WO1998022582A1 - Human cancer regression antigen protein - Google Patents

Human cancer regression antigen protein Download PDF

Info

Publication number
WO1998022582A1
WO1998022582A1 PCT/JP1997/004182 JP9704182W WO9822582A1 WO 1998022582 A1 WO1998022582 A1 WO 1998022582A1 JP 9704182 W JP9704182 W JP 9704182W WO 9822582 A1 WO9822582 A1 WO 9822582A1
Authority
WO
WIPO (PCT)
Prior art keywords
tumor antigen
cells
peptide
sequence
seq
Prior art date
Application number
PCT/JP1997/004182
Other languages
French (fr)
Japanese (ja)
Inventor
Kyogo Itoh
Yasuhisa Imai
Uhi Tho
Original Assignee
Kyogo Itoh
Yasuhisa Imai
Uhi Tho
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 Kyogo Itoh, Yasuhisa Imai, Uhi Tho filed Critical Kyogo Itoh
Priority to AU49667/97A priority Critical patent/AU4966797A/en
Publication of WO1998022582A1 publication Critical patent/WO1998022582A1/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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention belongs to the medical field, and more specifically, a method for treating cancer or an autoimmune disease, and more specifically, a cancer regression antigen that is regressed by being attacked by cytotoxic T cells, and an immunotherapy using the same. About.
  • T cells melanoma Toxic T cells
  • MHC major histocompatibility complex
  • TCR T cell receptor
  • MHC class I antigen binds to the above-mentioned tumor antigen peptide, moves to transgolgi on the mature side via cis Golgi, and is expressed on the cell surface (clinical immunity 27 (9): 1034-1042, 1995).
  • MHC class I antigen A tumor antigen protein, presented on MHC class I antigen on human cancer cells and targeted by host T cells, was identified by T. Boon in 1991 (Science 254: 1643-164 7,1991). This antigen causes cancer cells that express this antigen to be attacked by CTLs. It is called a cancer regression antigen because it regresses, and is named Melanoma antigen (MAGE) because it was identified from melanoma cells.
  • MAGE Melanoma antigen
  • Tumor antigen proteins recognized by CTL were successively identified from melanoma cells and the like. Tumor antigen proteins identified so far are classified into one of the following four categories according to their origin, structure (presence or absence of mutation) and expression mode (T. Bon et al., J. Exp. Med. 183). : 725-729, 1996):
  • the antigens classified here are expressed only in the testis and placenta in normal tissues, and are widely expressed in tumor tissues such as melanoma, head and neck cancer, non-small cell lung cancer, and bladder cancer. It is protein.
  • tumor antigen proteins include the above-mentioned MAGE and its one or more proteins that form two or more similar families (J. Exp. Med. 178: 489-495, 1993). ), BAGE (Immunity 2: 167-175, 1995) and GAGE (J. Exp. Med. 182: 689-698.1995), all of which have been identified from melanocytic cells. Recently, a widespread expression of NA17-A in melanoma was reported.
  • VLPOVF IRC HLA-A2-restricted antigen peptide
  • Tumor antigen proteins classified here are expressed in melanocytes in normal tissues and are a group of proteins expressed only in melanomas in tumor tissues. These tissue-specific proteins are strongly expressed in tumor cell melanomas. They are not found in other tissue types of cancer (adenocarcinoma or squamous cell carcinoma). It is considered a tumor antigen protein. Tumor antigen proteins in this category include tyrosinase (J. Exp. Med. 178: 489-495, 1993), MART-1 (Proc. Natl. Acad. Sci. USA 91: 3515, 1994), gp 1 0 0 (J. Exp. Med. 179: 1 005-1009, 1994) and g ⁇ 75 (J. Exp. Ed.
  • MART-1 Zmelan-A is considered to be a target molecule in von-Koyanagi-Ichiharada disease (S. Sugita, et al., Int. Immunol. 8: 799-803, 1996).
  • gp100 may act as an in vivo cancer regression antigen because the melanoma expressing it is highly sensitive to immunotherapy.
  • This category 1 tumor antigen protein is not expressed in tumors other than melanomas and cannot be applied to other tumors.
  • Tumor antigen proteins classified into this region are cancer-specific new antigens associated with genetic changes that occur during the transformation of normal cells into cancer.
  • Gene changes are known to be point mutation (mutant CDK4 antigen, mutation; 8-catenin antigen, MUM antigen) and alterative open reading frame (mutant gp75 antigen). Therefore, it is considered that such an antigen is specific to cancer and that specific immunity is easily established.
  • most of the genetic changes are expressed only in individual tumors or individual tumor cells. Therefore, it has the drawback that its expression frequency is extremely low and it is difficult to apply it clinically as a vaccine molecule for cancer treatment.
  • P15 is known as a case in which an antigen that is universally expressed as a non-mutant (ubiqui tous) in most normal cells and cancer cells is a cancer recognition molecule of CTL.
  • the p15 molecule has an HLA-A24 binding peptide.
  • pi5 is more strongly expressed in cancer cells than in normal cells.
  • the HER-2Zneu antigen which is an oncogene protein that is strongly expressed in adenocarcinoma and the like, is also classified as the same. That is, HER-2 / neu has an HLA-A2-binding peptide and is recognized by host killer T cells as a cancer regression antigen.
  • the antigen classified here is tumor antigen protein, it is considered to be applicable to a wide range of cancers due to its ubiquitous expression, but it will damage normal tissues due to poor disease specificity There is a possibility that CTL induction may be difficult (because of trans).
  • MUC-11 antigen-specific CTL which was thought to be MHC-unrestricted, recognized the peptide derived from the antigen, STAPPAHGV, as HLA-A11-restricted, and clinical studies using MAGE-3 peptide
  • STAPPAHGV the peptide derived from the antigen
  • HER-2 / neu Most of the above identified antigen peptides except HER-2 / neu have been found in melanomas, and have not been reported in squamous cell carcinomas or adenocarcinomas with high incidence.
  • HL A2661-restricted cancer antigen, a peptide is present and recognized by host CTLs from esophageal squamous cell carcinoma (M. Nakao, et a Cancer Res. 55: 4248-4252, 1995). Therefore, it is suggested that squamous cell carcinoma also has a tumor antigen protein that encodes a similar antigen peptide.
  • Squamous cell carcinoma is one of the most common human cancers.It is known that squamous cell carcinoma in esophageal and lung cancers is relatively resistant to current chemotherapy and radiation therapy. I have. From this point of view, the development of specific immunotherapy using tumor antigen peptides is expected. Disclosure of the invention The object of the present invention is to be applicable to a wide range of tumors such as squamous cell carcinoma, or to a large number of patients with tumors, even if the applicable tumors are limited, or to treat or diagnose such tumors. An object of the present invention is to provide a tumor antigen peptide and the like which assist complementation of cancer and can be applied to various tumors.
  • tumor antigen proteins that are highly expressed in tumors are also thought to be expressed in normal tissues and cause an autoimmune disease due to excessive immune responses derived from the tumor antigen proteins.
  • the antisense DNA or tumor antigen peptide that prevents expression of a gene that encodes the tumor antigen protein is blocked.
  • a therapeutic method that specifically blocks the immune response using evening gonists is expected.
  • tumor antigen proteins from squamous cell carcinomas to obtain tumor antigen proteins or their corresponding tumor antigen peptides that can be widely applied to the treatment and diagnosis of tumor cells other than melanoma cells, especially squamous cell carcinoma, etc. Tried.
  • the present inventors have established CTL (YAM-CTL) that recognizes an HLA-CW0102-restricted tumor antigen peptide from metastatic lymph node cancer tissue infiltrating lymphocytes of esophageal cancer patients. This CTL strongly impairs the HLA-CW0102-positive esophageal cancer cell line TE8 (Kumamoto University School of Medicine).
  • the recombinant plasmid of the cDNA library and the recombinant plasmid of the HLA-CW0102 cDNA created from the TE8 cancer cell line were simultaneously transfected into the fibroblast cell line C0S7 cells, and YAM-CTL was added to the transfectant.
  • the cells were allowed to act, and whether or not the YAM-CTL cells were activated was screened by measuring the amount of IFN-7 produced. The result As a result, a gene encoding the tumor antigen protein of the present invention was successfully cloned from tumor cells other than melanoma TE8.
  • the present invention relates to a partial peptide of a tumor antigen protein having an amino acid sequence of SEQ ID NO: 2, which binds to a major histocompatibility complex (MH C) class I antigen and has T cells.
  • MH C major histocompatibility complex
  • the present invention relates to a partial peptide of a tumor antigen protein having the amino acid sequence of SEQ ID NO: 4, which is recognized by T cells by binding to a major histocompatibility complex (MHC) class I antigen.
  • MHC major histocompatibility complex
  • An oligonucleotide molecule that encodes, preferably the oligonucleotide nucleotide sequence whose nucleotide sequence encodes a tumor antigen protein; SEQ ID NO: 4; An oligonucleotide consisting of a sequence complementary to the base sequence in the coding sequence of the coding polynucleotide molecule or its 5 'non-coding sequence.
  • FIG. 1 shows the amino acid sequences of fragments 1 and 2 obtained from clone 3. Underline indicates fragment 1 and bold diagonal line indicates fragment 2.
  • FIG. 2 is a graph showing the amount of IFN- 7 produced by fragments 1 and 2 obtained from clone 3 measured by EUSA.
  • ORF indicates open reading frame, F 1 indicates fragment 1, and F 2 indicates fragment 2.
  • (-) Indicates that the cotransfected plasmid was not transfected, A24 indicates the cotransfected plasmid of HLA-A24 cDNA, and Cwl indicates the recombinant plasmid of HLA-Cw0102 cDNA. The result of simultaneous transfection is shown.
  • FIG. 3 shows the amino acid sequences of fragments 1, 2 and 3 obtained from clone 6. Underline indicates fragment 1, bold italic indicates fragment 2, double line indicates fragment 3.
  • FIG. 4 is a graph showing the amount of IFN-7 produced by fragments 1, 2 and 3 obtained from clone 6 measured by ELIS A.
  • ORF indicates the open reading frame, F1 indicates fragment 1, F2 indicates fragment 2, and F3 indicates fragment 3.
  • (1) is not simultaneously transfected, A24 is a simultaneously transfected recombinant plasmid of HLA-A24 cDNA, and Cwl is a simultaneously transfected plasmid of HLA-Cw0102 cDNA. Shows what was transfected.
  • Partial peptide of tumor antigen protein, tumor antigen peptide recognized by T cells by binding to major histocompatibility complex (MHC) class I antigen A partial peptide consisting of at least 7-10, preferably 9 contiguous amino acid sequences of a tumor antigen protein, which is presented on the cell surface in association with the MHC class I antigen on the cell surface.
  • MHC major histocompatibility complex
  • a T cell that specifically binds to the conjugate binds, it can transmit a signal to the T cell, that is, form a conjugate with the MHC class I antigen recognized by the T cell.
  • the “bond” here is a non-covalent bond.
  • Methods for confirming that a peptide binds to MHC class I antigen and is recognized by T cells include, for example, endogenous expression of the peptide in appropriate cells, or by externally adding (pulsing) the peptide.
  • the peptide is presented on the cell surface by binding to the MHC class I antigen, followed by the action of tumor antigen protein-specific T cells on the peptide-presenting cells, which damage the peptide-presenting cells
  • There is a method to measure the site force in produced when receiving the drug interferon, TNF, and CTL.
  • cytotoxicity of peptide presentation cells it can also be used how to use a peptide-presenting cells labeled with 5 1 C r.
  • CTLs are preferably used as T cells to be recognized.
  • the tumor antigen protein or tumor antigen peptide according to the present invention can be identified, for example, as follows.
  • the tumor antigen peptide that binds to the MHC class I antigen of the tumor cells is oxidized and extracted, and various peptides separated by high-performance liquid chromatography express antigen-presenting MHC.
  • the tumor antigen protein for example, B cells of the same patient
  • mass spectrometry was performed. This method is used to determine the sequence.
  • the same amount of gp100 from melanoma cells A tumor antigen peptide derived from offspring Pmell7 has been identified (Science264: 716-719, 1994).
  • a method for determining a gene encoding a tumor antigen protein and further identifying the corresponding tumor antigen peptide involves cloning the gene encoding the tumor antigen protein using molecular biology techniques.
  • CDNA is prepared from tumor cells, and the cDNA is transfected with cells that do not express the tumor antigen protein (for example, COS cells) together with the antigen-presenting MHC class I antigen gene to express them temporarily. Screening based on the reactivity of the CTL against it is repeated to isolate the gene encoding the tumor antigen protein.
  • the above-mentioned genes of MAGE, tyrosinase, MART-1, gp100, and gp75 have been cloned.
  • the following method is used to estimate and identify the tumor antigen peptide actually bound to and presented to the MHC class I antigen from the information on the tumor antigen gene.
  • fragments of genes encoding tumor antigen proteins of various sizes were prepared by PCR, exonuclease, restriction enzymes, etc., and cells that did not express tumor antigen proteins together with antigen-presenting MHC class I antigen genes ( For example, transfected into COS cells, etc., and transiently expressed, and the region containing the tumor antigen peptide is limited by the reactivity of CTL.
  • the peptide was synthesized, and the antigen-presenting MHC class I antigen was expressed, but it was pulsed into cells that did not express the tumor antigen protein, and the tumor antigen peptide was identified by examining the reaction of c ⁇ L. Med. 176: 1453, 1992, J. Exp. Med. 179: 24, 759, 1994).
  • MHC class I antigens such as HLA-Al, -A0201, -A0205, -All, A31, -A6801, -B7, -B8, -B2705, -37, -CW0401, and -CW0602.
  • sequence regularities (motifs) of the peptides presented are known (seminars in IMMUNOLOGY 5: 81-94, 1993), and the tumor antigen peptide candidates are examined with reference to them.
  • a method in which the peptide is bound and confirmed by the same method as described above is also used (Eur. J. Immunol, 24: 759, 1994, J. Exp. Med. 180: 347, 1994).
  • peptides determined in this way can be produced by methods known in normal peptide chemistry. For example, “Peptide Syntheses”, Interscience, New York. 1996, “The Proteins", Vol. 2, Academic Press Inc., New York. 1976. “Peptide synthesis” Maruzen Co., Ltd., 1975, “Peptide Synthesis Basics and Experiments ", Maruzen, 1985, and the like. That is, either the liquid phase method or the solid phase method can be selected for synthesis depending on the configuration of the C-terminal site, and the liquid phase method is more preferable. That is, a peptide can be produced by appropriately protecting and deprotecting a functional group of an amino acid with an appropriate protecting group, and bonding the amino acid to each residue or several residues. The protective group for the functional group of the amino acid is described, for example, in the above-mentioned book describing peptide chemistry.
  • a derivative of the tumor antigen peptide of the present invention (a derivative of a tumor antigen peptide that is a partial peptide of a tumor antigen protein and that is recognized by T cells by binding to an MHC class I antigen)” Means a peptide in which one or several amino acid sequences of the tumor antigen peptide of the present invention have been substituted, deleted, inserted or added.
  • Preferred derivatives include the tumor antigen peptides, in which the epitope region involved in binding to CTL remains unchanged and amino acid residues involved in binding to MHC class I antigen are substituted, deleted, or deleted.
  • Inserted or added derivatives can be mentioned, and more preferably, derivatives thereof, in which only one amino acid residue is substituted (Im munol. 84: 298-303, 1995).
  • Such a derivative can bind strongly to the MHC class I antigen while maintaining the binding property to CTL as it is, so that it can be applied as a more useful tumor antigen peptide.
  • Such a derivative can be prepared, for example, by the method described in Molecular Cloning: A Laboratory Manual, 2nd edition, Volume 13, Sambrook, J. et al., Cold Spring Harber Labolatory Press, New York, 1989. Site-directed mutagenesis And can be prepared by a method such as PCR.
  • the tumor antigen peptide of the present invention or a derivative thereof is encoded by the below-described oligonucleotide of the present invention.
  • the “derivative of the present invention” includes a derivative of the tumor antigen peptide of the present invention or a derivative in which a partial amino acid residue of the peptide is substituted, deleted, inserted, or added, wherein the amino group or the carboxyl group is modified. are also included.
  • Examples of the modifying group for the amino group include an acyl group.
  • Specific examples include an alkanol group having 1 to 6 carbon atoms, 1 carbon atom substituted by a phenyl group, an alkanol group having 6 or more carbon atoms, and 5 to 5 carbon atoms.
  • Examples of the carboxyl group-modifying group include an ester group and an amide group.
  • Specific examples of the ester group include an alkyl ester group having 1 to 6 carbon atoms, a carbon atom having 0 carbon atoms substituted with a phenyl group
  • Examples of the amide group include an amide group, one or two alkyl groups having 1 to 6 carbon atoms, and a cycloalkyl ester group having 5 to 7 carbon atoms.
  • Substituted amide group phenyl group-substituted alkyl group having 0 to 6 carbon atoms, amide group substituted with 1 or 2 carbon atoms, 5- to 7-membered ring including amide group nitrogen atom Amide groups forming azacycloalkanes and the like can be mentioned.
  • the present invention further provides a medicament containing the tumor antigen peptide of the present invention or a derivative thereof.
  • the tumor antigen protein and the tumor antigen peptide of the present invention can be administered together with an adjuvant so as to effectively establish cellular immunity, or can be administered in the form of particles.
  • an adjuvant those described in the literature (Cl in. Microbiol. Rev. 7: 277-289. 1994) can be applied.
  • dosage forms include ribosome preparations, particulate preparations bound to beads with a diameter of several meters, and preparations linked to lipids to efficiently present antigen to MHC class I antigens. Any method of administration is used.
  • tumor antigen protein and tumor antigen peptide of the present invention in the preparation can be appropriately adjusted depending on the disease to be treated, the age and weight of the patient, etc., and is usually 0.0001 nig to 1000 nig, preferably 0. .000 mg to 1000 mg, preferably administered once every several days to several months.
  • the “antibody” against the tumor antigen peptide or a derivative thereof according to the present invention may be, for example, a tumor antigen by a method described in Antibodies; A Laboratory Manual, Lane. HD et al., Edited by Cold Spring Harber Laboratory Press, New York 1989.
  • an antibody that recognizes a tumor antigen protein or an antibody that neutralizes its activity can be easily prepared.
  • Uses of the antibody include affinity chromatography, screening of a cDNA library, immunological diagnostic methods, pharmaceuticals and the like.
  • the immunological diagnostic method can be appropriately selected from an immunoblot method, a radioimmunoassay method (RIA), an enzyme immunoassay method (ELISA), a fluorescence or luminescence measurement method, or the like.
  • the present invention further relates to an oligonucleotide molecule encoding the tumor antigen peptide of the present invention or a derivative thereof.
  • the oligonucleotide molecules of the present invention can be in the form of DNA or RNA, where DNA includes cDNA, genomic DNA and synthetic DNA. Also, DNA and RNA may be single-stranded or double-stranded, and in the case of single-stranded, may include both the sense strand and the antisense strand.
  • Oligonucleotide molecules in which a part of a certain nucleotide sequence has been substituted, deleted, inserted or added are described in Molecular Cloning: A Laboratory Manual, 2nd edition, Volume 1-3, Sambrook, J. et al., Cold Spring Harber Labolatory It can be produced by the method described in Press Publishing New York, 1980. For example, it can be produced by site-directed mutagenesis or PCR.
  • the oligonucleotide molecules of the present invention also include these mutant oligonucleotide molecules. Examples of such mutant oligonucleotide nucleotides include, for example, one or more nucleotides in the nucleotide sequence of SEQ ID NO: 2 or 4.
  • the oligonucleotide molecules of the present invention also include "oligonucleotide molecules that hybridize to the oligonucleotide molecules of the present invention under stringent conditions".
  • a DNA molecule that hybridizes to a DNA molecule can be obtained, for example, by the method described in the aforementioned Molecular Cloning.
  • “hybridizing under stringent conditions” means, for example, heating at 42 ° C. in a solution of 6 XSSC, 0.5% SDS and 50% formamide, followed by 0.1 XSSC Under the condition of washing at 68 ° C. in a 0.5% SDS solution, a positive hybridization signal was still observed.
  • DNA to produce a protein can be carried out, for example, based on many books and literature such as the aforementioned Molecular Cloning.
  • a translation initiation codon is added upstream of the DNA to be expressed and a translation stop codon is added downstream to control the promoter sequence (eg, trp, lac, T7, SV40 early promoter) that controls transcription.
  • an appropriate vector for example, pBR322, pUC19, pSV • SPORT1, etc.
  • an expression plasmid that replicates and functions in host cells is produced.
  • the expression plasmid is introduced into an appropriate host cell to obtain a transformant cell.
  • Examples of the host cell include prokaryotes such as Escherichia coli, unicellular eukaryotes such as yeast, and cells of multicellular eukaryotes such as insects and animals.
  • Methods for introducing genes into host cells include the calcium phosphate method, the DEAE-dextran method, and the electric pulse method.
  • the transformant produces the target protein by culturing in a suitable medium.
  • the protein obtained as described above can be isolated and purified by a general biochemical method.
  • Transformants transformed with the plasmid of the present invention are also within the scope of the present invention. Is included.
  • the present invention further relates to a medicament containing the oligonucleotide molecule of the present invention.
  • the “medicine” containing the oligonucleotide molecule of the present invention can treat or prevent a tumor by, for example, administering the DNA of the present invention to a tumor patient or the like.
  • Methods of administering the DNA of the present invention and introducing it into cells include viral vectors and other methods (Nikkei Science, April 1994, pp. 20-45, Monthly Pharmaceutical Affairs, 36 (1) 23-48 (1994). Any of the methods in Experimental Medical Special Edition, 12 (15), (1994), and references cited therein (etc.) can be applied.
  • the DNA of the present invention is added to an RNA virus or DNA virus such as retrovirus, adenovirus, adeno-associated virus, herpes virus, vaccinia virus, box virus, polio virus, and simbis virus.
  • retrovirus adenovirus, adeno-associated virus, herpes virus, vaccinia virus, box virus, polio virus, and simbis virus.
  • retrovirus adenovirus, adeno-associated virus, vaccinia virus and the like is particularly preferable.
  • ribosome method direct injection of the expression plasmid into the muscle
  • lipofectin method lipofectin method
  • microinjection method calcium phosphate method
  • electoral poration method ribosome method
  • Plasmids having these oligonucleotide nucleotide molecules of the present invention are also included in the scope of the present invention.
  • an in vivo method of directly introducing the gene into the body, and the use of certain cells from humans to transform the gene in vitro There is an ex vivo method to introduce cells into cells and return the cells to the body (Nikkei Science, April 1994, pp. 20-45, Monthly Pharmaceutical Affairs, 36 (1) 23-48 (1994), extra edition of experimental medicine, 12 (15), (1994), and references cited therein). In vivo methods are more preferred. When administered by the in vivo method, it can be administered by an appropriate administration route depending on the disease, symptom and the like for the purpose of treatment.
  • the DNA-containing ribosome or membrane fusion ribosome (such as Sendai virus (HVJ) -liposome) of the present invention is in the form of a ribosome preparation such as a suspending agent, a freezing agent, and a centrifugal concentrated freeze agent. be able to.
  • the content of the DNA of the present invention in the preparation can be appropriately adjusted depending on the disease to be treated, the age and weight of the patient, etc., and usually, the DNA of the present invention is 0.0001 mg to 100 mg, preferably O.OOlmg. 1010 mg, preferably administered once every few days or months.
  • the present invention further provides an oligonucleotide molecule comprising a coding sequence of a polynucleotide molecule encoding the tumor antigen protein of SEQ ID NO: 2 or 4, or a sequence complementary to a base sequence in the 5 ′ non-coding sequence thereof, or It relates to the chemically decorated body.
  • a DNA consisting of 9 or more bases having a sequence complementary to the base sequence in the coding sequence of the polynucleotide molecule consisting of the base sequence of SEQ ID NO: 2 or 4 (structural gene portion) or its 5 ′ non-coding sequence Or RNA.
  • Such DNA or RNA is the DNA of the antisense strand of double-stranded DNA or RNA corresponding to the DNA of the antisense strand, and is composed of 9 bases or more (hereinafter referred to as antisense oligonucleotide).
  • This antisense oligonucleotide can be easily prepared, for example, as DNA based on the nucleotide sequence of the gene encoding the tumor antigen peptide of the present invention, or by incorporating this DNA into a gene expression plasmid in the antisense direction. Can be produced.
  • This antisense oligonucleotide encodes the cDNA of the gene of the present invention. It may be a sequence complementary to any of the inking portion and the 5 'non-coding portion, but is preferably a transcription initiation site, a translation initiation site, a 5' untranslated region, a boundary region between exon and intron, or 5 '. It is desirable that the sequence be complementary to the CAP region.
  • “Chemical modification of an oligonucleotide molecule” refers to a chemically modified product that can enhance the translocation of DNA or RNA into cells or the stability in cells.
  • phosphothioate, phosphorodithioate Derivatives such as alkyl phosphotriesters, alkyl phosphonates and alkyl phosphoamidates ("Ant isense
  • antisense oligonucleotide DNA or RNA (hereinafter, referred to as antisense oligonucleotide) having a complementary sequence of the present invention. This method can treat or prevent autoimmune diseases by reducing the production of tumor antigen proteins. Pharmaceuticals containing such antisense oligonucleotides are also included in the present invention.
  • a preferable length of the antisense oligonucleotide is, for example, 5 to 200 bases.
  • preferred lengths of the antisense oligonucleotide include, for example, 100 bases or more, and preferably 300 bases or more, More preferably, it is 500 bases or more.
  • examples of a method for introducing this antisense oligonucleotide into cells include the method described in Experimental Medicine, Vol. 12, 1994, and Liposome Co., Ltd. There is a method using a replacement virus.
  • Expression plasmid for antisense oligonucleotides The gene is simply connected by connecting the gene of the present invention so that the gene of the present invention is transcribed in the reverse direction behind the promoter using a normal expression vector, that is, in the 3 ′ to 5 ′ direction. Can be manufactured.
  • a plasmid having such an antisense oligonucleotide is also included in the present invention.
  • the antisense oligonucleotide or its chemically modified product is administered as it is, it is formulated by mixing with a stabilizer, buffer, solvent, etc., and is used simultaneously with antibiotics, anti-inflammatory drugs, anesthetics, etc. You can also.
  • the preparations thus prepared can be administered in various ways. Preferably, the administration is performed daily or every few days to several weeks. In order to avoid such frequent administration, it is possible to prepare a sustained-release minipellet preparation and implant it near the affected area. Alternatively, it can be continuously and gradually administered to a patient using an osmotic pump or the like. The usual dosage should be adjusted so that the concentration at the site of action is 0.1%.
  • a medicament containing such an antisense oligonucleotide or a chemically modified product thereof is also included in the present invention.
  • the present invention will be described in more detail with reference to examples, but the present invention is not limited to these examples.
  • CTL cytotoxic T cell
  • the operatively removed 67-year-old male esophageal cancer metastatic lymph node tissue was aseptically minced and pasted into a paste, and then in the presence of interleukin 2 for about 60 days, 5% CO 2 (95) Incubate in an incubator. Meanwhile, culture 2 8 after day cytotoxic ability against various cancer cells of T cells to proliferate in culture frequently 5 1 C r release assay and IFN - were analyzed by 7 measurements. As a result, T cells from day 39 to day 49 of culture were mainly CD8-positive killer T cells, and were restricted by HLA-Cw0102 among MHC class I antigens. Was found to exhibit CTL activity.
  • the squamous cell carcinoma TE8 cell line (M. Nakao et al., Cancer Research 55, 4248-4252, 1995) showed the highest sensitivity to the CTL. Therefore, a large amount of the above CTL (named YA-CTL) was stored in a liquid nitrogen-containing cell cryopreservation tank to prepare for cloning of a cancer regression antigen gene recognized by CTL.
  • Reference example 2
  • HLA-Cw0102 cDNA derived from TE8 cells was incorporated into an expression vector pCR3 (manufactured by INVITROGEN) to produce a recombinant plasmid.
  • Reference example 3 HLA-Cw0102 cDNA derived from TE8 cells was incorporated into an expression vector pCR3 (manufactured by INVITROGEN) to produce a recombinant plasmid. : Reference example 3
  • RNA purification system manufactured by Pharmacia Biotech
  • separation of the total RNA fraction from TE8 cells and preparation of poly (A) + mRNA using an oligo (dT) column were performed according to the attached protocol.
  • oligo (dT) column were performed according to the attached protocol.
  • GIBC0 BRL Superscript Plasmid System
  • GIBCO BRL express this cDNA pSV-SP0RT1 (GIBCO BRL) and ligated to the restriction sites Not 1 and Sal 1 to obtain a recombinant plasmid.
  • This recombinant plasmid was subjected to an electric pulse under the conditions of 25 zF, 200 ⁇ , and 2.5 kV using a Gene Pulser (manufactured by Bio-Rad) and an E.coli elect-mouth Matsux DH10B / p3 TM cell (GIBCO BRL) Transformants with recombinant plasmids selected on LB medium (1% pactotryptone, 0.5% NaCl, pH 7.3) containing ampicillin (50 gZml) did.
  • LB medium 1% pactotryptone, 0.5% NaCl, pH 7.3
  • ampicillin 50 gZml
  • IFN-7 interferon
  • ELIS A Enzymimnoassay
  • recombinant plasmid DNA is recovered from the pool of transformants prepared in Reference Example 3.
  • Recombinant plasmid DNA from transformants cultured in TYGPN medium was prepared by microplate-based lysis method (FM Ausubel et al., Ed., CURRENT P0TC0LS IN M0LECULAR BIOLOGY, John Wiley & Sons, Inc.). .
  • Recombinant plasmid DNA recovered by isopropanol precipitation was suspended in a 10 mM Tris, ImM EDTA, pH 7.4 solution containing 50/1 20 ng / ml RNase.
  • the recombinant plasmid DNA prepared above and the recombinant plasmid of the HLA-Cw 0102 cDNA prepared in Reference Example 2 were used in C0S7 cells (Gluzan. Y. Cell, 23: 175-182, 1981).
  • C0S7 cells were added at 1 ⁇ 10 4 cells / well of a 96-well flat-bottom microplate and cultured for 1 day in 100 ⁇ l of RPMI medium containing 10% FCS.
  • 100 ng of the recombinant plasmid of HLA-Cw0102 cDNA prepared in Reference Example 2 was added to about 100 transformants of the recombinant plasmid TE8 cDNA 25 // 1 and the ribofectin reagent (about 100-fold diluted) was added.
  • Ribofectamine manufactured by GIBC0-BRL) 25; / 1.
  • the obtained mixed solution 501 fusion suspension of ribosome and recombinant plasmid was added to the cultured C0S7 cells, and double transfected.
  • Transfectants Two transfectants were prepared. Transfectants are cultured for 48-72 hours at 37 ° C, the culture medium is removed, and 1 x 10 4 TE8 CTL cells per well are added and 100 1 10% human serum and 50 UZml are added. The cells were cultured in a culture solution containing IL-2 at 37 ° C for 16 to 24 hours. The culture was collected and IFN-y was measured by ELISA.
  • the transformant pool is cultured for about 6 hours in LB (ampicillin 1 / ml) medium, and the culture is further cultured on a plate of LB agar medium containing ampicillin ( ⁇ ⁇ ⁇ ) for 1 day. Transfer 200 colonies from each colony to a total of 8 ⁇ 200 colonies in each well of a 96-well microplate, and incubate in the same manner as above under the condition that only one transformant per well is used. Then, recombinant plasmid DNA of TE8 cDNA was prepared.
  • the recombinant plasmid of TE8 cDNA and the recombinant plasmid of HLA-Cw0102 cDNA were double-transfected into C0S7 cells by the same method as described above, and then mixed and cultured with TE8 CTL cells. Reacts IFN-7 in the resulting culture was quantified and positive plasmids were selected. By this operation, two TE8 cell cDNA recombinant plasmid clones reacting with TE8 CTL cells were selected, and named clone 3 and clone 6, respectively. For clones 3 and 6, the same operation was repeated once more to confirm the production of IFN-7 by TE8 CTL cells.
  • Transformant clones 3 and 6 each having a recombinant plasmid containing the cDNA of the tumor antigen gene of interest were cloned for 14-16 at 37 ° C in LB medium containing 500 ml of ampicillin (50 ⁇ 1 / ml). After culturing for an hour, the cells were collected by centrifugation. Recombinant plasmid was recovered from the cells according to the PLASMID MAXI kit (QIAGBN).
  • the cDNA is integrated at the site between the SP6 RNA polymerase promoter sequence and the T7 RNA polymerase promoter sequence.
  • the SP6 Promoter primer and the T7 promoter primer described in the literature were synthesized.
  • a didoxyquencing reaction was performed by combining the SP6 Promoter primer or T7 promoter primer with Fluore-dATP Labeling Mix (Pharmacia Biotech) and AutoRead Sequencing Kit (Pharmacia Biotech).
  • the nucleotide sequence of the cDNA was determined from both ends using a fluorescent DNA sequencer (Pharmacia Biotech).
  • the nucleotide sequence of cDNA corresponding to clone 3 was determined to be 532 base pairs in total length, and the nucleotide sequence of cDNA corresponding to clone 6 was determined to be 756 base pairs in total length, as shown in SEQ ID NO: 1 and SEQ ID NO: 4, respectively. 7
  • the cDNA base sequence of clone 6 obtained had a 3'-UTR (untranslated portion) and a poly (A) tail added to the 3'-end, but the rest was that of histone H1. It had 100% homology with the nucleotide sequence.
  • the deduced amino acid sequence of the obtained cDNA base sequence also had 100% homology with the reported amino acid sequence of histone H1.
  • the cDNA sequence and deduced amino acid sequence are shown in SEQ ID NO: 3.
  • Fragment 1 (276 bp, 92 amino acids) and Fragment 2 (278 bp, 92 amino acids) were prepared using a TA cloning kit (Invitrogen). The amino acid sequences of fragments 1 and 2 are shown in FIG.
  • Fragment 1 (180 bp, 60 amino acids), Fragment 2 (362 bp, 100 amino acids) and Fragment 3 (281 bp, 93 amino acids) were prepared using a TA cloning kit. The amino acid sequences of fragments 1, 2 and 3 are shown in FIG.
  • a medicament for activating antitumor immunity using the tumor antigen protein and the tumor antigen peptide of the present invention a medicament for treating an autoimmune disease using an antibody against the tumor antigen protein of the present invention; And a medicament containing a DNA encoding a tumor antigen protein and the like, and a method for diagnosing a tumor or an autoimmune disease.
  • Sequence type nucleic acid
  • Organism Name Homo (Homo sapiens)
  • Tissue type Esophageal cancer tissue
  • AAG AAG AAG TCT TAC ACC ACT CCC AAG AAG AAT AAG CAC AAG AGA AAG 313 Lys Lys Lys Ser Tyr Thr Thr Pro Lys Lys Asn Lys His Lys Arg Lys
  • Sequence type nucleic acid
  • AAG AAG AAG TCT TAC ACC ACT CCC AAG AAG AAT AAG 276 Lys Lys Lys Ser Tyr Thr Thr Pro Lys Lys Asn Lys
  • Sequence type nucleic acid
  • Organism Homo sapiens
  • Tissue type Esophageal cancer tissue LZ
  • AAG AAG CCC AAG AAG GCG GCT GGC GGC GCA ACT CCG AAG AAG AGC GCT 48 Lys Lys Pro Lys Lys Ala Ala Gly Gly Ala Thr Pro Lys Lys Ser Ala
  • AAA GCT GCC AAA ACT GCT GCT AAG GCT GTG AAG CCC AAG GCC GCT AAG 192 Lys Ala Ala Lys Ser Ala Ala Lys Ala Val Lys Pro Lys Ala Ala Lys

Abstract

A tumor antigen protein, a tumor antigen peptide corresponding thereto, or the like, which can be applied to a wide variety of tumors including squamous cell carcinoma and, even when treatable tumors are limited, can be applied to a major part of patients among those suffering from such tumors, or can be applied to various tumors while supplementing this method with tumor immunization or vice versa. They can activate antitumor immunization, treat autoimmune diseases, and provide a diagnostic method for tumors and autoimmune diseases.

Description

明 細 書 ヒト癌退縮抗原タンパク質  Description Human cancer regression antigen protein
技術分野 Technical field
本発明は医療分野に属し、 詳しくは癌または自己免疫疾患を処置する方法、 さ らに詳しくは細胞傷害性 T細胞によつて攻撃を受けて退縮する癌退縮抗原および それを利用する免疫療法等に関する。  The present invention belongs to the medical field, and more specifically, a method for treating cancer or an autoimmune disease, and more specifically, a cancer regression antigen that is regressed by being attacked by cytotoxic T cells, and an immunotherapy using the same. About.
生体による腫瘍の排除には免疫系、 特に T細胞が重要な役割を果たしているこ とが知られている。 実際、 ヒ トの腫瘍局所には腫瘍細胞に対して傷害活性を示す リンパ球の浸潤が認められ (Arch. Surg.126:200-205, 1990) 、 メラノーマから自 己の腫瘍細胞を認識する細胞傷害性 T細胞 (CTL) カ分離されている(Immunol . Today 8:385、 1987、 J. Immunol.138:989, 1987. Int. J. Cancer 52:52-59、 1992 等) 。 また、 T細胞移入によるメラノ一マ治療の臨床結果も腫瘍排除による T細 胞の重要性を示唆している(J.Natl. Cancer. Inst.86: 1159, 1994) 。  It is known that the immune system, especially T cells, plays an important role in eliminating tumors by living organisms. Indeed, infiltration of lymphocytes, which show cytotoxic activity against tumor cells, was observed in human tumors (Arch. Surg. 126: 200-205, 1990), and cells that recognize their own tumor cells from melanoma Toxic T cells (CTL) have been isolated (Immunol. Today 8: 385, 1987, J. Immunol. 138: 989, 1987. Int. J. Cancer 52: 52-59, 1992, etc.). The clinical results of melanoma treatment with T cell transfer also suggest the importance of T cells for tumor elimination (J. Natl. Cancer. Inst. 86: 1159, 1994).
自己の腫瘍細胞を攻撃する CTLは、 腫瘍抗原べプチドが主要組織適合遺伝子 複合体 (MHC) クラス I抗原に結合した複合体を T細胞受容体 (TCR) を用 いて認識し、 自己の腫瘍細胞を攻撃している。 この腫瘍抗原ペプチドは、 腫瘍抗 原 (タンパク質) が細胞内で合成された後、 プロテオソームによって細胞質内で ペプチドに分解されることによって生成される。 一方、 MHCクラス I抗原は、 上記の腫瘍抗原べプチドと結合し、 シスゴルジを経て成熟側のトランスゴルジへ と移動し細胞表面に発現する (臨床免疫 27(9) :1034-1042、 1995) 。 背景技術  CTLs that attack their own tumor cells recognize their complexes of tumor antigen peptides with major histocompatibility complex (MHC) class I antigens using the T cell receptor (TCR), Is attacking. This tumor antigen peptide is produced by the synthesis of tumor antigens (proteins) in cells and the subsequent degradation by proteosomes into peptides in the cytoplasm. On the other hand, MHC class I antigen binds to the above-mentioned tumor antigen peptide, moves to transgolgi on the mature side via cis Golgi, and is expressed on the cell surface (clinical immunity 27 (9): 1034-1042, 1995). Background art
ヒト癌細胞上の MHCクラス I抗原上に提示され、 宿主 T細胞の標的分子とな る腫瘍抗原夕ンパク質が 1991年に T. Boonにより同定された(Science254:1643- 164 7,1991) 。 この抗原は、 この抗原を発現する癌細胞が CTLによって攻撃を受け 退縮することから癌退縮抗原と呼ばれ、 また、 メラノーマ細胞から同定されたこ とより Melanoma antigen (MAGE) と名付けられている。 A tumor antigen protein, presented on MHC class I antigen on human cancer cells and targeted by host T cells, was identified by T. Boon in 1991 (Science 254: 1643-164 7,1991). This antigen causes cancer cells that express this antigen to be attacked by CTLs. It is called a cancer regression antigen because it regresses, and is named Melanoma antigen (MAGE) because it was identified from melanoma cells.
その後、 C T Lにより認識される腫瘍抗原タンパク質がメラノ一マ細胞などか ら相次いで同定された。 今までに同定された腫瘍抗原タンパク質はその由来、 構 造 (変異の有無) や発現様式により以下の 4つのカテゴリ一に分類される(T.Boo n et al., J. Exp. Med.183:725-729, 1996):  Subsequently, tumor antigen proteins recognized by CTL were successively identified from melanoma cells and the like. Tumor antigen proteins identified so far are classified into one of the following four categories according to their origin, structure (presence or absence of mutation) and expression mode (T. Bon et al., J. Exp. Med. 183). : 725-729, 1996):
i ) 腫瘍特異的共有抗原 (Tumor - specific Shared Antigens)  i) Tumor-Specific Shared Antigens
ここに分類される抗原は正常組織では睾丸と胎盤でのみ発現され、 腫瘍組織で はメラノーマ、 頭頸部癌、 非小細胞性肺癌、 膀胱癌など各種の癌に広範に発現が 認められる一群の夕ンパク質である。 このカテゴリ一の腫瘍抗原夕ンパク質とし ては、 上記の MAG E、 その 1 2種類以上の類似するファミ リ一を形成するタン パク質群(J.Exp. Med.178:489- 495, 1993)、 B AGE (Immunity 2:167- 175、 1995 ) および GAGE (J. Exp.Med.182:689- 698.1995)があり、 いづれもメラノ一マ钿 胞から同定されている。 また最近、 メラノ一マに限って広範に発現される NA 1 7— Aが報告された。 それは N—ァセチルグルコサミニルトランスフェラ一ゼ V 遺伝子のィントロンに相当する部分が翻訳され H LA-A2拘束性に抗原べプチ ド (VLPOVF I RC) が癌退縮抗原として発現し CTLにより認識される。 ii) 分化抗原 (Differentiation antigens  The antigens classified here are expressed only in the testis and placenta in normal tissues, and are widely expressed in tumor tissues such as melanoma, head and neck cancer, non-small cell lung cancer, and bladder cancer. It is protein. Examples of this category of tumor antigen proteins include the above-mentioned MAGE and its one or more proteins that form two or more similar families (J. Exp. Med. 178: 489-495, 1993). ), BAGE (Immunity 2: 167-175, 1995) and GAGE (J. Exp. Med. 182: 689-698.1995), all of which have been identified from melanocytic cells. Recently, a widespread expression of NA17-A in melanoma was reported. It is translated from the intron portion of the N-acetylglucosaminyltransferase V gene, and the HLA-A2-restricted antigen peptide (VLPOVF IRC) is expressed as a cancer regression antigen and recognized by CTL. You. ii) Differentiation antigens
ここに分類される腫瘍抗原タンパク質は、 正常組織ではメラノサイ トで発現し ており、 腫瘍組織ではメラノーマでのみ発現が認められる一群の夕ンパク質であ る。 これらの組織特異的なタンパク質は腫瘍細胞のメラノ一マに強度に発現して いる力 他の組織型の癌 (腺癌や扁平上皮癌) には認められないことから、 メラ ノーマに特異的な腫瘍抗原タンパク質と考えられる。 このカテゴリ一の腫瘍抗原 タンパク質としては、 チロシナ一ゼ(J. Exp.Med.178:489- 495, 1993)、 MART— 1 (Proc. Natl. Acad. Sci.USA 91: 3515, 1994) 、 gp 1 0 0 (J. Exp. Med. 179: 1 005-1009, 1994) , g ρ 7 5 (J. Exp. ed. 181 : 799-352, 1995) があり、 これら の遺伝子はいづれもメラノ一マ細胞からクロ一ニングされている。 なお、 他に Me lan- A (J. Exp. Med. 180: 35, 1994) が同定されたが、 後に MA R T— 1 と同一 の分子であることが判明した。 Tumor antigen proteins classified here are expressed in melanocytes in normal tissues and are a group of proteins expressed only in melanomas in tumor tissues. These tissue-specific proteins are strongly expressed in tumor cell melanomas. They are not found in other tissue types of cancer (adenocarcinoma or squamous cell carcinoma). It is considered a tumor antigen protein. Tumor antigen proteins in this category include tyrosinase (J. Exp. Med. 178: 489-495, 1993), MART-1 (Proc. Natl. Acad. Sci. USA 91: 3515, 1994), gp 1 0 0 (J. Exp. Med. 179: 1 005-1009, 1994) and gρ75 (J. Exp. Ed. 181: 799-352, 1995), and these genes are all cloned from melanoma cells. In addition, Melan-A (J. Exp. Med. 180: 35, 1994) was identified, but it was later found to be the same molecule as MART-1.
また、 ここに分類される抗原は正常のメラノサイ トにも発現していることから メラノサイ ト破壊性自己免疫疾患での標的分子としての可能性が存在する。 特に MA R T - 1 Zmelan-Aは von —小柳一原田氏病における標的分子と考えられる (S. Sugi ta, et al., Int. Immunol. 8 :799-803, 1996) 。 g p 1 0 0はそれを発現 するメラノーマが免疫療法に高い感受性を示すので、 in vivoでの癌退縮抗原と して作用している可能性がある。  In addition, since the antigens classified here are also expressed in normal melanosite, there is a possibility that they may be used as target molecules in melanosite-destructive autoimmune diseases. In particular, MART-1 Zmelan-A is considered to be a target molecule in von-Koyanagi-Ichiharada disease (S. Sugita, et al., Int. Immunol. 8: 799-803, 1996). gp100 may act as an in vivo cancer regression antigen because the melanoma expressing it is highly sensitive to immunotherapy.
このカテゴリ一の腫瘍抗原夕ンパク質は、 メラノ一マ以外の腫瘍では発現して いないため、 他の腫瘍に応用することはできない。  This category 1 tumor antigen protein is not expressed in tumors other than melanomas and cannot be applied to other tumors.
i i i ) 個々の腫瘍に特異的な抗原 (antigens Specif ic for Individual T umors )  i i i) Antigens Specific for Individual Tumors
この領域に分類される腫瘍抗原タンパク質は、 正常細胞が癌化する過程でおこ る遺伝子変化に伴う癌特有の新しい抗原である。 遺伝子変化としては点突然変異 (point mutation, 変異 C D K 4抗原、 変異;8 - Catenin 抗原、 MUM 抗原) 、 alterative open reading frame (変異 g p 7 5抗原) 力 これまでに知られ ている。 したがって、 このような抗原は癌に特異的であり特異免疫の成立が容易 に成立するものと考えられる。 し力、しなカ ら、 一方で各々の遺伝子変化は個々の 腫瘍もしくは個々の腫瘍細胞に限って発現していることが殆どである。 したがつ て発現頻度はきわめて低く、 癌治療を目的としたワクチン分子として臨床応用さ れ難いという欠点を有する。  Tumor antigen proteins classified into this region are cancer-specific new antigens associated with genetic changes that occur during the transformation of normal cells into cancer. Gene changes are known to be point mutation (mutant CDK4 antigen, mutation; 8-catenin antigen, MUM antigen) and alterative open reading frame (mutant gp75 antigen). Therefore, it is considered that such an antigen is specific to cancer and that specific immunity is easily established. On the other hand, most of the genetic changes are expressed only in individual tumors or individual tumor cells. Therefore, it has the drawback that its expression frequency is extremely low and it is difficult to apply it clinically as a vaccine molecule for cancer treatment.
iv) 普遍性抗原 (Ubiqui tous Antigens)  iv) Ubiqui tous Antigens
殆どの正常細胞や癌細胞に非変異体として普遍的に発現される (ubiqui tous) 抗原が C T Lの癌認識分子になっているケースとしては p 1 5が知られている。 p 1 5分子は HL A— A 24結合性ペプチドを有する。 p i 5は正常細胞に比し て癌細胞により強く発現されている。 その観点からは、 腺癌等に強発現している 癌遺伝子タンパク質である HER— 2Zn e u抗原も同類として分類される。 即 ち HER— 2/n e uは HLA— A2結合性べプチドを有し癌退縮抗原として宿 主キラ一 T細胞により認識される。 P15 is known as a case in which an antigen that is universally expressed as a non-mutant (ubiqui tous) in most normal cells and cancer cells is a cancer recognition molecule of CTL. The p15 molecule has an HLA-A24 binding peptide. pi5 is more strongly expressed in cancer cells than in normal cells. From that point of view, the HER-2Zneu antigen, which is an oncogene protein that is strongly expressed in adenocarcinoma and the like, is also classified as the same. That is, HER-2 / neu has an HLA-A2-binding peptide and is recognized by host killer T cells as a cancer regression antigen.
ここに分類される抗原を腫瘍抗原夕ンパク質とした場合、 普遍的に発現してい るために広範な癌に応用可能と考えられるが、 疾病特異性に乏しいため、 正常組 織に障害を与える可能性があり、 また CTL誘導が困難である可能性 (トランス のため) が考えられる。  If the antigen classified here is tumor antigen protein, it is considered to be applicable to a wide range of cancers due to its ubiquitous expression, but it will damage normal tissues due to poor disease specificity There is a possibility that CTL induction may be difficult (because of trans).
MHC一非拘束性と考えられていた MU C一 1抗原特異的 C T Lが同抗原由来 ペプチド STAPPAHGVを HLA— A 1 1拘束性に認識すると報告され、 ま た MAGE— 3ぺプチドを用いての臨床試験の初期成績が報告された (M.Marcha nd.et aし, Int. J. Cancer 63:883-885, 1995 ) 。 これらは、 癌退縮抗原を用いて の癌ワクチン開発の可能性を示唆している。  It was reported that MUC-11 antigen-specific CTL, which was thought to be MHC-unrestricted, recognized the peptide derived from the antigen, STAPPAHGV, as HLA-A11-restricted, and clinical studies using MAGE-3 peptide Initial results of the trial were reported (M. Marchand et al., Int. J. Cancer 63: 883-885, 1995). These suggest the possibility of developing a cancer vaccine using a cancer regression antigen.
これまでに同定された上記の抗原べプチドは HER— 2/n e uを除き、 その 殆どがメラノ一マから発見されており、 発病頻度の高い扁平上皮癌や腺癌では全 く報告されていない。 しかし、 我々は最近、 食道扁平上皮癌より HL A 2 6 0 1 拘束性の癌抗原べプチドが存在し宿主 C T Lによって認識されることを報告した (M.Nakao, et aし Cancer Res. 55:4248-4252, 1995) 。 したがって、 扁平上皮 癌にも同様の抗原べプチドをコ一ドする腫瘍抗原夕ンパク質の存在することが示 唆される。 扁平上皮癌は、 ヒトの癌で最も多く認められる癌のひとつであり、 特 に食道癌や肺癌での扁平上皮癌は現在の化学療法や放射線療法に比較的抵抗性を 示すことが知られている。 その点からも腫瘍抗原べプチド等を用いた特異的免疫 療法の開発が期待される。 発明の開示 本発明の課題は、 扁平上皮癌等の幅広い腫瘍に応用でき、 または応用可能な腫 瘍が限られていてもその腫瘍の患者のうち多くの人に応用でき、 またはその腫瘍 の治療や診断を補完しつつ各種腫瘍に応用できる癌療法の一助となる腫瘍抗原べ プチド等を提供することにある。 Most of the above identified antigen peptides except HER-2 / neu have been found in melanomas, and have not been reported in squamous cell carcinomas or adenocarcinomas with high incidence. However, we recently reported that HL A2661-restricted cancer antigen, a peptide, is present and recognized by host CTLs from esophageal squamous cell carcinoma (M. Nakao, et a Cancer Res. 55: 4248-4252, 1995). Therefore, it is suggested that squamous cell carcinoma also has a tumor antigen protein that encodes a similar antigen peptide. Squamous cell carcinoma is one of the most common human cancers.It is known that squamous cell carcinoma in esophageal and lung cancers is relatively resistant to current chemotherapy and radiation therapy. I have. From this point of view, the development of specific immunotherapy using tumor antigen peptides is expected. Disclosure of the invention The object of the present invention is to be applicable to a wide range of tumors such as squamous cell carcinoma, or to a large number of patients with tumors, even if the applicable tumors are limited, or to treat or diagnose such tumors. An object of the present invention is to provide a tumor antigen peptide and the like which assist complementation of cancer and can be applied to various tumors.
また、 腫瘍において高発現している腫瘍抗原タンパク質は、 一方で、 正常組織 にも発現しその腫瘍抗原タンパク質に由来する免疫反応が過剰に起こることで、 自己免疫疾患を引き起こしているとも考えられている。 例えば、 化学療法剤と I L - 2を併用してメラノ一マの治療を行った場合、 白斑症状の出現が認められる との報告がある(J. Cl in. Oncol. 10 : 1338- 1343, 1992)。 これは、 メラノーマに出現 する腫瘍抗原夕ンパク質の断片べプチドと M H C複合体に対して C T Lまたは抗 体が誘導、 産生され、 正常組織である皮膚組織に作用することで自己免疫疾患様 の症状である白斑症状が出現したためと考えられる。  On the other hand, tumor antigen proteins that are highly expressed in tumors, on the other hand, are also thought to be expressed in normal tissues and cause an autoimmune disease due to excessive immune responses derived from the tumor antigen proteins. I have. For example, it has been reported that when melanomas are treated with a combination of a chemotherapeutic agent and IL-2, vitiligo symptoms appear (J. Clin. Oncol. 10: 1338-1343, 1992). ). This is because CTLs or antibodies are induced and produced against the tumor antigen protein fragment peptide and MHC complex appearing in melanoma, and they act on normal skin tissue, causing symptoms similar to autoimmune disease. This is probably due to the appearance of vitiligo symptoms.
腫瘍抗原タンパク質に由来する特異的免疫が過剰に惹起されることにより自己 免疫疾患が発症した場合には、 腫瘍抗原タンパク質をコ一ドする遺伝子の発現を 妨ぐアンチセンス DNA や腫瘍抗原ペプチドのアン夕ゴニストなどを用いて、 免疫 反応を特異的にプロックする治療法が期待される。  When an autoimmune disease develops due to excessive induction of specific immunity derived from a tumor antigen protein, the antisense DNA or tumor antigen peptide that prevents expression of a gene that encodes the tumor antigen protein is blocked. A therapeutic method that specifically blocks the immune response using evening gonists is expected.
メラノ一マ細胞以外の腫瘍細胞、 特に扁平上皮癌等の治療や診断に幅広く応用 できる腫瘍抗原タンパク質またはその対応する腫瘍抗原べプチド等を得るために 、 扁平上皮癌からの腫瘍抗原タンパク質の同定を試みた。  Identification of tumor antigen proteins from squamous cell carcinomas to obtain tumor antigen proteins or their corresponding tumor antigen peptides that can be widely applied to the treatment and diagnosis of tumor cells other than melanoma cells, especially squamous cell carcinoma, etc. Tried.
本発明者らは食道癌患者の転移性リンパ節癌組織浸潤リンパ球から HLA- CW0102 拘束性の腫瘍抗原ペプチドを認識する C T L (YAM- CTL) を樹立した。 この C T L は HLA- CW0102陽性の食道癌細胞株 TE8 (熊本大学医学部) を強く障害する。  The present inventors have established CTL (YAM-CTL) that recognizes an HLA-CW0102-restricted tumor antigen peptide from metastatic lymph node cancer tissue infiltrating lymphocytes of esophageal cancer patients. This CTL strongly impairs the HLA-CW0102-positive esophageal cancer cell line TE8 (Kumamoto University School of Medicine).
そこで、 繊維芽細胞株 C0S7細胞に、 TE8 癌細胞株から作成した cDNAライブラリ 一の組換えプラスミ ドと HLA-CW0102 cDNA の組換えプラスミ ドを同時にトランス フエク トし、 そのトランスフユクタントに YAM- CTL 細胞を作用させ、 YAM-CTL 細 胞が活性化されたか否かを IFN- 7の産生量で測定しスクリーニングした。 その結 果、 メラノ一マ以外の腫瘍細胞 TE8 から本発明の腫瘍抗原タンパク質をコードす る遺伝子をクロ一ユングすることに成功した。 Therefore, the recombinant plasmid of the cDNA library and the recombinant plasmid of the HLA-CW0102 cDNA created from the TE8 cancer cell line were simultaneously transfected into the fibroblast cell line C0S7 cells, and YAM-CTL was added to the transfectant. The cells were allowed to act, and whether or not the YAM-CTL cells were activated was screened by measuring the amount of IFN-7 produced. The result As a result, a gene encoding the tumor antigen protein of the present invention was successfully cloned from tumor cells other than melanoma TE8.
すなわち、 本発明は一つの態様として、 配列番号: 2のアミノ酸配列を有する 腫瘍抗原夕ンパク質の部分べプチドであって主要組織適合遺伝子複合体 (MH C ) クラス I抗原と結合して T細胞により認識される腫瘍抗原べプチドまたはその 誘導体;本発明の腫瘍抗原べプチドまたはその誘導体を含有する医薬;本発明の 腫瘍抗原べプチドまたはその誘導体に対する抗体;本発明の腫瘍抗原べプチドま たはその誘導体をコ一ドするオリゴヌクレオチド分子、 好ましくは腫瘍抗原夕ン パク質をコードしている塩基配列が配列番号: 2の塩基配列である該ォリゴヌク レオチド分子;配列番号: 2の腫瘍抗原タンパク質をコ一ドするポリヌクレオチ ド分子のコーディング配列またはその 5 ' ノンコーディング配列中の塩基配列と 相補的な配列からなるオリゴヌクレオチド分子またはその化学的修飾体;本発明 のォリゴヌクレオチド分子を含有する医薬;本発明のォリゴヌクレオチド分子を 有するプラスミ ド;および本発明のプラスミ ドによって形質転換された形質転換 体、 に関する。  That is, in one embodiment, the present invention relates to a partial peptide of a tumor antigen protein having an amino acid sequence of SEQ ID NO: 2, which binds to a major histocompatibility complex (MH C) class I antigen and has T cells. A tumor antigen peptide or a derivative thereof, a drug containing the tumor antigen peptide or a derivative thereof according to the present invention; an antibody against the tumor antigen peptide or a derivative thereof of the present invention; a tumor antigen peptide or the present invention An oligonucleotide molecule encoding the derivative thereof, preferably the oligonucleotide molecule whose nucleotide sequence encoding a tumor antigen protein is the nucleotide sequence of SEQ ID NO: 2; the tumor antigen protein of SEQ ID NO: 2; Consists of the coding sequence of the coding polynucleotide molecule or a sequence complementary to the base sequence in its 5 'non-coding sequence An oligonucleotide molecule or a chemically modified product thereof; a medicament containing the oligonucleotide molecule of the present invention; a plasmid having the oligonucleotide molecule of the present invention; and a transformant transformed with the plasmid of the present invention. .
別の態様として、 本発明は、 配列番号: 4のアミノ酸配列を有する腫瘍抗原タ ンパク質の部分ペプチドであって主要組織適合遺伝子複合体 (MH C ) クラス I 抗原と結合して T細胞により認識される腫瘍抗原べプチドまたはその誘導体;本 発明の腫瘍抗原べプチドまたはその誘導体を含有する医薬;本発明の腫瘍抗原べ プチドまたはその誘導体に対する抗体;本発明の腫瘍抗原べプチドまたはその誘 導体をコードするオリゴヌクレオチド分子、 好ましくは腫瘍抗原夕ンパク質をコ 一ドしている塩基配列が配列番号: 4の塩基配列である該ォリゴヌクレオチド分 子;配列番号: 4の腫瘍抗原夕ンパク質をコ一ドするポリヌクレオチド分子のコ 一ディング配列またはその 5 ' ノンコ一ディング配列中の塩基配列と相補的な配 列からなるォリゴヌクレオチド分子またはその化学的修飾体;本発明のォリゴヌ クレオチド分子を含有する医薬;本発明のォリゴヌクレオチド分子を有するブラ スミ ド;および本発明のプラスミ ドによって形質転換された形質転換体、 に関す る。 図面の簡単な説明 In another embodiment, the present invention relates to a partial peptide of a tumor antigen protein having the amino acid sequence of SEQ ID NO: 4, which is recognized by T cells by binding to a major histocompatibility complex (MHC) class I antigen. A tumor antigen peptide or a derivative thereof to be used; a medicament containing the tumor antigen peptide or a derivative thereof of the present invention; an antibody against the tumor antigen peptide of the present invention or a derivative thereof; and a tumor antigen peptide of the present invention or a derivative thereof. An oligonucleotide molecule that encodes, preferably the oligonucleotide nucleotide sequence whose nucleotide sequence encodes a tumor antigen protein; SEQ ID NO: 4; An oligonucleotide consisting of a sequence complementary to the base sequence in the coding sequence of the coding polynucleotide molecule or its 5 'non-coding sequence. Reochido molecule or chemical modifications thereof; pharmaceutical containing Origonu Kureochido molecules of the present invention; bra with O oligo nucleotide molecule of the present invention And a transformant transformed with the plasmid of the present invention. BRIEF DESCRIPTION OF THE FIGURES
第 1図は、 クローン 3から得た断片 1および 2のアミノ酸配列を示す。 下線は 断片 1、 太字斜線は断片 2を示す。  FIG. 1 shows the amino acid sequences of fragments 1 and 2 obtained from clone 3. Underline indicates fragment 1 and bold diagonal line indicates fragment 2.
第 2図は、 クローン 3から得た断片 1および 2による IFN- 7産生量を EU SA に よって測定したグラフである。 O R Fはオープンリーディ ングフレームを、 F 1 は断片 1を、 F 2は断片 2を示す。 また (―) は同時トランスフヱク 卜していな いもの、 A 2 4は HLA-A24 cDNA の組換えプラスミ ドを同時トランスフエク トし たもの、 C w lは、 HLA-Cw0102 cDNA の組換えプラスミ ドを同時トランスフエク トしたものを示す。 FIG. 2 is a graph showing the amount of IFN- 7 produced by fragments 1 and 2 obtained from clone 3 measured by EUSA. ORF indicates open reading frame, F 1 indicates fragment 1, and F 2 indicates fragment 2. (-) Indicates that the cotransfected plasmid was not transfected, A24 indicates the cotransfected plasmid of HLA-A24 cDNA, and Cwl indicates the recombinant plasmid of HLA-Cw0102 cDNA. The result of simultaneous transfection is shown.
第 3図は、 クローン 6から得た断片 1、 2および 3のアミノ酸配列を示す。 下 線は断片 1を、 太字斜体は断片 2を、 二重線は断片 3を示す。  FIG. 3 shows the amino acid sequences of fragments 1, 2 and 3 obtained from clone 6. Underline indicates fragment 1, bold italic indicates fragment 2, double line indicates fragment 3.
第 4図は、 クローン 6から得た断片 1、 2および 3による IFN- 7産生量を ELIS A によって測定したグラフである。 O R Fはオープンリーディ ングフレームを、 F 1は断片 1を、 F 2は断片 2を、 F 3は断片 3を示す。 また (一) は同時トラ ンスフェク トしていないもの、 A 2 4は HLA- A24 cDNA の組換えプラスミ ドを同 時トランスフヱク トしたもの、 C w lは、 HLA- Cw0102 cDNA の組換えプラスミ ド を同時トランスフ ク トしたものを示す。 発明を実施するための最良の形態  FIG. 4 is a graph showing the amount of IFN-7 produced by fragments 1, 2 and 3 obtained from clone 6 measured by ELIS A. ORF indicates the open reading frame, F1 indicates fragment 1, F2 indicates fragment 2, and F3 indicates fragment 3. (1) is not simultaneously transfected, A24 is a simultaneously transfected recombinant plasmid of HLA-A24 cDNA, and Cwl is a simultaneously transfected plasmid of HLA-Cw0102 cDNA. Shows what was transfected. BEST MODE FOR CARRYING OUT THE INVENTION
本明細書中で使用している用語の意義を明らかにするとともに、 発明の実施形 態を説明する。  The meaning of the terms used in this specification will be clarified, and embodiments of the invention will be described.
「腫瘍抗原夕ンパク質の部分べプチドであって主要組織適合遺伝子複合体 (M H C ) クラス I抗原と結合して T細胞により認識される腫瘍抗原ペプチド」 とは 、 腫瘍抗原タンパク質の少なくとも 7〜 1 0個、 好ましくは 9個の連続するアミ ノ酸配列からなる部分べプチドであって、 細胞表面の MH Cクラス I抗原と結合 して細胞表面に提示された場合、 その結合体に対して特異的に結合する T細胞が 結合するとその T細胞にシグナルを伝えることのできる、 即ち T細胞に認識され る MH Cクラス I抗原との結合体を形成できるそのようなぺプチドを意味する。 なお、 ここでいう 「結合」 とは非共有結合である。 "Partial peptide of tumor antigen protein, tumor antigen peptide recognized by T cells by binding to major histocompatibility complex (MHC) class I antigen" A partial peptide consisting of at least 7-10, preferably 9 contiguous amino acid sequences of a tumor antigen protein, which is presented on the cell surface in association with the MHC class I antigen on the cell surface In this case, when a T cell that specifically binds to the conjugate binds, it can transmit a signal to the T cell, that is, form a conjugate with the MHC class I antigen recognized by the T cell. Means peptide. The “bond” here is a non-covalent bond.
ぺプチドが MH Cクラス I抗原に結合して T細胞に認識されることを確かめる 方法としては、 例えば、 ペプチドを適当な細胞に内因性に発現させるか、 または 外部から加える (パルスする) ことにより MH Cクラス I抗原に結合させること で細胞表面にペプチドを提示させ、 つづいて、 そのペプチド提示細胞に対して腫 瘍抗原夕ンパク質特異的な T細胞を作用させ、 そのべプチド提示細胞が傷害を受 けた際に産生されるサイ ト力イン (インターフェロンひや TN F 、 および C T Lが産生するサイ ト力イン) を測定する方法などがある。 また、 ペプチド提示細 胞の傷害を測定する方法として、 5 1 C rで標識したぺプチド提示細胞を用いる方 法も使用できる。 ここで、 認識する T細胞としては、 C T Lを用いるのが好まし い。 Methods for confirming that a peptide binds to MHC class I antigen and is recognized by T cells include, for example, endogenous expression of the peptide in appropriate cells, or by externally adding (pulsing) the peptide. The peptide is presented on the cell surface by binding to the MHC class I antigen, followed by the action of tumor antigen protein-specific T cells on the peptide-presenting cells, which damage the peptide-presenting cells There is a method to measure the site force in produced when receiving the drug (interferon, TNF, and CTL). Further, as a method for measuring the cytotoxicity of peptide presentation cells, it can also be used how to use a peptide-presenting cells labeled with 5 1 C r. Here, CTLs are preferably used as T cells to be recognized.
本発明に係る腫瘍抗原タンパク質または腫瘍抗原べプチドは例えば、 以下のよ うにして同定することができる。  The tumor antigen protein or tumor antigen peptide according to the present invention can be identified, for example, as follows.
まず、 これらの同定に際し、 MH C —クラス Iアレルの一致した腫瘍細胞およ びこの細胞を攻撃する C T Lのセッ トを用意する。  To identify them, first prepare a set of tumor cells with matching MHC-class I alleles and CTLs that attack these cells.
次いで、 腫瘍細胞の MH Cクラス I抗原に結合している腫瘍抗原ペプチドを酸 性化して抽出し、 高速液体クロマトグラフィーで分離された種々のべプチドを、 抗原提示 MH Cを発現しているが腫瘍抗原タンパク質を発現していない細胞 (例 えば、 同一患者の B細胞など) にパルスし、 C T Lの反応を調べることにより腫 瘍抗原べプチドを同定し、 さらにマススぺク トロメ夕リ一などを用いて配列を決 定する方法である。 この方法によって、 メラノ一マ細胞から g p 1 0 0と同一分 子の Pmell7由来の腫瘍抗原べプチドが同定されている(Science264:716- 719, 1994Next, the tumor antigen peptide that binds to the MHC class I antigen of the tumor cells is oxidized and extracted, and various peptides separated by high-performance liquid chromatography express antigen-presenting MHC. By pulsing cells that do not express the tumor antigen protein (for example, B cells of the same patient), and examining the CTL response, the tumor antigen peptide was identified, and then mass spectrometry was performed. This method is used to determine the sequence. By this method, the same amount of gp100 from melanoma cells A tumor antigen peptide derived from offspring Pmell7 has been identified (Science264: 716-719, 1994).
) 0 ) 0
あるいは、 上記のような腫瘍抗原ペプチドを直接同定する方法とは異なり、 腫 瘍抗原タンパク質をコードする遺伝子を決定してさらにその対応する腫瘍抗原べ プチドを同定する方法もある。 これは、 分子生物学的手法を用いて腫瘍抗原タン パク質をコードする遺伝子をクローニングするものである。 腫瘍細胞から cDNAを 調製し、 その cDNAを腫瘍抗原タンパク質を発現していない細胞 (例えば COS細 胞など) に抗原提示 MHCクラス I抗原遺伝子とともにトランスフ ク トして一 過的にそれらを発現させ、 それに対する CTLの反応性によるスクリ一二ングを 繰り返し行い、 腫瘍抗原タンパク質をコードする遺伝子を単離する。 この方法に より、 上記の MAG E、 チロシナ一ゼ、 MART— 1、 gp l 0 0、 gp 75の 遺伝子がクローニングされている。  Alternatively, unlike the method for directly identifying a tumor antigen peptide as described above, there is also a method for determining a gene encoding a tumor antigen protein and further identifying the corresponding tumor antigen peptide. This involves cloning the gene encoding the tumor antigen protein using molecular biology techniques. CDNA is prepared from tumor cells, and the cDNA is transfected with cells that do not express the tumor antigen protein (for example, COS cells) together with the antigen-presenting MHC class I antigen gene to express them temporarily. Screening based on the reactivity of the CTL against it is repeated to isolate the gene encoding the tumor antigen protein. By this method, the above-mentioned genes of MAGE, tyrosinase, MART-1, gp100, and gp75 have been cloned.
この腫瘍抗原遺伝子の情報から実際に MH Cクラス I抗原に結合して提示され ている腫瘍抗原ペプチドを推定、 同定するためには次のような方法を用いる。 ま ず、 PCR、 ェキソヌクレアーゼ、 制限酵素などにより様々なサイズの腫瘍抗原 タンパク質をコ一ドする遺伝子のフラグメントを作製し、 抗原提示 M H Cクラス I抗原遺伝子とともに腫瘍抗原タンパク質を発現していない細胞 (例えば COS 細胞など) にトランスフ ク トして一過性に発現させ、 CTLの反応性により腫 瘍抗原ペプチドを含む領域を限定する。 その後、 ペプチドを合成し、 抗原提示 M HCクラス I抗原は発現しているが腫瘍抗原夕ンパク質を発現していない細胞に パルスし、 c τ Lの反応を調べることにより腫瘍抗原べプチドを同定できる . E xp.Med. 176: 1453, 1992、 J. Exp. Med.179:24, 759、 1994) 。  The following method is used to estimate and identify the tumor antigen peptide actually bound to and presented to the MHC class I antigen from the information on the tumor antigen gene. First, fragments of genes encoding tumor antigen proteins of various sizes were prepared by PCR, exonuclease, restriction enzymes, etc., and cells that did not express tumor antigen proteins together with antigen-presenting MHC class I antigen genes ( For example, transfected into COS cells, etc., and transiently expressed, and the region containing the tumor antigen peptide is limited by the reactivity of CTL. After that, the peptide was synthesized, and the antigen-presenting MHC class I antigen was expressed, but it was pulsed into cells that did not express the tumor antigen protein, and the tumor antigen peptide was identified by examining the reaction of cτL. Med. 176: 1453, 1992, J. Exp. Med. 179: 24, 759, 1994).
また、 HLA- Al, -A0201, -A0205, -All, A31 , - A6801, -B7 , -B8 , -B2705, -37, -CW0401 , -CW0602 などの MH Cクラス I抗原の型については、 結合して 提示されるペプチドの配列の規則性 (モチーフ) が判明しており(seminars in IMMUNOLOGY 5:81-94,1993)、 それを参考にして腫瘍抗原ペプチドの候補を調べ 、 そのべプチドを結合して上記と同様な方法で確認する方法も用いられる(Eur. J . Immunol, 24 : 759, 1994, J. Exp. Med. 180 : 347, 1994)。 For the types of MHC class I antigens such as HLA-Al, -A0201, -A0205, -All, A31, -A6801, -B7, -B8, -B2705, -37, -CW0401, and -CW0602, binding The sequence regularities (motifs) of the peptides presented are known (seminars in IMMUNOLOGY 5: 81-94, 1993), and the tumor antigen peptide candidates are examined with reference to them. A method in which the peptide is bound and confirmed by the same method as described above is also used (Eur. J. Immunol, 24: 759, 1994, J. Exp. Med. 180: 347, 1994).
この様にして決定されたべプチドは、 通常のぺプチド化学において知られてい る方法で製造することができる。 例えば、 "Pept ide Synthesi s" , Interscienc e, New York. 1996、 "The Proteins", Vol. 2, Academic Press Inc. , New Y ork. 1976. 「ペプチド合成」 丸善( 株) 、 1975、 「ペプチド合成の基礎と実験」 丸善(株) 、 1985、 等に記載されている方法等が挙げられる。 すなわち、 C末端 部位の構成により液相法、 固相法のいずれかを選択して合成することができ、 な かでも液相法がより好ましい。 すなわち、 アミノ酸の官能基を適当な保護基で適 宜保護および脱保護を行い、 アミノ酸を、 残基または数残基づつ結合させること でペプチドを製造することができる。 なお、 アミノ酸の官能基の保護基について は、 例えば前述のぺプチド化学について記載する書籍等に記載されている。  The peptides determined in this way can be produced by methods known in normal peptide chemistry. For example, "Peptide Syntheses", Interscience, New York. 1996, "The Proteins", Vol. 2, Academic Press Inc., New York. 1976. "Peptide synthesis" Maruzen Co., Ltd., 1975, "Peptide Synthesis Basics and Experiments ", Maruzen, 1985, and the like. That is, either the liquid phase method or the solid phase method can be selected for synthesis depending on the configuration of the C-terminal site, and the liquid phase method is more preferable. That is, a peptide can be produced by appropriately protecting and deprotecting a functional group of an amino acid with an appropriate protecting group, and bonding the amino acid to each residue or several residues. The protective group for the functional group of the amino acid is described, for example, in the above-mentioned book describing peptide chemistry.
本明細書中、 「本発明の腫瘍抗原べプチドの誘導体 (腫瘍抗原タンパク質の部 分べプチドであって MH Cクラス I抗原と結合して T細胞により認識される腫瘍 抗原ペプチドの誘導体) 」 とは、 本発明の腫瘍抗原ペプチドのアミノ酸配列のう ち 1つまたは数個が置換、 欠失、 挿入または付加されたペプチドを意味する。 好 ましい誘導体としては、 腫瘍抗原べプチドのうちで C T Lとの結合に関与するェ ピトープ領域はそのままであって MH Cクラス I抗原との結合に関与するアミノ 酸残基が置換、 欠失、 挿入または付加された誘導体が挙げられ、 さらに好ましく はその誘導体であって一つのァミノ酸残基のみを置換したものが挙げられる (Im munol. 84 :298-303, 1995 ) 。 かかる誘導体は、 C T Lとの結合性はそのまま維持 しつつ、 MH Cクラス I抗原により強く結合可能であるため、 さらに有用な腫瘍 抗原べプチドとして適用ができる。  In the present specification, “a derivative of the tumor antigen peptide of the present invention (a derivative of a tumor antigen peptide that is a partial peptide of a tumor antigen protein and that is recognized by T cells by binding to an MHC class I antigen)” Means a peptide in which one or several amino acid sequences of the tumor antigen peptide of the present invention have been substituted, deleted, inserted or added. Preferred derivatives include the tumor antigen peptides, in which the epitope region involved in binding to CTL remains unchanged and amino acid residues involved in binding to MHC class I antigen are substituted, deleted, or deleted. Inserted or added derivatives can be mentioned, and more preferably, derivatives thereof, in which only one amino acid residue is substituted (Im munol. 84: 298-303, 1995). Such a derivative can bind strongly to the MHC class I antigen while maintaining the binding property to CTL as it is, so that it can be applied as a more useful tumor antigen peptide.
このような誘導体は、 例えば Molecular Cloning : A Laboratory Manual第 2 版第 1 一 3巻 Sambrook, J. ら著、 Cold Spring Harber Labolatory Press 出 版 New York 1989 年に記載の方法で調製することができ、 部位特異的変異誘発 や P C R法などの方法によって調製することができる。 Such a derivative can be prepared, for example, by the method described in Molecular Cloning: A Laboratory Manual, 2nd edition, Volume 13, Sambrook, J. et al., Cold Spring Harber Labolatory Press, New York, 1989. Site-directed mutagenesis And can be prepared by a method such as PCR.
従って、 本発明の腫瘍抗原ペプチドまたはその誘導体は、 後述の本発明のオリ ゴヌクレオチドによりコードされるものである。  Therefore, the tumor antigen peptide of the present invention or a derivative thereof is encoded by the below-described oligonucleotide of the present invention.
また、 「本発明の誘導体」 には、 本発明の腫瘍抗原ペプチドまたは該ペプチド の一部のアミノ酸残基を置換、 欠失、 挿入または付加した誘導体のアミノ基もし くはカルボキシル基を修飾した誘導体も包含される。  In addition, the “derivative of the present invention” includes a derivative of the tumor antigen peptide of the present invention or a derivative in which a partial amino acid residue of the peptide is substituted, deleted, inserted, or added, wherein the amino group or the carboxyl group is modified. Are also included.
ァミノ基の修飾基としては、 例えばァシル基が挙げられ、 具体的には炭素数 1 から 6のアルカノィル基、 フエニル基で置換された炭素数 1力、ら 6のアルカノィ ル基、 炭素数 5から 7のシクロアルキル基で置換されたカルボニル基、 炭素数 1 から 6のアルキルスルホニル基、 フヱニルスルホニル基等が挙げられる。  Examples of the modifying group for the amino group include an acyl group.Specific examples include an alkanol group having 1 to 6 carbon atoms, 1 carbon atom substituted by a phenyl group, an alkanol group having 6 or more carbon atoms, and 5 to 5 carbon atoms. A carbonyl group substituted with 7 cycloalkyl groups, an alkylsulfonyl group having 1 to 6 carbon atoms, a phenylsulfonyl group, and the like.
カルボキシル基の修飾基としては、 例えばエステル基およびアミ ド基が挙げら れ、 エステル基の具体例としては、 炭素数 1かる 6のアルキルエステル基、 フエ ニル基で置換された炭素数 0力、ら 6のアルキルエステル基、 炭素数 5から 7のシ クロアルキルエステル基等が挙げられ、 アミ ド基の具体例としては、 アミ ド基、 炭素数 1から 6のアルキル基 1つまたは 2つで置換されたアミ ド基、 フヱニル基 で置換された炭素数 0から 6のアルキル基 1つまたは 2つで置換されたァミ ド基 、 アミ ド基の窒素原子を含んで 5から 7員環のァザシクロアルカンを形成するァ ミ ド基等が挙げられる。  Examples of the carboxyl group-modifying group include an ester group and an amide group. Specific examples of the ester group include an alkyl ester group having 1 to 6 carbon atoms, a carbon atom having 0 carbon atoms substituted with a phenyl group, Examples of the amide group include an amide group, one or two alkyl groups having 1 to 6 carbon atoms, and a cycloalkyl ester group having 5 to 7 carbon atoms. Substituted amide group, phenyl group-substituted alkyl group having 0 to 6 carbon atoms, amide group substituted with 1 or 2 carbon atoms, 5- to 7-membered ring including amide group nitrogen atom Amide groups forming azacycloalkanes and the like can be mentioned.
本発明はさらに、 本発明の腫瘍抗原べプチドまたはその誘導体を含有する医薬 を提供する。 本発明の腫瘍抗原タンパク質および腫瘍抗原ペプチドは、 細胞性免 疫が効果的に成立するようにアジュバントとともに投与したり、 粒子状の剤型に して投与することができる。 アジュバントとしては、 文献 (Cl in. Microbi ol. R ev. 7:277-289. 1994) に記載のものなどが応用可能である。 また、 剤型としては 、 リボソーム製剤、 直径数 m のビーズに結合させた粒子状の製剤、 リピッ トを 結合させた製剤など外因性の抗原べプチドを MH Cクラス I抗原へ効率良く抗原 提示させうる投与法が用いられる。 また、 腫瘍抗原ペプチドをパルスした樹状紬 胞ゃマイクロファ一ジなどの抗原提示細胞や腫瘍抗原夕ンパク質をコ一ドする DN A を導入した細胞を投与する方法も考えられる。 製剤中の本発明の腫瘍抗原タン パク質および腫瘍抗原ペプチドの投与量は、 治療目的の疾患、 患者の年齢、 体重 等により適宜調整することができるが、 通常 0. 0001nig〜1000nig、 好ましくは 0. 00 lmg〜1000mgであり、 これを数日ないし数月に 1 回投与するのが好ましい。 The present invention further provides a medicament containing the tumor antigen peptide of the present invention or a derivative thereof. The tumor antigen protein and the tumor antigen peptide of the present invention can be administered together with an adjuvant so as to effectively establish cellular immunity, or can be administered in the form of particles. As the adjuvant, those described in the literature (Cl in. Microbiol. Rev. 7: 277-289. 1994) can be applied. In addition, dosage forms include ribosome preparations, particulate preparations bound to beads with a diameter of several meters, and preparations linked to lipids to efficiently present antigen to MHC class I antigens. Any method of administration is used. In addition, dendritic pongee pulsed with tumor antigen peptide It is also conceivable to administer antigen-presenting cells such as vesicle microphages or cells transfected with DNA encoding tumor antigen protein. The dosage of the tumor antigen protein and tumor antigen peptide of the present invention in the preparation can be appropriately adjusted depending on the disease to be treated, the age and weight of the patient, etc., and is usually 0.0001 nig to 1000 nig, preferably 0. .000 mg to 1000 mg, preferably administered once every several days to several months.
本発明の腫瘍抗原ペプチドまたはその誘導体に対する 「抗体」 は、 例えば、 An t ibodies ; A Laboratory Manual, Lane. H. D.ら編、 Cold Spring Harber Laboratory Press 出版 New York 1989 年などに記載の方法により、 腫瘍抗原 夕ンパク質またはその断片べプチドを用いて適切な方法で適切な動物を免疫する ことにより、 腫瘍抗原タンパク質を認識する抗体、 あるいはその活性を中和する 抗体を容易に作成できる。 抗体の用途としては、 ァフィ二ティ一クロマトグラフ ィー、 cDNAライブラリーのスクリーニング、 免疫学的診断法、 医薬等が挙げられ る。 免疫学的診断法は、 ィムノブロッ ト法、 放射免疫測定法 (R I A) 、 酵素免 疫測定法 (E L I S A) 、 蛍光あるいは発光測定法等より適宜選択できる。  The “antibody” against the tumor antigen peptide or a derivative thereof according to the present invention may be, for example, a tumor antigen by a method described in Antibodies; A Laboratory Manual, Lane. HD et al., Edited by Cold Spring Harber Laboratory Press, New York 1989. By immunizing an appropriate animal with an appropriate method using the protein or its fragment peptide, an antibody that recognizes a tumor antigen protein or an antibody that neutralizes its activity can be easily prepared. Uses of the antibody include affinity chromatography, screening of a cDNA library, immunological diagnostic methods, pharmaceuticals and the like. The immunological diagnostic method can be appropriately selected from an immunoblot method, a radioimmunoassay method (RIA), an enzyme immunoassay method (ELISA), a fluorescence or luminescence measurement method, or the like.
本発明はさらに、 本発明の腫瘍抗原べプチドまたはその誘導体をコ一ドするォ リゴヌクレオチド分子に関する。 本発明のォリゴヌクレオチド分子は DNA または RNA の形態をとることができ、 DNA には cDNA、 ゲノム DNA および合成 DNA が包含 される。 また、 DNA および RNA は 1本鎖または 2本鎖であってよく、 1本鎖の場 合はセンス鎖またはアンチセンス鎖の両者が包含され得る。  The present invention further relates to an oligonucleotide molecule encoding the tumor antigen peptide of the present invention or a derivative thereof. The oligonucleotide molecules of the present invention can be in the form of DNA or RNA, where DNA includes cDNA, genomic DNA and synthetic DNA. Also, DNA and RNA may be single-stranded or double-stranded, and in the case of single-stranded, may include both the sense strand and the antisense strand.
ある塩基配列のうち一部が置換、 欠失、 挿入または付加されたオリゴヌクレオ チド分子は、 Molecular Cloning :A Laboratory Manual第 2版第 1— 3巻 Samb rook, J. ら著、 Cold Spring Harber Labolatory Press 出版 New York 198 9 年などに記載の方法によって製造することができ、 例えば部位特異的変異誘発 や P C R法などにより製造できる。 本発明のオリゴヌクレオチド分子はこれらの 変異型ォリゴヌクレオチド分子も包含する。 かかる変異型ォリゴヌクレオチド分 子としては、 例えば、 配列番号: 2または 4の塩基配列において 1 もしくは複数 個の塩基が置換、 欠失、 挿入または付加されたオリゴヌクレオチド分子が挙げら れる。 また、 本発明のオリゴヌクレオチド分子には 「本発明のオリゴヌクレオチ ド分子にストリンジヱン卜な条件下でハイプリダイズするオリゴヌクレオチド分 子」 も包含される。 オリゴヌクレオチド分子として DNA分子を代表例にとると、 「DNA分子にハイブリダィズする DNA分子」 は、 例えば前述の Molecular Cloni ngに記載の方法によって得ることができる。 ここで、 「ストリンジヱン卜な条件 下でハイブリダィズする」 とは、 例えば、 6 XSSC, 0. 5 %SDSおよび 5 0 %ホルムアミ ドの溶液中で 42°Cにて加温した後、 0. 1 XSSC、 0. 5 % SDSの溶液中で 6 8 °Cにて洗浄する条件でも依然として陽性のハイブリダイズ のシグナルが観察されることを表す。 Oligonucleotide molecules in which a part of a certain nucleotide sequence has been substituted, deleted, inserted or added are described in Molecular Cloning: A Laboratory Manual, 2nd edition, Volume 1-3, Sambrook, J. et al., Cold Spring Harber Labolatory It can be produced by the method described in Press Publishing New York, 1980. For example, it can be produced by site-directed mutagenesis or PCR. The oligonucleotide molecules of the present invention also include these mutant oligonucleotide molecules. Examples of such mutant oligonucleotide nucleotides include, for example, one or more nucleotides in the nucleotide sequence of SEQ ID NO: 2 or 4. Oligonucleotide molecules in which three bases have been substituted, deleted, inserted or added. The oligonucleotide molecules of the present invention also include "oligonucleotide molecules that hybridize to the oligonucleotide molecules of the present invention under stringent conditions". Taking a DNA molecule as a typical example of the oligonucleotide molecule, “a DNA molecule that hybridizes to a DNA molecule” can be obtained, for example, by the method described in the aforementioned Molecular Cloning. Here, “hybridizing under stringent conditions” means, for example, heating at 42 ° C. in a solution of 6 XSSC, 0.5% SDS and 50% formamide, followed by 0.1 XSSC Under the condition of washing at 68 ° C. in a 0.5% SDS solution, a positive hybridization signal was still observed.
また、 本発明の腫瘍抗原ペプチドをコードする DNA を発現させることによって 、 腫瘍抗原ペプチドを大量に製造することが可能となる。  In addition, by expressing the DNA encoding the tumor antigen peptide of the present invention, it becomes possible to produce the tumor antigen peptide in large quantities.
DNA を発現してタンパク質を生産するには、 例えば、 前述の Molecular Cloni ng等の多くの成書や文献に基づいて実施することができる。 発現させたい DNA の 上流に翻訳開始コドンを、 下流には翻訳終始コドンを付加し、 転写を制御するプ ロモ一夕一配列 (例えば、 t r p、 l a c、 T7、 S V 4 0初期プロモーター) 等の制御遺伝子を付加し、 適当なベクター (例えば、 p BR 322、 pUC 1 9 、 pSV · SPORT 1など) に組み込むことにより、 宿主細胞内で複製し、 機 能する発現プラスミ ドを作製する。 次に発現プラスミ ドを適当な宿主細胞に導入 して形質転換体細胞を得る。 宿主細胞としては、 大腸菌などの原核生物、 酵母の ような単細胞真核生物、 昆虫、 動物などの多細胞真核生物の細胞などが挙げられ る。 また、 宿主細胞への遺伝子導入法としては、 リン酸カルシウム法、 DEAE ーデキストラン法、 電気パルス法などがある。 形質転換体は、 適当な培地で培養 することによって目的とするタンパク質を生産する。 以上のようにして得られた タンパク質は一般的な生化学的方法によって単離精製することができる。  Expression of DNA to produce a protein can be carried out, for example, based on many books and literature such as the aforementioned Molecular Cloning. A translation initiation codon is added upstream of the DNA to be expressed and a translation stop codon is added downstream to control the promoter sequence (eg, trp, lac, T7, SV40 early promoter) that controls transcription. By adding the gene and incorporating it into an appropriate vector (for example, pBR322, pUC19, pSV • SPORT1, etc.), an expression plasmid that replicates and functions in host cells is produced. Next, the expression plasmid is introduced into an appropriate host cell to obtain a transformant cell. Examples of the host cell include prokaryotes such as Escherichia coli, unicellular eukaryotes such as yeast, and cells of multicellular eukaryotes such as insects and animals. Methods for introducing genes into host cells include the calcium phosphate method, the DEAE-dextran method, and the electric pulse method. The transformant produces the target protein by culturing in a suitable medium. The protein obtained as described above can be isolated and purified by a general biochemical method.
これらの本発明プラスミ ドによって形質転換された形質転換体も本発明の範囲 に包含される。 Transformants transformed with the plasmid of the present invention are also within the scope of the present invention. Is included.
本発明はさらに、 本発明ォリゴヌクレオチド分子を含有する医薬に関する。 本発明のオリゴヌクレオチド分子を含有する 「医薬」 は、 例えば、 本発明の DN A を腫瘍患者等に投与することで腫瘍を治療または予防することができる。 本発 明の DNA を投与し細胞内に導入する方法としでは、 ウィルスベクターによる方法 およびその他の方法 (日経サイエンス、 1994年 4 月号、 20-45 頁、 月刊薬事、 36 (1)23-48(1994). 実験医学増刊、 12(15)、 (1994)、 およびこれらの引用文献( 等 ) のいずれの方法も適用することができる。  The present invention further relates to a medicament containing the oligonucleotide molecule of the present invention. The “medicine” containing the oligonucleotide molecule of the present invention can treat or prevent a tumor by, for example, administering the DNA of the present invention to a tumor patient or the like. Methods of administering the DNA of the present invention and introducing it into cells include viral vectors and other methods (Nikkei Science, April 1994, pp. 20-45, Monthly Pharmaceutical Affairs, 36 (1) 23-48 (1994). Any of the methods in Experimental Medical Special Edition, 12 (15), (1994), and references cited therein (etc.) can be applied.
ウィルスベクターによる方法としては、 例えばレトロウイルス、 アデノウィル ス、 アデノ関連ウィルス、 ヘルぺスウィルス、 ワクシニアウィルス、 ボックスゥ ィルス、 ポリオウイルス、 シンビスウィルス等の RNA ウィルスまたは DNA ウィル スに本発明の DNA を組み込んで導入する方法が挙げられる。 この中で、 レトロゥ ィルス、 アデノウイルス、 アデノ関連ウィルス、 ワクシニアウィルス等を用いた 方法が特に好ましい。  As a method using a viral vector, for example, the DNA of the present invention is added to an RNA virus or DNA virus such as retrovirus, adenovirus, adeno-associated virus, herpes virus, vaccinia virus, box virus, polio virus, and simbis virus. There is a method of incorporating and introducing. Among them, a method using retrovirus, adenovirus, adeno-associated virus, vaccinia virus and the like is particularly preferable.
その他の方法としては、 発現プラスミ ドを直接筋肉内に投与する方法 (DNA ヮ クチン法) 、 リボソーム法、 リポフエクチン法、 マイクロインジェクション法、 リン酸カルシウム法、 エレク ト口ポレーシヨン法等が挙げられ、 特に DNA ヮクチ ン法、 リボソーム法が好ましい。  Other methods include direct injection of the expression plasmid into the muscle (DNA-injection method), ribosome method, lipofectin method, microinjection method, calcium phosphate method, and electoral poration method. The ribosome method is preferred.
これらの本発明ォリゴヌクレオチド分子を有するプラスミ ドも本発明の範囲に 含まれる。  Plasmids having these oligonucleotide nucleotide molecules of the present invention are also included in the scope of the present invention.
本発明の腫瘍抗原べプチドをコ一ドする遺伝子を実際に医薬として作用させる には、 遺伝子を直接体内に導入する in vivo方法、 およびヒ トからある種の細胞 を採用し体外で遺伝子を該細胞に導入しその細胞を体内に戻す ex vivo方法があ る (日経サイエンス、 1994年 4 月号、 20-45 頁、 月刊薬事、 36(1)23-48(1994)、 実験医学増刊、 12(15)、 (1994)、 およびこれらの引用文献等) 。 in vivo方法が より好ましい。 in vivo方法により投与する場合は、 治療目的の疾患、 症状等に応じた適当な 投与経路により投与され得る。 例えば、 静脈、 動脈、 皮下、 筋肉内などに投与す ることが出来る。 in vivo方法により投与する場合は、 例えば、 液剤等の製剤形 態をとりうるが、 一般的には有効成分である本発明の DNA を含有する注射剤等と され、 必要に応じて、 慣用の担体を加えてもよい。 また、 本発明の DNA を含有す るリボソームまたは膜融合リボソーム (センダイウィルス (H V J ) —リポソ一 ム等) においては、 懸濁剤、 凍結剤、 遠心分離濃縮凍結剤等のリボソーム製剤の 形態とすることができる。 In order for the gene encoding the tumor antigen peptide of the present invention to actually act as a medicament, an in vivo method of directly introducing the gene into the body, and the use of certain cells from humans to transform the gene in vitro There is an ex vivo method to introduce cells into cells and return the cells to the body (Nikkei Science, April 1994, pp. 20-45, Monthly Pharmaceutical Affairs, 36 (1) 23-48 (1994), extra edition of experimental medicine, 12 (15), (1994), and references cited therein). In vivo methods are more preferred. When administered by the in vivo method, it can be administered by an appropriate administration route depending on the disease, symptom and the like for the purpose of treatment. For example, it can be administered intravenously, intraarterially, subcutaneously, or intramuscularly. When administered by the in vivo method, for example, it can be in the form of a liquid preparation or the like.However, it is generally an injection containing the DNA of the present invention as an active ingredient. A carrier may be added. In addition, the DNA-containing ribosome or membrane fusion ribosome (such as Sendai virus (HVJ) -liposome) of the present invention is in the form of a ribosome preparation such as a suspending agent, a freezing agent, and a centrifugal concentrated freeze agent. be able to.
製剤中の本発明の DNA 含量は、 治療目的の疾患、 患者の年齢、 体重等により適 宜調整することができるが、 通常本発明の DNA として、 0. 0001mg〜100mg 、 好ま しくは O. OOlmg 〜10mgであり、 これを数日ないし数月に 1回投与するのが好まし い。  The content of the DNA of the present invention in the preparation can be appropriately adjusted depending on the disease to be treated, the age and weight of the patient, etc., and usually, the DNA of the present invention is 0.0001 mg to 100 mg, preferably O.OOlmg. 1010 mg, preferably administered once every few days or months.
本発明はさらに、 配列番号: 2または 4の腫瘍抗原タンパク質をコードするポ リヌクレオチド分子のコ一ディング配列またはその 5 ' ノンコ一ディング配列中 の塩基配列と相補的な配列からなるオリゴヌクレオチド分子またはその化学的修 飾体に関する。 好ましくは、 配列番号: 2または 4の塩基配列 (構造遺伝子部分 ) からなるポリヌクレオチド分子のコーディ ング配列またはその 5 ' ノンコーデ ィング配列中の塩基配列と相補的な配列をもつ 9塩基以上からなる DNA もしくは RNA である。 このような DNA もしくは RNA とは、 2 本鎖 DNA のアンチセンス鎖の DNA またはそのアンチセンス鎖の DNA に対応する RNA であって 9塩基以上からな るもの (以下、 アンチセンスオリゴヌクレオチドという。 ) をいう。  The present invention further provides an oligonucleotide molecule comprising a coding sequence of a polynucleotide molecule encoding the tumor antigen protein of SEQ ID NO: 2 or 4, or a sequence complementary to a base sequence in the 5 ′ non-coding sequence thereof, or It relates to the chemically decorated body. Preferably, a DNA consisting of 9 or more bases having a sequence complementary to the base sequence in the coding sequence of the polynucleotide molecule consisting of the base sequence of SEQ ID NO: 2 or 4 (structural gene portion) or its 5 ′ non-coding sequence Or RNA. Such DNA or RNA is the DNA of the antisense strand of double-stranded DNA or RNA corresponding to the DNA of the antisense strand, and is composed of 9 bases or more (hereinafter referred to as antisense oligonucleotide). Say.
このアンチセンスオリゴヌクレオチドは、 例えば本発明の腫瘍抗原べプチドを コードする遺伝子の塩基配列を基にして DNA として製造するか、 またこの DNA を アンチセンスの向きに遺伝子発現プラスミ ドに組み込むことで容易に対応する RN A を製造することができる。  This antisense oligonucleotide can be easily prepared, for example, as DNA based on the nucleotide sequence of the gene encoding the tumor antigen peptide of the present invention, or by incorporating this DNA into a gene expression plasmid in the antisense direction. Can be produced.
このアンチセンスオリゴヌクレオチドは、 本発明の遺伝子である cDNAのコーデ イング部分、 5 ' ノンコーディング部分のいずれの部分の相補的な配列であって もよいが、 好ましくは転写開始部位、 翻訳開始部位、 5 ' 非翻訳領域、 ェクソン とイントロンとの境界領域もしくは 5' CAP 領域に相補的配列であることが望まし い。 This antisense oligonucleotide encodes the cDNA of the gene of the present invention. It may be a sequence complementary to any of the inking portion and the 5 'non-coding portion, but is preferably a transcription initiation site, a translation initiation site, a 5' untranslated region, a boundary region between exon and intron, or 5 '. It is desirable that the sequence be complementary to the CAP region.
「オリゴヌクレオチド分子の化学的修飾」 としては、 DNA または RNA の細胞内 への移行性または細胞内での安定性を高めることができる化学的修飾体を表し、 例えば、 ホスホチォェ一ト、 ホスホロジチォェ一ト、 アルキルホスホトリエステ ル、 アルキルホスホナート、 アルキルホスホアミデート等の誘導体 ("Ant isense “Chemical modification of an oligonucleotide molecule” refers to a chemically modified product that can enhance the translocation of DNA or RNA into cells or the stability in cells. For example, phosphothioate, phosphorodithioate Derivatives such as alkyl phosphotriesters, alkyl phosphonates and alkyl phosphoamidates ("Ant isense
RNA and DNA" WILEY — USS刊 1992 P. 1-50) が挙げられる。 この化学的修 飾体は、 同文献等に従って製造することができる。 RNA and DNA "WILEY — USS, 1992, P. 1-50). This chemically-modified product can be produced according to the same literature.
本発明の相補的な配列を持つ DNA または RNA (以下、 アンチセンスオリゴヌク レオチドという) を用いて、 腫瘍抗原タンパク質をコードする遺伝子の発現を制 御することができる。 この方法によって腫瘍抗原タンパク質の生産量を減らすこ とで、 自己免疫疾患を治療または予防することができる。 このようなアンチセン スォリゴヌクレオチドを含有する医薬も本発明に包含される。  The expression of the gene encoding the tumor antigen protein can be controlled using DNA or RNA (hereinafter, referred to as antisense oligonucleotide) having a complementary sequence of the present invention. This method can treat or prevent autoimmune diseases by reducing the production of tumor antigen proteins. Pharmaceuticals containing such antisense oligonucleotides are also included in the present invention.
アンチセンスオリゴヌクレオチドをそのまま投与する場合は、 このアンチセン スオリゴヌクレオチドの好ましい長さとしては、 例えば 5— 2 0 0塩基のものが 挙げられる。  When the antisense oligonucleotide is administered as it is, a preferable length of the antisense oligonucleotide is, for example, 5 to 200 bases.
また、 アンチセンスオリゴヌクレオチドを発現プラスミ ドに組み込む場合は、 このアンチセンスォリゴヌクレオチドの好ましい長さとしては、 例えば 1 0 0塩 基以上が挙げられ、 好ましくは 3 0 0塩基以上が挙げられ、 さらに好ましくは 5 0 0塩基以上が挙げられる。  When an antisense oligonucleotide is incorporated into an expression plasmid, preferred lengths of the antisense oligonucleotide include, for example, 100 bases or more, and preferably 300 bases or more, More preferably, it is 500 bases or more.
アンチセンスオリゴヌクレオチドを発現プラスミ ドに組み込む場合、 このアン チセンスォリゴヌクレオチドを細胞に導入する方法としては例えば、 実験医学 12 巻 1994年に述べられている方法が挙げられ、 リポソ一厶ゃ組換えウィルスなど を利用した方法が挙げられる。 アンチセンスォリゴヌクレオチドの発現プラスミ ドは通常の発現べクタ一を用いてプロモータ一の後ろに逆向きに、 すなわち本発 明の遺伝子が 3 ' から 5 ' の向きに転写されるように、 本発明の遺伝子をつなぐ だけで簡単に作製できる。 When incorporating an antisense oligonucleotide into an expression plasmid, examples of a method for introducing this antisense oligonucleotide into cells include the method described in Experimental Medicine, Vol. 12, 1994, and Liposome Co., Ltd. There is a method using a replacement virus. Expression plasmid for antisense oligonucleotides The gene is simply connected by connecting the gene of the present invention so that the gene of the present invention is transcribed in the reverse direction behind the promoter using a normal expression vector, that is, in the 3 ′ to 5 ′ direction. Can be manufactured.
このようなアンチセンスォリゴヌクレオチドを有するプラスミ ドも本発明に包 含される。  A plasmid having such an antisense oligonucleotide is also included in the present invention.
アンチセンスオリゴヌクレオチドまたはその化学的修飾体をそのまま投与する 場合、 安定化剤、 緩衝液、 溶媒などと混合して製剤された後、 投与時には抗生物 質、 抗炎症剤、 麻酔薬などと同時に用いることもできる。 こうして作成された製 剤は様々な方法で投与可能である。 投与は連日または数日から数週間おきになさ れるのが好ましい。 また、 この様な頻回の投与を避けるために徐放性のミニペレ ッ ト製剤を作製し患部近くに埋め込むことも可能である。 あるいはォスモチック ポンプなどを用いて患者に連続的に徐々に投与することも可能である。 通常投与 量は作用部位における濃度が 0. ΙηΜ-10 ζ になるように調製する。  When the antisense oligonucleotide or its chemically modified product is administered as it is, it is formulated by mixing with a stabilizer, buffer, solvent, etc., and is used simultaneously with antibiotics, anti-inflammatory drugs, anesthetics, etc. You can also. The preparations thus prepared can be administered in various ways. Preferably, the administration is performed daily or every few days to several weeks. In order to avoid such frequent administration, it is possible to prepare a sustained-release minipellet preparation and implant it near the affected area. Alternatively, it can be continuously and gradually administered to a patient using an osmotic pump or the like. The usual dosage should be adjusted so that the concentration at the site of action is 0.1%.
このようなアンチセンスオリゴヌクレオチドまたはその化学的修飾体を含有す る医薬も本発明に包含される。 以下、 実施例により本発明をさらに詳しく説明するが、 本発明はこれらの実施 例によってなんら限定されるものではない。  A medicament containing such an antisense oligonucleotide or a chemically modified product thereof is also included in the present invention. Hereinafter, the present invention will be described in more detail with reference to examples, but the present invention is not limited to these examples.
参考例 1 Reference example 1
食道癌細胞株に対する細胞傷害性 T細胞 (C T L ) 株の樹立 Establishment of cytotoxic T cell (CTL) strain against esophageal cancer cell line
手術的に摘出された 6 7歳男性の食道癌転移性リンパ節組織を無菌的に細切し ペースト状にしたのち、 インタ一ロイキン 2存在下で約 6 0日間、 5 %炭酸ガス ( 9 5 %空気) 培養器にて培養する。 その間、 培養 2 8日目以降、 頻回に培養し て増殖する T細胞の各種癌細胞に対する細胞障害能を5 1 C r遊離法と I F N - 7 測定法にて解析した。 その結果、 培養 3 9日目〜 4 9日目の T細胞が C D 8陽性 のキラー T細胞を主体とし、 かつ MH Cクラス I抗原のうちの HLA- Cw0102拘束性 の C T L活性を示すことが判明した。 HLA- Cw0102陽性の癌細胞中、 扁平上皮癌で ある TE8 細胞株 (M. Nakao ら, Cancer Research 55, 4248-4252, 1995) が最も高 い感受性を上記 C T Lに対して示した。 そこで、 上記 C T L (YA -CTL と命名) を大量に液体窒素添加細胞凍結保存用タンクに保存し、 C T Lの認識する癌退縮 抗原遺伝子のクローニングに備えた。 参考例 2 The operatively removed 67-year-old male esophageal cancer metastatic lymph node tissue was aseptically minced and pasted into a paste, and then in the presence of interleukin 2 for about 60 days, 5% CO 2 (95) Incubate in an incubator. Meanwhile, culture 2 8 after day cytotoxic ability against various cancer cells of T cells to proliferate in culture frequently 5 1 C r release assay and IFN - were analyzed by 7 measurements. As a result, T cells from day 39 to day 49 of culture were mainly CD8-positive killer T cells, and were restricted by HLA-Cw0102 among MHC class I antigens. Was found to exhibit CTL activity. Among the HLA-Cw0102-positive cancer cells, the squamous cell carcinoma TE8 cell line (M. Nakao et al., Cancer Research 55, 4248-4252, 1995) showed the highest sensitivity to the CTL. Therefore, a large amount of the above CTL (named YA-CTL) was stored in a liquid nitrogen-containing cell cryopreservation tank to prepare for cloning of a cancer regression antigen gene recognized by CTL. Reference example 2
HLA-Cw0102 cDNAの調製  Preparation of HLA-Cw0102 cDNA
中尾ら著、 Cancer Res.55:4248-252(1995)の教示に従い、 TE8 細胞由来の HLA - Cw0102 cDNA を発現ベクター pCR3 ( INVITROGEN社製) に組込み、 組換えプラスミ ドを作製した。 : 参考例 3  According to the teaching of Nakao et al., Cancer Res. 55: 4248-252 (1995), HLA-Cw0102 cDNA derived from TE8 cells was incorporated into an expression vector pCR3 (manufactured by INVITROGEN) to produce a recombinant plasmid. : Reference example 3
TE8 細胞 cDNAライブラリ一作製  Preparation of TE8 cell cDNA library
mRNA精製システム (フアルマシアバイオテク社製) を用い添付のプロトコ一ル に従い、 TE8 細胞から全 RNA 画分の分離および ol igo(dT) カラムによる poly(A) + mRNAの調製を行った。 mRNAよりスーパースクリプトプラスミ ドシステム(GIBC0 BRL 社製) を用い添付のプロトコールに従い、 両端に Not 1アダプタ一と Sal 1ァダブターを連結した cDNAを作製した後、 この cDNAを発現べク夕一 pSV- SP0RT1 (GIBCO BRL社製) の制限酵素 Not 1および Sal 1の切断部位に連結して組換え プラスミ ドを得た。 この組換えプラスミ ドをジーンパルサー(Bio- Rad社製) を用 いて 25 zF, 200 Ω , 2. 5kV の条件で、 電気パルスにより大腸菌のエレク ト口マツ クス DH10B/p3TMセル (GIBCO BRL社製) に導入し、 アンピシリン (50 gZml) を含む LB培地 ( 1 %パクトトリプトン、 0. 5%NaCl、 pH7. 3 ) 上にて組換えプラス ミ ドが導入されている形質転換体を選択した。 参考例 4 Using an mRNA purification system (manufactured by Pharmacia Biotech), separation of the total RNA fraction from TE8 cells and preparation of poly (A) + mRNA using an oligo (dT) column were performed according to the attached protocol. Using Superscript Plasmid System (GIBC0 BRL) from mRNA and preparing cDNA with Not 1 adapter and Sal 1 adapter linked at both ends according to the attached protocol, express this cDNA pSV-SP0RT1 (GIBCO BRL) and ligated to the restriction sites Not 1 and Sal 1 to obtain a recombinant plasmid. This recombinant plasmid was subjected to an electric pulse under the conditions of 25 zF, 200 Ω, and 2.5 kV using a Gene Pulser (manufactured by Bio-Rad) and an E.coli elect-mouth Matsux DH10B / p3 cell (GIBCO BRL) Transformants with recombinant plasmids selected on LB medium (1% pactotryptone, 0.5% NaCl, pH 7.3) containing ampicillin (50 gZml) did. Reference example 4
インターフヱロン一 7の定量 Quantitation of Interferon-1
インタ一フエロン一 ( IFN- r ) の定量は、 ェンザィムィムノアッセィ (ELIS A ) により行った。 9 6ウェルマイク口プレートに一次抗体として抗ヒト IFN- 7 マウスモノクロール抗体を吸着させ、 ゥシ血清アルブミンで非特異的結合をプロ ックした後、 検体中の IFN- 7を抗体に結合させた。 次に二次抗体として抗ヒト IF Ν- γゥサギポリクロール抗体を結合させ、 さらにペルォキシダ一ゼ標識した抗ゥ サギ免疫グロブリンロバ抗体を結合した後、 発色剤として ΤΜ Β Ζ (テトラメチ ルペンジデイン) を反応させ、 2Ν H2 SO 4 を等量加えて反応を停止させた後、 吸 光度 (450 nm)を測定した。 これを標準品の IFN-ァより得られた値と比較すること により定量した。 実施例 1 Quantification of interferon (IFN-r) was performed by Enzymimnoassay (ELIS A). 9 Adsorb anti-human IFN-7 mouse monoclonal antibody as the primary antibody on the 6-well mic opening plate, block nonspecific binding with serum albumin, and bind IFN-7 in the sample to the antibody. Was. Then anti-human IF New as a secondary antibody - to bind the γ © heron poly crawl antibodies after binding the further Peruokishida Ichize labeled anti © heron immunoglobulin donkey antibody, ΤΜ Β Ζ as coloring agent (tetramethyl Rupenjidein) After the reaction, the reaction was stopped by adding an equal amount of 2ΝH2SO4, and the absorbance (450 nm) was measured. This was quantified by comparing it with the value obtained from the standard IFN-α. Example 1
腫瘍抗原タンパク質遺伝子のスクリーニング Screening for tumor antigen protein genes
まず、 参考例 3にて調製した形質転換体のプールから組換えプラスミ ド DNA を 回収する。  First, recombinant plasmid DNA is recovered from the pool of transformants prepared in Reference Example 3.
アンピシリン (50 /z g /ml) を含む LB培地の入った 96ゥヱル U底マイクロプレ 一卜にゥ ルあたり 100-200 個の形質転換体を加え培養後、 その一部をゥエル当 たり 0. 3ml の TYGPN 培地 (F. . Ausubel ら編、 CURRENT PR0TC0LS IN MOLECULAR BIOLOGY, John Wi ley & Sons, Inc. ) の入った別の 96ゥエル U底マイクロプレー 卜に移して 3 7でで 4 8時間培養し、 残りの LB培地のマイクロプレートは凍結保 存した。 TYGPN 培地で培養した形質転換体の組換えプラスミ ド DNA は、 マイクロ プレートでアル力リ溶解法 (F. M. Ausubel ら編、 CURRENT P 0TC0LS IN M0LECU LAR BIOLOGY, John Wi ley & Sons, Inc. ) により調製した。 イソプロパノール沈 殿で回収した組換えプラスミ ド DNA は、 50 / 1の 20 ng /ml RNaseを含む 10mM T ris , ImM EDTA, pH7. 4 溶液に懸濁した。 次に、 上記にて調製した組換えプラスミ ド DNA と参考例 2にて調製した HLA- Cw 0102 cDNA の組換えプラスミ ドを C0S7細胞 (Gluzan. Y. Cel l, 23 : 175- 182, 1981 ) にリポフエクチン法により同時にトランスフエク トする。 Add 100-200 transformants per well to a 96-well U-bottom microplate containing LB medium containing ampicillin (50 / zg / ml), culture, and aliquot 0.3 ml per well. TYGPN medium (F. Ausubel et al., Ed., CURRENT PR0TC0LS IN MOLECULAR BIOLOGY, John Wiley & Sons, Inc.), transferred to another 96-well U-bottom microplate and incubated at 37 for 48 hours. The remaining microplates of the LB medium were stored frozen. Recombinant plasmid DNA from transformants cultured in TYGPN medium was prepared by microplate-based lysis method (FM Ausubel et al., Ed., CURRENT P0TC0LS IN M0LECULAR BIOLOGY, John Wiley & Sons, Inc.). . Recombinant plasmid DNA recovered by isopropanol precipitation was suspended in a 10 mM Tris, ImM EDTA, pH 7.4 solution containing 50/1 20 ng / ml RNase. Next, the recombinant plasmid DNA prepared above and the recombinant plasmid of the HLA-Cw 0102 cDNA prepared in Reference Example 2 were used in C0S7 cells (Gluzan. Y. Cell, 23: 175-182, 1981). First, transfect the cells simultaneously by the lipofectin method.
C0S7細胞を 96ゥエル平底マイクロプレートのゥエル当たり 1 X 104 個を加え、 100 μ. 1の 10%FCS を含む RPMI培養液で 1 日培養した。 形質転換体約 100 個分の TE8cDNA の組換えプラスミ ド 25 // 1に参考例 2にて調製した HLA- Cw0102 cDNA の 組換えプラスミ ド 100 ngを加え、 さらに約 100 倍に希釈したリボフヱクチン試薬 (リボフェクタミン、 GIBC0-BRL 社製) 25 ;/ 1を加えた。 得られた混合液 50 1 (リボソームと組換えプラスミ ドの融合懸濁液) を、 培養した C0S7細胞に加え てダブルトランスフヱク 卜した。 トランスフエクタントは 2 点ずつ用意した。 ト ランスフヱクタントは 48〜72時間、 37°Cで培養した後、 培養液を除去し、 ゥエル 当たり 1 X 104 個の TE8 C T L細胞を加えて 100 1の 10%ヒト血清と 50UZml の IL-2を含む培養液で 37°Cで 16〜24時間培養した。 培養液を回収し、 IFN- yを EL ISA で測定した。 C0S7 cells were added at 1 × 10 4 cells / well of a 96-well flat-bottom microplate and cultured for 1 day in 100 μl of RPMI medium containing 10% FCS. 100 ng of the recombinant plasmid of HLA-Cw0102 cDNA prepared in Reference Example 2 was added to about 100 transformants of the recombinant plasmid TE8 cDNA 25 // 1 and the ribofectin reagent (about 100-fold diluted) was added. Ribofectamine, manufactured by GIBC0-BRL) 25; / 1. The obtained mixed solution 501 (fusion suspension of ribosome and recombinant plasmid) was added to the cultured C0S7 cells, and double transfected. Two transfectants were prepared. Transfectants are cultured for 48-72 hours at 37 ° C, the culture medium is removed, and 1 x 10 4 TE8 CTL cells per well are added and 100 1 10% human serum and 50 UZml are added. The cells were cultured in a culture solution containing IL-2 at 37 ° C for 16 to 24 hours. The culture was collected and IFN-y was measured by ELISA.
次いで、 ELISA によって高い IFN — 7産生が認められた 8群について、 該当す る凍結保存しておいた TE8CDNA の組換えプラスミ ドによる形質転換体約 100 〜20 0 クローン/ゥヱルのプールを用いてさらに以下のようにスクリ一二ングを行う ο  Next, for the 8 groups in which high IFN-7 production was observed by ELISA, using the pool of approximately 100 to 200 clones / milliliter of the transformants of the corresponding TE8CDNA recombinant plasmids that had been frozen and stored. Screen as follows ο
形質転換体のプールを約 6 時間 LB (アンピシリン 1 /ml) 培地にて培養し 、 さらに培養物をアンピシリン (δΟ ζ ΐ ΖπιΙ) を含む LB寒天培地のプレートにま いて 1 日培養し、 得られた単一コロニー各群 200 コロニー、 合計 8 X 200 コロニ —をそれぞれ 9 6穴マイクロプレー卜の各ゥヱルに移し、 ゥエル当たりの形質転 換体が 1種類となる条件で上記と同様の方法で培養し、 TE8 cDNAの組換えプラス ミ ド DNA を調製した。 さらに上記と同様な方法により TE8 cDNAの組換えプラスミ ドと HLA- Cw0102 cDNA の組換えプラスミ ドとを C0S7細胞にダブルトランスフエク トし、 引き続いて TE8 C T L細胞との混合培養を行い、 TE8 C T L細胞が反応し て産生した培養液中の IFN- 7を定量し、 陽性プラスミ ドを選択した。 この操作に より、 TE8 C T L細胞と反応する TE8 細胞 cDNA組換えプラスミ ドクローン 2つが 選択され、 それぞれクローン 3およびクローン 6と命名した。 クローン 3および 6について、 さらにもう一度、 同様な操作を繰り返して TE8 C T L細胞による IF N- 7の産生を確認した。 実施例 2 The transformant pool is cultured for about 6 hours in LB (ampicillin 1 / ml) medium, and the culture is further cultured on a plate of LB agar medium containing ampicillin (δΟ ΐ ΖπιΙ) for 1 day. Transfer 200 colonies from each colony to a total of 8 × 200 colonies in each well of a 96-well microplate, and incubate in the same manner as above under the condition that only one transformant per well is used. Then, recombinant plasmid DNA of TE8 cDNA was prepared. Further, the recombinant plasmid of TE8 cDNA and the recombinant plasmid of HLA-Cw0102 cDNA were double-transfected into C0S7 cells by the same method as described above, and then mixed and cultured with TE8 CTL cells. Reacts IFN-7 in the resulting culture was quantified and positive plasmids were selected. By this operation, two TE8 cell cDNA recombinant plasmid clones reacting with TE8 CTL cells were selected, and named clone 3 and clone 6, respectively. For clones 3 and 6, the same operation was repeated once more to confirm the production of IFN-7 by TE8 CTL cells. Example 2
腫瘍抗原遺伝子の塩基配列決定 Nucleotide sequencing of tumor antigen gene
目的の腫瘍抗原遺伝子の cDNAが組み込まれた組換えプラスミ ドを持つ形質転換 体クローン 3およびクローン 6をそれぞれ、 500ml のアンピシリン (50〃 1 /ml ) を含む LB培地で 37°Cで 14〜16時間培養し、 遠心分離にて菌体を回収した。 菌体 から PLASMID MAXI キッ ト (QIAGBN社製) に従い、 組換えプラスミ ドを回収した 。 cDNAは、 SP6 RNAポリメラーゼプロモーター配列と T7 RNA ポリメラーゼプロ モーター配列に挟まれた部位に組み込まれている。 そこで文献 (DNA 4 : 165, 198 5 ) に記載の SP6 プロモー夕一プライマーおよび T7プロモータープライマ一を合 成した。 次に、 SP6 プロモー夕一プライマーまたは T7プロモータープライマ一を Fluore-dATP Label ing Mix (フアルマシアバイオテク社製) および AutoRead Sequencing Ki t (フアルマシアバイオテク社製) と組み合わせてジデォキシシ —クェンシング反応を行い、 蛍光 DNA シーケンサ一 (フアルマシアバイオテク社 製) を使用し、 両端から cDNAの塩基配列を決定した。 クローン 3に相当する cDNA の塩基配列は全長が 532塩基対およびクローン 6に相当する cDNAの塩基配列は全 長が 756 塩基対と決定され、 それぞれ配列番号: 1および配列番号: 4の通りで あつ 7こ。  Transformant clones 3 and 6 each having a recombinant plasmid containing the cDNA of the tumor antigen gene of interest were cloned for 14-16 at 37 ° C in LB medium containing 500 ml of ampicillin (50〃1 / ml). After culturing for an hour, the cells were collected by centrifugation. Recombinant plasmid was recovered from the cells according to the PLASMID MAXI kit (QIAGBN). The cDNA is integrated at the site between the SP6 RNA polymerase promoter sequence and the T7 RNA polymerase promoter sequence. Thus, the SP6 Promoter primer and the T7 promoter primer described in the literature (DNA 4: 165, 1985) were synthesized. Next, a didoxyquencing reaction was performed by combining the SP6 Promoter primer or T7 promoter primer with Fluore-dATP Labeling Mix (Pharmacia Biotech) and AutoRead Sequencing Kit (Pharmacia Biotech). The nucleotide sequence of the cDNA was determined from both ends using a fluorescent DNA sequencer (Pharmacia Biotech). The nucleotide sequence of cDNA corresponding to clone 3 was determined to be 532 base pairs in total length, and the nucleotide sequence of cDNA corresponding to clone 6 was determined to be 756 base pairs in total length, as shown in SEQ ID NO: 1 and SEQ ID NO: 4, respectively. 7
Gene Worksデーターベースを使用し、 配列番号: 1および 4に記載の塩基配列 の検索を行った。 その結果、 以下のことが判明した:  Using the Gene Works database, the nucleotide sequences described in SEQ ID NOs: 1 and 4 were searched. The results revealed the following:
クローン 3 検索の結果、 得られたクローン 3の cDNA塩基配列はュビキチン前駆体 (P. Kay Lund et aし J. Bi ol. Chem. Vol. 260, No. 12, pp7609- 7613, 1985) の塩基配列と 1 0 0 %の相同性を有することが判明した。 得られた cDNA塩基配列の推定アミノ酸 配列も報告されているュビキチン前駆体 (P. Kay Lund et al ., 前掲) のァミノ 酸配列と 1 0 0 %の相同性を有していた。 この cDNA配列および推定アミノ酸配列 は配列番号: 1に示している。 Clone 3 As a result of the search, the cDNA nucleotide sequence of clone 3 obtained was the same as that of ubiquitin precursor (P. Kay Lund et al., J. Biol. Chem. Vol. 260, No. 12, pp7609-7613, 1985). It was found to have 100% homology. The deduced amino acid sequence of the obtained cDNA base sequence also had 100% homology with the amino acid sequence of the reported ubiquitin precursor (P. Kay Lund et al., Supra). The cDNA sequence and deduced amino acid sequence are shown in SEQ ID NO: 1.
クローン 6 Clone 6
検索の結果、 得られたクローン 6の cDNA塩基配列は 3' - 端に 3' - UTR (非翻訳部 ) と poly(A) テールが付加されているものの、 その残りの部分はヒストン H 1の 塩基配列と 1 0 0 %の相同性を有していた。 得られた cDNA塩基配列の推定ァミノ 酸配列も報告されているヒストン H 1のアミノ酸配列と 1 0 0 %の相同性を有し ていた。 この cDNA配列および推定アミノ酸配列は配列番号: 3に示している。 実施例 3  As a result of the search, the cDNA base sequence of clone 6 obtained had a 3'-UTR (untranslated portion) and a poly (A) tail added to the 3'-end, but the rest was that of histone H1. It had 100% homology with the nucleotide sequence. The deduced amino acid sequence of the obtained cDNA base sequence also had 100% homology with the reported amino acid sequence of histone H1. The cDNA sequence and deduced amino acid sequence are shown in SEQ ID NO: 3. Example 3
クローン 3の活性断片の特定 Identification of the active fragment of clone 3
1 . クローン 3の cDNAの断片の調製  1. Preparation of cDNA fragment of clone 3
T Aクロ一ニングキッ ト(Inv i trogen)を用い、 断片 1 (276 塩基対、 92ァミノ 酸) および断片 2 (278 塩基対、 92アミノ酸) を調製した。 断片 1および 2のァ ミノ酸配列を図 1に示す。  Fragment 1 (276 bp, 92 amino acids) and Fragment 2 (278 bp, 92 amino acids) were prepared using a TA cloning kit (Invitrogen). The amino acid sequences of fragments 1 and 2 are shown in FIG.
2 . 活性断片のスクリーニング  2. Screening of active fragments
上記にて調製した組換えプラスミ ド DNA と参考例 2にて調製した HLA-Cw0102 c DNA の組換えプラスミ ドを C0S7細胞に実施例 2に記載の手法により同時トランス フエク トし、 IFN-ァ産生量を ELISA によって測定した。 得られた結果を図 2に示 す。 これにより、 断片 1が活性であることが判明した。 断片 1の配列を配列番号 : 2に示す。 実施例 4 The recombinant plasmid DNA prepared above and the recombinant plasmid of HLA-Cw0102 cDNA prepared in Reference Example 2 were cotransfected into C0S7 cells by the method described in Example 2 to produce IFN-α. The amount was determined by ELISA. Figure 2 shows the results obtained. This revealed that fragment 1 was active. The sequence of fragment 1 is shown in SEQ ID NO: 2. Example 4
クローン 6の活性断片の特定 Identification of active fragment of clone 6
1 . クローン 6の cDNAの断片の調製  1. Preparation of cDNA fragment of clone 6
T Aクロ一ニングキッ トを用い、 断片 1 (180 塩基対、 60ァミノ酸) 、 断片 2 (362 塩基対、 100 アミノ酸) および断片 3 (281 塩基対、 93アミノ酸) を調製 した。 断片 1、 2および 3のアミノ酸配列を図 3に示す。  Fragment 1 (180 bp, 60 amino acids), Fragment 2 (362 bp, 100 amino acids) and Fragment 3 (281 bp, 93 amino acids) were prepared using a TA cloning kit. The amino acid sequences of fragments 1, 2 and 3 are shown in FIG.
2 . 活性断片のスクリ一ニング  2. Screening of active fragments
上記にて調製した組換えプラスミ ド DNA と参考例 2にて調製した HLA- Cw0102 c DNA の組換えプラスミ ドを C0S7細胞に実施例 2に記載の手法により同時トランス フエク トし、 IFN-ァ産生量を ELISA によって測定した。 得られた結果を図 4に示 す。 これにより、 断片 3が活性であることが判明した。 断片 3の配列を配列番号 : 4に示す。 産業上の利用可能性  The recombinant plasmid DNA prepared above and the HLA-Cw0102 cDNA prepared in Reference Example 2 were co-transfected into C0S7 cells by the method described in Example 2 to produce IFN-α. The amount was determined by ELISA. Figure 4 shows the obtained results. This revealed that fragment 3 was active. The sequence of fragment 3 is shown in SEQ ID NO: 4. Industrial applicability
本発明の腫瘍抗原タンパク質および腫瘍抗原べプチドを用いた抗腫瘍免疫を活 性化するための医薬、 本発明の腫瘍抗原夕ンパク質に対する抗体等を用いた自己 免疫疾患を治療するための医薬、 および腫瘍抗原タンパク質をコードする DNA 等 を含有する医薬を提供することができ、 また腫瘍または自己免疫疾患の診断方法 を提供することができる。 A medicament for activating antitumor immunity using the tumor antigen protein and the tumor antigen peptide of the present invention; a medicament for treating an autoimmune disease using an antibody against the tumor antigen protein of the present invention; And a medicament containing a DNA encoding a tumor antigen protein and the like, and a method for diagnosing a tumor or an autoimmune disease.
配列表 Sequence listing
配列番号: 1 SEQ ID NO: 1
配列の長さ : 532 Array length: 532
配列の型:核酸 Sequence type: nucleic acid
トポロジー:直鎖状  Topology: linear
配列の種類: cDNA Sequence type: cDNA
起源 Origin
生物名 : ヒ卜 (Homo sapiens )  Organism Name: Homo (Homo sapiens)
組織の種類:食道癌組織  Tissue type: Esophageal cancer tissue
配列: Array:
GCCATCTGCG GTGGAGCCGC ACAAA 25  GCCATCTGCG GTGGAGCCGC ACAAA 25
ATG CAG ATT TTC GTG AAA ACC CTT ACG GGG AAG ACC ATC ACC CTC GAG 73 Met Gin He Phe Val Lys Thr Leu Thr Gly Lys Thr l ie Thr Leu Glu ATG CAG ATT TTC GTG AAA ACC CTT ACG GGG AAG ACC ATC ACC CTC GAG 73 Met Gin He Phe Val Lys Thr Leu Thr Gly Lys Thr lie Thr Leu Glu
1 5 10 15  1 5 10 15
GTT GAA CCC TCG GAT ACG ATA GAA AAT GTA AAG GCC AAG ATC CAG GAT 121 Val Glu Pro Ser Asp Thr l ie Glu Asn Val Lys Ala Lys l ie Gin Asp  GTT GAA CCC TCG GAT ACG ATA GAA AAT GTA AAG GCC AAG ATC CAG GAT 121 Val Glu Pro Ser Asp Thr lie Glu Asn Val Lys Ala Lys lie Gin Asp
20 25 30 20 25 30
AAG GAA GGA ATT CCT CCT GAT CAG CAG AGA CTG ATC TTT GCT GGC AAG 169 Lys Glu Gly l ie Pro Pro Asp Gin Gin Arg Leu l ie Phe Ala Gly Lys AAG GAA GGA ATT CCT CCT GAT CAG CAG AGA CTG ATC TTT GCT GGC AAG 169 Lys Glu Gly lie Pro Pro Asp Gin Gin Arg Leu lie Phe Ala Gly Lys
35 40 45  35 40 45
CAG CTG GAA GAT GGA CGT ACT TTG TCT GAC TAC AAT ATT CAA AAG GAG 217 Gin Leu Glu Asp Gly Arg Thr Leu Ser Asp Tyr Asn l ie Gin Lys Glu  CAG CTG GAA GAT GGA CGT ACT TTG TCT GAC TAC AAT ATT CAA AAG GAG 217 Gin Leu Glu Asp Gly Arg Thr Leu Ser Asp Tyr Asn lie Gin Lys Glu
50 55 60  50 55 60
TCT ACT CTT CAT CTT GTG TTG AGA CTT CGT GGT GGT GCT AAG AAA AGG 265 Ser Thr Leu His Leu Val Leu Arg Leu Arg Gly Gly Ala Lys Lys Arg 65 70 75 80TCT ACT CTT CAT CTT GTG TTG AGA CTT CGT GGT GGT GCT AAG AAA AGG 265 Ser Thr Leu His Leu Val Leu Arg Leu Arg Gly Gly Ala Lys Lys Arg 65 70 75 80
AAG AAG AAG TCT TAC ACC ACT CCC AAG AAG AAT AAG CAC AAG AGA AAG 313 Lys Lys Lys Ser Tyr Thr Thr Pro Lys Lys Asn Lys His Lys Arg Lys AAG AAG AAG TCT TAC ACC ACT CCC AAG AAG AAT AAG CAC AAG AGA AAG 313 Lys Lys Lys Ser Tyr Thr Thr Pro Lys Lys Asn Lys His Lys Arg Lys
85 90 95  85 90 95
AAG GTT AAG CTG GCT GTC CTG AAA TAT TAT AAG GTG GAT GAG AAT GGC 361 Lys Val Lys Leu Ala Val Leu Lys Tyr Tyr Lys Val Asp Glu Asn Gly  AAG GTT AAG CTG GCT GTC CTG AAA TAT TAT AAG GTG GAT GAG AAT GGC 361 Lys Val Lys Leu Ala Val Leu Lys Tyr Tyr Lys Val Asp Glu Asn Gly
100 105 110 100 105 110
AAA ATT AGT CGC CTT CGT CGA GAG TGC CCT TCT GAT GAA TGT GGT GCT 409 Lys l ie Ser Arg Leu Arg Arg Glu Cys Pro Ser Asp Glu Cys Gly Ala AAA ATT AGT CGC CTT CGT CGA GAG TGC CCT TCT GAT GAA TGT GGT GCT 409 Lys lie Ser Arg Leu Arg Arg Glu Cys Pro Ser Asp Glu Cys Gly Ala
115 120 125  115 120 125
GGG GTG TTT ATG GCA AGT CAC TTT GAC AGA CAT TAT TGT GGC AAA TGT 457 Gly Val Phe Met Ala Ser His Phe Asp Arg His Tyr Cys Gly Lys Cys  GGG GTG TTT ATG GCA AGT CAC TTT GAC AGA CAT TAT TGT GGC AAA TGT 457 Gly Val Phe Met Ala Ser His Phe Asp Arg His Tyr Cys Gly Lys Cys
130 135 140  130 135 140
TGT CTG ACT TAC TGT TTC AAC AAA CCA GAA GAC AAG 493 Cys Leu Thr Tyr Cys Phe Asn Lys Pro Glu Asp Lys  TGT CTG ACT TAC TGT TTC AAC AAA CCA GAA GAC AAG 493 Cys Leu Thr Tyr Cys Phe Asn Lys Pro Glu Asp Lys
145 150 155 145 150 155
TAACTGTATG AGTTAATAAA AGACATGAAC TAACAAAAA 532 配列番号: 2 TAACTGTATG AGTTAATAAA AGACATGAAC TAACAAAAA 532 SEQ ID NO: 2
配列の長さ : 276 Sequence length: 276
配列の型:核酸 Sequence type: nucleic acid
トポロジー:直鎖状  Topology: linear
配列の種類: cDNA Sequence type: cDNA
配列: Array:
ATG CAG ATT TTC GTG AAA ACC CTT ACG GGG AAG ACC ATC ACC CTC GAG 48 Met Gin He Phe Val Lys Thr Leu Thr Gly Lys Thr l ie Thr Leu GluATG CAG ATT TTC GTG AAA ACC CTT ACG GGG AAG ACC ATC ACC CTC GAG 48 Met Gin He Phe Val Lys Thr Leu Thr Gly Lys Thr lie Thr Leu Glu
1 5 10 15 1 5 10 15
GTT GAA CCC TCG GAT ACG ATA GAA AAT GTA AAG GCC AAG ATC CAG GAT 96 Val Glu Pro Ser Asp Thr l ie Glu Asn Val Lys Ala Lys He Gin Asp  GTT GAA CCC TCG GAT ACG ATA GAA AAT GTA AAG GCC AAG ATC CAG GAT 96 Val Glu Pro Ser Asp Thr lie Glu Asn Val Lys Ala Lys He Gin Asp
20 25 30 20 25 30
AAG GAA GGA ATT CCT CCT GAT CAG CAG AGA CTG ATC TTT GCT GGC AAG 144 Lys Glu Gly l ie Pro Pro Asp Gin Gin Arg Leu l ie Phe Ala Gly Lys AAG GAA GGA ATT CCT CCT GAT CAG CAG AGA CTG ATC TTT GCT GGC AAG 144 Lys Glu Gly lie Pro Pro Asp Gin Gin Arg Leu lie Phe Ala Gly Lys
35 40 45  35 40 45
CAG CTG GAA GAT GGA CGT ACT TTG TCT GAC TAC AAT ATT CAA AAG GAG 192 Gin Leu Glu Asp Gly Arg Thr Leu Ser Asp Tyr Asn l ie Gin Lys Glu  CAG CTG GAA GAT GGA CGT ACT TTG TCT GAC TAC AAT ATT CAA AAG GAG 192 Gin Leu Glu Asp Gly Arg Thr Leu Ser Asp Tyr Asn lie Gin Lys Glu
50 55 60  50 55 60
TCT ACT CTT CAT CTT GTG TTG AGA CTT CGT GGT GGT GCT AAG AAA AGG 240 Ser Thr Leu His Leu Val Leu Arg Leu Arg Gly Gly Ala Lys Lys Arg 65 70 75 80 TCT ACT CTT CAT CTT GTG TTG AGA CTT CGT GGT GGT GCT AAG AAA AGG 240 Ser Thr Leu His Leu Val Leu Arg Leu Arg Gly Gly Ala Lys Lys Arg 65 70 75 80
AAG AAG AAG TCT TAC ACC ACT CCC AAG AAG AAT AAG 276 Lys Lys Lys Ser Tyr Thr Thr Pro Lys Lys Asn Lys AAG AAG AAG TCT TAC ACC ACT CCC AAG AAG AAT AAG 276 Lys Lys Lys Ser Tyr Thr Thr Pro Lys Lys Asn Lys
85 90 配列番号: 3  85 90 SEQ ID NO: 3
配列の長さ : 756 Array length: 756
配列の型:核酸 Sequence type: nucleic acid
トポロジー:直鎖状  Topology: linear
配列の種類: cDNA Sequence type: cDNA
起源 Origin
生物名: ヒ卜 (Homo sapiens )  Organism: Homo sapiens
組織の種類:食道癌組織 L Z Tissue type: Esophageal cancer tissue LZ
OJd sxq J¾ λιο ^IO BIV sAq sAq Λ sAq sA n jas 130 VVV 03V VOO 300030 OVV VVV X199VV 330 OVV 030 WO 000301 OJd sxq J¾ λιο ^ IO BIV sAq sAq Λ sAq sA n jas 130 VVV 03V VOO 300030 OVV VVV X199VV 330 OVV 030 WO 000301
0Π 901 001  0Π 901 001
BIV BIV sAq sAq usv na s aqj ¾iv λΐθ ^ΐθ sAq BIV BIV sAq sAq usv na s aqj ¾iv λΐθ ^ ΐθ sAq
8 δ 330 VOO OVV OVV OVV 013 VVV ILL 331300131130100 GOV 300 VVV 8 δ 330 VOO OVV OVV OVV 013 VVV ILL 331 300 131 130 100 GOV 300 VVV
96 06 98  96 06 98
UI9 1¾Λ n91 J¾ s jas Λ n sAq ΠΘつ na sAUI9 1¾Λ n91 J¾ s jas Λ n sAq na na sA
908 03V WO 010013 3V 399 OVV 39V 01001330V OVV 310100113 VVV908 03V WO 010013 3V 399 OVV 39V 01001330V OVV 310 100 113 VVV
08 9Z 01 9908 9Z 01 99
3Π 3JV usv usv sAq n^g 八 dsv JAl ¾iv ¾IV na ¾iv 92 31V 10339V OVV DVV VVV 0V9910 XV9 IVl 300330330130 Oil 000 3Π 3 J V usv usv sAq n ^ g 8 dsv JAl ¾iv ¾IV na ¾iv 92 31V 10339V OVV DVV VVV 0V9910 XV9 IVl 300 330 330 130 Oil 000
09 99 OS sXq s人 n na ¾ν BIV naq jgg JBA AI J3S Sjy nio sA J9S ¾IV ¾IV 09 99 OS sXq s people n na ¾ν BIV naq jgg JBA AI J3S Sjy nio sA J9S ¾IV ¾IV
60S VVV VVV 013130100013131119 VOO 30V 103 OVO VVV 13X 330330 60S VVV VVV 013130100013131119 VOO 30V 103 OVO VVV 13X 330 330
^ 0 9S  ^ 09S
ΙΒΛ BIV sA J¾ 9ii naq nio s ΐ¾Λ OJd OJJ XIO J9S ¾IV s q 8jy ΙΒΛ BIV sA J¾ 9ii naq nio s ΐ¾Λ OJd OJJ XIO J9S ¾IV s q 8jy
Ϊ9Ϊ 0101309VV 33V 3XV 3130V9 V310100333331000310309VV 103 Ϊ9Ϊ 0101309VV 33V 3XV 3130V9 V310100333331000310309VV 103
OS 02  OS 02
OJd J¾ Ai9 Aio BIV sAq sAq ¾iv ¾1V sAq sAq s Ι¾Λ OJJ BIV s OJd J¾ Ai9 Aio BIV sAq sAq ¾iv ¾1V sAq sAq s Ι¾Λ OJJ BIV s
SU 13303V 100090130 OVV VVV 300030 OVV OVV OVV VIO 1333309VV SU 13303V 100090130 OVV VVV 300030 OVV OVV OVV VIO 1333309VV
91 Οΐ 9 I niO ¾IV OJd OJd BIV ¾1V OJd ¾IV BIV ojj ¾iv J¾ J9S 91 Οΐ 9 I niO ¾IV OJd OJd BIV ¾1V OJd ¾IV BIV ojj ¾iv J¾ J9S
99 OVO 939133133039 D3013933313003013313013V OVO 03101V 99 OVO 939133133039 D3013933313003013313013V OVO 03101V
L\ 0VV31311V00001030 L \ 0VV31311V00001030
::
Z8TtO//.6df/XDd Z8SZZ/86 OAV 115 120 125 Z8TtO //. 6df / XDd Z8SZZ / 86 OAV 115 120 125
AAG AAG CCA GTT GGG GCA GCC AAG AAG CCC AAG AAG GCG GCT GGC GGC 449 Lys Lys Pro Val Gly Ala Ala Lys Lys Pro Lys lys Ala Ala Gly Gly  AAG AAG CCA GTT GGG GCA GCC AAG AAG CCC AAG AAG GCG GCT GGC GGC 449 Lys Lys Pro Val Gly Ala Ala Lys Lys Pro Lys lys Ala Ala Gly Gly
130 135 140  130 135 140
GCA ACT CCG AAG AAG AGC GCT AAG AAA ACA CCG AAG AAA GCG AAG AAG 497 Ala Thr Pro Lys Lys Ser Ala Lys Lys Thr Pro Lys Lys Ala Lys Lys 145 150 155 160 GCA ACT CCG AAG AAG AGC GCT AAG AAA ACA CCG AAG AAA GCG AAG AAG 497 Ala Thr Pro Lys Lys Ser Ala Lys Lys Thr Pro Lys Lys Ala Lys Lys 145 150 155 160
CCG GCC GCG GCC ACT GTA ACC AAG AAA GTG GCT AAG AGC CCA AAG AAG 545 Pro Ala Ala Ala Thr Val Thr Lys Lys Val Ala Lys Ser Pro Lys Lys CCG GCC GCG GCC ACT GTA ACC AAG AAA GTG GCT AAG AGC CCA AAG AAG 545 Pro Ala Ala Ala Thr Val Thr Lys Lys Val Ala Lys Ser Pro Lys Lys
165 170 175  165 170 175
GCC AAG GTT GCG AAG CCC AAG AAA GCT GCC AAA AGT GCT GCT AAG GCT 593 Ala Lys Val Ala Lys Pro Lys Lys Ala Ala Lys Ser Ala Ala Lys Ala  GCC AAG GTT GCG AAG CCC AAG AAA GCT GCC AAA AGT GCT GCT AAG GCT 593 Ala Lys Val Ala Lys Pro Lys Lys Ala Ala Lys Ser Ala Ala Lys Ala
180 185 190  180 185 190
GTG AAG CCC AAG GCC GCT AAG CCC AAG GTT GTC AAG CCT AAG AAG GCG 641 Val Lys Pro Lys Ala Ala Lys Pro Lys Val Val Lys Pro Lys Lys Ala  GTG AAG CCC AAG GCC GCT AAG CCC AAG GTT GTC AAG CCT AAG AAG GCG 641 Val Lys Pro Lys Ala Ala Lys Pro Lys Val Val Lys Pro Lys Lys Ala
195 200 205  195 200 205
GCG CCC AAG AAG AAA 656 Ala Pro Lys Lys Lys  GCG CCC AAG AAG AAA 656 Ala Pro Lys Lys Lys
210  210
TAGGCGAACG CCTACTTCTA AAACCCAAAA GGCTCTTTTC AGAGCCACCA CTGATCTCAA 716 TAAAAGAGCT GGATAATTTC TTTAAAAAAA AAAAAAAAAA 756 配列番号: 4 TAGGCGAACG CCTACTTCTA AAACCCAAAA GGCTCTTTTC AGAGCCACCA CTGATCTCAA 716 TAAAAGAGCT GGATAATTTC TTTAAAAAAA AAAAAAAAAA 756 SEQ ID NO: 4
配列の長さ : 234 Array length: 234
配列の型:核酸 トポロジー:直鎖状 Sequence type: nucleic acid Topology: linear
配列の種類: cDNA Sequence type: cDNA
配列: Array:
AAG AAG CCC AAG AAG GCG GCT GGC GGC GCA ACT CCG AAG AAG AGC GCT 48 Lys Lys Pro Lys Lys Ala Ala Gly Gly Ala Thr Pro Lys Lys Ser Ala  AAG AAG CCC AAG AAG GCG GCT GGC GGC GCA ACT CCG AAG AAG AGC GCT 48 Lys Lys Pro Lys Lys Ala Ala Gly Gly Ala Thr Pro Lys Lys Ser Ala
5 10 15  5 10 15
AAG AAA ACA CCG AAG AAA GCG AAG AAG CCG GCC GCG GCC ACT GTA ACC 96 Lys Lys Thr Pro Lys Lys Ala Lys Lys Pro Ala Ala Ala Thr Val Thr  AAG AAA ACA CCG AAG AAA GCG AAG AAG CCG GCC GCG GCC ACT GTA ACC 96 Lys Lys Thr Pro Lys Lys Ala Lys Lys Pro Ala Ala Ala Thr Val Thr
20 25 30 20 25 30
AAG AAA GTG GCT AAG AGC CCA AAG AAG GCC AAG GTT GCG AAG CCC AAG 144 Lys Lys Val Ala Lys Ser Pro Lys Lys Ala Lys Val Ala Lys Pro Lys AAG AAA GTG GCT AAG AGC CCA AAG AAG GCC AAG GTT GCG AAG CCC AAG 144 Lys Lys Val Ala Lys Ser Pro Lys Lys Ala Lys Val Ala Lys Pro Lys
35 40 45  35 40 45
AAA GCT GCC AAA ACT GCT GCT AAG GCT GTG AAG CCC AAG GCC GCT AAG 192 Lys Ala Ala Lys Ser Ala Ala Lys Ala Val Lys Pro Lys Ala Ala Lys  AAA GCT GCC AAA ACT GCT GCT AAG GCT GTG AAG CCC AAG GCC GCT AAG 192 Lys Ala Ala Lys Ser Ala Ala Lys Ala Val Lys Pro Lys Ala Ala Lys
50 55 60  50 55 60
CCC AAG GTT GTC AAG CCT AAG AAG GCG GCG CCC AAG AAG AAA 234 Pro Lys Val Val Lys Pro Lys Lys Ala Ala Pro Lys Lys Lys  CCC AAG GTT GTC AAG CCT AAG AAG GCG GCG CCC AAG AAG AAA 234 Pro Lys Val Val Lys Pro Lys Lys Ala Ala Pro Lys Lys Lys
65 70 75  65 70 75

Claims

請 求 の 範 囲 The scope of the claims
1 . 配列番号: 2のアミノ酸配列を有する腫瘍抗原タンパク質の部分べプチド であって主要組織適合遺伝子複合体 (MH C ) クラス I抗原と結合して T細胞に より認識される腫瘍抗原べプチドをコ一ドするオリゴヌクレオチド分子。 1. A partial peptide of a tumor antigen protein having the amino acid sequence of SEQ ID NO: 2, which binds to a major histocompatibility complex (MHC) class I antigen and recognizes a tumor antigen peptide recognized by T cells. The oligonucleotide molecule to be coded.
2 . 配列番号: 2のアミノ酸配列中、 連続した少なくとも 7個のアミノ酸残基 を含む腫瘍抗原べプチドをコ一ドする請求項 1記載のォリゴヌクレオチド分子。 2. The oligonucleotide molecule according to claim 1, which encodes a tumor antigen peptide comprising at least 7 consecutive amino acid residues in the amino acid sequence of SEQ ID NO: 2.
3 . 腫瘍抗原タンパク質をコードしている塩基配列が配列番号: 2の塩基配列 である、 請求項 1 または 2記載のオリゴヌクレオチド分子。 3. The oligonucleotide molecule according to claim 1 or 2, wherein the nucleotide sequence encoding the tumor antigen protein is the nucleotide sequence of SEQ ID NO: 2.
4 . 腫瘍抗原タンパク質をコードしている塩基配列が、 配列番号: 2の塩基配 列において 1 もしくは複数個の塩基が置換、 欠失、 挿入または付加されたもので ある請求項 3記載のォリゴヌクレオチド分子。 4. The oligonucleotide according to claim 3, wherein the nucleotide sequence encoding the tumor antigen protein is obtained by substituting, deleting, inserting or adding one or more nucleotides in the nucleotide sequence of SEQ ID NO: 2. Nucleotide molecule.
5 . 請求項 1〜 4いずれか記載のォリゴヌクレオチド分子とストリンジヱント な条件下でハイブリダイズするオリゴヌクレオチド分子。 5. An oligonucleotide molecule that hybridizes under stringent conditions with the oligonucleotide molecule according to any one of claims 1 to 4.
6 . 配列番号: 2のアミノ酸配列を有する腫瘍抗原タンパク質をコードするポ リヌクレオチド分子のコーディング配列またはその 5 ' ノンコ一ディング配列中 の塩基配列と相補的な配列からなるオリゴヌクレオチド分子またはその化学的修 飾体。 6. An oligonucleotide molecule comprising a coding sequence of a polynucleotide molecule encoding a tumor antigen protein having the amino acid sequence of SEQ ID NO: 2 or a sequence complementary to the base sequence in the 5 ′ non-coding sequence thereof, or a chemical thereof. Decorative body.
7 . 請求項 1〜 5いずれか記載のォリゴヌクレオチドによりコードされる腫瘍 抗原べプチドまたはその誘導体。 7. A tumor antigen peptide encoded by the oligonucleotide according to any one of claims 1 to 5, or a derivative thereof.
8 . 請求項 7記載の腫瘍抗原べプチドまたはその誘導体を含有する医薬。 8. A medicament comprising the tumor antigen peptide according to claim 7 or a derivative thereof.
9 . 請求項 7記載の腫瘍抗原べプチドまたはその誘導体に対する抗体。 9. An antibody against the tumor antigen peptide of claim 7 or a derivative thereof.
1 0 . 請求項 1〜6いずれか記載のオリゴヌクレオチド分子を含有する医薬。 10. A medicament comprising the oligonucleotide molecule according to any one of claims 1 to 6.
1 1 . 請求項 1〜6いずれか記載のオリゴヌクレオチド分子を有するプラスミ 11. A plasmid having the oligonucleotide molecule according to any one of claims 1 to 6.
1 2 . 請求項 1 1記載のプラスミ ドによって形質転換された形質転換体。 12. A transformant transformed with the plasmid of claim 11.
1 3 . 配列番号: 4のアミノ酸配列を有する腫瘍抗原タンパク質の部分べプチ ドであって主要組織適合遺伝子複合体 (MH C ) クラス I抗原と結合して Τ細胞 により認識される腫瘍抗原べプチドをコードするオリゴヌクレオチド分子。 13. A partial peptide of a tumor antigen protein having the amino acid sequence of SEQ ID NO: 4, which is recognized by Τ cells by binding to the major histocompatibility complex (MH C) class I antigen An oligonucleotide molecule encoding
1 4 . 配列番号: 4のァミノ酸配列中、 連続した少なくとも 7個のァミノ酸残 基を含む腫瘍抗原べプチドをコードする請求項 1 3記載のォリゴヌクレオチド分 子。 14. The oligonucleotide molecule according to claim 13, which encodes a tumor antigen peptide comprising at least 7 consecutive amino acid residues in the amino acid sequence of SEQ ID NO: 4.
1 5 . 腫瘍抗原タンパク質をコードしている塩基配列が配列番号: 4の塩基配 列である、 請求項 1 3または 1 4記載のオリゴヌクレオチド分子。 15. The oligonucleotide molecule according to claim 13 or 14, wherein the nucleotide sequence encoding the tumor antigen protein is the nucleotide sequence of SEQ ID NO: 4.
1 6 . 腫瘍抗原タンパク質をコードしている塩基配列が、 配列番号: 4の塩基 配列において 1 もしくは複数個の塩基が置換、 欠失、 挿入または付加されたもの である請求項 1 5記載のオリゴヌクレオチド分子。 16. The oligo according to claim 15, wherein the nucleotide sequence encoding the tumor antigen protein is obtained by substituting, deleting, inserting or adding one or more nucleotides in the nucleotide sequence of SEQ ID NO: 4. Nucleotide molecule.
1 7 . 請求項 1 3〜 1 6いずれか記載のオリゴヌクレオチド分子とストリンジ ェントな条件下でハイブリダイズするオリゴヌクレオチド分子。 17. An oligonucleotide molecule that hybridizes with the oligonucleotide molecule according to any one of claims 13 to 16 under stringent conditions.
1 8. 配列番号: 4のアミノ酸配列を有する腫瘍抗原タンパク質をコードする ポリヌクレオチド分子のコーディング配列またはその 5' ノンコ一ディング配列 中の塩基配列と相補的な配列からなるオリゴヌクレオチド分子またはその化学的 修飾体。 1 8. Oligonucleotide molecule consisting of a sequence complementary to the nucleotide sequence in the coding sequence of the polynucleotide molecule encoding the tumor antigen protein having the amino acid sequence of SEQ ID NO: 4 or its 5 'non-coding sequence or its chemical Modifier.
1 9. 請求項 1 3〜1 7いずれか記載のオリゴヌクレオチドによりコードされ る腫瘍抗原べプチドまたはその誘導体。 1 9. A tumor antigen peptide encoded by the oligonucleotide according to any one of claims 13 to 17, or a derivative thereof.
2 0. 請求項 1 9記載の腫瘍抗原ペプチドまたはその誘導体を含有する医薬。 20. A medicament comprising the tumor antigen peptide according to claim 19 or a derivative thereof.
2 1. 請求項 1 9記載の腫瘍抗原ペプチドまたはその誘導体に対する抗体。 2 1. An antibody against the tumor antigen peptide of claim 19 or a derivative thereof.
2 2. 請求項 1 3〜 1 8いずれか記載のオリゴヌクレオチド分子を含有する医 2 2. A medicine containing the oligonucleotide molecule according to any one of claims 13 to 18
2 3. 請求項 1 3〜1 8いずれか記載のオリゴヌクレオチド分子を有するブラ スミ ド。 23. A plasmid having the oligonucleotide molecule according to any one of claims 13 to 18.
2 4. 請求項 2 3記載のプラスミ ドによって形質転換された形質転換体。 2 4. A transformant transformed by the plasmid of claim 23.
PCT/JP1997/004182 1996-11-21 1997-11-17 Human cancer regression antigen protein WO1998022582A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU49667/97A AU4966797A (en) 1996-11-21 1997-11-17 Human cancer regression antigen protein

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP32778296 1996-11-21
JP8/327782 1996-11-21

Publications (1)

Publication Number Publication Date
WO1998022582A1 true WO1998022582A1 (en) 1998-05-28

Family

ID=18202930

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP1997/004182 WO1998022582A1 (en) 1996-11-21 1997-11-17 Human cancer regression antigen protein

Country Status (2)

Country Link
AU (1) AU4966797A (en)
WO (1) WO1998022582A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH01104193A (en) * 1986-06-10 1989-04-21 Kyowa Hakko Kogyo Co Ltd Novel polypeptide
WO1996040917A1 (en) * 1995-06-07 1996-12-19 Yale University NOVEL NuMA-INTERACTING PROTEINS AND METHODS FOR THEIR IDENTIFICATION

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH01104193A (en) * 1986-06-10 1989-04-21 Kyowa Hakko Kogyo Co Ltd Novel polypeptide
WO1996040917A1 (en) * 1995-06-07 1996-12-19 Yale University NOVEL NuMA-INTERACTING PROTEINS AND METHODS FOR THEIR IDENTIFICATION

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
CANCER RES., 54, (1994), PETER SCHRAML et al., "Identification of Genes Differentially Expressed in Normal Lung and Non-Small Cell Lung Carcinoma Tissue", pages 5236-5240. *
CANCER RES., 55, (1995), K. ITOH et al., "HLA A2601-Restricted CTLs Recognize a Peptide Antigen Expressed on Squamous Cell Carcinoma", pages 4248-4252. *
EUR. J. BIOCHEM., 223, (1994), D. DOENECKE et al., "Role of a Distal Promoter Element in the S-Phase Control of the Human H1.2 Histone Gene Transcription", pages 567-574. *
EUR. J. CELL. BIOL., 49(1), (1989), STEFAN EICK et al., "Human H1 Histones Conserved and Varied Sequence Elements in Two H1 Subtype Genes", pages 110-115. *
EUR. J. IMMUNOL., 21(11), (1991), CLAUDE AURIAULT et al., "Effect Ofubiquitin on Platelet Function: Possible Identify with Platelet Activity Suppressive Lymphokinase (PASL)", pages 2735-2741. *
J. BIOL. CHEM., 260(12), (1985), P. KAY LUND et al., "Nucleotide Sequence Analysis of a cDNA Encoding Human Ubiquitin Reveals that Ubiquitin is Synthesized as a Precursor", pages 7609-7613. *
SCIENCE, 254, (1991), C. TRAVERSARI et al., "A Gene Encoding an Antigen Recognized by Cytolytic T Lymphocytes on a Human Meianoma", pages 1643-1647. *

Also Published As

Publication number Publication date
AU4966797A (en) 1998-06-10

Similar Documents

Publication Publication Date Title
EP1536009B1 (en) Hla-a24-restricted cancer antigen peptide
JP4904384B2 (en) Novel tumor antigen protein SART-3 and its tumor antigen peptide
KR100581508B1 (en) Tumor antigen peptide derivatives
US6815531B1 (en) Tumor antigen proteins, genes thereof, and tumor antigen peptides
JP4138073B2 (en) Human cancer regression antigen protein
WO1998022582A1 (en) Human cancer regression antigen protein
KR20010080661A (en) Novel tumor antigen protein art-1 and tumor antigen peptide thereof
JP2000116383A (en) Human cancer regression antigen protein
KR100465546B1 (en) Tumor Antigen Proteins, Genes and Tumor Antigen Peptides thereof
AU748657B2 (en) Tumor antigen protein, gene thereof, and utilization thereof
US6444800B1 (en) Human gastric cancer antigen gene and gastric cancer antigen protein
AU3411097A (en) Brain glycogen phosphorylase cancer antigen
WO2000002907A1 (en) Tumor antigen peptide originating in sart-1

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH HU ID IL IS JP KE KG KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH KE LS MW SD SZ UG ZW AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL

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
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA