WO2010074049A1 - 抗hs6st2抗体及びその用途 - Google Patents
抗hs6st2抗体及びその用途 Download PDFInfo
- Publication number
- WO2010074049A1 WO2010074049A1 PCT/JP2009/071271 JP2009071271W WO2010074049A1 WO 2010074049 A1 WO2010074049 A1 WO 2010074049A1 JP 2009071271 W JP2009071271 W JP 2009071271W WO 2010074049 A1 WO2010074049 A1 WO 2010074049A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- antibody
- hs6st2
- amino acid
- seq
- acid sequence
- Prior art date
Links
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/30—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/40—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/574—Immunoassay; Biospecific binding assay; Materials therefor for cancer
- G01N33/57407—Specifically defined cancers
- G01N33/57423—Specifically defined cancers of lung
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/574—Immunoassay; Biospecific binding assay; Materials therefor for cancer
- G01N33/57407—Specifically defined cancers
- G01N33/57438—Specifically defined cancers of liver, pancreas or kidney
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/574—Immunoassay; Biospecific binding assay; Materials therefor for cancer
- G01N33/57407—Specifically defined cancers
- G01N33/57449—Specifically defined cancers of ovaries
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/73—Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/73—Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
- C07K2317/732—Antibody-dependent cellular cytotoxicity [ADCC]
Definitions
- the present invention relates to an antibody that binds to HS6ST2 protein and use thereof. More specifically, the present invention relates to an anti-HS6ST2 antibody conjugated with a cytotoxic substance that can be used as an anticancer agent.
- Proteoglycan is a glycoprotein formed by covalently binding a sugar chain (glycosaminoglycan) and protein (Non-patent Document 1).
- Glycosaminoglycans are polysaccharides whose basic disaccharide structure is usually repeated 40-100 times, and are characterized by being sulfated to various degrees.
- Glycosaminoglycans include chondroitin sulfate, dermatan sulfate, heparan sulfate, heparin, and keratan sulfate.
- heparan sulfate-bound proteoglycan is called heparan sulfate proteoglycan (HSPG), and syndecan and glypican are known to be expressed on the cell membrane, and the basement membrane Perlecan, agrin and the like are known to be secreted into (basement membrane).
- Heparan sulfate is highly expressed in the lungs and kidneys in mice and is less expressed in skeletal muscle, liver, skin, and brain (Non-patent Document 2).
- HSPG is known to influence the activity of growth factors and to be involved in cell proliferation and differentiation.
- Non-Patent Documents 3, 4, 5 fibroblast growth factor, heparin-binding epidermal growth factor-like ⁇ growth ⁇ factor, and amphiregulin transmit signals to cells through receptors for each growth factor while binding to heparan sulfate. It has been reported that HSPG is also involved in cancer cell proliferation and metastasis in cancer (Non-Patent Documents 7 and 8).
- Heparan-sulfate-6-O-sulfotransferase-2 is an enzyme that adds a sulfate group to the 6-O position of glucosamine that constitutes heparan sulfate.
- HS6ST1 and HS6ST3 are known as similar enzymes (sulfotransferase) (Non-patent Document 9).
- These HS6ST families are type II membrane proteins, but are present in intracellular Golgi and function as enzymes (Non-patent Document 10).
- HS6ST1 is secreted extracellularly by being cleaved in the vicinity of the transmembrane region (Non-Patent Documents 11, 12, and 13).
- Non-Patent Document 14 mouse HS6ST2 (mHS6ST2) is forcibly expressed in CHO cells, it is also secreted extracellularly (Non-patent Document 15).
- mHS6ST2 that remains in the Golgi and secreted mHS6ST2 have the same molecular weight, and mouse HS6ST3 may cleave the N-terminal region including the transmembrane region as a signal peptide
- mouse HS6ST3 may cleave the N-terminal region including the transmembrane region as a signal peptide
- Non-patent Document 15 a variant of mHS6ST2 whose N-terminal side is 146 amino acids long is not secreted extracellularly.
- 3′-phosphoadenosinephospho5′-phosphosulfate which is a donor of sulfate group is rapidly degraded in blood, it is considered that HS6ST2 secreted extracellularly does not work as an enzyme.
- Non-patent Document 16 Although the three-dimensional structure of HS6ST2 has not been elucidated, it is thought to recognize and bind to a partial sequence of heparan sulfate having 6 sugar chains or less (Non-patent Document 16).
- Membrane type HSPG is expressed in almost all cells. The expression level is about 10 5 -10 6 molecules per cell, most of which is taken up into cells with a half-life of 3-8 hours and degraded by lysosomes (Non-patent Document 18). It is known that peptides such as HIV-Tat and bFGF, nucleic acids such as polylysine-DNA complexes, polyamines, or anti-HSPG antibodies are actually taken into cells via HSPG (Non-patent Documents 19 and 20). , 21).
- An object of the present invention is to provide a novel anti-HS6ST2 antibody. Another object of the present invention is to clarify the physiological action of the obtained anti-HS6ST2 antibody and to use it for diagnosis and treatment of diseases, particularly cancer.
- the present inventors have produced a novel anti-HS6ST2 antibody having cytotoxic activity, and found that the antibody is useful for diagnosis and treatment of cancer. Was completed.
- the present invention provides the following.
- [5] The antibody according to any one of [1] to [3], which binds to HS6ST2 expressed on a cell membrane.
- [7] The region from the 379th amino acid to the 459th amino acid, the region from the 308th amino acid to the 393rd amino acid, or the 24th amino acid to the 175th amino acid of the HS6ST2 protein having the amino acid sequence of SEQ ID NO: 107
- a pharmaceutical composition comprising the antibody according to any one of [1] to [7] as an active ingredient.
- the pharmaceutical composition according to [8] which is an anticancer agent.
- a method for diagnosing cancer comprising the following steps: (a) providing a sample collected from the subject; (b) A step of detecting HS6ST2 protein or HS6ST2 gene contained in the sample of (a). [11] HS6ST2 protein to which a cytotoxic substance is bound. [12] A pharmaceutical composition comprising the HS6ST2 protein according to [11] as an active ingredient. [13] The pharmaceutical composition according to [12], which is an anticancer agent.
- the present inventors made an anti-HS6ST2 antibody and examined cancer diagnosis and treatment methods using this antibody. It has been found that the anti-HS6ST2 antibody of the present invention kills cells expressing HS6ST2 on the cell membrane by an antibody-dependent cellular cytotoxicity (ADCC) activity. In addition, cell proliferation was suppressed in the presence of a secondary antibody conjugated with toxin. This indicates that anti-HS6ST2 antibody having ADCC activity and immunotoxin targeting HS6ST2 are useful for cancer treatment.
- HS6ST2 has two variants with different N-terminal amino acid lengths (UniProt Q96MM7-1, Q96MM7-2), and the obtained antibody bound to both variants.
- HS6ST2 protein in cancer cells was evaluated using an anti-HS6ST2 antibody.
- Analysis using flow cytometry revealed that lung and hepatoma cell lines express HS6ST2 on the cell membrane.
- Western blot analysis revealed that lung and ovarian cancer cell lines express HS6ST2. Judging from the molecular weight, these HS6ST2s were considered to be variants (UniProt Q96MM7-2) lacking the N-terminal 146 amino acids.
- Analysis using immunohistochemical staining revealed that HS6ST2 was expressed on cell membranes in clinical lung adenocarcinoma and clinical lung squamous cell carcinoma.
- Analysis by Western blot revealed that lung and ovarian cancer cell lines secrete HS6ST2 in the culture supernatant.
- soluble HS6ST2 binds to heparan sulfate on the cell membrane.
- soluble HS6ST2 binds to heparan sulfate on the cell membrane.
- the anti-HS6ST2 antibody suppressed cell growth in the presence of a secondary antibody conjugated with toxin. Therefore, it was shown that immunotoxin targeting soluble HS6ST2 is useful for the treatment of cancer.
- Clinical lung cancer also expresses HSPG (Glycosaminoglycansaminoin human lung cancer. Cancer. 1981. 48: 2016; Heparan sulfate proteoglycan expression in human lung-cancer cells.
- a method for diagnosing cancer using soluble HS6ST2 was examined. Using the obtained anti-HS6ST2 antibody, an ELISA system capable of detecting soluble HS6ST2 in the order of ng / mL was established. Since this ELISA system was measurable even in the presence of human serum, it was shown that cancer can be diagnosed using blood as a specimen.
- FIG. 1A is a diagram showing an expression profile of HS6ST2 in normal tissues.
- FIG. 1B is a diagram showing an expression profile of HS6ST2 in normal tissues.
- FIG. 2A is a diagram showing an expression profile of HS6ST2 in a tumor part of a lung cancer cell line and a lung cancer removed tissue.
- the AF on the vertical axis represents the lung adenocarcinoma cell line, large cell lung cancer cell line, lung squamous cell carcinoma cell line, small cell lung cancer cell line, tumor part of small cell lung cancer isolated tissue, and tumor part of lung adenocarcinoma isolated tissue, respectively. Indicates.
- FIG. 1A is a diagram showing an expression profile of HS6ST2 in normal tissues.
- FIG. 1B is a diagram showing an expression profile of HS6ST2 in normal tissues.
- FIG. 2A is a diagram showing an expression profile of HS6ST2 in a tumor part of a lung cancer cell line and a lung cancer removed
- FIG. 2B is a view showing an expression profile of HS6ST2 in a tumor part of a lung cancer cell line and a lung cancer-extracted tissue.
- the AF on the vertical axis represents the lung adenocarcinoma cell line, large cell lung cancer cell line, lung squamous cell carcinoma cell line, small cell lung cancer cell line, tumor part of small cell lung cancer isolated tissue, and tumor part of lung adenocarcinoma isolated tissue, respectively.
- FIG. 3A is a diagram showing an expression profile of HS6ST2 in cell lines other than lung cancer.
- A-H on the vertical axis represents a colon cancer cell line, stomach cancer cell line, breast cancer cell line, ovarian cancer cell line, endometrial cancer cell line, lymphoma cell line, myeloma cell line, and liver cancer cell line, respectively.
- FIG. 3B is a diagram showing an expression profile of HS6ST2 in cell lines other than lung cancer.
- A-H on the vertical axis represents a colon cancer cell line, stomach cancer cell line, breast cancer cell line, ovarian cancer cell line, endometrial cancer cell line, lymphoma cell line, myeloma cell line, and liver cancer cell line, respectively.
- FIG. 4 is a diagram showing the results of flow cytometry analysis for evaluating the binding of anti-HS6ST2 antibodies to HS6ST2_N-short_ctV5_CHO and mHS6ST2_ctV5_CHO.
- the solid line is anti-HS6ST2 antibody, and the gray line is mIgG1.
- FIG. 5 is a diagram showing the results of flow cytometry analysis for evaluating the binding of anti-HS6ST2 antibodies A6 and C8 to HS6ST2_N-short_ctV5_CHO and mHS6ST2_ctV5_CHO. ⁇ ; HS6ST2_N-short_ctV5_CHO, ⁇ ; mHS6ST2_ctV5_CHO.
- FIG. 6 shows the results of evaluating ADCC activity of anti-HS6ST2 antibody against HS6ST2_N-short_ctV5_CHO.
- FIG. 7 is a graph showing the results of evaluating the growth inhibitory activity of anti-HS6ST2 antibody against HS6ST2_N-short_ctV5_CHO using Mab-ZAP.
- FIG. 8 is a diagram showing the results of flow cytometry analysis for evaluating the binding of anti-HS6ST2 antibodies to HS6ST2_N-short_ctV5_CHO and HS6ST2_N-long_ctV5_CHO.
- the solid line is anti-HS6ST2 antibody, and the gray line is mIgG1.
- FIG. 9 is a diagram showing the results of flow cytometry analysis in which the binding of anti-HS6ST2 antibody B6 to liver cancer cell line HuH6 and lung adenocarcinoma cell line ABC-1 was evaluated.
- the solid line is anti-HS6ST2 antibody B6, and the gray line is mIgG1.
- FIG. 10 shows the results of Western blotting of HS6ST2_N-short_ctV5_CHO and HS6ST2_N-long_ctV5_CHO using anti-HS6ST2 antibody C10 and anti-V5 tag antibody.
- FIG. 11 shows the results of Western blotting of cancer cell lines using anti-HS6ST2 antibody C10.
- FIG. 12 is a diagram showing the results of flow cytometry analysis for evaluating the expression of heparan sulfate on the cell membrane.
- the solid line is anti-heparan sulfate antibody, and the gray line is mIgM.
- FIG. 13 is a diagram showing the results of flow cytometry analysis for evaluating the binding of sHS6ST2_FLAG to heparan sulfate.
- heparan sulfate in DG44 cells was degraded with an enzyme, and the expression level of heparan sulfate was compared with and without enzyme treatment.
- the solid line is anti-heparan sulfate antibody, and the gray line is mIgM.
- the binding of sHS6ST2_FLAG to DG44 cells was compared with and without enzyme treatment.
- the solid line is anti-HS6ST2 antibody C8, and the gray line is mIgG1.
- FIG. 14 shows the binding of sHS6ST2_FLAG to cancer cell line A549 and the antitumor activity of anti-HS6ST2 antibody C8 in the presence of Mab-ZAP.
- FIG. 1 shows A549 cells reacted with sHS6ST2_FLAG at 100 ⁇ g / mL (thick line), 20 ⁇ g / mL (solid line), 4 ⁇ g / mL (dotted line), 0.8 ⁇ g / mL (dashed line), 0 ⁇ g / mL (solid line in gray) It is a figure which shows the result of having performed the flow cytometry analysis using the anti- HS6ST2 antibody C8 after having made it.
- FIG. 15 is a diagram showing a calibration curve of an ELISA system for detecting soluble HS6ST2.
- FIG. 16 is a diagram showing that heparan sulfate and human serum do not affect the ELISA system.
- Figure 17 shows the results of immunohistochemical staining that analyzed the expression of HS6ST2 protein in clinical lung adenocarcinoma (A), clinical lung squamous cell carcinoma (B), normal tissue adjacent to lung cancer (C), and skin (D).
- A clinical lung adenocarcinoma
- B clinical lung squamous cell carcinoma
- C normal tissue adjacent to lung cancer
- D skin
- the HS6ST2 protein used in the present invention is not particularly limited, and HS6ST2 protein known to those skilled in the art can be used.
- the HS6ST2 protein is preferably human HS6ST2.
- human HS6ST2 has a plurality of variants, HS6ST2 used in the present invention may be any variant.
- HS6ST2 variants include variants lacking exons 4 and 5 (GenBank Accession No: NM_147175, UniProt: Q96MM7-1, SEQ ID NO: 106), variants lacking amino acids 1 to 146 of Q96MM7-1 ( UniProt: Q96MM7-2, SEQ ID NO: 107), and third variant (UniProt: Q96MM7-3, SEQ ID NO: 108).
- HS6ST2 may be HS6ST2 protein expressed on the cell membrane or secreted HS6ST2 secreted.
- Anti-HS6ST2 antibody used in the present invention may be bound to the HS6ST2 protein, and its origin, type, shape, etc. are not limited. Specifically, known antibodies such as non-human animal antibodies (eg, mouse antibodies, rat antibodies, camel antibodies), human antibodies, chimeric antibodies, and humanized antibodies can be used. In the present invention, monoclonal or polyclonal antibodies can be used as antibodies, but monoclonal antibodies are preferred.
- the binding of the antibody to the HS6ST2 protein is preferably specific binding.
- the HS6ST2 protein recognized by the anti-HS6ST2 antibody of the present invention is not particularly limited, and examples thereof include HS6ST2 protein expressed on the cell membrane, HS6ST2 protein secreted from the cell, HS6ST2 protein bound to heparan sulfate, and the like.
- Heparan sulfate is generally a glycosaminoglycan obtained by subjecting the disaccharide unit repeating structure of D-glucuronic acid and D-glucosamine to N-sulfation and O-sulfation modification.
- the heparan sulfate is preferably heparan sulfate contained in heparan sulfate proteoglycan, and more preferably heparan sulfate contained in heparan sulfate proteoglycan on the cancer cell membrane.
- an antibody that binds to HS6ST2 protein bound to heparan sulfate on the cancer cell membrane can be exemplified.
- the anti-HS6ST2 antibody used in the present invention can be obtained as a polyclonal or monoclonal antibody using known means.
- a monoclonal antibody derived from a mammal is particularly preferable.
- Mammal-derived monoclonal antibodies include those produced by hybridomas and those produced by hosts transformed with expression vectors containing antibody genes by genetic engineering techniques.
- Monoclonal antibody-producing hybridomas can be basically produced using known techniques as follows. First, HS6ST2 protein is used as a sensitizing antigen, and this is immunized according to a normal immunization method. Immune cells obtained from an immunized animal are fused with a known parent cell by a conventional cell fusion method to obtain a hybridoma. Furthermore, from this hybridoma, a hybridoma producing an anti-HS6ST2 antibody can be selected by screening cells producing the target antibody by a usual screening method.
- HS6ST2 protein used as a sensitizing antigen for antibody acquisition can be obtained. That is, after inserting a gene sequence encoding HS6ST2 into a known expression vector and transforming an appropriate host cell, the target human HS6ST2 protein is purified from the host cell or culture supernatant by a known method. it can. Purified natural HS6ST2 protein can also be used as well. Further, a fusion protein obtained by fusing a desired partial polypeptide of HS6ST2 protein with a different polypeptide can also be used as an immunogen.
- a fusion protein as an immunogen, for example, an Fc fragment of an antibody, a peptide tag, or the like can be used.
- a vector that expresses the fusion protein can be prepared by fusing genes encoding two or more desired polypeptide fragments in-frame and inserting the fusion gene into an expression vector. The method for producing the fusion protein is described in Molecular® Cloning® 2nd® ed. (Sambrook, Jet et al., “Molecular® Cloning® 2nd® ed.,” 9.47-9.58, “Cold® Spring® Harbor® Lab.® press,” 1989).
- the HS6ST2 protein thus purified can be used as a sensitizing antigen used for immunization against mammals.
- a partial peptide of HS6ST2 can also be used as a sensitizing antigen.
- the following peptides can be used as the sensitizing antigen.
- the region and size of HS6ST2 used as a partial peptide are not limited.
- the number of amino acids constituting the peptide to be sensitized antigen is preferably at least 3 or more, for example, 5 or more, or 6 or more. More specifically, a peptide having 8 to 50, preferably 10 to 30 residues can be used as a sensitizing antigen.
- the mammal immunized with the sensitizing antigen is not particularly limited.
- an immunized animal in consideration of compatibility with a parent cell used for cell fusion.
- rodent animals are preferred as immunized animals. Specifically, mice, rats, hamsters, or rabbits can be used as immunized animals.
- monkeys and the like can be used as immunized animals.
- the above animals can be immunized with a sensitizing antigen.
- mammals can be immunized by injecting a sensitizing antigen intraperitoneally or subcutaneously. Specifically, the sensitizing antigen is administered to mammals several times every 4 to 21 days.
- the sensitizing antigen is diluted with PBS (Phosphate-Buffered Saline) or physiological saline at an appropriate dilution ratio and used for immunization.
- a sensitizing antigen can be administered with an adjuvant. For example, it can be mixed with Freund's complete adjuvant and emulsified to give a sensitizing antigen.
- An appropriate carrier can be used for immunization with the sensitizing antigen.
- a partial peptide having a small molecular weight is used as a sensitizing antigen, it is desirable to immunize the sensitizing antigen peptide by binding it to a carrier protein such as albumin or keyhole limpet hemocyanin.
- DNA immunization refers to immunization by administering a vector DNA constructed in such a manner that a gene encoding an antigen protein can be expressed in an immunized animal, and expressing the immunizing antigen in the body of the immunized animal. It is a method of giving a stimulus.
- DNA immunization can be expected to have the following advantages. ⁇ Implement immune stimulation by maintaining the structure of membrane proteins such as HS6ST2. -There is no need to purify immune antigens.
- DNA expressing HS6ST2 protein is first administered to an immunized animal.
- DNA encoding HS6ST2 can be synthesized by a known method such as PCR.
- the obtained DNA is inserted into an appropriate expression vector and administered to an immunized animal.
- the expression vector for example, a commercially available expression vector such as pcDNA3.1 can be used.
- a method of administering the vector to a living body a generally used method can be used.
- DNA immunization can be performed by driving gold particles adsorbed with an expression vector into cells with a gene gun.
- immune cells are collected from the mammal and subjected to cell fusion.
- spleen cells can be used.
- Mammalian myeloma cells are used as cells to be fused with the above immune cells.
- the myeloma cell is preferably provided with an appropriate selection marker for screening.
- a selectable marker refers to a trait that can (or cannot) survive under certain culture conditions.
- Known selection markers include hypoxanthine-guanine-phosphoribosyltransferase deficiency (hereinafter abbreviated as HGPRT deficiency) or thymidine kinase deficiency (hereinafter abbreviated as TK deficiency).
- HGPRT deficiency hypoxanthine-guanine-phosphoribosyltransferase deficiency
- TK deficiency thymidine kinase deficiency
- Cells having HGPRT or TK deficiency have hypoxanthine-aminopterin-thymidine sensitivity (hereinafter abbreviated as HAT sensitivity).
- HGPRT-deficient and TK-deficient cells can be selected in media containing 6 thioguanine, 8 azaguanine (hereinafter abbreviated as 8AG), or 5 'bromodeoxyuridine, respectively.
- 8AG 8 azaguanine
- Normal cells die because they incorporate these pyrimidine analogs into the DNA, but cells deficient in these enzymes cannot survive these pyrimidine analogs and can survive in selective media.
- a selectable marker called G418 resistance confers resistance to 2-deoxystreptamine antibiotics (gentamicin analogs) with a neomycin resistance gene.
- Various myeloma cells suitable for cell fusion are known. For example, the following myeloma cells can be used for the production of the monoclonal antibody in the present invention.
- cell fusion can be carried out in a normal nutrient culture medium in the presence of a cell fusion promoter.
- a cell fusion promoter for example, polyethylene glycol (PEG), Sendai virus (HVJ) or the like can be used.
- an auxiliary agent such as dimethyl sulfoxide can be added as desired in order to increase the fusion efficiency.
- the usage ratio of immune cells and myeloma cells can be set arbitrarily.
- the number of immune cells is preferably 1 to 10 times that of myeloma cells.
- the culture solution used for cell fusion for example, RPMI1640 culture solution suitable for growth of myeloma cell line, MEM culture solution, and other normal culture solutions used for this type of cell culture can be used.
- serum supplements such as fetal calf serum (FCS) can be added to the culture medium.
- a predetermined amount of immune cells and myeloma cells are mixed well in a culture solution, and a target PEG (hybridoma) is formed by mixing a PEG solution preheated to about 37 ° C.
- a target PEG hybrida
- PEG having an average molecular weight of about 1000 to 6000 can be usually added at a concentration of 30 to 60% (w / v).
- cell fusion agents and the like that are undesirable for the growth of hybridomas are removed by sequentially adding the appropriate culture medium listed above, and then centrifuging to remove the supernatant.
- the hybridoma obtained in this manner can be selected by using a selective culture solution corresponding to the selection marker possessed by the myeloma used for cell fusion.
- a selective culture solution corresponding to the selection marker possessed by the myeloma used for cell fusion.
- cells having HGPRT or TK deficiency can be selected by culturing in a HAT culture solution (a culture solution containing hypoxanthine, aminopterin and thymidine). That is, when HAT-sensitive myeloma cells are used for cell fusion, cells that have succeeded in cell fusion with normal cells can be selectively proliferated in the HAT culture solution.
- the culture using the HAT culture solution is continued for a time sufficient for cells other than the target hybridoma (non-fusion cells) to die.
- the target hybridoma can be selected by culturing for several days to several weeks. Subsequently, by carrying out the usual limiting dilution method, screening and single cloning of the hybridoma producing the target antibody can be performed.
- an antibody that recognizes HS6ST2 can be prepared by the method described in International Publication WO03 / 104453.
- Screening and single cloning of the target antibody can be suitably performed by a screening method based on a known antigen-antibody reaction.
- the antigen is bound to a carrier such as beads made of polystyrene or the like, or a commercially available 96-well microtiter plate, and reacted with the culture supernatant of the hybridoma.
- a secondary antibody labeled with an enzyme is reacted. If the culture supernatant contains an antibody of interest that reacts with the sensitizing antigen, the secondary antibody binds to the carrier via this antibody. By detecting the secondary antibody that finally binds to the carrier, it can be determined whether the antibody of interest is present in the culture supernatant.
- substantially the same HS6ST2 protein can be preferably used as the antigen, including those used for immunization.
- cell lines expressing HS6ST2, soluble HS6ST2 and the like can be used as antigens.
- a target antibody can be obtained by sensitizing human lymphocytes with an antigen. Specifically, first, human lymphocytes are sensitized with HS6ST2 protein in vitro. The immunized lymphocytes are then fused with an appropriate fusion partner. As the fusion partner, for example, a myeloma cell derived from human and having a permanent division ability can be used (see Japanese Patent Publication No. 1-59878).
- the anti-HS6ST2 antibody obtained by this method is a human antibody having binding activity to HS6ST2 protein.
- anti-HS6ST2 can be obtained by administering an HS6ST2 protein as an antigen to a transgenic animal having all repertoires of human antibody genes or by immunizing with DNA constructed to express HS6ST2 in the animal.
- Human antibodies can also be obtained.
- Antibody-producing cells of the immunized animal can be immortalized by treatment such as cell fusion with an appropriate fusion partner or Epstein-Barr virus infection. From the immortalized cells thus obtained, a human antibody against the HS6ST2 protein can be isolated (see International Publications WO 94/25585, WO 93/22727, WO 92/03918, WO 94/02602).
- the immortalized cells by cloning the immortalized cells, it is possible to clone cells that produce an antibody having the desired reaction specificity.
- the animal's immune system recognizes human HS6ST2 as a foreign substance. Therefore, a human antibody against human HS6ST2 can be easily obtained.
- the hybridoma producing the monoclonal antibody thus produced can be subcultured in a normal culture solution.
- the hybridoma can also be stored for a long time in liquid nitrogen.
- the hybridoma can be cultured according to a usual method, and the target monoclonal antibody can be obtained from the culture supernatant.
- a hybridoma can be administered to a mammal compatible therewith to proliferate and a monoclonal antibody can be obtained as its ascites.
- the former method is suitable for obtaining a highly pure antibody.
- an antibody encoded by an antibody gene cloned from an antibody-producing cell can also be used.
- the cloned antibody gene can be expressed as an antibody by incorporating it into an appropriate vector and introducing it into a host. Methods for isolation of antibody genes, introduction into vectors, and transformation of host cells have already been established (eg, Vandamme, A. M. et al., Eur.J. Biochem. (1990)). 192, 767-775).
- a cDNA encoding the variable region (V region) of an anti-HS6ST2 antibody can be obtained from a hybridoma cell that produces the anti-HS6ST2 antibody.
- total RNA is extracted from the hybridoma.
- the following method can be used. ⁇ Guanidine ultracentrifugation (Chirgwin, J. M. et al., Biochemistry (1979) 18, 5294-5299) ⁇ AGPC method (Chomczynski, P. et al., Anal. Biochem. (1987) 162, 156-159)
- Extracted mRNA can be purified using mRNA “Purification” Kit (manufactured by GE Healthcare Bioscience) or the like.
- kits for extracting total mRNA directly from cells such as QuickPrep mRNA Purification Kit (manufactured by GE Healthcare Bioscience) are also commercially available.
- total mRNA can also be obtained from the hybridoma.
- cDNA encoding the antibody V region can be synthesized using reverse transcriptase. In this case, any 15-30 base sequence selected from sequences common to antibody genes can be used as a primer.
- cDNA can be synthesized by AMV Reverse Transcriptase First-strand cDNA Synthesis Kit (manufactured by Seikagaku Corporation).
- AMV Reverse Transcriptase First-strand cDNA Synthesis Kit manufactured by Seikagaku Corporation.
- 5'-Ampli FINDER RACE Kit (Clontech) and 5'-RACE method using PCR (Frohman, M. A. et al., Proc. Natl. Acad. Sci. USA (1988) 85, 8998-9002, Belyavsky, A. et al., Nucleic Acids Res. (1989) 17, 2919-2932).
- appropriate restriction enzyme sites described later can be introduced at both ends of the cDNA.
- the desired cDNA fragment is purified from the obtained PCR product and then ligated with vector DNA.
- a recombinant vector is produced, introduced into Escherichia coli or the like and a colony is selected, a desired recombinant vector can be prepared from Escherichia coli that has formed the colony. Then, the base sequence of the cDNA can be confirmed by a known method such as the dideoxynucleotide chain termination method.
- a cDNA library can be used to obtain a gene encoding the variable region of an antibody.
- cDNA is synthesized using mRNA extracted from antibody-producing cells as a template to obtain a cDNA library. It is convenient to use a commercially available kit for the synthesis of the cDNA library. Actually, the amount of mRNA obtained from only a small number of cells is extremely small, and the yield is low when it is directly purified. Therefore, it is usually purified after adding carrier RNA that is apparently free of antibody genes. Alternatively, when a certain amount of RNA can be extracted, it is possible to efficiently extract only RNA of antibody-producing cells. For example, carrier RNA may not be necessary for RNA extraction from 10 or more, 30 or more, preferably 50 or more antibody-producing cells.
- Antibody gene is amplified by PCR using the obtained cDNA library as a template.
- Primers for amplifying antibody genes by PCR are known.
- primers for human antibody gene amplification can be designed based on the disclosure of a paper (J. Mol. Biol. (1991) 222, 581-597). These primers have different nucleotide sequences for each immunoglobulin subclass. Therefore, when a cDNA library whose subclass is unknown is used as a template, the PCR method is performed in consideration of all possibilities.
- a primer capable of amplifying genes encoding ⁇ 1 to ⁇ 5 as a heavy chain and ⁇ chain and ⁇ chain as a light chain may be used. it can.
- a primer that anneals to a portion corresponding to the hinge region is generally used as the 3′-side primer.
- a primer corresponding to each subclass can be used as the 5′-side primer.
- PCR products using primers for gene amplification of heavy and light chain subclasses should be independent libraries.
- an immunoglobulin comprising a combination of a heavy chain and a light chain can be reconstructed.
- the target antibody can be screened using the binding activity of the reconstituted immunoglobulin to HS6ST2 as an index.
- the antibody gene can be incorporated into an expression vector so as to be expressed under the control of the expression control region.
- An expression control region for expressing an antibody includes, for example, an enhancer and a promoter. Subsequently, by transforming an appropriate host cell with this expression vector, a recombinant cell expressing a DNA encoding an anti-HS6ST2 antibody can be obtained.
- DNAs encoding antibody heavy chains (H chains) and light chains (L chains) can be incorporated into separate expression vectors.
- An antibody molecule having an H chain and an L chain can be expressed by co-transfecting a vector incorporating the H chain and the L chain into the same host cell at the same time.
- DNA encoding the H chain and L chain may be incorporated into a single expression vector to transform host cells (see International Publication WO94 / 11523).
- a host and an expression vector for producing an antibody by introducing an isolated antibody gene into a suitable host are known. Any of these expression systems can be applied to the present invention.
- animal cells, plant cells, or fungal cells can be used.
- the following cells can be exemplified as animal cells that can be used in the present invention.
- Mammalian cells CHO, COS, myeloma, BHK (baby hamster kidney), Hela, Vero, HEK293, Ba / F3, HL-60, Jurkat, SK-HEP1, etc.
- Amphibian cells Xenopus oocytes and the like.
- Insect cells sf9, sf21, Tn5, etc.
- an antibody gene expression system using cells from the genus Nicotiana such as Nicotiana tabacum is known.
- Callus cultured cells can be used for transformation of plant cells.
- Yeast genus Saccharomyces such as Saccharomyces serevisiae, Pichia genus fungus such as methanol-utilizing yeast (Pichia pastoris): Aspergillus genus such as Aspergillus niger.
- antibody gene expression systems using prokaryotic cells are also known.
- bacterial cells such as E. coli and Bacillus subtilis can be used in the present invention.
- the promoter / enhancer includes human cytomegalovirus early promoter / enhancer (human cytomegalovirus immediate-promoter / enhancer).
- a promoter / enhancer derived from a mammalian cell such as a viral promoter / enhancer or human elongation factor 1 ⁇ (HEF1 ⁇ ) can be used for antibody expression.
- viruses that can utilize promoters / enhancers include retroviruses, polyomaviruses, adenoviruses, and simian virus 40 (SV40).
- the method of Mulligan et al. (Nature (1979) 277, 108) can be used. Further, the HEF1 ⁇ promoter / enhancer can be easily used for target gene expression by the method of Mizushima et al. (Nucleic Acids Res. (1990) 18, 5322).
- the gene can be expressed by functionally combining a useful promoter commonly used, a signal sequence for antibody secretion, and an antibody gene to be expressed.
- the promoter include lacZ promoter and araB promoter.
- the lacZ promoter the method of Ward et al. (Nature (1989) 341, 544-546; FASEBJ. (1992) 6, 2422-2427) can be used.
- the araB promoter can be used for the expression of the target gene by the method of Better et al. (Science (1988) 240, 1041-1043).
- a pelB signal sequence (Lei, S. P. et al., J. Bacteriol. (1987) 169, 4379) may be used when the periplasm of E. coli is produced. Then, after separating the antibody produced in the periplasm, the structure of the antibody is refolded so as to have a desired binding activity by using a protein denaturant such as urea or guanidine hydrochloride.
- the signal sequence of the antibody heavy chain gene or light chain gene is desirable to use as the signal sequence required for secretion outside the cell. It is also possible to use signal sequences possessed by secreted proteins such as IL-3 and IL-6.
- a selectable marker can be inserted into the expression vector for amplification of the gene copy number in the host cell system. Specifically, the following selection markers can be used. ⁇ Aminoglycoside transferase (APH) gene ⁇ Thymidine kinase (TK) gene ⁇ E. coli xanthine guanine phosphoribosyltransferase (Ecogpt) gene ⁇ Dihydrofolate reductase (dhfr) gene, etc.
- APH Aminoglycoside transferase
- TK Thymidine kinase
- Ecogpt E. coli xanthine guanine phosphoribosyltransferase
- dhfr Dihydrofolate reductase
- Host cells are cultured according to a known method.
- DMEM, MEM, RPMI1640, and IMDM can be used as the culture medium, and serum supplements such as fetal calf serum (FCS) can be used in combination.
- FCS fetal calf serum
- the antibody expressed and produced as described above can be purified by using a known method used in normal protein purification alone or in combination as appropriate.
- antibodies can be separated and purified by appropriately selecting and combining affinity columns such as protein A columns, chromatography columns, filters, ultrafiltration, salting out, dialysis, etc. (Antibodies A Laboratory Manual. Ed Harlow , David David Lane, Cold Spring Spring Laboratory, 1988).
- transgenic animals can also be used for the production of recombinant antibodies. That is, the antibody can be obtained from an animal into which a gene encoding the target antibody has been introduced.
- an antibody gene can be constructed as a fusion gene by inserting in frame into a gene that encodes a protein that is uniquely produced in milk.
- a protein secreted into milk for example, goat ⁇ -casein can be used.
- the DNA fragment containing the fusion gene into which the antibody gene has been inserted is injected into a goat embryo, and the injected embryo is introduced into a female goat.
- the desired antibody can be obtained as a fusion protein with milk protein from milk produced by a transgenic goat (or its offspring) born from a goat that has received the embryo.
- hormones can be used as appropriate in transgenic goats to increase the amount of milk containing the desired antibody produced from the transgenic goat (Ebert, KM et al., Bio / Technology (1994) 12, 699-702). ).
- a C region derived from a human antibody or a C region derived from a non-human animal antibody can be used.
- C ⁇ 1, C ⁇ 2a, C ⁇ 2b, C ⁇ 3, C ⁇ , C ⁇ , C ⁇ 1, C ⁇ 2, C ⁇ can be used as the H chain C region of the mouse antibody
- C ⁇ , C ⁇ can be used as the L chain C region.
- antibodies such as rats, rabbits, goats, sheep, camels and monkeys can be used as antibodies from animals other than mice. These sequences are known.
- the C region can be modified to improve the stability of the antibody or its production.
- an antibody when administered to a human, it can be a genetically modified antibody that has been artificially modified for the purpose of reducing the heterologous antigenicity of the human.
- the recombinant antibody includes, for example, a chimeric antibody and a humanized antibody. These modified antibodies can be produced using known methods.
- Chimeric antibody refers to an antibody in which variable regions and constant regions derived from each other are linked.
- an antibody consisting of the variable regions of the heavy and light chains of a mouse antibody and the constant regions of the heavy and light chains of a human antibody is a mouse-human-heterologous chimeric antibody.
- a recombinant vector that expresses a chimeric antibody can be prepared by linking DNA encoding the variable region of a mouse antibody to DNA encoding the constant region of a human antibody and incorporating it into an expression vector.
- the chimeric antibody produced in the culture can be obtained by culturing recombinant cells transformed with the vector and expressing the incorporated DNA.
- a human antibody is used for the C region of the chimeric antibody and the humanized antibody.
- C ⁇ 1, C ⁇ 2, C ⁇ 3, C ⁇ 4, C ⁇ , C ⁇ , C ⁇ 1, C ⁇ 2, and C ⁇ can be used as the C region.
- C ⁇ and C ⁇ can be used as the C region.
- the amino acid sequences of these C regions, as well as the base sequences encoding them, are known.
- the human antibody C region can be modified to improve the stability of the antibody itself or the production of the antibody.
- a chimeric antibody is composed of a V region of an antibody derived from a non-human animal and a C region derived from a human antibody.
- humanized antibodies include complementarity determining regions (CDRs) of non-human animal-derived antibodies, framework regions derived from human antibodies (FR; framework regions), and C regions derived from human antibodies. Consists of Since humanized antibodies have reduced antigenicity in the human body, they are useful as the active ingredient of the therapeutic agent of the present invention.
- Antibody variable regions are usually composed of three CDRs sandwiched between four FRs.
- CDRs are regions that substantially determine the binding specificity of an antibody.
- the amino acid sequence of CDR is rich in diversity.
- the amino acid sequence constituting FR often shows high homology among antibodies having different binding specificities. Therefore, it is generally said that the binding specificity of one antibody can be transplanted to another antibody by CDR grafting.
- Humanized antibodies are also referred to as reshaped human antibodies. Specifically, non-human animals, for example, humanized antibodies obtained by grafting mouse antibody CDRs to human antibodies are known. General genetic recombination techniques for obtaining humanized antibodies are also known.
- overlap extension PCR is known as a method for transplanting mouse antibody CDRs to human FRs.
- PCR extension the base sequence which codes CDR of the mouse antibody which should be transplanted is added to the primer for synthesize
- selection of human FRs having high homology with mouse FRs is advantageous in maintaining CDR function. That is, generally, it is preferable to use a human FR comprising an amino acid sequence having high homology with the amino acid sequence of the FR adjacent to the mouse CDR to be transplanted.
- the base sequences to be linked are designed to be connected to each other in frame.
- Human FRs are synthesized individually by each primer.
- a product in which DNA encoding mouse CDR is added to each FR is obtained.
- the base sequences encoding mouse CDRs of each product are designed to overlap each other.
- the overlapping CDR portions of the products synthesized using the human antibody gene as a template are annealed with each other to perform a complementary chain synthesis reaction. By this reaction, human FRs are linked via the mouse CDR sequence.
- a human-type antibody expression vector can be prepared by inserting the DNA obtained as described above and a DNA encoding the human antibody C region into an expression vector so as to be fused in frame. After introducing this vector into a host to establish recombinant cells, the recombinant cells are cultured, and the DNA encoding the humanized antibody is expressed, whereby the humanized antibody is produced in the culture of the cultured cells. (See European Patent Publication EP 239400, International Publication WO 96/02576).
- the CDR forms a favorable antigen-binding site when linked via CDR.
- a human antibody FR can be suitably selected.
- FR amino acid residues can be substituted so that the CDR of the reshaped human antibody forms an appropriate antigen-binding site.
- amino acid sequence mutations can be introduced into FRs by applying the PCR method used for transplantation of mouse CDRs into human FRs.
- partial nucleotide sequence mutations can be introduced into primers that anneal to the FR.
- a nucleotide sequence mutation is introduced into the FR synthesized by such a primer.
- a mutant FR sequence having a desired property can be selected by measuring and evaluating the antigen-binding activity of a mutant antibody substituted with an amino acid by the above method (Sato, K.et al., Cancer Res, 1993, 53 , 851-856).
- the antibody of the present invention includes not only a bivalent antibody typified by IgG but also a monovalent antibody or a multivalent antibody typified by IgM as long as it binds to the HS6ST2 protein.
- the multivalent antibodies of the present invention include multivalent antibodies that all have the same antigen-binding site, or multivalent antibodies that have some or all different antigen-binding sites.
- the antibody of the present invention is not limited to the full-length antibody molecule, and may be a low molecular weight antibody or a modified product thereof as long as it binds to the HS6ST2 protein.
- the low molecular weight antibody includes an antibody fragment in which a part of a full-length antibody (whole antibody, such as whole IgG) is deleted. As long as it has the ability to bind to the HS6ST2 antigen, partial deletion of the antibody molecule is tolerated.
- the antibody fragment in the present invention preferably contains either or both of a heavy chain variable region (VH) and a light chain variable region (VL).
- the antibody fragment in the present invention preferably contains CDR.
- the number of CDRs contained in the antibody fragment of the present invention is not particularly limited, but preferably contains at least six of heavy chain CDR1, CDR2, CDR3, light chain CDR1, CDR2, and CDR3.
- the amino acid sequence of VH or VL can contain substitutions, deletions, additions and / or insertions. Furthermore, as long as it has the ability to bind to the HS6ST2 antigen, either or both of VH and VL can be deleted.
- the variable region may be chimerized or humanized.
- Specific examples of antibody fragments include, for example, Fab, Fab ′, F (ab ′) 2, and Fv.
- Specific examples of the low molecular weight antibody include, for example, Fab, Fab ′, F (ab ′) 2, Fv, scFv (single chain Fv), diabody, sc (Fv) 2 (single chain (Fv) 2 ), ScFv-Fc, and the like. Multimers of these antibodies (eg, dimer, trimer, tetramer, polymer) are also included in the low molecular weight antibody of the present invention.
- Antibody fragments can be obtained by treating antibodies with enzymes to generate antibody fragments.
- enzymes that produce antibody fragments include, for example, papain, pepsin, and plasmin.
- genes encoding these antibody fragments can be constructed, introduced into an expression vector, and then expressed in an appropriate host cell (for example, Co, MS et al., J. Immunol. (1994) 152). , 2968-2976, Better, M. & Horwitz, A. H. Methods in Enzymology (1989) 178, 476-496, Plueckthun, A. & Skerra, A.
- the digestive enzyme cleaves a specific position of the antibody fragment to give an antibody fragment having a specific structure as follows, for example. If a genetic engineering technique is used for such an enzymatically obtained antibody fragment, any part of the antibody can be deleted. Papain digestion: F (ab) 2 or Fab Pepsin digestion: F (ab ') 2 or Fab'
- the low molecular weight antibody in the present invention can be an antibody fragment lacking any region as long as it has binding affinity for HS6ST2. Further, particularly in the treatment of cell proliferative diseases such as cancer according to the present invention, it is desirable that the antibody maintain its effector activity. That is, the preferred low molecular weight antibody in the present invention has both a binding affinity for HS6ST2 and an effector function.
- Antibody effector functions include ADCC activity and CDC activity.
- the therapeutic antibody in the present invention particularly preferably comprises one or both of ADCC activity and CDC activity as an effector function.
- Diabody refers to a bivalent antibody fragment constructed by gene fusion (Holliger Pet et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993), EP 404,097, WO93 / 11161 etc.).
- Diabodies are dimers composed of two polypeptide chains. Usually, in the polypeptide chain constituting the dimer, VL and VH are connected by a linker in the same chain. The linker in the diabody is generally so short that VL and VH cannot bind to each other. Specifically, the amino acid residues constituting the linker are, for example, about 5 residues. Therefore, VL and VH encoded on the same polypeptide chain cannot form a single chain variable region fragment but form a dimer with another single chain variable region fragment. As a result, the diabody has two antigen binding sites.
- ScFv can be obtained by linking antibody H chain V region and L chain V region.
- the H chain V region and the L chain V region are linked via a linker, preferably a peptide linker (Huston, J. S. et al., Proc. Natl. Acad. Sci. USA, 1988, 85 , 5879-5883).
- the H chain V region and L chain V region in scFv may be derived from any of the antibodies described herein.
- region For example, any single chain peptide consisting of about 3 to 25 residues can be used as a linker. Specifically, for example, a peptide linker described later can be used.
- the V regions of both chains can be linked by, for example, the PCR method as described above.
- the DNA encoding the desired partial amino acid sequence is used as a template.
- the DNAs encoding the V region of the H chain and the L chain are each amplified by PCR using a pair of primers having sequences corresponding to the sequences at both ends of the DNA to be amplified.
- DNA encoding a peptide linker portion is prepared.
- DNA encoding a peptide linker can also be synthesized using PCR.
- a base sequence that can be linked to the amplification product of each V region synthesized separately is added to the 5 ′ side of the primer to be used.
- PCR reaction is performed using each DNA of [H chain V region DNA]-[peptide linker DNA]-[L chain V region DNA] and assembly PCR primers.
- the primer for assembly PCR consists of a combination of a primer that anneals to the 5 'side of [H chain V region DNA] and a primer that anneals to the 3' side of [L chain V region DNA]. That is, the assembly PCR primer is a primer set that can amplify DNA encoding the full-length sequence of scFv to be synthesized. On the other hand, a base sequence that can be linked to each V region DNA is added to [peptide linker DNA]. As a result, these DNAs are ligated, and the full length of scFv is finally produced as an amplification product by the primers for assembly PCR.
- an expression vector containing them and a recombinant cell transformed with the expression vector can be obtained according to a conventional method. Further, the scFv can be obtained by culturing the resulting recombinant cells and expressing the DNA encoding the scFv.
- ScFv-Fc is a low molecular weight antibody in which an Fc region is fused to an scFv consisting of an H chain V region and an L chain V region of an antibody (Cellular® & “Molecular® Immunology” 2006; 3: 439-443).
- scFv derived from IgM
- the origin of Fc is not particularly limited, and for example, human IgG (human IgG1 etc.) can be used.
- scFv-Fc scFv- fragment obtained by linking the scFv fragment of IgM antibody and human IgG1 CH2 (eg, C ⁇ 2) and CH3 (eg, C ⁇ 3) at the hinge region (H ⁇ ) of human IgG1.
- Fc can be mentioned.
- sc (Fv) 2 is a low molecular weight antibody in which two VHs and two VLs are combined with a linker or the like to form a single chain (Hudsonudet al., J Immunol. Methods 1999; 231: 177-189) .
- sc (Fv) 2 can be prepared, for example, by linking scFv with a linker.
- VHs and two VLs are arranged in the order of VH, VL, VH, and VL ([VH] linker [VL] linker [VH] linker [VL]) starting from the N-terminal side of the single-chain polypeptide.
- An antibody characterized in that it is preferred.
- a peptide linker is preferred.
- the length of the peptide linker is not particularly limited, and can be appropriately selected by those skilled in the art according to the purpose.
- the amino acid residues constituting the peptide linker are 1 to 100 amino acids, preferably 3 to 50 amino acids, more preferably 5 to 30 amino acids, particularly preferably 12 to 18 amino acids (for example, 15 amino acids).
- the amino acid sequence constituting the peptide linker can be any sequence as long as it does not inhibit the scFv binding action.
- the following amino acid sequence can be used.
- Ser Gly ⁇ Ser Gly ⁇ Gly ⁇ Ser Ser ⁇ Gly ⁇ Gly Gly, Gly, Ser (SEQ ID NO: 109) Ser, Gly, Gly, Gly (SEQ ID NO: 110) Gly, Gly, Gly, Gly, Ser (SEQ ID NO: 111) Ser, Gly, Gly, Gly, Gly (SEQ ID NO: 112) Gly, Gly, Gly, Gly, Gly, Ser (SEQ ID NO: 113) Ser, Gly, Gly, Gly, Gly, Gly (SEQ ID NO: 114) Gly, Gly, Gly, Gly, Gly, Gly, Ser (SEQ ID NO: 115) Ser, Gly, Gly, Gly, Gly, Gly, Gly, Gly (SEQ ID NO: 115) Ser, G
- n which determines the length of the above peptide linker is usually 1 to 5, preferably 1 to 3, more preferably 1 or 2.
- sc (Fv) 2 in the present invention include the following sc (Fv) 2.
- V regions can be linked using a synthetic chemical linker (chemical cross-linking agent).
- a crosslinking agent usually used for crosslinking such as peptide compounds can be used in the present invention.
- the following chemical crosslinking agents are known. These crosslinking agents are commercially available.
- N-hydroxysuccinimide (NHS), Disuccinimidyl suberate (DSS), Bis (sulfosuccinimidyl) suberate (BS3), Dithiobis (succinimidyl propionate) (DSP), Dithiobis (sulfosuccinimidyl propionate) (DTSSP), Ethylene glycol bis (succinimidyl succinate) (EGS), Ethylene glycol bis (sulfosuccinimidyl succinate) (sulfo-EGS), Disuccinimidyl tartrate (DST), disulfosuccinimidyl tartrate (sulfo-DST), When binding four antibody variable regions such as bis [2- (succinimideoxycarbonyloxy) ethyl] sulfone (BSOCOES) and bis [2- (sulfosuccinimideoxycarbonyloxy) ethyl] sulfone (sul
- the preferred low molecular weight antibody in the present invention is diabody or sc (Fv) 2.
- the antibody is treated with an enzyme such as papain or pepsin to generate antibody fragments, or DNA encoding these antibody fragments is constructed and used as an expression vector. After the introduction, it may be expressed in an appropriate host cell (for example, Co, MS et al., J. Immunol. (1994) 152, 2968-2976; Better, M. and Horwitz, AH, Methods Enzymol. ( (1989) 178, 476-496; Pluckthun, A. and Skerra, A., Methods Enzymol.
- the antibody of the present invention includes not only a monovalent antibody but also a multivalent antibody.
- the multivalent antibodies of the present invention include multivalent antibodies that all have the same antigen-binding site, or multivalent antibodies that have some or all different antigen-binding sites.
- An antibody conjugated with various molecules such as polyethylene glycol (PEG) can also be used as a modified antibody. It is also possible to bind a cytotoxic substance such as a chemotherapeutic agent, a toxic peptide or a radioactive chemical substance to the antibody.
- a modified antibody hereinafter referred to as antibody conjugate
- antibody conjugate can be obtained by chemically modifying the obtained antibody.
- the modification method of an antibody has already been established in this field.
- the cytotoxic substance that binds to the anti-HS6ST2 antibody of the present invention to function the cytotoxic activity is specifically a chemotherapeutic agent such as the following chemotherapeutic agents: azaribine, anastro Anastrozole, azacytidine, bleomycin, bortezomib, bryostatin-1, busulfan, camptothecin, 10-hydroxycamptothecin, Carmustine, celebrex, chlorambucil, cisplatin, irinotecan, carboplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine (Dacarbazine), Docetaki Docetaxel, dactinomycin, daunomycin glucuronide, daunorubicin, dexamethasone, diethylstilbestrol, ethylstilbestrol, doxorubicin rubo doxorubicin Epirubicin, eth
- a preferable chemotherapeutic agent is a low-molecular chemotherapeutic agent.
- Small molecule chemotherapeutic agents are less likely to interfere with antibody function after binding to the antibody.
- the low-molecular chemotherapeutic agent usually has a molecular weight of 100 to 2000, preferably 200 to 1000.
- the chemotherapeutic agents exemplified here are all low-molecular chemotherapeutic agents.
- These chemotherapeutic agents in the present invention include a prodrug that is converted into an active chemotherapeutic agent in vivo. Activation of the prodrug may be enzymatic conversion or non-enzymatic conversion.
- the cytotoxic substance of the present invention may be a toxic peptide.
- the antibody is modified with the toxic peptide.
- a toxic peptide the following can be mentioned, for example. Diphtheria toxin A chain (Diphtheria toxin A Chain) (Langone JJ, et al., Methods in Enzymology, 93, 307-308, 1983), Pseudomonas Exotoxin (Nature Medicine, 2, 350-353,1996) , Ricin A Chain (Fulton RJ, ulet al., J.Biol.Chem., 261, 5314-5319, 1986; Sivam G., et al., Cancer Res., 47, 3169-3173, 1987 ; Cumber AJ et al., J.Immunol.Methods, 135, 15-24, 1990; Wawzynczak EJ, et al., Cancer Res., 50, 7519-7562
- FEBS letter 195, 1-8, 1986 ccModeccin (Stirpe F., Barbieri L., FEBS letter 195, 1-8, 1986); Viscumin (Stirpe F., Barbieri L., FEBS letter 195, 1-8, 1986); Volkesin (Stirpe F., Barbieri L., FEBS letter 195, 1-8, 1986); Dodecandrin (Stirpe F., Barbieri L., FEBS letter 195, 1-8, 1986); Tritin (Stirpe F., Barbieri L., FEBS letter 195, 1-8, 1986); Luffin (Stirpe F., Barbieri L., FEBS letter 195, 1-8, 1986); Trichokirin (Casellas P., et al., Eur.J. Biochem. 176, 581-588, 1988; B perfumesi A., et al., Clin.exp.Immunol., 89
- the cytotoxic substance in the present invention may be a radioactive chemical substance, and the radioactive chemical substance refers to a chemical substance containing a radioisotope.
- the radioactive isotope is not particularly limited, and any radioactive isotope may be used. For example, 32 P, 14 C, 125 I, 3 H, 131 I, 186 Re, 188 Re, etc. can be used. .
- one or two or more small molecule chemotherapeutic agents and toxic peptides can be used in combination to modify the antibody.
- Coupling between the anti-HS6ST2 antibody and the above small molecule chemotherapeutic agent can be covalent or noncovalent. Methods for producing antibodies bound with these chemotherapeutic agents are known.
- proteinaceous drugs and toxins can be bound to antibodies by genetic engineering techniques.
- a recombinant vector in which a DNA encoding the toxic peptide and a DNA encoding an anti-HS6ST2 antibody are fused in frame and incorporated into an expression vector can be constructed.
- a transformed cell obtained by introducing the vector into an appropriate host cell is cultured, and the incorporated DNA is expressed, whereby an anti-HS6ST2 antibody to which a toxic peptide is bound can be obtained as a fusion protein.
- a proteinaceous drug or toxin is generally arranged on the C-terminal side of the antibody.
- a peptide linker can be interposed between the antibody and the proteinaceous drug or toxin.
- the antibody of the present invention may be a bispecific antibody.
- Bispecific antibodies refer to antibodies that have variable regions that recognize different epitopes within the same antibody molecule.
- bispecific antibodies can have antigen binding sites that recognize different epitopes on the HS6ST2 molecule.
- Such a bispecific antibody can bind two antibody molecules to one molecule of HS6ST2. As a result, a stronger cytotoxic effect can be expected.
- a bispecific antibody in which one antigen-binding site recognizes HS6ST2 and the other antigen-binding site recognizes a cytotoxic substance can also be used.
- the cytotoxic substance includes a chemotherapeutic agent, a toxic peptide, a radioactive chemical substance, and the like.
- Such bispecific antibodies capture cytotoxic substances while binding to cells expressing HS6ST2.
- the cytotoxic substance can directly act on HS6ST2-expressing cells. That is, the bispecific antibody that recognizes the cytotoxic substance can specifically damage the tumor cell and suppress the growth of the tumor cell.
- bispecific antibodies that recognize antigens other than HS6ST2 can also be combined.
- a bispecific antibody that recognizes an antigen that is specifically expressed on the cell surface of the target cancer cell and is different from HS6ST2 can be combined.
- bispecific antibodies can be produced by combining two types of antibodies with different recognition antigens.
- the antibody to be bound may be a 1 ⁇ 2 molecule each having an H chain and an L chain, or may be a 1 ⁇ 4 molecule consisting only of an H chain.
- bispecific antibody-producing fused cells can be prepared by fusing hybridomas that produce different monoclonal antibodies.
- bispecific antibodies can be produced by genetic engineering techniques.
- ELISA enzyme-linked immunosorbent assay
- EIA enzyme immunoassay
- RIA radioimmunoassay
- fluorescent immunoassay can be used.
- the antibody of the present invention may be an antibody having a modified sugar chain. It is known that the cytotoxic activity of an antibody can be enhanced by modifying the sugar chain of the antibody.
- modified sugar chains for example, the following antibodies are known. Antibodies with modified glycosylation (eg WO99 / 54342), Antibodies lacking fucose added to the sugar chain (WO00 / 61739, WO02 / 31140 etc.), Antibodies with sugar chains with bisecting GlcNAc (WO02 / 79255 etc.) etc.
- the antibody of the present invention is used for therapeutic purposes, the antibody is preferably an antibody having cytotoxic activity.
- cytotoxic activity in the present invention examples include antibody-dependent cell-mediated cytotoxicity (ADCC) activity and complement-dependent cytotoxicity (CDC) activity.
- ADCC activity means cytotoxic activity by the complement system.
- Fc ⁇ receptor-bearing cells immunodeficiency virus, etc.
- Fc ⁇ receptor-bearing cells bind to the Fc portion via the Fc ⁇ receptor, causing damage to the target cell. Means activity.
- Whether or not the anti-HS6ST2 antibody has ADCC activity or CDC activity can be measured by a known method (for example, Current protocols in Immunology, Chapter7. Immunologic studies in humans, Editor, John E, Coligan et al., John Wiley & Sons, Inc., (1993)).
- effector cells are prepared.
- complement solutions are prepared.
- target cells are prepared.
- effector cells Spleens are removed from CBA / N mice and the like, and spleen cells are isolated in RPMI1640 medium (Invitrogen). After washing with the same medium containing 10% fetal bovine serum (FBS, HyClone), effector cells can be prepared by adjusting the cell concentration to 5 ⁇ 10 6 / ml.
- FBS fetal bovine serum
- Target cell preparation HS6ST2 protein-expressing cells are cultured with 0.2 mCi of 51 Cr-sodium chromate (GE Healthcare Biosciences) in DMEM medium containing 10% FBS for 1 hour at 37 ° C.
- the target cell can be radiolabeled.
- cells expressing HS6ST2 protein cells transformed with a gene encoding HS6ST2 protein, lung adenocarcinoma cells, lung cancer cells, liver cancer cells, ovarian cancer cells, lung squamous cell carcinoma cells, etc. can be used. .
- the target cells can be prepared by washing the cells three times with 10% FBS-containing RPMI1640 medium and adjusting the cell concentration to 2 ⁇ 10 5 / ml.
- ADCC activity or CDC activity can be measured by the method described below.
- 50 ⁇ l each of target cells and anti-HS6ST2 antibody are added to a 96-well U-bottom plate (BectonectDickinson) and allowed to react on ice for 15 minutes. Thereafter, 100 ⁇ l of effector cells are added and cultured for 4 hours in a carbon dioxide incubator. The final antibody concentration is 0 or 10 ⁇ g / ml. After incubation, 100 ⁇ l of the supernatant is collected, and the radioactivity is measured with a gamma counter (COBRAIIAAUTO-GAMMA, MODEL D5005, Packard Instruments Company).
- COBRAIIAAUTO-GAMMA MODEL D5005, Packard Instruments Company
- Cytotoxic activity (%) can be calculated based on the formula (A-C) / (B-C) x 100 using the obtained value.
- A is the radioactivity (cpm) in each sample
- B is the radioactivity (cpm) in the sample with 1% NP-40 (manufactured by nacalai tesque)
- C is the radioactivity (cpm) of the sample containing only the target cells Show.
- cytotoxic activity in the case of measuring the cytotoxic activity by the antibody conjugate, 50 ⁇ l each of the target cells and the anti-HS6ST2 antibody conjugate are added to a 96-well flat bottom plate (Becton-Dickinson) and allowed to react on ice for 15 minutes. Incubate for 1 to 4 hours in a carbon dioxide incubator. The final antibody concentration is 0 or 3 ⁇ g / ml. After incubation, 100 ⁇ l of the supernatant is collected, and the radioactivity is measured with a gamma counter. Cytotoxic activity can be calculated in the same manner as ADCC activity measurement.
- Another example of the antibody used in the present invention is an antibody having an internalizing activity.
- an antibody having an internalizing activity means an antibody that is transported into cells (cytoplasm, vesicle, other organelle, etc.) when bound to HS6ST2.
- an anti-HS6ST2 antibody bound with a labeling substance is contacted with a cell expressing HS6ST2, and the labeling substance
- a method for confirming whether or not it has been taken into the cell a method for confirming whether cell death has been induced in the HS6ST2-expressing cells by contacting an anti-HS6ST2 antibody conjugated with a cytotoxic substance with cells expressing HS6ST2, etc. Can be confirmed. More specifically, it is possible to confirm whether or not an antibody has an internalizing activity by the method described in the Examples below.
- An antibody having an internalizing activity can be used as a pharmaceutical composition such as an anticancer agent by binding the above-mentioned cytotoxic substance.
- a heavy chain comprising a heavy chain CDR1 having the amino acid sequence set forth in SEQ ID NO: 64, a heavy chain CDR2 having the amino acid sequence set forth in SEQ ID NO: 65, and a heavy chain CDR3 having the amino acid sequence set forth in SEQ ID NO: 66
- An antibody comprising a chain variable region (A1);
- An antibody comprising a chain variable region (A6);
- having the same activity as the antibody of the present invention means that the binding activity to HS6ST2 and / or the cytotoxic activity to cells expressing HS6ST2 are equivalent.
- the method of introducing a mutation into a polypeptide is one of methods well known to those skilled in the art for preparing a polypeptide functionally equivalent to a certain polypeptide.
- a person skilled in the art can perform site-directed mutagenesis (Hashimoto-Gotoh, T. et al. (1995) Gene 152, 271-275, Zoller, MJ, and Smith, M. (1983) Methods Enzymol. 100 , 468-500, Kramer, W. et al. (1984) Nucleic Acids Res. 12, 9441-9456, Kramer W, and Fritz HJ (1987) Methods. Enzymol. 154, 350-367, Kunkel, TA (1985) Proc Natl Acad Sci USA.
- Antibodies can be prepared. Amino acid mutations can also occur in nature. Thus, an antibody having an amino acid sequence in which one or more amino acids are mutated in the amino acid sequence of the antibody of the present invention and functionally equivalent to the antibody is also included in the antibody of the present invention.
- the number of amino acids to be mutated is usually within 50 amino acids, preferably within 30 amino acids, and more preferably within 10 amino acids (for example, within 5 amino acids).
- the amino acid residue to be mutated is preferably mutated to another amino acid in which the properties of the amino acid side chain are conserved.
- the following classification has been established based on the properties of amino acid side chains.
- Hydrophobic amino acids (A, I, L, M, F, P, W, Y, V)
- Hydrophilic amino acids (R, D, N, C, E, Q, G, H, K, S, T)
- Amino acids with aliphatic side chains G, A, V, L, I, P
- An amino acid having a hydroxyl group-containing side chain S, T, Y
- Amino acids having side chains containing sulfur atoms C, M
- Amino acids with carboxylic acid and amide-containing side chains (D, N, E, Q)
- Amino acids with base-containing side chains (R, K, H)
- Amino acids with aromatic-containing side chains (H, F, Y, W) (In parentheses all represent one letter of amino acid)
- amino acid sequence classified into each group in an amino acid sequence constituting a certain polypeptide is highly likely to maintain the activity of the polypeptide when substituted with each other.
- substitution between amino acids in the group of amino acids is referred to as conservative substitution.
- the present invention also provides an antibody that binds to the same epitope as that to which any of the above-mentioned antibodies (1) to (21) binds.
- Specific examples of the antibodies (1) to (21) described above include the antibodies A1, A6, A10, B5, B6, C8, and C10 described in the Examples of the present application. That is, the present invention also provides an antibody that recognizes the same epitope as that recognized by A1, A6, A10, B5, B6, C8, and C10.
- Such an antibody can be obtained, for example, by the following method.
- test antibody shares an epitope with a certain antibody by competition for the same epitope.
- Competition between antibodies is detected by a cross-blocking assay or the like.
- a competitive ELISA assay is a preferred cross-blocking assay.
- the anti-HS6ST2 antibody of the present invention is added after preincubation of HS6ST2 protein coated on the well of a microtiter plate in the presence or absence of a candidate competitive antibody. Is done.
- the amount of the anti-HS6ST2 antibody of the present invention bound to the HS6ST2 protein in the well is indirectly correlated with the binding ability of a candidate competitive antibody (test antibody) that competes for binding to the same epitope. That is, the greater the affinity of the test antibody for the same epitope, the lower the amount of binding of the anti-HS6ST2 antibody of the present invention to the well coated with the HS6ST2 protein, while the test antibody bound to the well coated with the HS6ST2 protein. The amount of binding increases.
- the amount of antibody bound to the well can be easily measured by labeling the antibody in advance.
- biotin-labeled antibodies can be measured by using an avidin peroxidase conjugate and an appropriate substrate.
- a cross-blocking assay using an enzyme label such as peroxidase is particularly referred to as a competitive ELISA assay.
- the antibody can be labeled with another labeling substance that can be detected or measured. Specifically, radiolabels or fluorescent labels are known.
- any antibody bound to the well can be measured by a labeled antibody that recognizes any constant region.
- the antibodies bound to the wells can be measured by the antibodies that identify the respective classes.
- Anti-HS6ST2 antibody binding is at least 20%, preferably at least 30%, more preferably at least 50% compared to the binding activity obtained in a control test performed in the absence of the candidate competing antibody Can block substantially the same epitope as the anti-HS6ST2 antibody of the invention, or is an antibody that competes for binding to the same epitope.
- the constant region of the anti-HS6ST2 antibody of the present invention is replaced with the same constant region as the test antibody. Also good.
- the test antibody has a human-derived constant region (human IgG1, IgG2, IgG3, IgG4, etc.)
- the constant region of the anti-HS6ST2 antibody of the present invention is replaced with the same constant region as the test antibody. be able to.
- A1, A10, B5, B6, C8 recognizes a peptide consisting of the 459th amino acid from the 379th amino acid of the HS6ST2 protein having the amino acid sequence of SEQ ID NO: 107
- A1, A10, B5, B6, As a preferred example of an antibody that recognizes the same epitope as that recognized by C8, an antibody that recognizes the region from the 379th amino acid to the 459th amino acid of the HS6ST2 protein having the amino acid sequence of SEQ ID NO: 107 can be mentioned. .
- A6 recognizes a peptide consisting of amino acids 308 to 393 of the HS6ST2 protein having the amino acid sequence of SEQ ID NO: 107
- an antibody that recognizes a site from the 308th amino acid to the 393rd amino acid of the HS6ST2 protein having the amino acid sequence of SEQ ID NO: 107 is provided.
- C10 recognizes a peptide consisting of amino acids 24 to 175 of the HS6ST2 protein having the amino acid sequence of SEQ ID NO: 107. Therefore, as a preferred example of an antibody that recognizes the same epitope as that recognized by C10 And an antibody that recognizes a site from the 24th amino acid to the 175th amino acid of the HS6ST2 protein having the amino acid sequence of SEQ ID NO: 107.
- the present invention further provides an HS6ST2 protein to which a cytotoxic substance is bound. Since the HS6ST2 protein to which a cytotoxic substance is bound is considered to be internalized in the cell after binding to heparin sulfate, it can be used as an anticancer agent or the like.
- the cytotoxic substance to be bound to the HS6ST2 protein is not particularly limited, and for example, the above-described cytotoxic substances can be used.
- the HS6ST2 protein may be a fragment or mutant as long as it has the ability to bind to heparin sulfate.
- the ability of HS6ST2 protein to bind to heparin sulfate and the internalizing activity can be confirmed by methods known to those skilled in the art. For example, it can be confirmed by the method described above.
- the present invention provides a pharmaceutical composition containing an antibody that binds to HS6ST2 protein as an active ingredient.
- the present invention also relates to a cell growth inhibitor, particularly an anticancer agent, containing an antibody that binds to HS6ST2 protein as an active ingredient.
- the cytostatic agent and anticancer agent of the present invention are preferably administered to a subject suffering from or possibly suffering from cancer. Since the expression level of HS6ST2 is increased in cancer cells, it is considered that cytotoxicity specific to cancer cells can be obtained by administration of anti-HS6ST2 antibody.
- the anti-HS6ST2 antibody used in the pharmaceutical composition (for example, anticancer agent) of the present invention is not particularly limited, and any anti-HS6ST2 antibody may be used.
- the above-mentioned anti-HS6ST2 antibody can be used.
- containing an antibody that binds to HS6ST2 as an active ingredient means that the anti-HS6ST2 antibody is contained as a main active ingredient, and does not limit the content of the anti-HS6ST2 antibody.
- the present invention provides a pharmaceutical composition comprising HS6ST2 protein bound with a cytotoxic substance as an active ingredient.
- the present invention also provides a cell growth inhibitor, particularly an anticancer agent, containing HS6ST2 protein bound with a cytotoxic substance as an active ingredient.
- the cytostatic agent and anticancer agent of the present invention are preferably administered to a subject suffering from or possibly suffering from cancer.
- containing a cytotoxic substance-bound HS6ST2 protein as an active ingredient means that a cytotoxic substance-bound HS6ST2 protein is included as a main active ingredient. It does not limit the content of bound HS6ST2 protein.
- the target cancer is not particularly limited, but is preferably lung adenocarcinoma, lung cancer, liver cancer, ovarian cancer, or lung squamous cell carcinoma.
- the cancer may be either a primary lesion or a metastatic lesion.
- the pharmaceutical composition of the present invention can be administered to a patient by either oral or parenteral administration. Preferably, it is parenteral administration. Specific examples of such administration methods include injection administration, nasal administration, transpulmonary administration, and transdermal administration.
- injection administration the pharmaceutical composition of the present invention can be administered systemically or locally by, for example, intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection and the like.
- the administration method can be appropriately selected depending on the age and symptoms of the patient.
- the dose for example, the dose can be selected in the range of 0.0001 mg to 1000 mg per kg of body weight per administration. Alternatively, for example, the dose can be selected in the range of 0.001 to 100,000 mg / body per patient.
- the pharmaceutical composition of the present invention is not limited to these doses.
- the pharmaceutical composition of the present invention can be formulated in accordance with a conventional method (for example, Remington's Pharmaceutical, Science, Latest Edition, Mark Publishing, Company, Easton, USA) together with pharmaceutically acceptable carriers and additives. It may be.
- a conventional method for example, Remington's Pharmaceutical, Science, Latest Edition, Mark Publishing, Company, Easton, USA
- pharmaceutically acceptable carriers and additives may be.
- it is not limited to these, and other commonly used carriers can be used as appropriate.
- silicic acid lactose, crystalline cellulose, mannitol, starch, carmellose calcium, carmellose sodium, hydroxypropylcellulose, hydroxypropylmethylcellulose, polyvinylacetal diethylaminoacetate, polyvinylpyrrolidone, gelatin, medium chain fatty acid triglyceride
- the carrier include polyoxyethylene hydrogenated castor oil 60, sucrose, carboxymethylcellulose, corn starch, and inorganic salts.
- the present invention also relates to a method of causing injury to HS6ST2-expressing cells by contacting an HS6ST2-expressing cell and an antibody that binds to HS6ST2 protein or cell proliferation Provide a method for suppressing
- the antibody used in the method of the present invention is not particularly limited, and for example, the above-described antibodies can be used.
- the cell to which the anti-HS6ST2 antibody binds is not particularly limited as long as it is a cell expressing HS6ST2.
- Preferred HS6ST2-expressing cells in the present invention are cancer cells. More preferred are lung adenocarcinoma cells, lung cancer cells, liver cancer cells, ovarian cancer cells, and lung squamous cell carcinoma cells.
- the method of the present invention can be applied to both primary lesions and metastatic lesions of these cancers.
- contact is performed, for example, by adding an antibody to a culture solution of HS6ST2-expressing cells cultured in a test tube.
- contact is also performed by administering to a non-human animal transplanted with HS6ST2-expressing cells or an animal having cancer cells that endogenously express HS6ST2.
- the following method is preferably used as a method for evaluating or measuring the cytotoxicity caused to HS6ST2-expressing cells by contact with the anti-HS6ST2 antibody.
- Methods for evaluating or measuring the cytotoxic activity in vitro include the above-described antibody-dependent cell-mediated cytotoxicity (ADCC) activity, complement-dependent cytotoxicity (complement-dependent cytotoxicity). : CDC) activity and the like. Whether or not the anti-HS6ST2 antibody has ADCC activity or CDC activity can be measured by a known method (for example, Current protocols in Immunology, Chapter 7. Immunologic studies in humans, Editor, John E Coligan et al., John Wiley & Sons, Inc., (1993)).
- a binding antibody having the same isotype as the anti-HS6ST2 antibody and having no cytotoxic activity is used as a control antibody in the same manner as the anti-HS6ST2 antibody, and the anti-HS6ST2 antibody is stronger than the control antibody.
- Activity can be determined by showing cytotoxic activity.
- Antibody isotype is defined by the sequence of the H chain constant region of the amino acid sequence of the antibody. In vivo, the antibody isotype is finally determined by class switching caused by genetic recombination on the chromosome that occurs during maturation of antibody-producing B cells. Differences in isotypes are reflected in differences in the physiological and pathological functions of antibodies. Specifically, for example, it is known that the intensity of cytotoxic activity is influenced by the antibody isotype as well as the antigen expression level. Therefore, in the measurement of the cytotoxic activity described above, it is preferable to use the same isotype as the test antibody as the antibody used as a control.
- the test antibody is administered daily or at intervals of several days from that day or the next day. Administer intravenously or intraperitoneally. Cytotoxic activity can be determined by measuring tumor size over time. Similar to the in vitro evaluation, a control antibody having the same isotype was administered, and the tumor size in the anti-HS6ST2 antibody administration group was significantly smaller than the tumor size in the control antibody administration group. Then it can be determined.
- a nude (nu / nu) mouse in which the thymus is genetically deleted and the function of the T lymphocyte is deleted can be preferably used.
- a method for diagnosing cancer comprising detecting HS6ST2 protein or a gene encoding HS6ST2 protein.
- HS6ST2 has been confirmed to be significantly upregulated in various cancer tissues and cancer cell lines. Therefore, HS6ST2 is useful as a marker for specifically detecting cancer.
- the present invention provides a method for diagnosing cancer comprising the following steps: (a) providing a sample collected from the subject; (b) A step of detecting HS6ST2 protein or HS6ST2 gene contained in the sample of (a).
- cancer is diagnosed by detecting HS6ST2 protein in a sample.
- the detection of the HS6ST2 protein is preferably performed using an antibody that recognizes the HS6ST2 protein.
- One specific example of the diagnostic method of the present invention is a cancer diagnostic method including the following steps. (A) providing a sample collected from the subject; (B) A step of detecting HS6ST2 protein contained in the collected sample using an antibody that binds to HS6ST2 protein.
- detection includes quantitative or qualitative detection.
- qualitative detection can include the following measurements. ⁇ Measurement of whether or not HS6ST2 protein is present ⁇ Measurement of whether or not HS6ST2 protein is present in a certain amount or more ⁇ Measurement of comparing the amount of HS6ST2 protein with other samples (for example, control samples)
- quantitative detection includes measurement of the concentration of HS6ST2 protein, measurement of the amount of HS6ST2 protein, and the like.
- the test sample in the present invention is not particularly limited as long as it is a sample that may contain HS6ST2 protein.
- a sample collected from the body of a living organism such as a mammal is preferable. Further preferred samples are samples taken from humans.
- Specific examples of the test sample include blood, interstitial fluid, plasma, extravascular fluid, cerebrospinal fluid, synovial fluid, pleural fluid, serum, lymph fluid, saliva, urine, tissue, and the like.
- a preferred sample is a sample obtained from a test sample such as a specimen in which tissue or cells collected from the body of an organism are fixed, or a culture solution of cells.
- the cancer diagnosed by the present invention is not particularly limited and may be any cancer. Specific examples include lung adenocarcinoma, lung cancer, liver cancer, ovarian cancer, and lung squamous cell carcinoma. In the present invention, both primary lesions and metastatic lesions of these cancers can be diagnosed.
- the present invention when a protein is detected in a test sample, cancer is diagnosed using the level as an index. Specifically, when the amount of HS6ST2 protein detected in a test sample is large compared to a negative control or a healthy subject, the subject is more likely to have cancer or suffer from cancer in the future. It is shown. That is, the present invention relates to a method for diagnosing cancer including the following steps. (1) detecting an HS6ST2 expression level in a biological sample collected from a subject, and (2) A step in which the subject is shown to have cancer when the expression level of HS6ST2 detected in (1) is higher than that of the control.
- the control refers to a sample serving as a reference for comparison, and includes a negative control and a biological sample of a healthy person.
- the negative control can be obtained by collecting a biological sample of a healthy person and mixing as necessary.
- the control expression level of HS6ST2 can be detected in parallel with the expression level of HS6ST2 in the biological sample of the subject.
- the expression level of HS6ST2 in biological samples of a large number of healthy subjects can be detected in advance, and the standard expression level in healthy subjects can be statistically determined. Specifically, for example, an average value ⁇ 2 ⁇ standard deviation (S.D.) or an average value ⁇ 3 ⁇ standard deviation (S.D.) can be used as the standard value.
- the mean ⁇ 2 ⁇ standard deviation (S.D.) contains 80% and the mean ⁇ 3 ⁇ standard deviation (S.D.) contains 90% of healthy subjects.
- the expression level of HS6ST2 in the control can be set using the ROC curve.
- An ROC curve (receiver operating characteristic curve; receiver operating characteristic curve) is a graph in which the vertical axis indicates detection sensitivity and the horizontal axis indicates a false positive rate (ie, “1-specificity”).
- an ROC curve can be obtained by plotting changes in sensitivity and false positive rate when the reference value for determining the expression level of HS6ST2 in a biological sample is continuously changed.
- the “reference value” for obtaining the ROC curve is a numerical value temporarily used for statistical analysis.
- the “reference value” for obtaining the ROC curve is generally changed continuously within a range that can cover all selectable reference values. For example, the reference value can be varied between the minimum and maximum values of HS6ST2 measurements in the population to be analyzed.
- a standard value that can be expected to have a desired detection sensitivity and accuracy is also called a cut-off value.
- the expression level of HS6ST2 detected in (1) is compared with the cut-off value.
- the expression level of HS6ST2 detected in (1) is higher than the cut-off value, the subject's cancer is detected.
- the expression level of HS6ST2 can be determined by any method. Specifically, the expression level of HS6ST2 can be known by evaluating the amount of HS6ST2 mRNA, the amount of HS6ST2 protein, and the biological activity of HS6ST2 protein. The amount of mRNA or protein of HS6ST2 can be determined by a method as described herein.
- a particularly suitable subject is a human.
- HS6ST2 protein of the animal species is detected.
- the method for detecting the HS6ST2 protein contained in the test sample is not particularly limited, but it is preferably detected by an immunological method as exemplified below using an anti-HS6ST2 antibody.
- the immunohistochemistry (IHC) method includes a step of detecting HS6ST2 protein on a section on which tissue or cells obtained from a patient suffering from cancer is immobilized, and is preferable as a method for diagnosing cancer.
- One of the immunological assay methods One of the immunological assay methods.
- the immunological methods described above, such as the immunohistochemistry (IHC) method, are methods known to those skilled in the art.
- HS6ST2 is a membrane protein whose expression is specifically enhanced in cancer cells
- cancer cells or cancer tissues can be detected by anti-HS6ST2 antibody.
- immunohistological analysis cancer cells contained in cells or tissues collected from a living body are detected.
- cancer tissue in a living body can be detected with an anti-HS6ST2 antibody.
- the present invention includes (1) a step of administering an antibody that binds to an HS6ST2 protein labeled with a labeling substance such as a radioisotope to a subject, and (2) a step of detecting accumulation of the labeling substance. It relates to the detection method.
- the antibodies can be detectably labeled. For example, the behavior of a fluorescent substance, a luminescent substance, or an antibody labeled with a radioisotope in a living body can be traced.
- An antibody labeled with a fluorescent substance or a luminescent substance can be observed using an endoscope or a laparoscope.
- a radioisotope can image the localization of an antibody by following its radioactivity.
- the localization of anti-HS6ST2 antibody in vivo indicates the presence of cancer cells.
- a positron emitting nuclide can be used as a radioisotope for labeling an antibody to detect cancer in vivo.
- antibodies can be labeled with positron emitting nuclides such as 18F, 55Co, 64Cu, 66Ga, 68Ga, 76Br, 89Zr, and 124I.
- positron emitting nuclides such as 18F, 55Co, 64Cu, 66Ga, 68Ga, 76Br, 89Zr, and 124I.
- a known method Acta Oncol. 32, 825-830, 1993
- the radiation emitted by the radionuclide is measured from outside the body by PET (Positron Tomography) and converted to an image by computer tomography. Is done.
- PET is a device for non-invasively obtaining data on the behavior of a drug in the body. With PET, radiation intensity can be quantitatively imaged as signal intensity.
- antigen molecules highly expressed in a specific cancer can be detected without collecting a sample from a patient.
- the anti-HS6ST2 antibody can also be radiolabeled with a short-lived nuclide using a positron emitting nuclide such as 11C, 13N, 15O, 18F, 45Ti in addition to the above-mentioned nuclide.
- a positron emitting nuclide such as 11C, 13N, 15O, 18F, 45Ti in addition to the above-mentioned nuclide.
- anti-HS6ST2 antibodies can be labeled with various radioisotopes.
- the anti-HS6ST2 antibody administered to the patient accumulates in the primary and metastatic lesions according to the specificity of the anti-HS6ST2 antibody for the pathological tissue at each site. If the anti-HS6ST2 antibody is labeled with a positron emitting nuclide, the presence of the primary and metastatic lesions is detected by the localization of the radioactivity by detecting the radioactivity.
- an activity value of 25-4000 keV gamma particles or positron emission can be used appropriately.
- an appropriate nuclide is selected and administered in a larger amount, a therapeutic effect can be expected.
- a gamma particle of 70-700 keV or a nuclide that gives a positron emission value can be used.
- the expression of HS6ST2 gene is detected.
- the gene detected in the present invention is not particularly limited, but mRNA is preferable.
- detection includes quantitative or qualitative detection. For example, the following measurement operation can be given as qualitative detection. -Simply measuring whether HS6ST2 mRNA is present, Measurement of whether or not HS6ST2 mRNA is present above a certain amount, ⁇ Measurements comparing the amount of HS6ST2 mRNA with other samples (eg, control samples)
- quantitative detection includes measurement of the concentration of HS6ST2 mRNA, measurement of the amount of HS6ST2 mRNA, and the like.
- test sample in the present invention any sample that may contain HS6ST2 mRNA can be used.
- a sample collected from the body of a living organism such as a mammal is preferable, and a sample collected from a human is more preferable.
- Specific examples of the test sample include blood, interstitial fluid, plasma, extravascular fluid, cerebrospinal fluid, synovial fluid, pleural fluid, serum, lymph fluid, saliva, urine, tissue, and the like.
- a preferable sample includes a sample obtained from a test sample, such as a specimen in which tissues or cells collected from the body of an organism are fixed, or a culture solution of cells, and the like.
- an in situ hybridization method is preferably used.
- the in situ hybridization method has been developed as a method for confirming the presence or distribution of specific DNA or RNA in cells or tissues and the intensity of its expression. In principle, this utilizes the property that a probe nucleic acid having a base sequence complementary to a specific nucleic acid sequence in a cell specifically forms a complex.
- RI radioisotope
- hapten antigenic substance
- an RI label can be preferably used.
- a fluorescent label using a non-radioactive material such as biotin or hapten such as digoxigenin can be used.
- a detection method by fluorescence in situ hybridization called FISH is used.
- the cancer to be diagnosed is not particularly limited. Specific examples include lung adenocarcinoma, lung cancer, liver cancer, ovarian cancer, and lung squamous cell carcinoma. In the present invention, both primary lesions and metastatic lesions of these cancers can be diagnosed.
- any animal species that expresses the HS6ST2 gene can be the subject.
- a particularly suitable subject is a human.
- the HS6ST2 gene of the animal species is detected.
- a sample is prepared from a subject.
- HS6ST2 mRNA contained in the sample is detected.
- cDNA synthesized from mRNA can also be detected.
- mRNA of HS6ST2 or cDNA encoding HS6ST2 is detected in a test sample, it is determined that there is a possibility of cancer. For example, when the amount of HS6ST2 mRNA or cDNA encoding HS6ST2 detected in a test sample is higher than in a negative control or a healthy subject, the subject has cancer or will suffer from future cancer. It is shown that there is a high probability of doing.
- Methods for detecting mRNA are known. Specifically, for example, Northern blotting method, RT-PCR method, DNA array method and the like can be used in the present invention.
- the detection method of the present invention described above can be automated using various automatic inspection apparatuses. By automation, a large number of samples can be inspected in a short time.
- the present invention also provides a diagnostic agent or kit for diagnosing cancer, comprising a reagent for detecting HS6ST2 protein in a test sample.
- the diagnostic agent of the present invention contains at least an anti-HS6ST2 antibody.
- kits for diagnosing cancer can be obtained by combining the diagnostic reagent for cancer of the present invention with other elements used for detection of HS6ST2. That is, the present invention includes a kit for diagnosing cancer, which comprises an antibody that binds to HS6ST2, a reagent for detecting the binding between the antibody and HS6ST2, and may further comprise a control sample comprising a biological sample containing HS6ST2. About.
- the kit of the present invention may further include instructions for explaining the measurement operation.
- Example 1 Expression analysis of HS6ST2 mRNA by Human Exon 1.0 ST Array
- Human Exon 1.0 ST Array Expression of HS6ST2 mRNA in clinical cancer, cancer cell lines and various normal tissues was analyzed using Human Exon 1.0 ST Array (Affymetrix).
- Human Exon 1.0 ST Array since at least one probe set is set for each exon of each gene, expression data of a plurality of probe sets can be obtained for one gene. Therefore, it is considered that the reliability of the expression data is improved as compared with the conventional expression array of Affymetrix, which basically has only one probe set per gene.
- Samples include 41 tumors of lung adenocarcinoma tissue, 13 tumors of small cell lung cancer tissue, 2 normal regions of lung adenocarcinoma tissue, 10 normal regions of colon cancer tissue, Normal part of one breast cancer-extracted tissue, 24 lung adenocarcinoma cell lines, 1 large cell lung cancer cell line, 2 lung squamous cell carcinoma cell lines, 5 small cell lung cancer cell lines, 89 Total cell RNA derived from cancer cell lines other than lung cancer and 69 normal tissues were used. Cancer cell lines were purchased from ATCC, JCRB or RIKEN, and total RNA from normal tissues was purchased from Clontech, Ambion, Stratagene, Cell Applications, Panomics, Chemicon and Biochain Institute.
- RNA was purified using Trizol® (Invitrogen) according to the method attached to the product. Expression analysis was performed using 1 ⁇ g of total RNA, according to GeneChip, Whole, Transcript (WT), Sense, Target, Labeling, Assay, and Manual (Affymetrix). ExACT (Exon Array Computational Tool) software (Affymetrix) was used to digitize the data.
- the core probe set of Human Exon 1.0 ST Array for HS6ST2 is one in exon 1 (probe set ID; 4022257), five in exon 2 (4022252, 4022253, 4022254, 4022255, 4022256), and one in exon 3 (4022247) One for exon 4 (4022221), two for exon 5 (4022212, 4022213), and seven for exon 6 (4022194, 4022195, 4022196, 4022197, 4022198, 4022198, 1022200).
- the average value of all the core probe sets of that exon was used as the expression data for that exon.
- probe set ID4022253 showed a negative numerical value in all samples, it was judged to be unreliable and was excluded from data analysis.
- Normal tissue expression data are shown in FIGS. 1A and 1B
- lung cancer cell lines and tumor tissue expression data of lung cancer excised tissues are shown in FIGS. 2A and 2B
- expression data of cancer cell lines other than lung cancer are shown in FIGS. 3A and 3B. Indicated.
- Example 2 Preparation of antibodies against HS6ST2 2-1. Cloning of HS6ST2 As shown in Example 1, it was considered that the expression in cancer cells was a variant lacking exons 4 and 5 of HS6ST2. The gene sequence of this variant is registered in RefSeq as NM_147175. UniProt has its amino acid sequence registered as Q96MM7-1. On the other hand, a variant in which amino acid 1-146 of Q96MM7-1 is deleted is registered as Q96MM7-2. Q96MM7-2 corresponds to a protein whose translation is initiated from the 11th base of exon 3.
- HS6ST2 As shown in Example 1, the expression of exons 1 and 2 could not be confirmed in the Human Exon 1.0 ST Array, and the first cloned HS6ST2 was an amino acid sequence corresponding to Q96MM7-2 (Biosynthesis of heparan sulphate with diverse structures and functions: two alternatively spliced forms of human heparan sulphate 6-O-sulphotransferase-2 having different expression patterns and properties.Biochem J. 2003. 371: 131), Q96MM7-2 is expressed dominantly It was considered. Therefore, Q96MM7-2 was named HS6ST2_N-short and cloned.
- Pyrobest DNA Polymerase (Takara Bio) was used for PCR amplification, 3 ⁇ L of 10 ⁇ Pyrobest buffer II, 3 ⁇ L of dNTP mixture, 3 ⁇ L of HuH6 cDNA, 1 ⁇ L of SEQ ID NO: 1 primer (50 ⁇ M), 1 ⁇ L of SEQ ID NO: 2 primer Prepare a solution containing (50 ⁇ M), 0.5 ⁇ L Pyrobest DNA Polymerase, and 18.5 ⁇ L nuclease-free water, and amplify at 94 ° C for 1 minute (94 ° C for 30 seconds, 62 ° C for 30 seconds, 72 ° C for 1.5 minutes) x 35 cycles Went. PCR reamplification was similarly performed using 1 ⁇ L of this PCR product as a template.
- the sequence of TOPO_HS6ST2_N-short was sequenced and confirmed to be the same as RefSeq Accession No. NM_147175.
- HS6ST2_N-short expression CHO cell line HS6ST2_N-short cDNA was cloned into an expression vector for mammalian cells (pMCDN2_ctV5).
- the pMCDN2_ctV5 expression vector is a vector in which expression can be induced under the control of a mouse CMV promoter (GenBank Accession No. U68299) and a neomycin resistance gene is incorporated.
- a V5 tag sequence is added to the 3 ′ side of the inserted target gene.
- the V5 tag is a sequence consisting of 14 amino acids of GKPIPNPLLGLDST that is recognized by an anti-V5 antibody (Invitrogen).
- SEQ ID NO: 3 The sequence of EcoRI recognition sequence-Kozak sequence-start codon-multicloning site (NheI, SalI, NotI) -V5 tag sequence-stop codon of pMCDN2_ctV5 expression vector is shown in SEQ ID NO: 3.
- Primer represented by SEQ ID NO: 4 (EcoRI recognition sequence-Kozak sequence-5 'end sequence of HS6ST2_N-short) and primer represented by SEQ ID NO: 5 (3' end excluding stop codon of NotI recognition sequence-HS6ST2_N-short) PCR amplification was performed using TOPO_HS6ST2_N-short as a template.
- the amplified fragment was digested with EcoRI and NotI and cloned into the EcoRI and NotI sites of pMCDN2_ctV5 (pMCDN2_HS6ST2_N-short_ctV5).
- the base sequence from the start codon to the stop codon of pMCDN2_HS6ST2_N-short_ctV5 is shown in SEQ ID NO: 6, and the amino acid sequence is shown in SEQ ID NO: 7.
- PMCDN2_HS6ST2_N-short_ctV5 digested with PvuI was introduced into the CHO cell line DG44 by electroporation.
- a C-terminal V5-tagged HS6ST2_N-short constant expression CHO cell line was established by selecting the introduced cell line with 500 ⁇ g / mL Geneticin (Invitrogen).
- CHO-S-SFM II medium Invitrogen
- HT supplement Invitrogen
- penicillin / streptomycin Invitrogen
- pMC mammalian cells
- pMC is a vector capable of inducing expression under the control of the mouse CMV promoter.
- PCR amplification using pMCDN2_HS6ST2_N-short_ctV5 as a template using the primer represented by SEQ ID NO: 4 and the primer represented by SEQ ID NO: 8 (SalI recognition sequence-stop codon-3 'end sequence excluding stop codon of HS6ST2_N-short) went.
- the amplified fragment was digested with EcoRI and SalI and cloned into the EcoRI and SalI sites of pMC (pMC_HS6ST2_N-short).
- HS6ST2_N-short is deleted from the N-terminal to the transmembrane region (16th amino acid of 8th to 23rd LLLALVMLFLFAVIVL) and replaced with epidermal growth factor receptor (EGFR, RefSeq Accession No. NM_005228) signal sequence (24 amino acid sequence of MRPSGTAGAALLALLAALCPASRA), and a secreted HS6ST2 (protein with 8 amino acids of DYKDDDDK recognized by anti-FLAG antibody) added to the C-terminus sHS6ST2_FLAG) expression vector was prepared.
- EGFR epidermal growth factor receptor
- Primer represented by SEQ ID NO: 9 (3 ′ end sequence of EGFR signal sequence-sequence after transmembrane region of HS6ST2_N-short) and primer represented by SEQ ID NO: 10 (NotI recognition sequence-stop codon-FLAG tag sequence- PCR amplification was performed using pMCDN2_HS6ST2_N-short_ctV5 as a template using 3 ′ terminal sequence excluding the stop codon of HS6ST2_N-short). Using this PCR product as a template, PCR amplification was performed using a primer represented by SEQ ID NO: 11 (EcoRI recognition sequence-Kozak sequence-5 'end sequence of EGFR signal sequence) and a primer represented by SEQ ID NO: 10.
- pMCDN2_sHS6ST2_FLAG The amplified fragment was digested with EcoRI and NotI and cloned into the EcoRI and NotI sites of pMCDN2 (pMCDN2_sHS6ST2_FLAG).
- pMCDN2 is a vector capable of inducing expression under the control of the mouse CMV promoter and incorporating a neomycin resistance gene.
- the base sequence from the start codon to the stop codon of pMCDN2_sHS6ST2_FLAG is shown in SEQ ID NO: 12, and the amino acid sequence is shown in SEQ ID NO: 13.
- PMCDN2_sHS6ST2_FLAG digested with PvuI was introduced into CHO cell line DG44 by electroporation.
- the sHS6ST2_FLAG constant expression CHO cell line was established by selecting the introduced cell line with Geneticin® (500 ⁇ g / mL). CHO medium was used for the culture.
- SHS6ST2_FLAG was purified from the culture supernatant of the established sHS6ST2_FLAG_CHO cells. Apply culture supernatant to anti-FLAG antibody-binding affinity gel (Sigma), wash with binding buffer (50 M Tris HCl, pH7.6, 150 mM NaCl), and then elution buffer (0.1 M glycine HCl, pH3.5) Eluted with. The eluate was immediately neutralized with a neutralization buffer (1M Tris HCl, pH 8.0) and then replaced with Dulbecco's phosphate-buffered saline (PBS, Invitrogen) using PD10 column (GE Healthcare).
- a neutralization buffer (1M Tris HCl, pH 8.0
- PBS Dulbecco's phosphate-buffered saline
- the concentration of purified sHS6ST2_FLAG was measured using DC-Protein-Assay-Kit-I (Bio-Rad) according to the method attached to the product.
- the attached bovine gamma globulin was used as the standard.
- sHS6ST2_FLAG emulsified with Freund's complete adjuvant (Becton Dickinson) was subcutaneously administered.
- 50 ⁇ g of sHS6ST2_FLAG was administered into the tail vein.
- spleen cells were removed, mixed with mouse myeloma cell line P3-X63Ag8U1 (P3U1, ATCC) to 2: 1, and PEG1500 (Roche Diagnostics) was gradually added to prepare a hybridoma. . After adding RPMI1640 medium (Invitrogen) and centrifugation, PEG1500 was removed by removing the supernatant.
- HAT medium RPMI1640 medium containing 10% fetal bovine serum (FBS), penicillin-streptomycin, 1 ⁇ HAT media supplement (Sigma), 0.5 ⁇ BM-Condimed H1 Hybridoma Cloning Supplement (Roche Diagnostics)
- FBS fetal bovine serum
- penicillin-streptomycin 1 ⁇ HAT media supplement
- 0.5 ⁇ BM-Condimed H1 Hybridoma Cloning Supplement (Roche Diagnostics)
- Screening was performed by measuring the binding of antibodies contained in the culture supernatant to HS6ST2_N-short_ctV5_CHO cells and the parent strain CHO cells using a flow cytometer (FACS Calibur, Becton Dickinson).
- the hybridomas that specifically bound to HS6ST2_N-short_ctV5_CHO cells were continuously cultured, screened again by the same method, and then cloned by the limiting dilution method. As described above, clones A1, A6, and A10 were established as antibodies that specifically bind to HS6ST2.
- mice female, 6 weeks old were subcutaneously administered with 100 ⁇ g of sHS6ST2_FLAG emulsified with Freund's complete adjuvant. After 15 days and 23 days, sHS6ST2_FLAG 50 ⁇ g emulsified with Freund's complete adjuvant was subcutaneously administered. For one animal, 50 ⁇ g of sHS6ST2_FLAG was administered into the tail vein 1 day after the last immunization, and a hybridoma was produced 3 days later.
- sHS6ST2_FLAG 50 ⁇ g was administered into the tail vein 36 days after the last immunization, and a hybridoma was produced 3 days later.
- screening was performed to establish clones B5, B6, C8, and C10 as antibodies that specifically bind to HS6ST2.
- hybridomas were cultured in a HAT medium containing Ultra Low IgG FBS (Invitrogen) instead of FBS, and antibodies were purified from the culture supernatant using a HiTrap Protein G HP 1 mL column (GE Healthcare).
- IsoStrip® Roche
- the antibody concentration was measured using DC-Protein-Assay-Kit-I (Bio-Rad).
- the attached bovine gamma globulin was used as the standard.
- the above-described antibody purification, isotyping, and antibody concentration measurement were performed according to the method attached to the product.
- Example 3 Evaluation of binding of anti-HS6ST2 antibody to mouse HS6ST2 3-1.
- Mouse HS6ST2 has two variants of transcript variant 1 (NM_001077202) and transcript variant 2 (NM_015819) registered in RefSeq. Yes.
- a variant corresponding to HS6ST2_N-short in which N-terminal 146 amino acids and exons 4 and 5 are deleted is transcript variant 2. Therefore, transcript variant 2 was cloned into mouse HS6ST2 (mHS6ST2).
- a primer represented by SEQ ID NO: 14 (EcoRI recognition sequence-Kozak sequence-5 'terminal sequence of mHS6ST2)
- a primer represented by SEQ ID NO: 15 (NotI recognition sequence- PCR amplification was performed using the 3 'terminal sequence of mHS6ST2)
- the amplified product was cloned into pGEM-T Easy vector using pGEM-T Easy Vector Systems (Promega) (pGEM-T_mHS6ST2).
- KOD Plus Ver.2 (Toyobo) was used, 5 ⁇ L of 10 ⁇ KOD Plus Ver.2 buffer, 5 ⁇ L of dNTP mixture, 4 ⁇ L of 25 mM MgSO 4 , 1.5 ⁇ L of the primer of SEQ ID NO: 14 (10 ⁇ M), Prepare a solution containing 1.5 ⁇ L of SEQ ID NO: 15 primer (10 ⁇ M), 4 ⁇ L of mouse spleen cDNA, 1 ⁇ L of KOD Plus Polymerase, 28 ⁇ L of nuclease-free water, 94 ° C. for 2 minutes (98 ° C. for 10 seconds, 72 ° C.
- pMCDN2_ntHA an expression vector for mammalian cells
- pMCDN2_ntHA is a vector capable of inducing expression under the control of the mouse CMV promoter and incorporating a neomycin resistance gene.
- An HA tag sequence is added to the 5 ′ side of the inserted target gene.
- the HA tag sequence is an HA epitope sequence (YPYDVPDYA) derived from influenza hemagglutinin protein, which is recognized by an HA-specific antibody.
- the sequence of EcoRI recognition sequence-Kozak sequence-start codon-HA tag sequence-multicloning site (NheI, SalI, NotI) -stop codon of pMCDN2_ntHA expression vector is shown in SEQ ID NO: 16.
- PCR amplification was performed using pGEM-T_mHS6ST2 as a template, using the primer represented by SEQ ID NO: 17 (NheI recognition sequence—the 5 ′ end sequence excluding the start codon of mHS6ST2) and the primer represented by SEQ ID NO: 15.
- the amplified fragment was digested with NheI and NotI and cloned into the NheI and NotI sites of pMCDN2_ntHA (pMCDN2_mHS6ST2_ntHA).
- mHS6ST2 cDNA was cloned into an expression vector for mammalian cells (pMCDN2_ctV5).
- PCR amplification was performed using pMCDN2_mHS6ST2_ntHA as a template using the primer represented by SEQ ID NO: 17 and the primer represented by SEQ ID NO: 18 (NotI recognition sequence—3 ′ terminal sequence excluding the stop codon of mHS6ST2).
- the amplified fragment was digested with NheI and NotI and cloned into the NheI and NotI sites of pMCDN2_ctV5 (pMCDN2_mHS6ST2_ctV5).
- the base sequence from the start codon to the stop codon of pMCDN2_mHS6ST2_ctV5 is shown in SEQ ID NO: 19, and the amino acid sequence is shown in SEQ ID NO: 20.
- PMCDN2_mHS6ST2_ctV5 digested with PvuI was introduced into CHO cell DG44 by electroporation.
- a C-terminal V5-tagged mHS6ST2 constant expression CHO cell line (mHS6ST2_ctV5_CHO) was established by selecting the introduced cell line with Geneticin® (500 ⁇ g / mL). CHO medium was used for the culture.
- PBS FACS buffer
- anti-HS6ST2 antibody or mouse IgG1 mouse IgG1 (mIgG1, BD Biosciences Pharmingen) was added as a negative control. After reacting for 1 hour on ice, the cells were washed with FACS buffer.
- a FITC-labeled anti-mouse antibody Goat F (ab ′) 2 Fragment Anti-mouse IgG (H + L) -FITC, Beckman Coulter
- FACS buffer supplemented with propidium iodide (PI) 10 ⁇ g / mL (Sigma) and measured with a flow cytometer (FACS Calibur, Becton Dickinson).
- PI propidium iodide
- the measurement data was analyzed using CELLQuest software (Becton Dickinson), and the geo-mean value of FITC fluorescence intensity was calculated for a live cell population that was negative for PI.
- Example 4 Analysis of epitope of anti-HS6ST2 antibody
- GST_HS6ST2_N is the HS6ST2_N-short amino acid sequence from 24 to 175
- GST_HS6ST2_mid is the amino acid sequence from 166 to 317
- GST_HS6ST2_C is the protein that added GST at the N-terminus and His tag at the C-terminus It is.
- the His tag is a tag peptide consisting of 6 consecutive histidine residues.
- Primer represented by SEQ ID NO: 21 (EcoRI recognition sequence-5 'terminal sequence of amino acid sequence 24-175 of HS6ST2_N-short) and primer represented by SEQ ID NO: 22 (NotI recognition sequence-stop codon-His tag sequence PCR amplification was carried out using pMCDN2_HS6ST2_N-short_ctV5 as a template, using the HS6ST2_N-short 24′-175th amino acid sequence (3 ′ terminal sequence). The amplified product was digested with EcoRI and NotI and cloned into the EcoRI and NotI sites of pGEX-6P-1 (pGEX_GST_HS6ST2_N).
- GST_HS6ST2_mid was cloned using the primers represented by SEQ ID NO: 23 and SEQ ID NO: 24, and GST_HS6ST2_C was cloned using the primers represented by SEQ ID NO: 25 and SEQ ID NO: 26 (pGEX_GST_HS6ST2_mid, pGEX_GST_HS6ST2_C).
- GST_HS6ST2_C sequence was divided into two, and a GST fusion protein was prepared for each.
- GST_HS6ST2_C1 is a protein in which 308-393th amino acid sequence is added, GST_HS6ST2_C2 is 379-459th amino acid sequence, GST is added to N-terminal and His tag is added to C-terminal.
- GST_HS6ST2_C1 was cloned using the primers represented by SEQ ID NO: 25 and SEQ ID NO: 27 (pGEX_GST_HS6ST2_C1).
- GST_HS6ST2_C2 was cloned using the primers represented by SEQ ID NO: 26 and SEQ ID NO: 28 (pGEX_GST_HS6ST2_C2).
- GST_HS6ST2_N, GST_HS6ST2_mid, GST_HS6ST2_C, GST_HS6ST2_C1 and GST_HS6ST2_C2 are expressed using BL21 (DE3) et Competent Cells (Takara Bio).
- Western blotting was performed with anti-HS6ST2 antibody.
- the anti-HS6ST2 antibody was used at 10 ⁇ g / mL, the secondary antibody (HRP-antiIgGmIgG, GE Healthcare) was used at a 3000-fold dilution, and detected using ECL Western Blotting Detection Reagents (GE Healthcare).
- anti-HS6ST2 antibodies A1, A10, B5, B6, and C8 bound to GST_HS6ST2_C2, A6 bound to GST_HS6ST2_C1, and C10 bound to GST_HS6ST2_N. Therefore, anti-HS6ST2 antibodies A1, A10, B5, B6, and C8 can bind to the amino acid sequence of 379-459 of HS6ST2_N-short, A6 can bind to the amino acid sequence of 308-393, and C10 can bind to the amino acid sequence of 24-175. It became clear.
- Example 5 Evaluation of antibody-dependent cellular cytotoxicity (ADCC) activity of anti-HS6ST2 antibody
- ADCC activity of the anti-HS6ST2 antibody prepared in Example 2 was measured.
- HS6ST2_N-short_ctV5_CHO cells were used as target cells.
- 1 ⁇ 10 6 HS6ST2_N-short_ctV5_CHO cells were cultured for 1 hour in the presence of Chromium-51 (GE Healthcare). After washing, the concentration was adjusted to 2 ⁇ 10 5 cells / mL with CHO medium, and 50 ⁇ L / well was added to a 96-well plate.
- anti-HS6ST2 antibody prepared to 4 ⁇ g / mL in CHO medium or mIgG1 (BD Biosciences Pharmingen) as a negative control was added at 50 ⁇ L / well.
- effector cells prepared to 5 ⁇ 10 5 / mL with CHO medium were added at 100 ⁇ L / well.
- chimeric proteins containing the extracellular region of mouse Fc-gamma receptor 3 (RefSeq Accession No. NM_010188) and the transmembrane region and intracellular region of human gamma chain RefSeq Accession No.
- NM_004106 were added to NK-92 cells ( A recombinant cell (Japanese Patent Application No. 2007-20155, WO2008 / 093688) forcibly expressed in ATCC was used. After culturing the plate for 4 hours in a 37 ° C, 5% CO 2 incubator, collect 100 ⁇ L / well of the culture supernatant and measure the radioactivity (cpm) using a gamma counter (1480 WIZARD 3 ⁇ , Wallac) The specific chromium release rate (%) was determined using the following formula.
- Specific chromium release rate (%) (AC) x 100 / (BC)
- A is the radioactivity in each well
- B is the average radioactivity of the wells in which cells were lysed at a final concentration of 1% Nonidet P-40
- C is the average radioactivity of wells to which only target cells were added. It is. B and C were performed in triplicate, and others were duplicated, and the average value and standard deviation of the specific chromium release rate were calculated.
- anti-HS6ST2 antibodies A6, B5, B6, and C8 have ADCC activity (FIG. 6).
- Example 6 Evaluation of anti-tumor activity of anti-HS6ST2 antibody using Mab-ZAP The potential of the anti-HS6ST2 antibody prepared in Example 2 as an immunotoxin was evaluated using Mab-ZAP (Advanced Targeting Systems).
- Mab-ZAP is an antibody obtained by labeling saporin with a goat anti-mouse IgG antibody. Saporin is a toxin that inhibits protein synthesis in the ribosome.
- a mouse antibody that binds to an antigen on the cell surface and Mab-ZAP are added to the culture system at the same time, when the mouse antibody is taken into the cell, Mab-ZAP is also taken into the cell and suppresses cell growth. .
- HS6ST2_N-short_ctV5_CHO cells were used as target cells.
- HS6ST2_N-short_ctV5_CHO cells prepared at 1 ⁇ 10 5 cells / mL in CHO medium were seeded in a 96-well plate at 50 ⁇ L / well and cultured in a 37 ° C., 5% CO 2 incubator.
- anti-HS6ST2 antibody and Mab-ZAP were added to make 100 ⁇ L / well, and further cultured for 2 days.
- the anti-HS6ST2 antibody had a final concentration of 20 ng / mL
- Mab-ZAP had a final concentration of 100 ng / mL.
- MIgG1 (BD Biosciences Pharmingen) was used as a negative control. After culturing, 10 ⁇ L / well of live cell measurement reagent SF (Nacalai Tesque) was added and further cultured for 1.5 hours, and then the absorbance at 450 nm to 655 nm was measured. The experiment was performed in duplicate, and the average value and standard deviation of absorbance were calculated.
- Example 7 Evaluation of binding activity to HS6ST2 variant 7-1.
- Cloning of HS6ST2 variant HS6ST2 has a variant with a long N-terminal amino acid sequence (UniProt Q96MM7-1, RefSeq Accession No. NM_147175). This variant was named HS6ST2_N-long, and it was confirmed that the anti-HS6ST2 antibody prepared in Example 2 was bound.
- HS6ST2_N-short In the open reading frame of HS6ST2_N-short, there is a NheI recognition sequence near the 5 ′ end. Therefore, the HS6ST2_N-short sequence already cloned downstream from this NheI recognition sequence was used, and the HS6ST2_N-long sequence upstream was further cloned.
- total RNA was extracted from cancer cell line HuH6 (RIKEN) using Trizol (Invitrogen) according to the method attached to the product, and further cDNA was prepared using SuperScript III Reverse Transcriptase (Invitrogen) according to the method attached to the product.
- the primer represented by SEQ ID NO: 29 (HS6ST2_N-long 5'-UTR sequence) and the primer represented by SEQ ID NO: 30 (sequence downstream from the NheI recognition sequence of HS6ST2_N-short) were used.
- PCR amplification was performed, and the amplified product was cloned into pGEM-T Easy vector using pGEM-T Easy Vector Systems (Promega) (pGEM-T_HS6ST2_N).
- KOD Plus Ver.2 (Toyobo) was used, 5 ⁇ L of 10 ⁇ KOD Plus Ver.2 buffer, 5 ⁇ L of dNTP mixture, 3 ⁇ L of 25 mM MgSO 4 , 1 ⁇ L of primer of SEQ ID NO: 29 (10 ⁇ M), 1 ⁇ L Prepare a solution containing the primer of SEQ ID NO: 30 (10 ⁇ M), 2 ⁇ L of HuH6 cDNA, 1 ⁇ L of KOD Plus Polymerase, 33 ⁇ L of nuclease-free water, 94 ° C. for 2 minutes, (94 ° C. for 30 seconds, 58 ° C. for 30 seconds, 68 Amplification was performed at 35 ° C. for 3 minutes and 68 ° C. for 3 minutes.
- the sequence of pGEM-T_HS6ST2_N was sequenced and confirmed to be the same as the sequence of the corresponding part of RefSeq Accession No. NM_147175.
- PCR amplification is performed using the primer represented by SEQ ID NO: 31 (EcoRI recognition sequence-Kozak sequence-5 'end sequence of HS6ST2_N-long) and the primer represented by SEQ ID NO: 30
- SEQ ID NO: 31 EcoRI recognition sequence-Kozak sequence-5 'end sequence of HS6ST2_N-long
- SEQ ID NO: 30 The product was cloned into a TOPO vector (TOPO_HS6ST2_N) using TOPOOTA Cloning Kit (Invitrogen).
- TOPO_HS6ST2_N was digested with EcoRI and NheI and cloned into the EcoRI and NheI sites of pMCDN2_HS6ST2_N-short_ctV5 to prepare a C6 V5 tagged HS6ST2_N-long expression vector (pMCDN2_HS6ST2_N-long_ctV5).
- the base sequence from the start codon to the stop codon of pMCDN2_HS6ST2_N-long_ctV5 is shown in SEQ ID NO: 32, and the amino acid sequence is shown in SEQ ID NO: 33.
- HS6ST2_N-long-expressing CHO cell line PvuI digested pMCDN2_HS6ST2_N-long_ctV5 was introduced into CHO cell DG44 by electroporation. By selecting the introduced cell line with Geneticin (500 ⁇ g / mL), a C-terminal V5-tagged HS6ST2_N-long constant expression CHO cell line (HS6ST2_N-long_ctV5_CHO) was established. CHO medium was used for the culture.
- Example 8 Expression analysis of HS6ST2 in cancer cell lines 8-1. Expression analysis of HS6ST2 by flow cytometry Using the anti-HS6ST2 antibody prepared in Example 2, the expression of HS6ST2 on the cell membrane of cancer cell lines is a flow site Evaluation was performed using a meter.
- the primary antibody is anti-HS6ST2 antibody B6 or negative control mIgG1 (BD Biosciences Pharmingen), and the cells are the lung adenocarcinoma cell line ABC-1 and the liver cancer cell line HuH6, which have the highest HS6ST2 expression in the Human Exon 1.0 ST Array. Used (Example 1).
- Flow cytometry was performed in the same manner as in Example 3, and the primary antibody was used at a concentration of 10 ⁇ g / mL. As a result, the expression of HS6ST2 was confirmed on the cell membrane in any cell (FIG. 9).
- N-Glycosidase F PNGaseF, New England Biolabs
- Samples were subjected to SDS-PAGE electrophoresis with Multigel II Mini (8/16, Cosmo Bio), transferred to PVDF membrane (Immobilon-P, Millipore), and anti-HS6ST2 antibody C10 or anti-V5 tag antibody (Invitrogen) was used.
- Western blot was performed.
- Anti-HS6ST2 antibody C10 was used at 5 ⁇ g / mL
- anti-V5 tag antibody was used at a 5000-fold dilution, and reacted at room temperature for 1 hour.
- an HRP-labeled anti-mouse IgG antibody (GE Healthcare) was used and reacted at room temperature for 1 hour. Finally, color was developed using ECL Western Blotting Detection Reagents (GE Healthcare), and the band was detected by exposing to X-ray film.
- the molecular weight estimated from the amino acid sequence is 55 kDa and 71 kDa respectively for HS6ST2_N-short_ctV5 and HS6ST2_N-long_ctV5.
- N-Glycosidase F By removing the N-type sugar chain with N-Glycosidase F, a band of this estimated molecular weight was confirmed (FIG. 10A).
- the anti-HS6ST2 antibody C10 showed the same band as the anti-V5 tag antibody, it was confirmed that it could be used for Western blotting.
- soluble HS6ST2 secreted in the culture supernatant of HS6ST2_N-short_ctV5_CHO cells and HS6ST2_N-long_ctV5_CHO cells was detected by Western blot.
- the culture supernatant of HS6ST2_N-short_ctV5_CHO cells and HS6ST2_N-long_ctV5_CHO cells was treated with N-Glycosidase F and Western blotted with anti-HS6ST2 antibody C10 and anti-V5 tag antibody, a band was confirmed around 50-60kDa ( Figure 10B).
- HS6ST2_N-short and HS6ST2_N-long had the same molecular weight, it was considered that both were cleaved at the same site.
- the downstream side of the transmembrane region (16 amino acids of LLLALVMLFLFAVIVL of HS6ST2_N-short from 8 to 23rd from the N terminus, and 1564 to 169th from HS6ST2_N-long) may be cleaved. It is known that the downstream side of the transmembrane region is also cleaved in HS6ST1 (Molecular characterization and expression expression of heparan-sulfate 6-sulfotransferase. J J Biol Chem. 1998. 273: 9208).
- HS6ST2 was expressed in lung adenocarcinoma cell lines ABC-1, NCI-H441, NCI-H1781 and ovarian cancer cell lines OVMANA (FIG. 11A). All were considered to be N-short variants because the molecular weight after N-Glycosidase F treatment was 50-60 kDa.
- A549 is a cell line that was confirmed not to express HS6ST2 in Example 1, and was used as a negative control.
- HS6ST2 in the culture supernatant of these cells was measured.
- the culture supernatant was filtered through 0.22 ⁇ m, concentrated 50 times using Amicon® Ultra® (10 kDa cut, Millipore), and used for Western blotting (FIG. 11B). Since the protein contained in FBS is affected, the cancer cell line cultured in a 10 cm culture dish was cultured overnight in a medium (5 ml) containing no FBS, and the culture supernatant was used. As a result, soluble HS6ST2 was detected in the culture supernatant of cancer cell lines, indicating that soluble HS6ST2 can be a diagnostic marker for cancer.
- the cells after recovering the culture supernatant were dissolved in lysis buffer and the protein concentration was measured with DC Protein Protein Assay Kit I (Bio-Rad).
- the amount of protein per petri dish was A549, ABC-1, NCI- H441, NCI-1781, and OVMANA were 1.4, 2.1, 1.8, 1.9, and 2.0 mg respectively, confirming that the number of cells was almost the same.
- Example 9 Binding of soluble HS6ST2 to heparan sulfate and anti-tumor effect of anti-HS6ST2 antibody 9-1. Binding of soluble HS6ST2 to heparan sulfate Heparan on cell membrane is soluble HS6ST2 secreted from cells It was confirmed to bind to sulfuric acid. First, the expression of heparan sulfate on the cell membrane was examined by flow cytometry in the same manner as in Example 3. The cells used were lung adenocarcinoma cell lines A549, ABC-1, NCI-H441, NCI-H1781, ovarian cancer cell line OVMANA and CHO cell DG44.
- Primary antibodies include anti-heparan sulfate antibody (HepSS-1, biochemical biobusiness) and mouse IgM (mIgM, BD Biosciences Pharmingen) as negative control at 20 ⁇ g / mL (NCI-H441, NCI-H1781) or 40 ⁇ g / mL (A549 , ABC-1, OVMANA, DG44). As a result, it was confirmed that A549, ABC-1 and DG44 express heparan sulfate (FIG. 12).
- heparan sulfate on the cell membrane was digested with enzymes, and the binding of soluble HS6ST2 to cells was examined.
- 1 ⁇ 10 6 DG44 cells in 37 mL of 3 mL of CHO-S-SFM II medium containing 1 mU / mL heparinase, heparitinase I and heparitinase II (both biochemical biobusiness), HT supplement, penicillin / streptomycin Culturing was carried out at 0 ° C. for 2 hours.
- Heparinase, heparitinase I and heparitinase II are enzymes that specifically degrade heparan sulfate. In the cells treated with the enzyme, the expression of heparan sulfate was greatly reduced (FIG. 13A).
- sHS6ST2_FLAG soluble HS6ST2
- DG44 cells were treated with enzyme, and reacted with sHS6ST2_FLAG prepared at 50 ⁇ g / mL in FACS buffer at 4 ° C. for 3 hours. After washing, flow cytometry analysis was performed in the same manner as in Example 3.
- the primary antibody used was anti-HS6ST2 antibody C8 and mIgG1 (BD Biosciences Pharmingen) as a negative control.
- A549 cells were prepared at a concentration of 1 ⁇ 10 4 cells / mL in DMEM medium (Invitrogen) containing 10% FBS and penicillin / streptomycin, and seeded at 50 ⁇ L / well in a 96-well plate. At this time, wells containing and not containing 50 ⁇ g / mL sHS6ST2_FLAG were prepared. After culturing at 37 ° C.
- Example 10 Measurement of soluble HS6ST2 by ELISA Since the cancer cell line secreted soluble HS6ST2 in Example 8 and could be a diagnostic marker for cancer, ELISA for detecting soluble HS6ST2 A system was constructed. Anti-HS6ST2 antibodies A6, B5, and C8 were used as antibodies. B5 and C8 bind to the 379-459th amino acid sequence of HS6ST2_N-short, and A6 binds to the 308-393th amino acid sequence (Example 4). C8 binds to mHS6ST2 but does not bind B5 (Example 3). Therefore, the binding sites of the three were considered different.
- anti-HS6ST2 antibodies B5 and C8 were labeled with biotin using Biotin Protein Labeling Kit (Roche) according to the method attached to the product (B5-biotin, C8-biotin).
- the concentration of the biotin-labeled antibody was measured using DC Protein Assay Kit I (Bio-Rad).
- Anti-HS6ST2 antibody A6 was prepared to 5 ⁇ g / mL with coating buffer (0.1 M NaHCO 3 , pH 9.6, 0.02% NaN 3 ) and added to ELISA 96-well plate (F96 Cert. Maxisorp, Nunc) at 100 ⁇ L / well. .
- the antibody was bound to the plate by allowing to stand at room temperature for 1 hour.
- dilution buffer 50 mM Tris-HCl, pH8.1 , 150 mM NaCl, 1 mM MgCl 2, 0.05% Tween20, 1% bovine serum albumin, 0.02% NaN 3) to each 200 [mu] L / well Then, blocking was performed by allowing to stand at room temperature for 1 hour. After removing the supernatant, 50 ⁇ L / well of sample was added and allowed to react at room temperature for 1 hour.
- a culture supernatant of lung adenocarcinoma cell line ABC-1 and a culture supernatant of HS6ST2_N-short_ctV5_CHO cells diluted 500-fold with RPMI1640 medium supplemented with 10% FBS were used. After washing the plate 3 times with rinse buffer (50 mM Tris-HCl, pH 7.6, 150 mM NaCl, 0.05% Tween20), 100 ⁇ L / well of dilution buffer containing 3 ⁇ g / mL B5-biotin and 3 ⁇ g / mL C8-biotin They were added one by one and reacted at room temperature for 1 hour.
- rinse buffer 50 mM Tris-HCl, pH 7.6, 150 mM NaCl, 0.05% Tween20
- the soluble HS6ST2 concentrations in the culture supernatants of lung adenocarcinoma cell lines ABC-1 and HS6ST2_N-short_ctV5_CHO cells were 9.5 ng / mL and 1.8 ⁇ g / mL, respectively.
- a calibration curve is shown in FIG.
- heparan sulfate As a diagnostic marker for cancer, it can be preferably measured using peripheral blood. Because heparan sulfate is present in the blood (Solubleynsyndecan-1 and serum basic fibroblastrowgrowth factor are newgnoprognostic factors in lung cancer. Cancer Res 62: 5210 (2002), Serum and urinary concentrations of heindiapatient Kidney Int 56: 650 (1999), Isolation and characterization of glycosaminoglycans in human plasma. J Clin Invest 76: 1984 (1985)), soluble HS6ST2 binds to heparan sulfate in blood and inhibits detection by ELISA The possibility was considered. Therefore, ELISA was examined in the presence of human serum or heparan sulfate.
- SHS6ST2_FLAG was adjusted to 1 ⁇ g / mL using Dilution buffer, dilution buffer added with 100 ⁇ g / mL heparan sulfate (Biochemical Biobusiness), or human serum (Cosmo Bio), and allowed to stand at room temperature for 1 hour.
- dilution buffer 4 steps of dilution from 100 ng / mL at a common ratio of 10 were measured by ELISA, and color development was confirmed even in the presence of heparan sulfate and human serum (FIG. 16). From the above, it was shown that the established ELISA system is not affected by human serum.
- Example 11 Analysis of HS6ST2 expression in lung cancer by immunohistochemical staining Because HS6ST2 gene expression was increased in lung adenocarcinoma (Example 1), expression of HS6ST2 protein in lung cancer was analyzed by immunohistochemical staining. . A 4% paraformaldehyde-fixed AMeX-embedded paraffin block was prepared from each specimen, and 5 ⁇ m sliced sections were prepared. These sections were stained immunohistochemically as follows using the Ventana HX Discovery System (Ventana Medical Systems).
- Blocker D Ventana Medical Systems
- streptavidin horseradish peroxidase Ventana Medical Systems
- DAB map solution Ventana Medical Systems
- DAB map solution Ventana Medical Systems
- DAB map solution Ventana Medical Systems
- Lung macrophages and bronchial epithelial cells are known to express heparan sulfate (Heterogeneity of heparan sulfates in human lung. Am J Respir Cell Mol Biol. 2004. 30: 166) soluble soluble secreted from cancer cells The possibility that type HS6ST2 was bound was also considered.
- Example 12 Determination of variable region gene sequence of anti-HS6ST2 antibody The nucleic acid sequence and amino acid sequence of the variable region of the anti-HS6ST2 antibody prepared in Example 2 were determined. Total RNA was purified from 1 ⁇ 10 6 hybridoma cells producing each antibody using Trizol (Invitrogen) according to the method attached to the product.
- Trizol Invitrogen
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Urology & Nephrology (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Hematology (AREA)
- Biomedical Technology (AREA)
- Biochemistry (AREA)
- Cell Biology (AREA)
- Analytical Chemistry (AREA)
- Pathology (AREA)
- Biotechnology (AREA)
- Oncology (AREA)
- Biophysics (AREA)
- Microbiology (AREA)
- Hospice & Palliative Care (AREA)
- Food Science & Technology (AREA)
- Physics & Mathematics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- General Physics & Mathematics (AREA)
- Genetics & Genomics (AREA)
- Gastroenterology & Hepatology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Peptides Or Proteins (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
Description
[1] HS6ST2タンパク質に結合する抗体。
[2] 細胞傷害活性を有することを特徴とする[1]に記載の抗体。
[3] 細胞傷害性物質が結合していることを特徴とする[1]または[2]に記載の抗体。
[4] ヘパラン硫酸に結合したHS6ST2に結合することを特徴とする[1]~[3]のいずれかに記載の抗体。
[5] 細胞膜上に発現したHS6ST2に結合することを特徴とする[1]~[3]のいずれかに記載の抗体。
[6] 以下のいずれかに記載の抗体であることを特徴とする[1]~[5]のいずれかに記載の抗体:
(1)配列番号:64に記載のアミノ酸配列を有する重鎖CDR1、配列番号:65に記載のアミノ酸配列を有する重鎖CDR2、配列番号:66に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(A1);
(2)配列番号:70に記載のアミノ酸配列を有する重鎖CDR1、配列番号:71に記載のアミノ酸配列を有する重鎖CDR2、配列番号:72に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(A6);
(3)配列番号:76に記載のアミノ酸配列を有する重鎖CDR1、配列番号:77に記載のアミノ酸配列を有する重鎖CDR2、配列番号:78に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(A10);
(4)配列番号:82に記載のアミノ酸配列を有する重鎖CDR1、配列番号:83に記載のアミノ酸配列を有する重鎖CDR2、配列番号:84に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(B5);
(5)配列番号:88に記載のアミノ酸配列を有する重鎖CDR1、配列番号:89に記載のアミノ酸配列を有する重鎖CDR2、配列番号:90に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(B6);
(6)配列番号:94に記載のアミノ酸配列を有する重鎖CDR1、配列番号:95に記載のアミノ酸配列を有する重鎖CDR2、配列番号:96に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(C8);
(7)配列番号:100に記載のアミノ酸配列を有する重鎖CDR1、配列番号:101に記載のアミノ酸配列を有する重鎖CDR2、配列番号:102に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(C10);
(8)配列番号:67に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:68に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:69に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(A1);
(9)配列番号:73に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:74に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:75に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(A6);
(10)配列番号:79に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:80に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:81に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(A10);
(11)配列番号:85に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:86に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:87に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(B5);
(12)配列番号:91に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:92に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:93に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(B6);
(13)配列番号:97に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:98に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:99に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(C8);
(14)配列番号:103に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:104に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:105に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(C10);
(15)(1)の重鎖可変領域と(8)の軽鎖可変領域を含む抗体(A1);
(16)(2)の重鎖可変領域と(9)の軽鎖可変領域を含む抗体(A6);
(17)(3)の重鎖可変領域と(10)の軽鎖可変領域を含む抗体(A10);
(18)(4)の重鎖可変領域と(11)の軽鎖可変領域を含む抗体(B5);
(19)(5)の重鎖可変領域と(12)の軽鎖可変領域を含む抗体(B6);
(20)(6)の重鎖可変領域と(13)の軽鎖可変領域を含む抗体(C8);
(21)(7)の重鎖可変領域と(14)の軽鎖可変領域を含む抗体(C10);
(22)(1)から(21)のいずれかに記載の抗体において1若しくは複数のアミノ酸が置換、欠失、付加および/または挿入された抗体であって、(1)から(21)のいずれかに記載の抗体と同等の活性を有する抗体;
(23)(1)から(21)のいずれかに記載の抗体が結合するHS6ST2タンパク質のエピトープと同じエピトープに結合する抗体。
[7] 配列番号:107のアミノ酸配列を有するHS6ST2タンパク質の379番目のアミノ酸から459番目のアミノ酸までの部位、308番目のアミノ酸から393番目のアミノ酸までの部位、または24番目のアミノ酸から175番目のアミノ酸までの部位を認識することを特徴とする請求項1~5いずれかに記載の抗体。
[8] [1]~[7]のいずれかに記載の抗体を有効成分として含む医薬組成物。
[9] 抗癌剤である[8]に記載の医薬組成物。
[10] 以下の工程を含む癌の診断方法:
(a)被験者から採取された試料を提供する工程、
(b) (a)の試料に含まれるHS6ST2タンパク質又はHS6ST2遺伝子を検出する工程。
[11] 細胞傷害性物質が結合したHS6ST2タンパク質。
[12] [11]に記載のHS6ST2タンパク質を有効成分として含む医薬組成物。
[13] 抗癌剤である[12]に記載の医薬組成物。
本発明で用いられるHS6ST2タンパク質は特に限定されず、当業者に公知のHS6ST2タンパク質を用いることが可能である。HS6ST2タンパク質は好ましくはヒトHS6ST2である。ヒトHS6ST2は複数のバリアントが存在しているが、本発明で用いられるHS6ST2は如何なるバリアントでもよい。HS6ST2のバリアントの例として、エクソン4と5が欠損したバリアント(GenBank Accession No:NM_147175、UniProt:Q96MM7-1、配列番号:106)、Q96MM7-1の1番目~146番目のアミノ酸が欠損したバリアント(UniProt:Q96MM7-2、配列番号:107)、第三のバリアント(UniProt:Q96MM7-3、配列番号:108)を挙げることができる。
本発明で用いられる抗HS6ST2抗体は、HS6ST2タンパク質に結合すればよく、その由来、種類、形状などは問われない。具体的には、非ヒト動物の抗体(例えば、マウス抗体、ラット抗体、ラクダ抗体)、ヒト抗体、キメラ抗体、ヒト化抗体などの公知の抗体が使用できる。本発明においては、モノクローナル、あるいはポリクローナルを抗体として利用することができるが好ましくはモノクローナル抗体である。抗体のHS6ST2タンパク質への結合は特異的な結合であることが好ましい。
・ヒトHS6ST2のアミノ酸配列に基づいて化学合成によって取得されたペプチド。
・HS6ST2遺伝子の一部を発現ベクターに組込んで発現させることによって取得されたペプチド。
・HS6ST2タンパク質をタンパク質分解酵素により分解することによって取得されたペプチド。
・HS6ST2のような膜タンパク質の構造を維持して免疫刺激を与えることができる。
・免疫抗原を精製する必要が無い。
・P3(P3x63Ag8.653)(J. Immunol.(1979)123, 1548-1550)
・P3x63Ag8U.1(Current Topics in Microbiology and Immunology(1978)81, 1-7)
・NS-1(Kohler. G. and Milstein, C. Eur. J. Immunol.(1976)6, 511-519)
・MPC-11(Margulies. D.H. et al., Cell(1976)8, 405-415)
・SP2/0(Shulman, M. et al., Nature(1978)276, 269-270)
・FO(de St. Groth, S. F. et al., J. Immunol. Methods(1980)35, 1-21)
・S194(Trowbridge, I. S. J. Exp. Med.(1978)148, 313-323)、
・R210(Galfre, G. et al., Nature(1979)277, 131-133)等
基本的には公知の方法、たとえば、ケーラーとミルステインらの方法(Kohler. G. and Milstein, C., Methods Enzymol.(1981)73, 3-46)等に準じて、免疫細胞とミエローマ細胞との細胞融合が行われる。
・グアニジン超遠心法(Chirgwin, J. M. et al., Biochemistry(1979)18, 5294-5299)
・AGPC法(Chomczynski, P.et al., Anal. Biochem.(1987)162, 156-159)
(1)哺乳類細胞、:CHO、COS、ミエローマ、BHK(baby hamster kidney)、Hela、Vero、HEK293、Ba/F3、HL-60、Jurkat、SK-HEP1など。
(2)両生類細胞:アフリカツメガエル卵母細胞など。
(3)昆虫細胞:sf9、sf21、Tn5など。
・アミノグリコシドトランスフェラーゼ(APH)遺伝子
・チミジンキナーゼ(TK)遺伝子
・大腸菌キサンチングアニンホスホリボシルトランスフェラーゼ(Ecogpt)遺伝子
・ジヒドロ葉酸還元酵素(dhfr)遺伝子等
パパイン消化:F(ab)2またはFab
ペプシン消化:F(ab’)2またはFab’
抗体のH鎖またはH鎖V領域をコードするDNA配列、および
抗体のL鎖またはL鎖V領域をコードするDNA配列
[VL]リンカー[VH]リンカー[VH]リンカー[VL]
[VH]リンカー[VL]リンカー[VL]リンカー[VH]
[VH]リンカー[VH]リンカー[VL]リンカー[VL]
[VL]リンカー[VL]リンカー[VH]リンカー[VH]
[VL]リンカー[VH]リンカー[VL]リンカー[VH]
Ser
Gly・Ser
Gly・Gly・Ser
Ser・Gly・Gly
Gly・Gly・Gly・Ser(配列番号:109)
Ser・Gly・Gly・Gly(配列番号:110)
Gly・Gly・Gly・Gly・Ser(配列番号:111)
Ser・Gly・Gly・Gly・Gly(配列番号:112)
Gly・Gly・Gly・Gly・Gly・Ser(配列番号:113)
Ser・Gly・Gly・Gly・Gly・Gly(配列番号:114)
Gly・Gly・Gly・Gly・Gly・Gly・Ser(配列番号:115)
Ser・Gly・Gly・Gly・Gly・Gly・Gly(配列番号:116)
(Gly・Gly・Gly・Gly・Ser(配列番号:111))n
(Ser・Gly・Gly・Gly・Gly(配列番号:112))n
[nは1以上の整数である]
[VH]ペプチドリンカー(15アミノ酸)[VL]ペプチドリンカー(15アミノ酸)[VH]ペプチドリンカー(15アミノ酸)[VL]
N-ヒドロキシスクシンイミド(NHS)、
ジスクシンイミジルスベレート(DSS)、
ビス(スルホスクシンイミジル)スベレート(BS3)、
ジチオビス(スクシンイミジルプロピオネート)(DSP)、
ジチオビス(スルホスクシンイミジルプロピオネート)(DTSSP)、
エチレングリコールビス(スクシンイミジルスクシネート)(EGS)、
エチレングリコールビス(スルホスクシンイミジルスクシネート)(スルホ-EGS)、
ジスクシンイミジル酒石酸塩(DST)、ジスルホスクシンイミジル酒石酸塩(スルホ-DST)、
ビス[2-(スクシンイミドオキシカルボニルオキシ)エチル]スルホン(BSOCOES)、および
ビス[2-(スルホスクシンイミドオキシカルボニルオキシ)エチル]スルホン(スルホ-BSOCOES)など
4つの抗体可変領域を結合する場合には、通常、3つのリンカーが必要となる。複数のリンカーは、同じでもよいし、異なるリンカーを用いることもできる。本発明において好ましい低分子化抗体はダイアボディー又はsc(Fv)2である。このような低分子化抗体を得るには、抗体を酵素、例えば、パパイン、ペプシンなどで処理し、抗体断片を生成させるか、又はこれら抗体断片をコードするDNAを構築し、これを発現ベクターに導入した後、適当な宿主細胞で発現させればよい(例えば、Co, M. S. et al., J. Immunol. (1994) 152, 2968-2976 ; Better, M. and Horwitz, A. H., Methods Enzymol. (1989) 178, 476-496 ; Pluckthun, A. and Skerra, A., Methods Enzymol. (1989) 178, 497-515 ; Lamoyi, E., Methods Enzymol. (1986) 121, 652-663 ; Rousseaux, J. et al., Methods Enzymol. (1986) 121, 663-669 ; Bird, R. E. and Walker, B. W., Trends Biotechnol. (1991) 9, 132-137参照)。
グリコシル化が修飾された抗体(WO99/54342など)、
糖鎖に付加するフコースが欠損した抗体(WO00/61739、WO02/31140など))、
バイセクティングGlcNAcを有する糖鎖を有する抗体(WO02/79255など)など
CBA/Nマウスなどから脾臓を摘出し、RPMI1640培地(Invitrogen社製)中で脾臓細胞が分離される。10%ウシ胎児血清(FBS、HyClone社製)を含む同培地で洗浄後、細胞濃度を5×106/mlに調製することによって、エフェクター細胞が調製できる。
Baby Rabbit Complement(CEDARLANE社製)を10% FBS含有培地(Invitrogen社製)にて10倍希釈し、補体溶液が調製できる。
HS6ST2タンパク質を発現する細胞を0.2 mCiの51Cr-クロム酸ナトリウム(GEヘルスケアバイオサイエンス社製)とともに、10% FBS含有DMEM培地中で37℃にて1時間培養することにより該標的細胞を放射性標識できる。HS6ST2タンパク質を発現する細胞としては、HS6ST2タンパク質をコードする遺伝子で形質転換された細胞、肺腺癌細胞、肺癌細胞、肝臓癌細胞、卵巣癌細胞、肺扁平上皮癌細胞等を利用することができる。放射性標識後、細胞を10% FBS含有RPMI1640培地にて3回洗浄し、細胞濃度を2×105/mlに調製することによって、該標的細胞が調製できる。
(1)配列番号:64に記載のアミノ酸配列を有する重鎖CDR1、配列番号:65に記載のアミノ酸配列を有する重鎖CDR2、配列番号:66に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(A1);
(2)配列番号:70に記載のアミノ酸配列を有する重鎖CDR1、配列番号:71に記載のアミノ酸配列を有する重鎖CDR2、配列番号:72に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(A6);
(3)配列番号:76に記載のアミノ酸配列を有する重鎖CDR1、配列番号:77に記載のアミノ酸配列を有する重鎖CDR2、配列番号:78に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(A10);
(4)配列番号:82に記載のアミノ酸配列を有する重鎖CDR1、配列番号:83に記載のアミノ酸配列を有する重鎖CDR2、配列番号:84に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(B5);
(5)配列番号:88に記載のアミノ酸配列を有する重鎖CDR1、配列番号:89に記載のアミノ酸配列を有する重鎖CDR2、配列番号:90に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(B6);
(6)配列番号:94に記載のアミノ酸配列を有する重鎖CDR1、配列番号:95に記載のアミノ酸配列を有する重鎖CDR2、配列番号:96に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(C8);
(7)配列番号:100に記載のアミノ酸配列を有する重鎖CDR1、配列番号:101に記載のアミノ酸配列を有する重鎖CDR2、配列番号:102に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(C10);
(8)配列番号:67に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:68に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:69に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(A1);
(9)配列番号:73に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:74に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:75に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(A6);
(10)配列番号:79に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:80に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:81に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(A10);
(11)配列番号:85に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:86に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:87に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(B5);
(12)配列番号:91に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:92に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:93に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(B6);
(13)配列番号:97に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:98に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:99に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(C8);
(14)配列番号:103に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:104に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:105に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(C10);
(15)(1)の重鎖可変領域と(8)の軽鎖可変領域を含む抗体(A1);
(16)(2)の重鎖可変領域と(9)の軽鎖可変領域を含む抗体(A6);
(17)(3)の重鎖可変領域と(10)の軽鎖可変領域を含む抗体(A10);
(18)(4)の重鎖可変領域と(11)の軽鎖可変領域を含む抗体(B5);
(19)(5)の重鎖可変領域と(12)の軽鎖可変領域を含む抗体(B6);
(20)(6)の重鎖可変領域と(13)の軽鎖可変領域を含む抗体(C8);
(21)(7)の重鎖可変領域と(14)の軽鎖可変領域を含む抗体(C10);
(22)(1)から(21)のいずれかに記載の抗体において1若しくは複数のアミノ酸が置換、欠失、付加および/または挿入された抗体であって、(1)から(21)のいずれかに記載の抗体と同等の活性を有する抗体;
(23)(1)から(21)のいずれかに記載の抗体が結合するHS6ST2タンパク質のエピトープと同じエピトープに結合する抗体。
疎水性アミノ酸(A、I、L、M、F、P、W、Y、V)、
親水性アミノ酸(R、D、N、C、E、Q、G、H、K、S、T)、
脂肪族側鎖を有するアミノ酸(G、A、V、L、I、P)、
水酸基含有側鎖を有するアミノ酸(S、T、Y)、
硫黄原子含有側鎖を有するアミノ酸(C、M)、
カルボン酸及びアミド含有側鎖を有するアミノ酸(D、N、E、Q)、
塩基含有側鎖を有するアミノ酸(R、K、H)、
芳香族含有側鎖を有するアミノ酸(H、F、Y、W)
(括弧内はいずれもアミノ酸の一文字標記を表す)
本発明はさらに、細胞傷害性物質が結合したHS6ST2タンパク質を提供する。細胞傷害性物質が結合したHS6ST2タンパク質は、ヘパリン硫酸に結合した後、細胞内にインターナライズされると考えられるので、抗癌剤等に用いることが可能である。
別の観点においては、本発明は、HS6ST2タンパク質に結合する抗体を有効成分として含有する医薬組成物を提供する。また、本発明はHS6ST2タンパク質に結合する抗体を有効成分として含有する細胞増殖抑制剤、特に抗癌剤に関する。本発明の細胞増殖抑制剤および抗癌剤は、癌を罹患している対象または罹患している可能性がある対象に投与されることが好ましい。HS6ST2の発現レベルは癌細胞で亢進していることから、抗HS6ST2抗体の投与によって、癌細胞特異的な細胞傷害作用が得られると考えられる。
また、本発明は、HS6ST2発現細胞とHS6ST2タンパク質に結合する抗体とを接触させることによりHS6ST2発現細胞に傷害を引き起こす方法又は細胞の増殖を抑制する方法を提供する。
癌の診断方法
また、本発明はHS6ST2タンパク質またはHS6ST2タンパク質をコードする遺伝子を検出することを特徴とする癌の診断方法を提供する。HS6ST2は種々の癌組織あるいは癌細胞株において顕著な発現亢進が確認されている。したがって、HS6ST2は、癌を特異的に検出するためのマーカーとして有用である。
(a)被験者から採取された試料を提供する工程、
(b) (a)の試料に含まれるHS6ST2タンパク質又はHS6ST2遺伝子を検出する工程。
(a) 被検者から採取された試料を提供する工程;
(b) 採取された試料に含まれるHS6ST2タンパク質を、HS6ST2タンパク質に結合する抗体を用いて検出する工程。
・単にHS6ST2タンパク質が存在するか否かの測定
・HS6ST2タンパク質が一定の量以上存在するか否かの測定
・HS6ST2タンパク質の量を他の試料(例えば、コントロール試料など)と比較する測定など
(1) 被検者から採取された生体試料中のHS6ST2発現レベルを検出する工程、および
(2) (1)で検出されたHS6ST2の発現レベルが、対照と比較して高い場合に被検者が癌を有することが示される工程。
ラジオイムノアッセイ(RIA)、
エンザイムイムノアッセイ(EIA)、
蛍光イムノアッセイ(FIA)、
発光イムノアッセイ(LIA)、
免疫沈降法(IP)、
免疫比濁法(TIA)、
ウエスタンブロット(WB)、
免疫組織化学(IHC)法、
免疫拡散法(SRID)
・単にHS6ST2のmRNAが存在するか否かの測定、
・HS6ST2のmRNAが一定の量以上存在するか否かの測定、
・HS6ST2のmRNAの量を他の試料(例えば、コントロール試料など)と比較する測定など
本発明は、被検試料中のHS6ST2タンパク質を検出するための試薬を含む、癌の診断のための診断薬またはキットも提供する。本発明の診断薬は、少なくとも抗HS6ST2抗体を含む。
Human Exon 1.0 ST Array (Affymetrix) を用いてHS6ST2 mRNAの臨床癌、癌細胞株および各種正常組織における発現を解析した。Human Exon 1.0 ST Arrayでは各遺伝子の各エクソンに少なくとも一つのプローブセットが設定されていることから、一遺伝子について複数のプローブセットの発現データを得ることができる。従って、基本的に一遺伝子につき一つのプローブセットしかなかったこれまでのAffymetrix社の発現アレイと比較して、発現データの信頼性が上がると考えられる。
2-1. HS6ST2のクローニング
実施例1に示した通り、癌細胞で発現するのはHS6ST2のエクソン4と5が欠失したバリアントと考えられた。このバリアントの遺伝子配列はRefSeqにNM_147175として登録されている。またUniProtにはそのアミノ酸配列がQ96MM7-1として登録されている。一方Q96MM7-1の1-146番目のアミノ酸が欠失したバリアントがQ96MM7-2として登録されている。Q96MM7-2はエクソン3の11番目の塩基から翻訳が開始されるタンパクに相当する。実施例1に示したようにHuman Exon 1.0 ST Arrayではエクソン1と2の発現が確認できなかったこと、最初にクローニングされたHS6ST2はQ96MM7-2に相当するアミノ酸配列であったことから(Biosynthesis of heparan sulphate with diverse structures and functions: two alternatively spliced forms of human heparan sulphate 6-O-sulphotransferase-2 having different expression patterns and properties. Biochem J. 2003. 371:131)、Q96MM7-2がドミナントに発現していると考えられた。そこでQ96MM7-2をHS6ST2_N-shortと名付け、そのクローニングを行った。まず癌細胞株HuH6 (理化学研究所)からTrizol (Invitrogen)を用いて製品添付の方法に従ってtotal RNAを抽出し、さらにSuperScript III Reverse Transcriptase (Invitrogen)を用いて製品添付の方法に従ってcDNAを作製した。このcDNAをテンプレートとして、配列番号1で表されるプライマー(HS6ST2_N-shortの5’末端配列)、配列番号2で表されるプライマー(HS6ST2_N-shortのストップコドンを除く3’末端配列)を用いてPCR増幅を行い、増幅産物をTOPO TA Cloning Kit (Invitrogen)を用いてTOPOベクターにクローニングした(TOPO_HS6ST2_N-short)。PCR増幅にはPyrobest DNA Polymerase (タカラバイオ)を用い、3μLの10×Pyrobest buffer II、3μLのdNTP mixture、3μLのHuH6 cDNA、1μLの配列番号1のプライマー(50μM)、1μLの配列番号2のプライマー(50μM)、0.5μLのPyrobest DNA Polymerase、18.5μLのヌクレアーゼフリー水を含む溶液を調製し、94℃ 1分、(94℃ 30秒、62℃ 30秒、72℃ 1.5分)×35サイクルで増幅を行った。このPCR産物1μLをテンプレートとして同様にPCR再増幅を行った。TOPO_HS6ST2_N-shortの配列をシークエンスしRefSeq Accession No. NM_147175と同じであることを確認した。
HS6ST2_N-short cDNAを哺乳動物細胞用発現ベクター(pMCDN2_ctV5)にクローニングした。pMCDN2_ctV5発現ベクターはマウスCMVプロモーター(GenBank Accession No. U68299)による制御下で発現誘導が可能で、かつネオマイシン耐性遺伝子が組み込まれたベクターである。また挿入した目的遺伝子の3’側にV5タグ配列が付加される。V5タグとは抗V5抗体(Invitrogen)によって認識される、GKPIPNPLLGLDSTの14アミノ酸から成る配列である。pMCDN2_ctV5発現ベクターのEcoRI認識配列-Kozak配列-開始コドン-マルチクローニングサイト(NheI、SalI、NotI)-V5タグ配列-ストップコドンの配列を配列番号3に示す。配列番号4で表されるプライマー(EcoRI認識配列-Kozak配列-HS6ST2_N-shortの5’末端配列)および配列番号5で表されるプライマー(NotI認識配列-HS6ST2_N-shortのストップコドンを除く3’末端配列)を用い、TOPO_HS6ST2_N-shortをテンプレートとしてPCR増幅を行った。増幅断片をEcoRI、NotIで消化し、pMCDN2_ctV5のEcoRI、NotIサイトにクローニングした(pMCDN2_HS6ST2_N-short_ctV5)。pMCDN2_HS6ST2_N-short_ctV5の開始コドンからストップコドンまでの塩基配列を配列番号6に、アミノ酸配列を配列番号7に示す。
HS6ST2_N-short cDNAを哺乳動物細胞用発現ベクター(pMC)にクローニングした。pMCはマウスCMVプロモーターによる制御下で発現誘導が可能なベクターである。配列番号4で表されるプライマーおよび配列番号8で表されるプライマー(SalI認識配列-ストップコドン-HS6ST2_N-shortのストップコドンを除く3’末端配列)を用い、pMCDN2_HS6ST2_N-short_ctV5をテンプレートとしてPCR増幅を行った。増幅断片をEcoRI、SalIで消化し、pMCのEcoRI、SalIサイトにクローニングした(pMC_HS6ST2_N-short)。
HS6ST2_N-shortのN末端から膜貫通領域(8-23番目のLLLALVMLFLFAVIVLの16アミノ酸)までを削除し、代わりにepidermal growth factor receptor (EGFR, RefSeq Accession No. NM_005228)のシグナル配列(MRPSGTAGAALLALLAALCPASRAの24アミノ酸配列)を付加し、さらにC末端にはFLAGタグ配列(抗FLAG抗体により認識されるDYKDDDDKの8アミノ酸からなる配列)を付加したタンパクである分泌型HS6ST2 (sHS6ST2_FLAG)の発現ベクターを作製した。配列番号9で表されるプライマー(EGFRシグナル配列の3’末端配列-HS6ST2_N-shortの膜貫通領域以降の配列)および配列番号10で表されるプライマー(NotI認識配列-ストップコドン-FLAGタグ配列- HS6ST2_N-shortのストップコドンを除く3’末端配列)を用い、pMCDN2_HS6ST2_N-short_ctV5をテンプレートとしてPCR増幅を行った。このPCR産物をテンプレートとして、配列番号11で表されるプライマー(EcoRI認識配列-Kozak配列-EGFRシグナル配列の5’末端配列)および配列番号10で表されるプライマーを用いてPCR増幅を行った。増幅断片をEcoRI、NotIで消化し、pMCDN2のEcoRI、NotIサイトにクローニングした(pMCDN2_sHS6ST2_FLAG)。pMCDN2はマウスCMVプロモーターによる制御下で発現誘導が可能で、かつネオマイシン耐性遺伝子が組み込まれたベクターである。pMCDN2_sHS6ST2_FLAGの開始コドンからストップコドンまでの塩基配列を配列番号12に、アミノ酸配列を配列番号13に示す。
BALB/cマウス(メス、6週令、日本チャールス・リバー)に対し、Helios Gene Gun (Bio-Rad)を用いて製品添付の方法により1週間に2回、合計11回のDNA免疫を行った。DNA免疫にはpMC_HS6ST2_N-short発現ベクターを用いた。DNA免疫に引き続きフロイントコンプリートアジュバント(Becton Dickinson)にてエマルジョン化したsHS6ST2_FLAG 40μgを皮下に投与した。2週間後にフロイントインコンプリートアジュバント(Becton Dickinson)にてエマルジョン化したsHS6ST2_FLAG 40μgを皮下に投与した。さらに1週間後にsHS6ST2_FLAG 50μgを尾静脈内に投与した。三日後に脾臓細胞を摘出し、マウスミエローマ細胞株P3-X63Ag8U1 (P3U1、ATCC)と2:1になるよう混合し、PEG1500 (ロシュ・ダイアグノスティックス株式会社)を徐々に加えハイブリドーマを作製した。RPMI1640培地(Invitrogen)を加え遠心後、上清を除去することによりPEG1500を除去した。次にHAT培地(10% fetal bovine serum (FBS)、penicillin-streptomycin、1×HAT media supplement (Sigma)、0.5×BM-Condimed H1 Hybridoma Cloning Supplement (ロシュ・ダイアグノスティックス)を含むRPMI1640培地)に懸濁し、96ウェルプレート8枚にP3U1細胞が1×105個/ウェルになるように播種した。これを37℃、5% CO2インキュベーターにて7日間培養後、培養上清を用いてスクリーニングを行った。スクリーニングは、培養上清に含まれる抗体のHS6ST2_N-short_ctV5_CHO細胞および親株CHO細胞に対する結合をフローサイトメーター(FACS Calibur、Becton Dickinson)を用いて測定することにより行った。HS6ST2_N-short_ctV5_CHO細胞に特異的に結合したハイブリドーマについては培養を継続し、同様な方法で再度スクリーニングを行った後、限界希釈法によりモノクローン化を行った。以上により、HS6ST2に特異的に結合する抗体としてクローンA1、A6、A10を樹立した。
3-1. マウスHS6ST2のクローニング
マウスHS6ST2はtranscript variant 1 (NM_001077202)およびtranscript variant 2 (NM_015819)の2種類のバリアントがRefSeqに登録されている。N末端146アミノ酸およびエクソン4、5が欠失している、HS6ST2_N-shortに相当するバリアントはtranscript variant 2である。そこでtranscript variant 2をマウスHS6ST2 (mHS6ST2)とし、クローニングを行った。Marathon-Ready mouse spleen cDNA (Clontech)をテンプレートとして、配列番号14で表されるプライマー(EcoRI認識配列-Kozak配列-mHS6ST2の5’末端配列)、配列番号15で表されるプライマー(NotI認識配列-mHS6ST2の3’末端配列)を用いてPCR増幅を行い、増幅産物をpGEM-T Easy Vector Systems (Promega)を用いてpGEM-T Easyベクターにクローニングした(pGEM-T_mHS6ST2)。PCR増幅にはKOD Plus Ver.2 (東洋紡)を用い、5μLの10×KOD Plus Ver.2 buffer、5μLのdNTP mixture、4μLの25 mM MgSO4、1.5μLの配列番号14のプライマー (10μM)、1.5μLの配列番号15のプライマー (10μM)、4μLのmouse spleen cDNA、1μLのKOD Plus Polymerase、28μLのヌクレアーゼフリー水を含む溶液を調製し、94℃ 2分、(98℃ 10秒、72℃ 30秒、68℃ 3分)×5サイクル、(98℃ 10秒、70℃ 30秒、68℃ 3分)×5サイクル、(98℃ 10秒、68℃ 3分)×27サイクルで増幅を行った。pGEM-T_mHS6ST2の配列をシークエンスしtranscript variant 2 (RefSeq Accession No. NM_015819)と同じであることを確認した。
mHS6ST2 cDNAを哺乳動物細胞用発現ベクター(pMCDN2_ntHA)にクローニングした。pMCDN2_ntHAはマウスCMVプロモーターによる制御下で発現誘導が可能で、かつネオマイシン耐性遺伝子が組み込まれたベクターである。挿入した目的遺伝子の5’側にHAタグ配列が付加される。HAタグ配列とはインフルエンザのヘマグルチニンタンパク質由来のHAエピトープ配列 (YPYDVPDYA)であり、HA特異的抗体により認識される。pMCDN2_ntHA発現ベクターのEcoRI認識配列-Kozak配列-開始コドン-HAタグ配列-マルチクローニングサイト(NheI、SalI、NotI)-ストップコドンの配列を配列番号16に示す。配列番号17で表されるプライマー(NheI認識配列-mHS6ST2の開始コドンを除く5’末端配列)および配列番号15で表されるプライマーを用い、pGEM-T_mHS6ST2をテンプレートとしてPCR増幅を行った。増幅断片をNheI、NotIで消化し、pMCDN2_ntHAのNheI、NotIサイトにクローニングした(pMCDN2_mHS6ST2_ntHA)。
実施例2で作製した抗HS6ST2抗体のmHS6ST2に対する結合をフローサイトメトリーにて評価した。細胞にはmHS6ST2_ctV5_CHOおよび陽性対照としてHS6ST2_N-short_ctV5_CHO、陰性対照としてCHO細胞DG44株を用いた。
実施例2で作製した抗HS6ST2抗体のエピトープを解析するため、HS6ST2_N-shortの膜貫通領域(8-23番目のLLLALVMLFLFAVIVLの16アミノ酸)より後ろを三分割し、それぞれglutathione S-transferase (GST)融合タンパクを作製した。GST_HS6ST2_NはHS6ST2_N-shortの24-175番目のアミノ酸配列、GST_HS6ST2_midは166-317番目のアミノ酸配列、GST_HS6ST2_Cは308-459番目のアミノ酸配列に加え、N末端にGST、C末端にHisタグを付加したタンパクである。Hisタグは6個の連続したヒスチジン残基からなるタグペプチドである。
実施例2で作製した抗HS6ST2抗体のADCC活性を測定した。標的細胞にはHS6ST2_N-short_ctV5_CHO細胞を用いた。1×106個のHS6ST2_N-short_ctV5_CHO細胞をChromium-51 (GE Healthcare)存在下で1時間培養した。洗浄後CHO培地で2×105個/mLに調製し、96ウェルプレートに50μL/ウェル加えた。次にCHO培地で4μg/mLに調製した抗HS6ST2抗体もしくは陰性対照としてmIgG1 (BD Biosciences Pharmingen)を50μL/ウェル加えた。室温にて15分間静置後、CHO培地で5×105/mLに調製したエフェクター細胞を100μL/ウェル加えた。エフェクター細胞にはマウスFc-ガンマ受容体3 (RefSeq Accession No. NM_010188)の細胞外領域およびヒトガンマ鎖(RefSeq Accession No. NM_004106)の膜貫通領域と細胞内領域を含むキメラタンパク質をNK-92細胞(ATCC)に強制発現させた組換え細胞(特願2007-20155、WO2008/093688)を用いた。プレートを37℃、5% CO2インキュベーターにて4時間培養後、100μL/ウェルの培養上清を回収しガンマカウンター(1480 WIZARD 3’’、Wallac)を用いて放射活性(cpm)を測定し、以下の式を用いて特異的クロム遊離率(%)を求めた。
ここで、Aは各ウェルにおける放射活性、Bは終濃度1% Nonidet P-40で細胞を溶解させたウェルの放射活性の平均値、Cは標的細胞のみを添加したウェルの放射活性の平均値である。B、Cはtriplicate、その他はduplicateで行い、特異的クロム遊離率の平均値と標準偏差を算出した。
実施例2で作製した抗HS6ST2抗体のイムノトキシンとしてのポテンシャルをMab-ZAP (Advanced Targeting Systems)を用いて評価した。Mab-ZAPはヤギ抗マウスIgG抗体にサポリン(saporin)が標識された抗体である。サポリンはリボソームにおけるタンパク質合成を阻害する毒素である。細胞表面上の抗原に結合するマウス抗体とMab-ZAPを同時に培養系に添加すると、そのマウス抗体が細胞内に取り込まれる場合にはMab-ZAPも同時に細胞内に取り込まれ細胞の増殖を抑制する。
7-1. HS6ST2バリアントのクローニング
HS6ST2にはN末端アミノ酸配列が長いバリアント(UniProt Q96MM7-1, RefSeq Accession No. NM_147175)が存在する。このバリアントをHS6ST2_N-longと名付け、実施例2で作製した抗HS6ST2抗体が結合することを確認した。
pMCDN2_HS6ST2_N-long_ctV5をPvuI消化したものをエレクトロポレーション法によりCHO細胞DG44株に導入した。Geneticin (500μg/mL)により導入細胞株を選抜することにより、C末端V5タグ付加HS6ST2_N-long定常発現CHO細胞株(HS6ST2_N-long_ctV5_CHO)を樹立した。培養にはCHO培地を用いた。
実施例2で作製した抗HS6ST2抗体のHS6ST2_N-longに対する結合活性をフローサイトメトリーにて評価した。細胞にはHS6ST2_N-long_ctV5_CHO細胞および陽性対照としてHS6ST2_N-short_ctV5_CHO細胞を用い、実施例3と同様の方法で行った。一次抗体は10μg/mLの濃度で用いた。
8-1. フローサイトメトリーによるHS6ST2の発現解析
実施例2で作製した抗HS6ST2抗体を用いて、癌細胞株の細胞膜上におけるHS6ST2の発現をフローサイトメトリーを用いて評価した。一次抗体には抗HS6ST2抗体B6または陰性対照mIgG1 (BD Biosciences Pharmingen)、細胞にはHuman Exon 1.0 ST ArrayにてHS6ST2の発現が最も高かった肺腺癌細胞株ABC-1および肝臓癌細胞株HuH6を用いた(実施例1)。フローサイトメトリーは実施例3と同様に行い、一次抗体は10μg/mLの濃度で用いた。その結果、いずれの細胞でも細胞膜上にHS6ST2の発現が確認された(図9)。
実施例2で作製した抗HS6ST2抗体を用いたウェスタンブロットを検討した。まず1×106個のHS6ST2_N-short_ctV5_CHO細胞およびHS6ST2_N-long_ctV5_CHO細胞をPBSで洗浄後、100μLのlysis buffer (50 mM Tris-HCl, pH7.4, 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, Protease Inhibitor Cocktail (Sigma))で溶解しwhole cell lysateを調製した。これをN-Glycosidase F (PNGaseF, New England Biolabs)を用いて処理することによりN型糖鎖を除去した。サンプルをMultigel II Mini (8/16、コスモバイオ)にてSDS-PAGE電気泳動後、PVDFメンブレン(Immobilon-P, Millipore)に転写し、抗HS6ST2抗体C10または抗V5タグ抗体(Invitrogen)を用いてウェスタンブロットを行った。抗HS6ST2抗体C10は5μg/mL、抗V5タグ抗体は5000倍希釈にて用い、室温にて1時間反応させた。二次抗体にはHRP標識抗マウスIgG抗体(GE Healthcare)を用い、室温にて1時間反応させた。最後にECL Western Blotting Detection Reagents (GE Healthcare)を用いて発色させ、X線フィルムに露光することによりバンドを検出した。
肺腺癌細胞株および卵巣癌細胞株におけるHS6ST2の発現とその分子量、および培養上清中への分泌を、抗HS6ST2抗体C10を用いたウェスタンブロットにより測定した。肺腺癌細胞株A549、ABC-1 (以上JCRB Cell Bank)、NCI-H441、NCI-H1781 (以上ATCC)、卵巣癌細胞株OVMANA (JCRB Cell Bank)から調製したwhole cell lysateをN-Glycosidase Fにて処理しウェスタンブロットを行った。Whole cell lysateのタンパク濃度はDC Protein Assay Kit I (Bio-Rad)にて測定し、A549は10μg/レーン、その他は15μg/レーンになるように泳動した。その結果、肺腺癌細胞株ABC-1、NCI-H441、NCI-H1781および卵巣癌細胞株OVMANAでHS6ST2が発現していた(図11A)。いずれもN-Glycosidase F処理後の分子量が50-60kDaであったことから、N-shortバリアントであると考えられた。なおA549は実施例1でHS6ST2を発現していないことを確認した細胞株であり、陰性対照として用いた。
9-1. 可溶型HS6ST2のヘパラン硫酸への結合
細胞から分泌された可溶型HS6ST2が細胞膜上のヘパラン硫酸に結合することを確認した。まず細胞膜上のヘパラン硫酸の発現を実施例3と同様にフローサイトメトリーにて検討した。細胞には肺腺癌細胞株A549、ABC-1、NCI-H441、NCI-H1781、卵巣癌細胞株OVMANAおよびCHO細胞DG44株を用いた。一次抗体には抗ヘパラン硫酸抗体(HepSS-1、生化学バイオビジネス)および陰性対照としてマウスIgM (mIgM、BD Biosciences Pharmingen)を20μg/mL (NCI-H441、NCI-H1781)または40μg/mL (A549、ABC-1、OVMANA、DG44)の濃度で用いた。その結果A549、ABC-1およびDG44がヘパラン硫酸を発現することが確認された(図12)。
ヘパラン硫酸の発現量が高かった肺腺癌細胞株A549へのsHS6ST2_FLAGの結合を評価した。A549細胞を100, 20, 4, 0.8, 0μg/mLのsHS6ST2_FLAGと氷上にて2時間反応させた後、フローサイトメトリー解析を行ったところ、濃度依存的にsHS6ST2_FLAGが結合した(図14A)。
実施例8にて癌細胞株が可溶型HS6ST2を分泌し、癌の診断マーカーになる可能性が考えられたため、可溶型HS6ST2を検出するELISA系を構築した。抗体には抗HS6ST2抗体A6、B5、C8を用いた。B5とC8はHS6ST2_N-shortの379-459番目のアミノ酸配列に結合し、A6は308-393番目のアミノ酸配列に結合する(実施例4)。またC8はmHS6ST2に結合するがB5は結合しない(実施例3)。従って三者の結合部位は異なると考えられた。
HS6ST2遺伝子の発現が肺腺癌で亢進していたことから(実施例1)、肺癌におけるHS6ST2タンパクの発現を免疫組織化学染色により解析した。各検体より4% paraformaldehyde固定AMeX包埋パラフィンブロックを作製し、5μmの薄切切片を作製した。これらの切片についてVentana HX Discovery System (Ventana Medical Systems)を用いて下記のように免疫組織化学的に染色した。各切片を脱パラフィン後に洗浄し、Cell Conditioner #1 (Ventana Medical Systems)を用いて100℃にて30分間加熱処理後、内因性peroxidaseの除去のため3.0% hydrogen peroxide solution (Inhibitor D、Ventana Medical Systems)を用いて37℃にて4分間反応させた。洗浄後、非特異的反応の除去のためProtein Block (Dako)を加え、室温にて30分間反応させた。洗浄後、一次抗体として25μg/mLの抗HS6ST2抗体C10を加え室温にて2時間反応させた。洗浄後、二次抗体(Ventana Universal Secondary Antibody、Ventana Medical Systems)を加え、室温にて30分間反応させた。洗浄後、非特異的反応の除去のためBlocker D (Ventana Medical Systems)にて室温にて2分間反応させ、続いてstreptavidin horseradish peroxidase (Ventana Medical Systems)を加え、37℃にて16分間反応させた。洗浄後、diaminobenzidine (DAB map solution、Ventana Medical Systems)とhydrogen peroxide solution (DAB map solution、Ventana Medical Systems)を混和させて加え、基質の発色のため42℃にて8分間反応させた。さらにCopper sulfate solution (Ventana Medical Systems)にて発色の増感を行った。洗浄後、ヘマトキシリンにて核染を行い、脱水、透徹、封入を行った。
実施例2にて作製した抗HS6ST2抗体の可変領域の核酸配列およびアミノ酸配列を決定した。それぞれの抗体を産生するハイブリドーマ細胞1×106個からTrizol (Invitrogen)を用いて製品添付の方法に従ってtotal RNAを精製した。Total RNA 1μgを使用し、SMART RACE cDNA Amplification Kit (Clontech)、マウスIgG1定常領域配列に相補的な合成オリゴヌクレオチドMHC-IgG1 (配列番号34)、およびマウスκ鎖定常領域配列に相補的な合成オリゴヌクレオチドMLC-kappa (配列番号35)を用い、抗体のH鎖、L鎖cDNAの上述オリゴヌクレオチド配列に相当する位置から5’-cDNA末端までの配列をPCR増幅した。増幅産物をpGEM-T Easy Vector Systems (Promega)を用いてpGEM-T Easyベクターにクローニングし、cDNA配列を決定した。下表に各抗体の可変領域配列の配列番号をまとめた。
Claims (13)
- HS6ST2タンパク質に結合する抗体。
- 細胞傷害活性を有することを特徴とする請求項1に記載の抗体。
- 細胞傷害性物質が結合していることを特徴とする請求項1または2に記載の抗体。
- ヘパラン硫酸に結合したHS6ST2に結合することを特徴とする請求項1~3のいずれかに記載の抗体。
- 細胞膜上に発現したHS6ST2に結合することを特徴とする請求項1~3のいずれかに記載の抗体。
- 以下のいずれかに記載の抗体であることを特徴とする請求項1~5のいずれかに記載の抗体:
(1)配列番号:64に記載のアミノ酸配列を有する重鎖CDR1、配列番号:65に記載のアミノ酸配列を有する重鎖CDR2、配列番号:66に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(A1);
(2)配列番号:70に記載のアミノ酸配列を有する重鎖CDR1、配列番号:71に記載のアミノ酸配列を有する重鎖CDR2、配列番号:72に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(A6);
(3)配列番号:76に記載のアミノ酸配列を有する重鎖CDR1、配列番号:77に記載のアミノ酸配列を有する重鎖CDR2、配列番号:78に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(A10);
(4)配列番号:82に記載のアミノ酸配列を有する重鎖CDR1、配列番号:83に記載のアミノ酸配列を有する重鎖CDR2、配列番号:84に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(B5);
(5)配列番号:88に記載のアミノ酸配列を有する重鎖CDR1、配列番号:89に記載のアミノ酸配列を有する重鎖CDR2、配列番号:90に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(B6);
(6)配列番号:94に記載のアミノ酸配列を有する重鎖CDR1、配列番号:95に記載のアミノ酸配列を有する重鎖CDR2、配列番号:96に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(C8);
(7)配列番号:100に記載のアミノ酸配列を有する重鎖CDR1、配列番号:101に記載のアミノ酸配列を有する重鎖CDR2、配列番号:102に記載のアミノ酸配列を有する重鎖CDR3を含む重鎖可変領域を含む抗体(C10);
(8)配列番号:67に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:68に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:69に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(A1);
(9)配列番号:73に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:74に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:75に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(A6);
(10)配列番号:79に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:80に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:81に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(A10);
(11)配列番号:85に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:86に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:87に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(B5);
(12)配列番号:91に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:92に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:93に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(B6);
(13)配列番号:97に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:98に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:99に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(C8);
(14)配列番号:103に記載のアミノ酸配列を有する軽鎖CDR1、配列番号:104に記載のアミノ酸配列を有する軽鎖CDR2、配列番号:105に記載のアミノ酸配列を有する軽鎖CDR3を含む軽鎖可変領域を含む抗体(C10);
(15)(1)の重鎖可変領域と(8)の軽鎖可変領域を含む抗体(A1);
(16)(2)の重鎖可変領域と(9)の軽鎖可変領域を含む抗体(A6);
(17)(3)の重鎖可変領域と(10)の軽鎖可変領域を含む抗体(A10);
(18)(4)の重鎖可変領域と(11)の軽鎖可変領域を含む抗体(B5);
(19)(5)の重鎖可変領域と(12)の軽鎖可変領域を含む抗体(B6);
(20)(6)の重鎖可変領域と(13)の軽鎖可変領域を含む抗体(C8);
(21)(7)の重鎖可変領域と(14)の軽鎖可変領域を含む抗体(C10);
(22)(1)から(21)のいずれかに記載の抗体において1若しくは複数のアミノ酸が置換、欠失、付加および/または挿入された抗体であって、(1)から(21)のいずれかに記載の抗体と同等の活性を有する抗体;
(23)(1)から(21)のいずれかに記載の抗体が結合するHS6ST2タンパク質のエピトープと同じエピトープに結合する抗体。 - 配列番号:107のアミノ酸配列を有するHS6ST2タンパク質の379番目のアミノ酸から459番目のアミノ酸までの部位、308番目のアミノ酸から393番目のアミノ酸までの部位、または24番目のアミノ酸から175番目のアミノ酸までの部位を認識することを特徴とする請求項1~5のいずれかに記載の抗体。
- 請求項1~7のいずれかに記載の抗体を有効成分として含む医薬組成物。
- 抗癌剤である請求項8に記載の医薬組成物。
- 以下の工程を含む癌の診断方法:
(a)被験者から採取された試料を提供する工程、
(b) (a)の試料に含まれるHS6ST2タンパク質又はHS6ST2遺伝子を検出する工程。 - 細胞傷害性物質が結合したHS6ST2タンパク質。
- 請求項11に記載のHS6ST2タンパク質を有効成分として含む医薬組成物。
- 抗癌剤である請求項12に記載の医薬組成物。
Priority Applications (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US12/998,988 US8846870B2 (en) | 2008-12-22 | 2009-12-22 | Anti-HS6ST2 antibodies and uses thereof |
JP2010544062A JP5756292B2 (ja) | 2008-12-22 | 2009-12-22 | 抗hs6st2抗体及びその用途 |
EP09834850.1A EP2388320B1 (en) | 2008-12-22 | 2009-12-22 | Anti-hs6st2 antibodies and uses thereof |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2008324883 | 2008-12-22 | ||
JP2008-324883 | 2008-12-22 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2010074049A1 true WO2010074049A1 (ja) | 2010-07-01 |
Family
ID=42287658
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2009/071271 WO2010074049A1 (ja) | 2008-12-22 | 2009-12-22 | 抗hs6st2抗体及びその用途 |
Country Status (4)
Country | Link |
---|---|
US (1) | US8846870B2 (ja) |
EP (1) | EP2388320B1 (ja) |
JP (1) | JP5756292B2 (ja) |
WO (1) | WO2010074049A1 (ja) |
Cited By (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2013205362A (ja) * | 2012-03-29 | 2013-10-07 | Forerunner Pharma Research Co Ltd | 大腸癌マーカー、および予後の予測方法 |
WO2015098112A1 (ja) * | 2013-12-27 | 2015-07-02 | 独立行政法人医薬基盤研究所 | 食道がんのマーカーおよびその利用 |
JP2020531045A (ja) * | 2017-07-14 | 2020-11-05 | シートムエックス セラピューティクス,インコーポレイテッド | 抗cd166抗体およびその使用 |
US11753466B2 (en) | 2015-05-04 | 2023-09-12 | Cytomx Therapeutics, Inc. | Anti-CD166 antibodies, activatable anti-CD166 antibodies, and methods of use thereof |
Families Citing this family (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US10676723B2 (en) | 2015-05-11 | 2020-06-09 | David Gordon Bermudes | Chimeric protein toxins for expression by therapeutic bacteria |
US20220017610A1 (en) * | 2018-11-16 | 2022-01-20 | The Brigham And Women`S Hospital, Inc. | Antibodies Blocking DLL4-Mediated Notch Signalling |
US20220026445A1 (en) * | 2018-12-07 | 2022-01-27 | Georgia Tech Research Corporation | Antibodies that bind to natively folded myocilin |
Citations (16)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP0239400A2 (en) | 1986-03-27 | 1987-09-30 | Medical Research Council | Recombinant antibodies and methods for their production |
EP0404097A2 (de) | 1989-06-22 | 1990-12-27 | BEHRINGWERKE Aktiengesellschaft | Bispezifische und oligospezifische, mono- und oligovalente Rezeptoren, ihre Herstellung und Verwendung |
WO1992003918A1 (en) | 1990-08-29 | 1992-03-19 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
WO1993011161A1 (en) | 1991-11-25 | 1993-06-10 | Enzon, Inc. | Multivalent antigen-binding proteins |
WO1993012227A1 (en) | 1991-12-17 | 1993-06-24 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
WO1994002602A1 (en) | 1992-07-24 | 1994-02-03 | Cell Genesys, Inc. | Generation of xenogeneic antibodies |
WO1994011523A2 (en) | 1992-11-13 | 1994-05-26 | Idec Pharmaceuticals Corporation | Fully impaired consensus kozac sequences for mammalian expression |
WO1994025585A1 (en) | 1993-04-26 | 1994-11-10 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
WO1996002576A1 (fr) | 1994-07-13 | 1996-02-01 | Chugai Seiyaku Kabushiki Kaisha | Anticorps humain reconstitue contre l'interleukine-8 humaine |
WO1999054342A1 (en) | 1998-04-20 | 1999-10-28 | Pablo Umana | Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity |
WO2000061739A1 (en) | 1999-04-09 | 2000-10-19 | Kyowa Hakko Kogyo Co., Ltd. | Method for controlling the activity of immunologically functional molecule |
WO2002031140A1 (fr) | 2000-10-06 | 2002-04-18 | Kyowa Hakko Kogyo Co., Ltd. | Cellules produisant des compositions d'anticorps |
WO2002079255A1 (en) | 2001-04-02 | 2002-10-10 | Idec Pharmaceuticals Corporation | RECOMBINANT ANTIBODIES COEXPRESSED WITH GnTIII |
WO2003104453A1 (ja) | 2002-06-05 | 2003-12-18 | 中外製薬株式会社 | 抗体作製方法 |
JP2007020155A (ja) | 2005-06-08 | 2007-01-25 | Canon Inc | 画像処理装置、画像処理方法、プログラム、及び記憶媒体 |
WO2008093688A1 (ja) | 2007-01-30 | 2008-08-07 | Forerunner Pharma Research Co., Ltd. | キメラFcγレセプター及び該レセプターを用いたADCC活性測定方法 |
Family Cites Families (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP1295946B1 (en) * | 2000-06-29 | 2007-05-16 | Seikagaku Corporation | Sulfate transferase and dna encoding this enzyme |
-
2009
- 2009-12-22 JP JP2010544062A patent/JP5756292B2/ja active Active
- 2009-12-22 US US12/998,988 patent/US8846870B2/en active Active
- 2009-12-22 EP EP09834850.1A patent/EP2388320B1/en active Active
- 2009-12-22 WO PCT/JP2009/071271 patent/WO2010074049A1/ja active Application Filing
Patent Citations (16)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP0239400A2 (en) | 1986-03-27 | 1987-09-30 | Medical Research Council | Recombinant antibodies and methods for their production |
EP0404097A2 (de) | 1989-06-22 | 1990-12-27 | BEHRINGWERKE Aktiengesellschaft | Bispezifische und oligospezifische, mono- und oligovalente Rezeptoren, ihre Herstellung und Verwendung |
WO1992003918A1 (en) | 1990-08-29 | 1992-03-19 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
WO1993011161A1 (en) | 1991-11-25 | 1993-06-10 | Enzon, Inc. | Multivalent antigen-binding proteins |
WO1993012227A1 (en) | 1991-12-17 | 1993-06-24 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
WO1994002602A1 (en) | 1992-07-24 | 1994-02-03 | Cell Genesys, Inc. | Generation of xenogeneic antibodies |
WO1994011523A2 (en) | 1992-11-13 | 1994-05-26 | Idec Pharmaceuticals Corporation | Fully impaired consensus kozac sequences for mammalian expression |
WO1994025585A1 (en) | 1993-04-26 | 1994-11-10 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
WO1996002576A1 (fr) | 1994-07-13 | 1996-02-01 | Chugai Seiyaku Kabushiki Kaisha | Anticorps humain reconstitue contre l'interleukine-8 humaine |
WO1999054342A1 (en) | 1998-04-20 | 1999-10-28 | Pablo Umana | Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity |
WO2000061739A1 (en) | 1999-04-09 | 2000-10-19 | Kyowa Hakko Kogyo Co., Ltd. | Method for controlling the activity of immunologically functional molecule |
WO2002031140A1 (fr) | 2000-10-06 | 2002-04-18 | Kyowa Hakko Kogyo Co., Ltd. | Cellules produisant des compositions d'anticorps |
WO2002079255A1 (en) | 2001-04-02 | 2002-10-10 | Idec Pharmaceuticals Corporation | RECOMBINANT ANTIBODIES COEXPRESSED WITH GnTIII |
WO2003104453A1 (ja) | 2002-06-05 | 2003-12-18 | 中外製薬株式会社 | 抗体作製方法 |
JP2007020155A (ja) | 2005-06-08 | 2007-01-25 | Canon Inc | 画像処理装置、画像処理方法、プログラム、及び記憶媒体 |
WO2008093688A1 (ja) | 2007-01-30 | 2008-08-07 | Forerunner Pharma Research Co., Ltd. | キメラFcγレセプター及び該レセプターを用いたADCC活性測定方法 |
Non-Patent Citations (103)
Title |
---|
"Affinity, kinetic, and structural study of the interaction of 3-O-sulfotransferase isoform 1 with heparan sulfate", BIOCHEMISTRY, vol. 45, 2006, pages 5122 |
"Antibodies A Laboratory Manual.", 1988, COLD SPRING HARBOR LABORATORY |
"Biosynthesis of heparan sulphate with diverse structures and functions: two alternatively spliced forms of human heparan sulphate 6.O-sulphotransferase-2 having different expression patterns and properties", BIOCHEM J., vol. 371, 2003, pages 131 |
"Biosynthesis of heparan sulphate with diverse structures and functions: two alternatively spliced forms of human heparan sulphate 6-0-sulphotransferase-2 having different expression patterns and properties", BIOCHEM J., vol. 371, 2003, pages 131 |
"Cell surface heparan sulfate proteoglycans", J BIOL CHEM., vol. 267, 1992, pages 9451 |
"Current protocols in Immunology", 1993, JOHN WILEY & SONS, INC. |
"Functions of cell surface heparan sulfate proteoglycans", ANNU REV BIOCHEM., vol. 68, 1999, pages 729 - 777 |
"Glycosaminoglycans in human lung cancer", CANCER., vol. 48, 1981, pages 2016 |
"Heparan sulfate is essential to amphiregulin-induced mitogenic signaling by the epidermal growth factor receptor", J BIOL CHEM., vol. 269, 1994, pages 27149 |
"Heparan sulfate proteoglycan as a plasma membrane carrier", TRENDS BIOCHEM SCI., vol. 28, 2003, pages 145 |
"Heparan sulfate proteoglycan expression in human lung-cancer cells", INT J CANCER., vol. 74, 1997, pages 335 |
"Heparan sulfate undergoes specific structural changes during the progression from human colon adenoma to carcinoma in vitro", J BIOL CHEM., vol. 273, 1998, pages 51 |
"Heparan sulfate: growth control with a restricted sequence menu", J CLIN INVEST., vol. 108, 2001, pages 357 - 361 |
"Heterogeneity of heparan sulfates in human lung", AM J RESPIR CELL MOL BIOL., vol. 30, 2004, pages 166 |
"Internalization and trafficking of cell surface proteoglycans and proteoglycan-binding ligands", TRAFFIC., vol. 8, 2007, pages 389 |
"Isolation and characterization of glycosaminoglycans in human plasma", J CLIN INVEST, vol. 76, 1985, pages 1984 |
"Molecular characterization and expression of heparan-sulfate 6-sulfotransferase", J BIOL CHEM., vol. 273, 1998, pages 9208 |
"Novel aspects of glypican glycobiology", CELL MOL LIFE SCI., vol. 61, 2004, pages 1016 |
"Oligosaccharide library-based assessment of heparan sulfate 6-O-sulfotransferase substrate specificity", J BIOL CHEM., vol. 278, 2003, pages 24371 |
"Purification and characterization of heparan sulfate 6- sulfotransferase from the culture medium of Chinese hamster ovary cells", J BIOL CHEM., vol. 270, 1995, pages 4172 |
"Regulated diversity of heparan sulfate", J BIOL CHEM., vol. 273, 1998, pages 24979 |
"Regulation of heparan sulfate 6-O-sulfation by ?-secretase activity", J BIOL CHEM., 2007, pages 282 |
"Remington's Pharmaceutical Science", MARK PUBLISHING COMPANY |
"Serum and urinary concentrations of heparan sulfate in patients with diabetic nephropathy", KIDNEY INT, vol. 56, 1999, pages 650 |
"Soluble syndecan-1 and serum basic fibroblast growth factor are new prognostic factors in lung cancer", CANCER RES, vol. 62, 2002, pages 5210 |
"Stem domains of heparan sulfate 6-O.sulfotransferase are required for golgi localization, oligomer formation and enzyme activity", J CELL SCI., vol. 117, 2004, pages 3331 |
"Structural differences between heparan sulphates of proteoglycan involved in the formation of basement membranes in vivo by Lewis-lung-carcinoma-derived cloned cells with different metastatic potentials", BIOCHEM J., vol. 288, 1992, pages 215 |
"The cell-surface heparan sulfate proteoglycan glypican-1 regulates growth factor action in pancreatic carcinoma cells and is overexpressed in human pancreatic cancer", J CLIN INVEST., vol. 102, 1998, pages 1662 |
"The occurrence of three isoforms of heparan sulfate 6-O-sulfotransferase having different specificities for hexuronic acid adjacent to the targeted N-sulfoglucosamine", J BIOL CHEM., vol. 275, 2000, pages 2859 |
ACTA ONCOL., vol. 32, 1993, pages 825 - 830 |
BACKEN A.C. ET AL: "Heparan sulphate synthetic and editing enzymes in ovarian cancer", BR. J. CANCER, vol. 96, no. 10, 2007, pages 1544 - 1548, XP055037801, DOI: doi:10.1038/sj.bjc.6603747 * |
BELYAVSKY, A. ET AL., NUCLEIC ACIDS RES., vol. 17, 1989, pages 2919 - 2932 |
BETTER ET AL., SCIENCE, vol. 240, 1988, pages 1041 - 1043 |
BETTER, M., HORWITZ, A. H., METHODS ENZYMOL., vol. 178, 1989, pages 476 - 496 |
BETTER, M., HORWITZ, A. H., METHODS IN ENZYMOLOGY, vol. 178, 1989, pages 476 - 496 |
BIOCHEM J., vol. 371, 2003, pages 131 |
BIRD, R. E. ET AL., TIBTECH, vol. 9, 1991, pages 132 - 137 |
BIRD, R. E., WALKER, B. W., TRENDS BIOTECHNOL., vol. 9, 1991, pages 132 - 137 |
BOLOGNESI A. ET AL., CLIN. EXP. IMMUNOL., vol. 89, 1992, pages 341 - 346 |
CASELLAS P. ET AL., EUR. J. BIOCHEM., vol. 176, 1988, pages 581 - 588 |
CELLULAR & MOLECULAR IMMUNOLOGY, vol. 3, 2006, pages 439 - 443 |
CHIRGWIN, J. M. ET AL., BIOCHEMISTRY, vol. 18, 1979 |
CHOMCZYNSKI, P. ET AL., ANAL. BIOCHEM., vol. 162, 1987, pages 156 - 159 |
CO, M. S. ET AL., J. IMMUNOL., vol. 152, 1994, pages 2968 - 2976 |
CO, M.S. ET AL., J. IMMUNOL., vol. 152, 1994, pages 2968 - 2976 |
CUMBER A.J. ET AL., J. IMMUNOL. METHODS, vol. 135, 1990, pages 15 - 24 |
CURRENT TOPICS IN MICROBIOLOGY AND IMMUNOLOGY, vol. 81, 1978, pages 1 - 7 |
DALBADIE-MCFARLAND, G. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 79, 1982, pages 6409 - 6413 |
DE ST. GROTH, S. F. ET AL., J. IMMUNOL. METHODS, vol. 35, 1980, pages 1 - 21 |
EBERT, K.M. ET AL., BIO/TECHNOLOGY, vol. 12, 1994, pages 699 - 702 |
FASEB J., vol. 10, 1996, pages 598 - 614 |
FASEBJ., vol. 6, 1992, pages 2422 - 2427 |
FROHMAN, M.A. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 8998 - 9002 |
FULTON R.J. ET AL., J. BIOL. CHEM., vol. 261, 1986, pages 5314 - 5319 |
GALFRE, G. ET AL., NATURE, vol. 277, 1979, pages 131 - 133 |
GHEEITE V. ET AL., J. IMMUNOL. METHODS, vol. 142, 1991, pages 223 - 230 |
GOICHIRO MIYAKE ET AL: "Heparan Ryusan-O- Ryusan Ten'i Koso-gun (HS2ST, HS6ST) no Hito Sodo Idenshi no Cloning to Gan Soshiki ni Okeru Hatsugen", CANCER SCIENCE, vol. 91, 1 September 2000 (2000-09-01), pages 145 * |
HABUCHI H. ET AL: "Biosynthesis of heparan sulphate with diverse structures and functions: two alternatively spliced forms of human heparansulphate 6-0-sulphotransferase-2 having different expression patterns and properties", BIOCHEM. J., vol. 371, 2003, pages 131 - 142, XP002683188, DOI: doi:10.1042/bj20021259 * |
HABUCHI H. ET AL: "The occurrence of three isoforms of heparan sulfate 6-0- sulfotransferase having different specificities for hexuronic acid adjacent to the targeted N-sulfoglucosamine", J. BIOL. CHEM., vol. 275, no. 4, 2000, pages 2859 - 2868, XP002148154, DOI: doi:10.1074/jbc.275.4.2859 * |
HASHIMOTO-GOTOH, T. ET AL., GENE, vol. 152, 1995, pages 271 - 275 |
HOLLIGER P ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448 |
HUDSON ET AL., J IMMUNOL. METHODS, vol. 231, 1999, pages 177 - 189 |
J BIOL CHEM., vol. 279, 2004, pages 42732 |
J. IMMUNOL., vol. 123, 1979, pages 1548 - 1550 |
J. MOL. BIOL., vol. 222, 1991, pages 581 - 597 |
KOHLER. G., MILSTEIN, C., EUR. J. IMMUNOL., vol. 6, 1976, pages 511 - 519 |
KOHLER. G., MILSTEIN, C., METHODS ENZYMOL., vol. 73, 1981, pages 3 - 46 |
KRAMER W, FRITZ HJ, METHODS. ENZYMOL., vol. 154, 1987, pages 350 - 367 |
KRAMER, W. ET AL., NUCLEIC ACIDS RES., vol. 12, 1984, pages 9441 - 9456 |
KUNKEL, METHODS ENZYMOL., vol. 85, 1988, pages 2763 - 2766 |
KUNKEL, TA, PROC NATL ACAD SCI USA., vol. 82, 1985, pages 488 - 492 |
LAMOYI, E., METHODS ENZYMOL., vol. 121, 1986, pages 652 - 663 |
LAMOYI, E., METHODS IN ENZYMOLOGY, vol. 121, 1989, pages 652 - 663 |
LANGONE J.J. ET AL., METHODS IN ENZYMOLOGY, vol. 93, 1983, pages 307 - 308 |
LEI, S. P. ET AL., J. BACTERIOL., vol. 169, 1987, pages 4379 |
MARGULIES. D.H. ET AL., CELL, vol. 8, 1976, pages 405 - 415 |
MARK, D. F. ET AL., PROC. NAT1. ACAD. SCI. USA, vol. 81, 1984, pages 5662 - 5666 |
MIZUSHIMA ET AL., NUCLEIC ACIDS RES., vol. 18, 1990, pages 5322 |
MULLIGAN ET AL., NATURE, vol. 277, 1979, pages 108 |
NATURE MEDICINE, vol. 2, 1996, pages 350 - 353 |
PLUCKTHUN, A., SKERRA, A., METHODS ENZYMOL., vol. 178, 1989, pages 497 - 515 |
PROC. NATL. ACAD. SCI. U.S.A, vol. 85, 1988, pages 5879 - 5883 |
PROTEIN ENGINEERING, vol. 9, no. 3, 1996, pages 299 - 305 |
ROUSSEAUX, J. ET AL., METHODS ENZYMOL., vol. 121, 1986, pages 663 - 669 |
ROUSSEAUX, J. ET AL., METHODS IN ENZYMOLOGY, vol. 121, 1989, pages 663 - 669 |
SAMBROOK, J ET AL.: "Molecular Cloning", 1989, COLD SPRING HARBOR LAB. PRESS |
SATO, K. ET AL., CANCER RES, vol. 53, 1993, pages 851 - 856 |
See also references of EP2388320A4 * |
SHULMAN, M. ET AL., NATURE, vol. 276, 1978, pages 269 - 270 |
SIVAM G. ET AL., CANCER RES., vol. 47, 1987, pages 3169 - 3173 |
SKERRA, A., METHODS IN ENZYMOLOGY, vol. 178, 1989, pages 476 - 496 |
STIRPE F., BARBIERI L, FEBS 1 LETTER, vol. 195, 1986, pages 1 - 8 |
STIRPE F., BARBIERI L., FEBS LETTER, vol. 195, 1986, pages 1 - 8 |
STIRPE, vol. 1-8, 1986, pages 1 |
THORPE P.E. ET AL., CANCER RES., vol. 47, 1987, pages 5924 - 5931 |
TROWBRIDGE, I. S., J. EXP. MED., vol. 148, 1978, pages 313 - 323 |
VANDAMME, A. M. ET AL., EUR. J. BIOCHEM., vol. 192, 1990, pages 767 - 775 |
WANG, A. ET AL., SCIENCE, vol. 224, pages 1431 - 1433 |
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546 |
WAWRZYNCZAK E.J. ET AL., BR. J. CANCER, vol. 66, 1992, pages 361 - 366 |
WAWRZYNCZAK E.J. ET AL., CANCER RES., vol. 50, 1990, pages 7519 - 7562 |
ZOLLER, M. J., SMITH, M., NUCLEIC ACIDS RESEARCH, vol. 10, 1982, pages 6487 - 6500 |
ZOLLER, MJ, SMITH, M., METHODS ENZYMOL., vol. 100, 1983, pages 468 - 500 |
Cited By (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2013205362A (ja) * | 2012-03-29 | 2013-10-07 | Forerunner Pharma Research Co Ltd | 大腸癌マーカー、および予後の予測方法 |
WO2015098112A1 (ja) * | 2013-12-27 | 2015-07-02 | 独立行政法人医薬基盤研究所 | 食道がんのマーカーおよびその利用 |
US11753466B2 (en) | 2015-05-04 | 2023-09-12 | Cytomx Therapeutics, Inc. | Anti-CD166 antibodies, activatable anti-CD166 antibodies, and methods of use thereof |
JP2020531045A (ja) * | 2017-07-14 | 2020-11-05 | シートムエックス セラピューティクス,インコーポレイテッド | 抗cd166抗体およびその使用 |
Also Published As
Publication number | Publication date |
---|---|
JP5756292B2 (ja) | 2015-07-29 |
EP2388320A4 (en) | 2012-11-07 |
JPWO2010074049A1 (ja) | 2012-06-14 |
EP2388320A1 (en) | 2011-11-23 |
US8846870B2 (en) | 2014-09-30 |
EP2388320B1 (en) | 2017-02-15 |
US20110262929A1 (en) | 2011-10-27 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20210380715A1 (en) | Anti-dll3 antibody | |
JP5986621B2 (ja) | 抗gpr49抗体を用いる癌の診断および治療 | |
JP6162864B2 (ja) | 抗Desmoglein3抗体を用いる癌の診断および治療 | |
JP5848863B2 (ja) | 抗cldn6抗体 | |
JP6104794B2 (ja) | 抗itm2a抗体を用いる癌の診断および治療 | |
JP6009733B2 (ja) | 抗tm4sf20抗体を用いた癌の診断と治療 | |
JP5756292B2 (ja) | 抗hs6st2抗体及びその用途 | |
WO2011105573A1 (ja) | 抗icam3抗体およびその用途 | |
JP5801557B2 (ja) | 抗lgr7抗体を用いる癌の診断および治療 | |
JP5746018B2 (ja) | 抗tmprss11e抗体を用いた癌の診断と治療 | |
JP5618172B2 (ja) | 抗prg−3抗体を用いる癌の診断および治療 |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 09834850 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2010544062 Country of ref document: JP Kind code of ref document: A |
|
WWE | Wipo information: entry into national phase |
Ref document number: 12998988 Country of ref document: US |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
REEP | Request for entry into the european phase |
Ref document number: 2009834850 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2009834850 Country of ref document: EP |