
  • modified nucleic acid include, but are not limited to, sulfur derivatives of nucleic acids, thiophosphate derivatives, and polynucleoside amides, which are resistant to degradation of oligonucleoside amides.
  • the antisense nucleic acid of the present invention can be preferably designed according to the following policy. That is, to make the antisense nucleic acid more stable in the cell, to increase the cell permeability of the antisense nucleic acid, to have a greater affinity for the target sense strand, and to be more toxic if it is toxic. Sense nucleic acid toxicity Make it smaller.
  • the antisense nucleic acids of the present invention may contain altered or modified sugars, bases, or bonds, may be provided in special forms such as ribosomes or microspheres, may be applied by gene therapy, It could be given in additional form.
  • additional forms include polycations, such as polylysine, which act to neutralize the charge on the phosphate backbone, and lipids, which enhance interaction with cell membranes and increase nucleic acid uptake (eg, , Phospholipids, cholesterol, etc.).
  • Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chromate formate, cholic acid, etc.).
  • Such a substance can be attached to the 3 'end or 5' end of a nucleic acid, and can be attached via a base, a sugar, or an intramolecular nucleoside bond.
  • Other groups include cap groups specifically arranged at the 3 'end or 5' end of nucleic acids, which prevent degradation by nucleases such as exonucleases and RNAses.
  • capping groups include, but are not limited to, hydroxyl-protecting groups known in the art, including glycols such as polyethylene glycol and tetraethylene glycol.
  • the inhibitory activity of the antisense nucleic acid can be examined using the transformant of the present invention, the in vivo or in vitro gene expression system of the present invention, or the in vivo or in vitro translation system of the protein of the present invention.
  • the nucleic acid can be applied to cells by various methods known per se.
  • the protein or partial peptide of the present invention or a salt thereof hereinafter sometimes abbreviated as the protein of the present invention
  • the DNA encoding the protein or partial peptide of the present invention hereinafter referred to as the D NA
  • an antibody against the protein or partial peptide of the present invention or a salt thereof hereinafter referred to as “the present invention”.
  • an antibody sometimes abbreviated as an antibody
  • the antisense polynucleotide of the DNA of the present invention hereinafter sometimes abbreviated as the antisense polynucleotide of the present invention
  • a medicament containing an antisense polynucleotide of the gene encoding the protein of the present invention, a compound that inhibits the activity of the protein of the present invention or a salt thereof, or an antibody against the protein of the present invention may be, for example, a large intestine.
  • Cancer breast cancer, lung cancer, prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid gland It can be used as a prophylactic / therapeutic agent for cancer such as cancer, brain tumor or blood tumor.
  • compounds or salts thereof that modulate (preferably inhibit) the activity of the protein of the present invention include, for example, colorectal cancer, breast cancer, lung cancer, prostate cancer, esophageal cancer, stomach cancer, liver cancer, and biliary tract.
  • a prophylactic and therapeutic agent for cancer such as cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testis cancer, thyroid cancer, kidney cancer, brain tumor, or blood tumor can do.
  • it is a prophylactic / therapeutic agent for cancer (eg, breast cancer, etc.) that is hormone-independent (eg, estrogen, etc.)-Independent, HER2-negative, etc.
  • the compound or a salt thereof that inhibits the activity of the protein of the present invention can also be used, for example, as an apoptosis action regulator (preferably an apoptosis action promoter).
  • the protein of the present invention is useful as a reagent for screening a compound or its salt that regulates the activity of the protein of the present invention.
  • the present invention provides a method for screening a compound or a salt thereof that regulates the activity (eg, ligand binding activity, signal transduction activity, etc.) of the protein of the present invention, characterized by using the protein of the present invention. More specifically, for example, (i) the ligand binding activity or signal transduction activity of a cell capable of producing the protein of the present invention; A method for screening a compound or a salt thereof that regulates (promotes or inhibits) the activity of the protein of the present invention, which is characterized by comparing the ligand binding activity or the signal transmission activity of the mixture, is used.
  • the ligand binding activity or signal transduction activity can be determined by a method known per se, for example, J. Immunol. 166 (1), pp. 15-19, 2001 Measure and compare according to the method described in or above.
  • the protein expression vector of the present invention is prepared by adding an NF ⁇ B binding sequence to a promoter or enhancer. And (ii) a reporter gene containing the protein expression vector of the present invention in the promoter or enhancer of the NF ⁇ B binding sequence.
  • the amount of reporter expression is compared. The repo overnight expression level can be measured by using a reporter protein activity using luciferase activity, alkaline phosphatase activity or the like as an index.
  • the protein expression vector of the present invention may be used to bind the NF ⁇ B binding sequence to a promoter or enhancer. Transfection into cells with the reporter gene contained therein. If necessary, (a) a solution containing ligands such as microbial cell disruption solution, microbial culture supernatant, eukaryotic cell disruption solution, and eukaryotic cell culture supernatant (B) culturing with the addition of a substance having the same binding activity as the ligand itself or (c) a natural ligand; and (ii) using the protein expression vector of the present invention to promote the binding sequence of NFKB.
  • ligands such as microbial cell disruption solution, microbial culture supernatant, eukaryotic cell disruption solution, and eukaryotic cell culture supernatant
  • Transfected cells or microbial cell culture supernatant, eukaryotic cell lysate, eukaryotic cells Youe liquid containing the ligand, such as supernatants, by adding a substance having a (b) a ligand itself or (c) a natural ligand comparable binding activity, trial Measure and compare reporter expression levels when cultured in the presence of the test compound.
  • the expression level of the repo overnight can be measured by using the repoprotein activity using luciferase activity, alkaline phosphatase activity and the like as an index.
  • a host transformed with a vector containing a DNA encoding the protein of the present invention described above is used.
  • a host for example, animal cells such as COS 7 cells, CHO cells, and HEK293 cells are preferably used.
  • a transformant in which the protein of the present invention is expressed on a cell membrane by culturing by the method described above is preferably used.
  • the method for culturing cells capable of expressing the protein of the present invention is the same as the above-described method for culturing the transformant of the present invention.
  • Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and the like.
  • the ligand binding activity or signal transduction activity in the case of (ii) is increased by about 20% or more, preferably 30% or more, more preferably about 50% or more, as compared with the case of (i).
  • the test compound to be used is a compound that promotes the activity of the protein of the present invention, wherein the ligand binding activity or signal transduction activity in the case (ii) above is about 20% or more, preferably A test compound that reduces the activity of the protein of the present invention by 30% or more, more preferably about 50% or more, can be selected as the compound that inhibits the activity.
  • the compound having the activity of promoting the activity of the protein of the present invention is useful as a safe and low-toxic drug for enhancing the action of the protein of the present invention.
  • the compound having an activity of inhibiting the activity of the protein of the present invention is used as a safe and low-toxicity drug for suppressing the physiological activity of the protein of the present invention, for example, a prophylactic / therapeutic agent for cancer, an apoptosis promoting agent and the like. Useful.
  • Compounds or salts thereof obtained using the screening method or screening kit of the present invention include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts And compounds selected from plasma and the like.
  • the salt of the compound includes the peptide of the present invention described above. The same ones as used in the above are used.
  • compounds or salts thereof that regulate (preferably inhibit) the expression of the gene encoding the protein of the present invention include, for example, breast cancer and colon Cancer, breast, lung, prostate, esophagus, stomach, liver, biliary, spleen, kidney, bladder, uterus, ovary, testis, thyroid It can be used as a prophylactic and therapeutic agent for cancer such as cancer, kidney cancer, brain tumor and blood tumor.
  • it is a prophylactic / therapeutic agent for cancers (eg, breast cancer, etc.) that are hormone-independent (eg, estrogen, etc.)-Independent, HER2-negative, etc.
  • the compound that inhibits the expression of the gene encoding the protein of the present invention or a salt thereof can be used, for example, as an apoptosis-controlling agent (preferably, an apoptosis-promoting agent).
  • the DNA of the present invention is useful as a reagent for screening a compound or a salt thereof that regulates the expression of a gene encoding the protein of the present invention.
  • the screening method includes (iii) culturing cells capable of producing the protein of the present invention, and (iv) culturing cells capable of producing the protein used in the present invention in the presence of the test compound. There is a screening method which is characterized by comparing with the case.
  • the expression level of the gene (specifically, the amount of the protein of the present invention or the amount of mRNA encoding the protein) in the cases (ii) and (iv) is measured and compared.
  • test compound and cells having the ability to produce the protein of the present invention include the same cells as described above.
  • the amount of the protein is measured by a known method, for example, using an antibody that recognizes the protein of the present invention, and measuring the protein present in a cell extract or the like according to a method such as Western analysis, ELISA, or a method analogous thereto. can do.
  • the mRNA amount can be measured by a known method, for example, Northern hybridization using a nucleic acid containing SEQ ID NO: 2 or a part thereof as a probe, or
  • the gene expression in the case of the above (iv) is about 20% or more, preferably 30% or more, as compared with the case of the above (iii).
  • Test conjugates that inhibit at least about 50%, more preferably at least about 50%, can be selected as compounds that inhibit the expression of the gene encoding the protein of the present invention.
  • the screening kit of the present invention contains cells capable of producing the protein or partial peptide used in the present invention or a salt thereof, or the protein or partial peptide used in the present invention.
  • the compound or a salt thereof obtained by using the screening method or the screening kit of the present invention is a test compound as described above, for example, a peptide, a protein, a non-peptidic compound, a synthetic compound, a fermentation product, a cell extract, or a plant extract.
  • salt of the compound those similar to the aforementioned salts of the protein of the present invention are used.
  • a compound or a salt thereof that regulates (preferably inhibits) the activity of the protein of the present invention and a compound or a salt thereof that regulates (preferably inhibits) the expression of a gene encoding the protein of the present invention are, for example, colon cancer, Breast, lung, prostate, esophagus, stomach, liver, biliary, spleen, kidney, bladder, uterus, ovary, testis, thyroid, It is useful as a prophylactic / therapeutic agent for cancer, such as ligament cancer, brain tumor or hematological tumor, and as an apoptosis promoter.
  • they are agents for preventing or treating cancers (eg, breast cancer, etc.) that are hormone-independent (eg, estrogen, etc.)-Independent, HER2-negative, etc., and for promoting apoptosis.
  • cancers eg, breast cancer, etc.
  • hormone-independent eg, estrogen, etc.
  • HER2-negative e.g.
  • apoptosis e.g., HER2-negative, etc.
  • compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (including soft capsules). ), Syrups, emulsions, suspensions and the like.
  • Such a composition is produced by a method known per se and contains a carrier, diluent or excipient commonly used in the field of formulation. For example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and tablets for tablets.
  • compositions for parenteral administration for example, injections, suppositories, etc. are used. Injections are intravenous, subcutaneous, intradermal, intramuscular, intravenous, intraarticular. And dosage forms such as agents. Such injections are prepared according to a method known per se, for example, by dissolving, suspending or emulsifying the antibody or a salt thereof in a sterile aqueous or oily liquid commonly used for injections.
  • aqueous solution for injection for example, physiological saline, isotonic solution containing glucose and other adjuvants and the like are used, and suitable solubilizing agents, for example, alcohol (eg, ethanol), polyalcohol ( For example, propylene glycol, polyethylene glycol), nonionic surfactants [for example, polysorbate 80, HCO-50 (po lyoxye thy 1 ene (50mo 1) adduc t of
  • oily liquid for example, sesame oil, soybean oil and the like are used, and benzyl benzoate, benzyl alcohol and the like may be used in combination as a solubilizing agent.
  • the prepared injection solution is usually filled in a suitable ampoule.
  • a suppository for rectal administration is prepared by mixing the above antibody or a salt thereof with a conventional suppository base.
  • the above-mentioned oral or parenteral pharmaceutical composition is conveniently prepared in a unit dosage form adapted to the dose of the active ingredient.
  • dosage unit dosage forms include tablets, pills, capsules, injections (ampoules), and suppositories, and usually 5 to 50 mg / dosage per dosage unit dosage form.
  • the injection contains 5 to 100 mg of the above-mentioned antibody, and other dosage forms contain 10 to 25 mg of the above-mentioned antibody.
  • compositions may cause an undesired interaction due to the combination with the above antibody.
  • other active ingredients may be contained.
  • the preparations obtained in this way are safe and low toxic and can be used, for example, in humans or in warm-blooded animals (eg mice, rats, puppies, sheep, bush, pussi, puma, birds, cats, dogs, Monkeys, chimpanzees, etc.) orally or parenterally.
  • warm-blooded animals eg mice, rats, puppies, sheep, bush, pussi, puma, birds, cats, dogs, Monkeys, chimpanzees, etc.
  • the dose of the compound or a salt thereof varies depending on its action, target disease, subject to be administered, route of administration, and the like.
  • a compound or a salt thereof that regulates the activity of the protein of the present invention for the purpose of treating breast cancer When the compound is orally administered, generally in an adult (assuming a body weight of 60 kg), the compound or a salt thereof is used in an amount of about 0.1 to 10 Omg, preferably about 1.0 to 5 Omg, more preferably about 1 to 10 Omg per day.
  • the single dose of the compound or a salt thereof varies depending on the administration subject, target disease, and the like, but, for example, regulates the activity of the protein of the present invention for the purpose of treating breast cancer.
  • a compound or a salt thereof is administered to an adult (with a body weight of 6 O kg) usually in the form of an injection, the compound or a salt thereof is about 0.01 to 3 Omg, preferably about 0.1 to 2 Omg per day. More preferably, about 0.1 to 1 Omg is administered by injection to the cancerous lesion. In the case of other animals, the dose can be administered in terms of the weight per 6 O kg.
  • an antibody against the protein of the present invention (hereinafter sometimes abbreviated as the antibody of the present invention) can specifically recognize the protein of the present invention. In particular, it can be used for quantification by sandwich immunoassay.
  • the present invention provides a method for quantifying the protein of the present invention in a test wave, characterized by measuring In the quantification method (ii) above, it is desirable that one antibody is an antibody that recognizes the N-terminal of the protein of the present invention and the other antibody is an antibody that reacts with the C-terminal of the protein of the present invention. .
  • the protein of the present invention can be quantified using a monoclonal antibody against the protein of the present invention (hereinafter sometimes referred to as the monoclonal antibody of the present invention), and can also be detected by tissue staining or the like.
  • the antibody molecule itself may be used, or the F (ab ') 2 , Fab', or Fab fraction of the antibody molecule may be used.
  • the method for quantifying the protein of the present invention using the antibody of the present invention is not particularly limited, and may be an antibody, an antigen, or an antibody-antigen complex corresponding to the amount of antigen (eg, the amount of protein) in the test solution. Any measurement method may be used as long as the amount of the body is detected by chemical or physical means, and this is calculated from a standard curve prepared using a standard solution containing a known amount of antigen. .
  • nephelometry, competition method, immunometric method and sandwich method are preferably used, but in terms of sensitivity and specificity, it is particularly preferable to use a sandwich method described later.
  • a labeling agent used in a measurement method using a labeling substance for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance and the like are used. Radioisotopes, if example embodiment, [125 iota], [131 1], [3 ⁇ 4], and [14 c] used.
  • the above enzyme a stable enzyme having a large specific activity is preferable.
  • 3-galactosidase,) 3-glucosidase, alkaline phosphatase, peroxidase, malate dehydrogenase and the like are used.
  • the fluorescent substance for example, fluorescamine, fluorescein isothiocyanate and the like are used.
  • the luminescent substance for example, luminol, luminol derivative, luciferin, lucigenin and the like are used.
  • a biotin-avidin system can be used for binding the antibody or antigen to the labeling agent.
  • Carriers such as agarose, dextran, and cellulose Examples include soluble polysaccharides, synthetic resins such as polystyrene, polyacrylamide, and silicon, and glass.
  • the test solution is reacted with the insolubilized monoclonal antibody of the present invention (primary reaction), and further reacted with another labeled monoclonal antibody of the present invention (secondary reaction).
  • primary reaction the insolubilized monoclonal antibody of the present invention
  • secondary reaction another labeled monoclonal antibody of the present invention
  • the primary reaction and the secondary reaction may be performed in the reverse order, may be performed simultaneously, or may be performed at staggered times.
  • the labeling agent and the method of insolubilization can be in accordance with those described above.
  • the antibody used for the solid phase antibody or the labeling antibody does not necessarily need to be one kind, and a mixture of two or more kinds of antibodies is used for the purpose of improving measurement sensitivity and the like. May be used.
  • the monoclonal antibody of the present invention used in the primary reaction and the secondary reaction is an antibody having different binding sites to the protein of the present invention. Is preferably used. That is, the antibody used in the primary reaction and the secondary reaction is, for example, when the antibody used in the secondary reaction recognizes the C-terminal of the protein of the present invention, the antibody used in the primary reaction is Preferably, an antibody that recognizes other than the C-terminal, for example, the N-terminal, is used.
  • the monoclonal antibody of the present invention can be used in a measurement system other than the San Deutsch method, for example, a competition method, an immunometric method, or a nephrometry.
  • a competition method the antigen in the test solution and the labeled antigen are allowed to react competitively with the antibody, and then the unreacted labeled antigen (F) and the recognized antigen (B) bound to the antibody are separated.
  • F labeled antigen
  • B recognized antigen
  • BZF separation Measure the amount of labeling of either B or F, and quantify the amount of Bobara in the test wave.
  • a soluble antibody was used as the antibody
  • B / F separation was performed using polyethylene glycol
  • a liquid phase method using a second antibody against the antibody was used as the first antibody.
  • a solid phase method using a soluble first antibody and a solid phase antibody as the second antibody is used.
  • the antigen in the test solution and the immobilized antigen are subjected to a competitive reaction with a certain amount of labeled antibody, and then the solid phase and the liquid phase are separated. Reaction with an excess amount of the labeled antibody, and then adding immobilized antigen to the unreacted labeled antibody. After binding the body to the solid phase, the solid and liquid phases are separated. Next, the amount of label in either phase is measured to determine the amount of antigen in the test solution.
  • the amount of insoluble sediment resulting from the antigen-antibody reaction in a gel or in a solution is measured. Even when the amount of antigen in the test solution is small and only a small amount of sediment is obtained, laser-nephrometry utilizing laser scattering is preferably used.
  • the protein measuring system of the present invention may be constructed by adding ordinary technical considerations of those skilled in the art to ordinary conditions and operation methods in each method. For details of these general technical means, reference can be made to reviews, documents, etc.
  • the protein of the present invention can be quantified with high sensitivity by using the antibody of the present invention.
  • an increase or decrease in the concentration of the protein of the present invention is detected by quantifying the concentration of the protein of the present invention using the antibody of the present invention, for example, colon cancer, breast cancer, lung cancer, Prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid cancer, It can be diagnosed as having cancer, such as kidney cancer, brain tumor or hematological tumor, or is more likely to be affected in the future.
  • the antibody of the present invention can be used for detecting the protein of the present invention present in a subject such as a body fluid or a tissue.
  • a subject such as a body fluid or a tissue.
  • the detection of the protein of the present invention in each fraction during purification, and the analysis of the behavior of the protein of the present invention in test cells, etc. Can be used.
  • the DNA of the present invention can be used, for example, as a probe to produce human or warm-blooded animals (e.g., rats, mice, guinea pigs, egrets, birds, higgies, pigs, pigs, dogs, cats, dogs, Abnormalities (genetic abnormalities) in the DNA or mRNA encoding the protein of the present invention or its partial peptide in monkeys, chimpanzees, etc.).
  • human or warm-blooded animals e.g., rats, mice, guinea pigs, egrets, birds, higgies, pigs, pigs, dogs, cats, dogs, Abnormalities (genetic abnormalities) in the DNA or mRNA encoding the protein of the present invention or its partial peptide in monkeys, chimpanzees, etc.
  • it is useful as a diagnostic agent for genes such as decreased expression, increased DNA or mRNA or excessive expression.
  • the above-described genetic diagnosis using the DNA of the present invention includes, for example, the known Northern Hybridization ⁇ PCR-SSCP method (Genomics, Vol. 5, pp. 874-879, Q989), Proceedings of the National Academy of Sciences of the United States of America, Vol. 86, pp. 2766-2770 (1989)).
  • PCR-SSCP method for example, colon cancer, breast cancer, lung cancer, and prostate Cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testis cancer, thyroid cancer, visceral cancer, It can be diagnosed as having a high possibility of cancer such as a brain tumor or a blood tumor.
  • the antisense polynucleotide of the present invention which can complementarily bind to the DNA of the present invention and suppress the expression of the DNA, has low toxicity, and functions of the protein of the present invention or the DNA of the present invention in vivo.
  • ligand binding activity or signal transduction activity such as colon cancer, breast cancer, lung cancer, prostate cancer, esophagus cancer, stomach cancer, liver cancer, biliary tract cancer, and spleen
  • cancer such as cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid cancer, kidney cancer, brain tumor or blood tumor .
  • the antisense polynucleotide of the present invention can be used, for example, as an apoptotic action regulator (preferably an apoptotic action promoter).
  • the antisense polynucleotide When used as the above-mentioned prophylactic / therapeutic agent or modulator, it can be formulated and administered according to a method known per se.
  • the above-mentioned antisense polynucleotide is inserted alone or into a suitable vector such as a retrovirus vector, an adenovirus vector, an adenovirus association virus vector, etc. It can be administered orally or parenterally to mammals (eg, rats, egrets, sheep, sheep, bush, birds, cats, dogs, monkeys, etc.).
  • the antisense polynucleotide can be administered as it is or in the form of a formulation together with a physiologically acceptable carrier such as an adjuvant for promoting uptake, and administered by a gene gun or a catheter such as a hydrogel catheter. Alternatively, they can be made into an aerosol and administered topically into the trachea as an inhalant.
  • the above-mentioned antisense polynucleotide is formulated alone or together with a carrier such as liposome (injection), and is intravenously or subcutaneously. Etc. may be administered.
  • the dose of the antisense polynucleotide varies depending on the disease to be treated, the subject of administration, the route of administration, and the like.For example, when the antisense polynucleotide of the present invention is administered for the purpose of treating breast cancer, the dose is generally used for adults. (Weight 60 kg), the antisense polynucleotide is administered in an amount of about 0.1 to 10 O mg per day. 03 007926
  • the antisense polynucleotide can be used as a diagnostic oligonucleotide probe for examining the presence or expression of the DNA of the present invention in tissues or cells.
  • RNA containing a part of the RNA encoding the protein of the present invention a double-stranded RNA containing a part of the RNA encoding the protein of the present invention, a lipozyme containing a part of the RNA encoding the protein of the present invention, etc.
  • the expression of the gene of the present invention can be suppressed and the function of the protein used in the present invention or the DNA used in the present invention in vivo can be suppressed, for example, for example, colon cancer, breast cancer, lung cancer, Prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, renal cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid cancer, thyroid cancer Prevention and treatment of cancer such as cancer, brain tumor or blood tumor, preferably prevention of cancer (eg, breast cancer, etc.) independent of hormones (eg, estrogen), HER2 negative, etc.Furthermore, it can be used as an apoptosis action regulator or the like.
  • the double-stranded RNA can be produced by designing based on the sequence of the polynucleotide of the present invention according to a known method (eg, Nature, 411, 494, 2001).
  • the lipozyme can be designed and produced based on the sequence of the polynucleotide of the present invention according to a known method (eg, TRENDS in Molecular Medicine, Vol. 7, pp. 221, 2001). For example, it can be produced by linking a known lipozyme to a part of RNA encoding the protein of the present invention.
  • a part of the RNA encoding the protein of the present invention includes a portion (RNA fragment) close to the cleavage site on the RNA of the present invention, which can be cleaved by a known lipozyme.
  • RNA or lipozyme When used as a therapeutic agent, it can be formulated and administered in the same manner as an antisense polynucleotide. (5) a drug containing the antibody of the present invention
  • the antibody of the present invention which has the activity of neutralizing the activity of the protein of the present invention, includes, for example, colon cancer, breast cancer, lung cancer, prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer It can be used as a prophylactic or therapeutic agent for cancers such as renal, bladder, uterine, ovarian, testicular, thyroid, knee, brain or blood tumors it can. Preferably, it is a prophylactic / therapeutic agent for cancer (eg, breast cancer, etc.) that is hormone-independent (eg, estrogen, etc.)-Independent, HER2-expressing negative, etc. Further, the antibody of the present invention can also be used, for example, as an apoptosis action regulator (preferably, an apoptosis action promoter).
  • an apoptosis action regulator preferably, an apoptosis action promoter
  • the prophylactic or therapeutic agent for the above-mentioned diseases which comprises the antibody of the present invention, has low toxicity, and is used as it is as a liquid or as a pharmaceutical composition in an appropriate dosage form, in humans or mammals (eg, rat, rabbit, etc.). It can be given orally or parenterally (eg, intravenously, subcutaneously, etc.) to cattle, higgins, bushes, puppies, cats, dogs, monkeys, etc.).
  • the antibody of the present invention may be administered as it is, or may be administered as a suitable pharmaceutical composition.
  • the pharmaceutical composition used for administration may contain the antibody of the present invention or a salt thereof and a pharmacologically acceptable carrier, diluent or excipient.
  • Such a pharmaceutical composition is provided as a dosage form suitable for oral or parenteral administration.
  • compositions for parenteral administration include injections, suppositories, etc., and injections include intravenous, subcutaneous, intradermal, intramuscular, and intravenous injections. Shapes may be included.
  • Such an injection can be prepared according to a known method. Injections can be prepared, for example, by dissolving, suspending or emulsifying the antibody of the present invention or a salt thereof in a sterile aqueous liquid or oily liquid commonly used for injections.
  • aqueous solution for injection for example, physiological saline, isotonic solution containing glucose and other adjuvants, and the like, suitable solubilizing agents, for example, alcohol (e.g., ethanol), polyalcohol (e.g., , Propylene glycol, polyethylene glycol), nonionic surfactants [eg, Polysorbate 80, HC0-50
  • a suppository for rectal administration may be prepared by mixing the antibody or a salt thereof with a usual suppository base.
  • compositions for oral administration include solid or liquid dosage forms, specifically tablets (including dragees and film-coated tablets), pills, granules, powders, capsules (including soft capsules), syrups Agents, emulsions, suspensions and the like.
  • Such compositions are prepared by known methods and may contain carriers, diluents or excipients commonly used in the field of formulation.
  • carriers and excipients for tablets for example, lactose, starch, sucrose, and magnesium stearate are used.
  • the above-mentioned parenteral or oral pharmaceutical composition is conveniently prepared in a unit dosage form so as to be compatible with the dosage of the active ingredient.
  • Such dosage unit forms include, for example, tablets, pills, capsules, injections (ampoules), and suppositories.
  • the content of the antibody is preferably 5 to 500 mg per dosage unit dosage form, particularly 5 to 100 mg for injection, and 10 to 25 Omg for other dosage forms.
  • the dosage of the prophylactic / therapeutic or diagnostic agent (pharmaceutical) containing the antibody of the present invention varies depending on the administration target, target disease, symptoms, administration route, and the like.
  • the antibody of the present invention is used in a single dose, usually about 0.01 to 2 OmgZkg body weight, preferably about 0 :! to about 1 OmgZkg body weight, and more preferably about 0.1 to 5 mgZkg body weight.
  • Is administered by intravenous injection about 1 to 5 times a day, preferably about 1 to 3 times a day. In the case of other parenteral administration and oral administration, an equivalent dose can be administered. If the symptoms are particularly severe, the dose may be increased accordingly.
  • the antibodies of the present invention can be administered by themselves or as a suitable pharmaceutical composition.
  • the pharmaceutical composition used for the administration contains the antibody or a salt thereof and a pharmacologically acceptable carrier, diluent or excipient.
  • a composition is provided as a dosage form suitable for oral or parenteral administration (eg, intravascular injection, subcutaneous injection, etc.).
  • compositions may contain another active ingredient as long as the composition does not cause an undesirable interaction with the above-mentioned antibody.
  • the “compound having an activity of regulating (preferably inhibiting) the activity of RI PK2” may be any compound having an activity of regulating (preferably inhibiting) the activity of RIPK2, such as colon cancer, breast cancer, lung cancer, Prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid cancer, hepatic cancer It is used as a prophylactic / therapeutic agent for cancer such as cancer, brain tumor, or blood tumor.
  • Any compound can be used as long as it has a K2 expression regulating (preferably inhibiting) action, such as colon cancer, breast cancer, lung cancer, prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, Used as a prophylactic and therapeutic agent for cancer such as spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid cancer, kidney cancer, brain tumor, or blood tumor Can be
  • the prophylactic / therapeutic agent is produced in the same manner as described above.
  • the present invention relates to a DNA encoding the exogenous protein of the present invention (hereinafter abbreviated as the exogenous DNA of the present invention) or a mutant DNA thereof (may be abbreviated as the exogenous mutant DNA of the present invention). And a non-human mammal having the formula:
  • Non-human mammals having the exogenous DNA of the present invention or the mutant DNA thereof include unfertilized eggs, fertilized eggs, spermatozoa, For germ cells and the like including progenitor cells, calcium phosphate is preferably used at the stage of embryonic development in non-human mammal development (more preferably, at the stage of single cells or fertilized egg cells and generally before the 8-cell stage). It can be produced by transferring the desired DNA by the method, electric pulse method, lipofection method, coagulation method, microinjection method, particle gun method, DEAE-dextran method, etc.
  • the exogenous DNA of the present invention intended for somatic cells, organs of living organisms, tissue cells, and the like can be transferred and used for cell culture, tissue culture, and the like.
  • non-human mammals for example, porcupines, pigs, higgins, goats, magpies, dogs, cats, guinea pigs, eight-muster, ma., Males, rats and the like are used.
  • mice for example, C57B LZ6 strains, DBA2 strains as pure strains, etc.
  • a cross line a B6C3F line, a BDFi line, a B6D2F line, a BALBZc line, an ICR line, etc., or a rat (for example, Wistar, SD, etc.) are preferable.
  • mammal in the recombinant vector that can be expressed in mammals, human and the like can be mentioned in addition to the above-mentioned non-human mammals.
  • the exogenous DNA of the present invention refers not to the DNA of the present invention originally possessed by a non-human mammal, but to the DNA of the present invention once isolated and extracted from a mammal.
  • mutant DNA of the present invention those having a mutation (for example, mutation) in the base sequence of the original DNA of the present invention, specifically, addition or deletion of a base, substitution with another base, etc. DNA that has been used is used, and also includes abnormal DNA.
  • a mutation for example, mutation
  • the abnormal DNA means a DNA that expresses an abnormal protein of the present invention, and for example, DNA that expresses a protein that suppresses the function of the normal protein of the present invention is used.
  • the exogenous DNA of the present invention may be derived from a mammal of the same species or a different species as the target animal.
  • a DNA linked to a downstream of a promoter capable of expressing the DNA in animal cells is used. It is generally advantageous to use it as an A construct.
  • various mammals having the DNA of the present invention having a high homology to the human DNA eg, egret, dog, cat, guinea pig, hamster, rat, mouse, etc.
  • a target mammal for example, a mouse fertilized egg
  • various promoters capable of expressing the derived DNA it is possible to create a DNA transgenic mammal that highly expresses the DNA of the present invention.
  • Examples of the expression vector of the protein of the present invention include a plasmid derived from Escherichia coli, a plasmid derived from Bacillus subtilis, a plasmid derived from yeast, a bacteriophage such as ⁇ phage, a retrovirus such as Moroni leukemia virus, a vaccinia virus or a baculovirus.
  • animal viruses such as E. coli are used.
  • a plasmid derived from Escherichia coli, a plasmid derived from Bacillus subtilis or a plasmid derived from yeast are preferably used.
  • promoters that regulate DNA expression include: (1) a promoter of DNA derived from a virus (eg, Simian virus, cytomegalovirus, Moroni leukemia virus, JC virus, breast cancer virus, poliovirus, etc.). Yuichi, 2 Promoters derived from various mammals (humans, egrets, dogs, cats, guinea pigs, hamsters, rats, mice, etc.), for example, albumin, insulin II, ⁇ roplakin II, Erasuyuze, erythropoietin, Endothelin, muscle creatine kinase, glial fibrillary acidic protein, daryuthion S-transferase, platelet-derived growth factor / 3, keratin Kl, 1 ⁇ 10 and 14, collagen I and II , Cyclic AMP-dependent protein kinase / 3 I-subunit, dystrophin, Tartrate-resistant alkaline phosphatase, atrial sodium diuretic factor
  • Myosin light chains 1 and 2 myelin basic protein, thyroglobulin, Thy-1, immunoglobulin, heavy chain variable region (VNP), serum amyloid P component, myoglobin, troponin C, smooth muscle ⁇ -actin, prebub enkephalin ⁇ , A promoter such as vasopressin is used.
  • cytomegalovirus promoter which can be highly expressed in the whole body, human peptide chain elongation factor 1 (EF-1) promoter, human and chicken j3 actin promoter, and the like are preferable.
  • the vector preferably has a sequence (generally called terminator) that terminates transcription of the target messenger RNA in the DNA-transferred mammal.
  • terminator a sequence that terminates transcription of the target messenger RNA in the DNA-transferred mammal.
  • it is derived from viruses and various mammals.
  • the sequence of each DNA can be used, and preferably, SV40 terminator of Simian virus or the like is used.
  • the splicing signal of each DNA, the enhancer region, a part of the intron of eukaryotic DNA, etc. are used to further express the target exogenous DNA at 5 'upstream of the promoter region, the promoter region and the translation region. It is also possible to ligate between them or at the 3 'downstream of the translation region.
  • the normal translation region of the protein of the present invention is derived from a liver, a kidney, a thyroid cell, a fibroblast derived from a human or various mammals (for example,- ⁇ egret, dog, cat, guinea pig, hamster, rat, mouse, etc.). All or part of genomic DNA from DNA and various commercially available genomic DNA libraries, or complementary DNA prepared by known methods from liver, kidney, thyroid cell, and fibroblast-derived RNA can be used as raw materials. I can do it.
  • a foreign abnormal DNA can produce a translation region obtained by mutating a normal protein translation region obtained from the above cells or tissues by a point mutagenesis method.
  • the translation region can be prepared as a DNA construct that can be expressed in a transgenic animal by a conventional DNA engineering technique in which the translation region is ligated downstream of the promoter and, if desired, upstream of the transcription termination site.
  • Transfer of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal.
  • Production after DNA transfer The presence of the exogenous DNA of the present invention in the germ cell of the product means that all the progeny of the produced animal retain the exogenous DNA of the present invention in all of the germinal and somatic cells .
  • the offspring of such animals that have inherited the exogenous DNA of the present invention have the exogenous DNA of the present invention in all of their germinal and somatic cells.
  • the non-human mammal to which the exogenous normal DNA of the present invention has been transferred is confirmed to stably maintain the exogenous DNA by mating, and is subcultured as an animal having the DNA in a normal breeding environment. I can do it.
  • Transfer of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in excess in all germ cells and somatic cells of the target mammal.
  • Excessive presence of the exogenous DNA of the present invention in the germinal cells of the animal after transfer of DNA indicates that all of the offspring of the animal produced have the extraneous DNA of the present invention in all of their germinal and somatic cells.
  • the progeny of this type of animal that has inherited the exogenous DNA of the present invention has an excess of the exogenous DNA of the present invention in all of its germ cells and somatic cells.
  • the non-human mammal having the normal DNA of the present invention expresses the normal DNA of the present invention at a high level, and finally promotes the function of endogenous normal DNA, thereby finally obtaining the protein of the present invention. May develop hyperfunction, and can be used as a model animal for the disease. For example, using the normal DNA-transferred animal of the present invention, it is possible to elucidate the pathological mechanism of the hyperactivity of the protein of the present invention and diseases associated with the protein of the present invention and to examine a method for treating these diseases. It is possible.
  • a prophylactic / therapeutic agent for a disease associated with the protein of the present invention Cancer, breast cancer, lung cancer, prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid gland It can also be used for screening tests for prophylactic and therapeutic agents for cancer such as cancer, kidney cancer, brain tumor, and blood tumor.
  • the non-human mammal having the foreign abnormal DNA of the present invention After confirming that sex DNA is stably maintained, the animal can be subcultured as a DNA-bearing animal in a normal breeding environment. Furthermore, the desired foreign DNA can be incorporated into the above-mentioned plasmid and used as a raw material.
  • the DNA construct with the promoter can be prepared by ordinary DNA engineering techniques. The transfer of the abnormal DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal.
  • the presence of the abnormal DNA of the present invention in the germinal cells of the produced animal after DNA transfer means that all the offspring of the produced animal have the abnormal DNA of the present invention in all of its germ cells and somatic cells.
  • the progeny of this type of animal that has inherited the exogenous DNA of the present invention has the abnormal DNA of the present invention in all of its germinal and somatic cells.
  • a homozygous animal having the introduced DNA on both homologous chromosomes is obtained, and by crossing these male and female animals, it is possible to breed so that all offspring have the DNA.
  • the abnormal DNA of the present invention is highly expressed, and the function of the protein of the present invention is ultimately achieved by inhibiting the function of endogenous normal DNA.
  • Inactive refractory disease may occur and can be used as a model animal for the disease.
  • using the abnormal DNA-transferred animal of the present invention it is possible to elucidate the pathological mechanism of the function-inactive refractory of the protein of the present invention and to examine a method for treating this disease.
  • the abnormal DNA highly expressing animal of the present invention can be used to inhibit the function of the normal protein by the abnormal protein of the present invention in the function-inactive refractory disease of the protein of the present invention (dominant negative activity). Action).
  • the agent for preventing or treating the protein of the present invention or a functionally inactive refractory agent for example, Colorectal, breast, lung, prostate, esophageal, stomach, liver, biliary, spleen, kidney, bladder, uterine, ovarian, testicular, It can also be used for screening tests for prophylactic and therapeutic agents for cancers such as thyroid cancer, knee cancer, brain tumor, and blood tumor.
  • ⁇ ⁇ Isolation and purification of the mutant protein of the present invention and production of its antibody can be considered. Furthermore, using the DNA-transferred animal of the present invention, it is possible to examine clinical symptoms of a disease related to the protein of the present invention, including a functionally inactive refractory disease of the protein of the present invention. More detailed pathological findings in each organ of the disease model related to the protein of the present invention can be obtained, which will contribute to the development of new treatment methods and the research and treatment of secondary diseases caused by the disease. it can.
  • each organ is removed from the DNA-transferred animal of the present invention, and after minced, it is possible to obtain free DNA-transferred cells, culture them, or systematize the cultured cells using a protease such as trypsin. It is possible.
  • the protein of the present invention can be identified, its relationship with apoptosis, differentiation or proliferation, or its signal transduction mechanism can be examined, and its abnormality can be examined. It is an effective research material for elucidation.
  • the DNA transgenic animal of the present invention in order to use the DNA transgenic animal of the present invention to develop a therapeutic agent for a disease associated with the protein of the present invention, including a refractory inactive type of the protein of the present invention, Using a quantitative method or the like, it is possible to provide an effective and rapid screening method for the therapeutic agent for the disease. Also, using the DNA transgenic animal of the present invention or the exogenous DNA expression vector of the present invention, it is possible to examine and develop a method for treating a DNA associated with the protein of the present invention.
  • the present invention provides a non-human mammalian embryonic stem cell in which the DNA of the present invention is inactivated and a non-human mammal deficient in expression of the DNA of the present invention.
  • the DNA is inactivated by introducing a reporter gene (eg, a / 3-galactosidase enzyme gene derived from Escherichia coli), and the repo allele gene is a promoter of the promoter of the present invention.
  • a reporter gene eg, a / 3-galactosidase enzyme gene derived from Escherichia coli
  • the repo allele gene is a promoter of the promoter of the present invention.
  • the non-human mammal according to (6) which can be expressed under control,
  • test compound is administered to the animal described in (7), and the expression of a reporter gene is detected.
  • a screening method is provided.
  • a non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated is an artificially mutated DNA of the present invention possessed by the non-human mammal, which suppresses the expression ability of the DNA, or By substantially losing the activity of the protein of the present invention encoded by the DNA, the DNA substantially does not have the ability to express the protein of the present invention (hereinafter referred to as the knockout DNA of the present invention).
  • Non-human mammalian embryonic stem cells hereinafter abbreviated as ES cells).
  • non-human mammal the same one as described above is used.
  • the method of artificially mutating the DNA of the present invention can be carried out, for example, by deleting a part or all of the DNA sequence or inserting or substituting another DNA by a genetic engineering technique. . These mutations can, for example, shift the reading frame of codons or disrupt the function of Promote or Exon. What is necessary is just to prepare the knockout DNA of the present invention.
  • Non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated include, for example, The DNA of the present invention possessed by a non-human mammal is isolated, and its exon portion is a drug resistance gene typified by a neomycin resistance gene, a hygromycin resistance gene, or 1acZ (j3-galactosidase gene), cat Inserting a reporter gene or the like represented by (chloramphenicylacetyltransferase gene) disrupts exon function, or terminates transcription of the gene in the intron portion between exons.
  • a drug resistance gene typified by a neomycin resistance gene, a hygromycin resistance gene, or 1acZ (j3-galactosidase gene), cat Inserting a reporter gene or the like represented by (chloramphenicylacetyltransferase gene) disrupts exon function, or terminates transcription of the gene in the intron portion between exons
  • the resulting A DNA chain having a DNA sequence constructed so as to disrupt the gene (hereinafter, abbreviated as “gathering vector 1”) is introduced into the chromosome of the animal by, for example, a homologous recombination method.
  • gathering vector 1 A DNA sequence on or near the DNA of the present invention used for Southern hybridization analysis or a targeting vector and a DNA sequence on a targeting vector other than the DNA of the present invention used for preparing a targeting vectorcan be obtained by analyzing by the PCR method using as primers and selecting the knockout ES cells of the present invention.
  • the original ES cells for inactivating the DNA of the present invention by the homologous recombination method or the like for example, those already established as described above may be used.
  • BDFi mice C57BL / 6 and DBAZ2 and C57BL / 6 and DBAZ2
  • BDFi mice have the advantage of high number of eggs collected and robust eggs, and have the background of 'C57BL / 6 mice, The ES cells obtained using this were used to generate C57 It can be used to advantage in that loss can replace its genetic background with C57BL / 6 mice.
  • blastocysts 3.5 days after fertilization are generally used.However, it is more efficient to collect 8-cell embryos and culture them up to blastocysts. Many early embryos can be obtained.
  • Either male or female ES cells may be used, but male ES cells are generally more convenient for producing a germline chimera. It is also desirable to discriminate between males and females as soon as possible in order to reduce the complexity of culturing.
  • a method for determining the sex of ES cells a method of amplifying and detecting a gene in the sex-determining region on the Y chromosome by PCR can be given as an example.
  • this method conventionally, for example G-banding method, it requires about 1 0 6 cells for karyotype analysis, since suffices ES cell number of about 1 colony (about 5 0)
  • the primary selection of ES cells at the initial stage of culture can be performed by gender discrimination, and the early selection of male cells can greatly reduce the labor required at the initial stage of culture.
  • the secondary selection can be performed, for example, by confirming the number of chromosomes by the G-banding method.
  • Embryonic stem cell lines obtained in this way usually have very good proliferative properties, but they must be carefully subcultured because they tend to lose their ability to generate individuals.
  • a suitable feeder cell such as STO fibroblasts
  • a carbon dioxide incubator preferably 5% carbon dioxide, 95% air or 5% oxygen, in the presence of LIF (1 to 10,000 U / ml)
  • Culture at about 37 ° C with 5% carbon dioxide gas and 90% air.
  • trypsin / EDTA solution usually 0.001 to 0.5% trypsin / 0.1 l
  • a 5 mM EDTA preferably, about 0.1% tris / L M EDTA
  • Such subculture is usually performed every 1 to 3 days. At this time, cells are observed, and morphologically abnormal cells are found. If so, the cultured cells should be discarded.
  • ES cells can be transformed into various types of cells, such as parietal, visceral, and cardiac muscle, by monolayer culture up to high density or suspension culture until cell clumps are formed under appropriate conditions. It is possible to differentiate [M.]. Evans and MH
  • the non-human mammal deficient in DNA expression of the present invention can be distinguished from a normal animal by measuring the mRNA level of the animal using a known method and indirectly comparing the expression level.
  • non-human mammal those similar to the aforementioned can be used.
  • the non-human mammal deficient in DNA expression of the present invention can be obtained, for example, by introducing the evening-getting vector prepared as described above into a mouse embryonic stem cell or a mouse egg cell and introducing the evening-getting vector into a non-human mammal.
  • the DNA of the present invention is inactivated by homologous recombination to replace the DNA of the present invention on the chromosome of mouse embryonic stem cells or mouse egg cells by gene homologous recombination, thereby obtaining the DNA of the present invention. Can be knocked out.
  • Cells in which the DNA of the present invention has been knocked out can be analyzed by Southern hybridization analysis or DNA analysis using a DNA sequence on or near the DNA of the present invention as a probe. It can be determined by analysis by PCR using, as a primer, the DNA sequence of a neighboring region other than the DNA of the present invention derived from the mouse used for the targeting vector.
  • the cell line in which the DNA of the present invention has been inactivated is cloned by homologous gene recombination, and the cell line is cloned at an appropriate time, for example, at the 8-cell stage.
  • the chimeric embryo is injected into a human mammalian embryo or blastocyst, and the resulting chimeric embryo is transplanted into the uterus of the pseudo-pregnant non-human mammal.
  • the resulting animal will be artificially altered from cells having a normal DNA locus of the present invention. It is a chimeric animal composed of both cells having different DNA loci of the present invention.
  • all tissues are artificially mutated from a population obtained by crossing such a chimeric individual with a normal individual.
  • the individuals obtained in this manner are usually individuals deficient in the hetero-expression of the protein of the present invention, and mated with individuals deficient in the hetero-expression of the protein of the present invention.
  • a homozygous deficient individual can be obtained.
  • a transgenic non-human mammal having a chromosome into which a gettering vector has been introduced can be obtained by injecting a DNA solution into a nucleus of an egg by a microinjection method. Compared to a transgenic non-human mammal, it can be obtained by selecting a gene having a mutation in the DNA locus of the present invention by homologous recombination.
  • the animal individuals obtained by mating should also be confirmed to be knocked out of the DNA, and reared in a normal breeding environment. Can be.
  • the germline can be obtained and maintained according to a standard method. That is, by mating male and female animals having the inactivated DNA, homozygous animals having the inactivated DNA on both homologous chromosomes can be obtained.
  • the obtained homozygous animal can be efficiently obtained by rearing the mother animal in such a manner that one normal individual and plural homozygotes are obtained.
  • homozygous and heterozygous animals having the inactivated DNA are bred and passaged.
  • the non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated are extremely useful for producing the non-human mammal deficient in expression of the DNA of the present invention.
  • the non-human mammal deficient in expression of the DNA of the present invention lacks various biological activities that can be induced by the protein of the present invention, a disease caused by inactivation of the biological activity of the protein of the present invention. Since it can be a model, it is useful for investigating the causes of these diseases and examining treatment methods.
  • the non-human mammal deficient in expression of the DNA of the present invention can be used for screening for a compound having a therapeutic / preventive effect against diseases caused by the deficiency or damage of the DNA of the present invention.
  • the present invention comprises administering a test compound to a non-human mammal deficient in expression of the DNA of the present invention, and observing and measuring changes in the animal.
  • a test compound to a non-human mammal deficient in expression of the DNA of the present invention, and observing and measuring changes in the animal.
  • the non-human mammal deficient in DNA expression of the present invention used in the screening method includes the same ones as described above.
  • Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and plasma, and these compounds are novel compounds. Or a known compound.
  • a non-human mammal deficient in expression of the DNA of the present invention is treated with a test compound, compared with a non-treated control animal, and changes in the organs, tissues, disease symptoms, etc. of the animal are used as indicators.
  • the test compound can be tested for its therapeutic and prophylactic effects.
  • test compound for example, oral administration, intravenous injection and the like are used, and it can be appropriately selected according to the symptoms of the test animal, the properties of the test compound, and the like.
  • the dose of the test compound can be appropriately selected according to the administration method, the properties of the test compound, and the like.
  • testis For example, colon, breast, lung, prostate, esophagus, stomach, liver, biliary tract, spleen, kidney, bladder, uterus, ovary, testis
  • a test compound is administered to a non-human mammal deficient in DNA expression of the present invention. Then, the difference in the degree of onset of cancer and the degree of cure of cancer in the non-administered test compound group are observed over time in the above tissues.
  • test compound when administered to a test animal, When the disease symptom of the test animal is improved by about 10% or more, preferably about 30% or more, more preferably about 50% or more, the test compound has a therapeutic / preventive effect on the above-mentioned diseases. It can be selected as a compound.
  • the compound obtained by using the screening method is a compound selected from the test compounds described above, and has a therapeutic / preventive effect on a disease caused by a deficiency or damage of the protein of the present invention. It can be used as a medicament such as a safe and low toxic prophylactic or therapeutic agent. Further, a compound derived from the compound obtained by the above screening can also be used.
  • the compound obtained by the screening method may form a salt.
  • the salt of the compound include physiologically acceptable acids (eg, inorganic acids, organic acids, etc.) and bases (eg, alkalis). Salts with metals and the like are used, and especially preferred are physiologically acceptable acid addition salts.
  • salts include salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.), and organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid) , Succinic acid, tartaric acid, citric acid, malic acid, succinic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc.).
  • inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.
  • organic acids eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid
  • Succinic acid tartaric acid
  • citric acid malic acid
  • succinic acid benzoic acid
  • methanesulfonic acid benzenesulfonic acid, etc.
  • a drug containing the compound or a salt thereof obtained by the screening method can be produced in the same manner as the drug containing the protein of the present invention described above.
  • the preparations obtained in this way are safe and low toxic and can be used, for example, in humans or mammals (eg, rats, mice, guinea pigs, egrets, sheep, sheep, bushus, dogs, dogs, cats, dogs). , Monkeys, etc.).
  • the dose of the compound or a salt thereof varies depending on the target disease, the administration subject, the administration route, and the like.
  • the compound when the compound is orally administered, generally, breast cancer of an adult (assuming a body weight of 60 kg) is used.
  • the single dose of the compound varies depending on the administration subject, the target disease and the like.
  • the compound is usually in the form of an injection and usually used for adult (with a body weight of 6 O kg) breast cancer.
  • the compound When administered to a patient, the compound is administered in a daily dose of about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 mg. It is convenient to administer about 1 to 10 mg by intravenous injection. In the case of other animals, the dose can be administered in terms of weight per 60 kg.
  • the present invention provides a test compound administered to a non-human mammal deficient in expression of a DNA of the present invention, and detects the expression of a reporter gene.
  • the non-human mammal deficient in expression of DNA of the present invention may be the non-human mammal deficient in expression of DNA of the present invention in which the DNA of the present invention is inactive by introducing a reporter gene.
  • a gene which can be expressed under the control of a motor for the DNA of the present invention is used.
  • test compound examples include the same compounds as described above.
  • the same gene as described above is used, and a / 3-galactosidase gene (1 acZ), a soluble alkaline phosphatase gene or a luciferase gene is preferable.
  • the repo overnight gene is under the control of the promoter for the DNA of the present invention.
  • the protein-deficient mouse of the present invention or a tissue section thereof is fixed with datalaldehyde or the like, and phosphate buffered physiological After washing with saline solution (PBS), react with a staining solution containing X-ga 1 at room temperature or around 37 ° C for about 30 minutes to 1 hour, and then tissue samples are washed with ImM ED TAZ PBS solution. By washing, the jS-galactosidase reaction is stopped, and coloration may be observed.
  • mRNA encoding 1 ac Z may be detected according to a conventional method.
  • the compound or a salt thereof obtained by using the above-mentioned screening method is a compound selected from the above-mentioned test compounds, and is a compound that promotes or inhibits the DNA promoter activity of the present invention.
  • the compound obtained by the screening method may form a salt
  • the salt of the compound may be a physiologically acceptable acid (eg, an inorganic acid) or a base (eg, an alkali metal). And the like, and particularly preferably a physiologically acceptable acid addition salt.
  • examples of such salts include salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.), and organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, Salts with succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc.
  • inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.
  • organic acids eg, acetic acid, formic acid, propionic acid, fumaric acid, male
  • the compound of the present invention which promotes or inhibits the activity of the promoter against DNA, or a salt thereof can regulate the expression of the protein of the present invention and regulate the function of the protein, such as colon cancer, breast cancer, lung cancer, Prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testis cancer, thyroid cancer, knee It is useful as a prophylactic and / or therapeutic agent for cancer such as cancer, brain tumor, or blood tumor.
  • a compound derived from the compound obtained by the above screening can also be used.
  • a drug containing the compound or a salt thereof obtained by the screening method can be produced in the same manner as the above-mentioned drug containing the protein of the present invention or a salt thereof.
  • the preparations obtained in this way are safe and low toxic and can be used, for example, in humans or mammals (eg, rats, mice, guinea pigs, egrets, sheep, sheep, bushus, dogs, dogs, cats, dogs). , Monkeys, etc.).
  • the dose of the compound or a salt thereof varies depending on the target disease, the subject of the administration, the administration route, and the like.For example, when the compound of the present invention that inhibits the promoter activity for DNA is orally administered, generally the adult For a breast cancer patient weighing 60 kg, the compound is administered at about 0.1-100 mg, preferably about 1.0-5 Omg, more preferably about 1.0-2 Omg per day.
  • the single dose of the compound varies depending on the administration subject, target disease, and the like.
  • a compound that inhibits the promoter activity of DNA of the present invention is usually administered in the form of an injection to an adult.
  • the compound When administered to a breast cancer patient weighing 6 O kg, the compound is administered in an amount of about 0.01 to 3 Omg per day, preferably about 0.:! To 20 mg, more preferably about 0.1 to 1 mg per day.
  • Omg is conveniently administered by intravenous injection. In the case of other animals, the dose can be administered in terms of the weight per 6 O kg.
  • the non-human mammal deficient in expression of the DNA of the present invention is extremely useful for screening a compound or a salt thereof that promotes or inhibits the activity of the promoter for the DNA of the present invention. It can greatly contribute to investigating or preventing the causes of various diseases caused by insufficient expression of DNA.
  • genes encoding various proteins are ligated downstream thereof and injected into egg cells of an animal to produce a so-called transgenic animal (gene). Creating an introduced animal) will allow the specific synthesis of the protein and the study of its effects in living organisms. Furthermore, by binding an appropriate repo overnight gene to a portion of the above promoter and establishing a cell line in which this is expressed, the ability of the protein of the present invention itself to be produced in the body can be specifically promoted or suppressed. It can be used as a search system for low molecular compounds that have an action.
  • bases, amino acids, and the like are indicated by abbreviations based on the abbreviations of the IUPAC- IUB Commission on Biochemical Nomenclature or commonly used abbreviations in the art, and examples thereof are described below.
  • optical isomer for an amino acid the L-form is indicated unless otherwise specified.
  • DNA Deoxylipo nucleic acid cDNA complementary deoxyliponucleic acid A adenine
  • Tr p Tribute fan
  • Trt Trityl
  • 1 shows the nucleotide sequence of DNA containing the full length gene encoding RIPK2.
  • Example 3 shows the nucleotide sequence of the oligonucleotide used in Example 2.
  • Example 3 shows the nucleotide sequence of a primer used in Example 2.
  • Example 3 shows the nucleotide sequence of a primer used in Example 2.
  • Example 1 shows the nucleotide sequence of a probe used in Example 2.
  • Example 2 shows the nucleotide sequence of a probe used in Example 2.
  • RNA total RNA extracted from two breast cancer tissues and 23 other normal tissues was used as a material. Gene expression analysis was performed using an oligonucleotide microarray (Human Genome U95A, U95B, U95C, U95D, U95E; Affymetrix).
  • Example 2 In order to analyze the effect of RIPK2 gene on the apoptosis observed in breast cancer tissues, RI PK2 antisense oligonucleotide transfection experiments were performed. First, after designing an antisense (SEQ ID NO: 4) for the base sequence represented by SEQ ID NO: 3, a phosphorothioated oligonucleotide was synthesized, purified by HPLC, and used for an introduction experiment (Amersham Pharmacia Biotech). As a control oligonucleotide, the reverse sequence (SEQ ID NO: 5) of the base sequence represented by SEQ ID NO: 4 was similarly phosphorothioated, purified by HPLC and used (Amersham Pharmacia Biotech).
  • the expression level of the RIM2 gene was determined using two types of primers (SEQ ID NO: 6 and SEQ ID NO: 7) and a probe (SEQ ID NO: 8), and the ABI7700 (Perkin-Elmer) manual was used. PCR was performed according to the procedure. The expression level of RIPK2 was subtracted from the data obtained using the non-reverse transcription control from the data obtained using the cDNA, and then corrected with the total RNA amount used for comparison.
  • the expression of the protein of the present invention is increased in cancer tissues, and when the activity of the protein of the present invention is inhibited, cancer cells undergo apoptosis. Therefore, the protein of the present invention and a polynucleotide encoding the protein are diagnostic markers for cancer.
  • Compounds or salts thereof that regulate (preferably inhibit) the activity of the protein, compounds or salts thereof that regulate (preferably inhibit) the expression of the protein gene include, for example, colorectal cancer, breast cancer, lung cancer, Prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid cancer, sarcoma It can be used safely as a prophylactic or therapeutic agent for cancer such as cancer, brain tumors or hematological tumors.
  • the prophylactic / therapeutic agent for cancer eg, breast cancer, etc.
  • hormone-independent eg, estrogen, etc.
  • HER2 expression negative e.g., HER2 expression negative
  • the compound that inhibits the activity of the protein or a salt thereof, or the compound that inhibits the expression of the protein gene or a salt thereof can be safely used, for example, as an apoptosis-regulating agent (preferably, an apoptosis-action promoting agent). You can do that too.
  • the antisense polynucleotide or antibody of the present invention can inhibit the expression of the protein used in the present invention.
  • Therapeutic agent preferably as a preventive agent for cancer (eg, breast cancer, etc.) that is hormone-independent (eg, estrogen) -independent, HER2-negative, etc., or an apoptotic agent (preferably It can be used safely as an apoptosis action promoter).

Abstract

A compound or its salt controlling (inhibiting) the activity of a protein having an amino acid sequence which is the same or substantially the same as the amino acid sequence represented by SEQ ID NO:1, a compound controlling (inhibiting) the expression of the gene of the above protein, an antisense nucleotide having a base sequence or a part thereof which is complementary or substantially complementary to the base sequence of a DNA encoding the above protein or its peptide fragment, an antibody against the above protein or its peptide fragment, etc. are usable as preventives/remedies for cancer and apoptosis promoters.

Description

がんの予防 ·治療剤 技術分野  Cancer prevention and treatment technology
本発明は、 がんの予防 ·治療剤および診断薬などに関する。 背景技術 明  The present invention relates to an agent for preventing and treating cancer, a diagnostic agent, and the like. Background art
がんの化学療法においては、 新しい抗細がん剤の開発により延命効果が向上し、 治癒に向かうケースも増えてきている。しかしながら、 現在使用されている抗が ん剤は D NAに傷害を与えたり、 細胞分裂を阻害する細胞毒性の強いものが殆ど であり、 このため正常細胞に対しても少なからず傷害を与え、 特に細胞分裂の盛 んな骨髄などにしばしば強い副作用が現れる。  In cancer chemotherapy, the development of new anticancer drugs has improved life-prolonging effects and is increasing the number of cases toward cure. However, most of the currently used anticancer drugs are highly cytotoxic, damaging DNA and inhibiting cell division. Strong side effects often appear in bone marrow, where cell division is active.
遺伝子発現を網羅的に解析するために、 cDNAまたはオリゴヌクレオチドを固定 化したマイクロアレイ法が開発され、 疾患特異的な遺伝子発現の変化を見出す技 術が普及し、 その有用性が確認されている。 例えば、 Af fymet r ix社の GeneChipシ ステムは、 がんなどの疾患の診断や創薬標的遺伝子の発見に多用されつつある。 アンチセンスオリゴヌクレオチドは、 細胞内に導入されると、 相補的な配列を 有する RNAとハイブリダィズし、 RnaseHによる RMの分解を誘導してタンパク質翻 訳を阻害する、 またはハイブリダィズによる直接的タンパク合成阻害ももたらす。 目的の遺伝子機能を特異的に抑えることが可能なことから、 遺伝子の機能解析手 段として頻用されていると共に、 いくつかのアンチセンスオリゴヌクレオチドは 臨床応用開発が進んでいる。  In order to comprehensively analyze gene expression, a microarray method in which cDNA or oligonucleotides are immobilized has been developed. Techniques for finding disease-specific changes in gene expression have become widespread, and their usefulness has been confirmed. For example, Af fymetrix's GeneChip system is being widely used for diagnosing diseases such as cancer and finding drug discovery target genes. When introduced into cells, antisense oligonucleotides hybridize to RNAs with complementary sequences and induce RNase H degradation of RMs to inhibit protein translation, or to inhibit direct protein synthesis by hybridization. Bring. Because it is possible to specifically suppress the function of the target gene, it is frequently used as a means of analyzing the function of genes, and some antisense oligonucleotides are being developed for clinical application.
主に免疫系において、 FASを介した恒常性の維持が重要な役割を担っているこ とが知られている。 FASの Deathドメインに結合する因子として Receptor - It is known that maintaining FAS-mediated homeostasis plays an important role mainly in the immune system. Receptor-a factor that binds to the FAS Death domain
Interac t ing Ser ine/Threonine Kinase 1 (以下、 RIPK1) が見出されていたが、 最近、 この RIPK1に相同性を示す分子、 RIPK2がクロ一ニングされた (J. Bi o l . Chem. 273巻、 12296- 12300頁、 1998年) 。 その後、 RIPK2はいくつかの Caspaseや c IAPといったアポトーシス関連分子と結合すること、 また、 RIPK2を MCF7や 293T 細胞に強制発現させることによってアポトーシスを誘導することが報告されてい る (J. Biol . Chem. 273巻、 12296- 12300頁、 1998年および J. Bio l . Chem. 273 巻、 16968- 16975頁、 1998年) 。 また、 McCar thyらは 8種のがん細胞株での発現を 見て、 Raj i (B細胞由来) 以外の発現は低いと報告している (J. Bi ol . Chem. 273巻、 16968 - 16975頁、 1998年) 。 さらに最近、 RI 2ノックアウトマウスの研 究から、 RIPK2は To l l - l ike Receptor (TLR) - 2, 3, 4の下流で作用し、 自然免疫に関 与する分子であることが報告された (Nature 416巻、 190-194頁、 2002年; Nature 416巻、 194- 199頁、 2002年) 。 Interacting Serine / Threonine Kinase 1 (hereinafter referred to as RIPK1) was discovered, but a molecule showing homology to this RIPK1, RIPK2, has recently been cloned (J. Biol. Chem. 273). , 12296-12300, 1998). Subsequently, RIPK2 binds to apoptosis-related molecules such as Caspase and c IAP, and also binds RIPK2 to MCF7 and 293T. It has been reported that apoptosis is induced by forced expression in cells (J. Biol. Chem. 273, 12296-12300, 1998 and J. Biol. Chem. 273, 16968-16975, 1998). McCarthy et al., Looking at expression in eight cancer cell lines, reported that expression was low except for Raj i (derived from B cells) (J. Biol. Chem. 273, 16968- 16975, 1998). More recently, studies on RI2 knockout mice have reported that RIPK2 is a molecule that acts downstream of Toll-like Receptor (TLR) -2, 3, 4 and is involved in innate immunity ( Nature 416, 190-194, 2002; Nature 416, 194-199, 2002).
がん細胞に特異的に発現する分子を標的とし、 がん細胞の増殖阻害、 またはァ ポトーシス誘導を行える薬剤が切望されている。 また、 RIM2遺伝子の研究は、 免疫系に関するものがほとんどで、 がん組織での発現亢進に関する報告はなく、 RIPK2は MCF7や 293T細胞株を用いた実験から、 アポトーシスを誘導する因子と考 えられてきた。 発明の開示  There is a strong need for drugs that target molecules that are specifically expressed in cancer cells and that can inhibit the growth of cancer cells or induce apoptosis. In addition, most studies on the RIM2 gene relate to the immune system, and there are no reports of increased expression in cancer tissues.RIPK2 is considered to be a factor that induces apoptosis based on experiments using MCF7 and 293T cell lines. Have been. Disclosure of the invention
本発明者らは、 上記の課題を解決するために鋭意研究を重ねた結果、 がん組織、 特に悪性のがん組織に発現が顕著に増加する遺伝子として RIM2遺伝子を見出し た。 さらに、 アポトーシスを誘導する因子と考えられてきた RIPK2遺伝子の機能 を抑制することにより、 アポトーシスが誘導されることを見出し、 この知見に基 づいて、 さらに検討を重ねた結果、 本発明を完成するに至った。  The present inventors have conducted intensive studies to solve the above problems, and as a result, have found the RIM2 gene as a gene whose expression is significantly increased in cancer tissues, particularly malignant cancer tissues. Furthermore, they found that apoptosis was induced by suppressing the function of the RIPK2 gene, which has been considered to be a factor that induces apoptosis.Based on this finding, the inventors conducted further studies and completed the present invention. Reached.
すなわち、 本発明は、  That is, the present invention
( 1 ) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質もしくはその部分べプチドまたはその塩の活性 を阻害する化合物またはその塩を含有してなるがんの予防 ·治療剤、  (1) A protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a compound that inhibits the activity of a salt thereof, or a salt thereof Prevention and treatment of cancer
( 2 ) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩の遺伝 子の発現を阻害する化合物またはその塩を含有してなるがんの予防 ·治療剤、 ( 3 ) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質またはその部分ペプチドをコードするポリヌク レオチドの塩基配列に相補的もしくは実質的に相補的な塩基配列またはその一部 を含有するァンチセンスポリヌクレオチド、 (2) Contains a compound or a salt thereof that inhibits the expression of a protein or a partial peptide thereof or a salt thereof, which has the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1. (3) a polynucleic acid encoding a protein containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof An antisense polynucleotide containing a nucleotide sequence complementary or substantially complementary to the nucleotide sequence of leotide or a part thereof,
(4) 配列番号: 4で表される塩基配列を有する上記 (3) 記載のアンチセン スポリヌクレオチド、  (4) the antisense polynucleotide according to (3), which has the base sequence represented by SEQ ID NO: 4;
(5) 上記 (3) 記載のアンチセンスポリヌクレオチドを含有してなるがんの 予防 ·治療剤、  (5) a prophylactic or therapeutic agent for cancer comprising the antisense polynucleotide according to (3) above,
(6) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質またはその部分べプチドまたはその塩に対する 抗体を含有してなるがんの予防 ·治療剤、  (6) Prevention and treatment of cancer comprising an antibody against a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide or a salt thereof Agent,
(7) がんが、 大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣 がん、 甲状腺がん、 滕臓がん、 脳腫瘍または血液腫瘍である上記 (1) 、 (2) 、 (7) Cancer is colorectal, breast, lung, prostate, esophagus, stomach, liver, biliary, spleen, kidney, bladder, uterus, ovary (1), (2), cancer, testis, thyroid, thyroid, brain or hematological
(5) またば ( 6 ) 記載の予防 ·治療剤、 (5) A prophylactic or therapeutic agent according to (6),
(7 a) がんが、 ホルモン非依存性がんである上記 (1) 、 (2) 、 (5) ま たは (6) 記載の予防 ·治療剤、  (7a) The prophylactic or therapeutic agent according to (1), (2), (5) or (6) above, wherein the cancer is a hormone-independent cancer.
(7 b) がんが、 HER2発現陰性がんである上記 (1) 、 (2) 、 (5) または (7b) The cancer (1), (2), (5) or
(6) 記載の予防 ·治療剤、 (6) The preventive and therapeutic agents described in
(7 c) がんが、、 乳がんであるである上記 (1) 、 (2) 、 (5) または (7c) The cancer is breast cancer, (1), (2), (5) or
(6) 記載の予防 ·治療剤、 (6) The preventive and therapeutic agents described in
(8) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質またはその部分ペプチドまたはその塩に対する 抗体を含有してなるがんの診断薬、  (8) a diagnostic agent for cancer comprising an antibody against a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof;
(9) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質またはその部分ペプチドをコードするポリヌク レオチドを含有してなるがんの診断薬、  (9) a diagnostic agent for cancer comprising a polynucleotide encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof,
(10) がんが、 大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣 がん、 甲状腺がん、 塍臓がん、 脳腫瘍または血液腫瘍である上記 (8) または (10) Cancer is colorectal, breast, lung, prostate, esophagus, stomach, liver, biliary, spleen, kidney, bladder, uterus, ovary (8) or cancer, testicular cancer, thyroid cancer, kidney cancer, brain tumor or blood tumor
(9) 記載の診断薬、 (10 a) がんが、 ホルモン非依存性がんである上記 (8) または (9) 記載 の診断薬、 (9) The diagnostic agent described, (10a) The diagnostic agent according to (8) or (9), wherein the cancer is a hormone-independent cancer,
(10 b) がんが、 HER2発現陰性がんである上記 (8) または (9) 記載の診 断薬、  (10 b) The diagnostic drug according to the above (8) or (9), wherein the cancer is a HER2-expressing cancer.
(10 c) がんが、 乳がんである上記 (8) または (9) 記載の診断薬、 (10c) The diagnostic agent according to (8) or (9), wherein the cancer is breast cancer,
(11) Receptor-Interacting Serine/Threonine Kinase 2の活性阻害作用を 有する化合物またはその塩を含有してなるがんの予防 ·治療剤、 (11) A prophylactic / therapeutic agent for cancer comprising a compound having a Receptor-Interacting Serine / Threonine Kinase 2 activity inhibitory activity or a salt thereof,
(12) Receptor-Interacting Serine/Threonine Kinase 2の発現阻害作用を 有する化合物またはその塩を含有してなるがんの予防 ·治療剤、  (12) A prophylactic / therapeutic agent for cancer comprising a compound having an inhibitory effect on the expression of Receptor-Interacting Serine / Threonine Kinase 2 or a salt thereof,
(13) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩を用 いることを特徴とするがんの予防 ·治療剤のスクリーニング方法、  (13) a prophylactic or therapeutic agent for cancer characterized by using a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof; Screening method,
(13 a) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一 のアミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩を 用いることを特徴とする医薬用化合物のスクリーニング方法、  (13a) a method for screening a pharmaceutical compound, which comprises using a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1, a partial peptide thereof, or a salt thereof,
(13 b) 医薬用化合物が、 がんの予防 ·治療用の化合物、 がんの予防 ·治療 に用いられる化合物および Zまたはがんの予防 ·治療効果を有する化合物である 上記 (13 a) 記載のスクリ一二ング方法、  (13b) The pharmaceutical compound described in (13a) above, wherein the pharmaceutical compound is a compound for preventing or treating cancer, a compound used for preventing or treating cancer, or a compound having Z or Z preventive or therapeutic effect. The screening method of
(14) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の ァミノ酸配列を含有する夕ンパク質もしくはその部分べプチドまたはその塩を含 有することを特徴とするがんの予防 ·治療剤のスクリーニング用キット、  (14) Prevention of cancer characterized by containing a protein containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof · Kits for screening therapeutic agents,
(14 a) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一 のアミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩を 含有することを特徴とする医薬用化合物のスクリ一二ング用キット、  (14a) A screen of a pharmaceutical compound characterized by containing a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof. Kit for
(1 b) 医薬用化合物が、 がんの予防 ·治療用の化合物、 がんの予防 ·治療 に用いられる化合物および Zまたはがんの予防 ·治療効果を有する化合物である 上記 (14 a) 記載のスクリーニング用キット、  (1b) The pharmaceutical compound described in (14a) above, wherein the pharmaceutical compound is a compound for cancer prevention / treatment, a compound used for cancer prevention / treatment, or a compound having Z or cancer prevention / treatment effects. Screening kit,
(15) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質またはその部分ペプチドをコードするポリヌ PC漏 00雇 7926 (15) a polynucleotide encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof PC leakage 00 hire 7926
5 クレオチドを用いることを特徴とするがんの予防 ·治療剤のスクリ一二ング方法、 (15 a) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一 のァミノ酸配列を含有するタンパク質またはその部分べプチドをコードするポリ ヌクレオチドを用いることを特徴とする医薬用化合物のスクリ一二ング方法、 (15 b) 医薬用化合物が、 がんの予防 ·治療用の化合物、 がんの予防 ·治療 に用いられる化合物および Zまたはがんの予防 ·治療効果を有する化合物である 上記 (15 a) 記載のスクリーニング方法、  (5a) A method for screening a cancer prophylactic and / or therapeutic agent characterized by using a nucleotide, (15a) containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1. A method for screening a pharmaceutical compound, which comprises using a polynucleotide encoding a protein or a partial peptide thereof, (15b) a compound for preventing or treating cancer, The method according to (15a) above, which is a compound used for the prevention and treatment of Z and a compound having Z or cancer prevention and treatment effects.
(16) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の ァミノ酸配列を含有するタンパク質またはその部分ペプチドをコードするポリヌ クレオチドを含有することを特徴とするがんの予防 ·治療剤のスクリーニング用 キッ卜、  (16) Prevention of cancer characterized by containing a polynucleotide encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof, Kit for screening therapeutic agents,
(16 a) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一 のアミノ酸配列を含有するタンパク質もしくはその部分ペプチドをコードするポ リヌクレオチドを含有することを特徴とする医薬用化合物のスクリーニング用キ ッ卜、  (16a) a pharmaceutical compound characterized by containing a polynucleotide encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof; Screening kit,
(16 b) 医薬用化合物が、 がんの予防 ·治療用の化合物、 がんの予防 ·治療 に用いられる化合物および Zまたはがんの予防 ·治療効果を有する化合物である 上記 (16 a) 記載のスクリーニング用キット、  (16b) The pharmaceutical compound described in (16a) above, wherein the pharmaceutical compound is a compound for cancer prevention / treatment, a compound used for cancer prevention / treatment, or a compound having Z or cancer prevention / treatment effects. Screening kit,
(17) 上記 (13) もしくは (15) 記載のスクリーニング方法または上記 (14) もしくは (16) 記載のスクリーニング用キットを用いて得られうるが んの予防 ·治療剤、  (17) An agent for preventing or treating cancer, which can be obtained by using the screening method according to (13) or (15) or the screening kit according to (14) or (16).
(17 a) 上記 (13 a) もしくは (15 a) 記載のスクリーニング方法また は上記 (14 a) もしくは (16 a) 記載のスクリーニング用キットを用いて得 られうる医薬用化合物、  (17a) a pharmaceutical compound obtainable by using the screening method according to (13a) or (15a) or the screening kit according to (14a) or (16a);
(17 b) 医薬用化合物が、 がんの予防 ·治療用の化合物、 がんの予防 ·治療 に用いられる化合物および/またはがんの予防 ·治療効果を有する化合物である 上記 (17 a) 記載の化合物またはその塩、  (17b) The pharmaceutical compound described in (17a) above, wherein the pharmaceutical compound is a compound for preventing or treating cancer, a compound used for preventing or treating cancer, and / or a compound having an effect of preventing or treating cancer. Or a salt thereof,
(17 c) 上記 (17 b) 記載の化合物またはその塩を含有してなるがんの予 防 ·治療剤、 (18) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質もしくはその部分べプチドまたはその塩の活 性を阻害する化合物またはその塩を含有してなるアポトーシス作用促進剤、(17c) an agent for preventing or treating cancer comprising the compound or salt thereof according to (17b) above, (18) a protein or a partial peptide which has the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a compound which inhibits the activity of the salt or a salt thereof Apoptosis action promoter,
(19) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩の遺 伝子の発現を阻害する化合物またはその塩を含有してなるアポトーシス作用促進 剤、 (19) a compound or a salt thereof which inhibits the expression of a protein or a partial peptide thereof or a salt thereof which has the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1; Apoptosis promoting agent
(20) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質もしくはその部分べプチドまたはその塩を用 いることを特徴とするアポトーシス作用促進剤のスクリーニング方法、  (20) A method for screening an apoptosis-activating agent, which comprises using a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof. ,
(20 a) 医薬用化合物が、 アポトーシス作用促進用の化合物、 アポ卜一シス 作用促進に用いられる化合物および/またはアポトーシス作用促進効果を有する 化合物である上記 (15 a) 記載のスクリーニング方法、  (20a) The screening method according to (15a) above, wherein the pharmaceutical compound is a compound for promoting an apoptotic effect, a compound used for promoting an apoptotic effect, and / or a compound having an apoptotic effect promoting effect.
(20 b) 上記 (20 a) 記載の化合物またはその塩を含有してなるアポト一 シス作用促進剤、  (20b) an apoptosis-enhancing agent comprising the compound according to (20a) or a salt thereof,
(21) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の ァミノ酸配列を含有するタンパク質またはその部分べプチドをコードするポリヌ クレオチドを用いることを特徴とするアポトーシス作用促進剤のスクリ一ニング 方法、  (21) an agent for promoting apoptosis, characterized by using a protein containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or a polynucleotide encoding a partial peptide thereof; Screening method,
(22) 上記 (3) 記載のアンチセンスポリヌクレオチドを含有してなるアポ 卜一シス作用促進剤、  (22) an apoptosis-enhancing agent comprising the antisense polynucleotide according to (3),
(23) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の ァミノ酸配列を含有するタンパク質またはその部分べプチドまたはその塩に対す る抗体を含有してなるアポト一シス作用促進剤、  (23) Promoting apoptosis effect comprising an antibody against a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide or a salt thereof Agent,
(24) 哺乳動物に対して、 配列番号: 1で表されるアミノ酸配列と同一もし くは実質的に同一のアミノ酸配列を含有するタンパク質もしくはその部分べプチ ドまたはその塩の活性を阻害する化合物またはその塩、 または該タンパク質の遺 伝子の発現を阻害する化合物またはその塩の有効量を投与することを特徴とする がんの予防 ·治療方法、 • ( 2 5 ) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩の活 性を阻害する、 または該タンパク質の遺伝子の発現を阻害することを特徴とする がんの予防 ·治療方法、 (24) a compound that inhibits the activity of a protein, or a partial peptide or a salt thereof, having the same or substantially the same amino acid sequence as that represented by SEQ ID NO: 1 in a mammal; Or a salt thereof, or a compound that inhibits the expression of a gene of the protein or an effective amount of a salt thereof, comprising the steps of: • (25) inhibits the activity of a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof, or expresses a gene of the protein Cancer prevention and treatment methods characterized by inhibiting
( 2 6 ) 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一の アミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩の活 性を阻害する、 または該タンパク質の遺伝子の発現を阻害することを特徴とする アポトーシス作用の促進方法、  (26) Inhibits the activity of a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof, or inhibits the expression of a gene of the protein. A method of promoting apoptosis, characterized by inhibiting
( 2 7 ) がんの予防 ·治療剤を製造するための、 配列番号: 1で表されるアミ ノ酸配列と同一もしくは実質的に同一のァミノ酸配列を含有するタンパク質もし くはその部分べプチドまたはその塩の活性を阻害する化合物またはその塩、 また は該タンパク質の遺伝子の発現を阻害する化合物またはその塩の使用、  (27) A protein or a part thereof containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 for producing a prophylactic or therapeutic agent for cancer. Use of a compound or a salt thereof that inhibits the activity of a peptide or a salt thereof, or a compound or a salt thereof that inhibits expression of a gene of the protein,
( 2 8 ) アポトーシス作用促進剤を製造するための、 配列番号: 1で表される ァミノ酸配列と同一もしくは実質的に同一のァミノ酸配列を含有するタンパク質 もしくはその部分べプチドまたはその塩の活性を阻害する化合物またはその塩、 または該タンパク質の遺伝子の発現を阻害する化合物またはその塩の使用などを 提供する。 発明を実施するための最良の形態  (28) Activity of a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide or a salt thereof for producing an apoptosis promoting agent And a salt thereof, or a compound inhibiting the expression of the gene of the protein or a salt thereof. BEST MODE FOR CARRYING OUT THE INVENTION
本発明で用いられる配列番号: 1で表されるアミノ酸配列と同一もしくは実質 的に同一のアミノ酸配列を含有するタンパク質 (以下、 本発明のタンパク質また は本発明で用いられるタンパク質と称することもある) は、 ヒトゃ温血動物 (例 えば、 モルモット、 ラット、 マウス、 ニヮトリ、 ゥサギ、 ブ夕、 ヒッジ、 ゥシ、 サルなど) の細胞 (例えば、 肝細胞、 脾細胞、 神経細胞、 グリア細胞、 滕臓 i3細 胞、 骨髄細胞、 メサンギゥム細胞、 ランゲルハンス細胞、 表皮細胞、 上皮細胞、 杯細胞、 内皮細胞、 平滑筋細胞、 繊維芽細胞、 ·繊維細胞、 筋細胞、 脂肪細胞、 免 疫細胞 (例、 マクロファージ、 T細胞、 B細胞、 ナチュラルキラ一細胞、 肥満細 胞、 好中球、 好塩基球、 好酸球、 単球) 、 巨核球、 滑膜細胞、 軟骨細胞、 骨細胞、 , 骨芽細胞、 破骨細胞、 乳腺細胞、 肝細胞もしくは間質細胞、 またはこれら細胞の P2003/007926 A protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 used in the present invention (hereinafter sometimes referred to as the protein of the present invention or the protein used in the present invention) Are cells of human warm-blooded animals (eg, guinea pig, rat, mouse, chicken, egret, bush, hidge, horse, monkey, etc.) (eg, hepatocytes, spleen cells, nerve cells, glial cells, Glands i3 cells, bone marrow cells, mesangial cells, Langerhans cells, epidermal cells, epithelial cells, goblet cells, endothelial cells, smooth muscle cells, fibroblasts, fibrocytes, muscle cells, fat cells, immune cells (eg, Macrophages, T cells, B cells, natural killer cells, obesity cells, neutrophils, basophils, eosinophils, monocytes), megakaryocytes, synovial cells, cartilage Cells, osteocytes, osteoblasts, osteoclasts, breast cells, hepatocytes or stromal cells, or any of these cells P2003 / 007926
8 前駆細胞、 幹細胞もしくはがん細胞など) もしくはそれらの細胞が存在するあら ゆる組織、 例えば、 脳、 脳の各部位 (例、 嗅球、 扁桃核、 大脳基底球、 海馬、 視 床、 視床下部、 大脳皮質、 延髄、 小脳) 、 脊髄、 下垂体、 胃、 脬臓、 腎臓、 肝臓、 生殖腺、 甲状腺、 胆のう、 骨髄、 副腎、 皮膚、 筋肉、 肺、 消化管 (例、 大腸、 小 腸) 、 血管、 心臓、 胸腺、 脾臓、 顎下腺、 末梢血、 前立腺、 睾丸、 卵巣、 胎盤、 子宮、 骨、 関節、 骨格筋などに由来するタンパク質であってもよく、 合成タンパ ク質であってもよい。  8 progenitor cells, stem cells or cancer cells, etc.) or any tissue where these cells are present, such as the brain, various parts of the brain (eg, olfactory bulb, amygdala, basal sphere, hippocampus, thalamus, hypothalamus, Cerebral cortex, medulla, cerebellum), spinal cord, pituitary, stomach, kidney, kidney, liver, gonad, thyroid, gall bladder, bone marrow, adrenal gland, skin, muscle, lung, digestive tract (eg, large intestine, small intestine), blood vessels , Heart, thymus, spleen, submandibular gland, peripheral blood, prostate, testis, ovary, placenta, uterus, bone, joints, skeletal muscle, etc., or a synthetic protein .
配列番号: 1で表されるアミノ酸配列と実質的に同一のアミノ酸配列としては、 配列番号: 1で表されるアミノ酸配列と約 5 0 %以上、 好ましくは約 6 0 %以上、 さらに好ましくは約 7 0 %以上、 より好ましくは約 8 0 %以上、 特に好ましくは 約 9 0 %以上、 最も好ましくは約 9 5 %以上の相同性を有するアミノ酸配列など が挙げられる。  The amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 is about 50% or more, preferably about 60% or more, and more preferably about 50% or more of the amino acid sequence represented by SEQ ID NO: 1. Amino acid sequences having a homology of 70% or more, more preferably about 80% or more, particularly preferably about 90% or more, and most preferably about 95% or more.
アミノ酸配列の相同性は、 相同性計算アルゴリズム NCBI BLAST (Nat ional Center for Biotechnology Informat ion Bas ic Local Al ignment Search Tool) を用いて計算することができる。  Amino acid sequence homology can be calculated using a homology calculation algorithm NCBI BLAST (National Center for Biotechnology Information Basic Local Search Tool).
配列番号: 1で表されるアミノ酸配列と実質的に同一のアミノ酸配列を含有す るタンパク質としては、 例えば、 前記の配列番号: 1で表されるアミノ酸配列と 実質的に同一のアミノ酸配列を含有し、 配列番号: 1で表されるアミノ酸配列を 含有するタンパク質と実質的に同質の活性を有するタンパク質などが好ましい。 実質的に同質の活性としては、 例えば、 シグナル伝達活性 (例、 R I P K 2細 胞内シグナル伝達活性) 、 リン酸化活性などが挙げられる。 実質的に同質とは、 それらの性質が性質的に (例、 生理学的に、 または薬理学的に) 同質であること を示す。 したがって、 シグナル伝達活性、 リン酸化活性が同等 (例、 約 0 . 0 1 〜 1 0 0倍、 好ましくは約 0 . 1〜 1 0倍、 より好ましくは 0 . 5〜 2倍) であ ることが好ましいが、 これらの活性の程度、 タンパク質の分子量などの量的要素 は異なっていてもよい。  Examples of the protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 include, for example, a protein having an amino acid sequence substantially the same as the amino acid sequence represented by SEQ ID NO: 1 described above. However, a protein having substantially the same activity as the protein containing the amino acid sequence represented by SEQ ID NO: 1 is preferable. Examples of substantially equivalent activities include, for example, signal transduction activity (eg, RIPK2 intracellular signal transduction activity), phosphorylation activity, and the like. Substantially identical indicates that the properties are qualitatively (eg, physiologically or pharmacologically) identical. Therefore, the signal transduction activity and the phosphorylation activity are equivalent (eg, about 0.01 to 100 times, preferably about 0.1 to 10 times, more preferably 0.5 to 2 times). However, the quantitative factors such as the degree of these activities and the molecular weight of the protein may be different.
シグナル伝達活性、 リン酸化活性の測定は、 自体公知の方法、 例えば J. Bi ol . Chem. 273巻、 16968- 16975頁、 1998年に記載の方法またはそれに準じる方法に従 つて測定することができる。 R I P K 2はシグナル伝達において自己リン酸化す ると共に、 NF K Bシグナルを活性化する。 The signal transduction activity and phosphorylation activity can be measured by a method known per se, for example, the method described in J. Biol. Chem. 273, 16968-16975, 1998 or a method analogous thereto. . RIPK 2 is autophosphorylated in signaling Activates the NF KB signal.
例えば、 リン酸化活性は、 本発明のタンパク質 (例、 R I PK2) の発現べク ターを細胞に導入後、 適当な可溶化緩衝液で細胞を可溶化し、 免疫沈降法などに よって本発明のタンパク質 (例、 R I PK2) を得た後、 放射能ラベルされた A TPと適当な緩衝液中反応させ、 この反応液を SDSゲル電気泳動し、オートラ ジオグラフィーなどを用いることによって本発明のタンパク質 (例、 R I PK 2) の自己リン酸化の程度を測定できる。  For example, phosphorylation activity can be determined by introducing an expression vector for the protein of the present invention (eg, RI PK2) into cells, solubilizing the cells with an appropriate solubilization buffer, and immunoprecipitation. After obtaining a protein (eg, RI PK2), it is reacted with radiolabeled ATP in an appropriate buffer, and the reaction mixture is subjected to SDS gel electrophoresis, and the protein of the present invention is obtained by autoradiography or the like. (Eg, RI PK 2) can be measured for the degree of autophosphorylation.
シグナル伝達活性は、 本発明のタンパク質 (例、 R I PK2) の発現ベクター を、 NF κ B結合配列をプロモータ一またはェンハンサ一中に含有するレポ一夕 —遺伝子などとともに細胞に導入し、 必要に応じ、 微生物細胞破碎液、 微生物培 養上清、 真核細胞破碎液、 真核細胞培養上清などシグナル伝達活性または抑制化 物質などが含まれる液を添加して、 該レポーター発現量を自体公知の方法により 測定する。 レポ一夕一発現量は、 例えば、 ルシフェラ一ゼ活性、 アルカリフォス ファタ一ゼ活性などを用いたレポータータンパク活性を指標とすることにより測 定できる。  The signal transduction activity can be determined by introducing an expression vector for the protein of the present invention (eg, RI PK2) into a cell together with a repo overnight gene containing an NFκB binding sequence in a promoter or an enhancer. A liquid containing a signal transduction activity or a suppressive substance such as a microorganism cell disruption solution, a microorganism culture supernatant, a eukaryotic cell disruption solution, or a eukaryotic cell culture supernatant is added, and the reporter expression level is known per se. Measure according to the method. The repo overnight expression level can be measured using, for example, a reporter protein activity using luciferase activity, alkaline phosphatase activity, or the like as an index.
また、 本発明で用いられるタンパク質としては、 例えば、 ①配列番号: 1で表 されるアミノ酸配列中の 1または 2個以上 (例えば 1〜1 00個程度、 好ましく は 1〜30個程度、 好ましくは 1〜1 0個程度、 さらに好ましくは数 (1〜5) 個) のアミノ酸が欠失したアミノ酸配列、 ②配列番号: 1で表されるアミノ酸配 列に 1または 2個以上 (例えば 1〜1 00個程度、 好ましくは 1〜30個程度、 好ましくは 1〜1 0個程度、 さらに好ましくは数 (1〜5) 個) のアミノ酸が付 加したアミノ酸配列、 ③配列番号: 1で表されるアミノ酸配列に 1または 2個以 上 (例えば 1〜1 0 0個程度、 好ましくは 1〜30個程度、 好ましくは 1〜1 0 個程度、 さらに好ましくは数 (1〜5) 個) のアミノ酸が挿入されたアミノ酸配 列、 ④配列番号: 1で表されるアミノ酸配列中の 1または 2個以上 (例えば 1〜 1 0 0個程度、 好ましくは 1〜3 0個程度、 好ましくは 1〜1 0個程度、 さらに 好ましくは数 (1〜5) 個) のアミノ酸が他のアミノ酸で置換されたアミノ酸配 列、 または⑤それらを組み合わせたァミノ酸配列を含有するタンパク質などのい わゆるムテインも含まれる。 上記のようにアミノ酸配列が挿入、 欠失または置換されている場合、 その挿入、 欠失または置換の位置としては、 とくに限定されない。 Examples of the protein used in the present invention include: (1) one or two or more amino acids in the amino acid sequence represented by SEQ ID NO: 1 (for example, about 1 to 100, preferably about 1 to 30, An amino acid sequence in which about 1 to 10 amino acids have been deleted, more preferably a number (1 to 5) of amino acids, and 2 or more amino acids in the amino acid sequence represented by SEQ ID NO: 1 (for example, 1 to 1) An amino acid sequence to which about 00, preferably about 1 to 30, preferably about 1 to 10, and more preferably a number (1 to 5) of amino acids are added; ③ represented by SEQ ID NO: 1 The amino acid sequence contains one or more (for example, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, and more preferably about 1 to 5) amino acids. Inserted amino acid sequence, ④ 1 in the amino acid sequence represented by SEQ ID NO: 1 Or two or more (for example, about 1 to 100, preferably about 1 to 30, preferably about 1 to 10, and more preferably the number (1 to 5)) of other amino acids So-called muteins such as proteins containing an amino acid sequence substituted with or an amino acid sequence obtained by combining them are also included. When the amino acid sequence is inserted, deleted or substituted as described above, the position of the insertion, deletion or substitution is not particularly limited.
本明細書におけるタンパク質は、 ペプチド標記の慣例に従って左端が N末端 (ァミノ末端) 、 右端が C末端 (力ルポキシル末端) である。 配列番号: 1で表 されるアミノ酸配列を含有するタンパク質をはじめとする、 本発明で用いられる タンパク質は、 C末端が力ルポキシル基 (-C00H) 、 カルポキシレート(-C00— ) 、 アミド (- C0NH2) またはエステル (- C00R) の何れであってもよい。 In the present specification, the protein has a N-terminus at the left end (amino terminus) and a C-terminus at the right end (capillary end) according to the convention of peptide labeling. The protein used in the present invention, including the protein containing the amino acid sequence represented by SEQ ID NO: 1, has a C-terminal lipoxyl group (-C00H), carboxylate (-C00-), amide (- It may be either C0NH 2 ) or ester (-C00R).
ここでエステルにおける Rとしては、 例えば、 メチル、 ェチル、 n—プロピル、 イソプロピル、 n—ブチルなどの C ,_6アルキル基、 例えば、 シクロペンチル、 シ クロへキシルなどの C3_8シクロアルキル基、 例えば、 フエニル、 α—ナフチルな どの C 6_l27リール基、 例えば、 ベンジル、 フエネチルなどのフエ二ルー C卜 2ァ ルキル基もしくは α—ナフチルメチルなどの 一ナフチルー C ,_2アルキル基など の C 7_14ァラルキル基、 ピバ口ィルォキシメチル基などが用いられる。 Here, as R in the ester, e.g., methyl, Echiru, n- propyl, isopropyl, C, such as n- butyl, _ 6 alkyl groups, such as cyclopentyl, C 3 _ 8 cycloalkyl group such as cyclohexyl cyclo, for example, phenyl, alpha-naphthyl of which C 6 _ l2 7 aryl group, e.g., benzyl, one Nafuchiru C, such as phenylene Lou C Bok 2 § alkyl group or alpha-naphthylmethyl, such as phenethyl, such as _ 2 alkyl groups C 7 _ 14 Ararukiru group, such as pin bar opening Iruokishimechiru group is used.
本発明で用いられるタンパク質が C末端以外に力ルポキシル基 (または力ルポ キシレート) を有している場合、 カルボキシル基がアミド化またはエステル化さ れているものも本発明で用いられるタンパク質に含まれる。 この場合のエステル としては、 例えば上記した C末端のエステルなどが用いられる。  When the protein used in the present invention has a lipoxyl group (or lipoxylate) other than the C-terminus, a protein in which the carboxyl group is amidated or esterified is also included in the protein used in the present invention. . As the ester in this case, for example, the above-mentioned C-terminal ester and the like are used.
さらに、 本発明で用いられるタンパク質には、 N末端のアミノ酸残基 (例、 メ チォニン残基) のァミノ基が保護基 (例えば、 ホルミル基、 ァセチル基などの C μ6アルカノィルなどの _6ァシル基など) で保護されているもの、 生体内で切断 されて生成する N末端のダル夕ミン残基がピ口ダル夕ミン酸化したもの、 分子内 のアミノ酸の側鎖上の置換基 (例えば - 0H、 -SH、 アミノ基、 イミダゾール基、 ィ ンドール基、 グァニジノ基など) が適当な保護基 (例えば、 ホルミル基、 ァセチ ル基などの C ,_6アルカノィル基などの C卜6ァシル基など) で保護されているもの、 あるいは糖鎖が結合したいわゆる糖タンパク質などの複合タンパク質なども含ま れる。 Furthermore, the protein used in the present invention, the amino acid residue (e.g., main Chionin residues) of N-terminal Amino group protecting groups (e.g., formyl group, _ 6 Ashiru groups such as C Myu6 Arukanoiru such Asechiru group ), N-terminal dalluminin residue generated by cleavage in vivo, oxidized with lipamine, substituents on the side chains of amino acids in the molecule (eg-0H , -SH, amino group, imidazole group, I Ndoru group, in Guanijino group, etc.) a suitable protecting group (e.g., formyl group, C such Asechi group, such as C WINCH 6 Ashiru groups such as _ 6 Arukanoiru group) Protected proteins and complex proteins such as so-called glycoproteins to which sugar chains are bound are also included.
本発明で用いられるタンパク質の具体例としては、 例えば、 配列番号: 1で表 されるァミノ酸配列を含有する夕ンパク質などがあげられる。  Specific examples of the protein used in the present invention include, for example, protein containing an amino acid sequence represented by SEQ ID NO: 1.
本発明で用いられる夕ンパク質の部分べプチドとしては、 前記した本発明で用 いられるタンパク質の部分ペプチドであって、 好ましくは、 前記した本発明で用 いられるタンパク質と同様の性質を有するものであればいずれのものでもよい。 具体的には、 後述する本発明の抗体を調製する目的には、 配列番号: 1で表さ れるアミノ酸配列において第 2 7〜5 7 8番目のアミノ酸配列を有するペプチド などがあげられる。 例えば、 本発明で用いられるタンパク質の構成アミノ酸配列 のうち少なくとも 2 0個以上、 好ましくは 5 0個以上、 さらに好ましくは 7 0個 以上、 より好ましくは 1 0 0個以上、 最も好ましくは 2 0 0個以上のアミノ酸配 列を有するぺプチドなどが用いられる。 Examples of the partial peptide of the protein used in the present invention include: It is a partial peptide of the protein used, and preferably may be any peptide having the same properties as the protein used in the present invention described above. Specifically, for the purpose of preparing the antibody of the present invention described below, a peptide having the 27th to 578th amino acid sequence in the amino acid sequence represented by SEQ ID NO: 1 and the like can be mentioned. For example, at least 20 or more, preferably 50 or more, more preferably 70 or more, more preferably 100 or more, and most preferably 200 or more of the constituent amino acid sequences of the protein used in the present invention. Peptides having more than one amino acid sequence are used.
また、 本発明で用いられる部分ペプチドは、 そのアミノ酸配列中の 1または 2 個以上 (好ましくは、 1〜1 0個程度、 さらに好ましくは数 (1〜5 ) 個) のァ ミノ酸が欠失し、 または、 そのアミノ酸配列に 1または 2個以上 (好ましくは、 1〜2 0個程度、 より好ましくは 1〜1 0個程度、 さらに好ましくは数 (1〜 5 ) 個) のアミノ酸が付加し、 または、 そのアミノ酸配列に 1または 2個以上 (好ましくは、 1〜2 0個程度、 より好ましくは 1〜1 0個程度、 さらに好まし くは数 (1〜5 ) 個) のアミノ酸が挿入され、 または、 そのアミノ酸配列中の 1 または 2個以上 (好ましくは、 1〜1 0個程度、 より好ましくは数個、 さらに好 ましくは 1〜 5個程度) のアミノ酸が他のアミノ酸で置換されていてもよい。 また、 本発明で用いられる部分ペプチドは C末端が力ルポキシル基 (- C00H) 、 カルポキシレ一ト (-C00") 、 アミド (- C0NH2) またはエステル (-C00R) の何れ であってもよい。 In the partial peptide used in the present invention, one or more (preferably about 1 to 10, more preferably a number (1 to 5)) amino acids in the amino acid sequence are deleted. Or 1 or 2 or more (preferably, about 1 to 20, more preferably, about 1 to 10, and more preferably, about 1 to 5) amino acids are added to the amino acid sequence. Or 1 or 2 or more (preferably, about 1 to 20; more preferably, about 1 to 10; more preferably, 1 to 5) amino acids are inserted into the amino acid sequence. Or one or more (preferably about 1 to 10, more preferably several, and more preferably about 1 to 5) amino acids in the amino acid sequence are replaced with other amino acids. It may be. The partial peptide used in the present invention, the C-terminus force Rupokishiru group (- C00H), Karupokishire Ichito (-C00 "), amido (- C0NH 2) or may be any of the ester (-C00R).
さらに、 本発明で用いられる部分ペプチドには、 前記した本発明で用いられる タンパク質と同様に、 C末端以外にカルボキシル基 (またはカルポキシレート) を有しているもの、 N末端のアミノ酸残基 (例、 メチォニン残基) のァミノ基が 保護基で保護されているもの、 N端側が生体内で切断され生成したグルタミン残 基がピロダルタミン酸化したもの、 分子内のアミノ酸の側鎖上の置換基が適当な 保護基で保護されているもの、 あるいは糖鎖が結合したいわゆる糖ペプチドなど の複合ペプチドなども含まれる。  Further, similar to the protein used in the present invention, the partial peptide used in the present invention includes those having a carboxyl group (or carboxylate) in addition to the C-terminal, N-terminal amino acid residues ( (E.g., methionine residue) whose amino group is protected by a protecting group, glutamine residue generated by cleavage of N-terminal side in vivo and pyrodartamine oxidation, substitution on the side chain of amino acid in the molecule is Also included are those protected with an appropriate protecting group, and complex peptides such as so-called glycopeptides to which sugar chains are bonded.
本発明で用いられる部分べプチドは抗体作成のための抗原としても用いること ができる。 本発明で用いられるタンパク質または部分ペプチドの塩としては、 生理学的に 許容される酸 (例、 無機酸、 有機酸) や塩基 (例、 アルカリ金属塩) などとの塩 が用いられ、 とりわけ生理学的に許容される酸付加塩が好ましい。 この様な塩と しては、 例えば、 無機酸 (例えば、 塩酸、 リン酸、 臭化水素酸、 硫酸) との塩、 あるいは有機酸 (例えば、 酢酸、 ギ酸、 プロピオン酸、 フマル酸、 マレイン酸、 コハク酸、 酒石酸、 クェン酸、 リンゴ酸、 蓚酸、 安息香酸、 メタンスルホン酸、 ベンゼンスルホン酸) との塩などが用いられる。 The partial peptide used in the present invention can also be used as an antigen for producing an antibody. As the salt of the protein or partial peptide used in the present invention, salts with physiologically acceptable acids (eg, inorganic acids, organic acids) and bases (eg, alkali metal salts) are used. Are preferred. Such salts include, for example, salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid) or organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid) Succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid).
本発明で用いられるタンパク質もしくはその部分ペプチドまたはその塩は、 前 述したヒトゃ温血動物の細胞または組織から自体公知のタンパク質の精製方法に よって製造することもできるし、 タンパク質をコードする D NAを含有する形質 転換体を培養することによつても製造することができる。 また、 後述のペプチド 合成法に準じて製造することもできる。  The protein, its partial peptide, or a salt thereof used in the present invention can be produced from the above-mentioned human or warm-blooded animal cell or tissue by a known method for purifying a protein, or a DNA encoding the protein. Can also be produced by culturing a transformant containing It can also be produced according to the peptide synthesis method described below.
ヒトゃ哺乳動物の組織または細胞から製造する場合、 ヒトゃ哺乳動物の組織ま たは細胞をホモジナイズした後、 酸などで抽出を行ない、 該抽出液を逆相クロマ 卜グラフィー、 ィォン交換ク口マトグラフィーなどのクロマトグラフィーを組み 合わせることにより精製単離することができる。  When producing from human or mammalian tissues or cells, the human or mammalian tissues or cells are homogenized, then extracted with an acid or the like, and the extract is subjected to reverse phase chromatography, ion exchange chromatography. Purification and isolation can be performed by combining chromatography such as chromatography.
本発明で用いられるタンパク質もしくは部分ペプチドまたはその塩、 またはそ のアミド体の合成には、 通常市販のタンパク質合成用樹脂を用いることができる。 そのような樹脂としては、 例えば、 クロロメチル榭脂、 ヒドロキシメチル樹脂、 ベンズヒドリルアミン榭脂、 アミノメチル樹脂、 4一べンジルォキシベンジルァ ルコール樹脂、 4—メチルベンズヒドリルアミン榭脂、 P AM樹脂、 4—ヒドロ キシメチルメチルフエ二ルァセ卜アミドメチル樹脂、 ポリアクリルアミド榭脂、 4一 (2, , 4, ージメトキシフエ二ルーヒドロキシメチル) フエノキシ樹脂、 4 - ( 2 ' , 4, 一ジメトキシフエニル一 F m o cアミノエチル) フエノキシ樹 脂などを挙げることができる。 このような樹脂を用い、 α—ァミノ基と側鎖官能 基を適当に保護したアミノ酸を、 目的とするタンパク質の配列通りに、 自体公知 の各種縮合方法に従い、 樹脂上で縮合させる。 反応の最後に樹脂からタンパク質 または部分べプチドを切り出すと同時に各種保護基を除去し、 さらに高希釈溶液 中で分子内ジスルフィド結合形成反応を実施し、 目的のタンパク質もしくは部分 ペプチドまたはそれらのアミド体を取得する。 For the synthesis of the protein or partial peptide or a salt thereof, or an amide thereof used in the present invention, a commercially available resin for protein synthesis can be usually used. Examples of such a resin include chloromethyl resin, hydroxymethyl resin, benzhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, and PAM. Resin, 4-hydroxymethylmethylphenylacetamidomethyl resin, polyacrylamide resin, 4- (2,, 4, dimethoxyphenylhydroxymethyl) phenoxy resin, 4- (2 ′, 4,1-dimethoxyphenylmethyl resin F moc aminoethyl) phenoxy resin. Using such a resin, an amino acid having an α-amino group and a side chain functional group appropriately protected is condensed on the resin in accordance with the sequence of the target protein according to various known condensation methods. At the end of the reaction, a protein or partial peptide is cleaved from the resin, and at the same time, various protecting groups are removed.In addition, an intramolecular disulfide bond formation reaction is performed in a highly diluted solution to obtain the target protein or partial Obtain peptides or their amides.
上記した保護アミノ酸の縮合に関しては、 タンパク質合成に使用できる各種活 性化試薬を用いることができるが、 特に、 カルポジイミド類がよい。 カルポジィ ミド類としては、 D C C、 N, N'—ジイソプロピルカルポジイミド、 N—ェチ ル— N,一 ( 3—ジメチルァミノプロリル) カルポジイミドなどが用いられる。 これらによる活性化にはラセミ化抑 添加剤 (例えば、 H O B t , HO O B t ) とともに保護アミノ酸を直接樹脂に添加するかまたは、 対称酸無水物または H〇 B tエステルあるいは H〇O B tエステルとしてあらかじめ保護アミノ酸の活性 化を行なつた後に樹脂に添加することができる。  For the condensation of the protected amino acids described above, various activating reagents that can be used for protein synthesis can be used, and carbodiimides are particularly preferable. As the carpoimides, DCC, N, N'-diisopropylcarpoimide, N-ethyl-N, 1- (3-dimethylaminoprolyl) carpoimide, and the like are used. For these activations, the protected amino acid is added directly to the resin along with a racemization inhibitor (eg, HOB t, HO OB t), or as a symmetrical anhydride or H〇B t ester or H〇OB t ester After the protected amino acid is activated in advance, it can be added to the resin.
保護アミノ酸の活性化や樹脂との縮合に用いられる溶媒としては、 タンパク質 縮合反応に使用しうることが知られている溶媒から適宜選択されうる。 例えば、 The solvent used for activating the protected amino acid or condensing with the resin can be appropriately selected from solvents known to be usable for the protein condensation reaction. For example,
N, N—ジメチルホルムアミド, N, N—ジメチルァセトアミド, N—メチルビ 口リドンなどの酸アミド類、 塩化メチレン, クロ口ホルムなどのハロゲン化炭化 水素類、 トリフルォロエタノールなどのアルコール類、 ジメチルスルホキシドな どのスルホキシド類、 ピリジン, ジォキサン, テトラヒドロフランなどのエーテ ル類、 ァセトニトリル, プロピオ二トリルなどの二トリル類、 酢酸メチル, 酢酸 ェチルなどのエステル類あるいはこれらの適宜の混合物などが用いられる。 反応 温度はタンパク質結合形成反応に使用され得ることが知られている範囲から適宜 選択され、 通常約ー2 0 °C〜5 0 °Cの範囲から適宜選択される。 活性化されたァ ミノ酸誘導体は通常 1 . 5〜4倍過剰で用いられる。 ニンヒドリン反応を用いた テストの結果、 縮合が不十分な場合には保護基の脱離を行なうことなく縮合反応 を繰り返すことにより十分な縮合を行なうことができる。 反応を繰り返しても十 分な縮合が得られないときには、 無水酢酸またはァセチルイミダゾ一ルを用いて 未反応アミノ酸をァセチル化することによって、 後の反応に影響を与えないよう にすることができる。 Acid amides such as N, N-dimethylformamide, N, N-dimethylacetamide and N-methylvinylidone; halogenated hydrocarbons such as methylene chloride and chloroform; alcohols such as trifluoroethanol; Sulfoxides such as dimethylsulfoxide, ethers such as pyridine, dioxane and tetrahydrofuran, nitriles such as acetonitrile and propionitrile, esters such as methyl acetate and ethyl acetate, or an appropriate mixture thereof are used. The reaction temperature is appropriately selected from a range that is known to be used for a protein bond formation reaction, and is usually appropriately selected from a range of about −20 ° C. to 50 ° C. The activated amino acid derivative is usually used in a 1.5 to 4-fold excess. As a result of the test using the ninhydrin reaction, if the condensation is insufficient, sufficient condensation can be performed by repeating the condensation reaction without removing the protecting group. If sufficient condensation cannot be obtained even after repeating the reaction, unreacted amino acids can be acetylated using acetic anhydride or acetylimidazole so that subsequent reactions are not affected. .
原料のァミノ基の保護基としては、 例えば、 Z、 B o c、 t—ペンチルォキシ 力ルポニル、 イソポルニルォキシカルポニル、 4ーメトキシベンジルォキシカル ポニル、 C 1 一 Z、 B r— Z、 ァダマンチルォキシカルボニル、 トリフルォロア セチル、 フタロイル、 ホルミル、 2一二トロフエニルスルフエニル、 ジフエ二ル ホスフイノチオイル、 Fmo cなどが用いられる。 ' Examples of the protecting group for the amino group of the starting material include Z, Boc, t-pentyloxycarbonyl, isopolnyoxycarbonyl, 4-methoxybenzyloxycarbonyl, C11Z, Br-Z, a Damantyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 212-trophenylsulfenyl, diphenyl Phosphinothio oil, Fmoc, and the like are used. '
力ルポキシル基は、 例えば、 アルキルエステル化 (例えば、 メチル、 ェチル、 プロピル、 ブチル、 tーブチル、 シクロペンチル、 シクロへキシル、 シクロヘプ チル、 シクロォクチル、 2—ァダマンチルなどの直鎖状、 分枝状もしくは環状ァ ルキルエステル化) 、 ァラルキルエステル化 (例えば、 ベンジルエステル、 4一 ニトロべンジルエステル、 4ーメトキシベンジルエステル、 4—クロ口べンジル エステル、 ベンズヒドリルエステル化) 、 フエナシルエステル化、 ベンジルォキ シカルポニルヒドラジド化、 t一ブトキシカルポニルヒドラジド化、 トリチルヒ ドラジド化などによつて保護することができる。  The lipoxyl group can be, for example, alkyl esterified (eg, methyl, ethyl, propyl, butyl, t-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.). Alkyl esterification), aralkyl esterification (eg, benzyl ester, 4-nitrobenzyl ester, 4-methoxybenzyl ester, 4-chlorobenzyl ester, benzhydryl esterification), phenacyl esterification, benzyloxycarponyl It can be protected by hydrazide, t-butoxycarbonyl hydrazide, trityl hydrazide and the like.
セリンの水酸基は、 例えば、 エステル化またはエーテル化によって保護するこ とができる。 このエステル化に適する基としては、 例えば、 ァセチル基などの低 級 (C,.6) アルカノィル基、 ベンゾィル基などのァロイル基、 ベンジルォキシカ ルポニル基、 エトキシカルボニル基などの炭酸から誘導される基などが用いられ る。 また、 エーテル化に適する基としては、 例えば、 ベンジル基、 テトラヒドロ ピラニル基、 t一ブチル基などである。 The hydroxyl group of serine can be protected, for example, by esterification or etherification. Examples of groups appropriately used for the esterification, for example, low-grade (C ,. 6) Arukanoiru groups such Asechiru group, Aroiru group such Benzoiru group, Benjiruokishika Ruponiru group, and a group derived from carbonic acid such as ethoxycarbonyl group Used. Examples of a group suitable for etherification include a benzyl group, a tetrahydropyranyl group, and a t-butyl group.
チロシンのフエノール性水酸基の保護基としては、 例えば、 Bz l、 C 12- Bz l、 2—二トロベンジル、 B r— Z、 t一ブチルなどが用いられる。 The protecting group of the phenolic hydroxyl group of tyrosine, for example, Bz l, C 1 2 - Bz l, 2- two Torobenjiru, B r- Z, such as t one-butyl is used.
ヒスチジンのイミダゾ一ルの保護基としては、 例えば、 To s、 4—メトキシ -2, 3, 6—トリメチルベンゼンスルホニル、 DNP、 ベンジルォキシメチル、 Bum, Bo c、 Tr t、 Fmo cなどが用いられる。  As the protecting group for histidine imidazole, for example, Tos, 4-methoxy-2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, Fmoc and the like are used. Can be
原料の力ルポキシル基の活性化されたものとしては、 例えば、 対応する酸無水 物、 アジド、 活性エステル 〔アルコール (例えば、 ペンタクロロフエノ一ル、 2, 4, 5 -トリクロ口フエノール、 2, 4ージニトロフエノ一ル、 シァノメチルァ ルコ一ル、 パラニトロフエノール、 H〇NB、 N—ヒドロキシスクシミド、 N— ヒドロキシフタルイミド、 H'OB t) とのエステル〕 などが用いられる。 原料の ァミノ基の活性化されたものとしては、 例えば、 対応するリン酸アミドが用いら れる。  Examples of the activated carbonyl group of the raw material include, for example, a corresponding acid anhydride, azide, active ester [alcohol (for example, pentachlorophenol, 2,4,5-trichloromouth phenol, 2,4 Dinitrophenol, cyanomethyl alcohol, paranitrophenol, H〇NB, N-hydroxysuccinimide, N-hydroxyphthalimide, and ester with H'OB t). As the activated amino group of the raw material, for example, a corresponding phosphoric amide is used.
保護基の除去 (脱離) 方法としては、 例えば、 Pd—黒あるいは Pd—炭素な どの触媒の存在下での水素気流中での接触還元や、 また、 無水フッ化水素、 メタ ンスルホン酸、 トリフルォロメタンスルホン酸、 トリフルォロ酢酸あるいはこれ らの混合液などによる酸処理や、 ジイソプロピルェチルァミン、 トリェチルアミ ン、 ピぺリジン、 ピぺラジンなどによる塩基処理、 また液体アンモニア中ナトリ ゥムによる還元なども用いられる。 上記酸処理による脱離反応は、 一般に約一 2 0 °C〜4 0 °Cの温度で行なわれるが、 酸処理においては、 例えば、 ァニソ一ル、 フエノール、 チオアニソール、 メタクレゾール、 パラクレゾ一ル、 ジメチルスル フイド、 1 , 4一ブタンジチオール、 1 , 2—エタンジチオールなどのような力 チオン捕捉剤の添加が有効である。 また、 ヒスチジンのイミダゾール保護基とし て用いられる 2 , 4—ジニトロフエニル基はチォフエノール処理により除去され、 トリブトファンのインド一ル保護基として用いられるホルミル基は上記の 1, 2 —エタンジチオール、 1, 4一ブタンジチォ一ルなどの存在下の酸処理による脱 保護以外に、 希水酸化ナトリウム溶液、 希アンモニアなどによるアルカリ処理に よっても除去される。 Methods for removing (eliminating) the protecting group include, for example, catalytic reduction in a hydrogen stream in the presence of a catalyst such as Pd-black or Pd-carbon, or anhydrous hydrogen fluoride, Acid treatment with sulfonic acid, trifluoromethanesulfonic acid, trifluoroacetic acid or a mixture thereof, base treatment with diisopropylethylamine, triethylamine, piperidine, piperazine, etc. Reduction by a system is also used. The elimination reaction by the above-mentioned acid treatment is generally carried out at a temperature of about 120 ° C. to 40 ° C. In the acid treatment, for example, anisol, phenol, thioanisole, methacresol, paracresol It is effective to add a force thione scavenger such as dimethyl sulfide, 1,4-butanedithiol, 1,2-ethanedithiol and the like. The 2,4-dinitrophenyl group used as an imidazole protecting group of histidine is removed by thiophenol treatment, and the formyl group used as an indole protecting group of tributofan is 1,2-ethanedithiol, 1,4- In addition to deprotection by acid treatment in the presence of butanedithiol, etc., it is also removed by alkali treatment with dilute sodium hydroxide solution, dilute ammonia and the like.
原料の反応に関与すべきでない官能基の保護ならびに保護基、 およびその保護 基の脱離、 反応に関与する官能基の活性化などは公知の基または公知の手段から 適宜選択しうる。  The protection of the functional group which should not be involved in the reaction of the raw materials, the protecting group, the elimination of the protective group, the activation of the functional group involved in the reaction, and the like can be appropriately selected from known groups or known means.
タンパク質または部分ペプチドのアミド体を得る別の方法としては、 例えば、 まず、 カルポキシ末端アミノ酸の α—力ルポキシル基をアミド化して保護した後、 アミノ基側にペプチド (タンパク質) 鎖を所望の鎖長まで延ばした後、 該ぺプチ ド鎖の Ν末端の K—ァミノ基の保護基のみを除いたタンパク質または部分べプチ ドと C末端のカルボキシル基の保護基のみを除去したタンパク質または部分ぺプ チドとを製造し、 これらのタンパク質またはペプチドを上記したような混合溶媒 中で縮合させる。 縮合反応の詳細については上記と同様である。 縮合により得ら れた保護タンパク質またはペプチドを精製した後、 上記方法によりすベての保護 基を除去し、 所望の粗タンパク質またはペプチドを得ることができる。 この粗夕 ンパク質またはべプチドは既知の各種精製手段を駆使して精製し、 主要画分を凍 結乾燥することで所望の夕ンパク質またはべプチドのアミド体を得ることができ る。  As another method for obtaining an amide of a protein or partial peptide, for example, first, after amidating and protecting the α-hydroxyl group of the amino acid at the carboxy terminal, a peptide (protein) chain is added to the amino group side with a desired chain length. The protein or partial peptide from which only the K-amino group protecting group at the Ν-terminal of the peptide chain has been removed, and the protein or partial peptide from which only the C-terminal carboxyl group-protecting group has been removed And condensing these proteins or peptides in a mixed solvent as described above. Details of the condensation reaction are the same as described above. After purifying the protected protein or peptide obtained by the condensation, all the protecting groups are removed by the above-mentioned method, and a desired crude protein or peptide can be obtained. The crude protein or peptide can be purified using various known purification means, and the main fraction can be freeze-dried to obtain the desired amide of the protein or peptide.
タンパク質またはペプチドのエステル体を得るには、 例えば、 カルポキシ末端 アミノ酸の α—力ルポキシル基を所望のアルコール類と縮合しアミノ酸エステル とした後、 タンパク質またはペプチドのアミド体と同様にして、 所望のタンパク 質またはべプチドのエステル体を得ることができる。 To obtain an ester of a protein or peptide, for example, After condensing the α-hydroxyl group of an amino acid with a desired alcohol to form an amino acid ester, a desired protein or peptide ester can be obtained in the same manner as in the case of a protein or peptide amide.
本発明で用いられる部分ペプチドまたはそれらの塩は、 自体公知のペプチドの 合成法に従って、 あるいは本発明で用いられるタンパク質を適当なぺプチダ一ゼ で切断することによって製造することができる。 ペプチドの合成法としては、 例 えば、 固相合成法、 液相合成法のいずれによっても良い。 すなわち、 本発明で用 いられる部分べプチドを構成し得る部分べプチドもしくはアミノ酸と残余部分と を縮合させ、 生成物が保護基を有する場合は保護基を脱離することにより目的の ペプチドを製造することができる。 公知の縮合方法や保護基の脱離としては、 例 えば、 以下の①〜⑤に記載された方法が挙げられる。  The partial peptide used in the present invention or a salt thereof can be produced according to a peptide synthesis method known per se, or by cleaving the protein used in the present invention with an appropriate peptidase. As a method for synthesizing a peptide, for example, any of a solid phase synthesis method and a liquid phase synthesis method may be used. That is, a partial peptide or an amino acid capable of constituting the partial peptide used in the present invention is condensed with the remaining portion, and when the product has a protecting group, the protecting peptide is removed to produce the desired peptide. can do. Known condensation methods and elimination of protecting groups include, for example, the methods described in the following ① to ⑤.
(DM. Bodanszky および M. A. Ondet t i、 ペプチド 'シンセシス (Pept i de Synthes i s) , Intersc i ence Pub l i shers, New York (1966%; (DM. Bodanszky and M.A. Ondet ti, Peptide de Synthesis (Pepti de Synthes is), Interscience Pub lishers, New York (1966%;
② Schroederおよび Luebke、 ザ ·ペプチド(The Pept i de) , Academic Press, New York (1965年)  ② Schroeder and Luebke, The Peptide, Academic Press, New York (1965)
③泉屋信夫他、 ペプチド合成の基礎と実験、 丸善 (株) (1975年)  (3) Nobuo Izumiya et al. Basics and experiments on peptide synthesis, Maruzen Co., Ltd. (1975)
④矢島治明 および榊原俊平、 生化学実験講座 1、 タンパク質の化学 IV、 205、 (1977年)  治 Haruaki Yajima and Shunpei Sakakibara, Laboratory for Biochemical Experiments 1, Protein Chemistry IV, 205, (1977)
⑤矢島治明監修、 続医薬品の開発、 第 14巻、 ペプチド合成、 広川書店  治 Supervised by Haruaki Yajima, Development of Continuing Drugs, Volume 14, Peptide Synthesis, Hirokawa Shoten
また、 反応後は通常の精製法、 例えば、 溶媒抽出 ·蒸留 ·カラムクロマトグラ フィ一'液体クロマトグラフィー '再結晶などを組み合わせて本発明で用いられ る部分べプチドを精製単離することができる。 上記方法で得られる部分べプチド が遊離体である場合は、 公知の方法あるいはそれに準じる方法によって適当な塩 に変換することができるし、 逆に塩で得られた場合は、 公知の方法あるいはそれ に準じる方法によって遊離体または他の塩に変換することができる。  After the reaction, the partial peptide used in the present invention can be purified and isolated by a combination of ordinary purification methods such as solvent extraction, distillation, column chromatography, 'liquid chromatography', and recrystallization. . When the partial peptide obtained by the above method is a free form, it can be converted to an appropriate salt by a known method or a method analogous thereto, and conversely, when the partial peptide is obtained by a salt, the known method or Can be converted into a free form or another salt by a method according to the above.
本発明で用いられるタンパク質をコードするポリヌクレオチドとしては、 前述 した本発明で用いられるタンパク質をコードする塩基配列を含有するものであれ ばいかなるものであってもよい。 好ましくは D NAである。 D NAとしては、 ゲ ノム D NA、 ゲノム D NAライブラリー、 前記した細胞'組織由来の c D NA、 前記した細胞 ·組織由来の c D NAライブラリー、 合成 D N Aのいずれでもよい。 ライブラリ一に使用するべクタ一は、 パクテリオファージ、 プラスミド、 コス ミド、 ファージミドなどいずれであってもよい。 また、 前記した細胞 '組織より total R N Aまたは mR N A画分を調製したものを用いて直接 Reverse The polynucleotide encoding the protein used in the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the protein used in the present invention. Preferably it is DNA. Examples of the DNA include genomic DNA, genomic DNA library, the above-described cell-tissue-derived cDNA, Any of the above-described cell / tissue-derived cDNA library and synthetic DNA may be used. The vector used for the library may be any of pacteriophage, plasmid, cosmid, phagemid and the like. In addition, reverse RNA was directly prepared using a preparation of total RNA or mRNA fraction from the above-mentioned cell 'tissue.
Transcriptase Polymerase Chain React ion (以下、 R T— P C R法と略称す る) によって増幅することもできる。 It can also be amplified by transcriptase polymerase chain reaction (hereinafter abbreviated as RT-PCR method).
本発明で用いられるタンパク質をコ一ドする D N Aとしては、 例えば、 配列番 号: 2で表される塩基配列を含有する D NA、 または配列番号: 2で表される塩 基配列とハイストリンジェントな条件下でハイブリダィズする塩基配列を含有し、 前記した配列番号: 1で表されるアミノ酸配列を含有するタンパク質と実質的に 同質の性質を有するタンパク質をコ一ドする D NAであれば何れのものでもよい。 配列番号: 2で表される塩基配列と八イストリンジエンドな条件下でハイプリ ダイズできる D NAとしては、 例えば、 配列番号: 2で表される塩基配列と約 5 0 %以上、 好ましくは約 6 0 %以上、 さらに好ましくは約 7 0 %以上、 より好ま しくは約 8 0 %以上、 特に好ましくは約 9 0 %以上、 最も好ましくは約 9 5 %以 上の相同性を有する塩基配列を含有する D N Aなどが用いられる。  Examples of the DNA encoding the protein used in the present invention include a DNA containing the base sequence represented by SEQ ID NO: 2 or the base sequence represented by SEQ ID NO: 2 and high stringency. Any DNA that contains a nucleotide sequence that hybridizes under the following conditions and encodes a protein having substantially the same properties as the protein containing the amino acid sequence represented by SEQ ID NO: 1 described above. It may be something. Examples of the DNA that can hybridize with the nucleotide sequence represented by SEQ ID NO: 2 under conditions of eight istrines include, for example, about 50% or more, preferably about 6% with the nucleotide sequence represented by SEQ ID NO: 2. 0% or more, more preferably about 70% or more, more preferably about 80% or more, particularly preferably about 90% or more, most preferably about 95% or more. DNA or the like is used.
ハイブリダィゼーシヨンは、 自体公知の方法あるいはそれに準じる方法、 例え は、 Molecular Cloning 2nd (J. Saibrook et al . , Cold Spring Harbor Lab. Press, 1989) に記載の方法などに従って行なうことができる。 また、 市販のラ イブラリーを使用する場合、 添付の使用説明書に記載の方法に従って行なうこと ができる。 より好ましくは、 ハイストリンジエンドな条件に従って行なうことが できる。  Hybridization can be performed according to a method known per se or a method analogous thereto, for example, the method described in Molecular Cloning 2nd (J. Saibrook et al., Cold Spring Harbor Lab. Press, 1989). When a commercially available library is used, it can be performed according to the method described in the attached instruction manual. More preferably, the reaction can be performed under high stringency conditions.
ハイストリンジエンドな条件とは、 例えば、 ナトリウム濃度が約 1 9〜4 0 m M、 好ましくは約 1 9〜 2 0 mMで、 温度が約 5 0〜7 0 :、 好ましくは約 6 0 〜 6 5 °Cの条件を示す。 特に、 ナトリゥム濃度が約 1 9 mMで温度が約 6 5 °Cの 場合が最も好ましい。  High stringent end conditions include, for example, a sodium concentration of about 19 to 40 mM, preferably about 19 to 20 mM, and a temperature of about 50 to 70: preferably about 60 to 6 The conditions at 5 ° C are shown. In particular, the case where the sodium concentration is about 19 mM and the temperature is about 65 ° C. is most preferable.
より具体的には、 配列番号: 1で表されるアミノ酸配列を含有するタンパク質 をコードする D NAとしては、 配列番号: 2で表される塩基配列を含有する D N Aなどが用いられる。 本発明で用いられる部分ペプチドをコードするポリヌクレオチド (例、 D N A) としては、 前述した本発明で用いられる部分ペプチドをコードする塩基配列 を含有するものであればいかなるものであってもよい。 また、 ゲノム D NA、 ゲ ノム D NAライブラリー、 前記した細胞 ·組織由来の c D NA、 前記した細胞 · 組織由来の c D N Aライブラリー、 合成 D N Aのいずれでもよい。 More specifically, as the DNA encoding the protein containing the amino acid sequence represented by SEQ ID NO: 1, a DNA containing the base sequence represented by SEQ ID NO: 2 or the like is used. The polynucleotide (eg, DNA) encoding the partial peptide used in the present invention may be any polynucleotide containing the above-described nucleotide sequence encoding the partial peptide used in the present invention. In addition, any of a genomic DNA, a genomic DNA library, the above-described cell / tissue-derived cDNA, the above-described cell / tissue-derived cDNA library, and synthetic DNA may be used.
本発明で用いられる部分ペプチドをコードする D NAとしては、 例えば、 配列 番号: 2で表される塩基配列を含有する D N Aの一部分を含有する D NA、 また は配列番号: 2で表される塩基配列とハイストリンジェン卜な条件下でハイプリ ダイズする塩基配列を含有し、 本発明のタンパク質と実質的に同質の活性を有す るタンパク質をコードする D N Aの一部分を含有する D N Aなどが用いられる。 配列番号: 2で表される塩基配列と八イブリダィズできる D NAは、 前記と同 意義を示" 5—。  Examples of the DNA encoding the partial peptide used in the present invention include, for example, a DNA containing a part of a DNA containing the base sequence represented by SEQ ID NO: 2, or a base represented by SEQ ID NO: 2. A DNA containing a base sequence that hybridizes with the sequence under high stringent conditions, and a DNA containing a part of a DNA encoding a protein having substantially the same activity as the protein of the present invention is used. The DNA that can be hybridized with the base sequence represented by SEQ ID NO: 2 has the same meaning as described above.
ハイブリダィゼ一シヨンの方法およびハイストリンジェントな条件は前記と同 様のものが用いられる。  The same hybridization method and high stringency conditions as described above are used.
本発明で用いられるタンパク質、 部分ペプチド (以下、 これらをコ一ドする D N Aのクローニングおよび発現の説明においては、 これらを単に本発明のタンパ ク質と略記する場合がある) を完全にコードする D NAのクローエングの手段と しては、 本発明のタンパク質をコードする塩基配列の一部分を含有する合成 D N Aプライマーを用いて P C R法によって増幅するか、 または適当なベクターに組 み込んだ D N Aを本発明のタンパク質の一部あるいは全領域をコードする D NA 断片もしくは合成 D NAを用いて標識したものとのハイブリダィゼーションによ つて選別することができる。 ハイブリダィゼ一シヨンの方法は、 例えば、  D which completely encodes the protein and partial peptide used in the present invention (hereinafter, these may be simply referred to as the protein of the present invention in the description of cloning and expression of the DNA encoding them). As a means for closing NA, DNA amplified by a PCR method using a synthetic DNA primer containing a part of the nucleotide sequence encoding the protein of the present invention, or a DNA incorporated into an appropriate vector is used as the DNA of the present invention. Can be selected by hybridization with a DNA fragment coding for a part or the entire region of the above-mentioned protein or labeled with a synthetic DNA. The method of hybridization is, for example,
Molecul ar Cl oning 2nd (J. Sambrook et al . , Cold Spring Harbor Lab. Press, 1989) に記載の方法などに従って行なうことができる。 また、 市販のライブラリ 一を使用する場合、 添付の使用説明書に記載の方法に従って行なうことができる。 Molecular Cloning 2nd (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). When a commercially available library is used, it can be performed according to the method described in the attached instruction manual.
D NAの塩基配列の変換は、 P C R、 公知のキット、 例えば、 Mutan™-super Express Km (宝酒造 (株) ) 、 MutanTM-K (宝酒造 (株) ) 等を用いて、 ODA- LA PCR法、 Gapped duplex法、 Kunkel法等の自体公知の方法あるいはそれらに準じる 方法に従って行なうことができる。 クローン化されたタンパク質をコードする DNAは目的によりそのまま、 また は所望により制限酵素で消化したり、 リンカーを付加したりして使用することが できる。 該 DNAはその 5' 末端側に翻訳開始コドンとしての ATGを有し、 ま た 3' 末端側には翻訳終止コドンとしての T A A、 TGAまたは TAGを有して いてもよい。 これらの翻訳開始コドンや翻訳終止コドンは、 適当な合成 DNAァ ダブ夕一を用いて付加することもできる。 The DNA base sequence can be converted using the ODA-LA PCR method using PCR, a known kit, for example, Mutan ™ -super Express Km (Takara Shuzo), Mutan TM- K (Takara Shuzo), or the like. , Gapped duplex method, Kunkel method and the like, or a method analogous thereto. The DNA encoding the cloned protein can be used as it is depending on the purpose, or can be used after digesting with a restriction enzyme or adding a linker, if desired. The DNA may have ATG as a translation initiation codon at the 5 'end and TAA, TGA or TAG as a translation termination codon at the 3' end. These translation initiation codon and translation termination codon can also be added using an appropriate synthetic DNA adapter.
本発明のタンパク質の発現ベクターは、 例えば、 (ィ) 本発明のタンパク質を コードする DNAから目的とする DNA断片を切り出し、 (口) 該 DNA断片を 適当な発現ベクター中のプロモーターの下流に連結することにより製造すること ができる。  The expression vector for the protein of the present invention includes, for example, (a) cutting out a DNA fragment of interest from DNA encoding the protein of the present invention, and (mouth) ligating the DNA fragment downstream of a promoter in an appropriate expression vector. It can be manufactured by
ベクターとしては、 大腸菌由来のプラスミド (例、 pBR322, pBR 32 5, pUC 12, pUC 13) 、 枯草菌由来のプラスミド (例、 pUB 110, pTP 5, p C 194) 、 酵母由来プラスミド (例、 p SH 19, p SH 15) 、 λファージなどのパクテリオファージ、 レトロウイルス, ワクシニアウィルス, バキュロウィルスなどの動物ウィルスなどの他、 pAl— 11、 pXTl、 pR c/CMV、 pRcZRSV、 p cDNA I /N e oなどが用いられる。  Examples of the vector include a plasmid derived from Escherichia coli (eg, pBR322, pBR325, pUC12, pUC13), a plasmid derived from Bacillus subtilis (eg, pUB110, pTP5, pC194), a plasmid derived from yeast (eg, p SH19, pSH15), pacteriophage such as phage λ, animal viruses such as retrovirus, vaccinia virus, baculovirus, etc., pAl-11, pXTl, pRc / CMV, pRcZRSV, pcDNA I / N eo or the like is used.
本発明で用いられるプロモーターとしては、 遺伝子の発現に用いる宿主に対 応して適切なプロモーターであればいかなるものでもよい。 例えば、 動物細胞を 宿主として用いる場合は、 SRo;プロモ一夕一、 SV40プロモータ一、 LTR プロモータ一、 CMVプロモーター、 HS V- TKプロモータ一などが挙げられ る。  The promoter used in the present invention may be any promoter as long as it is appropriate for the host used for gene expression. For example, when an animal cell is used as a host, SRo; Promoter, SV40 promoter, LTR promoter, CMV promoter, HSV-TK promoter and the like can be mentioned.
これらのうち、 CMV (サイトメガロウィルス) プロモータ一、 SRo;プロ モーターなどを用いるのが好ましい。 宿主がェシエリヒア属菌である場合は、 t r pプロモータ一、 l a cプロモーター、 r e cAプロモーター、 APLプロモ —ター、 1 ρ Pプロモータ一、 T7プロモーターなどが、 宿主がバチルス属菌で ある場合は、 SP01プロモ一夕一、 SPO 2プロモーター、 penPプロモ一 夕一など、 宿主が酵母である場合は、 PHO 5プロモーター、 PGKプロモータ 一、 GAPプロモータ一、 ADHプロモータ一などが好ましい。 宿主が昆虫細胞 である場合は、 ポリヘドリンプロモーター、 P 10プロモータ一などが好ましい。 発現ベクターには、 以上の他に、 所望によりェンハンサ一、 スプライシシグシ ダナル、 ポリ A付加シグナル、 選択マ一力一、 SV40複製オリジン (以下、 S V40 o r iと略称する場合がある) などを含有しているものを用いることがで きる。 選択マ一力一としては、 例えば、 ジヒドロ葉酸還元酵素 (以下、 dh f r と略称する場合がある) 遺伝子 〔メソトレキセ一ト (MTX) 耐性〕 、 アンピシ リン耐性遺伝子 (以下、 Amp1"と略称する場合がある) 、 ネオマイシン耐性遺 伝子 (以下、 Ne orと略称する場合がある、 G418耐性) 等が挙げられる。 特に、 dh f r遺伝子欠損チャイニーズハムスター細胞を用いて dh f r遺伝子 を選択マーカ一として使用する場合、 目的遺伝子をチミジンを含まない培地によ つても選択できる。 Of these, it is preferable to use the CMV (cytomegalovirus) promoter, SRo; When the host is Eshierihia genus bacterium, trp promoter one, lac promoter, re cA promoter, AP L promoter - ter, 1 [rho P promoter one, such as T7 promoter, if the host is a strain of the genus Bacillus, SP01 promoter When the host is yeast, such as overnight, SPO2 promoter, penP promoter, and the like, PHO5 promoter, PGK promoter, GAP promoter, ADH promoter, and the like are preferable. When the host is an insect cell, a polyhedrin promoter, a P10 promoter and the like are preferable. The expression vector may contain, in addition to the above, an enhancer, a splicing signal, a poly-A addition signal, a selection promoter, and an SV40 replication origin (hereinafter sometimes abbreviated as SV40 ori), if desired. Can be used. Selection methods include, for example, a dihydrofolate reductase (hereinafter sometimes abbreviated as dh fr) gene [methotrexate (MTX) resistance], an ampicillin resistance gene (hereinafter abbreviated as Amp 1 ) If there is), the neomycin resistance gene (hereinafter, Ne o sometimes abbreviated as r, G418 resistance), etc. in the like. in particular, dh fr gene deficient Chinese hamster cell select dh fr gene using the marker one When used as a target gene, the target gene can also be selected on a thymidine-free medium.
また、 必要に応じて、 宿主に合ったシグナル配列を、 本発明のタンパク質の N 端末側に付加する。 宿主がェシエリヒア属菌である場合は、 PhoA'シグナル配列、 OmpA*シグナル配列などが、 宿主がバチルス属菌である場合は、 α—アミラー ゼ ·シグナル配列、 サブチリシン ·シグナル配列などが、 宿主が酵母である場合 は、 MFo! ·シグナル配列、 SUC2 ·シグナル配列など、 宿主が動物細胞であ る場合には、 インシュリン 'シグナル配列、 ひ一インターフェロン ·シグナル配 列、 抗体分子 ·シグナル配列などがそれぞれ利用できる。  If necessary, a signal sequence suitable for the host is added to the N-terminal side of the protein of the present invention. When the host is a genus Escherichia, the PhoA 'signal sequence and the OmpA * signal sequence are used.When the host is a Bacillus genus, the α-amylase signal sequence and subtilisin signal sequence are used. Signal sequence, SUC2 signal sequence, etc. If the host is an animal cell, use insulin 'signal sequence, Hi-I interferon signal sequence, antibody molecule, signal sequence, etc. it can.
このようにして構築された本発明のタンパク質をコードする DNAを含有する ベクターを用いて、 形質転換体を製造することができる。  Using the vector containing the DNA encoding the protein of the present invention thus constructed, a transformant can be produced.
宿主としては、 例えば、 ェシエリヒア属菌、 バチルス属菌、 酵母、 昆虫細胞、 昆虫、 動物細胞などが用いられる。  As the host, for example, Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells, and the like are used.
ェシエリヒア属菌の具体例としては、 例えば、 ェシエリヒア 'コリ  Specific examples of the genus Escherichia include, for example, Escherichia coli.
(Escherichia coli) K12 - DH 1 [Proc. Natl. Acad. Sci. USA, 60 巻, 160(1968)〕 , JM103 [Nucleic Acids Research, 9巻, 309 (1981)〕 , J A 221 [Journal of Molecular Biology, 120巻, 517 (1978)〕 , HB 101  (Escherichia coli) K12-DH 1 [Proc. Natl. Acad. Sci. USA, 60, 160 (1968)], JM103 [Nucleic Acids Research, 9, 309 (1981)], JA 221 [Journal of Molecular Biology] , 120, 517 (1978)], HB 101
[Journal of Molecular Biology, 41巻, 459(1969)〕 , C 600 [Genetics, 39 巻, 440 (1954)〕 などが用いられる。  [Journal of Molecular Biology, 41, 459 (1969)], C600 [Genetics, 39, 440 (1954)] and the like are used.
バチルス属菌としては、 例えば、 バチルス 'サブチルス (Bacillus  Examples of Bacillus bacteria include, for example, Bacillus' subtilis (Bacillus
snbtilis) M I 1 14 〔Gene, 24巻, 255 (1983)〕 , 207 -21 [Journal of 03007926 snbtilis) MI 1 14 [Gene, 24, 255 (1983)], 207-21 [Journal of 03007926
21  twenty one
Biochemistry, 95卷, 87(1984)] などが用いられる。 Biochemistry, 95, 87 (1984)].
酵母としては、 例えば、 サッカロマイセス セレピシェ (Saccharomyces cerevisiae) AH 22, AH22R—, NA87 - 11 A, DKD— 5D, 20 B— 12、 シゾサッカロマイセス ボンべ (Schizosaccharomyces pombe) NC YC 1913, NCYC 2036、 ピキア パストリス (Pichia pastor is) K M71などが用いられる。  Examples of yeast include Saccharomyces cerevisiae AH22, AH22R—, NA87-11A, DKD—5D, 20B—12, Schizosaccharomyces pombe NC YC 1913, NCYC2036, and Pichia pastoris (Pichia pastor is) K M71 or the like is used.
昆虫細胞としては、 例えば、 ウィルスが Ac NPVの場合は、 夜盗蛾の幼虫由 来株化細胞 (Spodoptera frugiperda cell; S ί細胞) 、 Trichoplusia niの中 腸由来の MG1細胞、 Trichoplusia niの卵由来の High Five™細胞、 Mamestra brassicae由来の細胞または Estigmena acrea由来の細胞などが用いられる。 ウイ ルスが BmNPVの場合は、 蚕由来株化細胞 (Bombyx mori N細胞; BmN細 胞) などが用いられる。 該 S f細胞としては、 例えば、 S f 9細胞 (ATCC CRL1711) 、 S f 21細胞 (以上、 Vaughn, J.L.ら、 In Vivo, 13, 213- 217,(1977)) などが用いられる。 , 昆虫としては、 例えば、 カイコの幼虫などが用いられる 〔前田ら、 Nature, 315 卷, 592 (1985)〕 。  As insect cells, for example, when the virus is Ac NPV, the cell line derived from the larvae of the night moth (Spodoptera frugiperda cell; S ; cell), the MG1 cell derived from the midgut of Trichoplusia ni, High Five ™ cells, cells derived from Mamestra brassicae or cells derived from Estigmena acrea are used. When the virus is BmNPV, a cell line derived from silkworm (Bombyx mori N cell; BmN cell) is used. As the Sf cells, for example, Sf9 cells (ATCC CRL1711), Sf21 cells (Vaughn, J.L., et al., In Vivo, 13, 213-217, (1977)) and the like are used. As insects, for example, silkworm larvae are used [Maeda et al., Nature, Vol. 315, 592 (1985)].
動物細胞としては、 例えば、 サル細胞 COS— 7, Ve r o, チャイニーズノ、 ムスター細胞 CHO (以下、 CHO細胞と略記) , dh f r遺伝子欠損チヤィニ —ズハムスター細胞 CHO (以下、 CHO (dh f r") 細胞と略記) , マウス L細胞, マウス At T— 20, マウスミエローマ細胞, ラット GH3, ヒト FL 細胞などが用いられる。  As animal cells, for example, monkey cell COS-7, Vero, Chinese cell, Muster cell CHO (hereinafter abbreviated as CHO cell), dh fr gene-deficient chinini-zhamster cell CHO (hereinafter, CHO (dh fr)) Cells), mouse L cells, mouse AtT-20, mouse myeloma cells, rat GH3, human FL cells, and the like.
ェシエリヒア属菌を形質転換するには、 例えば、 Proc. Natl. Acad. Sci. USA, 69巻, 2110 (1972)や Gene, 17巻, 107 (1982)などに記載の方法に従って行なうこ とができる。  Natl. Acad. Sci. USA, 69, 2110 (1972) and Gene, 17, 107 (1982) can be used to transform Escherichia species. .
バチルス属菌を形質転換するには、 例えば、 Molecular & General  To transform Bacillus, for example, use Molecular & General
Genetics, 168巻, 111 (1979)などに記載の方法に従つて行なうことができる。  Genetics, vol. 168, 111 (1979) can be used.
酵母を形質転換するには、 例えば、 Methods in Enzymology, 194巻, 182- 187(1991)、 Proc. Natl. Acad. Sci. USA, 75巻, 1929 (1978)などに記載の方法に 従って行なうことができる。 昆虫細胞または昆虫を形質転換するには、 例えば、 Bio/Technology,6, 47 - 55 (1988)などに記載の方法に従つて行なうことができる。 The yeast can be transformed according to the method described in, for example, Methods in Enzymology, Vol. 194, 182-187 (1991), Proc. Natl. Acad. Sci. USA, Vol. 75, 1929 (1978). Can be. Insect cells or insects can be transformed, for example, according to the method described in Bio / Technology, 6, 47-55 (1988).
動物細胞を形質転換するには、 例えば、 細胞工学別冊 8 新細胞工学実験プロ トコール. 263-267(1995) (秀潤社発行) 、 Virology, 52巻, 456 (1973)に記載の方 法に従って行なうことができる。  To transform animal cells, for example, follow the method described in Cell Engineering Separate Volume 8, New Cell Engineering Experimental Protocol. 263-267 (1995) (published by Shujunsha), Virology, 52, 456 (1973). Can do it.
このようにして、 タンパク質をコードする DNAを含有する発現ベクターで形 質転換された形質転換体を得ることができる。  Thus, a transformant transformed with the expression vector containing the DNA encoding the protein can be obtained.
宿主がェシエリヒア属菌、 バチルス属菌である形質転換体を培養する際、 培養 に使用される培地としては液体培地が適当であり、 その中には該形質転換体の生 育に必要な炭素源、 窒素源、 無機物その他が含有せしめられる。 炭素源としては、 例えば、 グルコース、 デキストリン、 可溶性澱粉、 ショ糖など、 窒素源としては、 例えば、 アンモニゥム塩類、 硝酸塩類、 コーンスチ一プ ·リカー、 ペプトン、 力 ゼイン、 肉エキス、 大豆粕、 バレイショ抽出液などの無機または有機物質、 無機 物としては、 例えば、 塩化カルシウム、 リン酸二水素ナトリウム、 塩化マグネシ ゥムなどが挙げられる。 また、 酵母エキス、 ビタミン類、 成長促進因子などを添 加してもよい。 培地の pHは約 5〜8が望ましい。  When culturing a transformant whose host is a bacterium belonging to the genus Escherichia or Bacillus, a liquid medium is suitable as a medium used for the cultivation, and a carbon source necessary for the growth of the transformant is contained therein. , Nitrogen sources, inorganic substances and others. Examples of carbon sources include glucose, dextrin, soluble starch, and sucrose. Examples of nitrogen sources include ammonium salts, nitrates, corn chip liquor, peptone, potato zein, meat extract, soybean meal, and potato extract. Examples of the inorganic or organic substance such as a liquid and the inorganic substance include calcium chloride, sodium dihydrogen phosphate, and magnesium chloride. In addition, yeast extract, vitamins, growth promoting factors and the like may be added. The pH of the medium is preferably about 5-8.
ェシエリヒア属菌を培養する際の培地としては、 例えば、 グルコース、 カザミ ノ酸を含む M9培地 [Miller, Journal of Experiments in Molecular Genetics, 431-433, Cold Spring Harbor Laboratory, New York 1972] が好ましい。 ここ に必要によりプロモータ一を効率よく働かせるために、 例えば、 3 /3—インドリ ルアクリル酸のような薬剤を加えることができる。  As a medium for culturing the genus Escherichia, for example, an M9 medium containing glucose and casamino acids [Miller, Journal of Experiments in Molecular Genetics, 431-433, Cold Spring Harbor Laboratory, New York 1972] is preferable. Here, for example, a drug such as 3 / 3-indolylacrylic acid can be added to make the promoter work efficiently if necessary.
宿主がェシエリヒア属菌の場合、 培養は通常約 1 5〜43°Cで約 3〜24時間 行ない、 必要により、 通気や撹拌を加えることもできる。  When the host is a bacterium belonging to the genus Escherichia, the cultivation is usually performed at about 15 to 43 ° C for about 3 to 24 hours, and if necessary, aeration or stirring may be added.
宿主がバチルス属菌の場合、 培養は通常約 30〜40 で約6〜24時間行な レ 、 必要により通気や撹拌を加えることもできる。  When the host is a bacterium belonging to the genus Bacillus, the cultivation is usually performed at about 30 to 40 for about 6 to 24 hours, and if necessary, aeration and stirring may be added.
宿主が酵母である形質転換体を培養する際、 培地としては、 例えば、 バークホ —ルダ一 (Burkholder) 最小培地 CBostian, K. L. ら、 Proc. Natl. Acad. Sci. USA, 77巻, 4505 (1980)〕 や 0.5 %カザミノ酸を含有する S D培地 〔Bitter, G. A. ら、 Proc. Natl. Acad. Sci. USA, 81巻, 5330 (1984)〕 が挙げられる。 培地の pH は約 5〜 8に調整するのが好ましい。 培養は通常約 20〜35!0で約24〜72 時間行ない、 必要に応じて通気や撹拌を加える。 When culturing a transformant in which the host is yeast, for example, Burkholder's minimum medium (CBostian, KL et al., Proc. Natl. Acad. Sci. USA, 77, 4505 (1980)) And a SD medium containing 0.5% casamino acid [Bitter, GA et al., Proc. Natl. Acad. Sci. USA, 81, 5330 (1984)]. Medium pH Is preferably adjusted to about 5 to 8. The culture is carried out usually at about 20 to 35! 0 to about 24 to 72 hours, with aeration or agitation if necessary.
宿主が昆虫細胞または昆虫である形質転換体を培養する際、 培地としては、 Grace's Insect Medium (Grace, T.C.C. , Nature, 195,788 (1962)) に非動化した 10%ゥシ血清等の添加物を適宜加えたものなどが用いられる。 培地の pHは約 6. 2〜 6. 4に調整するのが好ましい。 培養は通常約 27 °Cで約 3〜 5日間行 ない、 必要に応じて通気や撹拌を加える。  When culturing an insect cell or a transformant in which the host is an insect, the culture medium is supplemented with Grace's Insect Medium (Grace, TCC, Nature, 195,788 (1962)). Those appropriately added are used. The pH of the medium is preferably adjusted to about 6.2 to 6.4. Culture is usually performed at about 27 ° C for about 3 to 5 days, and aeration and agitation are added as necessary.
宿主が動物細胞である形質転換体を培養する際、 培地としては、 例えば、 約 5 〜20 %の胎児牛血清を含む MEM培地 [Science, 122巻, 501 (1952)〕 , DME M培地 [Virology, 8巻, 396(1959)〕 , RPM I 1640培地 〔The 】ournal of the American Medical Association 199卷, 519 (1967)〕 , 199培地  When culturing a transformant in which the host is an animal cell, for example, a MEM medium containing about 5 to 20% fetal bovine serum [Science, 122, 501 (1952)], a DMEM medium [Virology , Vol. 8, 396 (1959)], RPMI 1640 medium [The] journal of the American Medical Association 199, 519 (1967)], 199 medium
[Proceeding of the Society for the Biological Medicine, 73巻, 1 (1950)〕 な どが用いられる。 pHは約 6〜8であるのが好ましい。 培養は通常約 30〜4 0°Cで約 15~60時間行ない、 必要に応じて通気や撹拌を加える。  [Proceeding of the Society for the Biological Medicine, 73, 1 (1950)]. Preferably, the pH is about 6-8. The cultivation is usually performed at about 30 to 40 ° C for about 15 to 60 hours, and aeration and stirring are added as necessary.
以上のようにして、 形質転換体の細胞内、 細胞膜または細胞外に本発明のタン パク質を生成せしめることができる。  As described above, the protein of the present invention can be produced in the cells, in the cell membrane, or outside the cells of the transformant.
上記培養物から本発明のタンパク質を分離精製するには、 例えば、 下記の方法 により行なうことができる。  The protein of the present invention can be separated and purified from the culture by, for example, the following method.
本発明のタンパク質を培養菌体あるいは細胞から抽出するに際しては、 培養後、 公知の方法で菌体あるいは細胞を集め、 これを適当な緩衝液に懸濁し、 超音波、 リゾチームおよび Zまたは凍結融解などによって菌体あるいは細胞を破壊したの ち、 遠心分離やろ過によりタンパク質の粗抽出液を得る方法などが適宜用いられ る。 緩衝液の中に尿素や塩酸グァニジンなどのタンパク質変性剤や、 トリトン X 一 100™などの界面活性剤が含まれていてもよい。 培養液中にタンパク質が 分泌される場合には、 培養終了後、 それ自体公知の方法で菌体あるいは細胞と上 清とを分離し、 上清を集める。  When extracting the protein of the present invention from cultured cells or cells, after culturing, the cells or cells are collected by a known method, suspended in an appropriate buffer, and subjected to sonication, lysozyme and Z or freeze-thawing. After the cells or cells are destroyed by the method, a method of obtaining a crude extract of the protein by centrifugation or filtration is appropriately used. The buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant such as Triton X-1100 ™. When the protein is secreted into the culture solution, after completion of the culture, the cells or cells are separated from the supernatant by a method known per se, and the supernatant is collected.
このようにして得られた培養上清、 あるいは抽出液中に含まれるタンパク質の 精製は、 自体公知の分離。精製法を適切に組み合わせて行なうことができる。 こ れらの公知の分離、 精製法としては、 塩析ゃ溶媒沈澱法などの溶解度を利用する P T/JP2003/007926 Purification of the protein contained in the culture supernatant or extract obtained in this manner is a separation known per se. Purification methods can be combined appropriately. As these known separation and purification methods, solubility such as salting-out and solvent precipitation is used. PT / JP2003 / 007926
24 方法、 透析法、 限外ろ過法、 ゲルろ過法、 および S D S—ポリアクリルアミドゲ ル電気泳動法などの主として分子量の差を利用する方法、 イオン交換クロマトグ ラフィ一などの荷電の差を利用する方法、 ァフィ二ティークロマトグラフィーな どの特異的親和性を利用する方法、 逆相高速液体ク口マトグラフィ一などの疎水 性の差を利用する方法、 等電点電気泳動法などの等電点の差を利用する方法など が用いられる。  24 methods, methods that mainly use differences in molecular weight, such as dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis, and methods that use differences in charge, such as ion-exchange chromatography. Methods that use specific affinity such as affinity chromatography, methods that use differences in hydrophobicity such as reversed-phase high-performance liquid chromatography, and methods that use differences in isoelectric points such as isoelectric focusing. The method used is used.
かくして得られるタンパク質が遊離体で得られた場合には、 自体公知の方法あ るいはそれに準じる方法によって塩に変換することができ、 逆に塩で得られた場 合には自体公知の方法あるいはそれに準じる方法により、 遊離体または他の塩に 変換することができる。  When the protein thus obtained is obtained as a free form, it can be converted to a salt by a method known per se or a method analogous thereto, and conversely, when the protein is obtained as a salt, a method known per se or It can be converted to a free form or other salts by a method analogous thereto.
なお、 組換え体が産生するタンパク質を、 精製前または精製後に適当な蛋白修 飾酵素を作用させることにより、 任意に修飾を加えたり、 ポリペプチドを部分的 に除去することもできる。 蛋白修飾酵素としては、 例えば、 トリプシン、 キモト リプシン、 アルギニルエンドべプチダ一ゼ、 プロテインキナーゼ、 グリコシダー ゼなどが用いられる。  The protein produced by the recombinant can be arbitrarily modified or the polypeptide can be partially removed by the action of an appropriate protein modifying enzyme before or after purification. As the protein modifying enzyme, for example, trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, glycosidase and the like are used.
かくして生成する本発明のタンパク質の存在は、 特異抗体を用いたェンザィム ィムノアツセィゃウエスタンプロッティングなどにより測定することができる。 本発明で用いられるタンパク質もしくは部分ペプチドまたはその塩に対する抗 体は、 本発明で用いられるタンパク質もしくは部分べプチドまたはその塩を認識 し得る抗体であれば、 ポリクロ一ナル抗体、 モノクローナル抗体の何れであって もよい。  The presence of the protein of the present invention thus produced can be measured by, for example, enzymatic immunoassay western blotting using a specific antibody. The antibody against the protein or partial peptide or a salt thereof used in the present invention is any of a polyclonal antibody and a monoclonal antibody as long as it can recognize the protein or partial peptide or a salt thereof used in the present invention. You may.
本発明で用いられるタンパク質もしくは部分ペプチドまたはその塩 (以下、 抗 体の説明においては、 これらを単に本発明のタンパク質と略記する場合がある) に対する抗体は、 本発明のタンパク質を抗原として用い、 自体公知の抗体または 抗血清の製造法に従つて製造することができる。  An antibody against the protein or partial peptide used in the present invention or a salt thereof (hereinafter sometimes simply referred to as the protein of the present invention in the description of the antibody) uses the protein of the present invention as an antigen, It can be produced according to a known antibody or antiserum production method.
〔モノクローナル抗体の作製〕  [Preparation of monoclonal antibody]
( a ) モノクローナル抗体産生細胞の作製  (a) Preparation of monoclonal antibody-producing cells
本発明のタンパク質は、 温血動物に対して投与により抗体産生が可能な部位に それ自体あるいは担体、 希釈剤とともに投与される。 投与に際して抗体産生能を TJP2003/007926 The protein of the present invention is administered to a warm-blooded animal itself or together with a carrier or diluent at a site capable of producing an antibody upon administration. Antibody production ability upon administration TJP2003 / 007926
25 高めるため、 完全フロイントアジュバントや不完全フロイントアジュバントを投 与してもよい。 投与は通常 2〜 6週毎に 1回ずつ、 計 2〜10回程度行われる。 用いられる温血動物としては、 例えば、 サル、 ゥサギ、 ィヌ、 モルモット、 マウ ス、 ラット、 ヒッジ、 ャギ、 ニヮトリが挙げられるが、 マウスおよびラットが好 ましく用いられる。  25 Complete Freund's adjuvant or incomplete Freund's adjuvant may be given to increase the level. Administration is usually performed once every 2 to 6 weeks, for a total of 2 to 10 times. Examples of the warm-blooded animal to be used include monkeys, egrets, dogs, guinea pigs, mice, rats, sheep, goats, and chickens, and mice and rats are preferably used.
モノクローナル抗体産生細胞の作製に際しては、 抗原で免疫された温血動物、 例えばマウスから抗体価の認められた個体を選択し最終免疫の 2〜 5日後に脾臓 またはリンパ節を採取し、 それらに含まれる坊体産生細胞を同種または異種動物 の骨髄腫細胞と融合させることにより、 モノクローナル抗体産生ハイプリドーマ を調製することができる。 抗血清中の抗体価の測定は、 例えば、 後記の標識化夕 ンパク質と抗血清とを反応させたのち、 抗体に結合した標識剤の活性を測定する ことにより行なうことができる。 融合操作は既知の方法、 例えば、 ケーラ一とミ ルスタインの方法 〔ネィチヤ一 (Nature)、 256、 495 (1975)] に従い実施するこ とができる。 融合促進剤としては、 例えば、 ポリエチレングリコール (PEG) やセンダイウィルスなどが挙げられるが、 好ましくは PEGが用いられる。  When preparing monoclonal antibody-producing cells, a warm-blooded animal immunized with an antigen, for example, an individual with an antibody titer is selected from a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization and contained in them. The hybridoma producing the monoclonal antibody can be prepared by fusing the animal-producing cells obtained with myeloma cells of the same or different species. The antibody titer in the antiserum can be measured, for example, by reacting the labeled protein described below with the antiserum, and then measuring the activity of the labeling agent bound to the antibody. The fusion operation can be performed according to a known method, for example, the method of Kohler and Milstein [Nature, 256, 495 (1975)]. Examples of the fusion promoter include polyethylene glycol (PEG) and Sendai virus, but PEG is preferably used.
骨髄腫細胞としては、 例えば、 NS— 1、 P 3U1、 SP 2/0、 AP— lな どの温血動物の骨髄腫細胞が挙げられるが、 P 3U1が好ましく用いられる。 用 いられる抗体産生細胞 (脾臓細胞) 数と骨髄腫細胞数との好ましい比率は 1 : 1 〜20 : 1程度であり、 PEG (好ましくは PEG 1000〜PEG6000) が 10〜80 %程度の濃度で添加され、 20〜40°C、 好ましくは 30〜37 で 1〜 10分間ィンキュベ一卜することにより効率よく細胞融合を実施できる。 モノクロ一ナル抗体産生ハイブリドーマのスクリーニングには種々の方法が使 用できるが、 例えば、 タンパク質抗原を直接あるいは担体とともに吸着させた固 相 (例、 マイクロプレート) にハイブリド一マ培養上清を添加し、 次に放射性物 質や酵素などで標識した抗免疫グロプリン抗体 (細胞融合に用いられる細胞がマ ウスの場合、 抗マウス免疫グロブリン抗体が用いられる) またはプロテイン Aを 加え、 固相に結合したモノクローナル抗体を検出する方法、 抗免疫グロブリン抗 体またはプロテイン Aを吸着させた固相にハイプリドーマ培養上清を添加し、 放 射性物質や酵素などで標識したタンパク質を加え、 固相に結合したモノクローナ ル抗体を検出する方法などが挙げられる。 Examples of myeloma cells include myeloma cells of warm-blooded animals such as NS-1, P3U1, SP 2/0, and AP-1, but P3U1 is preferably used. The preferred ratio between the number of antibody-producing cells (spleen cells) used and the number of myeloma cells used is about 1: 1 to 20: 1, and PEG (preferably PEG 1000 to PEG 6000) is used at a concentration of about 10 to 80%. The cell fusion can be carried out efficiently by adding the mixture and incubating at 20 to 40 ° C., preferably 30 to 37, for 1 to 10 minutes. Various methods can be used to screen for monoclonal antibody-producing hybridomas.For example, a hybridoma culture supernatant is added to a solid phase (eg, a microplate) on which a protein antigen is adsorbed directly or together with a carrier. Next, an anti-immunoglobulin antibody labeled with a radioactive substance or enzyme (anti-mouse immunoglobulin antibody is used if the cells used for cell fusion are mouse) or protein A, and a monoclonal antibody bound to the solid phase A monoclonal antibody bound to a solid phase is prepared by adding a hybridoma supernatant to a solid phase to which an anti-immunoglobulin antibody or protein A has been adsorbed, adding a protein labeled with a radioactive substance, an enzyme, or the like. And a method for detecting antibody.
モノクローナル抗体の選別は、 自体公知あるいはそれに準じる方法に従って行 なうことができる。 通常 HA T (ヒポキサンチン、 アミノプテリン、 チミジン) を添加した動物細胞用培地で行なうことができる。 選別および育種用培地として は、 ハイプリドーマが生育できるものならばどのような培地を用いても良い。 例 えば、 1〜2 0 %、 好ましくは 1 0〜2 0 %の牛胎児血清を含む R P M I 1 6 4 0培地、 1〜1 0 %の牛胎児血清を含む G I T培地 (和光純薬工業 (株) ) あ るいはハイプリドーマ培養用無血清培地 (S F M—1 0 1、 日水製薬 (株) ) な どを用いることができる。 培養温度は、 通常 2 0〜4 0 °C、 好ましくは約 3 7 である。 培養時間は、 通常 5日〜 3週間、 好ましくは 1週間〜 2週間である。 培 養は、 通常 5 %炭酸ガス下で行なうことができる。 ハイプリドーマ培養上清の抗 体価は、 上記の抗血清中の抗体価の測定と同様にして測定できる。  The selection of the monoclonal antibody can be performed according to a method known per se or a method analogous thereto. Usually, it can be performed in a medium for animal cells supplemented with HAT (hypoxanthine, aminopterin, thymidine). As a medium for selection and breeding, any medium can be used as long as the hybridoma can grow. For example, RPMI 164 medium containing 1 to 20%, preferably 10 to 20% fetal bovine serum, GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd. )) Or serum-free medium for hybridoma culture (SFM-101, Nissui Pharmaceutical Co., Ltd.). The culture temperature is usually from 20 to 40 ° C, preferably about 37. The culture time is generally 5 days to 3 weeks, preferably 1 week to 2 weeks. Cultivation can usually be performed under 5% CO2. The antibody titer of the hybridoma culture supernatant can be measured in the same manner as the measurement of the antibody titer in the antiserum described above.
( b ) モノクローナル抗体の精製  (b) Purification of monoclonal antibody
モノクローナル抗体の分離精製は、 自体公知の方法、 例えば、 免疫グロブリン の分離精製法 〔例、 塩析法、 アルコール沈殿法、 等電点沈殿法、 電気泳動法、 ィ オン交換体 (例、 D E A E) による吸脱着法、 超遠心法、 ゲルろ過法、 抗原結合 固相あるいはプロテイン Aあるいはプロテイン Gなどの活性吸着剤により抗体の みを採取し、 結合を解離させて抗体を得る特異的精製法〕 に従って行なうことが できる。  Monoclonal antibodies can be separated and purified by methods known per se, for example, immunoglobulin separation and purification methods (eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis method, ion exchanger (eg, DEAE) Absorption and desorption method, ultracentrifugation method, gel filtration method, antigen binding Solid phase or specific purification method of collecting antibody only with an active adsorbent such as protein A or protein G and dissociating the bond to obtain the antibody) Can do it.
〔ポリクローナル抗体の作製〕  (Preparation of polyclonal antibody)
本発明のポリクローナル抗体は、 それ自体公知あるいはそれに準じる方法に従 つて製造することができる。 例えば、 免疫抗原 (タンパク質抗原) 自体、 あるい はそれとキヤリア一タンパク質との複合体をつくり、 上記のモノクローナル抗体 の製造法と同様に温血動物に免疫を行ない、 該免疫動物から本発明のタンパク質 に対する抗体含有物を採取して、 抗体の分離精製を行なうことにより製造するこ とができる。  The polyclonal antibody of the present invention can be produced by a method known per se or a method analogous thereto. For example, an immunizing antigen (protein antigen) itself or a complex thereof with a carrier protein is formed, and immunization is performed on a warm-blooded animal in the same manner as in the above-described method for producing a monoclonal antibody. The antibody can be produced by collecting the antibody-containing substance of the antibody and separating and purifying the antibody.
温血動物を免疫するために用いられる免疫抗原とキャリアータンパク質との複 合体に関し、 キャリア一タンパク質の種類およびキャリアーとハプテンとの混合 比は、 キャリアーに架橋させて免疫したハプテンに対して抗体が効率良くできれ ば、 どの様なものをどの様な比率で架橋させてもよいが、 例えば、 ゥシ血清アル ブミンゃゥシサイログロプリン、 へモシァニン等を重量比でハプテン 1に対し、 約 0 . 1〜 2 0、 好ましくは約 1〜 5の割合でカプルさせる方法が用いられる。 また、 ハプテンとキャリアーの力プリングには、 種々の縮合剤を用いることが できるが、 ダルタルアルデヒドやカルポジイミド、 マレイミド活性エステル、 チ オール基、 ジチオビリジル基を含有する活性エステル試薬等が用いられる。 Regarding the complex of immunizing antigen and carrier protein used to immunize warm-blooded animals, the type of carrier-protein and the mixing ratio of carrier and hapten depend on the efficiency of antibody against hapten immunized by cross-linking with carrier. Well done For example, any substance may be cross-linked at any ratio.For example, serum serum albumin, psiloculopurine, hemocyanin, etc. may be used in a weight ratio of about 0.1 to 2 with respect to 1 hapten. A method of coupling at a rate of 0, preferably about 1 to 5 is used. Further, various condensing agents can be used for force coupling between the hapten and the carrier. For example, an active ester reagent containing a daltaraldehyde, a carbodiimide, a maleimide active ester, a thiol group or a dithioviridyl group is used.
縮合生成物は、 温血動物に対して、 抗体産生が可能な部位にそれ自体あるいは 担体、 希釈剤とともに投与される。 投与に際してお体産生能を高めるため、 完全 フロイントアジュバントゃ不完全フロイントアジュバントを投与してもよい。 投 与は、 通常約 2〜 6週毎に 1回ずつ、 計約 3〜1 0回程度行なわれる。  The condensation product is administered to a warm-blooded animal itself or together with a carrier or diluent at a site where antibody production is possible. Complete Freund's adjuvant ゃ Incomplete Freund's adjuvant may be administered to enhance body production during administration. The administration is usually made once every about 2 to 6 weeks, for a total of about 3 to 10 times.
ポリクローナル抗体は、 上記の方法で免疫された温血動物の血液、 腹水など、 好ましくは血液から採取することができる。  The polyclonal antibody can be collected from the blood, ascites, etc., preferably from the blood of a warm-blooded animal immunized by the above method.
抗血清中のポリクローナル抗体価の測定は、 上記の抗血清中の抗体価の測定と 同様にして測定できる。 ポリクロ一ナル抗体の分離精製は、 上記のモノクローナ ル抗体の分離精製と同様の免疫グロプリンの分離精製法に従って行なうことがで さる。  The measurement of the polyclonal antibody titer in the antiserum can be performed in the same manner as the measurement of the antibody titer in the antiserum described above. Separation and purification of the polyclonal antibody can be performed according to the same method for separation and purification of immunoglobulin as in the above-described separation and purification of the monoclonal antibody.
本発明で用いられるタンパク質または部分ペプチドをコードするポリヌクレオ チド (例、 D NA (以下、 アンチセンスポリヌクレオチドの説明においては、 こ れらの D NAを本発明の D NAと略記する場合がある) ) の塩基配列に相補的な、 または実質的に相補的な塩基配列またはその一部を含有するアンチセンスポリヌ クレオチドとしては、 本発明のポリヌクレオチド (例、 D NA) の塩基配列に相 補的な、 または実質的に相補的な塩基配列またはその一部を含有し、 該 D NAの 発現を抑制し得る作用を有するものであれば、 いずれのアンチセンスポリヌクレ ォチドであってもよいが、 アンチセンス D NAが好ましい。  Polynucleotide encoding the protein or partial peptide used in the present invention (eg, DNA (hereinafter, in the description of antisense polynucleotides, these DNAs may be abbreviated as the DNA of the present invention). The nucleotide sequence of the polynucleotide of the present invention (eg, DNA) is complementary to the nucleotide sequence of the polynucleotide of the present invention (eg, DNA). Any antisense polynucleotide may be used as long as it contains a basic or substantially complementary base sequence or a part thereof and has an action capable of suppressing the expression of the DNA. Antisense DNA is preferred.
本発明の D NAに実質的に相補的な塩基配列とは、 例えば、 本発明の D NAに 相補的な塩基配列 (すなわち、 本発明の D NAの相補鎖) の全塩基配列あるいは 部分塩基配列と約 7 0 %以上、 好ましくは約 8 0 %以上、 より好ましくは約 9 0 %以上、 最も好ましくは約 9 5 %以上の相同性を有する塩基配列などが挙げら れる。 特に、 本発明の D N Aの相補鎖の全塩基配列うち、 (ィ) 翻訳阻害を指向 したアンチセンスポリヌクレオチドの場合は、 本発明のタンパク質の N末端部位 をコードする部分の塩基配列 (例えば、 開始コドン付近の塩基配列など) の相補 鎖と約 7 0 %以上、 好ましくは約 8 0 %以上、 より好ましくは約 9 0 %以上、 最 も好ましくは約 9 5 %以上の相同性を有するアンチセンスポリヌクレオチドが、 (口) R N a s e Hによる R NA分解を指向するアンチセンスポリヌクレオチド の場合は、 イントロンを含む本発明の D NAの全塩基配列の相補鎖と約 7 0 %以 上、 好ましくは約 8 0 %以上、 より好ましくは約 9 0 %以上、 最も好ましくは約 9 5 %以上の相同性を有するアンチセンスポリヌクレオチドがそれぞれ好適であ る。 The nucleotide sequence substantially complementary to the DNA of the present invention is, for example, the entire nucleotide sequence or a partial nucleotide sequence of the nucleotide sequence complementary to the DNA of the present invention (that is, the complementary strand of the DNA of the present invention). And about 70% or more, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% or more. In particular, of the total nucleotide sequence of the complementary strand of the DNA of the present invention, In the case of the antisense polynucleotide thus obtained, it is at least about 70%, preferably about 80%, complementary to the complementary sequence of the nucleotide sequence (for example, the nucleotide sequence near the start codon) of the N-terminal site of the protein of the present invention. % Or more, more preferably about 90% or more, and most preferably about 95% or more of the antisense polynucleotide which is directed to RNA degradation by RNase H. In this case, it is at least about 70%, preferably about 80% or more, more preferably about 90% or more, and most preferably about 95% with the complementary strand of the entire nucleotide sequence of the DNA of the present invention including introns. Antisense polynucleotides having the above homology are each suitable.
具体的には、 配列番号: 2で表される塩基配列を含有する D NAの塩基配列に 相補的な、 もしくは実質的に相補的な塩基配列、 またはその一部分を含有するァ ンチセンスポリヌクレオチド、 好ましくは例えば、 配列番号: 2で表される塩基 配列を含有する D NAの塩基配列に相補的な塩基配列、 またはその一部分を含有 するアンチセンスポリヌクレオチド (より好ましくは、 配列番号: 2で表される 塩基配列を含有する D NAの塩基配列に相補的な塩基配列、 またはその一部分を 含有するアンチセンスポリヌクレオチド) が挙げられる。  Specifically, an antisense polynucleotide containing a nucleotide sequence complementary to or substantially complementary to the nucleotide sequence of DNA containing the nucleotide sequence represented by SEQ ID NO: 2, or a part thereof, Preferably, for example, an antisense polynucleotide containing a nucleotide sequence complementary to the nucleotide sequence of DNA containing the nucleotide sequence represented by SEQ ID NO: 2, or a part thereof (more preferably, a nucleotide sequence represented by SEQ ID NO: 2 A base sequence complementary to the base sequence of DNA containing the base sequence to be prepared, or an antisense polynucleotide containing a part thereof.
アンチセンスポリヌクレオチドは通常、 1 0〜4 0個程度、 好ましくは 1 5〜 3 0個程度の塩基から構成される。  An antisense polynucleotide is usually composed of about 10 to 40 bases, preferably about 15 to 30 bases.
ヌクレアーゼなどの加水分解酵素による分解を防ぐために、 アンチセンス D N Aを構成する各ヌクレオチドのリン酸残基 (ホスフェート) は、 例えば、 ホスホ 口チォェ一ト、 メチルホスホネート、 ホスホロジチォネートなどの化学修飾リン 酸残基に置換されていてもよい。 また、 各ヌクレオチドの糖 (デォキシリポ一 ス) は、 2, 一〇一メチル化などの化学修飾糖構造に置換されていてもよいし、 塩基部分 (ピリミジン、 プリン) も化学修飾を受けたものであってもよく、 配列 番号: 2で表される塩基配列を含有する D NAにハイブリダィズするものであれ ばいずれのものでもよい。 これらのアンチセンスポリヌクレオチドは、 公知の D N A合成装置などを用いて製造することができる。  In order to prevent degradation by nucleases and other hydrolases, the phosphate residues (phosphates) of each nucleotide constituting the antisense DNA are, for example, chemically modified phosphorus such as phospho-thiolate, methylphosphonate, and phosphorodithionate. It may be substituted by an acid residue. In addition, the sugar (deoxyliposome) of each nucleotide may be substituted with a chemically modified sugar structure such as 2,1-methylation, and the base portion (pyrimidine, purine) may also be chemically modified. Any one may be used as long as it hybridizes to DNA containing the nucleotide sequence represented by SEQ ID NO: 2. These antisense polynucleotides can be produced using a known DNA synthesizer or the like.
本発明に従えば、 本発明のタンパク質遺伝子の複製または発現を阻害すること のできるアンチセンスポリヌクレオチド (核酸) を、 クローン化した、 あるいは T JP2003/007926 According to the present invention, an antisense polynucleotide (nucleic acid) capable of inhibiting the replication or expression of the protein gene of the present invention has been cloned or T JP2003 / 007926
29 決定されたタンパク質をコードする D N Aの塩基配列情報に基づき設計し、 合成 しうる。 かかるポリヌクレオチド (核酸) は、 本発明のタンパク質遺伝子の R N Aとハイブリダィズすることができ、 該 R N Aの合成または機能を阻害すること ができるか、 あるいは本発明のタンパク質関連 R N Aとの相互作用を介して本発 明のタンパク質遺伝子の発現を調節 ·制御することができる。 本発明のタンパク 質関連 RN Aの選択された配列に相補的なポリヌクレオチド、 および本発明の夕 ンパク質関連 R N Aと特異的にハイプリダイズすることができるポリヌクレオチ ドは、 生体内および生体外で本発明のタンパク質遺伝子の発現を調節 ·制御する のに有用であり、 また病気などの治療または診断に有用である。 用語 「対応す る」 とは、 遺伝子を含めたヌクレオチド、 塩基配列または核酸の特定の配列に相 同性を有するあるいは相補的であることを意味する。 ヌクレオチド、 塩基配列ま たは核酸とペプチド (タンパク質) との間で 「対応する」 とは、 ヌクレオチド 29 It can be designed and synthesized based on the nucleotide sequence information of the DNA encoding the determined protein. Such a polynucleotide (nucleic acid) can hybridize with the RNA of the protein gene of the present invention, inhibit the synthesis or function of the RNA, or interact with the protein-related RNA of the present invention It can regulate and control the expression of the protein gene of the present invention. Polynucleotides complementary to the selected sequence of the protein-related RNA of the present invention, and polynucleotides capable of specifically hybridizing with the protein-related RNA of the present invention, can be used in vivo and in vitro. It is useful for regulating and controlling the expression of the protein gene of the present invention, and also useful for treating or diagnosing diseases and the like. The term "corresponding" means having homology or being complementary to a nucleotide, base sequence or a specific sequence of a nucleic acid including a gene. The “correspondence” between a nucleotide, base sequence or nucleic acid and a peptide (protein) is a nucleotide
(核酸) の配列またはその相補体から誘導される指令にあるペプチド (タンパク 質) のアミノ酸を通常指している。 タンパク質遺伝子の 5 ' 端ヘアピンループ、 5 ' 端 6—ベースペア ·リピート、 5 ' 端非翻訳領域、 ポリペプチド翻訳開始コ ドン、 タンパク質コード領域、 O R F翻訳終止コドン、 3 ' 端非翻訳領域、 3 ' 端パリンドローム領域、 および 3 ' 端ヘアピンループは好ましい対象領域として 選択しうるが、 タンパク質遺伝子内の如何なる領域も対象として選択しうる。 目的核酸と、 対象領域の少なくとも一部に相補的なポリヌクレオチドとの関係 は、 対象物とハイブリダィズすることができるポリヌクレオチドとの関係は、It usually refers to the amino acids of a peptide (protein) in a directive derived from the (nucleic acid) sequence or its complement. 5 'end hairpin loop of protein gene, 5' end 6—base pair repeat, 5 'end untranslated region, polypeptide translation start codon, protein coding region, ORF translation stop codon, 3' end untranslated region, 3 The palindrome region at the 'end and the hairpin loop at the 3' end can be selected as preferred regions of interest, but any region within the protein gene can be selected as the region of interest. The relationship between the target nucleic acid and the polynucleotide complementary to at least a part of the target region is as follows: The relationship between the target nucleic acid and the polynucleotide capable of hybridizing with the target is
「アンチセンス」 であるということができる。 7ンチセンスポリヌクレオチドは、 2ーデォキシー D—リボースを含有しているポリヌクレオチド、 D—リポースを 含有しているポリヌクレオチド、 プリンまたはピリミジン塩基の N—グリコシド であるその他のタイプのポリヌクレオチド、 あるいは非ヌクレオチド骨格を有す るその他のポリマ一 (例えば、 市販のタンパク質核酸および合成配列特異的な核 酸ポリマー) または特殊な結合を含有するその他のポリマー (但し、 該ポリマー は D N Aや R N A中に見出されるような塩基のペアリングゃ塩基の付着を許容す る配置をもつヌクレオチドを含有する) などが挙げられる。 それらは、 2本鎖 D NA、 1本鎖 D NA、 2本鎖 R NA、 1本鎖 R NA、 さらに D NA : R NAハイ ブリツドであることができ、 さらに非修飾ポリヌクレオチド (または非修飾オリ ゴヌクレオチド) 、 さらには公知の修飾の付加されたもの、 例えば当該分野で知 られた標識のあるもの、 キャップの付いたもの、 メチル化されたもの、 1個以上 の天然のヌクレオチドを類縁物で置換したもの、 分子内ヌクレオチド修飾のされ たもの、 例えば非荷電結合 (例えば、 メチルホスホネート、 ホスホトリエステル、 ホスホルアミデート、 力ルバメートなど) を持つもの、 電荷を有する結合または 硫黄含有結合 (例えば、 ホスホロチォエート、 ホスホロジチォェ一トなど) を持 つもの、 例えばタンパク質 (ヌクレアーゼ、 ヌクレア一ゼ ·インヒビター、 トキ シン、 抗体、 シグナルペプチド、 ポリ一 L一リジンなど) や糖 (例えば、 モノサ ッカライドなど) などの側鎖基を有しているもの、 インターカレント化合物 (例 えば、 ァクリジン、 ソラレンなど) を持つもの、 キレート化合物 (例えば、 金属、 放射活性をもつ金属、 ホウ素、 酸化性の金属など) を含有するもの、 アルキルィ匕 剤を含有するもの、 修飾された結合を持つもの (例えば、 αァノマ一型の核酸な ど) であってもよい。 ここで 「ヌクレオシド」 、 「ヌクレオチド」 および 「核 酸」 とは、 プリンおよびピリミジン塩基を含有するのみでなく、 修飾されたその 他の複素環型塩基をもつようなものを含んでいて良い。 こうした修飾物は、 メチ ル化されたプリンぉよびピリミジン、 ァシル化されたプリンぉよびピリミジン、 あるいはその他の複素環を含むものであってよい。 修飾されたヌクレオチドおよ び修飾されたヌクレオチドはまた糖部分が修飾されていてよく、 例えば、 1個以 上の水酸基がハロゲンとか、 脂肪族基などで置換されていたり、 あるいはェ一テ ル、 ァミンなどの官能基に変換されていてよい。 It can be said to be "antisense". Antisense polynucleotides are polynucleotides containing 2-dexoxy D-ribose, polynucleotides containing D-reports, other types of polynucleotides that are N-glycosides of purine or pyrimidine bases, or non-polynucleotides. Other polymers having a nucleotide backbone (eg, commercially available protein nucleic acids and synthetic sequence-specific nucleic acid polymers) or other polymers containing special bonds (provided that such polymers are found in DNA and RNA) Base pairing (including nucleotides having a configuration that allows base attachment)). They are double-stranded DNA, single-stranded DNA, double-stranded RNA, single-stranded RNA, and even DNA: RNA high. Bridges, and may also be unmodified polynucleotides (or unmodified oligonucleotides), or even those with known modifications, such as those with a label known in the art, capped, Methylated, one or more natural nucleotides replaced by analogs, modified intramolecular nucleotides, e.g., uncharged bonds (e.g., methylphosphonates, phosphotriesters, phosphoramidates, Those with a charged bond or a sulfur-containing bond (eg, phosphorothioate, phosphorodithioate, etc.), eg, proteins (nucleases, nuclease inhibitors, toxins, antibodies, signals) Peptides, poly-L-lysine, etc.) and sugars (eg, Compounds with side groups such as monosaccharides, etc., compounds with interactive compounds (eg, acridine, psoralen, etc.), chelating compounds (eg, metals, radioactive metals, boron, oxidizable compounds) Metal, etc.), one containing an alkylating agent, and one having a modified bond (for example, α-anomer type 1 nucleic acid). Here, the “nucleoside”, “nucleotide” and “nucleic acid” may include not only those containing purine and pyrimidine bases but also those having other modified heterocyclic bases. Such modifications may include methylated purines and pyrimidines, acylated purines and pyrimidines, or other heterocycles. Modified nucleotides and modified nucleotides may also be modified at the sugar moiety, e.g., where one or more hydroxyl groups have been replaced with halogens, aliphatic groups, etc., or ethers, It may be converted to a functional group such as amine.
本発明のアンチセンスポリヌクレオチド (核酸) は、 R NA、 D NA、 あるい は修飾された核酸 (R NA、 D NA) である。 修飾された核酸の具体例としては 核酸の硫黄誘導体ゃチォホスフェート誘導体、 そしてポリヌクレオシドアミドゃ オリゴヌクレオシドアミドの分解に抵抗性のものが挙げられるが、 それに限定さ れるものではない。 本発明のアンチセンス核酸は次のような方針で好ましく設計 されうる。 すなわち、 細胞内でのアンチセンス核酸をより安定なものにする、 ァ ンチセンス核酸の細胞透過性をより高める、 目標とするセンス鎖に対する親和性 をより大きなものにする、 そしてもし毒性があるならアンチセンス核酸の毒性を より小さなものにする。 The antisense polynucleotide (nucleic acid) of the present invention is an RNA, a DNA, or a modified nucleic acid (RNA, DNA). Specific examples of the modified nucleic acid include, but are not limited to, sulfur derivatives of nucleic acids, thiophosphate derivatives, and polynucleoside amides, which are resistant to degradation of oligonucleoside amides. The antisense nucleic acid of the present invention can be preferably designed according to the following policy. That is, to make the antisense nucleic acid more stable in the cell, to increase the cell permeability of the antisense nucleic acid, to have a greater affinity for the target sense strand, and to be more toxic if it is toxic. Sense nucleic acid toxicity Make it smaller.
こうして修飾は当該分野で数多く知られており、 例えば J. Kawakami et al ., Pharm Tech Japan, Vo l . 8, pp. 247, 1992 ; Vol . 8, pp. 395, 1992 ; S. T.  Thus, many modifications are known in the art, for example, J. Kawakami et al., Pharm Tech Japan, Vol. 8, pp. 247, 1992; Vol. 8, pp. 395, 1992; S. T.
Crooke et al . ed., Ant i sense Research and Appl i cat i ons, CRC Press, 1993 などに開示がある。 Crooke et al. Ed., Ant isense Research and Appli cations, CRC Press, 1993.
本発明のアンチセンス核酸は、 変化せしめられたり、 修飾された糖、 塩基、 結 合を含有していて良く、 リボゾーム、 ミクロスフエアのような特殊な形態で供与 されたり、 遺伝子治療により適用されたり、 付加された形態で与えられることが できうる。 こうして付加形態で用いられるものとしては、 リン酸基骨格の電荷を 中和するように働くポリリジンのようなポリカチォン体、 細胞膜との相互作用を 高めたり、 核酸の取込みを増大せしめるような脂質 (例えば、 ホスホリピド、 コ レステロールなど) といった疎水性のものが挙げられる。 付加するに好ましい脂 質としては、 コレステロールやその誘導体 (例えば、 コレステリルクロ口ホルメ ート、 コール酸など) が挙げられる。 こうしたものは、 核酸の 3 ' 端あるいは 5 ' 端に付着させることができ、 塩基、 糖、 分子内ヌクレオシド結合を介して付 着させることができうる。 その他の基としては、 核酸の 3 ' 端あるいは 5 ' 端に 特異的に配置されたキャップ用の基で、 ェキソヌクレアーゼ、 R N a s eなどの ヌクレアーゼによる分解を阻止するためのものが挙げられる。 こうしたキャップ 用の基としては、 ポリエチレングリコール、 テトラエチレングリコールなどのグ リコールをはじめとした当該分野で知られた水酸基の保護基が挙げられるが、 そ れに限定されるものではない。  The antisense nucleic acids of the present invention may contain altered or modified sugars, bases, or bonds, may be provided in special forms such as ribosomes or microspheres, may be applied by gene therapy, It could be given in additional form. Such additional forms include polycations, such as polylysine, which act to neutralize the charge on the phosphate backbone, and lipids, which enhance interaction with cell membranes and increase nucleic acid uptake (eg, , Phospholipids, cholesterol, etc.). Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chromate formate, cholic acid, etc.). Such a substance can be attached to the 3 'end or 5' end of a nucleic acid, and can be attached via a base, a sugar, or an intramolecular nucleoside bond. Other groups include cap groups specifically arranged at the 3 'end or 5' end of nucleic acids, which prevent degradation by nucleases such as exonucleases and RNAses. Such capping groups include, but are not limited to, hydroxyl-protecting groups known in the art, including glycols such as polyethylene glycol and tetraethylene glycol.
アンチセンス核酸の阻害活性は、 本発明の形質転換体、 本発明の生体内や生体 外の遺伝子発現系、 あるいは本発明のタンパク質の生体内や生体外の翻訳系を用 いて調べることができる。 該核酸それ自体公知の各種の方法で細胞に適用できる。 以下に、 本発明のタンパク質もしくは部分ペプチドまたはその塩 (以下、 本発 明のタンパク質と略記する場合がある) 、 本発明のタンパク質または部分べプチ ドをコードする D NA (以下、 本発明の D NAと略記する場合がある) 、 本発明 のタンパク質もしくは部分ペプチドまたはその塩に対する抗体 (以下、 本発明の 抗体と略記する場合がある) 、 および本発明の D N Aのアンチセンスポリヌクレ ォチド (以下、 本発明のアンチセンスポリヌクレオチドと略記する場合がある) の用途を説明する。 The inhibitory activity of the antisense nucleic acid can be examined using the transformant of the present invention, the in vivo or in vitro gene expression system of the present invention, or the in vivo or in vitro translation system of the protein of the present invention. The nucleic acid can be applied to cells by various methods known per se. Hereinafter, the protein or partial peptide of the present invention or a salt thereof (hereinafter sometimes abbreviated as the protein of the present invention), the DNA encoding the protein or partial peptide of the present invention (hereinafter referred to as the D NA), an antibody against the protein or partial peptide of the present invention or a salt thereof (hereinafter referred to as “the present invention”). The use of an antibody (sometimes abbreviated as an antibody) and the antisense polynucleotide of the DNA of the present invention (hereinafter sometimes abbreviated as the antisense polynucleotide of the present invention) will be described.
本発明のタンパク質は、 がん組織で発現が増加するので、 疾患マ一カーとして 利用することが出来る。 すなわち、 がん組織における早期診断、 症状の重症度の 判定、 疾患進行の予測のためのマーカーとして有用である。 よって、 本発明の夕 ンパク質をコードする遺伝子のアンチセンスポリヌクレオチド、 本発明のタンパ ク質の活性を阻害する化合物もしくはその塩または本発明のタンパク質に対する 抗体を含有する医薬は、 例えば、 大腸がん、 乳がん、 肺がん、 前立腺がん、 食道 がん、 胃がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣がん、 甲状腺がん、 勝臓がん、 脳腫瘍または血液腫瘍などのがん の予防 ·治療剤として使用することができる。  Since the expression of the protein of the present invention increases in cancer tissues, it can be used as a disease marker. In other words, it is useful as a marker for early diagnosis in cancer tissues, judgment of the severity of symptoms, and prediction of disease progression. Therefore, a medicament containing an antisense polynucleotide of the gene encoding the protein of the present invention, a compound that inhibits the activity of the protein of the present invention or a salt thereof, or an antibody against the protein of the present invention may be, for example, a large intestine. Cancer, breast cancer, lung cancer, prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid gland It can be used as a prophylactic / therapeutic agent for cancer such as cancer, brain tumor or blood tumor.
( 1 ) 疾病に対する医薬候補化合物のスクリーニング  (1) Screening of drug candidate compounds for diseases
本発明のタンパク質はがん組織で発現が増加しており、 さらに、 本発明のタン パク質の活性を阻害するとがん細胞がアポトーシスを起こす。 従って、 本発朗の タンパク質の活性を調節 (好ましぐは阻害) する化合物またはその塩は、 例えば 大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝臓がん、 胆道が ん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣がん、 甲状腺がん、 塍臓がん、 脳腫瘍または血液腫瘍などのがんの予防 ·治療剤として使用すること ができる。 好ましくは、 ホルモン (例、 エストロゲンなど) 非依存性、 HER2発現 陰性などのがん (例、 乳がんなど) の予防 ·治療剤である。  The expression of the protein of the present invention is increased in cancer tissues, and when the activity of the protein of the present invention is inhibited, cancer cells undergo apoptosis. Therefore, compounds or salts thereof that modulate (preferably inhibit) the activity of the protein of the present invention include, for example, colorectal cancer, breast cancer, lung cancer, prostate cancer, esophageal cancer, stomach cancer, liver cancer, and biliary tract. Used as a prophylactic and therapeutic agent for cancer such as cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testis cancer, thyroid cancer, kidney cancer, brain tumor, or blood tumor can do. Preferably, it is a prophylactic / therapeutic agent for cancer (eg, breast cancer, etc.) that is hormone-independent (eg, estrogen, etc.)-Independent, HER2-negative, etc.
また、 本発明のタンパク質の活性を阻害する化合物またはその塩は、 例えば、 アポトーシス作用調節剤 (好ましくはアポトーシス作用促進剤) として使用する こともできる。  Further, the compound or a salt thereof that inhibits the activity of the protein of the present invention can also be used, for example, as an apoptosis action regulator (preferably an apoptosis action promoter).
したがって、 本発明のタンパク質は、 本発明のタンパク質の活性を調節する化 合物またはその塩のスクリーニングのための試薬として有用である。  Therefore, the protein of the present invention is useful as a reagent for screening a compound or its salt that regulates the activity of the protein of the present invention.
すなわち、 本発明は、 本発明のタンパク質を用いることを特徴とする本発明の タンパク質の活性 (例えば、 リガンド結合活性、 シグナル伝達活性など) を調節 する化合物またはその塩のスクリーニング方法を提供する。 より具体的には、 例えば、 (i) 本発明のタンパク質を産生する能力を有する 細胞のリガンド結合活性またはシグナル伝達活性と、 (i i) 本発明のタンパク質 を産生する能力を有する細胞と試験化合物の混合物のリガンド結合活性またはシ グナル伝達活性の比較することを特徴する本発明のタンパク質の活性を調節 (促 進または阻害) する化合物またはその塩のスクリーニング方法が用いられる。. 上記スクリーニング方法においては、 例えば (i) と (U) の場合において、 リガンド結合活性またはシグナル伝達活性を自体公知の方法、 例えば J. Immunol . 166 (1)巻、 15- 19頁、 2001年に記載の方法またはそれに準じる方法に従って測定 し、 比較する。 That is, the present invention provides a method for screening a compound or a salt thereof that regulates the activity (eg, ligand binding activity, signal transduction activity, etc.) of the protein of the present invention, characterized by using the protein of the present invention. More specifically, for example, (i) the ligand binding activity or signal transduction activity of a cell capable of producing the protein of the present invention; A method for screening a compound or a salt thereof that regulates (promotes or inhibits) the activity of the protein of the present invention, which is characterized by comparing the ligand binding activity or the signal transmission activity of the mixture, is used. In the above screening method, for example, in the case of (i) and (U), the ligand binding activity or signal transduction activity can be determined by a method known per se, for example, J. Immunol. 166 (1), pp. 15-19, 2001 Measure and compare according to the method described in or above.
具体的には、 リガンド結合促進または細胞内シグナル伝達促進作用を有する化 合物をスクリーニングする場合は、 (i) 本発明のタンパク質発現ベクターを、 N F κ Bの結合配列をプロモーターまたはェンハンサ一中に含有するレポ一夕一 遺伝子とともに細胞に導入して培養した場合と、 (i i) 本発明のタンパク質発現 ベクタ一を、 N F κ Bの結合配列をプロモー夕一またはェンハンサ一中に含有す るレポーター遺伝子とともに細胞に導入し、 試験化合物の存在下、 培養した場合 の、 レポ一ター発現量を比較する。 レポ一夕一発現量は、 ルシフェラ一ゼ活性、 アルカリフォスファターゼ活性などを用いたレポータータンパク活性を指標とす ることにより測定できる。  Specifically, when screening for a compound having a ligand binding promoting action or an intracellular signaling promoting action, (i) the protein expression vector of the present invention is prepared by adding an NFκB binding sequence to a promoter or enhancer. And (ii) a reporter gene containing the protein expression vector of the present invention in the promoter or enhancer of the NFκB binding sequence. When the cells are introduced into cells and cultured in the presence of the test compound, the amount of reporter expression is compared. The repo overnight expression level can be measured by using a reporter protein activity using luciferase activity, alkaline phosphatase activity or the like as an index.
また、 リガンド結合阻害または細胞内シグナル伝達阻害作用を有する化合物を スクリーニングする場合は、 具体的には、 U) 本発明のタンパク質発現べクタ 一を、 N F κ Bの結合配列をプロモータ一またはェンハンサ一中に含有するレポ 一ター遺伝子とともに細胞に導入し、 必要に応じ、 (a) 微生物細胞破碎液、 微 生物培養上清、 真核細胞破碎液、 真核細胞培養上清などリガンドが含まれる液、 (b) リガンド自身または (c) 天然のリガンドと同等に結合活性を有する物質を 添加して培養した場合と、 (i i) 本発明のタンパク質発現べクタ一を、 N F K B の結合配列をプロモ一ターまたはェンハンサ一中に含有するレポ一タ一遺伝子と ともに細胞に導入し、 必要に応じ、 (a) 微生物細胞破砕液、 微生物培養上清、 真核細胞破碎液、 真核細胞培養上清などリガンドが含まれる液、 (b) リガンド 自身または (c) 天然のリガンドと同等に結合活性を有する物質を添加して、 試 験化合物の存在下、 培養した場合の、 レポーター発現量を測定し、 比較する。 レポ一夕一発現量は、 ルシフェラ一ゼ活性、 アルカリフォスファタ一ゼ活性な どを用いたレポ一タータンパク活性を指標とすることにより測定できる。 When screening for a compound having an inhibitory effect on ligand binding or intracellular signal transduction, specifically, U) the protein expression vector of the present invention may be used to bind the NFκB binding sequence to a promoter or enhancer. Transfection into cells with the reporter gene contained therein. If necessary, (a) a solution containing ligands such as microbial cell disruption solution, microbial culture supernatant, eukaryotic cell disruption solution, and eukaryotic cell culture supernatant (B) culturing with the addition of a substance having the same binding activity as the ligand itself or (c) a natural ligand; and (ii) using the protein expression vector of the present invention to promote the binding sequence of NFKB. Transfected cells or microbial cell culture supernatant, eukaryotic cell lysate, eukaryotic cells Youe liquid containing the ligand, such as supernatants, by adding a substance having a (b) a ligand itself or (c) a natural ligand comparable binding activity, trial Measure and compare reporter expression levels when cultured in the presence of the test compound. The expression level of the repo overnight can be measured by using the repoprotein activity using luciferase activity, alkaline phosphatase activity and the like as an index.
本発明のタンパク質を産生する能力を有する細胞としては、 例えば、 前述した 本発明のタンパク質をコードする D NAを含有するベクターで形質転換された宿 主 (形質転換体) が用いられる。 宿主としては、 例えば、 C O S 7細胞、 CH O 細胞、 H E K 2 9 3細胞などの動物細胞が好ましく用いられる。 該スクリ一ニン グには、 例えば、 前述の方法で培養することによって、 本発明のタンパク質を細 胞膜上に発現させた形質転換体が好ましく用いられる。 本発明のタンパク質を発 現し得る細胞の培養方法は、 前記した本発明の形質変換体の培養法と同様である。 試験化合物としては、 例えばペプチド、 タンパク質、 非ペプチド性化合物、 合 成化合物、 発酵生産物、 細胞抽出液、 植物抽出液、 動物組織抽出液などがあげら れる。  As a cell having the ability to produce the protein of the present invention, for example, a host (transformant) transformed with a vector containing a DNA encoding the protein of the present invention described above is used. As a host, for example, animal cells such as COS 7 cells, CHO cells, and HEK293 cells are preferably used. For the screening, for example, a transformant in which the protein of the present invention is expressed on a cell membrane by culturing by the method described above is preferably used. The method for culturing cells capable of expressing the protein of the present invention is the same as the above-described method for culturing the transformant of the present invention. Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and the like.
例えば、 上記 (i i) の場合におけるリガンド結合活性またはシグナル伝達活性 が上記 (i) の場合に比べて、 約 2 0 %以上、 好ましくは 3 0 %以上、 より好ま しくは約 5 0 %以上上昇させる試験化合物を、 本発明のタンパク質の活性を促進 する化合物として、 上記 (i i) の場合におけるリガンド結合活性またはシグナル 伝達活性が上記 (i) の場合に比べて、 約 2 0 %以上、 好ましくは 3 0 %以上、 より好ましくは約 5 0 %以上減少させる試験化合物を本発明のタンパク質の活性 を阻害する化合物として選択することができる。  For example, the ligand binding activity or signal transduction activity in the case of (ii) is increased by about 20% or more, preferably 30% or more, more preferably about 50% or more, as compared with the case of (i). The test compound to be used is a compound that promotes the activity of the protein of the present invention, wherein the ligand binding activity or signal transduction activity in the case (ii) above is about 20% or more, preferably A test compound that reduces the activity of the protein of the present invention by 30% or more, more preferably about 50% or more, can be selected as the compound that inhibits the activity.
本発明のタンパク質の活性を促進する活性を有する化合物は、 本発明のタンパ ク質の作用を増強するための安全で低毒性な医薬として有用である。  The compound having the activity of promoting the activity of the protein of the present invention is useful as a safe and low-toxic drug for enhancing the action of the protein of the present invention.
本発明のタンパク質の活性を阻害する活性を有する化合物は、 本発明のタンパ ク質の生理活性を抑制するための安全で低毒性な医薬、 例えばがんの予防 治療 剤、 アポトーシス作用促進剤などとして有用である。  The compound having an activity of inhibiting the activity of the protein of the present invention is used as a safe and low-toxicity drug for suppressing the physiological activity of the protein of the present invention, for example, a prophylactic / therapeutic agent for cancer, an apoptosis promoting agent and the like. Useful.
本発明のスクリーニング方法またはスクリーニング用キットを用いて得られる 化合物またはその塩は、 例えば、 ペプチド、 タンパク質、 非ペプチド性化合物、 合成化合物、 発酵生産物、 細胞抽出液、 植物抽出液、 動物組織抽出液、 血漿など から選ばれた化合物である。 該化合物の塩としては、 前記した本発明のペプチド の塩と同様のものが用いられる。 Compounds or salts thereof obtained using the screening method or screening kit of the present invention include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts And compounds selected from plasma and the like. The salt of the compound includes the peptide of the present invention described above. The same ones as used in the above are used.
さらに、 本発明のタンパク質をコードする遺伝子も、 がん組織において発現が 増加するので、 本発明のタンパク質をコードする遺伝子の発現を調節 (好ましく は阻害) する化合物またはその塩も、 例えば乳がん、 大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱 がん、 子宮がん、 卵巣がん、 精巣がん、 甲状腺がん、 塍臓がん、 脳腫瘍または血 液腫瘍などのがんの予防 ·治療剤として使用することができる。 好ましくは、 ホ ルモン (例、 エストロゲンなど) 非依存性、 HER2発現陰性などのがん (例、 乳が んなど) の予防 ·治療剤である。 また、 本発明のタンパク質をコードする遺伝子 の発現を阻害する化合物またはその塩は、 例えば、 アポトーシス作用調節剤 (好 ましくはアポトーシス作用促進剤) として使用することもできる。  Furthermore, since the expression of the gene encoding the protein of the present invention also increases in cancer tissues, compounds or salts thereof that regulate (preferably inhibit) the expression of the gene encoding the protein of the present invention include, for example, breast cancer and colon Cancer, breast, lung, prostate, esophagus, stomach, liver, biliary, spleen, kidney, bladder, uterus, ovary, testis, thyroid It can be used as a prophylactic and therapeutic agent for cancer such as cancer, kidney cancer, brain tumor and blood tumor. Preferably, it is a prophylactic / therapeutic agent for cancers (eg, breast cancer, etc.) that are hormone-independent (eg, estrogen, etc.)-Independent, HER2-negative, etc. Further, the compound that inhibits the expression of the gene encoding the protein of the present invention or a salt thereof can be used, for example, as an apoptosis-controlling agent (preferably, an apoptosis-promoting agent).
したがって、 本発明の D N Aは、 本発明のタンパク質をコードする遺伝子の発 現を調節する化合物またはその塩のスクリーニングのための試薬として有用であ る。  Therefore, the DNA of the present invention is useful as a reagent for screening a compound or a salt thereof that regulates the expression of a gene encoding the protein of the present invention.
スクリーニング方法としては、 (i i i) 本発明のタンパク質を産生する能力を 有する細胞を培養した場合と、 (iv) 試験化合物の存在下、 本発明で用いられる タンパク質を産生する能力を有する細胞を培養した場合との比較を行うことを特 徵とするスクリーニング方法が挙げられる。  The screening method includes (iii) culturing cells capable of producing the protein of the present invention, and (iv) culturing cells capable of producing the protein used in the present invention in the presence of the test compound. There is a screening method which is characterized by comparing with the case.
上記方法において、 (i i i) と (iv) の場合における、 前記遺伝子の発現量 (具体的には、 本発明のタンパク質量または前記タンパク質をコードする mR N A量) を測定して、 比較する。  In the above method, the expression level of the gene (specifically, the amount of the protein of the present invention or the amount of mRNA encoding the protein) in the cases (ii) and (iv) is measured and compared.
試験化合物および本発明のタンパク質を産生する能力を有する細胞としては、 上記と同様のものが挙げられる。  Examples of the test compound and cells having the ability to produce the protein of the present invention include the same cells as described above.
タンパク質量の測定は、 公知の方法、 例えば、 本発明のタンパク質を認識する 抗体を用いて、 細胞抽出液中などに存在する前記タンパク質を、 ウエスタン解析、 E L I S A法などの方法またはそれに準じる方法に従い測定することができる。 mR NA量の測定は、 公知の方法、 例えば、 プローブとして配列番号: 2また はその一部を含有する核酸を用いるノーザンハイブリダィゼーション、 あるいは  The amount of the protein is measured by a known method, for example, using an antibody that recognizes the protein of the present invention, and measuring the protein present in a cell extract or the like according to a method such as Western analysis, ELISA, or a method analogous thereto. can do. The mRNA amount can be measured by a known method, for example, Northern hybridization using a nucleic acid containing SEQ ID NO: 2 or a part thereof as a probe, or
'一として配列番号: 2またはその一部を含有する核酸を用いる P C R法 またはそれに準じる方法に従い測定することができる。 'PCR method using nucleic acid containing SEQ ID NO: 2 or a part thereof as one Or it can measure according to the method according to it.
例えば、 上記 (iv) の場合における遺伝子発現を、 上記 (i i i) の場合に比べ て、 約 2 0 %以上、 好ましくは 3 0 %以上、 より好ましくは約 5 0 %以上促進さ せる試験化合物を、 発明のタンパク質をコードする遺伝子の発現を促進する化 合物として、 上記 (iv) の場合における遺伝子発現を、 上記 (i i i) の場合に比 ベて、 約 2 0 %以上、 好ましくは 3 0 %以上、 より好ましくは約 5 0 %以上阻害 する試験ィ匕合物を、 本発明のタンパク質をコードする遺伝子の発現を阻害する化 合物として選択することができる。  For example, a test compound that promotes gene expression in the case of the above (iv) by about 20% or more, preferably 30% or more, more preferably about 50% or more as compared with the case of the above (iii) As a compound that promotes the expression of a gene encoding the protein of the present invention, the gene expression in the case of the above (iv) is about 20% or more, preferably 30% or more, as compared with the case of the above (iii). Test conjugates that inhibit at least about 50%, more preferably at least about 50%, can be selected as compounds that inhibit the expression of the gene encoding the protein of the present invention.
本発明のスクリーニング用キットは、 本発明で用いられるタンパク質もしくは 部分べプチドまたはその塩、 または本発明で用いられるタンパク質もしくは部分 ペプチドを産生する能力を有する細胞を含有するものである。  The screening kit of the present invention contains cells capable of producing the protein or partial peptide used in the present invention or a salt thereof, or the protein or partial peptide used in the present invention.
本発明のスクリーニング方法またはスクリーニング用キットを用いて得られる 化合物またはその塩は、 上記した試験化合物、 例えば、 ペプチド、 タンパク質、 非ペプチド性化合物、 合成化合物、 発酵生産物、 細胞抽出液、 植物抽出液、 動物 組織抽出液、 血漿などから選ばれた化合物またはその塩であり、 本発明のタンパ ク質の活性 (例、 リガンド結合活性またはシグナル伝達活性など) を調節する化 合物またはその塩である。  The compound or a salt thereof obtained by using the screening method or the screening kit of the present invention is a test compound as described above, for example, a peptide, a protein, a non-peptidic compound, a synthetic compound, a fermentation product, a cell extract, or a plant extract. A compound selected from animal tissue extract, plasma, or the like, or a salt thereof, which is a compound or a salt thereof that regulates the activity of the protein of the present invention (eg, ligand binding activity or signal transduction activity). .
該化合物の塩としては、 前記した本発明のタンパク質の塩と同様のものが用い られる。  As the salt of the compound, those similar to the aforementioned salts of the protein of the present invention are used.
本発明のタンパク質の活性を調節 (好ましくは阻害) する化合物またはその塩、 および本発明のタンパク質をコードする遺伝子の発現を調節 (好ましくは阻害) する化合物またはその塩はそれぞれ、 例えば大腸がん、 乳がん、 肺がん、 前立腺 がん、 食道がん、 胃がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣がん、 甲状腺がん、 滕臓がん、 脳腫瘍または血液腫瘍 などのがんの予防 ·治療剤として、 またアポトーシス作用促進剤として有用であ る。 好ましくは、 ホルモン (例、 エストロゲンなど) 非依存性、 HER2発現陰性な どのがん (例、 乳がんなど) の予防'治療剤、 アポトーシス作用促進剤である。 本発明のスクリ一ニング方法またはスクリーニング用キットを用いて得られる 化合物またはその塩を上述の予防。治療剤として使用する場合、 常套手段に従つ て製剤化することができる。 A compound or a salt thereof that regulates (preferably inhibits) the activity of the protein of the present invention and a compound or a salt thereof that regulates (preferably inhibits) the expression of a gene encoding the protein of the present invention are, for example, colon cancer, Breast, lung, prostate, esophagus, stomach, liver, biliary, spleen, kidney, bladder, uterus, ovary, testis, thyroid, It is useful as a prophylactic / therapeutic agent for cancer, such as ligament cancer, brain tumor or hematological tumor, and as an apoptosis promoter. Preferably, they are agents for preventing or treating cancers (eg, breast cancer, etc.) that are hormone-independent (eg, estrogen, etc.)-Independent, HER2-negative, etc., and for promoting apoptosis. The above-described prophylaxis of a compound or a salt thereof obtained by using the screening method or the screening kit of the present invention. When used as a therapeutic agent, follow conventional procedures Can be formulated.
例えば、 経口投与のための組成物としては、 固体または液体の剤形、 具体的に は錠剤 (糖衣錠、 フィルムコーティング錠を含む) 、 丸剤、 顆粒剤、 散剤、 カブ セル剤 (ソフトカプセル剤を含む) 、 シロップ剤、 乳剤、 懸濁剤などがあげられ る。 かかる組成物は自体公知の方法によって製造され、 製剤分野において通常用 いられる担体、 希釈剤もしくは賦形剤を含有するものである。 例えば、 錠剤用の 担体、 陚形剤としては、 乳糖、 でんぷん、 蔗糖、 ステアリン酸マグネシウムなど が用いられる。  For example, compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets and film-coated tablets), pills, granules, powders, capsules (including soft capsules). ), Syrups, emulsions, suspensions and the like. Such a composition is produced by a method known per se and contains a carrier, diluent or excipient commonly used in the field of formulation. For example, lactose, starch, sucrose, magnesium stearate and the like are used as carriers and tablets for tablets.
非経口投与のための組成物としては、 例えば、 注射剤、 坐剤などが用いられ、 注射剤は静脈注射剤、 皮下注射剤、 皮内注射剤、 筋肉注射剤、 点滴注射剤、 関節 内注射剤などの剤形を包含する。 かかる注射剤は、 自体公知の方法に従って、 例 えば、 上記抗体またはその塩を通常注射剤に用いられる無菌の水性もしくは油性 液に溶解、 懸濁または乳化することによって調製する。 注射用の水性液としては、 例えば、 生理食塩水、 ブドウ糖やその他の補助薬を含む等張液などが用いられ、 適当な溶解補助剤、 例えば、 アルコ一ル (例、 エタノール) 、 ポリアルコール (例、 プロピレングリコール、 ポリエチレングリコ一ル) 、 非イオン界面活性剤 〔例、 ポリソルベート 80、 HCO-50 (po lyoxye thy 1 ene (50mo 1) adduc t of  As compositions for parenteral administration, for example, injections, suppositories, etc. are used. Injections are intravenous, subcutaneous, intradermal, intramuscular, intravenous, intraarticular. And dosage forms such as agents. Such injections are prepared according to a method known per se, for example, by dissolving, suspending or emulsifying the antibody or a salt thereof in a sterile aqueous or oily liquid commonly used for injections. As an aqueous solution for injection, for example, physiological saline, isotonic solution containing glucose and other adjuvants and the like are used, and suitable solubilizing agents, for example, alcohol (eg, ethanol), polyalcohol ( For example, propylene glycol, polyethylene glycol), nonionic surfactants [for example, polysorbate 80, HCO-50 (po lyoxye thy 1 ene (50mo 1) adduc t of
hydrogenated cas tor oi l) 〕 などと併用してもよい。 油性液としては、 例えば、 ゴマ油、 大豆油などが用いられ、 溶解補助剤として安息香酸ベンジル、 ベンジル アルコールなどを併用してもよい。 調製された注射液は、 通常、 適当なアンプル に充填される。 直腸投与に用 られる坐剤は、 上記抗体またはその塩を通常の坐 薬用基剤に混合することによって調製される。 hydrogenated castoroi l)]]. As the oily liquid, for example, sesame oil, soybean oil and the like are used, and benzyl benzoate, benzyl alcohol and the like may be used in combination as a solubilizing agent. The prepared injection solution is usually filled in a suitable ampoule. A suppository for rectal administration is prepared by mixing the above antibody or a salt thereof with a conventional suppository base.
上記の経口用または非経口用医薬組成物は、 活性成分の投与量に適合するよう な投薬単位の剤形に調製されることが好都合である。 かかる投薬単位の剤形とし ては、 錠剤、 丸剤、 カプセル剤、 注射剤 (アンプル) 、 坐剤などが例示され、 そ れぞれの投薬単位剤形当たり通常 5〜5 0' 0 m g、 とりわけ注射剤では 5〜 1 0 0 m g、 その他の剤形では 1 0〜2 5 O m gの上記抗体が含有されていることが 好ましい。  The above-mentioned oral or parenteral pharmaceutical composition is conveniently prepared in a unit dosage form adapted to the dose of the active ingredient. Examples of such dosage unit dosage forms include tablets, pills, capsules, injections (ampoules), and suppositories, and usually 5 to 50 mg / dosage per dosage unit dosage form. In particular, it is preferable that the injection contains 5 to 100 mg of the above-mentioned antibody, and other dosage forms contain 10 to 25 mg of the above-mentioned antibody.
なお前記した各組成物は、 上記抗体との配合により好ましくない相互作用を生 じない限り他の活性成分を含有してもよい。 In addition, each of the above-mentioned compositions may cause an undesired interaction due to the combination with the above antibody. Unless otherwise specified, other active ingredients may be contained.
このようにして得られる製剤は安全で低毒性であるので、 例えば、 ヒトまたは 温血動物 (例えば、 マウス、 ラット、 ゥサギ、 ヒッジ、 ブ夕、 ゥシ、 ゥマ、 トリ、 ネコ、 ィヌ、 サル、 チンパンジーなど) に対して経口的にまたは非経口的に投与 することができる。  The preparations obtained in this way are safe and low toxic and can be used, for example, in humans or in warm-blooded animals (eg mice, rats, puppies, sheep, bush, pussi, puma, birds, cats, dogs, Monkeys, chimpanzees, etc.) orally or parenterally.
該化合物またはその塩の投与量は、 その作用、 対象疾患、 投与対象、 投与ルー トなどにより差異はあるが、 例えば、 乳がんの治療の目的で本発明のタンパク質 の活性を調節する化合物またはその塩を経口投与する場合、 一般的に成人 (体重 60 kgとして) においては、 一日につき該化合物またはその塩を約 0.1〜1 0 Omg、 好ましくは約 1. 0〜5 Omg、 より好ましくは約 1. 0〜20mg 投与する。 非経口的に投与する場合は、 該化合物またはその塩の 1回投与量は投 与対象、 対象疾患などによっても異なるが、 例えば、 乳がんの治療の目的で本発 明のタンパク質の活性を調節する化合物またはその塩を注射剤の形で通常成人 (体重 6 O kgとして) に投与する場合、 一日につき該化合物またはその塩を約 0. 01〜3 Omg、 好ましくは約 0. 1〜 2 Omg、 より好ましくは約 0. 1 〜1 Omgをがん病変部に注射により投与するのが好都合である。 他の動物の場 合も、 体重 6 O kg当たりに換算した量を投与することができる。  The dose of the compound or a salt thereof varies depending on its action, target disease, subject to be administered, route of administration, and the like.For example, a compound or a salt thereof that regulates the activity of the protein of the present invention for the purpose of treating breast cancer. When the compound is orally administered, generally in an adult (assuming a body weight of 60 kg), the compound or a salt thereof is used in an amount of about 0.1 to 10 Omg, preferably about 1.0 to 5 Omg, more preferably about 1 to 10 Omg per day. Administer 0 to 20 mg. When administered parenterally, the single dose of the compound or a salt thereof varies depending on the administration subject, target disease, and the like, but, for example, regulates the activity of the protein of the present invention for the purpose of treating breast cancer. When a compound or a salt thereof is administered to an adult (with a body weight of 6 O kg) usually in the form of an injection, the compound or a salt thereof is about 0.01 to 3 Omg, preferably about 0.1 to 2 Omg per day. More preferably, about 0.1 to 1 Omg is administered by injection to the cancerous lesion. In the case of other animals, the dose can be administered in terms of the weight per 6 O kg.
(2) 本発明のタンパク質、 その部分ペプチドまたはその塩の定量 (2) Quantification of the protein of the present invention, its partial peptide or its salt
本発明のタンパク質に対する抗体 (以下、 本発明の抗体と略記する場合があ る) は、 本発明のタンパク質を特異的に認識することができるので、 被検液中の 本発明のタンパク質の定量、 特にサンドイッチ免疫測定法による定量などに使用 することができる。  An antibody against the protein of the present invention (hereinafter sometimes abbreviated as the antibody of the present invention) can specifically recognize the protein of the present invention. In particular, it can be used for quantification by sandwich immunoassay.
すなわち、 本発明は、  That is, the present invention
(i) 本発明の抗体と、 被検液および標識化された本発明のタンパク質とを競合 的に反応させ、 該钪体に結合した標識化された本発明のタンパク質の割合を測定 することを特徴とする被検液中の本発明の夕ンパク質の定量法、 および  (i) reacting the antibody of the present invention with a test solution and the labeled protein of the present invention competitively, and measuring the ratio of the labeled protein of the present invention bound to the antibody. A method for quantifying the protein of the present invention in a test solution characterized by:
( i i ) 被検液と担体上に不溶化した本発明の抗体および標識化された本発明の別 の抗体とを同時あるいは連続的に反応させたのち、 不溶化担体上の標識剤の活性 を測定することを特徴とする被検波中の本発明のタンパク質の定量法を提供する。 上記 (i i) の定量法においては、 一方の抗体が本発明のタンパク質の N端部を 認識する抗体で、 他方の抗体が本発明のタンパク質の C端部に反応する抗体であ ることが望ましい。 (ii) After reacting the test solution with the antibody of the present invention insoluble on the carrier and another labeled antibody of the present invention simultaneously or continuously, the activity of the labeling agent on the insoluble carrier is determined. The present invention provides a method for quantifying the protein of the present invention in a test wave, characterized by measuring In the quantification method (ii) above, it is desirable that one antibody is an antibody that recognizes the N-terminal of the protein of the present invention and the other antibody is an antibody that reacts with the C-terminal of the protein of the present invention. .
また、 本発明のタンパク質に対するモノクローナル抗体 (以下、 本発明のモノ クローナル抗体と称する場合がある) を用いて本発明のタンパク質の定量を行な えるほか、 組織染色等による検出を行なうこともできる。 これらの目的には、 抗 体分子そのものを用いてもよく、 また、 抗体分子の F ( a b ' ) 2 、 F a b '、 あ るいは F a b画分を用いてもよい。 In addition, the protein of the present invention can be quantified using a monoclonal antibody against the protein of the present invention (hereinafter sometimes referred to as the monoclonal antibody of the present invention), and can also be detected by tissue staining or the like. For these purposes, the antibody molecule itself may be used, or the F (ab ') 2 , Fab', or Fab fraction of the antibody molecule may be used.
本発明の抗体を用いる本発明のタンパク質の定量法は、 特に制限されるべきも のではなく、 被測定液中の抗原量 (例えば、 タンパク質量) に対応した抗体、 抗 原もしくは抗体一抗原複合体の量を化学的または物理的手段により検出し、 これ を既知量の抗原を含む標準液を用いて作製した標準曲線より算出する測定法であ れば、 いずれの測定法を用いてもよい。 例えば、 ネフロメトリー、 競合法、 ィム ノメトリック法およびサンドィツチ法が好適に用いられるが、 感度、 特異性の点 で、 後述するサンドイッチ法を用いるのが特に好ましい。  The method for quantifying the protein of the present invention using the antibody of the present invention is not particularly limited, and may be an antibody, an antigen, or an antibody-antigen complex corresponding to the amount of antigen (eg, the amount of protein) in the test solution. Any measurement method may be used as long as the amount of the body is detected by chemical or physical means, and this is calculated from a standard curve prepared using a standard solution containing a known amount of antigen. . For example, nephelometry, competition method, immunometric method and sandwich method are preferably used, but in terms of sensitivity and specificity, it is particularly preferable to use a sandwich method described later.
標識物質を用いる測定法に用いられる標識剤としては、 例えば、 放射性同位元 素、 酵素、 蛍光物質、 発光物質などが用いられる。 放射性同位元素としては、 例 えば、 〔125ι〕 、 〔1311〕 、 〔¾〕 、 〔14c〕 などが用いられる。 上記酵素としては、 安定で比活性の大きなものが好ましく、 例えば、 3—ガラクトシダーゼ、 )3—グ ルコシダーゼ、 アルカリフォスファターゼ、 パーォキシダ一ゼ、 リンゴ酸脱水素 酵素などが用いられる。 蛍光物質としては、 例えば、 フルォレスカミン、 フルォ レツセンイソチオシァネ一トなどが用いられる。 発光物質としては、 例えば、 ル ミノ一ル、 ルミノール誘導体、 ルシフェリン、 ルシゲニンなどが用いられる。 さ らに、 抗体あるいは抗原と標識剤との結合にピオチン—アビジン系を用いること もできる。 As a labeling agent used in a measurement method using a labeling substance, for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance and the like are used. Radioisotopes, if example embodiment, [125 iota], [131 1], [¾], and [14 c] used. As the above enzyme, a stable enzyme having a large specific activity is preferable. For example, 3-galactosidase,) 3-glucosidase, alkaline phosphatase, peroxidase, malate dehydrogenase and the like are used. As the fluorescent substance, for example, fluorescamine, fluorescein isothiocyanate and the like are used. As the luminescent substance, for example, luminol, luminol derivative, luciferin, lucigenin and the like are used. Furthermore, a biotin-avidin system can be used for binding the antibody or antigen to the labeling agent.
抗原あるいは抗体の不溶化に当っては、 物理吸着を用いてもよく、 また通常夕 ンパク質あるいは酵素等を不溶化、 固定化するのに用いられる化学結合を用いる 方法でもよい。 担体としては、 ァガロース、 デキストラン、 セルロースなどの不 溶性多糖類、 ポリスチレン、 ポリアクリルアミド、 シリコン等の合成樹脂、 ある いはガラス等が挙げられる。 For the insolubilization of the antigen or antibody, physical adsorption may be used, or a method using a chemical bond usually used for insolubilizing and immobilizing proteins or enzymes may be used. Carriers such as agarose, dextran, and cellulose Examples include soluble polysaccharides, synthetic resins such as polystyrene, polyacrylamide, and silicon, and glass.
サンドイッチ法においては不溶化した本発明のモノクローナル抗体に被検液を 反応させ (1次反応) 、 さらに標識化した別の本発明のモノクローナル抗体を反 応させ (2次反応) たのち、 不溶化担体上の標識剤の活性を測定することにより 被検波中の本発明のタンパク質量を定量することができる。 1次反応と 2次反応 は逆の順序に行っても、 また、 同時に行なってもよいし時間をずらして行なって もよい。 標識化剤および不溶化の方法は前記のそれらに準じることができる。 ま た、 サンドイッチ法による免疫測定法において、 固相用抗体あるいは標識用抗体 に用いられる抗体は必ずしも 1種類である必要はなく、 測定感度を向上させる等 の目的で 2種類以上の抗体の混合物を用いてもよい。  In the sandwich method, the test solution is reacted with the insolubilized monoclonal antibody of the present invention (primary reaction), and further reacted with another labeled monoclonal antibody of the present invention (secondary reaction). By measuring the activity of the labeling agent, the amount of the protein of the present invention in the test wave can be quantified. The primary reaction and the secondary reaction may be performed in the reverse order, may be performed simultaneously, or may be performed at staggered times. The labeling agent and the method of insolubilization can be in accordance with those described above. Also, in the immunoassay by the sandwich method, the antibody used for the solid phase antibody or the labeling antibody does not necessarily need to be one kind, and a mixture of two or more kinds of antibodies is used for the purpose of improving measurement sensitivity and the like. May be used.
本発明のサンドィツチ法による本発明のタンパク質の測定法においては、 1次 反応と 2次反応に用いられる本発明のモノク口一ナル抗体は、 本発明の夕ンパク 質の結合する部位が相異なる抗体が好ましく用いられる。 すなわち、 1次反応お よび 2次反応に用いられる抗体は、 例えば、 2次反応で用いられる抗体が、 本発 明のタンパク質の C端部を認識する場合、 1次反応で用いられる抗体は、 好まし くは C端部以外、 例えば N端部を認識する抗体が用いられる。  In the method for measuring the protein of the present invention by the sandwich method of the present invention, the monoclonal antibody of the present invention used in the primary reaction and the secondary reaction is an antibody having different binding sites to the protein of the present invention. Is preferably used. That is, the antibody used in the primary reaction and the secondary reaction is, for example, when the antibody used in the secondary reaction recognizes the C-terminal of the protein of the present invention, the antibody used in the primary reaction is Preferably, an antibody that recognizes other than the C-terminal, for example, the N-terminal, is used.
本発明のモノクロ一ナル抗体をサンドイツチ法以外の測定システム、 例えば、 競合法、 ィムノメトリック法あるいはネフロメトリーなどに用いることができる。 競合法では、 被検液中の抗原と標識抗原とを抗体に対して競合的に反応させた のち、 未反応の標識抗原(F) と、 抗体と結合した檩識抗原 (B ) とを分離し The monoclonal antibody of the present invention can be used in a measurement system other than the San Deutsch method, for example, a competition method, an immunometric method, or a nephrometry. In the competition method, the antigen in the test solution and the labeled antigen are allowed to react competitively with the antibody, and then the unreacted labeled antigen (F) and the recognized antigen (B) bound to the antibody are separated. I
(BZ F分離) 、 B, Fいずれかの標識量を測定し、 被検波中の坊原量を定量す る。 本反応法には、.抗体として可溶性抗体を用い、 B /F分離をポリエチレング リコール、 前記抗体に対する第 2抗体などを用いる液相法、 および、 第 1抗体と して固相化抗体を用いるか、 あるいは、 第 1抗体は可溶性のものを用い第 2抗体 として固相化抗体を用いる固相化法とが用いられる。 (BZF separation) Measure the amount of labeling of either B or F, and quantify the amount of Bobara in the test wave. In this reaction method, a soluble antibody was used as the antibody, B / F separation was performed using polyethylene glycol, a liquid phase method using a second antibody against the antibody, and a solid-phased antibody was used as the first antibody. Alternatively, a solid phase method using a soluble first antibody and a solid phase antibody as the second antibody is used.
ィムノメトリック法では、 被検液中の抗原と固相化抗原とを一定量の標識化抗 体に対して競合反応させた後固相と液相を分離するか、 あるいは、 被検液中の抗 原と過剰量の標識化抗体とを反応させ、 次に固相化抗原を加え未反応の標識化抗 体を固相に結合させたのち、 固相と液相を分離する。 次に、 いずれかの相の標識 量を測定し被検液中の抗原量を定量する。 In the immunometric method, the antigen in the test solution and the immobilized antigen are subjected to a competitive reaction with a certain amount of labeled antibody, and then the solid phase and the liquid phase are separated. Reaction with an excess amount of the labeled antibody, and then adding immobilized antigen to the unreacted labeled antibody. After binding the body to the solid phase, the solid and liquid phases are separated. Next, the amount of label in either phase is measured to determine the amount of antigen in the test solution.
また、 ネフロメトリーでは、 ゲル内あるいは溶液中で抗原抗体反応の結果生じ た不溶性の沈降物の量を測定する。 被検液中の抗原量が僅かであり、 少量の沈降 物しか得られない場合にもレーザーの散乱を利用するレーザ一ネフロメトリーな どが好適に用いられる。  In nephelometry, the amount of insoluble sediment resulting from the antigen-antibody reaction in a gel or in a solution is measured. Even when the amount of antigen in the test solution is small and only a small amount of sediment is obtained, laser-nephrometry utilizing laser scattering is preferably used.
これら個々の免疫学的測定法を本発明の定量方法に適用するにあたっては、 特 別の条件、 操作等の設定は必要とされない。 それぞれの方法における通常の条件、 操作法に当業者の通常の技術的配慮を加えて本発明のタンパク質の測定系を構築 すればよい。 これらの一般的な技術手段の詳細については、 総説、 成書などを参 照することができる。  In applying these individual immunological measurement methods to the quantification method of the present invention, no special conditions, operations, and the like need to be set. The protein measuring system of the present invention may be constructed by adding ordinary technical considerations of those skilled in the art to ordinary conditions and operation methods in each method. For details of these general technical means, reference can be made to reviews, documents, etc.
例えば、 入江 寛編 「ラジオィムノアツセィ」 (講談社、 昭和 49年発行) 、 入 江 寛編 「続ラジオィムノアツセィ」 (講談社、 昭和 54年発行) 、 石川栄治ら編 「酵素免疫測定法」 (医学書院、 昭和 53年発行) 、 石川栄治ら編 「酵素免疫測定 法」 (第 2版) (医学書院、 昭和 57年発行) 、 石川栄治ら編 「酵素免疫測定法」 (第 3版) (医学書院、 昭和 62年発行) 、 「Methods in ENZYMOLOGYJ Vol .  For example, edited by Hiro Irie, "Radio Imuno Atsushi" (Kodansha, published in 1974), edited by Hiro Irie, "Radio Imuno Atssey" (Kodansha, published in 1974), edited by Eiji Ishikawa et al. "The Enzyme Immunoassay Method" (2nd edition) (Medical Shoin, published in 1982), "Enzyme Immunoassay Method" (ed. 3rd Edition), edited by Eiji Ishikawa et al. Edition) (Medical Shoin, published in 1987), "Methods in ENZYMOLOGYJ Vol.
70 (Immunochemical Techniques (Par t A))、 同書 Vol. 73 (Immunochemical Techniques (Part B))、 同書 Vol. 74 (Immunochemical Techniques (Part C))、 同 書 Vol. 84 (Immunochemical Techniques (Par t D: Selec ted Immunoassays) ) , 同 書 Vol. 92 (Iimunochemical Techniques (Par t E:Monoc lonal Ant ibodies and General Immunoassay Methods)) 同書 Vol. 121 (Immunochemical 70 (Immunochemical Techniques (Part A)), ibid.Vol. 73 (Immunochemical Techniques (Part B)), ibid.Vol. 74 (Immunochemical Techniques (Part C)), ibid.Vol. 84 (Immunochemical Techniques (Part d: Selected Immunoassays)), ibid.Vol. 92 (Iimunochemical Techniques (Part E: Monoclonal Ant ibodies and General Immunoassay Methods)) ibid.Vol. 121 (Immunochemical
Techniques (Part I: Hybr idoia Technology and Monoc lonal Ant ibodies) ) (以上、 アカデミックプレス社発行)などを参照することができる。 Techniques (Part I: Hybrid Technology and Monoclonal Ant ibodies)) (above, published by Academic Press) can be referred to.
以上のようにして、 本発明の抗体を用いることによって、 本発明のタンパク質 を感度良く定量することができる。  As described above, the protein of the present invention can be quantified with high sensitivity by using the antibody of the present invention.
さらには、 本発明の抗体を用いて本発明の夕ンパク質の濃度を定量することに よって、 本発明のタンパク質の濃度の増加または減少が検出された場合、 例えば 大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝臓がん、 胆道が ん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣がん、 甲状腺がん、 塍臓がん、 脳腫瘍または血液腫瘍などのがんである、 または将来罹患する可能性 が高いと診断することができる。 Furthermore, when an increase or decrease in the concentration of the protein of the present invention is detected by quantifying the concentration of the protein of the present invention using the antibody of the present invention, for example, colon cancer, breast cancer, lung cancer, Prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid cancer, It can be diagnosed as having cancer, such as kidney cancer, brain tumor or hematological tumor, or is more likely to be affected in the future.
また、 本発明の抗体は、 体液や組織などの被検体中に存在する本発明のタンパ 'ク質を検出するために使用することができる。 また、 本発明のタンパク質を精製 するために使用する抗体カラムの作製、 精製時の各分画中の本発明のタンパク質 の検出、 被検細胞内における本発明のタンパク質の挙動の分析などのために使用 することができる。  Further, the antibody of the present invention can be used for detecting the protein of the present invention present in a subject such as a body fluid or a tissue. In addition, for the preparation of an antibody column used for purifying the protein of the present invention, the detection of the protein of the present invention in each fraction during purification, and the analysis of the behavior of the protein of the present invention in test cells, etc. Can be used.
(3) 遺伝子診断薬 (3) Genetic diagnostics
本発明の DNAは、 例えば、 プロ一ブとして使用することにより、 ヒトまたは 温血動物 (例えば、 ラット、 マウス、 モルモット、 ゥサギ、 トリ、 ヒッジ、 ブタ、 ゥシ、 ゥマ、 ネコ、 ィヌ、 サル、 チンパンジ一など) における本発明のタンパク 質またはその部分ペプチドをコードする DNAまたは mRNAの異常 (遺伝子異 常) を検出することができるので、 例えば、 該 DN Aまたは mRNAの損傷、 突 然変異あるいは発現低下や、 該 DNAまたは mRNAの増加あるいは発現過多な どの遺伝子診断薬として有用である。  The DNA of the present invention can be used, for example, as a probe to produce human or warm-blooded animals (e.g., rats, mice, guinea pigs, egrets, birds, higgies, pigs, pigs, dogs, cats, dogs, Abnormalities (genetic abnormalities) in the DNA or mRNA encoding the protein of the present invention or its partial peptide in monkeys, chimpanzees, etc.). Alternatively, it is useful as a diagnostic agent for genes such as decreased expression, increased DNA or mRNA or excessive expression.
本発明の DNAを用いる上記の遺伝子診断は、 例えば、 自体公知のノーザンハ イブリダィゼーションゃ PCR— S S CP法 (Genomics,第 5巻, 874〜879頁 Q989 年) 、 Proceedings of the National Academy of Sciences of the United States of America,第 86巻, 2766~2770頁(1989年))などにより実施することがで さる。  The above-described genetic diagnosis using the DNA of the present invention includes, for example, the known Northern Hybridization ゃ PCR-SSCP method (Genomics, Vol. 5, pp. 874-879, Q989), Proceedings of the National Academy of Sciences of the United States of America, Vol. 86, pp. 2766-2770 (1989)).
例えば、 ノーザンハイブリダィゼ一ションにより発現過多または減少が検出さ れた場合や PCR— S SCP法により DNAの突然変異が検出された場合は、 例 えば大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝臓がん、 胆 道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣がん、 甲状腺 がん、 勝臓がん、 脳腫瘍または血液腫瘍などのがんである可能性が高いと診断す ることができる。  For example, if overexpression or decrease is detected by Northern hybridization or DNA mutation is detected by PCR-SSCP method, for example, colon cancer, breast cancer, lung cancer, and prostate Cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testis cancer, thyroid cancer, visceral cancer, It can be diagnosed as having a high possibility of cancer such as a brain tumor or a blood tumor.
(4) アンチセンスポリヌクレオチドを含有する医薬 本発明の D NAに相補的に結合し、 該 D N Aの発現を抑制することができる本 発明のアンチセンスポリヌクレオチドは低毒性であり、 生体内における本発明の タンパク質または本発明の D NAの機能 (例、 リガンド結合活性またはシグナル 伝達活性) を抑制することができるので、 例えば大腸がん、 乳がん、 肺がん、 前 立腺がん、 食道がん、 胃がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱が ん、 子宮がん、 卵巣がん、 精巣がん、 甲状腺がん、 塍臓がん、 脳腫瘍または血液 腫瘍などのがんの予防 ·治療剤として使用することができる。 好ましくは、 ホル モン (例、 エストロゲンなど) 非依存性、 HER2発現陰性などのがん (例、 乳がん など) の予防 ·治療剤である。 また、 本発明のアンチセンスポリヌクレオチドは 例えば、 アポト一シス作用調節剤 (好ましくはアポト一シス作用促進剤) として 使用することもできる。 (4) a drug containing an antisense polynucleotide The antisense polynucleotide of the present invention, which can complementarily bind to the DNA of the present invention and suppress the expression of the DNA, has low toxicity, and functions of the protein of the present invention or the DNA of the present invention in vivo. (Eg, ligand binding activity or signal transduction activity), such as colon cancer, breast cancer, lung cancer, prostate cancer, esophagus cancer, stomach cancer, liver cancer, biliary tract cancer, and spleen Can be used as a prophylactic / therapeutic agent for cancer such as cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid cancer, kidney cancer, brain tumor or blood tumor . Preferably, it is a preventive / therapeutic agent for cancer (eg, breast cancer, etc.) that is hormone-independent (eg, estrogen, etc.)-Independent, HER2 expression-negative, etc. In addition, the antisense polynucleotide of the present invention can be used, for example, as an apoptotic action regulator (preferably an apoptotic action promoter).
上記ァンチセンスポリヌクレオチドを上記の予防 ·治療剤、 調節剤として使用 する場合、 自体公知の方法に従って製剤化し、 投与することができる。  When the antisense polynucleotide is used as the above-mentioned prophylactic / therapeutic agent or modulator, it can be formulated and administered according to a method known per se.
また、 例えば、 前記のアンチセンスポリヌクレオチドを単独あるいはレトロゥ ィルスベクター、 アデノウイルスベクタ一、 アデノウィルスァソシエーテツドゥ ィルスべクタ一などの適当なベクターに挿入した後、 常套手段に従って、 ヒトま たは哺乳動物 (例、 ラット、 ゥサギ、 ヒッジ、 ブ夕、 ゥシ、 ネコ、 ィヌ、 サルな ど) に対して経口的または非経口的に投与することができる。 該アンチセンスポ リヌクレオチドは、 そのままで、 あるいは摂取促進のために補助剤などの生理学 的に認められる担体とともに製剤化し、 遺伝子銃やハイドロゲルカテーテルのよ うなカテーテルによって投与できる。 あるいは、 エア口ゾル化して吸入剤として 気管内に局所投与することもできる。  In addition, for example, the above-mentioned antisense polynucleotide is inserted alone or into a suitable vector such as a retrovirus vector, an adenovirus vector, an adenovirus association virus vector, etc. It can be administered orally or parenterally to mammals (eg, rats, egrets, sheep, sheep, bush, birds, cats, dogs, monkeys, etc.). The antisense polynucleotide can be administered as it is or in the form of a formulation together with a physiologically acceptable carrier such as an adjuvant for promoting uptake, and administered by a gene gun or a catheter such as a hydrogel catheter. Alternatively, they can be made into an aerosol and administered topically into the trachea as an inhalant.
さらに、 体内動態の改良、 半減期の長期化、 細胞内取り込み効率の改善を目的 に、 前記のアンチセンスポリヌクレオチドを単独またはリポゾ一ムなどの担体と ともに製剤 (注射剤) 化し、 静脈、 皮下等に投与してもよい。  Furthermore, for the purpose of improving pharmacokinetics, prolonging the half-life, and improving the efficiency of cellular uptake, the above-mentioned antisense polynucleotide is formulated alone or together with a carrier such as liposome (injection), and is intravenously or subcutaneously. Etc. may be administered.
該アンチセンスポリヌクレオチドの投与量は、 対象疾患、 投与対象、 投与ルー トなどにより差異はあるが、 例えば、 乳がんの治療の目的で本発明のアンチセン スポリヌクレオチドを投与する場合、 一般的に成人 (体重 6 0 k g ) においては、 一日につき該アンチセンスポリヌクレオチドを約 0 . 1 ~ 1 0 O m g投与する。 03 007926 The dose of the antisense polynucleotide varies depending on the disease to be treated, the subject of administration, the route of administration, and the like.For example, when the antisense polynucleotide of the present invention is administered for the purpose of treating breast cancer, the dose is generally used for adults. (Weight 60 kg), the antisense polynucleotide is administered in an amount of about 0.1 to 10 O mg per day. 03 007926
44 さらに、 該アンチセンスポリヌクレオチドは、 組織や細胞における本発明の D N Aの存在やその発現状況を調べるための診断用オリゴヌクレオチドプローブと して使用することもできる。  44 Further, the antisense polynucleotide can be used as a diagnostic oligonucleotide probe for examining the presence or expression of the DNA of the present invention in tissues or cells.
上記アンチセンスポリヌクレオチドと同様に、 本発明のタンパク質をコードす る R NAの一部を含有する二重鎖 R NA、 本発明のタンパク質をコードする R N Aの一部を含有するリポザィムなども、 本発明の遺伝子の発現を抑制することが でき、 生体内における本発明で用いられるタンパク質または本発明で用いられる D NAの機能を抑制することができるので、 例えば、 例えば大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝臓がん、 胆道がん、 脾臓がん、 腎が ん、 膀胱がん、,子宮がん、 卵巣がん、 精巣がん、 甲状腺がん、 滕臓がん、 脳腫瘍 または血液腫瘍などのがんの予防 ·治療剤、 好ましくは、 ホルモン (例、 ェスト ロゲンなど) 非依存性、 HER2発現陰性などのがん (例、 乳がんなど) の予防 *治 療剤、 さらにアポトーシス作用調節剤などとして使用することができる。  Similarly to the above-mentioned antisense polynucleotide, a double-stranded RNA containing a part of the RNA encoding the protein of the present invention, a lipozyme containing a part of the RNA encoding the protein of the present invention, etc. Since the expression of the gene of the present invention can be suppressed and the function of the protein used in the present invention or the DNA used in the present invention in vivo can be suppressed, for example, for example, colon cancer, breast cancer, lung cancer, Prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, renal cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid cancer, thyroid cancer Prevention and treatment of cancer such as cancer, brain tumor or blood tumor, preferably prevention of cancer (eg, breast cancer, etc.) independent of hormones (eg, estrogen), HER2 negative, etc.Furthermore, it can be used as an apoptosis action regulator or the like.
二重鎖 R NAは、 公知の方法 (例、 Nature, 411巻, 494頁, 2001年) に準じて、 本発明のポリヌクレオチドの配列を基に設計して製造することができる。  The double-stranded RNA can be produced by designing based on the sequence of the polynucleotide of the present invention according to a known method (eg, Nature, 411, 494, 2001).
リポザィムは、 公知の方法 (例、 TRENDS in Molecular Medic ine, 7巻, 221頁, 2001年) に準じて、 本発明のポリヌクレオチドの配列を基に設計して製造するこ とができる。 例えば、 本発明のタンパク質をコードする R N Aの一部に公知のリ ポザィムを連結することによって製造することができる。 本発明のタンパク質を コードする R NAの一部としては、 公知のリポザィムによって切断され得る本発 明の R NA上の切断部位に近接した部分 (R NA断片) が挙げられる。  The lipozyme can be designed and produced based on the sequence of the polynucleotide of the present invention according to a known method (eg, TRENDS in Molecular Medicine, Vol. 7, pp. 221, 2001). For example, it can be produced by linking a known lipozyme to a part of RNA encoding the protein of the present invention. A part of the RNA encoding the protein of the present invention includes a portion (RNA fragment) close to the cleavage site on the RNA of the present invention, which can be cleaved by a known lipozyme.
上記の二重鎖 R N Aまたはリポザィムを上記予防。治療剤として使用する場合、 ァンチセンスポリヌクレオチドと同様にして製剤化し、 投与することができる。 ( 5 ) 本発明の抗体を含有する医薬  Prevention of the above double-stranded RNA or lipozyme. When used as a therapeutic agent, it can be formulated and administered in the same manner as an antisense polynucleotide. (5) a drug containing the antibody of the present invention
本発明のタンパク質の活性を中和する作用を有する本発明の抗体は、 例えば大 腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣がん、 甲状腺がん、 膝 臓がん、 脳腫瘍または血液腫瘍などのがんの予防 ·治療剤として使用することが できる。 好ましくは、 ホルモン (例、 エストロゲンなど) 非依存性、 HER2発現陰 性などのがん (例、 乳がんなど) の予防 ·治療剤である。 また、 本発明の抗体は、 例えば、 アポトーシス作用調節剤 (好ましくはアポトーシス作用促進剤) として 使用することもできる。 The antibody of the present invention, which has the activity of neutralizing the activity of the protein of the present invention, includes, for example, colon cancer, breast cancer, lung cancer, prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer It can be used as a prophylactic or therapeutic agent for cancers such as renal, bladder, uterine, ovarian, testicular, thyroid, knee, brain or blood tumors it can. Preferably, it is a prophylactic / therapeutic agent for cancer (eg, breast cancer, etc.) that is hormone-independent (eg, estrogen, etc.)-Independent, HER2-expressing negative, etc. Further, the antibody of the present invention can also be used, for example, as an apoptosis action regulator (preferably, an apoptosis action promoter).
本発明の抗体を含有する上記疾患の予防'治療剤、 調節剤は低毒性であり、 そ のまま液剤として、 または適当な剤型の医薬組成物として、 ヒトまたは哺乳動物 (例、 ラット、 ゥサギ、 ヒッジ、 ブ夕、 ゥシ、 ネコ、 ィヌ、 サルなど) に対して 経口的または非経口的 (例、 血管内投与、 皮下投与など) に投与することができ る。  The prophylactic or therapeutic agent for the above-mentioned diseases, which comprises the antibody of the present invention, has low toxicity, and is used as it is as a liquid or as a pharmaceutical composition in an appropriate dosage form, in humans or mammals (eg, rat, rabbit, etc.). It can be given orally or parenterally (eg, intravenously, subcutaneously, etc.) to cattle, higgins, bushes, puppies, cats, dogs, monkeys, etc.).
本発明の抗体は、 それ自体を投与しても良いし、 または適当な医薬組成物とし て投与しても良い。 投与に用いられる医薬組成物としては、 本発明の抗体および その塩と薬理学的に許容され得る担体、 希釈剤もしくは賦形剤とを含むものであ つても良い。 このような医薬組成物は、 経口または非経口投与に適する剤形とし て提供される。  The antibody of the present invention may be administered as it is, or may be administered as a suitable pharmaceutical composition. The pharmaceutical composition used for administration may contain the antibody of the present invention or a salt thereof and a pharmacologically acceptable carrier, diluent or excipient. Such a pharmaceutical composition is provided as a dosage form suitable for oral or parenteral administration.
非経口投与のための組成物としては、 例えば、 注射剤、 坐剤等が用いられ、 注 射剤は静脈注射剤、 皮下注射剤、 皮内注射剤、 筋肉注射剤、 点滴注射剤等の剤形 を包含しても良い。 このような注射剤は、 公知の方法に従って調製できる。 注射 剤の調製方法としては、 例えば、 上記本発明の抗体またはその塩を通常注射剤に 用いられる無菌の水性液、 または油性液に溶解、 懸濁または乳化することによつ て調製できる。 注射用の水性液としては、 例えば、 生理食塩水、 ブドウ糖やその 他の補助薬を含む等張液等が用いられ、 適当な溶解補助剤、 例えば、 アルコール (例、 エタノール) 、 ポリアルコール (例、 プロピレングリコール、 ポリエチレ ングリコール) 、 非イオン界面活性剤 〔例、 ポリソルべ一ト 80、 HC0-50  Examples of compositions for parenteral administration include injections, suppositories, etc., and injections include intravenous, subcutaneous, intradermal, intramuscular, and intravenous injections. Shapes may be included. Such an injection can be prepared according to a known method. Injections can be prepared, for example, by dissolving, suspending or emulsifying the antibody of the present invention or a salt thereof in a sterile aqueous liquid or oily liquid commonly used for injections. As an aqueous solution for injection, for example, physiological saline, isotonic solution containing glucose and other adjuvants, and the like, suitable solubilizing agents, for example, alcohol (e.g., ethanol), polyalcohol (e.g., , Propylene glycol, polyethylene glycol), nonionic surfactants [eg, Polysorbate 80, HC0-50
(po lyoxye thy 1 ene (50mo 1) adduc t o f hydrogenated cas tor o i l) 〕 等と併用し てもよい。 油性液としては、 例えば、 ゴマ油、 大豆油等が用いられ、 溶解補助剤 として安息香酸ベンジル、 ベンジルアルコール等を併用してもよい。 調製された 注射液は、 適当なアンプルに充填されることが好ましい。 直腸投与に用いられる 坐剤は、 上記抗体またはその塩を通常の坐薬用基剤に混合することによって調製 されても良い。 経口投与のための組成物としては、 固体または液体の剤形、 具体的には錠剤 (糖衣錠、 フィルムコーティング錠を含む) 、 丸剤、 顆粒剤、 散剤、 カプセル剤 (ソフトカプセル剤を含む) 、 シロップ剤、 乳剤、 懸濁剤等が挙げられる。 この ような組成物は公知の方法によって製造され、 製剤分野において通常用いられる 担体、 希釈剤もしくは賦形剤を含有していても良い。 錠剤用の担体、 賦形剤とし ては、 例えば、 乳糖、 でんぷん、 蔗糖、 ステアリン酸マグネシウムが用いられる。 上記の非経口用または経口用医薬組成物は、 活性成分の投与量に適合するよう な投薬単位の剤形に調製されることが好都合である。 このような投薬単位の剤形 としては、 例えば、 錠剤、 丸剤、 カプセル剤、 注射剤 (アンプル) 、 坐剤が挙げ られる。 抗体の含有量としては、 投薬単位剤形当たり通常 5〜500mg、 とり わけ注射剤では 5〜100mg、 その他の剤形では 10〜25 Omgの上記抗体 が含有されていることが好ましい。 (polyoxye thy 1 ene (50mo 1) adduc tof hydrogenated cas tor oil)]. As the oily liquid, for example, sesame oil, soybean oil and the like are used, and benzyl benzoate, benzyl alcohol and the like may be used in combination as a solubilizing agent. The prepared injection solution is preferably filled in a suitable ampoule. A suppository for rectal administration may be prepared by mixing the antibody or a salt thereof with a usual suppository base. Compositions for oral administration include solid or liquid dosage forms, specifically tablets (including dragees and film-coated tablets), pills, granules, powders, capsules (including soft capsules), syrups Agents, emulsions, suspensions and the like. Such compositions are prepared by known methods and may contain carriers, diluents or excipients commonly used in the field of formulation. As carriers and excipients for tablets, for example, lactose, starch, sucrose, and magnesium stearate are used. The above-mentioned parenteral or oral pharmaceutical composition is conveniently prepared in a unit dosage form so as to be compatible with the dosage of the active ingredient. Such dosage unit forms include, for example, tablets, pills, capsules, injections (ampoules), and suppositories. The content of the antibody is preferably 5 to 500 mg per dosage unit dosage form, particularly 5 to 100 mg for injection, and 10 to 25 Omg for other dosage forms.
本発明の抗体を含有する予防 ·治療剤または診断剤 (医薬) の投与量は、 投与 対象、 対象疾患、 症状、 投与ルートなどによっても異なるが、 例えば、 成人の乳 がんの治療 '予防のために使用する場合には、 本発明の抗体を 1回量として、 通 常 0.01〜2 OmgZkg体重程度、 好ましくは 0.:!〜 1 OmgZkg体重程 度、 さらに好ましくは 0. l〜5mgZkg体重程度を、 1日 1〜5回程度、 好 ましくは 1日 1〜3回程度、 静脈注射により投与するのが好都合である。 他の非 経口投与および経口投与の場合もこれに準ずる量を投与することができる。 症状 が特に重い場合には、 その症状に応じて増量してもよい。  The dosage of the prophylactic / therapeutic or diagnostic agent (pharmaceutical) containing the antibody of the present invention varies depending on the administration target, target disease, symptoms, administration route, and the like. When used, the antibody of the present invention is used in a single dose, usually about 0.01 to 2 OmgZkg body weight, preferably about 0 :! to about 1 OmgZkg body weight, and more preferably about 0.1 to 5 mgZkg body weight. Is administered by intravenous injection about 1 to 5 times a day, preferably about 1 to 3 times a day. In the case of other parenteral administration and oral administration, an equivalent dose can be administered. If the symptoms are particularly severe, the dose may be increased accordingly.
本発明の抗体は、 それ自体または適当な医薬組成物として投与することができ る。 上記投与に用いられる医薬組成物は、 上記抗体またはその塩と薬理学的に許 容され得る担体、 希釈剤もしくは賦形剤とを含むものである。 かかる組成物は、 経口または非経口投与 (例、 血管内注射、 皮下注射など) に適する剤形として提 供される。  The antibodies of the present invention can be administered by themselves or as a suitable pharmaceutical composition. The pharmaceutical composition used for the administration contains the antibody or a salt thereof and a pharmacologically acceptable carrier, diluent or excipient. Such a composition is provided as a dosage form suitable for oral or parenteral administration (eg, intravascular injection, subcutaneous injection, etc.).
なお前記した各組成物は、 上記抗体との配合により好ましくない相互作用を生 じない限り他の活性成分を含有してもよい。  Each of the above-mentioned compositions may contain another active ingredient as long as the composition does not cause an undesirable interaction with the above-mentioned antibody.
(6) 本発明の 「RI PK2の活性調節 (好ましくは阻害) 作用を有する化合物 またはその塩を含有してなるがんの予防 ·治療剤」 および 「R I PK2の発現調 節 (好ましくは阻害) 作用を有する化合物またはその塩を含有してなるがんの予 防'治療剤」 について (6) The compound having the activity of regulating (preferably inhibiting) the activity of RI PK2 of the present invention Or a prophylactic / therapeutic agent for cancer containing a salt thereof or a "preventive / therapeutic agent for cancer containing a compound having an effect of regulating (preferably inhibiting) the expression of RI PK2 or a salt thereof". about
「R I PK2の活性調節 (好ましくは阻害) 作用を有する化合物」 は、 R I P K 2の活性調節 (好ましくは阻害) 作用を有する化合物であればいかなるもので もよく、 例えば大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝 臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣が ん、 甲状腺がん、 塍臓がん、 脳腫瘍または血液腫瘍などのがんの予防 ·治療剤と して用いられる。  The “compound having an activity of regulating (preferably inhibiting) the activity of RI PK2” may be any compound having an activity of regulating (preferably inhibiting) the activity of RIPK2, such as colon cancer, breast cancer, lung cancer, Prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid cancer, hepatic cancer It is used as a prophylactic / therapeutic agent for cancer such as cancer, brain tumor, or blood tumor.
「R I PK2の発現調節 (好ましくは阻害) 作用を有する化合物」 は、 R I P “Compound having an action of regulating (preferably inhibiting) the expression of R I PK2”
K2の発現調節 (好ましくは阻害) 作用を有する化合物であればいかなるもので もよく、 例えば大腸がん、 乳がん、 肺がん、前立腺がん、 食道がん、 胃がん、 肝 臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣が ん、 甲状腺がん、 塍臓がん、 脳腫瘍または血液腫瘍などのがんの予防 ·治療剤と して用いられる。 Any compound can be used as long as it has a K2 expression regulating (preferably inhibiting) action, such as colon cancer, breast cancer, lung cancer, prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, Used as a prophylactic and therapeutic agent for cancer such as spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid cancer, kidney cancer, brain tumor, or blood tumor Can be
該予防 ·治療剤は、 上記と同様にして製造される。  The prophylactic / therapeutic agent is produced in the same manner as described above.
(7) DNA転移動物 (7) DNA transfer animal
本発明は、 外来性の本発明のタンパク質をコードする DN A (以下、 本発明の 外来性 DN Aと略記する) またはその変異 DN A (本発明の外来性変異 DNAと 略記する場合がある) を有する非ヒ卜哺乳動物を提供する。  The present invention relates to a DNA encoding the exogenous protein of the present invention (hereinafter abbreviated as the exogenous DNA of the present invention) or a mutant DNA thereof (may be abbreviated as the exogenous mutant DNA of the present invention). And a non-human mammal having the formula:
すなわち、 本発明は、  That is, the present invention
(1) 本発明の外来性 DNAまたはその変異 DNAを有する非ヒト哺乳動物、 (1) a non-human mammal having the exogenous DNA of the present invention or a mutant DNA thereof,
(2) 非ヒト哺乳動物がゲッ歯動物である第 (1)記載の動物、 (2) the animal according to (1), wherein the non-human mammal is a rodent;
(3) ゲッ歯動物がマウスまたはラットである第 (2) 記載の動物、 および (3) The animal according to (2), wherein the rodent is a mouse or a rat, and
(4) 本発明の外来性 DN Aまたはその変異 DN Aを含有し、 哺乳動物において 発現しうる組換えべクタ一を提供するものである。 (4) It is intended to provide a recombinant vector containing the exogenous DNA of the present invention or its mutant DNA, which can be expressed in mammals.
本発明の外来性 DNAまたはその変異 DNAを有する非ヒト哺乳動物 (以下、 本発明の DNA転移動物と略記する) は、 未受精卵、 受精卵、 精子およびその始 原細胞を含む胚芽細胞などに対して、 好ましくは、 非ヒト哺乳動物の発生におけ る胚発生の段階 (さらに好ましくは、 単細胞または受精卵細胞の段階でかつ一般 に 8細胞期以前) に、 リン酸カルシウム法、 電気パルス法、 リポフエクシヨン法、 凝集法、 マイクロインジェクション法、 パ一ティクルガン法、 DEAE—デキス トラン法などにより目的とする DN Aを転移することによって作出することがで きる。 また、 該 DNA転移方法により、 体細胞、 生体の臓器、 組織細胞などに目 的とする本発明の外来性 DNAを転移し、 細胞培養、 組織培養などに利用するこ ともでき、 さらに、 これら細胞を上述の胚芽細胞と自体公知の細胞融合法により 融合させることにより本発明の D N A転移動物を作出することもできる。 Non-human mammals having the exogenous DNA of the present invention or the mutant DNA thereof (hereinafter abbreviated as the DNA transgenic animal of the present invention) include unfertilized eggs, fertilized eggs, spermatozoa, For germ cells and the like including progenitor cells, calcium phosphate is preferably used at the stage of embryonic development in non-human mammal development (more preferably, at the stage of single cells or fertilized egg cells and generally before the 8-cell stage). It can be produced by transferring the desired DNA by the method, electric pulse method, lipofection method, coagulation method, microinjection method, particle gun method, DEAE-dextran method, etc. Further, by the DNA transfer method, the exogenous DNA of the present invention intended for somatic cells, organs of living organisms, tissue cells, and the like can be transferred and used for cell culture, tissue culture, and the like. Can be fused with the above-mentioned germ cells by a cell fusion method known per se to produce the DNA-transferred animal of the present invention.
非ヒト哺乳動物としては、 例えば、 ゥシ、 ブタ、 ヒッジ、 ャギ、 ゥサギ、 ィヌ、 ネコ、 モルモット、 八ムスター、 マ.,ウス、 ラットなどが用いられる。 なかでも、 病体動物モデル系の作成の面から個体発生および生物サイクルが比較的短く、 ま た、 繁殖が容易なゲッ歯動物、 とりわけマウス (例えば、 純系として、 C 57B LZ6系統, DBA2系統など、 交雑系として、 B6C3F 系統, BDFi系 統, B 6D2F 系統, BALBZc系統, I CR系統など) またはラット (例 えば、 Wi s t a r, SDなど) などが好ましい。  As non-human mammals, for example, porcupines, pigs, higgins, goats, magpies, dogs, cats, guinea pigs, eight-muster, ma., Males, rats and the like are used. Above all, rodents with relatively short ontogeny and biological cycles in terms of the creation of disease animal model systems, and easily breeding rodents, especially mice (for example, C57B LZ6 strains, DBA2 strains as pure strains, etc.) As a cross line, a B6C3F line, a BDFi line, a B6D2F line, a BALBZc line, an ICR line, etc., or a rat (for example, Wistar, SD, etc.) are preferable.
哺乳動物において発現しうる組換えベクターにおける 「哺乳動物」 としては、 上記の非ヒト哺乳動物の他にヒトなどがあげられる。  As the “mammal” in the recombinant vector that can be expressed in mammals, human and the like can be mentioned in addition to the above-mentioned non-human mammals.
本発明の外来性 DNAとは、 非ヒト哺乳動物が本来有している本発明の DNA ではなく、 いったん哺乳 »物から単離'抽出された本発明の DNAをいう。  The exogenous DNA of the present invention refers not to the DNA of the present invention originally possessed by a non-human mammal, but to the DNA of the present invention once isolated and extracted from a mammal.
本発明の変異 DNAとしては、 元の本発明の DNAの塩基配列に変異 (例えば、 突然変異など) が生じたもの、 具体的には、 塩基の付加、 欠損、 他の塩基への置 換などが生じた DNAなどが用いられ、 また、 異常 DNAも含まれる。  As the mutant DNA of the present invention, those having a mutation (for example, mutation) in the base sequence of the original DNA of the present invention, specifically, addition or deletion of a base, substitution with another base, etc. DNA that has been used is used, and also includes abnormal DNA.
該異常 DNAとしては、 異常な本発明のタンパク質を発現させる DNAを意味 し、 例えば、 正常な本発明のタンパク質の機能を抑制するタンパク質を発現させ る D N Aなどが用いられる。  The abnormal DNA means a DNA that expresses an abnormal protein of the present invention, and for example, DNA that expresses a protein that suppresses the function of the normal protein of the present invention is used.
本発明の外来性 D N Aは、 対象とする動物と同種あるいは異種のどちらの哺乳 動物由来のものであってもよい。 本発明の DN Aを対象動物に転移させるにあた つては、 該 DNAを動物細胞で発現させうるプロモーターの下流に結合した DN Aコンストラクトとして用いるのが一般に有利である。 例えば、 本発明のヒト D NAを転移させる場合、 これと相同性が高い本発明の DN Aを有する各種哺乳動 物 (例えば、 ゥサギ、 ィヌ、 ネコ、 モルモット、 ハムスター、 ラット、 マウスな ど) 由来の DNAを発現させうる各種プロモーターの下流に、 本発明のヒト DN Aを結合した DNAコンストラクト (例、 ベクタ一など) を対象哺乳動物の受精 卵、 例えば、 マウス受精卵へマイクロインジェクションすることによって本発明 の DN Aを高発現する DN A転移哺乳動物を作出することができる。 The exogenous DNA of the present invention may be derived from a mammal of the same species or a different species as the target animal. In transferring the DNA of the present invention to a target animal, a DNA linked to a downstream of a promoter capable of expressing the DNA in animal cells is used. It is generally advantageous to use it as an A construct. For example, when the human DNA of the present invention is transferred, various mammals having the DNA of the present invention having a high homology to the human DNA (eg, egret, dog, cat, guinea pig, hamster, rat, mouse, etc.) By microinjecting the DNA construct (eg, vector 1) of the present invention bound to the human DNA into a fertilized egg of a target mammal, for example, a mouse fertilized egg, downstream of various promoters capable of expressing the derived DNA. It is possible to create a DNA transgenic mammal that highly expresses the DNA of the present invention.
本発明のタンパク質の発現ベクターとしては、 大腸菌由来のプラスミド、 枯草 菌由来のプラスミド、 酵母由来のプラスミド、 λファージなどのバクテリオファ ージ、 モロニ一白血病ウィルスなどのレトロウイルス、 ワクシニアウィルスまた はバキュロウィルスなどの動物ウィルスなどが用いられる。 なかでも、 大腸菌由 来のプラスミド、 枯草菌由来のプラスミドまたは酵母由来のプラスミドなどが好 ましく用いられる。  Examples of the expression vector of the protein of the present invention include a plasmid derived from Escherichia coli, a plasmid derived from Bacillus subtilis, a plasmid derived from yeast, a bacteriophage such as λ phage, a retrovirus such as Moroni leukemia virus, a vaccinia virus or a baculovirus. For example, animal viruses such as E. coli are used. Among them, a plasmid derived from Escherichia coli, a plasmid derived from Bacillus subtilis or a plasmid derived from yeast are preferably used.
上記の DNA発現調節を行なうプロモー夕一としては、 例えば、 ①ウィルス (例、 シミアンウィルス、 サイトメガロウィルス、 モロニ一白血病ウィルス、 J Cウィルス、 乳がんウィルス、 ポリオウイルスなど) に由来する DN Aのプロモ 一夕一、 ②各種哺乳動物 (ヒト、 ゥサギ、 ィヌ、 ネコ、 モルモット、 ハムスター、 ラット、 マウスなど) 由来のプロモ一夕一、 例えば、 アルブミン、 インスリン I I、 ゥロプラキン I I、 エラス夕一ゼ、 エリスロポエチン、 エンドセリン、 筋ク レアチンキナーゼ、 グリア線維性酸性タンパク質、 ダル夕チオン S—トランスフ エラ一ゼ、 血小板由来成長因子 /3、 ケラチン Kl, 1^10ぉょび 14、 コラ一 ゲン I型および I I型、 サイクリック AMP依存タンパク質キナーゼ /3 Iサブュ ニット、 ジストロフィン、 酒石酸抵抗性アル力リフォスファターゼ、 心房ナトリ ゥム利尿性因子、 内皮レセプターチ口シンキナーゼ (一般に T i e 2と略され る) 、 ナトリウムカリウムアデノシン 3リン酸化酵素 (Na, K-ATP a s e) 、 ニュ一口フィラメント軽鎖、 メタ口チォネイン Iおよび I IA、 メタロプ ロティナーゼ 1組織インヒピ夕一、 MHCクラス I抗原 (H—2L) 、 H— r a s、 レニン、 ドーパミン /3—水酸化酵素、 甲状腺ペルォキシダーゼ (TPO) 、 ペプチド鎖延長因子 la (EF- 1 α) 、 βァクチン、 ひおよび ]3ミオシン重鎖、 03007926 Examples of promoters that regulate DNA expression include: (1) a promoter of DNA derived from a virus (eg, Simian virus, cytomegalovirus, Moroni leukemia virus, JC virus, breast cancer virus, poliovirus, etc.). Yuichi, ② Promoters derived from various mammals (humans, egrets, dogs, cats, guinea pigs, hamsters, rats, mice, etc.), for example, albumin, insulin II, ゥ roplakin II, Erasuyuze, erythropoietin, Endothelin, muscle creatine kinase, glial fibrillary acidic protein, daryuthion S-transferase, platelet-derived growth factor / 3, keratin Kl, 1 ^ 10 and 14, collagen I and II , Cyclic AMP-dependent protein kinase / 3 I-subunit, dystrophin, Tartrate-resistant alkaline phosphatase, atrial sodium diuretic factor, endothelial receptor ostial synthase (generally abbreviated as Tie 2), sodium potassium adenosine 3 kinase (Na, K-ATPase), Monofilament light chain, metamouth thionine I and I IA, metalloproteinase 1 tissue in situ, MHC class I antigen (H-2L), H-ras, renin, dopamine / 3-hydroxylase, thyroid peroxidase (TPO) , Peptide chain elongation factor la (EF-1α), β-actin, 03007926
50 ミオシン軽鎖 1および 2、 ミエリン基礎タンパク質、 チログロブリン、 Thy— 1、 免疫グロブリン、 H鎖可変部 (VNP) 、 血清アミロイド Pコンポーネント、 ミオグロビン、 トロポニン C、 平滑筋 αァクチン、 プレブ口エンケフアリン Α、 バソプレシンなどのプロモーターなどが用いられる。 なかでも、 全身で高発現す ることが可能なサイトメガロウィルスプロモー夕一、 ヒトペプチド鎖延長因子 1 (EF- 1 ) のプロモーター、 ヒトおよびニヮトリ j3ァクチンプロモータ一 などが好適である。  50 Myosin light chains 1 and 2, myelin basic protein, thyroglobulin, Thy-1, immunoglobulin, heavy chain variable region (VNP), serum amyloid P component, myoglobin, troponin C, smooth muscle α-actin, prebub enkephalin Α, A promoter such as vasopressin is used. Among them, cytomegalovirus promoter, which can be highly expressed in the whole body, human peptide chain elongation factor 1 (EF-1) promoter, human and chicken j3 actin promoter, and the like are preferable.
上記べクタ一は、 DN A転移哺乳動物において目的とするメッセンジャー RN Aの転写を終結する配列 (一般にターミネタ一と呼ばれる) を有していることが 好ましく、 例えば、 ウィルス由来および各種哺乳動物由来の各 DNAの配列を用 いることができ、 好ましくは、 シミアンウィルスの SV40ターミネ夕一などが 用いられる。  The vector preferably has a sequence (generally called terminator) that terminates transcription of the target messenger RNA in the DNA-transferred mammal. For example, it is derived from viruses and various mammals. The sequence of each DNA can be used, and preferably, SV40 terminator of Simian virus or the like is used.
その他、 目的とする外来性 DNAをさらに高発現させる目的で各 DNAのスプ ライシングシグナル、 ェンハンサー領域、 真核 DNAのイントロンの一部などを プロモータ一領域の 5'上流、 プロモー夕一領域と翻訳領域間あるいは翻訳領域 の 3' 下流 に連結することも目的により可能である。  In addition, the splicing signal of each DNA, the enhancer region, a part of the intron of eukaryotic DNA, etc. are used to further express the target exogenous DNA at 5 'upstream of the promoter region, the promoter region and the translation region. It is also possible to ligate between them or at the 3 'downstream of the translation region.
正常な本発明のタンパク質の翻訳領域は、 ヒトまたは各種哺乳動物 (例えば、 -ゥサギ、 ィヌ、 ネコ、 モルモット、 ハムスター、 ラット、 マウスなど) 由来の肝 臓、 腎臓、 甲状腺細胞、 線維芽細胞由来 DNAおよび市販の各種ゲノム DNAラ イブラリーよりゲノム DNAの全てあるいは一部として、 または肝臓、 腎臓、 甲 状腺細胞、 線維芽細胞由来 RNAより公知の方法により調製された相補 DNAを 原料として取得することが出来る。 また、 外来性の異常 DNAは、 上記の細胞ま たは組織より得られた正常な夕ンパク質の翻訳領域を点突然変異誘発法により変 異した翻訳領域を作製することができる。  The normal translation region of the protein of the present invention is derived from a liver, a kidney, a thyroid cell, a fibroblast derived from a human or various mammals (for example,-ゥ egret, dog, cat, guinea pig, hamster, rat, mouse, etc.). All or part of genomic DNA from DNA and various commercially available genomic DNA libraries, or complementary DNA prepared by known methods from liver, kidney, thyroid cell, and fibroblast-derived RNA can be used as raw materials. I can do it. In addition, a foreign abnormal DNA can produce a translation region obtained by mutating a normal protein translation region obtained from the above cells or tissues by a point mutagenesis method.
該翻訳領域は転移動物において発現しうる DNAコンストラクトとして、 前記 のプロモーターの下流および所望により転写終結部位の上流に連結させる通常の D N A工学的手法により作製することができる。  The translation region can be prepared as a DNA construct that can be expressed in a transgenic animal by a conventional DNA engineering technique in which the translation region is ligated downstream of the promoter and, if desired, upstream of the transcription termination site.
受精卵細胞段階における本発明の外来性 DNAの転移は、 対象哺乳動物の胚芽 細胞および体細胞のすべてに存在するように確保される。 DNA転移後の作出動 物の胚芽細胞において、 本発明の外来性 D NAが存在することは、 作出動物の後 代がすべて、 その胚芽細胞および体細胞のすべてに本発明の外来性 D NAを保持 することを意味する。 本発明の外来性 D N Aを受け継いだこの種の動物の子孫は その胚芽細胞および体細胞のすべてに本発明の外来性 D N Aを有する。 Transfer of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal. Production after DNA transfer The presence of the exogenous DNA of the present invention in the germ cell of the product means that all the progeny of the produced animal retain the exogenous DNA of the present invention in all of the germinal and somatic cells . The offspring of such animals that have inherited the exogenous DNA of the present invention have the exogenous DNA of the present invention in all of their germinal and somatic cells.
本発明の外来性正常 D NAを転移させた非ヒト哺乳動物は、 交配により外来性 D NAを安定に保持することを確認して、 該 D N A保有動物として通常の飼育環 境で継代飼育することが出来る。  The non-human mammal to which the exogenous normal DNA of the present invention has been transferred is confirmed to stably maintain the exogenous DNA by mating, and is subcultured as an animal having the DNA in a normal breeding environment. I can do it.
受精卵細胞段階における本発明の外来性 D NAの転移は、 対象哺乳動物の胚芽 細胞および体細胞の全てに過剰に存在するように確保される。 D NA転移後の作 出動物の胚芽細胞において本発明の外来性 D N Aが過剰に存在することは、 作出 動物の子孫が全てその胚芽細胞および体細胞の全てに本発明の外来性 D N Aを過 剰に有することを意味する。 本発明の外来性 D N Aを受け継いだこの種の動物の 子孫はその胚芽細胞および体細胞の全てに本発明の外来性 D N Aを過剰に有する。 導入 D NAを相同染色体の両方に持つホモザィゴ一ト動物を取得し、 この雌雄 の動物を交配することによりすべての子孫が該 D N Aを過剰に有するように繁殖 継代することができる。  Transfer of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in excess in all germ cells and somatic cells of the target mammal. Excessive presence of the exogenous DNA of the present invention in the germinal cells of the animal after transfer of DNA indicates that all of the offspring of the animal produced have the extraneous DNA of the present invention in all of their germinal and somatic cells. To have. The progeny of this type of animal that has inherited the exogenous DNA of the present invention has an excess of the exogenous DNA of the present invention in all of its germ cells and somatic cells. By obtaining homozygous animals having the introduced DNA on both homologous chromosomes and mating the male and female animals, all offspring can be bred and passaged so as to have the DNA in excess.
本発明の正常 D NAを有する非ヒト哺乳動物は、 本発明の正常 D NAが高発現 させられており、 内在性の正常 D N Aの機能を促進することにより最終的に本発 明の夕ンパク質の機能亢進症を発症することがあり、 その病態モデル動物として 利用することができる。 例えば、 本発明の正常 D N A転移動物を用いて、 本発明 のタンパク質の機能亢進症や、 本発明のタンパク質が関連する疾患の病態機序の 解明およびこれらの疾患の治療方法の検討を行なうことが可能である。  The non-human mammal having the normal DNA of the present invention expresses the normal DNA of the present invention at a high level, and finally promotes the function of endogenous normal DNA, thereby finally obtaining the protein of the present invention. May develop hyperfunction, and can be used as a model animal for the disease. For example, using the normal DNA-transferred animal of the present invention, it is possible to elucidate the pathological mechanism of the hyperactivity of the protein of the present invention and diseases associated with the protein of the present invention and to examine a method for treating these diseases. It is possible.
また、 本発明の外来性正常 D NAを転移させた哺乳動物は、 遊離した本発明の タンパク質の増加症状を有することから、 本発明のタンパク質に関連する疾患に 対する予防 ·治療剤、 例えば大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣が ん、 精巣がん、 甲状腺がん、 塍臓がん、 脳腫瘍または血液腫瘍などのがんの予 防 ·治療剤のスクリーニング試験にも利用可能である。  In addition, since the mammal to which the exogenous normal DNA of the present invention has been transferred has an increased symptom of the released protein of the present invention, a prophylactic / therapeutic agent for a disease associated with the protein of the present invention, Cancer, breast cancer, lung cancer, prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid gland It can also be used for screening tests for prophylactic and therapeutic agents for cancer such as cancer, kidney cancer, brain tumor, and blood tumor.
一方、 本発明の外来性異常 D NAを有する非ヒト哺乳動物は、 交配により外来 性 D N Aを安定に保持することを確認して該 D NA保有動物として通常の飼育環 境で継代飼育することが出来る。 さらに、 目的とする外来 D NAを前述のプラス ミドに組み込んで原料として用いることができる。 プロモ一ターとの D NAコン ストラクトは、 通常の D NA工学的手法によって作製することができる。 受精卵 細胞段階における本発明の異常 D N Aの転移は、 対象哺乳動物の胚芽細胞および 体細胞の全てに存在するように確保される。 D N A転移後の作出動物の胚芽細胞 において本発明の異常 D N Aが存在することは、 作出動物の子孫が全てその胚芽 細胞および体細胞の全てに本発明の異常 D N Aを有することを意味する。 本発明 の外来性 D N Aを受け継いだこの種の動物の子孫は、 その胚芽細胞および体細胞 の全てに本発明の異常 D N Aを有する。 導入 D N Aを相同染色体の両方に持つホ モザィゴート動物を取得し、 この雌雄の動物を交配することによりすべての子孫 が該 D N Aを有するように繁殖継代することができる。 On the other hand, the non-human mammal having the foreign abnormal DNA of the present invention After confirming that sex DNA is stably maintained, the animal can be subcultured as a DNA-bearing animal in a normal breeding environment. Furthermore, the desired foreign DNA can be incorporated into the above-mentioned plasmid and used as a raw material. The DNA construct with the promoter can be prepared by ordinary DNA engineering techniques. The transfer of the abnormal DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal. The presence of the abnormal DNA of the present invention in the germinal cells of the produced animal after DNA transfer means that all the offspring of the produced animal have the abnormal DNA of the present invention in all of its germ cells and somatic cells. The progeny of this type of animal that has inherited the exogenous DNA of the present invention has the abnormal DNA of the present invention in all of its germinal and somatic cells. A homozygous animal having the introduced DNA on both homologous chromosomes is obtained, and by crossing these male and female animals, it is possible to breed so that all offspring have the DNA.
本発明の異常 D NAを有する非ヒト哺乳動物は、 本発明の異常 D NAが高発現 させられており、 内在性の正常 D N Aの機能を阻害することにより最終的に本発 明のタンパク質の機能不活性型不応症となることがあり、 その病態モデル動物と して利用することができる。 例えば、 本発明の異常 D NA転移動物を用いて、 本 発明のタンパク質の機能不活性型不応症の病態機序の解明およびこの疾患を治療 方法の検討を行なうことが可能である。  In the non-human mammal having the abnormal DNA of the present invention, the abnormal DNA of the present invention is highly expressed, and the function of the protein of the present invention is ultimately achieved by inhibiting the function of endogenous normal DNA. Inactive refractory disease may occur and can be used as a model animal for the disease. For example, using the abnormal DNA-transferred animal of the present invention, it is possible to elucidate the pathological mechanism of the function-inactive refractory of the protein of the present invention and to examine a method for treating this disease.
また、 具体的な利用可能性としては、 本発明の異常 D N A高発現動物は、 本発 明のタンパク質の機能不活性型不応症における本発明の異常タンパク質による正 常タンパク質の機能阻害 (dominant negat ive作用) を解明するモデルとなる。 また、 本発明の外来異常 D N Aを転移させた哺乳動物は、 遊離した本発明の夕 ンパク質の増加症状を有することから、 本発明のタンパク質または機能不活性型 不応症に対する予防 ·治療剤、 例えば大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮が ん、 卵巣がん、 精巣がん、 甲状腺がん、 膝臓がん、 脳腫瘍または血液腫瘍などの がんの予防 ·治療剤のスクリーニング試験にも利用可能である。  Further, as a specific possibility, the abnormal DNA highly expressing animal of the present invention can be used to inhibit the function of the normal protein by the abnormal protein of the present invention in the function-inactive refractory disease of the protein of the present invention (dominant negative activity). Action). Further, since the mammal to which the foreign abnormal DNA of the present invention has been transferred has an increased symptom of the released protein of the present invention, the agent for preventing or treating the protein of the present invention or a functionally inactive refractory agent, for example, Colorectal, breast, lung, prostate, esophageal, stomach, liver, biliary, spleen, kidney, bladder, uterine, ovarian, testicular, It can also be used for screening tests for prophylactic and therapeutic agents for cancers such as thyroid cancer, knee cancer, brain tumor, and blood tumor.
また、 上記 2種類の本発明の D N A転移動物のその他の利用可能性として、 例 えば、 ①組織培養のための細胞源としての使用、 Further, other possible uses of the above two DNA-transferred animals of the present invention include, for example, ① Use as a cell source for tissue culture,
②本発明の D NA転移動物の組織中の D NAもしくは R N Aを直接分析するか、 または D N Aにより発現されたペプチド組織を分析することによる、 本発明の夕 ンパク質により特異的に発現あるいは活性化するペプチドとの関連性についての 解析、  ②Specific expression or activation by the protein of the present invention by directly analyzing DNA or RNA in the tissue of the DNA-transferred animal of the present invention or by analyzing the peptide tissue expressed by DNA Analysis of the relationship with the peptide
③ D NAを有する組織の細胞を標準組織培養技術により培養し、 これらを使用し て、 一般に培養困難な組織からの細胞の機能の研究、  (3) Cells from tissues having DNA are cultured by standard tissue culture techniques, and these are used to study the function of cells from tissues that are generally difficult to culture.
④上記③記載の細胞を用いることによる細胞の機能を高めるような薬剤のスクリ 一二ング、 および  薬 剤 Screening of drugs that enhance cell function by using the cells described in ③ above, and
⑤本発明の変異タンパク質を単離精製およびその抗体作製などが考えられる。 さらに、 本発明の D NA転移動物を用いて、 本発明のタンパク質の機能不活性 型不応症などを含む、 本発明のタンパク質に関連する疾患の臨床症状を調べるこ とができ、 また、 本発明のタンパク質に関連する疾患モデルの各臓器におけるよ り詳細な病理学的所見が得られ、 新しい治療方法の開発、 さらには、 該疾患によ る二次的疾患の研究および治療に貢献することができる。 単 離 Isolation and purification of the mutant protein of the present invention and production of its antibody can be considered. Furthermore, using the DNA-transferred animal of the present invention, it is possible to examine clinical symptoms of a disease related to the protein of the present invention, including a functionally inactive refractory disease of the protein of the present invention. More detailed pathological findings in each organ of the disease model related to the protein of the present invention can be obtained, which will contribute to the development of new treatment methods and the research and treatment of secondary diseases caused by the disease. it can.
また、 本発明の D NA転移動物から各臓器を取り出し、 細切後、 トリプシンな どのタンパク質分解酵素により、 遊離した D NA転移細胞の取得、 その培養また はその培養細胞の系統化を行なうことが可能である。 さらに、 本発明のタンパク 質産生細胞の特定化、 アポトーシス、 分化あるいは増殖との関連性、 またはそれ らにおけるシグナル伝達機構を調べ、 それらの異常を調べることなどができ、 本 発明のタンパク質およびその作用解明のための有効な研究材料となる。  In addition, each organ is removed from the DNA-transferred animal of the present invention, and after minced, it is possible to obtain free DNA-transferred cells, culture them, or systematize the cultured cells using a protease such as trypsin. It is possible. Furthermore, the protein of the present invention can be identified, its relationship with apoptosis, differentiation or proliferation, or its signal transduction mechanism can be examined, and its abnormality can be examined. It is an effective research material for elucidation.
さらに、 本発明の D N A転移動物を用いて、 本発明のタンパク質の機能不活性 型不応症を含む、 本発明のタンパク質に関連する疾患の治療薬の開発を行なうた めに、 上述の検査法および定量法などを用いて、 有効で迅速な該疾患治療薬のス クリーニング法を提供することが可能となる。 また、 本発明の D N A転移動物ま たは本発明の外来性 D N A発現べクターを用いて、 本発明のタンパク質が関連す る疾患の D N A治療法を検討、 開発することが可能である。  Furthermore, in order to use the DNA transgenic animal of the present invention to develop a therapeutic agent for a disease associated with the protein of the present invention, including a refractory inactive type of the protein of the present invention, Using a quantitative method or the like, it is possible to provide an effective and rapid screening method for the therapeutic agent for the disease. Also, using the DNA transgenic animal of the present invention or the exogenous DNA expression vector of the present invention, it is possible to examine and develop a method for treating a DNA associated with the protein of the present invention.
( 8 ) ノックアウト動物 本発明は、 本発明の DN Aが不活性化された非ヒト哺乳動物胚幹細胞および本 発明の DNA発現不全非ヒト哺乳動物を提供する。 (8) Knockout animal The present invention provides a non-human mammalian embryonic stem cell in which the DNA of the present invention is inactivated and a non-human mammal deficient in expression of the DNA of the present invention.
すなわち、 本発明は、  That is, the present invention
( 1 ) 本発明の D N Aが不活性化された非ヒト哺乳動物胚幹細胞、  (1) a non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated,
(2) 該 DNAがレポ一夕一遺伝子 (例、 大腸菌由来の ]3—ガラクトシダ一ゼ遺 伝子) を導入することにより不活性化された第 (1) 項記載の胚幹細胞、  (2) The embryonic stem cell according to (1), wherein the DNA is inactivated by introducing a repo overnight gene (eg, a 3-galactosidase enzyme gene derived from Escherichia coli).
(3) ネオマイシン耐性である第 (1) 項記載の胚幹細胞、  (3) The embryonic stem cell according to (1), which is neomycin-resistant,
(4) 非ヒト哺乳動物がゲッ歯動物である第 (1) 項記載の胚幹細胞、  (4) the embryonic stem cell according to (1), wherein the non-human mammal is a rodent;
(5) ゲッ歯動物がマウスである第 (4) 項記載の胚幹細胞、  (5) The embryonic stem cell according to (4), wherein the rodent is a mouse,
(6) 本発明の DNAが不活性化された該 DNA発現不全非ヒト哺乳動物、 (6) a DNA-deficient non-human mammal in which the DNA of the present invention has been inactivated,
(7) 該 DNAがレポーター遺伝子 (例、 大腸菌由来の /3—ガラクトシダ一ゼ遺 伝子) を導入することにより不活性化され、 該レポ一夕一遺伝子が本発明の DN Aに対するプロモータ一の制御下で発現しうる第 (6) 項記載の非ヒト哺乳動物、(7) The DNA is inactivated by introducing a reporter gene (eg, a / 3-galactosidase enzyme gene derived from Escherichia coli), and the repo allele gene is a promoter of the promoter of the present invention. The non-human mammal according to (6), which can be expressed under control,
(8) 非ヒト哺乳動物がゲッ歯動物である第 (6) 項記載の非ヒト哺乳動物、 (9) ゲッ歯動物がマウスである第 (8) 項記載の非ヒト哺乳動物、 および(8) The non-human mammal according to (6), wherein the non-human mammal is a rodent, (9) the non-human mammal according to (8), wherein the rodent is a mouse, and
(10) 第 (7) 項記載の動物に、 試験化合物を投与し、 レポーター遺伝子の発 現を検出することを特徴とする本発明の DNAに対するプロモータ一活性を促進 または阻害する化合物またはその塩のスクリーニング方法を提供する。 (10) A test compound is administered to the animal described in (7), and the expression of a reporter gene is detected. A screening method is provided.
本発明の DNAが不活性化された非ヒト哺乳動物胚幹細胞とは、 該非ヒト哺乳 動物が有する本発明の DNAに人為的に変異を加えることにより、 DNAの発現 能を抑制するか、 もしくは該 D N Aがコ一ドしている本発明のタンパク質の活性 を実質的に喪失させることにより、 D N Aが実質的に本発明のタンパク質の発現 能を有さない (以下、 本発明のノックアウト DNAと称することがある) 非ヒト 哺乳動物の胚幹細胞 (以下、 ES細胞と略記する) をいう。  A non-human mammalian embryonic stem cell in which the DNA of the present invention has been inactivated is an artificially mutated DNA of the present invention possessed by the non-human mammal, which suppresses the expression ability of the DNA, or By substantially losing the activity of the protein of the present invention encoded by the DNA, the DNA substantially does not have the ability to express the protein of the present invention (hereinafter referred to as the knockout DNA of the present invention). Non-human mammalian embryonic stem cells (hereinafter abbreviated as ES cells).
非ヒト哺乳動物としては、 前記と同様のものが用いられる。  As the non-human mammal, the same one as described above is used.
本発明の DN Aに人為的に変異を加える方法としては、 例えば、 遺伝子工学的 手法により該 DN A配列の一部又は全部の削除、 他 DNAを揷入または置換させ ることによって行なうことができる。 これらの変異により、 例えば、 コドンの読 み取り枠をずらしたり、 プロモー夕一あるいはェキソンの機能を破壊することに より本発明のノックアウト DNAを作製すればよい。 The method of artificially mutating the DNA of the present invention can be carried out, for example, by deleting a part or all of the DNA sequence or inserting or substituting another DNA by a genetic engineering technique. . These mutations can, for example, shift the reading frame of codons or disrupt the function of Promote or Exon. What is necessary is just to prepare the knockout DNA of the present invention.
本発明の DNAが不活性化された非ヒト哺乳動物胚幹細胞 (以下、 本発明の D N A不活性化 E S細胞または本発明のノックァゥト E S細胞と略記する) の具体 例としては、 例えば、 目的とする非ヒト哺乳動物が有する本発明の DNAを単離 し、 そのェキソン部分にネオマイシン耐性遺伝子、 ハイグロマイシン耐性遺伝子 を代表とする薬剤耐性遺伝子、 あるいは 1 a c Z (j3—ガラクトシダ一ゼ遺伝 子) 、 c a t (クロラムフエニコ一ルァセチルトランスフェラ一ゼ遗伝子) を代 表とするレポーター遺伝子等を挿入することによりェキソンの機能を破壊するか、 あるいはェキソン間のイントロン部分に遺伝子の転写を終結させる DNA配列 (例えば、 p o 1 yA付加シグナルなど) を揷入し、 完全なメッセンジャ一 RN Aを合成できなくすることによって、 結果的に遺伝子を破壊するように構築した DNA配列を有する DNA鎖 (以下、 夕一ゲッティングベクタ一と略記する) を、 例えば相同組換え法により該動物の染色体に導入し、 得られた ES細胞について 本発明の D N A上あるいはその近傍の D N A配列をプローブとしたサザンハイブ リダィゼーション解析あるいはターゲッティングベクター上の DNA配列とタ一 ゲッティングベクタ一作製に使用した本発明の D N A以外の近傍領域の D N A配 列をプライマーとした P CR法により解析し、 本発明のノックアウト E S細胞を 選別することにより得ることができる。  Specific examples of the non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated (hereinafter abbreviated as the DNA-inactivated ES cells of the present invention or the knockout ES cells of the present invention) include, for example, The DNA of the present invention possessed by a non-human mammal is isolated, and its exon portion is a drug resistance gene typified by a neomycin resistance gene, a hygromycin resistance gene, or 1acZ (j3-galactosidase gene), cat Inserting a reporter gene or the like represented by (chloramphenicylacetyltransferase gene) disrupts exon function, or terminates transcription of the gene in the intron portion between exons. For example, by introducing a po 1 yA additional signal) and preventing the synthesis of a complete messenger RNA, the resulting A DNA chain having a DNA sequence constructed so as to disrupt the gene (hereinafter, abbreviated as “gathering vector 1”) is introduced into the chromosome of the animal by, for example, a homologous recombination method. A DNA sequence on or near the DNA of the present invention used for Southern hybridization analysis or a targeting vector and a DNA sequence on a targeting vector other than the DNA of the present invention used for preparing a targeting vector Can be obtained by analyzing by the PCR method using as primers and selecting the knockout ES cells of the present invention.
また、 相同組換え法等により本発明の DNAを不活化させる元の ES細胞とし ては、 例えば、 前述のような既に樹立されたものを用いてもよく、 また公知 As the original ES cells for inactivating the DNA of the present invention by the homologous recombination method or the like, for example, those already established as described above may be used.
Evansと Kaufmaの方法に準じて新しく樹立したものでもよい。 例えば、 マウスの ES細胞の場合、 現在、 一般的には 129系の ES細胞が使用されているが、 免 疫学的背景がはつきりしていないので、 これに代わる純系で免疫学的に遺伝的背 景が明らかな ES細胞を取得するなどの目的で例えば、 C57BL/6マウスや C 57 BLZ6の採卵数の少なさを DBAZ2との交雑により改善した BDFi マウス (C 57 BL/6と DBAZ2との を用いて樹立したものなども良 好に用いうる。 BDFiマウスは、 採卵数が多く、 かつ、 卵が丈夫であるという 利点に加えて、' C 57 BL/6マウスを背景に持つので、 これを用いて得られた ES細胞は病態モデルマウスを作出したとき、 C 57 ロスすることでその遺伝的背景を C 5 7 B L / 6マウスに代えることが可能であ る点で有利に用い得る。 It may be newly established according to the method of Evans and Kaufma. For example, in the case of mouse ES cells, currently, 129 ES cells are generally used, but since their immunological background has not been fixed, a pure line that substitutes them has been used for immunological inheritance. For example, for the purpose of obtaining ES cells with a clear background, BDFi mice (C57BL / 6 and DBAZ2 and C57BL / 6 and DBAZ2) BDFi mice have the advantage of high number of eggs collected and robust eggs, and have the background of 'C57BL / 6 mice, The ES cells obtained using this were used to generate C57 It can be used to advantage in that loss can replace its genetic background with C57BL / 6 mice.
また、 E S細胞を樹立する場合、 一般には受精後 3 . 5日目の胚盤胞を使用す るが、 これ以外に 8細胞期胚を採卵し胚盤胞まで培養して用いることにより効率 よく多数の初期胚を取得することができる。  In addition, when ES cells are established, blastocysts 3.5 days after fertilization are generally used.However, it is more efficient to collect 8-cell embryos and culture them up to blastocysts. Many early embryos can be obtained.
また、 雌雄いずれの E S細胞を用いてもよいが、 通常雄の E S細胞の方が生殖 系列キメラを作出するのに都合が良い。 また、 煩雑な培養の手間を削減するため にもできるだけ早く雌雄の判別を行なうことが望ましい。  Either male or female ES cells may be used, but male ES cells are generally more convenient for producing a germline chimera. It is also desirable to discriminate between males and females as soon as possible in order to reduce the complexity of culturing.
E S細胞の雌雄の判定方法としては、 例えば、 P C R法により Y染色体上の性 決定領域の遺伝子を増幅、 検出する方法が、 その 1例としてあげることができる。 この方法を使用すれば、 従来、 核型分析をするのに約 1 0 6個の細胞数を要して いたのに対して、 1コロニー程度の E S細胞数 (約 5 0個) で済むので、 培養初 期における E S細胞の第一次セレクションを雌雄の判別で行なうことが可能であ り、 早期に雄細胞の選定を可能にしたことにより培養初期の手間は大幅に削減で きる。 As an example of a method for determining the sex of ES cells, a method of amplifying and detecting a gene in the sex-determining region on the Y chromosome by PCR can be given as an example. Using this method, conventionally, for example G-banding method, it requires about 1 0 6 cells for karyotype analysis, since suffices ES cell number of about 1 colony (about 5 0) However, the primary selection of ES cells at the initial stage of culture can be performed by gender discrimination, and the early selection of male cells can greatly reduce the labor required at the initial stage of culture.
また、 第二次セレクションとしては、 例えば、 G—バンデイング法による染色 体数の確認等により行うことができる。 得られる E S細胞の染色体数は正常数の 1 0 0 %が望ましいが、 樹立の際の物理的操作等の関係上困難な場合は、 E S細 胞の遺伝子をノックアウトした後、 正常細胞 (例えば、 マウスでは染色体数が 2 n = 4 0である細胞) に再びクローニングすることが望ましい。  The secondary selection can be performed, for example, by confirming the number of chromosomes by the G-banding method. The number of chromosomes in the obtained ES cells is preferably 100% of the normal number. However, if it is difficult due to physical operations at the time of establishment, knock out the gene of the ES cells, and then use normal cells (for example, It is desirable to clone again into the mouse (cells with 2 n = 40 chromosomes).
このようにして得られた胚幹細胞株は、 通常その増殖性は大変良いが、 個体発 生できる能力を失いやすいので、 注意深く継代培養することが必要である。 例え ば、 S T O繊維芽細胞のような適当なフィーダ一細胞上で LIF (l〜10000U/ml) 存在下に炭酸ガス培養器内 (好ましくは、 5 %炭酸ガス、 95%空気または 5%酸素、 5 %炭酸ガス、 90%空気) で約 37°Cで培養するなどの方法で培養し、 継代時には、 例えば、 トリプシン/ EDTA溶液 (通常 0. 001〜0. 5%トリプシン/ 0. l〜5mM EDTA、 好ましくは約 0. 1 %トリフ°シン/ lmM EDTA) 処理に.より単細胞化し、 新たに用意し たフィーダ一細胞上に播種する方法などがとられる。 このような継代は、 通常 1 〜3日毎に行なうが、 この際に細胞の観察を行い、 形態的に異常な細胞が見受け られた場合はその培養細胞は放棄することが望まれる。 Embryonic stem cell lines obtained in this way usually have very good proliferative properties, but they must be carefully subcultured because they tend to lose their ability to generate individuals. For example, on a suitable feeder cell, such as STO fibroblasts, in a carbon dioxide incubator (preferably 5% carbon dioxide, 95% air or 5% oxygen, in the presence of LIF (1 to 10,000 U / ml)) Culture at about 37 ° C with 5% carbon dioxide gas and 90% air. At the time of subculture, for example, trypsin / EDTA solution (usually 0.001 to 0.5% trypsin / 0.1 l A 5 mM EDTA (preferably, about 0.1% tris / L M EDTA) treatment may be used to form a single cell and inoculate it on a newly prepared feeder cell. Such subculture is usually performed every 1 to 3 days. At this time, cells are observed, and morphologically abnormal cells are found. If so, the cultured cells should be discarded.
E S細胞は、 適当な条件により、 高密度に至るまで単層培養するか、 または細 胞集塊を形成するまで浮遊培養することにより、 頭頂筋、 内臓筋、 心筋などの 種々のタイプの細胞に分化させることが可能であり 〔M. 】. Evans及び M. H.  ES cells can be transformed into various types of cells, such as parietal, visceral, and cardiac muscle, by monolayer culture up to high density or suspension culture until cell clumps are formed under appropriate conditions. It is possible to differentiate [M.]. Evans and MH
Kaufman, Nature 第 292巻、 154頁、 1981年; G. R. Mart in, Proc. Nat l . Acad. Sc i . U. S. A.第 78卷、 7634頁、 1981年; T. C. Doetschman ら、 ジャーナル'ォ ブ ·ェンブリオロジ一 ·アンド ·ェクスペリメンタル ·モルフォロジ一、 第 87巻、 27頁、 1985年〕 、 本発明の E S細胞を分化させて得られる本発明の D NA発現不 全細胞は、 ィンビトロにおける本発明のタンパク質の細胞生物学的検討において 有用である。 Kaufman, Nature 292, 154, 1981; GR Mart in, Proc. Natl. Acad. Sci. USA 78, 7634, 1981; TC Doetschman et al., Journal of Ob. And experimental morphology, Vol. 87, p. 27, 1985), and the DNA-incompletely expressed cells of the present invention obtained by differentiating the ES cells of the present invention Useful in cell biology studies.
本発明の D N A発現不全非ヒト哺乳動物は、 該動物の mR N A量を公知方法を 用いて測定して間接的にその発現量を比較することにより、 正常動物と区別する ことが可能である。  The non-human mammal deficient in DNA expression of the present invention can be distinguished from a normal animal by measuring the mRNA level of the animal using a known method and indirectly comparing the expression level.
該非ヒト哺乳動物としては、 前記と同様のものが用いられる。  As the non-human mammal, those similar to the aforementioned can be used.
本発明の D NA発現不全非ヒト哺乳動物は、 例えば、 前述のようにして作製し た夕一ゲッティングベクターをマウス胚幹細胞またはマウス卵細胞に導入し、 導 入により夕ーゲッティングベクタ一の本発明の D N Aが不活性化された D N A配 列が遺伝子相同組換えにより、 マウス胚幹細胞またはマウス卵細胞の染色体上の 本発明の D N Aと入れ換わる相同組換えをさせることにより、 本発明の D NAを ノックアウトさせることができる。  The non-human mammal deficient in DNA expression of the present invention can be obtained, for example, by introducing the evening-getting vector prepared as described above into a mouse embryonic stem cell or a mouse egg cell and introducing the evening-getting vector into a non-human mammal. The DNA of the present invention is inactivated by homologous recombination to replace the DNA of the present invention on the chromosome of mouse embryonic stem cells or mouse egg cells by gene homologous recombination, thereby obtaining the DNA of the present invention. Can be knocked out.
本発明の D NAがノックアウトされた細胞は、 本発明の D NA上またはその近 傍の D N A配列をプローブとしたサザンハイプリダイゼーション解析または夕一 ゲッティングベクター上の D NA配列と、 夕ーゲッティングベクターに使用した マウス由来の本発明の D N A以外の近傍領域の D N A配列とをプライマーとした P C R法による解析で判定することができる。 非ヒト哺乳動物胚幹細胞を用いた 場合は、 遺伝子相同組換えにより、 本発明の D NAが不活性化された細胞株をク ローニングし、 その細胞を適当な時期、 例えば、 8細胞期の非ヒト哺乳動物胚ま たは胚盤胞に注入し、 作製したキメラ胚を偽妊娠させた該非ヒト哺乳動物の子宮 に移植する。 作出された動物は正常な本発明の D N A座をもつ細胞と人為的に変 異した本発明の D N A座をもつ細胞との両者から構成されるキメラ動物である。 該キメラ動物の生殖細胞の一部が変異した本発明の D NA座をもつ場合、 この ようなキメラ個体と正常個体を交配することにより得られた個体群より、 全ての 組織が人為的に変異を加えた本発明の D N A座をもつ細胞で構成された個体を、 例えば、 コートカラーの判定等により選別することにより得られる。 このように して得られた個体は、 通常、 本発明のタンパク質のヘテロ発現不全個体であり、 本発明のタンパク質のヘテロ発現不全個体同志を交配し、 それらの産仔から本発 明のタンパク質のホモ発現不全個体を得ることができる。 Cells in which the DNA of the present invention has been knocked out can be analyzed by Southern hybridization analysis or DNA analysis using a DNA sequence on or near the DNA of the present invention as a probe. It can be determined by analysis by PCR using, as a primer, the DNA sequence of a neighboring region other than the DNA of the present invention derived from the mouse used for the targeting vector. When a non-human mammalian embryonic stem cell is used, the cell line in which the DNA of the present invention has been inactivated is cloned by homologous gene recombination, and the cell line is cloned at an appropriate time, for example, at the 8-cell stage. The chimeric embryo is injected into a human mammalian embryo or blastocyst, and the resulting chimeric embryo is transplanted into the uterus of the pseudo-pregnant non-human mammal. The resulting animal will be artificially altered from cells having a normal DNA locus of the present invention. It is a chimeric animal composed of both cells having different DNA loci of the present invention. When a part of the germ cells of the chimeric animal has the mutated DNA locus of the present invention, all tissues are artificially mutated from a population obtained by crossing such a chimeric individual with a normal individual. Can be obtained by, for example, selecting an individual composed of cells having the DNA locus of the present invention to which the DNA is added by, for example, determining the coat color. The individuals obtained in this manner are usually individuals deficient in the hetero-expression of the protein of the present invention, and mated with individuals deficient in the hetero-expression of the protein of the present invention. A homozygous deficient individual can be obtained.
卵細胞を使用する場合は、 例えば、 卵細胞核内にマイクロインジェクション法 で D N A溶液を注入することにより夕一ゲッティングベクターを染色体内に導入 したトランスジエニック非ヒト哺乳動物を得ることができ、 これらのトランスジ エニック非ヒト哺乳動物に比べて、 遺伝子相同組換えにより本発明の D N A座に 変異のあるものを選択することにより得られる。  In the case of using an egg cell, for example, a transgenic non-human mammal having a chromosome into which a gettering vector has been introduced can be obtained by injecting a DNA solution into a nucleus of an egg by a microinjection method. Compared to a transgenic non-human mammal, it can be obtained by selecting a gene having a mutation in the DNA locus of the present invention by homologous recombination.
このようにして本発明の D NAがノックアウトされている個体は、 交配により 得られた動物個体も該 D NAがノックアウトされていることを確認して通常の飼 育環境で飼育継代を行なうことができる。  In the individual knocked out of the DNA of the present invention in this way, the animal individuals obtained by mating should also be confirmed to be knocked out of the DNA, and reared in a normal breeding environment. Can be.
さらに、 生殖系列の取得および保持についても常法に従えばよい。 すなわち、 該不活化 D N Aの保有する雌雄の動物を交配することにより、 該不活化 D N Aを 相同染色体の両方に持つホモザィゴート動物を取得しうる。 得られたホモザィゴ —ト動物は、 母親動物に対して、 正常個体 1, ホモザィゴ一ト複数になるような 状態で飼育することにより効率的に得ることができる。 ヘテロザィゴ一ト動物の 雌雄を交配することにより、 該不活化 D N Aを有するホモザィゴートおよびへテ ロザィゴート動物を繁殖継代する。  Furthermore, the germline can be obtained and maintained according to a standard method. That is, by mating male and female animals having the inactivated DNA, homozygous animals having the inactivated DNA on both homologous chromosomes can be obtained. The obtained homozygous animal can be efficiently obtained by rearing the mother animal in such a manner that one normal individual and plural homozygotes are obtained. By mating male and female heterozygous animals, homozygous and heterozygous animals having the inactivated DNA are bred and passaged.
本発明の D N Aが不活性化された非ヒト哺乳動物胚幹細胞は、 本発明の D NA 発現不全非ヒト哺乳動物を作出する上で、 非常に有用である。  The non-human mammalian embryonic stem cells in which the DNA of the present invention has been inactivated are extremely useful for producing the non-human mammal deficient in expression of the DNA of the present invention.
また、 本発明の D NA発現不全非ヒト哺乳動物は、 本発明のタンパク質により 誘導され得る種々の生物活性を欠失するため、 本発明のタンパク質の生物活性の 不活性化を原因とする疾病のモデルとなり得るので、 これらの疾病の原因究明及 び治療法の検討に有用である。 ( 8 a ) 本発明の D NAの欠損や損傷などに起因する疾病に対して治療 ·予防効 果を有する化合物のスクリーニング方法 In addition, since the non-human mammal deficient in expression of the DNA of the present invention lacks various biological activities that can be induced by the protein of the present invention, a disease caused by inactivation of the biological activity of the protein of the present invention. Since it can be a model, it is useful for investigating the causes of these diseases and examining treatment methods. (8a) A method for screening a compound having a therapeutic / preventive effect on diseases caused by DNA deficiency or damage, etc. of the present invention
本発明の D NA発現不全非ヒト哺乳動物は、 本発明の D NAの欠損や損傷など に起因する疾病に対して治療 ·予防効果を有する化合物のスクリーニングに用い ることができる。  The non-human mammal deficient in expression of the DNA of the present invention can be used for screening for a compound having a therapeutic / preventive effect against diseases caused by the deficiency or damage of the DNA of the present invention.
すなわち、 本発明は、 本発明の D NA発現不全非ヒト哺乳動物に試験化合物を 投与し、 該動物の変化を観察 ·測定することを特徴とする、 本発明の D NAの欠 損や損傷などに起因する疾病、 例えばがんなどに対して治療 ·予防効果を有する 化合物またはその塩のスクリーニング方法を提供する。  That is, the present invention comprises administering a test compound to a non-human mammal deficient in expression of the DNA of the present invention, and observing and measuring changes in the animal. To provide a method for screening a compound or a salt thereof having a therapeutic / preventive effect against diseases caused by, for example, cancer.
該スクリーニング方法において用いられる本発明の D NA発現不全非ヒト哺乳 動物としては、 前記と同様のものがあげられる。  The non-human mammal deficient in DNA expression of the present invention used in the screening method includes the same ones as described above.
試験化合物としては、 例えば、 ペプチド、 タンパク質、 非ペプチド性化合物、 合成化合物、 発酵生産物、 細胞抽出液、 植物抽出液、 動物組織抽出液、 血漿など があげられ、 これら化合物は新規な化合物であってもよいし、 公知の化合物であ つてもよい。  Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and plasma, and these compounds are novel compounds. Or a known compound.
具体的には、 本発明の D N A発現不全非ヒト哺乳動物を、 試験化合物で処理し、 無処理の対照動物と比較し、 該動物の各器官、 組織、 疾病の症状などの変化を指 標として試験化合物の治療 ·予防効果を試験することができる。  Specifically, a non-human mammal deficient in expression of the DNA of the present invention is treated with a test compound, compared with a non-treated control animal, and changes in the organs, tissues, disease symptoms, etc. of the animal are used as indicators. The test compound can be tested for its therapeutic and prophylactic effects.
試験動物を試験化合物で処理する方法としては、 例えば、 経口投与、 静脈注射 などが用いられ、 試験動物の症状、 試験化合物の性質などにあわせて適宜選択す ることができる。 また、 試験化合物の投与量は、 投与方法、 試験化合物の性質な どにあわせて適宜選択することができる。  As a method for treating a test animal with a test compound, for example, oral administration, intravenous injection and the like are used, and it can be appropriately selected according to the symptoms of the test animal, the properties of the test compound, and the like. The dose of the test compound can be appropriately selected according to the administration method, the properties of the test compound, and the like.
例えば大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣がん、 甲状 腺がん、 塍臓がん、 脳腫瘍または血液腫瘍などのがんに対して治療 ·予防効果を 有する化合物をスクリーニングする場合、 本発明の D N A発現不全非ヒト哺乳動 物に試験化合物を投与し、 試験化合物非投与群とがんの発症度合いの違いやがん の治癒度合いの違いを上記組織で経時的に観察する。  For example, colon, breast, lung, prostate, esophagus, stomach, liver, biliary tract, spleen, kidney, bladder, uterus, ovary, testis When screening for a compound having a therapeutic or preventive effect against cancers such as thyroid cancer, kidney cancer, brain tumor, or blood tumor, a test compound is administered to a non-human mammal deficient in DNA expression of the present invention. Then, the difference in the degree of onset of cancer and the degree of cure of cancer in the non-administered test compound group are observed over time in the above tissues.
該スクリーニング方法において、 試験動物に試験化合物を投与した場合、 該試 験動物の上記疾患症状が約 1 0 %以上、 好ましくは約 3 0 %以上、 より好ましく は約 5 0 %以上改善した場合、 該試験化合物を上記の疾患に対して治療 ·予防効 果を有する化合物として選択することができる。 In the screening method, when a test compound is administered to a test animal, When the disease symptom of the test animal is improved by about 10% or more, preferably about 30% or more, more preferably about 50% or more, the test compound has a therapeutic / preventive effect on the above-mentioned diseases. It can be selected as a compound.
該スクリーニング方法を用いて得られる化合物は、 上記した試験化合物から選 ばれた化合物であり、 本発明のタンパク質の欠損や損傷などによって引き起こさ れる疾患に対して治療 ·予防効果を有するので、 該疾患に対する安全で低毒性な 予防'治療剤などの医薬として使用することができる。 さらに、 上記スクリ一二 ングで得られた化合物から誘導される化合物も同様に用いることができる。  The compound obtained by using the screening method is a compound selected from the test compounds described above, and has a therapeutic / preventive effect on a disease caused by a deficiency or damage of the protein of the present invention. It can be used as a medicament such as a safe and low toxic prophylactic or therapeutic agent. Further, a compound derived from the compound obtained by the above screening can also be used.
該スクリ一ニング方法で得られた化合物は塩を形成していてもよく、 該化合物 の塩としては、 生理学的に許容される酸 (例、 無機酸、 有機酸など) や塩基 (例、 アルカリ金属など) などとの塩が用いられ、 とりわけ生理学的に許容される酸付 加塩が好ましい。 この様な塩としては、 例えば、 無機酸 (例えば、 塩酸、 リン酸、 臭化水素酸、 硫酸など) との塩、 あるいは有機酸 (例えば、 酢酸、 ギ酸、 プロピ オン酸、 フマル酸、 マレイン酸、 コハク酸、 酒石酸、 クェン酸、 リンゴ酸、 篠酸、 安息香酸、 メタンスルホン酸、 ベンゼンスルホン酸など) との塩などが用いられ る。  The compound obtained by the screening method may form a salt. Examples of the salt of the compound include physiologically acceptable acids (eg, inorganic acids, organic acids, etc.) and bases (eg, alkalis). Salts with metals and the like are used, and especially preferred are physiologically acceptable acid addition salts. Examples of such salts include salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.), and organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid) , Succinic acid, tartaric acid, citric acid, malic acid, succinic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc.).
該スクリーニング方法で得られた化合物またはその塩を含有する医薬は、 前記 した本発明のタンパク質を含有する医薬と同様にして製造することができる。 このようにして得られる製剤は、 安全で低毒性であるので、 例えば、 ヒトまた は哺乳動物 (例えば、 ラット、 マウス、 モルモット、 ゥサギ、 ヒッジ、 ブ夕、 ゥ シ、 ゥマ、 ネコ、 ィヌ、 サルなど) に対して投与することができる。  A drug containing the compound or a salt thereof obtained by the screening method can be produced in the same manner as the drug containing the protein of the present invention described above. The preparations obtained in this way are safe and low toxic and can be used, for example, in humans or mammals (eg, rats, mice, guinea pigs, egrets, sheep, sheep, bushus, dogs, dogs, cats, dogs). , Monkeys, etc.).
該化合物またはその塩の投与量は、 対象疾患、 投与対象、 投与ルートなどによ り差異はあるが、 例えば、 該化合物を経口投与する場合、 一般的に成人 (体重 6 0 k gとして) の乳がん患者においては、 一日につき該化合物を約 0. 1〜1 0 O m g、 好ましくは約 1 . 0〜5 O m g、 より好ましくは約 1 . 0〜2 0 mg投 与する。 非経口的に投与する場合は、 該化合物の 1回投与量は投与対象、 対象疾 患などによっても異なるが、 例えば、 該化合物を注射剤の形で通常成人 (体重 6 O k gとして) の乳がん患者に投与する場合、 一日につき該化合物を約 0 . 0 1 〜 3 O m g程度、 好ましくは約 0 . l〜2 0 m g程度、 より好ましくは約 0 . 1 〜1 O m g程度を静脈注射により投与するのが好都合である。 他の動物の場合も、 体重 6 0 k g当たりに換算した量を投与することができる。 The dose of the compound or a salt thereof varies depending on the target disease, the administration subject, the administration route, and the like. For example, when the compound is orally administered, generally, breast cancer of an adult (assuming a body weight of 60 kg) is used. In a patient, about 0.1 to 10 Omg, preferably about 1.0 to 5 Omg, more preferably about 1.0 to 20 mg of the compound is administered per day. In the case of parenteral administration, the single dose of the compound varies depending on the administration subject, the target disease and the like. For example, the compound is usually in the form of an injection and usually used for adult (with a body weight of 6 O kg) breast cancer. When administered to a patient, the compound is administered in a daily dose of about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 mg. It is convenient to administer about 1 to 10 mg by intravenous injection. In the case of other animals, the dose can be administered in terms of weight per 60 kg.
( 8 b ) 本発明の D NAに対するプロモ一夕一の活性を促進または阻害する化合 物をスクリーニング方法  (8b) A method for screening for a compound that promotes or inhibits the overnight activity of the promoter for DNA of the present invention
本発明は、 本発明の D NA発現不全非ヒト哺乳動物に、 試験化合物を投与し、 レポーター遺伝子の発現を検出することを特徴とする本発明の D NAに対するプ 口モーターの活性を促進または阻害する化合物またはその塩のスクリ—エング方 法を提供する。  The present invention provides a test compound administered to a non-human mammal deficient in expression of a DNA of the present invention, and detects the expression of a reporter gene. A method for screening a compound or a salt thereof.
上記スクリーニング方法において、 本発明の D N A発現不全非ヒト哺乳動物と しては、 前記した本発明の D NA発現不全非ヒト哺乳動物の中でも、 本発明の D N Aがレポーター遺伝子を導入することにより不活性化され、 該レポ一夕一遺伝 子が本発明の D N Aに対するプ口モータ一の制御下で発現しうるものが用いられ る。  In the above-described screening method, the non-human mammal deficient in expression of DNA of the present invention may be the non-human mammal deficient in expression of DNA of the present invention in which the DNA of the present invention is inactive by introducing a reporter gene. A gene which can be expressed under the control of a motor for the DNA of the present invention is used.
試験化合物としては、 前記と同様のものがあげられる。  Examples of the test compound include the same compounds as described above.
レポ一夕一遺伝子としては、 前記と同様のものが用いられ、 /3—ガラクトシダ ーゼ遺伝子 ( 1 a c Z ) 、 可溶性アルカリフォスファタ一ゼ遺伝子またはルシフ ェラーゼ遺伝子などが好適である。  As the repo overnight gene, the same gene as described above is used, and a / 3-galactosidase gene (1 acZ), a soluble alkaline phosphatase gene or a luciferase gene is preferable.
本発明の D N Aをレポ一夕一遺伝子で置換された本発明の D N A発現不全非ヒ ト哺乳動物では、 レポ一夕一遺伝子が本発明の D N Aに対するプロモーターの支 配下に存在するので、 レポ一夕一遺伝子がコードする物質の発現をトレースする ことにより、 プロモーターの活性を検出することができる。  In a non-human mammal deficient in expression of the DNA of the present invention in which the DNA of the present invention has been replaced with the repo overnight gene, the repo overnight gene is under the control of the promoter for the DNA of the present invention. By tracing the expression of a substance encoded by one gene, the activity of the promoter can be detected.
例えば、 本発明のタンパク質をコードする D N A領域の一部を大腸菌由来の )3 一ガラクトシダーゼ遺伝子 (1 a c Z ) で置換している場合、 本来、 本発明の夕 ンパク質の発現する組織で、 本発明のタンパク質の代わりに ;3—ガラクトシダ一 ゼが発現する。 従って、 例えば、 5—プロモー 4—クロロー 3—インドリル一 ;8 —ガラクトビラノシド (X— g a l ) のような 0—ガラクトシダ一ゼの基質とな る試薬を用いて染色することにより、 簡便に本発明のタンパク質の動物生体内に おける発現状態を観察することができる。 具体的には、 本発明のタンパク質欠損 マウスまたはその組織切片をダルタルアルデヒドなどで固定し、 リン酸緩衝生理 食塩液 ( P B S ) で洗浄後、 X— g a 1を含む染色液で、 室温または 3 7 °C付近 で、 約 3 0分ないし 1時間反応させた後、 組織標本を I mM E D TAZ P B S 溶液で洗浄することによって、 jS—ガラクトシダ一ゼ反応を停止させ、 呈色を観 察すればよい。 また、 常法に従い、 1 a c Zをコードする mR NAを検出しても よい。 For example, when a part of the DNA region encoding the protein of the present invention is replaced by a) 3 monogalactosidase gene (1 ac Z) derived from Escherichia coli, the tissue that expresses the protein of the present invention originally Instead of the protein of the invention,; 3-galactosidase is expressed. Therefore, by staining with a reagent that serves as a substrate for 0-galactosidase such as, for example, 5-promo 4-chloro-3-indolyl; 8-galactobilanoside (X-gal), it is easy to carry out The expression state of the protein of the present invention in an animal body can be observed. Specifically, the protein-deficient mouse of the present invention or a tissue section thereof is fixed with datalaldehyde or the like, and phosphate buffered physiological After washing with saline solution (PBS), react with a staining solution containing X-ga 1 at room temperature or around 37 ° C for about 30 minutes to 1 hour, and then tissue samples are washed with ImM ED TAZ PBS solution. By washing, the jS-galactosidase reaction is stopped, and coloration may be observed. In addition, mRNA encoding 1 ac Z may be detected according to a conventional method.
上記スクリーニング方法を用いて得られる化合物またはその塩は、 上記した試 験化合物から選ばれた化合物であり、 本発明の D N Aに対するプロモーター活性 を促進または阻害する化合物である。  The compound or a salt thereof obtained by using the above-mentioned screening method is a compound selected from the above-mentioned test compounds, and is a compound that promotes or inhibits the DNA promoter activity of the present invention.
該スクリーニング方法で得られた化合物は塩を形成していてもよく、 該化合物 の塩としては、 生理学的に許容される酸 (例、 無機酸など) や塩基 (例、 アル力 リ金属など) などとの塩が用いられ、 とりわけ生理学的に許容される酸付加塩が 好ましい。 この様な塩としては、 例えば、 無機酸 (例えば、 塩酸、 リン酸、 臭化 水素酸、 硫酸など) との塩、 あるいは有機酸 (例えば、 酢酸、 ギ酸、 プロピオン 酸、 フマル酸、 マレイン酸、 コハク酸、 酒石酸、 クェン酸、 リンゴ酸、 蓚酸、 安 息香酸、 メタンスルホン酸、 ベンゼンスルホン酸など) との塩などが用いられる。 本発明の D N Aに対するプロモータ一活性を促進または阻害する化合物または その塩は、 本発明のタンパク質の発現の調節、 該タンパク質の機能を調節するこ とができるので、 例えば大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃 がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣がん、 甲状腺がん、 膝臓がん、 脳腫瘍または血液腫瘍などのがんの予防 ·治 療剤として有用である。  The compound obtained by the screening method may form a salt, and the salt of the compound may be a physiologically acceptable acid (eg, an inorganic acid) or a base (eg, an alkali metal). And the like, and particularly preferably a physiologically acceptable acid addition salt. Examples of such salts include salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.), and organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, Salts with succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc. are used. The compound of the present invention, which promotes or inhibits the activity of the promoter against DNA, or a salt thereof can regulate the expression of the protein of the present invention and regulate the function of the protein, such as colon cancer, breast cancer, lung cancer, Prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testis cancer, thyroid cancer, knee It is useful as a prophylactic and / or therapeutic agent for cancer such as cancer, brain tumor, or blood tumor.
さらに、 上記スクリ一ニングで得られた化合物から誘導される化合物も同様に 用いることができる。  Further, a compound derived from the compound obtained by the above screening can also be used.
該スクリーニング方法で得られた化合物またはその塩を含有する医薬は、 前記 した本発明のタンパク質またはその塩を含有する医薬と同様にして製造すること ができる。  A drug containing the compound or a salt thereof obtained by the screening method can be produced in the same manner as the above-mentioned drug containing the protein of the present invention or a salt thereof.
このようにして得られる製剤は、 安全で低毒性であるので、 例えば、 ヒトまた は哺乳動物 (例えば、 ラット、 マウス、 モルモット、 ゥサギ、 ヒッジ、 ブ夕、 ゥ シ、 ゥマ、 ネコ、 ィヌ、 サルなど) に対して投与することができる。 該化合物またはその塩の投与量は、 対象疾患、 投与対象、 投与ルートなどによ り差異はあるが、 例えば、 本発明の DNAに対するプロモーター活性を阻害する 化合物を経口投与する場合、 一般的に成人 (体重 60 kgとして) の乳がん患者 においては、 一日につき該化合物を約 0.1〜100mg、 好ましくは約 1. 0 〜5 Omg、 より好ましくは約 1. 0〜2 Omg投与する。 非経口的に投与する 場合は、 該化合物の 1回投与量は投与対象、 対象疾患などによっても異なるが、 例えば、 本発明の DN Aに対するプロモーター活性を阻害する化合物を注射剤の 形で通常成人 (体重 6 O kgとして) の乳がん患者に投与する場合、 一日につき 該化合物を約 0. 01〜3 Omg、 好ましくは約 0. :!〜 20mg、 より好まし くは約 0. 1〜1 Omgを静脈注射により投与するのが好都合である。 他の動物 の場合も、 体重 6 O kg当たりに換算した量を投与することができる。 The preparations obtained in this way are safe and low toxic and can be used, for example, in humans or mammals (eg, rats, mice, guinea pigs, egrets, sheep, sheep, bushus, dogs, dogs, cats, dogs). , Monkeys, etc.). The dose of the compound or a salt thereof varies depending on the target disease, the subject of the administration, the administration route, and the like.For example, when the compound of the present invention that inhibits the promoter activity for DNA is orally administered, generally the adult For a breast cancer patient weighing 60 kg, the compound is administered at about 0.1-100 mg, preferably about 1.0-5 Omg, more preferably about 1.0-2 Omg per day. When administered parenterally, the single dose of the compound varies depending on the administration subject, target disease, and the like.For example, a compound that inhibits the promoter activity of DNA of the present invention is usually administered in the form of an injection to an adult. When administered to a breast cancer patient weighing 6 O kg, the compound is administered in an amount of about 0.01 to 3 Omg per day, preferably about 0.:! To 20 mg, more preferably about 0.1 to 1 mg per day. Omg is conveniently administered by intravenous injection. In the case of other animals, the dose can be administered in terms of the weight per 6 O kg.
このように、 本発明の DNA発現不全非ヒト哺乳動物は、 本発明の DNAに対 するプロモーターの活性を促進または阻害する化合物またはその塩をスクリー二 ングする上で極めて有用であり、 本発明の DN A発現不全に起因する各種疾患の 原因究明または予防 ·治療剤の開発に大きく貢献することができる。  As described above, the non-human mammal deficient in expression of the DNA of the present invention is extremely useful for screening a compound or a salt thereof that promotes or inhibits the activity of the promoter for the DNA of the present invention. It can greatly contribute to investigating or preventing the causes of various diseases caused by insufficient expression of DNA.
また、 本発明のタンパク質のプロモータ一領域を含有する DNAを使って、 そ の下流に種々のタンパクをコードする遺伝子を連結し、 これを動物の卵細胞に注 入していわゆるトランスジエニック動物 (遺伝子移入動物) を作成すれば、 特異 的にそのタンパク質を合成させ、 その生体での作用を検討することも可能となる。 さらに上記プロモ一夕一部分に適当なレポ一夕一遺伝子を結合させ、 これが発現 するような細胞株を樹立すれば、 本発明のタンパク質そのものの体内での産生能 力を特異的に促進もしくは抑制する作用を持つ低分子化合物の探索系として使用 できる。 本明細書において、 塩基やアミノ酸などを略号で表示する場合、 IUPAC- IUB Commission on Biochemical Nomenclature による略号あるいは当該分野におけ る慣用略号に基づくものであり、 その例を下記する。 またアミノ酸に関し光学異 性体があり得る場合は、 特に明示しなければ L体を示すものとする。  In addition, using a DNA containing a promoter region of the protein of the present invention, genes encoding various proteins are ligated downstream thereof and injected into egg cells of an animal to produce a so-called transgenic animal (gene). Creating an introduced animal) will allow the specific synthesis of the protein and the study of its effects in living organisms. Furthermore, by binding an appropriate repo overnight gene to a portion of the above promoter and establishing a cell line in which this is expressed, the ability of the protein of the present invention itself to be produced in the body can be specifically promoted or suppressed. It can be used as a search system for low molecular compounds that have an action. In the present specification, bases, amino acids, and the like are indicated by abbreviations based on the abbreviations of the IUPAC- IUB Commission on Biochemical Nomenclature or commonly used abbreviations in the art, and examples thereof are described below. When there is an optical isomer for an amino acid, the L-form is indicated unless otherwise specified.
DNA :デォキシリポ核酸 cDNA 相補的デォキシリポ核酸 A アデニン DNA: Deoxylipo nucleic acid cDNA complementary deoxyliponucleic acid A adenine
T チミン  T thymine
G グァニン  G Guanin
C C
RNA リポ核酸 RNA liponucleic acid
mRNA メッセ、ンジャーリポ核酸 dATP デォキシアデノシン三リン酸 dTTP デォキシチミジン三リン酸 dGTP デォキシグアノシン三リン酸 dCTP デォキシシチジン三リン酸mRNA Messe, Nja lipo nucleic acid dATP Deoxyadenosine triphosphate dTTP Deoxythymidine triphosphate dGTP Deoxyguanosine triphosphate dCTP Deoxycytidine triphosphate
ATP アデノシン三リン酸 ATP Adenosine triphosphate
EDTA エチレンジァミン四酢酸 EDTA ethylenediaminetetraacetic acid
SD S ドデシル硫酸ナトリウム G 1 y SD S Sodium dodecyl sulfate G 1 y
A 1 a ァラニン  A 1 a Alanin
Va 1 バリン  Va 1 Valine
Le u ロイシン  Le u leucine
I 1 e  I 1 e
S e r セリン S e r serine
Th r スレオニン  Th r threonine
Cy s  Cy s
Me t メチォニン  Me t Methionin
G 1 u グルタミン酸  G 1 u Glutamic acid
As p ァスパラギン酸 As p Aspartic acid
L y s リジン  Lys lysine
Ar g アルギニン  Ar g Arginine
H i s ヒスチジン  H is histidine
Ph e フエ二ルァラニン Ty r :チロシン Ph e feniralanin Ty r: Tyrosine
Tr p : トリブトファン  Tr p: Tribute fan
P r o :プロリン  Pro: Proline
As n :ァスパラギン  As n: Asparagine
G i n :グルタミン  G in: glutamine
pG 1 u :ピログルタミン酸  pG 1 u: pyroglutamic acid
S e c :セレノシスティン (selenocysteine  S e c: selenocysteine
■ また、 本明細書中で繁用される置換基、 保護基および試薬を下記の記号で表記 する。 ■ In addition, substituents, protecting groups and reagents frequently used in the present specification are represented by the following symbols.
Me :メチル基  Me: methyl group
E t :ェチル基  E t: ethyl group
B u :ブチル基  B u: butyl group
Ph :フエニル基  Ph: phenyl group
TC :チアゾリジン— 4 (R) —カルポキサミド基  TC: thiazolidine-4 (R) -carboxamide group
To s : P-トルエンスルフォニル  To s: P-toluenesulfonyl
CHO :ホルミル  CHO: Formyl
B z 1 :ベンジル  B z 1: benzyl
Cl2-Bzl : 2, 6—ジクロ口べンジル Cl 2 -Bzl: 2,6-dichlorobenzene
Bom :ベンジルォキシメチル  Bom: benzyloxymethyl
Z :ベンジルォキシカルポニル  Z: benzyloxycarponyl
C 1一 Z : 2—クロ口べンジルォキシカルポニル  C 1-Z: 2-clozen benzyloxycarponyl
B r— Z : 2—ブロモベンジルォキシカルボニル  B r— Z: 2-bromobenzyloxycarbonyl
Bo c : t一ブトキシカルポニル  Bo c: t-butoxycarponyl
DNP :ジニトロフエニル  DNP: dinitrophenyl
T r t : トリチル  Trt: Trityl
Bum : t一ブトキシメチル  Bum: t-butoxymethyl
Fmo c : N— 9一フルォレニルメトキシカルポニル  Fmo c: N-9-Fluorenylmethoxycarbonyl
HOB t : 1ーヒドロキシベンズトリアゾール  HOB t: 1-hydroxybenztriazole
HOOB t : 3, 4—ジヒドロ一 3—ヒドロキシ一 4—ォキソ一 1, 2, 3—ベンゾトリアジン HOOB t: 3, 4-dihydro-3-hydroxy-1 4-oxo-1 1, 2, 3-benzotriazine
HONB :卜ヒドロキシ- 5 -ノルポルネン- 2, 3-ジカルボキシイミド HONB: Trihydroxy-5-norporene-2,3-dicarboximide
DCC : N, N, ージシクロへキシルカルポジイミド 本願明細書の配列表の配列番号は、 以下の配列を示す。 DCC: N, N, dicyclohexylcarposimide The sequence numbers in the sequence listing in the present specification show the following sequences.
〔配列番号: 1〕  [SEQ ID NO: 1]
R I PK 2のアミノ酸配列を示す。  2 shows the amino acid sequence of R I PK2.
〔配列番号': 2〕  [SEQ ID NO: 2]
配列番号: 1で表されるアミノ酸配列を有する R I PK2をコードする DNA の塩基配列を示す。  This shows the base sequence of DNA encoding RIPK2 having the amino acid sequence represented by SEQ ID NO: 1.
〔配列番号: 3〕  [SEQ ID NO: 3]
R I PK 2をコードする全長遺伝子を含む DN Aの塩基配列を示す。  1 shows the nucleotide sequence of DNA containing the full length gene encoding RIPK2.
〔配列番号: 4〕 - 実施例 2で用いられたアンチセンスォリゴヌクレオチドの塩基配列を示す。 〔配列番号: 5〕  [SEQ ID NO: 4]-This shows the base sequence of the antisense oligonucleotide used in Example 2. [SEQ ID NO: 5]
実施例 2で用いられたオリゴヌクレオチドの塩基配列を示す。  3 shows the nucleotide sequence of the oligonucleotide used in Example 2.
〔配列番号: 6〕  [SEQ ID NO: 6]
実施例 2で用いられたプライマ一の塩基配列を示す。  3 shows the nucleotide sequence of a primer used in Example 2.
〔配列番号: 7〕  [SEQ ID NO: 7]
実施例 2で用いられたプライマーの塩基配列を示す。  3 shows the nucleotide sequence of a primer used in Example 2.
〔配列番号: 8〕  [SEQ ID NO: 8]
実施例 2で用いられたプローブの塩基配列を示す。 以下において、 実施例により本発明をより具体的にするが、 この発明はこれら に限定されるものではない。 実施例 1  4 shows the nucleotide sequence of a probe used in Example 2. Hereinafter, the present invention will be more specifically described with reference to examples, but the present invention is not limited thereto. Example 1
乳がん組織で特異的に発現亢進している遺伝子群を明らかにするため、 乳がん 組織 2例、 その他 23種類の正常組織から抽出された total RNA (表 1 ) を材料とし、 oligonucleotide microarray (Human Genome U95A, U95B, U95C, U95D, U95E; Af fyme t r ix社) を用いて遺伝子発現解析を行つた。 To identify genes that are specifically upregulated in breast cancer tissue, total RNA (Table 1) extracted from two breast cancer tissues and 23 other normal tissues was used as a material. Gene expression analysis was performed using an oligonucleotide microarray (Human Genome U95A, U95B, U95C, U95D, U95E; Affymetrix).
実験方法は、 Affymetrix社の実験手引き書 (Expression analysis technical manual) に従った。 その結果、 乳がん組織において RIM2遺伝子の発現亢進が検 出された (表 2) 。  The experimental method followed Affymetrix's experiment guide (Expression analysis technical manual). As a result, increased expression of the RIM2 gene was detected in breast cancer tissues (Table 2).
〔表 1〕 〔table 1〕
RNAを抽出した組織 販冗兀 RNA extracted tissue
乳がん組織 (患者番号 9) BioClinical Partners 社 Breast cancer tissue (patient number 9) BioClinical Partners
脂肪 BioChain Institute社 Fat BioChain Institute
骨格筋 CI on tech社 Skeletal muscle CI on tech
心臓 Clontechi Heart Clontechi
CI on tech社  CI on tech
副腎 Clontechfh Adrenal gland Clontechfh
肝臓 Clontech社 Liver Clontech
勝 Jfe Clontech社 Win Jfe Clontech
Clontech社  Clontech
CI on tech社  CI on tech
肺 CI on tech社 Lung CI on tech
全脳 CI on tech社 Whole brain CI on tech
小脳 Clontech社 Cerebellum Clontech
胸腺 CI on tech社 Thymus CI on tech
乳腺 Clontech社 Mammary gland Clontech
Clontech社  Clontech
Clontech社  Clontech
直腸 BioChain Institute社 Rectal BioChain Institute
大腸 BioChain Institute社 Large intestine BioChain Institute
子宮 CI on tech社 Uterus CI on tech
子宮頸 BioChain Institute社― 〔表 2〕 組織 遺伝子発現: Cervical BioChain Institute- (Table 2) Tissue gene expression:
乳がん組織 (患者番号 9) 1.9 Breast cancer tissue (patient number 9) 1.9
脂肪 ND Fat ND
骨格筋 ND Skeletal muscle ND
、 ND  , ND
腎臓 ND Kidney ND
副腎 ND Adrenal gland ND
肝臓 ND Liver ND
腾臓 ND Monkey ND
脾臓 ND Spleen ND
気管 ND Tracheal ND
肺 ND Lung ND
全脳 ND Whole brain ND
小脳 ND Cerebellar ND
胸腺 ND Thymus ND
乳腺 ND Mammary gland ND
睡液腺 ND Sleeping gland ND
ND  ND
直腸 ND Rectal ND
大腸 ND Large intestine ND
子宮 . ND Uterus .ND
子宮頸 ND Cervical ND
a遺伝子発現量は、 oligonucleotide microarrayで発現が検出された 全遺伝子の発現量の中央値を 1として標準化した。  aThe gene expression level was normalized with the median expression level of all genes whose expression was detected by the oligonucleotide microarray as 1.
ND; not detected  ND; not detected
実施例 2 乳がん組織に発現亢進が観察された RIPK2遺伝子のアポト一シスに及ぼす影響 を解析するために、 RI PK2アンチセンスオリゴヌクレオチド導入実験を行つた。 まず、 配列番号: 3で表される塩基配列に対するアンチセンス (配列番号: 4 ) を設計後、 phosphorothioate化オリゴヌクレオチドを合成し、 HPLC精製して導入 実験に用いた (Amersham Pharmacia Biotech) 。 コントロールオリゴヌクレオチ ドとしては配列番号: 4で示される塩基配列のリバース配列 (配列番号: 5 ) を 同様に phosphorothioate化し、 HPLC精製して用いた (Amersham Pharmac i a Biotech) 。 Example 2 In order to analyze the effect of RIPK2 gene on the apoptosis observed in breast cancer tissues, RI PK2 antisense oligonucleotide transfection experiments were performed. First, after designing an antisense (SEQ ID NO: 4) for the base sequence represented by SEQ ID NO: 3, a phosphorothioated oligonucleotide was synthesized, purified by HPLC, and used for an introduction experiment (Amersham Pharmacia Biotech). As a control oligonucleotide, the reverse sequence (SEQ ID NO: 5) of the base sequence represented by SEQ ID NO: 4 was similarly phosphorothioated, purified by HPLC and used (Amersham Pharmacia Biotech).
被験細胞として乳がん細胞株 MDA- MB- 231 (In Vi tro, 14 (11)巻、 911- 915頁、 1978年.、 ATCCより購入) を用い、 オリゴヌクレオチド導入前日に 4X 104個の細胞 を 24ゥエルプレ一ト (Falcon社製) に播種した。 オリゴヌクレオチド導入には Ol igofec tAMINE (Invi trogen社製) を使用し、 そのプロトコールに従った。 導入 後 20時間で RNeasy mini ki t (Qi agen社製) のプロトコールに従って RNAを抽出し、 TaqManRTキット (Perkin- Elmer社製) を用いて cDNAを調製した。 また同時に逆転 写酵素を添加せずに同様の反応を行い、 非逆転写コントロールとした。 2種のプ ライマー (配列番号: 6および配列番号: 7 ) 、 およびプロ一ブ (配列番号: 8 ) を用いて RIM2遺伝子の発現量を見ることとし、 ABI7700 (Perkin- Elmer社製) マ ニュアルに従い PCRを行った。 RIPK2の発現量は c DNAを用いて得られたデ一夕か ら非逆転写コントロールを用いて得られたデータを差し引いた後に用いた totalRNA量で補正して比較した。 Using the breast cancer cell line MDA-MB-231 (In Vitro, vol. 14 (11), 911-915, pp. 911-915, 1978., purchased from ATCC) as test cells, 4 × 10 4 cells were obtained on the day before the introduction of the oligonucleotide. The seeds were seeded on a 24-plate (Falcon). Oligonucleotides (Invitrogen) were used for oligonucleotide introduction, and the protocol was followed. Twenty hours after the introduction, RNA was extracted according to the protocol of RNeasy mini kit (Qiagen), and cDNA was prepared using TaqManRT kit (Perkin-Elmer). At the same time, a similar reaction was carried out without adding reverse transcriptase, and used as a non-reverse transcription control. The expression level of the RIM2 gene was determined using two types of primers (SEQ ID NO: 6 and SEQ ID NO: 7) and a probe (SEQ ID NO: 8), and the ABI7700 (Perkin-Elmer) manual was used. PCR was performed according to the procedure. The expression level of RIPK2 was subtracted from the data obtained using the non-reverse transcription control from the data obtained using the cDNA, and then corrected with the total RNA amount used for comparison.
一方、 アンチセンスオリゴを導入して RIPK2遺伝子の発現を抑えた際のアポト —シスに及ぼす影響を見るために、 アンチセンスオリゴヌクレオチド導入後 3日 目に Cel l death detec t ion EL ISA (Roche社製) を用いてコントロールオリゴ導 入サンプルと比較した。  On the other hand, on the third day after the introduction of the antisense oligonucleotide, a cell death detection ELISA (Roche Co., Ltd.) was conducted to observe the effect on apoptosis when the expression of the RIPK2 gene was suppressed by introducing the antisense oligo. Was compared with the control oligo-introduced sample.
その結果、 RIM2遺伝子に対するアンチセンス (配列番号: 4 ) を用いた場合、 導入後 20時間で、 そのコントロールオリゴヌクレオチド (配列番号: 5 ) に比べ、 RIPK2遺伝子の発現量は 78%に減少していた。 また導入後 3日目でアポト一シスは コント口一ルォリゴヌクレオチド (配列番号: 5 ) を 100%とするとアンチセン スオリゴヌクレオチド (配列番号: 4 ) 導入時には 154%に増加していた。 これらのことから、 がん組織で発現亢進している RIPK2遺伝子の発現を抑制す ることにより、 がん細胞がアポトーシスを起こすことが確認され、 RIPK2遺伝子 が、 がん細胞の増殖やアポト一シスに関与していることが明らかとなった。 産業上の利用可能性 As a result, when the antisense (SEQ ID NO: 4) for the RIM2 gene was used, the expression level of the RIPK2 gene was reduced to 78% at 20 hours after the introduction compared to the control oligonucleotide (SEQ ID NO: 5). Was. On the 3rd day after the introduction, apoptosis increased to 154% when the antisense oligonucleotide (SEQ ID NO: 4) was introduced, assuming that the control oligonucleotide (SEQ ID NO: 5) was 100%. These results confirm that suppressing the expression of the RIPK2 gene, which is upregulated in cancer tissues, causes cancer cells to undergo apoptosis, and that RIPK2 gene promotes cancer cell growth and apoptosis. It was clear that they were involved in Industrial applicability
本発明のタンパク質はがん組織で発現が増加しており、 さらに、 本発明のタン パク質の活性を阻害するとがん細胞がアポトーシスを起こす。 従って、 本発明の タンパク質および該タンパク質をコードするポリヌクレオチドなどは、 がんの診 断マーカーである。 該タンパク質の活性を調節 (好ましくは阻害) する化合物ま たはその塩、 該タンパク質遺伝子の発現を調節 (好ましくは阻害) する化合物ま だはその塩は、 例えば、 大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃 がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣がん、 甲状腺がん、 勝臓がん、 脳腫瘍または血液腫瘍などのがんの予防'治 療剤として安全に使用することができる。 好ましくは、 ホルモン (例、 エストロ ゲンなど) 非依存性、 HER2発現陰性などのがん (例、 乳がんなど) の予防 ·治療 剤である。 さらに、 該タンパク質の活性を阻害する化合物またはその塩、 該タン パク質遺伝子の発現を阻害する化合物またはその塩は、 例えば、 アポトーシス作 用調節剤 (好ましくはアポトーシス作用促進剤) として安全に使用することもで さる。  The expression of the protein of the present invention is increased in cancer tissues, and when the activity of the protein of the present invention is inhibited, cancer cells undergo apoptosis. Therefore, the protein of the present invention and a polynucleotide encoding the protein are diagnostic markers for cancer. Compounds or salts thereof that regulate (preferably inhibit) the activity of the protein, compounds or salts thereof that regulate (preferably inhibit) the expression of the protein gene include, for example, colorectal cancer, breast cancer, lung cancer, Prostate cancer, esophageal cancer, stomach cancer, liver cancer, biliary tract cancer, spleen cancer, kidney cancer, bladder cancer, uterine cancer, ovarian cancer, testicular cancer, thyroid cancer, sarcoma It can be used safely as a prophylactic or therapeutic agent for cancer such as cancer, brain tumors or hematological tumors. Preferably, it is a prophylactic / therapeutic agent for cancer (eg, breast cancer, etc.) that is hormone-independent (eg, estrogen, etc.)-Independent, HER2 expression negative, etc. Further, the compound that inhibits the activity of the protein or a salt thereof, or the compound that inhibits the expression of the protein gene or a salt thereof can be safely used, for example, as an apoptosis-regulating agent (preferably, an apoptosis-action promoting agent). You can do that too.
また、 本発明のアンチセンスポリヌクレオチドや抗体は、 本発明で用いられる タンパク質の発現を阻害することができ、 例えば大腸がん、 乳がん、 肺がん、 前 立腺がん、 食道がん、 胃がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱が ん、 子宮がん、 卵巣がん、 精巣がん、 甲状腺がん、 塍臓がん、 脳腫瘍または血液 腫瘍などのがんの予防 ·治療剤、 好ましくは、 ホルモン (例、 エストロゲンな ど) 非依存性、 HER2発現陰性などのがん (例、 乳がんなど) の予防'治療剤とし て、 あるいはアポト一シス作用調節剤 (好ましくはアポトーシス作用促進剤) と して安全に使用することができる。  In addition, the antisense polynucleotide or antibody of the present invention can inhibit the expression of the protein used in the present invention. For example, colorectal cancer, breast cancer, lung cancer, prostate cancer, esophageal cancer, stomach cancer, liver Prevention of cancers such as cancer, biliary tract, spleen, kidney, bladder, uterus, ovary, testis, thyroid, kidney, brain or blood tumors · Therapeutic agent, preferably as a preventive agent for cancer (eg, breast cancer, etc.) that is hormone-independent (eg, estrogen) -independent, HER2-negative, etc., or an apoptotic agent (preferably It can be used safely as an apoptosis action promoter).

Claims

請求 の 範 囲 The scope of the claims
1 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のアミ ノ酸配列を含有するタンパク質もしくはその部分べプチドまたはその塩の活性を 阻害する化合物またはその塩を含有してなるがんの予防 ·治療剤。 1. It comprises a compound or a salt thereof which inhibits the activity of a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof. Cancer prevention and treatment.
2 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のアミ ノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩の遺伝子 の発現を阻害する化合物またはその塩を含有してなるがんの予防 ·治療剤。 2. It contains a compound or a salt thereof that inhibits the expression of a protein or a partial peptide thereof or a salt thereof that has the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1. Prevention and treatment of cancer.
3 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のアミ ノ酸配列を含有するタンパク質またはその部分ペプチドをコードするポリヌクレ ォチドの塩基配列に相補的もしくは実質的に相補的な塩基配列またはその一部を 含有するァンチセンスポリヌクレオチド。 3. Complementary or substantially complementary to the nucleotide sequence of a polynucleotide encoding a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof Antisense polynucleotide containing a base sequence or a part thereof.
4. 配列番号: 4で表される塩基配列を有する請求項 3記載のアンチセンスポ リヌクレオチド。  4. The antisense polynucleotide according to claim 3, which has a base sequence represented by SEQ ID NO: 4.
'5 . 請求項 3記載のアンチセンスポリヌクレオチドを含有してなるがんの予 防 ·治療剤。 '5. An agent for preventing or treating cancer, comprising the antisense polynucleotide according to claim 3.
6 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のアミ ノ酸配列を含有するタンパク質またはその部分ペプチドまたはその塩に対する抗 体を含有してなるがんの予防 ·治療剤。  6. A prophylactic / therapeutic agent for cancer comprising an antibody against a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof. .
7 . がんが、 大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝 臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣が ん、 甲状腺がん、 塍臓がん、 脳腫瘍または血液腫瘍である請求項 1、 請求項 2、 請求項 5または請求項 6記載の予防 ·治療剤。 7. Cancer is colorectal, breast, lung, prostate, esophagus, stomach, liver, biliary, spleen, kidney, bladder, uterus, ovary 7. The prophylactic or therapeutic agent according to claim 1, claim 2, claim 5, or claim 6 which is a cancer, testicular cancer, thyroid cancer, kidney cancer, brain tumor or blood tumor.
8 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のアミ ノ酸配列を含有するタンパク質またはその部分ペプチドまたはその塩に対する抗 体を含有してなるがんの診断薬。  8. A diagnostic agent for cancer comprising an antibody to a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof.
9 . 配列番号: iで表されるアミノ酸配列と同 "^もしくは実質的に同一のアミ ノ酸配列を含有するタンパク質またはその部分べプチドをコ一ドするポリヌクレ ォチドを含有してなるがんの診断薬。 ' 9. A cancer comprising a protein containing an amino acid sequence identical to or substantially identical to the amino acid sequence represented by SEQ ID NO: i, or a polynucleotide encoding a partial peptide thereof. Diagnostics. '
1 0 . がんが、 大腸がん、 乳がん、 肺がん、 前立腺がん、 食道がん、 胃がん、 肝臓がん、 胆道がん、 脾臓がん、 腎がん、 膀胱がん、 子宮がん、 卵巣がん、 精巣 がん、 甲状腺がん、 滕臓がん、 脳腫瘍または血液腫瘍である請求項 8または請求 項 9記載の診断薬。 10 .Cancer is colorectal, breast, lung, prostate, esophagus, stomach, liver, biliary, spleen, kidney, bladder, uterus, ovary 10. The diagnostic agent according to claim 8 or 9, which is a cancer, testicular cancer, thyroid cancer, Teng's cancer, brain tumor or hematological tumor.
1 1 . Receptor-Interac t ing Ser ine/Threonine Kinase 2の活性阻害作用を有 する化合物またはその塩を含有してなるがんの予防 ·治療剤。  1 1. A prophylactic / therapeutic agent for cancer comprising a compound having an inhibitory activity on Receptor-Interacting Serine / Threonine Kinase 2 or a salt thereof.
1 2 . Receptor-Interact ing Serine/Threonine Kinase 2の発現阻害作用を有 する化合物またはその塩を含有してなるがんの予防 ·治療剤。  1 2. Receptor-Interacting Serine / Threonine Kinase 2 A compound for inhibiting or inhibiting the expression of Kinase 2 or a salt thereof, which is a prophylactic / therapeutic agent for cancer.
1 3 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質もしくはその部分べプチドまたはその塩を用い ることを特徴とするがんの予防 ·治療剤のスクリーニング方法。  13 3. Prevention of cancer characterized by using a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof · A method for screening a therapeutic agent.
1 4 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質もしくはその部分べプチドまたはその塩を含有 することを特徴とするがんの予防 ·治療剤のスクリーニング用キット。  14. Prevention of cancer characterized by containing a protein having an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof · Kit for screening therapeutic agents.
1 5 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質またはその部分ペプチドをコードするポリヌク レオチドを用いることを特徴とするがんの予防 ·治療剤のスクリーニング方法。  15 5. Prevention of cancer characterized by using a polynucleotide encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof · A method for screening a therapeutic agent.
1 6 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質またはその部分ペプチドをコードするポリヌク レオチドを含有することを特徴とするがんの予防 ·治療剤のスクリーニング用キ ッ卜。 16. Prevention of cancer characterized by containing a polynucleotide encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof · Kit for screening therapeutic agents.
1 7 . 請求項 1 3もしくは請求項 1 5記載のスクリ一二ング方法または請求項 1 4もしくは請求項 1 6記載のスクリーニング用キットを用いて得られうるがん の予防 ·治療剤。  17. An agent for preventing or treating cancer obtainable by using the screening method according to claim 13 or claim 15 or the screening kit according to claim 14 or claim 16.
1 8 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質もしくはその部分べプチドまたはその塩の活性 を阻害する化合物またはその塩を含有してなるアポト—シス作用促進剤。  18. A protein containing a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a compound which inhibits the activity of a salt thereof or a salt thereof Apoptotic action promoter.
1 9 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質もしくはその部分べプチドまたはその塩の遺伝 子の発現を阻害する化合物またはその塩を含有してなるアポトーシス作用促進剤。 1 9. Inheritance of a protein containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof An apoptosis-promoting agent comprising a compound that inhibits the expression of an offspring or a salt thereof.
2 0 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質もしくはその部分ペプチドまたはその塩を用い ることを特徴とするアポトーシス作用促進剤のスクリーニング方法。 20. Screening for an apoptosis-activator characterized by using a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof Method.
2 1 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質またはその部分べプチドをコードするポリヌク レオチドを用いることを特徴とするアポトーシス作用促進剤のスクリーニング方 法。 21. An agent for promoting apoptosis, characterized by using a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a polynucleotide encoding a partial peptide thereof. Screening method.
2 2 . 請求項 3記載のアンチセンスポリヌクレオチドを含有してなるアポト一 シス作用促進剤。  22. An apoptosis-activating agent comprising the antisense polynucleotide according to claim 3.
2 3 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質またはその部分ペプチドまたはその塩に対する 抗体を含有してなるアポトーシス作用促進剤。  23. An agent for promoting apoptosis comprising an antibody against a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof.
2 4 . 哺乳動物に対して、 配列番号: 1で表されるアミノ酸配列と同一もしく は実質的に同一のアミノ酸配列を含有するタンパク質もしくはその部分ペプチド またはその塩の活性を阻害する化合物またはその塩、 または該タンパク質の遺伝 子の発現を阻害する化合物またはその塩の有効量を投与することを特徴とするが んの予防 ·治療方法。  24. A compound that inhibits the activity of a protein or a partial peptide thereof or a salt thereof containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1, A method for preventing and treating cancer, which comprises administering an effective amount of a salt or a compound that inhibits the expression of a gene of the protein or a salt thereof.
2 5 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質もしくはその部分べプチドまたはその塩の活性 を阻害する、 または該タンパク質の遺伝子の発現を阻害することを特徴とするが んの予防 ·治療方法。  25. Inhibits the activity of a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof, or expresses a gene of the protein A method for preventing and treating cancer characterized by inhibiting cancer.
2 6 . 配列番号: 1で表されるアミノ酸配列と同一もしくは実質的に同一のァ ミノ酸配列を含有するタンパク質もしくはその部分べプチドまたはその塩の活性 を阻害する、 または該タンパク質の遺伝子の発現を阻害することを特徴とするァ ポト一シス作用の促進方法。  26. Inhibits the activity of a protein having the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 1 or a partial peptide thereof or a salt thereof, or expresses a gene of the protein A method for promoting an apoptosis effect, which comprises:
2 7 . がんの予防 ·治療剤を製造するための、 配列番号: 1で表されるァミノ 酸配列と同一もしくは実質的に同一のァミノ酸配列を含有するタンパク質もしく はその部分ぺプチドまたはその塩の活性を阻害する化合物またはその塩、 または 該タンパク質の遺伝子の発現を阻害する化合物またはその塩の使用。 27. A protein or partial peptide containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 for producing a prophylactic or therapeutic agent for cancer A compound that inhibits the activity of the salt or a salt thereof, or Use of a compound or a salt thereof that inhibits expression of the protein gene.
2 8 . アポトーシス作用促進剤を製造するための、 配列番号: 1で表されるァ ミノ酸配列と同一もしくは実質的に同一のァミノ酸配列を含有するタンパク質も しくはその部分べプチドまたはその塩の活性を阻害する化合物またはその塩、 ま たは該タンパク質の遺伝子の発現を阻害する化合物またはその塩の使用。 28. A protein or a partial peptide or a salt thereof containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 1 for producing an apoptosis-activating agent Use of a compound or a salt thereof that inhibits the activity of the compound, or a compound or a salt thereof that inhibits the expression of a gene of the protein.
PCT/JP2003/007926 2002-06-24 2003-06-23 Preventives/remedies for cancer WO2004000346A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003243951A AU2003243951A1 (en) 2002-06-24 2003-06-23 Preventives/remedies for cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2002183148 2002-06-24
JP2002-183148 2002-06-24

Publications (1)

Publication Number Publication Date
WO2004000346A1 true WO2004000346A1 (en) 2003-12-31

Family

ID=29996673

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2003/007926 WO2004000346A1 (en) 2002-06-24 2003-06-23 Preventives/remedies for cancer

Country Status (2)

Country Link
AU (1) AU2003243951A1 (en)
WO (1) WO2004000346A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999040102A1 (en) * 1998-02-06 1999-08-12 Millennium Pharmaceuticals, Inc. Novel molecules of the card-related protein family and uses thereof
WO2000055350A1 (en) * 1999-03-12 2000-09-21 Human Genome Sciences, Inc. Human cancer associated gene sequences and polypeptides
WO2001000826A2 (en) * 1999-06-28 2001-01-04 Millennium Pharmaceuticals, Inc. Novel molecules of the card-related protein family and uses thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999040102A1 (en) * 1998-02-06 1999-08-12 Millennium Pharmaceuticals, Inc. Novel molecules of the card-related protein family and uses thereof
WO2000055350A1 (en) * 1999-03-12 2000-09-21 Human Genome Sciences, Inc. Human cancer associated gene sequences and polypeptides
WO2001000826A2 (en) * 1999-06-28 2001-01-04 Millennium Pharmaceuticals, Inc. Novel molecules of the card-related protein family and uses thereof

Also Published As

Publication number Publication date
AU2003243951A1 (en) 2004-01-06

Similar Documents

Publication Publication Date Title
WO2007004692A1 (en) Prophylactic/therapeutic agent and diagnostic agent for non-small cell lung cancer
WO2003099331A1 (en) Insulin resistance improving agents
WO2005067971A1 (en) Drug for preventing and treating arteriosclerosis
JP2007224009A (en) Agent for prevention or treatment of cancer
WO2003055506A1 (en) Preventives/remedies for cancer
WO2004028558A1 (en) Preventives/remedies for neurodegenerative disease
JP4353697B2 (en) Cancer preventive / therapeutic agent
WO2004000346A1 (en) Preventives/remedies for cancer
JP2004217634A (en) Preventing/treating agent for cancer
JP4445291B2 (en) Novel protein and its DNA
WO2002033071A1 (en) Survivin-like polypeptides and dnas thereof
JP2004089182A (en) Preventive/therapeutic agent for cancer
JP2003189873A (en) Prophylactic/therapeutic agent for cancer
JP4300008B2 (en) Novel protein and its DNA
JP2005015460A (en) Use of sglt homolog
JP2004075675A (en) Prophylactic/remedy for osteal/arthral disease
JP2001299363A (en) New polypeptide and dna encoding the same
JP2004026692A (en) New use of mepe
JP2004059502A (en) Prophylactic/therapeutic agent for hormone independent cancer
WO2004058969A1 (en) Preventives/remedies for cancer
WO2003097686A1 (en) Novel protein, its dna and use thereof
WO2003061697A1 (en) Preventives/remedies for cancer
JP2003277288A (en) Preventive/remedial agent for cancer
JP2005151826A (en) Use of c1qtnf5
JP2004269512A (en) Preventing and treating agent of renal disease

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP